curcuminasananti inflammatoryagent:implications...

13
Received: 13 August 2018 | Accepted: 27 August 2018 DOI: 10.1002/jcp.27442 REVIEW ARTICLE Curcumin as an antiinflammatory agent: Implications to radiotherapy and chemotherapy Bagher Farhood 1 | Keywan Mortezaee 2 | Nasser Hashemi Goradel 3 | Neda Khanlarkhani 4 | Ensieh Salehi 4 | Maryam Shabani Nashtaei 4,5 | Masoud Najafi 6 | Amirhossein Sahebkar 7,8 1 Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran 2 Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran 3 Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran 4 Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran 5 Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran 6 Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran 7 Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran 8 Biotechnology Research Center, Pharmaceutical Technology Institute, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran Correspondence Masoud Najafi, Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6719851351, Iran. Email: [email protected] Amirhossein Sahebkar, Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran. Email: [email protected]; [email protected] Abstract Cancer is the second cause of death worldwide. Chemotherapy and radiotherapy are the most common modalities for the treatment of cancer. Experimental studies have shown that inflammation plays a central role in tumor resistance and the incidence of several side effects following both chemotherapy and radiotherapy. Inflammation resulting from radiotherapy and chemotherapy is responsible for adverse events such as dermatitis, mucositis, pneumonitis, fibrosis, and bone marrow toxicity. Chronic inflammation may also lead to the development of second cancer during years after treatment. A number of antiinflammatory drugs such as nonsteroidal antiinflammatory agents have been proposed to alleviate chronic inflammatory reactions after radiotherapy or chemotherapy. Curcumin is a welldocumented herbal antiinflammatory agents. Studies have proposed that curcumin can help management of inflammation during and after radiotherapy and chemotherapy. Curcumin targets various inflammatory mediators such as cyclooxygenase2, inducible nitric oxide synthase, and nuclear factor κB (NFκB), thereby attenuating the release of proinflammatory and profibrotic cytokines, and suppressing chronic production of free radicals, which culminates in the amelioration of tissue toxicity. Through modulation of NFκB and its downstream signaling cascade, curcumin can also reduce angiogenesis, tumor growth, and metastasis. Low toxicity of curcumin is linked to its cytoprotective effects in normal tissues. This protective action along with the capacity of this phytochemical to sensitize tumor cells to radiotherapy and chemotherapy makes it a potential candidate for use as an adjuvant in cancer therapy. There is also evidence from clinical trials suggesting the potential utility of curcumin for acute inflammatory reactions during radiotherapy such as dermatitis and mucositis. KEYWORDS cancer, chemotherapy, curcumin, inflammation, radiotherapy J Cell Physiol. 2018;113. wileyonlinelibrary.com/journal/jcp © 2018 Wiley Periodicals, Inc. | 1

Upload: others

Post on 28-Feb-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

Received: 13 August 2018 | Accepted: 27 August 2018

DOI: 10.1002/jcp.27442

R EV I EW ART I C L E

Curcumin as an anti‐inflammatory agent: Implicationsto radiotherapy and chemotherapy

Bagher Farhood1 | Keywan Mortezaee2 | Nasser Hashemi Goradel3 |Neda Khanlarkhani4 | Ensieh Salehi4 | Maryam Shabani Nashtaei4,5 | Masoud Najafi6 |Amirhossein Sahebkar7,8

1Departments of Medical Physics and

Radiology, Faculty of Paramedical Sciences,

Kashan University of Medical Sciences,

Kashan, Iran

2Department of Anatomy, School of Medicine,

Kurdistan University of Medical Sciences,

Sanandaj, Iran

3Department of Medical Biotechnology,

School of Advanced Technologies in Medicine,

Tehran University of Medical Sciences,

Tehran, Iran

4Department of Anatomy, School of Medicine,

Tehran University of Medical Sciences,

Tehran, Iran

5Department of Infertility, Shariati Hospital,

Tehran University of Medical Sciences,

Tehran, Iran

6Department of Radiology and Nuclear

Medicine, School of Paramedical Sciences,

Kermanshah University of Medical Sciences,

Kermanshah, Iran

7Neurogenic Inflammation Research Center,

Mashhad University of Medical Sciences,

Mashhad, Iran

8Biotechnology Research Center,

Pharmaceutical Technology Institute, School of

Pharmacy, Mashhad University of Medical

Sciences, Mashhad, Iran

Correspondence

Masoud Najafi, Department of Radiology and

Nuclear Medicine, School of Paramedical

Sciences, Kermanshah University of Medical

Sciences, Kermanshah 6719851351, Iran.

Email: [email protected]

Amirhossein Sahebkar, Biotechnology

Research Center, Pharmaceutical Technology

Institute, Mashhad University of Medical

Sciences, Mashhad 9177948564, Iran.

Email: [email protected];

[email protected]

Abstract

Cancer is the second cause of death worldwide. Chemotherapy and radiotherapy are

the most common modalities for the treatment of cancer. Experimental studies have

shown that inflammation plays a central role in tumor resistance and the incidence of

several side effects following both chemotherapy and radiotherapy. Inflammation

resulting from radiotherapy and chemotherapy is responsible for adverse events such

as dermatitis, mucositis, pneumonitis, fibrosis, and bone marrow toxicity. Chronic

inflammation may also lead to the development of second cancer during years after

treatment. A number of anti‐inflammatory drugs such as nonsteroidal anti‐inflammatory agents have been proposed to alleviate chronic inflammatory reactions

after radiotherapy or chemotherapy. Curcumin is a well‐documented herbal anti‐inflammatory agents. Studies have proposed that curcumin can help management of

inflammation during and after radiotherapy and chemotherapy. Curcumin targets

various inflammatory mediators such as cyclooxygenase‐2, inducible nitric oxide

synthase, and nuclear factor κB (NF‐κB), thereby attenuating the release of

proinflammatory and profibrotic cytokines, and suppressing chronic production of

free radicals, which culminates in the amelioration of tissue toxicity. Through

modulation of NF‐κB and its downstream signaling cascade, curcumin can also reduce

angiogenesis, tumor growth, and metastasis. Low toxicity of curcumin is linked to its

cytoprotective effects in normal tissues. This protective action along with the

capacity of this phytochemical to sensitize tumor cells to radiotherapy and

chemotherapy makes it a potential candidate for use as an adjuvant in cancer

therapy. There is also evidence from clinical trials suggesting the potential utility of

curcumin for acute inflammatory reactions during radiotherapy such as dermatitis

and mucositis.

K E YWORD S

cancer, chemotherapy, curcumin, inflammation, radiotherapy

J Cell Physiol. 2018;1–13. wileyonlinelibrary.com/journal/jcp © 2018 Wiley Periodicals, Inc. | 1

Page 2: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

1 | INTRODUCTION

After cardiovascular diseases, cancer is the second cause of death in

worldwide (Siegel, Miller, & Jemal, 2015). Each year several million

people undergo different modalities for treatment of cancer (Miller

et al., 2016). The widest modalities for cancer therapy are

chemotherapy and radiotherapy. Although, immunotherapy is the

most interesting modality for the eradication of tumor cells, it need

to more studies for development and confirmation of new drugs

(Kang, Demaria, & Formenti, 2016). Yet, radiotherapy and che-

motherapy are the more common compared with immunotherapy for

cancer therapy, especially in countries with low income (Bazargani,

de Boer, Schellens, Leufkens, & Mantel‐Teeuwisse, 2014). In spite of

beneficial role of these modalities for cancer treatment, there are

some concerns related to early and late side effects of them that may

affect quality of life of patients that undergo chemotherapy and

radiotherapy (Najafi, Motevaseli, et al., 2018). Emerging evidence

show that inflammation caused by radiotherapy and chemotherapy

has a central role for the development of various side effects that

may appear during or after treatment (Yahyapour, Motevaseli, et al.,

2018). Also, inflammation can significantly affect therapeutic out-

come of radiotherapy and chemotherapy (Barker, Paget, Khan, &

Harrington, 2015). Acute inflammation may lead to some severe

reactions in normal tissues that have high sensitivity or are under

expose to massive doses of ionizing radiation. Inflammation in some

of organs, such as tongue and gastrointestinal system, lead to

mucositis that potently affect the quality of life of patients. However,

inflammatory responses in some organs like lung may lead to acute

pneumonitis or fibrosis that threat life of patients (Cheki et al., 2018;

Yahyapour, Amini, Rezapoor, et al., 2018). Dermatitis is a common

side effect of radiotherapy that is resulting from damage to basal

layers of skin (Hymes, Strom, & Fife, 2006). An addition to severe

side effects, chronic inflammation has a potent link to carcinogenesis

(Farhood et al., 2018).

There is a growing interest in traditional medicine‐based therapy

for several diseases including inflammatory diseases. This is because of

low toxicity and lower side effects compared with chemical anti‐inflammatory drugs. The most of anti‐inflammation drugs such as

nonsteroidal anti‐inflammatory drugs (NSAIDs) may lead to increased

risk of cardiovascular diseases and also problems in calcium absorption

(Stock, Groome, Siemens, Rohland, & Song, 2008). Curcumin is the

Asian yellow spice, which is derived from the roots of the Curcuma

longa. Several studies have shown that curcumin can be proposed as a

promising anti‐inflammatory agent (Basnet & Skalko‐Basnet, 2011;Sahebkar, Cicero, Simental‐Mendia, Aggarwal, & Gupta, 2016). By

contrast to NSAIDs, curcumin not only has no a serious side effect, it

can reduce risk of cardiovascular disorders (Qadir, Naqvi, &

Muhammad, 2016). Curcumin has shown promising properties that

are interesting for patients with cancer. In experimental studies, it has

shown is able to ameliorate toxic effects of chemotherapy and

radiotherapy. Antioxidant effect of curcumin help to scavenging of free

radicals which are produced by ionizing radiation and some

chemotherapeutic agents such as cyclophosphamide (Hatcher, Planalp,

Cho, Torti, & Torti, 2008). This is associated with reduced chromosome

aberrations and genomic instability, which is a hallmark of second

primary cancers (Hatcher et al., 2008). Also, curcumin through

modulation several signaling pathways reduces appearance of several

early and late side effects of ionizing radiation and chemotherapy

agents (Bar‐Sela, Epelbaum, & Schaffer, 2010). In this review, we

aimed to explain molecular anti‐inflammation properties of curcumin

in cancer treatment with radiotherapy and chemotherapy.

2 | INFLAMMATION IN CANCER

In addition to tumor cells, a tumor is consisting of a mixture of

different types of cells such as immune cells and some other

nonmalignant cells like fibroblasts, endothelial, and epithelial cells.

The response of tumor cells to radiotherapy, chemotherapy, or

immunotherapy is highly depending to these cells. These cells

together to tumor cells make an environment named tumor

microenvironment (Junttila & de Sauvage, 2013). Inflammatory cells

including macrophages, lymphocytes, and dendritic cells play a key

role in response of tumor, as well as progression of it following

therapy (Jain, 2013). Although, these cells can eradicate tumor cells,

emerging evidence show that inflammatory responses by these cells

play a key role in tumor growth and angiogenesis (Kershaw, Devaud,

John, Westwood, & Darcy, 2013; Liyanage et al., 2002).

Following tumor exposure to therapeutic agents such as

radiation or chemotherapy, a large number of cells undergo death

through different mechanisms, including apoptosis, mitotic cata-

strophe, necrosis, autophagy, and senescence. Among them,

apoptosis and necrosis have pivotal role in the balance between

inflammation and tolerogenic responses. Apoptotic bodies may

digest by macrophages and do not able to stimulate inflammatory

responses by lymphocytes or dendritic cells. However, necrosis

cause release of several danger molecules that are able to induce

inflammation. Also, a high rate of cell death by apoptosis may

overwhelm this type of death of cells, leading to necroptosis, a

phenomenon that apoptosis followed by necrosis. Similar to

necrosis, necroptosis is able to stimulate inflammatory responses

(Yahyapour, Amini, Rezapour, et al., 2018). Inflammatory mediators

play a key role in the angiogenesis and growth of cancers. Nuclear

factor κB (NF‐κB) is regarded as one of the most important links

between cells death, inflammation, and tumor resistance. It has

been shown that cisplatin stimulate upregulation of NF‐κB via PI3/

Akt‐signaling cascade in human ovary cancer cells (Ohta et al.,

2006). Clinical studies also showed that chemotherapy by cisplatin

cause increased expression of NF‐κB in ovarian cancer patients

(Annunziata et al., 2010).

NF‐κB via upregulation of cyclooxygenase‐2 (COX‐2) stimulates

release of prostaglandins and resistance of tumor cells to apoptosis.

Also, it induces vascular endothelial growth factor (VEGF), which

promotes development of new vascular. Signal transducer and

activator of transcription 3 (STAT‐3) and hypoxia‐inducible factor 1

are other inflammation mediators that are involved in tumor

2 | FARHOOD ET AL.

Page 3: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

resistance through stimulation of cell proliferation and resistance to

apoptosis. It seems that upregulation of TLR‐4 is involved in

stimulation of NF‐κB and resistance to chemotherapy (Rajput, Volk‐Draper, & Ran, 2013). Inhibition of NF‐κB has proposed for

increasing efficiency of radiotherapy and chemotherapy drugs such

as paclitaxel (Mabuchi et al., 2004; Yahyapour et al., 2017).

3 | INFLAMMATION OF NORMAL TISSUESFOLLOWING RADIOTHERAPY

Inflammation is responsible for several side effects of exposure to

ionizing radiation in normal tissues. Studies have shown that chronic

inflammation that is a common side effect of radiotherapy can induce

genomic instability through stimulation of free‐radical production and

inhibition of DNA repair pathways (Najafi, Cheki, et al., 2018). Also,

chronic inflammation can appear as several side effects in different

organs, such as pneumonitis and fibrosis in the lung, dermatitis in the

skin, enteritis in intestine, proctitis in colon, edema in muscles, and so on

(Yahyapour, Shabeeb, et al., 2018). The origin of radiation‐inducedinflammation is related to DNA damage and cell death. Massive damage

to nucleus DNA by ionizing radiation interactions or free radicals may

lead to cells death because of overwhelm of DNA repair mechanisms.

Immunogenic and tolerogenic types of cells death lead to release

various contents of cells including damage‐associated molecular

patterns (DAMPs). DAMPs including uric acid, heat‐shock proteins,

high‐mobility‐group box 1, and so on, can recognized by some receptors

on the surface of macrophages and dendritic cells, which are named toll‐like receptors (TLRs). TLRs facilitate the response of lymphocytes to

danger alarms. This lead to upregulation of inflammatory mediators

such as NF‐κB, intracellular adhesion molecules (ICAM), STATs, and

some other, which lead to secretion of various inflammatory cytokines

(Najafi, Motevaseli, et al., 2018).

Abnormal increased level of inflammatory cytokines leads to

various side effects in different organs. Pneumonitis in the lung is

resulting from a massive release of interleukin‐1 (IL‐1), IL‐6, IL‐8,tumor necrosis factor α (TNF‐α), and interferon γ ( IFN‐γ). Thesecytokines also via regulation of some pro‐oxidant and profibrotic

enzymes stimulate accumulation of collagen leading to fibrosis.

Both pneumonitis and fibrosis are serious side effects that may

lead to death of patients with cancer. This issue has observed for

patients with chest cancer, as well as other cancers with lung

metastasis (Yahyapour, Shabeeb, et al., 2018). Inflammasome is a

complex that is involved in secretion of IL‐1 following exposure of

cells to ionizing radiation. Mitochondria injury by ionizing

radiation stimulate upregulation of inflammasome (Abderrazak

et al., 2015). Experimental studies have revealed that this

pathway is involved in radiation‐induced mucositis in the tongue

and intestine (Fernández‐Gil et al., 2017; Ortiz et al., 2015). Also,

it seems that dermatitis following skin irradiation has a potent

relation to inflammasome (Favero, Franceschetti, Bonomini,

Rodella, & Rezzani, 2017). Experimental studies show that

pericarditis in heart play a key role in the development of cardiac

disorders in patients with chest cancer. Studies have proposed

that an abnormal increase in the level of some cytokines such as

IL‐1 and TGF‐β play a key role in late effects of ionizing radiation

on cardiac function (Boerma et al., 2016; Eldabaje, Le, Huang, &

Yang, 2015). Increased inflammatory responses and subsequent

chronic production of free radicals following exposure to ionizing

radiation have observed for bone marrow, vascular, brain, joints,

mammary cells, and others (Robbins & Zhao, 2004).

4 | INFLAMMATION OF NORMAL TISSUESFOLLOWING CHEMOTHERAPY

Inflammation plays a key role in the appearance of side effects of

chemotherapy drugs. It is proposed that inflammation caused by

chemotherapy drugs may lead to stimulation of metastasis, cancer

relapse, and even fail of cancer treatment (Demaria et al., 2017;

Vyas, Laput, & Vyas, 2014). Moreover, some evidence show that

inflammation may is involved in some behavioral changes such as

cognitive problems, fatigue, and neuropathy (Vichaya et al., 2015).

It has been shown that NF‐κB and TNF‐α play a key role in

nephrotoxicity following injection of cisplatin to rats (Hagar,

Medany, Salam, Medany, & Nayal, 2015). TNF‐α and its down-

stream signaling such as TNFR1 and p38 have key role for

induction of apoptosis and necrosis in tubular cells following

exposure to chemotherapy agents (Luo et al., 2008; Ramesh &

Reeves, 2003). It seems that it through activation of caspase‐3pathway is involved in cisplatin nephrotoxicity. Suppression of

this mediators showed that attenuate cisplatin‐induced nephro-

toxicity (Arjumand, Seth, & Sultana, 2011). In addition, it is

proposed that cisplatin through stimulation of COX‐2 upregula-

tion is involved in appearance of nephrotoxicity (Domitrović et al.,

2013). Zhou et al. showed that injection of cisplatin to mice lead

to infiltration of inflammatory cells including macrophages and

lymphocytes in kidneys. Moreover, their results showed a

significant increase in the level of inflammatory cytokines

including IL‐1, IL‐6, and TNF‐α. This study showed that inhibition

of apoptosis pathway plays a key role in nephrotoxicity induced

by cisplatin (J. Zhou et al., 2017). Paclitaxel and methotrexate are

other major chemotherapy agents used in clinical oncology.

Experimental studies showed that they are able to induce

oxidative injury and increase of inflammatory cytokines such as

IL‐1, IL‐6, TNF‐α, and IL‐8 (Ibrahim, El‐Sheikh, Khalaf, & Abdelrah-

man, 2014; Pusztai et al., 2004). Lian et al. evaluated irinotecan

(CPT‐11) effect on inflammation pathway in mice intestine. They

showed that DNA damage by CPT‐11 lead to release of exosome

secretion and stimulation of innate immune responses. This lead

to secretion of IL‐1 and IL‐13 through stimulation of inflamma-

some pathway (Lian et al., 2017).

Activation of redox system and chronic oxidative stress

through a chronic inflammatory responses play central role for

chemotherapy‐induced tissues injury. El‐Naga (2014) showed that

injection of cisplatin to rat is associated with increased the

FARHOOD ET AL. | 3

Page 4: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

expression of NOX1 and inducible nitric oxide synthase (iNOS) in

kidney, two major reactive oxygen species (ROS) and nitric oxide

(NO) producing enzymes in inflammation conditions. Some other

studies showed increased free‐radical production by mitochondria,

upregulation of heme oxygenase‐1, as well as inhibition of

antioxidant enzymes (Chtourou, Aouey, Aroui, Kebieche, & Fetoui,

2016; Nafees, Rashid, Ali, Hasan, & Sultana, 2015; Ramesh &

Reeves, 2004; Santos et al., 2007). Increased oxidative injury

following administration of chemotherapy drugs such as cisplatin

and cyclophosphamide have confirmed in several studies (Y. Chen,

Jungsuwadee, Vore, Butterfield, & Clair, 2007; Conklin, 2004;

Tomar et al., 2017).

5 | CURCUMIN AS ANANTI ‐ INFLAMMATORY AGENT

Curcumin has been traditionally used for the treatment of several

inflammatory diseases (Aggarwal, Sundaram, Malani, & Ichikawa,

2007). These traditional applications have been translated in

modern pharmacological studies and randomized controlled trials

against a variety of human diseases including cancer (Iranshahi

et al., 2010; Mirzaei et al., 2016; Momtazi et al., 2016; Rezaee,

Momtazi, Monemi, & Sahebkar, 2017), respiratory diseases (Lelli,

Sahebkar, Johnston, & Pedone, 2017; Panahi, Ghanei, Bashiri,

Hajihashemi, & Sahebkar, 2014; Panahi, Ghanei, Hajhashemi, &

Sahebkar, 2016), osteoarthritis (Panahi, Alishiri, Parvin, & Sahebkar,

2016; Panahi, Rahimnia, et al., 2014; Sahebkar & Henrotin, 2016),

nonalcoholic fatty liver disease (Panahi, Kianpour, et al., 2016;

Rahmani et al., 2016; Zabihi, Pirro, Johnston, & Sahebkar, 2017),

and dyslipidemia (Cicero et al., 2017; Ganjali et al., 2017; Panahi,

Kianpour, et al., 2016). In the recent decades, biological studies

showed that it can affect more than 90 inflammatory targets in cells

(H. Zhou, Beevers, & Huang, 2011). Evidence show that curcumin is

able to attenuate the expression of some transcription factors

especially NF‐κB that has a central role for regulation of inflamma-

tion (Shehzad, Rehman, & Lee, 2013). Also, it can attenuate the

metabolism of prostaglandins and lipoxygenases, which are involved

in appearance of inflammatory signs and also lead to production of

free radicals (Aggarwal & Harikumar, 2009). iNOS is another target

for curcumin that it induces nitrative DNA damage and attenuation

of DNA damage response through nitroacetylation of 8‐oxoguanine‐DNA glycosylase 1 (Ogg1), an important modulator of base excision

repair pathway (Onoda & Inano, 2000). Curcumin through inhibition

of inflammatory cytokines such as IL‐1 and TNF‐α can prevent from

several inflammatory diseases (Aggarwal & Harikumar, 2009). Also,

it has antioxidant effects that prevents oxidative damage and

carcinogenesis (López‐Lázaro, 2008). Daily treatment with curcumin

has shown that can reverse reduction of bone marrow cells in

carcinoma‐bearing mice. Curcumin is able to reduces tumor‐inducedhepatic injury, and also is able to reverse reduction of hematopoie-

tic parameters such as total count of immune cells like lymphocytes

(Pal et al., 2005).

6 | PROTECTION AGAINSTINFLAMMATION IN RESPONSETO RADIATION AND CHEMOTHERAPYAGENTS: EXPERIMENTAL STUDIES

As mentioned, inflammation is originated form DNA damage and cell

death, especially necrosis, which is a common type of cell death in

radiotherapy and chemotherapy. It is confirmed that inflammatory

responses to cancer therapy may lead to chronic oxidative stress,

which may lead to damage to the normal function of organs. Also, it

may cause accumulation of unrepaired DNA damage, leading to

genomic instability and cancer (Najafi, Motevaseli, et al., 2018).

Inflammation can induce reduction–oxidation reactions, which itself

play a key role in triggering of inflammation responses. A positive

feedback between inflammation and redox responses lead to some

late side effects such as dermatitis, mucositis, enteritis, pneumonitis,

cardiac injury, and other (Yahyapour, Motevaseli, et al., 2018). As

curcumin has both antioxidant and anti‐inflammation properties, it

has proposed for preventing and treatment of various types of

inflammatory diseases in cancer radiotherapy and chemotherapy

(Verma, 2016).

6.1 | Bone marrow toxicity

Bone‐marrow stem cells are among the most sensitive cells to

ionizing radiation and chemotherapy drugs within the human body

(Wang et al., 2010). Studies have proposed that activation of some

cytokines and proapoptosis pathways are involved in high toxicity of

bone marrow cells to cancer therapy agents (Wang et al., 2010).

Exposure of bone marrow cells to ionizing radiation lead to a

significant increase in the apoptosis induction during some hours

(Meng, Wang, Brown, Van Zant, & Zhou, 2003). This is associated

with elevation of TGF‐β secretion, which can continue for long time

after exposure (Zhang et al., 2013). Chronic upregulation of TGF‐β is

associated with continuous production of ROS and NO by macro-

phages and lymphocytes, as well as some other cells (Anscher, 2010).

TGF‐β through stimulation of some ROS producing enzymes such as

nicotinamide adenine dinucleotide phosphate oxidase, and also via

upregulation of iNOS induces oxidative and nitrative damages in

bone marrow cells (Wang et al., 2010). The redox interactions

between immune mediators and ROS/NO‐producing enzymes may

continue for some weeks after exposure to radiation (Paraswani,

Ghosh, & Thoh, 2017). Bone marrow toxicity also has observed after

treatment with various chemotherapy agents such as cisplatin,

carboplatinum, busulfan (BU), 5‐fluorouracil (5‐FU), cyclophospha-mide, and other (Newman et al., 2016). In addition to oxidative injury,

these drugs may lead to apoptosis, senescence, and activation of

redox interactions (Hassanshahi, Hassanshahi, Khabbazi, Su, &

Xian, 2017).

Curcumin has been shown is able to protect bone marrow cells

against toxic effects of radiation and chemotherapy. X. Chen et al.

showed that curcumin can induce activity of some DNA repair

enzymes and attenuates myelosuppression following injection of

4 | FARHOOD ET AL.

Page 5: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

carboplatin to mice. Results indicated an increase in the expression of

BRCA1, BRCA2, and ERCC1 in the mice bone marrow cells in a

curcumin dose dependent manner. This was associated with

reduction of DNA damage in bone marrow cells (X. Chen et al.,

2017). Curcumin also reduces chromosome aberrations in rats bone

marrow cells following cisplatin injection (Antunes, Araújo, Darin, &

Bianchi, 2000). Zhou et al. showed that combination of curcumin and

mitomycin C reduces the side effects of mitomycin C on bone

marrow cells. They showed that curcumin through inhibition of

glucose regulatory protein (GRP58), a protein which mediate

mitomycin DNA cross link, reduces DNA damage and subsequent

toxicities in bone marrow cells. They showed that inhibition of

GRP58 by curcumin is mediate through extracellular signal‐regulatedkinase (ERK)/p38 MAPK pathway (Q. M. Zhou, Zhang, Lu, Wang, &

Su, 2009).

6.2 | Dermatitis

Dermatitis induced by ionizing radiation is resulting from massive

apoptosis and necrosis of basal cells in derma. These lead to complex‐signaling pathways that lead to appearance of dermatitis with signs

such as redness, pain, dry desquamation, moist desquamation, dry

crusting, ulcers, and so forth. Studies have shown that upregulation

of various inflammatory mediators such as COX‐2, NF‐κB and

inflammasome, cytokines such as IL‐1, IL‐6, IL‐8, TNF‐α, and TGF‐β,and also infiltration of mast cells and T cells are involved in

appearance of dermatitis following exposure to ionizing radiation

(J.‐S. Kim et al., 2015; J. H. Lee et al., 2009; Müller & Meineke, 2007).

Curcumin has shown promising effects for mitigation of radiation‐induced skin injury (Jagetia & Rajanikant, 2005). Okunieff et al.

evaluated protective effect of curcumin on radiation‐induced skin

toxicity in mice. They irradiated hind part of mice legs with a single

50 Gy radiation and treated mice with 50, 100, or 200mg/kg

curcumin before and after irradiation. Skin toxicity were evaluated

during 2–3 weeks for evaluating acute dermatitis, and 90 days after

irradiation for chronic skin damage. Results showed that adminis-

tration of 200mg/kg curcumin can attenuate pathological appear-

ance of dermatitis in both times. Also, results indicated that

amelioration of dermatitis was related to suppression of IL‐1β,IL‐1Ra1, IL‐6, and IL‐18 (Okunieff et al., 2006). Kim et al. used a

cream containing curcumin at 200mg/cm2 in a mini‐pig model. Pigs

were irradiated locally with 50 Gy cobalt‐60 γ rays and then treated

with cream containing curcumin twice daily for 35 days. Results

showed a significant improvement in wound healing associated with

suppression of COX‐2 and NF‐κB (J. Kim et al., 2016).

6.3 | Mucositis

Mucositis is one of the most common side effects of radiotherapy,

which affects the mucosa. This complication may appear in oral for

head and neck cancers, and also in the gastrointestinal system. This

complication may appear as acute inflammation, pain, and ulceration

(Peterson, Bensadoun, Roila, & On behalf of the EGWG, 2011;

Ps, Balan, Sankar, & Bose, 2009). So far, some experimental studies

have conducted to evaluate protective effect of curcumin against

radiation‐induced mucositis. Rezvani et al. showed that treatment of

rats with curcumin reduces oral mucositis with a dose reduction factor

equal to 9%. Rats received curcumin at 200mg·kg−1·day−1 following

irradiation with 13.5–18Gy radiation (Rezvani & Ross, 2004).

Inhibition of inflammatory mediators such as NF‐κB and protein

kinase B (Akt) has proposed for protective effect of curcumin in the

intestine. In an animal study Rafiee et al. showed that a low dose of

ionizing radiation (1–5Gy) cause upregulation of NF‐κB and PI3K/Akt

pathways in human intestinal microvascular endothelial cells. Also,

they showed that treatment of rats with curcumin can attenuate

edema in endothelial cells. However, irradiation or curcumin treatment

did not cause any effect on regulation of mouse double minute 2

homolog (MDM2; Rafiee et al., 2010). Administration of curcumin

45mg·kg−1·day−1 for 2 weeks before irradiation has shown can

attenuate histopathological damages such as oxidative injury and

accumulation of fibroblasts (El‐Tahawy, 2009). Similar results were

obtained by another study by Fukuda et al. (2016).

6.4 | Pneumonitis and lung fibrosis

Pneumonitis in the lung is resulting from chronic upregulation several

inflammatory mediators, chemokines and cytokines, and transcrip-

tion factors. These lead to the accumulation of inflammatory cells

such as macrophages, mast cells, and lymphocytes. Interactions

between inflammatory cytokines with macrophages and lymphocytes

cause the continuous production of free radicals, including ROS and

NO. chronic oxidative stress following inflammatory responses

stimulate upregulation of profibrotic cytokines such as TGF‐β, andalso growth factors such as epithelial growth factor receptor (EGFR),

connective tissue growth factor (CTGF) and platelet‐derived growth

factor. Pneumonitis appears some months after radiotherapy, while

fibrosis may appear years later (Proklou, Diamantaki, Pediaditis, &

Kondili, 2018).

Curcumin as a potent immune modulator agent has shown is

able to modulate radiation responses in the lung tissue. Lee et al. in

an experimental study evaluated the radioprotective effect of

curcumin on oxidative injury and fibrosis in the mice lung tissue.

Mice were treated with a diet containing 5% curcumin for 2 weeks

and then irradiated with 13.5 Gy X‐ray. Results showed that

treatment with curcumin attenuate production of ROS by

pulmonary endothelial cells following irradiation. Also, curcumin

diet caused significant reduction of fibrosis and increased survival,

while it could not ameliorate pneumonitis (J. C. Lee et al., 2010). In

another study has been used from 200 mg/kg curcumin before to 8

weeks after irradiation. Results showed that curcumin treatment

can attenuate the upregulation of profibrotic genes including

TGF‐β1 and CTGF, alleviate inflammatory mediators including

TNFR1 and COX‐2, and also suppresses NF‐κB. These were

associated with attenuation of inflammatory cells accumulation,

edema, alveolar and vascular injury, as well as collagen deposition

(Cho et al., 2013).

FARHOOD ET AL. | 5

Page 6: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

7 | ANTI ‐ INFLAMMATORY EFFECTOF CURCUMIN ON CANCER

Curcumin can induce apoptosis in some cancerous cells (Noorafshan

& Ashkani‐Esfahani, 2013). Treatment of human prostate cancer cell

line LNCaP with curcumin can induce apoptosis through upregulation

of proapoptosis genes (Deeb et al., 2003). Similarity, curcumin can

induce apoptosis in HepG2 cells, which it seems is resulting from

effects on mitochondrial function and increased superoxide produc-

tion (Cao et al., 2007). In addition to death signal pathways, curcumin

has shown that can attenuate inflammatory cytokines, which are

involved in the proliferation and differentiation of cancer stem cells.

Curcumin has shown that through suppression of some transcription

factors such as activator protein‐1, NF‐κB and STAT‐3, phosphodies-terases, some cytokines such as IL‐1, IL‐6, IL‐8, and also chemokine

receptors such as CXCR1 and CXCR2 can suppress differentiation of

cancer stem cells (Abusnina et al., 2015; C. Chen, Liu, Chen, & Xu,

2011; Deguchi, 2015; Sordillo & Helson, 2015; Teymouri, Barati,

Pirro, & Sahebkar, 2018). Suppression of Wnt/β‐catenin and Sonic

hedgehog pathways, and also some microRNAs by curcumin can

suppress cancer stem cells (Y. Li & Zhang, 2014; Zhu et al., 2017).

Curcumin has an inhibitory effect on angiogenesis and metastasis

factors such as angiopoetin‐1 and VEGF, and also matrix metallo-

proteinase‐3 (MMP‐3) that may be useful for suppression of tumor

growth (Boonrao, Yodkeeree, Ampasavate, Anuchapreeda, & Lim-

trakul, 2010; W. Li et al., 2018; Qadir et al., 2016; Ramezani,

Hatamipour, & Sahebkar, 2018; Saberi‐Karimian et al., 2017; Shakeri,

Ward, Panahi, & Sahebkar, 2018; You et al., 2017).

7.1 | Synergistic effect of curcumin with radiation

In addition to protection of normal tissues, sensitization of tumor

cells can cause decreases in demanded radiation dose, so as to

reduce the risk of normal tissues reactions and also secondary cancer

risk especially in the young people. So far, some agents have used for

sensitization of tumor cells to radiotherapy. However, severe toxicity

of these agents may lead to several side effects for patients. It has

been proposed that targeting of inflammation can attenuate normal

tissues injury, as well as sensitize tumor cells to ionizing radiation. On

the other hand, potent anti‐inflammation properties of curcumin can

be proposed for radiosensitization and radioprotection. As men-

tioned, curcumin is able to suppress several inflammation mediators

that are activated following exposure to ionizing radiation. As some

of inflammatory mediators are able to induce angiogenesis and

proliferation of cancer cells, inhibition of them by curcumin can

attenuate tumor cells growth and repopulation during radiotherapy

(Verma, 2016).

Curcumin has shown is able to sensitize cancer cells to ionizing

radiation through induction of proapoptosis and attenuation and

antiapoptosis genes. Qiao et al. showed that curcumin reduces the

expression of NF‐κB in human Burkitt’s lymphoma cells, leading to

increasing apoptosis induction in these cells. They showed that PI3K/

Akt pathway inhibition by curcumin is responsible for attenuation of

NF‐κB regulation (Qiao, Jiang, & Li, 2013). This was associated with

cell cycle arrest in G2‐M, which is more sensitive to kill by ionizing

radiation (Calaf, Echiburu‐Chau, Wen, Balajee, & Roy, 2012; Qiao,

Jiang, & Li, 2012). Similar results were obtained for nasopharyngeal

carcinoma (Pan et al., 2013).

Suppression of NF‐κB by curcumin has shown attenuate down-

stream genes such as COX‐2 and VEGF, which cause inhibition of

tumor growth. A study showed that treatment of tumor‐bearing mice

with curcumin that were exposed to radiation lead to 50% reduction

in COX‐2 and VEGF, and remarkable tumor regrowth delay in

xenograft colorectal cancer (Kunnumakkara et al., 2008). Similar

results were observed for HepG2, rhabdomyosarcoma, human oral

squamous cell carcinoma, and breast cancer cells (Chiang et al., 2014;

Gallardo & Calaf, 2016; Hsu, Liu, Liu, & Hwang, 2015; Orr et al.,

2013). In addition to inhibition of angiogenesis factors, curcumin has

shown attenuate telomerase activity, which itself is depend to NF‐κBactivity. Combination of curcumin with radiation has shown that

reduces the expression of telomerase reverse transcriptase (TERT)

gene in human neuroblastoma cells (Aravindan, Veeraraghavan,

Madhusoodhanan, Herman, & Natarajan, 2011). This property can

reduce survival of irradiated cells. Chendil, Ranga, Meigooni,

Sathishkumar, and Ahmed (2004) proposed that inhibitory effect of

curcumin on TNF‐α is involved in NF‐κB suppression and radio-

sensitization of PC3 cells. Also, in response to radiation, curcumin is

able to phosphorylate IkB directly, an inhibitor of NF‐κB (Orr et al.,

2013; Sandur et al., 2009). Inhibition of EGFR is another mechanism

for curcumin that may be involved in radiosensitization of cancer

cells (Khafif et al., 2009). Also, it is proposed that curcumin through

upregulation of ERK1/2 amplifies the production of ROS following

exposure of cancer cells to radiation (Javvadi, Segan, Tuttle, &

Koumenis, 2008).

Curcumin has shown that through inhibition of PI3K suppresses

the regulation of MDM2, a suppresser of p53. Suppression of MDM2

can increase activity of p53, which is necessary for initiation of

apoptosis in cancerous cells. Curcumin through modulation of this

pathway has shown sensitize PC3 cells to ionizing radiation and

chemotherapy (M. Li, Zhang, Hill, Wang, & Zhang, 2007). These

results were confirmed by Veeraraghavan, Natarajan, Herman, and

Aravindan (2010), which showed that curcumin induces apoptosis in

sarcoma cells through modulation of p53 and other related genes

such as p21 and Bax.

7.2 | Synergistic effect of curcumin withchemotherapy drugs

Several in vitro, animal and clinical studies have shown that curcumin

has a synergistic effect with some chemotherapy agents such as

bevacizumab, 5‐FU, and oxaliplatin (FOLOX; Anitha, Deepa, Chennazhi,

Lakshmanan, & Jayakumar, 2014; Anitha, Sreeranganathan, Chennazhi,

Lakshmanan, & Jayakumar, 2014; Irving et al., 2015; James et al., 2015;

Yue et al., 2016). Combination of curcumin with chemotherapy drugs

has shown is able to attenuate inflammation and oxidative signs

associated with tumor growth (Marjaneh et al., 2018). Sen et al. showed

6 | FARHOOD ET AL.

Page 7: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

that curcumin through targeting of NF‐κB help to antitumor effect of an

adjuvant chemotherapeutic drug. This study showed doxorubicin cause

degradation of Iκbα, leading to activation of NF‐κB and resistance of

Ehrlich ascites carcinoma cells, which injected to mice. Treatment of

mice with 50mg/kg curcumin lead to significant reduction of tumor

cells viability. This was associated with inhibition of nuclear transloca-

tion of NF‐κB and suppression of B‐cell lymphoma 2 (Bcl‐2), as well as,

induction of proapoptosis genes such as p53, Bax, p53 upregulated

modulator of apoptosis (PUMA), and phorbol‐12‐myristate‐13‐acetate‐induced protein 1 (NOXA) (Sen et al., 2011). Another study by Yang

et al. (2017) showed that inhibition of NF‐κB and COX‐2 by curcumin

enhance the cytotoxic effect of 5‐FU against gastric cancer MKN45 and

AGS cells. Similar results showed for esophageal squamous cell

carcinoma, breast, and colorectal cancer cells (Shakibaei et al., 2013;

Tian, Fan, Zhang, Jiang, & Zhang, 2012; Vinod et al., 2013). It is

confirmed that NF‐κB mediate tumor resistance through upregulation

of antiapoptosis factors, including Bcl‐2 (Bava et al., 2011).

In addition to NF‐κB, curcumin combination with chemotherapy

drugs has shown is able to sensitizes cells through inhibition growth

factors. VEGF and EGFR are important genes that play a central role

in tumor growth. However, it seems that regulation of these growth

factors are highly depend to NF‐κB too. Curcumin has shown inhibits

VEGF, ICAM‐1, MMP‐9, and other inflammatory mediators in

colorectal cancer cells in response to capecitabine (Kunnumakkara

et al., 2009). Another study showed that curcumin in combination of

5‐FU and doxorubicin through inhibition of EGFR‐ERK1/2 signaling

can attenuate proliferation of head and neck squamous cell

carcinoma (Sivanantham, Sethuraman, & Krishnan, 2016).

8 | WHAT IS THE EVIDENCE FROMCLINICAL TRIALS?

In addition to experimental studies, some clinical trials showed that

curcumin can be proposed for amelioration of side effects of cancer

therapy such as inflammatory disorders. Dermatitis and mucositis are

two inflammatory side effects of cancer therapy that proposed as

target diseases for curcumin. Dermatitis is one the most common

side effects of radiotherapy for different types of cancers. It is

estimated that 95% patients with breast cancer that undergo

radiotherapy show signs of dermatitis. Palatty et al. evaluated

possible protective effect of a cream containing curcumin and sandal

wood oil for patients with head and neck cancer that undergo

radiotherapy. The first pilot study showed promising results. They

showed that this cream can alleviate Grade 3 of dermatitis of

radiotherapy patients (Palatty et al., 2014). Using this cream showed

that it is able to reduce signs of dermatitis during Weeks 2 and 3.

Patients in this study received a total dose equal 50 Gy (2 Gy/day,

five times per week) for 5 consecutive weeks (Rao et al., 2017). Ryan

et al. (2013) evaluated therapeutic effect of curcumin on radiation‐induced dermatitis in a double‐blind clinical trial study including

30 patients with breast cancer. Patients received 6 g daily curcumin

or placebo as orally during course of radiotherapy. Results showed TABLE

1Su

mmaryofstudiesreportingan

ti‐in

flam

matory

effectsofcu

rcumin

innorm

alcells

Route/ce

lllin

eTarge

tModality

Dosage

/durationofcu

rcumin

Majorfindings

Referen

ces

Mice

Bonemarrow

Chem

otherap

y(carboplatin)

0.1ml5mM·m

ouse

−1·day

−1

Atten

uation

ofchromosom

eab

erration

san

dmyelosupp

ression,

andup

regu

lation

ofBRCA1,B

RCA2,and

ERCC1

(Chen

etal.,2017)

Rat

Bonemarrow

Chem

otherap

y(cisplatin)

8mg/kg

for2day

sRed

uctionofch

romosomeab

errations

(Antunes

etal.,2000)

Mice

Bonemarrow

Chem

otherap

y(m

itomycin

C)

100mg/kg

for4wee

ksInhibitingDNA

cross‐linking

(Zhouet

al.,2009)

Mice

Lung

Rad

iation

5%

indietstartingfrom

2

wee

ksbefore

Red

uctionofROSproductionbyen

dothelialcells,

attenuationofinflam

mationan

dfibrosis

(Lee

etal.,2010)

Rats

Tongu

esRad

iation

200mg·kg

−1·day

−1foren

dof

study

Red

uctionofmuco

sitisby9%

(Rezva

nian

d

Ross,2

004)

Invitro,

rat

Human

intestinal

microva

scular

endothelialcells/ratsintestine

Rad

iation

10μM

,2%

ofdaily

diet

Atten

uationofNF‐κBan

dPI3K/A

kt/m

TORsign

aling,

alleviationofed

emain

endothelialcells

(Rafieeet

al.,2010)

Mice

Intestine

Rad

iation

0.5%

(wt/wt)

Red

uctionofap

optosis,increa

sednumbersofvilli

(Fuku

daet

al.,2016)

Mice

legs

Rad

iation

200mg/kg

Atten

uationofdermatitis,suppressionofinflam

matory

cytokines

includingIL‐1β,

IL‐1Ra1

,IL‐6,an

dIL‐18

(Oku

nieffet

al.,2006)

Pig

Rad

iation

Curcumin

crea

mat

200mg/

cm2tw

icedaily

for35day

s

Improve

men

tin

woundhea

ling,

inhibitionofCOX‐2

and

NF‐κB

(Kim

etal.,2016)

Note.Akt:p

rotein

kinaseB;C

OX‐2:cyclooxy

genase‐2;IL‐1β:

interleu

kin‐1β;

NF‐κB:n

uclea

rfactorκB

;PI3K:p

hosphoinositide3‐kinase;

mTOR:m

ammaliantarget

ofrapam

ycin;R

OS:

reactive

oxy

genspecies.

FARHOOD ET AL. | 7

Page 8: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

that curcumin administration do not able to attenuate redness but it

causes significant reduction of dermatitis and moist desquamation.

However, reduction of dermatitis was observed after fifth weeks

(Ryan et al., 2013). Another clinical trial study on 686 patients

showed that administration of 2 g/day cannot attenuate dermatitis in

breast cancer patients that undergo radiotherapy (Ryan Wolf et al.,

2018). It seems that the main difference between these studies is

resulting from dose of curcumin that patients received.

In a pilot study comprising 20 patients with cancer who

underwent radiochemotherapy, the efficacy of curcumin in amelior-

ating oral mucositis was tested. Results showed that administration

of curcumin attenuated mucositis and improved wound healing (Patil,

Guledgud, Kulkarni, Keshari, & Tayal, 2015). Curcumin also attenu-

ated chemotherapy‐induced mucositis in pediatric patients (Elad

et al., 2013). In a clinical study comprising >200 patients with head

and neck cancers, the incidence of mucositis following chemora-

diotherapy with or without curcumin treatment was evaluated.

Administration of curcumin was started 3 days before radiotherapy

at a dose of 2 g/day, and was continued for 3 months. Curcumin

treatment delayed the initiation of mucositis by 7 days and reduced

mean duration of it by 20 days. The most obvious reduction of

mucositis duration was for Grades 3 and 4 by 22 days. Moreover,

treatment with curcumin reduced the incidence of mucositis from

89% to 51% (Adhvaryu & Reddy, 2018). In another study by Rao et al.

the efficacy of curcumin in reducing mucositis in patients with head

and neck cancer who underwent radiotherapy or radiochemotherapy

was evaluated. Patients were visited each week for a 7‐week follow‐up. Patients who received chemoradiation first received carboplatin

(1 dose/week). Radiotherapy was planned to provide 70Gy radiation

dose to tumor. Patients received a solution containing 300mg

curcumin at 6 doses/day during the treatment course. Results

showed that radiotherapy caused a significant mucositis from the

first week to the end of radiotherapy. Treatment with curcumin

showed delayed and reduced mucositis at all weeks (Rao et al., 2014).

9 | CONCLUSION

As mentioned in the above sections, curcumin possesses interesting

properties for the amelioration of inflammatory complications of

radiotherapy and chemotherapy. Inflammation is involved in acute

reactions to ionizing radiation and also chemotherapy drugs in

various organs such as bone marrow, lung, heart, and gastrointestinal

system. In addition to toxic effects on normal tissues, a large number

of studies have revealed pivotal role of inflammatory mediators in

tumor resistance and decreased efficiency of chemotherapy. Inflam-

mation can induce chronic upregulation of several cytokines and

continuous production of free radicals, leading to genomic instability,

pain, ulcer, and fibrosis. Curcumin is a nontoxic dietary polyphenol

that is able to target a large number of inflammatory mediators. It

seems that several therapeutic effects of curcumin are mediated

through modulation of NF‐κB. Targeting of NF‐κB by curcumin

attenuates the release of inflammatory cytokines, prostaglandins,TABLE

2Su

mmaryofstudiesreportingan

ti‐in

flam

matory

effectsofcu

rcumin

against

cancercells

Tumorce

llsModality

Majorfindings

Mec

han

isms

Referen

ce

Human

Burkitt’slymphomacells

Rad

iation

Apoptosisinduction

NF‐κBan

dPI3K/A

ktpathway

inhibition

(Qiaoet

al.,2013)

Human

Burkitt’slymphomacells

Rad

iation

Apoptosisinduction

Increa

sedG2/M

phasearrest

andNF‐κBinhibition

(Qiaoet

al.,2012)

MCF‐10F

Rad

iation

Proliferationsuppression,g

rowth

inhibition

Increa

sedG2/M

phasearrest,red

uctionofRasGRF1

expression,a

ndincrea

singDNA

dam

age

Calaf

etal.(2012)

PC3cells

Rad

iation

Apoptosisinduction

InhibitionofPI3K

andactiva

tionofp53

(Liet

al.,2007)

Sarcomacells

Rad

iation

Apoptosisinduction

Stim

ulationofp53,p

21,a

ndBax

(Vee

raragh

avan

etal.,2010)

Nasopharyn

geal

carcinoma

Rad

iation

Inhibitionoftumorgrowth,a

poptosisinduction

Inactiva

tionofJab1,G2/M

arrest

(Pan

etal.,2013)

Ehrlichascitescarcinomacells

Doxo

rubicin

Red

uctionoftumorcells

viab

ility

SuppressionofNF‐κBan

dBcl‐2,activa

tionofp53,

Bax

,PUMA,a

ndNOXA

(Sen

etal.,2011)

MKN45an

dAGScells

5‐FU

Inhibitionoftumorgrowth

InhibitionofNF‐κBan

dCOX‐2

(Yan

get

al.,2017)

HCT116an

dch

3cells

5‐FU

Apoptosisinduction

Mitoch

ondrial

deg

enerationan

dupregu

lationof

proap

optoticproteins

(Shak

ibae

i

etal.2

013)

HNSC

C5‐FU

andDOX

Apoptosisinduction,g

rowth

inhibition

Cellcyclegrowth

arrest

attheG1/S

phase,

increa

sed

p21,p

53,a

ndBax

(Sivan

antham

et

al.,2016)

Note.

Akt:protein

kinaseB;EAC:Ehrlichascitescarcinoma;

5‐FU:5‐fluorouracil;HNSC

C:hea

dan

dnecksquam

ouscellcarcinoma;

PI3K:phosphoinositide3‐kinase;

NF‐κB:nuclea

rfactorκB

.

8 | FARHOOD ET AL.

Page 9: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

pro‐oxidant enzymes and free radicals. These effects reduce damage

to DNA and cell death, leading to the attenuation of redox

interactions and chronic oxidative stress. As NF‐κB has a potent

antiapoptotic activity, targeting of NF‐κB by curcumin in tumor cells

sensitizes the cells to apoptosis, thereby reducing cell survival and

tumor growth. Upregulation of proapoptotic factors such as p53 and

Bax, and arrest of cell cycle in G1 or G2 phases are other antitumor

activities of curcumin that enhance the toxic effects of radiation and

chemotherapy agents on tumor cells. Although clinical studies of

curcumin administration in patients with cancer are limited, some

studies have proposed beneficial effects of this phytochemical in

reducing dermatitis and mucositis. However, additional robust

evidence is still required from proof‐of‐concept trials in patients

with cancer to determine the role of curcumin in preventing

chemotherapy‐ and radiotherapy‐induced inflammatory complica-

tions as well as the optimal dosing and formulation of curcumin to

elicit pharmacological effects (Table 1,2).

CONFLICTS OF INTEREST

The authors declare that there are no conflicts of interest.

ORCID

Amirhossein Sahebkar http://orcid.org/0000-0002-8656-1444

REFERENCES

Abderrazak, A., Syrovets, T., Couchie, D., El Hadri, K., Friguet, B., Simmet,

T., & Rouis, M. (2015). NLRP3 inflammasome: From a danger signal

sensor to a regulatory node of oxidative stress and inflammatory

diseases. Redox Biology, 4, 296–307.

Abusnina, A., Keravis, T., Zhou, Q., Justiniano, H., Lobstein, A., &

Lugnier, C. (2015). Tumour growth inhibition and anti‐angiogeniceffects using curcumin correspond to combined PDE2 and PDE4

inhibition. Thrombosis and Haemostasis, 113(2), 319–328.

Adhvaryu M, V. B., & Reddy, N. (2018). Curcumin prevents mucositis and

improves patient compliance in head & neck cancer patients under-

going radio‐chemotherapy. Annals of Medicinal Chemistry and Research,

4(1), 1022.

Aggarwal, B. B., & Harikumar, K. B. (2009). Potential therapeutic effects of

curcumin, the anti‐inflammatory agent, against neurodegenerative,

cardiovascular, pulmonary, metabolic, autoimmune and neoplastic

diseases. The International Journal of Biochemistry & Cell Biology, 41(1),

40–59.

Aggarwal, B. B., Sundaram, C., Malani, N., & Ichikawa, H. (2007). Curcumin:

The Indian solid gold. Advances in Experimental Medicine and Biology,

595, 1–75.

Anitha, A., Deepa, N., Chennazhi, K. P., Lakshmanan, V. K., & Jayakumar, R.

(2014). Combinatorial anticancer effects of curcumin and 5‐fluorour-acil loaded thiolated chitosan nanoparticles towards colon cancer

treatment. Biochimica et Biophysica Acta, 1840(9), 2730–2743.

Anitha, A., Sreeranganathan, M., Chennazhi, K. P., Lakshmanan, V. K., &

Jayakumar, R. (2014). In vitro combinatorial anticancer effects of 5‐fluorouracil and curcumin loaded N,O‐carboxymethyl chitosan nano-

particles toward colon cancer and in vivo pharmacokinetic studies.

European Journal of Pharmaceutics and Biopharmaceutics, 88(1),

238–251.

Annunziata, C. M., Stavnes, H. T., Kleinberg, L., Berner, A., Hernandez, L. F.,

Birrer, M. J., … Kohn, E. C. (2010). Nuclear factor κB transcription

factors are coexpressed and convey a poor outcome in ovarian cancer.

Cancer, 116(13), 3276–3284.

Anscher, M. S. (2010). Targeting the TGF‐β1 pathway to prevent normal

tissue injury after cancer therapy. The Oncologist, 15(4), 350–359.

Antunes, L. M. G., Araújo, M. C. P., Darin, J. D. C., & Bianchi, M. L. P.

(2000). Effects of the antioxidants curcumin and vitamin C on

cisplatin‐induced clastogenesis in Wistar rat bone marrow cells.

Mutation Research, 465(1), 131–137.

Aravindan, N., Veeraraghavan, J., Madhusoodhanan, R., Herman, T. S., &

Natarajan, M. (2011). Curcumin regulates low‐LET γ‐radiation induced

NFκB dependent telomerase activity in human neuroblastoma cells.

International Journal of Radiation Oncology, Biology, Physics, 79(4),

1206–1215.

Arjumand, W., Seth, A., & Sultana, S. (2011). Rutin attenuates cisplatin

induced renal inflammation and apoptosis by reducing NF‐κB, TNF‐αand caspase‐3 expression in wistar rats. Food and Chemical Toxicology,

49(9), 2013–2021.

Barker, H. E., Paget, J. T. E., Khan, A. A., & Harrington, K. J. (2015). The

tumour microenvironment after radiotherapy: Mechanisms of resis-

tance and recurrence. Nature Reviews Cancer, 15(7), 409–425.

Bar‐Sela, G., Epelbaum, R., & Schaffer, M. (2010). Curcumin as an anti‐cancer agent: Review of the gap between basic and clinical

applications. Current Medicinal Chemistry, 17(3), 190–197.

Basnet, P., & Skalko‐Basnet, N. (2011). Curcumin: An anti‐inflammatory

molecule from a curry spice on the path to cancer treatment.

Molecules, 16(6), 4567–4598.

Bava, S. V., Sreekanth, C. N., Thulasidasan, A. K., Anto, N. P., Cheriyan,

V. T., Puliyappadamba, V. T., … Anto, R. J. (2011). Akt is upstream and

MAPKs are downstream of NF‐κB in paclitaxel‐induced survival

signaling events, which are down‐regulated by curcumin contributing

to their synergism. The International Journal of Biochemistry & Cell

Biology, 43(3), 331–341.

Bazargani, Y. T., de Boer, A., Schellens, J. H. M., Leufkens, H. G. M., &

Mantel‐Teeuwisse, A. K. (2014). Selection of oncology medicines in

low‐ and middle‐income countries. Annals of Oncology, 25(1), 270–276.

Boerma, M., Sridharan, V., Mao, X. W., Nelson, G. A., Cheema, A. K.,

Koturbash, I., … Hauer‐Jensen, M. (2016). Effects of ionizing radiation

on the heart. Mutation Research, 770(Pt B), 319–327.

Boonrao, M., Yodkeeree, S., Ampasavate, C., Anuchapreeda, S., &

Limtrakul, P. (2010). The inhibitory effect of turmeric curcuminoids

on matrix metalloproteinase‐3 secretion in human invasive breast

carcinoma cells. Archives of Pharmacal Research, 33(7), 989–998.

Calaf, G. M., Echiburú‐Chau, C., Wen, G., Balajee, A. S., & Roy, D.

(2012). Effect of curcumin on irradiated and estrogen‐trans-formed human breast cell lines. International Journal of Oncology,

40(2), 436–442.

Cao, J., Liu, Y., Jia, L., Zhou, H. M., Kong, Y., Yang, G., … Zhong, L. F. (2007).

Curcumin induces apoptosis through mitochondrial hyperpolarization

and mtDNA damage in human hepatoma G2 cells. Free Radical

Biology & Medicine, 43(6), 968–975.

Cheki, M., Yahyapour, R., Farhood, B., Rezaeyan, A., Shabeeb, D., Amini, P.,

… Najafi, M. (2018). COX‐2 in radiotherapy: A potential target

for radioprotection and radiosensitization. Current Molecular

Pharmacology, 11(3), 173–183.

Chen , C., Liu, Y., Chen, Y., & Xu, J. (2011). C086, a novel analog of

curcumin, induces growth inhibition and down‐regulation of NF‐κBin colon cancer cells and xenograft tumors. Cancer Biology & Therapy,

12(9), 797–807.

Chen, X., Wang, J., Fu, Z., Zhu, B., Wang, J., Guan, S., & Hua, Z. (2017).

Curcumin activates DNA repair pathway in bone marrow to

improve carboplatin‐induced myelosuppression. Scientific Reports,

7(1), 17724.

FARHOOD ET AL. | 9

Page 10: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

Chen, Y., Jungsuwadee, P., Vore, M., Butterfield, D. A., St, & St. clair, D. K.

(2007). Collateral damage in cancer chemotherapy: Oxidative stress

in nontargeted tissues. Molecular Interventions, 7(3), 147–156.

Chendil, D., Ranga, R. S., Meigooni, D., Sathishkumar, S., & Ahmed, M. M.

(2004). Curcumin confers radiosensitizing effect in prostate cancer

cell line PC‐3. Oncogene, 23(8), 1599–1607.Chiang, I. T., Liu, Y. C., Hsu, F. T., Chien, Y. C., Kao, C. H. K., Lin, W. J., …

Hwang, J. J. (2014). Curcumin synergistically enhances the radio-

sensitivity of human oral squamous cell carcinoma via suppression of

radiation‐induced NF‐κB activity. Oncology Reports, 31(4), 1729–1737.

Cho, Y. J., Yi, C. O., Jeon, B. T., Jeong, Y. Y., Kang, G. M., Lee, J. E., … Lee, J.

D. (2013). Curcumin attenuates radiation‐induced inflammation and

fibrosis in Rat lungs. The Korean Journal of Physiology & Pharmacology,

17(4), 267–274.

Chtourou, Y., Aouey, B., Aroui, S., Kebieche, M., & Fetoui, H. (2016). Anti‐apoptotic and anti‐inflammatory effects of naringin on cisplatin‐induced renal injury in the rat. Chemico‐Biological Interactions, 243,1–9.

Cicero, A. F. G., Colletti, A., Bajraktari, G., Descamps, O., Djuric, D. M.,

Ezhov, M., … Banach, M. (2017). Lipid lowering nutraceuticals in

clinical practice: Position paper from an International Lipid Expert

Panel. Archives of Medical Science, 13(5), 965–1005.

Conklin, K. A. (2004). Chemotherapy‐associated oxidative stress: Impact

on chemotherapeutic effectiveness. Integrative Cancer Therapies, 3(4),

294–300.

Deeb, D., Xu, Y. X., Jiang, H., Gao, X., Janakiraman, N., Chapman, R. A., &

Gautam, S. C. (2003). Curcumin (diferuloyl‐methane) enhances tumor

necrosis factor‐related apoptosis‐inducing ligand‐induced apoptosis in

LNCaP prostate cancer cells. Molecular Cancer Therapeutics, 2(1),

95–103.

Deguchi, A. (2015). Curcumin targets in inflammation and cancer.

Endocrine, Metabolic & Immune Disorders Drug Targets, 15(2), 88–96.

Demaria, M., O’Leary, M. N., Chang, J., Shao, L., Liu, S., Alimirah, F., …

Campisi, J. (2017). Cellular senescence promotes adverse effects

of chemotherapy and cancer relapse. Cancer Discovery, 7(2),

165–176.

Domitrović, R., Cvijanović, O., Pugel, E. P., Zagorac, G. B., Mahmutefendić,

H., & Škoda, M. (2013). Luteolin ameliorates cisplatin‐inducednephrotoxicity in mice through inhibition of platinum accumulation,

inflammation and apoptosis in the kidney. Toxicology, 310, 115–123.

Elad, S., Meidan, I., Sellam, G., Simaan, S., Zeevi, I., Waldman, E., … Revel‐Vilk, S. (2013). Topical curcumin for the prevention of oral mucositis

in pediatric patients: Case series. Alternative Therapies in Health and

Medicine, 19(3), 21–24.

Eldabaje, R., Le, D. L., Huang, W., & Yang, L. X. (2015). Radiation‐associated cardiac injury. Anticancer Research, 35(5), 2487–2492.

El‐Naga, R. N. (2014). Pre‐treatment with cardamonin protects against

cisplatin‐induced nephrotoxicity in rats: Impact on NOX‐1, inflamma-

tion and apoptosis. Toxicology and Applied Pharmacology, 274(1),

87–95.

El‐Tahawy, N. (2009). Curcumin attenuates gamma radiation induced

intestinal damage in rats. Egyptian Journal of Basic and Applied Sciences,

22(1), 7–34.

Farhood, B., Goradel, N. H., Mortezaee, K., Khanlarkhani, N., Salehi, E.,

Nashtaei, M. S., … Najafi, M. (2018). Intercellular communications‐redox interactions in radiation toxicity; potential targets for radiation

mitigation. Journal of Cell Communication and Signaling

Favero, G., Franceschetti, L., Bonomini, F., Rodella, L. F., & Rezzani, R.

(2017). Melatonin as an anti‐inflammatory agent modulating inflam-

masome activation. International Journal of Endocrinology, 2017,

https://doi.org/10.1155/2017/1835195.

Fernández‐Gil, B., Moneim, A. E. A., Ortiz, F., Shen, Y. Q., Soto‐Mercado, V.,

Mendivil‐Perez, M., … Escames, G. (2017). Melatonin protects rats

from radiotherapy‐induced small intestine toxicity. PLOS One, 12(4),

e0174474.

Fukuda, K., Uehara, Y., Nakata, E., Inoue, M., Shimazu, K., Yoshida, T., …

Shibata, H. (2016). A diarylpentanoid curcumin analog exhibits

improved radioprotective potential in the intestinal mucosa. Interna-

tional Journal of Radiation Biology, 92(7), 388–394.

Gallardo, M., & Calaf, G. M. (2016). Curcumin and epithelial‐mesenchymal

transition in breast cancer cells transformed by low doses of radiation

and estrogen. International Journal of Oncology, 48(6), 2534–2542.

Ganjali, S., Blesso, C. N., Banach, M., Pirro, M., Majeed, M., & Sahebkar, A.

(2017). Effects of curcumin on HDL functionality. Pharmacological

Research, 119, 208–218.

Hagar, H., Medany, A. E., Salam, R., Medany, G. E., & Nayal, O. A. (2015).

Betaine supplementation mitigates cisplatin‐induced nephrotoxicity

by abrogation of oxidative/nitrosative stress and suppression of

inflammation and apoptosis in rats. Experimental and Toxicologic

Pathology, 67(2), 133–141.

Hassanshahi, M., Hassanshahi, A., Khabbazi, S., Su, Y.‐W., & Xian, C. J.

(2017). Bone marrow sinusoidal endothelium: Damage and potential

regeneration following cancer radiotherapy or chemotherapy. Angio-

genesis, 20(4), 427–442.

Hatcher, H., Planalp, R., Cho, J., Torti, F. M., & Torti, S. V. (2008).

Curcumin: From ancient medicine to current clinical trials. Cellular and

Molecular Life Sciences, 65(11), 1631–1652.

Hsu, F.‐T., Liu, Y.‐C., Liu, T.‐T., & Hwang, J.‐J. (2015). Curcumin sensitizes

hepatocellular carcinoma cells to radiation via suppression of

radiation‐induced NF‐κB activity. BioMed Research International,

2015, https://doi.org/10.1155/2015/363671.

Hymes, S. R., Strom, E. A., & Fife, C. (2006). Radiation dermatitis: Clinical

presentation, pathophysiology, and treatment 2006. Journal of the

American Academy of Dermatology, 54(1), 28–46.

Ibrahim, M. A., El‐Sheikh, A. A. K., Khalaf, H. M., & Abdelrahman, A. M.

(2014). Protective effect of peroxisome proliferator activator recep-

tor (PPAR)‐α and ‐γ ligands against methotrexate‐induced nephro-

toxicity. Immunopharmacology and Immunotoxicology, 36(2), 130–137.

Iranshahi, M., Sahebkar, A., Hosseini, S. T., Takasaki, M., Konoshima, T., &

Tokuda, H. (2010). Cancer chemopreventive activity of diversin from

Ferula diversivittata in vitro and in vivo. Phytomedicine, 17(3‐4),269–273.

Irving, G. R., Iwuji, C. O., Morgan, B., Berry, D. P., Steward, W. P., Thomas, A., …

Howells, L. M. (2015). Combining curcumin (C3‐complex, Sabinsa) with

standard care FOLFOX chemotherapy in patients with inoperable

colorectal cancer (CUFOX): Study protocol for a randomised control trial.

Trials, 16, 110.

Jagetia, G. C., & Rajanikant, G. K. (2005). Curcumin treatment enhances

the repair and regeneration of wounds in mice exposed to hemibody

gamma‐irradiation. Plastic and Reconstructive Surgery, 115(2),

515–528.

Jain, R. K. (2013). Normalizing tumor microenvironment to treat cancer:

Bench to bedside to biomarkers. Journal of Clinical Oncology, 31(17),

2205–2218.

James, M. I., Iwuji, C., Irving, G., Karmokar, A., Higgins, J. A., Griffin‐Teal, N., …Brown, K. (2015). Curcumin inhibits cancer stem cell phenotypes in ex

vivo models of colorectal liver metastases, and is clinically safe and

tolerable in combination with FOLFOX chemotherapy. Cancer Letters,

364(2), 135–141.

Javvadi, P., Segan, A. T., Tuttle, S. W., & Koumenis, C. (2008). The

chemopreventive agent curcumin is a potent radiosensitizer of human

cervical tumor cells via increased reactive oxygen species production

and overactivation of the mitogen‐activated protein kinase pathway.

Molecular Pharmacology, 73(5), 1491–1501.

Junttila, M. R., & de Sauvage, F. J. (2013). Influence of tumour micro‐environment heterogeneity on therapeutic response. Nature,

501(7467), 346–354.

Kang, J., Demaria, S., & Formenti, S. (2016). Current clinical trials testing

the combination of immunotherapy with radiotherapy. Journal for

Immunotherapy of Cancer, 4(1), 51.

10 | FARHOOD ET AL.

Page 11: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

Kershaw, M. H., Devaud, C., John, L. B., Westwood, J. A., & Darcy, P. K.

(2013). Enhancing immunotherapy using chemotherapy and radiation

to modify the tumor microenvironment. Oncoimmunology, 2(9),

e25962.

Khafif, A., Lev‐Ari, S., Vexler, A., Barnea, I., Starr, A., Karaush, V., … Ben‐Yosef, R. (2009). Curcumin: A potential radio‐enhancer in head and

neck cancer. The Laryngoscope, 119(10), 2019–2026.

Kim, J., Park, S., Jeon, B.‐S., Jang, W.‐S., Lee, S.‐J., Son, Y., … Lee, S.‐S.(2016). Therapeutic effect of topical application of curcumin during

treatment of radiation burns in a mini‐pig model. Journal of Veterinary

Science, 17(4), 435–444.

Kim, J.‐S., Rhim, K.‐J., Jang, W.‐S., Lee, S.‐J., Son, Y., Lee, S.‐S., … Lim, S. M.

(2015). β‐Irradiation ((166)Ho patch)‐induced skin injury in mini‐pigs:Effects on NF‐κB and COX‐2 expression in the skin. Journal of

Veterinary Science, 16(1), 1–9.

Kunnumakkara, A. B., Diagaradjane, P., Anand, P., Kuzhuvelil, H. B.,

Deorukhkar, A., Gelovani, J., … Aggarwal, B. B. (2009). Curcumin

sensitizes human colorectal cancer to capecitabine by modulation of

cyclin D1, COX‐2, MMP‐9, VEGF and CXCR4 expression in an

orthotopic mouse model. International Journal of Cancer, 125(9),

2187–2197.

Kunnumakkara, A. B., Diagaradjane, P., Guha, S., Deorukhkar, A., Shentu,

S., Aggarwal, B. B., & Krishnan, S. (2008). Curcumin sensitizes human

colorectal cancer xenografts in nude mice to gamma‐radiation by

targeting nuclear factor‐κB‐regulated gene products. Clinical Cancer

Research, 14(7), 2128–2136.

Lee, J. C., Kinniry, P. A., Arguiri, E., Serota, M., Kanterakis, S., Chatterjee, S., …

Christofidou‐Solomidou, M. (2010). Dietary curcumin increases antiox-

idant defenses in lung, ameliorates radiation‐induced pulmonary fibrosis,

and improves survival in mice. Radiation Research, 173(5), 590–601.

Lee, J. H., Kay, C. S., Maeng, L. S., Oh, S. J., Lee, A. H., Lee, J. D., … Cho, S. H.

(2009). The clinical features and pathophysiology of acute radiation

dermatitis in patients receiving tomotherapy. Annals of Dermatology,

21(4), 358–363.

Lelli, D., Sahebkar, A., Johnston, T. P., & Pedone, C. (2017). Curcumin use

in pulmonary diseases: State of the art and future perspectives.

Pharmacological Research, 115, 133–148.

Li, M., Zhang, Z., Hill, D. L., Wang, H., & Zhang, R. (2007). Curcumin, a

dietary component, has anticancer, chemosensitization, and radio-

sensitization effects by down‐regulating the MDM2 oncogene

through the PI3K/mTOR/ETS2 pathway. Cancer Research, 67(5),

1988–1996.

Li, W., Jiang, Z., Xiao, X., Wang, Z., Wu, Z., Ma, Q., & Cao, L. (2018).

Curcumin inhibits superoxide dismutase‐induced epithelial‐to‐me-

senchymal transition via the PI3K/Akt/NF‐κB pathway in pancreatic

cancer cells. International Journal of Oncology

Li, Y., & Zhang, T. (2014). Targeting cancer stem cells by curcumin and

clinical applications. Cancer Letters, 346(2), 197–205.

Lian, Q., Xu, J., Yan, S., Huang, M., Ding, H., Sun, X., … Geng, M. (2017).

Chemotherapy‐induced intestinal inflammatory responses are

mediated by exosome secretion of double‐strand DNA via AIM2

inflammasome activation. Cell Research, 27(6), 784–800.

Liyanage, U. K., Moore, T. T., Joo, H.‐G., Tanaka, Y., Herrmann, V.,

Doherty, G., … Linehan, D. C. (2002). Prevalence of regulatory T

cells is increased in peripheral blood and tumor microenvironment

of patients with pancreas or breast adenocarcinoma. The Journal of

Immunology, 169(5), 2756–2761.

Luo, J., Tsuji, T., Yasuda, H., Sun, Y., Fujigaki, Y., & Hishida, A. (2008). The

molecular mechanisms of the attenuation of cisplatin‐induced acute

renal failure by N‐acetylcysteine in rats. Nephrology, Dialysis,

Transplantation, 23(7), 2198–2205.

López‐Lázaro, M. (2008). Anticancer and carcinogenic properties of

curcumin: Considerations for its clinical development as a cancer

chemopreventive and chemotherapeutic agent. Molecular Nutrition &

Food Research, 52(Suppl 1), S103–S127.

Mabuchi, S., Ohmichi, M., Nishio, Y., Hayasaka, T., Kimura, A., Ohta, T., …

Murata, Y. (2004). Inhibition of inhibitor of nuclear factor‐κBphosphorylation increases the efficacy of paclitaxel in in vitro and in

vivo ovarian cancer models. Clinical Cancer Research, 10(22),

7645–7654.

Marjaneh, R. M., Rahmani, F., Hassanian, S. M., Rezaei, N., Hashemzehi, M.,

Bahrami, A., … Khazaei, M. (2018). Phytosomal curcumin inhibits

tumor growth in colitis‐associated colorectal cancer. Journal of Cellular

Physiology, 233, 6785–6798.

Meng, A., Wang, Y., Brown, S. A., Van Zant, G., & Zhou, D. (2003). Ionizing

radiation and busulfan inhibit murine bone marrow cell hematopoietic

function via apoptosis‐dependent and‐independent mechanisms.

Experimental Hematology, 31(12), 1348–1356.

Miller, K. D., Siegel, R. L., Lin, C. C., Mariotto, A. B., Kramer, J. L., Rowland,

J. H., … Jemal, A. (2016). Cancer treatment and survivorship statistics,

2016. CA: A Cancer Journal for Clinicians, 66(4), 271–289.

Mirzaei, H., Naseri, G., Rezaee, R., Mohammadi, M., Banikazemi, Z.,

Mirzaei, H. R., … Sahebkar, A. (2016). Curcumin: A new candidate for

melanoma therapy. International Journal of Cancer, 139(8), 1683–1695.

Momtazi, A. A., Shahabipour, F., Khatibi, S., Johnston, T. P., Pirro, M., &

Sahebkar, A. (2016). Curcumin as a microRNA regulator in cancer: A

review. Reviews of Physiology, Biochemistry and Pharmacology,

171, 1–38.

Müller, K., & Meineke, V. (2007). Radiation‐induced alterations in cytokine

production by skin cells. Experimental Hematology, 35(Suppl 1),

96–104.

Nafees, S., Rashid, S., Ali, N., Hasan, S. K., & Sultana, S. (2015). Rutin

ameliorates cyclophosphamide induced oxidative stress and inflam-

mation in Wistar rats: Role of NF‐κB/MAPK pathway. Chemico‐Biological Interactions, 231, 98–107.

Najafi, M., Cheki, M., Rezapoor, S., Geraily, G., Motevaseli, E., Carnovale, C.,

Clementi, E., & Shirazi, A. (2018). Metformin: Prevention of genomic

instability and cancer: A review. Mutation Research, https://doi.org/10.

1016/j.mrgentox.2018.01.007.

Najafi, M., Motevaseli, E., Shirazi, A., Geraily, G., Rezaeyan, A., Norouzi, F., …

Abdollahi, H. (2018). Mechanisms of inflammatory responses to radiation

and normal tissues toxicity: Clinical implications. International Journal of

Radiation Biology, 94(4), 335–356.

Newman, N. B., Sidhu, M. K., Baby, R., Moss, R. A., Nissenblatt, M. J., Chen, T.,…

Jabbour, S. K. (2016). Long‐term bone marrow suppression during

postoperative chemotherapy in rectal cancer patients after preoperative

chemoradiation therapy. International Journal of Radiation Oncology Biology

Physics, 94(5), 1052–1060.

Noorafshan, A., & Ashkani‐Esfahani, S. (2013). A review of therapeutic

effects of curcumin. Current Pharmaceutical Design, 19(11),

2032–2046.

Ohta, T., Ohmichi, M., Hayasaka, T., Mabuchi, S., Saitoh, M., Kawagoe, J., …

Kurachi, H. (2006). Inhibition of phosphatidylinositol 3‐kinaseincreases efficacy of cisplatin in in vivo ovarian cancer models.

Endocrinology, 147(4), 1761–1769.

Okunieff, P., Xu, J., Hu, D., Liu, W., Zhang, L., Morrow, G., … Ding, I. (2006).

Curcumin protects against radiation‐induced acute and chronic

cutaneous toxicity in mice and decreases mRNA expression of

inflammatory and fibrogenic cytokines. International Journal of Radia-

tion Oncology, Biology, Physics, 65(3), 890–898.

Onoda, M., & Inano, H. (2000). Effect of curcumin on the production of

nitric oxide by cultured rat mammary gland. Nitric Oxide: Biology and

Chemistry, 4(5), 505–515.

Orr, W. S., Denbo, J. W., Saab, K. R., Ng, C. Y., Wu, J., Li, K., … Davidoff, A.

M. (2013). Curcumin potentiates rhabdomyosarcoma radiosensitivity

by suppressing NF‐κB activity. PLOS One, 8(2), e51309.

Ortiz, F., Acuña‐Castroviejo, D., Doerrier, C., Dayoub, J. C., López, L. C.,

Venegas, C., … Escames, G. (2015). Melatonin blunts the mitochon-

drial/NLRP3 connection and protects against radiation‐induced oral

mucositis. Journal of Pineal Research, 58(1), 34–49.

FARHOOD ET AL. | 11

Page 12: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

Pal, S., Bhattacharyya, S., Choudhuri, T., Datta, G. K., Das, T., & Sa, G. (2005).

Amelioration of immune cell number depletion and potentiation of

depressed detoxification system of tumor‐bearing mice by curcumin.

Cancer Detection and Prevention, 29(5), 470–478.

Palatty, P. L., Azmidah, A., Rao, S., Jayachander, D., Thilakchand, K. R.,

Rai, M. P., … Baliga, M. S. (2014). Topical application of a sandal wood

oil and turmeric based cream prevents radiodermatitis in head and

neck cancer patients undergoing external beam radiotherapy: A pilot

study. The British Journal of Radiology, 87(1038), 20130490.

Pan, Y., Wang, M., Bu, X., Zuo, Y., Wang, S., Wang, D., … Yang, H. (2013).

Curcumin analogue T83 exhibits potent antitumor activity and

induces radiosensitivity through inactivation of Jab1 in nasophar-

yngeal carcinoma. BMC Cancer, 13, 323.

Panahi, Y., Alishiri, G. H., Parvin, S., & Sahebkar, A. (2016). Mitigation of

systemic oxidative stress by curcuminoids in osteoarthritis: Results of

a randomized controlled trial. Journal of Dietary Supplements, 13(2),

209–220.

Panahi, Y., Ghanei, M., Bashiri, S., Hajihashemi, A., & Sahebkar, A. (2014).

Short‐term curcuminoid supplementation for chronic pulmonary

complications due to sulfur mustard intoxication: Positive results of

a randomized double‐blind placebo‐controlled trial. Drug Research,

65(11), 567–573.

Panahi, Y., Ghanei, M., Hajhashemi, A., & Sahebkar, A. (2016). Effects of

curcuminoids‐piperine combination on systemic oxidative stress,

clinical symptoms and quality of life in subjects with chronic

pulmonary complications due to sulfur mustard: A randomized

controlled trial. Journal of Dietary Supplements, 13(1), 93–105.

Panahi, Y., Kianpour, P., Mohtashami, R., Jafari, R., Simental‐Mendía, L. E.,

& Sahebkar, A. (2016). Curcumin lowers serum lipids and uric acid in

subjects with nonalcoholic fatty liver disease: A randomized con-

trolled trial. Journal of Cardiovascular Pharmacology, 68(3), 223–229.

Panahi, Y., Rahimnia, A. R., Sharafi, M., Alishiri, G., Saburi, A., & Sahebkar,

A. (2014). Curcuminoid treatment for knee osteoarthritis: A rando-

mized double‐blind placebo‐controlled trial. Phytotherapy Research,

28(11), 1625–1631.

Paraswani, N., Ghosh, A., & Thoh, M. (2017). Cellular redox status

mediates adaptive response to ionizing radiation in human peripheral

blood mononuclear cells. Free Radical Biology and Medicine, 108, S22.

Patil, K., Guledgud, M. V., Kulkarni, P. K., Keshari, D., & Tayal, S. (2015).

Use of curcumin mouthrinse in radio‐chemotherapy induced oral

mucositis patients: A pilot study. Journal of Clinical and Diagnostic

Research, 9(8), ZC59–ZC62.

Peterson, D. E., Bensadoun, R. J., & Roila, F., ESMO Guidelines Working

Group (2011). Management of oral and gastrointestinal mucositis:

ESMO Clinical Practice Guidelines. Annals of Oncology, 22(Suppl 6),

vi78–vi84.

Proklou, A., Diamantaki, E., Pediaditis, E., & Kondili, E. (2018). Radiation

therapy: Impact on lung function and acute respiratory failure.

Mechanical Ventilation in Critically Ill Cancer Patients, 33–39.

Ps, S. K., Balan, A., Sankar, A., & Bose, T. (2009). Radiation induced oral

mucositis. Indian Journal of Palliative Care, 15(2), 95–102.

Pusztai, L., Mendoza, T. R., Reuben, J. M., Martinez, M. M., Willey, J. S.,

Lara, J., … Hortobagyi, G. N. (2004). Changes in plasma levels of

inflammatory cytokines in response to paclitaxel chemotherapy.

Cytokine, 25(3), 94–102.

Qadir, M. I., Naqvi, S. T., & Muhammad, S. A. (2016). Curcumin: A

polyphenol with molecular targets for cancer control. Asian Pacific

Journal of Cancer Prevention, 17(6), 2735–2739.

Qiao, Q., Jiang, Y., & Li, G. (2012). Curcumin improves the antitumor effect

of X‐ray irradiation by blocking the NF‐κB pathway: An in‐vitro study

of lymphoma. Anti‐Cancer Drugs, 23(6), 597–605.Qiao, Q., Jiang, Y., & Li, G. (2013). Inhibition of the PI3K/AKT‐NF‐κB

pathway with curcumin enhanced radiation‐induced apoptosis in

human Burkitt’s lymphoma. Journal of Pharmacological Sciences, 121(4),

247–256.

Rafiee, P., Binion, D. G., Wellner, M., Behmaram, B., Floer, M., Mitton, E., …

Otterson, M. F. (2010). Modulatory effect of curcumin on survival of

irradiated human intestinal microvascular endothelial cells: Role of

Akt/mTOR and NF‐κB. American Journal of Physiology Gastrointestinal

and Liver Physiology, 298(6), G865–G877.

Rahmani, S., Asgary, S., Askari, G., Keshvari, M., Hatamipour, M., Feizi, A.,

& Sahebkar, A. (2016). Treatment of Non‐alcoholic fatty liver disease

with curcumin: A randomized placebo‐controlled Trial. Phytotherapy

Research, 30, 1540–1548.

Rajput, S., Volk‐Draper, L. D., & Ran, S. (2013). TLR4 is a novel

determinant of the response to paclitaxel in breast cancer. Molecular

Cancer Therapeutics, 12(8), 1676–1687.

Ramesh, G., & Reeves, W. B. (2003). TNFR2‐mediated apoptosis and

necrosis in cisplatin‐induced acute renal failure. American Journal of

Physiology‐Renal Physiology, 285(4), F610–F618.Ramesh, G., & Reeves, W. B. (2004). Salicylate reduces cisplatin

nephrotoxicity by inhibition of tumor necrosis factor‐α. Kidney

International, 65(2), 490–499.

Ramezani, M., Hatamipour, M., & Sahebkar, A. (2018). Promising

anti‐tumor properties of bisdemethoxycurcumin: A naturally

occurring curcumin analogue. Journal of Cellular Physiology,

233(2), 880–887.

Rao, S., Dinkar, C., Vaishnav, L. K., Rao, P., Rai, M. P., Fayad, R., & Baliga, M.

S. (2014). The Indian spice turmeric delays and mitigates radiation‐induced oral mucositis in patients undergoing treatment for head and

neck cancer: An investigational study. Integrative Cancer Therapies,

13(3), 201–210.

Rao, S., Hegde, S., Baliga‐Rao, M., Lobo, J., Palatty, P., George, T., & Baliga,

M. (2017). Sandalwood oil and turmeric‐based cream prevents

ionizing radiation‐induced dermatitis in breast cancer patients:

Clinical study. Medicines, 4(3), 43.

Rezaee, R., Momtazi, A. A., Monemi, A., & Sahebkar, A. (2017). Curcumin:

A potentially powerful tool to reverse cisplatin‐induced toxicity.

Pharmacological Research, 117, 218–227.

Rezvani, M., & Ross, G. A. (2004). Modification of radiation‐induced acute

oral mucositis in the rat. International Journal of Radiation Biology, 80

(2), 177–182.

Robbins, M. E. C., & Zhao, W. (2004). Chronic oxidative stress and

radiation‐induced late normal tissue injury: A review. International

Journal of Radiation Biology, 80(4), 251–259.

Ryan, J. L., Heckler, C. E., Ling, M., Katz, A., Williams, J. P., Pentland, A. P.,

& Morrow, G. R. (2013). Curcumin for radiation dermatitis: a

randomized, double‐blind, placebo‐controlled clinical trial of thirty

breast cancer patients. Radiation Research, 180(1), 34–43.

Ryan Wolf, J., Heckler, C. E., Guido, J. J., Peoples, A. R., Gewandter, J. S.,

Ling, M., … Morrow, G. R. (2018). Oral curcumin for radiation

dermatitis: A URCC NCORP study of 686 breast cancer patients.

Supportive Care in Cancer, 26(5), 1543–1552.

Saberi‐Karimian, M., Katsiki, N., Caraglia, M., Boccellino, M., Majeed, M., &

Sahebkar, A. (2017). Vascular endothelial growth factor: An important

molecular target of curcumin. Critical Reviews in Food Science and

Nutrition, 30, 1–14.

Sahebkar, A., Cicero, A. F. G., Simental‐Mendía, L. E., Aggarwal, B. B., &

Gupta, S. C. (2016). Curcumin downregulates human tumor necrosis

factor‐alpha levels: A systematic review and meta‐analysis ofrando-

mized controlled trials. Pharmacological Research, 107, 234–242.

Sahebkar, A., & Henrotin, Y. (2016). Analgesic efficacy and safety of

curcuminoids in clinical practice: A systematic review and meta‐analysis of randomized controlled trials. Pain Medicine, 17(6),

1192–1202.

Sandur, S. K., Deorukhkar, A., Pandey, M. K., Pabón, A. M., Shentu, S.,

Guha, S., … Krishnan, S. (2009). Curcumin modulates the radio-

sensitivity of colorectal cancer cells by suppressing constitutive and

inducible NF‐κB activity. International Journal of Radiation Oncology,

Biology, Physics, 75(2), 534–542.

12 | FARHOOD ET AL.

Page 13: Curcuminasananti inflammatoryagent:Implications …eprints.kaums.ac.ir/3671/1/curcuminasanti-inflammation.pdf · 2019. 5. 29. · It seems that upregulation of TLR‐4 is involved

Santos, N. A. G., Catão, C. S., Martins, N. M., Curti, C., Bianchi, M. L. P., &

Santos, A. C. (2007). Cisplatin‐induced nephrotoxicity is associated

with oxidative stress, redox state unbalance, impairment of energetic

metabolism and apoptosis in rat kidney mitochondria. Archives of

Toxicology, 81(7), 495–504.

Sen, G. S., Mohanty, S., Hossain, D. M. S., Bhattacharyya, S., Banerjee, S.,

Chakraborty, J., … Sa, G. (2011). Curcumin enhances the efficacy of

chemotherapy by tailoring p65NFkappaB‐p300 cross‐talk in favor of

p53‐p300 in breast cancer. The Journal of Biological Chemistry, 286(49),

42232–42247.

Shakeri, A., Ward, N., Panahi, Y., & Sahebkar, A. (2018). Anti‐angiogenic activityof curcumin in cancer therapy: A narrative review. Current vascular

pharmacology, https://doi.org/10.2174/1570161116666180209113014.

Shakibaei, M., Mobasheri, A., Lueders, C., Busch, F., Shayan, P., & Goel, A.

(2013). Curcumin enhances the effect of chemotherapy against

colorectal cancer cells by inhibition of NF‐kappaB and Src protein

kinase signaling pathways. PLOS One, 8(2), e57218.

Shehzad, A., Rehman, G., & Lee, Y. S. (2013). Curcumin in inflammatory

diseases. BioFactors, 39(1), 69–77.

Siegel, R. L., Miller, K. D., & Jemal, A. (2015). Cancer statistics, 2015. CA: A

Cancer Journal for Clinicians, 65(1), 5–29.

Sivanantham, B., Sethuraman, S., & Krishnan, U. M. (2016). Combinator-

ial effects of curcumin with an anti‐neoplastic agent on head and

neck squamous cell carcinoma through the regulation of EGFR‐ERK1/2 and apoptotic signaling pathways. ACS Combinatorial Science,

18(1), 22–35.

Sordillo, P. P., & Helson, L. (2015). Curcumin and cancer stem cells:

Curcumin has asymmetrical effects on cancer and normal stem cells.

Anticancer Research, 35(2), 599–614.

Stock, D. C., Groome, P. A., Siemens, D. R., Rohland, S. L., & Song, Z. (2008).

Effects of non‐selective non‐steroidal anti‐inflammatory drugs on the

aggressiveness of prostate cancer. The Prostate, 68(15), 1655–1665.

Teymouri, M., Barati, N., Pirro, M., & Sahebkar, A. (2018). Biological and

pharmacological evaluation of dimethoxycurcumin: A metabolically

stable curcumin analogue with a promising therapeutic potential.

Journal of Cellular Physiology, 233(1), 124–140.

Tian, F., Fan, T., Zhang, Y., Jiang, Y., & Zhang, X. (2012). Curcumin

potentiates the antitumor effects of 5‐FU in treatment of esophageal

squamous carcinoma cells through downregulating the activation of

NF‐κB signaling pathway in vitro and in vivo. Acta Biochimica et

Biophysica Sinica, 44(10), 847–855.

Tomar, A., Vasisth, S., Khan, S. I., Malik, S., Nag, T. C., Arya, D. S., & Bhatia,

J. (2017). Galangin ameliorates cisplatin induced nephrotoxicity in

vivo by modulation of oxidative stress, apoptosis and inflammation

through interplay of MAPK signaling cascade. Phytomedicine,

34, 154–161.

Veeraraghavan, J., Natarajan, M., Herman, T. S., & Aravindan, N. (2010).

Curcumin‐altered p53‐response genes regulate radiosensitivity in

p53‐mutant Ewing’s sarcoma cells. Anticancer Research, 30(10),

4007–4015.

Verma, V. (2016). Relationship and interactions of curcumin with

radiation therapy. World Journal of Clinical Oncology, 7(3), 275–283.

Vichaya, E. G., Chiu, G. S., Krukowski, K., Lacourt, T. E., Kavelaars, A.,

Dantzer, R., … Walker, A. K. (2015). Mechanisms of chemotherapy‐induced behavioral toxicities. Frontiers in Neuroscience, 9, 131.

Vinod, B. S., Antony, J., Nair, H. H., Puliyappadamba, V. T., Saikia, M.,

Narayanan, S. S., … Anto, R. J. (2013). Mechanistic evaluation of the

signaling events regulating curcumin‐mediated chemosensitization of

breast cancer cells to 5‐fluorouracil. Cell Death & Disease, 4, e505.

Vyas, D., Laput, G., & Vyas, A. K. (2014). Chemotherapy‐enhancedinflammation may lead to the failure of therapy and metastasis.

OncoTargets and Therapy, 7, 1015–1023.

Wang, Y., Liu, L., Pazhanisamy, S. K., Li, H., Meng, A., & Zhou, D. (2010).

Total body irradiation causes residual bone marrow injury by

induction of persistent oxidative stress in murine hematopoietic stem

cells. Free Radical Biology and Medicine, 48(2), 348–356.

Yahyapour, R., Amini, P., Rezapoor, S., Rezaeyan, A., Farhood, B., Cheki, M., …

Najafi, M. (2017). Targeting of inflammation for radiation protection and

mitigation. Current Molecular Pharmacology

Yahyapour, R., Amini, P., Rezapoor, S., Rezaeyan, A., Farhood, B.,

Cheki, M., … Najafi, M. (2018). Targeting of Inflammation for

Radiation Protection and Mitigation. Current molecular pharmacol-

ogy, 11(3), 203–210.

Yahyapour, R., Amini, P., Rezapour, S., Cheki, M., Rezaeyan, A., Farhood, B., …

Najafi, M. (2018). Radiation‐induced inflammation and autoimmune

diseases. Military Medical Research, 5, 9.

Yahyapour, R., Motevaseli, E., Rezaeyan, A., Abdollahi, H., Farhood, B.,

Cheki, M., … Villa, V. (2018). Reduction–oxidation (redox) system in

radiation‐induced normal tissue injury: Molecular mechanisms and

implications in radiation therapeutics. Clinical and Translational

Oncology, 20(8), 975–988.

Yahyapour, R., Shabeeb, D., Cheki, M., Musa, A. E., Farhood, B., Rezaeyan, A., …

Najafi, M. (2018). Radiation protection and mitigation by natural

antioxidants and flavonoids; implications to radiotherapy and radiation

disasters. Current Molecular Pharmacology, 11

Yang, H., Huang, S., Wei, Y., Cao, S., Pi, C., Feng, T., … Ren, G. (2017).

Curcumin enhances the anticancer effect of 5‐fluorouracil against

gastric cancer through down‐regulation of COX‐2 and NF‐κB signaling

pathways. Journal of Cancer, 8(18), 3697–3706.

You, J., Sun, J., Ma, T., Yang, Z., Wang, X., Zhang, Z., … Shen, Z. (2017).

Curcumin induces therapeutic angiogenesis in a diabetic mouse

hindlimb ischemia model via modulating the function of endothelial

progenitor cells. Stem Cell Research & Therapy, 8, 182.

Yue, G. G. L., Kwok, H. F., Lee, J. K. M., Jiang, L., Wong, E. C. W., Gao, S., …

Lau, C. B. S. (2016). Combined therapy using bevacizumab and

turmeric ethanolic extract (with absorbable curcumin) exhibited

beneficial efficacy in colon cancer mice. Pharmacological Research,

111, 43–57.

Zabihi, N. A., Pirro, M., Johnston, T. P., & Sahebkar, A. (2017). Is there a

role for curcumin supplementation in the treatment of non‐alcoholicfatty liver disease? The data suggest yes. Current Pharmaceutical

Design, 23(7), 969–982.

Zhang, H., Wang, Y., Meng, A., Yan, H., Wang, X., Niu, J., … Wang, H.

(2013). Inhibiting TGFβ1 has a protective effect on mouse bone

marrow suppression following ionizing radiation exposure in vitro.

Journal of Radiation Research, 54(4), 630–636.

Zhou, H., S. beevers, C., & Huang, S. (2011). Targets of curcumin. Current

Drug Targets, 12(3), 332–347.

Zhou, J., Fan, Y., Tang, S., Wu, H., Zhong, J., Huang, Z., … Chen, H. (2017).

Inhibition of PTEN activity aggravates cisplatin‐induced acute kidney

injury. Oncotarget, 8(61), 103154–103166.

Zhou, Q. M., Zhang, H., Lu, Y. Y., Wang, X. F., & Su, S. B. (2009). Curcumin

reduced the side effects of mitomycin C by inhibiting GRP58‐mediated DNA cross‐linking in MCF‐7 breast cancer xenografts.

Cancer Science, 100(11), 2040–2045.

Zhu, J. Y., Yang, X., Chen, Y., Jiang, Y., Wang, S. J., Li, Y., … Han, H. Y.

(2017). Curcumin suppresses lung cancer stem cells via inhibiting

Wnt/β‐catenin and sonic hedgehog pathways. Phytotherapy Research,

31(4), 680–688.

How to cite this article: Farhood B, Mortezaee K, Goradel

NH, et al. Curcumin as an anti‐inflammatory agent:

Implications to radiotherapy and chemotherapy. J Cell Physiol.

2018;1‐13. https://doi.org/10.1002/jcp.27442

FARHOOD ET AL. | 13