Next Article in Journal
Immunity and Breast Cancer: Focus on Eosinophils
Previous Article in Journal
Oral Cavity as a Source of Mesenchymal Stem Cells Useful for Regenerative Medicine in Dentistry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Curcumin in Cancer Treatment

by
Vasiliki Zoi
1,2,
Vasiliki Galani
2,
Georgios D. Lianos
3,
Spyridon Voulgaris
1,4,
Athanasios P. Kyritsis
1 and
George A. Alexiou
1,4,*
1
Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
2
Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
3
Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece
4
Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
*
Author to whom correspondence should be addressed.
Biomedicines 2021, 9(9), 1086; https://doi.org/10.3390/biomedicines9091086
Submission received: 27 July 2021 / Revised: 19 August 2021 / Accepted: 23 August 2021 / Published: 26 August 2021
(This article belongs to the Section Drug Discovery and Development)

Abstract

:
Curcumin is a polyphenol extracted from the rhizomes of the turmeric plant, Curcuma longa which has anti-inflammatory, and anticancer properties. Chronic inflammation is associated with the development of cancer. Curcumin acts on the regulation of various immune modulators, including cytokines, cyclooxygenase-2 (COX-2), and reactive oxygen species (ROS), which partly explains its anticancer effects. It also takes part in the downregulation of growth factors, protein kinases, oncogenic molecules and various signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 3 (STAT3) signaling. Clinical trials of curcumin have been completed or are ongoing for various types of cancer. This review presents the molecular mechanisms of curcumin in different types of cancer and the evidence from the most recent clinical trials.

1. Introduction

Curcuma longa, commonly known as turmeric, is a herbaceous plant belonging to the ginger family. [1] The plant produces a variety of secondary metabolites including flavonoids, alkaloids, tannins and phenolic acids [2], among which the active hydrophobic polyphenol diferuloymethane, named curcumin, is of special notice [3]. Curcumin is used in the treatment of various health conditions, including inflammatory disorders, liver disease, metabolic syndrome, neurodegenerative diseases and, most importantly, in several types of cancer [4]. The chemical structure of curcumin, as well as the most important curcuminoids, namely demethoxycurcumin and bis-demethoxycurcumin, are presented in Figure 1.
Cancer is among the primary causes of death worldwide [5] and, despite the high level of global awareness and the development of multitargeted therapeutic options, death rates from cancer are still significantly high [6]. Cancer cells are characterized by disruption of several signaling pathways including those responsible for angiogenesis, proliferation, metabolism, migration, immune modulation and survival [7]. Curcumin has been found to affect cancer cells in a variety of ways leading to the prevention of cancer formation. Its most prominent effects on cancer are summarized in Figure 2.
Curcumin has shown promising results in the treatment of several types of cancer both alone and in combination with other antineoplastic agents. It affects several signaling pathways and can thus effectively modify both the development and the growth of various tumors. This review summarizes the immunomodulatory effects of curcumin and the most recent evidence on the effectiveness of curcumin in the treatment of different types of cancer in vitro both alone and in combination with other chemotherapeutic agents.

2. Immunomodulatory Effects of Curcumin

Chronic inflammation disorders and infectious diseases are responsible for the development of several types of cancer [8], contributing to genomic instability, which is considered a hallmark of cancer [9]. The inflammatory process results in the production of several pro-inflammatory molecules, including reactive oxygen species (ROS), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-Κb), cytokines, AKT, the transcription factor activator protein-1 (AP-1), and cyclooxygenase-2 (COX-2), which are involved in both the initiation and the growth of tumors [10]. Curcumin acts on the regulation of different immune modulators, resulting in some of its anticancer properties.
ROS are molecules derived from oxygen with the ability to act as secondary messengers in several cellular signaling pathways. They participate in inflammation and in cell survival, differentiation and proliferation, resulting in the progress of different types of cancer [11]. Curcumin binds directly to ROS scavengers and may thus suppress the growth and metastasis of some types of cancer [12].
NF-κB factor regulates and controls the expression of various proteins, including cytokines and interferons. Those proteins are closely related to both inflammation and cancer progression [13]. Curcumin has an inhibitory action on the NF-κB-dependent pathway, resulting in the suppression of tumors and induction of apoptosis [14,15]. Cytokines are important regulators of the immune system that allow cells to communicate over short distances, and they control the proliferation, survival, differentiation, and apoptosis of leucocytes. In recent studies, cytokines, such as interleukin (IL) IL-12, IL-15 and interferon-a (IFN)-a, have been investigated in murine cancer models [16,17]. The expression of pro-inflammatory cytokines has been shown to be downregulated by the action of curcumin on the binding of nuclear proteins with interleukins or interferons [18].
The transcription factor AP-1 has been related to pro- and antiapoptotic effects in different types of cancer and its expression is downregulated by curcumin in in vitro models [19]. In microglial cells, curcumin was shown to inhibit the expression of (COX-2), while in melanoma cancer cells a concentration-dependent decrease in COX-2 was observed [20,21].
Curcumin has been found to modulate the cellular response of several cell types of the immune system, including B and T lymphocytes, macrophages and natural killer cells [22]. Curcumin can also affect both the expression and action of immune cytokines. TNF-a is a pleiotropic cytokine that induces growth stimulation and plays a pivotal role as an immunostimulant. Curcumin can modulate the expression of this factor and inhibit LPS-induced expression of TNF-α [23]. Dendritic cells, known for their immunostimulatory role, are also a target of curcumin. In particular, curcumin has been found to inhibit myeloid DC maturation, mainly through the suppression of CD80 and CD86 expression [24].
In some initial studies, curcumin supplementation showed no effect on the activity of natural killer (NK) cells in rats when given at doses of up to 40 mg/kg for up to five weeks [25]. However, in another study, Yadav et al. explored the immunomodulatory effects of curcumin and showed that this compound can increase NK cell cytotoxicity in vitro and that this effect can be further enhanced when curcumin was given along with IFN-γ treatment [26]. Increasing evidence indicates that curcumin exhibits its anti-inflammatory effects through the downregulation of NLRP3 inflammasomes [27]. Inflammasomes are multiprotein complexes that are responsible for innate immunity. The NLRP3, as a component of inflammasomes, can trigger immune response after detecting the products of damaged cells [28]. Curcumin can suppress NLRP3 inflammasome activation through regulation of the NF-κB signaling, leading also to suppression of IL-1β secretion [29]. For example, in malignant mesothelioma, curcumin has been found to exhibit its anticancer properties through regulation of signaling pathways affecting inflammasomes, such as IL-1 and NF-κB [30].
Other studies have also determined the anti-inflammatory and anticancer properties of curcumin. In a clinical investigation where human chronic myelogenous leukemia cells (K562) were studied, curcumin treatment resulted in significant downregulation of IL-6, TLRs, IL-3 and STAT-1 [31]. In another randomized double-blind placebo-controlled trial, 80 patients with solid tumors were randomly assigned to 180 mg/day of curcuminoids supplementation or matched placebo for 8 weeks. All mediators of inflammation, including interleukins, TNF-α and monocyte chemotactic protein-1 were significantly lower in the curcuminoids versus placebo group [32]. When curcumin was given as part of a botanical drug containing multiple polyphenols, named APG-157 in a double-blind, randomized phase 1 clinical trial, where 12 patients with oral cancer were enrolled, reduced IL-1β, IL-6, and IL-8 concentrations were found in the saliva 24 h after treatment [33].

3. Lung Cancer

Lung cancer is among the most commonly occurring types of cancer in men and the third most common in women. The standard treatment for lung cancer includes surgery, followed by radiation therapy, chemotherapy, and immunotherapy [34].
Curcumin exhibits its therapeutic potential in lung cancer treatment by acting on the Wnt/β-catenin-dependent pathway in the human lung cancer cell line A549 [35]. Curcumin also suppresses the vascular endothelial growth factor (VEGF0 and NF-κB expression in the same cell line [36]. In addition, curcumin has been shown to inhibit the expression of enhancer of zeste homolog 2 (EZH2) in lung cancer cells, which was followed by a decrease in the expression of the Notch 1 gene [37]. Curcumin has exhibited cytotoxicity against non-small-cell lung cancer (NSCLC) through inhibition of cell proliferation, increased apoptosis, and G2/M arrest. Curcumin also induces ROS production, thus activating the DNA damage signaling pathway [38].
A recent study investigated the effects of long-term, low-dose curcumin administration in NSCLC cells. Doses of curcumin between 0.25 and 0.5 μΜ produced significant suppression of both the invasion and metastatic potential of the cancer cells [39]. Curcumin derivatives have also shown promising results against the growth and invasion of NSCLC cells with anaplastic lymphoma kinas (ALK) rearrangements even when they become resistant to ALK inhibitory drugs [40].
The extracellular signal-regulated kinase (ERK) pathway plays an important role in the sensitivity of cancer cells to chemotherapeutic agents [41]. Curcumin selectively targets the ERK 1/2 pathway, resulting in a 75% reduction in its expression [42]. The phosphoinositide 3-kinase (PI3K)/Akt-dependent pathway has been related to cell proliferation and apoptosis in several cancer cell lines, but currently the available data on the relationship between the PI3K/Akt pathway and apoptosis are conflicting. Some drugs trigger apoptosis in cancer cells via stimulation of this pathway [43], while other drugs exert an inhibitory effect [44]. A recent study showed that a new derivative of curcumin, T59, activated the PI3K/Akt pathway and induced apoptosis in lung cancer cells [45], but another study found that a combination of curcumin and paris saponin 2 (PS2) induced apoptosis in lung cancer cells through inhibition of the PI3K-Akt pathway.
Tobacco smoke plays a significant role in the development of lung cancer. It has been estimated that approximately 90% of all lung cancer cases are associated with tobacco smoke [46]. The mitogen-activated protein kinase (MAPK) pathway is involved in the epithelial–mesenchymal transition (EMT) that is induced by tobacco smoke (TS). [47] Liang et al. investigated the effects of curcumin on the tobacco smoke-induced EMT in nude mice and concluded that curcumin can attenuate EMT alterations through downregulation of the MAPK/AP-1-dependent pathway [48]. In another study, Xie et al. investigated the possible effect of curcumin on TAp63α transcription factor (tumor suppressor gene). This factor is repressed by TS-induced lung cancer EMT, and it was found that curcumin can increase TAp63α expression and thus play a positive role in smocking-induced carcinogenesis [49].

4. Breast Cancer

Breast cancer continues to be the commonest type of cancer among women worldwide. In 2018, 2.1 million women were newly diagnosed with breast cancer and approximately 627,000 died from this disease [50]. The current clinical approach is based on the use of compounds targeting a shared pathway between different types of cancer cells in the same tumor [51]. For example, common pathways, including Notch, human epidermal growth factor receptor 2 (Her-2), NF-κB, and signal transducer and activator of transcription 3 (STAT-3), are targets of various treatmentoptions used against breast cancer [52].
The Akt/mTOR-dependent pathway is a leading signaling pathway in the proliferation of breast cancer cells, and clinical evidence suggests that targeting of this pathway is a promising treatment option [53,54]. Curcumin was found to inhibit the phosphorylation of Akt, mTOR and their downstream proteins, resulting in cell cycle arrest of various breast cancer cell lines, including T47D and MCF7 [55].
NF-κB plays a key role in the proliferation, invasion, and metastasis of breast cancer cell lines. Curcumin was found to suppress the nuclear translocation of NF-κB and to downregulate the levels of p100 and p52 in MCF-7 and MDA-MB-453 breast cancer cells, respectively [56]. Curcumin was also observed to induce cell death in MCF-7 breast cancer cells via upregulation of the expression of the spermidine/spermine N1-acetyltransferase (SSAT) gene, which is strongly related to the NF-κB-dependent signaling pathway [57].
The autocrine growth hormone (GH) signaling pathway may promote breast cancer proliferation by inducing abnormal cell growth, metastasis, and drug resistance. Curcumin treatment prevented GH-mediated invasion and metastasis in T47D breast cancer cells through suppression of the miR-182-96-183 cluster expression and also induced downregulation in antiapoptotic proteins, including Bcl-2 and Bcl- xL in the same cell line [58].
Curcumin has shown promising results in counteracting the drug resistance commonly found in various types of cancer, including breast cancer. For example, Flap endonuclease 1 (FEN1) overexpression in breast cancer cell lines promotes resistance to the chemotherapeutic agent cisplatin. Curcumin was found to enhance the sensitivity of breast cancer cells to cisplatin by downregulating FEN1 expression in vitro [59]. Paclitaxel, another significant chemotherapeutic agent used in the treatment of breast cancer, has demonstrated acquired resistance in breast cancer cell lines, possibly through the overexpression of the multidrug resistance mutation 1 (MDR-1) gene and P-glycoprotein. Curcumin has shown promising results in decreasing this drug resistance by downregulating MDR-1 gene expression in MCF-7 breast cancer cells [60].

5. Prostate Cancer

Prostate cancer is currently the second cause of cancer death among men in the Western world [6]. The initiation and progression of the disease are primarily driven by androgen receptor (AR)-dependent signaling [61]. In recent years, clinical practice has been focused on the use of AR pathway inhibitors (ARPIs), such as abiraterone and enzalutamide, for the treatment of both metastatic and non-metastatic prostate cancer. Because various mechanisms of treatment resistance have emerged, new potent agents are being investigated as possible treatment options [62].
Several in vitro studies have identified curcumin as an effective agent in the treatment of prostate cancer. Curcumin interferes with various molecular signaling pathways, including the NF-κB, mitogen-activated protein kinase (MAPK) and epidermal growth factor (EGFR) pathways [63]. Abnormal NF-κB activity has been observed in human prostate cancer cells and xenografts [64]. During prostate carcinogenesis, the NF-κB pathway induces cancer cell survival, angiogenesis and metastasis, but also chemoresistance to several agents. Curcumin, acts on this pathway and prevents the NF-κB activation, thus downregulating cancer-related genes, including Bcl-2, Bcl-xL, IL-6, and COX-2 [65]. Moreover, curcumin downregulates C-X-C motif chemokine ligand 1 (CXCL-1) and CXCL-2 by targeting the NF-κB pathway in androgen independent prostate cancer (AIPC) cells [66].
In addition to these properties, curcumin has demonstrated the ability to suppress AR gene transcription in prostate cancer cells in vitro downregulating AR expression in the lymph node carcinoma of the prostate (LNCaP) cell line by inhibition of the expression of steroidogenic acute regulatory proteins, such as HSD3B2 and CYP11A1 [67]. AP-1 is a transcription factor that is primarily activated by MAPKs. The activation of this transcription factor in prostate cancer cells often leads to a poor clinical outcome [68]. Curcumin significantly hampers AP-1 protein in AIPC (PC-3) cells, thus suppressing tumor progression. A recent study used the LNCaP cell line to investigate the effects of curcumin in the JNK-dependent signaling pathway. Under curcumin treatment, JNK levels were reduced, as were the levels of epigenetic markers, including H3K4. These findings suggest that curcumin has the capability to affect the transcriptional regulation of genes by regulating the expression of epigenetic markers [69].
A variety of studies have examined the effects of curcumin in androgen-insensitive prostate cancer cells. Dorai et al. found that curcumin treatment results in a 60–80% inhibition of PC3-insensitive prostate cancer cell line development [70], whereas Deeb et al. found out that curcumin decreased viability of both PC3 and DU-145 prostate cancer cells in vitro [71]. Interestingly, in castration-resistant prostate cancer (CRPC), where patients are no longer responsive to androgen deprivation therapy, curcumin seems to play a significant role in preventing cancer development as well. Yang et al. reported that curcumin can induce apoptosis and protective autophagy in CRPC cells, and that this effect is closely related to its iron-chelating properties. [72] Chen et al. investigated the effects of different curcumin analogues in the treatment of castrate-resistant prostate cancer. They found that some of the analogues induced apoptosis and inhibited the expression of nuclear factor (NF)-κB, AKT and p-AKT and thus they are considered potent anticancer agents against this type of prostate cancer [73].

6. Brain Tumors

Several brain malignancies are among the most resistant tumors to all therapeutic modalities [74]. Glioblastoma (GBM) is the most common primary malignant central nervous system (CNS) tumor, accounting for 45.6% of primary malignant brain tumors [75]. Although curcumin has shown poor bioavailability due to its rapid metabolism, low stability and poor permeability to the BBB, in a free form [76], localized delivery of curcumin into brain with the use of nanoparticles has significantly increased its delivery into the targeted brain nuclei [77]. The effect of curcumin on glioma cells has been evaluated in vitro and found to possess inhibitory effects, which is enhanced by miR-378 [78]. MiR-378 belongs to a class of non-coding RNAs closely associated with post-transcriptional gene regulation and it is expressed at lower levels in different brain tumor tissues. In addition, curcumin decreases the proliferation of glioblastoma cells in vitro via inhibition of miR-21 which is a significant regulator in GBM progression [79].
GBM stem cells are a small population of tumor cells responsible for tumor progression, recurrence, and resistance to chemotherapeutic agents [80]. Curcumin affects both the differentiation and self-renewal of these stem cells, resulting in the prevention of GBM recurrence [81]. Curcumin causes an arrest in GBM stem cells by inhibition of both the STAT3 and the IAP-dependent pathway, along with activation of the MAPK pathway. Thus, curcumin can effectively decrease the malignant characteristics of GBM stem cells [82].
EGFR overexpression has been associated with several types of cancer, including malignant brain tumors. The overexpression of EGFR is closely related to several molecular pathways, including the JAK/STAT-dependent and PI3K/Akt-dependent pathways. [83] In several studies, curcumin caused significant dose-dependent antiproliferative effects on different glioblastoma cell lines in vitro, by inhibiting the overexpression of EGFR [84,85,86]. Bojko et al. reported the ability of curcumin to treat human GBM cells in combination with EGFR kinase inhibitors, such as tyrphostin AG1478. Curcumin induced the cytotoxic potential of AG1478 and when given together, those two compounds resulted in irreversible DNA damage and a significant decrease in the cell viability of GBM cells [87].

7. Pancreatic Cancer

Pancreatic cancer is a fatal condition and it accounts for 3% of all cancers. Its highly heterogeneous nature and high rates of metastasis make this type of malignancy difficult to treat despite novel treatment options [88,89]. The effect of curcumin has been studied, along with other phytochemicals, in the treatment of pancreatic cancer since it can effectively suppress different metastatic properties of cancer cells. Early studies revealed the ability of curcumin to inhibit both the phosphorylation of extracellular signal-related kinases and platelet-derived growth factor, thus resulting in the inhibition of pancreatic stellate cells (PSCs) [90]. More recent studies explored the ability of curcumin to suppress the formation of cancer stem cells (CSCs) which are responsible for the high proliferation rate and rapid tumor growth of pancreatic cancer with promising results in suppressing tumor growth [91,92].
Several pancreatic cell lines are susceptible to the effects of curcumin, with Panc-1, BXPC-3, and L3.6pl being the most well-studied. In 2015, Zhao et al. revealed that curcumin can induce apoptosis in various pancreatic cancer cells by inhibition of the PI3 K/Akt pathway and induction of forkhead box O1 [93]. Osterman et al. demonstrated the antitumor effects of curcumin in panc-1 cells through decreasing both IAP protein and mRNA expression [94]. Cell division cycle 20 (Cdc20) plays an important oncogenic role in the growth of pancreatic cancer [95] and higher expression of Cdc20 is closely related to a poor prognosis [96]. Curcumin can inhibit Cdc20 expression in PC cells through upregulation of both Bcl-2-like protein 11 (Bim) and p21, leading to cell apoptosis and decreased cell motility [97].

8. Gastric Cancer

Gastric cancer is the third most common cause of cancer-related death worldwide [98]. The pathogenesis and growth of gastric cancer are associated with several signaling molecules, genes and epigenetic patterns [99]. Recent evidence indicates promising anticancer effects of curcumin via its ability to inhibit several signal transduction pathways, such as the p53, Ras, Wnt-β, extracellular signal-regulated kinases (ERK), PI3K, MAPKs and protein kinase B (Akt) in gastric cancer cells [100].
Another mechanism of in vitro gastric cancer cell growth inhibition of curcumin is by downregulation of nuclear transcription factors such as NF-κB and reduced expression of pro-inflammatory cytokines, including TNF-α, interleukins and chemokines [101]. Moreover, it can induce p53 gene expression, promoting apoptosis of the malignant cells [102]. Jin et al. reported that curcumin can increase p53 gene expression with subsequent inhibition of hepatic stellate cell growth [103]. Based on this p53 effect, Fu and colleagues found that curcumin may indeed induce apoptosis in gastric cancer cells by activation of the p53-dependent signaling pathway [104].
Several studies have explored the role of the phosphatidylinositol-3 kinase (PI3K)-dependent signaling pathway on cell proliferation in different types of cancer [105]. Fu et al. reported a close relationship between the effects of curcumin on gastric cancer and the PI3K pathway in two gastric cancer cell lines, SGC-7901 and BGC-823 [104]. In addition, a recent study conducted by Liu et al. indicated that curcumin can inhibit the proliferation of the SGC7901, MKN45 and NCI N87 cells lines through inhibition of the Wnt3 a/β-catenin/EMT pathway, along with regulation of the Bcl-2 signaling and caspase pathways [106].

9. Leukemia

Leukemia accounts for 8% of all malignancies worldwide [107]. It is the most common type of childhood cancer, accounting for 30% of cancer in children [108]. Leukemia has been categorized in four main types: chronic myeloid leukemia (CML), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia (ALL) [109].
The pathogenesis of CML is based on the expression of P210 BCR-ABL protein, encoded by the Breakpoint Cluster Region-Abelson (BCR-ABL) gene. This protein takes part in the proliferation of progenitor cells through its connection with pathways such as the Ras/Raf/MAPK pathway [110]. Curcumin has been identified as a potent agent against CML through downregulation of p210 BCR-ABL, resulting in inhibition of the MAPK pathway [111]. In addition, curcumin increased the efficacy of the chemotherapeutic agent imatinib, through downregulation of both the p210 BCR-ABL protein and the heat shock protein 90 (Hsp90) [112]. NF-κB, a transcription factor responsible for the regulation of different genes [113], is inhibited by curcumin in KCL-22 myeloid cells, resulting in apoptosis. Curcumin also enhanced the anticancer properties of-TNFα-related apoptosis-inducing ligand (TRAIL) in the same cell line since both compounds effectively downregulate the NF-kB-dependent pathway [15].
AML is characterized by poor prognosis and resistance to chemotherapy [114]. The presence of CD34+ AML cells are partly responsible for the limited anticancer effects of chemotherapeutic agents. Rao et al. explored the effects of curcumin on CD34+ AML cell lines, and specifically KG1a and Kasumi-1 when given in combination with daunorubicin. Curcumin was shown to enhance the cytotoxic effects of daunorubicin and to suppress the expression of Bcl-2 protein [115]. FMS-like tyrosine kinase 3 (FLT3) is a well-known marker in AML and its overexpression has been found in other hematological cancers, including ALL. Tima and colleagues reported a dose-dependent effect of curcumin on the expression of this biomarker, suggesting that curcumin can decrease the levels of both FLT3 and STAT5A in AML cells [116]. Zhu et al. explored the molecular mechanism of action of curcumin on human M5 leukemia cells (SHI-1 cell line) and reported that this compound can inhibit the MAPK and matrix metalloproteinase (MMP)-dependent signaling pathways as a result of its anticancer effects [117].
CLL is currently the commonest hematological malignancy in the Western world [118]. The disease is characterized bydefective neoplastic B lymphocytes and elevated T cells and natural killer (NK) cells [119]. Curcumin is a potent agent against CLL since it can suppress pathways involved in the growth and survival of neoplastic B lymphocytes. Ghosh et al. found that curcumin inhibited the STAT3, AKT, and NF-κB-dependent pathways in vitro, and suppressed the expression of the X-linked inhibitor of apoptosis protein (XIAP) and Mcl-1 [120].
ALL is characterized by overexpression of adenosine diphosphate ribose (ADP-ribose) and polymerase-1 (PARP1) [121]. Curcumin has been reported to be a potent agent against ALL via cleavage of PARP1-dependent pathways [122]. It may also affect other signaling pathways associated with the development and growth of ALL tumor, including AKT/mTOR, STAT5, and RAF/MEK/ERK. Curcumin reduced the activation of both the AKT/mTOR- and ABL/STAT5-dependent pathways in vitro and downregulated the expression of BCR/ABL. The ability of curcumin to affect those pathways explains its synergistic antitumor effects when given along with imatinib in the treatment of ALL [123]. The effects of curcumin on different cell signaling pathways in different cancers are summarized in Table 1.

10. Clinical Trials

Curcumin has been investigated, or is currently under investigation, both as a monotherapy and in combination with other drugs in various clinical trials. In a phase 2 clinical trial, curcumin was used alone in 21 patients with advanced pancreatic cancer. After receiving 8 g curcumin orally daily for 8 weeks, two patients showed clinical evidence of biological activity. Specifically, one patient had ongoing stable disease for >18 months and the other experienced brief, but significant tumor regression (73%) [124]. In another phase 1 clinical trial, 14 patients with advanced or metastatic breast cancer were given a combination of docetaxel and curcumin. Patients received 100 mg/m2 of docetaxel as a 1 h i.v. infusion and doses of curcumin ranging from 500 mg/day until dose-limiting toxicity occurred. Patients received a combination of a standard dose of docetaxel and different doses of curcumin for 7 consecutive days. It was observed that a dose of 6g/day of curcumin in combination with a standard dose of docetaxel was effective and safe against advanced breast cancer [125].
Another phase I/II study investigated the efficacy and safety of oral administration of curcumin alone or in combination with bioperine in 29 patients with multiple myeloma (MM) Patients received escalated doses of curcumin (2, 4, 6, 8, or 12 g/day in 2 divided doses) alone or with 10 mg/day of bioperine for 12 weeks. Based on the expression of surrogate biomarkers, including NF-kB (p65), COX-2 and phospho-STAT3, the mixed treatment was more effective than bioperine alone and curcumin was not associated with any toxic effects [126].
In patients with advanced breast cancer, curcumin is under investigation as monotherapy (NCT03980509) or in combination with paclitaxel (NCT03072992). The main object of these clinical studies is to determine the effect of curcumin on the development of advanced breast cancer and to estimate the risk of adverse effects.
The use of curcumin in patients with low-risk, localized prostate cancer under active surveillance will be evaluated with the goal to slowing cancer progression (NCT03769766). The possible radio- sensitizing effect of curcumin in prostate patients undergoing radiotherapy, along with the role of curcumin as a radio-protector in normal tissues will also be explored (NCT02724618).
In 2019, the combination treatment of curcumin and Avastin was given to patients with colorectal cancer with unresectable metastasis with the aim of evaluating progression-free survival, and overall survival (NCT02439385).
The efficacy and safety of curcumin in patients with advanced cervical cancer will be investigated in a Phase 2 clinical trial in which the treatment response rate will be a major indicator of efficacy and the Common Terminology Criteria for Adverse Events (CTCAE) classification will be used as an index of the safety of the therapeutic scheme (NCT04294836).
The tolerability of a combination of curcumin and cholecalciferol in the treatment of patients with untreated stage 0–II small lymphocytic lymphoma (SLL) or CLL was evaluated in a phase 2 clinical trial. When given together, the two drugs suppressed the growth of cancer cells and increased the overall survival rate (NCT02100423).
Some of the most recent clinical trials on the use of curcumin as a therapeutic agent alone or in combination with chemotherapeutic agents in various cancers are reported in Table 2.

11. Curcumin and Cancer-Associated Fibroblasts/Tumor-Associated Fibroblasts (CAFs/TAFs)

Fibroblasts are significant components of connective tissue [127]. Both cancer-associated fibroblasts and tumor-associated fibroblasts (CAFs and TAFs, respectively) have been shown to induce tumor growth and development and are currently considered primary source of different cytokines and tumor-promoting growth factors [128].
Curcumin can inhibit the development of fibroblasts in a dose-dependent way. Zhang and colleagues studied the effect of curcumin in CAFs isolated from human oral mucosa and found that curcumin increased the expression of Bax and similar pro-apoptotic molecules [129]. Other studies have shown that curcumin can inhibit several molecules involved in the TGF-β-pathway-induced fibrosis, including NF-Kb and the p38 MAP kinase [130] and increase the expression of a variety of tumor-suppressor proteins, such as p21 and p53, thus leading to inhibition of tumor invasion and cell migration [131].
On prostate-CAFs, curcumin can increase the intracellular ROS levels and induce G2/M arrest [132], while in tongue squamous cell carcinoma, curcumin has been found to inhibit the secretion of pro-carcinogenic cytokines, including stromal cell-derived factor-1 (SDF-1) and matrix metalloproteinases 2 (MMP2) [133].

12. Potential Side Effects of Curcumin

Curcumin has been characterized as “generally safe” by the US Food and Drug Administration (FDA) [134]. Indeed, no significant side effects related to curcumin can be found in the literature. Some of the documented cases are of reversible side effects, including allergic dermatitis [135]. Dose-escalating studies have demonstrated that consumption of up to 12 g of curcumin daily presents no damaging effects [136]. In studies on patients with solid tumors, no adverse effects were reported when curcumin was given for 8 weeks at a dosage of 900 mg/day orally, except for mild gastrointestinal upset [137]. Oral intake of 6 g/day of curcumin for 7 weeks was also reported to be safe in patients with breast cancer, and 3 g/day of curcumin given for 9 weeks to patients with prostate cancer showed no adverse effects [138,139].
Curcumin also exhibits a strong iron-chelating activity [140]. Long-term supplementation with curcumin induced iron depletion in young mice and this effect was further enhanced when they were fed with diets containing low iron concentrations [141]. Apart from that, curcumin possesses anticoagulant properties and may increase bleeding time in patients receiving anticoagulants [142].
Despite the absence of overt adverse effects of curcumin, it has been reported that this compound can inhibit several cytochrome P450 subtypes, including CYP2C9 and CYP3A4 [143,144]. For this reason, curcumin has been known to interact with certain other medications, including anticoagulants, antibiotics and antidepressants. For example, curcumin was found to affect the pharmacokinetics of warfarin and clopidogrel in Wistar rats and to increase the elimination half-life and volume of distribution of norfloxacin in New Zealand white rabbits [145,146].

13. Curcumin as Chemoprotective Agent in Cancer Chemotherapy

Curcumin has been found to inhibit some of the most significant chemotherapy-induced side effects. When given in combination with cisplatin, curcumin increased the levels of superoxide dismutase (SOD), an enzyme responsible for hepatoprotection [147]. In doxorubicin-treated cells, curcumin was found to upregulate the expression of SOD, and downregulate the expression of cardiotoxic marker SCK [148]. In cisplatin-treated cells, curcumin managed to diminish nephrotoxicity mainly through the downregulation of creatinine expression [149].
Interestingly, when a lecithin delivery system of curcumin was evaluated in a study containing 160 patients with solid tumors, in order to determine its efficacy in alleviating the side effects of chemotherapeutic drugs, results showed that curcumin can prevent the appearance of side effects related to cytostatic agents. That effect was mainly attributed to the ability of curcumin to downregulate inflammatory pathways [150].

14. Curcumin Application: Limitations and Prospects

Despite its efficacy and safety, curcumin displays poor solubility in water, a lowly pharmacokinetic profile and serious instability. For those reasons, the therapeutic potential of this compound is still under review. Even when administrated at doses of 12 g/day, the bioavailability of curcumin remains exceedingly poor [76]. Moreover, its oral bioavailability is low due to its low absorption by small intestine, extensive phase I and II biotransformation and quick elimination through the gall bladder [151].
A series of studies have been conducted to assess curcumin absorption after oral administration. Lao et al. performed a dose escalation study in which 24 healthy volunteers were given escalating doses of curcumin, ranging from 500 to 12,000 mg. Curcumin was not detectable in the serum of those participants when given at a dosage lower than 8 g [136]. In another study where healthy volunteers (n = 12) were also used, curcumin was administered at doses of 10 or 12 g. After 30 min, free curcumin could be detected in the plasma of only one volunteer [152]. The bioavailability of oral curcumin was also examined in patients with pancreatic cancer, where 21 patients received 8 g curcumin orally in combination with the chemotherapeutic agent gemcitabine. Total curcumin levels were found at a range of 29 to 91 ng/mL [153].
Another study on refractory colorectal cancer included 15 patients who were given daily curcumin doses ranging from 0.45 and 3.6 g for up to 4 months. Curcumin as well as its glucuronide and sulfate metabolites were detected in plasma samples after a dose of 3.6 g/day at the following concentrations: 11.1 ± 0.6, 15.8 ± 0.9 and 8.9 ± 0.7, respectively [154]. Despite its poor oral bioavailability, curcumin can cross the blood–brain barrier (BBB) thanks to its lipophilic nature. However, only a few studies in murine models have been able to assess its brain concentration. In one study, mice were administered 50 mg/kg curcumin orally and displayed brain curcumin concentrations lower than the limit of detection even 120 min after administration. However, when 100 mg/kg curcumin was given via intraperitoneal injection, total concentration ranged between 4 and 5 µg/g tissue [155].
In order to overcome poor curcumin absorption, and low bioavailability, different strategies, mainly in the form of developing novel oral delivery systems, have been made. Those strategies aim to increase curcumin solubility, improve its pharmacokinetic profile and enhance cellular uptake [156].
A water-soluble curcumin formulation consisting of turmeric extract, cellulosic derivatives and a widely-used hydrophilic carrier was made and compared to standard curcumin treatment in healthy volunteers. This formulation managed to increase the solubility of curcumin considerably and thus provide a 46-fold increase in its oral absorption [157].
Curcumin in a lipid-based formulation containing nanostructured lipid carriers is another promising way of increasing the bioavailability of curcumin. When a solid lipid curcumin particle (SLCP) formulation was given in both healthy volunteers and late-stage osteosarcoma patients, plasma curcumin levels were detectable in both healthy individuals and osteosarcoma patients as opposed to unformulated curcumin [158].
In a single-blind crossover study, healthy volunteers were given 500 mg of either unformulated curcumin, or liquid micelles or micronized curcumin. Both liquid micelles and micronized powder showed a relatively higher curcumin plasma concentration compared to the unformulated compound (3228, 41.6 and 7.1 nmol/L, respectively) [159].
In another study, SBA mesoporous silica 15 was used in order to prepare inhalable curcumin and investigate its application in lung cancer. The therapeutic effect of this inhalation system was evaluated and it was confirmed that the inclusion of curcumin in this mesoporous material increased the compound’s bioavailability and had a certain inhibitory effect on a metastatic lung mouse model. In addition, curcumin loaded in SBA silica 15 was more effective in reducing the number of metastatic lung tumors in the lung mouse model compared to treatment with curcumin alone [160].
In order to increase curcumin bioavailability, it is also important to protect it from physical and chemical damage due to its unstable nature under neutral or alkaline conditions. A well-studied technique is based on the use of metallocomplexes of curcumin, including complexation of curcumin with divalent ions, such as Zn2+, and Cu2+ [161]. Zebib and colleagues, evaluated the stability of curcumin complexes with divalent ions when prepared in a glycerol/water solvent and found that its stability was indeed significantly enhanced in vitro compared to curcumin alone [162].
In another study aimed to increase curcumin physicochemical stability, Meng et al. used zein/carboxymethyl dextrin nanoparticles to encapsulate curcumin. The results showed that encapsulation of curcumin in zein/CMD nanoparticles increased its stability and delayed its release in simulated gastrointestinal fluids [163].

15. Conclusions

Many challenges accompany the research on new drugs for use against malignant diseases. Various natural products have attracted the notice of researchers as possible chemotherapeutic agents, thanks to their efficacy and safety. Curcumin, an active hydrophobic polyphenol extracted from the rhizomes of the plant Curcuma longa, has been investigated thoroughly and has exhibited an important role in the treatment of various health conditions, including several types of cancer. As reported in this review, curcumin targets multiple signaling pathways involved in the initiation, development, and growth of tumors. Growth factors, transcription factors, protein kinases, cytokines, and genes taking part in apoptosis are the molecular targets of curcumin, which appears to significantly affect the development of various different malignancies. However, further in vitro studies and clinical trials in humans are needed to determine the complete mechanism of action of this compound in each different type of cancer, while ensuring its safety for human use.

Author Contributions

Conceptualization, G.A.A., G.D.L., A.P.K. and V.G.; writing—original draft, V.Z., V.G., G.A.A., G.D.L. and A.P.K.; writing—review and editing, G.A.A., A.P.K., G.D.L., S.V. and V.G.; visualization and supervision, G.A.A., G.D.L., A.P.K. and V.G.; methodology, V.Z., G.A.A., G.D.L. and S.V.; data curation, V.Z., G.A.A. and V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Priyadarsini, K.I. The chemistry of curcumin: From extraction to therapeutic agent. Molecules 2014, 19, 20091–20112. [Google Scholar] [CrossRef] [Green Version]
  2. Dutta, B. Study of secondary metabolite constituents and curcumin contents of six different species of genus curcuma. J. Med. Plants 2015, 3, 116–119. [Google Scholar]
  3. Kyritsis, A.; Bondy, M.; Levin, V. Modulation of Glioma Risk and Progression by Dietary Nutrients and Antiinflammatory Agents. Nutr. Cancer 2011, 63, 174–184. [Google Scholar] [CrossRef] [PubMed]
  4. Heling’s, S.J.; Kalman, D.S. Curcumin: A Review of Its’ Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  5. World Health Organization. Global Action against Cancer; World Health Organization: Geneva, Switzerland, 2005. [Google Scholar]
  6. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  7. Pentimalli, F.; Grelli, S.; Daniele, N.; Melino, G.; Amelio, I. Cell death pathologies: Targeting death pathways and the immune system for cancer therapy. Genes Immun. 2019, 20, 539–554. [Google Scholar] [CrossRef]
  8. Murata, M. Inflammation and cancer. Environ. Health Prev. Med. 2018, 23, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Negrini, S.; Gorgoulis, V.; Halazonetis, T. Genomic instability—An evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 220–228. [Google Scholar] [CrossRef] [PubMed]
  10. Mohamed, S.I.A.; Jantan, I.; Haque, M.A. Naturally occurring immunomodulators with antitumor activity: An insight on their mechanisms of action. Int. Immunopharmacol. 2017, 50, 291–304. [Google Scholar] [CrossRef]
  11. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, 453–462. [Google Scholar] [CrossRef] [Green Version]
  12. Nakamae, I.; Morimoto, T.; Shima, H.; Shionyu, M.; Fujiki, H.; Yoneda-Kato, N.; Yokoyama, T.; Kanaya, S.; Kakiuchi, K.; Shirai, T.; et al. Curcumin Derivatives Verify the Essentiality of ROS Upregulation in Tumor Suppression. Molecules 2019, 24, 4067. [Google Scholar] [CrossRef] [Green Version]
  13. Sethi, G.; Tergaonkar, V. Potential pharmacological control of the NF-κB pathway. Trends Pharmacol. Sci. 2009, 30, 313–321. [Google Scholar] [CrossRef]
  14. Ghasemi, F.; Shafiee, M.; Banikazemi, Z.; Pourhanifeh, M.H.; Khanbabaei, H.; Shamshirian, A.; Amiri-Moghadam, S.; ArefNezhad, R.; Sahebkar, A.; Avan, A.; et al. Curcumin inhibits NF-kB and Wnt/β-catenin pathways in cervical cancer cells. Pathol. Res. Pract. 2019, 215, 152556. [Google Scholar] [CrossRef] [PubMed]
  15. Iqbal, B.; Ghildiyal, A.; Singh, S.; Siddiqui, S.; Kumari, P.; Arshad, M.; Mahdi, A. A Combinatorial effect of curcumin and tumor necrosis factor-α-related apoptosis-inducing ligand (TRAIL) in induction of apoptosis via inhibition of nuclear factor kappa activity and enhancement of caspase-3 activity in chronic myeloid cells: An in-vitro study. J. Cancer Res. Ther. 2018, 14, 1193–1200. [Google Scholar]
  16. Bessard, A.; Sole, V.; Bouchaud, G.; Quemener, A.; Jacques, Y. High antitumor activity of RLI, an interleukin-15 (IL-15)-IL-15 receptor α fusion protein, in metastatic melanoma and colorectal cancer. Mol. Cancer Ther. 2009, 8, 2736–2745. [Google Scholar] [CrossRef] [Green Version]
  17. Lee, S.; Margolin, K. Cytokines in cancer immunotherapy. Cancers 2011, 3, 3856–3893. [Google Scholar] [CrossRef]
  18. Das, L.; Vinayak, M. Curcumin Modulates Glycolytic Metabolism and Inflammatory Cytokines via Nrf 2 in Dalton’s Lymphoma Ascites Cells In Vivo. Anticancer Agents Med. Chem. 2018, 18, 1779–1791. [Google Scholar] [CrossRef] [PubMed]
  19. Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Roy, N.K.; Prasad, S.; Aggarwal, B.B. Curcumin, the golden nutraceutical: Multitargeting for multiple chronic diseases. Br. J. Pharmacol. 2017, 174, 1325–1348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Yu, Y.; Shen, Q.; Lai, Y.; Park, S.Y.; Ou, X.; Lin, D.; Jin, M.; Zhang, W. Anti-inflammatory Effects of Curcumin in Microglial Cells. Front Pharmacol. 2018, 9, 386. [Google Scholar] [CrossRef] [Green Version]
  21. Srivastava, N.S.; Srivastava, R.A.K. Curcumin and quercetin synergistically inhibit cancer cell proliferation in multiple cancer cells and modulate Wnt/β-catenin signaling and apoptotic pathways in A375 cells. Phytomedicine 2019, 52, 117–128. [Google Scholar] [CrossRef] [PubMed]
  22. Jagetia, G.C.; Aggarwal, B.B. ‘’Spicing Up” of the Immune System by Curcumin. J. Clin. Immunol. 2007, 27, 19–35. [Google Scholar] [CrossRef] [PubMed]
  23. Lantz, R.C.; Chena, G.J.; Solyomb, A.M.; Jolad, S.D.; Timmermann, B.N. The effect of turmeric extracts on inflammatory mediator production. Phytomedicine 2005, 12, 445–452. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, G.Y.; Kim, K.H.; Lee, S.H.; Yoon, M.S.; Lee, H.J.; Moon, D.O.; Lee, C.M.; Ahn, S.C.; Park, Y.C.; Park, Y.M. Curcumin inhibits immunostimulatory function of dendritic cells: MAPKs and translocation of NF-kappa B as potential targets. J. Immunol. 2005, 174, 8116–8124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. South, E.H.; Exon, J.H.; Hendrix, K. Dietary curcumin enhances antibody response in rats. Immunopharmacol. Immunotoxicol. 1997, 19, 105–119. [Google Scholar] [CrossRef] [PubMed]
  26. Yadav, V.S.; Mishra, K.P.; Singh, D.P.; Mehrotra, S.; Singh, V.K. Immunomodulatory effects of curcumin. Immunopharmacol. Immunotoxicol. 2005, 27, 485–497. [Google Scholar] [CrossRef] [PubMed]
  27. Hasanzadeh, S.; Read, M.I.; Bland, A.R.; Majeed, M.; Jamialahmadi, T.; Sahebkar, A. Curcumin: An inflammasome silencer. Pharmacol. Res. 2020, 159, 104921. [Google Scholar] [CrossRef] [PubMed]
  28. Kang, T.B.; Yang, S.H.; Toth, B.; Kovalenko, A.; Wallach, D. Activation of the NLRP3 inflammasome by proteins that signal for necroptosis. Methods Enzymol. 2014, 545, 67–81. [Google Scholar]
  29. Yin, H.; Guo, Q.; Li, X.; Tang, T.; Li, C.; Wang, H.; Sun, Y.; Feng, Q.; Ma, C.; Gao, C.; et al. Curcumin suppresses IL-1β secretion and prevents inflammation through inhibition of the NLRP3 inflammasome. J. Immunol. 2018, 200, 2835–2846. [Google Scholar] [CrossRef] [Green Version]
  30. Miller, J.M.; Thompson, J.K.; MacPherson, M.B.; Beuschel, S.L.; Westbom, C.M.; Sayan, M.; Shukla, A. Curcumin: A double hit on malignant mesothelioma. Cancer Prev. Res. 2014, 7, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Reuter, S.; Charlet, J.; Juncker, T.; Teiten, M.H.; Dicato, M.; Diederich, M. Effect of curcumin on nuclear factor κB signaling pathways in human chronic myelogenous K562 leukemia cells. Ann. N. Y. Acad. Sci. 2009, 1171, 436. [Google Scholar] [CrossRef] [PubMed]
  32. Panahi, Y.; Saadat, A.; Beiraghdar, F.; Sahebkar, A. Adjuvant Therapy with Bioavailability-Boosted Curcuminoids Suppresses Systemic Inflammation and Improves Quality of Life in Patients with Solid Tumors: A Randomized Double-Blind Placebo-Controlled Trial. Phytother. Res. 2014, 28, 1461–1467. [Google Scholar] [CrossRef] [PubMed]
  33. Basak, S.K.; Bera, A.; Yoon, A.J.; Morselli, M.; Jeong, C.; Tosevska, A.; Dong, T.S.; Eklund, M.; Russ, E.; Nasser, H.; et al. A randomized, phase 1, placebo-controlled trial of APG-157 in oral cancer demonstrates systemic absorption and an inhibitory effect on cytokines and tumor-associated microbes. Cancer 2020, 8, 1668–1682. [Google Scholar] [CrossRef]
  34. Memon, H.; Patel, B. Immune checkpoint inhibitors in non-small cell lung cancer: A bird’s eye view. Life Sci. 2019, 233, 116713. [Google Scholar] [CrossRef]
  35. Wang, J.W.; Wang, X.; Wang, X.J.; Zheng, B.Z.; Wang, Y.; Liang, B. Curcumin inhibits the growth via Wnt/β-catenin pathway in non-small-cell lung cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7492–7499. [Google Scholar]
  36. Li, X.; Ma, S.; Yang, P.; Sun, B.; Zhang, Y.; Sun, Y.; Hao, M.; Mou, R.; Jia, Y. Anticancer effects of curcumin on nude mice bearing lung cancer A549 cell subsets SP and NSP cells. Oncol. Lett. 2018, 16, 6756–6762. [Google Scholar] [CrossRef]
  37. Wu, G.Q.; Chai, K.Q.; Zhu, X.M.; Jiang, H.; Wang, X.; Xue, Q.; Zheng, A.H.; Zhou, H.Y.; Chen, Y.; Chen, X.C.; et al. Anti-cancer effects of curcumin on lung cancer through the inhibition of EZH2 and NOTCH1. Oncotarget 2016, 7, 26535–26550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Wang, C.; Song, X.; Shang, M.; Zou, W.; Zhang, M.; Wei, H.; Shao, H. Curcumin exerts cytotoxicity dependent on reactive oxygen species accumulation in non-small-cell lung cancer cells. Future Oncol. 2019, 15, 1243–1253. [Google Scholar] [CrossRef]
  39. Smagurauskaite, G.; Mahale, J.; Brown, K.; Thomas, A.L.; Howells, L.M. New paradigms to assess consequences of long-term, low-dose curcumin exposure in lung cancer cells. Molecules 2020, 25, 366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Bland, A.R.; Bower, R.L.; Nimick, M.; Hawkins, B.C.; Rosengren, R.J.; Ashton, J.C. Cytotoxicity of curcumin derivatives in ALK positive non-small cell lung cancer. Eur. J. Pharmacol. 2019, 865, 172749. [Google Scholar] [CrossRef] [PubMed]
  41. Li, X.; He, S.; Tian, Y.; Weiss, R.M.; Martin, D.T. Synergistic inhibition of GP130 and ERK signaling blocks chemoresistant bladder cancer cell growth. Cell. Signal. 2019, 63, 109381. [Google Scholar] [CrossRef]
  42. Jin, X.; Wang, J.; Shen, H.; Ran, R.; Xu, K.; Zhang, W.; Tong, X.; Feng, L. Curcumin co-treatment ameliorates resistance to gefitinib in drug-resistant NCI-H1975 lung cancer cells. J. Tradit. Chin. Med. 2017, 37, 355–360. [Google Scholar]
  43. Yoon, J.H.; Shin, J.W.; Pham, T.H.; Choi, Y.J.; Ryu, H.W.; Oh, S.R.; Oh, J.W.; Yoon, D.Y. Methyl lucidone induces apoptosis and G2/M phase arrest via the PI3K/Akt/NF-κB pathway in ovarian cancer cells. Pharm. Biol. 2020, 58, 51–59. [Google Scholar] [CrossRef] [Green Version]
  44. Gao, X.; Li, X.; Ho, C.T.; Lin, X.; Zhang, Y.; Li, B.; Chen, Z. Cocoa tea (Camellia ptilophylla) induces mitochondria-dependent apoptosis in HCT116 cells via ROS generation and PI3K/Akt signaling pathway. Food Res. Int. 2020, 129, 108854. [Google Scholar] [CrossRef]
  45. Zhou, G.Z.; Li, A.F.; Sun, Y.H.; Sun, G.C. A novel synthetic curcumin derivative MHMM-41 induces ROS-mediated apoptosis and migration blocking of human lung cancer cells A549. Biomed. Pharmacother. 2018, 103, 391–398. [Google Scholar] [CrossRef]
  46. Agudo, A.; Bonet, C.; Travier, N.; González, C.A.; Vineis, P.; Bueno-de-Mesquita, H.B.; Trichopoulos, D.; Boffetta, P.; Clavel-Chapelon, F.; Boutron-Ruault, M.C.; et al. Impact of cigarette smoking on cancer risk in the European prospective investigation into cancer and nutrition study. J. Clin. Oncol. 2012, 30, 4550–4557. [Google Scholar] [CrossRef] [PubMed]
  47. Liang, Z.; Xie, W.; Wu, R.; Geng, H.; Zhao, L.; Xie, C.; Li, X.; Zhu, M.; Zhu, W.; Zhu, J.; et al. Inhibition of tobacco smoke-induced bladder MAPK activation and epithelial-mesenchymal transition in mice by curcumin. Int. J. Clin. Exp. Pathol. 2015, 8, 4503–4513. [Google Scholar]
  48. Liang, Z.; Wu, R.; Xie, W.; Zhu, M.; Xie, C.; Li, X.; Zhu, J.; Zhu, W.; Wu, J.; Geng, S.; et al. Curcumin reverses tobacco smoke-induced epithelial-mesenchymal transition by suppressing the MAPK pathway in the lungs of mice. Mol. Med. Rep. 2018, 17, 2019–2025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Xie, C.; Zhu, J.; Yang, X.; Huang, C.; Zhou, L.; Meng, Z.; Li, X.; Zhong, C. TAp63α Is Involved in Tobacco Smoke-Induced Lung Cancer EMT and the Anti-Cancer Activity of Curcumin via miR-19 Transcriptional Suppression. Front. Cell Dev. Biol. 2021, 9, 645402. [Google Scholar] [CrossRef]
  50. Bray, F.; Ferlay, J.; Soerjomataram, I. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  51. Lheureux, S.; Denoyelle, C.; Ohashi, P.S.; De Bono, J.S.; Mottaghy, F.M. Molecularly targeted therapies in cancer: A guide for the nuclear medicine physician. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 41–54. [Google Scholar] [CrossRef] [PubMed]
  52. Januškevičienė, I.; Petrikaitė, V. Heterogeneity of breast cancer: The importance of interaction between different tumor cell popu lations. Life Sci. 2019, 239, 117009. [Google Scholar] [CrossRef]
  53. Yang, S.X.; Polley, E.; Lipkowitz, S. New insights on PI3K/AKT pathway alterations and clinical outcomes in breast cancer. Cancer Treat. Rev. 2016, 45, 87–96. [Google Scholar] [CrossRef] [Green Version]
  54. Steelman, L.S.; Martelli, A.M.; Cocco, L.; Libra, M.; Nicoletti, F.; Abrams, S.L.; McCubrey, J.A. The therapeutic potential of mTOR inhibitors in breast cancer. Br. J. Clin. Pharmacol. 2016, 82, 1189–1212. [Google Scholar] [CrossRef]
  55. Shan, H.; Yingchun, X.; Liwei, M.; Liming, H.; He, S. Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells. Exp. Ther. Med. 2018, 16, 1266–1272. [Google Scholar]
  56. Coker-Gurkan, A.; Celik, M.; Ugur, M.; Arisan, E.D.; Obakan-Yerlikaya, P.; Durdu, Z.B.; Palavan-Unsal, N. Curcumin inhibits autocrine growth hormone-mediated invasion and metastasis by targeting NF-κB signaling and polyamine metabolism in breast cancer cells. Amino Acids 2018, 50, 1045–1069. [Google Scholar] [CrossRef] [PubMed]
  57. Berrak, O.; Akkoc, Y.; Arisan, E.D.; Coker-Gurkan, A.; Obakan-Yerlikaya, P.; Palavan-Unsal, N. The inhibition of PI3K and NF-κB promoted curcumin-induced cell cycle arrest at G2/M via altering polyamine metabolism in Bcl-2 overexpressing MCF-7 breast cancer cells. Biomed. Pharm. 2016, 77, 150–160. [Google Scholar] [CrossRef] [PubMed]
  58. Coker-Gurkan, A.; Bulut, D.; Genc, R.; Arisan, E.D.; Obakan-Yerlikaya, P.; Palavan-Unsal, N. Curcumin prevented human autocrine growth hormone (GH) signaling mediated NF-κB activation and miR-183-96-182 cluster stimulated epithelial mesenchymal transition in T47D breast cancer cells. Mol. Biol. Rep. 2019, 46, 355–369. [Google Scholar] [CrossRef]
  59. Zou, J.; Zhu, L.; Jiang, X.; Wang, Y.; Wang, X.; Chen, B. Curcumin increases breast cancer cell sensitivity to cisplatin by decreasing FEN1 expression. Oncotarget 2018, 9, 11268–11278. [Google Scholar] [CrossRef] [Green Version]
  60. Attia, Y.M.; El-Kersh, D.M.; Ammar, R.A.; Adel, A.; Khalil, A.; Walid, H.; Eskander, K.; Hamdy, M.; Reda, N.; Mohsen, N.E.; et al. Inhibition of aldehyde dehydrogenase-1 and p-glycoprotein-mediated multidrug resistance by curcumin and vitamin D3 increases sensitivity to paclitaxel in breast cancer. Chem. Biol. Interact. 2020, 315, 108865. [Google Scholar] [CrossRef] [PubMed]
  61. Lonergan, P.E.; Tindall, D.J. Androgen receptor signaling in prostate cancer development and progression. J. Carcinog. 2010, 10, 20. [Google Scholar]
  62. Watson, P.A.; Arora, V.K.; Sawyers, C.L. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat. Rev. Cancer 2015, 15, 701–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Tomeh, M.A.; Hadianamrei, R.; Zhao, X. A review of curcumin and its derivatives as anticancer agents. Int. J. Mol. Sci. 2019, 20, 1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Thapa, D.; Ghosh, R. Chronic inflammatory mediators enhance prostate cancer development and progression. Biochem. Pharmacol. 2015, 94, 53–62. [Google Scholar] [CrossRef] [PubMed]
  65. Hayden, M.S.; Ghosh, S. NF-κB, the first quarter-century: Remarkable progress and outstanding questions. Genes Dev. 2012, 26, 203–234. [Google Scholar] [CrossRef] [Green Version]
  66. Killian, P.H.; Kronski, E.; Michalik, K.M.; Barbieri, O.; Astigiano, S.; Sommerhoff, C.P.; Pfeffer, U.; Nerlich, A.G.; Bachmeier, B.E. Curcumin inhibits prostate cancer metastasis in vivo by targeting the inflammatory cytokines CXCL1 and-2. Carcinogenesis 2012, 33, 2507–2519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Ide, H.; Lu, Y.; Noguchi, T.; Muto, S.; Okada, H.; Kawato, S.; Horie, S. Modulation of AKR 1C2 by curcumin decreases testosterone production in prostate cancer. Cancer Sci. 2018, 109, 1230–1238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kavya, K.; Kumar, M.N.; Patil, R.H.; Hegde, S.M.; Kiran-Kumar, K.M.; Nagesh, R.; Babu, R.L.; Ramesh, G.T.; Chidananda-Sharma, S. Differential expression of AP-1 transcription factors in human prostate LNCaP and PC-3 cells: Role of Fra-1 in transition to CRPC status. Mol. Cell. Biochem. 2017, 433, 13–26. [Google Scholar] [CrossRef] [Green Version]
  69. Wanli, Z.; Xudong, Z.; Guisong, Q.; Yuexian, G. Curcumin suppressed the prostate cancer by inhibiting JNK pathways via epigenetic regulation. J. Biochem. Mol. Toxicol. 2018, 32, e22049. [Google Scholar]
  70. Dorai, T.; Gehani, N.; Katz, A. Therapeutic potential of curcumin in human prostate cancer-I. curcumin induces apoptosis in both androgen-dependent and androgen-independent prostate cancer cells. Prostate Cancer Prostatic Dis. 2000, 3, 84–93. [Google Scholar] [CrossRef] [Green Version]
  71. Deeb, D.; Jiang, H.; Gao, X.; Hafner, M.S.; Wong, H.; Divine, G.; Chapman, R.A.; Dulchavsky, S.A.; Gautam, S.C. Curcumin sensitizes prostate cancer cells to tumor necrosis factor-related apoptosis-inducing ligand/Apo2L by inhibiting nuclear factor-kappaB through suppression of IkappaBalpha phosphorylation. Mol. Cancer Ther. 2004, 3, 803–812. [Google Scholar]
  72. Yang, C.; Ma, X.; Wang, Z.; Zeng, X.; Hu, Z.; Ye, Z.; Shen, G. Curcumin induces apoptosis and protective autophagy in castration-resistant prostate cancer cells through iron chelation. Drug Des. Dev. Ther. 2017, 11, 431–439. [Google Scholar] [CrossRef] [Green Version]
  73. Chen, S.; Nimick, M.; Cridge, A.G.; Hawkins, B.C.; Rosengren, R.J. Anticancer potential of novel curcumin analogs towards castrate-resistant prostate cancer. Int. J. Oncol. 2018, 52, 579–588. [Google Scholar] [CrossRef]
  74. Klinger, N.V.; Mittal, S. Therapeutic Potential of Curcumin for the Treatment of Brain Tumors. Oxid. Med. Cell. Longev. 2016, 2016, 9324085. [Google Scholar] [CrossRef]
  75. Weathers, S.P.; Gilbert, M.R. Advances in treating glioblastoma. F1000Prime Rep. 2014, 6, 46. [Google Scholar] [CrossRef]
  76. Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of curcumin: Problems and promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, N.; Yan, F.; Liang, X.; Wu, M.; Shen, Y.; Chen, M.; Xu, Y.; Zou, G.; Jiang, P.; Tang, C.; et al. Localized delivery of curcumin into brain with polysorbate 80-modified cerasomes by ultrasound-targeted microbubble destruction for improved Parkinson’s disease therapy. Theranostics 2018, 8, 2264–2277. [Google Scholar] [CrossRef] [Green Version]
  78. Li, W.; Yang, W.; Liu, Y.; Chen, S.; Chin, S.; Qi, X.; Zhao, Y.; Liu, H.; Wang, J.; Mei, X.; et al. MicroRNA-378 enhances inhibitory effect of curcumin on glioblastoma. Oncotarget 2017, 8, 73938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Yeh, W.L.; Lin, H.Y.; Huang, C.Y.; Huang, B.R.; Lin, C.; Lu, D.Y.; Wei, K.C. Migration-prone glioma cells show curcumin resistance associated with enhanced expression of miR-21 and invasion/anti-apoptosis-related proteins. Oncotarget 2015, 6, 37770. [Google Scholar] [CrossRef]
  80. Lathia, J.D.; Mack, S.C.; Mulkearns-Hubert, E.E.; Valentim, C.L.; Rich, J.N. Cancer stem cells in glioblastoma. Genes Dev. 2015, 29, 1203–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Zhuang, W.; Long, L.; Zheng, B.; Ji, W.; Yang, N.; Zhang, Q.; Liang, Z. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Sci. 2012, 103, 684–690. [Google Scholar] [CrossRef] [PubMed]
  82. Gersey, C.; Rodriguez, G.A.; Barbarite, E.; Sanchez, A.; Walters, W.M.; Ohaeto, K.C.; Komotar, R.J.; Graham, R.M. Curcumin decreases malignant characteristics of glioblastoma stem cells via induction of reactive oxygen species. BMC Cancer 2017, 17, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hatanpaa, K.J.; Burma, S.; Zhao, D.; Habib, A.A. Epidermal growth factor receptor in glioma: Signal transduction, neuropathology, imaging, and radioresistance. Neoplasia 2010, 12, 675–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Su, C.C.; Wang, M.J.; Chiu, T.L. The anti-cancer efficacy of curcumin scrutinized through core signaling pathways in glioblastoma. Int. J. Mol. Med. 2010, 26, 217–224. [Google Scholar]
  85. Zhao, J.; Zhu, J.; Lv, X.; Xing, J.; Liu, S.; Chen, C.; Xu, Y. Curcumin potentiates the potent antitumor activity of ACNU against glioblastoma by suppressing the PI3K/AKT and NF-kappaB/COX-2 signaling pathways. OncoTargets Ther. 2017, 10, 5471–5482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Zanotto-Filho, A.; Braganhol, E.; Klafke, K.; Figueiro, F.; Terra, S.R.; Paludo, F.J.; Morrone, M.; Bristot, I.J.; Battastini, A.M.; Forcelini, C.M.; et al. Autophagy inhibition improves the efficacy of curcumin/temozolomide combination therapy in glioblastomas. Cancer Lett. 2015, 358, 220–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bojko, A.; Cierniak, A.; Adamczyk, A.; Ligeza, J. Modulatory effects of curcumin and tyrphostins (AG494 and AG1478) on growth regulation and viability of LN229 human brain cancer cells. Nutr. Cancer 2015, 67, 1170–1182. [Google Scholar] [CrossRef]
  88. Oettle, H. Progress in the knowledge and treatment of advanced pancreatic cancer: From benchside to bedside. Cancer Treat. Rev. 2014, 40, 1039–1047. [Google Scholar] [CrossRef] [Green Version]
  89. Epelbaum, R.; Schaffer, M.; Vizel, B.; Badmaev, V.; Bar-Sela, G. Curcumin and gemcitabine in patients with advanced pancreatic cancer. Nutr. Cancer 2010, 62, 1137–1141. [Google Scholar] [CrossRef]
  90. Masamune, A.; Suzuki, N.; Kikuta, K.; Satoh, M.; Satoh, K.; Shimosegawa, T. Curcumin blocks activation of pancreatic stellate cells. J. Cell. Biochem. 2006, 97, 1080–1093. [Google Scholar] [CrossRef]
  91. Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bao, B.; Ali, S.; Banerjee, S.; Wang, Z.; Logna, F.; Azmi, A.S.; Kong, D.; Ahmad, A.; Li, Y.; Padhye, S.; et al. Curcumin analogue CDF inhibits pancreatic tumor growth by switching on suppressor microRNAs and attenuating EZH2 expression. Cancer Res. 2012, 72, 335–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhao, Z.; Li, C.; Xi, H.; Gao, Y.; Xu, D. Curcumin induces apoptosis in pancreatic cancer cells through the induction of forkhead box O1 and inhibition of the PI3 K/Akt pathway. Mol. Med. Rep. 2015, 12, 5415–5422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Díaz-Osterman, C.J.; Gonda, A.; Stiff, T.; Sigaran, U.; Valenzuela, M.M.A.; Bennit, H.R.; Moyron, R.B.; Khan, S.; Wall, N.R. Curcumin induces pancreatic adenocarcinoma cell death via reduction of the inhibitors of apoptosis. Pancreas 2016, 45, 101–109. [Google Scholar] [CrossRef] [Green Version]
  95. Wang, L.; Zhang, J.; Wan, L.; Zhou, X.; Wang, Z.; Wei, W. Targeting Cdc20 as a novel cancer therapeutic strategy. Pharmacol. Ther. 2015, 151, 141–151. [Google Scholar] [CrossRef] [Green Version]
  96. Chang, D.Z.; Ma, Y.; Ji, B.; Liu, Y.; Hwu, P.; Abbruzzese, J.L. Increased Cdc20 expression is associated with pancreatic ductal adenocarcinoma differentiation and progression. J. Hematol. Oncol. 2012, 5, 15. [Google Scholar] [CrossRef] [Green Version]
  97. Zhang, Y.; Xue, Y.B.; Li, H.; Qiu, D.; Wang, Z.W.; Tan, S.S. Inhibition of Cell Survival by Curcumin Is Associated with Downregulation of Cell Division Cycle 20 (Cdc20) in Pancreatic Cancer Cells. Nutrients 2017, 9, 109. [Google Scholar] [CrossRef] [PubMed]
  98. Rugge, M.; Fassan, M.; Graham, D.Y. Epidemiology of gastric cancer. In Gastric Cancer; Springer: Cham, Switzerland, 2015; pp. 23–34. [Google Scholar]
  99. Yamashita, K.; Sakuramoto, S.; Watanabe, M. Genomic and epigenetic profiles of gastric cancer: Potential diagnostic and therapeutic applications. Surg. Today 2011, 41, 24–38. [Google Scholar] [CrossRef]
  100. Kasi, P.D.; Tamilselvam, R.; Skalicka-Woźniak, K.; Nabavi, S.F.; Daglia, M.; Bishayee, A.; Pazoki-Toroudi, H.; Nabavi, S.M. Molecular targets of curcumin for cancer therapy: An updated review. Tumor Biol. 2016, 37, 13017–13028. [Google Scholar] [CrossRef]
  101. Basnet, P.; Skalko-Basnet, N. Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 2011, 16, 4567–4598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Gong, H.; Cao, Y.; Han, G.; Zhang, Y.; You, Q.; Wang, Y.; Pan, Y. P53/microRNA-374b/AKT1 regulates colorectal cancer cell apoptosis in response to DNA damage. Int. J. Oncol. 2017, 50, 1785–1791. [Google Scholar] [CrossRef] [Green Version]
  103. Jin, H.; Lian, N.; Zhang, F.; Chen, L.; Chen, Q.; Lu, C.; Bian, M.; Shao, J.; Wu, L.; Zheng, S. Activation of PPARgamma/P53 signaling is required for curcumin to induce hepatic stellate cell senescence. Cell Death Dis. 2017, 7, e2189. [Google Scholar] [CrossRef]
  104. Fu, H.; Wang, C.; Yang, D.; Wei, Z.; Xu, J.; Hu, Z.; Zhang, Y.; Wang, W.; Yan, R.; Cai, Q. Curcumin regulates proliferation, autophagy, and apoptosis in gastric cancer cells by affecting PI3K and P53 signaling. J. Cell. Physiol. 2017, 233, 4634–4642. [Google Scholar] [CrossRef] [PubMed]
  105. Engelman, J.A. Targeting PI3K signalling in cancer: Opportunities, challenges and limitations. Nat. Rev. Cancer 2009, 9, 550–562. [Google Scholar] [CrossRef]
  106. Liu, W.H.; Yuan, J.B.; Zhang, F.; Chang, J.X. Curcumin inhibits proliferation, migration and invasion of gastric cancer cells via Wnt3a/β-catenin/EMT signaling pathway. Zhongguo Zhong Yao Za Zhi 2019, 44, 3107–3115. [Google Scholar]
  107. Zand, A.; Imani, S.; Saadati, M.; Borna, H.; Ziaei, R.; Honari, H. Effect of age, gender and blood group on blood cancer types. Trauma Mon. 2010, 15, 111–114. [Google Scholar]
  108. Koohi, F.; Salehiniya, H.; Shamlou, R.; Eslami, S.; Ghojogh, Z.M.; Kor, Y.; Rafiemanesh, H. Leukemia in Iran: Epidemiology and morphology trends. Asian Pac. J. Cancer Prev. 2015, 16, 7759–7763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Rafiq, S.; Raza, M.H.; Younas, M.; Naeem, F.; Adeeb, R.; Iqbal, J.; Anwar, P.; Sajid, U.; Manzoor, H.M. Molecular targets of curcumin and future therapeutic role in leukemia. J. Biosci. Med. 2018, 6, 33–50. [Google Scholar] [CrossRef] [Green Version]
  110. Jabbour, E.; Kantarjian, H. Chronic myeloid leukemia: 2016 update on diagnosis, therapy, and monitoring. Am. J. Hematol. 2016, 91, 252–265. [Google Scholar] [CrossRef] [Green Version]
  111. Wu, L.X.; Xu, J.H.; Wu, G.H.; Chen, Y.Z. Inhibitory effect of curcumin on proliferation of K562 cells involves down-regulation of p210 (bcr/abl) initiated Ras signal transduction pathway. Acta Pharmacol. Sin. 2003, 24, 1155–1160. [Google Scholar]
  112. Mukherjee, A.; Sarkar, R.; Mukherjee, S.; Biswas, J.; Roy, M. Curcumin boosts up the efficacy of imatinib mesylate in chronic myelogenic leukemia cell line K-562 by modulation of various markers. Int. J. Curr. Microbiol. Appl. Sci. 2016, 5, 240–255. [Google Scholar] [CrossRef] [Green Version]
  113. Gupta, S.C.; Sundaram, C.; Reuter, S. Aggarwal BB: Inhibiting NF-κB activation by small molecules as a therapeutic strategy. Biochim. Biophys. Acta 2010, 1799, 775–787. [Google Scholar] [CrossRef] [Green Version]
  114. Pesakhov, S.; Nachliely, M.; Barvish, Z.; Aqaqe, N.; Schwartzman, B.; Voronov, E.; Sharoni, Y.; Studzinski, G.P.; Fiahman, D.; Danilenko, M. Cancer-selective cytotoxic Ca2+ overload in acute myeloid leukemia cells and attenuation of disease progression in mice by synergistically acting polyphenols curcumin and carnosic acid. Oncotarget 2016, 7, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Rao, J.; Xu, D.R.; Zheng, F.M.; Long, Z.J.; Huang, S.S.; Wu, X.; Zhou, W.H.; Hunag, R.W.; Liu, Q. Curcumin reduces expression of Bcl-2, leading to apoptosis in daunorubicin-insensitive CD34+ acute myeloid leukemia cell lines and primary sorted CD34+ acute myeloid leukemia cells. J. Transl. Med. 2011, 9, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Tima, S.; Ichikawa, H.; Ampasavate, C.; Okonogi, S.; Anuchapreeda, S. Inhibitory effect of turmeric curcuminoids on FLT3 expression and cell cycle arrest in the FLT3-overexpressing EoL-1 leukemic cell line. J. Nat. Prod. 2014, 77, 948–954. [Google Scholar] [CrossRef]
  117. Zhu, G.H.; Dai, H.P.; Shen, Q.; Ji, O.; Zhang, Q.; Zhai, Y.L. Curcumin induces apoptosis and suppresses invasion through MAPK and MMP signaling in human monocytic leukemia SHI-1 cells. Pharm. Biol. 2016, 4, 1303–1311. [Google Scholar] [CrossRef]
  118. Hallek, M. Chronic lymphocytic leukemia: 2013 update on diagnosis, risk stratification and treatment. Am. J. Hematol. 2013, 88, 803–816. [Google Scholar] [CrossRef]
  119. Gonzalez-Rodriguez, A.P.; Contesti, J.; Huergo-Zapico, L.; Lopez-Soto, A.; Fernández-Guizán, A.; Acebes-Huerta, A.; Gonzalez-Huerta, A.J.; Gonzalez, E.; Fernandez-Alvarez, C.; Gonzalez, S. Prognostic significance of CD8 and CD4 T cells in chronic lymphocytic leukemia. Leuk Lymphoma 2010, 51, 1829–1836. [Google Scholar] [CrossRef] [PubMed]
  120. Ghosh, A.K.; Kay, N.E.; Secreto, C.R.; Shanafelt, T.D. Curcumin inhibits prosurvival pathways in chronic lymphocytic leukemia B cells and may overcome their stromal protection in combination with EGCG. Clin. Cancer Res. 2009, 15, 1250–1258. [Google Scholar] [CrossRef] [Green Version]
  121. Chaitanya, G.; Alexander, J.S.; Babu, P. PARP-1 cleavage fragments: Signatures of cell-death proteases in neurodegeneration. Cell Commun. Signal. 2010, 8, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Mishra, D.; Singh, S.; Narayan, G. Curcumin induces apoptosis in Pre-B acute lymphoblastic leukemia cell lines Via PARP-1 cleavage. Asian Pac. J. Cancer Prev. 2016, 17, 3865–3869. [Google Scholar]
  123. Guo, Y.; Li, Y.; Shan, Q.; He, G.; Lin, J.; Gong, Y. Curcumin potentiates the anti-leukemia effects of imatinib by downregulation of the AKT/mTOR pathway and BCR/ABL gene expression in Ph+ acute lymphoblastic leukemia. Int. J. Biochem. Cell Biol. 2016, 65, 1–11. [Google Scholar] [CrossRef] [PubMed]
  124. Dhillon, N.; Aggarwal, B.B.; Newman, R.A.; Wolff, R.A.; Kunnumakkara, A.B.; Abbruzzese, J.L.; Ng, C.S.; Badmaev, V.; Kurzrock, R. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin. Cancer Res. 2008, 14, 4491–4499. [Google Scholar] [CrossRef] [Green Version]
  125. Bayet-Robert, M.; Kwiatkowski, F.; Leheurteur, M.; Gachon, F.; Planchat, E.; Abrial, C.; Mouret-Reynier, M.A.; Durando, X.; Barthomeuf, C.; Chollet, P. Phase I dose escalation trial of docetaxel plus curcumin in patients with advanced and metastatic breast cancer. Cancer Biol. Ther. 2010, 9, 8–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Vadhan-Raj, S.V.; Weber, D.M.; Wang, M.; Giralt, S.A.; Thomas, S.K.; Alexanian, R.; Zhou, X.; Patel, P.; Bueso-Ramos, C.E.; Newman, R.; et al. Curcumin downregulates NF-kB and related genes in patients with multiple myeloma: Results of a phase I/II study. Blood 2007, 110, 1177. [Google Scholar] [CrossRef]
  127. Kalluri, R.; Michael, Z. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef]
  128. Marsh, T.; Pietras, K.; McAllister, S.S. Fibroblasts as architects ofcancer pathogenesis. Biochim. Biophys. Acta 2013, 1832, 1070–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zhang, S.S.; Gond, Z.J.; Li, W.H.; Wang, X.; Ling, T.Y. Antifibrotic effect of curcumin in TGF-Β1-induced myofibroblastsfrom human Oral mucosa. Asian Pac. J. Cancer Prev. 2012, 13, 289–294. [Google Scholar] [CrossRef] [PubMed]
  130. Avasarala, S.; Zhang, F.; Liu, G.; Wang, R.; London, S.D.; London, L. Curcumin modulates the inflammatory response and inhibitssubsequent fibrosis in a mouse model of viral-induced acute respira-tory distress syndrome. PLoS ONE 2013, 8, e57285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Hendrayani, S.F.; Al-Khalaf, H.H.; Aboussekhra, A. Curcumintriggers P16-dependent senescence in active breast cancer-associatedfibroblasts and suppresses their paracrine procarcinogenic effects. Neoplasia 2013, 15, 631–640. [Google Scholar] [CrossRef] [Green Version]
  132. Zeng, Y.; Du, Q.; Zhang, Z.; Ma, J.; Han, L.; Wang, Y.; Yang, L.; Tao, N.; Qin, Z. Curcumin promotes cancer-associated fibroblasts apoptosis via ROS-mediated endoplasmic reticulum stress. Arch. Biochem. Biophys. 2020, 694, 108613. [Google Scholar] [CrossRef] [PubMed]
  133. Ba, P.; Xu, M.; Yu, M.; Li, L.; Duan, X.; Lv, S.; Fu, G.; Yang, J.; Yang, P.; Yang, C.; et al. Curcumin suppresses the proliferation and tumorigenicity of Cal27 by modulating cancer-associated fibroblasts of TSCC. Oral. Dis. 2020, 26, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  134. U.S. Food and Drug Administration. Agency Response Letter GRAS Notice No. GRN 00460; CFSAN/Office of Food Additive Safety, 1011 U.S.; Food and Drug Administration: Silver Spring, MD, USA, 2013.
  135. Lamb, S.R.; Wilkinson, S.M. Contact allergy to tetrahydrocurcumin. Contact Dermat. 2003, 48, 227. [Google Scholar] [CrossRef] [PubMed]
  136. Lao, C.D.; Ruffin, M.T.; Normolle, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E. Dose escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 2006, 6, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Panahi, Y.; Saadat, A.; Beiraghdar, F.; Nouzari, S.M.H.; Jalalian, H.R.; Sahebkar, A. Antioxidant effects of bioavailability-enhanced curcuminoids in patients with solid tumors: A randomized double-blind placebo-controlled trial. J. Funct. Foods 2014, 6, 615–622. [Google Scholar] [CrossRef]
  138. Ryan, J.L.; Heckler, C.E.; Ling, M.; Katz, A.; Williams, J.P.; Pentland, A.P.; Morrow, G.R. Curcumin for radiation dermatitis: A randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer patients. Radiat. Res. 2013, 180, 34–43. [Google Scholar] [CrossRef] [Green Version]
  139. Hejazi, J.; Rastmanesh, R.; Taleban, F.A.; Molana, S.H.; Ehtejab, G. A pilot clinical trial of radioprotective effects of curcumin supplementation in patients with prostate cancer. J. Cancer Sci. Ther. 2013, 5, 320–324. [Google Scholar]
  140. Borsari, M.; Ferrari, E.; Grandi, R.; Saladini, M. Curcuminoids as potential new iron-chelating agents: Spectroscopic, polarographic and potentiometric study on their Fe(III) complexing ability. Inorg. Chim. Acta 2002, 328, 61–68. [Google Scholar] [CrossRef]
  141. Jiao, Y.; Wilkinson, J.T.; Di, X.; Wang, W.; Hatcher, H.; Kock, N.D. Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood 2009, 113, 462–469. [Google Scholar] [CrossRef] [Green Version]
  142. Kim, D.C.; Ku, S.K.; Bae, J.S. Anticoagulant activities of curcumin and its derivative. BMB Rep. 2012, 45, 221–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wang, Z.; Sun, W.; Huang, C.K.; Wang, L.; Xia, M.M.; Cui, X.; Hu, G.X.; Wang, Z.S. Inhibitory effects of curcumin on activity of cytochrome P450 2C9 enzyme in human and 2C11 in rat liver microsomes. Drug Dev. Ind. Pharm. 2015, 41, 613–616. [Google Scholar] [CrossRef] [PubMed]
  144. Volak, L.P.; Ghirmai, S.; Cashman, J.R.; Court, M.H. Curcuminoids inhibit multiple human cytochromes P450 (CYP), UDP-glucuronosyltransferase (UGT), and sulfotransferase (SULT) enzymes, while piperine is a relatively selective CYP3A4 inhibitor. Drug Metab. Dispos. 2008, 36, 1594–1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, A.C.; Zhao, L.X.; Lou, H.X. Curcumin alters the pharmacokinetics of warfarin and clopidogrel in Wistar rats but has no effect on anticoagulation or antiplatelet aggregation. Planta Med. 2013, 79, 971–977. [Google Scholar] [CrossRef] [PubMed]
  146. Pavithra, B.H.; Prakash, N.; Jayakumar, K. Modification of pharmacokinetics of norfloxacin following oral administration of curcumin in rabbits. J. Vet. Sci. 2009, 10, 293–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Choudhari, A.S.; Mandave, P.C.; Deshpande, M.; Ranjekar, P.; Prakash, O. Phytochemicals in cancer treatment: From preclinical studies to clinical practice. Front. Pharmacol. 2020, 10, 1614. [Google Scholar] [CrossRef] [Green Version]
  148. Tan, B.L.; Norhaizan, M.E. Curcumin combination chemotherapy: The implication and efficacy in cancer. Molecules 2019, 24, 2527. [Google Scholar] [CrossRef] [Green Version]
  149. Kumar, P.; Sulakhiya, K.; Barua, C.C.; Mundhe, N. TNF-α, IL-6 and IL-10 expressions, responsible for disparity in action of curcumin against cisplatin-induced nephrotoxicity in rats. Mol. Cell. Biochem. 2017, 431, 113–122. [Google Scholar] [CrossRef]
  150. Belcaro, G.; Hosoi, M.; Pellegrini, L.; Appendino, G.; Ippolito, E.; Ricci, A.; Ledda, A.; Dugall, M.; Cesarone, M.R.; Maione, C.; et al. A controlled study of a lecithinized delivery system of curcumin (Meriva(R)) to alleviate the adverse effects of cancer treatment. Phytother. Res. 2014, 28, 444–450. [Google Scholar] [CrossRef]
  151. Heger, M.; van Golen, R.F.; Broekgaarden, M.; Michel, M.C. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol. Rev. 2014, 66, 222–307. [Google Scholar] [CrossRef]
  152. Vareed, S.K.; Kakarala, M.; Ruffin, M.T.; Crowell, J.A.; Normolle, D.P.; Djuric, Z.; Brenner, D.E. Pharmacokinetics of curcumin conjugate metabolites in healthy human subjects. Cancer Epidemiol. Prev. Biomark. 2008, 17, 1411–1417. [Google Scholar] [CrossRef] [Green Version]
  153. Kanai, M.; Yoshimura, K.; Asada, M.; Imaizumi, A.; Suzuki, C.; Matsumoto, S.; Nishimura, T.; Mori, Y.; Masui, T.; Kawaguchi, Y.; et al. A phase I/II study of gemcitabine-based chemotherapy plus curcumin for patients with gemcitabine-resistant pancreatic cancer. Cancer Chemother. Pharmacol. 2011, 68, 157–164. [Google Scholar] [CrossRef] [Green Version]
  154. Sharma, R.A.; Euden, S.A.; Platton, S.L.; Cooke, D.N.; Shafayat, A.; Hewitt, H.R.; Marczylo, T.H.; Morgan, B.; Hemingway, D.; Plummer, S.M.; et al. Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance. Clin. Cancer Res. 2004, 10, 6847–6854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Schiborr, C.; Eckert, G.P.; Rimbach, G.; Frank, J. A validated method for the quantification of curcumin in plasma and brain tissue by fast narrow-bore high-performance liquid chromatography with fluorescence detection. Anal. Bioanal. Chem. 2010, 397, 1917–1925. [Google Scholar] [CrossRef]
  156. Adiwidjaja, J.; McLachlan, A.J.; Boddy, A.V. Curcumin as a clinically-promising anti-cancer agent: Pharmacokinetics and drug interactions. Expert Opin. Drug Metab. Toxicol. 2017, 13, 953–972. [Google Scholar] [CrossRef]
  157. Jäger, R.; Lowery, R.P.; Calvanese, A.V.; Joy, J.M.; Purpura, M.; Wilson, J.M. Comparative absorption of curcumin formulations. Nutr. J. 2014, 13, 11. [Google Scholar] [CrossRef] [Green Version]
  158. Gota, V.S.; Maru, G.B.; Soni, T.G.; Gandhi, T.R.; Kochar, N.; Agarwal, M.G. Safety and pharmacokinetics of a solid lipid curcumin particle formulation in osteosarcoma patients and healthy volunteers. J. Agric. Food Chem. 2010, 58, 2095–2099. [Google Scholar] [CrossRef] [PubMed]
  159. Schiborr, C.; Kocher, A.; Behnam, D.; Jandasek, J.; Toelstede, S.; Frank, J. The oral bioavailability of curcumin from micronized powder and liquid micelles is significantly increased in healthy humans and differs between sexes. Mol. Nutr. Food Res. 2014, 58, 516–527. [Google Scholar] [CrossRef] [PubMed]
  160. Wei, T.; Lu, M.; Meng, Z.; Chen, X.; Jing, J.; Li, J.; Yao, W.; Zhu, H.; Fu, T. Treatment of metastatic lung cancer via inhalation administration of curcumin composite particles based on mesoporous silica. Eur. J. Pharm. Sci. 2019, 134, 246–255. [Google Scholar]
  161. Barik, A.; Mishra, B.; Shen, L.; Mohan, H.; Kadam, R.M.; Dutta, S.; Zhang, H.Y.; Priyadarsini, K.I. Evaluation of a new copper(II)-curcumin complex as superoxide dismutase mimic and its free radical reactions. Free Radic. Biol. Med. 2005, 6, 811–822. [Google Scholar] [CrossRef]
  162. Zebib, B.; Mouloungui, Z.; Noirot, V. Stabilization of Curcumin by Complexation with Divalent Cations in Glycerol/Water System. Bioinorg. Chem. Appl. 2010, 2010, 292760. [Google Scholar] [CrossRef] [PubMed]
  163. Meng, R.; Wu, Z.; Xie, Q.T.; Cheng, J.S.; Zhang, B. Preparation and characterization of zein/carboxymethyl dextrin nanoparticles to encapsulate curcumin: Physicochemical stability, antioxidant activity and controlled release properties. Food Chem. 2021, 340, 127893. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of (A) curcumin, (B) bis-demethoxycurcumin, (C) demethoxycurcumin. Structures were drawn using ChemSpider, an online free chemical structure database.
Figure 1. Chemical structure of (A) curcumin, (B) bis-demethoxycurcumin, (C) demethoxycurcumin. Structures were drawn using ChemSpider, an online free chemical structure database.
Biomedicines 09 01086 g001
Figure 2. Summarized effects of curcumin on cancer cells. Key: CDK2, cyclin-dependent kinase 2; MMP-2, matrix metallopeptidase 2; MMP-9, matrix metallopeptidase 9; VEGF, vascular endothelial growth factor; NF-kb, nuclear factor kappa-light-chain-enhancer of activated B cells; mTOR, mechanistic target of rapamycin; PTEN, phosphatase and tensin homolog; bcl-2, B-cell lymphoma-2; bax, Bcl-2-associated X protein.
Figure 2. Summarized effects of curcumin on cancer cells. Key: CDK2, cyclin-dependent kinase 2; MMP-2, matrix metallopeptidase 2; MMP-9, matrix metallopeptidase 9; VEGF, vascular endothelial growth factor; NF-kb, nuclear factor kappa-light-chain-enhancer of activated B cells; mTOR, mechanistic target of rapamycin; PTEN, phosphatase and tensin homolog; bcl-2, B-cell lymphoma-2; bax, Bcl-2-associated X protein.
Biomedicines 09 01086 g002
Table 1. Effects of curcumin on cell signaling pathways in different types of cancer.
Table 1. Effects of curcumin on cell signaling pathways in different types of cancer.
Cancer TypeCell Signaling
Pathways
Type of EffectUsed ModelTested DosageReferences
Lung cancerWnt/β-cateninDownregulation/inhibitionHuman cell line A54960 μM[35]
VEGFDownregulation/inhibitionNude mice100 mg/kg[36]
NF-κBDownregulation/inhibitionNude mice100 mg/kg[36]
NOTCH 1Downregulation/inhibitionHuman lung cancer cell lines6 μΜ[37]
ERK ½Downregulation/inhibitionHuman NCI-H1975 line10 ng/mL[41]
Breast cancerAkt/mTORDownregulation/inhibitionHuman breast cell lines10 or 30 μΜ[55]
NF-κBDownregulation/inhibitionHuman breast cell lines20 or 25 μΜ[56]
Autocrine GHDownregulation/inhibitionT47D human breast cells20 μΜ[58]
Bcl-2 and Bcl- xLDownregulation/inhibitionT47D human breast cells20 μΜ[58]
MDR-1Downregulation/inhibitionMCF-7 breast cancer cell line1.3 μΜ[60]
FEN1Downregulation/inhibitionMCF-7 breast cancer cell line0–50 μmol/L[59]
Brain cancerSTAT3Downregulation/inhibitionHuman GBM stem cells25 μΜ[82]
IAPDownregulation/inhibitionHuman GBM stem cells25 μΜ[82]
MAPKUpregulation/activationHuman GBM stem cells25 μΜ[82]
Pancreatic cancerPlatelet-derived growth factorDownregulation/inhibitionRat pancreatic stellate cells25 μΜ[90]
PI3 K/AktDownregulation/inhibitionPanc-1 human pancreatic cells20 μΜ[93]
IAPDownregulation/inhibitionPANC-1 human cells10/50/100μΜ[94]
Cdc20Downregulation/inhibitionPatu8988 and Panc-1
human cell lines
10 or 20 μΜ[97]
Gastric cancerPI3KDownregulation/inhibitionHuman SGC-7901 and BGC-823 cells10/20/40 μΜ[104]
Wnt3 a/β-catenin/EMTDownregulation/inhibitionHuman gastric cell lines20 μΜ[106]
BCL-2Downregulation/inhibitionHuman gastric cell lines20 μΜ[106]
Leukemia
-CMLMAPKDownregulation/inhibitionHuman K562 cell line5 or 10 mg/L[111]
Hsp90Downregulation/inhibitionHuman K562 cell line30 μΜ[112]
p210 BCR-ABLDownregulation/inhibitionHuman K562 cell line5 or 10 mg/L[111]
AMLBcl-2Downregulation/inhibitionPrimary human CD34+ AML cells0–80 μΜ[116]
MAPKDownregulation/inhibitionHuman SHI-1 cells6.25–25 μM[117]
MMPDownregulation/inhibitionHuman SHI-1 cells6.25–25 μM[117]
CLLNF-κBDownregulation/inhibitionHuman CLL B cells10–12.5 μΜ[120]
STAT3Downregulation/inhibitionHuman CLL B cells10–12.5 μΜ[120]
AKTDownregulation/inhibitionHuman CLL B cells10–12.5 μΜ[120]
XIAPDownregulation/inhibitionHuman CLL B cells10–12.5 μΜ[120]
Mcl-1Downregulation/inhibitionHuman CLL B cells10–12.5 μΜ[120]
ALLAKT/mTORDownregulation/inhibitionHuman ALL cell lines0–40 μΜ[123]
ABL/STAT5Downregulation/inhibitionHuman ALL cell lines0–40 μΜ[123]
BCR/ABLDownregulation/inhibitionHuman ALL cell lines0–40 μΜ[123]
Table 2. The most recent clinical trials of curcumin in different cancers.
Table 2. The most recent clinical trials of curcumin in different cancers.
CancerDrugTitleNCTPhaseEstimated/Actual Completion Date
BreastCurcuminA „Window Trial” on Curcumin for Invasive Breast Cancer Primary TumorsNCT03980509130 December 2022
Curcumin ® (CUC-01) and Paclitaxel‘’Curcumin” in Combination with Chemotherapy in Advanced Breast CancerNCT03072992230 June 2019
ProstateCurcuminTrial of Curcumin to Prevent Progression of Low-risk Prostate Cancer Under Active SurveillanceNCT037697663November 2026
Curcumin and radiationNanocurcumin for Prostate Cancer Patients Undergoing Radiotherapy (RT)NCT027246182April 2022
ColorectalAvastin/FOLFIRI and curcuminAvastin/FOLFIRI in Combination with Curcumin in Colorectal Cancer Patients with Unresectable MetastasisNCT0243938521 August 2019
CervicalCurcuminCurcumin in Advanced Cervical CancerNCT04294836231 December 2023
Chronic Lymphocytic Leukemia (CLL),
Small Lymphocytic Lymphoma (SLL)
Curcumin and cholecalciferolCurcumin and Cholecalciferol in Treating Patients With Previously Untreated Stage 0–II Chronic Lymphocytic Leukemia or Small Lymphocytic LymphomaNCT02100423213 December 2018
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zoi, V.; Galani, V.; Lianos, G.D.; Voulgaris, S.; Kyritsis, A.P.; Alexiou, G.A. The Role of Curcumin in Cancer Treatment. Biomedicines 2021, 9, 1086. https://doi.org/10.3390/biomedicines9091086

AMA Style

Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The Role of Curcumin in Cancer Treatment. Biomedicines. 2021; 9(9):1086. https://doi.org/10.3390/biomedicines9091086

Chicago/Turabian Style

Zoi, Vasiliki, Vasiliki Galani, Georgios D. Lianos, Spyridon Voulgaris, Athanasios P. Kyritsis, and George A. Alexiou. 2021. "The Role of Curcumin in Cancer Treatment" Biomedicines 9, no. 9: 1086. https://doi.org/10.3390/biomedicines9091086

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop