Next Article in Journal
Ribose and Non-Ribose A2A Adenosine Receptor Agonists: Do They Share the Same Receptor Recognition Mechanism?
Next Article in Special Issue
Investigating the Role of Circulating miRNAs as Biomarkers in Colorectal Cancer: An Epidemiological Systematic Review
Previous Article in Journal
IGF-1 as a Potential Therapy for Spinocerebellar Ataxia Type 3
Previous Article in Special Issue
Animal Models in Bladder Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Parthenolide and Its Soluble Analogues: Multitasking Compounds with Antitumor Properties

1
Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Biochemistry Building, University of Palermo, 90127 Palermo, Italy
2
Laboratory of Biochemistry, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90127 Palermo, Italy
*
Authors to whom correspondence should be addressed.
Biomedicines 2022, 10(2), 514; https://doi.org/10.3390/biomedicines10020514
Submission received: 26 January 2022 / Revised: 16 February 2022 / Accepted: 19 February 2022 / Published: 21 February 2022

Abstract

:
Due to its chemical properties and multiple molecular effects on different tumor cell types, the sesquiterpene lactone parthenolide (PN) can be considered an effective drug with significant potential in cancer therapy. PN has been shown to induce either classic apoptosis or alternative caspase-independent forms of cell death in many tumor models. The therapeutical potential of PN has been increased by chemical design and synthesis of more soluble analogues including dimethylaminoparthenolide (DMAPT). This review focuses on the molecular mechanisms of both PN and analogues action in tumor models, highlighting their effects on gene expression, signal transduction and execution of different types of cell death. Recent findings indicate that these compounds not only inhibit prosurvival transcriptional factors such as NF-κB and STATs but can also determine the activation of specific death pathways, increasing intracellular reactive oxygen species (ROS) production and modifications of Bcl-2 family members. An intriguing property of these compounds is its specific targeting of cancer stem cells. The unusual actions of PN and its analogues make these agents good candidates for molecular targeted cancer therapy.

1. Introduction

Parthenolide (PN) is a sesquiterpene lactone extracted from the medical herb feverfew (Tanacetum parthenium) that has been shown to exert both anti-inflammatory [1,2,3] and antitumor effects [4,5,6]. The biological activity of PN is correlated with its chemical structure, which includes an α-methylene-γ-lactone ring and an epoxide (Figure 1A). These components can interact with nucleophilic sites of specific cellular proteins [7]. In particular, the unsaturated double bonds conjugated with a carbonyl group have been shown to react with free protein thiol groups, causing modulation of the redox state in the cells and induction of cell death [8].
The anti-inflammatory action of PN has been widely considered a consequence of its inhibitory effect on the transcription factors belonging to NF-κB family [9,10,11]. These factors are responsible for the regulation of several diverse cellular events, including cell proliferation, differentiation and survival, and their role in inflammation is well established. NF-κB, upon specific stimuli, can promote the expression of proinflammatory mediators including cytokines such as TNF-α, IL-1 and IL-6 [12], adhesion molecules (e.g., ICAM, E-selectin) and enzymes including the inducible forms of cyclooxygenase (COX-2) and nitric oxide synthase (iNOS). Therefore, they behave as a central regulator to innate immunity and inflammatory processes [13,14].
Clinical and experimental data indicate that the NF-κB signaling pathway is the cause of cachexia that develops in cancer patients [15]. Indeed, patients often develop a wasting syndrome characterized by systemic inflammation and involuntary loss of body mass that cannot be reversed by normal nutritional support [15]. NF-κB inhibitors could also be used as anticancer therapy to treat cachexia [16,17].
As well as NF-κB inhibition, PN has been shown to directly inhibit platelet phospholipase A2, 5-lipoxygenase and inducible cyclo-oxigenase [18,19], thus contributing to inflammation prevention. In addition, evidence has been provided that PN can also inhibit the release of the proinflammatory mediators nitric oxide, PGE(2) and TNF-α from macrophages and TNF-α, IL-2, IFN-γ and IL-4 from human peripheral blood mononuclear cells [1].
According to its antiphlogistic action, PN, obtained by leaves or infusions of feverfew, has been widely used in folk medical practices for treatment of fever, arthritis and migraine [20,21].
In addition to its anti-inflammatory properties, PN has been shown to exert potent antitumor properties. Several recent papers highlight the role of PN as an antitumor agent and describe its ability to induce programmed cell death in a wide variety of tumor cell lines [22,23,24].
To make PN more usable in therapy, a more soluble derivative has been synthesized: dimethylaminoparthenolide (DMAPT) (Figure 1B), also known as LC-1 [25]. When formulated as a fumarate salt, DMAPT displays a solubility in water more than 1000 times greater than PN [26,27].
Using the main data present in the literature to date, this comprehensive review critically summarizes the biological effects of PN and its analogues in tumor cells, providing an up-to-date overview of their action mechanisms and selective antitumor potential that make these naturally derived compounds promising agents for cancer therapy.

2. The Effect of Parthenolide on Gene Expression Profile

Both the anti-inflammatory and antitumor effects of PN have been correlated with its ability to influence the expression of specific genes through modulation of some transcription factors.

2.1. Parthenolide and Nuclear Factor Kappa B (NF-κB)

The factors belonging to the NF-κB family play an important role in the transcriptional regulation of genes involved in cell survival and immune response [14]. The most well characterized NF-κB components are the heterodimers constituted by p65 and p50 subunits. Activation of this complex, as well as some other NF-κB dimeric forms, occurs as a prosurvival response following different proapoptotic stimuli including cytotoxic drugs [28,29], ionizing radiation [30,31], TNF-α [32,33], oxidative stress [34,35].
In resting cells, the NF-κB factors are sequestered in the cytoplasm by binding to their regulatory effectors’ IkB proteins. Upon specific stimulation, IkB proteins become phosphorylated on their N-terminal serine residues. This modification targets IkB through ubiquitination and consequent degradation by the 26S proteasome. NF-κB is thus released and can translocate to the nucleus where it promotes gene transcription [36].
The phosphorylation of IkB inhibitory proteins is controlled by specific kinases known as IkB kinases (IKK). In particular, the catalytic subunits IKKα and IKKβ together with the regulatory subunit NEMO, associate to form the active IKK complex (IKC) which can then phosphorylate IkB [37,38].
The inhibitory action of PN on NF-κB has been known for many years. Several lines of evidence indicate that PN can exert its inhibitory action by targeting IKC components [5,39]. A direct in vitro binding of PN to IKKβ has been demonstrated through the alpha-methylene gamma-lactone moiety present in the molecule [40]. Moreover, Idris et al. have shown that PN prevents the activation of both IKKα and IKKβ and suppresses growth and migration of mammary carcinosarcoma cells [41]. Inhibition of IKK results in the accumulation of IkB inhibitory proteins with consequent sequestration of NF-κB in the cytoplasm.
In addition, it has been shown that PN can directly affect the translocation of the p65 NF-κB subunit to the nucleus [42,43].
It is well known that NF-κB factors can promote the transcriptional activation of prosurvival/antiapoptotic genes including Bcl-XL, survivin and c-FLIP [44,45]. Inhibition of NF-κB by PN thus results in a reduction in the level of these gene products with consequent apoptotic effects. Many authors have recently shown that the antitumor/proapoptotic effects of PN in tumor cells are, at least in part, due to NF-κB inhibition. Considering that NF-κB factors are involved in malignant transformation and tumor progression [46,47], the use of PN as NF-κB inhibitor can be particularly adequate for those tumor forms where increased activity or constitutive activation of NF-κB have been characterized.
Interestingly, NF-κB inhibition by PN has been shown to sensitize tumor cells to the effects of chemotherapeutic drugs [22,48,49] or radiation [50,51,52,53].

2.2. Parthenolide and STAT Transcription Factors

Other transcription factors which are targeted by PN belong to the signal transducer and activator of transcription (STAT) family. These proteins modulate a wide variety of biological processes including cell proliferation, differentiation and apoptosis as well as inflammation and immunity [54,55]. Activation of these proteins occurs following tyrosine-phosphorylation by members of the Janus Kinase (JAK) family, which are cytokine receptor associated kinases. For a complete description of STAT family members and JAK signal transduction, review by Awasthi et al. and Hu et al. provide updated insights [56,57].
Phosphorylated STATs can then dimerize and translocate to the nucleus, where they exert their transcriptional activity. STATs transcriptional targets include several antiapoptotic and prosurvival factors [58,59], thus indicating their oncogenic potential. In particular, among STAT family members, STAT-3 seems to play a key role in tumor transformation and malignancy [60,61]. Inhibition of STAT-3 in tumor cells leads to down-regulation of its transcriptional targets belonging to the Bcl-2 family including the antiapoptotic Bcl-2, Bcl-XL and Mcl-1 [62]. Similar effects have been found in cutaneous T-cell lymphoma cells after treatment with a STAT-3 inhibitor [63]. Given the role of STAT proteins in contributing to cell survival, oncogenic activation of these factors, especially STAT-3, has been found in diverse types of tumors including hepatocellular carcinoma, colorectal cancer, breast cancer, bladder cancer and hematological malignancies [55,64].
Our previous studies performed in human hepatocellular carcinoma (HCC) cell lines, provided evidence that PN is able to sensitize TRAIL-induced apoptosis by reducing phosphorylated STAT-3 levels. This event appears to be related to the inhibitory effect exerted by PN on the activation of JAK proteins. Sensitization by PN to TRAIL stimulated the extrinsic mechanism of apoptosis in HCC cell lines with the activation of both caspases 8 and 3 [65].
PN has been shown to behave as an inhibitor of the JAK/STAT signaling pathway in response to cytokines of the IL-6 family [66]. Evidence has been provided that PN affects multiple steps of this pathway. For instance, the compound can either inhibit JAKs through binding to their SH groups, thus avoiding STAT phosphorylation [67] or directly block the DNA binding of STAT proteins in activated hepatic stellate cells [66].
In addition, recent evidence has been provided that PN can regulate crosstalk of toll-like receptor 4 (TLR4) with STAT-3, thus reducing the release of inflammatory cytokines inducing apoptosis in activated hepatic stellate cells [68].
It is interesting to note that STAT-3 inhibition by PN has also been correlated not only with apoptosis induction in tumor cells but also with reversed drug-resistance [69].

2.3. Parthenolide Interplay with Other Transcription Factors and Epigenetic Effects

Notably, the antitumor effect of PN has been correlated with reduced gene transcription of prosurvival mediators [70].
In this context, AP-1 is another important transcription factor which is mainly involved in cell survival and proliferation [71,72]. Interestingly, PN has been shown to inhibit AP-1 DNA binding and transcriptional activity induced by UVB in a skin cancer model, thus exerting a chemopreventive activity [73].
Microphthalmia-associated transcription factor (MITF) is strongly associated with regulation of proliferation, survival and senescence of melanoma cells [74,75]. Hartman et al. have recently provided evidence that PN efficiently decreases the MITF M isoform level in melanoma cells as a result of transcriptional regulation that results in melanoma cell senescence [73].
The Wnt/β-catenin pathway is well known to be involved in tumor cell proliferation [76,77]. PN was recently shown to inhibit Wnt signaling by decreasing the levels of the transcription factors TCF4/LEF1 without affecting β-catenin stability or subcellular localization [78]. However, other authors also showed that PN is capable of reducing the levels of β-catenin in myelomonocytic leukemic U937 cells [70].
These findings might represent a nice tool to inhibit cell proliferation of those tumor types bearing β-catenin oncogenic mutations or Wnt signaling hyperactivation.
In addition to inhibition of transcription factors with prosurvival/oncogenic action, PN has been also shown to exert a function in promoting apoptosis through the activation of specific death genes. One example is given by the effect of PN on the tumor suppressor/proapoptotic transcription factor p53. It is well known that p53 is key regulator in a molecular network establishing the cell fate due to its ability to promote cell cycle arrest and/or apoptosis [79,80,81]. The p53 pathway is often abrogated in cancer and sometimes this might depend on increased activity of its negative regulator MDM2 [82].
In this regard, Gopal et al. have reported that PN can stimulate the ubiquitination of MDM2 thereby activating p53 cellular functions [83]. More recently, activation or stabilization of p53 by PN in tumor cells has also been found by other authors [17,84,85].
However, other reports indicate that PN can also act independently of p53 [86] and promote epigenetic modifications. For instance, epigenetic modulation of the CDK inhibitor and tumor suppressor p21 by PN has been evidenced in cancer cells [87].
Through an integrated molecular profiling approach, a description of transcription factors modulated by PN was provided. PN was able to alter the binding of important transcription factors in prostate cancer including C/EBP-alpha, fos-related antigen-1 (FRA-1), HOXA-4, c-MYB, SNAIL, SP1, serum response factor (SRF), STAT3, X-box binding protein-1 (XBP1), and p53 [88]. PN interplay with some transcription factors and relative signaling is described in Figure 2.
Beyond direct modulation of transcription factors, PN has been shown to modulate gene expression through the controls of epigenetic events. Evidence has been provided, for instance, that PN is able to deplete histone deacetylase 1 protein (HDAC1) and to induce cell death through ataxia telangiectasia mutated (ATM) [89]. Liu et al. have shown that PN favors DNA hypomethylation through a dual effect on DNA methyltransferase 1 (DNMT1). These authors reported that the compound can either directly inhibit DNMT1 or repress its expression by targeting transcriptional factor Sp1 binding to the DNMT1 promoter [90]. Up-to-date and wide descriptions of epigenetic effects of PN are provided by the papers of Irshad et al. [91] and Freund et al. [2].

3. Parthenolide Can Specifically Modulate Signal Transduction Pathways

A number of papers have highlighted the ability of PN to modulate signal transduction components, thereby influencing cell response to various stimuli.
The role of PN in counteracting cytokine-mediated signaling through the inhibition of the JAK–STAT pathway has previously been noted [67]. Moreover, PN has been shown to inhibit IL-1- and TNFα-induced NF-κB activation [66]. Figure 3 summarizes the effects of PN on different signaling cascades.

3.1. Modulation of PKC and MAPKs by Parthenolide

In addition to its effect in counteracting proinflammatory cytokine-mediated signaling, PN has been shown to exert an influence on cascades involved in cell proliferation and differentiation.
Specifically, modulation of protein kinase C (PKC) and MAP kinase components (MAPKs) by PN has been described. For instance, papers published some time ago show the effects of PN on PKC. In particular, the activation of specific PKC isoforms by PN has been shown to increase the effects of ultraviolet B (UVB) on skin cancer [92]. Moreover, evidence has indicated that PN leads to activation of the PKC and MAPK pathways, thus enhancing the differentiation of leukemia cells induced by all trans retinoic acid [93].
Considering the complex and diverse role of PKC isoforms and MAPKs in the control of cell proliferation and apoptosis, it is not surprising that contradictory data can be found regarding the effect of PN on these mediators. In contrast to the findings of the previous authors, through a protein microarray analysis of PN action in prostate cancer, Kawasaki et al. have correlated the antiproliferative effect of the compound with its ability to downregulate prosurvival kinase effectors, including PKC isoforms, MAPKs, PI3 kinase (PI3-K) and CaM kinases [94].
More recently, in line with these findings, PN has been shown to potently inhibit the B-Raf/MAPK/Erk mitogenic pathway in tumor cells [95] and to target epidermal growth factor receptor and signaling either in vitro or in vivo in lung cancer models [96]. Evidence has also indicated that PN inhibits oncogenic focal adhesion kinase in breast cancer cells [97].
Moreover, in accordance with previous considerations regarding Wnt/beta-catenin signaling, direct interference by PN with this oncogenic pathway was evidenced in colorectal cancer [98].
Overall, it seems that targeting prosurvival/oncogenic signaling is among the modalities that account for the antitumor action of PN.

3.2. Effects of Parthenolide on Stress Kinases

The c-Jun N-terminal kinase (JNK) and the p38 kinase are members of the MAPK family that are involved in the response to cell stress and induction of apoptosis [99,100].
Many studies support a role of PN and its analogues in the activation of these kinases, an event that is often correlated with the induction of oxidative stress and the consequent execution of cell death. It has been demonstrated that the PN analogue DMAPT suppresses in vivo prostate cancer growth and induces apoptosis through reactive oxygen species (ROS) generation with subsequent JNK activation [27,43,101,102]. The same authors showed that interfering with JNK by shRNA reduces the antiproliferative and apoptotic effects of DMAPT [27]. More recently, in line with these findings, a novel PN analogue (CPPTL) displayed antineoplastic effects via the ROS/JNK pathway in acute myeloid leukemia [101]. However, other authors have previously shown that activation of JNK by PN can also occur independently of ROS generation and appears to be crucial to overcome resistance of breast cancer cells to TRAIL-induced apoptosis [102]. Similarly, Zhang et al. have demonstrated that PN can sensitize various human cancer cells to TNF-α-induced apoptosis and that this effect is counteracted by JNK-dominant negative overexpression or specific JNK inhibitor [43].
It has to be considered that the role of both JNK and p38 in apoptosis is rather controversial [103,104,105]. In contrast to the previous findings supporting an antiapoptotic role of JNK and p38, Won et al. have provided evidence that PN inhibits the activity of both the kinases, leading to the sensitization of JB6 cells to UVB-induced apoptosis [92]. In general, the exact role of JNK and p38 in cell death varies in relationship with a number of factors that include the cell type, the kind of stimulus and the crosstalk with other signaling pathways [106].
PN has also been shown to influence the apoptotic extrinsic pathway through the modulation of the signal cascade downstream death receptors such as Apo1/Fas and TRAIL.
In this regard, Qin et al. have found that PN is capable of reversing the suppression of Fas-mediated apoptosis by TNF-α in acute myeloid leukemia cells, an effect which is most likely due to NF-κB inhibition and the consequent repression of antiapoptotic genes [107].
Evidence has indicated that PN is able to overcome resistance to TRAIL-induced apoptosis. According to the findings of Nakshatri et al. [102] PN can induce sensitization to TRAIL in resistant breast cancer, an event that is correlated with JNK activity. The apoptotic effects of PN are specifically described in the next paragraph.

4. Parthenolide and Cell Death

4.1. Classic Apoptosis

Apoptosis represents the major form of programmed cell death in multicellular organisms, and its execution is prevalently dependent on the activation of caspases [108]. Upstream events that trigger caspase-dependent cell death include receptor-mediated apoptotic extrinsic pathway and mitochondria-mediated intrinsic pathway [109,110].
A number of papers have reported that PN induces apoptosis in tumor cells either stimulating the apoptotic extrinsic or intrinsic pathway (Figure 4) [111]. The involvement of caspases in PN-induced cell death has been widely documented [85]. In multiple myeloma, for instance, PN has been shown to be capable of inducing a rapid accumulation of cleaved products of caspase-8 and caspase-3 and, to a lesser extent, caspase-9. Consequently, PN induced caspase-dependent cleavage of the antiapoptotic factors MCL-1 and XIAP, thus promoting the execution of apoptosis [112]. However, the same authors suggested that PN cytotoxicity is likely partly caspase-dependent, as they found that pan-caspase inhibitor Z-VAD-fmk only partially protects the cells from the effect of PN.
Massive apoptosis was recently found following PN treatment in different both in vitro and in vivo tumor models [113].
Apoptotic activities of PN also account for synergistic action in combination with other antitumor compounds as described in the previous paragraph.

4.2. Caspase Independent Cell Death

It is widely recognized that the induction of nonapoptotic forms of programmed cell death (PCD) can represent a valid alternative when tumor cells appear to be resistant to classic apoptosis [114]. Caspase-independent PCD includes diverse forms of death that result from the activation of specific pathways and involve other proteases to trigger the terminal events that culminate in cell suicide.
The first evidence that PN could stimulate caspase-independent PCD with necrotic-like morphology (Figure 4) was provided by Pozarowski et al., who found that HL-60 cells died by necrosis, concurrent with atypical apoptosis, after exposure to PN [115]. These authors showed that the different effects induced by PN depended on the concentration used. They demonstrated the presence of activated caspase-3 in cells undergoing apoptosis after treatment with 10 µM PN. On the other hand, at 30 µM PN they observed that most cells did not show any evidence of caspase-3 activation and revealed a necrotic morphology. Necrotic-like features thus appeared at higher PN concentrations, although were not identified as real necrosis.
Nevertheless, necrotic-like phenotype induced by PN was not further characterized, and necroptosis, the main caspase independent necrosis-like PCD form, has not yet been documented to be specifically activated by the compound. However, we have previously found that PN is capable of increasing the expression of RIP1, a major kinase involved in necroptosis [116].
Among caspase-independent cell death types, the type that was originally called “apoptosis-like PCD” includes apoptosis inducing factor (AIF)-mediated cell death. This factor is released by mitochondria under specific stimuli and translocates to the nucleus, where it promotes chromatin condensation and cell death. It has been documented that PN is able to induce cell death through an AIF-dependent mechanism in melanoma and osteosarcoma cells. In this mechanism, dissipation of the mitochondrial transmembrane potential but not the activation of the caspases has been highlighted [117].
Supporting a possible switch toward PN-induced cell death without caspase involvement is the recent observation that, under particular circumstances, PN can reduce the expression of caspase 3 and caspase 9 and is also capable of suppressing their activation [118].
Considering these observations as well as strict crosstalk among different types of cell death, further studies are needed to clarify the mechanisms of PN-induced Caspase independent-PCD in tumor cells.

4.3. Autophagy

Autophagy is a physiological process that leads to the lysosomal degradation of cellular components and damaged organelles, representing a prosurvival cellular response to various types of stress [119,120]. Under certain conditions, this process can evolve into a death event, namely called autophagic cell death [121]. The activation of this pathway is fundamental for tumor cells that show resistance to classical apoptosis and can, therefore, induce recurrence [122,123].
PN can induce cell death through autophagy activation (Figure 4) without the involvement of caspases. Yang et al. [24] demonstrated that in human osteosarcoma cells, PN induces human osteosarcoma cell death by activating autophagy and mitophagy. PN induction of autophagic death is associated with increased ROS and does not involve caspases.
Activation of the autophagic mechanism by PN could be an excellent strategy to enhance the action of chemotherapeutic agents [124]. It has been shown that PN induces autophagy in human promyelocytic leukemia cell line HL-60 and in human epithelial carcinoma HeLa cell line. The researchers also highlighted that the autophagic mechanism is necessary for PN to activate apoptosis [125].
In glioblastoma (GBM) cells, the efficacy of chemotherapy is limited by intrinsic resistance [126]. PN can induce survival inhibition and trigger cell death through autophagic activation in the GBM U373 cell line [127].

5. Parthenolide and Oxidative Stress

Targeting intracellular redox pathways can be considered as a therapeutic approach for cancer [128]. Oxidative stress is a cell condition caused by increased production of reactive oxygen species (ROS) or reduced function of antioxidant defense systems [129]. It is well known that ROS can behave as mediators of apoptosis. Unbalanced intracellular redox status can, in fact, trigger specific events including alteration of mitochondrial function and activation of death signaling pathways [130].
Many lines of evidence indicate that PN can promote apoptosis in tumor cells through the induction of oxidative stress [131,132] (Figure 5). This was first observed by Wen et al., who found that PN-induced apoptosis in hepatoma cells was accompanied with depletion of glutathione (GSH), generation of ROS, reduction of mitochondrial transmembrane potential and activation of caspases. These effects were effectively abrogated by the antioxidant N-acetyl-l-cysteine (NAC) and enhanced by the GSH synthesis inhibitor buthionine sulfoximine (BSO) confirming the role of oxidative stress in PN-induced apoptosis [133].
The principal site of ROS production in the cell is represented by the mitochondrial respiratory chain [134]. It has been demonstrated that treatment of tumor cells with PN exerts profound effects on mitochondria through the involvement of proapoptotic members of the Bcl-2 family, such as Bid, Bax and Bak. Consequent modifications across the mitochondrial membrane, determined by the proapoptotic Bcl-2 family proteins, lead to the subsequent release of mitochondrial death effectors, including cytochrome c and Samc/Diablo [135].
Kim et al. [136] have further shown that Bax translocation to the mitochondria correlates with ROS production during PN-induced apoptosis in cholangiocarcinoma cells. In particular, these authors have found that Bcl-XL-mediated inhibition of Bax translocation can decrease PN-induced ROS generation and subsequently inhibits the reduction in mitochondrial membrane potential and the release of proapoptotic factors from the mitochondria [136].
In addition, it has been reported that PN-induced apoptosis in multiple myeloma (MM) cells involves oxidative stress and that cell sensitivity depends on catalase activity [137]. Interestingly, the same authors found a different expression of catalase in MM cells than in normal lymphocytes, thus explaining the different response to PN. The compound appeared to be ineffective in normal cells that displayed a higher level of catalase [138].
Other authors have also supported the evidence that PN preferentially acts on tumor cells promoting oxidative stress rather than in normal cells [139]. They showed that PN causes oxidative stress in prostate cancer cells but not in prostate epithelial cells. This effect seems to be dependent on a selective stimulation of NADPH oxidase (NOX) by PN in tumor cells with a consequent decrease in thioredoxin reduction and downregulation of FOXO3a targets antioxidant enzyme manganese superoxide dismutase (Mn-SOD) and catalase [139].
PN and DMAPT also induced oxidative stress through activation of NOX in breast cancer cells [116,140]. It was highlighted that after a short treatment (1–8 h) with PN in MDA-MB231, there was an increase in O2•− induced by the activation of NADPH oxidase. With a longer treatment time, PN also induced mitochondrial production of O2•−, likely blocking mitochondrial activities. The induction of oxidative stress by PN was the main cause of reduced cell viability of breast cancer cells [116,140]. Experimental evidence has suggested that NOX involvement is a consequence of PN-induced activation of the EGFR receptor [141].
According to the findings of Kurdi et al., in cardiac myocytes, PN generation of ROS can depend on either mitochondria or NADPH oxidase in relationship with the concentration of the compound used [142]. Specifically, PN generated superoxide anion, and at lower concentrations (<5 μM) the source of superoxide was mainly mitochondria; at higher concentrations (>5 μM) the primary source was NADPH oxidase [142].
Although the precise mechanism of PN induced oxidative stress remains unclear, it seems that its ability to bind thiol groups can account for its effect on reduction of intracellular GSH and protein thiols, such as thioredoxin [139]. In accordance with this hypothesis, a recent report has described the role of PN in modifying extracellular protein thiol groups and identified surface thioredoxin-1 as one of the targets of PN in lymphoma cells [8]. These authors, however, did not provide a precise explanation of how altering the redox state of exofacial thiols modulates cell death and speculated that surface thioredoxin can mediate the crosstalk between exofacial thiols and downstream intracellular events [8].
A direct action of PN on intracellular thiols, including both free GSH and protein thiols, has been previously described by Zhang et al. [7]. These authors found that colorectal cancer cells underwent apoptosis following treatment with PN through depletion of intracellular thiols and concomitant increase in ROS production and intracellular calcium [7].
It is evident that PN effects on the cell redox state and mitochondrial activity are key components of its action mechanism.
Interestingly, chemoresistance often correlates with an increase in antioxidant activity and a reduction in intracellular ROS level [134,143]. In fact, many cancer cells express high levels of the transcription factor Nrf2, which increases the antioxidant defenses of the cells [144,145]. PN inhibits chemoresistance by inhibiting the overexpression of both Nrf2 and downstream targets, such as MnSOD and catalase [146]. PN likely induces Nrf2 inhibition via increased expression of miR-29b-1-5p [147], a miRNA that regulates the inhibition of proliferation and invasion of triple-negative breast cancer cells (TNBC) [148].

6. Parthenolide Selectivity in Tumor Cells and Targeting Cancer Stem Cells

6.1. Selective Action of Parthenolide in Tumor Cells

Evidence has been provided that PN can selectively induce apoptosis in tumor cells rather than in the normal counterparts [86,112,137,139,149] thus displaying a high potential in cancer therapy.
In addition, in vivo studies have shown that PN is capable of suppressing tumor growth and, in some cases, preventing metastasis in different mice xenograft models including mammary carcinosarcoma [41], triple negative breast cancer [116,150], acute leukemia [26], prostate cancer [27,50,151] and renal carcinoma [152].
Interestingly, the antitumor efficacy of PN is also sustained by its ability to overcome cell resistance to conventional chemotherapeutic agents or other antitumor compounds.
The significant in vivo chemosensitizing properties of PN have been evidenced by Sweeney et al., who obtained reduction in metastasis and improved survival in a xenograft model of breast cancer following a combination treatment of PN with docetaxel [153].
Our previous studies demonstrated that PN and its soluble analogue DMAPT also have a selective effect against triple-negative breast cancer cells and not against normal cells (HMEC) [146]. Furthermore, it has been shown that the administration of DMAPT in nude mice carrying xenografts of MDA-MB231 cells results in inhibition of tumor growth, an increase in mouse survival and a reduction in the metastasis process [116].
To date, the only clinical trial based on PN administration to patients with cancer is reported by Curry et al. [154], who conducted a phase 1 trial to evaluate the pharmacokinetics and toxicity of the compound extracted by feverfew. PN was administered as a daily oral tablet in a 28-day cycle, and patients were evaluated for response after every two cycles. Based on their results, PN had no significant toxicity, but when administered in doses of up to 4 mg it did not provide detectable plasma concentrations. This limit was attributed by the authors to the fact that they could not provide desirable levels of PN with the feverfew preparation [154].
It should be noted that PN has no toxic effect in normal cells and can protect against oxidative stress. Some studies show that PN has scavenging activity against reactive oxygen species, thus protecting myoblast cells. In particular, it has been shown that PN modulates mitophagy induced by oxidative stress and protects C2C12 myoblasts from apoptosis, suggesting a potential protective effect against skeletal muscle diseases associated with oxidative stress [17].

6.2. Parthenolide and Cancer Stem Cells

Many studies have shown that in some forms of cancer, there are cells with specific stem-like properties called cancer stem cells (CSCs) [155]. CSCs are considered the “initiating” cells of the tumor and the main reasons behind tumor heterogeneity, resistance to therapy and recurrence [156,157].
Therefore, a drug capable of targeting both CSCs and cancer cells would be an excellent therapeutic strategy for the treatment of cancer patients. Several studies have shown that PN is a compound capable of acting on CSCs.
Breast cancer is the most common form of cancer diagnosed in women worldwide and, despite the development of new diagnostic and therapeutic methods, is a leading cause of death. The presence of CSCs is potentially the cause of treatment failure; drug resistance; metastasis; and relapse after surgery, chemotherapy and radiotherapy in breast cancer.
There are several studies showing the effect of PN in reducing the presence of CSCs in solid and hematological tumors [4,158,159,160], acting mainly on NF-κB inhibition.
The first studies on the cytotoxic effect of PN on CSCs focused on hematological tumors. Leukemia stem cells (LSCs) are the main cause of the onset, growth and recurrence of acute and chronic myeloid leukemia (AML and CML). PN induces apoptosis in primary human AML cells and CML cells, and in LSCs derived from AML without effect in normal hematopoietic cells. The apoptotic mechanism induced by PN is associated with the inhibition of NF-κB, the proapoptotic activation of p53 and the increase of reactive oxygen species (ROS) [26].
Recent studies have shown that PN is able to reduce the presence of LSCs in drug-resistant leukemia K562/ADM cells and increase their sensitivity to apoptosis induced by doxorubicin through the downregulation of P-gp mediated by NF-κB [161].
The cytotoxic effect of PN has also been highlighted in multiple myeloma cancer stem cells (MM-CSCs). Multiple myeloma (MM) is an incurable malignant plasma cell tumor in which nearly all patients succumb to relapse. In vitro studies conducted using a three-dimensional tissue culture system have shown that PN has a potent cytotoxic effect against MM-CSCs [162].
Triple-negative breast cancers (TNBCs) are aggressive forms of breast cancer associated with a high recurrence rate. Carlisi et al. [150] found that PN and its more soluble analog DMAPT induce cytotoxic effects in CSCs of TNBC. It has been shown that both PN and DMAPT reduce the formation of mammospheres in TNBC cell lines (MDA-MB231, BT20 and MDA-MB436). In particular, the compounds exerted a significant inhibitory effect on the viability of stem cells derived from the dissociation of mammosphere, inducing the generation of ROS, mitochondrial dysfunction and cell necrosis [150].
Zhou et al. [163] have demonstrated that PN can inhibit the formation of mammospheres in MCF-7, a breast cancer line responsive to estrogen and progesterone receptors. This effect is mediated by the inhibition of NF-κB activity.
PN was found to increase the cytotoxic effect in CSCs derived from breast tumors by vinorelbine, a semisynthetic vinca alkaloid that acts through disruption of microtubule assembly. Liu et al. [164] developed liposomes containing PN and vinorelbine. Their combined cytotoxic effect was evaluated in MCF-7 and MDA-MB-231 cells and in CSCs derived from them. The antitumor activity of the combined treatment of liposomes containing vinorelbine and PN was also evaluated on MCF-7 xenografts [164].
It is known that the 5-fluorouracil (5-FU) chemotherapeutic agent has a powerful cytotoxic effect on most cancer cells, but not on CSCs, which can make some cancers resistant to this drug. In general, resistance to chemotherapy is caused by the overexpression of one or more ABC transporters, including ABCG2 and ABCB1/multidrug resistance protein 1 (MDR1) [165]. Studies of nasopharyngeal carcinoma cells (NCS) have shown that while 5-FU treatment increases the CSCs population and COX-2 expression, PN reduces CSCs and downregulates COX-2. Studies conducted by Liao et al. [166] have shown that PN induces cell death in CSCs of NCS by inhibiting the NF-κB/COX-2 pathway.

7. Pathenolide Therapeutic Potential and Parthenolide Analogues

7.1. Synergistic Action of PN with Other Coumponds

From the evidence reported so far, PN represents a promising chemotherapy agent. Its effectiveness can be used in synergy with other anticancer agents of various types [22] (Table 1), with the aim of reducing the doses of the compounds used and preventing the onset of chemoresistance.
The combination of PN with Taxol has also been shown to be effective in lung cancer xenografts as described by Zang et al. [167]. Similarly, a PN analogue has been found to suppress in vivo prostate cancer growth and potentiate the effect of docetaxel [27]. The same authors have previously reported the in vivo effects of PN in combination with docetaxel in prostate cancer [168].
As described in the previous paragraph, significant results have also been found by Liu et al. [164], who combined PN stealthy liposomes with vinorelbine stealthy liposomes to treat breast cancer stem cells in both in vitro and in xenograft models.
In breast cancer, PN has also been shown to enhance sensitivity to antiestrogen such as tamoxifen [169] or fulvestrant [170], an effect which was mainly correlated with inhibition of NF-κB.
Other studies [171] of triple negative breast cancer cells have shown that PN sensitizes breast cancer cells to suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor. One study showed that, in MDA-MB231 cells, the association of PN to SAHA inhibits the cytoprotective responses induced by the single compounds, while the cytotoxic effects are enhanced [171].
In lung cancer cells, Fang et al. have observed an increased susceptibility to low doses of oxaliplatin [172].
In addition to its anticancer properties, PN seems to exert beneficial effects in combination with chemotherapeutic drugs as reported by Francescato et al. [173] who showed its ability to reduce cisplatin-induced renal damage in mice.
More recently, evidence has indicated that PN strongly potentiates the effect of epirubicin in breast cancer cells [174].
Preclinical studies have also highlighted the efficacy of PN, or its analogues, in combination with nonconventional antitumor agents including COX-2 inhibitors [175], sulindac [176], steroid anti-inflammatory agents [177], retinoic acid [93], arsenic trioxide [178] and TRAIL [65,102].
Due to inhibition of NF-κB, PN has been shown to increase X-ray sensitivity of tumor cells [50,179]. Moreover, thermoenhancement effects of PN in lung adenocarcinoma cells have suggested a possible combination with hyperthermia as a novel approach in cancer treatment [180,181].

7.2. Parthenolide Anologues

It should be noted that the studies conducted with PN have highlighted both its potential and limitations. The limits of PN are due to its poor solubility in water and poor stability. Researchers have tried to overcome these limits to make the compound even more effective. Several studies have been conducted that have aimed to make PN more soluble and stable while preserving its bioactivities [2,158] (Table 2).
For example, researchers attempted to make PN soluble in water by adding a methyl group and formulating the DMAPT in the form of fumarate salt [26,27].
In a recent study by Jia X et al. [182], the structure of PN was combined with semicarbazone/thiosemicarbazone groups. Semicarbazones and thiosemicarbazones have been extensively studied for their anticancer activities. In fact, both chemical substituents can inhibit kinase activity involved in cell proliferation and the development of tumors. Furthermore, thiosemicarbazones are able to chelate transition metals with effective antitumor activity with the substituted semicarbazone. In Jia X et al’s study, 21 new derivatives of PN semicarbazone or thiosemicarbazone were synthesized. Most semicarbazone derivatives showed greater cytotoxicity against human tumor cell lines than PN. Five synthesized compounds, including four semicarbazones and one thiosemicarbazone, were tested in MC38 tumor-bearing mice. In vivo results showed that a specific semicarbazone greatly reduced tumor burden [182].
A valid strategy to increase the solubility of a drug and make it more selective towards cancer cells is nanoencapsulation. Based on these considerations, the researchers sought to improve the effect of PN.
Karmakar et al. [183] used carboxyl-functionalized nanographene (fGn) delivery to overcome the extreme hydrophobicity of PN. The authors found that administration of fGn augments the anticancer and apoptotic effects of PN in human pancreatic cancer Panc-1 cell lines. This effect was accompanied by an increase in ROS production and mitochondrial dysfunction [183].
Darwish NHE et al. [184] designed polylactide co-glycolide (PLGA) nanoparticles conjugated with antiCD44, a highly expressed transmembrane glycoprotein in leukemia cells and encapsulating PN, in order to improve the selectivity by recognizing tumor cells rather than normal cells. Studies conducted on leukemia cells Kasumi-1, KG-1a, and THP-1 have shown that PLGA-antiCD44-PN nanoparticles significantly reduce cell viability compared to PN alone [184].
Given the abundance of PN in nature by several researchers, it is used to semisynthesize other sesquiterpene lactones such as arglabin [185] (Figure 6A). Arglabin is a sesquiterpene lactone belonging to the guaianolide subclass, isolated from Artemisia species [185,186]. It has strong anticancer activities on leukemia and human oral squamous and lung cancer cells [187,188]. The antitumor action of arglabin is due to its ability to inhibit farnesyl transferase, an enzyme that causes the activation of the proto-oncogene RAS [186].
In addition, micheliolide (MCL), a sesquiterpene lactone that can be produced from PN [185] (Figure 6B), has shown great potential in antitumor treatment [189]. The antitumor action of MCL is due, as well as for PN, to the inhibitions of the NF-κB and the STAT3 signaling pathways [190,191,192]. Furthermore, MCL induces death in leukemia cells by irreversibly activating pyruvate kinase [193]. ACT001 (Figure 6C) is the fumarate salt of dimethylaminomicheliolide as an orally available derivative of MCL, synthesized from PN by Accendatech Co., Ltd. (Tianjin, China) [194]. ACT001 has a strong anticancer action in breast cancer cells [195,196]. It has been shown that ACT001 is also able to inhibit the proliferation of glioma stem cells (GSCs) [197]. Studies conducted in GSC xenografts have shown that ACT001 acts by inhibiting the adipocyte enhancer 1 binding protein (AEBP1), resulting in inhibition of AKT phosphorylation and cell proliferation [197].

8. Conclusions

PN exerts its antitumor action through different mechanisms including transcriptional regulation, epigenetic effects, signal transduction modulation and induction of oxidative stress. Through these modalities, PN is able to induce apoptosis or alternative forms of cell death including “necrosis-like” and “apoptosis-like” programmed cell death as well as autophagic cell death. The compound appears to display selectivity to tumor cells and a particular efficacy in cancer stem cells, which are the cause of relapses and resistance following chemotherapy. Many lines of evidence indicate that PN potentiates the effect of conventional chemotherapeutics or other antitumor drugs in different tumor models. Studies are still underway to make PN a more effective drug by increasing its stability and solubility. For this purpose, some PN analogues have been developed and are currently under investigation in different tumor models. Overall, PN and its analogues represent promising agents in cancer therapy, and studies on these compounds have served as a basis to synthesize or isolate new sesquiterpene lactones that are increasingly effective to specifically target cancer cells.

Author Contributions

S.E. and D.C. conceived the central idea of the article and performed a preliminary extensive literature review; S.E. wrote the manuscript with support from D.C., M.L. and A.D.; A.C. and G.P. composed figures; A.N., M.G., A.D.B. and G.C. performed complementary literature reviews and revised the manuscript. 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.

Acknowledgments

This paper is dedicated to the memory of Giovanni Tesoriere, whose devotion to understanding the biochemical mechanisms of parthenolide action was a source of inspiration.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-FU5-fluorouracil
ABCB1ATP-binding cassette subfamily B member 1
ABCG2ATP-binding cassette subfamily G member 2
AEBP1Adipocyte enhancer-binding protein 1
AIFApoptosis inducing factor
AMLAcute myeloid leukemia
ATMAtaxia-telangiectasia mutated
B-Rafv-Raf murine sarcoma viral oncogene homolog B1
BSOButhionine sulfoximine
C/EBP-αCCAAT enhancer-binding protein alpha
CaM kinasesCa2+/calmodulin-dependent protein kinases
CDKCyclin-dependent kinase
c-FLIPCellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein
CMLChronic myeloid leukemia
c-MYBCellular myeloblastosis
COX-2Cyclooxygenase-2
CSCsCancer stem cells
DMAPTDimethylaminoparthenolide
DNMT1DNA (cytosine-5)-methyltransferase 1
ERKExtracellular signal-regulated kinase
fGnCarboxyl-functionalized nanographene
FRA-1Fos-related antigen 1
GBMGlioblastoma
GSCsGlioma stem cells
GSHGlutathione
HCCHepatocellular carcinoma
HDAC1Histone deacetylase 1
HMECHuman mammary epithelial cells
HOXA-4Homeobox A4
ICAMIntercellular adhesion molecule
IFN-γInterferon gamma
IkBInhibitor of NF-κB
IKCIkB kinase complex
IKKαIkappaB kinase alpha
IKKβIkappaB kinase beta
IL-1Interleukin-1
IL-2Interleukin-2
IL-4Interleukin-4
IL-6Interleukin-6
iNOSInducible nitric oxide synthase
JAKJanus kinase
JNKc-Jun N-terminal kinase
LEF1Lymphoid enhancer-binding factor 1
LSCsLeukemia stem cells
MAPKsMitogen-activated protein kinases
MCL-1Myeloid cell leukemia 1
MDM2Murine double minute 2
MDR1Multidrug resistance protein 1
MITFMelanocyte-inducing transcription factor
MMMultiple myeloma
MM-CSCsMultiple myeloma cancer stem cells
Mn-SODManganese superoxide dismutase
NACN-acetyl-l-cysteine
NEMONF-κB essential modulator
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NOXNADPH oxidase
NPCNasopharyngeal carcinoma cells
PCDProgrammed cell death
PGE2Prostaglandin E2
PI3-KPhosphoinositide 3-kinase
PKCProtein kinase C
PLGAPolylactide co-glycolide
PNParthenolide
RIP1Receptor-interacting protein 1
ROSReactive oxygen species
SAHASuberoylanilide hydroxamic acid
SP1Specificity protein 1
SRFSerum response factor
STATSignal transducer and activator of transcription
STAT-3Signal transducer and activator of transcription 3
TCF4Transcription factor 4
TLR4Toll-like receptor 4
TNBCTriple-negative breast cancer
TNF-αTumor necrosis factor alpha
TRAILTNF-related apoptosis-inducing ligand
UVBUltraviolet B-rays
XBP1X-box binding protein 1
XIAPX-linked inhibitor of apoptosis

References

  1. Sur, R.; Martin, K.; Liebel, F.; Lyte, P.; Shapiro, S.; Southall, M. Anti-Inflammatory Activity of Parthenolide-Depleted Feverfew (Tanacetum Parthenium). Inflammopharmacology 2009, 17, 42–49. [Google Scholar] [CrossRef]
  2. Freund, R.R.A.; Gobrecht, P.; Fischer, D.; Arndt, H.-D. Advances in Chemistry and Bioactivity of Parthenolide. Nat. Prod. Rep. 2020, 37, 541–565. [Google Scholar] [CrossRef]
  3. Juliana, C.; Fernandes-Alnemri, T.; Wu, J.; Datta, P.; Solorzano, L.; Yu, J.-W.; Meng, R.; Quong, A.A.; Latz, E.; Scott, C.P.; et al. Anti-Inflammatory Compounds Parthenolide and Bay 11-7082 Are Direct Inhibitors of the Inflammasome. J. Biol. Chem. 2010, 285, 9792–9802. [Google Scholar] [CrossRef] [Green Version]
  4. Ghantous, A.; Sinjab, A.; Herceg, Z.; Darwiche, N. Parthenolide: From Plant Shoots to Cancer Roots. Drug Discov. Today 2013, 18, 894–905. [Google Scholar] [CrossRef]
  5. Pajak, B.; Gajkowska, B.; Orzechowski, A. Molecular Basis of Parthenolide-Dependent Proapoptotic Activity in Cancer Cells. Folia Histochem. Cytobiol. 2008, 46, 129–135. [Google Scholar] [CrossRef] [Green Version]
  6. Li-Weber, M.; Palfi, K.; Giaisi, M.; Krammer, P.H. Dual Role of the Anti-Inflammatory Sesquiterpene Lactone: Regulation of Life and Death by Parthenolide. Cell Death Differ. 2005, 12, 408–409. [Google Scholar] [CrossRef]
  7. Zhang, S.; Ong, C.-N.; Shen, H.-M. Critical Roles of Intracellular Thiols and Calcium in Parthenolide-Induced Apoptosis in Human Colorectal Cancer Cells. Cancer Lett. 2004, 208, 143–153. [Google Scholar] [CrossRef]
  8. Skalska, J.; Brookes, P.S.; Nadtochiy, S.M.; Hilchey, S.P.; Jordan, C.T.; Guzman, M.L.; Maggirwar, S.B.; Briehl, M.M.; Bernstein, S.H. Modulation of Cell Surface Protein Free Thiols: A Potential Novel Mechanism of Action of the Sesquiterpene Lactone Parthenolide. PLoS ONE 2009, 4, e8115. [Google Scholar] [CrossRef]
  9. Dawood, M.; Ooko, E.; Efferth, T. Collateral Sensitivity of Parthenolide via NF-κB and HIF-α Inhibition and Epigenetic Changes in Drug-Resistant Cancer Cell Lines. Front. Pharmacol. 2019, 10, 542. [Google Scholar] [CrossRef] [Green Version]
  10. Nam, Y.J.; Lee, D.H.; Lee, M.S.; Lee, C.S. Sesquiterpene Lactone Parthenolide Attenuates Production of Inflammatory Mediators by Suppressing the Toll-like Receptor-4-Mediated Activation of the Akt, MTOR, and NF-κB Pathways. Naunyn-Schmiedeberg’s Arch. Pharm. 2015, 388, 921–930. [Google Scholar] [CrossRef]
  11. López-Franco, O.; Hernández-Vargas, P.; Ortiz-Muñoz, G.; Sanjuán, G.; Suzuki, Y.; Ortega, L.; Blanco, J.; Egido, J.; Gómez-Guerrero, C. Parthenolide Modulates the NF-κB–Mediated Inflammatory Responses in Experimental Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 1864–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Rahman, M.M.; Mohamed, M.R.; Kim, M.; Smallwood, S.; McFadden, G. Co-Regulation of NF-κB and Inflammasome-Mediated Inflammatory Responses by Myxoma Virus Pyrin Domain-Containing Protein M013. PLoS Pathog. 2009, 5, e1000635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB Signaling in Inflammation. Signal Transduct. Target. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zinatizadeh, M.R.; Schock, B.; Chalbatani, G.M.; Zarandi, P.K.; Jalali, S.A.; Miri, S.R. The Nuclear Factor Kappa B (NF-κB) Signaling in Cancer Development and Immune Diseases. Genes Dis. 2021, 8, 287–297. [Google Scholar] [CrossRef]
  15. Aoyagi, T.; Terracina, K.P.; Raza, A.; Matsubara, H.; Takabe, K. Cancer Cachexia, Mechanism and Treatment. World J. Gastrointest. Oncol. 2015, 7, 17–29. [Google Scholar] [CrossRef]
  16. Santos, J.M.O.; Moreira-Pais, A.; Neto, T.; Peixoto da Silva, S.; Oliveira, P.A.; Ferreira, R.; Mendes, J.; Bastos, M.M.S.M.; Lopes, C.; Casaca, F.; et al. Dimethylaminoparthenolide Reduces the Incidence of Dysplasia and Ameliorates a Wasting Syndrome in HPV16-Transgenic Mice. Drug Dev. Res. 2019, 80, 824–830. [Google Scholar] [CrossRef]
  17. Ren, Y.; Li, Y.; Lv, J.; Guo, X.; Zhang, J.; Zhou, D.; Zhang, Z.; Xue, Z.; Yang, G.; Xi, Q.; et al. Parthenolide Regulates Oxidative Stress-Induced Mitophagy and Suppresses Apoptosis through P53 Signaling Pathway in C2C12 Myoblasts. J. Cell. Biochem. 2019, 120, 15695–15708. [Google Scholar] [CrossRef]
  18. Mathema, V.B.; Koh, Y.-S.; Thakuri, B.C.; Sillanpää, M. Parthenolide, a Sesquiterpene Lactone, Expresses Multiple Anti-Cancer and Anti-Inflammatory Activities. Inflammation 2012, 35, 560–565. [Google Scholar] [CrossRef]
  19. Wang, M.; Li, Q. Parthenolide Could Become a Promising and Stable Drug with Anti-Inflammatory Effects. Nat. Prod. Res. 2015, 29, 1092–1101. [Google Scholar] [CrossRef]
  20. Chen, C.-F.C.; Leung, A.Y.L.Y. Gene Response of Human Monocytic Cells for the Detection of Antimigraine Activity of Feverfew ExtractsThis Article Is One of a Selection of Papers Published in This Special Issue (Part 2 of 2) on the Safety and Efficacy of Natural Health Products. Can. J. Physiol. Pharmacol. 2007. [Google Scholar] [CrossRef]
  21. Brown, R.P.; Gerbarg, P.L. Herbs and Nutrients in the Treatment of Depression, Anxiety, Insomnia, Migraine, and Obesity. J. Psychiatr. Pract. 2001, 7, 75–91. [Google Scholar] [CrossRef] [PubMed]
  22. Sztiller-Sikorska, M.; Czyz, M. Parthenolide as Cooperating Agent for Anti-Cancer Treatment of Various Malignancies. Pharmaceuticals 2020, 13, 194. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, Y.C.; Kim, S.L.; Park, Y.R.; Lee, S.-T.; Kim, S.W. Parthenolide Promotes Apoptotic Cell Death and Inhibits the Migration and Invasion of SW620 Cells. Intestig. Res. 2017, 15, 174–181. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, C.; Yang, Q.O.; Kong, Q.-J.; Yuan, W.; Yang, Y.-P.O. Parthenolide Induces Reactive Oxygen Species-Mediated Autophagic Cell Death in Human Osteosarcoma Cells. Cell. Physiol. Biochem. 2016, 40, 146–154. [Google Scholar] [CrossRef]
  25. Hexum, J.K.; Becker, C.M.; Kempema, A.M.; Ohlfest, J.R.; Largaespada, D.A.; Harki, D.A. Parthenolide Prodrug LC-1 Slows Growth of Intracranial Glioma. Bioorg. Med. Chem. Lett. 2015, 25, 2493–2495. [Google Scholar] [CrossRef]
  26. Guzman, M.L.; Rossi, R.M.; Neelakantan, S.; Li, X.; Corbett, C.A.; Hassane, D.C.; Becker, M.W.; Bennett, J.M.; Sullivan, E.; Lachowicz, J.L.; et al. An Orally Bioavailable Parthenolide Analog Selectively Eradicates Acute Myelogenous Leukemia Stem and Progenitor Cells. Blood 2007, 110, 4427–4435. [Google Scholar] [CrossRef]
  27. Shanmugam, R.; Kusumanchi, P.; Cheng, L.; Crooks, P.; Neelakantan, S.; Matthews, W.; Nakshatri, H.; Sweeney, C.J. A Water-Soluble Parthenolide Analogue Suppresses in Vivo Prostate Cancer Growth by Targeting NFκB and Generating Reactive Oxygen Species. Prostate 2010, 70, 1074–1086. [Google Scholar] [CrossRef]
  28. Avci, N.G.; Ebrahimzadeh-Pustchi, S.; Akay, Y.M.; Esquenazi, Y.; Tandon, N.; Zhu, J.-J.; Akay, M. NF-κB Inhibitor with Temozolomide Results in Significant Apoptosis in Glioblastoma via the NF-κB(P65) and Actin Cytoskeleton Regulatory Pathways. Sci. Rep. 2020, 10, 13352. [Google Scholar] [CrossRef]
  29. Arora, R.; Yates, C.; Gary, B.D.; McClellan, S.; Tan, M.; Xi, Y.; Reed, E.; Piazza, G.A.; Owen, L.B.; Dean-Colomb, W. Panepoxydone Targets NF-κB and FOXM1 to Inhibit Proliferation, Induce Apoptosis and Reverse Epithelial to Mesenchymal Transition in Breast Cancer. PLoS ONE 2014, 9, e98370. [Google Scholar] [CrossRef] [Green Version]
  30. Singh, V.; Gupta, D.; Arora, R. NF-κB as a Key Player in Regulation of Cellular Radiation Responses and Identification of Radiation Countermeasures. Discoveries 2015, 3, e35. [Google Scholar] [CrossRef] [Green Version]
  31. Hellweg, C.E. The Nuclear Factor ΚB Pathway: A Link to the Immune System in the Radiation Response. Cancer Lett. 2015, 368, 275–289. [Google Scholar] [CrossRef] [PubMed]
  32. Benedetti, G.; Fredriksson, L.; Herpers, B.; Meerman, J.; van de Water, B.; de Graauw, M. TNF-α-Mediated NF-κB Survival Signaling Impairment by Cisplatin Enhances JNK Activation Allowing Synergistic Apoptosis of Renal Proximal Tubular Cells. Biochem. Pharmacol. 2013, 85, 274–286. [Google Scholar] [CrossRef] [PubMed]
  33. Rex, J.; Lutz, A.; Faletti, L.E.; Albrecht, U.; Thomas, M.; Bode, J.G.; Borner, C.; Sawodny, O.; Merfort, I. IL-1β and TNFα Differentially Influence NF-κB Activity and FasL-Induced Apoptosis in Primary Murine Hepatocytes during LPS-Induced Inflammation. Front. Physiol. 2019, 10, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lingappan, K. NF-κB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef] [PubMed]
  35. Morgan, M.J.; Liu, Z. Crosstalk of Reactive Oxygen Species and NF-κB Signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [Green Version]
  36. Chaithongyot, S.; Jantaree, P.; Sokolova, O.; Naumann, M. NF-κB in Gastric Cancer Development and Therapy. Biomedicines 2021, 9, 870. [Google Scholar] [CrossRef]
  37. Zhang, Q.; Lenardo, M.J.; Baltimore, D. 30 Years of NF-κB: A Blossoming of Relevance to Human Pathobiology. Cell 2017, 168, 37–57. [Google Scholar] [CrossRef] [Green Version]
  38. Singh, S.; Singh, T.G. Role of Nuclear Factor Kappa B (NF-κB) Signalling in Neurodegenerative Diseases: An Mechanistic Approach. Curr. Neuropharmacol. 2020, 18, 918–935. [Google Scholar] [CrossRef]
  39. Hehner, S.P.; Hofmann, T.G.; Dröge, W.; Schmitz, M.L. The Antiinflammatory Sesquiterpene Lactone Parthenolide Inhibits NF-κB by Targeting the IκB Kinase Complex. J. Immunol. 1999, 163, 5617–5623. [Google Scholar]
  40. Kwok, B.H.B.; Koh, B.; Ndubuisi, M.I.; Elofsson, M.; Crews, C.M. The Anti-Inflammatory Natural Product Parthenolide from the Medicinal Herb Feverfew Directly Binds to and Inhibits IκB Kinase. Chem. Biol. 2001, 8, 759–766. [Google Scholar] [CrossRef] [Green Version]
  41. Idris, A.I.; Libouban, H.; Nyangoga, H.; Landao-Bassonga, E.; Chappard, D.; Ralston, S.H. Pharmacologic Inhibitors of IκB Kinase Suppress Growth and Migration of Mammary Carcinosarcoma Cells in Vitro and Prevent Osteolytic Bone Metastasis in Vivo. Mol. Cancer 2009, 8, 2339–2347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yip, K.H.M.; Zheng, M.H.; Feng, H.T.; Steer, J.H.; Joyce, D.A.; Xu, J. Sesquiterpene Lactone Parthenolide Blocks Lipopolysaccharide-Induced Osteolysis Through the Suppression of NF-κB Activity. J. Bone Miner. Res. 2004, 19, 1905–1916. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, S.; Lin, Z.-N.; Yang, C.-F.; Shi, X.; Ong, C.-N.; Shen, H.-M. Suppressed NF-κB and Sustained JNK Activation Contribute to the Sensitization Effect of Parthenolide to TNF-α-Induced Apoptosis in Human Cancer Cells. Carcinogenesis 2004, 25, 2191–2199. [Google Scholar] [CrossRef] [PubMed]
  44. Buhrmann, C.; Shayan, P.; Banik, K.; Kunnumakkara, A.B.; Kubatka, P.; Koklesova, L.; Shakibaei, M. Targeting NF-κB Signaling by Calebin A, a Compound of Turmeric, in Multicellular Tumor Microenvironment: Potential Role of Apoptosis Induction in CRC Cells. Biomedicines 2020, 8, 236. [Google Scholar] [CrossRef]
  45. Park, S.H.; Riley, P.; Frisch, S.M. Regulation of Anoikis by Deleted in Breast Cancer-1 (DBC1) through NF-κB. Apoptosis 2013, 18, 949–962. [Google Scholar] [CrossRef] [Green Version]
  46. Verzella, D.; Pescatore, A.; Capece, D.; Vecchiotti, D.; Ursini, M.V.; Franzoso, G.; Alesse, E.; Zazzeroni, F. Life, Death, and Autophagy in Cancer: NF-κB Turns up Everywhere. Cell Death Dis. 2020, 11, 210. [Google Scholar] [CrossRef] [Green Version]
  47. Xia, Y.; Shen, S.; Verma, I.M. NF-κB, an Active Player in Human Cancers. Cancer Immunol. Res. 2014, 2, 823–830. [Google Scholar] [CrossRef] [Green Version]
  48. Kim, S.-L.; Kim, S.H.; Park, Y.R.; Liu, Y.-C.; Kim, E.-M.; Jeong, H.-J.; Kim, Y.N.; Seo, S.Y.; Kim, I.H.; Lee, S.O.; et al. Combined Parthenolide and Balsalazide Have Enhanced Antitumor Efficacy Through Blockade of NF-κB Activation. Mol. Cancer Res. 2017, 15, 141–151. [Google Scholar] [CrossRef] [Green Version]
  49. Kim, S.-L.; Kim, S.H.; Trang, K.T.T.; Kim, I.H.; Lee, S.-O.; Lee, S.T.; Kim, D.G.; Kang, S.-B.; Kim, S.-W. Synergistic Antitumor Effect of 5-Fluorouracil in Combination with Parthenolide in Human Colorectal Cancer. Cancer Lett. 2013, 335, 479–486. [Google Scholar] [CrossRef]
  50. Mendonca, M.S.; Chin-Sinex, H.; Gomez-Millan, J.; Datzman, N.; Hardacre, M.; Comerford, K.; Nakshatri, H.; Nye, M.; Benjamin, L.; Mehta, S.; et al. Parthenolide Sensitizes Cells to X-Ray-Induced Cell Killing through Inhibition of NF-κB and Split-Dose Repair. Radiat. Res. 2007, 168, 689–697. [Google Scholar] [CrossRef]
  51. Sun, Y.; Clair, D.K.S.; Fang, F.; Warren, G.W.; Rangnekar, V.M.; Crooks, P.A.; Clair, W.H.S. The Radiosensitization Effect of Parthenolide in Prostate Cancer Cells Is Mediated by Nuclear Factor-ΚB Inhibition and Enhanced by the Presence of PTEN. Mol. Cancer 2007, 6, 2477–2486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Shao, L.; Wu, L.; Zhou, D. Sensitization of Tumor Cells to Cancer Therapy by Molecularly Targeted Inhibition of the Inhibitor of Nuclear Factor ΚB Kinase. Transl. Cancer Res. 2012, 1, 100–108. [Google Scholar] [CrossRef] [PubMed]
  53. Morel, K.L.; Ormsby, R.J.; Bezak, E.; Sweeney, C.J.; Sykes, P.J. Parthenolide Selectively Sensitizes Prostate Tumor Tissue to Radiotherapy While Protecting Healthy Tissues In Vivo. Radiat. Res. 2017, 187, 501–512. [Google Scholar] [CrossRef] [PubMed]
  54. Yin, Q.; Wang, L.; Yu, H.; Chen, D.; Zhu, W.; Sun, C. Pharmacological Effects of Polyphenol Phytochemicals on the JAK-STAT Signaling Pathway. Front. Pharmacol. 2021, 12, 2350. [Google Scholar] [CrossRef] [PubMed]
  55. Verhoeven, Y.; Tilborghs, S.; Jacobs, J.; De Waele, J.; Quatannens, D.; Deben, C.; Prenen, H.; Pauwels, P.; Trinh, X.B.; Wouters, A.; et al. The Potential and Controversy of Targeting STAT Family Members in Cancer. Semin. Cancer Biol. 2020, 60, 41–56. [Google Scholar] [CrossRef]
  56. Awasthi, N.; Liongue, C.; Ward, A.C. STAT Proteins: A Kaleidoscope of Canonical and Non-Canonical Functions in Immunity and Cancer. J. Hematol. Oncol. 2021, 14, 198. [Google Scholar] [CrossRef]
  57. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT Signaling Pathway: From Bench to Clinic. Signal Transduct. Target. 2021, 6, 402. [Google Scholar] [CrossRef]
  58. Arumuggam, N.; Bhowmick, N.A.; Rupasinghe, H.P.V. A Review: Phytochemicals Targeting JAK/STAT Signaling and IDO Expression in Cancer. Phytother. Res. 2015, 29, 805–817. [Google Scholar] [CrossRef]
  59. Heppler, L.N.; Frank, D.A. Targeting Oncogenic Transcription Factors: Therapeutic Implications of Endogenous STAT Inhibitors. Trends Cancer 2017, 3, 816–827. [Google Scholar] [CrossRef]
  60. Tolomeo, M.; Cascio, A. The Multifaced Role of STAT3 in Cancer and Its Implication for Anticancer Therapy. Int. J. Mol. Sci. 2021, 22, 603. [Google Scholar] [CrossRef]
  61. Tošić, I.; Frank, D.A. STAT3 as a Mediator of Oncogenic Cellular Metabolism: Pathogenic and Therapeutic Implications. Neoplasia 2021, 23, 1167–1178. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, Y.; Liao, S.; Bennett, S.; Tang, H.; Song, D.; Wood, D.; Zhan, X.; Xu, J. STAT3 and Its Targeting Inhibitors in Osteosarcoma. Cell Prolif. 2021, 54, e12974. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, C.; Li, B.; Gaikwad, A.S.; Haridas, V.; Xu, Z.; Gutterman, J.U.; Duvic, M. Avicin D Selectively Induces Apoptosis and Downregulates P-STAT-3, Bcl-2, and Survivin in Cutaneous T-Cell Lymphoma Cells. J. Investig. Dermatol. 2008, 128, 2728–2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Loh, C.-Y.; Arya, A.; Naema, A.F.; Wong, W.F.; Sethi, G.; Looi, C.Y. Signal Transducer and Activator of Transcription (STATs) Proteins in Cancer and Inflammation: Functions and Therapeutic Implication. Front. Oncol. 2019, 9, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Carlisi, D.; D’Anneo, A.; Angileri, L.; Lauricella, M.; Emanuele, S.; Santulli, A.; Vento, R.; Tesoriere, G. Parthenolide Sensitizes Hepatocellular Carcinoma Cells to Trail by Inducing the Expression of Death Receptors through Inhibition of STAT3 Activation. J. Cell. Physiol. 2011, 226, 1632–1641. [Google Scholar] [CrossRef] [PubMed]
  66. Sobota, R.; Szwed, M.; Kasza, A.; Bugno, M.; Kordula, T. Parthenolide Inhibits Activation of Signal Transducers and Activators of Transcription (STATs) Induced by Cytokines of the IL-6 Family. Biochem. Biophys. Res. Commun. 2000, 267, 329–333. [Google Scholar] [CrossRef]
  67. Liu, M.; Xiao, C.; Sun, M.; Tan, M.; Hu, L.; Yu, Q. Parthenolide Inhibits STAT3 Signaling by Covalently Targeting Janus Kinases. Molecules 2018, 23, 1478. [Google Scholar] [CrossRef] [Green Version]
  68. Cui, Z.-Y.; Wang, G.; Zhang, J.; Song, J.; Jiang, Y.-C.; Dou, J.-Y.; Lian, L.-H.; Nan, J.-X.; Wu, Y.-L. Parthenolide, Bioactive Compound of Chrysanthemum parthenium L., Ameliorates Fibrogenesis and Inflammation in Hepatic Fibrosis via Regulating the Crosstalk of TLR4 and STAT3 Signaling Pathway. Phytother. Res. 2021, 35, 5680–5693. [Google Scholar] [CrossRef]
  69. Li, H.; Lu, H.; Lv, M.; Wang, Q.; Sun, Y. Parthenolide Facilitates Apoptosis and Reverses Drug-Resistance of Human Gastric Carcinoma Cells by Inhibiting the STAT3 Signaling Pathway. Oncol. Lett. 2018, 15, 3572–3579. [Google Scholar] [CrossRef] [Green Version]
  70. Mohammadi, S.; Zahedpanah, M.; Nikbakht, M.; Shaiegan, M.; Hamidollah, G.; Nikugoftar, M.; Rahmani, B.; Hamedi Asl, D. Parthenolide Reduces Gene Transcription of Prosurvival Mediators in U937 Cells. Exp. Oncol. 2017, 39, 30–35. [Google Scholar] [CrossRef]
  71. Wu, Z.; Nicoll, M.; Ingham, R.J. AP-1 Family Transcription Factors: A Diverse Family of Proteins That Regulate Varied Cellular Activities in Classical Hodgkin Lymphoma and ALK+ ALCL. Exp. Hematol. Oncol. 2021, 10, 4. [Google Scholar] [CrossRef] [PubMed]
  72. Atsaves, V.; Leventaki, V.; Rassidakis, G.Z.; Claret, F.X. AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers 2019, 11, 1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hartman, M.L.; Talar, B.; Sztiller-Sikorska, M.; Nejc, D.; Czyz, M. Parthenolide Induces MITF-M Downregulation and Senescence in Patient-Derived MITF-M High Melanoma Cell Populations. Oncotarget 2016, 7, 9026–9040. [Google Scholar] [CrossRef]
  74. Zhang, J.; Mou, Y.; Gong, H.; Chen, H.; Xiao, H. Microphthalmia-Associated Transcription Factor in Senescence and Age-Related Diseases. Gerontology 2021, 67, 708–717. [Google Scholar] [CrossRef] [PubMed]
  75. Bourseguin, J.; Bonet, C.; Renaud, E.; Pandiani, C.; Boncompagni, M.; Giuliano, S.; Pawlikowska, P.; Karmous-Benailly, H.; Ballotti, R.; Rosselli, F.; et al. FANCD2 Functions as a Critical Factor Downstream of MiTF to Maintain the Proliferation and Survival of Melanoma Cells. Sci. Rep. 2016, 6, 36539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Trejo-Solis, C.; Escamilla-Ramirez, A.; Jimenez-Farfan, D.; Castillo-Rodriguez, R.A.; Flores-Najera, A.; Cruz-Salgado, A. Crosstalk of the Wnt/β-Catenin Signaling Pathway in the Induction of Apoptosis on Cancer Cells. Pharmaceuticals 2021, 14, 871. [Google Scholar] [CrossRef]
  77. Yu, F.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C.; Ye, L. Wnt/β-Catenin Signaling in Cancers and Targeted Therapies. Signal Transduct. Target. 2021, 6, 307. [Google Scholar] [CrossRef]
  78. Zhu, X.; Yuan, C.; Tian, C.; Li, C.; Nie, F.; Song, X.; Zeng, R.; Wu, D.; Hao, X.; Li, L. The Plant Sesquiterpene Lactone Parthenolide Inhibits Wnt/β-Catenin Signaling by Blocking Synthesis of the Transcriptional Regulators TCF4/LEF1. J. Biol. Chem. 2018, 293, 5335–5344. [Google Scholar] [CrossRef] [Green Version]
  79. Lees, A.; Sessler, T.; McDade, S. Dying to Survive—The P53 Paradox. Cancers 2021, 13, 3257. [Google Scholar] [CrossRef]
  80. Lukin, D.J.; Carvajal, L.A.; Liu, W.; Resnick-Silverman, L.; Manfredi, J.J. P53 Promotes Cell Survival Due to the Reversibility of Its Cell-Cycle Checkpoints. Mol. Cancer Res. 2015, 13, 16–28. [Google Scholar] [CrossRef] [Green Version]
  81. Guadagno, J.; Swan, P.; Shaikh, R.; Cregan, S.P. Microglia-Derived IL-1β Triggers P53-Mediated Cell Cycle Arrest and Apoptosis in Neural Precursor Cells. Cell Death Dis. 2015, 6, e1779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Hou, H.; Sun, D.; Zhang, X. The Role of MDM2 Amplification and Overexpression in Therapeutic Resistance of Malignant Tumors. Cancer Cell Int. 2019, 19, 216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Gopal, Y.N.V.; Chanchorn, E.; Dyke, M.W.V. Parthenolide Promotes the Ubiquitination of MDM2 and Activates P53 Cellular Functions. Mol. Cancer 2009, 8, 552–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Al-Fatlawi, A.A.; Al-Fatlawi, A.A.; Irshad, M.; Rahisuddin; Ahmad, A. Effect of parthenolide on growth and apoptosis regulatory genes of human cancer cell lines. Pharm. Biol. 2015, 53, 104–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Talib, W.H.; Al Kury, L.T. Parthenolide Inhibits Tumor-Promoting Effects of Nicotine in Lung Cancer by Inducing P53—Dependent Apoptosis and Inhibiting VEGF Expression. Biomed. Pharmacother. 2018, 107, 1488–1495. [Google Scholar] [CrossRef]
  86. Steele, A.J.; Jones, D.T.; Ganeshaguru, K.; Duke, V.M.; Yogashangary, B.C.; North, J.M.; Lowdell, M.W.; Kottaridis, P.D.; Mehta, A.B.; Prentice, A.G.; et al. The Sesquiterpene Lactone Parthenolide Induces Selective Apoptosis of B-Chronic Lymphocytic Leukemia Cells in Vitro. Leukemia 2006, 20, 1073–1079. [Google Scholar] [CrossRef]
  87. Ghantous, A.; Saikali, M.; Rau, T.; Gali-Muhtasib, H.; Schneider-Stock, R.; Darwiche, N. Inhibition of Tumor Promotion by Parthenolide: Epigenetic Modulation of P21. Cancer Prev. Res. 2012, 5, 1298–1309. [Google Scholar] [CrossRef] [Green Version]
  88. Dey, S.; Sarkar, M.; Giri, B. Anti-Inflammatory and Anti-Tumor Activities of Parthenolide: An Update. J. Chem. Biol. 2016, 1, 107. [Google Scholar] [CrossRef] [Green Version]
  89. Gopal, Y.N.V.; Arora, T.S.; Van Dyke, M.W. Parthenolide Specifically Depletes Histone Deacetylase 1 Protein and Induces Cell Death through Ataxia Telangiectasia Mutated. Chem. Biol. 2007, 14, 813–823. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, Z.; Liu, S.; Xie, Z.; Pavlovicz, R.E.; Wu, J.; Chen, P.; Aimiuwu, J.; Pang, J.; Bhasin, D.; Neviani, P.; et al. Modulation of DNA Methylation by a Sesquiterpene Lactone Parthenolide. J. Pharm. Exp. 2009, 329, 505–514. [Google Scholar] [CrossRef] [Green Version]
  91. Irshad, R.; Husain, M. Natural Products in the Reprogramming of Cancer Epigenetics. Toxicol. Appl. Pharmacol. 2021, 417, 115467. [Google Scholar] [CrossRef] [PubMed]
  92. Won, Y.-K.; Ong, C.-N.; Shen, H.-M. Parthenolide Sensitizes Ultraviolet (UV)-B-Induced Apoptosis via Protein Kinase C-Dependent Pathways. Carcinogenesis 2005, 26, 2149–2156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kim, S.H.; Danilenko, M.; Kim, T.S. Differential Enhancement of Leukaemia Cell Differentiation without Elevation of Intracellular Calcium by Plant-Derived Sesquiterpene Lactone Compounds. Br. J. Pharm. 2008, 155, 814–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kawasaki, B.T.; Hurt, E.M.; Kalathur, M.; Duhagon, M.A.; Milner, J.A.; Kim, Y.S.; Farrar, W.L. Effects of the Sesquiterpene Lactone Parthenolide on Prostate Tumor-Initiating Cells: An Integrated Molecular Profiling Approach. Prostate 2009, 69, 827–837. [Google Scholar] [CrossRef] [Green Version]
  95. Lin, M.; Bi, H.; Yan, Y.; Huang, W.; Zhang, G.; Zhang, G.; Tang, S.; Liu, Y.; Zhang, L.; Ma, J.; et al. Parthenolide Suppresses Non-Small Cell Lung Cancer GLC-82 Cells Growth via B-Raf/MAPK/Erk Pathway. Oncotarget 2017, 8, 23436–23447. [Google Scholar] [CrossRef] [Green Version]
  96. Li, X.; Huang, R.; Li, M.; Zhu, Z.; Chen, Z.; Cui, L.; Luo, H.; Luo, L. Parthenolide Inhibits the Growth of Non-Small Cell Lung Cancer by Targeting Epidermal Growth Factor Receptor. Cancer Cell Int. 2020, 20, 561. [Google Scholar] [CrossRef]
  97. Berdan, C.A.; Ho, R.; Lehtola, H.S.; To, M.; Hu, X.; Huffman, T.R.; Petri, Y.; Altobelli, C.R.; Demeulenaere, S.G.; Olzmann, J.A.; et al. Parthenolide Covalently Targets and Inhibits Focal Adhesion Kinase in Breast Cancer Cells. Cell Chem. Biol. 2019, 26, 1027–1035. [Google Scholar] [CrossRef]
  98. Li, X.; Kong, L.; Yang, Q.; Duan, A.; Ju, X.; Cai, B.; Chen, L.; An, T.; Li, Y. Parthenolide Inhibits Ubiquitin-Specific Peptidase 7 (USP7), Wnt Signaling, and Colorectal Cancer Cell Growth. J. Biol. Chem. 2020, 295, 3576–3589. [Google Scholar] [CrossRef]
  99. Yue, J.; López, J.M. Understanding MAPK Signaling Pathways in Apoptosis. Int. J. Mol. Sci. 2020, 21, 2346. [Google Scholar] [CrossRef] [Green Version]
  100. Ki, Y.-W.; Park, J.H.; Lee, J.E.; Shin, I.C.; Koh, H.C. JNK and P38 MAPK Regulate Oxidative Stress and the Inflammatory Response in Chlorpyrifos-Induced Apoptosis. Toxicol. Lett. 2013, 218, 235–245. [Google Scholar] [CrossRef]
  101. Gao, H.-E.; Sun, Y.; Ding, Y.-H.; Long, J.; Liu, X.-L.; Yang, M.; Ji, Q.; Li, Y.-H.; Chen, Y.; Zhang, Q.; et al. Antineoplastic Effects of CPPTL via the ROS/JNK Pathway in Acute Myeloid Leukemia. Oncotarget 2017, 8, 38990–39000. [Google Scholar] [CrossRef] [Green Version]
  102. Nakshatri, H.; Rice, S.E.; Bhat-Nakshatri, P. Antitumor Agent Parthenolide Reverses Resistance of Breast Cancer Cells to Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand through Sustained Activation of c-Jun N-Terminal Kinase. Oncogene 2004, 23, 7330–7344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Sui, X.; Kong, N.; Ye, L.; Han, W.; Zhou, J.; Zhang, Q.; He, C.; Pan, H. P38 and JNK MAPK Pathways Control the Balance of Apoptosis and Autophagy in Response to Chemotherapeutic Agents. Cancer Lett. 2014, 344, 174–179. [Google Scholar] [CrossRef] [PubMed]
  104. Achkar, I.W.; Abdulrahman, N.; Al-Sulaiti, H.; Joseph, J.M.; Uddin, S.; Mraiche, F. Cisplatin Based Therapy: The Role of the Mitogen Activated Protein Kinase Signaling Pathway. J. Transl. Med. 2018, 16, 96. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, S.; Rauch, J.; Kolch, W. Targeting MAPK Signaling in Cancer: Mechanisms of Drug Resistance and Sensitivity. Int. J. Mol. Sci. 2020, 21, 1102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wagner, E.F.; Nebreda, Á.R. Signal Integration by JNK and P38 MAPK Pathways in Cancer Development. Nat. Rev. Cancer 2009, 9, 537–549. [Google Scholar] [CrossRef] [PubMed]
  107. Qin, Y.; Auh, S.; Blokh, L.; Long, C.; Gagnon, I.; Hamann, K.J. TNF-α Induces Transient Resistance to Fas-Induced Apoptosis in Eosinophilic Acute Myeloid Leukemia Cells. Mol. Immunol. 2007, 4, 10. [Google Scholar]
  108. Chen, Y.-T.; Lin, C.-W.; Su, C.-W.; Yang, W.-E.; Chuang, C.-Y.; Su, S.-C.; Hsieh, M.-J.; Yang, S.-F. Magnolol Triggers Caspase-Mediated Apoptotic Cell Death in Human Oral Cancer Cells through JNK1/2 and P38 Pathways. Biomedicines 2021, 9, 1295. [Google Scholar] [CrossRef]
  109. D’Arcy, M.S. Cell Death: A Review of the Major Forms of Apoptosis, Necrosis and Autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef]
  110. Chen, Q.; Kang, J.; Fu, C. The Independence of and Associations among Apoptosis, Autophagy, and Necrosis. Signal Transduct. Target. 2018, 3, 18. [Google Scholar] [CrossRef] [Green Version]
  111. Zhao, X.; Liu, X.; Su, L. Parthenolide Induces Apoptosis via TNFRSF10B and PMAIP1 Pathways in Human Lung Cancer Cells. J. Exp. Clin. Cancer Res. 2014, 33, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Suvannasankha, A.; Crean, C.D.; Shanmugam, R.; Farag, S.S.; Abonour, R.; Boswell, H.S.; Nakshatri, H. Antimyeloma Effects of a Sesquiterpene Lactone Parthenolide. Clin. Cancer Res. 2008, 14, 1814–1822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Karam, L.; Abou Staiteieh, S.; Chaaban, R.; Hayar, B.; Ismail, B.; Neipel, F.; Darwiche, N.; Abou Merhi, R. Anticancer Activities of Parthenolide in Primary Effusion Lymphoma Preclinical Models. Mol. Carcinog. 2021, 60, 567–581. [Google Scholar] [CrossRef] [PubMed]
  114. Emanuele, S.; Oddo, E.; D’Anneo, A.; Notaro, A.; Calvaruso, G.; Lauricella, M.; Giuliano, M. Routes to Cell Death in Animal and Plant Kingdoms: From Classic Apoptosis to Alternative Ways to Die—A Review. Rend. Lincei. Sci. Fis. Nat. 2018, 29, 397–409. [Google Scholar] [CrossRef]
  115. Pozarowski, P.; Halicka, D.H.; Darzynkiewicz, Z. Cell Cycle Effects and Caspase-Dependent and Independent Death of HL-60 and Jurkat Cells Treated with the Inhibitor of NF-KappaB Parthenolide. Cell Cycle 2003, 2, 377–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. D’Anneo, A.; Carlisi, D.; Lauricella, M.; Puleio, R.; Martinez, R.; Di Bella, S.; Di Marco, P.; Emanuele, S.; Di Fiore, R.; Guercio, A.; et al. Parthenolide Generates Reactive Oxygen Species and Autophagy in MDA-MB231 Cells. A Soluble Parthenolide Analogue Inhibits Tumour Growth and Metastasis in a Xenograft Model of Breast Cancer. Cell Death Dis. 2013, 4, e891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. D’Anneo, A.; Carlisi, D.; Lauricella, M.; Emanuele, S.; Di Fiore, R.; Vento, R.; Tesoriere, G. Parthenolide Induces Caspase-Independent and AIF-Mediated Cell Death in Human Osteosarcoma and Melanoma Cells. J. Cell. Physiol. 2013, 228, 952–967. [Google Scholar] [CrossRef] [Green Version]
  118. Yao, H.; Tang, X.; Shao, X.; Feng, L.; Wu, N.; Yao, K. Parthenolide Protects Human Lens Epithelial Cells from Oxidative Stress-Induced Apoptosis via Inhibition of Activation of Caspase-3 and Caspase-9. Cell Res. 2007, 17, 565–571. [Google Scholar] [CrossRef]
  119. Parzych, K.R.; Klionsky, D.J. An Overview of Autophagy: Morphology, Mechanism, and Regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [Green Version]
  120. Emanuele, S.; Lauricella, M.; D’Anneo, A.; Carlisi, D.; De Blasio, A.; Di Liberto, D.; Giuliano, M. P62: Friend or Foe? Evidences for OncoJanus and NeuroJanus Roles. Int. J. Mol. Sci. 2020, 21, 5029. [Google Scholar] [CrossRef]
  121. Lei, Y.; Klionsky, D.J. The Emerging Roles of Autophagy in Human Diseases. Biomedicines 2021, 9, 1651. [Google Scholar] [CrossRef] [PubMed]
  122. Kriel, J.; Loos, B. The Good, the Bad and the Autophagosome: Exploring Unanswered Questions of Autophagy-Dependent Cell Death. Cell Death Differ. 2019, 26, 640–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Pellerito, C.; Emanuele, S.; Ferrante, F.; Celesia, A.; Giuliano, M.; Fiore, T. Tributyltin(IV) Ferulate, a Novel Synthetic Ferulic Acid Derivative, Induces Autophagic Cell Death in Colon Cancer Cells: From Chemical Synthesis to Biochemical Effects. J. Inorg. Biochem. 2020, 205, 110999. [Google Scholar] [CrossRef] [PubMed]
  124. Kocaturk, N.M.; Akkoc, Y.; Kig, C.; Bayraktar, O.; Gozuacik, D.; Kutlu, O. Autophagy as a Molecular Target for Cancer Treatment. Eur. J. Pharm. Sci. 2019, 134, 116–137. [Google Scholar] [CrossRef]
  125. Lan, B.; Wan, Y.-J.; Pan, S.; Wang, Y.; Yang, Y.; Leng, Q.-L.; Jia, H.; Liu, Y.; Zhang, C.-Z.; Cao, Y. Parthenolide Induces Autophagy via the Depletion of 4E-BP1. Biochem. Biophys. Res. Commun. 2015, 456, 434–439. [Google Scholar] [CrossRef]
  126. Eramo, A.; Ricci-Vitiani, L.; Zeuner, A.; Pallini, R.; Lotti, F.; Sette, G.; Pilozzi, E.; Larocca, L.M.; Peschle, C.; De Maria, R. Chemotherapy Resistance of Glioblastoma Stem Cells. Cell Death Differ. 2006, 13, 1238–1241. [Google Scholar] [CrossRef] [Green Version]
  127. Tang, T.K.; Chiu, S.C.; Lin, C.W.; Su, M.J.; Liao, M.H. Induction of survivin inhibition, G2/M cell cycle arrest and autophagic on cell death in human malignant glioblastoma cells. Chin. J. Physiol. 2015, 58, 95–103. [Google Scholar] [CrossRef] [Green Version]
  128. Gào, X.; Schöttker, B. Reduction–Oxidation Pathways Involved in Cancer Development: A Systematic Review of Literature Reviews. Oncotarget 2017, 8, 51888–51906. [Google Scholar] [CrossRef] [Green Version]
  129. Forman, H.J.; Zhang, H. Targeting Oxidative Stress in Disease: Promise and Limitations of Antioxidant Therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef]
  130. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of Apoptosis Signalling Pathways by Reactive Oxygen Species. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2016, 1863, 2977–2992. [Google Scholar] [CrossRef]
  131. Yun, B.-R.; Lee, M.-J.; Kim, J.-H.; Kim, I.-H.; Yu, G.-R.; Kim, D.-G. Enhancement of Parthenolide-Induced Apoptosis by a PKC-Alpha Inhibition through Heme Oxygenase-1 Blockage in Cholangiocarcinoma Cells. Exp. Mol. Med. 2010, 42, 787–797. [Google Scholar] [CrossRef] [PubMed]
  132. Duan, D.; Zhang, J.; Yao, J.; Liu, Y.; Fang, J. Targeting Thioredoxin Reductase by Parthenolide Contributes to Inducing Apoptosis of HeLa Cells. J. Biol. Chem. 2016, 291, 10021–10031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wen, J.; You, K.-R.; Lee, S.-Y.; Song, C.-H.; Kim, D.-G. Oxidative Stress-Mediated Apoptosis: The anticancer effect of the sesquiterpene lactone parthenolide. J. Biol. Chem. 2002, 277, 38954–38964. [Google Scholar] [CrossRef] [Green Version]
  134. Emanuele, S.; D’Anneo, A.; Calvaruso, G.; Cernigliaro, C.; Giuliano, M.; Lauricella, M. The Double-Edged Sword Profile of Redox Signaling: Oxidative Events as Molecular Switches in the Balance between Cell Physiology and Cancer. Chem. Res. Toxicol. 2018, 31, 201–210. [Google Scholar] [CrossRef] [PubMed]
  135. Kim, S.-L.; Trang, K.T.T.; Kim, S.H.; Kim, I.H.; Lee, S.O.; Lee, S.T.; Kim, D.G.; Kim, S.-W. Parthenolide Suppresses Tumor Growth in a Xenograft Model of Colorectal Cancer Cells by Inducing Mitochondrial Dysfunction and Apoptosis. Int. J. Oncol. 2012, 41, 1547–1553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kim, J.-H.; Liu, L.; Lee, S.-O.; Kim, Y.-T.; You, K.-R.; Kim, D.-G. Susceptibility of Cholangiocarcinoma Cells to Parthenolide-Induced Apoptosis. Cancer Res. 2005, 65, 6312–6320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Wang, W.; Adachi, M.; Kawamura, R.; Sakamoto, H.; Hayashi, T.; Ishida, T.; Imai, K.; Shinomura, Y. Parthenolide-Induced Apoptosis in Multiple Myeloma Cells Involves Reactive Oxygen Species Generation and Cell Sensitivity Depends on Catalase Activity. Apoptosis 2006, 11, 2225–2235. [Google Scholar] [CrossRef]
  138. Flores-Lopez, G.; Moreno-Lorenzana, D.; Ayala-Sanchez, M.; Aviles-Vazquez, S.; Torres-Martinez, H.; Crooks, P.A.; Guzman, M.L.; Mayani, H.; Chávez-González, A. Parthenolide and DMAPT Induce Cell Death in Primitive CML Cells through Reactive Oxygen Species. J. Cell. Mol. Med. 2018, 22, 4899–4912. [Google Scholar] [CrossRef]
  139. Sun, Y.; Clair, D.K.S.; Xu, Y.; Crooks, P.A.; Clair, W.H.S. A NADPH Oxidase–Dependent Redox Signaling Pathway Mediates the Selective Radiosensitization Effect of Parthenolide in Prostate Cancer Cells. Cancer Res. 2010, 70, 2880–2890. [Google Scholar] [CrossRef] [Green Version]
  140. Carlisi, D.; D’Anneo, A.; Martinez, R.; Emanuele, S.; Buttitta, G.; Di Fiore, R.; Vento, R.; Tesoriere, G.; Lauricella, M. The Oxygen Radicals Involved in the Toxicity Induced by Parthenolide in MDA-MB-231 Cells. Oncol. Rep. 2014, 32, 167–172. [Google Scholar] [CrossRef] [Green Version]
  141. D’Anneo, A.; Carlisi, D.; Emanuele, S.; Buttitta, G.; Di Fiore, R.; Vento, R.; Tesoriere, G.; Lauricella, M. Parthenolide Induces Superoxide Anion Production by Stimulating EGF Receptor in MDA-MB-231 Breast Cancer Cells. Int. J. Oncol. 2013, 43, 1895–1900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Kurdi, M.; Bowers, M.C.; Dado, J.; Booz, G.W. Parthenolide Induces a Distinct Pattern of Oxidative Stress in Cardiac Myocytes. Free Radic. Biol. Med. 2007, 42, 474–481. [Google Scholar] [CrossRef] [PubMed]
  143. Dauer, P.; Sharma, N.S.; Gupta, V.K.; Nomura, A.; Dudeja, V.; Saluja, A.; Banerjee, S. GRP78-mediated Antioxidant Response and ABC Transporter Activity Confers Chemoresistance to Pancreatic Cancer Cells. Mol. Oncol. 2018, 12, 1498–1512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Chio, I.I.C.; Jafarnejad, S.M.; Ponz-Sarvise, M.; Park, Y.; Rivera, K.; Palm, W.; Wilson, J.; Sangar, V.; Hao, Y.; Öhlund, D.; et al. NRF2 Promotes Tumor Maintenance by Modulating MRNA Translation in Pancreatic Cancer. Cell 2016, 166, 963–976. [Google Scholar] [CrossRef] [Green Version]
  145. Emanuele, S.; Celesia, A.; D’Anneo, A.; Lauricella, M.; Carlisi, D.; De Blasio, A.; Giuliano, M. The Good and Bad of Nrf2: An Update in Cancer and New Perspectives in COVID-19. Int. J. Mol. Sci. 2021, 22, 7963. [Google Scholar] [CrossRef]
  146. Carlisi, D.; De Blasio, A.; Drago-Ferrante, R.; Di Fiore, R.; Buttitta, G.; Morreale, M.; Scerri, C.; Vento, R.; Tesoriere, G. Parthenolide Prevents Resistance of MDA-MB231 Cells to Doxorubicin and Mitoxantrone: The Role of Nrf2. Cell Death Discov. 2017, 3, 17078. [Google Scholar] [CrossRef] [Green Version]
  147. De Blasio, A.; Di Fiore, R.; Pratelli, G.; Drago-Ferrante, R.; Saliba, C.; Baldacchino, S.; Grech, G.; Scerri, C.; Vento, R.; Tesoriere, G. A Loop Involving NRF2, MiR-29b-1-5p and AKT, Regulates Cell Fate of MDA-MB-231 Triple-Negative Breast Cancer Cells. J. Cell. Physiol. 2020, 235, 629–637. [Google Scholar] [CrossRef]
  148. Drago-Ferrante, R.; Pentimalli, F.; Carlisi, D.; Blasio, A.D.; Saliba, C.; Baldacchino, S.; Degaetano, J.; Debono, J.; Caruana-Dingli, G.; Grech, G.; et al. Suppressive Role Exerted by MicroRNA-29b-1-5p in Triple Negative Breast Cancer through SPIN1 Regulation. Oncotarget 2017, 8, 28939–28958. [Google Scholar] [CrossRef] [Green Version]
  149. Pei, S.; Minhajuddin, M.; D’Alessandro, A.; Nemkov, T.; Stevens, B.M.; Adane, B.; Khan, N.; Hagen, F.K.; Yadav, V.K.; De, S.; et al. Rational Design of a Parthenolide-Based Drug Regimen That Selectively Eradicates Acute Myelogenous Leukemia Stem Cells. J. Biol. Chem. 2016, 291, 21984–22000. [Google Scholar] [CrossRef] [Green Version]
  150. Carlisi, D.; Buttitta, G.; Di Fiore, R.; Scerri, C.; Drago-Ferrante, R.; Vento, R.; Tesoriere, G. Parthenolide and DMAPT Exert Cytotoxic Effects on Breast Cancer Stem-like Cells by Inducing Oxidative Stress, Mitochondrial Dysfunction and Necrosis. Cell Death Dis. 2016, 7, e2194. [Google Scholar] [CrossRef] [Green Version]
  151. Mendonca, M.S.; Turchan, W.T.; Alpuche, M.E.; Watson, C.N.; Estabrook, N.C.; Chin-Sinex, H.; Shapiro, J.B.; Imasuen-Williams, I.E.; Rangel, G.; Gilley, D.P.; et al. DMAPT Inhibits NF-κB Activity and Increases Sensitivity of Prostate Cancer Cells to X-Rays In Vitro and in Tumor Xenografts In Vivo. Free Radic. Biol. Med. 2017, 112, 318–326. [Google Scholar] [CrossRef] [PubMed]
  152. Oka, D.; Nishimura, K.; Shiba, M.; Nakai, Y.; Arai, Y.; Nakayama, M.; Takayama, H.; Inoue, H.; Okuyama, A.; Nonomura, N. Sesquiterpene Lactone Parthenolide Suppresses Tumor Growth in a Xenograft Model of Renal Cell Carcinoma by Inhibiting the Activation of NF-κB. Int. J. Cancer 2007, 120, 2576–2581. [Google Scholar] [CrossRef] [PubMed]
  153. Sweeney, C.J.; Mehrotra, S.; Sadaria, M.R.; Kumar, S.; Shortle, N.H.; Roman, Y.; Sheridan, C.; Campbell, R.A.; Murry, D.J.; Badve, S.; et al. The Sesquiterpene Lactone Parthenolide in Combination with Docetaxel Reduces Metastasis and Improves Survival in a Xenograft Model of Breast Cancer. Mol. Cancer 2005, 4, 1004–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Curry, E.A.; Murry, D.J.; Yoder, C.; Fife, K.; Armstrong, V.; Nakshatri, H.; O’Connell, M.; Sweeney, C.J. Phase I Dose Escalation Trial of Feverfew with Standardized Doses of Parthenolide in Patients with Cancer. Investig. New Drugs 2004, 22, 299–305. [Google Scholar] [CrossRef]
  155. Wan Kamarul Zaman, W.S.; Nurul, A.A.; Nordin, F. Stem Cells and Cancer Stem Cells: The Jekyll and Hyde Scenario and Their Implications in Stem Cell Therapy. Biomedicines 2021, 9, 1245. [Google Scholar] [CrossRef]
  156. Walcher, L.; Kistenmacher, A.-K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauß, A.; Blaudszun, A.-R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer Stem Cells—Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020, 11, 1280. [Google Scholar] [CrossRef]
  157. Kuşoğlu, A.; Biray Avcı, Ç. Cancer Stem Cells: A Brief Review of the Current Status. Gene 2019, 681, 80–85. [Google Scholar] [CrossRef]
  158. Zhang, S.; Ju, X.; Yang, Q.; Zhu, Y.; Fan, D.; Su, G.; Kong, L.; Li, Y. USP47 maintains the stemness of colorectal cancer cells and is inhibited by parthenolide. Biochem. Biophys. Res. Commun. 2021, 562, 21–28. [Google Scholar] [CrossRef]
  159. Dandawate, P.R.; Subramaniam, D.; Jensen, R.A.; Anant, S. Targeting Cancer Stem Cells and Signaling Pathways by Phytochemicals: Novel Approach for Breast Cancer Therapy. Semin. Cancer Biol. 2016, 40–41, 192–208. [Google Scholar] [CrossRef] [Green Version]
  160. Nguyen, N.P.; Almeida, F.S.; Chi, A.; Nguyen, L.M.; Cohen, D.; Karlsson, U.; Vinh-Hung, V. Molecular biology of breast cancer stem cells: Potential clinical applications. Cancer Treat. Rev. 2010, 36, 485–491. [Google Scholar] [CrossRef]
  161. Yi, J.; Wang, L.; Wang, X.-Y.; Sun, J.; Yin, X.-Y.; Hou, J.-X.; Chen, J.; Xie, B.; Wei, H.-L. Suppression Of Aberrant Activation Of NF-κB Pathway in Drug-Resistant Leukemia Stem Cells Contributes To Parthenolide-Potentiated Reversal Of Drug Resistance in Leukemia. J. Cancer 2021, 12, 5519–5529. [Google Scholar] [CrossRef] [PubMed]
  162. Gunn, E.J.; Williams, J.T.; Huynh, D.T.; Iannotti, M.J.; Han, C.; Barrios, F.J.; Kendall, S.; Glackin, C.A.; Colby, D.A.; Kirshner, J. The Natural Products Parthenolide and Andrographolide Exhibit Anti-Cancer Stem Cell Activity in Multiple Myeloma. Leuk. Lymphoma 2011, 52, 1085–1097. [Google Scholar] [CrossRef] [PubMed]
  163. Zhou, J.; Zhang, H.; Gu, P.; Bai, J.; Margolick, J.B.; Zhang, Y. NF-κB Pathway Inhibitors Preferentially Inhibit Breast Cancer Stem-like Cells. Breast Cancer Res. Treat 2008, 111, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Liu, Y.; Lu, W.-L.; Guo, J.; Du, J.; Li, T.; Wu, J.-W.; Wang, G.-L.; Wang, J.-C.; Zhang, X.; Zhang, Q. A Potential Target Associated with Both Cancer and Cancer Stem Cells: A Combination Therapy for Eradication of Breast Cancer Using Vinorelbine Stealthy Liposomes plus Parthenolide Stealthy Liposomes. J. Control. Release 2008, 129, 18–25. [Google Scholar] [CrossRef] [PubMed]
  165. Abdullah, L.N.; Chow, E.K.-H. Mechanisms of Chemoresistance in Cancer Stem Cells. Clin. Transl. Med. 2013, 2, 3. [Google Scholar] [CrossRef] [Green Version]
  166. Liao, K.; Xia, B.; Zhuang, Q.-Y.; Hou, M.-J.; Zhang, Y.-J.; Luo, B.; Qiu, Y.; Gao, Y.-F.; Li, X.-J.; Chen, H.-F.; et al. Parthenolide Inhibits Cancer Stem-Like Side Population of Nasopharyngeal Carcinoma Cells via Suppression of the NF-κB/COX-2 Pathway. Theranostics 2015, 5, 302–321. [Google Scholar] [CrossRef] [Green Version]
  167. Zhang, D.; Qiu, L.; Jin, X.; Guo, Z.; Guo, C. Nuclear Factor-ΚB Inhibition by Parthenolide Potentiates the Efficacy of Taxol in Non–Small Cell Lung Cancer In Vitro and In Vivo. Mol. Cancer Res. 2009, 7, 1139–1149. [Google Scholar] [CrossRef] [Green Version]
  168. Shanmugam, R.; Jayaprakasan, V.; Gokmen-Polar, Y.; Kelich, S.; Miller, K.D.; Yip-Schneider, M.; Cheng, L.; Bhat-Nakshatri, P.; Sledge, G.W., Jr.; Nakshatri, H.; et al. Restoring Chemotherapy and Hormone Therapy Sensitivity by Parthenolide in a Xenograft Hormone Refractory Prostate Cancer Model. Prostate 2006, 66, 1498–1511. [Google Scholar] [CrossRef]
  169. deGraffenried, L.A.; Chandrasekar, B.; Friedrichs, W.E.; Donzis, E.; Silva, J.; Hidalgo, M.; Freeman, J.W.; Weiss, G.R. NF-κB Inhibition Markedly Enhances Sensitivity of Resistant Breast Cancer Tumor Cells to Tamoxifen. Ann. Oncol. 2004, 15, 885–890. [Google Scholar] [CrossRef]
  170. Riggins, R.B.; Zwart, A.; Nehra, R.; Clarke, R. The Nuclear Factor ΚB Inhibitor Parthenolide Restores ICI 182,780 (Faslodex; Fulvestrant)–Induced Apoptosis in Antiestrogen-Resistant Breast Cancer Cells. Mol. Cancer 2005, 4, 33–41. [Google Scholar]
  171. Carlisi, D.; Lauricella, M.; D’Anneo, A.; Buttitta, G.; Emanuele, S.; di Fiore, R.; Martinez, R.; Rolfo, C.; Vento, R.; Tesoriere, G. The Synergistic Effect of SAHA and Parthenolide in MDA-MB231 Breast Cancer Cells. J. Cell. Physiol. 2015, 230, 1276–1289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Fang, L.-J.; Shao, X.-T.; Wang, S.; Lu, G.-H.; Xu, T.; Zhou, J.-Y. Sesquiterpene Lactone Parthenolide Markedly Enhances Sensitivity of Human A549 Cells to Low-Dose Oxaliplatin via Inhibition of NF-κB Activation and Induction of Apoptosis. Planta Med. 2010, 76, 258–264. [Google Scholar] [CrossRef] [PubMed]
  173. Francescato, H.D.C.; Costa, R.S.; Scavone, C.; Coimbra, T.M. Parthenolide Reduces Cisplatin-Induced Renal Damage. Toxicology 2007, 230, 64–75. [Google Scholar] [CrossRef]
  174. Ghorbani-Abdi-Saedabad, A.; Hanafi-Bojd, M.Y.; Parsamanesh, N.; Tayarani-Najaran, Z.; Mollaei, H.; Hoshyar, R. Anticancer and Apoptotic Activities of Parthenolide in Combination with Epirubicin in Mda-Mb-468 Breast Cancer Cells. Mol. Biol Rep. 2020, 47, 5807–5815. [Google Scholar] [CrossRef]
  175. Ralstin, M.C.; Gage, E.A.; Yip-Schneider, M.T.; Klein, P.J.; Wiebke, E.A.; Schmidt, C.M. Parthenolide Cooperates with NS398 to Inhibit Growth of Human Hepatocellular Carcinoma Cells through Effects on Apoptosis and G0-G1 Cell Cycle Arrest. Mol. Cancer Res. 2006, 4, 387–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Yip-Schneider, M.T.; Wu, H.; Ralstin, M.; Yiannoutsos, C.; Crooks, P.A.; Neelakantan, S.; Noble, S.; Nakshatri, H.; Sweeney, C.J.; Schmidt, C.M. Suppression of Pancreatic Tumor Growth by Combination Chemotherapy with Sulindac and LC-1 Is Associated with Cyclin D1 Inhibition in Vivo. Mol. Cancer 2007, 6, 1736–1744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Taguchi, T.; Takao, T.; Iwasaki, Y.; Nishiyama, M.; Asaba, K.; Hashimoto, K. Suppressive Effects of Dehydroepiandrosterone and the Nuclear Factor-ΚB Inhibitor Parthenolide on Corticotroph Tumor Cell Growth and Function In Vitro and In Vivo. J. Endocrinol. 2006, 188, 321–331. [Google Scholar] [CrossRef] [Green Version]
  178. Duechler, M.; Stańczyk, M.; Czyż, M.; Stępnik, M. Potentiation of Arsenic Trioxide Cytotoxicity by Parthenolide and Buthionine Sulfoximine in Murine and Human Leukemic Cells. Cancer Chemother. Pharm. 2008, 61, 727–737. [Google Scholar] [CrossRef]
  179. Watson, C.; Miller, D.A.; Chin-Sinex, H.; Losch, A.; Hughes, W.; Sweeney, C.; Mendonca, M.S. Suppression of NF-κB Activity by Parthenolide Induces X-Ray Sensitivity through Inhibition of Split-Dose Repair in TP53 Null Prostate Cancer Cells. Radiat. Res. 2009, 171, 389–396. [Google Scholar] [CrossRef]
  180. Hayashi, S.; Hatashita, M.; Hayashi, A.; Matsumoto, H.; Shioura, H.; Kitai, R. Thermosensitization by Parthenolide in Human Lung Adenocarcinoma A549 Cells and P53- and Hsp72-Independent Apoptosis Induction via the Nuclear Factor-ΚB Signal Pathway. Int. J. Mol. Med. 2008, 21, 585–592. [Google Scholar] [CrossRef]
  181. Hayashi, S.; Sakurai, H.; Hayashi, A.; Tanaka, Y.; Hatashita, M.; Shioura, H. Inhibition of NF-κB by Combination Therapy with Parthenolide and Hyperthermia and Kinetics of Apoptosis Induction and Cell Cycle Arrest in Human Lung Adenocarcinoma Cells. Int. J. Mol. Med. 2010, 25, 81–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Jia, X.; Liu, Q.; Wang, S.; Zeng, B.; Du, G.; Zhang, C.; Li, Y. Synthesis, Cytotoxicity, and in Vivo Antitumor Activity Study of Parthenolide Semicarbazones and Thiosemicarbazones. Bioorg. Med. Chem. 2020, 28, 115557. [Google Scholar] [CrossRef] [PubMed]
  183. Karmakar, A.; Xu, Y.; Mustafa, T.; Kannarpady, G.; Bratton, S.M.; Radominska-Pandya, A.; Crooks, P.A.; Biris, A.S. Nanodelivery of Parthenolide Using Functionalized Nanographene Enhances Its Anticancer Activity. RSC Adv. 2015, 5, 2411–2420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Darwish, N.H.E.; Sudha, T.; Godugu, K.; Bharali, D.J.; Elbaz, O.; El-ghaffar, H.A.A.; Azmy, E.; Anber, N.; Mousa, S.A. Novel Targeted Nano-Parthenolide Molecule against NF-κB in Acute Myeloid Leukemia. Molecules 2019, 24, 2103. [Google Scholar] [CrossRef] [Green Version]
  185. Zhai, J.-D.; Li, D.; Long, J.; Zhang, H.-L.; Lin, J.-P.; Qiu, C.-J.; Zhang, Q.; Chen, Y. Biomimetic Semisynthesis of Arglabin from Parthenolide. J. Org. Chem. 2012, 77, 7103–7107. [Google Scholar] [CrossRef]
  186. Lone, S.H.; Bhat, K.A.; Khuroo, M.A. Arglabin: From Isolation to Antitumor Evaluation. Chem.-Biol. Interact. 2015, 240, 180–198. [Google Scholar] [CrossRef]
  187. Zhang, Q.; Lu, Y.; Ding, Y.; Zhai, J.; Ji, Q.; Ma, W.; Yang, M.; Fan, H.; Long, J.; Tong, Z.; et al. Guaianolide Sesquiterpene Lactones, a Source to Discover Agents That Selectively Inhibit Acute Myelogenous Leukemia Stem and Progenitor Cells. J. Med. Chem. 2012, 55, 8757–8769. [Google Scholar] [CrossRef]
  188. He, W.; Lai, R.; Lin, Q.; Huang, Y.; Wang, L. Arglabin is a plant sesquiterpene lactone that exerts potent anticancer effects on human oral squamous cancer cells via mitochondrial apoptosis and downregulation of the mTOR/PI3K/Akt signaling pathway to inhibit tumor growth in vivo. J. Buon 2018, 23, 1679–1685. [Google Scholar]
  189. Yu, L.; Chen, W.; Tang, Q.; Ji, K.-Y. Micheliolide Inhibits Liver Cancer Cell Growth Via Inducing Apoptosis And Perturbing Actin Cytoskeleton. Cancer Manag. Res. 2019, 11, 9203–9212. [Google Scholar] [CrossRef] [Green Version]
  190. Viennois, E.; Xiao, B.; Ayyadurai, S.; Wang, L.; Wang, P.G.; Zhang, Q.; Chen, Y.; Merlin, D. Micheliolide, a New Sesquiterpene Lactone That Inhibits Intestinal Inflammation and Colitis-Associated Cancer. Lab. Investig. 2014, 94, 950–965. [Google Scholar] [CrossRef] [Green Version]
  191. Zhong, J.; Gong, W.; Chen, J.; Qing, Y.; Wu, S.; Li, H.; Huang, C.; Chen, Y.; Wang, Y.; Xu, Z.; et al. Micheliolide Alleviates Hepatic Steatosis in Db/Db Mice by Inhibiting Inflammation and Promoting Autophagy via PPAR-γ-Mediated NF-κB and AMPK/MTOR Signaling. Int. Immunopharmacol. 2018, 59, 197–208. [Google Scholar] [CrossRef] [PubMed]
  192. Tang, X.; Ding, Q.; Chen, C.; Chen, F.; Zhou, X.; Hong, C.J.; Pan, W. Micheliolide Inhibits Gastric Cancer Growth in Vitro and in Vivo via Blockade of the IL-6/STAT3 Pathway. Die Pharm. Int. J. Pharm. Sci. 2019, 74, 175–178. [Google Scholar] [CrossRef]
  193. Li, J.; Li, S.; Guo, J.; Li, Q.; Long, J.; Ma, C.; Ding, Y.; Yan, C.; Li, L.; Wu, Z.; et al. Natural Product Micheliolide (MCL) Irreversibly Activates Pyruvate Kinase M2 and Suppresses Leukemia. J. Med. Chem. 2018, 61, 4155–4164. [Google Scholar] [CrossRef] [PubMed]
  194. An, Y.; Guo, W.; Li, L.; Xu, C.; Yang, D.; Wang, S.; Lu, Y.; Zhang, Q.; Zhai, J.; Fan, H.; et al. Micheliolide Derivative DMAMCL Inhibits Glioma Cell Growth In Vitro and In Vivo. PLoS ONE 2015, 10, e0116202. [Google Scholar] [CrossRef] [Green Version]
  195. Liu, Y.; Wang, L.; Liu, J.; Xie, X.; Hu, H.; Luo, F. Anticancer Effects of ACT001 via NF-κB Suppression in Murine Triple-Negative Breast Cancer Cell Line 4T1. Cancer Manag. Res. 2020, 12, 5131–5139. [Google Scholar] [CrossRef]
  196. Jin, X.-H.; Jia, Y.-S.; Shi, Y.-H.; Li, Q.-Y.; Bao, S.-Q.; Lu, W.-P.; Tong, Z.-S. ACT001 Can Prevent and Reverse Tamoxifen Resistance in Human Breast Cancer Cell Lines by Inhibiting NF-κB Activation. J. Cell. Biochem. 2019, 120, 1386–1397. [Google Scholar] [CrossRef]
  197. Hou, Y.; Sun, B.; Liu, W.; Yu, B.; Shi, Q.; Luo, F.; Bai, Y.; Feng, H. Targeting of Glioma Stem-like Cells with a Parthenolide Derivative ACT001 through Inhibition of AEBP1/PI3K/AKT Signaling. Theranostics 2021, 11, 555–566. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of (A) parthenolide (PN) and its analogue (B) dimethylaminoparthenolide (DMAPT).
Figure 1. Chemical structures of (A) parthenolide (PN) and its analogue (B) dimethylaminoparthenolide (DMAPT).
Biomedicines 10 00514 g001
Figure 2. Effects of PN on prosurvival/oncogenic transcriptional factors and relative signaling: (A) NF-κB; (B) JAK/STAT; (C) β-catenin.
Figure 2. Effects of PN on prosurvival/oncogenic transcriptional factors and relative signaling: (A) NF-κB; (B) JAK/STAT; (C) β-catenin.
Biomedicines 10 00514 g002
Figure 3. The effects of PN on different signaling cascades.
Figure 3. The effects of PN on different signaling cascades.
Biomedicines 10 00514 g003
Figure 4. Different types of cell death induced by PN treatment.
Figure 4. Different types of cell death induced by PN treatment.
Biomedicines 10 00514 g004
Figure 5. Mechanisms of oxidative stress induction by PN.
Figure 5. Mechanisms of oxidative stress induction by PN.
Biomedicines 10 00514 g005
Figure 6. Chemical structures of (A) arglabin, (B) micheliolide (MCL) and its analogue (C) ACT001.
Figure 6. Chemical structures of (A) arglabin, (B) micheliolide (MCL) and its analogue (C) ACT001.
Biomedicines 10 00514 g006
Table 1. Main classes of compounds that have a synergistic action in association with PN.
Table 1. Main classes of compounds that have a synergistic action in association with PN.
Synergistic Action of PN
CoumpoundsClass of CompoundTested onRef.
Taxol, Docetaxel, VinorelbineAntimicrotubule agentsLung cancer, prostate cancer, breast cancer cell lines[164,167,168]
Tamoxifen, FulvestrantAntiestrogen agentsBreast cancer cell lines[169,170]
SAHAHistone deacetylase inhibitorBreast cancer cell lines[171]
OxaliplatinAntineoplastic platinum drugsLung cancer cell lines[172]
EpirubicinAnthracycline doxorubicin analogBreast cancer cell lines[174]
NS398, sulindacCOX-2 inhibitorsHepatocellular carcinoma cell lines[175,176]
DehydroepiandrosteroneSteroid anti-inflammatory agentsPituitary tumor cell lines[177]
Retinoic acidActive metabolite of vitamin ALeukaemia cell lines[93]
Arsenic trioxideToxic metalloidLeukaemia cell lines[178]
TRAILTumor necrosis factor familyHepatocellular carcinoma and breast cancer cell lines[50,65,179]
RadiotherapyX-rayLung adenocarcinoma, prostate cancer cell lines[50,179,180,181]
Table 2. Main compounds with structural and functional analogy to PN.
Table 2. Main compounds with structural and functional analogy to PN.
PN Anologues
CoumpoundsStructureTested onRef.
Dimethylaminoparthenolide (DMAPT), also known as LC-1PN with a methyl group also in the form of fumarate saltLeukemia, prostate cancer, breast cancer[26,27,116]
PN semicarbazone or thiosemicarbazonePN with semicarbazone/thiosemi-carbazone groupsColorectal carcinoma, glioblastoma, liver carcinoma, gastric cancer and lung cancer cell lines[182]
PN-fGn or DMAPT-fGnCarboxyl-functionalized nanographenePancreatic cancer cell lines[183]
PLGA-anti CD44-PN nanoparticlesPolylactide coglycolide (PLGA) nanoparticles conjugated with antiCD44Acute myeloid leukemia[184]
ArglabinGuaianolide sesquiterpene lactoneLeukemia, human oral squamous and lung cancer cell lines[185,186,187,188]
Micheliolide (MCL)Guaianolide sesquiterpene lactoneHepatocellular carcinoma, leukemia cells[185,189,190,191,192,193]
ACT001Fumarate salt of dimethylaminomicheliolideBreast cancer cells, glioma stem cells[194,195,196,197]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Carlisi, D.; Lauricella, M.; D’Anneo, A.; De Blasio, A.; Celesia, A.; Pratelli, G.; Notaro, A.; Calvaruso, G.; Giuliano, M.; Emanuele, S. Parthenolide and Its Soluble Analogues: Multitasking Compounds with Antitumor Properties. Biomedicines 2022, 10, 514. https://doi.org/10.3390/biomedicines10020514

AMA Style

Carlisi D, Lauricella M, D’Anneo A, De Blasio A, Celesia A, Pratelli G, Notaro A, Calvaruso G, Giuliano M, Emanuele S. Parthenolide and Its Soluble Analogues: Multitasking Compounds with Antitumor Properties. Biomedicines. 2022; 10(2):514. https://doi.org/10.3390/biomedicines10020514

Chicago/Turabian Style

Carlisi, Daniela, Marianna Lauricella, Antonella D’Anneo, Anna De Blasio, Adriana Celesia, Giovanni Pratelli, Antonietta Notaro, Giuseppe Calvaruso, Michela Giuliano, and Sonia Emanuele. 2022. "Parthenolide and Its Soluble Analogues: Multitasking Compounds with Antitumor Properties" Biomedicines 10, no. 2: 514. https://doi.org/10.3390/biomedicines10020514

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