Oncotarget

Reviews:

The importance of integrated therapies on cancer: Silibinin, an old and new molecule

PDF |  Full Text  |  How to cite  |  Press Release

Oncotarget. 2024; 15:345-353. https://doi.org/10.18632/oncotarget.28587

Metrics: PDF 230 views  |   Full Text 1286 views  |   ?  

Elisa Roca _, Giuseppe Colloca, Fiorella Lombardo, Andrea Bellieni, Alessandra Cucinella, Giorgio Madonia, Licia Martinelli, Maria Elisa Damiani, Ilaria Zampieri and Antonio Santo

Abstract

Elisa Roca1, Giuseppe Colloca2, Fiorella Lombardo1, Andrea Bellieni2, Alessandra Cucinella1, Giorgio Madonia1, Licia Martinelli1, Maria Elisa Damiani1, Ilaria Zampieri1 and Antonio Santo1

1 Oncologia Toracica - Lung Unit, Ospedale P. Pederzoli - Via Monte Baldo, Peschiera del Garda (VR), Italy

2 Dipartimento di Scienze dell’invecchiamento, Neurologiche, Ortopediche e della testa-collo, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy

Correspondence to:

Elisa Roca, email: [email protected],
ORCID: orcid.org/0000-0001-7625-7708

Keywords: silibinin; anti-inflammatory; inflammation; toxicity; integrated therapy

Received: March 19, 2024     Accepted: May 06, 2024     Published: May 23, 2024

Copyright: © 2024 Roca et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

ABSTRACT

In the landscape of cancer treatments, the efficacy of coadjuvant molecules remains a focus of attention for clinical research with the aim of reducing toxicity and achieving better outcomes.

Most of the pathogenetic processes causing tumour development, neoplastic progression, ageing, and increased toxicity involve inflammation. Inflammatory mechanisms can progress through a variety of molecular patterns. As is well known, the ageing process is determined by pathological pathways very similar and often parallel to those that cause cancer development. Among these complex mechanisms, inflammation is currently much studied and is often referred to in the geriatric field as ‘inflammaging’.

In this context, treatments active in the management of inflammatory mechanisms could play a role as adjuvants to standard therapies.

Among these emerging molecules, Silibinin has demonstrated its anti-inflammatory properties in different neoplastic types, also in combination with chemotherapeutic agents.

Moreover, this molecule could represent a breakthrough in the management of age-related processes.

Thus, Silibinin could be a valuable adjuvant to reduce drug-related toxicity and increase therapeutic potential.

For this reason, the main aim of this review is to collect and analyse data presented in the literature on the use of Silibinin, to better understand the mechanisms of the functioning of this molecule and its possible therapeutic role.


Introduction

Inflammation underlies many disease processes, and among these, ageing and the development of neoplasms would seem to have similarities and parallels underlying an important inflammatory substrate.

Silibinin was isolated from the seeds of milk thistle (Silybum marianum) and it a polyphenolic flavonolignan [1] used to date as a treatment for hepatopathies and for different exchange mechanisms; it is also a molecule with inflammatory, antioxidant and anti-fibrotic properties [1] (Figure 1).

Figure 1: Molecular structure of Silibinin.

Furthermore, there are promising data on the application of Silibinin as an anti-cancer coadjuvant in various types of neoplasia [2].

In this regard, the antitumour characteristics of Silibinin have been documented both in vivo and in vitro.

In particular, this molecule seems to play a role in inhibiting proliferation, blocking metastasis and tumour invasion, and inhibiting angiogenesis.

Furthermore, Silibinin could be useful as an inducer of programmed death (apoptosis) and autophagy.

Finally, studies show a role for Silibinin in cell cycle arrest and in the inhibition of certain signalling pathways such as MAPK, STAT3, Notch-1, ERK and Akt.

For this reason, Silibinin could be used in combination with antiblastic drugs to achieve a synergistic effect and improve the outcomes of neoplastic patients.

Since several studies show that Silibinin is implicated at some critical points in the processes of ageing and ‘inflammation’, this treatment could counteract ‘inflammation’ mechanisms above all in older adults [2].

ROLE OF SILIBININ IN DIFFERENT CANCER’S TYPE

Studies investigating the efficacy of Silibinin as an anticancer agent are promising [24]; in particular, the most interesting data concern its effectiveness as an adjuvant in the treatment of gliomas.

Gliomas are the most common brain tumors, which maintain a poor prognosis, despite the aggressive therapeutic strategies (surgery, radiotherapy, chemotherapy) that are implemented to increase the survival of patients affected by this neoplasm [5].

To reduce the side effects of these treatments and to increase their effectiveness, therapeutic strategies also based on natural compounds are currently being taken into consideration [6].

In this context, research regarding Silibinin has shown interesting results regarding its ability to inhibit cell proliferation [7] and its role in sensitizing glioma cells to apoptosis [8].

The promising results of these studies suggest that Silibinin could inhibit cell proliferation by increasing the intracellular Ca2+ concentration. This increase in intracellular Ca2+ appears to have a critical role in regulating cell death mediated by a calpain-dependent pathway and in inducing the generation of ROS. Several studies also show that Silibinin is able to generate ROS through a CA2+ -dependent mechanism by the respiratory chain in the inner mitochondrial membrane [9] or through direct stimulation of the generation of mitochondrial ROS [10].

Furthermore, this milk thistle-derived molecule can induce programmed cell death by activating mitogen-activated protein kinases (MAPKs) through the generation of ROS.

Moreover, Silibinin is able to activate (via ROS) and inhibit (via antioxidant N-acetylcysteine (NAC)) the extracellular signal-regulated kinase (ERK), the p38 kinase and the N-terminal c-Jun kinase (JNK) in time mode [11]. This suggests that MAPK activation is likely involved in Silibinin-induced cell death.

Another mechanism believed to be secondary to the action of Silibinin is the inhibition of glioma cell migration.

Regarding in vivo antineoplastic effects, some authors have reported data on Silibinin administered orally in mice with gliomas: these data demonstrated a decrease in glioma growth in vivo, thanks to the administration of this molecule.

In particular, the regression of tumor volume could be induced by Silibinin, leading cells to programmed death through a caspase-dependent mechanism, involving the Ca2+/ROS/MAPK pathways, [5, 12].

In addition, Silibinin appears to play a role in cell migration through mitochondrial fusion and inhibition of ROS [13].

Some studies have also shown the ability of Silibinin to promote the epithelial-mesenchymal transition: this transformation could be due to the decrease in the expression of proteins linked to migration (for example metalloproteinases 2 and 9) or to the increase in the expression of other biomarkers, including E-cadherin.

The discovery of the epidermal growth factor receptor (EGFR) and the possibility of using it as a therapeutic target has changed the natural history of many patients suffering from cancer.

In the context of gliomas, rat cell lines expressing EGFR were tested to evaluate the ability of Silibinin to inhibit mitogenic signaling, regulate cell survival and alter the cell cycle [14, 15]. The researchers noted that EGRF-positive cells demonstrated cytotoxicity in response to Silibinin, on the other hand, a lack of toxicity was observed in cell lines without EGFR sequences. Moreover Silibinin seems to be able to inhibit the binding of EGF to the EGFR resulting in cytotoxicity and with unknown effects on the dimerization of the receptor.

SILIBININ AS STAT3 INHIBITOR

Silibinin has been shown to be both in vitro and in vivo, a physiological down-regulator of STAT3, a protein which is part of “signal transducer and activator of transcription” family (STAT) activity [16]. This molecule appears to have a role in reducing the incidence of main chemotherapy-related toxicities, such as nephrotoxicity, neurotoxicity and cardiotoxicity in pre-clinical models, as well as in preventing drug resistance mechanisms.

Members of STAT family share a similar functional structure: they are cytoplasmic proteins, containing Src homology 2 (SH2) domains. Being responsive to different levels of cell growth factors and cytokines they work as transcriptional factors [17]. Currently STAT3 is the most known member of STAT family, since several studies have shown that altered regulation of this protein may underlie loss of cell cycle control, uncontrolled cell survival, carcinogenesis, and metastatic dissemination and immunoresistance processes. Accordingly, this protein has been largely considered as a potential therapeutic target in different tumor types, leading to the development of a new class of antineoplastic drugs, STAT3 inhibitors, classified as indirect or direct based on their mechanism of action [17, 18].

Given its modulatory function on STAT3, Silibinin is under evaluation in the oncological research field both as monotherapy and in combination with other available therapeutic regimens [19].

BRAIN METASTASIS GROWTH

Among intracranial tumors, metastases are the most common form of central nervous system (CNS) involvement in adults (more than 50%). The process of brain metastasis appears to be the most challenging for tumor cells, due to the presence of the blood-brain barrier (BBB) and the distinctive microenvironment [20]. The metastatic potential of cancer cells increase as they acquire the ability to escape natural defense mechanisms, interact with various cell types in different cellular microenvironments, affecting their activity in a pro-metastatic way, and lastly promoting their own survival [21, 22]. Within this specific microenvironment, astrocytes and microglia cells seem to play a critical role in the process of promoting brain metastatization. In fact, astrocytes are CNS cells able to react to tissue damage by the activation of a morphological and transcriptomic reactive state (Reactive astrocytes- Ras), which has been demonstrated to affect the course of several neurological diseases including secondary SNC lesions [23]. This reactive state also appears to be mediated by STAT3 activation, resulting in a pro-metastatic environment [24, 25]. Furthermore, STAT3 hyperactivation in reactive astrocytic phenotype (RAs) could act negatively on the activation of both innate and acquired immune response and in particular on CD8+ lymphocytes, of which also brain metastases are often infiltrated. Activated STAT3 RAs cells could therefore act by reducing the tumor infiltrate and creating an immunosuppressive microenvironment [26].

The importance of STAT3 activation for CNS colonization, microenvironment changes, and the potential utility of administering anti STAT 3 drugs also in combination with Immune Checkpoint Inhibitors (ICIs) is supported by the fact that Silibinin is able to cross the BBB and reduce STAT3 expression in the tumor microenvironment. It was used in an RCT test to test the overall survival from brain metastasis diagnosis compared to a control cohort.

COADJUVANT MECHANISM OF SILIBININ

Protector on chemotherapy-induced toxicity

Most antiblastic drugs are metabolized/excreted by the kidney, potentially resulting in dose-related renal toxicity, as acute or chronic kidney injury and electrolyte disorders. Sometimes renal damage may be irreversible [2, 27, 28]. Several studies have demonstrated a protective role of Silibinin toward renal function when administered together with nephrotoxic drugs such as cisplatin and vinorelbine [29] (Table 1).

Table 1: Fields of application and mechanisms of Silibinin

CANCER MECHANISMS

⇓ Ca2+/ROS/MAPK

⇓ STAT3

⇑ Sensibility to apoptosis

⇓ Tumor cell proliferation

⇓ Metastasis growth

⇓ Resistance to chemo/radio/target therapies

NEUROPATHY (CIPN)

⇓ Microtubule disruption

⇓ Oxidative stress

⇓ Mitochondrial damage

⇓ Altered ion channel activity

⇓ DNA Damage

⇓ Immunological processes

INFLAMMATION

⇓ IL-6

⇓ ROS

AGING

⇓ Oxidative stress

⇓ Mitochondrial Dysfunction

⇓ Shortened Telomeres

⇓ DNA Damage

⇓ Cell Senescence

Silibinin in reversing resistance to chemotherapy

STAT pathway activation has been associated with primary chemoresistance and the development of secondary chemoresistance [30]. Based on this evidence, it is expected that inactivation of STAT3 may play a synergistic role by potentiating the effect of chemotherapeutic drugs [31].

Among these, the greatest evidence in terms of synergistic efficacy was obtained combining Silibinin with paclitaxel: known as a microtubule stabilizer, paclitaxel has been shown to reduce STAT3 protein activity (Tyr705) and to inhibit the expression of STAT3-target genes via a negative feedback loop [32]. Further evidences have been obtained combining Silibinin with Arsenic Trioxide (ATO) in human glioblastoma cell line, in which a prevalence of apoptotic phenomena and reduced invasive capacity were observed [33].

Silibinin in reversing resistance to targeted therapies

The activation of STAT3 is among the pathways proposed to be involved in the development of drug resistance in oncogene-addicted tumor cells, although the specific mechanism is still unclear: STAT3 could be activated to prevent apoptosis and sustain cell viability but it may also be activated early in a cancer-cell sub-population, maybe immediately after drug exposure [34].

In this setting, elevated levels of pSTAT3 have been associated to poor prognosis and lower overall survival in patients affected by hepatocellular carcinoma, where this molecule plays a pivotal role in inflammation, and in sustaining survival, proliferation, and invasion capability of malignant cells [35]. Numerous studies have also demonstrated the essential role of STAT3 in the development of drug resistance toward epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs): high levels of STAT3 have been recently reported to predict worse progression-free survival.in patients affected by Non-Small Cell Lung Cancer (NSCLC) harboring EGFR activating mutations treated with this class of drugs [36, 37]. Interestingly, Silibinin have been shown to be able to overcome such resistance towards two different TKI, erlotinib and gefitinib, both in vivo and in vitro, although the ability of Silibinin to revert such resistance acting through the pSTAT pathtway inhibition is yet to be confirmed [3840].

Silibinin in reversing resistance to radiotherapy

Radio-resistance is one of the main limitations to the efficacy of radiation therapy and may be induced by the use of fractionated radiation. At a cellular level, this has been associated to EMT: cancer cells surviving to ionizing radiations often feature EMT-like phenotype, that appears to be essential for the development of radio-resistance [41]. Radiation-resistant cancer cells have been found to feature high levels of nuclear expression of STAT3 [42], and some studies suggested that the inhibition of this pathway can reverse the EMT process, thus overcoming radio resistance [4345]. Based on these data, the combination of ionizing radiation with Silibilin may represent a possible strategy to overcome radio resistance by inhibiting STAT3 signaling and therefore EM transition [46].

INFLAMMAGING

The process of aging is the combination of various factors, involving a variety of environmental, stochastic, and genetic-epigenetic events. One of the pivotal mechanisms involved is inflammation, that usually contributes to aging process through the development of the so-called “inflammaging”: a chronic, low-grade inflammation status [47, 48].

Various organs and tissues, including bone and muscle and adipose tissue, are able to produce pro-inflammatory compounds, and, as such, inflammaging appears to be a systemic, multifactor and multiorgan condition, determined by a complex balance between pro- and anti-inflammatory factors. It can be considered among the main factors in the pathogenesis of various age-related diseases, such as atherosclerosis, type II diabetes, as well as of many chronic conditions including sarcopenia [49], osteoporosis [50], frailty and disability [51]. Overall, this inflammatory process can contribute to increase the risk of multimorbidity and disease susceptibility, and to impair the ability to appropriately respond to stress as well as to treatments, ultimately favoring the development of geriatric syndromes, disabilities, and death.

Multiple processes, both at a cellular and molecular level, have been taken in account to explain inflammaging, among them inflammasome activation, dysbiosis, cellular senescence, mitochondrial dysfunction, defective autophagy and mitophagy, ubiquitin-proteasome system dysregulation, activation of the DNA damage response [52], and chronic activation of the innate immune system [48].

Multiple epidemiological and biodemographic studies have shown that inflammation biomarkers can be robust predictors of morbidity and mortality in the elderly [53, 54]. Among the multiple factors involved, interleukin-6 (IL-6), with its complex pro-and anti-inflammatory functions, deserves a primary role, and has been associated to multiple conditions and age-related disease as well as to mortality in the elderly [55].

Some authors have also suggested that inflammaging could be considered in some way a form of autoimmune condition in which the distinction between self and non-self structures get increasingly blurred with age, and that this process could be caused by the malfunction of some of the various mechanisms appointed to dispose of senescent or apoptotic cells [51]. Indeed, the process may be triggered by the oxidative stress caused by the accumulation of free radical within a cell can activate a network of distress sensors (Nlrp3 inflammasome) thus activating the immune response and the production of pro-inflammatory cytokines such as IL-1beta and IL-8. Hence, Inflammaging may be triggered by the slow accumulation with age of damaged macromolecules and cells, that lead with time to chronic stress.

The same aging process that affects the human body’s microbial constituents (i.e., gut microbiota), altering their physiological functions (i.e. elimination of toxic substances, production of important metabolites and obstacle to the growth of microorganisms harmful to the organism), is responsible for a state of chronic low-grade inflammation mediated by binding to pattern recognition receptors (PPR), the activation of peroxidation processes, membrane rupture and release of cellular components into the extracellular space and the triggering of autoimmune reactions.

Standard cell components [56], considered part of the cellular self, usually do not stimulate immune and inflammatory reactions. However, when they are misplaced and occur outside of their standard physiological location, they could sense and bind pattern recognition receptors (PPRs) inducing an inflammatory response.

The misplacement of self-molecules appears to increase with age, supporting the hypothesis that inflammaging is fueled by increased exposure of cell components. Given that mitochondria are reminiscent of their ancestral bacterial origin, their parts share with bacteria the ability to bind to PRRs leading to an inflammatory response.

The inflammatory phenotype originates inside cells with different organelles’ contributions, including the nucleus, and various mechanisms, such as telomere attrition, DNA damage response (DDR), mitochondrial dysfunction, proteasome/lysosome alteration, inflammasome activation, and endoplasmic reticulum (ER) stress. Supporting this idea, human longevity is characterized by the preserved function of proteasomes [57] and autophagy (both mechanisms involved to prevent the accumulation of cellular components): the activity of the ubiquitin-proteasome system and autophagy have also been associated with the rate of aging [58, 59], and a higher expression of the immunoproteasome is a sign of neuroinflammation, as found in the brains of patients with Alzheimer Disease (AD) but not of healthy older subjects [60].

These misplaced molecules are not confined within the cell and actively or passively leave them, being taken up by other distal cells and triggering a feed-forward propagation–amplification cycle of inflammation and inflammaging in distant cells by traveling via the circulatory and lymphatic systems. This suggests that inflammaging is propagated by the secretion of damaged cellular components produced by compromised, stressed, or senescent cells and organelles during a pathological event [61], transmitted in a paracrine fashion by activation of IL-1 signaling [62]. Local propagation can be significant in pathologies such as cancer in older subjects, where systemic inflammaging occurs [63]. Moreover, systemic inflammation can accelerate aging via reactive oxygen species (ROS)-mediated exacerbation of telomere dysfunction and cell senescence in the absence of any other genetic or environmental factors [64].

There is, therefore, excellent attention today towards all those products capable of reducing these inflammatory mechanisms at the base of aging and all those changes associated with it (Table 1).

Peripheral neuropathy

Inflammation is also the basis of chemotherapy-induced painful peripheral neuropathy (CIPN).

In particular, chemotherapeutics damage the nervous system through mechanisms involving microtubule disruption, oxidative stress, mitochondrial damage, altered ion channel activity, DNA damage, immunological processes, and neuroinflammation [65]. which leading to the CIPN.

CIPN is a predominantly sensory neuropathy as a typical “glove and stocking” neuropathy; it is characterized by high prevalence among cancer patients negatively impacting their quality of life and it varies in intensity and duration usually emerging weeks or months after chemotherapy completion.

For these reasons, chemotherapy-induced painful neuropathy (CIPN) is considered a significant dose-limiting side effect of several chemotherapeutic agents limiting therapeutic options for patients.

Six main agent groups can result in CIPN development: platinum-based antineoplastics (particularly oxaliplatin and cisplatin), vinca alkaloids, epothilones, taxanes, proteasome inhibitors, and immunomodulatory drugs.

Data suggest that, as a natural antioxidant compound, the administration of Silibinin in combination with oxaliplatin, the best known among chemotherapeutics for inducing CIPN through oxidative stress), can prevent oxidative damage reducing oxaliplatin-dependent pain neuropathy. Accordingly, Silibinin could be a valid therapeutic option for CIPN [66] (Table 1).

Conclusions and potential new fields

Inflammation is a critical process in tumor progression as demonstrated by the fact that cytokines released in chronic inflammatory conditions provide a microenvironment favourable to tumor growth and metastasis [67]. In particular, mitochondrial damage it would seem plays an essential role in inflammation as it is a prerequisite for the assembly and activation of inflammasome through oxidized mitochondrial DNA (mtDNA) released in a ROS-dependent manner [68, 69]. On the basis of these considerations, data suggest that Silibinin and NAC administration, limiting ROS generation, can decrease the production of ox-mtDNA in human triple-negative breast cancer cells, [13, 70, 71].

This consideration could be the starting point to study whether Silibinin could contrast tumor progression and aging and inflammaging through molecular and cellular mechanisms until now not completely clarified.

Furthermore, data suggest that the inhibition of STAT3 mediated by Silibilin, could be considered an interesting therapeutic approach also for patients with brain metastases. Indeed, this setting of patients maintains a poor prognosis with limited treatment options, therefore, further investigations on the role of Silibilin in this setting of patient could offer better outcomes and survival benefit.

With the aim of confirming this hypothesis a randomised phase 2 study is investigating the preventive role of Silibinin against placebo in patients who have undergone complete resection of a brain metastasis from NSCLC and breast cancers (NCT05689619).

AUTHOR CONTRIBUTIONS

Conceptualization and design of the work: Elisa Roca, Giuseppe Ferdinando Colloca, Andrea Bellieni. Methodology: Giuseppe Ferdinando Colloca, Licia Martinelli, Maria Elisa Damiani, Ilaria Zampieri. Writing-original draft preparation: Elisa Roca, Giuseppe Ferdinando Colloca, Fiorella Lombardo, Alessandra Cucinella, Giorgio Madonia. Writing-review and editing: Elisa Roca, Giuseppe Ferdinando Colloca. Supervision: Elisa Roca, Giuseppe Ferdinando Colloca, Antonio Santo.

CONFLICTS OF INTEREST

Authors have no conflicts of interest to declare.

FUNDING

No funding was used for this paper.

References

1. Cheung CW, Gibbons N, Johnson DW, Nicol DL. Silibinin--a promising new treatment for cancer. Anticancer Agents Med Chem. 2010; 10:186–95. https://doi.org/10.2174/1871520611009030186. [PubMed].

2. Singh RP, Gu M, Agarwal R. Silibinin inhibits colorectal cancer growth by inhibiting tumor cell proliferation and angiogenesis. Cancer Res. 2008; 68:2043–50. https://doi.org/10.1158/0008-5472.CAN-07-6247. [PubMed].

3. Ramasamy K, Agarwal R. Multitargeted therapy of cancer by silymarin. Cancer Lett. 2008; 269:352–62. https://doi.org/10.1016/j.canlet.2008.03.053. [PubMed].

4. Kaur M, Agarwal R. Silymarin and epithelial cancer chemoprevention: how close we are to bedside? Toxicol Appl Pharmacol. 2007; 224:350–59. https://doi.org/10.1016/j.taap.2006.11.011. [PubMed].

5. Kim KW, Choi CH, Kim TH, Kwon CH, Woo JS, Kim YK. Silibinin inhibits glioma cell proliferation via Ca2+/ROS/MAPK-dependent mechanism in vitro and glioma tumor growth in vivo. Neurochem Res. 2009; 34:1479–90. https://doi.org/10.1007/s11064-009-9935-6. [PubMed].

6. Chakrabarti M, Ray SK. Anti-tumor activities of luteolin and silibinin in glioblastoma cells: overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo. Apoptosis. 2016; 21:312–28. https://doi.org/10.1007/s10495-015-1198-x. [PubMed].

7. Qi L, Singh RP, Lu Y, Agarwal R, Harrison GS, Franzusoff A, Glode LM. Epidermal growth factor receptor mediates silibinin-induced cytotoxicity in a rat glioma cell line. Cancer Biol Ther. 2003; 2:526–31. https://doi.org/10.4161/cbt.2.5.452. [PubMed].

8. Son YG, Kim EH, Kim JY, Kim SU, Kwon TK, Yoon AR, Yun CO, Choi KS. Silibinin sensitizes human glioma cells to TRAIL-mediated apoptosis via DR5 up-regulation and down-regulation of c-FLIP and survivin. Cancer Res. 2007; 67:8274–84. https://doi.org/10.1158/0008-5472.CAN-07-0407. [PubMed].

9. Grijalba MT, Vercesi AE, Schreier S. Ca2+-induced increased lipid packing and domain formation in submitochondrial particles. A possible early step in the mechanism of Ca2+-stimulated generation of reactive oxygen species by the respiratory chain. Biochemistry. 1999; 38:13279–87. https://doi.org/10.1021/bi9828674. [PubMed].

10. Dykens JA. Isolated cerebral and cerebellar mitochondria produce free radicals when exposed to elevated CA2+ and Na+: implications for neurodegeneration. J Neurochem. 1994; 63:584–91. https://doi.org/10.1046/j.1471-4159.1994.63020584.x. [PubMed].

11. Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol. 2002; 192:1–15. https://doi.org/10.1002/jcp.10119. [PubMed].

12. Chakrabarti M, Ray SK. Synergistic anti-tumor actions of luteolin and silibinin prevented cell migration and invasion and induced apoptosis in glioblastoma SNB19 cells and glioblastoma stem cells. Brain Res. 2015; 1629:85–93. https://doi.org/10.1016/j.brainres.2015.10.010. [PubMed].

13. Si L, Fu J, Liu W, Hayashi T, Nie Y, Mizuno K, Hattori S, Fujisaki H, Onodera S, Ikejima T. Silibinin inhibits migration and invasion of breast cancer MDA-MB-231 cells through induction of mitochondrial fusion. Mol Cell Biochem. 2020; 463:189–201. https://doi.org/10.1007/s11010-019-03640-6. [PubMed].

14. Zi X, Zhang J, Agarwal R, Pollak M. Silibinin up-regulates insulin-like growth factor-binding protein 3 expression and inhibits proliferation of androgen-independent prostate cancer cells. Cancer Res. 2000; 60:5617–20. [PubMed].

15. Dhanalakshmi S, Singh RP, Agarwal C, Agarwal R. Silibinin inhibits constitutive and TNFalpha-induced activation of NF-kappaB and sensitizes human prostate carcinoma DU145 cells to TNFalpha-induced apoptosis. Oncogene. 2002; 21:1759–67. https://doi.org/10.1038/sj.onc.1205240. [PubMed].

16. Bosch-Barrera J, Menendez JA. Silibinin and STAT3: A natural way of targeting transcription factors for cancer therapy. Cancer Treat Rev. 2015; 41:540–46. https://doi.org/10.1016/j.ctrv.2015.04.008. [PubMed].

17. Furtek SL, Backos DS, Matheson CJ, Reigan P. Strategies and Approaches of Targeting STAT3 for Cancer Treatment. ACS Chem Biol. 2016; 11:308–18. https://doi.org/10.1021/acschembio.5b00945. [PubMed].

18. Chang Q, Bournazou E, Sansone P, Berishaj M, Gao SP, Daly L, Wels J, Theilen T, Granitto S, Zhang X, Cotari J, Alpaugh ML, de Stanchina E, et al. The IL-6/JAK/Stat3 feed-forward loop drives tumorigenesis and metastasis. Neoplasia. 2013; 15:848–62. https://doi.org/10.1593/neo.13706. [PubMed].

19. Thiagarajan A, Yamada Y. Radiobiology and radiotherapy of brain metastases. Clin Exp Metastasis. 2017; 34:411–19. https://doi.org/10.1007/s10585-017-9865-7. [PubMed].

20. Kienast Y, von Baumgarten L, Fuhrmann M, Klinkert WE, Goldbrunner R, Herms J, Winkler F. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010; 16:116–22. https://doi.org/10.1038/nm.2072. [PubMed].

21. Valiente M, Obenauf AC, Jin X, Chen Q, Zhang XH, Lee DJ, Chaft JE, Kris MG, Huse JT, Brogi E, Massagué J. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell. 2014; 156:1002–16. https://doi.org/10.1016/j.cell.2014.01.040. [PubMed].

22. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, Jacob L, Patwa R, Shah H, Xu K, Cross JR, Massagué J. Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016; 533:493–98. https://doi.org/10.1038/nature18268. [PubMed].

23. Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009; 32:638–47. https://doi.org/10.1016/j.tins.2009.08.002. [PubMed].

24. Priego N, Zhu L, Monteiro C, Mulders M, Wasilewski D, Bindeman W, Doglio L, Martínez L, Martínez-Saez E, Ramón Y Cajal S, Megías D, Hernández-Encinas E, Blanco-Aparicio C, et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat Med. 2018; 24:1024–35. https://doi.org/10.1038/s41591-018-0044-4. [PubMed].

25. Lee HT, Xue J, Chou PC, Zhou A, Yang P, Conrad CA, Aldape KD, Priebe W, Patterson C, Sawaya R, Xie K, Huang S. Stat3 orchestrates interaction between endothelial and tumor cells and inhibition of Stat3 suppresses brain metastasis of breast cancer cells. Oncotarget. 2015; 6:10016–29. https://doi.org/10.18632/oncotarget.3540. [PubMed].

26. Jones LM, Broz ML, Ranger JJ, Ozcelik J, Ahn R, Zuo D, Ursini-Siegel J, Hallett MT, Krummel M, Muller WJ. STAT3 Establishes an Immunosuppressive Microenvironment during the Early Stages of Breast Carcinogenesis to Promote Tumor Growth and Metastasis. Cancer Res. 2016; 76:1416–28. https://doi.org/10.1158/0008-5472.CAN-15-2770. [PubMed].

27. Janus N, Thariat J, Boulanger H, Deray G, Launay-Vacher V. Proposal for dosage adjustment and timing of chemotherapy in hemodialyzed patients. Ann Oncol. 2010; 21:1395–403. https://doi.org/10.1093/annonc/mdp598. [PubMed].

28. Malyszko J, Kozlowska K, Kozlowski L, Malyszko J. Nephrotoxicity of anticancer treatment. Nephrol Dial Transplant. 2017; 32:924–36. https://doi.org/10.1093/ndt/gfw338. [PubMed].

29. Sonnenbichler J, Scalera F, Sonnenbichler I, Weyhenmeyer R. Stimulatory effects of silibinin and silicristin from the milk thistle Silybum marianum on kidney cells. J Pharmacol Exp Ther. 1999; 290:1375–83. [PubMed].

30. Barré B, Vigneron A, Perkins N, Roninson IB, Gamelin E, Coqueret O. The STAT3 oncogene as a predictive marker of drug resistance. Trends Mol Med. 2007; 13:4–11. https://doi.org/10.1016/j.molmed.2006.11.001. [PubMed].

31. Su WP, Cheng FY, Shieh DB, Yeh CS, Su WC. PLGA nanoparticles codeliver paclitaxel and Stat3 siRNA to overcome cellular resistance in lung cancer cells. Int J Nanomedicine. 2012; 7:4269–83. https://doi.org/10.2147/IJN.S33666. [PubMed].

32. Walker SR, Chaudhury M, Frank DA. STAT3 Inhibition by Microtubule-Targeted Drugs: Dual Molecular Effects of Chemotherapeutic Agents. Mol Cell Pharmacol. 2011; 3:13–19. [PubMed].

33. Dizaji MZ, Malehmir M, Ghavamzadeh A, Alimoghaddam K, Ghaffari SH. Synergistic effects of arsenic trioxide and silibinin on apoptosis and invasion in human glioblastoma U87MG cell line. Neurochem Res. 2012; 37:370–80. https://doi.org/10.1007/s11064-011-0620-1. [PubMed].

34. Lee HJ, Zhuang G, Cao Y, Du P, Kim HJ, Settleman J. Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell. 2014; 26:207–21. https://doi.org/10.1016/j.ccr.2014.05.019. [PubMed].

35. Li MX, Bi XY, Huang Z, Zhao JJ, Han Y, Li ZY, Zhang YF, Li Y, Chen X, Hu XH, Zhao H, Cai JQ. Prognostic Role of Phospho-STAT3 in Patients with Cancers of the Digestive System: A Systematic Review and Meta-Analysis. PLoS One. 2015; 10:e0127356. https://doi.org/10.1371/journal.pone.0127356. [PubMed].

36. Zhang FQ, Yang WT, Duan SZ, Xia YC, Zhu RY, Chen YB. JAK2 inhibitor TG101348 overcomes erlotinib-resistance in non-small cell lung carcinoma cells with mutated EGF receptor. Oncotarget. 2015; 6:14329–43. https://doi.org/10.18632/oncotarget.3685. [PubMed].

37. Gao SP, Chang Q, Mao N, Daly LA, Vogel R, Chan T, Liu SH, Bournazou E, Schori E, Zhang H, Brewer MR, Pao W, Morris L, et al. JAK2 inhibition sensitizes resistant EGFR-mutant lung adenocarcinoma to tyrosine kinase inhibitors. Sci Signal. 2016; 9:ra33. https://doi.org/10.1126/scisignal.aac8460. [PubMed].

38. Cufí S, Bonavia R, Vazquez-Martin A, Corominas-Faja B, Oliveras-Ferraros C, Cuyàs E, Martin-Castillo B, Barrajón-Catalán E, Visa J, Segura-Carretero A, Bosch-Barrera J, Joven J, Micol V, Menendez JA. Silibinin meglumine, a water-soluble form of milk thistle silymarin, is an orally active anti-cancer agent that impedes the epithelial-to-mesenchymal transition (EMT) in EGFR-mutant non-small-cell lung carcinoma cells. Food Chem Toxicol. 2013; 60:360–68. https://doi.org/10.1016/j.fct.2013.07.063. [PubMed].

39. Corominas-Faja B, Oliveras-Ferraros C, Cuyàs E, Segura-Carretero A, Joven J, Martin-Castillo B, Barrajón-Catalán E, Micol V, Bosch-Barrera J, Menendez JA. Stem cell-like ALDH(bright) cellular states in EGFR-mutant non-small cell lung cancer: a novel mechanism of acquired resistance to erlotinib targetable with the natural polyphenol silibinin. Cell Cycle. 2013; 12:3390–404. https://doi.org/10.4161/cc.26417. [PubMed].

40. Chaib I, Karachaliou N, Pilotto S, Codony Servat J, Cai X, Li X, Drozdowskyj A, Servat CC, Yang J, Hu C, Cardona AF, Vivanco GL, Vergnenegre A, et al. Co-activation of STAT3 and YES-Associated Protein 1 (YAP1) Pathway in EGFR-Mutant NSCLC. J Natl Cancer Inst. 2017; 109:djx014. https://doi.org/10.1093/jnci/djx014. [PubMed].

41. Zhou S, Zhang M, Zhou C, Wang W, Yang H, Ye W. The role of epithelial-mesenchymal transition in regulating radioresistance. Crit Rev Oncol Hematol. 2020; 150:102961. https://doi.org/10.1016/j.critrevonc.2020.102961. [PubMed].

42. You S, Li R, Park D, Xie M, Sica GL, Cao Y, Xiao ZQ, Deng X. Disruption of STAT3 by niclosamide reverses radioresistance of human lung cancer. Mol Cancer Ther. 2014; 13:606–16. https://doi.org/10.1158/1535-7163.MCT-13-0608. [PubMed].

43. Zang C, Liu X, Li B, He Y, Jing S, He Y, Wu W, Zhang B, Ma S, Dai W, Li S, Peng Z. IL-6/STAT3/TWIST inhibition reverses ionizing radiation-induced EMT and radioresistance in esophageal squamous carcinoma. Oncotarget. 2017; 8:11228–38. https://doi.org/10.18632/oncotarget.14495. [PubMed].

44. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, Schwabe RF, Vahdat LT, Altorki NK, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015; 527:472–76. https://doi.org/10.1038/nature15748. [PubMed].

45. Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS, Kalluri R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015; 527:525–30. https://doi.org/10.1038/nature16064. [PubMed].

46. Nambiar DK, Rajamani P, Singh RP. Silibinin attenuates ionizing radiation-induced pro-angiogenic response and EMT in prostate cancer cells. Biochem Biophys Res Commun. 2015; 456:262–68. https://doi.org/10.1016/j.bbrc.2014.11.069. [PubMed].

47. Colloca G, Di Capua B, Bellieni A, Fusco D, Ciciarello F, Tagliaferri L, Valentini V, Balducci L. Biological and Functional Biomarkers of Aging: Definition, Characteristics, and How They Can Impact Everyday Cancer Treatment. Curr Oncol Rep. 2020; 22:115. https://doi.org/10.1007/s11912-020-00977-w. [PubMed].

48. Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000; 908:244–54. https://doi.org/10.1111/j.1749-6632.2000.tb06651.x. [PubMed].

49. Colloca G, Di Capua B, Bellieni A, Cesari M, Marzetti E, Valentini V, Calvani R. Muscoloskeletal aging, sarcopenia and cancer. J Geriatr Oncol. 2019; 10:504–9. https://doi.org/10.1016/j.jgo.2018.11.007. [PubMed].

50. Lencel P, Magne D. Inflammaging: the driving force in osteoporosis? Med Hypotheses. 2011; 76:317–21. https://doi.org/10.1016/j.mehy.2010.09.023. [PubMed].

51. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014 (Suppl 1); 69:S4–9. https://doi.org/10.1093/gerona/glu057. [PubMed].

52. Vitale G, Salvioli S, Franceschi C. Oxidative stress and the ageing endocrine system. Nat Rev Endocrinol. 2013; 9:228–40. https://doi.org/10.1038/nrendo.2013.29. [PubMed].

53. Singh T, Newman AB. Inflammatory markers in population studies of aging. Ageing Res Rev. 2011; 10:319–29. https://doi.org/10.1016/j.arr.2010.11.002. [PubMed].

54. Roxburgh CS, McMillan DC. Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol. 2010; 6:149–63. https://doi.org/10.2217/fon.09.136. [PubMed].

55. Sacheck JM, Cannon JG, Hamada K, Vannier E, Blumberg JB, Roubenoff R. Age-related loss of associations between acute exercise-induced IL-6 and oxidative stress. Am J Physiol Endocrinol Metab. 2006; 291:E340–49. https://doi.org/10.1152/ajpendo.00052.2005. [PubMed].

56. Dall’Olio F, Vanhooren V, Chen CC, Slagboom PE, Wuhrer M, Franceschi C. N-glycomic biomarkers of biological aging and longevity: a link with inflammaging. Ageing Res Rev. 2013; 12:685–98. https://doi.org/10.1016/j.arr.2012.02.002. [PubMed].

57. Chondrogianni N, Petropoulos I, Franceschi C, Friguet B, Gonos ES. Fibroblast cultures from healthy centenarians have an active proteasome. Exp Gerontol. 2000; 35:721–28. https://doi.org/10.1016/s0531-5565(00)00137-6. [PubMed].

58. Cuervo AM, Wong E. Chaperone-mediated autophagy: roles in disease and aging. Cell Res. 2014; 24:92–104. https://doi.org/10.1038/cr.2013.153. [PubMed].

59. Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK, Sadoshima J. Aging and Autophagy in the Heart. Circ Res. 2016; 118:1563–76. https://doi.org/10.1161/CIRCRESAHA.116.307474. [PubMed].

60. Mishto M, Bellavista E, Santoro A, Stolzing A, Ligorio C, Nacmias B, Spazzafumo L, Chiappelli M, Licastro F, Sorbi S, Pession A, Ohm T, Grune T, Franceschi C. Immunoproteasome and LMP2 polymorphism in aged and Alzheimer’s disease brains. Neurobiol Aging. 2006; 27:54–66. https://doi.org/10.1016/j.neurobiolaging.2004.12.004. [PubMed].

61. Nelson G, Wordsworth J, Wang C, Jurk D, Lawless C, Martin-Ruiz C, von Zglinicki T. A senescent cell bystander effect: senescence-induced senescence. Aging Cell. 2012; 11:345–49. https://doi.org/10.1111/j.1474-9726.2012.00795.x. [PubMed].

62. Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, Athineos D, Kang TW, Lasitschka F, Andrulis M, Pascual G, Morris KJ, Khan S, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013; 15:978–90. https://doi.org/10.1038/ncb2784. [PubMed].

63. Bonafè M, Storci G, Franceschi C. Inflamm-aging of the stem cell niche: breast cancer as a paradigmatic example: breakdown of the multi-shell cytokine network fuels cancer in aged people. Bioessays. 2012; 34:40–49. https://doi.org/10.1002/bies.201100104. [PubMed].

64. Jurk D, Wilson C, Passos JF, Oakley F, Correia-Melo C, Greaves L, Saretzki G, Fox C, Lawless C, Anderson R, Hewitt G, Pender SL, Fullard N, et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat Commun. 2014; 2:4172. https://doi.org/10.1038/ncomms5172. [PubMed].

65. Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J. Mechanisms of Chemotherapy-Induced Peripheral Neuropathy. Int J Mol Sci. 2019; 20:1451. https://doi.org/10.3390/ijms20061451. [PubMed].

66. Di Cesare Mannelli L, Zanardelli M, Failli P, Ghelardini C. Oxaliplatin-induced neuropathy: oxidative stress as pathological mechanism. Protective effect of silibinin. J Pain. 2012; 13:276–84. https://doi.org/10.1016/j.jpain.2011.11.009. [PubMed].

67. Ye K, Chen QW, Sun YF, Lin JA, Xu JH. Loss of BMI-1 dampens migration and EMT of colorectal cancer in inflammatory microenvironment through TLR4/MD-2/MyD88-mediated NF-κB signaling. J Cell Biochem. 2018; 119:1922–30. https://doi.org/10.1002/jcb.26353. [PubMed].

68. Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, Malech HL, Ledbetter JA, Elkon KB, Kaplan MJ. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med. 2016; 22:146–53. https://doi.org/10.1038/nm.4027. [PubMed].

69. Bordon Y. mtDNA synthesis ignites the inflammasome. Nat Rev Immunol. 2018; 18:539. https://doi.org/10.1038/s41577-018-0049-8. [PubMed].

70. West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol. 2017; 17:363–75. https://doi.org/10.1038/nri.2017.21. [PubMed].

71. Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin XJ, Wong J, Ding S, Seki E, Schnabl B, Hevener AL, Greenberg HB, Kisseleva T, Karin M. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature. 2018; 560:198–203. https://doi.org/10.1038/s41586-018-0372-z. [PubMed].


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 28587