Article Search
닫기

Journal of Cancer Prevention

Commentary

J Cancer Prev 2024; 29(1): 1-5

Published online March 30, 2024

https://doi.org/10.15430/JCP.24.001

© Korean Society of Cancer Prevention

Resveratrol Promotes Self-digestion to Put Cancer to Sleep

Alessandra Ferraresi , Suyanee Thongchot , Ciro Isidoro

Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy

Correspondence to :
Ciro Isidoro, E-mail: ciro.isidoro@med.uniupo.it, https://orcid.org/0000-0002-5494-3034

Received: January 8, 2024; Revised: January 15, 2024; Accepted: January 15, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Resveratrol, a natural polyphenol present in a variety of food stuff, has been shown to exert preventive and curative anticancer activity in several in vitro and in vivo models. Such chemopreventive/anticancer activity has been linked to biochemical and epigenetic modifications of multiple pathways involved in carcinogenesis and metastasization. In this commentary, we focus on the recent work done in our laboratory showing that resveratrol has potential to prevent and cure cancer by promoting epigenetic-mediated autophagy-dependent tumor dormancy, an effect associated with re-education of the cancer-associated fibroblasts and reduced production of inflammatory cytokines in the tumor microenvironment. The clinical translation of the current knowledge on resveratrol anticancer activity is also discussed.

Keywords: Natural products, Dormancy, Autophagy, Cytokines, Tumor microenvironment

Trans-resveratrol (3,4,5-trihydroxystilbene; RV), a natural polyphenol particularly enriched in berries and grapes, has been proven an effective nutraceutical for cancer prevention and treatment in several types of malignancy, both in in vitro and in vivo preclinical models [1,2]. The efficacy of RV in inhibiting cancer growth can be attributed to its ability to target multiple pathways [1,3,4]. To mention a few, RV has been shown to induce autophagy, apoptosis and autophagy-associated apoptosis in ovarian and colorectal cancer cells [5-8]. These led to inhibition of epithelial-mesenchymal transition and migration of ovarian cancer cells [9], inhibition of reduced glucose uptake and metabolism in ovarian cancer cells [10], or suppression of the insulin like growth factor 1 receptor/Akt/Wnt-β-catenin pathways in colon cancers [11] and in stem-like breast cancer cells [12]. RV regulates many of these pathways through epigenetic mechanisms [13]. For instance, we have shown that RV can modulate microRNAs to rescue the re-expression of aplasia Ras-homolog member I (ARH-I; aka DIRAS3), an imprinted tumor suppressor that positively regulates BECLIN-1-dependent autophagy [9].

Interestingly, two independent studies have shown that RV can also interrupt the malignant cross-talk between cancer-associated fibroblasts (CAFs) and cancer cells in the tumor microenvironment [14,15]. RV could inhibit breast cancer cell proliferation and migration and the stem-like properties that were promoted by CAFs, and this effect was due to inhibition of the expression of cyclin D1 and c-Myc, and of the STAT3 (a transcription factor triggered by interleukin [IL]-6), Akt and self-renewal pathways [14]. In our study, the conditioned medium of CAFs pre-treated with RV lost the ability to induce proliferation and invasiveness of cholangiocarcinoma cells [15]. The anticancer activity of RV has been proven in several preclinical in vivo models, including breast cancer [16,17], colorectal cancer [18], and lung cancer [19]. RV alone [19] or in combination with curcumin and quercetin [17] showed the ability to also modify the tumor microenvironment, reversing the infiltration of immunosuppressive Th2 lymphocytes, tumor-associated N2 neutrophils and tumor-associated M2 macrophages.

We have previously shown that RV acts as a protein-restriction mimetic to induce mTOR-dependent autophagy, a self-digestion lysosomal-mediated process which oversees cellular homeostasis [8]. Recently, we found that reduced overall survival of cholangiocarcinoma patients was associated with low expression of autophagy markers in cancer cells along with high expression of IL-6 and high infiltration of CAFs in the tumor microenvironment [20]. By pre-treating cholangiocarcinoma-derived CAFs with RV in vitro, we could inhibit the secretion of IL-6 so that their conditioned medium lost the ability to repress autophagy and to induce proliferation and migration of the exposed cholangiocarcinoma cells [15]. This prompted us to test in vivo the preventive and curative potential of RV. To this end, we set three groups of nude mice in which we transplanted subcutaneously human cholangiocarcinoma cells. Mice in the first group (that had not received any treatment for five weeks) served as controls, the mice in the second group were treated with 50 mg/kg RV in drinking water two weeks before tumor transplantation and for the next five weeks, and they served as the “preventive” group, and the mice in the third group started the RV treatment one week after tumor transplantation and served as the “curative” group. The results can be summarized as follows: (i) in controls, the tumors reached an average volume of 5.00 mm3 after 5 weeks in all the five mice, while in the “preventive” group, only three mice developed a tumor with an average volume of 0.15 mm3, and in the “curative” group the five mice developed a tumor with an average volume of 1.0 mm3; (ii) both in the “preventive” and “curative” groups, the infiltrate of CAFs and the presence of stromal IL-6 were drastically decreased; (iii) in the tumors from both the “preventive” and the “curative” groups, the autophagy markers microtubule-associated protein 1 light chain 3 (LC3) and BECLIN-1 were highly expressed [21].

It has been reported that RV can act epigenetically to modulate autophagy in cancer [22-24], and it is known that the secretions in the tumor microenvironment can epigenetically modulate autophagy to drive tumor dormancy [25]. In a separate study, we demonstrated that RV could keep ovarian cancer cells grown as 3D spheroids (a proxy of peritoneal metastatic colonies) in a dormant state even in the presence of IL-6, and this effect was due to the down-regulation of miR-1305 and concomitant re-expression of its target ARH-I (DIRAS3), which then rescued BECLIN-1-dependent autophagy [26]. By extracting the data from TCGA, we found that cholangiocarcinomas highly expressing BECN1 also showed high levels of DIRAS3 and CDKN1A mRNA, strongly suggesting a dormant state of the tumors [21]. This prompted us to check for the expression of ARH-I (DIRAS3) and autophagy and dormancy markers in the biopsies of the preclinical model of transplanted cholangiocarcinomas in nude mice described above. To this end, we searched for co-expression of the autophagy markers LC3 and BECLIN-1 with p21, the marker for cell cycle arrest encoded by CDKN1A, and with ARH-1/DIRAS3 (encoded by DIRAS3), a protein playing a pivotal role in the switch from autophagic cell death to autophagy-driven cell dormancy [27]. The images in Figure 1 show that RV-treated tumors, either via the preventive or the curative protocol, present high levels of LC3 and p21 co-expression, suggestive of the involvement of autophagy in cell cycle arrest. Further, RV greatly enhanced the expression of DIRAS3, which was shown to largely interact with BECLIN-1. To be noted, in RV-treated tumors, the expression of STAT3 (which is downstream of IL-6 signalling) was decreased, and this protein was found largely co-localized in the cytoplasm bound to DIRAS3. Taken together, these data suggest induction of a dormant state in association with induction of autophagy in the tumors exposed to RV.

Figure 1. The immunofluorescence co-staining for LC3/p21, BECLIN-1/DIRAS3, LC3/DIRAS3 and DIRAS3/STAT3 shows up-regulation of autophagy and autophagy-dependent dormancy in Pre-RV + RV and RV-treated mice (for treatment details see the text and reference [21]). RV, resveratrol; LC3, light chain 3. Scale bar = 20 µm.

Cancer develops following genetic and epigenetic alterations of the genes controlling cell cycle and cell proliferation, telomerase activity and stemness, cell metabolism, motility and invasiveness, autophagy, apoptosis, and other types (e.g., ferroptosis) of cell death, along with the complicity of the tumor microenvironment which promotes angiogenesis and inhibits the immune response [28]. Chemotherapy is the gold standard for cancer treatment, but unfortunately it has several drawbacks that discourages the patients from continuing the therapy [29]. Disappointingly, even precision therapy with molecular drugs targeting “driver” oncogenic proteins has a limited efficacy [30,31]. RV could represent a valid alternative and/or adjuvant therapy for synergizing with chemotherapeutics and at the same time mitigating their undesired side effects [32]. Two characteristics make RV an attractive cancer therapeutic worth of further investigations: (i) it is a hormetic drug, meaning that it elicits different effects in normal and diseased tissues depending on the concentration [33], and (ii) it causes negligible, if any, side effects [34]. Moreover, a recent systematic drug-target interaction network study found that RV can target up to 23 driver genes (including mTOR, BRCA1, TP53, PTEN, HGF, CTNB1, CDKN1A, and AKT1, among others) significantly mutated in up to 15 types of cancer [35]. Yet, clinical trials with RV (available at https://clinicaltrials.gov/ct2/) appear not to meet the expectations, likely because of the low bioavailability and rapid excretion of RV, which prevent it from reaching a pharmacologic concentration in the target tissue [36].

It is indeed puzzling that despite the vastness of in vitro and in vivo preclinical data supporting its anticancer activity, the clinical efficacy of RV in cancer patients remains ambiguous and inconclusive [37]. Possible reasons for the failure of the clinical trials include mistakes in the experimental design (time, dose, administration route, patient’s selection criteria, etc.), the supposed mechanisms of action, and the endpoints, which are based on in vitro and in animal studies [36,38]. First, the assumption that high dose administration is needed to obtain the desired therapeutic effect in the diseased organ has been challenged by a recent study showing that low dietary administration of RV (5 mg) is more effective than a pharmacologic dose (1 g) for a chemopreventive activity in colorectal cancer [38]. Interestingly, the chemopreventive effect was influenced by the diet, and it was associated with increased expression of LC3 and p21 (indicative of autophagy-mediated arrest of cell proliferation) in human colonic mucosa [38]. To be noted, the oral administration of 5 or 50 mgRV twice daily for 12 weeks in breast cancer women led to hypomethylation and re-expression of the RASSF-1α tumor suppressor gene encoding a RAS-associated protein that inhibits cell cycle and promotes apoptosis [39].

Regarding the mechanisms of action, it is conceivable that the pharmacologic inhibition of biochemical pathways attained in vitro at a relatively high concentration (10 to 200 µM) of RV unlikely occurs in patients, where its plasma concentration reached 20 nM after oral administration of 360 µg/kg [40]. As a comparison with the mouse, 15 mg/kg of RV administered in drinking water (0.01%) would lead to a putative peak in the plasma of 1.5 µM [36]. Another possible reason for the different outcome at a given dose in animal experiments and clinical trials is that orally administered RV synergizes with microbiota metabolites to exert anticancer activity via yet unidentified pathways [41]. The microbiota in rodents and in humans are different and influenced by the diet, and RV itself can modify the microbiota [41]. This fact may account for the difference in the dose-response between the two species. This hypothesis is being explored in our laboratory.

The dose of RV (50 mg/kg) used in our nude mice model (which would lead to a putative peak in the plasma of 5 µM) might be equivalent to approximately 500 to 600 mg per 60 to 70 kg individual. Based on our findings, we hypothesize that for curative purposes, oral administration of RV at 10 to 20 mg/kg (approximately 600 to 1,400 mg per 60 to 70 kg body weight) daily eventually leads to epigenetic changes of the gene expression and metabolism both in cancer and stromal cells. This can restore homeostasis and tumor dormancy in the affected tissue. Of course, the dose must be personalized to the patient, taking into account sensitive factors such as age, tumor stage, performance, diet, lifestyle, and concomitant therapies with drugs and/or other natural products.

As per the treatment endpoints, it is to be stressed that in our animal models treated with RV in drinking water, we obtained an effect not only in cancer cells (where markers of autophagy, apoptosis, cell cycle arrest and cell dormancy were induced) but also in the tumor microenvironment (where infiltration of CAFs and stromal IL-6 levels were decreased) [21]. In a separate study, we showed that RV also reduces angiogenesis and infiltration of immune-suppressive cells in the tumor microenvironment of transplanted lung cancer cells [19]. The latter finding is compatible with induction of angiogenic and immune-mediated tumor dormancy. The multiple processes targeted by RV leading to tumor dormancy are schematically summarized in Figure 2.

Figure 2. The cartoon illustrates the action of RV in the tumor microenvironment comprising fibroblasts and immune cells as well as the cancer cells which eventually leads to tumor dormancy. ATG, autophagy; CCD, cancer cell dormancy; MTX, metastasis; RV, resveratrol; TME, tumor microenvironment; Fb, fibroblasts; Th, T CD4 lymphocyte; M, macrophages; CAF, cancer-associated fibroblasts; BM, basement membrane; IL, interleukin.

In summary, the body of scientific evidence reported here demonstrates that RV can block tumor growth by inducing “drowsiness” of tumor cells and modifying the microenvironment to create a “comfortable bed” to put the tumor to sleep.

Alessandra Ferraresi is recipient of a post-doctoral fellowship granted by Fondazione Umberto Veronesi (FUV 2023). Suyanee Thongchot is a post-doc fellow at Siriraj Faculty of Medicine, Mahidol University (Bangkok, Thailand). The animal core facilities for animal experiments at the Center of Excellence for Cholangiocarcinoma in Khon Kaen (Thailand) and for cell imaging at Department of Health Sciences in Novara (Italy) are fully acknowledged.

No potential conflicts of interest were disclosed.

  1. Aggarwal BB, Bhardwaj A, Aggarwal RS, Seeram NP, Shishodia S, Takada Y. Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Res 2004;24(5A):2783-840.
  2. Ko JH, Sethi G, Um JY, Shanmugam MK, Arfuso F, Kumar AP, et al. The role of resveratrol in cancer therapy. Int J Mol Sci 2017;18:2589.
    Pubmed KoreaMed CrossRef
  3. Wu SX, Xiong RG, Huang SY, Zhou DD, Saimaiti A, Zhao CN, et al. Effects and mechanisms of resveratrol for prevention and management of cancers: an updated review. Crit Rev Food Sci Nutr 2023;63:12422-40.
    Pubmed CrossRef
  4. Kulkarni SS, Cantó C. The molecular targets of resveratrol. Biochim Biophys Acta 2015;1852:1114-23.
    Pubmed CrossRef
  5. Opipari AW Jr, Tan L, Boitano AE, Sorenson DR, Aurora A, Liu JR. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res 2004;64:696-703.
    Pubmed CrossRef
  6. Trincheri NF, Nicotra G, Follo C, Castino R, Isidoro C. Resveratrol induces cell death in colorectal cancer cells by a novel pathway involving lysosomal cathepsin D. Carcinogenesis 2007;28:922-31.
    Pubmed CrossRef
  7. Trincheri NF, Follo C, Nicotra G, Peracchio C, Castino R, Isidoro C. Resveratrol-induced apoptosis depends on the lipid kinase activity of Vps34 and on the formation of autophagolysosomes. Carcinogenesis 2008;29:381-9.
    Pubmed CrossRef
  8. Ferraresi A, Titone R, Follo C, Castiglioni A, Chiorino G, Dhanasekaran DN, et al. The protein restriction mimetic Resveratrol is an autophagy inducer stronger than amino acid starvation in ovarian cancer cells. Mol Carcinog 2017;56:2681-91.
    Pubmed CrossRef
  9. Ferraresi A, Phadngam S, Morani F, Galetto A, Alabiso O, Chiorino G, et al. Resveratrol inhibits IL-6-induced ovarian cancer cell migration through epigenetic up-regulation of autophagy. Mol Carcinog 2017;56:1164-81.
    Pubmed CrossRef
  10. Vidoni C, Ferraresi A, Vallino L, Salwa A, Ha JH, Seca C, et al. Glycolysis inhibition of autophagy drives malignancy in ovarian cancer: exacerbation by IL-6 and attenuation by resveratrol. Int J Mol Sci 2023;24:1723.
    Pubmed KoreaMed CrossRef
  11. Vanamala J, Reddivari L, Radhakrishnan S, Tarver C. Resveratrol suppresses IGF-1 induced human colon cancer cell proliferation and elevates apoptosis via suppression of IGF-1R/Wnt and activation of p53 signaling pathways. BMC Cancer 2010;10:238.
    Pubmed KoreaMed CrossRef
  12. Fu Y, Chang H, Peng X, Bai Q, Yi L, Zhou Y, et al. Resveratrol inhibits breast cancer stem-like cells and induces autophagy via suppressing Wnt/β-catenin signaling pathway. PLoS One 2014;9:e102535.
    Pubmed KoreaMed CrossRef
  13. Farooqi AA, Khalid S, Ahmad A. Regulation of cell signaling pathways and miRNAs by resveratrol in different cancers. Int J Mol Sci 2018;19:652.
    Pubmed KoreaMed CrossRef
  14. Suh J, Kim DH, Surh YJ. Resveratrol suppresses migration, invasion and stemness of human breast cancer cells by interfering with tumor-stromal cross-talk. Arch Biochem Biophys 2018;643:62-71.
    Pubmed CrossRef
  15. Thongchot S, Ferraresi A, Vidoni C, Loilome W, Yongvanit P, Namwat N, et al. Resveratrol interrupts the pro-invasive communication between cancer associated fibroblasts and cholangiocarcinoma cells. Cancer Lett 2018;430:160-71. Erratum in: Cancer Lett 2018;434:206-7.
    Pubmed CrossRef
  16. Garvin S, Ollinger K, Dabrosin C. Resveratrol induces apoptosis and inhibits angiogenesis in human breast cancer xenografts in vivo. Cancer Lett 2006;231:113-22.
    Pubmed CrossRef
  17. Li C, Xu Y, Zhang J, Zhang Y, He W, Ju J, et al. The effect of resveratrol, curcumin and quercetin combination on immuno-suppression of tumor microenvironment for breast tumor-bearing mice. Sci Rep 2023;13:13278.
    Pubmed KoreaMed CrossRef
  18. Sudha T, El-Far AH, Mousa DS, Mousa SA. Resveratrol and its nanoformulation attenuate growth and the angiogenesis of xenograft and orthotopic colon cancer models. Molecules 2020;25:1412.
    Pubmed KoreaMed CrossRef
  19. Savio M, Ferraresi A, Corpina C, Vandenberghe S, Scarlata C, Sottile V, et al. Resveratrol and Its analogue 4,4'-dihydroxy-trans-stilbene inhibit Lewis lung carcinoma growth in vivo through apoptosis, autophagy and modulation of the tumour microenvironment in a murine model. Biomedicines 2022;10:1784.
    Pubmed KoreaMed CrossRef
  20. Thongchot S, Vidoni C, Ferraresi A, Loilome W, Khuntikeo N, Sangkhamanon S, et al. Cancer-associated fibroblast-derived IL-6 determines unfavorable prognosis in cholangiocarcinoma by affecting autophagy-associated chemoresponse. Cancers (Basel) 2021;13:2134.
    Pubmed KoreaMed CrossRef
  21. Thongchot S, Ferraresi A, Vidoni C, Salwa A, Vallino L, Kittirat Y, et al. Preclinical evidence for preventive and curative effects of resveratrol on xenograft cholangiocarcinogenesis. Cancer Lett 2024;582:216589.
    Pubmed CrossRef
  22. Vidoni C, Ferraresi A, Secomandi E, Vallino L, Dhanasekaran DN, Isidoro C. Epigenetic targeting of autophagy for cancer prevention and treatment by natural compounds. Semin Cancer Biol 2020;66:34-44.
    Pubmed CrossRef
  23. Vallino L, Ferraresi A, Vidoni C, Secomandi E, Esposito A, Dhanasekaran DN, et al. Modulation of non-coding RNAs by resveratrol in ovarian cancer cells: in silico analysis and literature review of the anti-cancer pathways involved. J Tradit Complement Med 2020;10:217-29.
    Pubmed KoreaMed CrossRef
  24. Rajendran P, Abdelsalam SA, Renu K, Veeraraghavan V, Ben Ammar R, Ahmed EA. Polyphenols as potent epigenetics agents for cancer. Int J Mol Sci 2022;23:11712.
    Pubmed KoreaMed CrossRef
  25. Sosa MS, Bragado P, Debnath J, Aguirre-Ghiso JA. Regulation of tumor cell dormancy by tissue microenvironments and autophagy. Adv Exp Med Biol 2013;734:73-89.
    Pubmed KoreaMed CrossRef
  26. Esposito A, Ferraresi A, Salwa A, Vidoni C, Dhanasekaran DN, Isidoro C. Resveratrol contrasts IL-6 pro-growth effects and promotes autophagy-mediated cancer cell dormancy in 3D ovarian cancer: role of miR-1305 and of its target ARH-I. Cancers (Basel) 2022;14:2142.
    Pubmed KoreaMed CrossRef
  27. Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest 2008;118:3917-29.
    Pubmed KoreaMed CrossRef
  28. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov 2022;12:31-46.
    Pubmed CrossRef
  29. Vyas D, Laput G, Vyas AK. Chemotherapy-enhanced inflammation may lead to the failure of therapy and metastasis. Onco Targets Ther 2014;7:1015-23.
    Pubmed KoreaMed CrossRef
  30. Antoniou P, Ziogas DE, Mitsis M, Roukos DH. Precision oncology in patients with nonmetastatic disease: emerging reality or illusion. Future Oncol 2019;15:1805-10.
    Pubmed CrossRef
  31. Prasad V. Perspective: the precision-oncology illusion. Nature 2016;537:S63.
    Pubmed CrossRef
  32. Mittra I, Pal K, Pancholi N, Shaikh A, Rane B, Tidke P, et al. Prevention of chemotherapy toxicity by agents that neutralize or degrade cell-free chromatin. Ann Oncol 2017;28:2119-27.
    Pubmed CrossRef
  33. Borriello A, Bencivenga D, Caldarelli I, Tramontano A, Borgia A, Pirozzi AV, et al. Resveratrol and cancer treatment: is hormesis a yet unsolved matter? Curr Pharm Des 2013;19:5384-93.
    Pubmed CrossRef
  34. Cottart CH, Nivet-Antoine V, Laguillier-Morizot C, Beaudeux JL. Resveratrol bioavailability and toxicity in humans. Mol Nutr Food Res 2010;54:7-16.
    Pubmed CrossRef
  35. Fang J, Cai C, Wang Q, Lin P, Zhao Z, Cheng F. Systems pharmacology-based discovery of natural products for precision oncology through targeting cancer mutated genes. CPT Pharmacometrics Syst Pharmacol 2017;6:177-87.
    Pubmed KoreaMed CrossRef
  36. Gescher AJ, Steward WP. Relationship between mechanisms, bioavailibility, and preclinical chemopreventive efficacy of resveratrol: a conundrum. Cancer Epidemiol Biomarkers Prev 2003;12:953-7.
  37. Berman AY, Motechin RA, Wiesenfeld MY, Holz MK. The therapeutic potential of resveratrol: a review of clinical trials. NPJ Precis Oncol 2017;1:35.
    Pubmed KoreaMed CrossRef
  38. Cai H, Scott E, Kholghi A, Andreadi C, Rufini A, Karmokar A, et al. Cancer chemoprevention: evidence of a nonlinear dose response for the protective effects of resveratrol in humans and mice. Sci Transl Med 2015;7:298ra117.
    Pubmed KoreaMed CrossRef
  39. Zhu W, Qin W, Zhang K, Rottinghaus GE, Chen YC, Kliethermes B, et al. Trans-resveratrol alters mammary promoter hypermethylation in women at increased risk for breast cancer. Nutr Cancer 2012;64:393-400.
    Pubmed KoreaMed CrossRef
  40. Goldberg DM, Yan J, Soleas GJ. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin Biochem 2003;36:79-87.
    Pubmed CrossRef
  41. Wang P, Shang R, Ma Y, Wang D, Zhao W, Chen F, et al. Targeting microbiota-host interactions with resveratrol on cancer: effects and potential mechanisms of action. Crit Rev Food Sci Nutr 2024;64:311-33.
    Pubmed CrossRef

Share this article on :

Related articles in JCP

Most KeyWord