PP Mechanisms of Action as an Anti-Cancer Agent: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Pyrvinium, a lipophilic cation belonging to the cyanine dye family, has been used in the clinic as a safe and effective anthelminthic for over 70 years. Its structure, similar to some polyaminopyrimidines and mitochondrial-targeting peptoids, has been linked with mitochondrial localization and targeting. Unsurprisingly for a small-molecule compound, pyrvinium pamoate (PP) has been shown to act through multiple mechanisms. The majority of the published articles assessing PP specifically as an anticancer therapeutic have focused on two main MOA, which appear to be at the root of PP action: inhibition of the WNT pathway and inhibition of mitochondrial function.

  • pyrvinium
  • cancer
  • mitochondria
  • drug repurposing

1. WNT Signaling

The WNT/β-catenin signaling pathway is one of the essential pathways regulating the cell differentiation, development, and self-renewal of stem cells. As such, it has also been tightly associated with key events in cancer initiation and progression [1][2][3][4][5][6][7][8][9][10]. In 2010, Thorne et al. [11] found that PP inhibited the WNT signaling pathway at low nanomolar concentrations (in vitro and in vivo) in Xenopus laevis and in human colon cancer cells. They determined that PP bound and activated Casein kinase 1α (CK1α), thereby promoting the degradation of β-catenin, the primary effector of the WNT pathway. Supporting this work, Cui et al. [12], Li et al. [13], and Shen et al. [14] have all subsequently demonstrated that PP targeted the WNT pathway in a CK1α-dependent fashion in various cancer models. However, other studies have contested this mechanism and have suggested that PP acts through the upstream inhibition of the Akt/GSK-3β/β-catenin pathway [15][16].
In addition to pre-clinical studies assessing PP as a monotherapy in colon cancer [17][18], PP was found to act as a chemoprotective agent in the formation of intestinal polyps in an APCmin mouse model, presumably through its inhibition of the WNT pathway [19]. PP was also shown to synergize with the KRAS inhibitor salirasib in a WNT-dependent manner, describing a potentially actionable targeted combination of PP and salirasib in KRAS-driven colon cancers [20].

2. Mitochondrial Inhibition

Like many other cyanine dyes, and similar to mitochondrial-targeting peptoids [21], pyrvinium is a lipophilic cation, which allows it to preferentially target the mitochondria. Historically, the activity of PP as an anti-mitochondrial agent was linked to its efficacy at inhibiting protozoa including the plasmodium species associated with malaria [22][23][24]. To an extent, this was also linked to pyrvinium’s ability to inhibit various fungi [25][26]. From an evolutionary standpoint, the endosymbiotic theory holds that mitochondria were generated in a progenitor eukaryotic cell through endosymbiosis and that mitochondria maintain many similarities with bacteria [27]. Unsurprisingly, multiple antibiotics have displayed significant efficacy against cancer cells through inhibiting the mitochondria [28][29][30][31]. PP, interestingly, was first found to be an anti-mitochondrial agent, and only subsequently found to show significant activity against bacteria including Mycobacterium tuberculosis [22][32][33].
Multiple studies have implicated mitochondrial inhibition as a key factor in the activity of PP against cancer cells. Compelling support for this MOA lies in the facts that PP activity is increased in nutrient-poor microenvironments [34][35], and that mitochondrial DNA-depleted cell lines have shown significant resistance to PP therapy [36][37]. Several mechanisms of action have been suggested to explain the PP-dependent mitochondrial inhibition. A frequently proposed mechanism revolves around the inhibition of the mitochondrial respiratory complex I, with data showing PP doses of 0.1–1μM significantly inhibited complex I enzymatic activity, resulting in decreased ATP production and the increased production of reactive oxygen species (ROS) [37][38]. Tomitsuka et al. [39] have also shown that PP treatment of cancer cells (Panc-1, DLD-1, HepG2) resulted in the inhibition of the NADH–fumarate reductase system, a system primarily associated with cancer metabolism in dually hypoxic and hypoglycemic conditions, and increased activity of complex II (succinate–ubiquinone reductase).
In a recent study by Schultz et al. [35], a new MOA for PP-mediated mitochondrial inhibition was proposed. In the study, PP was shown to result in the significant depletion of mitochondrially encoded RNA transcripts and the downregulation of electron transport chain proteins. Here it was found that PP, similar to other G-quadruplex binding agents, bound G-quadruplex structures, which are over-represented in mitochondrial DNA and profoundly inhibited global mitochondrial transcription [40][41].
Overall, it is likely that all of these actions can occur with PP treatment. Acute treatment of PP will lead to the inhibition of complex I and the electron transport chain in normoxic conditions and the inhibition of the NADH–fumarate reductase system in hypoxic conditions. However, prolonged treatment will cause more global dysregulation of mitochondrial function through the inhibition of mitochondrial transcription in all conditions.

3. Tumor Stemness

An important feature of tumor cell population self-renewal and survival after chemotherapy treatment is through a specialized sub-population of cancer-initiating cells or “cancer stem cells” [42][43][44]. Interestingly, a common MOA attributed to the anti-cancer effect of PP is the inhibition of cancer stem cells. Xu et al. [45] found that PP targeted tumor-initiating cells and impaired tumor self-renewal in breast cancer. In vivo PP treatment slowed tumor growth, decreased lung metastasis, and synergized with radiotherapy. Zhang et al. have shown the inhibitory effects of pyrvinium pamoate (PP) on lung cancer stem cells in vitro [46][47]. Similar studies have found PP to inhibit tumor stemness in multiple cancer types including melanoma, leukemia, glioblastoma, and cancers of the prostate, pancreas, lung, ovary, and breast [36][48][49][50]. Interestingly, the inhibition of cancer stem and initiating cells appears to be derivative of PP’s inhibition of the WNT [48] and mitochondrial pathways [36][49], both of which are specifically critical for cancer initiating and stem cells. Venugopal et al. showed that CD133high and CD133-overexpressed cells displayed increased WNT signaling and were highly sensitive to PP treatment [48]. However, Xiang et al. [36] noted that although pyrvinium treatment showed selectivity to blast-phase chronic myeloid leukemia CD34+ progenitor cells, CK1α depletion did not affect PP sensitivity and WNT overexpression failed to rescue these cells. Instead, PP was shown to mainly accumulate in the mitochondria and the creation of chronic myeloid leukemia (CML) ρ0 cells that lacked mitochondrial DNA caused resistance to PP. Similar references to a mitochondrial-driven targeting of cancer stem cells were made by Datta et al. [49] in their assessment of PP in glioma-like stem cells. Recently, Dattilo et al. [51] showed that PP targeted triple-negative breast cancer stem cells through the inhibition of lipid anabolism, resulting in the increased death of cancer stem cells. Additionally, it seems as though the inhibition of stemness is not limited to cancer, but is also apparent in dysplastic tissues—as was described by Min et al. [52], showing that PP can inhibit Trop2+/CD133+/CD166+ dysplastic gastric mucosa stem cells in mouse and human organoids.
Interestingly, PP has been shown to promote wound repair, inhibit fibrotic tissue development [53][54][55], and protect from ischemic injury and promote healing [56][57]. This appears to be directly related to improved stem cell survival and the inhibition of inappropriate lineage commitment [58], which appears contradictory to the known efficacy of PP against cancer progenitor and stem cells [36][46][50]. However, it has been demonstrated that PP preferentially targets cancer progenitor cells compared to non-malignant normal progenitor cells [36]. This is likely due to the altered metabolism and reliance of cancer stem cells on mitochondrial-related pathways including oxidative phosphorylation [36][59] and lipid metabolism [51][60].

4. ELAVL1/HuR Inhibition

ELAVL1/HuR is an RNA-binding protein that has been shown to be an important post-transcriptional regulator of many cancer-associated pro-survival genes such as VEGF, WEE1, and IDH1 [61][62][63][64][65]. Its mechanism, at least in part, is associated with the nucleocytoplasmic translocation and stabilization of 3′UTR of target mRNA transcripts [66][67]. Pyrvinium pamoate has been described by Goa et al. [68] as a functional HuR inhibitor that shifts the HuR cytoplasmic/nuclear equilibrium in favor of the nuclear import of HuR by blocking HuR nucleo-cytoplasmic translocation through inhibiting the checkpoint kinase1/cyclin-dependent kinase 1 pathway and activating the AMP-activated kinase/importin α1 cascade.
It is important to note that a published byproduct of PP’s inhibition of mitochondrial function and ATP production is a relative increase in AMP and in AMP-related signaling, which raises the possibility that PP is not a direct activator of AMP-activated kinase/importin α1. Similarly, other studies have shown that the CRISPR knockout of HuR failed to rescue cancer cells from PP-induced cytotoxicity [35].

5. Androgen Receptor Inhibition

In 2008, PP was identified as a novel androgen inhibitor based on a high-throughput FRET screen [69]. It was shown to be a potent anti-prostate cancer agent that inhibited the function of androgen receptor (AR) splice variants, potentially through interaction with the AR DNA-binding domain (DBD), and the inhibition of several splicing factors (such as DDX17) [70][71][72]. Its DNA-binding domain interaction was thought to occur as a result of binding at the interface of the DBD dimer and the minor groove of the AR response element [72]. However, as noted by Li et al., PP treatment was toxic to LNCaP cells and AR-negative PC3 cells even at concentrations lower than the AR IC50, which is suggestive that PP’s anti prostate cancer efficacy was mediated through a non-AR-related mechanism [73].

6. Unfolded Protein Response

Cells, and cancer cells in particular, undergo physiological processes that require a high rate of protein production and degradation. During protein production, unfolded or misfolded proteins can accumulate in the endoplasmic reticulum, resulting in increased cellular stress (i.e., ER stress). ER stress can also be increased upon physiological, environmental, or pharmacological insults. The unfolded protein response (UPR) is an adaptive cellular survival mechanism that regulates the cell’s handling of ER stress. In routine function, the ER stress signaling activates the unfolded protein response to reduce the volume of unfolded protein load and to help maintain normal cell function and metabolism [74]. However, chronic or extreme ER stress can trigger cell death by apoptosis and has been linked to many diseases such as cancer, diabetes, and neurodegenerative disorders [75][76][77][78][79].
Yu et al. [80] have shown that after hypoglycemia induced UPR, PP suppressed the transcriptional activation of UPR-regulated apoptosis and autophagy targets including XBP-1 and ATF-4 [80][81] along with inhibiting the induction of UPR transcription factors GRP78 (HSPA5) and GRP94 (HSP90B1). Interestingly PP did not inhibit the UPR when it was initiated by non-metabolic agents [80], potentially indicating that the inhibition of the UPR is secondary to the mitochondrial and metabolic dysregulation associated with PP treatment. Another aspect that is yet to be assessed is the impact of PP on the mitochondrial unfolded protein response (UPRmt), a crucial pathway for the maintenance of mitochondrial homeostasis [82].

7. Attenuation of Hedgehog Signaling

The hedgehog (SHH) signaling pathway is one of the key developmental pathways regulating cell proliferation, differentiation, and stem cell renewal [83]. Not surprisingly, the SHH pathway has been shown to play a key role in multiple cancer types [84]. Li et al. [85] were the first to describe PP as an inhibitor of the SHH pathway, and determined that PP treatment decreased the expression of downstream targets of the SHH pathway including Patched1 and Gli-family transcription factors in vitro and in vivo. Subsequently, El-Derhany et al. [86] noted that PP caused the dual inhibition of the WNT and SHH pathways. However, as a specific SHH target has yet to be identified, it is possible that due to the reciprocal coregulation between the WNT and SHH pathways that the inhibition of SHH is secondary to PP’s effects on the WNT pathway [87].

8. Inhibition of PD-1/PDL-1 Interaction

Immunotherapies, and PD-1/PDL-1 inhibitors specifically, have revolutionized many fields of cancer therapy. PDL-1 (CD274)/PD1 (CD279) is an important modulatory/inhibitory immune checkpoint mechanism, utilized by some cancers to target and suppress the adaptive immune response [88]. Upon the binding of PDL-1, a transmembrane ligand, to its immune cell receptor PD-1 (which is highly expressed in activated T-cells), an inhibitory signaling cascade results in decreased T-cell proliferation, the induction of apoptosis in antigen-specific T-cells, and the inhibition of apoptosis in regulatory T-cells [88][89]. In a recent paper by Fattakhova et al. [90], a focused structure- and ligand-based drug library screen was employed that identified PP as an effective blocker of the PD-1/PDL-1 interaction. Of note, they found the IC50 for PP inhibition of PD-1/PDL-1 to be ∼30 μM, which is a promising starting point for PP to serve as a lead compound for the development of more potent inhibitors. However, since PP inhibits mitochondria, the WNT pathway and cancer cell viability at concentrations that are orders of magnitude lower than the noted IC50 for PD-1/PDL-1 inhibition, this immunoactive mechanism is likely not a relevant MOA for PP.

This entry is adapted from the peer-reviewed paper 10.3390/biomedicines10123249

References

  1. Luu, H.H.; Zhang, R.; Haydon, R.C.; Rayburn, E.; Kang, Q.; Si, W.; Park, J.K.; Wang, H.; Peng, Y.; Jiang, W.; et al. Wnt/beta-catenin signaling pathway as a novel cancer drug target. Curr. Cancer Drug Targets 2004, 4, 653–671.
  2. Jansson, E.A.; Are, A.; Greicius, G.; Kuo, I.C.; Kelly, D.; Arulampalam, V.; Pettersson, S. The Wnt/beta-catenin signaling pathway targets PPARgamma activity in colon cancer cells. Proc. Natl. Acad. Sci. USA 2005, 102, 1460–1465.
  3. Jiang, Y.G.; Luo, Y.; He, D.L.; Li, X.; Zhang, L.L.; Peng, T.; Li, M.C.; Lin, Y.H. Role of Wnt/beta-catenin signaling pathway in epithelial-mesenchymal transition of human prostate cancer induced by hypoxia-inducible factor-1alpha. Int. J. Urol. 2007, 14, 1034–1039.
  4. Yeh, C.T.; Yao, C.J.; Yan, J.L.; Chuang, S.E.; Lee, L.M.; Chen, C.M.; Yeh, C.F.; Li, C.H.; Lai, G.M. Apoptotic Cell Death and Inhibition of Wnt/beta-Catenin Signaling Pathway in Human Colon Cancer Cells by an Active Fraction (HS7) from Taiwanofungus camphoratus. Evid. Based Complement. Altern. Med. 2011, 2011, 750230.
  5. King, T.D.; Suto, M.J.; Li, Y. The Wnt/beta-catenin signaling pathway: A potential therapeutic target in the treatment of triple negative breast cancer. J. Cell Biochem. 2012, 113, 13–18.
  6. Abdel-Magid, A.F. Wnt/beta-Catenin Signaling Pathway Inhibitors: A Promising Cancer Therapy. ACS Med. Chem. Lett. 2014, 5, 956–957.
  7. Gedaly, R.; Galuppo, R.; Daily, M.F.; Shah, M.; Maynard, E.; Chen, C.; Zhang, X.; Esser, K.A.; Cohen, D.A.; Evers, B.M.; et al. Targeting the Wnt/beta-catenin signaling pathway in liver cancer stem cells and hepatocellular carcinoma cell lines with FH535. PLoS ONE 2014, 9, e99272.
  8. Vatansever, H.S.; Gumus, B.; Aydogdu, O.; Sivrikoz, O.N.; Turkoz-Uluer, E.; Kivanc, M.; Atesci, Y.Z.; Bugdayci, H. The role of stem/progenitor cells and Wnt/beta-catenin signaling pathway in the patients with prostate cancer. Minerva Urol. Nefrol. 2014, 66, 249–255.
  9. Wang, R.; Sun, Q.; Wang, P.; Liu, M.; Xiong, S.; Luo, J.; Huang, H.; Du, Q.; Geller, D.A.; Cheng, B. Notch and Wnt/beta-catenin signaling pathway play important roles in activating liver cancer stem cells. Oncotarget 2016, 7, 5754–5768.
  10. Zhang, Q.; Meng, X.K.; Wang, W.X.; Zhang, R.M.; Zhang, T.; Ren, J.J. The Wnt/beta-catenin signaling pathway mechanism for pancreatic cancer chemoresistance in a three-dimensional cancer microenvironment. Am. J. Transl. Res. 2016, 8, 4490–4498.
  11. Thorne, C.A.; Hanson, A.J.; Schneider, J.; Tahinci, E.; Orton, D.; Cselenyi, C.S.; Jernigan, K.K.; Meyers, K.C.; Hang, B.I.; Waterson, A.G.; et al. Small-molecule inhibition of Wnt signaling through activation of casein kinase 1alpha. Nat. Chem. Biol. 2010, 6, 829–836.
  12. Cui, L.; Zhao, J.; Liu, J. Pyrvinium Sensitizes Clear Cell Renal Cell Carcinoma Response to Chemotherapy Via Casein Kinase 1alpha-Dependent Inhibition of Wnt/beta-Catenin. Am. J. Med. Sci. 2018, 355, 274–280.
  13. Li, J.; Jiang, Y.; Wang, P.; Ke, S.; Yang, L.; Shen, Y. Casein kinase 1alpha-dependent inhibition of Wnt/beta-catenin selectively targets nasopharyngeal carcinoma and increases chemosensitivity. Anticancer Drugs 2019, 30, e0747.
  14. Shen, C.; Li, B.; Astudillo, L.; Deutscher, M.P.; Cobb, M.H.; Capobianco, A.J.; Lee, E.; Robbins, D.J. The CK1alpha Activator Pyrvinium Enhances the Catalytic Efficiency (kcat/Km) of CK1alpha. Biochemistry 2019, 58, 5102–5106.
  15. Zheng, L.; Liu, Y.; Pan, J. Inhibitory effect of pyrvinium pamoate on uveal melanoma cells involves blocking of Wnt/beta-catenin pathway. Acta Biochim. Biophys. Sin. 2017, 49, 890–898.
  16. Venerando, A.; Girardi, C.; Ruzzene, M.; Pinna, L.A. Pyrvinium pamoate does not activate protein kinase CK1, but promotes Akt/PKB down-regulation and GSK3 activation. Biochem. J. 2013, 452, 131–137.
  17. Wiegering, A.; Uthe, F.W.; Huttenrauch, M.; Muhling, B.; Linnebacher, M.; Krummenast, F.; Germer, C.T.; Thalheimer, A.; Otto, C. The impact of pyrvinium pamoate on colon cancer cell viability. Int. J. Colorectal. Dis. 2014, 29, 1189–1198.
  18. Zheng, W.; Hu, J.; Lv, Y.; Bai, B.; Shan, L.; Chen, K.; Dai, S.; Zhu, H. Pyrvinium pamoate inhibits cell proliferation through ROS-mediated AKT-dependent signaling pathway in colorectal cancer. Med. Oncol. 2021, 38, 21.
  19. Li, B.; Flaveny, C.A.; Giambelli, C.; Fei, D.L.; Han, L.; Hang, B.I.; Bai, F.; Pei, X.H.; Nose, V.; Burlingame, O.; et al. Repurposing the FDA-approved pinworm drug pyrvinium as a novel chemotherapeutic agent for intestinal polyposis. PLoS ONE 2014, 9, e101969.
  20. Mologni, L.; Brussolo, S.; Ceccon, M.; Gambacorti-Passerini, C. Synergistic effects of combined Wnt/KRAS inhibition in colorectal cancer cells. PLoS ONE 2012, 7, e51449.
  21. Nam, H.Y.; Hong, J.A.; Choi, J.; Shin, S.; Cho, S.K.; Seo, J.; Lee, J. Mitochondria-Targeting Peptoids. Bioconjug. Chem. 2018, 29, 1669–1676.
  22. Gaikwad, V.R.; Karale, U.B.; Govindarajalu, G.; Adhikari, N.; Krishna, E.V.; Krishna, V.S.; Misra, S.; Sriram, D.; Sijwali, P.S.; Rode, H.B. Synthesis and efficacy of pyrvinium-inspired analogs against tuberculosis and malaria pathogens. Bioorg. Med. Chem. Lett. 2020, 30, 127037.
  23. Teguh, S.C.; Klonis, N.; Duffy, S.; Lucantoni, L.; Avery, V.M.; Hutton, C.A.; Baell, J.B.; Tilley, L. Novel conjugated quinoline-indoles compromise Plasmodium falciparum mitochondrial function and show promising antimalarial activity. J. Med. Chem. 2013, 56, 6200–6215.
  24. Downey, A.S.; Chong, C.R.; Graczyk, T.K.; Sullivan, D.J. Efficacy of pyrvinium pamoate against Cryptosporidium parvum infection in vitro and in a neonatal mouse model. Antimicrob. Agents Chemother. 2008, 52, 3106–3112.
  25. Simm, C.; Weerasinghe, H.; Thomas, D.R.; Harrison, P.F.; Newton, H.J.; Beilharz, T.H.; Traven, A. Disruption of Iron Homeostasis and Mitochondrial Metabolism Are Promising Targets to Inhibit Candida auris. Microbiol. Spectr. 2022, 10, e0010022.
  26. Sun, Y.; Gao, L.; Zhang, Y.; Yang, J.; Zeng, T. Synergistic Effect of Pyrvinium Pamoate and Azoles Against Aspergillus fumigatus in vitro and in vivo. Front. Microbiol. 2020, 11, 579362.
  27. Boguszewska, K.; Szewczuk, M.; Kazmierczak-Baranska, J.; Karwowski, B.T. The Similarities between Human Mitochondria and Bacteria in the Context of Structure, Genome, and Base Excision Repair System. Molecules 2020, 25, 2857.
  28. Aminzadeh-Gohari, S.; Weber, D.D.; Vidali, S.; Catalano, L.; Kofler, B.; Feichtinger, R.G. From old to new—Repurposing drugs to target mitochondrial energy metabolism in cancer. Semin. Cell Dev. Biol. 2020, 98, 211–223.
  29. Wu, X.; Li, F.; Wang, X.; Li, C.; Meng, Q.; Wang, C.; Huang, J.; Chen, S.; Zhu, Z. Antibiotic bedaquiline effectively targets growth, survival and tumor angiogenesis of lung cancer through suppressing energy metabolism. Biochem. Biophys. Res. Commun. 2018, 495, 267–272.
  30. Tian, F.; Wang, C.; Tang, M.; Li, J.; Cheng, X.; Zhang, S.; Ji, D.; Huang, Y.; Li, H. The antibiotic chloramphenicol may be an effective new agent for inhibiting the growth of multiple myeloma. Oncotarget 2016, 7, 51934–51942.
  31. Tan, Q.; Yan, X.; Song, L.; Yi, H.; Li, P.; Sun, G.; Yu, D.; Li, L.; Zeng, Z.; Guo, Z. Induction of Mitochondrial Dysfunction and Oxidative Damage by Antibiotic Drug Doxycycline Enhances the Responsiveness of Glioblastoma to Chemotherapy. Med. Sci. Monit. 2017, 23, 4117–4125.
  32. Niu, H.; Yee, R.; Cui, P.; Tian, L.; Zhang, S.; Shi, W.; Sullivan, D.; Zhu, B.; Zhang, W.; Zhang, Y. Identification of Agents Active against Methicillin-Resistant Staphylococcus aureus USA300 from a Clinical Compound Library. Pathogens 2017, 6, 44.
  33. Guan, Q.; Zhan, L.; Liu, Z.H.; Pan, Q.; Chen, X.L.; Xiao, Z.; Qin, C.; Zhang, X.L. Identification of pyrvinium pamoate as an anti-tuberculosis agent in vitro and in vivo by SOSA approach amongst known drugs. Emerg. Microbes Infect. 2020, 9, 302–312.
  34. Esumi, H.; Lu, J.; Kurashima, Y.; Hanaoka, T. Antitumor activity of pyrvinium pamoate, 6-(dimethylamino)-2--1-methyl-qu inolinium pamoate salt, showing preferential cytotoxicity during glucose starvation. Cancer Sci. 2004, 95, 685–690.
  35. Schultz, C.W.; McCarthy, G.A.; Nerwal, T.; Nevler, A.; DuHadaway, J.B.; McCoy, M.D.; Jiang, W.; Brown, S.Z.; Goetz, A.; Jain, A.; et al. The FDA-Approved Anthelmintic Pyrvinium Pamoate Inhibits Pancreatic Cancer Cells in Nutrient-Depleted Conditions by Targeting the Mitochondria. Mol. Cancer Ther. 2021, 20, 2166–2176.
  36. Xiang, W.; Cheong, J.K.; Ang, S.H.; Teo, B.; Xu, P.; Asari, K.; Sun, W.T.; Than, H.; Bunte, R.M.; Virshup, D.M.; et al. Pyrvinium selectively targets blast phase-chronic myeloid leukemia through inhibition of mitochondrial respiration. Oncotarget 2015, 6, 33769–33780.
  37. Harada, Y.; Ishii, I.; Hatake, K.; Kasahara, T. Pyrvinium pamoate inhibits proliferation of myeloma/erythroleukemia cells by suppressing mitochondrial respiratory complex I and STAT3. Cancer Lett. 2012, 319, 83–88.
  38. Xiao, M.; Zhang, L.; Zhou, Y.; Rajoria, P.; Wang, C. Pyrvinium selectively induces apoptosis of lymphoma cells through impairing mitochondrial functions and JAK2/STAT5. Biochem. Biophys. Res. Commun. 2016, 469, 716–722.
  39. Tomitsuka, E.; Kita, K.; Esumi, H. An anticancer agent, pyrvinium pamoate inhibits the NADH-fumarate reductase system--a unique mitochondrial energy metabolism in tumour microenvironments. J. Biochem. 2012, 152, 171–183.
  40. Falabella, M.; Fernandez, R.J.; Johnson, F.B.; Kaufman, B.A. Potential Roles for G-Quadruplexes in Mitochondria. Curr. Med. Chem. 2019, 26, 2918–2932.
  41. Falabella, M.; Kolesar, J.E.; Wallace, C.; de Jesus, D.; Sun, L.; Taguchi, Y.V.; Wang, C.; Wang, T.; Xiang, I.M.; Alder, J.K.; et al. G-quadruplex dynamics contribute to regulation of mitochondrial gene expression. Sci. Rep. 2019, 9, 5605.
  42. Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284.
  43. Haraguchi, N.; Inoue, H.; Tanaka, F.; Mimori, K.; Utsunomiya, T.; Sasaki, A.; Mori, M. Cancer stem cells in human gastrointestinal cancers. Hum. Cell 2006, 19, 24–29.
  44. Collins, A.T.; Berry, P.A.; Hyde, C.; Stower, M.J.; Maitland, N.J. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005, 65, 10946–10951.
  45. Xu, W.; Lacerda, L.; Debeb, B.G.; Atkinson, R.L.; Solley, T.N.; Li, L.; Orton, D.; McMurray, J.S.; Hang, B.I.; Lee, E.; et al. The antihelmintic drug pyrvinium pamoate targets aggressive breast cancer. PLoS ONE 2013, 8, e71508.
  46. Zhang, X.; Lou, Y.; Zheng, X.; Wang, H.; Sun, J.; Dong, Q.; Han, B. Wnt blockers inhibit the proliferation of lung cancer stem cells. Drug Des. Dev. Ther. 2015, 9, 2399–2407.
  47. Zhang, X.; Zheng, X.; Lou, Y.; Wang, H.; Xu, J.; Zhang, Y.; Han, B. β-catenin inhibitors suppress cells proliferation and promote cells apoptosis in PC9 lung cancer stem cells. Int. J. Clin. Exp. Pathol. 2017, 10, 11968–11978.
  48. Venugopal, C.; Hallett, R.; Vora, P.; Manoranjan, B.; Mahendram, S.; Qazi, M.A.; McFarlane, N.; Subapanditha, M.; Nolte, S.M.; Singh, M.; et al. Pyrvinium Targets CD133 in Human Glioblastoma Brain Tumor-Initiating Cells. Clin. Cancer Res. 2015, 21, 5324–5337.
  49. Datta, S.; Sears, T.; Cortopassi, G.; Woolard, K.; Angelastro, J.M. Repurposing FDA approved drugs inhibiting mitochondrial function for targeting glioma-stem like cells. Biomed. Pharmacother. 2021, 133, 111058.
  50. Lamb, R.; Ozsvari, B.; Lisanti, C.L.; Tanowitz, H.B.; Howell, A.; Martinez-Outschoorn, U.E.; Sotgia, F.; Lisanti, M.P. Antibiotics that target mitochondria effectively eradicate cancer stem cells, across multiple tumor types: Treating cancer like an infectious disease. Oncotarget 2015, 6, 4569–4584.
  51. Dattilo, R.; Mottini, C.; Camera, E.; Lamolinara, A.; Auslander, N.; Doglioni, G.; Muscolini, M.; Tang, W.; Planque, M.; Ercolani, C.; et al. Pyrvinium Pamoate Induces Death of Triple-Negative Breast Cancer Stem-Like Cells and Reduces Metastases through Effects on Lipid Anabolism. Cancer Res. 2020, 80, 4087–4102.
  52. Min, J.; Zhang, C.; Bliton, R.J.; Caldwell, B.; Caplan, L.; Presentation, K.S.; Park, D.J.; Kong, S.H.; Lee, H.S.; Washington, M.K.; et al. Dysplastic Stem Cell Plasticity Functions as a Driving Force for Neoplastic Transformation of Precancerous Gastric Mucosa. Gastroenterology 2022, 163, 875–890.
  53. Cuevas, C.A.; Tapia-Rojas, C.; Cespedes, C.; Inestrosa, N.C.; Vio, C.P. beta-Catenin-Dependent Signaling Pathway Contributes to Renal Fibrosis in Hypertensive Rats. Biomed. Res. Int. 2015, 2015, 726012.
  54. Cuevas, C.A.; Gonzalez, A.A.; Inestrosa, N.C.; Vio, C.P.; Prieto, M.C. Angiotensin II increases fibronectin and collagen I through the beta-catenin-dependent signaling in mouse collecting duct cells. Am. J. Physiol. Renal. Physiol. 2015, 308, F358–F365.
  55. Jeon, K.I.; Phipps, R.P.; Sime, P.J.; Huxlin, K.R. Antifibrotic Actions of Peroxisome Proliferator-Activated Receptor gamma Ligands in Corneal Fibroblasts Are Mediated by beta-Catenin-Regulated Pathways. Am. J. Pathol. 2017, 187, 1660–1669.
  56. Bastakoty, D.; Saraswati, S.; Cates, J.; Lee, E.; Nanney, L.B.; Young, P.P. Inhibition of Wnt/beta-catenin pathway promotes regenerative repair of cutaneous and cartilage injury. FASEB J. 2015, 29, 4881–4892.
  57. Saraswati, S.; Alfaro, M.P.; Thorne, C.A.; Atkinson, J.; Lee, E.; Young, P.P. Pyrvinium, a potent small molecule Wnt inhibitor, promotes wound repair and post-MI cardiac remodeling. PLoS ONE 2010, 5, e15521.
  58. Saraswati, S.; Deskins, D.L.; Holt, G.E.; Young, P.P. Pyrvinium, a potent small molecule Wnt inhibitor, increases engraftment and inhibits lineage commitment of mesenchymal stem cells (MSCs). Wound Repair Regen. 2012, 20, 185–193.
  59. Valle, S.; Alcala, S.; Martin-Hijano, L.; Cabezas-Sainz, P.; Navarro, D.; Munoz, E.R.; Yuste, L.; Tiwary, K.; Walter, K.; Ruiz-Canas, L.; et al. Exploiting oxidative phosphorylation to promote the stem and immunoevasive properties of pancreatic cancer stem cells. Nat. Commun. 2020, 11, 5265.
  60. Li, H.; Feng, Z.; He, M.L. Lipid metabolism alteration contributes to and maintains the properties of cancer stem cells. Theranostics 2020, 10, 7053–7069.
  61. Blanco, F.F.; Jimbo, M.; Wulfkuhle, J.; Gallagher, I.; Deng, J.; Enyenihi, L.; Meisner-Kober, N.; Londin, E.; Rigoutsos, I.; Sawicki, J.A.; et al. The mRNA-binding protein HuR promotes hypoxia-induced chemoresistance through posttranscriptional regulation of the proto-oncogene PIM1 in pancreatic cancer cells. Oncogene 2016, 35, 2529–2541.
  62. Kurosu, T.; Ohga, N.; Hida, Y.; Maishi, N.; Akiyama, K.; Kakuguchi, W.; Kuroshima, T.; Kondo, M.; Akino, T.; Totsuka, Y.; et al. HuR keeps an angiogenic switch on by stabilising mRNA of VEGF and COX-2 in tumour endothelium. Br. J. Cancer 2011, 104, 819–829.
  63. Burkhart, R.A.; Pineda, D.M.; Chand, S.N.; Romeo, C.; Londin, E.R.; Karoly, E.D.; Cozzitorto, J.A.; Rigoutsos, I.; Yeo, C.J.; Brody, J.R.; et al. HuR is a post-transcriptional regulator of core metabolic enzymes in pancreatic cancer. RNA Biol. 2013, 10, 1312–1323.
  64. Lal, S.; Burkhart, R.A.; Beeharry, N.; Bhattacharjee, V.; Londin, E.R.; Cozzitorto, J.A.; Romeo, C.; Jimbo, M.; Norris, Z.A.; Yeo, C.J.; et al. HuR posttranscriptionally regulates WEE1: Implications for the DNA damage response in pancreatic cancer cells. Cancer Res. 2014, 74, 1128–1140.
  65. Zarei, M.; Lal, S.; Parker, S.J.; Nevler, A.; Vaziri-Gohar, A.; Dukleska, K.; Mambelli-Lisboa, N.C.; Moffat, C.; Blanco, F.F.; Chand, S.N.; et al. Posttranscriptional Upregulation of IDH1 by HuR Establishes a Powerful Survival Phenotype in Pancreatic Cancer Cells. Cancer Res. 2017, 77, 4460–4471.
  66. Fan, X.C.; Steitz, J.A. HNS, a nuclear-cytoplasmic shuttling sequence in HuR. Proc. Natl. Acad. Sci. USA 1998, 95, 15293–15298.
  67. Gorospe, M. HuR in the mammalian genotoxic response: Post-transcriptional multitasking. Cell Cycle 2003, 2, 412–414.
  68. Guo, J.; Lv, J.; Chang, S.; Chen, Z.; Lu, W.; Xu, C.; Liu, M.; Pang, X. Inhibiting cytoplasmic accumulation of HuR synergizes genotoxic agents in urothelial carcinoma of the bladder. Oncotarget 2016, 7, 45249–45262.
  69. Jones, J.O.; Diamond, M.I. A cellular conformation-based screen for androgen receptor inhibitors. ACS Chem. Biol. 2008, 3, 412–418.
  70. Jones, J.O.; Bolton, E.C.; Huang, Y.; Feau, C.; Guy, R.K.; Yamamoto, K.R.; Hann, B.; Diamond, M.I. Non-competitive androgen receptor inhibition in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2009, 106, 7233–7238.
  71. Pal, S.K.; Tew, B.Y.; Lim, M.; Stankavich, B.; He, M.; Pufall, M.; Hu, W.; Chen, Y.; Jones, J.O. Mechanistic Investigation of the Androgen Receptor DNA-Binding Domain Inhibitor Pyrvinium. ACS Omega 2019, 4, 2472–2481.
  72. Lim, M.; Otto-Duessel, M.; He, M.; Su, L.; Nguyen, D.; Chin, E.; Alliston, T.; Jones, J.O. Ligand-independent and tissue-selective androgen receptor inhibition by pyrvinium. ACS Chem. Biol. 2014, 9, 692–702.
  73. Li, H.; Ban, F.; Dalal, K.; Leblanc, E.; Frewin, K.; Ma, D.; Adomat, H.; Rennie, P.S.; Cherkasov, A. Discovery of small-molecule inhibitors selectively targeting the DNA-binding domain of the human androgen receptor. J. Med. Chem. 2014, 57, 6458–6467.
  74. Rutkowski, D.T.; Wu, J.; Back, S.H.; Callaghan, M.U.; Ferris, S.P.; Iqbal, J.; Clark, R.; Miao, H.; Hassler, J.R.; Fornek, J.; et al. UPR pathways combine to prevent hepatic steatosis caused by ER stress-mediated suppression of transcriptional master regulators. Dev. Cell 2008, 15, 829–840.
  75. Socha, L.; Silva, D.; Lesage, S.; Goodnow, C.; Petrovsky, N. The role of endoplasmic reticulum stress in nonimmune diabetes: NOD.k iHEL, a novel model of beta cell death. Ann. N. Y. Acad. Sci. 2003, 1005, 178–183.
  76. Harding, H.P.; Ron, D. Endoplasmic reticulum stress and the development of diabetes: A review. Diabetes 2002, 51 (Suppl. S3), S455–S461.
  77. Cabral-Miranda, F.; Tamburini, G.; Martinez, G.; Ardiles, A.O.; Medinas, D.B.; Gerakis, Y.; Hung, M.D.; Vidal, R.; Fuentealba, M.; Miedema, T.; et al. Unfolded protein response IRE1/XBP1 signaling is required for healthy mammalian brain aging. EMBO J. 2022, 41, e111952.
  78. Ajoolabady, A.; Lindholm, D.; Ren, J.; Pratico, D. ER stress and UPR in Alzheimer’s disease: Mechanisms, pathogenesis, treatments. Cell Death Dis. 2022, 13, 706.
  79. Di Domenico, F.; Lanzillotta, C. The disturbance of protein synthesis/degradation homeostasis is a common trait of age-related neurodegenerative disorders. Adv. Protein Chem. Struct. Biol. 2022, 132, 49–87.
  80. Yu, D.H.; Macdonald, J.; Liu, G.; Lee, A.S.; Ly, M.; Davis, T.; Ke, N.; Zhou, D.; Wong-Staal, F.; Li, Q.X. Pyrvinium targets the unfolded protein response to hypoglycemia and its anti-tumor activity is enhanced by combination therapy. PLoS ONE 2008, 3, e3951.
  81. Fu, Y.H.; Tseng, C.Y.; Lu, J.W.; Lu, W.H.; Lan, P.Q.; Chen, C.Y.; Ou, D.L.; Lin, L.I. Deciphering the Role of Pyrvinium Pamoate in the Generation of Integrated Stress Response and Modulation of Mitochondrial Function in Myeloid Leukemia Cells through Transcriptome Analysis. Biomedicines 2021, 9, 1869.
  82. Zhao, Q.; Wang, J.; Levichkin, I.V.; Stasinopoulos, S.; Ryan, M.T.; Hoogenraad, N.J. A mitochondrial specific stress response in mammalian cells. EMBO J. 2002, 21, 4411–4419.
  83. Jiang, J.; Hui, C.C. Hedgehog signaling in development and cancer. Dev. Cell 2008, 15, 801–812.
  84. Taipale, J.; Beachy, P.A. The Hedgehog and Wnt signalling pathways in cancer. Nature 2001, 411, 349–354.
  85. Li, B.; Fei, D.L.; Flaveny, C.A.; Dahmane, N.; Baubet, V.; Wang, Z.; Bai, F.; Pei, X.H.; Rodriguez-Blanco, J.; Hang, B.; et al. Pyrvinium attenuates Hedgehog signaling downstream of smoothened. Cancer Res. 2014, 74, 4811–4821.
  86. El-Derany, M.O.; El-Demerdash, E. Pyrvinium pamoate attenuates non-alcoholic steatohepatitis: Insight on hedgehog/Gli and Wnt/beta-catenin signaling crosstalk. Biochem. Pharmacol. 2020, 177, 113942.
  87. Ding, M.; Wang, X. Antagonism between Hedgehog and Wnt signaling pathways regulates tumorigenicity. Oncol. Lett. 2017, 14, 6327–6333.
  88. Iwai, Y.; Hamanishi, J.; Chamoto, K.; Honjo, T. Cancer immunotherapies targeting the PD-1 signaling pathway. J. Biomed. Sci. 2017, 24, 26.
  89. Salmaninejad, A.; Valilou, S.F.; Shabgah, A.G.; Aslani, S.; Alimardani, M.; Pasdar, A.; Sahebkar, A. PD-1/PD-L1 pathway: Basic biology and role in cancer immunotherapy. J. Cell Physiol. 2019, 234, 16824–16837.
  90. Fattakhova, E.; Hofer, J.; DiFlumeri, J.; Cobb, M.; Dando, T.; Romisher, Z.; Wellington, J.; Oravic, M.; Radnoff, M.; Patil, S.P. Identification of the FDA-Approved Drug Pyrvinium as a Small-Molecule Inhibitor of the PD-1/PD-L1 Interaction. ChemMedChem 2021, 16, 2769–2774.
More
This entry is offline, you can click here to edit this entry!
Video Production Service