CX-4945

Flavones and Flavonols May Have Clinical Potential as CK2 Inhibitors in Cancer Therapy

Mark F. McCarty1, Simon Iloki Assanga2 and Lidianys Lewis Lujan2

Abstract

The serine-threonine kinase CK2, which targets over 300 cellular proteins, is overexpressed in all cancers, presumably reflecting its ability to promote proliferation, spread, and survival through a wide range of complementary mechanisms. Via an activating phosphorylation of Cdc373, a co-chaperone which partners with Hsp90, CK2 prolongs the half-life of protein kinases that promote proliferation and survival in many cancers, including Akt, Src, EGFR, Raf1, and several cyclin-dependent kinases. CK2 works in other ways to boost the activity of signaling pathways that promote cancer aggressiveness and chemoresistance, including those driven by Akt, NF-kappaB, hypoxia-inducible factor-1, beta-catenin, TGF-beta, STAT3, hedgehog, Notch1, and the androgen receptor; it promotes the epidermal-mesenchymal transition and aids efficiency of DNA repair. Several potent and relatively specific inhibitors of CK2 are now being evaluated as potential cancer drugs; CX-4945 has shown impressive activity in cell culture studies and xenograft models, and is now entering clinical trials. Moreover, it has long been recognized that the natural flavone apigenin can inhibit CK2, with a Ki near 1 micromolar; more recent work indicates that a range of flavones and flavonols, characterized by a planar structure and hydroxylations at the 7 and 4’ positions – including apigenin, luteolin keampferol, fisetin, quercetin, and myricetin – can inhibit CK2 with Ki s in the sub-micromolar range. This finding is particularly intriguing in light of the numerous studies demonstrating that each of these agents can inhibit the growth of cancer cells lines in vitro and of human xenografts in nude mice. These studies attribute the cancer-retardant efficacy of flavones/flavonols to impacts on a bewildering array of cellular targets, including those whose activities are boosted by CK2; it is reasonable to suspect that, at least in physiologically achievable concentrations, these agents may be achieving these effects primarily via CK2 inhibition. Inefficient absorption and rapid conjugation limit the bioefficacy of orally administered flavonoids; however, the increased extracellular beta-glucuronidase of many tumors may give tumors privileged access to glucuronidated flavonoids, and nanopartical technology can improve the bioavailability of these agents. Enzymatically modified isoquercitrin has particular promise as a delivery vehicle for quercetin. Hence, it may be worthwhile to explore the clinical potential of flavones/flavonols as

Key words: cancer; CK2; CX-4945; Hsp90; flavones; flavonols; xenografts, beta-glucuronidase; HDA6; sulforaphane

CK2 inhibitors for cancer therapy.

CK2 is Over-expressed and Up-regulates Proliferation, Spread, and Survival in Cancer CK2, a serine-threonine kinase once known as casein kinase 2, is a ubiquitously expressed tetramer comprised of two catalytic subunits – α and/or α’ – and two regulatory β subunits that direct it to specific targets. CK2 is capable of phosphorylating a huge range of cellular proteins; over 300 physiological targets have been documented to date (though ironically casein is not one of them!).1 Its level of expression and its sub-cellular localization determine its activity, as posttranslational modifications or allosteric interactions are thought to have little impact in that regard; moreover, no gain-of-function mutants of this kinase are known.
Virtually all cancer cell lines studied to date overexpress CK2 protein, relative to its expression in normal tissues of origin; moreover, they tend to route a higher proportion of this protein to the cell nucleus.2 This is not likely to be accidental, as high CK2 activity works in a bewildering number of complementary ways to promote cellular proliferation and spread, while suppressing apoptosis and inducing chemoresistance. Hence, cancer cells which overexpress CK2 will tend to be selected for.

CK2 Modulates a Plethora of Signaling Pathways

One of CK2’s most intriguing and ramified effects is to phosphorylate, and thereby activate, the co-chaperone Cdc37.3, 4 Activated Cdc37 interacts with Hsp90 to provide chaperoning activity for a broad range of protein kinases, many of which play a role in promoting cell proliferation and survival. These include Akt, Src, EGFR, PDGFR, Raf-1, IKK, RIP1, Cdc2, Cdk2, Cdk4, and Cdk6. This chaperoning activity tends to slow the proteolytic degradation of these kinases, prolonging their effective half-lives; this activity is particularly crucial for the survival of certain mutant constitutively active forms of these kinases often found in cancers. CK2 is one of the few kinases known to confer activation on Cdc37 – for which reason assessment of Cdc37 phosphorylation at Ser13 has been proposed as a strategy for determining CK2 activity in vivo.5 (Phosphorylation of Akt1 at Ser129 has been found to be a more specific a marker for CK2 activity – and to respond more rapidly to CK2 inhibition.6-9)
But CK2 works in a number of additional ways to boost the activity of signaling pathways that make cancer more aggressive and harder to kill:Akt – While CK2 boosts Akt expression via Hsp90-cdc37-mediated stabilization, it can also work in various complementary ways to increase the phosphorylation and activation of this key kinase, which promotes cellular proliferation while acting in a number of ways to inhibit apoptosis. CK2 phosphorylates Akt directly at Ser129; this up-regulates the activation of Akt mediated by PDK1 and mTORC2, and facilitates its association with Hsp90.10, 11 And CK2 inhibits phosphatase activities that target Akt; it phosphorylates and thereby reduces the activity of the crucial cancer suppressor PTEN, and also promotes proteasomal degradation of PML, a protein which is an obligate component of a nuclear complex that dephosphorylates Akt within the nucleus.12-15 CK2 also has the potential to work upstream from Akt, enhancing its activation by up-regulating certain tyrosine kinase signaling pathways.
NF-kappaB – Numerous studies show that CK2 inhibition suppresses NF-kappaB activity in cancer cell lines, whereas overexpression of this kinase boosts NF-kappaB activity.16-31 CK2 promotes degradation of IkappaB; this can reflect an activating phosphorylation of IKKbeta, as well as a direct phosphorylation of IkappaB that renders it more sensitive to proteolytic cleavage
by calpain.17, 18, 23, 28 CK2 activity also has been reported to somehow boost the expression of IKK-i/IKKepsilon, an alternative IkappaB kinase complex capable of promoting IkappaB degradation.21 And the transcriptional activity of p65 is enhanced by a phosphorylation of Ser529 conferred by CK2.24
Hypoxia-inducible factor-1 (HIF-1) – CK2 enhances the transcriptional activity of HIF-1, even though it doesn’t increase the protein expression or nuclear binding of this factor.32-34 Some evidence suggests that this reflects a reduction of p53 levels; nuclear p53 somehow antagonizes the transcriptional activity of HIF-1.33 CK2’s impact on p53 level, in turn, may reflect phosphorylations of MDM2 that enhance its ability to promote proteasomal degradation of p53.
Beta-Catenin – Many studies show that CK2 inhibition decreases Wnt-beta-catenin signaling.26, 35-43 Activation of Akt, which stabilizes beta-catenin through inhibition of glycogen synthase kinase-3 and also via a direct phosphorylation on Ser552, evidently can contribute to this effect.41, 42 However, CK2 also phosphorylates beta-catenin directly on Thr393, an effect which likewise prolongs the half-life and promotes the transcriptional activity of this factor.36, 37
TGF-beta – Numerous studies have shown that CK2 supports the activity of signaling pathways downstream from TGF-beta, including those mediated by SMAD2/3, Akt, and ERK1. Its direct molecular targets in this regard have not yet been determined.44-47
STAT3 – There are several reports that inhibition of CK2 suppresses STAT3 phosphorylation and activation in cancer cell lines.4, 48, 49 The basis of this effect is not yet clear. In some cell lines, suppression of IL-6 expression may contribute to this effect.
Hedgehog – In human lung cancer cells, CK2 activity has been shown to boost the mRNA and protein expression of Gli1, and to enhance the half-life and transcriptional activity of this key mediator of hedgehog signaling.50 CK2 can directly phosphorylate Gli1, and it has been suggested that this may be responsible for the positive impact of CK2 on hedgehog signaling.
Notch1 – In human lung cancer cell lines expressing Notch1, inhibition of CK2 activity suppresses Notch1-driven transcription, whereas forced overexpression of CK2 has the opposite effect.51 This may reflect the fact that CK2 activity increases the half-life of Notch1 protein.
Androgen Receptor – CK2 inhibitors suppress androgen receptor-mediated transcription in prostate cancer cell lines, at least in part by blocking androgen-induced nuclear translocation of the receptor.52-54 The direct target of CK2 in this effect has not been identified.
Caspase-3 – Whereas CK2 can suppress apoptosis by promoting the activity of various kinases and transcription factors that suppress apoptosis, CK2alpha’ can strike at the very heart of the apoptotic process by conferring a phosphorylation on caspase-3 that prevents its cleavage and activation by upstream caspases.55
DNA Repair – CK2-mediated phosphorylations of XRCC1 and MDC1, nuclear proteins which play a key role in the repair of DNA single-strand and double-strand breaks, respectively, are required for their proper activity.56-61 Hence, inhibition of CK2 can boost the killing activity of DNA-damaging cytotoxins not only by up-regulating mechanisms of apoptosis, but also by impeding the efficiency of DNA repair. Not surprisingly, a number of reports indicate that concurrent CK2 inhibition can boost the sensitivity of various cell lines to chemotherapeutic agents.62-69
Epidermal-Mesenchymal Transition – Studies with CK2 inhibitors demonstrate that CK2 activity can promote the epidermal-mesenchymal transition (EMT) necessary for invasive behavior by boosting expression of vimentin, snail, and smad2/3, while suppressing that of Ecadherin.70-74 For some reason this effect is most prominent in cancer cells which overexpress CK2alpha catalytic subunits, relative to CK2beta regulatory subunits.75, 76 The amplifying impact of CK2 on EMT reflect its ability to support a number of signaling pathways that promote EMT in complementary ways, including those triggered by TGF-beta, Wnt-beta-catenin, Hedgehog, Notch1, HIF-1, NF-kappaB, and Akt.77 EMT not only aids metastasis, but also promotes the vessel co-option strategy of cancer spread whereby cancer cells migrate along the abluminal surface of capillaries, enabling them to develop resistance to anti-angiogenic cancer therapies.78, 79

New Drugs for Inhibition of CK2 – CX-4945

These considerations make it abundantly clear that well tolerated and effective pharmaceutical inhibitors of CK2 may have a bright future in oncology – both as agents for slowing cancer growth and spread, and as adjuvants to chemo- or radiotherapy. Some pharmaceutical companies are moving aggressively to evaluate the potential of this approach, and the highly potent and orally active CK2 inhibitor CX-4945 has shown impressive anti-cancer activity in mouse xenograft models, in doses which the animals appear to tolerate well.80, 81 Moreover, in doses that don’t greatly retard tumor growth, CX-4945 considerably amplifies response of an ovarian cancer xenograft to gemcitabine and cisplatin – though the somewhat greater weight loss in the mice receiving combination therapy suggests that toxicity might also be increased to a degree.58 This agent is now entering clinical trials, and its progress should be followed with the greatest interest.

Flavones/Flavonols as Natural Inhibitors of CK2

However, there are other known inhibitors of CK2, one being the dietary flavone apigenin. Indeed, long before the development of the more potent pharmaceutical inhibitors of CK2, this agent was employed as a relatively specific inhibitor of CK2 in cell culture studies, with a Ki near 1 µM.82 There are indeed a number of studies, both in cancer cell culture and in mouse xenograft models, showing that apigenin can exert cancer-retardant and chemo-potentiating effects. In xenograft models, apigenin has shown activity whether administered parenterally or orally, alone or as an adjuvant to chemotherapy.83-99 Intriguingly, many of the effects of apigenin on signaling pathways reported in cell culture or xenograft studies are parallel to those of CK2 inhibition, including down-regulated activity of Akt,92, 100-105 HIF-1,83, 85, 106-110 NF-kappaB,21, 25, 32, 100, 111, 111-114 STAT3,100, 113, 115 beta-catenin,116, 117 Gli1,118 AR,119, 120 and Cdc37,100 and up-regulated p53.113, 121-128 Indeed, Zhao and colleagues have recently proposed that inhibition of CK2 is a key mediator of apigenin’s anti-cancer activity in multiple myeloma cells.100 A survey of the burgeoning cancer research literature involving apigenin – 476 citations on Pubmed at present – reveals apigenin can influence a truly dizzying array of molecular targets in cancer cells; it is reasonable to suspect that, rather than directly inhibiting dozens of separate targets, it must be influencing one or more signaling factors that have a remarkably broad impact on the molecular biology of cancer cells. CK2 may be the crucial target in this regard. However, none of the studies in which apigenin has been administered in cancer-retardant doses to xenograftbearing mice have assessed the impact of apigenin on tumor CK2 activity. A study assessing this – perhaps by measuring Ser129 phosphorylation of Akt1 in tumors – would be worthwhile; and it would also be intriguing to see whether apigenin administration has any significant additional impact on cancer growth in animals that are already receiving potent doses of CX4945; if CK2 is apigenin’s key target, little additional benefit might be seen.
Although apigenin is considered the prototype flavone inhibitor of CK2, recent studies show that other naturally-occuring flavones and flavonols have similar or slightly more potent inhibitory activity. Working in vitro with human recombinant CK2, Lolli and colleagues have recently reported that apigenin, luteolin, kaempferol, fisetin, quercetin, and myricetin can inhibit CK2 with Kis of 0.8, 0.5, 0.4, 0.35, 0.55, and 0.92 µM, respectively.129 This inhibition is competitive with respect to the phosphodonor substrate ATP. All effective compounds are planar and are hydroxylated at the 7 and 4’ positions. Hydroxylations at 5, 3, and 3’ positions do not greatly add to or detract from activity.
These findings may help to explain the curious fact that every one of these flavones or flavonols has been reported to exert anti-cancer effects, both in cancer cell cultures, and in xenografted mice. Here are citations for the xenograft studies: apigenin,83-99 luteolin,130-141 kaempferol,142 fisetin,143-145 quercetin,146-164 myrcetin.165 There are at least 53 published studies in which flavones or flavonols have decreased the growth of human xenografts in nude mice.
It seems likely that, ultimately, a drug such as CX-4945 will offer the most convenient and effective way to address the CK2 activity of clinical cancer. However, this or comparable drugs will not be available for several years, and when available will initially only be approved for use in a limited number of cancers – and will doubtless be staggeringly expensive to use for off-label purposes. For this reason, it would be prudent to give serious attention to the possibility that apigenin or related flavones/flavonols might be clinically useful for suppressing CK2 activity in some sufficiently high dosage schedule. This might be assessed by pharmacokinetic studies in which a marker for CK2 activity, such as phosphorylation of Cdc37 – or Thr145 phosphorylation of p21, employed as a marker in studies with CX-454980 – is determined in leukocytes or some other accessible cell type. The efficacy of a given agent will presumably reflect it absorbability, the rapidity with which it is conjugated once absorbed (glucuronidation or sulfation), and its capacity to pass through cell walls. Pharmaceutical innovations which optimize absorbability might make this approach more feasible.160 With respect to quercetin, the approved food additive enzymatically-modified isoquercitrin (EMIQ), unlike quercetin, is highly soluble, but is metabolized to yield free quercetin at the intestinal brush border; a human pharmacokinetic study found that when equimolar amounts of quercetin and EMIQ were administered orally, the plasma levels of quercetin achieved were twenty-fold higher with EMIQ.166-170 This agent apparently has not yet been tested in rodent tumor models.
Rapid conjugation of absorbed flavonoids limits their capacity to exert intracellular effects.171 It is therefore fortunate that some tumors may have privileged access to flavone/flavonol glucuronide conjugates, owing to the fact that extracellular beta-glucuronidase activity tends to be elevated in tumors, particularly in their hypoxic/necrotic regions.172, 173 Infiltrating immune cells may be the chief source of this activity. Moreover, the tendency of extracellular pH to be acidic in such regions can be expected to amplify their beta-glucuronidase activity.174-176 Many investigators have proposed or presented evidence that glucuronide-masked anti-cancer agents –
including flavonoids – can be selectively activated within tumors.173, 176-184 Hence, the rapid glucuronidation of flavones and flavonols may not be an insuperable obstacle to the capacity of these compounds to inhibit CK2 in vivo. Perhaps this mechanism contributes to the demonstrable efficacy of flavones/flavonols in mouse xenograft studies; co-administration of a beta-glucuronidase inhibitor might clarify this. A corollary of this consideration, however, is that measurement of CK2 activity in healthy tissues following oral administration of flavones/flavonols may underestimate the capacity of these agents to inhibit CK2 within tumors.

Joint Inhibition of HDAC6 and CK2 to Target Hsp90 Function

In light of the fact that inhibition of the chaperoning function of Hsp90-Cdc37 plays a key role in the cancer-retardant efficacy of CK2 inhibitors, it is pertinent to note that acetylation of Hsp90 notably reduces its chaperoning activity.185-189 The cytosolic deacetylase HDAC6 targets these acetylations of Hsp90, restoring its activity. Hence, type II histone deacetylase inhibitors have the potential to complement the impact of CK2 inhibitors on the chaperoning of many prooncogenic kinases. Moreover, it has recently emerged that sulforaphane can function as an inhibitor of HDAC6 within cells;190 this phenomenon may be clinically relevant, as acute ingestion of 68 g of broccoli sprouts has been reported to suppress global histone deacetylase activity in peripheral blood mononuclear cells.185 This finding may be of particular interest, in light of the fact that HDAC6, rather like CK2, works in multifarious ways to sustain malignant cellular behavior, and is emerging as a key target for cancer therapy.191, 192 It would be of interest to determine whether flavones/flavonols and sulforaphane might complement each other’s efficacy in integrative cancer therapy.

A Potential Countervailing Effect – Nrf2 Activation

Although the great majority of reports examining quercetin’s impact on cancer, in vitro or in vivo, with or without concurrent chemotherapy, conclude that quercetin has cancer suppressive activity, one recent study found that, in low micromolar concentrations, quercetin protected a human ovarian cancer cell line from a range of cytotoxic drugs; concurrent quercetin administration decreased the cancer-retardant efficacy of cisplatin in a xenograft model. This effect was traced to quercetin’s ability to activate nrf2 and thereby increase the expression of antioxidant enzymes, glutathione, and glutathione-dependent detoxicant enzymes. The ability of phase 2 induction via nfr2 activation to promote chemoresistance in some cancers has been demonstrated. Hence, while a number of studies describe a chemosensitizing effect for quercetin
in cancer models154, 155, 193-195 – including a report that low concentrations of quercetin sensitize some ovarian cancer cell lines to cisplatin – the possibility remains that quercetin (and presumably other phase 2-inductive flavonols) may promote chemoresistance in some cancers. (The “flip side” of this observation is that quercetin has potential for protecting healthy tissues from chemotherapy drugs, as demonstrated in mice.196-200) These considerations, in any case, do not speak to quercetin’s potential utility as an adjuvant for slowing cancer growth.

Evaluating the Hypothesis

As noted above, EMIQ may be the most appropriate agent to study in pre-clinical and clinical trials, owing to its ability to promote absorption of quercetin. In cancer xenograft models, the impact of EMIQ administration on Ser129 phosphorylation of Akt1 in the tumor could be determined to assess this agent’s ability to suppress CK2 activity in vivo. Positive results in such studies could then encourage clinical cancer trials with EMIQ. Rather than expecting objective response, it would be more realistic to hope that flavonol administration will slow the growth and spread of cancer, as it does in rodent models. A placebo-controlled design might thus be required to establish clinical efficacy. The extent of Ser129 phosphorylation of Akt1 in leukocytes could be measured as a surrogate for CK2 inhibition in the cancer – bearing in mind, however, that quercetin metabolites might have greater activity within inflamed tumor tissue.
In regard to toxicity considerations, it should be noted that knockout of the alpha subunits of CK2 results in embryonic lethality.201, 202 However, flavonols in vivo would achieve at best only partial inhibition of CK2. In rodent studies with CX-4945, cancer control is noted with doses that are not overtly toxic to the animals. Phase I clinical trials with this agent have not yet been reported, so it is not clear what the dose-limiting toxicities of CK2 inhibitors will be. Flavonols are of course prominent phytochemicals in natural diets. The toxicological evaluation of EMIQ in rodents has been described by Valentova and colleagues;166 when fed at up to 2.5% of diet to rats for 13 weeks, yellowish discoloration of bones and urine was noted, and weight gain was slightly decreased at the highest doses. At 5% of diet, isoquercetin feeding to male rats was associated with significant declines in body weight, hemoglobin, triglycerides, bilirubin, and phosphorus, with small increases in the relative weights of the lungs and testes.203 EMIQ has been accorded GRAS status for use as a food additive. These considerations suggest that it would be reasonably safe to test EMIQ in doses of several grams daily in Phase I cancer trials.
While many different tumor types might reasonably be targeted in pre-clinical/clinical studies with EMIQ, the facts a substantial research literature finds ovarian cancers to be responsive to quercetin in vitro and in vivo, and that CK2 inhibitors, alone or as adjuvants to chemotherapy, have shown utility in such cancers, suggests that the impact of EMIQ in ovarian cancer models would be appropriate to study.204, 205 And, in light of the role of EMT in enabling cancers to escape control by anti-angiogenic therapies via vessel co-option, it would be of interest to study EMIQ as adjuvants to anti-angiogenic drugs in cancer lines that have become relatively resistant to control by those drugs alone.

References

(1) Meggio F, Pinna LA. One-thousand-and-one substrates of protein kinase CK2? FASEB J 2003 March;17(3):349-68.
(2) Tawfic S, Yu S, Wang H, Faust R, Davis A, Ahmed K. Protein kinase CK2 signal in neoplasia. Histol Histopathol 2001 April;16(2):573-82.
(3) Miyata Y, Nishida E. CK2 controls multiple protein kinases by phosphorylating a kinasetargeting molecular chaperone, Cdc37. Mol Cell Biol 2004 May;24(9):4065-74.
(4) Zhao M, Ma J, Zhu HY, Zhang XH, Du ZY, Xu YJ, Yu XD. Apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through targeting the trinity of CK2, Cdc37 and Hsp90. Mol Cancer 2011 August 29;10:104.
(5) Miyata Y, Nishida E. Evaluating CK2 activity with the antibody specific for the CK2phosphorylated form of a kinase-targeting cochaperone Cdc37. Mol Cell Biochem 2008 September;316(1-2):127-34.
(6) Di Maira G, Salvi M, Arrigoni G, Marin O, Sarno S, Brustolon F, Pinna LA, Ruzzene M. Protein kinase CK2 phosphorylates and upregulates Akt/PKB. Cell Death Differ 2005 June;12(6):668-77.
(7) Girardi C, James P, Zanin S, Pinna LA, Ruzzene M. Differential phosphorylation of Akt1 and Akt2 by protein kinase CK2 may account for isoform specific functions. Biochim Biophys Acta 2014 September;1843(9):1865-74.
(8) Franchin C, Borgo C, Cesaro L, Zaramella S, Vilardell J, Salvi M, Arrigoni G, Pinna LA. Reevaluation of protein kinase CK2 pleiotropy: new insights provided by a phosphoproteomics analysis of CK2 knockout cells. Cell Mol Life Sci 2018 June;75(11):2011-26.
(9) Zanin S, Borgo C, Girardi C, O’Brien SE, Miyata Y, Pinna LA, Donella-Deana A, Ruzzene M. Effects of the CK2 inhibitors CX-4945 and CX-5011 on drug-resistant cells. PLoS One 2012;7(11):e49193.
(10) Di Maira G, Brustolon F, Pinna LA, Ruzzene M. Dephosphorylation and inactivation of Akt/PKB is counteracted by protein kinase CK2 in HEK 293T cells. Cell Mol Life Sci 2009 October;66(20):3363-73.
(11) Siddiqui-Jain A, Drygin D, Streiner N, Chua P, Pierre F, O’Brien SE, Bliesath J, Omori M, Huser N, Ho C, Proffitt C, Schwaebe MK, Ryckman DM, Rice WG, Anderes K. CX-4945, an orally bioavailable selective inhibitor of protein kinase CK2, inhibits prosurvival and angiogenic signaling and exhibits antitumor efficacy. Cancer Res 2010 December 15;70(24):10288-98.
(12) Shehata M, Schnabl S, Demirtas D, Hilgarth M, Hubmann R, Ponath E, Badrnya S, Lehner C, Hoelbl A, Duechler M, Gaiger A, Zielinski C, Schwarzmeier JD, Jaeger U. Reconstitution of PTEN activity by CK2 inhibitors and interference with the PI3-K/Akt cascade counteract the antiapoptotic effect of human stromal cells in chronic lymphocytic leukemia. Blood 2010 October 7;116(14):2513-21.
(13) Barata JT. The impact of PTEN regulation by CK2 on PI3K-dependent signaling and leukemia cell survival. Adv Enzyme Regul 2011;51(1):37-49.
(14) Kang NI, Yoon HY, Kim HA, Kim KJ, Han MK, Lee YR, Hwang PH, Soh BY, Shin SJ, Im SY, Lee HK. Protein kinase CK2/PTEN pathway plays a key role in platelet-activating factormediated murine anaphylactic shock. J Immunol 2011 June 1;186(11):6625-32.
(15) Chatterjee A, Chatterjee U, Ghosh MK. Activation of protein kinase CK2 attenuates FOXO3a functioning in a PML-dependent manner: implications in human prostate cancer. Cell Death Dis 2013 March 14;4:e543.
(16) Romieu-Mourez R, Landesman-Bollag E, Seldin DC, Traish AM, Mercurio F, Sonenshein GE. Roles of IKK kinases and protein kinase CK2 in activation of nuclear factor-kappaB in breast cancer. Cancer Res 2001 May 1;61(9):3810-8.
(17) Shen J, Channavajhala P, Seldin DC, Sonenshein GE. Phosphorylation by the protein kinase CK2 promotes calpain-mediated degradation of IkappaBalpha. J Immunol 2001 November 1;167(9):4919-25.
(18) Romieu-Mourez R, Landesman-Bollag E, Seldin DC, Sonenshein GE. Protein kinase CK2 promotes aberrant activation of nuclear factor-kappaB, transformed phenotype, and survival of breast cancer cells. Cancer Res 2002 November 15;62(22):6770-8.
(19) Kato T, Jr., Delhase M, Hoffmann A, Karin M. CK2 Is a C-Terminal IkappaB Kinase Responsible for NF-kappaB Activation during the UV Response. Mol Cell 2003 October;12(4):829-39.
(20) Cavin LG, Romieu-Mourez R, Panta GR, Sun J, Factor VM, Thorgeirsson SS, Sonenshein GE, Arsura M. Inhibition of CK2 activity by TGF-beta1 promotes IkappaB-alpha protein stabilization and apoptosis of immortalized hepatocytes. Hepatology 2003 December;38(6):1540-51.
(21) Eddy SF, Guo S, Demicco EG, Romieu-Mourez R, Landesman-Bollag E, Seldin DC, Sonenshein GE. Inducible IkappaB kinase/IkappaB kinase epsilon expression is induced by CK2 and promotes aberrant nuclear factor-kappaB activation in breast cancer cells. Cancer Res 2005 December 15;65(24):11375-83.
(22) Piazza FA, Ruzzene M, Gurrieri C, Montini B, Bonanni L, Chioetto G, Di MG, Barbon F, Cabrelle A, Zambello R, Adami F, Trentin L, Pinna LA, Semenzato G. Multiple myeloma cell survival relies on high activity of protein kinase CK2. Blood 2006 September 1;108(5):1698707.
(23) Yu M, Yeh J, Van WC. Protein kinase casein kinase 2 mediates inhibitor-kappaB kinase and aberrant nuclear factor-kappaB activation by serum factor(s) in head and neck squamous carcinoma cells. Cancer Res 2006 July 1;66(13):6722-31.
(24) Parhar K, Morse J, Salh B. The role of protein kinase CK2 in intestinal epithelial cell inflammatory signaling. Int J Colorectal Dis 2007 June;22(6):601-9.
(25) Hamacher R, Saur D, Fritsch R, Reichert M, Schmid RM, Schneider G. Casein kinase II inhibition induces apoptosis in pancreatic cancer cells. Oncol Rep 2007 September;18(3):695701.
(26) Dominguez I, Sonenshein GE, Seldin DC. Protein kinase CK2 in health and disease: CK2 and its role in Wnt and NF-kappaB signaling: linking development and cancer. Cell Mol Life Sci 2009 June;66(11-12):1850-7.
(27) Brown MS, Diallo OT, Hu M, Ehsanian R, Yang X, Arun P, Lu H, Korman V, Unger G, Ahmed K, Van WC, Chen Z. CK2 modulation of NF-kappaB, TP53, and the malignant phenotype in head and neck cancer by anti-CK2 oligonucleotides in vitro or in vivo via sub-50nm nanocapsules. Clin Cancer Res 2010 April 15;16(8):2295-307.
(28) Tsuchiya Y, Asano T, Nakayama K, Kato T, Jr., Karin M, Kamata H. Nuclear IKKbeta is an adaptor protein for IkappaBalpha ubiquitination and degradation in UV-induced NF-kappaB activation. Mol Cell 2010 August 27;39(4):570-82.
(29) Trembley JH, Unger GM, Tobolt DK, Korman VL, Wang G, Ahmad KA, Slaton JW, Kren BT, Ahmed K. Systemic administration of antisense oligonucleotides simultaneously targeting CK2alpha and alpha’ subunits reduces orthotopic xenograft prostate tumors in mice. Mol Cell Biochem 2011 October;356(1-2):21-35.
(30) Trembley JH, Unger GM, Korman VL, Tobolt DK, Kazimierczuk Z, Pinna LA, Kren BT, Ahmed K. Nanoencapsulated anti-CK2 small molecule drug or siRNA specifically targets malignant cancer but not benign cells. Cancer Lett 2012 February 1;315(1):48-58.
(31) Zheng Y, McFarland BC, Drygin D, Yu H, Bellis SL, Kim H, Bredel M, Benveniste EN. Targeting Protein Kinase CK2 Suppresses Pro-survival Signaling Pathways and Growth of Glioblastoma. Clin Cancer Res 2013 September 13.
(32) Mottet D, Ruys SP, Demazy C, Raes M, Michiels C. Role for casein kinase 2 in the regulation of HIF-1 activity. Int J Cancer 2005 December 10;117(5):764-74.
(33) Hubert A, Paris S, Piret JP, Ninane N, Raes M, Michiels C. Casein kinase 2 inhibition decreases hypoxia-inducible factor-1 activity under hypoxia through elevated p53 protein level. J Cell Sci 2006 August 15;119(Pt 16):3351-62.
(34) Ampofo E, Kietzmann T, Zimmer A, Jakupovic M, Montenarh M, Gotz C. Phosphorylation of the von Hippel-Lindau protein (VHL) by protein kinase CK2 reduces its protein stability and affects p53 and HIF-1alpha mediated transcription. Int J Biochem Cell Biol 2010 October;42(10):1729-35.
(35) Song DH, Sussman DJ, Seldin DC. Endogenous protein kinase CK2 participates in Wnt signaling in mammary epithelial cells. J Biol Chem 2000 August 4;275(31):23790-7.
(36) Song DH, Dominguez I, Mizuno J, Kaut M, Mohr SC, Seldin DC. CK2 phosphorylation of the armadillo repeat region of beta-catenin potentiates Wnt signaling. J Biol Chem 2003 June 27;278(26):24018-25.
(37) Seldin DC, Landesman-Bollag E, Farago M, Currier N, Lou D, Dominguez I. CK2 as a positive regulator of Wnt signalling and tumourigenesis. Mol Cell Biochem 2005 June;274(1-2):63-7.
(38) Tapia JC, Torres VA, Rodriguez DA, Leyton L, Quest AF. Casein kinase 2 (CK2) increases survivin expression via enhanced beta-catenin-T cell factor/lymphoid enhancer binding factordependent transcription. Proc Natl Acad Sci U S A 2006 October 10;103(41):15079-84.
(39) Wang S, Jones KA. CK2 controls the recruitment of Wnt regulators to target genes in vivo. Curr Biol 2006 November 21;16(22):2239-44.
(40) Lee AK, Ahn SG, Yoon JH, Kim SA. Sox4 stimulates ss-catenin activity through induction of CK2. Oncol Rep 2011 February;25(2):559-65.
(41) Ponce DP, Maturana JL, Cabello P, Yefi R, Niechi I, Silva E, Armisen R, Galindo M, Antonelli M, Tapia JC. Phosphorylation of AKT/PKB by CK2 is necessary for the AKT-dependent upregulation of beta-catenin transcriptional activity. J Cell Physiol 2011 July;226(7):1953-9.
(42) Ponce DP, Yefi R, Cabello P, Maturana JL, Niechi I, Silva E, Galindo M, Antonelli M, Marcelain K, Armisen R, Tapia JC. CK2 functionally interacts with AKT/PKB to promote the beta-catenin-dependent expression of survivin and enhance cell survival. Mol Cell Biochem 2011 October;356(1-2):127-32.
(43) Kim J, Hwan KS. CK2 Inhibitor CX-4945 Blocks TGF-beta1-Induced Epithelial-toMesenchymal Transition in A549 Human Lung Adenocarcinoma Cells. PLoS ONE 2013;8(9):e74342.
(44) Kim S, Ham S, Yang K, Kim K. Protein kinase CK2 activation is required for transforming growth factor beta-induced epithelial-mesenchymal transition. Mol Oncol 2018 October;12(10):1811-26.
(45) Zdunek M, Silbiger S, Lei J, Neugarten J. Protein kinase CK2 mediates TGF-beta1-stimulated type IV collagen gene transcription and its reversal by estradiol. Kidney Int 2001 December;60(6):2097-108.
(46) Kim J, Hwan KS. CK2 inhibitor CX-4945 blocks TGF-beta1-induced epithelial-tomesenchymal transition in A549 human lung adenocarcinoma cells. PLoS One 2013;8(9):e74342.
(47) Zhang Y, Dees C, Beyer C, Lin NY, Distler A, Zerr P, Palumbo K, Susok L, Kreuter A, Distler O, Schett G, Distler JH. Inhibition of casein kinase II reduces TGFbeta induced fibroblast activation and ameliorates experimental fibrosis. Ann Rheum Dis 2015 May;74(5):936-43.
(48) Lin YC, Hung MS, Lin CK, Li JM, Lee KD, Li YC, Chen MF, Chen JK, Yang CT. CK2 inhibitors enhance the radiosensitivity of human non-small cell lung cancer cells through inhibition of stat3 activation. Cancer Biother Radiopharm 2011 June;26(3):381-8.
(49) Manni S, Brancalion A, Mandato E, Tubi LQ, Colpo A, Pizzi M, Cappellesso R, Zaffino F, Di Maggio SA, Cabrelle A, Marino F, Zambello R, Trentin L, Adami F, Gurrieri C, Semenzato G, Piazza F. Protein kinase CK2 inhibition down modulates the NF-kappaB and STAT3 survival pathways, enhances the cellular proteotoxic stress and synergistically boosts the cytotoxic effect of bortezomib on multiple myeloma and mantle cell lymphoma cells. PLoS One 2013;8(9):e75280.
(50) Zhang S, Wang Y, Mao JH, Hsieh D, Kim IJ, Hu LM, Xu Z, Long H, Jablons DM, You L. Inhibition of CK2alpha down-regulates Hedgehog/Gli signaling leading to a reduction of a stem-like side population in human lung cancer cells. PLoS ONE 2012;7(6):e38996.