Exp Hematol OncolExp Hematol OncolExperimental Hematology & Oncology2162-3619BioMed Central2321093035069892162-3619-1-410.1186/2162-3619-1-4ReviewDigitoxin and its analogs as novel cancer therapeuticsElbazHosam A1helbaz@hsc.wvu.eduStueckleTodd A14tstueckle@hsc.wvu.eduTseWilliam2tse@hsc.wvu.eduRojanasakulYon15yrojan@hsc.wvu.eduDinuCerasela Zoica36cerasela-zoica.dinu@mail.wvu.eduDepartment of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USAMary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, USADepartment of Chemical Engineering, West Virginia University, Morgantown, WV, USANational Institute for Occupational Safety and Health, Morgantown, WV, USADepartment of Basic Pharmaceutical Sciences, West Virginia University, PO Box 9530, 1 Medical Center Drive, Morgantown, WV 26506, USADepartment of Chemical Engineering, West Virginia University, College of Engineering and Mineral Resources, PO Box 6102, ESB 445, Morgantown, WV 26506, USA20125420121442822012542012Copyright ©2012 Elbaz et al; licensee BioMed Central Ltd.2012Elbaz et al; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

A growing body of evidence indicates that digitoxin cardiac glycoside is a promising anticancer agent when used at therapeutic concentrations. Digitoxin has a prolonged half-life and a well-established clinical profile. New scientific avenues have shown that manipulating the chemical structure of the saccharide moiety of digitoxin leads to synthetic analogs with increased cytotoxic activity. However, the anticancer mechanism of digitoxin or synthetic analogs is still subject to study while concerns about digitoxin's cardiotoxicity preclude its clinical application in cancer therapeutics. This review focuses on digitoxin and its analogs, and their cytotoxicity against cancer cells. Moreover, a new perspective on the pharmacological aspects of digitoxin and its analogs is provided to emphasize new research directions for developing potent chemotherapeutic drugs.

Introduction

Cardiac glycosides (CGs) are a large family of chemical compounds found in several plants and animal species [1]. Plants containing CGs were used for more than 1500 years as diuretics, emetics, abortifacients, antineoplastics, and heart tonics [2]. In the 18th century, English physician and scientist William Withering discovered that a patient with congestive heart failure, "dropsy", improved after administering foxglove extract (Digitalis purpurea L.) [3]. Since then, many CGs have been isolated and their pharmacological effects have been tested; subsequently, CGs were used for treating congestive heart failure, cardiac arrhythmias, and atrial fibrillation [4,5].

Generally, CGs share a common structural motif with a steroidal nucleus, a sugar moiety at position 3 (C3), and a lactone moiety at position 17 (C17) [1]. Figure 1 shows the common structural motif of CGs. The steroidal nucleus is the core structure of CG and is considered to be the active pharmacophoric moiety [6]. CGs also show an A/B and C/D cis-conformation that is different from mineralocorticoids, glycocorticoids, or sex hormones known to show trans-confirmation [6]. The presence of sugars at position 3 on the steroid ring significantly affects the pharmacological profile of each glycoside [7,8]. Free aglycones, for instance, show faster and less complex absorption and metabolism compared to their glycosylated counterparts [7]. Additionally, the type of sugar attached to the steroidal nucleus changes the potency of CG compounds [8,9].

Structural characteristics of CGs. The common structural motif of a CG molecule is characterized by a steroidal nucleus, a lactone moiety at C17, and a saccharide moiety at C3 linked to the steroidal nucleus by a glycosidic linkage.

It is well established that CGs inhibit Na+/K+-ATPase and increases intracellular sodium ions [10]. The Na+/K+-ATPase is a P-type pump that actively transports potassium ions inside and sodium ions outside cells in a 2:3 stoichiometry [11]. Such activity keeps intracellular sodium levels low, thus initiating and sustaining adequate electrochemical gradient across the plasma membrane [11]. Appropriate electrochemical gradient is essential for vital cellular processes such as ion homeostasis, neuronal communication, and apoptosis [10,12]. To maintain ion homeostasis after Na+/K+-ATPase inhibition by CGs, the cells have to restore intracellular sodium concentration to their basal levels by stimulating the Na+/Ca2+ exchange pump to extrude sodium ions out of the cell and introduce calcium ions into the cell [10]. This activity increases intracellular calcium ions and results in increased cellular phenomena such as calcium dependent signaling and myocardial contractility [5].

Cardiac glycosides and their anticancer action

A growing body of evidence suggests significant anticancer effects mediated by CGs. For instance, in the 8th century, CG plant extracts were used for treating malignant conditions [13]. An ancient Chinese remedy that employs an extract of Bufo bufo toad secretions contains several CGs and is still being used today for managing cancerous conditions [14-16]. Investigations in using CGs as anticancer drugs intensified in the 1960s [17,18]. Shiratori et al. examined the cytotoxicity of CGs in rodent cancer models and found inhibition of in vitro proliferation at concentrations that are relatively toxic to humans (0.1-10 μM) [17]. A series of landmark epidemiological studies by Stenkvist et al. compared breast cancer tissue samples from women maintained on digitalis (a CG) for cardiac conditions to tissue samples from control patients [19]. This study found that women on digitalis therapy developed more benign forms of breast tumors when compared to control patients [19-22]. Additionally, patients on digitalis treatment showed a 9.6-time lower cancer recurrence rate compared to control patients, 5 years after mastectomy for primary tumor [21]. In 1984, Goldin et al. examined 127 cancer patients treated with digitalis [23]. While patients in the control group had 21 cancer-related deaths, only one patient died in the digitalis group [23]. Twenty years later using follow-up data, Stenkvist et al. found that those patients on digitalis treatment showed significantly lower mortality rate than the control group [22]. Given the promising epidemiological results and the emergence of human cancer cell lines, several studies investigated the cytotoxic effect of different CGs on human cancer cells. Examples of the CGs of plant or animal origins that exhibit anticancer effects are shown in Table 1.

Summary of most studied CGs and their anticancer activity

CompoundStructure and Natural OriginSusceptible cancer typesIC50 RangeReferences
Bufalin• Prostate cancer (PC3, DU145, and LNCaP cells)• Leukemia (THP1, U937, and MOLT-3 cells)0.1-10 μM[14,15,24-28]
Cinobufagin• Prostate cancer (PC3, DU145, and LNCaP cells)0.1-10 μM[14,15,24,25,27,28]
Digitoxin• Prostate cancer (PC3, DU145, and LNCaP cells)• Breast cancer (MCF-7 cells)• Renal adenocarcinoma (TK-10 cells)• Melanoma (UACC-62 cells)• Leukemia (K-562 cells)• Lung (A549 cells, and NCI-H460 cells)0.01-10 μM[9,29-40]
Digoxin• Prostate cancer (PC3, DU145, and LNCaP cells)• Cervical cancer (Hela cells)• Lung (A549 cells, and NCI-H460 cells)0.1-10 μM[29-32,35,40,41]
Oleandrin• Prostate cancer (PC3, DU145, LNCaP)• Leukemia (U-937, and HL-60 cells)• Breast cancer (MCF-7)• Lung• Malignant Fibroblast (VA-13)• Liver carcinoma (HepG2 cells)• Pancreatic cancer (PANC-1 cells)0.1 - 1 μM[42-57]
Ouabain• Prostate cancer (PC3, DU145, LNCaP)• Breast cancer (MDA-MB-435scells)• Lung (NCI-H460 cells)0.1-10 μM[2,29,30,32-35,40,46,58-62]
Proscillaridin• Breast cancer (MCF-7 cells)• Fibroblasts30-100 nM[29,30,32,33,35,43,63-66]
Digitoxin: a well established CG

Digitoxin is one of the few CGs that have been extensively studied and have a well-established clinical profile [67,68]. Digitoxin is a cardiotonic drug with a narrow therapeutic window; thus, toxicity is a persistent concern whenever digitoxin is considered for therapy [69]. Cardiotoxicity is frequently encountered as the most significant toxicity following digitoxin's clinical administration [70,71]. However, digitoxin is perhaps one of the most promising anti-cancer CGs since it shows significant anticancer effect against several types of cancer including lung cancer, pancreatic cancer, leukemia, and breast cancer, all at therapeutic concentrations (see Table 1) [18,31,58,68,72]. Moreover, digitoxin has a prolonged half-life [69], which can potentially contribute to a less frequent and more stable pharmacokinetic profile in digitoxin-treated cancer patients. Such advantages qualify digitoxin for further laboratory investigations and clinical trials. However, to justify the anticancer clinical application of digitoxin, its anticancer mechanism ought to be clearly understood. Also, with the persistent concern about digitoxin's cardiotoxicity, further studies are needed to clarify the cytotoxic mechanism of digitoxin in cancer cells.

Digitoxin and its anticancer mechanism

To understand its anticancer mechanisms, initial research focused on the ability of digitoxin to inhibit Na+/K+-ATPase pump at concentrations between 0.5 - 5 μM [5,10-12,73]. Such inhibition can result in increased intracellular calcium via increased Na+/Ca2+ pump activity and subsequently induce apoptosis in cancer cells [74-76]. Figure 2A shows how inhibiting the Na+/K+-ATPase pump causes an accumulation of intracellular sodium, thus perturbation of cellular ion homeostasis [5,10-12]. For the cells to restore ion homeostasis, the Na+/Ca2+ exchanger needs to be activated to export excess sodium to the extracellular space while importing calcium, which in turn causes an accumulation of intracellular calcium that mediates cellular events such as myocardial contractility, cytoskeleton remodeling, and apoptosis [5,10-12]. Interestingly, however, epidemiological studies have shown that digitoxin inhibits cancer cell viability at nanomolar concentrations (10-100 nM) [18,31,36,58]. This suggests that digitoxin exhibits a different mechanism for its anticancer effect than the one initially proposed.

Effects of digitoxin on Na+/K+-ATPase at micromolar and nanomolar concentrations. (A) Old theory model summarizes the effect of digitoxin at 0.5-5 μM concentrations. (B) New theory model summarizes digitoxin's effect at 0.01-0.1 μM concentrations.

In 2003, Xie et al. suggested that the signaling characteristics of Na+/K+-ATPase are distinct from the ion pumping activity [77]. It was shown that Na+/K+-ATPase signalosome is a multiple-protein signaling complex of 3 alpha (α) subunits and 2 beta (β) subunits that controls cellular activities like apoptosis [78], cell proliferation [79], cell motility [80], and tight junctions [81]. Subsequently, it was proposed that digitoxin at nanomolar concentrations activates Na+/K+ATPase signalosome to transmit intracellular signals. Figure 2B summarizes the intracellular effects of digitoxin upon binding to Na+/K+-ATPase at concentrations between 10-100 nM. Upon binding, digitoxin modulates the Na+/K+-ATPase protein complex activating the associated downstream signaling pathways, i.e., activation of several signaling cascades such as phospholipase C (PLC) signaling, mitogen-activated protein kinase (MAPK) signaling, phosphatidyl-inositol-3-kinase (PI3K) signaling, and Src kinase signaling.

The signaling cascades that are stimulated upon the interaction of digitoxin with Na+/K+-ATPase have mixed functions. For example, the MAPK signaling is mainly a proliferative and pro-survival signaling that can also be complemented by a pro-apoptotic signaling capability [82-88]. Src kinase signaling is a non-receptor tyrosine kinase that exhibits pro-survival as well as pro-apoptotic functions [89-97]. Therefore, digitoxin's ability to affect the downstream Na+/K+-ATPase signalosome is very complex. Such complexity, together with the digitoxin's narrow therapeutic window and its known cardiotoxicity, led to a slow progress in developing digitoxin as anticancer therapeutics [1,2,18,98].

Designing digitoxin analogs as anticancer agents

Early on, it was suggested that a potential approach to circumvent digitoxin's cardiotoxicity is to design synthetic analogs that are either more effective or less toxic than digitoxin when used as anticancer agents [8,99]. Such analogs were designed by structural modifications of digitoxin's chemical entity. For example, using digitoxin as a template, Langenhan et al. developed MeON-neoglycosylation, a chemoselective method for glycorandomization that employs modifying the glycosidic bond that links the saccharide moiety to the core steroidal nucleus of digitoxin [99]. In 2008, Zhou and O'Doherty developed another method for modifying the glycosidic linkage of digitoxin using palladium-catalyzed glycosylation [100,101]. The O'Doherty analogs are characterized by an ether linkage between the saccharide moiety and the steroidal nucleus. As a result, novel synthetic digitoxin O-glycoside analogs were synthesized [9,100,101]. Figure 3 shows the core structures of digitoxin analogs synthesized by Langenhan et al. and O'Doherty et al. respectively [99-101].

The structure of novel digitoxin analogs. (A) The common structural motif of a neoglycoside molecule developed by Langenhan et al. and characterized by a tertiary amine bond linking the saccharide moiety to the steroidal nucleus. (B) The common structural motif of a digitoxin O-saccharide molecule developed by O'Doherty et al. and characterized by an ether bond linking the saccharide moiety to the steroidal nucleus.

The differential potency and cytotoxic selectivity of digitoxin MeON-neoglycosides and digitoxin O-glycosides have been demonstrated in various cancer cell lines. For instance, Iyer et al. compared a library of digitoxin MeON-neoglycosides and digitoxin O-glycosides in a panel of 60 different cancer cell lines [9]. They showed that O-glycosides are more potent anticancer agents when compared to MeON-neoglycosides in a variety of cancer cell lines from leukemia, to lung, pancreatic and breast cancer cells. Additionally, the authors also show that the potency of the analogs depends on their sugar moiety with the monosaccharide analog being more potent than the di- and tri-saccharide analogs, respectively. Wang et al. used artificial O-monosaccharide analogs that they tested against a panel of 60 different cancer cell lines [37-39]. They identified three digitoxin monosaccharide analogs that showed significantly greater cytotoxic potential against non-small cell lung cancer (NSCLC) cells [37-39]. The most promising of these analogs, D6-MA, was subsequently evaluated for its anticancer mechanism by Elbaz et al. [36].

Anticancer mechanism and selectivity of digitoxin and D6-MA analog

Elbaz et al. showed that the D6-MA analog was 4-5 folds more potent than digitoxin in inhibiting cell proliferation, inducing cell cycle arrest and apoptosis in NSCLC cells [36]. The authors also showed that digitoxin and D6-MA exhibited significantly greater cytotoxicity against NSCLC when compared to both primary and non-tumorigenic lung epithelial lines. The selective cytotoxicity in lung cancer cells included G2/M phase arrest and apoptosis [36]. Moreover, digitoxin and D6-MA inhibited the expression of p53, cdc2, cyclin B1, survivin, and Chk1/2, critical genes/proteins for cell proliferation.

In order to explain the observed anticancer effect, it is postulated that digitoxin and D6-MA activate several transcriptional regulatory cascades via the Na+/K+-ATPase signalosome [61,62,77,102-107]. This suggested mechanism of selective cytotoxic effect may be associated with reduced expression of cell cycle regulatory proteins that are specifically overexpressed in cancer cells. Figure 4 illustrates how digitoxin regulates the expression of these cancer specific proteins.

Digitoxin modulates Na+/K+-ATPase signalosome and inhibits the transcriptional activity of AP-1 and NF-κB which mediate the expression of cancer-specific cycle regulatory genes such as cyclin B1, cdc2, Chk1, Chk2, and surviving.

The above suggested mechanism is supported by previous work showing that digitoxin inhibits AP-1 and NF-κB signaling through the Na+/K+-ATPase signalosome [45,55,106,108-112]. AP-1 is a transcription factor that promotes the expression of cell cycle regulatory proteins such as cdc2 and cyclinB1 [113-115]. Several studies have shown that cyclinB1 is essential for cell viability [116,117]. CyclinB1 and cdc2 are overexpressed in cancer cells [118-123] and form a complex with each other prior to mitosis. They catalyze chromatin condensation as well as nuclear envelope breakdown during mitosis [124,125]. The cyclinB1/cdc2 complex performs a rate limiting function in G2/M phase transition and protects mitotic cells from apoptosis by activating survivin. Survivin is a vital cell cycle regulatory protein known to be overexpressed in different cancer types [126-131]. Survivin controls cell progression through mitosis by promoting the chromosomal passenger complex and regulating microtubule dynamics [132-135]. Additionally, survivin regulates the mitotic spindle checkpoint [136-139]. Several studies suggest survivin downregulation as a biomarker for mitotic catastrophe [140-142]. Also, NF-κB transcription factor regulates the expression of cell cycle regulatory proteins such as survivin [143]. Thus, by inhibiting both AP-1 and NF-κB signaling, digitoxin and D6-MA potentially inhibit the expression of cdc2, cyclin B1, survivin and Chk1/2, as shown in Figures 2 and 4. Moreover, the association of cyclin B1/cdc2 complex with survivin indicates that the complex is crucial for G2/M phase transition and cell viability, and inhibiting its expression by both digitoxin and D6-MA could potentially explain the selective cytotoxicity of these compounds towards cancer cells. Since inhibiting Chk1/2 typically elicits checkpoint abrogation and uncontrolled cell cycle progression abolished cell viability [144-146], a potential alternative mechanism of digitoxin and D6-MA's cytotoxicity may be their ability to cause checkpoint abrogation. It is unclear, however, how digitoxin and D6-MA inhibit checkpoint kinase proteins in cancer cells. Distinguishing between CG-induced cell cycle arrest and/or checkpoint abrogation will improve our understanding on how CGs including digitoxin and its analogs exert their anti-proliferative/cell death effect.

Conclusions and outlook

Digitoxin and D6-MA strongly modulate cell cycle machinery through the Na+/K+-ATPase signalosome in a manner that significantly undermines cell viability (Figure 4). However, several questions remain unanswered regarding their mechanistic control of cancer cell cycle. Delineating the mechanism of cell cycle arrest and selectivity of digitoxin and its analogs are particularly interesting. First, it will improve our understanding of how digitoxin and its analogs cause G2/M phase arrest in cancer cells. Second, it will help identify novel p53-independent therapeutic targets to mediate cancer cell death. Since many cancer types develop resistance to chemotherapy by reducing p53 expression and signaling [158-168], identifying p53-independent therapeutic targets will contribute to the development of new and more effective cancer treatments by circumventing p53 related resistance mechanisms. Third, detailing how digitoxin and D6-MA inhibit survivin expression in cancer cells will improve our understanding on how the drugs cause G2/M phase arrest and apoptosis. Last, dissecting the inhibitory effects of these compounds on survivin and p53 expression in cancer cells would explore whether and how the compounds induce mitotic catastrophe. Answering these questions will elevate our understanding on the antineoplastic mechanism of CGs at therapeutically relevant concentrations, and develop new and more effective cancer treatments [147].

Disclaimer

Research findings and conclusions are those of the authors and do not necessarily represent the views of the National Institute for Occupational Safety and Health.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

HAE drafted the original manuscript; TAS and WT coordinated and helped to draft the manuscript. YR and CZD have been involved in revising the manuscript critically for its scientific content and have given final approval of the version to be published. All the authors read and approved the final manuscript.

Acknowledgements

This work is supported by the NIH grants R01-HL076340, GM090259, GM088839 and the NSF grant EPS-1003907.

PrassasIDiamandisEPNovel therapeutic applications of cardiac glycosidesNat Rev Drug Discov200871192693510.1038/nrd268218948999NewmanRAYangPPawlusADBlockKICardiac Glycosides as Novel Cancer Therapeutic AgentsMol Interv200881364910.1124/mi.8.1.818332483HuxtableRJThe Erroneous Pharmacology of a CatMol Interv200112757714993326GheorghiadeMvan VeldhuisenDJColucciWSContemporary Use of Digoxin in the Management of Cardiovascular DisordersCirculation2006113212556256410.1161/CIRCULATIONAHA.105.56011016735690RahimtoolaSHTakTThe use of digitalis in heart failureCurr Probl Cardiol1996211278185310.1016/S0146-2806(96)80001-68968683SchonfeldWThe lead structure in cardiac glycosides is 5[beta],14[beta]-androstane-3[beta]14-diolNaunyn Schmiedebergs Arch Pharmacol198532941442610.1007/BF004963774033807MeleroCPMedardeaMFelicianoASA short review on cardiotonic steroids and their aminoguanidine analoguesMolecules20005518110.3390/50100051LangenhanJMPetersNRGuzeiIAHoffmannMThorsonJSEnhancing the anticancer properties of cardiac glycosides by neoglycorandomizationProc Natl Acad Sci USA200510235123051231010.1073/pnas.050327010216105948IyerAKVZhouMAzadNElbazHWangLRogalskyDKRojanasakulYO'DohertyGALangenhanJMA Direct Comparison of the Anticancer Activities of Digitoxin MeON-Neoglycosides and O-Glycosides: Oligosaccharide Chain Length-Dependent Induction of Caspase-9-Mediated ApoptosisACS Med Chem Lett20101732633010.1021/ml100093321103068BarryWHasinYSmithTSodium pump inhibition, enhanced calcium influx via sodium-calcium exchange, and positive inotropic response in cultured heart cellsCirc Res19855622312412578900KaplanJHBiochemistry of Na, K-ATPaseAnnu Rev Biochem20027151153510.1146/annurev.biochem.71.102201.14121812045105KaplanJGMembrane cation transport and the control of proliferation of mammalian cellsAnnu Rev Physiol197840194110.1146/annurev.ph.40.030178.000315147644BrewerHHistorical perspectives on healthJ R Soc Promot Health2004124418418710.1177/14664240041240041215301318WatabeMMasudaYNakajoSYoshidaTKuroiwaYNakayaKThe Cooperative Interaction of Two Different Signaling Pathways in Response to Bufalin Induces Apoptosis in Human Leukemia U937 CellsJ Biol Chem199627124140671407310.1074/jbc.271.24.140678662906YehJYHuangWJKanSFWangPSEffects of bufalin and cinobufagin on the proliferation of androgen dependent and independent prostate cancer cellsProstate200354211212410.1002/pros.1017212497584HanKQAnti-tumor activities and apoptosis-regulated mechanisms of bufalin on the orthotopic transplantation tumor model of human hepatocellular carcinoma in nude miceWorld J Gastroenterol2007133374337917659679ShiratoriOGrowth inhibitory effect of cardiac glycosides and aglycones on neoplastic cells: in vitro and in vivo studiesGann19675865215285589524López-LázaroMDigitoxin as an anticancer agent with selectivity for cancer cells: possible mechanisms involvedExpert Opin Ther Targets20071181043105310.1517/14728222.11.8.104317665977StenkvistBCardiac glycosides and breast cancerLancet1979156385158StenkvistBEvidence of a modifying influence of heart glucosides on the development of breast cancerAnal Quant Cytol1980249547377665StenkvistBCardiac glycosides and breast cancer, revisitedN Engl J Med19823064847057849StenkvistBIs digitalis a therapy for breast carcinoma?Oncol Rep1999649349610203580GoldinAGSafaARDigitalis and cancerLancet1984111346144872HashimotoSJingYKawazoeNMasudaYNakajoSYoshidaTKuroiwaYNakayaKBufalin reduces the level of topoisomerase II in human leukemia cells and affects the cytotoxicity of anticancer drugsLeuk Res199721987588310.1016/S0145-2126(97)00061-19393603JingYSelective inhibitory effect of bufalin on growth of human tumor cells in vitro: association with the induction of apoptosis in leukemia HL-60 cellsJpn J Cancer Res19948564565110.1111/j.1349-7006.1994.tb02408.x8063619KamanoYKotakeAHashimaHInoueMMoritaHTakeyaKItokawaHNandachiNSegawaTYukitaAStructure-cytotoxic activity relationship for the toad poison bufadienolidesBioorg Med Chem1998671103111510.1016/S0968-0896(98)00067-49730247KawazoeNWatabeMMasudaYNakajoSNakayaKTiam1 is involved in the regulation of bufalin-induced apoptosis in human leukemia cellsOncogene1999182413242110.1038/sj.onc.120255510229192MasudaYBufalin induces apoptosis and influences the expression of apoptosis-related genes in human leukemia cellsLeuk Res19951954955610.1016/0145-2126(95)00031-I7658701BielawskiKWinnickaKBielawskaAInhibition of DNA topoisomerases I and II, and growth inhibition of breast cancer MCF-7 cells by ouabain, digoxin and proscillaridin ABiol Pharm Bull20062971493149710.1248/bpb.29.149316819197JohanssonSLindholmPGullboJLarssonPBohlinLClaesonPCytotoxicity of digitoxin and related cardiac glycosides in human tumor cellsAnti-Cancer Drugs200112547548310.1097/00001813-200106000-0000911395576Lopez-LazaroMPastorNAzrakSSAyusoMJAustinCACortesFDigitoxin inhibits the growth of cancer cell lines at concentrations commonly found in cardiac patientsJ Nat Prod200568111642164510.1021/np050226l16309315WinnickaKBielawskiKBielawskaAMiltykWApoptosis-mediated cytotoxicity of ouabain, digoxin and proscillaridin A in the estrogen independent MDA-MB-231 breast cancer cellsArch Pharm Res2007301216122410.1007/BF0298026218038900WinnickaKBielawskiKBielawskaAMiltykWDual effects of ouabain, digoxin and proscillaridin A on the regulation of apoptosis in human fibroblastsNat Prod Res201024327428510.1080/1478641090299187820140806YehJYHuangWJKanSFWangPSInhibitory effects of digitalis on the proliferation of androgen dependent and independent prostate cancer cellsJ Urol200116651937194210.1016/S0022-5347(05)65724-211586264ZhangHFQianDZTanYSLeeKGaoPRenYRReySHammerHChangDPiliRDigoxin and other cardiac glycosides inhibit HIF-1 alpha synthesis and block tumor growthProc Natl Acad Sci USA200810550195791958610.1073/pnas.080976310519020076ElbazHAStueckleTAWangH-YLO'DohertyGLowryDTSargentLMWangLDinuCZRojanasakulYDigitoxin and a synthetic monosaccharide analog inhibit cell viability in lung cancer cellsToxicology and Applied Pharmacology20122581516010.1016/j.taap.2011.10.00722037315WangH-YLRojanasakulYO‚ÄôDohertyGASynthesis and Evaluation of the α-d-/α-l-Rhamnosyl and Amicetosyl Digitoxigenin Oligomers as Antitumor AgentsACS Med Chem Lett20112426426910.1021/ml100290d21660118WangH-YLWuBZhangQKangS-WRojanasakulYO‚ÄôDohertyGAC5'-Alkyl Substitution Effects on Digitoxigenin α-l-Glycoside Cancer CytotoxicityACS Med Chem Lett20112425926310.1021/ml100291n21572583WangH-YLXinWZhouMStueckleTARojanasakulYO'DohertyGAStereochemical Survey of Digitoxin MonosaccharidesACS Med Chem Lett201021737821643465WangZZhengMLiZLiRJiaLXiongXSouthallNWangSXiaMAustinCPCardiac Glycosides Inhibit p53 Synthesis by a Mechanism Relieved by Src or MAPK InhibitionCancer Res200969166556656410.1158/0008-5472.CAN-09-089119679550WinnickaKBielawskiKBielawskaAMiltykWDual effects of ouabain, digoxin and proscillaridin A on the regulation of apoptosis in human fibroblastsNat Prod Res201024327428510.1080/1478641090299187820140806AfaqFSaleemMAzizMHMukhtarHInhibition of 12-O-tetradecanoylphorbol-13-acetate-induced tumor promotion markers in CD-1 mouse skin by oleandrinToxicol Appl Pharmacol2004195336136910.1016/j.taap.2003.09.02715020199FelthJRickardsonLRosenJWickstromMFryknasMLindskogMBohlinLGullboJCytotoxic Effects of Cardiac Glycosides in Colon Cancer Cells, Alone and in Combination with Standard Chemotherapeutic DrugsJ Nat Prod200972111969197410.1021/np900210m19894733GuptaRSChopraAStetskoDKCellular basis for the species differences in sensitivity to cardiac glycosides (digitalis)J Cell Physiol1986127219720610.1002/jcp.10412702023009493MannaSKSahNKNewmanRACisnerosAAggarwalBBOleandrin suppresses activation of nuclear transcription factor-kappa B, activator protein-1, and c-Jun NH2-terminal kinaseCancer Res200060143838384710919658McConkeyDJLinYNuttLKOzelHZNewmanRACardiac glycosides stimulate Ca2+ increases and apoptosis in androgen-independent, metastatic human prostate adenocarcinoma cellsCancer Res200060143807381210919654NasuSMilasLKawabeSRajuUNewmanRAEnhancement of radiotherapy by oleandrin is a caspase-3 dependent processCancer Lett2002185214515110.1016/S0304-3835(02)00263-X12169388NewmanRAOleandrin-mediated oxidative stress in human melanoma cellsJ Exp Ther Oncol2006516718116528968NewmanRAKondoYYokoyamaTDixonSCartwrightCChanDJohansenMYangPAutophagic cell death of human pancreatic tumor cells mediated by oleandrin, a lipid-soluble cardiac glycosideIntegr Cancer Ther2007635436410.1177/153473540730962318048883PathakSMultaniASNarayanSKumarVNewmanRAAnvirzelTM, an extract of Nerium oleander, induces cell death in human but not murine cancer cellsAnti-Cancer Drugs200011645546310.1097/00001813-200007000-0000611001386RaghavendraPBSreenivasanYMannaSKOleandrin induces apoptosis in human, but not in murine cells: Dephosphorylation of Akt, expression of FasL, and alteration of membrane fluidityMol Immunol20074492292230210.1016/j.molimm.2006.11.00917173971SchonerWScheiner-BobisGEndogenous and exogenous cardiac glycosides: their roles in hypertension, salt metabolism, and cell growthAm J Physiol Cell Physiol20072932C509C53610.1152/ajpcell.00098.200717494630SmithJAMaddenTVijjeswarapuMNewmanRAInhibition of export of fibroblast growth factor-2 (FGF-2) from the prostate cancer cell lines PC3 and DU145 by Anvirzel and its cardiac glycoside component, oleandrinBiochem Pharmacol200162446947210.1016/S0006-2952(01)00690-611448457SreenivasanYRaghavendraPBMannaSKOleandrin-mediated expression of fas potentiates apoptosis in tumor cellsJ Clin Immunol200626430832210.1007/s10875-006-9028-016779680SreenivasanYSarkarAMannaSKOleandrin suppresses activation of nuclear transcription factor-kappa B and activator protein-1 and potentiates apoptosis induced by ceramideBiochem Pharmacol200366112223223910.1016/j.bcp.2003.07.01014609747WangXMPlomleyJBNewmanRACisnerosALC/MS/MS analyses of an oleander extract for cancer treatmentAnal Chem200072153547355210.1021/ac991425a10952541YangPYMenterDGCartwrightCChanDDixonSSuraokarMMendozaGLlansaNNewmanRAOleandrin-mediated inhibition of human tumor cell proliferation: importance of Na, K-ATPase alpha subunits as drug targetsMol Cancer Ther2009882319232810.1158/1535-7163.MCT-08-108519671733KometianiPLiuLAskariADigitalis-Induced Signaling by Na+/K + -ATPase in Human Breast Cancer CellsMol Pharmacol200567392993615602003LawrenceTSOuabain sensitizes tumor cells but not normal cells to radiationInt J Radiat Oncol Biol Phys198815495395810.1016/0360-3016(88)90132-03182336WinnickaKBielawskiKBielawskaASurazy ≈ ÑskiAAntiproliferative activity of derivatives of ouabain, digoxin and proscillaridin A in human MCF-7 and MDA-MB-231 breast cancer cellsBiol Pharm Bull20083161131114010.1248/bpb.31.113118520043XieZKometianiPLiuJLiJShapiroJIAskariAIntracellular reactive oxygen species mediate the linkage of Na+/K + -ATPase to hypertrophy and its marker genes in cardiac myocytesJ Biol Chem199927427193231932810.1074/jbc.274.27.1932310383443XuJ-WJinR-MWangY-RLinWMengBEffects of ouabain at different concentrations on growth of leukemia cellsZhongguo Dang Dai Er Ke Za Zhi200911425926219374807ContrerasRGFlores-MaldonadoCLázaroAShoshaniLFlores-BenitezDLarréICereijidoMOuabain Binding to Na+/K + -ATPase Relaxes Cell Attachment and Sends a SpecificSignal (NACos) to the NucleusJ Membr Biol2004198314715810.1007/s00232-004-0670-215216416JohanssonSLindholmPGullboJLarssonRBohlinLClaesonPCytotoxicity of digitoxin and related cardiac glycosides in human tumor cellsAnti-Cancer Drugs200112547548310.1097/00001813-200106000-0000911395576SchneiderRProscillaridin A immunoreactivity: its purification, transport in blood by a specific binding protein and its correlation with blood pressureClin Exp Hypertens19982059359910.3109/106419698090532379682915WinnickaKBielawskiKBielawskaASurazynskiAAntiproliferative activity of derivatives of ouabain, digoxin and proscillaridin a in human MCF-7 and MDA-MB-231 breast cancer cellsBiol Pharm Bull20083161131114010.1248/bpb.31.113118520043BelzGGBreithaupt-Gr√∂gler K, Osowski U: Treatment of congestive heart failure-current status of use of digitoxinEur J Clin Invest200131Suppl 2101711525233CastleMCPharmacokinetics of digoxin and digitoxin in humansEastern virginia med sch/med col hamp rd1977BøHMERTRøSETHAProlonged digitoxin half-life in very elderly patientsAge Ageing199827222222410.1093/ageing/27.2.22216296683SmithTWDigitalis toxicity: epidemiology and clinical use of serum concentration measurementsAm J Med197558447047610.1016/0002-9343(75)90118-71092162WilliamsJFJrPotterRDMathewBEffects of arrhythmia-producing concentrations of digitoxin on mechanical performance of cat myocardiumAm Heart J19831051212510.1016/0002-8703(83)90273-96849237HauxJDigitoxin is a potential anticancer agent for several types of cancerMed Hypotheses199953654354810.1054/mehy.1999.098510687899SchatzmannHJRassBInhibition of the active Na-K-transport and Na-K-activated membrane ATP-ase of erythrocyte stroma by ouabainHelv Physiol Pharmacol Acta196565C47C494220691ChangHTHuangJKWangJLChengJSLeeKCLoYKLiuCPChouKJChenWCSuWTamoxifen-induced increases in cytoplasmic free Ca-2+ levels in human breast cancer cellsBreast Cancer Res Treat200271212513110.1023/A:101380773164211881910KoumuraTNakamuraCNakagawaYRole of calcium-induced mitochondrial hydroperoxide in induction of apoptosis of RBL2H3 cells with eicosapentaenoic acid treatmentFree Radic Res200539101083108910.1080/1071576050026465416298733PigozziDTombalBDucretTVacherPGaillyPRole of store-dependent influx of Ca2+ and efflux of K + in apoptosis of CHO cellsCell Calcium200436542143010.1016/j.ceca.2004.04.00215451625XieZCaiTNa+/K + ATPase-Mediated Signal Transduction: From Protein Interaction to Cellular FunctionMol Interv20033315716810.1124/mi.3.3.15714993422WangXQApoptotic insults impair Na+, K + -ATPase activity as a mechanism of neuronal death mediated by concurrent ATP deficiency and oxidant stressJ Cell Sci20031162099211010.1242/jcs.0042012679386LiuLAbramowitzJAskariAAllenJCRole of caveolae in ouabain-induced proliferation of cultured vascular smooth muscle cells of the synthetic phenotypeAm J Physiol Heart Circ Physiol2004287H2173H218210.1152/ajpheart.00352.200415256370BarweSPNovel role for Na, K-ATPase in phosphatidylinositol 3-kinase signaling and suppression of cell motilityMol Biol Cell2005161082109410.1091/mbc.E04-05-042715616195LarreILazaroAContrerasRGBaldaMSMatterKFlores-MaldonadoCPonceAFlores-BenitezDRincon-HerediaRPadilla-BenavidesTOuabain modulates epithelial cell tight junctionProc Natl Acad Sci201010725113871139210.1073/pnas.100050010720534449BulavinDVFornaceAJJrp38 MAP kinase's emerging role as a tumor suppressorAdv Cancer Res2004929511815530558CagnolSChambardJ-CERK and cell death: mechanisms of ERK-induced cell death-apoptosis, autophagy and senescenceFEBS J2009277122119843174de PaulaRMLambTMBennettLBell-PedersenDA connection between MAPK pathways and circadian clocksCell Cycle20087172630263410.4161/cc.7.17.651618728391GotohINishidaESignal transductions by the MAP kinase cascadesNippon Rinsho1998567177917839702053HanJSunPThe pathways to tumor suppression via route p38Trends Biochem Sci200732836437110.1016/j.tibs.2007.06.00717624785KeyseSMDual-specificity MAP kinase phosphatases (MKPs) and cancerCancer Metastasis Rev200827225326110.1007/s10555-008-9123-118330678KhavariTARinnJRas/Erk MAPK signaling in epidermal homeostasis and neoplasiaCell Cycle20076232928293110.4161/cc.6.23.499818000402BolósVGasentJMLópez-TarruellaSGrandeEThe dual kinase complex FAK-Src as a promising therapeutic target in cancerOnco Targets Ther20103839720616959BurnhamMRBruce-StaskalPJHarteMTWeidowCLMaAWeedSABoutonAHRegulation of c-SRC activity and function by the adapter protein CASMol Cell Biol200020165865587810.1128/MCB.20.16.5865-5878.200010913170ChenC-YChangC-YLiuH-JLiaoM-HChangC-IHsuJ-LShihW-LApoptosis induction in BEFV-infected Vero and MDBK cells through Src-dependent JNK activation regulates caspase-3 and mitochondria pathwaysVet Res2010412Di FlorioACapursoGMilioneMPanzutoFGeremiaRDelle Fave G, Sette C: Src family kinase activity regulates adhesion, spreading and migration of pancreatic endocrine tumour cellsEndocr Relat Cancer200714111112410.1677/erc.1.0131817395980GötzRInter-cellular adhesion disruption and the RAS/RAF and beta-catenin signalling in lung cancer progressionCancer Cell Int200881710.1186/1475-2867-8-718492263LiuYGaoLGelmanIHSSeCKS/Gravin/AKAP12 attenuates expression of proliferative and angiogenic genes during suppression of v-Src-induced oncogenesisBMC Cancer20066110510.1186/1471-2407-6-10516638134OwenKAAbshireMYTilghmanRWCasanovaJEBoutonAHFAK Regulates Intestinal Epithelial Cell Survival and Proliferation during Mucosal Wound HealingPLoS One201168TianMSchiemannWThe TGF-β Paradox in Human Cancer: An UpdateFuture Oncol20095225927110.2217/14796694.5.2.25919284383WilliamsSPKarnezisTAchenMGStackerSATargeting lymphatic vessel functions through tyrosine kinasesJ Angiogenes Res20102131310.1186/2040-2384-2-1320698997LiangMCaiTTianJQuWXieZJFunctional characterization of Src-interacting Na/K-ATPase using RNA interference assayJ Biol Chem2006281197091971910.1074/jbc.M51224020016698801LangenhanJMEngleJMSlevinLKFayLRLuckerRWSmithKREndoMMModifying the glycosidic linkage in digitoxin analogs provides selective cytotoxinsBioorg Med Chem Lett200818267067310.1016/j.bmcl.2007.11.05818240383ZhouMO'DohertyGThe De novo Synthesis of Oligosaccharides - Application to the Medicinal Chemistry SAR Study of DigitoxinChemInform Abstract20083927ZhouMO'DohertyGThe De Novo Synthesis of Oligosaccharides: Application to the Medicinal Chemistry SAR-Study of DigitoxinCurr Top Med Chem2008811412510.2174/15680260878337882818289081MohammadiKKometianiPXieZAskariARole of protein kinase C in the signal pathways that link Na+/K + -ATPase to ERK1/2J Biol Chem2001276420504205610.1074/jbc.M10789220011562372YuanZNa/K-ATPase tethers phospholipase C and IP3 receptor into a calcium-regulatory complexMol Biol Cell2005164034404510.1091/mbc.E05-04-029515975899LiuLAskariAOn the importance and mechanism of amplification of digitalis signal through Na+/K+-ATPaseCell Mol Biol (Noisy-le-Grand)2006528283017535733KometianiPLiJGnudiLKahnBBAskariAXieZMultiple signal transduction pathways link Na+/K+-ATPase to growth-related genes in cardiac myocytes. The roles of Ras and mitogen-activated protein kinasesJ Biol Chem199827324152491525610.1074/jbc.273.24.152499614140PengMHuangLXieZHuangWHAskariAPartial inhibition of Na+/K + -ATPase by ouabain induces the Ca2 + -dependent expressions of early-response genes in cardiac myocytesJ Biol Chem199627117103721037810.1074/jbc.271.17.103728626609SchonerWScheiner-BobisGEndogenous and exogenous cardiac glycosides: their roles in hypertension, salt metabolism, and cell growthAm J Physiol Cell Physiol20072932C509C53610.1152/ajpcell.00098.200717494630Dueñas-GonzálezAGarcía-LópezPHerreraLAMedina-FrancoJLGonzález-FierroAMyrnaCThe prince and the pauper. A tale of anticancer targeted agentsMol Cancer2008718210.1186/1476-4598-7-8218947424ShullMMPughDGLingrelJBCharacterization of the human Na, K-ATPase alpha 2 gene and identification of intragenic restriction fragment length polymorphismsJ Biol Chem19892642917532175432477373JagielskaJSalgueroGSchiefferBBavendiekUDigitoxin elicits anti-inflammatory and vasoprotective properties in endothelial cells: Therapeutic implications for the treatment of atherosclerosis?Atherosclerosis2009206239039610.1016/j.atherosclerosis.2009.03.01919446813SrivastavaMEidelmanOZhangJPaweletzCCaohuyHYangQJacobsonKAHeldmanEHuangWJozwikCDigitoxin mimics gene therapy with CFTR and suppresses hypersecretion of IL-8 from cystic fibrosis lung epithelial cellsProc Natl Acad Sci USA2004101207693769810.1073/pnas.040203010115136726YangQHuangWJozwikCLinYGlasmanMCaohuyHSrivastavaMEspositoDGilletteWHartleyJCardiac glycosides inhibit TNF-alpha/NF-kappaB signaling by blocking recruitment of TNF receptor-associated death domain to the TNF receptorProc Natl Acad Sci USA2005102279631963610.1073/pnas.050409710215983368BambergerAMMilde-LangoschKR√∂ssingEGoemannCL√∂ningTExpression pattern of the AP-1 family in endometrial cancer: correlations with cell cycle regulatorsJ Cancer Res Clin Oncol2001127954555010.1007/s00432010025511570575DumesicPASchollFABarraganDIKhavariPAErk1/2 MAP kinases are required for epidermal G2/M progressionJ Cell Biol2009185340942210.1083/jcb.20080403819414607KaramouzisMVKonstantinopoulosPAPapavassiliouAGThe Activator Protein-1 Transcription Factor in Respiratory Epithelium CarcinogenesisMol Cancer Res20075210912010.1158/1541-7786.MCR-06-031117314269CrombezLMorrisMCDufortSAldrian-HerradaGNguyenQMc MasterGCollJ-LHeitzFDivitaGTargeting cyclin B1 through peptide-based delivery of siRNA prevents tumour growthNucleic Acids Res200937144559456910.1093/nar/gkp45119483097YuanJYanRKramerAEckerdtFRollerMKaufmannMStrebhardtKCyclin B1 depletion inhibits proliferation and induces apoptosis in human tumor cellsOncogene200423345843585210.1038/sj.onc.120775715208674ChaeSWSohnJHKimD-HChoiYJParkYLKimKChoYHPyoJ-SKimJHOverexpressions of Cyclin B1, cdc2, p16 and p53 in Human Breast Cancer: The Clinicopathologic Correlations and Prognostic ImplicationsYonsei Med J201152344545310.3349/ymj.2011.52.3.44521488187CooperWAKohonen-CorishMRJMcCaughanBKennedyCSutherlandRLLeeCSExpression and prognostic significance of cyclin B1 and cyclin A in non-small cell lung cancerHistopathology2009551283610.1111/j.1365-2559.2009.03331.x19614764EgloffAMWeissfeldJLandSRFinnOJEvaluation of anticyclin B1 serum antibody as a diagnostic and prognostic biomarker for lung cancerAnn N Y Acad Sci20051062294010.1196/annals.1358.00516461786KimD-HPrognostic implications of cyclin B1, p34cdc2, p27(Kip1) and p53 expression in gastric cancerYonsei Med J200748469470010.3349/ymj.2007.48.4.69417722244WongY-FCheungT-HTsaoGSWLoKWKYimS-FWangVWHeungMMSChanSCSChanLKYHoTWFGenome-wide gene expression profiling of cervical cancer in Hong Kong women by oligonucleotide microarrayInt J Cancer2006118102461246910.1002/ijc.2166016353136YoshidaTTanakaSMogiAShitaraYKuwanoHThe clinical significance of Cyclin B1 and Wee1 expression in non-small-cell lung cancerAnn Oncol200415225225610.1093/annonc/mdh07314760118De SouzaCPEllemKAGabrielliBGCentrosomal and cytoplasmic Cdc2/cyclin B1 activation precedes nuclear mitotic eventsExp Cell Res20002571112110.1006/excr.2000.487210854050StarkGTaylorWControl of the G2/M transitionMol Biotechnol200632322724810.1385/MB:32:3:22716632889DabrowskiAFilipAZgodzi ≈ ÑskiWDabrowskaMPola ≈ ÑskaDW√ ≥ jcikMZinkiewiczKWallnerGAssessment of prognostic significance of cytoplasmic survivin expression in advanced oesophageal cancerFolia Histochem Cytobiol200442316917215493578FalleniMPellegriniCMarchettiAOprandiBButtittaFBarassiFSantambrogioLCoggiGBosariSSurvivin gene expression in early-stage non-small cell lung cancerJ Pathol2003200562062610.1002/path.138812898598HeLHouMZhangJXuNChenPSubcellular localization of survivin in non-small cell lung cancerAi Zheng200928995596019728914Y-jRQ-yZExpression of survivin and its clinical significance in non-small cell lung cancerBeijing Da Xue Xue Bao200537550450716224523YangHFuJ-hHuYHuangW-zZhengBWangGRelationship between survivin expression and chemosensitivity of human lung cancer cellsZhonghua Yi Xue Za Zhi200787271934193717923022ZhouJ-MZhouJ-HDengZ-HZhengHJiangH-YCaoH-QExpression of survivin and proliferating cell nuclear antigen in human non-small cell lung cancerZhong Nan Da Xue Xue Bao Yi Xue Ban200530554454816320585BeardmoreVAAhonenLJGorbskyGJKallioMJSurvivin dynamics increases at centromeres during G2/M phase transition and is regulated by microtubule-attachment and Aurora B kinase activityJ Cell Sci2004117Pt 184033404215280424BeltramiEPlesciaJWilkinsonJCDuckettCSAltieriDCAcute Ablation of Survivin Uncovers p53-dependent Mitotic Checkpoint Functions and Control of Mitochondrial ApoptosisJ Biol Chem200427932077208410.1074/jbc.M30947920014581472MitaACMitaMMNawrockiSTGilesFJSurvivin: Key Regulator of Mitosis and Apoptosis and Novel Target for Cancer TherapeuticsClin Cancer Res200814165000500510.1158/1078-0432.CCR-08-074618698017WolaninKPiwockaKRole of survivin in mitosisPostepy Biochem2007531101817718383ColnaghiRWheatleySPLiaisons between survivin and Plk1 during cell division and cell deathJ Biol Chem201028529225922260410.1074/jbc.M109.06500320427271LensSMAWolthuisRMFKlompmakerRKauwJAgamiRBrummelkampTKopsGMedemaRHSurvivin is required for a sustained spindle checkpoint arrest in response to lack of tensionEMBO J200322122934294710.1093/emboj/cdg30712805209LiFAckermannEJBennettCFRothermelALPlesciaJTogninSVillaAMarchisioPCAltieriDCPleiotropic cell-division defects and apoptosis induced by interference with survivin functionNat Cell Biol19991846146610.1038/7024210587640LiFAmbrosiniGChuEYPlesciaJTogninSMarchisioPCAltieriDCControl of apoptosis and mitotic spindle checkpoint by survivinNature1998396671158058410.1038/251419859993CastedoMPerfettiniJ-LRoumierTAndreauKMedemaRKroemerGCell death by mitotic catastrophe: a molecular definitionOncogene200423162825283710.1038/sj.onc.120752815077146OkadaHMakTWPathways of apoptotic and non-apoptotic death in tumour cellsNat Rev Cancer20044859260310.1038/nrc141215286739VakifahmetogluHOlssonMZhivotovskyBDeath through a tragedy: mitotic catastropheCell Death Differ20081571153116210.1038/cdd.2008.4718404154Barre'BPerkinsNDA cell cycle regulatory network controlling NF-kappaB subunit activity and functionEMBO J200726234841485510.1038/sj.emboj.760189917962807BartekJLukasJChk1 and Chk2 kinases in checkpoint control and cancerCancer Cell2003342142910.1016/S1535-6108(03)00110-712781359ZhouB-BSBartekJTargeting the checkpoint kinases: chemosensitization versus chemoprotectionNat Rev Cancer20044321622510.1038/nrc129614993903ZhouB-BSSausvilleEADrug discovery targeting Chk1 and Chk2 kinasesProg Cell Cycle Res2003541342114593735HaasMWangHTianJXieZSrc-mediated inter-receptor cross-talk between the Na+/K + -ATPase and the epidermal growth factor receptor relays the signal from ouabain to mitogen-activated protein kinasesJ Biol Chem2002277186941870210.1074/jbc.M11135720011907028