Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Aug 15.
Published in final edited form as: Cell Cycle. 2010 May 1;9(9):1722–1728. doi: 10.4161/cc.9.9.11358

A therapeutic role for targeting c-Myc/Hif-1-dependent signaling pathways

Klaus Podar 1,2,*, Kenneth C Anderson 1
PMCID: PMC3155944  NIHMSID: NIHMS285091  PMID: 20404562

Abstract

Deregulated c-Myc occurs in ~30% of human cancers. Similarly, hypoxia-inducible factor (HIF) is commonly overexpressed in a variety of human malignancies. Under physiologic conditions, HIF inhibits c-Myc activity; however, when deregulated oncogenic c-Myc collaborates with HIF in inducing the expression of VEGF, PDK1 and hexokinase 2. Most of the knowledge of HIF derives from studies investigating a role of HIF under hypoxic conditions, however, HIF-1α stabilization is also found in normoxic conditions. Specifically, under hypoxic conditions HIF-1-mediated regulation of oncogenic c-Myc plays a pivotal role in conferring metabolic advantages to tumor cells as well as adaptation to the tumorigenic micromilieu. In addition, our own results show that under normoxic conditions oncogenic c-Myc is required for constitutive high HIF-1 protein levels and activity in Multiple Myeloma (MM) cells, thereby influencing VEGF secretion and angiogenic activity within the bone marrow microenvironment. Further studies are needed to delineate the functional relevance of HIF, MYC, and the HIF-MYC collaboration in MM and other malignancies, also integrating the tumor microenvironment and the cellular context. Importantly, early studies already demonstrate promising preclinical of novel agents, predominantly small molecules, which target c-Myc, HIF or both.

Keywords: HIF, c-Myc, targeted therapy, tumor microenvironment

Introduction

The basic helix-loop-helix leucine zipper (bHLH-LZ) transcription factor c-Myc regulates the expression of 10–15% of all genes of the genome.15 In addition, it directly regulates DNA replication.6 Deregulation of c-Myc occurs in ~30% of human cancers including breast, colon, cervical, small-cell lung cancer, osteosarcoma, glioblastoma, melanoma and myeloid leukemias; and is triggered by a variety of mechanisms including retroviral transduction, chromosomal translocation, gene amplification, as well as activation by hormones, their receptors, second messengers or transcriptional effectors.714 Myc-containing pathways are therefore an attractive target for cancer therapy.

Hypoxia-inducible factor (HIF) is a prominent transcription factor, which, similar to c-Myc, both promotes and represses the transcription of a broad range of genes involved not only in angiogenesis but also in other aspects of cancer biology including tumor cell survival and proliferation, migration, pH regulation, metabolism, drug and radiation resistance, and immune evasion.15 In addition, HIF-1 also promotes genetic instability by repressing transcription of the MSH2 and MSH6 genes via blockade of Myc-SP1 interaction. This effect does not require direct DNA binding or even the presence of HIF-1.16

In this review, we summarize the role of deregulated c-Myc and HIF in tumorigenesis in general, and MM in particular. Moreover, we focus on the proposed key pathophysiologic role of deregulated c-Myc-HIF collaboration in cell cycle progression, cell metabolism, DNA instability, as well as tumor angiogenesis. Finally, early preclinical data evaluating the role of derived novel agents targeting HIF-1 signaling are presented.

c-Myc

Functionally, c-Myc: (1) modulates cell cycle activity via blockade of transcription of cell cycle checkpoint genes (e.g., GADD45 and GADD153) and cyclin-dependent kinase (CDK) inhibitors, as well as promotion of cell cycle progression via activation of cyclin D1, D2, E1, A2, CDK4, cdc25A, E2F1 and E2F2; (2) stimulates differentiation and stem cell renewal; (3) induces as one of four factors the reprogramming of differentiated adult cells as one of four factors back to a pluripotent stem cell fate;17 (4) increases cell metabolism via regulation of glucose transporter GLUT1, hexokinase 2, phosphofruktokinase and enolase, as well as via glutamine transporters (ASCT2 and SLC7A25) and glutaminase (GLS) due to repression of miR-23a/b;1821 (5) contributes to chromosomal instability and ROS production;2224 and (6) sensitizes cells to apoptosis. This last effect is triggered via an ARF-Mdm2-p53 tumor suppressor pathway which is regulated by BMI1, TWIST1 and CUL7; the induction of the pro-apoptotic BH3-only protein BIM; and BAX-mediated suppression of anti-apoptotic proteins Bcl-2 and BCL-XL, followed by mitochondrial cytochrome c release. For example, Bcl-XL gain of function and p19 ARF loss of function cooperate oncogenetically with c-Myc to induce tumors in pancreatic islet cells.25,26

As a regulator of transcription, c-Myc can work both as a transcriptional activator as well as a transcriptional repressor of target genes. In vivo, Myc heterodimerizes with Myc-associated protein X (MAX). Binding to E-box (CACAGTG) sequences, the Myc-MAX complex activates or represses gene transcription or modulates chromatin.27,28 Transcription induced by the Myc-MAX complex is tightly regulated by the competitive complex formation of MAX with MAX dimerization proteins (MADs) Mad1, Mad2 (MAX interactor 1; MAXI1), Mad3, Mad4 and Mnt (Rox), followed by the subsequent recruitment of histone deacetylases (HDACs). Specifically, upon serum stimulation-enhanced c-Myc expression effectively competes with MAD for dimerization with MAX. c-Myc stability, and therefore expression and activity, is predominantly regulated by phosphorylation of residues Thr58 and Ser62. For example, activation of the Ras/MAPK/ERK pathway triggers phosphorylation of Ser62, thereby increasing Myc stability. When phosphorylated, Ser62 enables GSK3-mediated Thr58 phosphorylation, followed by FBW7/SCF/PP2A-dependent ubiquitylation and proteasomal degradation.2932 In addition, c-Myc activity is regulated by a short form of c-Myc,33 a cap-independent translation of c-Myc,34 and FOXO transcription factors.35

A requirement for transcriptional activation is the recruitment of co-activators including: positive transcription elongation factor b (P-TEFb); histone-acetyl-transferases (CREB-binding protein, CBP; p300; TRRAP; GCN5; TIP60); the ATPases TIP48 and TIP49; and the E3 ubiquitin ligase SKP2. Genes upregulated by Myc include HDAC2, CCND1, CCND2, CDK4, E2F2, LDHA and SHMT; whereas genes downregulated by Myc p21, p15, N-cadherins and integrins.14,27 In contrast, Myc expression decreases due to limited nutrients or high cellular density. In addition to E-box dependent transcription, c-Myc can also repress promoter activity by mechanisms independent from E-box binding sites, i.e., via nuclear factor Y (NF-Y), SP1 and Myc-interacting zinc finger 1 (MIZ1), TFII and yingyang-1. In summary, Myc-MAX heterodimers are predominant in proliferating cells; MAD-MAX and Mnt-MAX complexes are predominant in resting or differentiating cells.36,37

Besides directly coordinating multiple intracellular programs which mediate transformation, metabolic activity, and proliferation, c-Myc facilitates tumor cell growth via angiogenesis. Indeed, c-Myc has been postulated to be the master regulator of angiogenic factors, most prominently VEGF, and specifically of the angiogenic switch required for tumor progression and metastasis.3840 For example, targeted skin expression of c-Myc induces VEGF protein release and, in conjunction with hypoxia, further increases VEGF protein levels and angiogenesis.41 Moreover, lethality in c-myc (−/−) mouse embryos is due at least in part to the requirement for c-Myc for VEGF expression, since VEGF can partially rescue defects in differentiation and growth, including vasculogenesis.38,39

In MM, complex karyotypic abnormalities of the c-myc locus are present in the majority of MM cell lines (reviewed in ref. 42). Rearrangements of c-Myc are reported in nearly 40% of advanced human MM. Enforced expression of c-Myc using Ig enhancers with peak activity in plasma cells recapitulates some features of human MM in a murine model.43 Moreover, a conditional mouse model of sporadic MM strongly supports a pivotal role for c-Myc deregulation in the progression of benign MGUS to malignant MM.44 Functional mechanisms of c-Myc mediating disease progression are not fully elucidated, and a role of c-Myc in triggering MM BM angiogenesis may be a contributing factor.40,41,45

Approaches to target c-Myc activity include inhibition of c-Myc/Max dimerization and blockade of c-Myc/Max binding to the DNA binding motif. Small molecules inhibiting Myc-Max dimerization include IIA6B17, NY2267, 10058-F4, and 28RH-NCN-1; small molecules inhibiting Myc-Max binding to DNA include the pyrazolo [1,5] pyrimidine Mycro1, 1 (Mycro3), Myra-A and NSC308848.4649

HIF

HIF is composed of an oxygen-regulated-subunit and a constitutively expressed-subunit. Within the nucleus one of three HIF-1 subunits dimerizes with one of two isoforms of the subunit through the basic helix-loop-helix (bHLH) and PER-ARNT-SIM (PAS) A and B domains located in the N-terminal region of each subunit.50 These dimers bind to specific DNA sequences within the promoter, intron and/or enhancer regions of target genes called hypoxia-response elements (HREs), which are composed of 5′-RCGTG-3′ and recruit co-activators.51 Both HIF-1 and HIF-2 are overexpressed in common human cancers and their metastases.52 Importantly, increased levels of HIF-1 or HIF-2 are correlated with adverse prognosis in breast, cervical, endometrial, colorectal, NSCLC, ovarian, rectal, pancreatic, and prostate cancers.5359

Under normoxia, HIF expression is finely balanced between constitutive synthesis and proteasomal degradation.60 Expression and activity of HIF-1, HIF-2 and HIF-3 are regulated via posttranslational prolyl-hydroxylase domain proteins (PHDs: PHD1, PHD2, PHD2)-mediated prolyl-hydroxylation of the oxygen-dependent degradation domain (ODDD)61 and factor inhibiting HIF (FIH)-mediated asparaginyl-hydroxylation of the C-terminal end of HIF-1 and HIF-2 subunits.62 Specifically, hydroxylation on proline residue 402 and/or 564 of HIF-1 by PHD2 mediates HIF interaction with the von Hippel-Lindau tumor suppressor protein (VHL). In turn, VHL protein recruits an E3 ubiquitin-protein ligase and catalyzes poly-ubiquitination of HIF-1α, thereby triggering its inactivation by proteasomal degradation.63 Asparaginyl hydroxylation of HIF-1 and HIF-2 prevents binding of co-activators p300 and CREB-binding protein (CBP), thereby inhibiting HIF activity.61 Hydroxylation is dependent on the presence of oxygen, 2-oxoglutarate and cofactor FE2+. Due to their decrease under hypoxic conditions, first PHDs and later FIH become inactivated, followed by full HIF activation due to inhibition of proteasome-mediated degradation and nuclear accumulation.64,65 Moreover, transcriptional-activation domains (TADs), N-TAD and C-TAD in the C-terminus of HIF-1 and HIF-2 subunits regulate HIF transcriptional activity. Importantly and in contrast to N-TAD, C-TAD is inhibited by FIH hydroxylation thereby allowing bi-functional transcriptional activity of HIF-1.

Under hypoxia, reduced oxygen decreases the activity of PHDs and increases mitochondrial ROS release, thereby stabilizing HIF.66 Besides hypoxia, HIF activation is also triggered via oxygen-independent mechanisms: autocrine growth factor stimulation including cytokines, lipopolysaccharides, EGF, FGF2, IGF; loss of tumor-suppressor function (LOF) including ING4, p53, PTEN and VHL; gain of oncogene function (GOF) including Ras, Raf, Src, PI3K/Akt, mTOR, Myc15,52,6769; as well as reactive oxygen and nitrogen species.7072

In MM, prior studies found that bortezomib inhibits tumor adaptation by stimulating factor inhibiting HIF-1 (FIH);73 and that inhibitor of growth family member 4 (ING4) suppresses Hif-1 activity and angiogenesis under hypoxic conditions.74 Moreover, Asosingh et al. have suggested an important role of HIF-1 and BM hypoxia in MM progression.75

Both conventional and novel therapies inhibit HIF-1 activity including doxorubicin,76 geldanamycin/17-AAG, taxotere,77 topotecan78, imatinib,79,80 gefitinib, erlotinib, cetuximab,81,82 trastuzumab,67 rapamycin, temsirolimus, everolimus67,83 and histone deacetylase inhibitors.84,85 Functionally, these compounds inhibit m-TOR-dependent translation of HIF-1 mRNA into protein, target HIF-1 for proteasomal degradation, and inhibit either HIF-1 transactivation domain function or HIF-1 transcriptional activity by blocking DNA binding.

More specific HIF-1 inhibitors include: (1) YC-1,8691 and PX-478, which inhibit Hif-1 expression provide radiosensitization to cancer cells including glioma, squamous cell, prostate and pancreatic adenocarcinoma cell lines in vivo.9294 A phase I clinical trial using PX-478 in advanced solid tumors or lymphoma has now been initiated (http://www.clinicaltrials.gov). (2) Polyamide 2,95,96 which inhibits DNA binding and HIF-1 transcriptional activity by binding to the promoter of the HIF-1 target gene VEGF in nanomolar affinity; (3) NSC 50352, which blocks binding between the PAS-A domains of HIF-1 and HIF-1;97 (4) acriflavine, an antimicrobial used in WWII, which similarly to NSC 50352 decreases the interaction between endogenous HIF1 and HIF1 under hypoxic conditions;98 as well as (5) several compounds inhibiting HIF-PHD including oral FG-2216 and FG-4592;99,100 and the antisense oligo-nucleotide against HIF1 EZN-2968.101 A phase I clinical trial using EZN-2968 in advanced solid tumors or lymphoma has now been initiated (www.clinicaltrials.gov).

c-Myc and HIF

Under physiologic conditions Hif-1 inhibits c-Myc activity by direct interaction, induction of Mxi1, and stimulation of a proteasome-dependent pathway102104 however hypoaccentuates Myc-MAX-mediated transcriptional activation via Hif2-induced stabilization of Myc-MAX. Specifically, by increasing c-Myc/Max interactions, Hif-2 promotes c-Myc-mediated activation of cyclin D2, and triggers repression of p21 and p27.104,105

When deregulated, (oncogenic) c-Myc paradoxically collaborates with Hif-1. Specifically, c-Myc and Hif-1 collaborate in inducing the expression of PDK1 and hexokinase 2 followed by aerobic glycolysis (Warburg effect);68,104,106,107 and VEGF (angiogenesis).68,108

Based on c-Myc/HIF-1 associated metabolic differences in normal versus cancer cells induced by the Warburg effect, LDHA and PDK1 have been identified as potential novel therapeutic targets.109112 Growth inhibition was also triggered by inhibition of another c-Myc/Hif-1 target gene, the transferring receptor gene (TFRC).113,114 Moreover, echinomycin besides inhibiting DNA binding and transcriptional activity of HIF-1 also inhibits DNA binding of c-Myc/Max.115,116 Furthermore, promising results were observed by targeting Hif-1/c-Myc-dependent glutamine metabolism using antisense mRNA against glutaminase, acivicin and BPTES.117119

Given the importance of the BM microenvironment and BM angiogenesis in MM pathogenesis, we investigated the potential role of c-Myc and Hif-1 in triggering MM BM angiogenesis.120 Our studies demonstrate that c-Myc and Hif-1 are elevated in all MM cells, even under normoxic conditions. Moreover, we identified a link between oncogenic c-Myc and Hif-1 expression, VEGF production and poor prognosis in MM patients. The particular novelty of our data was the demonstration that Hif-1α protein level and activity in MM cells under normoxic conditions is regulated by oncogenic c-Myc to influence VEGF secretion and angiogenic activity. These data are consistent with previous studies in other tumor models.40,41,45,68,71 However, in contrast to previous data,121123 our data demonstrate c-Myc-dependent regulation of HIF-1 instead of the HIF-1α-dependent c-Myc regulation. In addition, our study identified c-Myc, HIF-1α, and the collaboration between both as a potential new therapeutic approach in MM. Using a drug screen we identified adaphostin, bortezomib, lenalidomide and enzastaurin to decrease HIF-1 levels and VEGF, dependent on c-Myc. Our in vitro results were then translated into in vivo demonstrating anti-angiogenic activity of our tool compound adaphostin124 using the zebrafish model. To further enhance the clinical relevance of this model, we are now establishing a xenograft zebrafish model for MM, similar to those previously described for other cancers.125 In summary, our study delineated a new c-Myc/Hif-1-dependent pathway, which triggers the release of VEGF and the induction of MM angiogenesis. It thereby supports the hypothesis that oncogenic c-Myc triggers MM progression, at least in part, by modulation of tumor angiogenesis.

Supporting our own data, inhibition of Hif-1 function by echinomycin or siRNA against Hif-1 resulted in enhanced sensitivity to melphalan in MM cells.126 Taken together these studies strongly indicate a potential therapeutic role for targeting the c-Myc/Hif-1 pathway in MM and other solid tumors and hematologic malignancies.

Conclusions

When acting alone, the transcription factors HIF and c-Myc play a complex roles in tumorigenesis by regulation of tumor cell metabolism, cell cycle progression and angiogenesis. Importantly, in contrast to HIF-induced c-Myc inhibition under physiologic conditions, deregulated c-Myc forms a functionally synergistic complex with HIF, which fine-tunes the cellular homeostasis of tumor cells i.e., adaptation to hypoxia, cell metabolism, pH regulation and angiogenesis.68,127 However, molecular sequelae underlying this synergism are elusive. One mechanism could be that high levels of c-Myc overcome the competing complex formation between c-Myc and Hif-1 through mass effect.68,128,129 Another mechanism could be the induction of genomic instability via a Hif-1-induced release of the transcription factor Sp1, which in turn inhibits c-Myc-induced expression of MSH2 and MSH6.130 Moreover, a recent study demonstrates that c-Myc decreases hypoxia-associated factor (HAF), which is responsible for oxygen-independent Hif-1 degradation.131,132 Finally, distinct binding partners, co-activators and repressors, may be differentially recruited dependent on the presence of deregulated c-Myc as well as the cellular and microenvironmental context.

Most of the knowledge of HIF derives from studies investigating a role of HIF under hypoxic conditions; however, HIF-1α stabilization is also found under normoxic conditions. To date, mechanisms mediating HIF stabilization are still elusive, NFKB has been proposed as a direct modular of HIF-1α expression.133 Our own data surprisingly demonstrate that deregulated c-Myc regulates Hif-1 levels in MM cells under normoxic conditions and thereby influences VEGF secretion and angiogenic activity within the bone marrow.120 In ongoing studies, we are therefore investigating c-Myc-dependent molecular mechanisms leading to the maintenance of Hif-1 levels in the MM cell including transcriptional, translational and post-translational modifications. Importantly, adaphostin inhibits both HIF and c-Myc in a clinically relevant in vivo murine xenograft model of human MM providing the framework for evolution of novel therapies functionally similar to adaphostin.

Although HIF is predominantly associated with tumor progression and poor clinical outcome, a recent study demonstrates that HIF-1 might also have tumor suppressive activity, via induction of mir-210 both in normoxic and hypoxic cells. Indeed, overexpression of mir-210 in pancreatic and head and neck cancer cell lines delayed tumor cell growth in a murine xenograft mouse model.134 Similarly and in contrast to our own data,120 Noguera recently showed that high Hif-1 levels correlate negatively with advanced clinical stage and tumor vascularization in neuroblastoma.135 One hypothesis for these discordant results may be tumor dependency, given the requirement for deregulated c-Myc in the tumorigenic c-Myc/Hif-1-complex, the anti-tumor effect in tumors lacking deregulated c-Myc remains to be evaluated. Moreover, further studies are needed to further delineate effect in cells that express Hif-1 and Hif-2, molecules with opposite effects.136

As our knowledge on the functions of c-Myc and HIF and their intimate punctional interaction grows, more efficient ways to target their molecular sequelae, dependent on the cellular context and on the tumor microenvironment, will be identified. Derived drugs have already demonstrated preclinical activity and are now in early clinical testing.

Acknowledgments

This work was supported by a National Institutes of Health Grants RO CA50947, PO-1 CA78378, and P50 CA100707; and the Doris Duke Distinguished Clinical Research Scientist Award (to K.C.A.).

Abbreviations

HIF

hypoxia-inducible factor

MM

multiple myeloma

VEGF

vascular endothelial growth factor

PHDs

prolyl-hydroxylase domain proteins

VHL

Hippel-Lindau tumor suppressor protein

References

  • 1.Fernandez PC, Frank SR, Wang L, Schroeder M, Liu S, Greene J, et al. Genomic targets of the human c-Myc protein. Genes Dev. 2003;17:1115–29. doi: 10.1101/gad.1067003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Li Z, Van Calcar S, Qu C, Cavenee WK, Zhang MQ, Ren B. A global transcriptional regulatory role for c-Myc in Burkitt’s lymphoma cells. Proc Natl Acad Sci USA. 2003;100:8164–9. doi: 10.1073/pnas.1332764100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.O’Connell BC, Cheung AF, Simkevich CP, Tam W, Ren X, Mateyak MK, et al. A large scale genetic analysis of c-Myc-regulated gene expression patterns. J Biol Chem. 2003;278:12563–73. doi: 10.1074/jbc.M210462200. [DOI] [PubMed] [Google Scholar]
  • 4.Zeller KI, Jegga AG, Aronow BJ, O’Donnell KA, Dang CV. An integrated database of genes responsive to the Myc oncogenic transcription factor: identification of direct genomic targets. Genome Biol. 2003;4:69. doi: 10.1186/gb-2003-4-10-r69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Patel JH, Loboda AP, Showe MK, Showe LC, McMahon SB. Analysis of genomic targets reveals complex functions of MYC. Nat Rev Cancer. 2004;4:562–8. doi: 10.1038/nrc1393. [DOI] [PubMed] [Google Scholar]
  • 6.Gusse M, Ghysdael J, Evan G, Soussi T, Mechali M. Translocation of a store of maternal cytoplasmic c-myc protein into nuclei during early development. Mol Cell Biol. 1989;9:5395–403. doi: 10.1128/mcb.9.12.5395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cheng AS, Jin VX, Fan M, Smith LT, Liyanarachchi S, Yan PS, et al. Combinatorial analysis of transcription factor partners reveals recruitment of c-MYC to estrogen receptor-alpha responsive promoters. Mol Cell. 2006;21:393–404. doi: 10.1016/j.molcel.2005.12.016. [DOI] [PubMed] [Google Scholar]
  • 8.Afar DE, Goga A, McLaughlin J, Witte ON, Sawyers CL. Differential complementation of Bcr-Abl point mutants with c-Myc. Science. 1994;264:424–6. doi: 10.1126/science.8153630. [DOI] [PubMed] [Google Scholar]
  • 9.He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, et al. Identification of c-MYC as a target of the APC pathway. Science. 1998;281:1509–12. doi: 10.1126/science.281.5382.1509. [DOI] [PubMed] [Google Scholar]
  • 10.Oster SK, Ho CS, Soucie EL, Penn LZ. The myc oncogene: MarvelouslY Complex. Adv Cancer Res. 2002;84:81–154. doi: 10.1016/s0065-230x(02)84004-0. [DOI] [PubMed] [Google Scholar]
  • 11.Roussel MF, Cleveland JL, Shurtleff SA, Sherr CJ. Myc rescue of a mutant CSF-1 receptor impaired in mitogenic signalling. Nature. 1991;353:361–3. doi: 10.1038/353361a0. [DOI] [PubMed] [Google Scholar]
  • 12.Barone MV, Courtneidge SA. Myc but not Fos rescue of PDGF signalling block caused by kinase-inactive Src. Nature. 1995;378:509–12. doi: 10.1038/378509a0. [DOI] [PubMed] [Google Scholar]
  • 13.Weng AP, Millholland JM, Yashiro-Ohtani Y, Arcangeli ML, Lau A, Wai C, et al. c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. Genes Dev. 2006;20:2096–109. doi: 10.1101/gad.1450406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer. 2008;8:976–90. doi: 10.1038/nrc2231. [DOI] [PubMed] [Google Scholar]
  • 15.Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3:721–32. doi: 10.1038/nrc1187. [DOI] [PubMed] [Google Scholar]
  • 16.Koshiji M, To KK, Hammer S, Kumamoto K, Harris AL, Huang LE, et al. HIF-1alpha induces genetic instability by transcriptionally downregulating MutSalpha expression. Mol Cell. 2005;17:793–803. doi: 10.1016/j.molcel.2005.02.015. [DOI] [PubMed] [Google Scholar]
  • 17.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 18.Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, et al. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem. 2000;275:21797–800. doi: 10.1074/jbc.C000023200. [DOI] [PubMed] [Google Scholar]
  • 19.Louro ID, Bailey EC, Li X, South LS, McKie-Bell PR, Yoder BK, et al. Comparative gene expression profile analysis of GLI and c-MYC in an epithelial model of malignant transformation. Cancer Res. 2002;62:5867–73. [PubMed] [Google Scholar]
  • 20.Yuneva M, Zamboni N, Oefner P, Sachidanandam R, Lazebnik Y. Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells. J Cell Biol. 2007;178:93–105. doi: 10.1083/jcb.200703099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–5. doi: 10.1038/nature07823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Herold S, Herkert B, Eilers M. Facilitating replication under stress: an oncogenic function of MYC? Nat Rev Cancer. 2009;9:441–4. doi: 10.1038/nrc2640. [DOI] [PubMed] [Google Scholar]
  • 23.Reimann M, Loddenkemper C, Rudolph C, Schildhauer I, Teichmann B, Stein H, et al. The Myc-evoked DNA damage response accounts for treatment resistance in primary lymphomas in vivo. Blood. 2007;110:2996–3004. doi: 10.1182/blood-2007-02-075614. [DOI] [PubMed] [Google Scholar]
  • 24.Vafa O, Wade M, Kern S, Beeche M, Pandita TK, Hampton GM, et al. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol Cell. 2002;9:1031–44. doi: 10.1016/s1097-2765(02)00520-8. [DOI] [PubMed] [Google Scholar]
  • 25.Brunner T, Martin SJ. c-Myc: where death and division collide. Cell Cycle. 2004;3:456–9. [PubMed] [Google Scholar]
  • 26.Finch A, Prescott J, Shchors K, et al. Bcl-xL gain of function and p19 ARF loss of function cooperate oncogenically with Myc in vivo by distinct mechanisms. Cancer Cell. 2006;10:113–20. doi: 10.1016/j.ccr.2006.06.017. [DOI] [PubMed] [Google Scholar]
  • 27.Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc proteins. Nat Rev Mol Cell Biol. 2005;6:635–45. doi: 10.1038/nrm1703. [DOI] [PubMed] [Google Scholar]
  • 28.Dang CV, O’Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F. The c-Myc target gene network. Semin Cancer Biol. 2006;16:253–64. doi: 10.1016/j.semcancer.2006.07.014. [DOI] [PubMed] [Google Scholar]
  • 29.Hann SR. Role of post-translational modifications in regulating c-Myc proteolysis, transcriptional activity and biological function. Semin Cancer Biol. 2006;16:288–302. doi: 10.1016/j.semcancer.2006.08.004. [DOI] [PubMed] [Google Scholar]
  • 30.Vervoorts J, Luscher-Firzlaff J, Luscher B. The ins and outs of MYC regulation by posttranslational mechanisms. J Biol Chem. 2006;281:34725–9. doi: 10.1074/jbc.R600017200. [DOI] [PubMed] [Google Scholar]
  • 31.Welcker M, Clurman BE. FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation. Nat Rev Cancer. 2008;8:83–93. doi: 10.1038/nrc2290. [DOI] [PubMed] [Google Scholar]
  • 32.Sears RC. The life cycle of C-myc: from synthesis to degradation. Cell Cycle. 2004;3:1133–7. [PubMed] [Google Scholar]
  • 33.Spotts GD, Patel SV, Xiao Q, Hann SR. Identification of downstream-initiated c-Myc proteins which are dominant-negative inhibitors of transactivation by full-length c-Myc proteins. Mol Cell Biol. 1997;17:1459–68. doi: 10.1128/mcb.17.3.1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cobbold LC, Spriggs KA, Haines SJ, Dobbyn HC, Hayes C, de Moor CH, et al. Identification of internal ribosome entry segment (IRES)-transacting factors for the Myc family of IRESs. Mol Cell Biol. 2008;28:40–9. doi: 10.1128/MCB.01298-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bouchard C, Marquardt J, Bras A, Medema RH, Eilers M. Myc-induced proliferation and transformation require Akt-mediated phosphorylation of FoxO proteins. EMBO J. 2004;23:2830–40. doi: 10.1038/sj.emboj.7600279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ayer DE, Kretzner L, Eisenman RN. Mad: a heterodimeric partner for Max that antagonizes Myc transcriptional activity. Cell. 1993;72:211–22. doi: 10.1016/0092-8674(93)90661-9. [DOI] [PubMed] [Google Scholar]
  • 37.Hurlin PJ, Queva C, Eisenman RN. Mnt, a novel Max-interacting protein is coexpressed with Myc in proliferating cells and mediates repression at Myc binding sites. Genes Dev. 1997;11:44–58. doi: 10.1101/gad.11.1.44. [DOI] [PubMed] [Google Scholar]
  • 38.Pelengaris S, Khan M, Evan GI. Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell. 2002;109:321–34. doi: 10.1016/s0092-8674(02)00738-9. [DOI] [PubMed] [Google Scholar]
  • 39.Pelengaris S, Khan M, Evan G. c-MYC: more than just a matter of life and death. Nat Rev Cancer. 2002;2:764–76. doi: 10.1038/nrc904. [DOI] [PubMed] [Google Scholar]
  • 40.Baudino TA, McKay C, Pendeville-Samain H, Nilsson JA, Maclean KH, White EL, et al. c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression. Genes Dev. 2002;16:2530–43. doi: 10.1101/gad.1024602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Knies-Bamforth UE, Fox SB, Poulsom R, Evan GI, Harris AL. c-Myc interacts with hypoxia to induce angiogenesis in vivo by a vascular endothelial growth factor-dependent mechanism. Cancer Res. 2004;64:6563–70. doi: 10.1158/0008-5472.CAN-03-3176. [DOI] [PubMed] [Google Scholar]
  • 42.Shou Y, Martelli ML, Gabrea A, Qi Y, Brents LA, Roschke A, et al. Diverse karyotypic abnormalities of the c-myc locus associated with c-myc dysregulation and tumor progression in multiple myeloma. Proc Natl Acad Sci USA. 2000;97:228–33. doi: 10.1073/pnas.97.1.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cheung WC, Kim JS, Linden M, Peng L, Van Ness B, Polakiewicz RD, et al. Novel targeted deregulation of c-Myc cooperates with Bcl-X(L) to cause plasma cell neoplasms in mice. J Clin Invest. 2004;113:1763–73. doi: 10.1172/JCI20369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chesi M, Robbiani DF, Sebag M, Chng WJ, Affer M, Tiedemann R, et al. AID-dependent activation of a MYC transgene induces multiple myeloma in a conditional mouse model of post-germinal center malignancies. Cancer Cell. 2008;13:167–80. doi: 10.1016/j.ccr.2008.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mezquita P, Parghi SS, Brandvold KA, Ruddell A. Myc regulates VEGF production in B cells by stimulating initiation of VEGF mRNA translation. Oncogene. 2005;24:889–901. doi: 10.1038/sj.onc.1208251. [DOI] [PubMed] [Google Scholar]
  • 46.Kiessling A, Sperl B, Hollis A, Eick D, Berg T. Selective inhibition of c-Myc/Max dimerization and DNA binding by small molecules. Chem Biol. 2006;13:745–51. doi: 10.1016/j.chembiol.2006.05.011. [DOI] [PubMed] [Google Scholar]
  • 47.Kiessling A, Wiesinger R, Sperl B, Berg T. Selective inhibition of c-Myc/Max dimerization by a pyrazolo[1,5-a]pyrimidine. ChemMedChem. 2007;2:627–30. doi: 10.1002/cmdc.200600294. [DOI] [PubMed] [Google Scholar]
  • 48.Mo H, Henriksson M. Identification of small molecules that induce apoptosis in a Myc-dependent manner and inhibit Myc-driven transformation. Proc Natl Acad Sci USA. 2006;103:6344–9. doi: 10.1073/pnas.0601418103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mo H, Vita M, Crespin M, Henriksson M. Myc overexpression enhances apoptosis induced by small molecules. Cell Cycle. 2006;5:2191–4. doi: 10.4161/cc.5.19.3320. [DOI] [PubMed] [Google Scholar]
  • 50.Chapman-Smith A, Lutwyche JK, Whitelaw ML. Contribution of the Per/Arnt/Sim (PAS) domains to DNA binding by the basic helix-loop-helix PAS transcriptional regulators. J Biol Chem. 2004;279:5353–62. doi: 10.1074/jbc.M310041200. [DOI] [PubMed] [Google Scholar]
  • 51.Pugh CW, Tan CC, Jones RW, Ratcliffe PJ. Functional analysis of an oxygen-regulated transcriptional enhancer lying 3′ to the mouse erythropoietin gene. Proc Natl Acad Sci USA. 1991;88:10553–7. doi: 10.1073/pnas.88.23.10553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59:5830–5. [PubMed] [Google Scholar]
  • 53.Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev. 2007;26:225–39. doi: 10.1007/s10555-007-9055-1. [DOI] [PubMed] [Google Scholar]
  • 54.Tan EY, Campo L, Han C, Turley H, Pezzella F, Gatter KC. Cytoplasmic location of factor-inhibiting hypoxia-inducible factor is associated with an enhanced hypoxic response and a shorter survival in invasive breast cancer. Breast Cancer Res. 2007;9:89. doi: 10.1186/bcr1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Silva P, Slevin NJ, Sloan P, Valentine H, Cresswell J, Ryder D, et al. Prognostic significance of tumor hypoxia inducible factor-1alpha expression for outcome after radiotherapy in oropharyngeal cancer. Int J Radiat Oncol Biol Phys. 2008;72:1551–9. doi: 10.1016/j.ijrobp.2008.07.051. [DOI] [PubMed] [Google Scholar]
  • 56.Nanni S, Benvenuti V, Grasselli A, Priolo C, Aiello A, Mattiussi S, et al. Endothelial NOS, estrogen receptor beta, and HIFs cooperate in the activation of a prognostic transcriptional pattern in aggressive human prostate cancer. J Clin Invest. 2009;119:1093–108. doi: 10.1172/JCI35079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Griffiths EA, Pritchard SA, Valentine HR, Whitchelo N, Bishop PW, Ebert MP, et al. Hypoxia-inducible factor-1alpha expression in the gastric carcinogenesis sequence and its prognostic role in gastric and gastro-oesophageal adenocarcinomas. Br J Cancer. 2007;96:95–103. doi: 10.1038/sj.bjc.6603524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA. Proliferation of immature tumor vessels is a novel marker of clinical progression in prostate cancer. Cancer Res. 2009;69:4708–15. doi: 10.1158/0008-5472.CAN-08-4417. [DOI] [PubMed] [Google Scholar]
  • 59.Daponte A, Ioannou M, Mylonis I, Simos G, Minas M, Messinis IE, et al. Prognostic significance of Hypoxia-Inducible Factor 1alpha (HIF-1alpha) expression in serous ovarian cancer: an immunohistochemical study. BMC Cancer. 2008;8:335. doi: 10.1186/1471-2407-8-335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brahimi-Horn MC, Chiche J, Pouyssegur J. Hypoxia signalling controls metabolic demand. Curr Opin Cell Biol. 2007;19:223–9. doi: 10.1016/j.ceb.2007.02.003. [DOI] [PubMed] [Google Scholar]
  • 61.Schofield CJ, Ratcliffe PJ. Oxygen sensing by HIF hydroxylases. Nat Rev Mol Cell Biol. 2004;5:343–54. doi: 10.1038/nrm1366. [DOI] [PubMed] [Google Scholar]
  • 62.Peet D, Linke S. Regulation of HIF: asparaginyl hydroxylation. Novartis Found Symp. 2006;272:37–49. [PubMed] [Google Scholar]
  • 63.Kaelin WG., Jr The von Hippel-Lindau tumour suppressor protein: O2 sensing and cancer. Nat Rev Cancer. 2008;8:865–73. doi: 10.1038/nrc2502. [DOI] [PubMed] [Google Scholar]
  • 64.Koivunen P, Hirsila M, Gunzler V, Kivirikko KI, Myllyharju J. Catalytic properties of the asparaginyl hydroxylase (FIH) in the oxygen sensing pathway are distinct from those of its prolyl 4-hydroxylases. J Biol Chem. 2004;279:9899–904. doi: 10.1074/jbc.M312254200. [DOI] [PubMed] [Google Scholar]
  • 65.Kallio PJ, Okamoto K, O’Brien S, Carrero P, Makino Y, Tanaka H, et al. Signal transduction in hypoxic cells: Inducible nuclear translocation and recruitment of the CBP/p300 coactivator by the hypoxia-inducible factor-1alpha. EMBO J. 1998;17:6573–86. doi: 10.1093/emboj/17.22.6573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lu H, Dalgard CL, Mohyeldin A, McFate T, Tait AS, Verma A. Reversible inactivation of HIF-1 prolyl hydroxylases allows cell metabolism to control basal HIF-1. J Biol Chem. 2005;280:41928–39. doi: 10.1074/jbc.M508718200. [DOI] [PubMed] [Google Scholar]
  • 67.Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol Cell Biol. 2001;21:3995–4004. doi: 10.1128/MCB.21.12.3995-4004.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dang CV, Kim JW, Gao P, Yustein J. The interplay between MYC and HIF in cancer. Nat Rev Cancer. 2008;8:51–6. doi: 10.1038/nrc2274. [DOI] [PubMed] [Google Scholar]
  • 69.Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–5. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
  • 70.Quintero M, Brennan PA, Thomas GJ, Moncada S. Nitric oxide is a factor in the stabilization of hypoxia-inducible factor-1alpha in cancer: role of free radical formation. Cancer Res. 2006;66:770–4. doi: 10.1158/0008-5472.CAN-05-0333. [DOI] [PubMed] [Google Scholar]
  • 71.Gao P, Zhang H, Dinavahi R, Li F, Xiang Y, Raman V, et al. HIF-dependent antitumorigenic effect of antioxidants in vivo. Cancer Cell. 2007;12:230–8. doi: 10.1016/j.ccr.2007.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dewhirst MW. Relationships between cycling hypoxia, HIF-1, angiogenesis and oxidative stress. Radiat Res. 2009;172:653–65. doi: 10.1667/RR1926.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Shin DH, Chun YS, Lee DS, Huang LE, Park JW. Bortezomib inhibits tumor adaptation to hypoxia by stimulating the FIH-mediated repression of hypoxia-inducible factor-1. Blood. 2008;111:3131–6. doi: 10.1182/blood-2007-11-120576. [DOI] [PubMed] [Google Scholar]
  • 74.Colla S, Tagliaferri S, Morandi F, Lunghi P, Donofrio G, Martorana D, et al. The new tumor-suppressor gene inhibitor of growth family member 4 (ING4) regulates the production of proangiogenic molecules by myeloma cells and suppresses hypoxia-inducible factor-1alpha (HIF-1alpha) activity: involvement in myeloma-induced angiogenesis. Blood. 2007;110:4464–75. doi: 10.1182/blood-2007-02-074617. [DOI] [PubMed] [Google Scholar]
  • 75.Asosingh K, De Raeve H, de Ridder M, Storme GA, Willems A, Van Riet I, et al. Role of the hypoxic bone marrow microenvironment in 5T2MM murine myeloma tumor progression. Haematologica. 2005;90:810–7. [PubMed] [Google Scholar]
  • 76.Lee K, Qian DZ, Rey S, Wei H, Liu JO, Semenza GL. Anthracycline chemotherapy inhibits HIF-1 transcriptional activity and tumor-induced mobilization of circulating angiogenic cells. Proc Natl Acad Sci USA. 2009;106:2353–8. doi: 10.1073/pnas.0812801106. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 77.Escuin D, Kline ER, Giannakakou P. Both microtubule-stabilizing and microtubule-destabilizing drugs inhibit hypoxia-inducible factor-1alpha accumulation and activity by disrupting microtubule function. Cancer Res. 2005;65:9021–8. doi: 10.1158/0008-5472.CAN-04-4095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rapisarda A, Uranchimeg B, Scudiero DA, Selby M, Sausville EA, Shoemaker RH, et al. Identification of small molecule inhibitors of hypoxia-inducible factor 1 transcriptional activation pathway. Cancer Res. 2002;62:4316–24. [PubMed] [Google Scholar]
  • 79.Mayerhofer M, Valent P, Sperr WR, Griffin JD, Sillaber C. BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor-1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood. 2002;100:3767–75. doi: 10.1182/blood-2002-01-0109. [DOI] [PubMed] [Google Scholar]
  • 80.Litz J, Krystal GW. Imatinib inhibits c-Kit-induced hypoxia-inducible factor-1alpha activity and vascular endothelial growth factor expression in small cell lung cancer cells. Mol Cancer Ther. 2006;5:1415–22. doi: 10.1158/1535-7163.MCT-05-0503. [DOI] [PubMed] [Google Scholar]
  • 81.Luwor RB, Lu Y, Li X, Mendelsohn J, Fan Z. The antiepidermal growth factor receptor monoclonal antibody cetuximab/C225 reduces hypoxia-inducible factor-1alpha, leading to transcriptional inhibition of vascular endothelial growth factor expression. Oncogene. 2005;24:4433–41. doi: 10.1038/sj.onc.1208625. [DOI] [PubMed] [Google Scholar]
  • 82.Pore N, Jiang Z, Gupta A, Cerniglia G, Kao GD, Maity A. EGFR tyrosine kinase inhibitors decrease VEGF expression by both hypoxia-inducible factor (HIF)-1-independent and HIF-1-dependent mechanisms. Cancer Res. 2006;66:3197–204. doi: 10.1158/0008-5472.CAN-05-3090. [DOI] [PubMed] [Google Scholar]
  • 83.Majumder PK, Febbo PG, Bikoff R, Berger R, Xue Q, McMahon LM, et al. mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways. Nat Med. 2004;10:594–601. doi: 10.1038/nm1052. [DOI] [PubMed] [Google Scholar]
  • 84.Liang D, Kong X, Sang N. Effects of histone deacetylase inhibitors on HIF-1. Cell Cycle. 2006;5:2430–5. doi: 10.4161/cc.5.21.3409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Verheul HM, Salumbides B, Van Erp K, Hammers H, Qian DZ, Sanni T, et al. Combination strategy targeting the hypoxia inducible factor-1alpha with mammalian target of rapamycin and histone deacetylase inhibitors. Clin Cancer Res. 2008;14:3589–97. doi: 10.1158/1078-0432.CCR-07-4306. [DOI] [PubMed] [Google Scholar]
  • 86.Yeo EJ, Chun YS, Cho YS, Kim J, Lee JC, Kim MS, et al. YC-1: a potential anticancer drug targeting hypoxia-inducible factor 1. J Natl Cancer Inst. 2003;95:516–25. doi: 10.1093/jnci/95.7.516. [DOI] [PubMed] [Google Scholar]
  • 87.Kato H, Inoue T, Asanoma K, Nishimura C, Matsuda T, Wake N. Induction of human endome-trial cancer cell senescence through modulation of HIF-1alpha activity by EGLN1. Int J Cancer. 2006;118:1144–53. doi: 10.1002/ijc.21488. [DOI] [PubMed] [Google Scholar]
  • 88.Sun HL, Liu YN, Huang YT, Pan SL, Huang DY, Guh JH, et al. YC-1 inhibits HIF-1 expression in prostate cancer cells: contribution of Akt/NFkappaB signaling to HIF-1alpha accumulation during hypoxia. Oncogene. 2007;26:3941–51. doi: 10.1038/sj.onc.1210169. [DOI] [PubMed] [Google Scholar]
  • 89.Zhao Q, Du J, Gu H, Teng X, Zhang Q, Qin H, et al. Effects of YC-1 on hypoxia-inducible factor 1-driven transcription activity, cell proliferative vitality and apoptosis in hypoxic human pancreatic cancer cells. Pancreas. 2007;34:242–7. doi: 10.1097/01.mpa.0000250135.95144.b6. [DOI] [PubMed] [Google Scholar]
  • 90.Hiraga T, Kizaka-Kondoh S, Hirota K, Hiraoka M, Yoneda T. Hypoxia and hypoxia-inducible factor-1 expression enhance osteolytic bone metastases of breast cancer. Cancer Res. 2007;67:4157–63. doi: 10.1158/0008-5472.CAN-06-2355. [DOI] [PubMed] [Google Scholar]
  • 91.Harada H, Itasaka S, Zhu Y, Zeng L, Xie X, Morinibu A, et al. Treatment regimen determines whether an HIF-1 inhibitor enhances or inhibits the effect of radiation therapy. Br J Cancer. 2009;100:747–57. doi: 10.1038/sj.bjc.6604939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Palayoor ST, Mitchell JB, Cerna D, Degraff W, John-Aryankalayil M, Coleman CN. PX-478, an inhibitor of hypoxia-inducible factor-1alpha, enhances radio-sensitivity of prostate carcinoma cells. Int J Cancer. 2008;123:2430–7. doi: 10.1002/ijc.23807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Schwartz DL, Powis G, Thitai-Kumar A, He Y, Bankson J, Williams R, et al. The selective hypoxia inducible factor-1 inhibitor PX-478 provides in vivo radiosensitization through tumor stromal effects. Mol Cancer Ther. 2009;8:947–58. doi: 10.1158/1535-7163.MCT-08-0981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Otrock ZK, Hatoum HA, Awada AH, Ishak RS, Shamseddine AI. Hypoxia-inducible factor in cancer angiogenesis: structure, regulation and clinical perspectives. Crit Rev Oncol Hematol. 2009;70:93–102. doi: 10.1016/j.critrevonc.2009.01.001. [DOI] [PubMed] [Google Scholar]
  • 95.Vinson C. A rationally designed small molecule that inhibits the HIF-1alpha-ARNT heterodimer from binding to DNA in vivo. Sci STKE. 2005;2005:23. doi: 10.1126/stke.2842005pe23. [DOI] [PubMed] [Google Scholar]
  • 96.Olenyuk BZ, Zhang GJ, Klco JM, Nickols NG, Kaelin WG, Jr, Dervan PB. Inhibition of vascular endothelial growth factor with a sequence-specific hypoxia response element antagonist. Proc Natl Acad Sci USA. 2004;101:16768–73. doi: 10.1073/pnas.0407617101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Park EJ, Kong D, Fisher R, Cardellina J, Shoemaker RH, Melillo G. Targeting the PAS-A domain of HIF-1alpha for development of small molecule inhibitors of HIF-1. Cell Cycle. 2006;5:1847–53. doi: 10.4161/cc.5.16.3019. [DOI] [PubMed] [Google Scholar]
  • 98.Lee K, Zhang H, Qian DZ, Rey S, Liu JO, Semenza GL. Acriflavine inhibits HIF-1 dimerization, tumor growth and vascularization. Proc Natl Acad Sci USA. 2009;106:17910–5. doi: 10.1073/pnas.0909353106. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 99.Hsieh MM, Linde NS, Wynter A, Metzger M, Wong C, Langsetmo I, et al. HIF prolyl hydroxylase inhibition results in endogenous erythropoietin induction, erythrocytosis and modest fetal hemoglobin expression in rhesus macaques. Blood. 2007;110:2140–7. doi: 10.1182/blood-2007-02-073254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cases A. The latest advances in kidney diseases and related disorders. Drug News Perspect. 2007;20:647–54. [PubMed] [Google Scholar]
  • 101.Greenberger LM, Horak ID, Filpula D, Sapra P, Westergaard M, Frydenlund HF, et al. A RNA antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhibits tumor cell growth. Mol Cancer Ther. 2008;7:3598–608. doi: 10.1158/1535-7163.MCT-08-0510. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, et al. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell. 2007;11:407–20. doi: 10.1016/j.ccr.2007.04.001. [DOI] [PubMed] [Google Scholar]
  • 103.Corn PG, Ricci MS, Scata KA, Arsham AM, Simon MC, Dicker DT, et al. Mxi1 is induced by hypoxia in a HIF-1-dependent manner and protects cells from c-Myc-induced apoptosis. Cancer Biol Ther. 2005;4:1285–94. doi: 10.4161/cbt.4.11.2299. [DOI] [PubMed] [Google Scholar]
  • 104.Gordan JD, Thompson CB, Simon MC. HIF and c-Myc: sibling rivals for control of cancer cell metabolism and proliferation. Cancer Cell. 2007;12:108–13. doi: 10.1016/j.ccr.2007.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gordan JD, Bertout JA, Hu CJ, Diehl JA, Simon MC. HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell. 2007;11:335–47. doi: 10.1016/j.ccr.2007.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Warburg O. On respiratory impairment in cancer cells. Science. 1956;124:269–70. [PubMed] [Google Scholar]
  • 107.Warburg O. On the origin of cancer cells. Science. 1956;123:309–14. doi: 10.1126/science.123.3191.309. [DOI] [PubMed] [Google Scholar]
  • 108.Kim JW, Dang CV. Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 2006;66:8927–30. doi: 10.1158/0008-5472.CAN-06-1501. [DOI] [PubMed] [Google Scholar]
  • 109.Shim H, Dolde C, Lewis BC, Wu CS, Dang G, Jungmann RA, et al. c-Myc transactivation of LDH-A: implications for tumor metabolism and growth. Proc Natl Acad Sci USA. 1997;94:6658–63. doi: 10.1073/pnas.94.13.6658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Bonnet S, Archer SL, Allalunis-Turner J, Haromy A, Beaulieu C, Thompson R, et al. A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell. 2007;11:37–51. doi: 10.1016/j.ccr.2006.10.020. [DOI] [PubMed] [Google Scholar]
  • 111.Fantin VR, St-Pierre J, Leder P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology and tumor maintenance. Cancer Cell. 2006;9:425–34. doi: 10.1016/j.ccr.2006.04.023. [DOI] [PubMed] [Google Scholar]
  • 112.Deck LM, Royer RE, Chamblee BB, Hernandez VM, Malone RR, Torres JE, et al. Selective inhibitors of human lactate dehydrogenases and lactate dehydrogenase from the malarial parasite Plasmodium falciparum. J Med Chem. 1998;41:3879–87. doi: 10.1021/jm980334n. [DOI] [PubMed] [Google Scholar]
  • 113.Kasibhatla S, Jessen KA, Maliartchouk S, Wang JY, English NM, Drewe J, et al. A role for transferrin receptor in triggering apoptosis when targeted with gambogic acid. Proc Natl Acad Sci USA. 2005;102:12095–100. doi: 10.1073/pnas.0406731102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Pandey MK, Sung B, Ahn KS, Kunnumakkara AB, Chaturvedi MM, Aggarwal BB. Gambogic acid, a novel ligand for transferrin receptor, potentiates TNF-induced apoptosis through modulation of the nuclear factor-kappaB signaling pathway. Blood. 2007;110:3517–25. doi: 10.1182/blood-2007-03-079616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kong D, Park EJ, Stephen AG, Calvani M, Cardellina JH, Monks A, et al. Echinomycin, a small-molecule inhibitor of hypoxia-inducible factor-1 DNA-binding activity. Cancer Res. 2005;65:9047–55. doi: 10.1158/0008-5472.CAN-05-1235. [DOI] [PubMed] [Google Scholar]
  • 116.Vlaminck B, Toffoli S, Ghislain B, Demazy C, Raes M, Michiels C. Dual effect of echinomycin on hypoxia-inducible factor-1 activity under normoxic and hypoxic conditions. Febs J. 2007;274:5533–42. doi: 10.1111/j.1742-4658.2007.06072.x. [DOI] [PubMed] [Google Scholar]
  • 117.Perez-Gomez C, Campos-Sandoval JA, Alonso FJ, Segura JA, Manzanares E, Ruiz-Sánchez P, et al. Co-expression of glutaminase K and L isoenzymes in human tumour cells. Biochem J. 2005;386:535–42. doi: 10.1042/BJ20040996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lobo C, Ruiz-Bellido MA, Aledo JC, Marquez J, Nunez De Castro I, Alonso FJ. Inhibition of glutaminase expression by antisense mRNA decreases growth and tumourigenicity of tumour cells. Biochem J. 2000;348:257–61. [PMC free article] [PubMed] [Google Scholar]
  • 119.Hidalgo M, Rodriguez G, Kuhn JG, Brown T, Weiss G, MacGovren JP, et al. A Phase I and pharmacological study of the glutamine antagonist acivicin with the amino acid solution aminosyn in patients with advanced solid malignancies. Clin Cancer Res. 1998;4:2763–70. [PubMed] [Google Scholar]
  • 120.Zhang J, Sattler M, Tonon G, Grabher C, Lababidi S, Zimmerhackl A, et al. Targeting angiogenesis via a c-Myc/hypoxia-inducible factor-1alpha-dependent pathway in multiple myeloma. Cancer Res. 2009;69:5082–90. doi: 10.1158/0008-5472.CAN-08-4603. [DOI] [PubMed] [Google Scholar]
  • 121.An WG, Kanekal M, Simon MC, Maltepe E, Blagosklonny MV, Neckers LM. Stabilization of wild-type p53 by hypoxia-inducible factor 1alpha. Nature. 1998;392:405–8. doi: 10.1038/32925. [DOI] [PubMed] [Google Scholar]
  • 122.Santore MT, McClintock DS, Lee VY, Budinger GR, Chandel NS. Anoxia-induced apoptosis occurs through a mitochondria-dependent pathway in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2002;282:727–34. doi: 10.1152/ajplung.00281.2001. [DOI] [PubMed] [Google Scholar]
  • 123.Jiang BH, Agani F, Passaniti A, Semenza GL. V-SRC induces expression of hypoxia-inducible factor 1 (HIF-1) and transcription of genes encoding vascular endothelial growth factor and enolase 1: involvement of HIF-1 in tumor progression. Cancer Res. 1997;57:5328–35. [PubMed] [Google Scholar]
  • 124.Avramis IA, Christodoulopoulos G, Suzuki A, Laug WE, Gonzalez-Gomez I, McNamara G, et al. In vitro and in vivo evaluations of the tyrosine kinase inhibitor NSC 680410 against human leukemia and glioblastoma cell lines. Cancer Chemother Pharmacol. 2002;50:479–89. doi: 10.1007/s00280-002-0507-6. [DOI] [PubMed] [Google Scholar]
  • 125.Nicoli S, Ribatti D, Cotelli F, Presta M. Mammalian tumor xenografts induce neovascularization in zebrafish embryos. Cancer Res. 2007;67:2927–31. doi: 10.1158/0008-5472.CAN-06-4268. [DOI] [PubMed] [Google Scholar]
  • 126.Hu Y, Kirito K, Yoshida K, Mitsumori T, Nakajima K, Nozaki Y, et al. Inhibition of hypoxia-inducible factor-1 function enhances the sensitivity of multiple myeloma cells to melphalan. Mol Cancer Ther. 2009;8:2329–38. doi: 10.1158/1535-7163.MCT-09-0150. [DOI] [PubMed] [Google Scholar]
  • 127.Yoo YG, Hayashi M, Christensen J, Huang LE. An essential role of the HIF-1alpha-c-Myc axis in malignant progression. Ann NY Acad Sci. 2009;1177:198–204. doi: 10.1111/j.1749-6632.2009.05043.x. [DOI] [PubMed] [Google Scholar]
  • 128.Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM, et al. Hypoxia-inducible factor-1alpha is a positive factor in solid tumor growth. Cancer Res. 2000;60:4010–5. [PubMed] [Google Scholar]
  • 129.Blouw B, Song H, Tihan T, Bosze J, Ferrara N, Gerber HP, et al. The hypoxic response of tumors is dependent on their microenvironment. Cancer Cell. 2003;4:133–46. doi: 10.1016/s1535-6108(03)00194-6. [DOI] [PubMed] [Google Scholar]
  • 130.Koshiji M, Kageyama Y, Pete EA, Horikawa I, Barrett JC, Huang LE. HIF-1alpha induces cell cycle arrest by functionally counteracting Myc. EMBO J. 2004;23:1949–56. doi: 10.1038/sj.emboj.7600196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Koh MY, Powis G. HAF: the new player in oxygen-independent HIF-1alpha degradation. Cell Cycle. 2009;8:1359–66. doi: 10.4161/cc.8.9.8303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Koh MY, Darnay BG, Powis G. Hypoxia-associated factor, a novel E3-ubiquitin ligase, binds and ubiquitinates hypoxia-inducible factor 1alpha, leading to its oxygen-independent degradation. Mol Cell Biol. 2008;28:7081–95. doi: 10.1128/MCB.00773-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.van Uden P, Kenneth NS, Rocha S. Regulation of hypoxia-inducible factor-1alpha by NFkappaB. Biochem J. 2008;412:477–84. doi: 10.1042/BJ20080476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Huang X, Ding L, Bennewith KL, Tong RT, Welford SM, Ang KK, et al. Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol Cell. 2009;35:856–67. doi: 10.1016/j.molcel.2009.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Noguera R, Fredlund E, Piqueras M, Pietras A, Beckman S, Navarro S, et al. HIF-1alpha and HIF-2alpha are differentially regulated in vivo in neuroblastoma: high HIF-1alpha correlates negatively to advanced clinical stage and tumor vascularization. Clin Cancer Res. 2009;15:7130–6. doi: 10.1158/1078-0432.CCR-09-0223. [DOI] [PubMed] [Google Scholar]
  • 136.Huang LE. Carrot and stick: HIF-alpha engages c-Myc in hypoxic adaptation. Cell Death Differ. 2008;15:672–7. doi: 10.1038/sj.cdd.4402302. [DOI] [PubMed] [Google Scholar]

RESOURCES