Tumor hypoxia is a well-described phenomenon during the progression of solid tumors affecting cell signaling pathways and cell metabolism; however, its role in hematologic malignancies has not been given the same attention in the literature. Therefore, this review focuses on the comparative differences between solid and hematologic malignancies with emphasis on the role of hypoxia during tumorigenesis and progression. In addition, contribution of the bone marrow and angiogenic environment are also discussed. Insight is provided into the role of hypoxia in metastatic spread, stemness, and drug resistance in hematologic conditions. Finally, emerging therapeutic strategies such as small-molecule prodrugs and hypoxia-inducible factor (HIF) targeting approaches are outlined to combat hypoxic cells and/or adaptive mechanisms in the treatment of hematologic malignancies. Mol Cancer Res; 12(10); 1347–54. ©2014 AACR.

Hypoxia is a level of oxygen tension below the physiologic level; physiologic range varies due to diversified blood vessel network in different organs (1). Hypoxic conditions develop during cancer progression due to rapidly proliferating tumor cells that reduce O2 diffusion as well as impaired perfusion of the abnormal blood vessels in the tumor. The oxygen level in hypoxic tumor tissues is found to be less than 1.3% O2, far below the physiologic oxygenation level (5%–10% O2; ref. 2).

Cellular adaptation to hypoxia is mediated through protein stabilization of hypoxia-inducible factor (HIF) subunits (HIF1α, HIF2α and HIF3α) that are regulated by prolyl hydroxylase domain (PHD) and factor-inhibiting HIF1 (FIH-1) enzymes. In oxygenated cells, HIFα subunits are hydroxylated by PHD and FIH-1, poly-ubiquitinated, and degraded by the proteasome; in hypoxia, the PHD enzymes lose their activity and the HIF degradation is halted. The stabilized, non-hydroxylated HIFα translocate to the nucleus, where they dimerize with constitutively expressed HIFβ subunit, and bind to DNA to initiate gene transcription of the adaptive pathways (3, 4). Cellular adaptations to hypoxia can also occur by HIF-independent mechanisms including mammalian target of rapamycin (mTOR) kinase, unfolded protein response (UPR) in response to endoplasmic reticulum (ER) stress (5), p38 MAPK, cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS; ref. 6), NF-κB (7), and alarmins (8).

Hypoxia in solid tumors is widely involved in solid tumor biology including angiogenesis, metastasis, and tumor resistance. The overexpression of HIFα subunits in solid tumors is associated with aggressive cancer cell behavior and is correlated with poor overall survival of patients (9). In this review, we focused on recent insights into the hypoxic aspect of tumorigenesis and therapy in hematologic malignancies.

Physiologically, the bone marrow shows the average oxygen tension between 5% and 7% O2 (10, 11). Because of the bone marrow tissue architecture, there is a heterogenous O2 level implied by the fact that the bone marrow consists of at least two different niches, the endosteal niche, the most hypoxic region of the bone marrow and closer to the bone, and the well-oxygenated vascular niche closer to the blood vessels (12). High levels of HIF1α were demonstrated at the border of endosteum, whereas lower levels of HIF1α were shown in the vascular niche (13). Interestingly, the hematopoietic stem cells (HSC) are localized mainly in the bone marrow with the endosteal niche, suggesting fundamental role of hypoxia in stem-cell function maintenance (14), such as homeostasis, low mitochondrial activity, and increased glycolysis (15).

Development of hypoxia due to tumor progression

The development of hypoxic conditions during the progression of solid tumors was very well documented (16–19), and was shown to be mediated through a complex network of signaling including HIF-dependent and -independent pathways (3, 5). Severe hypoxic conditions (beyond the physiologic bone marrow conditions) were shown to develop during the progression of hematologic malignancies including multiple myeloma, lymphoma, and leukemia.

Similar to solid tumors, bone marrow samples from patients with multiple myeloma were reported hypoxic by using endogenous hypoxic markers, HIF1α and HIF2α (11, 20–23). In animal models, the expression of exogenous (pimonidazole) and endogenous (HIF1α) hypoxia markers was significantly increased in the bone marrow of mice with high involvement of myeloma compared with the bone marrow of naïve mice or in areas with low tumor involvement (9, 18, 24). In addition, a direct correlation was found between the tumor burden and the levels of hypoxia in the bone marrow of mice with multiple myeloma (18). In addition to the bone marrow, circulating multiple myeloma cells were shown to have hypoxic phenotype (18). In multiple myeloma cell lines, including KMM1, U266, RPMI8226, JJN3, U266, H929, and MM1s, HIF1α was constitutively expressed and was further increased when the cells were exposed to hypoxia (9, 18, 21, 23). In addition to hypoxia in the multiple myeloma cells, the multiple myeloma bone marrow microenvironment also showed hypoxic response, in which strong HIF1α and HIF2α expression was detected in CD68+ macrophages (20, 22) and in stromal cells in areas with multiple myeloma progression (18).

HIF1α and HIF2α were detected in 54% of patients with Hodgkin lymphoma, and in 71% of patients with non-Hodgkin lymphoma; and the expression of HIFα and HIF2α was found to play a role in tumor progression (25). In diffuse large B-cell lymphoma (DLBCL), follicular lymphoma, and mantle cell lymphoma (MCL), HIF1α and HIF2α proteins were highly expressed in 70%, 73%, and 73.5% of the patients, respectively (26, 27).

High expression of HIF1α was detected in 67% of patients with acute myelogenous leukemia (AML; ref. 28), 66.7% of patients with pediatric acute lymphoblastic leukemia (ALL; ref. 29) with hypoxic areas widely present in ALL leukemic bone marrow (30), and in 100% (10 of 10) of patients with B-cell chronic lymphocytic leukemia (CLL; ref. 31). It was demonstrated that during leukemogenesis in rat xenograft model of AML, the bone marrow became more hypoxic, detected by nitroimidazol, in which hypoxia was found in 80% of AML cells in the bone marrow and 40% of circulating cells (32). Other reports showed that hypoxic regions also expanded in the bone marrow in the mice xenograft models of ALL, AML, and chronic myelogenous leukemia (CML; refs. 30, 33, 34). In CML model, hypoxia was shown to develop in the bone marrow in correlation with the tumor burden, while no hypoxia was found in circulating CML cells (34).

Hypoxia was shown to develop during the progression of most types of solid tumors; however, the development of hypoxia in hematologic malignancies was variable. In myeloma and lymphoma, which have a more solid nature in the bone marrow, hypoxia was shown to develop in the bone marrow of the patients in correlation with the tumor progression in the bone marrow, while in myeloma also circulating tumor cells had hypoxic phenotype. Leukemia involves both the bone marrow and circulation but the number of circulating cells is way much higher, and the dependency of leukemia cells on the bone marrow is less critical. Accordingly, hypoxia in the bone marrow was developed in different types of leukemia but less evidence was found for hypoxic phenotype in the circulating leukemic cells. We suggest that hypoxia plays a major role in the development of all three types of hematologic malignancies, but to a lower extent in leukemia.

The role of hypoxia in angiogenesis

In solid tumors, tumor neovasculogenesis plays a crucial role in the growth, progression, and metastasis of cancer (16). New blood vessels provide oxygen and nutrients, entrance for the immune cells, and exit of tumor cells into the circulation. Uncontrolled cancer cell proliferation causes hypoxia, which induces proliferation and sprouting of endothelial cells from preexisting vessels, supports vessel maturation (1), and contributes to the recruitment of endothelial progenitor cells from the bone marrow to tumor areas and induction of their differentiation into endothelial cells (35).

An increase in vascular density in the bone marrow, regulated at least, in part, by the HIF pathway, was demonstrated in patients during the monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma progression. A positive correlation was demonstrated between hypoxic markers, such as HIF1α and HIF2α expression, proangiogenic factors (VEGF) and its receptors, circulating chemokine SDF-1, and the level of bone marrow angiogenesis in patients with multiple myeloma (20, 22). Culturing multiple myeloma cell lines and CD138+ cells from myeloma patients in hypoxia increased CXCL2, VEGF, TGFβ, and IL8 secretion, induced expression of HIF1α or HIF2α, and augmented angiogenesis in vivo (11, 22). Subcutaneous injection of CD138+ multiple myeloma cells with overexpressed HIF1α and HIF2α into the nude mice induced a number of proangiogenic molecules, which resulted in multiple myeloma–induced angiogenesis in vivo (22). On the other hand, silencing HIF1α in multiple myeloma cells reduced new vessels formation both in vivo (23) and in vitro (11).

Angiogenesis was shown to be involved in the pathogenesis and progression of non-Hodgkin lymphomas; matched lymph node biopsies at diagnosis and recurrence of relapsed/refractory indolent non-Hodgkin lymphomas patients showed increased vascular network and HIF1α protein expression in the second biopsy, providing direct evidence that hypoxia-induced angiogenesis is an essential process for disease progression (37). In anaplastic large-cell lymphomas, secretion of VEGF and angiogenesis was shown to be mediated through hypoxia-induced downregulation of microRNA-16 (miR16) (38).

In leukemia, it was demonstrated that hypoxia contributes to angiogenesis through increased exosomes production by K562 leukemia cell line. This study has shown that exosomes derived from hypoxic leukemia cells increased tube formation when cultured with endothelial cells, partially via augmented exosomal miR210 expression (39). In addition, silencing of HIF1α in leukemic bone marrow stromal cells (BMSC) from AML and ALL decreased production of proangiogenic factor (such as VEGF), and decreased angiogenesis (40).

These results suggest that, similar to solid tumors, angiogenesis in hematologic malignancies is induced by tumor hypoxia and targeting one of the hypoxic pathways, namely HIF, shows promising results in controlling formation of the blood vessels. Hypoxia-induced angiogenesis in hematologic malignancies was shown to be mediated mainly through local secretion of angiogenic factors and sprouting of local vessels; unlike solid tumors, there is no evidence that hypoxia affects cell trafficking of endothelial progenitors in hematologic malignancies. The effect of tumor hypoxia on endothelial progenitors and their contribution to angiogenesis in hematologic malignancies is yet to be explored. We suggest that the inhibition of hypoxic response can provide an alternative approach to inhibit angiogenesis in which it will cause downregulation of multiple angiogenic factors at once, rather than inhibition of a specific factor.

The role of hypoxia in metastasis and spread

Hypoxia was shown to induce invasive behavior of epithelial cancer cells via epithelial-to-mesenchymal transition (EMT) in which cells acquire plastic and migratory abilities (17, 18). Tumor hypoxia was shown to activate EMT transcription factors (41, 42) and molecules responsible for polarization, adhesion, invasion of cells, such as vimentin, fibronectin, CXCR4, E-cadherin, MMP-2, and MMP-9 (17, 18).

Multiple myeloma is present at multiple sites of the bone marrow and the tumor cells spread throughout by continuous trafficking in and out of the bone marrow (18, 43–45). Steps of cell dissemination include: (i) cell detachment and acquisition of EMT-like phenotype (ii) invasion, (iii) egress to the bloodstream, (iv) circulation in the blood, (v) homing to the predetermined tissues, (vi) formation of a new tumor foci, and finally (vii) growth of micrometastasis (colonization; ref. 46). The selective choice is believed to be triggered by multiple factors such as chemoattractants (SDF-1 and its ligand, CXCR4), growth factors (IL6, TGFβ, IGFI, IGFII, FGF, PDGF, and VEGF), and calcium (46). Although not of an epithelial origin, Azab and colleagues (18) showed that increasing bone marrow tumor volume caused tumor hypoxia that induced multiple myeloma cell dissemination via hypoxia-induced EMT-like phenotype (increased expression of Snail1, FOXC2, and TGFβ). It was observed that, with increasing hypoxia, multiple myeloma cells seemed to be less adherent to bone marrow microenvironment (through downregulation of E-cadherin and downregulation of secretion of SDF1 from the stroma), and consequently, a higher number of circulating multiple myeloma cells in the peripheral blood was observed in mouse xenograft model (18). At the same time, hypoxic multiple myeloma cells (in the bone marrow and in the circulation) expressed high CXCR4 through increased expression of HIF1α and facilitated migration and homing of multiple myeloma cells to new bone marrow niches (18).

In lymphoma, HIF1α and HIF1 signaling were increased during liver metastasis of T-lymphoma cells in vivo, and HIF1α knockdown significantly impaired Met expression and phosphorylation and inhibited scattered liver metastasis (47).

In leukemia, Azab and colleagues (34) demonstrated that hypoxia regulated trafficking of CML cells. Hypoxia decreased the expression of E-cadherin in CML cells in the bone marrow and in vitro, which resulted in decreased adhesion to bone marrow stroma. In addition, hypoxia increased CXCR4 expression in vivo and in vitro and enhanced chemotaxis. These results suggest that hypoxia in the bone marrow induced an EMT-like phenotype in CML cells (34). It was also reported that in AML, hypoxia increased CXCR4 expression, thus augmented migration and survival of leukemic cells (48). Moreover, silencing HIF1α applied in BMSCs, resulted in VEGF and SDF-1 reduction, and as a consequence, decreased leukemia cell migration and adhesion (40). No changes were observed in the expression of CXCR4 and E-cadherin in circulating CML cells as a result of hypoxia in the bone marrow (34).

To sum up, these results show that metastatic spread of multiple myeloma is very similar to the metastatic spread of solid tumors, which is based on the fact that myeloma cells both in the bone marrow and in the circulation show hypoxic phenotype that facilitates metastasis through the EMT-like phenotype. Inhibition of the hypoxic response as a therapeutic strategy to prevent metastasis in multiple myeloma is yet to be explored. Although the literature in lymphoma is relatively limited, it seems that hypoxia plays an important role in the spread of lymphoma; more research to explore this role and its therapeutic applications is warranted. In leukemia, hypoxia seems to play a critical role of the progression of the disease in the bone marrow and it displays EMT-like phenotype, but not when the cells are circulating (in CML). However, the role of hypoxia in other leukemia types is still not clear.

The role of hypoxia in stemness and drug resistance

Hypoxia confers treatment resistance in solid tumors by regulating processes such as (i) cell-cycle arrest (quiescence), a state of reduced cell proliferation that protects the cells from external stress (49–51), (ii) inhibiting apoptosis and senescence of cells, (iii) controlling autophagy, ER-stress, and p53 and mitochondrial activity (50, 52). Hypoxia induces de-differentiated and immature phenotype of cells, as shown in neuroblastoma and breast cancer cells (53), as well as maintaining stem cell–like phenotype in both HSCs and brain stem cells (14, 54, 55). This was confirmed by activation of stemness-related genes and pathways, such as Oct4, HIF, Notch (56, 57), Wnt, and Hedgehog (58, 59). Hypoxic multiple myeloma cells were shown to undergo de-differentiation, which was demonstrated by decreased expression of plasma cell–specific markers (including CD138 and CS1) and transcription factors (IRF4, PRDM1, and XBP1), increased expression of B-cell–specific CD markers (CD20) and transcription markers (BCL6 and PAX5), and increased stem cell–like transcriptional program including Oct-4, SOX2, and NANOG (60). Drug resistance is one of the main features of stem cell–like phenotype that are associated with hypoxia in solid tumors. It was demonstrated that by inhibiting HIF1α in multiple myeloma cells using echinomycin or siRNA against HIF1α, myeloma cells were sensitized to melphalan, which happened through reduction of one of the most important antiapoptotic proteins, survivin (21).

Inhibition of HIF1α using echinomycin in mouse lymphoma stem cell population (characterized as c-Kit+Sca-1+) significantly reduced the colony formation of the lymphoma stem cells, indicating clear involvement of hypoxia in lymphoma tumor initiation (61). Interestingly, VHL protein (involved in HIFα degradation) transcript was undetectable in c-Kit+Sca-1+ lymphoma cells, which suggests the importance of HIFα in maintaining stem cell–like phenotype in lymphoma (61). In addition, HIFα protein stabilization was correlated with high expression of antiapoptotic protein, Bcl-(xL), indicating that HIFα is involved in apoptosis resistance of lymphoma cells conferring drug resistance in non-Hodgkin lymphoma (62).

In leukemia, using a mouse model of CML, it was demonstrated that HIF1α is a key player in maintaining the survival of leukemia stem cells that present the phenotype of LinSca-1+c-Kit+. HIF1α deletion hindered propagation of CML by inhibiting cell-cycle progression and inducing apoptosis. Interestingly, when compared with normal HSCs, leukemia stem cells seem to be more HIF1α-dependent (63). HIF2α was also demonstrated to be strongly involved in the long-term repopulating ability of human CD34+ HSCs as well as in survival and engraftment ability of primary human AML cells (64). Inducible HIF1α in AML cells changed the expression of 19 miRNAs, among which miR17 and miR20a were downregulated by repressing c-Myc expression. These two miRNAs alleviated hypoxia and HIF-induced AML cell differentiation. Studies in vivo revealed that miR20a contributes to HIF1α-inducible differentiation of AML cells (65). Overexpression of HIF1α in hypoxic AML cell lines conferred chemoresistance to antimycin due to quiescence induced by cell-cycle arrest (66). Coculture of ALL cells with stromal cells in hypoxia induced HIF1α and AKT/mTOR signaling; inhibition of mTOR decreased HIF1α and led to chemosensitization of ALL cells (33).

Altogether, in solid tumors the contribution of hypoxia to the drug-resistant stem-cell-like phenotype is well established. However, the results which suggest that hypoxia plays an important role in development of cancer stem cells (CSC) and drug resistance in hematologic malignancies are still preliminary and need further confirmation to provide direct evidence for the involvement of hypoxia in these processes. We suggest to further investigate the role of hypoxia in the development of drug-resistant stem cell-like phenotype; in hematologic malignancy this will have a huge impact on our understanding of the development of resistance to therapy, development of minimal residual disease, and recurrence in hematologic malignancies. Exploring hypoxia will open new therapeutic windows for enhanced treatments in the relapsed/refractory stages of these diseases. Special emphasis should be given to tumors involving stem cells and blasts.

Other effects induced by hypoxia

It was demonstrated that HIFα plays an important role in bone biology and bone disease, because the bone is physiologically a hypoxic tissue (67). Lytic lesions affect more than 80% of patients with multiple myeloma and are caused by imbalance between proliferation and activity of overstimulated osteoclasts and suppressed osteoblasts. Receptor activator of NF-κB ligand (RANKL) produced by multiple myeloma cells and activated osteoclasts was shown to be increased in patients with multiple myeloma (68). Recent reports suggest that osteoclast activity contributing to the osteolytic bone disease in multiple myeloma is hypoxia-regulated, because the increase in number and size of osteoclasts was, in part, regulated by HIF1α (23). Stable inhibition of HIF1α in multiple myeloma cells halted proosteoclastogenic cytokines secretion including IL7 and CCL3/MIP-1α, reduced the weight and tumor size in vivo, and significantly decreased bone destruction (23). Osteolytic lesions were reduced together with tumor burden by HIF inhibition that can be explained by the fact that active osteoclasts support multiple myeloma cell proliferation in vivo.

Targeting hypoxia is an emerging strategy for the treatment of hematologic malignancies, in which hypoxia-induced cellular adaptation mechanisms and hypoxic cells are being used as therapeutic targets for prevention of angiogenesis, metastasis, stemness, and drug resistance.

Targeting hypoxia is an emerging approach to target tumor tissue and microenvironment, mainly due to the fact that hypoxia induces resistance to anticancer therapies. Some of the mechanisms of therapy-resistance include (i) increased expression of drug efflux pumps (such as P-glycoprotein, Pgp), (ii) oxygen-dependent cytotoxicity of some drugs (such as bleomycin), (iii) mutations conferring resistance, (iv) selection of cells insensitive to p53-mediated apoptosis, (v) decreased cell proliferation rate in hypoxic regions, and (vi) drug diffusion limits due to the distance from the blood vessels and thus inadequate exposure of cells to anticancer drugs (see Fig. 1).

Figure 1.

Mechanisms of resistance to anticancer drugs developed by hypoxic cells. Hypoxia-associated mechanisms conferring therapy-resistance include (i) increased expression of drug efflux pumps (such as Pgp), (ii) oxygen-dependent cytotoxicity of drugs (such as bleomycin), (iii) mutations providing apoptosis resistance, (iv) selection of cells insensitive to p53-mediated apoptosis, (v) decreased cell proliferation rate in hypoxic regions, and (vi) inadequate exposure of cells to anticancer drugs due to diffusion limit.

Figure 1.

Mechanisms of resistance to anticancer drugs developed by hypoxic cells. Hypoxia-associated mechanisms conferring therapy-resistance include (i) increased expression of drug efflux pumps (such as Pgp), (ii) oxygen-dependent cytotoxicity of drugs (such as bleomycin), (iii) mutations providing apoptosis resistance, (iv) selection of cells insensitive to p53-mediated apoptosis, (v) decreased cell proliferation rate in hypoxic regions, and (vi) inadequate exposure of cells to anticancer drugs due to diffusion limit.

Close modal

Targeting hypoxic cells with prodrugs

Small-molecule prodrugs discovery in the 1970s was a result of an attempt to overcome the problem with hypoxia-induced radiation resistance. It led to the finding of nitroimidazoles that can mimic the effect of oxygen and enhance the effect of radiation. Recently, a paradigm has shifted and hypoxia-activated prodrugs are currently used as compounds that target hypoxia and HIF-mediated signaling pathways. This strategy is intended to penetrate the tissue and deliver cytotoxic agents to the hypoxic nutrient-starved regions of tumor areas with minimal toxicity for the surrounding normoxic cells (69). In principle, non-toxic prodrug requires one- or two-electron reduction to a far more toxic radical, or further to a downstream product.

In solid tumors, some of the first prodrugs such as metronidazole, misonidasole, etanidazole, and tirapazamine were shown to have a very high toxicity. Combination of these drugs with other chemotherapies enhanced the effectiveness of the treatment in vitro and in vivo (70–73). The combination of tirapazamine with cisplatin shows prolonged survival in phase III clinical trial; however, dosing is limited because of neutropenia and other toxicities (74). No literature was found describing the use of these compounds in hematologic malignancies.

Two new prodrugs, 2-nitroimidazole of the phosphoramide mustard, namely TH-302 and PR-104, also show potential in the clinical studies due to better dose potency compared with tirapazamine. TH-302 was shown to be cytotoxic against a number of human hypoxic cancer cell lines in vitro and in vivo (75–79). TH-302 is being clinically tested in solid tumors as a therapy in combination with chemotherapy (69, 75), and has completed phase I clinical trial. TH-302 and PR-104 have been tested in myeloma and leukemia and their activity was demonstrated both in vivo and in vitro. Preclinical studies on myeloma model (5T33MMvv) showed that TH-302 induced multiple myeloma cell apoptosis and decreased paraprotein secretion in vivo, and combination of TH-302 and bortezomib significantly improved survival of mice (24, 75). TH-302 also effectively inhibited onset, progression of AML, and extended the overall survival, both in systemic AML xenograft models and mice with advanced AML disease. In vitro, TH-302 decreased HIF1α protein expression and reduced ROS production, which resulted in decreased proliferation, increased cell-cycle arrest, and induced double-stranded DNA breaks (80). TH-302 is currently evaluated in phase I/II study in patients with relapsed/refractory multiple myeloma combination with dexamethasone, with or without bortezomib (NCT01522872). Dose-limiting toxicity with most common adverse events, such as leukopenia, thrombocytopenia, anemia, and stomatitis, was described. TH-302 in combination with dexamethasone scheduled twice weekly showed initial clinical activity with benefit rate in 33% patients who were relapsed/refractory to both bortezomib and thalidomide/lenalidomide regimens.

PR-104 is being evaluated in treating patients with refractory/relapsed AML in phase I/II study (NCT01037556), because leukemic bone marrow was reported to be sufficiently hypoxic to activate PR-104 to its active metabolite PR-104H and PR-104M. The final report demonstrated that PR-104 had moderate toxicity such as myelosuppression and enterocolitis (81).

The hypoxic prodrugs are showing promising results in preclinical and clinical studies in hematologic malignancies as well as solid tumors; however, their toxicity and pharmacokinetics appear to be a limiting factor in these treatments. We suggest further studies for the development of a new generation of hypoxic prodrugs with greater hypoxic selectivity, lower toxicity, and better pharmacokinetics, as well as more efficient vector for enhanced delivery of these drugs to hypoxic regions.

Targeting HIF pathway

HIFα-targeted therapy is another promising approach to overcome hypoxia-induced progression, metastasis, and drug resistance. Although hypoxia does not equate HIFα expression, because the HIFα pathway is also regulated in normoxic conditions, HIFα is strongly intertwined with almost every aspect of cancer biology due to its powerful potential of regulating gene transcription. Recently, a high-throughput screening of FDA-approved drugs disclosed digoxin, acriflavine, anthracyclines (doxorubicin), vincristine, paclitaxel, and many more as HIFα inhibitors, which suggest that currently used drugs are already inhibiting HIFα. However, based on their primary therapeutic functions, these inhibitors have also HIFα-independent effects, thus their usage is not specific (82).

Silencing HIF1α with shRNA successfully reduced multiple myeloma–induced new vessel formation and tumor burden (11, 23). In addition, inhibition of HIFα was proposed to target therapy-resistant stem cell–like phenotype in hematologic CSCs (83). Promising study in vivo demonstrated that HIF1α inhibition using echinomycin in lymphoma (61) and leukemia (63) stem cells, efficiently decreased tumor-initiating activity of these cells and improved survival of mice. It was also shown that blocking HIFα enhances the antiproliferative effect of lenalidomide (23) and melphalan (21).

Further studies are warranted to investigate the therapeutic applications of inhibition of HIFα in prevention of tumor progression, metastasis, and drug resistance in hematologic malignancies. Furthermore, the development of more specific small-molecule HIFα inhibitors is urgently needed in the field to test these therapeutic applications.

Tumor-hypoxia was shown to develop in correlation with the progression of hematologic malignancies and is associated with aggressive tumor behavior. Exploiting the distinctive features of hypoxia and hypoxia-regulated molecules in hematologic malignancies is a promising strategy to improve the treatment of hematologic malignancies; however, its full potential is yet to be determined. We hypothesize that targeting hypoxia in hematologic malignancies, especially in multiple myeloma and lymphoma, will improve the treatment in these diseases through inhibition of angiogenesis, metastasis, and drug resistance.

A.K. Azab reports receiving research funding from Selexys Pharmaceuticals, Verastem Inc., Karyopharm Therapeutics Inc., and Cellworks. No potential conflicts of interest were disclosed by other authors.

1.
Krock
BL
,
Skuli
N
,
Simon
MC
. 
Hypoxia-induced angiogenesis: good and evil
.
Genes Cancer
2011
;
2
:
1117
33
.
2.
Bertout
JA
,
Patel
SA
,
Simon
MC
. 
The impact of O2 availability on human cancer
.
Nat Rev Cancer
2008
;
8
:
967
75
.
3.
Semenza
GL
,
Wang
GL
. 
A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation
.
Mol Cell Biol
1992
;
12
:
5447
54
.
4.
Bruick
RK
,
McKnight
SL
. 
A conserved family of prolyl-4-hydroxylases that modify HIF
.
Science
2001
;
294
:
1337
40
.
5.
Wouters
BG
,
Koritzinsky
M
. 
Hypoxia signalling through mTOR and the unfolded protein response in cancer
.
Nat Rev Cancer
2008
;
8
:
851
64
.
6.
Sanchez
A
,
Tripathy
D
,
Yin
X
,
Desobry
K
,
Martinez
J
,
Riley
J
, et al
p38 MAPK: a mediator of hypoxia-induced cerebrovascular inflammation
.
J Alzheimers Dis
2012
;
32
:
587
97
.
7.
Cummins
EP
,
Berra
E
,
Comerford
KM
,
Ginouves
A
,
Fitzgerald
KT
,
Seeballuck
F
, et al
Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity
.
Proc Natl Acad Sci U S A
2006
;
103
:
18154
9
.
8.
Tafani
M
,
Schito
L
,
Pellegrini
L
,
Villanova
L
,
Marfe
G
,
Anwar
T
, et al
Hypoxia-increased RAGE and P2 × 7R expression regulates tumor cell invasion through phosphorylation of Erk1/2 and Akt and nuclear translocation of NF-{kappa}B
.
Carcinogenesis
2011
;
32
:
1167
75
.
9.
Semenza
GL
. 
Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy
.
Trends Pharmacol Sci
2012
;
33
:
207
14
.
10.
Grant
JL
,
Smith
B
. 
Bone marrow gas tensions, bone marrow blood flow, and erythropoiesis in man
.
Ann Intern Med
1963
;
58
:
801
9
.
11.
Colla
S
,
Storti
P
,
Donofrio
G
,
Todoerti
K
,
Bolzoni
M
,
Lazzaretti
M
, et al
Low bone marrow oxygen tension and hypoxia-inducible factor-1alpha overexpression characterize patients with multiple myeloma: role on the transcriptional and proangiogenic profiles of CD138(+) cells
.
Leukemia
2010
;
24
:
1967
70
.
12.
Sipkins
DA
,
Wei
X
,
Wu
JW
,
Runnels
JM
,
Côté
D
,
Means
TK
, et al
In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment
.
Nature
2005
;
435
:
969
73
.
13.
Levesque
JP
,
Winkler
IG
,
Hendy
J
,
Williams
B
,
Helwani
F
,
Barbier
V
, et al
Hematopoietic progenitor cell mobilization results in hypoxia with increased hypoxia-inducible transcription factor-1 alpha and vascular endothelial growth factor A in bone marrow
.
Stem Cells
2007
;
25
:
1954
65
.
14.
Parmar
K
,
Mauch
P
,
Vergilio
JA
,
Sackstein
R
,
Down
JD
. 
Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia
.
Proc Natl Acad Sci U S A
2007
;
104
:
5431
6
.
15.
Simsek
T
,
Kocabas
F
,
Zheng
J
,
Deberardinis
RJ
,
Mahmoud
AI
,
Olson
EN
, et al
The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche
.
Cell Stem Cell
2010
;
7
:
380
90
.
16.
Folkman
J
. 
Tumor angiogenesis: therapeutic implications
.
N Engl J Med
1971
;
285
:
1182
6
.
17.
Lu
X
,
Kang
Y
. 
Hypoxia and hypoxia-inducible factors: master regulators of metastasis
.
Clin Cancer Res
2010
;
16
:
5928
35
.
18.
Azab
AK
,
Hu
J
,
Quang
P
,
Azab
F
,
Pitsillides
C
,
Awwad
R
, et al
Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features
.
Blood
2012
;
119
:
5782
94
.
19.
Tannock
I
. 
Cell kinetics and chemotherapy: a critical review
.
Cancer Treat Rep
1978
;
62
:
1117
33
.
20.
Giatromanolaki
A
,
Bai
M
,
Margaritis
D
,
Bourantas
KL
,
Koukourakis
MI
,
Sivridis
E
, et al
Hypoxia and activated VEGF/receptor pathway in multiple myeloma
.
Anticancer Res
2010
;
30
:
2831
6
.
21.
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
.
22.
Martin
SK
,
Diamond
P
,
Williams
SA
,
To
LB
,
Peet
DJ
,
Fujii
N
, et al
Hypoxia-inducible factor-2 is a novel regulator of aberrant CXCL12 expression in multiple myeloma plasma cells
.
Haematologica
2010
;
95
:
776
84
.
23.
Storti
P
,
Bolzoni
M
,
Donofrio
G
,
Airoldi
I
,
Guasco
D
,
Toscani
D
, et al
Hypoxia-inducible factor (HIF)-1alpha suppression in myeloma cells blocks tumoral growth in vivo inhibiting angiogenesis and bone destruction
.
Leukemia
2013
;
27
:
1697
706
.
24.
Hu
J
,
Handisides
DR
,
Van Valckenborgh
E
,
De Raeve
H
,
Menu
E
,
Vande Broek
I
, et al
Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrug
.
Blood
2010
;
116
:
1524
7
.
25.
Passam
FH
,
Alexandrakis
MG
,
Kafousi
M
,
Fotinou
M
,
Darivianaki
K
,
Tsirakis
G
, et al
Histological expression of angiogenic factors: VEGF, PDGFRalpha, and HIF-1alpha in Hodgkin lymphoma
.
Pathol Res Pract
2009
;
205
:
11
20
.
26.
Evens
AM
,
Schumacker
PT
,
Helenowski
IB
,
Singh
AT
,
Dokic
D
,
Keswani
A
, et al
Hypoxia inducible factor-alpha activation in lymphoma and relationship to the thioredoxin family
.
Br J Haematol
2008
;
141
:
676
80
.
27.
Argyriou
P
,
Papageorgiou
SG
,
Panteleon
V
,
Psyrri
A
,
Bakou
V
,
Pappa
V
, et al
Hypoxia-inducible factors in mantle cell lymphoma: implication for an activated mTORC1–>HIF-1alpha pathway
.
Ann Hematol
2011
;
90
:
315
22
.
28.
Deeb
G
,
Vaughan
MM
,
McInnis
I
,
Ford
LA
,
Sait
SN
,
Starostik
P
, et al
Hypoxia-inducible factor-1alpha protein expression is associated with poor survival in normal karyotype adult acute myeloid leukemia
.
Leuk Res
2011
;
35
:
579
84
.
29.
Wellmann
S
,
Guschmann
M
,
Griethe
W
,
Eckert
C
,
von Stackelberg
A
,
Lottaz
C
, et al
Activation of the HIF pathway in childhood ALL, prognostic implications of VEGF
.
Leukemia
2004
;
18
:
926
33
.
30.
Benito
J
,
Shi
Y
,
Szymanska
B
,
Carol
H
,
Boehm
I
,
Lu
H
, et al
Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104
.
PLoS ONE
2011
;
6
:
e23108
.
31.
Ghosh
AK
,
Shanafelt
TD
,
Cimmino
A
,
Taccioli
C
,
Volinia
S
,
Liu
CG
, et al
Aberrant regulation of pVHL levels by microRNA promotes the HIF/VEGF axis in CLL B cells
.
Blood
2009
;
113
:
5568
74
.
32.
Jensen
PO
,
Mortensen
BT
,
Hodgkiss
RJ
,
Iversen
PO
,
Christensen
IJ
,
Helledie
N
, et al
Increased cellular hypoxia and reduced proliferation of both normal and leukaemic cells during progression of acute myeloid leukaemia in rats
.
Cell Prolif
2000
;
33
:
381
95
.
33.
Frolova
O
,
Samudio
I
,
Benito
JM
,
Jacamo
R
,
Kornblau
SM
,
Markovic
A
, et al
Regulation of HIF-1alpha signaling and chemoresistance in acute lymphocytic leukemia under hypoxic conditions of the bone marrow microenvironment
.
Cancer Biol Ther
2012
;
13
:
858
70
.
34.
Azab
AK
,
Weisberg
E
,
Sahin
I
,
Liu
F
,
Awwad
R
,
Azab
F
, et al
The influence of hypoxia on CML trafficking through modulation of CXCR4 and E-cadherin expression
.
Leukemia
2013
;
27
:
961
4
.
35.
de la Puente
P
,
Muz
B
,
Azab
F
,
Azab
AK
. 
Cell trafficking of endothelial progenitor cells in tumor progression
.
Clin Cancer Res
2013
;
19
:
3360
8
.
36.
Rajkumar
SV
,
Mesa
RA
,
Fonseca
R
,
Schroeder
G
,
Plevak
MF
,
Dispenzieri
A
, et al
Bone marrow angiogenesis in 400 patients with monoclonal gammopathy of undetermined significance, multiple myeloma, and primary amyloidosis
.
Clin Cancer Res
2002
;
8
:
2210
6
.
37.
Minoia
C
,
Quero
C
,
Asselti
M
,
Galise
I
,
Marzano
AL
,
Iacobazzi
A
, et al
Changes in angiogenesis and hypoxia-inducible factor-1alpha protein expression in relapsed/refractory indolent non-Hodgkin lymphomas
.
Br J Haematol
2013
;
163
:
640
5
.
38.
Dejean
E
,
Renalier
MH
,
Foisseau
M
,
Agirre
X
,
Joseph
N
,
de Paiva
GR
, et al
Hypoxia-microRNA-16 downregulation induces VEGF expression in anaplastic lymphoma kinase (ALK)-positive anaplastic large-cell lymphomas
.
Leukemia
2011
;
25
:
1882
90
.
39.
Tadokoro
H
,
Umezu
T
,
Ohyashiki
K
,
Hirano
T
,
Ohyashiki
JH
. 
Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells
.
J Biol Chem
2013
;
288
:
34343
51
.
40.
Dong-Feng
Z
,
Ting
L
,
Cheng
C
,
Xi
Z
,
Xue
L
,
Xing-Hua
C
, et al
Silencing HIF-1alpha reduces the adhesion and secretion functions of acute leukemia hBMSCs
.
Braz J Med Biol Res
2012
;
45
:
906
12
.
41.
Yang
MH
,
Wu
KJ
. 
TWIST activation by hypoxia inducible factor-1 (HIF-1): implications in metastasis and development
.
Cell Cycle
2008
;
7
:
2090
6
.
42.
Mak
P
,
Leav
I
,
Pursell
B
,
Bae
D
,
Yang
X
,
Taglienti
CA
, et al
ERbeta impedes prostate cancer EMT by destabilizing HIF-1alpha and inhibiting VEGF-mediated snail nuclear localization: implications for Gleason grading
.
Cancer Cell
2010
;
17
:
319
32
.
43.
Azab
AK
,
Azab
F
,
Blotta
S
,
Pitsillides
CM
,
Thompson
B
,
Runnels
JM
, et al
RhoA and Rac1 GTPases play major and differential roles in stromal cell-derived factor-1-induced cell adhesion and chemotaxis in multiple myeloma
.
Blood
2009
;
114
:
619
29
.
44.
Azab
AK
,
Quang
P
,
Azab
F
,
Pitsillides
C
,
Thompson
B
,
Chonghaile
T
, et al
P-selectin glycoprotein ligand regulates the interaction of multiple myeloma cells with the bone marrow microenvironment
.
Blood
2012
;
119
:
1468
78
.
45.
Azab
AK
,
Runnels
JM
,
Pitsillides
C
,
Moreau
AS
,
Azab
F
,
Leleu
X
, et al
CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy
.
Blood
2009
;
113
:
4341
4351
.
46.
Ghobrial
IM
. 
Myeloma as a model for the process of metastasis: implications for therapy
.
Blood
2012
;
120
:
20
30
.
47.
Schelter
F
,
Halbgewachs
B
,
Bäumler
P
,
Neu
C
,
Görlach
A
,
Schrötzlmair
F
, et al
Tissue inhibitor of metalloproteinases-1-induced scattered liver metastasis is mediated by hypoxia-inducible factor-1alpha
.
Clin Exp Metastasis
2011
;
28
:
91
9
.
48.
Fiegl
M
,
Samudio
I
,
Clise-Dwyer
K
,
Burks
JK
,
Mnjoyan
Z
,
Andreeff
M
. 
CXCR4 expression and biologic activity in acute myeloid leukemia are dependent on oxygen partial pressure
.
Blood
2009
;
113
:
1504
12
.
49.
Vaupel
P
,
Kelleher
DK
,
Hockel
M
. 
Oxygen status of malignant tumors: pathogenesis of hypoxia and significance for tumor therapy
.
Semin Oncol
2001
;
28
(
2 Suppl 8
):
29
35
.
50.
Das
B
,
Tsuchida
R
,
Malkin
D
,
Koren
G
,
Baruchel
S
,
Yeger
H
. 
Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction
.
Stem Cells
2008
;
26
:
1818
30
.
51.
Cipolleschi
MG
,
Dello Sbarba
P
,
Olivotto
M
. 
The role of hypoxia in the maintenance of hematopoietic stem cells
.
Blood
1993
;
82
:
2031
7
.
52.
Rohwer
N
,
Cramer
T
. 
Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways
.
Drug Resist Updat
2011
;
14
:
191
201
.
53.
Axelson
H
,
Fredlund
E
,
Ovenberger
M
,
Landberg
G
,
Påhlman
S
. 
Hypoxia-induced dedifferentiation of tumor cells—a mechanism behind heterogeneity and aggressiveness of solid tumors
.
Semin Cell Dev Biol
2005
;
16
:
554
63
.
54.
Diabira
S
,
Morandi
X
. 
Gliomagenesis and neural stem cells: key role of hypoxia and concept of tumor “neo-niche”
.
Med Hypotheses
2008
;
70
:
96
104
.
55.
Blazek
ER
,
Foutch
JL
,
Maki
G
. 
Daoy medulloblastoma cells that express CD133 are radioresistant relative to CD133 cells, and the CD133+ sector is enlarged by hypoxia
.
Int J Radiat Oncol Biol Phys
2007
;
67
:
1
5
.
56.
Gustafsson
MV
,
Zheng
X
,
Pereira
T
,
Gradin
K
,
Jin
S
,
Lundkvist
J
, et al
Hypoxia requires notch signaling to maintain the undifferentiated cell state
.
Dev Cell
2005
;
9
:
617
28
.
57.
Keith
B
,
Simon
MC
. 
Hypoxia-inducible factors, stem cells, and cancer
.
Cell
2007
;
129
:
465
72
.
58.
Tung
DC
,
Chao
KS
. 
Targeting hedgehog in cancer stem cells: how a paradigm shift can improve treatment response
.
Future Oncol
2007
;
3
:
569
74
.
59.
Katoh
M
. 
WNT signaling in stem cell biology and regenerative medicine
.
Curr Drug Targets
2008
;
9
:
565
70
.
60.
Kawano
Y
,
Kikukawa
Y
,
Fujiwara
S
,
Wada
N
,
Okuno
Y
,
Mitsuya
H
, et al
Hypoxia reduces CD138 expression and induces an immature and stem cell–like transcriptional program in myeloma cells
.
Int J Oncol
2013
;
43
:
1809
16
.
61.
Wang
Y
,
Liu
Y
,
Malek
SN
,
Zheng
P
,
Liu
Y
. 
Targeting HIF1alpha eliminates cancer stem cells in hematological malignancies
.
Cell Stem Cell
2011
;
8
:
399
411
.
62.
Hernandez-Luna
MA
,
Rocha-Zavaleta
L
,
Vega
MI
,
Huerta-Yepez
S
. 
Hypoxia inducible factor-1alpha induces chemoresistance phenotype in non-Hodgkin lymphoma cell line via up-regulation of Bcl-(xL)
.
Leuk Lymphoma
2013
;
54
:
1048
55
.
63.
Zhang
H
,
Li
H
,
Xi
HS
,
Li
S
. 
HIF1alpha is required for survival maintenance of chronic myeloid leukemia stem cells
.
Blood
2012
;
119
:
2595
607
.
64.
Rouault-Pierre
K
,
Lopez-Onieva
L
,
Foster
K
,
Anjos-Afonso
F
,
Lamrissi-Garcia
I
,
Serrano-Sanchez
M
, et al
HIF-2alpha protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress
.
Cell Stem Cell
2013
;
13
:
549
63
.
65.
He
M
,
Wang
QY
,
Yin
QQ
,
Tang
J
,
Lu
Y
,
Zhou
CX
, et al
HIF-1alpha downregulates miR-17/20a directly targeting p21 and STAT3: a role in myeloid leukemic cell differentiation
.
Cell Death Differ
2013
;
20
:
408
18
.
66.
Matsunaga
T
,
Imataki
O
,
Torii
E
,
Kameda
T
,
Shide
K
,
Shimoda
H
, et al
Elevated HIF-1alpha expression of acute myelogenous leukemia stem cells in the endosteal hypoxic zone may be a cause of minimal residual disease in bone marrow after chemotherapy
.
Leuk Res
2012
;
36
:
e122
4
.
67.
Rankin
EB
,
Giaccia
AJ
,
Schipani
E
. 
A central role for hypoxic signaling in cartilage, bone, and hematopoiesis
.
Curr Osteoporos Rep
2011
;
9
:
46
52
.
68.
Raje
N
,
Roodman
GD
. 
Advances in the biology and treatment of bone disease in multiple myeloma
.
Clin Cancer Res
2011
;
17
:
1278
86
.
69.
Duan
JX
,
Jiao
H
,
Kaizerman
J
,
Stanton
T
,
Evans
JW
,
Lan
L
, et al
Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs
.
J Med Chem
2008
;
51
:
2412
20
.
70.
Brown
JM
. 
Clinical trials of radiosensitizers: what should we expect?
Int J Radiat Oncol Biol Phys
1984
;
10
:
425
9
.
71.
Patterson
LH
. 
Bioreductively activated antitumor N-oxides: the case of AQ4N, a unique approach to hypoxia-activated cancer chemotherapy
.
Drug Metab Rev
2002
;
34
:
581
92
.
72.
Albertella
MR
,
Loadman
PM
,
Jones
PH
,
Phillips
RM
,
Rampling
R
,
Burnet
N
, et al
Hypoxia-selective targeting by the bioreductive prodrug AQ4N in patients with solid tumors: results of a phase I study
.
Clin Cancer Res
2008
;
14
:
1096
104
.
73.
Tredan
O
,
Garbens
AB
,
Lalani
AS
,
Tannock
IF
. 
The hypoxia-activated ProDrug AQ4N penetrates deeply in tumor tissues and complements the limited distribution of mitoxantrone
.
Cancer Res
2009
;
69
:
940
7
.
74.
von Pawel
J
,
von Roemeling
R
,
Gatzemeier
U
,
Boyer
M
,
Elisson
LO
,
Clark
P
, et al
Tirapazamine plus cisplatin versus cisplatin in advanced non-small-cell lung cancer: a report of the international CATAPULT I study group. Cisplatin and tirapazamine in subjects with advanced previously untreated non-small-cell lung tumors
.
J Clin Oncol
2000
;
18
:
1351
9
.
75.
Hu
J
,
Van Valckenborgh
E
,
Xu
D
,
Menu
E
,
De Raeve
H
,
De Bryune
E
, et al
Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro
.
Mol Cancer Ther
2013
;
12
:
1763
73
.
76.
Meng
F
,
Evans
JW
,
Bhupathi
D
,
Banica
M
,
Lan
L
,
Lorente
G
, et al
Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302
.
Mol Cancer Ther
2012
;
11
:
740
51
.
77.
Liu
Q
,
Sun
JD
,
Wang
J
,
Ahluwalia
D
,
Baker
AF
,
Cranmer
LD
, et al
TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules
.
Cancer Chemother Pharmacol
2012
;
69
:
1487
98
.
78.
Patterson
AV
,
Ferry
DM
,
Edmunds
SJ
,
Gu
Y
,
Singleton
RS
,
Patel
K
, et al
Mechanism of action and preclinical antitumor activity of the novel hypoxia-activated DNA cross-linking agent PR-104
.
Clin Cancer Res
2007
;
13
:
3922
32
.
79.
McKeage
MJ
,
Gu
Y
,
Wilson
WR
,
Hill
A
,
Amies
K
,
Melink
TJ
, et al
A phase I trial of PR-104, a pre-prodrug of the bioreductive prodrug PR-104A, given weekly to solid tumour patients
.
BMC Cancer
2011
;
11
:
432
.
80.
Portwood
S
,
Lal
D
,
Hsu
YC
,
Vargas
R
,
Johnson
MK
,
Wetzler
M
, et al
Activity of the hypoxia-activated prodrug, TH-302, in preclinical human acute myeloid leukemia (AML) models
.
Clin Cancer Res
2013
;
19
:
6506
19
.
81.
Yi
CYA
,
Borthakur
G
,
Thall
PF
. 
Final report of phase I/II study of PR104, a hypoxia-activated pro-drug, in relapsed/refractory acute leukemia 2013
J Clin Oncol
31
, 
2013
(
suppl; abstr 7074
).
82.
Zhang
H
,
Qian
DZ
,
Tan
YS
,
Lee
K
,
Gao
P
,
Ren
YR
, et al
Digoxin and other cardiac glycosides inhibit HIF-1alpha synthesis and block tumor growth
.
Proc Natl Acad Sci U S A
2008
;
105
:
19579
86
.
83.
Lee
KE
,
Simon
MC
. 
From stem cells to cancer stem cells: HIF takes the stage
.
Curr Opin Cell Biol
2012
;
24
:
232
5
.