You are on page 1of 10

Leukemia (2013) 27, 16971706

& 2013 Macmillan Publishers Limited All rights reserved 0887-6924/13


www.nature.com/leu

ORIGINAL ARTICLE
Hypoxia-inducible factor (HIF)-1a suppression in myeloma
cells blocks tumoral growth in vivo inhibiting angiogenesis
and bone destruction
P Storti1,11, M Bolzoni1,11, G Donofrio2, I Airoldi3, D Guasco1, D Toscani1, E Martella4, M Lazzaretti5, C Mancini4, L Agnelli6, K Patrene7,
S Maga8, V Franceschi2, S Colla9, J Anderson10, A Neri6, M Amiot8, F Aversa1, G David Roodman10 and N Giuliani1

Hypoxia-inducible transcription factor-1 (HIF-1a) is overexpressed in multiple myeloma (MM) cells within the hypoxic
microenvironment. Herein, we explored the effect of persistent HIF-1a inhibition by a lentivirus short hairpin RNA pool on MM cell
growth either in vitro or in vivo and on the transcriptional and pro-angiogenic proles of MM cells. HIF-1a suppression did not have
a signicant impact on MM cell proliferation and survival in vitro although, increased the antiproliferative effect of lenalidomide. On
the other hand, we found that HIF-1a inhibition in MM cells downregulates the pro-angiogenic genes VEGF, IL8, IL10, CCL2, CCL5
and MMP9. Pro-osteoclastogenic cytokines were also inhibited, such as IL-7 and CCL3/MIP-1a. The effect of HIF-1a inhibition was
assessed in vivo in nonobese diabetic/severe combined immunodeciency mice both in a subcutaneous and an intratibial MM
model. HIF-1a inhibition caused a dramatic reduction in the weight and volume of the tumor burden in both mouse models.
Moreover, a signicant reduction of the number of vessels and vascular endothelial growth factors (VEGFs) immunostaining was
observed. Finally, in the intratibial experiments, HIF-1a inhibition signicantly blocked bone destruction. Overall, our data indicate
that HIF-1a suppression in MM cells signicantly blocks MM-induced angiogenesis and reduces MM tumor burden and bone
destruction in vivo, supporting HIF-1a as a potential therapeutic target in MM.

Leukemia (2013) 27, 16971706; doi:10.1038/leu.2013.24


Keywords: myeloma; angiogenesis; bone disease; hypoxia; mice

INTRODUCTION hypoxia and is composed of two basic helix-loop-helix proteins,


Multiple myeloma (MM) is characterized by the accumulation of including the constitutive HIF-1b subunit and the hypoxia-
malignant plasma cells in the bone marrow (BM) microenviron- inducible a-subunit.1921 Under normoxic conditions HIF-1a has
ment that critically supports MM cell growth and survival.1 a very short half-life undergoing proteosomal degradation by
Particularly, an increase of BM angiogenesis and osteoclasto- oxygen-dependent hydroxylation. In contrast, under hypoxic
genesis occurs with MM cell growth and proliferation and plays a condition, hydroxylation is suppressed and HIF-1a protein
critical role in the pathophysiology and progression of MM.28 escapes proteasomal destruction and can accumulate and
The angiogenic switch in MM is mainly sustained by the translocate to the nucleus.1921 HIF-1a is a critical trigger and
overproduction of vascular endothelial growth factors (VEGFs) regulator of tumor associated angiogenesis.22,23 Interestingly
by MM cells and the BM microenvironment.2,9,10 Other pro- a critical role of HIF-1a in osteoclast formation and regulation
angiogenic molecules, such as basic broblast growth factor,11,12 has been shown24,25 as well as its role in the development of
interleukin-8 (IL-8),13 angiopoietin-1 14 and osteopontin15 are osteolytic metastasis in breast cancer model.26
involved in the pro-angiogenic process. Hypoxia is a common Recently it has been demonstrated that the BM microenvironment
feature of solid tumors and is associated with angiogenesis is hypoxic in MM patients27 and that HIF-1a is overexpressed by MM
and malignant phenotype. It is critical to the regulation of the cells, and modulates their transcriptional and pro-angiogenic
angiogenic switch in those cancers, by regulating the production proles.2729 Moreover, the role of hypoxia in tumor progression
of the pro-angiogenic molecules.1618 Tumor adaptation to and dissemination has been demonstrated in MM mouse
hypoxia is mainly due to the hypoxia-inducible factor (HIF)-1, a models.30,31 These ndings suggest that hypoxia could be a target
key transcription factor that regulates pro-angiogenic factors, in MM. The role of HIF-1a as a therapeutic target in MM is under
angiogenesis and tumor progression in solid tumors.1923 HIF-1 is investigation. In this study, we determined the effect of stable HIF-1a
a heterodimeric DNA binding complex highly inducible by inhibition in MM cells on cell proliferation and survival in vitro and

1
Hematology, Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; 2Dipartimento di Scienze Medico-Veterinarie, University of Parma, Parma, Italy;
3
A.I.R.C. Laboratory of Immunology and Tumors, Department of Experimental and Laboratory Medicine, Istituto Giannina Gaslini, Genova, Italy; 4Anatomia e Istologia patologica,
Azienda Ospedaliero-Universitaria di Parma, Parma, Italy; 5Department of Biosciences, University of Parma, Parma, Italy; 6Department of Clinical Sciences and Community Health,
University of Milan, and Hematology 1, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Milan, Italy; 7Department of Medicine, Division of Hematology/Oncology and
the Center for Bone Biology of UPMC, University of Pittsburgh, Pittsburgh, PA, USA; 8INSERM, U892, University of Nantes, CNRS, UMR 6299, Nantes, France; 9MD Anderson Cancer
Center, University of Texas, Houston, TX, USA and 10Division of Hematology Oncology, Indiana University School of Medicine Department of Medicine, Indianapolis, IN, USA.
Correspondence: Professor N Giuliani, Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, Parma 43126, Italy.
E-mail: nicola.giuliani@unipr.it
11
These authors contributed equally to this work.
Received 5 December 2012; revised 14 January 2013; accepted 17 January 2013; accepted article preview online 24 January 2013; advance online publication, 12 February 2013
Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1698
growth, angiogenesis and osteolysis in vivo using plasmocytoma Gene expression proling and microarray analysis
xenograft or intratibial mouse models previously used to evaluate The transcriptional proles of JJN3-anti-HIF-1a and JJN3-pLKO.1
the effect on myeloma cell growth and bone destruction.3234 cells exposed to hypoxic or normoxic conditions were analyzed.
To perform gene expression proles, total RNA was puried using the
RNeasy total RNA Isolation Kit (Qiagen, Valencia, CA, USA). Preparation of
MATERIALS AND METHODS biotin-labeled complementary RNA, hybridization to GeneChip Human
Genome U133 Plus2.0 Arrays and scanning (GeneChip Scanner 3000 7G,
Cells and cell culture conditions Affymetrix Inc., Santa Clara, CA, USA) were performed according to
Cell lines. Human myeloma cell lines (HMCLs) JJN3, Roswell Park Memorial manufacturers protocols. The raw intensity signals were extracted from
Institute medium (RPMI)-8226 and OPM-2 were purchased from DSMZ cell intensity (CELL) les and normalized using the robust multi-chip
(Braunschweig, Germany), while the U266 was obtained from the ATCC average (RMA) package for Bioconductor and custom GeneAnnot-based
(LGC Standards S.r.l., Venezia, Italy). HMCLs were cultured in RPMI media at Chip Denition Files version 2.2.0 in R software. The most differentially
10% fetal bovine serum with 2 nM of glutamine and antibiotics (Invitrogen expressed genes between two experimental conditions were dened as
Life Technologies, Milan, Italy). follows: for each gene, the ratio between the difference and the average
value in the expression signals under the two conditions was calculated.
HIF-1a knockdown. Lentivirus short hairpin RNA (shRNA) pool anti-HIF-1a Those genes with a minimum of 1.5 absolute fold change between hypoxic
(Sigma-Aldrich, Milan, Italy) was used for HIF-1a stable knockdown in and normoxic conditions were selected for further analysis. Then, those
HMCLs, whereas the pLKO.1 lentiviral vector was the empty control. genes with ratios exceeding two s.d. from the mean were considered
Recombinant lentivirus was produced by transient transfection of 293T differentially expressed. NetAffx (https://www.affymetrix.com/analysis/
cells following a standard protocol. HMCLs were infected as previously netaffx/) and Database for Annotation, Visualization and Integrated
described32, and selected in culture by the presence of 4 ug per ml Discovery (DAVID) (http://david.abcc.ncifcrf.gov) tools were used for the
puromycin for 21 days. Selected clones of HMCLs were then screened for functional annotation studies of the selected lists. The data have been
HIF-1a, HIF-1b, HIF-2a and HIF-3a mRNA and/or protein expression. Stably deposited at NCBI Gene Expression Omnibus repository (http://
transfected HMCLs were maintained in RPMI medium containing 10% of www.ncbi.nlm.mih.gov/geo) and are accessible through Gene Expression
fetal bovine serum with 4 ug per ml puromycin until use. Omnibus series accession number GSE40326.

Hypoxic and drugs treatments. HMCLs stably transfected with pLKO.1 or RNA isolation and reverse-transcriptase polymerase chain
anti-HIF-1a shRNA were incubated in the presence or absence of hypoxic reaction amplication
conditions (1%O2, 5%CO2) or treated with the hypoxic mimetic drug cobalt
chloride at 100 mM (Sigma-Aldrich, St Louis, MO, USA) or vehicle for 1224 h. In Total RNA was extracted from the cells using the RNeasy total RNA
selected experiments HMCLs stably transfected with pLKO.1 or anti- isolation kit (Qiagen, Valencia, CA, USA), and then quantied using a
HIF-1a were treated either in normoxic or in hypoxic conditions in the presence Nanodrop ND-100 (Celbio S.p.A., Milan, Italy). 1 mg of RNA was reverse-
of the absence of Bortezomib (supplied from Janssen-Cilag; Milan, Italy) at transcribed with 400 U Moloney murine leukemia reverse transcriptase
concentrations ranging from 150 nM of Lenalidomide (supplied by Celgene Italy (Applied Biosciences, Life Sciences, Carlsbad, CA, USA) in accordance with
srl, MiIan, Italy) (0.210 mM) or vehicle (dimethyl sulfoxide) for 2472 h. the manufacturers protocol. The complementary DNAs were amplied by
means of PCR using the following specic primer pairs:
Cell proliferation and viability assays. HMCLs transfected with pLKO.1 or
anti-HIF-1a were cultured in 96-well microtiter plates for 4896 h in the  HIF1A: F: 50 -CTCAAAGTCGGACAGCCTCA-30 ; R: 50 -CCCTGCAGTAGGTTTCTG
presence of 3H-thymidine (3H-TdR) (Biocompare South San Francisco, CA, CT-30
USA) and thymidine incorporation was detected by liquid scintillation  VEGFA: F:50 -CGAAGTGGTGAAGTTCATGGATG-30 ; R: 50 -TTCTGTTCAGTCTTT
spectroscopy (1205 Betaplate; Wallac; Markham, ON, Canada). Viability CCTGGTGAG-30
of HMCLs stably transfected with pLKO.1 or anti-HIF-1a was evaluated under  IL8: F: 50 -TACTCCAAACCTTTCCACCC-30 ; R: 50 -AACTTCTCCACAACCCTCT
both normoxic and hypoxic conditions after 2472 h of culture by adapted G-30
3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test assay  MMP9: F: 50 -CGCAGACATCGTCATCCAGT-30 ; R: 50 -GGATTGGCCTTGGAAG
(Cell Counting Kit-8; Alexis, Vinci-Biochem srl, Italy). Cell apoptosis was ATGA-30
determined by Apo 2.7 mAb staining (Immunotech, Fullerton, CA, USA) and  CCL2: F: 50 -ACTGAAGCTCGTACTCTC-30 ; R: 50 -CTTGGGTTGTGGAGTGAG-30
veried by FACScan, (BD Biosciences Italy, Milan, Italy).  GAPDH: F 5-CAACGGATTTGGTCGTATTG-3; R: 5-GGAAGATGGTGATGGG
ATTT-3

In vivo studies Annealing temperature: HIF1A: 59 1C; VEGFA: 66 1C; IL8: 64 1C; CCL2: 60 1C
Four to six-week-old males and females severe combined immunodeciency/ MMP9: 58 1C; GAPDH: 58 1C. Product size: HIF1A: 454 bp; VEGFA: 375 bp; IL8:
nonobese diabetic (SCID-NOD) mice (Harlan Laboratories, Udine, Italy) were 158 bp; CCL2: 354 bp MMP9: 369 bp GAPDH: 209 bp. Pictures of the
housed under specic pathogen-free conditions. All procedures involving electrophoresed complementary DNAs were recorded with a digital DC
animals were performed in accordance with the National 120 Kodak camera and quantied.
and International current regulations (D.l.vo 27/01/1992, n.116, European
Economic Community Council Directive 86/609, OJL 358, Dec. 1, 1987). Three Real-time quantitative PCR and angiogenesis PCR array
groups of six animals each were injected subcutaneously with 5  106 JJN3
cells stably transfected with anti-HIF1a containing plasmid vectors (JJN3-anti- Real-time PCR was performed using the TaqMan Gene Expression Assay
(Applied Biosystems Applera, Milan, Italy) for the following genes:
HIF-1a), or with JJN3 stably transfected with empty vector (JJN3-pLKO.1), or
JJN3 wild type (JJN3). Twenty days after tumor cell inoculation, mice were
killed and autopsies were performed. Tumor mass was measured as  HIF1A: HS00936379_M1
previously described.32 Maximum length and width of the tumor masses  HIF-1B (ARNT): HS00231048_M1
were measured with a caliper, and tumor volume (mm3) was calculated  HIF2A (EPAS1): HS01026149_M1
according to the following formula: 0.523  length  width2. Tumors were  HIF2B (ARNT2): HS00208298_M1
removed and subjected to immunohistochemical staining.  HIF3A: HS00223818_M1
In a separate set of experiments SCID and NIH-III nude mice (4 weeks of  BNIP3: HS00969291_M1
age) were injected intratibially with 20 ml of 5  104 cells of JJN3-anti-HIF-1a  VEGFA: HS99999034_M1
or JJN3-pLKO.1 or JJN3 or saline alone. All research protocols were approved  IL8: Hs99999034_M1
by the Pittsburgh VA Healthcare System Institutional Animal Care and Use  CCL2: HS00234140_M1
Committee. Four weeks after injection, the animals were killed and the tibias  MIP1A: Hs00234142_M1
were dissected out. Images of dissected tibias were acquired on a vivaCT 40  IL7: Hs00174202_M1
scanner (Scanco Medical, Bruttisellen, Switzerland) at resolution of 21 mm  DKK1: Hs00183740_M1
isotropic, reconstructed, and segmented for 3-dimensional display using the
instruments analysis algorithm software. Tissue samples and cell extracts The PCR amplications were performed in duplicate using the iCycler iQ
were obtained for immunohistochemical staining. Real-Time Detection System (Bio-Rad, Milan, Italy). The comparative

Leukemia (2013) 1697 1706 & 2013 Macmillan Publishers Limited


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1699
Ct method was applied to normalize the differences in the quantity RESULTS
and quality of RNA, and mRNA was quantied using the comparative Permanent HIF-1a silencing in HMCLs: effects on MM cell
DCt method using the endogenous reference gene ABL (DCt mean proliferation, survival and drug sensitivity
Ct genemean Ct ABL); DDCt was evaluated as the difference between the
DCt of a sample and the DCt of the control. The fold change in mRNA
HMCLs were initially checked for HIF1A mRNA and HIF-1a protein
expression was calculated as 2  DDCt. expression under normoxic conditions (Figure 1a). All HMCLs
The expression levels of the pro-angiogenic molecules were evaluated tested expressed HIF1A mRNA, whereas HIF-1a protein was
on mRNA extracted from tumors removed from SCID-NOD mice with the present at low levels in JJN3 and U266, but not in RPMI-8226
Human Angiogenesis RT2 Proler PCR Array and RT2 Real-Timer SyBR and OPM-2 (Figure 1a). All HMCLs overexpressed HIF-1a protein
green/ROX PCR Mix (PAHS-024, Superarray, SABiosciences, Frederick, MD, upon hypoxic treatment. The JJN3 and RPMI-8226 were used as
USA) that proles the expression of 84 key genes involved in modulating cell lines for HIF-1a inhibition studies. The selected clones were
the biological processes of angiogenesis. screened both under normoxic and hypoxic conditions for HIF1A
mRNA by qualitative PCR (Figure 1b), and for HIF1A, BNIP3, HIF1B,
HIF2A, HIF2B and HIF3A mRNA levels by real-time PCR. These
Western blot analysis
studies showed a selective inhibition of HIF1A and its target gene
Western blot analysis was performed as previously described.26 Nuclear BNIP3 by the shRNA pool used (Figure 1c left panel). HIF1B, HIF2A,
extracts were prepared using the Nuclear extraction kit (Active Motif,
Carlsbad, CA, USA) according to the manufacturers protocol. A total of
HIF2B and HIF3A mRNA were not inhibited by the shRNA pool
40 mg of nuclear extracts were tested. A polyclonal goat anti-HIF-1a Ab (Figure 1c). Western blot for HIF-1a, HIF-1b and HIF-3a proteins
(1:1000; R&D system, Minneapolis, MN, USA) or anti-HIF-1b or anti-HIF-2a showed specic inhibition of HIF-1a protein under both normoxia
or anti-HIF-3a (1:500; Novus Biologicals, Littleton, CO, USA) were used to and hypoxia (Figure 1d). HIF-2a and HIF-2 b were not expressed
detect HIF-1a, HIF-1b, HIF-2a and HIF-3a, respectively, whereas anti- either in normoxia or upon treatment with hypoxic mimetic
Histone H1 monoclonal Ab was used as internal control (Upstate, Lake (data not shown). Consistently HIF-1a activity was signicantly
Placid, NY, USA). A monoclonal anti-p27 (BD Pharmingen, San Jose, CA, inhibited in JJN3 anti-HIF-1a as compared with JJN3 pLKO.1 under
USA) was used as primary antibody to detect p27 on whole lysates. hypoxic conditions (Figure 1e). Similar results were obtained with
Appropriate secondary antibodies that were used: anti-goat RPMI-8226 (data not shown).
Immunoglobulin G (Rockland Immunochemicals, Gilberrtsville, PA, USA)
for HIF-1a, anti-mouse Immunoglobulin G (BD Biosciences) for HIF-1b,
The effect of HIF-1a suppression on HMCLs proliferation and
HIF-2a and p27 and anti-rabbit Immunoglobulin G (Chemicon survival was next investigated. Interestingly, there was no signi-
International, Millipore, Billerica, MA, USA) for HIF-3a. cant inhibition of cell proliferation either with JJN3 or RPMI-8226
(Figure 2a) at different time points. Cell viability was not
signicantly changed by HIF-1a suppression when checked at
HIF-1a activity 12 and 24 h either under normoxia or in hypoxia (Figure 2b).
HIF-1a activation in nuclear extracts of HMCLs was evaluated using an A statistically signicant effect on cell viability was observed in
Enzyme-linked immunosorbent assay based method (TransAM HIF-1 kit, normoxia condition at 72 h for JJN3 cells (  23%; P 0.01)
Active Motif) according to the manufacturers procedures. (Figure 2c upper panel) and maintained at the same entity for
longer culture period, but not for RPMI-8226 cells (Figure 2c, lower
panel). Treatment with lenalidomide at a wide range of
Enzyme-linked immunosorbent assays concentrations induced a signicantly higher inhibition of cell
Soluble VEGF, IL-8, IL-7 and CCL3 proteins were detected in the proliferation in JJN3 anti-HIF-1a as compared with JJN3 pLKO.1
conditioned media of HMCLs using Enzyme-linked immunosorbent assays (P 0.01) (Figure 2d) without changing their viability signicantly.
purchased from R&D system according to the manufacturers protocols. Consistently p27 expression was increased by lenalidomide in
Cytokine levels in the conditioned media were normalized to the number
JJN3 anti-HIF-1a as compared with JJN3 pLKO.1 (Figure 2e). On the
of cells at the end of culture period.
other hand, Bortezomib (410 nM) induced a similar rate of cell
proliferation and cell death in JJN3 anti-HIF-1a and JJN3 pLKO.1
Histological and Immunohistochemical analysis (data not shown).
Tissue samples obtained from tumors removed from mice injected either
subcutaneously or intratibially with JJN3-pLKO.1, JJN3-anti-HIF-1a and JJN3
were xed in 10% neutral buffered formalin, embedded in parafn, HIF-1a suppression affects the transcriptional prole of HMCLs:
sectioned at 3 mm, and stained with hematoxylin and eosin or with
inhibitory effect on the pro-angiogenic and pro-osteoclastogenic
Toluidine blu and Gomoris three-chromic methods. Tumors obtained from
genes
intratibial injections underwent EDTA decalcication before embedding in The transcriptional proles of JJN3 cells transduced with shRNA
parafn. On the basis of cell location and morphology, the number of all anti-HIF-1a (JJN3-anti-HIF-1a) were compared with those infected
Osteoclasts (OCLs) and active prismatic osteoblasts was evaluated on the with the control vector pLKO.1 (JJN3-pLKO.1) either under hypoxic
bone surface of each section (3  10 mm2). Osteocyte number and vitality or normoxic conditions. Among the signicantly modulated genes
were recorded on a total of 500 lacunae per histological section. (326 and 361 genes under hypoxic and normoxic condition,
Sections were immuno-stained either with mouse monoclonal anti-HIF- respectively) (Supplementary Table S1), we found downregulation
1a Ab (NOVUS Biologicals Littleton, CO; Working dilution 1:100) or mouse
anti-human VEGF (R&D; dilution 1:20) or with 1:100 diluted mouse anti- of the pro-angiogenic molecules VEGFA, IL8, CCL2, MMP9 in JJN3-
Ki67 primary antibody (Clone MIB-1, Dako, Carpinteria, CA, USA) for 30 min. anti-HIF-1a cells under both hypoxic and normoxic conditions.
HIF-1a and VEGF staining were revealed using the UltraVision LP Large Microarray data were further validated by both qualitative PCR
Volume Detection System HRP polymer (Thermo Scientic, Erembodegem, (Figure 3a) and real-time quantitative PCR, which showed that
Belgium) and quantied according to semiquantitative immunohisto- VEGFA and IL8 were induced by hypoxia and inhibited by HIF-1a
chemical score. Detection of Ki67 was performed using a high-sensitive suppression (Figure 3b), whereas CCL2 mRNA was not induced by
detection system (Advance-HRP, Dako Carpinteria, CA, USA) and 3,3- hypoxia but was inhibited by HIF-1a suppression either under
diaminobenzidine was used as chromogen substrate. Angiogenesis was normoxia or hypoxia. Finally, other genes with pro-angiogenic
evaluated on frozen tissues samples obtained from both series of mice. properties, such as IL10 and CCL5, were inhibited by HIF-1a
Tissues were xed in acetone and treated with rabbit anti-mouse CD34
(1:100; Santa Cruz Biotechnology, Santa Cruz, CA, USA). After washing suppression in JJN3 under both normoxic and hypoxic conditions
sections were incubated with a secondary antibody (1:250; Rat anti- (Supplementary Table S1). Consistent with our previous observa-
Immunoglobulin G horseradish peroxidase; Millipore) and reaction tion on small interfering RNA-mediated HIF-1a inhibition,27 stable
revealed with a solution of 3-30 -diaminobenzidine tetrahydrochloride HIF-1a suppression by shRNA signicantly blunted vessels
(liquid DAB substrate chromogen system, DAKO, Glostrup, DK). formation induced by the conditioned media of HMCLs as

& 2013 Macmillan Publishers Limited Leukemia (2013) 1697 1706


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1700

Figure 1. HIF-1a expression by HMCLs and validation of permanent HIF-1a knockdown by shRNA. HIF1A mRNA and HIF-1a protein expressions
were evaluated in RPMI-8226, U266, OPM-2 and JJN3 by qualitative PCR and western blot, respectively. JJN3 treated under hypoxic conditions
for 12 h was used as positive control for HIF-1a expression (a). Anti-HIF-1a Lentivirus shRNA pool was used for HIF-1a stable knockdown in
JJN3, whereas the pLKO.1 lentiviral vector was used as the empty control vector. Cells were infected and maintained in RPMI medium at 10%
of fetal bovine serum in the presence of puromycin at 4 ug/ml for 21 days. Cells were then incubated with the hypoxic mimetic drug cobalt
chloride or the vehicle for 12 h. HIF1A mRNA expression was evaluated by qualitative PCR using Glyceraldehyde 3-phosphate dehydrogenase
(GAPDH) mRNA as internal control (b). mRNA levels of HIF1A and BNIP3 as well as those of HIF1B, HIF2A, HIF2B and HIF3A were evaluated by real-
time PCR. Graphs represent the median fold change in the mRNA levels for three independent experiments performed in triplicate (c). HIF-1a,
HIF-1b and HIF-3a protein expression at the nuclear level were evaluated by western blot. Histone H1 was used as internal control (d). HIF-1a
activity in the nucleus was checked as described in the methods. Graphs represent the median optical density related to HIF-1a activity for
10 ug of proteins (e).

determined by AngioKit (TCS Biologies, London, UK) (data not of metastases to distant sites. Mice injected with the JJN3-anti-
shown). HIF-1a cells developed signicantly smaller tumors than mice
The effect of HIF-1a suppression on the production of inoculated with the JJN3-pLKO.1 (P 0.00018) or with JJN3
pro-osteoclastogenic cytokines by HMCLs was then investigated. (P 0.032) (Figure 4a-b). Both weight and volume of the tumors
A signicant inhibitory effect was observed for both MIP1A/CCL3 in JJN3-anti-HIF-1a mice were signicantly reduced as compared
and IL7 mRNA levels either under normoxia or in hypoxia as with JJN3-pLKO.1 (Figure 4c-d). The median weight of tumors
shown for JJN3 (Figure 3c). This inhibitory effect was also formed by JJN3-anti-HIF-1a was 0.05 g vs 0.3 g for JJN3-pLKO
conrmed at translational level under normoxia as shown for (P 0.0007) (Figure 4c). The median volume of tumors formed by
JJN3 (Figure 3d). In contrast, no signicant effect was observed on JJN3-anti-HIF-1a was 72.6 mm3 (range 3221 mm3) and that of
the expression of the osteoblast inhibitor gene DKK1 by JJN3 with JJN3-pLKO.1 was 369.8 mm3 (range 221.8671 mm3) (P 0.0012)
HIF-1a suppression (data not shown). (Figure 4d). Interestingly in the tumoral mass removed from
JJN3-anti-HIF-1a mice the microvascular density (number of
vessels positive for CD34/mm3) was signicantly reduced as
HIF-1a suppression in JJN3 cells blocks the growth of compared with that obtained from JJN3-pLKO.1 (JJN3-anti-HIF-1a
subcutaneous MM in SCID-NOD mice and inhibits angiogenesis vs. JJN3-pLKO.1:  76%; P 0.003). Similarly VEGF immunostain-
We next investigated whether inhibition of HIF1a in JJN3 may ing was reduced in JJN3-anti-HIF-1a derived tumors as compared
inuence tumor growth in vivo. Therefore, we analyzed the with JJN3 pLKO.1 injected mice (Figure 4e-f).
tumorigenicity of JJN3 wild type, JJN3 transfected with a plasmid
with silenced HIF1a (JJN3-anti-HIF-1a) and JJN3 with empty vector
(JJN3-pLKO.1) cells injected subcutaneously into SCID-NOD HIF-1a suppression in JJN3 cells blunts angiogenesis and bone
animals. Twenty days after cell inoculation, mice were killed, destruction in an intratibial mouse model
tumors removed and measured. At this time point, all animals The effect of HIF-1a suppression in MM cells was further
developed tumors that grew at the site of injection in the absence investigated in vivo in intratibial mouse model. The SCID mice

Leukemia (2013) 1697 1706 & 2013 Macmillan Publishers Limited


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1701

Figure 2. Effect of HIF-1a inhibition on the proliferation, viability and drug sensitivity of HMCLs. Cell proliferation was evaluated in JJN3-
pLKO.1, JJN3-anti-HIF-1a, RPMI-8226-pLKO.1 and RPMI-8226-anti-HIF-1a after 4896 h of culture by 3H-thymidine uptake as described in the
methods. Graphs and bars represent the means.d. of counts per minute of incorporated 3H-thymidine for three independent experiments
performed in six replicates (a). JJN3-pLKO.1 and JJN3-anti-HIF-1a were incubated under normoxic or hypoxic conditions for 1224 h and
viability evaluated by adapted MTT test. Graphs and bars represent the means.d. for cell viability in four independent experiments
performed in duplicate (b). Cell viability was evaluated in JJN3-pLKO.1, JJN3-anti-HIF-1a, RPMI-8226-pLKO.1 and RPMI-8226-anti-HIF-1a after
1272 h of culture. Graphs and bars represent the means.d. percentage change in the cell viability vs control (c). JJN3-pLKO.1 and JJN3-anti-
HIF-1a were incubated with Lenalidomide (0.210 mM) or vehicle (dimethyl sulfoxide, DMSO) under normoxic conditions for 48 h and cell
proliferation checked by 3H-thymidine uptake. Graph represents the mean % inhibition of cell proliferation (d). p27 protein expression was
evaluated by western blot analysis in cell lysates of JJN3-pLKO.1 and JJN3-anti-HIF-1a treated for 24 h with Lenalidomide (Len) (2 and 10 mM) or
vehicle (DMSO); actin was used as internal control (e).

were injected intratibially with saline or JJN3-pLKO.1 or JJN3-anti- subcutaneous model. Finally, micro-QCT analysis demonstrated
HIF-1a cells, and lytic lesions were allowed to develop for that mice injected with JJN3-pLKO.1 cells started developing
24 weeks before the mice were analyzed. Histological analysis detectable lytic lesions at 2 weeks after cell injection with
showed a signicant reduction in the tumor mass with an continued further bone deterioration through the 4 weeks, that
increase in the bone thickness in JJN3-anti-HIF-1a as compared ultimately involved the entire tibia leading to animal death from
with JJN3 pLKO.1 injected animals (Figure 5a). However, advanced disease. In contrast, the saline injected controls at 4
immunostaining for Ki67 showed a reduction in the number of weeks were similar to the 0-week time point, demonstrating that
Ki67-positive cells in JJN3-anti-HIF-1a as compared with JJN3 the effects detected were not the result of the injection process.
pLKO.1 mice although the difference did not reach statistical Interestingly, mice injected with JJN3-anti-HIF-1a cells showed a
signicance (Figure 5b). However, immunostaining for CD34 dramatic reduction of osteolytic lesions (Figure 5d). Finally,
showed a signicant reduction in the number of CD34-positive histological analysis showed a signicant reduction in the number
vessels in JJN3-anti-HIF-1a as compared with JJN3 pLKO.1 mice of OCLs in JJN3-anti-HIF-1a as compared with JJN3-pLKO.1 bone
(median number of vessels positive for CD34/mm3 22 vs 64, sections (Po0.05), whereas the number of active prismatic
P 0.001) (Figure 5c) similarly to what observed in the osteoblasts arranged in laminae did not signicantly changed as

& 2013 Macmillan Publishers Limited Leukemia (2013) 1697 1706


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1702

Figure 3. Effect of HIF-1a inhibition on the expression of the pro-angiogenic and pro-osteoclastogenic cytokines by JJN3 cells. The presence of
the mRNA transcripts for the pro-angiogenic genes VEGFA, IL8, MMP9, CCL2 was evaluated in JJN3-pLKO.1 and JJN3-anti-HIF-1a under both
normoxic and hypoxic conditions by qualitative PCR. GAPDH was used as internal control (a). mRNA levels of VEGFA, IL8 and CCL2 were
quantified by real-time PCR. Graph represents the median -DCt values of two independent experiments performed in triplicate (b). Gene
expression of the pro-osteoclastogenic genes MIP1A and IL7 was evaluated in JJN3-pLKO.1 and JJN3-anti-HIF-1a by real-time PCR. Graphs
represent the median fold change in mRNA levels for two independent experiments performed in triplicate (*Po0.05) (c). CCL3/MIP1a and
IL-7 protein levels were evaluated in the conditioned media of JJN3-pLKO.1 and JJN3-anti-HIF-1a. Graphs and bars represent the means.d.
protein levels for two independent experiments measured in duplicate. (*Po0.05) (d).

well as that of viable osteocytes for 500 lacunae (P 0.4; NS, data by shRNA was specic because no signicant inhibitory effect was
not shown). demonstrated on HIF1B, HIF2A, HIF2B and HIF3A expression.
Interestingly, we did not nd a signicant inhibitory in vitro effect
on MM cell proliferation by HIF-1a suppression only showing a
DISCUSSION signicant inhibitory effect on cell survival after 72 h in JJN3, but
It is well established that intratumoral hypoxia and consequently not in RPMI-8226. These results suggest that HIF-1a inhibition did
HIF-1a activation in solid tumors critically trigger the angiogenic not directly induce a signicant effect on MM cell proliferation and
switch, and induce modications of cancer cell metabolism survival. Similarly, it has been previously reported that small
leading to tumor progression and metastasis.1618,22 HIF-1a is interfering RNA anti-HIF-1a alone is not able to directly affect MM
overexpressed in many tumors22,23 including some hematological cell survival, but enhanced MM cell sensitivity to melphalan.36
malignancies such as MM.2729,35 We and others27,35 have recently Therefore, we further evaluated the potential effect of HIF-1a
shown that BM microenvironment is hypoxic both in MM stable suppression on the sensitivity of HMCLs to IMiDs and
patients27 and in mice.35 In addition HIF-1a also may be Bortezomib in vitro. Interestingly, we found that shRNA anti-HIF-1a
overexpressed by MM cells under normoxic conditions27,28 increased the inhibitory effect of IMiDs on HMCL proliferation.
dependent at least in part on c-myc upregulation.27 Finally, it This result was also supported by the previous nding that the
has been reported that hypoxia modulates the expression of pro- response to lenalidomide treatment involves oxidative
angiogenic genes such as VEGF and IL-827 by MM cells, and that stress response in MM cells.37 In contrast, we did not nd any
HIF-1a is involved in MM-induced angiogenesis in vitro.27 In turn, sensitization of MM cells to Bortezomib treatment. It has been
angiogenesis is able to support MM cell growth and survival.2 All reported that anti-MM effects of Bortezomib may be increased by
these results suggest that HIF-1a could be a potential therapeutic hypoxia and HIF-1a upregulation38 rather than by HIF-1a
target for MM. To test this hypothesis, we performed HIF-1a inhibition.
knockdown in HMCLs using a pool of shRNA anti-HIF-1a and Although, we did not nd a signicant effect on cell
evaluated the effect permanent suppression of HIF-1a on MM cell proliferation and survival by knockdown of HIF-1a, stable
proliferation, survival, the pro-angiogenic prole and tumor HIF-1a suppression by shRNA modied the pro-angiogenic and
growth in vivo. Firstly, we showed that the inhibition of HIF-1a osteoclastogenic proles of MM cells either under hypoxic or

Leukemia (2013) 1697 1706 & 2013 Macmillan Publishers Limited


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1703

Figure 4. HIF-1a inhibition in JJN3 blocks the development of subcutaneous plasmocytomas in SCID-NOD mice and inhibits angiogenesis.
Three groups of six SCID/NOD animals each were injected subcutaneously with 5  106 of JJN3, JJN3 transfected with a plasmid with silenced
HIF1a (JJN3-anti-HIF-1a) or JJN3 with empty vector (JJN3-pLKO.1). Twenty days after cell inoculation, mice were killed, tumors removed,
weighed and measured as described in the methods. Tissue samples from the masses were obtained for immunohistochemical staining.
Photographs represent either one representative mouse of each group of animals (JJN3, JJN3-anti-HIF-1a and JJN3-pLKO.1) after 20 days (a) or
tumor masses removed from JJN3-anti-HIF-1a and JJN3-pLKO.1 of two representative mice from each group (b). Box plots represent the
median weight (c) and volume (d) of the masses removed from all the mice injected with JJN3-anti-HIF-1a and JJN3-pLKO.1, respectively.
Images represent the immunostaining of CD34 (e) and VEGF (f ) antigens performed on frozen tissue and formalin fixed paraffin-embedded
samples, respectively, obtained from the plasmocytoma masses of two representative mice injected with JJN3-anti-HIF-1a and JJN3-pLKO.1.
Original magnification  400 (e) and  1000 (f ).

normoxic conditions. Microarray analysis and real-time PCR of MM-derived pro-osteoclastogenic factors that are known to be
showed that HIF-1a suppression signicantly inhibited some involved in osteoclast formation and bone destruction such as
pro-angiogenic factor genes such as VEGFA, IL-8, CCL2, MMP9. CCL3/MIP1a44 and IL-7.45 To the best of our knowledge, this is the
The capacity of HIF-1a to regulate VEGFA and IL8 gene expression rst time that the capacity of HIF-1a has been reported to regulate
by MM cells conrmed our previous data,27,39 and supports HIF-1a pro-osteoclastogenic cytokine in MM cells, although the evidence
as a key regulator of angiogenesis also in MM cells because that CCL3/MIP1a and IL-7 are potential target genes of HIF-1a has
VEGF9,10 and at least in part of IL-839,40 are well-known factors been shown in other cell types such as acute myeloid leukemia
involved in MM-induced angiogenesis. CCL2 and MMP9 could be cells for CCL3/MIP1a46 and mesenchymal/osteoblastic cells for
also involved in the pro-angiogenic role of HIF-1a, because IL-7.47
both cytokines are produced by MM cells and contribute to To evaluate the role of HIF-1a as a potential target in MM we
angiogenesis and BM spreading.4143 used two different in vivo models: the subcutaneously and
Interestingly, our data suggest that other than angiogenic the intratibial model. A dramatic effect on tumor burden was
cytokines, HIF-1a suppression affects expression and production demonstrated in both models by HIF-1a suppression showing a

& 2013 Macmillan Publishers Limited Leukemia (2013) 1697 1706


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1704

Figure 5. HIF-1a inhibition in JJN3 blunts myeloma-induced neo-angiogenesis and bone destruction after intratibial inoculation in mice. SCID
mice were injected intratibially with 20 ml containing 5  104 cells of JJN3-anti-HIF-1a or JJN3-pLKO.1 or saline alone. At 4 weeks after injection,
the animals were killed and the tibias were dissected out. Tissue samples were obtained for the histological evaluation and
immunoistochemistry. Hematoxylin and eosin staining of tissues samples obtained from two representative mice of each mice injected
with JJN3-anti-HIF-1a and JJN3-pLKO.1, respectively. Original magnification  400 (a). Immunostaining for Ki67 (b) and CD34 (c) antigens
performed on tissue samples of representative JJN3-anti-HIF-1a and JJN3-pLKO.1 mice (original magnification  630). Plots represent the
number of Ki67-positive cells (b) and the number of vessels X mm2 of tissues positive for CD34 (c), respectively (bars show the median value,
*Po0.001). Images show the dissected tibias of representative mice (saline, JJN3-anti-HIF-1a and JJN3-pLKO.1) acquired on a vivaCT 40 at
resolution of 21 mm isotropic, reconstructed and segmented for 3-dimensional display using the instruments analysis algorithm software (d).

signicant reduction of tumor mass. Because of the lack of a showing that HIF-1a suppression blocks MM-induced in vitro
signicant effect of HIF-1a suppression on cell proliferation and angiogenesis.27 The signicant inhibition of angiogenesis
survival of HMCLs in vitro, we guess that the anti-MM in vivo effect (evidenced by reduction in the number and length of vessels in
was mainly due to the inhibitory effect on angiogenesis. This was mice injected with JJN3-anti-HIF-1a) conrmed that the antitumor
supported by the capacity of HIF-1a to regulate the pro- effect of HIF-1a suppression was mainly due to the inhibition of
angiogenic proles of HMCLs in vitro and by our previous results angiogenesis. In support of these data, the inhibitory effect on

Leukemia (2013) 1697 1706 & 2013 Macmillan Publishers Limited


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1705
angiogenesis by HIF-1a suppression was accompanied by a 5 Munshi NC, Wilson C. Increased bone marrow microvessel density in newly
reduction of VEGF immunostaining and of the proliferative diagnosed multiple myeloma carries a poor prognosis. Semin Oncol 2001; 28:
index, Ki67, that could be related to angiogenesis in MM,6 565569.
although it did not reach a statistical signicance. Finally, in line 6 Alexandrakis MG, Passam FH, Dambaki C, Pappa CA, Stathopoulos EN. The relation
with our data, it has been previously reported that HIF-1a between bone marrow angiogenesis and the proliferation index Ki-67 in multiple
inhibition by RNA interference inhibits in vivo angiogenesis in myeloma. J Clin Pathol 2004; 57: 856860.
7 Yaccoby S. Advances in the understanding of myeloma bone disease and tumour
mice in other tumor models.48
growth. Br J Haematol 2010; 149: 311321.
In addition to the inhibition of angiogenesis, we showed in the 8 Roodman GD. Targeting the bone microenvironment in multiple myeloma.
intratibial bone model that HIF-1a suppression also affects MM- J Bone Miner Metab 2010; 28: 244250.
induced in vivo formation of osteolytic bone lesions. This could be 9 Dankbar B, Padro T, Leo R, Feldmann B, Kropff M, Mesters RM et al. Vascular
due in part to the inhibitory effect on MM growth via endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell
the inhibition of angiogenesis, but also to a direct effect on the interactions in multiple myeloma. Blood 2000; 95: 26302636.
production of pro-osteoclastogenic cytokines as shown in vitro. 10 Podar K, Anderson KC. The pathophisiologic role of VEGF in hematologic
Accordingly, histological analysis showed a signicant reduction of malignancies: therapeutic implications. Blood 2005; 105: 13831395.
the number of OCLs in JJN3 anti-HIF-1a as compared with JJN3- 11 Bisping G, Leo R, Wenning D, Dankbar B, Padro T, Kropff M et al. Paracrine
interactions of basic broblast growth factor and interleukin-6 in multiple
pLKO.1.
myeloma. Blood 2003; 101: 27752783.
The capacity of HIF-1a suppression to block the development of 12 Colla S, Morandi F, Lazzaretti M, Polistena P, Svaldi M, Coser P et al. Do human
osteolytic lesions in the intratibial MM model may explain in turn myeloma cells directly produce basic FGF? Blood 2003; 102: 30713072.
its anti-MM effect, because it is well-known that osteoclast 13 Shapiro VS, Mollenauer MN, Weiss A. Endogenous CD28 expressed on myeloma
activation supports MM cells growth in vivo.7 Similarly, it has cells up-regulates interleukin-8 production: implications for multiple myeloma
been previously reported that hypoxia and HIF-1a lead to the progression. Blood 2001; 98: 187193.
development of osteolytic lesions and tumor growth in breast 14 Giuliani N, Colla S, Lazzaretti M, Sala R, Roti G, Mancini C et al. Pro-angiogenic
cancer in vivo models.26 properties of human myeloma cells: production of angiopoietin-1 and its
In conclusion, as in other tumoral models where it has been potential relationship to myeloma-induced angiogenesis. Blood 2003; 102:
shown that HIF-1a inhibition exerts a potent in vivo antitumor 638645.
15 Colla S, Morandi F, Lazzaretti M, Rizzato R, Lunghi P, Bonomini S et al. Human
effect, including the squamous cell carcinoma, liver and lung
myeloma cells express the bone regulating gene Runx2/Cbfa1 and produce
cancer,48,49 our data indicate that selective HIF-1a inhibition osteopontin that is involved in angiogenesis in multiple myeloma patients.
results in a potent anti-MM effect by blocking angiogenesis and Leukemia 2005; 19: 21662176.
the development of osteolytic bone lesions. These results suggest 16 Brahimi-Horn MC, Chiche J, Pouyssegur J. Hypoxia and cancer. J Mol Med 2007;
that HIF-1a is a potential therapeutic target in MM. In this context, 85: 13011307.
it has been recently reported that adaphostin inhibits HIF-1a 17 Rankin EB, Giaccia AJ. The role of hypoxia-inducible factors in tumorigenesis.
expression and induces a signicant antiangiogenic and anti-MM Cell Death Differ 2008; 15: 678685.
activity in a MM xenograft mouse model.28 Furthermore, a 18 Liao D, Johnson RS. Hypoxia: a key regulator of angiogenesis in cancer. Cancer
growing number of novel anticancer agents have been Metastasis Rev 2007; 26: 281290.
19 Weidemann A, Johnson RS. Biology of HIF-1a. Cell Death Differ 2008; 15: 621627.
demonstrated to inhibit HIF-1a with different molecular
20 Lisy K, Peet DJ. Turn me on: regulating HIF transcriptional activity. Cell Death Differ
mechanisms including histone deacetylases, molecules acting on 2008; 15: 642649.
RAS-RAK-MEK-ERK pathways and PI3K-AKT, and mTOR inhibitors, 21 Yee Koh M, Spivak-Kroizman TR, Powis G. HIF-1 regulation: not so easy come, easy
that are also currently in phase I-II clinical trials in MM.1,29 Overall, go. Trends Biochem Sci 2008; 33: 526634.
our study provides the rationale for clinical trial with selective 22 Semenza GL. Regulation of cancer cell metabolism by hypoxia-inducible factor 1.
inhibitors of HIF-1a mRNA, such as EZN-296850 in MM patients. Semin Cancer Biol 2009; 19: 1216.
23 Hirota K, Semenza GL. ReguIation of angiogenesis by hypoxia-inducibIe factor 1.
Crit Rev OncoI Hematol 2006; 59: 1526.
CONFLICT OF INTEREST 24 Utting JC, Flanagan AM, Brandao-Burch A, Orriss IR, Arnett TR. Hypoxia stimulates
The authors declare no conict of interest. osteoclast formation from human peripheral blood. Cell Biochem Funct 2010; 28:
374380.
25 Bozec A, Bakiri L, Hoebertz A, Eferl R, Schilling AF, Komnenovic V et al. Osteoclast
size is controlled by Fra-2 through LIF/LIF-receptor signalling and hypoxia. Nature
ACKNOWLEDGEMENTS
2008; 454: 221225.
This study was supported by grants from the International Myeloma Foundation; 26 Hiraga T, Kizaka-Kondoh S, Hirota K, Hiraoka M, Yoneda T. Hypoxia and hypoxia-
and Associazione Italiana per la Ricerca sul Cancro A.I.R.C, IG2009 no. 8530 (N.G.), inducible factor-1 expression enhance osteolytic bone metastases of breast
no. IG4569 (A.N.) and no. 13018 (I.A.); Special Program Molecular Clinical Oncology 5 cancer. Cancer Res 2007; 67: 41574163.
per mille no. 9965 (N.G.) and no. 9980 (A.N.), the Multiple Myeloma Research 27 Colla S, Storti P, Donofrio G, Todoerti K, Bolzoni M, Lazzaretti M et al. Low bone
Foundation and grant support R01 AR059679, R21 CA141426, R01AR057308, and marrow oxygen tension and hypoxia-inducible factor-1a overexpression
Merit Review funds from the Veterans Administration. (G.D.R.). We thanks Ministero characterize patients with multiple myeloma: role on the transcriptional and
Italiano dell Istruzione, Universita e Ricerca (MIUR) and Italian Association against proangiogenic proles of CD138( ) cells. Leukemia 2010; 24: 19671970.
Leukemia, Lymphoma and Myeloma (AIL). We thanks Carla Palumbo and Marzia 28 Zhang J, Sattler M, Tonon G, Grabher C, Lababidi S, Zimmerhackl A et al. Targeting
Ferretti for their support in the histological analysis. angiogenesis via a c-Myc/hypoxia-inducible factor-1alpha-dependent pathway in
multiple myeloma. Cancer Res 2009; 69: 50825090.
29 Martin SK, Diamond P, Gronthos S, Peet DJ, Zannettino AC. The emerging role of
REFERENCES hypoxia, HIF-1 and HIF-2 in multiple myeloma. Leukemia 2011; 25: 15331542.
1 Podar K, Richardson PG, Hideshima T, Chauhan D, Anderson KC. The malignant 30 Azab AK, Hu J, Quang P, Azab F, Pitsillides C, Awwad R et al. Hypoxia promotes
clone and the bone-marrow environment. Best Pract Res Clin Haematol 2007; 20: dissemination of multiple myeloma through acquisition of epithelial to
597612. mesenchymal transition-like features. Blood 2012; 119: 57825794.
2 Vacca A, Ribatti D, Presta M, Minischetti M, Iurlaro M, Ria R et al. Bone marrow 31 Hu J, Handisides DR, Van Valckenborgh E, De Raeve H, Menu E, Vande Broek I et al.
neovascularization, plasma cell angiogenic potential, and matrix metalloprotei- Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated
nase-2 secretion parallel progression of human multiple myeloma. Blood 1999; 93: prodrug. Blood 2010; 116: 15241527.
30643073. 32 Storti P, Donofrio G, Colla S, Airoldi I, Bolzoni M, Agnelli L et al. HOXB7 expression
3 Vacca A, Ribatti D. Bone marrow angiogenesis in multiple myeloma. Leukemia by myeloma cells regulates their pro-angiogenic properties in multiple myeloma
2006; 20: 193199. patients. Leukemia 2011; 25: 527537.
4 Rajkumar SV, Leong T, Roche PC. Prognostic value of bone marrow angiogenesis 33 Campbell RA, Berenson JR. Animal models of multiple myeloma and their utility in
in multiple myeloma. Clin Cancer Res 2000; 6: 31113116. drug discovery. Curr Protoc Pharmacol 2008, Chapter 14, Unit 14: 9.

& 2013 Macmillan Publishers Limited Leukemia (2013) 1697 1706


Targeting HIF-1a in multiple myeloma in vivo
P Storti et al
1706
34 DSouza S, del Prete D, Jin S, Sun Q, Huston AJ, Kostov FE et al. G1 expressed in 42 Van Valckenborgh E, Bakkus M, Munaut C, Noel St A, Pierre Y, Asosingh K et al.
bone marrow stromal cells is a novel osteoblast suppressor in patients with Upregulation of matrix metalloproteinase-9 in murine 5T33 multiple myeloma
multiple myeloma bone disease. Blood 2011; 118: 68716880. cells by interaction with bone marrow endothelial cells. Int J Cancer 2002; 101:
35 Asosingh K, De Raeve H, de Ridder M, Storme GA, Willems A, Van Riet I et al. Role 512518.
of the hypoxic bone marrow microenvironment in 5T2MM murine myeloma 43 Purushothaman A, Chen L, Yang Y, Sanderson RD. Heparanase stimulation of
tumor progression. Haematologica 2005; 90: 810817. protease expression implicates it as a master regulator of the aggressive tumor
36 Hu Y, Kirito K, Yoshida K, Mitsumori T, Nakajima K, Nozaki Y et al. Inhibition of phenotype in myeloma. J Biol Chem 2008; 283: 3262832636.
hypoxia-inducible factor-1 function enhances the sensitivity of multiple myeloma 44 Choi SJ, Cruz JC, Craig F, Chung H, Devlin RD, Roodman GD et al. Macrophage
cells to melphalan. Mol Cancer Ther 2009; 8: 23292338. inammatory protein 1-alpha is a potential osteoclast stimulatory factor in
37 Colla S, Zhan F, Xiong W, Wu X, Xu H, Stephens O et al. The oxidative stress multiple myeloma. Blood 2000; 96: 671675.
response regulates DKK1 expression through the JNK signaling cascade in mul- 45 Giuliani N, Colla S, Sala R, Moroni M, Lazzaretti M, La Monica S et al. Human
tiple myeloma plasma cells. Blood 2007; 109: 44704477. myeloma cells stimulate the receptor activator of nuclear factor-kappa B ligand
(RANKL) in T lymphocytes: a potential role in multiple myeloma bone disease.
38 Veschini L, Belloni D, Foglieni C, Cangi MG, Ferrarini M, Caligaris-Cappio F et al.
Blood 2002; 100: 46154621.
Hypoxia-inducible transcription factor-1 alpha determines sensitivity of
46 Hateld KJ, Bedringsaas SL, Ryningen A, Gjertsen BT, Bruserud O. Hypoxia
endothelial cells to the proteosome inhibitor bortezomib. Blood 2007; 109:
increases HIF-1a expression and constitutive cytokine release by primary human
25652570.
acute myeloid leukaemia cells. Eur Cytokine Netw 2010; 21: 154164.
39 Colla S, Tagliaferri S, Morandi F, Lunghi P, Donofrio G, Martorana D et al. The new
47 Nishida C, Kusubata K, Tashiro Y, Gritli I, Sato A, Ohki-Koizumi M et al. MT1-MMP
tumor-suppressor gene inhibitor of growth family member 4 (ING4) regulates the
plays a critical role in hematopoiesis by regulating HIF-mediated chemokine/
production of proangiogenic molecules by myeloma cells and suppresses
cytokine gene transcription within niche cells. Blood 2012; 119: 54055416.
hypoxia-inducible factor-1 alpha (HIF-1alpha) activity: involvement in myeloma-
48 Krishnamachary B, Berg-Dixon S, Kelly B, Agani F, Feldser D, Ferreira G et al.
induced angiogenesis. Blood 2007; 110: 44644475.
Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer
40 Negaard HF, Iversen N, Bowitz-Lothe IM, Sandset PM, Steinsvik B, Ostenstad B et al. Res 2003; 63: 11381143.
Increased bone marrow microvascular density in haematological malignancies 49 Li L, Lin X, Staver M, Shoemaker A, Semizarov D, Fesik SW et al. Evaluating
is associated with differential regulation of angiogenic factors. Leukemia 2009; 23: hypoxia-inducible factor-1alpha as a cancer therapeutic target via inducible RNA
162169. interference in vivo. Cancer Res 2005; 65: 72497258.
41 Arendt BK, Velazquez-Dones A, Tschumper RC, Howell KG, Ansell SM, Witzig TE et 50 Greenberger LM, Horak ID, Filpula D, Sapra P, Westergaard M, Frydenlund HF et al.
al. Interleukin 6 induces monocyte chemoattractant protein-1 expression in A RNA antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhibits tumor
myeloma cells. Leukemia 2002; 16: 21422147. cell growth. Mol Cancer Ther 2008; 7: 35983608.

Supplementary Information accompanies the paper on the Leukemia website (http://www.nature.com/leu)

Leukemia (2013) 1697 1706 & 2013 Macmillan Publishers Limited

You might also like