10 1038@onc2012599 PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Oncogene (2013) 32, 4191–4202

& 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13


www.nature.com/onc

REVIEW
Thalidomide-analogue biology: immunological, molecular
and epigenetic targets in cancer therapy
J Shortt1,2,3, AK Hsu4 and RW Johnstone1,5

Thalidomide and its analogues (lenalidomide and pomalidomide) are small molecule glutamic acid derivatives of the
immunomodulatory drug (IMiD) class. In addition to the immuno-adjuvant and anti-inflammatory properties that define an IMiD,
the thalidomide analogues demonstrate an overlapping and diverse range of biological activities, including anti-angiogenic,
teratogenic and epigenetic effects. Importantly, the IMiDs possess anti-cancer activity with selectivity for molecularly defined
subgroups of hematological malignancies, specifically mature B-cell neoplasms and myelodysplasia with deletion of chromosome
5q. Emerging insight into the pathophysiological drivers of these IMiD-responsive disease states can now be synthesized using
previously disclosed IMiD activities and recently discovered thalidomide targets to build unifying models of IMiD mechanism of
action. Attention to mechanisms of IMiD-induced clinical toxicities, in particular the recently identified association of lenalidomide
with second primary malignancies, provides an additional tool for determination of drug mechanism. This review seeks to define
the molecular IMiD targets and biological outputs that underpin their anti-neoplastic activity. It is anticipated that elucidation of
important IMiD targets will allow the rational development of new-generation therapeutics with the potential to separate
thalidomide-analogue efficacy from clinical toxicity.

Oncogene (2013) 32, 4191–4202; doi:10.1038/onc.2012.599; published online 14 January 2013


Keywords: immune modulation; therapeutics; hematological malignancies

INTRODUCTION widespread withdrawal of thalidomide from clinical use in 1961.1–3


Thalidomide analogues are immunomodulatory drugs (IMiDs) with The serendipitous discovery of thalidomide’s efficacy in erythema
a range of biological effects and putative molecular targets. nodosum leprosum (ENL) reawakened interest in the drug.4,5
Thalidomide itself has clinical efficacy as an anti-inflammatory and Emerging insight into thalidomide’s anti-inflammatory and anti-
in a distinct cohort of neoplastic diseases including myelodyspla- angiogenic activity led to clinical trials in AIDS-related aphthous
sia (MDS) and multiple myeloma (MM). Despite an initially ulceration,6,7 Behcet’s syndrome,8 Crohn’s disease,9 cutaneous
infamous track record that spans 450 years of human exposure, lupus10 and various malignancies. The greatest testament to
thalidomide’s precise mechanisms of action remain undefined. thalidomide’s efficacy was that despite its teratogenicity it was re-
The diverse biological properties of thalidomide analogues instated as an FDA-approved treatment for ENL and MM (in 1998
indicate that they might possess divergent mechanistic activities and 2006, respectively, both available at http://www.fda.gov; last
in different diseases. However, thalidomide analogues are not accessed 28/05/2012).
indiscriminate anti-neoplastic agents. Rather, their clinical
specificity for a relatively narrow range of clearly definable
malignancies suggests a molecularly targeted mechanism of DEFINITION OF IMID
action, intricately linked to the biology of these susceptible The term IMiD refers to IMiDs licensed by Celgene Corporation
disease states. (Summit, NJ, USA) for the treatment of several inflammatory and
neoplastic diseases.11 The IMiDs were initially defined by their
capacity to inhibit lipopolysaccharide (LPS)-induced monocyte
HISTORICAL NOTE tumor necrosis factor (TNF)-a secretion, and further stratified by
It is speculated that thalidomide was tested as an antidote to their relative effects on other cytokines. Thus ‘class I’ IMiDs were
neurotoxins during World War II. This is consistent with characterized as broad inhibitors of LPS-induced inflammatory
thalidomide’s initial clinical indication as a sedative with anti- cytokines (for example, TNFa, interleukin (IL)-1b, IL-6 and IL-12),
emetic properties.1 Thalidomide was subsequently prescribed for whereas ‘class II’ compounds (so-called SelCIDS) showed
anxiety- and nausea-related conditions, including ‘morning selectivity for TNFa suppression.12 The SelCIDS are structurally
sickness’ to pregnant women in the 1950s.1 The ensuing diverse compounds unified by their capacity to antagonize
tragedy of birth defects including characteristic limb deformities phosphodiesterase-4. Class I (non-phosphodiesterase-4 inhibitory)
(dysmelia; stunted limb growth) is well documented and led to the IMiDs comprise thalidomide and its amino-substituted analogues
1
Gene Regulation Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Locked Bag 1, A’Beckett St, Melbourne, Victoria, Australia; 2Department of Clinical
Haematology, Monash Medical Centre, Clayton, Victoria, Australia; 3Department of Medicine, Southern Clinical School, Monash University, Clayton, Victoria, Australia;
4
Haematology Immunology Translational Research Laboratory, Cancer Immunology Program, Peter MacCallum Cancer Centre, Locked Bag 1, A’Beckett St, Melbourne, Victoria,
Australia and 5Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia. Correspondence: Professor RW Johnstone, The Peter
MacCallum Cancer Institute, Cancer Therapeutics Program, St Andrews Place, East Melbourne 3002, Victoria, Australia.
E-mail: ricky.johnstone@petermac.org
Received 17 September 2012; revised 4 November 2012; accepted 5 November 2012; published online 14 January 2013
Molecular IMiD targets
J Shortt et al
4192
O Mycobacterial infections such as tuberculous meningitis are
likewise attenuated in tandem with TNFa levels by IMiDs.21
N O Thalidomide pre-treatment also abrogates LPS-induced septic
NH shock in animal models of gram-negative septicemia.22 The in vitro
correlate is that thalidomide suppresses the release of TNFa from
O O human monocytes in response to mycobacterial antigens or LPS.23
thalidomide (Thalomid) TNFa suppression occurs downstream of receptor activation
and transcriptional events by acceleration of TNFa mRNA
O degradation,24 a mechanism that is specific to thalidomide
analogues (Figure 3a). In addition to enhancing the degradation
N O TNFa mRNA, the IMiDs were recently implicated as having a wider
NH role in translational regulation through downregulation of the
NH2 O eukaryotic elongation initiation factor (eIF)-4E.25 As the availability
lenalidomide (Revlimid, IMiD3, CC5013) of eIF4E is a rate limiting step in the recruitment of mRNAs-
bearing complex 50 -untranslated regions to the ribosome, IMiDs
NH2 O may preferentially downregulate the translation of a range of
proteins with ‘fragile’ 50 -untranslated regions. The translational
N O modulation of transcription factors upstream of pro-inflammatory
NH cascades is important in IMiD-related suppression of inflammation.
O
Cyclooxygenase (COX)-2 is a key regulator of arachidonic acid
O
metabolism and rate-limits prostaglandin production. LPS-
pomalidomide (Actimid, IMiD1, CC4047) stimulated COX-2 expression is transcriptionally regulated by
Figure 1. Chemical structures of the ‘IMiDs’, thalidomide and its CCAAT/enhancer-binding protein isoforms26,27 that are subject to
amino-phthaloyl-substituted analogues. 50 -cap-dependent translation in association with eIF4E.25
Therefore, the effects of IMiDs on eIF4E-regulated transcription
(Figure 1). These thalidomide analogues have an established anti- factors are likely to directly regulate the expression of
neoplastic role, and will be the focus of this review. pro-inflammatory gene loci, including COX-2.
The role of thalidomide’s anti-inflammatory targets in cancer
therapy is uncertain. Non-steroidal COX-2 enzymatic inhibitors
IMID CHEMISTRY AND DEVELOPMENT have demonstrable anti-cancer activities including anti-angiogenic
and anti-metastatic effects.28 COX-2 inhibitors have efficacy in
Thalidomide (a-(N-phthalimido)glutarimide) is a synthetic chemoprophylaxis of familial colorectal polyposis syndromes,29
glutamic acid derivative. Thalidomide is a chiral molecule; this is but have not shown significant benefit when added to
biologically important as the S(  ) and R( þ ) enantiomers mediate chemotherapy for established COX-2 overexpressing epithelial
different activities.13 However, segregation of biological effects by tumors.30 Myeloma cells harboring oncogenic RAS mutations also
administration of pure enantiomer is not feasible, as rapid show selective upregulation of COX-2 expression, and in these
physiological inter-conversion occurs in vivo.14,15 The cells COX-2 inhibition abrogates the protective effects of bone
second generation IMiDs CC-5013 (Revlimid, IMiD3, hereafter marrow stromal cells on myeloma following chemotherapy
lenalidomide) and CC-4047 (Actimid, IMiD1, hereafter treatment.31 There is some clinical evidence that COX-2 selective
pomalidomide), are amino-phthaloyl-substituted thalidomide inhibition has activity in MM,32 indicating that the anti-
analogues developed on the basis of augmented TNFa inflammatory effects of IMiDs may be relevant in this disease.
inhibition (Figure 1).16,17 In this respect, second generation IMiDs
are up to 50 000 times more potent than thalidomide in vitro12
and subsequent clinical development has disclosed divergent (2) Costimulation of T-cells
efficacy and toxicity profiles. Although TNFa suppression is an Correlative pre-clinical33 and clinical34 evidence suggests that
anti-inflammatory mechanism of IMiDs in ENL,18 TNFa inhibition is IMiD-mediated T-cell stimulation might augment anti-tumor
thought to be less relevant in other diseases such as MM (where immunity. The IMiDs modulate cytokine expression in a cell-type
TNFa-directed therapies do not have efficacy19). Thus, the notion and context-dependent manner to either suppress or stimulate
that second generation IMiDs are generally more potent than the release of selected interleukins. In contrast to the suppression
thalidomide is an oversimplification of thalidomide-analogue of LPS-induced monocyte inflammatory cytokine release, IMiDs
biology. Although thalidomide analogues share important costimulate the release of IL-2 and interferon (IFN)-g from
immunomodulatory hallmarks (Figure 2), they differ in toxicity T lymphocytes undergoing CD3 ligation, mitogenic stimulation
profiles and clinical efficacy between different disease types. or in response to dendritic cell presented super- or allo-antigens
Moreover, thalidomide resistant myeloma can be rescued by (Figure 3b).35,36 This results in T-cell proliferation, T-helper-1
treatment by second generation IMiDs, potentially indicating subset skewing and immuno-adjuvant anti-tumor effects33
divergent mechanistic activities within the same disease.20 including augmentation of cytotoxic effector functions.37,38 T-cell
costimulation by lenalidomide is only partially inhibited by
CTLA-4-Ig blockade of the B7/CD28 costimulatory axis.36
IMMUNOLOGICAL HALLMARKS OF IMID ACTIVITY Moreover, lenalidomide induces tyrosine-phosphorylation of
(1) Suppression of LPS-induced monocyte TNFa production and CD28, facilitating the recruitment and activation of upstream
associated anti-inflammatory activity nodes in intracellular signaling cascades including
The IMiDs possess clinically relevant anti-inflammatory activity. phosphatidylinositol-3-kinase (PI3K) and nuclear factor (NF)-
ENL is an immune complication of Mycobacterium leprae infection kB.36,37 As neither CD28 nor the IMiDs possess intrinsic kinase
manifesting with fever, skin lesions and multi-organ dysfunction activity, an indirect mechanism of CD28 phosphorylation, such as
because of antibody complex deposition, complement activation recruitment of other signaling proteins including protein
and cytokine dysregulation.5 Patients with ENL have increased kinase-C (PKC), is postulated. Pomalidomide costimulation
cellular and circulating levels of TNFa that rapidly reduce with activates PKC and PI3K to enhance nuclear translocation of
thalidomide therapy.18 Inflammatory complications of other nuclear factor of activated T-cells (NFAT)-2 and activated protein

Oncogene (2013) 4191 – 4202 & 2013 Macmillan Publishers Limited


Molecular IMiD targets
J Shortt et al
4193
Immunomodulatory
Anti-neoplastic
Anti-inflammatory (TNFα suppression)
Multiple myeloma
Co-stimulation of T-cells
Mature B-cell lymphomas
Augmentation of NK-cell effector function
Myelodysplasia with del 5q
T-helper subset skewing (TH1 response)

Teratogenic Clinical toxicities


Interspecies specificity Second primary malignancy
Limb deformities (phocomelia) O O
Venous thromboembolic disease
Cereblon binding NH
Peripheral neuropathy
Oxidative stress N O
Bone marrow suppression

Anti-angiogenic Epigenetic
VEGF expression DNA intercalation
FGF signaling Promoter acetylation / demethylation
Sphingolipid metabolism

Figure 2. The pleotropic effects of thalidomide analogues.

LPS

TLR

MYD88
TCR
AKT CD28
p
PKC
PI3K

mTOR

eIF4E 5’ cap-dependent translation


AP1 nuclear translocation

TNFα
NFAT2 NFAT2 IL2
NFκB AP1 IL2
TNFα

Figure 3. Mechanisms of thalidomide-analogue immunomodulatory activity. (a) Suppression of LPS-induced TNFa secretion: Activation of
Toll-like receptors (TLR) by LPS activates NF-kB signaling downstream of MYD88. Transactivation of the TNFa promoter by NF-kB results in
TNFa mRNA expression. Thalidomide accelerates the degradation of TNFa mRNA, while reducing the availability of eIF4E for 50 -cap-dependent
translation of the same TNFa mRNA transcripts. (b) Costimulation of T-cells: in conjunction with T-cell receptor (TCR) ligation IMiD exposure
induces phosphorylation (p) of the CD28 costimulatory receptor, possibly by recruitment of PKC. Downstream activation of signaling
pathways, including PI3K results in nuclear translocation of nuclear factor of activated T-cells (NFAT)-2 and activated protein (AP-1) to the IL-2
promoter and increased IL-2 secretion.

(AP)-1 resulting in increased DNA-binding at the IL-2 cells. Clinical responses to thalidomide in myeloma patients
promoter.37,39 Accordingly, PI3K inhibitors can reduce IL-2 likewise correspond with increasing numbers of peripheral NK
secretion in response to IMiD costimulation.37 cells.38

(3) Augmentation of cytotoxic T- and NK-cell effector functions


The augmentation of cytotoxic effector cells is a direct conse- OTHER DEFINING FEATURES OF THALIDOMIDE ANALOGUES
quence of the immunostimulatory activity of IMiDs on T-cells. The (1) Anti-angiogenic properties
secretion of IL-2 and IFN-g by CD4 T-cells following IMiD exposure Inflammatory cytokine networks and angiogenic pathways are
augments NK-cell mediated killing against both (allogeneic) inherently linked, as neo-vascularization is a consequence of the
leukaemic cell lines and autologous MM cells in vitro.38,40 The inflammatory response. Angiogenesis is important in tumor
importance of T-cell help in IMiD responses is highlighted by the growth and metastasis. Experiments initially designed to dissect
suppressive effects of calcineurin inhibitors and corticosteroids on thalidomide’s teratogenic potential subsequently demonstrated
NK-cell cytotoxicity assays.37,40,41 In vivo administration of IMiDs it’s anti-angiogenic properties.43 Thus, suppression of angio-
expands circulating NK-cell populations and synergizes with anti- genesis linked IMiD cytokine modulation and anti-cancer
CD20 antibody therapy against B-cell lymphoma xenografts in an activity. However, in contrast to the anti-inflammatory and
NK-cell-dependent fashion.42 This is potentially explained by the immunomodulatory properties that are conserved across
observation that lenalidomide augments antibody-dependent cell primate and rodent species’, anti-angiogenic effects appear to
cytotoxicity against rituximab (anti-CD20) treated CD20 þ tumor be relatively species specific. As the teratogenic effects of

& 2013 Macmillan Publishers Limited Oncogene (2013) 4191 – 4202


Molecular IMiD targets
J Shortt et al
4194
Notwithstanding the putative role of interspecies metabolism in
VEGFR FGFR
IMiD-mediated suppression of vascularization, thalidomide and its
analogues have demonstrable activity against several key
PI3K angiogenesis regulators. In vivo suppression of pro-angiogenic
Ceramide
factors including IL-6, TNFa, VEGF, NF-kB and prostaglandin
synthesis is predicted to indirectly downregulate angiogen-
esis.12,26,47 At the cellular level, the IMiDs downregulate
NFκB WNT AKT paracrine production of VEGF and IL-6 by both bone marrow
u stroma and MM cells, alone or in co-culture.48 VEGF-induced
u
Apoptosis
u
DDB1 angiogenesis is modulated by intracellular pro-survival signaling
CRBN
Cul4
pathways and sphingolipid metabolism.44,49 Thalidomide activates
H O H O
neutral sphingomyelinase to generate ceramide in human
u
Oxidative stress u umbilical vein endothelial cells resulting in both decreased
u CRBN cell growth and reduced expression of the VEGF receptors,
??? binding
neurophilin-1 and FLK-1 (Figure 4).49 Lenalidomide downregulates
the migratory response of human umbilical vein endothelial cells
Teratogenesis to VEGF and bFGF.44 Here, reduced endothelial cell migration
correlated with downregulation of AKT phosphorylation,
Figure 4. Overlapping mechanisms of teratogenesis and indicating that lenalidomide might uncouple the permissive
anti-angiogenesis conveyed by thalidomide targets. The anti- effects of VEGF-receptor ligation from intracellular signaling
angiogenic and teratogenic pathways modulated by thalidomide pathways. Together, these results indicate that the IMiDs
are inherently linked. Thalidomide downregulates VEGF receptor
(VEGFR) and fibroblast growth factor receptor (FGFR) signaling by modulate angiogenesis at multiple levels including systemic
several mechanisms, including generation of ceramide and down- cytokine networks, local tumor/stroma interactions, surface
regulation of effector pathways downstream of receptor signaling. expression of VEGF receptors and intracellular signaling
Anti-apoptotic effectors antagonized in this fashion include AKT, pathways thereof.
WNT and NF-kB. Concurrently, thalidomide induces oxidative stress
(pro-apoptotic) and binds cereblon (CRBN) to inhibit the E3
(2) Teratogenicity
ubiquitin–ligase activity of the CRBN/DDB-1/Cul4 complex. Direct
ubiquitination (u) targets of this complex outside of its Intense scientific activity has been dedicated to determining the
auto-ubiquitination function are not known. Together these molecular mechanisms of thalidomide teratogenicity.50 From a
pro-apoptotic, anti-angiogenic, oxidative and CRBN-mediated translational perspective, a non-teratogenic yet efficacious IMiD
effects combine to induce teratogenicity. would be highly desirable. Indeed the thalidomide enantiomers,
metabolites and analogues vary in their teratogenic potential51
and the anti-angiogenic and immunomodulatory properties of
IMiDs appear to be separable.52 Whether the anti-neoplastic
thalidomide show similar species specificity, it is likely that
activities of IMiDs are separable from teratogenic effects remains a
thalidomide’s anti-angiogenic capacity and teratogenicity are
point of conjecture. Proposed embryotoxic mechanisms include
also linked (Figure 4).
effects on angiogenesis,51 intracellular signaling,53–55 gene
D’Amato et al.43 first described anti-angiogenic activity of orally
regulation56 and the induction of oxidative stress.57 For an
administered thalidomide and a related teratogenic analogue,
excellent synopsis that attempts to unify existing conceptual
EM-12 that was apparent in experimental systems using rabbit but
frameworks, the reader is directed to a recent review by Ito et al.50
not chicken cells. Later, the same investigators demonstrated
As mechanisms of thalidomide teratogenicity have been
intraperitoneal, but not oral administration of thalidomide
extensively reviewed elsewhere, they will only be covered here
inhibited angiogenesis in bFGF and vascular endothelial growth
in brief:
factor (VEGF)-driven cornea micro-pocket assays in the mouse.13
Conversely, orally administered lenalidomide reduced basic (a) Angiogenesis. Thalidomide’s anti-angiogenic effects provide
fibroblast growth factor (bFGF)-induced angiogenesis in a rat insight into the predilection for limb deformities in its teratogenic
mesenteric window assay.44 The observation that route of phenotype. Developing limb buds comprise an immature vessel
administration (oral versus intraperitoneal) resulted in divergent network that is hypersensitive to thalidomide’s anti-angiogenic
anti-angiogenic effects with species specificity supported the effects.51 Thalidomide induces loss of newly formed vessels at
notion that in vivo IMiD metabolism modulates anti-angiogenic critical stages in limb development before the onset of
effects. This hypothesis was further substantiated using in vitro rat malformations.51 Changes in vasculature appear to precede
aorta and human endothelial cell culture systems that are changes in gene expression, cytostatic effects and cell death in
insensitive to the presence of non-metabolized thalidomide.45 the developing limb. This suggests defects in vascular
Here, supplementation of thalidomide with human or rabbit (but development are an upstream event in teratogenicity.
not rat) hepatic microsomes resulted in reduced microvessel
formation and endothelial cell growth. In a humanized myeloma (b) Oxidative stress. There is much evidence linking thalidomide
xenograft model, co-engraftment of human fetal liver cells into a teratogenicity to oxidative stress. Bioactivation of thalidomide
severe combined immunodeficient mouse augmented the anti- induces glutathione depletion and DNA-oxidation consistent with
myeloma activity of thalidomide.46 Myeloma xenografts from the capacity to generate reactive oxygen species.57 Interestingly,
thalidomide-treated, human fetal liver bearing mice showed the threshold for thalidomide-induced free-radical generation and
reduced microvessel density compared with non-fetal liver embryonic DNA-oxidation is much lower in rabbits than mice,
transplanted hosts. It was concluded that human hepatic corresponding with greater sensitivity to dysmorphogenesis. Pre-
metabolism was required for thalidomide’s anti-myeloma activity treatment of rabbits with the free-radical spin trapping agent,
in a mouse. However, this interpretation requires caution as alpha-phenyl-N-t-butylnitrone abolishes thalidomide-induced
thalidomide metabolites were not assayed, the immunological DNA-oxidation and teratogenicity.57 The induction of oxidative
effects of fetal liver grafts (which contain hematopoietic stem stress in developing limb buds (potentially compounded by tissue
cells) were not accounted for, and thalidomide was administered hypoxia) induces alterations pro-survival signaling pathways
parenterally rather than orally. including NF-kB,58 bone morphogenic protein/Dickkopf-1/Wnt54

Oncogene (2013) 4191 – 4202 & 2013 Macmillan Publishers Limited


Molecular IMiD targets
J Shortt et al
4195
and PI3K/AKT55 culminating in downregulation of FGF and inhibition to species-specific oxidative stress were elucidated, this
apoptosis induction (Figure 4). might finally provide a unifying model (Figure 4).

(c) Dysfunction of the cereblon/DNA-binding protein 1 ubiquitin–


ligase complex. Cereblon is a ubiquitously expressed protein that EVIDENCE FOR GENETIC AND EPIGENETIC EFFECTS OF IMIDS
was identified as a candidate gene in a mild form of autosomal Although not mutagenic,66 thalidomide intercalates DNA.67,68
recessive, non-syndromal mental retardation.59,60 Cereblon binds Specifically, S(  ) thalidomide is predicted to intercalate into the
voltage-gated chloride channels in the retina61 and calcium- major groove of the DNA-double helix structure at purine sites
activated potassium channels in the brain.62 It also modulates (with greater affinity for guanine over adenine) where its
AMP-activated protein kinase activity.63 In autosomal recessive, glutarimide moiety oriented in a 30 -direction.56 The preferential
non-syndromal mental retardation, a truncating cereblon point affinity of thalidomide for guanine nucleotides potentially explains
mutation interrupts C-terminal myristoylation and phosphory- differential regulation of genes bearing GC-rich gene promoters.
lation sequences, potentially disrupting intracellular localization The transcription factor Sp1 binds GC-boxes within promoter
and signaling activity, respectively.59,60 regions and docks within the major groove of DNA that would be
Cereblon is required for the E3 ubiquitin–ligase activity of a occupied by intercalated thalidomide.69,70 Candidate genes
multi-protein complex containing DNA-binding protein 1 and regulated by Sp1 include insulin-like growth factor and its
Cullin 4 (Figure 4). Thalidomide was recently demonstrated to receptor, insulin receptor substrate-1, FGF-2, FGF receptors and
directly modulate FGF-mediated angiogenesis via a cereblon- several integrins. As these have a central roles in angiogenesis and
dependent pathway.53 Using thalidomide to ‘pull down’ potential embryo development, modulation of gene expression via
drug targets from crude cell extracts, Ito et al.53 identified cereblon intercalation at GC-rich promoters has been proposed as a
as a thalidomide-binding protein. The antagonism of cereblon by mechanism for thalidomide teratogenesis (reviewed in Stephens
thalidomide was subsequently shown to be necessary but et al.56).
insufficient to induce teratogenicity in Zebrafish and chick The DNA-intercalation model provides a conceptual framework
embryos. In the Zebrafish model, thalidomide-induced terato- for how thalidomide might reduce the efficiency of transcription
genicity could be phenocopied by cereblon knockdown and at GC-rich promoters. However, emerging evidence suggests the
rescued by overexpression of a thalidomide-insensitive cereblon IMiDs epigenetically promote the transactivation of genes at the
mutant protein.53 Thalidomide exposure or cereblon knockdown same GC-rich loci. Lenalidomide and pomalidomide induce
resulted in reduced FGF expression in the apical ectodermal ridge, growth arrest in selected myeloma and B-cell lymphoma cell
providing a mechanistic link between thalidomide, cereblon and lines. This correlates with the induction of the cell cycle regulator
FGF-mediated angiogenesis in teratogenicity. p21WAF-1 and concomitant inhibition of cyclin-dependent kinases,
Although the characterization of cereblon as a thalidomide- hypo-phosphorylation of Rb and G0–G1 arrest.71 Pomalidomide
target provided much insight into teratogenic mechanisms, the rapidly upregulates CDKN1A mRNA expression via p53-
role of complementary pathways remains undefined. In the independent recruitment of Sp1 to a proximal GC-rich region in
human, cereblon mutations causing autosomal recessive, non- the CDKN1A promoter (Figure 5a).72 Sp1 recruitment occurs in the
syndromal mental retardation do not associate with limb absence of a change in absolute Sp1 levels and is facilitated by the
defects.60 Conversely, persons suffering from thalidomide- acetylation of histone H3 on lysines 9 and 14. As the IMiDs do not
induced dysmelia do not demonstrate intellectual disability.1 As inhibit histone deacetylases, an indirect mechanism whereby
the protein structure of cereblon is highly conserved, thalidomide recruitment of the histone demethylase, LSD-1 to H3K9 was
should not discriminate between rodent and human (or Zebrafish) suggested.72 In this model, the acetylation of H3K9/14 is co-
protein.53 Thus, the well described associations between species- regulated by LSD-1 demethylation at the same loci.
specific thalidomide metabolism and angiogenic effects are not Epigenetic mechanisms are also implicated in IMiD-related
well explained by the cereblon-model of teratogenicity. Another modulation of cytokine expression (Figure 5b).73 Cytokine
caveat to the interpretation of Zebrafish modeling is the receptor ligation activates intracellular signaling through the
supraphysiological thalidomide dosing required to induce anti- JAK/STAT pathway. STAT target genes include the suppressor of
angiogenic and teratogenic phenotypes. Myeloma patients cytokine signaling (SOCS) genes that attenuate JAK/STAT
receive 50–200 mg/day of thalidomide. A dose-escalation study activity.74 SOCS1 is a negative regulator of IFN-g, IL-2 and IL-6.
in elderly prostate cancer patients recorded maximum steady- Hypermethylation of the SOCS1 promoter is reported in myeloma
state plasma concentrations of 11 mg/ml (B40 mM) in persons patients, leading to augmented expression of IL-6, a potent
receiving 1.2 g of thalidomide daily.64 However, Ito et al.53 plasma cell growth factor.75 Pomalidomide upregulated myeloma
reported dysmorphogenesis with 400 mM of thalidomide applied cell SOCS1 expression concomitant with demethylation of its
directly to dechorionated embryos, diluted from a 400 mM DMSO silenced promoter region.73 Demethylation of the SOCS1 promoter
stock solution. As thalidomide is relatively insoluble, repeated by pomalidomide or 5-azacitidine correlated with increased
cycles of heating (to 65 1C) and agitation were required to myeloma cell sensitivity to subsequent lysis by cytotoxic
solubilize thalidomide, introducing the potential for pre-analytical T lymphocytes. Interestingly, the epigenetic modulation of
drug modification. Others have reported Zebrafish embryos to be SOCS1 was tumor specific, as the promoter methylation was
much less sensitive to thalidomide application. For example, unchanged in effector lymphocyte populations.73
Cheng et al.65 reported that thalidomide concentrations of
2 mg/ml (B7–8 mM) were the minimum requirement to induce
vascular deformities in transgenic fli1a: EGFP Zebrafish. As both ACTIVITY IN MDS WITH DEL 5Q
investigators used similar methodology, the reasons for this MDS refers to a group of clonal bone marrow stem-cell disorders
10-fold discrepancy in potency are unclear. In either case, the characterized by ineffective hematopoiesis and potential trans-
concentrations used are much higher than those measured in formation to acute myeloid leukemia. MDS with associated
human subjects. deletion of the long arm of chromosome 5 (del 5q) is an MDS
Although it is apparent that cereblon is a potentially important subtype characterized by refractory macrocytic anemia, well
IMiD target, there are still many unanswered questions regarding preserved thrombopoiesis and low bone marrow blasts.76 Unlike
mechanisms of thalidomide embryotoxicity. In particular, the other MDS, 5q- syndrome has a high response rate to
grossly divergent interspecies sensitivity to teratogenesis is lenalidomide, including cytogenetic remissions and improved
unresolved. If a pathway linking thalidomide-related cereblon erythropoiesis.77 The clinical phenotype of MDS with del 5q is

& 2013 Macmillan Publishers Limited Oncogene (2013) 4191 – 4202


Molecular IMiD targets
J Shortt et al
4196
megakaryopoiesis to co-operate with RPS14 deletion in the
Sp1 induction of anemia while promoting platelet production.
The predominant clinical response to lenalidomide in MDS
p21 is improved erythropoiesis with a significant proportion of
Me Me Ac Sp1 patients achieving independence from red cell transfusions.77
CDKN1A CDKN1A This is associated with cytogenetic improvement, but occurs at the
Me
LSD-1 Ac expense of thrombocytopenia and neutropenia. Despite detailed
insight into the molecular mechanism of anemia and
Me Me thrombocytosis in MDS with del 5q, the mechanism of
Me
lenalidomide activity remains something of an enigma as RPS14
is not an IMiD-responsive gene.85 Gene-expression profiling of
lenalidomide-treated del 5q erythroblasts revealed significant
IL6 upregulation of only one CDR gene, SPARC.85 SPARC regulates
interactions with the extracellular matrix, mediating anti-
proliferative and anti-angiogenic effects in common with IMiD-
related activity. An alternative hypothesis is that lenalidomide
JAK further downregulates the function of haploinsufficient proteins
coded in the CDR by post-transcriptional and post-translational
STAT SOCS1
mechanisms. This might explain selective toxicity for del 5q cells
over normal cells with a diploid gene complement at the same
locus. For example, del 5q cells are haplodeficient for the protein
phosphatases Cdc25C and PP2A and the expression of
these genes is lower in MDS with del 5q than with other
MDS.86 Lenalidomide increases the retention of inhibitory
BCL2 phosphorylation residues on cell cycle regulators targeted by
STAT
IL6 target genes
Cdc25C and PP2A.86 This correlates with potent inhibition
of recombinant human Cdc25C phosphatase activity by
SOCS1 lenalidomide in the nanomolar concentration range in vitro.86
Me Me Lenalidomide’s effects on PP2A activity appear to be indirect, as
SOCS1 SOCS1
Me
they cannot be demonstrated on purified protein in the same
assays. As Cdc25C and PP2A regulate the cell cycle, antagonism by
Figure 5. Examples of epigenetic modulation of gene targets by lenalidomide explains the selective induction of growth arrest and
IMiDs. (a) Genes bearing GC-rich promoter regions are preferentially apoptosis in del 5q cells compared with their normal counterparts.
targeted by IMiDs either by direct DNA intercalation (not shown) or The erythroid phenotype of MDS with del 5q is p53
alterations in methylation status (Me) and histone acetylation (Ac). dependent.82 Moreover, neoplastic escape from lenalidomide
The transcription factor Sp1 is excluded from a GC-rich region of the in vivo is associated with selection for sub-clones with mutant
CDKN1A promoter by methylation. Recruitment of the demethylase
LSD-1 allows histone H3 acetylation at lysines 9 and 14, opening p53.87 Ribosomal biogenesis occurs in the nucleolus, which also
the promoter to transcription of the cell cycle inhibitor p21WAF-1. provides a sink for the sequestration of the p53 ubiquitin ligase,
(b) SOCS1 feedback suppresses JAK/STAT signaling in response to MDM2 (Figure 6a). Nucleolar stress due to a range of causes
cytokine receptor ligation including IL-6. In myeloma cells, the including ribosomal protein deficiency results in the dispersion of
SOCS1 promoter is silenced by hypermethylation, amplifying the free RPs that are able sequester MDM2 independently of its co-
transcription of IL-6 regulated pro-survival effectors (for example, regulator ARF. The sequestration of MDM2 allows stabilization of
BCL2). IMiD treatment demethylates the SOCS1 promoter, restoring p53. Lenalidomide might therefore attenuate the severity of
SOCS1 expression and dampening pathophysiological JAK/STAT dyserythropoiesis indirectly by either reducing ribosomal stress or
signaling. promoting p53 degradation. The suppressive effects of lenalido-
mide on mRNA translation25 potentially reduces the biosynthetic
demands of a cell on haploinsufficient ribosome biogenesis.
Lenalidomide also promoted degradation of p53 in the context of
specifically mapped to loss of a common deleted region (CDR) on 5q deletion by antagonising PP2A-enhanced ubiquitin-mediated
chromosome 5.78 This CDR provides a list of 40 genes implicated clearance of MDM2.88 Thus, it is possible that lenalidomide
in the biology of 5q- syndrome and therefore a template for the treatment reduces ribosomal stress and the p53 response thereof
discovery of lenalidomide-responsive elements. In a seminal to ameliorate dyserythropoiesis (Figure 6a). The observation that
paper, Ebert et al.79 used an RNA-mediated interference lenalidomide can destabilize p53 is clinically significant, as p53
approach to interrogate the CDR. They identified ribosomal suppression is a highly oncogenic effect. IMiD-mediated p53
protein (RP) S14 haploinsufficiency as a key determinate of the suppression provides a mechanistic explanation for the recently
erythroid differentiation defect in MDS with del 5q. This correlated published observation that prolonged exposure to lenalidomide is
well with prior descriptions of heterozygous germline mutations associated with the development of second cancers (discussed
for several other RPs in patients with Diamond–Blackfan anemia, a further below).89–92
congenital bone marrow failure syndrome.80,81 The erythroid
defect conveyed by RPS14 loss is p53 dependent, indicating that
activation of the p53 tumor suppressor pathway is necessary for ACTIVITY IN MM AND MATURE B-CELL NEOPLASMS
RP haploinsufficiency to induce anemia.82,83 Although RP MM is a malignancy of immunoglobulin producing plasma cells
haploinsufficiency contributes to the p53-dependent anemia of associated with systemic toxicities including renal failure, anemia,
MDS with del 5q, other genes are implicated in the generation immunoparesis and destructive skeletal lesions.93 The mechanism
of dysplastic megakaryocytic hyperplasia and thrombocytosis. of action of IMiDs in MM is complex and likely reflects the
In particular, loss of microRNA-145 in the CDR results in increased sum-total of a range of independent targets including direct
expression of the transcription factor Fli-1 (ref. 84). Fli-1 toxicity to MM cells, disruption of the protective bone marrow
overexpression causes a shift from erythropoiesis to microenvironment and augmentation of host-mediated anti-

Oncogene (2013) 4191 – 4202 & 2013 Macmillan Publishers Limited


Molecular IMiD targets
J Shortt et al
4197
IL6
SPARC

AKT

Ineffective erythropoiesis SOCS1 PI3K


(macrocytic anemia) MYD88

mTOR

AKT

p p53
RP eIF4E PP2A NFκB
CRBN
mTOR

Ribosomal stress
(Haploinsuffcient RPS14) eIF4E

RP RP MDM2
RP
+
RP IRF-4 cMYC

Free ribosomal proteins

Figure 6. Cell intrinsic mechanisms of IMiD activity in specific disease states. (a) MDS with del 5q: haploinsufficiency for RPS14 induces
ribosomal stress and releases free ribosomal proteins to sequester the p53 ubiquitin ligase, MDM2. Activation of the p53 tumor suppressor
pathway causes ineffective erythropoiesis (macrocytic anemia). IMiD treatment may ameliorate ribosomal stress by antagonism of mTOR/
eIF4E-regulated protein translation while concurrently inhibiting PP2A activity to stabilize MDM2 levels and dampen the p53 response. In
addition, upregulation of SPARC expression has potential anti-proliferative effects through interactions with the tumor microenvironment.
RPS14, PP2A and SPARC are all genes in the 5q common deleted region. (b) Mature B-cell neoplasms (for example, MM and diffuse large B-cell
lymphoma of activated B-cell phenotype (DLBCL-ABC) subtype) show activation of pro-survival NF-kB and IRF-4 signaling through diverse
mechanisms including MYD88 mutation and IL-6 receptor signaling. IRF-4 and cMYC translation is rate limited by the availability of eIF4E; the
mutually permissive effects of IRF-4, cMYC and eIF4E form an oncogenic ‘feed-forward loop’. IMiD treatment disrupts this pathophysiological
network at multiple levels including upregulation of SOCS1, and antagonism of NF-kB dependent IRF-4/cMYC axis. Cereblon binding is
required for some of this cell intrinsic anti-neoplastic activity.

tumor immunity. Myeloma is a clinically and molecularly pathway. Similarly, Mitsiades et al.98 correlated IMiD-induced
heterogeneous disease.93,94 Despite this heterogeneity, IMiD- downregulation of NF-kB with activation of the extrinsic apoptotic
responsiveness is a defining feature of the majority of MM at pathway and sensitization to death-receptor agonists including
presentation; a phenotype shared with selected other mature TNF-related apoptosis related ligand in human myeloma cell lines.
‘post-germinal center’ B-cell malignancies.95 This potentially Interestingly, the direct cytostatic and apoptotic effects of IMiDs
indicates that the IMiDs target fundamental aspects of mature are less evident in lymphomas and myelomas after depletion of
B-cell biology. cereblon, linking this mediator of teratogenicity to anti-tumor
effects.95,99,100 Acutely, shRNA-mediated cereblon knockdown
induces cytostatic and apoptotic effects in MM cell lines that
(a) Myeloma cell autonomous effects correlates with a reduction in IRF-4 expression.100 Those MM cells
An intriguing property of the IMiDs is anti-tumor activity with surviving sustained cereblon knockdown are lenalidomide
selectivity for neoplasms of mature B-cell origin. In rare instances resistant when compared with their parental counterparts.
this is explained by 5q loss,71 however, a more complex molecular Conversely, prolonged (4 to 6 months) in vitro treatment with
interaction is implicated in the majority of cases. Mature B-cell lenalidomide generates IMiD-resistant MM cell lines that show
malignancies including MM and diffuse large B-cell lymphoma of reduced cereblon expression relative to non-lenalidomide treated
activated B-cell phenotype show dependence on constitutive controls.99,100 The resistance phenotype is specific to IMiDs, as
NF-kB signaling and upregulation of interferon regulatory factor lenalidomide-conditioned MM cell lines remained sensitive to
(IRF)-4 (Figure 6b).95 Mutually permissive ‘feed-forward loops’ apoptosis induction by dexamethasone. Cereblon mRNA
linking IRF-4 to cMYC implicate the former as an ‘oncogene of expression was also reportedly reduced in primary patient MM
addiction’ in MM.96 MYD88 is an adaptor protein that mediates samples with clinically resistant disease following a period of
interleukin and toll-like receptor signaling. Gain-of-function lenalidomide therapy.100 However, the effects of cereblon
MYD88 mutations and copy number amplification are observed knockdown did not entirely phenocopy IMiD treatment. For
in more than half of diffuse large B-cell lymphoma of activated example, gene expression profiling of OPM2 MM cells identified
B-cell phenotype and are associated with increased IRF-4 2398 differentially regulated genes following lenalidomide
expression.97 Genomic profiling of MM revealed frequent treatment, but only 123 were concordant with the cereblon
mutations of genes implicated in NF-kB signaling and IRF-4 knockdown gene set of 768 genes.100 Thus, although it is apparent
itself.94 Thus, diverse oncogenic mechanism with the net effect of that cereblon mediates some of the cell-intrinsic activity of IMiDs
NF-kB and IRF-4 activation link these IMiD-responsive diseases. in MM, complementary mechanisms and other molecular targets
Recent evidence suggests that IMiDs can downregulate the are likely to be at play. This is consistent with observations in
NF-kB/IRF-4 axis at multiple levels. In MM, antagonism of the teratogenicity, where cereblon binding is necessary but
eIF4E translational axis decreases CCAAT/enhancer-binding pro- insufficient to explain the teratogenic phenotype.53
teinb protein expression to impair transcription at the IRF-4
locus.25 In diffuse large B-cell lymphoma of activated B-cell
phenotype, lenalidomide relieves the suppressive effects of (b) Disruption of the myeloma niche and supportive cytokine
constitutive MYD88 signaling on interferon-b by downregulating networks
IRF-4 (ref. 95). The ensuing lethal type-1 interferon response The bone marrow microenvironment provides a permissive niche
mediates apoptosis through the extrinsic (death receptor) for MM growth and promotes resistance to the apoptotic effects

& 2013 Macmillan Publishers Limited Oncogene (2013) 4191 – 4202


Molecular IMiD targets
J Shortt et al
4198
of conventional therapeutics.101,102 Elaborate co-culture systems The contribution of T-cells to immune-mediated control of MM
have defined both direct interactions with bone marrow stromal in vivo has been a difficult question to address, with conflicting
cells and humoral components of cytokine networks as essential reports characterizing both impaired/exhausted and activated
to MM viability.101,103–105 In particular, osteoclastogenic cytokines clonal expansions of MM-specific T-cells in MM patients.126–129
of the TNF-family and IL-6 are implicated in myeloma cell growth There is agreement, however, on the responsiveness of T-cells to
and skeletal destruction.102,106 Paracrine and autocrine interleukin immunomodulatory agents. The stimulatory effect of IMiDs on
networks combine with cell adhesion molecules to activate T-cell function is driven by PI3K-mediated activation of nuclear
intracellular pro-survival pathways including PI3K/AKT, NF-kB factor of activated T-cells-2.37,130 This results in increased
and RAS/MAPK.107,108 Myeloma cell adherence to bone marrow production of IL-2 and NK-cell stimulation.37,40 Further to this,
stroma also upregulates VEGF secretion to promote IMiDs appear to polarize T-cells towards a Th-1 phenotype
angiogenesis.48 By downregulating cytoprotective cytokines (for through costimulation, resulting in the production of IFN-g and
example, IL-1b, IL-6, IL-10 and VEGF), the IMiDs may disrupt suppression of IL-10.131,132 The first study to show protective anti-
protection by the microenvironment, sensitizing myeloma to tumor immunity by IMiDs in conjunction with whole-tumor cell
concurrent cytotoxic therapies.109 vaccination was demonstrated in a CT-26 colon mouse model.33
As conventional therapies are initially successful in debulking a More recently, Noonan et al.34 showed single-agent lenalidomide
high proportion of the myeloma burden, the bone marrow niche therapy in MM patients augmented endogenous MM-specific CD8
is postulated to rescue clonogenic myeloma stem cells ultimately T-cell responses.34 Both Th-17 and regulatory T-cells have a
responsible for disease relapse.110 Although the literature on the pathogenic role in MM, with higher T-regulatory cells conferring
identification of myeloma stem cells are somewhat nebulous and poorer outcomes in MM patients.133,134 The ability of IMiDs to
without consensus, studies have eluded to several populations of reduce Th-17 cells, and inhibit the generation and function of
myeloma cells that have clonogenic capacity. These include the Foxp3 T-regulatory cells may result in the mediation of T-cell
CD138  CD27 þ cells that are lenalidomide, dexamethasone, responses against MM.34,135
cyclophosphamide and bortezomib resistant with a high level of
hedgehog signaling.111,112 Furthermore, a small population of
CD34 þ CD45low myeloma cells has been shown to have CLINICAL TOXICITIES PROVIDE MECHANISTIC INSIGHT INTO
clonogenic properties in NOD/SCID mice.113 ‘Side-population IMID BIOLOGY
cells’ defined by the capacity to efflux Hoechst 3342 stain,114 The differing side effect profiles of thalidomide and its IMiD
have also been identified as a source of stem-cell like derivatives highlight important biological distinctions between
chemoresistant cancer-initiating cells. The proliferation and these closely related compounds not merely explained by issues
survival of MM side-population cells is augmented by bone of potency. Elucidation of the mechanisms responsible for
marrow stroma through pathways that are suppressible by divergent clinical toxicities could potentially inform the biology
thalidomide and lenalidomide.110 Lenalidomide also reduces the of tumor-specific responses to the respective analogues. The
clonogenic potential of myeloma side-population cells.110 This major cumulative dose-limiting toxicity of thalidomide is periph-
indicates that suppression of the myeloma ‘stem-cell’ may be an eral neuropathy136 with minimal bone marrow suppression.
important facet to IMiD activity, particularly in the prevention of Although lenalidomide is more potent than thalidomide (for
relapse from minimal residual disease states (for example, example, in TNFa suppression assays), it does not cause
following autologous stem-cell transplantation). neuropathy.89 Rather, lenalidomide suppresses normal hemato-
poiesis resulting in neutropenia and thrombocytopenia.
Pomalidomide also causes neutropenia, with a lower incidence
(c) Immunological disease control
of neuropathy than thalidomide.137 All of the IMiDs are
Hematological malignancies, particularly MM are characterized by prothrombotic, with prophylactic anticoagulant therapy often
progressive immune cell dysregulation, of which T- and NK-cell administered concurrent to the IMiDs to prevent venous
suppression is well described.115–117 Lenalidomide’s ability to thromboembolic disease. The greatest emerging concern with
potentiate T- and NK-cell activation37,38,40 has resulted in chronic lenalidomide exposure is a measurable increase in the
exploratory studies elucidating the role of IMiDs in the incidence of second primary malignancies.89–92
immunological control of MM. NK-cell interaction with target
cells involves a complex set of activating and inhibitory receptors.
Activating receptors involved in MM cell recognition and lysis are (a) Peripheral neuropathy
largely governed by NKG2D, with a lesser role for DNAM-1 and Neuropathy occurs in most patients receiving chronic thalidomide
NKp46.118 Interestingly, IMiDs do not appear to alter the therapy with an actuarial incidence of over 70% at 12 months.136
expression of these receptors to confer functional changes in The etiology of thalidomide-induced neuropathy is multifactorial,
NK-cell activity.40,118,119 IMiDs upregulate CD16 expression on NK as patients often have pre-existing nerve damage and many
cells to increase antibody-dependent cell cytotoxicity against receive concurrent neurotoxic therapies. In particular, the
MM.120,121 It is becoming evident that IMiDs are able to decrease combination of thalidomide with bortezomib is associated with
the activation threshold of NK-cell cytotoxicity through a high risk of nerve damage.138 However, the potent new-
modulation of inhibitory receptor interactions. MM-induced generation proteasome inhibitor carfilzomib is not neuropathic,
expression of the programmed cell death receptor 1 inhibits NK- indicating that the neurotoxicity of bortezomib may be ‘off target’
cell cytotoxicity.122 Treatment with lenalidomide reduces inhibition of other serine proteases.139 HtrA2/Omi was identified
programmed cell death receptor 1 ligand on MM cells and thus as a non-proteasomal bortezomib target that is spared by
sensitizes MM cells to lysis by autologous NK cells.122 Killer-cell carfilzomib and protects neurons from oxidative stress.139 Thus,
Immunoglobulin-like receptors (KIR) are well described as the thalidomide-induced free-radical generation57 and bortezomib-
major functional inhibitory receptors on NK cells.123 As MM cells induced depletion of neuroprotective serine proteases (for
express high levels of the KIR ligand, HLA class I, it is likely that this example, HtrA2/Omi) could combine to cause the mitochondrial
interaction contributes to NK-cell dysfunction in vivo.124,125 IMiDs toxicity observed in neuropathic states. Indeed, genomic profiling
are able to reduce the expression of KIR on NK cells.119 identified a single-nucleotide polymorphism of glutathione
Furthermore, a recent study demonstrated the therapeutic S-transferase theta-1 (GSTT1) as predictive of neuropathy in a
efficacy of combination IMiD and anti-KIR blocking therapy in a cohort of myeloma patients receiving thalidomide.140 As GSTT1 is
mouse model of MM.125 a cellular regulator of reactive oxygen species, this further

Oncogene (2013) 4191 – 4202 & 2013 Macmillan Publishers Limited


Molecular IMiD targets
J Shortt et al
4199
implicated thalidomide-induced oxidative stress in the etiology of CONCLUSIONS
nerve damage. Despite their simple chemical structures, thalidomide and its
analogues exhibit pleotropic immunomodulatory and anti-neo-
plastic effects. This conveys remarkable clinical efficacy in clearly
(b) Hematological toxicities defined disease entities, specifically granulomatous inflammation,
Neutropenia and the risk of sepsis is a major dose-limiting toxicity mature B-cell neoplasms and MDS with del 5q. The molecular
of lenalidomide. Lenalidomide treatment is also associated with mechanisms of IMiD activity are likely to be both divergent and
impaired stem-cell mobilization for autologous CD34 þ collection complementary across these distinct diseases. In particular the
before transplantation in MM.141 This is despite the observation combined ability to modulate multiple targets from the level of
that IMiDs induce the expansion of CD34 þ bone marrow the epigenome to post-translational protein modulations and
progenitors in vitro.142 However, the transcriptional changes that systemic cytokine networks highlights the complexity of IMiD
evoke CD34 þ proliferation in vitro are also associated with a biology. Unfortunately, the beneficial properties of IMiDs may be
differentiation block within the granulocytic lineage.143 This inseparable from serious clinical toxicities including teratogenicity,
apparent ‘maturation arrest’ is associated with downregulation secondary malignancy, myelosuppression and nerve-damage,
of the transcription factor PU.1, and results in a relative excess ostensibly because they are mediated by shared molecular
of promyelocytes. Promyelocytes are rich in prothrombotic targets. It is hoped that the next chapter in the ‘thalidomide
mediators including cathepsin G, a platelet agonist implicated in saga’1 will be the identification of new targets that can be
the pathogenesis of disseminated intravascular coagulation therapeutically manipulated by novel non-IMiD pharmaceuticals
complicating acute promyelocytic leukemia.144 Therefore, to derive clinical benefit without toxicity.
IMiD-induced changes in granulocyte maturation may directly
contribute to an increased risk of venous thrombosis.143 Other
well-described IMiD activities are pro-coagulant. Suppression of CONFLICT OF INTEREST
COX-2 has been clearly implicated in adverse thrombotic events in The authors declare no conflict of interest.
patients treated for non-malignant inflammatory conditions.26,145
On-target suppression of angiogenesis is also pro-thrombotic, as
endothelial cells that become stressed in response to VEGF REFERENCES
withdrawal release a range of pro-coagulant mediators.44,146 A
1 Mellin GW, Katzenstein M. The saga of thalidomide. Neuropathy to embryopathy,
targeted genetic screen of myeloma patients identified single- with case reports of congenital anomalies. N Engl J Med 1962; 267: 1184–1192.
nucleotide polymorphisms associated with thalidomide- 2 McBride WG. Thalidomide and congenital abnormalities. Lancet 1961; 278: 1358.
associated thrombosis in genes and pathways important in drug 3 Lenz W, Pfeiffer RA, Kosenow W, Hayman DJ. Thalidomide and congenital
metabolism, DNA-repair and cytokine regulation.146 The authors abnormalities. Lancet 1962; 279: 45–46.
suggested that susceptibility to IMiD-associated thrombosis 4 Iyer CG, Languillon J, Ramanujam K, Tarabini-Castellani G, De las Aguas JT,
was secondary to rapid dissolution of myeloma in the context Bechelli LM et al. WHO co-ordinated short-term double-blind trial with thalido-
of apoptosis induction by concurrent DNA-damaging stimuli. mide in the treatment of acute lepra reactions in male lepromatous patients. Bull
The thrombogenicity of IMiDs is therefore multifactorial and World Health Organ 1971; 45: 719–732.
5 Sheskin J. Thalidomide in the treatment of lepra reactions. Clin Pharmacol Ther
contributed to by underlying malignancy and associated
1965; 6: 303–306.
treatments. 6 Wohl DA, Aweeka FT, Schmitz J, Pomerantz R, Cherng DW, Spritzler J et al. Safety,
tolerability, and pharmacokinetic effects of thalidomide in patients infected with
human immunodeficiency virus: AIDS Clinical Trials Group 267. J Infect Dis 2002;
(c) Lenalidomide and second primary malignancies 185: 1359–1363.
With the benefit of longer myeloma-related progression free 7 Jacobson JM, Greenspan JS, Spritzler J, Ketter N, Fahey JL, Jackson JB et al.
survival, patients receiving chronic lenalidomide maintenance Thalidomide for the treatment of oral aphthous ulcers in patients with human
therapy have an increased risk of both hematopoietic and solid immunodeficiency virus infection. National Institute of Allergy and Infectious
organ cancers.89 Several major clinical studies identified an Diseases AIDS Clinical Trials Group. N Engl J Med 1997; 336: 1487–1493.
8 Hamuryudan V, Mat C, Saip S, Ozyazgan Y, Siva A, Yurdakul S et al. Thalidomide
increased incidence of second primary malignancies in MM
in the treatment of the mucocutaneous lesions of the Behcet syndrome. A
patients receiving chronic ‘maintenance’ lenalidomide.90–92 The randomized, double-blind, placebo-controlled trial. Ann Intern Med 1998; 128:
incidence of second cancers in these studies was as high as 7% 443–450.
with a hazard ratio close to double that of placebo-treated 9 Ehrenpreis ED, Kane SV, Cohen LB, Cohen RD, Hanauer SB. Thalidomide therapy
controls. Elucidation of the precise molecular oncogenic for patients with refractory Crohn’s disease: an open-label trial. Gastroenterology
mechanisms is confounded by prior or concurrent exposure to 1999; 117: 1271–1277.
genotoxic agents, in particular the alkylator melphalan. Second 10 Cortes-Hernandez J, Torres-Salido M, Castro-Marrero J, Vilardell-Tarres M, Ordi-
primary cancers have not yet been associated with other IMiDs, Ros J. Thalidomide in the treatment of refractory cutaneous lupus erythemato-
and lenalidomide does not appear to accelerate the progression sus: prognostic factors of clinical outcome. Br J Dermatol 2012; 166: 616–623.
11 Bartlett JB, Dredge K, Dalgleish AG. The evolution of thalidomide and its IMiD
of MDS with del 5q to acute leukemia.89 However, as the ‘danger
derivatives as anticancer agents. Nat Rev Cancer 2004; 4: 314–322.
signal’ from these large lenalidomide trials has only recently 12 Corral LG, Haslett PA, Muller GW, Chen R, Wong LM, Ocampo CJ et al. Differential
emerged, mechanistic insights remain speculative. Lenalidomide cytokine modulation and T cell activation by two distinct classes of thalidomide
is not considered directly mutagenic, but could plausibly synergize analogues that are potent inhibitors of TNF-alpha. J Immunol 1999; 163:
with DNA-damaging cytotoxics in oncogenesis. For example, 380–386.
lenalidomide destabilizes p53,88 an important tumor suppressor in 13 Kenyon BM, Browne F, D’Amato RJ. Effects of thalidomide and related meta-
the context of DNA-damage. This might also explain the lack bolites in a mouse corneal model of neovascularization. Exp Eye Res 1997; 64:
of an association between thalidomide and second cancers, 971–978.
as it is less potent in this regard. However, all of the IMiDs bind 14 Eriksson T, Bjorkman S, Roth B, Fyge A, Hoglund P. Stereospecific determination,
chiral inversion in vitro and pharmacokinetics in humans of the enantiomers of
cereblon and dysfunction of the cereblon/DDB-1 ubiquitin–ligase
thalidomide. Chirality 1995; 7: 44–52.
complex might also impair the DNA-damage response, as DDB-1 15 Lepper ER, Smith NF, Cox MC, Scripture CD, Figg WD. Thalidomide metabolism and
functions in nucleotide excision DNA-repair.147 Elucidation of the hydrolysis: mechanisms and implications. Curr Drug Metab. 2006; 7: 677–685.
capacity for IMiDs to potentiate mutagens requires urgent 16 Muller GW, Corral LG, Shire MG, Wang H, Moreira A, Kaplan G et al. Structural
scientific attention in order to inform the safe delivery of modifications of thalidomide produce analogs with enhanced tumor necrosis
IMiD/chemotherapy combinations in the clinic. factor inhibitory activity. J Med Chem 1996; 39: 3238–3240.

& 2013 Macmillan Publishers Limited Oncogene (2013) 4191 – 4202


Molecular IMiD targets
J Shortt et al
4200
17 Muller GW, Chen R, Huang SY, Corral LG, Wong LM, Patterson RT et al. Amino- T cells by increasing PKC-theta activation and enhancing the DNA-binding
substituted thalidomide analogs: potent inhibitors of TNF-alpha production. activity of AP-1 but not NF-kappaB, OCT-1, or NF-AT. J Interferon Cytokine Res
Bioorg Med Chem Lett 1999; 9: 1625–1630. 2005; 25: 604–616.
18 Barnes PF, Chatterjee D, Brennan PJ, Rea TH, Modlin RL. Tumor necrosis factor 40 Hsu AK, Quach H, Tai T, Prince HM, Harrison SJ, Trapani JA et al. The immu-
production in patients with leprosy. Infect Immun 1992; 60: 1441–1446. nostimulatory effect of lenalidomide on NK-cell function is profoundly inhibited
19 Tsimberidou AM, Waddelow T, Kantarjian HM, Albitar M, Giles FJ. Pilot study of by concurrent dexamethasone therapy. Blood 2011; 117: 1605–1613.
recombinant human soluble tumor necrosis factor (TNF) receptor (p75) fusion 41 Gandhi AK, Kang J, Capone L, Parton A, Wu L, Zhang LH et al. Dexamethasone
protein (TNFR:Fc; Enbrel) in patients with refractory multiple myeloma: increase synergizes with lenalidomide to inhibit multiple myeloma tumor growth, but
in plasma TNF alpha levels during treatment. Leuk Res 2003; 27: 375–380. reduces lenalidomide-induced immunomodulation of T and NK cell function.
20 Madan S, Lacy MQ, Dispenzieri A, Gertz MA, Buadi F, Hayman SR et al. Efficacy of Curr Cancer Drug Targets 2010; 10: 155–167.
retreatment with immunomodulatory drugs (IMiDs) in patients receiving 42 Hernandez-Ilizaliturri FJ, Reddy N, Holkova B, Ottman E, Czuczman MS. Immu-
IMiDs for initial therapy of newly diagnosed multiple myeloma. Blood 2011; 118: nomodulatory drug CC-5013 or CC-4047 and rituximab enhance antitumor
1763–1765. activity in a severe combined immunodeficient mouse lymphoma model. Clin
21 Tsenova L, Mangaliso B, Muller G, Chen Y, Freedman VH, Stirling D et al. Use of Cancer Res 2005; 11: 5984–5992.
IMiD3, a thalidomide analog, as an adjunct to therapy for experimental tuber- 43 D’Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is an inhibitor of
culous meningitis. Antimicrob Agents Chemother 2002; 46: 1887–1895. angiogenesis. Proc Natl Acad Sci USA 1994; 91: 4082–4085.
22 Giamarellos-Bourboulis EJ, Poulaki H, Kostomitsopoulos N, Dontas I, Perrea D, 44 Dredge K, Horsfall R, Robinson SP, Zhang LH, Lu L, Tang Y et al. Orally admi-
Karayannacos PE et al. Effective immunomodulatory treatment of Escherichia coli nistered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endo-
experimental sepsis with thalidomide. Antimicrob Agents Chemother 2003; 47: thelial cell migration and Akt phosphorylation in vitro. Microvasc Res 2005; 69:
2445–2449. 56–63.
23 Sampaio EP, Sarno EN, Galilly R, Cohn ZA, Kaplan G. Thalidomide selectively 45 Bauer KS, Dixon SC, Figg WD. Inhibition of angiogenesis by thalidomide requires
inhibits tumor necrosis factor alpha production by stimulated human mono- metabolic activation, which is species-dependent. Biochem Pharmacol 1998; 55:
cytes. J Exp Med 1991; 173: 699–703. 1827–1834.
24 Moreira AL, Sampaio EP, Zmuidzinas A, Frindt P, Smith KA, Kaplan G. Thalido- 46 Yaccoby S, Johnson CL, Mahaffey SC, Wezeman MJ, Barlogie B, Epstein J.
mide exerts its inhibitory action on tumor necrosis factor alpha by enhancing Antimyeloma efficacy of thalidomide in the SCID-hu model. Blood 2002; 100:
mRNA degradation. J Exp Med 1993; 177: 1675–1680. 4162–4168.
25 Li S, Pal R, Monaghan SA, Schafer P, Ouyang H, Mapara M et al. IMiD immuno- 47 Lu L, Payvandi F, Wu L, Zhang LH, Hariri RJ, Man HW et al. The anti-cancer drug
modulatory compounds block C/EBP{beta} translation through eIF4E down- lenalidomide inhibits angiogenesis and metastasis via multiple inhibitory effects
regulation resulting in inhibition of MM. Blood 2011; 117: 5157–5165. on endothelial cell function in normoxic and hypoxic conditions. Microvasc Res
26 Ferguson GD, Jensen-Pergakes K, Wilkey C, Jhaveri U, Richard N, Verhelle D et al. 2009; 77: 78–86.
Immunomodulatory drug CC-4047 is a cell-type and stimulus-selective tran- 48 Gupta D, Treon SP, Shima Y, Hideshima T, Podar K, Tai YT et al. Adherence of
scriptional inhibitor of cyclooxygenase 2. J Clin Immunol 2007; 27: 210–220. multiple myeloma cells to bone marrow stromal cells upregulates vascular
27 Welm AL, Mackey SL, Timchenko LT, Darlington GJ, Timchenko NA.. Translational endothelial growth factor secretion: therapeutic applications. Leukemia 2001; 15:
induction of liver-enriched transcriptional inhibitory protein during acute phase 1950–1961.
response leads to repression of CCAAT/enhancer binding protein alpha mRNA. 49 Yabu T, Tomimoto H, Taguchi Y, Yamaoka S, Igarashi Y, Okazaki T. Thalidomide-
J Biol Chem 2000; 275: 27406–27413. induced antiangiogenic action is mediated by ceramide through depletion of
28 Masferrer JL, Leahy KM, Koki AT, Zweifel BS, Settle SL, Woerner BM et al. Anti- VEGF receptors, and is antagonized by sphingosine-1-phosphate. Blood 2005;
angiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res 106: 125–134.
2000; 60: 1306–1311. 50 Ito T, Ando H, Handa H. Teratogenic effects of thalidomide: molecular
29 Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB et al. The mechanisms. Cell Mol Life Sci 2011; 68: 1569–1579.
effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous 51 Therapontos C, Erskine L, Gardner ER, Figg WD, Vargesson N. Thalidomide
polyposis. N Engl J Med 2000; 342: 1946–1952. induces limb defects by preventing angiogenic outgrowth during early limb
30 Groen HJ, Sietsma H, Vincent A, Hochstenbag MM, van Putten JW, van den Berg formation. Proc Natl Acad Sci USA 2009; 106: 8573–8578.
A et al. Randomized, placebo-controlled phase III study of docetaxel plus car- 52 Dredge K, Marriott JB, Macdonald CD, Man HW, Chen R, Muller GW et al. Novel
boplatin with celecoxib and cyclooxygenase-2 expression as a biomarker for thalidomide analogues display anti-angiogenic activity independently of
patients with advanced non-small-cell lung cancer: the NVALT-4 study. J Clin immunomodulatory effects. Br J Cancer 2002; 87: 1166–1172.
Oncol 2011; 29: 4320–4326. 53 Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y et al. Identification of a
31 Hoang B, Zhu L, Shi Y, Frost P, Yan H, Sharma S et al. Oncogenic RAS mutations in primary target of thalidomide teratogenicity. Science 2010; 327: 1345–1350.
myeloma cells selectively induce cox-2 expression, which participates in 54 Knobloch J, Shaughnessy Jr. JD, Ruther U. Thalidomide induces limb deformities by
enhanced adhesion to fibronectin and chemoresistance. Blood 2006; 107: perturbing the Bmp/Dkk1/Wnt signaling pathway. Faseb J 2007; 21: 1410–1421.
4484–4490. 55 Knobloch J, Schmitz I, Gotz K, Schulze-Osthoff K, Ruther U. Thalidomide induces
32 Prince HM, Mileshkin L, Roberts A, Ganju V, Underhill C, Catalano J et al. limb anomalies by PTEN stabilization, Akt suppression, and stimulation of cas-
A multicenter phase II trial of thalidomide and celecoxib for patients with pase-dependent cell death. Mol Cell Biol 2008; 28: 529–538.
relapsed and refractory multiple myeloma. Clin Cancer Res 2005; 11: 5504–5514. 56 Stephens TD, Bunde CJ, Fillmore BJ. Mechanism of action in thalidomide ter-
33 Dredge K, Marriott JB, Todryk SM, Muller GW, Chen R, Stirling DI et al. Protective atogenesis. Biochem Pharmacol 2000; 59: 1489–1499.
antitumor immunity induced by a costimulatory thalidomide analog in con- 57 Parman T, Wiley MJ, Wells PG. Free radical-mediated oxidative DNA damage in
junction with whole tumor cell vaccination is mediated by increased Th1-type the mechanism of thalidomide teratogenicity. Nat Med 1999; 5: 582–585.
immunity. J Immunol 2002; 168: 4914–4919. 58 Hansen JM, Gong SG, Philbert M, Harris C. Misregulation of gene expression in
34 Noonan K, Rudraraju L, Ferguson A, Emerling A, Pasetti MF, Huff CA et al. the redox-sensitive NF-kappab-dependent limb outgrowth pathway by thali-
Lenalidomide-induced immunomodulation in multiple myeloma: impact on domide. Dev Dyn 2002; 225: 186–194.
vaccines and antitumor responses. Clin Cancer Res 2012; 18: 1426–1434. 59 Xin W, Xiaohua N, Peilin C, Xin C, Yaqiong S, Qihan W. Primary function analysis
35 Haslett PA, Corral LG, Albert M, Kaplan G. Thalidomide costimulates primary of human mental retardation gene CRBN. Mol Biol Rep 2008; 35: 251–256.
human T lymphocytes, preferentially inducing proliferation, cytokine production, 60 Higgins JJ, Pucilowska J, Lombardi RQ, Rooney JP. A mutation in a novel ATP-
and cytotoxic responses in the CD8 þ subset. J Exp Med 1998; 187: 1885–1892. dependent Lon protease gene in a kindred with mild mental retardation. Neu-
36 LeBlanc R, Hideshima T, Catley LP, Shringarpure R, Burger R, Mitsiades N et al. rology 2004; 63: 1927–1931.
Immunomodulatory drug costimulates T cells via the B7-CD28 pathway. Blood 61 Hohberger B, Enz R. Cereblon is expressed in the retina and binds to voltage-
2004; 103: 1787–1790. gated chloride channels. FEBS Lett 2009; 583: 633–637.
37 Hayashi T, Hideshima T, Akiyama M, Podar K, Yasui H, Raje N et al. Molecular 62 Jo S, Lee KH, Song S, Jung YK, Park CS. Identification and functional character-
mechanisms whereby immunomodulatory drugs activate natural killer cells: ization of cereblon as a binding protein for large-conductance calcium-activated
clinical application. Br J Haematol 2005; 128: 192–203. potassium channel in rat brain. J Neurochem 2005; 94: 1212–1224.
38 Davies FE, Raje N, Hideshima T, Lentzsch S, Young G, Tai YT et al. Thalidomide 63 Lee KM, Jo S, Kim H, Lee J, Park CS. Functional modulation of AMP-activated
and immunomodulatory derivatives augment natural killer cell cytotoxicity in protein kinase by cereblon. Biochem Biophys Acta 2011; 1813: 448–455.
multiple myeloma. Blood 2001; 98: 210–216. 64 Figg WD, Raje S, Bauer KS, Tompkins A, Venzon D, Bergan R et al. Pharmacoki-
39 Payvandi F, Wu L, Naziruddin SD, Haley M, Parton A, Schafer PH et al. Immu- netics of thalidomide in elderly prostate cancer population. J Pharm Sci 1999;
nomodulatory drugs (IMiDs) increase the production of IL-2 from stimulated 88: 121–125.

Oncogene (2013) 4191 – 4202 & 2013 Macmillan Publishers Limited


Molecular IMiD targets
J Shortt et al
4201
65 Cheng J, Gu YJ, Wang Y, Cheng SH, Wong WT. Nanotherapeutics in angiogenesis: 89 Palumbo A, Freeman J, Weiss L, Fenaux P. The clinical safety of lenalidomide in
synthesis and in vivo assessment of drug efficacy and biocompatibility in zeb- multiple myeloma and myelodysplastic syndromes. Expert Opin Drug Saf 2012;
rafish embryos. Int J Nanomedicine 2011; 6: 2007–2021. 11: 107–120.
66 Ashby J, Tinwell H, Callander RD, Kimber I, Clay P, Galloway SM et al. Thalido- 90 Palumbo A, Hajek R, Delforge M, Kropff M, Petrucci MT, Catalano J et al. Con-
mide: lack of mutagenic activity across phyla and genetic endpoints. Mutat Res tinuous lenalidomide treatment for newly diagnosed multiple myeloma. N Engl J
1997; 396: 45–64. Med 2012; 366: 1759–1769.
67 Huang PH, McBride WG. Interaction of [glutarimide-2-14C]-thalidomide with rat 91 Attal M, Lauwers-Cances V, Marit G, Caillot D, Moreau P, Facon T et al. Lenali-
embryonic DNA in vivo. Teratog Carcinog Mutagen 1997; 17: 1–5. domide maintenance after stem-cell transplantation for multiple myeloma.
68 Jonsson NA. Chemical structure and teratogenic properties. IV. An outline of a N Engl J Med 2012; 366: 1782–1791.
chemical hypothesis for the teratogenic action of thalidomide. Acta Pharm Suec 92 McCarthy PL, Owzar K, Hofmeister CC, Hurd DD, Hassoun H, Richardson PG et al.
1972; 9: 543–562. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med
69 Jackson SP, MacDonald JJ, Lees-Miller S, Tjian R. GC box binding induces 2012; 366: 1770–1781.
phosphorylation of Sp1 by a DNA-dependent protein kinase. Cell 1990; 63: 93 Kyle RA, Rajkumar SV. Multiple myeloma. Blood 2008; 111: 2962–2972.
155–165. 94 Chapman MA, Lawrence MS, Keats JJ, Cibulskis K, Sougnez C, Schinzel AC et al.
70 Narayan VA, Kriwacki RW, Caradonna JP. Structures of zinc finger domains from Initial genome sequencing and analysis of multiple myeloma. Nature 2011; 471:
transcription factor Sp1. Insights into sequence-specific protein-DNA recogni- 467–472.
tion. J Biol Chem 1997; 272: 7801–7809. 95 Yang Y, Shaffer 3rd AL, Emre NC, Ceribelli M, Zhang M, Wright G et al. Exploiting
71 Gandhi AK, Kang J, Naziruddin S, Parton A, Schafer PH, Stirling DI. Lenalidomide synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer
inhibits proliferation of Namalwa CSN.70 cells and interferes with Gab1 phos- Cell 2012; 21: 723–737.
phorylation and adaptor protein complex assembly. Leuk Res 2006; 30: 849–858. 96 Shaffer AL, Emre NC, Lamy L, Ngo VN, Wright G, Xiao W et al. IRF4 addiction in
72 Escoubet-Lozach L, Lin I-L, Jensen-Pergakes K, Brady HA, Gandhi AK, Schafer PH multiple myeloma. Nature 2008; 454: 226–231.
et al. Pomalidomide and lenalidomide induce p21WAF-1 expression in both 97 Ngo VN, Young RM, Schmitz R, Jhavar S, Xiao W, Lim KH et al. Oncogenically
lymphoma and multiple myeloma through a LSD1-mediated epigenetic active MYD88 mutations in human lymphoma. Nature 2011; 470: 115–119.
mechanism. Cancer Res 2009; 69: 7347–7356. 98 Mitsiades N, Mitsiades CS, Poulaki V, Chauhan D, Richardson PG, Hideshima T
73 Gorgun G, Calabrese E, Soydan E, Hideshima T, Perrone G, Bandi M et al. et al. Apoptotic signaling by immunomodulatory thalidomide analogs
Immunomodulatory effects of lenalidomide and pomalidomide on interaction of in human multiple myeloma cells: therapeutic implications. Blood 2002; 99:
tumor and bone marrow accessory cells in multiple myeloma. Blood 2010; 116: 4525–4530.
3227–3237. 99 Lopez-Girona A, Mendy D, Ito T, Miller K, Gandhi AK, Kang J et al. Cereblon is a
74 Croker BA, Kiu H, Nicholson SE. SOCS regulation of the JAK/STAT signalling direct protein target for immunomodulatory and antiproliferative activities of
pathway. Semin Cell Dev Biol 2008; 19: 414–422. lenalidomide and pomalidomide. Leukemia 2012; 26: 2326–2335.
75 Depil S, Saudemont A, Quesnel B. SOCS-1 gene methylation is frequent but does 100 Zhu YX, Braggio E, Shi CX, Bruins LA, Schmidt JE, Van Wier S et al. Cereblon
not appear to have prognostic value in patients with multiple myeloma. Leu- expression is required for the antimyeloma activity of lenalidomide and
kemia 2003; 17: 1678–1679. pomalidomide. Blood 2011; 118: 4771–4779.
76 Van den Berghe H, Cassiman JJ, David G, Fryns JP, Michaux JL, Sokal G. Distinct 101 Nefedova Y, Landowski TH, Dalton WS. Bone marrow stromal-derived soluble
haematological disorder with deletion of long arm of no. 5 chromosome. Nature factors and direct cell contact contribute to de novo drug resistance of myeloma
1974; 251: 437–438. cells by distinct mechanisms. Leukemia 2003; 17: 1175–1182.
77 List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E et al. Lenalidomide 102 Mitsiades CS, Mitsiades NS, Richardson PG, Munshi NC, Anderson KC. Multiple
in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med myeloma: a prototypic disease model for the characterization and therapeutic
2006; 355: 1456–1465. targeting of interactions between tumor cells and their local microenvironment.
78 Boultwood J, Fidler C, Strickson AJ, Watkins F, Gama S, Kearney L et al. Narrowing J Cell Biochem 2007; 101: 950–968.
and genomic annotation of the commonly deleted region of the 5q- syndrome. 103 Cheung WC, Van Ness B. The bone marrow stromal microenvironment influ-
Blood 2002; 99: 4638–4641. ences myeloma therapeutic response in vitro. Leukemia 2001; 15: 264–271.
79 Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N et al. Identification of 104 Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion medi-
RPS14 as a 5q- syndrome gene by RNA interference screen. Nature 2008; 451: ated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in
335–339. human myeloma cell lines. Blood 1999; 93: 1658–1667.
80 Draptchinskaia N, Gustavsson P, Andersson B, Pettersson M, Willig TN, Dianzani I 105 McMillin DW, Delmore J, Weisberg E, Negri JM, Geer DC, Klippel S et al. Tumor
et al. The gene encoding ribosomal protein S19 is mutated in Diamond-Blackfan cell-specific bioluminescence platform to identify stroma-induced changes to
anaemia. Nat Genet 1999; 21: 169–175. anticancer drug activity. Nat Med 2010; 16: 483–489.
81 Gazda HT, Grabowska A, Merida-Long LB, Latawiec E, Schneider HE, Lipton JM 106 Shi Y, Frost P, Hoang B, Benavides A, Gera J, Lichtenstein A. IL-6-induced
et al. Ribosomal protein S24 gene is mutated in Diamond-Blackfan anemia. Am J enhancement of c-Myc translation in multiple myeloma cells: critical role of
Hum Genet 2006; 79: 1110–1118. cytoplasmic localization of the RNA-binding protein hnRNP A1. J Biol Chem 2011;
82 Barlow JL, Drynan LF, Hewett DR, Holmes LR, Lorenzo-Abalde S, Lane AL et al. A 286: 67–78.
p53-dependent mechanism underlies macrocytic anemia in a mouse model of 107 Landowski TH, Megli CJ, Nullmeyer KD, Lynch RM, Dorr RT. Mitochondrial-
human 5q- syndrome. Nat Med 2010; 16: 59–66. mediated disregulation of Ca2 þ is a critical determinant of Velcade
83 Dutt S, Narla A, Lin K, Mullally A, Abayasekara N, Megerdichian C et al. Hap- (PS-341/bortezomib) cytotoxicity in myeloma cell lines. Cancer Res 2005; 65:
loinsufficiency for ribosomal protein genes causes selective activation of p53 in 3828–3836.
human erythroid progenitor cells. Blood 2011; 117: 2567–2576. 108 Mitsiades CS, Mitsiades N, Poulaki V, Schlossman R, Akiyama M, Chauhan D et al.
84 Kumar MS, Narla A, Nonami A, Mullally A, Dimitrova N, Ball B et al. Coordinate Activation of NF-kappaB and upregulation of intracellular anti-apoptotic proteins
loss of a microRNA and protein-coding gene cooperate in the pathogenesis of via the IGF-1/Akt signaling in human multiple myeloma cells: therapeutic
5q- syndrome. Blood 2011; 118: 4666–4673. implications. Oncogene 2002; 21: 5673–5683.
85 Pellagatti A, Jadersten M, Forsblom A-M, Cattan H, Christensson B, Emanuelsson 109 Hideshima T, Chauhan D, Shima Y, Raje N, Davies FE, Tai Y-T et al. Thalidomide
EK et al. Lenalidomide inhibits the malignant clone and up-regulates the SPARC and its analogs overcome drug resistance of human multiple myeloma cells to
gene mapping to the commonly deleted region in 5q- syndrome patients. Proc conventional therapy. Blood 2000; 96: 2943–2950.
Natl Acad Sci USA 2007; 104: 11406–11411. 110 Jakubikova J, Adamia S, Kost-Alimova M, Klippel S, Cervi D, Daley JF et al.
86 Wei S, Chen X, Rocha K, Epling-Burnette PK, Djeu JY, Liu Q et al. A critical role for Lenalidomide targets clonogenic side population in multiple myeloma: patho-
phosphatase deficiency in the selective suppression of deletion 5q MDS by physiologic and clinical implications. Blood 2011; 117: 4409–4419.
lenalidomide. Proc Natl Acad Sci USA 2009; 106: 12974–12979. 111 Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, Borrello I et al. Clonogenic
87 Jadersten M, Saft L, Pellagatti A, Gohring G, Wainscoat JS, Boultwood J et al. multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer
Clonal heterogeneity in the 5q- syndrome: p53 expressing progenitors prevail Res. 2008; 68: 190–197.
during lenalidomide treatment and expand at disease progression. Haemato- 112 Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, Kim J et al.
logica 2009; 91: 1762–1766. Hedgehog signaling maintains a tumor stem cell compartment in multiple
88 Wei S, Chen X, McGraw K, Zhang L, Komrokji R, Clark J et al. Lenalidomide myeloma. Proc Natl Acad Sci USA 2007; 104: 4048–4053.
promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in 113 Pilarski LM, Belch AR. Clonotypic myeloma cells able to xenograft myeloma to
myelodysplastic syndrome with chromosome 5q deletion. Oncogene 2013; 32: nonobese diabetic severe combined immunodeficient mice copurify with CD34
1110–1120. ( þ ) hematopoietic progenitors. Clin Cancer Res 2002; 8: 3198–3204.

& 2013 Macmillan Publishers Limited Oncogene (2013) 4191 – 4202


Molecular IMiD targets
J Shortt et al
4202
114 Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional 131 Luptakova K, Rosenblatt J, Glotzbecker B, Mills H, Stroopinsky D, Kufe T et al.
properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Lenalidomide enhances anti-myeloma cellular immunity. Cancer Immunol
Med 1996; 183: 1797–1806. Immunother (e-pub ahead of print 24 June 2012).
115 Bernal M, Garrido P, Jimenez P, Carretero R, Almagro M, Lopez P et al. Changes in 132 Haslett PA, Hanekom WA, Muller G, Kaplan G. Thalidomide and a thalidomide
activatory and inhibitory natural killer (NK) receptors may induce progression to analogue drug costimulate virus-specific CD8 þ T cells in vitro. J Infect Dis 2003;
multiple myeloma: implications for tumor evasion of T and NK cells. Hum 187: 946–955.
Immunol 2009; 70: 854–857. 133 Giannopoulos K, Kaminska W, Hus I, Dmoszynska A. The frequency of T reg-
116 Jinushi M, Vanneman M, Munshi NC, Tai YT, Prabhala RH, Ritz J et al. MHC class I ulatory cells modulates the survival of multiple myeloma patients: detailed
chain-related protein A antibodies and shedding are associated with the pro- characterisation of immune status in multiple myeloma. Br J Cancer 2012; 106:
gression of multiple myeloma. Proc Natl Acad Sci USA 2008; 105: 1285–1290. 546–552.
117 Racanelli V, Leone P, Frassanito MA, Brunetti C, Perosa F, Ferrone S et al. 134 Noonan K, Marchionni L, Anderson J, Pardoll D, Roodman GD, Borrello I. A novel
Alterations in the antigen processing-presenting machinery of transformed role of IL-17 producing lymphocytes in mediating lytic bone disease in multiple
plasma cells are associated with reduced recognition by CD8 þ T cells and myeloma. Blood 2010; 116: 3554–3563.
characterize the progression of MGUS to multiple myeloma. Blood 2010; 115: 135 Galustian C, Meyer B, Labarthe M-C, Dredge K, Klaschka D, Henry J et al.
1185–1193. The anti-cancer agents lenalidomide and pomalidomide inhibit the prolife-
118 El-Sherbiny YM, Meade JL, Holmes TD, McGonagle D, Mackie SL, Morgan AW ration and function of T regulatory cells. Cancer Immunol Immunother 2009; 58:
et al. The requirement for DNAM-1, NKG2D, and NKp46 in the natural killer cell- 1033–1045.
mediated killing of myeloma cells. Cancer Res 2007; 67: 8444–8449. 136 Mileshkin L, Stark R, Day B, Seymour JF, Zeldis JB, Prince HM. Development of
119 Dauguet N, Fournie JJ, Poupot R, Poupot M. Lenalidomide down regulates the neuropathy in patients with myeloma treated with thalidomide: patterns of
production of interferon-gamma and the expression of inhibitory cytotoxic occurrence and the role of electrophysiologic monitoring. J Clin Oncol 2006; 24:
receptors of human Natural Killer cells. Cell Immunol 2010; 264: 163–170. 4507–4514.
120 Tai Y-T, Li X-F, Catley L, Coffey R, Breitkreutz I, Bae J et al. Immunomodulatory 137 Lacy MQ, Hayman SR, Gertz MA, Dispenzieri A, Buadi F, Kumar S et al. Pomali-
drug lenalidomide (CC-5013, IMiD3) augments anti-CD40 SGN-40-induced domide (CC4047) plus low-dose dexamethasone as therapy for relapsed multi-
cytotoxicity in human multiple myeloma: clinical implications. Cancer Res 2005; ple myeloma. J Clin Oncol 2009; 27: 5008–5014.
65: 11712–11720. 138 Chaudhry V, Cornblath DR, Polydefkis M, Ferguson A, Borrello I. Characteristics of
121 Tai YT, Horton HM, Kong SY, Pong E, Chen H, Cemerski S et al. Potent in vitro and bortezomib- and thalidomide-induced peripheral neuropathy. J Peripher Nerv
in vivo activity of an Fc-engineered humanized anti-HM1.24 antibody against Syst 2008; 13: 275–282.
multiple myeloma via augmented effector function. Blood 2012; 119: 2074–2082. 139 Arastu-Kapur S, Anderl JL, Kraus M, Parlati F, Shenk KD, Lee SJ et al. Non-
122 Benson Jr DM, Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B et al. The proteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a
PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: link to clinical adverse events. Clin Cancer Res 2011; 17: 2734–2743.
a therapeutic target for CT-011, a novel, monoclonal anti-PD-1 antibody. Blood 140 Cibeira MT, de Larrea CF, Navarro A, Diaz T, Fuster D, Tovar N et al. Impact on
2010; 116: 2286–2294. response and survival of DNA repair single nucleotide polymorphisms in
123 Lanier LL. Up on the tightrope: natural killer cell activation and inhibition. Nat relapsed or refractory multiple myeloma patients treated with thalidomide. Leuk
Immunol 2008; 9: 495–502. Res 2011; 35: 1178–1183.
124 Bernal M, Garrido P, Jiménez P, Carretero R, Almagro M, López P et al. Changes in 141 Kumar S, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, Gastineau DA et al.
activatory and inhibitory natural killer (NK) receptors may induce progression to Impact of lenalidomide therapy on stem cell mobilization and engraftment post-
multiple myeloma: implications for tumor evasion of T and NK cells. Hum peripheral blood stem cell transplantation in patients with newly diagnosed
Immunol 2009; 70: 854–857. myeloma. Leukemia 2007; 21: 2035–2042.
125 Benson Jr. DM, Bakan CE, Zhang S, Collins SM, Liang J, Srivastava S et al. IPH2101, 142 Verhelle D, Corral LG, Wong K, Mueller JH, Moutouh-de Parseval L, Jensen-
a novel anti-inhibitory KIR antibody, and lenalidomide combine to enhance the Pergakes K et al. Lenalidomide and CC-4047 inhibit the proliferation of malig-
natural killer cell versus multiple myeloma effect. Blood 2011; 118: 6387–6391. nant B cells while expanding normal CD34 þ progenitor cells. Cancer Res 2007;
126 Cook G, Campbell JD, Carr CE, Boyd KS, Franklin IM. Transforming growth factor 67: 746–755.
beta from multiple myeloma cells inhibits proliferation and IL-2 responsiveness 143 Pal R, Monaghan SA, Hassett AC, Mapara MY, Schafer P, Roodman GD et al.
in T lymphocytes. J Leukoc Biol 1999; 66: 981–988. Immunomodulatory derivatives induce PU.1 down-regulation, myeloid matura-
127 Frassanito MA, Cusmai A, Dammacco F. Deregulated cytokine network and tion arrest, and neutropenia. Blood 2010; 115: 605–614.
defective Th1 immune response in multiple myeloma. Clin Exp Immunol 2001; 144 Hirata RK, Chen ST, Weil SC. Expression of granule protein mRNAs in acute
125: 190–197. promyelocytic leukemia. Hematol Pathol 1993; 7: 225–238.
128 Sze DM, Giesajtis G, Brown RD, Raitakari M, Gibson J, Ho J et al. Clonal cytotoxic 145 Solomon DH, Schneeweiss S, Glynn RJ, Kiyota Y, Levin R, Mogun H et al. Rela-
T cells are expanded in myeloma and reside in the CD8( þ )CD57( þ )CD28(  ) tionship between selective cyclooxygenase-2 inhibitors and acute myocardial
compartment. Blood 2001; 98: 2817–2827. infarction in older adults. Circulation 2004; 109: 2068–2073.
129 Raitakari M, Brown RD, Sze D, Yuen E, Barrow L, Nelson M et al. T-cell expansions 146 Johnson DC, Corthals S, Ramos C, Hoering A, Cocks K, Dickens NJ et al. Genetic
in patients with multiple myeloma have a phenotype of cytotoxic T cells. Br J associations with thalidomide mediated venous thrombotic events in myeloma
Haematol 2000; 110: 203–209. identified using targeted genotyping. Blood 2008; 112: 4924–4934.
130 Schafer PH, Gandhi AK, Loveland MA, Chen RS, Man H-W, Schnetkamp PPM et al. 147 Wittschieben BO, Iwai S, Wood RD. DDB1-DDB2 (xeroderma pigmentosum group
Enhancement of cytokine production and AP-1 transcriptional activity in T cells E) protein complex recognizes a cyclobutane pyrimidine dimer, mismatches,
by thalidomide-related immunomodulatory drugs. J Pharmacol Exp Ther 2003; apurinic/apyrimidinic sites, and compound lesions in DNA. J Biol Chem 2005;
305: 1222–1232. 280: 39982–39989.

Oncogene (2013) 4191 – 4202 & 2013 Macmillan Publishers Limited

You might also like