ArticlePDF Available

Immunomodulatory drugs and their therapeutic effect in hematological malignancies through cereblon

Authors:
ISSN: 2398-8495
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
Review Article
Hematology & Medical Oncology
Volume 2(3): 1-8
Immunomodulatory drugs and their therapeutic eect in
hematological malignancies through cereblon
Ota Fuchs1*
1Institute of Hematology and Blood Transfusion, Prague, Czech Republic
Abstract
Immunomodulatory drugs (IMiDs), today also known as cereblon (CRBN) binding drugs, are therapeutically important anti-cancer and anti-inammatory drugs.
IMiDs are analogs of their prototype compound thalidomide. IMiDs have immune-modulation, anti-angiogenic, anti-inammatory and anti-proliferative eects.
CRBN is a component and substrate receptor of the Cullin 4 Ring E3 Ubiquitin Protein Ligase complex (CRL4). CRL4 consists of Cullin 4, RING nger protein
(Roc1), and DNA damage binding protein 1 (DDB1). CRBN binds to its substrate proteins and it leads to ubiquitination of these substrates by the CRL4. CRBN
is also involved in IMiDs-mediated T-cell co-stimulation and cytokine production. CRBN is a primary target of thalidomide teratogenicity. e binding of IMiDs
to CRBN is associated with cytotoxicity of IMiDs and is used to treat multiple myeloma (MM), myelodysplastic syndromes (MDS), lymphomas and chronic
lymphocytic leukemia. CRBN is composed of an N-terminal ATP-dependent serine protease Lon-like domain, which links to the E3 ubiquitin protein ligase
complex CRL4, and a C-terminal domain, which binds IMiDs. CRBN binding is mediated by a glutarimide ring in thalidomide, lenalidomide, pomalidomide,
CC-122, CC-220, CC-885 and CC-90009. Development of eector molecules mediating targeted ubiquitination of disease related proteins through cereblon is a
new important way in pharmacology.
Correspondence to: Ota Fuchs, Institute of Hematology and Blood Transfusion,
Prague, Czech Republic, Tel: +420 221 977 313; E-mail: Ota.Fuchs@uhkt.cz
Key words: immunomodulatory drugs, cereblon, cullin 4 E3 ubiquitin protein
ligase, Ikaros transcription factors famil, casein kinase 1A1, translation termination
factor GSPT1, ubiquitination, proteasome
Received: May 13, 2017; Accepted: May 29, 2017; Published: May 31, 2017
Introduction
IMiDs include thalidomide, lenalidomide, pomalidomide
(Figure 1), CC-122, CC-220, CC-885 and CC-90009 till now.
Both, lenalidomide (Revlimid®, initially known as CC-5013), and
pomalidomide (Pomalyst®, initially known as CC-4047, Actimid) are a
synthetic derivative of thalidomide (alomid®, Inmunoprin, Talidex,
Talizer).
Thalidomide [(RS)-2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-
1,3(2H)-dione] was synthesized in Germany, in 1954, from
α-phtaloylisoglutamine, to be used as a non-barbiturate sedative and
antimetic drug (Contergan). In 1957, aer a short period of preclinical
studies, thalidomide was approved for rst trimester gestational sickness
in humans. e appearance of malformations such as phocomelia in the
newborn banned its use three years later. alidomide was responsible
for birth defects in more than 10,000 children [1,2]. e US Food and
Drug Administration (FDA) approved thalidomide in 1998 for the
treatment of erythema nodosum leprosum [3]. alidomide exhibits
potent antiangiogenic and immunomodulatory eects, and is currently
used around the world to treat a range of conditions, mainly multiple
myeloma. However, long-term use of thalidomide has detrimental side
eects, such as peripheral neuropathy [4,5].
Lenalidomide was developed in order to avoid thalidomide side
eects (sedation and neuropathy), and to increase ecacy [6,7].
Lenalidomide shares a number of structural and biological properties
with thalidomide but is safer and more potent than thalidomide
Lenalidomide [3-(4-amino-1-oxo1,3-dihydro-2H-isoindol-2-yl)
piperidine-2,6-dione] is 4-amino-glutarimide analog of thalidomide
with potent immunomodulatory, antiangiogenic and direct neoplastic
cell inhibitory activity [6-26].
Pomalidomide [4-Amino-2-(2,6-dioxopiperidin-3-yl) isoindole-
1,3-dione] is a potent second-generation IMiD [27-34]. Pomalidomide
has direct antiproliferative, pro-apoptotic, and antiangiogenic eects,
as well as modulatory eects on bone resorption and on the immune
system. Chemical structure of the IMiD drugs is shown in Figure 1.
CC-122 hydrochloride is a novel immunomodulatory agent-like
thalidomide analog which directly binds to CRBN and promotes
ubiquitination and degradation of zinc nger transcription factors
Figure 1. Chemical structures of immunomodulatory drugs (IMiDs) including thalidomide,
lenalidomide and pomalidomide. Lenalidomide and pomalidomide are synthetic compounds
derived by modifying the chemical structure of thalidomide.
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 2-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
Aiolos and Ikaros in difuse large B cell-lymphoma (DLBCL) [35-39].
CC-220 is a further IMiD that binds to CRBN and is currently in
phase 1b/2aclinical trials for the treatment of relapsed and refractory
multiple myeloma (MM) and in phase 2 clinical trials for systemic
lupus erythematosus (SLE) [40]. CC-220 binds tighter to the CRBN
in the complex of CRL4 E3 ubiquitin protein ligase than lenalidomide
and causes more potent degradation of transcription factors Aiolos
and Ikaros. CC-885 binds also to CRL4CRBN but mediates not only
degradation of Aiolos but also specically binds to the eukaryotic
translation termination factor 3a (eRF3a), also known as GSPT1 (G1 to
S phase transition 1) and mediates degradation of this protein [41,42].
GSPT1 together with the eukaryotic translation termination factor 1
(eRF1) functions in stop codon recognition and nascent protein release
from ribosome in the process of mRNA translation. Lenalidomide
and pomalidomide do not mediate the degradation of GSPT1. CC-
90009 is a further IMiD that binds the E3 protein ubiquitin ligase
CRL4CRBN and mediates degradation of certain lymphoid transcription
factors, including Ikaros (IKZF1) and Aiolos (IKZF3), which are
transcriptional repressors in T-cells. is reduces the levels of these
transcription factors, and modulates the activity of the immune system,
which may include the activation of T-lymphocytes. In addition, this
downregulates the expression of other proteins, including interferon
regulatory factor 4 (IRF4) and c-myc, which plays a key role in the
proliferation of certain cancer cell types.CC-90009 is now in phase 1
clinical trials (NCT 02848001) in subjects with relapsed or refractory
acute myeloid leukemia (AML) in Canada and USA.
Mechanism of immunomodulatory drugs action in the
treatment of multiple myeloma
IMiDs target both MM cells and their microenvironment, while also
modulating the immune system. e exact molecular mechanisms of
the antitumor eects of IMiDs remain uncertain. IMiDs aect various
molecular and cellular elements within the tumor microenvironment.
IMiDS change the concentration of various cytokines that support
tumor cell growth [8-10,27,43,44]. IMiDs disrupt bone marrow stromal
support for malignant MM cells, although the exact mechanisms of
these actions remain unclear. IMiDs decrease the expression of adhesion
molecules that facilitate the interaction between MM cells and bone
marrow stromal cells (BMSCs) [45]. Importantly, the downregulation
of MM cells adherence to BMSCs can overcome the cellular adhesion-
mediated drug resistence by malignant MM cells. is eect of IMiDs
is further increased by their ability to downregulate TNFα [9,43,46,47].
Surprisingly, in contrast to the inhibitory eect of lenalidomide in
BMSCs, lenalidomide was totally ineective in inhibiting TNFα mRNA
expression in MM cells [44].
Immunomodulatory activities of IMiDs
IMiDs are a potent co-stimulator of primary human T cells,
synergizing with stimulation via T-cell receptor complex to increase
IL-2-mediated T-cell proliferation and interferon gamma (IFN-γ)
production [12-14,48-50]. Secretion of IL-2 and IFN-γ increases
the number of natural killer (NK) cells, improves their function and
mediates lysis of MM cells (Figure 2). NKT cells are a heterogeneous
Figure 2. Schematic diagram of ubiquitination and degradation of zinc nger lymphoid transcription factors Ikaros and Aiolos by CRL4cereblonE3 ubiquitin ligase and proteasomes. Binding
of cereblon (CRBN) by lenalidomide induces ubiquitination (marking IKZF1 /Ikaros/ and IKZF3 /Aiolos/) and degradation of both, Ikaros and Aiolos transcription factors. CRBN functions
as a substrate recognition component (substrate receptor) of this E3 ubiquitin ligase enzyme complex. CRL4cerebloncomplex consists of cullin 4A, RING nger protein regulator of cullins
(Roc1), and DNA damage binding protein 1 (DDB1).
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 3-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
group of T-cells that recognize lipids and glycolipids presented by
CD1d molecules. NKT cells activated in presence of lenalidomide,
have greater ability to secrete IFN-γ. Lenalidomide enhances antigen-
specic expansion of NKT cells [51]. Regulatory T-cells (Tregs) are a
component of the immune system that suppresses immune response of
other cells. Tregs were elevated in MM patients. IMiDs strongly inhibits
Tregs proliferation via decreased FOXP3 mRNA expression [52].
Anti-angiogenic properties of IMiDs
All IMiDs have anti-angiogenic activity. alidomide has
predominant anti-angiogenic activity while lenalidomide and
pomalidomide have far greater immune enhancing eects [43].
It appears that anti-angiogenesis occurs via the modulation of
chemotactic factors involved in endothelial cells migration including
TNFα, vascular endothelial growth factor (VEGF) and basic broblast
growth factor (bFGF) from BMSCs rather than a direct inhibition
of endothelial cells proliferation [53,54]. Inhibition of VEGF and
bFGF production by IMiDs is likely to have multiple other biological
eects beyond that of anti-angiogenesis, including inhibition of IL-6
production by BMSC. VEGF and bFGF up-regulate IL-6 and other
pro-inammatory cytokines [55].
Inhibition of cell cycle and induction of apoptosis by
IMiDs
IMiDs induce cell cycle arrest via upregulation of tumor suppressor
genes (cyclin dependent kinase inhibitors p15INK4b, p16INK4a, p21Cip1,Waf1,
p27Kip1; early response transcription factors / Erg1, Erg2 and Erg3/) and
apoptosis by caspase activation [56,57]. e activity of the transcription
factor NF-κB inhibition by IMiDs resulted in reduced expression of
anti-apoptotic proteins including cellular inhibitor of apoptosis protein
2 (cIAP2) [58] and FLIP [Fas-associated protein with death protein
(FADD)-like interleukin-1β-converting enzyme (FLICE) inhibitor
protein] [59]. ese anti-apoptotic proteins inhibit caspase-8, that is
on the other hand stimulated by IMiDs [57].
Disruption of bone marrow stromal support for malig-
nant MM cells
IMiDs down-regulate the expression of adhesion molecules
[leukocyte function-associated antigen 1 (LFA-1, CD11a), intercellular
adhesion molecule 1 (ICAM-1, CD54), vascular cell adhesion molecule
1 (VCAM-1, CD106) and very late antigen 4 (VLA-4)] that facilitate
the interaction between MM cells and BMSC. As we described, IMiDs
inhibit NF-κB, a transription factor that has important growth and
anti-apoptotic roles and which is connected with the upregulation of
intracellular adhesion molecules and many cytokines [60].
IMiDs eect on myeloma cell proliferation
e direct anti-MM eect of IMiDs was shown to occur through
the induction of a G1 phase of cell cycle growth arrest of MM cells
[54] and was associated with a decrease in interferon regulatory factor
4 (IRF4), a transcription factor that is critical for MM cell growth and
survival, (Figure 2), [61,62].
Raje, et al. [63] showed strong synergism of anti-MM activity of
rapamycin (Rapamune), a specic mTOR inhibitor, combined with
CC-5013. Importantly, this combination was able to overcome drug
resistance when tested against MM cell lines resistant to conventional
chemotherapy. Moreover, the combination, but not rapamycin alone,
was able to overcome the growth advantage conferred on MM cells by
interleukin-6 (IL-6), insulin-like growth factor-1 (IGF-1), or adherence
to bone marrow stromal cells (BMSCs). Combining rapamycin
and CC-5013 induced apoptosis of MM cells. Dierential signaling
cascades, including the mitogen-activated protein kinase (MAPK) and
the phosphatidylinositol 3’-kinase / Akt kinase (PI3K /Akt) pathways
[64-67], were targeted by these drugs individually and in combination,
suggesting the molecular mechanism by which they interfere with MM
growth and survival. ese studies, therefore, provide the framework
for clinical evaluation of mTOR inhibitors combined with IMiDs to
improve patient outcome in MM.
IMiDs down-regulate CCAAT/enhancer-binding protein-β (C/
EBPβ) resulting in abrogation of cell proliferation [68]. Overexpression
of C/EBPβ rescued MM cells from IMiD-induced inhibition
of proliferation, indicating that C/EBPβ is critical in mediating
antiproliferative eects. IMiD-induced decrease of C/EBPβ protein led
to impaired transcription of interferon regulatory factor 4 (IRF4).
Down-regulation of IRF4 by lenalidomide was conrmed by
longitudinal studies of bone marrow samples from 23 patients obtained
before and during lenalidomide treatment using CD138⁺/IRF4⁺ double
labeling. In contrast to down-regulation of C/EBPβ protein, IMiD
compounds did not alter C/EBPβ mRNA levels or protein stability,
suggesting translational regulation of C/EBPβ. We could demonstrate
that C/EBPβ protein expression is under eIF4E-translational control
in MM. Furthermore, inhibition of the eIF4E-C/EBPβ axis by IMiD
compounds was not observed in IMiD-resistant MM cells.
However, targeting translation at a dierent level by inhibiting
eukaryotic translation initiation factor 4E-binding protein 1
phosphorylation overcame resistance, suggesting that this pathway is
critical and might be a target to overcome drug resistance. MM cell lines
and primary MM cells strongly expressed C/EBPβ, whereas normal B
cells and plasma cells had little or no detectable levels of C/EBPβ [69].
Silencing of C/EBPβ led to down-regulation of transcription factors
such as IRF4, XBP1, and BLIMP1 accompanied by a strong inhibition
of proliferation. Further, silencing of C/EBPβ led to a complete down-
regulation of antiapoptotic B-cell lymphoma 2 (BCL2) expression. In
chromatin immunoprecipitation assays, C/EBPβ directly bound to
the promoter region of IRF4, BLIMP1, and BCL2. C/EBPβ is involved
in the regulatory network of transcription factors that are critical for
plasma cell dierentiation and survival. Targeting C/EBPβ may provide
a novel therapeutic strategy in the treatment of multiple myeloma.
Exposure plasma cells to lenalidomide activated the Wnt/β-catenin
pathway and its downstream targets such as cyclin D1 and MYC [70].
e accumulation of β-catenin during treatment with lenalidomide
might be cause of drug resistance [70]. N-cadherin-based interaction
between MM cells and osteoblasts block MM cell growth. erefore,
the high levels of N-cadherin expression in osteoblasts confers strong
proliferation block on MM cells. Since β-catenin associates with
N-cadherin at the cell membrane, N-cadherin adhesion is disrupted,
β-catenin is released and will translocate to the nucleus leading to
the transcription of target genes, and ultimately, cell proliferation
[71]. Resistance of myeloma to lenalidomide is an emerging clinical
problem, and though it has been associated in part with activation of
Wnt/β-catenin signaling, the mediators of this phenotype remained
undened. Lenalidomide-resistant models were found to overexpress
the hyaluronan (HA)-binding protein CD44, a downstream Wnt/β-
catenin transcriptional target [72]. Consistent with a role of CD44 in
cell adhesion-mediated drug resistance (CAM-DR), lenalidomide-
resistant myeloma cells were more adhesive to bone marrow stroma
and HA-coated plates. Blockade of CD44 with monoclonal antibodies,
free HA or CD44 knockdown reduced adhesion and sensitized to
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 4-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
lenalidomide. Wnt/β-catenin inhibition by FH535, a compound that
suppresses both Wnt/β-catenin and peroxisome proliferator-activated
receptor (PPAR) signaling, enhanced the activity of lenalidomide, as
did interleukin-6 neutralization with siltuximab.
Cereblon as the direct target protein of IMiDs
Ito, et al. [73-76] developed a new anity bead technology
for isolating ligand-binding proteins. Polymer-coated beads were
constructed that allow single-step purication of ligand target
molecules. ese beads include styrene-glycidyl-methacrylate (SG)
beads and ferrite-glycidyl-methacrylate (FG) beads. FG beads were
used for the purication of thalidomide-binding proteins from various
cell extracts. alidomide-modied beads were incubated with cell
extracts and then washed with buer. Bound proteins were eluted with
free thalidomide and analyzed by gel electrophoresis. Only two specic
protein bands were detected (55 kDa and 127 kDa). ese proteins
were identied as CRBN and DDB1. As we described in Abstract and
Introduction, CRBN, like DDB1, is a component of the cullin 4 ring E3
ubiquitin ligase complex (CRL4). Auto-ubiquitination of CRBN was
inhibited by thalidomide in vitro, suggesting that thalidomide is an
inhibitor of E3 ubiquitin ligase [77].
Cereblon as a primary target for thalidomide teratoge-
nicity
e zebrash was adopted as a model animal for in vivo study of
thalidomide teratogenicity [73-76]. Zebrash have a protein zCrbn,
which is 70% homologous to human CRBN and possesses thalidomide
binding activity. alidomide exposure induces n and ear (otic
vesicle) defects in zebrash. A reduction in in the size of otic vesicle was
observed in zebrash following thalidomide exposure. Zebrash ns
and tetrapod limbs are homologous with respect to early patterning
and gene expression, although skeletal structures in adult sh and
tetrapods are quite dierent. Shortening of the pectoral ns along
the proximo-distal axis and the inhibition of broblast growth factor
8 (Fgf8) gene expression were found in thalidomide-treated zebrash
embryos. Finally, chicks were employed for the investigation of the
conserved role of CRBN. Down-regulation of Fgf8 and Fgf10 cause
multiple birth defects and limb deformities.
e role of Ikaros family proteins in IMiDs and CRBN
mechanism
Using distinct but complementary proteomic techniques and
systems, three groups have recently simultaneously reported that
IMiDs induced the CRBN-dependent proteasomal degradation of
IKZF1 (Ikaros) and IKZF3 (Aiolos) [50,78,79]. Schema is shown in
Figure 2. IKZF1, a zinc nger transcription factor initially discovered
as a regulator of the T cell receptor, is required for hematopoiesis,
particularly lymphocyte development and plasma cell maturation. Loss
of function mutations of IKZF1 and IKZF3 are associated with acute
lymphoblastic leukemia, consistent with a tumor suppressor function.
On the other hand, IKZF1 and IKZF3 are required for the viability of
many MM cell lines. IKZF1 and IKZF3 are also involved in the complex
process of chromatin remodeling, and the nature of their interactions
is poorly understood.
IKZF1 bound and activated the IRF4 gene promoter and loss
of IKZF1 led to decreased IRF4 and MYC expression. However,
lenalidomide could also inhibit MM cell lines with high basal levels
of IRF4 unchanged by drug treatment, suggesting that other IKZF1/3
targets can play a role in the therapeutic response to IMiDs. IKZF1/3 are
known repressors of IL-2 gene promoter. e degradation of IKZF1/3
in response to IMiDs explains enhanced T cell IL-2 production. Hence,
many of the eects of IMiDs can be explained by a unied mechanism:
IMiDs re-target the cullin 4 ring E3 ubiquitin ligase activity toward
IKZF1/3 in a change-of-function eect (Figure 2).
An alternative CRL4CRBN substrate in the lenalidomide-
responsive myelodysplastic syndrome with del(5q)
Krӧnke, et al [18] identied a novel target casein kinase1A1
(CSNK1A1) by quantitative proteomics in the myeloid cell line KG-1.
CSNK1A1 is encoded in the del(5q) commonly deleted region and the
gene is haploinsucient. Lenalidomide treatment leads to increased
ubiquitination of CSNK1A1 and decreased protein abundance (Figure
3). CSNK1A1 was shown as a therapeutic target in a murine model of
AML [80,81] and in MDS with del(5q) [82,83]. CSNK1A1 negatively
regulates β-catenin which drives stem cell self-renewal and CSNK1A1
haploinsuciency causes the initial clonal expansion in patients with
the del(5q) MDS and contributes to the pathogenesis of del(5q) MDS.
Further inhibition of CSNK1A1 in del(5q) MDS is associated with
del(5q) cells apoptosis and p53 activation. e inhibition of CSNK1A1
reduced RPS6 phosphorylation, induced p53 expression, and triggered
myeloid dierentiation program. TP53-null leukemia did not respond
to CSNK1A1 inhibition, strongly supporting the importance of the p53
expression for the yield of CSNK1A1 inhibition. CSNK1A1 mutations
have been recently found in 5-18% of MDS patients with del(5q)
[82,84-87]. ese mutations are associated similarly to the eect of
TP53 mutations with rise to a poor prognosis in del(5q) MDS [87].
While CSNK1A1 is CRL4CRBN target in del(5q) MDS,
CRL4CRBN targets in lower risk non-del(5q) remain to be determined.
e mechanism of action of lenalidomide is still unclear in non-del(5q)
MDS cells.
Importance of valosin-containing protein/p97 for the
degradation of all known CRL4CRBN targets
Valosin-containing protein (VCP)/p97 is ATPase which delivers
ubiquitinated proteins for degradation in proteasomes. VCP is
required for degradation of all known CRL4CRBN targets (IKZF1,
IKZF3, casein kinase 1α, and the translation termination factor
GSPT1) [88]. VCP promotes also degradation of glutamine synthetase
(GS). GS is important for de novo synthesis of glutamine and functions
tn detoxification of glutamate and ammonia. GS is associated with
diseases including cancer, Alzheimer´s disease and Huntington
disease [89].
Immunomodulatory drugs disrupt the cereblon-
CD147-MCT1 axis to exert anti-cancer activity and ter-
atogenicity in a ubiquitin-independent way
CRBN promotes the activation of the CD147 (basigin; BSG) -
MCT1 (solute carrier family 16 members 1; SLC16A1) transmembrane
complex. is complex activatesvarious biological functions, including
angiogenesis, proliferation, invasion and lactate export. Binding of
IMiDs to CRBN leads to destabilization of the CD147-MCT1 complex
and to the inhibition of tumor growth [90].
Measurement of cereblon mRNA and cereblon protein
levels as biomarkers for IMiDs response
e measurement of CRBN mRNA by quantitative RT-PCR
typically uses commercial assays, such as predesigned TaqMan assays,
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 5-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
where primers and probe are optimized with respect to known gene
splicing information. For CRBN mRNA, the current “best coverage
assay” detects the exon 8-exon 9 junction and exon 10 as a measure of
gene expression [91]. is assay (Hs00372271_m1; Applied Biosystems,
Life Technologies Corp.) measures all CRBN mRNA variants that
are translated to functional protein with exception of the variants
with removed exon 10 (part of IMIDs binding region). e presence
of multiple CRBN mRNA splice variants complicates the transcript
measurement by Aymetrix array.
We have found that MDS patients with isolated del(5q) (the so-
called 5q minus syndrome) have higher levels of full-length CRBN
mRNA than other patients with lower risk MDS, linking higher levels
of a known lenalidomide target CRBN and an MDS subgroup known
to be especially sensitive to lenalidomide [92].
Currently available commercial antibodies are neither sensitive
nor specic for reliable detection of CRBN protein levels. Gandhi et
al. [91] characterized a monoclonal antibody CRBN65 and compared
its properties with the commonly used, currently available commercial
antibodies against CRBN. is antibody is the most sensitive and
specic and can detect as little as 200 pg of CRBN protein via Western
blot [91].
Appropriate antibodies and validated assays for cereblon protein
detection and CRBN gene expression that account for the known gene
splicing information are needed for CRBN measurements in the clinic.
While it has been shown that CRBN and IRF4 levels correlate
with lenalidomide responsiveness in MM patients, previous in vitro
investigations using cytogenetically discrete human myeloma cell
lines have not been able to replicate this phenomenon [93,94]. e
implications of this apparent discordance are two-fold. First, these
results show that it is important to investigate CRBN and IRF4 gene
expression in both CD138- and CD138+ myeloma cells. Second,
because there are multiple isoforms of CRBN, it may be important to
Figure 3. Schematic diagram of ubiquitination and degradation of casein kinase 1α by CRL4cereblon E3 ubiquitin ligase and proteasomes. Binding of cereblon (CRBN) by lenalidomide induces
ubiquitination (marking CK1α) and degradation of casein kinase 1α. CRBN functions as a substrate recognition component (substrate receptor) of this E3 ubiquitin ligase enzyme complex.
CRL4cereblon complex consists of cullin 4A, RING nger protein regulator of cullins (Roc1), and DNA damage binding protein 1 (DDB1).
study expression levels of each one in the context of both in vivo and
in vitro settings [91].
Conclusion and perspectives
e studies showing IMiDs induced the CRBN-dependent
proteasomal degradation of IKZF1, IKZF3, and caein kinase
[18,50,78,79] have greatly advanced our understanding of the
mechanism of action of IMiDs in MM and MDS with del(5q). e
small-molecule drug lenalidomide modulates the activity of the CRBN-
CRL4 E3 ubiquitin ligase complex to increase ubiquitination of two
transcription factors, IKZF1 and IKZF3, or casein kinase 1α. It does
so by specic binding to one component of the system, cereblon.
Crystal structures of these complexes were studied in detail [83,94,95].
All these studies may have wider implications for the targeting of E3
ubiquitin ligases in drug discovery for other diseases. ere are very
few approved and experimental drugs that modulate the ubiquitin
system like lenalidomide in this case. Other small molecules could be
developed to alter the specicity of ubiquitination complexes with the
aim of inducing the specic degradation of previously “undruggable”
oncoproteins such as Ras and Myc in many dierent types of cancer
[96-99]. Nevertheless, there are still gaps in our understanding of
the mechanism of action of IMiDs in MM and other hematological
malignancies. For example, the proteasomal inhibitor bortezomib
(VelcadeTM, PS-341) is used for the treatment of MM. Bortezomib
is used eciently also in combination with lenalidomide. ere is an
apparent paradox, because inhibition of proteasomal destruction of
IKZF1 and IKZF3 by bortezomib contradicts the preposed mechanism.
Lu et al. [80] hypothesize that since the proteasomal inhibition by
bortezomib is incomplete with therapeutic dosing, this might allow
sucient destruction of IKZF1 and IKZF3 while retaining other
therapeutic eect of bortezomib. Alternatively, they hypothesize that
IKZF1 and IKZF3 once polyubiquitylated, may be inactive or act as
dominant-negatives.
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 6-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
Future dissection of CRBN direct down-stream substrates and
CRBN indirect down-stream factors will help to identify mechanisms
of IMiD action and nd new biomarkers for prediction of IMiD
response and IMiD resistence as well as developing a new therapy to
treat the patients with MM.
It is also possible to speculate whether or not anti-myeloma
activity of IMiDs is associated with the interaction between CRBN
and AMPK (AMP-activated protein kinase) or KCNT1 (Potassium
Sodium-Activated Channel Subfamily T Member 1). Both AMPK and
KCNT1 are potential substrates of the CRBN-CRL4 E3 ubiquitin ligase
complex, but they may not be associated with downstream signaling
that leads to anti-myeloma activity of IMiDs.
Acknowledgements
is work was supported by the project for conceptual development
of research organisation No 00023736 (Institute of Hematology and
Blood Transfusion) from the Ministry of Health of the Czech Republic.
References
1. Smithells RW, Newman CG (1992) Recognition of thalidomide defects. J Med Genet
29: 716-723.[Crossref]
2. Vargesson N (2009) Thalidomide-induced limb defects: resolving a 50-year-old
puzzle. Bioessays31: 1327-1336.[Crossref]
3. Franks ME, Macpherson GR, Figg WD (2004) Thalidomide. Lancet363: 1802-1811.
[Crossref]
4. Delforge M, Bladé J, Dimopoulos MA, Facon T, Kropff, M, et al. (2010) Treatment-
related peripheral neuropathy in multiple myeloma: the challenge continues. Lancet
Oncol 11: 1086-1095.
5. Richardson PG, Delforge M, Beksac M, Wen P, Jongen JL, et al. (2012) Management
of treatment-emergent peripheral neuropathy in multiple myeloma. Leukemia 26: 595-
608.
6. Andhavarapu S, Roy V (2013) Immunomodulatory drugs in multiple myeloma. Expert
Rev Hematol6: 69-82.
7. Saini N, Mahindra A (2013) Novel immunomodulatory compounds in multiple
myeloma. Expert Opin Investig Drugs 22: 207-215.[Crossref]
8. Davies F, Baz R (2010) Lenalidomide mode of action: linking bench and clinical
ndings. Blood Rev 24 Suppl 1: S13-19.[Crossref]
9. Quach H, Kalff A, Spencer A (2012) Lenalidomide in multiple myeloma: Current
status and future potential. Amer J Hematol87: 1089-1095.
10. Chan AC, Neeson P, Leeansyah E, Tainton K, Quach H, et al. (2013) Natural killer T
cell defects in multiple myeloma and the impact of lenalidomide therapy. Clin Exper
Immunol175: 49-58.
11. Strati P, Keating MJ, Wierda WG, Badoux XC, Calin S, et al. (2013) Lenalidomide
induces long-lasting responses in elderly patients with chronic lymphocytic leukemia.
Blood 122: 734-737.
12. Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, et al. (2013) Trial Watch:
Lenalidomide-based immunochemotherapy. Oncoimmunology2: e26494.[Crossref]
13. Chang X, Zhu Y, Shi C, Stewart AK (2014) Mechanism of immunomodulatory drugs
action in the treatment of multiple myeloma. Acta Biochim Biophys Sin46: 240-253.
14. Keevan J, Figg WD (2014) New mechanism of lenalidomide activity. Cancer Biol Ther
15: 968-969.[Crossref]
15. Desai M, Newberry K, Ou Z, Wang M, Zhang L (2014) Lenalidomide in relapsed or
refractory mantle cell lymphoma: overview and perspective. Ther Adv Hematol 5: 91-
101.[Crossref]
16. Gribben JG, Fowler N, Morschhauser F (2015) Mechanisms of Action of Lenalidomide
in B-Cell Non-Hodgkin Lymphoma. J Clin Oncol 33: 2803-2811.[Crossref]
17. Ghosh N, Grunwald MR, Fasan O, Bhutani M (2015) Expanding role of lenalidomide
in hematologic malignancies. Cancer Management Res7: 105-119.
18. Krönke J, Fink EC, Hollenbach PW, MacBeth KJ, Hurst SN (2015) Lenalidomide
induces ubiquitination and degradation of CK1a in del(5q) MDS. Nature 523: 183-188.
19. Guirguis AA, Ebert BL (2015) Lenalidomide: deciphering mechanisms of action in
myeloma, myelodysplastic syndrome and beyond. Curr Opin Cell Biol37: 61-67.
20. Liang L, Zhao M, Zhu YC, Hu X, Yang LP, et al. (2016) Efcacy of lenalidomide
in relapsed/refractory chronic lymphocytic leukemia patient: a systematic review and
meta-analysis. Ann Hematol95: 1473-1482.
21. Arora M, Gowda S, Tuscano J (2016) A comprehensive review of lenalidomide in
B-cell non-Hodgkin lymphoma. Ther Adv Hematol 7: 209-221.[Crossref]
22. Lindner S, Krönke J (2016) The molecular mechanism of thalidomide analogs in
hematologic malignancies. J Mol Med (Berl) 94: 1327-1334.[Crossref]
23. Manasanch EE (2016) Recommend maintenance therapy with lenalidomide in multiple
myeloma. Semin Oncol43: 712-713.[Crossref]
24. Holstein SA, McCarthy PL (2017) Immunomodulatory Drugs in Multiple Myeloma:
Mechanisms of Action and Clinical Experience. Drugs77: 505-520.[Crossref]
25. Itchaki G, Brown JR (2017) Lenalidomide in the treatment of chronic lymphocytic
leukemia. Expert Opin Investig Drugs 26: 633-650.[Crossref]
26. Stahl M, Zeidan AM (2017) Lenalidomide use in myelodysplastic syndromes: Insights
into the biologic mechanisms and clinical applications. Cancer123: 1703-1713.
[Crossref]
27. Lacy MQ, McCurdy AR (2013) Pomalidomide. Blood122: 2305-2309.[Crossref]
28. Richardson PG, Mark TM, Lacy MQ (2013) Pomalidomide: new immunomodulatory
agent with potent antiproliferative effects. Crit Rev Oncol Hematol 88 Suppl 1: S36-44.
[Crossref]
29. Chanan-Khan AA, Swaika A, Paulus A, Kumar SK, Mikhael JR, et al. (2013)
Pomalidomide: the new immunomodulatory agent for the treatment of multiple
myeloma. Blood Cancer J 3: e143.[Crossref]
30. McCurdy AR, Lacy MQ (2013) Pomalidomide and its clinical potential for relapsed
or refractory multiple myeloma: an update for the hematologist. Ther Adv Hematol 4:
211-216.[Crossref]
31. Mark TM, Coleman M, Niesvizky R (2014) Preclinical and clinical results with
pomalidomide in the treatment of relapsed/refractory multiple myeloma. Leuk Res38:
517-524.
32. Fouquet G, Bories C, Guidez S, Renaud L, Herbaux C, et al. (2014) Pomalidomide for
multiple myeloma. Expert Rev Hematol 7: 719-731.[Crossref]
33. Jones JR, Pawlyn C, Davies FE, Morgan GJ (2016) The safety of pomalidomide for the
treatment of multiple myeloma. Expert Opin Drug Saf 15: 535-547.[Crossref]
34. Touzeau C, Moreau P (2016) Pomalidomide in the management of relapsed multiple
myeloma. Future Oncol 12: 1975-1983.[Crossref]
35. Hagner PR, Man HW, Fontanillo C, Wang M, Couto S, et al. (2015) CC-122, a
pleiotropic pathway modier, mimics an interferon response and has antitumor activity
in DLBCL. Blood 126: 779-789.
36. Nowakowski GS1 (2015) Evolution: IMiDs to PPMs, revolution in DLBCL? Blood126:
698-700.[Crossref]
37. Camicia R, Winkler HC (2015) Novel drug targets for personalized precision medicine
in relapsed/refractory diffuse large B-cell lymphoma: a comprehensive review. Mol
Cancer 14: 207.[Crossref]
38. Cubillos-Zapata C, Córdoba R, Avendaño-Ortiz J, Arribas-Jiménez C, Hernández-
Jiménez E, et al. (2016) CC-122 immunomodulatory effects in refractory patients with
difuse large B-cell lymphoma. Oncoimmunology 5: e1231290.
39. Cubillos-Zapata C, Córdoba R, Avendaño-Ortiz J, Lopez-Collazo E (2017)
Thalidomide analog CC-122 induces a refractory state in monocytes from patients with
difuse large B-cell lymphoma. Leuk Lymphoma58: 1999-2001.
40. Matyskiela ME1, Zhang W1, Man HW1, Muller G1, Khambatta G1, et al. (2017) A
Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos. J Med
Chem.[Crossref]
41. Matyskiela ME, Lu G, Ito T, Pagarigan B, Lu CC, et al. (2016) A novel cereblon
modulator recruits GSPT1 to the CRL4 (CRBN) ubiquitin ligase. Nature 535: 252-257.
42. Hansen JD, Condroski K, Correa M, Muller G, Man HW, et al. (2017) Protein
Degradation via CRL4CRBN Ubiquitin Ligase: Discovery and Structure-Activity
Relationships of Novel Glutarimide Analogs That Promote Degradation of Aiolos and/
or GSPT1. J Med Chem.[Crossref]
43. Quach H, Ritchie D, Stewart AK, Neeson P, Harrison S, et al. (2010) Mechanism of action
of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia24: 22-32.[Crossref]
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 7-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
44. Maiga S, Gomez-Bougie P, Bonnand S, Grates C, Moreau P, et al. (2013) Paradoxical
effect of lenalidomide on cytokine/growth factor proles in multiple myeloma. Br J
Cancer108: 1801-1806.
45. Geitz H, Handt S, Zwingenberger K (1996) Thalidomide selectively modulates
the, density of cell surface molecules involved in the adhesion cascade.
Immunopharmacology 31: 213-221.
46. Muller GW, Chen R, Huang SY, Corral LG, Wong LM, et al. (1999) Amino-substituted
thalidomide analogs: potent inhibitors of TNF-alpha production. Bioorg Med Chem
Lett9: 1625-1630.
47. Hideshima T, Chauhan D, Schlossman R, Richardson P, Anderson KC (2001) The role
of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma:
therapeutic applications. Oncogene 20: 4519-4527.
48. Handa K, Suzuki R, Matsui H, Shimizu Y, Kumagai K (1983)Natural killer (NK) cells
as a responder to interleukin 2 (IL 2). II. IL 2-induced interferon gamma production. J
Immunol130: 988-992.
49. Haslett PA, Colrral LG, Albert M, Kaplan G (1998) Thalidomide costimulates primary
human T lymphocytes, preferentially inducing proliferation, cytokine production, and
cytotoxic responses in the CD8+ subset. J Exp Med187: 1885-1892.
50. Gandhi AK, Kang J, Havens CG, Conklin T, Yuhong N, et al. (2014) Immunomodulatory
agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of
T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex
CRL4CRBN. Br J Haematol164: 811-821.
51. Chang DH, Liu D, Klimek V, Hassoun H, Mazumder A, et al. (2006) Enhancement
of ligand-dependent activation of human natural killer T cells by lenalidomide.:
therapeutic implications. Blood 108: 618-621.
52. Galustian C, Meyer B, Labarte MC, Dredge K, Klaschka D, et al. (2009) The anti-
cancer agents lenalidomide and pomalidomide inhibits the proliferation and function of
T regulatory cells. Cancer Immunol Immunother 58: 1033-1045.
53. Dredge K, Marriott JB, Macdonald CD, Man HW, Chen R, et al. (2002) Novel
thalidomide analogues display anti-angiogenic activity independently of
immunomodulatory effects. Br J Cancer87: 1166-1172.
54. Dredge K, Horsfall R, Robinson SP, Zhang LH, Lu L, et al. (2005) Orally administrated
lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell
migration and Akt phosphorylation in vitro. Microvascular Res69: 56-63.
55. Dankbar B, Paadro T, Leo R, Feldman B, Kropff M, et al. (2000) Vascular endothelial
growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple
myeloma. Blood 95: 2630-2636.
56. Escoubet-Lozach L, Lin IL, Jensen-Pergakes K, Brady HA, Gandhi AK, et al. (2009)
Pomalidomide and lenalidomide induce p21 WAF-1 expression in both lymphoma and
multiple myeloma through a LSD1-mediated epigenetic mechanism. Cancer Res69:
7347-7356.
57. Mitsiades N, Mitsiades CS, Poulaki V, Chauhan D, Richardson PG, et al. (2002)
Apoptotic signaling induced by immunomodulatory thalidomide analogs in human
multiple myeloma cells: Therapeutic implications. Blood 99: 4525-4530.
58. Chu ZL, McKinsey TA, Liu L, Gentry JJ, Malim MH, et al. (1997) Suppression of
tumor necrosis factor-induced cell death by inhibitor of apoptosis c-IAP2 is under NF-
kB control. Proc Natl Acad Sci USA94: 10057-10062.
59. Kreuz S, Siegmund D, Scheurich P, Wajant H (2001) NF-kappaB inducers upregulate
cFLIP, a cycloheximide-sensitive inhibitor of dech receptor signaling. Mol Cell Biol
21: 3964-3973.
60. Li ZW, Chen H, Campbell RA, Bonavida B, Berenson JR (2008) NF-kappaB in the
pathogenesis and treatment of multiple myeloma. Curr Opin Hematol 15: 391-399.
[Crossref]
61. Lopez-Girona A, Heintel D, Zhang LH, Mendy D, Gaidarova S, et al. (2011) Lenalidomide
downregulates the cell survival factor, interferon regulatory factor-4 providing a potential
mechanistic link for predicting response. Br J Haematol154: 325-336.
62. Shaffer AL, Emre NC, Lamy L, Ngo VN, Wright G, et al. (2008) IRF4 addiction in
multiple myeloma. Nature454: 226-231.[Crossref]
63. Raje N, Kumar S, Hideshima T, Ishitsuka K, Chauhan D, et al. (2004) Combination
of the mTOR inhibitor rapamycin and CC-5013 has synergistic activity in multiple
myeloma. Blood 104: 4188-4193.
64. Steinbrunn T, Stühmer T, Sayehli C, Chatterjee M, Einsele H, et al. (2012) Combined
targeting of MEK/MAPK and PI3K/Akt signalling in multiple myeloma. Br J Haematol
159: 430-440.[Crossref]
65. Yang J, He J, Wang J, Cao Y, Ling J, et al. (2012) Constitutive activation of p38 MAPK
in tumor cells contributes to osteolytic bone lesions in multiple myeloma. Leukemia
26: 2114-2123.
66. Tu Y, Gardner A, Lichtenstein A (2000) The phosphatidylinositol 3-kinase/AKT
kinase pathway in multiple myeloma plasma cells: roles in cytokine-dependent survival
and proliferative responses. Cancer Res 60: 6763-6770.
67. Younes H, Leleu X, Hatjiharissi E, Moreau AS, Hideshima T, et al. (2007) Targeting
the phosphatidylinositol 3-kinase pathway in multiple myeloma. Clin Cancer Res 13:
3771-3775.[Crossref]
68. Li S, Pal R, Monaghan SA, Schafer P, Ouyang H, et al. (2011) IMiD immunomodulatory
compounds block C/EBPß translation through eIF4E down-regulation resulting in
inhibition of MM. Blood 117: 5157-5165.
69. Pal R, Janz, M, Galson DL, Gries M, Li S, et al. (2009) C/EBPß regulates transcription
factors critical for proliferation and survival of multiple myeloma cells. Blood 114:
3890-3898.
70. Bjorklund CC, Ma W, Wang ZQ, Davis RE, Kuhn DJ, et al. (2011) Evidence of a
role for activation of Wnt/ß-catenin signaling in the resistance of plasma cells to
lenalidomide. J Biol Chem13: 11009-11020.
71. Sadler NM, Harris BR, Metzger BA, Kirshner J (2013) N-cadherin impedes
proliferation of the multiple myeloma cancer stem cells. Am J Blood Res3: 271-285.
72. Bjorklund CC, Baladandazuthapani V, Lin HY, Jones RJ, Kuiatse I, et al. (2014)
Evidence of a role for CD44 and cell adhesion in mediating resistance to lenalidomide
in multiple myeloma: therapeutic implications. Leukemia 28: 373-383.
73. Ito T, Ando H, Suzuki T, Ogura T, Hotta K, et al. (2010) Identication of a primary
target of thalidomide teratogenicity. Science 327: 1345-1350.
74. Ito T, Ando H, Handa H (2011) Teratogenic effects of thalidomide: molecular
mechanisms. Cell Mol Life Sci 68: 1569-1579.
75. Ito T, Handa H (2012) Deciphering the mystery of thalidomide teratogenicity. Congenit
Anom (Kyoto) 52: 1-7.[Crossref]
76. Ito T, Handa H (2016) Cereblon and its downstream substrates as molecular targets of
immunomodulatory drugs. Int J Hematol104: 293-299.
77. Liu Y, Huang X, He X, Zhou Y, Jiang X, et al. (2015) A novel effect of thalidomide and
its analogs: suppression of cereblon ubiquitination enhances ubiquitin ligase function.
FASEB J 29: 4829-4839.
78. Kr?nke J, Udeshi ND, Narla A, Grauman P, Hurst SN, et al. (2014) Lenalidomide
causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science
343: 301-305.
79. Lu G, Middleton RE, Sun H, Naniong M, Ott CJ, et al. (2014) The myeloma drug
lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science
343: 305-309.
80. Stegmaier K (2014) Targeting Csnk1a1 in leukemia. J Exp Med 211: 594.[Crossref]
81. Järås M, Miller PG, Chu LP, Puram RV, Fink EC, et al. Csnk1a1 inhibition has p53-
dependent therapeutic efcacy in acute myeloid leukemia. (2014) J Exp Med211: 605-612.
82. Schneider RK, Ademà V, Heckl D, Järås M, Mallo M, et al. (2014) Role of casein
kinase 1A1 in the biology and targeted therapy of del(5q) MDS. Cancer Cell26: 509-
520.
83. Petzold G, Fischer ES, Thomä NH (2016) Structural basis of lenalidomide-induced
CK1a degradation by the CRL4(CRBN) ubiquitin ligase. Nature 532:127-130.
84. Heuser M, Meggendorfer M, Cruz MM, Fabisch J, Klesse S, et al. (2015) Frequency
and prognostic impact of casein kinase 1A1 mutations in MDS patients with deletion of
chromosome 5q. Leukemia 29: 1942-1945.
85. Bello E, Pellagatti A, Shaw J, Mecucci C, Kušec R, et al. (2015) CSNK1A1 mutations
and gene expression analysis in myelodysplastic syndromes with del(5q). Br J
Haematol171: 210-214.
86. Boultwood J, Pellagatti A (2015) Clinical associations of CSNK1A1 mutation in
myelodysplastic syndrome. Lancet Haematol2: e182-183.
87. Smith AE, Kulasekararaj AG, Jiang J, Mian S, Mohamedali A, et al. (2015)
CSNK1A1 mutations and isolated del(5q) abnormality in myelodysplastic syndrome: a
retrospective mutational analysis. Lancet Haematol 2: e212-221.
88. Van Nguyen T, Li J, Lu CCJ, Mamrosh JJ, Lu G, et al. (2017) p97/VCP promotes
degradation of CRBN substrate glutamine synthetase and neosubstrates. Proc Natl
Acad Sci USA114: 3565-3571.
Fuchs O (2017) Immunomodulatory drugs and their therapeutic eect in hematological malignancies through cereblon
Volume 2(3): 8-8
Hematol Med Oncol, 2017 doi: 10.15761/HMO.1000129
Copyright: ©2017 Fuchs O. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use,
distribution, and reproduction in any medium, provided the original author and source are credited.
89. Kim HK, Ko TH, Nyamaa B, Lee SR (2016) Cereblon in health and disease. Pugers
Arch 468: 1299-1309.[Crossref]
90. Eichner R, Heider M, Fern8ndey/S8iy V, van Bebber F, Gary AK, et al. (2016)
Immunomodulatory drugs disrupt the cereblon-CD147-MCT1 axis to exert antitumor
activity and teratogenicity. Nat Med 22: 735-743.
91. Gandhi AK, Mendy D, Waldman M, Chen G, Rychak E, et al. (2014) Measuring
cereblon as a biomarker of response or resistance to lenalidomide and pomalidomide
requires use of standardized reagents and understanding of gene complexity. Br J
Haematol 164: 233-244.[Crossref]
92. Jonasova A, Bokorova R, Polak J, Vostry M (2015) High level of full-length cereblon
mRNA in lower risk myelodysplastic syndrome with isolated 5q deletion is implicated
in the efcacy of lenalidomide. Eur J Haematol 95: 27-34.[Crossref]
93. Lopez-Girona A, Mendy D, Ito T, Miller K, Gandhi AK, et al. (2012) Cereblon is
a direct protein target for immunomodulatory and antiproliferative activities of
lenalidomide and pomalidomide. Leukemia 26: 2326-2335.
94. Greenberg AJ, Walters DK, Kumar SK, Vincent Rajkumar S, Jelinek DF, et al. (2013)
Responsiveness of cytogenetically discrete human myeloma cell lines to lenalidomide:
lack of correlation with cereblon and interferon regulatory factor 4 expression levels.
Eur J Haematol91: 504-513.
95. Fischer ES, Böhm K, Lydeard JR, Yang H, Stadler MB, et al. (2014) Structure of the DDB1-
CRBN E3 ubiquitin ligase in complex with thalidomide. Nature512: 49-53.[Crossref]
96. Chamberlain PP, Lopez-Girona A, Miller K, Carmel G, Pagarigan B, et al. (2014)
Structure of the human cereblon-DDB1-lenalidomide complex reveals basis for
responsiveness to thalidomide analogs. Nat Str Mol Biol 21: 803-809.
97. Bulatov E, Ciulli A (2015) Targeting cullin-ring E3 ubiquirin ligases for drug discovery:
structure, assembly and small-molecule modulation. Biochem J 467: 365-386.
98. Huang X, Dixit VM (2016) Drugging the undruggables: exploring the ubiquitin system
for drug development. Cell Res26: 484-498.
99. Collins I, Wang H, Caldwell JJ, Chopra R (2017) Chemical approaches to targeted
protein degradation through modulation of the ubiquitin-proteasome pathway. Biochem
J474: 1127-1147.[Crossref]
... Lenalidomide (CC-5013) is the 4-amino-glutamyl analogue of thalidomide. Unlike thalidomide, lenalidomide is not sedative, and the incidence of sensory axonal neuropathy is less frequent, but still possible [13,14]. ...
Article
Full-text available
Over the past two decades, the improvement in our understanding of the biology of MM and the introduction of new drug classes, including immunomodulatory drugs (IMiDs), proteasome inhibitors (PI), and monoclonal antibodies (MoAb), have significantly improved outcomes. The first IMiD introduced to treat MM was thalidomide. The side effects observed during treatment with thalidomide initiated work on the synthesis of IMiD analogs. Subsequently, lenalidomide and pomalidomide were developed, both with different safety profiles, and they have better tolerability than thalidomide. In 2010, the cereblon (CRBN) protein was discovered as a direct target of IMiDs. By binding to CRBN, IMiDs change the substrate specificity of the CRBN E3 ubiquitin ligase complex, which results in the breakdown of internal Ikaros and Aiolos proteins. Most clinical trials conducted, both in newly diagnosed, post-transplant maintenance and relapsed/refractory MM, report a beneficial effect of IMiDs on the extension of progression-free survival and overall survival in patients with MM. Due to side effects, thalidomide is used less frequently. Currently, lenalidomide is used at every phase of MM treatment. Lenalidomide is used in conjunction with other agents such as PIs and MoAb as induction and relapsed therapy. Pomalidomide is currently used to treat relapsed/refractory MM, also with PIs and monoclonal antibodies. Current clinical trials are evaluating the efficacy of IMiD derivatives, the CRBN E3 ligase modulators (CELMoDs). This review focuses on the impact of IMiDs for the treatment of MM.
... The anti-myeloma activity of IMiDs has been attributed to several mechanisms of action, including anti-angiogenic, pro-apoptotic, and anti-proliferative effects 1,7 . Until recently, however, the precise molecular mechanisms by which thalidomide and its analogs act remained elusive. ...
Article
Full-text available
Carfilzomib-lenalidomide-dexamethasone (KRd) therapy has yielded promising results in patients with newly diagnosed multiple myeloma (NDMM). Cereblon (CRBN) is the direct molecular target of lenalidomide and genetic polymorphisms in CRBN have been associated with lenalidomide efficacy. In this study, we assessed the correlation of five single nucleotide variants (SNVs) in the CRBN gene with clinical response and outcomes in patients with NDMM administered KRd therapy with lenalidomide maintenance, achieving favorable trial endpoints in a prospective Phase II study (NCT01402284). Of the observed SNVs, no associations with KRd therapy response were found in this patient cohort, although strong trends in hypoalbuminemia grade and hyperbilirubinemia grade emerged across the CRBN rs1672753 genotype (P = 0.0008) and the rs1714327 genotype (P = 0.0010), respectively. Our results do not provide conclusive support for the predictive utility of CRBN gene polymorphisms as potential biomarkers of clinical response to lenalidomide-based therapy in our patient population. However, these findings remain to be validated in prospective studies using larger patient populations.
Article
A persistent challenge in developing personalized treatments for hematologic cancers is the lack of patient specific, physiologically relevant disease models to test investigational drugs in clinical trials and to select therapies in a clinical setting. Biomicrofluidic systems and organ-on-a-chip technologies have the potential to change how researchers approach the fundamental study of hematologic cancers and select clinical treatment for individual patient. Here, we review microfluidics cell-based technology with application toward studying hematologic tumor microenvironments (TMEs) for the purpose of drug discovery and clinical treatment selection. We provide an overview of state-of-the-art microfluidic systems designed to address questions related to hematologic TMEs and drug development. Given the need to develop personalized treatment platforms involving this technology, we review pharmaceutical drugs and different modes of immunotherapy for hematologic cancers, followed by key considerations for developing a physiologically relevant microfluidic companion diagnostic tool for mimicking different hematologic TMEs for testing with different drugs in clinical trials. Opportunities lie ahead for engineers to revolutionize conventional drug discovery strategies of hematologic cancers, including integrating cell-based microfluidics technology with machine learning and automation techniques, which may stimulate pharma and regulatory bodies to promote research and applications of microfluidics technology for drug development.
Article
Full-text available
Manipulation of the ubiquitin–proteasome system to achieve targeted degradation of proteins within cells using chemical tools and drugs has the potential to transform pharmacological and therapeutic approaches in cancer and other diseases. An increased understanding of the molecular mechanism of thalidomide and its analogues following their clinical use has unlocked small-molecule modulation of the substrate specificity of the E3 ligase cereblon (CRBN), which in turn has resulted in the advancement of new immunomodulatory drugs (IMiDs) into the clinic. The degradation of multiple context-specific proteins by these pleiotropic small molecules provides a means to uncover new cell biology and to generate future drug molecules against currently undruggable targets. In parallel, the development of larger bifunctional molecules that bring together highly specific protein targets in complexes with CRBN, von Hippel–Lindau, or other E3 ligases to promote ubiquitin-dependent degradation has progressed to generate selective chemical compounds with potent effects in cells and in vivo models, providing valuable tools for biological target validation and with future potential for therapeutic use. In this review, we survey recent breakthroughs achieved in these two complementary methods and the discovery of new modes of direct and indirect engagement of target proteins with the proteasome. We discuss the experimental characterisation that validates the use of molecules that promote protein degradation as chemical tools, the preclinical and clinical examples disclosed to date, and the future prospects for this exciting area of chemical biology.
Article
The introduction of novel immunomodulatory drugs (IMiDs), such as lenalidomide, has drastically improved the survival of patients with multiple myeloma (MM). While it has been shown that patients with specific cytogenetic subtypes, namely t(4;14), have the best outcomes when treated with bortezomib-based regimens, the relationship between cytogenetic subtypes and response to IMiDs remains unclear. Using DNA synthesis and apoptosis assays, we investigated the relationship between cytogenetic subtype and lenalidomide response in a representative panel of human myeloma cell lines (HMCLs). Additionally, we examined HMCL protein expression levels of the lenalidomide target cereblon (CRBN) and its putative downstream target interferon regulatory factor 4 (IRF4), both of which have previously been shown to be predictive of lenalidomide response in HMCLs. Our results reveal that response to lenalidomide did not correlate with specific cytogenetic translocations. There were, however, distinct groups of lenalidomide-responsive and non-responsive HMCLs, as defined by inhibition of cellular proliferation; notably, all of the hyperdiploid HMCLs fell into the latter category. Repeated dosing of lenalidomide significantly lowered the IC50 of the responsive HMCL ALMC-1 (IC50 = 2.6 μM versus 0.005 μM, p=0.004), but did not have an effect on the IC50 of the non-responsive HMCL DP-6 (p>0.05). Moreover, no association was found between lenalidomide responsiveness and CRBN or IRF4 expression levels. In summary, our data indicate that lenalidomide sensitivity is independent of cytogenetic subtype in HMCLs. Additionally, while CRBN and IRF-4 have been shown to be associated with response to lenalidomide in patients, these findings do not translate back to HMCLs, which could be attributable to factors present in the bone marrow microenvironment. These results suggest that the development of a new treatment paradigm based on cytogenetic subtype of MM alone may be insufficient, and that additional markers may need to be incorporated. Furthermore, the disconnect between our findings of expression of CRBN and IRF-4 in lenalidomide-treated HMCLs and patient data highlights the need for further investigation into the role of the bone marrow microenvironment on the regulation of these previously demonstrated markers of lenalidomide response. Disclosures No relevant conflicts of interest to declare.
Article
The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4–CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degradation. Here we describe CC-220 (compound 6), a cereblon modulator in clinical development for systemic lupus erythematosis and relapsed/refractory multiple myeloma. Compound 6 binds cereblon with a higher affinity than lenalidomide or pomalidomide. Consistent with this, the cellular degradation of Ikaros and Aiolos is more potent and the extent of substrate depletion is greater. The crystal structure of cereblon in complex with DDB1 and compound 6 reveals that the increase in potency correlates with increased contacts between compound 6 and cereblon away from the modeled binding site for Ikaros/Aiolos. These results describe a new cereblon modulator which achieves greater substrate degradation via tighter binding to the cereblon E3 ligase and provides an example of the effect of E3 ligase binding affinity with relevance to other drug discovery efforts in targeted protein degradation.
Article
Introduction: Lenalidomide is an immunomodulatory drug (IMiD) with a unique mode of action (MOA) that may vary across disease-type. It is currently approved in multiple myeloma (MM), myelodysplastic syndrome (MDS) and mantle cell lymphoma (MCL), yet is also clinically active in a host of lymphoproliferative diseases, including chronic lymphocytic leukemia (CLL). Due to its protean effects on the immune system, lenalidomide may be particularly appealing in CLL, which is distinct in its ability to evade immune recognition and cause immunosuppression. Areas covered: This review recaps the biological mechanisms of lenalidomide specific for CLL, and summarizes the clinical data in previously untreated and relapsed/refractory (R/R) CLL patients, with emphasis on toxicity. Moreover, lenalidomide treatment is put into the context of the highly effective targeted agents that are drastically changing the therapeutic approach in CLL. Expert Opinion: Lenalidomide is a potent drug in CLL, both in first line and relapse. However, in comparison to other newly available agents, lenalidomide has slow onset of efficacy and notable toxicity profile that limits both its single agent use and combinations with chemotherapy. Future trials will hopefully direct our ability to harness lenalidomide MOA to best incorporate it in the rapidly evolving landscape of CLL treatment.
Article
We previously disclosed the identification of cereblon modulator CC-885, with potent antitumor activity mediated through the degradation of GSPT1. We describe herein the structure-activity relationships of CC-885 analogs with exploration of the structurally related dioxoisoindoline class. The observed activity of protein degradation could in part be rationalized through docking into the previously disclosed CC-885-CRBN-GSPT1 co-crystal ternary complex. SAR that could not be rationalized through the co-crystal complex we sought to predict through a QSAR model developed in house. Through these analyses, selective protein degradation could be achieved between two proteins of interest, GSPT1, and Aiolos.
Article
Over the last two decades, the outcomes for patients with multiple myeloma, a plasma cell malignancy, have dramatically improved. The development of the immunomodulatory drugs (IMiDs), which include thalidomide, lenalidomide, and pomalidomide, has contributed significantly to these improved outcomes. While thalidomide is now less commonly prescribed, lenalidomide is widely used in the treatment of newly diagnosed transplant-eligible and transplant-ineligible patients, in the maintenance setting post-transplant and in the relapsed/refractory setting, while pomalidomide is currently utilized in the relapsed/refractory setting. The IMiDs have been reported to have a multitude of activities, including anti-angiogenic, cytotoxic, and immunomodulatory. However, the more recent discoveries that the IMiDs bind to cereblon and thus regulate the ubiquitination of key transcription factors including IKZF1 and IKZF3 have provided greater insight into their mechanism of action. Here, the clinical efficacy of these agents in myeloma is reviewed and the structure-function relationship, the molecular mechanisms of action, and the association of IMiDs with second primary malignancies and thrombosis are discussed.
Article
Myelosysplastic syndromes (MDS) include a heterogeneous group of clonal myeloid neoplasms characterized by ineffective hematopoiesis leading to blood cytopenias and a variable risk of progression into acute myeloid leukemia (AML). Although the hypomethylating agent azacitidine prolongs survival among patients with higher risk (HR)-MDS compared with conventional care, no drug has been shown conclusively to prolong survival or delay progression to AML among patients with lower-risk MDS (LR-MDS). Lenalidomide is the drug with the most impressive clinical activity in the subset of anemic LR-MDS patients who harbor a deletion of the long arm of chromosome 5 (5q-), where it leads to high rates of transfusion independence and cytogenetic responses. Furthermore, lenalidomide delays progression to AML and prolongs survival among responders. In this article, we review the recently recognized mechanisms of action of lenalidomide and discuss the most recent clinical data regarding its use in patients with both 5q- MDS as well as non-5q- MDS. Finally, we forecast the future directions to improve the efficacy of lenalidomide in MDS with and without 5q-. Cancer 2017. © 2017 American Cancer Society.
Article
Thalidomide was the first immunomodulatory drug used as maintenance after autologous stem cell transplant (ASCT) in multiple myeloma (MM). This showed improved progression-free survival (PFS) and in some cases, overall survival (OS). Despite this, use of thalidomide was limited due to toxicity and high rates of therapy discontinuation. Lenalidomide, an analog of thalidomide, had a more favorable toxicity profile making its use in maintenance a potential approach. The use of lenalidomide as a maintenance therapy after ASCT in newly diagnosed MM patients has been investigated in four phase III randomized control studies.
Article
In the three patients included in a phase I clinical trial (NCT01421524) we report the immunomodulatory effects and efficacy of CC-122, a novel pleiotropic pathway modifier compound originally developed for broad diffuse large B-cell lymphoma (DLBCL). The chemical structure of CC-122 includes the glutarimide moiety that is known to modulate the immune response. The immunomodulatory agents including lenalidomide represent a promising therapeutic strategy targeting tumours in B-cell lymphoid malignancies. We observed that CC-122 might regulate the NK phenotype and its activity due to the reduced accumulation of myeloid derived suppressor cell and eventually decrease the Tregs subsets. Finally, the activation of T cells through co-stimulatory molecule (CD28) was detected as a delayed CC-122 effect. In this context, CC-122 arises as an alternative option for DLBCL patients refractory to the traditional chemotherapeutic agents.