scholarly journals The novel mechanism of lenalidomide activity

Blood ◽  
2015 ◽  
Vol 126 (21) ◽  
pp. 2366-2369 ◽  
Author(s):  
Emma C. Fink ◽  
Benjamin L. Ebert

Abstract Lenalidomide acts by a novel drug mechanism—modulation of the substrate specificity of the CRL4CRBN E3 ubiquitin ligase. In multiple myeloma, lenalidomide induces the ubiquitination of IKZF1 and IKZF3 by CRL4CRBN. Subsequent proteasomal degradation of these transcription factors kills multiple myeloma cells. In del(5q) myelodysplastic syndrome, lenalidomide induces the degradation of CK1α, which preferentially affects del(5q) cells because they express this gene at haploinsufficient levels. In the future, modulation of ubiquitin ligase function may enable us to target previously “undruggable” proteins.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. SCI-51-SCI-51
Author(s):  
Jan Krönke

Abstract Lenalidomide is a derivative of thalidomide, a drug developed in the 1950s as a sedative and treatment for morning sickness that became infamous for causing limb deformations (phocomelia) and other birth defects when used by pregnant women. Following the discovery in the 1990s that thalidomide inhibits the release of tumor necrosis factor (TNF) and blocks angiogenesis, researchers began studying thalidomide in other diseases, including cancer. Thalidomide demonstrated high in vitro activity in multiple myeloma and high response rates in clinical trials, leading to its accelerated approval by the FDA in 2006. In addition to the direct antiproliferative effects on multiple myeloma cells, thalidomide and its more potent derivatives, lenalidomide and pomalidomide, have pleiotropic effects on immune cells and are therefore called immunomodulatory drugs (IMiDs). IMiDs enhance the release of interleukin-2 (IL-2) and interferon-γ (IFN-γ) from activated T cells, inhibit the immunosuppressive activity of regulatory T cells, and increase natural killer (NK) cell-mediated cytotoxicity. In peripheral blood monocytes (PBMCs) IMiDs inhibit the release of TNF and other cytokines including interleukin-6 (IL-6), a critical growth factor for multiple myeloma cells. Recently, cereblon (CRBN) was identified as the common primary target for all IMiDs. CRBN forms an E3 ubiquitin ligase together with DNA damage-binding protein 1 (DDB1), cullin 4A (CUL4A), and regulators of cullins (ROC1) CRBN-CRL4. Interaction with this enzymatic complex has been shown to be essential for most properties of IMiDs including teratogenicity, antiproliferative effects in multiple myeloma and some of the immunomodulatory properties. More recently, it was demonstrated that lenalidomide and its analogues activate the CRBN-CRL4 E3 ligase to ubiquitinate and degrade two members of the Ikaros family of zinc finger transcription factors: Ikaros (IKZF1) and Aiolos (IKZF3). IKZF1 and IKZF3 are key regulators in lymphopoiesis and essential for lymphoid progenitor differentiation into effector cells. While IKZF1 and IKZF3 deletions and loss of function mutations are frequent in acute lymphoblastic leukemia, mature B-cell lymphomas like multiple myeloma and chronic lymphocytic leukemia have high IKZF1 and IKZF3 expression. Inactivation of IKZF1 and IKZF3 results in growth inhibition in multiple myeloma. Conversely, over-expression of IKZF1 or IKZF3 confers lenalidomide resistance, demonstrating that degradation of IKZF1 and IKZF3 is responsible for the direct cytotoxic effects of lenalidomide in multiple myeloma. One of the transcriptional targets of IKZF1 and IKZF3 is interferon regulatory factor 4 (IRF4), a transcription factor that is essential for proliferation and survival of multiple myeloma cells that is down-regulated after lenalidomide-induced degradation of IKZF1 and IKZF3. At the IL-2 locus, IKZF3 is a transcriptional repressor that is de-repressed after lenalidomide-induced degradation of IKZF3; explaining one of the immunomodulatory properties of lenalidomide. While IKZF1 and IKZF3 degradation is likely involved in the other effects of lenalidomide on T cell subsets and NK cells, it is unlikely that this accounts for all of its properties. Limb deformations, for instance, do not occur in mice with germline genetic inactivation of IKZF1 or IKZF3. Similarly, it is unlikely that degradation of the lymphoid transcription factors IKZF1 and IKZF3 accounts for the specific activity of lenalidomide in myelodysplastic syndrome with chromosome 5q deletion. Since most of these effects have been shown to depend on CRBN, it is conceivable that they result from lenalidomide-induced alteration of other substrates of the CRBN-CRL4 E3 ubiquitin ligase. Future studies aiming to identify the substrates responsible for each of the biological effects of IMiDs could enable the development of more specific drugs that modify ubiquitination of different sets of proteins with fewer side effects. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 7 (6) ◽  
pp. eabd6263
Author(s):  
Vidyasagar Koduri ◽  
Leslie Duplaquet ◽  
Benjamin L. Lampson ◽  
Adam C. Wang ◽  
Amin H. Sabet ◽  
...  

Most intracellular proteins lack hydrophobic pockets suitable for altering their function with drug-like small molecules. Recent studies indicate that some undruggable proteins can be targeted by compounds that can degrade them. For example, thalidomide-like drugs (IMiDs) degrade the critical multiple myeloma transcription factors IKZF1 and IKZF3 by recruiting them to the cereblon E3 ubiquitin ligase. Current loss of signal (“down”) assays for identifying degraders often exhibit poor signal-to-noise ratios, narrow dynamic ranges, and false positives from compounds that nonspecifically suppress transcription or translation. Here, we describe a gain of signal (“up”) assay for degraders. In arrayed chemical screens, we identified novel IMiD-like IKZF1 degraders and Spautin-1, which, unlike the IMiDs, degrades IKZF1 in a cereblon-independent manner. In a pooled CRISPR-Cas9–based screen, we found that CDK2 regulates the abundance of the ASCL1 oncogenic transcription factor. This methodology should facilitate the identification of drugs that directly or indirectly degrade undruggable proteins.


Author(s):  
Ota Fuchs

Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.


2004 ◽  
Vol 23 (19) ◽  
pp. 3780-3792 ◽  
Author(s):  
Su Ryeon Seo ◽  
François Lallemand ◽  
Nathalie Ferrand ◽  
Marcia Pessah ◽  
Sébastien L'Hoste ◽  
...  

Haematologica ◽  
2019 ◽  
Vol 105 (5) ◽  
pp. e237-e241 ◽  
Author(s):  
Santiago Barrio ◽  
Umair Munawar ◽  
Yuan Xiao Zhu ◽  
Nicola Giesen ◽  
Chang-Xin Shi ◽  
...  

eLife ◽  
2018 ◽  
Vol 7 ◽  
Author(s):  
Gang Lu ◽  
Stephanie Weng ◽  
Mary Matyskiela ◽  
Xinde Zheng ◽  
Wei Fang ◽  
...  

The cereblon modulating agents (CMs) including lenalidomide, pomalidomide and CC-220 repurpose the Cul4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase complex to induce the degradation of specific neomorphic substrates via polyubiquitination in conjunction with E2 ubiquitin-conjugating enzymes, which have until now remained elusive. Here we show that the ubiquitin-conjugating enzymes UBE2G1 and UBE2D3 cooperatively promote the K48-linked polyubiquitination of CRL4CRBN neomorphic substrates via a sequential ubiquitination mechanism. Blockade of UBE2G1 diminishes the ubiquitination and degradation of neomorphic substrates, and consequent antitumor activities elicited by all tested CMs. For example, UBE2G1 inactivation significantly attenuated the degradation of myeloma survival factors IKZF1 and IKZF3 induced by lenalidomide and pomalidomide, hence conferring drug resistance. UBE2G1-deficient myeloma cells, however, remained sensitive to a more potent IKZF1/3 degrader CC-220. Collectively, it will be of fundamental interest to explore if loss of UBE2G1 activity is linked to clinical resistance to drugs that hijack the CRL4CRBN to eliminate disease-driving proteins.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2856-2856 ◽  
Author(s):  
Dirk Hose ◽  
Anja Seckinger ◽  
Hartmut Goldschmidt ◽  
Tobias Meiβner ◽  
Blanka Rebacz ◽  
...  

Abstract Abstract 2856 Poster Board II-832 BACKGROUND. Molecular profiling of multiple myeloma allows the identification of novel targets, including HIF1A, and evaluation of their expression within large cohorts of patients. We report here the expression of HIF1A in myeloma and for the first time the preclinical testing of 4 members of a novel class of sulfonanilide HIF1A signaling inhibitors. PATIENTS AND METHODS. Expression of HIF1A was assessed using Affymetrix DNA-microarrays in 329 samples of CD138-purified myeloma cells from previously untreated patients. Chromosomal aberrations were assessed by comprehensive iFISH using a set of probes for the chromosomal regions 1q21, 6q21, 8p21, 9q34, 11q23, 11q13, 13q14.3, 14q32, 15q22, 17p13, 19q13, 22q11, as well as the translocations t(4;14)(p16.3;q32.3) and t(11;14)(q13;q32.3). Proliferation of primary myeloma cells (n=67) was determined by propidium iodine staining. The effect of the novel HIF1A signaling inhibitors ELR510490, ELR510454, ELR510444 and ELR105813 on the proliferation of 12 human myeloma cell lines and the first three on the survival of 5 primary myeloma cell-samples cultured within their microenvironment was tested, and their ability to inhibit HIF1A signaling was examined using a cell-based reporter assay. Studies were also conducted to determine in vitro stability (in plasma and microsomes), as well as single-dose PK (SDPK) parameters and maximum tolerated dose (MTD) levels after dosing in mice. RESULTS. We found (i) HIF1A to be expressed by 95.4% of CD138-purified primary myeloma cell samples from previously untreated patients. (ii) HIF1A expression shows a weak but significant correlation (r=0.3, p<0.001) with a gene expression based proliferation index. (iii) Of the chromosomal aberrations tested, myeloma cells of patients with presence of a translocation t(4,14) show a significantly higher expression of HIF1A (p<0.001) vs. patients without. Myeloma cells of hyperdiploid patients show a significantly lower expression of HIF1A (p=0.02) vs. non hyperdiploid patients. (iii) HIF1A expression does not show a correlation with event-free or overall survival. (iv) The sulfonanilides ELR510490, ELR510444, ELR510454 and ELR105813 completely inhibit proliferation of all tested myeloma cell lines at nM concentrations. (v) The compounds tested, i.e. ELR510490, ELR510444, ELR510454, are active on all primary myeloma cell-samples tested. (vi) The compounds show a pronounced effect on the HIF1A signaling pathway at EC50s of 1-25nM. (vii) Pre-clinical pharmacology data for the compounds ELR510444 and ELR510490 in mice indicate favorable absorption, distribution, metabolism, and excretion (ADME) profiles as well as exposure levels upon dosing at well-tolerated levels that are significantly above the in vitro EC50 in all the cell lines tested. CONCLUSION. HIF1A is expressed in almost all primary myeloma cells. The novel HIF1A signaling inhibitors tested are very active on myeloma cell lines as well as primary myeloma cells and show favorable in vivo profiles with exposure levels in mice significantly higher than the concentrations required for the inhibition of cell proliferation or apoptosis induction in vitro. This class of compounds thus represents a promising weapon in the therapeutic arsenal against multiple myeloma. Disclosures: Rebacz: ELARA Pharmaceuticals: Employment. Lewis:ELARA Pharmaceuticals: Employment. Schultes:ELARA Pharmaceuticals: Employment.


Sign in / Sign up

Export Citation Format

Share Document