Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 145(7): 1075-87, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21683433

ABSTRACT

In the ubiquitin-proteasome system (UPS), E2 enzymes mediate the conjugation of ubiquitin to substrates and thereby control protein stability and interactions. The E2 enzyme hCdc34 catalyzes the ubiquitination of hundreds of proteins in conjunction with the cullin-RING (CRL) superfamily of E3 enzymes. We identified a small molecule termed CC0651 that selectively inhibits hCdc34. Structure determination revealed that CC0651 inserts into a cryptic binding pocket on hCdc34 distant from the catalytic site, causing subtle but wholesale displacement of E2 secondary structural elements. CC0651 analogs inhibited proliferation of human cancer cell lines and caused accumulation of the SCF(Skp2) substrate p27(Kip1). CC0651 does not affect hCdc34 interactions with E1 or E3 enzymes or the formation of the ubiquitin thioester but instead interferes with the discharge of ubiquitin to acceptor lysine residues. E2 enzymes are thus susceptible to noncatalytic site inhibition and may represent a viable class of drug target in the UPS.


Subject(s)
Amino Acids/pharmacology , Biphenyl Compounds/pharmacology , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Allosteric Site , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , DNA Mutational Analysis , Humans , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligase Complexes/chemistry , Ubiquitin-Protein Ligase Complexes/genetics
2.
Blood ; 137(2): 216-231, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33024998

ABSTRACT

Cancer treatment has been transformed by checkpoint blockade therapies, with the highest anti-tumor activity of anti-programmed death 1 (PD-1) antibody therapy seen in Hodgkin lymphoma. Disappointingly, response rates have been low in the non-Hodgkin lymphomas, with no activity seen in relapsed/refractory chronic lymphocytic leukemia (CLL) with PD-1 blockade. Thus, identifying more powerful combination therapy is required for these patients. Here, we preclinically demonstrate enhanced anti-CLL activity following combinational therapy with anti-PD-1 or anti-PD-1 ligand (PD-L1) and avadomide, a cereblon E3 ligase modulator (CELMoD). Avadomide induced type I and II interferon (IFN) signaling in patient T cells, triggering a feedforward cascade of reinvigorated T-cell responses. Immune modeling assays demonstrated that avadomide stimulated T-cell activation, chemokine expression, motility and lytic synapses with CLL cells, as well as IFN-inducible feedback inhibition through upregulation of PD-L1. Patient-derived xenograft tumors treated with avadomide were converted to CD8+ T cell-inflamed tumor microenvironments that responded to anti-PD-L1/PD-1-based combination therapy. Notably, clinical analyses showed increased PD-L1 expression on T cells, as well as intratumoral expression of chemokine signaling genes in B-cell malignancy patients receiving avadomide-based therapy. These data illustrate the importance of overcoming a low inflammatory T-cell state to successfully sensitize CLL to checkpoint blockade-based combination therapy.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Activation/drug effects , Piperidones/pharmacology , Quinazolinones/pharmacology , T-Lymphocytes/drug effects , Animals , Antineoplastic Agents/pharmacology , Humans , Immunotherapy/methods , Interferons/immunology , Mice , Signal Transduction/drug effects , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
3.
Blood ; 137(5): 661-677, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33197925

ABSTRACT

A number of clinically validated drugs have been developed by repurposing the CUL4-DDB1-CRBN-RBX1 (CRL4CRBN) E3 ubiquitin ligase complex with molecular glue degraders to eliminate disease-driving proteins. Here, we present the identification of a first-in-class GSPT1-selective cereblon E3 ligase modulator, CC-90009. Biochemical, structural, and molecular characterization demonstrates that CC-90009 coopts the CRL4CRBN to selectively target GSPT1 for ubiquitination and proteasomal degradation. Depletion of GSPT1 by CC-90009 rapidly induces acute myeloid leukemia (AML) apoptosis, reducing leukemia engraftment and leukemia stem cells (LSCs) in large-scale primary patient xenografting of 35 independent AML samples, including those with adverse risk features. Using a genome-wide CRISPR-Cas9 screen for effectors of CC-90009 response, we uncovered the ILF2 and ILF3 heterodimeric complex as a novel regulator of cereblon expression. Knockout of ILF2/ILF3 decreases the production of full-length cereblon protein via modulating CRBN messenger RNA alternative splicing, leading to diminished response to CC-90009. The screen also revealed that the mTOR signaling and the integrated stress response specifically regulate the response to CC-90009 in contrast to other cereblon modulators. Hyperactivation of the mTOR pathway by inactivation of TSC1 and TSC2 protected against the growth inhibitory effect of CC-90009 by reducing CC-90009-induced binding of GSPT1 to cereblon and subsequent GSPT1 degradation. On the other hand, GSPT1 degradation promoted the activation of the GCN1/GCN2/ATF4 pathway and subsequent apoptosis in AML cells. Collectively, CC-90009 activity is mediated by multiple layers of signaling networks and pathways within AML blasts and LSCs, whose elucidation gives insight into further assessment of CC-90009s clinical utility. These trials were registered at www.clinicaltrials.gov as #NCT02848001 and #NCT04336982).


Subject(s)
Acetamides/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Isoindoles/pharmacology , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Piperidones/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Acetamides/therapeutic use , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Humans , Isoindoles/therapeutic use , Mice , Mice, Inbred NOD , Mice, SCID , Models, Molecular , Neoplastic Stem Cells/enzymology , Nuclear Factor 45 Protein/physiology , Nuclear Factor 90 Proteins/physiology , Peptide Termination Factors/metabolism , Piperidones/therapeutic use , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , Protein Processing, Post-Translational/drug effects , Proteolysis , Small Molecule Libraries , Stress, Physiological , TOR Serine-Threonine Kinases/physiology , U937 Cells , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
4.
Nature ; 535(7611): 252-7, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27338790

ABSTRACT

Immunomodulatory drugs bind to cereblon (CRBN) to confer differentiated substrate specificity on the CRL4(CRBN) E3 ubiquitin ligase. Here we report the identification of a new cereblon modulator, CC-885, with potent anti-tumour activity. The anti-tumour activity of CC-885 is mediated through the cereblon-dependent ubiquitination and degradation of the translation termination factor GSPT1. Patient-derived acute myeloid leukaemia tumour cells exhibit high sensitivity to CC-885, indicating the clinical potential of this mechanism. Crystallographic studies of the CRBN-DDB1-CC-885-GSPT1 complex reveal that GSPT1 binds to cereblon through a surface turn containing a glycine residue at a key position, interacting with both CC-885 and a 'hotspot' on the cereblon surface. Although GSPT1 possesses no obvious structural, sequence or functional homology to previously known cereblon substrates, mutational analysis and modelling indicate that the cereblon substrate Ikaros uses a similar structural feature to bind cereblon, suggesting a common motif for substrate recruitment. These findings define a structural degron underlying cereblon 'neosubstrate' selectivity, and identify an anti-tumour target rendered druggable by cereblon modulation.


Subject(s)
Antineoplastic Agents/pharmacology , Peptide Hydrolases/metabolism , Peptide Termination Factors/metabolism , Phenylurea Compounds/pharmacology , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Antineoplastic Agents/chemistry , Binding Sites , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Ikaros Transcription Factor/chemistry , Ikaros Transcription Factor/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Peptide Hydrolases/chemistry , Peptide Termination Factors/chemistry , Peptide Termination Factors/deficiency , Phenylurea Compounds/chemistry , Protein Binding , Proteolysis/drug effects , Substrate Specificity , Thalidomide/chemistry , Thalidomide/pharmacology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
5.
Blood ; 126(6): 779-89, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26002965

ABSTRACT

Cereblon (CRBN), a substrate receptor of the Cullin 4 RING E3 ubiquitin ligase complex, is the target of the immunomodulatory drugs lenalidomide and pomalidomide. Recently, it was demonstrated that binding of these drugs to CRBN promotes the ubiquitination and subsequent degradation of 2 common substrates, transcription factors Aiolos and Ikaros. Here we report that CC-122, a new chemical entity termed pleiotropic pathway modifier, binds CRBN and promotes degradation of Aiolos and Ikaros in diffuse large B-cell lymphoma (DLBCL) and T cells in vitro, in vivo, and in patients, resulting in both cell autonomous as well as immunostimulatory effects. In DLBCL cell lines, CC-122-induced degradation or short hairpin RNA-mediated knockdown of Aiolos and Ikaros correlates with increased transcription of interferon (IFN)-stimulated genes independent of IFN-α, -ß, and -γ production and/or secretion and results in apoptosis in both activated B-cell (ABC) and germinal center B-cell DLBCL cell lines. Our results provide mechanistic insight into the cell-of-origin independent antilymphoma activity of CC-122, in contrast to the ABC subtype selective activity of lenalidomide.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Ikaros Transcription Factor/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Peptide Hydrolases/genetics , Piperidones/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/chemistry , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Ikaros Transcription Factor/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferons/genetics , Interferons/metabolism , Lenalidomide , Lentivirus/genetics , Lentivirus/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, SCID , Molecular Mimicry , Peptide Hydrolases/metabolism , Piperidones/chemistry , Proteolysis/drug effects , Quinazolinones/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
6.
Br J Haematol ; 172(6): 889-901, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26914976

ABSTRACT

Pomalidomide is an IMiD(®) immunomodulatory agent, which has shown clinically significant benefits in relapsed and/or refractory multiple myeloma (rrMM) patients when combined with dexamethasone, regardless of refractory status to lenalidomide or bortezomib. (Schey et al, ; San Miguel et al, 2013; Richardson et al, 2014; Scott, ) In this work, we present preclinical data showing that the combination of pomalidomide with dexamethasone (PomDex) demonstrates potent anti-proliferative and pro-apoptotic activity in both lenalidomide-sensitive and lenalidomide-resistant MM cell lines. PomDex also synergistically inhibited tumour growth compared with single-agent treatment in xenografts of lenalidomide-resistant H929 R10-1 cells. Typical hallmarks of IMiD compound activity, including IKZF3 (Aiolos) degradation, and the downregulation of interferon regulatory factor (IRF) 4 and MYC, seen in lenalidomide-sensitive H929 MM cell lines, were also observed in PomDex-treated lenalidomide-resistant H929 MM cells. Remarkably, this resulted in strong, synergistic effects on the induction of apoptosis in both lenalidomide-sensitive and resistant MM cells. Furthermore, gene expression profiling revealed a unique differential gene expression pattern in PomDex-treated samples, highlighted by the modulation of pro-apoptotic pathways in lenalidomide-resistant cells. These results provide key insights into molecular mechanisms of PomDex in the lenalidomide-resistant setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunomodulation/drug effects , Lenalidomide , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
7.
Blood ; 124(10): 1637-44, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-24990888

ABSTRACT

Lenalidomide has demonstrated clinical activity in patients with chronic lymphocytic leukemia (CLL), even though it is not cytotoxic for primary CLL cells in vitro. We examined the direct effect of lenalidomide on CLL-cell proliferation induced by CD154-expressing accessory cells in media containing interleukin-4 and -10. Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21). Silencing p21 with small interfering RNA impaired the capacity of lenalidomide to inhibit CLL-cell proliferation. Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. We isolated CLL cells from the blood of patients before and after short-term treatment with low-dose lenalidomide (5 mg per day) and found the leukemia cells were also induced to express p21 in vivo. These results indicate that lenalidomide can directly inhibit proliferation of CLL cells in a cereblon/p21-dependent but p53-independent manner, at concentrations achievable in vivo, potentially contributing to the capacity of this drug to inhibit disease-progression in patients with CLL.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Peptide Hydrolases/physiology , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing , Gene Expression Regulation, Leukemic/drug effects , HeLa Cells , Humans , Lenalidomide , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Signal Transduction/drug effects , Thalidomide/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/physiology , Ubiquitin-Protein Ligases
8.
Br J Haematol ; 164(2): 233-44, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24206017

ABSTRACT

Cereblon, a member of the cullin 4 ring ligase complex (CRL4), is the molecular target of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide and is required for the antiproliferative activity of these agents in multiple myeloma (MM) and immunomodulatory activity in T cells. Cereblon's central role as a target of lenalidomide and pomalidomide suggests potential utility as a predictive biomarker of response or resistance to IMiD therapy. Our studies characterized a cereblon monoclonal antibody CRBN65, with high sensitivity and specificity in Western analysis and immunohistochemistry that is superior to commercially available antibodies. We identified multiple cereblon splice variants in both MM cell lines and primary cells, highlighting challenges with conventional gene expression assays given this gene complexity. Using CRBN65 antibody and TaqMan quantitative reverse transcription polymerase chain reaction assays, we showed lack of correlation between cereblon protein and mRNA levels. Furthermore, lack of correlation between cereblon expression in MM cell lines and sensitivity to lenalidomide was shown. In cell lines made resistant to lenalidomide and pomalidomide, cereblon protein is greatly reduced. These studies show limitations to the current approaches of cereblon measurement that rely on commercial reagents and assays. Standardized reagents and validated assays are needed to accurately assess the role of cereblon as a predictive biomarker.


Subject(s)
Drug Resistance, Neoplasm/genetics , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Alternative Splicing , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Multiple Myeloma/drug therapy , Peptide Hydrolases/immunology , RNA Isoforms , Thalidomide/therapeutic use , Ubiquitin-Protein Ligases
9.
Nat Commun ; 13(1): 4121, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35840578

ABSTRACT

The myeloma surface proteome (surfaceome) determines tumor interaction with the microenvironment and serves as an emerging arena for therapeutic development. Here, we use glycoprotein capture proteomics to define the myeloma surfaceome at baseline, in drug resistance, and in response to acute drug treatment. We provide a scoring system for surface antigens and identify CCR10 as a promising target in this disease expressed widely on malignant plasma cells. We engineer proof-of-principle chimeric antigen receptor (CAR) T-cells targeting CCR10 using its natural ligand CCL27. In myeloma models we identify proteins that could serve as markers of resistance to bortezomib and lenalidomide, including CD53, CD10, EVI2B, and CD33. We find that acute lenalidomide treatment increases activity of MUC1-targeting CAR-T cells through antigen upregulation. Finally, we develop a miniaturized surface proteomic protocol for profiling primary plasma cell samples with low inputs. These approaches and datasets may contribute to the biological, therapeutic, and diagnostic understanding of myeloma.


Subject(s)
Multiple Myeloma , Drug Resistance , Humans , Immunotherapy/methods , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Proteomics , Tumor Microenvironment
10.
Br J Haematol ; 154(3): 325-36, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21707574

ABSTRACT

Overexpression of the transcription factor interferon regulatory factor-4 (IRF4), which is common in multiple myeloma (MM), is associated with poor prognosis. Patients with higher IRF4 expression have significantly poorer overall survival than those with low IRF4 expression. Lenalidomide is an IMiD immunomodulatory compound that has both tumouricidal and immunomodulatory activity in MM. This study showed that lenalidomide downregulated IRF4 levels in MM cell lines and bone marrow samples within 8 h of drug exposure. This was associated with a decrease in MYC levels, as well as an initial G1 cell cycle arrest, decreased cell proliferation, and cell death by day 5 of treatment. In eight MM cell lines, high IRF4 levels correlated with increased lenalidomide sensitivity. The clinical significance of this observation was investigated in 154 patients with MM. Among MM patients with high levels of IRF4 expression, treatment with lenalidomide led to a significantly longer overall survival than other therapies in a retrospective analysis. These data confirm the central role of IRF4 in MM pathogenesis; indicate that this is an important mechanism by which lenalidomide exerts its antitumour effects; and may provide a mechanistic biomarker to predict response to lenalidomide.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/biosynthesis , Interferon Regulatory Factors/biosynthesis , Multiple Myeloma/metabolism , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Cell Proliferation , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Genes, myc , Humans , Interferon Regulatory Factors/antagonists & inhibitors , Interferon Regulatory Factors/genetics , Lenalidomide , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prognosis , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction/methods , Thalidomide/pharmacology , Thalidomide/therapeutic use , Treatment Outcome , Tumor Cells, Cultured
11.
Blood ; 114(2): 338-45, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19417207

ABSTRACT

IMiDs immunomodulatory drugs, including lenalidomide and pomalidomide represent a novel class of small molecule anticancer and anti-inflammatory drugs with broad biologic activities. However, the molecular mechanism through which these drugs exert their effects is largely undefined. Using pomalidomide and primary human monocytes, we report that pomalidomide rapidly and selectively activated RhoA and Rac1, but not Cdc42 or Ras, in the absence of any costimulation. Consistent with the activation of Rho GTPases, we found that pomalidomide enhanced F-actin formation, stabilized microtubules, and increased cell migration, all of which were blocked by selective inhibitors of ROCK1 and Rac1. Further, we showed that in Swiss 3T3 cells, pomalidomide only activated RhoA, not Rac1 or Cdc42, and potently induced stress fiber formation. The pomalidomide effect on actin cytoskeleton was blocked by the ROCK1 inhibitor, but not Rac1 inhibitor. Finally, we demonstrated that pomalidomide was able to regulate the activity of Rho GTPases and the formation of F-actin in primary human T cells as it did in monocytes and showed that the activation of RhoA was essential for pomalidomide-induced interleukin-2 expression in T cells. These novel activities provide what we believe a critical mechanism by which IMiDs drugs function as therapeutic immunomodulatory agents.


Subject(s)
Cytoskeleton/drug effects , Cytoskeleton/enzymology , Immunosuppressive Agents/pharmacology , Thalidomide/analogs & derivatives , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/immunology , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/enzymology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Thalidomide/pharmacology
12.
J Med Chem ; 64(4): 1835-1843, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33591756

ABSTRACT

Acute myeloid leukemia (AML) is marked by significant unmet clinical need due to both poor survival and high relapse rates where long-term disease control for most patients with relapsed or refractory AML remain dismal. Inspired to bring novel therapeutic options to these patients, we envisioned protein degradation as a potential therapeutic approach for the treatment of AML. Following this course, we discovered and pioneered a novel mechanism of action which culminated in the discovery of CC-90009. CC-90009 represents a novel protein degrader and the first cereblon E3 ligase modulating drug to enter clinical development that specifically targets GSPT1 (G1 to S phase transition 1) for proteasomal degradation. This manuscript briefly summarizes the mechanism of action, scientific rationale, medicinal chemistry, pharmacokinetic properties, and efficacy data for CC-90009, which is currently in phase 1 clinical development.


Subject(s)
Acetamides/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/therapeutic use , Isoindoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Peptide Termination Factors/antagonists & inhibitors , Piperidones/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Acetamides/chemistry , Acetamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isoindoles/chemistry , Isoindoles/pharmacology , Macaca fascicularis , Male , Mice , Molecular Structure , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Piperidones/chemistry , Piperidones/pharmacology , Proteolysis/drug effects , Structure-Activity Relationship
13.
Blood ; 111(9): 4690-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18305219

ABSTRACT

Decreased p27(Kip1) levels are a poor prognostic factor in many malignancies, and can occur through up-regulation of SCF(Skp2) E3 ligase function, resulting in enhanced p27 ubiquitination and proteasome-mediated degradation. While proteasome inhibitors stabilize p27(Kip1), agents inhibiting SCF(Skp2) may represent more directly targeted drugs with the promise of enhanced efficacy and reduced toxicity. Using high-throughput screening, we identified Compound A (CpdA), which interfered with SCF(Skp2) ligase function in vitro, and induced specific accumulation of p21 and other SCF(Skp2) substrates in cells without activating a heat-shock protein response. CpdA prevented incorporation of Skp2 into the SCF(Skp2) ligase, and induced G(1)/S cell-cycle arrest as well as SCF(Skp2)- and p27-dependent cell killing. This programmed cell death was caspase-independent, and instead occurred through activation of autophagy. In models of multiple myeloma, CpdA overcame resistance to dexamethasone, doxorubicin, and melphalan, as well as to bortezomib, and also acted synergistically with this proteasome inhibitor. Importantly, CpdA was active against patient-derived plasma cells and both myeloid and lymphoblastoid leukemia blasts, and showed preferential activity against neoplastic cells while relatively sparing other marrow components. These findings provide a rational framework for further development of SCF(Skp2) inhibitors as a novel class of antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p27/physiology , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Cell Line, Tumor , Drug Delivery Systems/methods , Drug Resistance, Neoplasm , Humans , Multiple Myeloma/drug therapy
14.
J Med Chem ; 63(13): 6648-6676, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32130004

ABSTRACT

Many patients with multiple myeloma (MM) initially respond to treatment with modern combination regimens including immunomodulatory agents (lenalidomide and pomalidomide) and proteasome inhibitors. However, some patients lack an initial response to therapy (i.e., are refractory), and although the mean survival of MM patients has more than doubled in recent years, most patients will eventually relapse. To address this need, we explored the potential of novel cereblon E3 ligase modulators (CELMoDs) for the treatment of patients with relapsed or refractory multiple myeloma (RRMM). We found that optimization beyond potency of degradation, including degradation efficiency and kinetics, could provide efficacy in a lenalidomide-resistant setting. Guided by both phenotypic and protein degradation data, we describe a series of CELMoDs for the treatment of RRMM, culminating in the discovery of CC-92480, a novel protein degrader and the first CELMoD to enter clinical development that was specifically designed for efficient and rapid protein degradation kinetics.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Multiple Myeloma/drug therapy , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Inhibitory Concentration 50 , Mice , Multiple Myeloma/pathology , Recurrence , Stereoisomerism , Treatment Failure , Ubiquitin-Protein Ligases/antagonists & inhibitors , Xenograft Model Antitumor Assays
15.
Cancer Res ; 67(2): 746-55, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234786

ABSTRACT

Clinical studies involving patients with myelodysplastic syndromes or multiple myeloma have shown the efficacy of lenalidomide by reducing and often eliminating malignant cells while restoring the bone marrow function. To better understand these clinical observations, we investigated and compared the effects of lenalidomide and a structurally related analogue, CC-4047, on the proliferation of two different human hematopoietic cell models: the Namalwa cancer cell line and normal CD34+ progenitor cells. Both compounds had antiproliferative effects on Namalwa cells and pro-proliferative effects on CD34+ cells, whereas p21WAF-1 expression was up-regulated in both cell types. In Namalwa cells, the up-regulation of p21WAF-1 correlated well with the inhibition of cyclin-dependent kinase (CDK) 2, CDK4, and CDK6 activity leading to pRb hypophosphorylation and cell cycle arrest, whereas in CD34+ progenitor cells the increase of p21WAF-1 did not inhibit proliferation. Similarly, antiproliferation results were observed in two B lymphoma cell lines (LP-1 and U266) but interestingly not in normal B cells where a protection of apoptosis was found. Finally, CC-4047 and lenalidomide had synergistic effects with valproic acid [a histone deacetylase (HDAC) inhibitor] by increasing the apoptosis of Namalwa cells and enhancing CD34+ cell expansion. Our results indicate that lenalidomide and CC-4047 have opposite effects in tumor cells versus normal cells and could explain, at least in part, the reduction of malignant cells and the restoration of bone marrow observed in patients undergoing lenalidomide treatment. Moreover, this study provides new insights on the cellular pathways affected by lenalidomide and CC-4047, proposes new potential clinical uses, such as bone marrow regeneration, and suggests that the combination of lenalidomide or CC-4047 with certain HDAC inhibitors may elevate the therapeutic index in the treatment of hematologic malignancies.


Subject(s)
Antigens, CD34/biosynthesis , Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Hematopoietic Stem Cells/drug effects , Lymphoma, B-Cell/drug therapy , Thalidomide/analogs & derivatives , B-Lymphocytes/pathology , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Lenalidomide , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , Thalidomide/pharmacology , Up-Regulation/drug effects
16.
Mol Cell Biol ; 23(16): 5939-46, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897162

ABSTRACT

Genome integrity is protected by Cds1 (Chk2), a checkpoint kinase that stabilizes arrested replication forks. How Cds1 accomplishes this task is unknown. We report that Cds1 interacts with Rad60, a protein required for recombinational repair in fission yeast. Cds1 activation triggers Rad60 phosphorylation and nuclear delocalization. A Rad60 mutant that inhibits regulation by Cds1 renders cells specifically sensitive to replication fork arrest. Genetic and biochemical studies indicate that Rad60 functions codependently with Smc5 and Smc6, subunits of an SMC (structural maintenance of chromosomes) complex required for recombinational repair. These studies indicate that regulation of Rad60 is an important part of the replication checkpoint response controlled by Cds1. We propose that control of Rad60 regulates recombination events at stalled forks.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Recombination, Genetic , Schizosaccharomyces pombe Proteins/metabolism , Amino Acid Sequence , Cell Nucleus/metabolism , Cell Survival , Checkpoint Kinase 2 , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Immunoblotting , Models, Biological , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Saccharomycetales
17.
Appl Immunohistochem Mol Morphol ; 24(10): 695-702, 2016.
Article in English | MEDLINE | ID: mdl-26186254

ABSTRACT

Clinical interest in the measurement of Cereblon (CRBN), the primary target of the IMiDs immunomodulatory drugs lenalidomide and pomalidomide, has been fueled by its essential requirement for antitumor or immunomodulatory activity of both drugs in multiple myeloma (MM). However, limited analyses of clinical samples for CRBN gene expression or protein levels have utilized unvalidated reagents and assays, raising uncertainty about the interpretation of these results. We previously described a highly specific rabbit monoclonal antibody CRBN65 against 65-76 AA of human Cereblon. Here we describe a validated dual color bright-field Cereblon/CD138 immunohistochemical (IHC) assay utilizing CRBN65 and a commercial mouse monoclonal CD138 antibody. Sensitivity and specificity of the assay was determined and assay precision was shown for both cytoplasmic and nuclear Cereblon in MM bone marrow samples with coefficient of variation values of 5% and 2%, respectively. The dual IHC assay was effective for detecting a continuous range of Cereblon levels in 22 MM patient bone marrow core biopsies and aspirate clots, as shown by average cytoplasmic H-scores ranging from 63 to 267 and nuclear H-scores ranging from 17 to 250. Interpathologist comparison of MM sample H-scores by 3 pathologists demonstrated good concordance (R=0.73). This dual assay demonstrated superior Cereblon IHC measurement in MM samples compared with the single IHC assay using a published commercial rabbit polyclonal Cereblon antibody and could be used to explore the potential utility of Cereblon as a biomarker in the clinic.


Subject(s)
Biomarkers, Tumor/metabolism , Color , Immunohistochemistry/methods , Multiple Myeloma/metabolism , Peptide Hydrolases/metabolism , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , Humans , Ubiquitin-Protein Ligases
18.
Mol Cancer Ther ; 13(2): 504-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24431075

ABSTRACT

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth, proliferation, metabolism, and cell survival, and plays those roles by forming two functionally distinct multiprotein complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Deregulation of the mTOR pathway has been found in different cancers, including multiple myeloma. Agents acting on mTORC1, such as rapamycin and derivatives, are being explored as antitumoral strategies. However, whether targeting mTOR would be a more effective antimyeloma strategy than exclusively acting on the mTORC1 branch remains to be established. In this report, we explored the activation status of mTOR routes in malignant plasma cells, and analyzed the contribution of mTOR and its two signaling branches to the proliferation of myeloma cells. Gene expression profiling demonstrated deregulation of mTOR pathway-related genes in myeloma plasma cells from patients. Activation of the mTOR pathway in myelomatous plasma cells was corroborated by flow cytometric analyses. RNA interference (RNAi) experiments indicated that mTORC1 predominated over mTORC2 in the control of myeloma cell proliferation. However, mTOR knockdown had a superior antiproliferative effect than acting only on mTORC1 or mTORC2. Pharmacologic studies corroborated that the neutralization of mTOR has a stronger antimyeloma effect than the individual inhibition of mTORC1 or mTORC2. Together, our data support the clinical development of agents that widely target mTOR, instead of agents, such as rapamycin or its derivatives, that solely act on mTORC1.


Subject(s)
Multiple Myeloma/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Imidazoles/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Pyrazines/pharmacology , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured
19.
Nat Struct Mol Biol ; 21(9): 803-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108355

ABSTRACT

The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. These drugs directly bind Cereblon (CRBN) and promote the recruitment of substrates Ikaros (IKZF1) and Aiolos (IKZF3) to the E3 complex, thus leading to substrate ubiquitination and degradation. Here we present the crystal structure of human CRBN bound to DDB1 and the drug lenalidomide. A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. We also solved the structures of the mouse TBD in the apo state and with thalidomide or pomalidomide. Site-directed mutagenesis in lentiviral-expression myeloma models showed that key drug-binding residues are critical for antiproliferative effects.


Subject(s)
Angiogenesis Inhibitors/pharmacology , DNA-Binding Proteins/metabolism , Peptide Hydrolases/metabolism , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Animals , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Humans , Lenalidomide , Mice , Molecular Docking Simulation , Molecular Sequence Data , Peptide Hydrolases/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Thalidomide/chemistry , Thalidomide/pharmacology , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL