Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Analyst ; 149(11): 3178-3185, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38639441

ABSTRACT

In this application of native mass spectrometry (nMS) to investigate complexes formed by molecular glues (MGs), we have demonstrated its efficiency in delineating stoichiometric rearrangements of E3 ligases that occur during targeted protein degradation (TPD). MGs stabilise interactions between an E3 ligase and a protein of interest (POI) targeted for degradation, and these ternary interactions are challenging to characterise. We have shown that nMS can unambiguously identify complexes formed between the CRBN : DDB1 E3 ligase and the POI GSPT1 upon the addition of lenalidomide, pomalidomide or thalidomide. Ternary complex formation was also identified involving the DCAF15 : DDA1 : DDB1 E3 ligase in the presence of MG (E7820 or indisulam) and POI RBM39. Moreover, we uncovered that the DCAF15 : DDA1 : DDB1 E3 ligase self-associates into dimers and trimers when analysed alone at low salt concentrations (100 mM ammonium acetate) which dissociate into single copies of the complex at higher salt concentrations (500 mM ammonium acetate), or upon the addition of MG and POI, forming a 1 : 1 : 1 ternary complex. This work demonstrates the strength of nMS in TPD research, reveals novel binding mechanisms of the DCAF15 E3 ligase, and its self-association into dimers and trimers at reduced salt concentration during structural analysis.


Subject(s)
Mass Spectrometry , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/chemistry , Mass Spectrometry/methods , Thalidomide/chemistry , Thalidomide/analogs & derivatives , Humans , Lenalidomide/chemistry , Protein Multimerization , Protein Binding
2.
J Med Chem ; 66(9): 6037-6046, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37083375

ABSTRACT

Targeted protein degradation is a promising therapeutic strategy, spearheaded by the anti-myeloma drugs lenalidomide and pomalidomide. These drugs stabilize very efficiently the complex between the E3 ligase Cereblon (CRBN) and several non-native client proteins (neo-substrates), including the transcription factors Ikaros and Aiolos and the enzyme Caseine Kinase 1α (CK1α,), resulting in their degradation. Although the structures for these complexes have been determined, there are no evident interactions that can account for the high efficiency of formation of the ternary complex. We show that lenalidomide's stabilization of the CRBN-CK1α complex is largely due to hydrophobic shielding of intermolecular hydrogen bonds. We also find a quantitative relationship between hydrogen bond robustness and binding affinities of the ternary complexes. These results pave the way to further understand cooperativity effects in drug-induced protein-protein complexes and could help in the design of improved molecular glues and more efficient protein degraders.


Subject(s)
Multiple Myeloma , Humans , Lenalidomide/pharmacology , Lenalidomide/chemistry , Multiple Myeloma/drug therapy , Ubiquitin-Protein Ligases/metabolism , Proteolysis , Transcription Factors/metabolism , Peptide Hydrolases/metabolism
3.
Bioorg Chem ; 127: 105923, 2022 10.
Article in English | MEDLINE | ID: mdl-35717803

ABSTRACT

Molecular glue degraders, such as lenalidomide and pomalidomide, bind to cereblon (CRBN) E3 ligase and subsequently recruit neosubstrate proteins, Ikaros (IKZF1) and Aiolos (IKZF3), for the ubiquitination-proteasomal degradation process. In this study, we explored structure-activity relationship analysis for novel GSPT1 degraders utilizing a benzotriazinone scaffold previously discovered as a novel CRBN binder. In particular, we focused on the position of the ureido group on the benzotriazinone scaffold, substituent effect on the phenylureido group, and methyl substitution on the benzylic position of benzotriazinone. As a result, we identified 34f (TD-522), which exhibits strong anti-proliferative effects in both KG-1 (EC50 = 0.5 nM) and TMD-8 (EC50 = 5.2 nM) cell lines. Compound 34f effectively induced GSPT1 degradation with a DC50 of 0.269 nM and Dmax of >95 % at 10 nM concentration in KG-1 cells. An in vivo xenograft study showed that compound 34f effectively suppressed TMD8-driven tumor growth, suggesting a potential role in the development of novel GSPT1 degraders.


Subject(s)
Adaptor Proteins, Signal Transducing , Animals , Disease Models, Animal , Heterografts , Humans , Lenalidomide/chemistry , Lenalidomide/pharmacology , Mice , Proteolysis , Structure-Activity Relationship
4.
Bioorg Chem ; 119: 105508, 2022 02.
Article in English | MEDLINE | ID: mdl-34959180

ABSTRACT

Clinical FLT3 mutations caused poor therapeutic benefits toward the present FLT3 inhibitors, and degradation of the FLT3 mutant protein may be a promising alternative approach to protect against acute myeloid leukemia (AML). Herein, we report the discovery of small molecule FLT3 degraders based on the proteolysis targeting chimera (PROTAC). FLT3 degraders were designed, synthesized, and evaluated for FLT3 degradation. Promising PF15 significantly inhibited the proliferation of FLT3-ITD-positive cells, induced FLT3 degradation and downregulated the phosphorylation of FLT3 and STAT5. An in vivo xenograft model and survival period evaluation verified the efficacy of PROTAC. These findings laid a robust foundation for FLT3-PROTAC molecules as an effective strategy for treating AML.


Subject(s)
Antineoplastic Agents/pharmacology , Lenalidomide/pharmacology , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Lenalidomide/chemical synthesis , Lenalidomide/chemistry , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Structure , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proteolysis/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
5.
Drug Des Devel Ther ; 15: 5035-5059, 2021.
Article in English | MEDLINE | ID: mdl-34949914

ABSTRACT

INTRODUCTION: With the aim of repositioning commercially available drugs for the inhibition of the anti-apoptotic myeloid cell leukemia protein, Mcl-1, implied in various cancers, five molecules, highlighted from a published theoretical screening, were selected to experimentally validate their affinity toward Mcl-1. RESULTS: A detailed NMR study revealed that only two of the five tested drugs, Torsemide and Deferasirox, interacted with Mcl-1. NMR data analysis allowed the complete characterization of the binding mode of both drugs to Mcl-1, including the estimation of their affinity for Mcl-1. Biological assays evidenced that the biological activity of Torsemide was lower as compared to the Deferasirox, which was able to efficiently and selectively inhibit the anti-apoptotic activity of Mcl-1. Finally, docking and molecular dynamics led to a 3D model for the Deferasirox:Mcl-1 complex and revealed the positioning of the drug in the Mcl-1 P2/P3 pockets as well as almost all synthetic Mcl-1 inhibitors. Interestingly, contrary to known synthetic Mcl-1 inhibitors which interact through Arg263, Deferasirox, establishes a salt bridge with Lys234. CONCLUSION: Deferasirox could be a potential candidate for drug repositioning as Mcl-1 inhibitor.


Subject(s)
Apoptosis Regulatory Proteins/drug effects , Deferasirox/pharmacology , Drug Repositioning , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Deferasirox/chemistry , Lenalidomide/chemistry , Lenalidomide/pharmacology , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Oxcarbazepine/chemistry , Oxcarbazepine/pharmacology , Risperidone/chemistry , Risperidone/pharmacology , Torsemide/chemistry , Torsemide/pharmacology
6.
Nat Commun ; 12(1): 5263, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489457

ABSTRACT

Immunomodulatory drugs (IMiDs) are important for the treatment of multiple myeloma and myelodysplastic syndrome. Binding of IMiDs to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase, induces cancer cell death by targeting key neo-substrates for degradation. Despite this clinical significance, the physiological regulation of CRBN remains largely unknown. Herein we demonstrate that Wnt, the extracellular ligand of an essential signal transduction pathway, promotes the CRBN-dependent degradation of a subset of proteins. These substrates include Casein kinase 1α (CK1α), a negative regulator of Wnt signaling that functions as a key component of the ß-Catenin destruction complex. Wnt stimulation induces the interaction of CRBN with CK1α and its resultant ubiquitination, and in contrast with previous reports does so in the absence of an IMiD. Mechanistically, the destruction complex is critical in maintaining CK1α stability in the absence of Wnt, and in recruiting CRBN to target CK1α for degradation in response to Wnt. CRBN is required for physiological Wnt signaling, as modulation of CRBN in zebrafish and Drosophila yields Wnt-driven phenotypes. These studies demonstrate an IMiD-independent, Wnt-driven mechanism of CRBN regulation and provide a means of controlling Wnt pathway activity by CRBN, with relevance for development and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Peptide Hydrolases/genetics , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway/physiology , Zebrafish Proteins/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Casein Kinase Ialpha/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Evolution, Molecular , HEK293 Cells , Humans , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Lenalidomide/chemistry , Lenalidomide/pharmacology , Mice , Organoids , Peptide Hydrolases/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism
7.
Eur J Med Chem ; 223: 113645, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34217059

ABSTRACT

Protein degradation is a promising strategy for drug development. Proteolysis-targeting chimeras (PROTACs) hijacking the E3 ligase cereblon (CRBN) exhibit enormous potential and universal degradation performance due to the small molecular weight of CRBN ligands. In this study, the CRBN-recruiting PROTACs were explored on the degradation of oncogenic fusion protein BCR-ABL, which drives the pathogenesis of chronic myeloid leukemia (CML). A series of novel PROTACs were synthesized by conjugating BCR-ABL inhibitor dasatinib to the CRBN ligand including pomalidomide and lenalidomide, and the extensive structure-activity relationship (SAR) studies were performed focusing on optimization of linker parameters. Therein, we uncovered that pomalidomide-based degrader 17 (SIAIS056), possessing sulfur-substituted carbon chain linker, exhibits the most potent degradative activity in vitro and favorable pharmacokinetics in vivo. Besides, degrader 17 also degrades a variety of clinically relevant resistance-conferring mutations of BCR-ABL. Furthermore, degrader 17 induces significant tumor regression against K562 xenograft tumors. Our study indicates that 17 as an efficacious BCR-ABL degrader warrants intensive investigation for the future treatment of BCR-ABL+ leukemia.


Subject(s)
Drug Design , Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Ubiquitin-Protein Ligases/chemistry , Animals , Cell Proliferation/drug effects , Dasatinib/pharmacology , Fusion Proteins, bcr-abl/metabolism , Half-Life , Humans , K562 Cells , Lenalidomide/chemistry , Lenalidomide/metabolism , Ligands , Mice , Neoplasms/drug therapy , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proteolysis , Structure-Activity Relationship , Thalidomide/analogs & derivatives , Thalidomide/chemistry , Thalidomide/metabolism , Transplantation, Heterologous , Ubiquitin-Protein Ligases/metabolism
8.
Eur J Med Chem ; 209: 112912, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33328101

ABSTRACT

Lenalidomide and its analogs are well-known for treating multiple myeloma. In this work, designed sulfide-modified lenalidomide and pomalidomide were synthesized and evaluated. The anti-proliferative activity against MM.1S cell line of 3ak (IC50 = 79 nM) was similar to lenalidomide (IC50 = 81 nM). Compared to benzylic thioether substituted lenalidomide 3a, the half-live (T1/2) of 4-F-phenyl-thioether analogs 3ak in human liver microsomes was promoted from 3 min to 416.7 min. The corresponding metabolic factor of 3ak was increased from 2.8% to 79.5%, which was slightly lower than lenalidomide (91.5%). Moreover, the IKZF1 degradation of 3y and 3ak was well related with corresponding IC50 values, which suggested the IKZF1 degradation efficiency is correlated to the responses of MM1. S cells. Furthermore, the oral administration of compounds 3y and 3ak at dosages of 60 mg/kg could delay tumor growth in female CB-17 SCID mice. This research helped to prompt the stability of thioether lenalidomide analogs, which paved the way for developing better molecules for treating multiple myeloma.


Subject(s)
Drug Design , Lenalidomide/chemistry , Lenalidomide/pharmacology , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Animals , Cell Proliferation/drug effects , Female , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Lenalidomide/chemical synthesis , Lenalidomide/therapeutic use , Mice , Mice, SCID , Sulfides/chemistry , Thalidomide/chemical synthesis , Thalidomide/chemistry , Thalidomide/pharmacology , Thalidomide/therapeutic use , Xenograft Model Antitumor Assays
9.
Int J Nanomedicine ; 15: 6873-6886, 2020.
Article in English | MEDLINE | ID: mdl-32982239

ABSTRACT

PURPOSE: In a previous study, we demonstrated that the combination of fenretinide with lenalidomide, administered by a novel nanomicellar formulation (FLM), provided a strong antitumor effect in a neuroblastoma TrkB-expressing tumor. In this study, we tested the nanomicellar combination in an MYCN amplified neuroblastoma xenograft to assess its efficacy in different tumor genotypes and evaluate the interactions of the nanomicelles with the tumor cells. EXPERIMENTAL DESIGN: FLM was administered to mice bearing human NLF xenografts to evaluate its efficacy in comparison with the nanomicelles containing fenretinide alone (FM). Confocal laser-scanning fluorescence microscopy images of the NLF cells treated with FLM and FM allowed us to estimate the nanomicelle ability to transport the encapsulated drugs inside the tumor cells. Flow cytometric analysis of the cells from treated tumors was performed to assess the effect of treatment on GD2 expression and NK cell infiltration. RESULTS: FLM and FM decreased the growth of NLF xenografts at comparable extents during the treatment period. Afterwards, FLM induced a progressive tumor regression without regrowth, while FM treatment was followed by regrowth within 15-20 days after the end of treatment. Both FLM and FM were able to penetrate the tumor cells transporting the encapsulated drugs. FLM transported higher amount of fenretinide inside the cells. Also, FLM treatment strongly increased GD2 expression in treated tumors and slightly decreased the NK infiltration compared to FM. CONCLUSION: FLM treatment induced a superior antitumor response than FM in NLF xenografts, presumably due to the combined effects of fenretinide cytotoxicity and lenalidomide antiangiogenic activity. The ability of FLM to penetrate tumor cells, transporting the encapsulated drugs, substantially improved the therapeutic efficiency of this system. Moreover, the enhancement of GD2 expression in FLM treated tumors offers the possibility to further increase the antitumor effect by the use of anti-GD2 CAR-T cells and anti-GD2 antibodies in combination with FLM in multimodal therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Fenretinide/administration & dosage , Fenretinide/chemistry , Gene Expression Regulation, Neoplastic , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Lenalidomide/administration & dosage , Lenalidomide/chemistry , Mice, Nude , Micelles , Microscopy, Confocal , Nanostructures/chemistry , Neuroblastoma/genetics , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
10.
Chemistry ; 26(70): 16818-16823, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32706492

ABSTRACT

A palladium-catalysed Buchwald-Hartwig amination for lenalidomide-derived aryl bromides was optimised using high throughput experimentation (HTE). The substrate scope of the optimised conditions was evaluated for a range of alkyl- and aryl- amines and functionalised aryl bromides. The methodology allows access to new cereblon-based bifunctional proteolysis targeting chimeras with a reduced step count and improved yields.


Subject(s)
Amines/chemistry , Bromides/chemistry , Lenalidomide/chemistry , Proteolysis/drug effects , Amination , Ligands , Ubiquitin-Protein Ligases/metabolism
11.
Cell Chem Biol ; 27(7): 866-876.e8, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32413286

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) is a paradigm shift for small-molecule drug discovery. However, limited E3 ubiquitin ligase ligands with cellular activity are available. In vitro binding assays involve the expression and purification of a large amount of proteins and they often yield ligands that are inactive in cell-based assays due to poor cell permeability, stability, and other reasons. Herein, we report the development of a practical and efficient cell-based target engagement assay to evaluate the binding affinity of a small library of cereblon ligands to its E3 ligase in cells. Selected cell-permeable E3 ligase ligands derived from this assay are then used to construct HDAC6 degraders with cellular protein degradation activity. Because the assay does not involve any genetic engineering, it is relatively easy to transfer from one cell type to a different one.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Histone Deacetylase 6/metabolism , Ligands , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Cell Line , Histone Deacetylase 6/antagonists & inhibitors , Humans , Lenalidomide/chemistry , Lenalidomide/metabolism , Protein Binding , Proteolysis , Thalidomide/analogs & derivatives , Thalidomide/chemistry , Thalidomide/metabolism , Ubiquitin-Protein Ligases/chemistry
12.
Bioorg Med Chem Lett ; 30(12): 127167, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32317208

ABSTRACT

Epidermal growth factor receptor (EGFR) is one of the important and valuable drug targets. Overexpression of EGFR is associated with the development of many types of cancer. In this study, three PROTACs small molecules (16a-16c) were designed, synthesized and evaluated for their cytotoxicity against the growth in different NSCLC cell line and the degradation effect. The bioassay results indicated that 16c has a good inhibition in PC9 cells and H1975 cells, and the corresponding IC50 value was 0.413 µM and 0.657 µM, respectively. Western blotting results demonstrated that compound 16c could serve as an effective EGFRdel19-targeting degrader in PC9 cells.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Chimera/metabolism , Lenalidomide/pharmacology , Lung Neoplasms/drug therapy , Acrylamides/chemistry , Amino Acid Sequence , Aniline Compounds/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , ErbB Receptors/metabolism , Humans , Lenalidomide/chemistry , Protein Binding , Protein Conformation , Proteolysis , Ubiquitin-Protein Ligases/metabolism
13.
Arch Pharm (Weinheim) ; 353(7): e1900376, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32342558

ABSTRACT

Lenalidomide is a cereblon modulator known for its antitumor, anti-inflammatory, and immunomodulatory properties in clinical applications. Recently, some reported lenalidomide analogs could exhibit a significant bioactivity through various modifications in the isoindolinone ring. In this study, we designed and synthesized a series of novel lenalidomide analogs on the basis of the installation of a methylene chain at the C-4 position of isoindolinone via the Suzuki cross-coupling reaction. These new compounds were further evaluated for their in vitro antiproliferative activities against two tumor cell lines (MM.1S and Mino). Specifically, compound 4c displayed the strongest antiproliferative activity against the MM.1S (IC50 = 0.27 ± 0.03 µM) and Mino (IC50 = 5.65 ± 0.58 µM) tumor cell lines. In summary, we have developed a new synthetic strategy for C-4 derivatization of lenalidomide, providing a bioactive scaffold that could be used to discover further potential antitumor lead compounds in pharmaceutical research.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Lenalidomide/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Lenalidomide/chemical synthesis , Lenalidomide/chemistry , Molecular Structure , Structure-Activity Relationship
14.
Bioorg Med Chem ; 28(1): 115228, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31813613

ABSTRACT

BRD4 has emerged as an attractive target for anticancer therapy. However, BRD4 inhibitors treatment leads to BRD4 protein accumulation, together with the reversible nature of inhibitors binding to BRD4, which may limit the efficacy of BRD4 inhibitors. To address these problems, a protein degradation strategy based on the proteolysis targeting chimera (PROTAC) technology has been developed to target BRD4 recently. Herein, we present our design, synthesis and biological evaluation of a new class of PROTAC BRD4 degraders, which were based on a potent dihydroquinazolinone-based BRD4 inhibitor compound 6 and lenalidomide/pomalidomide as ligand for E3 ligase cereblon. Gratifyingly, several compounds showed excellent inhibitory activity against BRD4, and high anti-proliferative potency against human monocyte lymphoma cell line THP-1. Especially, compound 21 (BRD4 BD1, IC50 = 41.8 nM) achieved a submicromolar IC50 value of 0.81 µM in inhibiting the growth of THP-1 cell line, and was 4 times more potent than compound 6. Moreover, the mechanism study established that 21 could effectively induce the degradation of BRD4 protein and suppression of c-Myc. All of these results suggested that 21 was an efficacious BRD4 degrader for further investigation.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Discovery , Lenalidomide/pharmacology , Quinazolinones/pharmacology , Thalidomide/analogs & derivatives , Transcription Factors/antagonists & inhibitors , Cell Line , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Lenalidomide/chemical synthesis , Lenalidomide/chemistry , Models, Molecular , Molecular Structure , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Structure-Activity Relationship , THP-1 Cells , Thalidomide/chemical synthesis , Thalidomide/chemistry , Thalidomide/pharmacology
15.
Pharm Dev Technol ; 24(9): 1175-1180, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31282833

ABSTRACT

Purpose: Lenalidomide (LDM) is a blockbuster drug for multiple myeloma and non-Hodgkin's lymphoma, and contributed $ 6.974 billion in sales for Celgene in 2016. The aim of this research is to expand the crystal form landscape, characterize the physicochemical properties and thoroughly investigate the potential solid forms transformation for this famous drug. Materials and methods: In this study, a comprehensive solid-state screening was carried out. The physicochemical properties, stability and phase transformation were fully investigated using powder X-ray diffraction (PXRD), thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), solid state nuclear magnetic resonance (solid state NMR) and Infrared Spectroscopic Analysis (IR). Finally the differences of dissolution behavior were compared through powder dissolution test. Results: Two new anhydrous forms (α and ß) and one new dihydrate form (DH) of LDM were discovered through a comprehensive solid-state screening experiment. The new discovered DH showed better stability under accelerated storage condition (40 °C/75% RH) and in most organic solvents than other forms. The new discovered form α exhibited faster dissolution rate in the early phase and larger apparent solubility than the currently marketed form. Conclusions: These new forms exhibit a new chance for drug development in view of their pharmaceutical properties and intellectual property.


Subject(s)
Angiogenesis Inhibitors/chemistry , Lenalidomide/chemistry , Crystallization , Drug Liberation , Phase Transition , Powder Diffraction , Powders , Solubility , Thermogravimetry , Water/chemistry , X-Ray Diffraction
16.
J Sep Sci ; 41(20): 3840-3847, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30095219

ABSTRACT

Semipreparative separation of lenalidomide has been performed through supercritical fluid chromatography. In regard to retention and resolution of lenalidomide, effects of chromatographic conditions, such as chiral stationary phases, organic co-solvents, mobile phases, and column temperature, have been studied in detail. Amylose tris(3, 5-dimethylphenylcarbamate)-coated and the single-urea-bound ß-cyclodextrin chiral stationary phases exhibited good separation performances for lenalidomide in the CO2 /methanol mixture. Then, a comparative study of semipreparative separation of lenalidomide has been carried out on these two chiral stationary phases. As indicated, separation of lenalidomide on the ß-cyclodextrin-bound column was much better than the other. Under the optimized conditions, the loading per injection was 30 mg, the cycle time was 5 min, and the recoveries of two enantiomers were about 81.7 and 79.5%, respectively. Moreover, the vibrational circular dichroism spectrum of the first-eluted enantiomer in d6 -dimethylsulfoxide solution was consistent with the calculated pattern based on the S configuration, revealing that it should be (S)-(-)-lenalidomide. Briefly, this separation method through supercritical fluid chromatography might provide favorable information for rapid separation, enantioselective assessment, and absolute configurations of chiral pharmaceuticals.


Subject(s)
Angiogenesis Inhibitors/isolation & purification , Angiogenesis Inhibitors/therapeutic use , Chromatography, Supercritical Fluid , Lenalidomide/isolation & purification , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Angiogenesis Inhibitors/chemistry , Circular Dichroism , Lenalidomide/chemistry , Vibration
17.
Blood ; 132(14): 1535-1544, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30064974

ABSTRACT

Thalidomide and its derivatives, lenalidomide and pomalidomide, are clinically effective treatments for multiple myeloma and myelodysplastic syndrome with del(5q). These molecules lack activity in murine models, limiting investigation of their therapeutic activity or toxicity in vivo. Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). In human cells, thalidomide and its analogs bind CRBN and recruit protein targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and subsequent degradation by the proteasome. We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1α), both in vitro and in vivo. We use the Crbn I391V model to demonstrate that the in vivo therapeutic activity of lenalidomide in del(5q) myelodysplastic syndrome can be explained by heterozygous expression of Ck1α in del(5q) cells. We found that lenalidomide acts on hematopoietic stem cells with heterozygous expression of Ck1α and inactivation of Trp53 causes lenalidomide resistance. We further demonstrate that Crbn I391V is sufficient to confer thalidomide-induced fetal loss in mice, capturing a major toxicity of this class of drugs. Further study of the Crbn I391V model will provide valuable insights into the in vivo efficacy and toxicity of this class of drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Lenalidomide/pharmacology , Myelodysplastic Syndromes/drug therapy , Nerve Tissue Proteins/genetics , Point Mutation , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/chemistry , Casein Kinase I/metabolism , Disease Models, Animal , Female , Hematopoiesis/drug effects , Lenalidomide/chemistry , Male , Mice , Mice, Inbred C57BL , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Nerve Tissue Proteins/metabolism , Thalidomide/analogs & derivatives
18.
J Med Chem ; 61(2): 535-542, 2018 01 25.
Article in English | MEDLINE | ID: mdl-28425720

ABSTRACT

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.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Ikaros Transcription Factor/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Proteolysis/drug effects , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Fluorescence Resonance Energy Transfer , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/metabolism , Humans , Lenalidomide/chemistry , Lenalidomide/metabolism , Morpholines , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Phthalimides , Piperidones , Protein Binding , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...