Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell ; 186(21): 4528-4545.e18, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37788669

ABSTRACT

MLL/KMT2A amplifications and translocations are prevalent in infant, adult, and therapy-induced leukemia. However, the molecular contributor(s) to these alterations are unclear. Here, we demonstrate that histone H3 lysine 9 mono- and di-methylation (H3K9me1/2) balance at the MLL/KMT2A locus regulates these amplifications and rearrangements. This balance is controlled by the crosstalk between lysine demethylase KDM3B and methyltransferase G9a/EHMT2. KDM3B depletion increases H3K9me1/2 levels and reduces CTCF occupancy at the MLL/KMT2A locus, in turn promoting amplification and rearrangements. Depleting CTCF is also sufficient to generate these focal alterations. Furthermore, the chemotherapy doxorubicin (Dox), which associates with therapy-induced leukemia and promotes MLL/KMT2A amplifications and rearrangements, suppresses KDM3B and CTCF protein levels. KDM3B and CTCF overexpression rescues Dox-induced MLL/KMT2A alterations. G9a inhibition in human cells or mice also suppresses MLL/KMT2A events accompanying Dox treatment. Therefore, MLL/KMT2A amplifications and rearrangements are controlled by epigenetic regulators that are tractable drug targets, which has clinical implications.


Subject(s)
Epigenesis, Genetic , Myeloid-Lymphoid Leukemia Protein , Adult , Animals , Humans , Infant , Mice , Doxorubicin/pharmacology , Gene Rearrangement , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia/metabolism , Lysine/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Translocation, Genetic
2.
J Biol Chem ; 299(6): 104825, 2023 06.
Article in English | MEDLINE | ID: mdl-37196766

ABSTRACT

Aberrant overexpression of nonreceptor tyrosine kinase FER (Fps/Fes Related) has been reported in various ovarian carcinoma-derived tumor cells and is a poor prognosis factor for patient survival. It plays an essential role in tumor cell migration and invasion, acting concurrently in both kinase-dependent and -independent manners, which is not easily suppressed by conventional enzymatic inhibitors. Nevertheless, the PROteolysis-TArgeting Chimera (PROTAC) technology offers superior efficacy over traditional activity-based inhibitors by simultaneously targeting enzymatic and scaffold functions. Hence in this study, we report the development of two PROTAC compounds that promote robust FER degradation in a cereblon-dependent manner. Both PROTAC degraders outperform a Food and Drug Administration-approved drug, brigatinib, in ovarian cancer cell motility suppression. Importantly, these PROTAC compounds also degrade multiple oncogenic FER fusion proteins identified in human tumor samples. These results lay an experimental foundation to apply the PROTAC strategy to antagonize cell motility and invasiveness in ovarian and other types of cancers with aberrant expression of FER kinase and highlight PROTACs as a superior strategy for targeting proteins with multiple tumor-promoting functions.


Subject(s)
Ovarian Neoplasms , Protein-Tyrosine Kinases , Humans , Female , Protein-Tyrosine Kinases/metabolism , Proteolysis Targeting Chimera , Proteins/metabolism , Ovarian Neoplasms/drug therapy , Cell Movement , Proteolysis
3.
Eur J Med Chem ; 247: 115016, 2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36577219

ABSTRACT

CRBN E3 ligase modulators, also anteriorly called immunomodulatory drugs (IMiDs), exhibit excellent pharmacological activity by degrading cereblon (CRBN) associated multiple substrates and have become an important field for drug development. These modulators such as Thalidomide, Lenalidomide and CC-122 abduct CRBN to adhere to IKZF1/3 and other neosubstrates, and then induce the degradation of these substrates, thus retarding the further development of related diseases. Herein, we reported a series of CC-122 derivatives that inhibit the proliferation of hematological malignant tumor cell lines. Studies further confirmed that several derivatives which exhibit strong anti-proliferation effect induce the significant degradation of IKZF1/3. In addition, we found that the best compound 14 (SIAIS355035) exhibits better degradation activity and better anti-proliferation activities than CC-122, especially in diffuse large B lymphoma cell lines. Moreover, the PK properties of compound 14 are pretty promising with excellent oral bioavailability. These results clarified the SAR of CC-122 derivatives preliminarily and suggested that compound 14 has great value for further studies as an ideal novel CRBN E3 ligase modulation drug.


Subject(s)
Peptide Hydrolases , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Peptide Hydrolases/metabolism , Thalidomide , Lenalidomide , Ubiquitination , Transcription Factors/metabolism
4.
Theranostics ; 12(17): 7476-7490, 2022.
Article in English | MEDLINE | ID: mdl-36438482

ABSTRACT

Rationale: Primary and acquired resistance to Smoothened (Smo) inhibitors largely hampered their clinical efficacy. Given the important functions of hedgehog (Hh) pathway in bone formation and development, the permanent defects in bone growth caused by Smo inhibitors further restrict the use of Smo inhibitors for pediatric tumor patients. Anti-apoptotic Bcl-2 proteins regulate Hh activity by engaging a Bcl-2 homology (BH) domain sequence found in suppressor of fused (Sufu). In this study, we tested the effect of SIAIS361034, a Proteolysis Targeting Chimera (PROTAC) specifically targeting B-cell lymphoma extra large (Bcl-xL) to the celeblon (CRBN) E3 ligase for degradation, on combating the resistance and reducing the toxicity of bone growth caused by Hh inhibition. Methods: Fluorescence polarization, homogeneous time-resolved fluorescence (HTRF) assay, immunoblot, and immunoprecipitation (IP) were used to evaluate whether SIAIS361034 is an appropriate Bcl-xL PROTAC. Dual luciferase reporter assay, real-time quantitative PCR (RT-qPCR), depilatory model, and SmoA1 model were established to assess the effect of SIAIS361034 on the activity of Hh signaling pathway and its ability to overcome drug resistance in vitro and in vivo. Molecular mechanisms of SIAIS361034 for inhibiting Hh activity were demonstrated by dual luciferase reporter assay, immunoblot, and immunofluorescence staining. PET-CT and histopathology of bone tissues were used to assess the effects of SIAIS361034 on bone growth. Results: We observed that SIAIS361034 efficiently and selectively inhibits the activity of the Hh pathway in vitro and in vivo, by interrupting Bcl-xL/Sufu interaction, therefore, promoting the interaction of Sufu with Gli1. Moreover, SIAIS361034 possesses the ability of combating resistance to current Smo inhibitors caused by Smo mutations and Gli2 amplification and remarkably inhibits the growth of SmoA1 tumors in vivo. In contrast to von Hippel-Lindau (VHL) E3 ligase, our result further reveals little detectable expression of CRBN in two types of cells critical for bone development, human articular chondrocytes and human fetal osteoblastic cells. Moreover, treatment with SIAIS361034 results in no impairment on the bone growth of young mice, accompanying no alteration of the expression of Bcl-xL and Gli1 proteins. Conclusion: Our findings demonstrate that selectively targeting Bcl-xL by PROTAC is a promising strategy for combating resistance to Smo inhibitors without causing on-target drug toxicities of bone growth.


Subject(s)
Antineoplastic Agents , Neoplasms , Child , Humans , Mice , Animals , Hedgehog Proteins/metabolism , Zinc Finger Protein GLI1/genetics , Proteolysis , Positron Emission Tomography Computed Tomography , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Bone Development , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Nat Commun ; 13(1): 7133, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36414666

ABSTRACT

The rational design of PROTACs is difficult due to their obscure structure-activity relationship. This study introduces a deep neural network model - DeepPROTACs to help design potent PROTACs molecules. It can predict the degradation capacity of a proposed PROTAC molecule based on structures of given target protein and E3 ligase. The experimental dataset is mainly collected from PROTAC-DB and appropriately labeled according to the DC50 and Dmax values. In the model of DeepPROTACs, the ligands as well as the ligand binding pockets are generated and represented with graphs and fed into Graph Convolutional Networks for feature extraction. While SMILES representations of linkers are fed into a Bidirectional Long Short-Term Memory layer to generate the features. Experiments show that DeepPROTACs model achieves 77.95% average prediction accuracy and 0.8470 area under receiver operating characteristic curve on the test set. DeepPROTACs is available online at a web server ( https://bailab.siais.shanghaitech.edu.cn/services/deepprotacs/ ) and at github ( https://github.com/fenglei104/DeepPROTACs ).


Subject(s)
Deep Learning , Neural Networks, Computer , Proteins , Ubiquitin-Protein Ligases/metabolism
6.
Chem Commun (Camb) ; 58(72): 10072-10075, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-35993284

ABSTRACT

By introducing a reactive oxygen species (ROS) triggered leaving group (arylboronic acid) to the parent PROTACs, ROS-responsive Pre-PROTACs were designed and evaluated. Pre-PROTAC (7) efficiently degraded the target protein BRD3 according to ROS levels. Our research provides an effective approach to control PROTAC activation by the endogenous ROS-related microenvironment.


Subject(s)
Neoplasms , Proteolysis , Humans , Neoplasm Proteins , Reactive Oxygen Species , Tumor Microenvironment
7.
J Med Chem ; 64(13): 9152-9165, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34138566

ABSTRACT

Proteolysis-targeting chimera (PROTAC) is an attractive technology in drug discovery. Canonically, targets act as a basic starting point in the most previous PROTAC design. Here, we designed degraders considering from the view of clinical benefits. With this novel design, Brigatinib was turned into a degrader SIAIS164018 and endowed with unique features. First, SIAIS164018 could degrade not only ALK fusion proteins in activating or G1202R-mutated form but also mutant EGFR with L858R + T790M, which are two most important targets in non-small-cell lung cancer. Second, SIAIS164018 strongly inhibited cell migration and invasion of Calu-1 and MDA-MB-231. Third and surprisingly, SIAIS164018 degrades several important oncoproteins involved in metastasis such as FAK, PYK2, and PTK6. Interestingly, SIAIS164018 reshuffled the kinome ranking profile when compared to Brigatinib. Finally, SIAIS164018 is orally bioavailable and well tolerated in vivo. SIAIS164018 is an enlightening degrader for us to excavate the charm of protein degradation.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Anaplastic Lymphoma Kinase/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Org Biomol Chem ; 19(1): 166-170, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33226388

ABSTRACT

As a promising protein degradation strategy, PROTAC technology is increasingly becoming a new star in cancer treatment. Here we report the efficient construction of an IMiD-based azide library via a quick one-step conversion of the existing IMiD-based amine library. This new azide library can act as a kit to endow PROTAC libraries with triazole moieties for various POIs through a highly effective 'click reaction' and then help to rapidly screen out lead degraders that are valuable for drug development. Its power in fleetly identifying potent degraders has been verified on two oncogenic proteins, BCR-ABL and BET, the degraders of which showed comparable potency to or even higher potency than the reported PROTACs in degrading target proteins and effectively inhibiting cancer cell proliferation.

9.
J Med Chem ; 62(20): 9281-9298, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31539241

ABSTRACT

The oncogenic fusion protein BCR-ABL is the driving force of leukemogenesis in chronic myeloid leukemia (CML). Despite great progress for CML treatment through application of tyrosine kinase inhibitors (TKIs) against BCR-ABL, long-term drug administration and clinical resistance continue to be an issue. Herein, we described the design, synthesis, and evaluation of novel proteolysis-targeting chimeric (PROTAC) small molecules targeting BCR-ABL which connect dasatinib and VHL E3 ubiquitin ligase ligand by extensive optimization of linkers. Our efforts have yielded SIAIS178 (19), which induces proper interaction between BCR-ABL and VHL ligase leading to effective degradation of BCR-ABL protein, achieves significant growth inhibition of BCR-ABL+ leukemic cells in vitro, and induces substantial tumor regression against K562 xenograft tumors in vivo. In addition, SIAIS178 also degrades several clinically relevant resistance-conferring mutations. Our data indicate that SIAIS178 as efficacious BCR-ABL degrader warrants extensive further investigation for the treatment of BCR-ABL+ leukemia.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , Drug Evaluation, Preclinical , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Piperazines/metabolism , Piperazines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Transplantation, Heterologous
10.
Chemistry ; 22(47): 16765-16769, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27643803

ABSTRACT

A Ni/BPh3 catalyzed [2+2+2] cycloaddition of alkyne-nitriles with alkynes has been developed, which provides an efficient route to fused pyridines under mild reaction conditions. Mechanistic studies indicate that an azanickelacycle via heterocoupling of an alkyne with a nitrile moiety is possibly formed as a key reaction intermediate. The Lewis acid catalyst is crucial to the successful transformation, which is suggested to promote the oxidative cyclization process.

11.
Chem Commun (Camb) ; 51(29): 6426-9, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25765939

ABSTRACT

A titanium-catalyzed cis-hydroalumination of propargylic amines with Red-Al is described, which provides an efficient way to produce Z-configured allylic amines in good to excellent yields with high stereoselectivity and good regioselectivity. The hydride-bridged Al/Ti bimetallic species may act as a real catalyst in this reaction.


Subject(s)
Organometallic Compounds/chemistry , Pargyline/analogs & derivatives , Propylamines/chemistry , Propylamines/chemical synthesis , Catalysis , Chemistry Techniques, Synthetic , Models, Molecular , Molecular Conformation , Pargyline/chemical synthesis , Pargyline/chemistry , Stereoisomerism
12.
Chemistry ; 21(4): 1420-4, 2015 Jan 19.
Article in English | MEDLINE | ID: mdl-25470360

ABSTRACT

Zirconocene-mediated multicomponent reactions of 1,3-butadiynes with ylidenemalononitriles in the absence or presence of CuCl have been developed. In the absence of CuCl, 1,3-butadiyne couples with three molecules of ylidenemalononitriles to yield azazirconacycles bearing a hexahydro-1,8-naphthyridine skeleton with high stereoselectivity. In the presence of CuCl, cyclopenta[b]pyridine or cyclopenta[b]quinolin-1-one derivatives are obtained via transmetalation of Zr-C bond to Cu-C bond as the key reaction step. These domino-type reactions proceed with high chemo-, regio- and/or stereoselectivities, and allowing the formation of multiple C-N and C-C bonds in a single operation.

13.
Org Biomol Chem ; 10(9): 1922-30, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22278679

ABSTRACT

The aryne [3 + 2] cycloaddition process with pyridinium imides breaks the aromaticity of the pyridine ring. By equipping the imide nitrogen with a sulfonyl group, the intermediate readily eliminates a sulfinate anion to restore the aromaticity, leading to the formation of pyrido[1,2-b]indazoles. The scope and limitation of this reaction are discussed. As an extension of this chemistry, N-tosylisoquinolinium imides, generated in situ from N'-(2-alkynylbenzylidene)-tosylhydrazides via an AgOTf-catalyzed 6-endo-dig electrophilic cyclization, readily undergo aryne [3 + 2] cycloaddition to afford indazolo[3,2-a]-isoquinolines in the same pot, offering a highly efficient route to these potential anticancer agents.


Subject(s)
Imides/chemistry , Indazoles/chemistry , Isoquinolines/chemistry , Phenazopyridine/chemistry , Sulfur Compounds/chemistry , Cyclization , Molecular Structure , Nitrogen/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...