Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mol Cell ; 74(3): 436-451.e7, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30926242

ABSTRACT

The evolutionarily related deubiquitinating enzymes (DUBs) USP25 and USP28 comprise an identical overall domain architecture but are functionally non-redundant: USP28 stabilizes c-MYC and other nuclear proteins, and USP25 regulates inflammatory TRAF signaling. We here compare molecular features of USP25 and USP28. Active enzymes form distinctively shaped dimers, with a dimerizing insertion spatially separating independently active catalytic domains. In USP25, but not USP28, two dimers can form an autoinhibited tetramer, where a USP25-specific, conserved insertion sequence blocks ubiquitin binding. In full-length enzymes, a C-terminal domain with a previously unknown fold has no impact on oligomerization, but N-terminal regions affect the dimer-tetramer equilibrium in vitro. We confirm oligomeric states of USP25 and USP28 in cells and show that modulating oligomerization affects substrate stabilization in accordance with in vitro activity data. Our work highlights how regions outside of the catalytic domain enable a conceptually intriguing interplay of DUB oligomerization and activity.


Subject(s)
Inflammation/genetics , Protein Conformation , Ubiquitin Thiolesterase/genetics , Amino Acid Sequence/genetics , Catalytic Domain/genetics , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Humans , Inflammation/pathology , Mutation/genetics , Protein Binding/genetics , Protein Domains/genetics , Protein Multimerization/genetics , Proto-Oncogene Proteins c-myb/chemistry , Proto-Oncogene Proteins c-myb/genetics , Signal Transduction/genetics , Substrate Specificity , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Ubiquitin/genetics , Ubiquitin Thiolesterase/chemistry
2.
Proc Natl Acad Sci U S A ; 110(36): E3445-54, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23946421

ABSTRACT

Stapled α-helical peptides have emerged as a promising new modality for a wide range of therapeutic targets. Here, we report a potent and selective dual inhibitor of MDM2 and MDMX, ATSP-7041, which effectively activates the p53 pathway in tumors in vitro and in vivo. Specifically, ATSP-7041 binds both MDM2 and MDMX with nanomolar affinities, shows submicromolar cellular activities in cancer cell lines in the presence of serum, and demonstrates highly specific, on-target mechanism of action. A high resolution (1.7-Å) X-ray crystal structure reveals its molecular interactions with the target protein MDMX, including multiple contacts with key amino acids as well as a role for the hydrocarbon staple itself in target engagement. Most importantly, ATSP-7041 demonstrates robust p53-dependent tumor growth suppression in MDM2/MDMX-overexpressing xenograft cancer models, with a high correlation to on-target pharmacodynamic activity, and possesses favorable pharmacokinetic and tissue distribution properties. Overall, ATSP-7041 demonstrates in vitro and in vivo proof-of-concept that stapled peptides can be developed as therapeutically relevant inhibitors of protein-protein interaction and may offer a viable modality for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Binding, Competitive , Cell Line, Tumor , Crystallography, X-Ray , Female , HCT116 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Peptides/chemistry , Peptides/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/therapeutic use , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Rats , Rats, Long-Evans , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 109(29): 11788-93, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22745160

ABSTRACT

Activation of p53 tumor suppressor by antagonizing its negative regulator murine double minute (MDM)2 has been considered an attractive strategy for cancer therapy and several classes of p53-MDM2 binding inhibitors have been developed. However, these compounds do not inhibit the p53-MDMX interaction, and their effectiveness can be compromised in tumors overexpressing MDMX. Here, we identify small molecules that potently block p53 binding with both MDM2 and MDMX by inhibitor-driven homo- and/or heterodimerization of MDM2 and MDMX proteins. Structural studies revealed that the inhibitors bind into and occlude the p53 pockets of MDM2 and MDMX by inducing the formation of dimeric protein complexes kept together by a dimeric small-molecule core. This mode of action effectively stabilized p53 and activated p53 signaling in cancer cells, leading to cell cycle arrest and apoptosis. Dual MDM2/MDMX antagonists restored p53 apoptotic activity in the presence of high levels of MDMX and may offer a more effective therapeutic modality for MDMX-overexpressing cancers.


Subject(s)
Apoptosis/physiology , Hydantoins/pharmacology , Models, Molecular , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Cycle Proteins , Cell Line, Tumor , Crystallization , Dimerization , Fluorescence Resonance Energy Transfer , Humans , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/chemistry , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles
4.
Bioorg Med Chem ; 22(15): 4001-9, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24997575

ABSTRACT

The field of small-molecule inhibitors of protein-protein interactions is rapidly advancing and the specific area of inhibitors of the p53/MDM2 interaction is a prime example. Several groups have published on this topic and multiple compounds are in various stages of clinical development. Building on the strength of the discovery of RG7112, a Nutlin imidazoline-based compound, and RG7388, a pyrrolidine-based compound, we have developed additional scaffolds that provide opportunities for future development. Here, we report the discovery and optimization of a highly potent and selective series of spiroindolinone small-molecule MDM2 inhibitors, culminating in RO8994.


Subject(s)
Indoles/chemistry , Indolizidines/chemistry , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Spiro Compounds/chemistry , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Imidazolines/chemistry , Indoles/therapeutic use , Indoles/toxicity , Indolizidines/therapeutic use , Indolizidines/toxicity , Molecular Dynamics Simulation , Neoplasms/drug therapy , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrrolidines/chemistry , Spiro Compounds/therapeutic use , Spiro Compounds/toxicity , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , para-Aminobenzoates/chemistry
5.
Lancet Oncol ; 13(11): 1133-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23084521

ABSTRACT

BACKGROUND: We report a proof-of-mechanism study of RG7112, a small-molecule MDM2 antagonist, in patients with chemotherapy-naive primary or relapsed well-differentiated or dedifferentiated MDM2-amplified liposarcoma who were eligible for resection. METHODS: Patients with well-differentiated or dedifferentiated liposarcoma were enrolled at four centres in France. Patients received up to three 28-day neoadjuvant treatment cycles of RG7112 1440 mg/m(2) per day for 10 days. If a patient progressed at any point after the first cycle, the lesion was resected or, if unresectable, an end-of-study biopsy was done. The primary endpoint was to assess markers of RG7112-dependent MDM2 inhibition and P53 pathway activation (P53, P21, MDM2, Ki-67, macrophage inhibitory cytokine-1 [MIC-1], and apoptosis). All analyses were per protocol. This trial is registered with EudraCT, number 2009-015522-10. RESULTS: Between June 3, and Dec 14, 2010, 20 patients were enrolled and completed pretreatment and day 8 biopsies. 18 of 20 patients had TP53 wild-type tumours and two carried missense TP53 mutations. 14 of 17 assessed patients had MDM2 gene amplification. Compared with baseline, P53 and P21 concentrations, assessed by immunohistochemistry, had increased by a median of 4·86 times (IQR 4·38-7·97; p=0·0001) and 3·48 times (2·05-4·09; p=0·0001), respectively, at day 8 (give or take 2 days). At the same timepoint, relative MDM2 mRNA expression had increased by a median of 3·03 times (1·23-4·93; p=0·003) that at baseline. The median change from baseline for Ki-67-positive tumour cells was -5·05% (IQR -12·55 to 0·05; p=0·01). Drug exposure correlated with blood concentrations of MIC-1 (p<0·0001) and haematological toxicity. One patient had a confirmed partial response and 14 had stable disease. All patients experienced at least one adverse event, mostly nausea (14 patients), vomiting (11 patients), asthenia (nine patients), diarrhoea (nine patients), and thrombocytopenia (eight patients). There were 12 serious adverse events in eight patients, the most common of which were neutropenia (six patients) and thrombocytopenia (three patients). DISCUSSION: MDM2 inhibition activates the P53 pathway and decreases cell proliferation in MDM2-amplified liposarcoma. This study suggests that it is feasible to undertake neoadjuvant biopsy-driven biomarker studies in liposarcoma. FUNDING: F Hoffmann-La Roche.


Subject(s)
Antineoplastic Agents , Liposarcoma/drug therapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53 , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Apoptosis , Cell Differentiation , Cell Proliferation/drug effects , Disease-Free Survival , Female , Growth Differentiation Factor 15/metabolism , Humans , Male , Middle Aged , Mutation , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Young Adult
6.
Target Oncol ; 18(2): 269-285, 2023 03.
Article in English | MEDLINE | ID: mdl-36826464

ABSTRACT

BACKGROUND: Patients with triple-negative breast cancer (TNBC) expressing the androgen receptor (AR) respond poorly to neoadjuvant chemotherapy, although AR antagonists have shown promising clinical activity, suggesting these tumors are AR-dependent. cAMP responsive element binding protein (CREB)-binding protein (CBP) and p300 are transcriptional co-activators for the AR, a key driver of AR+ breast and prostate cancer, and may provide a novel therapeutic target in AR+ TNBC. OBJECTIVES: The aim of this study was to determine the therapeutic potential of FT-6876, a new CBP/p300 bromodomain inhibitor, in breast cancer models with a range of AR levels in vitro and in vivo. METHODS: Effects of FT-6876 on the CBP/p300 pathway were determined by combining chromatin immunoprecipitation (ChIP) with precision run-on sequencing (PRO-seq) complemented with H3K27 acetylation (Ac) and transcriptional profiling. The antiproliferative effect of FT-6876 was also measured in vitro and in vivo. RESULTS: We describe the discovery of FT-6876, a potent and selective CBP/p300 bromodomain inhibitor. The combination of ChIP and PRO-seq confirmed the reduction in H3K27Ac at specific promoter sites concurrent with a decrease in CBP/p300 on the chromatin and a reduction in nascent RNA and enhancer RNA. This was associated with a time- and concentration-dependent reduction in H3K37Ac associated with a decrease in AR and estrogen receptor (ER) target gene expression. This led to a time-dependent growth inhibition in AR+ models, correlated with AR expression. Tumor growth inhibition was also observed in AR+ tumor models of TNBC and ER+ breast cancer subtypes with consistent pharmacokinetics and pharmacodynamics. CONCLUSION: Our findings demonstrate FT-6876 as a promising new CBP/p300 bromodomain inhibitor, with efficacy in preclinical models of AR+ breast cancer.


Subject(s)
Receptors, Androgen , Triple Negative Breast Neoplasms , Male , Humans , Receptors, Androgen/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Protein Binding , RNA/metabolism
7.
J Med Chem ; 66(14): 9401-9417, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37439511

ABSTRACT

We report the discovery of sulanemadlin (ALRN-6924), the first cell-permeating, stabilized α-helical peptide to enter clinical trials. ALRN-6924 is a "stapled peptide" that mimics the N-terminal domain of the p53 tumor suppressor protein. It binds with high affinity to both MDM2 and MDMX (also known as MDM4), the endogenous inhibitors of p53, to activate p53 signaling in cells having a non-mutant, or wild-type TP53 genotype (TP53-WT). Iterative structure-activity optimization endowed ALRN-6924 with favorable cell permeability, solubility, and pharmacokinetic and safety profiles. Intracellular proteolysis of ALRN-6924 forms a long-acting active metabolite with potent MDM2 and MDMX binding affinity and slow dissociation kinetics. At high doses, ALRN-6924 exhibits on-mechanism anticancer activity in TP53-WT tumor models. At lower doses, ALRN-6924 transiently arrests the cell cycle in healthy tissues to protect them from chemotherapy without protecting the TP53-mutant cancer cells. These results support the continued clinical evaluation of ALRN-6924 as an anticancer and chemoprotection agent.


Subject(s)
Antineoplastic Agents , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Protein Binding , Peptides/chemistry , Antineoplastic Agents/chemistry , Cell Cycle Proteins/metabolism
8.
Curr Top Med Chem ; 6(5): 423-34, 2006.
Article in English | MEDLINE | ID: mdl-16719801

ABSTRACT

Binding affinity optimization of small molecules interacting with polar binding sites on target proteins is a formidable, but not uncommon challenge in drug discovery. The challenge relates to the difficulty of integrating favourable and unfavourable polar, non-polar and conformation contributions into overall favourable binding energies. This review describes the surprising breakthrough findings leading to the development of Tamiflu, a clinically efficacious orally bioavailable drug targeting the active site of influenza neuraminidase (NA). The NA active site is highly polar and formed mostly by arginine, aspartate and glutamate residues. This active site structure evolved for efficient interaction with charged sialic acid moieties on glycoproteins and stabilization of an oxocarbonium ion in the transition state of the neuraminidase reaction. The initial strategy of optimizing polar interactions in transition state analogs led to NA inhibitors (NAIs) with sub-nanomolar binding affinities, but such compounds were highly polar and lacked oral bioavailability. The realization of the possibility to achieve high affinity binding in a highly polar active site through optimization of non-polar and van-der-Waals interactions initially appeared counterintuitive and required a few serendipitous findings, but was key to reduce the polarity of drug candidates, avoid large desolvation penalties and achieve oral bioavailability.


Subject(s)
Acetamides/pharmacology , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Acetamides/chemical synthesis , Acetamides/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Influenza, Human/drug therapy , Influenza, Human/prevention & control , Models, Molecular , Molecular Conformation , Molecular Weight , Neuraminidase/chemistry , Oseltamivir , Structure-Activity Relationship
9.
J Med Chem ; 49(22): 6549-60, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-17064073

ABSTRACT

The cyclin-dependent kinases (CDKs) and their cyclin partners are key regulators of the cell cycle. Since deregulation of CDKs is found with high frequency in many human cancer cells, pharmacological inhibition of CDKs with small molecules has the potential to provide an effective strategy for the treatment of cancer. The 2,4-diamino-5-ketopyrimidines 6 reported here represent a novel class of potent and ATP-competitive inhibitors that selectively target the cyclin-dependent kinase family. This diaminopyrimidine core with a substituted 4-piperidine moiety on the C2-amino position and 2-methoxybenzoyl at the C5 position has been identified as the critical structure responsible for the CDK inhibitory activity. Further optimization has led to a good number of analogues that show potent inhibitory activities against CDK1, CDK2, and CDK4 but are inactive against a large panel of serine/threonine and tyrosine kinases (K(i) > 10 microM). As one of these representative analogues, compound 39 (R547) has the best CDK inhibitory activities (K(i) = 0.001, 0.003, and 0.001 microM for CDK1, CDK2, and CDK4, respectively) and excellent in vitro cellular potency, inhibiting the growth of various human tumor cell lines including an HCT116 cell line (IC(50) = 0.08 microM). An X-ray crystal structure of 39 bound to CDK2 has been determined in this study, revealing a binding mode that is consistent with our SAR. Compound 39 demonstrates significant in vivo efficacy in the HCT116 human colorectal tumor xenograft model in nude mice with up to 95% tumor growth inhibition. On the basis of its superior overall profile, 39 was chosen for further evaluation and has progressed into Phase I clinical trial for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chemical Phenomena , Chemistry, Physical , Cyclin-Dependent Kinase 2/chemistry , Drug Design , Female , Humans , Indicators and Reagents , Kinetics , Mice , Mice, Nude , Models, Molecular , Structure-Activity Relationship , X-Ray Diffraction
10.
Clin Cancer Res ; 22(4): 868-76, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26459177

ABSTRACT

PURPOSE: RG7112 is a small-molecule MDM2 antagonist. MDM2 is a negative regulator of the tumor suppressor p53 and frequently overexpressed in leukemias. Thus, a phase I study of RG7112 in patients with hematologic malignancies was conducted. EXPERIMENTAL DESIGN: Primary study objectives included determination of the dose and safety profile of RG7112. Secondary objectives included evaluation of pharmacokinetics; pharmacodynamics, such as TP53-mutation status and MDM2 expression; and preliminary clinical activity. Patients were divided into two cohorts: Stratum A [relapsed/refractory acute myeloid leukemia (AML; except acute promyelocytic leukemia), acute lymphoblastic leukemia, and chronic myelogenous leukemia] and Stratum B (relapsed/refractory chronic lymphocytic leukemia/small cell lymphocytic leukemia; CLL/sCLL). Some Stratum A patients were treated at the MTD to assess clinical activity. RESULTS: RG7112 was administered to 116 patients (96 patients in Stratum A and 20 patients in Stratum B). All patients experienced at least 1 adverse event, and 3 dose-limiting toxicities were reported. Pharmacokinetic analysis indicated that twice-daily dosing enhanced daily exposure. Antileukemia activity was observed in the 30 patients with AML assessed at the MTD, including 5 patients who met International Working Group (IWG) criteria for response. Exploratory analysis revealed TP53 mutations in 14% of Stratum A patients and in 40% of Stratum B patients. Two patients with TP53 mutations exhibited clinical activity. p53 target genes were induced only in TP53 wild-type leukemic cells. Baseline expression levels of MDM2 correlated positively with clinical response. CONCLUSIONS: RG7112 demonstrated clinical activity against relapsed/refractory AML and CLL/sCLL. MDM2 inhibition resulted in p53 stabilization and transcriptional activation of p53-target genes. We provide proof-of-concept that MDM2 inhibition restores p53 function and generates clinical responses in hematologic malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Imidazolines/therapeutic use , Leukemia, Lymphoid/drug therapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Apoptosis , DNA Mutational Analysis , Drug Administration Schedule , Gene Expression , Humans , Imidazolines/pharmacokinetics , Imidazolines/toxicity , Leukemia, Lymphoid/genetics , Maximum Tolerated Dose , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Methods Enzymol ; 399: 622-33, 2005.
Article in English | MEDLINE | ID: mdl-16338385

ABSTRACT

Inhibition of E3 ligase-substrate binding is the most direct approach for blocking protein ubiquitylation and degradation. However, protein-protein interactions have proven to be difficult targets for discovery of small molecules that bind at the interface and modulate protein activity in a selective manner. Recently, we developed the first potent and selective small-molecule inhibitors of the binding between MDM2 E3 ligase and its substrate p53 (Vassilev et al., 2004). This process was aided significantly by the acquisition and use of structural information. We describe herein how such information was obtained and used at various stages in the program. These applications included assessment of MDM2 as a target, evaluation of hits from high-throughput screening and the selection of lead molecules, and analysis of binding strategies used by the inhibitors as a basis for guiding studies of similar systems. These tools are likely to be useful in any attempt to find and develop druglike compounds that modulate the function of a protein-protein interaction.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/chemistry , Substrate Specificity , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/chemistry
12.
Curr Top Med Chem ; 5(2): 159-65, 2005.
Article in English | MEDLINE | ID: mdl-15853644

ABSTRACT

The discovery of the key negative regulator MDM2 (mouse double minute 2, also termed HDM2 for its human equivalent) provided a great opportunity to manipulate the levels of the tumor suppressor p53 in cancer cells. Activation of p53 in tumor cells by inhibiting the interaction of MDM2 with p53 has therefore been the focus of a large effort in drug discovery. The modulation of protein-protein interactions, however, has historically been very difficult to achieve owing to the large surface area of interaction. In this article, we review the recent accomplishments in this area and our quest for a clinically viable MDM2 inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Humans , Models, Molecular , Peptides/pharmacology , Proto-Oncogene Proteins c-mdm2
13.
ACS Med Chem Lett ; 5(2): 124-7, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24900784

ABSTRACT

The development of small-molecule MDM2 inhibitors to restore dysfunctional p53 activities represents a novel approach for cancer treatment. In a previous communication, the efforts leading to the identification of a non-imidazoline MDM2 inhibitor, RG7388, was disclosed and revealed the desirable in vitro and in vivo pharmacological properties that this class of pyrrolidine-based inhibitors possesses. Given this richness and the critical need for a wide variety of chemical structures to ensure success in the clinic, research was expanded to evaluate additional derivatives. Here we report two new potent, selective, and orally active p53-MDM2 antagonists, RO5353 and RO2468, as follow-ups with promising potential for clinical development.

14.
ACS Med Chem Lett ; 4(7): 660-5, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-24900726

ABSTRACT

Protein-protein interaction (PPI) systems represent a rich potential source of targets for drug discovery, but historically have proven to be difficult, particularly in the lead identification stage. Application of the fragment-based approach may help toward success with this target class. To provide an example toward understanding the potential issues associated with such an application, we have deconstructed one of the best established protein-protein inhibitors, the Nutlin series that inhibits the interaction between MDM2 and p53, into fragments, and surveyed the resulting binding properties using heteronuclear single quantum coherence nuclear magnetic resonance (HSQC NMR), surface plasmon resonance (SPR), and X-ray crystallography. We report the relative contributions toward binding affinity for each of the key substituents of the Nutlin molecule and show that this series could hypothetically have been discovered via a fragment approach. We find that the smallest fragment of Nutlin that retains binding accesses two subpockets of MDM2 and has a molecular weight at the high end of the range that normally defines fragments.

15.
ACS Med Chem Lett ; 4(5): 466-9, 2013 May 09.
Article in English | MEDLINE | ID: mdl-24900694

ABSTRACT

The p53 tumor suppressor is a potent transcription factor that plays a key role in the regulation of cellular responses to stress. It is controlled by its negative regulator MDM2, which binds directly to p53 and inhibits its transcriptional activity. MDM2 also targets p53 for degradation by the proteasome. Many tumors produce high levels of MDM2, thereby impairing p53 function. Restoration of p53 activity by inhibiting the p53-MDM2 interaction may represent a novel approach to cancer treatment. RG7112 (2g) is the first clinical small-molecule MDM2 inhibitor designed to occupy the p53-binding pocket of MDM2. In cancer cells expressing wild-type p53, RG7112 stabilizes p53 and activates the p53 pathway, leading to cell cycle arrest, apoptosis, and inhibition or regression of human tumor xenografts.

16.
Cancer Res ; 73(8): 2587-97, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23400593

ABSTRACT

MDM2 negatively regulates p53 stability and many human tumors overproduce MDM2 as a mechanism to restrict p53 function. Thus, inhibitors of p53-MDM2 binding that can reactivate p53 in cancer cells may offer an effective approach for cancer therapy. RG7112 is a potent and selective member of the nutlin family of MDM2 antagonists currently in phase I clinical studies. RG7112 binds MDM2 with high affinity (K(D) ~ 11 nmol/L), blocking its interactions with p53 in vitro. A crystal structure of the RG7112-MDM2 complex revealed that the small molecule binds in the p53 pocket of MDM2, mimicking the interactions of critical p53 amino acid residues. Treatment of cancer cells expressing wild-type p53 with RG7112 activated the p53 pathway, leading to cell-cycle arrest and apoptosis. RG7112 showed potent antitumor activity against a panel of solid tumor cell lines. However, its apoptotic activity varied widely with the best response observed in osteosarcoma cells with MDM2 gene amplification. Interestingly, inhibition of caspase activity did not change the kinetics of p53-induced cell death. Oral administration of RG7112 to human xenograft-bearing mice at nontoxic concentrations caused dose-dependent changes in proliferation/apoptosis biomarkers as well as tumor inhibition and regression. Notably, RG7112 was highly synergistic with androgen deprivation in LNCaP xenograft tumors. Our findings offer a preclinical proof-of-concept that RG7112 is effective in treatment of solid tumors expressing wild-type p53.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazolines/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Imidazolines/chemistry , Mice , Molecular Docking Simulation , Neoplasms/pathology , Protein Binding/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Burden/drug effects , Tumor Suppressor Protein p53/chemistry , Xenograft Model Antitumor Assays
17.
J Med Chem ; 56(14): 5979-83, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23808545

ABSTRACT

Restoration of p53 activity by inhibition of the p53-MDM2 interaction has been considered an attractive approach for cancer treatment. However, the hydrophobic protein-protein interaction surface represents a significant challenge for the development of small-molecule inhibitors with desirable pharmacological profiles. RG7112 was the first small-molecule p53-MDM2 inhibitor in clinical development. Here, we report the discovery and characterization of a second generation clinical MDM2 inhibitor, RG7388, with superior potency and selectivity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrrolidines/chemical synthesis , Tumor Suppressor Protein p53/antagonists & inhibitors , para-Aminobenzoates/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Humans , Male , Mice , Mice, Inbred C57BL , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , para-Aminobenzoates/pharmacology , para-Aminobenzoates/therapeutic use
18.
J Med Chem ; 54(7): 2255-65, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21375264

ABSTRACT

The development of a new series of p38α inhibitors resulted in the identification of two clinical candidates, one of which was advanced into a phase 2 clinical study for rheumatoid arthritis. The original lead, an lck inhibitor that also potently inhibited p38α, was a screening hit from our kinase inhibitor library. This manuscript describes the optimization of the lead to p38-selective examples with good pharmacokinetic properties.


Subject(s)
Drug Discovery/methods , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Pyridones/pharmacology , Pyridones/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Administration, Oral , Arthritis, Rheumatoid/drug therapy , Biological Availability , Cell Line , Clinical Trials as Topic , Humans , Mitogen-Activated Protein Kinase 14/chemistry , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyridones/administration & dosage , Pyridones/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Structure-Activity Relationship , Substrate Specificity
19.
Science ; 303(5659): 844-8, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14704432

ABSTRACT

MDM2 binds the p53 tumor suppressor protein with high affinity and negatively modulates its transcriptional activity and stability. Overexpression of MDM2, found in many human tumors, effectively impairs p53 function. Inhibition of MDM2-p53 interaction can stabilize p53 and may offer a novel strategy for cancer therapy. Here, we identify potent and selective small-molecule antagonists of MDM2 and confirm their mode of action through the crystal structures of complexes. These compounds bind MDM2 in the p53-binding pocket and activate the p53 pathway in cancer cells, leading to cell cycle arrest, apoptosis, and growth inhibition of human tumor xenografts in nude mice.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Imidazoles/pharmacology , Neoplasms, Experimental/pathology , Nuclear Proteins , Piperazines/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Crystallization , Crystallography, X-Ray , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dose-Response Relationship, Drug , Gene Expression , Genes, p53 , Humans , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemistry , Imidazoles/metabolism , Mice , Mice, Nude , Models, Molecular , Molecular Weight , NIH 3T3 Cells , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Phosphorylation , Piperazines/chemistry , Piperazines/metabolism , Protein Conformation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Stereoisomerism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL