Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
ACS Omega ; 6(34): 22073-22102, 2021 Aug 31.
Article En | MEDLINE | ID: mdl-34497901

Following the identification of thieno[2,3-d]pyrimidine-based selective and potent inhibitors of MCL-1, we explored the effect of core swapping at different levels of advancement. During hit-to-lead optimization, X-ray-guided S-N replacement in the core provided a new vector, whose exploration led to the opening of the so-called deep-S2 pocket of MCL-1. Unfortunately, the occupation of this region led to a plateau in affinity and had to be abandoned. As the project approached selection of a clinical candidate, a series of core swap analogues were also prepared. The affinity and cellular activity of these compounds showed a significant dependence on the core structure. In certain cases, we also observed an increased and accelerated epimerization of the atropoisomers. The most potent core replacement analogues showed considerable in vivo PD response. One compound was progressed into efficacy studies and inhibited tumor growth.

2.
J Med Chem ; 63(22): 13762-13795, 2020 11 25.
Article En | MEDLINE | ID: mdl-33146521

Myeloid cell leukemia 1 (Mcl-1) has emerged as an attractive target for cancer therapy. It is an antiapoptotic member of the Bcl-2 family of proteins, whose upregulation in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy. Here we report the discovery of our clinical candidate S64315, a selective small molecule inhibitor of Mcl-1. Starting from a fragment derived lead compound, we have conducted structure guided optimization that has led to a significant (3 log) improvement of target affinity as well as cellular potency. The presence of hindered rotation along a biaryl axis has conferred high selectivity to the compounds against other members of the Bcl-2 family. During optimization, we have also established predictive PD markers of Mcl-1 inhibition and achieved both efficient in vitro cell killing and tumor regression in Mcl-1 dependent cancer models. The preclinical candidate has drug-like properties that have enabled its development and entry into clinical trials.


Antineoplastic Agents/chemistry , Drug Discovery/methods , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Animals , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Female , HCT116 Cells , HeLa Cells , Humans , Mice , Mice, SCID , Protein Structure, Secondary , Protein Structure, Tertiary
3.
Blood Adv ; 4(12): 2762-2767, 2020 06 23.
Article En | MEDLINE | ID: mdl-32569380

Improving survival outcomes in adult B-cell acute lymphoblastic leukemia (B-ALL) remains a clinical challenge. Relapsed disease has a poor prognosis despite the use of tyrosine kinase inhibitors (TKIs) for Philadelphia chromosome positive (Ph+ ALL) cases and immunotherapeutic approaches, including blinatumomab and chimeric antigen receptor T cells. Targeting aberrant cell survival pathways with selective small molecule BH3-mimetic inhibitors of BCL-2 (venetoclax, S55746), BCL-XL (A1331852), or MCL1 (S63845) is an emerging therapeutic option. We report that combined targeting of BCL-2 and MCL1 is synergistic in B-ALL in vitro. The combination demonstrated greater efficacy than standard chemotherapeutics and TKIs in primary samples from adult B-ALL with Ph+ ALL, Ph-like ALL, and other B-ALL. Moreover, combined BCL-2 or MCL1 inhibition with dasatinib showed potent killing in primary Ph+ B-ALL cases, but the BH3-mimetic combination appeared superior in vitro in a variety of Ph-like ALL samples. In PDX models, combined BCL-2 and MCL1 targeting eradicated ALL from Ph- and Ph+ B-ALL cases, although fatal tumor lysis was observed in some instances of high tumor burden. We conclude that a dual BH3-mimetic approach is highly effective in diverse models of high-risk human B-ALL and warrants assessment in clinical trials that incorporate tumor lysis precautions.


Precursor Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Proteins c-bcl-2 , Adult , B-Lymphocytes , Cell Line, Tumor , Humans , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/genetics
4.
ACS Omega ; 4(5): 8892-8906, 2019 May 31.
Article En | MEDLINE | ID: mdl-31459977

We describe our work to establish structure- and fragment-based drug discovery to identify small molecules that inhibit the anti-apoptotic activity of the proteins Mcl-1 and Bcl-2. This identified hit series of compounds, some of which were subsequently optimized to clinical candidates in trials for treating various cancers. Many protein constructs were designed to identify protein with suitable properties for different biophysical assays and structural methods. Fragment screening using ligand-observed NMR experiments identified several series of compounds for each protein. The series were assessed for their potential for subsequent optimization using 1H and 15N heteronuclear single-quantum correlation NMR, surface plasmon resonance, and isothermal titration calorimetry measurements to characterize and validate binding. Crystal structures could not be determined for the early hits, so NMR methods were developed to provide models of compound binding to guide compound optimization. For Mcl-1, a benzodioxane/benzoxazine series was optimized to a K d of 40 µM before a thienopyrimidine hit series was identified which subsequently led to the lead series from which the clinical candidate S 64315 (MIK 665) was identified. For Bcl-2, the fragment-derived series were difficult to progress, and a compound derived from a published tetrahydroquinone compound was taken forward as the hit from which the clinical candidate (S 55746) was obtained. For both the proteins, the work to establish a portfolio of assays gave confidence for identification of compounds suitable for optimization.

5.
J Med Chem ; 62(15): 6913-6924, 2019 08 08.
Article En | MEDLINE | ID: mdl-31339316

Myeloid cell leukemia 1 (Mcl-1), an antiapoptotic member of the Bcl-2 family of proteins, whose upregulation when observed in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy, has emerged as an attractive target for cancer therapy. Here, we report the discovery of selective small molecule inhibitors of Mcl-1 that inhibit cellular activity. Fragment screening identified thienopyrimidine amino acids as promising but nonselective hits that were optimized using nuclear magnetic resonance and X-ray-derived structural information. The introduction of hindered rotation along a biaryl axis has conferred high selectivity to the compounds, and cellular activity was brought on scale by offsetting the negative charge of the anchoring carboxylate group. The obtained compounds described here exhibit nanomolar binding affinity and mechanism-based cellular efficacy, caspase induction, and growth inhibition. These early research efforts illustrate drug discovery optimization from thienopyrimidine hits to a lead compound, the chemical series leading to the identification of our more advanced compounds S63845 and S64315.


Cell Survival/physiology , Drug Discovery/methods , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Thiophenes/chemistry , Thiophenes/metabolism , Cell Survival/drug effects , HCT116 Cells , HeLa Cells , Humans , Protein Structure, Tertiary , Pyrimidines/pharmacology , Structure-Activity Relationship , Thiophenes/pharmacology
6.
Leukemia ; 33(4): 905-917, 2019 04.
Article En | MEDLINE | ID: mdl-30214012

Improving outcomes in acute myeloid leukemia (AML) remains a major clinical challenge. Overexpression of pro-survival BCL-2 family members rendering transformed cells resistant to cytotoxic drugs is a common theme in cancer. Targeting BCL-2 with the BH3-mimetic venetoclax is active in AML when combined with low-dose chemotherapy or hypomethylating agents. We now report the pre-clinical anti-leukemic efficacy of a novel BCL-2 inhibitor S55746, which demonstrates synergistic pro-apoptotic activity in combination with the MCL1 inhibitor S63845. Activity of the combination was caspase and BAX/BAK dependent, superior to combination with standard cytotoxic AML drugs and active against a broad spectrum of poor risk genotypes, including primary samples from patients with chemoresistant AML. Co-targeting BCL-2 and MCL1 was more effective against leukemic, compared to normal hematopoietic progenitors, suggesting a therapeutic window of activity. Finally, S55746 combined with S63845 prolonged survival in xenograft models of AML and suppressed patient-derived leukemia but not normal hematopoietic cells in bone marrow of engrafted mice. In conclusion, a dual BH3-mimetic approach is feasible, highly synergistic, and active in diverse models of human AML. This approach has strong clinical potential to rapidly suppress leukemia, with reduced toxicity to normal hematopoietic precursors compared to chemotherapy.


Biomimetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Drug Therapy, Combination , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Peptide Fragments , Proto-Oncogene Proteins , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Oncotarget ; 9(28): 20075-20088, 2018 Apr 13.
Article En | MEDLINE | ID: mdl-29732004

Escape from apoptosis is one of the major hallmarks of cancer cells. The B-cell Lymphoma 2 (BCL-2) gene family encodes pro-apoptotic and anti-apoptotic proteins that are key regulators of the apoptotic process. Overexpression of the pro-survival member BCL-2 is a well-established mechanism contributing to oncogenesis and chemoresistance in several cancers, including lymphoma and leukemia. Thus, BCL-2 has become an attractive target for therapeutic strategy in cancer, as demonstrated by the recent approval of ABT-199 (Venclexta™) in relapsed or refractory Chronic Lymphocytic Leukemia with 17p deletion. Here, we describe a novel orally bioavailable BCL-2 selective and potent inhibitor called S55746 (also known as BCL201). S55746 occupies the hydrophobic groove of BCL-2. Its selectivity profile demonstrates no significant binding to MCL-1, BFL-1 (BCL2A1/A1) and poor affinity for BCL-XL. Accordingly, S55746 has no cytotoxic activity on BCL-XL-dependent cells, such as platelets. In a panel of hematological cell lines, S55746 induces hallmarks of apoptosis including externalization of phosphatidylserine, caspase-3 activation and PARP cleavage. Ex vivo, S55746 induces apoptosis in the low nanomolar range in primary Chronic Lymphocytic Leukemia and Mantle Cell Lymphoma patient samples. Finally, S55746 administered by oral route daily in mice demonstrated robust anti-tumor efficacy in two hematological xenograft models with no weight lost and no change in behavior. Taken together, these data demonstrate that S55746 is a novel, well-tolerated BH3-mimetic targeting selectively and potently the BCL-2 protein.

8.
Sci Transl Med ; 9(401)2017 Aug 02.
Article En | MEDLINE | ID: mdl-28768804

The development of BH3 mimetics, which antagonize prosurvival proteins of the BCL-2 family, represents a potential breakthrough in cancer therapy. Targeting the prosurvival member MCL-1 has been an area of intense interest because it is frequently deregulated in cancer. In breast cancer, MCL-1 is often amplified, and high expression predicts poor patient outcome. We tested the MCL-1 inhibitor S63845 in breast cancer cell lines and patient-derived xenografts with high expression of MCL-1. S63845 displayed synergistic activity with docetaxel in triple-negative breast cancer and with trastuzumab or lapatinib in HER2-amplified breast cancer. Using S63845-resistant cells combined with CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated 9) technology, we identified deletion of BAK and up-regulation of prosurvival proteins as potential mechanisms that confer resistance to S63845 in breast cancer. Collectively, our findings provide a strong rationale for the clinical evaluation of MCL-1 inhibitors in breast cancer.


Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Gene Amplification , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Pyrimidines/therapeutic use , Receptor, ErbB-2/genetics , Thiophenes/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Docetaxel , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Lapatinib , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Pyrimidines/pharmacology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Survival Analysis , Taxoids/pharmacology , Taxoids/therapeutic use , Thiophenes/pharmacology , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Treatment Outcome , Triple Negative Breast Neoplasms/pathology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism
9.
PLoS One ; 9(1): e80836, 2014.
Article En | MEDLINE | ID: mdl-24454684

PURPOSE: Uveal melanoma (UM) is associated with a high risk of metastases and lack of efficient therapies. Reduced capacity for apoptosis induction by chemotherapies is one obstacle to efficient treatments. Human UM is characterized by high expression of the anti-apoptotic protein Bcl-2. Consequently, regulators of apoptosis such as Bcl-2 family inhibitors may constitute an attractive approach to UM therapeutics. In this aim, we have investigated the efficacy of the Bcl-2/Bcl-XL inhibitor S44563 on 4 UM Patient-Derived Xenografts (PDXs) and derived-cell lines. EXPERIMENTAL DESIGN: Four well characterized UM PDXs were used for in vivo experiments. S44563 was administered alone or combined with fotemustine either concomitantly or after the alkylating agent. Bcl-2, Bcl-XL, and Mcl-1 expressions after S44563 administration were evaluated by immunohistochemistry (IHC). RESULTS: S44563 administered alone by at 50 and 100 mg/kg i.p. induced a significant tumour growth inhibition in only one xenograft model with a clear dose effect. However, when S44563 was concomitantly administered with fotemustine, we observed a synergistic activity in 3 out of the 4 tested models. In addition, S44563 administered after fotemustine induced a tumour growth delay in 2 out of 3 tested xenografts. Finally, IHC analyses showed that Bcl-2, Bcl-XL, and Mcl-1 expression were not modified after S44563 administration. CONCLUSION: The novel anti-apoptotic experimental compound S44563, despite a relative low efficacy when administered alone, increased the efficacy of fotemustine in either concomitant or sequential combinations or indeed subsequent to fotemustine. These data support further exploration of potential therapeutic effect of Bcl-2/Bcl-xl inhibition in human UM.


Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Melanoma/drug therapy , Molecular Targeted Therapy , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Uveal Neoplasms/drug therapy , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Fluorescence Polarization , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Immunohistochemistry , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nitrosourea Compounds/pharmacology , Nitrosourea Compounds/therapeutic use , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Peptides/metabolism , Protein Binding/drug effects , Sulfonamides/administration & dosage , Survival Analysis , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , bcl-X Protein/metabolism
10.
Nat Rev Cancer ; 13(7): 455-65, 2013 Jul.
Article En | MEDLINE | ID: mdl-23783119

Cancer cells are subject to many apoptotic stimuli that would kill them were it not for compensatory prosurvival alterations. BCL-2-like (BCL-2L) proteins contribute to such aberrant behaviour by engaging a network of interactions that is potent at promoting survival but that is also fragile: inhibition of a restricted number of interactions may suffice to trigger cancer cell death. Currently available and novel compounds that inhibit these interactions could be efficient therapeutic agents if this phenotype of BCL-2L dependence was better understood at a molecular, cellular and systems level and if it could be diagnosed by relevant biomarkers.


Antineoplastic Agents/therapeutic use , Neoplasms/etiology , Neoplasms/prevention & control , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/physiology , Antineoplastic Agents/pharmacology , Humans , Molecular Targeted Therapy , Neoplasms/metabolism
11.
J Biol Chem ; 284(44): 30257-63, 2009 Oct 30.
Article En | MEDLINE | ID: mdl-19759007

Unlike other antiapoptotic members of the Bcl-2 family, Bfl-1 does not contain a well defined C-terminal transmembrane domain, and whether the C-terminal tail of Bfl-1 functions as a membrane anchor is not yet clearly established. The molecular modeling study of the full-length Bfl-1 performed within this work suggests that Bfl-1 may co-exist in two distinct conformational states: one in which its C-terminal helix alpha9 is inserted in the hydrophobic groove formed by the BH1-3 domains of Bfl-1 and one with its C terminus. Parallel analysis of the subcellular localization of Bfl-1 indicates that even if Bfl-1 may co-exist in two distinct conformational states, most of the endogenous protein is tightly associated with the mitochondria by its C terminus in both healthy and apoptotic peripheral blood lymphocytes as well as in malignant B cell lines. However, the helix alpha9 of Bfl-1, and therefore the binding of Bfl-1 to mitochondria, is not absolutely required for the antiapoptotic activity of Bfl-1. A particular feature of Bfl-1 is the amphipathic character of its C-terminal helix alpha9. Our data clearly indicate that this property of helix alpha9 is required for the anchorage of Bfl-1 to the mitochondria but also regulates the antiapoptotic function Bfl-1.


Apoptosis Regulatory Proteins , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , Amino Acids , Animals , Apoptosis , B-Lymphocytes/pathology , Cell Line , Cell Line, Tumor , Humans , Lymphocytes/cytology , Mice , Minor Histocompatibility Antigens , Models, Molecular , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
J Cell Biol ; 185(2): 279-90, 2009 Apr 20.
Article En | MEDLINE | ID: mdl-19380879

It is still unclear whether the BH3-only protein Puma (p53 up-regulated modulator of apoptosis) can prime cells to death and render antiapoptotic BH3-binding Bcl-2 homologues necessary for survival through its ability to directly interact with proapoptotic Bax and activate it. In this study, we provide further evidence, using cell-free assays, that the BH3 domain of Puma binds Bax at an activation site that comprises the first helix of Bax. We also show that, in yeast, Puma interacts with Bax and triggers its killing activity when Bcl-2 homologues are absent but not when Bcl-xL is expressed. Finally, endogenous Puma is involved in the apoptotic response of human colorectal cancer cells to the Bcl-2/Bcl-xL inhibitor ABT-737, even in conditions where the expression of Mcl-1 is down-regulated. Thus, Puma is competent to trigger Bax activity by itself, thereby promoting cellular dependence on prosurvival Bcl-2 family members.


Apoptosis Regulatory Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Biphenyl Compounds/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Knockdown Techniques , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/metabolism , Piperazines/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Sulfonamides/metabolism , Yeasts/physiology , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics
13.
Cell Cycle ; 6(18): 2263-7, 2007 Sep 15.
Article En | MEDLINE | ID: mdl-17890908

When added to cells, a variety of autophagy inducers that operate through distinct mechanisms and target different organelles for autophagic destruction (mitochondria in mitophagy, endoplasmic reticulum in reticulophagy) rarely induce autophagic vacuolization in more than 50% or the cells. Here we show that this heterogeneity may be explained by cell cycle-specific effects. The BH3 mimetic ABT737, lithium, rapamycin, tunicamycin or nutrient depletion stereotypically induce autophagy preferentially in the G(1) and S phases of the cell cycle, as determined by simultaneous monitoring of cell cycle markers and the cytoplasmic aggregation of GFP-LC3 in autophagic vacuoles. These results point to a hitherto neglected crosstalk between autophagic vacuolization and cell cycle regulation.


Autophagy/physiology , Cell Cycle/physiology , Endoplasmic Reticulum/physiology , Mitochondria/physiology , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Endoplasmic Reticulum/pathology , HCT116 Cells , Humans , Mitochondria/pathology , Rats
14.
Autophagy ; 3(4): 374-6, 2007.
Article En | MEDLINE | ID: mdl-17438366

Beclin 1 has recently been identified as novel BH3-only protein, meaning that it carries one Bcl-2-homology-3 (BH3) domain. As other BH3-only proteins, Beclin 1 interacts with anti-apoptotic multidomain proteins of the Bcl-2 family (in particular Bcl-2 and its homologue Bcl-X(L)) by virtue of its BH3 domain, an amphipathic alpha-helix that binds to the hydrophobic cleft of Bcl-2/Bcl-X(L). The BH3 domains of other BH3-only proteins such as Bad, as well as BH3-mimetic compounds such as ABT737, competitively disrupt the inhibitory interaction between Beclin 1 and Bcl-2/Bcl-X(L). This causes autophagy of mitochondria (mitophagy) but not of the endoplasmic reticulum (reticulophagy). Only ER-targeted (not mitochondrion-targeted) Bcl-2/Bcl-X(L) can inhibit autophagy induced by Beclin 1, and only Beclin 1-Bcl-2/Bcl-X(L) complexes present in the ER (but not those present on heavy membrane fractions enriched in mitochondria) are disrupted by ABT737. These findings suggest that the Beclin 1-Bcl-2/Bcl-X(L) complexes that normally inhibit autophagy are specifically located in the ER and point to an organelle-specific regulation of autophagy. Furthermore, these data suggest a spatial organization of autophagy and apoptosis control in which BH3-only proteins exert two independent functions. On the one hand, they can induce apoptosis, by (directly or indirectly) activating the mitochondrion-permeabilizing function of pro-apoptotic multidomain proteins from the Bcl-2 family. On the other hand, they can activate autophagy by liberating Beclin 1 from its inhibition by Bcl-2/Bcl-X(L) at the level of the endoplasmic reticulum.


Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , BH3 Interacting Domain Death Agonist Protein/metabolism , Membrane Proteins/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Beclin-1 , Humans , Mitochondria/metabolism , Models, Biological , Models, Chemical
15.
EMBO J ; 26(10): 2527-39, 2007 May 16.
Article En | MEDLINE | ID: mdl-17446862

The anti-apoptotic proteins Bcl-2 and Bcl-X(L) bind and inhibit Beclin-1, an essential mediator of autophagy. Here, we demonstrate that this interaction involves a BH3 domain within Beclin-1 (residues 114-123). The physical interaction between Beclin-1 and Bcl-X(L) is lost when the BH3 domain of Beclin-1 or the BH3 receptor domain of Bcl-X(L) is mutated. Mutation of the BH3 domain of Beclin-1 or of the BH3 receptor domain of Bcl-X(L) abolishes the Bcl-X(L)-mediated inhibition of autophagy triggered by Beclin-1. The pharmacological BH3 mimetic ABT737 competitively inhibits the interaction between Beclin-1 and Bcl-2/Bcl-X(L), antagonizes autophagy inhibition by Bcl-2/Bcl-X(L) and hence stimulates autophagy. Knockout or knockdown of the BH3-only protein Bad reduces starvation-induced autophagy, whereas Bad overexpression induces autophagy in human cells. Gain-of-function mutation of the sole BH3-only protein from Caenorhabditis elegans, EGL-1, induces autophagy, while deletion of EGL-1 compromises starvation-induced autophagy. These results reveal a novel autophagy-stimulatory function of BH3-only proteins beyond their established role as apoptosis inducers. BH3-only proteins and pharmacological BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin-1 and Bcl-2 or Bcl-X(L).


Apoptosis Regulatory Proteins/chemistry , Membrane Proteins/chemistry , Proteins/chemistry , bcl-X Protein/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/ultrastructure , Autophagy , Beclin-1 , Blotting, Western , Cells, Cultured , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , HeLa Cells , Humans , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Mice , Mutation , Precipitin Tests , Protein Structure, Tertiary/genetics , Proteins/metabolism , Proteins/ultrastructure , RNA Interference , Two-Hybrid System Techniques , bcl-X Protein/ultrastructure
16.
J Biomol Screen ; 11(8): 949-58, 2006 Dec.
Article En | MEDLINE | ID: mdl-17092915

Fluorescence polarization is a screening technology that is radioactivity free, homogeneous, and ratiometric. The signal measured with this technology is a weighted value of free and bound ligand. As a consequence, saturation curves are accessible only after calculation of the corresponding concentrations of free and bound ligand. To make this technology more accessible to assay development, the authors propose a simple mathematical model that predicts fluorescence polarization values from ligand and receptor total concentrations, depending on the corresponding dissociation constant. This model was validated using data of Bodipy-NDP-alphaMSH binding to MC(5), obtained after either ligand saturation of a receptor preparation or, conversely, receptor saturation of a ligand solution. These experimental data were also used to calculate the actual concentration of free and bound ligand and receptor and to obtain pharmacological constants by Scatchard analysis. A general method is proposed, which facilitates the design of fluorescence polarization binding assays by relying on the representation of theoretical polarization values. This approach is illustrated by the application to 2 systems of very different affinities.


Fluorescence Polarization Immunoassay/methods , Models, Theoretical , Receptors, Cytoplasmic and Nuclear/metabolism , Binding Sites , Binding, Competitive , Ligands , Protein Binding
17.
Cancer Res ; 66(5): 2757-64, 2006 Mar 01.
Article En | MEDLINE | ID: mdl-16510597

A functional imbalance between proapoptotic Bax and antiapoptotic Bcl-2 is likely to participate in the resistance of cancer cells to therapy. We show here that ethyl 2-amino-6-bromo-4-(1-cyano-2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (HA14-1), a small organic compound recently proposed to function as an inhibitor of Bcl-2, increases the sensitivity of human glioblastoma cells to radiotherapy and chemotherapy. This sensitizing effect is lost if Bcl-2 expression, but not Bcl-xL expression, is knocked down or if cells only express a mutant of Bax that does not interact with Bcl-2. This points to a specific Bcl-2 inhibitory function of HA14-1 and implies that it selectively involves hindrance of Bcl-2 binding to Bax, which HA14-1 inhibits in cell-free assays and in cells in receipt of an apoptotic stimulation. Moreover, HA14-1, in combination with a cytotoxic treatment, slows down the growth of glioblastoma in vivo. Thus, the inhibition of Bcl-2 achieved by HA14-1 might improve treatment outcome.


Benzopyrans/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Nitriles/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , bcl-2-Associated X Protein/antagonists & inhibitors , Amino Acid Sequence , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Combined Modality Therapy , DNA Damage , Drug Synergism , Etoposide/pharmacology , Female , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Mice , Mice, Nude , Molecular Sequence Data , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/metabolism
18.
J Soc Biol ; 199(3): 253-65, 2005.
Article Fr | MEDLINE | ID: mdl-16471266

Programmed cell death or apoptosis is a crucial process for normal embryonic development and homeostasis. Apoptosis is known to be coupled to multiple signalling pathways. Identification of critical points in the regulation of apoptosis is of major interest both for the understanding of control of cell fate and for the discovery of new pharmacological targets, particularly in oncology. Indeed, defects in the execution of apoptosis are known to participate in tumour initiation and progression as well as in chemoresistance. The Bcl-2 family members constitute essential intracellular players in the apoptotic machinery. Those proteins are either pro or anti-apoptotic, they interact with each other to regulate apoptosis. Inhibiting the heterodimerisation between pro- and anti-apoptotic members is sufficient to promote apoptosis in mammalian cells. Small molecules, antagonists or peptidomimetics inhibiting this heterodimerisation, represent a therapeutic prototype targeting the apoptotic cascade. They induce cell death by activating directly the mitochondrial apoptotic pathway. Considerable evidence indicate that such Bcl-2 antagonists could be useful drugs to induce apoptosis preferentially in neoplastic cells.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/drug effects , Antineoplastic Agents/therapeutic use , Antisense Elements (Genetics)/therapeutic use , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/physiology , Drug Design , Flavonoids/therapeutic use , Genes, bcl-2 , Genetic Therapy , Humans , Models, Molecular , Molecular Structure , Multigene Family , Neoplasms/pathology , Neoplasms/therapy , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Phenols/therapeutic use , Polyphenols , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/physiology , Structure-Activity Relationship
20.
J Cell Biol ; 157(5): 831-8, 2002 May 27.
Article En | MEDLINE | ID: mdl-12034774

The small GTPase RhoA controls activity of serum response factor (SRF) by inducing changes in actin dynamics. We show that in PC12 cells, activation of SRF after serum stimulation is RhoA dependent, requiring both actin polymerization and the Rho kinase (ROCK)-LIM kinase (LIMK)-cofilin signaling pathway, previously shown to control F-actin turnover. Activation of SRF by overexpression of wild-type LIMK or ROCK-insensitive LIMK mutants also requires functional RhoA, indicating that a second RhoA-dependent signal is involved. This is provided by the RhoA effector mDia: dominant interfering mDia1 derivatives inhibit both serum- and LIMK-induced SRF activation and reduce the ability of LIMK to induce F-actin accumulation. These results demonstrate a role for LIMK in SRF activation, and functional cooperation between RhoA-controlled LIMK and mDia effector pathways.


Actins/metabolism , Carrier Proteins/metabolism , Neurons/enzymology , Protein Kinases/metabolism , Serum Response Factor/metabolism , Actin Depolymerizing Factors , Animals , Intracellular Signaling Peptides and Proteins , Lim Kinases , Microfilament Proteins/metabolism , Neurons/cytology , PC12 Cells , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats , rho-Associated Kinases , rhoA GTP-Binding Protein/metabolism
...