Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
Add more filters

Publication year range
1.
Arch Pharm (Weinheim) ; 357(3): e2300583, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38110703

ABSTRACT

Immunotherapy has emerged as a game-changing approach for cancer treatment. Although monoclonal antibodies (mAbs) targeting the programmed cell death protein 1/programmed cell death protein 1 ligand 1 (PD-1/PD-L1) axis have entered the market revolutionizing the treatment landscape of many cancer types, small molecules, although presenting several advantages including the possibility of oral administration and/or reduced costs, struggled to enter in clinical trials, suffering of water insolubility and/or inadequate potency compared with mAbs. Thus, the search for novel scaffolds for both the design of effective small molecules and possible synergistic strategies is an ongoing field of interest. In an attempt to find novel chemotypes, a virtual screening approach was employed, resulting in the identification of new chemical entities with a certain binding capability, the most versatile of which was the benzimidazole-containing compound 10. Through rational design, a small library of its derivatives was synthesized and evaluated. The homogeneous time-resolved fluorescence (HTRF) assay revealed that compound 17 shows the most potent inhibitory activity (IC50 ) in the submicromolar range and notably, differently from the major part of PD-L1 inhibitors, exhibits satisfactory water solubility properties. These findings highlight the potential of benzimidazole-based compounds as novel promising candidates for PD-L1 inhibition.


Subject(s)
Biphenyl Compounds , Immune Checkpoint Inhibitors , Programmed Cell Death 1 Receptor , B7-H1 Antigen , Ligands , Structure-Activity Relationship , Benzimidazoles/pharmacology , Water
3.
Molecules ; 29(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38893521

ABSTRACT

The PD-1/PD-L1 complex is an immune checkpoint responsible for regulating the natural immune response, but also allows tumors to escape immune surveillance. Inhibition of the PD-1/PD-L1 axis positively contributes to the efficacy of cancer treatment. The only available therapeutics targeting PD-1/PD-L1 are monoclonal antibody-based drugs, which have several limitations. Therefore, small molecule compounds are emerging as an attractive alternative that can potentially overcome the drawbacks of mAb-based therapy. In this article, we present a novel class of small molecule compounds based on the terphenyl scaffold that bind to PD-L1. The general architecture of the presented structures is characterized by axial symmetry and consists of three elements: an m-terphenyl core, an additional aromatic ring, and a solubilizing agent. Using molecular docking, we designed a series of final compounds, which were subsequently synthesized and tested in HTRF assay and NMR binding assay to evaluate their activity. In addition, we performed an in-depth analysis of the mutual arrangement of the phenyl rings of the terphenyl core within the binding pocket of PD-L1 and found several correlations between the plane angle values and the affinity of the compounds towards the protein.


Subject(s)
B7-H1 Antigen , Molecular Docking Simulation , Programmed Cell Death 1 Receptor , Protein Binding , Terphenyl Compounds , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/chemistry , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/chemistry , Humans , Terphenyl Compounds/chemistry , Terphenyl Compounds/pharmacology , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/pharmacology , Molecular Structure , Structure-Activity Relationship , Binding Sites
4.
Mol Cancer ; 22(1): 150, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679783

ABSTRACT

Recent advances in immuno-oncology have opened up new and impressive treatment options for cancer. Notwithstanding, overcoming the limitations of the current FDA-approved therapies with monoclonal antibodies (mAbs) that block the PD-1/PD-L1 pathway continues to lead to the testing of multiple approaches and optimizations. Recently, a series of macrocyclic peptides have been developed that exhibit binding strengths to PD-L1 ranging from sub-micromolar to micromolar. In this study, we present the most potent non-antibody-based PD-1/PD-L1 interaction inhibitor reported to date. The structural and biological characterization of this macrocyclic PD-L1 targeting peptide provides the rationale for inhibition of both PD-1/PD-L1 and CD80/PD-L1 complexes. The IC50 and EC50 values obtained in PD-L1 binding assays indicate that the pAC65 peptide has potency equivalent to the current FDA-approved mAbs and may have similar activity to the BMS986189 peptide, which entered the clinical trial and has favorable safety and pharmacokinetic data. The data presented here delineate the generation of similar peptides with improved biological activities and applications not only in the field of cancer immunotherapy but also in other disorders related to the immune system.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Humans , Antibodies, Monoclonal/pharmacology , Immune Checkpoint Inhibitors , Peptides/pharmacology
5.
Chembiochem ; 24(6): e202300006, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36602436

ABSTRACT

Nutlin-3a is a reversible inhibitor of the p53/MDM2 interaction. We have synthesized the derivative Nutlin-3a-aa bearing an additional exocyclic methylene group in the piperazinone moiety. Nutlin-3a-aa is more active than Nutlin-3a against purified wild-type MDM2, and is more effective at increasing p53 levels and releasing transcription of p53 target genes from MDM2-induced repression. X-ray analysis of wild-type MDM2-bound Nutlin-3a-aa indicated that the orientation of its modified piperazinone ring was altered in comparison to the piperazinone ring of MDM2-bound Nutlin-3a, with the exocyclic methylene group of Nutlin-3a-aa pointing away from the protein surface. Our data point to the introduction of exocyclic methylene groups as a useful approach by which to tailor the conformation of bioactive molecules for improved biological activity.


Subject(s)
Antineoplastic Agents , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-mdm2 , Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Imidazoles/metabolism , Cell Line, Tumor , Apoptosis
6.
Molecules ; 28(22)2023 Nov 10.
Article in English | MEDLINE | ID: mdl-38005242

ABSTRACT

Lysosome-targeting chimeras (LYTACs) have recently been developed to facilitate the lysosomal degradation of specific extracellular and transmembrane molecular targets. However, the LYTAC particles described to date are based on glycopeptide conjugates, which are difficult to prepare and produce on a large scale. Here, we report on the development of pure protein LYTACs based on the non-glycosylated IGF2 peptides, which can be readily produced in virtually any facility capable of monoclonal antibody production. These chimeras utilize the IGF2R/CI-M6PR pathway for lysosomal shuttling and, in our illustrative example, target programmed death ligand 1 (PD-L1), eliciting physiological effects analogous to immune checkpoint blockade. Results from in vitro assays significantly exceed the effects of anti-PD-L1 antibodies alone.


Subject(s)
Antibodies, Monoclonal , Peptides , Peptides/chemistry , Antibodies, Monoclonal/metabolism , Glycopeptides/metabolism , Membrane Proteins/metabolism , Lysosomes/metabolism
7.
Cell Commun Signal ; 20(1): 10, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35057808

ABSTRACT

BACKGROUND: A universal adaptor protein, MyD88, orchestrates the innate immune response by propagating signals from toll-like receptors (TLRs) and interleukin-1 receptor (IL-1R). Receptor activation seeds MyD88 dependent formation of a signal amplifying supramolecular organizing center (SMOC)-the myddosome. Alternatively spliced variant MyD88S, lacking the intermediate domain (ID), exhibits a dominant negative effect silencing the immune response, but the mechanistic understanding is limited. METHODS: Luciferase reporter assay was used to evaluate functionality of MyD88 variants and mutants. The dimerization potential of MyD88 variants and myddosome nucleation process were monitored by co-immunoprecipitation and confocal microscopy. The ID secondary structure was characterized in silico employing I-TASSER server and in vitro using nuclear magnetic resonance (NMR) and circular dichroism (CD). RESULTS: We show that MyD88S is recruited to the nucleating SMOC and inhibits its maturation by interfering with incorporation of additional components. Biophysical analysis suggests that important functional role of ID is not supported by a well-defined secondary structure. Mutagenesis identifies Tyr116 as the only essential residue within ID required for myddosome nucleation and signal propagation (NF-κB activation). CONCLUSIONS: Our results argue that the largely unstructured ID of MyD88 is not only a linker separating toll-interleukin-1 receptor (TIR) homology domain and death domain (DD), but contributes intermolecular interactions pivotal in MyD88-dependent signaling. The dominant negative effect of MyD88S relies on quenching the myddosome nucleation and associated signal transduction. Video abstract.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Myeloid Differentiation Factor 88/metabolism , Cell Line , Humans , Interleukin-1 Receptor-Associated Kinases/chemistry , Interleukin-1 Receptor-Associated Kinases/metabolism , Myeloid Differentiation Factor 88/genetics , Protein Structure, Tertiary , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Toll-Like Receptors/metabolism
8.
Molecules ; 27(23)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36500376

ABSTRACT

By binding to the spliceosomal protein Snu66, the human ubiquitin-like protein Hub1 is a modulator of the spliceosome performance and facilitates alternative splicing. Small molecules that bind to Hub1 would be of interest to study the protein-protein interaction of Hub1/Snu66, which is linked to several human pathologies, such as hypercholesterolemia, premature aging, neurodegenerative diseases, and cancer. To identify small molecule ligands for Hub1, we used the interface analysis, peptide modeling of the Hub1/Snu66 interaction and the fragment-based NMR screening. Fragment-based NMR screening has not proven sufficient to unambiguously search for fragments that bind to the Hub1 protein. This was because the Snu66 binding pocket of Hub1 is occupied by pH-sensitive residues, making it difficult to distinguish between pH-induced NMR shifts and actual binding events. The NMR analyses were therefore verified experimentally by microscale thermophoresis and by NMR pH titration experiments. Our study found two small peptides that showed binding to Hub1. These peptides are the first small-molecule ligands reported to interact with the Hub1 protein.


Subject(s)
Alternative Splicing , Spliceosomes , Humans , Spliceosomes/metabolism , Ubiquitins/genetics , Magnetic Resonance Spectroscopy , Computers , Protein Binding , Ligands , Binding Sites
9.
Molecules ; 27(11)2022 May 27.
Article in English | MEDLINE | ID: mdl-35684392

ABSTRACT

New biphenyl-based chimeric compounds containing pomalidomide were developed and evaluated for their activity to inhibit and degrade the programmed cell death-1/programmed cell death- ligand 1 (PD-1/PD-L1) complex. Most of the compounds displayed excellent inhibitory activity against PD-1/PD-L1, as assessed by the homogenous time-resolved fluorescence (HTRF) binding assay. Among them, compound 3 is one of the best with an IC50 value of 60 nM. Using an ex vivo PD-1/PD-L1 blockade cell line bioassay that expresses human PD-1 and PD-L1, we show that compounds 4 and 5 significantly restore the repressed immunity in this co-culture model. Western blot data, however, demonstrated that these anti-PD-L1/pomalidomide chimeras could not reduce the protein levels of PD-L1.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Thalidomide , B7-H1 Antigen/antagonists & inhibitors , Biphenyl Compounds , Humans , Ligands , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
10.
Int J Mol Sci ; 22(21)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34769226

ABSTRACT

Targeting the programmed cell death protein 1/programmed cell death 1 ligand 1 (PD-1/PD-L1) interaction has become an established strategy for cancer immunotherapy. Although hundreds of small-molecule, peptide, and peptidomimetic inhibitors have been proposed in recent years, only a limited number of drug candidates show good PD-1/PD-L1 blocking activity in cell-based assays. In this article, we compare representative molecules from different classes in terms of their PD-1/PD-L1 dissociation capacity measured by HTRF and in vitro bioactivity determined by the immune checkpoint blockade (ICB) co-culture assay. We point to recent discoveries that underscore important differences in the mechanisms of action of these molecules and also indicate one principal feature that needs to be considered, which is the eventual human PD-L1 specificity.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors , Peptidomimetics , Animals , B7-H1 Antigen/metabolism , CHO Cells , Cricetulus , Drug Evaluation , Humans , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/pharmacology , Jurkat Cells , Peptidomimetics/chemistry , Peptidomimetics/pharmacology
11.
Molecules ; 26(16)2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34443436

ABSTRACT

The clinical success of PD-1/PD-L1 immune checkpoint targeting antibodies in cancer is followed by efforts to develop small molecule inhibitors with better penetration into solid tumors and more favorable pharmacokinetics. Here we report the crystal structure of a macrocyclic peptide inhibitor (peptide 104) in complex with PD-L1. Our structure shows no indication of an unusual bifurcated binding mode demonstrated earlier for another peptide of the same family (peptide 101). The binding mode relies on extensive hydrophobic interactions at the center of the binding surface and an electrostatic patch at the side. An interesting sulfur/π interaction supports the macrocycle-receptor binding. Overall, our results allow a better understanding of forces guiding macrocycle affinity for PD-L1, providing a rationale for future structure-based inhibitor design and rational optimization.


Subject(s)
B7-H1 Antigen/metabolism , Immune Checkpoint Proteins/metabolism , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Peptides/chemistry , Peptides/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Animals , CHO Cells , Cricetulus , Humans , Jurkat Cells , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding
12.
Molecules ; 25(13)2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32630327

ABSTRACT

Screening for small-molecule fragments that can lead to potent inhibitors of protein-protein interactions (PPIs) is often a laborious step as the fragments cannot dissociate the targeted PPI due to their low µM-mM affinities. Here, we describe an NMR competition assay called w-AIDA-NMR (weak-antagonist induced dissociation assay-NMR), which is sensitive to weak µM-mM ligand-protein interactions and which can be used in initial fragment screening campaigns. By introducing point mutations in the complex's protein that is not targeted by the inhibitor, we lower the effective affinity of the complex, allowing for short fragments to dissociate the complex. We illustrate the method with the compounds that block the Mdm2/X-p53 and PD-1/PD-L1 oncogenic interactions. Targeting the PD-/PD-L1 PPI has profoundly advanced the treatment of different types of cancers.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Magnetic Resonance Spectroscopy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Interaction Maps/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , B7-H1 Antigen/metabolism , Humans , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
13.
Angew Chem Int Ed Engl ; 59(13): 5235-5241, 2020 03 23.
Article in English | MEDLINE | ID: mdl-31944488

ABSTRACT

Stapled peptides are chemical entities in-between biologics and small molecules, which have proven to be the solution to high affinity protein-protein interaction antagonism, while keeping control over pharmacological performance such as stability and membrane penetration. We demonstrate that the multicomponent reaction-based stapling is an effective strategy for the development of α-helical peptides with highly potent dual antagonistic action of MDM2 and MDMX binding p53. Such a potent inhibitory activity of p53-MDM2/X interactions was assessed by fluorescence polarization, microscale thermophoresis, and 2D NMR, while several cocrystal structures with MDM2 were obtained. This MCR stapling protocol proved efficient and versatile in terms of diversity generation at the staple, as evidenced by the incorporation of both exo- and endo-cyclic hydrophobic moieties at the side chain cross-linkers. The interaction of the Ugi-staple fragments with the target protein was demonstrated by crystallography.


Subject(s)
Peptides/chemistry , Peptides/metabolism , Proto-Oncogene Proteins c-mdm2/chemistry , Tumor Suppressor Protein p53/chemistry , Aldehydes/chemistry , Amines/chemistry , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Cyanides/chemistry , Fluorescence Polarization , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Conformation
14.
Bioorg Chem ; 82: 284-289, 2019 02.
Article in English | MEDLINE | ID: mdl-30396062

ABSTRACT

The cell-surface protein CD44, a primary receptor for hyaluronic acid (HA), is one of the most promising targets for cancer therapies. It is prominently involved in the process of tumor growth and metastasis. The possibility of modulating the CD44-HA interaction with a pharmacological inhibitor is therefore of great importance, yet until now there are only few small molecules reported to bind to CD44. Here, we describe the results of the NMR fragment-based screening conducted against CD44 by which we found eight new hit compounds that bind to the receptor with the affinity in milimolar range. The NMR-based characterization revealed that there are two possible binding modes for these compounds, and for some of them the binding is no longer possible in the presence of hyaluronic acid. This could provide an interesting starting point for the development of new high-affinity ligands targeting the CD44-HA axis.


Subject(s)
Aniline Compounds/metabolism , Hyaluronan Receptors/metabolism , Thiazoles/metabolism , Aniline Compounds/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Hyaluronan Receptors/chemistry , Hyaluronic Acid/chemistry , Ligands , Protein Binding , Proton Magnetic Resonance Spectroscopy , Thiazoles/chemistry
15.
Molecules ; 24(11)2019 May 30.
Article in English | MEDLINE | ID: mdl-31151293

ABSTRACT

Cancer immunotherapy based on antibodies targeting the immune checkpoint PD-1/PD-L1 pathway has seen unprecedented clinical responses and constitutes the new paradigm in cancer therapy. The antibody-based immunotherapies have several limitations such as high production cost of the antibodies or their long half-life. Small-molecule inhibitors of the PD-1/PD-L1 interaction have been highly anticipated as a promising alternative or complementary therapeutic to the monoclonal antibodies (mAbs). Currently, the field of developing anti-PD-1/PD-L1 small-molecule inhibitors is intensively explored. In this paper, we review anti-PD-1/PD-L1 small-molecule and peptide-based inhibitors and discuss recent structural and preclinical/clinical aspects of their development. Discovery of the therapeutics based on small-molecule inhibitors of the PD-1/PD-L1 interaction represents a promising but challenging perspective in cancer treatment.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/metabolism , Drug Development , Peptides/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents, Immunological/chemistry , Drug Development/methods , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Peptides/chemistry , Protein Binding/drug effects , Quantitative Structure-Activity Relationship , Signal Transduction/drug effects
16.
Molecules ; 24(15)2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31374878

ABSTRACT

CA-170 is currently the only small-molecule modulator in clinical trials targeting PD-L1 and VISTA proteins - important negative checkpoint regulators of immune activation. The reported therapeutic results to some extent mimic those of FDA-approved monoclonal antibodies overcoming the limitations of the high production costs and adverse effects of the latter. However, no conclusive biophysical evidence proving the binding to hPD-L1 has ever been presented. Using well-known in vitro methods: NMR binding assay, HTRF and cell-based activation assays, we clearly show that there is no direct binding between CA-170 and PD-L1. To strengthen our reasoning, we performed control experiments on AUNP-12 - a 29-mer peptide, which is a precursor of CA-170. Positive controls consisted of the well-documented small-molecule PD-L1 inhibitors: BMS-1166 and peptide-57.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immunotherapy , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , B7 Antigens/antagonists & inhibitors , B7 Antigens/chemistry , B7-H1 Antigen/chemistry , Humans , Magnetic Resonance Spectroscopy , Neoplasms/immunology , Protein Binding/drug effects
17.
Beilstein J Org Chem ; 15: 513-520, 2019.
Article in English | MEDLINE | ID: mdl-30873235

ABSTRACT

Macrocycles were designed to antagonize the protein-protein interaction p53-MDM2 based on the three-finger pharmacophore F19W23L25. The synthesis was accomplished by a rapid, one-pot synthesis of indole-based macrocycles based on Ugi macrocyclization. The reaction of 12 different α,ω-amino acids and different indole-3-carboxaldehyde derivatives afforded a unique library of macrocycles otherwise difficult to access. Screening of the library for p53-MDM2 inhibition by fluorescence polarization and 1H,15N HSQC NMR measurements confirm MDM2 binding.

18.
Int J Mol Sci ; 19(9)2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30134556

ABSTRACT

Fibroblast growth factor 1 (FGF1) and its receptors (FGFRs) regulate crucial biological processes such as cell proliferation and differentiation. Aberrant activation of FGFRs by their ligands can promote tumor growth and angiogenesis in many tumor types, including lung or breast cancer. The development of FGF1-targeting molecules with potential implications for the therapy of FGF1-driven tumors is recently being considered a promising approach in the treatment of cancer. In this study we have used phage display selection to find scFv antibody fragments selectively binding FGF1 and preventing it from binding to its receptor. Three identified scFv clones were expressed and characterized with regard to their binding to FGF1 and ability to interfere with FGF1-induced signaling cascades activation. In the next step the scFvs were cloned to scFv-Fc format, as dimeric Fc fusions prove beneficial in prospective therapeutic application. As expected, scFvs-Fc exhibited significantly increased affinity towards FGF1. We observed strong antiproliferative activity of the scFvs and scFvs-Fc in the in vitro cell models. Presented antibody fragments serve as novel FGF1 inhibitors and can be further utilized as powerful tools to use in the studies on the selective cancer therapy.


Subject(s)
Fibroblast Growth Factor 1/antagonists & inhibitors , Peptide Library , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Clone Cells , Cloning, Molecular , Cross Reactions , Escherichia coli/genetics , Escherichia coli/metabolism , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Mice , NIH 3T3 Cells , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Signal Transduction , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics
19.
Angew Chem Int Ed Engl ; 57(18): 4840-4848, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29178534

ABSTRACT

The PD-1/PD-L1 interaction has emerged as a significant target in cancer immunotherapy. Current medications include monoclonal antibodies, which have shown impressive clinical results in the treatment of several types of tumors. The cocrystal structure of human PD-1 and PD-L1 is expected to be a valuable starting point for the design of novel inhibitors, along with the recent crystal structures with monoclonal antibodies, small molecules, and macrocycles.


Subject(s)
Antibodies, Monoclonal/immunology , B7-H1 Antigen/chemistry , B7-H1 Antigen/immunology , Drug Design , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Humans , Immunotherapy , Ligands , Programmed Cell Death 1 Receptor/antagonists & inhibitors
20.
Nature ; 474(7350): 173-8, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21614000

ABSTRACT

Alternative splicing of pre-messenger RNAs diversifies gene products in eukaryotes and is guided by factors that enable spliceosomes to recognize particular splice sites. Here we report that alternative splicing of Saccharomyces cerevisiae SRC1 pre-mRNA is promoted by the conserved ubiquitin-like protein Hub1. Structural and biochemical data show that Hub1 binds non-covalently to a conserved element termed HIND, which is present in the spliceosomal protein Snu66 in yeast and mammals, and Prp38 in plants. Hub1 binding mildly alters spliceosomal protein interactions and barely affects general splicing in S. cerevisiae. However, spliceosomes that lack Hub1, or are defective in Hub1-HIND interaction, cannot use certain non-canonical 5' splice sites and are defective in alternative SRC1 splicing. Hub1 confers alternative splicing not only when bound to HIND, but also when experimentally fused to Snu66, Prp38, or even the core splicing factor Prp8. Our study indicates a novel mechanism for splice site utilization that is guided by non-covalent modification of the spliceosome by an unconventional ubiquitin-like modifier.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Fungal , Ligases/metabolism , RNA Splice Sites/genetics , RNA, Fungal/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , Gene Deletion , Humans , Ligases/deficiency , Ligases/genetics , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/deficiency , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/deficiency , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/deficiency , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Spliceosomes/chemistry , Spliceosomes/metabolism , Ubiquitin-Protein Ligase Complexes/deficiency , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitins
SELECTION OF CITATIONS
SEARCH DETAIL