Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Angew Chem Int Ed Engl ; 56(44): 13732-13735, 2017 10 23.
Article in English | MEDLINE | ID: mdl-28881104

ABSTRACT

Blockade of the immunoinhibitory PD-1/PD-L1 pathway using monoclonal antibodies has shown impressive results with durable clinical antitumor responses. Anti-PD-1 and anti-PD-L1 antibodies have now been approved for the treatment of a number of tumor types, whereas the development of small molecules targeting immune checkpoints lags far behind. We characterized two classes of macrocyclic-peptide inhibitors directed at the PD-1/PD-L1 pathway. We show that these macrocyclic compounds act by directly binding to PD-L1 and that they are capable of antagonizing PD-L1 signaling and, similarly to antibodies, can restore the function of T-cells. We also provide the crystal structures of two of these small-molecule inhibitors bound to PD-L1. The structures provide a rationale for the checkpoint inhibition by these small molecules, and a description of their small molecule/PD-L1 interfaces provides a blueprint for the design of small-molecule inhibitors of the PD-1/PD-L1 pathway.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , B7-H1 Antigen/immunology , Drug Discovery , Humans , Jurkat Cells , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Molecular Docking Simulation , Programmed Cell Death 1 Receptor/immunology , Protein Interaction Maps/drug effects , T-Lymphocytes/immunology
12.
Expert Opin Ther Pat ; 34(8): 627-650, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38903044

ABSTRACT

INTRODUCTION: PD-L1, via its interactions with PD-1, constitutes a key immune checkpoint that allows cancer cells to escape immune surveillance. Targeting PD-1/PD-L1 with monoclonal antibodies (mAbs) led to spectacular success in clinical oncology. However, the inherent limitations of mAbs and increasing findings about immune-related adverse events (iRAEs) prompted intense research in the field of small-molecule inhibitors of PD-L1. AREAS COVERED: This review covers inhibitors of PD-L1 reported in patents published in the online databases of the World Intellectual Property Organization and European Patent Office in the 2022-2023 period. This review provides a landscape of available inhibitors, including their chemical structures, activity, and stage of development. EXPERT OPINION: Small-molecule inhibitors impairing PD-L1/PD-1 interaction represent an attractive alternative to mAbs. In recent years, the field of small-molecule and macrocyclic inhibitors targeting PD-L1 has grown rapidly. The majority (if not all) of small-molecule inhibitors developed recently, similarly to their predecessors, act through a dimerization mechanism of PD-L1, followed by its internalization into the cytosol. In contrast, macrocyclic peptides act purely through a competition mechanism known as protein-protein interaction inhibitors. The ongoing clinical trials should ultimately reveal which strategy has real clinical potential and may complement or even replace mAbs-based therapies.


Subject(s)
B7-H1 Antigen , Drug Development , Immune Checkpoint Inhibitors , Neoplasms , Patents as Topic , Programmed Cell Death 1 Receptor , Humans , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/adverse effects , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Neoplasms/drug therapy , Antibodies, Monoclonal/pharmacology
13.
RSC Med Chem ; 15(4): 1210-1215, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38665826

ABSTRACT

The progress in cancer survival and treatment has witnessed a remarkable transformation through the innovative approach of targeting the inhibitory immune checkpoint protein PD-1/PD-L1 complex by mAbs, e.g. pembrolizumab (Keytruda). While generating 17.2 billion U.S. dollars in revenue in 2021, the true significance of these developments lies in their ability to enhance cancer patient outcomes. Despite the proven efficacy of mAbs in inhibiting the PD-1/PD-L1 signaling pathways, they face significant challenges, including limited response rates, high production costs, missing oral bioavailability, and extended half-lives that can lead to immune-related adverse effects. A promising alternative approach involves the use of small molecules acting as PD-1/PD-L1 antagonists to stimulate PD-L1 dimerization. However, the precise mechanisms of action of these molecules remain partially understood, posing challenges to their development. In this context, our research focuses on the creation of a novel scaffold based on the Ugi tetrazole four-component reaction (UT-4CR) to develop low-molecular-weight inhibitors of PD-L1. Employing structure-based methods, we synthesized a library of small compounds using biphenyl vinyl isocyanide, leading to the discovery of a structure-activity relationship among 1,5-disubstituted tetrazole-based inhibitors. Supported by a cocrystal structure with PD-L1, these inhibitors underwent biophysical testing, including HTRF and protein NMR experiments, resulting in the identification of potent candidates with sub-micromolar PD-L1 affinities. This finding opens opportunities to the further development of a new class of PD-L1 antagonists, holding promise for improved cancer immunotherapy strategies.

14.
Transl Oncol ; 42: 101892, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38359715

ABSTRACT

The PD-1/PD-L1 complex belongs to the group of inhibitory immune checkpoints and plays a critical role in immune regulation. The PD-1/PD-L1 axis is also responsible for immune evasion of cancer cells, and this complex is one of the main targets of immunotherapies used in oncology. Treatment using immune checkpoint inhibitors is mainly based on antibodies. This approach has great therapeutic potential; however, it also has major drawbacks and can induce immune-related adverse events. Thus, there is a strong need for alternative, non-antibody-based therapies using small molecules, peptides, or peptidomimetics. In the present study, we designed, synthesized, and evaluated a set of PD-1-targeting peptides based on the sequence and structure of PD-L1. The binding of these peptides to PD-1 was investigated using SPR and ELISA. We also assessed their ability to compete with PD-L1 for binding to PD-1 and their inhibitory properties against the PD-1/PD-L1 complex at the cellular level. The best results were obtained for the peptide PD-L1(111-127)(Y112C-I126C), named (L11), which displaced PD-L1 from binding to PD-1 in the competitive assay and inhibited the formation of the PD-1/PD-L1 complex. The (L11) peptide also exhibited strong affinity for PD-1. NMR studies revealed that (L11) does not form a well-defined secondary structure; however, MD simulation indicated that (L11) binds to PD-1 at the same place as PD-L1. After further optimization of the structure, the peptide inhibitor obtained in this study could also be used as a potential therapeutic compound targeting the PD-1/PD-L1 axis.

15.
Int J Biol Macromol ; 264(Pt 1): 130458, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38423421

ABSTRACT

The PD-1/PD-L1 protein-protein interaction (PPI) controls an adaptive immune resistance mechanism exerted by tumor cells to evade immune responses. The large-molecule nature of current commercial monoclonal antibodies against this PPI hampers their effectiveness by limiting tumor penetration and inducing severe immune-related side effects. Synthetic small-molecule inhibitors may overcome such limitations and have demonstrated promising clinical translation, but their design is challenging. Microbial natural products (NPs) are a source of small molecules with vast chemical diversity that have proved anti-tumoral activities, but which immunotherapeutic properties as PD-1/PD-L1 inhibitors had remained uncharacterized so far. Here, we have developed the first cell-based PD-1/PD-L1 blockade reporter assay to screen NPs libraries. In this study, 6000 microbial extracts of maximum biosynthetic diversity were screened. A secondary metabolite called alpha-cyclopiazonic acid (α-CPA) of a bioactive fungal extract was confirmed as a new PD-1/PD-L1 inhibitor with low micromolar range in the cellular assay and in an additional cell-free competitive assay. Thermal denaturation experiments with PD-1 confirmed that the mechanism of inhibition is based on its stabilization upon binding to α-CPA. The identification of α-CPA as a novel PD-1 stabilizer proves the unprecedented resolution of this methodology at capturing specific PD-1/PD-L1 PPI inhibitors from chemically diverse NP libraries.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/metabolism , Antibodies, Monoclonal
16.
ACS Med Chem Lett ; 15(1): 36-44, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38229762

ABSTRACT

Although heavily studied, the subject of anti-PD-L1 small-molecule inhibitors is still elusive. Here we present a systematic overview of the principles behind successful anti-PD-L1 small-molecule inhibitor design on the example of the m-terphenyl scaffold, with a particular focus on the neglected influence of the solubilizer tag on the overall affinity toward PD-L1. The inhibitor developed according to the proposed guidelines was characterized through its potency in blocking PD-1/PD-L1 complex formation in homogeneous time-resolved fluorescence and cell-based assays. The affinity is also explained based on the crystal structure of the inhibitor itself and its costructure with PD-L1 as well as a molecular modeling study. Our results structuralize the knowledge related to the strong pharmacophore feature of the m-terphenyl scaffold preferential geometry and the more complex role of the solubilizer tag in PD-L1 homodimer stabilization.

17.
Eur J Med Chem ; 276: 116683, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39032403

ABSTRACT

A series of novel 2-arylmethoxy-4-(2-fluoromethyl-biphenyl-3-ylmethoxy) benzylamine derivatives was designed, synthesized, and evaluated for their antitumor effects as PD-1/PD-L1 inhibitors both in vitro and in vivo. Firstly, the ability of these compounds to block the PD-1/PD-L1 immune checkpoint was assessed using the homogeneous time-resolved fluorescence (HTRF) assay. Two of the compounds can strongly block the PD-1/PD-L1 interaction, with IC50 values of less than 10 nM, notably, compound HD10 exhibited significant clinical potential by inhibiting the PD-1/PD-L1 interaction with an IC50 value of 3.1 nM. Further microscale thermophoresis (MST) analysis demonstrated that HD10 had strong interaction with PD-L1 protein. Co-crystal structure (2.7 Å) analysis of HD10 in complex with the PD-L1 protein revealed a strong affinity between the compound and the target PD-L1 dimer. This provides a solid theoretical basis for further in vitro and in vivo studies. Next, a typical cell-based experiment demonstrated that HD10 could remarkably prevent the interaction of hPD-1 293 T cells from human recombinant PD-L1 protein, effectively restoring T cell function, and promoting IFN-γ secretion in a dose-dependent manner. Moreover, HD10 was effective in suppressing tumor growth (TGI = 57.31 %) in a PD-1/PD-L1 humanized mouse model without obvious toxicity. Flow cytometry, qPCR, and immunohistochemistry data suggested that HD10 inhibits tumor growth by activating the immune system in vivo. Based on these results, it seems likely that HD10 is a promising clinical candidate that should be further investigated.


Subject(s)
Antineoplastic Agents , B7-H1 Antigen , Benzylamines , Drug Design , Drug Screening Assays, Antitumor , Programmed Cell Death 1 Receptor , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Animals , Mice , Structure-Activity Relationship , Benzylamines/pharmacology , Benzylamines/chemistry , Benzylamines/chemical synthesis , Molecular Structure , Dose-Response Relationship, Drug , Cell Proliferation/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/chemical synthesis , Immune Checkpoint Inhibitors/chemistry , Cell Line, Tumor , Female , Models, Molecular
18.
J Med Chem ; 66(15): 10579-10603, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37496104

ABSTRACT

Novel 2-arylmethoxy-4-(2,2'-dihalogen-substituted biphenyl-3-ylmethoxy) benzylamine derivatives were designed, synthesized, and evaluated in vitro and in vivo against cancers as PD-1/PD-L1 inhibitors. Through the computer-aided structural optimization and the homogeneous time-resolved fluorescence (HTRF) assay, compound A56 was found to most strongly block the PD-1/PD-L1 interaction with an IC50 value of 2.4 ± 0.8 nM and showed the most potent activity. 1H NMR titration results indicated that A56 can tightly bind to the PD-L1 protein with KD < 1 µM. The X-ray diffraction data for the cocrystal structure of the A56/PD-L1 complex (3.5 Å) deciphered a novel binding mode in detail, which can account for its most potent inhibitory activity. Cell-based assays further demonstrated the strong ability of A56 as an hPD-1/hPD-L1 blocker. Especially in an hPD-L1 MC38 humanized mouse model, A56 significantly inhibited tumor growth without obvious toxicity, with a TGI rate of 55.20% (50 mg/kg, i.g.). In conclusion, A56 is a promising clinical candidate worthy of further development.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Animals , Mice , B7-H1 Antigen , Benzylamines/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Hydrocarbons, Halogenated/chemistry , Hydrocarbons, Halogenated/pharmacology
19.
J Med Chem ; 66(14): 9577-9591, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37450644

ABSTRACT

In search of a potent small molecular PD-L1 inhibitor, we designed and synthesized a compound based on a 2-hydroxy-4-phenylthiophene-3-carbonitrile moiety. Ligand's performance was tested in vitro and compared side-by-side with a known PD-L1 antagonist with a proven bioactivity BMS1166. Subsequently, we modified both compounds to allow 18F labeling that could be used for PET imaging. Radiolabeling, which is used in drug development and diagnosis, was applied to investigate the properties of those ligands and test them against tissue sections with diverse expression levels of PD-L1. We confirmed biological activity toward hPD-L1 for this inhibitor, comparable with BMS1166, while holding enhanced pharmacological properties.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors
20.
ACS Chem Biol ; 17(9): 2655-2663, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36073782

ABSTRACT

Development of small molecules targeting the PD-L1/PD-1 interface is advancing both in industry and academia, but only a few have reached early-stage clinical trials. Here, we take a closer look at the general druggability of PD-L1 using in silico hot spot mapping and nuclear magnetic resonance (NMR)-based characterization. We found that the conformational elasticity of the PD-L1 surface strongly influences the formation of hot spots. We deconstructed several generations of known inhibitors into fragments and examined their binding properties using differential scanning fluorimetry (DSF) and protein-based nuclear magnetic resonance (NMR). These biophysical analyses showed that not all fragments bind to the PD-L1 ectodomain despite having the biphenyl scaffold. Although most of the binding fragments induced PD-L1 oligomerization, two compounds, TAH35 and TAH36, retain the monomeric state of proteins upon binding. Additionally, the presence of the entire ectodomain did not affect the binding of the hit compounds and dimerization of PD-L1. The data demonstrated here provide important information on the PD-L1 druggability and the structure-activity relationship of the biphenyl core moiety and therefore may aid in the design of novel inhibitors and focused fragment libraries for PD-L1.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , B7-H1 Antigen/metabolism , Biphenyl Compounds , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Small Molecule Libraries/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL