Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
ACS Chem Biol ; 19(1): 101-109, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38069818

ABSTRACT

Protein-protein interactions (PPIs) are intriguing targets in drug discovery and development. Peptides are well suited to target PPIs, which typically present with large surface areas lacking distinct features and deep binding pockets. To improve binding interactions with these topologies and advance the development of PPI-focused therapeutics, potential ligands can be equipped with electrophilic groups to enable binding through covalent mechanisms of action. We report a strategy termed electrophile scanning to identify reactivity hotspots in a known peptide ligand and demonstrate its application in a model PPI. Cysteine mutants of a known ligand are used to install protein-reactive modifiers via a palladium oxidative addition complex (Pd-OAC). Reactivity hotspots are revealed by cross-linking reactions with the target protein under physiological conditions. In a model PPI with the 9-mer peptide antigen VL9 and major histocompatibility complex (MHC) class I protein HLA-E, we identify two reactivity hotspots that afford up to 87% conversion to the protein-peptide conjugate within 4 h. The reactions are specific to the target protein in vitro and dependent on the peptide sequence. Moreover, the cross-linked peptide successfully inhibits molecular recognition of HLA-E by CD94-NKG2A possibly due to structural changes enacted at the PPI interface. The results illustrate the potential application of electrophile scanning as a tool for rapid discovery and development of covalent peptide binders.


Subject(s)
HLA-E Antigens , Histocompatibility Antigens Class I , Ligands , Histocompatibility Antigens Class I/metabolism , Peptides/chemistry , Protein Binding
2.
Chem Sci ; 14(44): 12484-12497, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38020382

ABSTRACT

Human papillomavirus (HPV) infections account for nearly all cervical cancer cases, which is the fourth most common cancer in women worldwide. High-risk variants, including HPV16, drive tumorigenesis in part by promoting the degradation of the tumor suppressor p53. This degradation is mediated by the HPV early protein 6 (E6), which recruits the E3 ubiquitin ligase E6AP and redirects its activity towards ubiquitinating p53. Targeting the protein interaction interface between HPV E6 and E6AP is a promising modality to mitigate HPV-mediated degradation of p53. In this study, we designed a covalent peptide inhibitor, termed reactide, that mimics the E6AP LXXLL binding motif by selectively targeting cysteine 58 in HPV16 E6 with quantitative conversion. This reactide provides a starting point in the development of covalent peptidomimetic inhibitors for intervention against HPV-driven cancers.

3.
Science ; 379(6635): 939-945, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36862767

ABSTRACT

Encoding small-molecule information in DNA has been leveraged to accelerate the discovery of ligands for therapeutic targets such as proteins. However, oligonucleotide-based encoding is hampered by inherent limitations of information stability and density. In this study, we establish abiotic peptides for next-generation information storage and apply them for the encoding of diverse small-molecule synthesis. The chemical stability of the peptide-based tag allows the use of palladium-mediated reactions to efficiently synthesize peptide-encoded libraries (PELs) with broad chemical diversity and high purity. We demonstrate the successful de novo discovery of small-molecule protein ligands from PELs by affinity selection against carbonic anhydrase IX and the oncogenic protein targets BRD4(1) and MDM2. Collectively, this work establishes abiotic peptides as carriers of information for the encoding of small-molecule synthesis, leveraged herein for the discovery of protein ligands.


Subject(s)
Drug Discovery , Peptide Library , Peptides , Small Molecule Libraries , Ligands , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptides/chemical synthesis , Peptides/chemistry , Transcription Factors/chemistry , Transcription Factors/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Protein Stability , Carbonic Anhydrase IX
4.
Cancers (Basel) ; 13(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34071914

ABSTRACT

The cholecystokinin-2 receptor (CCK2R) is an attractive target in nuclear medicine due to its overexpression by different tumors. Several radiolabeled peptidic ligands targeting the CCK2R have been investigated in the past; however, their low stability against proteases can limit their uptake in tumors and metastases. Substitution of single or multiple amide bonds with metabolically stable 1,4-disubstituted 1,2,3-triazoles as amide bond bioisosteres proved a promising strategy for improving the tumor-targeting properties of a truncated analog of minigastrin. In this study, we applied the previously studied structural modifications to improve the pharmacokinetic and pharmacodynamic properties of PP-F11N, a minigastrin analog currently in clinical trials. Novel minigastrins (NMGs) as analogs of PP-F11N with one or two amide bonds substituted by 1,2,3-triazoles were synthesized, radiolabeled with 177Lu3+, and subjected to full evaluation in vitro (cell internalization, receptor affinity, stability in blood plasma) and in vivo (stability, biodistribution, SPECT/CT imaging). NMGs with triazoles inserted between the amino acids DGlu10-Ala11 and/or Tyr12-Gly13 showed a significantly increased cellular uptake and affinity toward the CCK2R in vitro. Resistance against the metabolic degradation of the NMGs was comparable to those of the clinical candidate PP-F11N. Imaging by SPECT/CT and biodistribution studies demonstrated a higher uptake in CCK2R-positive tumors but also in the CCK2R-positive stomach. The peptidomimetic compounds showed a slow tumor washout and high tumor-to-kidney ratios. The structural modifications led to the identification of analogs with promising properties for progression to clinical applications in the diagnosis and therapy of CCK2R-positive neoplasms.

5.
ACS Med Chem Lett ; 12(4): 585-592, 2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33859799

ABSTRACT

1,5-Disubstituted 1,2,3-triazoles (1,5-Tz) are considered bioisosteres of cis-amide bonds. However, their use for enhancing the pharmacological properties of peptides or proteins is not yet well established. Aiming to illustrate their utility, we chose the peptide conjugate [Nle15]MG11 (DOTA-dGlu-Ala-Tyr-Gly-Trp-Nle-Asp-Phe-NH2) as a model compound since it is known that the cholecystokinin-2 receptor (CCK2R) is able to accommodate turn conformations. Analogs of [Nle15]MG11 incorporating 1,5-Tz in the backbone were synthesized and radiolabeled with lutetium-177, and their pharmacological properties (cell internalization, receptor binding affinity and specificity, plasma stability, and biodistribution) were evaluated and compared with [Nle15]MG11 as well as their previously reported analogs bearing 1,4-disubstituted 1,2,3-triazoles. Our investigations led to the discovery of novel triazole-modified analogs of [Nle15]MG11 with nanomolar CCK2R-binding affinity and 2-fold increased tumor uptake. This study illustrates that substitution of amides by 1,5-disubstituted 1,2,3-triazoles is an effective strategy to enhance the pharmacological properties of biologically active peptides.

6.
Chemistry ; 26(47): 10690-10694, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32691857

ABSTRACT

Mutating the side-chains of amino acids in a peptide ligand, with unnatural amino acids, aiming to mitigate its short half-life is an established approach. However, it is hypothesized that mutating specific backbone peptide bonds with bioisosters can be exploited not only to enhance the proteolytic stability of parent peptides, but also to tune its receptor subtype selectivity. Towards this end, four [Y]6 -Angiotensin II analogues are synthesized where amide bonds have been replaced by 1,4-disubstituted 1,2,3-triazole isosteres in four different backbone locations. All the analogues possessed enhanced stability in human plasma in comparison with the parent peptide, whereas only two of them achieved enhanced AT2 R/AT1 R subtype selectivity. This diversification has been studied through 2D NMR spectroscopy and unveiled a putative more structured microenvironment for the two selective ligands accompanied with increased number of NOE cross-peaks. The most potent analogue, compound 2, has been explored regarding its neurotrophic potential and resulted in an enhanced neurite growth with respect to the established agent C21.


Subject(s)
Angiotensin II/chemistry , Angiotensin II/metabolism , Mutation , Peptides/genetics , Receptors, Angiotensin/chemistry , Receptors, Angiotensin/metabolism , Amino Acids/genetics , Angiotensin II/genetics , Animals , HEK293 Cells , Humans , Ligands , Peptides/chemistry , Peptides/metabolism , Substrate Specificity
7.
J Med Chem ; 63(9): 4496-4505, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32302130

ABSTRACT

The insertion of single 1,4-disubstituted 1,2,3-triazoles as metabolically stable bioisosteres of trans-amide bonds (triazole scan) was recently applied to the 177Lu-labeled tumor-targeting analog of minigastrin, [Nle15]MG11. The reported novel mono-triazolo-peptidomimetics of [Nle15]MG11 showed either improved resistance against enzymatic degradation or a significantly increased affinity toward the target receptor but never both. To enhance further the tumor-targeting properties of the minigastrin analogs, we studied conjugates with multiple amide-to-triazole substitutions for additive or synergistic effects. Promising candidates were identified by modification of two or three amide bonds, which yielded both improved stability and increased receptor affinity of the peptidomimetics in vitro. Biodistribution studies of radiolabeled multi-triazolo-peptidomimetics in mice bearing receptor-positive tumor xenografts revealed up to 4-fold increased tumor uptake in comparison to the all-amide reference compound [Nle15]MG11. In addition, we report here for the first time a linear peptidomimetic with three triazole insertions in its backbone and maintained biological activity.


Subject(s)
Antineoplastic Agents/pharmacology , Gastrins/pharmacology , Peptidomimetics/pharmacology , Radiopharmaceuticals/pharmacology , Triazoles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Design , Gastrins/chemical synthesis , Gastrins/metabolism , Gastrins/pharmacokinetics , Humans , Lutetium/chemistry , Mice , Neoplasms/metabolism , Peptidomimetics/chemical synthesis , Peptidomimetics/metabolism , Peptidomimetics/pharmacokinetics , Protein Binding , Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Receptor, Cholecystokinin B/metabolism , Triazoles/chemical synthesis , Triazoles/metabolism , Triazoles/pharmacokinetics
8.
J Med Chem ; 63(9): 4484-4495, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32302139

ABSTRACT

MG11 is a truncated analog of minigastrin, a peptide with high affinity and specificity toward the cholecystokinin-2 receptor (CCK2R), which is overexpressed by different tumors. Thus, radiolabeled MG11 derivatives have great potential for use in cancer diagnosis and therapy. A drawback of MG11 is its fast degradation by proteases, leading to moderate tumor uptake in vivo. We introduced 1,4-disubstituted 1,2,3-triazoles as metabolically stable bioisosteres to replace labile amide bonds of the peptide. The "triazole scan" yielded peptidomimetics with improved resistance to enzymatic degradation and/or enhanced affinity toward the CCK2R. Remarkably, our lead compound achieved a 10-fold increase in receptor affinity, resulting in a 2.6-fold improved tumor uptake in vivo. Modeling of the ligand-CCK2R complex suggests that an additional cation-π interaction of the aromatic triazole moiety with the Arg356 residue of the receptor is accountable for these observations. We show for the first time that the amide-to-triazole substitution strategy offers new opportunities in drug development that go beyond the metabolic stabilization of bioactive peptides.


Subject(s)
Antineoplastic Agents/pharmacology , Gastrins/pharmacology , Peptidomimetics/pharmacology , Radiopharmaceuticals/pharmacology , Triazoles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Female , Gastrins/chemical synthesis , Gastrins/metabolism , Gastrins/pharmacokinetics , Humans , Lutetium/chemistry , Mice , Neoplasms/metabolism , Peptidomimetics/chemical synthesis , Peptidomimetics/metabolism , Peptidomimetics/pharmacokinetics , Protein Binding , Protein Conformation , Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Receptor, Cholecystokinin B/metabolism , Triazoles/chemical synthesis , Triazoles/metabolism , Triazoles/pharmacokinetics
10.
J Pept Sci ; 23(1): 38-44, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28054429

ABSTRACT

Radiolabelled peptides with high specificity and affinity towards receptors that are overexpressed by tumour cells are used in nuclear medicine for the diagnosis (imaging) and therapy of cancer. In some cases, the sequences of peptides under investigations contain methionine (Met), an amino acid prone to oxidation during radiolabelling procedures. The formation of oxidative side products can affect the purity of the final radiopharmaceutical product and/or impair its specificity and affinity towards the corresponding receptor. The replacement of Met with oxidation resistant amino acid analogues, for example, norleucine (Nle), can provide a solution. While this approach has been applied successfully to different radiolabelled peptides, a Met → Nle switch only preserves the length of the amino acid side chain important for hydrophobic interactions but not its hydrogen-bonding properties. We report here the use of methoxinine (Mox), a non-canonical amino acid that resembles more closely the electronic properties of Met in comparison to Nle. Specifically, we replaced Met15 by Mox15 and Nle15 in the binding sequence of a radiometal-labelled human gastrin derivative [d-Glu10 ]HG(10-17), named MG11 (d-Glu-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH2 ). A comparison of the physicochemical properties of 177 Lu-DOTA[X15 ]MG11 (X = Met, Nle, Mox) in vitro (cell internalization/externalization properties, receptor affinity (IC50 ), blood plasma stability and logD) showed that Mox indeed represents a suitable, oxidation-stable amino acid substitute of Met in radiolabelled peptide conjugates. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Gastrins/chemical synthesis , Heterocyclic Compounds, 1-Ring/chemistry , Homoserine/analogs & derivatives , Lutetium/chemistry , Oligopeptides/chemical synthesis , Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Amino Acid Substitution , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gastrins/metabolism , Gastrins/pharmacology , Heterocyclic Compounds, 1-Ring/metabolism , Heterocyclic Compounds, 1-Ring/pharmacology , Homoserine/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Isotope Labeling , Methionine/chemistry , Norleucine/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacology , Oxidation-Reduction , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacology , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL