Discovery of CZY43 as a new small-molecule degrader of pseudokinase HER3.

  • Abstract
  • References
  • Similar Papers
Abstract
Translate article icon Translate Article Star icon
Take notes icon Take Notes

Discovery of CZY43 as a new small-molecule degrader of pseudokinase HER3.

ReferencesShowing 10 of 24 papers
  • Open Access Icon
  • Cite Count Icon 36
  • 10.15252/emmm.201911498
HER3 targeting with an antibody‐drug conjugate bypasses resistance to anti‐HER2 therapies
  • Apr 24, 2020
  • EMBO Molecular Medicine
  • Lucía Gandullo‐Sánchez + 5 more

  • Cite Count Icon 326
  • 10.1073/pnas.0912101106
Structural analysis of the catalytically inactive kinase domain of the human EGF receptor 3
  • Dec 22, 2009
  • Proceedings of the National Academy of Sciences
  • Natalia Jura + 4 more

  • Cite Count Icon 50
  • 10.1021/acs.jmedchem.3c00736
Small-Molecule Hydrophobic Tagging: A Promising Strategy of Druglike Technology for Targeted Protein Degradation.
  • Aug 3, 2023
  • Journal of Medicinal Chemistry
  • Shaowen Xie + 6 more

  • Cite Count Icon 65
  • 10.1016/j.pharmthera.2014.01.005
HER3, serious partner in crime: Therapeutic approaches and potential biomarkers for effect of HER3-targeting
  • Feb 7, 2014
  • Pharmacology & Therapeutics
  • Arjan Kol + 6 more

  • Open Access Icon
  • Cite Count Icon 67
  • 10.7554/elife.32271
Inhibitor-induced HER2-HER3 heterodimerisation promotes proliferation through a novel dimer interface.
  • May 1, 2018
  • eLife
  • Jeroen Claus + 19 more

  • Open Access Icon
  • Cite Count Icon 950
  • 10.1073/pnas.1537685100
The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation.
  • Jul 9, 2003
  • Proceedings of the National Academy of Sciences
  • Thomas Holbro + 5 more

  • Cite Count Icon 6674
  • 10.1038/35052073
Untangling the ErbB signalling network.
  • Feb 1, 2001
  • Nature Reviews Molecular Cell Biology
  • Yosef Yarden + 1 more

  • Open Access Icon
  • Cite Count Icon 35
  • 10.1080/15548627.2022.2111083
The role of LC3B in autophagy as an RNA-binding protein
  • Aug 14, 2022
  • Autophagy
  • Hyun Jung Hwang + 1 more

  • Open Access Icon
  • Cite Count Icon 184
  • 10.1038/nchembio.1658
Pharmacological targeting of the pseudokinase Her3.
  • Oct 19, 2014
  • Nature Chemical Biology
  • Ting Xie + 14 more

  • Open Access Icon
  • Cite Count Icon 91
  • 10.1158/1078-0432.ccr-20-4465
Thirty Years of HER3: From Basic Biology to Therapeutic Interventions.
  • Feb 19, 2021
  • Clinical Cancer Research
  • Heidi M Haikala + 1 more

Similar Papers
  • Research Article
  • Cite Count Icon 780
  • 10.1016/j.ccr.2010.08.009
Targeting Mitochondrial Glutaminase Activity Inhibits Oncogenic Transformation
  • Sep 1, 2010
  • Cancer Cell
  • Jian-Bin Wang + 10 more

Targeting Mitochondrial Glutaminase Activity Inhibits Oncogenic Transformation

  • Research Article
  • Cite Count Icon 27
  • 10.1016/j.ejmech.2021.113689
Platinum complexes inhibit HER-2 enriched and triple-negative breast cancer cells metabolism to suppress growth, stemness and migration by targeting PKM/LDHA and CCND1/BCL2/ATG3 signaling pathways.
  • Nov 1, 2021
  • European Journal of Medicinal Chemistry
  • Xue Bai + 7 more

Platinum complexes inhibit HER-2 enriched and triple-negative breast cancer cells metabolism to suppress growth, stemness and migration by targeting PKM/LDHA and CCND1/BCL2/ATG3 signaling pathways.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 24
  • 10.3390/cancers12102850
Protein Disulphide Isomerase A1 Is Involved in the Regulation of Breast Cancer Cell Adhesion and Transmigration via Lung Microvascular Endothelial Cells
  • Oct 2, 2020
  • Cancers
  • Marta Stojak + 11 more

Simple SummaryMetastasis is one of the most devastating aspect of cancer progression and involves biochemical and physical interactions between cancer cells and surrounding microenvironment. In particular, cancer cells adhesion to endothelium and their subsequent transendothelial migration represent important steps in the metastatic process in target organs. In this study we characterized the functional role of Protein Disulfide Isomerase A1 (PDIA1) in breast cancer cells adhesion and transendothelial migration. We identified the full repertoire of protein disulfide isomerases in endothelial cells as well as in breast cancer cells. We provided insight into the mechanisms involved in cancer-endothelial cells interactions and suggested that PDIA1 regulates the adhesion and transendothelial migration of breast cancer cells by disulphide exchange involving most likely the activation of integrins. Our results suggest that the inhibition of extracellular PDIA1 or other PDIs represents an interesting target for anti-metastatic treatment. Cancer cell cross-talk with the host endothelium plays a crucial role in metastasis, but the underlying mechanisms are still not fully understood. We studied the involvement of protein disulphide isomerase A1 (PDIA1) in human breast cancer cell (MCF-7 and MDA-MB-231) adhesion and transendothelial migration. For comparison, the role of PDIA1 in proliferation, migration, cell cycle and apoptosis was also assessed. Pharmacological inhibitor, bepristat 2a and PDIA1 silencing were used to inhibit PDIA1. Inhibition of PDIA1 by bepristat 2a markedly decreased the adhesion of breast cancer cells to collagen type I, fibronectin and human lung microvascular endothelial cells. Transendothelial migration of breast cancer cells across the endothelial monolayer was also inhibited by bepristat 2a, an effect not associated with changes in ICAM-1 expression or changes in cellular bioenergetics. The silencing of PDIA1 produced less pronounced anti-adhesive effects. However, inhibiting extracellular free thiols by non-penetrating blocker p-chloromercuribenzene sulphonate substantially inhibited adhesion. Using a proteomic approach, we identified that β1 and α2 integrins were the most abundant among all integrins in breast cancer cells as well as in lung microvascular endothelial cells, suggesting that integrins could represent a target for PDIA1. In conclusion, extracellular PDIA1 plays a major role in regulating the adhesion of cancer cells and their transendothelial migration, in addition to regulating cell cycle and caspase 3/7 activation by intracellular PDIA1. PDIA1-dependent regulation of cancer–endothelial cell interactions involves disulphide exchange and most likely integrin activation but is not mediated by the regulation of ICAM-1 expression or changes in cellular bioenergetics in breast cancer or endothelial cells.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 26
  • 10.1074/jbc.m110.197533
Disruption of a Nuclear NFATc2 Protein Stabilization Loop Confers Breast and Pancreatic Cancer Growth Suppression by Zoledronic Acid
  • Aug 1, 2011
  • Journal of Biological Chemistry
  • Shiv K Singh + 11 more

The aminobisphosphonate zoledronic acid has elicited significant attention due to its remarkable anti-tumoral activity, although its detailed mechanism of action remains unclear. Here, we demonstrate the existence of a nuclear GSK-3β-NFATc2 stabilization pathway that promotes breast and pancreatic cancer growth in vitro and in vivo and serves as a bona fide target of zoledronic acid. Specifically, the serine/threonine kinase GSK-3β stabilizes nuclear NFATc2 through phosphorylation of the serine-rich SP2 domain, thus protecting the transcription factor from E3-ubiquitin ligase HDM2-mediated proteolysis. Zoledronic acid disrupts this NFATc2 stabilization pathway through two mechanisms, namely GSK-3β inhibition and induction of HDM2 activity. Upon nuclear accumulation, HDM2 targets unphosphorylated NFATc2 for ubiquitination at acceptor lysine residues Lys-684/Lys-897 and hence labels the factor for subsequent proteasomal degradation. Conversely, mutagenesis-induced constitutive serine phosphorylation (Ser-215, Ser-219, and Ser-223) of the SP2 domain prevents NFATc2 from HDM2-mediated ubiquitination and degradation and consequently rescues cancer cells from growth suppression by zoledronic acid. In conclusion, this study demonstrates a critical role of the GSK-3β-HDM2 signaling loop in the regulation of NFATc2 protein stability and growth promotion and suggests that double targeting of this pathway is responsible, at least to a significant part, for the potent and reliable anti-tumoral effects of zoledronic acid.

  • Research Article
  • Cite Count Icon 129
  • 10.2353/ajpath.2007.070535
In Vivo Evidence for the Role of CD44s in Promoting Breast Cancer Metastasis to the Liver
  • Dec 1, 2007
  • The American Journal of Pathology
  • Allal Ouhtit + 4 more

In Vivo Evidence for the Role of CD44s in Promoting Breast Cancer Metastasis to the Liver

  • PDF Download Icon
  • Addendum
  • Cite Count Icon 40
  • 10.1074/jbc.m408708200
Bone Marrow Stromal Cell-derived Growth Inhibitor Inhibits Growth and Migration of Breast Cancer Cells via Induction of Cell Cycle Arrest and Apoptosis
  • Feb 1, 2005
  • Journal of Biological Chemistry
  • Tao Wang + 7 more

Genes encoding growth-inhibitory proteins are postulated to be candidate tumor suppressors. The identification of such proteins may benefit the early diagnosis and therapy of tumors. Here we report the cloning and functional characterization of a novel human bone marrow stromal cell (BMSC)-derived growth inhibitor (BDGI) by large scale random sequencing of a human BMSC cDNA library. Human BDGI cDNA encodes a 477-amino acid residue protein that shares high homology with rat and mouse pregnancy-induced growth inhibitors. The C-terminal of BDGI is identical to a novel human pregnancy-induced growth inhibitor, OKL38. BDGI is also closely related to many other eukaryotic proteins, which together form a novel and highly conserved family of BDGI-like proteins. BDGI overexpression inhibits the proliferation, decreases anchorage-dependent growth, and reduces migration of MCF-7 human breast cancer cells, whereas down-regulation of BDGI expression promotes the proliferation of MCF-7 and HeLa cervix epitheloid carcinoma cells. Interestingly, the inhibitory effect of BDGI on MCF-7 cells is more potent than that of OKL38. We demonstrate that BDGI induces cell cycle arrest in S phase and subsequent apoptosis of MCF-7 cells, which is likely to account for the antiproliferative effects of BDGI. This process may involve up-regulation of p27Kip1 and down-regulation of cyclin A, Bcl-2, and Bcl-xL. The inhibitory effect of BDGI on cell proliferation and the induction of apoptosis were also observed in A549 lung cancer cells but not HeLa cells. These results indicate that BDGI might be a growth inhibitor for human tumor cells, especially breast cancer cells, possibly contributing to the development of new therapeutic strategies for breast cancer.

  • Research Article
  • Cite Count Icon 92
  • 10.1016/j.ajpath.2012.02.020
Spontaneous Formation of Tumorigenic Hybrids between Breast Cancer and Multipotent Stromal Cells Is a Source of Tumor Heterogeneity
  • Apr 27, 2012
  • The American Journal of Pathology
  • Germana Rappa + 2 more

Spontaneous Formation of Tumorigenic Hybrids between Breast Cancer and Multipotent Stromal Cells Is a Source of Tumor Heterogeneity

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 46
  • 10.1074/jbc.c110.114124
EGF-induced Grb7 Recruits and Promotes Ras Activity Essential for the Tumorigenicity of Sk-Br3 Breast Cancer Cells
  • Sep 1, 2010
  • Journal of Biological Chemistry
  • Pei-Yu Chu + 4 more

Co-amplification and co-overexpression of ErbB2 and Grb7 are frequently found in various cancers, including breast cancer. Biochemical and functional correlations of the two molecules have identified Grb7 to be a pivotal mediator downstream of ErbB2-mediated oncogenesis. However, it remains largely unknown how Grb7 is involve in the ErbB2-mediated tumorigenesis. In this study, we show that Grb7-mediated cell proliferation and growth are essential for the tumorigenesis that occurs in ErbB2-Grb7-overexpressing breast cancer cells. Intrinsically, EGF-induced de novo Grb7 tyrosine phosphorylation/activation recruits and activates Ras-GTPases and subsequently promotes the phosphorylation of ERK1/2, thereby stimulating tumor growth. Furthermore, we also found the anti-tumor effect could be synergized by co-treatment with Herceptin plus Grb7 knockdown in Sk-Br3 breast cancer cells. Our findings illustrate an underlying mechanism by which Grb7 promotes tumorigenesis through the formation of a novel EGFR-Grb7-Ras signaling complex, thereby highlighting the potential strategy of targeting Grb7 as an anti-breast cancer therapy.

  • Research Article
  • Cite Count Icon 51
  • 10.1016/j.gene.2016.04.023
High-throughput screening of Sirtuin family of genes in breast cancer
  • Apr 11, 2016
  • Gene
  • Mehri Igci + 7 more

High-throughput screening of Sirtuin family of genes in breast cancer

  • Research Article
  • Cite Count Icon 37
  • 10.1074/jbc.r110.203026
Small Molecule Inhibitors as Probes for Estrogen and Androgen Receptor Action
  • Feb 1, 2011
  • Journal of Biological Chemistry
  • David J Shapiro + 2 more

Because activated estrogen (ER) and androgen (AR) receptors stimulate cell proliferation in breast and prostate cancer, inhibiting their actions represents a major therapeutic goal. Most efforts to modulate ER and AR activity have focused on inhibiting the synthesis of estrogens or androgens or on the identification of small molecules that act by competing with agonist hormones for binding in the ligand-binding pocket of the receptor. An alternative approach is to implement screens for small molecule inhibitors that target other sites in the pathway of steroid receptor action. Many of these second-site inhibitors directly target ER or AR; others have still unknown sites of action. Small molecule inhibitors that target second sites represent new leads with clinical potential; they serve as novel modulators of receptor action; and they can reveal new and as yet unidentified interactions and pathways that modulate ER and AR action.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 76
  • 10.1074/jbc.m109.028407
RGS16 Inhibits Breast Cancer Cell Growth by Mitigating Phosphatidylinositol 3-Kinase Signaling
  • Aug 1, 2009
  • Journal of Biological Chemistry
  • Genqing Liang + 3 more

Aberrant activity of the phosphatidylinositol 3-kinase (PI3K) pathway supports growth of many tumors including those of breast, lung, and prostate. Resistance of breast cancer cells to targeted chemotherapies including tyrosine kinase inhibitors (TKI) has been linked to persistent PI3K activity, which may in part be due to increased membrane expression of epidermal growth factor (EGF) receptors (HER2 and HER3). Recently we found that proteins of the RGS (regulator of G protein signaling) family suppress PI3K activity downstream of the receptor by sequestering its p85alpha subunit from signaling complexes. Because a substantial percentage of breast tumors have RGS16 mutations and reduced RGS16 protein expression, we investigated the link between regulation of PI3K activity by RGS16 and breast cancer cell growth. RGS16 overexpression in MCF7 breast cancer cells inhibited EGF-induced proliferation and Akt phosphorylation, whereas shRNA-mediated extinction of RGS16 augmented cell growth and resistance to TKI treatment. Exposure to TKI also reduced RGS16 expression in MCF7 and BT474 cell lines. RGS16 bound the amino-terminal SH2 and inter-SH2 domains of p85alpha and inhibited its interaction with the EGF receptor-associated adapter protein Gab1. These results suggest that the loss of RGS16 in some breast tumors enhances PI3K signaling elicited by growth factors and thereby promotes proliferation and TKI evasion downstream of HER activation.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 68
  • 10.1186/bcr2900
NOTCH-1 and NOTCH-4 are novel gene targets of PEA3 in breast cancer: novel therapeutic implications
  • Jun 1, 2011
  • Breast Cancer Research
  • Anthony G Clementz + 4 more

IntroductionWomen with triple-negative breast cancer have the worst prognosis, frequently present with metastatic tumors and have few targeted therapy options. Notch-1 and Notch-4 are potent breast oncogenes that are overexpressed in triple-negative and other subtypes of breast cancer. PEA3, an ETS transcription factor, is also overexpressed in triple-negative and other breast cancer subtypes. We investigated whether PEA3 could be the critical transcriptional activator of Notch receptors in MDA-MB-231 and other breast cancer cells.MethodsReal-time PCR and Western blot analysis were performed to detect Notch-1, Notch-2, Notch-3 and Notch-4 receptor expression in breast cancer cells when PEA3 was knocked down by siRNA. Chromatin immunoprecipitation was performed to identify promoter regions for Notch genes that recruited PEA3. TAM-67 and c-Jun siRNA were used to identify that c-Jun was necessary for PEA3 enrichment on the Notch-4 promoter. A Notch-4 luciferase reporter was used to confirm that endogenous PEA3 or AP-1 activated the Notch-4 promoter region. Cell cycle analysis, trypan blue exclusion, annexin V flow cytometry, colony formation assay and an in vivo xenograft study were performed to determine the biological significance of targeting PEA3 via siRNA, Notch signaling via a γ-secretase inhibitor, or both.ResultsHerein we provide new evidence for transcriptional regulation of Notch by PEA3 in breast cancer. PEA3 activates Notch-1 transcription in MCF-7, MDA-MB-231 and SKBr3 breast cancer cells. PEA3 activates Notch-4 transcription in MDA-MB-231 cells where PEA3 levels are endogenously high. In SKBr3 and BT474 breast cancer cells where PEA3 levels are low, overexpression of PEA3 increases Notch-4 transcripts. Chromatin immunoprecipitation confirmed the enrichment of PEA3 on Notch-1 and Notch-4 promoters in MDA-MB-231 cells. PEA3 recruitment to Notch-1 was AP-1-independent, whereas PEA3 recruitment to Notch-4 was c-JUN-dependent. Importantly, the combined inhibition of Notch signaling via a γ-secretase inhibitor (MRK-003 GSI) and knockdown of PEA3 arrested growth in the G1 phase, decreased both anchorage-dependent and anchorage-independent growth and significantly increased apoptotic cells in vitro. Moreover, either PEA3 knockdown or MRK-003 GSI treatment significantly reduced tumor growth of MDA-MB-231 xenografts in vivo.ConclusionsTaken together, the results from this study demonstrate for the first time that Notch-1 and Notch-4 are novel transcriptional targets of PEA3 in breast cancer cells. Targeting of PEA3 and/or Notch pathways might provide a new therapeutic strategy for triple-negative and possibly other breast cancer subtypes.

  • Research Article
  • Cite Count Icon 14
  • 10.1038/s41598-018-22364-z
B49, a BST-2-based peptide, inhibits adhesion and growth of breast cancer cells
  • Mar 9, 2018
  • Scientific Reports
  • Wadie D Mahauad-Fernandez + 1 more

Bone marrow stromal antigen 2 (BST-2) also known as Tetherin has been implicated in the growth and progression of many cancers. BST-2 employs its pro-tumor effects through the formation of BST-2:BST-2 dimers which ultimately promotes cell to cell and cell to matrix adhesion, cell motility, survival, and growth. The aim of this study was to evaluate the effect of a novel BST-2-based peptide—B49 on adhesion and growth of breast cancer cells. Homotypic/heterotypic adhesion, three-dimensional spheroid formation, and anchorage-independent growth were used to assess the effect of B49 on cell adhesion and growth. Additionally, we provide evidence of the anti-tumor effect of B49 in a preclinical mouse model of breast cancer. Results show that breast cancer cell adhesion to other cancer cells or components of the tumor microenvironment were inhibited by B49. Most well-known evaluation indexes of cancer cell growth, including spheroid formation, anchorage-independent, and primary tumor growth were significantly inhibited by B49. These data affirm that i) BST-2 plays a key role in mediating breast cancer cell adhesion and growth, and ii) B49 and its analog B49Mod1 significantly inhibits BST-2-mediated cancer cell adhesion and growth. Therefore, B49 and its analogs offer a promising anti-adhesion and therapeutic lead for BST-2-dependent cancers.

  • Research Article
  • Cite Count Icon 23
  • 10.3390/cancers13112682
NR4A1 Ligands as Potent Inhibitors of Breast Cancer Cell and Tumor Growth
  • May 29, 2021
  • Cancers
  • Keshav Karki + 5 more

Simple SummaryBis-indole derived (CDIMs) bind the orphan nuclear receptor 4A1 (NR4A1) and inhibit NR4A1-regulated cancer cell and tumor growth. In this study a subset of 3,5-disubstituted phenyl CDIM compounds that bound NR4A1 were investigated in a breast cancer model. All of these analogs were potent inhibitors of breast tumor growth in a xenograft model using MDA-MB-231 cells at doses ≤ 1 mg/kg/d.Nuclear receptor 4A1 (NR4A1, Nur77, TR3) is more highly expressed in breast and solid tumors compared to non-tumor tissues and is a pro-oncogenic factor in solid tumor-derived cancers. NR4A1 regulates cancer cell growth, survival, migration, and invasion, and bis-indole-derived compounds (CDIMs) that bind NR4A1 act as antagonists and inhibit tumor growth. Preliminary structure-binding studies identified 1,1-bis(3′-indolyl)-1-(3,5-disubstitutedphenyl)methane analogs as NR4A1 ligands with low KD values; we further investigated the anticancer activity of the four most active analogs (KD’s ≤ 3.1 µM) in breast cancer cells and in athymic mouse xenograft models. The treatment of MDA-MB-231 and SKBR3 breast cancer cells with the 3-bromo-5-methoxy, 3-chloro-5-trifluoromethoxy, 3-chloro-5-trifluoromethyl, and 3-bromo-5-trifluoromethoxy phenyl-substituted analogs decreased cell growth and the expression of epidermal of growth factor receptor (EGFR), hepatocyte growth factor receptor (cMET), and PD-L1 as well as inhibited mTOR phosphorylation. In addition, all four compounds inhibited tumor growth in athymic nude mice bearing MDA-MB-231 cells (orthotopic) at a dose of 1 mg/kg/d, which was not accompanied by changes in body weight. These 3,5-disubstituted analogs were the most potent CDIM/NR4A1 ligands reported and are being further developed for clinical applications.

  • Research Article
  • Cite Count Icon 183
  • 10.1074/jbc.m607094200
Autophagy for Cancer Therapy through Inhibition of Pro-apoptotic Proteins and Mammalian Target of Rapamycin Signaling
  • Dec 1, 2006
  • Journal of Biological Chemistry
  • Kwang Woon Kim + 6 more

Autophagy is an alternative cell death pathway that is induced by mammalian target of rapamycin (mTOR) inhibitors and up-regulated when apoptosis is defective. We investigated radiation-induced autophagy in the presence or absence of Bax/Bak with or without an mTOR inhibitor, Rad001. Two isogenic cell lines, wild type (WT) and Bak/Bak(-/-) mouse embryonic fibroblasts and tumor cell lines were used for this study. Irradiated Bak/Bak(-/-) cells had a decrease of Akt/mTOR signaling and a significant increase of pro-autophagic proteins ATG5-ATG12 COMPLEX and Beclin-1. These molecular events resulted in an up-regulation of autophagy. Bax/Bak(-/-) cells were defective in undergoing apoptosis but were more radiosensitive than the WT cells in autophagy. Both autophagy and sensitization of Bak/Bax(-/-) cells were further enhanced in the presence of Rad001. In contrast, inhibitors of autophagy rendered the Bak/Bax(-/-) cells radioresistant, whereas overexpression of ATG5 and Beclin-1 made the WT cells radiosensitive. When this novel concept of radiosensitization was tested in cancer models, small interfering RNAs against Bak/Bax also led to increased autophagy and sensitization of human breast and lung cancer cells to gamma radiation, which was further enhanced by Rad001. This is the first report to demonstrate that inhibition of pro-apoptotic proteins and induction of autophagy sensitizes cancer cells to therapy. Therapeutically targeting this novel pathway may yield significant benefits for cancer patients.

More from: European journal of medicinal chemistry
  • Research Article
  • 10.1016/j.ejmech.2025.118046
Recent advances of the mechanistic target of rapamycin (mTOR) inhibitors for drug discovery.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Sha Ni + 5 more

  • Research Article
  • 10.1016/j.ejmech.2025.117913
Highly potent dipeptidyl peptidase 8/9 (DPP8/9) inhibitors designed via relative binding free energy calculations.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Vanesa Nozal + 7 more

  • Research Article
  • 10.1016/j.ejmech.2025.117920
Local enhancement of cationic charge density via polyamine side chain incorporation improves the selectivity of antimicrobial peptoids.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Jinyoung Oh + 6 more

  • Research Article
  • 10.1016/j.ejmech.2025.118040
Discovery, structural modification and structure-activity relationship study of Echinulin derivatives as tyrosyl-DNA phosphodiesterase 1 inhibitors and their potential antitumor activity.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Chuan-Sheng Yao + 10 more

  • Research Article
  • 10.1016/j.ejmech.2025.117923
Design and synthesis of Riluzole-Ciprofloxacin hybrids as selective MST3 inhibitors for cancer treatment.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Deping Li + 5 more

  • Research Article
  • 10.1016/j.ejmech.2025.118000
trans-Styrylquinazolinone CYP1B1 inhibitors as potential therapeutics in A549 cells.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Jiajing Cai + 3 more

  • Research Article
  • 10.1016/j.ejmech.2025.117955
Analogues of gramicidin S: A promising direction for future antibacterial drug development?
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Tatiana S Shkuratova + 3 more

  • Research Article
  • 10.1016/j.ejmech.2025.118340
Drug repurposing identifies novel Wee1 kinase inhibitors for triple negative breast cancer therapeutics
  • Nov 1, 2025
  • European Journal of Medicinal Chemistry
  • Jisha Pillai U + 6 more

  • Research Article
  • 10.1016/j.ejmech.2025.117993
Design, synthesis, and pharmacological evaluation of novel M4 muscarinic receptor positive allosteric modulators for schizophrenia treatment.
  • Nov 1, 2025
  • European journal of medicinal chemistry
  • Yi Luo + 11 more

  • Research Article
  • 10.1016/j.ejmech.2025.118336
Synthesis of Novel β-Boswellic Acid Amino Acid Conjugates in Targeting KRASG13D Mutant Colon Cancer
  • Nov 1, 2025
  • European Journal of Medicinal Chemistry
  • Arem Qayum + 11 more

Save Icon
Up Arrow
Open/Close
  • Ask R Discovery Star icon
  • Chat PDF Star icon

AI summaries and top papers from 250M+ research sources.

Search IconWhat is the difference between bacteria and viruses?
Open In New Tab Icon
Search IconWhat is the function of the immune system?
Open In New Tab Icon
Search IconCan diabetes be passed down from one generation to the next?
Open In New Tab Icon