21 results on '"Thomas W. Gero"'
Search Results
2. Table S2 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
KINOMEscan selectivity profiling of JBJ-04-125-02 and IC50 of EAI045, JBJ-02-112-05 and JBJ-04-125-02 in BaF3 cells.
- Published
- 2023
- Full Text
- View/download PDF
3. Figure S1 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
JBJ-04-125-02 is more potent than EAI045 and JBJ-02-112-05 in the presence of cetuximab and is effective in inhibiting EGFR-dependent proliferation and phosphorylation in L858R/ C797S Ba/F3 cells.
- Published
- 2023
- Full Text
- View/download PDF
4. Figure S5 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
The combination treatment of JBJ-04-125-02 and osimertinib do not result in any toxicity issues associated with weight loss but osimertinib at clinically relevant dose (25 mg/kg) is too potent to exhibit additive effect when combined with JBJ-04-125-02.
- Published
- 2023
- Full Text
- View/download PDF
5. Figure S2 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
JBJ-04-125-02 as a single agent does not result in any toxicity issues associated with weight loss.
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
Allosteric kinase inhibitors offer a potentially complementary therapeutic strategy to ATP-competitive kinase inhibitors due to their distinct sites of target binding. In this study, we identify and study a mutant-selective EGFR allosteric inhibitor, JBJ-04-125-02, which as a single agent can inhibit cell proliferation and EGFRL858R/T790M/C797S signaling in vitro and in vivo. However, increased EGFR dimer formation limits treatment efficacy and leads to drug resistance. Remarkably, osimertinib, an ATP-competitive covalent EGFR inhibitor, uniquely and significantly enhances the binding of JBJ-04-125-02 for mutant EGFR. The combination of osimertinib and JBJ-04-125-02 results in an increase in apoptosis, a more effective inhibition of cellular growth, and an increased efficacy in vitro and in vivo compared with either single agent alone. Collectively, our findings suggest that the combination of a covalent mutant–selective ATP-competitive inhibitor and an allosteric EGFR inhibitor may be an effective therapeutic approach for patients with EGFR-mutant lung cancer.Significance:The clinical efficacy of EGFR tyrosine kinase inhibitors (TKI) in EGFR-mutant lung cancer is limited by acquired drug resistance, thus highlighting the need for alternative strategies to inhibit EGFR. Here, we identify a mutant EGFR allosteric inhibitor that is effective as a single agent and in combination with the EGFR TKI osimertinib.This article is highlighted in the In This Issue feature, p. 813
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Methods from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
Supplemental methods for structural modeling of inhibitor binding, pharmacokinetic assays, in vivo studies, crosslinking and biotinylated drug pull-down assays.
- Published
- 2023
- Full Text
- View/download PDF
8. Table S1 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
Crystallographic data collection and refinement statistics.
- Published
- 2023
- Full Text
- View/download PDF
9. Figure S4 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
Not all tyrosine kinase inhibitors (TKIs) can enhance or bind at the same time as an allosteric inhibitor (AI) and co-binding of TKIs and AIs do not necessarily translate into increased drug potency.
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S3 from Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Pasi A. Jänne, Nathanael S. Gray, Michael J. Eck, Kwok-Kin Wong, Suzanne E. Dahlberg, Annan Yang, Thomas W. Gero, Bo Hee Shin, David E. Heppner, Michael D. Cameron, Stephen Wang, Man Xu, Dries J.H. De Clercq, Mierzhati Mushajiang, Eunyoung Park, Ting Chen, Jaebong Jang, and Ciric To
- Abstract
The relative allelic fraction of T790M does not change the potency of JBJ-04-125-02 in H3255 cells but the presence of EGF blunts the efficacy of JBJ-04-125-02 in Ba/F3 and H1975 cells.
- Published
- 2023
- Full Text
- View/download PDF
11. Discovery of sterically-hindered phenol compounds with potent cytoprotective activities against ox-LDL–induced retinal pigment epithelial cell death as a potential pharmacotherapy
- Author
-
Franco Aparecido Rossato, Eun Young Choi, Andrew J. Urquhart, David A. Scott, Yin Shan E. Ng, Gopalan Gnanaguru, Ashley Mackey, Anthoula Arta, Patricia A. D'Amore, and Thomas W. Gero
- Subjects
Programmed cell death ,Retinal Pigment Epithelium ,Pharmacology ,Biochemistry ,Article ,chemistry.chemical_compound ,Phenols ,Physiology (medical) ,medicine ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,Gene knockdown ,Retinal pigment epithelium ,Chemistry ,Troglitazone ,Epithelial Cells ,Cytoprotection ,eye diseases ,Lipoproteins, LDL ,medicine.anatomical_structure ,sense organs ,Trolox ,Retinal Pigments ,Lipoprotein ,medicine.drug - Abstract
Late-stage dry age-related macular degeneration (AMD) or geographic atrophy (GA) is an irreversible blinding condition characterized by degeneration of retinal pigment epithelium (RPE) and the associated photoreceptors. Clinical and genetic evidence supports a role for dysfunctional lipid processing and accumulation of harmful oxidized lipids in the pathogenesis of GA. Using an oxidized low-density lipoprotein (ox-LDL)-induced RPE death assay, we screened and identified sterically-hindered phenol compounds with potent protective activities for RPE. The phenol-containing PPARγ agonist, troglitazone, protected against ox-LDL–induced RPE cell death, whereas other more potent PPARγ agonists did not protect RPE cells. Knockdown of PPARγ did not affect the protective activity of troglitazone in RPE, confirming the protective function is not due to the thiazolidine (TZD) group of troglitazone. Prototypical hindered phenol trolox and its analogs potently protected against ox-LDL–induced RPE cell death whereas potent antioxidants without the phenol group failed to protect RPE. Hindered phenols preserved lysosomal integrity against ox-LDL–induced damage and FITC-labeled trolox was localized to the lysosomes in RPE cells. Analogs of trolox inhibited reactive oxygen species (ROS) formation induced by ox-LDL uptake in a dose-dependent fashion and were effective at sub-micromolar concentrations. Treatment with trolox analog 2,2,5,7,8-pentamethyl-6-chromanol (PMC) significantly induced the expression of the lysosomal protein NPC-1 and reduced intracellular cholesterol level upon ox-LDL uptake. Our data indicate that the lysosomal-localized hindered phenols are uniquely potent in protecting the RPE against the toxic effects of ox-LDL, and may represent a novel pharmacotherapy to preserve the vision in patients with GA.
- Published
- 2022
- Full Text
- View/download PDF
12. Quinazolinones as allosteric fourth-generation EGFR inhibitors for the treatment of NSCLC
- Author
-
Thomas W. Gero, David E. Heppner, Tyler S. Beyett, Ciric To, Seth C. Azevedo, Jaebong Jang, Thomas Bunnell, Frederic Feru, Zhengnian Li, Bo Hee Shin, Kara M. Soroko, Prafulla C. Gokhale, Nathanael S. Gray, Pasi A. Jänne, Michael J. Eck, and David A. Scott
- Subjects
Lung Neoplasms ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,ErbB Receptors ,Phenols ,Drug Resistance, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Drug Discovery ,Mutation ,Molecular Medicine ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,Quinazolinones - Abstract
The C797S mutation confers resistance to covalent EGFR inhibitors used in the treatment of lung tumors with the activating L858R mutation. Isoindolinones such as JBJ-4-125-02 bind in an allosteric pocket and are active against this mutation, with high selectivity over wild-type EGFR. The most potent examples we developed from that series have a potential chemical instability risk from the combination of the amide and phenol groups. We explored a scaffold hopping approach to identify new series of allosteric EGFR inhibitors that retained good potency in the absence of the phenol group. The 5-F quinazolinone 34 demonstrated tumor regression in an H1975 efficacy model upon once daily oral dosing at 25 mg/kg.
- Published
- 2021
13. An allosteric inhibitor against the therapy-resistant mutant forms of EGFR in non-small cell lung cancer
- Author
-
Ciric To, Tyler S. Beyett, Jaebong Jang, William W. Feng, Magda Bahcall, Heidi M. Haikala, Bo H. Shin, David E. Heppner, Jaimin K. Rana, Brittaney A. Leeper, Kara M. Soroko, Michael J. Poitras, Prafulla C. Gokhale, Yoshihisa Kobayashi, Kamal Wahid, Kari J. Kurppa, Thomas W. Gero, Michael D. Cameron, Atsuko Ogino, Mierzhati Mushajiang, Chunxiao Xu, Yanxi Zhang, David A. Scott, Michael J. Eck, Nathanael S. Gray, and Pasi A. Jänne
- Subjects
ErbB Receptors ,Cancer Research ,Adenosine Triphosphate ,Lung Neoplasms ,Oncology ,Carcinoma, Non-Small-Cell Lung ,Mutation ,Humans ,Protein Kinase Inhibitors - Abstract
Epidermal growth factor receptor (EGFR) therapy using small-molecule tyrosine kinase inhibitors (TKIs) is initially efficacious in patients with EGFR-mutant lung cancer, although drug resistance eventually develops. Allosteric EGFR inhibitors, which bind to a different EGFR site than existing ATP-competitive EGFR TKIs, have been developed as a strategy to overcome therapy-resistant EGFR mutations. Here we identify and characterize JBJ-09-063, a mutant-selective allosteric EGFR inhibitor that is effective across EGFR TKI-sensitive and resistant models, including those with EGFR T790M and C797S mutations. We further uncover that EGFR homo- or heterodimerization with other ERBB family members, as well as the EGFR L747S mutation, confers resistance to JBJ-09-063, but not to ATP-competitive EGFR TKIs. Overall, our studies highlight the potential clinical utility of JBJ-09-063 as a single agent or in combination with EGFR TKIs to define more effective strategies to treat EGFR-mutant lung cancer.
- Published
- 2021
14. Discovery of a series of benzopyrimidodiazepinone TNK2 inhibitors via scaffold morphing
- Author
-
Thomas W. Gero, John Thomas Feutrill, Bailing Chen, Nathanael S. Gray, Feru Frederic, Brian J. Groendyke, Hilary Szabo, Zhengnian Li, Bin Li, David A. Scott, and Chelsea E. Powell
- Subjects
Male ,Scaffold ,Clinical Biochemistry ,Pharmaceutical Science ,Crystallography, X-Ray ,01 natural sciences ,Biochemistry ,Compound 32 ,TNK2 ,Cell Line ,Residue (chemistry) ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Drug Stability ,Drug Discovery ,Animals ,Humans ,Protein kinase A ,Molecular Biology ,Protein Kinase Inhibitors ,Alanine ,Benzodiazepinones ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Effector ,Organic Chemistry ,Tyrosine phosphorylation ,Protein-Tyrosine Kinases ,0104 chemical sciences ,Cell biology ,010404 medicinal & biomolecular chemistry ,Pyrimidines ,Microsomes, Liver ,Molecular Medicine ,Protein Binding - Abstract
The protein kinase TNK2 (ACK1) is an emerging drug target for a variety of indications, in particular for cancer where it plays a key role transmitting cell survival, growth and proliferative signals via modification of multiple downstream effectors by unique tyrosine phosphorylation events. Scaffold morphing based on our previous TNK2 inhibitor XMD8-87 identified urea 17 from which we developed the potent and selective compound 32. A co-crystal structure was obtained showing 32 interacting primarily with the main chain atoms of an alanine residue of the hinge region. Additional H-bonds exist between the urea NHs and the Thr205 and Asp270 residues.
- Published
- 2020
15. Benzopyrimidodiazepinone inhibitors of TNK2
- Author
-
Brian J. Groendyke, John Thomas Feutrill, Thomas W. Gero, Zhengnian Li, David A. Scott, Feru Frederic, Chelsea E. Powell, Hilary Szabo, Kevin Pang, Bailing Chen, Bin Li, and Nathanael S. Gray
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,Inhibitory Concentration 50 ,Mice ,Structure-Activity Relationship ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,Diimine ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Azepines ,Protein-Tyrosine Kinases ,Combinatorial chemistry ,0104 chemical sciences ,Quinone ,010404 medicinal & biomolecular chemistry ,Microsomes, Liver ,Molecular Medicine ,Half-Life ,Protein Binding - Abstract
The SAR of a series of benzopyrimidodiazepinone inhibitors of TNK2 was developed, starting from the potent and selective compound XMD8-87. A diverse set of anilines was introduced in an effort to improve the in vivo PK profile and minimize the risk of quinone diimine formation.
- Published
- 2019
16. Discovery and Optimization of Dibenzodiazepinones as Allosteric Mutant-Selective EGFR Inhibitors
- Author
-
David E. Heppner, Cai-Hong Yun, Nathanael S. Gray, Ciric To, Michael J. Eck, Dries De Clercq, Bo Hee Shin, Pasi A. Jänne, Mierzhati Mushajiang, David A. Scott, Jaebong Jang, Thomas W. Gero, and Eun Young Park
- Subjects
010405 organic chemistry ,Kinase ,Chemistry ,Organic Chemistry ,Mutant ,Allosteric regulation ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,respiratory tract diseases ,010404 medicinal & biomolecular chemistry ,T790M ,Egfr mutation ,Drug Discovery ,Cancer research ,Kinome ,EGFR inhibitors ,Therapeutic strategy - Abstract
[Image: see text] Allosteric kinase inhibitors represent a promising new therapeutic strategy for targeting kinases harboring oncogenic driver mutations in cancers. Here, we report the discovery, optimization, and structural characterization of allosteric mutant-selective EGFR inhibitors comprising a 5,10-dihydro-11H-dibenzo[b,e][1,4]diazepin-11-one scaffold. Our structure-based medicinal chemistry effort yielded an inhibitor (3) of the EGFR(L858R/T790M) and EGFR(L858R/T790M/C797S) mutants with an IC(50) of ∼10 nM and high selectivity, as assessed by kinome profiling. Further efforts to develop allosteric dibenzodiazepinone inhibitors may serve as the basis for new therapeutic options for targeting drug-resistant EGFR mutations.
- Published
- 2019
17. Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
- Author
-
Bo Hee Shin, Michael D. Cameron, Thomas W. Gero, Michael J. Eck, Man Xu, Kwok-Kin Wong, Stephen Wang, Annan Yang, Ciric To, Jaebong Jang, Mierzhati Mushajiang, Suzanne E. Dahlberg, Eun Young Park, Ting Chen, David E. Heppner, Pasi A. Jänne, Nathanael S. Gray, and Dries De Clercq
- Subjects
0301 basic medicine ,Lung Neoplasms ,Cell Survival ,Allosteric regulation ,Mutant ,Benzeneacetamides ,Mice, Transgenic ,Article ,03 medical and health sciences ,T790M ,Mice ,0302 clinical medicine ,Allosteric Regulation ,In vivo ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Humans ,Osimertinib ,Protein Kinase Inhibitors ,EGFR inhibitors ,Acrylamides ,Aniline Compounds ,Kinase ,Chemistry ,Cell growth ,Xenograft Model Antitumor Assays ,ErbB Receptors ,Thiazoles ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,NIH 3T3 Cells - Abstract
Allosteric kinase inhibitors offer a potentially complementary therapeutic strategy to ATP-competitive kinase inhibitors due to their distinct sites of target binding. In this study, we identify and study a mutant-selective EGFR allosteric inhibitor, JBJ-04-125-02, which as a single agent can inhibit cell proliferation and EGFRL858R/T790M/C797S signaling in vitro and in vivo. However, increased EGFR dimer formation limits treatment efficacy and leads to drug resistance. Remarkably, osimertinib, an ATP-competitive covalent EGFR inhibitor, uniquely and significantly enhances the binding of JBJ-04-125-02 for mutant EGFR. The combination of osimertinib and JBJ-04-125-02 results in an increase in apoptosis, a more effective inhibition of cellular growth, and an increased efficacy in vitro and in vivo compared with either single agent alone. Collectively, our findings suggest that the combination of a covalent mutant–selective ATP-competitive inhibitor and an allosteric EGFR inhibitor may be an effective therapeutic approach for patients with EGFR-mutant lung cancer. Significance: The clinical efficacy of EGFR tyrosine kinase inhibitors (TKI) in EGFR-mutant lung cancer is limited by acquired drug resistance, thus highlighting the need for alternative strategies to inhibit EGFR. Here, we identify a mutant EGFR allosteric inhibitor that is effective as a single agent and in combination with the EGFR TKI osimertinib. This article is highlighted in the In This Issue feature, p. 813
- Published
- 2018
18. Potent and Selective Covalent Quinazoline Inhibitors of KRAS G12C
- Author
-
Nathanael S. Gray, Chiara Ambrogio, Thomas W. Gero, Scott B. Ficarro, Feru Frederic, Yuan Xiong, Raymond M. Paranal, Kwok-Kin Wong, Chunshan Quan, Marco Catalano, Jarrod A. Marto, David A. Scott, Jay Shao, Kenneth D. Westover, Jia Lu, Eric S. Fischer, Pasi A. Jänne, Lianbo Li, and Mei Zeng
- Subjects
0301 basic medicine ,GTP' ,Stereochemistry ,MAP Kinase Signaling System ,Clinical Biochemistry ,Allosteric regulation ,Apoptosis ,Molecular Dynamics Simulation ,medicine.disease_cause ,Crystallography, X-Ray ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,medicine ,Quinazoline ,Structure–activity relationship ,Humans ,Phosphorylation ,Molecular Biology ,Pharmacology ,Acrylamides ,Binding Sites ,Chemistry ,Drug discovery ,HCT116 Cells ,Small molecule ,Amides ,Protein Structure, Tertiary ,030104 developmental biology ,Covalent bond ,A549 Cells ,Mutagenesis, Site-Directed ,Quinazolines ,ras Proteins ,Molecular Medicine ,KRAS - Abstract
Targeted covalent small molecules have shown promise for cancers driven by KRAS G12C. Allosteric compounds that access an inducible pocket formed by movement of a dynamic structural element in KRAS, switch II, have been reported, but these compounds require further optimization to enable their advancement into clinical development. We demonstrate that covalent quinazoline-based switch II pocket (SIIP) compounds effectively suppress GTP loading of KRAS G12C, MAPK phosphorylation, and the growth of cancer cells harboring G12C. Notably we find that adding an amide substituent to the quinazoline scaffold allows additional interactions with KRAS G12C, and remarkably increases the labeling efficiency, potency, and selectivity of KRAS G12C inhibitors. Structural studies using X-ray crystallography reveal a new conformation of SIIP and key interactions made by substituents located at the quinazoline 2-, 4-, and 7-positions. Optimized lead compounds in the quinazoline series selectively inhibit KRAS G12C-dependent signaling and cancer cell growth at sub-micromolar concentrations.
- Published
- 2017
19. Exploring Targeted Degradation Strategy for Oncogenic KRASG12C
- Author
-
Feru Frederic, Katherine A. Donovan, Nathanael S. Gray, Mei Zeng, Nozhat Safaee, Milka Kostic, Eric S. Fischer, Pasi A. Jänne, Sudershan R. Gondi, Kenneth D. Westover, Behnam Nabet, Christine Yuan, Lincoln J. Ombelets, Yuan Xiong, David A. Scott, Chunshan Quan, Thomas W. Gero, Lianbo Li, and Radosław P. Nowak
- Subjects
Pharmacology ,biology ,Oncogene ,010405 organic chemistry ,Clinical Biochemistry ,Endogeny ,medicine.disease ,medicine.disease_cause ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,Ubiquitin ,Drug Discovery ,medicine ,biology.protein ,Cancer research ,Molecular Medicine ,KRAS ,Lung cancer ,Molecular Biology ,Cysteine - Abstract
Summary KRAS is the most frequently mutated oncogene found in pancreatic, colorectal, and lung cancers. Although it has been challenging to identify targeted therapies for cancers harboring KRAS mutations, KRASG12C can be targeted by small-molecule inhibitors that form covalent bonds with cysteine 12 (C12). Here, we designed a library of C12-directed covalent degrader molecules (PROTACs) and subjected them to a rigorous evaluation process to rapidly identify a lead compound. Our lead degrader successfully engaged CRBN in cells, bound KRASG12C in vitro, induced CRBN/KRASG12C dimerization, and degraded GFP-KRASG12C in reporter cells in a CRBN-dependent manner. However, it failed to degrade endogenous KRASG12C in pancreatic and lung cancer cells. Our data suggest that inability of the lead degrader to effectively poly-ubiquitinate endogenous KRASG12C underlies the lack of activity. We discuss challenges for achieving targeted KRASG12C degradation and proposed several possible solutions which may lead to efficient degradation of endogenous KRASG12C.
- Published
- 2020
- Full Text
- View/download PDF
20. Structure-activity studies of analogs of ?, ?-methylene-ATP at P2X-purinoceptors in the rabbit ear central artery
- Author
-
Noel J. Cusack, Pauline L. Martin, Alice Anne Potts, and Thomas W. Gero
- Subjects
Agonist ,Purine ,chemistry.chemical_classification ,medicine.drug_class ,Stereochemistry ,Antagonist ,Partial agonist ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,Ribose ,Prazosin ,medicine ,Potency ,Nucleotide ,medicine.drug - Abstract
Structure-activity studies using naphthylmethyl analogs of β,γ-methylene-ATP were conducted at the P 2x -purinoceptor that mediates contraction of the rabbit ear central artery by ATP, α,β-m-ATP, and β,γ-m-ATP. On the adenine base, substitution at the C 2 -position (WRC-0440) increased the agonist potency 2-fold and substitution at the C 8 -position (WRC-0431) did not change agonist potency, and both compounds had the same maximal response as β,γ-m-ATP, whereas substitution at the N 6 -position (WRC-0416) abolished activity. On the D-ribose sugar, substitution on the 2'-hydroxyl generated a partial agonist (WRC-0479), which had a maximal effect of only 39% of that of β-γ-m-ATP. Attempts to substitute the 3'-hydroxyl by naphthylmethyl failed, but substitution by p-methoxybenzyl (WRC-0617) did not change potency or the maximal response. Cyclic substitution of both the 2'- and 3'-hydroxyls by naphthylmethylidine (WRC-0498) had no effect on the agonist potency or the maximal response relative to β-γ-m-ATP. On the β,γ-methylenetriphosphonate chain, substitution on the methylene linkage by naphthylmethyl (WRC-0433) had no effect on agonist potency, but the maximal response increased to 122% that of β,γ-m-ATP. However, the contractile response to WRC-0433 was not desensitized by α,β-m-ATP (contractile responses to all other agonists were abolished by α-β-m-ATP pretreatment), but was blocked by the α 1 antagonist prazosin (10 -6 M). WRC-0433 appears to act at a prejunctional site that mediates ATP-induced release of norepinephrine. Purine nucleotides with substituents at the 2'-position of the ribose sugar could provide a lead to the generation of P 2x -purinoceptor antagonists.
- Published
- 1995
- Full Text
- View/download PDF
21. Synthesis and biological activity of a cyclic pseudohexapeptide analog of somatostatin
- Author
-
Andrew V. Schally, Ignacio Torres-Aleman, Thomas W. Gero, and Arno F. Spatola
- Subjects
Chemical Phenomena ,Hydrogen bond ,Stereochemistry ,Protein Conformation ,Biophysics ,Biological activity ,Cell Biology ,Growth hormone ,Ring (chemistry) ,Biochemistry ,Peptide Fragments ,Rats ,chemistry.chemical_compound ,Chemistry ,Structure-Activity Relationship ,Somatostatin ,chemistry ,Intramolecular force ,Growth Hormone ,Diphenylphosphoryl azide ,Animals ,Molecular Biology - Abstract
A cyclic pseudohexapeptide analog of somatostatin, cyclo(Pro psi[ CH2S ]Phe-D-Trp-Lys-Thr-Phe) was synthesized by solid phase methods and diphenylphosphoryl azide ring closure. The resulting crystalline compound possessed 23% of the growth hormone inhibitory activity of the parent tetradecapeptide and approximately 6% of the activity of the all-amide cyclic hexapeptide analog in spite of the absence of one of two postulated intramolecular hydrogen bonds.
- Published
- 1984
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.