Covalent Proximity Scanning of a Distal Cysteine to Target PI3KαClick to copy article linkArticle link copied!
- Chiara BorsariChiara BorsariDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Chiara Borsari
- Erhan KelesErhan KelesDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Erhan Keles
- Jacob A. McPhailJacob A. McPhailDepartment of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, CanadaMore by Jacob A. McPhail
- Alexander SchaeferAlexander SchaeferDepartment of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, SwitzerlandMore by Alexander Schaefer
- Rohitha SriramaratnamRohitha SriramaratnamDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Rohitha Sriramaratnam
- Wojciech GochWojciech GochDepartment of Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, PolandMore by Wojciech Goch
- Thorsten SchaeferThorsten SchaeferDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Thorsten Schaefer
- Martina De PascaleMartina De PascaleDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Martina De Pascale
- Wojciech BalWojciech BalInstitute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, PolandMore by Wojciech Bal
- Matthias GstaigerMatthias GstaigerDepartment of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, SwitzerlandMore by Matthias Gstaiger
- John E. BurkeJohn E. BurkeDepartment of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, CanadaMore by John E. Burke
- Matthias P. Wymann*Matthias P. Wymann*Email: [email protected]. Tel: +41 61 207 5046. Fax: +41 61 207 3566.Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Matthias P. Wymann
Abstract
Covalent protein kinase inhibitors exploit currently noncatalytic cysteines in the adenosine 5′-triphosphate (ATP)-binding site via electrophiles directly appended to a reversible-inhibitor scaffold. Here, we delineate a path to target solvent-exposed cysteines at a distance >10 Å from an ATP-site-directed core module and produce potent covalent phosphoinositide 3-kinase α (PI3Kα) inhibitors. First, reactive warheads are used to reach out to Cys862 on PI3Kα, and second, enones are replaced with druglike warheads while linkers are optimized. The systematic investigation of intrinsic warhead reactivity (kchem), rate of covalent bond formation and proximity (kinact and reaction space volume Vr), and integration of structure data, kinetic and structural modeling, led to the guided identification of high-quality, covalent chemical probes. A novel stochastic approach provided direct access to the calculation of overall reaction rates as a function of kchem, kinact, Ki, and Vr, which was validated with compounds with varied linker lengths. X-ray crystallography, protein mass spectrometry (MS), and NanoBRET assays confirmed covalent bond formation of the acrylamide warhead and Cys862. In rat liver microsomes, compounds 19 and 22 outperformed the rapidly metabolized CNX-1351, the only known PI3Kα irreversible inhibitor. Washout experiments in cancer cell lines with mutated, constitutively activated PI3Kα showed a long-lasting inhibition of PI3Kα. In SKOV3 cells, compounds 19 and 22 revealed PI3Kβ-dependent signaling, which was sensitive to TGX221. Compounds 19 and 22 thus qualify as specific chemical probes to explore PI3Kα-selective signaling branches. The proposed approach is generally suited to develop covalent tools targeting distal, unexplored Cys residues in biologically active enzymes.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Results and Discussion
Design of Covalent PI3Kα Inhibitors
Tuning of Warhead Reactivity
Scan of Protein Target Site and Assessment of Proximity
Linker Optimization Using Druglike Warheads
compound | kchem × 104 (M–1·s–1) | kinact PI3Kα × 104 (s–1)a | Ki PI3Kα (nM)a | kinact/Ki × 105 (nM–1·s–1)a | IC50 pPKB (S473) SKOV3c |
---|---|---|---|---|---|
16 | 4.10 | 0.58 | 10.8 | 0.54 | 51 |
17 | 2.49 | 0.98 | 10.7 | 0.93 | 47 |
18 | 3.07 | 2.92 | 8.56 | 3.43 | 48 |
19 | 3.70 | 12.0 | 2.93 | 41.4 | 82 |
20 | 4.52 | 2.52 | 14.0 | 1.80 | 664 |
21 | 4.37 | 1.95 | 10.6 | 1.87 | 686 |
22 | 3.54 | 10.8 | 5.14 | 21.0 | 86 |
23 | 3.88 | 3.78 | 14.5 | 2.61 | 250 |
24 | 3.86 | 4.06 | 12.2 | 3.36 | 260 |
19r | 0b | 2.55 | 0b | 200 | |
22r | 0b | 6.15 | 0b | 227 | |
CNX-1351 | 2.22 | 6.57 | 38.0 | 1.74 | 165 |
ibrutinib | 10.4 |
Mean (and SD) were calculated from three independent measurements.
Values modeled with KinTek Global Kinetic Explorer are lower than 10–11 (nM–1·s–1), no covalent reaction was detected. The full dataset and SD are reported in Table S5.
Mean (and SD) were calculated from three independent measurements.
Stochastic Approach to Calculation of Covalent Reaction Rates as f(kchem, kinact, Ki, and Vr)
Confirmation of Covalent Cys862 Engagement
NanoBRET Assay Detects Drug-Target Engagement in Cells
Validation of Extended Duration of Action in Cellular Washout Studies
Lipophilicity and Lipophilic Ligand Efficiency (LipE)
Assessment of Metabolic Stability
Profiling of Protein and Lipid Kinase Selectivity
Conclusions
Methods
Compounds Synthesis and Characterization
Determination of kchem Using β-Mercaptoethanol (βME)
Structure Modeling of PI3K and mTOR Kinase Complexes
Determination of Inhibitor Binding by TR-FRET
Determination of Inhibitor Dissociation and Kinetic Constants
KinTek Modeling of On-/Off-Target Reactions
Stochastic Approach to Calculation of Covalent Reaction Rates
Protein Mass Spectrometry
Purification of PI3Kα Protein for Structural Studies
Crystallography
NanoBRET Target Engagement Assay
Determination of Cellular PKB/Akt Phosphorylation
In-Cell Western (ICW) Washout Experiments
Metabolic Stability in Rat Liver Microsomes
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacs.1c13568.
Comparison of PI3Kα X-ray crystallographic complexes (Table S1); reaction rate constants (Table S2); KinTek modeling (Figure S1); time- and concentration-dependent TR-FRET data and time-dependent IC50 shift calculations (Figures S2 and S4); summary data (Tables S3 and S5); modeling into PI3Kα (Figures S3 and S5); estimation of reactive volume (Vr) and local concentration of warhead (Table S4); CovDock Schrödinger modeling (Figure S6); kchem/kinact ratio vs reactive volume relations (Figure S7); calculation of σ factors (Table S6); protein mass spectrometry (Figure S8); summary of compound-modified peptides (Table S7); MS/MS spectra (Figure S9); data collection and refinement statistics (molecular replacement, Table S8); NanoBRET experiments with CNX-1351 (Figure S10); lipophilic efficiency (Table S9); metabolic stability (Table S10); affinities to PI3K and PI3K-related kinases (PIKK, Table S11); TREEspot data visualization of KINOMEScan interactions (Figure S11); selectivity profile (Table S12); protein kinase interactions (KINOMEscan data, Table S13); time- and concentration-dependent TR-FRET determination of IC50s (Figures S12 and S13); comparison of chemical structures (Figure S14); synthetic Schemes S1–S3; detailed experimental procedures; synthesis and characterization of intermediates and final compounds;1H NMR,13C{1H} NMR, MALDI-MS, and high-resolution mass spectrometry (HRMS) spectra; HPLC chromatograms; MALDI-MS spectra of β-mercaptoethanol adducts; chemical structures of final compounds, intermediates, and reference compounds; KinTek global kinetic explorer data fitting of TR-FRET traces; and chemical structures of final compounds, intermediates, and reference compounds (PDF)
Proteomics data set file: peptide probabilities assigned with PeptideProphet and iProphet, MS1 XIC traces of the Cys862-containing peptides with different covalent modifications extracted using Skyline v4.2 (XLSX)
The coordinates of compounds 19 and 22 covalently bound to the PI3Kα catalytic subunit p110α have been deposited at www.pdb.org and www.rcsb.org with PDB ID code 7R9V and 7R9Y, respectively.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
The authors thank P. Hebeisen, D. Rageot, A. Melone, and T. Bohnacker for advice and discussions; A. Ontani, A. Christy, J. Klein, P. Knecht, L. Wisson, and R. Triaud for discussions and contribution to synthetic efforts and kinetic experiments; and M. Pfeffer and the mass spectrometry and elemental analysis team at the University of Basel for HRMS data. They also thank A. Chaikuad and S. Knapp for kindly providing the pdb coordinates of their cysteinome template kinase used to compose Figure 7a.
References
This article references 83 other publications.
- 1Miller, V. A.; Hirsh, V.; Cadranel, J.; Chen, Y.-M.; Park, K.; Kim, S.-W.; Zhou, C.; Su, W.-C.; Wang, M.; Sun, Y.; Heo, D. S.; Crino, L.; Tan, E.-H.; Chao, T.-Y.; Shahidi, M.; Cong, X. J.; Lorence, R. M.; Yang, J. C.-H. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol. 2012, 13, 528– 538, DOI: 10.1016/S1470-2045(12)70087-6Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmslyhsbg%253D&md5=6ddd7f552e8ab1ebb43fb699e27894f5Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trialMiller, Vincent A.; Hirsh, Vera; Cadranel, Jacques; Chen, Yuh-Min; Park, Keunchil; Kim, Sang-We; Zhou, Caicun; Su, Wu-Chou; Wang, Mengzhao; Sun, Yan; Heo, Dae Seog; Crino, Lucio; Tan, Eng-Huat; Chao, Tsu-Yi; Shahidi, Mehdi; Cong, Xiuyu Julie; Lorence, Robert M.; Yang, James Chih-HsinLancet Oncology (2012), 13 (5), 528-538CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)Summary: Background: Afatinib, an irreversible ErbB-family blocker, has shown preclin. activity when tested in EGFR mutant models with mutations that confer resistance to EGFR tyrosine-kinase inhibitors. We aimed to assess its efficacy in patients with advanced lung adenocarcinoma with previous treatment failure on EGFR tyrosine-kinase inhibitors. Methods: In this phase 2b/3 trial, we enrolled patients with stage IIIB or IV adenocarcinoma and an Eastern Cooperative Oncol. Group performance (ECOG) performance score of 0-2 who had received one or two previous chemotherapy regimens and had disease progression after at least 12 wk of treatment with erlotinib or gefitinib. We used a computer-generated sequence to randomly allocate patients (2:1) to either afatinib (50 mg per day) or placebo; all patients received best supportive care. Randomisation was done in blocks of three and was stratified by sex and baseline ECOG performance status (0-1 vs 2). Investigators, patients, and the trial sponsor were masked to treatment assignment. The primary endpoint was overall survival (from date of randomisation to death), analyzed on an intention-to-treat basis. This study is registered with ClinicalTrials.gov, no. NCT00656136. Findings: Between May 26, 2008, and Sept 21, 2009, we identified 697 patients, 585 of whom were randomly allocated to treatment (390 to afatinib, 195 to placebo). Median overall survival was 10·8 mo (95% CI 10·0-12·0) in the afatinib group and 12·0 mo (10·2-14·3) in the placebo group (hazard ratio 1·08, 95% CI 0·86-1·35; p=0·74). Median progression-free survival was longer in the afatinib group (3·3 mo, 95% CI 2·79-4·40) than it was in the placebo group (1·1 mo, 0·95-1·68; hazard ratio 0·38, 95% CI 0·31-0·48; p<0·0001). No complete responses to treatment were noted; 29 (7%) patients had a partial response in the afatinib group, as did one patient in the placebo group. Subsequent cancer treatment was given to 257 (68%) patients in the afatinib group and 153 (79%) patients in the placebo group. The most common adverse events in the afatinib group were diarrhoea (339 [87%] of 390 patients; 66 [17%] were grade 3) and rash or acne (305 [78%] patients; 56 [14%] were grade 3). These events occurred less often in the placebo group (18 [9%] of 195 patients had diarrhoea; 31 [16%] had rash or acne), all being grade 1 or 2. Drug-related serious adverse events occurred in 39 (10%) patients in the afatinib group and one (<1%) patient in the placebo group. We recorded two possibly treatment-related deaths in the afatinib group. Interpretation: Although we recorded no benefit in terms of overall survival with afatinib (which might have been affected by cancer treatments given after progression in both groups), our findings for progression-free survival and response to treatment suggest that afatinib could be of some benefit to patients with advanced lung adenocarcinoma who have failed at least 12 wk of previous EGFR tyrosine-kinase inhibitor treatment. Funding: Boehringer Ingelheim Inc.
- 2Pan, Z.; Scheerens, H.; Li, S.-J.; Schultz, B. E.; Sprengeler, P. A.; Burrill, L. C.; Mendonca, R. V.; Sweeney, M. D.; Scott, K. C. K.; Grothaus, P. G.; Jeffery, D. A.; Spoerke, J. M.; Honigberg, L. A.; Young, P. R.; Dalrymple, S. A.; Palmer, J. T. Discovery of Selective Irreversible Inhibitors for Bruton’s Tyrosine Kinase. ChemMedChem 2007, 2, 58– 61, DOI: 10.1002/cmdc.200600221Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjsVWjsbg%253D&md5=023ba8bca9942ad0127d7cf32293e9d2Discovery of selective irreversible inhibitors for bruton's tyrosine kinasePan, Zhengying; Scheerens, Heleen; Li, Shyr-Jiann; Schultz, Brian E.; Sprengeler, Paul A.; Burrill, L. Chuck; Mendonca, Rohan V.; Sweeney, Michael D.; Scott, Keana C. K.; Grothaus, Paul G.; Jeffery, Douglas A.; Spoerke, Jill M.; Honigberg, Lee A.; Young, Peter R.; Dalrymple, Stacie A.; Palmer, James T.ChemMedChem (2007), 2 (1), 58-61CODEN: CHEMGX; ISSN:1860-7179. (Wiley-VCH Verlag GmbH & Co. KGaA)Synthesis and pharmacol. evaluation of a series of peptidomimetic quinoline derivs. was undertaken to evaluate their efficacy in acting as selective irreversible inhibitors of Bruton's tyrosine kinase (Btk). With the relative scarcity of knowledge on the inhibition of Btk it is crucial to discover a potent and selective tool compd. for this kinase. Herein is described the discovery of selective irreversible Btk inhibitors and their efficacy in a mouse RA model.
- 3Yver, A. Osimertinib (AZD9291)─a science-driven, collaborative approach to rapid drug design and development. Ann. Oncol. 2016, 27, 1165– 1170, DOI: 10.1093/annonc/mdw129Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28joslCgsA%253D%253D&md5=903bbb3edf2d109af352ef0354e4bca6Osimertinib (AZD9291)-a science-driven, collaborative approach to rapid drug design and developmentYver AAnnals of oncology : official journal of the European Society for Medical Oncology (2016), 27 (6), 1165-1170 ISSN:.There is no expanded citation for this reference.
- 4Miles, J.; White, Y. Neratinib for the Treatment of Early-Stage HER2-Positive Breast Cancer. J. Adv. Pract. Oncol. 2018, 9, 750– 754Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MzhslyrsQ%253D%253D&md5=0e0f12297730802dfd5806865209deefNeratinib for the Treatment of Early-Stage HER2-Positive Breast CancerMiles Jennifer; White YahsinJournal of the advanced practitioner in oncology (2018), 9 (7), 750-754 ISSN:2150-0878.The treatment of breast cancer has been revolutionized by the development of HER2-targeted treatments for patients who are HER2 positive. The HER2 protein is present at high levels in about 30% of breast cancer patients. These high levels are associated with a greater chance of metastasis, relapse, and decreased survival. The current standard of care for early-stage HER2-positive patients includes treatment with 1 year of trastuzumab therapy. Although trastuzumab has improved outcomes, there is still a 20% chance for tumor recurrence and a 16% chance of death. Neratinib was developed to give patients with early-stage HER2-positive breast cancer an option to increase the disease-free survival rate. The 5-year invasive disease-free survival rate was 90.2% (95% confidence interval = 88.3-91.8) in the neratinib group and 87.7% (95% confidence interval = 85.7-89.4) in the placebo group.
- 5Lau, S. C. M.; Batra, U.; Mok, T. S. K.; Loong, H. H. Dacomitinib in the Management of Advanced Non-Small-Cell Lung Cancer. Drugs 2019, 79, 823– 831, DOI: 10.1007/s40265-019-01115-yGoogle Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFWmtrfI&md5=e09f6baf1fed49df33397d74698b4c52Dacomitinib in the Management of Advanced Non-Small-Cell Lung CancerLau, Sally C. M.; Batra, Ullas; Mok, Tony S. K.; Loong, Herbert H.Drugs (2019), 79 (8), 823-831CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. The use of targeted therapy in the management of epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer is an important milestone in the management of advanced lung cancer. There are several generations of EGFR tyrosine kinase inhibitors available for clin. use. Dacomitinib is a second-generation irreversible EGFR tyrosine kinase inhibitor with early-phase clin. studies showing efficacy in non-small-cell lung cancer. In the recently published ARCHER 1050 phase III study, dacomitinib given at 45 mg/day orally was superior to gefitinib, a first-generation reversible EGFR tyrosine kinase inhibitor, in improving both progression-free survival and overall survival when given as first-line therapy. There is no prospective evidence to support the use of dacomitinib as subsequent therapy in patients previously treated with chemotherapy or a first-generation EGFR tyrosine kinase inhibitor such as gefitinib and erlotinib. Dacomitinib has not demonstrated any benefit in unselected patients with non-small-cell lung cancer, and its use should be limited to those with known EGFR-sensitizing mutations. Dacomitinib is assocd. with increased toxicities of diarrhea, rash, stomatitis, and paronychia compared with first-generation EGFR inhibitors. Global quality of life was maintained when assessed in phase III studies. Overall, dacomitinib is an important first- line agent in EGFR-mutated non-small-cell lung cancer in otherwise fit patients whose toxicities can be well managed.
- 6Davids, M. S. Acalabrutinib for the initial treatment of chronic lymphocytic leukaemia. Lancet 2020, 395, 1234– 1236, DOI: 10.1016/s0140-6736(20)30372-xGoogle Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsVWmsrY%253D&md5=b174e8413a465839fdb071616c793227Acalabrutinib for the initial treatment of chronic lymphocytic leukaemiaDavids, Matthew S.Lancet (2020), 395 (10232), 1234-1236CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A review. This article describes about the acalabrutinib for initial treatment of chronic lymphocytic leukemia.
- 7Guo, Y.; Liu, Y.; Hu, N.; Yu, D.; Zhou, C.; Shi, G.; Zhang, B.; Wei, M.; Liu, J.; Luo, L.; Tang, Z.; Song, H.; Guo, Y.; Liu, X.; Su, D.; Zhang, S.; Song, X.; Zhou, X.; Hong, Y.; Chen, S.; Cheng, Z.; Young, S.; Wei, Q.; Wang, H.; Wang, Q.; Lv, L.; Wang, F.; Xu, H.; Sun, H.; Xing, H.; Li, N.; Zhang, W.; Wang, Z.; Liu, G.; Sun, Z.; Zhou, D.; Li, W.; Liu, L.; Wang, L.; Wang, Z. Discovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase. J. Med. Chem. 2019, 62, 7923– 7940, DOI: 10.1021/acs.jmedchem.9b00687Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsFSku7bP&md5=5cdb551d296e60eb1e82b03b7a0384eeDiscovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton's Tyrosine KinaseGuo, Yunhang; Liu, Ye; Hu, Nan; Yu, Desheng; Zhou, Changyou; Shi, Gongyin; Zhang, Bo; Wei, Min; Liu, Junhua; Luo, Lusong; Tang, Zhiyu; Song, Huipeng; Guo, Yin; Liu, Xuesong; Su, Dan; Zhang, Shuo; Song, Xiaomin; Zhou, Xing; Hong, Yuan; Chen, Shuaishuai; Cheng, Zhenzhen; Young, Steve; Wei, Qiang; Wang, Haisheng; Wang, Qiuwen; Lv, Lei; Wang, Fan; Xu, Haipeng; Sun, Hanzi; Xing, Haimei; Li, Na; Zhang, Wei; Wang, Zhongbo; Liu, Guodong; Sun, Zhijian; Zhou, Dongping; Li, Wei; Liu, Libin; Wang, Lai; Wang, ZhiweiJournal of Medicinal Chemistry (2019), 62 (17), 7923-7940CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Aberrant activation of Bruton's tyrosine kinase (BTK) plays an important role in pathogenesis of B-cell lymphomas, suggesting that inhibition of BTK is useful in the treatment of hematol. malignancies. The discovery of a more selective on-target covalent BTK inhibitor is of high value. Herein, we disclose the discovery and preclin. characterization of a potent, selective, and irreversible BTK inhibitor as our clin. candidate by using in vitro potency, selectivity, pharmacokinetics (PK), and in vivo pharmacodynamic for prioritizing compds. Compd. BGB-3111 (31a, Zanubrutinib) demonstrates (i) potent activity against BTK and excellent selectivity over other TEC, EGFR and Src family kinases, (ii) desirable ADME, excellent in vivo pharmacodynamic in mice and efficacy in OCI-LY10 xenograft models.
- 8Roskoski, R., Jr. Properties of FDA-approved small molecule protein kinase inhibitors: A 2022 update. Pharmacol. Res. 2022, 175, 106037 DOI: 10.1016/j.phrs.2021.106037Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XovVahtr4%253D&md5=d12f02086d0761cc387762f1ca06a95bProperties of FDA-approved small molecule protein kinase inhibitors: A 2022 updateRoskoski Jr., RobertPharmacological Research (2022), 175 (), 106037CODEN: PHMREP; ISSN:1043-6618. (Elsevier Ltd.)A review. Owing to the dysregulation of protein kinase activity in many diseases including cancer, this enzyme family has become one of the most important drug targets in the 21st century. There are 68 FDA-approved therapeutic agents that target about two dozen different protein kinases and six of these drugs were approved in 2021. Of the approved drugs, twelve target protein-serine/threonine protein kinases, four are directed against dual specificity protein kinases (MEK1/2), thirteen block nonreceptor protein-tyrosine kinases, and 39 target receptor protein-tyrosine kinases. The data indicate that 58 of these drugs are prescribed for the treatment of neoplasms (49 against solid tumors including breast, lung, and colon, five against nonsolid tumors such as leukemias, and four against both solid and nonsolid tumors: acalabrutinib, ibrutinib, imatinib, and midostaurin). Three drugs (baricitinib, tofacitinib, upadacitinib) are used for the treatment of inflammatory diseases including rheumatoid arthritis. Of the 68 approved drugs, eighteen are used in the treatment of multiple diseases. The following six drugs received FDA approval in 2021 for the treatment of these specified diseases: belumosudil (graft vs. host disease), infigratinib (cholangiocarcinomas), mobocertinib and tepotinib (specific forms of non-small cell lung cancer), tivozanib (renal cell carcinoma), and trilaciclib (to decrease chemotherapy-induced myelosuppression). All of the FDA-approved drugs are orally effective with the exception of netarsudil, temsirolimus, and the newly approved trilaciclib. This review summarizes the physicochem. properties of all 68 FDA-approved small mol. protein kinase inhibitors including lipophilic efficiency and ligand efficiency.
- 9Riely, G. J.; Neal, J. W.; Camidge, D. R.; Spira, A. I.; Piotrowska, Z.; Costa, D. B.; Tsao, A. S.; Patel, J. D.; Gadgeel, S. M.; Bazhenova, L.; Zhu, V. W.; West, H. L.; Mekhail, T.; Gentzler, R. D.; Nguyen, D.; Vincent, S.; Zhang, S.; Lin, J.; Bunn, V.; Jin, S.; Li, S.; Jänne, P. A. Activity and Safety of Mobocertinib (TAK-788) in Previously Treated Non-Small Cell Lung Cancer with EGFR Exon 20 Insertion Mutations from a Phase I/II Trial. Cancer Discovery 2021, 11, 1688– 1699, DOI: 10.1158/2159-8290.Cd-20-1598Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFyntL7E&md5=adf33e0cf63304cada04d78c187afdf9Activity and safety of mobocertinib (TAK-788) in previousiytreated non-small cell lung cancer with EGFR exon 20 insertion mutations from a phase I/ll trialRiely, Gregory J.; Neal, Joel W.; Ross Camidge, D.; Spira, Alexander I.; Piotrowska, Zofia; Costa, Daniel B.; Tsao, Anne S.; Patel, Jyoti D.; Gadgeel, Shirish M.; Bazhenova, Lyudmila; Zhu, Viola W.; West, Howard L.; Mekhail, Tarek; Gentzler, Ryan D.; Nguyen, Danny; Vincent, Sylvie; Zhang, Steven; Lin, Jianchang; Bunn, Veronica; Jin, Shu; Li, Shuanglian; Janne, Pasi A.Cancer Discovery (2021), 11 (7), 1688-1699CODEN: CDAIB2; ISSN:2159-8274. (American Association for Cancer Research)Mobocertinib, an oral epidermal growth factor receptor (EGFR) inhibitor targeting EGFR gene mutations, including exon 20 insertions (EGFRex20ins), in nonsmall cell lung cancer, was evaluated in a phase I/ll dose-escalation/expansion trial (ClinicalTrials. gov NCT02716116). Dose escalation identified 160 mg/d as the recommended phase 2 dose and max. tolerated dose. Among 136 patients treated with 160 mg/d, the most common any-grade treatment-related adverse events (TRAE; >25%) were diarrhea (83%), nausea (43%), rash (33%), and vomiting (26%), with diarrhea (21%) the only grade ≥3 TRAE >5%. Among 28 EGFRex20ins patients treated at 160 mg/d, the investigator-assessed confirmed response rate was 43% (12/28; 95% confidence interval, 24%-63%) with median duration of response of 14 mo (5.0-not reached) and median progression-free survival of 7.3 mo (4.4-15.6). Mobocertinib demonstrated antitumor activity in patients with diverse EGFRex20ins variants with a safety profile consistent with other EGFR inhibitors. SIGNIFICANCE: No oral EGFR-targeted therapies are currently approved for patients with EGFRex- 20ins NSCLC. Mobocertinib demonstrated antitumor activity with manageable toxicity in patients with advanced EGFRex20ins NSCLC in this study, supporting addnl. development of mobocertinib in this patient population.
- 10Singh, J.; Petter, R. C.; Baillie, T. A.; Whitty, A. The resurgence of covalent drugs. Nat. Rev. Drug Discovery 2011, 10, 307– 317, DOI: 10.1038/nrd3410Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXktVGmu7g%253D&md5=2190289081e151416c097be4a5b04460The resurgence of covalent drugsSingh, Juswinder; Petter, Russell C.; Baillie, Thomas A.; Whitty, AdrianNature Reviews Drug Discovery (2011), 10 (4), 307-317CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Covalent drugs haveproved to be successful therapies for various indications, but largely owing to safety concerns, they are rarely considered when initiating a target-directed drug discovery project. There is a need to reassess this important class of drugs, and to reconcile the discordance between the historic success of covalent drugs and the reluctance of most drug discovery teams to include them in their armamentarium. This Review surveys the prevalence and pharmacol. advantages of covalent drugs, discusses how potential risks and challenges may be addressed through innovative design, and presents the broad opportunities provided by targeted covalent inhibitors.
- 11London, N.; Miller, R. M.; Krishnan, S.; Uchida, K.; Irwin, J. J.; Eidam, O.; Gibold, L.; Cimermančič, P.; Bonnet, R.; Shoichet, B. K.; Taunton, J. Covalent docking of large libraries for the discovery of chemical probes. Nat. Chem. Biol. 2014, 10, 1066– 1072, DOI: 10.1038/nchembio.1666Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVSqsbjE&md5=4e5fbeb8f67cb82d464d57069ba0c93aCovalent docking of large libraries for the discovery of chemical probesLondon, Nir; Miller, Rand M.; Krishnan, Shyam; Uchida, Kenji; Irwin, John J.; Eidam, Oliv; Gibold, Lucie; Cimermancic, Peter; Bonnet, Richard; Shoichet, Brian K.; Taunton, JackNature Chemical Biology (2014), 10 (12), 1066-1072CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Chem. probes that form a covalent bond with a protein target often show enhanced selectivity, potency and utility for biol. studies. Despite these advantages, protein-reactive compds. are usually avoided in high-throughput screening campaigns. Here we describe a general method (DOCKovalent) for screening large virtual libraries of electrophilic small mols. We apply this method prospectively to discover reversible covalent fragments that target distinct protein nucleophiles, including the catalytic serine of AmpC β-lactamase and noncatalytic cysteines in RSK2, MSK1 and JAK3 kinases. We identify submicromolar to low-nanomolar hits with high ligand efficiency, cellular activity and selectivity, including what are to our knowledge the first reported reversible covalent inhibitors of JAK3. Crystal structures of inhibitor complexes with AmpC and RSK2 confirm the docking predictions and guide further optimization. As covalent virtual screening may have broad utility for the rapid discovery of chem. probes, we have made the method freely available through an automated web server (http://covalent.docking.org/).
- 12Zhao, Z.; Liu, Q.; Bliven, S.; Xie, L.; Bourne, P. E. Determining Cysteines Available for Covalent Inhibition Across the Human Kinome. J. Med. Chem. 2017, 60, 2879– 2889, DOI: 10.1021/acs.jmedchem.6b01815Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkvVGjsLg%253D&md5=22ea95ce38eb0836d32c1332dba54fadDetermining Cysteines Available for Covalent Inhibition Across the Human KinomeZhao, Zheng; Liu, Qingsong; Bliven, Spencer; Xie, Lei; Bourne, Philip E.Journal of Medicinal Chemistry (2017), 60 (7), 2879-2889CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Covalently bound protein kinase inhibitors have been frequently designed to target non-catalytic Cys residues at the ATP binding site. Thus, it is important to know if a given Cys residue can form a covalent bond. Here, we combined a function-site interaction fingerprint method and DFT calcns. to det. the potential of Cys residues to form a covalent interaction with an inhibitor. By harnessing the human structural kinome, a comprehensive structure-based binding site Cys dataset was assembled. The orientation of the Cys SH group indicated which Cys residues could potentially form covalent bonds. These covalent inhibitor accessible Cys residues were located within 5 kinase regions (P-loop, roof of pocket, front pocket, catalytic-2 of the catalytic loop and DFG-3 close to the DFG peptide). In an independent test set, these Cys residues covered 95% of covalent kinase inhibitors. Thus, this study provides new insights into Cys reactivity and preference which is important for the prospective development of covalent kinase inhibitors.
- 13Liu, Q.; Sabnis, Y.; Zhao, Z.; Zhang, T.; Buhrlage, S. J.; Jones, L. H.; Gray, N. S. Developing irreversible inhibitors of the protein kinase cysteinome. Chem. Biol. 2013, 20, 146– 159, DOI: 10.1016/j.chembiol.2012.12.006Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtFWitbs%253D&md5=cef80823123bae88362e816a0a638133Developing Irreversible Inhibitors of the Protein Kinase CysteinomeLiu, Qingsong; Sabnis, Yogesh; Zhao, Zheng; Zhang, Tinghu; Buhrlage, Sara J.; Jones, Lyn H.; Gray, Nathanael S.Chemistry & Biology (Oxford, United Kingdom) (2013), 20 (2), 146-159CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)A review. Protein kinases are a large family of approx. 530 highly conserved enzymes that transfer a γ-phosphate group from ATP to a variety of amino acid residues, such as tyrosine, serine, and threonine, that serves as a ubiquitous mechanism for cellular signal transduction. The clin. success of a no. of kinase-directed drugs and the frequent observation of disease causing mutations in protein kinases suggest that a large no. of kinases may represent therapeutically relevant targets. To date, the majority of clin. and preclin. kinase inhibitors are ATP competitive, noncovalent inhibitors that achieve selectivity through recognition of unique features of particular protein kinases. Recently, there has been renewed interest in the development of irreversible inhibitors that form covalent bonds with cysteine or other nucleophilic residues in the ATP-binding pocket. Irreversible kinase inhibitors have a no. of potential advantages including prolonged pharmacodynamics, suitability for rational design, high potency, and ability to validate pharmacol. specificity through mutation of the reactive cysteine residue. Here, we review recent efforts to develop cysteine-targeted irreversible protein kinase inhibitors and discuss their modes of recognizing the ATP-binding pocket and their biol. activity profiles. In addn., we provided an informatics assessment of the potential "kinase cysteinome" and discuss strategies for the efficient development of new covalent inhibitors.
- 14Barf, T.; Covey, T.; Izumi, R.; van de Kar, B.; Gulrajani, M.; van Lith, B.; van Hoek, M.; de Zwart, E.; Mittag, D.; Demont, D.; Verkaik, S.; Krantz, F.; Pearson, P. G.; Ulrich, R.; Kaptein, A. Acalabrutinib (ACP-196): A Covalent Bruton Tyrosine Kinase Inhibitor with a Differentiated Selectivity and In Vivo Potency Profile. J. Pharmacol. Exp. Ther. 2017, 363, 240– 252, DOI: 10.1124/jpet.117.242909Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtleltrnO&md5=73cbd3a87396abbf8cb20e4361f96f64Acalabrutinib (ACP-196): a covalent Bruton tyrosine kinase inhibitor with a differentiated selectivity and in vivo potency profileBarf, Tjeerd; Covey, Todd; Izumi, Raquel; van de Kar, Bas; Gulrajani, Michael; van Lith, Bart; van Hoek, Maaike; de Zwart, Edwin; Mittag, Diana; Demont, Dennis; Verkaik, Saskia; Krantz, Fanny; Pearson, Paul G.; Ulrich, Roger; Kaptein, AllardJournal of Pharmacology and Experimental Therapeutics (2017), 363 (2), 240-252CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)Several small-mol. Bruton tyrosine kinase (BTK) inhibitors are in development for B cell malignancies and autoimmune disorders, each characterized by distinct potency and selectivity patterns. Herein we describe the pharmacol. characterization of BTK inhibitor acalabrutinib [compd. 1, ACP-196 (4-[8- amino-3-[(2S)-1-but-2-ynoylpyrrolidin-2-yl]imidazo[1,5-a]pyrazin1-yl]-N-(2-pyridyl)benzamide)]. Acalabrutinib possesses a reactive butynamide group that binds covalently to Cys481 in BTK. Relative to the other BTK inhibitors described here, the reduced intrinsic reactivity of acalabrutinib helps to limit inhibition of off-target kinases having cysteine-mediated covalent binding potential. Acalabrutinib demonstrated higher biochem. and cellular selectivity than ibrutinib and spebrutinib (compds. 2 and 3, resp.). Importantly, off-target kinases, such as epidermal growth factor receptor (EGFR) and interleukin 2-inducible T cell kinase (ITK), were not inhibited. Detn. of the inhibitory potential of anti-IgM-induced CD69 expression in human peripheral blood mononuclear cells and whole blood demonstrated that acalabrutinib is a potent functional BTK inhibitor. In vivo evaluation in mice revealed that acalabrutinib is more potent than ibrutinib and spebrutinib. Preclin. and clin. studies showed that the level and duration of BTK occupancy correlates with in vivo efficacy. Evaluation of the pharmacokinetic properties of acalabrutinib in healthy adult volunteers demonstrated rapid absorption and fast elimination. In these healthy individuals, a single oral dose of 100 mg showed approx. 99% median target coverage at 3 and 12 h and around 90% at 24 h in peripheral B cells. In conclusion, acalabrutinib is a BTK inhibitor with key pharmacol. differentiators vs. ibrutinib and spebrutinib and is currently being evaluated in clin. trials.
- 15Cheng, H.; Planken, S. Precedence and Promise of Covalent Inhibitors of EGFR and KRAS for Patients with Non-Small-Cell Lung Cancer. ACS Med. Chem. Lett. 2018, 9, 861– 863, DOI: 10.1021/acsmedchemlett.8b00311Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVagtrjE&md5=0b1f65c9994dc226a5c6d72aa7789071Precedence and Promise of Covalent Inhibitors of EGFR and KRAS for Patients with Non-Small-Cell Lung CancerCheng, Hengmiao; Planken, SimonACS Medicinal Chemistry Letters (2018), 9 (9), 861-863CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (KRAS) oncogenic mutations are leading causes for lung cancer. Extensive drug discovery efforts targeting EGFR have led to the discovery and FDA approval of both reversible and covalent inhibitors. Second and third generation covalent inhibitors for EGFR have also been described, with the latter targeting specific emerging mutations. After decades of extensive effort, KRAS is widely regarded as an intractable therapeutic target; however, recent publications suggest covalent inhibition is a promising strategy to deliver inhibitors of the KRASG12C mutation.
- 16Tomassi, S.; Lategahn, J.; Engel, J.; Keul, M.; Tumbrink, H. L.; Ketzer, J.; Mühlenberg, T.; Baumann, M.; Schultz-Fademrecht, C.; Bauer, S.; Rauh, D. Indazole-Based Covalent Inhibitors To Target Drug-Resistant Epidermal Growth Factor Receptor. J. Med. Chem. 2017, 60, 2361– 2372, DOI: 10.1021/acs.jmedchem.6b01626Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1WltL0%253D&md5=25e24252dc5a03ec5349359289ebd808Indazole-Based Covalent Inhibitors To Target Drug-Resistant Epidermal Growth Factor ReceptorTomassi, Stefano; Lategahn, Jonas; Engel, Julian; Keul, Marina; Tumbrink, Hannah L.; Ketzer, Julia; Muehlenberg, Thomas; Baumann, Matthias; Schultz-Fademrecht, Carsten; Bauer, Sebastian; Rauh, DanielJournal of Medicinal Chemistry (2017), 60 (6), 2361-2372CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The specific targeting of oncogenic mutant epidermal growth factor receptor (EGFR) is a breakthrough in targeted cancer therapy and marks a drastic change in the treatment of non-small cell lung cancer (NSCLC). The recurrent emergence of resistance to these targeted drugs requires the development of novel chem. entities that efficiently inhibit drug-resistant EGFR. Herein, the authors report the optimization process for a hit compd. that has emerged from a phenotypic screen resulting in indazole-based compds. These inhibitors are conformationally less flexible, target gatekeeper mutated drug resistant EGFR-L858R/T790M and covalently alkylate Cys 797. Western blot anal., as well as characterization of the binding kinetics and kinase selectivity profiling, substantiates the authors' approach of targeting drug-resistant EGFR-L858R/T790M with inhibitors incorporating the indazole as hinge binder.
- 17Planken, S.; Behenna, D. C.; Nair, S. K.; Johnson, T. O.; Nagata, A.; Almaden, C.; Bailey, S.; Ballard, T. E.; Bernier, L.; Cheng, H.; Cho-Schultz, S.; Dalvie, D.; Deal, J. G.; Dinh, D. M.; Edwards, M. P.; Ferre, R. A.; Gajiwala, K. S.; Hemkens, M.; Kania, R. S.; Kath, J. C.; Matthews, J.; Murray, B. W.; Niessen, S.; Orr, S. T.; Pairish, M.; Sach, N. W.; Shen, H.; Shi, M.; Solowiej, J.; Tran, K.; Tseng, E.; Vicini, P.; Wang, Y.; Weinrich, S. L.; Zhou, R.; Zientek, M.; Liu, L.; Luo, Y.; Xin, S.; Zhang, C.; Lafontaine, J. Discovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFR. J. Med. Chem. 2017, 60, 3002– 3019, DOI: 10.1021/acs.jmedchem.6b01894Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXktFOmtrw%253D&md5=14a6700ed770621bdbd84f7931276f0eDiscovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFRPlanken, Simon; Behenna, Douglas C.; Nair, Sajiv K.; Johnson, Theodore O.; Nagata, Asako; Almaden, Chau; Bailey, Simon; Ballard, T. Eric; Bernier, Louise; Cheng, Hengmiao; Cho-Schultz, Sujin; Dalvie, Deepak; Deal, Judith G.; Dinh, Dac M.; Edwards, Martin P.; Ferre, Rose Ann; Gajiwala, Ketan S.; Hemkens, Michelle; Kania, Robert S.; Kath, John C.; Matthews, Jean; Murray, Brion W.; Niessen, Sherry; Orr, Suvi T. M.; Pairish, Mason; Sach, Neal W.; Shen, Hong; Shi, Manli; Solowiej, James; Tran, Khanh; Tseng, Elaine; Vicini, Paolo; Wang, Yuli; Weinrich, Scott L.; Zhou, Ru; Zientek, Michael; Liu, Longqing; Luo, Yiqin; Xin, Shuibo; Zhang, Chengyi; Lafontaine, JenniferJournal of Medicinal Chemistry (2017), 60 (7), 3002-3019CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mutant epidermal growth factor receptor (EGFR) is a major driver of non-small-cell lung cancer (NSCLC). Marketed first generation inhibitors, such as erlotinib, effect a transient beneficial response in EGFR mutant NSCLC patients before resistance mechanisms render these inhibitors ineffective. Secondary oncogenic EGFR mutations account for approx. 50% of relapses, the most common being the gatekeeper T790M substitution that renders existing therapies ineffective. The discovery of PF-06459988 (1), an irreversible pyrrolopyrimidine inhibitor of EGFR T790M mutants, was recently disclosed. Herein, we describe our continued efforts to achieve potency across EGFR oncogenic mutations and improved kinome selectivity, resulting in the discovery of clin. candidate PF-06747775 (21), which provides potent EGFR activity against the four common mutants (exon 19 deletion (Del), L858R, and double mutants T790M/L858R and T790M/Del), selectivity over wild-type EGFR, and desirable ADME properties. Compd. 21 is currently being evaluated in phase-I clin. trials of mutant EGFR driven NSCLC.
- 18Zapf, C. W.; Gerstenberger, B. S.; Xing, L.; Limburg, D. C.; Anderson, D. R.; Caspers, N.; Han, S.; Aulabaugh, A.; Kurumbail, R.; Shakya, S.; Li, X.; Spaulding, V.; Czerwinski, R. M.; Seth, N.; Medley, Q. G. Covalent inhibitors of interleukin-2 inducible T cell kinase (itk) with nanomolar potency in a whole-blood assay. J. Med. Chem. 2012, 55, 10047– 10063, DOI: 10.1021/jm301190sGoogle Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFOlsbnP&md5=7e39a17151dc9493d13439bc48fa83a3Covalent Inhibitors of Interleukin-2 Inducible T Cell Kinase (Itk) with Nanomolar Potency in a Whole-Blood AssayZapf, Christoph W.; Gerstenberger, Brian S.; Xing, Li; Limburg, David C.; Anderson, David R.; Caspers, Nicole; Han, Seungil; Aulabaugh, Ann; Kurumbail, Ravi; Shakya, Subarna; Li, Xin; Spaulding, Vikki; Czerwinski, Robert M.; Seth, Nilufer; Medley, Quintus G.Journal of Medicinal Chemistry (2012), 55 (22), 10047-10063CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We wish to report a strategy that targets interleukin-2 inducible T cell kinase (Itk) with covalent inhibitors. Thus far, covalent inhibition of Itk has not been disclosed in the literature. Structure-based drug design was utilized to achieve low nanomolar potency of the disclosed series even at high ATP concns. Kinetic measurements confirmed an irreversible binding mode with off-rate half-lives exceeding 24 h and moderate on-rates. The analogs are highly potent in a cellular IP1 assay as well as in a human whole-blood (hWB) assay. Despite a half-life of approx. 2 h in resting primary T cells, the covalent inhibition of Itk resulted in functional silencing of the TCR pathway for more than 24 h. This prolonged effect indicates that covalent inhibition is a viable strategy to target the inactivation of Itk.
- 19Ward, R. A.; Colclough, N.; Challinor, M.; Debreczeni, J. E.; Eckersley, K.; Fairley, G.; Feron, L.; Flemington, V.; Graham, M. A.; Greenwood, R.; Hopcroft, P.; Howard, T. D.; James, M.; Jones, C. D.; Jones, C. R.; Renshaw, J.; Roberts, K.; Snow, L.; Tonge, M.; Yeung, K. Structure-Guided Design of Highly Selective and Potent Covalent Inhibitors of ERK1/2. J. Med. Chem. 2015, 58, 4790– 4801, DOI: 10.1021/acs.jmedchem.5b00466Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXosVChs7c%253D&md5=40b47d8671c966434434f056ac88b3a7Structure-Guided Design of Highly Selective and Potent Covalent Inhibitors of ERK1/2Ward, Richard A.; Colclough, Nicola; Challinor, Mairi; Debreczeni, Judit E.; Eckersley, Kay; Fairley, Gary; Feron, Lyman; Flemington, Vikki; Graham, Mark A.; Greenwood, Ryan; Hopcroft, Philip; Howard, Tina D.; James, Michael; Jones, Clifford D.; Jones, Christopher R.; Renshaw, Jonathan; Roberts, Karen; Snow, Lindsay; Tonge, Michael; Yeung, KayJournal of Medicinal Chemistry (2015), 58 (11), 4790-4801CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The RAS/RAF/MEK/ERK signaling pathway has been targeted with a no. of small mol. inhibitors in oncol. clin. development across multiple disease indications. Importantly, cell lines with acquired resistance to B-RAF and MEK inhibitors have been shown to maintain sensitivity to ERK1/2 inhibition by small mol. inhibitors. There are a no. of selective, noncovalent ERK1/2 inhibitors reported along with the promiscuous hypothemycin (and related analogs) that act via a covalent mechanism of action. This article reports the identification of multiple series of highly selective covalent ERK1/2 inhibitors informed by structure-based drug design (SBDD). As a starting point for these covalent inhibitors, reported ERK1/2 inhibitors and a chem. series identified via high-throughput screening were exploited. These approaches resulted in the identification of selective covalent tool compds. for potential in vitro and in vivo studies to assess the risks and or benefits of targeting this pathway through such a mechanism of action.
- 20Angst, D.; Gessier, F.; Janser, P.; Vulpetti, A.; Wälchli, R.; Beerli, C.; Littlewood-Evans, A.; Dawson, J.; Nuesslein-Hildesheim, B.; Wieczorek, G.; Gutmann, S.; Scheufler, C.; Hinniger, A.; Zimmerlin, A.; Funhoff, E. G.; Pulz, R.; Cenni, B. Discovery of LOU064 (Remibrutinib), a Potent and Highly Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase. J. Med. Chem. 2020, 63, 5102– 5118, DOI: 10.1021/acs.jmedchem.9b01916Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjs1KiurY%253D&md5=9122b310c294d6071ae7cd73cdb91b3dDiscovery of LOU064 (Remibrutinib), a Potent and Highly Selective Covalent Inhibitor of Bruton's Tyrosine KinaseAngst, Daniela; Gessier, Francois; Janser, Philipp; Vulpetti, Anna; Walchli, Rudolf; Beerli, Christian; Littlewood-Evans, Amanda; Dawson, Janet; Nuesslein-Hildesheim, Barbara; Wieczorek, Grazyna; Gutmann, Sascha; Scheufler, Clemens; Hinniger, Alexandra; Zimmerlin, Alfred; Funhoff, Enrico G.; Pulz, Robert; Cenni, BrunoJournal of Medicinal Chemistry (2020), 63 (10), 5102-5118CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bruton's tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a central role in immunity and is considered an attractive target for treating autoimmune diseases. The use of currently marketed covalent BTK inhibitors is limited to oncol. indications based on their suboptimal kinase selectivity. We describe the discovery and preclin. profile of LOU064 (remibrutinib, 25), a potent, highly selective covalent BTK inhibitor. LOU064 exhibits an exquisite kinase selectivity due to binding to an inactive conformation of BTK and has the potential for a best-in-class covalent BTK inhibitor for the treatment of autoimmune diseases. It demonstrates potent in vivo target occupancy with an EC90 of 1.6 mg/kg and dose-dependent efficacy in rat collagen-induced arthritis. LOU064 is currently being tested in phase 2 clin. studies for chronic spontaneous urticaria and Sjoegren's syndrome.
- 21Gurbani, D.; Du, G.; Henning, N. J.; Rao, S.; Bera, A. K.; Zhang, T.; Gray, N. S.; Westover, K. D. Structure and Characterization of a Covalent Inhibitor of Src Kinase. Front. Mol. Biosci. 2020, 7, 81 DOI: 10.3389/fmolb.2020.00081Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFKksLbF&md5=0b1b4bbea40bb1cfaad8fb8fbbff45e8Structure and characterization of a covalent inhibitor of Src kinaseGurbani, Deepak; Du, Guangyan; Henning, Nathaniel J.; Rao, Suman; Bera, Asim K.; Zhang, Tinghu; Gray, Nathanael S.; Westover, Kenneth D.Frontiers in Molecular Biosciences (2020), 7 (), 00081CODEN: FMBRBS; ISSN:2296-889X. (Frontiers Media S.A.)Unregulated Src activity promotes malignant processes in cancer, but no Src-directed targeted therapies are used clin., possibly because early Src inhibitors produce off-target effects leading to toxicity. Improved selective Src inhibitors may enable Src-directed therapies. Previously, we reported an irreversible Src inhibitor, DGY-06-116, based on the hybridization of dasatinib and a promiscuous covalent kinase probe SM1-71. Here, we report biochem. and biophys. characterization of this compd. An x-ray co-crystal structure of DGY-06-116: Src shows a covalent interaction with the kinase p-loop and occupancy of the back hydrophobic kinase pocket, explaining its high potency, and selectivity. However, a reversible analog also shows similar potency. Kinetic anal. shows a slow inactivation rate compared to other clin. approved covalent kinase inhibitors, consistent with a need for p-loop movement prior to covalent bond formation. Overall, these results suggest that a strong reversible interaction is required to allow sufficient time for the covalent reaction to occur. Further optimization of the covalent linker may improve the kinetics of covalent bond formation.
- 22Abdeldayem, A.; Raouf, Y. S.; Constantinescu, S. N.; Moriggl, R.; Gunning, P. T. Advances in covalent kinase inhibitors. Chem. Soc. Rev. 2020, 49, 2617– 2687, DOI: 10.1039/c9cs00720bGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvVygsr8%253D&md5=88c6e097cb0b3619a53ceb1e03d19971Advances in covalent kinase inhibitorsAbdeldayem, Ayah; Raouf, Yasir S.; Constantinescu, Stefan N.; Moriggl, Richard; Gunning, Patrick T.Chemical Society Reviews (2020), 49 (9), 2617-2687CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)Over the past decade, covalent kinase inhibitors (CKI) have seen a resurgence in drug discovery. Covalency affords a unique set of advantages as well as challenges relative to their non-covalent counterpart. After reversible protein target recognition and binding, covalent inhibitors irreversibly modify a proximal nucleophilic residue on the protein via reaction with an electrophile. To date, the acrylamide group remains the predominantly employed electrophile in CKI development, with its incorporation in the majority of clin. candidates and FDA approved covalent therapies. Nonetheless, in recent years considerable efforts have ensued to characterize alternative electrophiles that exhibit irreversible or reversibly covalent binding mechanisms towards cysteine thiols and other amino acids. This review article provides a comprehensive overview of CKIs reported in the literature over a decade period, 2007-2018. Emphasis is placed on the rationale behind warhead choice, optimization approach, and inhibitor design. Current FDA approved CKIs are also highlighted, in addn. to a detailed anal. of the common trends and themes obsd. within the listed data set.
- 23Yan, G.; Zhong, X.; Pu, C.; Yue, L.; Shan, H.; Lan, S.; Zhou, M.; Hou, X.; Yang, J.; Li, D.; Fan, S.; Li, R. Targeting Cysteine Located Outside the Active Site: An Effective Strategy for Covalent ALKi Design. J. Med. Chem. 2021, 64, 1558– 1569, DOI: 10.1021/acs.jmedchem.0c01707Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFWisr4%253D&md5=216a6fc20ae93c196947e73e9c933e85Targeting Cysteine Located Outside the Active Site: An Effective Strategy for Covalent ALKi DesignYan, Guoyi; Zhong, Xinxin; Pu, Chunlan; Yue, Lin; Shan, Huifang; Lan, Suke; Zhou, Meng; Hou, Xueyan; Yang, Jie; Li, Deyu; Fan, Shilong; Li, RuiJournal of Medicinal Chemistry (2021), 64 (3), 1558-1569CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Potent inhibitors of ALK are highly desired because of the occurrence of drug resistance. We herein firstly report the development of a rationally designed inhibitor, Con B-1, which can covalently bind to Cys1259, a cysteine located outside the ALK active site by linking a warhead with Ceritinib through a 2,2'-Oxybis(ethylamine) linker. The in vitro and in vivo assays showed ConB-1 is a potent selective ALKi with low toxicity to normal cells. In addn., the mol. showed significant improvement of anticancer activities and potential antidrug resistant activity compared with Ceritinib, demonstrating the covalent inhibitor of ALK can be a promising drug candidate for the treatment of NSCLC. This work may provide a novel perspective on the design of covalent inhibitors.
- 24Jiang, B.; Jiang, J.; Kaltheuner, I. H.; Iniguez, A. B.; Anand, K.; Ferguson, F. M.; Ficarro, S. B.; Seong, B. K. A.; Greifenberg, A. K.; Dust, S.; Kwiatkowski, N. P.; Marto, J. A.; Stegmaier, K.; Zhang, T.; Geyer, M.; Gray, N. S. Structure-activity relationship study of THZ531 derivatives enables the discovery of BSJ-01-175 as a dual CDK12/13 covalent inhibitor with efficacy in Ewing sarcoma. Eur. J. Med. Chem. 2021, 221, 113481 DOI: 10.1016/j.ejmech.2021.113481Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVWqtb3N&md5=d6e8546a93cb97657e9cb24bb86dc5bcStructure-activity relationship study of THZ531 derivatives enables the discovery of BSJ-01-175 as a dual CDK12/13 covalent inhibitor with efficacy in Ewing sarcomaJiang, Baishan; Jiang, Jie; Kaltheuner, Ines H.; Iniguez, Amanda Balboni; Anand, Kanchan; Ferguson, Fleur M.; Ficarro, Scott B.; Seong, Bo Kyung Alex; Greifenberg, Ann Katrin; Dust, Sofia; Kwiatkowski, Nicholas P.; Marto, Jarrod A.; Stegmaier, Kimberly; Zhang, Tinghu; Geyer, Matthias; Gray, Nathanael S.European Journal of Medicinal Chemistry (2021), 221 (), 113481CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Development of inhibitors targeting CDK12/13 is of increasing interest as a potential therapy for cancers as these compds. inhibit transcription of DNA damage response (DDR) genes. We previously described THZ531, a covalent inhibitor with selectivity for CDK12/13. In order to elucidate structure-activity relationship (SAR), we have undertaken a medicinal chem. campaign and established a focused library of THZ531 analogs. Among these analogs, BSJ-01-175 demonstrates exquisite selectivity, potent inhibition of RNA polymerase II phosphorylation, and downregulation of CDK12-targeted genes in cancer cells. A 3.0 Å co-crystal structure with CDK12/CycK provides a structural rational for selective targeting of Cys1039 located in a C-terminal extension from the kinase domain. With moderate pharmacokinetic properties, BSJ-01-175 exhibits efficacy against an Ewing sarcoma tumor growth in a patient-derived xenograft (PDX) mouse model following 10 mg/kg once a day, i.p. administration. Taken together, BSJ-01-175 represents the first selective CDK12/13 covalent inhibitor with in vivo efficacy reported to date.
- 25Zhang, T.; Kwiatkowski, N.; Olson, C. M.; Dixon-Clarke, S. E.; Abraham, B. J.; Greifenberg, A. K.; Ficarro, S. B.; Elkins, J. M.; Liang, Y.; Hannett, N. M.; Manz, T.; Hao, M.; Bartkowiak, B.; Greenleaf, A. L.; Marto, J. A.; Geyer, M.; Bullock, A. N.; Young, R. A.; Gray, N. S. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat. Chem. Biol. 2016, 12, 876– 884, DOI: 10.1038/nchembio.2166Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlyqsbbJ&md5=5ac265105d1654e129c3c1325572f389Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitorsZhang, Tinghu; Kwiatkowski, Nicholas; Olson, Calla M.; Dixon-Clarke, Sarah E.; Abraham, Brian J.; Greifenberg, Ann K.; Ficarro, Scott B.; Elkins, Jonathan M.; Liang, Yanke; Hannett, Nancy M.; Manz, Theresa; Hao, Mingfeng; Bartkowiak, Bartlomiej; Greenleaf, Arno L.; Marto, Jarrod A.; Geyer, Matthias; Bullock, Alex N.; Young, Richard A.; Gray, Nathanael S.Nature Chemical Biology (2016), 12 (10), 876-884CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Cyclin-dependent kinases 12 and 13 (CDK12 and CDK13) play crit. roles in the regulation of gene transcription. However, the absence of CDK12 and CDK13 inhibitors has hindered the ability to investigate the consequences of their inhibition in healthy cells and cancer cells. Here we describe the rational design of a first-in-class CDK12 and CDK13 covalent inhibitor, THZ531. Co-crystn. of THZ531 with CDK12-cyclin K indicates that THZ531 irreversibly targets a cysteine located outside the kinase domain. THZ531 causes a loss of gene expression with concurrent loss of elongating and hyperphosphorylated RNA polymerase II. In particular, THZ531 substantially decreases the expression of DNA damage response genes and key super-enhancer-assocd. transcription factor genes. Coincident with transcriptional perturbation, THZ531 dramatically induced apoptotic cell death. Small mols. capable of specifically targeting CDK12 and CDK13 may thus help identify cancer subtypes that are particularly dependent on their kinase activities.
- 26Kwiatkowski, N.; Zhang, T.; Rahl, P. B.; Abraham, B. J.; Reddy, J.; Ficarro, S. B.; Dastur, A.; Amzallag, A.; Ramaswamy, S.; Tesar, B.; Jenkins, C. E.; Hannett, N. M.; McMillin, D.; Sanda, T.; Sim, T.; Kim, N. D.; Look, T.; Mitsiades, C. S.; Weng, A. P.; Brown, J. R.; Benes, C. H.; Marto, J. A.; Young, R. A.; Gray, N. S. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014, 511, 616– 620, DOI: 10.1038/nature13393Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1ChurnF&md5=895a28fb536663a31e7558fe79ce628bTargeting transcription regulation in cancer with a covalent CDK7 inhibitorKwiatkowski, Nicholas; Zhang, Tinghu; Rahl, Peter B.; Abraham, Brian J.; Reddy, Jessica; Ficarro, Scott B.; Dastur, Anahita; Amzallag, Arnaud; Ramaswamy, Sridhar; Tesar, Bethany; Jenkins, Catherine E.; Hannett, Nancy M.; McMillin, Douglas; Sanda, Takaomi; Sim, Taebo; Kim, Nam Doo; Look, Thomas; Mitsiades, Constantine S.; Weng, Andrew P.; Brown, Jennifer R.; Benes, Cyril H.; Marto, Jarrod A.; Young, Richard A.; Gray, Nathanael S.Nature (London, United Kingdom) (2014), 511 (7511), 616-620CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Tumor oncogenes include transcription factors that co-opt the general transcriptional machinery to sustain the oncogenic state, but direct pharmacol. inhibition of transcription factors has so far proven difficult. However, the transcriptional machinery contains various enzymic cofactors that can be targeted for the development of new therapeutic candidates, including cyclin-dependent kinases (CDKs). Here the authors present the discovery and characterization of a covalent CDK7 inhibitor, THZ1, which has the unprecedented ability to target a remote cysteine residue located outside of the canonical kinase domain, providing an unanticipated means of achieving selectivity for CDK7. Cancer cell-line profiling indicates that a subset of cancer cell lines, including human T-cell acute lymphoblastic leukemia (T-ALL), have exceptional sensitivity to THZ1. Genome-wide anal. in Jurkat T-ALL cells shows that THZ1 disproportionally affects transcription of RUNX1 and suggests that sensitivity to THZ1 may be due to vulnerability conferred by the RUNX1 super-enhancer and the key role of RUNX1 in the core transcriptional regulatory circuitry of these tumor cells. Pharmacol. modulation of CDK7 kinase activity may thus provide an approach to identify and treat tumor types that are dependent on transcription for maintenance of the oncogenic state.
- 27Nacht, M.; Qiao, L.; Sheets, M. P.; St Martin, T.; Labenski, M.; Mazdiyasni, H.; Karp, R.; Zhu, Z.; Chaturvedi, P.; Bhavsar, D.; Niu, D.; Westlin, W.; Petter, R. C.; Medikonda, A. P.; Singh, J. Discovery of a Potent and Isoform-Selective Targeted Covalent Inhibitor of the Lipid Kinase PI3Kα. J. Med. Chem. 2013, 56, 712– 721, DOI: 10.1021/jm3008745Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWjsr4%253D&md5=42bf6d9221fdb9eafe010064807d22b9Discovery of a Potent and Isoform-Selective Targeted Covalent Inhibitor of the Lipid Kinase PI3KαNacht, Mariana; Qiao, Lixin; Sheets, Michael P.; St. Martin, Thia; Labenski, Matthew; Mazdiyasni, Hormoz; Karp, Russell; Zhu, Zhendong; Chaturvedi, Prasoon; Bhavsar, Deepa; Niu, Deqiang; Westlin, William; Petter, Russell C.; Medikonda, Aravind Prasad; Singh, JuswinderJournal of Medicinal Chemistry (2013), 56 (3), 712-721CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallog., that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kβ, -γ, and -δ are not covalently modified. 3 is able to potently (EC50 < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI50 < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biol. impact of selectively targeting PI3Kα.
- 28Wymann, M. P.; Schneiter, R. Lipid signalling in disease. Nat. Rev. Mol. Cell Biol. 2008, 9, 162– 176, DOI: 10.1038/nrm2335Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOgug%253D%253D&md5=c96aabfdfa8e93c9e98386f1a407de85Lipid signalling in diseaseWymann, Matthias P.; Schneiter, RogerNature Reviews Molecular Cell Biology (2008), 9 (2), 162-176CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Signaling lipids such as eicosanoids, phosphoinositides, sphingolipids and fatty acids control important cellular processes, including cell proliferation, apoptosis, metab. and migration. Extracellular signals from cytokines, growth factors and nutrients control the activity of a key set of lipid-modifying enzymes: phospholipases, prostaglandin synthase, 5-lipoxygenase, phosphoinositide 3-kinase, sphingosine kinase and sphingomyelinase. These enzymes and their downstream targets constitute a complex lipid signaling network with multiple nodes of interaction and cross-regulation. Imbalances in this network contribute to the pathogenesis of human disease. Although the function of a particular signaling lipid is traditionally studied in isolation, this review attempts a more integrated overview of the key role of these signaling lipids in inflammation, cancer and metabolic disease, and discusses emerging strategies for therapeutic intervention.
- 29Fruman, D. A.; Rommel, C. PI3K and cancer: lessons, challenges and opportunities. Nat. Rev. Drug Discovery 2014, 13, 140– 156, DOI: 10.1038/nrd4204Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1SnsLc%253D&md5=c4f6b4b9f9fcacd6ef55eb17d3a4f425PI3K and cancer: lessons, challenges and opportunitiesFruman, David A.; Rommel, ChristianNature Reviews Drug Discovery (2014), 13 (2), 140-156CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The central role of phosphoinositide 3-kinase (PI3K) activation in tumor cell biol. has prompted a sizeable effort to target PI3K and/or downstream kinases such as AKT and mammalian target of rapamycin (mTOR) in cancer. However, emerging clin. data show limited single-agent activity of inhibitors targeting PI3K, AKT or mTOR at tolerated doses. One exception is the response to PI3Kδ inhibitors in chronic lymphocytic leukemia, where a combination of cell-intrinsic and -extrinsic activities drive efficacy. Here, we review key challenges and opportunities for the clin. development of inhibitors targeting the PI3K-AKT-mTOR pathway. Through a greater focus on patient selection, increased understanding of immune modulation and strategic application of rational combinations, it should be possible to realize the potential of this promising class of targeted anticancer agents.
- 30Samuels, Y.; Wang, Z.; Bardelli, A.; Silliman, N.; Ptak, J.; Szabo, S.; Yan, H.; Gazdar, A.; Powell, S. M.; Riggins, G. J.; Willson, J. K.; Markowitz, S.; Kinzler, K. W.; Vogelstein, B.; Velculescu, V. E. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004, 304, 554, DOI: 10.1126/science.1096502Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjsVGmsbk%253D&md5=be8d63bf0c909971c3b7a5f475ce8fabBrevia: High frequency of mutations of the PIK3Ca gene in human cancersSamuels, Yardena; Wang, Zhenghe; Bardelli, Alberto; Silliman, Natalie; Ptak, Janine; Szabo, Steve; Yan, Hai; Gazdar, Adi; Powell, Steven M.; Riggins, Gregory J.; Willson, James K. V.; Markowitz, Sanford; Kinzler, Kenneth W.; Vogelstein, Bert; Velculescu, Victor E.Science (Washington, DC, United States) (2004), 304 (5670), 554CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)There is no expanded citation for this reference.
- 31Vanhaesebroeck, B.; Perry, M. W. D.; Brown, J. R.; André, F.; Okkenhaug, K. PI3K inhibitors are finally coming of age. Nat. Rev. Drug Discovery 2021, 20, 741– 769, DOI: 10.1038/s41573-021-00209-1Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlaitrjL&md5=6186f01192ecbd4d16bf820b0c3259c0Phosphoinositide 3-kinase inhibitors are finally coming of ageVanhaesebroeck, Bart; Perry, Matthew W. D.; Brown, Jennifer R.; Andre, Fabrice; Okkenhaug, KlausNature Reviews Drug Discovery (2021), 20 (10), 741-769CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)A review. Overactive phosphoinositide 3-kinase (PI3K) in cancer and immune dysregulation has spurred extensive efforts to develop therapeutic PI3K inhibitors. Although progress has been hampered by issues such as poor drug tolerance and drug resistance, several PI3K inhibitors have now received regulatory approval-the PI3Kα isoform-selective inhibitor alpelisib for the treatment of breast cancer and inhibitors mainly aimed at the leukocyte-enriched PI3Kδ in B cell malignancies. In addn. to targeting cancer cell-intrinsic PI3K activity, emerging evidence highlights the potential of PI3K inhibitors in cancer immunotherapy. This Review summarizes key discoveries that aid the clin. translation of PI3Kα and PI3Kδ inhibitors, highlighting lessons learnt and future opportunities.
- 32Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26 DOI: 10.1186/s12943-019-0954-xGoogle Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cflt1Kluw%253D%253D&md5=4267e4df49544ebe11ab4255e69e5bbeTargeting PI3K in cancer: mechanisms and advances in clinical trialsYang Jing; Nie Ji; Ma Xuelei; Wei Yuquan; Peng Yong; Wei XiaweiMolecular cancer (2019), 18 (1), 26 ISSN:.Phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling is one of the most important intracellular pathways, which can be considered as a master regulator for cancer. Enormous efforts have been dedicated to the development of drugs targeting PI3K signaling, many of which are currently employed in clinical trials evaluation, and it is becoming increasingly clear that PI3K inhibitors are effective in inhibiting tumor progression. PI3K inhibitors are subdivided into dual PI3K/mTOR inhibitors, pan-PI3K inhibitors and isoform-specific inhibitors. In this review, we performed a critical review to summarize the role of the PI3K pathway in tumor development, recent PI3K inhibitors development based on clinical trials, and the mechanisms of resistance to PI3K inhibition.
- 33Massacesi, C.; Di Tomaso, E.; Urban, P.; Germa, C.; Quadt, C.; Trandafir, L.; Aimone, P.; Fretault, N.; Dharan, B.; Tavorath, R.; Hirawat, S. PI3K inhibitors as new cancer therapeutics: implications for clinical trial design. OncoTargets Ther. 2016, 9, 203– 210, DOI: 10.2147/OTT.S89967Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlvFyqsr4%253D&md5=aa0e646fb23513ab825d1d0aed50dd16PI3K inhibitors as new cancer therapeutics: implications for clinical trial designMassacesi, Cristian; Di Tomaso, Emmanuelle; Urban, Patrick; Germa, Caroline; Quadt, Cornelia; Trandafir, Lucia; Aimone, Paola; Fretault, Nathalie; Dharan, Bharani; Tavorath, Ranjana; Hirawat, SamitOncoTargets and Therapy (2016), 9 (), 203-210CODEN: OTNHAZ; ISSN:1178-6930. (Dove Medical Press Ltd.)The PI3K-AKT-mTOR pathway is frequently activated in cancer. PI3K inhibitors, including the pan-PI3K inhibitor buparlisib (BKM120) and the PI3Ka-selective inhibitor alpelisib (BYL719), currently in clin. development by Novartis Oncol., may therefore be effective as anticancer agents. Early clin. studies with PI3K inhibitors have demonstrated preliminary antitumor activity and acceptable safety profiles. However, a no. of unanswered questions regarding PI3K inhibition in cancer remain, including: what is the best approach for different tumor types, and which biomarkers will accurately identify the patient populations most likely to benefit from specific PI3K inhibitors. This review summarizes the strategies being employed by Novartis Oncol. to help maximize the benefits of clin. studies with buparlisib and alpelisib, including stratification according to PI3K pathway activation status, selective enrollment/target enrichment (where patients with PI3K pathway-activated tumors are specifically recruited), nonselective enrollment with mandatory tissue collection, and enrollment of patients who have progressed on previous targeted agents, such as mTOR inhibitors or endocrine therapy. An overview of Novartis-sponsored and Novartis-supported trials that are utilizing these approaches in a range of cancer types, including breast cancer, head and neck squamous cell carcinoma, non-small cell lung carcinoma, lymphoma, and glioblastoma multiforme, is also described.
- 34Mishra, R.; Patel, H.; Alanazi, S.; Kilroy, M. K.; Garrett, J. T. PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects. Int. J. Mol. Sci. 2021, 22, 3464 DOI: 10.3390/ijms22073464Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXht1Gku7bE&md5=b3551463f09f9086323dfac7f9804fc1PI3K inhibitors in cancer: clinical implications and adverse effectsMishra, Rosalin; Patel, Hima; Alanazi, Samar; Kilroy, Mary Kate; Garrett, Joan T.International Journal of Molecular Sciences (2021), 22 (7), 3464CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)A review. The phospatidylinositol-3 kinase (PI3K) pathway is a crucial intracellular signaling pathway which is mutated or amplified in a wide variety of cancers including breast, gastric, ovarian, colorectal, prostate, glioblastoma and endometrial cancers. PI3K signaling plays an important role in cancer cell survival, angiogenesis and metastasis, making it a promising therapeutic target. There are several ongoing and completed clin. trials involving PI3K inhibitors (pan, isoform-specific and dual PI3K/mTOR) with the goal to find efficient PI3K inhibitors that could overcome resistance to current therapies. This review focuses on the current landscape of various PI3K inhibitors either as monotherapy or in combination therapies and the treatment outcomes involved in various phases of clin. trials in different cancer types. There is a discussion of the drug-related toxicities, challenges assocd. with these PI3K inhibitors and the adverse events leading to treatment failure. In addn., novel PI3K drugs that have potential to be translated in the clinic are highlighted.
- 35Fritsch, C.; Huang, A.; Chatenay-Rivauday, C.; Schnell, C.; Reddy, A.; Liu, M.; Kauffmann, A.; Guthy, D.; Erdmann, D.; De Pover, A.; Furet, P.; Gao, H.; Ferretti, S.; Wang, Y.; Trappe, J.; Brachmann, S. M.; Maira, S. M.; Wilson, C.; Boehm, M.; Garcia-Echeverria, C.; Chene, P.; Wiesmann, M.; Cozens, R.; Lehar, J.; Schlegel, R.; Caravatti, G.; Hofmann, F.; Sellers, W. R. Characterization of the novel and specific PI3Kα inhibitor NVP-BYL719 and development of the patient stratification strategy for clinical trials. Mol. Cancer Ther. 2014, 13, 1117– 1129, DOI: 10.1158/1535-7163.Mct-13-0865Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnsFWjsb4%253D&md5=58d9132052dd9d430361021b0f877475Characterization of the Novel and Specific PI3Kα Inhibitor NVP-BYL719 and Development of the Patient Stratification Strategy for Clinical TrialsFritsch, Christine; Huang, Alan; Chatenay-Rivauday, Christian; Schnell, Christian; Reddy, Anupama; Liu, Manway; Kauffmann, Audrey; Guthy, Daniel; Erdmann, Dirk; De Pover, Alain; Furet, Pascal; Gao, Hui; Ferretti, Stephane; Wang, Youzhen; Trappe, Joerg; Brachmann, Saskia M.; Maira, Sauveur-Michel; Wilson, Christopher; Boehm, Markus; Garcia-Echeverria, Carlos; Chene, Patrick; Wiesmann, Marion; Cozens, Robert; Lehar, Joseph; Schlegel, Robert; Caravatti, Giorgio; Hofmann, Francesco; Sellers, William R.Molecular Cancer Therapeutics (2014), 13 (5), 1117-1129CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Somatic PIK3CA mutations are frequently found in solid tumors, raising the hypothesis that selective inhibition of PI3Kα may have robust efficacy in PIK3CA-mutant cancers while sparing patients the side-effects assocd. with broader inhibition of the class I phosphoinositide 3-kinase (PI3K) family. Here, we report the biol. properties of the 2-aminothiazole deriv. NVP-BYL719, a selective inhibitor of PI3Kα and its most common oncogenic mutant forms. The compd. selectivity combined with excellent drug-like properties translates to dose- and time-dependent inhibition of PI3Kα signaling in vivo, resulting in robust therapeutic efficacy and tolerability in PIK3CA-dependent tumors. Novel targeted therapeutics such as NVP-BYL719, designed to modulate aberrant functions elicited by cancer-specific genetic alterations upon which the disease depends, require well-defined patient stratification strategies in order to maximize their therapeutic impact and benefit for the patients. Here, we also describe the application of the Cancer Cell Line Encyclopedia as a preclin. platform to refine the patient stratification strategy for NVP-BYL719 and found that PIK3CA mutation was the foremost pos. predictor of sensitivity while revealing addnl. pos. and neg. assocns. such as PIK3CA amplification and PTEN mutation, resp. These patient selection determinants are being assayed in the ongoing NVP-BYL719 clin. trials. Mol Cancer Ther; 13(5); 1117-29. ©2014 AACR.
- 36Markham, A. Alpelisib: First Global Approval. Drugs 2019, 79, 1249– 1253, DOI: 10.1007/s40265-019-01161-6Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlCiur%252FN&md5=4d9e9d1e3742a9bce160ca2c7987ce6cAlpelisib: First Global ApprovalMarkham, AnthonyDrugs (2019), 79 (11), 1249-1253CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. Alpelisib (Piqray)-an orally available phosphatidylinositol 3-kinase (PI3K) inhibitor with specific activity against PI3K alpha (PI3Kα)-is being developed by Novartis for the treatment of breast cancer. Alpelisib has demonstrated efficacy in combination with fulvestrant as treatment for hormone receptor (HR)-pos., human epidermal growth factor receptor-2 (HER2)-neg. breast cancer in patients with a PIK3CA mutation and was recently approved for this indication in the USA. This article summarizes the milestones in the development of alpelisib leading to this first approval.
- 37Narayan, P.; Prowell, T. M.; Gao, J. J.; Fernandes, L. L.; Li, E.; Jiang, X.; Qiu, J.; Fan, J.; Song, P.; Yu, J.; Zhang, X.; King-Kallimanis, B. L.; Chen, W.; Ricks, T. K.; Gong, Y.; Wang, X.; Windsor, K.; Rhieu, S. Y.; Geiser, G.; Banerjee, A.; Chen, X.; Reyes Turcu, F.; Chatterjee, D. K.; Pathak, A.; Seidman, J.; Ghosh, S.; Philip, R.; Goldberg, K. B.; Kluetz, P. G.; Tang, S.; Amiri-Kordestani, L.; Theoret, M. R.; Pazdur, R.; Beaver, J. A. FDA Approval Summary: Alpelisib Plus Fulvestrant for Patients with HR-positive, HER2-negative, PIK3CA-mutated, Advanced or Metastatic Breast Cancer. Clin. Cancer Res. 2021, 27, 1842– 1849, DOI: 10.1158/1078-0432.Ccr-20-3652Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFGjtL%252FO&md5=3220d285c0f3c135cf229b67554295d7FDA approval summary: alpelisib plus fulvestrant for patients with HR-positive, HER2-negative, PIK3CA-mutated, advanced or metastatic breast cancerNarayan, Preeti; Prowell, Tatiana M.; Gao, Jennifer J.; Fernandes, Laura L.; Li, Emily; Jiang, Xiling; Qiu, Junshan; Fan, Jianghong; Song, Pengfei; Yu, Jingyu; Zhang, Xinyuan; King-Kallimanis, Bellinda L.; Chen, Wei; Ricks, Tiffany K.; Gong, Yutao; Wang, Xing; Windsor, Katherine; Rhieu, Steve Y.; Geiser, Gerlie; Banerjee, Anamitro; Chen, Xiaohong; Turcu, Francisca Reyes; Chatterjee, Deb K.; Pathak, Anand; Seidman, Jeffrey; Ghosh, Soma; Philip, Reena; Goldberg, Kirsten B.; Kluetz, Paul G.; Tang, Shenghui; Amiri-Kordestani, Laleh; Theoret, Marc R.; Pazdur, Richard; Beaver, Julia A.Clinical Cancer Research (2021), 27 (7), 1842-1849CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)On May 24, 2019, the FDA granted regular approval to alpelisib in combination with fulvestrant for postmenopausal women, and men, with hormone receptor (HR)-pos., HER2-neg., phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA)-mutated, advanced or metastatic breast cancer as detected by an FDA-approved test following progression on or after an endocrine-based regimen. Approval was based on the SOLAR-1 study, a randomized, double-blind, placebo-controlled trial of alpelisib plus fulvestrant vs. placebo plus fulvestrant. The primary endpoint was investigator-assessed progression-free survival (PFS) per RECIST v1.1 in the cohort of trial participants whose tumors had a PIK3CA mutation. The estd. median PFS by investigator assessment in the alpelisib plus fulvestrant arm was 11 mo [95% confidence interval (CI), 7.5-14.5] compared with 5.7 mo (95% CI, 3.7-7.4) in the placebo plus fulvestrant arm (HR, 0.65; 95% CI, 0.50-0.85; two-sided P = 0.001). The median overall survival was not yet reached for the alpelisib plus fulvestrant arm (95% CI, 28.1-NE) and was 26.9 mo (95% CI, 21.9-NE) for the fulvestrant control arm. No PFS benefit was obsd. in trial participants whose tumors did not have a PIK3CA mutation (HR, 0.85; 95% CI, 0.58-1.25). The most common adverse reactions, including lab. abnormalities, on the alpelisib plus fulvestrant arm were increased glucose, increased creatinine, diarrhea, rash, decreased lymphocyte count, increased gamma glutamyl transferase, nausea, increased alanine aminotransferase, fatigue, decreased Hb, increased lipase, decreased appetite, stomatitis, vomiting, decreased wt., decreased calcium, decreased glucose, prolonged activated partial thromboplastin time, and alopecia.
- 38André, F.; Ciruelos, E. M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I. A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y. S.; Inoue, K.; Pápai, Z.; Takahashi, M.; Ghaznawi, F.; Mills, D.; Kaper, M.; Miller, M.; Conte, P. F.; Iwata, H.; Rugo, H. S. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1. Ann. Oncol. 2021, 32, 208– 217, DOI: 10.1016/j.annonc.2020.11.011Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvVKrtbc%253D&md5=11ec0f2b91cad4b709edfb314de7e577Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1Andre, F.; Ciruelos, E. M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I. A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y.-S.; Inoue, K.; Papai, Z.; Takahashi, M.; Ghaznawi, F.; Mills, D.; Kaper, M.; Miller, M.; Conte, P. F.; Iwata, H.; Rugo, H. S.Annals of Oncology (2021), 32 (2), 208-217CODEN: AONNAY; ISSN:1569-8041. (Elsevier Ltd.)Activation of the phosphatidylinositol-3-kinase (PI3K) pathway via PIK3CA mutations occurs in 28%-46% of hormone receptor-pos. (HR+), human epidermal growth factor receptor-2-neg. (HER2-) advanced breast cancers (ABCs) and is assocd. with poor prognosis. The SOLAR-1 trial showed that the addn. of alpelisib to fulvestrant treatment provided statistically significant and clin. meaningful progression-free survival (PFS) benefit in PIK3CA-mutated, HR+, HER2- ABC. Men and postmenopausal women with HR+, HER2- ABC whose disease progressed on or after aromatase inhibitor (AI) were randomized 1 : 1 to receive alpelisib (300 mg/day) plus fulvestrant (500 mg every 28 days and once on day 15) or placebo plus fulvestrant. Overall survival (OS) in the PIK3CA-mutant cohort was evaluated by Kaplan-Meier methodol. and a one-sided stratified log-rank test was carried out with an O'Brien-Fleming efficacy boundary of P ≤ 0.0161. In the PIK3CA-mutated cohort (n = 341), median OS [95% confidence interval (CI)] was 39.3 mo (34.1-44.9) for alpelisib-fulvestrant and 31.4 mo (26.8-41.3) for placebo-fulvestrant [hazard ratio (HR) = 0.86 (95% CI, 0.64-1.15; P = 0.15)]. OS results did not cross the prespecified efficacy boundary. Median OS (95% CI) in patients with lung and/or liver metastases was 37.2 mo (28.7-43.6) and 22.8 mo (19.0-26.8) in the alpelisib-fulvestrant and placebo-fulvestrant arms, resp. [HR = 0.68 (0.46-1.00)]. Median times to chemotherapy (95% CI) for the alpelisib-fulvestrant and placebo-fulvestrant arms were 23.3 mo (15.2-28.4) and 14.8 mo (10.5-22.6), resp. [HR = 0.72 (0.54-0.95)]. No new safety signals were obsd. with longer follow-up. Although the anal. did not cross the prespecified boundary for statistical significance, there was a 7.9-mo numeric improvement in median OS when alpelisib was added to fulvestrant treatment of patients with PIK3CA-mutated, HR+, HER2- ABC. Overall, these results further support the statistically significant prolongation of PFS obsd. with alpelisib plus fulvestrant in this population, which has a poor prognosis due to a PIK3CA mutation.
- 39Turner, S.; Chia, S.; Kanakamedala, H.; Hsu, W. C.; Park, J.; Chandiwana, D.; Ridolfi, A.; Yu, C. L.; Zarate, J. P.; Rugo, H. S. Effectiveness of Alpelisib + Fulvestrant Compared with Real-World Standard Treatment Among Patients with HR+, HER2-, PIK3CA-Mutated Breast Cancer. Oncologist 2021, 26, e1133– e1142, DOI: 10.1002/onco.13804Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1antb7P&md5=14e986b4e02d772d01cc9b6762ffa8feEffectiveness of Alpelisib + Fulvestrant Compared with Real-World Standard Treatment Among Patients with HR +, HER2 -, PIK3CA-Mutated Breast CancerTurner, Stuart; Chia, Stephen; Kanakamedala, Hemanth; Hsu, Wei-Chun; Park, Jinhee; Chandiwana, David; Ridolfi, Antonia; Yu, Chu-Ling; Zarate, Juan Pablo; Rugo, Hope S.Oncologist (2021), 26 (7), e1133-e1142CODEN: OCOLF6; ISSN:1549-490X. (AlphaMed Press)The BYLieve trial (NCT03056755) confirmed efficacy and safety of alpelisib with fulvestrant for hormone receptor-pos. (HR+), human epidermal growth factor receptor-2-neg. (HER2-), PIK3CA-mutated advanced breast cancer (ABC), after cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) with an aromatase inhibitor (AI) as immediate prior therapy. Further analyses were performed to compare efficacy from BYLieve with effectiveness of std. treatment in the real-world setting. Patients who progressed on a CDK4/6i plus AI and were treated with alpelisib with fulvestrant in BYLieve were matched with a real-world patient cohort who received std.-of-care from a deidentified clinico-genomics database (CGDB). Primary and secondary endpoints were to compare progression-free survival (PFS), estd. by the Kaplan-Meier method, and the proportion of patients remaining progression-free at 6 mo, resp., between the two cohorts. A total of 855 patients with PIK3CA-mutant disease who had prior CDK4/6i plus hormone therapy were selected from the CGDB; further matching to 120 patients from BYLieve selected 95 patients without exposure to HER2-targeting agents, clin. study drug, or alpelisib. In unadjusted and postmatching results, primary and secondary endpoints favored treatment with alpelisib with fulvestrant in BYLieve more than std. treatments in the real-world cohort. Postadjustment, median PFS for patients treated with alpelisib in BYLieve was 7.3 vs. 3.7 mo in the real-world cohort, and 6-mo PFS was 54.6% vs. 40.1%, resp. Matched/weighted anal. comparing BYLieve with the real-world setting further supports the clin. benefit of alpelisib with fulvestrant for treatment of HR+, HER2-, PIK3CA-mutant ABC after CDK4/6i treatment. Approx. 40% of patients with hormone receptor-pos. (HR+), human epidermal growth factor receptor-2-neg. (HER2-) advanced breast cancer (ABC) have PIK3CA-mutated tumors, which have been assocd. with endocrine therapy resistance. Alpelisib, an α-selective phosphatidylinositol-3-kinase inhibitor, demonstrated significantly improved progression-free survival in SOLAR-1 and demonstrated clin. efficacy in BYLieve when combined with fulvestrant. Data are limited in comparing the efficacy of alpelisib combined with fulvestrant with effectiveness of std. therapy after CDK4/6i treatment. Using real-world data, this is the first anal. comparing alpelisib combined with fulvestrant with std. treatments for HR+, HER2-, PIK3CA-mutant ABC in the post-CDK4/6i setting.
- 40Verret, B.; Cortes, J.; Bachelot, T.; Andre, F.; Arnedos, M. Efficacy of PI3K inhibitors in advanced breast cancer. Ann. Oncol. 2019, 30, x12– x20, DOI: 10.1093/annonc/mdz381Google ScholarThere is no corresponding record for this reference.
- 41Venot, Q.; Blanc, T.; Rabia, S. H.; Berteloot, L.; Ladraa, S.; Duong, J. P.; Blanc, E.; Johnson, S. C.; Hoguin, C.; Boccara, O.; Sarnacki, S.; Boddaert, N.; Pannier, S.; Martinez, F.; Magassa, S.; Yamaguchi, J.; Knebelmann, B.; Merville, P.; Grenier, N.; Joly, D.; Cormier-Daire, V.; Michot, C.; Bole-Feysot, C.; Picard, A.; Soupre, V.; Lyonnet, S.; Sadoine, J.; Slimani, L.; Chaussain, C.; Laroche-Raynaud, C.; Guibaud, L.; Broissand, C.; Amiel, J.; Legendre, C.; Terzi, F.; Canaud, G. Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature 2018, 558, 540– 546, DOI: 10.1038/s41586-018-0217-9Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFeqtbjO&md5=ce1b6f4dd6c5b0f31d4332e5fec21f42Targeted therapy in patients with PIK3CA-related overgrowth syndromeVenot, Quitterie; Blanc, Thomas; Rabia, Smail Hadj; Berteloot, Laureline; Ladraa, Sophia; Duong, Jean-Paul; Blanc, Estelle; Johnson, Simon C.; Hoguin, Clement; Boccara, Olivia; Sarnacki, Sabine; Boddaert, Nathalie; Pannier, Stephanie; Martinez, Frank; Magassa, Sato; Yamaguchi, Junna; Knebelmann, Bertrand; Merville, Pierre; Grenier, Nicolas; Joly, Dominique; Cormier-Daire, Valerie; Michot, Caroline; Bole-Feysot, Christine; Picard, Arnaud; Soupre, Veronique; Lyonnet, Stanislas; Sadoine, Jeremy; Slimani, Lotfi; Chaussain, Catherine; Laroche-Raynaud, Cecile; Guibaud, Laurent; Broissand, Christine; Amiel, Jeanne; Legendre, Christophe; Terzi, Fabiola; Canaud, GuillaumeNature (London, United Kingdom) (2018), 558 (7711), 540-546CODEN: NATUAS; ISSN:0028-0836. (Nature Research)CLOVES syndrome (congenital lipomatous overgrowth, vascular malformations, epidermal naevi, scoliosis/skeletal and spinal syndrome) is a genetic disorder that results from somatic, mosaic gain-of-function mutations of the PIK3CA gene, and belongs to the spectrum of PIK3CA-related overgrowth syndromes (PROS). This rare condition has no specific treatment and a poor survival rate. Here, we describe a postnatal mouse model of PROS/CLOVES that partially recapitulates the human disease, and demonstrate the efficacy of BYL719, an inhibitor of PIK3CA, in preventing and improving organ dysfunction. On the basis of these results, we used BYL719 to treat nineteen patients with PROS. The drug improved the disease symptoms in all patients. Previously intractable vascular tumors became smaller, congestive heart failure was improved, hemihypertrophy was reduced, and scoliosis was attenuated. The treatment was not assocd. with any substantial side effects. In conclusion, this study provides the first direct evidence supporting PIK3CA inhibition as a promising therapeutic strategy in patients with PROS.
- 42Bohnacker, T.; Prota, A. E.; Beaufils, F.; Burke, J. E.; Melone, A.; Inglis, A. J.; Rageot, D.; Sele, A. M.; Cmiljanovic, V.; Cmiljanovic, N.; Bargsten, K.; Aher, A.; Akhmanova, A.; Diaz, J. F.; Fabbro, D.; Zvelebil, M.; Williams, R. L.; Steinmetz, M. O.; Wymann, M. P. Deconvolution of Buparlisib’s mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nat. Commun. 2017, 8, 14683 DOI: 10.1038/ncomms14683Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czksV2ruw%253D%253D&md5=97d27a7d83bf6ab6094a587565f82bf6Deconvolution of Buparlisib's mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic interventionBohnacker Thomas; Beaufils Florent; Melone Anna; Rageot Denise; Sele Alexander M; Cmiljanovic Vladimir; Cmiljanovic Natasa; Wymann Matthias P; Prota Andrea E; Bargsten Katja; Steinmetz Michel O; Burke John E; Inglis Alison J; Williams Roger L; Aher Amol; Akhmanova Anna; Diaz J Fernando; Fabbro Doriano; Zvelebil MarketaNature communications (2017), 8 (), 14683 ISSN:.BKM120 (Buparlisib) is one of the most advanced phosphoinositide 3-kinase (PI3K) inhibitors for the treatment of cancer, but it interferes as an off-target effect with microtubule polymerization. Here, we developed two chemical derivatives that differ from BKM120 by only one atom. We show that these minute changes separate the dual activity of BKM120 into discrete PI3K and tubulin inhibitors. Analysis of the compounds cellular growth arrest phenotypes and microtubule dynamics suggest that the antiproliferative activity of BKM120 is mainly due to microtubule-dependent cytotoxicity rather than through inhibition of PI3K. Crystal structures of BKM120 and derivatives in complex with tubulin and PI3K provide insights into the selective mode of action of this class of drugs. Our results raise concerns over BKM120's generally accepted mode of action, and provide a unique mechanistic basis for next-generation PI3K inhibitors with improved safety profiles and flexibility for use in combination therapies.
- 43Beaufils, F.; Cmiljanovic, N.; Cmiljanovic, V.; Bohnacker, T.; Melone, A.; Marone, R.; Jackson, E.; Zhang, X.; Sele, A.; Borsari, C.; Mestan, J.; Hebeisen, P.; Hillmann, P.; Giese, B.; Zvelebil, M.; Fabbro, D.; Williams, R. L.; Rageot, D.; Wymann, M. P. 5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a potent, brain-penetrant, orally bioavailable, pan-class I PI3K/mTOR inhibitor as clinical candidate in oncology. J. Med. Chem. 2017, 60, 7524– 7538, DOI: 10.1021/acs.jmedchem.7b00930Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlKhurrP&md5=54d4db75f6a8bda3f12eaf1d196352fd5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a Potent, Brain-Penetrant, Orally Bioavailable, Pan-Class I PI3K/mTOR Inhibitor as Clinical Candidate in OncologyBeaufils, Florent; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Bohnacker, Thomas; Melone, Anna; Marone, Romina; Jackson, Eileen; Zhang, Xuxiao; Sele, Alexander; Borsari, Chiara; Mestan, Jurgen; Hebeisen, Paul; Hillmann, Petra; Giese, Bernd; Zvelebil, Marketa; Fabbro, Doriano; Williams, Roger L.; Rageot, Denise; Wymann, Matthias P.Journal of Medicinal Chemistry (2017), 60 (17), 7524-7538CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Phosphoinositide 3-kinase (PI3K) is deregulated in a wide variety of human tumors and triggers activation of protein kinase B (PKB/Akt) and mammalian target of rapamycin (mTOR). Here we describe the preclin. characterization of compd. 1 (PQR309, bimiralisib), a potent 4,6-dimorpholino-1,3,5-triazine-based pan-class I PI3K inhibitor, which targets mTOR kinase in a balanced fashion at higher concns. No off-target interactions were detected for 1 in a wide panel of protein kinase, enzyme, and receptor ligand assays. Moreover, 1 did not bind tubulin, which was obsd. for the structurally related 4 (BKM120, buparlisib). Compd. 1 is orally available, crosses the blood-brain barrier, and displayed favorable pharmacokinetic parameters in mice, rats, and dogs. Compd. 1 demonstrated efficiency in inhibiting proliferation in tumor cell lines and a rat xenograft model. This, together with the compd.'s safety profile, identifies 1 as a clin. candidate with a broad application range in oncol., including treatment of brain tumors or CNS metastasis. Compd. 1 is currently in phase II clin. trials for advanced solid tumors and refractory lymphoma.
- 44Tarantelli, C.; Gaudio, E.; Arribas, A. J.; Kwee, I.; Hillmann, P.; Rinaldi, A.; Cascione, L.; Spriano, F.; Bernasconi, E.; Guidetti, F.; Carrassa, L.; Pittau, R. B.; Beaufils, F.; Ritschard, R.; Rageot, D.; Sele, A.; Dossena, B.; Rossi, F. M.; Zucchetto, A.; Taborelli, M.; Gattei, V.; Rossi, D.; Stathis, A.; Stussi, G.; Broggini, M.; Wymann, M. P.; Wicki, A.; Zucca, E.; Cmiljanovic, V.; Fabbro, D.; Bertoni, F. PQR309 is a novel dual PI3K/mTOR inhibitor with preclinical antitumor activity in lymphomas as a single agent and in combination therapy. Clin. Cancer Res. 2018, 24, 120– 129, DOI: 10.1158/1078-0432.CCR-17-1041Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVWktw%253D%253D&md5=730305c33b7f669b0eedd0614edd886cPQR309 Is a Novel Dual PI3K/mTOR Inhibitor with Preclinical Antitumor Activity in Lymphomas as a Single Agent and in Combination TherapyTarantelli, Chiara; Gaudio, Eugenio; Arribas, Alberto J.; Kwee, Ivo; Hillmann, Petra; Rinaldi, Andrea; Cascione, Luciano; Spriano, Filippo; Bernasconi, Elena; Guidetti, Francesca; Carrassa, Laura; Pittau, Roberta Bordone; Beaufils, Florent; Ritschard, Reto; Rageot, Denise; Sele, Alexander; Dossena, Barbara; Rossi, Francesca Maria; Zucchetto, Antonella; Taborelli, Monica; Gattei, Valter; Rossi, Davide; Stathis, Anastasios; Stussi, Georg; Broggini, Massimo; Wymann, Matthias P.; Wicki, Andreas; Zucca, Emanuele; Cmiljanovic, Vladimir; Fabbro, Doriano; Bertoni, FrancescoClinical Cancer Research (2018), 24 (1), 120-129CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Activation of the PI3K/mTOR signaling pathway is recurrent in different lymphoma types, and pharmacol. inhibition of the PI3K/mTOR pathway has shown activity in lymphoma patients. Here, we extensively characterized the in vitro and in vivo activity and the mechanism of action of PQR309 (bimiralisib), a novel oral selective dual PI3K/mTOR inhibitor under clin. evaluation, in preclin. lymphoma models. Exptl. Design: This study included preclin. in vitro activity screening on a large panel of cell lines, both as single agent and in combination, validation expts. on in vivo models and primary cells, proteomics and gene-expression profiling, and comparison with other signaling inhibitors. Results: PQR309 had in vitro antilymphoma activity as single agent and in combination with venetoclax, panobinostat, ibrutinib, lenalidomide, ARV-825, marizomib, and rituximab. Sensitivity to PQR309 was assocd. with specific baseline gene-expression features, such as high expression of transcripts coding for the BCR pathway. Combining proteomics and RNA profiling, we identified the different contribution of PQR309-induced protein phosphorylation and gene expression changes to the drug mechanism of action. Gene-expression signatures induced by PQR309 and by other signaling inhibitors largely overlapped. PQR309 showed activity in cells with primary or secondary resistance to idelalisib. Conclusions: On the basis of these results, PQR309 appeared as a novel and promising compd. that is worth developing in the lymphoma setting. Clin Cancer Res; 24(1); 120-9. ©2017 AACR.
- 45Wicki, A.; Brown, N.; Xyrafas, A.; Bize, V.; Hawle, H.; Berardi, S.; Cmiljanovic, N.; Cmiljanovic, V.; Stumm, M.; Dimitrijevic, S.; Herrmann, R.; Pretre, V.; Ritschard, R.; Tzankov, A.; Hess, V.; Childs, A.; Hierro, C.; Rodon, J.; Hess, D.; Joerger, M.; von Moos, R.; Sessa, C.; Kristeleit, R. First-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13). Eur. J. Cancer 2018, 96, 6– 16, DOI: 10.1016/j.ejca.2018.03.012Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1Snu70%253D&md5=acbdfdb59498f5543ba7f02ed9709d0eFirst-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13)Wicki, Andreas; Brown, Nicholas; Xyrafas, Alexandros; Bize, Vincent; Hawle, Hanne; Berardi, Simona; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Stumm, Michael; Dimitrijevic, Sasa; Herrmann, Richard; Pretre, Vincent; Ritschard, Reto; Tzankov, Alexandar; Hess, Viviane; Childs, Alexa; Hierro, Cinta; Rodon, Jordi; Hess, Dagmar; Joerger, Markus; von Moos, Roger; Sessa, Cristiana; Kristeleit, RebeccaEuropean Journal of Cancer (2018), 96 (), 6-16CODEN: EJCAEL; ISSN:0959-8049. (Elsevier Ltd.)PQR309 is an orally bioavailable, balanced pan-phosphatidylinositol-3-kinase (PI3K), mammalian target of rapamycin (mTOR) C1 and mTORC2 inhibitor. This is an accelerated titrn., 3 D 3 dose-escalation, open-label phase Itrial of continuous once-daily (OD) PQR309 administration to evaluate the safety, pharmacokinetics (PK) and pharmacodynamics in patients with advanced solid tumors. Primary objectives were to det. the max. tolerated dose (MTD) and recommended phase 2 dose (RP2D).Twenty-eight patients were included in six dosing cohorts and treated at a daily PQR309 dose ranging from 10 to 150 mg. Common adverse events (AEs; ≥30% patients) included fatigue, hyperglycemia, nausea, diarrhea, constipation, rash, anorexia and vomiting. Grade (G) 3 or 4 drug-related AEs were seen in 13 (46%) and three (11%) patients, resp. Dose-limiting toxicity (DLT) was obsd. in two patients at 100 mg OD (>14-d interruption in PQR309 due to G3 rash, G2 hyperbilirubinemia, G4 suicide attempt; dose redn. due to G3 fatigue, G2 diarrhoea, G4 transaminitis) and one patient at 80 mg (G3 hyperglycemia >7 d). PK shows fast absorption (Tmax 1-2 h) and dose proportionality for Cmax and area under the curve. A partial response in a patient with metastatic thymus cancer, 24% disease vol. redn. in a patient with sinonasal cancer and stable disease for more than 16 wk in a patient with clear cell Bartholin's gland cancer were obsd.The MTD and RP2D of PQR309 is 80 mg of orally OD. PK is dose-proportional. PD shows PI3K pathway phosphoprotein downregulation in paired tumor biopsies. Clin. activity was obsd. in patients with and without PI3K pathway dysregulation.
- 46Borsari, C.; Rageot, D.; Beaufils, F.; Bohnacker, T.; Keles, E.; Buslov, I.; Melone, A.; Sele, A. M.; Hebeisen, P.; Fabbro, D.; Hillmann, P.; Wymann, M. P. Preclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl-Pyrimidine Moiety. ACS Med. Chem. Lett. 2019, 10, 1473– 1479, DOI: 10.1021/acsmedchemlett.9b00333Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslSqtrzP&md5=3423dce039aa5b68a178ecea264e6f1fPreclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl-Pyrimidine MoietyBorsari, Chiara; Rageot, Denise; Beaufils, Florent; Bohnacker, Thomas; Keles, Erhan; Buslov, Ivan; Melone, Anna; Sele, Alexander M.; Hebeisen, Paul; Fabbro, Doriano; Hillmann, Petra; Wymann, Matthias P.ACS Medicinal Chemistry Letters (2019), 10 (10), 1473-1479CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is a crit. regulator of cell growth and is frequently hyperactivated in cancer. Therefore, PI3K inhibitors represent a valuable asset in cancer therapy. Herein we have developed a novel anticancer agent, the potent pan-PI3K inhibitor PQR514 (4), which is a follow-up compd. for the phase-II clin. compd. PQR309 (1). Compd. 4 has an improved potency both in vitro and in cellular assays with respect to its predecessor compds. It shows superiority in the suppression of cancer cell proliferation and demonstrates significant antitumor activity in an OVCAR-3 xenograft model at concns. approx. eight times lower than PQR309 (1). The favorable pharmacokinetic profile and a minimal brain penetration promote PQR514 (4) as an optimized candidate for the treatment of systemic tumors.
- 47Jackson, P. A.; Widen, J. C.; Harki, D. A.; Brummond, K. M. Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition Reactions. J. Med. Chem. 2017, 60, 839– 885, DOI: 10.1021/acs.jmedchem.6b00788Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitV2rtbnM&md5=36d3be5bf688c4e4a8cf4bcf42073009Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition ReactionsJackson, Paul A.; Widen, John C.; Harki, Daniel A.; Brummond, Kay M.Journal of Medicinal Chemistry (2017), 60 (3), 839-885CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Although Michael acceptors display a potent and broad spectrum of bioactivity, they have largely been ignored in drug discovery because of their presumed indiscriminate reactivity. As such, a dearth of information exists relevant to the thiol reactivity of natural products and their analogs possessing this moiety. In the midst of recently approved acrylamide-contg. drugs, it is clear that a good understanding of the hetero-Michael addn. reaction and the relative reactivities of biol. thiols with Michael acceptors under physiol. conditions is needed for the design and use of these compds. as biol. tools and potential therapeutics. This Perspective provides information that will contribute to this understanding, such as kinetics of thiol addn. reactions, bioactivities, as well as steric and electronic factors that influence the electrophilicity and reversibility of Michael acceptors. This Perspective is focused on α,β-unsatd. carbonyls given their preponderance in bioactive natural products.
- 48Flanagan, M. E.; Abramite, J. A.; Anderson, D. P.; Aulabaugh, A.; Dahal, U. P.; Gilbert, A. M.; Li, C.; Montgomery, J.; Oppenheimer, S. R.; Ryder, T.; Schuff, B. P.; Uccello, D. P.; Walker, G. S.; Wu, Y.; Brown, M. F.; Chen, J. M.; Hayward, M. M.; Noe, M. C.; Obach, R. S.; Philippe, L.; Shanmugasundaram, V.; Shapiro, M. J.; Starr, J.; Stroh, J.; Che, Y. Chemical and computational methods for the characterization of covalent reactive groups for the prospective design of irreversible inhibitors. J. Med. Chem. 2014, 57, 10072– 10079, DOI: 10.1021/jm501412aGoogle Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVyjtLbM&md5=3ccf59ab7a494655185e5eb5becf8c48Chemical and Computational Methods for the Characterization of Covalent Reactive Groups for the Prospective Design of Irreversible InhibitorsFlanagan, Mark E.; Abramite, Joseph A.; Anderson, Dennis P.; Aulabaugh, Ann; Dahal, Upendra P.; Gilbert, Adam M.; Li, Chao; Montgomery, Justin; Oppenheimer, Stacey R.; Ryder, Tim; Schuff, Brandon P.; Uccello, Daniel P.; Walker, Gregory S.; Wu, Yan; Brown, Matthew F.; Chen, Jinshan M.; Hayward, Matthew M.; Noe, Mark C.; Obach, R. Scott; Philippe, Laurence; Shanmugasundaram, Veerabahu; Shapiro, Michael J.; Starr, Jeremy; Stroh, Justin; Che, YeJournal of Medicinal Chemistry (2014), 57 (23), 10072-10079CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Interest in drugs that covalently modify their target is driven by the desire for enhanced efficacy that can result from the silencing of enzymic activity until protein resynthesis can occur, along with the potential for increased selectivity by targeting uniquely positioned nucleophilic residues in the protein. However, covalent approaches carry addnl. risk for toxicities or hypersensitivity reactions that can result from covalent modification of unintended targets. Here we describe methods for measuring the reactivity of covalent reactive groups (CRGs) with a biol. relevant nucleophile, glutathione (GSH), along with kinetic data for a broad array of electrophiles. We also describe a computational method for predicting electrophilic reactivity, which taken together can be applied to the prospective design of thiol-reactive covalent inhibitors.
- 49Martin, J. S.; MacKenzie, C. J.; Fletcher, D.; Gilbert, I. H. Characterising covalent warhead reactivity. Bioorg. Med. Chem. 2019, 27, 2066– 2074, DOI: 10.1016/j.bmc.2019.04.002Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntVGgsbo%253D&md5=487334d8d0a8b03d0a25d6d87c1b4329Characterising covalent warhead reactivityMartin, James S.; MacKenzie, Claire J.; Fletcher, Daniel; Gilbert, Ian H.Bioorganic & Medicinal Chemistry (2019), 27 (10), 2066-2074CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Many drugs currently used are covalent inhibitors and irreversibly inhibit their targets. Most of these were discovered through serendipity. Covalent inhibitions can have many advantages from a pharmacokinetic perspective. However, until recently most organizations have shied away from covalent compd. design due to fears of non-specific inhibition of off-target proteins leading to toxicity risks. However, there has been a renewed interest in covalent modifiers as potential drugs, as it possible to get highly selective compds. It is therefore important to know how reactive a warhead is and to be able to select the least reactive warhead possible to avoid toxicity. A robust NMR based assay was developed and used to measure the reactivity of a variety of covalent warheads against serine and cysteine - the two most common targets for covalent drugs. A selection of these warheads also had their reactivity measured against threonine, tyrosine, lysine, histidine and arginine to better understand our ability to target non-traditional residues. The reactivity was also measured at various pHs to assess what effect the environment in the active site would have on these reactions. The reactivity of a covalent modifier was found to be very dependent on the amino acid residue.
- 50Lonsdale, R.; Burgess, J.; Colclough, N.; Davies, N. L.; Lenz, E. M.; Orton, A. L.; Ward, R. A. Expanding the Armory: Predicting and Tuning Covalent Warhead Reactivity. J. Chem. Inf. Model. 2017, 57, 3124– 3137, DOI: 10.1021/acs.jcim.7b00553Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsl2gsLjF&md5=722e83699c58dee1009b87c8c31bdeccExpanding the Armory: Predicting and Tuning Covalent Warhead ReactivityLonsdale, Richard; Burgess, Jonathan; Colclough, Nicola; Davies, Nichola L.; Lenz, Eva M.; Orton, Alexandra L.; Ward, Richard A.Journal of Chemical Information and Modeling (2017), 57 (12), 3124-3137CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Targeted covalent inhibition is an established approach for increasing the potency and selectivity of potential drug candidates, as well as identifying potent and selective tool compds. for target validation studies. It is evident that identification of reversible recognition elements is essential for selective covalent inhibition, but this must also be achieved with the appropriate level of inherent reactivity of the reactive functionality (or "warhead"). Structural changes that increase or decrease warhead reactivity, guided by methods to predict the effect of those changes, have the potential to tune warhead reactivity and negate issues related to potency and/or toxicity. The half-life to adduct formation with glutathione (GSH t1/2) is a useful assay for measuring the reactivity of cysteine-targeting covalent warheads but is limited to synthesized mols. In this manuscript the authors assess the ability of several exptl. and computational approaches to predict GSH t1/2 for a range of cysteine targeting warheads, including a novel method based on pKa. Furthermore, matched mol. pairs anal. has been performed against the internal compd. collection, revealing structure-activity relationships between a selection of different covalent warheads. These observations and methods of prediction will be valuable in the design of new covalent inhibitors with desired levels of reactivity.
- 51Ma, S.; Devi-Kesavan, L. S.; Gao, J. Molecular dynamics simulations of the catalytic pathway of a cysteine protease: a combined QM/MM study of human cathepsin K. J. Am. Chem. Soc. 2007, 129, 13633– 13645, DOI: 10.1021/ja074222+Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtFKhtbjK&md5=051a894ff0cf93fcd72bb572088284a2Molecular Dynamics Simulations of the Catalytic Pathway of a Cysteine Protease: A Combined QM/MM Study of Human Cathepsin KMa, Shuhua; Devi-Kesavan, Lakshmi S.; Gao, JialiJournal of the American Chemical Society (2007), 129 (44), 13633-13645CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Mol. dynamics simulations using a combined QM/MM potential have been performed to study the catalytic mechanism of human cathepsin K, a member of the papain family of cysteine proteases. We have detd. the two-dimensional free energy surfaces of both acylation and deacylation steps to characterize the reaction mechanism. These free energy profiles show that the acylation step is rate limiting with a barrier height of 19.8 kcal/mol in human cathepsin K and of 29.3 kcal/mol in aq. soln. The free energy of activation for the deacylation step is 16.7 kcal/mol in cathepsin K and 17.8 kcal/mol in aq. soln. The redn. of free energy barrier is achieved by stabilization of the oxyanion in the transition state. Interestingly, although the "oxyanion hole" has been formed in the Michaelis complex, the amide units do not donate hydrogen bonds directly to the carbonyl oxygen of the substrate, but they stabilize the thiolate anion nucleophile. Hydrogen-bonding interactions are induced as the substrate amide group approaches the nucleophile, moving more than 2 Å and placing the oxyanion in contact with Gln19 and the backbone amide of Cys25. The hydrolysis of peptide substrate shares a common mechanism both for the catalyzed reaction in human cathepsin K and for the uncatalyzed reaction in water. Overall, the nucleophilic attack by Cys25 thiolate and the proton-transfer reaction from His162 to the amide nitrogen are highly coupled, whereas a tetrahedral intermediate is formed along the nucleophilic reaction pathway.
- 52Cusack, K. P.; Arnold, L. D.; Barberis, C. E.; Chen, H.; Ericsson, A. M.; Gaza-Bulseco, G. S.; Gordon, T. D.; Grinnell, C. M.; Harsch, A.; Pellegrini, M.; Tarcsa, E. A 13C NMR approach to categorizing potential limitations of alpha,beta-unsaturated carbonyl systems in drug-like molecules. Bioorg. Med. Chem. Lett. 2004, 14, 5503– 5507, DOI: 10.1016/j.bmcl.2004.09.007Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXos1eju70%253D&md5=ee93901b0f81e811ff86c86210a8286cA 13C NMR approach to categorizing potential limitations of α,β-unsaturated carbonyl systems in drug-like moleculesCusack, Kevin P.; Arnold, Lee D.; Barberis, Claude E.; Chen, Haipeng; Ericsson, Anna M.; Gaza-Bulseco, Georgeen S.; Gordon, Thomas D.; Grinnell, Christine M.; Harsch, Andreas; Pellegrini, Maria; Tarcsa, EditBioorganic & Medicinal Chemistry Letters (2004), 14 (22), 5503-5507CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Compds. that contain an α,β-unsatd. carbonyl moiety are often flagged as potential Michael acceptors. All α,β-unsatd. carbonyl moieties are not equiv., however, and we sought to better understand this system and its potential implications in drug-like mols. Measurement of the 13C NMR shift of the β-carbon and correlation to in vitro results allowed compds. in our collection to be categorized as potential Michael acceptors, potential substrates for NADPH, or as photoisomerizable.
- 53MacFaul, P. A.; Morley, A. D.; Crawford, J. J. A simple in vitro assay for assessing the reactivity of nitrile containing compounds. Bioorg. Med. Chem. Lett. 2009, 19, 1136– 1138, DOI: 10.1016/j.bmcl.2008.12.105Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXit1WrtLs%253D&md5=d949b5d0e76c6d268f3f57cc4584f0b9A simple in vitro assay for assessing the reactivity of nitrile containing compoundsMacFaul, Philip A.; Morley, Andrew D.; Crawford, James J.Bioorganic & Medicinal Chemistry Letters (2009), 19 (4), 1136-1138CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)A quant. assay involving the reaction of nitriles with glutathione and cysteine has been used as a simple in vitro screen to assess potential toxicity risk of candidate compds. in drug discovery. Studies have indicated that, when benchmarked with selected compds., the reaction of the nitriles with glutathione can provide a useful tool for deciding whether or not to progress compds. in the absence of radiolabelling studies.
- 54Brameld, K. A.; Owens, T. D.; Verner, E.; Venetsanakos, E.; Bradshaw, J. M.; Phan, V. T.; Tam, D.; Leung, K.; Shu, J.; LaStant, J.; Loughhead, D. G.; Ton, T.; Karr, D. E.; Gerritsen, M. E.; Goldstein, D. M.; Funk, J. O. Discovery of the Irreversible Covalent FGFR Inhibitor 8-(3-(4-Acryloylpiperazin-1-yl)propyl)-6-(2,6-dichloro-3,5-dimethoxyphenyl)-2-(methylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (PRN1371) for the Treatment of Solid Tumors. J. Med. Chem. 2017, 60, 6516– 6527, DOI: 10.1021/acs.jmedchem.7b00360Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVykt7fK&md5=987069b645d5e0e36ccda5e5c81520b9Discovery of the Irreversible Covalent FGFR Inhibitor 8-(3-(4-Acryloylpiperazin-1-yl)propyl)-6-(2,6-dichloro-3,5-dimethoxyphenyl)-2-(methylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (PRN1371) for the Treatment of Solid TumorsBrameld, Ken A.; Owens, Timothy D.; Verner, Erik; Venetsanakos, Eleni; Bradshaw, J. Michael; Phan, Vernon T.; Tam, Danny; Leung, Kwan; Shu, Jin; LaStant, Jacob; Loughhead, David G.; Ton, Tony; Karr, Dane E.; Gerritsen, Mary E.; Goldstein, David M.; Funk, Jens OliverJournal of Medicinal Chemistry (2017), 60 (15), 6516-6527CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Aberrant signaling of the FGF/FGFR pathway occurs frequently in cancers and is an oncogenic driver in many solid tumors. Clin. validation of FGFR as a therapeutic target has been demonstrated in bladder, liver, lung, breast, and gastric cancers. Our goal was to develop an irreversible covalent inhibitor of FGFR1-4 for use in oncol. indications. An irreversible covalent binding mechanism imparts many desirable pharmacol. benefits including high potency, selectivity, and prolonged target inhibition. Herein we report the structure-based design, medicinal chem. optimization, and unique ADME assays of our irreversible covalent drug discovery program which culminated in the discovery of compd. 34 (PRN1371), a highly selective and potent FGFR1-4 inhibitor.
- 55Cee, V. J.; Volak, L. P.; Chen, Y.; Bartberger, M. D.; Tegley, C.; Arvedson, T.; McCarter, J.; Tasker, A. S.; Fotsch, C. Systematic Study of the Glutathione (GSH) Reactivity of N-Arylacrylamides: 1. Effects of Aryl Substitution. J. Med. Chem. 2015, 58, 9171– 9178, DOI: 10.1021/acs.jmedchem.5b01018Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFGrsLfN&md5=2f3a816446dd73d5be061ad8d2195e2dSystematic Study of the Glutathione (GSH) Reactivity of N-Arylacrylamides: 1. Effects of Aryl SubstitutionCee, Victor J.; Volak, Laurie P.; Chen, Yuping; Bartberger, Michael D.; Tegley, Chris; Arvedson, Tara; McCarter, John; Tasker, Andrew S.; Fotsch, ChristopherJournal of Medicinal Chemistry (2015), 58 (23), 9171-9178CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Success in the design of targeted covalent inhibitors depends in part on a knowledge of the factors influencing electrophile reactivity. In an effort to further develop an understanding of structure-reactivity relations among N-arylacrylamides, the authors detd. glutathione (GSH) reaction rates for a family of N-arylacrylamides independently substituted at ortho-, meta-, and para-positions with 11 different groups common to inhibitor design. Substituent effects on reaction rates show a linear Hammett correlation for ortho-, meta-, and para-substitution. The authors note a correlation between 1H and 13C NMR chem. shifts of the acrylamide with GSH reaction rates, suggesting that NMR chem. shifts may be a convenient surrogate measure of relative acrylamide reactivity. D. functional theory calcns. reveal a correlation between computed activation parameters and exptl. detd. reaction rates, validating the use of such methodol. for the screening of synthetic candidates in a prospective fashion.
- 56Birkholz, A.; Kopecky, D. J.; Volak, L. P.; Bartberger, M. D.; Chen, Y.; Tegley, C. M.; Arvedson, T.; McCarter, J. D.; Fotsch, C.; Cee, V. J. Systematic Study of the Glutathione Reactivity of N-Phenylacrylamides: 2. Effects of Acrylamide Substitution. J. Med. Chem. 2020, 63, 11602– 11614, DOI: 10.1021/acs.jmedchem.0c00749Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvV2ns7zJ&md5=051ff03cf309131266a52f6f3c55087aSystematic Study of the Glutathione Reactivity of N-Phenylacrylamides: 2. Effects of Acrylamide SubstitutionBirkholz, Adam; Kopecky, David J.; Volak, Laurie P.; Bartberger, Michael D.; Chen, Yuping; Tegley, Christopher M.; Arvedson, Tara; McCarter, John D.; Fotsch, Christopher; Cee, Victor J.Journal of Medicinal Chemistry (2020), 63 (20), 11602-11614CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A comprehensive understanding of structure-reactivity relationships is crit. to the design and optimization of cysteine-targeted covalent inhibitors. Herein, we report glutathione (GSH) reaction rates for N-Ph acrylamides with varied substitutions at the α- and β-positions of the acrylamide moiety. We find that the GSH reaction rates can generally be understood in terms of the electron donating or withdrawing ability of the substituent. When installed at the β-position, aminomethyl substituents with amine pKa's > 7 accelerate, while those with pKa's < 7 slow the rate of GSH addn. at pH 7.4, relative to a hydrogen substituent. Although a computational model was able to only approx. capture exptl. reactivity trends, our calcns. do not support a frequently invoked mechanism of concerted amine/thiol proton transfer and C-S bond formation and instead suggest that protonated aminomethyl functions as an electron-withdrawing group to reduce the barrier for thiolate addn. to the acrylamide.
- 57Zhu, K.; Borrelli, K. W.; Greenwood, J. R.; Day, T.; Abel, R.; Farid, R. S.; Harder, E. Docking covalent inhibitors: a parameter free approach to pose prediction and scoring. J. Chem. Inf. Model. 2014, 54, 1932– 1940, DOI: 10.1021/ci500118sGoogle Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpsVKksLs%253D&md5=3633c29fa5dc29817b178fd10a4c7b15Docking Covalent Inhibitors: A Parameter Free Approach To Pose Prediction and ScoringZhu, Kai; Borrelli, Kenneth W.; Greenwood, Jeremy R.; Day, Tyler; Abel, Robert; Farid, Ramy S.; Harder, EdwardJournal of Chemical Information and Modeling (2014), 54 (7), 1932-1940CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Although many popular docking programs include a facility to account for covalent ligands, large-scale systematic docking validation studies of covalent inhibitors have been sparse. In this paper, we present the development and validation of a novel approach for docking and scoring covalent inhibitors, which consists of conventional noncovalent docking, heuristic formation of the covalent attachment point, and structural refinement of the protein-ligand complex. This approach combines the strengths of the docking program Glide and the protein structure modeling program Prime and does not require any parameter fitting for the study of addnl. covalent reaction types. We first test this method by predicting the native binding geometry of 38 covalently bound complexes. The av. RMSD of the predicted poses is 1.52 Å, and 76% of test set inhibitors have an RMSD of less than 2.0 Å. In addn., the apparent affinity score constructed herein is tested on a virtual screening study and the characterization of the SAR properties of two different series of congeneric compds. with satisfactory success.
- 58Toledo Warshaviak, D.; Golan, G.; Borrelli, K. W.; Zhu, K.; Kalid, O. Structure-based virtual screening approach for discovery of covalently bound ligands. J. Chem. Inf. Model. 2014, 54, 1941– 1950, DOI: 10.1021/ci500175rGoogle Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpslegtbw%253D&md5=0ff8b6e682473abd2e2dadb49d95539aStructure-Based Virtual Screening Approach for Discovery of Covalently Bound LigandsToledo Warshaviak, Dora; Golan, Gali; Borrelli, Kenneth W.; Zhu, Kai; Kalid, OriJournal of Chemical Information and Modeling (2014), 54 (7), 1941-1950CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)We present a fast and effective covalent docking approach suitable for large-scale virtual screening (VS). We applied this method to four targets (HCV NS3 protease, Cathepsin K, EGFR, and XPO1) with known crystal structures and known covalent inhibitors. We implemented a customized "VS mode" of the Schrodinger Covalent Docking algorithm (CovDock), which we refer to as CovDock-VS. Known actives and target-specific sets of decoys were docked to selected X-ray structures, and poses were filtered based on noncovalent protein-ligand interactions known to be important for activity. We were able to retrieve 71%, 72%, and 77% of the known actives for Cathepsin K, HCV NS3 protease, and EGFR within 5% of the decoy library, resp. With the more challenging XPO1 target, where no specific interactions with the protein could be used for postprocessing of the docking results, we were able to retrieve 95% of the actives within 30% of the decoy library and achieved an early enrichment factor (EF1%) of 33. The poses of the known actives bound to existing crystal structures of 4 targets were predicted with an av. RMSD of 1.9 Å. To the best of our knowledge, CovDock-VS is the first fully automated tool for efficient virtual screening of covalent inhibitors. Importantly, CovDock-VS can handle multiple chem. reactions within the same library, only requiring a generic SMARTS-based predefinition of the reaction. CovDock-VS provides a fast and accurate way of differentiating actives from decoys without significantly deteriorating the accuracy of the predicted poses for covalent protein-ligand complexes. Therefore, we propose CovDock-VS as an efficient structure-based virtual screening method for discovery of novel and diverse covalent ligands.
- 59Rageot, D.; Bohnacker, T.; Keles, E.; McPhail, J. A.; Hoffmann, R. M.; Melone, A.; Borsari, C.; Sriramaratnam, R.; Sele, A. M.; Beaufils, F.; Hebeisen, P.; Fabbro, D.; Hillmann, P.; Burke, J. E.; Wymann, M. P. (S)-4-(Difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530), a potent, orally bioavailable, and brain-penetrable dual inhibitor of class I PI3K and mTOR kinase. J. Med. Chem. 2019, 62, 6241– 6261, DOI: 10.1021/acs.jmedchem.9b00525Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFWgtLrM&md5=48e0d98bf33ad5c98a1911b82bcccf03(S)-4-(Difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530), a Potent, Orally Bioavailable, and Brain-Penetrable Dual Inhibitor of Class I PI3K and mTOR KinaseRageot, Denise; Bohnacker, Thomas; Keles, Erhan; McPhail, Jacob A.; Hoffmann, Reece M.; Melone, Anna; Borsari, Chiara; Sriramaratnam, Rohitha; Sele, Alexander M.; Beaufils, Florent; Hebeisen, Paul; Fabbro, Doriano; Hillmann, Petra; Burke, John E.; Wymann, Matthias P.Journal of Medicinal Chemistry (2019), 62 (13), 6241-6261CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is frequently overactivated in cancer, and drives cell growth, proliferation, survival, and metastasis. Here, we report a structure-activity relationship study, which led to the discovery of a drug-like ATP-site PI3K/mTOR kinase inhibitor: (S)-4-(difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530, compd. 6), which qualifies as a clin. candidate due to its potency and specificity for PI3K and mTOR kinases, and its pharmacokinetic properties, including brain penetration. Compd. 6 showed excellent selectivity over a wide panel of kinases and an excellent selectivity against unrelated receptor enzymes and ion channels. Moreover, compd. 6 prevented cell growth in a cancer cell line panel. The preclin. in vivo characterization of compd. 6 in an OVCAR-3 xenograft model demonstrated good oral bioavailability, excellent brain penetration, and efficacy. Initial toxicity studies in rats and dogs qualify 6 for further development as a therapeutic agent in oncol.
- 60Goel, P.; Alam, O.; Naim, M. J.; Nawaz, F.; Iqbal, M.; Alam, M. I. Recent advancement of piperidine moiety in treatment of cancer- A review. Eur. J. Med. Chem. 2018, 157, 480– 502, DOI: 10.1016/j.ejmech.2018.08.017Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFahsrrL&md5=098009e3143fa4e14f98b02bbdc27a15Recent advancement of piperidine moiety in treatment of cancer- A reviewGoel, Pallavi; Alam, Ozair; Naim, Mohd Javed; Nawaz, Farah; Iqbal, Muzaffar; Alam, Md IqbalEuropean Journal of Medicinal Chemistry (2018), 157 (), 480-502CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A review. Piperidine is an important pharmacophore, a privileged scaffold and an excellent heterocyclic system in the field of drug discovery which provides numerous opportunities in studying/exploring this moiety as an anticancer agent by acting on various receptors of utmost importance. Cancer is an uncontrolled division of cells that results in the formation of tumor which have the potential to migrate to other parts of the body (metastasis) finally becoming a major threat to human population. Since piperidine displayed great potential in this area it is being further probed to get novel entities for the treatment of cancer. This review throws light on recent biol. expansions of piperidine along with structure activity relationships to deliver assocn. between various synthesized newer/novel derivs. and receptor sites.
- 61Copeland, R. A. Evaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologists. Methods Biochem. Anal. 2005, 46, 1– 265Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2MnmtlyjsA%253D%253D&md5=4bb6759b5155aa6c2b6a462432afda7fEvaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologistsCopeland Robert AMethods of biochemical analysis (2005), 46 (), 1-265 ISSN:0076-6941.There is no expanded citation for this reference.
- 62Strelow, J. M. A Perspective on the Kinetics of Covalent and Irreversible Inhibition. SLAS Discovery 2017, 22, 3– 20, DOI: 10.1177/1087057116671509Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlSntrbJ&md5=bd404e6b26df20b8761cd38f0f3c3541A perspective on the kinetics of covalent and irreversible inhibitionStrelow, John M.SLAS Discovery (2017), 22 (1), 3-20CODEN: SDLIBT; ISSN:2472-5560. (Sage Publications)The clin. and com. success of covalent drugs has prompted a renewed and more deliberate pursuit of covalent and irreversible mechanisms within drug discovery. A covalent mechanism can produce potent inhibition in a biochem., cellular, or in vivo setting. In many cases, teams choose to focus on the consequences of the covalent event, defined by an IC50 value. In a biochem. assay, the IC50 may simply reflect the target protein concn. in the assay. What has received less attention is the importance of the rate of covalent modification, defined by kinact/KI. The kinact/KI is a rate const. describing the efficiency of covalent bond formation resulting from the potency (KI) of the first reversible binding event and the max. potential rate (kinact) of inactivation. In this perspective, it is proposed that the kinact/KI should be employed as a crit. parameter to identify covalent inhibitors, interpret structure-activity relationships (SARs), translate activity from biochem. assays to the cell, and more accurately define selectivity. It is also proposed that a physiol. relevant kinact/KI and an (unbound) AUC generated from a pharmacokinetic profile reflecting direct exposure of the inhibitor to the target protein are two crit. determinants of in vivo covalent occupancy. A simple equation is presented to define this relationship and improve the interpretation of covalent and irreversible kinetics.
- 63Dale, N. C.; Johnstone, E. K. M.; White, C. W.; Pfleger, K. D. G. NanoBRET: The Bright Future of Proximity-Based Assays. Front. Bioeng. Biotechnol. 2019, 7, 56 DOI: 10.3389/fbioe.2019.00056Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252FktFOgsA%253D%253D&md5=31e05be574d590da21049e92b8e1509aNanoBRET: The Bright Future of Proximity-Based AssaysDale Natasha C; Johnstone Elizabeth K M; White Carl W; Pfleger Kevin D G; Dale Natasha C; Johnstone Elizabeth K M; White Carl W; Pfleger Kevin D G; Dale Natasha C; Johnstone Elizabeth K M; White Carl W; Pfleger Kevin D G; Pfleger Kevin D GFrontiers in bioengineering and biotechnology (2019), 7 (), 56 ISSN:2296-4185.Bioluminescence resonance energy transfer (BRET) is a biophysical technique used to monitor proximity within live cells. BRET exploits the naturally occurring phenomenon of dipole-dipole energy transfer from a donor enzyme (luciferase) to an acceptor fluorophore following enzyme-mediated oxidation of a substrate. This results in production of a quantifiable signal that denotes proximity between proteins and/or molecules tagged with complementary luciferase and fluorophore partners. BRET assays have been used to observe an array of biological functions including ligand binding, intracellular signaling, receptor-receptor proximity, and receptor trafficking, however, BRET assays can theoretically be used to monitor the proximity of any protein or molecule for which appropriate fusion constructs and/or fluorophore conjugates can be produced. Over the years, new luciferases and approaches have been developed that have increased the potential applications for BRET assays. In particular, the development of the small, bright and stable Nanoluciferase (NanoLuc; Nluc) and its use in NanoBRET has vastly broadened the potential applications of BRET assays. These advances have exciting potential to produce new experimental methods to monitor protein-protein interactions (PPIs), protein-ligand interactions, and/or molecular proximity. In addition to NanoBRET, Nluc has also been exploited to produce NanoBiT technology, which further broadens the scope of BRET to monitor biological function when NanoBiT is combined with an acceptor. BRET has proved to be a powerful tool for monitoring proximity and interaction, and these recent advances further strengthen its utility for a range of applications.
- 64Vasta, J. D.; Corona, C. R.; Wilkinson, J.; Zimprich, C. A.; Hartnett, J. R.; Ingold, M. R.; Zimmerman, K.; Machleidt, T.; Kirkland, T. A.; Huwiler, K. G.; Ohana, R. F.; Slater, M.; Otto, P.; Cong, M.; Wells, C. I.; Berger, B. T.; Hanke, T.; Glas, C.; Ding, K.; Drewry, D. H.; Huber, K. V. M.; Willson, T. M.; Knapp, S.; Muller, S.; Meisenheimer, P. L.; Fan, F.; Wood, K. V.; Robers, M. B. Quantitative, Wide-Spectrum Kinase Profiling in Live Cells for Assessing the Effect of Cellular ATP on Target Engagement. Cell Chem. Biol. 2018, 25, 206.e11– 214.e11, DOI: 10.1016/j.chembiol.2017.10.010Google ScholarThere is no corresponding record for this reference.
- 65Ikediobi, O. N.; Davies, H.; Bignell, G.; Edkins, S.; Stevens, C.; O’Meara, S.; Santarius, T.; Avis, T.; Barthorpe, S.; Brackenbury, L.; Buck, G.; Butler, A.; Clements, J.; Cole, J.; Dicks, E.; Forbes, S.; Gray, K.; Halliday, K.; Harrison, R.; Hills, K.; Hinton, J.; Hunter, C.; Jenkinson, A.; Jones, D.; Kosmidou, V.; Lugg, R.; Menzies, A.; Mironenko, T.; Parker, A.; Perry, J.; Raine, K.; Richardson, D.; Shepherd, R.; Small, A.; Smith, R.; Solomon, H.; Stephens, P.; Teague, J.; Tofts, C.; Varian, J.; Webb, T.; West, S.; Widaa, S.; Yates, A.; Reinhold, W.; Weinstein, J. N.; Stratton, M. R.; Futreal, P. A.; Wooster, R. Mutation analysis of 24 known cancer genes in the NCI-60 cell line set. Mol. Cancer Ther. 2006, 5, 2606– 2612, DOI: 10.1158/1535-7163.Mct-06-0433Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1els7jK&md5=f943d16d3e147357815de0ec3c29c30dMutation analysis of 24 known cancer genes in the NCI-60 cell line setIkediobi, Ogechi N.; Davies, Helen; Bignell, Graham; Edkins, Sarah; Stevens, Claire; O'Meara, Sarah; Santarius, Thomas; Avis, Tim; Barthorpe, Syd; Brackenbury, Lisa; Buck, Gemma; Butler, Adam; Clements, Jody; Cole, Jennifer; Dicks, Ed; Forbes, Simon; Gray, Kristian; Halliday, Kelly; Harrison, Rachel; Hills, Katy; Hinton, Jonathan; Hunter, Chris; Jenkinson, Andy; Jones, David; Kosmidou, Vivienne; Lugg, Richard; Menzies, Andrew; Mironenko, Tatiana; Parker, Adrian; Perry, Janet; Raine, Keiran; Richardson, David; Shepherd, Rebecca; Small, Alex; Smith, Raffaella; Solomon, Helen; Stephens, Philip; Teague, Jon; Tofts, Calli; Varian, Jennifer; Webb, Tony; West, Sofie; Widaa, Sara; Yates, Andy; Reinhold, William; Weinstein, John N.; Stratton, Michael R.; Futreal, P. Andrew; Wooster, RichardMolecular Cancer Therapeutics (2006), 5 (11), 2606-2612CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The panel of 60 human cancer cell lines (the NCI-60) assembled by the National Cancer Institute for anticancer drug discovery is a widely used resource. The NCI-60 has been characterized pharmacol. and at the mol. level more extensively than any other set of cell lines. However, no systematic mutation anal. of genes causally implicated in oncogenesis has been reported. This study reports the sequence anal. of 24 known cancer genes in the NCI-60 and an assessment of 4 of the 24 genes for homozygous deletions. One hundred thirty-seven oncogenic mutations were identified in 14 (APC, BRAF, CDKN2, CTNNB1, HRAS, KRAS, NRAS, SMAD4, PIK3CA, PTEN, RB1, STK11, TP53, and VHL) of the 24 genes. All lines have at least one mutation among the cancer genes examd., with most lines (73%) having more than one. Identification of those cancer genes mutated in the NCI-60, in combination with pharmacol. and mol. profiles of the cells, will allow for more informed interpretation of anticancer agent screening and will enhance the use of the NCI-60 cell lines for molecularly targeted screens.
- 66Gall, V. A.; Philips, A. V.; Qiao, N.; Clise-Dwyer, K.; Perakis, A. A.; Zhang, M.; Clifton, G. T.; Sukhumalchandra, P.; Ma, Q.; Reddy, S. M.; Yu, D.; Molldrem, J. J.; Peoples, G. E.; Alatrash, G.; Mittendorf, E. A. Trastuzumab Increases HER2 Uptake and Cross-Presentation by Dendritic Cells. Cancer Res. 2017, 77, 5374– 5383, DOI: 10.1158/0008-5472.Can-16-2774Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFOls7bO&md5=70f3d634f03212ebd5e026a5acdc1079trastuzumab increases her2 uptake and cross-presentation by dendritic cellsGall, Victor A.; Philips, Anne V.; Qiao, Na; Clise-Dwyer, Karen; Perakis, Alexander A.; Zhang, Mao; Clifton, Guy T.; Sukhumalchandra, Pariya; Ma, Qing; Reddy, Sangeetha M.; Yu, Dihua; Molldrem, Jeffrey J.; Peoples, George E.; Alatrash, Gheath; Mittendorf, Elizabeth A.Cancer Research (2017), 77 (19), 5374-5383CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Early-phase clin. trials evaluating CD8+ T cell-eliciting, HER2-derived peptide vaccines administered to HER2+ breast cancer patients in the adjuvant setting suggest synergy between the vaccines and trastuzumab, the mAb targeting the HER2 protein. Among 60 patients enrolled in clin. trials evaluating the E75 + GM-CSF and GP2 + GM-CSF vaccines, there have been no recurrences in patients vaccinated after receiving trastuzumab as part of std. therapy in the per treatment analyses conducted after a median follow-up of greater than 34 mo. Here, we describe a mechanism by which this synergy may occur. Flow cytometry showed that trastuzumab facilitated uptake of HER2 by dendritic cells (DC), which was mediated by the Fc receptor and was specific to trastuzumab. In vitro, increased HER2 uptake by DC increased cross-presentation of E75, the immunodominant epitope derived from the HER2 protein, an observation confirmed in two in vivo mouse models. This increased E75 cross-presentation, mediated by trastuzumab treatment, enabled more efficient expansion of E75-specific cytotoxic T cells (E75-CTL). These results demonstrate a mechanism by which trastuzumab links innate and adaptive immunity by facilitating activation of antigen-specific T cells. On the basis of these data, we conclude that HER2-pos. breast cancer patients that have been treated with trastuzumab may experience a more robust antitumor immune response by restimulation of T cells with the E75 peptide vaccine, thereby accounting for the improved disease-free survival obsd. with combination therapy.
- 67Chaussade, C.; Rewcastle, G. W.; Kendall, J. D.; Denny, W. A.; Cho, K.; Grønning, L. M.; Chong, M. L.; Anagnostou, S. H.; Jackson, S. P.; Daniele, N.; Shepherd, P. R. Evidence for functional redundancy of class IA PI3K isoforms in insulin signalling. Biochem. J. 2007, 404, 449– 458, DOI: 10.1042/BJ20070003Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXlvVSgsr8%253D&md5=b74b9308f1e9e36010ccb4163e3a957bEvidence for functional redundancy of class IA PI3K isoforms in insulin signallingChaussade, Claire; Rewcastle, Gordon W.; Kendall, Jackie D.; Denny, William A.; Cho, Kitty; Gronning, Line M.; Chong, Mei Ling; Anagnostou, Sasha H.; Jackson, Shaun P.; Daniele, Nathalie; Shepherd, Peter R.Biochemical Journal (2007), 404 (3), 449-458CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)Recent genetic knock-in and pharmacol. approaches have suggested that, of class IA PI3Ks (phosphatidylinositol 3-kinases), it is the p110α isoform (PIK3CA) that plays the predominant role in insulin signaling. We have used isoform-selective inhibitors of class IA PI3K to dissect further the roles of individual p110 isoforms in insulin signaling. These include a p110α-specific inhibitor (PIK-75), a p110α-selective inhibitor (PI-103), a p110β-specific inhibitor (TGX-221) and a p110δ-specific inhibitor (IC87114). Although we find that p110α is necessary for insulin-stimulated phosphorylation of PKB (protein kinase B) in several cell lines, we find that this is not the case in HepG2 hepatoma cells. Inhibition of p110β or p110δ alone was also not sufficient to block insulin signaling to PKB in these cells, but, when added in combination with p110α inhibitors, they are able to significantly attenuate insulin signaling. Surprisingly, in J774.2 macrophage cells, insulin signaling to PKB was inhibited to a similar extent by inhibitors of p110α, p110β or p110δ. These results provide evidence that p110β and p110δ can play a role in insulin signaling and also provide the first evidence that there can be functional redundancy between p110 isoforms. Further, our results indicate that the degree of functional redundancy is linked to the relative levels of expression of each isoform in the target cells.
- 68Freeman-Cook, K. D.; Hoffman, R. L.; Johnson, T. W. Lipophilic efficiency: the most important efficiency metric in medicinal chemistry. Future Med. Chem. 2013, 5, 113– 115, DOI: 10.4155/fmc.12.208Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1aiu7g%253D&md5=36170decfa1f1e3ef09ecf772322a210Lipophilic efficiency: the most important efficiency metric in medicinal chemistryFreeman-Cook, Kevin D.; Hoffman, Robert L.; Johnson, Ted W.Future Medicinal Chemistry (2013), 5 (2), 113-115CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review is given on the crit. role of lipophilicity in drug discovery and optimization of general absorption, distribution, metab. and excretion properties, toxicol. profiles and ultimately pharmacol. response. Math. equations are presented concerning the impact of lipophilicity on the dose and the calcn. of the lipophilic ligand efficiency (LipE).
- 69Leeson, P. D.; Springthorpe, B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat. Rev. Drug Discovery 2007, 6, 881– 890, DOI: 10.1038/nrd2445Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1els7rL&md5=5665a9e2dc266a1ee096e20625757ef7The influence of drug-like concepts on decision-making in medicinal chemistryLeeson, Paul D.; Springthorpe, BrianNature Reviews Drug Discovery (2007), 6 (11), 881-890CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Despite the wide acceptance of drug-like principles such as the 'rule of five', this anal. of mols. currently being synthesized in leading pharmaceutical companies reveals that their phys. properties differ significantly from those of recently discovered oral drugs. The marked increase in lipophilicity in particular could increase the likelihood of attrition in drug development. The application of guidelines linked to the concept of drug-likeness, such as the 'rule of five', has gained wide acceptance as an approach to reduce attrition in drug discovery and development. However, despite this acceptance, anal. of recent trends reveals that the phys. properties of mols. that are currently being synthesized in leading drug discovery companies differ significantly from those of recently discovered oral drugs and compds. in clin. development. The consequences of the marked increase in lipophilicity - the most important drug-like phys. property - include a greater likelihood of lack of selectivity and attrition in drug development. Tackling the threat of compd.-related toxicol. attrition needs to move to the mainstream of medicinal chem. decision-making.
- 70Chaikuad, A.; Koch, P.; Laufer, S. A.; Knapp, S. The Cysteinome of Protein Kinases as a Target in Drug Development. Angew. Chem., Int. Ed. 2018, 57, 4372– 4385, DOI: 10.1002/anie.201707875Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitFGgs7c%253D&md5=311ba5ab53dc4a5dc1dc92ddd4b6d4c2The Cysteinome of Protein Kinases as a Target in Drug DevelopmentChaikuad, Apirat; Koch, Pierre; Laufer, Stefan A.; Knapp, StefanAngewandte Chemie, International Edition (2018), 57 (16), 4372-4385CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Drugs that function through covalent bond formation represent a considerable fraction of the authors' repository of effective medicines but safety concerns and the complexity of developing covalent inhibitors has rendered covalent targeting a less attractive strategy for rational drug design. The recent approval of four covalent kinase inhibitors and the development of highly potent covalent kinase probes with exceptional selectivity has raised significant interest in industry and academic research and validated the concept of covalent kinase targeting for clin. applications. The abundance of cysteines at diverse positions in and around the kinase active site suggests that a large fraction of kinases can be targeted by covalent inhibitors. Herein, the authors review recent developments of this rapidly growing area in kinase drug development and highlight the unique opportunities and challenges of this strategy.
- 71Zhang, T.; Inesta-Vaquera, F.; Niepel, M.; Zhang, J.; Ficarro, S. B.; Machleidt, T.; Xie, T.; Marto, J. A.; Kim, N.; Sim, T.; Laughlin, J. D.; Park, H.; LoGrasso, P. V.; Patricelli, M.; Nomanbhoy, T. K.; Sorger, P. K.; Alessi, D. R.; Gray, N. S. Discovery of potent and selective covalent inhibitors of JNK. Chem. Biol. 2012, 19, 140– 154, DOI: 10.1016/j.chembiol.2011.11.010Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1Ghurw%253D&md5=f7d9647fb2be162c637b1d31a778ceb4Discovery of Potent and Selective Covalent Inhibitors of JNKZhang, Tinghu; Inesta-Vaquera, Francisco; Niepel, Mario; Zhang, Jianming; Ficarro, Scott B.; Machleidt, Thomas; Xie, Ting; Marto, Jarrod A.; Kim, Nam Doo; Sim, Taebo; Laughlin, John D.; Park, Hajeung; LoGrasso, Philip V.; Patricelli, Matt; Nomanbhoy, Tyzoon K.; Sorger, Peter K.; Alessi, Dario R.; Gray, Nathanael S.Chemistry & Biology (Oxford, United Kingdom) (2012), 19 (1), 140-154CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)The mitogen-activated kinases JNK1/2/3 are key enzymes in signaling modules that transduce and integrate extracellular stimuli into coordinated cellular response. Here, we report the discovery of irreversible inhibitors of JNK1/2/3. We describe two JNK3 cocrystal structures at 2.60 and 2.97 Å resoln. that show the compds. form covalent bonds with a conserved cysteine residue. JNK-IN-8 is a selective JNK inhibitor that inhibits phosphorylation of c-Jun, a direct substrate of JNK, in cells exposed to submicromolar drug in a manner that depends on covalent modification of the conserved cysteine residue. Extensive biochem., cellular, and pathway-based profiling establish the selectivity of JNK-IN-8 for JNK and suggests that the compd. will be broadly useful as a pharmacol. probe of JNK-dependent signal transduction. Potential lead compds. have also been identified for kinases, including IRAK1, PIK3C3, PIP4K2C, and PIP5K3.
- 72Zhang, T.; Hatcher, J. M.; Teng, M.; Gray, N. S.; Kostic, M. Recent Advances in Selective and Irreversible Covalent Ligand Development and Validation. Cell Chem. Biol. 2019, 26, 1486– 1500, DOI: 10.1016/j.chembiol.2019.09.012Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitVSlsrzO&md5=ce957a868ca27f545243d28e60dc5868Recent Advances in Selective and Irreversible Covalent Ligand Development and ValidationZhang, Tinghu; Hatcher, John M.; Teng, Mingxing; Gray, Nathanael S.; Kostic, MilkaCell Chemical Biology (2019), 26 (11), 1486-1500CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)A review. Some of the most widely used drugs, such as aspirin and penicillin, are covalent drugs. Covalent binding can improve potency, selectivity, and duration of the effects, but the intrinsic reactivity represents a potential liability and may result in idiosyncratic toxicity. For decades, the cons were believed to outweigh the pros, and covalent targeting was deprioritized in drug discovery. Recently, several covalent inhibitors have been approved for cancer treatment, thus rebooting the field. In this review, we briefly reflect on the history of selective covalent targeting, and provide a comprehensive overview of emerging developments from a chem. biol. stand-point. Our discussion will reflect on efforts to validate irreversible covalent ligands, expand the scope of targets, and discover new ligands and warheads. We conclude with a brief commentary of remaining limitations and emerging opportunities in selective covalent targeting.
- 73Johnson, K. A. Fitting enzyme kinetic data with KinTek Global Kinetic Explorer. Computer Methods Part B; Methods in Enzymology; Elsevier B.V., 2009; Vol. 467, pp 601– 626.Google ScholarThere is no corresponding record for this reference.
- 74Johnson, K. A.; Simpson, Z. B.; Blom, T. Global kinetic explorer: a new computer program for dynamic simulation and fitting of kinetic data. Anal. Biochem. 2009, 387, 20– 29, DOI: 10.1016/j.ab.2008.12.024Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXisFKjt7c%253D&md5=1d0227c91a334f47edae70f5a89478ffGlobal Kinetic Explorer: A new computer program for dynamic simulation and fitting of kinetic dataJohnson, Kenneth A.; Simpson, Zachary B.; Blom, ThomasAnalytical Biochemistry (2009), 387 (1), 20-29CODEN: ANBCA2; ISSN:0003-2697. (Elsevier B.V.)We describe a new dynamic kinetic simulation program that allows multiple data sets to be fit simultaneously to a single model based on numerical integration of the rate equations describing the reaction mechanism. Unlike other programs that allow fitting based on numerical integration of rate equations, in the dynamic simulation rate consts., output factors, and starting concns. of reactants can be scrolled while observing the change in the shape of the simulated reaction curves. Fast dynamic simulation facilitates the exploration of initial parameters that serve as the starting point for nonlinear regression in fitting data and facilitates exploration of the relationships between individual consts. and observable reactions. The exploration of parameter space by dynamic simulation provides a powerful tool for learning kinetics and for evaluating the extent to which parameters are constrained by the data. This feature is crit. to avoid overly complex models that are not supported by the data.
- 75Janes, M. R.; Zhang, J.; Li, L. S.; Hansen, R.; Peters, U.; Guo, X.; Chen, Y.; Babbar, A.; Firdaus, S. J.; Darjania, L.; Feng, J.; Chen, J. H.; Li, S.; Li, S.; Long, Y. O.; Thach, C.; Liu, Y.; Zarieh, A.; Ely, T.; Kucharski, J. M.; Kessler, L. V.; Wu, T.; Yu, K.; Wang, Y.; Yao, Y.; Deng, X.; Zarrinkar, P. P.; Brehmer, D.; Dhanak, D.; Lorenzi, M. V.; Hu-Lowe, D.; Patricelli, M. P.; Ren, P.; Liu, Y. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell 2018, 172, 578.e17– 589.e17, DOI: 10.1016/j.cell.2018.01.006Google ScholarThere is no corresponding record for this reference.
- 76Müller, M. P.; Jeganathan, S.; Heidrich, A.; Campos, J.; Goody, R. S. Nucleotide based covalent inhibitors of KRas can only be efficient in vivo if they bind reversibly with GTP-like affinity. Sci. Rep. 2017, 7, 3687 DOI: 10.1038/s41598-017-03973-6Google ScholarThere is no corresponding record for this reference.
- 77Chen, P.; Deng, Y. L.; Bergqvist, S.; Falk, M. D.; Liu, W.; Timofeevski, S.; Brooun, A. Engineering of an isolated p110α subunit of PI3Kα permits crystallization and provides a platform for structure-based drug design. Protein Sci. 2014, 23, 1332– 1340, DOI: 10.1002/pro.2517Google Scholar77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsFelsb3I&md5=4edd5247eb9e8729dc63d50713fd5314Engineering of an isolated p110α subunit of PI3Kα permits crystallization and provides a platform for structure-based drug designChen, Ping; Deng, Ya-Li; Bergqvist, Simon; Falk, Matthew D.; Liu, Wei; Timofeevski, Sergei; Brooun, AlexeiProtein Science (2014), 23 (10), 1332-1340CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)PI3Kα remains an attractive target for the development of anticancer targeted therapy. A no. of p110α crystal structures in complex with the nSH2-iSH2 fragment of p85 regulatory subunit have been reported, including a few small mol. co-crystal structures, but the utilization of this crystal form is limited by low diffraction resoln. and a crystal packing artifact that partially blocks the ATP binding site. Taking advantage of recent data on the functional characterization of the lipid binding properties of p110α, we designed a set of novel constructs allowing prodn. of isolated stable p110α subunit missing the adapter-binding domain and lacking or featuring a modified C-terminal lipid binding motif. While this protein is not catalytically competent to phosphorylate its substrate PIP2, it retains ligand binding properties as indicated by direct binding studies with a pan-PI3Kα inhibitor. Addnl., we detd. apo and PF-04691502 bound crystal structures of the p110α (105-1048) subunit at 2.65 and 2.85 Å, resp. Comparison of isolated p110α(105-1048) with the p110α/p85 complex reveals a high degree of structural similarity, which validates suitability of this catalytically inactive p110α for iterative SBDD. Importantly, this crystal form of p110α readily accommodates the binding of noncovalent inhibitor by means of a fully accessible ATP site. The strategy presented here can be also applied to structural studies of other members of PI3KIA family.
- 78Marcotte, D. J.; Liu, Y. T.; Arduini, R. M.; Hession, C. A.; Miatkowski, K.; Wildes, C. P.; Cullen, P. F.; Hong, V.; Hopkins, B. T.; Mertsching, E.; Jenkins, T. J.; Romanowski, M. J.; Baker, D. P.; Silvian, L. F. Structures of human Bruton’s tyrosine kinase in active and inactive conformations suggest a mechanism of activation for TEC family kinases. Protein Sci. 2010, 19, 429– 439, DOI: 10.1002/pro.321Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXisFOhtbs%253D&md5=8c2dae6949f8bdb627d68ee14edc8d4fStructures of human Bruton's tyrosine kinase in active and inactive conformations suggest a mechanism of activation for TEC family kinasesMarcotte, Douglas J.; Liu, Yu-Ting; Arduini, Robert M.; Hession, Catherine A.; Miatkowski, Konrad; Wildes, Craig P.; Cullen, Patrick F.; Hong, Victor; Hopkins, Brian T.; Mertsching, Elisabeth; Jenkins, Tracy J.; Romanowski, Michael J.; Baker, Darren P.; Silvian, Laura F.Protein Science (2010), 19 (3), 429-439CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Bruton's tyrosine kinase (BTK), a member of the TEC family of kinases, plays a crucial role in B-cell maturation and mast cell activation. Although the structures of the unphosphorylated mouse BTK kinase domain and the unphosphorylated and phosphorylated kinase domains of human ITK are known, understanding the kinase selectivity profiles of BTK inhibitors has been hampered by the lack of availability of a high resoln., ligand-bound BTK structure. Here, we report the crystal structures of the human BTK kinase domain bound to either Dasatinib (BMS-354825) at 1.9 Å resoln. or to 4-amino-5-(4-phenoxyphenyl)-7H-pyrrolospyrimidin-7-yl-cyclopentane at 1.6 Å resoln. This data provides information relevant to the development of small mol. inhibitors targeting BTK and the TEC family of nonreceptor tyrosine kinases. Anal. of the structural differences between the TEC and Src families of kinases near the Trp-Glu-Ile motif in the N-terminal region of the kinase domain suggests a mechanism of regulation of the TEC family members.
- 79Gajiwala, K. S.; Feng, J.; Ferre, R.; Ryan, K.; Brodsky, O.; Weinrich, S.; Kath, J. C.; Stewart, A. Insights into the aberrant activity of mutant EGFR kinase domain and drug recognition. Structure 2013, 21, 209– 219, DOI: 10.1016/j.str.2012.11.014Google Scholar79https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXotFeq&md5=439bfda2e4e33786a15742e53cc271bcInsights into the Aberrant Activity of Mutant EGFR Kinase Domain and Drug RecognitionGajiwala, Ketan S.; Feng, Junli; Ferre, RoseAnn; Ryan, Kevin; Brodsky, Oleg; Weinrich, Scott; Kath, John C.; Stewart, AlStructure (Oxford, United Kingdom) (2013), 21 (2), 209-219CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)The oncogenicity of the L858R mutant form of the epidermal growth factor receptor (EGFR) in non-small-cell lung cancer is thought to be due to the constitutive activation of its kinase domain. The selectivity of the marketed drugs gefitinib and erlotinib for L858R mutant is attributed to their specific recognition of the active kinase and to weaker ATP binding by L858R EGFR. We present crystal structures showing that neither L858R nor the drug-resistant L858R+T790M EGFR kinase domain is in the constitutively active conformation. Addnl. co-crystal structures show that gefitinib and dacomitinib, an irreversible anilinoquinazoline deriv. currently in clin. development, may not be conformation specific for the active state of the enzyme. Structural data further reveal the potential mode of recognition of one of the autophosphorylation sites in the C-terminal tail, Tyr-1016, by the kinase domain. Biochem. and biophys. evidence suggest that the oncogenic mutations impact the conformational dynamics of the enzyme.
- 80Montero, D.; Tachibana, C.; Rahr Winther, J.; Appenzeller-Herzog, C. Intracellular glutathione pools are heterogeneously concentrated. Redox Biol. 2013, 1, 508– 513, DOI: 10.1016/j.redox.2013.10.005Google Scholar80https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXptFGlsg%253D%253D&md5=cb40f4e69e51f8b6f09f133f7b1b87f3Intracellular glutathione pools are heterogeneously concentratedMontero, Davide; Tachibana, Christine; Rahr Winther, Jakob; Appenzeller-Herzog, ChristianRedox Biology (2013), 1 (1), 508-513CODEN: RBEIB3; ISSN:2213-2317. (Elsevier B.V.)Glutathione is present in millimolar concns. in the cell, but its relative distribution among cellular compartments remains elusive. We have chosen the endoplasmic reticulum (ER) as an example organelle to study compartment-specific glutathione levels. Using a glutaredoxin sensor (sCGrx1pER), which rapidly and specifically equilibrates with the reduced glutathione (GSH)-glutathione disulfide (GSSG) redox couple with known equil. const., we showed that the [GSH]:[GSSG] ratio in the ER of intact HeLa cells is less than 7:1. Taking into consideration the previously detd. value for [GSH]2:[GSSG] in the ER of 83 mM, this translates into a total glutathione concn. in the ER ([GStot]=[GSH]+2[GSSG]) of greater than 15 mM. Since the integrated, intracellular [GStot] was measured as ∼7 mM, we conclude the existence of a [GStot] gradient across the ER membrane. A possible homeostatic mechanism by which cytosol-derived glutathione is trapped in the ER is discussed. We propose a high [GStot] as a distinguishing feature of the ER environment compared to the extracellular space.
- 81Choi, B.; Rempala, G. A.; Kim, J. K. Beyond the Michaelis-Menten equation: Accurate and efficient estimation of enzyme kinetic parameters. Sci. Rep. 2017, 7, 17018 DOI: 10.1038/s41598-017-17072-zGoogle Scholar81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M3otlWmug%253D%253D&md5=24801e3eb36b6bd649d17583ae5db895Beyond the Michaelis-Menten equation: Accurate and efficient estimation of enzyme kinetic parametersChoi Boseung; Rempala Grzegorz A; Kim Jae KyoungScientific reports (2017), 7 (1), 17018 ISSN:.Examining enzyme kinetics is critical for understanding cellular systems and for using enzymes in industry. The Michaelis-Menten equation has been widely used for over a century to estimate the enzyme kinetic parameters from reaction progress curves of substrates, which is known as the progress curve assay. However, this canonical approach works in limited conditions, such as when there is a large excess of substrate over enzyme. Even when this condition is satisfied, the identifiability of parameters is not always guaranteed, and often not verifiable in practice. To overcome such limitations of the canonical approach for the progress curve assay, here we propose a Bayesian approach based on an equation derived with the total quasi-steady-state approximation. In contrast to the canonical approach, estimates obtained with this proposed approach exhibit little bias for any combination of enzyme and substrate concentrations. Importantly, unlike the canonical approach, an optimal experiment to identify parameters with certainty can be easily designed without any prior information. Indeed, with this proposed design, the kinetic parameters of diverse enzymes with disparate catalytic efficiencies, such as chymotrypsin, fumarase, and urease, can be accurately and precisely estimated from a minimal amount of timecourse data. A publicly accessible computational package performing such accurate and efficient Bayesian inference for enzyme kinetics is provided.
- 82Schwartz, P. A.; Kuzmic, P.; Solowiej, J.; Bergqvist, S.; Bolanos, B.; Almaden, C.; Nagata, A.; Ryan, K.; Feng, J.; Dalvie, D.; Kath, J. C.; Xu, M.; Wani, R.; Murray, B. W. Covalent EGFR inhibitor analysis reveals importance of reversible interactions to potency and mechanisms of drug resistance. Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 173– 178, DOI: 10.1073/pnas.1313733111Google Scholar82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXps1eluw%253D%253D&md5=47197c5e7876a238eefb2e8e7de305eaCovalent EGFR inhibitor analysis reveals importance of reversible interactions to potency and mechanisms of drug resistanceSchwartz, Phillip A.; Kuzmic, Petr; Solowiej, James; Bergqvist, Simon; Bolanos, Ben; Almaden, Chau; Nagata, Asako; Ryan, Kevin; Feng, Junli; Dalvie, Deepak; Kath, John C.; Xu, Meirong; Wani, Revati; Murray, Brion WilliamProceedings of the National Academy of Sciences of the United States of America (2014), 111 (1), 173-178CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Covalent inhibition is a reemerging paradigm in kinase drug design, but the roles of inhibitor binding affinity and chem. reactivity in overall potency are not well-understood. To characterize the underlying mol. processes at a microscopic level and det. the appropriate kinetic consts., specialized exptl. design and advanced numerical integration of differential equations are developed. Previously uncharacterized investigational covalent drugs reported here are shown to be extremely effective epidermal growth factor receptor (EGFR) inhibitors (kinact/Ki in the range 105-107 M-1s-1), despite their low specific reactivity (kinact ≤ 2.1 × 10-3 s-1), which is compensated for by high binding affinities (Ki < 1 nM). For inhibitors relying on reactivity to achieve potency, noncovalent enzyme-inhibitor complex partitioning between inhibitor dissocn. and bond formation is central. Interestingly, reversible binding affinity of EGFR covalent inhibitors is highly correlated with antitumor cell potency. Furthermore, cellular potency for a subset of covalent inhibitors can be accounted for solely through reversible interactions. One reversible interaction is between EGFR-Cys797 nucleophile and the inhibitor's reactive group, which may also contribute to drug resistance. Because covalent inhibitors target a cysteine residue, the effects of its oxidn. on enzyme catalysis and inhibitor pharmacol. are characterized. Oxidn. of the EGFR cysteine nucleophile does not alter catalysis but has widely varied effects on inhibitor potency depending on the EGFR context (e.g., oncogenic mutations), type of oxidn. (sulfinylation or glutathiolation), and inhibitor architecture. These methods, parameters, and insights provide a rational framework for assessing and designing effective covalent inhibitors.
- 83Yu, C.; Miao, W.; Wang, J.; Hao, E.; Jiao, L. PyrrolylBODIPYs: Syntheses, Properties, and Application as Environment-Sensitive Fluorescence Probes. ACS Omega 2017, 2, 3551– 3561, DOI: 10.1021/acsomega.7b00444Google Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFGhu73P&md5=711bec97efed886779c0a8e12835d2f1PyrrolylBODIPYs: Syntheses, Properties, and Application as Environment-Sensitive Fluorescence ProbesYu, Changjiang; Miao, Wei; Wang, Jun; Hao, Erhong; Jiao, LijuanACS Omega (2017), 2 (7), 3551-3561CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)Four pyrrole B-ring functionalized pyrrolylBODIPYs and their B-ring unsubstituted analogs were synthesized from easily accessible starting 5-halo-2-formylpyrroles and were characterized by NMR, HRMS, X-ray anal., optical/electronic properties. In great contrast to the substitution(s) at the other two pyrrolic units, electron donating substituent(s) at pyrrole B-ring bring significantly blue-shift of the absorption and emission bands. Cyclic voltammetry and DFT calcns. indicate that this blue-shift may be attributed to the increased HOMO and the LUMO energy levels and the overall increase of the energy band gaps. These pyrrolylBODIPYs generally show intense absorption (centered at 570-624 nm) and fluorescence emission (582-654 nm) in nonpolar solvents. A gradual decrease of the fluorescence intensity was obsd. for these dyes with the increase of solvent dipolar moment, which combines with the red to far-red absorption/emission rendering these pyrrolylBODIPYs potential applications as environment-sensitive fluorescence probes as demonstrated in this work for BSA.
Cited By
This article is cited by 19 publications.
- Yasir S. Raouf. Covalent Inhibitors: To Infinity and Beyond. Journal of Medicinal Chemistry 2024, 67
(13)
, 10513-10516. https://doi.org/10.1021/acs.jmedchem.4c01308
- Laura Hillebrand, Xiaojun Julia Liang, Ricardo A. M. Serafim, Matthias Gehringer. Emerging and Re-emerging Warheads for Targeted Covalent Inhibitors: An Update. Journal of Medicinal Chemistry 2024, 67
(10)
, 7668-7758. https://doi.org/10.1021/acs.jmedchem.3c01825
- Moritz Schwarz, Maksym Kurkunov, Florian Wittlinger, Ramona Rudalska, Guiqun Wang, Martin Peter Schwalm, Alexander Rasch, Benedikt Wagner, Stefan A. Laufer, Stefan Knapp, Daniel Dauch, Matthias Gehringer. Development of Highly Potent and Selective Covalent FGFR4 Inhibitors Based on SNAr Electrophiles. Journal of Medicinal Chemistry 2024, 67
(8)
, 6549-6569. https://doi.org/10.1021/acs.jmedchem.3c02483
- Ingo V. Hartung, Joachim Rudolph, Mary M. Mader, Monique P. C. Mulder, Paul Workman. Expanding Chemical Probe Space: Quality Criteria for Covalent and Degrader Probes. Journal of Medicinal Chemistry 2023, 66
(14)
, 9297-9312. https://doi.org/10.1021/acs.jmedchem.3c00550
- Aarion Romany, Ruibin Liu, Shaoqi Zhan, Joseph Clayton, Jana Shen. Analysis of the ERK Pathway Cysteinome for Targeted Covalent Inhibition of RAF and MEK Kinases. Journal of Chemical Information and Modeling 2023, 63
(8)
, 2483-2494. https://doi.org/10.1021/acs.jcim.3c00014
- Bente A. Somsen, Rick J.C. Schellekens, Carlo J.A. Verhoef, Michelle R. Arkin, Christian Ottmann, Peter J. Cossar, Luc Brunsveld. Reversible Dual-Covalent Molecular Locking of the 14-3-3/ERRγ Protein–Protein Interaction as a Molecular Glue Drug Discovery Approach. Journal of the American Chemical Society 2023, 145
(12)
, 6741-6752. https://doi.org/10.1021/jacs.2c12781
- Johannes Karges, Seth M. Cohen. Rhenium(V) Complexes as Cysteine-Targeting Coordinate Covalent Warheads. Journal of Medicinal Chemistry 2023, 66
(4)
, 3088-3105. https://doi.org/10.1021/acs.jmedchem.2c02074
- Jindi Huang, Liye Chen, Jiangxia Wu, Daiqiao Ai, Ji-Quan Zhang, Tie-Gen Chen, Ling Wang. Targeting the PI3K/AKT/mTOR Signaling Pathway in the Treatment of Human Diseases: Current Status, Trends, and Solutions. Journal of Medicinal Chemistry 2022, 65
(24)
, 16033-16061. https://doi.org/10.1021/acs.jmedchem.2c01070
- Tanel Sõrmus, Darja Lavogina, Anu Teearu, Erki Enkvist, Asko Uri, Kaido Viht. Construction of Covalent Bisubstrate Inhibitor of Protein Kinase Reacting with Cysteine Residue at Substrate-Binding Site. Journal of Medicinal Chemistry 2022, 65
(16)
, 10975-10991. https://doi.org/10.1021/acs.jmedchem.2c00067
- Xiaoteng Feng, Min Du, Sijin Li, Yifan Zhang, Jie Ding, Jiarou Wang, Yiru Wang, Ping Liu. Hydroxysafflor yellow A regulates lymphangiogenesis and inflammation via the inhibition of PI3K on regulating AKT/mTOR and NF-κB pathway in macrophages to reduce atherosclerosis in ApoE-/- mice. Phytomedicine 2023, 112 , 154684. https://doi.org/10.1016/j.phymed.2023.154684
- Martina De Pascale, Lukas Bissegger, Chiara Tarantelli, Florent Beaufils, Alessandro Prescimone, Hayget Mohamed Seid Hedad, Omar Kayali, Clara Orbegozo, Luka Raguž, Thorsten Schaefer, Paul Hebeisen, Francesco Bertoni, Matthias P. Wymann, Chiara Borsari. Investigation of morpholine isosters for the development of a potent, selective and metabolically stable mTOR kinase inhibitor. European Journal of Medicinal Chemistry 2023, 248 , 115038. https://doi.org/10.1016/j.ejmech.2022.115038
- Uros Stojiljkovic, Claudio Meyer, Pierre Boulay, Paul Hebeisen, Denise Rageot, Matthias P. Wymann, Chiara Borsari. Stereospecific Synthesis of Substituted Sulfamidates as Privileged Morpholine Building Blocks. Synthesis 2023, 55
(03)
, 499-509. https://doi.org/10.1055/a-1915-7794
- Chuan Wan, Zhanfeng Hou, Dongyan Yang, Ziyuan Zhou, Hongkun Xu, Yuena Wang, Chuan Dai, Mingchan Liang, Jun Meng, Jiean Chen, Feng Yin, Rui Wang, Zigang Li. The thiol-sulfoxonium ylide photo-click reaction for bioconjugation. Chemical Science 2023, 14
(3)
, 604-612. https://doi.org/10.1039/D2SC05650J
- Alba Ramos-Llorca, Lisse Decraecker, Valérie M. Y. Cacheux, Irena Zeiburlina, Michelle De bruyn, Louise Battut, Carlos Moreno-Cinos, Davide Ceradini, Eric Espinosa, Gilles Dietrich, Maya Berg, Ingrid De Meester, Pieter Van Der Veken, Guy Boeckxstaens, Anne-Marie Lambeir, Alexandre Denadai-Souza, Koen Augustyns. Chemically diverse activity-based probes with unexpected inhibitory mechanisms targeting trypsin-like serine proteases. Frontiers in Chemistry 2023, 10 https://doi.org/10.3389/fchem.2022.1089959
- Ricardo A.M. Serafim, Lisa Haarer, Júlia G.B. Pedreira, Matthias Gehringer. Covalent chemical probes for protein kinases. Current Research in Chemical Biology 2023, 3 , 100040. https://doi.org/10.1016/j.crchbi.2022.100040
- Zheng Zhao, Philip E. Bourne. Systematic Exploration of Privileged Warheads for Covalent Kinase Drug Discovery. Pharmaceuticals 2022, 15
(11)
, 1322. https://doi.org/10.3390/ph15111322
- Kirsten McAulay, Alan Bilsland, Marta Bon. Reactivity of Covalent Fragments and Their Role in Fragment Based Drug Discovery. Pharmaceuticals 2022, 15
(11)
, 1366. https://doi.org/10.3390/ph15111366
- Dongfang Lv, Yun Ji, Qian Zhang, Zhuozhuo Shi, Tengfei Chen, Chao Zhang, Xiangyun Wang, Taotao Ren, Zhaowang Gao, Chongfu Zhong. Mailuoshutong pill for varicocele-associated male infertility—Phytochemical characterisation and multitarget mechanism. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.961011
- Hongyan Du, Dejun Jiang, Junbo Gao, Xujun Zhang, Lingxiao Jiang, Yundian Zeng, Zhenxing Wu, Chao Shen, Lei Xu, Dongsheng Cao, Tingjun Hou, Peichen Pan. Proteome-Wide Profiling of the Covalent-Druggable Cysteines with a Structure-Based Deep Graph Learning Network. Research 2022, 2022 https://doi.org/10.34133/2022/9873564
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 83 other publications.
- 1Miller, V. A.; Hirsh, V.; Cadranel, J.; Chen, Y.-M.; Park, K.; Kim, S.-W.; Zhou, C.; Su, W.-C.; Wang, M.; Sun, Y.; Heo, D. S.; Crino, L.; Tan, E.-H.; Chao, T.-Y.; Shahidi, M.; Cong, X. J.; Lorence, R. M.; Yang, J. C.-H. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol. 2012, 13, 528– 538, DOI: 10.1016/S1470-2045(12)70087-61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmslyhsbg%253D&md5=6ddd7f552e8ab1ebb43fb699e27894f5Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trialMiller, Vincent A.; Hirsh, Vera; Cadranel, Jacques; Chen, Yuh-Min; Park, Keunchil; Kim, Sang-We; Zhou, Caicun; Su, Wu-Chou; Wang, Mengzhao; Sun, Yan; Heo, Dae Seog; Crino, Lucio; Tan, Eng-Huat; Chao, Tsu-Yi; Shahidi, Mehdi; Cong, Xiuyu Julie; Lorence, Robert M.; Yang, James Chih-HsinLancet Oncology (2012), 13 (5), 528-538CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)Summary: Background: Afatinib, an irreversible ErbB-family blocker, has shown preclin. activity when tested in EGFR mutant models with mutations that confer resistance to EGFR tyrosine-kinase inhibitors. We aimed to assess its efficacy in patients with advanced lung adenocarcinoma with previous treatment failure on EGFR tyrosine-kinase inhibitors. Methods: In this phase 2b/3 trial, we enrolled patients with stage IIIB or IV adenocarcinoma and an Eastern Cooperative Oncol. Group performance (ECOG) performance score of 0-2 who had received one or two previous chemotherapy regimens and had disease progression after at least 12 wk of treatment with erlotinib or gefitinib. We used a computer-generated sequence to randomly allocate patients (2:1) to either afatinib (50 mg per day) or placebo; all patients received best supportive care. Randomisation was done in blocks of three and was stratified by sex and baseline ECOG performance status (0-1 vs 2). Investigators, patients, and the trial sponsor were masked to treatment assignment. The primary endpoint was overall survival (from date of randomisation to death), analyzed on an intention-to-treat basis. This study is registered with ClinicalTrials.gov, no. NCT00656136. Findings: Between May 26, 2008, and Sept 21, 2009, we identified 697 patients, 585 of whom were randomly allocated to treatment (390 to afatinib, 195 to placebo). Median overall survival was 10·8 mo (95% CI 10·0-12·0) in the afatinib group and 12·0 mo (10·2-14·3) in the placebo group (hazard ratio 1·08, 95% CI 0·86-1·35; p=0·74). Median progression-free survival was longer in the afatinib group (3·3 mo, 95% CI 2·79-4·40) than it was in the placebo group (1·1 mo, 0·95-1·68; hazard ratio 0·38, 95% CI 0·31-0·48; p<0·0001). No complete responses to treatment were noted; 29 (7%) patients had a partial response in the afatinib group, as did one patient in the placebo group. Subsequent cancer treatment was given to 257 (68%) patients in the afatinib group and 153 (79%) patients in the placebo group. The most common adverse events in the afatinib group were diarrhoea (339 [87%] of 390 patients; 66 [17%] were grade 3) and rash or acne (305 [78%] patients; 56 [14%] were grade 3). These events occurred less often in the placebo group (18 [9%] of 195 patients had diarrhoea; 31 [16%] had rash or acne), all being grade 1 or 2. Drug-related serious adverse events occurred in 39 (10%) patients in the afatinib group and one (<1%) patient in the placebo group. We recorded two possibly treatment-related deaths in the afatinib group. Interpretation: Although we recorded no benefit in terms of overall survival with afatinib (which might have been affected by cancer treatments given after progression in both groups), our findings for progression-free survival and response to treatment suggest that afatinib could be of some benefit to patients with advanced lung adenocarcinoma who have failed at least 12 wk of previous EGFR tyrosine-kinase inhibitor treatment. Funding: Boehringer Ingelheim Inc.
- 2Pan, Z.; Scheerens, H.; Li, S.-J.; Schultz, B. E.; Sprengeler, P. A.; Burrill, L. C.; Mendonca, R. V.; Sweeney, M. D.; Scott, K. C. K.; Grothaus, P. G.; Jeffery, D. A.; Spoerke, J. M.; Honigberg, L. A.; Young, P. R.; Dalrymple, S. A.; Palmer, J. T. Discovery of Selective Irreversible Inhibitors for Bruton’s Tyrosine Kinase. ChemMedChem 2007, 2, 58– 61, DOI: 10.1002/cmdc.2006002212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjsVWjsbg%253D&md5=023ba8bca9942ad0127d7cf32293e9d2Discovery of selective irreversible inhibitors for bruton's tyrosine kinasePan, Zhengying; Scheerens, Heleen; Li, Shyr-Jiann; Schultz, Brian E.; Sprengeler, Paul A.; Burrill, L. Chuck; Mendonca, Rohan V.; Sweeney, Michael D.; Scott, Keana C. K.; Grothaus, Paul G.; Jeffery, Douglas A.; Spoerke, Jill M.; Honigberg, Lee A.; Young, Peter R.; Dalrymple, Stacie A.; Palmer, James T.ChemMedChem (2007), 2 (1), 58-61CODEN: CHEMGX; ISSN:1860-7179. (Wiley-VCH Verlag GmbH & Co. KGaA)Synthesis and pharmacol. evaluation of a series of peptidomimetic quinoline derivs. was undertaken to evaluate their efficacy in acting as selective irreversible inhibitors of Bruton's tyrosine kinase (Btk). With the relative scarcity of knowledge on the inhibition of Btk it is crucial to discover a potent and selective tool compd. for this kinase. Herein is described the discovery of selective irreversible Btk inhibitors and their efficacy in a mouse RA model.
- 3Yver, A. Osimertinib (AZD9291)─a science-driven, collaborative approach to rapid drug design and development. Ann. Oncol. 2016, 27, 1165– 1170, DOI: 10.1093/annonc/mdw1293https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28joslCgsA%253D%253D&md5=903bbb3edf2d109af352ef0354e4bca6Osimertinib (AZD9291)-a science-driven, collaborative approach to rapid drug design and developmentYver AAnnals of oncology : official journal of the European Society for Medical Oncology (2016), 27 (6), 1165-1170 ISSN:.There is no expanded citation for this reference.
- 4Miles, J.; White, Y. Neratinib for the Treatment of Early-Stage HER2-Positive Breast Cancer. J. Adv. Pract. Oncol. 2018, 9, 750– 7544https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MzhslyrsQ%253D%253D&md5=0e0f12297730802dfd5806865209deefNeratinib for the Treatment of Early-Stage HER2-Positive Breast CancerMiles Jennifer; White YahsinJournal of the advanced practitioner in oncology (2018), 9 (7), 750-754 ISSN:2150-0878.The treatment of breast cancer has been revolutionized by the development of HER2-targeted treatments for patients who are HER2 positive. The HER2 protein is present at high levels in about 30% of breast cancer patients. These high levels are associated with a greater chance of metastasis, relapse, and decreased survival. The current standard of care for early-stage HER2-positive patients includes treatment with 1 year of trastuzumab therapy. Although trastuzumab has improved outcomes, there is still a 20% chance for tumor recurrence and a 16% chance of death. Neratinib was developed to give patients with early-stage HER2-positive breast cancer an option to increase the disease-free survival rate. The 5-year invasive disease-free survival rate was 90.2% (95% confidence interval = 88.3-91.8) in the neratinib group and 87.7% (95% confidence interval = 85.7-89.4) in the placebo group.
- 5Lau, S. C. M.; Batra, U.; Mok, T. S. K.; Loong, H. H. Dacomitinib in the Management of Advanced Non-Small-Cell Lung Cancer. Drugs 2019, 79, 823– 831, DOI: 10.1007/s40265-019-01115-y5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFWmtrfI&md5=e09f6baf1fed49df33397d74698b4c52Dacomitinib in the Management of Advanced Non-Small-Cell Lung CancerLau, Sally C. M.; Batra, Ullas; Mok, Tony S. K.; Loong, Herbert H.Drugs (2019), 79 (8), 823-831CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. The use of targeted therapy in the management of epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer is an important milestone in the management of advanced lung cancer. There are several generations of EGFR tyrosine kinase inhibitors available for clin. use. Dacomitinib is a second-generation irreversible EGFR tyrosine kinase inhibitor with early-phase clin. studies showing efficacy in non-small-cell lung cancer. In the recently published ARCHER 1050 phase III study, dacomitinib given at 45 mg/day orally was superior to gefitinib, a first-generation reversible EGFR tyrosine kinase inhibitor, in improving both progression-free survival and overall survival when given as first-line therapy. There is no prospective evidence to support the use of dacomitinib as subsequent therapy in patients previously treated with chemotherapy or a first-generation EGFR tyrosine kinase inhibitor such as gefitinib and erlotinib. Dacomitinib has not demonstrated any benefit in unselected patients with non-small-cell lung cancer, and its use should be limited to those with known EGFR-sensitizing mutations. Dacomitinib is assocd. with increased toxicities of diarrhea, rash, stomatitis, and paronychia compared with first-generation EGFR inhibitors. Global quality of life was maintained when assessed in phase III studies. Overall, dacomitinib is an important first- line agent in EGFR-mutated non-small-cell lung cancer in otherwise fit patients whose toxicities can be well managed.
- 6Davids, M. S. Acalabrutinib for the initial treatment of chronic lymphocytic leukaemia. Lancet 2020, 395, 1234– 1236, DOI: 10.1016/s0140-6736(20)30372-x6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsVWmsrY%253D&md5=b174e8413a465839fdb071616c793227Acalabrutinib for the initial treatment of chronic lymphocytic leukaemiaDavids, Matthew S.Lancet (2020), 395 (10232), 1234-1236CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A review. This article describes about the acalabrutinib for initial treatment of chronic lymphocytic leukemia.
- 7Guo, Y.; Liu, Y.; Hu, N.; Yu, D.; Zhou, C.; Shi, G.; Zhang, B.; Wei, M.; Liu, J.; Luo, L.; Tang, Z.; Song, H.; Guo, Y.; Liu, X.; Su, D.; Zhang, S.; Song, X.; Zhou, X.; Hong, Y.; Chen, S.; Cheng, Z.; Young, S.; Wei, Q.; Wang, H.; Wang, Q.; Lv, L.; Wang, F.; Xu, H.; Sun, H.; Xing, H.; Li, N.; Zhang, W.; Wang, Z.; Liu, G.; Sun, Z.; Zhou, D.; Li, W.; Liu, L.; Wang, L.; Wang, Z. Discovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase. J. Med. Chem. 2019, 62, 7923– 7940, DOI: 10.1021/acs.jmedchem.9b006877https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsFSku7bP&md5=5cdb551d296e60eb1e82b03b7a0384eeDiscovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton's Tyrosine KinaseGuo, Yunhang; Liu, Ye; Hu, Nan; Yu, Desheng; Zhou, Changyou; Shi, Gongyin; Zhang, Bo; Wei, Min; Liu, Junhua; Luo, Lusong; Tang, Zhiyu; Song, Huipeng; Guo, Yin; Liu, Xuesong; Su, Dan; Zhang, Shuo; Song, Xiaomin; Zhou, Xing; Hong, Yuan; Chen, Shuaishuai; Cheng, Zhenzhen; Young, Steve; Wei, Qiang; Wang, Haisheng; Wang, Qiuwen; Lv, Lei; Wang, Fan; Xu, Haipeng; Sun, Hanzi; Xing, Haimei; Li, Na; Zhang, Wei; Wang, Zhongbo; Liu, Guodong; Sun, Zhijian; Zhou, Dongping; Li, Wei; Liu, Libin; Wang, Lai; Wang, ZhiweiJournal of Medicinal Chemistry (2019), 62 (17), 7923-7940CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Aberrant activation of Bruton's tyrosine kinase (BTK) plays an important role in pathogenesis of B-cell lymphomas, suggesting that inhibition of BTK is useful in the treatment of hematol. malignancies. The discovery of a more selective on-target covalent BTK inhibitor is of high value. Herein, we disclose the discovery and preclin. characterization of a potent, selective, and irreversible BTK inhibitor as our clin. candidate by using in vitro potency, selectivity, pharmacokinetics (PK), and in vivo pharmacodynamic for prioritizing compds. Compd. BGB-3111 (31a, Zanubrutinib) demonstrates (i) potent activity against BTK and excellent selectivity over other TEC, EGFR and Src family kinases, (ii) desirable ADME, excellent in vivo pharmacodynamic in mice and efficacy in OCI-LY10 xenograft models.
- 8Roskoski, R., Jr. Properties of FDA-approved small molecule protein kinase inhibitors: A 2022 update. Pharmacol. Res. 2022, 175, 106037 DOI: 10.1016/j.phrs.2021.1060378https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XovVahtr4%253D&md5=d12f02086d0761cc387762f1ca06a95bProperties of FDA-approved small molecule protein kinase inhibitors: A 2022 updateRoskoski Jr., RobertPharmacological Research (2022), 175 (), 106037CODEN: PHMREP; ISSN:1043-6618. (Elsevier Ltd.)A review. Owing to the dysregulation of protein kinase activity in many diseases including cancer, this enzyme family has become one of the most important drug targets in the 21st century. There are 68 FDA-approved therapeutic agents that target about two dozen different protein kinases and six of these drugs were approved in 2021. Of the approved drugs, twelve target protein-serine/threonine protein kinases, four are directed against dual specificity protein kinases (MEK1/2), thirteen block nonreceptor protein-tyrosine kinases, and 39 target receptor protein-tyrosine kinases. The data indicate that 58 of these drugs are prescribed for the treatment of neoplasms (49 against solid tumors including breast, lung, and colon, five against nonsolid tumors such as leukemias, and four against both solid and nonsolid tumors: acalabrutinib, ibrutinib, imatinib, and midostaurin). Three drugs (baricitinib, tofacitinib, upadacitinib) are used for the treatment of inflammatory diseases including rheumatoid arthritis. Of the 68 approved drugs, eighteen are used in the treatment of multiple diseases. The following six drugs received FDA approval in 2021 for the treatment of these specified diseases: belumosudil (graft vs. host disease), infigratinib (cholangiocarcinomas), mobocertinib and tepotinib (specific forms of non-small cell lung cancer), tivozanib (renal cell carcinoma), and trilaciclib (to decrease chemotherapy-induced myelosuppression). All of the FDA-approved drugs are orally effective with the exception of netarsudil, temsirolimus, and the newly approved trilaciclib. This review summarizes the physicochem. properties of all 68 FDA-approved small mol. protein kinase inhibitors including lipophilic efficiency and ligand efficiency.
- 9Riely, G. J.; Neal, J. W.; Camidge, D. R.; Spira, A. I.; Piotrowska, Z.; Costa, D. B.; Tsao, A. S.; Patel, J. D.; Gadgeel, S. M.; Bazhenova, L.; Zhu, V. W.; West, H. L.; Mekhail, T.; Gentzler, R. D.; Nguyen, D.; Vincent, S.; Zhang, S.; Lin, J.; Bunn, V.; Jin, S.; Li, S.; Jänne, P. A. Activity and Safety of Mobocertinib (TAK-788) in Previously Treated Non-Small Cell Lung Cancer with EGFR Exon 20 Insertion Mutations from a Phase I/II Trial. Cancer Discovery 2021, 11, 1688– 1699, DOI: 10.1158/2159-8290.Cd-20-15989https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFyntL7E&md5=adf33e0cf63304cada04d78c187afdf9Activity and safety of mobocertinib (TAK-788) in previousiytreated non-small cell lung cancer with EGFR exon 20 insertion mutations from a phase I/ll trialRiely, Gregory J.; Neal, Joel W.; Ross Camidge, D.; Spira, Alexander I.; Piotrowska, Zofia; Costa, Daniel B.; Tsao, Anne S.; Patel, Jyoti D.; Gadgeel, Shirish M.; Bazhenova, Lyudmila; Zhu, Viola W.; West, Howard L.; Mekhail, Tarek; Gentzler, Ryan D.; Nguyen, Danny; Vincent, Sylvie; Zhang, Steven; Lin, Jianchang; Bunn, Veronica; Jin, Shu; Li, Shuanglian; Janne, Pasi A.Cancer Discovery (2021), 11 (7), 1688-1699CODEN: CDAIB2; ISSN:2159-8274. (American Association for Cancer Research)Mobocertinib, an oral epidermal growth factor receptor (EGFR) inhibitor targeting EGFR gene mutations, including exon 20 insertions (EGFRex20ins), in nonsmall cell lung cancer, was evaluated in a phase I/ll dose-escalation/expansion trial (ClinicalTrials. gov NCT02716116). Dose escalation identified 160 mg/d as the recommended phase 2 dose and max. tolerated dose. Among 136 patients treated with 160 mg/d, the most common any-grade treatment-related adverse events (TRAE; >25%) were diarrhea (83%), nausea (43%), rash (33%), and vomiting (26%), with diarrhea (21%) the only grade ≥3 TRAE >5%. Among 28 EGFRex20ins patients treated at 160 mg/d, the investigator-assessed confirmed response rate was 43% (12/28; 95% confidence interval, 24%-63%) with median duration of response of 14 mo (5.0-not reached) and median progression-free survival of 7.3 mo (4.4-15.6). Mobocertinib demonstrated antitumor activity in patients with diverse EGFRex20ins variants with a safety profile consistent with other EGFR inhibitors. SIGNIFICANCE: No oral EGFR-targeted therapies are currently approved for patients with EGFRex- 20ins NSCLC. Mobocertinib demonstrated antitumor activity with manageable toxicity in patients with advanced EGFRex20ins NSCLC in this study, supporting addnl. development of mobocertinib in this patient population.
- 10Singh, J.; Petter, R. C.; Baillie, T. A.; Whitty, A. The resurgence of covalent drugs. Nat. Rev. Drug Discovery 2011, 10, 307– 317, DOI: 10.1038/nrd341010https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXktVGmu7g%253D&md5=2190289081e151416c097be4a5b04460The resurgence of covalent drugsSingh, Juswinder; Petter, Russell C.; Baillie, Thomas A.; Whitty, AdrianNature Reviews Drug Discovery (2011), 10 (4), 307-317CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Covalent drugs haveproved to be successful therapies for various indications, but largely owing to safety concerns, they are rarely considered when initiating a target-directed drug discovery project. There is a need to reassess this important class of drugs, and to reconcile the discordance between the historic success of covalent drugs and the reluctance of most drug discovery teams to include them in their armamentarium. This Review surveys the prevalence and pharmacol. advantages of covalent drugs, discusses how potential risks and challenges may be addressed through innovative design, and presents the broad opportunities provided by targeted covalent inhibitors.
- 11London, N.; Miller, R. M.; Krishnan, S.; Uchida, K.; Irwin, J. J.; Eidam, O.; Gibold, L.; Cimermančič, P.; Bonnet, R.; Shoichet, B. K.; Taunton, J. Covalent docking of large libraries for the discovery of chemical probes. Nat. Chem. Biol. 2014, 10, 1066– 1072, DOI: 10.1038/nchembio.166611https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVSqsbjE&md5=4e5fbeb8f67cb82d464d57069ba0c93aCovalent docking of large libraries for the discovery of chemical probesLondon, Nir; Miller, Rand M.; Krishnan, Shyam; Uchida, Kenji; Irwin, John J.; Eidam, Oliv; Gibold, Lucie; Cimermancic, Peter; Bonnet, Richard; Shoichet, Brian K.; Taunton, JackNature Chemical Biology (2014), 10 (12), 1066-1072CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Chem. probes that form a covalent bond with a protein target often show enhanced selectivity, potency and utility for biol. studies. Despite these advantages, protein-reactive compds. are usually avoided in high-throughput screening campaigns. Here we describe a general method (DOCKovalent) for screening large virtual libraries of electrophilic small mols. We apply this method prospectively to discover reversible covalent fragments that target distinct protein nucleophiles, including the catalytic serine of AmpC β-lactamase and noncatalytic cysteines in RSK2, MSK1 and JAK3 kinases. We identify submicromolar to low-nanomolar hits with high ligand efficiency, cellular activity and selectivity, including what are to our knowledge the first reported reversible covalent inhibitors of JAK3. Crystal structures of inhibitor complexes with AmpC and RSK2 confirm the docking predictions and guide further optimization. As covalent virtual screening may have broad utility for the rapid discovery of chem. probes, we have made the method freely available through an automated web server (http://covalent.docking.org/).
- 12Zhao, Z.; Liu, Q.; Bliven, S.; Xie, L.; Bourne, P. E. Determining Cysteines Available for Covalent Inhibition Across the Human Kinome. J. Med. Chem. 2017, 60, 2879– 2889, DOI: 10.1021/acs.jmedchem.6b0181512https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkvVGjsLg%253D&md5=22ea95ce38eb0836d32c1332dba54fadDetermining Cysteines Available for Covalent Inhibition Across the Human KinomeZhao, Zheng; Liu, Qingsong; Bliven, Spencer; Xie, Lei; Bourne, Philip E.Journal of Medicinal Chemistry (2017), 60 (7), 2879-2889CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Covalently bound protein kinase inhibitors have been frequently designed to target non-catalytic Cys residues at the ATP binding site. Thus, it is important to know if a given Cys residue can form a covalent bond. Here, we combined a function-site interaction fingerprint method and DFT calcns. to det. the potential of Cys residues to form a covalent interaction with an inhibitor. By harnessing the human structural kinome, a comprehensive structure-based binding site Cys dataset was assembled. The orientation of the Cys SH group indicated which Cys residues could potentially form covalent bonds. These covalent inhibitor accessible Cys residues were located within 5 kinase regions (P-loop, roof of pocket, front pocket, catalytic-2 of the catalytic loop and DFG-3 close to the DFG peptide). In an independent test set, these Cys residues covered 95% of covalent kinase inhibitors. Thus, this study provides new insights into Cys reactivity and preference which is important for the prospective development of covalent kinase inhibitors.
- 13Liu, Q.; Sabnis, Y.; Zhao, Z.; Zhang, T.; Buhrlage, S. J.; Jones, L. H.; Gray, N. S. Developing irreversible inhibitors of the protein kinase cysteinome. Chem. Biol. 2013, 20, 146– 159, DOI: 10.1016/j.chembiol.2012.12.00613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtFWitbs%253D&md5=cef80823123bae88362e816a0a638133Developing Irreversible Inhibitors of the Protein Kinase CysteinomeLiu, Qingsong; Sabnis, Yogesh; Zhao, Zheng; Zhang, Tinghu; Buhrlage, Sara J.; Jones, Lyn H.; Gray, Nathanael S.Chemistry & Biology (Oxford, United Kingdom) (2013), 20 (2), 146-159CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)A review. Protein kinases are a large family of approx. 530 highly conserved enzymes that transfer a γ-phosphate group from ATP to a variety of amino acid residues, such as tyrosine, serine, and threonine, that serves as a ubiquitous mechanism for cellular signal transduction. The clin. success of a no. of kinase-directed drugs and the frequent observation of disease causing mutations in protein kinases suggest that a large no. of kinases may represent therapeutically relevant targets. To date, the majority of clin. and preclin. kinase inhibitors are ATP competitive, noncovalent inhibitors that achieve selectivity through recognition of unique features of particular protein kinases. Recently, there has been renewed interest in the development of irreversible inhibitors that form covalent bonds with cysteine or other nucleophilic residues in the ATP-binding pocket. Irreversible kinase inhibitors have a no. of potential advantages including prolonged pharmacodynamics, suitability for rational design, high potency, and ability to validate pharmacol. specificity through mutation of the reactive cysteine residue. Here, we review recent efforts to develop cysteine-targeted irreversible protein kinase inhibitors and discuss their modes of recognizing the ATP-binding pocket and their biol. activity profiles. In addn., we provided an informatics assessment of the potential "kinase cysteinome" and discuss strategies for the efficient development of new covalent inhibitors.
- 14Barf, T.; Covey, T.; Izumi, R.; van de Kar, B.; Gulrajani, M.; van Lith, B.; van Hoek, M.; de Zwart, E.; Mittag, D.; Demont, D.; Verkaik, S.; Krantz, F.; Pearson, P. G.; Ulrich, R.; Kaptein, A. Acalabrutinib (ACP-196): A Covalent Bruton Tyrosine Kinase Inhibitor with a Differentiated Selectivity and In Vivo Potency Profile. J. Pharmacol. Exp. Ther. 2017, 363, 240– 252, DOI: 10.1124/jpet.117.24290914https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtleltrnO&md5=73cbd3a87396abbf8cb20e4361f96f64Acalabrutinib (ACP-196): a covalent Bruton tyrosine kinase inhibitor with a differentiated selectivity and in vivo potency profileBarf, Tjeerd; Covey, Todd; Izumi, Raquel; van de Kar, Bas; Gulrajani, Michael; van Lith, Bart; van Hoek, Maaike; de Zwart, Edwin; Mittag, Diana; Demont, Dennis; Verkaik, Saskia; Krantz, Fanny; Pearson, Paul G.; Ulrich, Roger; Kaptein, AllardJournal of Pharmacology and Experimental Therapeutics (2017), 363 (2), 240-252CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)Several small-mol. Bruton tyrosine kinase (BTK) inhibitors are in development for B cell malignancies and autoimmune disorders, each characterized by distinct potency and selectivity patterns. Herein we describe the pharmacol. characterization of BTK inhibitor acalabrutinib [compd. 1, ACP-196 (4-[8- amino-3-[(2S)-1-but-2-ynoylpyrrolidin-2-yl]imidazo[1,5-a]pyrazin1-yl]-N-(2-pyridyl)benzamide)]. Acalabrutinib possesses a reactive butynamide group that binds covalently to Cys481 in BTK. Relative to the other BTK inhibitors described here, the reduced intrinsic reactivity of acalabrutinib helps to limit inhibition of off-target kinases having cysteine-mediated covalent binding potential. Acalabrutinib demonstrated higher biochem. and cellular selectivity than ibrutinib and spebrutinib (compds. 2 and 3, resp.). Importantly, off-target kinases, such as epidermal growth factor receptor (EGFR) and interleukin 2-inducible T cell kinase (ITK), were not inhibited. Detn. of the inhibitory potential of anti-IgM-induced CD69 expression in human peripheral blood mononuclear cells and whole blood demonstrated that acalabrutinib is a potent functional BTK inhibitor. In vivo evaluation in mice revealed that acalabrutinib is more potent than ibrutinib and spebrutinib. Preclin. and clin. studies showed that the level and duration of BTK occupancy correlates with in vivo efficacy. Evaluation of the pharmacokinetic properties of acalabrutinib in healthy adult volunteers demonstrated rapid absorption and fast elimination. In these healthy individuals, a single oral dose of 100 mg showed approx. 99% median target coverage at 3 and 12 h and around 90% at 24 h in peripheral B cells. In conclusion, acalabrutinib is a BTK inhibitor with key pharmacol. differentiators vs. ibrutinib and spebrutinib and is currently being evaluated in clin. trials.
- 15Cheng, H.; Planken, S. Precedence and Promise of Covalent Inhibitors of EGFR and KRAS for Patients with Non-Small-Cell Lung Cancer. ACS Med. Chem. Lett. 2018, 9, 861– 863, DOI: 10.1021/acsmedchemlett.8b0031115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVagtrjE&md5=0b1f65c9994dc226a5c6d72aa7789071Precedence and Promise of Covalent Inhibitors of EGFR and KRAS for Patients with Non-Small-Cell Lung CancerCheng, Hengmiao; Planken, SimonACS Medicinal Chemistry Letters (2018), 9 (9), 861-863CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (KRAS) oncogenic mutations are leading causes for lung cancer. Extensive drug discovery efforts targeting EGFR have led to the discovery and FDA approval of both reversible and covalent inhibitors. Second and third generation covalent inhibitors for EGFR have also been described, with the latter targeting specific emerging mutations. After decades of extensive effort, KRAS is widely regarded as an intractable therapeutic target; however, recent publications suggest covalent inhibition is a promising strategy to deliver inhibitors of the KRASG12C mutation.
- 16Tomassi, S.; Lategahn, J.; Engel, J.; Keul, M.; Tumbrink, H. L.; Ketzer, J.; Mühlenberg, T.; Baumann, M.; Schultz-Fademrecht, C.; Bauer, S.; Rauh, D. Indazole-Based Covalent Inhibitors To Target Drug-Resistant Epidermal Growth Factor Receptor. J. Med. Chem. 2017, 60, 2361– 2372, DOI: 10.1021/acs.jmedchem.6b0162616https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1WltL0%253D&md5=25e24252dc5a03ec5349359289ebd808Indazole-Based Covalent Inhibitors To Target Drug-Resistant Epidermal Growth Factor ReceptorTomassi, Stefano; Lategahn, Jonas; Engel, Julian; Keul, Marina; Tumbrink, Hannah L.; Ketzer, Julia; Muehlenberg, Thomas; Baumann, Matthias; Schultz-Fademrecht, Carsten; Bauer, Sebastian; Rauh, DanielJournal of Medicinal Chemistry (2017), 60 (6), 2361-2372CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The specific targeting of oncogenic mutant epidermal growth factor receptor (EGFR) is a breakthrough in targeted cancer therapy and marks a drastic change in the treatment of non-small cell lung cancer (NSCLC). The recurrent emergence of resistance to these targeted drugs requires the development of novel chem. entities that efficiently inhibit drug-resistant EGFR. Herein, the authors report the optimization process for a hit compd. that has emerged from a phenotypic screen resulting in indazole-based compds. These inhibitors are conformationally less flexible, target gatekeeper mutated drug resistant EGFR-L858R/T790M and covalently alkylate Cys 797. Western blot anal., as well as characterization of the binding kinetics and kinase selectivity profiling, substantiates the authors' approach of targeting drug-resistant EGFR-L858R/T790M with inhibitors incorporating the indazole as hinge binder.
- 17Planken, S.; Behenna, D. C.; Nair, S. K.; Johnson, T. O.; Nagata, A.; Almaden, C.; Bailey, S.; Ballard, T. E.; Bernier, L.; Cheng, H.; Cho-Schultz, S.; Dalvie, D.; Deal, J. G.; Dinh, D. M.; Edwards, M. P.; Ferre, R. A.; Gajiwala, K. S.; Hemkens, M.; Kania, R. S.; Kath, J. C.; Matthews, J.; Murray, B. W.; Niessen, S.; Orr, S. T.; Pairish, M.; Sach, N. W.; Shen, H.; Shi, M.; Solowiej, J.; Tran, K.; Tseng, E.; Vicini, P.; Wang, Y.; Weinrich, S. L.; Zhou, R.; Zientek, M.; Liu, L.; Luo, Y.; Xin, S.; Zhang, C.; Lafontaine, J. Discovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFR. J. Med. Chem. 2017, 60, 3002– 3019, DOI: 10.1021/acs.jmedchem.6b0189417https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXktFOmtrw%253D&md5=14a6700ed770621bdbd84f7931276f0eDiscovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFRPlanken, Simon; Behenna, Douglas C.; Nair, Sajiv K.; Johnson, Theodore O.; Nagata, Asako; Almaden, Chau; Bailey, Simon; Ballard, T. Eric; Bernier, Louise; Cheng, Hengmiao; Cho-Schultz, Sujin; Dalvie, Deepak; Deal, Judith G.; Dinh, Dac M.; Edwards, Martin P.; Ferre, Rose Ann; Gajiwala, Ketan S.; Hemkens, Michelle; Kania, Robert S.; Kath, John C.; Matthews, Jean; Murray, Brion W.; Niessen, Sherry; Orr, Suvi T. M.; Pairish, Mason; Sach, Neal W.; Shen, Hong; Shi, Manli; Solowiej, James; Tran, Khanh; Tseng, Elaine; Vicini, Paolo; Wang, Yuli; Weinrich, Scott L.; Zhou, Ru; Zientek, Michael; Liu, Longqing; Luo, Yiqin; Xin, Shuibo; Zhang, Chengyi; Lafontaine, JenniferJournal of Medicinal Chemistry (2017), 60 (7), 3002-3019CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mutant epidermal growth factor receptor (EGFR) is a major driver of non-small-cell lung cancer (NSCLC). Marketed first generation inhibitors, such as erlotinib, effect a transient beneficial response in EGFR mutant NSCLC patients before resistance mechanisms render these inhibitors ineffective. Secondary oncogenic EGFR mutations account for approx. 50% of relapses, the most common being the gatekeeper T790M substitution that renders existing therapies ineffective. The discovery of PF-06459988 (1), an irreversible pyrrolopyrimidine inhibitor of EGFR T790M mutants, was recently disclosed. Herein, we describe our continued efforts to achieve potency across EGFR oncogenic mutations and improved kinome selectivity, resulting in the discovery of clin. candidate PF-06747775 (21), which provides potent EGFR activity against the four common mutants (exon 19 deletion (Del), L858R, and double mutants T790M/L858R and T790M/Del), selectivity over wild-type EGFR, and desirable ADME properties. Compd. 21 is currently being evaluated in phase-I clin. trials of mutant EGFR driven NSCLC.
- 18Zapf, C. W.; Gerstenberger, B. S.; Xing, L.; Limburg, D. C.; Anderson, D. R.; Caspers, N.; Han, S.; Aulabaugh, A.; Kurumbail, R.; Shakya, S.; Li, X.; Spaulding, V.; Czerwinski, R. M.; Seth, N.; Medley, Q. G. Covalent inhibitors of interleukin-2 inducible T cell kinase (itk) with nanomolar potency in a whole-blood assay. J. Med. Chem. 2012, 55, 10047– 10063, DOI: 10.1021/jm301190s18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFOlsbnP&md5=7e39a17151dc9493d13439bc48fa83a3Covalent Inhibitors of Interleukin-2 Inducible T Cell Kinase (Itk) with Nanomolar Potency in a Whole-Blood AssayZapf, Christoph W.; Gerstenberger, Brian S.; Xing, Li; Limburg, David C.; Anderson, David R.; Caspers, Nicole; Han, Seungil; Aulabaugh, Ann; Kurumbail, Ravi; Shakya, Subarna; Li, Xin; Spaulding, Vikki; Czerwinski, Robert M.; Seth, Nilufer; Medley, Quintus G.Journal of Medicinal Chemistry (2012), 55 (22), 10047-10063CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We wish to report a strategy that targets interleukin-2 inducible T cell kinase (Itk) with covalent inhibitors. Thus far, covalent inhibition of Itk has not been disclosed in the literature. Structure-based drug design was utilized to achieve low nanomolar potency of the disclosed series even at high ATP concns. Kinetic measurements confirmed an irreversible binding mode with off-rate half-lives exceeding 24 h and moderate on-rates. The analogs are highly potent in a cellular IP1 assay as well as in a human whole-blood (hWB) assay. Despite a half-life of approx. 2 h in resting primary T cells, the covalent inhibition of Itk resulted in functional silencing of the TCR pathway for more than 24 h. This prolonged effect indicates that covalent inhibition is a viable strategy to target the inactivation of Itk.
- 19Ward, R. A.; Colclough, N.; Challinor, M.; Debreczeni, J. E.; Eckersley, K.; Fairley, G.; Feron, L.; Flemington, V.; Graham, M. A.; Greenwood, R.; Hopcroft, P.; Howard, T. D.; James, M.; Jones, C. D.; Jones, C. R.; Renshaw, J.; Roberts, K.; Snow, L.; Tonge, M.; Yeung, K. Structure-Guided Design of Highly Selective and Potent Covalent Inhibitors of ERK1/2. J. Med. Chem. 2015, 58, 4790– 4801, DOI: 10.1021/acs.jmedchem.5b0046619https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXosVChs7c%253D&md5=40b47d8671c966434434f056ac88b3a7Structure-Guided Design of Highly Selective and Potent Covalent Inhibitors of ERK1/2Ward, Richard A.; Colclough, Nicola; Challinor, Mairi; Debreczeni, Judit E.; Eckersley, Kay; Fairley, Gary; Feron, Lyman; Flemington, Vikki; Graham, Mark A.; Greenwood, Ryan; Hopcroft, Philip; Howard, Tina D.; James, Michael; Jones, Clifford D.; Jones, Christopher R.; Renshaw, Jonathan; Roberts, Karen; Snow, Lindsay; Tonge, Michael; Yeung, KayJournal of Medicinal Chemistry (2015), 58 (11), 4790-4801CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The RAS/RAF/MEK/ERK signaling pathway has been targeted with a no. of small mol. inhibitors in oncol. clin. development across multiple disease indications. Importantly, cell lines with acquired resistance to B-RAF and MEK inhibitors have been shown to maintain sensitivity to ERK1/2 inhibition by small mol. inhibitors. There are a no. of selective, noncovalent ERK1/2 inhibitors reported along with the promiscuous hypothemycin (and related analogs) that act via a covalent mechanism of action. This article reports the identification of multiple series of highly selective covalent ERK1/2 inhibitors informed by structure-based drug design (SBDD). As a starting point for these covalent inhibitors, reported ERK1/2 inhibitors and a chem. series identified via high-throughput screening were exploited. These approaches resulted in the identification of selective covalent tool compds. for potential in vitro and in vivo studies to assess the risks and or benefits of targeting this pathway through such a mechanism of action.
- 20Angst, D.; Gessier, F.; Janser, P.; Vulpetti, A.; Wälchli, R.; Beerli, C.; Littlewood-Evans, A.; Dawson, J.; Nuesslein-Hildesheim, B.; Wieczorek, G.; Gutmann, S.; Scheufler, C.; Hinniger, A.; Zimmerlin, A.; Funhoff, E. G.; Pulz, R.; Cenni, B. Discovery of LOU064 (Remibrutinib), a Potent and Highly Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase. J. Med. Chem. 2020, 63, 5102– 5118, DOI: 10.1021/acs.jmedchem.9b0191620https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjs1KiurY%253D&md5=9122b310c294d6071ae7cd73cdb91b3dDiscovery of LOU064 (Remibrutinib), a Potent and Highly Selective Covalent Inhibitor of Bruton's Tyrosine KinaseAngst, Daniela; Gessier, Francois; Janser, Philipp; Vulpetti, Anna; Walchli, Rudolf; Beerli, Christian; Littlewood-Evans, Amanda; Dawson, Janet; Nuesslein-Hildesheim, Barbara; Wieczorek, Grazyna; Gutmann, Sascha; Scheufler, Clemens; Hinniger, Alexandra; Zimmerlin, Alfred; Funhoff, Enrico G.; Pulz, Robert; Cenni, BrunoJournal of Medicinal Chemistry (2020), 63 (10), 5102-5118CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bruton's tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a central role in immunity and is considered an attractive target for treating autoimmune diseases. The use of currently marketed covalent BTK inhibitors is limited to oncol. indications based on their suboptimal kinase selectivity. We describe the discovery and preclin. profile of LOU064 (remibrutinib, 25), a potent, highly selective covalent BTK inhibitor. LOU064 exhibits an exquisite kinase selectivity due to binding to an inactive conformation of BTK and has the potential for a best-in-class covalent BTK inhibitor for the treatment of autoimmune diseases. It demonstrates potent in vivo target occupancy with an EC90 of 1.6 mg/kg and dose-dependent efficacy in rat collagen-induced arthritis. LOU064 is currently being tested in phase 2 clin. studies for chronic spontaneous urticaria and Sjoegren's syndrome.
- 21Gurbani, D.; Du, G.; Henning, N. J.; Rao, S.; Bera, A. K.; Zhang, T.; Gray, N. S.; Westover, K. D. Structure and Characterization of a Covalent Inhibitor of Src Kinase. Front. Mol. Biosci. 2020, 7, 81 DOI: 10.3389/fmolb.2020.0008121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFKksLbF&md5=0b1b4bbea40bb1cfaad8fb8fbbff45e8Structure and characterization of a covalent inhibitor of Src kinaseGurbani, Deepak; Du, Guangyan; Henning, Nathaniel J.; Rao, Suman; Bera, Asim K.; Zhang, Tinghu; Gray, Nathanael S.; Westover, Kenneth D.Frontiers in Molecular Biosciences (2020), 7 (), 00081CODEN: FMBRBS; ISSN:2296-889X. (Frontiers Media S.A.)Unregulated Src activity promotes malignant processes in cancer, but no Src-directed targeted therapies are used clin., possibly because early Src inhibitors produce off-target effects leading to toxicity. Improved selective Src inhibitors may enable Src-directed therapies. Previously, we reported an irreversible Src inhibitor, DGY-06-116, based on the hybridization of dasatinib and a promiscuous covalent kinase probe SM1-71. Here, we report biochem. and biophys. characterization of this compd. An x-ray co-crystal structure of DGY-06-116: Src shows a covalent interaction with the kinase p-loop and occupancy of the back hydrophobic kinase pocket, explaining its high potency, and selectivity. However, a reversible analog also shows similar potency. Kinetic anal. shows a slow inactivation rate compared to other clin. approved covalent kinase inhibitors, consistent with a need for p-loop movement prior to covalent bond formation. Overall, these results suggest that a strong reversible interaction is required to allow sufficient time for the covalent reaction to occur. Further optimization of the covalent linker may improve the kinetics of covalent bond formation.
- 22Abdeldayem, A.; Raouf, Y. S.; Constantinescu, S. N.; Moriggl, R.; Gunning, P. T. Advances in covalent kinase inhibitors. Chem. Soc. Rev. 2020, 49, 2617– 2687, DOI: 10.1039/c9cs00720b22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvVygsr8%253D&md5=88c6e097cb0b3619a53ceb1e03d19971Advances in covalent kinase inhibitorsAbdeldayem, Ayah; Raouf, Yasir S.; Constantinescu, Stefan N.; Moriggl, Richard; Gunning, Patrick T.Chemical Society Reviews (2020), 49 (9), 2617-2687CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)Over the past decade, covalent kinase inhibitors (CKI) have seen a resurgence in drug discovery. Covalency affords a unique set of advantages as well as challenges relative to their non-covalent counterpart. After reversible protein target recognition and binding, covalent inhibitors irreversibly modify a proximal nucleophilic residue on the protein via reaction with an electrophile. To date, the acrylamide group remains the predominantly employed electrophile in CKI development, with its incorporation in the majority of clin. candidates and FDA approved covalent therapies. Nonetheless, in recent years considerable efforts have ensued to characterize alternative electrophiles that exhibit irreversible or reversibly covalent binding mechanisms towards cysteine thiols and other amino acids. This review article provides a comprehensive overview of CKIs reported in the literature over a decade period, 2007-2018. Emphasis is placed on the rationale behind warhead choice, optimization approach, and inhibitor design. Current FDA approved CKIs are also highlighted, in addn. to a detailed anal. of the common trends and themes obsd. within the listed data set.
- 23Yan, G.; Zhong, X.; Pu, C.; Yue, L.; Shan, H.; Lan, S.; Zhou, M.; Hou, X.; Yang, J.; Li, D.; Fan, S.; Li, R. Targeting Cysteine Located Outside the Active Site: An Effective Strategy for Covalent ALKi Design. J. Med. Chem. 2021, 64, 1558– 1569, DOI: 10.1021/acs.jmedchem.0c0170723https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFWisr4%253D&md5=216a6fc20ae93c196947e73e9c933e85Targeting Cysteine Located Outside the Active Site: An Effective Strategy for Covalent ALKi DesignYan, Guoyi; Zhong, Xinxin; Pu, Chunlan; Yue, Lin; Shan, Huifang; Lan, Suke; Zhou, Meng; Hou, Xueyan; Yang, Jie; Li, Deyu; Fan, Shilong; Li, RuiJournal of Medicinal Chemistry (2021), 64 (3), 1558-1569CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Potent inhibitors of ALK are highly desired because of the occurrence of drug resistance. We herein firstly report the development of a rationally designed inhibitor, Con B-1, which can covalently bind to Cys1259, a cysteine located outside the ALK active site by linking a warhead with Ceritinib through a 2,2'-Oxybis(ethylamine) linker. The in vitro and in vivo assays showed ConB-1 is a potent selective ALKi with low toxicity to normal cells. In addn., the mol. showed significant improvement of anticancer activities and potential antidrug resistant activity compared with Ceritinib, demonstrating the covalent inhibitor of ALK can be a promising drug candidate for the treatment of NSCLC. This work may provide a novel perspective on the design of covalent inhibitors.
- 24Jiang, B.; Jiang, J.; Kaltheuner, I. H.; Iniguez, A. B.; Anand, K.; Ferguson, F. M.; Ficarro, S. B.; Seong, B. K. A.; Greifenberg, A. K.; Dust, S.; Kwiatkowski, N. P.; Marto, J. A.; Stegmaier, K.; Zhang, T.; Geyer, M.; Gray, N. S. Structure-activity relationship study of THZ531 derivatives enables the discovery of BSJ-01-175 as a dual CDK12/13 covalent inhibitor with efficacy in Ewing sarcoma. Eur. J. Med. Chem. 2021, 221, 113481 DOI: 10.1016/j.ejmech.2021.11348124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVWqtb3N&md5=d6e8546a93cb97657e9cb24bb86dc5bcStructure-activity relationship study of THZ531 derivatives enables the discovery of BSJ-01-175 as a dual CDK12/13 covalent inhibitor with efficacy in Ewing sarcomaJiang, Baishan; Jiang, Jie; Kaltheuner, Ines H.; Iniguez, Amanda Balboni; Anand, Kanchan; Ferguson, Fleur M.; Ficarro, Scott B.; Seong, Bo Kyung Alex; Greifenberg, Ann Katrin; Dust, Sofia; Kwiatkowski, Nicholas P.; Marto, Jarrod A.; Stegmaier, Kimberly; Zhang, Tinghu; Geyer, Matthias; Gray, Nathanael S.European Journal of Medicinal Chemistry (2021), 221 (), 113481CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Development of inhibitors targeting CDK12/13 is of increasing interest as a potential therapy for cancers as these compds. inhibit transcription of DNA damage response (DDR) genes. We previously described THZ531, a covalent inhibitor with selectivity for CDK12/13. In order to elucidate structure-activity relationship (SAR), we have undertaken a medicinal chem. campaign and established a focused library of THZ531 analogs. Among these analogs, BSJ-01-175 demonstrates exquisite selectivity, potent inhibition of RNA polymerase II phosphorylation, and downregulation of CDK12-targeted genes in cancer cells. A 3.0 Å co-crystal structure with CDK12/CycK provides a structural rational for selective targeting of Cys1039 located in a C-terminal extension from the kinase domain. With moderate pharmacokinetic properties, BSJ-01-175 exhibits efficacy against an Ewing sarcoma tumor growth in a patient-derived xenograft (PDX) mouse model following 10 mg/kg once a day, i.p. administration. Taken together, BSJ-01-175 represents the first selective CDK12/13 covalent inhibitor with in vivo efficacy reported to date.
- 25Zhang, T.; Kwiatkowski, N.; Olson, C. M.; Dixon-Clarke, S. E.; Abraham, B. J.; Greifenberg, A. K.; Ficarro, S. B.; Elkins, J. M.; Liang, Y.; Hannett, N. M.; Manz, T.; Hao, M.; Bartkowiak, B.; Greenleaf, A. L.; Marto, J. A.; Geyer, M.; Bullock, A. N.; Young, R. A.; Gray, N. S. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat. Chem. Biol. 2016, 12, 876– 884, DOI: 10.1038/nchembio.216625https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlyqsbbJ&md5=5ac265105d1654e129c3c1325572f389Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitorsZhang, Tinghu; Kwiatkowski, Nicholas; Olson, Calla M.; Dixon-Clarke, Sarah E.; Abraham, Brian J.; Greifenberg, Ann K.; Ficarro, Scott B.; Elkins, Jonathan M.; Liang, Yanke; Hannett, Nancy M.; Manz, Theresa; Hao, Mingfeng; Bartkowiak, Bartlomiej; Greenleaf, Arno L.; Marto, Jarrod A.; Geyer, Matthias; Bullock, Alex N.; Young, Richard A.; Gray, Nathanael S.Nature Chemical Biology (2016), 12 (10), 876-884CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Cyclin-dependent kinases 12 and 13 (CDK12 and CDK13) play crit. roles in the regulation of gene transcription. However, the absence of CDK12 and CDK13 inhibitors has hindered the ability to investigate the consequences of their inhibition in healthy cells and cancer cells. Here we describe the rational design of a first-in-class CDK12 and CDK13 covalent inhibitor, THZ531. Co-crystn. of THZ531 with CDK12-cyclin K indicates that THZ531 irreversibly targets a cysteine located outside the kinase domain. THZ531 causes a loss of gene expression with concurrent loss of elongating and hyperphosphorylated RNA polymerase II. In particular, THZ531 substantially decreases the expression of DNA damage response genes and key super-enhancer-assocd. transcription factor genes. Coincident with transcriptional perturbation, THZ531 dramatically induced apoptotic cell death. Small mols. capable of specifically targeting CDK12 and CDK13 may thus help identify cancer subtypes that are particularly dependent on their kinase activities.
- 26Kwiatkowski, N.; Zhang, T.; Rahl, P. B.; Abraham, B. J.; Reddy, J.; Ficarro, S. B.; Dastur, A.; Amzallag, A.; Ramaswamy, S.; Tesar, B.; Jenkins, C. E.; Hannett, N. M.; McMillin, D.; Sanda, T.; Sim, T.; Kim, N. D.; Look, T.; Mitsiades, C. S.; Weng, A. P.; Brown, J. R.; Benes, C. H.; Marto, J. A.; Young, R. A.; Gray, N. S. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014, 511, 616– 620, DOI: 10.1038/nature1339326https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1ChurnF&md5=895a28fb536663a31e7558fe79ce628bTargeting transcription regulation in cancer with a covalent CDK7 inhibitorKwiatkowski, Nicholas; Zhang, Tinghu; Rahl, Peter B.; Abraham, Brian J.; Reddy, Jessica; Ficarro, Scott B.; Dastur, Anahita; Amzallag, Arnaud; Ramaswamy, Sridhar; Tesar, Bethany; Jenkins, Catherine E.; Hannett, Nancy M.; McMillin, Douglas; Sanda, Takaomi; Sim, Taebo; Kim, Nam Doo; Look, Thomas; Mitsiades, Constantine S.; Weng, Andrew P.; Brown, Jennifer R.; Benes, Cyril H.; Marto, Jarrod A.; Young, Richard A.; Gray, Nathanael S.Nature (London, United Kingdom) (2014), 511 (7511), 616-620CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Tumor oncogenes include transcription factors that co-opt the general transcriptional machinery to sustain the oncogenic state, but direct pharmacol. inhibition of transcription factors has so far proven difficult. However, the transcriptional machinery contains various enzymic cofactors that can be targeted for the development of new therapeutic candidates, including cyclin-dependent kinases (CDKs). Here the authors present the discovery and characterization of a covalent CDK7 inhibitor, THZ1, which has the unprecedented ability to target a remote cysteine residue located outside of the canonical kinase domain, providing an unanticipated means of achieving selectivity for CDK7. Cancer cell-line profiling indicates that a subset of cancer cell lines, including human T-cell acute lymphoblastic leukemia (T-ALL), have exceptional sensitivity to THZ1. Genome-wide anal. in Jurkat T-ALL cells shows that THZ1 disproportionally affects transcription of RUNX1 and suggests that sensitivity to THZ1 may be due to vulnerability conferred by the RUNX1 super-enhancer and the key role of RUNX1 in the core transcriptional regulatory circuitry of these tumor cells. Pharmacol. modulation of CDK7 kinase activity may thus provide an approach to identify and treat tumor types that are dependent on transcription for maintenance of the oncogenic state.
- 27Nacht, M.; Qiao, L.; Sheets, M. P.; St Martin, T.; Labenski, M.; Mazdiyasni, H.; Karp, R.; Zhu, Z.; Chaturvedi, P.; Bhavsar, D.; Niu, D.; Westlin, W.; Petter, R. C.; Medikonda, A. P.; Singh, J. Discovery of a Potent and Isoform-Selective Targeted Covalent Inhibitor of the Lipid Kinase PI3Kα. J. Med. Chem. 2013, 56, 712– 721, DOI: 10.1021/jm300874527https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWjsr4%253D&md5=42bf6d9221fdb9eafe010064807d22b9Discovery of a Potent and Isoform-Selective Targeted Covalent Inhibitor of the Lipid Kinase PI3KαNacht, Mariana; Qiao, Lixin; Sheets, Michael P.; St. Martin, Thia; Labenski, Matthew; Mazdiyasni, Hormoz; Karp, Russell; Zhu, Zhendong; Chaturvedi, Prasoon; Bhavsar, Deepa; Niu, Deqiang; Westlin, William; Petter, Russell C.; Medikonda, Aravind Prasad; Singh, JuswinderJournal of Medicinal Chemistry (2013), 56 (3), 712-721CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallog., that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kβ, -γ, and -δ are not covalently modified. 3 is able to potently (EC50 < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI50 < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biol. impact of selectively targeting PI3Kα.
- 28Wymann, M. P.; Schneiter, R. Lipid signalling in disease. Nat. Rev. Mol. Cell Biol. 2008, 9, 162– 176, DOI: 10.1038/nrm233528https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOgug%253D%253D&md5=c96aabfdfa8e93c9e98386f1a407de85Lipid signalling in diseaseWymann, Matthias P.; Schneiter, RogerNature Reviews Molecular Cell Biology (2008), 9 (2), 162-176CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Signaling lipids such as eicosanoids, phosphoinositides, sphingolipids and fatty acids control important cellular processes, including cell proliferation, apoptosis, metab. and migration. Extracellular signals from cytokines, growth factors and nutrients control the activity of a key set of lipid-modifying enzymes: phospholipases, prostaglandin synthase, 5-lipoxygenase, phosphoinositide 3-kinase, sphingosine kinase and sphingomyelinase. These enzymes and their downstream targets constitute a complex lipid signaling network with multiple nodes of interaction and cross-regulation. Imbalances in this network contribute to the pathogenesis of human disease. Although the function of a particular signaling lipid is traditionally studied in isolation, this review attempts a more integrated overview of the key role of these signaling lipids in inflammation, cancer and metabolic disease, and discusses emerging strategies for therapeutic intervention.
- 29Fruman, D. A.; Rommel, C. PI3K and cancer: lessons, challenges and opportunities. Nat. Rev. Drug Discovery 2014, 13, 140– 156, DOI: 10.1038/nrd420429https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1SnsLc%253D&md5=c4f6b4b9f9fcacd6ef55eb17d3a4f425PI3K and cancer: lessons, challenges and opportunitiesFruman, David A.; Rommel, ChristianNature Reviews Drug Discovery (2014), 13 (2), 140-156CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The central role of phosphoinositide 3-kinase (PI3K) activation in tumor cell biol. has prompted a sizeable effort to target PI3K and/or downstream kinases such as AKT and mammalian target of rapamycin (mTOR) in cancer. However, emerging clin. data show limited single-agent activity of inhibitors targeting PI3K, AKT or mTOR at tolerated doses. One exception is the response to PI3Kδ inhibitors in chronic lymphocytic leukemia, where a combination of cell-intrinsic and -extrinsic activities drive efficacy. Here, we review key challenges and opportunities for the clin. development of inhibitors targeting the PI3K-AKT-mTOR pathway. Through a greater focus on patient selection, increased understanding of immune modulation and strategic application of rational combinations, it should be possible to realize the potential of this promising class of targeted anticancer agents.
- 30Samuels, Y.; Wang, Z.; Bardelli, A.; Silliman, N.; Ptak, J.; Szabo, S.; Yan, H.; Gazdar, A.; Powell, S. M.; Riggins, G. J.; Willson, J. K.; Markowitz, S.; Kinzler, K. W.; Vogelstein, B.; Velculescu, V. E. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004, 304, 554, DOI: 10.1126/science.109650230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjsVGmsbk%253D&md5=be8d63bf0c909971c3b7a5f475ce8fabBrevia: High frequency of mutations of the PIK3Ca gene in human cancersSamuels, Yardena; Wang, Zhenghe; Bardelli, Alberto; Silliman, Natalie; Ptak, Janine; Szabo, Steve; Yan, Hai; Gazdar, Adi; Powell, Steven M.; Riggins, Gregory J.; Willson, James K. V.; Markowitz, Sanford; Kinzler, Kenneth W.; Vogelstein, Bert; Velculescu, Victor E.Science (Washington, DC, United States) (2004), 304 (5670), 554CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)There is no expanded citation for this reference.
- 31Vanhaesebroeck, B.; Perry, M. W. D.; Brown, J. R.; André, F.; Okkenhaug, K. PI3K inhibitors are finally coming of age. Nat. Rev. Drug Discovery 2021, 20, 741– 769, DOI: 10.1038/s41573-021-00209-131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlaitrjL&md5=6186f01192ecbd4d16bf820b0c3259c0Phosphoinositide 3-kinase inhibitors are finally coming of ageVanhaesebroeck, Bart; Perry, Matthew W. D.; Brown, Jennifer R.; Andre, Fabrice; Okkenhaug, KlausNature Reviews Drug Discovery (2021), 20 (10), 741-769CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)A review. Overactive phosphoinositide 3-kinase (PI3K) in cancer and immune dysregulation has spurred extensive efforts to develop therapeutic PI3K inhibitors. Although progress has been hampered by issues such as poor drug tolerance and drug resistance, several PI3K inhibitors have now received regulatory approval-the PI3Kα isoform-selective inhibitor alpelisib for the treatment of breast cancer and inhibitors mainly aimed at the leukocyte-enriched PI3Kδ in B cell malignancies. In addn. to targeting cancer cell-intrinsic PI3K activity, emerging evidence highlights the potential of PI3K inhibitors in cancer immunotherapy. This Review summarizes key discoveries that aid the clin. translation of PI3Kα and PI3Kδ inhibitors, highlighting lessons learnt and future opportunities.
- 32Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26 DOI: 10.1186/s12943-019-0954-x32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cflt1Kluw%253D%253D&md5=4267e4df49544ebe11ab4255e69e5bbeTargeting PI3K in cancer: mechanisms and advances in clinical trialsYang Jing; Nie Ji; Ma Xuelei; Wei Yuquan; Peng Yong; Wei XiaweiMolecular cancer (2019), 18 (1), 26 ISSN:.Phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling is one of the most important intracellular pathways, which can be considered as a master regulator for cancer. Enormous efforts have been dedicated to the development of drugs targeting PI3K signaling, many of which are currently employed in clinical trials evaluation, and it is becoming increasingly clear that PI3K inhibitors are effective in inhibiting tumor progression. PI3K inhibitors are subdivided into dual PI3K/mTOR inhibitors, pan-PI3K inhibitors and isoform-specific inhibitors. In this review, we performed a critical review to summarize the role of the PI3K pathway in tumor development, recent PI3K inhibitors development based on clinical trials, and the mechanisms of resistance to PI3K inhibition.
- 33Massacesi, C.; Di Tomaso, E.; Urban, P.; Germa, C.; Quadt, C.; Trandafir, L.; Aimone, P.; Fretault, N.; Dharan, B.; Tavorath, R.; Hirawat, S. PI3K inhibitors as new cancer therapeutics: implications for clinical trial design. OncoTargets Ther. 2016, 9, 203– 210, DOI: 10.2147/OTT.S8996733https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlvFyqsr4%253D&md5=aa0e646fb23513ab825d1d0aed50dd16PI3K inhibitors as new cancer therapeutics: implications for clinical trial designMassacesi, Cristian; Di Tomaso, Emmanuelle; Urban, Patrick; Germa, Caroline; Quadt, Cornelia; Trandafir, Lucia; Aimone, Paola; Fretault, Nathalie; Dharan, Bharani; Tavorath, Ranjana; Hirawat, SamitOncoTargets and Therapy (2016), 9 (), 203-210CODEN: OTNHAZ; ISSN:1178-6930. (Dove Medical Press Ltd.)The PI3K-AKT-mTOR pathway is frequently activated in cancer. PI3K inhibitors, including the pan-PI3K inhibitor buparlisib (BKM120) and the PI3Ka-selective inhibitor alpelisib (BYL719), currently in clin. development by Novartis Oncol., may therefore be effective as anticancer agents. Early clin. studies with PI3K inhibitors have demonstrated preliminary antitumor activity and acceptable safety profiles. However, a no. of unanswered questions regarding PI3K inhibition in cancer remain, including: what is the best approach for different tumor types, and which biomarkers will accurately identify the patient populations most likely to benefit from specific PI3K inhibitors. This review summarizes the strategies being employed by Novartis Oncol. to help maximize the benefits of clin. studies with buparlisib and alpelisib, including stratification according to PI3K pathway activation status, selective enrollment/target enrichment (where patients with PI3K pathway-activated tumors are specifically recruited), nonselective enrollment with mandatory tissue collection, and enrollment of patients who have progressed on previous targeted agents, such as mTOR inhibitors or endocrine therapy. An overview of Novartis-sponsored and Novartis-supported trials that are utilizing these approaches in a range of cancer types, including breast cancer, head and neck squamous cell carcinoma, non-small cell lung carcinoma, lymphoma, and glioblastoma multiforme, is also described.
- 34Mishra, R.; Patel, H.; Alanazi, S.; Kilroy, M. K.; Garrett, J. T. PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects. Int. J. Mol. Sci. 2021, 22, 3464 DOI: 10.3390/ijms2207346434https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXht1Gku7bE&md5=b3551463f09f9086323dfac7f9804fc1PI3K inhibitors in cancer: clinical implications and adverse effectsMishra, Rosalin; Patel, Hima; Alanazi, Samar; Kilroy, Mary Kate; Garrett, Joan T.International Journal of Molecular Sciences (2021), 22 (7), 3464CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)A review. The phospatidylinositol-3 kinase (PI3K) pathway is a crucial intracellular signaling pathway which is mutated or amplified in a wide variety of cancers including breast, gastric, ovarian, colorectal, prostate, glioblastoma and endometrial cancers. PI3K signaling plays an important role in cancer cell survival, angiogenesis and metastasis, making it a promising therapeutic target. There are several ongoing and completed clin. trials involving PI3K inhibitors (pan, isoform-specific and dual PI3K/mTOR) with the goal to find efficient PI3K inhibitors that could overcome resistance to current therapies. This review focuses on the current landscape of various PI3K inhibitors either as monotherapy or in combination therapies and the treatment outcomes involved in various phases of clin. trials in different cancer types. There is a discussion of the drug-related toxicities, challenges assocd. with these PI3K inhibitors and the adverse events leading to treatment failure. In addn., novel PI3K drugs that have potential to be translated in the clinic are highlighted.
- 35Fritsch, C.; Huang, A.; Chatenay-Rivauday, C.; Schnell, C.; Reddy, A.; Liu, M.; Kauffmann, A.; Guthy, D.; Erdmann, D.; De Pover, A.; Furet, P.; Gao, H.; Ferretti, S.; Wang, Y.; Trappe, J.; Brachmann, S. M.; Maira, S. M.; Wilson, C.; Boehm, M.; Garcia-Echeverria, C.; Chene, P.; Wiesmann, M.; Cozens, R.; Lehar, J.; Schlegel, R.; Caravatti, G.; Hofmann, F.; Sellers, W. R. Characterization of the novel and specific PI3Kα inhibitor NVP-BYL719 and development of the patient stratification strategy for clinical trials. Mol. Cancer Ther. 2014, 13, 1117– 1129, DOI: 10.1158/1535-7163.Mct-13-086535https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnsFWjsb4%253D&md5=58d9132052dd9d430361021b0f877475Characterization of the Novel and Specific PI3Kα Inhibitor NVP-BYL719 and Development of the Patient Stratification Strategy for Clinical TrialsFritsch, Christine; Huang, Alan; Chatenay-Rivauday, Christian; Schnell, Christian; Reddy, Anupama; Liu, Manway; Kauffmann, Audrey; Guthy, Daniel; Erdmann, Dirk; De Pover, Alain; Furet, Pascal; Gao, Hui; Ferretti, Stephane; Wang, Youzhen; Trappe, Joerg; Brachmann, Saskia M.; Maira, Sauveur-Michel; Wilson, Christopher; Boehm, Markus; Garcia-Echeverria, Carlos; Chene, Patrick; Wiesmann, Marion; Cozens, Robert; Lehar, Joseph; Schlegel, Robert; Caravatti, Giorgio; Hofmann, Francesco; Sellers, William R.Molecular Cancer Therapeutics (2014), 13 (5), 1117-1129CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Somatic PIK3CA mutations are frequently found in solid tumors, raising the hypothesis that selective inhibition of PI3Kα may have robust efficacy in PIK3CA-mutant cancers while sparing patients the side-effects assocd. with broader inhibition of the class I phosphoinositide 3-kinase (PI3K) family. Here, we report the biol. properties of the 2-aminothiazole deriv. NVP-BYL719, a selective inhibitor of PI3Kα and its most common oncogenic mutant forms. The compd. selectivity combined with excellent drug-like properties translates to dose- and time-dependent inhibition of PI3Kα signaling in vivo, resulting in robust therapeutic efficacy and tolerability in PIK3CA-dependent tumors. Novel targeted therapeutics such as NVP-BYL719, designed to modulate aberrant functions elicited by cancer-specific genetic alterations upon which the disease depends, require well-defined patient stratification strategies in order to maximize their therapeutic impact and benefit for the patients. Here, we also describe the application of the Cancer Cell Line Encyclopedia as a preclin. platform to refine the patient stratification strategy for NVP-BYL719 and found that PIK3CA mutation was the foremost pos. predictor of sensitivity while revealing addnl. pos. and neg. assocns. such as PIK3CA amplification and PTEN mutation, resp. These patient selection determinants are being assayed in the ongoing NVP-BYL719 clin. trials. Mol Cancer Ther; 13(5); 1117-29. ©2014 AACR.
- 36Markham, A. Alpelisib: First Global Approval. Drugs 2019, 79, 1249– 1253, DOI: 10.1007/s40265-019-01161-636https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlCiur%252FN&md5=4d9e9d1e3742a9bce160ca2c7987ce6cAlpelisib: First Global ApprovalMarkham, AnthonyDrugs (2019), 79 (11), 1249-1253CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. Alpelisib (Piqray)-an orally available phosphatidylinositol 3-kinase (PI3K) inhibitor with specific activity against PI3K alpha (PI3Kα)-is being developed by Novartis for the treatment of breast cancer. Alpelisib has demonstrated efficacy in combination with fulvestrant as treatment for hormone receptor (HR)-pos., human epidermal growth factor receptor-2 (HER2)-neg. breast cancer in patients with a PIK3CA mutation and was recently approved for this indication in the USA. This article summarizes the milestones in the development of alpelisib leading to this first approval.
- 37Narayan, P.; Prowell, T. M.; Gao, J. J.; Fernandes, L. L.; Li, E.; Jiang, X.; Qiu, J.; Fan, J.; Song, P.; Yu, J.; Zhang, X.; King-Kallimanis, B. L.; Chen, W.; Ricks, T. K.; Gong, Y.; Wang, X.; Windsor, K.; Rhieu, S. Y.; Geiser, G.; Banerjee, A.; Chen, X.; Reyes Turcu, F.; Chatterjee, D. K.; Pathak, A.; Seidman, J.; Ghosh, S.; Philip, R.; Goldberg, K. B.; Kluetz, P. G.; Tang, S.; Amiri-Kordestani, L.; Theoret, M. R.; Pazdur, R.; Beaver, J. A. FDA Approval Summary: Alpelisib Plus Fulvestrant for Patients with HR-positive, HER2-negative, PIK3CA-mutated, Advanced or Metastatic Breast Cancer. Clin. Cancer Res. 2021, 27, 1842– 1849, DOI: 10.1158/1078-0432.Ccr-20-365237https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFGjtL%252FO&md5=3220d285c0f3c135cf229b67554295d7FDA approval summary: alpelisib plus fulvestrant for patients with HR-positive, HER2-negative, PIK3CA-mutated, advanced or metastatic breast cancerNarayan, Preeti; Prowell, Tatiana M.; Gao, Jennifer J.; Fernandes, Laura L.; Li, Emily; Jiang, Xiling; Qiu, Junshan; Fan, Jianghong; Song, Pengfei; Yu, Jingyu; Zhang, Xinyuan; King-Kallimanis, Bellinda L.; Chen, Wei; Ricks, Tiffany K.; Gong, Yutao; Wang, Xing; Windsor, Katherine; Rhieu, Steve Y.; Geiser, Gerlie; Banerjee, Anamitro; Chen, Xiaohong; Turcu, Francisca Reyes; Chatterjee, Deb K.; Pathak, Anand; Seidman, Jeffrey; Ghosh, Soma; Philip, Reena; Goldberg, Kirsten B.; Kluetz, Paul G.; Tang, Shenghui; Amiri-Kordestani, Laleh; Theoret, Marc R.; Pazdur, Richard; Beaver, Julia A.Clinical Cancer Research (2021), 27 (7), 1842-1849CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)On May 24, 2019, the FDA granted regular approval to alpelisib in combination with fulvestrant for postmenopausal women, and men, with hormone receptor (HR)-pos., HER2-neg., phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA)-mutated, advanced or metastatic breast cancer as detected by an FDA-approved test following progression on or after an endocrine-based regimen. Approval was based on the SOLAR-1 study, a randomized, double-blind, placebo-controlled trial of alpelisib plus fulvestrant vs. placebo plus fulvestrant. The primary endpoint was investigator-assessed progression-free survival (PFS) per RECIST v1.1 in the cohort of trial participants whose tumors had a PIK3CA mutation. The estd. median PFS by investigator assessment in the alpelisib plus fulvestrant arm was 11 mo [95% confidence interval (CI), 7.5-14.5] compared with 5.7 mo (95% CI, 3.7-7.4) in the placebo plus fulvestrant arm (HR, 0.65; 95% CI, 0.50-0.85; two-sided P = 0.001). The median overall survival was not yet reached for the alpelisib plus fulvestrant arm (95% CI, 28.1-NE) and was 26.9 mo (95% CI, 21.9-NE) for the fulvestrant control arm. No PFS benefit was obsd. in trial participants whose tumors did not have a PIK3CA mutation (HR, 0.85; 95% CI, 0.58-1.25). The most common adverse reactions, including lab. abnormalities, on the alpelisib plus fulvestrant arm were increased glucose, increased creatinine, diarrhea, rash, decreased lymphocyte count, increased gamma glutamyl transferase, nausea, increased alanine aminotransferase, fatigue, decreased Hb, increased lipase, decreased appetite, stomatitis, vomiting, decreased wt., decreased calcium, decreased glucose, prolonged activated partial thromboplastin time, and alopecia.
- 38André, F.; Ciruelos, E. M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I. A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y. S.; Inoue, K.; Pápai, Z.; Takahashi, M.; Ghaznawi, F.; Mills, D.; Kaper, M.; Miller, M.; Conte, P. F.; Iwata, H.; Rugo, H. S. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1. Ann. Oncol. 2021, 32, 208– 217, DOI: 10.1016/j.annonc.2020.11.01138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvVKrtbc%253D&md5=11ec0f2b91cad4b709edfb314de7e577Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1Andre, F.; Ciruelos, E. M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I. A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y.-S.; Inoue, K.; Papai, Z.; Takahashi, M.; Ghaznawi, F.; Mills, D.; Kaper, M.; Miller, M.; Conte, P. F.; Iwata, H.; Rugo, H. S.Annals of Oncology (2021), 32 (2), 208-217CODEN: AONNAY; ISSN:1569-8041. (Elsevier Ltd.)Activation of the phosphatidylinositol-3-kinase (PI3K) pathway via PIK3CA mutations occurs in 28%-46% of hormone receptor-pos. (HR+), human epidermal growth factor receptor-2-neg. (HER2-) advanced breast cancers (ABCs) and is assocd. with poor prognosis. The SOLAR-1 trial showed that the addn. of alpelisib to fulvestrant treatment provided statistically significant and clin. meaningful progression-free survival (PFS) benefit in PIK3CA-mutated, HR+, HER2- ABC. Men and postmenopausal women with HR+, HER2- ABC whose disease progressed on or after aromatase inhibitor (AI) were randomized 1 : 1 to receive alpelisib (300 mg/day) plus fulvestrant (500 mg every 28 days and once on day 15) or placebo plus fulvestrant. Overall survival (OS) in the PIK3CA-mutant cohort was evaluated by Kaplan-Meier methodol. and a one-sided stratified log-rank test was carried out with an O'Brien-Fleming efficacy boundary of P ≤ 0.0161. In the PIK3CA-mutated cohort (n = 341), median OS [95% confidence interval (CI)] was 39.3 mo (34.1-44.9) for alpelisib-fulvestrant and 31.4 mo (26.8-41.3) for placebo-fulvestrant [hazard ratio (HR) = 0.86 (95% CI, 0.64-1.15; P = 0.15)]. OS results did not cross the prespecified efficacy boundary. Median OS (95% CI) in patients with lung and/or liver metastases was 37.2 mo (28.7-43.6) and 22.8 mo (19.0-26.8) in the alpelisib-fulvestrant and placebo-fulvestrant arms, resp. [HR = 0.68 (0.46-1.00)]. Median times to chemotherapy (95% CI) for the alpelisib-fulvestrant and placebo-fulvestrant arms were 23.3 mo (15.2-28.4) and 14.8 mo (10.5-22.6), resp. [HR = 0.72 (0.54-0.95)]. No new safety signals were obsd. with longer follow-up. Although the anal. did not cross the prespecified boundary for statistical significance, there was a 7.9-mo numeric improvement in median OS when alpelisib was added to fulvestrant treatment of patients with PIK3CA-mutated, HR+, HER2- ABC. Overall, these results further support the statistically significant prolongation of PFS obsd. with alpelisib plus fulvestrant in this population, which has a poor prognosis due to a PIK3CA mutation.
- 39Turner, S.; Chia, S.; Kanakamedala, H.; Hsu, W. C.; Park, J.; Chandiwana, D.; Ridolfi, A.; Yu, C. L.; Zarate, J. P.; Rugo, H. S. Effectiveness of Alpelisib + Fulvestrant Compared with Real-World Standard Treatment Among Patients with HR+, HER2-, PIK3CA-Mutated Breast Cancer. Oncologist 2021, 26, e1133– e1142, DOI: 10.1002/onco.1380439https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1antb7P&md5=14e986b4e02d772d01cc9b6762ffa8feEffectiveness of Alpelisib + Fulvestrant Compared with Real-World Standard Treatment Among Patients with HR +, HER2 -, PIK3CA-Mutated Breast CancerTurner, Stuart; Chia, Stephen; Kanakamedala, Hemanth; Hsu, Wei-Chun; Park, Jinhee; Chandiwana, David; Ridolfi, Antonia; Yu, Chu-Ling; Zarate, Juan Pablo; Rugo, Hope S.Oncologist (2021), 26 (7), e1133-e1142CODEN: OCOLF6; ISSN:1549-490X. (AlphaMed Press)The BYLieve trial (NCT03056755) confirmed efficacy and safety of alpelisib with fulvestrant for hormone receptor-pos. (HR+), human epidermal growth factor receptor-2-neg. (HER2-), PIK3CA-mutated advanced breast cancer (ABC), after cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) with an aromatase inhibitor (AI) as immediate prior therapy. Further analyses were performed to compare efficacy from BYLieve with effectiveness of std. treatment in the real-world setting. Patients who progressed on a CDK4/6i plus AI and were treated with alpelisib with fulvestrant in BYLieve were matched with a real-world patient cohort who received std.-of-care from a deidentified clinico-genomics database (CGDB). Primary and secondary endpoints were to compare progression-free survival (PFS), estd. by the Kaplan-Meier method, and the proportion of patients remaining progression-free at 6 mo, resp., between the two cohorts. A total of 855 patients with PIK3CA-mutant disease who had prior CDK4/6i plus hormone therapy were selected from the CGDB; further matching to 120 patients from BYLieve selected 95 patients without exposure to HER2-targeting agents, clin. study drug, or alpelisib. In unadjusted and postmatching results, primary and secondary endpoints favored treatment with alpelisib with fulvestrant in BYLieve more than std. treatments in the real-world cohort. Postadjustment, median PFS for patients treated with alpelisib in BYLieve was 7.3 vs. 3.7 mo in the real-world cohort, and 6-mo PFS was 54.6% vs. 40.1%, resp. Matched/weighted anal. comparing BYLieve with the real-world setting further supports the clin. benefit of alpelisib with fulvestrant for treatment of HR+, HER2-, PIK3CA-mutant ABC after CDK4/6i treatment. Approx. 40% of patients with hormone receptor-pos. (HR+), human epidermal growth factor receptor-2-neg. (HER2-) advanced breast cancer (ABC) have PIK3CA-mutated tumors, which have been assocd. with endocrine therapy resistance. Alpelisib, an α-selective phosphatidylinositol-3-kinase inhibitor, demonstrated significantly improved progression-free survival in SOLAR-1 and demonstrated clin. efficacy in BYLieve when combined with fulvestrant. Data are limited in comparing the efficacy of alpelisib combined with fulvestrant with effectiveness of std. therapy after CDK4/6i treatment. Using real-world data, this is the first anal. comparing alpelisib combined with fulvestrant with std. treatments for HR+, HER2-, PIK3CA-mutant ABC in the post-CDK4/6i setting.
- 40Verret, B.; Cortes, J.; Bachelot, T.; Andre, F.; Arnedos, M. Efficacy of PI3K inhibitors in advanced breast cancer. Ann. Oncol. 2019, 30, x12– x20, DOI: 10.1093/annonc/mdz381There is no corresponding record for this reference.
- 41Venot, Q.; Blanc, T.; Rabia, S. H.; Berteloot, L.; Ladraa, S.; Duong, J. P.; Blanc, E.; Johnson, S. C.; Hoguin, C.; Boccara, O.; Sarnacki, S.; Boddaert, N.; Pannier, S.; Martinez, F.; Magassa, S.; Yamaguchi, J.; Knebelmann, B.; Merville, P.; Grenier, N.; Joly, D.; Cormier-Daire, V.; Michot, C.; Bole-Feysot, C.; Picard, A.; Soupre, V.; Lyonnet, S.; Sadoine, J.; Slimani, L.; Chaussain, C.; Laroche-Raynaud, C.; Guibaud, L.; Broissand, C.; Amiel, J.; Legendre, C.; Terzi, F.; Canaud, G. Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature 2018, 558, 540– 546, DOI: 10.1038/s41586-018-0217-941https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFeqtbjO&md5=ce1b6f4dd6c5b0f31d4332e5fec21f42Targeted therapy in patients with PIK3CA-related overgrowth syndromeVenot, Quitterie; Blanc, Thomas; Rabia, Smail Hadj; Berteloot, Laureline; Ladraa, Sophia; Duong, Jean-Paul; Blanc, Estelle; Johnson, Simon C.; Hoguin, Clement; Boccara, Olivia; Sarnacki, Sabine; Boddaert, Nathalie; Pannier, Stephanie; Martinez, Frank; Magassa, Sato; Yamaguchi, Junna; Knebelmann, Bertrand; Merville, Pierre; Grenier, Nicolas; Joly, Dominique; Cormier-Daire, Valerie; Michot, Caroline; Bole-Feysot, Christine; Picard, Arnaud; Soupre, Veronique; Lyonnet, Stanislas; Sadoine, Jeremy; Slimani, Lotfi; Chaussain, Catherine; Laroche-Raynaud, Cecile; Guibaud, Laurent; Broissand, Christine; Amiel, Jeanne; Legendre, Christophe; Terzi, Fabiola; Canaud, GuillaumeNature (London, United Kingdom) (2018), 558 (7711), 540-546CODEN: NATUAS; ISSN:0028-0836. (Nature Research)CLOVES syndrome (congenital lipomatous overgrowth, vascular malformations, epidermal naevi, scoliosis/skeletal and spinal syndrome) is a genetic disorder that results from somatic, mosaic gain-of-function mutations of the PIK3CA gene, and belongs to the spectrum of PIK3CA-related overgrowth syndromes (PROS). This rare condition has no specific treatment and a poor survival rate. Here, we describe a postnatal mouse model of PROS/CLOVES that partially recapitulates the human disease, and demonstrate the efficacy of BYL719, an inhibitor of PIK3CA, in preventing and improving organ dysfunction. On the basis of these results, we used BYL719 to treat nineteen patients with PROS. The drug improved the disease symptoms in all patients. Previously intractable vascular tumors became smaller, congestive heart failure was improved, hemihypertrophy was reduced, and scoliosis was attenuated. The treatment was not assocd. with any substantial side effects. In conclusion, this study provides the first direct evidence supporting PIK3CA inhibition as a promising therapeutic strategy in patients with PROS.
- 42Bohnacker, T.; Prota, A. E.; Beaufils, F.; Burke, J. E.; Melone, A.; Inglis, A. J.; Rageot, D.; Sele, A. M.; Cmiljanovic, V.; Cmiljanovic, N.; Bargsten, K.; Aher, A.; Akhmanova, A.; Diaz, J. F.; Fabbro, D.; Zvelebil, M.; Williams, R. L.; Steinmetz, M. O.; Wymann, M. P. Deconvolution of Buparlisib’s mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nat. Commun. 2017, 8, 14683 DOI: 10.1038/ncomms1468342https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czksV2ruw%253D%253D&md5=97d27a7d83bf6ab6094a587565f82bf6Deconvolution of Buparlisib's mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic interventionBohnacker Thomas; Beaufils Florent; Melone Anna; Rageot Denise; Sele Alexander M; Cmiljanovic Vladimir; Cmiljanovic Natasa; Wymann Matthias P; Prota Andrea E; Bargsten Katja; Steinmetz Michel O; Burke John E; Inglis Alison J; Williams Roger L; Aher Amol; Akhmanova Anna; Diaz J Fernando; Fabbro Doriano; Zvelebil MarketaNature communications (2017), 8 (), 14683 ISSN:.BKM120 (Buparlisib) is one of the most advanced phosphoinositide 3-kinase (PI3K) inhibitors for the treatment of cancer, but it interferes as an off-target effect with microtubule polymerization. Here, we developed two chemical derivatives that differ from BKM120 by only one atom. We show that these minute changes separate the dual activity of BKM120 into discrete PI3K and tubulin inhibitors. Analysis of the compounds cellular growth arrest phenotypes and microtubule dynamics suggest that the antiproliferative activity of BKM120 is mainly due to microtubule-dependent cytotoxicity rather than through inhibition of PI3K. Crystal structures of BKM120 and derivatives in complex with tubulin and PI3K provide insights into the selective mode of action of this class of drugs. Our results raise concerns over BKM120's generally accepted mode of action, and provide a unique mechanistic basis for next-generation PI3K inhibitors with improved safety profiles and flexibility for use in combination therapies.
- 43Beaufils, F.; Cmiljanovic, N.; Cmiljanovic, V.; Bohnacker, T.; Melone, A.; Marone, R.; Jackson, E.; Zhang, X.; Sele, A.; Borsari, C.; Mestan, J.; Hebeisen, P.; Hillmann, P.; Giese, B.; Zvelebil, M.; Fabbro, D.; Williams, R. L.; Rageot, D.; Wymann, M. P. 5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a potent, brain-penetrant, orally bioavailable, pan-class I PI3K/mTOR inhibitor as clinical candidate in oncology. J. Med. Chem. 2017, 60, 7524– 7538, DOI: 10.1021/acs.jmedchem.7b0093043https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlKhurrP&md5=54d4db75f6a8bda3f12eaf1d196352fd5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a Potent, Brain-Penetrant, Orally Bioavailable, Pan-Class I PI3K/mTOR Inhibitor as Clinical Candidate in OncologyBeaufils, Florent; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Bohnacker, Thomas; Melone, Anna; Marone, Romina; Jackson, Eileen; Zhang, Xuxiao; Sele, Alexander; Borsari, Chiara; Mestan, Jurgen; Hebeisen, Paul; Hillmann, Petra; Giese, Bernd; Zvelebil, Marketa; Fabbro, Doriano; Williams, Roger L.; Rageot, Denise; Wymann, Matthias P.Journal of Medicinal Chemistry (2017), 60 (17), 7524-7538CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Phosphoinositide 3-kinase (PI3K) is deregulated in a wide variety of human tumors and triggers activation of protein kinase B (PKB/Akt) and mammalian target of rapamycin (mTOR). Here we describe the preclin. characterization of compd. 1 (PQR309, bimiralisib), a potent 4,6-dimorpholino-1,3,5-triazine-based pan-class I PI3K inhibitor, which targets mTOR kinase in a balanced fashion at higher concns. No off-target interactions were detected for 1 in a wide panel of protein kinase, enzyme, and receptor ligand assays. Moreover, 1 did not bind tubulin, which was obsd. for the structurally related 4 (BKM120, buparlisib). Compd. 1 is orally available, crosses the blood-brain barrier, and displayed favorable pharmacokinetic parameters in mice, rats, and dogs. Compd. 1 demonstrated efficiency in inhibiting proliferation in tumor cell lines and a rat xenograft model. This, together with the compd.'s safety profile, identifies 1 as a clin. candidate with a broad application range in oncol., including treatment of brain tumors or CNS metastasis. Compd. 1 is currently in phase II clin. trials for advanced solid tumors and refractory lymphoma.
- 44Tarantelli, C.; Gaudio, E.; Arribas, A. J.; Kwee, I.; Hillmann, P.; Rinaldi, A.; Cascione, L.; Spriano, F.; Bernasconi, E.; Guidetti, F.; Carrassa, L.; Pittau, R. B.; Beaufils, F.; Ritschard, R.; Rageot, D.; Sele, A.; Dossena, B.; Rossi, F. M.; Zucchetto, A.; Taborelli, M.; Gattei, V.; Rossi, D.; Stathis, A.; Stussi, G.; Broggini, M.; Wymann, M. P.; Wicki, A.; Zucca, E.; Cmiljanovic, V.; Fabbro, D.; Bertoni, F. PQR309 is a novel dual PI3K/mTOR inhibitor with preclinical antitumor activity in lymphomas as a single agent and in combination therapy. Clin. Cancer Res. 2018, 24, 120– 129, DOI: 10.1158/1078-0432.CCR-17-104144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVWktw%253D%253D&md5=730305c33b7f669b0eedd0614edd886cPQR309 Is a Novel Dual PI3K/mTOR Inhibitor with Preclinical Antitumor Activity in Lymphomas as a Single Agent and in Combination TherapyTarantelli, Chiara; Gaudio, Eugenio; Arribas, Alberto J.; Kwee, Ivo; Hillmann, Petra; Rinaldi, Andrea; Cascione, Luciano; Spriano, Filippo; Bernasconi, Elena; Guidetti, Francesca; Carrassa, Laura; Pittau, Roberta Bordone; Beaufils, Florent; Ritschard, Reto; Rageot, Denise; Sele, Alexander; Dossena, Barbara; Rossi, Francesca Maria; Zucchetto, Antonella; Taborelli, Monica; Gattei, Valter; Rossi, Davide; Stathis, Anastasios; Stussi, Georg; Broggini, Massimo; Wymann, Matthias P.; Wicki, Andreas; Zucca, Emanuele; Cmiljanovic, Vladimir; Fabbro, Doriano; Bertoni, FrancescoClinical Cancer Research (2018), 24 (1), 120-129CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Activation of the PI3K/mTOR signaling pathway is recurrent in different lymphoma types, and pharmacol. inhibition of the PI3K/mTOR pathway has shown activity in lymphoma patients. Here, we extensively characterized the in vitro and in vivo activity and the mechanism of action of PQR309 (bimiralisib), a novel oral selective dual PI3K/mTOR inhibitor under clin. evaluation, in preclin. lymphoma models. Exptl. Design: This study included preclin. in vitro activity screening on a large panel of cell lines, both as single agent and in combination, validation expts. on in vivo models and primary cells, proteomics and gene-expression profiling, and comparison with other signaling inhibitors. Results: PQR309 had in vitro antilymphoma activity as single agent and in combination with venetoclax, panobinostat, ibrutinib, lenalidomide, ARV-825, marizomib, and rituximab. Sensitivity to PQR309 was assocd. with specific baseline gene-expression features, such as high expression of transcripts coding for the BCR pathway. Combining proteomics and RNA profiling, we identified the different contribution of PQR309-induced protein phosphorylation and gene expression changes to the drug mechanism of action. Gene-expression signatures induced by PQR309 and by other signaling inhibitors largely overlapped. PQR309 showed activity in cells with primary or secondary resistance to idelalisib. Conclusions: On the basis of these results, PQR309 appeared as a novel and promising compd. that is worth developing in the lymphoma setting. Clin Cancer Res; 24(1); 120-9. ©2017 AACR.
- 45Wicki, A.; Brown, N.; Xyrafas, A.; Bize, V.; Hawle, H.; Berardi, S.; Cmiljanovic, N.; Cmiljanovic, V.; Stumm, M.; Dimitrijevic, S.; Herrmann, R.; Pretre, V.; Ritschard, R.; Tzankov, A.; Hess, V.; Childs, A.; Hierro, C.; Rodon, J.; Hess, D.; Joerger, M.; von Moos, R.; Sessa, C.; Kristeleit, R. First-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13). Eur. J. Cancer 2018, 96, 6– 16, DOI: 10.1016/j.ejca.2018.03.01245https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1Snu70%253D&md5=acbdfdb59498f5543ba7f02ed9709d0eFirst-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13)Wicki, Andreas; Brown, Nicholas; Xyrafas, Alexandros; Bize, Vincent; Hawle, Hanne; Berardi, Simona; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Stumm, Michael; Dimitrijevic, Sasa; Herrmann, Richard; Pretre, Vincent; Ritschard, Reto; Tzankov, Alexandar; Hess, Viviane; Childs, Alexa; Hierro, Cinta; Rodon, Jordi; Hess, Dagmar; Joerger, Markus; von Moos, Roger; Sessa, Cristiana; Kristeleit, RebeccaEuropean Journal of Cancer (2018), 96 (), 6-16CODEN: EJCAEL; ISSN:0959-8049. (Elsevier Ltd.)PQR309 is an orally bioavailable, balanced pan-phosphatidylinositol-3-kinase (PI3K), mammalian target of rapamycin (mTOR) C1 and mTORC2 inhibitor. This is an accelerated titrn., 3 D 3 dose-escalation, open-label phase Itrial of continuous once-daily (OD) PQR309 administration to evaluate the safety, pharmacokinetics (PK) and pharmacodynamics in patients with advanced solid tumors. Primary objectives were to det. the max. tolerated dose (MTD) and recommended phase 2 dose (RP2D).Twenty-eight patients were included in six dosing cohorts and treated at a daily PQR309 dose ranging from 10 to 150 mg. Common adverse events (AEs; ≥30% patients) included fatigue, hyperglycemia, nausea, diarrhea, constipation, rash, anorexia and vomiting. Grade (G) 3 or 4 drug-related AEs were seen in 13 (46%) and three (11%) patients, resp. Dose-limiting toxicity (DLT) was obsd. in two patients at 100 mg OD (>14-d interruption in PQR309 due to G3 rash, G2 hyperbilirubinemia, G4 suicide attempt; dose redn. due to G3 fatigue, G2 diarrhoea, G4 transaminitis) and one patient at 80 mg (G3 hyperglycemia >7 d). PK shows fast absorption (Tmax 1-2 h) and dose proportionality for Cmax and area under the curve. A partial response in a patient with metastatic thymus cancer, 24% disease vol. redn. in a patient with sinonasal cancer and stable disease for more than 16 wk in a patient with clear cell Bartholin's gland cancer were obsd.The MTD and RP2D of PQR309 is 80 mg of orally OD. PK is dose-proportional. PD shows PI3K pathway phosphoprotein downregulation in paired tumor biopsies. Clin. activity was obsd. in patients with and without PI3K pathway dysregulation.
- 46Borsari, C.; Rageot, D.; Beaufils, F.; Bohnacker, T.; Keles, E.; Buslov, I.; Melone, A.; Sele, A. M.; Hebeisen, P.; Fabbro, D.; Hillmann, P.; Wymann, M. P. Preclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl-Pyrimidine Moiety. ACS Med. Chem. Lett. 2019, 10, 1473– 1479, DOI: 10.1021/acsmedchemlett.9b0033346https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslSqtrzP&md5=3423dce039aa5b68a178ecea264e6f1fPreclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl-Pyrimidine MoietyBorsari, Chiara; Rageot, Denise; Beaufils, Florent; Bohnacker, Thomas; Keles, Erhan; Buslov, Ivan; Melone, Anna; Sele, Alexander M.; Hebeisen, Paul; Fabbro, Doriano; Hillmann, Petra; Wymann, Matthias P.ACS Medicinal Chemistry Letters (2019), 10 (10), 1473-1479CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is a crit. regulator of cell growth and is frequently hyperactivated in cancer. Therefore, PI3K inhibitors represent a valuable asset in cancer therapy. Herein we have developed a novel anticancer agent, the potent pan-PI3K inhibitor PQR514 (4), which is a follow-up compd. for the phase-II clin. compd. PQR309 (1). Compd. 4 has an improved potency both in vitro and in cellular assays with respect to its predecessor compds. It shows superiority in the suppression of cancer cell proliferation and demonstrates significant antitumor activity in an OVCAR-3 xenograft model at concns. approx. eight times lower than PQR309 (1). The favorable pharmacokinetic profile and a minimal brain penetration promote PQR514 (4) as an optimized candidate for the treatment of systemic tumors.
- 47Jackson, P. A.; Widen, J. C.; Harki, D. A.; Brummond, K. M. Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition Reactions. J. Med. Chem. 2017, 60, 839– 885, DOI: 10.1021/acs.jmedchem.6b0078847https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitV2rtbnM&md5=36d3be5bf688c4e4a8cf4bcf42073009Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition ReactionsJackson, Paul A.; Widen, John C.; Harki, Daniel A.; Brummond, Kay M.Journal of Medicinal Chemistry (2017), 60 (3), 839-885CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Although Michael acceptors display a potent and broad spectrum of bioactivity, they have largely been ignored in drug discovery because of their presumed indiscriminate reactivity. As such, a dearth of information exists relevant to the thiol reactivity of natural products and their analogs possessing this moiety. In the midst of recently approved acrylamide-contg. drugs, it is clear that a good understanding of the hetero-Michael addn. reaction and the relative reactivities of biol. thiols with Michael acceptors under physiol. conditions is needed for the design and use of these compds. as biol. tools and potential therapeutics. This Perspective provides information that will contribute to this understanding, such as kinetics of thiol addn. reactions, bioactivities, as well as steric and electronic factors that influence the electrophilicity and reversibility of Michael acceptors. This Perspective is focused on α,β-unsatd. carbonyls given their preponderance in bioactive natural products.
- 48Flanagan, M. E.; Abramite, J. A.; Anderson, D. P.; Aulabaugh, A.; Dahal, U. P.; Gilbert, A. M.; Li, C.; Montgomery, J.; Oppenheimer, S. R.; Ryder, T.; Schuff, B. P.; Uccello, D. P.; Walker, G. S.; Wu, Y.; Brown, M. F.; Chen, J. M.; Hayward, M. M.; Noe, M. C.; Obach, R. S.; Philippe, L.; Shanmugasundaram, V.; Shapiro, M. J.; Starr, J.; Stroh, J.; Che, Y. Chemical and computational methods for the characterization of covalent reactive groups for the prospective design of irreversible inhibitors. J. Med. Chem. 2014, 57, 10072– 10079, DOI: 10.1021/jm501412a48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVyjtLbM&md5=3ccf59ab7a494655185e5eb5becf8c48Chemical and Computational Methods for the Characterization of Covalent Reactive Groups for the Prospective Design of Irreversible InhibitorsFlanagan, Mark E.; Abramite, Joseph A.; Anderson, Dennis P.; Aulabaugh, Ann; Dahal, Upendra P.; Gilbert, Adam M.; Li, Chao; Montgomery, Justin; Oppenheimer, Stacey R.; Ryder, Tim; Schuff, Brandon P.; Uccello, Daniel P.; Walker, Gregory S.; Wu, Yan; Brown, Matthew F.; Chen, Jinshan M.; Hayward, Matthew M.; Noe, Mark C.; Obach, R. Scott; Philippe, Laurence; Shanmugasundaram, Veerabahu; Shapiro, Michael J.; Starr, Jeremy; Stroh, Justin; Che, YeJournal of Medicinal Chemistry (2014), 57 (23), 10072-10079CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Interest in drugs that covalently modify their target is driven by the desire for enhanced efficacy that can result from the silencing of enzymic activity until protein resynthesis can occur, along with the potential for increased selectivity by targeting uniquely positioned nucleophilic residues in the protein. However, covalent approaches carry addnl. risk for toxicities or hypersensitivity reactions that can result from covalent modification of unintended targets. Here we describe methods for measuring the reactivity of covalent reactive groups (CRGs) with a biol. relevant nucleophile, glutathione (GSH), along with kinetic data for a broad array of electrophiles. We also describe a computational method for predicting electrophilic reactivity, which taken together can be applied to the prospective design of thiol-reactive covalent inhibitors.
- 49Martin, J. S.; MacKenzie, C. J.; Fletcher, D.; Gilbert, I. H. Characterising covalent warhead reactivity. Bioorg. Med. Chem. 2019, 27, 2066– 2074, DOI: 10.1016/j.bmc.2019.04.00249https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntVGgsbo%253D&md5=487334d8d0a8b03d0a25d6d87c1b4329Characterising covalent warhead reactivityMartin, James S.; MacKenzie, Claire J.; Fletcher, Daniel; Gilbert, Ian H.Bioorganic & Medicinal Chemistry (2019), 27 (10), 2066-2074CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Many drugs currently used are covalent inhibitors and irreversibly inhibit their targets. Most of these were discovered through serendipity. Covalent inhibitions can have many advantages from a pharmacokinetic perspective. However, until recently most organizations have shied away from covalent compd. design due to fears of non-specific inhibition of off-target proteins leading to toxicity risks. However, there has been a renewed interest in covalent modifiers as potential drugs, as it possible to get highly selective compds. It is therefore important to know how reactive a warhead is and to be able to select the least reactive warhead possible to avoid toxicity. A robust NMR based assay was developed and used to measure the reactivity of a variety of covalent warheads against serine and cysteine - the two most common targets for covalent drugs. A selection of these warheads also had their reactivity measured against threonine, tyrosine, lysine, histidine and arginine to better understand our ability to target non-traditional residues. The reactivity was also measured at various pHs to assess what effect the environment in the active site would have on these reactions. The reactivity of a covalent modifier was found to be very dependent on the amino acid residue.
- 50Lonsdale, R.; Burgess, J.; Colclough, N.; Davies, N. L.; Lenz, E. M.; Orton, A. L.; Ward, R. A. Expanding the Armory: Predicting and Tuning Covalent Warhead Reactivity. J. Chem. Inf. Model. 2017, 57, 3124– 3137, DOI: 10.1021/acs.jcim.7b0055350https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsl2gsLjF&md5=722e83699c58dee1009b87c8c31bdeccExpanding the Armory: Predicting and Tuning Covalent Warhead ReactivityLonsdale, Richard; Burgess, Jonathan; Colclough, Nicola; Davies, Nichola L.; Lenz, Eva M.; Orton, Alexandra L.; Ward, Richard A.Journal of Chemical Information and Modeling (2017), 57 (12), 3124-3137CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Targeted covalent inhibition is an established approach for increasing the potency and selectivity of potential drug candidates, as well as identifying potent and selective tool compds. for target validation studies. It is evident that identification of reversible recognition elements is essential for selective covalent inhibition, but this must also be achieved with the appropriate level of inherent reactivity of the reactive functionality (or "warhead"). Structural changes that increase or decrease warhead reactivity, guided by methods to predict the effect of those changes, have the potential to tune warhead reactivity and negate issues related to potency and/or toxicity. The half-life to adduct formation with glutathione (GSH t1/2) is a useful assay for measuring the reactivity of cysteine-targeting covalent warheads but is limited to synthesized mols. In this manuscript the authors assess the ability of several exptl. and computational approaches to predict GSH t1/2 for a range of cysteine targeting warheads, including a novel method based on pKa. Furthermore, matched mol. pairs anal. has been performed against the internal compd. collection, revealing structure-activity relationships between a selection of different covalent warheads. These observations and methods of prediction will be valuable in the design of new covalent inhibitors with desired levels of reactivity.
- 51Ma, S.; Devi-Kesavan, L. S.; Gao, J. Molecular dynamics simulations of the catalytic pathway of a cysteine protease: a combined QM/MM study of human cathepsin K. J. Am. Chem. Soc. 2007, 129, 13633– 13645, DOI: 10.1021/ja074222+51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtFKhtbjK&md5=051a894ff0cf93fcd72bb572088284a2Molecular Dynamics Simulations of the Catalytic Pathway of a Cysteine Protease: A Combined QM/MM Study of Human Cathepsin KMa, Shuhua; Devi-Kesavan, Lakshmi S.; Gao, JialiJournal of the American Chemical Society (2007), 129 (44), 13633-13645CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Mol. dynamics simulations using a combined QM/MM potential have been performed to study the catalytic mechanism of human cathepsin K, a member of the papain family of cysteine proteases. We have detd. the two-dimensional free energy surfaces of both acylation and deacylation steps to characterize the reaction mechanism. These free energy profiles show that the acylation step is rate limiting with a barrier height of 19.8 kcal/mol in human cathepsin K and of 29.3 kcal/mol in aq. soln. The free energy of activation for the deacylation step is 16.7 kcal/mol in cathepsin K and 17.8 kcal/mol in aq. soln. The redn. of free energy barrier is achieved by stabilization of the oxyanion in the transition state. Interestingly, although the "oxyanion hole" has been formed in the Michaelis complex, the amide units do not donate hydrogen bonds directly to the carbonyl oxygen of the substrate, but they stabilize the thiolate anion nucleophile. Hydrogen-bonding interactions are induced as the substrate amide group approaches the nucleophile, moving more than 2 Å and placing the oxyanion in contact with Gln19 and the backbone amide of Cys25. The hydrolysis of peptide substrate shares a common mechanism both for the catalyzed reaction in human cathepsin K and for the uncatalyzed reaction in water. Overall, the nucleophilic attack by Cys25 thiolate and the proton-transfer reaction from His162 to the amide nitrogen are highly coupled, whereas a tetrahedral intermediate is formed along the nucleophilic reaction pathway.
- 52Cusack, K. P.; Arnold, L. D.; Barberis, C. E.; Chen, H.; Ericsson, A. M.; Gaza-Bulseco, G. S.; Gordon, T. D.; Grinnell, C. M.; Harsch, A.; Pellegrini, M.; Tarcsa, E. A 13C NMR approach to categorizing potential limitations of alpha,beta-unsaturated carbonyl systems in drug-like molecules. Bioorg. Med. Chem. Lett. 2004, 14, 5503– 5507, DOI: 10.1016/j.bmcl.2004.09.00752https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXos1eju70%253D&md5=ee93901b0f81e811ff86c86210a8286cA 13C NMR approach to categorizing potential limitations of α,β-unsaturated carbonyl systems in drug-like moleculesCusack, Kevin P.; Arnold, Lee D.; Barberis, Claude E.; Chen, Haipeng; Ericsson, Anna M.; Gaza-Bulseco, Georgeen S.; Gordon, Thomas D.; Grinnell, Christine M.; Harsch, Andreas; Pellegrini, Maria; Tarcsa, EditBioorganic & Medicinal Chemistry Letters (2004), 14 (22), 5503-5507CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Compds. that contain an α,β-unsatd. carbonyl moiety are often flagged as potential Michael acceptors. All α,β-unsatd. carbonyl moieties are not equiv., however, and we sought to better understand this system and its potential implications in drug-like mols. Measurement of the 13C NMR shift of the β-carbon and correlation to in vitro results allowed compds. in our collection to be categorized as potential Michael acceptors, potential substrates for NADPH, or as photoisomerizable.
- 53MacFaul, P. A.; Morley, A. D.; Crawford, J. J. A simple in vitro assay for assessing the reactivity of nitrile containing compounds. Bioorg. Med. Chem. Lett. 2009, 19, 1136– 1138, DOI: 10.1016/j.bmcl.2008.12.10553https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXit1WrtLs%253D&md5=d949b5d0e76c6d268f3f57cc4584f0b9A simple in vitro assay for assessing the reactivity of nitrile containing compoundsMacFaul, Philip A.; Morley, Andrew D.; Crawford, James J.Bioorganic & Medicinal Chemistry Letters (2009), 19 (4), 1136-1138CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)A quant. assay involving the reaction of nitriles with glutathione and cysteine has been used as a simple in vitro screen to assess potential toxicity risk of candidate compds. in drug discovery. Studies have indicated that, when benchmarked with selected compds., the reaction of the nitriles with glutathione can provide a useful tool for deciding whether or not to progress compds. in the absence of radiolabelling studies.
- 54Brameld, K. A.; Owens, T. D.; Verner, E.; Venetsanakos, E.; Bradshaw, J. M.; Phan, V. T.; Tam, D.; Leung, K.; Shu, J.; LaStant, J.; Loughhead, D. G.; Ton, T.; Karr, D. E.; Gerritsen, M. E.; Goldstein, D. M.; Funk, J. O. Discovery of the Irreversible Covalent FGFR Inhibitor 8-(3-(4-Acryloylpiperazin-1-yl)propyl)-6-(2,6-dichloro-3,5-dimethoxyphenyl)-2-(methylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (PRN1371) for the Treatment of Solid Tumors. J. Med. Chem. 2017, 60, 6516– 6527, DOI: 10.1021/acs.jmedchem.7b0036054https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVykt7fK&md5=987069b645d5e0e36ccda5e5c81520b9Discovery of the Irreversible Covalent FGFR Inhibitor 8-(3-(4-Acryloylpiperazin-1-yl)propyl)-6-(2,6-dichloro-3,5-dimethoxyphenyl)-2-(methylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (PRN1371) for the Treatment of Solid TumorsBrameld, Ken A.; Owens, Timothy D.; Verner, Erik; Venetsanakos, Eleni; Bradshaw, J. Michael; Phan, Vernon T.; Tam, Danny; Leung, Kwan; Shu, Jin; LaStant, Jacob; Loughhead, David G.; Ton, Tony; Karr, Dane E.; Gerritsen, Mary E.; Goldstein, David M.; Funk, Jens OliverJournal of Medicinal Chemistry (2017), 60 (15), 6516-6527CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Aberrant signaling of the FGF/FGFR pathway occurs frequently in cancers and is an oncogenic driver in many solid tumors. Clin. validation of FGFR as a therapeutic target has been demonstrated in bladder, liver, lung, breast, and gastric cancers. Our goal was to develop an irreversible covalent inhibitor of FGFR1-4 for use in oncol. indications. An irreversible covalent binding mechanism imparts many desirable pharmacol. benefits including high potency, selectivity, and prolonged target inhibition. Herein we report the structure-based design, medicinal chem. optimization, and unique ADME assays of our irreversible covalent drug discovery program which culminated in the discovery of compd. 34 (PRN1371), a highly selective and potent FGFR1-4 inhibitor.
- 55Cee, V. J.; Volak, L. P.; Chen, Y.; Bartberger, M. D.; Tegley, C.; Arvedson, T.; McCarter, J.; Tasker, A. S.; Fotsch, C. Systematic Study of the Glutathione (GSH) Reactivity of N-Arylacrylamides: 1. Effects of Aryl Substitution. J. Med. Chem. 2015, 58, 9171– 9178, DOI: 10.1021/acs.jmedchem.5b0101855https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFGrsLfN&md5=2f3a816446dd73d5be061ad8d2195e2dSystematic Study of the Glutathione (GSH) Reactivity of N-Arylacrylamides: 1. Effects of Aryl SubstitutionCee, Victor J.; Volak, Laurie P.; Chen, Yuping; Bartberger, Michael D.; Tegley, Chris; Arvedson, Tara; McCarter, John; Tasker, Andrew S.; Fotsch, ChristopherJournal of Medicinal Chemistry (2015), 58 (23), 9171-9178CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Success in the design of targeted covalent inhibitors depends in part on a knowledge of the factors influencing electrophile reactivity. In an effort to further develop an understanding of structure-reactivity relations among N-arylacrylamides, the authors detd. glutathione (GSH) reaction rates for a family of N-arylacrylamides independently substituted at ortho-, meta-, and para-positions with 11 different groups common to inhibitor design. Substituent effects on reaction rates show a linear Hammett correlation for ortho-, meta-, and para-substitution. The authors note a correlation between 1H and 13C NMR chem. shifts of the acrylamide with GSH reaction rates, suggesting that NMR chem. shifts may be a convenient surrogate measure of relative acrylamide reactivity. D. functional theory calcns. reveal a correlation between computed activation parameters and exptl. detd. reaction rates, validating the use of such methodol. for the screening of synthetic candidates in a prospective fashion.
- 56Birkholz, A.; Kopecky, D. J.; Volak, L. P.; Bartberger, M. D.; Chen, Y.; Tegley, C. M.; Arvedson, T.; McCarter, J. D.; Fotsch, C.; Cee, V. J. Systematic Study of the Glutathione Reactivity of N-Phenylacrylamides: 2. Effects of Acrylamide Substitution. J. Med. Chem. 2020, 63, 11602– 11614, DOI: 10.1021/acs.jmedchem.0c0074956https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvV2ns7zJ&md5=051ff03cf309131266a52f6f3c55087aSystematic Study of the Glutathione Reactivity of N-Phenylacrylamides: 2. Effects of Acrylamide SubstitutionBirkholz, Adam; Kopecky, David J.; Volak, Laurie P.; Bartberger, Michael D.; Chen, Yuping; Tegley, Christopher M.; Arvedson, Tara; McCarter, John D.; Fotsch, Christopher; Cee, Victor J.Journal of Medicinal Chemistry (2020), 63 (20), 11602-11614CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A comprehensive understanding of structure-reactivity relationships is crit. to the design and optimization of cysteine-targeted covalent inhibitors. Herein, we report glutathione (GSH) reaction rates for N-Ph acrylamides with varied substitutions at the α- and β-positions of the acrylamide moiety. We find that the GSH reaction rates can generally be understood in terms of the electron donating or withdrawing ability of the substituent. When installed at the β-position, aminomethyl substituents with amine pKa's > 7 accelerate, while those with pKa's < 7 slow the rate of GSH addn. at pH 7.4, relative to a hydrogen substituent. Although a computational model was able to only approx. capture exptl. reactivity trends, our calcns. do not support a frequently invoked mechanism of concerted amine/thiol proton transfer and C-S bond formation and instead suggest that protonated aminomethyl functions as an electron-withdrawing group to reduce the barrier for thiolate addn. to the acrylamide.
- 57Zhu, K.; Borrelli, K. W.; Greenwood, J. R.; Day, T.; Abel, R.; Farid, R. S.; Harder, E. Docking covalent inhibitors: a parameter free approach to pose prediction and scoring. J. Chem. Inf. Model. 2014, 54, 1932– 1940, DOI: 10.1021/ci500118s57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpsVKksLs%253D&md5=3633c29fa5dc29817b178fd10a4c7b15Docking Covalent Inhibitors: A Parameter Free Approach To Pose Prediction and ScoringZhu, Kai; Borrelli, Kenneth W.; Greenwood, Jeremy R.; Day, Tyler; Abel, Robert; Farid, Ramy S.; Harder, EdwardJournal of Chemical Information and Modeling (2014), 54 (7), 1932-1940CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Although many popular docking programs include a facility to account for covalent ligands, large-scale systematic docking validation studies of covalent inhibitors have been sparse. In this paper, we present the development and validation of a novel approach for docking and scoring covalent inhibitors, which consists of conventional noncovalent docking, heuristic formation of the covalent attachment point, and structural refinement of the protein-ligand complex. This approach combines the strengths of the docking program Glide and the protein structure modeling program Prime and does not require any parameter fitting for the study of addnl. covalent reaction types. We first test this method by predicting the native binding geometry of 38 covalently bound complexes. The av. RMSD of the predicted poses is 1.52 Å, and 76% of test set inhibitors have an RMSD of less than 2.0 Å. In addn., the apparent affinity score constructed herein is tested on a virtual screening study and the characterization of the SAR properties of two different series of congeneric compds. with satisfactory success.
- 58Toledo Warshaviak, D.; Golan, G.; Borrelli, K. W.; Zhu, K.; Kalid, O. Structure-based virtual screening approach for discovery of covalently bound ligands. J. Chem. Inf. Model. 2014, 54, 1941– 1950, DOI: 10.1021/ci500175r58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpslegtbw%253D&md5=0ff8b6e682473abd2e2dadb49d95539aStructure-Based Virtual Screening Approach for Discovery of Covalently Bound LigandsToledo Warshaviak, Dora; Golan, Gali; Borrelli, Kenneth W.; Zhu, Kai; Kalid, OriJournal of Chemical Information and Modeling (2014), 54 (7), 1941-1950CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)We present a fast and effective covalent docking approach suitable for large-scale virtual screening (VS). We applied this method to four targets (HCV NS3 protease, Cathepsin K, EGFR, and XPO1) with known crystal structures and known covalent inhibitors. We implemented a customized "VS mode" of the Schrodinger Covalent Docking algorithm (CovDock), which we refer to as CovDock-VS. Known actives and target-specific sets of decoys were docked to selected X-ray structures, and poses were filtered based on noncovalent protein-ligand interactions known to be important for activity. We were able to retrieve 71%, 72%, and 77% of the known actives for Cathepsin K, HCV NS3 protease, and EGFR within 5% of the decoy library, resp. With the more challenging XPO1 target, where no specific interactions with the protein could be used for postprocessing of the docking results, we were able to retrieve 95% of the actives within 30% of the decoy library and achieved an early enrichment factor (EF1%) of 33. The poses of the known actives bound to existing crystal structures of 4 targets were predicted with an av. RMSD of 1.9 Å. To the best of our knowledge, CovDock-VS is the first fully automated tool for efficient virtual screening of covalent inhibitors. Importantly, CovDock-VS can handle multiple chem. reactions within the same library, only requiring a generic SMARTS-based predefinition of the reaction. CovDock-VS provides a fast and accurate way of differentiating actives from decoys without significantly deteriorating the accuracy of the predicted poses for covalent protein-ligand complexes. Therefore, we propose CovDock-VS as an efficient structure-based virtual screening method for discovery of novel and diverse covalent ligands.
- 59Rageot, D.; Bohnacker, T.; Keles, E.; McPhail, J. A.; Hoffmann, R. M.; Melone, A.; Borsari, C.; Sriramaratnam, R.; Sele, A. M.; Beaufils, F.; Hebeisen, P.; Fabbro, D.; Hillmann, P.; Burke, J. E.; Wymann, M. P. (S)-4-(Difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530), a potent, orally bioavailable, and brain-penetrable dual inhibitor of class I PI3K and mTOR kinase. J. Med. Chem. 2019, 62, 6241– 6261, DOI: 10.1021/acs.jmedchem.9b0052559https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFWgtLrM&md5=48e0d98bf33ad5c98a1911b82bcccf03(S)-4-(Difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530), a Potent, Orally Bioavailable, and Brain-Penetrable Dual Inhibitor of Class I PI3K and mTOR KinaseRageot, Denise; Bohnacker, Thomas; Keles, Erhan; McPhail, Jacob A.; Hoffmann, Reece M.; Melone, Anna; Borsari, Chiara; Sriramaratnam, Rohitha; Sele, Alexander M.; Beaufils, Florent; Hebeisen, Paul; Fabbro, Doriano; Hillmann, Petra; Burke, John E.; Wymann, Matthias P.Journal of Medicinal Chemistry (2019), 62 (13), 6241-6261CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is frequently overactivated in cancer, and drives cell growth, proliferation, survival, and metastasis. Here, we report a structure-activity relationship study, which led to the discovery of a drug-like ATP-site PI3K/mTOR kinase inhibitor: (S)-4-(difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530, compd. 6), which qualifies as a clin. candidate due to its potency and specificity for PI3K and mTOR kinases, and its pharmacokinetic properties, including brain penetration. Compd. 6 showed excellent selectivity over a wide panel of kinases and an excellent selectivity against unrelated receptor enzymes and ion channels. Moreover, compd. 6 prevented cell growth in a cancer cell line panel. The preclin. in vivo characterization of compd. 6 in an OVCAR-3 xenograft model demonstrated good oral bioavailability, excellent brain penetration, and efficacy. Initial toxicity studies in rats and dogs qualify 6 for further development as a therapeutic agent in oncol.
- 60Goel, P.; Alam, O.; Naim, M. J.; Nawaz, F.; Iqbal, M.; Alam, M. I. Recent advancement of piperidine moiety in treatment of cancer- A review. Eur. J. Med. Chem. 2018, 157, 480– 502, DOI: 10.1016/j.ejmech.2018.08.01760https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFahsrrL&md5=098009e3143fa4e14f98b02bbdc27a15Recent advancement of piperidine moiety in treatment of cancer- A reviewGoel, Pallavi; Alam, Ozair; Naim, Mohd Javed; Nawaz, Farah; Iqbal, Muzaffar; Alam, Md IqbalEuropean Journal of Medicinal Chemistry (2018), 157 (), 480-502CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A review. Piperidine is an important pharmacophore, a privileged scaffold and an excellent heterocyclic system in the field of drug discovery which provides numerous opportunities in studying/exploring this moiety as an anticancer agent by acting on various receptors of utmost importance. Cancer is an uncontrolled division of cells that results in the formation of tumor which have the potential to migrate to other parts of the body (metastasis) finally becoming a major threat to human population. Since piperidine displayed great potential in this area it is being further probed to get novel entities for the treatment of cancer. This review throws light on recent biol. expansions of piperidine along with structure activity relationships to deliver assocn. between various synthesized newer/novel derivs. and receptor sites.
- 61Copeland, R. A. Evaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologists. Methods Biochem. Anal. 2005, 46, 1– 26561https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2MnmtlyjsA%253D%253D&md5=4bb6759b5155aa6c2b6a462432afda7fEvaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologistsCopeland Robert AMethods of biochemical analysis (2005), 46 (), 1-265 ISSN:0076-6941.There is no expanded citation for this reference.
- 62Strelow, J. M. A Perspective on the Kinetics of Covalent and Irreversible Inhibition. SLAS Discovery 2017, 22, 3– 20, DOI: 10.1177/108705711667150962https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlSntrbJ&md5=bd404e6b26df20b8761cd38f0f3c3541A perspective on the kinetics of covalent and irreversible inhibitionStrelow, John M.SLAS Discovery (2017), 22 (1), 3-20CODEN: SDLIBT; ISSN:2472-5560. (Sage Publications)The clin. and com. success of covalent drugs has prompted a renewed and more deliberate pursuit of covalent and irreversible mechanisms within drug discovery. A covalent mechanism can produce potent inhibition in a biochem., cellular, or in vivo setting. In many cases, teams choose to focus on the consequences of the covalent event, defined by an IC50 value. In a biochem. assay, the IC50 may simply reflect the target protein concn. in the assay. What has received less attention is the importance of the rate of covalent modification, defined by kinact/KI. The kinact/KI is a rate const. describing the efficiency of covalent bond formation resulting from the potency (KI) of the first reversible binding event and the max. potential rate (kinact) of inactivation. In this perspective, it is proposed that the kinact/KI should be employed as a crit. parameter to identify covalent inhibitors, interpret structure-activity relationships (SARs), translate activity from biochem. assays to the cell, and more accurately define selectivity. It is also proposed that a physiol. relevant kinact/KI and an (unbound) AUC generated from a pharmacokinetic profile reflecting direct exposure of the inhibitor to the target protein are two crit. determinants of in vivo covalent occupancy. A simple equation is presented to define this relationship and improve the interpretation of covalent and irreversible kinetics.
- 63Dale, N. C.; Johnstone, E. K. M.; White, C. W.; Pfleger, K. D. G. NanoBRET: The Bright Future of Proximity-Based Assays. Front. Bioeng. Biotechnol. 2019, 7, 56 DOI: 10.3389/fbioe.2019.0005663https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252FktFOgsA%253D%253D&md5=31e05be574d590da21049e92b8e1509aNanoBRET: The Bright Future of Proximity-Based AssaysDale Natasha C; Johnstone Elizabeth K M; White Carl W; Pfleger Kevin D G; Dale Natasha C; Johnstone Elizabeth K M; White Carl W; Pfleger Kevin D G; Dale Natasha C; Johnstone Elizabeth K M; White Carl W; Pfleger Kevin D G; Pfleger Kevin D GFrontiers in bioengineering and biotechnology (2019), 7 (), 56 ISSN:2296-4185.Bioluminescence resonance energy transfer (BRET) is a biophysical technique used to monitor proximity within live cells. BRET exploits the naturally occurring phenomenon of dipole-dipole energy transfer from a donor enzyme (luciferase) to an acceptor fluorophore following enzyme-mediated oxidation of a substrate. This results in production of a quantifiable signal that denotes proximity between proteins and/or molecules tagged with complementary luciferase and fluorophore partners. BRET assays have been used to observe an array of biological functions including ligand binding, intracellular signaling, receptor-receptor proximity, and receptor trafficking, however, BRET assays can theoretically be used to monitor the proximity of any protein or molecule for which appropriate fusion constructs and/or fluorophore conjugates can be produced. Over the years, new luciferases and approaches have been developed that have increased the potential applications for BRET assays. In particular, the development of the small, bright and stable Nanoluciferase (NanoLuc; Nluc) and its use in NanoBRET has vastly broadened the potential applications of BRET assays. These advances have exciting potential to produce new experimental methods to monitor protein-protein interactions (PPIs), protein-ligand interactions, and/or molecular proximity. In addition to NanoBRET, Nluc has also been exploited to produce NanoBiT technology, which further broadens the scope of BRET to monitor biological function when NanoBiT is combined with an acceptor. BRET has proved to be a powerful tool for monitoring proximity and interaction, and these recent advances further strengthen its utility for a range of applications.
- 64Vasta, J. D.; Corona, C. R.; Wilkinson, J.; Zimprich, C. A.; Hartnett, J. R.; Ingold, M. R.; Zimmerman, K.; Machleidt, T.; Kirkland, T. A.; Huwiler, K. G.; Ohana, R. F.; Slater, M.; Otto, P.; Cong, M.; Wells, C. I.; Berger, B. T.; Hanke, T.; Glas, C.; Ding, K.; Drewry, D. H.; Huber, K. V. M.; Willson, T. M.; Knapp, S.; Muller, S.; Meisenheimer, P. L.; Fan, F.; Wood, K. V.; Robers, M. B. Quantitative, Wide-Spectrum Kinase Profiling in Live Cells for Assessing the Effect of Cellular ATP on Target Engagement. Cell Chem. Biol. 2018, 25, 206.e11– 214.e11, DOI: 10.1016/j.chembiol.2017.10.010There is no corresponding record for this reference.
- 65Ikediobi, O. N.; Davies, H.; Bignell, G.; Edkins, S.; Stevens, C.; O’Meara, S.; Santarius, T.; Avis, T.; Barthorpe, S.; Brackenbury, L.; Buck, G.; Butler, A.; Clements, J.; Cole, J.; Dicks, E.; Forbes, S.; Gray, K.; Halliday, K.; Harrison, R.; Hills, K.; Hinton, J.; Hunter, C.; Jenkinson, A.; Jones, D.; Kosmidou, V.; Lugg, R.; Menzies, A.; Mironenko, T.; Parker, A.; Perry, J.; Raine, K.; Richardson, D.; Shepherd, R.; Small, A.; Smith, R.; Solomon, H.; Stephens, P.; Teague, J.; Tofts, C.; Varian, J.; Webb, T.; West, S.; Widaa, S.; Yates, A.; Reinhold, W.; Weinstein, J. N.; Stratton, M. R.; Futreal, P. A.; Wooster, R. Mutation analysis of 24 known cancer genes in the NCI-60 cell line set. Mol. Cancer Ther. 2006, 5, 2606– 2612, DOI: 10.1158/1535-7163.Mct-06-043365https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1els7jK&md5=f943d16d3e147357815de0ec3c29c30dMutation analysis of 24 known cancer genes in the NCI-60 cell line setIkediobi, Ogechi N.; Davies, Helen; Bignell, Graham; Edkins, Sarah; Stevens, Claire; O'Meara, Sarah; Santarius, Thomas; Avis, Tim; Barthorpe, Syd; Brackenbury, Lisa; Buck, Gemma; Butler, Adam; Clements, Jody; Cole, Jennifer; Dicks, Ed; Forbes, Simon; Gray, Kristian; Halliday, Kelly; Harrison, Rachel; Hills, Katy; Hinton, Jonathan; Hunter, Chris; Jenkinson, Andy; Jones, David; Kosmidou, Vivienne; Lugg, Richard; Menzies, Andrew; Mironenko, Tatiana; Parker, Adrian; Perry, Janet; Raine, Keiran; Richardson, David; Shepherd, Rebecca; Small, Alex; Smith, Raffaella; Solomon, Helen; Stephens, Philip; Teague, Jon; Tofts, Calli; Varian, Jennifer; Webb, Tony; West, Sofie; Widaa, Sara; Yates, Andy; Reinhold, William; Weinstein, John N.; Stratton, Michael R.; Futreal, P. Andrew; Wooster, RichardMolecular Cancer Therapeutics (2006), 5 (11), 2606-2612CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The panel of 60 human cancer cell lines (the NCI-60) assembled by the National Cancer Institute for anticancer drug discovery is a widely used resource. The NCI-60 has been characterized pharmacol. and at the mol. level more extensively than any other set of cell lines. However, no systematic mutation anal. of genes causally implicated in oncogenesis has been reported. This study reports the sequence anal. of 24 known cancer genes in the NCI-60 and an assessment of 4 of the 24 genes for homozygous deletions. One hundred thirty-seven oncogenic mutations were identified in 14 (APC, BRAF, CDKN2, CTNNB1, HRAS, KRAS, NRAS, SMAD4, PIK3CA, PTEN, RB1, STK11, TP53, and VHL) of the 24 genes. All lines have at least one mutation among the cancer genes examd., with most lines (73%) having more than one. Identification of those cancer genes mutated in the NCI-60, in combination with pharmacol. and mol. profiles of the cells, will allow for more informed interpretation of anticancer agent screening and will enhance the use of the NCI-60 cell lines for molecularly targeted screens.
- 66Gall, V. A.; Philips, A. V.; Qiao, N.; Clise-Dwyer, K.; Perakis, A. A.; Zhang, M.; Clifton, G. T.; Sukhumalchandra, P.; Ma, Q.; Reddy, S. M.; Yu, D.; Molldrem, J. J.; Peoples, G. E.; Alatrash, G.; Mittendorf, E. A. Trastuzumab Increases HER2 Uptake and Cross-Presentation by Dendritic Cells. Cancer Res. 2017, 77, 5374– 5383, DOI: 10.1158/0008-5472.Can-16-277466https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFOls7bO&md5=70f3d634f03212ebd5e026a5acdc1079trastuzumab increases her2 uptake and cross-presentation by dendritic cellsGall, Victor A.; Philips, Anne V.; Qiao, Na; Clise-Dwyer, Karen; Perakis, Alexander A.; Zhang, Mao; Clifton, Guy T.; Sukhumalchandra, Pariya; Ma, Qing; Reddy, Sangeetha M.; Yu, Dihua; Molldrem, Jeffrey J.; Peoples, George E.; Alatrash, Gheath; Mittendorf, Elizabeth A.Cancer Research (2017), 77 (19), 5374-5383CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Early-phase clin. trials evaluating CD8+ T cell-eliciting, HER2-derived peptide vaccines administered to HER2+ breast cancer patients in the adjuvant setting suggest synergy between the vaccines and trastuzumab, the mAb targeting the HER2 protein. Among 60 patients enrolled in clin. trials evaluating the E75 + GM-CSF and GP2 + GM-CSF vaccines, there have been no recurrences in patients vaccinated after receiving trastuzumab as part of std. therapy in the per treatment analyses conducted after a median follow-up of greater than 34 mo. Here, we describe a mechanism by which this synergy may occur. Flow cytometry showed that trastuzumab facilitated uptake of HER2 by dendritic cells (DC), which was mediated by the Fc receptor and was specific to trastuzumab. In vitro, increased HER2 uptake by DC increased cross-presentation of E75, the immunodominant epitope derived from the HER2 protein, an observation confirmed in two in vivo mouse models. This increased E75 cross-presentation, mediated by trastuzumab treatment, enabled more efficient expansion of E75-specific cytotoxic T cells (E75-CTL). These results demonstrate a mechanism by which trastuzumab links innate and adaptive immunity by facilitating activation of antigen-specific T cells. On the basis of these data, we conclude that HER2-pos. breast cancer patients that have been treated with trastuzumab may experience a more robust antitumor immune response by restimulation of T cells with the E75 peptide vaccine, thereby accounting for the improved disease-free survival obsd. with combination therapy.
- 67Chaussade, C.; Rewcastle, G. W.; Kendall, J. D.; Denny, W. A.; Cho, K.; Grønning, L. M.; Chong, M. L.; Anagnostou, S. H.; Jackson, S. P.; Daniele, N.; Shepherd, P. R. Evidence for functional redundancy of class IA PI3K isoforms in insulin signalling. Biochem. J. 2007, 404, 449– 458, DOI: 10.1042/BJ2007000367https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXlvVSgsr8%253D&md5=b74b9308f1e9e36010ccb4163e3a957bEvidence for functional redundancy of class IA PI3K isoforms in insulin signallingChaussade, Claire; Rewcastle, Gordon W.; Kendall, Jackie D.; Denny, William A.; Cho, Kitty; Gronning, Line M.; Chong, Mei Ling; Anagnostou, Sasha H.; Jackson, Shaun P.; Daniele, Nathalie; Shepherd, Peter R.Biochemical Journal (2007), 404 (3), 449-458CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)Recent genetic knock-in and pharmacol. approaches have suggested that, of class IA PI3Ks (phosphatidylinositol 3-kinases), it is the p110α isoform (PIK3CA) that plays the predominant role in insulin signaling. We have used isoform-selective inhibitors of class IA PI3K to dissect further the roles of individual p110 isoforms in insulin signaling. These include a p110α-specific inhibitor (PIK-75), a p110α-selective inhibitor (PI-103), a p110β-specific inhibitor (TGX-221) and a p110δ-specific inhibitor (IC87114). Although we find that p110α is necessary for insulin-stimulated phosphorylation of PKB (protein kinase B) in several cell lines, we find that this is not the case in HepG2 hepatoma cells. Inhibition of p110β or p110δ alone was also not sufficient to block insulin signaling to PKB in these cells, but, when added in combination with p110α inhibitors, they are able to significantly attenuate insulin signaling. Surprisingly, in J774.2 macrophage cells, insulin signaling to PKB was inhibited to a similar extent by inhibitors of p110α, p110β or p110δ. These results provide evidence that p110β and p110δ can play a role in insulin signaling and also provide the first evidence that there can be functional redundancy between p110 isoforms. Further, our results indicate that the degree of functional redundancy is linked to the relative levels of expression of each isoform in the target cells.
- 68Freeman-Cook, K. D.