Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kathleen Aertgeerts is active.

Publication


Featured researches published by Kathleen Aertgeerts.


Protein Science | 2004

Crystal structure of human dipeptidyl peptidase IV in complex with a decapeptide reveals details on substrate specificity and tetrahedral intermediate formation

Kathleen Aertgeerts; Sheng Ye; Mike Tennant; Michelle L. Kraus; Joe Rogers; Bi-Ching Sang; Robert J. Skene; David R. Webb; G. Sridhar Prasad

Dipeptidyl peptidase IV (DPPIV) is a member of the prolyl oligopeptidase family of serine proteases. DPPIV removes dipeptides from the N terminus of substrates, including many chemokines, neuropeptides, and peptide hormones. Specific inhibition of DPPIV is being investigated in human trials for the treatment of type II diabetes. To understand better the molecular determinants that underlie enzyme catalysis and substrate specificity, we report the crystal structures of DPPIV in the free form and in complex with the first 10 residues of the physiological substrate, Neuropeptide Y (residues 1–10; tNPY). The crystal structure of the free form of the enzyme reveals two potential channels through which substrates could access the active site—a so‐called propeller opening, and side opening. The crystal structure of the DPPIV/tNPY complex suggests that bioactive peptides utilize the side opening unique to DPPIV to access the active site. Other structural features in the active site such as the presence of a Glu motif, a well‐defined hydrophobic S1 subsite, and minimal long‐range interactions explain the substrate recognition and binding properties of DPPIV. Moreover, in the DPPIV/tNPY complex structure, the peptide is not cleaved but trapped in a tetrahedral intermediate that occurs during catalysis. Conformational changes of S630 and H740 between DPPIV in its free form and in complex with tNPY were observed and contribute to the stabilization of the tetrahedral intermediate. Our results facilitate the design of potent, selective small molecule inhibitors of DPPIV that may yield compounds for the development of novel drugs to treat type II diabetes.


Journal of Medicinal Chemistry | 2011

Design and Synthesis of Pyrimidinone and Pyrimidinedione Inhibitors of Dipeptidyl Peptidase IV.

Zhiyuan Zhang; Michael B. Wallace; Jun Feng; Jeffrey A. Stafford; Robert J. Skene; Lihong Shi; Bumsup Lee; Kathleen Aertgeerts; Andy Jennings; Rongda Xu; Daniel B. Kassel; Stephen W. Kaldor; Marc Navre; David R. Webb; Stephen L. Gwaltney

The discovery of two classes of heterocyclic dipeptidyl peptidase IV (DPP-4) inhibitors, pyrimidinones and pyrimidinediones, is described. After a single oral dose, these potent, selective, and noncovalent inhibitors provide sustained reduction of plasma DPP-4 activity and lowering of blood glucose in animal models of diabetes. Compounds 13a, 27b, and 27j were selected for development.


Protein Science | 2004

N-linked glycosylation of dipeptidyl peptidase IV (CD26): Effects on enzyme activity, homodimer formation, and adenosine deaminase binding

Kathleen Aertgeerts; Sheng Ye; Lihong Shi; Sridhar Prasad; Darbi Witmer; Ellen Chi; Bi-Ching Sang; Robert A. Wijnands; David R. Webb; Ronald V. Swanson

The type II transmembrane serine protease dipeptidyl peptidase IV (DPPIV), also known as CD26 or adenosine deaminase binding protein, is a major regulator of various physiological processes, including immune, inflammatory, nervous, and endocrine functions. It has been generally accepted that glycosylation of DPPIV and of other transmembrane dipeptidyl peptidases is a prerequisite for enzyme activity and correct protein folding. Crystallographic studies on DPPIV reveal clear N‐linked glycosylation of nine Asn residues in DPPIV. However, the importance of each glycosylation site on physiologically relevant reactions such as dipeptide cleavage, dimer formation, and adenosine deaminase (ADA) binding remains obscure. Individual Asn→Ala point mutants were introduced at the nine glycosylation sites in the extracellular domain of DPPIV (residues 39–766). Crystallographic and biochemical data demonstrate that N‐linked glycosylation of DPPIV does not contribute significantly to its peptidase activity. The kinetic parameters of dipeptidyl peptidase cleavage of wild‐type DPPIV and the N‐glycosylation site mutants were determined by using Ala‐Pro‐AFC and Gly‐Pro‐pNA as substrates and varied by <50%. DPPIV is active as a homodimer. Size‐exclusion chromatographic analysis showed that the glycosylation site mutants do not affect dimerization. ADA binds to the highly glycosylated β‐propeller domain of DPPIV, but the impact of glycosylation on binding had not previously been determined. Our studies indicate that glycosylation of DPPIV is not required for ADA binding. Taken together, these data indicate that in contrast to the generally accepted view, glycosylation of DPPIV is not a prerequisite for catalysis, dimerization, or ADA binding.


Journal of Medicinal Chemistry | 2010

Discovery of a Tetrahydropyrimidin-2(1H)-one Derivative (TAK-442) as a Potent, Selective, and Orally Active Factor Xa Inhibitor

Takuya Fujimoto; Yasuhiro Imaeda; Noriko Konishi; Katsuhiko Hiroe; Masaki Kawamura; Garret P. Textor; Kathleen Aertgeerts; Keiji Kubo

Coagulation enzyme factor Xa (FXa) is a particularly promising target for the development of new anticoagulant agents. We previously reported the imidazo[1,5-c]imidazol-3-one derivative 1 as a potent and orally active FXa inhibitor. However, it was found that 1 predominantly undergoes hydrolysis upon incubation with human liver microsomes, and the human specific metabolic pathway made it difficult to predict the human pharmacokinetics. To address this issue, our synthetic efforts were focused on modification of the imidazo[1,5-c]imidazol-3-one moiety of the active metabolite 3a, derived from 1, which resulted in the discovery of the tetrahydropyrimidin-2(1H)-one derivative 5k as a highly potent and selective FXa inhibitor. Compound 5k showed no detectable amide bond cleavage in human liver microsomes, exhibited a good pharmacokinetic profile in monkeys, and had a potent antithrombotic efficacy in a rabbit model without prolongation of bleeding time. Compound 5k is currently under clinical development with the code name TAK-442.


Bioorganic & Medicinal Chemistry | 2011

Identification of 3-aminomethyl-1,2-dihydro-4-phenyl-1-isoquinolones: a new class of potent, selective, and orally active non-peptide dipeptidyl peptidase IV inhibitors that form a unique interaction with Lys554.

Yoshihiro Banno; Yasufumi Miyamoto; Mitsuru Sasaki; Satoru Oi; Tomoko Asakawa; Osamu Kataoka; Koji Takeuchi; Nobuhiro Suzuki; Koji Ikedo; Takuo Kosaka; Shigetoshi Tsubotani; Akiyoshi Tani; Miyuki Funami; Michiko Tawada; Yoshio Yamamoto; Kathleen Aertgeerts; Jason Yano; Hironobu Maezaki

The design, synthesis, and structure-activity relationships of a new class of potent and orally active non-peptide dipeptidyl peptidase IV (DPP-4) inhibitors, 3-aminomethyl-1,2-dihydro-4-phenyl-1-isoquinolones, are described. We hypothesized that the 4-phenyl group of the isoquinolone occupies the S1 pocket of the enzyme, the 3-aminomethyl group forms an electrostatic interaction with the S2 pocket, and the introduction of a hydrogen bond donor onto the 6- or 7-substituent provides interaction with the hydrophilic region of the enzyme. Based on this hypothesis, intensive research focused on developing new non-peptide DPP-4 inhibitors has been carried out. Among the compounds designed in this study, we identified 2-[(3-aminomethyl-2-(2-methylpropyl)-1-oxo-4-phenyl-1,2-dihydro-6-isoquinolinyl)oxy]acetamide (35a) as a potent, selective, and orally bioavailable DPP-4 inhibitor, which exhibited in vivo efficacy in diabetic model rats. Finally, X-ray crystallography of 35a in a complex with the enzyme validated our hypothesized binding mode and identified Lys554 as a new target-binding site available for DPP-4 inhibitors.


Bioorganic & Medicinal Chemistry | 2011

Design and synthesis of 3-pyridylacetamide derivatives as dipeptidyl peptidase IV (DPP-4) inhibitors targeting a bidentate interaction with Arg125

Yasufumi Miyamoto; Yoshihiro Banno; Tohru Yamashita; Tatsuhiko Fujimoto; Satoru Oi; Yusuke Moritoh; Tomoko Asakawa; Osamu Kataoka; Koji Takeuchi; Nobuhiro Suzuki; Koji Ikedo; Takuo Kosaka; Shigetoshi Tsubotani; Akiyoshi Tani; Miyuki Funami; Michiko Amano; Yoshio Yamamoto; Kathleen Aertgeerts; Jason Yano; Hironobu Maezaki

We have previously discovered nicotinic acid derivative 1 as a structurally novel dipeptidyl peptidase IV (DPP-4) inhibitor. In this study, we obtained the X-ray co-crystal structure between nicotinic acid derivative 1 and DPP-4. From these X-ray co-crystallography results, to achieve more potent inhibitory activity, we targeted Arg125 as a potential amino acid residue because it was located near the pyridine core, and some known DPP-4 inhibitors were reported to interact with this residue. We hypothesized that the guanidino group of Arg125 could interact with two hydrogen-bond acceptors in a bidentate manner. Therefore, we designed a series of 3-pyridylacetamide derivatives possessing an additional hydrogen-bond acceptor that could have the desired bidentate interaction with Arg125. We discovered the dihydrochloride of 1-{[5-(aminomethyl)-2-methyl-4-(4-methylphenyl)-6-(2-methylpropyl)pyridin-3-yl]acetyl}-l-prolinamide (13j) to be a potent and selective DPP-4 inhibitor that could interact with the guanidino group of Arg125 in a unique bidentate manner.


Bioorganic & Medicinal Chemistry | 2011

Discovery of potent, selective, and orally bioavailable quinoline-based dipeptidyl peptidase IV inhibitors targeting Lys554.

Hironobu Maezaki; Yoshihiro Banno; Yasufumi Miyamoto; Yuusuke Moritou; Tomoko Asakawa; Osamu Kataoka; Koji Takeuchi; Nobuhiro Suzuki; Koji Ikedo; Takuo Kosaka; Masako Sasaki; Shigetoshi Tsubotani; Akiyoshi Tani; Miyuki Funami; Yoshio Yamamoto; Michiko Tawada; Kathleen Aertgeerts; Jason Yano; Satoru Oi

Dipeptidyl peptidase IV (DPP-4) inhibition is a validated therapeutic option for type 2 diabetes, exhibiting multiple antidiabetic effects with little or no risk of hypoglycemia. In our studies involving non-covalent DPP-4 inhibitors, a novel series of quinoline-based inhibitors were designed based on the co-crystal structure of isoquinolone 2 in complex with DPP-4 to target the side chain of Lys554. Synthesis and evaluation of designed compounds revealed 1-[3-(aminomethyl)-4-(4-methylphenyl)-2-(2-methylpropyl)quinolin-6-yl]piperazine-2,5-dione (1) as a potent, selective, and orally active DPP-4 inhibitor (IC₅₀=1.3 nM) with long-lasting ex vivo activity in dogs and excellent antihyperglycemic effects in rats. A docking study of compound 1 revealed a hydrogen-bonding interaction with the side chain of Lys554, suggesting this residue as a potential target site useful for enhancing DPP-4 inhibition.


Bioorganic & Medicinal Chemistry | 2012

Design and synthesis of novel DFG-out RAF/vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors: 3. Evaluation of 5-amino-linked thiazolo[5,4-d]pyrimidine and thiazolo[5,4-b]pyridine derivatives.

Masaaki Hirose; Masanori Okaniwa; Tohru Miyazaki; Takashi Imada; Tomohiro Ohashi; Yuta Tanaka; Takeo Arita; Masato Yabuki; Tomohiro Kawamoto; Shunichirou Tsutsumi; Akihiko Sumita; Terufumi Takagi; Bi-Ching Sang; Jason Yano; Kathleen Aertgeerts; Sei Yoshida; Tomoyasu Ishikawa

Our aim was to discover RAF/vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors that possess strong activity and sufficient oral absorption, and thus, we selected a 5-amino-linked thiazolo[5,4-d]pyrimidine derivative as the lead compound because of its potential kinase inhibitory activities and its desired solubility. The novel tertiary 1-cyano-1-methylethoxy substituent was designed to occupy the hydrophobic region of back pocket of BRAF on the basis of the X-ray co-crystal structure data of BRAF. In addition, we found that N-methylation of the amine linker could control the twisted molecular conformation leading to improved solubility. These approaches produced N-methyl thiazolo[5,4-b]pyridine-5-amine derivative 5. To maximize the in vivo efficacy, we attempted salt formation of 5. Our result indicated that the besylate monohydrate salt form (5c) showed significant improvement of both solubility and oral absorption. Owing to the improved physicochemical properties, compound 5c demonstrated regressive antitumor efficacy in a HT-29 xenograft model.


Molecular Cancer Therapeutics | 2013

Abstract C255: Discovery of TAK-632: A selective kinase inhibitor of pan-RAF with potent antitumor activity against BRAF and NRAS mutant melanomas.

Masanori Okaniwa; Masaaki Hirose; Takeo Arita; Masato Yabuki; Akito Nakamura; Terufumi Takagi; Tomohiro Kawamoto; Noriko Uchiyama; Akihiko Sumita; Shunichirou Tsutsumi; Tsuneaki Tottori; Yoshitaka Inui; Bi-Ching Sang; Jason Yano; Kathleen Aertgeerts; Sei Yoshida; Tomoyasu Ishikawa

The RAF family kinases play critical roles in cancer progression. Recently, BRAF selective inhibitors have shown significant clinical efficacy in melanoma patients bearing oncogenic BRAFV600E mutation. However, several studies reported that RAF inhibitors instinctively transactivate RAF homodimers (CRAF-CRAF) or heterodimers (CRAF-BRAF(wt)) and activate RAS dependent MAPK signaling. Along with this mechanism, it has been reported that selective BRAF inhibitors have not shown potent anti-proliferative activity against cancer cell lines such as NRAS mutant melanoma in which RAS dependent MAPK signaling is activated (Hong Yang et al., Cancer Res., 2010, 70, 5518-5527). However, our initial investigation using fibroblast CsFb (BRAFwt) cells indicated that phosphorylation of MEK and ERK was inhibited by some DFG-out inhibitors, but not by DFG-in inhibitors. These results led to the hypothesis: continuous inhibition of pan-RAF (BRAF and CRAF) with DFG-out type inhibitors could suppress the feedback activation. Here we report the discovery and characterization of pan-RAF inhibitor TAK-632. We designed novel 1,3-benzothiazole class derivatives using knowledge of structure-activity relationships gained from studies of our thiazolo[5,4-b]pyridine class RAF/VEGFR2 inhibitor (Masanori Okaniwa et al., J. Med. Chem., 2012, 55, 3452-3478). To enrich RAF kinase selectivity vs. VEGFR2, we utilized the cocrystal structures of our lead compound with both BRAF and VEGFR2. Eventually, we designed and selected 7-cyano derivative TAK-632 as a development candidate. Cocrystal structure analysis of BRAF bearing TAK-632 revealed that accommodation of the 7-cyano group into the BRAF-selectivity pocket and the 3-(trifluoromethyl)phenyl acetamide moiety into the hydrophobic back pocket of BRAF in the DFG-out conformation contributed to enhanced RAF inhibition and selectivity vs. VEGFR2. Reflecting its potent pan-RAF inhibition (IC50: BRAFV600E 2.4 nM, CRAF 1.4 nM) and slow dissociation (koff) profile measured by surface plasmon resonance (SPR) spectroscopy, TAK-632 demonstrated significant cellular activity against mutated BRAF or mutated NRAS cancer cell lines. Furthermore, in both A375 (BRAFV600E) and HMVII (NRASQ61K) xenograft models in rats, TAK-632 demonstrated regressive antitumor activity by twice daily, 14-day repetitive administration without significant body weight loss. In conclusion, these results raise the possibility of using slow off-rate pan-RAF inhibitors such as TAK-632 for the treatment of human cancers harboring either BRAFV600E or NRAS mutant. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C255. Citation Format: Masanori Okaniwa, Masaaki Hirose, Takeo Arita, Masato Yabuki, Akito Nakamura, Terufumi Takagi, Tomohiro Kawamoto, Noriko Uchiyama, Akihiko Sumita, Shunichirou Tsutsumi, Tsuneaki Tottori, Yoshitaka Inui, Bi-Ching Sang, Jason Yano, Kathleen Aertgeerts, Sei Yoshida, Tomoyasu Ishikawa. Discovery of TAK-632: A selective kinase inhibitor of pan-RAF with potent antitumor activity against BRAF and NRAS mutant melanomas. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C255.


Journal of Biological Chemistry | 2005

Structural and kinetic analysis of the substrate specificity of human fibroblast activation protein Alpha

Kathleen Aertgeerts; I Levin; L Shi; G.P Snell; Andy Jennings; G.S Prasad; Y Zhang; M.L Kraus; S Salakian; Sridhar; R Wijnands; Mike Tennant

Collaboration


Dive into the Kathleen Aertgeerts's collaboration.

Top Co-Authors

Avatar

Jason Yano

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Akiyoshi Tani

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Bi-Ching Sang

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Hironobu Maezaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Koji Ikedo

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Koji Takeuchi

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Miyuki Funami

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Osamu Kataoka

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Robert J. Skene

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge