Lakshmaiah Gingipalli
AstraZeneca
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Lakshmaiah Gingipalli.
Bioorganic & Medicinal Chemistry Letters | 2012
Tao Wang; Michael A. Block; Scott Cowen; Audrey Davies; Erik Devereaux; Lakshmaiah Gingipalli; Jeffrey W. Johannes; Nicholas A. Larsen; Qibin Su; Julie A. Tucker; David Whitston; Jiaquan Wu; Hai-Jun Zhang; Michael Zinda; Claudio Chuaqui
The design, synthesis and biological evaluation of a series of azabenzimidazole derivatives as TBK1/IKKε kinase inhibitors are described. Starting from a lead compound 1a, iterative design and SAR exploitation of the scaffold led to analogues with nM enzyme potencies against TBK1/IKKε. These compounds also exhibited excellent cellular activity against TBK1. Further structure-based design to improve selectivity over CDK2 and Aurora B resulted in compounds such as 5b-e. These probe compounds will facilitate study of the complex cancer biology of TBK1 and IKKε.
ACS Medicinal Chemistry Letters | 2015
Jeffrey W. Johannes; Lynsie Almeida; Bernard Barlaam; P. Ann Boriack-Sjodin; Robert Casella; Rosemary A. Croft; Allan Dishington; Lakshmaiah Gingipalli; Chungang Gu; Janet Hawkins; Jane L. Holmes; Tina Howard; Jian Huang; Stephanos Ioannidis; Steven Kazmirski; Michelle L. Lamb; Thomas M. McGuire; Jane E. Moore; Derek Ogg; Anil Patel; Kurt Gordon Pike; Timothy Pontz; Graeme R. Robb; Nancy Su; Haiyun Wang; Xiaoyun Wu; Hai-Jun Zhang; Yue Zhang; Xiaolan Zheng; Tao Wang
The canonical Wnt pathway plays an important role in embryonic development, adult tissue homeostasis, and cancer. Germline mutations of several Wnt pathway components, such as Axin, APC, and ß-catenin, can lead to oncogenesis. Inhibition of the poly(ADP-ribose) polymerase (PARP) catalytic domain of the tankyrases (TNKS1 and TNKS2) is known to inhibit the Wnt pathway via increased stabilization of Axin. In order to explore the consequences of tankyrase and Wnt pathway inhibition in preclinical models of cancer and its impact on normal tissue, we sought a small molecule inhibitor of TNKS1/2 with suitable physicochemical properties and pharmacokinetics for hypothesis testing in vivo. Starting from a 2-phenyl quinazolinone hit (compound 1), we discovered the pyrrolopyrimidinone compound 25 (AZ6102), which is a potent TNKS1/2 inhibitor that has 100-fold selectivity against other PARP family enzymes and shows 5 nM Wnt pathway inhibition in DLD-1 cells. Moreover, compound 25 can be formulated well in a clinically relevant intravenous solution at 20 mg/mL, has demonstrated good pharmacokinetics in preclinical species, and shows low Caco2 efflux to avoid possible tumor resistance mechanisms.
Bioorganic & Medicinal Chemistry Letters | 2014
Jeffrey W. Johannes; Claudio Chuaqui; Scott Cowen; Erik Devereaux; Lakshmaiah Gingipalli; Audrey Molina; Tao Wang; David Whitston; Xiaoyun Wu; Hai-Jun Zhang; Michael Zinda
The discovery and optimization of a series of 6-aryl-azabenzimidazole inhibitors of TBK1 and IKK-ε is described. Various internal azabenzimidazole leads and reported TBK1/IKK-ε inhibitors were docked into a TBK1 homology model. The resulting overlays inspired a focused screen of 6-substituted azabenzimidazoles against TBK1/IKK-ε. This screen resulted in initial hit compound 3. The TBK1/IKK-ε enzyme and cell potency of this compound was further improved using structure guided drug design. Systematic exploration of the C6 aryl group led to compound 19, a potent inhibitor of TBK1 with selectivity against cell cycle kinases CDK2 and Aurora B. Further elaboration and optimization gave compound 25, a single digit nM inhibitor of TBK1. These compounds may serve as in vitro probes to evaluate TBK1/IKK-ε as an oncology target.
Bioorganic & Medicinal Chemistry Letters | 2015
Kerry E. Murphy-Benenato; Lakshmaiah Gingipalli; P. Ann Boriack-Sjodin; Gabriel Martinez-Botella; Dan Carcanague; Charles J. Eyermann; Madhu Gowravaram; Jenna Harang; Michael R. Hale; Georgine Ioannidis; Harris Jahic; Amy Kutschke; Valerie A. Laganas; James T. Loch; Matthew D. Miller; Herbert Oguto; Sahil Joe Patel
Two novel compounds, pyridopyrimidines (1) and naphthyridines (2) were identified as potent inhibitors of bacterial NAD(+)-dependent DNA ligase (Lig) A in a fragment screening. SAR was guided by molecular modeling and X-ray crystallography. It was observed that the diaminonitrile pharmacophore made a key interaction with the ligase enzyme, specifically residues Glu114, Lys291, and Leu117. Synthetic challenges limited opportunities for diversification of the naphthyridine core, therefore most of the SAR was focused on a pyridopyrimidine scaffold. The initial diversification at R(1) improved both enzyme and cell potency. Further SAR developed at the R(2) position using the Negishi cross-coupling reaction provided several compounds, among these compounds 22g showed good enzyme potency and cellular potency.
ACS Medicinal Chemistry Letters | 2016
Edward J. Hennessy; Claudio Chuaqui; Susan Ashton; Nicola Colclough; Darren Cross; J.E. Debreczeni; Cath Eberlein; Lakshmaiah Gingipalli; Teresa Klinowska; Jonathan P. Orme; Li Sha; Xiaoyun Wu
A novel series of covalent inhibitors of EGFR (epidermal growth factor receptor) kinase was discovered through a combination of subset screening and structure-based design. These compounds preferentially inhibit mutant forms of EGFR (activating mutant and T790M mutant) over wild-type EGFR in cellular assays measuring EGFR autophosphorylation and proliferation, suggesting an improved therapeutic index in non-small cell lung cancer patients would be achievable relative to established EGFR inhibitors. We describe our design approaches, resulting in the identification of the lead compound 5, and our efforts to develop an understanding of the structure-activity relationships within this series. In addition, strategies to overcome challenges around metabolic stability and aqueous solubility are discussed. Despite limitations in its physical properties, 5 is orally bioavailable in mice and demonstrates pronounced antitumor activity in in vivo models of mutant EGFR-driven cancers.
Bioorganic & Medicinal Chemistry Letters | 2018
Lakshmaiah Gingipalli; Michael Howard Block; Larry Bao; Emma L. Cooke; Les A. Dakin; Christopher R. Denz; Andrew D. Ferguson; Jeffrey W. Johannes; Nicholas A. Larsen; Paul Lyne; Timothy Pontz; Tao Wang; Xiaoyun Wu; Allan Wu; Hai-Jun Zhang; Xiaolan Zheng; James E. Dowling; Michelle Lamb
The design and synthesis of a novel series of 2,6-disubstituted pyrazine derivatives as CK2 kinase inhibitors is described. Structure-guided optimization of a 5-substituted-3-thiophene carboxylic acid screening hit (3a) led to the development of a lead compound (12b), which shows inhibition in both enzymatic and cellular assays. Subsequent design and hybridization efforts also led to the unexpected identification of analogs with potent PIM kinase activity (14f).
Molecular Cancer Therapeutics | 2015
Dorin Toader; Jay Harper; Chris Lloyd; Rose Marwood; David Bannister; Shenlan Mao; Cui Chen; Haihon (Helen) Zhong; Vahe Bedian; Fengjiang Wang; Lakshmaiah Gingipalli; Melisa Vasbinder; Pamela Thompson; Ryan Fleming; Byniam Bezabeh; Nazzareno Dimasi; Changshou Gao; Adeela Kamal
Antibody drug conjugates (ADCs) combine the specificity of antibodies with the potency of small molecule cytotoxic drugs and have the potential to provide significant efficacy as a treatment for cancer. The objective of this work was to identify potent new cytotoxic ADC payloads that can be used to target diverse tumor types. Here we report for the first time the discovery of fully synthetic tubulysin payloads which belong to a class of highly cytotoxic natural products that disrupt the cellular microtubule network leading to apoptosis of tumor cells. Our fully synthetic tubulysin payloads are comprised of: (i) a tubulysin warhead that displays pM potency, (ii) a protease cleavable amino-acid sequence and (iii) a tether bearing a reactive maleimide group. Tubulysin-based ADCs were generated via site-specific conjugation of these payloads to cysteines engineered into antibodies against cancer antigen target oncofetal protein 5T4. The resulting ADCs showed potent in vitro cell killing and in vivo efficacy in multiple solid tumor xenograft models including prostate cancer, non-small cell lung adenocarcinoma, breast cancer and gastric carcinoma. Furthermore, specific structural features of the tubulysin warhead, linker design and antibody engineering were shown to impact the overall in vitro and in vivo properties of the ADCs. Thus, these synthetic tubulysin payloads represent novel microtubule network disrupting compounds that display potent preclinical anti-tumor activity as an ADC that could be advanced to the clinic. Citation Format: Dorin Toader, Jay Harper, Chris Lloyd, Rose Marwood, David Bannister, Shenlan Mao, Cui (Tracy) Chen, Haihon (Helen) Zhong, Vahe Bedian, Fengjiang Wang, Lakshmaiah Gingipalli, Melisa Vasbinder, Pamela Thompson, Ryan Fleming, Byniam Bezabeh, Nazzareno Dimasi, Changshou Gao, Adeela Kamal. Discovery of tubulysin payloads for antibody drug conjugates with potent in vitro activity and in vivo efficacy in solid tumor models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B170.
Cancer Research | 2011
Emma-Louise Cooke; Marat Alimzhanov; Lynsie Almeida; Larry Bao; Vahe Bedian; Jeffrey L. Brown; Chun Deng; Christopher R. Denz; Lakshmaiah Gingipalli; Jeffrey W. Johannes; Steven Kazmirski; Michelle Lamb; Zhong-Ying Liu; Vicki Racicot; Nancy Su; Haiyun Wang; Tao Wang; Xiaoyun Wu; Lihua Yu; Hai-Jun Zhang; Weijia Zheng; Paul Lyne
Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Deregulated Wnt signaling has been implicated in a wide range of cancer types, both through truncating mutations in the tumor suppressor protein Adenomatous Polyposis Coli (APC), most prevalent in colorectal cancer, as well as overexpression of Wnt ligands and receptors. Canonical Wnt signaling involves the regulated degradation of the beta catenin protein. In the absence of Wnt signaling, cytosolic beta catenin levels are maintained at low levels, as a result of its phosphorylation-dependent ubiquitination and subsequent proteasomal degradation. This degradation is mediated by the ‘destruction complex’, whose constituents include Glycogen Synthase Kinase 3 (GSK3β), Casein Kinase 1 (CK1) and the scaffolding proteins Axin and APC. The efficient assembly of this complex depends upon the steady state levels of its components, such as Axin, which when overexpressed has been shown to decrease beta catenin levels and inhibit Wnt signaling. Recent reports suggest that the poly-ADP-ribosylating Tankyrase enzymes can bind to and regulate Axin levels, hence providing promising targets for treating Wnt-dependent tumors. We have identified potent, low nM small molecule inhibitors of the Wnt pathway, which inhibit Tankyrases 1 and 2, stabilize Axin, deplete beta catenin protein levels and modulate a set of Wnt-regulated genes in colorectal cell lines in a similar manner as seen when using siRNA to beta catenin. Furthermore, using siRNAs to Tankyrase 1 and 2 we have shown that co-depletion of both enzymes is required for Axin stabilization and Wnt pathway inhibition. We have also successfully demonstrated that our lead molecules cause prolonged Axin stabilization and Wnt pathway inhibition in vivo following oral BID dosing in colon xenografts carrying APC mutations. However, in contrast to previous reports, using molecular tools and our lead compounds (e.g. inducible Axin cell lines, compound rescue experiments using siRNAs to Axin 1/2), we have shown that Axin stabilization has no effect on the viability/proliferation of APC mutant colorectal cell lines, under conditions where siRNA to beta catenin was able to cause significant growth inhibition. Some potential mechanistic rationales for this lack of phenotypic effect will be presented. Understanding the consequences of Tankyrase inhibition in vivo in relevant disease models will be important to establish the clinical utility of these molecules in colorectal cancer and beyond. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5196. doi:10.1158/1538-7445.AM2011-5196
Journal of Medicinal Chemistry | 2016
Dorin Toader; Fengjiang Wang; Lakshmaiah Gingipalli; Melissa Vasbinder; Mark Roth; Shenlan Mao; Michael Howard Block; Jay Harper; Sambaiah Thota; Mei Su; Jianquo Ma; Vahe Bedian; Adeela Kamal
Tetrahedron Letters | 2015
Michael R. Hale; Claire Brassington; Dan Carcanague; Kevin J. Embrey; Charles J. Eyermann; Robert A. Giacobbe; Lakshmaiah Gingipalli; Madhusudhan Gowravaram; Jenna Harang; Tina Howard; Georgine Ioannidis; Haris Jahić; Amy Kutschke; Valerie A. Laganas; James T. Loch; Matthew D. Miller; Kerry E. Murphy-Benenato; Herbert Oguto; Ludovic R. Otterbein; Sahil Joe Patel; Adam B. Shapiro; P. Ann Boriack-Sjodin