Network


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

Hotspot


Dive into the research topics where Shailaja Kasibhatla is active.

Publication


Featured researches published by Shailaja Kasibhatla.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974

Jun Liu; Shifeng Pan; Mindy H. Hsieh; Nicholas Ng; Fangxian Sun; Tao Wang; Shailaja Kasibhatla; Alwin Schuller; Allen Li; Dai Cheng; Jie Li; Celin Tompkins; Anne Marie Pferdekamper; Auzon Steffy; Jane Cheng; Colleen Kowal; Van Phung; Gui-Rong Guo; Yan Wang; Martin P. Graham; Shannon Flynn; J. Chad Brenner; Chun Li; M. Cristina Villarroel; Peter G. Schultz; Xu Wu; Peter McNamara; William R. Sellers; Lilli Petruzzelli; Anthony L. Boral

Significance Targeting the Wnt pathway in cancer is an attractive therapeutic approach. However, success has been limited because of the lack of effective therapeutic agents and the lack of biomarkers to define the patient population that would benefit from such a therapy. Herein, we report the discovery of LGK974, a drug that targets Porcupine, a Wnt-specific acyltransferase. We show that LGK974 potently inhibits Wnt signaling, has strong efficacy in rodent tumor models, and is well-tolerated. We also show that head and neck cancer cell lines with loss-of-function mutations in the Notch signaling pathway have a high response rate to LGK974. Together, these findings provide a strategy and tools for targeting Wnt-driven cancer. Wnt signaling is one of the key oncogenic pathways in multiple cancers, and targeting this pathway is an attractive therapeutic approach. However, therapeutic success has been limited because of the lack of therapeutic agents for targets in the Wnt pathway and the lack of a defined patient population that would be sensitive to a Wnt inhibitor. We developed a screen for small molecules that block Wnt secretion. This effort led to the discovery of LGK974, a potent and specific small-molecule Porcupine (PORCN) inhibitor. PORCN is a membrane-bound O-acyltransferase that is required for and dedicated to palmitoylation of Wnt ligands, a necessary step in the processing of Wnt ligand secretion. We show that LGK974 potently inhibits Wnt signaling in vitro and in vivo, including reduction of the Wnt-dependent LRP6 phosphorylation and the expression of Wnt target genes, such as AXIN2. LGK974 is potent and efficacious in multiple tumor models at well-tolerated doses in vivo, including murine and rat mechanistic breast cancer models driven by MMTV–Wnt1 and a human head and neck squamous cell carcinoma model (HN30). We also show that head and neck cancer cell lines with loss-of-function mutations in the Notch signaling pathway have a high response rate to LGK974. Together, these findings provide both a strategy and tools for targeting Wnt-driven cancers through the inhibition of PORCN.


Cancer Research | 2016

EGF816 Exerts Anticancer Effects in Non–Small Cell Lung Cancer by Irreversibly and Selectively Targeting Primary and Acquired Activating Mutations in the EGF Receptor

Yong Jia; José Juárez; Jie Li; Mari Manuia; Matthew J. Niederst; Celin Tompkins; Noelito Timple; Mei-Ting Vaillancourt; AnneMarie Culazzo Pferdekamper; Elizabeth L. Lockerman; Chun Li; Jennifer Anderson; Carlotta Costa; Debbie Liao; Eric Murphy; Michael DiDonato; Badry Bursulaya; Gerald Lelais; Jordi Barretina; Matthew McNeill; Robert Epple; Thomas H. Marsilje; Nuzhat Pathan; Jeffrey A. Engelman; Pierre-Yves Michellys; Peter McNamara; Jennifer L. Harris; Steven L. Bender; Shailaja Kasibhatla

Non-small cell lung cancer patients carrying oncogenic EGFR mutations initially respond to EGFR-targeted therapy, but later elicit minimal response due to dose-limiting toxicities and acquired resistance. EGF816 is a novel, irreversible mutant-selective EGFR inhibitor that specifically targets EGFR-activating mutations arising de novo and upon resistance acquisition, while sparing wild-type (WT) EGFR. EGF816 potently inhibited the most common EGFR mutations L858R, Ex19del, and T790M in vitro, which translated into strong tumor regressions in vivo in several patient-derived xenograft models. Notably, EGF816 also demonstrated antitumor activity in an exon 20 insertion mutant model. At levels above efficacious doses, EGF816 treatment led to minimal inhibition of WT EGFR and was well tolerated. In single-dose studies, EGF816 provided sustained inhibition of EGFR phosphorylation, consistent with its ability for irreversible binding. Furthermore, combined treatment with EGF816 and INC280, a cMET inhibitor, resulted in durable antitumor efficacy in a xenograft model that initially developed resistance to first-generation EGFR inhibitors via cMET activation. Thus, we report the first preclinical characterization of EGF816 and provide the groundwork for its current evaluation in phase I/II clinical trials in patients harboring EGFR mutations, including T790M.


ACS Medicinal Chemistry Letters | 2016

Discovery of Pyridinyl Acetamide Derivatives as Potent, Selective, and Orally Bioavailable Porcupine Inhibitors

Dai Cheng; Jun Liu; Dong Han; Guobao Zhang; Wenqi Gao; Mindy H. Hsieh; Nicholas Ng; Shailaja Kasibhatla; Celin Tompkins; Jie Li; Auzon Steffy; Fangxian Sun; Chun Li; H. Martin Seidel; Jennifer L. Harris; Shifeng Pan

Blockade of aberrant Wnt signaling is an attractive therapeutic approach in multiple cancers. We developed and performed a cellular high-throughput screen for inhibitors of Wnt secretion and pathway activation. A lead structure (GNF-1331) was identified from the screen. Further studies identified the molecular target of GNF-1331 as Porcupine, a membrane bound O-acyl transferase. Structure-activity relationship studies led to the discovery of a novel series of potent and selective Porcupine inhibitors. Compound 19, GNF-6231, demonstrated excellent pathway inhibition and induced robust antitumor efficacy in a mouse MMTV-WNT1 xenograft tumor model.


Molecular Cancer Therapeutics | 2011

Abstract B232: Activity of a potent and selective phase I ALK inhibitor LDK378 in naive and crizotinib-resistant preclinical tumor models.

Nanxin Li; Pierre-Yves Michellys; Sungjoon Kim; AnneMarie Culazzo Pferdekamper; Jie Li; Shailaja Kasibhatla; Celin Tompkins; Auzon Steffy; Allen Li; Frank Sun; Xiuying Sun; Su Hua; Ralph Tiedt; Yelena Sarkisova; Thomas H. Marsilje; Peter McNamara; Jennifer L. Harris

A c-MET/ALK kinase inhibitor crizotinib has demonstrated significant activity in lung cancer patients with EML4-ALK in clinical studies. However relapse (or acquired resistance) has also been reported. We have developed crizotinib resistant tumor models and used the models to evaluate the ALK inhibitor LDK378. LDK378 is a potent and selective ALK inhibitor that does not cross react with c-MET. In a mouse xenograft tumor model derived from the EML4-ALK+ lung cancer cell line NCI-H2228, LDK378 caused complete tumor regression when dosed orally at 25 mg/kg/day. After tumor bearing animals had been treated with LDK378 at 50 mg/kg/day for 14 days, remission was maintained for more than 4 months. In several NCI-H2228 tumor models that were induced to become resistant to crizotinib, LDK378 demonstrated efficacy at 50 mg/kg/day. Based on 4-wk GLP toxicology studies the drug exposure associated with this dose is predicted to be below the exposure at the MTD in humans. ALK resistance mutations reported in crizotinib relapsed patients were also found in the crizotinib resistant NCI-H2228 tumor models. The results from these preclinical studies suggest that LDK378 may be active in crizotinib-relapsed patients. A phase I clinical study of LDK378 has recently begun in both crizotinib-relapsed and crizotinib-naive patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr B232.


eLife | 2017

Combined ALK and MDM2 inhibition increases antitumor activity and overcomes resistance in human ALK mutant neuroblastoma cell lines and xenograft models

Hui Qin Wang; Ensar Halilovic; Xiaoyan Li; Jinsheng Liang; Yichen Cao; Daniel Rakiec; David A. Ruddy; Sébastien Jeay; Jens Wuerthner; Noelito Timple; Shailaja Kasibhatla; Nanxin Li; Juliet Williams; William R. Sellers; Alan Huang; Fang Li

The efficacy of ALK inhibitors in patients with ALK-mutant neuroblastoma is limited, highlighting the need to improve their effectiveness in these patients. To this end, we sought to develop a combination strategy to enhance the antitumor activity of ALK inhibitor monotherapy in human neuroblastoma cell lines and xenograft models expressing activated ALK. Herein, we report that combined inhibition of ALK and MDM2 induced a complementary set of anti-proliferative and pro-apoptotic proteins. Consequently, this combination treatment synergistically inhibited proliferation of TP53 wild-type neuroblastoma cells harboring ALK amplification or mutations in vitro, and resulted in complete and durable responses in neuroblastoma xenografts derived from these cells. We further demonstrate that concurrent inhibition of MDM2 and ALK was able to overcome ceritinib resistance conferred by MYCN upregulation in vitro and in vivo. Together, combined inhibition of ALK and MDM2 may provide an effective treatment for TP53 wild-type neuroblastoma with ALK aberrations. DOI: http://dx.doi.org/10.7554/eLife.17137.001


Cancer Research | 2011

PD08-11: Targeting Porcupine, a Critical Node for Wnt Signalling in Cancer.

Jun Liu; Shifeng Pan; F Sun; Shailaja Kasibhatla; Alwin Schuller; A Li; Chun Li; X Cui; T Wang; M Villarroel; Margaret E. McLaughlin; William R. Sellers; Anthony L. Boral; Jennifer L. Harris

Wnt ligands were first discovered based upon their transforming activity in the setting of mouse mammary tumor virus (MMTV) induced mammary carcinogenesis. Wnt signalling is required for normal development of the mammary gland and dysregulated Wnt signalling is implicated in cancers of the breast and colon among others. Furthermore, lobular breast cancer commonly shows reduced expression of E-cadherin that can lead to release of membrane-bound b-catenin into the cytoplasm and potentially increase Wnt signalling in the presence of Wnt ligand. To date, however, low-molecular weight therapeutic inhibitors of the Wnt pathway have not been developed. We set out to discover key points of intervention in the Wnt pathway and to develop low molecular weight inhibitors against such nodes. To this end, a cellular high-throughput screen for small molecule inhibitors of the Wnt pathway was performed. In this assay, cells secreting Wnt3a were co-cultured with cells harboring a luciferase reporter driven by the Wnt/beta-catenin responsive elements. Potent and specific Wnt pathway inhibitors were identified from this screen. To discern the mechanism of action of these compounds, a lead inhibitor was radiolabeled and used to probe protein targets in a radioligand binding assay. From these assays a candidate target of the inhibitor was identified as Porcupine. Porcupine is a membrane bound O-acyltransferase enzyme that is required for and dedicated to palmitoylating Wnt ligands, a necessary step in the process of Wnt ligand secretion. Inhibition of Porcupine blocks Wnt dependent activities, including LRP6 phosphorylation and the expression of Wnt target genes, such as Axin2, which in turn reduces the growth of cancer cells dependent on autocrine or paracrine Wnt signaling. A selective and orally bioavailable Porcupine inhibitor (NVP-LGK974) that inhibits the Wnt pathway in vitro and in vivo was developed through medicinal chemistry optimization. In preclinical evaluation, the compound robustly suppressed Wnt pathway signaling in vivo resulting in tumor regression in a murine breast cancer model driven by MMTV-Wnt1. In addition, this inhibitor attenuated tumor growth as a single agent and induced tumor regression in combination with Taxol in a human primary breast tumor model. Wnt signalling is also important for the maintenance and homeostasis of normal tissues including gastrointestinal tissue. However, preclinical rodent efficacy models of cancer demonstrate that intermittent pharmacological inhibition of Porcupine can be effective against the tumor and mostly spare the normal gastrointestinal tissue, with an acceptable therapeutic window. A Phase I dose escalation study of NVP-LGK974 in patients with melanoma and lobular breast cancer to evaluate the safety, tolerability, PK, and PD properties is expected to begin this year. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr PD08-11.


Cancer Research | 2016

Abstract 1445: STING activation in the tumor microenvironment with a synthetic human STING-activating cyclic dinucleotide leads to potent anti-tumor immunity

Laura Hix Glickman; David B. Kanne; Shailaja Kasibhatla; Jie Li; AnneMarie Culazzo Pferdekamper; Kelsey Sivick Gauthier; Weiwen Deng; Anthony L. Desbien; George E. Katibah; Justin J. Leong; Leonard Sung; Ken Metchette; Chudi Ndubaku; Lianxing Zheng; Charles Y. Cho; Yan Feng; Jeffrey Mckenna; John A. Tallarico; Steven L. Bender; Thomas W. Dubensky; Sarah M. McWhirter

Stimulator of Interferon Genes (STING) is a critical signaling sensor of the innate immune system. STING binds cyclic dinucleotides (CDN) produced by an intracellular enzyme in response to presence of intracellular DNA, including tumor-derived DNA. STING-mediated production of host type I interferon within the tumor microenvironment (TME) leads to the priming and activation of systemic tumor antigen-specific CD8+ T-cell immunity and tumor regression. A novel synthetic CDN derivative (ADU-S100), with superior STING-activating and anti-tumor properties, was developed for clinical translation. ADU-S100 has enhanced cellular uptake properties and stability, as compared to bacterial- and mammalian-derived CDNs. Induced cytokine expression from a panel of donor human peripheral blood mononuclear cells (PBMCs) expressing a variety of STING alleles, including a homozygous haplotype for the most refractory human allele (R232H), indicate that ADU-S100 activates STING across a diverse human population. Direct engagement of STING through intratumoral (IT) administration of ADU-S100 results in effective anti-tumor therapy and long-term survival in various mouse syngeneic tumor models. IT injection of ADU-S100 also generates substantial systemic immune responses capable of rejecting distant metastases and provides long-lived immunologic memory. Mechanistic studies demonstrate that STING-mediated anti-tumor immunity is due in part to an acute pro-inflammatory cytokine response as well as a tumor-specific CD8+ T cell response. Anti-tumor efficacy is enhanced by combination with immune checkpoint inhibitors, for example anti-PD1, informing future clinical development. By virtue of the ability to elicit innate and T cell-mediated anti-tumor immunity in the TME, these results demonstrate that CDNs have high translational potential for the treatment of patients with advanced/metastatic solid tumors. Citation Format: Laura Hix Glickman, David B. Kanne, Shailaja Kasibhatla, Jie Li, AnneMarie Culazzo Pferdekamper, Kelsey Sivick Gauthier, Weiwen Deng, Anthony L. Desbien, George E. Katibah, Justin J. Leong, Leonard Sung, Ken Metchette, Chudi Ndubaku, Lianxing Zheng, Charles Cho, Yan Feng, Jeffrey M. McKenna, John A. Tallarico, Steven L. Bender, Thomas W. Dubensky, Sarah M. McWhirter. STING activation in the tumor microenvironment with a synthetic human STING-activating cyclic dinucleotide leads to potent anti-tumor immunity. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1445.


Cancer Research | 2015

Abstract 2585: Discovery of a potent covalent mutant-selective EGFR inhibitor - the journey from high throughput screening to EGF816

Gerald Lelais; Robert Epple; Pierre-Yves Michellys; Thomas H. Marsilje; Yun Long; Matthew McNeill; Bei Chen; Wenshuo Lu; Badry Bursulaya; Michael DiDonato; Yong Jia; Shailaja Kasibhatla; Chun Li; Igor Matushansky; Steven L. Bender

Epidermal growth factor receptor (EGFR) is a validated therapeutic target for lung cancer. First and second generation EGFR inhibitors (e.g., gefitinib, erlotinib and afatinib) have revolutionized treatment paradigms of non-small cell lung cancer (NSCLC) patients with oncogenic EGFR mutations. The use of EGFR tyrosine kinase inhibitors (TKI) provides superior efficacy compared to chemotherapy in patients with EGFR L858R or exon 19 deletion tumors. However, resistance inevitably develops after 8-12 months of treatment; most commonly via a secondary T790M point mutation at the gatekeeper residue of EGFR. Furthermore, responses are hindered due to treatment intolerance in the form of rash and diarrhea that are mediated by simultaneous inhibition of wild-type (WT) EGFR at doses required for mutant EGFR suppression. To overcome these limitations, we initiated a project to identify mutant-selective EGFR inhibitors that potently inhibit both activating and T790M resistance EGFR mutations while sparing WT EGFR. In this presentation, we report our medicinal chemistry approach and optimization that led to the discovery of EGF816, a selective and potent covalent mutant-selective EGFR inhibitor with single digit nanomolar cellular target modulation on both activating and T790M resistance mutations. In addition, we will also report validated clinical efficacy data from the first patient treated with EGF816. Citation Format: Gerald Lelais, Robert Epple, Pierre-Yves Michellys, Thomas H. Marsilje, Yun Long, Matthew McNeill, Bei Chen, Wenshuo Lu, Badry Bursulaya, Michael DiDonato, Yong Jia, Shailaja Kasibhatla, Chun Li, Igor Matushansky, Steven Bender. Discovery of a potent covalent mutant-selective EGFR inhibitor - the journey from high throughput screening to EGF816. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2585. doi:10.1158/1538-7445.AM2015-2585


Cancer immunology research | 2016

Abstract B020: STING activation in the tumor microenvironment using a synthetic human STING-activating cyclic dinucleotide induces potent antitumor immunity

Sarah M. McWhirter; Laura Hix Glickman; Tony Desbien; Kelsey Sivick Gauthier; David B. Kanne; Shailaja Kasibhatla; Jie Li; AnneMarie Culazzo Pferdekamper; George E. Katibah; Ed Lemmens; Leticia Corrales; Meredith Leong; Chudi Ndubaku; Justin J. Leong; Leonard Sung; Lianxing Zheng; Charles Y. Cho; Yan Feng; Jeffery M. McKenna; John A. Tallarico; Steven L. Bender; Thomas W. Dubensky

Stimulator of Interferon Genes (STING) is a critical component of the cytosolic DNA sensing pathway of the innate immune system. STING is activated by cyclic dinucleotides (CDNs), a product of the intracellular enzyme, cyclic GMP-AMP synthase (cGAS), in response to presence of cytosolic DNA, including tumor-derived DNA. Production of type I interferon within the tumor microenvironment (TME), mediated by the STING pathway, leads to the priming and activation of systemic tumor antigen-specific CD8 + T-cell immunity and tumor regression. Therapeutic activation of STING through intratumoral (IT) administration of CDNs results in anti-tumor efficacy and long-lived survival in several mouse syngeneic tumor models. Rational design of synthetic CDN derivatives has shown that certain modifications alter STING binding, increase cellular potency, enhance maturation of human dendritic cells to promote in vitro T cell expansion, and are able to broadly activate all human STING haplotypes. Mechanistic studies in mouse tumor models demonstrate that CDNs mediate anti-tumor immunity by inducing an acute innate immune response, leading to collapse of the injected tumor, and promoting a tumor-specific CD8 + T cell response that protects against tumor re-challenge. Anti-tumor efficacy is enhanced by combination with immune checkpoint inhibitors, informing future clinical development. The ability to elicit innate and adaptive anti-tumor immunity via activation of STING in the TME demonstrates that CDNs have high translational potential for the treatment of patients with advanced/metastatic solid tumors. The design of an ongoing Phase 1 first-in-human clinical study to evaluate the safety, tolerability and possible antitumor activity of ADU-S100 in subjects with cutaneously accessible tumors and lymphomas will also be presented. Citation Format: Sarah M. McWhirter, Laura Hix Glickman, Tony Desbien, Kelsey Sivick Gauthier, David Kanne, Shailaja Kasibhatla, Jie Li, AnneMarie Culazzo Pferdekamper, George Katibah, Ed Lemmens, Leticia Corrales, Meredith Leong, Chudi Ndubaku, Justin Leong, Leonard Sung, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John A. Tallarico, Steven L. Bender, Thomas W. Dubensky, Jr.. STING activation in the tumor microenvironment using a synthetic human STING-activating cyclic dinucleotide induces potent antitumor immunity [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr B020.


Cancer Research | 2016

Abstract IA35: Discovery of porcupine inhibitors targeting Wnt signaling in cancer

Shifeng Pan; Jun Liu; Dai Cheng; Dong Han; Guobao Zhang; Mindy H. Hsieh; Nicholas Ng; Chun Li; Shailaja Kasibhatla; Peter McNamara; H. Martin Seidel; Jennifer L. Harris

Wnt signaling is tightly controlled during cellular proliferation, differentiation and embryonic morphogenesis. Aberrant activation of this pathway plays a critical role in a variety of cancers. Blockade of Wnt signaling is therefore an attractive therapeutic approach for anticancer therapy. In this presentation, we will discuss our approach to search for inhibitors of Wnt ligand secretion. We developed and performed a cellular high-throughput screen using a co-culture system. Lead structure (GNF-1331) was identified and further target elucidation revealed Porcupine, a membrane bound O-acyl transferase, as its molecular target. Further structure-activity relationship studies led to the discovery of WNT974, a potent and specific Porcupine inhibitor. Treatment of WNT974 leads to tumor regression in a Wnt dependent MMTV-Wnt1 mouse model at well tolerated doses. WNT974 is currently in Phase 1 clinical trials. Citation Format: Shifeng Pan, Jun Liu, Dai Cheng, Dong Han, Guobao Zhang, Mindy Hsieh, Nicholas Ng, Chun Li, Shailaja Kasibhatla, Peter McNamara, H. Martin Seidel, Jennifer Harris. Discovery of porcupine inhibitors targeting Wnt signaling in cancer. [abstract]. In: Proceedings of the Fourth AACR International Conference on Frontiers in Basic Cancer Research; 2015 Oct 23-26; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2016;76(3 Suppl):Abstract nr IA35.

Collaboration


Dive into the Shailaja Kasibhatla's collaboration.

Top Co-Authors

Avatar

Jie Li

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Chun Li

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Harris

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Steven L. Bender

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

AnneMarie Culazzo Pferdekamper

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Celin Tompkins

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Jun Liu

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Peter McNamara

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Shifeng Pan

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Auzon Steffy

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Researchain Logo
Decentralizing Knowledge