Lars D. Engstrom
Pfizer
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Lars D. Engstrom.
Journal of Medicinal Chemistry | 2014
Ted W. Johnson; Paul F. Richardson; Simon Bailey; Alexei Brooun; Benjamin J. Burke; Michael Raymond Collins; J. Jean Cui; Judith Gail Deal; Ya-Li Deng; Dac M. Dinh; Lars D. Engstrom; Mingying He; Jacqui Elizabeth Hoffman; Robert Louis Hoffman; Qinhua Huang; Robert Steven Kania; John Charles Kath; Hieu Lam; Justine L. Lam; Phuong Thi Quy Le; Laura Lingardo; Wei Liu; Michele McTigue; Cynthia Louise Palmer; Neal W. Sach; Tod Smeal; Graham L. Smith; Albert E. Stewart; Sergei Timofeevski; Huichun Zhu
Although crizotinib demonstrates robust efficacy in anaplastic lymphoma kinase (ALK)-positive non-small-cell lung carcinoma patients, progression during treatment eventually develops. Resistant patient samples revealed a variety of point mutations in the kinase domain of ALK, including the L1196M gatekeeper mutation. In addition, some patients progress due to cancer metastasis in the brain. Using structure-based drug design, lipophilic efficiency, and physical-property-based optimization, highly potent macrocyclic ALK inhibitors were prepared with good absorption, distribution, metabolism, and excretion (ADME), low propensity for p-glycoprotein 1-mediated efflux, and good passive permeability. These structurally unusual macrocyclic inhibitors were potent against wild-type ALK and clinically reported ALK kinase domain mutations. Significant synthetic challenges were overcome, utilizing novel transformations to enable the use of these macrocycles in drug discovery paradigms. This work led to the discovery of 8k (PF-06463922), combining broad-spectrum potency, central nervous system ADME, and a high degree of kinase selectivity.
Cancer Cell | 2015
Helen Y. Zou; Luc Friboulet; David P. Kodack; Lars D. Engstrom; Qiuhua Li; Melissa West; Ruth W. Tang; Hui Wang; Konstantinos Tsaparikos; Jinwei Wang; Sergei Timofeevski; Ryohei Katayama; Dac M. Dinh; Hieu Lam; Justine L. Lam; Shinji Yamazaki; Wenyue Hu; Bhushankumar Patel; Divya Bezwada; Rosa L. Frias; Eugene Lifshits; Sidra Mahmood; Justin F. Gainor; Timothy Affolter; Patrick B. Lappin; Hovhannes J. Gukasyan; Nathan V. Lee; Shibing Deng; Rakesh K. Jain; Ted W. Johnson
We report the preclinical evaluation of PF-06463922, a potent and brain-penetrant ALK/ROS1 inhibitor. Compared with other clinically available ALK inhibitors, PF-06463922 displayed superior potency against all known clinically acquired ALK mutations, including the highly resistant G1202R mutant. Furthermore, PF-06463922 treatment led to regression of EML4-ALK-driven brain metastases, leading to prolonged mouse survival, in a superior manner. Finally, PF-06463922 demonstrated high selectivity and safety margins in a variety of preclinical studies. These results suggest that PF-06463922 will be highly effective for the treatment of patients with ALK-driven lung cancers, including those who relapsed on clinically available ALK inhibitors because of secondary ALK kinase domain mutations and/or brain metastases.
Proceedings of the National Academy of Sciences of the United States of America | 2015
Helen Y. Zou; Qiuhua Li; Lars D. Engstrom; Melissa West; Vicky Appleman; Katy A. Wong; Michele McTigue; Ya-Li Deng; Wei Liu; Alexei Brooun; Sergei Timofeevski; Scott R. McDonnell; Ping Jiang; Matthew D. Falk; Patrick B. Lappin; Timothy Affolter; Tim Nichols; Wenyue Hu; Justine L. Lam; Ted W. Johnson; Tod Smeal; Al Charest; Valeria R. Fantin
Significance Overcoming resistance to targeted kinase inhibitors is a major clinical challenge in oncology. Development of crizotinib resistance through the emergence of a secondary ROS1 mutation, ROS1G2032R, was observed in patients with ROS1 fusion-positive lung cancer. In addition, a novel ROS1 fusion recently has been identified in glioblastoma. A new agent with robust activity against the ROS1G2032R mutation and with CNS activity is needed to address these unmet medical needs. Here we report the identification of PF-06463922, a ROS1/anaplastic lymphoma kinase (ALK) inhibitor, with exquisite potency against ROS1 fusion kinases, capable of inhibiting the ROS1G2032R mutation and FIG-ROS1–driven glioblastoma tumor growth in preclinical models. PF-06463922 demonstrated excellent therapeutic potential against ROS1 fusion-driven cancers, and it currently is undergoing phase I/II clinical trial investigation. Oncogenic c-ros oncogene1 (ROS1) fusion kinases have been identified in a variety of human cancers and are attractive targets for cancer therapy. The MET/ALK/ROS1 inhibitor crizotinib (Xalkori, PF-02341066) has demonstrated promising clinical activity in ROS1 fusion-positive non-small cell lung cancer. However, emerging clinical evidence has shown that patients can develop resistance by acquiring secondary point mutations in ROS1 kinase. In this study we characterized the ROS1 activity of PF-06463922, a novel, orally available, CNS-penetrant, ATP-competitive small-molecule inhibitor of ALK/ROS1. In vitro, PF-06463922 exhibited subnanomolar cellular potency against oncogenic ROS1 fusions and inhibited the crizotinib-refractory ROS1G2032R mutation and the ROS1G2026M gatekeeper mutation. Compared with crizotinib and the second-generation ALK/ROS1 inhibitors ceritinib and alectinib, PF-06463922 showed significantly improved inhibitory activity against ROS1 kinase. A crystal structure of the PF-06463922-ROS1 kinase complex revealed favorable interactions contributing to the high-affinity binding. In vivo, PF-06463922 showed marked antitumor activity in tumor models expressing FIG-ROS1, CD74-ROS1, and the CD74-ROS1G2032R mutation. Furthermore, PF-06463922 demonstrated antitumor activity in a genetically engineered mouse model of FIG-ROS1 glioblastoma. Taken together, our results indicate that PF-06463922 has potential for treating ROS1 fusion-positive cancers, including those requiring agents with CNS-penetrating properties, as well as for overcoming crizotinib resistance driven by ROS1 mutation.
Journal of Medicinal Chemistry | 2014
Qinhua Huang; Ted W. Johnson; Simon Bailey; Alexei Brooun; Kevin D. Bunker; Benjamin J. Burke; Michael Raymond Collins; Andrew Simon Cook; J. Jean Cui; Kevin Neil Dack; Judith Gail Deal; Ya-Li Deng; Dac M. Dinh; Lars D. Engstrom; Mingying He; Jacqui Elizabeth Hoffman; Robert Louis Hoffman; Patrick Stephen Johnson; Robert Steven Kania; Hieu Lam; Justine L. Lam; Phuong Thi Quy Le; Qiuhua Li; Laura Lingardo; Wei Liu; Melissa West Lu; Michele McTigue; Cynthia Louise Palmer; Paul F. Richardson; Neal W. Sach
Crizotinib (1), an anaplastic lymphoma kinase (ALK) receptor tyrosine kinase inhibitor approved by the U.S. Food and Drug Administration in 2011, is efficacious in ALK and ROS positive patients. Under pressure of crizotinib treatment, point mutations arise in the kinase domain of ALK, resulting in resistance and progressive disease. The successful application of both structure-based and lipophilic-efficiency-focused drug design resulted in aminopyridine 8e, which was potent across a broad panel of engineered ALK mutant cell lines and showed suitable preclinical pharmacokinetics and robust tumor growth inhibition in a crizotinib-resistant cell line (H3122-L1196M).
Clinical Cancer Research | 2017
Lars D. Engstrom; Ruth Aranda; Matthew Randolf Lee; Elizabeth A. Tovar; Curt Essenburg; Zachary Madaj; Harrah Chiang; David Briere; Jill Hallin; Pedro P. Lopez-Casas; Natalia Baños; Camino Menéndez; Manuel Hidalgo; Vanessa Tassell; Richard C. Chao; Darya Chudova; Richard B. Lanman; Peter Olson; L. Bazhenova; Sandip Pravin Patel; Carrie R. Graveel; Mizuki Nishino; Geoffrey I. Shapiro; Nir Peled; Mark M. Awad; Pasi A. Jänne; James G. Christensen
Purpose: MET exon 14 deletion (METex14 del) mutations represent a novel class of non–small cell lung cancer (NSCLC) driver mutations. We evaluated glesatinib, a spectrum-selective MET inhibitor exhibiting a type II binding mode, in METex14 del–positive nonclinical models and NSCLC patients and assessed its ability to overcome resistance to type I MET inhibitors. Experimental Design: As most MET inhibitors in clinical development bind the active site with a type I binding mode, we investigated mechanisms of acquired resistance to each MET inhibitor class utilizing in vitro and in vivo models and in glesatinib clinical trials. Results: Glesatinib inhibited MET signaling, demonstrated marked regression of METex14 del-driven patient-derived xenografts, and demonstrated a durable RECIST partial response in a METex14 del mutation-positive patient enrolled on a glesatinib clinical trial. Prolonged treatment of nonclinical models with selected MET inhibitors resulted in differences in resistance kinetics and mutations within the MET activation loop (i.e., D1228N, Y1230C/H) that conferred resistance to type I MET inhibitors, but remained sensitive to glesatinib. In vivo models exhibiting METex14 del/A-loop double mutations and resistance to type I inhibitors exhibited a marked response to glesatinib. Finally, a METex14 del mutation-positive NSCLC patient who responded to crizotinib but later relapsed, demonstrated a mixed response to glesatinib including reduction in size of a MET Y1230H mutation-positive liver metastasis and concurrent loss of detection of this mutation in plasma DNA. Conclusions: Together, these data demonstrate that glesatinib exhibits a distinct mechanism of target inhibition and can overcome resistance to type I MET inhibitors. Clin Cancer Res; 23(21); 6661–72. ©2017 AACR.
Cancer Research | 2011
Amy Jackson-Fisher; Melissa McMahon; Justine L. Lam; Chunze Li; Lars D. Engstrom; Konstantinos Tsaparikos; David J. Shields; Douglas D. Fang; Maruja E. Lira; Zhou Zhu; Michael D. Robbins; Richard Schwab; Michael John Munchhof; Todd VanArsdale
Aberrant activation of the Hedgehog (Hh) signaling pathway has been implicated in several human cancers. Mutations in the Patched (PTCH1) gene are responsible for basal cell nevus syndrome, and are commonly found in sporadic basal cell carcinoma and in medulloblastoma. In this study we evaluated PF-04449913, an inhibitor of the Hh signaling pathway, in a Ptch1+/-p53 mouse model of medulloblastoma and in human patient derived xenograft models. Treatment of Ptch1+/-p53+/- or Ptch1+/-p53-/- medulloblastoma allografts with PF-04449913 produced potent dose-dependent inhibition of Hh pathway activity resulting in stable tumor regression. Using Gli1 transcript levels as a surrogate for Hh pathway activity, the pharmacodynamic effects of PF-04449913 were evaluated in medulloblastoma allografts following single dose and multi dose administrations of compound. PF-04449913 treated medulloblastoma allografts had reduced levels of Gli1 gene expression and down regulation of genes linked to the Hh signaling pathway. PF-04449913 was also effective when combined with a chemotherapeutic agent in a colon patient derived xenograft model and a pancreatic patient derived xenograft model, resulting in 63% and 73% tumor growth inhibition respectively. Collectively, our study demonstrates the therapeutic efficacy of a small molecule inhibitor of Hh pathway in preclinical models of multiple cancer types in either single or combination treatments. 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 4504. doi:10.1158/1538-7445.AM2011-4504
Molecular Cancer Therapeutics | 2013
Ted W. Johnson; Simon Bailey; Benjamin J. Burke; Michael Raymond Collins; J. Jean Cui; Judy G. Deal; Ya-Li Deng; Martin Paul Edwards; Mingying He; Jacqui Elizabeth Hoffman; Robert L. Hoffman; Qinhua Huang; Robert Steven Kania; Phuong T. Le; Michele McTigue; Cynthia Louise Palmer; Paul F. Richardson; Neal W. Sach; Graham L. Smith; Lars D. Engstrom; Wenyue Hu; Hieu Lam; Justine L. Lam; Tod Smeal; Helen Y. Zou
Oncogenic fusions of anaplastic lymphoma kinase (ALK) define a subset of human lung adenocarcinoma. The 1st generation ALK inhibitor crizotinib demonstrated impressive clinical benefit in ALK-fusion positive lung cancers and was approved by the FDA for the treatment of ALK-fusion positive NSCLC in 2011. However, as seen with most kinase inhibitors, patients treated with crizotinib eventually develop resistance to therapy. Acquired ALK kinase domain mutations and disease progression in the central nervous system (CNS) are reported as main contributors to patient relapse after ALK inhibitor therapy. Preclinically, crizotinib lacks significant brain penetration and does not potently inhibit activity of ALK kinase domain mutants, so a drug discovery program was initiated aimed to develop a second generation ALK inhibitor that is more potent than existing ALK inhibitors, capable of inhibiting the resistant ALK mutants and penetrating the blood-brain-barrier. These objectives present a considerable challenge in kinase inhibitor chemical space. Here we report that PF-06463922, a novel small molecule ATP-competitive inhibitor of ALK/ROS1, showed exquisite potencies against non-mutant ALK (Ki 100 fold kinase selectivity against 95% of the kinases tested in a 207 recombinant kinase panel. Specific design considerations were developed leading to novel ATP-competitive kinase inhibitors with desired low efflux in cell lines over-expressing p-glycoprotein and breast cancer resistance protein, providing excellent blood-brain-barrier and cell penetration properties. Efforts to optimize ligand efficiency and lipophilic efficiency leveraging structure based drug design techniques led to ligands with overlapping broad spectrum potency and low efflux. Single and repeat dose preclinical rat in vivo studies of PF-06463922 demonstrated excellent oral bioavailability and CNS availability with free brain exposure approximately 30% of free plasma levels. In addition, CNS-directed safety studies showed no adverse events at predicted efficacious concentrations. It is anticipated that PF-06463922 with its potent activities on non-mutant ALK, ALK kinase domain mutations and CNS metastases would provide great promise for patients with ALK and ROS1 positive cancers. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):PR10. Citation Format: Ted W. Johnson, Simon Bailey, Benjamin J. Burke, Michael R. Collins, J. Jean Cui, Judy Deal, Ya-Li Deng, Martin P. Edwards, Mingying He, Jacqui Hoffman, Robert L. Hoffman, Qinhua Huang, Robert S. Kania, Phuong Le, Michele McTigue, Cynthia L. Palmer, Paul F. Richardson, Neal W. Sach, Graham L. Smith, Lars Engstrom, Wenyue Hu, Hieu Lam, Justine L. Lam, Tod Smeal, Helen Y. Zou. Is CNS availability for oncology a no-brainer? Discovery of PF-06463922, a novel small molecule inhibitor of ALK/ROS1 with preclinical brain availability and broad spectrum potency against ALK-resistant mutations. [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 PR10.
Cancer Research | 2013
Nathan V. Lee; Joan Cao; Timothy Scheffelin; Lars D. Engstrom; Stephen Huang; Maruja E. Lira; Scott J. Garza; Jing Yuan; Blake Enyart; James G. Christensen; Julie Kan
Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The Anaplastic Lymphoma Kinase (ALK) fusion proteins such as EML4-ALK act as oncogenic drivers in approximately 5-7% of non-small cell lung carcinomas (NSCLC). The rationale for targeting this kinase as a means of treatment has been validated by the clinical efficacy and subsequent approval of the ALK inhibitor, crizotinib (Xalkori ®). The overall response rate of 61% suggests potential intrinsic resistance to crizotinib. In addition, like many tyrosine kinase inhibitors, acquired resistance inevitably leads to treatment refractory cancers. To better understand resistance mechanisms (both intrinsic and acquired), and circumvent resistance to crizotinib, we developed in vitro resistance models through prolonged crizotinib exposure. We isolated resistance clones and characterization identified different resistance mechanisms, including pathway bypass as well as an ALK kinase mutation (G1269A), similar to those that have been reported from refractory patient samples. The majority of the resistance clones escaped crizotinib suppression via pathway bypass. In several resistance clones, simultaneous activation of EGFR and IGF1R receptors limited the efficacy of double combination strategies and required triple combinations. In addition, phospho-proteomic analysis revealed simultaneous activation of different receptor tyrosine kinases, suggesting that combination treatment with inhibitors of downstream mediators such as PI3K and MEK may be effective strategies. Our resistance models demonstrate the heterogeneity of cancers and reveal the multiplicity of escape mechanisms. Our results provide support for the evaluation of combination therapy with downstream mediator inhibitors as a way to restore as well as enhance sensitivity to crizotinib. Citation Format: Nathan V. Lee, Joan Cao, Timothy Scheffelin, Lars Engstrom, Stephen Huang, Maruja Lira, Scott Garza, Jing Yuan, Blake Enyart, James Christensen, Julie Kan. Elucidation of crizotinib resistance in NCI-H3122 and strategies to circumvent bypass resistance. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4464. doi:10.1158/1538-7445.AM2013-4464
Cancer Research | 2011
Helen Y. Zou; Qiuhua Li; Joseph Lee; Lars D. Engstrom; Melissa West Lu; Alex Young; Sergei Timofeevski; Dac M. Dinh; Shinji Yamazaki; Justine L. Lam; Zheng Feng; Jeffrey Nickel; Patrick B. Lappin; Tim Nichols; Anthony Wong; Brittany Snider; Hovhannes J. Gukasyan; Steve Bender; Sonya D. Zabludoff; Tod Smeal; James R. Christensen
EML4-ALK fusion was recently characterized as an “Addicted Oncogene” in a subset of human lung adenocarcinoma, and it plays an essential role in regulation of the tumor cell survival, growth and metastasis. Crizotinib is a potent and selective ATP competitive small molecule inhibitor of ALK and c-Met. It is currently in clinical trials for advanced non-small cell lung cancers positive for ALK fusion. This report summarizes the pre-clinical pharmacology studies for crizotinib in the EML4-Alk positive tumor models to assess the pharmacodynamic inhibition of EML4-ALK, antitumor efficacy, PKPD relationships and antitumor mechanism of action in vitro and in vivo. Crizotinib potently inhibited the catalytic activity of ALK kinase (Ki = 0.5 nM) and the autophosphorylation of cellular EML4-ALK V1, V2, V3a and V3b with IC 50 values ranging from 26–74 nM. Crizotinib also inhibited cell proliferation and induced apoptosis in NCI-H3122 human NSCLC cells harboring EML4-ALK V1 fusion with IC 50s of 63 nM and 110 nM respectively. In the EML4-ALK V3a/b positive NCI-H2228 human NSCLC cells, crizotinib completely inhibited ALK phosphorylation (IC 50 = 74 nM) but only partial inhibited cell proliferation and survival. This observation is consistent with the notion that only a portion of H2228 cells are EML4-ALK positive by Exon Array analysis. Crizotinib demonstrated marked tumor growth inhibition and regression in H3122 xenograft model at well tolerated dose levels. The antitumor efficacy of crizotinib was dose dependent and demonstrated a strong correlation to pharmacodynamic inhibition of ALK phosphorylation in vivo. PKPD modeling was conducted to understand the relationships between crizotinib plasma concentration to ALK target inhibition (EC 50 = 19 nM) and antitumor efficacy (EC 50 = 23 nM). Collectively, the results from H3122 model indicated that significant inhibition of EML4-ALK during the entire treatment period was necessary to achieve robust antitumor efficacy. Additional in vivo studies with crizotinib demonstrated dose dependent inhibition of EML4-ALK mediated signal transduction (STAT3, AKT, Erk, PLCγ, c-Myc), tumor cell proliferation (Ki67) and induction of apoptosis (caspase-3). Crizotinib also dose dependently inhibited total EML4-ALK levels in H3122 tumors indicating an additional antitumor mechanism of action by crizotinib in these tumors. Furthermore, a dose dependent increase in phospho-EGFR levels was observed after 4 days of drug treatment in H3122 model, indicating a compensation mechanism of “oncogene switching” in tumor cell signaling, and a potential resistance mechanism that may compromise “patient” responses to crizotinib treatment in these tumors. In conclusion, crizotinib was shown to be a potent inhibitor of EML4-ALK. It demonstrated marked antitumor efficacy in EML4-ALK dependent human NSCLC tumors in vitro and in vivo. 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 LB-390. doi:10.1158/1538-7445.AM2011-LB-390
Cancer Research | 2016
Lars D. Engstrom; Ruth W. Tang; David Briere; Harrah Chiang; Peter Olson; James G. Christensen
MET splice site mutations that result in the deletion of exon 14 (METex14del) are implicated as oncogenic drivers in a subset of non-small cell lung cancer (NSCLC). MET exon 14 contains the Y1003 CBL ubiquitin ligase regulatory binding site that normally mediates CBL-dependent MET degradation and signal attenuation. Deletion of this exon results in sustained activation of MET and its downstream signaling pathways. The diverse splice site mutations leading to exon 14 skipping comprise a unique and unprecedented class of RTK activating mutations and the molecular mechanism by which these genetic alterations transform cancer cells is not fully understood. One major challenge in understanding the utility of MET inhibition of the METex14del class has been the lack of available pre-clinical models. In the present study, we generated and characterized multiple METex14del-driven cancer models to study the mechanism of MET-dependent cellular transformation as well as the response to MGCD265, a small molecule inhibitor of MET and AXL. METex14del models were identified via mining patient-derived xenograft (PDX) databases or were engineered using genome editing techniques to generate isogenic pairs of METex14del and WT cell lines. The METex14del cell lines formed increased size and number of colonies in anchorage independent growth assays compared to their WT counterparts. The transformation of METex14del cells was associated with an increase in durable HGF-dependent activation of MET and downstream signaling pathways potentially due to dysregulated MET processing and signaling attenuation. MGCD265 was shown to effectively inhibit this growth and MET-dependent signal transduction in a concentration-dependent manner. When evaluated in the amplified METex14del-driven gastric xenograft model Hs746T, significant tumor regression was observed following MGCD265 treatment. In addition, MGCD265 demonstrated substantial regression of large established tumors, in two novel NSCLC METex14del-positive PDX models. Together, these data confirm METex14del mutations are bona fide oncogenic drivers and sensitive to targeted therapeutics. Moreover, the models described in this study represent a relevant pre-clinical platform to further study receptor hyper-activation and drug action that is clinically actionable. Identification, development, and understanding of METex14del models will likely help further guide precision medicine strategies to treat NSCLC patients harboring these mutations. Citation Format: Lars D. Engstrom, Ruth W. Tang, David M. Briere, Harrah Chiang, Peter Olson, James G. Christensen. Evaluation of the mechanism of MET-dependent cellular transformation and potent cytoreductive activity of MGCD265 in novel MET exon 14 mutation positive cancer models. [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 2642.