Network


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

Hotspot


Dive into the research topics where Elizabeth L. Lockerman is active.

Publication


Featured researches published by Elizabeth L. Lockerman.


Cancer Discovery | 2014

The ALK Inhibitor Ceritinib Overcomes Crizotinib Resistance in Non–Small Cell Lung Cancer

Luc Friboulet; Nanxin Li; Ryohei Katayama; Christian C. Lee; Justin F. Gainor; Adam S. Crystal; Pierre-Yves Michellys; Mark M. Awad; Noriko Yanagitani; Sungjoon Kim; AnneMarie C. Pferdekamper; Jie Li; Shailaja Kasibhatla; Frank Sun; Xiuying Sun; Su Hua; Peter McNamara; Sidra Mahmood; Elizabeth L. Lockerman; Naoya Fujita; Makoto Nishio; Jennifer L. Harris; Alice T. Shaw; Jeffrey A. Engelman

UNLABELLED Non-small cell lung cancers (NSCLC) harboring anaplastic lymphoma kinase (ALK) gene rearrangements invariably develop resistance to the ALK tyrosine kinase inhibitor (TKI) crizotinib. Herein, we report the first preclinical evaluation of the next-generation ALK TKI, ceritinib (LDK378), in the setting of crizotinib resistance. An interrogation of in vitro and in vivo models of acquired resistance to crizotinib, including cell lines established from biopsies of patients with crizotinib-resistant NSCLC, revealed that ceritinib potently overcomes crizotinib-resistant mutations. In particular, ceritinib effectively inhibits ALK harboring L1196M, G1269A, I1171T, and S1206Y mutations, and a cocrystal structure of ceritinib bound to ALK provides structural bases for this increased potency. However, we observed that ceritinib did not overcome two crizotinib-resistant ALK mutations, G1202R and F1174C, and one of these mutations was identified in 5 of 11 biopsies from patients with acquired resistance to ceritinib. Altogether, our results demonstrate that ceritinib can overcome crizotinib resistance, consistent with clinical data showing marked efficacy of ceritinib in patients with crizotinib-resistant disease. SIGNIFICANCE The second-generation ALK inhibitor ceritinib can overcome several crizotinib-resistant mutations and is potent against several in vitro and in vivo laboratory models of acquired resistance to crizotinib. These findings provide the molecular basis for the marked clinical activity of ceritinib in patients with ALK-positive NSCLC with crizotinib-resistant disease. Cancer Discov; 4(6); 662-73. ©2014 AACR. See related commentary by Ramalingam and Khuri, p. 634 This article is highlighted in the In This Issue feature, p. 621.


Science | 2014

Patient-derived models of acquired resistance can identify effective drug combinations for cancer

Adam S. Crystal; Alice T. Shaw; Lecia V. Sequist; Luc Friboulet; Matthew J. Niederst; Elizabeth L. Lockerman; Rosa L. Frias; Justin F. Gainor; Arnaud Amzallag; Patricia Greninger; Dana Lee; Anuj Kalsy; Maria Gomez-Caraballo; Leila Elamine; Emily Howe; Wooyoung Hur; Eugene Lifshits; Hayley Robinson; Ryohei Katayama; Anthony C. Faber; Mark M. Awad; Sridhar Ramaswamy; Mari Mino-Kenudson; A. John Iafrate; Cyril H. Benes; Jeffrey A. Engelman

Targeted cancer therapies have produced substantial clinical responses, but most tumors develop resistance to these drugs. Here, we describe a pharmacogenomic platform that facilitates rapid discovery of drug combinations that can overcome resistance. We established cell culture models derived from biopsy samples of lung cancer patients whose disease had progressed while on treatment with epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors and then subjected these cells to genetic analyses and a pharmacological screen. Multiple effective drug combinations were identified. For example, the combination of ALK and MAPK kinase (MEK) inhibitors was active in an ALK-positive resistant tumor that had developed a MAP2K1 activating mutation, and the combination of EGFR and fibroblast growth factor receptor (FGFR) inhibitors was active in an EGFR mutant resistant cancer with a mutation in FGFR3. Combined ALK and SRC (pp60c-src) inhibition was effective in several ALK-driven patient-derived models, a result not predicted by genetic analysis alone. With further refinements, this strategy could help direct therapeutic choices for individual patients. Secondary chemotherapies can be developed by screening drug-resistant cells from individual cancer patients. Drug resistance, up close and personal Cancer therapies that target specific genetic mutations driving tumor growth have shown promising results in patients; however, the response is often short-lived because the tumors acquire new mutations that render them resistant to these therapies. Complicating matters, the mechanism of resistance can vary from patient to patient. To identify drugs most likely to be effective against resistant tumors, Crystal et al. established cell lines from the tumors of individual patients after resistance occurred and performed a drug screen and genetic analysis on the cultured cells. This strategy successfully identified drug combinations that halted the growth of resistant tumor cells both in culture and in mice. In the future, pharmacological profiling of patient-derived cells could be an efficient way to direct therapeutic choices for individual cancer patients. Science, this issue p. 1480


The New England Journal of Medicine | 2013

Acquired Resistance to Crizotinib from a Mutation in CD74–ROS1

Mark M. Awad; Ryohei Katayama; Michele McTigue; Wei Liu; Ya-Li Deng; Alexei Brooun; Luc Friboulet; Donghui Huang; Matthew D. Falk; Sergei Timofeevski; Keith D. Wilner; Elizabeth L. Lockerman; Tahsin M. Khan; Sidra Mahmood; Justin F. Gainor; Subba R. Digumarthy; James R. Stone; Mari Mino-Kenudson; James G. Christensen; A. John Iafrate; Jeffrey A. Engelman; Alice T. Shaw

Crizotinib, an inhibitor of anaplastic lymphoma kinase (ALK), has also recently shown efficacy in the treatment of lung cancers with ROS1 translocations. Resistance to crizotinib developed in a patient with metastatic lung adenocarcinoma harboring a CD74-ROS1 rearrangement who had initially shown a dramatic response to treatment. We performed a biopsy of a resistant tumor and identified an acquired mutation leading to a glycine-to-arginine substitution at codon 2032 in the ROS1 kinase domain. Although this mutation does not lie at the gatekeeper residue, it confers resistance to ROS1 kinase inhibition through steric interference with drug binding. The same resistance mutation was observed at all the metastatic sites that were examined at autopsy, suggesting that this mutation was an early event in the clonal evolution of resistance. (Funded by Pfizer and others; ClinicalTrials.gov number, NCT00585195.).


Nature Medicine | 2016

Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition

Aaron N. Hata; Matthew J. Niederst; Hannah L. Archibald; Maria Gomez-Caraballo; Faria Siddiqui; Hillary Mulvey; Yosef E. Maruvka; Fei Ji; Hyo Eun C Bhang; Viveksagar Krishnamurthy Radhakrishna; Giulia Siravegna; Haichuan Hu; Sana Raoof; Elizabeth L. Lockerman; Anuj Kalsy; Dana Lee; Celina L. Keating; David A. Ruddy; Leah Damon; Adam S. Crystal; Carlotta Costa; Zofia Piotrowska; Alberto Bardelli; Anthony John Iafrate; Ruslan I. Sadreyev; Frank Stegmeier; Gad Getz; Lecia V. Sequist; Anthony C. Faber; Jeffrey A. Engelman

Although mechanisms of acquired resistance of epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancers to EGFR inhibitors have been identified, little is known about how resistant clones evolve during drug therapy. Here we observe that acquired resistance caused by the EGFRT790M gatekeeper mutation can occur either by selection of pre-existing EGFRT790M-positive clones or via genetic evolution of initially EGFRT790M-negative drug-tolerant cells. The path to resistance impacts the biology of the resistant clone, as those that evolved from drug-tolerant cells had a diminished apoptotic response to third-generation EGFR inhibitors that target EGFRT790M; treatment with navitoclax, an inhibitor of the anti-apoptotic factors BCL-xL and BCL-2 restored sensitivity. We corroborated these findings using cultures derived directly from EGFR inhibitor–resistant patient tumors. These findings provide evidence that clinically relevant drug-resistant cancer cells can both pre-exist and evolve from drug-tolerant cells, and they point to therapeutic opportunities to prevent or overcome resistance in the clinic.


Cancer Discovery | 2015

Heterogeneity Underlies the Emergence of EGFRT790 Wild-Type Clones Following Treatment of T790M-Positive Cancers with a Third-Generation EGFR Inhibitor

Zofia Piotrowska; Matthew J. Niederst; Chris Karlovich; Heather A. Wakelee; Joel W. Neal; Mari Mino-Kenudson; L. Fulton; Aaron N. Hata; Elizabeth L. Lockerman; Anuj Kalsy; Subba R. Digumarthy; Alona Muzikansky; Mitch Raponi; Angel R. Garcia; Hillary Mulvey; Melissa Parks; Richard H. DiCecca; Dora Dias-Santagata; Anthony John Iafrate; Alice T. Shaw; Andrew R. Allen; J. A. Engelman; Lecia V. Sequist

UNLABELLED Rociletinib is a third-generation EGFR inhibitor active in lung cancers with T790M, the gatekeeper mutation underlying most first-generation EGFR drug resistance. We biopsied patients at rociletinib progression to explore resistance mechanisms. Among 12 patients with T790M-positive cancers at rociletinib initiation, six had T790-wild-type rociletinib-resistant biopsies. Two T790-wild-type cancers underwent small cell lung cancer transformation; three T790M-positive cancers acquired EGFR amplification. We documented T790-wild-type and T790M-positive clones coexisting within a single pre-rociletinib biopsy. The pretreatment fraction of T790M-positive cells affected response to rociletinib. Longitudinal circulating tumor DNA (ctDNA) analysis revealed an increase in plasma EGFR-activating mutation, and T790M heralded rociletinib resistance in some patients, whereas in others the activating mutation increased but T790M remained suppressed. Together, these findings demonstrate the role of tumor heterogeneity when therapies targeting a singular resistance mechanism are used. To further improve outcomes, combination regimens that also target T790-wild-type clones are required. SIGNIFICANCE This report documents that half of T790M-positive EGFR-mutant lung cancers treated with rociletinib are T790-wild-type upon progression, suggesting that T790-wild-type clones can emerge as the dominant source of resistance. We show that tumor heterogeneity has important clinical implications and that plasma ctDNA analyses can sometimes predict emerging resistance mechanisms.


Cancer Discovery | 2016

Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer

Justin F. Gainor; Leila Dardaei; Satoshi Yoda; Luc Friboulet; Ignaty Leshchiner; Ryohei Katayama; Ibiayi Dagogo-Jack; Shirish M. Gadgeel; Katherine Schultz; Manrose Singh; Emily Chin; Melissa Parks; Dana Lee; Richard H. DiCecca; Elizabeth L. Lockerman; Tiffany Huynh; Jennifer A. Logan; Lauren L. Ritterhouse; Long P. Le; Ashok Muniappan; Subba R. Digumarthy; Colleen L. Channick; Colleen Keyes; Gad Getz; Dora Dias-Santagata; Rebecca S. Heist; Jochen K. Lennerz; Lecia V. Sequist; Cyril H. Benes; A. John Iafrate

Advanced, anaplastic lymphoma kinase (ALK)-positive lung cancer is currently treated with the first-generation ALK inhibitor crizotinib followed by more potent, second-generation ALK inhibitors (e.g., ceritinib, alectinib) upon progression. Second-generation inhibitors are generally effective even in the absence of crizotinib-resistant ALK mutations, likely reflecting incomplete inhibition of ALK by crizotinib in many cases. Herein, we analyzed 103 repeat biopsies from ALK-positive patients progressing on various ALK inhibitors. We find that each ALK inhibitor is associated with a distinct spectrum of ALK resistance mutations and that the frequency of one mutation - ALK G1202R - increases significantly after treatment with second-generation agents. To investigate strategies to overcome resistance to second-generation ALK inhibitors, we examine the activity of the third-generation ALK inhibitor lorlatinib in a series of ceritinib-resistant, patient-derived cell lines, and observe that the presence of ALK resistance mutations is highly predictive for sensitivity to lorlatinib, whereas those cell lines without ALK mutations are resistant.


Nature Communications | 2015

RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer

Matthew J. Niederst; Lecia V. Sequist; John T. Poirier; Craig H. Mermel; Elizabeth L. Lockerman; Angel R. Garcia; Ryohei Katayama; Carlotta Costa; Kenneth N. Ross; Teresa Moran; Emily Howe; L. Fulton; Hillary Mulvey; Lindsay A. Bernardo; Farhiya Mohamoud; Norikatsu Miyoshi; Paul A. VanderLaan; Daniel B. Costa; Pasi A. Jänne; Darrell R. Borger; Sridhar Ramaswamy; Toshi Shioda; Anthony John Iafrate; Gad Getz; Charles M. Rudin; Mari Mino-Kenudson; Jeffrey A. Engelman

Tyrosine kinase inhibitors are effective treatments for non-small-cell lung cancers (NSCLCs) with epidermal growth factor receptor (EGFR) mutations. However, relapse typically occurs after an average of 1 year of continuous treatment. A fundamental histological transformation from NSCLC to small-cell lung cancer (SCLC) is observed in a subset of the resistant cancers, but the molecular changes associated with this transformation remain unknown. Analysis of tumour samples and cell lines derived from resistant EGFR mutant patients revealed that Retinoblastoma (RB) is lost in 100% of these SCLC transformed cases, but rarely in those that remain NSCLC. Further, increased neuroendocrine marker and decreased EGFR expression as well as greater sensitivity to BCL2 family inhibition are observed in resistant SCLC transformed cancers compared with resistant NSCLCs. Together, these findings suggest that this subset of resistant cancers ultimately adopt many of the molecular and phenotypic characteristics of classical SCLC.


Clinical Cancer Research | 2014

Two novel ALK mutations mediate acquired resistance to the next generation ALK inhibitor alectinib

Ryohei Katayama; Luc Friboulet; Sumie Koike; Elizabeth L. Lockerman; Tahsin M. Khan; Justin F. Gainor; Anthony John Iafrate; Kengo Takeuchi; Taiji M; Yasushi Okuno; Naoya Fujita; J. A. Engelman; Alice T. Shaw

Purpose: The first-generation ALK tyrosine kinase inhibitor (TKI) crizotinib is a standard therapy for patients with ALK-rearranged non–small cell lung cancer (NSCLC). Several next-generation ALK-TKIs have entered the clinic and have shown promising activity in crizotinib-resistant patients. As patients still relapse even on these next-generation ALK-TKIs, we examined mechanisms of resistance to the next-generation ALK-TKI alectinib and potential strategies to overcome this resistance. Experimental Design: We established a cell line model of alectinib resistance, and analyzed a resistant tumor specimen from a patient who had relapsed on alectinib. We developed Ba/F3 models harboring alectinib-resistant ALK mutations and evaluated the potency of other next-generation ALK-TKIs in these models. We tested the antitumor activity of the next-generation ALK-TKI ceritinib in the patient with acquired resistance to alectinib. To elucidate structure–activity relationships of ALK mutations, we performed computational thermodynamic simulation with MP-CAFEE. Results: We identified a novel V1180L gatekeeper mutation from the cell line model and a second novel I1171T mutation from the patient who developed resistance to alectinib. Both ALK mutations conferred resistance to alectinib as well as to crizotinib, but were sensitive to ceritinib and other next-generation ALK-TKIs. Treatment of the patient with ceritinib led to a marked response. Thermodynamics simulation suggests that both mutations lead to distinct structural alterations that decrease the binding affinity with alectinib. Conclusions: We have identified two novel ALK mutations arising after alectinib exposure that are sensitive to other next-generation ALK-TKIs. The ability of ceritinib to overcome alectinib-resistance mutations suggests a potential role for sequential therapy with multiple next-generation ALK-TKIs. Clin Cancer Res; 20(22); 5686–96. ©2014 AACR.


Clinical Cancer Research | 2015

The allelic context of the C797S mutation acquired upon treatment with third generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies

Matthew J. Niederst; Haichuan Hu; Hillary Mulvey; Elizabeth L. Lockerman; Angel R. Garcia; Zofia Piotrowska; Lecia V. Sequist; Jeffrey A. Engelman

Purpose: A secondary EGFR mutation, T790M, is the most common resistance mechanism in EGFR-mutant adenocarcinomas that have progressed on erlotinib. Third-generation EGFR inhibitors capable of inhibiting mutant EGFR with T790M produce responses in nearly two thirds of patients. However, acquired resistance mechanisms in patients treated with these drugs are yet to be described. Experimental Design: To study acquired resistance to third-generation EGFR inhibitors, T790M-positive cells derived from an erlotinib-resistant cancer were made resistant to a third-generation TKI and then characterized using cell and molecular analyses. Results: Cells resistant to a third-generation TKI acquired an additional EGFR mutation, C797S, which prevented suppression of EGFR. Our results demonstrate that the allelic context in which C797S was acquired may predict responsiveness to alternative treatments. If the C797S and T790M mutations are in trans, cells will be resistant to third-generation EGFR TKIs, but will be sensitive to a combination of first- and third-generation TKIs. If the mutations are in cis, no EGFR TKIs alone or in combination can suppress activity. If C797S develops in cells wild-type for T790 (when third-generation TKIs are administered in the first-line setting), the cells are resistant to third-generation TKIs, but retain sensitivity to first-generation TKIs. Conclusions: Mutation of C797S in EGFR is a novel mechanism of acquired resistance to third-generation TKIs. The context in which the C797S develops with respect to the other EGFR alleles affects the efficacy of subsequent treatments. Clin Cancer Res; 21(17); 3924–33. ©2015 AACR. See related commentary by Ayeni et al., p. 3818


Cancer Discovery | 2015

Clinical Acquired Resistance to RAF Inhibitor Combinations in BRAF-Mutant Colorectal Cancer through MAPK Pathway Alterations

Leanne G. Ahronian; Erin M. Sennott; Eliezer M. Van Allen; Nikhil Wagle; Eunice L. Kwak; Jason E. Faris; Jason T. Godfrey; Koki Nishimura; Kerry Lynch; Craig H. Mermel; Elizabeth L. Lockerman; Anuj Kalsy; Joseph M. Gurski; Samira Bahl; Kristin Anderka; Lisa Green; Niall J. Lennon; Tiffany Huynh; Mari Mino-Kenudson; Gad Getz; Dora Dias-Santagata; A. John Iafrate; Jeffrey A. Engelman; Levi A. Garraway; Ryan B. Corcoran

UNLABELLED BRAF mutations occur in approximately 10% of colorectal cancers. Although RAF inhibitor monotherapy is highly effective in BRAF-mutant melanoma, response rates in BRAF-mutant colorectal cancer are poor. Recent clinical trials of combined RAF/EGFR or RAF/MEK inhibition have produced improved efficacy, but patients ultimately develop resistance. To identify molecular alterations driving clinical acquired resistance, we performed whole-exome sequencing on paired pretreatment and postprogression tumor biopsies from patients with BRAF-mutant colorectal cancer treated with RAF inhibitor combinations. We identified alterations in MAPK pathway genes in resistant tumors not present in matched pretreatment tumors, including KRAS amplification, BRAF amplification, and a MEK1 mutation. These alterations conferred resistance to RAF/EGFR or RAF/MEK combinations through sustained MAPK pathway activity, but an ERK inhibitor could suppress MAPK activity and overcome resistance. Identification of MAPK pathway reactivating alterations upon clinical acquired resistance underscores the MAPK pathway as a critical target in BRAF-mutant colorectal cancer and suggests therapeutic options to overcome resistance. SIGNIFICANCE RAF inhibitor combinations represent promising approaches in clinical development for BRAF-mutant colorectal cancer. Initial characterization of clinical acquired resistance mechanisms to these regimens identified several MAPK pathway alterations driving resistance by reactivating MAPK signaling, highlighting the critical dependence of BRAF-mutant colorectal cancers on MAPK signaling and offering potential strategies to overcome resistance.

Collaboration


Dive into the Elizabeth L. Lockerman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryohei Katayama

Japanese Foundation for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge