20 results on '"Christie J. Lau"'
Search Results
2. Data from Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Pasi A. Jänne, Stephen Wang, Sujuan Guo, Jiannan Guo, Paul T. Kirschmeier, Jens Köhler, Zihan Wei, Fangxin Hong, Pratik R. Chopade, Prafulla C. Gokhale, Christie J. Lau, Kshiti H. Dholakia, Nam Doo Kim, Taebo Sim, Luke J. Taus, Yanan Kuang, Cloud P. Paweletz, and Magda Bahcall
- Abstract
MET-targeted therapies are clinically effective in MET-amplified and MET exon 14 deletion mutant (METex14) non–small cell lung cancers (NSCLCs), but their efficacy is limited by the development of drug resistance. Structurally distinct MET tyrosine kinase inhibitors (TKIs) (type I/II) have been developed or are under clinical evaluation, which may overcome MET-mediated drug resistance mechanisms. In this study, we assess secondary MET mutations likely to emerge in response to treatment with single-agent or combinations of type I/type II MET TKIs using TPR-MET transformed Ba/F3 cell mutagenesis assays. We found that these inhibitors gave rise to distinct secondary MET mutant profiles. However, a combination of type I/II TKI inhibitors (capmatinib and merestinib) yielded no resistant clones in vitro. The combination of capmatinib/merestinib was evaluated in vivo and led to a significant reduction in tumor outgrowth compared with either MET inhibitor alone. Our findings demonstrate in vitro and in vivo that a simultaneous treatment with a type I and type II MET TKI may be a clinically viable approach to delay and/or diminish the emergence of on target MET-mediated drug-resistance mutations.
- Published
- 2023
3. Supplementary Data from Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Pasi A. Jänne, Stephen Wang, Sujuan Guo, Jiannan Guo, Paul T. Kirschmeier, Jens Köhler, Zihan Wei, Fangxin Hong, Pratik R. Chopade, Prafulla C. Gokhale, Christie J. Lau, Kshiti H. Dholakia, Nam Doo Kim, Taebo Sim, Luke J. Taus, Yanan Kuang, Cloud P. Paweletz, and Magda Bahcall
- Abstract
Supplementary Figures S1-3; Supplementary Tables S1-3
- Published
- 2023
4. Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Magda Bahcall, Luke J. Taus, Taebo Sim, Prafulla C. Gokhale, Jens Köhler, Stephen Wang, Nam Doo Kim, Cloud P. Paweletz, Zihan Wei, Yanan Kuang, Christie J. Lau, Pasi A. Jänne, Pratik R Chopade, Kshiti Dholakia, Sujuan Guo, Fangxin Hong, Jiannan Guo, and Paul Kirschmeier
- Subjects
Cancer Research ,business.industry ,Cell ,Mutant ,High-Throughput Nucleotide Sequencing ,Merestinib ,Mutagenesis (molecular biology technique) ,Drug resistance ,Article ,In vitro ,respiratory tract diseases ,Molecular Docking Simulation ,Mice ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,In vivo ,medicine ,Cancer research ,Animals ,Humans ,Female ,business ,Protein Kinase Inhibitors ,Tyrosine kinase - Abstract
MET-targeted therapies are clinically effective in MET-amplified and MET exon 14 deletion mutant (METex14) non–small cell lung cancers (NSCLCs), but their efficacy is limited by the development of drug resistance. Structurally distinct MET tyrosine kinase inhibitors (TKIs) (type I/II) have been developed or are under clinical evaluation, which may overcome MET-mediated drug resistance mechanisms. In this study, we assess secondary MET mutations likely to emerge in response to treatment with single-agent or combinations of type I/type II MET TKIs using TPR-MET transformed Ba/F3 cell mutagenesis assays. We found that these inhibitors gave rise to distinct secondary MET mutant profiles. However, a combination of type I/II TKI inhibitors (capmatinib and merestinib) yielded no resistant clones in vitro. The combination of capmatinib/merestinib was evaluated in vivo and led to a significant reduction in tumor outgrowth compared with either MET inhibitor alone. Our findings demonstrate in vitro and in vivo that a simultaneous treatment with a type I and type II MET TKI may be a clinically viable approach to delay and/or diminish the emergence of on target MET-mediated drug-resistance mutations.
- Published
- 2022
5. Validation of ctDNA Quality Control Materials Through a Precompetitive Collaboration of the Foundation for the National Institutes of Health
- Author
-
Robert T. McCormack, Daniel Stetson, Dana E. Connors, J. Carl Barrett, Kenneth D. Cole, Steven P. Lund, Chris Karlovich, Thomas Forbes, P. Mickey Williams, Megan H. Cleveland, Laura M. Yee, Christie J. Lau, Cloud P. Paweletz, Gary J. Kelloff, Benoit Destenaves, Hua-Jun He, and Susan M. Keating
- Subjects
Quality Control ,0301 basic medicine ,Cancer Research ,Engineering ,DNA Copy Number Variations ,media_common.quotation_subject ,Control (management) ,MEDLINE ,Polymerase Chain Reaction ,Circulating Tumor DNA ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Neoplasms ,Original Reports ,Biomarkers, Tumor ,Humans ,Quality (business) ,Diagnostics ,media_common ,business.industry ,Foundation (engineering) ,High-Throughput Nucleotide Sequencing ,United States ,Engineering management ,030104 developmental biology ,National Institutes of Health (U.S.) ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,business - Abstract
PURPOSEWe report the results from a Foundation for the National Institutes of Health Biomarkers Consortium project to address the absence of well-validated quality control materials (QCMs) for circulating tumor DNA (ctDNA) testing. This absence is considered a cause of variance and inconsistencies in translating ctDNA results into clinical actions.METHODSIn this phase I study, QCMs with 14 clinically relevant mutations representing single nucleotide variants, insertions or deletions (indels), translocations, and copy number variants were sourced from three commercial manufacturers with variant allele frequencies (VAFs) of 5%, 2.5%, 1%, 0.1%, and 0%. Four laboratories tested samples in quadruplicate using two allele-specific droplet digital polymerase chain reaction and three (amplicon and hybrid capture) next-generation sequencing (NGS) panels.RESULTSThe two droplet digital polymerase chain reaction assays reported VAF values very close to the manufacturers’ claimed concentrations for all QCMs. NGS assays reported most single nucleotide variants and indels, but not translocations, close to the expected VAF values. Notably, two NGS assays reported lower VAF than expected for all translocations in all QCM mixtures, possibly related to technical challenges detecting these variants. The ability to call ERBB2 copy number amplifications varied across assays. All three QCMs provided valuable insight into assay precision. Each assay across all variant types demonstrated dropouts at 0.1%, suggesting that the QCM can serve for testing of an assay’s limit of detection with confidence claims for specific variants.CONCLUSIONThese results support the utility of the QCM in testing ctDNA assay analytical performance. However, unique designs and manufacturing methods for the QCM, and variations in a laboratory’s testing configuration, may require testing of multiple QCMs to find the best reagents for accurate result interpretation.
- Published
- 2021
6. Plasma ctDNA Response Is an Early Marker of Treatment Effect in Advanced NSCLC
- Author
-
Michael Cheng, Pasi A. Jänne, Christie J. Lau, Marina S.D. Milan, Cloud P. Paweletz, Jonathan W. Riess, Julianna Supplee, Geoffrey R. Oxnard, and Penelope A. Bradbury
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Circulating Tumor DNA ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Original Reports ,Biomarkers, Tumor ,medicine ,Humans ,Treatment effect ,Lung cancer ,Genotyping ,Neoplasm Staging ,Acrylamides ,Aniline Compounds ,business.industry ,medicine.disease ,Response assessment ,Treatment Outcome ,030104 developmental biology ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,business - Abstract
PURPOSE Plasma circulating tumor DNA (ctDNA) analysis is routine for genotyping of advanced non–small-cell lung cancer (NSCLC); however, early response assessment using plasma ctDNA has yet to be well characterized. MATERIALS AND METHODS Patients with advanced EGFR-mutant NSCLC across three phase I NCI osimertinib combination trials were analyzed in this study, and an institutional cohort of patients with KRAS-, EGFR-, and BRAF-mutant advanced NSCLC receiving systemic treatment was used for validation. Plasma was collected before treatment initiation and serially before each cycle of therapy, and key driver mutations in ctDNA were characterized by droplet digital polymerase chain reaction. Timing of plasma versus imaging response was compared in a separate cohort of patients with EGFR-mutant NSCLC treated with osimertinib. Across cohorts, we also studied ctDNA variability before treatment start. RESULTS In the NCI cohort, 14/16 (87.5%) patients exhibited ≥ 90% decrease in mutation abundance by the first on-treatment timepoint (20-28 days from treatment start) with minimal subsequent change. Similarly, 47/56 (83.9%) patients with any decrease in the institutional cohort demonstrated ≥ 90% decrease in mutation abundance by the first follow-up draw (7-30 days from treatment start). All 16 patients in the imaging cohort with radiographic partial response showed best plasma response within one cycle, preceding best radiographic response by a median of 24 weeks (range: 3-147 weeks). Variability in ctDNA levels before treatment start was observed. CONCLUSION Plasma ctDNA response is an early phenomenon, with the majority of change detectable within the first cycle of therapy. These kinetics may offer an opportunity for early insight into treatment effect before standard imaging timepoints.
- Published
- 2021
7. Phase IB Study of Osimertinib in Combination with Navitoclax in EGFR-mutant NSCLC Following Resistance to Initial EGFR Therapy (ETCTN 9903)
- Author
-
Pasi A. Jänne, Christie J. Lau, Erin M. Bertino, D. Ross Camidge, Lynette M. Sholl, Jeffrey A. Moscow, Geoffrey R. Oxnard, Zofia Piotrowska, Jyoti Malhotra, Jill M. Kolesar, Christine L. Hann, Thomas E. Stinchcombe, Liza C. Villaruz, Geoffrey I. Shapiro, Sarah E. Clifford, Alona Muzikansky, Cloud P. Paweletz, Ryan D. Gentzler, Eric B. Haura, and Naoko Takebe
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Maximum Tolerated Dose ,Combination therapy ,Nausea ,medicine.drug_class ,Drug resistance ,Article ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Tissue Distribution ,Osimertinib ,030212 general & internal medicine ,Adverse effect ,Aged ,Aged, 80 and over ,Acrylamides ,Sulfonamides ,Aniline Compounds ,Navitoclax ,business.industry ,Middle Aged ,Prognosis ,ErbB Receptors ,chemistry ,Tolerability ,030220 oncology & carcinogenesis ,Mutation ,Feasibility Studies ,Female ,medicine.symptom ,business ,Follow-Up Studies - Abstract
Purpose: Osimertinib is an effective therapy in EGFR-mutant non–small cell lung cancer (NSCLC), but resistance invariably develops. Navitoclax is an oral inhibitor of BCL-2/BCL-xL that has exhibited synergy with osimertinib in preclinical models of EGFR-mutant NSCLC. In hematologic malignancies, BCL-2 family inhibitors in combination therapy effectively increase cellular apoptosis and decrease drug resistance. Patients and Methods: This single-arm phase Ib study evaluated safety, tolerability, and feasibility of osimertinib and navitoclax, including dose expansion in T790M-positive patients at the recommended phase II dose (RP2D). Eligible patients had advanced EGFR-mutant NSCLC with prior tyrosine kinase inhibitor exposure. Five dose levels were planned with osimertinib from 40 to 80 mg orally daily and navitoclax from 150 to 325 mg orally daily. Results: A total of 27 patients were enrolled (18 in the dose-escalation cohort and nine in the dose-expansion cohort): median age 65, 67% female, 48% exon 19 del, and 37% L858R, median one prior line of therapy. The most common adverse events were lymphopenia (37%), fatigue (22%), nausea (22%), and thrombocytopenia (37%). No dose-limiting toxicities were seen in dose-escalation cohort; osimertinib 80 mg, navitoclax 150 mg was chosen as the RP2D. Most patients (78%) received >95% of planned doses through three cycles. In expansion cohort, objective response rate was 100% and median progression-free survival was 16.8 months. A proapoptotic effect from navitoclax was demonstrated by early-onset thrombocytopenia. Conclusions: Oral combination therapy with navitoclax and osimertinib was safe and feasible at RP2D with clinical efficacy. Early thrombocytopenia was common, supporting an target engagement by navitoclax. Further study of BCL-2/BCL-xL inhibition to enhance osimertinib activity is warranted.
- Published
- 2020
8. Does Testing Error Underlie Liquid Biopsy Discordance?
- Author
-
Geoffrey R. Oxnard, Cloud P. Paweletz, and Christie J. Lau
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,medicine ,Radiology ,Liquid biopsy ,business - Published
- 2019
9. EGFR-Mutated Lung Cancers Resistant to Osimertinib through EGFR C797S Respond to First-Generation Reversible EGFR Inhibitors but Eventually Acquire EGFR T790M/C797S in Preclinical Models and Clinical Samples
- Author
-
Jason Shpilsky, Pasi A. Jänne, Christie J. Lau, Geoffrey R. Oxnard, Ciric To, Daniel B. Costa, Paul A. VanderLaan, Cloud P. Paweletz, Deepa Rangachari, Mierzhati Mushajiang, and Susumu Kobayashi
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,business.industry ,medicine.disease ,respiratory tract diseases ,03 medical and health sciences ,T790M ,030104 developmental biology ,0302 clinical medicine ,Gefitinib ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Adenocarcinoma ,Osimertinib ,Erlotinib ,Liquid biopsy ,Lung cancer ,business ,neoplasms ,medicine.drug ,EGFR inhibitors - Abstract
Introduction Osimertinib is approved for advanced EGFR-mutated NSCLC, and identification of on-target mechanisms of resistance (i.e., EGFR C797S) to this third-generation EGFR inhibitor are evolving. Whether durable control of subsequently osimertinib-resistant NSCLC with the EGFR-sensitizing mutation (SM)/C797S is possible with first-generation EGFR inhibitors (such as gefitinib or erlotinib) remains underreported, as does the resultant acquired resistance profile. Methods We used N-ethyl-N-nitrosourea mutagenesis to determine the profile of EGFR SM/C797S preclinical models exposed to reversible EGFR inhibitors. In addition, we retrospectively probed a case of EGFR SM lung adenocarcinoma treated with first-line osimertinib, followed by second-line erlotinib in the setting of EGFR SM/C797S. Results Use of N-ethyl-N-nitrosourea mutagenesis against the background of EGFR L858R/C797S in conjunction with administration of gefitinib revealed preferential outgrowth of cells with EGFR L858R/T790M/C797S. A patient with EGFR delE746_T751insV NSCLC was treated with osimertinib with sustained response for 10 months before acquiring EGFR C797S. The patient was subsequently treated with erlotinib, with response for a period of 4 months, but disease progression ensued. Liquid biopsy disclosed EGFR delE746_T751insV with T790M and C797S present in cis. Conclusion EGFR SM NSCLC can acquire resistance to osimertinib through development of the EGFR C797S mutation. In this clinical scenario, the tumor may respond transiently to reversible first-generation EGFR inhibitors (gefitinib or erlotinib), but evolving mechanisms of on-target resistance—in clinical specimens and preclinical systems—indicate that EGFR C797S along with EGFR T790M can evolve. This report adds to the growing understanding of tumor evolution or adaptability to sequential EGFR inhibition and augments support for exploring combination therapies to delay or prevent on-target resistance.
- Published
- 2019
10. Dynamic single-cell RNA sequencing identifies immunotherapy persister cells following PD-1 blockade
- Author
-
Jonathan R. Greene, Elena Ivanova, Moataz Noureddine, Luke J. Taus, David Liu, Tyler Teceno, Maria Pinzon-Ortiz, Navin R. Mahadevan, Tran C. Thai, Derek Liu, Andrew Portell, Amir Vajdi, Shunsuke Kitajima, Prafulla C. Gokhale, Paul Kirschmeier, Juan J. Miret, David A. Barbie, Patrick H. Lizotte, Russell W. Jenkins, Cloud P. Paweletz, Kartik Sehgal, Peter S. Hammerman, Christie J. Lau, Carino Gurjao, William D. Hastings, Tetsuo Tani, Silvia Goldoni, and Marios Giannakis
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Cell ,Programmed Cell Death 1 Receptor ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Animals ,RNA-Seq ,General Medicine ,Immunotherapy ,Neoplasms, Experimental ,Blockade ,Neoplasm Proteins ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Stem cell ,Single-Cell Analysis ,CD8 ,Ex vivo ,Research Article - Abstract
Resistance to oncogene-targeted therapies involves discrete drug-tolerant persister cells, originally discovered through in vitro assays. Whether a similar phenomenon limits efficacy of programmed cell death 1 (PD-1) blockade is poorly understood. Here, we performed dynamic single-cell RNA-Seq of murine organotypic tumor spheroids undergoing PD-1 blockade, identifying a discrete subpopulation of immunotherapy persister cells (IPCs) that resisted CD8(+) T cell–mediated killing. These cells expressed Snai1 and stem cell antigen 1 (Sca-1) and exhibited hybrid epithelial-mesenchymal features characteristic of a stem cell–like state. IPCs were expanded by IL-6 but were vulnerable to TNF-α–induced cytotoxicity, relying on baculoviral IAP repeat-containing protein 2 (Birc2) and Birc3 as survival factors. Combining PD-1 blockade with Birc2/3 antagonism in mice reduced IPCs and enhanced tumor cell killing in vivo, resulting in durable responsiveness that matched TNF cytotoxicity thresholds in vitro. Together, these data demonstrate the power of high-resolution functional ex vivo profiling to uncover fundamental mechanisms of immune escape from durable anti–PD-1 responses, while identifying IPCs as a cancer cell subpopulation targetable by specific therapeutic combinations.
- Published
- 2021
11. 248 Immunotherapy persister cells uncovered by dynamic single-cell RNA-sequencing
- Author
-
David Liu, Tyler Teceno, Shunsuke Kitajima, Tran C. Thai, Kartik Sehgal, Carino Gurjao, Moataz Noureddine, Cloud P. Paweletz, Navin R. Mahadevan, William D. Hastings, Prafulla C. Gokhale, Tetsuo Tani, Silvia Goldoni, Andrew Portell, Derek Liu, Peter S. Hammerman, Christie J. Lau, Patrick H. Lizotte, Marios Giannakis, Jonathan R. Greene, Maria Pinzon-Ortiz, Elena Ivanova, Paul Kirschmeier, Juan J. Miret, Russell W. Jenkins, Amir Vadji, David A. Barbie, and Luke J. Taus
- Subjects
business.industry ,T cell ,medicine.medical_treatment ,Antigen presentation ,Immunotherapy ,Immune checkpoint ,medicine.anatomical_structure ,In vivo ,Cancer research ,medicine ,Stem cell ,Autocrine signalling ,business ,Ex vivo - Abstract
Background To understand fundamental mechanisms of immune escape, we leveraged our functional ex vivo platform of murine derived organotypic tumor spheroids (DOTS)1 to determine if drug-tolerant persister cells analogous to oncogene targeted therapies limit efficacy of programmed death (PD)-1 blockade, and to identify therapeutic vulnerabilities to overcome anti-PD-1 (αPD-1) resistance. Methods Murine syngeneic cancer models with well-characterized response to αPD-1 therapy were chosen: MC38 (sensitive) and CT26 (partially resistant). Bulk and single-cell (sc) RNA-sequencing (RNA-seq) were performed on αPD-1 treated DOTS. In vitro culture studies were conducted with or without cytokines (100 ng/ml) or drugs (500 nM). In vivo studies in mice bearing MC38 or CT26 tumors evaluated the combinatorial strategy with PD-1 blockade. We further evaluated our findings in scRNA-seq of an αPD-1 refractory colorectal cancer (CRC) patient tumor.2 Results Bulk RNA-seq of αPD-1 treated DOTS revealed a mesenchymal resistant phenotype with upregulated TNF-α/NFκB signaling (figure 1). scRNA-seq further identified a discrete sub-population of immunotherapy persister cells (IPCs). These cells expressed a stem-like phenotype including downregulation of E2F targets indicative of quiescence, suppression of interferon-γ response genes, induction of hybrid epithelial-to-mesenchymal state, and active IL-6 signaling (figure 1). Ly6a/stem cell antigen-1 (Sca-1) and Snai1 were found to be differentially upregulated in IPCs resistant to PD-1 blockade (not shown). Sca-1 positivity was confirmed in pre-existing tumor populations in vitro (figure 2). When enriched via sorting, these cells remained more persistently Sca-1+ at 96 hours in culture of CT26 compared to MC38 cells, related to increased autocrine IL-6 production by CT26 Sca-1+ cells. Indeed, IL-6 supplementation was capable of expanding Sca-1+ cells in culture (figure 2). Sca-1+ cells expressing ovalbumin peptide were refractory to OT-1 T cell mediated killing and failed to upregulate MHC class-1 antigen presentation (H-2Kb) in response to IL-6, in contrast to interferon-γ (not shown). Analysis of RNA-seq data further identified Birc2/3 as potential targets limiting TNF-mediated apoptosis of these cells (not shown). Notably, Birc2/3 antagonism depleted Sca-1+ IPCs in vitro and significantly potentiated the impact of PD-1 blockade in vivo in MC38, and less robustly in CT26 (figure 3). Evaluation in a microsatellite-instability high CRC patient identified a pre-existent IPC subpopulation within the αPD-1 refractory pre-treatment tumor, with high SNAI1 expression compared to CRC samples in TCGA (figure 4). Conclusions High-resolution functional ex vivo profiling identified Sca-1+/Snai1high stem-like ‘immunotherapy persister cells‘ and uncovered their anti-apoptotic dependencies targetable with Birc2/3 antagonism to augment αPD-1 efficacy. Ethics Approval This study was approved by the Dana-Farber Animal Care and Use Committee and Novartis Institutional Animal Care and Use Committee. Informed written consent to participate in Dana-Farber/Harvard Cancer Center institutional review board (IRB)-approved research protocols was obtained from the human subject. A copy of the written consent is available for review by the Editor of this journal. The study was conducted per the WMA Declaration of Helsinki and IRB-approved protocols. References Jenkins RW, Aref AR, Lizotte PH, Ivanova E, Stinson S, Zhou CW, et al. Ex Vivo Profiling of PD-1 Blockade using organotypic tumor spheroids. Cancer Discov. 2018;8(2):196–668 215. Gurjao C, Liu D, Hofree M, AlDubayan SH, Wakiro I, Su MJ, et al. intrinsic resistance to immune checkpoint blockade in a mismatch repair-deficient colorectal cancer. Cancer Immunol Res 2019;7(8):1230–6.
- Published
- 2020
12. Effect of Erlotinib Plus Bevacizumab vs Erlotinib Alone on Progression-Free Survival in Patients With Advanced EGFR-Mutant Non-Small Cell Lung Cancer: A Phase 2 Randomized Clinical Trial
- Author
-
Anthony J. Jaslowski, Christie J. Lau, Stephen L. Graziano, Maria Q. Baggstrom, Cloud P. Paweletz, Lin Gu, Gregory J. Gerstner, Erin M. Bertino, Everett E. Vokes, Thomas E. Stinchcombe, Pasi A. Jänne, James D. Bearden, Jared Weiss, Xiaofei Wang, and Lyudmila Bazhenova
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,medicine ,030212 general & internal medicine ,Progression-free survival ,Epidermal growth factor receptor ,Lung cancer ,Adverse effect ,neoplasms ,Original Investigation ,biology ,business.industry ,Hazard ratio ,medicine.disease ,respiratory tract diseases ,030220 oncology & carcinogenesis ,biology.protein ,Erlotinib ,business ,medicine.drug - Abstract
IMPORTANCE: Erlotinib is a standard first-line therapy for patients with epidermal growth factor receptor (EGFR)–mutant non–small cell lung cancer (NSCLC). Median progression-free survival (PFS) with erlotinib is approximately 10 months. OBJECTIVE: To determine whether adding bevacizumab to erlotinib treatment results in superior progression-free survival compared with erlotinib alone. DESIGN, SETTING, AND PARTICIPANTS: This phase 2 randomized clinical trial compared erlotinib plus bevacizumab with erlotinib alone in EGFR-mutant NSCLC. The trial was conducted in 17 US academic and community medical centers among 88 patients with EGFR exon 19 deletion or exon 21 L858R mutation based on local testing and stage 4 NSCLC who were eligible for bevacizumab. Patients were enrolled between November 2, 2012, and August 22, 2016, and followed up for a median (range) of 33 (0.7-62.5) months. Data were analyzed on August 28, 2018, and included data from November 2, 2012, to August 20, 2018. INTERVENTIONS: Patients were randomized with equal allocation to 150 mg of oral erlotinib daily alone or with 15 mg/kg of intravenous bevacizumab every 3 weeks. Study therapy continued until disease progression, unacceptable adverse event, or withdrawal of consent. MAIN OUTCOMES AND MEASURES: The primary outcome was PFS as assessed by the investigator; secondary outcomes were objective response rate (ORR), adverse events, and overall survival (OS). Analysis was designed to detect a hazard ratio (HR) of 0.667 for PFS (an improvement from a median PFS of 10 to 15 months). RESULTS: Among 88 patients enrolled, the median (range) age was 63 (31-84) years; 62 patients (70%) were female; 75 (85%) were white, 8 (9%) were African American, 3 (3%) were Asian, and for 2 (2%), data on race were not available. Forty-eight patients (55%) were never smokers, 45 patients (51%) were of Eastern Cooperative Oncology Group performance status 1, and 59 patients (67%) had EGFR exon 19 deletion. Compared with erlotinib, the combination did not result in a significant difference in PFS (HR, 0.81; 95% CI, 0.50-1.31; P = .39; median PFS 17.9 [combination] and 13.5 months [erlotinib]), ORR (81% vs 83%; P = .81), and OS (HR, 1.41; 95% CI, 0.71-2.81; P = .33; median OS, 32.4 months [combination] and 50.6 months [erlotinib]). Adverse events of grade 3 or higher observed in 5 or more patients in the combination and erlotinib arms were skin eruption in 11 (26%) vs 7 (16%) patients, diarrhea in 4 (9%) vs 6 (13%) patients, hypertension in 17 (40%) vs 9 (20%) patients, and proteinuria in 5 (12%) vs 0 (0%) patients. CONCLUSIONS AND RELEVANCE: Erlotinib plus bevacizumab compared with erlotinib did not result in a significant improvement in PFS in EGFR-mutant NSCLC. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01532089.
- Published
- 2019
13. Immuno-PET identifies the myeloid compartment as a key contributor to the outcome of the antitumor response under PD-1 blockade
- Author
-
Xia Bu, Robert A. Weinberg, Mohammad Rashidian, Vincent L. Verschoor, Hidde L. Ploegh, Amir Reza Aref, Stephen C. Kolifrath, Anushka Dongre, Christie J. Lau, Arlene H. Sharpe, Martin W. LaFleur, Gordon J. Freeman, Thao H. Nguyen, M. Inmaculada Barrasa, Yun Zhang, and Cloud P. Paweletz
- Subjects
Myeloid ,medicine.medical_treatment ,Population ,Programmed Cell Death 1 Receptor ,Adenocarcinoma ,CD8-Positive T-Lymphocytes ,Mice ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Distribution (pharmacology) ,Animals ,education ,education.field_of_study ,Multidisciplinary ,CD11b Antigen ,biology ,Chemistry ,RNA ,Immunotherapy ,Neoplasms, Experimental ,Neoplasm Proteins ,medicine.anatomical_structure ,Integrin alpha M ,PNAS Plus ,Positron-Emission Tomography ,Cancer research ,biology.protein ,Female ,Antibody ,Colorectal Neoplasms ,CD8 - Abstract
Immunotherapy using checkpoint-blocking antibodies against PD-1 has produced impressive results in a wide range of cancers. However, the response remains heterogeneous among patients. We used noninvasive immuno-positron emission tomography (PET), using (89)Zr-labeled PEGylated single-domain antibody fragments (nanobodies or VHHs), to explore the dynamics and distribution of intratumoral CD8(+) T cells and CD11b(+) myeloid cells in response to anti–PD-1 treatment in the MC38 colorectal mouse adenocarcinoma model. Responding and nonresponding tumors showed consistent differences in the distribution of CD8(+) and CD11b(+) cells. Anti–PD-1 treatment mobilized CD8(+) T cells from the tumor periphery to a more central location. Only those tumors fully infiltrated by CD8(+) T cells went on to complete resolution. All tumors contained CD11b(+) myeloid cells from the outset of treatment, with later recruitment of additional CD11b(+) cells. As tumors grew, the distribution of intratumoral CD11b(+) cells became more heterogeneous. Shrinkage of tumors in responders correlated with an increase in the CD11b(+) population in the center of the tumors. The changes in distribution of CD8(+) and CD11b(+) cells, as assessed by PET, served as biomarkers to gauge the efficacy of anti–PD-1 treatment. Single-cell RNA sequencing of RNA from intratumoral CD45(+) cells showed that CD11b(+) cells in responders and nonresponders were markedly different. The responders exhibited a dominant population of macrophages with an M1-like signature, while the CD45(+) population in the nonresponders displayed an M2-like transcriptional signature. Thus, by using immuno-PET and single-cell RNA sequencing, we show that anti–PD-1 treatment not only affects interactions of CD8(+) T cells with the tumor but also impacts the intratumoral myeloid compartment.
- Published
- 2019
14. Salivary HPV DNA informs locoregional disease status in advanced HPV-associated oropharyngeal cancer
- Author
-
Julianna Supplee, Cloud P. Paweletz, Abhishek Mogili, Umair Mahmood, Christie J. Lau, Glenn J. Hanna, Robert I. Haddad, and Pasi A. Jänne
- Subjects
Oncology ,Male ,Cancer Research ,Saliva ,medicine.medical_specialty ,Pilot Projects ,Disease ,Circulating Tumor DNA ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Biomarkers, Tumor ,Medicine ,Humans ,Prospective Studies ,030223 otorhinolaryngology ,Papillomaviridae ,Aged ,Neoplasm Staging ,business.industry ,Head and neck cancer ,Papillomavirus Infections ,Liquid Biopsy ,Cancer ,Middle Aged ,medicine.disease ,Prognosis ,Survival Analysis ,Tumor Burden ,Hpv testing ,Oropharyngeal Neoplasms ,Treatment Outcome ,chemistry ,030220 oncology & carcinogenesis ,Cohort ,DNA, Viral ,Biomarker (medicine) ,Feasibility Studies ,Female ,Oral Surgery ,business ,DNA - Abstract
Quantifying tumor DNA in tissue and circulating in blood permits high-quality molecular monitoring to detect and track cancer progression. Evaluating tumor DNA in both blood and saliva in human papillomavirus (HPV)-associated oropharyngeal cancer (OPC) could provide a non-invasive and clinically actionable method for real-time disease detection.We previously validated an ultrasensitive droplet-digital (dd)PCR assay targeting the dominant high-risk HPV subtypes causally linked to OPC. Here we enrolled an observational cohort to evaluate the predictive and prognostic potential of paired plasma-salivary tumor DNA among 21 patients with advanced HPV+OPC.In patients with recurrent, persistent locoregional (LR) disease, median baseline normalized salivary HPV DNA was 10.9 copies/ng total DNA, nearly 20x higher compared with those with distant disease only (p = 0.01). A cutoff of 5 copies/ng yielded 87% sensitivity and 67% specificity for accurately predicting LR disease. Total tumor burden among those with LR disease strongly correlated with salivary HPV DNA levels (R = 0.83, p = 0.02). The rise and fall of salivary HPV DNA predicted treatment failure and response, respectively, in all patients with LR disease, and predated imaging findings. Among paired salivary-plasma (cell-free) cfDNA samples, only higher plasma HPV cfDNA levels were associated with poor outcomes (p 0.01), suggesting that each bodily fluid provides unique information about HPV disease status.Salivary HPV DNA provides valuable information about tumor burden and predicts treatment response in advanced HPV+OPC. Paired blood-saliva samples could be used to monitor HPV DNA with broad applications to inform diagnosis, prognosis, and surveillance in HPV-associated diseases.
- Published
- 2019
15. Concurrent osimertinib plus gefitinib for first-line treatment of EGFR-mutated non-small cell lung cancer (NSCLC)
- Author
-
David M. Jackman, Deepa Rangachari, Daniel B. Costa, Mark M. Awad, Julia K Rotow, Monika D Izdebski, Jacob Sands, Bruce E. Johnson, Geoffrey R. Oxnard, Michael Cheng, Kenneth L. Kehl, Cloud P. Paweletz, David A. Barbie, Paul Marcoux, Christie J. Lau, David J. Kwiatkowski, Pasi A. Jänne, and Kevin A Vasquez
- Subjects
Cancer Research ,Standard of care ,business.industry ,non-small cell lung cancer (NSCLC) ,medicine.disease ,respiratory tract diseases ,First line treatment ,03 medical and health sciences ,Egfr tki ,0302 clinical medicine ,Gefitinib ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Osimertinib ,business ,Egfr tyrosine kinase ,030215 immunology ,medicine.drug - Abstract
9507 Background: First-line treatment with an EGFR tyrosine kinase inhibitor (TKI) is standard of care for patients (pts) with EGFR-mutated NSCLC. The EGFR TKI osimertinib is active against the acquired gefitinib-resistant mutation EGFR T790M, as is gefitinib against the osimertinib-resistant EGFR C797S. Preclinical evidence suggests dual EGFR inhibition with gefitinib + osimertinib may delay emergence of acquired resistance. Methods: This ongoing phase I/II study enrolled pts with stage IV EGFR-mutated (L858R or del19) NSCLC, without prior therapy for metastatic disease. Treatment in dose escalation (n = 6): concurrent osimertinib 40 mg or 80 mg + gefitinib 250 mg daily. In dose expansion (n = 21): osimertinib + gefitinib at the maximum tolerated dose (MTD). Prior to protocol amendment 6 pts received alternating monthly cycles of TKI monotherapy and were excluded from this analysis. The primary endpoints in the dose escalation and expansion phases were, respectively, identification of the MTD and feasibility, defined as receipt of combination therapy for ≥ 6 four-week cycles. Secondary endpoints included overall response rate (ORR), survival outcomes, plasma EGFR mutation clearance (cell free DNA by droplet digital PCR (ddPCR)), and mechanisms of acquired resistance. Results: From May 2017 to July 2019 27 pts were enrolled and evaluable for the primary endpoints. The MTD was osimertinib 80 mg plus gefitinib 250 mg orally daily. In feasibility analysis, 81.5% completed ≥6 cycles combination therapy (1 pt discontinued for progression, 4 for toxicity). The ORR was 85.2% (95% CI 67.5%-94.1%). Best response: 85.2% partial response, 14.8% stable disease. The most common treatment-related adverse effects (TRAEs) (% any grade, % grade 3) were rash (96.3%, 3.7%), diarrhea (85.2%, 11.1%) and dry skin (70.4%, 0%). Plasma ddPCR (n = 25 pts) detected the driver EGFR mutation at baseline in 68% of pts. In these pts, plasma EGFR cleared to undetectable at 2 weeks treatment in 82.4%. At 14.8 months median follow up the median progression free survival was not yet reached. Conclusions: Combination therapy with osimertinib and gefitinib is tolerable for first-line treatment of EGFR-mutated NSCLC and resulted in rapid plasma clearance of the EGFR mutation. The observed ORR is consistent with previously reported first-line response rates to osimertinib. Analysis of survival outcomes and acquired resistance mechanisms are pending data maturity and will facilitate understanding of the role of first-line dual EGFR TKI therapy for this pt population. Clinical trial information: NCT03122717 .
- Published
- 2020
16. Plasma HPV cell-free DNA monitoring in advanced HPV-associated oropharyngeal cancer
- Author
-
Yanan Kuang, Julianna Supplee, Glenn J. Hanna, Umar Mahmood, Robert I. Haddad, Christie J. Lau, Cloud P. Paweletz, and Pasi A. Jänne
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Tumor burden ,Antineoplastic Agents ,Pilot Projects ,Disease ,Kaplan-Meier Estimate ,Polymerase Chain Reaction ,Sensitivity and Specificity ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Papillomaviridae ,Aged ,business.industry ,Head and neck cancer ,Papillomavirus Infections ,Cancer ,Hematology ,Middle Aged ,Viral Load ,medicine.disease ,Precision medicine ,Prognosis ,Tumor Burden ,Oropharyngeal Neoplasms ,030104 developmental biology ,Cell-free fetal DNA ,030220 oncology & carcinogenesis ,Cohort ,DNA, Viral ,Disease Progression ,Female ,business ,Viral load ,Cell-Free Nucleic Acids ,Follow-Up Studies - Abstract
Background Measuring cell-free (cf)DNA in blood and tissues holds significant potential as a minimally invasive method for disease monitoring in cancer. Cancers arising in the oropharynx and causally linked to human papillomavirus (HPV) represent an ideal model in which to interrogate these methods. Patients and methods We designed an ultrasensitive and quantitative droplet digital (dd)PCR assay to detect the five dominant high-risk HPV subtypes linked to oropharyngeal cancer (OPC). We enrolled a pilot observational cohort of 22 patients with advanced HPV+ OPC to evaluate the clinical utility of our assay and explore its predictive and prognostic potential. Results Total tumor burden (TTB) strongly correlated with HPV cfDNA levels (R = 0.91, P = 2.3×10−6) at this cohort size, and in most cases more distant anatomic disease locations predicted increasing HPV cfDNA levels. All participants demonstrated a corresponding change in their HPV cfDNA levels at a median of 16 days (range 12–38) before restaging scans confirming treatment response or progression. Patients with locoregional disease in the head and neck or pulmonary-only metastases had worse outcomes (P = 0.01). Both TTB and median plasma HPV cfDNA levels negatively correlated with survival (R=−0.65, P = 0.01; R=−0.48, P = 0.05, respectively). Conclusion(s) Plasma HPV cfDNA monitoring recapitulates fluctuations in disease status. While blood-based HPV DNA monitoring does not currently have a role in managing HPV+ OPC, these data speak to their broad clinical potential in an era of precision medicine.
- Published
- 2018
17. P1.01-46 Response Assessment Using Plasma Cell-Free DNA (cfDNA) – When Is the Optimal Time to Assess Response?
- Author
-
Geoffrey R. Oxnard, Penelope A. Bradbury, Marina S.D. Milan, Christie J. Lau, Jonathan W. Riess, Cloud P. Paweletz, Julianna Supplee, and Michael Cheng
- Subjects
Pulmonary and Respiratory Medicine ,Response assessment ,medicine.anatomical_structure ,Oncology ,business.industry ,Cancer research ,Medicine ,Plasma cell ,business ,Time optimal ,Free dna - Published
- 2019
18. Abstract 452: Salivary HPV cell free DNA levels predict locoregional disease burden and response in oropharyngeal cancer
- Author
-
Glenn J. Hanna, Christie J. Lau, Umair Mahmood, Robert I. Haddad, Cloud P. Paweletz, and Pasi A. Janne
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: The human papillomavirus (HPV) is linked to the majority of oropharyngeal squamous cell carcinomas (OPSCC). We have previously shown that tumor-derived HPV cell-free (cf)DNA can be detected and quantified in circulation with high sensitivity and specificity using droplet digital (dd)PCR1. Further, we have shown that plasma cfDNA levels correlate with disease burden and indicate treatment response in advanced OPSCC. In this study we pair plasma with salivary HPV cfDNA to understand its clinical application in monitoring locoregional spread of disease. Methods: We present a proof-of-concept prospective observational cohort of recurrent, metastatic (R/M) HPV+ OPSCC patients treated with standard systemic therapies or on active surveillance. We utilized ultrasensitive ddPCR to identify and quantify both plasma and salivary HPV cfDNA (subtypes 16, 18, 31, 33 and 45) at multiple time points. Salivary HPV cfDNA was normalized to total DNA concentration as measured by fluorometric quantification. We then compared matched plasma and normalized salivary HPV cfDNA concentrations at various timepoints with clinical parameters, such as tumor burden and therapeutic response. Results: Clinicopathologic data from 15 R/M patients revealed a male cohort with a median age of 57 at diagnosis. Five (33%) were on immunotherapy and 6 (40%) on standard chemotherapy during the study period (3 months). Salivary HPV cfDNA was detected in 13/15 (87%) samples (range 0-729 copies/mL) in at least one timepoint during the study (plasma cfDNA was detected in 12/15 or 80% of the cohort, range 0-12,949 copies/mL). All patients with measurable locoregional (LR) disease had detectable salivary cfDNA. While matched plasma and salivary cfDNA levels showed no correlation among individual patients, plasma cfDNA levels correlated with site of disease (LR, pulmonary or extra-pulmonary) (p < 0.001), while median salivary cfDNA levels strongly correlated with LR tumor burden (R = 0.81, p < 0.001). Salivary cfDNA levels declined by 80% of their baseline value within a median of 6.5 days among all patients with LR disease experiencing a response to treatment. Page 1 of 1 Conclusion: Our results suggest that high sensitivity salivary HPV cfDNA levels correlate with locoregional disease burden and can be an early indicator of locoregional treatment response, while our prior work has supported the role of plasma HPV cfDNA levels in monitoring metastatic disease. Evaluating paired plasma and salivary HPV cfDNA levels in a curative HPV OPSCC population is underway to further validate their prognostic and predictive potential. 1Hanna GJ, Supplee JG, Kuang Y, Mahmood U, Lau CJ, Haddad RI, et al. Plasma HPV cell-free DNA monitoring in advanced HPV-associated oropharyngeal cancer. Ann Oncol. 2018;29:1980–6. Page 2 of 1 Citation Format: Glenn J. Hanna, Christie J. Lau, Umair Mahmood, Robert I. Haddad, Cloud P. Paweletz, Pasi A. Janne. Salivary HPV cell free DNA levels predict locoregional disease burden and response in oropharyngeal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 452.
- Published
- 2019
19. Osimertinib (Osi) plus necitumumab (Neci) in EGFR-mutant NSCLC: An ETCTN California cancer consortium phase I study
- Author
-
Jonathan W. Riess, Geoffrey R. Oxnard, Cloud P. Paweletz, David R. Gandara, Sukhmani K. Padda, Philip C. Mack, Karen L. Reckamp, Jill M. Kolesar, Edward M. Newman, Heather A. Wakelee, Marianna Koczywas, Zofia Piotrowska, Lynette M. Sholl, Pasi A. Jänne, Primo N. Lara, Jeffrey A. Moscow, Susan Groshen, and Christie J. Lau
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Mutant ,Cancer ,medicine.disease ,Phase i study ,03 medical and health sciences ,Egfr tki ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Osimertinib ,business ,Progressive disease ,030215 immunology ,Necitumumab - Abstract
9057 Background: Osi (3rd gen EGFR TKI) has robust activity in 1st line EGFR mutant NSCLC and TKI resistant T790Mpos NSCLC but progressive disease (PD) occurs and outcomes with Osi alone are poor in T790Mneg, C797Xpos and EGFR exon 20 insertion (ins20) disease. This study examined the EGFR monoclonal antibody Neci with Osi in select settings of EGFR TKI resistance. Methods: Using a 3+3 design, Neci was examined in advanced EGFR mutant NSCLC at dose levels (DL) of 600 mg (DL1) & 800 mg (DL2) D1, D8 IV q21 days + Osi 80 mg qd. Four expansion cohorts (ExC; 18 each) included: A) T790Mneg PD on 1st/2nd gen TKI as last therapy, B) T790Mneg PD on 3rd gen TKI, C) T790Mpos PD on 3rd gen TKI, D) EGFR ex20ins PD on chemotherapy. Central T790M testing by ddPCR in ExC A-C. Additional studies performed include: NGS panel of > 400 genes, EGFR FISH, plasma for PK and serial EGFR ctDNA by ddPCR as exploratory analyses. Adverse events (AEs) graded (Gr) by CTCAEv5 with ORR and PFS by RECIST 1.1. Results: Dose escalation and ExC B completed accrual. In total 55 pts were evaluable (Table). 1 pt had DLT at DL2, Gr 3 sinus bradycardia. Drug related Gr 3 AEs were seen in 27% (15) of pts, mainly rash (7;13%). ctDNA showed decreased mutant allele frequency with therapy in all pts studied with detected EGFR at baseline, with complete plasma clearance in a pt with detectable C797S. Conclusions: The RP2D (Osi 80 mg qd and Neci 800 mg D1, D8 IV on q21d cycle) is feasible and tolerable. In ExC A,T790Mneg pts with 1st/2nd gen EGFR TKI as last treatment, using curtailed sampling, the pre-specified efficacy signal was reached for Osi + Neci comparing favorably to Osi alone in analogous pts from the AURA trial. Clinical activity was also seen in EGFR-dependent resistance (T790Mpos C797Spos) after PD on 3rd gen TKI and in EGFR ins 20. Clinical trial information: NCT02496663. [Table: see text]
- Published
- 2019
20. A randomized phase II trial of erlotinib or erlotinib and bevacizumab in patients with advanced EGFR mutant non-small cell lung cancer (NSCLC)
- Author
-
Stephen L. Graziano, Tom Stinchcombe, Lin Gu, A.J. Jaslowski, Cloud P. Paweletz, Maria Q. Baggstrom, Lyudmila Bazhenova, James D. Bearden, Pasi A. Jänne, G.J. Gerstner, Everett E. Vokes, Xiaofei Wang, Erin M. Bertino, Christie J. Lau, and Jared Weiss
- Subjects
Oncology ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Mutant ,non-small cell lung cancer (NSCLC) ,Hematology ,medicine.disease ,Internal medicine ,medicine ,In patient ,Erlotinib ,business ,medicine.drug - Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.