Lynn Navale
Amgen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Lynn Navale.
Lancet Oncology | 2014
Bradley J. Monk; Andres Poveda; Ignace Vergote; Francesco Raspagliesi; Keiichi Fujiwara; Duk Soo Bae; Isabelle Ray-Coquard; Diane Provencher; Beth Y. Karlan; Catherine Lhommé; Gary Richardson; Dolores Gallardo Rincon; Robert L. Coleman; Thomas J. Herzog; Christian Marth; Arija Brize; Michel Fabbro; Andrés Redondo; Aristotelis Bamias; Marjan Tassoudji; Lynn Navale; Douglas Warner; Amit M. Oza
BACKGROUND Angiogenesis is a valid target in the treatment of epithelial ovarian cancer. Trebananib inhibits the binding of angiopoietins 1 and 2 to the Tie2 receptor, and thereby inhibits angiogenesis. We aimed to assess whether the addition of trebananib to single-agent weekly paclitaxel in patients with recurrent epithelial ovarian cancer improved progression-free survival. METHODS For this randomised, double-blind phase 3 study undertaken between Nov 10, 2010, and Nov 19, 2012, we enrolled women with recurrent epithelial ovarian cancer from 32 countries. Patient eligibility criteria included having been treated with three or fewer previous regimens, and a platinum-free interval of less than 12 months. We enrolled patients with a computerised interactive voice response system, and patients were randomly assigned using a permuted block method (block size of four) in a 1:1 ratio to receive weekly intravenous paclitaxel (80 mg/m(2)) plus either weekly masked intravenous placebo or trebananib (15 mg/kg). Patients were stratified on the basis of platinum-free interval (≥0 and ≤6 months vs >6 and ≤12 months), presence or absence of measurable disease, and region (North America, western Europe and Australia, or rest of world). The sponsor, investigators, site staff, and patients were masked to the treatment assignment. The primary endpoint was progression-free survival assessed in the intention-to-treat population. The trial is registered with ClinicalTrials.gov, NCT01204749, and is no longer accruing patients. FINDINGS 919 patients were enrolled, of whom 461 were randomly assigned to the trebananib group and 458 to the placebo group. Median progression-free survival was significantly longer in the trebananib group than in the placebo group (7·2 months [5·8-7·4] vs 5·4 months [95% CI 4·3-5·5], respectively, hazard ratio 0·66, 95% CI 0·57-0·77, p<0·0001). Incidence of grade 3 or higher adverse events was similar between treatment groups (244 [54%] of 452 patients in the placebo group vs 258 [56%] of 461 patients in the trebananib group). Trebananib was associated with more adverse event-related treatment discontinuations than was placebo (77 [17%] patients vs 27 [6%], respectively) and higher incidences of oedema (294 [64%] patients had any-grade oedema in the trebananib group vs 127 [28%] patients in the placebo group). Grade 3 or higher adverse events included ascites (34 [8%] in the placebo group vs 52 [11%] in the trebananib group), neutropenia (40 [9%] vs 26 [6%]), and abdominal pain (21 [5%] vs 22 [5%]). We recorded serious adverse events in 125 (28%) patients in the placebo group and 159 (34%) patients in the trebananib group. There was a difference of 2% or less in class-specific adverse events associated with anti-VEGF therapy (hypertension, proteinuria, wound-healing complications, thrombotic events, gastrointestinal perforations), except bleeding, which was more common in the placebo group than in the trebananib group (75 [17%] vs 46 [10%]). INTERPRETATION Inhibition of angiopoietins 1 and 2 with trebananib provided a clinically meaningful prolongation in progression-free survival. This non-VEGF anti-angiogenesis option for women with recurrent epithelial ovarian cancer should be investigated in other settings and in combination with additional agents. Although oedema was increased, typical anti-VEGF associated adverse events were not prominent. FUNDING Amgen.
Cancer | 2007
J. Randolph Hecht; Amita Patnaik; Jordan Berlin; Alan P. Venook; Imtiaz Malik; Simon Tchekmedyian; Lynn Navale; Rafael G. Amado; Neal J. Meropol
The safety and efficacy of the fully human antibody panitumumab was evaluated in patients with metastatic colorectal cancer refractory to available therapies.
Clinical Cancer Research | 2008
Louis M. Weiner; Arie S. Belldegrun; Jeffrey Crawford; Anthony W. Tolcher; Pamela Lockbaum; Rosalin H. Arends; Lynn Navale; Rafael G. Amado; Gisela Schwab; Robert A. Figlin
Purpose: This phase 1 study evaluated the safety, pharmacokinetics, and activity of panitumumab, a fully human, IgG2 monoclonal antibody that targets the epidermal growth factor receptor in patients with previously treated epidermal growth factor receptor–expressing advanced solid tumors. Experimental Design: Sequential cohorts were enrolled to receive four i.v. infusions of panitumumab monotherapy at various doses and schedules. Safety was continuously monitored. Serum samples for pharmacokinetic, immunogenicity, and chemistry assessments were drawn at preset intervals. Tumor response was assessed at week 8. Results: Ninety-six patients received panitumumab. Median (range) age was 61 years (32-79 years), and 72 (75%) patients were male. Tumor types were 41% colorectal cancer, 22% prostate, 16% renal, 15% non–small cell lung, 3% pancreatic, 3% esophageal/gastroesophageal, and 1% anal. The overall incidence of grade 3 or 4 adverse events was 32% and 7%, respectively. The incidence of skin-related toxicities was dose dependent. No maximum tolerated dose was reached. No human anti-panitumumab antibodies were detected. No investigator-determined panitumumab infusion-related reactions were reported. Serum panitumumab concentrations were similar in the 2.5 mg/kg weekly, 6.0 mg/kg every 2 weeks, and 9.0 mg/kg every 3 weeks dose cohorts. Five of 39 patients (13%) with colorectal cancer had a confirmed partial response, and 9 of 39 patients (23%) with colorectal cancer had stable disease. Conclusions: Panitumumab was well tolerated with comparable exposure and safety profiles for the weekly, every 2 weeks, and every 3 weeks administration schedules. Rash and dry skin occurred more frequently in the dose cohorts receiving ≥2.5 mg/kg weekly dose. Panitumumab has single-agent antitumor activity, most notably in patients with advanced colorectal cancer.
Journal of Clinical Oncology | 2015
Michael B. Atkins; Gwenaelle Gravis; Kazimierz Drosik; Tomasz Demkow; Piotr Tomczak; Shirley Wong; M. Dror Michaelson; Toni K. Choueiri; Benjamin Wu; Lynn Navale; Douglas Warner; Alain Ravaud
PURPOSE Trebananib, an investigational recombinant peptide-Fc fusion protein, neutralizes the receptor-ligand interaction between Tie2 and angiopoietin-1/2. This phase II study was conducted to evaluate trebananib plus sunitinib, a vascular endothelial growth factor receptor inhibitor, in patients with metastatic clear cell renal cell carcinoma. PATIENTS AND METHODS Adults with metastatic renal cell carcinoma were enrolled sequentially onto two cohorts that received sunitinib 50 mg once per day for 4 weeks on and 2 weeks off and intravenous trebananib once per week at a dose of 10 mg/kg in cohort A or 15 mg/kg in cohort B. The primary end points were incidences of adverse events (AEs) and dose interruptions of sunitinib during the first 12 weeks of treatment. Secondary end points included objective response rate and progression-free survival. RESULTS Eighty-five patients were enrolled: 43 in cohort A, and 42 in cohort B. During the first 12 weeks of treatment, 58% and 57% of patients in cohorts A and B, respectively, had sunitinib dose interruptions (dose decrease, withholding, or withdrawal). The most frequent AEs were diarrhea (cohort A, 74%; cohort B, 67%), mucosal inflammation (cohort A, 49%; cohort B, 60%), and hypertension (cohort A, 52%; cohort B, 45%). AEs of grade 3 or greater occurred in 58% of patients in cohort A and in 69% of patients in cohort B. The objective response rate was 58% and 63% in cohorts A and B, respectively. The median progression-free survival time was 13.9 months (95% CI, 10.4 to 19.2) and 16.3 months (95% CI, 13.1 to 21.4) in cohorts A and B, respectively. The median overall survival time was 36 months (95% CI, 25.2 to not estimable) in cohort A and was not estimable (median follow-up, 25 months) in cohort B. CONCLUSION Trebananib plus sunitinib seemed to increase toxicity at the tested doses. Efficacy results suggest a potential benefit for the addition of trebananib to sunitinib.
Cancer Research | 2017
Frederick L. Locke; Sattva S. Neelapu; Nancy L. Bartlett; Lazaros J. Lekakis; David B. Miklos; Caron A. Jacobson; Ira Braunschweig; Olalekan O. Oluwole; Tanya Siddiqi; Yi Lin; John M. Timmerman; Jonathan W. Friedberg; Adrian Bot; John J. Rossi; Lynn Navale; Yizhou Jiang; Jeff Aycock; Meg Elias; Jeff Wiezorek; William Y. Go
Introduction: Outcomes for pts with refractory aggressive NHL are poor with current therapies (Crump, ASCO 2016). Results from the interim analysis of (n=62) of ZUMA-1, the 1st multicenter trial of an anti-CD19 chimeric antigen receptor (CAR) T cell, axi-cel, in refractory aggressive NHL, showed an objective response rate (ORR) of 79% (complete response [CR] 52%; Blood 2016;128:LBA-6). Here we present results from the primary analysis of ZUMA-1. Methods: Pts received a target dose of 2 × 106 anti-CD19 CAR T cells/kg after lowdose conditioning with cy/flu. Eligible pts (≥18 y) had diffuse large B cell lymphoma (DLBCL), primary mediastinal B cell lymphoma (PMBCL) or transformed follicular lymphoma (TFL); an ECOG performance status (PS) 0-1; and refractory disease (progressive or stable disease as best response to last prior therapy, or relapsed ≤12 m of autologous stem cell transplant [ASCT]). The primary endpoint for this analysis was ORR in the combined DLBCL, PMBCL, and TFL population. Key secondary endpoints were duration of response (DOR), overall survival (OS), and frequency of adverse events (AEs). The primary analysis was triggered when 92 pts had at least 6 m of follow-up. Results: As of January 27, 2017, 111 pts from 22 institutions were enrolled; 101 pts (91%) received axi-cel. Median age was 58 y (range, 23-76), 67% male, 85% stage IIIIV, 47% IPI 3-4, 77% refractory to ≥2nd line of therapy, and 21% relapsed ≤12 m of ASCT. Axi-cel was successfully manufactured in 110/111 (99%) pts with an average turnaround time from apheresis to the clinical site of 17 d. With an ORR of 82% (n = 92; P Conclusions: Axi-cel significantly improved ORR in patients with refractory aggressive NHL. The CR rate was 7-fold higher compared to historical controls (Crump, ASCO 2016) and nearly half the patients have an ongoing response. Axi-cel demonstrated significant clinical benefit with a manageable safety profile in pts lacking curative treatment options.[F.L.L. and S.S.N. contributed equally to this study.] Funding source: Kite Pharma and in part by funding from the Leukemia & Lymphoma Society (LLS) Therapy Acceleration Program treatment options. Citation Format: Frederick L. Locke, Sattva S. Neelapu, Nancy L. Bartlett, Lazaros J. Lekakis, David Miklos, Caron A. Jacobson, Ira Braunschweig, Olalekan Oluwole, Tanya Siddiqi, Yi Lin, John Timmerman, Jonathan W. Friedberg, Adrian Bot, John Rossi, Lynn Navale, Yizhou Jiang, Jeff Aycock, Meg Elias, Jeff Wiezorek, William Y. Go. Primary results from ZUMA-1: a pivotal trial of axicabtagene ciloleucel (axicel; KTE-C19) in patients with refractory aggressive non-Hodgkin lymphoma (NHL) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT019. doi:10.1158/1538-7445.AM2017-CT019
Cancer Research | 2017
Frederick L. Locke; John J. Rossi; Xiaodong Xue; Sattva S. Neelapu; Daniel H. Ryan; Armin Ghobadi; Lazaros J. Lekakis; David B. Miklos; Caron A. Jacobson; Ira Braunschweig; Olalekan O. Oluwole; Tanya Siddiqi; Yi Lin; John M. Timmerman; Patrick M. Reagan; Marika Sherman; Janice Nagatani; Xiao Zhang; Lynn Navale; William Y. Go; Jeff Wiezorek; Adrian Bot
Introduction: Axicabtagene ciloleucel (axi-cel) is an autologous anti-CD19 chimeric antigen receptor (CAR) T cell therapy. ZUMA-1 is a multicenter registrational trial of axi-cel in patients with refractory, aggressive B-cell non-Hodgkin lymphoma. In a pre-specified interim analysis, ZUMA-1 met its primary endpoint with a 76% objective response rate and 47% complete response. The incidence of Grade 3 and higher (Gr 3+) cytokine release syndrome (CRS) and neurologic events (NE) was 13% and 29%, respectively, in 93 patients with 1 month follow-up (Neelapu ASH 2016). We present novel immune signatures of Gr 3+ CRS and NE by analyzing the axi-cel-related biomarker profile in association with clinical outcomes. Methods: In this interim analysis of 62 patients, 44 serum analytes pre- and post-axi-cel treatment were measured via ELISA at multiple timepoints during the first month. The number of CAR+ cells in blood was determined by qPCR. Kinetics and association of these markers with CRS and NE were analyzed. The treatment-related profile was defined by analytes with at least double an increase over baseline in at least 50% of patients. These analytes were selected and evaluated for association with Gr 3+ CRS or NE. Immune signatures were determined based on p-values applied to peak or cumulative levels of analytes and adjusted for multiplicity (stepdown method). Results: Out of 44 analytes measured, 12 demonstrated an increase by double over baseline in at least 50% of patients. This axi-cel-related biomarker profile includes immune proliferative/modulating cytokines, pro-inflammatory cytokines, markers of myeloid activation, and chemokines. Markers with the highest median-fold increase were IFNγ (44x), IL-10 (31x), IL-6 (26x), IL-15 (20x), and GRZB (17x). These peaked within 7-14 days and generally returned to baseline within 1 month post-treatment. Within this panel of 12 analytes elevated post-axi-cel treatment, markers associated with Gr 3+ CRS (p Citation Format: Frederick L. Locke, John Rossi, Xiaodong Xue, Sattva S. Neelapu, Daniel H. Ryan, Armin Ghobadi, Lazaros J. Lekakis, David Miklos, Caron A. Jacobson, Ira Braunschweig, Olalekan Oluwole, Tanya Siddiqi, Yi Lin, John Timmerman, Patrick M. Reagan, Marika Sherman, Janice Nagatani, Xiao Zhang, Lynn Navale, William Y. Go, Jeff Wiezorek, Adrian Bot. Immune signatures of cytokine release syndrome and neurologic events in a multicenter registrational trial (ZUMA-1) in subjects with refractory diffuse large B cell lymphoma treated with axicabtagene ciloleucel (KTE-C19) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT020. doi:10.1158/1538-7445.AM2017-CT020
Molecular Therapy | 2016
Marc Better; Vijay Chiruvolu; James Oliver; Emily Lowe; John M. Rossi; Arianne Perez; Lynn Navale; Adrian Bot
Introduction: An ongoing anti-CD19 CAR T cell study with KTE-C19 in subjects with refractory, aggressive NHL achieved objective responses in 5/7 subjects, including 4 complete remissions (Locke, ASH 2015). To support this trial (ZUMA-1, NCT02348216) and other multicenter trials in lymphoma, we developed a robust and efficient strategy to generate autologous anti-CD19 CAR-engineered T cell products. The newly developed process aims to minimize the time between subject leukapheresis and product administration, and generate a KTE-C19 lot for every enrolled subject. Methods: Upon confirmation of eligibility, leukapheresis was performed to process 12-15 L of blood targeting collection of 5-10 × 109 peripheral blood mononuclear cells (PBMC). After collection at the investigational site, subject apheresis material was shipped to the central manufacturing site, where it was processed to enrich for the T cell-containing PBMC fraction on a closed system Ficoll™ gradient. In a closed bag system, T cells in the PBMC fraction were then activated using an anti-CD3 monoclonal antibody and cultured in serum-free medium containing 300 IU/ml of IL-2. Magnetic beads were not used for either cell selection or activation. Activated T cells were transduced with a gamma retroviral vector that contains the anti-CD19 CAR gene and further expanded for 4 to 6 days to achieve the target cell dose of 2 × 106 CAR-positive T cells/kg (minimum of 1 × 106). Final KTE-C19 product was washed, cryopreserved and tested for identity, potency, and adventitious agents. In-process samples were collected for analysis by flow cytometry for CAR expression and other characteristics. After meeting acceptance criteria, the KTE-C19 product was shipped back to the clinical sites using a validated cryo-shipper. Results: 7 subjects were dosed in the phase 1 portion of ZUMA-1. KTE-C19 was successfully manufactured in all cases despite a broad range of baseline leukocyte counts (median 5.4×103 cells/µl, 2.1-11.1) and low numbers of baseline lymphocytes (median 0.9×103 lymphocytes/µl, 0.1-1.4) prior to apheresis. Apheresed cell populations used to generate clinical lots had a broad range of phenotypic characteristics including lymphocyte / monocyte ratio (median 1.1, 0.04-3.5), CD8/CD4 ratio (median 4.2, 0.3-7.7), and naive plus central memory (Tcm) / more differentiated T cells (median 0.3, 0.04-1.3). Fold-expansion of T cells was consistent among the 7 product lots (average 6-fold) from transduction to harvest. All KTE-C19 lots contained predominantly CD3+ T cells (median 96%; 90-99%), with CD8+ T cells (median 57%, 27-82%) and CD4+ T cells (median 43%, 18-73%). Both CD4+ and CD8+ T cells expressed CAR at similar levels. The product lots contained predominantly effector memory (median 42%, range 30-56%) and Tcm cells (median 34%, 15-58%), with the remainder being naive (median 14%, 6-19%) and effector T cells (median 5%, 1-22%). All KTE-C19 lots met release specifications and were available for clinical administration within ~2 weeks of apheresis. Conclusions: We developed a robust KTE-C19 manufacturing process that successfully generated biologically and clinically active product irrespective of the characteristics of the starting cell populations processed to date. Product lots met release specifications and were available for subject treatment within the target timeframe. This centralized manufacturing process is well suited to support multicenter clinical trials.
Cancer Research | 2016
Armin Ghobadi; Frederick L. Locke; Sattva S. Neelapu; Tanya Siddiqi; Julio C. Chavez; Chitra Hosing; Nancy L. Bartlett; Lihua E. Budde; Adrian Bot; John M. Rossi; Marika Sherman; Lynn Navale; Meg Elias; Jeff Wiezorek; William Y. Go
Introduction: Anti-CD19 CAR T cells with CD3 zeta and CD28 signaling domains showed durable remissions in subjects with relapsed/refractory advanced B cell malignancies at the NCI (Kochenderfer, JCO 2014). KTE-C19 utilizes the same CAR construct as investigated in the NCI study in a 6-8 day manufacturing process (Better, ASCO 2014). Updated results from the phase 1 portion of ZUMA-1 are presented here. Methods: Subjects received KTE-C19 at a target dose of 2 × 10⁁6 anti-CD19 CAR T cells/kg after a fixed dose conditioning chemo regimen of cyclophosphamide and fludarabine. The primary objective of phase 1 was to evaluate the safety of KTE-C19 as determined by the incidence of dose-limiting toxicities (DLT). Key secondary objectives were ORR, duration of response, and biomarkers. Key inclusion criteria were ? 18 years old, ECOG 0-1, and chemo-refractory disease defined as progressive disease or stable disease as best response to last line of therapy, or disease progression ? 12 months after ASCT. Results: As of 20 Nov 2015, 7 subjects were dosed. All subjects were evaluable for safety and 6 were evaluable for efficacy with a median follow up time of 13 weeks post KTE-C19 infusion. 1 subject experienced a DLT of grade (gr) 4 encephalopathy and gr 4 cytokine release syndrome (CRS) and died due to an intracranial hemorrhage deemed unrelated to KTE-C19 per the investigator. CRS and neurotoxicity were managed with supportive care, α-IL6R and steroids. 5 of 7 (71%) subjects responded including 4 CRs (57%). CAR T cells peaked within 2 weeks and were detectable 1-3+ months post infusion. Conclusions: The KTE-C19 regimen evaluated was safe for further study. The predominant toxicities include CRS and neurotoxicity which are generally reversible. CRs and PRs have been observed in subjects with refractory disease. The potentially pivotal phase 2 portion of the study is ongoing (NCT02348216). This study is supported in part by funding from The Leukemia & Lymphoma Society (LLS) Therapy Acceleration Program® Citation Format: Armin Ghobadi, Frederick L. Locke, Sattva S. Neelapu, Tanya Siddiqi, Julio C. Chavez, Chitra M. Hosing, Nancy L. Bartlett, Lihua E. Budde, Adrian Bot, John M. Rossi, Marika Sherman, Lynn Navale, Meg Elias, Jeff Wiezorek, William Y. Go. Updated phase I results from ZUMA-1: a phase I-II multicenter study evaluating the safety and efficacy of KTE-C19 (anti-CD19 CAR T cells) in subjects with refractory aggressive non-Hodgkin lymphoma (NHL). [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 CT135.
Journal of Clinical Oncology | 2005
I. Malik; J. R. Hecht; Amita Patnaik; Alan P. Venook; Jordan Berlin; G. Croghan; Lynn Navale; M. MacDonald; S. Jerian; Neal J. Meropol
Cancer Chemotherapy and Pharmacology | 2012
Jian-Feng Lu; Erik Rasmussen; Beth Y. Karlan; Ignace Vergote; Lynn Navale; Mita Kuchimanchi; Rebeca Melara; Daniel E. Stepan; David M. Weinreich; Yu-Nien Sun