Network


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

Hotspot


Dive into the research topics where Nancy L. Lewis is active.

Publication


Featured researches published by Nancy L. Lewis.


Journal of Clinical Oncology | 2010

Phase I Pharmacologic and Biologic Study of Ramucirumab (IMC-1121B), a Fully Human Immunoglobulin G1 Monoclonal Antibody Targeting the Vascular Endothelial Growth Factor Receptor-2

Jennifer Spratlin; Roger B. Cohen; Matthew Eadens; Lia Gore; D. Ross Camidge; Sami G. Diab; Stephen Leong; Cindy L. O'Bryant; Laura Chow; Natalie J. Serkova; N. J. Meropol; Nancy L. Lewis; E. Gabriela Chiorean; Floyd Fox; Hagop Youssoufian; Eric K. Rowinsky; S. Gail Eckhardt

PURPOSE To evaluate the safety, maximum-tolerated dose (MTD), pharmacokinetics (PKs), pharmacodynamics, and preliminary anticancer activity of ramucirumab (IMC-1121B), a fully human immunoglobulin G(1) monoclonal antibody targeting the vascular endothelial growth factor receptor (VEGFR)-2. PATIENTS AND METHODS Patients with advanced solid malignancies were treated once weekly with escalating doses of ramucirumab. Blood was sampled for PK studies throughout treatment. The effects of ramucirumab on circulating vascular endothelial growth factor-A (VEGF-A), soluble VEGFR-1 and VEGFR-2, tumor perfusion, and vascularity using dynamic contrast-enhanced magnetic resonance imaging were assessed. Results Thirty-seven patients were treated with 2 to 16 mg/kg of ramucirumab. After one patient each developed dose-limiting hypertension and deep venous thrombosis at 16 mg/kg, the next lower dose (13 mg/kg) was considered the MTD. Nausea, vomiting, headache, fatigue, and proteinuria were also noted. Four (15%) of 27 patients with measurable disease had a partial response (PR), and 11 (30%) of 37 patients had either a PR or stable disease lasting at least 6 months. PKs were characterized by dose-dependent elimination and nonlinear exposure consistent with saturable clearance. Mean trough concentrations exceeded biologically relevant target levels throughout treatment at all dose levels. Serum VEGF-A increased 1.5 to 3.5 times above pretreatment values and remained in this range throughout treatment at all dose levels. Tumor perfusion and vascularity decreased in 69% of evaluable patients. CONCLUSION Objective antitumor activity and antiangiogenic effects were observed over a wide range of dose levels, suggesting that ramucirumab may have a favorable therapeutic index in treating malignancies amenable to VEGFR-2 inhibition.


Journal of Clinical Oncology | 2003

Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma.

Steven J. Cohen; Linus Ho; Sulabha Ranganathan; James L. Abbruzzese; R. Katherine Alpaugh; Mary Beard; Nancy L. Lewis; Susan McLaughlin; André Rogatko; Juan Jose Perez-Ruixo; Amanda M. Thistle; Tom Verhaeghe; Hao Wang; Louis M. Weiner; John J. Wright; Gary R. Hudes; N. J. Meropol

PURPOSE R115777 is a selective nonpeptidomimetic inhibitor of farnesyltransferase (FTase), one of several enzymes responsible for posttranslational modification that is required for the function of p21(ras) and other proteins. Given that RAS mutations are nearly universal in pancreatic cancer and R115777 demonstrated preclinical activity against pancreatic cell lines and xenografts, this phase II study was undertaken to determine its clinical activity and effect on target proteins in patients with measurable metastatic pancreatic adenocarcinoma. PATIENTS AND METHODS Twenty patients who had not received prior therapy for metastatic disease were treated with 300 mg of R115777 orally every 12 hours for 21 of 28 days. Inhibition of FTase activity in peripheral-blood mononuclear cells was measured using a lamin B C-terminus peptide as substrate. Western blot analysis was performed to monitor farnesylation status of the chaperone protein HDJ-2. RESULTS No objective responses were seen. Median time to progression was 4.9 weeks, and median survival time was 19.7 weeks. The estimated 6-month survival rate was 25%, with no patients progression-free at 6 months. Grade 3/4 toxicities were liver enzyme elevation, anemia, neutropenia, thrombocytopenia, fatigue, nausea/vomiting, rash, and anorexia. FTase activity (mean +/- SD) decreased by 49.8% +/- 9.8% 4 hours after treatment on day 1 and 36.1% +/- 24.8% before treatment on day 15. HDJ-2 farnesylation (mean +/- SD) decreased by 33.4% +/- 19.8% on day 15. CONCLUSION Although treatment with R115777 resulted in partial inhibition of FTase activity in mononuclear cells, it did not exhibit single-agent antitumor activity in patients with previously untreated metastatic pancreatic cancer.


Journal of Clinical Oncology | 2009

Effects of Food on the Relative Bioavailability of Lapatinib in Cancer Patients

Kevin M. Koch; Nandi J. Reddy; Roger B. Cohen; Nancy L. Lewis; Bonnie Whitehead; Kathleen Mackay; Andrew G. Stead; Andrew P. Beelen; Lionel D. Lewis

PURPOSE This study was conducted to characterize the effect of food on the relative bioavailability of lapatinib. PATIENTS AND METHODS A single 1,500-mg, oral dose of lapatinib was administered to 27 patients with advanced solid tumors on each of three occasions that were 1 week apart, in random order: after an overnight fast, with a low-fat breakfast, and with a high-fat breakfast. RESULTS The low-fat breakfast produced mean increases in lapatinib area under the concentration-time curve (AUC) of 167% (2.67-fold) and maximum concentration (C(max)) of 142% (2.42-fold). The high-fat breakfast produced mean increases in lapatinib AUC of 325% (4.25-fold) and C(max) of 203% (3.03-fold) compared with the fasted state. Increased bioavailability in the fed state did not significantly decrease relative variability. Therefore, absolute variability in systemic exposure was increased. CONCLUSION These large increases in lapatinib bioavailability and absolute variability support the recommendation for dosing in the fasted state to achieve consistent therapeutic exposure. Prescribers and patients should consider the potential consequences of toxicity or diminished efficacy that might result from dosing without regard to variations in diet.


Journal of Clinical Oncology | 2009

Phase I Study of the Safety, Tolerability, and Pharmacokinetics of Oral CP-868,596, a Highly Specific Platelet-Derived Growth Factor Receptor Tyrosine Kinase Inhibitor in Patients With Advanced Cancers

Nancy L. Lewis; Lionel D. Lewis; Joseph Paul Eder; Nandi J. Reddy; Feng Guo; Kristen J. Pierce; Anthony J. Olszanski; Roger B. Cohen

PURPOSE This phase I, first-in-human study evaluated the safety, tolerability, pharmacokinetics, and maximum-tolerated dose (MTD) of an oral platelet-derived growth factor receptor inhibitor, CP-868,596. PATIENTS AND METHODS Patients with advanced solid tumors were eligible. Dose escalations were performed in three groups with two formulations: uncoated on an empty stomach (UES), uncoated with food (UFED), and film-coated (FC) without food. Initial dose escalation in the UES group was followed by parallel escalations in the UFED and FC groups. RESULTS Fifty-nine patients enrolled. CP-868,596 was escalated from 100 mg to 340 mg daily in the UES group, from 60 mg to 100 mg twice daily in the UFED group, and from 100 mg once daily to 140 mg twice daily in the FC group. MTDs were 200 mg daily in the UES group and 100 mg twice daily in the FC group; MTD was not reached at 100 mg twice daily in the UFED group. Dose-limiting toxicities included hematuria, increased gamma-glutamyltransferase or ALT, insomnia, and nausea/vomiting. Most treatment-related AEs were of grades 1 to 2 severity; nausea, vomiting, and diarrhea were reported most frequently. Administration with food generally improved tolerability. CP-868,596 was absorbed slowly; systemic exposure parameters appeared to increase greater than proportionally with dose. Mean serum concentrations exceeded the preclinically predicted minimal efficacious concentration (ie, 16 ng/mL) at all dosages. Food and film coating apparently increased interpatient variability of the maximum observed plasma concentration and the area under the concentration-time curve. No objective responses were reported, and eight patients achieved stable disease (mean duration, 5.7 months). CONCLUSION CP-868,596 potentially demonstrated greater than dose-proportional pharmacokinetics. The recommended dosage of 100 mg twice daily with food was well tolerated. Additional development as a single agent in selected populations or in combination with chemotherapy in broader populations is warranted.


Annals of Oncology | 2016

Ramucirumab combined with FOLFOX as front-line therapy for advanced esophageal, gastroesophageal junction, or gastric adenocarcinoma: a randomized, double-blind, multicenter Phase II trial

H. H. Yoon; Johanna C. Bendell; F. S. Braiteh; I. Firdaus; Philip A. Philip; Allen Lee Cohn; Nancy L. Lewis; D. M. Anderson; E. Arrowsmith; Jonathan D. Schwartz; L. Gao; Yanzhi Hsu; Y. Xu; David Ferry; Steven R. Alberts; Zev A. Wainberg

BACKGROUND We report the first randomized, Phase II trial of ramucirumab, an anti-vascular endothelial growth factor receptor-2 monoclonal antibody, as front-line therapy in patients with advanced adenocarcinoma of the esophagus or gastric/gastroesophageal junction (GEJ). PATIENTS AND METHODS Patients from the USA with advanced esophageal, gastric, or GEJ adenocarcinoma randomly received (1:1) mFOLFOX6 plus ramucirumab (8 mg/kg) or mFOLFOX6 plus placebo every 2 weeks. The primary end point was progression-free survival (PFS) with 80% power to detect a hazard ratio (HR) of 0.71 (one-sided α = 0.15). Secondary end points included evaluation of response and overall survival (OS); an exploratory ramucirumab exposure-response analysis was undertaken. RESULTS Of 168 randomized patients, 52% of tumors were located in the stomach/GEJ and 48% in the esophagus. The trial did not meet the primary end point of PFS [6.4 versus 6.7 months, HR 0.98 (95% confidence interval 0.69-1.37)] or the secondary end point of OS (11.7 versus 11.5 months) in the intent-to-treat (ITT) population. Objective response rates (45.2% versus 46.4%) were similar between arms. Most Grade ≥3 toxicities did not differ significantly between arms, yet premature discontinuation of FOLFOX and ramucirumab (for reasons other than progressive disease) was more common among ramucirumab- versus placebo-treated patients. In an exploratory analysis that censored for premature discontinuation, the HR for PFS favored the ramucirumab arm (HR 0.76), particularly in patients with gastric/GEJ cancer. An exploratory exposure-response analysis indicated that patients with higher ramucirumab exposure had longer OS. CONCLUSION The addition of ramucirumab to front-line mFOLFOX6 did not improve PFS in the ITT population. CLINICALTRIALSGOV IDENTIFIER NCT01246960.


Annals of Oncology | 2012

Multicenter phase II study of trabectedin in patients with metastatic castration-resistant prostate cancer

M. D. Michaelson; Joaquim Bellmunt; Gary R. Hudes; Shom Goel; R. J. Lee; Philip W. Kantoff; C. A. Stein; P. Lardelli; I. Pardos; C. Kahatt; Antonio Nieto; M. Cullell-Young; Nancy L. Lewis; Matthew R. Smith

BACKGROUND This multicenter phase II trial evaluated the efficacy and safety of trabectedin in metastatic castration-resistant prostate cancer (CRPC). PATIENTS AND METHODS Two schedules were evaluated in three cohorts: weekly as 3-h i.v. infusion at 0.58 mg/m(2) for 3 out of 4 weeks (Cohort A, n = 33), and every 3 weeks (q3wk) as 24-h infusion at 1.5 mg/m(2) (Cohort B1, n = 5) and 1.2 mg/m(2) (Cohort B2, n = 20). The primary end point was prostate-specific antigen (PSA) response; secondary end points included safety, tolerability and time to progression (TTP). RESULTS Trabectedin resulted in PSA declines ≥ 50% in 12.5% (Cohort A) and 10.5% (Cohort B2) of patients. Among men pretreated with taxane-based chemotherapy, PSA response was 13.6% (Cohort A) and 15.4% (Cohort B2). PSA responses lasted 4.1-8.6 months, and median TTP was 1.5 months (Cohort A) and 1.9 months (Cohort B2). The dose of 1.5 mg/m(2) (approved for soft tissue sarcoma) given as 24-h infusion q3wk was not tolerable in these patients. At 1.2 mg/m(2) q3wk and 0.58 mg/m(2) weekly, the most common adverse events were nausea, fatigue and transient neutropenia and transaminase increase. CONCLUSIONS Two different trabectedin schedules showed modest activity in metastatic CRPC. Further studies may require identification of predictive factors of response in prostate cancer.BACKGROUND This multicenter phase II trial evaluated the efficacy and safety of trabectedin in metastatic castration-resistant prostate cancer (CRPC). PATIENTS AND METHODS Two schedules were evaluated in three cohorts: weekly as 3-h i.v. infusion at 0.58 mg/m2 for 3 out of 4 weeks (Cohort A, n = 33), and every 3 weeks (q3wk) as 24-h infusion at 1.5 mg/m2 (Cohort B1, n = 5) and 1.2 mg/m2 (Cohort B2, n = 20). The primary end point was prostate-specific antigen (PSA) response; secondary end points included safety, tolerability and time to progression (TTP). RESULTS Trabectedin resulted in PSA declines ≥50% in 12.5% (Cohort A) and 10.5% (Cohort B2) of patients. Among men pretreated with taxane-based chemotherapy, PSA response was 13.6% (Cohort A) and 15.4% (Cohort B2). PSA responses lasted 4.1-8.6 months, and median TTP was 1.5 months (Cohort A) and 1.9 months (Cohort B2). The dose of 1.5 mg/m2 (approved for soft tissue sarcoma) given as 24-h infusion q3wk was not tolerable in these patients. At 1.2 mg/m2 q3wk and 0.58 mg/m2 weekly, the most common adverse events were nausea, fatigue and transient neutropenia and transaminase increase. CONCLUSIONS Two different trabectedin schedules showed modest activity in metastatic CRPC. Further studies may require identification of predictive factors of response in prostate cancer.


Oncologist | 2016

Phase I Study of the Prolactin Receptor Antagonist LFA102 in Metastatic Breast and Castration-Resistant Prostate Cancer

Neeraj Agarwal; Jean-Pascal Machiels; Cristina Suárez; Nancy L. Lewis; Michaela J. Higgins; Kari Wisinski; Ahmad Awada; Michela Maur; Mark N. Stein; Andy Hwang; Rebecca Mosher; Ernesto Wasserman; Gang Wu; Hefei Zhang; Renata Zieba; Mohamed Elmeliegy

Lessons Learned Despite evidence for a role for prolactin signaling in breast and prostate tumorigenesis, a prolactin receptor-binding monoclonal antibody has not produced clinical efficacy. Increased serum prolactin levels may be a biomarker for prolactin receptor inhibition. Results from the pharmacokinetic and pharmacodynamics (PD) studies suggest that inappropriately long dosing intervals and insufficient exposure to LFA102 may have resulted in lack of antitumor efficacy. Based on preclinical data, combination therapy of LFA102 with those novel agents targeting hormonal pathways in metastatic castration-resistant prostate cancer and metastatic breast cancer is promising. Given the PD evidence of prolactin receptor blockade by LFA102, this drug has the potential to be used in conditions such as hyperprolactinemia that are associated with high prolactin levels. Background. Prolactin receptor (PRLR) signaling is implicated in breast and prostate cancer. LFA102, a humanized monoclonal antibody (mAb) that binds to and inhibits the PRLR, has exhibited promising preclinical antitumor activity. Methods. Patients with PRLR-positive metastatic breast cancer (MBC) or metastatic castration-resistant prostate cancer (mCRPC) received doses of LFA102 at 3–60 mg/kg intravenously once every 4 weeks. Objectives were to determine the maximum tolerated dose (MTD) and/or recommended dose for expansion (RDE) to investigate the safety/tolerability of LFA102 and to assess pharmacokinetics (PK), pharmacodynamics (PD), and antitumor activity. Results. A total of 73 patients were enrolled at 5 dose levels. The MTD was not reached because of lack of dose-limiting toxicities. The RDE was established at 60 mg/kg based on PK and PD analysis and safety data. The most common all-cause adverse events (AEs) were fatigue (44%) and nausea (33%) regardless of relationship. Grade 3/4 AEs reported to be related to LFA102 occurred in 4% of patients. LFA102 exposure increased approximately dose proportionally across the doses tested. Serum prolactin levels increased in response to LFA102 administration, suggesting its potential as a biomarker for PRLR inhibition. No antitumor activity was detected. Conclusion. Treatment with LFA102 was safe and well tolerated, but did not show antitumor activity as monotherapy at the doses tested.


Archives of Physical Medicine and Rehabilitation | 2009

The Effect of Stretching on Muscle Responses and Postural Sway Responses During Computerized Dynamic Posturography in Women and Men

Nancy L. Lewis; Jean-Michel Brismée; C. Roger James; Phillip S. Sizer; Steven F. Sawyer

OBJECTIVE To evaluate the effect of stretching on the parameters of postural sway and on the kinematic variables associated with balance control in women and men. DESIGN Mixed repeated measures design with 2 levels. SETTING Research laboratory. PARTICIPANTS Fifteen women and fifteen men (mean age 23.4+/-2.2). INTERVENTION Two separate sessions of (1) lower extremity stretching and (2) no-stretching, immediately prior to balance testing with simultaneous surface electromyographic (EMG) recordings of muscle responses. MAIN OUTCOME MEASURES EMG latencies and average amplitudes for 4 lower extremity muscles for the preferred stance limb during computerized dynamic posturography (CDP) tests, specifically the Postural Evoked Response Test, Adaptation Test, Motor Control Test, Sensory Organization Test, and Unilateral Stance Test. RESULTS Analyses of variance indicated no significant main effect for stretching and 2 significant main effects for gender for the Motor Control Test (P=.021) and latency of tibialis anterior (P=.009). Analyses of covariance with covariants of height and active knee extension revealed no significant main effect of stretching or of gender on muscles responses or CDP performance. CONCLUSIONS In both women and men, lower extremity stretching did not significantly affect muscle responses or performance during CDP.


American Journal of Clinical Oncology | 2008

Phase I study of capecitabine and oxaliplatin in combination with the proteasome inhibitor bortezomib in patients with advanced solid tumors.

Steven J. Cohen; Paul F. Engstrom; Nancy L. Lewis; Corey J. Langer; Susan McLaughlin; Mary Beard; Louis M. Weiner; Neal J. Meropol

Purpose:The combination of capecitabine and oxaliplatin has clinical benefit in a variety of gastrointestinal malignancies. The proteasome inhibitor bortezomib enhances the cytotoxic activity of fluoropyrimidines and platinum agents in vivo, and targeting of NF-κB may overcome chemotherapy resistance. Thus, we performed this phase I study to document the safety and obtain preliminary efficacy data for the combination of capecitabine, oxaliplatin, and bortezomib. Patients and Methods:Patients with advanced solid tumors were treated with oxaliplatin 130 mg/m2 intravenously on day 1, capecitabine 750–900 mg/m2 twice daily orally for 14 days, and bortezomib 1.0, 1.3, or 1.6 mg/m2 intravenously on days 1 and 8 of 21 day cycles. CT scans were repeated every 2 cycles. Results:Thirteen patients received 45 cycles of treatment (median, 2; range, 1–8). No dose-limiting toxicities were noted at all bortezomib dose levels when administered with full dose capecitabine and oxaliplatin. The most common grade 3 nonhematologic toxicities during any cycle of therapy included elevated transaminases (3), vomiting, diarrhea, and dehydration (2 each). Only one patient experienced grade 3 peripheral neuropathy in cycle 8. Three objective tumor responses were noted (squamous cell of anus, adenocarcinoma of unknown primary, adenocarcinoma of rectum). Conclusions:Weekly bortezomib can be safely combined with full doses of capecitabine and oxaliplatin. As 1.6 mg/m2 weekly of bortezomib is the maximum tolerated dose in single-agent studies, no further dose escalation was performed in this study. Preliminary evidence of antitumor activity is demonstrated. The further evaluation of this combination in diseases for which capecitabine and oxaliplatin have efficacy should be considered.


Cancer Investigation | 2000

Oral fluoropyrimidines in cancer treatment.

Nancy L. Lewis; Neal J. Meropol

Since its introduction more than 40 years ago, S-fluorouracil(5-FU) has become a component of the standard therapy for a variety of malignancies, including gastrointestinal cancers, breast cancer, and head and neck cancer. Insights into the mechanisms of 5-FU activity have led to strategies involving biochemical modulation with agents such as leucovorin (LV) (1,2) and altering dosing schedules to improve therapeutic index (3). Current efforts include the development of oral fluoropyrimidines, which have the added benefit of ease of administration, patient preference (4), and perhaps lower cost. These oral strategies must overcome the erratic absorption of 5-FU ( 5 ) . Four new oral fluoropyrimidine-based therapies have shown promise in the treatment of various malignancies. Capecitabine (XelodaTM, Hoffman-LaRoche), a fluoropyrimidine carbamate that is ultimately converted in vivo to 5-FU, is approved by the U.S. Food and Drug Administration for patients with refractory breast cancer and has shown activity in colorectal cancer. UFT (Taiho Pharmaceuticals), an oral combination of the 5-FU prodrug ftorafur (tegafur) and uracil (a naturally occurring pyrimidine that serves to competitively inhibit 5-FU degradation), has been studied alone and in combination with LV (OrzelTM, Bristol-Myers/Squibb) in colorectal, gastric, breast, and head and neck cancers. Eniluracil (ethynyluracil, 776C85, GlaxoWellcome) is a potent irreversible in-

Collaboration


Dive into the Nancy L. Lewis's collaboration.

Top Co-Authors

Avatar

Edith P. Mitchell

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ashwin Reddy Sama

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Neal J. Meropol

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Charles J. Yeo

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Roger B. Cohen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Benjamin E. Leiby

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Harish Lavu

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Jordan M. Winter

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge