Network


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

Hotspot


Dive into the research topics where Gayle S. Jameson is active.

Publication


Featured researches published by Gayle S. Jameson.


The Lancet | 2016

Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial

Andrea Wang-Gillam; Chung-Pin Li; G. Bodoky; Andrew Peter Dean; Yan Shen Shan; Gayle S. Jameson; Teresa Macarulla; Kyung-Hun Lee; David Cunningham; Jean Frédéric Blanc; Richard Hubner; Chang Fang Chiu; Gilberto Schwartsmann; Jens T. Siveke; Fadi S. Braiteh; Victor Moyo; Bruce Belanger; Navreet Dhindsa; Eliel Bayever; Daniel D. Von Hoff; Li-Tzong Chen

BACKGROUND Nanoliposomal irinotecan showed activity in a phase 2 study in patients with metastatic pancreatic ductal adenocarcinoma previously treated with gemcitabine-based therapies. We assessed the effect of nanoliposomal irinotecan alone or combined with fluorouracil and folinic acid in a phase 3 trial in this population. METHODS We did a global, phase 3, randomised, open-label trial at 76 sites in 14 countries. Eligible patients with metastatic pancreatic ductal adenocarcinoma previously treated with gemcitabine-based therapy were randomly assigned (1:1) using an interactive web response system at a central location to receive either nanoliposomal irinotecan monotherapy (120 mg/m(2) every 3 weeks, equivalent to 100 mg/m(2) of irinotecan base) or fluorouracil and folinic acid. A third arm consisting of nanoliposomal irinotecan (80 mg/m(2), equivalent to 70 mg/m(2) of irinotecan base) with fluorouracil and folinic acid every 2 weeks was added later (1:1:1), in a protocol amendment. Randomisation was stratified by baseline albumin, Karnofsky performance status, and ethnic origin. Treatment was continued until disease progression or intolerable toxic effects. The primary endpoint was overall survival, assessed in the intention-to-treat population. The primary analysis was planned after 305 events. Safety was assessed in all patients who had received study drug. This trial is registered at ClinicalTrials.gov, number NCT01494506. FINDINGS Between Jan 11, 2012, and Sept 11, 2013, 417 patients were randomly assigned either nanoliposomal irinotecan plus fluorouracil and folinic acid (n=117), nanoliposomal irinotecan monotherapy (n=151), or fluorouracil and folinic acid (n=149). After 313 events, median overall survival in patients assigned nanoliposomal irinotecan plus fluorouracil and folinic acid was 6.1 months (95% CI 4.8-8.9) vs 4.2 months (3.3-5.3) with fluorouracil and folinic acid (hazard ratio 0.67, 95% CI 0.49-0.92; p=0.012). Median overall survival did not differ between patients assigned nanoliposomal irinotecan monotherapy and those allocated fluorouracil and folinic acid (4.9 months [4.2-5.6] vs 4.2 months [3.6-4.9]; 0.99, 0.77-1.28; p=0.94). The grade 3 or 4 adverse events that occurred most frequently in the 117 patients assigned nanoliposomal irinotecan plus fluorouracil and folinic acid were neutropenia (32 [27%]), diarrhoea (15 [13%]), vomiting (13 [11%]), and fatigue (16 [14%]). INTERPRETATION Nanoliposomal irinotecan in combination with fluorouracil and folinic acid extends survival with a manageable safety profile in patients with metastatic pancreatic ductal adenocarcinoma who previously received gemcitabine-based therapy. This agent represents a new treatment option for this population. FUNDING Merrimack Pharmaceuticals.


Clinical Cancer Research | 2013

A Multicenter, Phase I, Dose-Escalation Study to Assess the Safety, Tolerability, and Pharmacokinetics of Etirinotecan Pegol in Patients with Refractory Solid Tumors

Gayle S. Jameson; John T. Hamm; Glen J. Weiss; Carlos Alemany; Stephen P. Anthony; Michele Basche; Ramesh K. Ramanathan; Mitesh J. Borad; Raoul Tibes; Allen Lee Cohn; Ioana Hinshaw; Robert M. Jotte; Lee S. Rosen; Ute Hoch; Michael A. Eldon; Robert A. Medve; Katrina Schroeder; Erica White; Daniel D. Von Hoff

Purpose: This study was designed to establish the maximum tolerated dose (MTD) and to evaluate tolerability, pharmacokinetics, and antitumor activity of etirinotecan pegol. Experimental Design: Patients with refractory solid malignancies were enrolled and assigned to escalating-dose cohorts. Patients received 1 infusion of etirinotecan pegol weekly 3 times every 4 weeks (w × 3q4w), or every 14 days (q14d), or every 21 days (q21d), with MTD as the primary end point using a standard 3 + 3 design. Results: Seventy-six patients were entered onto 3 dosing schedules (58–245 mg/m2). The MTD was 115 mg/m2 for the w × 3q4w schedule and 145 mg/m2 for both the q14d and q21d schedules. Most adverse events related to study drug were gastrointestinal disorders and were more frequent at higher doses of etirinotecan pegol. Late onset diarrhea was observed in some patients, the frequency of which generally correlated with dose density. Cholinergic diarrhea commonly seen with irinotecan treatment did not occur in patients treated with etirinotecan pegol. Etirinotecan pegol administration resulted in sustained and controlled systemic exposure to SN-38, which had a mean half-life of approximately 50 days. Overall, the pharmacokinetics of etirinotecan pegol are predictable and do not require complex dosing adjustments. Confirmed partial responses were observed in 8 patients with breast, colon, lung (small and squamous cell), bladder, cervical, and neuroendocrine cancer. Conclusion: Etirinotecan pegol showed substantial antitumor activity in patients with various solid tumors and a somewhat different safety profile compared with the irinotecan historical profile. The MTD recommended for phase II clinical trials is 145 mg/m2 q14d or q21d. Clin Cancer Res; 19(1); 268–78. ©2012 AACR.


Investigational New Drugs | 2009

Blackberry-induced hand-foot skin reaction to sunitinib

Susan L. Boone; Gayle S. Jameson; Daniel D. Von Hoff; Mario E. Lacouture

SummarySunitinib is an orally administered small molecule that was approved by the US Food and Drug Administration in January 2006 as monotherapy for the treatment of patients with advanced renal cell carcinoma (RCC) and patients with gastrointestinal stromal tumor after disease progression on or intolerance to imatinib mesylate. Data pooled from multiple sunitinib dose-escalation trials showed that sunitinib is associated with various adverse events, with HFSR occurring in up to 20% of patients. We describe a 48-year-old woman with a history of metastatic colorectal cancer treated with single-agent sunitinib who developed pain and tenderness in areas of friction secondary to Blackberry use, and was diagnosed with trauma-induced hand-foot skin reaction (HFSR) secondary to sunitinib therapy.


Clinical Cancer Research | 2017

Correlation between Ferumoxytol Uptake in Tumor Lesions by MRI and Response to Nanoliposomal Irinotecan in Patients with Advanced Solid Tumors: A Pilot Study

Ramesh K. Ramanathan; Ronald L. Korn; Natarajan Raghunand; Jasgit C. Sachdev; Ronald G. Newbold; Gayle S. Jameson; Gerald J. Fetterly; Joshua Prey; Stephan Klinz; Jaeyeon Kim; Jason E. Cain; Bart S. Hendriks; Daryl C. Drummond; Eliel Bayever; Jonathan Fitzgerald

Purpose: To determine whether deposition characteristics of ferumoxytol (FMX) iron nanoparticles in tumors, identified by quantitative MRI, may predict tumor lesion response to nanoliposomal irinotecan (nal-IRI). Experimental Design: Eligible patients with previously treated solid tumors had FMX-MRI scans before and following (1, 24, and 72 hours) FMX injection. After MRI acquisition, R2* signal was used to calculate FMX levels in plasma, reference tissue, and tumor lesions by comparison with a phantom-based standard curve. Patients then received nal-IRI (70 mg/m2 free base strength) biweekly until progression. Two percutaneous core biopsies were collected from selected tumor lesions 72 hours after FMX or nal-IRI. Results: Iron particle levels were quantified by FMX-MRI in plasma, reference tissues, and tumor lesions in 13 of 15 eligible patients. On the basis of a mechanistic pharmacokinetic model, tissue permeability to FMX correlated with early FMX-MRI signals at 1 and 24 hours, while FMX tissue binding contributed at 72 hours. Higher FMX levels (ranked relative to median value of multiple evaluable lesions from 9 patients) were significantly associated with reduction in lesion size by RECIST v1.1 at early time points (P < 0.001 at 1 hour and P < 0.003 at 24 hours FMX-MRI, one-way ANOVA). No association was observed with post-FMX levels at 72 hours. Irinotecan drug levels in lesions correlated with patients time on treatment (Spearman ρ = 0.7824; P = 0.0016). Conclusions: Correlation between FMX levels in tumor lesions and nal-IRI activity suggests that lesion permeability to FMX and subsequent tumor uptake may be a useful noninvasive and predictive biomarker for nal-IRI response in patients with solid tumors. Clin Cancer Res; 23(14); 3638–48. ©2017 AACR.


Journal of Cancer | 2012

The impact of concomitant medication use on patient eligibility for phase I cancer clinical trials.

Mitesh J. Borad; Kelly K. Curtis; Hani M. Babiker; Martin Benjamin; Raoul Tibes; Ramesh K. Ramanathan; Karen Wright; Amylou C. Dueck; Gayle S. Jameson; Daniel D. Von Hoff

Concomitant medication (CM) use may result in Phase I cancer clinical trial ineligibility due to concern for potential CM-investigational drug interactions or alteration of investigational drug absorption. Few studies have examined the impact of CM use on trial eligibility. Methods: We reviewed records of 274 patients on Phase I trials at a single academic institution. Demographics, CM identities and classes, CM discontinuation, reasons, and incidence of CM substitution were recorded. CM-investigational drug cytochrome P450 (CYP) enzyme interactions were documented. Statistical analysis was performed using descriptive statistics. Results: 273 of 274 patients (99.6%, 95% confidence interval [CI] 98.9-100%) took CM, with a median of 8 CM per patient (range 0 - 42). CM discontinuation occurred in 67 cases (25%, 95% CI 19-30%). The most common CM classes discontinued were herbal (17 cases, 25%, 95% CI 16-37%) and proton pump inhibitors (15 cases, 22%, 95% CI 12-32%). CM discontinuation reasons were: protocol prohibition (32 cases, 48%, 95% CI 36-60%); potential CM-investigational drug interaction (25 cases, 37%, 95% CI 26-49%); other (10 cases, 15%, 95% CI 6-23%). A potential CM-investigational drug CYP interaction was noted in 122 cases (45%, 95% CI 39-50%). CM potentially weakly decreased investigational drug metabolism in 52 cases (43%, 95% CI 34-51%), and potentially strongly decreased investigational drug metabolism in 17 cases (14%, 95% CI 8-20%). Investigational drug potentially weakly decreased CM metabolism in 39 cases (32%, 95% CI 24-40%), and potentially strongly decreased CM metabolism in 28 cases (23%, 95% CI 15-30%). CM substitution occurred in 36/67 cases (54%, 95% CI 41-66%) where CM were discontinued to allow for eventual participation in clinical trials. Overall in 2 cases (0.7%, 95% CI 0.1-2.6%), patients were protocol ineligible because CM could not be discontinued or substituted. Conclusions: This study highlights the high prevalence of concomitant medication use among cancer patients enrolled in phase I clinical trials. Most patients did meet trial eligibility criteria with careful substitution and discontinuation of CM. The most common reason for discontinuation of CM was protocol prohibition. The most common medications discontinued were herbal, proton pump inhibitors, selective serotonin reuptake inhibitor anti-depressants, and non-steroidal anti-inflammatory drugs.


The Lancet Gastroenterology & Hepatology | 2017

Safety and tolerability of veliparib combined with capecitabine plus radiotherapy in patients with locally advanced rectal cancer: a phase 1b study

Brian G. Czito; Dustin A. Deming; Gayle S. Jameson; Mary F. Mulcahy; Houman Vaghefi; Matthew W. Dudley; Kyle D. Holen; Angela DeLuca; Rajendar K. Mittapalli; Wijith Munasinghe; Lei He; John Zalcberg; S. Ngan; Philip Komarnitsky; Michael Michael

BACKGROUND Further optimisation of present standard chemoradiation is needed in patients with locally advanced rectal cancer. Veliparib, an oral poly(ADP-ribose) polymerase inhibitor, has been shown to enhance the antitumour activity of chemotherapy and radiotherapy in preclinical models. We aimed to establish the maximum tolerated dose and establish the recommended phase 2 dose of veliparib combined with neoadjuvant capecitabine and radiotherapy. METHODS This phase 1b, open-label, multicentre, dose-escalation study was done at six hospitals (one in Australia and five in the USA). Patients were eligible if they were aged 18 years or more and were newly diagnosed with stage II to III locally advanced, resectable adenocarcinoma of the rectum with a distal tumour border of less than 12 cm from anal verge. Patients were ineligible if they had received anticancer therapy or surgery (except colostomy or ileostomy) 28 days or less before the first dose of study drug, previous pelvic radiotherapy, or previous treatment with poly (ADP-ribose) polymerase inhibitors. Enrolled patients received capecitabine (825 mg/m2 orally twice daily) with radiotherapy (50·4 Gy in 1·8 Gy fractions daily, approximately 5 days consecutively per week for about 5·5 weeks). Veliparib (20-400 mg orally twice daily) was administered daily starting on day 2 of week 1 and continuing until 2 days after radiotherapy completion. Patients underwent total mesorectal excision 5-10 weeks after radiotherapy completion. The primary objectives were to establish the maximum tolerated dose and recommended phase 2 dose of veliparib plus capecitabine and radiotherapy, with an exposure-adjusted continual reassessment methodology. Efficacy and safety analyses were done per protocol. The reported study has completed accrual and all analyses are final. This trial is registered with ClinicalTrials.gov, number NCT01589419. FINDINGS Between June 12, 2012, and Jan 13, 2015, 32 patients received veliparib (22 in the dose-escalation group; ten in the safety expansion group); 31 were assessable for efficacy (<400 mg, n=16; 400 mg, n=15). During dose escalation, grade 2 dose-limiting toxic effects occurred in two patients; no grade 3-4 dose-limiting toxic effects were noted. Therefore, the maximum tolerated dose was not reached; the recommended phase 2 dose was selected as 400 mg twice daily. The most common treatment-emergent adverse events in all 32 patients were nausea (17 [53%]), diarrhoea (16 [50%]), and fatigue (16 [50%]). Grade 3 diarrhoea was noted in three (9%) of 32 patients; no grade 4 events were reported. Veliparib pharmacokinetics were dose proportional, with no effect on capecitabine pharmacokinetics. Tumour downstaging after surgery was noted in 22 (71%) of 31 patients; nine (29%) of 31 patients achieved a pathological complete response. INTERPRETATION Veliparib plus capecitabine and radiotherapy had an acceptable safety profile and showed a dose-proportional pharmacokinetic profile with no effect on the pharmacokinetics of capecitabine. Preliminary antitumour activity warrants further evaluation. FUNDING AbbVie Inc.


Dermatology Reports | 2011

Long-term safety, tolerability, and efficacy of vismodegib in two patients with metastatic basal cell carcinoma and basal cell nevus syndrome

Glen J. Weiss; Raoul Tibes; Lisa Blaydorn; Gayle S. Jameson; Molly Downhour; Erica White; Ivor Caro; Daniel D. Von Hoff

Tumor responses in advanced basal cell carcinoma (BCC) have been observed in clinical trials with vismodegib, a SMO antagonist. The result of SMO antagonism is inhibition Hedgehog Signaling Pathway (HHSP) downstream target genes. HHSP inhibition has been shown to affect stem cells responsible for blood, mammary, and neural development. We report on our experience of treating two patients with advanced BCC participating. These two patients have had no new BCCs develop for at least 2.25 years. Both patients have been receiving ongoing daily treatment with vismodegib for greater than 2.75 years without experiencing any significant side effects. After prolonged continuous daily dosing with a SMO antagonist, we have not observed a significant alteration in hematologic parameters or physical abnormalities of the pectoral regions of two patients with advanced BCC.


Case Reports in Gastroenterology | 2009

Marantic Endocarditis Associated with Pancreatic Cancer: A Case Series

Gayle S. Jameson; Ramesh K. Ramanathan; Mitesh J. Borad; Molly Downhour; Ronald L. Korn; Daniel D. Von Hoff

Marantic endocarditis, otherwise known as nonbacterial thrombotic endocarditis (NBTE), is a well-documented phenomenon due to hypercoagulability from an underlying cause. It has been associated with a variety of inflammatory states including malignancy. Surprisingly, although hypercoagulability is often seen in patients with pancreatic cancer, marantic endocarditis has rarely been reported antemortem in this population. We report three cases of marantic endocarditis in patients with advanced pancreatic cancer. In two instances, the patients’ neurological symptoms preceded the diagnosis of advanced pancreatic cancer. Health care professionals should be alert to the possibility of marantic endocarditis in any patient with cancer, especially pancreatic cancer, who presents with symptoms of neurological dysfunction or an arterial thrombotic event. Prompt diagnosis and treatment with heparin, unfractionated or low molecular weight, may prevent catastrophic CNS events and decrease morbidity in patients with pancreatic cancer and other malignancies.


British Journal of Cancer | 2017

Clinical study of genomic drivers in pancreatic ductal adenocarcinoma

Michael T. Barrett; Ray Deiotte; Elizabeth Lenkiewicz; Smriti Malasi; Tara Holley; Lisa Evers; Richard G. Posner; Timothy Jones; Haiyong Han; Mark Sausen; Victor E. Velculescu; Jeffrey A. Drebin; P. J. O'Dwyer; Gayle S. Jameson; Ramesh K. Ramanathan; Daniel D. Von Hoff

Background:Pancreatic ductal adenocarcinoma (PDA) is a lethal cancer with complex genomes and dense fibrotic stroma. This study was designed to identify clinically relevant somatic aberrations in pancreatic cancer genomes of patients with primary and metastatic disease enrolled and treated in two clinical trials.Methods:Tumour nuclei were flow sorted prior to whole genome copy number variant (CNV) analysis. Targeted or whole exome sequencing was performed on most samples. We profiled biopsies from 68 patients enrolled in two Stand Up to Cancer (SU2C)-sponsored clinical trials. These included 38 resected chemoradiation naïve tumours (SU2C 20206-003) and metastases from 30 patients who progressed on prior therapies (SU2C 20206-001). Patient outcomes including progression-free survival (PFS) and overall survival (OS) were observed.Results:We defined: (a) CDKN2A homozygous deletions that included the adjacent MTAP gene, only its’ 3′ region, or excluded MTAP; (b) SMAD4 homozygous deletions that included ME2; (c) a pancreas-specific MYC super-enhancer region; (d) DNA repair-deficient genomes; and (e) copy number aberrations present in PDA patients with long-term (⩾ 40 months) and short-term (⩽ 12 months) survival after surgical resection.Conclusions:We provide a clinically relevant framework for genomic drivers of PDA and for advancing novel treatments.


Molecular Cancer Therapeutics | 2011

Abstract A94: Dose determination of LY2603618, a Chk1 inhibitor, administered in combination with gemcitabine in patients with advanced cancer.

Emiliano Calvo; Donald A. Richards; F. Braiteh; Daniel D. Von Hoff; Robert R. McWilliams; Carlos Becerra; Matthew D. Galsky; Gayle S. Jameson; Karla Hurt; Scott M. Hynes; Ignacio Garcia-Ribas; Aimee K. Bence; Eric Westin

Background: LY2603618 is a selective inhibitor of Chk1, a protein kinase that plays a key role in the DNA damage checkpoint. Inhibition of Chk1 is predicted to enhance the effects of antimetabolites, such as gemcitabine. Methods: This study is a Phase 1–2 study in patients with solid tumors (Phase 1) and advanced pancreatic adenocarcinoma (Phase 2). In Phase 1, gemcitabine (1000 mg/m2) was administered on Days 1, 8, and 15 of a 28-day cycle. LY2603618 was administered on Days 2, 9, and 16. Patients were assessed for safety, tolerability, and dose-limiting toxicity (DLT). A recommended Phase 2 dose (RP2D) was determined based on safety, dose intensity, and pharmacokinetics (PK). Results: A total of 50 patients were enrolled. Patients were treated at 70 (n =3), 105 (n=3), 150 (n=7), 200 (n=11), 250 (n=6) mg/m2and at 2 additional flat-fixed dose cohorts of 200 (n=10) and 230 (n=10) mg. The most frequent AEs reported included fatigue, thrombocytopenia, anemia, nausea, neutropenia, and constipation, which are consistent with those reported with gemcitabine monotherapy. During escalation, DLTs included neutropenia, infusion-related reaction and thrombocytopenia, with thrombocytopenia being dose limiting. The maximum tolerated dose (MTD) was determined to be 200 mg/m2. The systemic exposure of LY2603618 increased in a dose-dependent manner and the LY2603618 systemic clearance was dose-independent across all doses on average. The mean LY2603618 half-life varied across doses but was consistent with a half-life (i.e., >10 hr and 21,000 ng hr/mL and Cmax > 2000 ng/mL) were achieved by all but one patient. Based on safety/tolerability, the ability to maintain dose intensity, and PK, a RP2D of 230 mg was selected. A total of 17 of 30 patients received more than 2 cycles of therapy. Conclusions: LY2603618 administered in combination with gemcitabine demonstrated an acceptable safety profile; the MTD for this regimen was defined at 200 mg/m2 in the originally designed study. However at a fixed dose of 230 mg, LY2603618 in combination with gemcitabine had an acceptable safety profile and the observed exposures exceed those required for biological effect in nonclinical models. This dose is being evaluated in the Phase 2 component of the study. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A94.

Collaboration


Dive into the Gayle S. Jameson's collaboration.

Top Co-Authors

Avatar

Daniel D. Von Hoff

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Glen J. Weiss

Cancer Treatment Centers of America

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Wang-Gillam

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Erkut Borazanci

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

David Cunningham

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. D. Von Hoff

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge