Network


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

Hotspot


Dive into the research topics where Katrina H. Smith is active.

Publication


Featured researches published by Katrina H. Smith.


Neuro-oncology | 2013

Phase 2 study of dose-intense temozolomide in recurrent glioblastoma

Andrew D. Norden; Glenn J. Lesser; Jan Drappatz; Keith L. Ligon; Samantha Hammond; Eudocia Q. Lee; David Reardon; Camilo E. Fadul; Scott R. Plotkin; Tracy T. Batchelor; Jay Jiguang Zhu; Rameen Beroukhim; Alona Muzikansky; Lisa Doherty; Debra C. LaFrankie; Katrina H. Smith; Vida Tafoya; Rosina T. Lis; Edward C. Stack; Myrna R. Rosenfeld; Patrick Y. Wen

BACKGROUND Among patients with glioblastoma (GBM) who progress on standard temozolomide, the optimal therapy is unknown. Resistance to temozolomide is partially mediated by O(6)-methylguanine-DNA methyltransferase (MGMT). Because MGMT may be depleted by prolonged temozolomide administration, dose-intense schedules may overcome resistance. METHODS This was a multicenter, phase 2, single-arm study of temozolomide (75-100 mg/m(2)/day) for 21 days of each 28-day cycle. Patients had GBM in first recurrence after standard therapy. The primary end point was 6-month progression-free survival (PFS6). RESULTS Fifty-eight participants were accrued, 3 of whom were ineligible for analysis; one withdrew before response assessment. There were 33 men (61%), with a median age of 57 years (range, 25-79 years) and a median Karnofsky performance score of 90 (range, 60-100). Of 47 patients with MGMT methylation results, 36 (65%) had methylated tumors. There were 7 (13%) partial responses, and PFS6 was only 11%. Response and PFS did not depend on MGMT status; MSH2, MLH1, or ERCC1 expression; the number of prior temozolomide cycles; or the time off temozolomide. Treatment was well tolerated, with limited grade 3 neutropenia (n = 2) or thrombocytopenia (n = 2). CONCLUSIONS Dose-intense temozolomide on this schedule is safe in recurrent GBM. However, efficacy is marginal and predictive biomarkers are needed.


Neuro-oncology | 2015

Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma

Eudocia Q. Lee; David A. Reardon; David Schiff; Jan Drappatz; Alona Muzikansky; Sean Grimm; Andrew D. Norden; Lakshmi Nayak; Rameen Beroukhim; Mikael L. Rinne; Andrew S. Chi; Tracy T. Batchelor; Kelly Hempfling; Christine McCluskey; Katrina H. Smith; Sarah C. Gaffey; Brendan Wrigley; Keith L. Ligon; Jeffrey Raizer; Patrick Y. Wen

BACKGROUND Panobinostat is a histone deacetylase inhibitor with antineoplastic and antiangiogenic effects in glioma that may work synergistically with bevacizumab. We conducted a multicenter phase II trial of panobinostat combined with bevacizumab in patients with recurrent high-grade glioma (HGG). METHODS Patients with recurrent HGG were treated with oral panobinostat 30 mg 3 times per week, every other week, in combination with bevacizumab 10 mg/kg every other week. The primary endpoint was a 6-month progression-fee survival (PFS6) rate for participants with recurrent glioblastoma (GBM). Patients with recurrent anaplastic glioma (AG) were evaluated as an exploratory arm of the study. RESULTS At interim analysis, the GBM arm did not meet criteria for continued accrual, and the GBM arm was closed. A total of 24 patients with GBM were accrued prior to closure. The PFS6 rate was 30.4% (95%, CI 12.4%-50.7%), median PFS was 5 months (range, 3-9 months), and median overall survival (OS) was 9 months (range, 6-19 months). Accrual in the AG arm continued to completion, and a total of 15 patients were enrolled. The PFS6 rate was 46.7% (range, 21%-73%), median PFS was 7 months (range, 2-10 months), and median OS was 17 months (range, 5 months-27 months). CONCLUSIONS This phase II study of panobinostat and bevacizumab in participants with recurrent GBM did not meet criteria for continued accrual, and the GBM cohort of the study was closed. Although it was reasonably well tolerated, the addition of panobinostat to bevacizumab did not significantly improve PFS6 compared with historical controls of bevacizumab monotherapy in either cohort.


Neurology | 2015

Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma

Andrew D. Norden; Keith L. Ligon; Samantha Hammond; Alona Muzikansky; David A. Reardon; Thomas Kaley; Tracy T. Batchelor; Scott R. Plotkin; Jeffrey Raizer; Eric T. Wong; Jan Drappatz; Glenn J. Lesser; Sam Haidar; Rameen Beroukhim; Eudocia Q. Lee; Lisa Doherty; Debra C. LaFrankie; Sarah C. Gaffey; Mary Gerard; Katrina H. Smith; Christine McCluskey; Surasak Phuphanich; Patrick Y. Wen

Objective: A subset of meningiomas recur after surgery and radiation therapy, but no medical therapy for recurrent meningioma has proven effective. Methods: Pasireotide LAR is a long-acting somatostatin analog that may inhibit meningioma growth. This was a phase II trial in patients with histologically confirmed recurrent or progressive meningioma designed to evaluate whether pasireotide LAR prolongs progression-free survival at 6 months (PFS6). Patients were stratified by histology (atypical [World Health Organization grade 2] and malignant [grade 3] meningiomas in cohort A and benign [grade 3] in cohort B). Results: Eighteen patients were accrued in cohort A and 16 in cohort B. Cohort A had median age 59 years, median Karnofsky performance status 80, 17 (94%) had previous radiation therapy, and 11 (61%) showed high octreotide uptake. Cohort B had median age 52 years, median Karnofsky performance status 90, 11 (69%) had previous radiation therapy, and 12 (75%) showed high octreotide uptake. There were no radiographic responses to pasireotide LAR therapy in either cohort. Twelve patients (67%) in cohort A and 13 (81%) in cohort B achieved stable disease. In cohort A, PFS6 was 17% and median PFS 15 weeks (95% confidence interval: 8–20). In cohort B, PFS6 was 50% and median PFS 26 weeks (12–43). Treatment was well tolerated. Octreotide uptake and insulin-like growth factor–1 levels did not predict outcome. Expression of somatostatin receptor 3 predicted favorable PFS and overall survival. Conclusions: Pasireotide LAR has limited activity in recurrent meningiomas. The finding that somatostatin receptor 3 is associated with favorable outcomes warrants further investigation. Classification of evidence: This study provides Class IV evidence that in patients with recurrent or progressive meningioma, pasireotide LAR does not significantly increase the proportion of patients with PFS at 6 months.


Clinical Cancer Research | 2015

A Multicenter, Phase II, Randomized, Noncomparative Clinical Trial of Radiation and Temozolomide with or without Vandetanib in Newly Diagnosed Glioblastoma Patients

Eudocia Q. Lee; Thomas Kaley; Dan G. Duda; David Schiff; Andrew B. Lassman; Eric T. Wong; Tom Mikkelsen; Benjamin Purow; Alona Muzikansky; Marek Ancukiewicz; Jason T. Huse; Shakti Ramkissoon; Jan Drappatz; Andrew D. Norden; Rameen Beroukhim; Stephanie E. Weiss; Brian M. Alexander; Christine McCluskey; Mary Gerard; Katrina H. Smith; Rakesh K. Jain; Tracy T. Batchelor; Keith L. Ligon; Patrick Y. Wen

Purpose: Vandetanib, a tyrosine kinase inhibitor of KDR (VEGFR2), EGFR, and RET, may enhance sensitivity to chemotherapy and radiation. We conducted a randomized, noncomparative, phase II study of radiation (RT) and temozolomide with or without vandetanib in patients with newly diagnosed glioblastoma (GBM). Experimental Design: We planned to randomize a total of 114 newly diagnosed GBM patients in a ratio of 2:1 to standard RT and temozolomide with (76 patients) or without (38 patients) vandetanib 100 mg daily. Patients with age ≥ 18 years, Karnofsky performance status (KPS) ≥ 60, and not on enzyme-inducing antiepileptics were eligible. Primary endpoint was median overall survival (OS) from the date of randomization. Secondary endpoints included median progression-free survival (PFS), 12-month PFS, and safety. Correlative studies included pharmacokinetics as well as tissue and serum biomarker analysis. Results: The study was terminated early for futility based on the results of an interim analysis. We enrolled 106 patients (36 in the RT/temozolomide arm and 70 in the vandetanib/RT/temozolomide arm). Median OS was 15.9 months [95% confidence interval (CI), 11.0–22.5 months] in the RT/temozolomide arm and 16.6 months (95% CI, 14.9–20.1 months) in the vandetanib/RT/temozolomide (log-rank P = 0.75). Conclusions: The addition of vandetanib at a dose of 100 mg daily to standard chemoradiation in patients with newly diagnosed GBM or gliosarcoma was associated with potential pharmacodynamic biomarker changes and was reasonably well tolerated. However, the regimen did not significantly prolong OS compared with the parallel control arm, leading to early termination of the study. Clin Cancer Res; 21(16); 3610–8. ©2015 AACR.


Clinical Cancer Research | 2018

Phase I and biomarker study of plerixafor and bevacizumab in recurrent high-grade glioma

Eudocia Q. Lee; Dan G. Duda; Alona Muzikansky; Elizabeth R. Gerstner; John G. Kuhn; David A. Reardon; Lakshmi Nayak; Andrew D. Norden; Lisa Doherty; Debra C. LaFrankie; Jennifer Stefanik; Trupti Vardam; Katrina H. Smith; Christine McCluskey; Sarah C. Gaffey; Tracy T. Batchelor; Rakesh K. Jain; Patrick Y. Wen

Purpose: Although antiangiogenic therapy for high-grade glioma (HGG) is promising, responses are not durable. Correlative clinical studies suggest that the SDF-1α/CXCR4 axis may mediate resistance to VEGFR inhibition. Preclinical data have demonstrated that plerixafor (a reversible CXCR4 inhibitor) could inhibit glioma progression after anti-VEGF pathway inhibition. We conducted a phase I study to determine the safety of plerixafor and bevacizumab in recurrent HGG. Patients and Methods: Part 1 enrolled 23 patients with a 3 × 3 dose escalation design to a maximum planned dose of plerixafor 320 μg/kg subcutaneously on days 1 to 21 and bevacizumab 10 mg/kg intravenously on days 1 and 15 of each 28-day cycle. Cerebrospinal fluid (CSF) and plasma samples were obtained for pharmacokinetic analyses. Plasma and cellular biomarkers were evaluated before and after treatment. Part 2 enrolled 3 patients and was a surgical study to determine plerixafors penetration in tumor tissue. Results: In Part 1, no dose-limiting toxicities were seen at the maximum planned dose of plerixafor + bevacizumab. Treatment was well tolerated. After plerixafor 320 μg/kg treatment, the average CSF drug concentration was 26.8 ± 19.6 ng/mL. Plerixafor concentration in resected tumor tissue from patients pretreated with plerixafor was 10 to 12 μg/g. Circulating biomarker data indicated that plerixafor + bevacizumab induces rapid and persistent increases in plasma SDF-1α and placental growth factor. Progression-free survival correlated with pretreatment plasma soluble mesenchymal–epithelial transition receptor and sVEGFR1, and overall survival with the change during treatment in CD34+ progenitor/stem cells and CD8 T cells. Conclusions: Plerixafor + bevacizumab was well tolerated in HGG patients. Plerixafor distributed to both the CSF and brain tumor tissue, and treatment was associated with biomarker changes consistent with VEGF and CXCR4 inhibition. Clin Cancer Res; 24(19); 4643–9. ©2018 AACR.


Neuro-oncology | 2014

A randomized, placebo-controlled pilot trial of armodafinil for fatigue in patients with gliomas undergoing radiotherapy

Eudocia Q. Lee; Alona Muzikansky; Jan Drappatz; Santosh Kesari; Eric T. Wong; Camilo E. Fadul; David A. Reardon; Andrew D. Norden; Lakshmi Nayak; Mikael L. Rinne; Brian M. Alexander; Nils D. Arvold; Lisa Doherty; Jennifer Stefanik; Debra C. LaFrankie; Sandra Ruland; Julee Pulverenti; Katrina H. Smith; Sarah C. Gaffey; Samantha Hammond; Patrick Y. Wen


Journal of Neuro-oncology | 2015

Phase II trial of triple tyrosine kinase receptor inhibitor nintedanib in recurrent high-grade gliomas

Andrew D. Norden; David Schiff; Manmeet S. Ahluwalia; Glenn J. Lesser; Lakshmi Nayak; Eudocia Q. Lee; Mikael L. Rinne; Alona Muzikansky; Jorg Dietrich; Benjamin Purow; Lisa Doherty; Debra C. LaFrankie; Julee Pulverenti; Jennifer Rifenburg; Sandra Ruland; Katrina H. Smith; Sarah C. Gaffey; Christine McCluskey; Keith L. Ligon; David A. Reardon; Patrick Y. Wen


Journal of Clinical Oncology | 2011

Preliminary results from a multicenter, phase II, randomized, noncomparative clinical trial of radiation and temozolomide with or without vandetanib in newly diagnosed glioblastoma (GBM).

Eudocia C. Quant; Tracy T. Batchelor; Andrew B. Lassman; David Schiff; Thomas Kaley; Eric T. Wong; T. Mikkelsen; Jan Drappatz; Andrew D. Norden; Rameen Beroukhim; Stephanie E. Weiss; Brian M. Alexander; C. Sceppa; Mary Gerard; S. D. Hallisey; C. A. Bochacki; Katrina H. Smith; Alona Muzikansky; Patrick Y. Wen


Journal of Clinical Oncology | 2011

Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma.

Andrew D. Norden; Samantha Hammond; Jan Drappatz; Surasak Phuphanich; David A. Reardon; Eric T. Wong; Scott R. Plotkin; Glenn J. Lesser; Jeffrey Raizer; Tracy T. Batchelor; Eudocia C. Quant; Rameen Beroukhim; Thomas Kaley; Alona Muzikansky; A. S. Ciampa; Lisa Doherty; Katrina H. Smith; Mary Gerard; C. Sceppa; Patrick Y. Wen


Journal of Clinical Oncology | 2017

Phase I study of plerixafor and bevacizumab in recurrent high-grade glioma.

Eudocia Q. Lee; Alona Muzikansky; Elizabeth R. Gerstner; John G. Kuhn; David A. Reardon; Lakshmi Nayak; Andrew D. Norden; Lisa Doherty; Jennifer Rifenburg; Debra C. LaFrankie; Julee Pulverenti; Trupti Vardam-Kaur; Deirdre Stokes; Katrina H. Smith; Christine McCluskey; Sarah C. Gaffey; Tracy T. Batchelor; Dan G. Duda; Rakesh K. Jain; Patrick Y. Wen

Collaboration


Dive into the Katrina H. Smith's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge