Network


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

Hotspot


Dive into the research topics where Craig Nolan is active.

Publication


Featured researches published by Craig Nolan.


Journal of Clinical Oncology | 2009

Randomized Phase II Trial of Chemoradiotherapy Followed by Either Dose-Dense or Metronomic Temozolomide for Newly Diagnosed Glioblastoma

Jennifer Clarke; Fabio M. Iwamoto; Joohee Sul; Katherine S. Panageas; Andrew B. Lassman; Lisa M. DeAngelis; Adília Hormigo; Craig Nolan; Igor T. Gavrilovic; Sasan Karimi; Lauren E. Abrey

PURPOSE Alternative dosing schedules of temozolomide may improve survival in patients with newly diagnosed glioblastoma (GBM) by increasing the therapeutic index, overcoming common mechanisms of temozolomide resistance, or both. The goal of this randomized phase II study was to evaluate two different temozolomide regimens in the adjuvant treatment of newly diagnosed GBM. PATIENTS AND METHODS Adult patients with newly diagnosed GBM were randomly assigned to receive standard radiotherapy with concurrent daily temozolomide followed by six adjuvant cycles of either dose-dense (150 mg/m(2) days 1 to 7 and 15 to 21) or metronomic (50 mg/m(2) continuous daily) temozolomide. Maintenance doses of 13-cis-retinoic acid were then administered until tumor progression. The primary end point was overall survival (OS) at 1 year. Tumor tissue was assayed to determine O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation status. RESULTS Eighty-five eligible patients were enrolled; 42 were randomly assigned to dose-dense and 43 to metronomic temozolomide. The 1-year survival rate was 80% for the dose-dense arm and 69% for the metronomic arm; median OS was 17.1 months (95% CI, 14.0 to 28.1 months) and 15.1 months (95% CI, 12.3 to 18.9 months), respectively. The most common toxicities were myelosuppression (leukopenia, neutropenia, and thrombocytopenia) and elevated liver enzymes. Pseudoprogression was observed in 37% of assessable patients and may have had an impact on estimates of progression-free survival (6.6 months in the dose-dense arm and 5.0 months in the metronomic arm). CONCLUSION Both dose-dense and metronomic temozolomide regimens were well tolerated with modest toxicity. The dose-dense regimen appears promising, with 1-year survival of 80%.


Blood | 2015

R-MPV followed by high-dose chemotherapy with TBC and autologous stem-cell transplant for newly diagnosed primary CNS lymphoma

Antonio Omuro; Denise D. Correa; Lisa M. DeAngelis; Craig H. Moskowitz; Matthew J. Matasar; Thomas Kaley; Igor T. Gavrilovic; Craig Nolan; Elena Pentsova; Christian Grommes; Katherine S. Panageas; Raymond E. Baser; Geraldine Faivre; Lauren E. Abrey; Craig S. Sauter

High-dose methotrexate-based chemotherapy is the mainstay of treatment of primary central nervous system lymphoma (PCNSL), but relapses remain frequent. High-dose chemotherapy (HDC) with autologous stem-cell transplant (ASCT) may provide an alternative to address chemoresistance and overcome the blood-brain barrier. In this single-center phase-2 study, newly diagnosed PCNSL patients received 5 to 7 cycles of chemotherapy with rituximab, methotrexate (3.5 g/m(2)), procarbazine, and vincristine (R-MPV). Those with a complete or partial response proceeded with consolidation HDC with thiotepa, cyclophosphamide, and busulfan, followed by ASCT and no radiotherapy. Primary end point was 1-year progression-free survival (PFS), N = 32. Median age was 57, and median Karnofsky performance status 80. Following R-MPV, objective response rate was 97%, and 26 (81%) patients proceeded with HDC-ASCT. Among all patients, median PFS and overall survival (OS) were not reached (median follow-up: 45 months). Two-year PFS was 79% (95% confidence interval [CI], 58-90), with no events observed beyond 2 years. Two-year OS was 81% (95% CI, 63-91). In transplanted patients, 2-year PFS and OS were 81%. There were 3 treatment-related deaths. Prospective neuropsychological evaluations suggested relatively stable cognitive functions posttransplant. In conclusion, this treatment was associated with excellent disease control and survival, an acceptable toxicity profile, and no evidence of neurotoxicity thus far. This trial was registered at www.clinicaltrials.gov as NCT00596154.


Neuro-oncology | 2015

Phase II trial of sunitinib for recurrent and progressive atypical and anaplastic meningioma

Thomas Kaley; Patrick Y. Wen; David Schiff; Keith L. Ligon; Sam Haidar; Sasan Karimi; Andrew B. Lassman; Craig Nolan; Lisa M. DeAngelis; Igor T. Gavrilovic; Andrew D. Norden; Jan Drappatz; Eudocia Q. Lee; Benjamin Purow; Scott R. Plotkin; Tracy T. Batchelor; Lauren E. Abrey; Antonio Omuro

BACKGROUND No proven effective medical therapy for surgery and radiation-refractory meningiomas exists. Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. METHODS This was a prospective, multicenter, investigator-initiated single-arm phase II trial. The primary cohort enrolled patients with surgery and radiation-refractory recurrent World Health Organization (WHO) grades II-III meningioma. An exploratory cohort enrolled patients with WHO grade I meningioma, hemangiopericytoma, or hemangioblastoma. Sunitinib was administered at 50 mg/d for days 1-28 of every 42-day cycle. The primary endpoint was the rate of 6-month progression-free survival (PFS6), with secondary endpoints of radiographic response rate, safety, PFS, and overall survival. Exploratory objectives include analysis of tumoral molecular markers and MR perfusion imaging. RESULTS Thirty-six patients with high-grade meningioma (30 atypical and 6 anaplastic) were enrolled. Patients were heavily pretreated (median number of 5 recurrences, range 2-10). PFS6 rate was 42%, meeting the primary endpoint. Median PFS was 5.2 months (95% CI: 2.8-8.3 mo), and median overall survival was 24.6 months (95% CI: 16.5-38.4 mo). Thirteen patients enrolled in the exploratory cohort. Overall toxicity included 1 grade 5 intratumoral hemorrhage, 2 grade 3 and 1 grade 4 CNS/intratumoral hemorrhages, 1 grade 3 and 1 grade 4 thrombotic microangiopathy, and 1 grade 3 gastrointestinal perforation. Expression of VEGFR2 predicted PFS of a median of 1.4 months in VEGFR2-negative patients versus 6.4 months in VEGFR2-positive patients (P = .005). CONCLUSION Sunitinib is active in recurrent atypical/malignant meningioma patients. A randomized trial should be performed.


Neuro-oncology | 2016

Integration of 2-hydroxyglutarate-proton magnetic resonance spectroscopy into clinical practice for disease monitoring in isocitrate dehydrogenase-mutant glioma

Mac Arena I De La Fuente; Robert J. Young; Jennifer Rubel; Marc K. Rosenblum; Jamie Tisnado; Samuel Briggs; Julio Arevalo-Perez; Justin R. Cross; Carl Campos; Kimberly Straley; Dongwei Zhu; Chuanhui Dong; Alissa A. Thomas; Antonio Omuro; Craig Nolan; Elena Pentsova; Thomas Kaley; Jung H. Oh; Ralph Noeske; Elizabeth A. Maher; Changho Choi; Philip H. Gutin; Andrei I. Holodny; Katharine Yen; Lisa M. DeAngelis; Ingo K. Mellinghoff; Sunitha B. Thakur

BACKGROUND The majority of WHO grades II and III gliomas harbor a missense mutation in the metabolic gene isocitrate dehydrogenase (IDH) and accumulate the metabolite R-2-hydroxyglutarate (R-2HG). Prior studies showed that this metabolite can be detected in vivo using proton magnetic-resonance spectroscopy (MRS), but the sensitivity of this methodology and its clinical implications are unknown. METHODS We developed an MR imaging protocol to integrate 2HG-MRS into routine clinical glioma imaging and examined its performance in 89 consecutive glioma patients. RESULTS Detection of 2-hydroxyglutarate (2HG) in IDH-mutant gliomas was closely linked to tumor volume, with sensitivity ranging from 8% for small tumors (<3.4 mL) to 91% for larger tumors (>8 mL). In patients undergoing 2HG-MRS prior to surgery, tumor levels of 2HG corresponded with tumor cellularity but not with tumor grade or mitotic index. Cytoreductive therapy resulted in a gradual decrease in 2HG levels with kinetics that closely mirrored changes in tumor volume. CONCLUSIONS Our study demonstrates that 2HG-MRS can be linked with routine MR imaging to provide quantitative measurements of 2HG in glioma and may be useful as an imaging biomarker to monitor the abundance of IDH-mutant tumor cells noninvasively during glioma therapy and disease monitoring.


Clinical Cancer Research | 2014

Phase II Study of Bevacizumab, Temozolomide and Hypofractionated Stereotactic Radiotherapy for Newly Diagnosed Glioblastoma

Antonio Omuro; Kathryn Beal; Philip H. Gutin; Sasan Karimi; Denise D. Correa; Thomas Kaley; Lisa M. DeAngelis; Timothy A. Chan; Igor T. Gavrilovic; Craig Nolan; Adília Hormigo; Andrew B. Lassman; Ingo K. Mellinghoff; Christian Grommes; Anne S. Reiner; Katherine S. Panageas; Raymond E. Baser; Viviane Tabar; Elena Pentsova; Juan Sánchez; Renata Barradas-Panchal; Jianan Zhang; Geraldine Faivre; Cameron Brennan; Lauren E. Abrey; Jason T. Huse

Purpose: Bevacizumab is associated with decreased vascular permeability that allows for more aggressive radiotherapy schedules. We conducted a phase II trial in newly diagnosed glioblastoma utilizing a novel hypofractionated stereotactic radiotherapy (HFSRT) schedule combined with temozolomide and bevacizumab. Experimental Design: Patients with tumor volume ≤60 cc were treated with HFSRT (6 × 6 Gy to contrast enhancement and 6 × 4 Gy to FLAIR hyperintensity with dose painting) combined with concomitant/adjuvant temozolomide and bevacizumab at standard doses. Primary endpoint was 1-year overall survival (OS): promising = 70%; nonpromising = 50%; α = 0.1; β = 0.1. Results: Forty patients were enrolled (median age: 55 years; methylated MGMT promoter: 23%; unmethylated: 70%). The 1-year OS was 93% [95% confidence interval (CI), 84–100] and median OS was 19 months. The median PFS was 10 months, with no pseudo-progression observed. The objective response rate (ORR) was 57%. Analysis of The Cancer Genome Atlas glioblastoma transcriptional subclasses (Nanostring assay) suggested patients with a proneural phenotype (26%) fared worse (ORR = 14%, vs. 77% for other subclasses; P = 0.009). Dynamic susceptibility-contrast perfusion MRI showed marked decreases in relative cerebral blood volume over time (P < 0.0001) but had no prognostic value, whereas higher baseline apparent diffusion coefficient (ADC) ratios and persistent hypermetabolism at the 6-month FDG-PET predicted poor OS (P = 0.05 and 0.0001, respectively). Quality-of-life (FACT-BR-4) and neuropsychological test scores were stable over time, although some domains displayed transient decreases following HFSRT. Conclusions: This aggressive radiotherapy schedule was safe and more convenient for patients, achieving an OS that is comparable with historical controls. Analysis of advanced neuroimaging parameters suggests ADC and FDG-PET as potentially useful biomarkers, whereas tissue correlatives uncovered the poor prognosis associated with the proneural signature in non–IDH-1–mutated glioblastoma. Clin Cancer Res; 20(19); 5023–31. ©2014 AACR.


Cancer Discovery | 2017

Ibrutinib Unmasks Critical Role of Bruton Tyrosine Kinase in Primary CNS Lymphoma

Christian Grommes; Alessandro Pastore; Nicolaos Palaskas; Sarah S Tang; Carl Campos; Derrek Schartz; Paolo Codega; Donna Nichol; Owen Clark; Wan-Ying Hsieh; Daniel Rohle; Marc K. Rosenblum; Agnes Viale; Viviane Tabar; Cameron Brennan; Igor T. Gavrilovic; Thomas Kaley; Craig Nolan; Antonio Omuro; Elena Pentsova; Alissa A. Thomas; Elina Tsyvkin; Ariela Noy; M. Lia Palomba; Paul A. Hamlin; Craig S. Sauter; Craig H. Moskowitz; Julia Wolfe; Ahmet Dogan; Minhee Won

Bruton tyrosine kinase (BTK) links the B-cell antigen receptor (BCR) and Toll-like receptors with NF-κB. The role of BTK in primary central nervous system (CNS) lymphoma (PCNSL) is unknown. We performed a phase I clinical trial with ibrutinib, the first-in-class BTK inhibitor, for patients with relapsed or refractory CNS lymphoma. Clinical responses to ibrutinib occurred in 10 of 13 (77%) patients with PCNSL, including five complete responses. The only PCNSL with complete ibrutinib resistance harbored a mutation within the coiled-coil domain of CARD11, a known ibrutinib resistance mechanism. Incomplete tumor responses were associated with mutations in the B-cell antigen receptor-associated protein CD79B. CD79B-mutant PCNSLs showed enrichment of mammalian target of rapamycin (mTOR)-related gene sets and increased staining with PI3K/mTOR activation markers. Inhibition of the PI3K isoforms p110α/p110δ or mTOR synergized with ibrutinib to induce cell death in CD79B-mutant PCNSL cells.Significance: Ibrutinib has substantial activity in patients with relapsed or refractory B-cell lymphoma of the CNS. Response rates in PCNSL were considerably higher than reported for diffuse large B-cell lymphoma outside the CNS, suggesting a divergent molecular pathogenesis. Combined inhibition of BTK and PI3K/mTOR may augment the ibrutinib response in CD79B-mutant human PCNSLs. Cancer Discov; 7(9); 1018-29. ©2017 AACR.See related commentary by Lakshmanan and Byrd, p. 940This article is highlighted in the In This Issue feature, p. 920.


Clinical Cancer Research | 2016

Molecular and Clinical Effects of Notch Inhibition in Glioma Patients: A Phase 0/I Trial

Xu R; Fumiko Shimizu; Koos E. Hovinga; Kathryn Beal; Sasan Karimi; Droms L; Peck Kk; P.H. Gutin; Iorgulescu Jb; Thomas Kaley; Lisa M. DeAngelis; Elena Pentsova; Craig Nolan; Christian Grommes; Timothy A. Chan; Dylan Bobrow; Hormigo A; Cross; Nian Wu; Naoko Takebe; K. S. Panageas; Ivy P; Jeffrey G. Supko; Tabar; Antonio Omuro

Purpose: High-grade gliomas are associated with a dismal prognosis. Notch inhibition via the gamma-secretase inhibitor RO4929097 has emerged as a potential therapeutic option based on modulation of the cancer-initiating cell (CIS) population and a presumed antiangiogenic role. Experimental Design: In this phase 0/I trial, 21 patients with newly diagnosed glioblastoma or anaplastic astrocytoma received RO4929097 combined with temozolomide and radiotherapy. In addition to establishing the MTD, the study design enabled exploratory studies evaluating tumor and brain drug penetration and neuroimaging parameters. We also determined functional effects on the Notch pathway and targeting of CISs through analysis of tumor tissue sampled from areas with and without blood–brain barrier disruption. Finally, recurrent tumors were also sampled and assessed for Notch pathway responses while on treatment. Results: Treatment was well tolerated and no dose-limiting toxicities were observed. IHC of treated tumors showed a significant decrease in proliferation and in the expression of the Notch intracellular domain (NICD) by tumor cells and blood vessels. Patient-specific organotypic tumor explants cultures revealed a specific decrease in the CD133+ CIS population upon treatment. Perfusion MRI demonstrated a significant decrease in relative plasma volume after drug exposure. Gene expression data in recurrent tumors suggested low Notch signaling activity, the upregulation of key mesenchymal genes, and an increase in VEGF-dependent angiogenic factors. Conclusions: The addition of RO4929097 to temozolomide and radiotherapy was well tolerated; the drug has a variable blood–brain barrier penetration. Evidence of target modulation was observed, but recurrence occurred, associated with alterations in angiogenesis signaling pathways. Clin Cancer Res; 22(19); 4786–96. ©2016 AACR.


Aminoff's Neurology and General Medicine (Fifth Edition) | 2015

Neurologic Complications of Chemotherapy and Radiation Therapy

Craig Nolan; Lisa M. DeAngelis

Purpose of Review:Nervous system injury from cancer therapeutics is second only to myelosuppression as the most common toxicity caused by chemotherapy and radiation therapy. Recent Findings:Neurologic complications can be seen throughout the course of cancer diagnosis and treatment; some toxicities may not develop for years or even decades after completion of treatment, making diagnosis challenging. However, early recognition is critical as modification of therapeutic regimens can diminish long-term consequences of nervous system injury and improve a patient’s quality of life. This article addresses the most common neurologic complications seen as a consequence of chemotherapy and radiation therapy in patients with systemic cancer. These common toxicities are likely to be seen with increasing frequency as patients with systemic cancer are living longer and receiving multiple courses of antineoplastic regimens, some of which are lifelong. Summary:Neurotoxicity from cancer therapeutics is common, and early recognition is critical to proper diagnosis and intervention.


Cancer treatment and research | 2005

Leptomeningeal Metastases from Leukemias and Lymphomas

Craig Nolan; Lauren E. Abrey

Leptomeningeal dissemination of lymphoma and leukemia differs from that of solid tumors in a number of clinically important aspects. Specific histologic variants of lymphoma and leukemia have such a high incidence of cerebrospinal fluid (CSF) dissemination that assessing CSF cytology at diagnosis is crucial and prophylactic therapy of the CSF compartment is required. Furthermore, while the overall prognosis for patients with leptomeningeal metastases from leukemia and lymphoma is similar to solid tumors, selected patients have excellent response to therapy and attain durable remission. Therefore, aggressive treatment is warranted.


CNS oncology | 2013

Bevacizumab for acute neurologic deterioration in patients with glioblastoma.

Thomas Kaley; Craig Nolan; Alan Carver; Antonio Omuro

AIM Glioblastoma causes neurologic dysfunction owing to the tumors location and peritumoral edema. Bevacizumab improves symptoms and steroid dependence, and may rescue glioblastoma patients hospitalized for acute neurologic dysfunction, allowing them to return home for outpatient treatment. MATERIALS & METHODS We carried out a retrospective review of glioblastoma patients with severe neurologic dysfunction who received bevacizumab as inpatients. RESULTS Nine patients (median age: 51 years; median Karnofsky Performance Status [KPS]: 40%) received one dose of bevacizumab while admitted for neurologic deterioration. Seven patients were treated at recurrence, two at diagnosis. Six patients clinically improved and continued outpatient treatment, while five decreased/discontinued dexamethasone. CONCLUSION Single bevacizumab treatment administered to naive hospitalized patients with glioblastoma improves function and quality of life through avoidance of prolonged hospitalization and rehabilitation admissions, and decreased dexamethasone administration.

Collaboration


Dive into the Craig Nolan's collaboration.

Top Co-Authors

Avatar

Lisa M. DeAngelis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Antonio Omuro

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Igor T. Gavrilovic

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Thomas Kaley

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ingo K. Mellinghoff

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christian Grommes

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elena Pentsova

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lauren E. Abrey

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrew B. Lassman

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adília Hormigo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge