Network


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

Hotspot


Dive into the research topics where Brian Weiss is active.

Publication


Featured researches published by Brian Weiss.


Pediatrics | 2016

Efficacy and Safety of Sirolimus in the Treatment of Complicated Vascular Anomalies.

Denise M. Adams; Cameron C. Trenor; Adrienne M. Hammill; Alexander A. Vinks; Manish N. Patel; Gulraiz Chaudry; Mary Sue Wentzel; Paula S. Mobberley-Schuman; Lisa M. Campbell; Christine Brookbank; Anita Gupta; Carol Chute; Jennifer Eile; Jesse McKenna; Arnold C. Merrow; Lin Fei; Lindsey Hornung; Michael Seid; A. Roshni Dasgupta; Belinda Dickie; Ravindhra G. Elluru; Anne W. Lucky; Brian Weiss; Richard G. Azizkhan

BACKGROUND AND OBJECTIVES: Complicated vascular anomalies have limited therapeutic options and cause significant morbidity and mortality. This Phase II trial enrolled patients with complicated vascular anomalies to determine the efficacy and safety of treatment with sirolimus for 12 courses; each course was defined as 28 days. METHODS: Treatment consisted of a continuous dosing schedule of oral sirolimus starting at 0.8 mg/m2 per dose twice daily, with pharmacokinetic-guided target serum trough levels of 10 to 15 ng/mL. The primary outcomes were responsiveness to sirolimus by the end of course 6 (evaluated according to functional impairment score, quality of life, and radiologic assessment) and the incidence of toxicities and/or infection-related deaths. RESULTS: Sixty-one patients were enrolled; 57 patients were evaluable for efficacy at the end of course 6, and 53 were evaluable at the end of course 12. No patient had a complete response at the end of course 6 or 12 as anticipated. At the end of course 6, a total of 47 patients had a partial response, 3 patients had stable disease, and 7 patients had progressive disease. Two patients were taken off of study medicine secondary to persistent adverse effects. Grade 3 and higher toxicities attributable to sirolimus included blood/bone marrow toxicity in 27% of patients, gastrointestinal toxicity in 3%, and metabolic/laboratory toxicity in 3%. No toxicity-related deaths occurred. CONCLUSIONS: Sirolimus was efficacious and well tolerated in these study patients with complicated vascular anomalies. Clinical activity was reported in the majority of the disorders.


Leukemia Research | 2009

Juvenile Myelomonocytic Leukemia: A Report from the 2nd International JMML Symposium

Rebecca J. Chan; Todd Cooper; Christian P. Kratz; Brian Weiss; Mignon L. Loh

Juvenile myelomonocytic leukemia (JMML) is an aggressive childhood myeloproliferative disorder characterized by the overproduction of myelomonocytic cells. JMML incidence approaches 1.2/million persons in the United States (Cancer Incidence and Survival Among Children and Adolescents: United States SEER Program 1975-1995). Although rare, JMML is innately informative as the molecular genetics of this disease implicates hyperactive Ras as an essential initiating event. Given that Ras is one of the most frequently mutated oncogenes in human cancer, findings from this disease are applicable to more genetically diverse and complex adult leukemias. The JMML Foundation (www.jmmlfoundation.org) was founded by parent advocates dedicated to finding a cure for this disease. They work to bring investigators together in a collaborative manner. This article summarizes key presentations from The Second International JMML Symposium, on 7-8 December 2007 in Atlanta, GA. A list of all participants is in Supplementary Table.


The New England Journal of Medicine | 2016

Activity of Selumetinib in Neurofibromatosis Type 1–Related Plexiform Neurofibromas

Eva Dombi; Andrea Baldwin; Leigh Marcus; Michael J. Fisher; Brian Weiss; AeRang Kim; Patricia Whitcomb; Staci Martin; Lindsey Aschbacher-Smith; Tilat A. Rizvi; Jianqiang Wu; Rachel Ershler; Pamela L. Wolters; Janet Therrien; John Glod; Jean B. Belasco; Elizabeth K. Schorry; Alessandra Brofferio; Amy J. Starosta; Andrea Gillespie; Austin L. Doyle; Nancy Ratner; Brigitte C. Widemann

BACKGROUND Effective medical therapies are lacking for the treatment of neurofibromatosis type 1-related plexiform neurofibromas, which are characterized by elevated RAS-mitogen-activated protein kinase (MAPK) signaling. METHODS We conducted a phase 1 trial of selumetinib (AZD6244 or ARRY-142886), an oral selective inhibitor of MAPK kinase (MEK) 1 and 2, in children who had neurofibromatosis type 1 and inoperable plexiform neurofibromas to determine the maximum tolerated dose and to evaluate plasma pharmacokinetics. Selumetinib was administered twice daily at a dose of 20 to 30 mg per square meter of body-surface area on a continuous dosing schedule (in 28-day cycles). We also tested selumetinib using a mouse model of neurofibromatosis type 1-related neurofibroma. Response to treatment (i.e., an increase or decrease from baseline in the volume of plexiform neurofibromas) was monitored by using volumetric magnetic resonance imaging analysis to measure the change in size of the plexiform neurofibroma. RESULTS A total of 24 children (median age, 10.9 years; range, 3.0 to 18.5) with a median tumor volume of 1205 ml (range, 29 to 8744) received selumetinib. Patients were able to receive selumetinib on a long-term basis; the median number of cycles was 30 (range, 6 to 56). The maximum tolerated dose was 25 mg per square meter (approximately 60% of the recommended adult dose). The most common toxic effects associated with selumetinib included acneiform rash, gastrointestinal effects, and asymptomatic creatine kinase elevation. The results of pharmacokinetic evaluations of selumetinib among the children in this trial were similar to those published for adults. Treatment with selumetinib resulted in confirmed partial responses (tumor volume decreases from baseline of ≥20%) in 17 of the 24 children (71%) and decreases from baseline in neurofibroma volume in 12 of 18 mice (67%). Disease progression (tumor volume increase from baseline of ≥20%) has not been observed to date. Anecdotal evidence of decreases in tumor-related pain, disfigurement, and functional impairment was observed. CONCLUSIONS Our early-phase data suggested that children with neurofibromatosis type 1 and inoperable plexiform neurofibromas benefited from long-term dose-adjusted treatment with selumetinib without having excess toxic effects. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01362803 .).


Journal of Pediatric Hematology Oncology | 2003

Secondary myelodysplastic syndrome and leukemia following 131I-metaiodobenzylguanidine Therapy for relapsed neuroblastoma

Brian Weiss; Amish Vora; John P. Huberty; Randall A. Hawkins; Katherine K. Matthay

Purpose To describe three patients with secondary leukemia after treatment with 131I-metaiodobenzylguanidine (MIBG) for neuroblastoma. Methods Of 95 children with refractory neuroblastoma treated with 131I-MIBG at UCSF, 3 have been identified with secondary myelodysplasia/leukemia. The case records and bone marrow results were reviewed, along with a review of the literature. Results Three patients developed secondary myelodysplasia/leukemia, at 7, 11, and 12 months following 131I-MIBG therapy. Cytogenetic abnormalities included −7q/−5, −7/+2q37, −11 and +12. Three additional cases were found in literature review of 509 reported patients treated with 131I-MIBG for neuroblastoma. Conclusions Therapy with 131I-MIBG may contribute to the risk of secondary leukemia in patients who have received intensive chemotherapy, thought the risk of this complication is far lower than the risk of disease progression. Further monitoring for this complication is indicated.


Neuro-oncology | 2015

Sirolimus for progressive neurofibromatosis type 1–associated plexiform neurofibromas: a Neurofibromatosis Clinical Trials Consortium phase II study

Brian Weiss; Brigitte C. Widemann; Pamela L. Wolters; Eva Dombi; Alexander A. Vinks; Alan Cantor; John P. Perentesis; Elizabeth K. Schorry; Nicole J. Ullrich; David H. Gutmann; James H. Tonsgard; David Viskochil; Bruce R. Korf; Roger J. Packer; Michael J. Fisher

BACKGROUND Plexiform neurofibromas (PNs) are benign peripheral nerve sheath tumors that arise in one-third of individuals with neurofibromatosis type 1 (NF1). They may cause significant disfigurement, compression of vital structures, neurologic dysfunction, and/or pain. Currently, the only effective management strategy is surgical resection. Converging evidence has demonstrated that the NF1 tumor suppressor protein, neurofibromin, negatively regulates activity in the mammalian Target of Rapamycin pathway. METHODS We employed a 2-strata clinical trial design. Stratum 1 included subjects with inoperable, NF1-associated progressive PN and sought to determine whether sirolimus safely and tolerably increases time to progression (TTP). Volumetric MRI analysis conducted at regular intervals was used to determine TTP relative to baseline imaging. RESULTS The estimated median TTP of subjects receiving sirolimus was 15.4 months (95% CI: 14.3-23.7 mo), which was significantly longer than 11.9 months (P < .001), the median TTP of the placebo arm of a previous PN clinical trial with similar eligibility criteria. CONCLUSIONS This study demonstrated that sirolimus prolongs TTP by almost 4 months in patients with NF1-associated progressive PN. Although the improvement in TTP is modest, given the lack of significant or frequent toxicity and the availability of few other treatment options, the use of sirolimus to slow the growth of progressive PN could be considered in select patients.


Current Neurology and Neuroscience Reports | 2012

Molecular Therapies for Tuberous Sclerosis and Neurofibromatosis

David Neal Franz; Brian Weiss

Neurofibromatosis type 1 (NF1) and tuberous sclerosis complex (TSC) are autosomal-dominant genetic disorders that result from dysregulation of the PI3K/AKT/mammalian target of rapamycin (mTOR) pathway. NF1 is caused by mutations in the NF1 gene on chromosome 17q11.2. Its protein product, neurofibromin, functions as a tumor suppressor and ultimately produces constitutive upregulation of mTOR. TSC is caused by mutations in either the TSC1 (chromosome 9q34) or TSC2 (chromosome 16p.13.3) genes. Their protein products, hamartin and tuberin, respectively, form a dimer that acts via the GAP protein Rheb (Ras homolog enhanced in brain) to directly inhibit mTOR, again resulting in upregulation. Specific inhibitors of mTOR are in clinical use, including sirolimus, everolimus, temsirolimus, and deforolimus. Everolimus has been shown to reduce the volume and appearance of subependymal giant cell astrocytomas (SEGA), facial angiofibromas, and renal angiomyolipomas associated with TSC, with a recent FDA approval for SEGA not suitable for surgical resection. This article reviews the use of mTOR inhibitors in these diseases, which have the potential to be a disease-modifying therapy in these and other conditions.


The Journal of Nuclear Medicine | 2012

Dose Escalation Study of No-Carrier-Added 131I-Metaiodobenzylguanidine for Relapsed or Refractory Neuroblastoma: New Approaches to Neuroblastoma Therapy Consortium Trial

Katherine K. Matthay; Brian Weiss; Judith G. Villablanca; John M. Maris; Gregory A. Yanik; Steven G. DuBois; James B. Stubbs; Susan Groshen; Denice D. Tsao-Wei; Randall A. Hawkins; Hollie A. Jackson; Fariba Goodarzian; Heike E. Daldrup-Link; Ashok Panigrahy; Alexander J. Towbin; Hiroyuki Shimada; John Barrett; Norman LaFrance; John W. Babich

131I-metaiodobenzylguanidine (MIBG) is specifically taken up in neuroblastoma, with a response rate of 20%–37% in relapsed disease. Nonradioactive carrier MIBG molecules inhibit uptake of 131I-MIBG, theoretically resulting in less tumor radiation and increased risk of cardiovascular toxicity. Our aim was to establish the maximum tolerated dose of no-carrier-added (NCA) 131I-MIBG, with secondary aims of assessing tumor and organ dosimetry and overall response. Methods: Eligible patients were 1–30 y old with resistant neuroblastoma, 131I-MIBG uptake, and cryopreserved hematopoietic stem cells. A diagnostic dose of NCA 131I-MIBG was followed by 3 dosimetry scans to assess radiation dose to critical organs and soft-tissue tumors. The treatment dose of NCA 131I-MIBG (specific activity, 165 MBq/μg) was adjusted as necessary on the basis of critical organ tolerance limits. Autologous hematopoietic stem cells were infused 14 d after therapy to abrogate prolonged myelosuppression. Response and toxicity were evaluated on day 60. The NCA 131I-MIBG was escalated from 444 to 777 MBq/kg (12–21 mCi/kg) using a 3 + 3 design. Dose-limiting toxicity (DLT) was failure to reconstitute neutrophils to greater than 500/μL within 28 d or platelets to greater than 20,000/μL within 56 d, or grade 3 or 4 nonhematologic toxicity by Common Terminology Criteria for Adverse Events (version 3.0) except for predefined exclusions. Results: Three patients each were evaluable at 444, 555, and 666 MBq/kg without DLT. The dose of 777 MBq/kg dose was not feasible because of organ dosimetry limits; however, 3 assigned patients were evaluable for a received dose of 666 MBq/kg, providing a total of 6 patients evaluable for toxicity at 666 MBq/kg without DLT. Mean whole-body radiation was 0.23 mGy/MBq, and mean organ doses were 0.92, 0.82, and 1.2 mGy/MBq of MIBG for the liver, lung, and kidney, respectively. Eight patients had 13 soft-tissue lesions with tumor-absorbed doses of 26–378 Gy. Four of 15 patients had a complete (n = 1) or partial (n = 3) response, 1 had a mixed response, 4 had stable disease, and 6 had progressive disease. Conclusion: NCA 131I-MIBG with autologous peripheral blood stem cell transplantation is feasible at 666 MBq/kg without significant nonhematologic toxicity and with promising activity.


Pediatric Blood & Cancer | 2014

Sirolimus for non-progressive NF1-associated plexiform neurofibromas: An NF clinical trials consortium phase II study

Brian Weiss; Brigitte C. Widemann; Pamela L. Wolters; Eva Dombi; Alexander A. Vinks; Alan Cantor; Bruce R. Korf; John P. Perentesis; David H. Gutmann; Elizabeth K. Schorry; Roger J. Packer; Michael J. Fisher

Patients with Neurofibromatosis Type 1 (NF1) have an increased risk of developing tumors of the central and peripheral nervous system, including plexiform neurofibromas (PN), which are benign nerve sheath tumors that are among the most debilitating complications of NF1. There are no standard treatment options for PN other than surgery, which is often difficult due to the extensive growth and invasion of surrounding tissues. Mammalian Target of Rapamcyin (mTOR) acts as a master switch of cellular catabolism and anabolism and controls protein translation, angiogenesis, cell motility, and proliferation. The NF1 tumor suppressor, neurofibromin, regulates the mTOR pathway activity. Sirolimus is a macrolide antibiotic that inhibits mTOR activity.


Pediatrics | 2014

A Team-Based Approach to Reducing Cardiac Monitor Alarms

Christopher E. Dandoy; Stella M. Davies; Laura Flesch; Melissa Hayward; Connie Koons; Kristen M. Coleman; Jodi Jacobs; Lori Ann McKenna; Alero Olomajeye; Chad Olson; Jessica Powers; Kimberly Shoemaker; Sonata Jodele; Evaline A. Alessandrini; Brian Weiss

BACKGROUND AND OBJECTIVES: Excessive cardiac monitor alarms lead to desensitization and alarm fatigue. We created and implemented a standardized cardiac monitor care process (CMCP) on a 24-bed pediatric bone marrow transplant unit. The aim of this project was to decrease monitor alarms through the use of team-based standardized care and processes. METHODS: Using small tests of change, we developed and implemented a standardized CMCP that included: (1) a process for initial ordering of monitor parameters based on age-appropriate standards; (2) pain-free daily replacement of electrodes; (3) daily individualized assessment of cardiac monitor parameters; and (4) a reliable method for appropriate discontinuation of monitor. The Model for Improvement was used to design, test, and implement changes. The changes that were implemented after testing and adaptation were: family/patient engagement in the CMCP; creation of a monitor care log to address parameters, lead changes, and discontinuation; development of a pain-free process for electrode removal; and customized monitor delay and customized threshold parameters. RESULTS: From January to November 2013, percent compliance with each of the 4 components of the CMCP increased. Overall compliance with the CMCP increased from a median of 38% to 95%. During this time, the median number of alarms per patient-day decreased from 180 to 40. CONCLUSIONS: Implementation of the standardized CMCP resulted in a significant decrease in cardiac monitor alarms per patient day. We recommend a team-based approach to monitor care, including individualized assessment of monitor parameters, daily lead change, and proper discontinuation of the monitors.


Pediatric Blood & Cancer | 2013

Pilot study of vincristine, oral irinotecan, and temozolomide (VOIT regimen) combined with bevacizumab in pediatric patients with recurrent solid tumors or brain tumors

Lars M. Wagner; Brian Turpin; Rajaram Nagarajan; Brian Weiss; Timothy P. Cripe; James I. Geller

The combination of vincristine, oral irinotecan, and temozolomide (VOIT regimen) has shown antitumor activity in a pediatric Phase I trial. To further potentiate synergy, we assessed the safety and feasibility of adding bevacizumab to VOIT for children and young adults with recurrent tumors.

Collaboration


Dive into the Brian Weiss's collaboration.

Top Co-Authors

Avatar

Brigitte C. Widemann

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael J. Fisher

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Araz Marachelian

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

John P. Perentesis

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Judith G. Villablanca

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Susan Groshen

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Eva Dombi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John M. Maris

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge