Network


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

Hotspot


Dive into the research topics where Todd Cooper is active.

Publication


Featured researches published by Todd Cooper.


Blood | 2012

Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children's Oncology Group

Michael R. Loken; Todd A. Alonzo; Laura Pardo; Robert B. Gerbing; Susana C. Raimondi; Betsy Hirsch; Phoenix A. Ho; Todd Cooper; Alan S. Gamis; Soheil Meshinchi

Early response to induction chemotherapy is a predictor of outcome in acute myeloid leukemia (AML). We determined the prevalence and significance of postinduction residual disease (RD) by multidimensional flow cytometry (MDF) in children treated on Childrens Oncology Group AML protocol AAML03P1. Postinduction marrow specimens at the end of induction (EOI) 1 or 2 or at the end of therapy from 249 patients were prospectively evaluated by MDF for RD, and presence of RD was correlated with disease characteristics and clinical outcome. Of the 188 patients in morphologic complete remission at EOI1, 46 (24%) had MDF-detectable disease. Those with and without RD at the EOI1 had a 3-year relapse risk of 60% and 29%, respectively (P < .001); the corresponding relapse-free survival was 30% and 65% (P < .001). Presence of RD at the EOI2 and end of therapy was similarly predictive of poor outcome. RD was detected in 28% of standard-risk patients in complete remission and was highly associated with poor relapse-free survival (P = .008). In a multivariate analysis, including cytogenetic and molecular risk factors, RD was an independent predictor of relapse (P < .001). MDF identifies patients at risk of relapse and poor outcome and can be incorporated into clinical trials for risk-based therapy allocation. This study was registered at www.clinicaltrials.gov as NCT00070174.


Cancer | 2012

AAML03P1, a pilot study of the safety of gemtuzumab ozogamicin in combination with chemotherapy for newly diagnosed childhood acute myeloid leukemia: a report from the Children's Oncology Group.

Todd Cooper; Robert B. Gerbing; Todd A. Alonzo; Craig A. Hurwitz; Susana C. Raimondi; Betsy Hirsch; Franklin O. Smith; Prasad Mathew; Robert J. Arceci; James H. Feusner; Robert Iannone; Robert S. Lavey; Soheil Meshinchi; Alan S. Gamis

The development of antigen‐targeted therapies may provide additional options to improve outcomes in children with acute myeloid leukemia (AML). The Childrens Oncology Group AAML03P1 trial sought to determine the safety of adding 2 doses of gemtuzumab ozogamicin, a humanized anti‐CD33 antibody‐targeted agent, to intensive chemotherapy during remission induction and postremission intensification for children with de novo AML.


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.


Blood | 2011

Phase 2 trial of clofarabine in combination with etoposide and cyclophosphamide in pediatric patients with refractory or relapsed acute lymphoblastic leukemia

Nobuko Hijiya; Blythe Thomson; Michael S. Isakoff; Lewis B. Silverman; Peter G. Steinherz; Michael J. Borowitz; Richard Kadota; Todd Cooper; Violet Shen; Gary V. Dahl; Jaideep V. Thottassery; Sima Jeha; Kelly W. Maloney; Jo Anne Paul; Elly Barry; William L. Carroll; Paul S. Gaynon

The outcomes in children with refractory/relapsed (R/R) acute lymphoblastic leukemia (ALL) are dismal. The efficacy and safety of intravenous clofarabine 40 mg/m(2) per day, cyclophosphamide 440 mg/m(2) per day, and etoposide 100 mg/m(2) per day for 5 consecutive days in pediatric patients with R/R ALL was evaluated in this phase 2 study. The primary endpoint was overall response rate (complete remission [CR] plus CR without platelet recovery [CRp]). Among the 25 patients (median age, 14 years; pre-B cell ALL, 84%; ≥ 2 prior regimens: 84%; refractory to previous regimen: 60%), the overall response rate was 44% (7 CR, 4 CRp) with a 67.3-week median duration or remission censored at last follow-up. Most patients proceeded to alternative therapy, and 10 patients (40%) received hematopoietic stem cell transplantation. Six patients (24%) died because of treatment-related adverse events associated with infection, hepatotoxicity, and/or multiorgan failure. The study protocol was amended to exclude patients with prior hematopoietic stem cell transplantation after 4 of the first 8 patients developed severe hepatotoxicity suggestive of veno-occlusive disease. No additional cases of veno-occlusive disease occurred. The regimen offered encouraging response rates and sustained remission in R/R patients. Future investigation should include exploration of patient selection, dosing, and supportive care. This trial was registered at www.clinicaltrials.gov as #NCT00315705.


Leukemia | 2009

A multi-center phase I study of clofarabine, etoposide and cyclophosphamide in combination in pediatric patients with refractory or relapsed acute leukemia

Nobuko Hijiya; Paul S. Gaynon; Elly Barry; Lewis B. Silverman; Blythe Thomson; Roland Chu; Todd Cooper; Richard Kadota; Michael Rytting; Peter G. Steinherz; Violet Shen; Sima Jeha; R. Abichandani; William L. Carroll

This Phase I study of clofarabine with etoposide and cyclophosphamide for children with relapsed/refractory acute lymphoblastic leukemia (ALL) or acute myelogenous leukemia (AML) was conducted to determine the maximum tolerated dose (MTD), dose-limiting toxicities and the recommended phase 2 doses (RP2Ds). All three drugs were administered for five consecutive days in induction and four consecutive days in consolidation, for a maximum of eight cycles. A total of 25 patients (20 ALL and 5 AML) were enrolled in five cohorts. An MTD was not reached. The RP2Ds of clofarabine, cyclophosphamide and etoposide were 40, 440 and 100 mg/m2/day, respectively. Complete remission (CR) was achieved in 10 patients (ALL: nine; AML: one), and CR without platelet recovery in six patients (ALL: two; AML: four) for an overall response rate of 64% (ALL: 55%; AML: 100%). Of the 16 responders, 9 patients proceeded to hematopoietic stem cell transplantation. In conclusion, the combination of clofarabine, etoposide and cyclophosphamide was well tolerated and effective in pediatric patients with relapsed/refractory leukemia. Of note, the phase II portion of the trial was amended after the occurrence of unexpected hepatotoxicity. The ongoing phase II study will evaluate the efficacy and safety of this regimen in ALL patients.


Journal of Clinical Oncology | 2016

Phase I/Phase II Study of Blinatumomab in Pediatric Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia

Arend von Stackelberg; Franco Locatelli; Gerhard Zugmaier; Rupert Handgretinger; Tanya M. Trippett; Carmelo Rizzari; Peter Bader; Maureen M. O’Brien; Benoit Brethon; Deepa Bhojwani; Paul G. Schlegel; Arndt Borkhardt; Susan R. Rheingold; Todd Cooper; Christian M. Zwaan; Phillip Barnette; Chiara Messina; Gérard Michel; Steven G. DuBois; Kuolung Hu; Min Zhu; James A. Whitlock; Lia Gore

Purpose Blinatumomab is a bispecific T-cell engager antibody construct targeting CD19 on B-cell lymphoblasts. We evaluated the safety, pharmacokinetics, recommended dosage, and potential for efficacy of blinatumomab in children with relapsed/refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Methods This open-label study enrolled children < 18 years old with relapsed/refractory BCP-ALL in a phase I dosage-escalation part and a phase II part, using 6-week treatment cycles. Primary end points were maximum-tolerated dosage (phase I) and complete remission rate within the first two cycles (phase II). Results We treated 49 patients in phase I and 44 patients in phase II. Four patients had dose-limiting toxicities in cycle 1 (phase I). Three experienced grade 4 cytokine-release syndrome (one attributed to grade 5 cardiac failure); one had fatal respiratory failure. The maximum-tolerated dosage was 15 µg/m2/d. Blinatumomab pharmacokinetics was linear across dosage levels and consistent among age groups. On the basis of the phase I data, the recommended blinatumomab dosage for children with relapsed/refractory ALL was 5 µg/m2/d for the first 7 days, followed by 15 µg/m2/d thereafter. Among the 70 patients who received the recommended dosage, 27 (39%; 95% CI, 27% to 51%) achieved complete remission within the first two cycles, 14 (52%) of whom achieved complete minimal residual disease response. The most frequent grade ≥ 3 adverse events were anemia (36%), thrombocytopenia (21%), and hypokalemia (17%). Three patients (4%) and one patient (1%) had cytokine-release syndrome of grade 3 and 4, respectively. Two patients (3%) interrupted treatment after grade 2 seizures. Conclusion This trial, which to the best of our knowledge was the first such trial in pediatrics, demonstrated antileukemic activity of single-agent blinatumomab with complete minimal residual disease response in children with relapsed/refractory BCP-ALL. Blinatumomab may represent an important new treatment option in this setting, requiring further investigation in curative indications.


Nature Genetics | 2015

The genomic landscape of juvenile myelomonocytic leukemia

Elliot Stieglitz; Amaro Taylor-Weiner; Tiffany Y. Chang; Laura C. Gelston; Yong Dong Wang; Tali Mazor; Emilio Esquivel; Ariel Yu; Sara Seepo; Scott R. Olsen; Mara Rosenberg; Sophie Archambeault; Ghada Abusin; Kyle Beckman; Patrick Brown; Michael Briones; Benjamin Carcamo; Todd Cooper; Gary V. Dahl; Peter D. Emanuel; Mark Fluchel; Rakesh K. Goyal; Robert J. Hayashi; Johann Hitzler; Christopher Hugge; Y. Lucy Liu; Yoav Messinger; Donald H. Mahoney; Philip Monteleone; Eneida R. Nemecek

Juvenile myelomonocytic leukemia (JMML) is a myeloproliferative neoplasm (MPN) of childhood with a poor prognosis. Mutations in NF1, NRAS, KRAS, PTPN11 or CBL occur in 85% of patients, yet there are currently no risk stratification algorithms capable of predicting which patients will be refractory to conventional treatment and could therefore be candidates for experimental therapies. In addition, few molecular pathways aside from the RAS-MAPK pathway have been identified that could serve as the basis for such novel therapeutic strategies. We therefore sought to genomically characterize serial samples from patients at diagnosis through relapse and transformation to acute myeloid leukemia to expand knowledge of the mutational spectrum in JMML. We identified recurrent mutations in genes involved in signal transduction, splicing, Polycomb repressive complex 2 (PRC2) and transcription. Notably, the number of somatic alterations present at diagnosis appears to be the major determinant of outcome.


Pediatric Blood & Cancer | 2015

Sorafenib treatment following hematopoietic stem cell transplant in pediatric FLT3/ITD acute myeloid leukemia

Katherine Tarlock; Bill H. Chang; Todd Cooper; Thomas G. Gross; Sumit Gupta; Steven Neudorf; Kathleen Adlard; Phoenix A. Ho; Suzanne Mcgoldrick; Tanya Watt; Tina Templeman; India Sisler; Amy Garee; Blythe Thomson; Ann E. Woolfrey; Elihu H. Estey; Soheil Meshinchi; Jessica A. Pollard

FLT3/ITD is associated with poor outcomes in adult and pediatric acute myeloid leukemia (AML). Allogeneic hematopoietic stem cell transplantation (HSCT) can improve cure rates, however relapse is still common. Recent studies demonstrate the activity of FLT3 inhibitors, including sorafenib, in targeting the underlying mutation.


Pediatric Blood & Cancer | 2012

Sorafenib as treatment for relapsed or refractory pediatric acute myelogenous leukemia

Tanya Watt; Todd Cooper

The prognosis for children with acute myelogenous leukemia (AML) has improved with overall survival rates of up to 65% [Pui et al. J Clin Oncol 2011; 29: 551–565]. However, the cure rate for AML lags behind that of acute lymphoblastic leukemia. Advances in AML leukemogenesis are leading to refined risk stratification. FMS like tyrosine kinase 3 (FLT3) mutations are independently associated with a poor prognosis. Newer kinase inhibitors, including sorafenib, have shown promise in adult studies. We report three pediatric patients with relapsed AML who achieved a sustained remission with sorafenib. Further trials are necessary to understand the role of sorafenib in pediatric AML. Pediatr Blood Cancer 2012;59:756–757.


Pediatric Blood & Cancer | 2015

Phase II/III trial of a pre-transplant farnesyl transferase inhibitor in juvenile myelomonocytic leukemia: a report from the Children's Oncology Group.

Elliot Stieglitz; Ashley F. Ward; Robert B. Gerbing; Todd A. Alonzo; Robert J. Arceci; Y. Lucy Liu; Peter D. Emanuel; Brigitte C. Widemann; Jennifer W. Cheng; Nalini Jayaprakash; Frank M. Balis; Robert P. Castleberry; Nancy Bunin; Mignon L. Loh; Todd Cooper

Juvenile myelomonocytic leukemia (JMML) is not durably responsive to chemotherapy, and approximately 50% of patients relapse after hematopoietic stem cell transplant (HSCT). Here we report the activity and acute toxicity of the farnesyl transferase inhibitor tipifarnib, the response rate to 13‐cis retinoic acid (CRA) in combination with cytoreductive chemotherapy, and survival following HSCT in children with JMML.

Collaboration


Dive into the Todd Cooper's collaboration.

Top Co-Authors

Avatar

Lia Gore

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Soheil Meshinchi

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Todd A. Alonzo

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Alan S. Gamis

Children's Mercy Hospital

View shared research outputs
Top Co-Authors

Avatar

Betsy Hirsch

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mignon L. Loh

University of California

View shared research outputs
Top Co-Authors

Avatar

Phoenix A. Ho

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Susana C. Raimondi

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge