Network


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

Hotspot


Dive into the research topics where Yunfeng Dai is active.

Publication


Featured researches published by Yunfeng Dai.


Blood | 2015

Prognostic significance of minimal residual disease in high risk B-ALL: a report from Children’s Oncology Group study AALL0232

Michael J. Borowitz; Brent L. Wood; Meenakshi Devidas; Mignon L. Loh; Elizabeth A. Raetz; Wanda L. Salzer; James Nachman; Andrew J. Carroll; Nyla A. Heerema; Julie M. Gastier-Foster; Cheryl L. Willman; Yunfeng Dai; Naomi J. Winick; Stephen P. Hunger; William L. Carroll; Eric Larsen

Minimal residual disease (MRD) is highly prognostic in pediatric B-precursor acute lymphoblastic leukemia (B-ALL). In Childrens Oncology Group high-risk B-ALL study AALL0232, we investigated MRD in subjects randomized in a 2 × 2 factorial design to receive either high-dose methotrexate (HD-MTX) or Capizzi methotrexate (C-MTX) during interim maintenance (IM) or prednisone or dexamethasone during induction. Subjects with end-induction MRD ≥0.1% or those with morphologic slow early response were nonrandomly assigned to receive a second IM and delayed intensification phase. MRD was measured by 6-color flow cytometry in 1 of 2 reference labs, with excellent agreement between the two. Subjects with end-induction MRD <0.01% had a 5-year event-free survival (EFS) of 87% ± 1% vs 74% ± 4% for those with MRD 0.01% to 0.1%; increasing MRD amounts was associated with progressively worse outcome. Subjects converting from MRD positive to negative by end consolidation had a relatively favorable 79% ± 5% 5-year disease-free survival vs 39% ± 7% for those with MRD ≥0.01%. Although HD-MTX was superior to C-MTX, MRD retained prognostic significance in both groups (86% ± 2% vs 58% ± 4% for MRD-negative vs positive C-MTX subjects; 88% ± 2% vs 68% ± 4% for HD-MTX subjects). Intensified therapy given to subjects with MRD >0.1% did not improve either 5-year EFS or overall survival (OS). However, these subjects showed an early relapse rate similar to that seen in MRD-negative ones, with EFS/OS curves for patients with 0.1% to 1% MRD crossing those with 0.01% to 0.1% MRD at 3 and 4 years, thus suggesting that the intensified therapy altered the disease course of MRD-positive subjects. Additional interventions targeted at the MRD-positive group may further improve outcome. This trial was registered at www.clinicaltrials.gov as #NCT00075725.


Blood | 2017

Targetable kinase gene fusions in high risk B-ALL: a study from the Children's Oncology Group

Shalini C. Reshmi; Richard C. Harvey; Kathryn G. Roberts; Eileen Stonerock; Amy Smith; Heather Jenkins; I. Ming Chen; Marc Valentine; Yu Liu; Yongjin Li; Ying Shao; John Easton; Debbie Payne-Turner; Zhaohui Gu; Thai Hoa Tran; Jonathan V. Nguyen; Meenakshi Devidas; Yunfeng Dai; Nyla A. Heerema; Andrew J. Carroll; Elizabeth A. Raetz; Michael J. Borowitz; Brent L. Wood; Anne L. Angiolillo; Michael J. Burke; Wanda L. Salzer; Patrick A. Zweidler-McKay; Karen R. Rabin; William L. Carroll; Jinghui Zhang

Philadelphia chromosome-like (Ph-like) acute lymphoblastic leukemia (ALL) is a high-risk subtype characterized by genomic alterations that activate cytokine receptor and kinase signaling. We examined the frequency and spectrum of targetable genetic lesions in a retrospective cohort of 1389 consecutively diagnosed patients with childhood B-lineage ALL with high-risk clinical features and/or elevated minimal residual disease at the end of remission induction therapy. The Ph-like gene expression profile was identified in 341 of 1389 patients, 57 of whom were excluded from additional analyses because of the presence of BCR-ABL1 (n = 46) or ETV6-RUNX1 (n = 11). Among the remaining 284 patients (20.4%), overexpression and rearrangement of CRLF2 (IGH-CRLF2 or P2RY8-CRLF2) were identified in 124 (43.7%), with concomitant genomic alterations activating the JAK-STAT pathway (JAK1, JAK2, IL7R) identified in 63 patients (50.8% of those with CRLF2 rearrangement). Among the remaining patients, using reverse transcriptase polymerase chain reaction or transcriptome sequencing, we identified targetable ABL-class fusions (ABL1, ABL2, CSF1R, and PDGFRB) in 14.1%, EPOR rearrangements or JAK2 fusions in 8.8%, alterations activating other JAK-STAT signaling genes (IL7R, SH2B3, JAK1) in 6.3% or other kinases (FLT3, NTRK3, LYN) in 4.6%, and mutations involving the Ras pathway (KRAS, NRAS, NF1, PTPN11) in 6% of those with Ph-like ALL. We identified 8 new rearrangement partners for 4 kinase genes previously reported to be rearranged in Ph-like ALL. The current findings provide support for the precision-medicine testing and treatment approach for Ph-like ALL implemented in Childrens Oncology Group ALL trials.


Nature Communications | 2016

Genomic analyses identify recurrent MEF2D fusions in acute lymphoblastic leukaemia.

Zhaohui Gu; Michelle L. Churchman; Kathryn G. Roberts; Yongjin Li; Yu Liu; Richard C. Harvey; Kelly McCastlain; Shalini C. Reshmi; Debbie Payne-Turner; Ilaria Iacobucci; Ying Shao; I. Ming Chen; Marcus B. Valentine; Deqing Pei; Karen Mungall; Andrew J. Mungall; Yussanne Ma; Richard A. Moore; Marco A. Marra; Eileen Stonerock; Julie M. Gastier-Foster; Meenakshi Devidas; Yunfeng Dai; Brent L. Wood; Michael J. Borowitz; Eric E. Larsen; Kelly W. Maloney; Leonard A. Mattano; Anne L. Angiolillo; Wanda L. Salzer

Chromosomal rearrangements are initiating events in acute lymphoblastic leukaemia (ALL). Here using RNA sequencing of 560 ALL cases, we identify rearrangements between MEF2D (myocyte enhancer factor 2D) and five genes (BCL9, CSF1R, DAZAP1, HNRNPUL1 and SS18) in 22 B progenitor ALL (B-ALL) cases with a distinct gene expression profile, the most common of which is MEF2D-BCL9. Examination of an extended cohort of 1,164 B-ALL cases identified 30 cases with MEF2D rearrangements, which include an additional fusion partner, FOXJ2; thus, MEF2D-rearranged cases comprise 5.3% of cases lacking recurring alterations. MEF2D-rearranged ALL is characterized by a distinct immunophenotype, DNA copy number alterations at the rearrangement sites, older diagnosis age and poor outcome. The rearrangements result in enhanced MEF2D transcriptional activity, lymphoid transformation, activation of HDAC9 expression and sensitive to histone deacetylase inhibitor treatment. Thus, MEF2D-rearranged ALL represents a distinct form of high-risk leukaemia, for which new therapeutic approaches should be considered.


Journal of Clinical Medicine Research | 2013

Outcomes of Salvage Autologous Versus Allogeneic Hematopoietic Cell Transplantation for Relapsed Multiple Myeloma After Initial Autologous Hematopoietic Cell Transplantation

Baldeep Wirk; Michael Byrne; Yunfeng Dai; Jan S. Moreb

Background Standard therapy for multiple myeloma (MM) includes initial autologous hematopoietic cell transplantation (autoHCT1) but this is not curative and most patients will relapse. Role of salvage autoHCT2 or allogeneic HCT (alloHCT2) is undefined. Methods MM patients who relapsed after autoHCT1 and had salvage autoHCT2 (N = 27) or alloHCT2 (N = 19) between 1995 - 2011 at our institution were studied retrospectively. Results Complete and very good partial remission (CR/VGPR) improved from 7% to 56% after autoHCT2 and from 26% to 37% after alloHCT2. Nonrelapse mortality (NRM) at 3 years was 3.7% for autoHCT2 and 5.3% for alloHCT2 (P = 0.901). Median progression free survival (PFS) and overall survival (OS) for autoHCT2 (19 months, 23 months) and alloHCT2 (6 months, 19 months) were not significantly different. On multivariate analysis, time from autoHCT1 to relapse ≥ 1year (HR 24.81, 95% CI 2.4 - 249.9) and maintenance therapy after autoHCT2 (HR 12.19, 95% CI 2.5 - 249.9) impacted OS after autoHCT2. Time from autoHCT1 to relapse < 1 year vs. ≥ 1 year (HR 18.55, 95% CI 2.28 - 150.57) impacted PFS after autoHCT2. For alloHCT2, no factors impacted NRM, PFS or OS. For those with relapse from autoHCT1 < 1 year vs. ≥ 1 year undergoing autoHCT2, median OS was 15 months (range, 1 - 53) vs. not yet reached at 143 months and median PFS was 5 months (range, 1 - 49) vs. not yet reached at 88 months. Conclusions Salvage autoHCT2 and alloHCT2 are both feasible for post autoHCT1 MM relapse. Relapse ≥ 1 year from autoHCT1 predicts for better PFS and OS after autoHCT2. Maintenance therapy after autoHCT2 is beneficial.


Blood | 2018

Preclinical efficacy of daratumumab in T-cell acute lymphoblastic leukemia (T-ALL)

Karen L. Bride; Tiffaney Vincent; Soo-Yeon L. Im; Richard Aplenc; David M. Barrett; William L. Carroll; Robin Carson; Yunfeng Dai; Meenakshi Devidas; Kimberly P. Dunsmore; Tori Fuller; Tina Glisovic-Aplenc; Terzah M. Horton; Stephen P. Hunger; Mignon L. Loh; Shannon L. Maude; Elizabeth A. Raetz; Stuart S. Winter; Stephan A. Grupp; Michelle L. Hermiston; Brent L. Wood; David T. Teachey

As a consequence of acquired or intrinsic disease resistance, the prognosis for patients with relapsed or refractory T-cell acute lymphoblastic leukemia (T-ALL) is dismal. Novel, less toxic drugs are clearly needed. One of the most promising emerging therapeutic strategies for cancer treatment is targeted immunotherapy. Immune therapies have improved outcomes for patients with other hematologic malignancies including B-cell ALL; however no immune therapy has been successfully developed for T-ALL. We hypothesize targeting CD38 will be effective against T-ALL. We demonstrate that blasts from patients with T-ALL have robust surface CD38 surface expression and that this expression remains stable after exposure to multiagent chemotherapy. CD38 is expressed at very low levels on normal lymphoid and myeloid cells and on a few tissues of nonhematopoietic origin, suggesting that CD38 may be an ideal target. Daratumumab is a human immunoglobulin G1κ monoclonal antibody that binds CD38, and has been demonstrated to be safe and effective in patients with refractory multiple myeloma. We tested daratumumab in a large panel of T-ALL patient-derived xenografts (PDX) and found striking efficacy in 14 of 15 different PDX. These data suggest that daratumumab is a promising novel therapy for pediatric T-ALL patients.


Journal of Clinical Oncology | 2018

TP53 germline variations influence the predisposition and prognosis of b-cell acute lymphoblastic leukemia in children

Maoxiang Qian; Xueyuan Cao; Meenakshi Devidas; Wenjian Yang; Cheng Cheng; Yunfeng Dai; Andrew J. Carroll; Nyla A. Heerema; Hui Zhang; Takaya Moriyama; Julie M. Gastier-Foster; Heng Xu; Elizabeth A. Raetz; Eric Larsen; Naomi J. Winick; W. Paul Bowman; Paul L. Martin; Elaine R. Mardis; Robert S. Fulton; Gerard P. Zambetti; Michael J. Borowitz; Brent L. Wood; Kim E. Nichols; William L. Carroll; Ching-Hon Pui; Charles G. Mullighan; William E. Evans; Stephen P. Hunger; Mary V. Relling; Mignon L. Loh

Purpose Germline TP53 variation is the genetic basis of Li-Fraumeni syndrome, a highly penetrant cancer predisposition condition. Recent reports of germline TP53 variants in childhood hypodiploid acute lymphoblastic leukemia (ALL) suggest that this type of leukemia is another manifestation of Li-Fraumeni syndrome; however, the pattern, prevalence, and clinical relevance of TP53 variants in childhood ALL remain unknown. Patients and Methods Targeted sequencing of TP53 coding regions was performed in 3,801 children from the Childrens Oncology Group frontline ALL clinical trials, AALL0232 and P9900. TP53 variant pathogenicity was evaluated according to experimentally determined transcriptional activity, in silico prediction of damaging effects, and prevalence in non-ALL control populations. TP53 variants were analyzed for their association with ALL presenting features and treatment outcomes. Results We identified 49 unique nonsilent rare TP53 coding variants in 77 (2.0%) of 3,801 patients sequenced, of which 22 variants were classified as pathogenic. TP53 pathogenic variants were significantly over-represented in ALL compared with non-ALL controls (odds ratio, 5.2; P < .001). Children with TP53 pathogenic variants were significantly older at ALL diagnosis (median age, 15.5 years v 7.3 years; P < .001) and were more likely to have hypodiploid ALL (65.4% v 1.2%; P < .001). Carrying germline TP53 pathogenic variants was associated with inferior event-free survival and overall survival (hazard ratio, 4.2 and 3.9; P < .001 and .001, respectively). In particular, children with TP53 pathogenic variants were at a dramatically higher risk of second cancers than those without pathogenic variants, with 5-year cumulative incidence of 25.1% and 0.7% ( P < .001), respectively. Conclusion Loss-of-function germline TP53 variants predispose children to ALL and to adverse treatment outcomes with ALL therapy, particularly the risk of second malignant neoplasms.


Blood | 2017

Measurable residual disease detection by high-throughput sequencing improves risk stratification for pediatric B-ALL

Brent L. Wood; David Wu; Beryl Crossley; Yunfeng Dai; David Williamson; Charles Gawad; Michael J. Borowitz; Meenakshi Devidas; Kelly W. Maloney; Eric Larsen; Naomi J. Winick; Elizabeth A. Raetz; William L. Carroll; Stephen P. Hunger; Mignon L. Loh; Harlan Robins; Ilan Kirsch

Early response to induction chemotherapy is an important prognostic factor in B-lymphoblastic leukemia (B-ALL). Here, we compare high-throughput sequencing (HTS) of IGH and TRG genes vs flow cytometry (FC) for measurable residual disease (MRD) detection at the end of induction chemotherapy in pediatric patients with newly diagnosed B-ALL. Six hundred nineteen paired pretreatment and end-of-induction bone marrow samples from Childrens Oncology Group studies AALL0331 (clinicaltrials.gov #NCT00103285) (standard risk [SR]; with MRD by FC at any level) and AALL0232 (clinicaltrials.gov #NCT00075725) (high risk; with day 29 MRD <0.1% by FC) were evaluated by HTS and FC for event-free (EFS) and overall survival (OS). HTS and FC showed similar 5-year EFS and OS for MRD-positive and -negative patients using an MRD threshold of 0.01%. However, there was a high discordant rate with HTS identifying 55 (38.7%) more patients MRD positive at this threshold. These discrepant patients have worse outcomes than FC MRD-negative patients. In addition, the increased analytic sensitivity of HTS permitted identification of 19.9% of SR patients without MRD at any detectable level who had excellent 5-year EFS (98.1%) and OS (100%). The higher analytic sensitivity and lower false-negative rate of HTS improves upon FC for MRD detection in pediatric B-ALL by identifying a novel subset of patients at end of induction who are essentially cured using current chemotherapy and identifying MRD at 0.01% in up to one-third of patients who are missed at the same threshold by FC.


Blood | 2018

Genomic and outcome analyses of Ph-like ALL in NCI standard-risk patients: a report from the Children’s Oncology Group

Kathryn G. Roberts; Shalini C. Reshmi; Richard C. Harvey; I-Ming Chen; Kinnari Patel; Eileen Stonerock; Heather Jenkins; Yunfeng Dai; Marc Valentine; Zhaohui Gu; Yaqi Zhao; Jinghui Zhang; Debbie Payne-Turner; Meenakshi Devidas; Nyla A. Heerema; Andrew J. Carroll; Elizabeth A. Raetz; Michael J. Borowitz; Brent L. Wood; Leonard A. Mattano; Kelly W. Maloney; William L. Carroll; Mignon L. Loh; Cheryl L. Willman; Julie M. Gastier-Foster; Charles G. Mullighan; Stephen P. Hunger

Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL; BCR-ABL1-like ALL) in children with National Cancer Institute (NCI) intermediate- or high-risk (HR) ALL is associated with poor outcome. Ph-like ALL is characterized by genetic alterations that activate cytokine receptor and kinase signaling and may be amenable to treatment with tyrosine kinase inhibitors. The prevalence, outcome, and potential for targeted therapy of Ph-like ALL in standard-risk (SR) ALL is less clear. We retrospectively analyzed a cohort of 1023 SR childhood B-ALL consecutively enrolled in the Childrens Oncology Group AALL0331 clinical trial. The Ph-like ALL gene expression profile was identified in 206 patients, and 67 patients with either BCR-ABL1 (n = 6) or ETV6-RUNX1 (n = 61) were excluded from downstream analysis, leaving 139 of 1023 (13.6%) as Ph-like. Targeted reverse transcription polymerase chain reaction assays and RNA-sequencing identified kinase-activating alterations in 38.8% of SR Ph-like cases, including CRLF2 rearrangements (29.5% of Ph-like), ABL-class fusions (1.4%), JAK2 fusions (1.4%), an NTRK3 fusion (0.7%), and other sequence mutations (IL7R, KRAS, NRAS; 5.6%). Patients with Ph-like ALL had inferior 7-year event-free survival compared with non-Ph-like ALL (82.4 ± 3.6% vs 90.7 ± 1.0%, P = .0022), with no difference in overall survival (93.2 ± 2.4% vs 95.8 ± 0.7%, P = .14). These findings illustrate the significant differences in the spectrum of kinase alterations and clinical outcome of Ph-like ALL based on presenting clinical features and establish that genomic alterations potentially targetable with approved kinase inhibitors are less frequent in SR than in HR ALL.


British Journal of Haematology | 2017

Klinefelter syndrome and 47,XYY syndrome in children with B cell acute lymphoblastic leukaemia

Rachel E. Rau; Andrew J. Carroll; Nyla A. Heerema; Lesley C. Arland; William L. Carroll; Naomi J. Winick; Elizabeth A. Raetz; Mignon L. Loh; Wenjian Yang; Mary V. Relling; Yunfeng Dai; Meenakshi Devidas; Stephen P. Hunger

Keywords: n nacute lymphoblastic leukaemia; nKlinefelter syndrome; n47,XYY syndrome; ncytogenetics; nSNP analysis


Biology of Blood and Marrow Transplantation | 2013

Outcomes of Salvage Autologous Versus Allogeneic Hematopoietic Cell Transplantation for Multiple Myeloma Relapsed After Initial Autologous Hematopoietic Cell Transplantation

Baldeep Wirk; Michael Byrne; Yunfeng Dai; Jan S. Moreb

Collaboration


Dive into the Yunfeng Dai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brent L. Wood

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mignon L. Loh

University of California

View shared research outputs
Top Co-Authors

Avatar

Stephen P. Hunger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew J. Carroll

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie M. Gastier-Foster

Nationwide Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge