Network


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

Hotspot


Dive into the research topics where Deqing Pei is active.

Publication


Featured researches published by Deqing Pei.


Nature | 2012

The genetic basis of early T-cell precursor acute lymphoblastic leukaemia.

Jinghui Zhang; Li Ding; Linda Holmfeldt; Gang Wu; Susan L. Heatley; Debbie Payne-Turner; John Easton; Xiang Chen; Jianmin Wang; Michael Rusch; Charles Lu; Shann Ching Chen; Lei Wei; J. Racquel Collins-Underwood; Jing Ma; Kathryn G. Roberts; Stanley Pounds; Anatoly Ulyanov; Jared Becksfort; Pankaj Gupta; Robert Huether; Richard W. Kriwacki; Matthew Parker; Daniel J. McGoldrick; David Zhao; Daniel Alford; Stephen Espy; Kiran Chand Bobba; Guangchun Song; Deqing Pei

Early T-cell precursor acute lymphoblastic leukaemia (ETP ALL) is an aggressive malignancy of unknown genetic basis. We performed whole-genome sequencing of 12 ETP ALL cases and assessed the frequency of the identified somatic mutations in 94 T-cell acute lymphoblastic leukaemia cases. ETP ALL was characterized by activating mutations in genes regulating cytokine receptor and RAS signalling (67% of cases; NRAS, KRAS, FLT3, IL7R, JAK3, JAK1, SH2B3 and BRAF), inactivating lesions disrupting haematopoietic development (58%; GATA3, ETV6, RUNX1, IKZF1 and EP300) and histone-modifying genes (48%; EZH2, EED, SUZ12, SETD2 and EP300). We also identified new targets of recurrent mutation including DNM2, ECT2L and RELN. The mutational spectrum is similar to myeloid tumours, and moreover, the global transcriptional profile of ETP ALL was similar to that of normal and myeloid leukaemia haematopoietic stem cells. These findings suggest that addition of myeloid-directed therapies might improve the poor outcome of ETP ALL.


The New England Journal of Medicine | 2009

Treating Childhood Acute Lymphoblastic Leukemia without Cranial Irradiation

Ching-Hon Pui; Dario Campana; Deqing Pei; W. Paul Bowman; John T. Sandlund; Sue C. Kaste; Raul C. Ribeiro; Jeffrey E. Rubnitz; Susana C. Raimondi; Mihaela Onciu; Elaine Coustan-Smith; Larry E. Kun; Sima Jeha; Cheng Cheng; Scott C. Howard; Vickey Simmons; Amy Bayles; Monika L. Metzger; James M. Boyett; Wing Leung; Rupert Handgretinger; James R. Downing; William E. Evans; Mary V. Relling

BACKGROUND Prophylactic cranial irradiation has been a standard treatment in children with acute lymphoblastic leukemia (ALL) who are at high risk for central nervous system (CNS) relapse. METHODS We conducted a clinical trial to test whether prophylactic cranial irradiation could be omitted from treatment in all children with newly diagnosed ALL. A total of 498 patients who could be evaluated were enrolled. Treatment intensity was based on presenting features and the level of minimal residual disease after remission-induction treatment. The duration of continuous complete remission in the 71 patients who previously would have received prophylactic cranial irradiation was compared with that of 56 historical controls who received it. RESULTS The 5-year event-free and overall survival probabilities for all 498 patients were 85.6% (95% confidence interval [CI], 79.9 to 91.3) and 93.5% (95% CI, 89.8 to 97.2), respectively. The 5-year cumulative risk of isolated CNS relapse was 2.7% (95% CI, 1.1 to 4.3), and that of any CNS relapse (including isolated relapse and combined relapse) was 3.9% (95% CI, 1.9 to 5.9). The 71 patients had significantly longer continuous complete remission than the 56 historical controls (P=0.04). All 11 patients with isolated CNS relapse remained in second remission for 0.4 to 5.5 years. CNS leukemia (CNS-3 status) or a traumatic lumbar puncture with blast cells at diagnosis and a high level of minimal residual disease (> or = 1%) after 6 weeks of remission induction were significantly associated with poorer event-free survival. Risk factors for CNS relapse included the genetic abnormality t(1;19)(TCF3-PBX1), any CNS involvement at diagnosis, and T-cell immunophenotype. Common adverse effects included allergic reactions to asparaginase, osteonecrosis, thrombosis, and disseminated fungal infection. CONCLUSIONS With effective risk-adjusted chemotherapy, prophylactic cranial irradiation can be safely omitted from the treatment of childhood ALL. (ClinicalTrials.gov number, NCT00137111.)


Lancet Oncology | 2009

Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia

Elaine Coustan-Smith; Charles G. Mullighan; Mihaela Onciu; Frederick G. Behm; Susana C. Raimondi; Deqing Pei; Cheng Cheng; Xiaoping Su; Jeffrey E. Rubnitz; Giuseppe Basso; Andrea Biondi; Ching-Hon Pui; James R. Downing; Dario Campana

BACKGROUND About a fifth of children with acute T-lymphoblastic leukaemia (T-ALL) succumb to the disease, suggesting an unrecognised biological heterogeneity that might contribute to drug resistance. We postulated that T-ALL originating from early T-cell precursors (ETPs), a recently defined subset of thymocytes that retain stem-cell-like features, would respond poorly to lymphoid-cell-directed therapy. We studied leukaemic cells, collected at diagnosis, to identify cases with ETP features and determine their clinical outcome. METHODS Leukaemic cells from 239 patients with T-ALL enrolled at St Jude Childrens Research Hospital (n=139) and in the Italian national study Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP) ALL-2000 (n=100) were assessed by gene-expression profiling, flow cytometry, and single nucleotide polymorphism array analysis. Probabilities of survival and treatment failure were calculated for subgroups considered to have ETP-ALL or typical T-ALL. FINDINGS 30 patients (12.6%) had leukaemic lymphoblasts with an ETP-related gene-expression signature or its associated distinctive immunophenotype (CD1a(-), CD8(-), CD5(weak) with stem-cell or myeloid markers). Cases of ETP-ALL showed increased genomic instability, in terms of number and size of gene lesions, compared with those with typical T-ALL. Patients with this form of leukaemia had high risk of remission failure or haematological relapse (72% [95% CI 40-100] at 10 years vs 10% [4-16] at 10 years for patients with typical T-ALL treated at St Jude Childrens Research Hospital; and 57% [25-89] at 2 years vs 14% [6-22] at 2 years for patients treated in the AIEOP trial). INTERPRETATION ETP-ALL is a distinct, previously unrecognised, pathobiological entity that confers a poor prognosis with use of standard intensive chemotherapy. Its early recognition, by use of the gene expression and immunophenotypic criteria outlined here, is essential for the development of an effective clinical management strategy. FUNDING US National Cancer Institute, Cariplo Foundation, Citta della Speranza Foundation, Italian Association for Cancer Research (AIRC), Italian Ministry for University and Research, and American Lebanese Syrian Associated Charities (ALSAC).


Nature Genetics | 2013

The genomic landscape of hypodiploid acute lymphoblastic leukemia

Linda Holmfeldt; Lei Wei; Ernesto Diaz-Flores; Michael D. Walsh; Jinghui Zhang; Li Ding; Debbie Payne-Turner; Michelle L. Churchman; Anna Andersson; Shann Ching Chen; Kelly McCastlain; Jared Becksfort; Jing Ma; Gang Wu; Samir N. Patel; Susan L. Heatley; Letha A. Phillips; Guangchun Song; John Easton; Matthew Parker; Xiang Chen; Michael Rusch; Kristy Boggs; Bhavin Vadodaria; Erin Hedlund; Christina D. Drenberg; Sharyn D. Baker; Deqing Pei; Cheng Cheng; Robert Huether

The genetic basis of hypodiploid acute lymphoblastic leukemia (ALL), a subtype of ALL characterized by aneuploidy and poor outcome, is unknown. Genomic profiling of 124 hypodiploid ALL cases, including whole-genome and exome sequencing of 40 cases, identified two subtypes that differ in the severity of aneuploidy, transcriptional profiles and submicroscopic genetic alterations. Near-haploid ALL with 24–31 chromosomes harbor alterations targeting receptor tyrosine kinase signaling and Ras signaling (71%) and the lymphoid transcription factor gene IKZF3 (encoding AIOLOS; 13%). In contrast, low-hypodiploid ALL with 32–39 chromosomes are characterized by alterations in TP53 (91.2%) that are commonly present in nontumor cells, IKZF2 (encoding HELIOS; 53%) and RB1 (41%). Both near-haploid and low-hypodiploid leukemic cells show activation of Ras-signaling and phosphoinositide 3-kinase (PI3K)-signaling pathways and are sensitive to PI3K inhibitors, indicating that these drugs should be explored as a new therapeutic strategy for this aggressive form of leukemia.


Journal of Clinical Oncology | 2009

Germline Genetic Variation in an Organic Anion Transporter Polypeptide Associated With Methotrexate Pharmacokinetics and Clinical Effects

Lisa R. Treviño; Noriko Shimasaki; Wenjian Yang; John C. Panetta; Cheng Cheng; Deqing Pei; Diana Chan; Alex Sparreboom; Kathleen M. Giacomini; Ching-Hon Pui; William E. Evans; Mary V. Relling

PURPOSE Methotrexate plasma concentration is related to its clinical effects. Our aim was to identify the genetic basis of interindividual variability in methotrexate pharmacokinetics in children with newly diagnosed acute lymphoblastic leukemia (ALL). PATIENTS AND METHODS We performed a genome-wide analysis of 500,568 germline single-nucleotide polymorphisms (SNPs) to identify how inheritance affects methotrexate plasma disposition among 434 children with ALL who received 3,014 courses of methotrexate at 2 to 5 g/m(2). SNPs were validated in an independent cohort of 206 patients. RESULTS Adjusting for age, race, sex, and methotrexate regimen, the most significant associations were with SNPs in the organic anion transporter polypeptide, SLCO1B1. Two SNPs in SLCO1B1, rs11045879 (P = 1.7 x 10(-10)) and rs4149081 (P = 1.7 x 10(-9)), were in linkage disequilibrium (LD) with each other (r(2) = 1) and with a functional polymorphism in SLCO1B1, T521C (rs4149056; r(2) > 0.84). rs11045879 and rs4149081 were validated in an independent cohort of 206 patients (P = .018 and P = .017), as were other SLCO1B1 SNPs residing in different LD blocks. SNPs in SLCO1B1 were also associated with GI toxicity (odds ratio, 15.3 to 16.4; P = .03 to .004). CONCLUSION A genome-wide interrogation identified inherited variations in a plausible, yet heretofore low-priority candidate gene, SLCO1B1, as important determinants of methotrexates pharmacokinetics and clinical effects.


JAMA | 2009

Genome-wide interrogation of germline genetic variation associated with treatment response in childhood acute lymphoblastic leukemia.

Jun Yang; Cheng Cheng; Wenjian Yang; Deqing Pei; Xueyuan Cao; Yiping Fan; Stanley Pounds; Geoffrey Neale; Lisa R. Treviño; Deborah L. French; Dario Campana; James R. Downing; William E. Evans; Ching-Hon Pui; Meenakshi Devidas; W P Bowman; Bruce M. Camitta; Cheryl L. Willman; Stella M. Davies; Michael J. Borowitz; William L. Carroll; Stephen P. Hunger; Mary V. Relling

CONTEXT Pediatric acute lymphoblastic leukemia (ALL) is the prototype for a drug-responsive malignancy. Although cure rates exceed 80%, considerable unexplained interindividual variability exists in treatment response. OBJECTIVES To assess the contribution of inherited genetic variation to therapy response and to identify germline single-nucleotide polymorphisms (SNPs) associated with risk of minimal residual disease (MRD) after remission induction chemotherapy. DESIGN, SETTING, AND PATIENTS Genome-wide interrogation of 476,796 germline SNPs to identify genotypes that were associated with MRD in 2 independent cohorts of children with newly diagnosed ALL: 318 patients in St Jude Total Therapy protocols XIIIB and XV and 169 patients in Childrens Oncology Group trial P9906. Patients were enrolled between 1994 and 2006 and last follow-up was in 2006. MAIN OUTCOME MEASURES Minimal residual disease at the end of induction therapy, measured by flow cytometry. RESULTS There were 102 SNPs associated with MRD in both cohorts (median odds ratio, 2.18; P < or = .0125), including 5 SNPs in the interleukin 15 (IL15) gene. Of these 102 SNPs, 21 were also associated with hematologic relapse (P < .05). Of 102 SNPs, 21 were also associated with antileukemic drug disposition, generally linking MRD eradication with greater drug exposure. In total, 63 of 102 SNPs were associated with early response, relapse, or drug disposition. CONCLUSION Host genetic variations are associated with treatment response for childhood ALL, with polymorphisms related to leukemia cell biology and host drug disposition associated with lower risk of residual disease.


Clinical Pharmacology & Therapeutics | 2009

Genetic polymorphism of inosine triphosphate pyrophosphatase is a determinant of mercaptopurine metabolism and toxicity during treatment for acute lymphoblastic leukemia.

Gabriele Stocco; Meyling Cheok; Kristine R. Crews; Dervieux T; Deborah L. French; Deqing Pei; Wenjian Yang; Cheng Cheng; Ching-Hon Pui; Mary V. Relling; William E. Evans

The influence of genetic polymorphism in inosine triphosphate pyrophosphatase (ITPA) on thiopurine‐induced adverse events has not been investigated in the context of combination chemotherapy for acute lymphoblastic leukemia (ALL). This study investigated the effects of a common ITPA variant allele (rs41320251) on mercaptopurine metabolism and toxicity during treatment of children with ALL. Significantly higher concentrations of methyl mercaptopurine nucleotides were found in patients with the nonfunctional ITPA allele. Moreover, there was a significantly higher probability of severe febrile neutropenia in patients with a variant ITPA allele among patients whose dose of mercaptopurine had been adjusted for TPMT genotype. In a cohort of patients whose mercaptopurine dose was not adjusted for TPMT phenotype, the TPMT genotype had a greater effect than the ITPA genotype. In conclusion, genetic polymorphism of ITPA is a significant determinant of mercaptopurine metabolism and of severe febrile neutropenia, after combination chemotherapy for ALL in which mercaptopurine doses are individualized on the basis of TPMT genotype.


Clinical Pharmacology & Therapeutics | 2003

A pharmacogenetic study of uridine diphosphate-glucuronosyltransferase 2B7 in patients receiving morphine

Michael Sawyer; Federico Innocenti; Soma Das; Cheng Cheng; Jacqueline Ramírez; Friedl H. Pantle‐Fisher; Constance Wright; Judith Badner; Deqing Pei; James M. Boyett; Edwin H. Cook; Mark J. Ratain

We investigated the variation in the uridine diphosphate–glucuronosyltransferase 2B7 (UGT2B7) gene in patients receiving patient‐controlled analgesia with morphine. UGT2B7 was sequenced in phenotypic extremes (n = 12) of the distribution of morphine‐6‐glucuronide/morphine plasma ratios. A new −161C/T promoter variant was in complete linkage disequilibrium with the 802C/T variant and was more frequent in low glucuronidators (P = .039). Both variants were genotyped in all patients (n = 86), and complete linkage disequilibrium was confirmed. Trend analysis showed reduced morphine‐6‐glucuronide/morphine ratios in patients with T/T, C/T, and C/C genotypes (T/T > C/T > C/C) (P = .031). Morphine levels were lower in T/T patients (median, 18 ng/mL [range, 18–1490 ng/mL]) as compared with C/T and C/C patients combined (median, 66 ng/m; range, 18–3995 ng/mL) (P = .04). Morphine‐6‐glucuronide and morphine‐3‐glucuronide concentrations were significantly lower in C/C patients (median, 18 ng/mL; range, 0–66 ng/mL; and median, 152 ng/mL; range, 30–434 ng/mL; respectively) compared with C/T and T/T patients combined (median, 43 ng/mL; range, 0–193 ng/mL; and median, 242 ng/mL; range, 33–1381 ng/mL; respectively) (P = .045 and P = .004, respectively). Interindividual differences in morphine glucuronidation may be the result of genetic variation in UGT2B7, and further studies are indicated.


Blood | 2011

Pharmacokinetic, pharmacodynamic, and pharmacogenetic determinants of osteonecrosis in children with acute lymphoblastic leukemia.

Jitesh D. Kawedia; Sue C. Kaste; Deqing Pei; John C. Panetta; Xiangjun Cai; Cheng Cheng; Geoffrey Neale; Scott C. Howard; William E. Evans; Ching-Hon Pui; Mary V. Relling

Osteonecrosis is a severe glucocorticoid-induced complication of acute lymphoblastic leukemia treatment. We prospectively screened children (n = 364) with magnetic resonance imaging of hips and knees, regardless of symptoms; the cumulative incidence of any (grade 1-4) versus symptomatic (grade 2-4) osteonecrosis was 71.8% versus 17.6%, respectively. We investigated whether age, race, sex, acute lymphoblastic leukemia treatment arm, body mass, serum lipids, albumin and cortisol levels, dexamethasone pharmacokinetics, and genome-wide germline genetic polymorphisms were associated with symptomatic osteonecrosis. Age more than 10 years (odds ratio, = 4.85; 95% confidence interval, 2.5-9.2; P = .00001) and more intensive treatment (odds ratio = 2.5; 95% confidence interval, 1.2-4.9; P = .011) were risk factors and included as covariates in all analyses. Lower albumin (P = .05) and elevated cholesterol (P = .02) associated with symptomatic osteonecrosis, and severe (grade 3 or 4) osteonecrosis was linked to poor dexamethasone clearance (P = .0005). Adjusting for clinical features, polymorphisms of ACP1 (eg, rs12714403, P = 1.9 × 10(-6), odds ratio = 5.6; 95% confidence interval, 2.7-11.3), which regulates lipid levels and osteoblast differentiation, were associated with risk of osteonecrosis as well as with lower albumin and higher cholesterol. Overall, older age, lower albumin, higher lipid levels, and dexamethasone exposure were associated with osteonecrosis and may be linked by inherited genomic variation.


Nature Genetics | 2015

The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias.

Anna Andersson; Jing Ma; Jianmin Wang; Xiang Chen; Amanda Larson Gedman; Jinjun Dang; Joy Nakitandwe; Linda Holmfeldt; Matthew A. Parker; John Easton; Robert Huether; Richard W. Kriwacki; Michael Rusch; Gang Wu; Yongjin Li; Heather L. Mulder; Susana C. Raimondi; Stanley Pounds; Guolian Kang; Lei Shi; Jared Becksfort; Pankaj Gupta; Debbie Payne-Turner; Bhavin Vadodaria; Kristy Boggs; Donald Yergeau; Jayanthi Manne; Guangchun Song; Michael Edmonson; Panduka Nagahawatte

Infant acute lymphoblastic leukemia (ALL) with MLL rearrangements (MLL-R) represents a distinct leukemia with a poor prognosis. To define its mutational landscape, we performed whole-genome, exome, RNA and targeted DNA sequencing on 65 infants (47 MLL-R and 18 non–MLL-R cases) and 20 older children (MLL-R cases) with leukemia. Our data show that infant MLL-R ALL has one of the lowest frequencies of somatic mutations of any sequenced cancer, with the predominant leukemic clone carrying a mean of 1.3 non-silent mutations. Despite this paucity of mutations, we detected activating mutations in kinase-PI3K-RAS signaling pathway components in 47% of cases. Surprisingly, these mutations were often subclonal and were frequently lost at relapse. In contrast to infant cases, MLL-R leukemia in older children had more somatic mutations (mean of 6.5 mutations/case versus 1.3 mutations/case, P = 7.15 × 10−5) and had frequent mutations (45%) in epigenetic regulators, a category of genes that, with the exception of MLL, was rarely mutated in infant MLL-R ALL.

Collaboration


Dive into the Deqing Pei's collaboration.

Top Co-Authors

Avatar

Cheng Cheng

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary V. Relling

University Of Tennessee System

View shared research outputs
Top Co-Authors

Avatar

William E. Evans

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Sima Jeha

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenjian Yang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jeffrey E. Rubnitz

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge