167 results on '"John K. Choi"'
Search Results
2. Author Correction: Proteasome inhibition targets the KMT2A transcriptional complex in acute lymphoblastic leukemia
- Author
-
Jennifer L. Kamens, Stephanie Nance, Cary Koss, Beisi Xu, Anitria Cotton, Jeannie W. Lam, Elizabeth A. R. Garfinkle, Pratima Nallagatla, Amelia M. R. Smith, Sharnise Mitchell, Jing Ma, Duane Currier, William C. Wright, Kanisha Kavdia, Vishwajeeth R. Pagala, Wonil Kim, LaShanale M. Wallace, Ji-Hoon Cho, Yiping Fan, Aman Seth, Nathaniel Twarog, John K. Choi, Esther A. Obeng, Mark E. Hatley, Monika L. Metzger, Hiroto Inaba, Sima Jeha, Jeffrey E. Rubnitz, Junmin Peng, Taosheng Chen, Anang A. Shelat, R. Kiplin Guy, and Tanja A. Gruber
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
3. Treatment response and outcome of children with T-cell acute lymphoblastic leukemia expressing the gamma-delta T-cell receptor
- Author
-
Ching-Hon Pui, Deqing Pei, Cheng Cheng, Suzanne L. Tomchuck, Scarlett N. Evans, Hiroto Inaba, Sima Jeha, Susana C. Raimondi, John K. Choi, Paul G. Thomas, and Mari Hashitate Dallas
- Subjects
γδ t cells ,tcr repertoire ,γδ t-all ,pediatric t-all ,hct ,risk stratification ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
T-cell malignancies expressing the γδ T-cell receptor (TCR) are often associated with poor prognosis. Here, we determined the clinical outcome of pediatric patients with T-cell acute lymphoblastic leukemia (T-ALL) expressing the γδ TCR. Of 100 newly diagnosed T-ALL patients, 93 had γδ TCR analysis performed at diagnosis. Repertoire was evaluated by paired sequencing of the rearranged TCR. All patients received intensified chemotherapy and those with minimal residual disease (MRD) ≥ 1% on day 42–46 became candidates for hematopoietic cell transplantation. Of the 93 T-ALL patients, 12 (13%) had γδ T-ALL and 11 (12%) had early T-cell precursor (ETP) ALL. Compared to the remaining 70 T-ALL patients, the γδ T-ALL patients were more likely to have MRD ≥ 1% on day 15–19 (67% vs. 33%, P = 0.03) and day 42–49 (33% vs. 7%; P = 0.007) of remission induction. The 10-year overall survival for γδ T-ALL patients (66.7% ± 22.2%) were lower than that of T-ALL patients (93.3% ± 7.3%, P = 0.001). TCR analysis demonstrated a conserved clonotype. In conclusion, the data suggest that children with γδ T-ALL may have a poor response to remission induction, based on MRD levels and decreased survival than the other T-ALL patients, despite receiving risk-directed therapy.
- Published
- 2019
- Full Text
- View/download PDF
4. γδ T-Cell Acute Lymphoblastic Leukemia/Lymphoma: Discussion of Two Pediatric Cases and Its Distinction from Other Mature γδ T-Cell Malignancies
- Author
-
Eric X. Wei, Vasiliki Leventaki, John K. Choi, Susana C. Raimondi, Elizabeth M. Azzato, Sheila A. Shurtleff, Menchu G. Ong, Diana M. Veillon, James D. Cotelingam, and Rodney E. Shackelford
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Gamma delta (γδ) T-cell antigen receptor (TCR) expression and its related T-cell differentiation are not commonly reported in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL). Here we report two pediatric T-ALL cases and present their clinical features, histology, immunophenotypes, cytogenetics, and molecular diagnostic findings. The first patient is a two-year-old girl with leukocytosis, circulating lymphoblasts, and a cryptic insertion of a short-arm segment at 10p12 into the long-arm segment of 11q23 resulting in an MLL and AF10 fusion transcript, which may be the first reported in γδ T-ALL. She responded to the chemotherapy protocol poorly and had persistent diseases. Following an allogeneic bone marrow transplant, she went into remission. The second patient is an eleven-year-old boy with a normal white cell count, circulating blasts, and a normal karyotype, but without any immature cellular markers by flow cytometric analysis. He responded to the chemotherapy well and achieved a complete remission. These cases demonstrate the diverse phenotypic, cytogenetic, and molecular aspects of γδ T-ALL. Early T-precursor- (ETP-) ALL and their differential diagnosis from other mature γδ T-cell leukemia/lymphomas are also discussed.
- Published
- 2017
- Full Text
- View/download PDF
5. Laboratory Aspects of Minimal / Measurable Residual Disease Testing in B-Lymphoblastic Leukemia
- Author
-
John K. Choi and Paul E. Mead
- Subjects
Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,medicine.diagnostic_test ,B lymphoblastic leukemia ,business.industry ,Treatment outcome ,Biochemistry (medical) ,Clinical Biochemistry ,Disease ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Flow Cytometry ,medicine.disease ,Residual ,Minimal residual disease ,Immunophenotyping ,Flow cytometry ,Leukemia ,Internal medicine ,medicine ,Humans ,Child ,Laboratories ,business - Abstract
Minimal residual disease detection provides critical prognostic predictor of treatment outcome and is the standard of care for B lymphoblastic leukemia. Flow cytometry-based minimal residual disease detection is the most common test modality and has high sensitivity (0.01%) and a rapid turnaround time (24 hours). This article details the leukemia associated immunophenotype analysis approach for flow cytometry-based minimal residual disease detection used at St. Jude Children's Research Hospital and importance of using guide gates and back-gating.
- Published
- 2023
6. Pathologic, cytogenetic, and molecular features of acute myeloid leukemia with megakaryocytic differentiation: A report from the Children's Oncology Group
- Author
-
Karen M. Chisholm, Jenny Smith, Amy E. Heerema‐McKenney, John K. Choi, Rhonda E. Ries, Betsy A. Hirsch, Susana C. Raimondi, Yi‐Cheng Wang, Alice Dang, Todd A. Alonzo, Lillian Sung, Richard Aplenc, Alan S. Gamis, Soheil Meshinchi, and Samir B. Kahwash
- Subjects
Oncology ,Pediatrics, Perinatology and Child Health ,Hematology - Published
- 2023
7. Clinicopathologic and prognostic features of TdT-negative pediatric B-lymphoblastic leukemia
- Author
-
Cheng Cheng, Yinmei Zhou, Ching-Hon Pui, Matthew M. Klairmont, Tanja A. Gruber, Sima Jeha, John K. Choi, Hiroto Inaba, and Yiwei Liu
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Pathology ,Adolescent ,Context (language use) ,Gastroenterology ,Disease-Free Survival ,Article ,Immunophenotyping ,Pathology and Forensic Medicine ,Cytogenetics ,Leukocyte Count ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,DNA Nucleotidylexotransferase ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,White blood cell ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Child ,Gene Rearrangement ,Acute leukemia ,biology ,business.industry ,Infant ,Nuclear Proteins ,Histone-Lysine N-Methyltransferase ,Flow Cytometry ,Prognosis ,medicine.disease ,Neoplasm Proteins ,Leukemia ,030104 developmental biology ,KMT2A ,medicine.anatomical_structure ,Terminal deoxynucleotidyl transferase ,Child, Preschool ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Hyperdiploidy ,business ,Myeloid-Lymphoid Leukemia Protein ,Transcription Factors - Abstract
Little is known about B-lymphoblastic leukemia (B-ALL) that lacks expression of terminal deoxynucleotidyl transferase (TdT). To address this, we performed the largest study to date of TdT-negative B-ALL using data from St. Jude Total XV and XVI clinical trials. Compared to TdT-positive B-ALL (n = 896), TdT-negative B-ALL (n = 21) was associated with younger age (median, 1.4 versus 6.8 years, P
- Published
- 2021
8. The 5th edition of the World Health Organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms
- Author
-
Joseph D. Khoury, Eric Solary, Oussama Abla, Yassmine Akkari, Rita Alaggio, Jane F. Apperley, Rafael Bejar, Emilio Berti, Lambert Busque, John K. C. Chan, Weina Chen, Xueyan Chen, Wee-Joo Chng, John K. Choi, Isabel Colmenero, Sarah E. Coupland, Nicholas C. P. Cross, Daphne De Jong, M. Tarek Elghetany, Emiko Takahashi, Jean-Francois Emile, Judith Ferry, Linda Fogelstrand, Michaela Fontenay, Ulrich Germing, Sumeet Gujral, Torsten Haferlach, Claire Harrison, Jennelle C. Hodge, Shimin Hu, Joop H. Jansen, Rashmi Kanagal-Shamanna, Hagop M. Kantarjian, Christian P. Kratz, Xiao-Qiu Li, Megan S. Lim, Keith Loeb, Sanam Loghavi, Andrea Marcogliese, Soheil Meshinchi, Phillip Michaels, Kikkeri N. Naresh, Yasodha Natkunam, Reza Nejati, German Ott, Eric Padron, Keyur P. Patel, Nikhil Patkar, Jennifer Picarsic, Uwe Platzbecker, Irene Roberts, Anna Schuh, William Sewell, Reiner Siebert, Prashant Tembhare, Jeffrey Tyner, Srdan Verstovsek, Wei Wang, Brent Wood, Wenbin Xiao, Cecilia Yeung, Andreas Hochhaus, HAL UVSQ, Équipe, Département d'hématologie [Gustave Roussy], Institut Gustave Roussy (IGR), Service de pathologie [CHU Ambroise Paré], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Ambroise Paré [AP-HP], Université de Versailles Saint-Quentin-en-Yvelines - UFR Sciences de la santé Simone Veil (UVSQ Santé), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), and Open Access funding enabled and organized by Projekt DEAL.
- Subjects
[SDV.MHEP.HEM] Life Sciences [q-bio]/Human health and pathology/Hematology ,Cancer Research ,Haematological cancer ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Hematology ,World Health Organization ,Oncology ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Hematologic Neoplasms ,Diagnosis ,Humans ,Histiocytosis - Abstract
The upcoming 5th edition of the World Health Organization (WHO) Classification of Haematolymphoid Tumours is part of an effort to hierarchically catalogue human cancers arising in various organ systems within a single relational database. This paper summarizes the new WHO classification scheme for myeloid and histiocytic/dendritic neoplasms and provides an overview of the principles and rationale underpinning changes from the prior edition. The definition and diagnosis of disease types continues to be based on multiple clinicopathologic parameters, but with refinement of diagnostic criteria and emphasis on therapeutically and/or prognostically actionable biomarkers. While a genetic basis for defining diseases is sought where possible, the classification strives to keep practical worldwide applicability in perspective. The result is an enhanced, contemporary, evidence-based classification of myeloid and histiocytic/dendritic neoplasms, rooted in molecular biology and an organizational structure that permits future scalability as new discoveries continue to inexorably inform future editions.
- Published
- 2022
9. Bortezomib with standard chemotherapy for children with acute myeloid leukemia does not improve treatment outcomes: a report from the Children’s Oncology Group
- Author
-
Soheil Meshinchi, Betsy A. Hirsch, Samir B. Kahwash, Jessica A. Pollard, John E. Levine, Brian T. Fisher, Terzah M. Horton, Richard Aplenc, Lillian Sung, Susana C. Raimondi, Todd A. Alonzo, Lisa Eidenschink Brodersen, Alan S. Gamis, Michael R. Loken, Edward A. Kolb, John K. Choi, and Robert B. Gerbing
- Subjects
Acute Myeloid Leukemia ,Oncology ,medicine.medical_specialty ,Myeloid ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Disease-Free Survival ,law.invention ,Bortezomib ,Randomized controlled trial ,law ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Child ,neoplasms ,Chemotherapy ,business.industry ,Standard treatment ,Remission Induction ,Cytarabine ,Articles ,Hematology ,Reference Standards ,medicine.disease ,Intensive care unit ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,medicine.anatomical_structure ,business ,medicine.drug - Abstract
New therapeutic strategies are needed for pediatric acute myeloid leukemia (AML) to reduce disease recurrence and treatment-related morbidity. The Children’s Oncology Group Phase III AAML1031 trial tested whether the addition of bortezomib to standard chemotherapy improves survival in pediatric patients with newly diagnosed AML. AAML1031 randomized patients younger than 30 years of age with de novo AML to standard treatment with or without bortezomib. All patients received the identical chemotherapy backbone with either four intensive chemotherapy courses or three courses followed by allogeneic hematopoietic stem cell transplantation for high-risk patients. For those randomized to the intervention arm, bortezomib 1.3 mg/m2 was given on days 1, 4 and 8 of each chemotherapy course. For those randomized to the control arm, bortezomib was not administered. In total, 1,097 patients were randomized to standard chemotherapy (n=542) or standard chemotherapy with bortezomib (n=555). There was no difference in remission induction rate between the bortezomib and control treatment arms (89% vs. 91%, P=0.531). Bortezomib failed to improve 3-year event-free survival (44.8±4.5% vs. 47.0±4.5%, P=0.236) or overall survival (63.6±4.5 vs. 67.2±4.3, P=0.356) compared with the control arm. However, bortezomib was associated with significantly more peripheral neuropathy (P=0.006) and intensive care unit admissions (P=0.025) during the first course. The addition of bortezomib to standard chemotherapy increased toxicity but did not improve survival. These data do not support the addition of bortezomib to standard chemotherapy in children with de novo AML. (Trial registered at clinicaltrials.gov NCT01371981; https://www.cancer.gov/clinicaltrials/ NCT01371981).
- Published
- 2020
10. Cytarabine dose reduction in patients with low-risk acute myeloid leukemia: A report from the Children's Oncology Group
- Author
-
Alan S. Gamis, Robert B. Gerbing, Betsy A. Hirsch, Richard Aplenc, John K. Choi, Lillian Sung, Michael R. Loken, Soheil Meshinchi, Kelly D. Getz, Samir B. Kahwash, E. Anders Kolb, Todd A. Alonzo, Susana C. Raimondi, and Lisa Eidenschink Brodersen
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Disease-Free Survival ,Article ,Recurrence ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Child ,Chemotherapy ,Drug Tapering ,business.industry ,Pediatric acute myeloid leukemia ,Hazard ratio ,Remission Induction ,Cytarabine ,Myeloid leukemia ,Hematology ,Prognosis ,Minimal residual disease ,Confidence interval ,Leukemia, Myeloid, Acute ,Pediatrics, Perinatology and Child Health ,Dose reduction ,business ,medicine.drug - Abstract
Background The optimal number of chemotherapy courses for low-risk (LR) pediatric acute myeloid leukemia (AML) is not known. Objective To compare outcomes for four (21.6 g/m2 cytarabine) versus five (45.6 g/m2 cytarabine) chemotherapy courses for LR-AML using data from Children's Oncology Group (COG) AAML0531 and AAML1031. Methods We compared relapse risk (RR), disease-free survival (DFS), and overall survival (OS), and the differential impact in LR subgroups for patients receiving four versus five chemotherapy courses. Cox (OS and DFS) and risk (RR) regressions were used to estimate hazard ratios (HR) to compare outcomes. Results A total of 923 LR-AML patients were included; 21% received five courses. Overall, LR-AML patients who received four courses had higher RR (40.9% vs. 31.4%; HR = 1.40, 95% confidence interval [CI]: 1.06-1.85), and worse DFS (56.0% vs. 67.0%; HR = 1.45, 95% CI: 1.10-1.91). There was a similar decrement in OS though it was not statistically significant (77.0% vs. 83.5%; HR = 1.45, 95% CI: 0.97-2.17). Stratified analyses revealed the detrimental effects of cytarabine dose de-escalation to be most pronounced in the LR-AML subgroup with uninformative cytogenetic/molecular features who were minimal residual disease (MRD) negative after the first induction course (EOI1). The absolute decrease in DFS with four courses for patients with favorable cytogenetic/molecular features and positive MRD was similar to that observed for patients with uninformative cytogenetic/molecular features and negative MRD at EOI1, though not statistically significant. Conclusions Our results support de-escalation of cytarabine exposure through the elimination of a fifth chemotherapy course only for LR-AML patients who have both favorable cytogenetic/molecular features and negative MRD after the first induction cycle.
- Published
- 2021
11. Clofarabine Can Replace Anthracyclines and Etoposide in Remission Induction Therapy for Childhood Acute Myeloid Leukemia: The AML08 Multicenter, Randomized Phase III Trial
- Author
-
Jeffrey W. Taub, Susana C. Raimondi, Deborah Schiff, Lei Wang, Jeffery Klco, Jeffrey E. Rubnitz, Kenneth Heym, Barbara A. Degar, Ching-Hon Pui, Elaine Coustan-Smith, Allen Eng Juh Yeoh, Hiroto Inaba, Norman J. Lacayo, John K. Choi, Brandon M. Triplett, Raul C. Ribeiro, Stanley Pounds, and Jennifer L. McNeer
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Young Adult ,Internal medicine ,Remission Induction Therapy ,medicine ,Humans ,Clofarabine ,Anthracyclines ,Child ,Etoposide ,business.industry ,Remission Induction ,Childhood Acute Myeloid Leukemia ,Infant, Newborn ,Cytarabine ,Infant ,Myeloid leukemia ,ORIGINAL REPORTS ,medicine.disease ,Clinical trial ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Child, Preschool ,Female ,Arabinonucleosides ,business ,medicine.drug - Abstract
PURPOSE To identify effective and less toxic therapy for children with acute myeloid leukemia, we introduced clofarabine into the first course of remission induction to reduce exposure to daunorubicin and etoposide. PATIENTS AND METHODS From 2008 through 2017, 285 patients were enrolled at eight centers; 262 were randomly assigned to receive clofarabine and cytarabine (Clo+AraC, n = 129) or high-dose cytarabine, daunorubicin, and etoposide (HD-ADE, n = 133) as induction I. Induction II consisted of low-dose ADE given alone or combined with sorafenib or vorinostat. Consolidation therapy comprised two or three additional courses of chemotherapy or hematopoietic cell transplantation. Genetic abnormalities and the level of minimal residual disease (MRD) at day 22 of initial remission induction determined final risk classification. The primary end point was MRD at day 22. RESULTS Complete remission was induced after two courses of therapy in 263 (92.3%) of the 285 patients; induction failures included four early deaths and 15 cases of resistant leukemia. Day 22 MRD was positive in 57 of 121 randomly assigned evaluable patients (47%) who received Clo+AraC and 42 of 121 patients (35%) who received HD-ADE (odds ratio, 1.86; 95% CI, 1.03 to 3.41; P = .04). Despite this result, the 3-year event-free survival rate (52.9% [44.6% to 62.8%] for Clo+AraC v 52.4% [44.0% to 62.4%] for HD-ADE, P = .94) and overall survival rate (74.8% [67.1% to 83.3%] for Clo+AraC v 64.6% [56.2% to 74.2%] for HD-ADE, P = .1) did not differ significantly across the two arms. CONCLUSION Our findings suggest that the use of clofarabine with cytarabine during remission induction might reduce the need for anthracycline and etoposide in pediatric patients with acute myeloid leukemia and may reduce rates of cardiomyopathy and treatment-related cancer.
- Published
- 2019
12. Integrative network analysis reveals USP7 haploinsufficiency inhibits E-protein activity in pediatric T-lineage acute lymphoblastic leukemia (T-ALL)
- Author
-
James R. Downing, Li Dong, Bridget Shaner, Shaina N. Porter, Junmin Peng, Laura J. Janke, Anthony A. High, Wei Gu, John K. Choi, Timothy I. Shaw, Deqing Pei, Jinghui Zhang, Cheng Cheng, Xiaotu Ma, Jiyang Yu, Liqing Tian, Yu Liu, Vishwajeeth Pagala, Shondra M. Pruett-Miller, Kanisha Kavdia, A. Thomas Look, Chenxi Qian, Bensheng Ju, and John Easton
- Subjects
Transcriptional Activation ,Somatic cell ,Science ,Haploinsufficiency ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Pediatrics ,Article ,law.invention ,Gene regulatory networks ,Transcriptome ,Ubiquitin-Specific Peptidase 7 ,law ,Cell Line, Tumor ,Humans ,Cell Lineage ,Gene ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Cell Proliferation ,Multidisciplinary ,Acute lymphocytic leukaemia ,Oncogene ,Cell growth ,Gene Expression Regulation, Leukemic ,Oncogenes ,Computational biology and bioinformatics ,Cancer research ,Suppressor ,Medicine ,CRISPR-Cas Systems ,TAL1 - Abstract
USP7, which encodes a deubiquitylating enzyme, is among the most frequently mutated genes in pediatric T-ALL, with somatic heterozygous loss-of-function mutations (haploinsufficiency) predominantly affecting the subgroup that has aberrant TAL1 oncogene activation. Network analysis of > 200 T-ALL transcriptomes linked USP7 haploinsufficiency with decreased activities of E-proteins. E-proteins are also negatively regulated by TAL1, leading to concerted down-regulation of E-protein target genes involved in T-cell development. In T-ALL cell lines, we showed the physical interaction of USP7 with E-proteins and TAL1 by mass spectrometry and ChIP-seq. Haploinsufficient but not complete CRISPR knock-out of USP7 showed accelerated cell growth and validated transcriptional down-regulation of E-protein targets. Our study unveiled the synergistic effect of USP7 haploinsufficiency with aberrant TAL1 activation on T-ALL, implicating USP7 as a haploinsufficient tumor suppressor in T-ALL. Our findings caution against a universal oncogene designation for USP7 while emphasizing the dosage-dependent consequences of USP7 inhibitors currently under development as potential cancer therapeutics.
- Published
- 2020
13. Acute erythroid leukemia is enriched in NUP98 fusions: a report from the Children's Oncology Group
- Author
-
Samir B. Kahwash, John K. Choi, Alan S. Gamis, Richard Aplenc, Lillian Sung, Jenny L. Smith, Soheil Meshinchi, Karen M. Chisholm, Betsy A. Hirsch, Amy Heerema-McKenney, Yi-Cheng Wang, Susana C. Raimondi, Rhonda E. Ries, and Todd A. Alonzo
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Child ,Aged ,Myeloid Neoplasia ,Genetic heterogeneity ,business.industry ,Myelodysplastic syndromes ,Acute erythroid leukemia ,Myeloid leukemia ,Karyotype ,Hematology ,medicine.disease ,Nuclear Pore Complex Proteins ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Leukemia, Erythroblastic, Acute ,Hematopathology ,business - Abstract
Acute erythroid leukemia (AEL) is a rare subtype of acute myeloid leukemia (AML) primarily affecting older adults and was previously classified into erythroid/myeloid and pure erythroid subtypes. In this pediatric AEL study, we evaluated morphologic, immunophenotypic, cytogenetic, molecular, and clinical data of 24 (1.2%) cases from all cases undergoing central pathology review in Children’s Oncology Group trials AAML0531 and AAML1031. Of 24 cases, 5 had a pure erythroid phenotype, and 19 had an erythroid/myeloid phenotype. NUP98 fusions were highly enriched in patients with AEL, occurring in 7 of 22 cases for which molecular data were available (31.8% vs 6.7% in other AML subtypes). Of 5 cases of pure erythroid leukemias (PELs), 3 had NUP98 fusions, and 4 had complex karyotypes. Erythroid/myeloid leukemias were reclassified by using the 2017 World Health Organization hematopathology classification as: myelodysplastic syndrome (MDS) with excess blasts-1 (n = 3), MDS with excess blasts-2 (n = 7), AML (nonerythroid, n = 5), and unknown MDS/AML (n = 4); the 5 cases of nonerythroid AML included 1 with an NUP98–NSD1 fusion, 2 with myelodysplasia-related changes, and 1 with a complex karyotype. Three cases of MDS with excess blasts-2 also had NUP98 rearrangements. WT1 mutations were present in 5 of 14 cases, all erythroid/myeloid leukemia. Outcomes assessment revealed statistically poorer overall survival (5-year, 20% ± 36% vs 66% ± 23%; P = .004) and event-free survival (5-year, 20% ± 36% vs 46% ± 23%; P = .019) for those with PEL than those with erythroid/myeloid leukemia. Our study supports that AEL is a morphologically and genetically heterogeneous entity that is enriched in NUP98 fusions, with the pure erythroid subtype associated with particularly adverse outcomes.
- Published
- 2020
14. The Comparative Sensitivity of Immunohistochemical Markers of Megakaryocytic Differentiation in Acute Megakaryoblastic Leukemia
- Author
-
Matthew M. Klairmont, John K. Choi, Nour Yadak, and Deepthi Hoskoppal
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cellular differentiation ,Sensitivity and Specificity ,03 medical and health sciences ,Acute megakaryoblastic leukemia ,0302 clinical medicine ,Von Willebrand factor ,Bone Marrow ,Leukemia, Megakaryoblastic, Acute ,hemic and lymphatic diseases ,von Willebrand Factor ,Biopsy ,medicine ,Humans ,Child ,biology ,medicine.diagnostic_test ,business.industry ,Integrin beta3 ,Cell Differentiation ,General Medicine ,medicine.disease ,Immunohistochemistry ,Staining ,Leukemia ,030104 developmental biology ,Platelet Glycoprotein GPIb-IX Complex ,030220 oncology & carcinogenesis ,biology.protein ,Biopsy, Large-Core Needle ,Reagent Kits, Diagnostic ,business ,Megakaryocytes ,CD61 ,Biomarkers - Abstract
Objectives Immunohistochemistry (IHC) staining of core biopsy sections often plays an essential role in the diagnosis of acute megakaryoblastic leukemia (AMKL). The goal of this study was to define the relative sensitivities of commonly used stains for markers of megakaryocytic differentiation. Methods The sensitivities of IHC stains for CD42b, CD61, and von Willebrand factor (vWF) were compared in 32 cases of pediatric AMKL. Results The sensitivities of CD42b, CD61, and vWF were 90.6%, 78.1% and 62.5%, respectively. When CD42b and CD61 were used together, the combined sensitivity increased to 93.6%. There were no cases in which vWF was positive when both CD42b and CD61 were negative. Conclusions CD42b can reliably be used as a solitary first-line marker for blasts of megakaryocytic lineage, whereas CD61 may be reserved for infrequent cases that are CD42b negative. There is no role for the routine use of vWF when CD42b and CD61 are available.
- Published
- 2018
15. NUP98-BPTF gene fusion identified in primary refractory acute megakaryoblastic leukemia of infancy
- Author
-
Mathieu Roussy, Mélanie Bilodeau, Loubna Jouan, Pauline Tibout, Louise Laramée, Emmanuelle Lemyre, France Léveillé, Frédérique Tihy, Sophie Cardin, Camille Sauvageau, Françoise Couture, Isabelle Louis, Aurélien Choblet, Natalie Patey, Patrick Gendron, Michel Duval, Pierre Teira, Josée Hébert, Brian T. Wilhelm, John K. Choi, Tanja A. Gruber, Henrique Bittencourt, and Sonia Cellot
- Subjects
Male ,0301 basic medicine ,Cancer Research ,RNA Splicing ,Nerve Tissue Proteins ,Chimeric gene ,Biology ,Fusion gene ,03 medical and health sciences ,Acute megakaryoblastic leukemia ,Leukemia, Megakaryoblastic, Acute ,Genetics ,medicine ,Humans ,Clofarabine ,Transcription factor ,Gene Expression Profiling ,Infant ,Antigens, Nuclear ,medicine.disease ,Chromatin ,Nuclear Pore Complex Proteins ,Gene expression profiling ,030104 developmental biology ,Drug Resistance, Neoplasm ,Karyotyping ,RNA splicing ,Disease Progression ,Cancer research ,Transcription Factors ,medicine.drug - Abstract
The advent of large scale genomic sequencing technologies significantly improved the molecular classification of acute megakaryoblastic leukaemia (AMKL). AMKL represents a subset (∼10%) of high fatality pediatric acute myeloid leukemia (AML). Recurrent and mutually exclusive chimeric gene fusions associated with pediatric AMKL are found in 60%-70% of cases and include RBM15-MKL1, CBFA2T3-GLIS2, NUP98-KDM5A and MLL rearrangements. In addition, another 4% of AMKL harbor NUP98 rearrangements (NUP98r), with yet undetermined fusion partners. We report a novel NUP98-BPTF fusion in an infant presenting with primary refractory AMKL. In this NUP98r, the C-terminal chromatin recognition modules of BPTF, a core subunit of the NURF (nucleosome remodeling factor) ATP-dependent chromatin-remodeling complex, are fused to the N-terminal moiety of NUP98, creating an in frame NUP98-BPTF fusion, with structural homology to NUP98-KDM5A. The leukemic blasts expressed two NUP98-BPTF splicing variants, containing one or two tandemly spaced PHD chromatin reader domains. Our study also identified an unreported wild type BPTF splicing variant encoding for 2 PHD domains, detected both in normal cord blood CD34+ cells and in leukemic blasts, as with the fly BPTF homolog, Nurf301. Disease course was marked by rapid progression and primary chemoresistance, with ultimately significant tumor burden reduction following treatment with a clofarabine containing regimen. In sum, we report 2 novel NUP98-BPTF fusion isoforms that contribute to refine the NUP98r subgroup of pediatric AMKL. Multicenter clinical trials are critically required to determine the frequency of this fusion in AMKL patients and explore innovative treatment strategies for a disease still plagued with poor outcomes.
- Published
- 2018
16. #75: Microbial Cell-free DNA Sequencing for Prediction of Culture-Negative Infection Events in Children with Cancer
- Author
-
Joshua Wolf, Desiree Hollemon, Yilun Sun, Gabriela Maron, Li Tang, Kim J Allison, Gonzalez-Pena, Yuki Inaba, Christina Kohler, John K. Choi, Amanda griffen, Asim A. Ahmed, Elisa Margolis, Jeffrey E. Rubnitz, Charles Gawad, Abigail Brenner, and Kathryn Goggin
- Subjects
biology ,business.industry ,Cancer ,General Medicine ,Helicobacter pylori ,biology.organism_classification ,medicine.disease ,Microbiology ,Sepsis ,Leukemia ,Infectious Diseases ,Cell-free fetal DNA ,Pediatrics, Perinatology and Child Health ,medicine ,Prevotella ,Neisseria ,Fusobacterium nucleatum ,business - Abstract
Background New culture-independent diagnostics are being investigated for diagnosis and prediction of infection. One method is microbial cell-free DNA sequencing (mcfDNA-seq), which can detect a wide range of pathogens directly from plasma. Immunocompromised children who develop febrile neutropenia (FN) without documented bloodstream infection (BSI) may have an undiagnosed bacterial infection, but identification of this subset is difficult. The percentage of episodes of FN caused by bacterial pathogens that did not grow in culture is unknown, as is the relative contribution of other specific etiologies such as viruses, fungi, immunotherapies, and immune reconstitution or graft vs. host disease. The value of mcfDNA-seq in FN patients is also unknown. As part of a larger study evaluating the ability of mcfDNA-seq to predict BSI in a cohort of pediatric patients with relapsed or refractory leukemia, we analyzed mcfDNA-seq results in a sample of cases of FN for which the definitive etiology was unknown. Methods Eligible participants were Results mcfDNA-seq results were obtained from eight episodes of FN in seven patients. Five episodes occurred in participants awaiting engraftment after HCT and three were receiving chemotherapy only. All participants receiving chemotherapy were receiving antibacterial prophylaxis with vancomycin and ciprofloxacin, antifungal prophylaxis with micafungin or voriconazole, and PJP prophylaxis with TMP-SMX or pentamidine. No HCT recipients were receiving antibacterial prophylaxis, but all received PJP prophylaxis, antifungal prophylaxis or treatment, and antiviral prophylaxis or treatment. Of 8 FN episodes, 4 (50%) had a common bacterial pathogen identified by mcfDNA-seq on Day 0 (Table 1). In 2 (50%) of these cases, the same organism was also identified on Day –1, at a lower concentration. One fungal pathogen was identified prior to and at the onset of FN. A common bacterial pathogen was identified in 3/64 (5%) control samples from the study population. Culture-negative sepsis was the final diagnosis in one episode; in this case, Streptococcus mitis, an important cause of sepsis in neutropenic patients, was identified in both Day 0 and Day –1 samples. In another episode where E. coli was identified, antibiotics were discontinued after 48 hours, but the patient was re-admitted within 24 hours for recurrent FN. Conclusions In this sample of culture-negative FN episodes in pediatric patients with relapsed or refractory leukemia, mcfDNA-seq identified a common bacterial pathogen in 50% of cases. The same organism was identifiable on the day prior to FN in 50% of cases, suggesting that predictive testing might be feasible. More data regarding sensitivity and specificity of mcfDNA-seq to diagnose and predict FN are needed.
- Published
- 2021
17. Clofarabine-Based Chemotherapy for KMT2Ar Infantile Acute Lymphoblastic Leukemia
- Author
-
Dario Campana, Jennifer L. Pauley, Raul C. Ribeiro, Cheng Cheng, Deqing Pei, Ching-Hon Pui, Jeffrey E. Rubnitz, William E. Evans, Tanja A. Gruber, Hiroto Inaba, Susana C. Raimondi, Monika L. Metzger, John K. Choi, Mary V. Relling, Elaine Coustan-Smith, Hope D. Swanson, and Sima Jeha
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,Lymphoblastic Leukemia ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Internal medicine ,medicine ,Clofarabine ,business ,medicine.drug - Abstract
Rearrangements in KMT2A (KMT2Ar) are associated with pediatric, adult and therapy-induced acute leukemias. Infants with KMT2Ar acute lymphoblastic leukemia (ALL) have a poor prognosis, with an event-free-survival (EFS) of 33.6-36.9%. In the context of the remarkable improvements in the treatment of childhood ALL, the dismal outcome of infantile KMT2Ar ALL and the lack of any significant progress for more than two decades are striking . The St. Jude Total Therapy 16 study (the most recently reported study of a program for childhood ALL that begun in 1962) yielded a 5-year EFS of 88.2% overall. Total 16 enrolled all subtypes of newly diagnosed pediatric ALL patients including infants, with intensity of treatment adapted to presenting clinical and genomic features, and early treatment response as determined by minimal/measurable residual disease (MRD). KMT2Ar infants were treated on an intensified high-risk arm and received clofarabine in combination with cyclophosphamide and etoposide (CCE) at two points during treatment: Induction days 22-25 and Reinduction I. Infants who lacked KMT2Ar and KMT2Ar patients who were one year of age or older received the same risk-directed treatment plan given to all other patients enrolled on study. A total of 28 patients with KMT2Ar were enrolled on Total 16; the 19 patients > 1 year of age received standard-risk therapy, and the 9 patients < 1 year of age received high-risk therapy on the infant arm with CCE. The probabilities of 5-year EFS and overall survival in KMT2Ar patients > 1 year of age and those < 1 year of age were 73.3% vs. 44.4% (p=0.071) and 84.2% vs. 55.6% (p=0.060), respectively. Six of the nine infants were MRD-positive on Induction day 15 prior to CCE (MRD-positive range, 0.012% to 13.7%; median, 2.13%) with MRD negative status ( 1 year of age and 12.5% for those < 1 year of age (p=0.454). Five infants remain alive (four in CR1, one in CR2), while four expired in CR1. Three deaths were secondary to infection, including a multi-drug resistant soft tissue bacterial infection during Induction days 1-21, a respiratory syncytial virus pneumonia during Reinduction II, and a chronic parainfluenza 3 infection during Continuation weeks 70-101 that led to chronic pneumonitis and interstitial fibrosis. The fourth patient developed grade 5 pulmonary hypertension following induction, a complication potentially compounded by their presenting WBC count of 905 x 10 9/L and pulmonary leukostasis. A comparison of 3-year cumulative risk of selected major toxic effects of treatment revealed that high-risk infants had a lower incidence of asparaginase allergic reactions, osteonecrosis, hyperglycemia, and pancreatitis; in contrast, the incidence of fever and neutropenia, hepatic toxicity and seizures, was similar in high-risk patients regardless of age. Infants had a higher risk of thrombosis (46.7% vs. 23.1%, p 1 year of age that received one or more clofarabine-containing Reintensification chemotherapy cycles prior to hematopoietic stem cell transplant in first remission (CR1). This revealed a higher frequency of infections in infants, suggesting a greater susceptibility to this complication independent from clofarabine exposure (mean number of episodes, 2.39 vs. 1, p Disclosures Gruber: Kura Oncology: Consultancy. Coustan-Smith: Juno Therapeutics: Patents & Royalties; Nkarta Therapeutics: Current holder of individual stocks in a privately-held company; Medisix Therapeutics: Current holder of individual stocks in a privately-held company. Campana: Nkarta Therapeutics: Current holder of stock options in a privately-held company; Medisix Therapeutics: Current holder of stock options in a privately-held company; Juno: Other: patent licensing payments; Juno Therapeutics (a Bristol-Myers Squibb company),: Other: patents on methods for minimal residual disease detection.. Evans: St. Jude Children's Research Hospital, Emeritus Member (began Jan 2021): Ended employment in the past 24 months; BioSkryb, Inc.: Membership on an entity's Board of Directors or advisory committees; Princess Máxima Center for Pediatric Oncology, Scientific Advisory Board, Chair: Membership on an entity's Board of Directors or advisory committees. Pui: Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; Novartis: Other: Data Monitoring Committee.
- Published
- 2021
18. AML-430: Utility of End-of-Induction Bone Marrow Biopsy in APL Patients Treated with ATRA/ATO Regimen
- Author
-
Pankit Vachhani, Kathleen Purdy, Fady M. Mikhail, Sydney Dunn-Valadez, Diana Morlote, John K. Choi, Omer Jamy, Ravi Bhatia, Srilakshmi Bathini, Josh Oliver, Kelly N. Godby, Sravanti Rangaraju, Kimo Bachiashvili, Vishnu Reddy, Gaurav Goyal, Amitkumar Mehta, and Sarah Worth
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Anthracycline ,medicine.diagnostic_test ,business.industry ,Gemtuzumab ozogamicin ,Age at diagnosis ,Hematology ,Fixed dose ,Regimen ,medicine.anatomical_structure ,Internal medicine ,Biopsy ,medicine ,End of induction ,Bone marrow ,business ,neoplasms ,medicine.drug - Abstract
Background: Significant variations exist related to end-of-induction (EOI) practices in APL management. These include fixed dose/duration ATRA/ATO vs continuation of ATRA/ATO until hematologic complete remission (CR) and performance vs omission of EOI bone marrow biopsy (BMBx) to confirm CR. These variations arise from the original protocols (notably, APL0406), as well as ELN-2019 and NCCN-2020 guidelines. Objectives: To assess utility of EOI BMBx morphologic findings in APL patients treated with a 28-day ATRA/ATO induction regimen. Methods: Patients ≥ 18 years treated for APL with ATRA/ATO ± cytoreductive agent at a tertiary center from 12/2012 to 03/2020 were identified. Patients who received >30 doses of ATO or those who died in the first 4 weeks of treatment were excluded. Demographics, hematologic parameters, treatment details, and BMBx results were collected. Time-to-event endpoints were calculated from day 1 of ATO. Results: Sixty-one patients (42 low/intermediate-risk, 19 high-risk) were included in the study. Median age at diagnosis was 51 years (21–81 years); 47 (77%) were Caucasian. No low/intermediate-risk APL patients received gemtuzumab ozogamicin (GO); 2 received anthracycline. Five high-risk APL patients received GO; 10 received anthracycline. Median time-to-dose 28 ± 2 of ATO was 28 days (27–40). Day 28 hematologic parameters showed that 38 (62.3%) patients had ANC Conclusions: A 28-dose fixed ATO-containing ATRA/ATO regimen delivers excellent outcomes. Our data suggest that EOI-BMBx can be safely omitted, irrespective of hematologic parameters.
- Published
- 2021
19. Abstract CT146: Prognostic and therapeutic significance of leukemia subtypes in the context of risk-directed therapy based on minimal residual disease levels in pediatric acute lymphoblastic leukemia
- Author
-
Seth E. Karol, William E. Evans, Elaine Coustan-Smith, Raul C. Ribiero, Mary V. Relling, John K. Choi, Sima Jeha, Dario Campana, James R. Downing, Hiroto Inaba, Susana C. Raimondi, Kathryn G. Roberts, Cheng Cheng, Ching-Hon Pui, Tanja A. Gruber, Deqing Pei, Charles G. Mullighan, Jeffrey E. Rubnitz, and Jun J. Yang
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Lymphoblastic Leukemia ,Cancer ,Retrospective cohort study ,Context (language use) ,medicine.disease ,Single Center ,Minimal residual disease ,Leukemia ,Pediatric Acute Lymphoblastic Leukemia ,hemic and lymphatic diseases ,Internal medicine ,medicine ,business - Abstract
Determination of prognostic and therapeutic implications of novel leukemia subtypes in children with acute lymphoblastic leukemia (ALL) treated with contemporary minimal residual disease (MRD)-directed therapy can improve outcome. In this study, we evaluated the clinical impact of identification of the full genomic spectrum of leukemia subtypes and MRD assessment to guide risk-directed therapy. A retrospective cohort study was conducted in 598 consecutive patients enrolled on Total Therapy Study 16 in a single center from October 29, 2007 to March 26, 2017, with a median follow-up of 7 years. High-hyperdiploid and ETV6-RUNX1 ALL were provisionally classified to be low-risk; TCF3-PBX1, hypodiploid Citation Format: Sima Jeha, John K. Choi, Deqing Pei, Elaine Coustan-Smith, Hiroto Inaba, Jeffrey E. Rubnitz, Raul C. Ribiero, Tanja A. Gruber, Susana C. Raimondi, Seth E. Karol, Kathryn G. Roberts, Jun J. Yang, Cheng Cheng, James R. Downing, William E. Evans, Mary V. Relling, Dario Campana, Charles G. Mullighan, Ching-Hon Pui. Prognostic and therapeutic significance of leukemia subtypes in the context of risk-directed therapy based on minimal residual disease levels in pediatric acute lymphoblastic leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT146.
- Published
- 2021
20. CD30 Expression in Pediatric Neoplasms, Study of 585 Cases
- Author
-
John K. Choi, Jinjun Cheng, and Haiqing Zhu
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Myeloid ,CD30 ,Ki-1 Antigen ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Neoplasms ,hemic and lymphatic diseases ,Neuroblastoma ,medicine ,Humans ,Neoplasm ,Child ,Anaplastic large-cell lymphoma ,Retrospective Studies ,integumentary system ,business.industry ,Myeloid leukemia ,General Medicine ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Chondrosarcoma ,business ,Diffuse large B-cell lymphoma - Abstract
CD30 is a member of the tumor necrosis factor receptor superfamily, member 8 (TNFRSF8), and its normal expression is restricted to activated T and B cells. In tumor cells, CD30 expression is most commonly associated with lymphoid malignancies (Hodgkin and non-Hodgkin lymphomas) and is a therapeutic target using anti-CD30 antibody. CD30 expression has been reported also in mostly adult non-lymphoid malignancies, raising the possibility of CD30-targeted therapy for additional tumors. In this study, we examined the incidence of CD30 expression in 251 hematopoietic and 334 non-hematopoietic cases of pediatric tumors. As expected, strong and membranous CD30 staining was seen in anaplastic large cell lymphoma, classical Hodgkin lymphoma, and embryonal carcinoma while variable staining was seen in diffuse large B cell lymphoma. In addition, positive CD30 staining was also seen in cases of neuroblastoma (33 of 56), neoplasm with chondroid differentiation (8 of 25), myeloid neoplasms (11 of 120), hemangioma (2 of 12), and mature teratoma (1 of 11). In neuroblastoma, the CD30 expression was generally restricted to cells with ganglion differentiation; staining of ganglion cells was also seen in the one positive case of mature teratoma. In neoplasm with chondroid differentiation, the positive cases were chondrosarcoma (3 of 5), chondroblastic osteosarcoma (2 of 10), and chondroblastoma (3 of 7). In acute myeloid leukemia, the CD30 positive cases were more common in AML with monocytic differentiation but did not correlate with any specific molecular change. We conclude that CD30 expression in pediatric tumors is more general than anticipated and future studies are warranted to understand the biologic and therapeutic significances.
- Published
- 2017
21. BCL-W has a fundamental role in B cell survival and lymphomagenesis
- Author
-
Jerald Z. Gong, Annette S. Kim, Clare M. Adams, Christine M. Eischen, John K. Choi, and Ramkrishna Mitra
- Subjects
0301 basic medicine ,Cell type ,Apoptosis ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins c-myc ,Mice ,03 medical and health sciences ,medicine ,Animals ,Humans ,B-cell lymphoma ,B cell ,Mice, Knockout ,B-Lymphocytes ,Proteins ,General Medicine ,medicine.disease ,Burkitt Lymphoma ,3. Good health ,Lymphoma ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Cancer cell ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Apoptosis Regulatory Proteins ,Carcinogenesis ,Diffuse large B-cell lymphoma ,Research Article - Abstract
Compromised apoptotic signaling is a prerequisite for tumorigenesis. The design of effective therapies for cancer treatment depends on a comprehensive understanding of the mechanisms that govern cell survival. The antiapoptotic proteins of the BCL-2 family are key regulators of cell survival and are frequently overexpressed in malignancies, leading to increased cancer cell survival. Unlike BCL-2 and BCL-XL, the closest antiapoptotic relative BCL-W is required for spermatogenesis, but was considered dispensable for all other cell types. Here, however, we have exposed a critical role for BCL-W in B cell survival and lymphomagenesis. Loss of Bcl-w conferred sensitivity to growth factor deprivation–induced B cell apoptosis. Moreover, Bcl-w loss profoundly delayed MYC-mediated B cell lymphoma development due to increased MYC-induced B cell apoptosis. We also determined that MYC regulates BCL-W expression through its transcriptional regulation of specific miR. BCL-W expression was highly selected for in patient samples of Burkitt lymphoma (BL), with 88.5% expressing BCL-W. BCL-W knockdown in BL cell lines induced apoptosis, and its overexpression conferred resistance to BCL-2 family–targeting BH3 mimetics. Additionally, BCL-W was overexpressed in diffuse large B cell lymphoma and correlated with decreased patient survival. Collectively, our results reveal that BCL-W profoundly contributes to B cell lymphoma, and its expression could serve as a biomarker for diagnosis and aid in the development of better targeted therapies.
- Published
- 2017
22. γδ T-Cell Acute Lymphoblastic Leukemia/Lymphoma: Discussion of Two Pediatric Cases and Its Distinction from Other Mature γδ T-Cell Malignancies
- Author
-
Rodney E. Shackelford, Vasiliki Leventaki, Elizabeth M Azzato, Menchu Ong, Sheila A. Shurtleff, Diana M Veillon, James D. Cotelingam, Susana C. Raimondi, John K. Choi, and Eric X Wei
- Subjects
0301 basic medicine ,medicine.medical_specialty ,T cell ,education ,Case Report ,behavioral disciplines and activities ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Medicine ,Leukocytosis ,lcsh:RC633-647.5 ,business.industry ,Lymphoblast ,T-cell receptor ,Cytogenetics ,lcsh:Diseases of the blood and blood-forming organs ,General Medicine ,medicine.disease ,Lymphoma ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Fusion transcript ,030220 oncology & carcinogenesis ,Immunology ,medicine.symptom ,business - Abstract
Gamma delta (γδ) T-cell antigen receptor (TCR) expression and its related T-cell differentiation are not commonly reported in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL). Here we report two pediatric T-ALL cases and present their clinical features, histology, immunophenotypes, cytogenetics, and molecular diagnostic findings. The first patient is a two-year-old girl with leukocytosis, circulating lymphoblasts, and a cryptic insertion of a short-arm segment at 10p12 into the long-arm segment of 11q23 resulting in an MLL and AF10 fusion transcript, which may be the first reported in γδ T-ALL. She responded to the chemotherapy protocol poorly and had persistent diseases. Following an allogeneic bone marrow transplant, she went into remission. The second patient is an eleven-year-old boy with a normal white cell count, circulating blasts, and a normal karyotype, but without any immature cellular markers by flow cytometric analysis. He responded to the chemotherapy well and achieved a complete remission. These cases demonstrate the diverse phenotypic, cytogenetic, and molecular aspects of γδ T-ALL. Early T-precursor- (ETP-) ALL and their differential diagnosis from other mature γδ T-cell leukemia/lymphomas are also discussed.
- Published
- 2017
23. Germline Gain-of-Function JAK3 Mutation in Familial Chronic Lymphoproliferative Disorder of NK Cells
- Author
-
Kim E. Nichols, John K. Choi, Sara Lewis, Charnise Goodings-Harris, Marcin W. Wlodarski, Mihaela Onciu, Harry Lesmana, Sushree S. Sahoo, Marcela Popescu, and Clifford Takemoto
- Subjects
Severe combined immunodeficiency ,Mutation ,Large granular lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Germline ,Germline mutation ,medicine.anatomical_structure ,medicine ,Primary immunodeficiency ,T-cell prolymphocytic leukemia ,B cell - Abstract
Chronic lymphoproliferative disorder of NK-cells (CLPD-NK) predominantly occurs in adults with a median age of diagnosis at 60 years. It is characterized by a persistent increase (≥2 x 109/L, for > 6 months) of mature NK-cells in peripheral blood with an indolent clinical course similar to T-cell large granular lymphocytic leukemia (T-LGL). Somatic gain-of-function (GOF) mutations in STAT3 have been identified in approximately one-third of patients with CLPD-NK. On the other hand, somatic GOF mutations in JAK3 recurrently occur in various types of T-cell neoplasms and exert a GOF effect, unlike biallelic germline loss-of-function mutations found in severe combined immunodeficiency (Figure 1). Here we report on the discovery of a germline GOF JAK3 mutation as a first germline cause of CLPD-NK. Two individuals from one nonconsanguineous family (mother and son) presented at ages 35 and 12 years old with NK cell lymphoproliferation, lymphadenopathy, splenomegaly and autoimmune symptoms. The mother had history of vasculitis while the son was diagnosed with CVID, recurrent multilineage autoimmune cytopenia and subsequently developed psoriasis at 18 years old. The immunological phenotype was assessed in depth in the son and revealed hypogammaglobulinemia with normal vaccine response, expanded NK cells (between 40-60% of total lymphocytes), decreased FOXP3 expression in regulatory T cells and B cell subsets showing decreased total and isotype-switched memory B cells. Flow cytometry revealed expanded population of aberrant NK cells with normal KIR panel. Marrow studies revealed normal karyotype, cellularity and maturation but prominent large granular lymphocytes with benign cytology. Genomic studies identified a novel germline heterozygous JAK3 variant (c.1520A>C/p.Q507P) located at the linker between SH2 and pseudokinase domain (Figure 1). No additional somatic mutations were found. The JAK3 variant was not present in gnomAD database but previously reported as somatic mutation in a patient with T cell prolymphocytic leukemia (Bergmann, Genes Chromosomes Cancer 2014) and predicted to exert a GOF effect. It is well known that JAK3 activation promotes STAT signaling, a known key player in lymphoproliferation. To better understand the biological effect in patient cells, we performed pSTAT5 phosphorylation assay in primary blood lymphocytes after IL2 stimulation, revealing increased pSTAT5 phosphorylation in patient's NK cells. The IL3-dependent BaF3 cell line (containing human wild type JAK3) has been previously used as a robust model to study the effect of JAK3 mutations (Elliott et al. Blood 2011). We therefore introduced the p.Q507P mutation using CRISPR/Cas9 system and used known GOF mutation p.A572V as positive control. While untransduced BaF3 cells died without IL3, p.Q507P-mutant BaF3 cells survived and rapidly expanded without IL3, showing comparable results to positive control. Finally, using western blot we identified constitutive phosphorylation as expected mechanism underlying the lymphoproliferation p.Q507P-mutant cells. In summary, we identify JAK3 as the first germline cause underlying familial CLPD-NK and describe a novel primary immune dysregulatory disorder characterized by non-malignant NK lymphoproliferation with CVID and autoimmune dysregulation. These findings broaden the genetic spectrum of primary immunodeficiency and immune dysregulatory conditions. Disclosures Takemoto: Genentech: Membership on an entity's Board of Directors or advisory committees; Novartis: Other: DSMB Aplastic Anemia Trial. Nichols:Incyte corporation: Research Funding.
- Published
- 2020
24. Prognostic Significance of Major Histocompatibility Complex Class II Expression in Pediatric Adrenocortical Tumors: A St. Jude and Children's Oncology Group Study
- Author
-
John Hicks, David Finkelstein, Gerard P. Zambetti, Carlos Rodriguez-Galindo, Emilia M. Pinto, John K. Choi, Raul C. Ribeiro, Stanley Pounds, Bonald C. Figueiredo, Alberto S. Pappo, Zhifa Liu, and Geoffrey Neale
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Adolescent ,Major histocompatibility complex ,medicine.disease_cause ,Article ,Disease-Free Survival ,MHC Class II Gene ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Adrenocortical Carcinoma ,medicine ,Humans ,Child ,MHC class II ,biology ,Cluster of differentiation ,Histocompatibility Antigens Class II ,Infant ,Prognosis ,Adrenal Cortex Neoplasms ,Haematopoiesis ,030104 developmental biology ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Immunohistochemistry ,Female ,Carcinogenesis ,Biomarkers - Abstract
Purpose: Histologic markers that differentiate benign and malignant pediatric adrenocortical tumors are lacking. Previous studies have implicated an association of MHC class II expression with adrenocortical tumor prognosis. Here, we determined the expression of MHC class II as well as the cell of origin of these immunologic markers in pediatric adrenocortical tumor. The impact of MHC class II gene expression on outcome was determined in a cohort of uniformly treated children with adrenocortical carcinomas. Experimental Design: We analyzed the expression of MHC class II and a selected cluster of differentiation genes in 63 pediatric adrenocortical tumors by Affymetrix Human U133 Plus 2.0 or HT HG-U133+PM gene chip analyses. Cells expressing MHC class II were identified by morphologic and immunohistochemical assays. Results: MHC class II expression was significantly greater in adrenocortical adenomas than in carcinomas (P = 4.8 ×10−6) and was associated with a higher progression-free survival (PFS) estimate (P = 0.003). Specifically, HLA-DPA1 expression was most significantly associated with PFS after adjustment for tumor weight and stage. HLA-DPA1 was predominantly expressed by hematopoietic infiltrating cells and undetectable in tumor cells in 23 of 26 cases (88%). Conclusions: MHC class II expression, which is produced by tumor-infiltrating immune cells, is an indicator of disease aggressiveness in pediatric adrenocortical tumor. Our results suggest that immune responses modulate adrenocortical tumorigenesis and may allow the refinement of risk stratification and treatment for this disease. Clin Cancer Res; 22(24); 6247–55. ©2016 AACR.
- Published
- 2016
25. Improved CNS Control of Childhood Acute Lymphoblastic Leukemia Without Cranial Irradiation: St Jude Total Therapy Study 16
- Author
-
Ching-Hon Pui, Mary V. Relling, Deqing Pei, Raja B. Khan, Tanja A. Gruber, John T. Sandlund, John K. Choi, Cheng Cheng, Dario Campana, William E. Evans, Jeffrey E. Rubnitz, James R. Downing, Sima Jeha, Jun J. Yang, Raul C. Ribeiro, Hiroto Inaba, Susana C. Raimondi, Charles G. Mullighan, and Elaine Coustan-Smith
- Subjects
Male ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,Adolescent ,Lymphoblastic Leukemia ,law.invention ,Polyethylene Glycols ,Central Nervous System Neoplasms ,Remission induction ,Neoplasm Recurrence ,Randomized controlled trial ,Cranial Irradiation ,law ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Asparaginase ,Humans ,Child ,Childhood Acute Lymphoblastic Leukemia ,Survival rate ,Extramural ,business.industry ,Remission Induction ,Cytarabine ,Infant, Newborn ,Infant ,ORIGINAL REPORTS ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Survival Rate ,Methotrexate ,Oncology ,Child, Preschool ,Female ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
PURPOSE Despite contemporary treatment, up to 10% of children with acute lymphoblastic leukemia still experience relapse. We evaluated whether a higher dosage of PEG-asparaginase and early intensification of triple intrathecal therapy would improve systemic and CNS control. PATIENTS AND METHODS Between 2007 and 2017, 598 consecutive patients age 0 to 18 years received risk-directed chemotherapy without prophylactic cranial irradiation in the St Jude Total Therapy Study 16. Patients were randomly assigned to receive PEG-asparaginase 3,500 U/m2 versus the conventional 2,500 U/m2. Patients presenting features that were associated with increased risk of CNS relapse received two extra doses of intrathecal therapy during the first 2 weeks of remission induction. RESULTS The 5-year event-free survival and overall survival rates for the 598 patients were 88.2% (95% CI, 84.9% to 91.5%) and 94.1% (95% CI, 91.7% to 96.5%), respectively. Cumulative risk of any—isolated or combined—CNS relapse was 1.5% (95% CI, 0.5% to 2.5%). Higher doses of PEG-asparaginase did not affect treatment outcome. T-cell phenotype was the only independent risk factor for any CNS relapse (hazard ratio, 5.15; 95% CI, 1.3 to 20.6; P = . 021). Among 359 patients with features that were associated with increased risk for CNS relapse, the 5-year rate of any CNS relapse was significantly lower than that among 248 patients with the same features treated in the previous Total Therapy Study 15 (1.8% [95% CI, 0.4% to 3.3%] v 5.7% [95% CI, 2.8% to 8.6%]; P = .008). There were no significant differences in the cumulative risk of seizure or infection during induction between patients who did or did not receive the two extra doses of intrathecal treatment. CONCLUSION Higher doses of PEG-asparaginase failed to improve outcome, but additional intrathecal therapy during early induction seemed to contribute to improved CNS control without excessive toxicity for high-risk patients.
- Published
- 2019
26. Genomic subtyping and therapeutic targeting of acute erythroleukemia
- Author
-
Marcus B. Valentine, L. Bik To, Ian D. Lewis, Stephen P. Hunger, Guangchun Song, Eric J. Enemark, Elliot Stieglitz, Laura J. Janke, Edgar Sioson, Andrew H. Wei, Yongjin Li, Ji Wen, Lei Shi, Catherine Carmichael, Hamish S. Scott, Katherine Masih, Richard J D'Andrea, Rhonda E. Ries, Shirley Kow Yin Kham, Virginia Valentine, Anna L. Brown, R. Coleman Lindsley, Sarah M. Morris, Benjamin L. Ebert, Chunxu Qu, Manja Meggendorfer, Franco Locatelli, Giuseppe Basso, Ilaria Iacobucci, Daisuke Tomizawa, Benjamin T. Kile, John K. Choi, Michael Rusch, Paula Marlton, Thomas B. Alexander, Stanley Pounds, Torsten Haferlach, Allen Eng Juh Yeoh, Nobutaka Kiyokawa, Deqing Pei, Xiaotu Ma, Debbie Payne-Turner, Mignon L. Loh, Charles G. Mullighan, Soheil Meshinchi, Christopher N. Hahn, Cheng Cheng, Xin Zhou, Iacobucci, Ilaria, Wen, Ji, Meggendorfer, Manja, Choi, John K, Lewis, Ian D, D'Andrea, Richard J, Brown, Anna L, Scott, Hamish S, Hahn, Christopher N, and Mullighan, Charles G
- Subjects
Male ,Myeloid ,Erythroblastic ,Medical and Health Sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,2.1 Biological and endogenous factors ,Aetiology ,Child ,Cancer ,Pediatric ,0303 health sciences ,Leukemia ,biology ,Acute erythroid leukemia ,Myeloid leukemia ,Nuclear Proteins ,Hematology ,Genomics ,Biological Sciences ,Prognosis ,KMT2A ,medicine.anatomical_structure ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,Child, Preschool ,Myeloid-Lymphoid Leukemia Protein ,Female ,acute myeloid-leukemia ,Nucleophosmin ,Biotechnology ,Adult ,Pediatric Research Initiative ,NPM1 ,Adolescent ,Pediatric Cancer ,Childhood Leukemia ,erythroleukemia ,Acute ,Article ,03 medical and health sciences ,Young Adult ,Rare Diseases ,acute erythroid leukemia ,acute lymphoblastic-leukemia ,world-health-organization ,myelodysplastic syndrome ,clonal hematopoiesis ,crystal-structures ,cell-line ,gene ,mutations ,Genetics ,medicine ,Humans ,Preschool ,030304 developmental biology ,Homeodomain Proteins ,Infant, Newborn ,Infant ,Newborn ,medicine.disease ,fms-Like Tyrosine Kinase 3 ,Fms-Like Tyrosine Kinase 3 ,Mutation ,Cancer research ,biology.protein ,Leukemia, Erythroblastic, Acute ,Tumor Suppressor Protein p53 ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Acute erythroid leukemia (AEL) is a high-risk leukemia of poorly understood genetic basis, with controversy regarding diagnosisin the spectrum of myelodysplasia and myeloid leukemia. We compared genomic features of 159 childhood and adult AEL cases with non-AEL myeloid disorders and defined five age-related subgroups with distinct transcriptional profiles: adult, TP53mutated; NPM1 mutated; KMT2A mutated/rearranged; adult, DDX41 mutated; and pediatric, NUP98 rearranged. Genomic features influenced outcome, with NPM1 mutations and HOXB9 overexpression being associated with a favorable prognosis andTP53, FLT3 or RB1 alterations associated with poor survival. Targetable signaling mutations were present in 45% of cases and included recurrent mutations of ALK and NTRK1, the latter of which drives erythroid leukemogenesis sensitive to TRK inhibition.This genomic landscape of AEL provides the framework for accurate diagnosis and risk stratification of this disease, and the rationale for testing targeted therapies in this high-risk leukemia. Refereed/Peer-reviewed
- Published
- 2019
27. Treatment response and outcome of children with T-cell acute lymphoblastic leukemia expressing the gamma-delta T-cell receptor
- Author
-
Paul G. Thomas, Cheng Cheng, Scarlett Evans, Suzanne L. Tomchuck, John K. Choi, Hiroto Inaba, Susana C. Raimondi, Deqing Pei, Sima Jeha, Ching-Hon Pui, and Mari Hashitate Dallas
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,medicine.medical_specialty ,γδ t-all ,T cell ,Lymphoblastic Leukemia ,medicine.medical_treatment ,Immunology ,Gamma-delta T-Cell Receptor ,tcr repertoire ,risk stratification ,Gastroenterology ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,hct ,medicine ,Immunology and Allergy ,Receptor ,Original Research ,Chemotherapy ,business.industry ,pediatric t-all ,γδ t cells ,T-cell receptor ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Minimal residual disease ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,business ,lcsh:RC581-607 - Abstract
T-cell malignancies expressing the γδ T-cell receptor (TCR) are often associated with poor prognosis. Here, we determined the clinical outcome of pediatric patients with T-cell acute lymphoblastic leukemia (T-ALL) expressing the γδ TCR. Of 100 newly diagnosed T-ALL patients, 93 had γδ TCR analysis performed at diagnosis. Repertoire was evaluated by paired sequencing of the rearranged TCR. All patients received intensified chemotherapy and those with minimal residual disease (MRD) ≥ 1% on day 42-46 became candidates for hematopoietic cell transplantation. Of the 93 T-ALL patients, 12 (13%) had γδ T-ALL and 11 (12%) had early T-cell precursor (ETP) ALL. Compared to the remaining 70 T-ALL patients, the γδ T-ALL patients were more likely to have MRD ≥ 1% on day 15-19 (67% vs. 33%, P = 0.03) and day 42-49 (33% vs. 7%; P = 0.007) of remission induction. The 10-year overall survival for γδ T-ALL patients (66.7% ± 22.2%) were lower than that of T-ALL patients (93.3% ± 7.3%, P = 0.001). TCR analysis demonstrated a conserved clonotype. In conclusion, the data suggest that children with γδ T-ALL may have a poor response to remission induction, based on MRD levels and decreased survival than the other T-ALL patients, despite receiving risk-directed therapy.
- Published
- 2018
28. No evidence that G6PD deficiency affects the efficacy or safety of daunorubicin in acute lymphoblastic leukemia induction therapy
- Author
-
Dario Campana, Robert S. Fulton, Mignon L. Loh, Cheng Cheng, Naomi J. Winick, Michael J. Borowitz, William L. Carroll, Dennis W. Jay, Seth E. Karol, Nancy Kornegay, Brent L. Wood, Julie M. Gastier-Foster, Sima Jeha, Ching-Hon Pui, Katherine M. Robinson, John K. Choi, Eric C. Larsen, Meenakshi Devidas, Elaine R. Mardis, Stephen P. Hunger, Wenjian Yang, Elizabeth A. Raetz, and Mary V. Relling
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Neoplasm, Residual ,Daunorubicin ,medicine.medical_treatment ,Glucosephosphate Dehydrogenase ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,hemic and lymphatic diseases ,Medicine ,Humans ,Doxorubicin ,Child ,Neoadjuvant therapy ,Antibiotics, Antineoplastic ,business.industry ,Induction chemotherapy ,Hematology ,Induction Chemotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Prognosis ,Minimal residual disease ,Neoadjuvant Therapy ,Leukemia ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Female ,Safety ,business ,Pharmacogenetics ,030215 immunology ,Cohort study ,medicine.drug ,Follow-Up Studies - Abstract
Background/objectives Anthracyclines are used in induction therapy of pediatric acute lymphoblastic leukemia (ALL) and are known to generate oxidative stress; whether this translates into enhanced antileukemic activity or hemolytic effects in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency is unknown. Design/methods Among 726 pediatric patients with newly diagnosed ALL treated at St. Jude Children's Research Hospital, 22 had deficient G6PD activity. We compared the prevalence of positive minimal residual disease (MRD) ≥1% at Day 15/Day 19 of induction or ≥0.01% at Day 42/Day 46 (end of induction) and the number of red blood cell (RBC) transfusions after daunorubicin in induction between patients with or without G6PD deficiency, adjusting for ALL risk group, treatment protocol, age, and gender. Results There was no difference in Day 15/19 (P = 1) or end of induction MRD (P = 0.76) nor in the number of RBC transfusions (P = 0.73); the lack of association with MRD was confirmed in a dataset of 1192 newly diagnosed male patients enrolled in a Children's Oncology Group trial (P = 0.78). Conclusion We found no evidence that G6PD deficiency affects daunorubicin activity during induction treatment for ALL.
- Published
- 2018
29. Mixed-phenotype acute leukemia, T/megakaryoblastic
- Author
-
John K. Choi and Matthew M. Klairmont
- Subjects
Male ,Anemia ,T-Lymphocytes ,Immunology ,CD33 ,CD34 ,Biochemistry ,Antigen ,Antigens, CD ,Bone Marrow ,Leukemia, Megakaryoblastic, Acute ,hemic and lymphatic diseases ,medicine ,Humans ,Leukocytosis ,Lymphocytes ,Megakaryocyte Progenitor Cells ,biology ,business.industry ,CD117 ,Infant, Newborn ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Leukemia ,Phenotype ,biology.protein ,Female ,medicine.symptom ,business ,Cytometry ,Stem Cell Transplantation - Abstract
[Figure][1] A newborn boy presented with leukocytosis (45 × 109/L), anemia (10 g/dL), and thrombocytopenia (12 × 109/L). Peripheral blood flow cytometry showed CD45 dim-to-negative blasts (panel A) positive for CD13, CD33 (subset), CD117, CD34, CD42b (panel B; >50% CD34-positive blasts
- Published
- 2018
30. Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma
- Author
-
Jeremy S. Abramson, Constance Namirembe, Tara M. Lichtenberg, Patrick Kerchan, Steven J. Reynolds, Julie M. Gastier-Foster, Christopher Rushton, George E. Wright, Cynthia Taylor, Thomas G. Gross, Charles G. Mullighan, Marie Reine Martin, Benjamin Hanf, Steven J.M. Jones, Jackson Orem, Louis M. Staudt, Roland Schmitz, Elaine S. Jaffe, Timothy C. Greiner, Aixiang Jiang, Martin D. Ogwang, Thomas B. Alexander, Bruno M. Grande, Leona W. Ayers, Fabio E. Leal, Tanja Davidsen, Nicholas B. Griner, John T. Sandlund, Ariela Noy, Patee Gesuwan, Andrew J. Mungall, Jay Bowen, Daniela S. Gerhard, Sam M. Mbulaiteye, Hilary Allen, Luka Culibrk, Yussanne Ma, J Martín, Karen Novik, Nancy L. Harris, Yiwen He, Jeffrey M. Bethony, Ryan D. Morin, Eric Y. Zhao, Marco A. Marra, Abraham Omoding, John K. Choi, Maureen A. Dyer, Kishor Bhatia, Wyndham H. Wilson, John D. Irvin, Nicole Knoetze, and Corey Casper
- Subjects
0301 basic medicine ,Male ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,medicine.disease_cause ,Biochemistry ,Cohort Studies ,0302 clinical medicine ,hemic and lymphatic diseases ,Activation-induced (cytidine) deaminase ,Child ,Antigens, Viral ,Mutation ,Lymphoid Neoplasia ,Genes, Immunoglobulin ,Hematology ,Cytidine deaminase ,Prognosis ,Phenotype ,Burkitt Lymphoma ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,Adult ,Adolescent ,Immunology ,Somatic hypermutation ,Biology ,03 medical and health sciences ,Young Adult ,Cytidine Deaminase ,medicine ,Biomarkers, Tumor ,Humans ,Gene ,Genome, Human ,Infant, Newborn ,Infant ,Cell Biology ,medicine.disease ,Diploidy ,Lymphoma ,030104 developmental biology ,Cancer research ,biology.protein ,Transcriptome ,Burkitt's lymphoma ,Follow-Up Studies - Abstract
Although generally curable with intensive chemotherapy in resource-rich settings, Burkitt lymphoma (BL) remains a deadly disease in older patients and in sub-Saharan Africa. Epstein-Barr virus (EBV) positivity is a feature in more than 90% of cases in malaria-endemic regions, and up to 30% elsewhere. However, the molecular features of BL have not been comprehensively evaluated when taking into account tumor EBV status or geographic origin. Through an integrative analysis of whole-genome and transcriptome data, we show a striking genome-wide increase in aberrant somatic hypermutation in EBV-positive tumors, supporting a link between EBV and activation-induced cytidine deaminase (AICDA) activity. In addition to identifying novel candidate BL genes such as SIN3A, USP7, and CHD8, we demonstrate that EBV-positive tumors had significantly fewer driver mutations, especially among genes with roles in apoptosis. We also found immunoglobulin variable region genes that were disproportionally used to encode clonal B-cell receptors (BCRs) in the tumors. These include IGHV4-34, known to produce autoreactive antibodies, and IGKV3-20, a feature described in other B-cell malignancies but not yet in BL. Our results suggest that tumor EBV status defines a specific BL phenotype irrespective of geographic origin, with particular molecular properties and distinct pathogenic mechanisms. The novel mutation patterns identified here imply rational use of DNA-damaging chemotherapy in some patients with BL and targeted agents such as the CDK4/6 inhibitor palbociclib in others, whereas the importance of BCR signaling in BL strengthens the potential benefit of inhibitors for PI3K, Syk, and Src family kinases among these patients.
- Published
- 2018
31. Significance of CNS 2 cerebrospinal fluid status post-induction in pediatric and adolescent patients with acute lymphoblastic leukemia
- Author
-
Ami V. Desai, Susan R. Rheingold, Yimei Li, John K. Choi, Arun Gurunathan, and L. Charles Bailey
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Adolescent ,Lymphoblastic Leukemia ,Status post ,03 medical and health sciences ,Leukocyte Count ,0302 clinical medicine ,Cerebrospinal fluid ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Relapse risk ,Child ,Cerebrospinal Fluid ,Retrospective Studies ,business.industry ,CYTOCENTRIFUGE ,Infant ,Retrospective cohort study ,Hematology ,Induction Chemotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Survival Rate ,Current practice ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Female ,Neoplasm Recurrence, Local ,business ,Leukemic Blasts ,030215 immunology ,Follow-Up Studies - Abstract
Background At diagnosis, there are prognostic implications of low-level leukemic blasts (CNS 2) in the cerebrospinal fluid (CSF) of patients with acute lymphoblastic leukemia (ALL). However, the significance of post-induction CNS 2 results and the impact of equipment on CNS 2 prevalence have not been well studied. Procedure A single-institution retrospective cohort study was conducted to analyze the outcome of patients with ≥1 post-induction CNS 2. A subanalysis compared the proportion of CNS 2 CSF results using 2 different cytocentrifuges; the Shandon Cytospin used from 2005 to 2008 and the Wescor Cytopro used from 2010 to 2014. Results Over 4500 post-induction CSF samples were analyzed, of which 59 were CNS 2. In covariate-adjusted analyses, post-induction CNS 2 did not significantly increase relapse risk. The proportion of CNS 2 results increased 4.3-fold in noninfants and 6.3-fold in infants using the Wescor Cytopro. Cytocentrifuge machine did not affect CNS 3 prevalence. Conclusions These findings support our current practice of not changing management based on a post-induction CNS 2 CSF and highlight how equipment changes can significantly influence testing results. More data are needed to analyze relapse by subpopulations, such as those with repeated CNS 2 findings.
- Published
- 2018
32. Precursor Lymphoid Neoplasms
- Author
-
John K. Choi
- Subjects
Chemotherapy ,medicine.medical_specialty ,Response to therapy ,business.industry ,medicine.medical_treatment ,Lymphoblastic Leukemia ,Lymphoblastic lymphoma ,Cytogenetics ,hemic and immune systems ,medicine.disease ,Risk groups ,hemic and lymphatic diseases ,Cancer research ,Medicine ,Acute lymphoblastic leukemias ,Lymphoid neoplasms ,business - Abstract
Acute lymphoblastic leukemia (ALL) and lymphoblastic lymphoma (LBL) constitute a biologic continuum of neoplastic lymphoid disorders characterized by the proliferation of immature (blast) cells of B-cell or T-cell lineage. Cases with tissue involvement and less than 25% replacement of the marrow cellularity by lymphoid blasts have been designated as lymphoblastic lymphomas. Cases with 25% or greater marrow involvement have been designated acute lymphoblastic leukemias. The modern approach to the treatment of ALL involves tailoring the intensity of the chemotherapy to risk groups defined by the presenting clinical features, lineage, cytogenetics, and molecular findings, as well as by the early response to therapy. This chapter reviews the clinical and pathologic features of lymphoblastic neoplasms.
- Published
- 2018
33. Acute Undifferentiated Leukemia and Mixed-Phenotype Acute Leukemias
- Author
-
John K. Choi
- Subjects
Lineage (genetic) ,Myeloid ,medicine.anatomical_structure ,Hematopoietic cell ,Antigen ,hemic and lymphatic diseases ,medicine ,Cancer research ,Acute Undifferentiated Leukemia ,Biology ,Phenotype ,Leukemic Blasts ,World health - Abstract
Acute leukemias of ambiguous lineage may be divided into acute undifferentiated leukemia (AUL) and mixed-phenotype acute leukemias (MPALs). AULs are leukemias with very primitive phenotypes that lack definitive lineage-specific antigens. MPAL encompasses rare blastic hematopoietic cell neoplasms that express a mixture of myeloid and lymphoid (B- or T-lineage) antigens. The 2008 World Health Organization (WHO) classification defines several categories of MPALs based on the presence of recurrent cytogenetic lesions or in their absence, on the lineage of the leukemic blasts, defined as B-myeloid, T-myeloid, B-T, or B-T-myeloid. Any of these leukemias can develop as biphenotypic or bilineal (bilineage) acute leukemias. The WHO 2016 update will maintain WHO 2008 criteria for MPAL with additional clarifications. This chapter summarizes the clinical and pathologic features of this rare leukemic subentity.
- Published
- 2018
34. International cooperative study identifies treatment strategy in childhood ambiguous lineage leukemia
- Author
-
Simone Stokley, Alexandra Kolenova, Sarah Elitzur, Jan Trka, Barbora Vakrmanova, Neda Marinov, Kirsten Bleckmann, Ales Luks, Karen R. Rabin, Benigna Konatkowska, Hiroto Inaba, Julie Irving, Elaine da Costa, Tamar Feuerstein, Shai Izraeli, Dirk Reinhardt, Ondrej Hrusak, Ester Mejstrikova, Valerie de Haas, Jan Stary, Barbara Buldini, Myriam Campbell, Luciano Dalla-Pozza, Jessa Morales, Olena Kreminska, Marketa Zaliova, Vaclav Capek, John K. Choi, Zuzana Zemanova, Sophia Polychronopoulou, Richard Ratei, Anthony V. Moorman, Kjeld Schmiegelow, Antonis Kattamis, Jorge Rossi, Martin Schrappe, Iveta Janotova, Maria Elena Cabrera, Hanne Vibeke Marquart, Maria S. Felice, Giuseppe Basso, Jitka Stancikova, Peter Svec, Thomas B. Alexander, Anja Möricke, Michael Dworzak, and Drorit Luria
- Subjects
Male ,medicine.medical_specialty ,Lineage (genetic) ,Adolescent ,Biochemistry ,Immunology ,Hematology ,Cell Biology ,Medizin ,World health ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,hemic and lymphatic diseases ,Internal medicine ,Biomarkers, Tumor ,Medicine ,Humans ,Child ,Proportional Hazards Models ,business.industry ,Proportional hazards model ,Infant, Newborn ,Myeloid leukemia ,Disease Management ,Infant ,medicine.disease ,Prognosis ,Combined Modality Therapy ,Leukemia, Biphenotypic, Acute ,Transplantation ,Leukemia ,Treatment Outcome ,030220 oncology & carcinogenesis ,Child, Preschool ,Treatment strategy ,Female ,Disease Susceptibility ,business ,Biomarkers ,030215 immunology - Abstract
Despite attempts to improve the definitions of ambiguous lineage leukemia (ALAL) during the last 2 decades, general therapy recommendations are missing. Herein, we report a large cohort of children with ALAL and propose a treatment strategy. A retrospective multinational study (International Berlin-Frankfurt-Munster Study of Leukemias of Ambiguous Lineage [iBFM-AMBI2012]) of 233 cases of pediatric ALAL patients is presented. Survival statistics were used to compare the prognosis of subsets and types of treatment. Five-year event-free survival (EFS) of patients with acute lymphoblastic leukemia (ALL)-type primary therapy (80% ± 4%) was superior to that of children who received acute myeloid leukemia (AML)-type or combined-type treatment (36% ± 7.2% and 50% ± 12%, respectively). When ALL- or AML-specific gene fusions were excluded, 5-year EFS of CD19+ leukemia was 83% ± 5.3% on ALL-type primary treatment compared with 0% ± 0% and 28% ± 14% on AML-type and combined-type primary treatment, respectively. Superiority of ALL-type treatment was documented in single-population mixed phenotype ALAL (using World Health Organization and/or European Group for Immunophenotyping of Leukemia definitions) and bilineal ALAL. Treatment with ALL-type protocols is recommended for the majority of pediatric patients with ALAL, including cases with CD19+ ALAL. AML-type treatment is preferred in a minority of ALAL cases with CD19- and no other lymphoid features. No overall benefit of transplantation was documented, and it could be introduced in some patients with a poor response to treatment. As no clear indicator was found for a change in treatment type, this is to be considered only in cases with ≥5% blasts after remission induction. The results provide a basis for a prospective trial.
- Published
- 2017
35. Atypical cutaneous γδ T cell proliferation with morphologic features of lymphoma but with clinical features and course of PLEVA or lymphomatoid papulosis
- Author
-
John K. Choi, Rebecca L. King, Albert C. Yan, and Debora R. Sekiguchi
- Subjects
Pathology ,medicine.medical_specialty ,Systemic disease ,Histology ,Atypical Lymphocyte ,business.industry ,T cell ,Dermatology ,Pityriasis lichenoides et varioliformis acuta ,medicine.disease ,Pathology and Forensic Medicine ,Lymphoma ,medicine.anatomical_structure ,Medicine ,T-cell lymphoma ,Lymphomatoid papulosis ,business ,Hematopathology - Abstract
Reactive lymphoid infiltrates of the skin composed predominantly of gamma-delta (γδ) T cells are not well described in the literature. Herein we report a case of an otherwise healthy 4-year-old male who presented with a waxing and waning papular rash characterized by small, discrete crusted papules spread across his trunk, face and extremities. Clinical evaluation revealed no evidence of systemic disease. Microscopic examination revealed a dermal, perivascular infiltrate of highly atypical lymphocytes with a γδ T cell phenotype, worrisome for primary cutaneous γδ T cell lymphoma. The clinical course, however, was that of a reactive condition and prompted consideration of a diagnosis of pityriasis lichenoides et varioliformis acuta (PLEVA) and lymphomatoid papulosis (LyP). In many ways, this case defies current classification schemes and seems to expand the spectrum of reactive γδ T cell infiltrates of the skin.
- Published
- 2015
36. Metachronous T-Lymphoblastic Lymphoma and Burkitt Lymphoma in a Child With Constitutional Mismatch Repair Deficiency Syndrome
- Author
-
Mary Beth McCarville, John T. Sandlund, Cary P. Cavender, Rose B. McGee, Erica C. Kaye, John K. Choi, Thomas B. Alexander, and Kim E. Nichols
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,business.industry ,Cancer predisposition ,Hematology ,medicine.disease ,Lymphoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Nonossifying fibroma ,Oncology ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Café au lait spot ,Pediatrics, Perinatology and Child Health ,MISMATCH REPAIR DEFICIENCY ,Medicine ,Pilomatrixoma ,medicine.symptom ,business ,T-Lymphoblastic Lymphoma ,Early onset - Abstract
Constitutional mismatch repair deficiency (CMMRD) is a cancer predisposition syndrome associated with a high risk of developing early-onset malignancies of the blood, brain, and intestinal tract. We present the case of a patient with T-lymphoblastic lymphoma at the age of 3 years, followed by Burkitt lymphoma 10 years later. This patient also exhibited numerous nonmalignant findings including cafe au lait spots, lipomas, bilateral renal nodules, a nonossifying fibroma, multiple colonic adenomas, and a rapidly enlarging pilomatrixoma. The spectrum of malignant and nonmalignant neoplasms in this patient highlights the remarkable diversity, and early onset, of lesions seen in children with CMMRD.
- Published
- 2016
37. The genetic basis and cell of origin of mixed phenotype acute leukaemia
- Author
-
Richard A. Moore, Deqing Pei, Daniela S. Gerhard, Beisi Xu, Hiroto Inaba, Tanja M. Davidsen, Andrew J. Mungall, Ching-Hon Pui, Jeffrey E. Rubnitz, Marco A. Marra, C. Michel Zwaan, Hiroki Yoshihara, Steven J.M. Jones, Ilaria Iacobucci, Liang Ding, Brent L. Wood, Laura J. Janke, Patee Gesuwan, Mignon L. Loh, Sarah Elitzur, Jaime M. Guidry Auvil, Yung-Li Yang, Xueyuan Cao, Meenakshi Devidas, James R. Downing, Yussanne Ma, Kirsten Dickerson, Tim Lammens, Stephen P. Hunger, John T. Horan, Marcus B. Valentine, Stanley Pounds, Etan Orgel, Ondrej Hrusak, Thomas B. Alexander, John K. Choi, Yu Liu, Debbie Payne-Turner, Soheil Meshinchi, Andrew S. Moore, Zhaohui Gu, Anthony V. Moorman, Leandro C. Hermida, Daniel Catchpoole, Lei Shi, Scott Newman, Valerie de Haas, Barbara Buldini, Allen Eng Juh Yeoh, Michael J. Borowitz, Barbara De Moerloose, Malcolm A. Smith, Nobutaka Kiyokawa, Dirk Reinhardt, Hiroki Hori, Andrew Carrol, Jinghui Zhang, Charles G. Mullighan, Kim E. Nichols, Daisuke Tomizawa, Giuseppe Basso, Nyla A. Heerema, and Pediatrics
- Subjects
0301 basic medicine ,Male ,Myeloid ,Lineage (genetic) ,General Science & Technology ,DNA Mutational Analysis ,Medizin ,Biology ,ZNF384 ,Article ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Genetic variation ,medicine ,Humans ,Cell Lineage ,Multidisciplinary ,Models, Genetic ,Genome, Human ,Genetic Variation ,Genomics ,medicine.disease ,Phenotype ,Leukemia, Biphenotypic, Acute ,Haematopoiesis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Neoplastic Stem Cells ,Trans-Activators ,Female - Abstract
Mixed phenotype acute leukaemia (MPAL) is a high-risk subtype of leukaemia with myeloid and lymphoid features, limited genetic characterization, and a lack of consensus regarding appropriate therapy. Here we show that the two principal subtypes of MPAL, T/myeloid (T/M) and B/myeloid (B/M), are genetically distinct. Rearrangement of ZNF384 is common in B/M MPAL, and biallelic WT1 alterations are common in T/M MPAL, which shares genomic features with early T-cell precursor acute lymphoblastic leukaemia. We show that the intratumoral immunophenotypic heterogeneity characteristic of MPAL is independent of somatic genetic variation, that founding lesions arise in primitive haematopoietic progenitors, and that individual phenotypic subpopulations can reconstitute the immunophenotypic diversity in vivo. These findings indicate that the cell of origin and founding lesions, rather than an accumulation of distinct genomic alterations, prime tumour cells for lineage promiscuity. Moreover, these findings position MPAL in the spectrum of immature leukaemias and provide a genetically informed framework for future clinical trials of potential treatments for MPAL.
- Published
- 2017
38. Trisomy 21–Associated Transient Abnormal Myelopoiesis Involving the Maternal Space of the Placenta: A Case Report and Literature Review
- Author
-
Ona Faye-Petersen, John K. Choi, Vishnu Reddy, and Qian Dai
- Subjects
Adult ,Male ,medicine.medical_specialty ,Placenta Diseases ,Chromosomes, Human, Pair 21 ,Placenta ,Trisomy ,Context (language use) ,Malignancy ,Leukemoid Reaction ,Pathology and Forensic Medicine ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Prenatal Diagnosis ,medicine ,Humans ,Fetus ,030219 obstetrics & reproductive medicine ,Obstetrics ,business.industry ,Transient abnormal myelopoiesis ,Infant, Newborn ,General Medicine ,medicine.disease ,Peripheral blood ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Female ,Down Syndrome ,business - Abstract
We report a case of a male newborn with trisomy 21 and transient abnormal myelopoiesis at birth whose placenta showed extravasated fetal blasts in the perivillous (maternal) space. Concern for possible maternal spread of fetal malignancy prompted a Kleihauer-Betke test and flow cytometric analysis of the maternal peripheral blood on postpartum day 2. Notably, no evidence of the persistence of fetal cells in the maternal blood was identified, a finding that likely reflected successful maternal immunologic clearance of the fetal blasts and erythrocytes, and/or blast cellular fragility and limited viability. Ours is the first report, to our knowledge, documenting maternal peripheral-blood follow-up evaluation of this disorder in the English literature. We discuss our case in the context of a comprehensive review of fetoneonatal solid tumor and leukemic proliferative disorders with placental involvement and evidence of maternal metastasis.
- Published
- 2014
39. Prognostic factors in children with acute myeloid leukaemia and excellent response to remission induction therapy
- Author
-
Dario Campana, Wing Leung, Jeffrey E. Rubnitz, Jeffrey W. Taub, James R. Downing, Raul C. Ribeiro, W P Bowman, Seth E. Karol, John K. Choi, Hiroto Inaba, Susana C. Raimondi, Elaine Coustan-Smith, Xueyuan Cao, Sheila A. Shurtleff, Ching-Hon Pui, Gary V. Dahl, and Barbara A. Degar
- Subjects
Oncology ,medicine.medical_specialty ,Prognostic factor ,Neoplasm, Residual ,Adolescent ,Karyotype ,Article ,Remission induction ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Remission Induction Therapy ,medicine ,Humans ,In patient ,Child ,Chromosome Aberrations ,business.industry ,Remission Induction ,Hematology ,Prognosis ,Minimal residual disease ,body regions ,Leukemia, Myeloid, Acute ,Treatment Outcome ,fms-Like Tyrosine Kinase 3 ,Child, Preschool ,Immunology ,Fms-Like Tyrosine Kinase 3 ,Cohort ,Myeloid leukaemia ,business - Abstract
Minimal residual disease (MRD) is a strong prognostic factor in children and adolescents with acute myeloid leukaemia (AML) but nearly one-quarter of patients who achieve MRD-negative status still relapse. The adverse prognostic factors among MRD-negative patients remain unknown. We analysed the AML02 study cohort to identify demographic and genetic prognostic factors. Among the presenting features, certain 11q23 abnormalities, such as t(6;11) and t(10;11), acute megakaryoblastic leukaemia without the t(1;22), and age ≥10 years were associated with inferior outcome in patients who had MRD-negative status after either remission induction I or II. By contrast, those with rearrangement of CBF genes had superior outcome. Our study identifies patient populations for whom close post-remission MRD monitoring to detect and treat emerging relapse and adjustment in treatment intensity might be indicated.
- Published
- 2014
40. Phase I Study of the Safety and Pharmacokinetics of Plerixafor in Children Undergoing a Second Allogeneic Hematopoietic Stem Cell Transplantation for Relapsed or Refractory Leukemia
- Author
-
Mari Hashitate Dallas, Guolian Kang, Shane J Cross, Jeffrey Jacobsen, Anusha Sunkara, Asha Pillai, David Shook, John C. Panetta, Ashok Srinivasan, Wing Leung, Brandon M. Triplett, John K. Choi, and Christine Hartford
- Subjects
Male ,Melphalan ,Oncology ,Benzylamines ,medicine.medical_specialty ,Transplantation Conditioning ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,ThioTEPA ,Cyclams ,Biomarkers, Pharmacological ,Article ,Immunophenotyping ,Heterocyclic Compounds ,Recurrence ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Children ,Allogeneic ,Transplantation ,Leukemia ,business.industry ,Plerixafor ,Hematopoietic Stem Cell Transplantation ,Stem cell transplantation ,Hematology ,medicine.disease ,3. Good health ,Surgery ,Fludarabine ,Treatment Outcome ,Tolerability ,Female ,business ,medicine.drug - Abstract
The safety, pharmacokinetics and biological effect of plerixafor in children as part of a conditioning regimen for chemo-sensitization in allogeneic hematopoietic stem cell transplantation (HSCT) have not been studied. This is a phase I study of plerixafor designed to evaluate its tolerability at dose of 0.24 mg/kg given intravenously on day -4 (level 1), day -4, and day -3 (level 2), or day -4, -3, and day -2 (level 3) in combination with fludarabine, thiotepa, melphalan, and rabbit anti-thymocytic globulin for a second allogeneic HSCT in children with refractory or relapsed leukemia. Immunophenotype analysis was performed on blood and bone marrow prior to and after plerixafor administration. Twelve patients were enrolled. Plerixafor at all 3 levels was well tolerated without dose-limiting toxicity. Transient gastrointestinal side effects of National Cancer Institute grade 1 or 2 in severity were the most common adverse events. The area under the concentration-time curve increased proportionally to the dose level. Plerixafor clearance was higher in males, and increased linearly with body weight, and glomerular filtration rate. The clearance decreased and the elimination half-life increased significantly from dose level 1 to 3 (P < 0.001). Biologically, the proportion of CXCR4-positive blasts and lymphocytes both in the bone marrow and peripheral blood, increased after plerixafor administration.
- Published
- 2014
41. Impact of tyrosine kinase inhibitors on minimal residual disease and outcome in childhood Philadelphia chromosome-positive acute lymphoblastic leukemia
- Author
-
Dario Campana, Sheila A. Shurtleff, Jeffrey E. Rubnitz, Raul C. Ribeiro, Deqing Pei, Deepa Bhojwani, John K. Choi, Elaine Coustan-Smith, Ching-Hon Pui, Hiroto Inaba, Susana C. Raimondi, Cheng Cheng, John T. Sandlund, Scott C. Howard, Sima Jeha, Monika L. Metzger, and Jeffrey Jacobsen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Induction chemotherapy ,Imatinib ,Hematopoietic stem cell transplantation ,Philadelphia chromosome ,medicine.disease ,Minimal residual disease ,respiratory tract diseases ,Dasatinib ,Leukemia ,hemic and lymphatic diseases ,Internal medicine ,Immunology ,medicine ,business ,Survival rate ,medicine.drug - Abstract
BACKGROUND Breakpoint cluster region-Abelson murine leukemia viral oncogene homolog 1 (BCR-ABL1) tyrosine kinase inhibitors (TKIs) improve the outcome of patients with childhood Philadelphia chromosome (Ph)-positive acute lymphoblastic leukemia (ALL) when they are incorporated into postremission induction chemotherapy. To date, no data are available on the impact of TKIs on minimal residual disease (MRD) at the end of induction therapy among patients who have a poor early response to 2 weeks of induction therapy that does not include TKIs. METHODS The authors analyzed the early response to TKIs during remission induction in children with Ph-positive ALL who were treated at St. Jude Children's Research Hospital. MRD was measured on days 15 and 42 of induction. TKIs were incorporated into induction therapy on day 22 in the post-TKI era. RESULTS TKIs produced a marked drop in MRD levels: at the end of remission induction, 9 of 11 patients who received imatinib or dasatinib and conventional induction chemotherapy achieved MRD-negative status compared with only 2 of 16 patients who received chemotherapy alone (P
- Published
- 2014
42. Adult T-Cell Leukemia/Lymphoma
- Author
-
Sohail Qayyum and John K. Choi
- Subjects
Adult ,Endemic Diseases ,Adult population ,Adult T-cell leukemia/lymphoma ,Virus ,Pathology and Forensic Medicine ,Diagnosis, Differential ,Retrovirus ,Japan ,hemic and lymphatic diseases ,medicine ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Neoplasm ,Human T-lymphotropic virus 1 ,biology ,business.industry ,General Medicine ,Prognosis ,medicine.disease ,biology.organism_classification ,HTLV-I Infections ,Virology ,United States ,Lymphoma ,Medical Laboratory Technology ,Leukemia ,Latin America ,Caribbean Region ,Virus type ,Immunology ,business - Abstract
Adult T-cell leukemia/lymphoma is a rare mature CD4+ T-cell neoplasm caused by the retrovirus human T-lymphotrophic virus type 1. At present there are approximately 20 million people infected globally with this virus, and most of these individuals belong to the endemic areas in southern Japan, Africa, the Caribbean basin, and Latin America. In the United States, it is usually seen in immigrants from these endemic regions. Adult T-cell leukemia/lymphoma predominantly affects the adult population and is rare in children. Adult T-cell leukemia/lymphoma has 4 subtypes: acute, lymphomatous, chronic, and smoldering. Clinically, the first 2 variants are classified as aggressive, and the latter two are classified as indolent. Given the rare occurrence and diagnostic challenges associated with adult T-cell leukemia/lymphoma, this review will highlight its salient features to aid in recognition of this entity and perform a comprehensive diagnostic workup.
- Published
- 2014
43. Sorafenib in Combination with Standard Chemotherapy for Children with High Allelic Ratio FLT3/ITD+ AML Improves Event-Free Survival and Reduces Relapse Risk: A Report from the Children's Oncology Group Protocol AAML1031
- Author
-
Jessica A. Pollard, Anders Kolb, Susana C. Raimondi, John K. Choi, Michael R. Loken, John E. Levine, Richard Aplenc, Lillian Sung, Betsy A. Hirsch, Elizabeth Fox, Todd A. Alonzo, Soheil Meshinchi, Andrew C. Wood, Samir B. Kahwash, Robert B. Gerbing, Brian T. Fisher, Patrick A. Brown, Alan S. Gamis, and Katherine Tarlock
- Subjects
Oncology ,Sorafenib ,medicine.medical_specialty ,Chemotherapy ,Randomization ,business.industry ,Proportional hazards model ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Biochemistry ,Chemotherapy regimen ,Maintenance therapy ,Statistical significance ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
INTRODUCTION Sorafenib, a multi-kinase tyrosine kinase inhibitor (TKI) targets FLT3 internal tandem duplication (FLT3/ITD) mutations and has efficacy in adult FLT3/ITD+ AML. High allelic ratio (HAR) FLT3/ITD mutations (allelic ratio of > 0.4, referred to as FLT3/ITD+) confer poor prognosis in de novo pediatric AML. COG AAML1031 Arm C studied the feasibility and efficacy of adding sorafenib to standard chemotherapy and as a single agent maintenance therapy in pediatric HAR FLT3/ITD+ patients. METHODS The AAML1031 primary randomization (Arm A vs. B) has been previously reported (Aplenc et al. Blood 2016;128:899). Patients with FLT3/ITD+ AML were offered secondary enrollment on Arm C and toxicities were monitored on respective treatment arms. An initial safety phase (Cohort 1, C1; N=12) defined the maximum tolerated dose (MTD) of sorafenib when added to induction II chemotherapy and subsequent courses. Following completion, the study was amended (Cohort 2; C2) to start sorafenib on day 11 of induction I and concomitantly with chemotherapy in subsequent cycles. A year of sorafenib maintenance was also added. Due to concerns for cardiac toxicity on interim analyses, the study was amended (Cohort 3; C3) to initiate sorafenib after chemotherapy completion in all courses. Sorafenib pharmacokinetics (PK) and plasma inhibitory activity (PIA) were measured in a subset of patients. Clinical outcome analysis was limited to cohorts 2/3 given lack of induction I sorafenib exposure in C1. Arm C results were compared to N=34 HAR FLT3/ITD+ AML patients on the control arm of COG AAML0531 (Arm A), a group that received comparable chemotherapy without sorafenib. Complete remission (CR), event-free survival (EFS), overall survival (OS) and relapse risk (RR) of patients with HAR FLT3/ITD AML enrolled on the 2 trials were compared to assess the impact of sorafenib use. Cox proportional hazards were used for multivariable analyses. RESULTS In the dose finding phase (C1), 12 patient were enrolled and the MTD of sorafenib defined as 200 mg/m2; dose limiting toxicities included rash (N=2; 1 grade III, 1 grade II), grade II hand foot syndrome and grade III fever. For the evaluation of efficacy, 80 C2/C3 (C2: N=33, C3: N=47) patients were enrolled. 30/80 (37.5%) received at least 1 maintenance cycle; 20/80 (25%) completed all courses. As outcomes were compared to N=34 HAR FLT3/ITD+ AML patients enrolled on the preceding trial AAML0531 Arm A as a historical comparison, clinical characteristics were described for the 2 groups (Table 1). For AAML1031 C2/C3, rates of induction I CR were 73% vs. 56% for AAML0531 Arm A (p=0.078). Following induction II, CR rates were 91% and 70% respectively (p=0.007). 3 year EFS from study entry was 57.5% for Arm C vs. 34.3% for AAML0531 Arm A; (p=0.007) whereas 3 year OS from study entry was not significantly different (63.9% vs. 54.1%, p=0.375, Figure 1A). RR from CR was reduced with sorafenib treatment (Arm C 18.2% vs. historical 52.5%, p=0.006, Figure 1B). The improvement in EFS and RR observed with sorafenib was retained in those with wild-type nucleophosmin (NPM-) FLT3/ITD+ AML but lacked statistical significance compared to historical in NPM+ FLT3/ITD+ disease (Figure 2A/2B). As 21/34 (62%) patients in the historical control did not undergo HSCT there was concern that the improved outcome observed for Arm C reflected increased use of HSCT in AAML1031. Multivariable analysis that adjusted for HSCT resulted in significantly lower EFS and higher RR for FLT3/ITD+ patients on AAML0531, suggesting that increased use of HSCT on Arm C did not explain the improved outcomes observed (EFS: HR 2.04, CI 1.18 - 3.53; p=0.01, RR: HR 3.09, CI 1.28 - 7.49; p=0.012). While dose modifications were more frequent for FLT3/ITD+ patients on Arm C versus arms A/B of AAML1031, targeted toxicity rates were otherwise no higher (Table 2). PIA assays demonstrated median trough FLT3 inhibition of 92% (n=29, 95% CI 79-94%), 91% (n=84, 95% CI 87-94%) and 81% (n=70, 95% CI 76-90%) for induction I, induction II and intensification I, respectively. CONCLUSION Addition of sorafenib to Arm C of AAML1031 was safe and resulted in potent FLT3 inhibition, particularly early in therapy. Sorafenib improved rates of induction II CR as well as 3 year EFS and reduced RR from CR compared to historical controls. These data support use of sorafenib in pediatric patients with HAR FLT3/ITD+ AML. Disclosures Fisher: Pfizer: Research Funding; Astellas: Other: Data Safety Monitoring Board Chair for an antifungal study; Merck: Research Funding. Levine:Viracor: Patents & Royalties: biomarker patent; Biogen: Other: non-financial support; Ironwood: Consultancy; Bluebird Bio: Consultancy; National Cancer Institute: Research Funding; Novartis: Consultancy; Incyte: Consultancy, Research Funding; Kamada: Research Funding. Loken:Hematologics, Inc: Employment, Equity Ownership. OffLabel Disclosure: sorafenib for high allelic ratio FLT3/ITD+ AML
- Published
- 2019
44. Abstract 3652: USP7 heterozygous loss-of-function affects T-cell differentiation in pediatric T-ALL
- Author
-
Li Dong, Junmin Peng, John K. Choi, Bensheng Ju, Kanisha Kavdia, John Easton, Timothy I. Shaw, Janke Janke, Anthony A. High, Yu Liu, Jinghui Zhang, Wei Gu, Vishwajeeth Pagala, Chenxi Qian, Jiyang Yu, James R. Downing, and Bridget Shaner
- Subjects
Cancer Research ,Oncology ,Transgene ,T cell differentiation ,Conditional gene knockout ,Gene expression ,Biology ,Haploinsufficiency ,Molecular biology ,Recombination-activating gene ,CD8 ,TAL1 - Abstract
Ubiquitin-specific-processing protease 7 (USP7), a protein deubiquitinase, is one of the most frequently mutated genes (33%) in the TAL1 subtype of pediatric T-lineage acute lymphoblastic leukemia (T-ALL). However, the functional effect of USP7 haploinsufficiency on T-ALL pathogenesis remains elusive. To understand USP7 haploinsufficiency’s impact on T-ALL, we performed gene expression analysis on 42 non-early T-cell precursor T-ALL RNAseq samples downloaded from phs000218. The gene expression analysis suggested that USP7 haploinsufficiency (USP7-mut N = 12; USP7-wt N = 30) down-regulated the expression of T-cell maturation markers (e.g. RAG1, RAG2, and CD1B), which were negatively regulated by TAL1 [1]. RNAseq on a T-ALL cell line with USP7 knocked down by shRNA also found the same TAL1 negatively regulated gene set, further supporting an increase of TAL1 activity in the USP7 mutated T-ALLs. To examine whether the T-cell maturation was affected by USP7 heterozygous knockout, we generated a conditional knockout (cKO) mouse model by cross-breeding the transgenic vav1-cre mice with the USP7fl/fl mice to obtain heterozygous USP7fl/wt-vav1-cre. Thymocytes isolated from cKO mice were co-cultured with the OP9-Δ1 cells in the medium supplied with cytokines. The cell surface differentiation markers, CD4 and CD8 were stained for flow cytometry detection, and we observed an increase of double-negative cells and a decrease of double-positive cells in USP7-het-KO mice versus control (N = 4 in each group; p-value < 0.05), consolidating the disrupted T-cell development ex vivo. To further understand USP7’s mechanism to regulate TAL1, we analyzed proteins that interacted with USP7 by affinity purification-mass spectrometry (AP-MS) and immune-precipitated followed by western-blotting (IP/WB). AP-MS with the anti-USP7 antibody revealed that USP7 directly interacts with TAL1 in Jurkat cells; AP-MS with the anti-TAL1 antibody also reciprocated the USP7-TAL1 interactions. Whole proteome analysis, through tandem-mass-tag and two-dimensional liquid chromatography-tandem mass spectrometry, revealed that USP7 knockdown down-regulated TRIM27, a deubiquitin target of USP7. IP/WB further confirmed the interaction between USP7, TRIM27, and TAL1, suggesting a possible synergistic relationship between USP7 and TRIM27 to regulate TAL1. In conclusion, our finding demonstrates heterozygous loss of function of USP7 dysregulates T-cell maturation by enhancing TAL1 activity. Future work on TRIM27 could further shed light on the mechanism underlying USP7’s ability to regulate TAL1. Reference: [1] Sanda et al. Cancer Cell 22, 209 (2012). Citation Format: Timothy I. Shaw, Li Dong, Anthony High, Yu Liu, Bensheng Ju, Kanisha Kavdia, Vishwajeeth Pagala, Bridget Shaner, John Easton, Chenxi Qian, Jiyang Yu, Janke Janke, John Kim Choi, Junmin Peng, Wei Gu, James R. Downing, Jinghui Zhang. USP7 heterozygous loss-of-function affects T-cell differentiation in pediatric T-ALL [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3652.
- Published
- 2019
45. Patient-derived induced pluripotent stem cells recapitulate hematopoietic abnormalities of juvenile myelomonocytic leukemia
- Author
-
Deborah L. French, Chiaka Aribeana, Julie Weng, Sarah K. Tasian, Stella T. Chou, Helene Favre, Silvia Bresolin, Mignon L. Loh, Spencer K. Sullivan, Shilpa Gandre-Babbe, Lin Lu, Mitchell J. Weiss, Prasuna Paluru, and John K. Choi
- Subjects
Male ,Heterozygote ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Immunology ,Mutation, Missense ,Biology ,Biochemistry ,Cohort Studies ,STAT5 Transcription Factor ,Tumor Cells, Cultured ,medicine ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Induced pluripotent stem cell ,Myeloproliferative neoplasm ,Interleukin 3 ,Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,Juvenile myelomonocytic leukemia ,Granulocyte-Macrophage Colony-Stimulating Factor ,Cell Differentiation ,Cell Biology ,Hematology ,medicine.disease ,Leukemia ,Haematopoiesis ,Granulocyte macrophage colony-stimulating factor ,Leukemia, Myelomonocytic, Juvenile ,Neoplastic Stem Cells ,Cancer research ,Female ,medicine.drug - Abstract
Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative neoplasm of young children initiated by mutations that deregulate cytokine receptor signaling. Studies of JMML are constrained by limited access to patient tissues. We generated induced pluripotent stem cells (iPSCs) from malignant cells of two JMML patients with somatic heterozygous p.E76K missense mutations in PTPN11, which encodes SHP-2, a nonreceptor tyrosine phosphatase. In vitro differentiation of JMML iPSCs produced myeloid cells with increased proliferative capacity, constitutive activation of granulocyte macrophage colony-stimulating factor (GM-CSF), and enhanced STAT5/ERK phosphorylation, similar to primary JMML cells from patients. Pharmacological inhibition of MEK kinase in iPSC-derived JMML cells reduced their GM-CSF independence, providing rationale for a potential targeted therapy. Our studies offer renewable sources of biologically relevant human cells in which to explore the pathophysiology and treatment of JMML. More generally, we illustrate the utility of iPSCs for in vitro modeling of a human malignancy.
- Published
- 2013
46. Intestinal γδ T-cell lymphomas are most frequently of type II enteropathy-associated T-cell type
- Author
-
John K. Choi, Massimo Trucco, Steven H. Swerdlow, Urvashi Surti, Amanda L. Wilson, S. Branden Van Oss, Elias Campo, Grzegorz K. Przybylski, and Raymond E. Felgar
- Subjects
Adult ,Male ,T cell ,CD3 ,Mucocutaneous zone ,Human leukocyte antigen ,Biology ,Lymphoma, T-Cell ,Article ,Immunophenotyping ,Pathology and Forensic Medicine ,Enteropathy-Associated T-Cell Lymphoma ,HLA Antigens ,hemic and lymphatic diseases ,Intestinal Neoplasms ,Biomarkers, Tumor ,medicine ,Humans ,In Situ Hybridization, Fluorescence ,Aged ,Oligonucleotide Array Sequence Analysis ,Aged, 80 and over ,Receptors, Antigen, T-Cell, gamma-delta ,medicine.disease ,Lymphoma ,medicine.anatomical_structure ,Immunology ,biology.protein ,Enteropathy-associated T-cell lymphoma ,Female ,CD8 - Abstract
Summary Enteropathy-associated T-cell lymphoma includes type I cases and distinctive type II cases that, according to 2008 and 2010 World Health Organization descriptions, are T-cell receptor β +. Although T-cell receptor γδ enteropathy-associated T-cell lymphomas are reported, it is unknown if they have distinctive features and if they should be categorized as enteropathy-associated T-cell lymphoma or as a mucocutaneous γδ T-cell lymphoma. To address these questions, the clinicopathologic, immunophenotypic, molecular, and cytogenetic features of 5 γδ -enteropathy-associated T-cell lymphomas were investigated. Only 1 patient had celiac disease and had type I enteropathy-associated T-cell lymphoma, and the others fulfilled the histopathologic criteria for type II enteropathy-associated T-cell lymphoma. All lacked cutaneous involvement. A celiac disease–associated HLA type was found in the patient with CD and one of four others. All were T-cell receptor γ +, T-cell receptor δ +, β F1−, CD3+, CD7+, CD5−, CD4−, and TIA-1+ with variable staining for CD2 (3/5), CD8 (2/5), Granzyme B (1/5), and CD56 (4/5). Fluorescence in situ hybridization demonstrated 9q34 gains in 4 cases, with 9q33-34 gains by single nucleotide polymorphism in 3 of these. Single nucleotide polymorphism analysis also demonstrated gains in 5q34-q35.1/5q35.1 (4/5), 8q24 (3/5), and in 32 other regions in 3 of 5 cases. V δ 1 rearrangements were identified in 4 of 4 cases with documented clonality showing the same clone in normal-appearing distant mucosa (3/3 tested cases). Thus, γδ -enteropathy-associated T-cell lymphomas share many features with other enteropathy-associated T-cell lymphoma and are mostly of type II. Their usual nonactivated cytotoxic phenotype and V δ 1 usage are features unlike many other mucocutaneous γδ T-cell lymphomas but shared with hepatosplenic T-cell lymphoma. These findings support the conclusion that a γδ T-cell origin at extracutaneous sites does not define a specific entity.
- Published
- 2013
47. CD30 Staining in Pediatric Neoplasms, Study of 586 Cases
- Author
-
John K. Choi and Jinjun Cheng
- Subjects
Pathology ,medicine.medical_specialty ,CD30 ,business.industry ,Medicine ,General Medicine ,business ,Staining - Published
- 2016
48. Phase I Study of Selinexor, a Selective Inhibitor of Nuclear Export, in Combination With Fludarabine and Cytarabine, in Pediatric Relapsed or Refractory Acute Leukemia
- Author
-
Raul C. Ribeiro, John K. Choi, Ching-Hon Pui, Norman J. Lacayo, Thomas B. Alexander, and Jeffrey E. Rubnitz
- Subjects
0301 basic medicine ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Active Transport, Cell Nucleus ,Administration, Oral ,Pharmacology ,Gastroenterology ,Drug Administration Schedule ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Pharmacokinetics ,Recurrence ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Child ,Acute leukemia ,Leukemia ,business.industry ,Remission Induction ,Cytarabine ,Infant ,ORIGINAL REPORTS ,Triazoles ,medicine.disease ,Fludarabine ,030104 developmental biology ,Hydrazines ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Child, Preschool ,Toxicity ,Acute Disease ,Female ,business ,Vidarabine ,medicine.drug - Abstract
Purpose To characterize the toxicity, pharmacokinetics, and pharmacodynamics of selinexor, a selective inhibitor of nuclear export, when combined with fludarabine and cytarabine, in children with relapsed or refractory leukemia. Patients and Methods Eighteen patients with relapsed or refractory acute leukemia were enrolled in the SELHEM (Selinexor With Fludarabine and Cytarabine for Treatment of Refractory or Relapsed Leukemia or Myelodysplastic Syndrome) clinical trial (NCT02212561). Selinexor, initially at 30 mg/m2 per dose, was given orally on days 1, 3, 8, 10, 22, and 24 and was escalated according to a rolling-six design. Fludarabine 30 mg/m2 and cytarabine 2 g/m2 were administered on days 15 to 19. Pharmacokinetic and pharmacodynamic studies were performed on days 1 and 22. Response evaluations were performed on day 15 and at the completion of course 1. Results Among the 17 patients who were evaluable for toxicity, three were treated at 30 mg/m2, three at 40 mg/m2, six at 55 mg/m2, and five at 70 mg/m2. The most common grade 3 nonhematologic toxicity was asymptomatic hyponatremia. Two patients who were treated at 70 mg/m2 experienced reversible cerebellar toxicity, thereby defining the dose-limiting toxicity. Pharmacokinetic parameters demonstrated that plasma exposure was dose proportional. Fifteen of 16 patients demonstrated at least a twofold increase of XPO1 mRNA, indicating inhibition of the XPO1 protein. In this group of heavily pretreated, relapsed, and refractory patients, seven of 15 evaluable patients (47%) achieved complete response or complete response with incomplete count recovery. Conclusion Selinexor, in combination with fludarabine and cytarabine, is tolerable at doses up to 55 mg/m2 in pediatric patients with relapsed or refractory leukemia. All patients who received selinexor at ≥ 40 mg/m2 demonstrated XPO1 target inhibition. Response rates are promising and will be further explored in a phase II trial.
- Published
- 2016
49. Metachronous T-Lymphoblastic Lymphoma and Burkitt Lymphoma in a Child With Constitutional Mismatch Repair Deficiency Syndrome
- Author
-
Thomas B, Alexander, Rose B, McGee, Erica C, Kaye, Mary Beth, McCarville, John K, Choi, Cary P, Cavender, Kim E, Nichols, and John T, Sandlund
- Subjects
Male ,stomatognathic diseases ,Adolescent ,Brain Neoplasms ,Neoplastic Syndromes, Hereditary ,hemic and lymphatic diseases ,Humans ,Genetic Predisposition to Disease ,Neoplasms, Second Primary ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Colorectal Neoplasms ,Burkitt Lymphoma ,Article - Abstract
Constitutional mismatch repair deficiency (CMMRD) is a cancer predisposition syndrome associated with a high risk of developing early-onset malignancies of the blood, brain, and intestinal tract. We present the case of a patient with T-lymphoblastic lymphoma at the age of 3 years, followed by Burkitt lymphoma 10 years later. This patient also exhibited numerous nonmalignant findings including café au lait spots, lipomas, bilateral renal nodules, a nonossifying fibroma, multiple colonic adenomas, and a rapidly enlarging pilomatrixoma. The spectrum of malignant and nonmalignant neoplasms in this patient highlights the remarkable diversity, and early onset, of lesions seen in children with CMMRD.
- Published
- 2016
50. Trisomy 21-associated defects in human primitive hematopoiesis revealed through induced pluripotent stem cells
- Author
-
Richard L. Nemiroff, Joanna Tober, Paul Gadue, Joanna B. Opalinska, Deborah L. French, Ross C. Hardison, John K. Choi, Stella T. Chou, Nancy A. Speck, Daniel VanDorn, Yu Yao, Jason A. Mills, Marta Byrska-Bishop, and Mitchell J. Weiss
- Subjects
Multidisciplinary ,Cellular differentiation ,Preleukemia ,Biology ,medicine.disease ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,medicine ,Erythropoiesis ,Myelopoiesis ,Yolk sac ,Trisomy ,Induced pluripotent stem cell - Abstract
Patients with Down syndrome (trisomy 21, T21) have hematologic abnormalities throughout life. Newborns frequently exhibit abnormal blood counts and a clonal preleukemia. Human T21 fetal livers contain expanded erythro-megakaryocytic precursors with enhanced proliferative capacity. The impact of T21 on the earliest stages of embryonic hematopoiesis is unknown and nearly impossible to examine in human subjects. We modeled T21 yolk sac hematopoiesis using human induced pluripotent stem cells (iPSCs). Blood progenitor populations generated from T21 iPSCs were present at normal frequency and proliferated normally. However, their developmental potential was altered with enhanced erythropoiesis and reduced myelopoiesis, but normal megakaryocyte production. These abnormalities overlap with those of T21 fetal livers, but also reflect important differences. Our studies show that T21 confers distinct developmental stage- and species-specific hematopoietic defects. More generally, we illustrate how iPSCs can provide insight into early stages of normal and pathological human development.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.