16 results on '"Ravandi F"'
Search Results
2. The Impact of Smoking on Survival in Patients (Pts) with Newly Diagnosed Philadelphia Chromosome Positive (Ph plus ) Acute Lymphoblastic Leukemia (ALL) Treated with the Combination of Intensive Therapy with Tyrosine Kinase Inhibitor (TKI)
- Author
-
Sasaki, K, Ribera, JM, Figueroa, M, Ravandi, F, Short, NJ, Garcia-Manero, G, Daver, NG, Kadia, TM, Konopleva, MY, Jain, N, Issa, GC, Estrov, ZE, Garris, R, Khouri, R, Nasnas, P, DiNardo, CD, Naqvi, K, Kornblau, SM, Montalban-Bravo, G, Pemmaraju, N, Cortes, JE, O'Brien, SM, Chandra, J, Kantarjian, HM, and Jabbour, E
- Published
- 2019
3. Prolonged survival in patients >= 60 years with first relapsed/refractory acute myeloid leukemia treated with vosaroxin plus cytarabine vs placebo plus cytarabine: Results from the phase 3 VALOR study
- Author
-
Krug, U, Ravandi, F, Ritchie, EK, Sayar, H, Lancet, JE, Craig, MD, Vey, N, Strickland, SA, Schiller, GJ, Jabbour, E, Erba, HP, Pigneux, A, Horst, H-A, Recher, C, Klimek, VM, Cortes, J, Roboz, GJ, Craig, AR, Fox, JA, Ward, R, Smith, JA, Acton, G, Mehta, C, Kantarjian, HM, and Stuart, RK
- Published
- 2015
4. Improvement in Clinical Outcome of FLT3 Mutated AML Patients over the Last One and a Half Decade
- Author
-
Talha Badar, Kantarjian H, Borthakur G, Garcia-Manero G, Andreeff M, Konopleva M, Kadia T, Daver N, Wierda W, Ravandi F, and Cortes J
- Published
- 2014
- Full Text
- View/download PDF
5. Clinical Outcome of De Novo Adult Acute Lymphoblastic Leukemia (ALL) with 11q23/Mixed Lineage Leukemia (MLL) Gene Rearrangements
- Author
-
Talha Badar, Kantarjian HM, O'Brien S, Garcia-Manero G, Jabbour E, Garris R, Pemmaraju N, Daver N, Ravandi F, Cortes J, and DA, Thomas
- Published
- 2014
- Full Text
- View/download PDF
6. Phase 1 and Extension Study of Clofarabine plus Cyclophosphamide in Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL)
- Author
-
Faderl, S, Balakrishnan, K, Thomas, DA, Ravandi, F, Borthakur, G, Burger, J, Ferrajoli, A, Cortes, J, O’Brien, S, Kadia, T, Feliu, J, Plunkett, W, Gandhi, V, and Kantarjian, HM
- Subjects
Adult ,Male ,Maximum Tolerated Dose ,Adenine Nucleotides ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Drug Administration Schedule ,Young Adult ,Treatment Outcome ,Recurrence ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Female ,Arabinonucleosides ,Cyclophosphamide ,Aged ,Clofarabine - Abstract
Clofarabine is a nucleoside analogue with activity in children with acute lymphoblastic leukemia (ALL). Based on the hypothesis that clofarabine inhibits DNA repair after exposure to DNA-damaging agents, we designed a phase I and extension study to evaluate the combination of clofarabine and cyclophosphamide in adult patients with relapsed/refractory ALL.The continual reassessment method (CRM) was used to define the maximum tolerated dose (MTD).Fifty patients with a median age of 30 years (range, 21-72 years) were enrolled, 30 of whom were part of the phase I group. Clofarabine 40 mg/m(2) intravenously daily × 3 days and cyclophosphamide 200 mg/m(2) intravenously every 12 hours × 3 days were established as the MTDs. Dose limiting toxicity (DLT) included diarrhea, transaminase elevations, and skin rashes. The response rate of the whole study group was 14%, including 10% of patients who achieved complete remission (CR) or CR without platelet recovery (CRp). Three responses occurred in patients with primary refractory disease. Early mortality (30 days) was 6%. The median duration of response was 69 days (range, 5-315 days). Median overall survival was about 3 months. Compared with day 1 (cyclophosphamide alone), H2AX phosphorylation was increased on day 2 when clofarabine and cyclophosphamide were administered as a couplet (n = 8).The combination of clofarabine plus cyclophosphamide at the doses used in this study in a group of heavily pretreated patients with ALL is only moderately effective. Other doses, alternative schedules, or a more favorable patient population may achieve better results.
- Published
- 2013
7. A Phase I Study of a Combination of anti-CD19 and anti-CD22 Immunotoxins (Combotox) in Adult Patients with Refractory B-Lineage Acute Lymphoblastic Leukaemia
- Author
-
Schindler, J., Gajavelli, S., Ravandi, F., Shen, Y., Parekh, S., Braunchweig, I., Barta, S, Ghetie, V., Vitetta, E., and Verma, A.
- Subjects
Adult ,Male ,Dose-Response Relationship, Drug ,Immunotoxins ,Sialic Acid Binding Ig-like Lectin 2 ,Antigens, CD19 ,Antibodies, Monoclonal ,Antineoplastic Agents ,Ricin ,Middle Aged ,Article ,Young Adult ,Treatment Outcome ,Recurrence ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Humans ,Female ,Aged - Abstract
Novel agents are needed for patients with refractory and relapsed acute lymphoblastic leukaemia (ALL). Combotox is a 1:1 mixture of two immunotoxins (ITs), prepared by coupling deglycosylated ricin A chain (dgRTA) to monoclonal antibodies directed against CD22 (RFB4-dgRTA) and CD19 (HD37-dgRTA). Pre-clinical data demonstrated that Combotox was effective in killing both pre-B-ALL cell lines and cells from patients with pre-B ALL. A clinical study of paediatric patients in which 3 of 17 patients with ALL experienced complete remission, supported the preclinical work and motivated this study. This study was a Phase I, dose-escalation trial using Combotox in adults with refractory or relapsed B-lineage-ALL. A cycle consisted of three doses, with one dose given every other day. Dose levels were 3, 5, 6, 7 and 8 mg/m(2) per dose. Seventeen patients, aged 19-72 years, were enrolled in this multi-institution study. The maximum tolerated dose was 7 mg/m(2) /dose (21 mg/m(2) /cycle) and vascular leak syndrome was the dose-limiting toxicity. Two patients developed reversible grade 3 elevations in liver function tests. One patient achieved partial remission and proceeded to allogeneic stem cell transplantation. All patients with peripheral blasts experienced decreased blast counts following the administration of Combotox. Thus, Combotox can be safely administered to adults with refractory leukaemia.
- Published
- 2011
8. Clinical Impact of Dose Reductions and Interruptions of Second-Generation Tyrosine Kinase Inhibitors in Patients with Chronic Myeloid Leukaemia
- Author
-
Santos, Fp, Kantarjian, H, Fava, Carmen, O'Brien, S, Garcia Manero, G, Ravandi, F, Wierda, W, Thomas, D, Shan, J, and Cortes, J.
- Subjects
Adult ,Aged, 80 and over ,Male ,Adolescent ,Dasatinib ,Antineoplastic Agents ,Middle Aged ,Protein-Tyrosine Kinases ,Survival Analysis ,Article ,Drug Administration Schedule ,Thiazoles ,Young Adult ,Pyrimidines ,Treatment Outcome ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Humans ,Female ,Protein Kinase Inhibitors ,Aged - Abstract
Second (2nd)-generation tyrosine kinase inhibitors (TKI) (dasatinib, nilotinib) are effective in patients with all phases of chronic myeloid leukaemia (CML). Dose reductions and treatment interruptions are frequently required due to toxicity, but their significance is unknown. We analysed the impact of dose reductions/interruptions and dose intensity of 2nd-generation TKI on response and survival. A total of 280 patients with CML (all phases) were analysed. Dose reductions were considered when the daily dose was below the standard dose. Dose intensity was determined based on the percentage of the ideal dose intensity. Overall, 176 patients (63%) required treatment interruptions and/or dose reduction at least once during therapy. Dose reductions/interruptions, analysed as a time-dependent covariate, were associated with worse failure-free survival only in patients with untreated CML. Dose intensity analysis did not reveal a worse response or survival in patients who received a lower dose intensity (
- Published
- 2010
9. IMPROVED SURVIVAL IN PATIENTS >= 60 WITH FIRST RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA TREATED WITH VOSAROXIN PLUS CYTARABINE VS PLACEBO PLUS CYTARABINE: RESULTS FROM THE PHASE 3 VALOR STUDY
- Author
-
Carella, A. -M, Ravandi, F., Ritchie, E. K., Sayar, H., Lancet, J. E., Craig, M. D., Norbert Vey, Strickland, S. A., Schiller, G. J., Jabbour, E., Erba, H. P., Pigneux, A., Horst, H. -A, Recher, C., Klimek, V. M., Cortes, J., Roboz, G. J., Craig, A. R., Fox, J. A., Ward, R., Smith, J. A., Acton, G., Mehta, C., Kantarjian, H. M., and Stuart, R. K.
10. Acute myeloid leukemia: Clinical practice guidelines in oncology
- Author
-
O Donnell, M. R., Abboud, C. N., Altman, J., Appelbaum, F. R., Coutre, S. E., Damon, L. E., Foran, J. M., Goorha, S., Maness, L. J., Marcucci, G., Maslak, P., Millenson, M. M., Moore, J. O., Ravandi, F., Shami, P. J., Smith, B. D., Stone, R. M., Stephen Strickland, Tallman, M. S., Wang, E. S., Gregory, K., and Kumar, R.
11. Resistance prediction in AML: analysis of 4601 patients from MRC/NCRI, HOVON/SAKK, SWOG and MD Anderson Cancer Center
- Author
-
Hills, R K, Pabst, Thomas, Ravandi, F, Estey, E H, Pierce, S R, Walter, R B, Othus, M, Burnett, A K, Kantarjian, H M, Evans, A, Vekemans, M-C, Appelbaum, F R, Ossenkoppele, G J, and Löwenberg, B
- Subjects
610 Medicine & health ,3. Good health - Abstract
Therapeutic resistance remains the principal problem in acute myeloid leukemia (AML). We used area under receiver-operating characteristic curves (AUCs) to quantify our ability to predict therapeutic resistance in individual patients, where AUC=1.0 denotes perfect prediction and AUC=0.5 denotes a coin flip, using data from 4601 patients with newly diagnosed AML given induction therapy with 3+7 or more intense standard regimens in UK Medical Research Council/National Cancer Research Institute, Dutch–Belgian Cooperative Trial Group for Hematology/Oncology/Swiss Group for Clinical Cancer Research, US cooperative group SWOG and MD Anderson Cancer Center studies. Age, performance status, white blood cell count, secondary disease, cytogenetic risk and FLT3-ITD/NPM1 mutation status were each independently associated with failure to achieve complete remission despite no early death (‘primary refractoriness’). However, the AUC of a bootstrap-corrected multivariable model predicting this outcome was only 0.78, indicating only fair predictive ability. Removal of FLT3-ITD and NPM1 information only slightly decreased the AUC (0.76). Prediction of resistance, defined as primary refractoriness or short relapse-free survival, was even more difficult. Our limited ability to forecast resistance based on routinely available pretreatment covariates provides a rationale for continued randomization between standard and new therapies and supports further examination of genetic and posttreatment data to optimize resistance prediction in AML.
12. Chronic myeloid leukemia 2011: Successes, challenges, and strategies-Proceedings of the 5th annual BCR-ABL1 positive and BCR-ABL1 negative myeloproliferative neoplasms workshop
- Author
-
Jerald P. Radich, Tariq I. Mughal, Giovanni Martinelli, Ayalew Tefferi, Daniel J. DeAngelo, Alfonso Quintás-Cardama, Carlo Gambacorti-Passerini, Richard A. Van Etten, Farhad Ravandi, Tomasz Skorski, Mughal T.I., Radich J.P., Van Etten R.A., Quintás-Cardama A., Skorski T., Ravandi F., Deangelo D.J., Gambacorti-Passerini C., Martinelli G., Tefferi A., Mughal, T, Radich, J, Van Etten, R, Quintás Cardama, A, Skorski, T, Ravandi, F, Deangelo, D, GAMBACORTI PASSERINI, C, Martinelli, G, and Tefferi, A
- Subjects
medicine.medical_specialty ,Gene mutation ,Article ,Genomic Instability ,Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative ,Antineoplastic Agent ,chemistry.chemical_compound ,Bcr abl1 ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animal ,business.industry ,Ponatinib ,Myeloid leukemia ,Imatinib ,Hematology ,Dasatinib ,Imatinib mesylate ,chemistry ,Drug Resistance, Neoplasm ,Family medicine ,Immunology ,Drug Monitoring ,business ,Bosutinib ,CHRONIC MYELOID LEUKEMIA (CML) ,Human ,medicine.drug - Abstract
NIH Public Access Author Manuscript Am J Hematol. Author manuscript; available in PMC 2012 November 01. Published in final edited form as: Am J Hematol. 2011 September ; 86(9): 811–819. doi:10.1002/ajh.22097. $watermark-text Chronic Myeloid Leukemia 2011: Successes, challenges, and strategies – Proceedings of the 5 th Annual BCR-ABL1 positive and BCR-ABL1 negative myeloproliferative neoplasms workshop Tariq I Mughal 1 , Jerald P Radich 2 , Richard A. Van Etten 3 , Alfonso Quintas-Cardama 4 , Tomasz Skorski 5 , Farhad Ravandi 6 , Daniel J. DeAngelo 7 , Carlo Gambacorti-Passerini 8 , Giovanni Martinelli 9 , and Ayalew Tefferi 10 1 University of Colorado School of Medicine, Denver, USA 2 Fred Hutchinson Cancer Center, Seattle, USA 3 Tufts Medical Center, Boston, USA 4 MD Anderson Cancer Center, Houston, USA $watermark-text 5 Temple 6 MD University, Philadelphia, USA Anderson Cancer Center, Houston, USA 7 Dana Farber Cancer Center at Harvard Medical School, Boston, USA 8 University 9 Insitute 10 Mayo of Milan, Monza, Italy Le A. Seragnoli, Bologna, Italy Clinic, Rochester, MN, USA. Abstract $watermark-text This report is based on the presentations and discussions at the 5 th annual BCR-ABL1 positive and BCR-ABL1 negative myeloproliferative neoplasms (MPN) workshop, which took place immediately following the 52 nd American Society of Hematology (ASH) meeting in Orlando, Florida on December 7 th -8 th , 2011. Relevant data which was presented at the ASH meeting as well as all other recent publications were presented and discussed at the workshop. This report covers front-line therapies of BCR-ABL1-positive leukemias, in addition to addressing some topical biological, pre-clinical and clinical issues, such as new insights into genomic instability and resistance to tyrosine kinase inhibitors (TKIs), risk stratification and optimizing molecular monitoring. A report pertaining to the new therapies and other pertinent preclinical and clinical issues in the BCR-ABL1 negative MPNs is published separately. Introduction For patients with BCR-ABL1-positive leukemias, which comprise of all the Philadelphia (Ph) chromosome-positive and some Ph-negative leukemias, the introduction of the original tyrosine kinase inhibitor (TKI), imatinib mesylate (IM) into the clinics in 1998, resulted in being a classic therapeutic landmark (1,2). After 12 months of therapy with IM, 69% of Correspondence to: Tariq Mughal MD FRCP FACP 23655 Currant Drive, Golden, Colorado 80401, USA Tel +1 303 526 8586 tmughal911@hotmail.com.
- Published
- 2011
13. Hairy cell leukemia and COVID-19 adaptation of treatment guidelines
- Author
-
Francesco Forconi, Jae H. Park, Martin S. Tallman, Brunangelo Falini, Robert J. Kreitman, James B. Johnston, Sameer A. Parikh, Timothy G. Call, Xavier Troussard, Seema A. Bhat, James S. Blachly, Sasha Dietrich, Gerard Lozanski, Matthew Cross, Jacqueline C. Barrientos, Thorsten Zenz, Claire Dearden, Sunil Iyengar, Alan Saven, Francesco Lauria, Judit Demeter, Gunnar Juliusson, Tadeusz Robak, Douglas E. Gladstone, Versha Banerji, Kerry A. Rogers, Enrico Tiacci, Tamar Tadmor, Pier Luigi Zinzani, John F. Seymour, Farhad Ravandi, Bernhard Wörmann, Constantine S. Tam, Michael R. Grever, Aaron Polliack, Alessandro Gozzetti, Clive S. Zent, Eric H. Kraut, Leslie A. Andritsos, Grever M., Andritsos L., Banerji V., Barrientos J.C., Bhat S., Blachly J.S., Call T., Cross M., Dearden C., Demeter J., Dietrich S., Falini B., Forconi F., Gladstone D.E., Gozzetti A., Iyengar S., Johnston J.B., Juliusson G., Kraut E., Kreitman R.J., Lauria F., Lozanski G., Parikh S.A., Park J., Polliack A., Ravandi F., Robak T., Rogers K.A., Saven A., Seymour J.F., Tadmor T., Tallman M.S., Tam C.S., Tiacci E., Troussard X., Zent C., Zenz T., Zinzani P.L., and Wormann B.
- Subjects
Cancer Research ,medicine.medical_specialty ,Consensus ,Hairy Cell ,medicine.medical_treatment ,Diseases ,Consensu ,Review Article ,Disease ,Severity of Illness Index ,Internal medicine ,medicine ,Leukaemia ,Humans ,Hairy cell leukemia ,Intensive care medicine ,Cladribine ,Pandemics ,Leukemia, Hairy Cell ,Leukemia ,Hematology ,Pandemic ,SARS-CoV-2 ,business.industry ,Standard treatment ,COVID-19 ,Immunosuppression ,Practice Guidelines as Topic ,medicine.disease ,Oncology ,business ,Human ,medicine.drug - Abstract
Standard treatment options in classic HCL (cHCL) result in high response rates and near normal life expectancy. However, the disease itself and the recommended standard treatment are associated with profound and prolonged immunosuppression, increasing susceptibility to infections and the risk for a severe course of COVID-19. The Hairy Cell Leukemia Foundation (HCLF) has recently convened experts and discussed different clinical strategies for the management of these patients. The new recommendations adapt the 2017 consensus for the diagnosis and management with cHCL to the current COVID-19 pandemic. They underline the option of active surveillance in patients with low but stable blood counts, consider the use of targeted and non-immunosuppressive agents as first-line treatment for cHCL, and give recommendations on preventive measures against COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
14. AML-389 Phase 1/2 Study of SEL24/MEN1703, a First-in-Class Dual PIM/FLT3 Kinase Inhibitor, in Patients With IDH1/2Mutated Acute Myeloid Leukemia: The DIAMOND-01 Trial
- Author
-
Giovanni Martinelli, Armando Santoro, Carlo Gambacorti-Passerini, Susana Vives Polo, Scott R. Solomon, Sudipto Mukherjee, Ewa Lech-Maranda, Moshe Yair Levy, Agnieszka Wierzbowska, María Calbacho-Robles, Giovanni Marconi, Maria Benedetta Giannini, Isabel Cano, Laura Torres Miñana, Evelyn Acuña-Cruz, Noemi Angelosanto, Tariq I. Mughal, Antonio Galleu, Simona Blotta, Farhad Ravandi, Pau Montesinos, Martinelli, G, Santoro, A, Gambacorti Passerini, C, Polo, S, Solomon, S, Mukherjee, S, Lech-Maranda, E, Levy, M, Wierzbowska, A, Calbacho-Robles, M, Marconi, G, Giannini, M, Cano, I, Minana, L, Acuna-Cruz, E, Angelosanto, N, Mughal, T, Galleu, A, Blotta, S, Ravandi, F, and Montesinos, P
- Subjects
Cancer Research ,Oncology ,AML ,FLT3-ITD, FLT3 inhibitor ,Trial-in-Progre ,Hematology ,acute myeloid leukemia ,IDH mutation ,PIM kinase - Abstract
Context: Mutations in the FLT3 tyrosine kinase and in IDH1/IDH2 (collectively IDHm) co-occur in up to 30% of adults with acute myeloid leukemia (AML). SEL24/MEN1703 is an orally available, first-in-class, dual PIM/FLT3 kinase inhibitor. Preliminary results from the phase 1/2 first-in-human DIAMOND-01 trial (NCT03008187) demonstrated antitumor activity of single-agent SEL24/MEN1703 in adult patients with relapsed/refractory (R/R) IDHm AML, where 3 of 8 IDHm patients responded. Objective: To report the first safety and efficacy results from an additional expansion cohort of the DIAMOND-01 trial in 20 patients with R/R IDHm AML. Design: DIAMOND-01 is a phase 1/2, open-label, multicenter study consisting of 2 parts: dose escalation and cohort expansion, including an additional expansion cohort (IDHm) that is ongoing. Patients: Patients with R/R IDHm AML and no standard therapeutic options were eligible. Intervention(s): Patients received the recommended dose of 125 mg SEL24/MEN1703 orally, once daily for 14 days over a 21-day cycle until disease progression or unacceptable toxicity. Main Outcome Measure(s): The number and frequency of adverse events (AEs; primary) and overall response rate (ORR; secondary). Results: As of 10 January 2022, 14 patients were enrolled in the IDHm cohort. Seven patients had IDH2, 1 had IDH1/2, and 4 had IDH1 mutations. Two patients had a concomitant FLT3-ITD mutation. Safety data (N=12) showed that grade ≥3 TEAEs (≥10% of patients) were pneumonia (33%) and asthenia (17%), both unrelated to the study drug. Of the 7 patients who completed ≥1 treatment cycle and had ≥1 post-baseline assessment or clear disease progression, ORR was 28.6%; 1 patient achieved a complete response with incomplete blood count recovery at cycle 3 and underwent hematopoietic stem cell transplant, and 1 patient had a partial response at cycle 4 (confirmed at cycle 7 and still on treatment). Among the 7 remaining patients, 3 discontinued before completion of cycle 1 without progression or response, and 4 patients are ongoing and have not yet undergone post-baseline assessments. Conclusions: SEL24/MEN1703 had a manageable safety profile and single-agent activity in adult patients with R/R IDHm AML and may be a feasible therapeutic option in this difficult-to-treat population.
- Published
- 2022
15. An Inv(16)(p13.3q24.3)-Encoded CBFA2T3-GLIS2 Fusion Protein Defines an Aggressive Subtype of Pediatric Acute Megakaryoblastic Leukemia
- Author
-
Timothy J. Ley, James R. Downing, Xiaoping Su, Andrea Biondi, Jianmin Wang, Li Ding, Stanley Pounds, Tanja A. Gruber, Matthew Parker, Der Cherng Liang, Heather L. Mulder, Giovanni Cazzaniga, Jeffrey E. Rubnitz, Michael Rusch, Paola Ballerini, John Easton, Hartmut Döhner, Ching-Hon Pui, Ramapriya Ganti, Michael I. Barbato, Sheila A. Shurtleff, Farhad Ravandi, Jinghui Zhang, Richard K. Wilson, Yasuhide Hayashi, Lee Yung Shih, Huy Q. Ta, Ina Radtke, Steven M. Kornblau, Stacey K. Ogden, Amanda Larson Gedman, Anna Andersson, Daisuke Tomizawa, Jayanthi Manne, Vedant Gupta, Swati Ranade, Akio Tawa, Stephen D. Nimer, Shann Ching Chen, Gang Wu, Souichi Adachi, Konstanze Döhner, Lei Shi, Jing Ma, Suresh Marada, Jinjun Dang, Elaine R. Mardis, Hagop M. Kantarjian, Cary Koss, Gruber, T, Larson Gedman, A, Zhang, J, Koss, C, Marada, S, Ta, H, Chen, S, Su, X, Ogden, S, Dang, J, Wu, G, Gupta, V, Andersson, A, Pounds, S, Sh, L, Easton, J, Barbato, M, Mulder, H, Manne, J, Wang, J, Rusch, M, Ranade, S, Ganti, R, Parker, M, Ma, J, Radtke, I, Ding, L, Cazzaniga, G, Biondi, A, Kornblau, S, Ravandi, F, Kantarjian, H, Nimer, S, Döhner, K, Döhner, H, Ley, T, Ballerini, P, Shurtleff, S, Tomizawa, D, Adachi, S, Hayashi, Y, Tawa, A, Shih, L, Liang, D, Rubnitz, J, Pui, C, Mardis, E, Wilson, R, and Downing, J
- Subjects
0303 health sciences ,Mutation ,Cancer Research ,Cell Biology ,Biology ,medicine.disease_cause ,medicine.disease ,Bone morphogenetic protein ,Fusion protein ,Molecular biology ,Article ,3. Good health ,Gene expression profiling ,03 medical and health sciences ,Acute megakaryoblastic leukemia ,Haematopoiesis ,0302 clinical medicine ,Chromosome 16 ,AML ,Oncology ,030220 oncology & carcinogenesis ,medicine ,030304 developmental biology ,Chromosomal inversion - Abstract
To define the mutation spectrum in non-Down syndrome acute megkaryoblastic leukemia (non-DS-AMKL), we performed transcriptome sequencing on diagnostic blasts from 14 pediatric patients and validated our findings in a recurrency/validation cohort consisting of 34 pediatric and 28 adult AMKL leukemia samples. Our analysis identified a cryptic chromosome 16 inversion [inv(16)(p13.3q24.3)] in 27% of pediatric cases, which encodes a CBFA2T3-GLIS2 fusion protein. Expression of CBFA2T3-GLIS2 in Drosophila and murine hematopoietic cells induced bone morphogenic protein (BMP) signaling, and resulted in a marked increase in the self-renewal capacity of hematopoietic progenitors. These data suggest that expression of CBFA2T3-GLIS2 directly contributes to leukemogenesis.
- Published
- 2012
- Full Text
- View/download PDF
16. Transcriptome Sequence Analysis of Pediatric Acute Megakaryoblastic Leukemia Identifies An Inv(16)(p13.3;q24.3)-Encoded CBFA2T3-GLIS2 Fusion Protein As a Recurrent Lesion in 39% of Non-Infant Cases: A Report From the St. Jude Children's Research Hospital – Washington University Pediatric Cancer Genome Project
- Author
-
Richard K. Wilson, John Easton, Vedant Gupta, Michael Rusch, Konstanze Doehner, Huy Q. Ta, Farhad Ravandi, Tanja A. Gruber, Jinghui Zhang, Jeffrey E. Rubnitz, Sheila A. Shurtleff, Hartmut Doehner, Shann-Ching Chen, Timothy J. Ley, Stephen D. Nimer, Anna Andersson, Xiaoping Su, Elaine R. Mardis, Li Ding, Stacey K. Ogden, Amanda Larson Gedman, Giovanni Cazzaniga, Hagop M. Kantarjian, Cary Koss, Andrea Biondi, Steven M. Kornblau, Ching-Hon Pui, James R. Downing, Jianmin Wang, Gruber, T, Gedman, A, Ta, H, Zhang, J, Koss, C, Chen, S, Su, X, Gupta, V, Ogden, S, Andersson, A, Easton, J, Wang, J, Rusch, M, Ding, L, Cazzaniga, G, Biondi, A, Kornblau, S, Ravandi, F, Kantarjian, H, Nimer, S, Doehner, K, Doehner, H, Ley, T, Shurtleff, S, Rubnitz, J, Pui, C, Mardis, E, Wilson, R, and Downing, J
- Subjects
Myeloid ,Immunology ,GATA2 ,Transcriptome Sequence Analysis ,Cell Biology ,Hematology ,Chimeric gene ,Biology ,medicine.disease ,Biochemistry ,Fusion protein ,Pediatric cancer ,Leukemia ,Acute megakaryoblastic leukemia ,medicine.anatomical_structure ,Pediatric Acute Megakaryoblastic Leukemia ,FLI1 ,medicine ,Cancer research - Abstract
Abstract 757 Acute Megakaryoblastic Leukemia (AMKL) accounts for ∼10% of childhood acute myeloid leukemia (AML). Although AMKL patients with down syndrome (DS-AMKL) have an excellent 5 year event-free survival (EFS), non-DS-AMKL patients have an extremely poor outcome with a 3 year EFS of less than 40%. With the exception of the t(1;22) translocation seen in infant non-DS-AMKL, little is known about the molecular genetic lesions that underlie this leukemia subtype. To define the landscape of mutations that occur in non-DS-AMKL, we performed transcriptome sequencing on diagnostic blasts from 14 cases (discovery cohort) using the illumina platform. Our results identified chromosomal rearrangements resulting in the expression of novel fusion transcripts in 12/14 cases. Remarkably, in 7/14 cases we detected an inversion on chromosome 16 [inv(16)(p13.3;q24.3)] that resulted in the juxtaposition of the CBFA2T3, a member of the ETO family of transcription factors, next to GLIS2 resulting in a CBFA2T3-GLIS2 chimeric gene encoding an in frame fusion protein. 6 cases in the discovery cohort fused exon 10 of CBFA2T3 to exon 3 of GLIS2, while 1 case carried a larger product that fused exon 11 of CBFA2T3 to exon 1 of GLIS2. Both products retain the 3 CBFA2T3 N-terminal nervy homology regions that mediate protein interactions, and the 5 GLIS2 C-terminal zinc finger domains that bind the Glis DNA consensus sequence, along with one of its N-terminal transcriptional regulatory domains. GLIS2 is a member of the GLI super family of transcription factors and has been demonstrated to play a role in regulating expression of GLI target genes as well as inhibiting WNT signaling through the binding of beta catenin. Although GLIS2 is not normally expressed in hematopoietic cells, the translocation results in high level expression of the CBFA2T3-GLIS2 fusion protein. In addition to CBFA2T3-GLIS2, chimeric transcripts were detected in 6/7 cases that lacked evidence of the inv(16)(p13.3;q24.3). Specifically, we detected GATA2-HOXA9, MN1-FLI1, NIPBL-HOXB9, NUP98-KDM5A, GRB10-SDK1 and C8orf76-HOXA11AS, each in an individual case. Importantly, several of the genes involved in these translocations either play a direct role in normal megakaryocytic differentiation (GATA2 and FLI1), or have been previously shown to be involved in leukemogenesis (HOXA9, MN1, HOXB9). Evaluation of a recurrency cohort of 42 samples including 14 additional pediatric cases and 28 adult cases by RT-PCR revealed 4 additional pediatric samples carrying CBFA2T3-GLIS2 for an overall frequency of 39% in pediatric AMKL. In addition to these somatic structural variations, we also identified mutations in genes previously shown to play a role in megakaryoblastic leukemia including activating mutations in JAK2 and MPL (36%). To gain insight into the mechanism whereby CBFA2T3-GLIS2 promotes leukemogenesis, we introduced the fusion into murine hematopoietic cells and assessed its effect on in vitro colony replating as a surrogate measure of self-renewal. Hematopoietic cells transduced with a mCherry expressing retroviral vector failed to form colonies after the second replating. By contrast, expression of either wild-type GLIS2 or the CBFA2T3-GLIS2 fusion resulted in a marked increase in the self-renewal capacity, with colony formation persisting through eight replatings. Immunophenotypic analysis of the CBFA2T3-GLIS2 expressing colonies revealed evidence of megakaryocytic differentiation. Importantly, the CBFA2T3-GLIS2 cells remained growth factor dependent suggesting that cooperating mutations in growth factor signaling pathways are required for full leukemic transformation. Taken together these data identify a novel cryptic inv(16)-encoded CBFA2T3-GLIS2 fusion protein as a recurrent driver mutation in approximately 40% of non-infant pediatric non-DS-AMKLs. Moreover, the majority of pediatric cases that lacked this lesion were shown by transcriptome sequence analysis to contain other chromosomal rearrangements that encoded fusion proteins that directly alter megakaryocytic differentiation and/or myeloid cell growth. The alteration of a key transcriptional regulator within the hedgehog signaling pathways in a substantial percentage of pediatric AMKL raises the possibility that inhibition of this pathway may have a therapeutic benefit in this aggressive form of AML. *TAG and ALG contributed equally to this work. Disclosures: Biondi: BMS, Novartis, Micromed: Consultancy, Membership on an entity's Board of Directors or advisory committees. Ravandi:Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria. Kantarjian:Novartis: Consultancy, Research Funding; Pfizer: Research Funding; BMS: Research Funding. Doehner:Hoffmann La Roche: Honoraria.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.