109 results on '"G. Garcia"'
Search Results
2. Mutant IL7R collaborates with MYC to induce T-cell acute lymphoblastic leukemia
- Author
-
Mariana L. Oliveira, Alexandra Veloso, Elaine G. Garcia, Sowmya Iyer, Clara Pereira, Vasco M. Barreto, David M. Langenau, João T. Barata, Repositório da Universidade de Lisboa, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), and Centro de Estudos de Doenças Crónicas (CEDOC)
- Subjects
EXPRESSION ,Cancer Research ,Carcinogenesis ,T-Lymphocytes ,BIOLOGY ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,ACTIVATION ,Animals, Genetically Modified ,Interleukin-7 Receptor alpha Subunit ,SDG 3 - Good Health and Well-being ,NOTCH1 ,OF-FUNCTION MUTATIONS ,C-MYC ,KINASE ,Animals ,Humans ,Child ,Zebrafish ,Science & Technology ,IL-7 ,Receptors, Interleukin-7 ,Hematology ,GENE ,Oncology ,GROWTH ,Life Sciences & Biomedicine ,Signal Transduction - Abstract
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/., T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive pediatric cancer. Amongst the wide array of driver mutations, 10% of T-ALL patients display gain-of-function mutations in the IL-7 receptor α chain (IL-7Rα, encoded by IL7R), which occur in different molecular subtypes of this disease. However, it is still unclear whether IL-7R mutational activation is sufficient to transform T-cell precursors. Also, which genes cooperate with IL7R to drive leukemogenesis remain poorly defined. Here, we demonstrate that mutant IL7R alone is capable of inducing T-ALL with long-latency in stable transgenic zebrafish and transformation is associated with MYC transcriptional activation. Additionally, we find that mutant IL7R collaborates with Myc to induce early onset T-ALL in transgenic zebrafish, supporting a model where these pathways collaborate to drive leukemogenesis. T-ALLs co-expressing mutant IL7R and Myc activate STAT5 and AKT pathways, harbor reduced numbers of apoptotic cells and remake tumors in transplanted zebrafish faster than T-ALLs expressing Myc alone. Moreover, limiting-dilution cell transplantation experiments reveal that activated IL-7R signaling increases the overall frequency of leukemia propagating cells. Our work highlights a synergy between mutant IL7R and Myc in inducing T-ALL and demonstrates that mutant IL7R enriches for leukemia propagating potential., This work was supported by NIH grant R01CA211734 (DML), the MGH Research Scholar Award (DML), the ERC consolidator CoG-648455 and proof-of-concept PoC-862545 grants from the European Research Council, under the European Union’s Horizon 2020 research and innovation programme (JTB), and the FCT grants FAPESP/ 20015/2014, PTDC/MEC-HEM/31588/2017 and PTDC/MEC-ONC/4606/2021 (JTB)
- Published
- 2021
3. Cell of origin dictates aggression and stem cell number in acute lymphoblastic leukemia
- Author
-
Franki Speleman, Elaine G. Garcia, Sara P. Garcia, David M. Langenau, Sowmya Iyer, Siebe Loontiens, and Ruslan I. Sadreyev
- Subjects
0301 basic medicine ,Cancer Research ,T-Lymphocytes ,Lymphoblastic Leukemia ,Cell of origin ,Biology ,Article ,Animals, Genetically Modified ,03 medical and health sciences ,Transgenic zebrafish ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cell Lineage ,Zebrafish ,B-Lymphocytes ,Aggression ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Clone Cells ,030104 developmental biology ,Oncology ,Neoplastic Stem Cells ,Cancer research ,medicine.symptom ,Stem cell - Published
- 2018
4. Achievement of a negative minimal residual disease state after hypomethylating agent therapy in older patients with AML reduces the risk of relapse
- Author
-
Marina Konopleva, Courtney D. DiNardo, Jeffrey L. Jorgensen, Farhad Ravandi, Mark Brandt, Musa Yilmaz, Prithviraj Bose, Sherry Pierce, E. Jabbour, G. Garcia-Manero, G. Borthakur, Naveen Pemmaraju, Prajwal Boddu, Naval Daver, Yesid Alvarado, Kiran Naqvi, Hagop M. Kantarjian, Jorge E. Cortes, and Tapan M. Kadia
- Subjects
Male ,Risk ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Myeloid ,Antineoplastic Agents ,Methylation ,03 medical and health sciences ,0302 clinical medicine ,Neoplasm Recurrence ,Older patients ,Recurrence ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Medicine ,Relapse risk ,Psychiatry ,neoplasms ,Aged ,Aged, 80 and over ,business.industry ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Minimal residual disease ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Hypomethylating agent ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business ,030215 immunology - Abstract
Achievement of a negative minimal residual disease state after hypomethylating agent therapy in older patients with AML reduces the risk of relapse
- Published
- 2017
5. A phase 1b/2b multicenter study of oral panobinostat plus azacitidine in adults with MDS, CMML or AML with ⩽30% blasts
- Author
-
Zita Borbényi, Suddhasatta Acharyya, Karen W.L. Yee, Je-Hwan Lee, Massimo Breccia, S. Ide, Árpád Illés, Alan Macwhannell, Carlos Graux, Miklos Egyed, Nancy Zhu, David Valcárcel, James D. Cavenagh, Reinhard Stauder, Huda Salman, Daniel J. DeAngelo, Mikkael A. Sekeres, M. Marker, Oliver G. Ottmann, G. Garcia-Manero, Lucien Gazi, and Pierre Fenaux
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Myeloid ,Indoles ,Administration, Oral ,Kaplan-Meier Estimate ,Hydroxamic Acids ,Gastroenterology ,chemistry.chemical_compound ,0302 clinical medicine ,Bone Marrow ,Antineoplastic Combined Chemotherapy Protocols ,Panobinostat ,Aged, 80 and over ,Leukemia, Myelomonocytic, Chronic ,Hematology ,Orvostudományok ,Middle Aged ,3. Good health ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Azacitidine ,Original Article ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Maximum Tolerated Dose ,Chronic myelomonocytic leukemia ,Klinikai orvostudományok ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,neoplasms ,Aged ,business.industry ,Myelodysplastic syndromes ,medicine.disease ,Demethylating agent ,Regimen ,030104 developmental biology ,chemistry ,Myelodysplastic Syndromes ,business - Abstract
Treatment with azacitidine (AZA), a demethylating agent, prolonged overall survival (OS) vs conventional care in patients with higher-risk myelodysplastic syndromes (MDS). As median survival with monotherapy is
- Published
- 2017
6. ITD mutation in FLT3 tyrosine kinase promotes Warburg effect and renders therapeutic sensitivity to glycolytic inhibition
- Author
-
Amin Huang, Yameng Sun, G. Garcia-Manero, Wen Hua Lu, Yizhuo Li, Shijun Wen, Peng Huang, Rui-Hua Xu, G. Zhan, Jing Yang, Huai-Qiang Ju, Jinyuan Li, and Yumin Hu
- Subjects
0301 basic medicine ,Cancer Research ,Tyrosine-kinase inhibitor ,Mice ,chemistry.chemical_compound ,Adenosine Triphosphate ,Hexokinase ,hemic and lymphatic diseases ,Glycolysis ,Molecular Targeted Therapy ,Mice, Inbred BALB C ,Myeloid leukemia ,hemic and immune systems ,Hematology ,Sorafenib ,Warburg effect ,Hydrocarbons, Brominated ,Mitochondria ,Neoplasm Proteins ,Leukemia ,Cell Transformation, Neoplastic ,Oncology ,embryonic structures ,Original Article ,Tyrosine kinase ,psychological phenomena and processes ,Niacinamide ,medicine.drug_class ,Antineoplastic Agents ,Deoxyglucose ,Biology ,Cell Line ,03 medical and health sciences ,medicine ,Animals ,Humans ,Leukemia, Experimental ,Phenylurea Compounds ,Hematopoietic Stem Cells ,medicine.disease ,body regions ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,chemistry ,Anaerobic glycolysis ,Cancer research ,Propionates ,Proto-Oncogene Proteins c-akt ,Microsatellite Repeats - Abstract
Internal tandem duplication (ITD) mutation in Fms-like tyrosine kinase 3 gene (FLT3/ITD) represents an unfavorable genetic change in acute myeloid leukemia (AML) and is associated with poor prognosis. Metabolic alterations have been involved in tumor progression and attracted interest as a target for therapeutic intervention. However, few studies analyzed the adaptations of cellular metabolism in the context of FLT3/ITD mutation. Here, we report that FLT3/ITD causes a significant increase in aerobic glycolysis through AKT-mediated upregulation of mitochondrial hexokinase (HK2), and renders the leukemia cells highly dependent on glycolysis and sensitive to pharmacological inhibition of glycolytic activity. Inhibition of glycolysis preferentially causes severe ATP depletion and massive cell death in FLT3/ITD leukemia cells. Glycolytic inhibitors significantly enhances the cytotoxicity induced by FLT3 tyrosine kinase inhibitor sorafenib. Importantly, such combination provides substantial therapeutic benefit in a murine model bearing FLT3/ITD leukemia. Our study suggests that FLT3/ITD mutation promotes Warburg effect, and such metabolic alteration can be exploited to develop effective therapeutic strategy for treatment of AML with FLT3/ITD mutation via metabolic intervention.
- Published
- 2017
7. Comparison of clinical outcomes and prognostic utility of risk stratification tools in patients with therapy-related vs de novo myelodysplastic syndromes: a report on behalf of the MDS Clinical Research Consortium
- Author
-
Alan F. List, Amy E. DeZern, David P. Steensma, J.E. Lancet, John Barnard, E. Jabbour, Mikkael A. Sekeres, G. Garcia-Manero, Amer M. Zeidan, Eric Padron, N. Al Ali, Gail J. Roboz, and Rami S. Komrokji
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Biomedical Research ,medicine.medical_treatment ,Risk Assessment ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Survival rate ,Neoplasm Staging ,Chemotherapy ,Therapy related ,Hematology ,business.industry ,De novo Myelodysplastic Syndrome ,Middle Aged ,Prognosis ,Combined Modality Therapy ,Survival Rate ,Clinical research ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Risk stratification ,Physical therapy ,Female ,business ,Follow-Up Studies ,030215 immunology - Abstract
While therapy-related (t)-myelodysplastic syndromes (MDS) have worse outcomes than de novo MDS (d-MDS), some t-MDS patients have an indolent course. Most MDS prognostic models excluded t-MDS patients during development. The performances of the International Prognostic Scoring System (IPSS), revised IPSS (IPSS-R), MD Anderson Global Prognostic System (MPSS), WHO Prognostic Scoring System (WPSS) and t-MDS Prognostic System (TPSS) were compared among patients with t-MDS. Akaike information criteria (AIC) assessed the relative goodness of fit of the models. We identified 370 t-MDS patients (19%) among 1950 MDS patients. Prior therapy included chemotherapy alone (48%), chemoradiation (31%), and radiation alone in 21%. Median survival for t-MDS patients was significantly shorter than for d-MDS (19 vs 46 months, P
- Published
- 2017
8. Comparison of risk stratification tools in predicting outcomes of patients with higher-risk myelodysplastic syndromes treated with azanucleosides
- Author
-
Jaroslaw P. Maciejewski, Hagop M. Kantarjian, G. Garcia-Manero, Katrina Zell, Steven D. Gore, Alan F. List, Amer M. Zeidan, Rami S. Komrokji, Gail J. Roboz, Najla Al Ali, Cassie Zimmerman, Mikkael A. Sekeres, E. Jabbour, David P. Steensma, John Barnard, Aziz Nazha, and Amy E. DeZern
- Subjects
Male ,Research design ,Oncology ,Cancer Research ,medicine.medical_specialty ,Databases, Factual ,MEDLINE ,Decitabine ,Antineoplastic Agents ,Article ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,Humans ,Medicine ,Survival analysis ,Aged ,business.industry ,Myelodysplastic syndromes ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,Transplantation ,Clinical research ,Research Design ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Azacitidine ,Physical therapy ,Female ,business ,Follow-Up Studies ,030215 immunology ,medicine.drug - Abstract
Established prognostic tools in patients with myelodysplastic syndromes (MDS) were largely derived from untreated patient cohorts. Although azanucleosides are standard therapies for higher-risk (HR)-MDS, the relative prognostic performance of existing prognostic tools among patients with HR-MDS receiving azanucleoside therapy is unknown. In the MDS Clinical Research Consortium database, we compared the prognostic utility of the International Prognostic Scoring System (IPSS), revised IPSS (IPSS-R), MD Anderson Prognostic Scoring System (MDAPSS), World Health Organization-based Prognostic Scoring System (WPSS) and the French Prognostic Scoring System (FPSS) among 632 patients who presented with HR-MDS and were treated with azanucleosides as the first-line therapy. Median follow-up from diagnosis was 15.7 months. No prognostic tool predicted the probability of achieving an objective response. Nonetheless, all five tools were associated with overall survival (OS, P = 0.025 for the IPSS, P = 0.011 for WPSS and P < 0.001 for the other three tools). The corrected Akaike Information Criteria, which were used to compare OS with the different prognostic scoring systems as covariates (lower is better) were 4138 (MDAPSS), 4156 (FPSS), 4196 (IPSS-R), 4186 (WPSS) and 4196 (IPSS). Patients in the highest-risk groups of the prognostic tools had a median OS from diagnosis of 11 – 16 months and should be considered for up-front transplantation or experimental approaches.
- Published
- 2015
9. Efficacy and safety of extended dosing schedules of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes
- Author
-
Kyle J. MacBeth, Barry S. Skikne, Tao Shi, William Jeffery Edenfield, G. Garcia-Manero, Eric Laille, A Tefferi, Keshava Kumar, Bart L. Scott, Joel Hetzer, Steven D. Gore, Suman Kambhampati, and Christopher R. Cogle
- Subjects
Adult ,Male ,Antimetabolites, Antineoplastic ,Cancer Research ,medicine.medical_specialty ,Azacitidine ,Administration, Oral ,Pharmacology ,Lower risk ,Gastroenterology ,Drug Administration Schedule ,Oral Azacitidine ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Tissue Distribution ,Dosing ,Aged ,Neoplasm Staging ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Therapeutic effect ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Platelet transfusion ,Oncology ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Original Article ,Female ,Safety ,business ,Follow-Up Studies ,030215 immunology ,medicine.drug - Abstract
CC-486, the oral formulation of azacitidine (AZA), is an epigenetic modifier and DNA methyltransferase inhibitor in clinical development for treatment of hematologic malignancies. CC-486 administered for 7 days per 28-day treatment cycle was evaluated in a phase 1 dose-finding study. AZA has a short plasma half-life and DNA incorporation is S-phase-restricted; extending CC-486 exposure may increase the number of AZA-affected diseased target cells and maximize therapeutic effects. Patients with lower-risk myelodysplastic syndromes (MDS) received 300 mg CC-486 once daily for 14 days (n=28) or 21 days (n=27) of repeated 28-day cycles. Median patient age was 72 years (range 31-87) and 75% of patients had International Prognostic Scoring System Intermediate-1 risk MDS. Median number of CC-486 treatment cycles was 7 (range 2-24) for the 14-day dosing schedule and 6 (1-24) for the 21-day schedule. Overall response (complete or partial remission, red blood cell (RBC) or platelet transfusion independence (TI), or hematologic improvement) (International Working Group 2006) was attained by 36% of patients receiving 14-day dosing and 41% receiving 21-day dosing. RBC TI rates were similar with both dosing schedules (31% and 38%, respectively). CC-486 was generally well-tolerated. Extended dosing schedules of oral CC-486 may provide effective long-term treatment for patients with lower-risk MDS.
- Published
- 2015
10. Comprehensive analysis of factors impacting risks and outcomes of therapy-related myeloid neoplasms following breast cancer treatment
- Author
-
JM Reuben, Sara S. Strom, G. Garcia-Manero, Katarina Sevcikova, Hagop M. Kantarjian, L. J. Medeiros, Gabriel N. Hortobagyi, Ricardo H. Alvarez, Joseph D. Khoury, Zuo Zhuang, Constance Albarracin, Guiling Tang, and Michal Mego
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Humans ,Medicine ,skin and connective tissue diseases ,Aged ,Proportional Hazards Models ,Therapy related ,business.industry ,Hematopoietic Stem Cell Transplantation ,Neoplasms, Second Primary ,Hematology ,Middle Aged ,medicine.disease ,Myelodysplastic-Myeloproliferative Diseases ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Female ,business ,030215 immunology - Abstract
Comprehensive analysis of factors impacting risks and outcomes of therapy-related myeloid neoplasms following breast cancer treatment
- Published
- 2015
11. Deregulation of innate immune and inflammatory signaling in myelodysplastic syndromes
- Author
-
Zachary S. Bohannan, Irene Ganan-Gomez, M Cabrero-Calvo, Simona Colla, H. Yang, G. Garcia-Manero, Amit Verma, Daniel T. Starczynowski, Yue Wei, and Ulrich Steidl
- Subjects
Inflammation ,Cancer Research ,Innate immune system ,Myelodysplastic syndromes ,Hematology ,Biology ,medicine.disease ,Article ,Immunity, Innate ,Immune system ,medicine.anatomical_structure ,Oncology ,Immunity ,Myelodysplastic Syndromes ,Immunology ,medicine ,Animals ,Humans ,Bone marrow ,Stem cell ,Progenitor cell ,medicine.symptom ,Signal Transduction - Abstract
Myelodysplastic syndromes (MDSs) are a group of heterogeneous clonal hematologic malignancies that are characterized by defective bone marrow (BM) hematopoiesis and by the occurrence of intramedullary apoptosis. During the past decade, the identification of key genetic and epigenetic alterations in patients has improved our understanding of the pathophysiology of this disease. However, the specific molecular mechanisms leading to the pathogenesis of MDS have largely remained obscure. Recently, essential evidence supporting the direct role of innate immune abnormalities in MDS has been obtained, including the identification of multiple key regulators that are overexpressed or constitutively activated in BM hematopoietic stem and progenitor cells. Mounting experimental results indicate that the dysregulation of these molecules leads to abnormal hematopoiesis, unbalanced cell death and proliferation in patients' BM, and has an important role in the pathogenesis of MDS. Furthermore, there is compelling evidence that the deregulation of innate immune and inflammatory signaling also affects other cells from the immune system and the BM microenvironment, which establish aberrant associations with hematopoietic precursors and contribute to the MDS phenotype. Therefore, the deregulation of innate immune and inflammatory signaling should be considered as one of the driving forces in the pathogenesis of MDS. In this article, we review and update the advances in this field, summarizing the results from the most recent studies and discussing their clinical implications.
- Published
- 2015
12. A clinical trial for patients with acute myeloid leukemia or myelodysplastic syndromes not eligible for standard clinical trials
- Author
-
Farhad Ravandi, X. Huang, Ana Alfonso, Sherry Pierce, Zachary S. Bohannan, Srdan Verstovsek, G. Borthakur, Guillermo Montalban-Bravo, Mark Brandt, William G. Wierda, Courtney D. DiNardo, Nitin Jain, E. Jabbour, Hsiang-Chun Chen, Marina Konopleva, Eli Estey, H. Yang, Kiran Naqvi, Naval Daver, Hagop M. Kantarjian, Yesid Alvarado, Jorge E. Cortes, Zeev Estrov, C. E. Bueso-Ramos, G. Garcia-Manero, Naveen Pemmaraju, Troy Sneed, and Tapan M. Kadia
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Azacitidine ,Comorbidity ,Kaplan-Meier Estimate ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,medicine ,Humans ,Adverse effect ,Survival rate ,Aged ,Aged, 80 and over ,Chromosome Aberrations ,business.industry ,Myelodysplastic syndromes ,Organ dysfunction ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,Clinical trial ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Physical therapy ,Female ,medicine.symptom ,business ,Biomarkers ,Follow-Up Studies ,medicine.drug ,030215 immunology - Abstract
Most clinical trials exclude patients with poor performance or comorbidities. To study whether patients with these characteristics can be treated within a clinical trial, we conducted a study for patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) with poor performance, organ dysfunction or comorbidities. Primary endpoint was 60-day survival. Study included stopping rules for survival and response. Treatment consisted on a combination of azacitidine and vorinostat. Thirty patients (16 with MDS, 14 with AML) were enrolled. Median follow-up was 7.4 months (0.3-29). Sixty-day survival was 83%. No stopping rules were met. Main adverse events (AEs) were grades 1 and 2 gastrointestinal toxicities. In view of these results, we expanded the study and treated 79 additional patients: 27 with azacitidine (AZA) and 52 with azacitidine and vorinostat (AZA+V). Median follow-up was 22.7 months (12.6-47.5). Sixty-day survival rate was 79% (AZA=67%, AZA+V=85%, P=0.07). Median overall survival was 7.6 months (4.5-10.7). Median event-free survival was 4.5 months (3.5-5.6). Main AEs included grades 1 and 2 gastrointestinal toxicities. Our results suggest this subset of patients can be safely treated within clinical trials and derive clinical benefit. Relaxation of standard exclusion criteria may increase the pool of patients likely to benefit from therapy.
- Published
- 2017
13. Myelodysplastic/Myeloproliferative Neoplasms, Unclassifiable (MDS/MPN, U): Natural history and clinical outcome by treatment strategy
- Author
-
Courtney D. DiNardo, Naval Daver, G. Garcia-Manero, Cheng Cameron Yin, Naveen Pemmaraju, Srdan Verstovsek, C. E. Bueso-Ramos, Neeraj Jain, Hagop M. Kantarjian, Jorge E. Cortes, Sherry Pierce, and E. Jabbour
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Unclassifiable MDS ,MEDLINE ,myelofibrosis ,thrombocytosis ,Article ,Myelodysplastic–myeloproliferative diseases ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Aged ,Janus kinase 2 ,biology ,business.industry ,MDS/MPD ,food and beverages ,Hematology ,Janus Kinase 2 ,Middle Aged ,medicine.disease ,Prognosis ,Myelodysplastic-Myeloproliferative Diseases ,Natural history ,JAK2 ,Immunology ,biology.protein ,Treatment strategy ,Female ,business - Abstract
Myelodysplastic/myeloproliferative neoplasms, unclassifiable (MDS/MPN, U): natural history and clinical outcome by treatment strategy
- Published
- 2014
14. Application of the International Prognostic Scoring System-Revised in therapy-related myelodysplastic syndromes and oligoblastic acute myeloid leukemia
- Author
-
Krina K. Patel, Ken H. Young, Patricia S. Fox, G. Garcia-Manero, L. J. Medeiros, Robert P. Hasserjian, Sa A. Wang, Chi Young Ok, Zhuang Zuo, and Francesco C. Stingo
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,education ,MEDLINE ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Intensive care medicine ,health care economics and organizations ,Therapy related ,Hematology ,business.industry ,Myelodysplastic syndromes ,Myeloid leukemia ,Middle Aged ,Prognosis ,medicine.disease ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,Female ,business - Abstract
Application of the International Prognostic Scoring System-Revised in therapy-related myelodysplastic syndromes and oligoblastic acute myeloid leukemia
- Published
- 2013
15. Randomized, dose-escalation study of the p38α MAPK inhibitor SCIO-469 in patients with myelodysplastic syndrome
- Author
-
Lubomir Sokol, Ann M. Lowe, Vikas Bhushan, Y. Li, Jamile M. Shammo, Mikkael A. Sekeres, Alan F. List, G. Garcia-Manero, Peter L. Greenberg, Simrit Parmar, Raymond J. Hohl, Joy Zhu, S. L. Goldberg, Amit Verma, Larry D. Cripe, and Hagop M. Kantarjian
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,P38α mapk ,Internal medicine ,Immunology ,medicine ,Dose escalation ,In patient ,Hematology ,Biology - Abstract
Randomized, dose-escalation study of the p38α MAPK inhibitor SCIO-469 in patients with myelodysplastic syndrome
- Published
- 2012
16. Final report of phase II study of sorafenib, cytarabine and idarubicin for initial therapy in younger patients with acute myeloid leukemia
- Author
-
Zeev Estrov, G. Garcia-Manero, Sherry Pierce, M. Andreeff, Keith W. Pratz, E. Jabbour, Hagop M. Kantarjian, Jorge E. Cortes, R. Luthra, Deborah A. Thomas, Marina Konopleva, Mark J. Levis, Mark Brandt, Farhad Ravandi, Stephan Faderl, Susan O'Brien, G. Borthakur, and C. Arana Yi
- Subjects
Sorafenib ,Cancer Research ,medicine.medical_specialty ,business.industry ,Induction chemotherapy ,Phases of clinical research ,Salvage therapy ,Hematology ,Pharmacology ,Gastroenterology ,Article ,Transplantation ,Regimen ,Oncology ,Internal medicine ,Cytarabine ,medicine ,Idarubicin ,business ,medicine.drug - Abstract
Sorafenib is a multikinase inhibitor and a potent FLT3 inhibitor that inhibits tumor growth and angiogenesis by inhibiting intracellular Raf kinases and cell surface kinase receptors such as VEGFR, PDGFR-beta, cKIT, and FLT-3.1 It has been approved by the U.S. Food and Drug Administration for the treatment of advanced renal cell carcinoma and hepatocellular carcinoma at a standard dose of 400 mg twice daily. The antileukemic activity of sorafenib has been evaluated in preclinical models and a number of recently reported clinical trials.2 In a phase I/II study, combination therapy with sorafenib, cytarabine, and idarubicin in younger patients with AML was able to induce a high complete response (CR) rate in those with FLT3 mutations and a 1-year probability of survival of 74%.3 We here report long-term outcome for adults with newly diagnosed AML who were treated with a combination of sorafenib, cytarabine, and idarubicin. Details of the patient population and the treatment regimen have been previously published.3 Survival curves were plotted by the Kaplan-Meier method and compared using the log-rank test. Differences in subgroups by different covariates were evaluated using the x2 test for nominal values and Fisher's exact test for continuous variables. Between February 1, 2008, and March 31, 2010, 62 patients with previously untreated AML were enrolled in this study. Patient characteristics are summarized in Table 1a. The median age of the patients was 53 years (range, 18 to 66 years); 12 were older than or equal to 60 years. Twenty-three had FLT3 mutations, including 17 with FLT3-ITD (10 with low mutation burden), 4 with D835 mutation, and 2 with both mutations; 11 patients had antecedent hematologic disorder and 10 had unfavorable cytogenetics. Herein, we will refer to the patients as FLT3-ITD-positive or negative including the 4 patients with sole D835 mutation in the latter group and the 2 with both mutations in the former group. Table 1a Patient characteristics The median white blood cell count (WBC) at presentation was 7.25 × 109/L (range, 0.6 to 228.5 × 109/L). Eight patients were FLT3-ITD–positive/nucleophosmin-1 (NPM1)–negative. The median age of patients with FLT3-ITD mutation was 52 years (range, 20 to 66 years); one had adverse and 18 had intermediate cytogenetics (17 with normal karyotype) based on MRC criteria. Sixty-one patients had response assessment; one patient died before response assessment could be performed. Among these, 49 (79%) achieved CR, and 5 (8%) CRp. Seven (11%) patients were non-responders. Among the 54 responders, 51 patients received consolidation and maintenance courses; 3 proceeded to allogeneic stem cell transplant straight after induction. Patients with FLT3-ITD were more likely to achieve a CR/CRp than were patients without FLT3-ITD [18 of 19 patients (95%) vs. 36 of 43 (84%), respectively (P = 0.23)] (Table 1b). Seventeen FLT3-ITD patients achieved CR and one had CRp, three FLT3-D835 patients achieved a CR and one had CRp, and 32 of 43 patients without FLT3-ITD achieved a CR and four had CRp; one died at induction, and six were resistant to therapy. CR was achieved after one induction cycle in 44 patients and after two induction cycles in five patients. Sixteen patients with the FLT3-ITD mutation achieved CR/CRp after one induction cycle, and two achieved it after two cycles. Altogether, sixteen patients proceeded to allogeneic stem-cell transplantation in first CR/CRp, including nine with a FLT3 mutation (six with FLT3-ITD, three with FLT3-D835). To date, 35 (65%) of the responders have experienced relapse including 11 of 18 patients (61%) with FLT3-ITD and 24 of 36 (67%) without FLT3-ITD (P = 0.69); 33 patients received salvage therapy: 2 died while receiving first salvage therapy, 11 achieved a second CR, 2 CRp, 1 PR and 17 had refractory disease. Ultimately 22 of these 33 patients died after their first or subsequent salvage therapies. Table 1b Response to induction according to FLT3 status With a median follow-up of 52 months (range, 2.3-62.9 months) for all patients, the median OS and DFS were 29 and 13.8 months, respectively (Figure 1a and and1b).1b). Figure 1c and and1d1d demonstrates the OS and DFS for all patients and for patients with FLT3-ITD. Among patients with FLT3-ITD who achieved CR/CRp, 11 have relapsed and 5 remain in CR; 2 patients died in CR. The median OS and DFS among the 19 patients with FLT3-ITD was 15.5 and 9.9 months, respectively (Figure 1c and and1d1d). Figure 1 (a) OS for all patients; (b) DFS for all patients; (c) OS according to FLT3-ITD status; (d) DFS according to FLT3-ITD status. Hematopoietic stem cell transplantation was performed in first CR in 16 of 54 patients who achieved a CR/CRp (30%); six of the 16 relapsed and 7 died following the SCT. The 3-year disease-free survival (DFS) rate (in 54 patients achieving CR/CRp) was 34%; the 3-year OS rate for all patients was 48% (Figure 1a and and1b1b). The regimen was generally well tolerated, the adverse events being those expected in patients receiving induction chemotherapy with the combination of cytarabine and idarubicin. The most common grade 3 and 4 adverse events possibly associated with the addition of sorafenib to the induction regimen occurred in 15 patients and included nausea and vomiting (7), cardiac/hypertension (6), diarrhea (5), infections (5), hand and foot syndrome (4), liver toxicity (5), and pancreatitis (2). One patient died of massive myocardial infarction during induction; this was not considered to be related to sorafenib (Table 1c). Table 1c Toxicity Mutations in FLT3 are well-established indicators of poor prognosis in AML 4,5 and occur in approximately 20% to 30% of patients. Small-molecule FLT3 kinase inhibitors have been developed with different efficacies against FLT3-mutant cell lines. They also have been evaluated in FLT3-ITD+ primary leukemia cells, alone or in combination with cytarabine and anthracyclines such as idarubicin. 6,7,8,9 This study reports on the long-term follow-up of patients with newly diagnosed AML who received upfront treatment consisting of sorafenib combined with cytarabine and idarubicin. Nineteen patients had the FLT3-ITD mutation. The response rates (CR and CRp) for patients with FLT3-ITD were higher than that for patients without FLT3-ITD including those with FLT3-D835 mutants and those with wild-type FLT3 (18/19 patients [95%] vs. 36/43 [83%], respectively) although this was not statistically significant (P = 0.23). Similarly, there was no significant difference in OS or DFS when comparing patients with and without FLT3-ITD (Figure 1c and and1d).1d). This is in contrast to previous studies reporting significantly shorter DFS and OS for patients with FLT3-ITD and can potentially be attributable to the addition of sorafenib to the regimen. This may be due to the possible increased likelihood of the patients to be able to undergo an allogeneic stem cell transplant as a result of the addition of sorafenib. We also analyzed the outcomes censoring patients at the time of allogeneic stem cell transplant (n=16) and found no significant difference between the groups' outcomes (data not shown). In this study, the sorafenib containing induction regimen produced a high response rate but was associated with relapse in the majority of the patients. Man et al. 10 reported activation of alternative survival pathways in sorafenib-resistant leukemic clones. In their study, 12 of 13 patients with AML and FLT3-ITD responded to treatment with sorafenib monotherapy but nine lost their response at a median of 72 days. We had a similar high response rate in this study but the median DFS was significantly longer at 9.9 months suggesting a potential benefit for combining sorafenib with chemotherapy, especially that overall, the treatment was well tolerated with limited grade 3 and 4 toxicity. A recent report, however, suggested that the addition of sorafenib to standard cytarabine and daunorubicin induction regimen did not improve EFS or OS and was associated with increased toxicity during induction.11 However, the patients treated in that study were older than 60 which may account for the higher incidence of toxicity. Our study is limited due to the inability to discern the impact of other interventions such as allogeneic stem cell transplant, related to its non-randomized design and limited patient numbers. Rollig and colleagues have reported preliminary results of SORAML trial in which patients received cytarabine 100 mg/m2 for 7 days plus daunorubicin 60 mg/m2 for 3 days followed by high dose cytarabine consolidation.12 Patients were randomized to receive Sorafenib at a dose of 800 mg/day or placebo during induction and consolidation and for a 12 months maintenance period after the end of consolidation. Among the 276 patients randomized, 264 were evaluable for event-free survival (EFS), 132 in each arm.12 Patient and disease characteristics were similar between the two arms, with the FLT3-ITD incidence of 16%. The CR rates were 56% and 60% in the placebo and sorafenib arms, respectively (p=0.62). With a median follow-up of 18 months, the median EFS was 12.2 months in the placebo arm and was not reached in the sorafenib arm; the 2-year OS was 66% versus 72% in placebo and sorafenib arms, respectively (p=0.37). Although there was an increased risk of hepatotoxicity and bleeding events in the sorafenib arm, the authors concluded that the addition of sorafenib was associated with a prolongation of EFS which was most marked in the patients with FLT3-ITD.12 The eventual result of this study will better clarify the potential role of sorafenib in the treatment of patients with AML and FLT3-ITD.
- Published
- 2014
17. Impact of adjunct cytogenetic abnormalities for prognostic stratification in patients with myelodysplastic syndrome and deletion 5q
- Author
-
Carlo Aul, Lourdes Florensa, Otto Krieger, Christian Steidl, Guillermo Sanz, Claudia Haferlach, G. Garcia-Manero, Michael Lübbert, Reinhard Stauder, Rosa Collado, Peter Valent, Miguel A. Sanz, José Cervera, Thomas Noesslinger, Maria-Jose Calasanz, Julie Schanz, Kazuma Ohyashiki, Leonor Arenillas, J M Hernández, Barbara Hildebrandt, Carmen Pedro, María-Luisa Martín, Esperanza Such, Teresa Vallespi, Javier Grau, Ana Valencia, E. Luño, A.A.N. Giagounidis, Ulrich Germing, Sabine Blum, Michael Pfeilstöcker, Mar Mallo, D. Haase, Blanca Espinet, C. Fonatsch, and Francesc Solé
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Fusion gene ,Internal medicine ,medicine ,Humans ,Anemia, Macrocytic ,Aged ,Retrospective Studies ,Aged, 80 and over ,Chromosome Aberrations ,Hematology ,business.industry ,Anemia, Macrocytic/genetics ,Anemia, Macrocytic/mortality ,Chromosome Deletion ,Chromosomes, Human, Pair 5/genetics ,Female ,Karyotyping ,Middle Aged ,Myelodysplastic Syndromes/genetics ,Myelodysplastic Syndromes/mortality ,Prognosis ,Myelodysplastic syndromes ,Cytogenetics ,Myeloid leukemia ,De novo Myelodysplastic Syndrome ,medicine.disease ,Lymphoma ,Leukemia ,Myelodysplastic Syndromes ,Chromosomes, Human, Pair 5 ,business - Abstract
This cooperative study assessed prognostic factors for overall survival (OS) and risk of transformation to acute myeloid leukemia (AML) in 541 patients with de novo myelodysplastic syndrome (MDS) and deletion 5q. Additional chromosomal abnormalities were strongly related to different patients' characteristics. In multivariate analysis, the most important predictors of both OS and AML transformation risk were number of chromosomal abnormalities (P
- Published
- 2010
18. Trisomy 11 in myelodysplastic syndromes defines a unique group of disease with aggressive clinicopathologic features
- Author
-
Dan Jones, Hagop M. Kantarjian, Guohui Lu, Sa A. Wang, K. Jabbar, G. Garcia-Manero, S. S. Chen, N. Galili, L. J. Medeiros, Azra Raza, Timothy J. McDonnell, and Francisco Vega
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Aneuploidy ,MLL Partial Tandem Duplication ,Chromosome Disorders ,Trisomy ,Biology ,Proto-Oncogene Proteins p21(ras) ,Gene Duplication ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Survival rate ,In Situ Hybridization, Fluorescence ,Aged ,Retrospective Studies ,Aged, 80 and over ,Cytopenia ,Chromosomes, Human, Pair 11 ,Myelodysplastic syndromes ,DNA, Neoplasm ,Histone-Lysine N-Methyltransferase ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,Blotting, Southern ,Leukemia, Myeloid, Acute ,Genes, ras ,fms-Like Tyrosine Kinase 3 ,Dysplasia ,International Prognostic Scoring System ,Karyotyping ,Myelodysplastic Syndromes ,Mutation ,ras Proteins ,Female ,Myeloid-Lymphoid Leukemia Protein - Abstract
Trisomy 11 in myelodysplastic syndromes (MDS) is rare, with undefined clinical significance and is currently assigned to the International Prognostic Scoring System (IPSS) intermediate-risk group. Over a 15-year period, we identified 17 MDS patients with trisomy 11 either as a sole abnormality (n=10) or associated with one or two additional alterations (n=7), comprising 0.3% of all MDS cases reviewed. Of 16 patients with Bone Marrow material available for review, 14 (88%) patients presented with excess blasts, 69% patients evolved to acute myeloid leukemia (AML) in a 5-month median interval and the median survival was 14 months. For comparison, we studied 19 AML patients with trisomy 11 in a noncomplex karyotype, of which, a substantial subset of patients had morphologic dysplasia, and/or preexisting cytopenia(s)/MDS. Genomic DNA PCR showed MLL partial tandem duplication in 5 of 10 MDS and 7 of 11 AML patients. A review of literature identified 17 additional cases of MDS with trisomy 11, showing similar clinicopathologic features to our patients. Compared with our historical data comprising 1165 MDS patients, MDS patients with trisomy 11 had a significantly inferior survival to patients in the IPSS intermediate-risk cytogenetic group (P=0.0002), but comparable to the poor-risk group (P=0.97). We conclude that trisomy 11 in MDS correlates with clinical aggressiveness, may suggest an early/evolving AML with myelodysplasia-related changes and is best considered a high-risk cytogenetic abnormality in MDS prognostication.
- Published
- 2010
19. Adult acute erythroleukemia: an analysis of 91 patients treated at a single institution
- Author
-
Michael J. Keating, Farhad Ravandi, J. E. Cortes, Charles Koller, Sherry Pierce, Fabio P.S. Santos, G. Borthakur, Stephan Faderl, Hagop M. Kantarjian, Emil J. Freireich, C. E. Bueso-Ramos, G. Garcia-Manero, M. de Lima, Miloslav Beran, Susan O'Brien, and X. Huang
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Myelodysplastic syndromes ,Case-control study ,Acute erythroid leukemia ,Cancer ,Retrospective cohort study ,Hematology ,medicine.disease ,Gastroenterology ,Surgery ,Leukemia ,Oncology ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Risk factor ,business ,neoplasms ,Survival analysis - Abstract
Acute erythroleukemia (AML-M6) is an uncommon subtype of acute myeloid leukemia (AML); it is considered to have a poor prognosis. From 1 January 1980 to 21 May 2008, 91 patients with newly diagnosed AML-M6 were seen at the University of Texas–M.D. Anderson Cancer Center (UT–MDACC). Forty-five patients (50%) had a history of myelodysplatic syndrome (MDS), compared with 41% in our control group (patients with other AML subtypes) (P=0.08). Poor-risk cytogenetics were more common in patients with AML-M6 (61% versus 38%, P=0.001). Complete remission rates were 62% for patients with AML-M6, comparing with 58% for the control group (P=0.35). Median disease free survival (DFS) for patients with AML-M6 was 32 weeks, versus 49 weeks for the control group (P=0.05). Median overall survival (OS) of patients with AML-M6 was 36 weeks, compared with 43 weeks for the control group (P=0.60). On multivariate analysis for DFS and OS, AML-M6 was not an independent risk factor. AML-M6 is commonly associated with a previous diagnosis of MDS and poor-risk karyotype. The diagnosis of AML-M6 does not impart by itself a worse prognosis, and treatment decisions on this disease should be guided by well known AML prognostic factors.
- Published
- 2009
20. Aberrant DNA methylation of the Src kinase Hck, but not of Lyn, in Philadelphia chromosome negative acute lymphocytic leukemia
- Author
-
H. Yang, Blanca Sanchez-Gonzalez, Alfonso Quintás-Cardama, Koyu Hoshino, and G. Garcia-Manero
- Subjects
Cancer Research ,Decitabine ,Biology ,Philadelphia chromosome ,LYN ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,medicine ,Humans ,Philadelphia Chromosome ,Promoter Regions, Genetic ,Hematology ,Methylation ,DNA Methylation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Leukemia ,src-Family Kinases ,Oncology ,DNA methylation ,Azacitidine ,Proto-Oncogene Proteins c-hck ,Cancer research ,CpG Islands ,Chronic myelogenous leukemia ,medicine.drug - Abstract
Hck and Lyn are required in Philadelphia chromosome (Ph) positive acute lymphocytic leukemia (ALL). Here, we present evidence that the promoter CpG island of Hck, but not of Lyn, is aberrantly methylated in leukemia. Hck promoter DNA methylation was detected in 13 out of 23 (56.5%) hematopoietic and eight out of 10 (80%) non-hematopoietic cell lines, but not in normal controls. Treatment with 5-aza-2'-deoxycytidine induced demethylation and restoration of Hck mRNA and protein expression. Hck methylation (> or =15%) was detected in nine out of 44 (20%) patients with Ph negative ALL, and in one out 16 (6%) patients with Ph positive ALL, but not in patients with AML or chronic myelogenous leukemia. In this subset of patients, low levels of Hck methylation (10-15%) were observed in 26-30% of patients. Lyn methylation was observed in three out of 28 (10.7%) cell lines, but only in one out of 71 (1.4%) patients. Patients with Ph negative ALL and Hck methylation had a poorer prognosis. These data indicate that Hck may have tumor suppressor properties in BCR-ABL negative leukemia.
- Published
- 2007
21. Novel drugs for older patients with acute myeloid leukemia
- Author
-
G. Garcia-Manero and Guillermo Montalban-Bravo
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Pracinostat ,Decitabine ,Antineoplastic Agents ,Pharmacology ,Sapacitabine ,Vosaroxin ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Midostaurin ,Protein Kinase Inhibitors ,Quizartinib ,Aged ,business.industry ,Cytotoxins ,Patient Selection ,Rigosertib ,Age Factors ,Antibodies, Monoclonal ,Volasertib ,Hematology ,Drugs, Investigational ,Histone Deacetylase Inhibitors ,Leukemia, Myeloid, Acute ,chemistry ,Clinical Trials, Phase III as Topic ,business ,medicine.drug - Abstract
Acute myeloid leukemia (AML) is the second most common form of leukemia and the most frequent cause of leukemia-related deaths in the United States. The incidence of AML increases with advancing age and the prognosis for patients with AML worsens substantially with increasing age. Many older patients are ineligible for intensive treatment and require other therapeutic approaches to optimize clinical outcome. To address this treatment gap, novel agents with varying mechanisms of action targeting different cellular processes are currently in development. Hypomethylating agents (azacitidine, decitabine, SGI-110), histone deacetylase inhibitors (vorinostat, pracinostat, panobinostat), FMS-like tyrosine kinase receptor-3 inhibitors (quizartinib, sorafenib, midostaurin, crenolanib), cytotoxic agents (clofarabine, sapacitabine, vosaroxin), cell cycle inhibitors (barasertib, volasertib, rigosertib) and monoclonal antibodies (gentuzumab ozogamicin, lintuzumab-Ac225) represent some of these promising new treatments. This review provides an overview of novel agents that have either completed or are currently in ongoing phase III trials in patients with previously untreated AML for whom intensive treatment is not an option. Other potential drugs in earlier stages of development will also be addressed in this review.
- Published
- 2014
22. Prognostic significance of 11q23 aberrations in adult acute myeloid leukemia and the role of allogeneic stem cell transplantation
- Author
-
Naval Daver, Hagop M. Kantarjian, Jorge E. Cortes, G. Garcia-Manero, E. Jabbour, Wei Qiao, Sherry Pierce, Tapan M. Kadia, Yiming Chen, M. de Lima, L. V. Abruzzo, Susan O'Brien, Farhad Ravandi, Partow Kebriaei, Stephan Faderl, and Zeev Estrov
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Chromosomal translocation ,Biology ,Translocation, Genetic ,Article ,Young Adult ,Risk Factors ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Aged ,Aged, 80 and over ,Chromosomes, Human, Pair 11 ,Myeloid leukemia ,Adult Acute Myeloid Leukemia ,Karyotype ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Transplantation ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Karyotyping ,Immunology ,Female ,Bone marrow ,Stem Cell Transplantation - Abstract
The clinical features and outcomes of 148 patients with acute myeloid leukemia (AML) and 11q23 chromosomal abnormalities were compared with those of 2640 patients with non-11q23 AML. Patients with t(9;11)), t(6;11), or other 11q23 balanced translocations [t(11;v)(q23;v)] presented at a younger age and with higher percentage of bone marrow blasts. Unbalanced 11q23 abnormalities were commonly associated with deletions of chromosomes 5q, 7q and/or complex karyotypes. In multivariate analysis, when compared to patients with non-11q23 AML and unfavorable risk karyotype, there was a significant difference in overall survival (OS) for patients with t(9;11) (P = .004), whereas there were no difference in OS for patients with t(6;11) (P = .62), t(11;19) (P = 0.20), and unbalanced 11q23 aberrations (P = .85) or t(11;v)(q23;v) (P = 0.59), indicating that t(9;11) has an independent intermediate prognostic factor, with all others poor prognostic factors for OS; this was further confirmed by comparing them with patients with non-11q23 AML and intermediate risk karyotype. Using intention-to treat analysis based on donor availability, we also noted that allogeneic stem cell transplant (SCT) in first remission had a significant benefit towards improving OS (P < 0.001) and relapse-free survival (P
- Published
- 2012
23. Activity of tyrosine kinase inhibitors against human NUP214-ABL1-positive T cell malignancies
- Author
-
Francisco Vega, Patrick A. Lennon, Hagop M. Kantarjian, D. G. Gilliland, G. Garcia-Manero, Weigang Tong, Jorge E. Cortes, Alfonso Quintás-Cardama, Taghi Manshouri, Jan Cools, and Francis Y. Lee
- Subjects
Adult ,Male ,Cancer Research ,Oncogene Proteins, Fusion ,medicine.drug_class ,Cell Survival ,Blotting, Western ,Dasatinib ,Apoptosis ,Mice, SCID ,Biology ,Tyrosine-kinase inhibitor ,Piperazines ,Mice ,Mice, Inbred NOD ,hemic and lymphatic diseases ,medicine ,STAT5 Transcription Factor ,Tumor Cells, Cultured ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Phosphorylation ,Protein Kinase Inhibitors ,In Situ Hybridization, Fluorescence ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,ABL ,Leukemia, Experimental ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Nuclear Proteins ,Imatinib ,Hematology ,Protein-Tyrosine Kinases ,medicine.disease ,Thiazoles ,Imatinib mesylate ,Pyrimidines ,Oncology ,Nilotinib ,Benzamides ,Cancer research ,Imatinib Mesylate ,Female ,Tyrosine kinase ,medicine.drug ,Chronic myelogenous leukemia - Abstract
Amplification of the NUP214-ABL1 oncogene can be detected in patients with T cell acute lymphoblastic leukemia (T-ALL). We screened 29 patients with T cell malignancies for the expression of NUP214-ABL1 by reverse transcription-polymerase chain reaction (RT-PCR). NUP214-ABL1 was detected in three (10%) patients. These results were confirmed by fluorescence in situ hybridization techniques. We also studied the activity of imatinib, nilotinib and dasatinib against the human NUP214-ABL1-positive cell lines PEER and BE-13. All three tyrosine kinase inhibitors decreased the viability of PEER and BE-13 cells, but nilotinib and dasatinib had >1-log lower IC(50) values than imatinib (P
- Published
- 2008
24. MicroRNAs and noncoding RNAs in hematological malignancies: molecular, clinical and therapeutic implications
- Author
-
Hagop M. Kantarjian, George A. Calin, M. Andreeff, Ramiro Garzon, G. Garcia-Manero, and Muller Fabbri
- Subjects
Cancer Research ,RNA, Untranslated ,Oncogene ,Tumor suppressor gene ,RNA ,Hematology ,Biology ,Bioinformatics ,medicine.disease_cause ,Hematopoiesis ,MicroRNAs ,Oncology ,RNA interference ,Hematologic Neoplasms ,microRNA ,medicine ,Gene silencing ,Humans ,Carcinogenesis ,Gene - Abstract
MicroRNAs (miRNAs) are a family of 19-24 nucleotide noncoding RNAs (ncRNAs) with posttranscriptional regulatory functions. Increasing evidences from the literature show that miRNAs play a pivotal role in human tumorigenesis. Many studies have addressed the role of miRNAs in normal hematopoiesis, giving an interpretative key to the aberrancies of expression observed in human hematological malignancies. Moreover, the recent demonstration that other ncRNAs, the ultraconserved genes (UCGs) or transcribed ultraconserved regions (T-UCRs), are involved in human cancerogenesis, suggests that the wider family of ncRNAs (including both miRNAs and UCGs) could contribute to the development of the malignant phenotype. Here we review the main studies investigating the role of miRNAs and UCRs in both normal hemopoiesis and hematological malignancies, and identify the molecular, clinical and therapeutic implications of these recent findings.
- Published
- 2008
25. A prognostic score for patients with lower risk myelodysplastic syndrome
- Author
-
Hagop M. Kantarjian, Jorge E. Cortes, William G. Wierda, G. Garcia-Manero, Eli Estey, Xuelin Huang, J. Shan, Farhad Ravandi, Stephan Faderl, Jinyun Liu, Sherry Pierce, and G. Borthakur
- Subjects
Adult ,Male ,Risk ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Anemia ,Lower risk ,Severity of Illness Index ,Bone Marrow ,Internal medicine ,Epidemiology ,Severity of illness ,medicine ,Humans ,Survival analysis ,Aged ,Aged, 80 and over ,Chromosome Aberrations ,business.industry ,Platelet Count ,Myelodysplastic syndromes ,Age Factors ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Survival Analysis ,Surgery ,Leukemia ,Oncology ,International Prognostic Scoring System ,Leukemia, Myeloid ,Myelodysplastic Syndromes ,Acute Disease ,Ferritins ,Disease Progression ,Female ,business ,beta 2-Microglobulin ,Follow-Up Studies - Abstract
Current prognostic models for myelodysplastic syndromes (MDS) do not allow the identification of patients with lower risk disease and poor prognosis that may benefit from early therapeutic intervention. We evaluated the characteristics of 856 patients with low or intermediate-1 disease by the International Prognostic Scoring System. Mean follow-up was 19.6 months (range 1-262). Of these patients, 87 (10%) transformed to acute myelogenous leukemia, and 429 (50%) had died. By multivariate analysis, characteristics associated with worse survival (P
- Published
- 2007
26. Methylation, aging, and pediatric acute lymphocytic leukemia
- Author
-
G. Garcia-Manero
- Subjects
Cancer Research ,GC Rich Sequence ,Aging ,Infant ,Hematology ,Methylation ,Biology ,DNA Methylation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Polymerase Chain Reaction ,Pediatric Acute Lymphocytic Leukemia ,Oncology ,Acute lymphocytic leukemia ,DNA methylation ,Immunology ,medicine ,Humans ,Gene ,Dinucleoside Phosphates - Published
- 2003
27. In reply to ‘Improving the prognostic evaluation of patients with lower-risk myelodysplastic syndromes’ by Kuendgen et al
- Author
-
G. Garcia-Manero
- Subjects
Cancer Research ,Pediatrics ,medicine.medical_specialty ,Oncology ,business.industry ,Myelodysplastic syndromes ,medicine ,Hematology ,Lower risk ,business ,medicine.disease - Abstract
In reply to ‘Improving the prognostic evaluation of patients with lower-risk myelodysplastic syndromes’ by Kuendgen et al.
- Published
- 2008
28. A phase II trial of ipilimumab, nivolumab, or ipilimumab and nivolumab with or without azacitidine in relapsed or refractory myelodysplastic neoplasms.
- Author
-
Bouligny IM, Montalban-Bravo G, Sasaki K, Daver N, Jabbour E, Alvarado Y, DiNardo CD, Ravandi F, Borthakur G, Pemmaraju N, Kadia T, Masarova L, Takahashi K, Andreeff M, Bazinet A, Yang H, Kanagal R, Pierce S, Meyer M, Huang X, and Garcia-Manero G
- Abstract
Competing Interests: Competing interests The authors declare no competing interests. Ethics approval and consent to participate The MD Anderson Institutional Review Board approved this study. All patients provided written informed consent according to the Declaration of Helsinki.
- Published
- 2024
- Full Text
- View/download PDF
29. Venetoclax in combination with hypomethylating agents in chronic myelomonocytic leukemia: a propensity score matched multicenter cohort study.
- Author
-
Tremblay D, Csizmar C, DiNardo CD, Ball S, Rippel N, Hammond D, Kadia TM, Ravandi F, Chien K, Van Hyfte G, Mazumdar M, Saliba A, Mangaonkar A, Lasho T, Al-Kali A, Kremyanskaya M, Feld J, Silverman LR, Komrokji R, Mascarenhas J, Padron E, Garcia-Manero G, Sallman DA, Patnaik MM, and Montalban-Bravo G
- Abstract
Competing Interests: Competing interests DT received research funding from Sobi, Sumitomo, Cogent Biosciences and Gilead and honoraria from Sobi, Novartis, AbbVie, PharmaEssentia, Sierra Oncology, GSK and Cogent Biosciences. CDD received honoraria from Abbvie, AstraZeneca, BMS, Genentech, GenMab, GSK, Immunogen, Notable Labs, Rigel, Schrodinger, and Servier and grant support from LLS Scholar in Research Award. TMK received research funding from AbbVie and Genentech and honoraria from AbbVie. FR received research funding from AbbVie and BMS and honoraria from AbbVie and BMS. AM received research funding from BMS, Incyte and Novartis. AA received research funding from Novartis, BMS, Onconova, Medimmune, Ariad, GSK, Celgene, Eisai, ALX Oncology, H3B Biomedicine/Hemavant. MK has received honoraria from Protagonist, Silence Therapeutics, Morphosys, Incyte, AbbVie and Kura. JF received research funding from Oryzon Genomics, Taiho Oncology, and Syros. RK received research funding from BMS and honoraria from Abbvie, BMS, DSI, Geron, Janssen, Jazz, PharmaEssentia, Rigel, Servio, Sobi, and Sumitomo. JM received research funding from Incyte, BMS, Novartis, Abbvie, Geron, Kartos, Karyopharm, Sobi and PharmaEssentia and honoraria from Incyte, BMS, Abbvie, Kartos, Geron, GSK, Roche, Merck, Pfizer, PharmaEssentia, MorphoSys, Novartis, Galecto, Sobi, Sumitomo, and Karyopharm. EP received research funding from Incyte, BMS, and Blueprint and honoraria from BMS, GSK, Sobi, Blueprint, Taiho, PharmaEssentia. DAS received research funding from Aprea and Jazz and honoraria from AbbVie, Aprea, Agios, Celyad, Froghorn, Gilead,Incyte, Intellisphere LLC, Kite, Megenta, Novartis, AvenCell, Astellas, BlueBird Bio, BMS, Dark Blue Therapeutics, Intellia, Jasper Therapeutics, Kite, Magenta Therapeutics, NKARTA, Novartis, Orbital Therapeutics, Rigel Pharmaceuticals, Shattuck Labs, Servier, Syndax, and Syros. MMP received research funding from Stemline, Kura Oncology, Solu Therapeutics, Epigenetix and Polaris Pharmaceuticals. GMB received research funding from IFM Therapeutics, Takeda Oncology, Solu Theraputics. The remaining of the authors have no potential competing interests to disclose. Ethics approval and consent to participate This study received approval by the Institutional Review Boards from all participating institutions and research was conducted in accordance with the Declaration of Helsinki. Given the retrospective nature of this analysis, informed consent was not required by local regulatory authorities.
- Published
- 2024
- Full Text
- View/download PDF
30. Downregulation of UBA1 expression in myelodysplastic neoplasm.
- Author
-
Wei Y, Zheng H, Li Z, Lockyer PP, Darbaniyan F, Kanagal-Shamanna R, Yang H, Hammond D, and Garcia-Manero G
- Subjects
- Humans, Aged, Male, Middle Aged, Female, Ubiquitin-Activating Enzymes genetics, Ubiquitin-Activating Enzymes metabolism, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology, Myelodysplastic Syndromes metabolism, Down-Regulation
- Published
- 2024
- Full Text
- View/download PDF
31. Menin inhibitors in pediatric acute leukemia: a comprehensive review and recommendations to accelerate progress in collaboration with adult leukemia and the international community.
- Author
-
Cuglievan B, Kantarjian H, Rubnitz JE, Cooper TM, Zwaan CM, Pollard JA, DiNardo CD, Kadia TM, Guest E, Short NJ, McCall D, Daver N, Nunez C, Haddad FG, Garcia M, Bhalla KN, Maiti A, Catueno S, Fiskus W, Carter BZ, Gibson A, Roth M, Khazal S, Tewari P, Abbas HA, Bourgeois W, Andreeff M, Shukla NN, Truong DD, Connors J, Ludwig JA, Stutterheim J, Salzer E, Juul-Dam KL, Sasaki K, Mahadeo KM, Tasian SK, Borthakur G, Dickson S, Jain N, Jabbour E, Meshinchi S, Garcia-Manero G, Ravandi F, Stein EM, Kolb EA, and Issa GC
- Subjects
- Humans, Child, Adult, Leukemia drug therapy, Leukemia genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Nucleophosmin
- Abstract
Aberrant expression of HOX and MEIS1 family genes, as seen in KMT2A-rearranged, NUP98-rearranged, or NPM1-mutated leukemias leads to arrested differentiation and leukemia development. HOX family genes are essential gatekeepers of physiologic hematopoiesis, and their expression is regulated by the interaction between KMT2A and menin. Menin inhibitors block this interaction, downregulate the abnormal expression of MEIS1 and other transcription factors and thereby release the differentiation block. Menin inhibitors show significant clinical efficacy against KMT2A-rearranged and NPM1-mutated acute leukemias, with promising potential to address unmet needs in various pediatric leukemia subtypes. In this collaborative initiative, pediatric and adult hematologists/oncologists, and stem cell transplant physicians have united their expertise to explore the potential of menin inhibitors in pediatric leukemia treatment internationally. Our efforts aim to provide a comprehensive clinical overview of menin inhibitors, integrating preclinical evidence and insights from ongoing global clinical trials. Additionally, we propose future international, inclusive, and efficient clinical trial designs, integrating pediatric populations in adult trials, to ensure broad access to this promising therapy for all children and adolescents with menin-dependent leukemias., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
32. Longitudinal follow up of a phase 2 trial of venetoclax added to hyper-CVAD, nelarabine and pegylated asparaginase in patients with T-cell acute lymphoblastic leukemia and lymphoma.
- Author
-
Ravandi F, Senapati J, Jain N, Short NJ, Kadia T, Borthakur G, Konopleva M, Wierda W, Huang X, Maiti A, Issa G, Balkin H, Garris R, Ferrajoli A, Garcia-Manero G, Alvarado Y, Kebriaei P, Jabbour E, and Kantarjian HM
- Abstract
Optimal frontline use of active agents in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL) is prudent to improve outcomes. We report the long-term follow-up of the phase 2 trial of HyperCVAD with nelarabine and pegylated asparaginase (Original cohort). In the latest protocol iteration venetoclax was added to the induction/consolidation regimen (Venetoclax cohort). Eligible patients were adults with untreated T-ALL/LBL or after minimal therapy and with adequate organ function. Primary endpoint of this analysis was improvement in 2-year progression free survival (PFS) and overall survival (OS) with venetoclax. From Aug 2007 to Dec 2024, 145 patients, at a median age of 35.4 years, were treated; 46 (33.8%) were in the venetoclax cohort. At median follow-up (mFU) of 62.4 months, 5-year PFS, duration of response (DOR), and OS were 63.7%, 72.0% and 66.2% respectively. In the venetoclax cohort (mFU 24.4 months) 2-year PFS (87.9% versus 64.1%, p = 0.03) and 2-year DOR (93.6% versus 69.2%, p = 0.005) were superior to the original cohort (mFU 89.4 months) and 2-year OS appeared better (87.8% versus 73.9%, p = 0.16). Febrile neutropenia was the most common serious adverse event, seen in 60% patients. The addition of venetoclax to HyperCVAD-nelarabine-pegylated asparaginase was tolerable and led to improvement in DOR and PFS., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
33. Association between class III obesity and overall survival in previously untreated younger patients with acute myeloid leukemia enrolled on SWOG S1203.
- Author
-
Zhang MY, Othus M, McMillen K, Erba HP, Garcia-Manero G, Pagel JM, Sorror ML, and Percival MM
- Subjects
- Humans, Female, Male, Adult, Middle Aged, Young Adult, Hematopoietic Stem Cell Transplantation, Adolescent, Prognosis, Survival Rate, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute complications, Obesity complications, Obesity mortality, Body Mass Index
- Abstract
There has been ongoing debate on the association between obesity and outcomes in acute myeloid leukemia (AML). Currently few studies have stratified outcomes by class I obesity, class II obesity, and class III obesity, and a more nuanced understanding is becoming increasingly important with the rising prevalence of obesity. We examined the association between body mass index (BMI) and outcomes in previously untreated AML in younger patients (age ≤60) enrolled in SWOG S1203 (n = 729). Class III obesity was associated with an increased rate of early death (p = 0.004) and worse overall survival (OS) in multivariate analysis (hazard ratio (HR) 2.48, 95% confidence interval (CI) 1.62-3.80 versus normal weight). Class III obesity was also associated with worse OS after allogeneic hematopoietic cell transplant (HR 2.37, 95% CI 1.24-4.54 versus normal weight). These findings highlight the unique risk of class III obesity in AML, and the importance of further investigation to better characterize this patient population., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
34. Influence of co-mutational patterns in disease phenotype and clinical outcomes of chronic myelomonocytic leukemia.
- Author
-
Montalban-Bravo G, Rodriguez-Sevilla JJ, Swanson DM, Kanagal-Shamanna R, Hammond D, Chien K, Sasaki K, Jabbour E, DiNardo C, Takahashi K, Short N, Issa GC, Pemmaraju N, Kadia T, Ravandi F, Daver N, Borthakur G, Loghavi S, Pierce S, Bueso-Ramos C, Kantarjian H, and Garcia-Manero G
- Subjects
- Humans, Prognosis, Male, Female, Aged, Middle Aged, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic pathology, Phenotype, Mutation
- Published
- 2024
- Full Text
- View/download PDF
35. Clinical evaluation of complete remission (CR) with partial hematologic recovery (CRh) in acute myeloid leukemia: a report of 7235 patients from seven cohorts.
- Author
-
Appelbaum JS, Wei AH, Mandrekar SJ, Tiong IS, Chua CC, Teh TC, Fong CY, Ting SB, Weber D, Benner A, Hill H, Saadati M, Yin J, Stone RM, Garcia-Manero G, Erba HP, Uy GL, Marcucci G, Larson RA, Thomas A, Freeman SD, Almuina NM, Döhner K, Thomas I, Russel NH, Döhner H, Othus M, Estey EH, and Walter RB
- Subjects
- Humans, Remission Induction, Pathologic Complete Response, Antineoplastic Combined Chemotherapy Protocols, Leukemia, Myeloid, Acute drug therapy
- Published
- 2024
- Full Text
- View/download PDF
36. A randomized phase III study of standard versus high-dose cytarabine with or without vorinostat for AML.
- Author
-
Garcia-Manero G, Podoltsev NA, Othus M, Pagel JM, Radich JP, Fang M, Rizzieri DA, Marcucci G, Strickland SA, Litzow MR, Savoie ML, Medeiros BC, Sekeres MA, Lin TL, Uy GL, Powell BL, Kolitz JE, Larson RA, Stone RM, Claxton D, Essell J, Luger SM, Mohan SR, Moseley A, Appelbaum FR, and Erba HP
- Subjects
- Humans, Vorinostat therapeutic use, Daunorubicin, Idarubicin therapeutic use, Remission Induction, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cytarabine, Leukemia, Myeloid, Acute
- Abstract
Prior experience indicated that use of higher doses of cytarabine during induction for acute myeloid leukemia (AML) with a histone deacetylase inhibitor resulted in high response rates. S1203 was a randomized multicenter trial for previously untreated patients aged 18-60 with AML which compared daunorubicin and cytarabine (DA), idarubicin with higher dose cytarabine (IA) and IA with vorinostat (IA + V). The primary endpoint was event free survival (EFS). 738 patients were randomized: 261 to each DA and IA arms and 216 to the IA + V arm. 96, 456, and 150 patients had favorable-, intermediate-, and unfavorable-risk cytogenetics, respectively. 152 were NPM1 and 158 FLT3 mutated. The overall remission rate was 77.5% including 62.5% CR and 15.0% CRi. No differences in remission, EFS, or overall survival were observed among the 3 arms except for the favorable cytogenetics subset who had improved outcomes with DA and postremission high dose cytarabine. A trend towards increased toxicity was observed with the IA and IA + V arms. The use of higher dose cytarabine during induction therapy in younger patients with AML, with or without vorinostat, does not result in improved outcomes. (Funded by the US National Institutes of Health and others, ClinicalTrials.gov number, NCT01802333.)., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
37. Long-term utilization and benefit of luspatercept in transfusion-dependent, erythropoiesis-stimulating agent-refractory or -intolerant patients with lower-risk myelodysplastic syndromes with ring sideroblasts.
- Author
-
Platzbecker U, Santini V, Komrokji RS, Zeidan AM, Garcia-Manero G, Buckstein R, Miteva D, Keeperman K, Holot N, Nadal JA, Lai Y, Vodala S, Rosettani B, Giuseppi AC, Yucel A, and Fenaux P
- Subjects
- Humans, Erythropoiesis, Activin Receptors, Type II pharmacology, Hematinics therapeutic use, Hematinics pharmacology, Myelodysplastic Syndromes drug therapy
- Published
- 2023
- Full Text
- View/download PDF
38. Characteristics of patients with myelodysplastic neoplasm and spliceosome mutations.
- Author
-
Urrutia S, Li Z, Almanza E, Bataller A, Kanagal-Shamanna R, Senapati J, Sasaki K, Chien K, Montalban-Bravo G, DiNardo C, Borthakur G, Bueso-Ramos C, Pierce S, Kantarjian H, and Garcia-Manero G
- Subjects
- Humans, Spliceosomes genetics, Spliceosomes metabolism, Mutation, RNA Splicing genetics, RNA Splicing Factors genetics, RNA Splicing Factors metabolism, Neoplasms, Myelodysplastic Syndromes genetics
- Published
- 2023
- Full Text
- View/download PDF
39. High-resolution structural variant profiling of myelodysplastic syndromes by optical genome mapping uncovers cryptic aberrations of prognostic and therapeutic significance.
- Author
-
Yang H, Garcia-Manero G, Sasaki K, Montalban-Bravo G, Tang Z, Wei Y, Kadia T, Chien K, Rush D, Nguyen H, Kalia A, Nimmakayalu M, Bueso-Ramos C, Kantarjian H, Medeiros LJ, Luthra R, and Kanagal-Shamanna R
- Subjects
- Chromosome Banding, Chromosome Mapping, Homeodomain Proteins, Humans, Prognosis, Chromosome Aberrations, Myelodysplastic Syndromes
- Abstract
Chromosome banding analysis (CBA) remains the standard-of-care for structural variant (SV) assessment in MDS. Optical genome mapping (OGM) is a novel, non-sequencing-based technique for high-resolution genome-wide SV profiling (SVP). We explored the clinical value of SVP by OGM in 101 consecutive, newly diagnosed MDS patients from a single-center, who underwent standard-of-care cytogenetic and targeted NGS studies. OGM detected 383 clinically significant, recurrent and novel SVs. Of these, 224 (51%) SVs, seen across 34% of patients, were cryptic by CBA (included rearrangements involving MECOM, NUP98::PRRX2, KMT2A partial tandem duplications among others). SVP decreased the proportion of normal karyotype by 16%, identified complex genomes (17%), chromothripsis (6%) and generated informative results in both patients with insufficient metaphases. Precise gene/exon-level mapping allowed assessment of clinically relevant biomarkers (TP53 allele status, KMT2A-PTD) without additional testing. SV data was complementary to NGS. When applied in retrospect, OGM results changed the comprehensive cytogenetic scoring system (CCSS) and R-IPSS risk-groups in 21% and 17% patients respectively with an improved prediction of prognosis. By multivariate analysis, CCSS by OGM only (not CBA), TP53 mutation and BM blasts independently predicted survival. This is the first and largest study reporting the value of combined SVP and NGS for MDS prognostication., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
40. Cooperation between KDM6B overexpression and TET2 deficiency in the pathogenesis of chronic myelomonocytic leukemia.
- Author
-
Wei Y, Kanagal-Shamanna R, Zheng H, Bao N, Lockyer PP, Class CA, Darbaniyan F, Lu Y, Lin K, Yang H, Montalban-Bravo G, Ganan-Gomez I, Soltysiak KA, Do KA, Colla S, and Garcia-Manero G
- Subjects
- Animals, Gene Expression Profiling, Genome, Humans, Loss of Function Mutation, Mice, Mutation, Proto-Oncogene Proteins genetics, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Dioxygenases deficiency, Dioxygenases genetics, Dioxygenases metabolism, Jumonji Domain-Containing Histone Demethylases biosynthesis, Jumonji Domain-Containing Histone Demethylases genetics, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Juvenile genetics, Leukemia, Myelomonocytic, Juvenile metabolism
- Abstract
Loss-of-function TET2 mutations are recurrent somatic lesions in chronic myelomonocytic leukemia (CMML). KDM6B encodes a histone demethylase involved in innate immune regulation that is overexpressed in CMML. We conducted genomic and transcriptomic analyses in treatment naïve CMML patients and observed that the patients carrying both TET2 mutations and KDM6B overexpression constituted 18% of the cohort and 42% of patients with TET2 mutations. We therefore hypothesized that KDM6B overexpression cooperated with TET2 deficiency in CMML pathogenesis. We developed a double-lesion mouse model with both aberrations, and discovered that the mice exhibited a more prominent CMML-like phenotype than mice with either Tet2 deficiency or KDM6B overexpression alone. The phenotype includes monocytosis, anemia, splenomegaly, and increased frequencies and repopulating activity of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs). Significant transcriptional alterations were identified in double-lesion mice, which were associated with activation of proinflammatory signals and repression of signals maintaining genome stability. Finally, KDM6B inhibitor reduced BM repopulating activity of double-lesion mice and tumor burden in mice transplanted with BM-HSPCs from CMML patients with TET2 mutations. These data indicate that TET2 deficiency and KDM6B overexpression cooperate in CMML pathogenesis of and that KDM6B could serve as a potential therapeutic target in this disease., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
41. Genetic correlates in patients with Philadelphia chromosome-positive acute lymphoblastic leukemia treated with Hyper-CVAD plus dasatinib or ponatinib.
- Author
-
Sasaki Y, Kantarjian HM, Short NJ, Wang F, Furudate K, Uryu H, Garris R, Jain N, Sasaki K, Ravandi F, Konopleva M, Garcia-Manero G, Little L, Gumbs C, Zhao L, Futreal PA, Takahashi K, and Jabbour E
- Subjects
- Acute Disease, Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dasatinib therapeutic use, Dexamethasone, Humans, Imidazoles, Pyridazines, Recurrence, Philadelphia Chromosome, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Recurring genetic abnormalities have been identified in Philadelphia chromosome (Ph)-positive acute lymphoblastic leukemia (ALL). Among them, IKZF1 deletion was associated with poor prognosis in patients treated with imatinib-based or dasatinib-based regimens. However, the molecular determinants for clinical outcomes in ponatinib-treated patients remain unknown. We systematically analyzed genetic alterations in adults with Ph-positive ALL uniformly treated in clinical trials with dasatinib-based regimens or a ponatinib-based regimen and investigated the molecular determinants for treatment outcomes using pretreatment specimens collected from adults with Ph-positive ALL treated with Hyper-CVAD plus dasatinib or ponatinib. DNA sequencing and SNP microarray were performed and recurrent genetic abnormalities were found in 84% of the patients, among whom IKZF1 deletion was most frequently detected (60%). IKZF1 deletion frequently co-occurred with other copy-number abnormalities (IKZF1
plus , 46%) and was significantly associated with unfavorable overall survival (OS) (false discovery rate < 0.1) and increased cumulative incidence of relapse (p = 0.01). In a multivariate analysis, dasatinib therapy, lack of achievement of 3-month complete molecular response, and the presence of IKZF1plus status were significantly associated with poor OS. The differential impact of IKZF1plus was largely restricted to patients given Hyper-CVAD plus ponatinib; dasatinib-based regimens had unfavorable outcomes regardless of the molecular abnormalities., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
42. Luspatercept for myelodysplastic syndromes/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis.
- Author
-
Komrokji RS, Platzbecker U, Fenaux P, Zeidan AM, Garcia-Manero G, Mufti GJ, Santini V, Díez-Campelo M, Finelli C, Jurcic JG, Greenberg PL, Sekeres MA, DeZern AE, Savona MR, Shetty JK, Ito R, Zhang G, Ha X, Backstrom JT, and Verma A
- Subjects
- Activin Receptors, Type II, Humans, Immunoglobulin Fc Fragments, Mutation, Recombinant Fusion Proteins, Anemia, Sideroblastic, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Myelodysplastic-Myeloproliferative Diseases drug therapy, Myelodysplastic-Myeloproliferative Diseases genetics, Neoplasms, Thrombocytosis drug therapy
- Published
- 2022
- Full Text
- View/download PDF
43. Correction: Genetic correlates in patients with Philadelphia chromosome-positive acute lymphoblastic leukemia treated with Hyper-CVAD plus dasatinib or ponatinib.
- Author
-
Sasaki Y, Kantarjian HM, Short NJ, Wang F, Furudate K, Uryu H, Garris R, Jain N, Sasaki K, Ravandi F, Konopleva M, Garcia-Manero G, Little L, Gumbs C, Zhao L, Futreal PA, Takahashi K, and Jabbour E
- Published
- 2022
- Full Text
- View/download PDF
44. Marrow ring sideroblasts are highly predictive for TP53 mutation in MDS with excess blasts.
- Author
-
Swoboda DM, Kanagal-Shamanna R, Brunner AM, Cluzeau T, Chan O, Al Ali N, Montalban-Bravo G, Gesiotto QJ, Gavralidis A, Hunter AM, Lee JH, Kuykendall AT, Talati C, Sweet KL, Lancet JE, Padron E, Hussaini M, Song J, Garcia-Manero G, Komrokji RS, and Sallman DA
- Subjects
- Humans, Mutation, RNA Splicing Factors genetics, Tumor Suppressor Protein p53 genetics, Anemia, Sideroblastic genetics, Bone Marrow
- Published
- 2022
- Full Text
- View/download PDF
45. Correction to: Donor clonal hematopoiesis increases risk of acute graft versus host disease after matched sibling transplantation.
- Author
-
Oran B, Champlin RE, Wang F, Tanaka T, Saliba RM, Al-Atrash G, Garcia-Manero G, Kantarjian H, Cao K, Shpall EJ, Alousi AM, Mehta RS, Popat U, Futreal A, and Takahashi K
- Published
- 2022
- Full Text
- View/download PDF
46. Donor clonal hematopoiesis increases risk of acute graft versus host disease after matched sibling transplantation.
- Author
-
Oran B, Champlin RE, Wang F, Tanaka T, Saliba RM, Al-Atrash G, Garcia-Manero G, Kantarjian H, Cao K, Shpall EJ, Alousi AM, Mehta RS, Popat U, Futreal A, and Takahashi K
- Subjects
- Aged, Female, Follow-Up Studies, Graft vs Host Disease etiology, Humans, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Myelodysplastic Syndromes pathology, Prognosis, Survival Rate, Tissue Donors statistics & numerical data, Clonal Hematopoiesis, Graft vs Host Disease pathology, Hematopoietic Stem Cell Transplantation adverse effects, Leukemia, Myeloid, Acute therapy, Myelodysplastic Syndromes therapy, Siblings, Transplantation Conditioning adverse effects
- Abstract
Clonal hematopoiesis (CH) is associated with older age and an increased risk of myeloid malignancies and cardiovascular complications. We analyzed donor DNA samples in patients with AML/MDS who underwent first allogeneic stem cell transplant (SCT) to investigate the association between donor CH and transplant outcomes. We performed targeted deep sequencing of 300 genes on donor blood samples and identified CH with the minimum variant allele frequency of 2%. Among 363 donors, 65 (18%) had CH. The most frequently mutated genes were DNMT3A (31 of 65; 48%), TET2 (16 of 65; 25%), PPM1D (5 of 65, 8%), and ASXL1 (7 of 65; 11%). Transplant outcomes: time to neutrophil and platelet recovery, relapse incidence, transplant-related mortality and progression-free survival, were comparable by donor CH. However, risk of grade II-IV and III-IV acute graft versus host disease (aGvHD) at 6 months after transplant was higher with donor CH vs. without donor CH (hazard ratio (HR) = 2.4, 95% Confidence Interval (CI) = 1.6-3.6, p < 0.001 and HR = 3.8, 95% CI = 1.6-8.9, p = 0.003). In this homogenous population of AML/MDS patients, donor CH was associated with increased risk of grade II-IV and III-IV aGvHD. Further studies to investigate the mechanisms of increased aGvHD and therapeutic interventions to improve aGvHD in the context of donor CH are warranted., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
47. Phase I First-in-Human Dose Escalation Study of the oral SF3B1 modulator H3B-8800 in myeloid neoplasms.
- Author
-
Steensma DP, Wermke M, Klimek VM, Greenberg PL, Font P, Komrokji RS, Yang J, Brunner AM, Carraway HE, Ades L, Al-Kali A, Alonso-Dominguez JM, Alfonso-Piérola A, Coombs CC, Deeg HJ, Flinn I, Foran JM, Garcia-Manero G, Maris MB, McMasters M, Micol JB, De Oteyza JP, Thol F, Wang ES, Watts JM, Taylor J, Stone R, Gourineni V, Marino AJ, Yao H, Destenaves B, Yuan X, Yu K, Dar S, Ohanjanian L, Kuida K, Xiao J, Scholz C, Gualberto A, and Platzbecker U
- Subjects
- Administration, Oral, Aged, Aged, 80 and over, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Dose-Response Relationship, Drug, Female, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Mutation, Missense, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes pathology, Patient Safety, Phosphoproteins genetics, Phosphoproteins metabolism, Piperazines adverse effects, Pyridines adverse effects, RNA Splicing Factors genetics, RNA Splicing Factors metabolism, Treatment Outcome, Leukemia, Myeloid, Acute drug therapy, Myelodysplastic Syndromes drug therapy, Phosphoproteins antagonists & inhibitors, Piperazines therapeutic use, Pyridines therapeutic use, RNA Splicing Factors antagonists & inhibitors
- Abstract
We conducted a phase I clinical trial of H3B-8800, an oral small molecule that binds Splicing Factor 3B1 (SF3B1), in patients with MDS, CMML, or AML. Among 84 enrolled patients (42 MDS, 4 CMML and 38 AML), 62 were red blood cell (RBC) transfusion dependent at study entry. Dose escalation cohorts examined two once-daily dosing regimens: schedule I (5 days on/9 days off, range of doses studied 1-40 mg, n = 65) and schedule II (21 days on/7 days off, 7-20 mg, n = 19); 27 patients received treatment for ≥180 days. The most common treatment-related, treatment-emergent adverse events included diarrhea, nausea, fatigue, and vomiting. No complete or partial responses meeting IWG criteria were observed; however, RBC transfusion free intervals >56 days were observed in nine patients who were transfusion dependent at study entry (15%). Of 15 MDS patients with missense SF3B1 mutations, five experienced RBC transfusion independence (TI). Elevated pre-treatment expression of aberrant transcripts of Transmembrane Protein 14C (TMEM14C), an SF3B1 splicing target encoding a mitochondrial porphyrin transporter, was observed in MDS patients experiencing RBC TI. In summary, H3B-8800 treatment was associated with mostly low-grade TAEs and induced RBC TI in a biomarker-defined subset of MDS., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
48. Autologous CD33-CAR-T cells for treatment of relapsed/refractory acute myelogenous leukemia.
- Author
-
Tambaro FP, Singh H, Jones E, Rytting M, Mahadeo KM, Thompson P, Daver N, DiNardo C, Kadia T, Garcia-Manero G, Chan T, Shah RR, and Wierda WG
- Subjects
- Adolescent, Adult, Aged, Autoantigens, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local pathology, Prognosis, Young Adult, Drug Resistance, Neoplasm, Immunotherapy, Adoptive methods, Leukemia, Myeloid, Acute therapy, Neoplasm Recurrence, Local therapy, Salvage Therapy, Sialic Acid Binding Ig-like Lectin 3 immunology
- Published
- 2021
- Full Text
- View/download PDF
49. Activity of venetoclax-based therapy in chronic myelomonocytic leukemia.
- Author
-
Montalban-Bravo G, Hammond D, DiNardo CD, Konopleva M, Borthakur G, Short NJ, Ramos-Perez J, Guerra V, Kanagal-Shamanna R, Naqvi K, Sasaki K, Jabbour E, Pemmaraju N, Kadia TM, Ravandi F, Daver N, Estrov Z, Pierce S, Kantarjian H, and Garcia-Manero G
- Subjects
- Adult, Aged, Aged, 80 and over, Female, High-Throughput Nucleotide Sequencing methods, Humans, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Myelomonocytic, Chronic genetics, Male, Middle Aged, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Antineoplastic Agents therapeutic use, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Leukemia, Myelomonocytic, Chronic drug therapy, Sulfonamides therapeutic use
- Published
- 2021
- Full Text
- View/download PDF
50. The Clinical impact of PTPN11 mutations in adults with acute myeloid leukemia.
- Author
-
Alfayez M, Issa GC, Patel KP, Wang F, Wang X, Short NJ, Cortes JE, Kadia T, Ravandi F, Pierce S, Assi R, Garcia-Manero G, DiNardo CD, Daver N, Pemmaraju N, Kantarjian H, and Borthakur G
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Cohort Studies, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Nucleophosmin, Prognosis, Survival Rate, Young Adult, Biomarkers, Tumor genetics, Leukemia, Myeloid, Acute mortality, Mutation, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics
- Abstract
While germline and somatic mutations in the gene PTPN11, encoding a phosphatase which regulates the RAS signaling pathway, are well characterized in children with Noonan syndrome and juvenile myelomonocytic leukemia, less is known about their clinical impact in adults with acute myeloid leukemia (AML). To elucidate the effect of PTPN11 mutations (PTPN11
mut ) on clinical outcomes, we screened adult patients with AML treated at our institution using targeted next-generation sequencing. Among 1406 consecutive patients, 112 (8%) had PTPN11mut . These mutations were more commonly associated with the acute myelomonocytic/monocytic leukemia subtype than was wild-type PTPN11, while none were detected in patients with core-binding factor AML. They co-occurred more commonly with NPM1 mutations and FLT3 internal tandem duplications and less commonly with mutations in IDH2 and a complex karyotype. Compared with the wild-type allele, PTPN11mut was associated with lower complete response rates (54% vs 40%; P = 0.04), and shorter overall survival (median 13.6 vs 8.4 months; P = 0.008). In a multivariate analysis, PTPN11mut independently increased the risk of death, with a hazard ratio of 1.69 (95% CI, 1.25-2.29; P = 0.0007). In summary, mutations in PTPN11 have a characteristic phenotype in adults with AML and are associated with an adverse prognosis.- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.