32 results on '"Wang, Eunice S."'
Search Results
2. Hematopoiesis and Hematopoietic Failure
- Author
-
Wang, Eunice S., primary and Berliner, Nancy, additional
- Published
- 2010
- Full Text
- View/download PDF
3. Lead Authors and Contributors
- Author
-
Benjamin, Ivor J., primary, Tariq, Sara G., additional, Beland, Susan S., additional, Benjamin, Ivor J., additional, Bull, David, additional, Hamdan, Mohamed H., additional, Li, Dean Y., additional, Litwin, Sheldon E., additional, Michaels, Andrew D., additional, Morshedzadeh, Jack H., additional, Stehlik, Josef, additional, Whitehead, Kevin J., additional, Victor, Ronald G., additional, Vongpatanasin, Wanpen, additional, Rounds, Sharon I., additional, Aliotta, Jason M., additional, Casserly, Brian, additional, Jankowich, Matthew D., additional, McCool, F. Dennis, additional, Eagle, Kim A., additional, Lau, Wei C., additional, Harris, Raymond C., additional, Andreoli, Thomas E., additional, Basford, Amanda W., additional, Cavanaugh, Kerri L., additional, Dwyer, Jamie P., additional, Golper, Thomas A., additional, Krause, Michelle W., additional, Ikizler, T. Alp, additional, Lewis, Julia B., additional, Luther, James M., additional, Pirkle, James L., additional, Portilla, Didier, additional, Safirstein, Robert L., additional, Schulman, Gerald, additional, Shah, Sudhir V., additional, Zent, Roy, additional, Wolfe, M. Michael, additional, Blanton, Wanda P., additional, Bliss, Charles M., additional, Farraye, Francis A., additional, Huang, Christopher S., additional, Jacobson, Brian C., additional, Lichtenstein, David R., additional, Lowe, Robert, additional, Mishkin, Daniel S., additional, Moore, T. Carlton, additional, Oviedo, Jaime A., additional, Pedrosa, Marcos C., additional, Schimmel, Elihu M., additional, Schroy, Paul C., additional, Singh, Satish K., additional, Tseng, Chi-Chuan, additional, Fallon, Michael B., additional, Arguedas, Miguel R., additional, Garcia-Gallont, Rudolf, additional, Kochar, Rajan, additional, McGuire, Brendan M., additional, Mönkemüller, Klaus, additional, Neumann, Helmut, additional, M. Sheikh, Aasim, additional, Varadarajulu, Shyam, additional, Berliner, Nancy, additional, Lacy, Jill, additional, S. Rinder, Christine, additional, M. Rinder, Henry, additional, G. Rose, Michal, additional, E. Seropian, Stuart, additional, Tormey, Christopher, additional, Torres, Richard, additional, Wang, Eunice S., additional, Griggs, Jennifer J., additional, Burtness, Barbara A., additional, Khorana, Alok A., additional, Lantz, Paula M., additional, Todd, Robert F., additional, Smith, Robert J., additional, Brooks, David G., additional, Gopalakrishnan, Geetha, additional, Hamdy, Osama, additional, Warren, Michelle P., additional, Ziegler, Thomas R., additional, Braunstein, Glenn D., additional, Barnett, Philip S., additional, Herman-Bonert, Vivien S., additional, Friedman, Theodore C., additional, Charney, Pamela A., additional, Carney, Patricia I., additional, Ehrenthal, Deborah B., additional, Kottenhahn, Renee K., additional, Smith, Joseph A., additional, Milam, Douglas F., additional, Starkman, Johnathan S., additional, Stewart, Andrew F., additional, Greenspan, Susan L., additional, Hodak, Steven P., additional, Horwitz, Mara J., additional, LeBeau, Shane O., additional, Roodman, G. David, additional, Moreland, Larry W., additional, Agarwal, Surabhi, additional, Ascherman, Dana P., additional, Domsic, Robyn T., additional, Elliott, Jennifer Rae, additional, Kao, Amy H., additional, Koumpouras, Fotios, additional, Kwoh, C. Kent, additional, Lienesch, Douglas W., additional, McKinnon-Maksimowicz, Kathleen, additional, Medsger, Thomas A., additional, Mohan, Niveditha, additional, Wing, Edward J., additional, Armitage, Keith B., additional, Beckwith, Curt G., additional, Bobak, David A., additional, Fairley, Jessica K., additional, Fulton, Scott A., additional, Hileman, Corrilynn O., additional, Lange, Christoph, additional, Lederman, Michael M., additional, Lemonovich, Tracy L., additional, Lisagaris, Michelle V., additional, Ray, Amy J., additional, Rodriguez, Benigno, additional, Salata, Robert A., additional, Watkins, Richard R., additional, Bradsher, Robert W., additional, Griggs, Robert C., additional, Berg, Michel J., additional, Ciafaloni, Emma, additional, Counihan, Timothy J., additional, Cheshire, William P., additional, de los Reyes, Emily C., additional, Jackson, Carlayne E., additional, Kerber, Kevin A., additional, Liu, Lynn C., additional, Ling, Geoffrey S.F., additional, Lyness, Jeffery M., additional, Lynn, Deborah Joanne, additional, Marshall, Frederick J., additional, McCarthy, Allan, additional, Murphy, Sinéad M., additional, Nath, Avindra, additional, Roach, E. Steve, additional, Rogers, Lisa R., additional, Simon, Roger P., additional, Cohen, Harvey J., additional, Heflin, Mitchell T., additional, Quill, Timothy E., additional, Holloway, Robert G., additional, Hillis, L. David, additional, and Lange, Richard A., additional
- Published
- 2010
- Full Text
- View/download PDF
4. Asciminib plus dasatinib and prednisone for Philadelphia chromosome-positive acute leukemia.
- Author
-
Luskin MR, Murakami MA, Keating JH, Flamand Y, Winer ES, Garcia JS, Stahl M, Stone RM, Wadleigh M, Jaeckle SL, Hagopian E, Weinstock DM, Liegel J, McMasters M, Wang ES, Stock W, and DeAngelo DJ
- Abstract
Dasatinib is effective treatment for Philadelphia chromosome-positive (Ph+) acute leukemia but some patients develop resistance. Combination treatment with dasatinib and asciminib, an allosteric inhibitor of BCR::ABL1, may deepen responses and prevent the emergence of dasatinib-resistant clones. In this phase 1 study (NCT03595017), 24 adults with Ph+ acute lymphoblastic leukemia (ALL, n=22; p190, n=16; p210, n=6) and chronic myeloid leukemia in lymphoid blast crisis (CML-LBC, n=2) were treated with escalating daily doses of asciminib in combination with dasatinib 140 mg daily plus prednisone 60 mg/m2 daily to determine the maximum tolerated dose (MTD). After a 28-day induction, dasatinib and asciminib continued indefinitely or until hematopoietic stem cell transplant. The median age was 64.5 years (range, 33-85; 50% ³65). The recommended phase 2 dose of asciminib was 80 mg daily in combination with dasatinib and prednisone. The dose limiting toxicity at 160 mg daily was asymptomatic grade 3 pancreatic enzyme elevation without symptomatic pancreatitis. There were no vaso-occlusive events. Among patients with de novo ALL, the complete hematologic remission rate at day 28 and 84 was 84% and 100%, respectively. At day 84, 100% of patients achieved complete cytogenetic remission, 89% achieved measurable residual disease negativity (<0.01%) by multicolor flow cytometry, and 74% and 26% achieved BCR::ABL1 RT-PCR <0.1% and <0.01%. Dual BCR::ABL1 inhibition with dasatinib and asciminib is safe with encouraging activity in patients with de novo Ph+ ALL. ClinicalTrials.gov NCT02081378., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
5. How I Treat: Secondary Acute Myeloid Leukemia.
- Author
-
Green SD and Wang ES
- Abstract
Secondary acute myeloid leukemia (sAML) has traditionally been used to designate any AML disease arising from an antecedent hematologic disorder or following prior cytotoxic or radiation therapy. We now know sAML comprises multiple disease entities with distinct clinical and biological features: AML-myelodysplastic related (AML-MR), myeloproliferative neoplasm-blast phase (MPN-BP), and AML post-cytotoxic therapy (AML-pCT). These entities largely represent adverse-risk phenotypes with the majority of patients experiencing suboptimal outcomes with standard therapeutic options. Given the aging general population and the increased lifespan of individuals receiving DNA damaging agents for other medical conditions, the incidence of these diseases is steadily rising and now comprise approximately 25-30% of all new AML diagnoses. Despite the plethora of novel agents approved for AML since 2017, many are either not applicable to sAML (i.e. lacking a targetable mutation), have limited efficacy, or have not been studied in these specific entities. Furthermore, these patients are under-represented in clinical trials, and novel therapeutic options are critically needed. Here we present multiple patient cases exemplifying the new nomenclature and classification of the diseases comprising sAML and highlighting their diverse presentations. We provide our therapeutic approach for each clinical scenario and discuss the challenges of treatment with the currently available armamentarium., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
6. Curing APL in Latin America: more than just ATRA.
- Author
-
Wang ES
- Subjects
- Humans, Latin America epidemiology, Antineoplastic Agents therapeutic use, Leukemia, Promyelocytic, Acute drug therapy, Tretinoin therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
7. Acute myeloid leukemia with LRRFIP1::FGFR1 rearrangement and a complex karyotype.
- Author
-
Qian YW, Wang ES, Sait SJ, and Glenn ST
- Subjects
- Male, Humans, Young Adult, Adult, In Situ Hybridization, Fluorescence, Karyotyping, Karyotype, Translocation, Genetic, RNA-Binding Proteins genetics, Receptor, Fibroblast Growth Factor, Type 1 genetics, Leukemia, Myeloid, Acute genetics
- Abstract
We report a case of a 20-year-old man who presented with splenomegaly, hyperleukocytosis, anemia, and thrombocytopenia. A diagnosis of acute myeloid leukemia (AML) with LRRFIP1::FGFR1 rearrangement with complex karyotype was determined. Chromosome analysis showed a male karyotype: 46,XY,i(1)(q10),t(2;8)(q37;p11.2),der(5)t(1;5) (p22;q13)[17]46,XY[3]. Fluorescence in situ hybridization (FISH) analysis using the Cytocell FGFR1 break apart/amplification probe detected FGFR1 rearrangement with t(2:8) in 126/200 cells analyzed. Other FISH probes including 1p36/ 1q25 probes, del(5q) deletion probe, TLX3 break apart probe, and PDGFRB break apart probe were also utilized to confirm the other karyotypic abnormalities. Next-generation sequencing (NGS) SureSelectXT Custom DNA Target Somatic Detection detected RUNX1 gene mutation. NGS Archer FusionPlex (RNA) confirmed the LRRFIP1::FGFR1 rearrangement. This is the second reported case of AML with LRRFIP1::FGFR1 rearrangement and the first with a complex karyotype., Competing Interests: Declaration of Competing Interest The authors declare no conflict of interest., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. Safety and efficacy of CPX-351 in younger patients (<60 years old) with secondary acute myeloid leukemia.
- Author
-
Przespolewski A, Goldberg AD, Talati C, Fazal S, Vachhani P, Sanikommu SR, Thota S, Waksal J, Ball B, Famulare C, Stahl M, Baron J, Griffiths EA, Thompson JE, Sweet K, and Wang ES
- Subjects
- Humans, Middle Aged, Daunorubicin adverse effects, Cytarabine adverse effects, Antineoplastic Combined Chemotherapy Protocols, Leukemia, Myeloid, Acute drug therapy, Neoplasms, Second Primary
- Published
- 2023
- Full Text
- View/download PDF
9. MDM2 and BCL-2: to p53 or not to p53?
- Author
-
Wang ES
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic, Tumor Suppressor Protein p53 genetics, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism
- Published
- 2023
- Full Text
- View/download PDF
10. An 8-year pragmatic observation evaluation of the benefits of allogeneic HCT in older and medically infirm patients with AML.
- Author
-
Sorror ML, Gooley TA, Storer BE, Gerds AT, Sekeres MA, Medeiros BC, Wang ES, Shami PJ, Adekola K, Luger S, Baer MR, Rizzieri DA, Wildes TM, Koprivnikar J, Smith J, Garrison M, Kojouri K, Schuler TA, Leisenring WM, Onstad LE, Becker PS, McCune JS, Lee SJ, Sandmaier BM, Appelbaum FR, and Estey EH
- Subjects
- Humans, Aged, Quality of Life, Prospective Studies, Remission Induction, Retrospective Studies, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute therapy
- Abstract
We designed a prospective, observational study enrolling patients presenting for treatment of acute myeloid leukemia (AML) at 13 institutions to analyze associations between hematopoietic cell transplantation (HCT) and survival, quality of life (QOL), and function in: the entire cohort, those aged ≥65 years, those with high comorbidity burden, intermediate cytogenetic risk, adverse cytogenetic risk, and first complete remission with or without measurable residual disease. Patient were assessed 8 times over 2 years. Time-dependent regression models were used. Among 692 patients that were evaluable, 46% received HCT with a 2-year survival of 58%. In unadjusted models, HCT was associated with reduced risks of mortality most of the subgroups. However, after accounting for covariates associated with increased mortality (age, comorbidity burden, disease risks, frailty, impaired QOL, depression, and impaired function), the associations between HCT and longer survival disappeared in most subgroups. Although function, social life, performance status, and depressive symptoms were better for those selected for HCT, these health advantages were lost after receiving HCT. Recipients and nonrecipients of HCT similarly ranked and expected cure as main goal of therapy, whereas physicians had greater expectations for cure than the former. Accounting for health impairments negates survival benefits from HCT for AML, suggesting that the unadjusted observed benefit is mostly owing to selection of the healthier candidates. Considering patients' overall expectations of cure but also the QOL burdens of HCT motivate the need for randomized trials to identify the best candidates for HCT. This trial was registered at www.clinicaltrials.gov as #NCT01929408., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
11. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy.
- Author
-
Wang ES, Montesinos P, Minden MD, Lee JH, Heuser M, Naoe T, Chou WC, Laribi K, Esteve J, Altman JK, Havelange V, Watson AM, Gambacorti-Passerini C, Patkowska E, Liu S, Wu R, Philipose N, Hill JE, Gill SC, Rich ES, and Tiu RV
- Subjects
- Adult, Humans, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Pyrazines adverse effects, Azacitidine adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute diagnosis
- Abstract
Treatment results for patients with newly diagnosed FMS-like tyrosine kinase 3 (FLT3)-mutated (FLT3mut+) acute myeloid leukemia (AML) ineligible for intensive chemotherapy are disappointing. This multicenter, open-label, phase 3 trial randomized (2:1) untreated adults with FLT3mut+ AML ineligible for intensive induction chemotherapy to receive gilteritinib (120 mg/d orally) and azacitidine (GIL + AZA) or azacitidine (AZA) alone. The primary end point was overall survival (OS). At the interim analysis (August 26, 2020), a total of 123 patients were randomized to treatment (GIL + AZA, n = 74; AZA, n = 49). Subsequent AML therapy, including FLT3 inhibitors, was received by 20.3% (GIL + AZA) and 44.9% (AZA) of patients. Median OS was 9.82 (GIL + AZA) and 8.87 (AZA) months (hazard ratio, 0.916; 95% CI, 0.529-1.585; P = .753). The study was closed based on the protocol-specified boundary for futility. Median event-free survival was 0.03 month in both arms. Event-free survival defined by using composite complete remission (CRc) was 4.53 months for GIL + AZA and 0.03 month for AZA (hazard ratio, 0.686; 95% CI, 0.433-1.087; P = .156). CRc rates were 58.1% (GIL + AZA) and 26.5% (AZA) (difference, 31.4%; 95% CI, 13.1-49.7; P < .001). Adverse event (AE) rates were similar for GIL + AZA (100%) and AZA (95.7%); grade ≥3 AEs were 95.9% and 89.4%, respectively. Common AEs with GIL + AZA included pyrexia (47.9%) and diarrhea (38.4%). Gilteritinib steady-state trough concentrations did not differ between GIL + AZA and gilteritinib. GIL + AZA resulted in significantly higher CRc rates, although similar OS compared with AZA. Results support the safety/tolerability and clinical activity of upfront therapy with GIL + AZA in older/unfit patients with FLT3mut+ AML. This trial was registered at www.clinicaltrials.gov as #NCT02752035., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
12. Impact of diagnostic genetics on remission MRD and transplantation outcomes in older patients with AML.
- Author
-
Murdock HM, Kim HT, Denlinger N, Vachhani P, Hambley B, Manning BS, Gier S, Cho C, Tsai HK, McCurdy S, Ho VT, Koreth J, Soiffer RJ, Ritz J, Carroll MP, Vasu S, Perales MA, Wang ES, Gondek LP, Devine S, Alyea EP, Lindsley RC, and Gibson CJ
- Subjects
- Aged, Humans, Neoplasm, Residual diagnosis, Neoplasm, Residual genetics, Recurrence, Retrospective Studies, Transplantation Conditioning, Transplantation, Homologous, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
Older patients with acute myeloid leukemia (AML) have high relapse risk and poor survival after allogeneic hematopoietic cell transplantation (HCT). Younger patients may receive myeloablative conditioning to mitigate relapse risk associated with high-risk genetics or measurable residual disease (MRD), but older adults typically receive reduced-intensity conditioning (RIC) to limit toxicity. To identify factors that drive HCT outcomes in older patients, we performed targeted mutational analysis (variant allele fraction ≥2%) on diagnostic samples from 295 patients with AML aged ≥60 years who underwent HCT in first complete remission, 91% of whom received RIC, and targeted duplex sequencing at remission in a subset comprising 192 patients. In a multivariable model for leukemia-free survival (LFS) including baseline genetic and clinical variables, we defined patients with low (3-year LFS, 85%), intermediate (55%), high (35%), and very high (7%) risk. Before HCT, 79.7% of patients had persistent baseline mutations, including 18.3% with only DNMT3A or TET2 (DT) mutations and 61.4% with other mutations (MRD positive). In univariable analysis, MRD positivity was associated with increased relapse and inferior LFS, compared with DT and MRD-negative mutations. However, in a multivariable model accounting for baseline risk, MRD positivity had no independent impact on LFS, most likely because of its significant association with diagnostic genetic characteristics, including MDS-associated gene mutations, TP53 mutations, and high-risk karyotype. In summary, molecular associations with MRD positivity and transplant outcomes in older patients with AML are driven primarily by baseline genetics, not by mutations present in remission. In this group of patients, where high-intensity conditioning carries substantial risk of toxicity, alternative approaches to mitigating MRD-associated relapse risk are needed., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
13. Follow-up of patients with R/R FLT3-mutation-positive AML treated with gilteritinib in the phase 3 ADMIRAL trial.
- Author
-
Perl AE, Larson RA, Podoltsev NA, Strickland S, Wang ES, Atallah E, Schiller GJ, Martinelli G, Neubauer A, Sierra J, Montesinos P, Récher C, Yoon SS, Hosono N, Onozawa M, Chiba S, Kim HJ, Hasabou N, Lu Q, Tiu R, and Levis MJ
- Subjects
- Follow-Up Studies, Humans, Mutation, Pyrazines, Recurrence, fms-Like Tyrosine Kinase 3 genetics, Aniline Compounds pharmacology, Aniline Compounds therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
The phase 3 ADMIRAL (NCT02421939; Study ID: 2215-CL-0301) trial showed superior overall survival in patients with relapsed/refractory FLT3-mutation-positive acute myeloid leukemia (AML) randomized 2:1 to receive the oral FMS-like tyrosine kinase 3 inhibitor gilteritinib vs those randomized to receive salvage chemotherapy (SC). Here we provide a follow-up of the ADMIRAL trial 2 years after the primary analysis to clarify the long-term treatment effects and safety of gilteritinib in these patients with AML. At the time of this analysis, the median survival follow-up was 37.1 months, with deaths in 203 of 247 and 97 of 124 patients in the gilteritinib and SC arms, respectively; 16 gilteritinib-treated patients remained on treatment. The median overall survival for the gilteritinib and SC arms was 9.3 and 5.6 months, respectively (hazard ratio, 0.665; 95% confidence interval [CI], 0.518, 0.853; two-sided P = .0013); 2-year estimated survival rates were 20.6% (95% CI, 15.8, 26.0) and 14.2% (95% CI, 8.3, 21.6). The gilteritinib-arm 2-year cumulative incidence of relapse after composite complete remission was 75.7%, with few relapses occurring after 18 months. Overall, 49 of 247 patients in the gilteritinib arm and 14 of 124 patients in the SC arm were alive for ≥2 years. Twenty-six gilteritinib-treated patients remained alive for ≥2 years without relapse; 18 of these patients underwent transplantation (hematopoietic stem cell transplantation [HSCT]) and 16 restarted gilteritinib as post-HSCT maintenance therapy. The most common adverse events of interest during years 1 and 2 of gilteritinib therapy were increased liver transaminase levels; adverse event incidence decreased in year 2. Thus, continued and post-HSCT gilteritinib maintenance treatment sustained remission with a stable safety profile. These findings confirm that prolonged gilteritinib therapy is safe and is associated with superior survival vs SC. This trial was registered at www.clinicaltrials.gov as #NCT02421939., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
14. Outcomes of Adult Acute Myeloid Leukemia Treated With Gemtuzumab-Ozogamicin: Cue To Optimized Chemotherapy Backbone.
- Author
-
Singh A, Thota S, Bradley T, Griffiths EA, Faber MG, Sadek S, Przespolewski A, Thompson JE, Baron J, Cronin T, Attwood K, Madarang EC, Watts J, and Wang ES
- Subjects
- Adult, Aged, Female, Gemtuzumab pharmacology, Humans, Male, Middle Aged, Treatment Outcome, Gemtuzumab therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
Background: The optimal approach to combine gemtuzumab-ozogamicin (GO) with various chemotherapy backbones and other newer agents safely remains to be determined., Materials and Methods: We performed a retrospective analysis of the safety and outcomes of adult patients with newly diagnosed acute myeloid leukemia (AML) treated with GO with intensified versus standard anthracycline doses (daunorubicin dose 90 mg/m
2 vs 60 mg/m2 ) ± FLT3 inhibitors. The χ2 test and Mann-Whitney U test were used to compare categorical and continuous data. Survival estimates were calculated by Kaplan-Meier method and survival comparisons made using log-rank test., Results: We report a 97% overall response rate in 34 patients with newly diagnosed AML with a median age of 54 years (19-75 years) treated with GO and standard induction. The 11 patients (100%) receiving GO plus daunorubicin dose 90 mg/m2 as part of 7 + 3 induction achieved complete response versus 91% (20/22) complete response in the standard daunorubicin dose group (P = NS). No increased toxicity was noted with the higher daunorubicin dose or when GO and 7 + 3 were combined with FLT3 inhibitors in 3 younger patients (<60 years). Two older patients treated with GO+7 + 3 and FLT3i experienced grade 3 or higher cardiotoxicity. We observed a longer estimated event-free survival for patients with newly diagnosed AML in our cohort (median, 24 months; 95% confidence interval, 17.2 to not reached) compared with historical data., Conclusion: We demonstrate that anthracycline dose intensification with GO may offer higher response rates without increased toxicity in younger patients presenting with de novo AML across European Leukemia Net risk categories., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
15. Acute Myeloid Leukemia: Historical Perspective and Progress in Research and Therapy Over 5 Decades.
- Author
-
Kantarjian HM, Short NJ, Fathi AT, Marcucci G, Ravandi F, Tallman M, Wang ES, and Wei AH
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Prognosis, Time Factors, Treatment Outcome, Leukemia, Myeloid, Acute therapy
- Abstract
With the Food and Drug Administration approval of 9 agents for different acute myeloid leukemia (AML) indications, the prognosis and management of AML is evolving rapidly. Herein, we review the important milestones in the history of AML research and therapy, discuss insights regarding prognostic assessment and prediction of treatment outcome, detail practical supportive care measures, and summarize the current treatment landscape and areas of evolving research., Competing Interests: Disclosure H.K. reports research grants and honoraria from AbbVie, Amgen, Ascentage, BMS, Daiichi-Sankyo, Immunogen, Jazz, Novartis, Pfizer, and Sanofi; and honoraria from Actinium (advisory board), Adaptive Biotechnologies, Aptitude Health, Bio Ascend, Delta Fly, Janssen Global, Oxford Biomedical, and Takeda Oncology. A.H.W. has served on advisory boards for Novartis, Janssen, Amgen, Roche, Pfizer, AbbVie, Servier, Celgene-BMS, MacroGenics, Agios, and Gilead; receives research funding to the institution from Novartis, AbbVie, Servier, Celgene-BMS, Astra Zeneca, and Amgen; serves on speakers bureaus for AbbVie, Novartis, and Celgene; and receives royalty payments from the Walter and Eliza Hall Institute of Medical Research related to venetoclax. M.S.T. has served on advisory boards for AbbVie, Daiichi-Sankyo, Orsenix, KAHR, Delta Fly Pharma, Jazz Pharma, Roche, BioSight, Novartis, Innate Pharmaceuticals, Kura, and Syros Pharmaceuticals; received research funding from AbbVie, Orsenix, BioSight, GlycoMimetics, Rafael Pharmaceuticals, and Amgen; and received royalty from UpToDate. N.J.S. has received consulting fees from Takeda Oncology and AstraZeneca, research funding from Takeda Oncology and Astellas Pharma Inc, and honoraria from Amgen. A.T.F. has provided consulting/advisory services for Agios, BMS/Celgene, Astellas, Seattle Genetics, AbbVie, Pfizer, Genentech, Kite, Amgen, Takeda, Kura, Blueprint, Trillium, Foghorn, Amphivena, NewLink Genetics, Trovagene, Novartis, and MorphoSys. He is receiving research support from Agios, Celgene/BMS, and AbbVie. G.M. has received honoraria from Agios, AbbVie, and Novartis; research support from Merck ESW; has served on advisory boards for AbbVie, Astellas, BMS/Celgene, Genentech, GlaxoSmithKline, Jazz, Kite Pharmaceuticals, Kura Oncology, Novartis, Pfizer, Stemline, and Takeda; provided consulting for Mana Therapeutics; served as a speaker for Stemline, Kura, Pfizer, and DAVA Oncology; and served on data safety monitoring committees for AbbVie and Rafael Pharmaceuticals., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
16. Multisite 11-year experience of less-intensive vs intensive therapies in acute myeloid leukemia.
- Author
-
Sorror ML, Storer BE, Fathi AT, Brunner A, Gerds AT, Sekeres MA, Mukherjee S, Medeiros BC, Wang ES, Vachhani P, Shami PJ, Peña E, Elsawy M, Adekola K, Luger S, Baer MR, Rizzieri D, Wildes TM, Koprivnikar J, Smith J, Garrison M, Kojouri K, Leisenring W, Onstad L, Nyland JE, Becker PS, McCune JS, Lee SJ, Sandmaier BM, Appelbaum FR, and Estey EH
- Subjects
- Adolescent, Adult, Age Factors, Aged, Aged, 80 and over, Disease-Free Survival, Female, Humans, Male, Middle Aged, Prospective Studies, Retrospective Studies, Risk Factors, Survival Rate, Critical Care, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute therapy, Quality of Life
- Abstract
Less-intensive induction therapies are increasingly used in older patients with acute myeloid leukemia (AML). Using an AML composite model (AML-CM) assigning higher scores to older age, increased comorbidity burdens, and adverse cytogenetic risks, we defined 3 distinct prognostic groups and compared outcomes after less-intensive vs intensive induction therapies in a multicenter retrospective cohort (n = 1292) treated at 6 institutions from 2008 to 2012 and a prospective cohort (n = 695) treated at 13 institutions from 2013 to 2017. Prospective study included impacts of Karnofsky performance status (KPS), quality of life (QOL), and physician perception of cure. In the retrospective cohort, recipients of less-intensive therapies were older and had more comorbidities, more adverse cytogenetics, and worse KPS. Less-intensive therapies were associated with higher risks of mortality in AML-CM scores of 4 to 6, 7 to 9, and ≥10. Results were independent of allogeneic transplantation and similar in those age 70 to 79 years. In the prospective cohort, the 2 groups were similar in baseline QOL, geriatric assessment, and patient outcome preferences. Higher mortality risks were seen after less-intensive therapies. However, in models adjusted for age, physician-assigned KPS, and chance of cure, mortality risks and QOL were similar. Less-intensive therapy recipients had shorter length of hospitalization (LOH). Our study questions the survival and QOL benefits (except LOH) of less-intensive therapies in patients with AML, including those age 70 to 79 years or with high comorbidity burdens. A randomized trial in older/medically infirm patients is required to better assess the value of less-intensive and intensive therapies or their combination. This trial was registered at www.clinicaltrials.gov as #NCT01929408., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
17. A Phase II Trial of Imatinib Mesylate as Maintenance Therapy for Patients With Newly Diagnosed C-kit-positive Acute Myeloid Leukemia.
- Author
-
Advani AS, Tse W, Li H, Jia X, Elson P, Cooper B, Ali-Osman F, Park J, Rao AV, Rizzieri DA, Wang ES, Cotta CV, Kalaycio M, Sobecks RM, Rouphail B, Maciejewski JP, Fensterl J, Carew JS, Foster B, Rush ML, Tripp B, Adams D, Corrigan D, Griffiths EA, and Sekeres MA
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Imatinib Mesylate adverse effects, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Maintenance Chemotherapy methods, Male, Middle Aged, Neoplasm Recurrence, Local mortality, Neoplasm Recurrence, Local pathology, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Proto-Oncogene Proteins c-kit metabolism, Young Adult, Imatinib Mesylate administration & dosage, Leukemia, Myeloid, Acute drug therapy, Neoplasm Recurrence, Local drug therapy, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins c-kit antagonists & inhibitors
- Abstract
Introduction: Adults with acute myeloid leukemia (AML) have a high rate of remission; however, more than 50% relapse. C-kit is expressed in approximately 60% of patients with de novo AML and represents a potential therapeutic target., Materials and Methods: Patients with newly diagnosed AML received 12 months of imatinib mesylate as maintenance therapy after the completion of post-remission therapy. The primary objective was to determine whether this approach improved progression-free survival (defined as no relapse and no death) compared with historical controls., Results: The median progression-free survival of patients < 60 years of age was 52.1 months (historical control, 13 months) and for patients ≥ 60 years of age was 10.7 months (historical control, 8 months). The median level of AF1q expression was high (9.59), and 84% of patients had moderate or high levels of drug-resistance factors., Conclusions: Imatinib maintenance therapy may improve the outcome of newly diagnosed patients with AML who are < 60 years of age., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. PARP goes the weasel! Emerging role of PARP inhibitors in acute leukemias.
- Author
-
Fritz C, Portwood SM, Przespolewski A, and Wang ES
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Biomarkers, Tumor, Combined Modality Therapy, DNA Repair drug effects, Drug Synergism, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute metabolism, Mutation, Neoplasm Staging, Poly(ADP-ribose) Polymerase Inhibitors administration & dosage, Poly(ADP-ribose) Polymerase Inhibitors adverse effects, Poly(ADP-ribose) Polymerases metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma etiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Treatment Outcome, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Molecular Targeted Therapy methods, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors, which induce synthetic lethality of BRCA mutant breast and ovarian cancers, are now under active exploration for treatment of acute leukemias, specifically acute myeloid leukemia (AML). Experimental data has revealed that DNA repair deficiencies similar to those found in BRCA mutant solid tumors function in malignant hematopoietic cells to enhance cell survival and promote therapy resistance. Preclinical studies have demonstrated that inhibition of PARP with a variety of agents can dramatically enhance the efficacy of other therapeutic approaches including cytotoxic and epigenetic chemotherapy, small molecule inhibitors (IDH and FLT3 inhibitors) and antibody drug conjugates. This has led to early stage clinical trials of multiple PARP inhibitors (PARPi) for AML patients. Despite small patient numbers, evidence of modest clinical efficacy and tolerability in combinatorial regimens support the further development of PARP inhibition as a novel therapeutic strategy for AML, particularly in select molecular subsets (MLL rearranged, FLT3 and IDH1 mutant disease., (Copyright © 2020. Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
19. Beyond midostaurin: Which are the most promising FLT3 inhibitors in AML?
- Author
-
Wang ES
- Subjects
- Allografts, Humans, Leukemia, Myeloid, Acute enzymology, Protein Kinase Inhibitors, Staurosporine therapeutic use, Antineoplastic Agents therapeutic use, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute therapy, Staurosporine analogs & derivatives, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Abstract
Mutations of FLT3 occur in around a third of acute myeloid leukemia (AML) patients and are associated with poor outcomes. Multiple targeted tyrosine kinase inhibitors (TKI) have been developed with different selectivity and potency for FLT3 mutant clones. Indications for which FLT3 inhibitor to use depend on the clinical setting and disease status. Patients with relapsed or refractory AML benefit from a different TKI than those with de novo AML or following stem cell transplant. Moreover, each FLT3 TKI displays a different toxicity and inhibitory profile and may be most useful in patients with varying comorbidities and types of FLT3 mutations., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
20. Incorporating FLT3 inhibitors in the frontline treatment of FLT3 mutant acute myeloid leukemia.
- Author
-
Wang ES
- Subjects
- Humans, Practice Guidelines as Topic, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute genetics, Mutation, Protein Kinase Inhibitors therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
FLT3 mutations occur in up to a third of newly diagnosed patients with acute myeloid leukemia (AML) and confer poor prognosis. Clinical development of FLT3 tyrosine kinase inhibitors for AML initially involved broad-spectrum inhibitors (midostaurin, sorafenib) targeting multiple kinases. Addition of midostaurin to upfront intensive chemotherapy for younger patients with FLT3 mutant AML significantly improved overall survival and validated FLT3 as a therapeutic target. Other regimens such as sorafenib and hypomethylating agents (azacitidine, decitabine) have expanded the use of FLT3 inhibitors to other populations with FLT3 mutant AML. However, emerging data on new highly potent and specific FLT3 inhibitors such as quizartinib, gilteritinib, and crenolanib suggest that these agents may soon supplant midostaurin and sorafenib in the upfront setting. Using case presentations, this review provides guidelines and practical management strategies for frontline therapy of patients with newly diagnosed FLT3 mutant AML in the current era., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
21. Evolution of acute myelogenous leukemia stem cell properties after treatment and progression.
- Author
-
Ho TC, LaMere M, Stevens BM, Ashton JM, Myers JR, O'Dwyer KM, Liesveld JL, Mendler JH, Guzman M, Morrissette JD, Zhao J, Wang ES, Wetzler M, Jordan CT, and Becker MW
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Biomarkers, Tumor immunology, Cohort Studies, Disease Progression, Female, Humans, Immunophenotyping, Leukemia, Myeloid, Acute immunology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Middle Aged, Neoplasm Transplantation, Neoplastic Stem Cells immunology, Prospective Studies, Recurrence, Young Adult, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Neoplastic Stem Cells pathology
- Abstract
Most cancers evolve over time as patients initially responsive to therapy acquire resistance to the same drugs at relapse. Cancer stem cells have been postulated to represent a therapy-refractory reservoir for relapse, but formal proof of this model is lacking. We prospectively characterized leukemia stem cell populations (LSCs) from a well-defined cohort of patients with acute myelogenous leukemia (AML) at diagnosis and relapse to assess the effect of the disease course on these critical populations. Leukemic samples were collected from patients with newly diagnosed AML before therapy and after relapse, and LSC frequency was assessed by limiting dilution analyses. LSC populations were identified using fluorescent-labeled cell sorting and transplantation into immunodeficient NOD/SCID/interleukin 2 receptor γ chain null mice. The surface antigen expression profiles of pretherapy and postrelapse LSCs were determined for published LSC markers. We demonstrate a 9- to 90-fold increase in LSC frequency between diagnosis and relapse. LSC activity at relapse was identified in populations of leukemic blasts that did not demonstrate this activity before treatment and relapse. In addition, we describe genetic instability and exceptional phenotypic changes that accompany the evolution of these new LSC populations. This study is the first to characterize the evolution of LSCs in vivo after chemotherapy, identifying a dramatic change in the physiology of primitive AML cells when the disease progresses. Taken together, these findings provide a new frame of reference by which to evaluate candidate AML therapies in which both disease control and the induction of more advanced forms of disease should be considered., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
22. Swallowing a bitter pill-oral arsenic trioxide for acute promyelocytic leukemia.
- Author
-
Torka P, Al Ustwani O, Wetzler M, Wang ES, and Griffiths EA
- Subjects
- Administration, Intravenous, Administration, Oral, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Arsenic Trioxide, Arsenicals administration & dosage, Arsenicals adverse effects, Arsenicals pharmacokinetics, Hematologic Neoplasms diagnosis, Hematologic Neoplasms drug therapy, Hematologic Neoplasms mortality, Humans, Leukemia, Promyelocytic, Acute mortality, Leukemia, Promyelocytic, Acute pathology, Oxides administration & dosage, Oxides adverse effects, Oxides pharmacokinetics, Remission Induction, Treatment Outcome, Antineoplastic Agents therapeutic use, Arsenicals therapeutic use, Leukemia, Promyelocytic, Acute drug therapy, Oxides therapeutic use
- Abstract
Parenteral arsenic trioxide (ATO) has been firmly established as a standard therapy for acute promyelocytic leukemia (APL). Despite widespread use of oral arsenicals in medicine historically, they had disappeared from modern pharmacopeia until oral ATO was redeveloped in Hong Kong in 2000. Since then, over 200 patients with leukemia (predominantly APL) have been treated with oral ATO in Hong Kong and China. Oral arsenic trioxide and other formulations of arsenic appear to have a clinical efficacy comparable to that of IV formulations. These drugs given orally also appear to have a slightly better safety profile, lower operational costs and improved convenience for patients. The clinical experience with oral ATO has previously been reported piecemeal as case series, pilot studies or subgroup analyses rather than in a comprehensive cohort. In this report we attempt to synthesize the published English language literature on oral arsenicals and present the argument for further development of these compounds. Systematic study of this drug with well-designed randomized multi-center clinical trials is needed to accelerate its development and incorporation into clinical practice., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Pharmacogenetics predictive of response and toxicity in acute lymphoblastic leukemia therapy.
- Author
-
Mei L, Ontiveros EP, Griffiths EA, Thompson JE, Wang ES, and Wetzler M
- Subjects
- Female, Humans, Male, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Pharmacogenetics methods, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications
- Abstract
Acute lymphoblastic leukemia (ALL) is a relatively rare disease in adults accounting for no more than 20% of all cases of acute leukemia. By contrast with the pediatric population, in whom significant improvements in long term survival and even cure have been achieved over the last 30years, adult ALL remains a significant challenge. Overall survival in this group remains a relatively poor 20-40%. Modern research has focused on improved pharmacokinetics, novel pharmacogenetics and personalized principles to optimize the efficacy of the treatment while reducing toxicity. Here we review the pharmacogenetics of medications used in the management of patients with ALL, including l-asparaginase, glucocorticoids, 6-mercaptopurine, methotrexate, vincristine and tyrosine kinase inhibitors. Incorporating recent pharmacogenetic data, mainly from pediatric ALL, will provide novel perspective of predicting response and toxicity in both pediatric and adult ALL therapies., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
24. Decitabine and Sorafenib Therapy in FLT-3 ITD-Mutant Acute Myeloid Leukemia.
- Author
-
Muppidi MR, Portwood S, Griffiths EA, Thompson JE, Ford LA, Freyer CW, Wetzler M, and Wang ES
- Subjects
- Azacitidine administration & dosage, Azacitidine therapeutic use, Cell Line, Tumor, Decitabine, Female, Humans, Leukemia, Myeloid, Acute mortality, Middle Aged, Mutation, Niacinamide administration & dosage, Niacinamide therapeutic use, Phenylurea Compounds administration & dosage, Protein Kinase Inhibitors administration & dosage, Sorafenib, Survival Analysis, Azacitidine analogs & derivatives, Leukemia, Myeloid, Acute drug therapy, Niacinamide analogs & derivatives, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors therapeutic use, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
Background: Acute myeloid leukemia (AML) characterized by Feline McDonough Sarcoma-like tyrosine kinase-3 (FLT-3) internal tandem duplication (ITD) mutations have poor outcomes. Treatment options are limited, because these mutations confer resistance to conventional chemotherapy. FLT-3 inhibitors such as sorafenib have been studied as a single agent and in combination with conventional chemotherapy or azacytidine with fair responses., Patients and Methods: Here we describe our preclinical and clinical experience with the combination of the DNA hypomethylating agent, decitabine and sorafenib for the treatment of FLT-3 ITD-mutant AML., Results: In vitro treatment of the human FLT-3 ITD-mutant AML cell line, MV4-11, with both drugs significantly improved growth inhibition over single-agent therapy and resulted in synergistic antitumor effects (combination index < 1). A case series of 6 patients treated with off protocol combination of decitabine and sorafenib demonstrated overall responses in 5 patients (83%) with a median survival of 155 days. Four of the 5 patients (80%) with relapsed/refractory AML achieved complete responses with incomplete count recovery. The combination was also well tolerated., Conclusion: Further investigation is warranted to confirm these responses., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. How we will treat chronic myeloid leukemia in 2016.
- Author
-
Talati C, Ontiveros EP, Griffiths EA, Wang ES, and Wetzler M
- Subjects
- Antineoplastic Agents economics, Drug Resistance, Neoplasm, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive economics, Protein Kinase Inhibitors economics, Antineoplastic Agents therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Protein Kinase Inhibitors therapeutic use
- Abstract
Imatinib will become generic in 2016; assuming that its price will decrease precipitously, we expect that the economic forces will change our current practice habits. We reviewed the literature on the current recommendations to treat chronic myeloid leukemia and highlight how we plan to deal with these changes. Specifically, we propose to better characterize patients according to prognostic scores, to allow more attention to those at high risk for disease progression, e.g., 3-month guidelines and BCR/ABL1 message half-time, emphasize compliance by using contemporary technologies, and increase the importance of early monitoring. We hope that our message will open communication between providers, insurance companies and healthcare authorities to offer the best care for our patients., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
26. Phase I/II trial of nanomolecular liposomal annamycin in adult patients with relapsed/refractory acute lymphoblastic leukemia.
- Author
-
Wetzler M, Thomas DA, Wang ES, Shepard R, Ford LA, Heffner TL, Parekh S, Andreeff M, O'Brien S, and Kantarjian HM
- Subjects
- Adult, Aged, Doxorubicin administration & dosage, Doxorubicin adverse effects, Female, Humans, Liposomes administration & dosage, Liposomes adverse effects, Male, Middle Aged, Nanoparticles administration & dosage, Nanoparticles adverse effects, Neoplasm Grading, Recurrence, Young Adult, Antibiotics, Antineoplastic administration & dosage, Antibiotics, Antineoplastic adverse effects, Doxorubicin analogs & derivatives, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Background: Treatment options for relapsed/refractory ALL in adult patients remain challenging. Annamycin is a highly lipophilic form of the anthracycline doxorubicin with the ability to bypass multidrug resistance mechanisms of cellular drug resistance., Patients and Methods: We performed a phase I/II multicenter, open-label, study to determine the maximally tolerated dose (MTD) of nanomolecular liposomal annamycin in adult patients with refractory ALL., Results: Thirty-one patients were enrolled; the MTD was determined to be 150 mg/m(2)/d for 3 days. Other than tumor lysis syndrome, there were 3 grade 3 mucositis which comprised the MTD determination. There was also 1 case each of grade 3 diarrhea, typhlitis, and nausea. After determining the MTD, a 10-patient phase IIA trial was conducted. Eight of the patients completed 1 cycle of the 3 days of treatment at the MTD. Of these, 5 (62%) had an efficacy signal with complete clearing of circulating peripheral blasts. Three of these subjects also cleared bone marrow blasts with 1 subsequently proceeding onto successful stem cell transplantation., Conclusion: Single-agent nanomolecular liposomal annamycin appears to be well tolerated, and shows evidence of clinical activity as a single agent in refractory adult ALL., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
27. Acute myeloid leukemia secondary to oxaliplatin treatment for esophageal cancer.
- Author
-
Damodaran S, Bellavia T, Sait SN, Wang ES, Wetzler M, and Khushalani NI
- Subjects
- Abnormal Karyotype, Adenocarcinoma radiotherapy, Adenocarcinoma surgery, Capecitabine, Clinical Trials, Phase II as Topic, Combined Modality Therapy, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Esophageal Neoplasms radiotherapy, Esophageal Neoplasms surgery, Esophagectomy, Fluorouracil administration & dosage, Fluorouracil analogs & derivatives, Humans, Leukemia, Myeloid, Acute genetics, Male, Middle Aged, Mutation, Oxaliplatin, Radiotherapy, Translocation, Genetic, Adenocarcinoma drug therapy, Antineoplastic Agents adverse effects, Esophageal Neoplasms drug therapy, Leukemia, Myeloid, Acute chemically induced, Organoplatinum Compounds adverse effects
- Published
- 2012
- Full Text
- View/download PDF
28. Interpretation of cytogenetic and molecular results in patients treated for CML.
- Author
-
Vigil CE, Griffiths EA, Wang ES, and Wetzler M
- Subjects
- Benzamides, Cytogenetics, Dasatinib, Disease-Free Survival, Humans, Imatinib Mesylate, Piperazines therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyrimidines therapeutic use, Thiazoles therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics
- Abstract
The International Randomized Study of Interferon vs. STI571 (IRIS) trial that investigated the use of the tyrosine kinase inhibitor (TKI) imatinib (versus interferon) changed the treatment and outcome of chronic myeloid leukemia (CML). Long-term follow-up of IRIS patients has defined response parameters and methods of tracking residual disease with cytogenetic testing of bone marrow metaphases and molecular monitoring of BCR-ABL transcripts using quantitative reverse-transcriptase polymerase chain reaction. Cytogenetic and molecular responses are now considered useful surrogates for long-term outcome. Early and robust response to imatinib predicts positive long-term outcomes. However, 15-25% of patients fail initial treatment or become intolerant of imatinib and need increased doses or alternate treatment. Second-line treatment with the second-generation TKIs nilotinib and dasatinib have resulted in favorable rates of progression-free survival (PFS) and overall survival. Data from the ENESTnd (nilotinib) and DASISION (dasatinib) trials in newly diagnosed chronic-phase CML patients demonstrated more robust and rapid complete cytogenetic (77-80%) and major molecular responses (43-46%) at 12 months compared with imatinib (65-66% and 22-28%). The relationship between a complete cytogenetic response at 12 months and long-term PFS supports a role for second-generation TKIs as first-line treatment of newly diagnosed chronic-phase CML., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
29. Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse.
- Author
-
Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S, Erba H, Stuart RK, Baccarani M, Cripe LD, Tallman MS, Meloni G, Godley LA, Langston AA, Amadori S, Lewis ID, Nagler A, Stone R, Yee K, Advani A, Douer D, Wiktor-Jedrzejczak W, Juliusson G, Litzow MR, Petersdorf S, Sanz M, Kantarjian HM, Sato T, Tremmel L, Bensen-Kennedy DM, Small D, and Smith BD
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carbazoles administration & dosage, Carbazoles adverse effects, Chemotherapy, Adjuvant, Female, Furans, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Mutant Proteins genetics, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors therapeutic use, Recurrence, Remission Induction, Salvage Therapy, Survival Analysis, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carbazoles therapeutic use, Leukemia, Myeloid, Acute drug therapy, fms-Like Tyrosine Kinase 3 genetics
- Abstract
In a randomized trial of therapy for FMS-like tyrosine kinase-3 (FLT3) mutant acute myeloid leukemia in first relapse, 224 patients received chemotherapy alone or followed by 80 mg of the FLT3 inhibitor lestaurtinib twice daily. Endpoints included complete remission or complete remission with incomplete platelet recovery (CR/CRp), overall survival, safety, and tolerability. Correlative studies included pharmacokinetics and analysis of in vivo FLT3 inhibition. There were 29 patients with CR/CRp in the lestaurtinib arm and 23 in the control arm (26% vs 21%; P = .35), and no difference in overall survival between the 2 arms. There was evidence of toxicity in the lestaurtinib-treated patients, particularly those with plasma levels in excess of 20 μM. In the lestaurtinib arm, FLT3 inhibition was highly correlated with remission rate, but target inhibition on day 15 was achieved in only 58% of patients receiving lestaurtinib. Given that such a small proportion of patients on this trial achieved sustained FLT3 inhibition in vivo, any conclusions regarding the efficacy of combining FLT3 inhibition with chemotherapy are limited. Overall, lestaurtinib treatment after chemotherapy did not increase response rates or prolong survival of patients with FLT3 mutant acute myeloid leukemia in first relapse. This study is registered at www.clinicaltrials.gov as #NCT00079482.
- Published
- 2011
- Full Text
- View/download PDF
30. Phase 2 study of romiplostim in patients with low- or intermediate-risk myelodysplastic syndrome receiving azacitidine therapy.
- Author
-
Kantarjian HM, Giles FJ, Greenberg PL, Paquette RL, Wang ES, Gabrilove JL, Garcia-Manero G, Hu K, Franklin JL, and Berger DP
- Subjects
- Aged, Aged, 80 and over, Female, Humans, Incidence, Male, Middle Aged, Recombinant Fusion Proteins adverse effects, Thrombocytopenia complications, Thrombocytopenia epidemiology, Thrombopoietin adverse effects, Antimetabolites, Antineoplastic therapeutic use, Azacitidine therapeutic use, Myelodysplastic Syndromes complications, Myelodysplastic Syndromes drug therapy, Receptors, Fc therapeutic use, Receptors, Thrombopoietin agonists, Recombinant Fusion Proteins therapeutic use, Thrombocytopenia prevention & control, Thrombopoietin therapeutic use
- Abstract
We evaluated the efficacy and safety of romiplostim, a thrombopoietin mimetic, in patients with low- or intermediate-risk myelodysplastic syndromes (MDS) receiving azacitidine therapy. Forty patients with low- or intermediate-risk MDS were stratified by baseline platelet counts (< 50 vs ≥ 50 × 10(9)/L) and randomized to romiplostim 500 μg or 750 μg or placebo subcutaneously once weekly during 4 cycles of azacitidine. The primary endpoint was the incidence of clinically significant thrombocytopenic events, defined by grade 3 or 4 thrombocytopenia starting on day 15 of the first cycle or platelet transfusion at any time during the 4-cycle treatment period. No formal hypothesis testing was planned. The incidence of clinically significant thrombocytopenic events in patients receiving romiplostim 500 μg, romiplostim 750 μg, or placebo was 62%, 71%, and 85%, respectively. The incidence of platelet transfusions was 46%, 36%, and 69%, respectively. These differences were not statistically significant with the small numbers in each group. Romiplostim 750 μg significantly raised median platelet counts during cycle 3 on day 1 (P = .0373) and at the nadir (P = .0035) compared with placebo. Grade 3 rash and arthralgia each were reported in 1 romiplostim-treated patient (4%). This study suggests romiplostim may provide clinical benefits in MDS patients during azacitidine therapy.
- Published
- 2010
- Full Text
- View/download PDF
31. Targeting autocrine and paracrine VEGF receptor pathways inhibits human lymphoma xenografts in vivo.
- Author
-
Wang ES, Teruya-Feldstein J, Wu Y, Zhu Z, Hicklin DJ, and Moore MA
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Autocrine Communication physiology, Cell Proliferation drug effects, Drug Delivery Systems, Humans, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Non-Hodgkin pathology, Mice, Mice, SCID, Neoplasm Transplantation, Paracrine Communication physiology, Receptors, Vascular Endothelial Growth Factor immunology, Transplantation, Heterologous, Vascular Endothelial Growth Factor Receptor-1 immunology, Vascular Endothelial Growth Factor Receptor-1 metabolism, Vascular Endothelial Growth Factor Receptor-2 immunology, Vascular Endothelial Growth Factor Receptor-2 metabolism, Autocrine Communication drug effects, Lymphoma, Large B-Cell, Diffuse pathology, Paracrine Communication drug effects, Receptors, Vascular Endothelial Growth Factor metabolism
- Abstract
The role of angiogenesis in lymphoproliferative diseases is not well established. We demonstrate here that human lymphoma cells secrete vascular endothelial growth factor (VEGF) and express VEGF receptor 1 (VEGFR-1) and VEGFR-2. Proliferation of non-Hodgkin lymphoma (NHL) cells under serum-free conditions was enhanced by the addition of VEGF and was blocked by VEGFR-1- and VEGFR-2-specific antibodies. To differentiate between VEGF-mediated autocrine and paracrine effects on lymphoma growth, NOD/SCID mice engrafted with human diffuse large B-cell lymphoma (DLBCL) were treated with species-specific antibodies against human VEGFR-1 (6.12), human VEGFR-2 (IMC-1C11), murine VEGFR-1 (MF-1), or murine VEGFR-2 (DC101). Treatment with 6.12 or DC101 (targeting tumor VEGFR-1 and host VEGFR-2) reduced established DLBCL xenograft growth, whereas treatment with IMC-1C11 or MF-1 (targeting tumor VEGFR-1 and host VEGFR-1) had no effect. Decreased tumor volumes after 6.12 and DC101 treatment correlated with increased tumor apoptosis and reduced vascularization, respectively, supporting the presence of autocrine VEGFR-1- and paracrine VEGFR-2-mediated pathways in lymphomagenesis. Inhibition of paracrine VEGF interactions (DC101) in these models was equivalent to their inhibition with rituximab. Combining DC101 with therapeutic agents (rituximab, 6.12, methotrexate) consistently improved tumor responses over those of single-agent therapy. These data support the further clinical development of VEGFR-targeted approaches for the therapy of aggressive DLBCL.
- Published
- 2004
- Full Text
- View/download PDF
32. Telomerase inhibition with an oligonucleotide telomerase template antagonist: in vitro and in vivo studies in multiple myeloma and lymphoma.
- Author
-
Wang ES, Wu K, Chin AC, Chen-Kiang S, Pongracz K, Gryaznov S, and Moore MA
- Subjects
- Animals, Apoptosis drug effects, Base Sequence, Cell Division drug effects, Cell Line, Tumor, Humans, In Vitro Techniques, Lymphoma, Non-Hodgkin pathology, Mice, Mice, Inbred NOD, Mice, SCID, Multiple Myeloma pathology, Neoplasm Transplantation, Oligodeoxyribonucleotides, Antisense genetics, Telomere drug effects, Telomere pathology, Transplantation, Heterologous, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Non-Hodgkin enzymology, Multiple Myeloma drug therapy, Multiple Myeloma enzymology, Oligodeoxyribonucleotides, Antisense pharmacology, Telomerase antagonists & inhibitors, Telomerase genetics
- Abstract
The effects of telomerase inhibition with an oligonucleotide N3' --> P5' thiophosphoramidate (GRN163) complementary to the telomerase template region were examined on human multiple myeloma (MM) and non-Hodgkin lymphoma (NHL) cell lines, primary MM cells, and tumor xenografts. GRN163 treatment reduced telomerase levels in all cells and induced more rapid telomeric shortening. Continuous GRN163 treatment for 7 to 14 days resulted in proliferative arrest, morphologic changes, and apoptosis characteristic of cell crisis in tumor cell lines with short (1.7-5.4 kb) but not long (9-11 kb) telomeres. Intratumoral administration of GRN163 also inhibited the growth of MM and NHL xenografts established from cell lines with short telomeres (Hs602 lymphoma, 2.7 kb; CAG myeloma, 2.7 kb) and increased tumor apoptosis. However, GRN163 therapy of NHL xenografts established from cells with long telomeres (11.0 kb) had equivocal effects on tumor growth and did not induce apoptosis during this time frame. Systemic daily intraperitoneal administration of GRN163 in myeloma xenografts with short telomere lengths also decreased tumor telomerase levels and reduced tumor volumes. These data demonstrate that telomerase is important for the replication of mature B-cell neoplasia by stabilizing short telomeres, and they suggest that telomerase inhibition represents a novel therapeutic approach to MM and NHL.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.