429 results on '"Wang, Eunice S."'
Search Results
2. Standardising acute myeloid leukaemia classification systems: a perspective from a panel of international experts
- Author
-
Shallis, Rory M, Daver, Naval, Altman, Jessica K, Komrokji, Rami S, Pollyea, Daniel A, Badar, Talha, Bewersdorf, Jan P, Bhatt, Vijaya R, de Botton, Stéphane, de la Fuente Burguera, Adolfo, Carraway, Hetty E, Desai, Pinkal, Dillon, Richard, Duployez, Nicolas, El Chaer, Firas, Fathi, Amir T, Freeman, Sylvie D, Gojo, Ivana, Grunwald, Michael R, Jonas, Brian A, Konopleva, Marina, Lin, Tara L, Mannis, Gabriel N, Mascarenhas, John, Michaelis, Laura C, Mims, Alice S, Montesinos, Pau, Pozdnyakova, Olga, Pratz, Keith W, Schuh, Andre C, Sekeres, Mikkael A, Smith, Catherine C, Stahl, Maximilian, Subklewe, Marion, Uy, Geoffrey L, Voso, Maria Teresa, Walter, Roland B, Wang, Eunice S, Zeidner, Joshua F, Žučenka, Andrius, and Zeidan, Amer M
- Abstract
The existence of two acute myeloid leukaemia classification systems—one put forth by WHO and one by the International Consensus Classification in 2022—is concerning. Although both systems appropriately move towards genomic disease definitions and reduced emphasis on blast enumeration, there are consequential disagreements between the two systems on what constitutes a diagnosis of acute myeloid leukaemia. This fundamental problem threatens the ability of heath-care providers to diagnose acute myeloid leukaemia, communicate with patients and other health-care providers, and deliver appropriate and consistent management strategies for patients with the condition. Clinical trial eligibility, standardised response assessments, and eventual drug development and regulatory pathways might also be negatively affected by the discrepancies. In this Viewpoint, we review the merits and limitations of both classification systems and illustrate how the coexistence, as well as application of both systems is an undue challenge to patients, clinicians, hematopathologists, sponsors of research, and regulators. Lastly, we emphasise the urgency and propose a roadmap, by which the two divergent classification systems can be harmonised.
- Published
- 2023
- Full Text
- View/download PDF
3. Curing APL in Latin America: more than just ATRA
- Author
-
Wang, Eunice S.
- Published
- 2024
- Full Text
- View/download PDF
4. Safety and efficacy of CPX-351 in younger patients (<60 years old) with secondary acute myeloid leukemia
- Author
-
Przespolewski, Amanda, Goldberg, Aaron D., Talati, Chetasi, Fazal, Salman, Vachhani, Pankit, Sanikommu, Srinivasa R., Thota, Swapna, Waksal, Julian, Ball, Brian, Famulare, Christopher, Stahl, Maximilian, Baron, Jeffrey, Griffiths, Elizabeth A., Thompson, James E., Sweet, Kendra, and Wang, Eunice S.
- Published
- 2023
- Full Text
- View/download PDF
5. CYAD-01, an autologous NKG2D-based CAR T-cell therapy, in relapsed or refractory acute myeloid leukaemia and myelodysplastic syndromes or multiple myeloma (THINK): haematological cohorts of the dose escalation segment of a phase 1 trial
- Author
-
Sallman, David A, Kerre, Tessa, Havelange, Violaine, Poiré, Xavier, Lewalle, Philippe, Wang, Eunice S, Brayer, Jason B, Davila, Marco L, Moors, Ine, Machiels, Jean-Pascal, Awada, Ahmad, Alcantar-Orozco, Erik M, Borissova, Rossitza, Braun, Nathalie, Dheur, Marie-Sophie, Gilham, David E, Lonez, Caroline, Lehmann, Frédéric F, and Flament, Anne
- Abstract
CYAD-01 is an autologous chimeric antigen receptor (CAR) T-cell product based on the natural killer (NK) group 2D (NKG2D) receptor, which binds eight ligands that are overexpressed in a wide range of haematological malignancies but are largely absent on non-neoplastic cells. Initial clinical evaluation of a single infusion of CYAD-01 at a low dose in patients with relapsed or refractory acute myeloid leukaemia, myelodysplastic syndromes, and multiple myeloma supported the feasibility of the approach and prompted further evaluation of CYAD-01. The aim of the present study was to determine the safety and recommended phase 2 dosing of CYAD-01 administered without preconditioning or bridging chemotherapy.
- Published
- 2023
- Full Text
- View/download PDF
6. Asciminib plus dasatinib and prednisone for Philadelphia chromosome–positive acute leukemia
- Author
-
Luskin, Marlise R., Murakami, Mark A., Keating, Julia, Flamand, Yael, Winer, Eric S., Garcia, Jacqueline S., Stahl, Maximilian, Stone, Richard M., Wadleigh, Martha, Jaeckle, Stella L., Hagopian, Ella, Weinstock, David M., Liegel, Jessica, McMasters, Malgorzata, Wang, Eunice S., Stock, Wendy, and DeAngelo, Daniel J.
- Abstract
•Dasatinib, asciminib, and prednisone can be safely combined for the dual targeting of BCR::ABL1 in Ph+acute leukemia.•Dasatinib and asciminib combination treatment has promising efficacy for the upfront treatment of Ph+ALL.
- Published
- 2025
- Full Text
- View/download PDF
7. An 8-year pragmatic observation evaluation of the benefits of allogeneic HCT in older and medically infirm patients with AML
- Author
-
Sorror, Mohamed L., Gooley, Ted A., Storer, Barry E., Gerds, Aaron T., Sekeres, Mikkael A., Medeiros, Bruno C., Wang, Eunice S., Shami, Paul J., Adekola, Kehinde, Luger, Selina, Baer, Maria R., Rizzieri, David A., Wildes, Tanya M., Koprivnikar, Jamie, Smith, Julie, Garrison, Mitchell, Kojouri, Kiarash, Schuler, Tammy A., Leisenring, Wendy M., Onstad, Lynn E., Becker, Pamela S., McCune, Jeannine S., Lee, Stephanie J., Sandmaier, Brenda M., Appelbaum, Frederick R., and Estey, Elihu H.
- 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.
- Published
- 2023
- Full Text
- View/download PDF
8. Olutasidenib alone or with azacitidine in IDH1-mutated acute myeloid leukaemia and myelodysplastic syndrome: phase 1 results of a phase 1/2 trial
- Author
-
Watts, Justin M, Baer, Maria R, Yang, Jay, Prebet, Thomas, Lee, Sangmin, Schiller, Gary J, Dinner, Shira N, Pigneux, Arnaud, Montesinos, Pau, Wang, Eunice S, Seiter, Karen P, Wei, Andrew H, De Botton, Stephane, Arnan, Montserrat, Donnellan, Will, Schwarer, Anthony P, Récher, Christian, Jonas, Brian A, Ferrell, P Brent, Marzac, Christophe, Kelly, Patrick, Sweeney, Jennifer, Forsyth, Sanjeev, Guichard, Sylvie M, Brevard, Julie, Henrick, Patrick, Mohamed, Hesham, and Cortes, Jorge E
- Abstract
Olutasidenib (FT-2102) is a potent, selective, oral, small-molecule inhibitor of mutant isocitrate dehydrogenase 1 (IDH1). The aims for phase 1 of this phase 1/2 study were to assess the safety, pharmacokinetics, pharmacodynamics, and clinical activity of olutasidenib, as monotherapy or in combination with azacitidine, in patients with acute myeloid leukaemia or myelodysplastic syndrome, harbouring mutant IDH1.
- Published
- 2023
- Full Text
- View/download PDF
9. KDM6B protects T-ALL cells from NOTCH1-induced oncogenic stress
- Author
-
Issa, Nancy, Bjeije, Hassan, Wilson, Elisabeth R., Krishnan, Aishwarya, Dunuwille, Wangisa M. B., Parsons, Tyler M., Zhang, Christine R., Han, Wentao, Young, Andrew L., Ren, Zhizhong, Ge, Kai, Wang, Eunice S., Weng, Andrew P., Cashen, Amanda, Spencer, David H., and Challen, Grant A.
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematopoietic neoplasm resulting from the malignant transformation of T-cell progenitors. While activating NOTCH1mutations are the dominant genetic drivers of T-ALL, epigenetic dysfunction plays a central role in the pathology of T-ALL and can provide alternative mechanisms to oncogenesis in lieu of or in combination with genetic mutations. The histone demethylase enzyme KDM6A(UTX) is also recurrently mutated in T-ALL patients and functions as a tumor suppressor. However, its gene paralog, KDM6B(JMJD3), is never mutated and can be significantly overexpressed, suggesting it may be necessary for sustaining the disease. Here, we used mouse and human T-ALL models to show that KDM6Bis required for T-ALL development and maintenance. Using NOTCH1gain-of-function retroviral models, mouse cells genetically deficient for Kdm6bwere unable to propagate T-ALL. Inactivating KDM6Bin human T-ALL patient cells by CRISPR/Cas9 showed KDM6B-targeted cells were significantly outcompeted over time. The dependence of T-ALL cells on KDM6Bwas proportional to the oncogenic strength of NOTCH1mutation, with KDM6B required to prevent stress-induced apoptosis from strong NOTCH1 signaling. These studies identify a crucial role for KDM6Bin sustaining NOTCH1-driven T-ALL and implicate KDM6B as a novel therapeutic target in these patients.
- Published
- 2023
- Full Text
- View/download PDF
10. Update on a Phase 1/2 First-in-Human Study of the Menin-KMT2A (MLL) Inhibitor Ziftomenib (KO-539) in Patients with Relapsed or Refractory Acute Myeloid Leukemia
- Author
-
Erba, Harry P., Fathi, Amir T., Issa, Ghayas C., Altman, Jessica K., Montesinos, Pau, Patnaik, Mrinal M., Foran, James M., De Botton, Stephane, Baer, Maria R., Schiller, Gary J., Walter, Roland B., Kremyanskaya, Marina, Pettit, Kristen M., Strickland, Stephen A, Tomkinson, Blake, Tabachri, Marilyn, Leoni, Mollie, Dale, Stephen, and Wang, Eunice S.
- Published
- 2022
- Full Text
- View/download PDF
11. Broad Activity for the Pivekimab Sunirine (PVEK, IMGN632), Azacitidine, and Venetoclax Triplet in High-Risk Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML)
- Author
-
Daver, Naval, Montesinos, Pau, Aribi, Ahmed, Marconi, Giovanni, Altman, Jessica K., Wang, Eunice S., Roboz, Gail J., Burke, Patrick W., Gaidano, Gianluca, Walter, Roland B., Thomas, Xavier, Jeyakumar, Deepa, DeAngelo, Daniel J., Erba, Harry P., Todisco, Elisabetta, Begna, Kebede H., Advani, Anjali, Gastaud, Lauris, De La Fuente, Adolfo, Curti, Antonio, Mendez, Lourdes M., Vyas, Paresh, Boissel, Nicolas, Vey, Norbert, Recher, Christian, Longval, Thomas, Platzbecker, Uwe, Kapp-Schwoerer, Silke, Schliemann, Christoph, Konopleva, Marina, Torres, Laura, Sallman, David A., Marcucci, Guido, Pemmaraju, Naveen, Martinelli, Giovanni, Kantarjian, Hagop, Sloss, Callum M, Malcolm, Kara E, Zweidler-McKay, Patrick A, and Sweet, Kendra
- Published
- 2022
- Full Text
- View/download PDF
12. Novel Data Analytics Identify Predictors of Quality-of-Life Trajectories in Patients with AML or High-Risk Myeloid Neoplasms
- Author
-
Gauthier, Jordan, Furtuna, Bianca, Mangiavacchi, Jacopo, Gholami, Shahrzad, Lavista Ferres, Juan, Dodhia, Rahul, Fathi, Amir T., Brunner, Andrew M., Gerds, Aaron T., Sekeres, Mikkael A., Medeiros, Bruno C., Wang, Eunice S., Shami, Paul J, Adekola, Kehinde, Luger, Selina M., Baer, Maria R., Rizzieri, David A, Wildes, Tanya, Koprivnikar, Jamie L., Smith, Julie, Garrison, Mitchell A., Kojouri, Kiarash, Appelbaum, Frederick R., Percival, Mary-Elizabeth M., Lee, Stephanie J., and Sorror, Mohamed L.
- Published
- 2022
- Full Text
- View/download PDF
13. Treating octogenarian and nonagenarian acute myeloid leukemia patients - predictive prognostic models
- Author
-
Harb, Antoine J., Tan, Wei, Wilding, Gregory E., Ford, LaurieAnn, Sait, Sheila N.J., Block, AnneMarie W., Barcos, Maurice, Wallace, Paul K., Wang, Eunice S., and Wetzler, Meir
- Subjects
Cancer patients -- Demographic aspects ,Cancer patients -- Prognosis ,Cancer patients -- Research ,Aged -- Health aspects ,Aged -- Research ,Aged patients -- Prognosis ,Aged patients -- Physiological aspects ,Aged patients -- Research ,Health - Published
- 2009
14. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy
- Author
-
Wang, Eunice S., Montesinos, Pau, Minden, Mark D., Lee, Je-Hwan, Heuser, Michael, Naoe, Tomoki, Chou, Wen-Chien, Laribi, Kamel, Esteve, Jordi, Altman, Jessica K., Havelange, Violaine, Watson, Anne-Marie, Gambacorti-Passerini, Carlo, Patkowska, Elzbieta, Liu, Shufang, Wu, Ruishan, Philipose, Nisha, Hill, Jason E., Gill, Stanley C., Rich, Elizabeth Shima, and Tiu, Ramon V.
- 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.
- Published
- 2022
- Full Text
- View/download PDF
15. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+AML ineligible for intensive chemotherapy
- Author
-
Wang, Eunice S., Montesinos, Pau, Minden, Mark D., Lee, Je-Hwan, Heuser, Michael, Naoe, Tomoki, Chou, Wen-Chien, Laribi, Kamel, Esteve, Jordi, Altman, Jessica K., Havelange, Violaine, Watson, Anne-Marie, Gambacorti-Passerini, Carlo, Patkowska, Elzbieta, Liu, Shufang, Wu, Ruishan, Philipose, Nisha, Hill, Jason E., Gill, Stanley C., Rich, Elizabeth Shima, and Tiu, Ramon V.
- Abstract
•In patients with FLT3mut+AML unfit for intensive chemotherapy, CRc rate was higher with GIL + AZA than with AZA.•OS was not significantly different between GIL + AZA and AZA in patients with FLT3mut+AML unfit for intensive chemotherapy.
- Published
- 2022
- Full Text
- View/download PDF
16. Intensive chemotherapy with cyclophosphamide, doxorubicin, high-dose methotrexate/ifosfamide, etoposide, and high-dose cytarabine (CODOX-M/IVAC) for human immunodeficiency virus-associated burkitt lymphoma
- Author
-
Wang, Eunice S., Straus, David J., Teruya-Feldstein, Julie, Qin, Jing, Portlock, Carol, Moskowitz, Craig, Goy, Andre, Hedrick, Eric, Zelenetz, Andrew D., and Noy, Ariela
- Subjects
Chemotherapy -- Methods ,Chemotherapy -- Usage ,HIV (Viruses) -- Risk factors ,HIV (Viruses) -- Care and treatment ,Burkitt's lymphoma -- Risk factors ,Health - Published
- 2003
17. Allelic complexity of KMT2A partial tandem duplications in acute myeloid leukemia and myelodysplastic syndromes
- Author
-
Tsai, Harrison K., Gibson, Christopher J., Murdock, H. Moses, Davineni, Phani, Harris, Marian H., Wang, Eunice S., Gondek, Lukasz P., Kim, Annette S., Nardi, Valentina, and Lindsley, R. Coleman
- Abstract
KMT2A partial tandem duplication (KMT2A-PTD) is an adverse risk factor in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), a potential therapeutic target, and an attractive marker of measurable residual disease. High initial KMT2A-PTD RNA levels have been linked to poor prognosis, but mechanisms regulating KMT2A-PTD expression are not well understood. Although KMT2A-PTD has been reported to affect only a single allele, it has been theorized but not proven that genomic gains of a monoallelic KMT2A-PTD may occur, thereby potentially driving high expression and disease progression. In this study, we identified 94 patients with KMT2A-PTDs using targeted DNA next-generation sequencing (NGS) and found that 16% (15/94) had complex secondary events, including copy-neutral loss of heterozygosity and selective gain involving the KMT2A-PTD allele. High copy numbers indicating complexity were significantly enriched in AML vs MDS and correlated with higher RNA expression. Moreover, in serial samples, complexity was associated with relapse and secondary transformation. Taken together, we provide approaches to integrate quantitative and allelic assessment of KMT2A-PTDs into targeted DNA NGS and demonstrate that secondary genetic events occur in KMT2A-PTD by multiple mechanisms that may be linked to myeloid disease progression by driving increased expression from the affected allele.
- Published
- 2022
- Full Text
- View/download PDF
18. Impact of diagnostic genetics on remission MRD and transplantation outcomes in older patients with AML
- Author
-
Murdock, H. Moses, Kim, Haesook T., Denlinger, Nathan, Vachhani, Pankit, Hambley, Bryan, Manning, Bryan S., Gier, Shannon, Cho, Christina, Tsai, Harrison K., McCurdy, Shannon, Ho, Vincent T., Koreth, John, Soiffer, Robert J., Ritz, Jerome, Carroll, Martin P., Vasu, Sumithira, Perales, Miguel-Angel, Wang, Eunice S., Gondek, Lukasz P., Devine, Steven, Alyea, Edwin P., Lindsley, R. Coleman, and Gibson, Christopher J.
- 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 DNMT3Aor 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, TP53mutations, 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.
- Published
- 2022
- Full Text
- View/download PDF
19. Follow-up of patients with R/R FLT3-mutation–positive AML treated with gilteritinib in the phase 3 ADMIRAL trial
- Author
-
Perl, Alexander E., Larson, Richard A., Podoltsev, Nikolai A., Strickland, Stephen, Wang, Eunice S., Atallah, Ehab, Schiller, Gary J., Martinelli, Giovanni, Neubauer, Andreas, Sierra, Jorge, Montesinos, Pau, Récher, Christian, Yoon, Sung-Soo, Hosono, Naoko, Onozawa, Masahiro, Chiba, Shigeru, Kim, Hee-Je, Hasabou, Nahla, Lu, Qiaoyang, Tiu, Ramon, and Levis, Mark J.
- 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.
- Published
- 2022
- Full Text
- View/download PDF
20. Prediction of life-threatening and disabling bleeding in patients with AML receiving intensive induction chemotherapy
- Author
-
Versluis, Jurjen, Pandey, Manu, Flamand, Yael, Haydu, J. Erika, Belizaire, Roger, Faber, Mark, Vedula, Rahul S., Charles, Anne, Copson, Kevin M., Shimony, Shai, Rozental, Alon, Bendapudi, Pavan K., Wolach, Ofir, Griffiths, Elizabeth A., Thompson, James E., Stone, Richard M., DeAngelo, Daniel J., Neuberg, Donna, Luskin, Marlise R., Wang, Eunice S., and Lindsley, R. Coleman
- Abstract
Bleeding in patients with acute myeloid leukemia (AML) receiving intensive induction chemotherapy is multifactorial and contributes to early death. We sought to define the incidence and risk factors of grade 4 bleeding to support strategies for risk mitigation. Bleeding events were retrospectively assessed between day-14 and day +60 of induction treatment according to the World Health Organization (WHO) bleeding assessment scale, which includes grade 4 bleeding as fatal, life-threatening, retinal with visual impairment, or involving the central nervous system. Predictors were considered pretreatment or prior to grade 4 bleeding. Using multivariable competing-risk regression analysis with grade 4 bleeding as the primary outcome, we identified risk factors in the development cohort (n = 341), which were tested in an independent cohort (n = 143). Grade 4 bleeding occurred in 5.9% and 9.8% of patients in the development and validation cohort, respectively. Risk factors that were independently associated with grade 4 bleeding included baseline platelet count ≤40 × 109/L compared with >40 × 109/L, and baseline international normalized ratio of prothrombin time (PT-INR) >1.5 or 1.3 > 1.5 compared with ≤1.3. These variables were allocated points, which allowed for stratification of patients with low- and high-risk for grade 4 bleeding. Cumulative incidence of grade 4 bleeding at day+60 was significantly higher among patients with high- vs low-risk (development: 31 ± 7% vs 2 ± 1%; P < .001; validation: 25 ± 9% vs 7 ± 2%; P = .008). In both cohorts, high bleeding risk was associated with disseminated intravascular coagulation (DIC) and proliferative disease. We developed and validated a simple risk model for grade 4 bleeding, which enables the development of rational risk mitigation strategies to improve early mortality of intensive induction treatment.
- Published
- 2022
- Full Text
- View/download PDF
21. Molecular, clinical, and prognostic implications of PTPN11 mutations in acute myeloid leukemia
- Author
-
Fobare, Sydney, Kohlschmidt, Jessica, Ozer, Hatice Gulcin, Mrózek, Krzysztof, Nicolet, Deedra, Mims, Alice S., Garzon, Ramiro, Blachly, James S., Orwick, Shelley, Carroll, Andrew J., Stone, Richard M., Wang, Eunice S., Kolitz, Jonathan E., Powell, Bayard L., Oakes, Christopher C., Eisfeld, Ann-Kathrin, Hertlein, Erin, and Byrd, John C.
- Abstract
Prognostic factors associated with chemotherapy outcomes in patients with acute myeloid leukemia (AML) are extensively reported, and one gene whose mutation is recognized as conferring resistance to several newer targeted therapies is protein tyrosine phosphatase non-receptor type 11 (PTPN11). The broader clinical implications of PTPN11 mutations in AML are still not well understood. The objective of this study was to determine which cytogenetic abnormalities and gene mutations co-occur with PTPN11 mutations and how PTPN11 mutations affect outcomes of patients treated with intensive chemotherapy. We studied 1725 patients newly diagnosed with AML (excluding acute promyelocytic leukemia) enrolled onto the Cancer and Leukemia Group B/Alliance for Clinical Trials in Oncology trials. In 140 PTPN11-mutated patient samples, PTPN11 most commonly co-occurred with mutations in NPM1, DNMT3A, and TET2. PTPN11 mutations were relatively common in patients with an inv(3)(q21q26)/t(3;3)(q21;q26) and a normal karyotype but were very rare in patients with typical complex karyotype and core-binding factor AML. Mutations in the N-terminal SH2 domain of PTPN11 were associated with a higher early death rate than those in the phosphatase domain. PTPN11 mutations did not affect outcomes of NPM1-mutated patients, but these patients were less likely to have co-occurring kinase mutations (ie, FLT3-ITD), suggesting activation of overlapping signaling pathways. However, in AML patients with wild-type NPM1, PTPN11 mutations were associated with adverse patient outcomes, providing a rationale to study the biology and treatment approaches in this molecular group. This trial was registered at www.clinicaltrials.gov as #NCT00048958 (CALGB 8461), #NCT00899223 (CALGB 9665), and #NCT00900224 (CALGB 20202).
- Published
- 2022
- Full Text
- View/download PDF
22. MRD Status and ELN 2022 Predicts for Outcomes of Venetoclax + Hypomethylating Therapy in AML: Single-Center Real-World Analysis
- Author
-
Faber, Mark G., Baron, Jeffrey, Yu, Han, Awada, Hassan, Cronin, Tara L., Thompson, James E., Przespolewski, Amanda, Green, Steven D., Sung, Pamela J., Griffiths, Elizabeth A., and Wang, Eunice S.
- Published
- 2022
- Full Text
- View/download PDF
23. Tagraxofusp, an Anti-CD123 Therapy, in Patients with Blastic Plasmacytoid Dendritic Cell Neoplasm and Prior or Concomitant Hematologic Malignancies: Subgroup Analysis of a Pivotal Trial
- Author
-
Pemmaraju, Naveen, Konopleva, Marina, Sweet, Kendra, Stein, Anthony S., Vasu, Sumithira, Rizzieri, David A, Wang, Eunice S., Kantarjian, Hagop, Brooks, Christopher L., Paley, Carole, Mughal, Tariq I., and Lane, Andrew A.
- Published
- 2022
- Full Text
- View/download PDF
24. Ameli-01: A Phase I Trial of UCART123v1.2, an Anti-CD123 Allogeneic CAR-T Cell Product, in Adult Patients with Relapsed or Refractory (R/R) CD123+ Acute Myeloid Leukemia (AML)
- Author
-
Sallman, David A., DeAngelo, Daniel J., Pemmaraju, Naveen, Dinner, Shira, Gill, Saar, Olin, Rebecca L., Wang, Eunice S., Konopleva, Marina, Stark, Eileen, Korngold, Ana, Haider, Asifa, Backhouse, Kate, Figliola, Carolyn, Lee, Daniel J., Frattini, Mark G., Brownstein, Carrie, and Roboz, Gail J.
- Published
- 2022
- Full Text
- View/download PDF
25. Menin Inhibitors in Acute Myeloid Leukemia—What Does the Future Hold?
- Author
-
Swaminathan, Mahesh, Bourgeois, Wallace, Armstrong, Scott A., and Wang, Eunice S.
- Abstract
Menin inhibitors constitute a novel class of agents targeting the underlying biology of nucleophosmin (NPM1) mutant and KMT2A(formerly known as MLL1) rearranged (KMT2Ar) acute leukemias. KMT2Aracute leukemias constitute 5% to 10% of acute leukemias, and NPM1mutations are identified in 30% of newly diagnosed acute myeloid leukemias (AMLs). In preclinical AML models, small molecule inhibitors of the menin-KMT2A protein-protein interaction induce differentiation, downregulate critical gene expression programs, and confer a survival advantage in patient-derived xenograft models of NPM1mutant and KMT2ArAML. Multiple clinical trials evaluating oral menin inhibitors in acute leukemias are ongoing. Preliminary results in relapsed/refractory NPM1mutant and KMT2ArAML have shown on-target effects, tolerable toxicity, and promising clinical activity. This review details the current clinical experience of menin inhibitors in AML and discusses how these agents can be successfully integrated into future therapeutic approaches.
- Published
- 2022
- Full Text
- View/download PDF
26. Harnessing the benefits of available targeted therapies in acute myeloid leukaemia
- Author
-
Kantarjian, Hagop, Short, Nicholas J, DiNardo, Courtney, Stein, Eytan M, Daver, Naval, Perl, Alexander E, Wang, Eunice S, Wei, Andrew, and Tallman, Martin
- Abstract
Research has resulted in regulatory approval of nine agents for acute myeloid leukaemia indications by the US Food and Drug Administration since 2017: the Bcl-2 inhibitor, venetoclax; two FLT3 inhibitors, midostaurin and gilteritinib; two IDH inhibitors, ivosidenib (IDH1 inhibitor) and enasidenib (IDH2 inhibitor); the anti-CD33 antibody–drug conjugate, gemtuzumab ozogamicin; the oral, poorly absorbable hypomethylating agent, azacitidine; the liposomal formulation of cytarabine and daunorubicin (5:1 ratio), CPX-351; and the hedgehog signalling pathway inhibitor, glasdegib. A 100% absorbable oral formulation of the hypomethylating agent decitabine was approved for the treatment of myelodysplastic syndrome and chronic myelomonocytic leukaemia, and might be used as an alternative to parenteral hypomethylating agents. Several of the approvals are as single-agent therapies or in specific combinations for narrow indications, thus offering poor treatment value. In this Review, we discuss ongoing research into combinations containing these commercially available targeted therapies for acute myeloid leukaemia.
- Published
- 2021
- Full Text
- View/download PDF
27. Multisite 11-year experience of less-intensive vs intensive therapies in acute myeloid leukemia
- Author
-
Sorror, Mohamed L., Storer, Barry E., Fathi, Amir T., Brunner, Andrew, Gerds, Aaron T., Sekeres, Mikkael A., Mukherjee, Sudipto, Medeiros, Bruno C., Wang, Eunice S., Vachhani, Pankit, Shami, Paul J., Peña, Esteban, Elsawy, Mahmoud, Adekola, Kehinde, Luger, Selina, Baer, Maria R., Rizzieri, David, Wildes, Tanya M., Koprivnikar, Jamie, Smith, Julie, Garrison, Mitchell, Kojouri, Kiarash, Leisenring, Wendy, Onstad, Lynn, Nyland, Jennifer E., Becker, Pamela S., McCune, Jeannine S., Lee, Stephanie J., Sandmaier, Brenda M., Appelbaum, Frederick R., and Estey, Elihu H.
- 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.
- Published
- 2021
- Full Text
- View/download PDF
28. Germline variants drive myelodysplastic syndrome in young adults
- Author
-
Feurstein, Simone, Churpek, Jane E., Walsh, Tom, Keel, Sioban, Hakkarainen, Marja, Schroeder, Thomas, Germing, Ulrich, Geyh, Stefanie, Heuser, Michael, Thol, Felicitas, Pohlkamp, Christian, Haferlach, Torsten, Gao, Juehua, Owen, Carolyn, Goehring, Gudrun, Schlegelberger, Brigitte, Verma, Divij, Krause, Daniela S., Gao, Guimin, Cronin, Tara, Gulsuner, Suleyman, Lee, Ming, Pritchard, Colin C., Subramanian, Hari Prasanna, del Gaudio, Daniela, Li, Zejuan, Das, Soma, Kilpivaara, Outi, Wartiovaara-Kautto, Ulla, Wang, Eunice S., Griffiths, Elizabeth A., Döhner, Konstanze, Döhner, Hartmut, King, Mary-Claire, and Godley, Lucy A.
- Published
- 2021
- Full Text
- View/download PDF
29. Phase 3 randomized trial of chemotherapy with or without oblimersen in older AML patients: CALGB 10201 (Alliance)
- Author
-
Walker, Alison R., Marcucci, Guido, Yin, Jun, Blum, William, Stock, Wendy, Kohlschmidt, Jessica, Mrózek, Krzysztof, Carroll, Andrew J., Eisfeld, Ann-Kathrin, Wang, Eunice S., Jacobson, Sawyer, Kolitz, Jonathan E., Thakuri, Mohan, Sutamtewagul, Grerk, Vij, Ravi, Stuart, Robert K., Byrd, John C., Bloomfield, Clara D., Stone, Richard M., and Larson, Richard A.
- Abstract
Overexpression of B-cell leukemia/lymphoma 2 (BCL2) renders acute myeloid leukemia (AML) cells resistant to chemotherapy and has been associated with unfavorable outcomes. Oblimersen (G3139) is a phosphorothioate 18-mer antisense oligonucleotide directed against the first 6 BCL2 codons. In a phase 1 study of AML patients treated with G3139, cytarabine, and daunorubicin induction with cytarabine consolidation, no antisense-related toxicity was reported, and BCL2 downregulation occurred in patients achieving complete remission. In this phase 3 trial, untreated older AML patients were randomized to cytarabine (100 mg/m2 per day on days 4-10) and daunorubicin (60 mg/m2 per day on days 4-6) followed by cytarabine consolidation (2000 mg/m2 per day on days 4-8) with (arm A) or without (arm B) G3139 (7 mg/m2 per day on days 1-10 [induction] or days 1-8 [consolidation]). A total of 506 patients were enrolled. No differences in toxicity were observed between arms. Estimated overall survival (OS) at 1 year was 43% for arm A and 40% for arm B (1-sided log rank P = .13), with no differences in disease-free (DFS; P = .26) or event-free survival (P = .80). Subgroup analyses showed patients age <70 years in arm A had improved OS by 1 month vs those in arm B (P = .04), and patients with secondary AML in arm A had better DFS vs those in arm B (P = .04). We conclude that addition of G3139 to chemotherapy failed to improve outcomes of older AML patients. However, more effective means of inhibiting BCL2 are showing promising results in combination with chemotherapy in AML. This trial was registered at www.clinicaltrials.gov as #NCT00085124.
- Published
- 2021
- Full Text
- View/download PDF
30. Inhibiting autophagy targets human leukemic stem cells and hypoxic AML blasts by disrupting mitochondrial homeostasis
- Author
-
Dykstra, Kaitlyn M., Fay, Hannah R. S., Massey, Ashish C., Yang, Neng, Johnson, Matthew, Portwood, Scott, Guzman, Monica L., and Wang, Eunice S.
- Abstract
Leukemia stem cells (LSCs) and therapy-resistant acute myeloid leukemia (AML) blasts contribute to the reinitiation of leukemia after remission, necessitating therapeutic interventions that target these populations. Autophagy is a prosurvival process that allows for cells to adapt to a variety of stressors. Blocking autophagy pharmacologically by using mechanistically distinct inhibitors induced apoptosis and prevented colony formation in primary human AML cells. The most effective inhibitor, bafilomycin A1 (Baf A1), also prevented the in vivo maintenance of AML LSCs in NSG mice. To understand why Baf A1 exerted the most dramatic effects on LSC survival, we evaluated mitochondrial function. Baf A1 reduced mitochondrial respiration and stabilized PTEN-induced kinase-1 (PINK-1), which initiates autophagy of mitochondria (mitophagy). Interestingly, with the autophagy inhibitor chloroquine, levels of enhanced cell death and reduced mitochondrial respiration phenocopied the effects of Baf A1 only when cultured in hypoxic conditions that mimic the marrow microenvironment (1% O2). This indicates that increased efficacy of autophagy inhibitors in inducing AML cell death can be achieved by concurrently inducing mitochondrial damage and mitophagy (pharmacologically or by hypoxic induction) and blocking mitochondrial degradation. In addition, prolonged exposure of AML cells to hypoxia induced autophagic flux and reduced chemosensitivity to cytarabine (Ara-C), which was reversed by autophagy inhibition. The combination of Ara-C with Baf A1 also decreased tumor burden in vivo. These findings demonstrate that autophagy is critical for mitochondrial homeostasis and survival of AML cells in hypoxia and support the development of autophagy inhibitors as novel therapeutic agents for AML.
- Published
- 2021
- Full Text
- View/download PDF
31. Phase I First-in-Human Dose Escalation Study of the oral SF3B1 modulator H3B-8800 in myeloid neoplasms
- Author
-
Steensma, David P., Wermke, Martin, Klimek, Virginia M., Greenberg, Peter L., Font, Patricia, Komrokji, Rami S., Yang, Jay, Brunner, Andrew M., Carraway, Hetty E., Ades, Lionel, Al-Kali, Aref, Alonso-Dominguez, Juan M., Alfonso-Piérola, Ana, Coombs, Catherine C., Deeg, H. Joachim, Flinn, Ian, Foran, James M., Garcia-Manero, Guillermo, Maris, Michael B., McMasters, Malgorzata, Micol, Jean-Baptiste, De Oteyza, Jaime Perez, Thol, Felicitas, Wang, Eunice S., Watts, Justin M., Taylor, Justin, Stone, Richard, Gourineni, Vikram, Marino, Alyssa J., Yao, Huilan, Destenaves, Benoit, Yuan, Xiaobin, Yu, Kun, Dar, Sara, Ohanjanian, Lernik, Kuida, Keisuke, Xiao, Jianjun, Scholz, Catherine, Gualberto, Antonio, and Platzbecker, Uwe
- 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.
- Published
- 2021
- Full Text
- View/download PDF
32. Management of toxicities associated with targeted therapies for acute myeloid leukemia: when to push through and when to stop
- Author
-
Wang, Eunice S. and Baron, Jeffrey
- Abstract
The recent advent of myriad targeted therapies for acute myeloid leukemia (AML) has led to new hope for our patients but has also introduced new challenges in managing the disease. For clinicians, the ability to treat AML in the outpatient setting with novel agents of equal or greater efficacy than 7+3 has been transformative. Despite the enthusiasm, however, the reality is that many patients are still frail and remain at risk for treatment-related complications. Translating the results of clinical trials into improved outcomes for these individuals requires an understanding of how best to manage the adverse effects of these agents. Which patients benefit most and what to watch for? When to stop therapy? Using illustrative case presentations, this review details the unique toxicities associated with each of the approved mutation-specific and nonspecific targeted drugs for AML. The goal of this review is to help clinicians determine the risk:benefit ratio in decision making for individual patients with AML.
- Published
- 2020
- Full Text
- View/download PDF
33. Safety and efficacy of BAY1436032 in IDH1-mutant AML: phase I study results
- Author
-
Heuser, Michael, Palmisiano, Neil, Mantzaris, Ioannis, Mims, Alice, DiNardo, Courtney, Silverman, Lewis R., Wang, Eunice S., Fiedler, Walter, Baldus, Claudia, Schwind, Sebastian, Pardee, Timothy, Perl, Alexander E., Cai, Charles, Kaulfuss, Stefan, Lagkadinou, Eleni, Rentzsch, Christine, Wagner, Markus, Wilkinson, Gary, Wu, Bingyan, Jeffers, Michael, Genvresse, Isabelle, and Krämer, Alwin
- Abstract
The mutant IDH1 (mIDH1) inhibitor BAY1436032 demonstrated robust activity in preclinical AML models, supporting clinical evaluation. In the current dose-escalation study, BAY1436032 was orally administered to 27 mIDH1 AML subjects across 4 doses ranging from 300 to 1500 mg twice-daily. BAY1436032 exhibited a relatively short half-life and apparent non-linear pharmacokinetics after continuous dosing. Most subjects experienced only partial target inhibition as indicated by plasma R-2HG levels. BAY1436032 was safe and a maximum tolerated dose was not identified. The median treatment duration for all subjects was 3.0 months (0.49–8.5). The overall response rate was 15% (4/27; 1 CRp, 1 PR, 2 MLFS), with responding subjects experiencing a median treatment duration of 6.0 months (3.9–8.5) and robust R-2HG decreases. Thirty percent (8/27) achieved SD, with a median treatment duration of 5.5 months (3.1–7.0). Degree of R-2HG inhibition and clinical benefit did not correlate with dose. Although BAY1436032 was safe and modestly effective as monotherapy, the low overall response rate and incomplete target inhibition achieved at even the highest dose tested do not support further clinical development of this investigational agent in AML.
- Published
- 2020
- Full Text
- View/download PDF
34. Phase 2 study of ruxolitinib and decitabine in patients with myeloproliferative neoplasm in accelerated and blast phase
- Author
-
Mascarenhas, John O., Rampal, Raajit K., Kosiorek, Heidi E., Bhave, Rupali, Hexner, Elizabeth, Wang, Eunice S., Gerds, Aaron, Abboud, Camille N., Kremyanskaya, Marina, Berenzon, Dimitry, Odenike, Olatoyosi, Farnoud, Noushin, Krishnan, Aishwarya, Weinberg, Rona Singer, McGovern, Erin, Salama, Mohamed E., Najfeld, Vesna, Medina-Martinez, Juan S., Arango Ossa, Juan E., Levine, Max F., Zhou, Yangyu, Sandy, Lonette, Heaney, Mark L., Levine, Ross L., Mesa, Ruben A., Dueck, Amylou C., and Hoffman, Ronald
- Abstract
Myeloproliferative neoplasms (MPN) that have evolved into accelerated or blast phase disease (MPN-AP/BP) have poor outcomes with limited treatment options and therefore represent an urgent unmet need. We have previously demonstrated in a multicenter, phase 1 trial conducted through the Myeloproliferative Neoplasms Research Consortium that the combination of ruxolitinib and decitabine is safe and tolerable and is associated with a favorable overall survival (OS). In this phase 2 trial, 25 patients with MPN-AP/BP were treated at the recommended phase 2 dose of ruxolitinib 25 mg twice daily for the induction cycle followed by 10 mg twice daily for subsequent cycles in combination with decitabine 20 mg/m2 for 5 consecutive days in a 28-day cycle. Nineteen patients died during the study follow-up. The median OS for all patients on study was 9.5 months (95% confidence interval, 4.3-12.0). Overall response rate (complete remission + incomplete platelet recovery + partial remission) was 11/25 (44%) and response was not associated with improved survival. We conclude that the combination of decitabine and ruxolitinib was well tolerated, demonstrated favorable OS, and represents a therapeutic option for this high-risk patient population. This trial was registered at www.clinicaltrials.gov as #NCT02076191.
- Published
- 2020
- Full Text
- View/download PDF
35. Special considerations in the management of adult patients with acute leukaemias and myeloid neoplasms in the COVID-19 era: recommendations from a panel of international experts
- Author
-
Zeidan, Amer M, Boddu, Prajwal C, Patnaik, Mrinal M, Bewersdorf, Jan Philipp, Stahl, Maximilian, Rampal, Raajit K, Shallis, Rory, Steensma, David P, Savona, Michael R, Sekeres, Mikkael A, Roboz, Gail J, DeAngelo, Daniel J, Schuh, Andre C, Padron, Eric, Zeidner, Joshua F, Walter, Roland B, Onida, Francesco, Fathi, Amir, DeZern, Amy, Hobbs, Gabriela, Stein, Eytan M, Vyas, Paresh, Wei, Andrew H, Bowen, David T, Montesinos, Pau, Griffiths, Elizabeth A, Verma, Amit K, Keyzner, Alla, Bar-Natan, Michal, Navada, Shyamala C, Kremyanskaya, Marina, Goldberg, Aaron D, Al-Kali, Aref, Heaney, Mark L, Nazha, Aziz, Salman, Huda, Luger, Selina, Pratz, Keith W, Konig, Heiko, Komrokji, Rami, Deininger, Michael, Cirici, Blanca Xicoy, Bhatt, Vijaya Raj, Silverman, Lewis R, Erba, Harry P, Fenaux, Pierre, Platzbecker, Uwe, Santini, Valeria, Wang, Eunice S, Tallman, Martin S, Stone, Richard M, and Mascarenhas, John
- Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 is a global public health crisis. Multiple observations indicate poorer post-infection outcomes for patients with cancer than for the general population. Herein, we highlight the challenges in caring for patients with acute leukaemias and myeloid neoplasms amid the COVID-19 pandemic. We summarise key changes related to service allocation, clinical and supportive care, clinical trial participation, and ethical considerations regarding the use of lifesaving measures for these patients. We recognise that these recommendations might be more applicable to high-income countries and might not be generalisable because of regional differences in health-care infrastructure, individual circumstances, and a complex and highly fluid health-care environment. Despite these limitations, we aim to provide a general framework for the care of patients with acute leukaemias and myeloid neoplasms during the COVID-19 pandemic on the basis of recommendations from international experts.
- Published
- 2020
- Full Text
- View/download PDF
36. A phase 1/2 study of the oral FLT3 inhibitor pexidartinib in relapsed/refractory FLT3-ITD–mutant acute myeloid leukemia
- Author
-
Smith, Catherine C., Levis, Mark J., Frankfurt, Olga, Pagel, John M., Roboz, Gail J., Stone, Richard M., Wang, Eunice S., Severson, Paul L., West, Brian L., Le, Mai H., Kayser, Sabine, Lam, Bao, Hsu, Henry H., Zhang, Chao, Bollag, Gideon, and Perl, Alexander E.
- Abstract
FMS-like tyrosine kinase 3 (FLT3) tyrosine kinase inhibitors (TKIs) have activity in acute myeloid leukemia (AML) patients with FLT3 internal tandem duplication (ITD) mutations, but efficacy is limited by resistance-conferring kinase domain mutations. This phase 1/2 study evaluated the safety, tolerability, and efficacy of the oral FLT3 inhibitor PLX3397 (pexidartinib), which has activity against the FLT3 TKI–resistant F691L gatekeeper mutation in relapsed/refractory FLT3-ITD–mutant AML. Ninety patients were treated: 34 in dose escalation (part 1) and 56 in dose expansion (part 2). Doses of 800 to 5000 mg per day in divided doses were tested. No maximally tolerated dose was reached. Plasma inhibitory assay demonstrated that patients dosed with ≥3000 mg had sufficient levels of active drug in their trough plasma samples to achieve 95% inhibition of FLT3 phosphorylation in an FLT3-ITD AML cell line. Based on a plateau in drug exposure, the 3000-mg dose was chosen as the recommended phase 2 dose. The most frequently reported treatment-emergent adverse events were diarrhea (50%), fatigue (47%), and nausea (46%). Based on modified response criteria, the overall response rate to pexidartinib among all patients was 21%. Twenty-three percent of patients treated at ≥2000 mg responded. The overall composite complete response rate for the study was 11%. Six patients were successfully bridged to transplantation. Median overall survival (OS) of patients treated in dose expansion was 112 days (90% confidence interval [CI], 77-150 days), and median OS of responders with complete remission with or without recovery of blood counts was 265 days (90% CI, 170-422 days). This trial was registered at www.clinicaltrials.gov as #NCT01349049.
- Published
- 2020
- Full Text
- View/download PDF
37. Combination of dasatinib with chemotherapy in previously untreated core binding factor acute myeloid leukemia: CALGB 10801
- Author
-
Marcucci, Guido, Geyer, Susan, Laumann, Kristina, Zhao, Weiqiang, Bucci, Donna, Uy, Geoffrey L., Blum, William, Eisfeld, Ann-Kathrin, Pardee, Timothy S., Wang, Eunice S., Stock, Wendy, Kolitz, Jonathan E., Kohlschmidt, Jessica, Mrózek, Krzysztof, Bloomfield, Clara D., Stone, Richard M., and Larson, Richard A.
- Abstract
Acute myeloid leukemia (AML) with either t(8;21)(q22;q22) or inv(16)(p13q22)/t(16;16)(p13;q22) is referred to as core binding factor (CBF) AML. Although categorized as favorable risk, long-term survival for these patients is only ∼50% to 60%. Mutated (mut) or overexpressed KIT, a gene encoding a receptor tyrosine kinase, has been found almost exclusively in CBF AML and may increase the risk of disease relapse. We tested the safety and clinical activity of dasatinib, a multi-kinase inhibitor, in combination with chemotherapy. Sixty-one adult patients with AML and CBF fusion transcripts (RUNX1/RUNX1T1 or CBFB/MYH11) were enrolled on Cancer and Leukemia Group B (CALGB) 10801. Patients received cytarabine/daunorubicin induction on days 1 to 7 and oral dasatinib 100 mg/d on days 8 to 21. Upon achieving complete remission, patients received consolidation with high-dose cytarabine followed by dasatinib 100 mg/d on days 6 to 26 for 4 courses, followed by dasatinib 100 mg/d for 12 months. Fifteen (25%) patients were older (aged ≥60 years); 67% were CBFB/MYH11–positive, and 19% harbored KITmut. There were no unexpected or dose-limiting toxicities. Fifty-five (90%) patients achieved complete remission. With a median follow-up of 45 months, only 16% have relapsed. The 3-year disease-free survival and overall survival rates were 75% and 77% (79% and 85% for younger patients [aged <60 years], and 60% and 51% for older patients). Patients with KITmut had comparable outcome to those with wild-type KIT (3-year rates: disease-free survival, 67% vs 75%; overall survival, 73% vs 76%), thereby raising the question of whether dasatinib may overcome the negative impact of these genetic lesions. CALGB 10801 was registered at www.clinicaltrials.gov as #NCT01238211.
- Published
- 2020
- Full Text
- View/download PDF
38. Clinical and functional significance of circular RNAs in cytogenetically normal AML
- Author
-
Papaioannou, Dimitrios, Volinia, Stefano, Nicolet, Deedra, Świerniak, Michał, Petri, Andreas, Mrózek, Krzysztof, Bill, Marius, Pepe, Felice, Walker, Christopher J., Walker, Allison E., Carroll, Andrew J., Kohlschmidt, Jessica, Eisfeld, Ann-Kathrin, Powell, Bayard L., Uy, Geoffrey L., Kolitz, Jonathan E., Wang, Eunice S., Kauppinen, Sakari, Dorrance, Adrienne, Stone, Richard M., Byrd, John C., Bloomfield, Clara D., and Garzon, Ramiro
- Abstract
Circular RNAs (circRNAs) are noncoding RNA molecules that display a perturbed arrangement of exons, called backsplicing. To examine the prognostic and biologic significance of circRNA expression in cytogenetically normal acute myeloid leukemia (CN-AML), we conducted whole-transcriptome profiling in 365 younger adults (age 18-60 years) with CN-AML. We applied a novel pipeline, called Massive Scan for circRNA, to identify and quantify circRNA expression. We validated the high sensitivity and specificity of our pipeline by performing RNase R treatment and RNA sequencing in samples of AML patients and cell lines. Unsupervised clustering analyses identified 3 distinct circRNA expression–based clusters with different frequencies of clinical and molecular features. After dividing our cohort into training and validation data sets, we identified 4 circRNAs (circCFLAR, circKLHL8, circSMC1A, and circFCHO2) that were prognostic in both data sets; high expression of each prognostic circRNA was associated with longer disease-free, overall, and event-free survival. In multivariable analyses, high circKLHL8 and high circFCHO2 expression were independently associated with better clinical outcome of CN-AML patients, after adjusting for other covariates. To examine the biologic relevance of circRNA expression, we performed knockdown screening experiments in a subset of prognostic and gene mutation–related candidate circRNAs. We identified circFBXW7, but not its linear messenger RNA, as a regulator of the proliferative capacity of AML blasts. In summary, our findings underscore the molecular associations, prognostic significance, and functional relevance of circRNA expression in CN-AML.
- Published
- 2020
- Full Text
- View/download PDF
39. What potential is there for LSD1 inhibitors to reach approval for AML?
- Author
-
Pandey, Manu R. and Wang, Eunice S.
- Published
- 2019
- Full Text
- View/download PDF
40. Phase 1b study of the MDM2 inhibitor AMG 232 with or without trametinib in relapsed/refractory acute myeloid leukemia
- Author
-
Erba, Harry P., Becker, Pamela S., Shami, Paul J., Grunwald, Michael R., Flesher, Donna L., Zhu, Min, Rasmussen, Erik, Henary, Haby A., Anderson, Abraham A., and Wang, Eunice S.
- Abstract
This open-label, phase 1 study evaluated the safety, pharmacokinetics, and maximum tolerated dose of AMG 232, an investigational oral, selective mouse double minute 2 homolog inhibitor in relapsed/refractory acute myeloid leukemia (AML). AMG 232 was administered orally once daily for 7 days every 2 weeks (7 on/off) at 60, 120, 240, 360, 480, or 960 mg as monotherapy (arm 1) or at 60 mg with trametinib 2 mg (arm 2). Dose-limiting toxicities (DLTs), adverse events (AEs), pharmacokinetics, clinical and pharmacodynamic response, and expression of p53 target genes were assessed. All 36 patients received AMG 232. No DLTs occurred in arm 1, and 360 mg was the highest test dose; dose escalation was halted due to gastrointestinal AEs at higher doses. One of ten patients in arm 2 had a DLT (grade 3 fatigue); 60 mg was the highest dose tested with trametinib. Common treatment-related AEs (any grade) included nausea (58%), diarrhea (56%), vomiting (33%), and decreased appetite (25%). AMG 232 exhibited linear pharmacokinetics unaffected by coadministration with trametinib. Serum macrophage inhibitor cytokine-1 and bone marrow expression of BAX, PUMA, P21, and MDM2 increased during treatment. Of 30 evaluable patients, 1 achieved complete remission, 4 had morphologic leukemia-free state, and 1 had partial remission. Four of 13 (31%) TP53-wild-type patients and 0 of 3 (0%) TP53-mutant patients were responders. AMG 232 was associated with gastrointestinal AEs at higher doses but had acceptable pharmacokinetics, on-target effects, and promising clinical activity warranting further investigation in patients with relapsed/refractory AML. This trial was registered at www.clinicaltrials.gov as #NCT02016729.
- Published
- 2019
- Full Text
- View/download PDF
41. Complex karyotype in de novo acute myeloid leukemia: typical and atypical subtypes differ molecularly and clinically
- Author
-
Mrózek, Krzysztof, Eisfeld, Ann-Kathrin, Kohlschmidt, Jessica, Carroll, Andrew J., Walker, Christopher J., Nicolet, Deedra, Blachly, James S., Bill, Marius, Papaioannou, Dimitrios, Wang, Eunice S., Uy, Geoffrey L., Kolitz, Jonathan E., Powell, Bayard L., Blum, William, Stone, Richard M., Byrd, John C., and Bloomfield, Clara D.
- Abstract
Complex karyotype (CK) with ≥ 3 abnormalities is detected in 10–12% of patients with acute myeloid leukemia (AML) and associated with poor prognosis. The most common unbalanced abnormalities found in CK result in loss of material from the 5q, 7q, and/or 17p chromosome arms. The presence of 5q, 7q, and/or 17p abnormalities denotes typical CK and their absence denotes atypical CK. Since molecular features of CK-AML are not well characterized, we investigated mutational status of 81 leukemia/cancer-associated genes in 160 clinically well-characterized patients. They included 136 patients with ≥ 3 exclusively unbalanced chromosome abnormalities, 96 of whom had a typical CK and 40 atypical CK, and 24 patients with ≥ 1 balanced abnormality in addition to ≥ 2 unbalanced ones. Patients with atypical CK-AML differed from those with typical CK-AML: they carried TP53mutations less often (P< 0.001) and more often PHF6(P= 0.008), FLT3-TKD (P= 0.02), MED12(P= 0.02), and NPM1(P= 0.02) mutations. They were younger (P=0.007), had higher WBC (P= 0.001) and percentages of marrow (P< 0.001) and blood (P= 0.006) blasts, higher complete remission rates (P= 0.02), and longer overall survival (P< 0.001), thus indicating that atypical and typical CK-AMLs constitute distinct disease subtypes. We also identified smaller patient subsets within both typical and atypical CK-AML that differed molecularly and clinically.
- Published
- 2019
- Full Text
- View/download PDF
42. Genome-wide association study identifies an acute myeloid leukemia susceptibility locus near BICRA
- Author
-
Walker, Christopher J., Oakes, Christopher C., Genutis, Luke K., Giacopelli, Brian, Liyanarachchi, Sandya, Nicolet, Deedra, Eisfeld, Ann-Kathrin, Scholz, Markus, Brock, Pamela, Kohlschmidt, Jessica, Mrózek, Krzysztof, Bill, Marius, Carroll, Andrew J., Kolitz, Jonathan E., Powell, Bayard L., Wang, Eunice S., Niederwieser, Dietger W., Stone, Richard M., Byrd, John C., Schwind, Sebastian, de la Chapelle, Albert, and Bloomfield, Clara D.
- Published
- 2019
- Full Text
- View/download PDF
43. How I treat: secondary acute myeloid leukemia
- Author
-
Green, Steven D. and Wang, Eunice S.
- Abstract
[Display omitted]
- Published
- 2024
- Full Text
- View/download PDF
44. Phase 2 study of add-on parsaclisib for patients with myelofibrosis and suboptimal response to ruxolitinib: final results
- Author
-
Yacoub, Abdulraheem, Borate, Uma, Rampal, Raajit K., Ali, Haris, Wang, Eunice S., Gerds, Aaron T., Hobbs, Gabriela, Kremyanskaya, Marina, Winton, Elliott, O’Connell, Casey, Goel, Swati, Oh, Stephen T., Schiller, Gary, McCloskey, James, Palmer, Jeanne, Holmes, Houston, Hager, Steven, Assad, Albert, Erickson-Viitanen, Susan, Zhou, Feng, and Daver, Naval
- Abstract
•Parsaclisib, a PI3Kδ inhibitor, reduced spleen volume and improved symptom scores when added to ruxolitinib for patients with myelofibrosis.•The safety and tolerability of the combination was acceptable, and daily parsaclisib dosing may provide the greatest benefit.
- Published
- 2024
- Full Text
- View/download PDF
45. Optimal Therapeutic Strategies in Relapsed/Refractory AML with Prior Exposure to Venetoclax Based Therapy
- Author
-
Bawek, Sawyer, Patel, Prutha, Wang, Katy, Attwood, Kristopher, Cronin, Tara, Fos, Melissa, Green, Steven, Sung, Pamela Jeannette, Thompson, James E, Griffiths, Elizabeth A., Wang, Eunice S, and Przespolewski, Amanda C.
- Abstract
Background:Acute myeloid leukemia (AML) is an aggressive hematological malignancy arising from immature myeloid progenitors with notoriously poor outcomes, particularly in the elderly. Venetoclax (Ven) plays an essential role in acute myeloid leukemia (AML) directed therapy as it is approved for frontline therapy in combination with azacitidine (Aza), decitabine (Dec), or low-dose cytarabine (LDAC) for patients aged 75 years and older, or those with significant comorbidities. However, venetoclax-based therapy is not curative, and the ideal salvage regimen is unclear in these patients. We performed a retrospective review that analyzed a single center's venetoclax-based regimens to identify rational subsequent lines of therapy after failing venetoclax.
- Published
- 2023
- Full Text
- View/download PDF
46. Outcomes of Extramedullary Acute Myeloid Leukemia with Gemtuzumab Ozogamicin: Real-World Experience
- Author
-
Patel, Riya, Shalaby, Khalid, Roche, Charles, Awada, Hassan, Griffiths, Elizabeth A., Thompson, James E, Green, Steven, Sung, Pamela Jeannette, Wang, Eunice S., and Przespolewski, Amanda C.
- Abstract
Introduction:
- Published
- 2023
- Full Text
- View/download PDF
47. Pivekimab Sunirine (PVEK, IMGN632), a CD123-Targeting Antibody-Drug Conjugate, in Combination with Azacitidine and Venetoclax in Patients with Newly Diagnosed Acute Myeloid Leukemia
- Author
-
Daver, Naval, Montesinos, Pau, Altman, Jessica K., Wang, Eunice S., Martinelli, Giovanni, Roboz, Gail J., Begna, Kebede, Vyas, Paresh, Lunghi, Monia, Platzbecker, Uwe, Burke, Patrick W., Walter, Roland B., Advani, Anjali S., Garciaz, Sylvain, Gastaud, Lauris, Sallman, David A, Pemmaraju, Naveen, Torres, Laura, Abaza, Yasmin, Chan, Onyee, Kantarjian, Hagop M., Oshrine, Benjamin, Du, Yining, Malcolm, Kara, and Sweet, Kendra
- Abstract
Background
- Published
- 2023
- Full Text
- View/download PDF
48. Conditioning Impacts on Regulatory T-Cells (Tregs) in the Microenvironment of AML: Observations from the Phase 1 Cynk-001-AML-001 Trial
- Author
-
Wang, Eunice S, Liu, Hongtao, Egan, Daniel N., Kilcoyne, Adrian, Stout, Bhavani, Ruggeri Barbaro, Natalia, Daly, Cherie, Kilcoyne, Siyara, Hariri, Robert, and McCloskey, James
- Abstract
Introduction:Evidence shows that NK cells can exhibit potent anti-tumor activity against acute myeloid leukemia (AML). CYNK-001 is a CD56+CD3- enriched, off-the-shelf, allogeneic natural killer (NK) cell therapy expanded from placental CD34 cells. The use of recombinant IL-2 (rhIL-2) therapy has induced regression of tumors by stimulating the activation and proliferation of NK cells. Low dose RhIL-2 has been shown to selectively expand and enhance the expression of maturation markers on the surface of NK cells, making them more potent killers in vitro and in vivo. CYNK-001-AML-001 is a Phase I multi-dose study evaluating the safety, tolerability, and persistence of CYNK-001 with or without rhIL-2 in adults with primary or secondary acute myeloid leukemia (AML) in morphologic complete remission (CR) with minimal residual disease (MRD) or relapsed/refractory (R/R) AML
- Published
- 2023
- Full Text
- View/download PDF
49. Ponatinib Versus Imatinib in Patients with Newly Diagnosed Ph+ ALL: Subgroup Analysis of the Phase 3 Phallcon Study
- Author
-
Aldoss, Ibrahim, Ribera, Josep-Maria, Kantarjian, Hagop, Montesinos, Pau, Leonard, Jessica T., Gomez-Almaguer, David, Baer, Maria R., Gambacorti-Passerini, Carlo, McCloskey, James, Minami, Yosuke, Papayannidis, Cristina, Rocha, Vanderson, Rousselot, Philippe, Vachhani, Pankit, Wang, Eunice S., Hennessy, Meliessa, Patel, Niti, Vorog, Alexander, Wang, Bingxia, and Jabbour, Elias
- Abstract
Background:BCR::ABL1 tyrosine kinase inhibitors (TKIs) in combination with chemotherapy or steroids remain the standard of care in patients with newly diagnosed Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Most patients treated with first- or second-generation TKIs eventually experience disease progression due to treatment resistance. Ponatinib is the only currently approved pan-BCR::ABL1 inhibitory TKI that potently inhibits wild-type and single resistance mutation variants of BCR::ABL1, including T315I. Multiple studies have shown promising minimal residual disease (MRD) negativity (neg) rates and survival outcomes with ponatinib in combination with chemotherapy or chemotherapy-free regimens. PhALLCON (NCT03589326) is the first randomized study comparing frontline TKIs in adults with Ph+ ALL. The primary endpoint for PhALLCON was met, with a clinically significantly higher MRD-neg complete remission (CR) rate at end of induction (EOI) with ponatinib vs imatinib (34.4% vs 16.7%; risk difference: 0.18 [95% confidence interval (CI): 0.06‒0.29]; P=0.0021) and a manageable safety profile comparable with imatinib. Here we report subgroup efficacy analyses from PhALLCON.
- Published
- 2023
- Full Text
- View/download PDF
50. Factors Impacting Response to Olutasidenib in Patients with mIDH1 Acute Myeloid Leukemia
- Author
-
Wang, Eunice S., Jonas, Brian A., Watts, Justin M., Jurcic, Joseph, Ferrell, P. Brent, Dinner, Shira N., Yang, Jay, Seiter, Karen, Mims, Alice, Donnellan, William B., Blum, William, and Cortes, Jorge
- Abstract
Introduction. Olutasidenib is an FDA-approved, targeted therapy for IDH1-mutated (mIDH1) AML. Among 147 patients with relapsed/refractory mIDH1 AML, 51 (35%) achieved a complete response (CR) or CR with partial hematologic recovery (CRh) after treatment with olutasidenib, with a duration of response of 25.9 months. We conducted sub-analyses to better understand which factors might be predictive of response to therapy.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.