1,621 results on '"Levis, MJ"'
Search Results
2. Smith CC, Levis MJ, Perl AE, et al. Molecular profile of FLT3-mutated relapsed/refractory patients with AML in the phase 3 ADMIRAL study of gilteritinib. Blood Adv. 2022;6(7):2144-2155.
- Published
- 2022
- Full Text
- View/download PDF
3. Smith CC, Levis MJ, Perl AE, et al. Molecular profile of FLT3-mutated relapsed/refractory patients with AML in the phase 3 ADMIRAL study of gilteritinib. Blood Adv. 2022;6(7):2144-2155
- Subjects
Leukemia, Myeloid, Acute ,Aniline Compounds ,fms-Like Tyrosine Kinase 3 ,Clinical Trials, Phase III as Topic ,Pyrazines ,Humans ,Neoplasm Recurrence, Local - Published
- 2022
4. Targeting rapid TKI-induced AXL upregulation overcomes adaptive ERK reactivation and exerts antileukemic effects in FLT3/ITD acute myeloid leukemia.
- Author
-
Seale TS, Li L, Bruner JK, Chou M, Nguyen B, Seo J, Zhu R, Levis MJ, Pratilas CA, and Small D
- Abstract
Acute myeloid leukemia (AML) patients with the FMS-related receptor tyrosine kinase 3 internal tandem duplication (FLT3/ITD) mutation have a poorer prognosis, and treatment with FLT3 tyrosine kinase inhibitors (TKIs) has been hindered by resistance mechanisms. One such mechanism is known as adaptive resistance, in which downstream signaling pathways are reactivated after initial inhibition. Past work has shown that FLT3/ITD cells undergo adaptive resistance through the reactivation of extracellular signal-regulated kinase (ERK) signaling within 24 h of sustained FLT3 inhibition. We investigated the mechanism(s) responsible for this ERK reactivation and hypothesized that targeting tyrosine-protein kinase receptor UFO (AXL), another receptor tyrosine kinase that has been implicated in cancer resistance, may overcome the adaptive ERK reactivation. Experiments revealed that AXL is upregulated and activated in FLT3/ITD cell lines mere hours after commencing TKI treatment. AXL inhibition combined with FLT3 inhibition to decrease the ERK signal rebound and to exert greater anti-leukemia effects than with either treatment alone. Finally, we observed that TKI-induced AXL upregulation occurs in patient samples, and combined inhibition of both AXL and FLT3 increased efficacy in our in vivo models. Taken together, these data suggest that AXL plays a role in adaptive resistance in FLT3/ITD AML and that combined AXL and FLT3 inhibition might improve FLT3/ITD AML patient outcomes., (© 2024 The Author(s). Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
5. Outcome of adult acute myeloid leukemia patients with extramedullary disease and treatment with venetoclax/ hypomethylating agents.
- Author
-
Kayser S, Sanber K, Marconi G, Mattei A, Luskin MR, Kelkar A, Cerrano M, Kristensen DT, Roug AS, Sartor C, Giglio F, Riva M, Rizzo L, Saraceni F, Guerzoni S, Lessi F, Borlenghi E, Levis MJ, Schlenk RF, Jain T, and Papayannidis C
- Subjects
- Humans, Middle Aged, Aged, Adult, Male, Female, Aged, 80 and over, Young Adult, Treatment Outcome, Hematopoietic Stem Cell Transplantation, Remission Induction, DNA Methylation, Sulfonamides therapeutic use, Sulfonamides administration & dosage, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
We evaluated response to venetoclax/hypomethylating agents (HMA) in 46 patients with acute myeloid leukemia (AML) characterized by extramedullary disease. The median age of these patients was 65 years (range, 19-81). The patients had a median of two sites of extramedullary disease (range, 1-5) and 35 (76%) had concurrent bone marrow involvement. Twenty (43%) patients had high-risk genetic features according to the European LeukemiaNet 2022 classification. Twenty-nine (63%) had relapsed or were refractory after intensive chemotherapy, including 13 (28%) who had undergone prior allogeneic hematopoietic cell transplantation. Patients received a median of two cycles of venetoclax/HMA (range, 1-31). Twenty (43%) patients achieved complete remission (CR) or CR with incomplete hematologic recovery (CRi) after venetoclax/HMA and five (11%) achieved a partial remission (PR). Six patients were subsequently consolidated with allogeneic hematopoietic cell transplantation (CR/CRi, N=4; PR, N=2). The median follow-up was 49.1 months (95% confidence interval [95% CI]: 26.1 months - not reached) and the median overall survival was 6.4 months (95% CI: 5.1-11 months). One-year and 2-year overall survival rates were 29.3% (95% CI: 18.6-46.2%) and 12.3% (95% CI: 5.5-27.6%), respectively. Age, with a cutoff of 60 years, did not have an impact on overall survival (P=0.90). Relapse occurred in 12 of 20 (60%) patients who achieved CR/CRi after venetoclax/ HMA treatment. Of those, all except one succumbed to their disease. Six (30%) patients were in CR/CRi at last follow-up and two (10%) died in CR. In our cohort of patients with AML with extramedullary disease with high-risk features, treatment with venetoclax/HMA resulted in an encouraging overall response rate of 54% with a CR/CRi rate of 43.5%. However, venetoclax/ HMA alone may not be effective in maintaining disease control.
- Published
- 2025
- Full Text
- View/download PDF
6. Measurable residual disease and posttransplantation gilteritinib maintenance for patients with FLT3-ITD-mutated AML.
- Author
-
Levis MJ, Hamadani M, Logan BR, Jones RJ, Singh AK, Litzow MR, Wingard JR, Papadopoulos EB, Perl AE, Soiffer RJ, Ustun C, Ueda Oshima M, Uy GL, Waller EK, Vasu S, Solh MM, Mishra A, Muffly L, Kim HJ, Stelljes M, Najima Y, Onozawa M, Thomson KJ, Nagler A, Wei AH, Marcucci G, Chen C, Hasabou N, Rosales M, Hill JE, Gill SC, Nuthethi R, King D, Mendizabal AM, Devine SM, Horowitz MM, and Chen YB
- Abstract
BMT CTN 1506 ("MORPHO"; NCT02997202) was a randomized phase 3 study of gilteritinib compared to placebo as maintenance therapy after hematopoietic stem cell transplantation (HCT) for patients with FLT3-ITD-mutated acute myeloid leukemia (AML). A key secondary endpoint was to determine the impact on survival of pre- and/or post-HCT measurable residual disease (MRD), as determined using a highly sensitive assay for FLT3-ITD mutations. Generally, gilteritinib maintenance therapy was associated with improved relapse-free survival (RFS) for participants with detectable peri-HCT MRD, whereas no benefit was evident for those lacking detectable MRD. We conducted a post-hoc analysis of the data and found that the level of MRD detected with this approach correlated remarkably with RFS and relapse risk, and that MRD detectable at any level negatively impacted RFS. In the placebo arm, 42.2% of participants with detectable FLT3-ITD MRD relapsed compared to 13.4% of those without detectable MRD. We found that 14.8% of participants had multiple FLT3-ITD clones detected as MRD and had worse survival irrespective of treatment arm. Finally, we examined the kinetics of FLT3-ITD clonal relapse or eradication and found that participants on the placebo arm with detectable MRD relapsed rapidly after HCT, often within a few weeks. MRD-positive participants on the gilteritinib arm relapsed either with FLT3 wild type clones (as assessed by capillary electrophoresis), after cessation of gilteritinib with persistent MRD, or on progression of multi-clonal disease. These data demonstrate the potential of using FLT3-ITD MRD to guide therapy with gilteritinib for this subtype of AML., (Copyright © 2025 American Society of Hematology.)
- Published
- 2025
- Full Text
- View/download PDF
7. Reply to S. Fuji.
- Author
-
Levis MJ, Hamadani M, Horowitz MM, and Chen YB
- Published
- 2024
- Full Text
- View/download PDF
8. Health-related quality of life with gilteritinib vs placebo posttransplant for FLT3-ITD+ acute myeloid leukemia.
- Author
-
Hamilton BK, Pandya BJ, Ivanescu C, Elsouda D, Hamadani M, Chen YB, Levis MJ, Ueda Oshima M, Litzow MR, Soiffer RJ, Ustun C, Perl AE, Singh AK, Geller N, Hasabou N, Rosales M, Cella D, Corredoira L, Pestana C, Horowitz MM, and Logan B
- Subjects
- Humans, Middle Aged, Female, Male, Adult, Aged, Quality of Life, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute therapy, fms-Like Tyrosine Kinase 3 genetics, Pyrazines therapeutic use, Aniline Compounds therapeutic use, Hematopoietic Stem Cell Transplantation methods
- Abstract
Abstract: The Blood and Marrow Transplant (BMT) Clinical Trials Network conducted a phase 3 randomized trial comparing gilteritinib with placebo after allogeneic hematopoietic cell transplantation (HCT) for FLT3-ITD+ acute myeloid leukemia (AML). The primary analysis demonstrated no statistically significant difference in relapse-free survival (RFS); however, patients with FLT3-ITD measurable residual disease (MRD) peri-HCT had significantly longer RFS with gilteritinib. This analysis investigates the effect of post-HCT gilteritinib vs placebo on health-related quality of life (HRQOL). HRQOL was measured with Functional Assessment of Cancer Therapy-Bone Marrow Transplant (FACT-BMT), FACT-Leukemia (FACT-Leu), and EuroQOL-5 Dimensions (EQ-5D-5L) at post-HCT randomization; day 29; months 3, 6, 12, 18, 24; and/or end of therapy. HRQOL and clinically meaningful differences were summarized using descriptive statistics and compared using mixed model repeated measures to evaluate longitudinal change from baseline and stratified Cox model to evaluate time to improvement. HRQOL completion rate was acceptable (>70%) across all time points and measures. There were no differences in HRQOL scores at any time point between cohorts. Clinically meaningful and time to improvement in HRQOL were similar in both arms. Despite higher treatment-emergent adverse effects with gilteritinib, response to the question of being "bothered by side effects of treatment" did not differ between groups. Subgroup analysis of MRD-positive and negative patients demonstrated no differences in HRQOL between arms. For patients with FLT3-ITD+ AML undergoing HCT, gilteritinib maintenance was not associated with any difference in HRQOL or patient-reported impact of side effects. This trial was registered at www.ClinicalTrials.gov as #NCT02997202., (Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution.)
- Published
- 2024
- Full Text
- View/download PDF
9. Complete morphologic response to gilteritinib in ALK-rearranged acute myeloid leukemia.
- Author
-
Adashek JJ, Brodsky M, and Levis MJ
- Abstract
The cytogenetic abnormality inv(2)(p23q13) in acute myeloid leukemia (AML) results in a fusion of RANBP2 with ALK. This fusion makes ALK constitutively active and acts as a driver for the proliferation of AML cell lines. Gilteritinib, a FLT3 inhibitor approved in AML, also can inhibit ALK among other receptor tyrosine kinases. A 75-year-old-woman with a history of essential thrombocythemia (ET) and a presumed germline DDX41 mutation developed ALK-fusion positive AML and despite standard therapies was transfusion-dependent and globally declining. The patient has been on gilteritinib with an ongoing response of more than one year with near normal blood counts and no evidence of AML. The fact that she was found to harbor a presumed germline DDX41 alteration may account for why she developed, and yet survived, two myeloid neoplasms (ET and AML). Additionally, this demonstrates that gilteritinib is clinically active as an ALK inhibitor, and could be considered for use in any AML patient presenting with an inv(2(p23q13)) translocation. Finally, it is an example of using a disease-agnostic, precision medicine approach to arrive at a beneficial treatment., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. 1q jumping translocation as a biomarker in myeloid malignancy: frequently mutated genes associated with bad prognosis and low survival.
- Author
-
Halper-Stromberg E, Stinnett V, Morsberger L, Pallavajjala A, Levis MJ, DeZern AE, Lei M, Phan B, Xian RR, Gocke CD, Tang G, and Zou YS
- Abstract
1q jumping translocation (JT) is rare and its molecular profiles in myeloid malignancies are not well-known. This study evaluated gene mutations in 1q-JT cohorts (0.38%) from hematological malignant specimens that underwent genetic analysis at the Johns Hopkins Hospital (n = 11,908) and the MD Anderson Cancer Center. 1q-JT had frequent mutations in eleven genes, most of which are associated with worse prognosis. BCOR mutations significantly co-occurred with others. Patients tended to have mutations in DNA-repair, spliceosome, and epigenetic modification pathways, though genes utilized within each of these pathways were not randomly distributed. Multi-, albeit overlapping, pathway interruptions tended to manifest in mutations of two gene sets. One gene set consisted of SF3B1 (spliceosome) and TET2 (epigenetic modification), while the other consisted of STAG2 (DNA repair), SRSF2, U2AF (spliceosome), ASXL1, KMT2D (epigenetic modification), BCOR, and GATA2 (transcription factors). An "intermediate" JT-like rearrangement may represent an early sign of occurring 1q-JT. Treatments (hypomethylating agents) and unique structures of the short arms of acrocentric chromosomes may contribute to 1q-JT formation in myeloid malignancies. The median overall survival after identification of a JT was 10 months (95% confidence interval, 5-15 months). Our cohort represents the largest number of myeloid malignancies from multi-centers with before and after the 1q-JT event analyzed to date. Overall, this study identified specific molecular profiles that are associated with 1q-JT in myeloid malignancies. 1q-JT could serve as a poor prognosis biomarker in myeloid malignancies, which could be important in making well-informed clinical decisions and treatment strategies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. Sorafenib plus intensive chemotherapy in newly diagnosed FLT3-ITD AML:a randomized, placebo-controlled study by the ALLG.
- Author
-
Loo, S, Roberts, AW, Anstee, NS, Kennedy, GA, He, SZ-X, Schwarer, AP, Enjeti, AK, D'Rozario, J, Marlton, P, Bilmon, I, Taper, JM, Cull, G, Tiley, C, Verner, E, Hahn, U, Hiwase, DK, Iland, HJ, Murphy, NE, Ramanathan, S, Reynolds, J, Ong, DM, Tiong, IS, Wall, M, Murray, M, Rawling, T, Leadbetter, J, Rowley, L, Latimer, M, Yuen, SLS, Ting, SB, Fong, CY, Morris, KL, Bajel, A, Seymour, JF, Levis, MJ, Wei, AH, Loo, S, Roberts, AW, Anstee, NS, Kennedy, GA, He, SZ-X, Schwarer, AP, Enjeti, AK, D'Rozario, J, Marlton, P, Bilmon, I, Taper, JM, Cull, G, Tiley, C, Verner, E, Hahn, U, Hiwase, DK, Iland, HJ, Murphy, NE, Ramanathan, S, Reynolds, J, Ong, DM, Tiong, IS, Wall, M, Murray, M, Rawling, T, Leadbetter, J, Rowley, L, Latimer, M, Yuen, SLS, Ting, SB, Fong, CY, Morris, KL, Bajel, A, Seymour, JF, Levis, MJ, and Wei, AH
- Abstract
Sorafenib maintenance improves outcome after hematopoietic cell transplant (HCT) for patients with FLT3-ITD acute myeloid leukemia (AML). Although promising outcomes have been reported for sorafenib plus intensive chemotherapy, randomized data are limited. This placebo-controlled, phase 2 study (ACTRN12611001112954) randomized 102 patients 18-65 years (2:1) to sorafenib vs placebo (days 4-10) combined with intensive induction; idarubicin 12mg/m2 days 1-3 plus cytarabine 1.5g/m2 twice daily on days 1,3,5,7 (18-55 years) or 100mg/m2 days 1-7 (56-65 years), consolidation therapy, followed by maintenance treatment for 12 months (post-HCT excluded) in newly diagnosed FLT3-ITD AML. Four patients were excluded from modified intention-to-treat final analysis (3 not dosed and 1 later found to be FLT3-ITD negative). Rates of complete remission (CR)/CR with incomplete hematologic recovery (CR/CRi) were high in both arms (sorafenib 78%/9%, placebo 70%/24%). With 49.1 months median follow-up, the primary endpoint of event-free survival (EFS) was not improved by sorafenib (2-year EFS 47.9% vs 45.4%)(hazard ratio [HR] 0.87;95% confidence interval [CI] 0.51-1.51, p=0.61). Two-year overall survival (OS) was 67% in the sorafenib arm and 58% in the placebo arm (HR 0.76; 95% CI 0.42-1.39). For patients transplanted in first remission, 2-year OS was 84% and 67% in the sorafenib and placebo arms, respectively (HR 0.45;95% CI 0.18-1.12, p=0.08). In exploratory analyses, FLT3-ITD measurable residual disease negative status (<0.001%) post-induction was associated with improved 2-year OS (83% vs 60%) (HR 0.4;95% CI 0.17-0.93, p=0.028). In conclusion, routine use of pre-transplant sorafenib plus chemotherapy in unselected patients with FLT3-ITD AML is not supported by this study.
- Published
- 2023
12. Gilteritinib as Post-Transplant Maintenance for AML With Internal Tandem Duplication Mutation of FLT3 .
- Author
-
Levis MJ, Hamadani M, Logan B, Jones RJ, Singh AK, Litzow M, Wingard JR, Papadopoulos EB, Perl AE, Soiffer RJ, Ustun C, Ueda Oshima M, Uy GL, Waller EK, Vasu S, Solh M, Mishra A, Muffly L, Kim HJ, Mikesch JH, Najima Y, Onozawa M, Thomson K, Nagler A, Wei AH, Marcucci G, Geller NL, Hasabou N, Delgado D, Rosales M, Hill J, Gill SC, Nuthethi R, King D, Wittsack H, Mendizabal A, Devine SM, Horowitz MM, and Chen YB
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Aged, Tandem Repeat Sequences, Young Adult, Neoplasm, Residual, Protein Kinase Inhibitors therapeutic use, Maintenance Chemotherapy, Gene Duplication, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute mortality, Pyrazines therapeutic use, Aniline Compounds therapeutic use, Hematopoietic Stem Cell Transplantation, Mutation
- Abstract
Purpose: Allogeneic hematopoietic cell transplantation (HCT) improves outcomes for patients with AML harboring an internal tandem duplication mutation of FLT3 ( FLT3-ITD ) AML. These patients are routinely treated with a FLT3 inhibitor after HCT, but there is limited evidence to support this. Accordingly, we conducted a randomized trial of post-HCT maintenance with the FLT3 inhibitor gilteritinib (ClinicalTrials.gov identifier: NCT02997202) to determine if all such patients benefit or if detection of measurable residual disease (MRD) could identify those who might benefit., Methods: Adults with FLT3-ITD AML in first remission underwent HCT and were randomly assigned to placebo or 120 mg once daily gilteritinib for 24 months after HCT. The primary end point was relapse-free survival (RFS). Secondary end points included overall survival (OS) and the effect of MRD pre- and post-HCT on RFS and OS., Results: Three hundred fifty-six participants were randomly assigned post-HCT to receive gilteritinib or placebo. Although RFS was higher in the gilteritinib arm, the difference was not statistically significant (hazard ratio [HR], 0.679 [95% CI, 0.459 to 1.005]; two-sided P = .0518). However, 50.5% of participants had MRD detectable pre- or post-HCT, and, in a prespecified subgroup analysis, gilteritinib was beneficial in this population (HR, 0.515 [95% CI, 0.316 to 0.838]; P = .0065). Those without detectable MRD showed no benefit (HR, 1.213 [95% CI, 0.616 to 2.387]; P = .575)., Conclusion: Although the overall improvement in RFS was not statistically significant, RFS was higher for participants with detectable FLT3-ITD MRD pre- or post-HCT who received gilteritinib treatment. To our knowledge, these data are among the first to support the effectiveness of MRD-based post-HCT therapy.
- Published
- 2024
- Full Text
- View/download PDF
13. Impact of inotuzumab ozogamicin on outcome in relapsed or refractory acute B-cell lymphoblastic leukemia patients prior to allogeneic hematopoietic stem cell transplantation and risk of sinusoidal obstruction syndrome/venous occlusive disease.
- Author
-
Kayser S, Sartor C, Giglio F, Bruno A, Webster J, Chiusolo P, Saraceni F, Guerzoni S, Pochintesta L, Borlenghi E, Marconi G, Zacheo I, Cerrano M, Salutari P, Restuccia F, Abbenante M, Levis MJ, Schlenk RF, and Papayannidis C
- Subjects
- Humans, Adult, Middle Aged, Male, Female, Adolescent, Aged, Young Adult, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Treatment Outcome, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological administration & dosage, Recurrence, Transplantation Conditioning methods, Transplantation Conditioning adverse effects, Inotuzumab Ozogamicin therapeutic use, Hematopoietic Stem Cell Transplantation adverse effects, Hepatic Veno-Occlusive Disease etiology, Hepatic Veno-Occlusive Disease diagnosis, Hepatic Veno-Occlusive Disease chemically induced, Transplantation, Homologous
- Abstract
We evaluated 58 patients with relapsed or refractory (r/r) acute B-lymphoblastic leukemia (B-ALL; median age 42.5 years; range, 16-69 years), treated with inotuzumab ozogamicin (INO) between 2016-2022 and who received an allogeneic hematopoietic stem cell transplantation (allo-HCT) consecutively. Forty-seven (81%) of the 58 patients were heavily pretreated receiving intensive chemotherapy +/- tyrosine kinase inhibitor, blinatumomab in 24 (41%) and allo-HCT at first-line in 11 (19%) patients. Complete remission rate prior to allo-HCT was 84%. Median follow-up was 30.5 months and median overall survival (OS) measured from start of INO was 11.2 months. One- and 2-year OS rates were 50% (95% confidence interval [CI]: 38.4-56.1) and 36.7% (95% CI: 25.5-52.9), respectively. Sinusoidal obstruction syndrome/venous occlusive disease (SOS/ VOD) after allo-HCT occurred in 17 (29%) patients. Of those, nine (53%) patients died due to SOS/VOD and multi-organ failure. Two had received >2 INO cycles (3 cycles, 5 cycles, N=1, each), all others ≤2 INO cycles prior to allo-HCT. Logistic regression analysis revealed conditioning with double alkylators (P=0.038) and allo-HCT during first-line therapy (P=0.050) as significant risk factors for SOS/VOD and in trend allo-HCT ≤60 days from last INO application (P=0.07), whereas number of INO cycles before allo-HCT and time between last INO application and allo-HCT were not significant. Relapse/progressive disease occurred in 20 (34%) patients. Of those, five (25%) patients are still alive, whereas 15 succumbed of their disease. Treatment with INO seems to be an effective approach with successful bridge-to-transplant. However, risk of SOS/VOD is high, necessitating continuous monitoring and recognition of SOS/VOD risk factors.
- Published
- 2024
- Full Text
- View/download PDF
14. Outcome heterogeneity of TP53 -mutated myeloid neoplasms and the role of allogeneic hematopoietic cell transplantation.
- Author
-
Pasca S, Haldar SD, Ambinder A, Webster JA, Jain T, Dalton WB, Prince GT, Ghiaur G, DeZern AE, Gojo I, Smith BD, Karantanos T, Schulz C, Stokvis K, Levis MJ, Jones RJ, and Gondek LP
- Subjects
- Humans, Tumor Suppressor Protein p53 genetics, Hematopoietic Stem Cell Transplantation, Neoplasms
- Published
- 2024
- Full Text
- View/download PDF
15. Sorafenib plus intensive chemotherapy in newly diagnosed FLT3-ITD AML: a randomized, placebo-controlled study by the ALLG.
- Author
-
Loo S, Roberts AW, Anstee NS, Kennedy GA, He S, Schwarer AP, Enjeti AK, D'Rozario J, Marlton P, Bilmon IA, Taper J, Cull G, Tiley C, Verner E, Hahn U, Hiwase DK, Iland HJ, Murphy N, Ramanathan S, Reynolds J, Ong DM, Tiong IS, Wall M, Murray M, Rawling T, Leadbetter J, Rowley L, Latimer M, Yuen S, Ting SB, Fong CY, Morris K, Bajel A, Seymour JF, Levis MJ, and Wei AH
- Subjects
- Humans, Sorafenib, fms-Like Tyrosine Kinase 3 genetics, Retrospective Studies, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Sorafenib maintenance improves outcomes after hematopoietic cell transplant (HCT) for patients with FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) acute myeloid leukemia (AML). Although promising outcomes have been reported for sorafenib plus intensive chemotherapy, randomized data are limited. This placebo-controlled, phase 2 study (ACTRN12611001112954) randomized 102 patients (aged 18-65 years) 2:1 to sorafenib vs placebo (days 4-10) combined with intensive induction: idarubicin 12 mg/m2 on days 1 to 3 plus either cytarabine 1.5 g/m2 twice daily on days 1, 3, 5, and 7 (18-55 years) or 100 mg/m2 on days 1 to 7 (56-65 years), followed by consolidation and maintenance therapy for 12 months (post-HCT excluded) in newly diagnosed patients with FLT3-ITD AML. Four patients were excluded in a modified intention-to-treat final analysis (3 not commencing therapy and 1 was FLT3-ITD negative). Rates of complete remission (CR)/CR with incomplete hematologic recovery were high in both arms (sorafenib, 78%/9%; placebo, 70%/24%). With 49.1-months median follow-up, the primary end point of event-free survival (EFS) was not improved by sorafenib (2-year EFS 47.9% vs 45.4%; hazard ratio [HR], 0.87; 95% confidence interval [CI], 0.51-1.51; P = .61). Two-year overall survival (OS) was 67% in the sorafenib arm and 58% in the placebo arm (HR, 0.76; 95% CI, 0.42-1.39). For patients who received HCT in first remission, the 2-year OS rates were 84% and 67% in the sorafenib and placebo arms, respectively (HR, 0.45; 95% CI, 0.18-1.12; P = .08). In exploratory analyses, FLT3-ITD measurable residual disease (MRD) negative status (<0.001%) after induction was associated with improved 2-year OS (83% vs 60%; HR, 0.4; 95% CI, 0.17-0.93; P = .028). In conclusion, routine use of pretransplant sorafenib plus chemotherapy in unselected patients with FLT3-ITD AML is not supported by this study., (© 2023 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
- 2023
- Full Text
- View/download PDF
16. Clinical outcomes in patients with relapsed/refractory FLT3-mutated acute myeloid leukemia treated with gilteritinib who received prior midostaurin or sorafenib
- Author
-
Perl, AE, Perl, AE, Hosono, N, Montesinos, P, Podoltsev, N, Martinelli, G, Panoskaltsis, N, Recher, C, Smith, CC, Levis, MJ, Strickland, S, Röllig, C, Groß-Langenhoff, M, Chou, WC, Lee, JH, Yokoyama, H, Hasabou, N, Lu, Q, Tiu, RV, Altman, JK, Perl, AE, Perl, AE, Hosono, N, Montesinos, P, Podoltsev, N, Martinelli, G, Panoskaltsis, N, Recher, C, Smith, CC, Levis, MJ, Strickland, S, Röllig, C, Groß-Langenhoff, M, Chou, WC, Lee, JH, Yokoyama, H, Hasabou, N, Lu, Q, Tiu, RV, and Altman, JK
- Abstract
The fms-like tyrosine kinase 3 (FLT3) inhibitor gilteritinib is indicated for relapsed or refractory (R/R) FLT3-mutated acute myeloid leukemia (AML), based on its observed superior response and survival outcomes compared with salvage chemotherapy (SC). Frontline use of FLT3 tyrosine kinase inhibitors (TKIs) midostaurin and sorafenib may contribute to cross-resistance to single-agent gilteritinib in the R/R AML setting but has not been well characterized. To clarify the potential clinical impact of prior TKI use, we retrospectively compared clinical outcomes in patients with R/R FLT3-mutated AML in the CHRYSALIS and ADMIRAL trials who received prior midostaurin or sorafenib against those without prior FLT3 TKI exposure. Similarly high rates of composite complete remission (CRc) were observed in patients who received a FLT3 TKI before gilteritinib (CHRYSALIS, 42%; ADMIRAL, 52%) and those without prior FLT3 TKI therapy (CHRYSALIS, 43%; ADMIRAL, 55%). Among patients who received a prior FLT3 TKI in ADMIRAL, a higher CRc rate (52%) and trend toward longer median overall survival was observed in the gilteritinib arm versus the SC arm (CRc = 20%; overall survival, 5.1 months; HR = 0.602; 95% CI: 0.299, 1.210). Remission duration was shorter with prior FLT3 TKI exposure. These findings support gilteritinib for FLT3-mutated R/R AML after prior sorafenib or midostaurin.
- Published
- 2022
17. Gilteritinib in Combination With Induction and Consolidation Chemotherapy and as Maintenance Therapy: A Phase IB Study in Patients With Newly Diagnosed AML.
- Author
-
Pratz KW, Cherry M, Altman JK, Cooper BW, Podoltsev NA, Cruz JC, Lin TL, Schiller GJ, Jurcic JG, Asch A, Wu R, Hill JE, Gill SC, James AJ, Rich ES, Hasabou N, Perl AE, and Levis MJ
- Subjects
- Adult, Humans, Neoplasm Recurrence, Local drug therapy, Idarubicin, Protein Kinase Inhibitors therapeutic use, fms-Like Tyrosine Kinase 3 genetics, Mutation, Consolidation Chemotherapy, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Purpose: Gilteritinib is a type 1 FLT3 inhibitor active as monotherapy for relapsed or refractory FLT3 -mutated AML. We investigated the safety, tolerability, and efficacy of gilteritinib incorporated into intensive induction and consolidation chemotherapy, and as maintenance therapy for adult patients with newly diagnosed, non-favorable-risk AML., Methods: In this phase IB study (2215-CL-0103; ClinicalTrials.gov identifier: NCT02236013), 103 participants were screened and 80 were allocated to treatment. The study was divided into four parts: dose escalation, dose expansion, investigation of alternate anthracycline and gilteritinib schedule, and continuous gilteritinib during consolidation., Results: After dose escalation, 120 mg gilteritinib once daily was chosen for further study. There were 58 participants evaluable for response at this dose, 36 of whom harbored FLT3 mutations. For participants with FLT3 -mutated AML, the composite complete response (CRc) rate was 89% (83% were conventional complete responses), all achieved after a single induction cycle. The median overall survival time was 46.1 months. Gilteritinib was well-tolerated in this context although the median time to count recovery during induction was approximately 40 days. Longer time-to-count recovery was associated with higher trough levels of gilteritinib, which, in turn, were associated with azole use. The recommended regimen is gilteritinib at a dose of 120 mg once daily from days 4 to 17 or 8 to 21 of a 7 + 3 induction with either idarubicin or daunorubicin and from day 1 continuously with high-dose cytarabine consolidation. Maintenance therapy with gilteritinib was well-tolerated., Conclusion: These results demonstrated the safety and tolerability of gilteritinib incorporated into an induction and consolidation chemotherapy regimen, and as single-agent maintenance therapy for patients with newly diagnosed FLT3 -mutant AML. The data herein provide an important framework for the design of randomized trials comparing gilteritinib with other FLT3 inhibitors.
- Published
- 2023
- Full Text
- View/download PDF
18. Differentiation syndrome with lower-intensity treatments for acute myeloid leukemia.
- Author
-
Fathi AT, Stein EM, DiNardo CD, Levis MJ, Montesinos P, and de Botton S
- Subjects
- Acute Kidney Injury chemically induced, Adrenal Cortex Hormones therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Arsenic Trioxide adverse effects, Arsenic Trioxide pharmacology, Arsenic Trioxide therapeutic use, Cell Differentiation drug effects, Clinical Trials as Topic, Cytokine Release Syndrome diagnosis, Cytokine Release Syndrome drug therapy, Cytokine Release Syndrome mortality, Edema chemically induced, Enzyme Inhibitors adverse effects, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Epigenesis, Genetic drug effects, Fever chemically induced, Humans, Hypotension chemically induced, Isocitrate Dehydrogenase antagonists & inhibitors, Leukemia, Myeloid, Acute complications, Molecular Targeted Therapy adverse effects, Neoplasm Proteins antagonists & inhibitors, Pleural Effusion chemically induced, Respiration Disorders chemically induced, Tretinoin adverse effects, Tretinoin pharmacology, Tretinoin therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, Antineoplastic Agents adverse effects, Cytokine Release Syndrome chemically induced, Leukemia, Myeloid, Acute drug therapy, Myelopoiesis drug effects
- Abstract
Differentiation Syndrome (DS) has been identified in a subset of patients undergoing treatment with novel classes of differentiating therapies for acute myeloid leukemia (AML) such as IDH and FLT3 inhibitors. While DS is a well-known treatment-related complication in acute promyelocytic leukemia (APL), efforts are still ongoing to standardize diagnostic and treatment parameters for DS in AML. Though the rates of incidence vary, many of the signs and symptoms of DS are common between APL and AML. So, DS can lead to fatal complications in AML, but prompt management is usually effective and rarely necessitates interruption or discontinuation of AML therapy., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
19. A Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FMS-like Tyrosine Kinase (FLT3) Mutation
- Published
- 2024
20. Co-occurring mutations in ASXL1, SRSF2, and SETBP1 define a subset of myelodysplastic/ myeloproliferative neoplasm with neutrophilia.
- Author
-
Jain T, Ware AD, Dalton WB, Pasca S, Tsai HL, Gocke CD, Gondek LP, Xian RR, Borowitz MJ, and Levis MJ
- Subjects
- Male, Female, Humans, Leukocytosis, Hyperplasia, Mutation, Serine-Arginine Splicing Factors genetics, Repressor Proteins genetics, Carrier Proteins genetics, Nuclear Proteins genetics, Myelodysplastic-Myeloproliferative Diseases genetics, Myelodysplastic-Myeloproliferative Diseases pathology, Leukemia, Myeloid, Acute genetics
- Abstract
Identification of genomic signatures with consistent clinicopathological features in myelodysplastic/myeloproliferative neoplasm (MDS/MPN) is critical for improved diagnosis, elucidation of biology, inclusion in clinical trials, and development of therapies. We describe clinical and pathological features with co-existence of mutations in ASXL1 (missense or nonsense), SRSF2, and SKI homologous region of SETBP1, in 18 patients. Median age was 68 years with a male predominance (83%). Leukocytosis and neutrophilia were common at presentation. Marrow features included hypercellularity, granulocytic hyperplasia with megakaryocytic atypia, while the majority had myeloid hyperplasia and/or erythroid hypoplasia, myeloid dysplasia, and aberrant CD7 expression on blasts. Mutations in growth signaling pathways (RAS or JAK2) were noted at diagnosis or acquired during the disease course in 83% of patients. Two patients progressed upon acquisition of FLT3-TKD (acute myeloid leukemia) or KIT (aggressive systemic mastocytosis) mutations. The prognosis is poor with only two long-term survivors, thus far, who underwent blood or marrow transplantation. We propose that the presence of co-occurring ASXL1, SRSF2, and SETBP1 mutations can be diagnostic of a subtype of MDS/MPN with neutrophilia if clinical and morphological findings align. Our report underscores the association between genotype and phenotype within MDS/MPN and that genomic signatures should guide categorization of these entities., Competing Interests: Declaration of Competing Interest Jain: Institutional research support from CTI Biopharma, Kartos Therapeutics, Incyte; Advisory board participation with Care Dx, Bristol Myers Squibb, Incyte, Abbvie, CTI, Kite, Cogent Biosciences, Blueprint Medicine, Telios pharma, Protagonist therapeutics. Levis: Research funding from Astellas, FujiFilm; Consultancy for Abbvie, Amgen, Bristol-Myers Squibb, Daiichi-Sankyo, Jazz, Menarini, Syndax. The remaining authors report no potential relevant conflict of interest., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. CCRL2 affects the sensitivity of myelodysplastic syndrome and secondary acute myeloid leukemia cells to azacitidine.
- Author
-
Karantanos T, Teodorescu P, Arvanitis M, Perkins B, Jain T, DeZern AE, Dalton WB, Christodoulou I, Paun BC, Varadhan R, Esteb C, Rajkhowa T, Bonifant C, Gondek LP, Levis MJ, Yegnasubramanian S, Ghiaur G, and Jones RJ
- Subjects
- Humans, Azacitidine pharmacology, Azacitidine therapeutic use, Cell Line, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Better understanding of the biology of resistance to DNA methyltransferase (DNMT) inhibitors is required to identify therapies that can improve their efficacy for patients with high-risk myelodysplastic syndrome (MDS). CCRL2 is an atypical chemokine receptor that is upregulated in CD34+ cells from MDS patients and induces proliferation of MDS and secondary acute myeloid leukemia (sAML) cells. In this study, we evaluated any role that CCRL2 may have in the regulation of pathways associated with poor response or resistance to DNMT inhibitors. We found that CCRL2 knockdown in TF-1 cells downregulated DNA methylation and PRC2 activity pathways and increased DNMT suppression by azacitidine in MDS/sAML cell lines (MDS92, MDS-L and TF-1). Consistently, CCRL2 deletion increased the sensitivity of these cells to azacitidine in vitro and the efficacy of azacitidine in an MDS-L xenograft model. Furthermore, CCRL2 overexpression in MDS-L and TF-1 cells decreased their sensitivity to azacitidine. Finally, CCRL2 levels were higher in CD34+ cells from MDS and MDS/myeloproliferative neoplasm patients with poor response to DNMT inhibitors. In conclusion, we demonstrated that CCRL2 modulates epigenetic regulatory pathways, particularly DNMT levels, and affects the sensitivity of MDS/sAML cells to azacitidine. These results support CCRL2 targeting as having therapeutic potential in MDS/sAML.
- Published
- 2023
- Full Text
- View/download PDF
22. Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia.
- Author
-
Fathi AT, Kim HT, Soiffer RJ, Levis MJ, Li S, Kim AS, DeFilipp Z, El-Jawahri A, McAfee SL, Brunner AM, Amrein PC, Mims AS, Knight LW, Kelley D, Bottoms AS, Perry LH, Wahl JL, Brock J, Breton E, Marchione DM, Ho VT, and Chen YB
- Subjects
- Humans, Recurrence, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Hematopoietic Stem Cell Transplantation, Long QT Syndrome
- Abstract
Purpose: Isocitrate dehydrogenase 1 (IDH1) mutations occur in 5% to 10% of patients with acute myeloid leukemia (AML). Ivosidenib is an IDH1 inhibitor, approved for use in patients with IDH1-mutated AML., Patients and Methods: We conducted a multicenter, phase I trial of maintenance ivosidenib following allogeneic hematopoietic cell transplantation (HCT) in patients with IDH1-mutated AML. Ivosidenib was initiated between days 30 and 90 following HCT and continued for up to 12 28-day cycles. The first dose level was 500 mg daily, with level reduction to 250 mg daily, if needed, in a 3 × 3 de-escalation design. Ten additional patients would then receive the MTD or recommended phase 2 dose (RP2D). The primary endpoint was establishing the MTD or RP2D of ivosidenib., Results: Eighteen patients were enrolled, of whom 16 initiated post-HCT ivosidenib. One dose-limiting toxicity, grade(g) 3 QTc prolongation, was observed. The RP2D was established at 500 mg daily. Attributable g≥3 adverse events were uncommon, with the most common being QTc prolongation in 2 patients. Eight patients discontinued maintenance, with only one due to adverse event. Six-month cumulative incidence (CI) of gII-IV aGVHD was 6.3%, and 2-year CI of all cGVHD was 63%. Two-year CI of relapse and nonrelapse mortality (NRM) were 19% and 0%, respectively. Two-year progression-free (PFS) was 81%, and 2-year overall survival (OS) was 88%., Conclusions: Ivosidenib is safe and well-tolerated as maintenance therapy following HCT. Cumulative incidence of relapse and NRM, as well as estimations of PFS and OS, were promising in this phase I study., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
23. Quizartinib plus chemotherapy in newly diagnosed patients with FLT3-internal-tandem-duplication-positive acute myeloid leukaemia (QuANTUM-First): a randomised, double-blind, placebo-controlled, phase 3 trial.
- Author
-
Erba HP, Montesinos P, Kim HJ, Patkowska E, Vrhovac R, Žák P, Wang PN, Mitov T, Hanyok J, Kamel YM, Rohrbach JEC, Liu L, Benzohra A, Lesegretain A, Cortes J, Perl AE, Sekeres MA, Dombret H, Amadori S, Wang J, Levis MJ, and Schlenk RF
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cytarabine, Double-Blind Method, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 genetics, Treatment Outcome, Benzothiazoles therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Phenylurea Compounds therapeutic use
- Abstract
Background: Patients with acute myeloid leukaemia (AML) positive for internal tandem duplication (ITD) mutations of FLT3 have poor outcomes. Quizartinib, an oral, highly potent, selective, type 2 FLT3 inhibitor, plus chemotherapy showed antitumour activity with an acceptable safety profile in patients with FLT3-ITD-positive newly diagnosed AML. The aim of the study was to compare the effect of quizartinib versus placebo on overall survival in patients with FLT3-ITD-positive newly diagnosed AML aged 18-75 years., Methods: We conducted a randomised, double-blind, placebo-controlled, phase 3 trial comparing quizartinib and placebo in combination with chemotherapy in induction and consolidation, followed by quizartinib or placebo single-agent continuation, in patients with FLT3-ITD-positive newly diagnosed AML at 193 hospitals and clinics in 26 countries in Europe; North America; and Asia, Australia, and South America. Patients aged 18-75 years were eligible. Patients were randomly assigned (1:1) to the quizartinib group or the placebo group by an independent biostatistician through an interactive web and voice response system, stratified by region, age, and white blood cell count at diagnosis. Patients, investigators, funders, and contract research organisations were masked to treatments assigned. Induction therapy comprised a standard 7 + 3 induction regimen of cytarabine 100 mg/m
2 per day (or 200 mg/m2 per day allowed if institutional or local standard) by continuous intravenous infusion from day 1 to day 7 and anthracycline (daunorubicin 60 mg/m2 per day or idarubicin 12 mg/m2 per day) by intravenous infusion on days 1, 2, and 3, then quizartinib 40 mg orally or placebo once per day, starting on day 8, for 14 days. Patients with complete remission or complete remission with incomplete neutrophil or platelet recovery received standard consolidation with high-dose cytarabine plus quizartinib (40 mg per day orally) or placebo, allogeneic haematopoietic cell transplantation (allo-HCT), or both as consolidation therapy, followed by continuation of single-agent quizartinib or placebo for up to 3 years. The primary outcome was overall survival, defined as time from randomisation until death from any cause and assessed in the intention-to-treat population. Safety was evaluated in all patients who received at least one dose of quizartinib or placebo. This study is registered with ClinicalTrials.gov (NCT02668653)., Findings: Between Sept 27, 2016, and Aug 14, 2019, 3468 patients with AML were screened and 539 patients (294 [55%] male patients and 245 [45%] female patients) with FLT3-ITD-positive AML were included and randomly assigned to the quizartinib group (n=268) or placebo group (n=271). 148 (55%) of 268 patients in the quizartinib group and 168 (62%) of 271 patients in the placebo group discontinued the study, primarily because of death (133 [90%] of 148 in the quizartinib group vs 158 [94%] of 168 in the placebo group) or withdrawal of consent (13 [9%] of 148 in the quizartinib group vs 9 [5%] of 168 in the placebo group). Median age was 56 years (range 20-75, IQR 46·0-65·0). At a median follow-up of 39·2 months (IQR 31·9-45·8), median overall survival was 31·9 months (95% CI 21·0-not estimable) for quizartinib versus 15·1 months (13·2-26·2) for placebo (hazard ratio 0·78, 95% CI 0·62-0·98, p=0·032). Similar proportions of patients in the quizartinib and placebo groups had at least one adverse event (264 [100%] of 265 in the quizartinib group and 265 [99%] of 268 in the placebo group) and one grade 3 or higher adverse event (244 [92%] of 265 in the quizartinib group and 240 [90%] of 268 in the placebo group). The most common grade 3 or 4 adverse events were febrile neutropenia, hypokalaemia, and pneumonia in both groups and neutropenia in the quizartinib group., Interpretation: The addition of quizartinib to standard chemotherapy with or without allo-HCT, followed by continuation monotherapy for up to 3 years, resulted in improved overall survival in adults aged 18-75 years with FLT3-ITD-positive newly diagnosed AML. Based on the results from the QuANTUM-First trial, quizartinib provides a new, effective, and generally well tolerated treatment option for adult patients with FLT3-ITD-positive newly diagnosed AML., Funding: Daiichi Sankyo., Competing Interests: Declaration of interests HPE reports research grants from AbbVie, Agios Pharmaceuticals, ALX Oncology, Amgen, Ascentage, Celgene, Daiichi Sankyo, Forma Therapeutics, Forty Seven, Gilead, GlycoMimetics, ImmunoGen, Jazz Pharmaceuticals, Kura Oncology, MacroGenics, Novartis, PTC Therapeutics, Servier, and Sumitomo Dainippon Pharma; consulting fees for participation on advisory boards for AbbVie, Agios Pharmaceuticals, Astellas, Bristol Myers Squibb, Celgene, Daiichi Sankyo, Genentech, GlycoMimetics, ImmunoGen, Incyte, Jazz Pharmaceuticals, Kura Oncology, MacroGenics, Novartis, Pfizer, Servier, Syros Pharmaceuticals, Takeda, and Trillium Therapeutics; payment for speakers bureaus from AbbVie, Agios Pharmaceuticals, Bristol Myers Squibb, Celgene, Incyte, Jazz Pharmaceuticals, Novartis, and Servier; and has served as a steering committee member for GlycoMimetics, as chair of the Myeloid Neoplasms Repository study for Bristol Myers Squibb and Celgene, and as chair of the independent review committee of the VIALE A and VIALE C studies for AbbVie. PM reports research grants from AbbVie, Bristol Myers Squibb, Jazz Pharmaceuticals, Menarini, Stemline Therapeutics, Novartis, Pfizer, and Takeda; consulting fees from AbbVie, Astellas, BeiGene, Bristol Myers Squibb, Gilead, Incyte, Jazz Pharmaceuticals, Kura Oncology, Menarini, Stemline Therapeutics, Nerviano Medical Sciences, Novartis, Otsuka Pharmaceutical, Pfizer, Ryvu Therapeutics, and Takeda; and payment for speakers bureaus from AbbVie, Astellas, Bristol Myers Squibb, Gilead, Jazz Pharmaceuticals, and Pfizer. H-JK reports research grants from BL&H; consulting fees from AbbVie, AIMS BioScience, Amgen, AMLHub, Astellas, Aston BioSciences, Bristol Myers Squibb, Celgene, Boryung Pharmaceutical, Daiichi Sankyo, Handok, Ingenium, Janssen, LG Chem, Novartis, Pfizer, Sanofi Genzyme, SL VaxiGen, and VigenCell; is on a data safety monitoring board or advisory board for AbbVie, the Asia Pacific Leukemia Consortium, Astellas, Bristol Myers Squibb, Celgene, Daiichi Sankyo, Handok, Janssen, Novartis, Pfizer, and Sanofi Genzyme; and is a leader in other board, society, committee, or advocacy groups for AMLHub, the Asia Pacific Leukemia Consortium, the Asia Pacific Blood and Marrow Transplantation Group, and the Korean Society of Blood and Marrow Transplantation. EP reports consulting fees from KCR US; payment for lectures from Amgen, Angelini, Astellas, Novartis, Pfizer, and Servier; and support for attending meetings from Angelini, Astellas, Bristol Myers Squibb, Jazz Pharmaceuticals, Novartis, Pfizer, and Servier. RV reports consulting fees from AbbVie, Astellas, Pfizer, and PharmaS; and payment for lectures from AbbVie, Astellas, Merck Sharp & Dohme, Novartis, Pfizer, PharmaS, Servier, and Teva. JC reports research grants from AbbVie, Daiichi Sankyo, Novartis, Sun Pharma, and Pfizer; consulting fees from AbbVie, Bio-Path, Daiichi Sankyo, Gilead, Forma Therapeutics, Novartis, Pfizer, and Takeda; payment for lectures from Novartis, Pfizer, and Takeda; and has stock options with Bio-Path. AEP reports research grants from AbbVie, Actinium Pharmaceuticals, Astellas, Bayer, BioMed Valley Discoveries, and Daiichi Sankyo; personal fees from Actinium Pharmaceuticals, Agios Pharmaceuticals, Astellas, Daiichi Sankyo, Forma Therapeutics, Jazz Pharmaceuticals, Leukemia & Lymphoma Society (Beat AML Master Clinical Trial), Loxo Oncology, NewLink Genetics, Novartis, and Takeda; and non-financial support from Arog Pharmaceuticals, Astellas, Jazz Pharmaceuticals, NewLink Genetics, Novartis, and Takeda. MAS reports consulting fees from Bristol Myers Squibb, Kurome Therapeutics, and Novartis; and has stock options with Kurome Therapeutics. HD reports personal fees from Incyte and Servier. JW reports payment for participation on an advisory board from Abbvie and for participation on a data safety monitoring committee from AstraZeneca. MJL reports research grants from Astellas and FujiFilm Pharmaceuticals; consulting fees from AbbVie, Amgen, Bristol Myers Squibb, Daiichi Sankyo, Jazz Pharmaceuticals, Menarini, Pfizer, and Takeda; and payment for lectures from Astellas. RFS reports consulting fees from Daiichi Sankyo for participation on a steering committee and from AbbVie, Jazz Pharmaceuticals, and Pfizer for participation on advisory boards; payment for lectures from Daiichi Sankyo, Novartis, and Pfizer; is on a data safety monitoring board or advisory board for BerGenBio and Novartis; and has been provided with equipment by AbbVie, AstraZeneca, Boehringer Ingelheim, Daiichi Sankyo, PharmaMar, Pfizer, and Roche. TM, JH, YMK, JECR, LL, AB, and AL are employees of Daiichi Sankyo. All other authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
24. Venetoclax in combination with hypomethylating agent for the treatment of advanced myeloproliferative neoplasms and acute myeloid leukemia with extramedullary disease.
- Author
-
Sanber K, Ye K, Tsai HL, Newman M, Webster JA, Gojo I, Ghiaur G, Prince GT, Gondek LP, Smith BD, Levis MJ, DeZern AE, Ambinder AJ, Dalton WB, and Jain T
- Subjects
- Humans, Retrospective Studies, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Sulfonamides, Antineoplastic Combined Chemotherapy Protocols adverse effects, Azacitidine therapeutic use, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
The combination of venetoclax and hypomethylating agent (HMA/venetoclax) has emerged as a treatment option for patients with de novo acute myeloid leukemia (AML) who are unfit to receive intensive chemotherapy. In this single-center retrospective study, we evaluated clinical outcomes following treatment with HMA/venetoclax in 35 patients with advanced myeloproliferative neoplasms, myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes or AML with extramedullary disease. The composite complete remission (CR) rate (including confirmed/presumed complete cytogenetic response, acute leukemia response-complete, CR and CR with incomplete hematologic recovery) was 42.9% with median overall survival (OS) of 9.7 months. Complex karyotype was associated with inferior median OS (3.7 versus 12.2 months; p = 0.0002) and composite CR rate (22% versus 50.0%; p = 0.2444). Although SRSF2 mutations were associated with higher composite CR rate (80.0% versus 28.0%; p = 0.0082), this was not associated with longer median OS (10.9 versus 8.0 months; p = 0.2269). Future studies should include these patient subgroups.
- Published
- 2023
- Full Text
- View/download PDF
25. An Update on FLT3 in Acute Myeloid Leukemia: Pathophysiology and Therapeutic Landscape.
- Author
-
Bystrom R and Levis MJ
- Subjects
- Humans, Nuclear Proteins genetics, Nucleophosmin, Mutation, fms-Like Tyrosine Kinase 3 genetics, Prognosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Purpose of Review: This review aims to summarize the pathophysiology, clinical presentation, and management of acute myeloid leukemia (AML) with FMS-like tyrosine kinase-3 (FLT3) mutations., Recent Findings: The recent European Leukemia Net (ELN2022) recommendations re-classified AML with FLT3 internal tandem duplications (FLT3-ITD) as intermediate risk regardless of Nucleophosmin 1 (NPM1) co-mutation or the FLT3 allelic ratio. Allogeneic hematopoietic cell transplantation (alloHCT) is now recommended for all eligible patients with FLT3-ITD AML. This review outlines the role of FLT3 inhibitors in induction and consolidation, as well as for post-alloHCT maintenance. It outlines the unique challenges and advantages of assessing FLT3 measurable residual disease (MRD) and discusses the pre-clinical basis for the combination of FLT3 and menin inhibitors. And, for the older or unfit patient ineligible for upfront intensive chemotherapy, it discusses the recent clinical trials incorporating FLT3 inhibitors into azacytidine- and venetoclax-based regimens. Finally, it proposes a rational sequential approach for integrating FLT3 inhibitors into less intensive regimens, with a focus on improved tolerability in the older and unfit patient population. The management of AML with FLT3 mutation remains a challenge in clinical practice. This review provides an update on the pathophysiology and therapeutic landscape of FLT3 AML, as well as a clinical management framework for managing the older or unfit patient ineligible for intensive chemotherapy., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
26. Outcomes in Patients with FLT3-Mutated Relapsed/ Refractory Acute Myelogenous Leukemia Who Underwent Transplantation in the Phase 3 ADMIRAL Trial of Gilteritinib versus Salvage Chemotherapy.
- Author
-
Perl AE, Larson RA, Podoltsev NA, Strickland S, Wang ES, Atallah E, Schiller GJ, Martinelli G, Neubauer A, Sierra J, Montesinos P, Recher C, Yoon SS, Maeda Y, Hosono N, Onozawa M, Kato T, Kim HJ, Hasabou N, Nuthethi R, Tiu R, and Levis MJ
- Subjects
- Adult, Humans, Aniline Compounds therapeutic use, Aniline Compounds pharmacology, Recurrence, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
The fms-like tyrosine kinase 3 (FLT3) inhibitor gilteritinib improved the survival of patients with relapsed or refractory (R/R) FLT3-mutated acute myelogenous leukemia (AML) in the phase 3 ADMIRAL trial. In this study, we assessed survival and relapse rates of patients in the ADMIRAL trial who underwent hematopoietic stem cell transplantation (HSCT), as well as safety outcomes in patients who received post-transplantation gilteritinib maintenance therapy. ADMIRAL was a global phase 3 randomized controlled trial that enrolled adult patients with FLT3-mutated R/R AML. Patients with R/R AML who harbored FLT3 internal tandem duplication mutations in the juxtamembrane domain or D835/I836 point mutations in the tyrosine kinase domain were randomized (2:1) to gilteritinib (120 mg/day) or to preselected high- or low-intensity salvage chemotherapy (1 or 2 cycles). Patients in the gilteritinib arm who proceeded to HSCT could receive post-transplantation gilteritinib maintenance therapy if they were within 30 to 90 days post-transplantation and had achieved composite complete remission (CRc) with successful engraftment and no post-transplantation complications. Adverse events (AEs) during HSCT were recorded in the gilteritinib arm only. Survival outcomes and the cumulative incidence of relapse were assessed in patients who underwent HSCT during the trial. Treatment-emergent AEs were evaluated in patients who restarted gilteritinib as post-transplantation maintenance therapy. Patients in the gilteritinib arm underwent HSCT more frequently than those in the chemotherapy arm (26% [n = 64] versus 15% [n = 19]). For all transplantation recipients, 12- and 24-month overall survival (OS) rates were 68% and 47%, respectively. Despite a trend toward longer OS after pretransplantation CRc, post-transplantation survival was comparable in the 2 arms. Patients who resumed gilteritinib after HSCT had a low relapse rate after pretransplantation CRc (20%) or CR (0%). The most common AEs observed with post-transplantation gilteritinib therapy were increased alanine aminotransferase level (45%), pyrexia (43%), and diarrhea (40%); grade ≥3 AEs were related primarily to myelosuppression. The incidences of grade ≥III acute graft-versus-host disease and related mortality were low. Post-transplantation survival was similar across the 2 study arms in the ADMIRAL trial, but higher remission rates with gilteritinib facilitated receipt of HSCT. Gilteritinib as post-transplantation maintenance therapy had a stable safety and tolerability profile and was associated with low relapse rates. Taken together, these data support a preference for bridging therapy with gilteritinib over chemotherapy in transplantation-eligible patients., (Copyright © 2022 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
27. Allogeneic Blood or Marrow Transplantation with High-Dose Post-Transplantation Cyclophosphamide for Acute Lymphoblastic Leukemia in Patients Age ≥55 Years.
- Author
-
Webster JA, Reed M, Tsai HL, Ambinder A, Jain T, Dezern AE, Levis MJ, Showel MM, Prince GT, Hourigan CS, Gladstone DE, Bolanos-Meade J, Gondek LP, Ghiaur G, Dalton WB, Paul S, Fuchs EJ, Gocke CB, Ali SA, Huff CA, Borrello IM, Swinnen L, Wagner-Johnston N, Ambinder RF, Luznik L, Gojo I, Smith BD, Varadhan R, Jones RJ, and Imus PH
- Subjects
- Humans, Middle Aged, Bone Marrow, Cyclophosphamide therapeutic use, Recurrence, Acute Disease, Hematopoietic Stem Cell Transplantation, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Graft vs Host Disease drug therapy
- Abstract
Patients age ≥55 years with acute lymphoblastic leukemia (ALL) fare poorly with conventional chemotherapy, with a 5-year overall survival (OS) of ∼20%. Tyrosine kinase inhibitors and novel B cell-targeted therapies can improve outcomes, but rates of relapse and death in remission remain high. Allogeneic blood or marrow transplantation (alloBMT) provides an alternative consolidation strategy, and post-transplantation cyclophosphamide (PTCy) facilitates HLA-mismatched transplantations with low rates of nonrelapse mortality (NRM) and graft-versus-host disease (GVHD). The transplantation database at Johns Hopkins was queried for patients age ≥55 years who underwent alloBMT for ALL using PTCy. The database included 77 such patients. Most received reduced-intensity conditioning (RIC) (88.3%), were in first complete remission (CR1) (85.7%), and had B-lineage disease (90.9%). For the entire cohort, 5-year relapse-free survival (RFS) and overall survival (OS) were 46% (95% confidence interval [CI], 34% to 57%) and 49% (95% CI, 37% to 60%), respectively. Grade III-IV acute GVHD occurred in only 3% of patients, and chronic GVHD occurred in 13%. In multivariable analysis, myeloablative conditioning led to worse RFS (hazard ratio [HR], 4.65; P = .001), whereas transplantation in CR1 (HR, .30; P = .004) and transplantation for Philadelphia chromosome-positive (Ph
+ ) ALL versus T-ALL (HR, .29; P = .03) were associated with improved RFS. Of the 54 patients who underwent RIC alloBMT in CR1 for B-ALL, the 5-year RFS and OS were 62% (95% CI, 47% to 74%) and 65% (95% CI, 51% to 77%), respectively, with a 5-year relapse incidence of 16% (95% CI, 7% to 27%) and an NRM of 24% (95% CI, 13% to 36%). RIC alloBMT with PTCy in CR1 represents a promising consolidation strategy for B-ALL patients age ≥55 years., (Copyright © 2022 The American Society for Transplantation and Cellular Therapy. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
28. The clinical impact of the molecular landscape of acute myeloid leukemia.
- Author
-
Kayser S and Levis MJ
- Subjects
- Humans, Gemtuzumab, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Research into the underlying pathogenic mechanisms of acute myeloid leukemia (AML) has led to remarkable advances in our understanding of the disease. Mutations now allow us to explore the enormous diversity among cytogenetically defined subsets of AML, particularly the large subset of cytogenetically normal AML. Despite the progress in unraveling the tumor genome, only a small number of recurrent mutations have been incorporated into risk-stratification schemes and have been proven to be clinically relevant, targetable lesions. The current World Health Organization Classification of myeloid neoplasms and leukemia includes eight AML categories defined by recurrent genetic abnormalities as well as three categories defined by gene mutations. We here discuss the utility of molecular markers in AML in prognostication and treatment decision-making. New therapies based on targetable markers include IDH inhibitors (ivosidenib, enasidenib), venetoclax-based therapy, FLT3 inhibitors (midostaurin, gilteritinib, and quizartinib), gemtuzumab ozogamicin, magrolimab and menin inhibitors.
- Published
- 2023
- Full Text
- View/download PDF
29. Enasidenib as maintenance following allogeneic hematopoietic cell transplantation for IDH2-mutated myeloid malignancies.
- Author
-
Fathi AT, Kim HT, Soiffer RJ, Levis MJ, Li S, Kim AS, Mims AS, DeFilipp Z, El-Jawahri A, McAfee SL, Brunner AM, Narayan R, Knight LW, Kelley D, Bottoms AS, Perry LH, Wahl JL, Brock J, Breton E, Ho VT, and Chen YB
- Subjects
- Humans, Recurrence, Graft vs Host Disease, COVID-19, Hematopoietic Stem Cell Transplantation adverse effects, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute drug therapy, Myeloproliferative Disorders
- Abstract
IDH2 (isocitrate dehydrogenase 2) mutations occur in approximately 15% of patients with acute myeloid leukemia (AML). The IDH2 inhibitor enasidenib was recently approved for IDH2-mutated relapsed or refractory AML. We conducted a multi-center, phase I trial of maintenance enasidenib following allogeneic hematopoietic cell transplantation (HCT) in patients with IDH2-mutated myeloid malignancies. Two dose levels, 50mg and 100mg daily were studied in a 3 × 3 dose-escalation design, with 10 additional patients treated at the recommended phase 2 dose (RP2D). Enasidenib was initiated between days 30 and 90 following HCT and continued for twelve 28-day cycles. Twenty-three patients were enrolled, of whom 19 initiated post-HCT maintenance. Two had myelodysplastic syndrome, and 17 had AML. All but 3 were in first complete remission. No dose limiting toxicities were observed, and the RP2D was established at 100mg daily. Attributable grade ≥3 toxicities were rare, with the most common being cytopenias. Eight patients stopped maintenance before completing 12 cycles, due to adverse events (n=3), pursuing treatment for graft-vs-host disease (GVHD) (n=2), clinician choice (n=1), relapse (n=1), and COVID infection (n=1). No cases of grade ≥3 acute GVHD were seen, and 12-month cumulative incidence of moderate/severe chronic GVHD was 42% (20-63%). Cumulative incidence of relapse was 16% (95% CI: 3.7-36%); 1 subject relapsed while receiving maintenance. Two-year progression-free and overall survival were 69% (95% CI: 39-86%) and 74% (95% CI, 44-90%), respectively. Enasidenib is safe, well-tolerated, with preliminary activity as maintenance therapy following HCT, and merits additional study. The study was registered at www.clinicaltrials.gov (#NCT03515512)., (© 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
30. Arsenic trioxide dose capping to decrease toxicity in the treatment of acute promyelocytic leukemia.
- Author
-
Zacholski K, Hambley B, Hickey E, Kashanian S, Li A, Baer MR, Duong VH, Newman MJ, DeZern A, Gojo I, Smith BD, Levis MJ, Varadhan R, Gehrie E, Emadi A, and Ghiaur G
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Arsenic Trioxide adverse effects, Humans, Oxides adverse effects, Retrospective Studies, Treatment Outcome, Tretinoin adverse effects, Arsenicals adverse effects, Leukemia, Promyelocytic, Acute drug therapy
- Abstract
Arsenic trioxide (ATO) and all-trans retinoic acid (ATRA) combination therapy yields high complete remission and disease-free survival rates in acute promyelocytic leukemia (APL). ATO is dosed on actual body weight and high ATO doses in overweight patients may contribute to increased toxicity. We performed a retrospective, two-center study comparing toxicities in patients who received the Lo-Coco et al ATRA/ATO regimen with capped ATO, ≤10 mg/dose, and non-capped ATO, >10 mg/dose. A total of 44 patients were included; 15 received doses ≤10 mg and 29 received >10 mg. During induction, there was no difference in the incidence of grade ≥3 hepatotoxicity, grade ≥3 QTc prolongation, neurotoxicity, and cardiac toxicity between groups. In consolidation, patients receiving >10 mg/dose experienced a greater incidence of neurotoxicity (66.7% vs 22.2%; p = 0.046). Capping doses saved $24634.37/patient and reduced waste of partially-used vials. At a median follow-up of 27 months, no disease relapses occurred in either group. This represents an opportunity to improve the safety profile of this highly effective regimen.
- Published
- 2022
- Full Text
- View/download PDF
31. Outcome of relapsed or refractory acute B-lymphoblastic leukemia patients and BCR-ABL -positive blast cell crisis of B-lymphoid lineage with extramedullary disease receiving inotuzumab ozogamicin.
- Author
-
Kayser S, Sartor C, Luskin MR, Webster J, Giglio F, Panitz N, Brunner AM, Fante M, Lutz C, Wolff D, Ho AD, Levis MJ, Schlenk RF, and Papayannidis C
- Subjects
- Adult, Blast Crisis, Humans, Inotuzumab Ozogamicin, Middle Aged, Remission Induction, Hematopoietic Stem Cell Transplantation adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Acute lymphoblastic leukemia (ALL) can relapse in the extramedullary compartment, with or without medullary involvement. Response to treatment may be individual. We evaluated response to inotuzumab ozogamicin in 31 patients with relapsed/refractory B-ALL with extramedullary disease. Median age was 31 years (range, 19-81). All patients were heavily pretreated, including allogeneic hematopoietic stem cell transplantation (HSCT; n=18). Overall response rate after two cycles of inotuzumab ozogamicin was 84% (complete remission, 55%; partial remission, 29%; resistant disease, 13%; early death, 3%). The median follow-up was 29 months and median overall survival was 12.8 months. One-year and 2-year overall survival rates were 53% (95% CI: 37-76%) and 18% (95% CI: 8-43%), respectively. Age had no impact on overall survival when assessed as a continuous variable or dichotomized at 60 years. Twelve patients proceeded to allogeneic HSCT (complete remission, n=6; partial remission, n=3; resistant disease, n=3). Prior to allogeneic HSCT, eight patients received two or fewer cycles and four patients received three or four cycles of inotuzumab ozogamicin. Sinusoidal obstruction syndrome was reported in three patients, including one after transplantation. Allogeneic HSCT, evaluated as a time-dependent variable, had no impact on overall survival. Inotuzumab ozogamicin seems to be effective as a debulking strategy in relapsed/refractory ALL with extramedullary disease. However, inotuzumab ozogamicin followed by allogeneic HSCT seems not to be effective in maintaining long-term disease control.
- Published
- 2022
- Full Text
- View/download PDF
32. Emerging Mutations at Relapse in Patients with FLT3-Mutated Relapsed/Refractory Acute Myeloid Leukemia Who Received Gilteritinib Therapy in the Phase 3 Admiral Trial
- Author
-
Smith CC, Levis MJ, Perl AE, Martinelli G, Neubauer A, Berman E, Montesinos P, Baer MR, Larson RA, Chou WC, Yokoyama H, Recher C, Yoon SS, Hill JE, Rosales M, and Bahceci E
- Abstract
Smith: Revolution Medicines: Research Funding; Astellas Pharma: Research Funding; fujiFilm: Research Funding; Abbvie: Research Funding. Levis:Astellas: Consultancy, Research Funding; FUJIFILM: Consultancy, Research Funding; Menarini: Consultancy, Honoraria; Novartis: Consultancy, Research Funding; Agios: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Daiichi Sankyo Inc: Consultancy, Honoraria. Perl:Astellas: Consultancy, Honoraria, Other: Non-financial support included travel costs for advisory board meetings as well as a medical writing company that assisted with manuscript preparation/submission and slide deck assembly for academic meeting presentations of trial data., Research Funding; BioMed Valley Discoveries: Research Funding; Daiichi Sankyo: Consultancy, Honoraria, Other, Research Funding; Arog: Consultancy, Other: Non-financial support included travel costs for advisory board meetings.; AbbVie: Consultancy, Honoraria, Other: Non-financial support included travel costs for advisory board meetings.; Actinium Pharmaceuticals: Consultancy, Honoraria, Other: Clinical Advisory Board member, Research Funding; Agios: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Non-financial support included travel costs for advisory board meetings.; Jazz: Consultancy, Honoraria, Other: Non-financial support included travel costs for advisory board meetings.; NewLink Genetics: Consultancy, Honoraria, Other: Non-financial support included travel costs for advisory board meetings.; Takeda: Consultancy, Honoraria, Other: Non-financial support included travel costs for advisory board meetings.; Bayer: Research Funding; FujiFilm: Research Funding; Novartis: Honoraria, Other: Advisory board, Non-financial support included travel costs for advisory board meetings as well as a medical writing company that assisted with manuscript preparation/submission and slide deck assembly for academic meeting presentations of the data., Research Funding. Martinelli:Daiichi Sankyo: Consultancy, Honoraria; Roche: Consultancy, Other: trial grant; Ariad: Consultancy, Other: trial grant; Janssen: Consultancy, Other: trial grant; Amgen: Consultancy, Other: trial grant; Pfizer: Consultancy, Other: trial grant; Abbvie: Consultancy, Honoraria, Other: trial grant; Celgene: Consultancy, Honoraria, Other: trial grant; Novartis: Consultancy, Other: trial grant; Incyte: Consultancy, Other: trial grant. Berman:Astellas: Membership on an entity's Board of Directors or advisory committees, Research Funding. Montesinos:Karyopharm: Membership on an entity's Board of Directors or advisory committees, Other: Research support; Pfizer: Membership on an entity's Board of Directors or advisory committees, Other: Research support, Research Funding, Speakers Bureau; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Research support, Speakers Bureau; Teva: Membership on an entity's Board of Directors or advisory committees, Other: Research support, Research Funding, Speakers Bureau; Abbvie: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Other: Research support, Research Funding, Speakers Bureau; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Research support, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Other: Research support, Research Funding, Speakers Bureau; Incyte: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Baer:Abbvie: Research Funding; Astellas: Research Funding; Al Therapeutics: Research Funding; Forma: Research Funding; Incyte: Research Funding; Kite: Research Funding; Takeda: Research Funding. Larson:Celgene: Consultancy; Agios: Consultancy; Novartis: Honoraria, Other: Contracts for clinical trials. Yokoyama:Astellas: Other: Travel expenses. Recher:Incyte: Honoraria; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sunesis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Jazz: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; Macrogenics: Consultancy, Membership on an entity's Board of Directors or advisory committees. Yoon:MSD: Consultancy; Kyowa Hako Kirin: Research Funding; Janssen: Consultancy; Genentech, Inc.: Research Funding; Amgen: Consultancy, Honoraria; Yuhan Pharma: Research Funding; Novartis: Consultancy, Honoraria. Hill:Astellas: Employment; Ligacept, LLC.: Other: Stock, Patents Royalties. Rosales:Astellas: Employment. Bahceci:Astellas: Employment, Patents Royalties.
- Published
- 2019
33. Genomic landscape of myelodysplastic/myeloproliferative neoplasm can predict response to hypomethylating agent therapy.
- Author
-
Karantanos T, Tsai HL, Gondek LP, DeZern AE, Ghiaur G, Dalton WB, Gojo I, Prince GT, Webster J, Ambinder A, Smith BD, Levis MJ, Varadhan R, Jones RJ, and Jain T
- Subjects
- Core Binding Factor Alpha 2 Subunit genetics, Genomics, Humans, Mutation, Myelodysplastic-Myeloproliferative Diseases diagnosis, Myelodysplastic-Myeloproliferative Diseases drug therapy, Myelodysplastic-Myeloproliferative Diseases genetics, Neoplasms
- Abstract
There are currently no known predictors of myelodysplastic syndrome (MDS)/myeloproliferative overlap neoplasm (MPN) patients' response to hypomethylating agents (HMA). Forty-three patients with MDS/MPN who were treated with HMA during chronic phase and had next-generation sequencing using the established 63-genes panel were identified. Complete and partial remission and marrow response were assessed based on the MDS/MPN International Working Group response criteria. On univariate analysis, younger age, higher number of mutations, and mutations in SETBP1 , RUNX1 , or EZH2 were associated with no response. Multivariable analysis for modeling response were conducted via least absolute shrinkage and selection operator logistic regression approach, and showed that mutations in SETBP1 , RUNX1 , or EZH2 predict lack of HMA response. While limited by sample size, our findings suggest that genomic landscape can potentially identify MDS/MPN patients with lower likelihood of response to HMA.
- Published
- 2022
- Full Text
- View/download PDF
34. 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
35. Targeting FLT3 mutations in AML: review of current knowledge and evidence.
- Author
-
Daver N, Schlenk RF, Russell NH, and Levis MJ
- Subjects
- Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Prognosis, Leukemia, Myeloid, Acute drug therapy, Molecular Targeted Therapy, Mutation, Protein Kinase Inhibitors therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Genomic investigations of acute myeloid leukemia (AML) have demonstrated that several genes are recurrently mutated, leading to new genomic classifications, predictive biomarkers, and new therapeutic targets. Mutations of the FMS-like tyrosine kinase 3 (FLT3) gene occur in approximately 30% of all AML cases, with the internal tandem duplication (ITD) representing the most common type of FLT3 mutation (FLT3-ITD; approximately 25% of all AML cases). FLT3-ITD is a common driver mutation that presents with a high leukemic burden and confers a poor prognosis in patients with AML. The prognostic value of a FLT3 mutation in the tyrosine kinase domain (FLT3-TKD), which has a lower incidence in AML (approximately 7-10% of all cases), is uncertain. Accumulating evidence demonstrates that FLT3 mutational status evolves throughout the disease continuum. This so-called clonal evolution, together with the identification of FLT3-ITD as a negative prognostic marker, serves to highlight the importance of FLT3-ITD testing at diagnosis and again at relapse. Earlier identification of FLT3 mutations will help provide a better understanding of the patient's disease and enable targeted treatment that may help patients achieve longer and more durable remissions. First-generation FLT3 inhibitors developed for clinical use are broad-spectrum, multikinase inhibitors; however, next-generation FLT3 inhibitors are more specific, more potent, and have fewer toxicities associated with off-target effects. Primary and secondary acquired resistance to FLT3 inhibitors remains a challenge and provides a rationale for combining FLT3 inhibitors with other therapies, both conventional and investigational. This review focuses on the pathological and prognostic role of FLT3 mutations in AML, clinical classification of the disease, recent progress with next-generation FLT3 inhibitors, and mechanisms of resistance to FLT3 inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
36. Clinical outcomes in patients with relapsed/refractory FLT3-mutated acute myeloid leukemia treated with gilteritinib who received prior midostaurin or sorafenib.
- Author
-
Perl AE, Hosono N, Montesinos P, Podoltsev N, Martinelli G, Panoskaltsis N, Recher C, Smith CC, Levis MJ, Strickland S, Röllig C, Groß-Langenhoff M, Chou WC, Lee JH, Yokoyama H, Hasabou N, Lu Q, Tiu RV, and Altman JK
- Subjects
- Aniline Compounds, Humans, Mutation, Protein Kinase Inhibitors adverse effects, Pyrazines, Retrospective Studies, Sorafenib pharmacology, Sorafenib therapeutic use, Staurosporine analogs & derivatives, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
The fms-like tyrosine kinase 3 (FLT3) inhibitor gilteritinib is indicated for relapsed or refractory (R/R) FLT3-mutated acute myeloid leukemia (AML), based on its observed superior response and survival outcomes compared with salvage chemotherapy (SC). Frontline use of FLT3 tyrosine kinase inhibitors (TKIs) midostaurin and sorafenib may contribute to cross-resistance to single-agent gilteritinib in the R/R AML setting but has not been well characterized. To clarify the potential clinical impact of prior TKI use, we retrospectively compared clinical outcomes in patients with R/R FLT3-mutated AML in the CHRYSALIS and ADMIRAL trials who received prior midostaurin or sorafenib against those without prior FLT3 TKI exposure. Similarly high rates of composite complete remission (CRc) were observed in patients who received a FLT3 TKI before gilteritinib (CHRYSALIS, 42%; ADMIRAL, 52%) and those without prior FLT3 TKI therapy (CHRYSALIS, 43%; ADMIRAL, 55%). Among patients who received a prior FLT3 TKI in ADMIRAL, a higher CRc rate (52%) and trend toward longer median overall survival was observed in the gilteritinib arm versus the SC arm (CRc = 20%; overall survival, 5.1 months; HR = 0.602; 95% CI: 0.299, 1.210). Remission duration was shorter with prior FLT3 TKI exposure. These findings support gilteritinib for FLT3-mutated R/R AML after prior sorafenib or midostaurin., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
37. Molecular profile of FLT3-mutated relapsed/refractory patients with AML in the phase 3 ADMIRAL study of gilteritinib.
- Author
-
Smith CC, Levis MJ, Perl AE, Hill JE, Rosales M, and Bahceci E
- Subjects
- Aniline Compounds therapeutic use, Humans, Protein Kinase Inhibitors therapeutic use, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Pyrazines therapeutic use
- Abstract
The phase 3 Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FMS-like Tyrosine Kinase (FLT3) Mutation (ADMIRAL) trial demonstrated the superiority of the FLT3 inhibitor, gilteritinib, to salvage chemotherapy (SC) in patients with FLT3-mutated relapsed or refractory (R/R) AML. Baseline comutations, FLT3-internal tandem duplication (ITD) allelic ratio and length, and treatment-emergent mutations were analyzed in patients in the ADMIRAL trial. Baseline comutations were grouped according to gene subgroups (DNA methylation/hydroxymethylation, transcription, chromatin-spliceosome, receptor tyrosine kinase-Ras signaling, TP53-aneuploidy, NPM1, DNMT3A, DNMT3A/NPM1, WT-1, and IDH1/IDH2). Across all but 1 gene subgroup (TP53-aneuploidy), higher pretransplant response rates and a trend toward longer overall survival were observed with gilteritinib vs SC. Patients with DNMT3A/NPM1 comutations who received gilteritinib had the most favorable outcomes of any molecular subgroup analyzed. Survival outcomes with gilteritinib were not adversely affected by FLT3-ITD allelic ratio, FLT3-ITD length, or multiple FLT3-ITD mutations. Among patients who relapsed on gilteritinib, Ras/mitogen-activated protein kinase (MAPK) pathway and FLT3 F691L gene mutations were the most common mutational events associated with treatment resistance. However, the occurrence of Ras/MAPK pathway gene mutations at baseline did not preclude a clinical benefit from gilteritinib. Acquisition of multiple Ras/MAPK pathway gene mutations at relapse suggests a high level of pathway reactivation is needed to overcome the gilteritinib treatment effect. These findings provide insight into the R/R AML molecular profile and the impact of FLT3 inhibitors on mutational evolution associated with treatment resistance and benefit of gilteritinib across a wide spectrum of molecular and genetic subgroups in FLT3-mutated R/R AML. This trial was registered at www.clinicaltrials.gov as #NCT02421939., (© 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
38. Characteristics and outcome of patients with core-binding factor acute myeloid leukemia and FLT3-ITD: results from an international collaborative study.
- Author
-
Kayser S, Kramer M, Martínez-Cuadrón D, Grenet J, Metzeler KH, Sustkova Z, Luskin MR, Brunner AM, Elliott MA, Gil C, Marini SC, Ráčil Z, Cetkovsky P, Novak J, Perl AE, Platzbecker U, Stölzel F, Ho AD, Thiede C, Stone RM, Röllig C, Montesinos P, Schlenk RF, and Levis MJ
- Subjects
- Adult, Aged, Aged, 80 and over, Core Binding Factors genetics, Humans, Middle Aged, Mutation, Prognosis, Remission Induction, Retrospective Studies, Young Adult, fms-Like Tyrosine Kinase 3 genetics, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
The aim of this study was to evaluate the prognostic impact of FLT3-ITD in core-binding factor acute myeloid leukemia (CBFAML) in an international, multicenter survey of 97 patients of whom 52% had t(8;21)(q22;q22) and 48% had inv(16)(p13q22)/t(16;16)(p13;q22). The median age of the patients was 53 years (range, 19-81). Complete remission after anthracycline-based induction (n=86) and non-intensive therapy (n=11) was achieved in 97% and 36% of the patients, respectively. The median follow-up was 4.43 years (95% confidence interval [95% CI]: 3.35-7.39 years). The median survival after intensive and non-intensive treatment was not reached and 0.96 years, respectively. Among intensively treated patients, inv(16) with trisomy 22 (n=11) was associated with a favorable 4-year relapse-free survival rate of 80% (95% CI: 59-100%) as compared to 38% (95% CI: 27-54%; P=0.02) in all other patients with CBFAML/ FLT3-ITD (n=75). Overall, 24 patients underwent allogeneic hematopoietic cell transplantation (HCT), 12 in first complete remission and 12 after relapse. Allogeneic HCT in first complete remission was not beneficial (P=0.60); however, allogeneic HCT seemed to improve median survival in relapsed patients compared to that of patients treated with chemotherapy (not reached vs. 0.6 years, respectively; P=0.002). Excluding patients with inv(16) with trisomy 22, our data indicate that compathe outcome of CBF-AML patients with FLT3-ITD may be inferior to that of patients without FLT3-ITD (based on previously published data), suggesting that prognostically CBF-AML patients with FLT3-ITD should not be classified as favorable-risk. FLT3-inhibitors may improve the outcome of these patients.
- Published
- 2022
- Full Text
- View/download PDF
39. The role of the atypical chemokine receptor CCRL2 in myelodysplastic syndrome and secondary acute myeloid leukemia.
- Author
-
Karantanos T, Teodorescu P, Perkins B, Christodoulou I, Esteb C, Varadhan R, Helmenstine E, Rajkhowa T, Paun BC, Bonifant C, Dalton WB, Gondek LP, Moliterno AR, Levis MJ, Ghiaur G, and Jones RJ
- Subjects
- Cell Proliferation, Female, Hematopoiesis, Humans, Male, Signal Transduction, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology
- Abstract
The identification of new pathways supporting the myelodysplastic syndrome (MDS) primitive cells growth is required to develop targeted therapies. Within myeloid malignancies, men have worse outcomes than women, suggesting male sex hormone-driven effects in malignant hematopoiesis. Androgen receptor promotes the expression of five granulocyte colony-stimulating factor receptor-regulated genes. Among them, CCRL2 encodes an atypical chemokine receptor regulating cytokine signaling in granulocytes, but its role in myeloid malignancies is unknown. Our study revealed that CCRL2 is up-regulated in primitive cells from patients with MDS and secondary acute myeloid leukemia (sAML). CCRL2 knockdown suppressed MDS92 and MDS-L cell growth and clonogenicity in vitro and in vivo and decreased JAK2/STAT3/STAT5 phosphorylation. CCRL2 coprecipitated with JAK2 and potentiated JAK2-STAT interaction. Erythroleukemia cells expressing JAK2V617F showed less effect of CCRL2 knockdown, whereas fedratinib potentiated the CCRL2 knockdown effect. Conclusively, our results implicate CCRL2 as an MDS/sAML cell growth mediator, partially through JAK2/STAT signaling.
- Published
- 2022
- Full Text
- View/download PDF
40. Updates on targeted therapies for acute myeloid leukaemia.
- Author
-
Kayser S and Levis MJ
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor, Clinical Decision-Making, Combined Modality Therapy, Disease Management, Disease Susceptibility, Drug Resistance, Genetic Predisposition to Disease, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute mortality, Mutation, Prognosis, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Recurrence, Retreatment, Treatment Outcome, Leukemia, Myeloid, Acute drug therapy, Molecular Targeted Therapy adverse effects, Molecular Targeted Therapy methods
- Abstract
In the past few years research in the underlying pathogenic mechanisms of acute myeloid leukaemia (AML) has led to remarkable advances in our understanding of the disease. Cytogenetic and molecular aberrations are the most important factors in determining response to chemotherapy as well as long-term outcome, but beyond prognostication are potential therapeutic targets. Our increased understanding of the pathogenesis of AML facilitated by next-generation sequencing has spurred the development of new compounds in the treatment of AML, particularly the creation of small molecules that target the disease on a molecular level. Many of the hopeful predictions outlined in our AML review of 2018 are now therapeutic realities: gemtuzumab ozogamicin, venetoclax, FLT3 inhibitors (midostaurin, gilteritinib), IDH inhibitors (ivosidenib, enasidenib), CPX-351, glasdegib, oral decitabine, and oral azacitidine. Others may soon be (quizartinib, APR246 magrolimab, menin inhibitors). The wealth of positive data allows reconsideration of what might soon be new standards of care in younger and older patients with AML. In this review we give an overview of recently approved therapies in AML and address present and future research directions., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
41. Publisher Correction: Outcome of older (≥70 years) APL patients frontline treated with or without arsenic trioxide-an International Collaborative Study.
- Author
-
Kayser S, Rahmé R, Martínez-Cuadrón D, Ghiaur G, Thomas X, Sobas M, Guerci-Bresler A, Garrido A, Pigneux A, Gil C, Raffoux E, Tormo M, Vey N, de la Serna J, Salamero O, Lengfelder E, Levis MJ, Fenaux P, Sanz MA, Platzbecker U, Schlenk RF, Adès L, and Montesinos P
- Published
- 2021
- Full Text
- View/download PDF
42. Phase 1 study of the histone deacetylase inhibitor entinostat plus clofarabine for poor-risk Philadelphia chromosome-negative (newly diagnosed older adults or adults with relapsed refractory disease) acute lymphoblastic leukemia or biphenotypic leukemia.
- Author
-
Carraway HE, Sawalha Y, Gojo I, Lee MJ, Lee S, Tomita Y, Yuno A, Greer J, Smith BD, Pratz KW, Levis MJ, Gore SD, Ghosh N, Dezern A, Blackford AL, Baer MR, Gore L, Piekarz R, Trepel JB, and Karp JE
- Subjects
- Adult, Aged, Benzamides administration & dosage, Clofarabine administration & dosage, Female, Follow-Up Studies, Histone Deacetylase Inhibitors therapeutic use, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Recurrence, Local pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Prognosis, Pyridines administration & dosage, Salvage Therapy, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Lineage, Drug Resistance, Neoplasm, Neoplasm Recurrence, Local drug therapy, Philadelphia Chromosome, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Purpose: Despite advances in immunotherapies, the prognosis for adults with Philadelphia chromosome-negative, newly diagnosed (ND) or relapsed/refractory (R/R) acute lymphoblastic leukemia/acute biphenotypic leukemia (ALL/ABL) remains poor. The benzamide derivative entinostat inhibits histone deacetylase and induces histone hyperacetylation. The purine nucleoside analogue clofarabine is FDA-approved for R/R ALL in children 1-21 years of age. Low doses of clofarabine have been reported to induce DNA hypomethylation. We conducted a phase 1 study of low dose clofarabine with escalating doses of entinostat in adults with ND or R/R ALL/ABL., Experimental Design: Adults ≥60 years with ND ALL/ABL or ≥21 years with R/R ALL/ABL received repeated cycles every 3 weeks of entinostat (4 mg, 6 mg or 8 mg orally days 1 and 8) and clofarabine (10 mg/m
2 /day IV for 5 days, days 3-7) (Arm A). Adults aged 40-59 years with ND ALL/ABL or age ≥21 years in first relapse received entinostat and clofarabine prior to traditional chemotherapy on day 11 (Arm B). Changes in DNA damage, global protein lysine acetylation, myeloid-derived suppressor cells and monocytes were measured in PBMCs before and during therapy., Results: Twenty-eight patients were treated at three entinostat dose levels with the maximum administered dose being entinostat 8 mg. The regimen was well tolerated with infectious and metabolic derangements more common in the older population versus the younger cohort. There was no severe hyperglycemia and no peripheral neuropathy in this small study. There were 2 deaths (1 sepsis, 1 intracranial bleed). Overall response rate was 32 %; it was 50 % for ND ALL/ABL. Entinostat increased global protein acetylation and inhibited immunosuppressive monocyte subpopulations, while clofarabine induced DNA damage in all cell subsets examined., Conclusion: Entinostat plus clofarabine appears to be tolerable and active in older adults with ND ALL/ABL, but less active in R/R patients. Further evaluation of this regimen in ND ALL/ABL appears warranted., (Copyright © 2021. Published by Elsevier Ltd.)- Published
- 2021
- Full Text
- View/download PDF
43. Phase II Trial of Pembrolizumab after High-Dose Cytarabine in Relapsed/Refractory Acute Myeloid Leukemia.
- Author
-
Zeidner JF, Vincent BG, Ivanova A, Moore D, McKinnon KP, Wilkinson AD, Mukhopadhyay R, Mazziotta F, Knaus HA, Foster MC, Coombs CC, Jamieson K, Van Deventer H, Webster JA, Prince GT, DeZern AE, Smith BD, Levis MJ, Montgomery ND, Luznik L, Serody JS, and Gojo I
- Subjects
- Antibodies, Monoclonal, Humanized, Antineoplastic Combined Chemotherapy Protocols adverse effects, CD8-Positive T-Lymphocytes, Drug Resistance, Neoplasm, Humans, Cytarabine adverse effects, Leukemia, Myeloid, Acute drug therapy
- Abstract
Immune suppression, exhaustion, and senescence are frequently seen throughout disease progression in acute myeloid leukemia (AML). We conducted a phase II study of high-dose cytarabine followed by pembrolizumab 200 mg i.v. on day 14 to examine whether PD-1 inhibition improves clinical responses in relapsed/refractory (R/R) AML. Overall responders could receive pembrolizumab maintenance up to 2 years. Among 37 patients enrolled, the overall response rate, composite complete remission (CRc) rate (primary endpoint), and median overall survival (OS) were 46%, 38%, and 11.1 months, respectively. Patients with refractory/early relapse and those receiving treatment as first salvage had encouraging outcomes (median OS, 13.2 and 11.3 months, respectively). Grade ≥3 immune-related adverse events were rare (14%) and self-limiting. Patients who achieved CRc had a higher frequency of progenitor exhausted CD8
+ T cells expressing TCF-1 in the bone marrow prior to treatment. A multifaceted correlative approach of genomic, transcriptomic, and immunophenotypic profiling offers insights on molecular correlates of response and resistance to pembrolizumab., Significance: Immune-checkpoint blockade with pembrolizumab was tolerable and feasible after high-dose cytarabine in R/R AML, with encouraging clinical activity, particularly in refractory AML and those receiving treatment as first salvage regimen. Further study of pembrolizumab and other immune-checkpoint blockade strategies after cytotoxic chemotherapy is warranted in AML. See related commentary by Wei et al., p. 551 . This article is highlighted in the In This Issue feature, p. 549 ., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
44. Correction to: Pharmacokinetic Profile of Gilteritinib: A Novel FLT-3 Tyrosine Kinase Inhibitor.
- Author
-
James AJ, Smith CC, Litzow M, Perl AE, Altman JK, Shepard D, Kadokura T, Souda K, Patton M, Lu Z, Liu C, Moy S, Levis MJ, and Bahceci E
- Published
- 2021
- Full Text
- View/download PDF
45. A method for overcoming plasma protein inhibition of tyrosine kinase inhibitors.
- Author
-
Young DJ, Nguyen B, Li L, Higashimoto T, Levis MJ, Liu JO, and Small D
- Subjects
- Animals, Blood Proteins therapeutic use, Mice, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute drug therapy, Protein Kinase Inhibitors pharmacology
- Abstract
FMS-like tyrosine kinase 3 (FLT3) is the most frequently-mutated gene in acute myeloid leukemia and a target for tyrosine kinase inhibitors (TKI). FLT3 TKI have yielded limited improvements to clinical outcomes. One reason for this is TKI inhibition by endogenous factors. We characterized plasma protein binding of FLT3 TKI, specifically staurosporine-derivatives (STS-TKI) by alpha-1-acid glycoprotein (AGP); simulating its effects upon drug efficacy. Human AGP inhibits the anti-proliferative activity of STS-TKI in FLT3-ITD-dependent cells, with IC
50 shifts higher than clinically achievable. This is not seen with non-human plasma. Mifepristone co-treatment, with its higher AGP affinity, improves TKI activity despite AGP, yielding IC50 s predicted to be clinically effective. In a mouse model of AGP drug inhibition, mifepristone restores midostaurin activity. This suggests combinatorial methods for overcoming plasma protein inhibition of existing TKIs for leukemia as well as providing a platform for investigating the drug-protein interaction space for developing more potent small-molecule agents., Competing Interests: Conflict of Interest The authors report no subject specific conflicts of interest. D.S. does serve on the SAB of InSilico Medicine and receives research support and serves as a consultant for an unrelated project from PHarosI&BT co, Ltd.. D.J.Y. is now the principal investigator on an unrelated trial sponsored by Novartis, begun after his involvement in this work, and does not receive compensation.- Published
- 2021
- Full Text
- View/download PDF
46. Deep learning for diagnosis of acute promyelocytic leukemia via recognition of genomically imprinted morphologic features.
- Author
-
Sidhom JW, Siddarthan IJ, Lai BS, Luo A, Hambley BC, Bynum J, Duffield AS, Streiff MB, Moliterno AR, Imus P, Gocke CB, Gondek LP, DeZern AE, Baras AS, Kickler T, Levis MJ, and Shenderov E
- Abstract
Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia (AML), classified by a translocation between chromosomes 15 and 17 [t(15;17)], that is considered a true oncologic emergency though appropriate therapy is considered curative. Therapy is often initiated on clinical suspicion, informed by both clinical presentation as well as direct visualization of the peripheral smear. We hypothesized that genomic imprinting of morphologic features learned by deep learning pattern recognition would have greater discriminatory power and consistency compared to humans, thereby facilitating identification of t(15;17) positive APL. By applying both cell-level and patient-level classification linked to t(15;17) PML/RARA ground-truth, we demonstrate that deep learning is capable of distinguishing APL in both discovery and prospective independent cohort of patients. Furthermore, we extract learned information from the trained network to identify previously undescribed morphological features of APL. The deep learning method we describe herein potentially allows a rapid, explainable, and accurate physician-aid for diagnosing APL at the time of presentation in any resource-poor or -rich medical setting given the universally available peripheral smear.
- Published
- 2021
- Full Text
- View/download PDF
47. Deletions in FLT-3 juxtamembrane domain define a new class of pathogenic mutations: case report and systematic analysis.
- Author
-
Young DJ, Nguyen B, Zhu R, Seo J, Li L, Levis MJ, Pratz KW, Duffield AS, and Small D
- Subjects
- Humans, Mutation, Signal Transduction, Tandem Repeat Sequences, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
The FMS-like tyrosine kinase 3 (FLT-3) is the most frequently mutated gene in acute myeloid leukemia (AML), a high-risk feature, and now the target of tyrosine kinase inhibitors (TKIs), which are approved and in development. The most common mutation is the internal tandem duplication (ITD). We present a novel mutation, FLT-3/Q575Δ, identified in a patient with AML through next-generation sequencing (NGS). This mutation is activating, drives downstream signaling comparable to FLT-3/ITD, and can be targeted using available FLT-3 TKIs. We present the results of a systematic analysis that identified Y572Δ, E573Δ, and S574Δ as similarly activating and targetable deletions located in the FLT-3 juxtamembrane domain (JMD). These mutations target key residues in the JMD involved in the interactions within FLT-3 that regulate its activation. Our results suggest a new class of FLT-3 mutations that may have an impact on patient care and highlight the increasing importance of a systematic understanding of FLT-3 mutations other than ITD. It is likely that, as NGS becomes more commonly used in the diagnosis of patients with AML, these and other activating mutations will be discovered with increasing frequency., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
48. FLT3 -ITD Allelic Burden and Acute Promyelocytic Leukemia Risk Stratification.
- Author
-
Li AY, Kashanian SM, Hambley BC, Zacholski K, Duong VH, El Chaer F, Holtzman NG, Gojo I, Webster JA, Norsworthy KJ, Smith BD, DeZern AE, Levis MJ, Baer MR, Kamangar F, Ghiaur G, and Emadi A
- Abstract
The significance of FLT3 -ITD in acute promyelocytic leukemia (APL) is not well-established. We performed a bi-center retrospective study of 138 APL patients, 59 (42.8%) of whom had FLT3 -ITD. APL patients with FLT3 -ITD had higher baseline white blood cell counts (WBCs) ( p < 0.001), higher hemoglobin, ( p = 0.03), higher aspartate aminotransferase ( p = 0.001), lower platelets ( p = 0.004), lower fibrinogen ( p = 0.003), and higher incidences of disseminated intravascular coagulation ( p = 0.005), M3v variant morphology ( p < 0.001), and the bcr3 isoform ( p < 0.001). FLT3 -ITD was associated with inferior post-consolidation complete remission (CR) ( p = 0.02) and 5-year overall survival (OS) of 79.7%, compared to 94.4% for FLT3 -WT (wild-type) ( p = 0.02). FLT3 -ITD was strongly associated with baseline WBCs ≥ 25 × 10
9 /L (odds ratio (OR): 54.4; 95% CI: 10.4-286.1; p < 0.001). High FLT3 -ITD allelic burdens correlated with high-risk (HR) Sanz scores and high WBCs, with every 1% increase in allelic burden corresponding to a 0.6 × 109 /L increase in WBC. HR APL was associated with a 38.5% increase in allelic burden compared with low-risk (LR) APL (95% CI: 19.8-57.2; p < 0.001). Our results provide additional evidence that FLT3 -ITD APL is a distinct subtype of APL that warrants further study to delineate potential differences in therapeutic approach.- Published
- 2021
- Full Text
- View/download PDF
49. Single-cell DNA sequencing reveals complex mechanisms of resistance to quizartinib.
- Author
-
Peretz CAC, McGary LHF, Kumar T, Jackson H, Jacob J, Durruthy-Durruthy R, Levis MJ, Perl A, Huang BJ, and Smith CC
- Subjects
- Sequence Analysis, DNA, Benzothiazoles, Phenylurea Compounds
- Published
- 2021
- Full Text
- View/download PDF
50. Characteristics and outcome of patients with acute myeloid leukaemia and t(8;16)(p11;p13): results from an International Collaborative Study.
- Author
-
Kayser S, Hills RK, Langova R, Kramer M, Guijarro F, Sustkova Z, Estey EH, Shaw CM, Ráčil Z, Mayer J, Zak P, Baer MR, Brunner AM, Szotkowski T, Cetkovsky P, Grimwade D, Walter RB, Burnett AK, Ho AD, Ehninger G, Müller-Tidow C, Platzbecker U, Thiede C, Röllig C, Schulz A, Warsow G, Brors B, Esteve J, Russell NH, Schlenk RF, and Levis MJ
- Subjects
- Abnormal Karyotype, Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Combined Modality Therapy, Consolidation Chemotherapy, Disease Progression, Disease-Free Survival, Female, Follow-Up Studies, Hematopoietic Stem Cell Transplantation, Humans, International Cooperation, Leukemia, Myeloid, Acute epidemiology, Leukemia, Myeloid, Acute therapy, Male, Middle Aged, Mutation, Myelodysplastic Syndromes epidemiology, Neoplasms, Second Primary chemically induced, Neoplasms, Second Primary epidemiology, Oncogene Proteins, Fusion genetics, Remission Induction, Survival Analysis, Whole Genome Sequencing, Chromosomes, Human, Pair 16 ultrastructure, Chromosomes, Human, Pair 8 ultrastructure, Leukemia, Myeloid, Acute genetics, Translocation, Genetic
- Abstract
In acute myeloid leukaemia (AML) t(8;16)(p11;p13)/MYST3-CREBBP is a very rare abnormality. Previous small series suggested poor outcome. We report on 59 patients with t(8;16) within an international, collaborative study. Median age was 52 (range: 16-75) years. AML was de novo in 58%, therapy-related (t-AML) in 37% and secondary after myelodysplastic syndrome (s-AML) in 5%. Cytogenetics revealed a complex karyotype in 43%. Besides MYST3-CREBBP, whole-genome sequencing on a subset of 10 patients revealed recurrent mutations in ASXL1, BRD3, FLT3, MLH1, POLG, TP53, SAMD4B (n = 3, each), EYS, KRTAP9-1 SPTBN5 (n = 4, each), RUNX1 and TET2 (n = 2, each). Complete remission after intensive chemotherapy was achieved in 84%. Median follow-up was 5·48 years; five-year survival rate was 17%. Patients with s-/t-AML (P = 0·01) and those with complex karyotype (P = 0·04) had an inferior prognosis. Allogeneic haematopoietic cell transplantation (allo-HCT) was performed in 21 (36%) patients, including 15 in first complete remission (CR1). Allo-HCT in CR1 significantly improved survival (P = 0·04); multivariable analysis revealed that allo-HCT in CR1 was effective in de novo AML but not in patients with s-AML/t-AML and less in patients exhibiting a complex karyotype. In summary, outcomes of patients with t(8;16) are dismal with chemotherapy, and may be substantially improved with allo-HCT performed in CR1., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.