47 results on '"Wang, Eunice S."'
Search Results
2. Olutasidenib in combination with azacitidine induces durable complete remissions in patients with relapsed or refractory mIDH1 acute myeloid leukemia: a multicohort open-label phase 1/2 trial.
- Author
-
Cortes, Jorge E., Roboz, Gail J., Baer, Maria R., Jonas, Brian A., Schiller, Gary J., Yee, Karen, Ferrell, P. Brent, Yang, Jay, Wang, Eunice S., Blum, William G., Mims, Alice, Tian, Hua, Sheppard, Aaron, de Botton, Stéphane, Montesinos, Pau, Curti, Antonio, and Watts, Justin M.
- Subjects
BLOOD cell count ,HEMATOPOIETIC stem cells ,ACUTE myeloid leukemia ,ERYTHROCYTES ,STEM cell transplantation - Abstract
Background: Olutasidenib is a potent, selective, oral, small molecule inhibitor of mutant IDH1 (mIDH1) which induced durable remissions in high-risk, relapsed/refractory (R/R) mIDH1 AML patients in a phase 1/2 trial. We present a pooled analysis from multiple cohorts of the phase 1/2 trial of patients with R/R AML who received combination olutasidenib and azacitidine therapy. Methods: Adult patients with mIDH1
R132 AML received 150 mg olutasidenib twice daily plus standard-of-care azacitidine (OLU + AZA) and were evaluated for response and safety. Results: Sixty-seven patients with R/R mIDH1R132 AML received combination OLU + AZA. Median age was 66 years (range 28–82) and 54% were male. Most patients (83%) had 2 + prior regimens, including a hypomethylating agent in 40%, IDH1 inhibitor therapy in 31% (olutasidenib in 24%), and hematopoietic stem cell transplant in 10%. Cytogenetic risk was intermediate in 72%, poor in 18% and unknown in 10%. CR/CRh was achieved in 21/67 (31%; 95% CI 21–44) patients, with a median duration of 14.7 months (95% CI 4.6-not reached). CR was achieved in 18/67 (27%; 95% CI 17–39) patients, with median duration of 20.3 months (95% CI 3.7-not reached). Overall response (partial remission or better) was achieved in 34/67 (51%; 95% CI 38–63) patients. Median overall survival was 12.9 months (95% CI 18.7–19.3). In a subset analysis excluding patients who had prior OLU exposure (N = 51), CR/CRh was achieved in 19/51 (37%; 95% CI 24–52) patients, CR was achieved in 16/51 (31%; 95% CI 19–46), and overall response was achieved in 30/51 (59%; 95% CI 44–72). In patients who achieved CR/CRh and were transfusion-dependent at baseline, transfusion independence (RBC and platelets) was achieved in 64% (7/11) and 57% (4/7) of patients, respectively. The most common Grade 3 or 4 adverse events (> 20% patients) were decreased platelet count (37%), red blood cell count (25%), and neutrophil count (24%). Six patients (9%) experienced differentiation syndrome. Four (6%) discontinued treatment due to an adverse event. Conclusions: Olutasidenib plus azacitidine induced high response rates and durable remissions with a tolerable side effect profile in patients with R/R AML with diverse treatment histories. The results represent another therapeutic option for patients with mIDH1 AML who may benefit from a targeted therapy. Trial registration: NCT02719574. [ABSTRACT FROM AUTHOR]- Published
- 2025
- Full Text
- View/download PDF
3. Ziftomenib in relapsed or refractory acute myeloid leukaemia (KOMET-001): a multicentre, open-label, multi-cohort, phase 1 trial.
- Author
-
Wang, Eunice S, Issa, Ghayas C, Erba, Harry P, Altman, Jessica K, Montesinos, Pau, DeBotton, Stephane, Walter, Roland B, Pettit, Kristen, Savona, Michael R, Shah, Mithun Vinod, Kremyanskaya, Marina, Baer, Maria R, Foran, James M, Schiller, Gary, Adès, Lionel, Heiblig, Mael, Berthon, Celine, Peterlin, Pierre, Rodríguez-Arbolí, Eduardo, and Salamero, Olga
- Subjects
- *
ACUTE myeloid leukemia , *CARDIAC arrest , *DEATH rate , *CLINICAL trials , *ADULTS - Abstract
Ziftomenib (KO-539) is an oral selective menin inhibitor with known preclinical activity in menin-dependent acute myeloid leukaemia models. The primary objective of this study was to determine the recommended phase 2 dose in patients with relapsed or refractory acute myeloid leukaemia based on safety, pharmacokinetics, pharmacodynamics, and preliminary activity. KOMET-001 is a multicentre, open-label, multi-cohort, phase 1/2 clinical trial of ziftomenib in adults with relapsed or refractory acute myeloid leukaemia. Results of the phase 1 study, conducted at 22 hospitals in France, Italy, Spain, and the USA, are presented here and comprise the dose-escalation (phase 1a) and dose-validation and expansion (phase 1b) phases. Eligible patients were aged 18 years or older, had relapsed or refractory acute myeloid leukaemia, and had an Eastern Cooperative Oncology Group performance status of 2 or less. For phase 1a, patients (all molecular subtypes) received ziftomenib (50–1000 mg) orally once daily in 28-day cycles. For phase 1b, patients with NPM1 mutations or with KMT2A rearrangements were randomly assigned (1:1) using third-party interactive response technology to two parallel dose cohorts (200 mg and 600 mg ziftomenib). Primary endpoints were maximum tolerated dose or recommended phase 2 dose in phase 1a, and safety, remission rates, and pharmacokinetics supporting recommended phase 2 dose determination in phase 1b. Analyses were performed in all patients who received at least one dose of ziftomenib (modified intention-to-treat population). Phase 1a/1b is complete. This trial is registered with ClinicalTrials.gov , NCT04067336 , and the EU Clinical Trials register, EudraCT 2019-001545-41. From Sept 12, 2019, to Aug 19, 2022, 83 patients received 50–1000 mg ziftomenib (39 [47%] were male and 44 [53%] were female). Median follow-up was 22·3 months (IQR 15·4–30·2). Of 83 patients, the most common grade 3 or worse treatment-emergent adverse events were anaemia (20 [24%]), febrile neutropenia (18 [22%]), pneumonia (16 [19%]), differentiation syndrome (12 [15%]), thrombocytopenia (11 [13%]), and sepsis (ten [12%]). Overall, 68 of 83 patients had serious adverse events, with two reported treatment-related deaths (one differentiation syndrome and one cardiac arrest). Differentiation syndrome rate and severity influenced the decision to halt enrolment of patients with KMT2A rearrangements. In Phase 1b, no responses were reported in patients treated at the 200 mg dose level. At the recommended phase 2 dose of 600 mg, nine (25%) of 36 patients with KMT2A rearrangement or NPM1 mutation had complete remission or complete remission with partial haematologic recovery. Seven (35%) of 20 patients with NPM1 mutation treated at the recommended phase 2 dose had a complete remission. Ziftomenib showed promising clinical activity with manageable toxicity in heavily pretreated patients with relapsed or refractory acute myeloid leukaemia. Phase 2 assessment of ziftomenib combination therapy in the upfront and relapsed or refractory setting is ongoing. Kura Oncology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. A precision medicine classification for treatment of acute myeloid leukemia in older patients
- Author
-
Mims, Alice S., Kohlschmidt, Jessica, Borate, Uma, Blachly, James S., Orwick, Shelley, Eisfeld, Ann-Kathrin, Papaioannou, Dimitrios, Nicolet, Deedra, Mrόzek, Krzysztof, Stein, Eytan, Bhatnagar, Bhavana, Stone, Richard M., Kolitz, Jonathan E., Wang, Eunice S., Powell, Bayard L., Burd, Amy, Levine, Ross L., Druker, Brian J., Bloomfield, Clara D., and Byrd, John C.
- Published
- 2021
- Full Text
- View/download PDF
5. Phase 1 dose escalation study of the MDM2 inhibitor milademetan as monotherapy and in combination with azacitidine in patients with myeloid malignancies.
- Author
-
DiNardo, Courtney D., Olin, Rebecca, Wang, Eunice S., Skikne, Barry, Rosenthal, Joseph, Kumar, Prasanna, Sumi, Hiroyuki, Hizukuri, Yoshiyuki, Hong, Ying, Patel, Parul, Seki, Takahiko, Duan, Tao, Lesegretain, Arnaud, and Andreeff, Michael
- Subjects
MYELODYSPLASTIC syndromes ,ACUTE myeloid leukemia ,HEMATOLOGIC malignancies ,MOUSE leukemia ,POLYMERASE chain reaction - Abstract
Background: Mouse double minute‐2 homolog (MDM2) plays a key role in downregulating p53 activity in hematologic malignancies, and its overexpression is associated with poor outcomes. Methods: This phase 1 study assessed the safety and efficacy of different dosing regimens of the MDM2 inhibitor milademetan as monotherapy and in combination with azacitidine (AZA) in patients with relapsed or refractory acute myeloid leukemia or high‐risk myelodysplastic syndromes. Results: Seventy‐four patients (monotherapy, n = 57; milademetan‐AZA combination, n = 17) were treated. The maximum tolerated dose of milademetan was 160 mg once daily given for the first 14–21 days of 28‐day cycles as monotherapy and on Days 5–14 in combination with AZA. Dose‐limiting toxicities were gastrointestinal, fatigue, or renal/electrolyte abnormalities. Treatment‐emergent adverse events related to milademetan occurred in 82.5% and 64.7% of participants in the monotherapy and AZA combination arms, respectively. Two participants (4.2%) in the monotherapy arm achieved complete remission (CR), and 1 (2.1%) achieved CR with incomplete blood count recovery (CRi). Two participants (13.3%) achieved CRi in the combination arm. New TP53 mutations, detected only during milademetan monotherapy, were found pre‐existing below standard detection frequency by droplet digital polymerase chain reaction. Interpretation: Milademetan was relatively well tolerated in this population; however, despite signals of activity, clinical efficacy was minimal. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Prognostic and biological significance of the proangiogenic factor EGFL7 in acute myeloid leukemia
- Author
-
Papaioannou, Dimitrios, Shen, Changxian, Nicolet, Deedra, McNeil, Betina, Bill, Marius, Karunasiri, Malith, Burke, Matthew H., Ozer, Hatice Gulcin, Yilmaz, Selen A., Zitzer, Nina, Behbehani, Gregory K., Oakes, Christopher C., Steiner, Damian J., Marcucci, Guido, Powell, Bayard L., Kolitz, Jonathan E., Carter, Thomas H., Wang, Eunice S., Mrózek, Krzysztof, Croce, Carlo M., Caligiuri, Michael A., Bloomfield, Clara D., Garzon, Ramiro, and Dorrance, Adrienne M.
- Published
- 2017
7. Prevention, recognition, and management of adverse events associated with gemtuzumab ozogamicin use in acute myeloid leukemia
- Author
-
Cortes, Jorge E., de Lima, Marcos, Dombret, Hervé, Estey, Elihu H., Giralt, Sergio A., Montesinos, Pau, Röllig, Christoph, Venditti, Adriano, and Wang, Eunice S.
- Published
- 2020
- Full Text
- View/download PDF
8. Acute myeloid leukemia with LRRFIP1::FGFR1 rearrangement and a complex karyotype.
- Author
-
Qian, You-Wen, Wang, Eunice S., Sait, Sheila Jani, and Glenn, Sean T.
- Subjects
- *
KARYOTYPES , *ACUTE myeloid leukemia , *CHROMOSOME analysis , *FLUORESCENCE in situ hybridization , *NUCLEOTIDE sequencing , *GENETIC mutation - Abstract
We report a case of a 20-year-old man who presented with splenomegaly, hyperleukocytosis, anemia, and thrombocytopenia. A diagnosis of acute myeloid leukemia (AML) with LRRFIP1::FGFR1 rearrangement with complex karyotype was determined. Chromosome analysis showed a male karyotype: 46,XY,i(1)(q10),t(2;8)(q37;p11.2),der(5)t(1;5) (p22;q13)[17]46,XY [3]. Fluorescence in situ hybridization (FISH) analysis using the Cytocell FGFR1 break apart/amplification probe detected FGFR1 rearrangement with t(2:8) in 126/200 cells analyzed. Other FISH probes including 1p36/ 1q25 probes, del(5q) deletion probe, TLX3 break apart probe, and PDGFRB break apart probe were also utilized to confirm the other karyotypic abnormalities. Next-generation sequencing (NGS) SureSelectXT Custom DNA Target Somatic Detection detected RUNX1 gene mutation. NGS Archer FusionPlex (RNA) confirmed the LRRFIP1::FGFR1 rearrangement. This is the second reported case of AML with LRRFIP1::FGFR1 rearrangement and the first with a complex karyotype. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Spliceosome mutations are associated with clinical response in a phase 1b/2 study of the PLK1 inhibitor onvansertib in combination with decitabine in relapsed or refractory acute myeloid leukemia.
- Author
-
Croucher, Peter J P, Ridinger, Maya, Becker, Pamela S., Lin, Tara L., Silberman, Sandra L., Wang, Eunice S., and Zeidan, Amer M.
- Subjects
DECITABINE ,ACUTE myeloid leukemia ,GENE expression ,GENOMICS ,GENETIC mutation ,BONE marrow - Abstract
PLK1 is overexpressed in acute myeloid leukemia (AML). A phase 1b trial of the PLK1 inhibitor onvansertib (ONV) combined with decitabine (DAC) demonstrated initial safety and efficacy in patients with relapsed/refractory (R/R) AML. The current study aimed to identify molecular predictors of response to ONV + DAC in R/R AML patients. A total of 44 R/R AML patients were treated with ONV + DAC and considered evaluable for efficacy. Bone marrow (BM) samples were collected at baseline for genomic and transcriptomic analysis (n = 32). A 10-gene expression signature, predictive of response to ONV + DAC, was derived from the leading-edge genes of gene set enrichment analyses (GSEA). The gene signature was evaluated in independent datasets and used to identify associated mutated genes. Twenty percent of the patients achieved complete remission, with or without hematologic count recovery (CR/CRi), and 32% exhibited a ≥50% reduction in bone marrow blasts. Patients who responded to treatment had elevated mitochondrial function and OXPHOS. The gene signature was not associated with response to DAC alone in an independent dataset. By applying the signature to the BeatAML cohort (n = 399), we identified a positive association between predicted ONV + DAC response and mutations in splicing factors (SF). In the phase 1b/2 trial, patients with SF mutations (SRSF2, SF3B1) had a higher CR/CRi rate (50%) compared to those without SF mutations (9%). PLK1 inhibition with ONV in combination with DAC could be a potential therapy in R/R AML patients, particularly those with high OXPHOS gene expression and SF mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Gemtuzumab ozogamicin plus standard induction chemotherapy improves outcomes of newly diagnosed intermediate cytogenetic risk acute myeloid leukemia.
- Author
-
Awada, Hassan, Abdelmalek, Mina, Cronin, Tara, Baron, Jeffrey, Kashour, Zakariya, Azad, Farhan, Faisal, Muhammad Salman, Faber, Mark, Gravina, Matthew, Sung, Pamela J., Green, Steven D., Przespolewski, Amanda, Thompson, James E., Griffiths, Elizabeth A., and Wang, Eunice S.
- Subjects
HEPATIC veno-occlusive disease ,ACUTE myeloid leukemia ,INDUCTION chemotherapy - Abstract
To the Editor: Gemtuzumab ozogamicin (GO) is a CD33-directed antibody-drug conjugate approved for the treatment of newly diagnosed (ND) and refractory/relapsed CD33-positive acute myeloid leukemia (AML) [[1]-[6]]. A meta-analysis of individual patient data from 5 randomized controlled trials ( I n i = 3325) confirmed a significant survival benefit of GO added to intensive induction chemotherapy for patients with AML without adverse cytogenetics [[7]]. While patients with favorable cytogenetic ND-AML, specifically core-binding factor AML (CBF-AML), particularly benefited with a 5-year overall survival improvement of 20.7% (log-rank I p i = 0.0006), patients with intermediate cytogenetic risk AML also showed a significant 5-year survival improvement of 5.7% ( I p i = 0.005). [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
11. Development of a Preclinical PK/PD Model to Assess Antitumor Response of a Sequential Aflibercept and Doxorubicin-Dosing Strategy in Acute Myeloid Leukemia
- Author
-
Fetterly, Gerald J., Aras, Urvi, Lal, Deepika, Murphy, Michael, Meholick, Patricia D., and Wang, Eunice S.
- Published
- 2013
- Full Text
- View/download PDF
12. Is obesity a prognostic factor for acute myeloid leukemia outcome?
- Author
-
Lee, Hun Ju, Licht, Andrea S., Hyland, Andrew J., Ford, Laurie A., Sait, Sheila N. J., Block, AnneMarie W., Barcos, Maurice, Baer, Maria R., Wang, Eunice S., and Wetzler, Meir
- Published
- 2012
- Full Text
- View/download PDF
13. Can decreasing smoking prevalence reduce leukemia mortality?
- Author
-
Varadarajan, Ramya, Cummings, Michael K., Hyland, Andrew J., Wang, Eunice S., and Wetzler, Meir
- Published
- 2010
- Full Text
- View/download PDF
14. Targeting acute myeloid leukemia through multimodal immunotherapeutic approaches.
- Author
-
Przespolewski, Amanda C., Portwood, Scott, and Wang, Eunice S.
- Subjects
ACUTE myeloid leukemia ,REGULATORY T cells ,TYPE I interferons ,IMMUNE checkpoint inhibitors ,HEMATOLOGIC malignancies ,PRELEUKEMIA ,PEMBROLIZUMAB - Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with a dismal prognosis. Immunotherapeutic approaches using single agent checkpoint inhibitors have thus far shown limited success. We hypothesized that successful adaptive anti-AML specific immune responses require additional modulation of innate immunity. DMXAA exposure resulted in modest apoptosis of C1498 AML cells with a subtle increase in PD-L1 expression and limited production of IL-6 and IFN-β. In contrast, DMXAA + anti-PD-1 ab, but not either agent alone, significantly decreased in vivo disease burden and prolonged overall survival in C1498 engrafted leukemic mice. Combination-treated mice demonstrated increased memory T-cells and mature dendritic cells, lower numbers of regulatory T-cells and evidence of leukemia apoptosis. Furthermore, these effects were associated with markedly increased serum levels of type I interferon (IFN) and IFN gamma. We demonstrate that combining an innate immune agonist with a checkpoint inhibitor synergistically improved anti-tumor activity in a preclinical AML model. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
15. Menin Inhibitors in Acute Myeloid Leukemia-What Does the Future Hold?
- Author
-
Swaminathan, Mahesh, Bourgeois, Wallace, Armstrong, Scott A., and Wang, Eunice S.
- Abstract
Abstract: Menin inhibitors constitute a novel class of agents targeting the underlying biology of nucleophosmin (NPM1) mutant and KMT2A (formerly known as MLL1) rearranged (KMT2Ar) acute leukemias. KMT2Ar acute leukemias constitute 5% to 10% of acute leukemias, and NPM1 mutations are identified in 30% of newly diagnosed acute myeloid leukemias (AMLs). In preclinical AML models, small molecule inhibitors of the menin-KMT2A protein-protein interaction induce differentiation, downregulate critical gene expression programs, and confer a survival advantage in patient-derived xenograft models of NPM1 mutant and KMT2Ar AML. Multiple clinical trials evaluating oral menin inhibitors in acute leukemias are ongoing. Preliminary results in relapsed/refractory NPM1 mutant and KMT2Ar AML have shown on-target effects, tolerable toxicity, and promising clinical activity. This review details the current clinical experience of menin inhibitors in AML and discusses how these agents can be successfully integrated into future therapeutic approaches. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
16. Emerging trends of therapy related myeloid neoplasms following modern cancer therapeutics in the United States.
- Author
-
Singh, Abhay, Herr, Megan M., Griffiths, Elizabeth A., Przespolewski, Amanda, Faber, Mark G., Mrad, Chebli, Wang, Eunice S., Hahn, Theresa, and Thota, Swapna
- Subjects
NON-small-cell lung carcinoma ,RENAL cell carcinoma ,THERAPEUTICS ,TUMORS ,ACUTE myeloid leukemia ,HEMATOPOIESIS - Abstract
Clonal hematopoiesis (CH) is a risk factor for the development of therapy-related myelodysplastic syndromes (tMDS) and acute myeloid leukemia (tAML). Adoption of targeted-immunotherapeutics since 2011, may alter the risk of CH progression to tMDS/AML. To study this, we evaluated risk of tMDS and tAML in 667 588 ≥ 1-year survivors of non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), melanoma and multiple-myeloma (MM) diagnosed during: 2000–2005, 2006–2010 and 2011–2016. The risk of tMDS increased significantly after NSCLC across all time periods (P
trend = 0.002) while tAML risk decreased from 2006–2010 to 2011–2016, coinciding with increasing use of non-chemotherapeutic agents. tAML risk after RCC decreased (Ptrend = 0.007) whereas tMDS risk did not significantly change over time. After melanoma, tMDS and tAML risks were similar to the general population. tMDS and tAML risk after MM increased from the first to second time-period, however, only risk of tMDS decreased during last period. We report diverging trends in the risk of tAML and tMDS after adoption of modern cancer therapies for specific cancers. It is imperative to further explore impact of contemporary treatment strategies on clonal evolution. Modern treatments via their discrete mechanism of actions on pre-existing CH may alter the risk of subsequent tMDS and tAML. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
17. A prospective biomarker analysis of alvocidib followed by cytarabine and mitoxantrone in MCL-1-dependent relapsed/refractory acute myeloid leukemia.
- Author
-
Zeidner, Joshua F., Lin, Tara L., Vigil, Carlos E., Fine, Gil, Yair Levy, M., Nazha, Aziz, Esteve, Jordi, Lee, Daniel J., Yee, Karen, Dalovisio, Andrew, Wang, Eunice S., Bergua Burgues, Juan M., Schriber, Jeffrey, Litzow, Mark R., Frankfurt, Olga, Castillo, Teresa Bernal Del, Bhatt, Vijaya Raj, Bhatnagar, Bhavana, Mehta, Priyanka, and Dillon, Richard
- Subjects
ACUTE myeloid leukemia ,CELL death ,BIOMARKERS ,CYTARABINE ,MITOXANTRONE - Abstract
Notably, refractory AML patients with MCL-1 dependence had a CRc rate of 52% compared with 38% in refractory AML patients without MCL-1 dependence (Supplemental Fig. A comparison of clinical outcomes among patients with MCL-1-dependent R/R AML and non-MCL-1-dependent R/R AML patients is outlined in Supplemental Tables 3-5. Eighty-two patients were enrolled onto one of the five cohorts: (1) MCL-1 <15% ( I n i = 15), (2) MCL-1: 15-<30% ( I n i = 14), (3) MCL-1: 30-<40% ( I n i = 14), (4) MCL-1: >=40% ( I n i = 25), and (5) NDHR AML with MCL-1 >=40% ( I n i = 14). [Extracted from the article]
- Published
- 2021
- Full Text
- View/download PDF
18. Cladribine, cytarabine, and GCSF with and without mitoxantrone (CLAG ± M) is highly effective for poor risk acute myeloid leukemia with adverse karyotype and prior hypomethylating therapy.
- Author
-
Przespolewski, Amanda, Muppidi, Monica R., Freyer, Craig W., Ji, Wenyan, Cronin, Tara L., Thota, Swapna, Griffiths, Elizabeth A., Thompson, James E., Ontiveros, Evelena P., Baron, Jeffrey, Elshoury, Amro, and Wang, Eunice S.
- Subjects
ACUTE myeloid leukemia ,MITOXANTRONE ,KARYOTYPES ,CYTARABINE ,PROGNOSIS - Abstract
Forty-seven (73.3%) had r/rAML with a median of 2 prior AML therapies (range 1-5), and 33 patients (62.3%) received prior HMA (see Supplementary Data for details of previous regimens). The regimen of cladribine, cytarabine, and filgrastrim with mitoxantrone (CLAG-M, GCLAM) has shown significant promise for upfront therapy of secondary acute myeloid leukemia (now known as AML with myelodysplastic related changes (AML-MRC)) [[1]] and relapsed/refractory AML (r/rAML) with high reported response rates in multiple prospective trials [[2]]. Cladribine, cytarabine, and GCSF with and without mitoxantrone (CLAG ± M) is highly effective for poor risk acute myeloid leukemia with adverse karyotype and prior hypomethylating therapy Overall response rate (ORR = CR + CRi + PR) following one cycle of CLAG ± M induction was 80.3% ( I n i = 49) with CR in 20 patients (32.8%) and CRi in 21 (34.4%) patients, respectively (Table 1). [Extracted from the article]
- Published
- 2021
- Full Text
- View/download PDF
19. Novel therapies for AML: a round-up for clinicians.
- Author
-
Swaminathan, Mahesh and Wang, Eunice S.
- Subjects
ACUTE myeloid leukemia ,EPIGENETICS ,ANTIBODY-drug conjugates ,MEDICAL literature ,SMALL molecules ,TARGETED drug delivery - Abstract
Introduction: Acute myeloid leukemia (AML) is a deadly disease associated with poor outcomes. For over four decades, therapeutic options for AML were limited to high dose cytotoxic chemotherapy. Scientific breakthroughs have not only enhanced our understanding of the molecular underpinnings of this disease but also resulted in the development of several targeted therapies with superior efficacy and lesser toxicities than conventional chemotherapy. The FDA approval of small molecule inhibitors for specific AML subsets highlights the importance of genetic and molecular profiling to optimally personalize AML therapy in the modern era. Areas covered: In this article, we review the medical literature from PubMed on recent FDA approved drugs for AML by their mechanism of action: small molecule inhibitors, antibody-drug conjugate, cytotoxic, and epigenetic agents. We describe how to incorporate these agents into the current treatment paradigm for specific AML patients. Expert opinion: Knowing the molecular characteristics of patients with AML is of utmost importance to plan the best management. There are promising drugs targeting leukemogenesis by various mechanisms. It is important to consider clinical trial options for patients if and when available. We have provided a brief overview of the most promising agents on the horizon for AML therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. Favorable outcomes of acute leukemias of ambiguous lineage treated with hyperCVAD: a multi-center retrospective study.
- Author
-
Duong, Vu H., Begna, Kebede H., Kashanian, Sarah, Sweet, Kendra, Wang, Eunice S., Caddell, Ryan, Shafer, Danielle A., Singh, Zeba N., Baer, Maria R., and Al-Kali, Aref
- Subjects
ACUTE leukemia ,HEMATOPOIETIC stem cell transplantation ,LYMPHOBLASTIC leukemia ,HEMATOLOGIC malignancies ,ACUTE myeloid leukemia - Abstract
Acute leukemias of ambiguous lineage (ALAL) are rare hematologic malignancies with poor outcomes. Retrospective studies have suggested that acute lymphoblastic leukemia (ALL) regimens are more effective than acute myeloid leukemia (AML) regimens. We retrospectively examined the effectiveness of the widely-used adult ALL regimen hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (hyperCVAD) as initial therapy in patients with ALAL at five academic institutions. Twenty-five patients were identified, including 23 with mixed phenotype acute leukemia (MPAL) and two with acute undifferentiated leukemia. Five of 8 tested (63%) had FLT3-ITD and 3 of 25 (12%) were Philadelphia chromosome-positive. The complete remission (CR) rate was 76%, with CR with incomplete count recovery (CRi) in an additional 8%, for an overall response rate of 84%. Median number of cycles to CR/CRi was 1. There were no deaths in the first 30 days. Of the 21 patients achieving CR or CRi, 14 (66%) proceeded to allogeneic hematopoietic stem cell transplantation. With a median follow-up time of 31.6 months, median overall survival for the entire cohort was not reached, and the estimated 2-year survival was 63%. HyperCVAD can be considered an effective and tolerable front-line regimen for patients with ALAL, and warrants further prospective study. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
21. Safety of gemtuzumab ozogamicin as monotherapy or combination therapy in an expanded-access protocol for patients with relapsed or refractory acute myeloid leukemia.
- Author
-
Wang, Eunice S., Aplenc, Richard, Chirnomas, Deborah, Dugan, Michael, Fazal, Salman, Iyer, Swaminathan, Lin, Tara L., Nand, Sucha, Pierce, Kristen J., Shami, Paul J., Vermette, Jennifer J., and Abboud, Camille N.
- Subjects
- *
ACUTE myeloid leukemia , *ACUTE promyelocytic leukemia , *MYELODYSPLASTIC syndromes , *TERMINATION of treatment , *HEPATIC veno-occlusive disease - Abstract
Gemtuzumab ozogamicin (GO) remained available to US clinicians through an open-label expanded-access protocol (NCT02312037) until GO was reapproved. Patients were aged ≥3 months with relapsed/refractory (R/R) acute myeloid leukemia (AML), high-risk myelodysplastic syndrome, or acute promyelocytic leukemia (APL), and had exhausted other treatment options. Three hundred and thirty one patients received GO as monotherapy for R/R AML (n = 139), combination therapy for R/R AML (n = 183), or treatment for R/R APL (n = 9). Corresponding treatment discontinuations occurred in 68, 39, and 33% of patients. All-causality grade 5 AEs occurred in 52, 22, and 22% of patients in the monotherapy, combination, and APL groups, respectively. Corresponding grades 3 and 4 treatment-related AEs were reported in 60, 55 and 78% of patients. Hepatotoxicity occurred in five patients: veno-occlusive disease (n = 4) and drug-induced liver injury (n = 1). GO was generally well tolerated in patients with R/R AML or APL. Most frequent treatment-related grade ≥3 AEs were hematologic AEs. Clinicaltrials.gov identifier: NCT02312037 [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. Incorporating FLT3 inhibitors in the frontline treatment of FLT3 mutant acute myeloid leukemia.
- Author
-
Wang, Eunice S.
- Abstract
FLT3 mutations occur in up to a third of newly diagnosed patients with acute myeloid leukemia (AML) and confer poor prognosis. Clinical development of FLT3 tyrosine kinase inhibitors for AML initially involved broad-spectrum inhibitors (midostaurin, sorafenib) targeting multiple kinases. Addition of midostaurin to upfront intensive chemotherapy for younger patients with FLT3 mutant AML significantly improved overall survival and validated FLT3 as a therapeutic target. Other regimens such as sorafenib and hypomethylating agents (azacitidine, decitabine) have expanded the use of FLT3 inhibitors to other populations with FLT3 mutant AML. However, emerging data on new highly potent and specific FLT3 inhibitors such as quizartinib, gilteritinib, and crenolanib suggest that these agents may soon supplant midostaurin and sorafenib in the upfront setting. Using case presentations, this review provides guidelines and practical management strategies for frontline therapy of patients with newly diagnosed FLT3 mutant AML in the current era. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
23. Advancing treatment of acute myeloid leukemia: the future of FLT3 inhibitors.
- Author
-
Elshoury, Amro, Przespolewski, Amanda, Baron, Jeffrey, and Wang, Eunice S.
- Abstract
Introduction: Mutations of the FLT3 gene are among most common genetic abnormalities occurring in acute myeloid leukemia (AML) and are associated with dismal prognosis. Tremendous effort has been devoted to developing clinically effective FLT3 inhibitors. First generation inhibitors consisted of multi-kinase inhibitors (sorafenib, lestaurtinib, midostaurin), which blocked FLT3 as well as multiple other kinase receptors. The failure of these agents to induce durable responses led to the development of second generation FLT3 tyrosine kinase inhibitors (quizartinib, crenolinib, gilteritinib) exhibiting high potency and specificity for mutant FLT3 kinases and sustained in vivo FLT3 inhibition. These myriad FLT3 inhibitors possess diverse kinase inhibitory properties, toxicity profiles, and pharmacokinetics, which impact on their incorporation into therapeutic regimens. Areas covered: This article reviews the medical literature on current and future FLT3 inhibitors for AML therapy. We provide algorithms for which kinase inhibitor should be utilized for different FLT3 mutations (ITD±TKD) and clinical scenarios (de novo, relapsed/refractory, fit vs. unfit) and discuss novel FLT3 targeted therapeutic approaches. Expert commentary: Integration of clinically active FLT3 inhibitors into all stages of therapy for all individuals with FLT3 mutant AML promises to significantly improve outcomes for this poor prognosis disease. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
24. Gemtuzumab ozogamicin for the treatment of acute myeloid leukemia.
- Author
-
Baron, Jeffrey and Wang, Eunice S.
- Subjects
ACUTE myeloid leukemia treatment ,ANTIBODY-drug conjugates ,MONOCLONAL antibody biotechnology ,ANTINEOPLASTIC agents ,CALICHEAMICIN ,ACUTE myeloid leukemia diagnosis - Abstract
Introduction: Gemtuzumab ozogamicin (GO) is an antibody-drug conjugate consisting of a monoclonal antibody targeting CD33 linked to a cytotoxic derivative of calicheamicin. Despite the known clinical efficacy in relapsed/refractory acute myeloid leukemia (AML), GO was withdrawn from the market in 2010 due to increased early deaths witnessed in newly diagnosed AML patients receiving GO + intensive chemotherapy. In 2017, new data on the clinical efficacy and safety of GO administered on a fractionated-dosing schedule led to re-approval for newly diagnosed and relapsed/refractory AML. Areas covered: Addition of fractionated GO to chemotherapy significantly improved event-free survival of newly diagnosed AML patients with favorable and intermediate cytogenetic-risk disease. GO monotherapy also prolonged survival in newly diagnosed unfit patients and relapse-free survival in relapsed/refractory AML. This new dosing schedule was associated with decreased incidence of hepatotoxicity, veno-occlusive disease, and early mortality. Expert commentary: GO represents the first drug-antibody conjugate approved (twice) in the United States for AML. Its re-emergence adds a valuable agent back into the armamentarium for AML. The approval of GO as well as three other agents for AML in 2017 highlights the need for rapid cytogenetic and molecular characterization of AML and incorporation into new treatment algorithms. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
25. Low 25(OH) vitamin D3 levels are associated with adverse outcome in newly diagnosed, intensively treated adult acute myeloid leukemia.
- Author
-
Lee, Hun Ju, Muindi, Josephia R, Tan, Wei, Hu, Qiang, Wang, Dan, Liu, Song, Wilding, Gregory E, Ford, Laurie A, Sait, Sheila N J, Block, Annemarie W, Adjei, Araba A, Barcos, Maurice, Griffiths, Elizabeth A, Thompson, James E, Wang, Eunice S, Johnson, Candace S, Trump, Donald L, and Wetzler, Meir
- Subjects
CANCER relapse ,CELL receptors ,GENES ,GENETIC polymorphisms ,PROGNOSIS ,RESEARCH funding ,RNA ,VITAMIN D ,CHOLECALCIFEROL ,ACUTE myeloid leukemia ,TREATMENT effectiveness ,GENE expression profiling - Abstract
Background: Several studies have suggested that low 25(OH) vitamin D3 levels may be prognostic in some malignancies, but no studies have evaluated their impact on treatment outcome in patients with acute myeloid leukemia (AML).Methods: Vitamin D levels were evaluated in 97 consecutive, newly diagnosed, intensively treated patients with AML. MicroRNA expression profiles and single nucleotide polymorphisms (SNPs) in the 25(OH) vitamin D3 pathway genes were evaluated and correlated with 25(OH) vitamin D3 levels and treatment outcome.Results: Thirty-four patients (35%) had normal 25(OH) vitamin D3 levels (32-100 ng/mL), 34 patients (35%) had insufficient levels (20-31.9 ng/mL), and 29 patients (30%) had deficient levels (<20 ng/mL). Insufficient/deficient 25(OH) vitamin D3 levels were associated with worse relapse-free survival (RFS) compared with normal vitamin D3 levels. In multivariate analyses, deficient 25(OH) vitamin D3 , smoking, European Leukemia Network genetic group, and white blood cell count retained their statistical significance for RFS. Several microRNAs and SNPs were associated with 25(OH) vitamin D3 levels, although none remained significant after multiple test corrections; one 25(OH) vitamin D3 receptor SNP, rs10783219, was associated with a lower complete remission rate (P = .0442) and with shorter RFS (P = .0058) and overall survival (P = .0011).Conclusions: It remains to be determined what role microRNA and SNP profiles play in contributing to low 25(OH) vitamin D3 level and/or outcome and whether supplementation will improve outcomes for patients with AML. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
26. Low 25(OH) vitamin D3 levels are associated with adverse outcome in newly diagnosed, intensively treated adult acute myeloid leukemia.
- Author
-
Lee, Hun Ju, Muindi, Josephia R., Tan, Wei, Hu, Qiang, Wang, Dan, Liu, Song, Wilding, Gregory E., Ford, Laurie A., Sait, Sheila N. J., Block, Annemarie W., Adjei, Araba A., Barcos, Maurice, Griffiths, Elizabeth A., Thompson, James E, Wang, Eunice S., Johnson, Candace S., Trump, Donald L., and Wetzler, Meir
- Subjects
ACUTE myeloid leukemia treatment ,CHOLECALCIFEROL ,TREATMENT effectiveness ,MICRORNA ,GENE expression ,SINGLE nucleotide polymorphisms - Abstract
BACKGROUND Several studies have suggested that low 25(OH) vitamin D
3 levels may be prognostic in some malignancies, but no studies have evaluated their impact on treatment outcome in patients with acute myeloid leukemia (AML). METHODS Vitamin D levels were evaluated in 97 consecutive, newly diagnosed, intensively treated patients with AML. MicroRNA expression profiles and single nucleotide polymorphisms (SNPs) in the 25(OH) vitamin D3 pathway genes were evaluated and correlated with 25(OH) vitamin D3 levels and treatment outcome. RESULTS Thirty-four patients (35%) had normal 25(OH) vitamin D3 levels (32-100 ng/mL), 34 patients (35%) had insufficient levels (20-31.9 ng/mL), and 29 patients (30%) had deficient levels (<20 ng/mL). Insufficient/deficient 25(OH) vitamin D3 levels were associated with worse relapse-free survival (RFS) compared with normal vitamin D3 levels. In multivariate analyses, deficient 25(OH) vitamin D3 , smoking, European Leukemia Network genetic group, and white blood cell count retained their statistical significance for RFS. Several microRNAs and SNPs were associated with 25(OH) vitamin D3 levels, although none remained significant after multiple test corrections; one 25(OH) vitamin D3 receptor SNP, rs10783219, was associated with a lower complete remission rate ( P = .0442) and with shorter RFS ( P = .0058) and overall survival ( P = .0011). CONCLUSIONS It remains to be determined what role microRNA and SNP profiles play in contributing to low 25(OH) vitamin D3 level and/or outcome and whether supplementation will improve outcomes for patients with AML. Cancer 2014;120:521-529. © 2013 American Cancer Society. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
27. CD19 expression in acute leukemia is not restricted to the cytogenetically aberrant populations.
- Author
-
Francis, Jawad, Dharmadhikari, Avinash V., Sait, Sheila N. J., Deeb, George, Wallace, Paul K., Thompson, James E., Wang, Eunice S., and Wetzler, Meir
- Subjects
ACUTE leukemia ,HUMAN cytogenetics ,GENE expression ,LYMPHOID tissue ,ABERRANT crypt foci ,CLONE cells ,GENETICS - Abstract
Aberrant expression of the B lymphoid marker, CD19, in acute myeloid leukemia (AML) has frequently been associated with t(8;21)(q22;q22). However, AML cases lacking t(8;21) may occasionally express CD19. We asked whether CD19 expression is restricted to the karyotypically abnormal leukemic cells in primary leukemia samples. We compared, by fluorescence in situ hybridization, CD19-positive and CD19-negative cells from nine patients with acute leukemia: three non-t(8;21) AML, three t(8;21) AML and three cases of acute lymphoblastic leukemia. There were no significant differences in karyotypic pattern between the CD19-positive and CD19-negative leukemic cells, raising the concern that therapeutically targeting CD19 for acute leukemia may not eradicate all malignant clones. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
28. Image cytometry-based detection of aneuploidy by fluorescence in situ hybridization in suspension.
- Author
-
Minderman, Hans, Humphrey, Kristen, Arcadi, Jane K., Wierzbicki, Andrzej, Maguire, Orla, Wang, Eunice S., Block, AnneMarie W., Sait, Sheila N. J., George, Thaddeus C., and Wallace, Paul K.
- Abstract
Cytogenetic abnormalities are important diagnostic and prognostic criteria for hematologic malignancies. Karyotyping and fluorescence in situ hybridization (FISH) are the conventional methods by which these abnormalities are detected. The sensitivity of these microscopy-based methods is limited by the abundance of the abnormal cells in the samples and therefore these analyses are commonly not applicable to minimal residual disease (MRD) stages. A flow cytometry-based imaging approach was developed to detect chromosomal abnormalities following FISH in suspension (FISH-IS), which enables the automated analysis of several log-magnitude higher number of cells compared with the microscopy-based approaches. This study demonstrates the applicability of FISH-IS for detecting numerical chromosome aberrations, establishes accuracy, and sensitivity of detection compared with conventional FISH, and feasibility to study procured clinical samples of acute myeloid leukemia (AML). Male and female healthy donor peripheral blood mononuclear cells hybridized with combinations of chromosome enumeration probes (CEP) 8, X, and Y served as models for disomy, monosomy, and trisomy. The sensitivity of detection of monosomies and trisomies amongst 20,000 analyzed cells was determined to be 1% with a high level of precision. A high correlation ( R
2 = 0.99) with conventional FISH analysis was found based on the parallel analysis of diagnostic samples procured from 10 AML patients with trisomy 8 (+8). Additionally, FISH-IS analysis of samples procured at the time of clinical remission demonstrated the presence of residual +8 cells indicating that this approach may be used to detect MRD and associated chromosomal defects. © 2012 International Society for Advancement of Cytometry [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
29. Phase 1 trial of linifanib (ABT-869) in patients with refractory or relapsed acute myeloid leukemia.
- Author
-
Wang, Eunice S., Yee, Karen, Koh, Liang Piu, Hogge, Donna, Enschede, Sari, Carlson, Dawn M., Dudley, Matthew, Glaser, Keith, McKeegan, Evelyn, Albert, Daniel H., Li, Xiaohui, Pradhan, Rajendra, and Stock, Wendy
- Subjects
- *
MYELOID leukemia , *NEUTROPENIA , *CYTOKINES , *FEBRILE neutropenia , *PROTEIN-tyrosine kinases - Abstract
Linifanib, a potent oral inhibitor of fms-like tyrosine kinase 3 (FLT3) and vascular endothelial growth factor receptor tyrosine kinases, has demonstrated promising preclinical single-agent and synergistic anti-leukemic activity in combination with cytarabine. In this phase 1, multicenter, open-label, dose-escalation study, 45 adults with relapsed/refractory acute myeloid leukemia (AML) received linifanib alone in arm A ( n = 29) and linifanib plus intermediate-dose cytarabine in arm B ( n = 16). Median treatment duration was 21 days (range 5-110). Linifanib was well tolerated overall. The most common grade 3/4 events were fatigue (arm A) and febrile neutropenia (arm B). The recommended phase 2 dose was 15 mg (alone), and 10 mg (with cytarabine). Evidence of on-target kinase inhibition in patients with FLT3-mutant and wild-type AML was seen. Decreased phosphorylated FLT3 was seen in 3/3 patients with FLT3-internal tandem duplication (ITD) with peripheral blast reductions and in 8/24 (33%) patients with wild-type, D835 or unknown FLT3 mutation. Eight/29 (28%) patients had decreased phosphorylated extracellular signal-regulated kinase (ERK). [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
30. Characterization of vancomycin pharmacokinetics in the adult acute myeloid leukemia population.
- Author
-
Jarkowski, Anthony, Forrest, Alan, Sweeney, Richard P, Tan, Wei, Segal, Brahm H, Almyroudis, Nikolaos, Wang, Eunice S, and Wetzler, Meir
- Subjects
NEPHROTOXICOLOGY ,CANCER patients ,CANCER treatment ,CREATININE ,DRUG monitoring ,DOSE-effect relationship in pharmacology ,KIDNEYS ,PHARMACEUTICAL arithmetic ,SCALES (Weighing instruments) ,TIME ,GRAM-positive bacterial infections ,VANCOMYCIN ,SPECIALTY hospitals ,MYELOID leukemia ,DESCRIPTIVE statistics ,STANDARDS ,PREVENTION - Abstract
Current vancomycin dosing guidelines in our acute myeloid leukemia population too often achieve suboptimal initial drug concentrations. Our aim was to assess vancomycin pharmacokinetic parameters in acute myeloid leukemia patients and develop an improved dosing equation to attain more accurate initial therapeutic trough levels. Acute myeloid leukemia patients receiving vancomycin for a presumed or documented gram positive infection were eligible. Patients hospitalized in the intensive care unit, those with creatinine clearance <30 mL/min or with limb amputation were excluded. Three samples were collected at the following post-infusion time ranges: 1 h, 3–8 h, and 8–24 h post-infusion, contingent on the dosing interval. Pharmacokinetic data were then fit using a Bayesian-based population pharmacokinetic model. A total of 25 acute myeloid leukemia patients were studied with a mean volume in the central compartment (Vc; L/65 kg), volume of distribution at steady state (Vss; L/65 kg) and distributional clearance (CLd; L/h/65 kg) of 15, 38.9, and 9.32, respectively. CLslope was 0.59 (mg of vancomycin clearance per unit of creatinine clearance in mL/min); this value is 21.4% lower than the established literature value (0.75). The derived equation, based upon these values, was reasonably precise at achieving the desired trough concentration using a priori dosing. The mean (CV%) of the achieved trough was 94% (29%) with a range of 66–188%; 3/25 at <75% and >125%]. We have established that the derived dosing equation can place ∼75% of adult acute myeloid leukemia patients at vancomycin trough levels within 75–125% of the target trough level. [ABSTRACT FROM PUBLISHER]
- Published
- 2012
- Full Text
- View/download PDF
31. Phase 1 study of arsenic trioxide, high-dose cytarabine, and idarubicin to down-regulate constitutive signal transducer and activator of transcription 3 activity in patients aged <60 years with acute myeloid leukemia.
- Author
-
Wetzler, Meir, Andrews, Chris, Ford, Laurie A., Tighe, Sheila, Barcos, Maurice, Sait, Sheila N. J., Block, AnneMarie W., Nowak, Norma J., Baer, Maria R., Wang, Eunice S., and Baumann, Heinz
- Subjects
CANCER treatment ,ACUTE myeloid leukemia ,ARSENIC trioxide ,EVALUATION of clinical trials ,COMPARATIVE studies - Abstract
BACKGROUND: Constitutive activation of signal transducer and activator of transcription-3 (STAT3) was detected in blasts from approximately 50% of patients with acute myeloid leukemia (AML) and was correlated with an adverse outcome. In vitro treatment of AML blasts with arsenic trioxide (ATO) down-regulated STAT3 activity within 6 hours associated with a reduced viability within 48 hours. METHODS: A phase 1 clinical trial to evaluate the biologically effective dose and/or the maximally tolerated dose (MTD) of ATO in vivo in conjunction with high-dose cytarabine (Hidac) and idarubicin (Ida) in patients with AML aged <60 years was conducted. Data were compared with 117 historic AML patients who had received treatment with Hidac/Ida. RESULTS: In total, 61 patients were enrolled onto 11 different dose levels (from 0.01 to 0.65 mg/kg ideal body weight). The MTD was 0.5 mg/kg. Compared with historic controls, patients who received ATO/Hidac/Ida, although they had similar pretreatment characteristics, had better overall survival ( P = .039). CONCLUSIONS: ATO priming may have improved the outcome of patients aged <60 years with AML who received Hidac/Ida. The current data suggested that ATO may enhance the effect of chemotherapy. The authors concluded that further studies of this novel combination are warranted. Cancer 2011;. © 2011 American Cancer Society. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
32. Genomic, immunophenotypic, and NPM1/FLT3 mutational studies on 17 patients with normal karyotype acute myeloid leukemia (AML) followed by aberrant karyotype AML at relapse
- Author
-
Wang, Eunice S., Sait, Sheila N.J., Gold, David, Mashtare, Terry, Starostik, Petr, Ford, Laurie Ann, Wetzler, Meir, Nowak, Norma J., and Deeb, George
- Subjects
- *
ACUTE myeloid leukemia , *GENOMICS , *IMMUNOPHENOTYPING , *GENETIC mutation , *KARYOTYPES , *CANCER relapse , *HUMAN cytogenetics , *LEUKEMIA etiology - Abstract
Abstract: Normal karyotype (NK) is the most common cytogenetic group in acute myeloid leukemia (AML) diagnosis; however, up to 50% of these patients at relapse will have aberrant karyotype (AK) AML. To determine the etiology of relapsed AK AML cells, we evaluated cytogenetic, immunophenotypic, and molecular results of 17 patients with diagnostic NK AML and relapsed AK AML at our institute. AK AML karyotype was diverse, involving no favorable and largely (8 of 17) complex cytogenetics. Despite clear cytogenetic differences, immunophenotype and NPM1/FLT3 gene mutation status did not change between presentation and relapse in 83% (10 of 12) and 94% (15 of 16) cases, respectively. High-resolution array-based comparative genomic hybridization (aCGH) performed via paired aCGH on NK AML and AK AML samples from the same patient confirmed cytogenetic aberrations only in the relapse sample. Analysis of 16 additional diagnostic NK AML samples revealed no evidence of submicroscopic aberrations undetected by conventional cytogenetics in any case. These results favor evolution of NK AML leukemia cells with acquisition of novel genetic changes as the most common etiology of AK AML relapse as opposed to secondary leukemogenesis. Additional studies are needed to confirm whether AK AML cells represent selection of rare preexisting clones below aCGH detection and to further characterize the molecular lesions found at time of AK AML relapse. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
33. Treating Octogenarian and Nonagenarian Acute Myeloid Leukemia Patients—Predictive Prognostic Models.
- Author
-
Harb, Antoine J., Wei Tan, Wilding, Gregory F., Ford, LaurieAnn, Sait, Sheila N. J., Block, AnneMarie W., Barcos, Maurice, Wallace, Paul K., Wang, Eunice S., and Wetzler, Meir
- Subjects
COMORBIDITY ,MEDICAL research ,RETROSPECTIVE studies ,PROGNOSIS - Abstract
The article presents a study which assessed treatment for octogenarian and nonagenarian patients. It mentions that the retrospective analysis was developed in assisting treatment decision for octogenarian and nonagenarian patients. The study also proposed several models in predicting outcomes which includes the Charlson commorbidity index (CCI) and hematopoietic cell transplant comorbidity index (HCTCI).
- Published
- 2009
- Full Text
- View/download PDF
34. PARP goes the weasel! Emerging role of PARP inhibitors in acute leukemias.
- Author
-
Fritz, Claire, Portwood, Scott M., Przespolewski, Amanda, and Wang, Eunice S.
- Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors, which induce synthetic lethality of BRCA mutant breast and ovarian cancers, are now under active exploration for treatment of acute leukemias, specifically acute myeloid leukemia (AML). Experimental data has revealed that DNA repair deficiencies similar to those found in BRCA mutant solid tumors function in malignant hematopoietic cells to enhance cell survival and promote therapy resistance. Preclinical studies have demonstrated that inhibition of PARP with a variety of agents can dramatically enhance the efficacy of other therapeutic approaches including cytotoxic and epigenetic chemotherapy, small molecule inhibitors (IDH and FLT3 inhibitors) and antibody drug conjugates. This has led to early stage clinical trials of multiple PARP inhibitors (PARPi) for AML patients. Despite small patient numbers, evidence of modest clinical efficacy and tolerability in combinatorial regimens support the further development of PARP inhibition as a novel therapeutic strategy for AML, particularly in select molecular subsets (MLL rearranged, FLT3 and IDH1 mutant disease. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. Beyond midostaurin: Which are the most promising FLT3 inhibitors in AML?
- Author
-
Wang, Eunice S.
- Abstract
Mutations of FLT3 occur in around a third of acute myeloid leukemia (AML) patients and are associated with poor outcomes. Multiple targeted tyrosine kinase inhibitors (TKI) have been developed with different selectivity and potency for FLT3 mutant clones. Indications for which FLT3 inhibitor to use depend on the clinical setting and disease status. Patients with relapsed or refractory AML benefit from a different TKI than those with de novo AML or following stem cell transplant. Moreover, each FLT3 TKI displays a different toxicity and inhibitory profile and may be most useful in patients with varying comorbidities and types of FLT3 mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
36. Pivekimab sunirine (IMGN632), a novel CD123-targeting antibody–drug conjugate, in relapsed or refractory acute myeloid leukaemia: a phase 1/2 study.
- Author
-
Daver, Naval G, Montesinos, Pau, DeAngelo, Daniel J, Wang, Eunice S, Papadantonakis, Nikolaos, Todisco, Elisabetta, Sweet, Kendra L, Pemmaraju, Naveen, Lane, Andrew A, Torres-Miñana, Laura, Thompson, James E, Konopleva, Marina Y, Sloss, Callum M, Watkins, Krystal, Bedse, Gaurav, Du, Yining, Malcolm, Kara E, Zweidler-McKay, Patrick A, and Kantarjian, Hagop M
- Subjects
- *
ACUTE myeloid leukemia , *ANTIBODY-drug conjugates , *HEPATIC veno-occlusive disease , *ADVERSE health care events , *FEBRILE neutropenia - Abstract
Pivekimab sunirine (IMGN632) is a first-in-class antibody–drug conjugate comprising a high-affinity CD123 antibody, cleavable linker, and novel indolinobenzodiazepine pseudodimer payload. CD123 is overexpressed in several haematological malignancies, including acute myeloid leukaemia. We present clinical data on pivekimab sunirine in relapsed or refractory acute myeloid leukaemia. This first-in-human, phase 1/2 dose-escalation and dose-expansion study enrolled participants aged 18 years or older at nine hospitals in France, Italy, Spain, and the USA with CD123+ haematological malignancies (Eastern Cooperative Oncology Group performance status of 0–1); participants reported here were in a cohort of participants with acute myeloid leukaemia who were refractory to or had relapsed on one or more previous treatments for acute myeloid leukaemia. The 3 + 3 dose-escalation phase evaluated two dosing schedules: schedule A (once every 3 weeks, on day 1 of a 3-week cycle) and fractionated schedule B (days 1, 4, and 8 of a 3-week cycle). The dose-expansion phase evaluated two cohorts: one cohort given 0·045 mg/kg of bodyweight (schedule A) and one cohort given 0·090 mg/kg of bodyweight (schedule A). The primary endpoints were the maximum tolerated dose and the recommended phase 2 dose. Antileukaemia activity (overall response and a composite complete remission assessment) was a secondary endpoint. The study is ongoing and registered with ClinicalTrials.gov , NCT03386513. Between Dec 29, 2017, and May 27, 2020, 91 participants were enrolled (schedule A, n=68; schedule B, n=23). 30 (44%) of schedule A participants were female and 38 (56%) were male; 60 (88%) were White, six (9%) were Black or African American, and two (3%) were other races. Pivekimab sunirine at doses of 0·015 mg/kg to 0·450 mg/kg in schedule A was administered in six escalating doses with no maximum tolerated dose defined; three dose-limiting toxicities were observed (reversible veno-occlusive disease; 0·180 mg/kg, n=1 and 0·450 mg/kg, n=1; and neutropenia; 0·300 mg/kg, n=1). Schedule B was not pursued further on the basis of comparative safety and antileukaemia findings with schedule A. The recommended phase 2 dose was selected as 0·045 mg/kg once every 3 weeks. At the recommended phase 2 dose (n=29), the most common grade 3 or worse treatment-related adverse events were febrile neutropenia (three [10%]), infusion-related reactions (two [7%]), and anaemia (two [7%]). Treatment-related serious adverse events occurring in 5% or more of participants treated at the recommended phase 2 dose were febrile neutropenia (two [7%]) and infusion-related reactions (two [7%]). Among 68 participants who received schedule A, one death (1%) was considered to be treatment-related (cause unknown; 0·300 mg/kg cohort). At the recommended phase 2 dose, the overall response rate was 21% (95% CI 8–40; six of 29) and the composite complete remission rate was 17% (95% CI 6–36; five of 29). Pivekimab sunirine showed single-agent activity across multiple doses, with a recommended phase 2 dose of 0·045 mg/kg once every 3 weeks. These findings led to a phase 1b/2 study of pivekimab sunirine plus azacitidine and venetoclax in patients with CD123-positive acute myeloid leukaemia. ImmunoGen. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Intensive chemotherapy vs. hypomethylating agents in older adults with newly diagnosed high-risk acute myeloid leukemia: A single center experience.
- Author
-
Vachhani, Pankit, Al Yacoub, Raed, Miller, Austin, Zhang, Fan, Cronin, Tara L., Ontiveros, Evelena P., Thompson, James E., Griffiths, Elizabeth A., and Wang, Eunice S.
- Subjects
- *
CANCER chemotherapy , *OLDER patients , *ACUTE myeloid leukemia , *CYTARABINE , *ANTHRACYCLINES - Abstract
Highlights Older patients with secondary AML or AML-MRC have poor prognosis. Hypomethylating agents offer survival outcomes comparable to intensive chemotherapy. Transplant eligibility and potential effects on QoL should drive therapy selection. Abstract Acute myeloid leukemia (AML) in older patients is often associated with biologic and clinical characteristics that predict poor outcomes to cytarabine and anthracycline based induction chemotherapy (IC). The impact of hypomethylating agents (HMA) in the treatment of these high-risk patients is unknown. Here we retrospectively examined the remission rates and survival outcomes of 201 newly diagnosed patients ≥60 years old with therapy-related (t-AML), secondary (s-AML), or AML with myelodysplasia-related changes (AML-MRC). Ninety-eight patients received IC, and 103 received HMA. Patients in the IC cohort were younger than those who received HMA (68 vs. 74 years; p < 0.01) with lower comorbidity burden. Composite complete remission rates (CR) were 39% in IC and 27% in the HMA cohorts (p = 0.10). Overall survival (OS) was not significantly different between the two cohorts (7.59 mos vs. 5.49 mos; HR 0.75 95% CI 0.55–1.02) despite the fact that more patients in the IC cohort (33% versus 5%, p < 0.01) underwent allogeneic stem cell transplant. Patients with t-AML (HR 0.56; 95% CI 0.33–0.97) and complex karyotype without monosomal karyotype (CK + MK-; HR 0.37; 95% CI 0.19–0.75) had better OS following IC. Patients with CK + MK+ (HR 2.00; 95% CI 1.08–3.70) had improved OS following HMA. Our results support the use of HMA as an alternative upfront regimen in older individuals with newly diagnosed high-risk AML based on similar clinical outcomes to IC. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
38. Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial.
- Author
-
Cortes, Jorge, Perl, Alexander E, Döhner, Hartmut, Kantarjian, Hagop, Martinelli, Giovanni, Kovacsovics, Tibor, Rousselot, Philippe, Steffen, Björn, Dombret, Hervé, Estey, Elihu, Strickland, Stephen, Altman, Jessica K, Baldus, Claudia D, Burnett, Alan, Krämer, Alwin, Russell, Nigel, Shah, Neil P, Smith, Catherine C, Wang, Eunice S, and Ifrah, Norbert
- Subjects
- *
PROTEIN-tyrosine kinases , *ACUTE myeloid leukemia , *GENETIC mutation , *PLATELET count , *PNEUMONIA , *ACUTE myeloid leukemia diagnosis , *THIAZOLES , *UREA , *CANCER relapse , *CLINICAL trials , *COMPARATIVE studies , *DRUG administration , *DRUG dosage , *DOSE-effect relationship in pharmacology , *DRUG toxicity , *INTERNATIONAL relations , *RESEARCH methodology , *MEDICAL cooperation , *ORAL drug administration , *PROGNOSIS , *RESEARCH , *RESEARCH funding , *SURVIVAL , *TRANSFERASES , *EVALUATION research , *TREATMENT effectiveness , *THERAPEUTICS - Abstract
Background: Old age and FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutations in patients with acute myeloid leukaemia are associated with early relapse and poor survival. Quizartinib is an oral, highly potent, and selective next-generation FLT3 inhibitor with clinical antileukaemic activity in relapsed or refractory acute myeloid leukaemia. We aimed to assess the efficacy and safety of single-agent quizartinib in patients with relapsed or refractory acute myeloid leukaemia.Methods: We did an open-label, multicentre, single-arm, phase 2 trial at 76 hospitals and cancer centres in the USA, Europe, and Canada. We enrolled patients with morphologically documented primary acute myeloid leukaemia or acute myeloid leukaemia secondary to myelodysplastic syndromes and an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 into two predefined, independent cohorts: patients who were aged 60 years or older with relapsed or refractory acute myeloid leukaemia within 1 year after first-line therapy (cohort 1), and those who were 18 years or older with relapsed or refractory disease following salvage chemotherapy or haemopoietic stem cell transplantation (cohort 2). Patients with an FLT3-ITD allelic frequency of more than 10% were considered as FLT3-ITD positive, whereas all other patients were considered as FLT3-ITD negative. Patients received quizartinib once daily as an oral solution; the initial 17 patients received 200 mg per day but the QTcF interval was prolonged for more than 60 ms above baseline in some of these patients. Subsequently, doses were amended for all patients to 135 mg per day for men and 90 mg per day for women. The co-primary endpoints were the proportion of patients who achieved a composite complete remission (defined as complete remission + complete remission with incomplete platelet recovery + complete remission with incomplete haematological recovery) and the proportion of patients who achieved a complete remission. Efficacy and safety analyses included all patients who received at least one dose of quizartinib (ie, the intention-to-treat population). Patients with a locally assessed post-treatment bone marrow aspirate or biopsy were included in efficacy analyses by response; all other patients were considered to have an unknown response. This study is registered with ClinicalTrials.gov, number NCT00989261, and with the European Clinical Trials Database, EudraCT 2009-013093-41, and is completed.Findings: Between Nov 19, 2009, and Oct 31, 2011, a total of 333 patients were enrolled (157 in cohort 1 and 176 in cohort 2). In cohort 1, 63 (56%) of 112 FLT3-ITD-positive patients and 16 (36%) of 44 FLT3-ITD-negative patients achieved composite complete remission, with three (3%) FLT3-ITD-positive patients and two (5%) FLT3-ITD-negative patients achieving complete remission. In cohort 2, 62 (46%) of 136 FLT3-ITD-positive patients achieved composite complete remission with five (4%) achieving complete remission, whereas 12 (30%) of 40 FLT3-ITD-negative patients achieved composite complete remission with one (3%) achieving complete remission. Across both cohorts (ie, the intention-to-treat population of 333 patients), grade 3 or worse treatment-related treatment-emergent adverse events in 5% or more of patients were febrile neutropenia (76 [23%] of 333), anaemia (75 [23%]), thrombocytopenia (39 [12%]), QT interval corrected using Fridericia's formula (QTcF) prolongation (33 [10%]), neutropenia (31 [9%]), leucopenia (22 [7%]), decreased platelet count (20 [6%]), and pneumonia (17 [5%]). Serious adverse events occurring in 5% or more of patients were febrile neutropenia (126 [38%] of 333; 76 treatment related), acute myeloid leukaemia progression (73 [22%]), pneumonia (40 [12%]; 14 treatment related), QTcF prolongation (33 [10%]; 32 treatment related), sepsis (25 [8%]; eight treatment related), and pyrexia (18 [5%]; nine treatment related). Notable serious adverse events occurring in less than 5% of patients were torsades de pointes (one [<1%]) and hepatic failure (two [1%]). In total, 125 (38%) of 333 patients died within the study treatment period, including the 30-day follow-up. 18 (5%) patients died because of an adverse event considered by the investigator to be treatment related (ten [6%] of 157 patients in cohort 1 and eight [5%] of 176 in cohort 2.Interpretation: Single-agent quizartinib was shown to be highly active and generally well tolerated in patients with relapsed or refractory acute myeloid leukaemia, particularly those with FLT3-ITD mutations. These findings confirm that targeting the FLT3-ITD driver mutation with a highly potent and selective FLT3 inhibitor is a promising clinical strategy to help improve clinical outcomes in patients with very few options. Phase 3 studies (NCT02039726; NCT02668653) will examine quizartinib at lower starting doses.Funding: Ambit Biosciences/Daiichi Sankyo. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
39. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study.
- Author
-
Perl, Alexander E, Altman, Jessica K, Cortes, Jorge, Smith, Catherine, Litzow, Mark, Baer, Maria R, Claxton, David, Erba, Harry P, Gill, Stan, Goldberg, Stuart, Jurcic, Joseph G, Larson, Richard A, Liu, Chaofeng, Ritchie, Ellen, Schiller, Gary, Spira, Alexander I, Strickland, Stephen A, Tibes, Raoul, Ustun, Celalettin, and Wang, Eunice S
- Subjects
- *
CARBOXAMIDES , *ACUTE myeloid leukemia , *ACUTE myeloid leukemia treatment , *DRUG dosage , *MEDICATION safety , *GENETICS , *AMINES , *ANTINEOPLASTIC agents , *BLOOD platelets , *CLINICAL trials , *COMPARATIVE studies , *DRUG toxicity , *RESEARCH methodology , *HETEROCYCLIC compounds , *MEDICAL cooperation , *PHOSPHORYLATION , *REOPERATION , *RESEARCH , *RESEARCH funding , *TRANSFERASES , *DISEASE relapse , *EVALUATION research , *PHARMACODYNAMICS , *THERAPEUTICS - Abstract
Background: Internal tandem duplication mutations in FLT3 are common in acute myeloid leukaemia and are associated with rapid relapse and short overall survival. The clinical benefit of FLT3 inhibitors in patients with acute myeloid leukaemia has been limited by rapid generation of resistance mutations, particularly in codon Asp835 (D835). We aimed to assess the highly selective oral FLT3 inhibitor gilteritinib in patients with relapsed or refractory acute myeloid leukaemia.Methods: In this phase 1-2 trial, we enrolled patients aged 18 years or older with acute myeloid leukaemia who either were refractory to induction therapy or had relapsed after achieving remission with previous treatment. Patients were enrolled into one of seven dose-escalation or dose-expansion cohorts assigned to receive once-daily doses of oral gilteritinib (20 mg, 40 mg, 80 mg, 120 mg, 200 mg, 300 mg, or 450 mg). Cohort expansion was based on safety and tolerability, FLT3 inhibition in correlative assays, and antileukaemic activity. Although the presence of an FLT3 mutation was not an inclusion criterion, we required ten or more patients with locally confirmed FLT3 mutations (FLT3mut+) to be enrolled in expansion cohorts at each dose level. On the basis of emerging findings, we further expanded the 120 mg and 200 mg dose cohorts to include FLT3mut+ patients only. The primary endpoints were the safety, tolerability, and pharmacokinetics of gilteritinib. Safety and tolerability were assessed in the safety analysis set (all patients who received at least one dose of gilteritinib). Responses were assessed in the full analysis set (all patients who received at least one dose of study drug and who had at least one datapoint post-treatment). Pharmacokinetics were assessed in a subset of the safety analysis set for which sufficient data for concentrations of gilteritinib in plasma were available to enable derivation of one or more pharmacokinetic variables. This study is registered with ClinicalTrials.gov, number NCT02014558, and is ongoing.Findings: Between Oct 15, 2013, and Aug 27, 2015, 252 adults with relapsed or refractory acute myeloid leukaemia received oral gilteritinib once daily in one of seven dose-escalation (n=23) or dose-expansion (n=229) cohorts. Gilteritinib was well tolerated; the maximum tolerated dose was established as 300 mg/day when two of three patients enrolled in the 450 mg dose-escalation cohort had two dose-limiting toxicities (grade 3 diarrhoea and grade 3 elevated aspartate aminotransferase). The most common grade 3-4 adverse events irrespective of relation to treatment were febrile neutropenia (97 [39%] of 252), anaemia (61 [24%]), thrombocytopenia (33 [13%]), sepsis (28 [11%]), and pneumonia (27 [11%]). Commonly reported treatment-related adverse events were diarrhoea (92 [37%] of 252]), anaemia (86 [34%]), fatigue (83 [33%]), elevated aspartate aminotransferase (65 [26%]), and increased alanine aminotransferase (47 [19%]). Serious adverse events occurring in 5% or more of patients were febrile neutropenia (98 [39%] of 252; five related to treatment), progressive disease (43 [17%]), sepsis (36 [14%]; two related to treatment), pneumonia (27 [11%]), acute renal failure (25 [10%]; five related to treatment), pyrexia (21 [8%]; three related to treatment), bacteraemia (14 [6%]; one related to treatment), and respiratory failure (14 [6%]). 95 people died in the safety analysis set, of which seven deaths were judged possibly or probably related to treatment (pulmonary embolism [200 mg/day], respiratory failure [120 mg/day], haemoptysis [80 mg/day], intracranial haemorrhage [20 mg/day], ventricular fibrillation [120 mg/day], septic shock [80 mg/day], and neutropenia [120 mg/day]). An exposure-related increase in inhibition of FLT3 phosphorylation was noted with increasing concentrations in plasma of gilteritinib. In-vivo inhibition of FLT3 phosphorylation occurred at all dose levels. At least 90% of FLT3 phosphorylation inhibition was seen by day 8 in most patients receiving a daily dose of 80 mg or higher. 100 (40%) of 249 patients in the full analysis set achieved a response, with 19 (8%) achieving complete remission, ten (4%) complete remission with incomplete platelet recovery, 46 (18%) complete remission with incomplete haematological recovery, and 25 (10%) partial remission INTERPRETATION: Gilteritinib had a favourable safety profile and showed consistent FLT3 inhibition in patients with relapsed or refractory acute myeloid leukaemia. These findings confirm that FLT3 is a high-value target for treatment of relapsed or refractory acute myeloid leukaemia; based on activity data, gilteritinib at 120 mg/day is being tested in phase 3 trials.Funding: Astellas Pharma, National Cancer Institute (Leukemia Specialized Program of Research Excellence grant), Associazione Italiana Ricerca sul Cancro. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
40. A phase I study of intermediate dose cytarabine in combination with lenalidomide in relapsed/refractory acute myeloid leukemia.
- Author
-
Griffiths, Elizabeth A., Brady, William E., Tan, Wei, Vigil, Carlos E., Thompson, James E., Ford, Laurie A., Dickey, Noelle M., L. Bashaw, Heather, Sperrazza, Jill, Wetzler, Meir, and Wang, Eunice S.
- Subjects
- *
CYTARABINE , *ACUTE myeloid leukemia treatment , *ACUTE leukemia , *DISEASE relapse , *HYPOKALEMIA - Abstract
Relapsed/refractory (r/r) Acute Myeloid Leukemia (AML) remains a therapeutic challenge. Cytarabine arabinoside (AraC) forms the backbone of most regimens, with complete responses (CR) ranging from 17 to 20%. Lenalidomide (Len) is approved by the FDA for multiple myeloma and myelodysplasia and has demonstrated activity in AML. We developed a phase I study to evaluate the safety and tolerability of Len in combination with intermediate dose AraC (1.5 g/m 2 /day given on days 1–5) in adults with r/r AML. The maximally tolerated dose for this combination was 10 mg daily on days 6–26 of a 28 day cycle. Dose de-escalation from 25 mg was required due to rash, liver function abnormalities, and hypokalemia. Of 32 evaluable patients, five achieved CR (16%), 5CRi (16%) and 3 had hematological improvements for an overall response rate of 41% (13/32). Median overall survival (95% confidence interval) for patients treated on study was 5.8 (2.5–10.6) months and disease free survival was 3.4 (2.3–6.2) months. This single institute phase I trial of Len and intermediate dose AraC was associated with marked skin and other toxicities. At the dose and schedule tested, this combination did not appear to result in improved CR over single agent AraC for r/r AML. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
41. Down-regulation of signal transducer and activator of transcription 3 improves human acute myeloid leukemia-derived dendritic cell function.
- Author
-
Brady, Michael T., Miller, Austin, Sait, Sheila N., Ford, Laurie A., Minderman, Hans, Wang, Eunice S., Lee, Kelvin P., Baumann, Heinz, and Wetzler, Meir
- Subjects
- *
CELLULAR signal transduction , *GENETIC transcription , *ACUTE myeloid leukemia , *DENDRITIC cells , *CELL differentiation , *T cells - Abstract
Abstract: Signal transducer and activator of transcription (STAT) 3 inhibits dendritic cell (DC) differentiation and is constitutively activated in blasts of approximately half of AML patients. We investigated the correlation between STAT3 activity, DC maturation and the ability to stimulate T-cells in primary acute myeloid leukemia (AML)-derived DCs. STAT3 knock-down by shRNAmir increased the ability of AML-DCs to stimulate T-cells. Treatment of AML-DC with arsenic trioxide, but not AG490, JSI-124 or NSC-74859, led to a more mature phenotype and enhanced T-cell stimulation, while having minimal effect on normal DC. We conclude that AML-DCs have improved immunogenicity after reducing STAT3. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
42. Myeloid blastic transformation of myeloproliferative neoplasms—A review of 112 cases
- Author
-
Noor, Syed J., Tan, Wei, Wilding, Gregory E., Ford, Laurie A., Barcos, Maurice, Sait, Sheila N.J., Block, AnneMarie W., Thompson, James E., Wang, Eunice S., and Wetzler, Meir
- Subjects
- *
MYELOPROLIFERATIVE neoplasms , *CELL transformation , *ERYTHROPOIETIN , *THROMBOCYTOSIS , *ANTINEOPLASTIC agents , *ALKYLATING agents , *BONE marrow transplantation , *MEDICAL records , *ACUTE myeloid leukemia - Abstract
Abstract: Blastic transformation of myeloproliferative neoplasms (MPN) is still poorly understood. We describe a cohort of 23 Roswell Park Cancer Institute (RPCI) patients and 89 additional cases from the English literature for whom biologic features were described. We initially compared our 23 patients to the 89 cases from the literature. Our population had significantly less patients with prior history of polycythemia vera (PV), shorter time from MPN diagnosis to blastic transformation, <3 prior therapies, more frequent use of hydroxyurea and erythropoietin and less frequent use of alkylating agents. Interestingly, the overall survival of the two cohorts from the time of blastic transformation was similar. We therefore looked at the outcome of the entire cohort (n =112). Patients with prior history of essential thrombocythemia survived longer than patients with prior history of myelofibrosis or PV. Further, patients with <3 prior therapies, those who lacked complex karyotype and those <60 year old at MPN diagnosis had significantly longer survival. Among the PRCI population, 20/23 patients underwent induction treatment with cytarabine and an anthracycline containing regimens; 12 achieved remission and their overall survival was significantly longer than those who did not. Three patients underwent an allogeneic transplantation and their survival was significantly longer than those who did not. Patients with <3 prior therapies, those who lack complex karyotype and those <60 at MPN diagnosis have longer survival following blastic transformation. Finally, allogeneic transplantation represents the only chance for long-term survival in these patients. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
43. Hypoxia-inducible factor-1α protein expression is associated with poor survival in normal karyotype adult acute myeloid leukemia
- Author
-
Deeb, George, Vaughan, Mary M., McInnis, Ian, Ford, Laurie Ann, Sait, Sheila N.J., Starostik, Petr, Wetzler, Meir, Mashtare, Terry, and Wang, Eunice S.
- Subjects
- *
TRANSCRIPTION factors , *GENE expression , *ACUTE myeloid leukemia , *VASCULAR endothelial growth factors , *CELL membranes , *HYPOXEMIA , *HEALTH outcome assessment , *IMMUNOHISTOCHEMISTRY - Abstract
Abstract: We examined the predictive impact of HIF-1α protein expression on clinical outcome of 84 normal karyotype acute myeloid leukemia (NK-AML) patients (median age 66.5 years) at our institute. Thirty percent of NK-AML cells expressed cytoplasmic HIF-1α. In univariate analysis, low HIF-1α (≤5%, n =66) was associated with improved event-free survival (p =0.0453, HR=0.22). Multivariate analysis incorporating age, complete remission, FLT3-ITD mutation, and marrow blast percentage demonstrated that HIF-1α was independently associated with poorer overall and event-free survival. HIF-1α expression correlated with VEGF-C but not VEGF-A, marrow angiogenesis, FLT3 ITD or NPM1 mutations. These results support HIF-1α as an outcome marker for NK-AML. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
44. Metachronous and synchronous presentation of acute myeloid leukemia and lung cancer
- Author
-
Varadarajan, Ramya, Ford, LaurieAnn, Sait, Sheila N.J., Block, AnneMarie W., Barcos, Maurice, Wallace, Paul K., Ramnath, Nithya, Wang, Eunice S., and Wetzler, Meir
- Subjects
- *
ACUTE myeloid leukemia treatment , *LUNG cancer , *PHYSIOLOGICAL effects of tobacco , *CANCER patients , *SMOKING cessation - Abstract
Abstract: Smoking is associated with both acute myeloid leukemia (AML) and lung cancer. We therefore searched our database for concomitant presentation of AML and lung cancer. Among 775 AML cases and 5225 lung cancer cases presenting to Roswell Park Cancer Institute between the years January 1992 and May 2008 we found 12 (1.5% of AML cases; 0.23% of lung cancer cases) cases (seven metachronous and five synchronous) with AML and lung cancer. All but one patient were smokers. There were no unique characteristic of either AML or lung cancer in these patients. Nine patients succumbed to AML, one died from an unrelated cause while undergoing treatment for AML, one died of lung cancer and one patient is alive after allogeneic transplantation for AML. In summary, this study supports the need for effective smoking cessation programs. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
45. Acute myeloid leukemia and diabetes insipidus with monosomy 7
- Author
-
Harb, Antoine, Tan, Wei, Wilding, Gregory E., Battiwalla, Minoo, Sait, Sheila N.J., Wang, Eunice S., and Wetzler, Meir
- Subjects
- *
ACUTE myeloid leukemia , *DIABETES , *ANEUPLOIDY , *KARYOTYPES , *MEDICAL literature , *HEALTH outcome assessment , *PATIENTS - Abstract
Abstract: The predisposition of monosomy 7 to diabetes insipidus (DI) in acute myeloid leukemia (AML) led us to ask whether AML associated with monosomy 7 and DI will differ from AML associated with other karyotype aberrations and DI and whether the outcome of patients with AML and DI will differ from those without DI. We describe 2 patients from Roswell Park Cancer Institute and discuss 29 additional cases from the literature. AML with monosomy 7 and DI (n = 25) had a trend towards a lower complete remission (p = 0.0936) and worse survival (p = 0.0480) than AML with other karyotype changes and DI (n = 6). Further, AML with monosomy 7 and DI had worse complete remission rate and overall survival than AML with monosomy 7 but without DI. In conclusion, it appears that AML with monosomy 7 and DI is a disease entity with specifically poor outcome. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
46. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1–2 study
- Author
-
Raoul Tibes, Stan Gill, Chaofeng Liu, Christoph Röllig, Stephen A. Strickland, Joseph G. Jurcic, Stuart L. Goldberg, Maria R. Baer, Alexander E. Perl, Alexander I. Spira, Andreas Neubauer, Gary J. Schiller, Mark R. Litzow, Richard A. Larson, Catherine C. Smith, Jessica K. Altman, Harry P. Erba, Mark J. Levis, Erkut Bahceci, Giovanni Martinelli, Robert K. Stuart, Eunice S. Wang, Jorge E. Cortes, David F. Claxton, Celalettin Ustun, Ellen K. Ritchie, Perl, Alexander E, Altman, Jessica K, Cortes, Jorge, Smith, Catherine, Litzow, Mark, Baer, Maria R, Claxton, David, Erba, Harry P, Gill, Stan, Goldberg, Stuart, Jurcic, Joseph G, Larson, Richard A, Liu, Chaofeng, Ritchie, Ellen, Schiller, Gary, Spira, Alexander I, Strickland, Stephen A, Tibes, Raoul, Ustun, Celalettin, Wang, Eunice S, Stuart, Robert, Röllig, Christoph, Neubauer, Andrea, Martinelli, Giovanni, Bahceci, Erkut, and Levis, Mark
- Subjects
Male ,Myeloid ,0301 basic medicine ,Gastroenterology ,chemistry.chemical_compound ,tyrosine kinase inhibitor ,0302 clinical medicine ,Recurrence ,hemic and lymphatic diseases ,Midostaurin ,Phosphorylation ,Lung ,Cancer ,Aniline Compounds ,Leukemia ,Hematology ,Middle Aged ,3. Good health ,Leukemia, Myeloid, Acute ,Infectious Diseases ,medicine.anatomical_structure ,Oncology ,Tolerability ,Pyrazines ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,embryonic structures ,Retreatment ,Female ,Patient Safety ,Blood Platelets ,medicine.medical_specialty ,Maximum Tolerated Dose ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antineoplastic Agents ,acute myeloid leukemia ,Acute ,Neutropenia ,Article ,relapsed/refractory ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Genetics ,medicine ,Humans ,Oncology & Carcinogenesis ,FLT3 inhibition ,Aged ,Quizartinib ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Surgery ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,chemistry ,Fms-Like Tyrosine Kinase 3 ,business ,Progressive disease ,Febrile neutropenia - Abstract
Summary Background Internal tandem duplication mutations in FLT3 are common in acute myeloid leukaemia and are associated with rapid relapse and short overall survival. The clinical benefit of FLT3 inhibitors in patients with acute myeloid leukaemia has been limited by rapid generation of resistance mutations, particularly in codon Asp835 (D835). We aimed to assess the highly selective oral FLT3 inhibitor gilteritinib in patients with relapsed or refractory acute myeloid leukaemia. Methods In this phase 1–2 trial, we enrolled patients aged 18 years or older with acute myeloid leukaemia who either were refractory to induction therapy or had relapsed after achieving remission with previous treatment. Patients were enrolled into one of seven dose-escalation or dose-expansion cohorts assigned to receive once-daily doses of oral gilteritinib (20 mg, 40 mg, 80 mg, 120 mg, 200 mg, 300 mg, or 450 mg). Cohort expansion was based on safety and tolerability, FLT3 inhibition in correlative assays, and antileukaemic activity. Although the presence of an FLT3 mutation was not an inclusion criterion, we required ten or more patients with locally confirmed FLT3 mutations ( FLT3 mut+ ) to be enrolled in expansion cohorts at each dose level. On the basis of emerging findings, we further expanded the 120 mg and 200 mg dose cohorts to include FLT3 mut+ patients only. The primary endpoints were the safety, tolerability, and pharmacokinetics of gilteritinib. Safety and tolerability were assessed in the safety analysis set (all patients who received at least one dose of gilteritinib). Responses were assessed in the full analysis set (all patients who received at least one dose of study drug and who had at least one datapoint post-treatment). Pharmacokinetics were assessed in a subset of the safety analysis set for which sufficient data for concentrations of gilteritinib in plasma were available to enable derivation of one or more pharmacokinetic variables. This study is registered with ClinicalTrials.gov, number NCT02014558, and is ongoing. Findings Between Oct 15, 2013, and Aug 27, 2015, 252 adults with relapsed or refractory acute myeloid leukaemia received oral gilteritinib once daily in one of seven dose-escalation (n=23) or dose-expansion (n=229) cohorts. Gilteritinib was well tolerated; the maximum tolerated dose was established as 300 mg/day when two of three patients enrolled in the 450 mg dose-escalation cohort had two dose-limiting toxicities (grade 3 diarrhoea and grade 3 elevated aspartate aminotransferase). The most common grade 3–4 adverse events irrespective of relation to treatment were febrile neutropenia (97 [39%] of 252), anaemia (61 [24%]), thrombocytopenia (33 [13%]), sepsis (28 [11%]), and pneumonia (27 [11%]). Commonly reported treatment-related adverse events were diarrhoea (41 [16%] of 252]), fatigue (37 [15%]), elevated aspartate aminotransferase (33 [13%]), and elevated alanine aminotransferase (24 [10%]). Serious adverse events occurring in 5% or more of patients were febrile neutropenia (78 [31%] of 252; five related to treatment), progressive disease (43 [17%]), sepsis (36 [14%]; two related to treatment), pneumonia (27 [11%]), acute renal failure (25 [10%]; five related to treatment), pyrexia (21 [8%]; three related to treatment), bacteraemia (14 [6%]; one related to treatment), and respiratory failure (14 [6%]). 95 people died in the safety analysis set, of which seven deaths were judged possibly or probably related to treatment (pulmonary embolism [200 mg/day], respiratory failure [120 mg/day], haemoptysis [80 mg/day], intracranial haemorrhage [20 mg/day], ventricular fibrillation [120 mg/day], septic shock [80 mg/day], and neutropenia [120 mg/day]). An exposure-related increase in inhibition of FLT3 phosphorylation was noted with increasing concentrations in plasma of gilteritinib. In-vivo inhibition of FLT3 phosphorylation occurred at all dose levels. At least 90% of FLT3 phosphorylation inhibition was seen by day 8 in most patients receiving a daily dose of 80 mg or higher. 100 (40%) of 249 patients in the full analysis set achieved a response, with 19 (8%) achieving complete remission, ten (4%) complete remission with incomplete platelet recovery, 46 (18%) complete remission with incomplete haematological recovery, and 25 (10%) partial remission. Interpretation Gilteritinib had a favourable safety profile and showed consistent FLT3 inhibition in patients with relapsed or refractory acute myeloid leukaemia. These findings confirm that FLT3 is a high-value target for treatment of relapsed or refractory acute myeloid leukaemia; based on activity data, gilteritinib at 120 mg/day is being tested in phase 3 trials. Funding Astellas Pharma, National Cancer Institute (Leukemia Specialized Program of Research Excellence grant), Associazione Italiana Ricerca sul Cancro.
- Published
- 2017
- Full Text
- View/download PDF
47. A phase 2 trial of single low doses of rasburicase for treatment of hyperuricemia in adult patients with acute leukemia.
- Author
-
Vachhani, Pankit, Baron, Jeffrey, Freyer, Craig W., Miller, Austin, Wetzler, Meir, Thompson, James E., Griffiths, Elizabeth A., and Wang, Eunice S.
- Subjects
- *
ACUTE leukemia , *ADULTS , *ACUTE promyelocytic leukemia , *TUMOR lysis syndrome , *ACUTE myeloid leukemia - Abstract
• Rasburicase is a synthetic urate oxidase that oxidates uric acid to allantoin which is more soluble in urine than uric acid. • Rasburicase can be expensive especially when administered as per the recommended dosage of 0.2 mg/kg/day for up to 5 days. • Single doses of rasburicase (1.5−3 mg) in addition to adjunct measures are safe and efficacious for prevention and treatment of TLS. Rasburicase can markedly and rapidly decrease uric acid (UA) levels, thereby preventing and treating tumor lysis syndrome. However, rasburicase is expensive, especially when used as per the manufacturer's recommended dosage of 0.2 mg/kg/day for up to 5 days. Numerous reports have shown that lower, and even single doses are effective in lowering UA levels but prospective randomized studies comparing low doses have not been performed. To prospectively determine the efficacy and safety of two single low doses of rasburicase in adult patients (pts) with acute leukemia and elevated plasma UA. Eligible pts aged ≥ 18 years old with acute leukemia and UA ≥ 7.5 mg/dL were randomized to receive an initial single dose of rasburicase 1.5 mg (Arm A) or 3 mg (Arm B) on day 1 in an unblinded fashion. All pts received allopurinol 300 mg daily on days 1–6. Twenty-four pts (median age 69 years; 14 males and 10 females) were enrolled in this phase 2 study (12 on each arm). Twenty pts had acute myeloid leukemia while 3 had acute lymphoblastic leukemia, and 1 had acute promyelocytic leukemia. Median initial UA level was 9.8 mg/dL. Eighty-three percent of pts in both arms achieved UA < 7.5 mg/dL by 24 h after therapy. Five pts (21 %; 2 from Arm A and 3 from Arm B) required additional doses of rasburicase. The majority (23/24) of pts achieved UA goals after 1–2 doses of rasburicase. None had worsening renal function. Both doses were well tolerated, and no treatment related adverse events were reported. Single doses of rasburicase (as low as 1.5−3 mg) used in addition to allopurinol were well tolerated and highly efficacious (83 % response rate) in decreasing UA levels within 24 h of administration in adult acute leukemia pts with hyperuricemia. [ABSTRACT FROM AUTHOR]
- 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.