scholarly journals Gilteritinib induces differentiation in relapsed and refractory FLT3-mutated acute myeloid leukemia

2019 ◽  
Vol 3 (10) ◽  
pp. 1581-1585 ◽  
Author(s):  
Christine M. McMahon ◽  
Jonathan Canaani ◽  
Bryan Rea ◽  
Rachel L. Sargent ◽  
Julianne N. Qualtieri ◽  
...  

Key Points Gilteritinib induces 2 distinct marrow responses in FLT3-mutated AML: responses with and without differentiation. Ongoing clonal hematopoiesis is ubiquitous during gilteritinib therapy and may promote genetic evolution and drug resistance.

Blood ◽  
2015 ◽  
Vol 126 (3) ◽  
pp. 363-372 ◽  
Author(s):  
Rongqing Pan ◽  
Vivian R. Ruvolo ◽  
Jun Wei ◽  
Marina Konopleva ◽  
John C. Reed ◽  
...  

Key Points Mcl-1 inhibition by pan-active Bcl-2 inhibitor (–)BI97D6 kills AML cells via induction of mitochondrial apoptosis. Mcl-1 inhibition by (–)BI97D6 overcomes intrinsic and extrinsic drug resistance to ABT-737.


Blood ◽  
2015 ◽  
Vol 126 (22) ◽  
pp. 2484-2490 ◽  
Author(s):  
Jane E. Churpek ◽  
Khateriaa Pyrtel ◽  
Krishna-Latha Kanchi ◽  
Jin Shao ◽  
Daniel Koboldt ◽  
...  

Key Points Known pathogenic germ line variants in 12 genes can explain nearly 30% of families with inherited predisposition to MDS/AML. Asymptomatic carriers of germ line RUNX1 mutations develop detectable clonal hematopoiesis with a cumulative risk of >80% by age 50 years.


Blood ◽  
2014 ◽  
Vol 123 (14) ◽  
pp. 2209-2219 ◽  
Author(s):  
Hee Kyu Lee ◽  
Hong Woo Kim ◽  
In Yong Lee ◽  
Jungmi Lee ◽  
Jaekyoo Lee ◽  
...  

Key Points A novel inhibitor G-749 is very potent against FLT3 kinase mutants including D835Y and ITD/F691L that confer resistance to PKC412 and AC220. G-749 shows several desirable characteristics to overcome other drug resistances conferred by patient plasma, FLT3 ligand, and stromal cells.


Nature Cancer ◽  
2021 ◽  
Vol 2 (5) ◽  
pp. 527-544 ◽  
Author(s):  
Marina Scheller ◽  
Anne Kathrin Ludwig ◽  
Stefanie Göllner ◽  
Christian Rohde ◽  
Stephen Krämer ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (7) ◽  
pp. 1316-1324 ◽  
Author(s):  
Margaret L. Green ◽  
Wendy M. Leisenring ◽  
Hu Xie ◽  
Roland B. Walter ◽  
Marco Mielcarek ◽  
...  

Key Points CMV reactivation after HCT is associated with a reduced risk of early relapse in patients with AML but not other disease groups. The benefit, however, is offset by an increased risk of nonrelapse mortality.


Blood ◽  
2015 ◽  
Vol 125 (15) ◽  
pp. 2386-2396 ◽  
Author(s):  
Francis Mussai ◽  
Sharon Egan ◽  
Joseph Higginbotham-Jones ◽  
Tracey Perry ◽  
Andrew Beggs ◽  
...  

Key Points Arginase depletion with BCT-100 pegylated recombinant human arginase is cytotoxic to AML blasts.


2015 ◽  
Vol 39 (1) ◽  
pp. 92-99 ◽  
Author(s):  
Bing Xia ◽  
Chen Tian ◽  
Shanqi Guo ◽  
Le Zhang ◽  
Dandan Zhao ◽  
...  

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yongliang Liu ◽  
Guiqin Wang ◽  
Jiasi Zhang ◽  
Xue Chen ◽  
Huailong Xu ◽  
...  

Abstract Background Leukemia stem cells (LSCs) are responsible for the initiation, progression, and relapse of acute myeloid leukemia (AML). Therefore, a therapeutic strategy targeting LSCs is a potential approach to eradicate AML. In this study, we aimed to identify LSC-specific surface markers and uncover the underlying mechanism of AML LSCs. Methods Microarray gene expression data were used to investigate candidate AML-LSC-specific markers. CD9 expression in AML cell lines, patients with AML, and normal donors was evaluated by flow cytometry (FC). The biological characteristics of CD9-positive (CD9+) cells were analyzed by in vitro proliferation, chemotherapeutic drug resistance, migration, and in vivo xenotransplantation assays. The molecular mechanism involved in CD9+ cell function was investigated by gene expression profiling. The effects of alpha-2-macroglobulin (A2M) on CD9+ cells were analyzed with regard to proliferation, drug resistance, and migration. Results CD9, a cell surface protein, was specifically expressed on AML LSCs but barely detected on normal hematopoietic stem cells (HSCs). CD9+ cells exhibit more resistance to chemotherapy drugs and higher migration potential than do CD9-negative (CD9−) cells. More importantly, CD9+ cells possess the ability to reconstitute human AML in immunocompromised mice and promote leukemia growth, suggesting that CD9+ cells define the LSC population. Furthermore, we identified that A2M plays a crucial role in maintaining CD9+ LSC stemness. Knockdown of A2M impairs drug resistance and migration of CD9+ cells. Conclusion Our findings suggest that CD9 is a new biomarker of AML LSCs and is a promising therapeutic target.


Sign in / Sign up

Export Citation Format

Share Document