scholarly journals Targeted Therapies and Druggable Genetic Anomalies in Acute Myeloid Leukemia: From Diagnostic Tools to Therapeutic Interventions

Cancers ◽  
2021 ◽  
Vol 13 (18) ◽  
pp. 4698
Author(s):  
Francesco Lanza ◽  
Ali Bazarbachi

Acute myeloid leukemia (AML) is a clonal disorder resulting from acquired somatic mutations in hematopoietic progenitor cells that lead to the dysregulation of differentiation and the proliferation of hematopoietic cells [...]

2021 ◽  
Vol 15 (1) ◽  
pp. 1
Author(s):  
Hendra Asputra

Acute myeloid leukemia (AML) is a disease characterized by neoplastic transformation and abnormality of progenitor cells differentiation from myeloid cells. In AML there is a heterogeneous abnormality in hematopoietic progenitor cells, this abnormality has been identified and affects the balance between proliferation, survival and differentiation of body cells. Mutation of FMS-like tyrosin kinase 3 (FLT3) gene causes overexpression which is one of the most often found mechanism that has been identified could trigger genetic changes that can disturb intracellular signaling tissue in pathogenesis of leukemia. Besides being used as a diagnostic tool, FLT3 overexpression can also be used to assess the prognosis of AML.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3516-3516
Author(s):  
Jens-Uwe Hartmann ◽  
Daniela Braeuer-Hartmann ◽  
Cindy Schödel ◽  
Dennis Gerloff ◽  
Christiane Katzerke ◽  
...  

Abstract Abstract 3516 Mitogen-activated protein kinase (MAPK) pathways are a family of related and sometimes interconnected pathways and one of the most studied. Over the last years, extensive work has established that these proteins play a critical role in G-CSF mediated maturation of neutrophil granulocytes. Understanding the mechanisms by which the MAPK pathways are regulated represents an important area of investigation. MicroRNAs, a class of small non-coding RNAs, have been found to play an important role in the regulation of diverse cellular processes by binding to target mRNAs leading to their translational repression. Deregulation of certain microRNAs, thereby, may lead to disrupted signal pathways, such as MAPK-signaling, and to tumorigenesis. However, the role of microRNAs in hematopoietic differentiation and development of leukemia remains largely unknown. In this study we performed a global screen to identify microRNAs involved in G-CSF-regulated MAPK-pathways in primary human CD34+ hematopoietic progenitor cells. Here we found microRNA-143 (miR-143) to be frequently upregulated in G-CSF stimulated CD34+ cells with a strong correlation to CD15 expression. We could also show the granulopoietic association of miR-143 in several hematopoietic cell line models and acute myeloid leukemia (AML) patient samples. Especially, AML patient samples FAB M4 and M5, which show monocytic phenotypes, had a significant lower expression level of miR-143 compared to the AML FAB types M0, M1, M2, and M3. In general, miR-143 expression was shown to be downregulated in AML patient samples in comparison to normal CD34+ hematopoietic progenitor cells. Most interestingly, we show that miR-143 expression is upregulated in acute promyelocytic leukemia (APL) patients after ATRA treatment. By in silico prediction we found MAPK protein family members (eg. MAPK1, MAPK3 and MAPK7) as predicted targets of miR-143. Western blot analysis of AML patient samples and G-CSF stimulated CD34+ cells clearly show an inverse correlation of miR-143 and MAPK7 (ERK5) protein expression. Finally, by transient overexpression of miR-143 we could show a strong downregulation of ERK protein expression in NB4 cells. Our study suggest that miR-143 upregulation by G-CSF may be an important regulatory step for permitting neutrophil differentiation. MicroRNA-143 blocks ERK5 signaling in G-CSF-induced granulopoiesis of CD34+ hematopoietic stem cells, is downregulated in myelo-monocytic acute myeloid leukemia subtypes, and upregulated after ATRA treatment in APL patients. This information may prove useful for the understanding of conditions in which neutrophil proliferation/differentiation balancing is dysregulated, such as in myeloid leukemia and myelodysplastic disorders. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3937-3937
Author(s):  
Alicia Cole ◽  
Zezhou Wang ◽  
Rachel Mattson ◽  
Etienne Coyaud ◽  
Rose Hurren ◽  
...  

Abstract Caseinolytic protease (ClpP) is a mitochondrial enzyme complex with structural similarity to the cytoplasmic proteasome, but little is known about its function in the mitochondria. We identified ClpP as a potential therapeutic target for acute myeloid leukemia (AML) through an shRNA screen to identify mitochondrial proteins that are necessary for the viability of AML cells. We measured ClpP expression in 511 AML samples and 21 samples of normal CD34+ hematopoietic cells using a reverse phase protein array. ClpP was over-expressed in 45% of primary AML samples and expression occurred across FAB subtypes, cytogenetic risk groups, molecular mutations, and CD34+ expression subsets. Next, we evaluated the effects of ClpP knockdown on the growth and viability of AML cells using 3 independent shRNA constructs in lentiviral vectors. In leukemic cell lines that express high levels of ClpP, (OCI-AML2 and K562), knockdown of ClpP reduced growth and viability by > 80%. Importantly, no changes in growth or viability were observed following knockdown of ClpP in HL60 cells, which have lower basal levels of ClpP expression. As a chemical approach to evaluate the effects of ClpP inhibition on AML and normal hematopoietic cells, we synthesized a beta-lactone bacterial ClpP inhibitor, (3RS,4RS)-3-(non-8-en-1-yl)-4-(2-(pyridin-3-yl)ethyl)oxetan-2-one (also known as A2-32-01). Demonstrating specificity for the target, A2-32-01 inhibited the enzymatic activity of human recombinant ClpP, but not chymotrypsin-, trypsin-, or capsase-like enzymatic activity. A2-32-01 induced cell death in TEX, OCI-AML2, and K562 leukemia cells at concentrations that matched its ability to inhibit ClpP activity. Similar to the genetic studies, A2-32-01 did not kill HL60 cells. A2-32-01 was selectively cytotoxic to primary AML cells expressing ClpP over normal hematopoietic cells or AML cells with low ClpP expression. In addition, A2-32-01 reduced the clonogenic growth and bone marrow engraftment of primary AML cells, demonstrating the ability of ClpP inhibition to target AML progenitor/stem cells. Mechanistically, we demonstrated that A2-32-01 disrupted mitochondrial membrane potential in TEX cells and primary AML samples sensitive to A2-32-01, but not in normal hematopoietic cells. To date, the substrates of the mitochondrial ClpP are unknown. Therefore, we defined the interactome map of ClpP in HEK293 cells using mass spectrometry and the BirA tagging method whereby near-neighbors of ClpP are marked with biotin. Fifty-eight mitochondrial proteins preferentially interacted with ClpP over controls and the proteins were primarily components of the respiratory chain and mitochondrial translation apparatus. Thus, ClpP appears to be important to maintain the integrity of mitochondrial respiration. In support of this hypothesis, genetic or chemical ClpP inhibition was cytotoxic to 143B rhabdomyosarcoma cells, but not their rho-zero counterparts that lack mitochondrial DNA and oxidative phosphorylation. Next, we evaluated whether ClpP was required for the growth of AML cells in vivo. We knocked down ClpP in TEX cells with shRNA and injected the cells into the femur of NSGF mice. Compared to cells infected with control shRNA, knockdown of ClpP significantly reduced the engraftment of the cells (control shRNA 15.12 ± 4.576 % vs. ClpP knockdown 0.6180 ± 0.1976 % engraftment). Finally, to evaluate the toxicity of ClpP inhibition, we generated ClpP -/- mice. ClpP -/- mice were viable with normal peripheral blood counts and hematopoietic progenitor cells isolated from their bone marrow showed no significant reduction in clonogenic growth compared to those from wild type mice. Moreover, the abundance of Lin-, Sca-1+, c-kit+ hematopoietic progenitor cells in the bone marrow of ClpP -/- mice was equivalent to that in ClpP +/+ controls. Thus, these data suggest that ClpP inhibition can effectively target a subset of AML, while sparing normal hematopoietic cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1646-1646
Author(s):  
Marijn A. Gillissen ◽  
Martijn Kedde ◽  
Greta de Jong ◽  
Etsuko Yasuda ◽  
Sophie E. Levie ◽  
...  

Abstract Background: Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are high-risk diseases with a poor prognosis. Even with intensive treatment regimens less than 50% of patients can be cured, and for the majority of patients - those over 65 years of age and/or patients with comorbidities - such intensive regimens are not feasible. Novel therapeutic approaches such as immunotherapy directed against a specific tumor target are highly needed. Aims: The aim of our study was to identify antibodies that are highly specific for AML and to discover novel tumor-specific antigens, widely expressed on AML and MDS but not on healthy hematopoietic and non-hematopoietic cells. Methods: Allogeneic bone marrow transplantation is an immunotherapy with proven therapeutic efficacy. We selected a patient with high-risk AML who remained disease free, now more than 5 years after receiving an allogeneic HSCT and therefore can be considered to have mounted a potent graft versus leukemia response. To study the antibody repertoire of this patient we isolated CD27+ IgG+ memory B lymphocytes, about 2 years after the transplant. These cells were transduced with Bcl-6 and Bcl-xL to generate plasmablast B cell clones that produce antibodies and express the B cell receptor on the cell surface. Supernatants of these B cell clones were used to screen for binding to surface antigens on the AML cell line THP-1. Results: We identified an donor derived IgG1 antibody, AT1413, that specifically bound to AML cell lines THP-1, MOLM-13, SH-2 and others, but not to normal bone marrow cells or non-hematopoietic cells. The antibody also interacted with AML blasts from the allogeneic HSCT patient from whom the antibody was derived, and with leukemic blasts isolated from newly diagnosed AML and MDS patients. Biochemical analysis revealed that AT1413 recognizes a sialylated epitope on CD43 which is specifically expressed on all types of AML and MDS cells, as illustrated by its reactivity with blasts of each of 60 randomly selected AML and MDS patients in our clinic. Since the target it is also expressed by CD34+ hematopoietic progenitor cells obtained from fetal liver and fetal bone marrow, but not by post-natal hematopoietic progenitor cells, it can be considered to be a oncofetal epitope. AT1413 induced antibody-dependent cell-mediated cytotoxicity and complement dependent cytotoxicity of AML cell lines and primary blasts. Summary and conclusion: We have identified oncofetal-sialylated CD43 (CD43os) as a novel tumor-specific target that is widely expressed on AML and MDS blasts. Antibodies against this target have therefore high potential as therapeutic antibodies, either as a naked antibody or manufactured into an antibody-drug conjugate, bispecific T cell engager or CAR (chimeric antigen receptor) T cell. Disclosures Kersten: Celgene: Research Funding; Amgen: Honoraria.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 724-724
Author(s):  
Michihiro Kobayashi ◽  
Yunpeng Bai ◽  
Sisi Chen ◽  
Sarah C Nabinger ◽  
Chonghua Yao ◽  
...  

Abstract Acute myeloid leukemia (AML) is a heterogeneous disease with multiple signaling pathways contributing to its pathogenesis. Mutations in receptor tyrosine kinase KIT and FLT3 are found in approximately 40% of AML patients and targeted therapies for inhibiting KIT and FLT3 have failed, thus new targets for therapeutic intervention need to be identified. The phosphatase of regenerating liver (PRL) family of phosphatases, consisting of PRL1, PRL2, and PRL3, represents an intriguing group of proteins being validated as biomarkers and therapeutic targets in human cancer. While PRL2 is highly expressed in some subtypes of human AML, including AML1-ETO+ AML and AML with mixed lineage leukemia (MLL) translocations, its role in AML is largely unknown. To determine the role of PRL2 in the pathogenesis of AML, we utilized two murine models of human AML induced by transducing mouse HSCs with AML1-ETO or MLL-AF9. We found that PRL2 is important for the progression and maintenance of leukemia induced by AML1-ETO or MLL-AF9 through enhancing leukemia stem cell (LSC) self-renewal. To elucidate the mechanisms by which PRL2 promotes LSC maintenance, we performed genome wide RNA-seq analysis of MLL-AF9+ LSCs. Gene Set Enrichment Analysis (GESA) indicates that PRL2 deficiency alters the MLL-AF9 signature essential for LSC self-renewal. We have recently identified PRL2 to be important for the proliferation and self-renewal of hematopoietic stem cells (HSCs) through the regulation of KIT signaling. Notably, PRL2 null hematopoietic progenitor cells showed decreased KIT phosphorylation as well as ERK phosphorylation following SCF stimulation, suggesting that PRL2 is important for KIT activation. Given that KIT inactivation could be mediated by removal from the cell surface and intracellular degradation, we reasoned that PRL2 may regulate KIT receptor internalization and stability. That was indeed the case. We found that the KIT protein half-life in PRL2 null hematopoietic progenitor cells (Kit+) was significantly decreased compared to WT cells. Furthermore, PRL2 null progenitor cells showed enhanced KIT ubiquitination compared to WT cells and less KIT was found on the surface of PRL2 null progenitor cells compared to WT cells following SCF stimulation. We also found that loss of PRL2 in human AML cells resulted in enhanced internalization of KIT. These observations demonstrate that PRL2 deficiency results in less KIT on the cell surface and a lower global KIT level in the cell. Upon SCF stimulation, KIT binds to and induces the phosphorylation of CBL proteins, which in turn act as E3 ligases, mediating the ubiquitination and degradation of KIT. To understand how PRL2 modulates the turnover of KIT in hematopoietic cells, we performed GST-pulldown assays and found that the substrate-trapping mutant PRL2/CS-DA showed an increased association with KIT and CBL compared to wild-type PRL2 in Kasumi-1 cells, suggesting that KIT and CBL may be PRL2 substrates. Furthermore, we found that PRL2/CS-DA mutant showed enhanced association with FLT3 and CBL compared to wild-type PRL2 in MV4-11 cells. Our data suggest that PRL2 dephosphorylates CBL and inhibits CBL activity toward KIT and FLT3, leading to sustained activation of downstream signaling pathways. To determine the functional significance of PRL2 in human AML with KIT and FLT3 mutations, we utilized two well-established murine model of myeloproliferative neoplasms (MPN) induced by KITD814V or FLT3-ITD. We found that loss of Prl2 decreased the ability of oncogenic KITD814V and FLT3-ITD to promote mouse hematopoietic stem and progenitor cell (HSPC) proliferation in vitro andthe development of MPN in vivo. Furthermore, we found that genetic and pharmacological inhibition of PRL2 decreased the proliferation and survival of human AML cells bearing KIT or FLT3 mutations. Taken together, we demonstrate that PRL2 promotes leukemia stem cell (LSC) self-renewal and maintenance through sustaining the activity of oncogenic KIT and FLT3 signals. Our findings suggest that pharmacological inhibition of PRL2 holds potential as a novel therapy for acute myeloid leukemia, and might also be applicable to the treatment of other human cancers. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2736-2736
Author(s):  
Tina Nilsson ◽  
Ahmed Waraky ◽  
Anders Östlund ◽  
Laleh Arabanian ◽  
Julia Asp ◽  
...  

Introduction Acute myeloid leukemia (AML) is the result of aberrant hematopoietic processes, such as enhanced proliferation, blocked differentiation, and dysregulated apoptosis of hematopoietic stem and progenitor cells, and frequently these changes are initiated by chromosomal translocations in leukemia. Efficient methods for modelling leukemia and to recreate leukemia-associated genetic aberrations, such as chromosome translocations, are therefore crucial for investigating how leukemia is initiated. Today, most such models are murine and usually based on introduction of fusion gene transcripts of interest under the control of a constitutive active promoter using lenti- or retroviral transduction rather than the chromosomal translocation itself. The aim of the current project was to create a human cellular model of a chromosomal translocation that is typically found in AML in children under 24 months of age, the translocation t(7;12)(q36;p13). This translocation has been associated with poor prognosis, and leads to a gene fusion MNX1-ETV6 but also aberrant MNX1 gene expression. Its mechanism for leukemia initiation is so far unknown, mainly due to lack of a suitable experimental model. Material and methods CRISPR/Cas9 was used to reconstruct the genetics of the t(7;12)(q36;p13) rearrangement in human induced pluripotent stem cells (iPSC) while maintaining the genomic architecture and regulatory elements. Ribonucleoprotein (RNP) complex was delivered by lipofection (Nucleofection, Amaxa 4D system) into undifferentiated iPSC (ChiPSC 22, Cellartis). An ATTO550 tag on tracrRNA/RNP complex was used to sort out positive cells by flow cytometry and then seeded as single-cells in 96-well plates. Genomic DNA from the single-cell derived iPSC clones were screened by PCR for the presence of the translocation and positive clones were verified with a FISH probe specific for t(7;12)(q36;p13) (Double Fusion Break Apart probe, Metasystem). RT-qPCR was used to detect and quantify the expression of MNX1-ETV6 fusion and MNX1 transcripts. Differentiation potential was tested with the Trilineage Differentiation and Hematopoietic Kits (STEMdiff, STEMCELL Technologies). Results Using CRISPR/Cas9, we could successfully generate iPSC with the t(7;12)(q36;p13) translocation. The translocation was confirmed using conventional karyotyping and FISH and the mRNA expression of the fusion was confirmed with RT-qPCR. No additional chromosomal aberrations were seen. The t(7;12)(q36;p13) iPSC showed similar growth and differentiation properties as the parental iPSC. They showed propensity to differentiate into all three germ layers, confirming their pluripotent stem cell properties. The potential for differentiation into hematopoietic progenitor cells was shown by expression of CD34+, CD43+ and CD45+. In AML with t(7;12)(q36;p13), MNX1 mRNA expression is increased and this may play a role for leukemia development. In the t(7;12)(q36;p13) iPSC, RT-qPCR indeed showed increased expression of MNX1 expression compared with iPSC without the translocation. This increase of MNX1 was not seen in murine adult bone marrow or fetal liver cells transduced with retrovirus expressing the MNX1-ETV6 fusion. Further characterization of the t(7;12)(q36;p13) iPSC, e.g. whole exome and transcriptome sequencing and engraftment potential in immunocompromised mice (NSG-SGM), is ongoing. Conclusion In summary, we have using CRISPR/Cas9 successfully created a t(7;12)(q36;p13) iPSC line with potential to differentiate into hematopoietic progenitor cells and with gene expression pattern similar to what is seen in human AML samples with the t(7;12)(q36;p13). The introduction of the MNX1-ETV6 fusion in its correct genomic context could recapitulate local gene regulation, making it superior to models based on lenti- or retroviral introduction of fusion genes transcripts. In conclusion, this created cell line will be a valuable tool to study the mechanisms behind t(7;12)(q36;p13) AML, a severe form of AML associated with poor prognosis. Disclosures No relevant conflicts of interest to declare.


Haematologica ◽  
2007 ◽  
Vol 92 (4) ◽  
pp. 542-545 ◽  
Author(s):  
G. Bug ◽  
K. Schwarz ◽  
C. Schoch ◽  
M. Kampfmann ◽  
R. Henschler ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document