Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Med Mycol J ; 65(1): 17-21, 2024.
Article in English | MEDLINE | ID: mdl-38417883

ABSTRACT

Disseminated trichosporonosis is a rare fungal infection whose risk factors are hematological malignancies and neutropenia. Recently, breakthrough Trichosporon infections after administration of micafungin, the first-line systemic antifungal agent in compromised hosts, have been widely recognized. A man in his seventies about 1 month into chemotherapy for acute megakaryoblastic leukemia presented with a worsening fever and dyspnea. The patient was being administered with empirical micafungin therapy for suspected candidiasis. As the symptoms progressed, scattered erythema appeared on the trunk, some with a dark red vesicle at the center. Blood cultures identified Trichosporon asahii, as did the specimen of the skin biopsy. On the basis also of the presence of pneumonia on chest computed tomography, we confirmed the diagnosis of disseminated trichosporonosis and changed the antifungal agent from micafungin to voriconazole. Blood culture turned out to be negative 1 month after administrating voriconazole. However, the patient died of the leukemia. Our review of previous reports on cutaneous manifestations of disseminated trichosporonosis revealed that despite their morphological diversity, erythema with a red papule or vesicle at the center, implying necrosis, was also observed in previous cases. Our case report suggests that dermatologists should be aware of skin manifestations of disseminated trichosporonosis after micafungin administration, especially in cases of hematological malignancies.


Subject(s)
Hematologic Neoplasms , Leukemia, Megakaryoblastic, Acute , Trichosporon , Trichosporonosis , Male , Humans , Micafungin , Antifungal Agents/therapeutic use , Voriconazole , Trichosporonosis/diagnosis , Trichosporonosis/drug therapy , Trichosporonosis/microbiology , Leukemia, Megakaryoblastic, Acute/complications , Leukemia, Megakaryoblastic, Acute/drug therapy , Hematologic Neoplasms/complications , Hematologic Neoplasms/drug therapy , Erythema/complications , Erythema/drug therapy
2.
Blood Adv ; 8(1): 112-129, 2024 01 09.
Article in English | MEDLINE | ID: mdl-37729615

ABSTRACT

ABSTRACT: Acute megakaryoblastic leukemia (AMKL) is a rare, developmentally restricted, and highly lethal cancer of early childhood. The paucity and hypocellularity (due to myelofibrosis) of primary patient samples hamper the discovery of cell- and genotype-specific treatments. AMKL is driven by mutually exclusive chimeric fusion oncogenes in two-thirds of the cases, with CBFA2T3::GLIS2 (CG2) and NUP98 fusions (NUP98r) representing the highest-fatality subgroups. We established CD34+ cord blood-derived CG2 models (n = 6) that sustain serial transplantation and recapitulate human leukemia regarding immunophenotype, leukemia-initiating cell frequencies, comutational landscape, and gene expression signature, with distinct upregulation of the prosurvival factor B-cell lymphoma 2 (BCL2). Cell membrane proteomic analyses highlighted CG2 surface markers preferentially expressed on leukemic cells compared with CD34+ cells (eg, NCAM1 and CD151). AMKL differentiation block in the mega-erythroid progenitor space was confirmed by single-cell profiling. Although CG2 cells were rather resistant to BCL2 genetic knockdown or selective pharmacological inhibition with venetoclax, they were vulnerable to strategies that target the megakaryocytic prosurvival factor BCL-XL (BCL2L1), including in vitro and in vivo treatment with BCL2/BCL-XL/BCL-W inhibitor navitoclax and DT2216, a selective BCL-XL proteolysis-targeting chimera degrader developed to limit thrombocytopenia in patients. NUP98r AMKL were also sensitive to BCL-XL inhibition but not the NUP98r monocytic leukemia, pointing to a lineage-specific dependency. Navitoclax or DT2216 treatment in combination with low-dose cytarabine further reduced leukemic burden in mice. This work extends the cellular and molecular diversity set of human AMKL models and uncovers BCL-XL as a therapeutic vulnerability in CG2 and NUP98r AMKL.


Subject(s)
Antineoplastic Agents , Leukemia, Megakaryoblastic, Acute , Humans , Child , Child, Preschool , Animals , Mice , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Proteomics , Transcription Factors , Proto-Oncogene Proteins c-bcl-2 , Repressor Proteins
3.
Blood Rev ; 64: 101154, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38016838

ABSTRACT

Children with Down syndrome (DS) have a 10- to 20-fold greater predisposition to develop acute leukemia compared to the general population, with a skew towards myeloid leukemia (ML-DS). While ML-DS is known to be a subtype with good outcome, patients who relapse face a dismal prognosis. Acute lymphocytic leukemia in DS (DS-ALL) is considered to have poor prognosis. The relapse rate is high in DS-ALL compared to their non-DS counterparts. We have a better understanding about the mutational spectrum of DS leukemia. Studies using animal, embryonic stem cell- and induced pluripotent stem cell-based models have shed light on the mechanism by which these mutations contribute to disease initiation and progression. In this review, we list the currently available treatment strategies for DS-leukemias along with their outcome with emphasis on challenges with chemotherapy-related toxicities in children with DS. We focus on the mechanisms of initiation and progression of leukemia in children with DS and highlight the novel molecular targets with greater success in preclinical trials that have the potential to progress to the clinic.


Subject(s)
Down Syndrome , Leukemia, Megakaryoblastic, Acute , Child , Animals , Humans , Down Syndrome/complications , Down Syndrome/genetics , Down Syndrome/therapy , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , GATA1 Transcription Factor/genetics , Mutation , Recurrence , Biology
4.
Cell Rep ; 42(9): 113084, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37716355

ABSTRACT

Pediatric acute megakaryoblastic leukemia (AMKL) is an aggressive blood cancer associated with poor therapeutic response and high mortality. Here we describe the development of CBFA2T3-GLIS2-driven mouse models of AMKL that recapitulate the phenotypic and transcriptional signatures of the human disease. We show that an activating Ras mutation that occurs in human AMKL increases the penetrance and decreases the latency of CBF2AT3-GLIS2-driven AMKL. CBFA2T3-GLIS2 and GLIS2 modulate similar transcriptional networks. We identify the dominant oncogenic properties of GLIS2 that trigger AMKL in cooperation with oncogenic Ras. We find that both CBFA2T3-GLIS2 and GLIS2 alter the expression of a number of BH3-only proteins, causing AMKL cell sensitivity to the BCL2 inhibitor navitoclax both in vitro and in vivo, suggesting a potential therapeutic option for pediatric patients suffering from CBFA2T3-GLIS2-driven AMKL.


Subject(s)
Leukemia, Megakaryoblastic, Acute , Animals , Mice , Child , Humans , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Aniline Compounds , Sulfonamides , Oncogene Proteins, Fusion/metabolism , Repressor Proteins
5.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3101-3110, 2023 11.
Article in English | MEDLINE | ID: mdl-37162543

ABSTRACT

Acute megakaryocytic leukemia (AMKL) is a rare neoplasm caused by abnormal megakaryoblasts. Megakaryoblasts keep dividing and avoid undergoing polyploidization to escape maturation. Small-molecule probes inducing polyploidization of megakaryocytic leukemia cells accelerate the differentiation of megakaryocytes. This study aims to determine that Rho kinase (ROCK) inhibition on megakaryoblasts enhances polyploidization and the inhibition of ROCK1 by fasudil benefits AMKL mice. The study investigated fasudil on the megakaryoblast cells in vitro and in vivo. With the differentiation and apoptosis induction, fasudil was used to treat 6133/MPLW515L mice, and the differentiation level was evaluated. Fasudil could reduce proliferation and promote the polyploidization of megakaryoblasts. Meanwhile, fasudil reduced the disease burden of 6133/MPLW515L AMKL mice at a dose that is safe for healthy mice. Combination therapy of ROCK1 inhibitor fasudil and reported clinical AURKA inhibitor MLN8237 achieved a better antileukemia effect in vivo, which alleviated hepatosplenomegaly and promoted the differentiation of megakaryoblast cells. ROCK1 inhibitor fasudil is a good proliferation inhibitor and polyploidization inducer of megakaryoblast cells and might be a novel rationale for clinical AMKL treatment.


Subject(s)
Leukemia, Megakaryoblastic, Acute , Megakaryocytes , Animals , Mice , Megakaryocytes/physiology , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Megakaryocyte Progenitor Cells , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , rho-Associated Kinases
6.
Blood ; 141(13): 1610-1625, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36508699

ABSTRACT

Myeloid neoplasms with erythroid or megakaryocytic differentiation include pure erythroid leukemia, myelodysplastic syndrome with erythroid features, and acute megakaryoblastic leukemia (FAB M7) and are characterized by poor prognosis and limited treatment options. Here, we investigate the drug sensitivity landscape of these rare malignancies. We show that acute myeloid leukemia (AML) cells with erythroid or megakaryocytic differentiation depend on the antiapoptotic protein B-cell lymphoma (BCL)-XL, rather than BCL-2, using combined ex vivo drug sensitivity testing, genetic perturbation, and transcriptomic profiling. High-throughput screening of >500 compounds identified the BCL-XL-selective inhibitor A-1331852 and navitoclax as highly effective against erythroid/megakaryoblastic leukemia cell lines. In contrast, these AML subtypes were resistant to the BCL-2 inhibitor venetoclax, which is used clinically in the treatment of AML. Consistently, genome-scale CRISPR-Cas9 and RNAi screening data demonstrated the striking essentiality of BCL-XL-encoding BCL2L1 but not BCL2 or MCL1, for the survival of erythroid/megakaryoblastic leukemia cell lines. Single-cell and bulk transcriptomics of patient samples with erythroid and megakaryoblastic leukemias identified high BCL2L1 expression compared with other subtypes of AML and other hematological malignancies, where BCL2 and MCL1 were more prominent. BCL-XL inhibition effectively killed blasts in samples from patients with AML with erythroid or megakaryocytic differentiation ex vivo and reduced tumor burden in a mouse erythroleukemia xenograft model. Combining the BCL-XL inhibitor with the JAK inhibitor ruxolitinib showed synergistic and durable responses in cell lines. Our results suggest targeting BCL-XL as a potential therapy option in erythroid/megakaryoblastic leukemias and highlight an AML subgroup with potentially reduced sensitivity to venetoclax-based treatments.


Subject(s)
Leukemia, Megakaryoblastic, Acute , Leukemia, Myeloid, Acute , Lymphoma, B-Cell , Animals , Mice , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Cell Line, Tumor , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , bcl-X Protein/genetics , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Cell Differentiation , Apoptosis
7.
Zhonghua Xue Ye Xue Za Zhi ; 43(6): 499-505, 2022 Jun 14.
Article in Chinese | MEDLINE | ID: mdl-35968594

ABSTRACT

Objective: To investigate the effect of blocking P21 activated kinase 1 (PAK1) activity on the proliferation, differentiation, and apoptosis of acute megakaryocytic leukemia (AMKL) cell lines (CHRF and CMK) . Methods: Cell counts were used to detect the effects of PAK1 inhibitors (IPA-3 and G5555) on AMKL cell proliferation inhibition and colony formation, and flow cytometry was used to detect its effects on AMKL cell cycle. The effect of PAK1 inhibitor on the expression of cyclin D1 and apoptosis-related protein Cleaved caspase 3 was detected using Western blot, while interference with the protein expression level of PAK1 in AMKL cells was assessed using lentivirus-mediated shRNA transfection technology. Flow cytometry was used to detect the effects of knockdown of PAK1 kinase activity on the ability of polyploid DNA formation and cell apoptosis in AMKL cells. Results: PAK1 inhibitors inhibited the proliferation of AMKL cells in a dose-dependent manner and reduced the ability of cell colony formation, and the difference was statistically significant when compared with the control group (P<0.05) . Moreover, they also reduced the percentage of AMKL cells in S phase, and Western blot detection showed that the expression levels of phosphorylated PAK1 and cyclin D1 decreased significantly. Finally, PAK1 inhibitors induced AMKL cell apoptosis by up-regulating Cleaved caspase 3 and showed different abilities to increase the content of polyploid DNA in megakaryocytes. Only high concentrations of IPA-3 and low doses of G5555 increased the number of polyploid megakaryocytes, while knockdown of PAK1 kinase activity promoted AMKL cell differentiation and increased the apoptosis rate. Conclusion: PAK1 inhibitor significantly arrests AMKL cell growth and promotes cell apoptosis. Knocking down the expression of PAK1 promotes the formation of polyploid DNA and induces AMKL cell apoptosis. The above findings indicate that inhibiting the activity of PAK1 may control AMKL effectively.


Subject(s)
Leukemia, Megakaryoblastic, Acute , p21-Activated Kinases , Apoptosis , Caspase 3/metabolism , Cell Differentiation , Cell Line, Tumor , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/metabolism , Polyploidy , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
8.
Leuk Res ; 120: 106920, 2022 09.
Article in English | MEDLINE | ID: mdl-35872339

ABSTRACT

Acute megakaryoblastic leukaemia (AMKL) is characterized by expansion of megakaryoblasts, which are hyper-proliferative cells that fail to undergo differentiation. Insight to the cell-cycle regulation revealed important events in early or late megakaryocytes (MKs) maturation; the cyclin-dependent kinases 4 and 6 (CDK4/6) have been reported to participate in the development of progenitor megakaryocytes, mainly by promoting cell cycle progression and DNA polyploidization. However, it remains unclear whether the continuous proliferation, but not differentiation, of megakaryoblasts is related to an aberrant regulation of CDK4/6 in AMKL. Here, we found that CDK4/6 were up regulated in patients with AMKL, and persistently maintained at a high level during the differentiation of abnormal megakaryocytes in vitro, according to a database and western blot. Additionally, AMKL cells were exceptionally reliant on the cell cycle regulators CDK4 or 6, as blocking their activity using an inhibitor or short hairpin RNA (shRNA) significantly reduced the proliferation of 6133/MPL megakaryocytes, reduced DNA polyploidy, induced apoptosis, decreased the level of phosphorylated retinoblastoma protein (p-Rb), and activation of caspase 3. Additionally, CDK4/6 inhibitors and shRNA reduced the numbers of leukemia cells in the liver and bone marrow (BM), alleviated hepatosplenomegaly, and prolonged the survival of AMKL-transplanted mice. These results suggested that blocking the activity of CDK4/6 may represent an effective approach to control megakaryoblasts in AMKL.


Subject(s)
Leukemia, Megakaryoblastic, Acute , Animals , Cyclins , DNA , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Megakaryocyte Progenitor Cells , Mice , RNA, Small Interfering
9.
Cancer Immunol Immunother ; 70(12): 3477-3488, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33890137

ABSTRACT

Acute megakaryocytic leukemia (AMKL) is one of the rarest sub-types of acute myeloid leukemia (AML). AMKL is characterized by high proliferation of megakaryoblasts and myelofibrosis of bone marrow, this disease is also associated with poor prognosis. Previous analyses have reported that the human megakaryoblastic cells can be differentiated into cells with megakaryocyte (MK)-like characteristics by phorbol 12-myristate 13-acetate (PMA). However, little is known about the mechanism responsible for regulating this differentiation process. We performed long non-coding RNA (lncRNA) profiling to investigate the differently expressed lncRNAs in megakaryocyte blast cells treated with and without PMA and examined those that may be responsible for the PMA-induced differentiation of megakaryoblasts into MKs. We found 30 out of 90 lncRNA signatures to be differentially expressed after PMA treatment of megakaryoblast cells, including the highly expressed JPX lncRNA. Further, in silico lncRNA-miRNA and miRNA-mRNA interaction analysis revealed that the JPX is likely involved in unblocking the expression of TGF-ß receptor (TGF-ßR) by sponging oncogenic miRNAs (miR-9-5p, miR-17-5p, and miR-106-5p) during MK differentiation. Further, we report the activation of TGF-ßR-induced non-canonical ERK1/2 and PI3K/AKT pathways during PMA-induced MK differentiation and ploidy development. The present study demonstrates that TGF-ßR-induced non-canonical ERK1/2 and PI3K/AKT pathways are associated with PMA-induced MK differentiation and ploidy development; in this molecular mechanism, JPX lncRNA could act as a decoy for miR-9-5p, miR-17-5p, and miR-106-5p, titrating them away from TGF-ßR mRNAs. Importantly, this study reveals the activation of ERK1/2 and PI3K/AKT pathway in PMA-induced Dami cell differentiation into MK. The identified differentially expressed lncRNA signatures may facilitate further study of the detailed molecular mechanisms associated with MK development. Thus, our data provide numerous targets with therapeutic potential for the modulation of the differentiation of megakaryoblastic cells in AMKL.


Subject(s)
Leukemia, Megakaryoblastic, Acute/drug therapy , Megakaryocytes/drug effects , Phorbol Esters/pharmacology , RNA, Long Noncoding/drug effects , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukemia, Megakaryoblastic, Acute/genetics , MAP Kinase Signaling System/drug effects , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Long Noncoding/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta/genetics
10.
J Pediatr Hematol Oncol ; 43(8): e1164-e1167, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-33122587

ABSTRACT

BACKGROUND: There is no established effective treatment for patients with t(1;22)(p13;q13) acute megakaryoblastic leukemia (AMKL) and hepatic fibrosis. OBSERVATION: Here we report the outcomes of 2 t(1;22)(p13;q13) AMKL patients with hepatic fibrosis. One patient died from liver failure despite the control of leukemia. The other patient was successfully treated with reduced-intensity chemotherapy and antifibrosis therapy with tretinoin and α-tocopheryl acetate, the hepatic fibrosis resolved and leukemia was in remission for 3 years. CONCLUSIONS: Reduced-intensity chemotherapy plus antifibrosis therapy with tretinoin and α-tocopheryl acetate could be a treatment option for these patients with t(1;22)(p13;q13) AMKL and hepatic fibrosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 22/genetics , Leukemia, Megakaryoblastic, Acute/drug therapy , Liver Cirrhosis/drug therapy , Translocation, Genetic , Tretinoin/therapeutic use , alpha-Tocopherol/therapeutic use , Antioxidants/therapeutic use , Child, Preschool , Drug Therapy, Combination , Female , Humans , Infant, Newborn , Keratolytic Agents/therapeutic use , Leukemia, Megakaryoblastic, Acute/complications , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Prognosis
13.
Trends Cancer ; 5(9): 547-557, 2019 09.
Article in English | MEDLINE | ID: mdl-31474360

ABSTRACT

GLI-similar 1-3 (GLIS1-3), a subfamily of Krüppel-like zinc finger transcription factors, function as key regulators of several biological processes important to oncogenesis, including control of cell proliferation, differentiation, self-renewal, and epithelial-mesenchymal transition. This review provides a short overview of the critical roles genetic changes in GLIS1-3 play in the development of several malignancies. This includes intrachromosomal translocations involving GLIS2 and ETO2/CBFA2T3 in the development of pediatric non-Down's syndrome (DS), acute megakaryoblastic leukemia (AMKL), a malignancy with poor prognosis, and an association of interchromosomal translocations between GLIS3, GLIS1, and PAX8, and between GLIS3 and CLPTM1L with hyalinizing trabecular tumors (HTTs) and fibrolamellar hepatocellular carcinoma (FHCC), respectively. Targeting upstream signaling pathways that regulate GLIS signaling may offer new therapeutic strategies in the management of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Liver Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Thyroid Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Leukemic , Gene Rearrangement , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia, Megakaryoblastic, Acute/drug therapy , Liver Neoplasms/drug therapy , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/metabolism , PAX8 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Thyroid Neoplasms/drug therapy , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers/genetics
14.
Pediatr Blood Cancer ; 66(8): e27785, 2019 08.
Article in English | MEDLINE | ID: mdl-31044544

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous disease regarding morphology, immunophenotyping, genetic abnormalities, and clinical behavior. The overall survival rate of pediatric AML is 60% to 70%, and has not significantly improved over the past two decades. Children with Down syndrome (DS) are at risk of developing acute megakaryoblastic leukemia (AMKL), which can be preceded by a transient myeloproliferative disorder during the neonatal period. Intensification of current treatment protocols is not feasible due to already high treatment-related morbidity and mortality. Instead, more targeted therapies with less severe side effects are highly needed. PROCEDURE: To identify potential novel therapeutic targets for myeloid disorders in children, including DS-AMKL and non-DS-AML, we performed an unbiased compound screen of 80 small molecules targeting epigenetic regulators in three pediatric AML cell lines that are representative for different subtypes of pediatric AML. Three candidate compounds were validated and further evaluated in normal myeloid precursor cells during neutrophil differentiation and in (pre-)leukemic pediatric patient cells. RESULTS: Candidate drugs LMK235, NSC3852, and bromosporine were effective in all tested pediatric AML cell lines with antiproliferative, proapoptotic, and differentiation effects. Out of these three compounds, the pan-histone deacetylase inhibitor NSC3852 specifically induced growth arrest and apoptosis in pediatric AML cells, without disrupting normal neutrophil differentiation. CONCLUSION: NSC3852 is a potential candidate drug for further preclinical testing in pediatric AML and DS-AMKL.


Subject(s)
Drug Screening Assays, Antitumor/methods , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Hydroxyquinolines/pharmacology , Leukemia, Myeloid, Acute/pathology , Nitroso Compounds/pharmacology , Apoptosis , Cell Proliferation , Child , Down Syndrome/drug therapy , Down Syndrome/genetics , Down Syndrome/pathology , High-Throughput Screening Assays , Histone Deacetylases/genetics , Humans , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemoid Reaction/drug therapy , Leukemoid Reaction/genetics , Leukemoid Reaction/pathology , Prognosis , Tumor Cells, Cultured
15.
Medicine (Baltimore) ; 97(27): e11331, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29979407

ABSTRACT

RATIONALE: The JAK2 V617F mutation is frequently found in ET, while it is rare in de novo AML. ET has a low frequency of leukemic transformation. Both secondary AML (sAML) from ET and AML with JAK2 V617F mutation have poor prognoses. Because of the low incidence of JAK2 mutation in acute myeloid leukemia (AML), the clinical features of AML with JAK2 mutation are rarely reported so far, either transformed from essential thrombocythemia (ET) or de novo AML. PATIENT CONCERNS: In this article, we present a pediatric AML patient with the JAK2 V617F mutation. DIAGNOSES: A diagnosis of acute megakaryoblastic leukemia was made and sAML was ruled out. INTERVENTIONS: The patient underwent chemotherapy. OUTCOMES: In the first two complete remission periods, we found significantly increased numbers of platelets and bone marrow megakaryocytes, which are characteristic of ET. After the third chemotherapy phase, the disease relapsed; the platelet count was reduced and continued to decrease. When disease relapsed, her family abandoned treatment. LESSONS: These observations of our case raise two possibilities: either transient posttreatment thrombocythemia is a feature of AML with JAK2 V617F mutation, or this was a case of secondary AML. Additional information is required to reach better conclusions on the connection between AML and JAK2 mutations.


Subject(s)
Leukemia, Megakaryoblastic, Acute/diagnosis , Leukemia, Myeloid, Acute/diagnosis , Thrombocythemia, Essential/diagnosis , Antineoplastic Agents/therapeutic use , Diagnosis, Differential , Fatal Outcome , Female , Humans , Infant , Janus Kinase 2/genetics , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Platelet Count , Thrombocythemia, Essential/drug therapy
16.
Anticancer Agents Med Chem ; 18(10): 1440-1447, 2018.
Article in English | MEDLINE | ID: mdl-29623856

ABSTRACT

BACKGROUND: Many studies suggested that Acetylcholine (ACh) might serve as an autocrine/ paracrine growth factor in several types of tumors or tumor cell lines. High levels of Acetylcholinesterase (AChE) activity have been reported in primary brain tumors, ovarian, colon and lung tumors. OBJECTIVES: The role of cholinergic signaling needs to be clarified in in leukemia. METHOD: K562 cells were derived from a chronic myelogenous leukemia patient during blast crisis serving as pluripotent hematopoietic stem cells. K562 cells were incubated with various cholinergic agonists or antagonists to investigate the role of ACh in different differentiated cell lines. RESULTS: Our experiments showed that AChE activity was increased in response to ACh in undifferentiated K562 cells, but in the erythroid differentiated K562 cells a high concentration of ACh (1 mM) decreased the AChE activity. ACh failed to elevate the AChE activity in the megakaryocytic differentiated K562 cells. An AChE inhibitor, eserine, also suppressed the AChE activity in a concentration-dependent manner. Choline uptake inhibition by hemicholinium did increase the AChE activity but not in the erythroid differentiated K562 cell line. Likewise, megakaryocytic differentiated K562 cells also displayed a similar pattern. Vesamicole, a vesicular choline uptake inhibitor, produced similar results. Curare, a nicotinic antagonist, elevated the cell counts of the megakaryocytic differentiated cells. CONCLUSION: Our findings may suggest excess extracellular ACh will decrease the cell growth in undifferentiated and megakaryocytic differentiated K562 cell lines through nicotinic type cholinoceptors.


Subject(s)
Acetylcholine/pharmacology , Antineoplastic Agents/pharmacology , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Acetylcholine/chemistry , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , K562 Cells , Leukemia, Megakaryoblastic, Acute/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Models, Molecular , Molecular Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Genes Chromosomes Cancer ; 57(6): 311-319, 2018 06.
Article in English | MEDLINE | ID: mdl-29427526

ABSTRACT

The advent of large scale genomic sequencing technologies significantly improved the molecular classification of acute megakaryoblastic leukaemia (AMKL). AMKL represents a subset (∼10%) of high fatality pediatric acute myeloid leukemia (AML). Recurrent and mutually exclusive chimeric gene fusions associated with pediatric AMKL are found in 60%-70% of cases and include RBM15-MKL1, CBFA2T3-GLIS2, NUP98-KDM5A and MLL rearrangements. In addition, another 4% of AMKL harbor NUP98 rearrangements (NUP98r), with yet undetermined fusion partners. We report a novel NUP98-BPTF fusion in an infant presenting with primary refractory AMKL. In this NUP98r, the C-terminal chromatin recognition modules of BPTF, a core subunit of the NURF (nucleosome remodeling factor) ATP-dependent chromatin-remodeling complex, are fused to the N-terminal moiety of NUP98, creating an in frame NUP98-BPTF fusion, with structural homology to NUP98-KDM5A. The leukemic blasts expressed two NUP98-BPTF splicing variants, containing one or two tandemly spaced PHD chromatin reader domains. Our study also identified an unreported wild type BPTF splicing variant encoding for 2 PHD domains, detected both in normal cord blood CD34+ cells and in leukemic blasts, as with the fly BPTF homolog, Nurf301. Disease course was marked by rapid progression and primary chemoresistance, with ultimately significant tumor burden reduction following treatment with a clofarabine containing regimen. In sum, we report 2 novel NUP98-BPTF fusion isoforms that contribute to refine the NUP98r subgroup of pediatric AMKL. Multicenter clinical trials are critically required to determine the frequency of this fusion in AMKL patients and explore innovative treatment strategies for a disease still plagued with poor outcomes.


Subject(s)
Antigens, Nuclear/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Nerve Tissue Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Transcription Factors/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Infant , Karyotyping , Leukemia, Megakaryoblastic, Acute/drug therapy , Male , RNA Splicing
18.
Malawi Med J ; 30(4): 298-301, 2018 Dec.
Article in English | MEDLINE | ID: mdl-31798811

ABSTRACT

Acute Megakaryoblastic Leukaemia (AML, M7) is a rare type of acute myeloid leukemia (AML) evolving from primitive megakaryoblasts. It accounted for 1.2% of newly diagnosed AML according to Eastern Cooperative Oncology Group (ECOG) trials between 1984 and 1997. Patients may present with a broad variety of symptoms including low-grade fever, easy bruising, and life-threatening conditions. We report a rare case of AML, M7 in a 19-year-old lady who presented with weakness and fatigue. She was diagnosed as a case of AML, M7 on the basis of peripheral blood finding, bone marrow examination report, radiological findings and immunophenotyping.


Subject(s)
Leukemia, Megakaryoblastic, Acute/diagnosis , Anemia/diagnosis , Anemia/therapy , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Blood Transfusion , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Fatigue/etiology , Female , Humans , Immunophenotyping , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/immunology , Leukemia, Megakaryoblastic, Acute/pathology , Lost to Follow-Up , Muscle Weakness/etiology , Thrombocytopenia/diagnosis , Thrombocytopenia/therapy , Young Adult
19.
J Pharmacol Sci ; 135(1): 14-21, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28927780

ABSTRACT

Metformin has received increasing attention for its potential anticancer activity against certain human leukemia cells, but its effects on human megakaryoblastic cells are unclear. This study aimed to investigate the effects of metformin on proliferation and apoptosis of human megakaryoblastic cells (Dami and MEG-01) and the underlying molecular mechanisms. CCK8 assay was employed to measure cell proliferation. Flow cytometry was adopted to detect cell apoptosis. Western blot was further employed to measure apoptosis-related proteins. In Dami and MEG-01 cells, metformin significantly inhibited proliferation and promoted apoptosis in a dose- and time-dependent manner, and metformin (4 mM) was selected for subsequent experiments. Metformin inhibited ERK1/2, JNK, and PI3K/Akt, but activated p38 pathway in these two cells. Moreover, inhibition of ERK1/2, JNK or PI3K/Akt pathway alone induced cell apoptosis compared to the control group. The combination of specific inhibitors of ERK1/2, JNK or PI3K/Akt pathway and metformin further promoted cell apoptosis and the up-regulation of p21, Bax, Bad, cleaved caspase-3 and -9 as well as the down-regulation of Bcl-2 mediated by metformin alone, but inhibition of p38 pathway exhibited the opposite results. These findings support the possibility of metformin treatment as a new therapeutic strategy against acute megakaryoblastic leukemia (AMKL).


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Hypoglycemic Agents/pharmacology , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/pathology , Metformin/pharmacology , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/therapeutic use , JNK Mitogen-Activated Protein Kinases , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Metformin/therapeutic use , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases , Signal Transduction/drug effects , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases
20.
Br J Pharmacol ; 174(23): 4308-4328, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28901537

ABSTRACT

BACKGROUND AND PURPOSE: The poor prognosis of acute megakaryoblastic leukaemia (AMKL) means there is a need to develop novel therapeutic methods to treat this condition. It was recently shown that inducing megakaryoblasts to undergo terminal differentiation is effective as a treatment for AMKL. This encouraged us to identify a compound that induces megakaryocyte differentiation, which could then act as a potent anti-leukaemia agent. EXPERIMENTAL APPROACH: The effects of tetrandrine on the expression of CD41 and cell morphology were investigated in AMKL cells. We used CRISPR/Cas9 knockout system to knock out ATG7 and verify the role of autophagy in tetrandrine-induced megakaryocyte differentiation. shNotch1 and CA-Akt were transfected into K562 cells to examine the downstream pathways of ROS signalling and the mechanistic basis of the tetrandrine-induced megakaryocyte differentiation. The anti-leukaemia effects of tetrandrine were analysed both in vitro and in vivo. KEY RESULTS: A low dose of tetrandrine induced cell cycle arrest and megakaryocyte differentiation in AMKL cells via activation of autophagy. Molecularly, we demonstrated that this effect is mediated by activation of Notch1 and Akt and subsequent accumulation of ROS. In contrast, in normal mouse fetal liver cells, although tetrandrine induced autophagy, it did not affect cell proliferation or promote megakaryocyte differentiation, suggesting a specific effect of tetrandrine in malignant megakaryoblasts. Finally, tetrandrine also showed in vivo efficacy in an AMKL xenograft mouse model. CONCLUSIONS AND IMPLICATIONS: Modulating autophagy-mediated differentiation may be a novel strategy for treating AMKL, and tetrandrine has the potential to be developed as a differentiation-inducing agent for AMKL chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzylisoquinolines/pharmacology , CRISPR-Cas Systems/genetics , Leukemia, Megakaryoblastic, Acute/drug therapy , Animals , Autophagy/drug effects , Cell Differentiation/drug effects , Female , Gene Knockout Techniques , Humans , K562 Cells , Leukemia, Megakaryoblastic, Acute/genetics , Mice , Mice, Nude , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...