scholarly journals Acute Hypotension after Loxoprofen Sodium Co-administration in Two Acute Myeloid Leukemia Patients Undergoing Chemotherapy Treatment.

Author(s):  
Akito Suzuki ◽  
Jun Moriwaki ◽  
Sachiko Nagae ◽  
Kiyomi Ishiga ◽  
Ichiro Ieiri ◽  
...  
2009 ◽  
Vol 48 (18) ◽  
pp. 1629-1633 ◽  
Author(s):  
Hiroyuki Kobayashi ◽  
Tomohiro Matsuyama ◽  
Masuzu Ueda ◽  
Takahiro Suzuki ◽  
Katsutoshi Ozaki ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 599-599
Author(s):  
Franck Rapaport ◽  
Marc Robert de Massy ◽  
Adil al Hinai ◽  
Mathijs A. Sanders ◽  
Todd Hricik ◽  
...  

Abstract Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. Unfortunately, a significant proportion of patients relapse after responding to initial treatment reflecting our poor understanding of the mechanisms mediating therapy resistance and relapse. We hypothesized that understanding the evolution of the mutational landscape between diagnosis and relapse is essential in order to identify mutational markers associated with sensitivity or resistance to treatment. To address this hypothesis we assembled a cohort of 53 clinically annotated, paired AML patient samples (diagnosis, relapse and patient-matched germline samples; mean age = 52 years). All patients achieved clinical remission after treatment with combination chemotherapy (cytarabine arabinoside and an anthracycline) during induction phase followed by consolidation chemotherapy treatment with or without a stem cell transplantation in first remission. Serial samples were collected at the time of initial diagnosis and within three months of relapse (mean time to relapse 455 days). We performed whole-exome and targeted capture followed by high-throughput sequencing. We aligned samples with BWA, recalibrated them with The Genome Analysis Toolkit (GATK) and then compiled integrated calls from substitution and indel callers (Mutect, Scalpel, Strelka, Varscan and Somatic Sniper). We performed several layers of post-processing filtering on these calls, including removing non-oncogenic mutations and previously documented non-somatic variants, and correcting for the variant allele fraction of indel calls. We filtered out the variants that were found to occur in non-copy number neutral re-arrangements using the clinically determined cytogenetic data. Furthermore, we assessed for copy number events, including loss of heterozygosity events, and for the presence and the variant allele frequency of the FLT3-ITD in our samples. We observed a median of 4.5 and 5 mutations per patient at diagnosis and relapse, respectively, with 3.5 mutations being shared by paired diagnosis and relapse samples. When limiting our analysis to genes previously shown to contribute to leukemogenesis, we found a median of 1.5 and 2 mutations per patient at diagnosis and relapse, with 1 mutation being shared. FLT3, DNMT3A, IDH2, NRAS, RUNX1 and TET2 were among the most commonly mutated genes, with a detected presence rate of 28%, 25%, 19%, 19%, 11% and 11%, respectively, in the diagnosis samples and 39%, 23%, 19%, 4%, 13% and 11% in the relapse samples. We identified significant variation in the variant allele frequency (VAF) for several of the mutations related to these genes and others, denoting variations in the cellular prevalence of the related clones after adjustment for tumor content using the mutations with the highest VAF to delineate clonal architecture. Specifically, we observed that DNMT3A, IDH2, TET2 variants are most commonly present in the bulk AML clone, and persist after treatment. WT1, GATA2 and FLT3mutations are predicted to confer relative resistance to standard combination chemotherapy treatment based on their increased VAF at relapse, whereas KRAS and NRAS subclone(s) are more sensitive to chemotherapy since their VAFs decrease following multiagent chemotherapy. Fifteen patients presented new events in leukemogenesis-related genes at relapse. Overall, our results support a model of AML as a disease with a complex mutational hierarchy and clonal architecture and provide further insight into how these change in response to standard induction therapy. Our data suggests that future efforts to develop targeted therapies with maximal clinical benefit in combination with standard induction treatments should be placed on mutated genes identified to be more strongly associated with disease relapse. Authors contributed equally: F. Rapaport and M.R. De Massy Authors contributed equally: A. al Hinai and M.A. Sanders Disclosures Guzman: Cellectis: Research Funding. Roboz:Cellectis: Research Funding; Agios, Amgen, Amphivena, Astex, AstraZeneca, Boehringer Ingelheim, Celator, Celgene, Genoptix, Janssen, Juno, MEI Pharma, MedImmune, Novartis, Onconova, Pfizer, Roche/Genentech, Sunesis, Teva: Consultancy. Melnick:Janssen: Research Funding. Levine:Qiagen: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy.


2020 ◽  
Vol 91 ◽  
pp. 106339 ◽  
Author(s):  
Elizabeth A. Griffiths ◽  
Hetty E. Carraway ◽  
Namrata S. Chandhok ◽  
Thomas Prebet

2019 ◽  
Vol 8 (2) ◽  
pp. IJH16 ◽  
Author(s):  
Mark W Drummond

Mark Drummond obtained his medical degree from the University of Glasgow (UK) and since then has trained as a hematologist in the West of Scotland and the Canterbury District Health Board (Christchurch, New Zealand) and back at Glasgow University, where he also gained a PhD in chronic myeloid leukemia. He is currently a consultant hematologist and honorary senior lecturer at the Beatson Cancer Centre (Glasgow, UK) as well as an investigator on multiple UK and international clinical trials. Here he talks to Commissioning Editor Jennifer Straiton, commenting on the recent announcement from the Scottish Medicines Consortium, regarding their acceptance of the use of the combination chemotherapy treatment Vyxeos® (daunorubicin and cytarabine) for the treatment of high-risk acute myeloid leukemia in adults. He then discusses the ongoing and future clinical trials in this disease area and then turns to more personalized treatments.


2018 ◽  
Vol 19 (4) ◽  
pp. 299-309
Author(s):  
Alex José de Melo Silva

Abstract The family of Bcl-2 proteins is one of the most responsible for apoptosis pathway, that is a critical process to the maintenance of tissue homeostasis. Bcl-2 is an essential apoptotic regulator belonging to a family of functionally and structurally related proteins known as the Bcl-2 family. Some members of this family act as anti-apoptotic regulators, whereas others act in pro-apoptotic function. The relationship between the pro and anti-apoptotic proteins can regulate whether cells begin the apoptosis or remain its life cycle. Increasing of Bcl-2 expression has been found in some hematologic diseases, such as Acute Myeloid Leukemia (AML) and their effects on responsiveness to anticancer therapy have been recently described. Thus, this review aims to discuss apoptosis and the role of the Bcl-2 family of proteins in chemoresistance when overexpressed in patients committed with Acute Myeloid Leukemia submitted to chemotherapy treatment.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 879-879
Author(s):  
Xiangguo Shi ◽  
Daisuke Nakada ◽  
Ayumi Kitano ◽  
Rebecca Murdaugh ◽  
Yu-Jung Tseng ◽  
...  

Acute myeloid leukemia (AML) is primarily a disease of older adults with poor treatment outcomes. Despite years of intensive research, the standard induction therapy for AML has remained largely unchanged for decades. Thus, the development of new and efficacious therapeutic targets for AML is urgently needed. Leukemia cells exhibit multiple metabolic aberrations that may be therapeutically targeted. Here, we show that nicotinamide adenine dinucleotide (NAD+) promotes leukemogenesis and causes chemotherapy treatment resistance through fueling energetic metabolism, and pinpoints nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) is a novel therapeutic target for AML. To identify novel genes essential for AML, we performed a whole genome CRISPR dropout screen by using MOLM13 cell line and identified 1,951 essential genes (Fig. A). By searching druggable targets among these genes, we narrowed down to 345 genes, among which we found two genes, NMNAT1 (nicotinamide nucleotide adenylyltransferase 1) and NAMPT (nicotinamide phosphoribosyltransferase), both involved in key steps in NAD+ biosynthesis. We comprehensively analyzed dependency scores for all genes involved in the NAD+ biosynthetic pathways (de novo synthesis pathway, the Preiss-Handler pathway and the salvage pathway) across a broad panel of cancer cell lines from the Dependency Map database (https://depmap.org/portal/). The results showed that NMNAT1 and NAMPT are both strongly selective and uniquely required for hematological malignancies compared to other cancers (Fig. B). Since little success has been achieved for NAMPT inhibitors in clinical trials, our attention was drawn to NMNAT1, which encodes a nuclear localized enzyme catalyzing the final step in NAD+ biosynthesis. We confirmed that deletion of NMNAT1 in AML cells significantly reduced nuclear NAD+ level and cell viability over time while sparing normal hematopoietic progenitor cells, suggesting that NMNAT1 is targetable to AML. Overexpression of wild-type Nmnat1 but not the enzymatically inactive forms rescued NMNAT1-KO AML, indicating that the catalytic activity of NMNAT1 is required for AML. To study the role of NAD+ in AML, we first measured NAD+ levels in leukemic and normal cells, and found higher NAD+ levels in leukemia-initiating cells from a murine MLL-AF9-induced AML model compared to normal cells. Supplementation of NAD+ metabolites (NMN, NAM and NR) increased AML proliferation, enhanced glycolysis (lactate production) and oxidative phosphorylation (ATP production), resulting in chemotherapy resistance (Fig. C). Deletion of NMNAT1 sensitized AML cell to chemotherapy treatment. To study the role of NMNAT1 in leukemogenesis in vivo, we genetically deleted NMNAT1 in murine or human leukemia cells, transplanted them into recipient mice, and found that deletion of NMNAT1 reduced leukemic burden and extended leukemia-free survival (Fig. D). Finally, to reveal the molecular mechanisms underlying NMNAT1 KO-mediated cell death (increased levels of gamma-H2AX), RNA-seq and functional assay of NAD+ dependent enzymes were performed. We found that the reduction of nuclear NAD+ resulting from NMNAT1 deletion upregulated genes involved in DNA repair pathway, which may be linked to impaired PARPs and Sirtuins activity. Our findings reveal the important function of NAD+ in leukemogenesis and chemoresistance, and identify NMANT1 as a novel therapeutic target for AML. Figure Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 20 (1) ◽  
pp. 219 ◽  
Author(s):  
Yulia Michailov ◽  
Eitan Lunenfeld ◽  
Joseph Kapilushnik ◽  
Shevach Friedler ◽  
Eckart Meese ◽  
...  

Leukemia is one of the most common cancers in patients of reproductive age. It is well known that chemotherapy, used as anti-cancer therapy, adversely affects male fertility. Moreover, the negative effect of leukemia on sperm quality, even before chemotherapy treatment, has been reported. However, the mechanisms behind this disease’s effect on sperm quality remains unknown. In this study, we examine the direct effect of leukemia and chemotherapy alone and in combination on sperm parameters and male fertility. For this, we developed an acute myeloid leukemia (AML) mouse model (mice were treated with AML cells C1498 and developed leukemia); these mice then received cytarabine chemotherapy. Our findings reveal a significant reduction in sperm concentration and motility and a significant increase in abnormal morphology and spontaneous acrosome reaction of the sperm following AML and chemotherapy treatment, alone and in combination. We also found a reduction in male fertility and the number of delivered offspring. Our results support previous findings that AML impairs sperm parameters and show for the first time that AML increases spontaneous acrosome reaction and decreases male fertility capacity and number of offspring.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2178-2178
Author(s):  
Robin L. Perry ◽  
Jean-Emmanuel Sarry ◽  
Gwenn-ael Danet-Desnoyers ◽  
Martin Carroll

Abstract Abstract 2178 The major therapeutic barrier in acute myeloid leukemia is chemotherapy resistance. Although most patients will respond to treatment with chemotherapy over 50% of responders will relapse and eventually die of disease. Many hypotheses have been proposed to explain chemotherapy resistance but none of these have lead to new therapies or a complete understanding of the molecular mechanisms of AML chemotherapy resistance. In order to develop an improved understanding of chemotherapy resistance in AML, we have developed a chemotherapy model of human AML in NSG mice. Mice are engrafted with primary AML samples from patients seen at diagnosis or relapse of disease or who demonstrated primary chemotherapy resistance. After demonstrating AML engraftment, mice are treated with cytosine arabinoside (Ara-C) given IP daily for 5 days as a single agent at 10mg/kg daily, 30mg/kg daily, and 60 mg/kg daily which correlates with human dosing. In >75% of mice treated with 10 mg/kg of Ara-C there is a cytoreductive effect at 2 weeks post-treatment with relapse at 4 weeks post-treatment. In all mice treated with 30 mg/kg of Ara-C there was a cytoreductive effect at 2 weeks post-treatment with relapse at 4 weeks post-treatment. In all mice treated with 60 mg/kg of Ara-C there was a cytoreductive effect at 2 weeks post-treatment with relapse 4–13 weeks post-treatment when relapse occurred, demonstrating that there is a dose response relationship in the model in terms of nadir leukemic burden and time to peripheral blood relapse. Two weeks after treatment, there was up to a 50-fold decrease in total AML cell burden in the peripheral blood of mice treated with 30 mg/kg and and up to a 70-fold decrease in the peripheral blood of mice treated with 60 mg/kg of Ara-C. We found no enhancement in quiescent or G0 cells after chemotherapy treatment. We did however, in 1/5 samples tested see a change in phenotype after chemotherapy treatment with an increase in the total number of CD34+38+ cells with a concomitant decrease in CD34-38+ cells. In all other samples tested, there was no change in phenotype after chemotherapy treatment. These results are in contrast to recent studies using a 1 gm/kg dose of Ara-C and analysis at Day 3 following a single treatment. These results have implications for understanding the physiologic response to Ara-C at different doses. We are currently analyzing mRNA expression arrays of AML without or with Ara-C treatment to identify novel mechanisms of chemotherapy resistance. Taken together, this model provides a novel approach for development of new therapies in AML. Disclosures: Carroll: Sanofi Aventis Corporation: Research Funding; Kyowa Hakko Kirin Pharmaceuticals: Research Funding; Agios Pharmaceuticals: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document