Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (4)

Search Parameters:
Keywords = revumenib

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 2205 KiB  
Article
An Ultra-Fast Validated Green UPLC-MS/MS Approach for Assessing Revumenib in Human Liver Microsomes: In Vitro Absorption, Distribution, Metabolism, and Excretion and Metabolic Stability Evaluation
by Mohamed W. Attwa, Ali S. Abdelhameed and Adnan A. Kadi
Medicina 2024, 60(12), 1914; https://doi.org/10.3390/medicina60121914 - 21 Nov 2024
Abstract
Background and Objectives: Revumenib (SNDX-5613) is a powerful and specific inhibitor of the menin–KMT2A binding interaction. It is a small molecule that is currently being researched to treat KMT2A-rearranged (KMT2Ar) acute leukemias. Revumenib (RVB) has received Orphan Drug Designation from the US FDA [...] Read more.
Background and Objectives: Revumenib (SNDX-5613) is a powerful and specific inhibitor of the menin–KMT2A binding interaction. It is a small molecule that is currently being researched to treat KMT2A-rearranged (KMT2Ar) acute leukemias. Revumenib (RVB) has received Orphan Drug Designation from the US FDA for treating patients with AML. It has also been granted Fast Track designation by the FDA for treating pediatric and adult patients with R/R acute leukemias that have a KMT2Ar or NPM1 mutation. Materials and Methods: The target of this research was to create a fast, precise, green, and extremely sensitive UPLC-MS/MS technique for the estimation of the RVB level in human liver microsomes (HLMs), employing an ESI source. The validation procedures were carried out in accordance with the bioanalytical technique validation requirements established by the US Food and Drug Administration that involve linearity, selectivity, precision, accuracy, stability, matrix effect, and extraction recovery. The outcome data of the validation features of the UPLC-MS/MS approach were acceptable according to FDA guidelines. RVB parent ions were formed in the positive ESI source and its two fragment ions were estimated employing multiple reaction monitoring (MRM) mode. The separation of RVB and encorafenib was achieved using a C8 column (2.1 mm, 50 mm, and 3.5 µm) and isocratic mobile phase. Results: The RVB calibration curve linearity ranged from 1 to 3000 ng/mL (y = 0.6515x − 0.5459 and R2 = 0.9945). The inter-day precision and accuracy spanned from −0.23% to 11.33%, while the intra-day precision and accuracy spanned from −0.88% to 11.67%, verifying the reproducibility of the UPLC-MS/MS analytical technique. The sensitivity of the developed methodology demonstrated its capability to quantify RVB levels at an LOQ of 0.96 ng/mL. The AGREE score was 0.77, confirming the greenness of the current method. The low in vitro t1/2 (14.93 min) and high intrinsic clearance (54.31 mL/min/kg) of RVB revealed that RVB shares similarities with medications that have a high extraction ratio. Conclusions: The present LC-MS/MS approach is considered the first analytical approach with the application of metabolic stability assessment for RVB estimation in HLMs. These methods are essential for advancing the development of new pharmaceuticals, particularly in enhancing metabolic stability. Full article
(This article belongs to the Special Issue Acute Myeloid Leukemia: Update on Diagnosis, Therapy, and Monitoring)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of the target analyte, revumenib, and the encorafenib that was used as an internal standard in the UPLC-MS/MS analysis of RVB.</p>
Full article ">Figure 2
<p>The RVB ADME radar chart produced from the in silico SwissADME program.</p>
Full article ">Figure 3
<p>MRM spectrum showing PI mass scan of RVB as protonated molecular ion [M + H]<sup>+</sup> (<b>A</b>) and MRM spectrum showing PI mass spectrum scan of ENF [M + H]<sup>+</sup> (<b>B</b>). The probable dissociations behaviours are elucidated.</p>
Full article ">Figure 4
<p>The MRM chromatogram of the first control sample (negative control HLMs) demonstrated the lack of any interference in the retention times of RVB and ENF (<b>A</b>). The MRM chromatogram of the second control sample, positive control (Blank HLMs combined with ENF at 1000 ng/mL) (<b>B</b>). The superimposed MRM chromatograms of the 9 RVB CSs, as well as the 3 QCs (<b>C</b>). The MRM chromatograms revealed analytical peaks conforming to RVB (at 0.34 min) and ENF at 1000 ng/mL and a retention time of 0.66 min).</p>
Full article ">Figure 5
<p>RVB LLOQ chromatographic peak (1 ng/mL) (<b>A</b>). The ENF (1000 ng/mL) peak that was used as IS (<b>B</b>).</p>
Full article ">Figure 6
<p>The AGREE programme was employed to demonstrate the greenness scale profile of the established UPLC-MS/MS approach, shown in the form of a circular diagram of twelve separate characteristics.</p>
Full article ">Figure 7
<p>(<b>A</b>) RVB metabolic stability curve representing percentage of RVB residual concentration against time intervals; (<b>B</b>) linear segment of the metabolic stability curve representing the LN of the percentage of RVB residual level against time intervals, showing the regression equation of the linear part.</p>
Full article ">
14 pages, 4700 KiB  
Article
Distinct Responses to Menin Inhibition and Synergy with DOT1L Inhibition in KMT2A-Rearranged Acute Lymphoblastic and Myeloid Leukemia
by Fabienne R. S. Adriaanse, Pauline Schneider, Susan T. C. J. M. Arentsen-Peters, Ana M. Neves da Fonseca, Janine Stutterheim, Rob Pieters, C. Michel Zwaan and Ronald W. Stam
Int. J. Mol. Sci. 2024, 25(11), 6020; https://doi.org/10.3390/ijms25116020 - 30 May 2024
Cited by 2 | Viewed by 1283
Abstract
Pediatric acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) exhibit favorable survival rates. However, for AML and ALL patients carrying KMT2A gene translocations clinical outcome remains unsatisfactory. Key players in KMT2A-fusion-driven leukemogenesis include menin and DOT1L. Recently, menin inhibitors like revumenib have [...] Read more.
Pediatric acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) exhibit favorable survival rates. However, for AML and ALL patients carrying KMT2A gene translocations clinical outcome remains unsatisfactory. Key players in KMT2A-fusion-driven leukemogenesis include menin and DOT1L. Recently, menin inhibitors like revumenib have garnered attention for their potential therapeutic efficacy in treating KMT2A-rearranged acute leukemias. However, resistance to menin inhibition poses challenges, and identifying which patients would benefit from revumenib treatment is crucial. Here, we investigated the in vitro response to revumenib in KMT2A-rearranged ALL and AML. While ALL samples show rapid, dose-dependent induction of leukemic cell death, AML responses are much slower and promote myeloid differentiation. Furthermore, we reveal that acquired resistance to revumenib in KMT2A-rearranged ALL cells can occur either through the acquisition of MEN1 mutations or independently of mutations in MEN1. Finally, we demonstrate significant synergy between revumenib and the DOT1L inhibitor pinometostat in KMT2A-rearranged ALL, suggesting that such drug combinations represent a potent therapeutic strategy for these patients. Collectively, our findings underscore the complexity of resistance mechanisms and advocate for precise patient stratification to optimize the use of menin inhibitors in KMT2A-rearranged acute leukemia. Full article
(This article belongs to the Special Issue New Agents and Novel Drugs Use for the Oncological Diseases Treatment)
Show Figures

Figure 1

Figure 1
<p>Responses to revumenib in <span class="html-italic">KMT2A</span>-rearranged AML and ALL cells. (<b>A</b>) Cell viability in response to increasing concentrations of revumenib as assessed by 4-day MTT assays in <span class="html-italic">KMT2A</span>-rearranged (n = 6) and wildtype <span class="html-italic">KMT2A</span> (n = 3) AML cell line models. The dashed line shows the 50% viability threshold. Experiments were performed in technical triplicates and data consisted of three biological replicates. (<b>B</b>) IC<sub>50</sub>-values (i.e., the inhibitory concentration to 50% of the leukemic cells) for revumenib as determined by nonlinear regression in <span class="html-italic">KMT2A</span>-rearranged and wildtype <span class="html-italic">KMT2A</span> AML cell lines, statistically evaluated by an unpaired two-tailed <span class="html-italic">t</span>-test, with ns showing no significant differences. (<b>C</b>) Cell viability in response to increasing concentrations of revumenib using 4-day MTT assays in ex vivo pediatric <span class="html-italic">KMT2A</span>-rearranged AML patient samples obtained from patient-derived xenograft mouse models (n = 3). The dashed line shows the 50% viability threshold. Experiments were performed in technical triplicates. (<b>D</b>) Cell viability in response to increasing concentrations of revumenib as assessed by 4-day MTT assays in <span class="html-italic">KMT2A</span>-rearranged ALL cell line models (n = 5) and wildtype <span class="html-italic">KMT2A</span> ALL cell lines (n = 2). The dashed line shows the 50% viability threshold. Experiments were performed in technical triplicates and data consisted of three biological replicates. (<b>E</b>) IC<sub>50</sub>-values for revumenib as determined by nonlinear regression in <span class="html-italic">KMT2A</span>-rearranged and wildtype <span class="html-italic">KMT2A</span> ALL cell lines, statistically evaluated by an unpaired two-tailed <span class="html-italic">t</span>-test; * <span class="html-italic">p</span> &lt; 0.05 (<b>F</b>) Cell viability in response to increasing concentrations of revumenib using 4-day MTT assays in ex vivo pediatric <span class="html-italic">KMT2A</span>-rearranged ALL patient samples (n = 5). The dashed line shows the 50% viability threshold Experiments were performed in technical triplicates.</p>
Full article ">Figure 2
<p>Prolonged revumenib exposure induces myeloid differentiation in <span class="html-italic">KMT2A</span>-rearranged AML cells. (<b>A</b>) Percentage of viable cells after 7 and 14-day exposures to indicated concentrations of revumenib as determined by trypan blue exclusion in <span class="html-italic">KMT2A</span>-rearranged AML cell line models (n = 5). Responsive cell lines are in blue and unresponsive cell lines are in purple. The dashed line shows the 50% viability threshold. Experiments were performed in technical duplicates and data consisted of two biological replicates. (<b>B</b>) Percentages of live (grey), apoptotic (orange), and dead (red) cells after 7 and 14-day exposures to indicated concentrations of revumenib as determined by an Annexin V/7AAD staining and flowcytometry in <span class="html-italic">KMT2A</span>-rearranged AML cell line models. Differences in live, apoptotic, and death cells induced by revumenib as compared to untreated controls were statistically verified by two-way ANOVA Tukey’s multiple comparisons tests. Data consisted of two biological replicates. (<b>C</b>) Flow cytometric assessment of the expression of the myeloid differentiation marker CD14 and of stem cell marker CD117 (c-Kit) after 7 and 14 days of revumenib exposure in the responsive <span class="html-italic">KMT2A</span>-rearranged AML cell lines SHI-1, MONO-MAC-1, and NOMO-1, from duplicate experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.00005 and ns for no significant <span class="html-italic">p</span>.</p>
Full article ">Figure 3
<p>Revumenib readily induces apoptosis in <span class="html-italic">KMT2A</span>-rearranged ALL cells. (<b>A</b>) Percentage of viable cells after 4-day exposures to indicated concentrations of revumenib as determined by trypan blue exclusion in <span class="html-italic">KMT2A</span>-rearranged ALL cell line models (n = 5; in orange) and the highly sensitive <span class="html-italic">KMT2A</span>-rearranged AML cell line MV4-11 (in blue). The dashed line shows the 50% viability threshold. Experiments were performed in technical duplicates and data consisted of two biological replicates. (<b>B</b>) Percentages of live (grey), apoptotic (orange), and dead (red) cells after 4-day exposures to indicated concentrations of revumenib as determined by flow cytometry and Annexin V/7AAD staining in the <span class="html-italic">KMT2A</span>-rearranged AML cell line MV4-11 and (<b>C</b>) in the <span class="html-italic">KMT2A</span>-rearranged ALL cell lines. Differences in live, apoptotic, and death cells induced by revumenib as compared to untreated controls were statistically verified by two-way ANOVA Tukey’s multiple comparisons tests. Data consisted of two biological replicates. (<b>D</b>) Cell cycle analysis showing the percentages of cells residing in the G1-phase, S-phase, and GM2-phase as determined by Hoechst 33342/7AAD staining and flow cytometry after 4-day exposures to indicated concentrations of revumenib. Differences in cell cycle phases induced by revumenib as compared to untreated controls were statistically verified by two-way ANOVA Tukey’s multiple comparisons tests. Data consisted of two biological replicates. (<b>E</b>) Cell viability (as determined by trypan blue exclusion) and cell cycle analysis (as determined by Hoechst 33342/7AAD staining and flow cytometry) after 4-day exposures to indicated concentrations of revumenib in a representative <span class="html-italic">KMT2A</span>-rearranged infant ALL patient sample obtained from a patient-derived xenograft mouse model. The dashed line shows the 50% viability threshold. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, **** <span class="html-italic">p</span> &lt; 0.00005 and ns for no significant <span class="html-italic">p</span>.</p>
Full article ">Figure 4
<p>Induction of acquired resistance to revumenib in <span class="html-italic">KMT2A</span>-rearranged ALL cells. Induction of acquired resistance to revumenib was accomplished by exposing the <span class="html-italic">KMT2A::AFF1</span><sup>+</sup> ALL cell lines SEM and RS4;11 to increasing concentrations of revumenib of up to 10 µM for 10 weeks. Revumenib-resistant daughter cell lines are indicated as SEM<sup>REV_RES#1–4</sup> and RS4;11<sup>REV_RES#1–4</sup>, respectively. (<b>A</b>) Cell viability in response to increasing concentrations of revumenib as assessed by 4-day MTT assays in SEM (orange) and revumenib-resistant daughter cell lines SEM<sup>REV_RES#1–4</sup>(purple). The dashed line shows the 50% viability threshold. Experiments were performed in technical triplicates and data consisted of three biological replicates. (<b>B</b>) IC<sub>50</sub>-values for revumenib as determined by nonlinear regression in SEM (orange) and revumenib-resistant daughter cell lines SEM<sup>REV_RES#1–4</sup> (purple) evaluated by an unpaired two-tailed <span class="html-italic">t</span>-test. (<b>C</b>) <span class="html-italic">MEN1</span> mutation analysis (i.e., Sanger sequencing results) showing an M322T <span class="html-italic">MEN1</span> mutation in all revumenib-resistant SEM daughter lines SEM<sup>REV_RES#1–4</sup> (blue). (<b>D</b>) Cell viability in response to increasing concentrations of revumenib as assessed by 4-day MTT assays in RS4;11 (orange) and revumenib-resistant daughter cell lines RS4;11<sup>REV_RES#1–4</sup> (purple). The dashed line shows the 50% viability threshold. Experiments were performed in technical triplicates and data consisted of three biological replicates. (<b>E</b>) IC<sub>50</sub>-values for revumenib as determined by nonlinear regression in RS4;11 (orange) and revumenib-resistant daughter cell lines RS4;11<sup>REV_RES#1–4</sup> (purple) evaluated by an unpaired two-tailed <span class="html-italic">t</span>-test. (<b>F</b>) <span class="html-italic">MEN1</span> mutation analysis reveals an M322T <span class="html-italic">MEN1</span> mutation only in revumenib-resistant RS4;11<sup>REV_RES#2</sup> cells (blue).</p>
Full article ">Figure 5
<p>Synergy between revumenib and pinometostat in <span class="html-italic">KMT2A</span>-rearranged ALL. Three-dimensional synergy plots showing drug synergy (red) or antagonism (green) between indicated concentrations of revumenib (x-axis) and pinometostat (y-axis) as determined by 6-day pinometostat pre-treated cells followed by 4-day MTT assays of revumenib exposures in (<b>A</b>) <span class="html-italic">KMT2A</span>-rearranged ALL cell lines, (<b>B</b>) a PDX derived <span class="html-italic">KMT2A</span>-rearranged infant ALL patient sample, and (<b>C</b>) <span class="html-italic">KMT2A</span>-rearranged AML cell lines. Drug synergy/antagonism is expressed as Zero Interaction Potency (ZIP) scores (z-axis), with scores of &gt;5 being considered as synergistic effects (red areas), and ZIP scores below −5 are deemed as antagonistic effects (green areas). On top of each 3D synergy plot, the average ZIP score over the entire range of pinometostat and revumenib concentrations is listed.</p>
Full article ">
11 pages, 1460 KiB  
Review
A 2024 Update on Menin Inhibitors. A New Class of Target Agents against KMT2A-Rearranged and NPM1-Mutated Acute Myeloid Leukemia
by Anna Candoni and Gabriele Coppola
Hematol. Rep. 2024, 16(2), 244-254; https://doi.org/10.3390/hematolrep16020024 - 18 Apr 2024
Cited by 7 | Viewed by 3741
Abstract
Menin inhibitors are new and promising agents currently in clinical development that target the HOX/MEIS1 transcriptional program which is critical for leukemogenesis in histone-lysine N-methyltransferase 2A-rearranged (KMT2Ar) and in NPM1-mutated (NPM1mut) acute leukemias. The mechanism of action of this new class of agents [...] Read more.
Menin inhibitors are new and promising agents currently in clinical development that target the HOX/MEIS1 transcriptional program which is critical for leukemogenesis in histone-lysine N-methyltransferase 2A-rearranged (KMT2Ar) and in NPM1-mutated (NPM1mut) acute leukemias. The mechanism of action of this new class of agents is based on the disruption of the menin–KMT2A complex (consisting of chromatin remodeling proteins), leading to the differentiation and apoptosis of AML cells expressing KMT2A or with mutated NPM1. To date, this new class of drugs has been tested in phase I and II clinical trials, both alone and in combination with synergistic drugs showing promising results in terms of response rates and safety in heavily pre-treated acute leukemia patients. In this brief review, we summarize the key findings on menin inhibitors, focusing on the mechanism of action and preliminary clinical data on the treatment of acute myeloid leukemia with this promising new class of agents, particularly revumenib and ziftomenib. Full article
Show Figures

Figure 1

Figure 1
<p>Cofactor menin is necessary for KMT2A to bind HOX gene promoters. In detail, KMT2A binding menin acts as a transcriptional upregulator of HOX genes and their cofactor MEIS1 (<b>A</b>). Disruption of this chromatin complex (by binding of the menin inhibitors and menin protein) leads to inhibition of the leukemogenic transcription program (downregulation of HOX genes) without affecting normal hematopoiesis (<b>B</b>).</p>
Full article ">
16 pages, 3100 KiB  
Article
Synergistic Effects of the RARalpha Agonist Tamibarotene and the Menin Inhibitor Revumenib in Acute Myeloid Leukemia Cells with KMT2A Rearrangement or NPM1 Mutation
by Maximilian Fleischmann, Julia Bechwar, Diana Voigtländer, Mike Fischer, Ulf Schnetzke, Andreas Hochhaus and Sebastian Scholl
Cancers 2024, 16(7), 1311; https://doi.org/10.3390/cancers16071311 - 28 Mar 2024
Cited by 2 | Viewed by 1756
Abstract
Inhibition of menin in acute myeloid leukemia (AML) harboring histone-lysine-N-methyltransferase 2A rearrangement (KMT2Ar) or the mutated Nucleophosmin gene (NPM1c) is considered a novel and effective treatment approach in these patients. However, rapid acquisition of resistance mutations can impair treatment success. In [...] Read more.
Inhibition of menin in acute myeloid leukemia (AML) harboring histone-lysine-N-methyltransferase 2A rearrangement (KMT2Ar) or the mutated Nucleophosmin gene (NPM1c) is considered a novel and effective treatment approach in these patients. However, rapid acquisition of resistance mutations can impair treatment success. In patients with elevated retinoic acid receptor alpha (RARA) expression levels, promising effects are demonstrated by the next-generation RARalpha agonist tamibarotene, which restores differentiation or induces apoptosis. In this study, the combination of revumenib and tamibarotene was investigated in various KMT2Ar or NPM1c AML cell lines and patient-derived blasts, focusing on the potential synergistic induction of differentiation or apoptosis. Both effects were analyzed by flow cytometry and validated by Western blot analysis. Synergy calculations were performed using viability assays. Regulation of the relevant key mediators for the MLL complex were quantified by RT-qPCR. In MV4:11 cells characterized by the highest relative mRNA levels of RARA, highly synergistic induction of apoptosis is demonstrated upon combination treatment. Induction of apoptosis by combined treatment of MV4:11 cells is accompanied by pronounced induction of the pro-apoptotic protein BAX and a synergistic reduction in CDK6 mRNA levels. In MOLM13 and OCI-AML3 cells, an increase in differentiation markers like PU.1 or a decreased ratio of phosphorylated to total CEBPA is demonstrated. In parts, corresponding effects were observed in patient-derived AML cells carrying either KMT2Ar or NPM1c. The impact of revumenib on KMT2Ar or NPM1c AML cells was significantly enhanced when combined with tamibarotene, demonstrating synergistic differentiation or apoptosis initiation. These findings propose promising strategies for relapsed/refractory AML patients with defined molecular characteristics. Full article
(This article belongs to the Special Issue Novel Combination Therapies for Acute Leukemia)
Show Figures

Figure 1

Figure 1
<p>Determination of differentiation and apoptosis induction following single or combination treatment with revumenib and tamibarotene in different AML cell lines. (<b>A</b>–<b>D</b>) Flow cytometry analysis of differentiation (<b>A</b>,<b>B</b>) and apoptosis (<b>C</b>,<b>D</b>) in MV4:11, MOLM13, OCI-AML3, and HL-60 cells after 72 h incubation with DMSO (0.02% <span class="html-italic">v</span>/<span class="html-italic">v</span>), 50 nM revumenib, 100 nM tamibarotene, and their combination. The anti-CD11b ratio is calculated relative to the respective isotype control. Graphs represent the mean ± SD from five independent experiments with technical duplicates. Statistical significance was assessed using a two-sided ANOVA (ns, not significant).</p>
Full article ">Figure 2
<p>Time-dependent induction of differentiation and apoptosis following single or combination treatment with revumenib and tamibarotene in different AML cell lines (<b>A</b>–<b>D</b>). Flow cytometry analysis of differentiation (upper panel) and apoptosis (lower panel) following 24 h, 48 h, 72 h, and 96 h incubation of MV4:11, MOLM13, OCI-AML3, and HL-60 cells with 0.2% <span class="html-italic">v</span>/<span class="html-italic">v</span> DMSO, 50 nM revumenib, 100 nM tamibarotene, and their combination. CD11b expression for MV4:11 is displayed up to 72 h due to elevated apoptosis rates at 96 h. The anti-CD11b ratio is normalized to the respective isotype control. Graphs depict the mean ± SD from three independent experiments with technical duplicates. Statistical significance was determined using a two-sided ANOVA test (ns, not significant).</p>
Full article ">Figure 3
<p>Impact of tamibarotene and revumenib on cellular morphology and surface expression of CD14, CD38, and CD135 (<b>A</b>,<b>B</b>). MOLM13 and OCI-AML3 cells were treated with specified concentrations of revumenib, tamibarotene, their combination, and 0.2% <span class="html-italic">v</span>/<span class="html-italic">v</span> DMSO for 4 days. Subsequently, cells were cytospun onto glass slides and stained with hematoxylin and eosin. Representative images were captured with a 40X objective and a CCD camera. (<b>C</b>,<b>D</b>) Flow cytometry analysis of CD14 expression after 24 h and 72 h incubation of MV4:11, MOLM13, and OCI-AML3 cells with specified concentrations of revumenib, tamibarotene, their combination, and 0.2% <span class="html-italic">v</span>/<span class="html-italic">v</span> DMSO, normalized to the respective isotype control. (<b>E</b>,<b>F</b>) Flow cytometry analysis of anti-CD135 and anti-CD38 staining after 24 h incubation for indicated conditions. Antibody ratios are normalized to the respective isotype control. Graphs represent the mean ± SD from three independent experiments with technical duplicates. Statistical significance was determined using a two-sided ANOVA test (ns, not significant).</p>
Full article ">Figure 4
<p>Western blot analysis of differentiation and apoptosis proteins in response to revumenib and tamibarotene treatments. Protein lysates from MV4:11 (<b>A</b>), MOLM13 (<b>B</b>,<b>C</b>), OCI-AML3 (<b>D</b>), and HL-60 (<b>E</b>) cells—treated for 24 h or 72 h with DMSO 0.2% <span class="html-italic">v</span>/<span class="html-italic">v</span>, 50 nM revumenib, 100 nM tamibarotene and their combination—underwent immunoblotting to assess the expression and phosphorylation status of specified proteins. GAPDH levels were detected as a loading control. Corresponding densitometry analysis was conducted, and fold changes normalized to the loading control and DMSO control are presented within the blots. Representative blots from three independent experiments are displayed. Next to the blots, the quantification of densitometry analysis is summarized from all replicates (at least two representative blots per protein) and indicated as log<sub>2</sub>fold change. The uncropped blots are shown in <a href="#app1-cancers-16-01311" class="html-app">File S1</a>. Except for (<b>F</b>), where the p-CEBPA-to-CEBPA ratio is illustrated, all proteins are normalized to the DMSO control, which is set to 1 and therefore not shown. Statistical significance was determined using a two-sided ANOVA (*** <span class="html-italic">p</span> &lt; 0.001; ** <span class="html-italic">p</span> &lt; 0.01; * <span class="html-italic">p</span> &lt; 0.05; ns, not significant).</p>
Full article ">Figure 5
<p>Synergistic reduction in metabolic activity with the combination of revumenib and tamibarotene (<b>A</b>–<b>D</b>). Indicated cell lines were incubated for 48 h with concentration series of revumenib (0, 10, 50, 100 nM) and tamibarotene (0, 50, 100, 300, 500 nM). Metabolic activity was assessed by PrestoBlue viability assay in a 96-well plate, and results were normalized to the DMSO control. Graphs represent the mean ± SD from two independent experiments in technical triplicates. Statistical significance was determined using a two-sided ANOVA (ns, not significant). (<b>E</b>–<b>H</b>) Cell lines were incubated for 48 h with a revumenib concentration series (0, 50, 100, 300, and 500 nM), either in combination with DMSO (0.01% <span class="html-italic">v</span>/<span class="html-italic">v</span>) or 50 nM tamibarotene. Metabolic activity was measured by PrestoBlue viability assay and graphs depict the mean ± SD from three independent experiments in technical triplicates. Data were analyzed using SynergyFinder software (Version 3) to calculate ZIP synergy scores (0–10 additive effects, &gt;10 synergistic effects). Corresponding heat maps are illustrated on the right side.</p>
Full article ">Figure 6
<p>Effects of revumenib and tamibarotene on primary AML cells. PBMCs were thawed and rested in IMDM medium for 24 h before incubating with 50 nM revumenib, 100 nM tamibarotene, and DMSO (0.02% <span class="html-italic">v</span>/<span class="html-italic">v</span>) for 72 h. Subsequently, protein lysates underwent immunoblotting for specified proteins, quantified by densitometry, and normalized to the loading and DMSO control. Flow cytometry measured Annexin V, CD11b, CD34, and CD38 surface expression (all presented as ratios to isotype control from geometric means) after 72 h of incubation. The uncropped blots are shown in <a href="#app1-cancers-16-01311" class="html-app">File S1</a>.</p>
Full article ">
Back to TopTop