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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,641)

Search Parameters:
Keywords = acute myeloid leukemia

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 793 KiB  
Systematic Review
Nurse-Led Interventions for Improving Medication Adherence in Chronic Diseases: A Systematic Review
by Daniela Berardinelli, Alessio Conti, Anis Hasnaoui, Elena Casabona, Barbara Martin, Sara Campagna and Valerio Dimonte
Healthcare 2024, 12(23), 2337; https://doi.org/10.3390/healthcare12232337 - 22 Nov 2024
Abstract
Background: Poor medication adherence results in negative health outcomes and increased healthcare costs. Several healthcare professionals provide interventions to improve medication adherence, with the effectiveness of nurse-led interventions in people with chronic diseases remaining unclear. Objective: This study sought to evaluate [...] Read more.
Background: Poor medication adherence results in negative health outcomes and increased healthcare costs. Several healthcare professionals provide interventions to improve medication adherence, with the effectiveness of nurse-led interventions in people with chronic diseases remaining unclear. Objective: This study sought to evaluate the effectiveness of nurse-led interventions for improving medication adherence in adults with chronic conditions. Methods: Five databases (MEDLINE, CINAHL, EMBASE, Cochrane Library, SCOPUS) were searched without applying a temporal limit. Studies evaluating the effects of nurse-led interventions on medication adherence in adults with one or multiple chronic conditions were included. Interventions only targeting a single acute disease were excluded. Results: A total of twenty-two studies with 5975 participants were included. Statistically significant improvements in adherence were reported in five out of seven studies involving face-to-face visits to patients with heart failure (n = 2), chronic myeloid leukemia (n = 1), hypertension (n = 1) and multimorbidity (n = 1) and in four out of nine studies adopting a mixed method involving face-to-face visits and telephone follow-up for patients with heart failure (n = 1), hypertension (n = 1), coronary disease (n = 1) and multimorbidity (n = 1). Remote interventions were effective in improving medication adherence in one out of six studies. No statistically significant differences were found between tablet computer-based patient education and nurse-led educational sessions. The motivational approach was found to be one of the most common strategies used to promote patient medication adherence. Conclusions: Nurse-led face-to-face visits may be effective in improving medication adherence in people with chronic diseases. However, further research is needed because current methods for measuring medication adherence may not accurately capture patient behaviour and medication consumption patterns. Full article
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the study selection.</p>
Full article ">Figure 2
<p>Bias assessment of the selected studies. Symbols and colours represent the risk-of-bias assessment for individual studies: “+” (green) indicates low risk of bias, “!” (yellow) indicates unclear risk of bias, and “-“ (red) indicates high risk of bias.</p>
Full article ">
15 pages, 1715 KiB  
Review
Senescent T Cells: The Silent Culprit in Acute Myeloid Leukemia Progression?
by Xiaolan Zhang and Lingbo Liu
Int. J. Mol. Sci. 2024, 25(23), 12550; https://doi.org/10.3390/ijms252312550 - 22 Nov 2024
Abstract
Malignant tumors can evade immune surveillance and elimination through multiple mechanisms, with the induction of immune cell dysfunction serving as a crucial strategy. Mounting evidence indicates that T cell senescence constitutes the primary mechanism underlying T cell dysfunction in acute myeloid leukemia (AML) [...] Read more.
Malignant tumors can evade immune surveillance and elimination through multiple mechanisms, with the induction of immune cell dysfunction serving as a crucial strategy. Mounting evidence indicates that T cell senescence constitutes the primary mechanism underlying T cell dysfunction in acute myeloid leukemia (AML) and represents one of the potential causes of immunotherapy failure. AML usually progresses rapidly and is highly susceptible to drug resistance, thereby resulting in recurrence and patient mortality. Hence, disrupting the immune interface within the bone marrow microenvironment of AML has emerged as a critical objective for synergistically enhancing tumor immunotherapy. In this review, we summarize the general characteristics, distinctive phenotypes, and regulatory signaling networks of senescent T cells and highlight their potential clinical significance in the bone marrow microenvironment of AML. Additionally, we discuss potential therapeutic strategies for alleviating and reversing T cell senescence. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>T cell senescence. T cell senescence can manifest as the senescence of individual cells and dysregulation of the cell population. In addition to the common characteristics of general cell senescence, senescent T cells can be characterized by the loss of surface CD27 and CD28 and the upregulation of terminal differentiation markers. Functionally, senescent T cells demonstrate impaired self-killing and immunosuppressive effects. ↑ represents the activation or upregulation; ↓ represents suppression or downregulation.</p>
Full article ">Figure 2
<p>Timeline of events related to the progression of T cell senescence in AML.</p>
Full article ">Figure 3
<p>Key signaling pathways involved in T cell senescence within the tumor microenvironment. In the immunosuppressive microenvironment formed by tumor cells, the MAPK signaling pathway of T cells is persistently activated by the continuous stimulation of tumor antigens and metabolic competition. Glucose deprivation results in the phosphorylation of P38, resulting in telomere shortening, DNA damage, and cell cycle arrest. Additionally, TCR signal transduction in senescent T cells is downregulated, leading to autophagy inactivation and further exacerbating mitochondrial dysfunction. Treg: regulatory T cell, IFN-γ: interferon-gamma, cAMP: cyclic adenosine monophosphate, ILT4: immunoglobulin-like transcript 4, HLA-G: Human leukocyte antigen-G, PKA: phosphorylase kinase A, CREB: cAMP response element-binding protein, ATM: ataxia-telangiectasia mutated, AMPK: adenosine 5′monophosphate-activated protein kinase, TERT: telomerase reverse transcriptase, ERK: extracellular signal-regulated protein kinase, STAT: signal transducer and activator of transcription, LCK: lymphocyte-specific protein tyrosine kinase, ZAP70: zeta-chain-associated protein kinase 70, ROS: reactive oxygen species, SASP: senescence-associated secretory phenotype, TCR: T cell receptor, AKT: protein kinase B, PI3K: phosphatidylinositide 3-kinase, mTOR: mammalian target of rapamycin, NKG2D: natural killer group 2 member D.</p>
Full article ">
14 pages, 1292 KiB  
Article
The Evolution of Treatment Policies and Outcomes for Patients Aged 60 and Older with Acute Myeloid Leukemia: A Population-Based Analysis over Two Decades
by Benno Diekmann, Nic Veeger, Johanne Rozema, Robby Kibbelaar, Bas Franken, Yasemin Güler, Bram Adema, Eric van Roon and Mels Hoogendoorn
Cancers 2024, 16(23), 3907; https://doi.org/10.3390/cancers16233907 - 21 Nov 2024
Viewed by 224
Abstract
Background: Acute myeloid leukemia (AML) is a malignancy of the bone marrow with a median age at diagnosis of 70 years. AML is difficult to treat, especially in older patients, among whom outcomes have historically been poor. Over the last two decades, a [...] Read more.
Background: Acute myeloid leukemia (AML) is a malignancy of the bone marrow with a median age at diagnosis of 70 years. AML is difficult to treat, especially in older patients, among whom outcomes have historically been poor. Over the last two decades, a greater understanding of the molecular mechanisms of the pathology has led to the development of new drugs and multiple updates to treatment guidelines. Methods: A population-based retrospective cohort study was conducted for all patients aged 60 and older who were newly diagnosed with AML (n = 370) as defined by the European Leukemia Net 2022 criteria in Friesland, a Dutch province, between 2005 and 2023. Results: In this cohort of patients with a median age of 73 years, complete bone marrow analysis to classify the AML according to ELN increased in time from 49% (2005–2011) to 86% (2022–2023). The rate of patients receiving antileukemic therapy increased over time (2005–2011: 19%; 2012–2016: 64%; 2017–2021: 75%; 2022–2023: 74%), mainly driven by the introduction of hypomethylating agents. Over these time periods, the use of intensive chemotherapy (13%, 27%, 27%, and 5%) and rates of stem cell transplantation (3%, 9%, 27%, and 14%) underwent similar development as more patients were deemed eligible for these interventions from 2012 onwards, but usage declined again after the introduction of venetoclax in 2022. The median overall survival was 3.7, 7.3, 8.0, and 9.4 months over the four time periods, respectively. Conclusions: Our study demonstrates how outcomes of patients with newly diagnosed AML aged 60 and older improved over the last two decades. Full article
(This article belongs to the Special Issue Advancements in Treatment Approaches for AML)
Show Figures

Figure 1

Figure 1
<p>The proportion of first-line therapies chosen per period as given in the bar chart diagram [<a href="#B33-cancers-16-03907" class="html-bibr">33</a>].</p>
Full article ">Figure 2
<p>Kaplan–Meier survival analysis per period.</p>
Full article ">Figure 3
<p>Kaplan–Meier survival analysis per first-line therapy.</p>
Full article ">Figure A1
<p>Patients and their first-line therapies in groups of ~50 consecutive patients [<a href="#B33-cancers-16-03907" class="html-bibr">33</a>].</p>
Full article ">
18 pages, 3893 KiB  
Article
Modulatory Effect of Cucurbitacin D from Elaeocarpus hainanensis on ZNF217 Oncogene Expression in NPM-Mutated Acute Myeloid Leukemia
by Sabrina Adorisio, Alessandra Fierabracci, Ba Thi Cham, Vu Dinh Hoang, Nguyen Thi Thuy Linh, Le Thi Hong Nhung, Maria Paola Martelli, Emira Ayroldi, Simona Ronchetti, Lucrezia Rosati, Silvia Di Giacomo, Trinh Thi Thuy and Domenico Vittorio Delfino
Pharmaceuticals 2024, 17(12), 1561; https://doi.org/10.3390/ph17121561 - 21 Nov 2024
Viewed by 200
Abstract
Background/Objectives: The expression of oncogene zinc-finger protein 217 (ZNF217) has been reported to play a central role in cancer development, resistance, and recurrence. Therefore, targeting ZNF217 has been proposed as a possible strategy to fight cancer, and there has been much research on [...] Read more.
Background/Objectives: The expression of oncogene zinc-finger protein 217 (ZNF217) has been reported to play a central role in cancer development, resistance, and recurrence. Therefore, targeting ZNF217 has been proposed as a possible strategy to fight cancer, and there has been much research on compounds that can target ZNF217. The present work investigates the chemo-preventive properties of cucurbitacin D, a compound with a broad range of anticancer effects, in hematological cancer cells, specifically with regard to its ability to modulate ZNF217 expression. Methods: Different cucurbitacins were isolated from the Vietnamese plant Elaeocarpus hainanensis. The purified compounds were tested on nucleophosmin-mutated acute myeloid leukemia and other hematological cancer cell lines to assess their effects on the cell cycle, cell viability and apoptosis, and the expression of ZNF217. Results: Cucurbitacin D resulted in a reduction in the number of acute myeloid leukemia cells by inducing an increase in apoptosis and blocking cell cycle progression. It also led to a significant decrease in ZNF217 expression in the nucleophosmin-mutated acute myeloid leukemia cell line but not in the other hematologic cancer cell lines. The reduction in ZNF217 expression contributed significantly to the blocking of cell cycle progression but did not affect apoptosis. Conclusions: The obtained results suggest that cucurbitacin D is a promising molecule for targeting mutated nucleophosmin or its pathway in acute myeloid leukemia cells, although further studies are needed for in-depth investigations into its specific mechanisms. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of cucurbitacins on OCI-AML3 cell count. The panels on the left depict the chemical structures of cucurbitacin D (<b>A</b>), 3-epi-isocucurbitacin D (<b>B</b>), and cucurbitacin D + cucurbitacin I (<b>C</b>). In the panels on the right, the bars represent the number of viable cells counted after 24 h of treatment with control vehicle (Control), cucurbitacin D (<b>A</b>), 3-epi-isocucurbitacin D (<b>B</b>), or cucurbitacin D + cucurbitacin I (<b>C</b>) at the concentrations shown on the x-axis. The mean ± SEM values were determined from data from five independent experiments. *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 are indicative of a significant decrease in cell viability in comparison to the control (calculated by one-way ANOVA).</p>
Full article ">Figure 2
<p>Effect of CucD and IsocucD on OCI-AML3 cell apoptosis. The bars represent the percentage of apoptotic cells after 24 h of treatment with the control vehicle (Control), CucD (<b>A</b>), or IsocucD (<b>B</b>), at the concentrations shown on the x-axis. The percentage of apoptotic cells was determined based on propidium iodide (PI) staining and is shown on the x-axis as the logarithmic scale values (FL3). (<b>C</b>) The bars represent the fold changes in TNF-α (left panel) and Bcl2 (right panel) after 24 h of treatment with the control vehicle (Control), CucD, or IsocucD at the concentrations reported on the x-axis. The graph shows the mean ± SEM values calculated from data from five independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 indicate a significant increase in the apoptosis rate in comparison to the control (calculated by one-way ANOVA).</p>
Full article ">Figure 3
<p>Effect of CucD and IsocucD on OCI-AML3 cell cycle progression. (<b>A</b>). The bars represent the percentage of cells in different phases of the cell cycle (left panels: G0/G1 phase, middle panels: S phase, right panels: G2/M phase) after 24 h of treatment with DMSO as the vehicle (Control), CucD (<b>A</b>), or IsoCucD (<b>B</b>) at the concentrations shown on the x-axis. Histograms from representative experiments show the values of PI staining on a logarithmic scale (FL2) on the X axes. The mean ± SEM values from five independent experiments are shown. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 indicate significant differences in comparison to the control group (calculated by one-way ANOVA).</p>
Full article ">Figure 4
<p>Effect of CucD and IsocucD on OCI-AML2. The bars represent the number of cells (<b>A</b>), the percentage of apoptotic cells (<b>B</b>), or, in (<b>C</b>), the percentage of cells in the G0/G1 (left), S (middle), or G2/M (right) phases of the cell cycle after 24 h of treatment with the control vehicle (Control), CucD, or IsoCucD, at the concentrations shown on the x-axis. The graphs show the mean ± SEM values calculated from data of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 indicate a significant difference in comparison to the control (calculated by one-way ANOVA).</p>
Full article ">Figure 5
<p>Effects of CucD and IsocucD on the expression of proteins involved in the cell cycle. Western blot illustrating the expression of p21 (in the first panel starting from the upper side), p53 (second line panels), phosphorylated p38 (pp38), total p38 (p38) (third line panels), phosphorylated ERK (pERK), and total ERK (ERK) (fourth line panels) in OCI-AML3 cells treated with vehicle (DMSO), CucD, or IsocucD for 24 h. The Western blots on the left side are representative of five independent experiments, and the corresponding data are quantified in the bar graphs on the right. GADPH served as a loading control. Data are reported as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Effects of the tested compounds on ZNF217 expression in OCI-AML3 cells. (<b>A</b>) Real-time PCR of ZNF217 transcripts in OCI-AML3 cells treated with the vehicle (DMSO, Control), CucD, or IsocucD for 24 h. Gene expression was normalized to the expression of 18 S, and the normalized expression levels are reported (white bar, fold change = 1). The mean ± SEM values from five independent experiments are reported. *** <span class="html-italic">p</span> &lt; 0.001 indicates significant differences in comparison to the control (calculated by one-way ANOVA). (<b>B</b>) Expression of the protein level of ZNF217 in OCI-AML3 cells treated with vehicle (DMSO, Control), Cuc-D, or IsocucD for 24 h. The expression level was normalized to that of laminin expression. The Western blot on the left is representative of four independent experiments, and the corresponding data are quantitatively analyzed in the right panel. The ZNF217/laminin ratio was calculated by densitometric quantification of the specific bands detected in four independent experiments. The mean ± SEM values from four independent experiments are reported. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.01 indicate significant differences in comparison to the control group (calculated by one-way ANOVA).</p>
Full article ">Figure 7
<p>Expression of the protein level of ZNF217 in different cell lines treated with cucurbitacins. The PGA1 (<b>A</b>), MEC1 (<b>B</b>), U9370 (<b>C</b>), and Jurkat (<b>D</b>) cell lines were treated with vehicle (DMSO, Control), CucD, or IsocucD for 24 h. The expression level was normalized to that of laminin expression. The Western blots are representative of three independent experiments. The ZNF217/laminin ratio is calculated by densitometric quantification of the specific bands detected in three independent experiments. Data (mean ± SEM) are reported as fold change in ZNF217 protein expression in samples treated with vehicle (DMSO), CucD, or IsocucD.</p>
Full article ">Figure 8
<p>Comparison of the expression of the protein level of ZNF217 in OCI-AML2 and OCI-AML3 cell lines. OCI-AML2 and OCI-AML3 cell lines were treated with vehicle (DMSO, Control), CucD, or IsocucD for 24 h. Th expression level was normalized to that of GAPDH expression. The Western blots are representative of three independent experiments. The ZNF217/GAPDH ratio is calculated by densitometric quantification of the specific bands detected in three independent experiments. Data (mean ± SEM) are reported as fold change in ZNF217 protein expression in samples treated with vehicle (DMSO), CucD, or IsocucD. * <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 9
<p>Effect of silencing ZNF217 on the viability of OCI-AML3 cells. (<b>A</b>) Expression of the ZNF217 protein in OCI-AML3 cells transfected with ZNF217 (siRNA) for 24 h, 48 h, and 72 h. ZNF217 expression was measured using Western blot analysis and normalized to laminin expression. (<b>B</b>) The Western blot is representative of five independent experiments. The ZNF217/laminin ratio was calculated by densitometric quantification of the specific bands detected in five independent experiments. Data (mean ± SEM) are reported as fold changes in expression in samples transfected with ZNF217 siRNA. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 indicate significant differences in comparison to the control (calculated by one-way ANOVA). (<b>C</b>) At 24 h, 48 h, and 72 h after treatment with the ZNF217 siRNA, the number of viable OCI-AML3 cells was determined by the trypan blue exclusion method. (<b>D</b>) Apoptosis and (<b>E</b>) cell cycle progression were evaluated by PI staining experiments. Data from five independent experiments are reported as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001, and *** <span class="html-italic">p</span> &lt; 0.0001 indicate significant differences in comparison to the control group (calculated by one-way ANOVA).</p>
Full article ">
11 pages, 421 KiB  
Article
Cladribine-Based Therapy for Acute Myeloid Leukemia in Child, Adolescent, and Early Young Adult Patients: The MD Anderson Cancer Center Experience
by David McCall, Shaikha Alqahtani, Moriah Budak, Irtiza Sheikh, Aaron E. Fan, Ramya Ramakrishnan, Cesar Nunez, Michael Roth, Miriam B. Garcia, Amber Gibson, Naval Daver, Sofia Garces, Nicholas J. Short, Ghayas C. Issa, Farhad Ravandi, Courtney D. DiNardo, Guillermo Montalban Bravo, Guillermo Garcia-Manero, Branko Cuglievan and Tapan Kadia
Cancers 2024, 16(22), 3886; https://doi.org/10.3390/cancers16223886 - 20 Nov 2024
Viewed by 248
Abstract
Background: Cladribine-based combination chemotherapy has demonstrated promising efficacy in patients with relapsed/refractory adult acute myeloid leukemia (AML), prompting its increased utilization in the frontline; in pediatrics, it has been typically reserved for relapsed or refractory cases. While fludarabine has been used more commonly [...] Read more.
Background: Cladribine-based combination chemotherapy has demonstrated promising efficacy in patients with relapsed/refractory adult acute myeloid leukemia (AML), prompting its increased utilization in the frontline; in pediatrics, it has been typically reserved for relapsed or refractory cases. While fludarabine has been used more commonly as a purine analog in intensive regimens, cladribine may be an important alternative. Methods: We performed a retrospective study at MD Anderson Cancer Center from January 2015 to July 2023, which included patients aged 1–21 years with refractory or relapsed AML who received cladribine outside of a transplant conditioning. Results: A total of 30 patients were included, with a median age of 20 years (range, 2–21), and 55% being male. Similar to adults, cladribine exhibited good tolerability in pediatric and adolescent patients, with the most common adverse events being febrile neutropenia and myelosuppression. The most common grade 3 or 4 adverse events included febrile neutropenia (55%) and sepsis (26%), and there were no treatment discontinuations due to adverse events. Among patients with a median number of 2 (0–7) prior treatments, the overall response rate (CR/CRi) was 45%, and median event-free and overall survival were 6 and 12 months, respectively. Disease progression resulted in 4 deaths within 30 days of treatment. Conclusions: Cladribine was tolerated in pediatrics. No new safety signals were seen with cladribine regimens in this cohort. Response assessment is limited due to the heavily pretreated cohort. Further prospective studies are warranted on the safety and efficacy of cladribine and establish its role in pediatric, adolescent, and early young adult patients with AML. Full article
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier curve showing overall survival from the start of cladribine therapy for the cohort.</p>
Full article ">
32 pages, 5401 KiB  
Review
Unveiling the Role of New Molecules in Acute Myeloid Leukemia: Insights into Disease Pathogenesis and Therapeutic Potential
by Diana Martinez, Nicole Santoro and Annalisa Paviglianiti
Targets 2024, 2(4), 396-427; https://doi.org/10.3390/targets2040023 - 20 Nov 2024
Viewed by 417
Abstract
This review article explores the current landscape of acute myeloid leukemia treatment, including novel target molecules and recent advancements in cell therapy and immunotherapy focused on T cell activity. Advances in treatment have been promising in recent years, driven by the development of [...] Read more.
This review article explores the current landscape of acute myeloid leukemia treatment, including novel target molecules and recent advancements in cell therapy and immunotherapy focused on T cell activity. Advances in treatment have been promising in recent years, driven by the development of therapies targeting new molecular and genetic therapeutic targets. These findings allowed for the approval of several target therapies by the European and American drug agencies in the last 5 years. However, mortality remains very high, particularly in relapsed or refractory (R/R) patients. In recent years, the development of immunotherapy has expanded this field, leading to the introduction of new drugs and treatments. Full article
(This article belongs to the Special Issue Advances in Targeted Therapy for Hematological Malignancies)
Show Figures

Figure 1

Figure 1
<p>Emerging target therapies for AML.</p>
Full article ">
14 pages, 2426 KiB  
Article
Dose-Reduced FLA-IDA in Combination with Venetoclax Is an Effective and Safe Salvage Therapy in Relapsed and Refractory Acute Myeloid Leukemia (R/R AML)
by Martin Schönrock, Piet Sonnemann, Nina Michalowski, Michael Heuser, Felicitas Thol, Francis Ayuketang Ayuk, Christine Wolschke, Evgeny Klyuchnikov, Carsten Bokemeyer, Walter Fiedler and Sophia Cichutek
Cancers 2024, 16(22), 3872; https://doi.org/10.3390/cancers16223872 - 19 Nov 2024
Viewed by 296
Abstract
Background: Despite the development of targeted therapies in first-line AML, complete remissions (CR) cannot be achieved in 30–40%, and relapse rates remain high. In R/R AML the intensive treatment regimen of fludarabine, cytarabine, idarubicin combined with venetoclax (FLA-VIDA) showed improved remission rates compared [...] Read more.
Background: Despite the development of targeted therapies in first-line AML, complete remissions (CR) cannot be achieved in 30–40%, and relapse rates remain high. In R/R AML the intensive treatment regimen of fludarabine, cytarabine, idarubicin combined with venetoclax (FLA-VIDA) showed improved remission rates compared to FLA-IDA. In this retrospective single-center analysis, we investigated the efficacy and safety of dose-reduced FLA-IDA with and without venetoclax to minimize the risk of infectious complications and excessive myelosuppression; Methods: Between 2011 and 2023, 89 R/R AML patients were treated with dose-reduced FLA-IDA (fludarabine 30 mg/m2 day 1–4, cytarabine 2000 mg/m2 day 1–4, idarubicin 10 mg/m2 day 1 + 4). From 2019 onwards, venetoclax was added (day 1 100 mg, day 2 200 mg, day 3–14 400 mg); Results: Significantly improved response rates were observed with 60.0% vs. 38.8% CR/CRi (p = 0.0297) and 74.5% vs. 47.3% (p = 0.032) CR/CRi/MLFS for FLA-VIDA vs. FLA-IDA. Further, with FLA-VIDA significantly improved event-free survival (EFS) was observed (p = 0.026). Overall survival (OS) was similar in FLA-VIDA and FLA-IDA treated patients. The most common treatment-related toxicities were hematological adverse events, but they were comparable between groups. The time to neutrophil and platelet recovery were similar in responding patients treated with FLA-VIDA vs. FLA-IDA; Conclusions: Dose-reduced FLA-VIDA significantly improved response rates without increases in toxicity, showing promise for an improved R/R AML treatment. Full article
(This article belongs to the Special Issue The Clinical Trials and Management of Acute Myeloid Leukemia)
Show Figures

Figure 1

Figure 1
<p>Comparison of remission rates and overall response rates (ORR) in patients treated with FLA-VIDA vs. FLA-IDA regimen. The response analysis for overall response rate was performed with Fisher’s exact test. N/A: Remission status not evaluable; RD: Refractory disease; ORR: Overall response rate; CR: Complete remission; CRi: Complete remission with incomplete hematological recovery; MLFS: Morphologic leukemia free state.; FLA-VIDA: fludarabine, cytarabine, idarubicin and venetoclax; FLA-IDA: fludarabine, cytarabine, idarubicin without venetoclax.</p>
Full article ">Figure 2
<p>Subgroup analysis for overall response in patients treated with FLA-VIDA vs. FLA-IDA regimen. Odds ratio and confidence intervals calculated according to Altmann 1991. For zero event cells Haldane Correction was performed. Patients without remission evaluation were excluded from analysis (<span class="html-italic">n</span> = 1 in FLA-VIDA; <span class="html-italic">n</span> = 2 in FLA-IDA cohort). Patients without complete NGS were excluded from ELN stratification. Subgroup analysis for IDH1/2 and TP53 omitted due to small sample size. ORR: Overall response rate (CR/CRi/MLFS); FLA-VIDA: fludarabine, cytarabine, idarubicin and venetoclax; FLA-IDA: fludarabine, cytarabine, idarubicin without venetoclax; ELN 2022: European Leukemia Net 2022; AHSCT: allogeneic hematopoietic stem cell transplantation.</p>
Full article ">Figure 3
<p>Event-free survival (EFS) in patients treated with FLA-VIDA vs. FLA-IDA regimen. Events were defined as death of any cause, relapse, or failure to achieve remission/refractory disease. For patients failing to achieve remission time of event was defined as date of first response assessment. HR: Hazard ratio.</p>
Full article ">Figure 4
<p>Overall survival (OS) in patients treated with FLA-VIDA vs. FLA-IDA regimen. HR: Hazard ratio.</p>
Full article ">Figure 5
<p>Time to neutrophil and platelet recovery in responding patients treated with FLA-VIDA vs. FLA-IDA regimen. (<b>A</b>) Absolute neutrophil count (ANC) recovery &gt; 1/nL. (<b>B</b>) Platelet recovery (PLT) &gt; 100/nL. Only patients achieving CR, Cri, or MLFS were included in the analysis. Patients who did not achieve complete recovery within 60 days of administration of FLA-VIDA/FLA-IDA were included in the analysis. Patients who proceeded to transplantation prior to hematological recovery were censored. <span class="html-italic">p</span>-values were calculated via log-rank test.</p>
Full article ">
26 pages, 1685 KiB  
Review
Myeloproliferative Neoplasms: Challenging Dogma
by Jerry L. Spivak
J. Clin. Med. 2024, 13(22), 6957; https://doi.org/10.3390/jcm13226957 - 19 Nov 2024
Viewed by 459
Abstract
Myeloproliferative neoplasms, polycythemia vera, essential thrombocytosis, and primary myelofibrosis are a unique group of clonal hematopoietic stem cell neoplasms that share somatic, gain-in-function driver mutations in JAK2, CALR, and MPL. As a consequence, these disorders exhibit similar phenotypic features, the [...] Read more.
Myeloproliferative neoplasms, polycythemia vera, essential thrombocytosis, and primary myelofibrosis are a unique group of clonal hematopoietic stem cell neoplasms that share somatic, gain-in-function driver mutations in JAK2, CALR, and MPL. As a consequence, these disorders exhibit similar phenotypic features, the most common of which are the ceaseless production of normal erythrocytes, myeloid cells, platelets alone or in combination, extramedullary hematopoiesis, myelofibrosis, and a potential for leukemic transformation. In the case of polycythemia vera and essential thrombocytosis, however, prolonged survival is possible. With an incidence value in the range of 0.5–2.0/100,000, myeloproliferative neoplasms are rare disorders, but they are not new disorders, and after a century of scrutiny, their clinical features and natural histories are well-defined, though their individual management continues to be controversial. With respect to polycythemia vera, there has been a long-standing dispute between those who believe that the suppression of red blood cell production by chemotherapy is superior to phlebotomy to prevent thrombosis, and those who do not. With respect to essential thrombocytosis, there is a similar dispute about the role of platelets in veinous thrombosis, and the role of chemotherapy in preventing thrombosis by suppressing platelet production. Linked to these disputes is another: whether therapy with hydroxyurea promotes acute leukemia in disorders with a substantial possibility of longevity. The 21st century revealed new insights into myeloproliferative neoplasms with the discovery of their three somatic, gain-of-function driver mutations. Almost immediately, this triggered changes in the diagnostic criteria for myeloproliferative neoplasms and their therapy. Most of these changes, however, conflicted with prior well-validated, phenotypically driven diagnostic criteria and the management of these disorders. The aim of this review is to examine these conflicts and demonstrate how genomic discoveries in myeloproliferative neoplasms can be used to effectively complement the known phenotypic features of these disorders for their diagnosis and management. Full article
Show Figures

Figure 1

Figure 1
<p>The hematopoietic stem cell (HSC) hierarchy (modified from Yamamoto et al. [<a href="#B14-jcm-13-06957" class="html-bibr">14</a>]). HSCs are organized in a hierarchy in which the most primitive HSC, the long-term HSC (CD34+CD38-LT-HSC), is mainly dormant in endosteal bone marrow niches, tethered to osteoblasts through adhesive proteins and thrombopoietin. These LT-HSCs also require the presence of closely opposed megakaryocytes, which maintain HSC quiescence though the secretion of thrombopoietin and CXCL4 [<a href="#B15-jcm-13-06957" class="html-bibr">15</a>]. The daily requirement of committed hematopoietic progenitor cells is provided by short-term HSCs (ST-HSCs), which have a limited self-renewal capacity, but a large proliferative capacity. Importantly, the only hematopoietic growth factor receptor expressed by LT-HSCs is the thrombopoietin receptor, MPL. The default commitment pathway for LT-HSCs is to give rise to myeloid repopulating HSCs (MyRPs) with restricted lineage specificity, including megakaryocyte HSCs (MkRPs); megakaryocyte–erythroid HSCs (MERPs), and a common myeloid HSCs (CMRPs), which gives rise to myeloid, erythroid, and megakaryocytic cells. This arrangement reflects the need of LT-HSCs for megakaryocytes as well as thrombopoietin to remain quiescent. It also explains why thrombocytosis is a common presenting manifestation of all three MPNs, and also the pleiomorphic presenting manifestations of MPNs and MPN clonal succession. MPC, multipotent myeloid progenitor cell; LPC, multipotent lymphoid progenitor cell; n/m, neutrophil/monocyte progenitor cell; ery, erythroid progenitor cell; mk, megakaryocytic progenitor cell; B, B-cell progenitor cell; T, T-cell progenitor cell; G-CSFR, granulocyte colony stimulating factor receptor; EPOR, erythropoietin receptor; MPL, thrombopoietin receptor. * Represents the presence of an MPN driver mutation.</p>
Full article ">Figure 2
<p>The correlation between the total body red cell mass measured directly and the peripheral blood hematocrit level in PV patients. The data from reference [<a href="#B24-jcm-13-06957" class="html-bibr">24</a>] have been replotted to illustrate the important fact that the peripheral blood hematocrit in untreated PV patients fails to correlate with the actual total body hematocrit, even at values falling within the normal hematocrit ranges for normal men and women. As a corollary, even for hematocrits greater than normal, until the hematocrit is greater than 59%, the possibility of a reduced plasma volume still exists (pseudopolycythemia). The red arrows represent the recommended phlebotomy target hematocrits for men and women PV patients, as well as for PV patients with splanchnic vein thrombosis or who are pregnant, respectively.</p>
Full article ">Figure 3
<p>An algorithm for the evaluation of isolated erythrocytosis. In this situation, the frequency of benign causes for an elevated hematocrit or red cell count far outnumbers PV as a cause of isolated erythrocytosis, and an assay for the <span class="html-italic">JAK</span>2 or <span class="html-italic">LNK</span> mutation is rarely necessary. * this MAB distinguishes PV from CHIP.</p>
Full article ">
25 pages, 15916 KiB  
Review
Acute Myeloid Leukemia: Diagnosis and Evaluation by Flow Cytometry
by Feras Ally and Xueyan Chen
Cancers 2024, 16(22), 3855; https://doi.org/10.3390/cancers16223855 - 17 Nov 2024
Viewed by 315
Abstract
With recent technological advances and significant progress in understanding the pathogenesis of acute myeloid leukemia (AML), the updated fifth edition WHO Classification (WHO-HAEM5) and the newly introduced International Consensus Classification (ICC), as well as the European LeukemiaNet (ELN) recommendations in 2022, require the [...] Read more.
With recent technological advances and significant progress in understanding the pathogenesis of acute myeloid leukemia (AML), the updated fifth edition WHO Classification (WHO-HAEM5) and the newly introduced International Consensus Classification (ICC), as well as the European LeukemiaNet (ELN) recommendations in 2022, require the integration of immunophenotypic, cytogenetic, and molecular data, alongside clinical and morphologic findings, for accurate diagnosis, prognostication, and guiding therapeutic strategies in AML. Flow cytometry offers rapid and sensitive immunophenotyping through a multiparametric approach and is a pivotal laboratory tool for the classification of AML, identification of therapeutic targets, and monitoring of measurable residual disease (MRD) post therapy. The association of immunophenotypic features and recurrent genetic abnormalities has been recognized and applied in informing further diagnostic evaluation and immediate therapeutic decision-making. Recently, the evolving role of machine learning models in assisting flow cytometric data analysis for the automated diagnosis and prediction of underlying genetic alterations has been illustrated. Full article
(This article belongs to the Special Issue Flow Cytometry of Hematological Malignancies)
Show Figures

Figure 1

Figure 1
<p>Gating strategy and normal myeloid maturation in normal bone marrow. (<b>A</b>) All the events are displayed on a CD45 versus event number plot to ensure stable data acquisition. (<b>B</b>) A singlet gate is applied to exclude doublets. (<b>C</b>) A viability gate is applied to exclude poorly viable cells and debris. (<b>D</b>) The CD45 versus side scatter (SSC) plot defines the blast gate (low CD45 and low SSC), lymphocyte gate (high CD45 and low SSC), and myelomonocytic gate (maturing myeloid cells colored green and monocytes in pink). (<b>E</b>) Events in the blast gate are displayed. A CD34-positive myeloid blast gate is generated to separate myeloid blasts from CD19-positive hematogones (colored aqua), as well as myeloid and monocytic cells. (<b>F</b>–<b>H</b>) Normal myeloid maturation from early progenitors to promyelocytes. The black line follows changes in the intensity of antigen expression. Early progenitors (highlighted in orange) express bright CD34, dim CD38, and dim CD33 without CD15. During differentiation, the progenitors gradually lose CD34 while acquiring CD15 and express higher levels of CD33 and CD38 as they reach the promyelocyte stage (highlighted in purple).</p>
Full article ">Figure 2
<p>Acute promyelocytic leukemia with <span class="html-italic">PML::RARA</span> fusion. The upper left dot plot displays the total viable white blood cells with blasts/blast equivalents colored in red, lymphocytes in blue, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts/blast equivalents. The leukemic blasts/blast equivalents (colored red; ~74% of the white blood cells) in the peripheral blood show variably increased SSC and express CD13 (increased, heterogeneous), CD15 (dim, variable), CD33 (uniform), CD34 (decreased to mostly absent), CD38 (dim), CD56 (major subset), CD64 (variable), CD117 (dim to absent), CD123, and HLA-DR (dim to mostly absent).</p>
Full article ">Figure 3
<p>Expanded basophilic population in a patient with APL, ~7 days post all-trans retinoic acid (ATRA) treatment. All the dot plots display the total viable white blood cells with basophilic cells colored in orange, lymphocytes in blue, and granulocytic cells in green. Monocytes are few in number. The basophilic population (colored orange; ~16% of the white blood cells) in the peripheral blood expresses CD13 (bright), CD33, CD34 (dim), CD38, CD45, CD117 (dim), CD123 (bright), and CD203c without HLA-DR.</p>
Full article ">Figure 4
<p>Acute myeloid leukemia with basophilic/mast cell differentiation. The upper left dot plot displays the total viable white blood cells with blasts colored in red, lymphocytes in blue, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts. The leukemic blasts (colored red; ~55% of the white blood cells) in the bone marrow express CD13 (increased, uniform), CD15 (dim, variable), CD33 (increased), CD34 (bright), CD38 (variably decreased), CD64 (dim, variable), CD117, CD123, and HLA-DR (variable). Additionally, a CD117-positive progenitor subset (colored purple; ~7% of the white blood cells) with basophilic and/or mast cell differentiation that appears in a continuum with the first abnormal blast population is present with the expression of CD13, CD33 (dim, variable), CD34 (dim, variable), CD38, CD117, and CD123 without CD15, CD64, or HLA-DR.</p>
Full article ">Figure 5
<p>Acute myeloid leukemia with <span class="html-italic">FLT3 ITD</span> and <span class="html-italic">NPM1</span> mutations. The upper left dot plot displays the total viable white blood cells with blasts colored in red, lymphocytes in blue, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts. The leukemic blasts (colored red; ~63% of the white blood cells) in the bone marrow express CD7, CD13 (variably decreased to absent), CD33 (uniform), CD38 (bright), CD117 (dim to mostly absent), CD123 (uniform), and HLA-DR (variably decreased to absent) without CD34 or CD64. While some immunophenotypic features overlap with acute promyelocytic leukemia (APL), SSC and the levels of CD13 of the leukemic blasts are lower than what is typically seen in APL.</p>
Full article ">Figure 6
<p>Acute myeloid leukemia with plasmacytoid dendritic cell (pDC) expansion and a <span class="html-italic">RUNX1</span> mutation. The upper left dot plot displays the total viable white blood cells with blasts colored in red, pDCs in aqua (highlighted), lymphocytes in blue, monocytes in pink, and granulocytic cells in green. The rest of the dot plots in the upper panel selectively display CD34+ leukemic blasts. The leukemic blasts (colored red; ~20% of the white blood cells) in the peripheral blood express CD13 (bright), CD33 (dim to mostly absent), CD34 (bright), CD38, CD56 (small subset), CD117 (decreased), CD123, and HLA-DR (variably decreased). In the lower panel, all the dot plots display the total white cells. The pDCs (highlighted in aqua; ~15% of the white blood cells) express CD4, CD5 (major subset), CD7 (major subset), CD34 (dim, variable), CD38, CD45, CD56, CD123, and HLA-DR (dim, variable) without CD2, CD13, CD14, CD33, CD64, and CD117.</p>
Full article ">Figure 7
<p>Acute myeloid leukemia with erythroid differentiation. The upper left dot plot displays the total viable white blood cells, with blasts colored in red, lymphocytes in blue, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts. The leukemic blasts (colored red; ~22% of the white blood cells) in the bone marrow abnormally express CD4 (dim on a small subset), CD13 (dim, variable), CD33 (variable), CD38 (variably decreased), CD45 (dim), CD71 (bright on a major subset), CD117, CD123 (dim), and HLA-DR (dim to absent) without CD34.</p>
Full article ">Figure 8
<p>Acute myeloid leukemia with megakaryocytic differentiation. The upper left dot plot displays the total viable white blood cells, with blasts colored in red, lymphocytes in blue, hematogones in aqua, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts. The leukemic blasts (colored red; ~31% of the white blood cells) in the bone marrow express CD2 (small subset), CD7 (subset), CD33 (dim to mostly absent), CD34 (bright), CD38 (dim), CD41 (dim), CD45 (variably decreased to absent), CD61, CD71, and CD123 (dim) without CD13, CD15, CD64, CD117, or HLA-DR.</p>
Full article ">Figure 9
<p>Acute myeloid leukemia with RAM phenotype. The left dot plot displays the total viable white blood cells, with blasts colored in red, lymphocytes in blue, hematogones in aqua, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts. The leukemic blasts (colored red; ~21% of the white blood cells) in the bone marrow express CD33 (bright), CD34 (variably decreased), CD38 (dim to absent), CD45 (dim to absent), CD56 (bright), and CD117 without HLA-DR. A small normal myeloid blast population (colored purple) is also present.</p>
Full article ">Figure 10
<p>Mixed phenotype acute leukemia, B/myeloid. The left dot plot displays the total viable white blood cells, with two blast populations shown in purple and red, lymphocytes in blue, monocytes in pink, and granulocytic cells in green. The rest of the dot plots selectively display leukemic blasts. The blasts with B lineage differentiation (purple) strongly express B-cell markers CD10, CD19, and CD79a, while lacking monocytic markers CD14 and CD64. In contrast, the blasts with myelomonocytic differentiation (red) variably express CD14 and CD64 and do not express B-cell markers.</p>
Full article ">
14 pages, 913 KiB  
Review
Decoding Acute Myeloid Leukemia: A Clinician’s Guide to Functional Profiling
by Prasad Iyer, Shaista Shabbir Jasdanwala, Yuhan Wang, Karanpreet Bhatia and Shruti Bhatt
Diagnostics 2024, 14(22), 2560; https://doi.org/10.3390/diagnostics14222560 - 14 Nov 2024
Viewed by 418
Abstract
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains [...] Read more.
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains a significant therapeutic challenge. Functional profiling techniques, such as BH3 profiling (BP), gene expression profiling (GEP), proteomics, metabolomics, drug sensitivity/resistance testing (DSRT), CRISPR/Cas9, and RNAi screens offer valuable insights into the functional behavior of leukemia cells. BP evaluates the mitochondrial response to pro-apoptotic BH3 peptides, determining a cell’s apoptotic threshold and its reliance on specific anti-apoptotic proteins. This knowledge can pinpoint vulnerabilities in the mitochondria-mediated apoptotic pathway in leukemia cells, potentially informing treatment strategies and predicting therapeutic responses. GEP, particularly RNA sequencing, evaluates the transcriptomic landscape and identifies gene expression alterations specific to AML subtypes. Proteomics and metabolomics, utilizing mass spectrometry and nuclear magnetic resonance (NMR), provide a detailed view of the active proteins and metabolic pathways in leukemia cells. DSRT involves exposing leukemia cells to a panel of chemotherapeutic and targeted agents to assess their sensitivity or resistance profiles and potentially guide personalized treatment strategies. CRISPR/Cas9 and RNAi screens enable systematic disruption of genes to ascertain their roles in leukemia cell survival and proliferation. These techniques facilitate precise disease subtyping, uncover novel biomarkers and therapeutic targets, and provide a deeper understanding of drug-resistance mechanisms. Recent studies utilizing functional profiling have identified specific mutations and gene signatures associated with aggressive AML subtypes, aberrant signaling pathways, and potential opportunities for drug repurposing. The integration of multi-omics approaches, advances in single-cell sequencing, and artificial intelligence is expected to refine the precision of functional profiling and ultimately improve patient outcomes in AML. This review highlights the diverse landscape of functional profiling methods and emphasizes their respective advantages and limitations. It highlights select successes in how these methods have further advanced our understanding of AML biology, identifies druggable targets that have improved outcomes, delineates challenges associated with these techniques, and provides a prospective view of the future where these techniques are likely to be increasingly incorporated into the routine care of patients with AML. Full article
(This article belongs to the Section Clinical Laboratory Medicine)
Show Figures

Figure 1

Figure 1
<p>Overview of different types of functional profiling methods and their utility in target identification, disease subtyping, and potential to improve precision medicine.</p>
Full article ">
20 pages, 1427 KiB  
Review
Current Understanding of the Role of Autophagy in the Treatment of Myeloid Leukemia
by Yasushi Kubota and Shinya Kimura
Int. J. Mol. Sci. 2024, 25(22), 12219; https://doi.org/10.3390/ijms252212219 - 14 Nov 2024
Viewed by 386
Abstract
The most important issues in acute myeloid leukemia are preventing relapse and treating relapse. Although the remission rate has improved to approximately 80%, the 5-year survival rate is only around 30%. The main reasons for this are the high relapse rate and the [...] Read more.
The most important issues in acute myeloid leukemia are preventing relapse and treating relapse. Although the remission rate has improved to approximately 80%, the 5-year survival rate is only around 30%. The main reasons for this are the high relapse rate and the limited treatment options. In chronic myeloid leukemia patients, when a deep molecular response is achieved for a certain period of time through tyrosine kinase inhibitor treatment, about half of them will reach treatment-free remission, but relapse is still a problem. Therefore, potential therapeutic targets for myeloid leukemias are eagerly awaited. Autophagy suppresses the development of cancer by maintaining cellular homeostasis; however, it also promotes cancer progression by helping cancer cells survive under various metabolic stresses. In addition, autophagy is promoted or suppressed in cancer cells by various genetic mutations. Therefore, the development of therapies that target autophagy is also being actively researched in the field of leukemia. In this review, studies of the role of autophagy in hematopoiesis, leukemogenesis, and myeloid leukemias are presented, and the impact of autophagy regulation on leukemia treatment and the clinical trials of autophagy-related drugs to date is discussed. Full article
(This article belongs to the Special Issue The Role of Autophagy in Disease and Cancer)
Show Figures

Figure 1

Figure 1
<p>Autophagy pathway. The membrane that forms at the contact site between the mitochondria and endoplasmic reticulum extends to surround the degradation products at both ends and closes (autophagosome). After that, it fuses with the lysosome to become an autolysosome, and the internal contents are broken down by digestive enzymes. Lysosomes are regenerated from the autolysosome.</p>
Full article ">Figure 2
<p>Formation of autophagosomes. The ULK1 complex, which is involved in the initiation of autophagy, is inhibited by the mTORC1 kinase complex, so autophagy is induced when TORC1 is inactivated by factors such as nutrient starvation. When the ULK1 complex migrates to a subdomain of the endoplasmic reticulum (ER), the PI3K complex I is recruited, and the production of PI3P production increases. The PI3P-binding protein WIPI binds to it and accumulates at the site of autophagosome formation together with its partner ATG2. ATG2 anchors the ER and the phagophore and transports lipids. The ATG12 system is a system in which ATG12 and ATG5 are covalently bound to each other via a ubiquitin-like binding reaction. The ATG12–ATG5 complex forms a ternary complex with ATG16L and localizes to the phagophore, where it determines the location of amide bond formation between ATG8 family proteins (LC3B) and PE. LC3B-PE localizes to the inner and outer membranes of the phagophore and autophagosome, and it functions in membrane elongation and closure. TORC1, target of rapamycin complex 1; ULK1, Unc51-like kinase 1; PI3K, phosphatidylinositol-3 kinase; PI3P, phosphatidylinositol-3-phosphate; WIPI, WD repeat domain phosphoinositide-interacting; ATG, autophagy-related protein; LC3B, light chain 3B; FIP200, focal adhesion kinase interacting protein; VPS34, vacuolar protein sorting 34.</p>
Full article ">Figure 3
<p>Effects of activating and inhibiting autophagy on leukemia cells. Although various anticancer drugs and radiation therapy have antitumor effects, they also activate autophagy in leukemia cells. This leads to resistance to antileukemia therapy and the progression of leukemia. It is thought that the use of autophagy inhibitors in combination can compensate for this drawback. However, drugs that promote autophagy are thought to promote the activation of autophagy beyond the maintenance of leukemia homeostasis, leading to cell death.</p>
Full article ">
14 pages, 1129 KiB  
Review
A Review of Limbic System-Associated Membrane Protein in Tumorigenesis
by Kayleigh Wittmann Sinopole, Kevin Babcock, Albert Dobi and Gyorgy Petrovics
Biomedicines 2024, 12(11), 2590; https://doi.org/10.3390/biomedicines12112590 - 13 Nov 2024
Viewed by 410
Abstract
Purpose of Review: This review aims to describe the role of limbic system-associated membrane protein (LSAMP) in normal- and pathophysiology, and its potential implications in oncogenesis. We have summarized research articles reporting the role of LSAMP in the development of a variety of [...] Read more.
Purpose of Review: This review aims to describe the role of limbic system-associated membrane protein (LSAMP) in normal- and pathophysiology, and its potential implications in oncogenesis. We have summarized research articles reporting the role of LSAMP in the development of a variety of malignancies, such as clear cell renal cell carcinoma, prostatic adenocarcinoma, lung adenocarcinoma, osteosarcoma, neuroblastoma, acute myeloid leukemia, and epithelial ovarian cancer. We also examine the current understanding of how defects in LSAMP gene function may contribute to oncogenesis. Finally, this review discusses the implications of future LSAMP research and clinical applications. Recent Findings: LSAMP has been originally described as a surface adhesion glycoprotein expressed on cortical and subcortical neuronal somas and dendrites during the development of the limbic system. It is categorized as part of the IgLON immunoglobulin superfamily of cell-adhesion molecules and is involved in regulating neurite outgrowth and neural synapse generation. LSAMP is both aberrantly expressed and implicated in the development of neuropsychiatric disorders due to its role in the formation of specific neuronal connections within the brain. Additionally, LSAMP has been shown to support brain plasticity via the formation of neuronal synapses and is involved in modulating the hypothalamus in anxiogenic environments. In murine studies, the loss of LSAMP expression was associated with decreased sensitivity to amphetamine, increased sensitivity to benzodiazepines, increased hyperactivity in new environments, abnormal social behavior, decreased aggressive behavior, and decreased anxiety. Findings have suggested that LSAMP plays a role in attuning serotonergic activity as well as GABA activity. Given its importance to limbic system development, LSAMP has also been studied in the context of suicide. In malignancies, LSAMP may play a significant role as a putative tumor suppressor, the loss of which leads to more aggressive phenotypes and mortality from metastatic disease. Loss of the LSAMP gene facilitates epithelial-mesenchymal transition, or EMT, where epithelial cells lose adhesion and gain the motile properties associated with mesenchymal cells. Additionally, LSAMP and the function of the RTK pathway have been implicated in tumorigenesis through the modulation of RTK expression in cell membranes and the activation of second messenger pathways and β-catenin. Summary: Beyond its many roles in the limbic system, LSAMP functions as a putative tumor suppressor protein. Loss of the LSAMP gene is thought to facilitate epithelial-mesenchymal transition, or EMT, where cells lose adhesion and migrate to distant organs. LSAMP’s role in modulating RTK activity and downstream ERK and Akt pathways adds to a large body of data investigating RTK expression in oncogenesis. The characteristics of LSAMP defects and their association with aggressive and metastatic disease are evident in reports on clear cell renal cell carcinoma, prostatic adenocarcinoma, lung adenocarcinoma, osteosarcoma, neuroblastoma, acute myeloid leukemia, and epithelial ovarian cancer. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment: Second Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>LSAMP in prostate cancer. The loss of LSAMP in the cell membrane upregulates RTK signaling and downstream ERK and Akt pathways. Deletion in LSAMP results in the loss of adhesion to normal extracellular matrix elements, leading to detachment of a nest of tumor cells from the primary tumor site and attachment to lymphatic or vascular endothelium where the LSAMP-deleted cancer cells spread to distant sites, such as lymph node and bone. Abbreviations: ERK, extracellular signal-regulated kinase; FAK, focal adhesion kinase; MEK, mitogen-activated protein kinase; RTK, receptor tyrosine kinase.</p>
Full article ">Figure 2
<p>LSAMP losses in the setting of cancer may increase the expression of receptor tyrosine kinases on the surface of the cell, leading to the increased activation of tumorigenic ERK and Akt pathways and increased activation of β-catenin.</p>
Full article ">
18 pages, 1512 KiB  
Review
Targeting Menin in Acute Myeloid Leukemia: Therapeutic Advances and Future Directions
by Sandhya Dhiman, Vikram Dhillon and Suresh Kumar Balasubramanian
Cancers 2024, 16(22), 3743; https://doi.org/10.3390/cancers16223743 - 6 Nov 2024
Viewed by 716
Abstract
Germline mutations in the MEN1 gene encoding menin protein cause multiple endocrine neoplasia type 1 (MEN1) syndrome. Recent evidence suggests that inhibiting the interaction of menin with its crucial oncogenic protein partners represents a promising therapeutic strategy to AML. Menin plays a critical [...] Read more.
Germline mutations in the MEN1 gene encoding menin protein cause multiple endocrine neoplasia type 1 (MEN1) syndrome. Recent evidence suggests that inhibiting the interaction of menin with its crucial oncogenic protein partners represents a promising therapeutic strategy to AML. Menin plays a critical role in lysine methyltransferase 2A (KMT2A)-gene-rearranged and NPM1-m acute leukemias, both associated with adverse outcomes with current standard therapies, especially in the relapsed/refractory setting. Disrupting the menin–KMT2A interaction affects the proleukemogenic HOX/MEIS transcription program. This disruption leads to the differentiation of KMT2Ar and NPM1-m AML cells. Small molecular inhibitors of the menin–KMT2A interaction target the central cavity of MEN1 to inhibit the MEN1-KMT2A interaction and could target a similar transcriptional dependency in other leukemia subsets, broadening their therapeutic potential. These agents, both as monotherapies and in combination with synergistic drugs, are undergoing preclinical and clinical evaluation with promising early results. With the growing literature around menin inhibitors in AML, we discussed the biology of menin, its mechanism of action, its interacting partners in leukemia, possible inhibitors, their implications, synergistic drugs, and future therapeutic strategies in this review. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Structure of menin and its functions: (<b>A</b>) Chromosomal location of <span class="html-italic">MEN1</span> on chromosome 11q13 with ten exons. (<b>B</b>) The scaffolding function of menin regulating gene expression by interacting with different classes of chromatin regulators and transcription factors (protein fusion partners in red; gene fusion partners in green; MLL-Fp is MLL fusion partners represented above the funnel). (<b>C</b>) Structural representation of wild-type MLL with the different functional domains indicated by colors. MBD is the menin-binding domain; AT is the AT hooks; SNL are the Speckled Nuclear Localization domains; RDs are the repression domains; the black line in the first RD is the CXXC domain; BCR is the breakpoint cluster region; MLL fusion proteins are the result of chromosomal rearrangements between N-terminal MLL up to the BCR and any of the 80 fusion partners; PHDs are the four PHD fingers; BD is the bromodomain; CS1 and CS2 are the taspase-1 cleavage sites; FYRN to FYRC is the region where MLL-N and MLL-C will interact after cleavage; TAD is the transactivation domain; SET is histone methyltransferase domain.</p>
Full article ">Figure 2
<p>Menin and signaling pathways. (A) Menin participates in Wnt signaling by interacting with β-catenin which is located at the cell membrane and translocated to the nucleus in presence of Wnt. (B) Menin also participates in nuclear receptor signaling, directly interacting with ERα in a hormone-dependent manner and is recruited to the ERα target gene (TFF)1. It increases the active histone mark H3K4me3, recruiting MLL and activating target gene expression. (C) Menin inhibits extracellular signal-regulated kinase (ERK)-dependent phosphorylation, a downstream target in the Ras pathway. It inhibits receptor tyrosine kinase signaling in the cytoplasm by inhibiting AKT, SOS1-dependent activation of Ras, and suppression of extracellular signal-regulated kinase (ERK) activation. (D) In Akt signaling, menin suppresses both Akt1-dependent proliferation and antiapoptotic activity by reducing the translocation of Akt1 from the cytoplasm to the plasma membrane. (E) Menin interacts with nuclear factor (NF)kB and recruits Sirtuin (Sirt1) to deacetylate p65b and suppress NFkB-induced gene expression. (F) Menin regulates BMP signaling and TGF-β/SMAD pathways by interacting with SMAD3 or SMAD1/5 as a transcriptional co-regulator, enhancing or modulating the transcriptional activity of SMADs in response to the signaling cues from either TGF-β or BMP ligands. CDKN2B (p15) and CDKN1B (p27); TGF-β target genes; JUNB; and TGFB1 are some of the genes regulated by the MEN1-SMADs interaction. EGFR, epidermal growth factor receptor; GRB, growth factor receptor-bound; GSK3b, glycogen synthase kinase; ERα, Estrogen Receptor; SOS, son of Sevenless; RAF, rapidly accelerated fibrosarcoma; IKK, inhibitor of nuclear factor k-B kinase; BMP, Bone Morphogenic Protein; IL, interleukin; PI3K, phosphoinositide 3-kinase; TFF, trefoil factor; TNF, tumor necrosis factor. Bold arrows show strong activation or direct stimulatory action, whereas dotted arrows show indirect effect or tentative stimulatory action.</p>
Full article ">Figure 3
<p>Menin and its interacting partners: (<b>A</b>) Menin enhances MYC-mediated transcription. It facilitates RNA Pol II phosphorylation to enhance MYC-mediated transcription and promote MYC-mediated cell proliferation. (<b>B</b>) Transcription factor JunD interacts with activated protein 1 (AP-1) and forms an active transcription complex. Interaction of menin with JunD downregulates cyclinD1, reducing cell proliferation.</p>
Full article ">Figure 4
<p>Mechanism of action of small-molecule menin inhibitors in <span class="html-italic">KMT2A</span>r AML and <span class="html-italic">NPM1</span>-m AML. Menin binds with KMT2A and overexpresses the <span class="html-italic">HOX</span> gene along with their cofactor <span class="html-italic">MEIS1</span>. <span class="html-italic">NPM1</span>-m turns exclusively cytoplasmic when mutated (NPM1c) with upregulation of <span class="html-italic">HOX</span> genes. Ziftomenib and revumenib (menin inhibitors) disrupt the menin–KMT2A interaction, suppressing <span class="html-italic">HOX</span> gene expression and inhibiting aberrant leukemogenesis.</p>
Full article ">
13 pages, 9132 KiB  
Article
Fluorescent Aerolysin (FLAER) Binding Is Abnormally Low in the Clonal Precursors of Acute Leukemias, with Binding Particularly Low or Absent in Acute Promyelocytic Leukemia
by María Beatriz Álvarez Flores, María Sopeña Corvinos, Raquel Guillén Santos and Fernando Cava Valenciano
Int. J. Mol. Sci. 2024, 25(22), 11898; https://doi.org/10.3390/ijms252211898 - 5 Nov 2024
Viewed by 612
Abstract
Flow cytometry plays a fundamental role in the diagnosis of leukemias and lymphomas, as well as in the follow-up and evaluation of minimally measurable disease after treatment. In some instances, such as in the case of acute promyelocytic leukemia (APL), rapid diagnosis is [...] Read more.
Flow cytometry plays a fundamental role in the diagnosis of leukemias and lymphomas, as well as in the follow-up and evaluation of minimally measurable disease after treatment. In some instances, such as in the case of acute promyelocytic leukemia (APL), rapid diagnosis is required to avoid death due to serious blood clotting or bleeding complications. Given that promyelocytes do not express the glycophosphatidylinositol (GPI)-anchored protein CD16 and that deficient CD16 expression is a feature of some CD16 polymorphisms and paroxysmal nocturnal hemoglobinuria (PNH), we included the GPI anchor probe FLAER aerolysin in the APL flow cytometry probe panel. Initial tests showed that FLAER binding was absent in pathological promyelocytes from APL patients but was consistently detected with high intensity in healthy promyelocytes from control bone marrow. FLAER binding was studied in 71 hematologic malignancies. Appropriate control cells were obtained from 16 bone marrow samples from patients with idiopathic thrombocytopenic purpura and non-infiltrated non-Hodgkin’s lymphoma. Compared with the positive FLAER signal in promyelocytes from healthy bone marrow, malignant promyelocytes from APL patients showed weak or negative FLAER binding. The FLAER signal in APL promyelocytes was also lower than that in control myeloid progenitors and precursors from patients with other forms of acute myeloid leukemia (AML), B-cell acute lymphoblastic leukemia, or myelodysplastic syndrome. Minimal measurable disease studies performed in APL patients after treatment found normal promyelocyte expression when minimal measurable disease was negative and FLAER-negative promyelocytes when disease relapse was detected. The inclusion of FLAER in the flow cytometry diagnosis and follow-up of APL could be very helpful. Decreased FLAER binding was found in all cases of APL, confirmed by the detection of the PML-RARA fusion transcript and, to a lesser extent, in the other AMLs studied. This study also revealed FLAER differences in other acute leukemias and even between different precursors (myeloid and lymphoid) from healthy controls. However, the reason for FLAER’s non-binding to the malignant precursors of these leukemias remains unknown, and future studies should explore the possible relation with an immune escape phenomenon in these leukemias. Full article
(This article belongs to the Special Issue Flow Cytometry: Applications and Challenges)
Show Figures

Figure 1

Figure 1
<p>Differential FLAER means fluorescence intensity (MFI) (<b>A</b>) and FLAER ratio (<b>B</b>) in malignant promyelocytes from patients with acute promyelocytic leukemia (APL, M3), B-ALL, and control samples derived from bone marrow, including myeloid precursors, promyelocytes, and B-cell precursors. **** <span class="html-italic">p</span> &lt; 0.0001; *** <span class="html-italic">p</span> &lt; 0.005; ** <span class="html-italic">p</span> &lt; 0.005; ns, non-significant (Mann Whitney U test). B-ALL, B-cell acute lymphoblastic leukemia; M3, APL with PML-RARA fusion (FAB classification).</p>
Full article ">Figure 2
<p>FLAER binding to promyelocytes from four control bone marrow samples (C1–C4, top row), contrasting with low binding to malignant promyelocytes from four bone marrow aspirates obtained from APL patients at diagnosis (APL 1–APL 4). Promyelocytes are depicted in fucsia.</p>
Full article ">Figure 3
<p>Comparison of FLAER MFI (<b>A</b>) and FLAER ratio (<b>B</b>) in the studied AMLs and control myeloid precursors. Control myeloid precursor samples were derived from bone marrow controls.**** <span class="html-italic">p</span> &lt; 0.0001; *** <span class="html-italic">p</span> &lt; 0.005; ** <span class="html-italic">p</span> &lt; 0.005; * <span class="html-italic">p</span> &lt; 0.05; ns, non-significant (Mann–Whitney U test).</p>
Full article ">Figure 4
<p>Representative promyelocyte analysis in a patient at the time of APL diagnosis (top row, (<b>a</b>–<b>d</b>)) and upon completion of remission induction therapy with ATRA (bottom row, (<b>e</b>,<b>f</b>)). The analysis shows cellular complexity in relation to CD16, CD15, and HLA-DR expression and FLAER binding. Recovery of cellular complexity after treatment is evident from the granulocytic component populations (<b>e</b>) and the increases in CD15 expression and FLAER binding (<b>f</b>,<b>h</b>). The HLA-DR expression remains negative, like normal promyelocytes (<b>g</b>).</p>
Full article ">Figure 5
<p>Differences in FLAER binding between AML samples relative to control myeloid precursors.</p>
Full article ">Figure 6
<p>Proposed workflow algorithm for the identification of APL. If immunophenotyping of a suspected case of APL-derived AML identifies pathological cells with an APL-like phenotype, the FLAER MFI is assessed. Cells lacking an APL-like phenotype or having a FLAER MFI &gt; 2500 exclude suspicion of APL. Conversely, if the FLAER MFI is ≤2500, the CD13/CD33 expression pattern is analyzed. A compatible expression pattern strongly suggests a diagnosis of APL, to be confirmed by PML-RARA rearrangement analysis. An incompatible CD13/CD33 pattern suggests that APL is unlikely; nevertheless, PML-RARA rearrangement analysis is recommended to exclude APL.</p>
Full article ">Figure 7
<p>(<b>A</b>) Representative differential expression of CD13 and CD33 in normal myeloid precursors (cyan), promyelocytes from a patient with APL (fuchsia), and myeloid precursors with low FLAER MFI from a patient with non-promyelocytic AML (khaki). The pattern compatible with APL AML is characterized by high expression of CD33 and, to a lesser degree, CD13. (<b>B</b>) Histograms showing the expression of the APL phenotype (CD117+, CD34−/+, and HLA-DR-) alongside CD13, CD33, CD45, and CD16 in the defined populations. Circles denote median values.</p>
Full article ">Figure 8
<p>Representative SSC-A/FLAER plots in A) non-APL AML and B) APL AML. (<b>A</b>) The orange population corresponds to non-APL AML cells, while the green represents lymphocytes in the same sample. (<b>B</b>) Fuchsia denotes the APL promyelocyte population, and blue represents lymphocytes in the same sample. Circles correspond to median values, and the squares represent the mean fluorescence intensity of FLAER, confirming that they are comparable.</p>
Full article ">
15 pages, 2303 KiB  
Article
The Immunomodulatory Effect of Different FLT3 Inhibitors on Dendritic Cells
by Sebastian Schlaweck, Alea Radcke, Sascha Kampmann, Benjamin V. Becker, Peter Brossart and Annkristin Heine
Cancers 2024, 16(21), 3719; https://doi.org/10.3390/cancers16213719 - 4 Nov 2024
Viewed by 565
Abstract
Background: FMS-like tyrosine kinase 3 (FLT3) mutations or internal tandem duplication occur in 30% of acute myeloid leukemia (AML) cases. In these cases, FLT3 inhibitors (FLT3i) are approved for induction treatment and relapse. Allogeneic hematopoietic stem cell transplantation (alloHSCT) remains the recommended post-induction [...] Read more.
Background: FMS-like tyrosine kinase 3 (FLT3) mutations or internal tandem duplication occur in 30% of acute myeloid leukemia (AML) cases. In these cases, FLT3 inhibitors (FLT3i) are approved for induction treatment and relapse. Allogeneic hematopoietic stem cell transplantation (alloHSCT) remains the recommended post-induction therapy for suitable patients. However, the role of FLT3i therapy after alloHSCT remains unclear. Therefore, we investigated the three currently available FLT3i, gilteritinib, midostaurin, and quizartinib, in terms of their immunosuppressive effect on dendritic cells (DCs). DCs are professional antigen-presenting cells inducing T-cell responses to infectious stimuli. Highly activated DCs can also cause complications after alloHSCT, such as triggering Graft versus Host disease, a serious and potentially life-threatening complication after alloHSCT. Methods: To study the immunomodulatory effects on DCs, we differentiated murine and human DCs in the presence of FLT3i and performed immunophenotyping by flow cytometry and cytokine measurements and investigated gene and protein expression. Results: We detected a dose-dependent immunosuppressive effect of midostaurin, which decreased the expression of costimulatory markers like CD86, and found a reduced secretion of pro-inflammatory cytokines such as IL-12, TNFα, and IL-6. Mechanistically, we show that midostaurin inhibits TLR and TNF signaling and NFκB, PI3K, and MAPK pathways. The immunosuppressive effect of gilteritinib was less pronounced, while quizartinib did not show truncation of relevant signaling pathways. Conclusions: Our results suggest different immunosuppressive effects of these three FLT3i and may, therefore, provide an additional rationale for optimal maintenance therapy after alloHSCT of FLT3-positive AML patients to prevent infectious complications and GvHD mediated by DCs. Full article
(This article belongs to the Special Issue Advancements in Treatment Approaches for AML)
Show Figures

Figure 1

Figure 1
<p>Effect of FLT3 inhibition on DC differentiation and surface expression of costimulatory molecules. DCs generated from murine bone marrow cells were differentiated in the presence of 100 nM gilteritinib, midostaurin or quizartinib and were gated using forward and sideward scatter. Dead cells were excluded by life/dead staining. Proportion of double-positive CD11c+/CD11b+ cells is markedly reduced after differentiation in the presence of midostaurin (<b>a</b>). Human PBMCs were cultured to obtain DCs as described. Midostaurin-induced monocyte differentiation indicated by reduced CD1a and increased CD14 expression in the presence of midostaurin (<span class="html-italic">p</span> &lt; 0.0001) and gilteritinib (<span class="html-italic">p</span> &lt; 0.05) (<b>b</b>). Differentiation of murine bone marrow cells in the presence of midostaurin (M1 = 1 nM; M10 = 10 nM; M100 = 100 nM), gilteritinib (G1 = 1 nM; G10 = 10 nM; G100 = 100 nM) or quizartinib (Q1 = 1 nM; Q10 = 10 nM; Q100 = 100 nM) in different concentrations did not induce apoptosis (<b>c</b>). The mean fluorescence intensity of CD40 and CD86 on bmDCs after LPS stimulation was reduced in a dose-dependent manner by midostaurin (<span class="html-italic">p</span> &lt; 0.0001) and gilteritinib (<span class="html-italic">p</span> &lt; 0.05) exposure (<b>d</b>). MoDCs showed reduced CD80, CD83 and CD86 expression after midostaurin treatment with 100 nM. The effect was less pronounced after gilteritinib (100 nM) and only minimal after quizartinib (100 nM) treatment. MFI and rates of positive cells are depicted in the right upper corner (<b>e</b>). ns not significant, * <span class="html-italic">p</span> &lt; 0.05, **** = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>FTL3 inhibition suppresses cytokine secretion by dendritic cells. After 6 days of differentiation in the presence of FLT3i at 100 nM concentration, cytokine release of bmDCs was measured 18 h after LPS stimulation. IL-12 levels were only reduced after midostaurin treatment (<span class="html-italic">p</span> &lt; 0.01), while IL-6 (<span class="html-italic">p</span> &lt; 0.01 for midostaurin and <span class="html-italic">p</span> &lt; 0.001 for gilteritinib) and CCL-2 levels (<span class="html-italic">p</span> &lt; 0.0001 for both FLT3i) were also negatively affected by gilteritinib treatment. TNFα secretion was not affected. The generation of bmDCs is illustrated. Created with BioRender.com/j07f023 (<b>a</b>). For moDCs, midostaurin (100 nM) and gilteritinib (100 nM) treatment reduces TNFα (<span class="html-italic">p</span> &lt; 0.0001), IL-6 (<span class="html-italic">p</span> &lt; 0.01) and IL-12 levels (<span class="html-italic">p</span> &lt; 0.0001), quizartinib (100 nM) treatment did not affect TNFα levels but lowered IL-12 (<span class="html-italic">p</span> &lt; 0.01) and IL-6 (<span class="html-italic">p</span> &lt; 0.05) levels. Schematic illustration of moDC generation created with BioRender.com/u60g276 (<b>b</b>). ns not significant, * = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, *** = <span class="html-italic">p</span> &lt; 0.001, **** = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>RNA sequencing reveals inhibition of important immune response pathways by FLT3i, which is supported by protein expression analysis. BmDCs were cultured in the presence of DMSO or FLT3i. After 6 days, cells were stimulated with LPS, when indicated, and RNA was harvested 18 h later. Using hierarchical clustering, the top 20 differentially expressed genes found in RNA sequencing analysis reveal specific clustering of DMSO control samples and after exposure to midostaurin but not for other treatment groups. Expression values are depicted from low (blue) to high (red) (<b>a</b>). Concomitantly, principal component analysis (PCA) based on all analyzed gene sets also showed high similarity of midaustaurin-treated samples (<b>b</b>). Log-fold change in gene expression of midostaurin-, gilteritinib- and quizartinib-treated moDCs after LPS maturation is depicted in a heatmap. Up- (blue) and downregulation (red) are color-coded (<b>c</b>). To prove the inhibition of relevant pathways, protein was extracted from human moDCs after FLT3i treatment and LPS stimulation. Stat3 and Stat5 phosphorylation was impaired by midostaurin treatment (<b>d</b>). Inhibition of Akt phosphorylation was observed after midostaurin and quizartinib treatment (<b>e</b>). The NFκB pathway was inhibited by midostaurin and gilteritinib treatment as cRel and relB expression were reduced. Quizartinib did not affect relB or cRel expression (<b>f</b>). The uncropped bolts are shown in <a href="#app1-cancers-16-03719" class="html-app">Supplementary Materials</a>.</p>
Full article ">
Back to TopTop