[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
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

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (367)

Search Parameters:
Keywords = imatinib

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 1640 KiB  
Article
Impact of First- and Second-Generation Tyrosine Kinase Inhibitors on the Development of Graft-Versus-Host Disease in Individuals with Chronic Myeloid Leukemia: A Retrospective Analysis on Behalf of the Polish Adult Leukemia Group
by Ugo Giordano, Agnieszka Piekarska, Witold Prejzner, Lidia Gil, Jan Maciej Zaucha, Joanna Kujawska, Zuzanna Dybko, Krzysztof Dudek, Sebastian Giebel and Jarosław Dybko
Biomedicines 2025, 13(1), 163; https://doi.org/10.3390/biomedicines13010163 - 11 Jan 2025
Viewed by 448
Abstract
Background: The implementation of tyrosine kinase inhibitors (TKIs) in the treatment of chronic myeloid leukemia (CML) has brought a significant improvement in the prognosis for CML patients and a decrease in the number of patients requiring allogeneic hematopoietic stem cell transplantation (allo-HCT). [...] Read more.
Background: The implementation of tyrosine kinase inhibitors (TKIs) in the treatment of chronic myeloid leukemia (CML) has brought a significant improvement in the prognosis for CML patients and a decrease in the number of patients requiring allogeneic hematopoietic stem cell transplantation (allo-HCT). Nevertheless, the impact of TKIs on allo-HCT outcomes has not been thoroughly explored. Objectives: The main endpoint of our research was to assess the impact of prior TKI treatment on acute graft-versus-host disease (aGvHD) and chronic graft-versus-host disease (cGvHD). Methods: In our retrospective analysis, we included 240 patients treated between 1993 and 2013 and divided them into three groups according to the therapy administered prior to haploidentical, matched-related, or matched-unrelated donor allo-HCT (imatinib group n = 41, dasatinib/nilotinib group n = 28, TKI-naïve group n = 171). Results: Both the cumulative incidence of aGvHD (p = 0.044) and cGvHD (p < 0.001) in individuals receiving second-generation TKIs (2G-TKIs) prior to allo-HCT were decreased compared to patients receiving no TKIs or imatinib (IMA) (40.7% vs. 61.4% vs. 70.7%, p = 0.044; 25.0% vs. 76.4% vs. 51.2%, p < 0.001, respectively). In the case of the 2G-TKI cohort, the number of low-grade aGvHD and cGvHD was significantly lower compared to the IMA and TKI-naïve groups (p = 0.018, p = 0.004; p < 0.001 versus TKI-naïve, respectively). In terms of 3-year overall survival (OS), there were no important variations between TKI-naïve, IMA, and 2G-TKI (55% vs. 49.9% vs. 69.6%, p = 0.740). Conclusions: The results of our study suggest that TKI treatment prior to allo-HCT may have a protective impact on immune-mediated outcomes. Full article
(This article belongs to the Special Issue Pathogenesis, Diagnosis and Treatment of Hematologic Malignancies)
Show Figures

Figure 1

Figure 1
<p>Impact of 1st generation and 2nd generation TKIs on the cumulative HR of aGvHD (12 months) and cGvHD (36 months).</p>
Full article ">Figure 2
<p>Impact of 1st generation and 2nd generation TKIs on the grade of aGvHD (<b>A</b>) and cGvHD (<b>B</b>).</p>
Full article ">Figure 3
<p>Impact of 1st generation and 2nd generation TKIs on OS probability.</p>
Full article ">
14 pages, 5285 KiB  
Article
Tyrosine Kinase Inhibitor Therapy Enhances Stem Cells Profile and May Contribute to Survival of Chronic Myeloid Leukemiastem Cells
by Simone Rocco, Alessandro Maglione, Valentina Schiavo, Alessandro Ferrando, Carmen Fava, Daniela Cilloni, Barbara Pergolizzi and Cristina Panuzzo
J. Clin. Med. 2025, 14(2), 392; https://doi.org/10.3390/jcm14020392 - 10 Jan 2025
Viewed by 330
Abstract
Background/Objectives: Treatment with tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML) has revolutionized disease management and has transformed CML from a life-threatening disease to a chronic condition for many patients. However, overcoming resistance, particularly related to leukemic stem cells (LSC) that [...] Read more.
Background/Objectives: Treatment with tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML) has revolutionized disease management and has transformed CML from a life-threatening disease to a chronic condition for many patients. However, overcoming resistance, particularly related to leukemic stem cells (LSC) that can persist even when the bulk of the leukemic cells are eliminated, remains a significant challenge. Methods: K562 and KU812 cell lines were treated in vitro with the TKI Imatinib (IM). Gene expression, protein analysis, and metabolomic screening were conducted to investigate the ability of the drug to enhance stem cell (SC) features. Moreover, a gene ontology analysis was performed on different available datasets, to further consolidate our data. Results: 48 h of IM treatment can significantly increase the expression of genes related to SC self-renewal, particularly SOX2 and OCT 3/4. Interestingly, these modulations occur in cells that remain alive after drug treatment and that displayed features consistent with leukemia stem-like CML cells, suggesting that SC genes levels are crucial even in cell population survived upon TKI treatment. Moreover, after in silico analysis of available data, we observed an enrichment of SOX2/NANOG and OCT 3/4 signatures after TKI treatment, thus strengthening our results. Conclusions: Our results confirmed the relevance of LSC features after TKI treatment, highlighting the need for more effective and potentially curative strategies targeting LSCs to overcome resistance in CML. Full article
(This article belongs to the Special Issue Blood Disorders: Diagnosis, Management, and Future Opportunities)
Show Figures

Figure 1

Figure 1
<p>Imatinib treatment induces a significant increase of genes related to stemness. (<b>A</b>) K562 and KU182 cell lines were treated with different concentrations of IM (0, 0.5, 1, 2 μM) to evaluate cell proliferation rate. Not treated cells are considered 100% vitality. (<b>B</b>) Western blot analysis of BCR/ABL phosphorylation level demonstrates its decrease in a dose dependent manner, confirming the activity of TKI. (<b>C</b>,<b>D</b>) FITC-Annexin V and PI staining was performed to confirm by FACS analysis the vitality of cells after Ficoll stratification. (<b>E</b>,<b>F</b>) OCT3/4 and SOX2 gene expression were evaluated through RT-qPCR, after 48 h of IM treatment cells extracted with Ficoll. Abbreviations: NT: not treated; IM: Imatinib; Ann V, Annexin V. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 2
<p>Imatinib treatment increases protein levels of SOX2 in K562 and KU812. (<b>A</b>,<b>B</b>) The green signal corresponding to Sox2 or OCT 3/4 showed a strong nuclear increase after IM treatment in alive cells extracted with Ficoll gradient. Red propidium (PI) is used to detect nuclei (63X magnification). (<b>C</b>) ROS levels in cytosolic extract of cells treated with IM, ROS is expressed as nmol/mg of proteins. (<b>D</b>) Heatmap of the most significant expressed metabolites in the groups of alive and total cells after Imatinib treatment. Significantly reduced activities are represented in blue while those with a significantly increased activity are in red. K562 IM alive R1, 2, and 3 corresponded to viable cells condition triplicates while K562 IM tot R1, 2, and 3 represented the total cells after treatment condition triplicates. (<b>E</b>) Pathway enrichment analysis obtained with MetaboAnalyst and KEGG for pathway identification. The significance of a particular pathway was expressed as enrichment ratio. The <span class="html-italic">p</span>-value is represented with a red scale. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 3
<p>Imatinib treatment activate few SC transcription factors in cell lines and in CD34+ CML cells after in vitro treatment. (<b>A</b>) Differentially expressed genes among K562 sensitive and resistant to IM clusters enriched according to Gene Ontology terms and ordered by adjusted <span class="html-italic">p</span>-value. (<b>B</b>) Heatmap of all genes modulated by Sox2 emerged after comparison between control and Imatinib treatment K562 cells. (<b>C</b>) Differentially expressed genes after CD34+ CML cells treatment in vitro with IM clusters enriched according to Gene Ontology terms and ordered by adjusted <span class="html-italic">p</span>-value. * adjusted <span class="html-italic">p</span>-value.</p>
Full article ">
9 pages, 2893 KiB  
Case Report
The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome
by Livia Vida, Bálint Horváth, Miklós Egyed, Béla Kajtár and Hussain Alizadeh
Hematol. Rep. 2025, 17(1), 1; https://doi.org/10.3390/hematolrep17010001 - 6 Jan 2025
Viewed by 368
Abstract
Background: T-cell prolymphocytic leukemia (T-PLL) is a rare mature T-cell lymphoma that is usually associated with poor prognosis and short overall survival. Methods: We present a case of a 61-year-old woman presenting with T-PLL and the leukemic cells harboring BCR::ABL1 (BCR—breakpoint [...] Read more.
Background: T-cell prolymphocytic leukemia (T-PLL) is a rare mature T-cell lymphoma that is usually associated with poor prognosis and short overall survival. Methods: We present a case of a 61-year-old woman presenting with T-PLL and the leukemic cells harboring BCR::ABL1 (BCR—breakpoint cluster region; ABL1—ABL protooncogene 1) fusion transcripts as the result of a variant of t(9;22)(q34;q11) called Philadelphia translocation: t(9;22;18)(q34;q11;q21). Sequencing revealed a rare BCR transcript with an exon 6 breakpoint corresponding to e6a2 transcripts, which has thus far been reported in only 26 cases of leukemias. Results: After 9 months of follow-up, the disease progressed and required treatment. Following alemtuzumab and chemotherapy, a short course of imatinib therapy stabilized the disease for six months, which was followed by progression and the demise of the patient. Conclusions: To the best of our knowledge, this is the first report of a mature T-cell lymphoma with a variant Philadelphia-translocation and a very rare type of BCR::ABL1 transcript. This case highlights the importance of comprehensive genetic testing of malignancies, as abnormal molecular pathways may be uncovered that may be specifically targeted by drugs. Full article
Show Figures

Figure 1

Figure 1
<p>Morphology and immunophenotype of neoplastic cells in peripheral blood and bone marrow. (<b>A</b>) Peripheral smears revealed neoplastic lymphocytes of 9–10 µm, with irregular, often angulated nuclei and prominent nucleoli (1000× magnification). (<b>B</b>) Flow cytometry at diagnosis showed 54% lymphocytes, the majority of which demonstrated the following phenotypes: CD2<sup>+</sup>/CD3<sup>+</sup>/CD4<sup>+</sup>/CD5<sup>+</sup>/CD7<sup>+</sup>/CD8<sup>−</sup>/CD52<sup>+</sup>/CD56<sup>−</sup>. SSC indicates side scatter. Green gate: lymphocytes; orange gate: monocytes; blue gate: granulocytes. The numbers on the other scatter plots represent percentages of the quadrants. (<b>C</b>) Histology showed normocellular bone marrow with approximately 15% interstitial and vaguely nodular lymphoid infiltrate in the bone marrow (200× magnification, Naphthol AS-D chloracetate esterase staining. (<b>D</b>) Immunohistochemistry using TCL1 antibody (Abcam, Cambridge, UK, 1:100 dilution), 200× magnification.</p>
Full article ">Figure 2
<p>Cytogenetic findings. (<b>A</b>) 47,XX, del(6)(q22q23), i(8)(q10), +i(8)(q10), t(9;22;18)(q34;q11;q21), inv(14)(q11;q32), der(15)t(8;15)(q22;p13), add(16)(p13) karyotype was seen in 5 out of 30 metaphases obtained from peripheral blood (an arrow marks the Philadelphia chromosome). Four metaphases showed the same abnormalities with the addition of add(17)(q25), while one metaphase showed the addition of +der(22). (<b>B</b>) Interphase cells showed one fusion, two red, and two green signals with <span class="html-italic">BCR::ABL1</span> dual fusion probe (Abbott, Chicago, United States of America). Approximately half of the cells showed an additional fusion signal representing +der(22) (630×).</p>
Full article ">Figure 3
<p>Results of Sanger sequencing. Based on alignment with reference RNA sequences, the transcript was identified as in-frame with the breakpoint showing fusion of <span class="html-italic">BCR</span> exon 6 to <span class="html-italic">ABL1</span> exon 2.</p>
Full article ">Figure 4
<p>Summary of therapies and clinical data. ALEM: alemtuzumab; CYCLO: cyclophosphamide; IMA: imatinib; CHOP: cyclophosphamide-, doxorubicin-, vincristine-, and prednisone-containing regimen; NILO: nilotinib.</p>
Full article ">
33 pages, 3272 KiB  
Review
Caught in the Crossfire: Unmasking the Silent Renal Threats of Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia
by Maria Benkhadra, Rola Ghasoub, Reem Hajeomar, Awni Alshurafa, Nabeel Mohammad Qasem, Giuseppe Saglio, Jorge Cortes, Islam Elkonaissi, Rasha Kaddoura and Mohamed A. Yassin
Cancers 2025, 17(1), 92; https://doi.org/10.3390/cancers17010092 - 30 Dec 2024
Viewed by 587
Abstract
Background: Renal adverse drug reactions (ADRs) associated with tyrosine kinase inhibitors (TKIs) in the treatment of chronic myeloid leukemia (CML) are relatively rare, and there is currently no standardized protocol for their management. Therefore, this study aimed to summarize renal ADRs related to [...] Read more.
Background: Renal adverse drug reactions (ADRs) associated with tyrosine kinase inhibitors (TKIs) in the treatment of chronic myeloid leukemia (CML) are relatively rare, and there is currently no standardized protocol for their management. Therefore, this study aimed to summarize renal ADRs related to TKIs use in CML and propose an evidence-based approach to monitor and manage these ADRs. Methods: A systematic literature review was performed to identify renal ADRs associated with TKIs in CML. Two authors screened the search results and extracted data from 37 eligible studies. These findings were then used to develop a scheme for clinicians to monitor and manage these ADRs. Results: Overall, imatinib seemed to be significantly linked to renal adverse events compared to other TKIs, and switching to dasatinib or nilotinib significantly improved renal function. Similar events were reported with bosutinib, although they were not statistically significant. However, most of the renal events reported on dasatinib were described as nephrotic syndrome that resolved with switching to imatinib. Few cases were reported with nilotinib that described tumor lysis syndrome (TLS)-related kidney injury. Conclusions: Recommendations include monitoring for progressive decline in the estimated glomerular filtration rate with imatinib, nephrotic syndrome with dasatinib, and TLS with nilotinib. Additionally, holding the offending TKI and managing renal ADRs according to local guidelines were adopted more frequently than reducing the TKI dose. Full article
(This article belongs to the Section Systematic Review or Meta-Analysis in Cancer Research)
Show Figures

Figure 1

Figure 1
<p>Record screening process (PRISMA flow chart).</p>
Full article ">Figure 2
<p>Suggestions on the initial choice of TKI in CML therapy and related monitoring parameters based on renal risk factors.</p>
Full article ">Figure 3
<p>Suggestions on patient management following the occurrence of a TKI-related renal event.</p>
Full article ">
24 pages, 3866 KiB  
Article
Antitumor Activity and Multi-Target Mechanism of Phenolic Schiff Bases Bearing Methanesulfonamide Fragments: Cell Cycle Analysis and a Molecular Modeling Study
by Alaa A.-M. Abdel-Aziz, Adel S. El-Azab, Simone Brogi, Rezk R. Ayyad, Ibrahim A. Al-Suwaidan and Mohamed Hefnawy
Int. J. Mol. Sci. 2024, 25(24), 13621; https://doi.org/10.3390/ijms252413621 - 19 Dec 2024
Viewed by 414
Abstract
Five phenolic Schiff bases (711) incorporating a fragment of methanesulfonamide were synthesized and evaluated for their efficacy as antitumor agents. Compounds 7 and 8 demonstrated the most potent antitumor action, with a positive cytotoxic effect (PCE) of 54/59 and [...] Read more.
Five phenolic Schiff bases (711) incorporating a fragment of methanesulfonamide were synthesized and evaluated for their efficacy as antitumor agents. Compounds 7 and 8 demonstrated the most potent antitumor action, with a positive cytotoxic effect (PCE) of 54/59 and 59/59 and a mean growth percentage (MG%) of 67.3% and 19.5%, respectively, compared with imatinib (PCE = 20/59 and MG% = 92.6%). The PCE values for derivatives 911 were 3/59, 4/59, and 4/59, respectively, indicating poor antitumor effect. Compound 8 exhibited the most significant efficacy, suppressing cell proliferation by an average of 50% at a dosage of 0.501 µM, in comparison with the reference drugs sorafenib (2.33 µM), gefitinib (2.10 µM), erlotinib (7.68 µM), and celecoxib (17.5 µM). Compounds 7 and 8 had substantial inhibitory effects on the human epidermal growth factor receptor 2 (HER2), with IC50 values of 0.183 μM and 0.464 μM, respectively. Furthermore, they exhibited significant inhibition of the epidermal growth factor receptor (EGFR), with IC50 values of 0.752 μM and 0.166 μM, respectively. Compound 8 exhibited the highest COX-2 inhibition (IC50 = 12.76 μM). We performed molecular docking dynamic experiments to examine the precise interaction and structural prerequisites for the anticancer activity of derivatives 7 and 8 by targeting EGFR and HER2. Full article
(This article belongs to the Special Issue Design, Synthesis and Applications of the Task-Specific Molecules)
Show Figures

Figure 1

Figure 1
<p>The reported anticancer agents and designed phenolic Schiff bases <b>7</b>–<b>11</b>.</p>
Full article ">Figure 2
<p>Dose−response curve of compound <b>8</b>.</p>
Full article ">Figure 3
<p>Effects of compound <b>8</b> (<b>upper right panel</b>), Staurosporine (<b>middle right panel</b>), and DMSO (<b>lower right panel</b>) on the percentage of annexin V-FITC-positive staining in MCF-7 cells and cell cycle analysis of MCF-7 cells treated with compound <b>8</b> (<b>upper left panel</b>), Staurosporine (<b>middle left panel</b>), and DMSO (<b>lower left panel</b>).</p>
Full article ">Figure 4
<p>Effect of compound <b>8,</b> lapatinib, and staurosporine on <span class="html-italic">Bax</span>, <span class="html-italic">Bcl</span>−<span class="html-italic">2</span>, and <span class="html-italic">Caspase</span>−<span class="html-italic">8/</span>−<span class="html-italic">9</span> levels inside MCF−7 cells.</p>
Full article ">Figure 5
<p>Docked poses of compound <b>7</b> (magenta sticks) within the EGFR (panel (<b>A</b>), PDB ID 1M17) and HER2 (panel (<b>B</b>), PDB ID 3RCD) binding sites. The interacting residues at the binding sites are indicated by lines and are labeled. The black dotted lines represent H-bonds. The pictures were generated by PyMOL (the PyMOL Molecular Graphics System, v1.8; Schrödinger, LLC, New York, NY, USA 2015).</p>
Full article ">Figure 6
<p>Docked poses of compound <b>8</b> (gray sticks) within the EGFR (panel (<b>A</b>), PDB ID 1M17) and HER2 (panel (<b>B</b>), PDB ID 3RCD) binding sites. The interacting residues at the binding sites are indicated by lines and are labeled. The black dotted lines represent H-bonds. The pictures were generated by PyMOL (the PyMOL Molecular Graphics System, v1.8; Schrödinger, LLC, New York, 2015).</p>
Full article ">Figure 7
<p>Docked poses of compound <b>7</b> (magenta sticks, panel (<b>A</b>)) and <b>8</b> (gray sticks, panel (<b>B</b>)) within the COX-2 (PDB ID 5KIR) binding sites. The interacting residues at the binding sites are indicated by lines and are labeled. The black dotted lines represent H-bonds. The pictures were generated by PyMOL (the PyMOL Molecular Graphics System, v1.8; Schrödinger, LLC, New York, 2015).</p>
Full article ">Figure 8
<p>(<b>A</b>) RMSD analysis (ligand: red line; protein: blue line; compound <b>7</b> is identified by the in-house code <b>ss31</b>). (<b>B</b>) RMSF evaluation of the complex <b>7</b>/EGFR after a 100 ns MD simulation. Compound <b>7</b> is shown in (<b>C</b>,<b>D</b>) during the MD run. It is possible to distinguish between four types of interactions: H-bonds (green), ionic (magenta), hydrophobic (gray), and water bridges (blue). The stacked bar charts are standardized over the trajectory. For example, if the contact is maintained 70% of the time during the simulation, its value is 0.7. Values &gt; 1.0 could occur if a protein residue uses the same subtype to bind with the ligand more than once. The diagram in the figure provides a temporal explanation of the main interactions. The results show residues that interact with the ligand in each trajectory frame. A deeper orange indicates several interactions between certain residues and the ligand. The images were created using software tools from Maestro and Desmond (Maestro, Schrödinger LLC, release 2020-3).</p>
Full article ">Figure 9
<p>(<b>A</b>) RMSD analysis (ligand: red line; protein: blue line; compound <b>7</b> is identified by the in-house code <b>ss31</b>). (<b>B</b>) RMSF evaluation of the complex <b>7</b>/HER2 after a 100 ns MD simulation. Compound <b>7</b> is shown in (<b>C</b>,<b>D</b>) during the MD run. It is possible to distinguish between four types of interactions: H-bonds (green), ionic (magenta), hydrophobic (gray), and water bridges (blue). The stacked bar charts are standardized over the trajectory. For example, if the contact is maintained 70% of the time during the simulation, its value is 0.7. Values &gt; 1.0 could occur if a protein residue uses the same subtype to bind with the ligand more than once. The diagram in the figure provides a temporal explanation of the main interactions. The results show residues that interact with the ligand in each trajectory frame. A deeper orange indicates several interactions between certain residues and the ligand. The images were created using software tools from Maestro and Desmond (Maestro, Schrödinger LLC, release 2020-3).</p>
Full article ">Figure 10
<p>(<b>A</b>) RMSD analysis (ligand: red line; protein: blue line; compound <b>8</b> is identified by the in-house code <b>ss32</b>). (<b>B</b>) RMSF evaluation of the complex <b>8</b>/EGFR after a 100 ns MD simulation. Compound <b>8</b> is shown in (<b>C</b>,<b>D</b>) during the MD run. It is possible to distinguish between four types of interactions: H-bonds (green), ionic (magenta), hydrophobic (gray), and water bridges (blue). The stacked bar charts are standardized over the trajectory. For example, if the contact is maintained 70% of the time during the simulation, its value is 0.7. Values &gt; 1.0 could occur if a protein residue uses the same subtype to bind with the ligand more than once. The diagram in the figure provides a temporal explanation of the main interactions. The results show residues that interact with the ligand in each trajectory frame. A deeper orange indicates several interactions between certain residues and the ligand. The images were created using software tools from Maestro and Desmond (Maestro, Schrödinger LLC, release 2020-3).</p>
Full article ">Figure 11
<p>(<b>A</b>) RMSD analysis (ligand: red line; protein: blue line; compound <b>8</b> is identified by the in-house code <b>ss32</b>). (<b>B</b>) RMSF evaluation of the complex <b>8</b>/HER2 after a 100 ns MD simulation. Compound <b>8</b> is shown in (<b>C</b>,<b>D</b>) during the MD run. It is possible to distinguish between four types of interactions: H-bonds (green), ionic (magenta), hydrophobic (gray), and water bridges (blue). The stacked bar charts are standardized over the trajectory. For example, if the contact is maintained 70% of the time during the simulation, its value is 0.7. Values &gt; 1.0 could occur if a protein residue uses the same subtype to bind with the ligand more than once. The diagram in the figure provides a temporal explanation of the main interactions. The results show residues that interact with the ligand in each trajectory frame. A deeper orange indicates several interactions between certain residues and the ligand. The images were created using software tools from Maestro and Desmond (Maestro, Schrödinger LLC, release 2020-3).</p>
Full article ">Scheme 1
<p>Synthesis of the designed phenolic Schiff bases <b>7</b>–<b>11</b>.</p>
Full article ">
17 pages, 540 KiB  
Article
Dynamics of Chronic Myeloid Leukemia Under Imatinib Treatment: A Study of Resistance Development
by Irina Badralexi, Ana-Maria Bordei, Andrei Halanay and Ileana Rodica Rădulescu
Mathematics 2024, 12(24), 3937; https://doi.org/10.3390/math12243937 - 14 Dec 2024
Viewed by 436
Abstract
Chronic myeloid leukemia (CML) is a hematological disorder characterized by the abnormal proliferation of leukemic cells. This study aims to model the dynamics of leukemic and healthy cell populations in CML, considering the role of the immune system and the effects of treatment [...] Read more.
Chronic myeloid leukemia (CML) is a hematological disorder characterized by the abnormal proliferation of leukemic cells. This study aims to model the dynamics of leukemic and healthy cell populations in CML, considering the role of the immune system and the effects of treatment with Imatinib. The model also addresses the development of treatment resistance in cells, following the Goldie–Coldman hypothesis. We employ a system of delay differential equations to simulate the interactions between leukemic cells, healthy cells, and the immune system under treatment. The results provide insights into the dynamic balance between leukemic cells, healthy cells, and immune responses, and the impact of developing resistance on treatment outcomes. Full article
(This article belongs to the Section Mathematical Biology)
Show Figures

Figure 1

Figure 1
<p>The influence of the resistance near the point <math display="inline"><semantics> <msub> <mi>E</mi> <mn>3</mn> </msub> </semantics></math>. (<b>a</b>) The concentration of stem-like healthy cells. (<b>b</b>) The concentration of mature healthy cells. (<b>c</b>) The concentration of stem-like leukemic cells. (<b>d</b>) The concentration of mature leukemic cells. (<b>e</b>) The concentration of anti-leukemia cells and CD8+ cytotoxic T cells.</p>
Full article ">Figure 2
<p>The influence of the resistance near the point (0.0001, 0.01, 2.0527, 23.52980, 0.1742). (<b>a</b>) The concentration of stem-like healthy cells. (<b>b</b>) The concentration of mature healthy cells. (<b>c</b>) The concentration of stem-like leukemic cells. (<b>d</b>) The concentration of mature leukemic cells. (<b>e</b>) The concentration of anti-leukemia cells and CD8+ cytotoxic T cells.</p>
Full article ">Figure 3
<p>The influence of the resistance near the point (0.38, 6.21, 0.01, 0.512, 0.1). (<b>a</b>) The concentration of stem-like healthy cells. (<b>b</b>) The concentration of mature healthy cells. (<b>c</b>) The concentration of stem-like leukemic cells. (<b>d</b>) The concentration of mature leukemic cells. (<b>e</b>) The concentration of anti-leukemia cells and CD8+ cytotoxic T cells.</p>
Full article ">
10 pages, 1015 KiB  
Article
Low Renalase Levels in Newly Diagnosed CML: Dysregulation Sensitive to Modulation by Tyrosine Kinase Inhibitors
by Jelena Milenkovic, Dijana Stojanovic, Sanja Velickovic, Branka Djordjevic, Goran Marjanovic and Maja Milojkovic
Pathophysiology 2024, 31(4), 787-796; https://doi.org/10.3390/pathophysiology31040053 - 10 Dec 2024
Viewed by 777
Abstract
Background: A dysregulated proinflammatory microenvironment is considered one of the reasons why current therapies of chronic myeloid leukemia (CML) with tyrosine kinase inhibitors (TKI) do not secure disease control. Therefore, the development of BCR-ABL1-independent therapies is encouraged. Renalase (RNLS) is a multifunctional protein [...] Read more.
Background: A dysregulated proinflammatory microenvironment is considered one of the reasons why current therapies of chronic myeloid leukemia (CML) with tyrosine kinase inhibitors (TKI) do not secure disease control. Therefore, the development of BCR-ABL1-independent therapies is encouraged. Renalase (RNLS) is a multifunctional protein that exhibits both enzymatic and non-enzymatic cytokine-like properties, along with potent anti-inflammatory and anti-apoptotic effects. It is expressed in various tissues, including tumors. Methods: We investigated the levels of RNLS in the blood of CML patients in the chronic phase, treatment naïve patients, and those in remission under TKI treatment (either imatinib or nilotinib) and compared them to healthy individuals. Results: Renalase concentration was markedly decreased in treatment-naive CML patients compared to other groups (p = 0.000), while lower levels in the TKI group were not statistically significant compared to controls. The levels correlated negatively with the total leukocyte and neutrophil count (p < 0.05), while a positive correlation was present with CRP levels in treatment naïve patients. Conclusions: Dynamic regulation of RNLS expression and activity is coupled with transcription factors NF-κB and STAT3. Interpretation of our results might rely on differential requirements of activated STATs (STAT3/5) during CML clone development and maintenance, including the observation of RNLS rise upon TKI introduction. Overall, our research provides new insights into the field of hematological malignancies. Unlike other malignancies studied, RNLS plasma levels are significantly decreased in CML. In future perspectives, RNLS could potentially serve as a diagnostic, prognostic, or therapeutic option for these patients. Full article
Show Figures

Figure 1

Figure 1
<p>Comparison of the RNLS concentration between the groups. * <span class="html-italic">p</span> = 0.000 compared to the CML treatment-naïve group.</p>
Full article ">Figure 2
<p>Correlation between the RNLS concentration and leukocyte count. (<b>a</b>) The correlation of RNLS to white blood cell (WBC) count; (<b>b</b>) The correlation of RNLS to neutrophil granulocyte count.</p>
Full article ">
33 pages, 2624 KiB  
Review
Searching for Old and New Small-Molecule Protein Kinase Inhibitors as Effective Treatments in Pulmonary Hypertension—A Systematic Review
by Magdalena Jasińska-Stroschein and Paulina Glajzner
Int. J. Mol. Sci. 2024, 25(23), 12858; https://doi.org/10.3390/ijms252312858 - 29 Nov 2024
Viewed by 908
Abstract
Treatment options for pulmonary arterial hypertension (PAH) have improved substantially in the last 30 years, but there is still a need for novel molecules that can regulate the excessive accumulation of pulmonary artery smooth muscle cells (PASMCs) and consequent vascular remodeling. One set [...] Read more.
Treatment options for pulmonary arterial hypertension (PAH) have improved substantially in the last 30 years, but there is still a need for novel molecules that can regulate the excessive accumulation of pulmonary artery smooth muscle cells (PASMCs) and consequent vascular remodeling. One set of possible candidates are protein kinases. The study provides an overview of existing preclinical and clinical data regarding small-molecule protein kinase inhibitors in PAH. Online databases were searched from 2001 to 2023 according to PRISMA. The corpus included preclinical studies demonstrating alterations in at least one PH-related parameter following chronic exposure to an individual protein kinase inhibitor, as well as prospective clinical reports including healthy adults or those with PAH, with primary outcomes defined as safety or efficacy of an individual small-molecule protein kinase inhibitor. Several models in preclinical protocols (93 papers) have been proposed for studying small-molecule protein kinase inhibitors in PAH. In total, 51 kinase inhibitors were tested. Meta-analysis of preclinical results demonstrated seralutinib, sorafenib, fasudil hydrochloride, and imatinib had the most comprehensive effects on PH with anti-inflammatory, anti-oxidant, and anti-proliferative potential. Fasudil demonstrated more than 70% animal survival with the longest experimental period, while dasatinib, nintedanib, and (R)-crizotinib could deteriorate PAH. The substances targeting the same kinases often varied considerably in their activity, and such heterogeneity may be due to the variety of causes. Recent studies have addressed the molecules that affect multiple networks such as PDG-FRα/β/CSF1R/c-KIT/BMPR2 or FKBP12/mTOR. They also focus on achieving a satisfactory safety profile using innovative inhalation formulations Many small-molecule protein kinase inhibitors are able to control migration, proliferation and survival in PASMCs in preclinical observations. Standardized animal models can successfully reduce inter-study heterogeneity and thereby facilitate successful identification of candidate drugs for further evaluations. Full article
Show Figures

Figure 1

Figure 1
<p>PRISMA 2020 flowchart of the preclinical and clinical study selection process.</p>
Full article ">Figure 2
<p>Efficacy of small-molecule protein kinase inhibitors against PH-related features according to primary target (kinase family), as noted in preclinical studies. (<b>a</b>) Most therapeutic agents, except for JAK and Alk kinase inhibitors, demonstrated a reversal of RV hemodynamics with a composite end-point, including a decrease in right ventricle systolic pressure (RVSP) and mean pulmonary arterial pressure (mPAP) (<b>b</b>) and decrease in RV hypertrophy. (<b>c</b>) Pulmonary artery remodeling was non-significantly reversed following exposure to Alk kinase inhibitors only. NS—non-significant. An effect size (R) &lt; 1 (Equation (S2)) indicates a decrease in the mean value of a parameter in PH animals exposed chronically to individual agent as compared to PH subjects treated with placebo; R = 0.50 would indicate an approximately two-fold reversal in PH manifestations. The Q measure (<span class="html-italic">p</span> &lt; 0.05) indicates pronounced heterogeneity between subgroups of animals treated with agents targeting different kinases. The analyses were performed according to extracted data addressing individual parameters: the mean (+/−SD, or +/−SEM) and number of animals per group (n). Where the range of animal subjects (e.g., 8–12) was given in an individual study, the lowest number was used; where the study results did not include any data about the number of subjects, the number of subjects at randomization was considered.</p>
Full article ">Figure 2 Cont.
<p>Efficacy of small-molecule protein kinase inhibitors against PH-related features according to primary target (kinase family), as noted in preclinical studies. (<b>a</b>) Most therapeutic agents, except for JAK and Alk kinase inhibitors, demonstrated a reversal of RV hemodynamics with a composite end-point, including a decrease in right ventricle systolic pressure (RVSP) and mean pulmonary arterial pressure (mPAP) (<b>b</b>) and decrease in RV hypertrophy. (<b>c</b>) Pulmonary artery remodeling was non-significantly reversed following exposure to Alk kinase inhibitors only. NS—non-significant. An effect size (R) &lt; 1 (Equation (S2)) indicates a decrease in the mean value of a parameter in PH animals exposed chronically to individual agent as compared to PH subjects treated with placebo; R = 0.50 would indicate an approximately two-fold reversal in PH manifestations. The Q measure (<span class="html-italic">p</span> &lt; 0.05) indicates pronounced heterogeneity between subgroups of animals treated with agents targeting different kinases. The analyses were performed according to extracted data addressing individual parameters: the mean (+/−SD, or +/−SEM) and number of animals per group (n). Where the range of animal subjects (e.g., 8–12) was given in an individual study, the lowest number was used; where the study results did not include any data about the number of subjects, the number of subjects at randomization was considered.</p>
Full article ">Figure 3
<p>Schematics describing the role of particular small-molecule protein kinases and their inhibitors in PAH microenvironment, including such molecular hallmarks as inflammation, apoptosis, proliferation, or mitochondrial function. Many of these compounds have multidirectional effects. The molecular background involves anti-inflammatory activity by influencing TNF-α, MCP-1, IL-10 factors. (<b>a</b>) Molecules that reduce the expression of tyrosine kinase receptors with their main target defined as EGF, FGF, or VEGF receptors, inhibiting the proliferation of smooth muscle cells and, consequently, the process of lung fibrosis. (<b>b</b>) Rho A kinase inhibitors normalize PAH-related hemodynamic and hypertrophic lesions in the heart and the process of proliferation in the pulmonary arteries. They can beneficially activate anti-inflammatory cytokines, but decrease vasoconstrictors—endothelin ET-1, pro-inflammatory interleukin IL-17—and reduce oxidative stress. Additionally, fasudil dichloroacetate is responsible for the reduced influx of fibroblasts and inhibition of the process leading to mitochondrial dysfunction. Sirolimus, an inhibitor of the FKBP12/mTOR complex, intensifies the process of proliferation and migration of fibroblasts and yet inhibits the ongoing hypertrophic changes in the heart. Inhibition of pulmonary fibrosis is also a primary mechanism for inhibitors of the JAK/STAT signaling pathway (ruxolitinib). The molecule exerts anti-inflammatory activity (IL-1β, IL-6, IL-10), with slight effect on the PASMC remodeling. The compound IN-1233, an ALK kinase inhibitor, is distinguished by its inhibitory effect on the production of pro-inflammatory factors (TGF-β1, PAI-1). Molecules that block BCR-ABL kinase inhibit PDGF-induced proliferation and migration within smooth muscle cells of vascular walls and reduce collagen deposition in vascular walls. However, imatinib might intensify the inflammatory process (↑INF-γ, TNF-α). Similarly, dasatinib increases the formation of free radicals and does not inhibit hemodynamic and hypertrophic changes in the heart. (<b>c</b>) Seralutinib, which targets PDGF/c-KIT/BMP receptor type 2 (BMPR2) signaling, reduces smooth muscle proliferation and fibroblast migration and inhibits the process of narrowing the walls of pulmonary vessels. The molecular background involves anti-inflammatory activity by influencing TNF-α, MCP-1, IL-10 factors. Plus in a circle—positive effect of candidate drugs on the normalization of PAH-related lesions; minus in a circle—negative effect of candidate drugs that manifest the worsening of PAH-related lesions; sharp arrow—increase in activity/signaling of an individual pathway/target; blunt arrow—decrease in activity/signaling of an individual pathway/target. The untargeted pathways/factors by an individual agent are marked in gray [<a href="#B8-ijms-25-12858" class="html-bibr">8</a>,<a href="#B19-ijms-25-12858" class="html-bibr">19</a>,<a href="#B25-ijms-25-12858" class="html-bibr">25</a>,<a href="#B31-ijms-25-12858" class="html-bibr">31</a>,<a href="#B74-ijms-25-12858" class="html-bibr">74</a>,<a href="#B94-ijms-25-12858" class="html-bibr">94</a>,<a href="#B101-ijms-25-12858" class="html-bibr">101</a>,<a href="#B105-ijms-25-12858" class="html-bibr">105</a>]. ALK—activin receptor-like kinase; BMPR2—bone morphogenetic protein receptor 2; CDK—cyclin-dependent kinase; EGFR—endothelial growth factor receptor; Eph2A—erythropoietin-producing human hepatocellular type 2A receptor; ER—estrogen receptor; ErbB2/HER2—human epidermal growth factor receptor-2; ET-1—endothelin type 1; FGFR—fibroblast growth factor receptor; FLT3—FMS-like tyrosine kinase 3; HIF1a—hypoxia inducible factor 1 subunit alpha; IL-10—interleukin type 10; INF-γ—interferon gamma; LDH—dehydrogenase lactate; LP—left pneumonectomy; MAPK—mitogen-activated protein kinase; MCP-1—monocyte chemoattractant protein-1; MHC—major histocompatibility complex; mPAP—mean pulmonary artery pressure; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; NK—natural killer; NO—nitric oxide; PAH—pulmonary arterial hypertension; PAI-1—plasminogen activator inhibitor; PDGFR—platelet-derived growth factor receptor; ROS—reactive oxygen species; RV—right ventricle; RVH—right ventricle hypertrophy; RVSP—right ventricle systolic pressure; SAPK/JNK—stress-activated protein kinase/c-Jun NH(2)-terminal kinase; SMA—smooth muscle alpha-actin; TGF—transforming growth factor beta; TNF-α—tumor necrosis factor alpha; VEGFR—vascular endothelial growth factor receptor.</p>
Full article ">Figure 3 Cont.
<p>Schematics describing the role of particular small-molecule protein kinases and their inhibitors in PAH microenvironment, including such molecular hallmarks as inflammation, apoptosis, proliferation, or mitochondrial function. Many of these compounds have multidirectional effects. The molecular background involves anti-inflammatory activity by influencing TNF-α, MCP-1, IL-10 factors. (<b>a</b>) Molecules that reduce the expression of tyrosine kinase receptors with their main target defined as EGF, FGF, or VEGF receptors, inhibiting the proliferation of smooth muscle cells and, consequently, the process of lung fibrosis. (<b>b</b>) Rho A kinase inhibitors normalize PAH-related hemodynamic and hypertrophic lesions in the heart and the process of proliferation in the pulmonary arteries. They can beneficially activate anti-inflammatory cytokines, but decrease vasoconstrictors—endothelin ET-1, pro-inflammatory interleukin IL-17—and reduce oxidative stress. Additionally, fasudil dichloroacetate is responsible for the reduced influx of fibroblasts and inhibition of the process leading to mitochondrial dysfunction. Sirolimus, an inhibitor of the FKBP12/mTOR complex, intensifies the process of proliferation and migration of fibroblasts and yet inhibits the ongoing hypertrophic changes in the heart. Inhibition of pulmonary fibrosis is also a primary mechanism for inhibitors of the JAK/STAT signaling pathway (ruxolitinib). The molecule exerts anti-inflammatory activity (IL-1β, IL-6, IL-10), with slight effect on the PASMC remodeling. The compound IN-1233, an ALK kinase inhibitor, is distinguished by its inhibitory effect on the production of pro-inflammatory factors (TGF-β1, PAI-1). Molecules that block BCR-ABL kinase inhibit PDGF-induced proliferation and migration within smooth muscle cells of vascular walls and reduce collagen deposition in vascular walls. However, imatinib might intensify the inflammatory process (↑INF-γ, TNF-α). Similarly, dasatinib increases the formation of free radicals and does not inhibit hemodynamic and hypertrophic changes in the heart. (<b>c</b>) Seralutinib, which targets PDGF/c-KIT/BMP receptor type 2 (BMPR2) signaling, reduces smooth muscle proliferation and fibroblast migration and inhibits the process of narrowing the walls of pulmonary vessels. The molecular background involves anti-inflammatory activity by influencing TNF-α, MCP-1, IL-10 factors. Plus in a circle—positive effect of candidate drugs on the normalization of PAH-related lesions; minus in a circle—negative effect of candidate drugs that manifest the worsening of PAH-related lesions; sharp arrow—increase in activity/signaling of an individual pathway/target; blunt arrow—decrease in activity/signaling of an individual pathway/target. The untargeted pathways/factors by an individual agent are marked in gray [<a href="#B8-ijms-25-12858" class="html-bibr">8</a>,<a href="#B19-ijms-25-12858" class="html-bibr">19</a>,<a href="#B25-ijms-25-12858" class="html-bibr">25</a>,<a href="#B31-ijms-25-12858" class="html-bibr">31</a>,<a href="#B74-ijms-25-12858" class="html-bibr">74</a>,<a href="#B94-ijms-25-12858" class="html-bibr">94</a>,<a href="#B101-ijms-25-12858" class="html-bibr">101</a>,<a href="#B105-ijms-25-12858" class="html-bibr">105</a>]. ALK—activin receptor-like kinase; BMPR2—bone morphogenetic protein receptor 2; CDK—cyclin-dependent kinase; EGFR—endothelial growth factor receptor; Eph2A—erythropoietin-producing human hepatocellular type 2A receptor; ER—estrogen receptor; ErbB2/HER2—human epidermal growth factor receptor-2; ET-1—endothelin type 1; FGFR—fibroblast growth factor receptor; FLT3—FMS-like tyrosine kinase 3; HIF1a—hypoxia inducible factor 1 subunit alpha; IL-10—interleukin type 10; INF-γ—interferon gamma; LDH—dehydrogenase lactate; LP—left pneumonectomy; MAPK—mitogen-activated protein kinase; MCP-1—monocyte chemoattractant protein-1; MHC—major histocompatibility complex; mPAP—mean pulmonary artery pressure; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; NK—natural killer; NO—nitric oxide; PAH—pulmonary arterial hypertension; PAI-1—plasminogen activator inhibitor; PDGFR—platelet-derived growth factor receptor; ROS—reactive oxygen species; RV—right ventricle; RVH—right ventricle hypertrophy; RVSP—right ventricle systolic pressure; SAPK/JNK—stress-activated protein kinase/c-Jun NH(2)-terminal kinase; SMA—smooth muscle alpha-actin; TGF—transforming growth factor beta; TNF-α—tumor necrosis factor alpha; VEGFR—vascular endothelial growth factor receptor.</p>
Full article ">Figure 4
<p>Kaplan–Meier survival curve of the overall survival of animals with monocrotaline-induced PH treated with agents targeting different kinase families (n = 461 animals). The placebo group was exposed to monocrotaline and saline. The treatment group received BCR-ABL blocker (imatinib at 1, 10, or 50 mg/kg bw), ROCK inhibitor (fasudil hydrochloride at 30 and 100 mg/kg bw), or other agents: suramin (10 mg/kg bw), BIBF1000 (50 mg/kg bw), masitinib (50 mg/kg bw), PKI166 (50 mg/kg bw), and TGFBRII-Fc (5 mg/kg bw). All substances significantly improved animal survival as compared to placebo (Chi2 = 77.19; df = 3; <span class="html-italic">p</span> &lt; 0.0001). No differences were found between individual therapeutic subgroups (Chi2 = 4.87; df = 2; <span class="html-italic">p</span> = 0.09) [<a href="#B13-ijms-25-12858" class="html-bibr">13</a>,<a href="#B16-ijms-25-12858" class="html-bibr">16</a>,<a href="#B26-ijms-25-12858" class="html-bibr">26</a>,<a href="#B28-ijms-25-12858" class="html-bibr">28</a>,<a href="#B43-ijms-25-12858" class="html-bibr">43</a>,<a href="#B52-ijms-25-12858" class="html-bibr">52</a>,<a href="#B66-ijms-25-12858" class="html-bibr">66</a>,<a href="#B84-ijms-25-12858" class="html-bibr">84</a>,<a href="#B103-ijms-25-12858" class="html-bibr">103</a>].</p>
Full article ">
20 pages, 934 KiB  
Review
What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies
by Tomasz Stein, Paulina Cieplewicz-Guźla, Katarzyna Iżykowska, Monika Pieniawska, Ryszard Żaba, Aleksandra Dańczak-Pazdrowska and Adriana Polańska
J. Clin. Med. 2024, 13(23), 7134; https://doi.org/10.3390/jcm13237134 - 25 Nov 2024
Viewed by 901
Abstract
Morphea, also known as localized scleroderma, is an autoimmune chronic connective tissue disease. It is characterized by excessive collagen deposition in the dermis and/or subcutaneous tissue. The etiopathogenesis of this disease is not fully understood, with endothelial cell damage, immunological disorders, extracellular matrix [...] Read more.
Morphea, also known as localized scleroderma, is an autoimmune chronic connective tissue disease. It is characterized by excessive collagen deposition in the dermis and/or subcutaneous tissue. The etiopathogenesis of this disease is not fully understood, with endothelial cell damage, immunological disorders, extracellular matrix disorders and factors such as infection, trauma and other autoimmune diseases being considered. As medicine advances, there is increasing evidence that genetic factors play a significant role in disease risk and progression. In addition to environmental factors and genetic predisposition, epigenetic factors may be potential triggers for morphea. Epigenetics studies changes that affect gene expression without altering the DNA sequence, such as microRNAs, long non-coding RNAs or DNA methylation. Understanding the pathogenesis of this disease is key to identifying potential new treatments. There are anecdotal reports of good therapeutic effects following the use of biological drugs such as tocilizumab, a humanized IgG monoclonal antibody; abatacept, a recombinant soluble fusion protein; JAK inhibitors, such as tofacitinib and baricitinib; and a drug used successfully in cancer treatment, imatinib, a tyrosine kinase receptor inhibitor. In this article, we aim to review up-to-date knowledge on the pathogenesis of morphea, with particular emphasis on genetic and epigenetic factors. In addition, we present the new options of morphea treatment based on several case series treated with new drugs that are potential targets for the development of therapies for this disease. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>,<b>B</b>)—Clinical picture of morphea. (<b>A</b>)—active lesion, characteristic lilac ring, courtesy of Prof. Polańska.</p>
Full article ">Figure 2
<p>Pathogenesis of morphea. Factors such as infection and skin trauma (red arrow) can trigger inflammation in predisposed patients. T lymphocytes and eosinophils are involved in this process, appearing in the dermis and also in the vascular area. During the inflammatory phase, the endothelium is damaged, adhesion molecules appear and pro-inflammatory Th1 and Th17 cells are recruited, which activate fibroblasts via cytokines. Th2 cells then recruit T cells that produce profibrotic cytokines such as IL-6, IL-4. This leads to an increase in collagen fibres and subsequent sclerosis. The vessel walls thicken and the vessel lumen narrows. The final phase is atrophy, when the thickness of the epidermis is reduced and melanophages appear. There is a reduction in or atrophy of blood vessels and skin appendages. (The <a href="#jcm-13-07134-f002" class="html-fig">Figure 2</a> was made using Canva).</p>
Full article ">
26 pages, 19592 KiB  
Article
Integration of Machine Learning and Experimental Validation to Identify Anoikis-Related Prognostic Signature for Predicting the Breast Cancer Tumor Microenvironment and Treatment Response
by Longpeng Li, Longhui Li, Yaxin Wang, Baoai Wu, Yue Guan, Yinghua Chen and Jinfeng Zhao
Genes 2024, 15(11), 1458; https://doi.org/10.3390/genes15111458 - 12 Nov 2024
Viewed by 976
Abstract
Background/Objectives: Anoikis-related genes (ANRGs) are crucial in the invasion and metastasis of breast cancer (BC). The underlying role of ANRGs in the prognosis of breast cancer patients warrants further study. Methods: The anoikis-related prognostic signature (ANRS) was generated using a variety of machine [...] Read more.
Background/Objectives: Anoikis-related genes (ANRGs) are crucial in the invasion and metastasis of breast cancer (BC). The underlying role of ANRGs in the prognosis of breast cancer patients warrants further study. Methods: The anoikis-related prognostic signature (ANRS) was generated using a variety of machine learning methods, and the correlation between the ANRS and the tumor microenvironment (TME), drug sensitivity, and immunotherapy was investigated. Moreover, single-cell analysis and spatial transcriptome studies were conducted to investigate the expression of prognostic ANRGs across various cell types. Finally, the expression of ANRGs was verified by RT-PCR and Western blot analysis (WB), and the expression level of PLK1 in the blood was measured by the enzyme-linked immunosorbent assay (ELISA). Results: The ANRS, consisting of five ANRGs, was established. BC patients within the high-ANRS group exhibited poorer prognoses, characterized by elevated levels of immune suppression and stromal scores. The low-ANRS group had a better response to chemotherapy and immunotherapy. Single-cell analysis and spatial transcriptomics revealed variations in ANRGs across cells. The results of RT-PCR and WB were consistent with the differential expression analyses from databases. NU.1025 and imatinib were identified as potential inhibitors for SPIB and PLK1, respectively. Additionally, findings from ELISA demonstrated increased expression levels of PLK1 in the blood of BC patients. Conclusions: The ANRS can act as an independent prognostic indicator for BC patients, providing significant guidance for the implementation of chemotherapy and immunotherapy in these patients. Additionally, PLK1 has emerged as a potential blood-based diagnostic marker for breast cancer patients. Full article
(This article belongs to the Section Bioinformatics)
Show Figures

Figure 1

Figure 1
<p>Screening of the prognostic ANRGs. (<b>A</b>) Venn diagram showing the intersection of DEGs with ANRGs. (<b>B</b>) Univariate Cox analysis of ANRGs. (<b>C</b>) Screening of the prognostic ANRGs by the Enet algorithm. (<b>D</b>) Plot of ten-fold cross-validations. (<b>E</b>) Plot of LASSO coefficient. Screening of the prognostic ANRGs by random forest algorithm (<b>F</b>) and Coxboost (<b>G</b>). (<b>H</b>) Top 10 most significant genes screened by the xgboost algorithm. (<b>I</b>) Top 10 most significant genes screened by the GBM algorithm. (<b>J</b>) Venn plot shows the intersected prognostic ANRGs identified by six machine learning algorithms for survival. (<b>K</b>) Multivariate Cox analysis of the prognostic ANRGs. * <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.</p>
Full article ">Figure 2
<p>Construction of the ANRS. (<b>A</b>–<b>D</b>) Scatter plots of ANRS and survival status in TCGA-BRCA, METABRIC, GSE96058, and GSE86166. (<b>E</b>–<b>H</b>) Survival curves of high- and low-ANRS groups in TCGA-BRCA, METABRIC, GSE96058, and GSE86166. (<b>I</b>–<b>L</b>) ROC curves for predicting 1, 3, and 5-year survival for the ANRS in TCGA-BRCA, METABRIC, GSE96058, and GSE86166.</p>
Full article ">Figure 3
<p>Relationship between ANRS and clinical characteristics. Box plots demonstrate the differences in ANRS across Status (<b>A</b>), Age (<b>B</b>), Stage (<b>C</b>), T stage (<b>D</b>), and N stage (<b>E</b>) in the TCGA-BRCA cohort. Survival curves for the TCGA-BRCA cohort for age &lt;60 (<b>F</b>), age ≥60 (<b>G</b>), stage I–II (<b>H</b>), stage III–IV (<b>I</b>), T1–T2 (<b>J</b>), T3–T4 (<b>K</b>), N0–N1 (<b>L</b>), and N2–N3 (<b>M</b>). * <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 4
<p>Nomogram construction. (<b>A</b>–<b>D</b>) Univariate and multivariate Cox regression analyses were performed to evaluate the independence of the ANRS. (<b>E</b>) A nomogram was developed based on the ANRS and clinical characteristics. (<b>F</b>) ROC curves were plotted to assess the performance of the nomogram. (<b>G</b>) Calibration curves were generated to compare the predicted survival probabilities with the actual outcomes. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Differences in immune cell infiltration between high- and low-ANRS groups. (<b>A</b>) A heatmap illustrating the variation in immune cell infiltration levels calculated using five algorithms between the different ANRS groups. (<b>B</b>) Comparison of tumor microenvironmental scores (Immune, Stromal, and ESTIMATE scores) between the high- and low-ANRS groups. (<b>C</b>) Differential expression of immune checkpoints between the high- and low-ANRS groups. (<b>D</b>) Correlation analysis between the prognostic ANRGs and immune checkpoints. * <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 6
<p>Immunotherapy and drug sensitivity. (<b>A</b>) Differences in immunophenotype score (IPS) between different ANRS groups. IPS predicts patient response to anti-PD-1/PD-L1 or anti-CTLA4 therapy. Analysis of sensitivity differences (IC50) of chemotherapeutic drugs between high- and low-ANRS groups. (<b>B</b>–<b>F</b>) cisplatin, epirubicin, 5-Fluorouracil, vinorelbine, gemcitabine. ** <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 7
<p>Single-cell analysis of prognostic ANRGs. (<b>A</b>) Different cell types within the EMTAB8107 dataset. Expression levels of PLK1 (<b>B</b>), SPIB (<b>C</b>), CD24 (<b>D</b>), EDA2R (<b>E</b>), and NTRK3 (<b>F</b>) in different cells.</p>
Full article ">Figure 8
<p>Spatial transcriptomic analysis of prognostic ANRGs. (<b>A</b>) Distribution of different cells in GSE203612-GSM6177603-NYU-BRCA2. (<b>B</b>–<b>F</b>) Distribution of PLK1, CD24, SPIB, EDA2R, and NTRK3 in different cells.</p>
Full article ">Figure 9
<p>Prediction of potential drugs. (<b>A</b>–<b>C</b>) CMap analysis reveals the top three potential drugs targeting SPIB, PLK1, and CD24. (<b>D</b>,<b>E</b>) Diagrams of the molecular docking models of drugs with proteins, their active sites, and binding distances.</p>
Full article ">Figure 10
<p>IHC images of the prognostic ANRGs. Protein staining levels of CD24 (<b>A</b>), EDA2R (<b>B</b>), NTRK3 (<b>C</b>), PLK1 (<b>D</b>), and SPIB (<b>E</b>) in normal and BC tissues.</p>
Full article ">Figure 11
<p>Experimental validation of the prognostic ANRGs. (<b>A</b>–<b>E</b>) The mRNA expression levels of the prognostic ANRGs in breast cancer tissues and paracancerous tissues. (<b>F</b>) Protein expression levels of the prognostic ANRGs in breast cancer tissues and paracancerous tissues. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 12
<p>Comprehensive analysis of PLK1. (<b>A</b>) Error rate plots and variable significance plots of the random forest algorithm. Differential expression of PLK1 in different status (<b>B</b>), age (<b>C</b>), stage (<b>D</b>), T-stage (<b>E</b>), N-stage (<b>F</b>), and M-stage (<b>G</b>). (<b>H</b>–<b>K</b>) Diagnostic ROC for PLK1 in the TCGA-BRCA, GSE45827, GSE57297, and GSE24124 cohorts. (<b>L</b>) Expression levels of PLK1 in the serum of normal subjects and breast cancer patients. * <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; 0.0001.</p>
Full article ">
23 pages, 7695 KiB  
Article
Rational Approach to New Chemical Entities with Antiproliferative Activity on Ab1 Tyrosine Kinase Encoded by the BCR-ABL Gene: An Hierarchical Biochemoinformatics Analysis
by Vitor H. da S. Sanches, Cleison C. Lobato, Luciane B. Silva, Igor V. F. dos Santos, Elcimar de S. Barros, Alexandre de A. Maciel, Elenilze F. B. Ferreira, Kauê S. da Costa, José M. Espejo-Román, Joaquín M. C. Rosa, Njogu M. Kimani and Cleydson B. R. Santos
Pharmaceuticals 2024, 17(11), 1491; https://doi.org/10.3390/ph17111491 - 6 Nov 2024
Viewed by 847
Abstract
Background: This study began with a search in three databases, totaling six libraries (ChemBridge-DIVERSet, ChemBridge-DIVERSet-EXP, Zinc_Drug Database, Zinc_Natural_Stock, Zinc_FDA_BindingDB, Maybridge) with approximately 2.5 million compounds with the aim of selecting potential inhibitors with antiproliferative activity on the chimeric tyrosine kinase encoded by the [...] Read more.
Background: This study began with a search in three databases, totaling six libraries (ChemBridge-DIVERSet, ChemBridge-DIVERSet-EXP, Zinc_Drug Database, Zinc_Natural_Stock, Zinc_FDA_BindingDB, Maybridge) with approximately 2.5 million compounds with the aim of selecting potential inhibitors with antiproliferative activity on the chimeric tyrosine kinase encoded by the BCR-ABL gene. Methods: Through hierarchical biochemoinformatics, ADME/Tox analyses, biological activity prediction, molecular docking simulations, synthetic accessibility and theoretical synthetic routes of promising compounds and their lipophilicity and water solubility were realized. Results: Predictions of toxicological and pharmacokinetic properties (ADME/Tox) using the top100/base (600 structures), in comparison with the commercial drug imatinib, showed that only nine exhibited the desired properties. In the prediction of biological activity, the results of the nine selected structures ranged from 13.7% < Pa < 65.8%, showing them to be potential protein kinase inhibitors. In the molecular docking simulations, the promising molecules LMQC01 and LMQC04 showed significant values in molecular targeting (PDB 1IEP—resolution 2.10 Å). LMQC04 presented better binding affinity (∆G = −12.2 kcal mol−1 with a variation of ±3.6 kcal mol−1) in relation to LMQC01. The LMQC01 and LMQC04 molecules were advanced for molecular dynamics (MD) simulation followed by Molecular Mechanics with generalized Born and Surface Area solvation (MM-GBSA); the comparable, low and stable RMSD and ΔE values for the protein and ligand in each complex suggest that the selected compounds form a stable complex with the Abl kinase domain. This stability is a positive indicator that LMQC01 and LMQC04 can potentially inhibit enzyme function. Synthetic accessibility (SA) analysis performed on the AMBIT and SwissADME webservers showed that LMQC01 and LMQC04 can be considered easy to synthesize. Our in silico results show that these molecules could be potent protein kinase inhibitors with potential antiproliferative activity on tyrosine kinase encoded by the BCR-ABL gene. Conclusions: In conclusion, the results suggest that these ligands, particularly LMQC04, may bind strongly to the studied target and may have appropriate ADME/Tox properties in experimental studies. Considering future in vitro or in vivo assays, we elaborated the theoretical synthetic routes of the promising compounds identified in the present study. Based on our in silico findings, the selected ligands show promise for future studies in developing chronic myeloid leukemia treatments. Full article
(This article belongs to the Special Issue Chemoinformatics and Drug Design, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>General scheme summarizing the methodological steps.</p>
Full article ">Figure 2
<p>Superpositions of the ligand with crystallographic pose (in red) with the calculated poses (in green)—Abl Kinase Domain (organism <span class="html-italic">Mus musculus</span>, PDB ID 1IEP), showing an RMSD value equal to 0.4721 Å.</p>
Full article ">Figure 3
<p>Interactions of imatinib with key amino acid residues in the active site of the Ab1 kinase domain.</p>
Full article ">Figure 4
<p>Predicted interactions between the BCR-ABL tyrosine kinase active site and compound LMQC01.</p>
Full article ">Figure 5
<p>Predicted interactions between the active site of BCR-ABL tyrosine kinase and the compound LMQC04.</p>
Full article ">Figure 6
<p>RMSD alignment analysis among apo-protein and ligand-complexes for C-Abl kinase domain (PDB ID: 1IEP) (<b>a</b>) based on 300 ns MD analysis. MMGBSA_∆G_Binding value line chart for 300 ns MD simulation (<b>b</b>).</p>
Full article ">Figure 7
<p>Result of 300 ns MD analysis for imatinib binding to the Abl-kinase domain. The protein–ligand RMSD plot of imatinib bound to the Abl-kinase domain (<b>a</b>) (PDB ID: 1IEP). Ligand–protein contact interactions scheme with the protein residues of imatinib bound to Abl-kinase (<b>b</b>). Protein–ligand contacts histogram of the interaction fraction of H-bond (green), hydrophobic bond (purple), ionic bond (magenta), and water bridges (blue) for imatinib (<b>c</b>). RMSF plot of imatinib (<b>d</b>) protein–ligand complex.</p>
Full article ">Figure 8
<p>Result of 300 ns MD analysis for LMQC01 and LMQC04 binding to the Abl-kinase domain. The protein–ligand RMSD plot of LMQC01 (<b>a</b>) and LMQC04 (<b>b</b>) bound to the Abl-kinase domain (PDB ID: 1IEP). Ligand–protein contact interactions scheme with the protein residues of LMQC01 (<b>c</b>) and LMQC04 (<b>d</b>) bound to Abl-kinase. Protein–ligand contacts histogram of the interaction fraction of H-bond (green), hydrophobic bond (purple), ionic bond (magenta), water bridges (blue), and halogen bonds (orange) for LMQC01 (<b>e</b>) and LMQC04 (<b>f</b>). RMSF plot of LMQC01 (<b>g</b>) and LMQC04 (<b>h</b>) protein–ligand complex.</p>
Full article ">Figure 9
<p>Graphical representation of the molecular overlay analysis between molecules (<b>a</b>) LMQC01 (yellow) and (<b>b</b>) LMQC04 (blue) with the reference molecule (imatinib—green).</p>
Full article ">Figure 10
<p>Synthetic route of the compound <b>LMQC 01</b>. Starting materials <b>I</b> and <b>II</b> are commercially available.</p>
Full article ">Figure 11
<p>Alternative synthetic route of the compound <b>LMQC 01</b>. 4 Å MS (molecular sieves). Starting materials <b>I</b>, <b>IV</b> and <b>V</b> are commercially available.</p>
Full article ">Figure 12
<p>Synthetic route of the compound <b>LMQC 04</b>. Starting materials <b>XIII, XV</b> and <b>XVII</b> are commercially available.</p>
Full article ">
12 pages, 3752 KiB  
Article
Vitamin K2 Protects Against SARS-CoV-2 Envelope Protein-Induced Cytotoxicity in Chronic Myeloid Leukemia Cells and Enhances Imatinib Activity
by Seiichi Okabe, Yuya Arai and Akihiko Gotoh
Int. J. Mol. Sci. 2024, 25(21), 11800; https://doi.org/10.3390/ijms252111800 - 2 Nov 2024
Viewed by 1045
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm characterized by excessive proliferation of myeloid cells. The COVID-19 pandemic has raised concerns regarding the impact of SARS-CoV-2 on patients with malignancies, particularly those with CML. This study aimed to investigate the effects of SARS-CoV-2 [...] Read more.
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm characterized by excessive proliferation of myeloid cells. The COVID-19 pandemic has raised concerns regarding the impact of SARS-CoV-2 on patients with malignancies, particularly those with CML. This study aimed to investigate the effects of SARS-CoV-2 proteins on CML cell viability and the protective role of vitamin K2 (VK2) in conjunction with imatinib. Experiments conducted on K562 CML cells demonstrated that the SARS-CoV-2 envelope protein induces cytotoxicity and activates caspase 3/7, which are key markers of apoptosis. VK2 mitigated these cytotoxic effects and decreased cytokine production while inhibiting colony formation. Furthermore, the combination of VK2 with imatinib significantly reduced cellular proliferation, diminished mitochondrial membrane potential, and markedly suppressed colony formation. These findings suggest that VK2 protects CML cells from SARS-CoV-2-induced cytotoxicity and enhances the therapeutic efficacy of imatinib, presenting a potential strategy to improve CML treatment during the COVID-19 pandemic. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Expression of COVID-19-related genes in chronic myeloid leukemia (CML) cells. (<b>A</b>) Volcano plot analysis of datasets retrieved from a public database (GSE100026). (<b>B</b>) Dendrogram analysis of datasets retrieved from a public database (GSE100026). (<b>C</b>) Gene Ontology (GO) analysis of biological processes and pathway analysis conducted on RNA-Seq data. (<b>D</b>) Expression of inflammation-related genes validated using data from the GEO database (GSE227341). ns, not significant.</p>
Full article ">Figure 2
<p>Cytotoxic and apoptotic effects of the SARS-CoV-2 envelope protein on CML cells. (<b>A</b>) CML cell lines were cultured with the indicated concentrations of EP or S1 protein for 24 h. Cell growth was evaluated using the Cell Counting Kit-8. (<b>B</b>) CML cell lines were cultured in RPMI 1640 medium with 10% fetal calf serum and the indicated concentrations of EP or S1 protein for 24 h. Cytotoxicity was evaluated using the Cytotoxicity LDH Assay Kit. (<b>C</b>) CML cell lines were treated with the indicated concentrations of EP for 24 h. Caspase 3/7 activity was measured using the Caspase Glo 3/7 Assay Kit. * <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 compared to the control. (<b>D</b>,<b>E</b>) Time-dependent effects of EP on CML cell lines. Cell growth (<b>D</b>) and cytotoxicity (<b>E</b>) were evaluated using the Cell Counting Kit-8. ns, not significant.</p>
Full article ">Figure 3
<p>Protective effects of VK2 on CML cells treated with SARS-CoV-2 EP. (<b>A</b>–<b>D</b>) CML cell lines were cultured with the indicated concentrations of EP and/or VK2 for 24 h. Cell growth (<b>A</b>), caspase 3/7 activity (<b>B</b>), cytotoxicity (<b>C</b>), and apoptosis (<b>D</b>) were evaluated. ** <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 compared to EP-treated cells. ns, not significant. (<b>E</b>) Expression levels of inflammatory-related genes in cells treated with EP and/or VK2 for 24 h. Gene expression was validated using RT-PCR. *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 compared to the control. ns, not significant.</p>
Full article ">Figure 4
<p>VK2 suppresses CML cell proliferation and colony formation. (<b>A</b>,<b>B</b>) CML cell lines were incubated with VK2 for 72 h. Cell growth (<b>A</b>) and cytotoxicity (<b>B</b>) were evaluated. * <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; 0.0001 compared to EP-treated cells. ns, not significant. (<b>C</b>) CML cells were treated with the indicated concentrations of VK2 for 7 days. Colonies were photographed with a digital camera and counted using an EVOS™ FL Digital Inverted Fluorescence Microscope (Thermo Fisher Scientific Inc., Waltham, MA, USA). The quantitative graph shows the number of colonies, and representative images are displayed. Scale bar: 1000 μm. **** <span class="html-italic">p</span> &lt; 0.0001 compared to the control. ns, not significant.</p>
Full article ">Figure 5
<p>Synergistic effects of imatinib and VK2 in CML cells. (<b>A</b>–<b>C</b>) CML cell lines were cultured with 1 µM imatinib and/or 10 µM VK2 for 48 h or 72 h. Cell growth (<b>A</b>), caspase 3/7 activity (<b>B</b>), and cytotoxicity (<b>C</b>) were measured. ** <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 compared to the control. ns, not significant. (<b>D</b>) Mitochondrial membrane potential (MMP) was analyzed in CML cell lines treated with 10 nM asciminib and/or 10 µM VK2 for 48 h using a Mitochondria Staining Kit. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 compared to the control. ns, not significant. (<b>E</b>,<b>F</b>) Colony formation in CML cells that were treated with VK2 for 7 to 9 days. Colonies were counted (<b>E</b>,<b>F</b>) photographed using a digital camera and an EVOS™ FL Digital Inverted Fluorescence Microscope. Scale bar: 1000 μm. **** <span class="html-italic">p</span> &lt; 0.0001 compared to the control.</p>
Full article ">
11 pages, 564 KiB  
Review
Bone Mineral Density, C-Terminal Telopeptide of Type I Collagen, and Osteocalcin as Monitoring Parameters of Bone Remodeling in CML Patients Undergoing Imatinib Therapy: A Basic Science and Clinical Review
by Nurita Indarwulan, Merlyna Savitri, Ami Ashariati, Siprianus Ugroseno Yudho Bintoro, Muhammad Noor Diansyah, Putu Niken Ayu Amrita and Pradana Zaky Romadhon
Diseases 2024, 12(11), 275; https://doi.org/10.3390/diseases12110275 - 2 Nov 2024
Viewed by 1085
Abstract
Background: Chronic myeloid leukemia (CML) is one of the most commonly found types of myeloproliferative neoplasms, characterized by increased proliferation of granulocytic cells without losing their differentiation ability. Imatinib, a tyrosine kinase inhibitor (TKI), can be effectively used as therapy for CML. However, [...] Read more.
Background: Chronic myeloid leukemia (CML) is one of the most commonly found types of myeloproliferative neoplasms, characterized by increased proliferation of granulocytic cells without losing their differentiation ability. Imatinib, a tyrosine kinase inhibitor (TKI), can be effectively used as therapy for CML. However, Imatinib can affect bone turnover thus having clinical implications on the bones of CML patients undergoing long-term Imatinib therapy. However, parameters that can accurately describe the bone condition in CML patients receiving Imatinib still need further study. A combination of imaging techniques such as bone mineral density (BMD) and bone turnover activity markers such as C-terminal telopeptide of type I collagen (CTX-1) and osteocalcin has the potential to be used as monitoring parameters for bone density abnormalities in CML patients receiving Imatinib. Objectives: This article explains the rationale for using BMD, CTX-1, and osteocalcin as monitoring parameters of bone remodeling in CML patients receiving Imatinib. Results: First, the physiological process of bone turnover will be explained. Then, we describe the role of tyrosine kinase in bone metabolism. Next, the impact of Imatinib on BMD, CTX-1, and osteocalcin will be explained. Conclusion: The assessment of bone health of CML patients on Imatinib should include both BMD tests and bone turnover marker assays such as CTX-1 and osteocalcin. Full article
Show Figures

Figure 1

Figure 1
<p>Interaction of CTX-1, osteocalcin, and bone mineral density. Osteoclasts produce CTX-1, which is a marker of bone resorption. Osteoblasts produce osteocalcin, which is a marker of bone formation. When the bone formation rate surpasses the bone resorption rate, bone mineral density (BMD) increases. On the other hand, when the bone resorption rate surpasses the bone formation rate, BMD decreases. CTX-1: C-terminal telopeptide of type I collagen. Created in BioRender.</p>
Full article ">
12 pages, 2811 KiB  
Article
Targeting Oxidative Phosphorylation with a Novel Thiophene Carboxamide Increases the Efficacy of Imatinib against Leukemic Stem Cells in Chronic Myeloid Leukemia
by Kana Kusaba, Tatsuro Watanabe, Keisuke Kidoguchi, Yuta Yamamoto, Ayaka Tomoda, Toshimi Hoshiko, Naoto Kojima, Susumu Nakata and Shinya Kimura
Int. J. Mol. Sci. 2024, 25(20), 11093; https://doi.org/10.3390/ijms252011093 - 15 Oct 2024
Viewed by 1157
Abstract
Patients with chronic myeloid leukemia (CML) respond to tyrosine kinase inhibitors (TKIs); however, CML leukemic stem cells (LSCs) exhibit BCR::ABL kinase-independent growth and are insensitive to TKIs, leading to disease relapse. To prevent this, new therapies targeting CML-LSCs are needed. Rates of mitochondria-mediated [...] Read more.
Patients with chronic myeloid leukemia (CML) respond to tyrosine kinase inhibitors (TKIs); however, CML leukemic stem cells (LSCs) exhibit BCR::ABL kinase-independent growth and are insensitive to TKIs, leading to disease relapse. To prevent this, new therapies targeting CML-LSCs are needed. Rates of mitochondria-mediated oxidative phosphorylation (OXPHOS) in CD34+CML cells within the primitive CML cell population are higher than those in normal undifferentiated hematopoietic cells; therefore, the inhibition of OXPHOS in CML-LSCs may be a potential cure for CML. NK-128 (C33H61NO5S) is a structurally simplified analog of JCI-20679, the design of which was based on annonaceous acetogenins. NK-128 exhibits antitumor activity against glioblastoma and human colon cancer cells by inhibiting OXPHOS and activating AMP-activated protein kinase (AMPK). Here, we demonstrate that NK-128 effectively suppresses the growth of CML cell lines and that the combination of imatinib and NK-128 is more potent than either alone in a CML xenograft mouse model. We also found that NK-128 inhibits colony formation by CD34+ CML cells isolated from the bone marrow of untreated CML patients. Taken together, these findings suggest that targeting OXPHOS is a beneficial approach to eliminating CML-LSCs, and may improve the treatment of CML. Full article
(This article belongs to the Collection Anticancer Drug Discovery and Development)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of thiophene carboxamide (NK-128).</p>
Full article ">Figure 2
<p>NK-128 inhibits the proliferation of CML and Philadelphia chromosome-positive acute lymphoblastic leukemia cell lines. (<b>A</b>) Cells were incubated with NK-128 and cell numbers were counted on Days 4 and 8. The number of viable cells is shown. NK-128 inhibited the proliferation of BV173 (<span class="html-italic">p</span> &lt; 0.01) and SUP-B15 (<span class="html-italic">p</span> &lt; 0.05) cells significantly, even at concentrations as low as 0.1 μM. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). (<b>B</b>) Experimental scheme showing the generation of the K562 xenograft model using NOD/Shi-scid IL-2Rγ KO Jic mice. On Day 0, K562 cells (4.0 × 10<sup>6</sup> cells/mouse) were injected subcutaneously into NOD/Shi-scid IL-2Rγ KO Jic mice. Daily ip administration of the vehicle and NK-128 began on Day 8. (<b>C</b>) Tumor growth curves and (<b>D</b>) body weight for each group; vehicle (black; n = 10), 10 mg/kg NK-128 (blue; n = 10), and 20 mg/kg NK-128 (red; n = 10).</p>
Full article ">Figure 3
<p>Combination therapy with imatinib and NK-128 inhibits the proliferation of CML cell lines and Philadelphia chromosome-positive acute lymphoblastic leukemia cell lines to a greater extent than imatinib alone. (<b>A</b>) CML and Ph<sup>+</sup>ALL cell lines were treated with medium (black), imatinib (blue) or a combination of imatinib and 0.1 μM NK-128 (red). The concentration of imatinib was 0.25 μM for K562, 0.2 μM for MYL, 0.1 μM for BV173, 0.4 μM for SUP-B15, and 1 μM for MYL-R. The combination of NK-128 plus imatinib also inhibited the proliferation to a greater extent than imatinib alone. (** <span class="html-italic">p</span> &lt; 0.01) (<b>B</b>) Each cell line was treated with or without NK-128 and imatinib (IM) for 3 days. The number of cells stained with APC-annexin V was measured by flow cytometric analysis, as described in the <a href="#sec4-ijms-25-11093" class="html-sec">Section 4</a>. Results are the mean of three independent experiments with SD (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>ATP and lactate production by K562, BV173, MYL, and SUP-B15 cells. Oligomycin inhibits OXPHOS, and 2-DG inhibits glycolysis. ATP and lactate production levels were determined in K562 and MYL cell lines, which are less sensitive to NK-128 (<b>A</b>) or in the sensitive cell lines, BV173 and SUP-B15 (<b>B</b>), as described in the <a href="#sec4-ijms-25-11093" class="html-sec">Section 4</a>. The main metabolic pathways of K562 and MYL are glycolysis-dependent. Conversely, those of BV173 and SUP-B15 are OXPHOS-dependent. NK-128 inhibits the production of ATP in BV173 and SUP-B15 cells, which are mainly OXPHOS dependent.</p>
Full article ">Figure 5
<p>Combined treatment with NK-128 and TKI effectively suppresses tumor growth in xenograft model mice. (<b>A</b>) Experimental scheme showing the generation of the K562 xenograft model using NOD/Shi-scid IL-2Rγ KO Jic mice. On Day 0, K562 cells (5.0 × 10<sup>6</sup> cells/mouse) were injected subcutaneously into NOD/Shi-scid IL-2Rγ KO Jic mice. Vehicle, imatinib, and NK-128 were administered on Day 6 and continued daily for a total of 11 days (nothing was administered on Day 12). (<b>B</b>) The number of subjects in each group was determined by the following formula. (<b>B</b>) Tumor growth curves for each group. Vehicle (black; n = 10), imatinib (blue; n = 10), NK-128 (green; n = 10), and NK-128 + imatinib (red; n = 10). NK-128 + imatinib inhibited tumor growth significantly (versus vehicle: * <span class="html-italic">p</span> = 0.03). (<b>C</b>) Xenograft tumors were isolated on day 17 in each treatment group. They were arranged in descending order from the right side. (<b>D</b>) The weight of the isolated tumors was measured. NK-128 + imatinib inhibited xenograft tumor growth to a greater extent than vehicle (* <span class="html-italic">p</span> = 0.04) Each circle shows the sample value and the cross shows the median. (<b>E</b>) Bodyweight was measured from day 6 to day 17. (<b>F</b>) White blood cell count, (<b>G</b>) red blood cell count, (<b>H</b>) hemoglobin levels, and (<b>I</b>) platelet count after treatment. Differences between the vehicle and each treatment group were tested using Dunnett’s test. (* <span class="html-italic">p</span> &lt; 0.05. n.s. not significant).</p>
Full article ">Figure 6
<p>Colony formation by CML CD34+ cells in the presence of different concentrations of imatinib and NK-128. NK-128 monotherapy inhibited colony formation in a concentration-dependent manner (compared with no treatment). NK-128 (500 nM) combined with imatinib (200 nM) inhibited colony formation by CML CD34+ cells significantly compared with imatinib alone. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
11 pages, 1307 KiB  
Case Report
Blast Transformation of Chronic Myeloid Leukemia Driven by Acquisition of t(8;21)(q22;q22)/RUNX1::RUNX1T1: Selecting Optimal Treatment Based on Clinical and Molecular Findings
by Adolfo Fernández-Sánchez, Alberto Hernández-Sánchez, Cristina De Ramón, María-Carmen Chillón, María Belén Vidriales, Mónica Baile-González, Cristina-Teresa Fuentes-Morales, Magdalena Sierra-Pacho, Lucía López-Corral and Fermín Sánchez-Guijo
Biomedicines 2024, 12(10), 2339; https://doi.org/10.3390/biomedicines12102339 - 15 Oct 2024
Viewed by 1241
Abstract
The advent of tyrosine kinase inhibitors (TKIs) has changed the natural history of chronic myeloid leukemia (CML), and the transformation from the chronic phase to the blast phase (BP) is currently an uncommon situation. However, it is one of the major remaining challenges [...] Read more.
The advent of tyrosine kinase inhibitors (TKIs) has changed the natural history of chronic myeloid leukemia (CML), and the transformation from the chronic phase to the blast phase (BP) is currently an uncommon situation. However, it is one of the major remaining challenges in the management of this disease, as it is associated with dismal outcomes. We report the case of a 63-year-old woman with a history of CML with poor response to imatinib who progressed to myeloid BP-CML, driven by the acquisition of t(8;21)(q22;q22)/RUNX1::RUNX1T1. The patient received intensive chemotherapy and dasatinib, followed by allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, she suffered an early relapse after allo-HSCT with the acquisition of the T315I mutation in ABL1. Ponatinib and azacitidine were started as salvage treatment, allowing for the achievement of complete remission with deep molecular response after five cycles. Advances in the knowledge of disease biology and clonal evolution are crucial for optimal treatment selection, which ultimately translates into better patient outcomes. Full article
(This article belongs to the Special Issue Advances in the Pathogenesis and Treatment of Acute Myeloid Leukemia)
Show Figures

Figure 1

Figure 1
<p>Cytogenetic studies of the patient at myeloid BP-CML, showing the acquisition of t(8;21)(q22;q22)/<span class="html-italic">RUNX1::RUNX1T1</span> as the driver event of BP. (<b>a</b>): <span class="html-italic">BCR::ABL1</span> fusion probe (Dual Color, Dual Fusion, Vysis LSI) in the FISH study, showing two fusions in most of the cells, representative of the presence of t(9;22)(q34.1;q11.2)/<span class="html-italic">BCR::ABL1</span> (<b>b</b>): <span class="html-italic">RUNX1::RUNX1T1</span> fusion probe (Dual Color, Dual Fusion, Vysis LSI) in the FISH study, showing two fusions in approximately half of the cells, representative of the presence of t(8;21)(q22;q22)/<span class="html-italic">RUNX1::RUNX1T1</span> (<b>c</b>): A Circos plot of optical genome mapping (Bionano) showing concomitant t(8;21)(q22;q22)/<span class="html-italic">RUNX1::RUNX1T1</span> and t(9;22)(q34.1;q11.2)/<span class="html-italic">BCR::ABL1</span> in the patient sample.</p>
Full article ">Figure 2
<p>Polymerase chain reaction and Sanger sequencing analysis of T315I mutation in <span class="html-italic">BCR::ABL1</span> fusion gene (c.944C &gt; T, p.Thr315Ile).</p>
Full article ">Figure 3
<p>A summary of the clinical case report. The temporal evolution of the <span class="html-italic">BCR::ABL1/ABL1</span> ratio according to the International Scale (represented in blue) and the number of <span class="html-italic">RUNX1::RUNX1T1</span> transcripts (represented in red) in peripheral blood samples are shown, together with the main events that the patient presented during the disease evolution. CML: chronic myeloid leukemia; BP: blast phase; FLAG-IDA: fludarabine, cytarabine, idarubicin and granulocyte colony-stimulating factor; allo-HCST: allogenic hematopoietic stem cell transplantation; MR: molecular response.</p>
Full article ">
Back to TopTop