[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
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
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
remove_circle_outline

Search Results (3,930)

Search Parameters:
Keywords = EGFR

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1447 KiB  
Article
Impact of Uric Acid Levels on Mortality and Cardiovascular Outcomes in Relation to Kidney Function
by Young-Eun Kwon, Shin-Young Ahn, Gang-Jee Ko, Young-Joo Kwon and Ji-Eun Kim
J. Clin. Med. 2025, 14(1), 20; https://doi.org/10.3390/jcm14010020 - 24 Dec 2024
Abstract
Background: Uric acid levels are linked to cardiovascular outcomes and mortality, especially in chronic kidney disease (CKD). However, their impact across varying kidney function remains unclear. Methods: We conducted a retrospective cohort study using the Observational Medical Outcomes Partnership Common Data [...] Read more.
Background: Uric acid levels are linked to cardiovascular outcomes and mortality, especially in chronic kidney disease (CKD). However, their impact across varying kidney function remains unclear. Methods: We conducted a retrospective cohort study using the Observational Medical Outcomes Partnership Common Data Model (OMOP-CDM) database from a single center. Adult patients with at least one serum uric acid measurement between 2002 and 2021 were included and categorized by estimated glomerular filtration rate (eGFR): normal kidney function (≥90 mL/min/1.73 m2), mild dysfunction (60–89 mL/min/1.73 m2), moderate dysfunction (30–59 mL/min/1.73 m2), and advanced dysfunction (<30 mL/min/1.73 m2). The primary outcome was all-cause mortality with secondary outcomes being myocardial infarction (MI) and heart failure (HF). Results: A total of 242,793 participants were analyzed. Uric acid levels showed a U-shaped association with all-cause mortality in advanced kidney dysfunction, where both low (<3 mg/dL) and high (>10 mg/dL) levels increased mortality risk. In mild kidney dysfunction, lower uric acid levels were linked to better survival. HF risk increased linearly with higher uric acid, particularly in normal kidney function, while no significant association was found between uric acid and MI in any group. Conclusions: Uric acid levels are associated with mortality in a U-shaped pattern for advanced kidney dysfunction, while lower levels appear protective in mild dysfunction. These findings suggest the need for personalized uric acid management in CKD patients based on their kidney function. Full article
(This article belongs to the Section Nephrology & Urology)
Show Figures

Figure 1

Figure 1
<p>Flowchart of study participants selection process.</p>
Full article ">Figure 2
<p>Adjusted hazard ratios for mortality according to uric acid levels by kidney function group. The figure shows adjusted cubic spline curves for the association between uric acid levels and all-cause mortality across four kidney function groups: normal kidney function, mild kidney dysfunction, moderate kidney dysfunction, and advanced kidney dysfunction. The solid black line represents the adjusted hazard ratios (HRs), while the dashed lines indicate the 95% confidence intervals. Adjustments were made for age, sex, hypertension, diabetes, cancer, myocardial infarction, body mass index, hemoglobin, blood urea nitrogen, creatinine, total cholesterol, and albumin. The reference uric acid level is 6–7 mg/dL.</p>
Full article ">Figure 3
<p>Mediation analysis of the association between uric acid levels and mortality. This figure illustrates the mediation pathways in the relationship between uric acid levels and 1-year mortality with the estimated glomerular filtration rate (eGFR) and serum albumin as mediators. Standardized coefficients for each path are displayed, and statistically significant mediation effects (<span class="html-italic">p</span> &lt; 0.05) are indicated.</p>
Full article ">
9 pages, 3967 KiB  
Case Report
Metabolic Complete Response of Metastatic Oncogene-Negative, PDL1-Negative Non-Small Cell Lung Cancer After Chemo-Immunotherapy and Radiotherapy: A Case Report
by Alessia Surgo, Valerio Davì, Maria Paola Ciliberti, Roberta Carbonara, Morena Caliandro, Fiorella Cristina Di Guglielmo, Nicola Sasso, Roberto Calbi, Maria Annunziata Gentile, Tiziana Talienti, Isabella Bruno, Michele Troia, Ilaria Bonaparte, Giuseppe Mario Ludovico, Giammarco Surico and Alba Fiorentino
Curr. Oncol. 2024, 31(12), 8118-8126; https://doi.org/10.3390/curroncol31120598 - 23 Dec 2024
Abstract
A 71-year-old male ex-smoker presented in October 2021 to our department with a brain and bone metastatic adenocarcinoma NSCLC. PDL1, ROS, EGFR, and ALK were negative. He underwent stereotactic radiotherapy for brain metastases. In November 2021, he started a chemotherapy (CHT) regimen with [...] Read more.
A 71-year-old male ex-smoker presented in October 2021 to our department with a brain and bone metastatic adenocarcinoma NSCLC. PDL1, ROS, EGFR, and ALK were negative. He underwent stereotactic radiotherapy for brain metastases. In November 2021, he started a chemotherapy (CHT) regimen with cisplatin (75 mg/m2 every 21 days) and pemetrexed (500 mg/m2 every 21 days), and ICI with Atezolizumab (1200 mg every 21 days). In July 2022, RT to the lung tumor and mediastinal nodal was performed with a total dose of 45 Gy in 15 fractions. He continued with immunotherapy until December 2022, when a grade 3–4 toxicity from immunotherapy was observed (hypothyroidism, psoriasis, and cystitis). He achieved a complete clinical response to the therapy. To date, the patient is alive, with a complete metabolic response, without treatment at 37 months from diagnosis. Full article
Show Figures

Figure 1

Figure 1
<p>Brain magnetic resonance at diagnosis. Legend: (<b>a</b>) right cerebellar lesion; (<b>b</b>) left cerebellar lesion; (<b>c</b>) left frontal lobe lesion.</p>
Full article ">Figure 2
<p>Lung tumor imaging at diagnosis. Legend: (<b>a</b>) lung tumor in the right upper lobe on CT scan; (<b>b</b>) PET-FDG hyper-capturing lesion on right superior lobe and mediastinal nodes.</p>
Full article ">Figure 3
<p>Radiotherapy treatment plans. Legend: (<b>a</b>) SRT treatment plan for cerebellar lesions; (<b>b</b>) SRT treatment plan for frontal right lesion; (<b>c</b>) mediastinal hypofractionated treatment plan. The colored areas show the isodoses which highlight the progressive decrease in the radiation dose moving from the center of the lesion to the periphery (from red to blue).</p>
Full article ">Figure 4
<p>Bladder biopsy. Legend: biopsy evidence of inflammatory process.</p>
Full article ">Figure 5
<p>MRI response to treatment. Legend: brain MRI complete response to treatments for cerebellar and frontal lesions 35 months after diagnosis.</p>
Full article ">Figure 6
<p>Response to treatment. Legend: (<b>a</b>) CT scan mediastinal and lung complete response to treatments 35 months after diagnosis; (<b>b</b>) PET-FDG complete response to treatments 35 months after diagnosis.</p>
Full article ">
11 pages, 755 KiB  
Article
Limited Efficacy of Anti-EGFR Monoclonal Antibodies in Colorectal Cancer Patients with Rare RAS Variants: Analysis of the C-CAT Database
by Shuhei Suzuki, Yosuke Saito, Koki Saito, Yuta Yamada, Koshi Takahashi, Ryosuke Kumanishi, Tadahisa Fukui and Takashi Yoshioka
Curr. Issues Mol. Biol. 2024, 46(12), 14476-14486; https://doi.org/10.3390/cimb46120869 - 23 Dec 2024
Abstract
Epidermal growth factor receptor (EGFR) inhibition is crucial in treating RAS wild-type metastatic colorectal cancer, yet current testing methods may miss rare RAS variants affecting treatment efficacy. We analyzed 4122 colorectal cancer patients receiving anti-EGFR antibodies from the Center for Cancer Genomics and [...] Read more.
Epidermal growth factor receptor (EGFR) inhibition is crucial in treating RAS wild-type metastatic colorectal cancer, yet current testing methods may miss rare RAS variants affecting treatment efficacy. We analyzed 4122 colorectal cancer patients receiving anti-EGFR antibodies from the Center for Cancer Genomics and Advanced Therapeutics database, identifying 54 patients (1.3%) with rare RAS variants undetectable by standard testing. These patients showed significantly lower response rates to anti-EGFR therapy (28.3%) compared to RAS wild-type cases (44.6%, p = 0.003). Disease control rates were also lower in rare variant cases (60.9%) versus wild-type cases (80.0%). Most common rare variants included KRAS Q22K, A59E, and A11_G12insGA. Comprehensive genomic profiling revealed additional alterations in TP53 (90.7%), APC (87.0%), and non-V600E BRAF mutations (25.9%). Our findings suggest that rare RAS variants predict poor anti-EGFR therapy response, highlighting the potential benefit of comprehensive genomic profiling before treatment initiation. This study provides real-world evidence supporting the clinical relevance of rare RAS variants in treatment decision-making for colorectal cancer. Future studies should focus on developing cost-effective comprehensive testing strategies and evaluating alternative treatment approaches for patients with rare RAS variants. Full article
Show Figures

Figure 1

Figure 1
<p>Genomic alterations and anti-EGFR antibodies (Upper, cetuximab; Lower, Panitumumab) treatment responses of rare RAS variants cases registered in the Center for Cancer Genomics and Advanced Therapeutics. TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated.</p>
Full article ">Figure 1 Cont.
<p>Genomic alterations and anti-EGFR antibodies (Upper, cetuximab; Lower, Panitumumab) treatment responses of rare RAS variants cases registered in the Center for Cancer Genomics and Advanced Therapeutics. TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated.</p>
Full article ">Figure 1 Cont.
<p>Genomic alterations and anti-EGFR antibodies (Upper, cetuximab; Lower, Panitumumab) treatment responses of rare RAS variants cases registered in the Center for Cancer Genomics and Advanced Therapeutics. TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated.</p>
Full article ">
9 pages, 412 KiB  
Article
Accuracy of Estimated Glomerular Filtration Rate Equations in Potential Vietnamese Living Kidney Donors
by Thang Diep, Tam Thai Thanh Tran, Chuan Khac Hoang and Sam Minh Thai
Transplantology 2024, 5(4), 312-320; https://doi.org/10.3390/transplantology5040031 - 21 Dec 2024
Viewed by 246
Abstract
Background: The accurate assessment of the glomerular filtration rate (GFR) in potential living kidney donors (PLKDs) is essential for successful transplantation and safeguarding kidney donation practice. Scintigraphy-measured GFR (mGFR) is widely regarded as the clinical reference standard. Various estimated GFR (eGFR) equations, [...] Read more.
Background: The accurate assessment of the glomerular filtration rate (GFR) in potential living kidney donors (PLKDs) is essential for successful transplantation and safeguarding kidney donation practice. Scintigraphy-measured GFR (mGFR) is widely regarded as the clinical reference standard. Various estimated GFR (eGFR) equations, such as the Modification of Diet in Renal Disease (MDRD), Cockcroft–Gault (CG), and Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equations, have been developed; however, none have been specifically validated for Vietnamese PLKDs. This study aimed to evaluate the accuracy of eGFR formulas compared to mGFR in PLKDs. Methods: This convenience retrospective study analyzed 189 PLKDs at Cho Ray Hospital in Vietnam from January 2014 to December 2020. The eGFR was calculated using various formulas and compared to the mGFR assessed using 99mTechnetium-diethylenetriaminepentaacetic acid. Bias, accuracy, and Bland–Altman plots were used to assess the significance of the eGFR values. Results: The median mGFR was 94.20 mL/min/1.73 m2 (interquartile range [IQR]: 88.40–100.50). The eGFR values were as follows: 77.52 mL/min/1.73 m2 (IQR: 70.50–86.33) for CG; 76.14 mL/min/1.73 m2 (IQR: 68.05–83.37) for MDRD; 106.80 ± 15.24 mL/min/1.73 m2 for CKD-EPI cystatin C 2012; 96.44 ± 13.40 mL/min/1.73 m2 for CKD-EPI creatinine cystatin C 2012; 88.74 ± 13.27 mL/min/1.73 m2 for CKD-EPI creatinine 2021; and 101.32 ± 12.82 mL/min/1.73 m2 for CKD-EPI creatinine cystatin C 2021. Among these formulas, the CKD-EPI creatinine cystatin C 2012 (P30 = 98.96%) and 2021 (P30 = 97.92%) showed the best consistency with the mGFR, owing to their high accuracy, low bias, and narrow limits of agreement in the Bland–Altman plots. Conclusions: The CKD-EPI equations based on creatinine and cystatin C are reliable tools for donor screening. Full article
(This article belongs to the Section Solid Organ Transplantation)
Show Figures

Figure 1

Figure 1
<p>Bland–Altman plot (agreement between glomerular filtration rate estimated by various equations and mGFR by <sup>99m</sup>Tc-DTPA).</p>
Full article ">
13 pages, 1050 KiB  
Article
Efficacy of the Once-Daily Tacrolimus Formulation LCPT Compared to the Immediate-Release Formulation in Preventing Early Post-Transplant Diabetes in High-Risk Kidney Transplant Patients: A Randomized, Controlled, Open-Label Pilot Study (EUDRACT: 2017-000718-52)
by Armando Torres, Concepción Rodríguez-Adanero, Constantino Fernández-Rivera, Domingo Marrero-Miranda, Eduardo de Bonis-Redondo, Aurelio P. Rodríguez-Hernández, Lourdes Pérez-Tamajón, Ana González-Rinne, Diego Álvarez-Sosa, Alejandra Álvarez-González, Nuria Sanchez-Dorta, Estefanía Pérez-Carreño, Laura Díaz-Martín, Sergio Luis-Lima, Ana E. Rodríguez-Rodríguez, Antonia María de Vera González, Cristina Romero-Delgado, María Calvo-Rodríguez, Rocío Seijo-Bestilleiro, Consuelo Rodríguez-Jiménez, Manuel Arturo Prieto López, Antonio Manuel Rivero-González, Domingo Hernández-Marrero and Esteban Porriniadd Show full author list remove Hide full author list
J. Clin. Med. 2024, 13(24), 7802; https://doi.org/10.3390/jcm13247802 - 20 Dec 2024
Viewed by 253
Abstract
Background/Objectives: Post-transplant diabetes mellitus (PTDM) and prediabetes (PreDM) are common after renal transplantation and increase the risk of cardiovascular events and mortality. Compared to immediate-release tacrolimus (IR-Tac), the LCPT formulation, with delayed absorption, offers higher bioavailability and a smoother time–concentration curve, potentially [...] Read more.
Background/Objectives: Post-transplant diabetes mellitus (PTDM) and prediabetes (PreDM) are common after renal transplantation and increase the risk of cardiovascular events and mortality. Compared to immediate-release tacrolimus (IR-Tac), the LCPT formulation, with delayed absorption, offers higher bioavailability and a smoother time–concentration curve, potentially reducing beta-cell stress. Methods: This randomized pilot trial compared de novo immunosuppression with IR-Tac (twice daily) and LCPT (once daily). At-risk recipients (age ≥ 60 years or 18–59 years with metabolic syndrome) were enrolled and followed for 3 months. The primary and secondary outcomes were the incidence of PTDM and PreDM, respectively. Results: 27 patients were randomized to IR-Tac and 25 to LCPT. The incidence of PTDM was comparable between groups [IR Tac: 18.5% (95% CI: 8.2–36.7%) vs. LCPT: 24% (95% CI: 11.5–43.4%); p = 0.7]. Although not statistically significant, the LCPT group exhibited a trend toward a reduction in PreDM incidence [IR-Tac: 40.7% (95% CI: 25–59%) vs. LCPT: 20% (95% CI: 9–39%); p = 0.1]. A sensitivity analysis showed similar results, with no significant differences in cumulative corticosteroid doses or baseline body mass index (BMI) between groups. The LCPT group showed a trend toward higher tacrolimus exposure at the end of the study [trough levels: IR-Tac group 8.3 (6.9–9.2) vs. LCPT group 9.4 (7.4–11.4) ng/mL; p = 0.05)], as well as fewer acute rejection episodes (none vs. three). Delayed graft function was more common in the IR-Tac group (37% vs. 8%; p = 0.01), and the eGFR was lower. Adverse events were comparable between groups. Conclusions: The potential biological activity of LCPT in preventing glucose metabolic alterations in at-risk patients warrants further investigation. Full article
(This article belongs to the Special Issue Advances in Kidney Transplantation)
Show Figures

Figure 1

Figure 1
<p>Patients’ disposition. IS: Immunosuppression; OGT: Oral glucose tolerance test. PKD-1: Autosomal Dominant Polycystic Kidney Disease type I; GN: Glomerulonephritis.</p>
Full article ">Figure 2
<p>Distribution of glucose metabolism abnormalities at the end of the study in each group. Prediabetes: Impaired Fasting Glucose and Impaired Glucose Tolerance, isolated or combined. (<b>A</b>): All patients; (<b>B</b>): Excluding patients with acute rejection or a baseline BMI &lt; 22 Kg/m<sup>2</sup>. IR-Tac: Immediate-release tacrolimus; LCPT: LCP Tacrolimus.</p>
Full article ">
13 pages, 1324 KiB  
Article
A Tri-Component (Glomerular, Tubular, and Metabolic) Assessment of Renal Function in Acute Heart Failure
by Gracjan Iwanek, Barbara Ponikowska, Husam Salah, Marat Fudim, Mateusz Guzik, Robert Zymliński, Krzysztof Aleksandrowicz, Beata Ponikowska and Jan Biegus
J. Clin. Med. 2024, 13(24), 7796; https://doi.org/10.3390/jcm13247796 - 20 Dec 2024
Viewed by 220
Abstract
Background: Despite the prevalence of impaired renal function in acute heart failure (AHF) patients, the intricate relationship between glomerular, tubular, and metabolic renal function remains unexplored. We aimed to investigate the co-occurrence of glomerular, tubular, and metabolic renal dysfunction in AHF and [...] Read more.
Background: Despite the prevalence of impaired renal function in acute heart failure (AHF) patients, the intricate relationship between glomerular, tubular, and metabolic renal function remains unexplored. We aimed to investigate the co-occurrence of glomerular, tubular, and metabolic renal dysfunction in AHF and their impact on prognosis. Methods: eGFR, spot urine sodium, and HCO3− were measured in 243 patients hospitalized for AHF. The population was stratified by the 4-point renal dysfunction score and linked with outcomes. Results: Glomerular dysfunction exhibited an elevated risk of death (HR of 2.04; 95% CI [1.24–3.36]; p = 0.006), combined risk of death, and HF rehospitalization (HR of 2.03; 95% CI [1.34–3.05]; p = 0.005). Similarly, tubular dysfunction correlated with a higher death risk (HR of 1.72; 95% CI [1.04–2.82]; p = 0.03) and a higher combined risk (HR of 1.82; 95% CI [1.21–2.74]; p = 0.004). While renal metabolic dysfunction was linked to increased death risk (HR of 1.82; 95% CI [1.07–3.11]; p = 0.028), it was not associated with composite risk (HR of 1.37; 95% CI [0.88–2.15]; p = 0.174). Multivariate analysis revealed a direct association between the renal dysfunction score and death risk (HR of 1.92 per 1 point; 95% CI [1.47–2.52]; p < 0.0001) and the combined risk of death and HF rehospitalization (HR of 1.78 per 1 point; 95% CI [1.43–2.22]; p < 0.0001). Conclusions: Renal dysfunction is common, with varied overlaps. Glomerular, tubular, and metabolic dysfunctions predict adverse outcomes in AHF. The established renal score may aid patient stratification and prognosis. Full article
(This article belongs to the Special Issue Clinical Challenges in Heart Failure Management)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier curves for one-year mortality by the renal dysfunction score. Log-rank, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Kaplan–Meier curves for death or heart failure rehospitalization (whichever occurred first) by the renal dysfunction score. Log-rank, <span class="html-italic">p</span> &lt; 0.0001.</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 262
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 ">
18 pages, 6611 KiB  
Article
The Impact of Bevacizumab and miR200c on EMT and EGFR-TKI Resistance in EGFR-Mutant Lung Cancer Organoids
by Nobuaki Kobayashi, Seigo Katakura, Nobuhiko Fukuda, Kohei Somekawa, Ayami Kaneko and Takeshi Kaneko
Genes 2024, 15(12), 1624; https://doi.org/10.3390/genes15121624 - 19 Dec 2024
Viewed by 229
Abstract
Objectives: This research aims to investigate the mechanisms of resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in non-small-cell lung cancer (NSCLC), particularly focusing on the role of the epithelial–mesenchymal transition (EMT) within the tumor microenvironment (TME). Materials and Methods [...] Read more.
Objectives: This research aims to investigate the mechanisms of resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in non-small-cell lung cancer (NSCLC), particularly focusing on the role of the epithelial–mesenchymal transition (EMT) within the tumor microenvironment (TME). Materials and Methods: We employed an in vitro three-dimensional organoid model that mirrors the physiology of human lung cancer. These organoids consist of lung cancer cells harboring specific EGFR mutations, human mesenchymal stem cells, and human umbilical vein endothelial cells. We analyzed EMT and drug resistance markers, and evaluated the effects of the anti-angiogenic agent Bevacizumab and micro-RNA miR200c. Results: The study identified a significant link between EMT and EGFR-TKI resistance. Notable findings included a decrease in E-cadherin and an increase in Zinc Finger E-Box Binding Homeobox 1 (ZEB1), both of which influenced EMT and resistance to treatment. Bevacizumab showed promise in improving drug resistance and mitigating EMT, suggesting an involvement of the Vascular Endothelial Growth Factor (VEGF) cascade. Transfection with miR200c was associated with improved EMT and drug resistance, further highlighting the role of EMT in TKI resistance. Conclusions: Our research provides significant insights into the EMT-driven EGFR-TKI resistance in NSCLC and offers potential strategies to overcome resistance, including the use of Bevacizumab and miR200c. However, due to the limitations in organoid models in replicating precise human cancer TME and the potential influence of specific EGFR mutations, further in vivo studies and clinical trials are necessary for validation. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Figure 1
<p>Construction and microscopic findings of spheroids and organoids using <span class="html-italic">Epidermal Growth Factor Receptor (EGFR)-</span>mutant lung cancer cells. (<b>A</b>) Hematoxylin and eosin (HE) staining and EGFR immunostaining of HCC827-derived spheroids and organoids. (<b>B</b>) HE and EGFR staining of structures generated from H1975 cells.</p>
Full article ">Figure 2
<p>Assessment of cell viability in response to various EGFR-TKIs in spheroids and organoids derived from cancer cells, determined by a luciferase assay. Each panel displays the influence of an individual EGFR-TKI on spheroids or organoids: (<b>A</b>) Gefitinib on HCC827, (<b>B</b>) Afatinib on HCC827, (<b>C</b>) Osimertinib on HCC827, and (<b>D</b>) Osimertinib on H1975. Each experiment was repeated three times (n = 3), with * <span class="html-italic">p</span> &lt; 0.05 (determined by the Mann–Whitney U test) indicating a statistically significant difference in cell viability.</p>
Full article ">Figure 3
<p>Immunofluorescence imaging of Epithelial Cadherin (E-cadherin) and zinc-finger-enhancer Binding Protein 1 (ZEB-1) in spheroids and organoids derived from lung cancer cell lines. (<b>A</b>) Visualization of these markers in HCC827-derived structures. The images illustrate cellular staining patterns for DAPI (blue, indicating cell nuclei), E-cadherin (green, an epithelial marker), and ZEB-1 (red, a mesenchymal marker). (<b>B</b>) Quantitative analysis of fluorescence intensity for e-cadherin and ZEB-1 in spheroids and organoids. the bar graphs depict the fluorescence intensity of E-cadherin and ZEB-1 in spheroids and organoids derived from <span class="html-italic">EGFR</span>-mutant lung cancer cells. Fluorescence intensity was measured and background-subtracted. Data represent the mean ± standard deviation (SD) of five independent experiments. Asterisks (*) indicate statistically significant differences between spheroids and organoids (<span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>C</b>,<b>D</b>) The same images for H1975-derived structures.</p>
Full article ">Figure 3 Cont.
<p>Immunofluorescence imaging of Epithelial Cadherin (E-cadherin) and zinc-finger-enhancer Binding Protein 1 (ZEB-1) in spheroids and organoids derived from lung cancer cell lines. (<b>A</b>) Visualization of these markers in HCC827-derived structures. The images illustrate cellular staining patterns for DAPI (blue, indicating cell nuclei), E-cadherin (green, an epithelial marker), and ZEB-1 (red, a mesenchymal marker). (<b>B</b>) Quantitative analysis of fluorescence intensity for e-cadherin and ZEB-1 in spheroids and organoids. the bar graphs depict the fluorescence intensity of E-cadherin and ZEB-1 in spheroids and organoids derived from <span class="html-italic">EGFR</span>-mutant lung cancer cells. Fluorescence intensity was measured and background-subtracted. Data represent the mean ± standard deviation (SD) of five independent experiments. Asterisks (*) indicate statistically significant differences between spheroids and organoids (<span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>C</b>,<b>D</b>) The same images for H1975-derived structures.</p>
Full article ">Figure 4
<p>Analysis of the combined effects of Bevacizumab and EGFR-TKI on EGFR-mutant cancer cells. (<b>A</b>,<b>B</b>) The results of a luciferase assay measuring cell viability following Osimertinib treatment alone or in combination with Bevacizumab in HCC827 and H1975 organoids, respectively (n = 3, * <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>C</b>) Immunofluorescence analysis of E-cadherin and ZEB-1 in HCC827. E-cadherin (green; left panels) and ZEB-1 (red; middle panels) expression and localization in HCC827 organoids, with and without Bevacizumab treatment. Merged images are shown in the right-hand panels. (<b>D</b>) Quantitative analysis of fluorescence intensity for E-cadherin and ZEB-1 expression in organoids. Values represent mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>E</b>,<b>F</b>) The corresponding analysis in H1975 organoids.</p>
Full article ">Figure 4 Cont.
<p>Analysis of the combined effects of Bevacizumab and EGFR-TKI on EGFR-mutant cancer cells. (<b>A</b>,<b>B</b>) The results of a luciferase assay measuring cell viability following Osimertinib treatment alone or in combination with Bevacizumab in HCC827 and H1975 organoids, respectively (n = 3, * <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>C</b>) Immunofluorescence analysis of E-cadherin and ZEB-1 in HCC827. E-cadherin (green; left panels) and ZEB-1 (red; middle panels) expression and localization in HCC827 organoids, with and without Bevacizumab treatment. Merged images are shown in the right-hand panels. (<b>D</b>) Quantitative analysis of fluorescence intensity for E-cadherin and ZEB-1 expression in organoids. Values represent mean ± SD from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>E</b>,<b>F</b>) The corresponding analysis in H1975 organoids.</p>
Full article ">Figure 5
<p>Western blot analysis examining E-cadherin and ZEB-1 expression in spheroids and organoids, with and without Bevacizumab treatment. (<b>A</b>) ZEB-1 (top) and E-cadherin (middle) levels in HCC827 spheroids and organoids. β-actin is utilized as a loading control (bottom panel). (<b>B</b>) Quantification of relative E-cadherin expression in HCC827 spheroids and organoids with or without Bevacizumab treatment. Data represent mean ± SD from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001 (Student’s <span class="html-italic">t</span>-test). (<b>C</b>) Western blot analysis of ZEB-1, E-cadherin, and β-actin expression in H1975 spheroids and organoids treated with or without Bevacizumab. (<b>D</b>) Quantification of relative E-cadherin expression in H1975 spheroids and organoids with or without Bevacizumab treatment. Data represent the mean ± SD from three independent experiments. ** <span class="html-italic">p</span> &lt; 0.001 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>The effects of miR200c on <span class="html-italic">EGFR</span>-mutant lung cancer organoids. (<b>A</b>) RT-PCR analysis comparing the expression of miR200c between organoids transfected with miR200c mimic and control organoids (n = 3, * <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>B</b>) Luciferase assay depicting cell viability in organoids treated with Osimertinib with or without the addition of the miR200c mimic (n = 3, * <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test). (<b>C</b>) Western blot analysis in spheroids and organoids with or without the introduction of the miR200c mimic, assessing the expression of ZEB-1 and β-actin. The bar graph, created using Image J, compares protein quantifications (n = 3, * <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney U test).</p>
Full article ">
21 pages, 316 KiB  
Review
Updates in Management of Unresectable Stage III Non Small Cell Lung Cancer: A Radiation Oncology Perspective
by Lakshmi Rekha Narra, Ritesh Kumar, Matthew P. Deek and Salma K. Jabbour
Cancers 2024, 16(24), 4233; https://doi.org/10.3390/cancers16244233 - 19 Dec 2024
Viewed by 358
Abstract
Unresectable stage III non-small-cell lung cancer (NSCLC) remains a clinical challenge, due to the need for optimal local and systemic control. The management of unresectable Stage III NSCLC has evolved with advancements in radiation therapy (RT), systemic therapies, and immunotherapy. For patients with [...] Read more.
Unresectable stage III non-small-cell lung cancer (NSCLC) remains a clinical challenge, due to the need for optimal local and systemic control. The management of unresectable Stage III NSCLC has evolved with advancements in radiation therapy (RT), systemic therapies, and immunotherapy. For patients with locally advanced NSCLC who are not surgical candidates, concurrent chemoradiotherapy (CRT) has modest survival outcomes, due to both local progression and distant metastasis. Efforts to enhance outcomes have led to dose-escalation trials, advances in modern RT techniques such as intensity-modulated RT (IMRT) and proton beam therapy (PBT), and the integration of adaptive RT to optimize target coverage while sparing organs at risk. Concurrent and consolidative immunotherapy, particularly with PD-L1 inhibitors, has shown promise, as evidenced by the PACIFIC trial, which demonstrated improved progression-free survival (PFS) and overall survival (OS) with durvalumab following CRT. Ongoing trials are now investigating novel immunotherapy combinations and targeted therapies in this setting, including dual checkpoint inhibition, DNA repair inhibitors, and molecularly targeted agents like osimertinib for EGFR-mutated NSCLC. Emerging biomarkers, such as circulating tumor DNA and radiomics, offer potential for personalizing treatment and predicting outcomes. Additionally, PBT and MR-guided adaptive RT have shown the potential to reduce toxicities while maintaining efficacy. Integrating these novel approaches may offer opportunities for optimizing treatment responses and minimizing adverse effects in this challenging patient population. Further investigation into patient stratification, biomarker-driven therapy, and refined therapeutic combinations is essential to improve long-term outcomes in unresectable Stage III NSCLC. This narrative review explores the current management strategies for unresectable Stage III NSCLC, from a radiation oncology perspective. Full article
22 pages, 8084 KiB  
Article
Optimization of Extraction of Luteolin from Schisandra chinensis by Ionic Liquid–Enzyme Complex System and Antioxidant Study Analysis
by Jingwei Hao, Nan Dong, Yifan Sun, Xiaoxia Lu, Yingying Pei, Yi Zhou, Xiangkun Zhou and Heming Liu
Separations 2024, 11(12), 354; https://doi.org/10.3390/separations11120354 - 19 Dec 2024
Viewed by 359
Abstract
The luteolin in Schisandra chinensis [Schisandraceae Schisandra (Turcz.) Baill.] were extracted by ultrasonic extraction assisted by an ionic liquid–enzyme composite system, and the content of luteolins was determined using high-performance liquid chromatography (HPLC). This process was initially conducted through a one-factor experiment and [...] Read more.
The luteolin in Schisandra chinensis [Schisandraceae Schisandra (Turcz.) Baill.] were extracted by ultrasonic extraction assisted by an ionic liquid–enzyme composite system, and the content of luteolins was determined using high-performance liquid chromatography (HPLC). This process was initially conducted through a one-factor experiment and a Box–Behnken combinatorial design of response surface method. The extraction process was optimized, and the results demonstrated that the optimal extraction conditions were 13.31% enzyme addition, 0.53 mol/L ionic liquid concentration, 173.47 min ultrasonic shaking, and 0.2266 mg/g, which was 4.88 times higher than that of the traditional reflux extraction. Secondly, the antioxidant function of luteolins was studied based on network pharmacology. For the study of the antioxidant mechanism of luteolin, the herb group identification database, SwissTargetPrediction on luteolins target prediction, and GeneCards database to achieve the antioxidant target were used. For the analysis of the intersection of the target protein interactions, GO bioanalysis and KEGG signaling pathway enrichment analysis were used. There were 57 overlapping targets of luteolin and antioxidants, including AKT1, MMP9, ESR1, EGFR, and SRC. GO function and KEGG pathway enrichment analysis showed that luteolin antioxidants were related to zoerythromycin metabolic process, adriamycin metabolic process, negative regulation of apoptotic process, endocrine resistance and oxidoreductase. The key targets in the pathways, such as luteolin AKT1 and MMP9, exert antioxidant effects. The antioxidant activity of luteolins was investigated by determining the scavenging ability of luteolins against two types of free radicals: 2,2-bipyridine-bis(3-ethyl-benzothiazole-6-sulfonic acid) diammonium salt (ABTS+) free radicals and 1,1-diphenyl-2-trinitrophenylhydrazine free radicals (DPPH-). The results of the antioxidant test demonstrated that the ABTS radical scavenging rate was 87.26%, and the DPPH radical scavenging rate was 93.85% when the quality concentration of Schisandra luteolins was 0.1 mg/g, indicating the potential of this natural antioxidant. This method of extracting Schisandra chinensis luteolins is highly productive, environmentally friendly, and practical, and it facilitates the development and utilization of industrial Schisandra chinensis. Full article
Show Figures

Figure 1

Figure 1
<p>Standard curve of luteolin.</p>
Full article ">Figure 2
<p>The effect of ionic liquid type on luteolin yield (<b>a</b>); the effect of ionic liquid concentration on luteolin yield (<b>b</b>); the effect of enzyme addition amount on luteolin yield (<b>c</b>); the effect of solid–liquid ratio on luteolin yield (<b>d</b>), the effect of ultrasonic time on luteolin yield (<b>e</b>); and the effect of enzyme addition amount on luteolin yield. Effect of ultrasonic extraction temperature on luteolin yield (<b>f</b>); effect of enzymolysis time on luteolin yield (<b>g</b>); effect of enzymolysis temperature on luteolin yield (<b>h</b>). Different letters labeled in the bar graph indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05) and the same letter indicates a non-significant difference (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 2 Cont.
<p>The effect of ionic liquid type on luteolin yield (<b>a</b>); the effect of ionic liquid concentration on luteolin yield (<b>b</b>); the effect of enzyme addition amount on luteolin yield (<b>c</b>); the effect of solid–liquid ratio on luteolin yield (<b>d</b>), the effect of ultrasonic time on luteolin yield (<b>e</b>); and the effect of enzyme addition amount on luteolin yield. Effect of ultrasonic extraction temperature on luteolin yield (<b>f</b>); effect of enzymolysis time on luteolin yield (<b>g</b>); effect of enzymolysis temperature on luteolin yield (<b>h</b>). Different letters labeled in the bar graph indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05) and the same letter indicates a non-significant difference (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 3
<p>Response surface contours and curves between each factor and yield.</p>
Full article ">Figure 3 Cont.
<p>Response surface contours and curves between each factor and yield.</p>
Full article ">Figure 4
<p>Interactive target information of luteolin and antioxidant.</p>
Full article ">Figure 5
<p>Drug—component—gene target network of luteolin antioxidant. The red triangle represents schisandra, the yellow triangle represents luteolin, and the blue rectangle represents the intersection target.</p>
Full article ">Figure 6
<p>PPI network of luteolin antioxidant function.</p>
Full article ">Figure 7
<p>Top 10 core targets in the PPI network of luteolin antioxidant function. The ranking of core targets is inversely proportional to the darkness of the color.</p>
Full article ">Figure 8
<p>Bubble diagram of GO function and KEGG path enrichment analysis. (<b>A</b>): BP; (<b>B</b>): MF; (<b>C</b>): CC; (<b>D</b>): KEGG.</p>
Full article ">Figure 8 Cont.
<p>Bubble diagram of GO function and KEGG path enrichment analysis. (<b>A</b>): BP; (<b>B</b>): MF; (<b>C</b>): CC; (<b>D</b>): KEGG.</p>
Full article ">Figure 9
<p>Scavenging ability of luteolin on free radicals. Different letters labeled in the bar graph indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05) and the same letter indicates a non-significant difference (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">
14 pages, 4542 KiB  
Article
Novel Cyclic Peptide–Drug Conjugate P6-SN38 Toward Targeted Treatment of EGFR Overexpressed Non-Small Cell Lung Cancer
by Andrii Bazylevich, Ayala Miller, Iryna Tkachenko, Maia Merlani, Leonid Patsenker, Gary Gellerman and Bat Chen R. Lubin
Pharmaceutics 2024, 16(12), 1613; https://doi.org/10.3390/pharmaceutics16121613 - 19 Dec 2024
Viewed by 370
Abstract
Background/Objectives: Here, we report on the synthesis and biological evaluation of a novel peptide–drug conjugate, P6-SN38, which consists of the EGFR-specific short cyclic peptide, P6, and the Topo I inhibitor SN38, which is a bioactive metabolite of the anticancer drug irinotecan. Methods: SN38 [...] Read more.
Background/Objectives: Here, we report on the synthesis and biological evaluation of a novel peptide–drug conjugate, P6-SN38, which consists of the EGFR-specific short cyclic peptide, P6, and the Topo I inhibitor SN38, which is a bioactive metabolite of the anticancer drug irinotecan. Methods: SN38 is attached to the peptide at position 20 of the E ring’s tertiary hydroxyl group via a mono-succinate linker. Results: The developed peptide–drug conjugate (PDC) exhibited sub-micromolar anticancer activity on EGFR-positive (EGFR+) cell lines but no effect on EGFR-negative (EGFR−) cells. In vivo studies have shown that this PDC specifically accumulates in EGFR+ non-small cell lung cancer (NSCLC) xenografts and presents superior anticancer activity compared to the EGFR-specific antibody cetuximab (ErbituxTM) and free SN38. The 10 mg/kg dose of P6-SN38 in a side-by-side EGFR+/EGFR− xenograft shows eradication of the EGFR+ tumor with good tolerance, but no inhibition of tumor growth of the EGFR− counterpart. Conclusions: The PDC examined in this study was proven to be highly efficient for NSCLC, broadening its utilization for targeted cancer therapy in EGFR overexpressed cancers. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure of P6 and SN38. To present the locations of the functional groups more clearly, the key positions of the SN38 structure were numbered.</p>
Full article ">Figure 2
<p>Cell viability after 6 h of pre-incubation of EGFR+ H1299 (<b>A</b>) and EGFR− HEK293 (<b>B</b>) cell lines with P6-SN38 and SN38. H1299 and HEK293 cells were cultured in a 96-well plate and pre-incubated with six different concentrations (0.5, 2.5, 5, 10, 15, and 25 μM) of P6-SN38 or SN38 for 6 h, washed out, and incubated additionally for 24 h, 48 h, and 72 h. Cell viability was assessed using an XTT assay, with the viability of untreated control cells set at 100%. The viability of treated cells was expressed as a percentage relative to the control. Data are presented as the mean ± standard deviation (SD).</p>
Full article ">Figure 3
<p>Apoptosis of H1299 cells estimated through cell survival percentage by binding with annexin V-FITC. H1299 cells were pre-incubated with P6-SN38 and SN38 as described in <a href="#sec2dot6-pharmaceutics-16-01613" class="html-sec">Section 2.6</a>. Cells were stained with annexin and PI and analyzed by flow cytometry; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0005. “ns” means non-significant.</p>
Full article ">Figure 4
<p>Cell cycle arrest analysis of treatment with SN38 and P6-SN38. Cell cycle distribution of H1299 cells detected by flow cytometry after 24 h (<b>A</b>) and 48 h (<b>B</b>) treatment with SN38 and P6-SN38 at 2.5 µM. A total of 1 × 10<sup>6</sup> cells were pre-incubated with P6-SN38 or SN38 for 6 h, followed by additional incubation for 24 h and 48 h. The cells were then stained with propidium iodide (PI) and analyzed by flow cytometry (<b>C</b>). The data in A and B are presented as the mean ± SD (<span class="html-italic">n</span> = 3). “ns” means non-significant. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0005. “ns” means non-significant.</p>
Full article ">Figure 4 Cont.
<p>Cell cycle arrest analysis of treatment with SN38 and P6-SN38. Cell cycle distribution of H1299 cells detected by flow cytometry after 24 h (<b>A</b>) and 48 h (<b>B</b>) treatment with SN38 and P6-SN38 at 2.5 µM. A total of 1 × 10<sup>6</sup> cells were pre-incubated with P6-SN38 or SN38 for 6 h, followed by additional incubation for 24 h and 48 h. The cells were then stained with propidium iodide (PI) and analyzed by flow cytometry (<b>C</b>). The data in A and B are presented as the mean ± SD (<span class="html-italic">n</span> = 3). “ns” means non-significant. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0005. “ns” means non-significant.</p>
Full article ">Figure 5
<p>Representative side-by-side xenograft bearing K562/NSCLC tumors before injection (<b>A</b>), white light image of resected tumors and organs at 24 h post-injection (<b>B</b>), fluorescence image of resected tumors and organs at 24 h post-injection (<b>C</b>).</p>
Full article ">Figure 6
<p>In vivo effect of P6-SN38 conjugate in NSCLC model (10 mg/kg, once a week). Effect of P6-SN38, free SN38, and Erbitux on tumor volume compared to non-treated control (<b>A</b>) and mice weight (<b>B</b>). The resected tumors are shown in (<b>C</b>). <span class="html-italic">N</span> = 4 in each group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6 Cont.
<p>In vivo effect of P6-SN38 conjugate in NSCLC model (10 mg/kg, once a week). Effect of P6-SN38, free SN38, and Erbitux on tumor volume compared to non-treated control (<b>A</b>) and mice weight (<b>B</b>). The resected tumors are shown in (<b>C</b>). <span class="html-italic">N</span> = 4 in each group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Scheme 1
<p>Synthesis of TBDMS-SN38-suc intermediate.</p>
Full article ">Scheme 2
<p>Solid phase synthesis of P6-SN38 peptide conjugate. The N and C terminus of peptide are highlighted with bold.</p>
Full article ">
13 pages, 724 KiB  
Article
Uric Acid Correlates with Serum Levels of Mineral Bone Metabolism and Inflammation Biomarkers in Patients with Stage 3a–5 Chronic Kidney Disease
by Francisco Mendoza Carrera, Gloria Elizabeth Vázquez Rivera, Caridad A. Leal Cortés, Lourdes del Carmen Rizo De la Torre, Renato Parra Michel, Rosalba Orozco Sandoval and Mariana Pérez Coria
Medicina 2024, 60(12), 2081; https://doi.org/10.3390/medicina60122081 - 19 Dec 2024
Viewed by 332
Abstract
Background and Objectives: Uric acid (UA) and the markers of mineral bone metabolism and inflammation are commonly altered in patients with chronic kidney disease (CKD) and are associated with the risk of cardiovascular complications and death. Studies point to a link between [...] Read more.
Background and Objectives: Uric acid (UA) and the markers of mineral bone metabolism and inflammation are commonly altered in patients with chronic kidney disease (CKD) and are associated with the risk of cardiovascular complications and death. Studies point to a link between high serum UA and mineral bone homeostasis and inflammation, but controversy remains. The aim of this study was to evaluate the relationship between UA levels and mineral bone metabolism and inflammation biomarkers in a sample of Mexican patients with CKD 3a–5. Materials and Methods: This cross-sectional study included 146 Mexican patients with CKD 3a–5. In addition, 25 healthy subjects were included in the study with the aim of generating reference data for comparisons. Metabolic parameters including UA serum concentrations, mineral bone metabolism (parathormone (PTH), fibroblast growth factor 23 (FGF23), calcium, and phosphate), and inflammation (interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α)) biomarkers were measured in all of the samples and compared as a function of the estimated glomerular function rate (eGFR) or UA levels. Results: Intact PTH, FGF23, and cytokines were higher in advanced CKD stages. Patients with hyperuricemia had significantly higher values of FGF23 and TNF-α compared with those without hyperuricemia. The eGFR was found to be significantly and negatively correlated with all markers. Uric acid was significantly correlated with phosphate, iPTH, FGF23, and TNF-α, whereas iPTH was significantly correlated with FGF23, TNF-α, and FGF23. Finally, a multivariate analysis confirmed the relationship of eGFR with all the tested biomarkers, as well as other relationships of iPTH with UA and TNF-α and of FGF23 with UA and TNF-α. Conclusions: This study supports the relationship between uric acid and levels of mineral bone metabolism and inflammation biomarkers in patients with CKD at middle to advanced stages. In the follow-up of patients with CKD, monitoring and controlling UA levels through nutritional or pharmacological interventions could help in the prevention of alterations related to mineral bone metabolism. Full article
(This article belongs to the Section Urology & Nephrology)
Show Figures

Figure 1

Figure 1
<p>Serum concentrations of uric acid, mineral bone metabolism, and inflammation biomarkers according to the CKD stage. The <span class="html-italic">p-</span>values are obtained from ANOVA or Kruskal–Wallis tests, as appropriate. Abbreviations: HS: healthy subjects; iPTH, intact parathyroid hormone; FGF23, fibroblast growth factor 23; IL, interleukin; TNF, tumor necrosis factor.</p>
Full article ">
18 pages, 16257 KiB  
Article
Effectiveness and Mechanism of Resibufogenin on Human Renal Cancer Cell Caki-1
by Yuqi Wu, Yue Yang, Run Huang, Tao Li, Chunlei Wan and Lei Zhang
Biology 2024, 13(12), 1064; https://doi.org/10.3390/biology13121064 - 19 Dec 2024
Viewed by 356
Abstract
In this study, we investigated the effect and mechanism of Resibufogenin on renal cell carcinoma based on network pharmacology, molecular docking, and in vitro experiments. The results showed that there were 35 cross-targets between Resibufogenin and renal cell carcinoma. GO and KEGG pathway [...] Read more.
In this study, we investigated the effect and mechanism of Resibufogenin on renal cell carcinoma based on network pharmacology, molecular docking, and in vitro experiments. The results showed that there were 35 cross-targets between Resibufogenin and renal cell carcinoma. GO and KEGG pathway analyses indicated that Resibufogenin inhibited renal cancer cells through the vascular smooth muscle contraction signalling pathway and EGFR tyrosine kinase inhibitor resistance signaling pathway, and MAPK1, PRKCB, and Resibufogenin had strong associative activities. After different concentrations of Resibufogenin were applied to human renal cancer cells, it was found that the IC50 value was 408.2 nM, 10 nM resibufogenin could significantly inhibit cell migration (p < 0.0001), the percentage of apoptosis and necrosis increased dose-dependently, and the expression of genes of MAPK1 and PRKCB in the cells was significantly reduced (p < 0.001) in a dose-dependent manner. The above results indicate that Resibufogenin can inhibit human renal cell carcinoma through multi-targets and multi-methods, which provides a theoretical basis for the application of Resibufogenin in the treatment of renal cell carcinoma and the development of novel drugs in the future. Full article
(This article belongs to the Section Cell Biology)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of the Resibufogenin.</p>
Full article ">Figure 2
<p>GSE66272 Volcano Map. (<b>a</b>) GSE66272 Volcano Plot (<b>b</b>) GSE66272 Volcano Plot.</p>
Full article ">Figure 3
<p>Protein-protein interaction network of renal cell carcinoma and Resibufogenin.</p>
Full article ">Figure 4
<p>The top five ranked targets across three algorithms ((<b>a</b>). Degree; (<b>b</b>). MCC; (<b>c</b>). NNC).</p>
Full article ">Figure 5
<p>GO enrichment analysis of intersecting targets.</p>
Full article ">Figure 6
<p>KEGG enrichment analysis of intersecting targets.</p>
Full article ">Figure 7
<p>Molecular docking results ((<b>a</b>). Resibufogenin with MAPK1; (<b>b</b>). Resibufogenin with PRKCB).</p>
Full article ">Figure 8
<p>Resibufogenin drug acts on Caki-1 12 h (100×). ((<b>A</b>). CK; (<b>B</b>). 10 nmol/L; (<b>C</b>). 50 nmol/L; (<b>D</b>). 100 nmol/L; (<b>E</b>). 200 nmol/L).</p>
Full article ">Figure 9
<p>Resibufogenin drug acts on Caki-1 12 h (400×). ((<b>A</b>). CK; (<b>B</b>). 10 nmol/L; (<b>C</b>). 50 nmol/L; (<b>D</b>). 100 nmol/L; (<b>E</b>). 200 nmol/L).</p>
Full article ">Figure 10
<p>Resibufogenin drug acts on Caki-1 24 h (100×). (<b>A</b>). CK; (<b>B</b>). 10 nmol/L; (<b>C</b>). 50 nmol/L; (<b>D</b>). 100 nmol/L; (<b>E</b>). 200 nmol/L).</p>
Full article ">Figure 11
<p>Resibufogenin drug acts on Caki-1 24 h (400×) ((<b>A</b>). CK; (<b>B</b>). 10 nmol/L; (<b>C</b>). 50 nmol/L; (<b>D</b>). 100 nmol/L; (<b>E</b>). 200 nmol/L).</p>
Full article ">Figure 12
<p>The aspect ratio on Caki-1 24 h. (* <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 13
<p>The effect of Resibufogenin on the activity of Caki-1 cells. (*** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 14
<p>The effect of Resibufogenin on the migration rate of Caki-1 cells. (**** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 15
<p>The effect of Resibufogenin on the apoptosis rate of Caki-1 cells. ((<b>a</b>). CK; (<b>b</b>). 50 nmol/L; (<b>c</b>). 100 nmol/L; (<b>d</b>). 200 nmol/L).</p>
Full article ">Figure 16
<p>Changes in the gene expression of Caki-1 cells under the action of Resibufogenin ((<b>a</b>). MAPK1; (<b>b</b>). PRKCB). (*** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
15 pages, 3006 KiB  
Article
Au@109Pd Core–Shell Nanoparticles Conjugated to Panitumumab for the Combined β—Auger Electron Therapy of Triple-Negative Breast Cancer
by Nasrin Abbasi Gharibkandi, Agnieszka Majkowska-Pilip, Rafał Walczak, Mateusz Wierzbicki and Aleksander Bilewicz
Int. J. Mol. Sci. 2024, 25(24), 13555; https://doi.org/10.3390/ijms252413555 - 18 Dec 2024
Viewed by 276
Abstract
Apart from HER2-positive, triple-negative breast cancer (TNBC) is the second most highly invasive type of breast cancer. Although TNBC does not overexpress HER2 receptors, it has been observed that EGFR protein expression is present in this specific type of tumor, making it an [...] Read more.
Apart from HER2-positive, triple-negative breast cancer (TNBC) is the second most highly invasive type of breast cancer. Although TNBC does not overexpress HER2 receptors, it has been observed that EGFR protein expression is present in this specific type of tumor, making it an attractive target for immune and radiopharmaceutical treatments. In our current study, we used 109Pd (T1/2 = 13.7 h) in the form of a 109Pd/109mAg in vivo generator as a source of β particles and Auger electrons in targeted radionuclide therapy for TNBC. 109Pd, obtained through neutron irradiation of the 108Pd target, was deposited onto 15 nm gold nanoparticles to form Au@109Pd core–shell nanoparticles, which were then conjugated to the panitumumab antibody. Au@109Pd-PEG-panitumumab nanoparticles were bound, internalized, and partially routed to the nucleus in MDA-MB-231 human breast cancer cells overexpressing EGFR receptors. The Au@109Pd-panitumumab radioconjugate significantly reduced the metabolic activity of MDA-MB-231 cells in a dose-dependent manner. In conclusion, we have found that Au@109Pd-PEG-panitumumab nanoparticles show potential as a therapeutic agent for combined β–Auger electron targeted radionuclide therapy of TNBC. The simultaneous emission of β, conversion, and Auger electrons from the 109Pd/109mAg generator, similar to 161Tb conjugates, significantly enhances the therapeutic effect. The partial localization of these nanoparticles into the cell nucleus, provided by the panitumumab vector, ensures effective therapy with Auger electrons. This is particularly important for the treatment of drug-resistant TNBC cells. Full article
(This article belongs to the Special Issue New Advances in Nanomedicine Innovation in Cancer Treatment)
Show Figures

Figure 1

Figure 1
<p>The scheme of the developed Au@<sup>109</sup>Pd-PEG-panitumumab radiobioconjugate.</p>
Full article ">Figure 2
<p>Changes in the hydrodynamic diameter of the Au@Pd-PEG-panitumumab nanoparticles incubated in 10 mM PBS buffer and 0.9% NaCl.</p>
Full article ">Figure 3
<p>Binding studies of Au@<sup>109</sup>PdNP-PEG-panitumumab on MDA-MB-231 cell line.</p>
Full article ">Figure 4
<p>Internalization of Au@Pd-PEG-panitumumab NPs into the MDA-MB-231 cell overexpressing EGFR receptor (<b>left</b>) and intranuclear uptake of radioconjugate (<b>right</b>).</p>
Full article ">Figure 5
<p>Internalization of panitumumab, Au@Pd-PEG-COOH, and Au@Pd-PEG-panitumumab in MDA-MB-231 cells determined by confocal microscopy. The fluorescence signals indicate the following: subcellular panitumumab distribution (green) and nuclei intracellular localization (blue). Au@Pd-containing particles (dark spots) were also visualized with a transmitted light detector (T-PMT).</p>
Full article ">Figure 6
<p>Metabolic viability of MDA-MB-231 cells after treatment with different concentrations of Au@Pd-PEG-panitumumab non-radioactive conjugates (<b>a</b>) and with different radioactive doses of Au@<sup>109</sup>Pd-PEG-panitumumab NPs (<b>b</b>) and of Au@<sup>109</sup>Pd-PEG radioactive conjugates (<b>c</b>) after 24 h, 48 h, and 72 h of incubation. ). The results are expressed as mean ± SD. <span class="html-italic">p</span>-values are presented as follows: (*) <span class="html-italic">p</span> ≤ 0.05, (**) <span class="html-italic">p</span> ≤ 0.001, (***), and <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 7
<p>Confocal immunofluorescence microscopy exhibiting γH2A.X foci (red) in the nucleus (counterstained blue with DAPI) and merged (purple) of MDA-MB-231 breast cancer cells treated with various radioactivity of Au@<sup>109</sup>Pd-PEG-panitumumab recorded after 4 h (<b>left</b>) and 24 h (<b>right</b>) of incubation.</p>
Full article ">Figure 8
<p>Quantification of γH2AX foci from the images shown in <a href="#ijms-25-13555-f007" class="html-fig">Figure 7</a>.</p>
Full article ">
21 pages, 5449 KiB  
Article
Rational Design of an Epidermal Growth Factor Receptor Vaccine: Immunogenicity and Antitumor Research
by Yifei Liu, Zehui Liu and Zhongliang Zheng
Biomolecules 2024, 14(12), 1620; https://doi.org/10.3390/biom14121620 - 18 Dec 2024
Viewed by 451
Abstract
The epidermal growth factor receptor (EGFR) is frequently overexpressed in a variety of human epithelial tumors, and its aberrant activation plays a pivotal role in promoting tumor growth, invasion, and metastasis. The clinically approved passive EGFR-related therapies have numerous limitations. Seven EGFR-ECD epitope [...] Read more.
The epidermal growth factor receptor (EGFR) is frequently overexpressed in a variety of human epithelial tumors, and its aberrant activation plays a pivotal role in promoting tumor growth, invasion, and metastasis. The clinically approved passive EGFR-related therapies have numerous limitations. Seven EGFR-ECD epitope peptides (EG1-7) were selected through bioinformatics epitope prediction tools including NetMHCpan-4.1, NetMHCIIpan-3.2, and IEDB Consensus (v2.18 and v2.22) and fused to the translocation domain of diphtheria toxin (DTT). The A549 tumor model was successfully established in a murine mouse model. The vaccine was formulated by combining the adjuvants Alum and CpG and subsequently assessed for its immunogenicity and anti-tumor efficacy. DTT-EG (3;5;6;7) vaccines elicited specific humoral and cellular immune responses and effectively suppressed tumor growth in both prophylactic and therapeutic mouse tumor models. The selected epitopes EG3 (HGAVRFSNNPALCNV145-159), EG5 (KDSLSINATNIKHFK346-360), EG6 (VKEITGFLLIQAWPE398-412), and EG7 (LCYANTINWKKLFGT469-483) were incorporated into vaccines for active immunization, representing a promising strategy for the treatment of tumors with overexpressed epidermal growth factor receptor (EGFR). The vaccine design and fusion method employed in this study demonstrate a viable approach toward the development of cancer vaccines. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Immunization with DTT-EG vaccines in combination with tumor cell injection protocol. (<b>A</b>) Prophylactic tumor model: tumor cell injection and vaccine immunization protocol (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a>) and (<b>B</b>) therapeutic tumor model: tumor cell injection and vaccine immunization protocol (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a> (accessed on 9 June 2024)).</p>
Full article ">Figure 2
<p>The predicted epitope EG in EGFR-ECD is demonstrated, along with the design and expression purification of DTT-EG utilizing DTT as a vector. (<b>A</b>) Displaying the EG epitope within EGFR-ECD (EGFR PDB id:3njp): EG1 is represented by a yellow sequence, EG2 by a red sequence, EG3 by a green sequence, EG4 by a blue and orange sequence, EG5 by an orange and purple sequence, EG6 by a cyan sequence, and EG7 by a pink sequence. (<b>B</b>) Design of DTT-EG tandem recombinant protein. DTT (202–373) denotes the amino acid fragment spanning from 202 to 373 of the DTT protein. The epitope prediction tool identified seven human-specific epitope peptides in the form of EG1, EG2, EG3, EG4, and E5G5G6G7 consisting of 15 amino acid residues each. GS represents the GS-linker sequence (GGTGGTGGTGGTAGTGGTGGTGGTGGTAGT). (<b>C</b>) Analysis of purified recombinant protein using 12% SDS-PAGE (M: Marker; A: DTT-EG1; B: DTT-EG2; C: DTT-EG3; D: DTT-EG4; E: DTT-EG5; F: DTT-EG6; G: DTT-EG7; H: DTT).</p>
Full article ">Figure 3
<p>Serum antibodies were detected using enzyme-linked immunosorbent assay (ELISA) following immunization of mice with the DTT-EG tandem recombinant protein. (Significance levels were denoted as ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, while “ns” indicated no significant difference). (<b>A</b>) Schematic representation of the immunization protocol for mice (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a> (accessed on 9 June 2024)). (<b>B</b>) ELISA analysis was conducted on the serum of immunized mice, with the coated proteins being DTT or EGFR. (<b>C</b>) The titer of antibodies against EGFR in the mouse serum following vaccination was determined by ELISA.</p>
Full article ">Figure 4
<p>The splenic lymphocytes of immunized mice were tested for cell proliferation, toxicity, and interferon-gamma release, and the spleens were tested for CD4 and CD8 immunohistochemistry. (Significance levels were denoted as * <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, while “ns” indicated no significant difference). (<b>A</b>) Schematic representation of the immunization protocol for mice (mouse diagram by <a href="http://medpeer.cn" target="_blank">medpeer.cn</a> (accessed on 9 June 2024)). (<b>B</b>) Cell proliferation detection experiment by CCK-8 method. (<b>C</b>) Cell cytotoxicity detection experiment by lactate dehydrogenase method. (<b>D</b>) Detection of IFN-γ release by ELISA method. (<b>E</b>) The spleen of immunized mice was stained by immunohistochemistry with anti-CD4 specific antibody. (<b>F</b>) The spleen of immunized mice was stained by immunohistochemistry with anti-CD8 specific antibody. (<b>G</b>) CD4+ T and CD8+ T-cell density was quantified using ImageJ.</p>
Full article ">Figure 5
<p>Antitumor effects of the DTT-EG vaccine in a prophylactic and therapeutic mouse A549 tumor model. (Significance levels were denoted as * <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, while “ns” indicated no significant difference). (<b>A</b>) prophylactic tumor model tumor growth curve, (<b>B</b>) prophylactic tumor model tumor weight, (<b>C</b>) therapeutic tumor model tumor growth curve, and (<b>D</b>) therapeutic tumor model tumor weight.</p>
Full article ">Figure 6
<p>The DTT-EG vaccine modulates the infiltration of CD8+ T cells and induces necrosis within the intratumoral tissue. (Significance levels were denoted as ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001, while “ns” indicated no significant difference). (<b>A</b>) Tumor tissues underwent immunohistochemical staining using an anti-CD4 specific antibody. (<b>B</b>) Tumor tissues underwent immunohistochemical staining using an anti-CD8 specific antibody. (<b>C</b>) CD4+ T and CD8+ T cell density was quantified using ImageJ. (<b>D</b>) Revealing the histopathological features of tumor tissue through hematoxylin and eosin (H&amp;E) staining.</p>
Full article ">
Back to TopTop