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

Journals

Article Types

Countries / Regions

Search Results (256)

Search Parameters:
Keywords = AZD9291

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
32 pages, 13931 KiB  
Article
Alisertib and Barasertib Induce Cell Cycle Arrest and Mitochondria-Related Cell Death in Multiple Myeloma with Enhanced Efficacy Through Sequential Combination with BH3-Mimetics and Panobinostat
by Andrea Benedi, Manuel Beltrán-Visiedo, Nelia Jiménez-Alduán, Alfonso Serrano-Del Valle, Alberto Anel, Javier Naval and Isabel Marzo
Cancers 2025, 17(14), 2290; https://doi.org/10.3390/cancers17142290 - 9 Jul 2025
Viewed by 605
Abstract
Background: The treatment landscape for multiple myeloma (MM) has significantly evolved in recent decades with novel therapies like proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies. However, MM remains incurable, necessitating new pharmacological strategies. Mitotic kinases, such as Aurora proteins, have emerged as potential [...] Read more.
Background: The treatment landscape for multiple myeloma (MM) has significantly evolved in recent decades with novel therapies like proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies. However, MM remains incurable, necessitating new pharmacological strategies. Mitotic kinases, such as Aurora proteins, have emerged as potential targets. Selective inhibitors of Aurora A and B,- alisertib (MLN8237) and barasertib (AZD1152), respectively, have shown anti-myeloma activity in preclinical studies, with alisertib demonstrating modest efficacy in early clinical trials. Methods and Results: This study investigated the mechanisms of action of alisertib and barasertib and their combination with antitumor agents in a panel of five MM cells lines. Both drugs induced cell cycle arrest phase and abnormal nuclear morphologies. Alisertib caused prolonged mitotic arrest, whereas barasertib induced transient arrest, both resulting in the activation of mitotic catastrophe. These findings revealed three potential outcomes: cell death, senescence, or polyploidy. High mitochondrial reactive oxygen species (mROS) were identified as possible drivers of cell death. Caspase inhibition reduced caspase-3 activation but did not prevent cell death. Interestingly, alisertib at low doses remained toxic to Bax/BakDKO cells, although mitochondrial potential disruption and cytochrome c release were observed. Sequential combinations of high-dose Aurora kinase inhibitors with BH3-mimetics, and in specific cases with panobinostat, showed a synergistic effect. Conversely, the simultaneous combination of alisertib and barasertib showed mostly antagonistic effects. Conclusions: Alisertib and barasertib emerge as potential in vitro candidates against MM, although further studies are needed to validate their efficacy and to find the best combinations with other molecules. Full article
(This article belongs to the Special Issue Advances in Molecular Oncology and Therapeutics)
Show Figures

Figure 1

20 pages, 18100 KiB  
Article
Targeting p-FGFR1Y654 Enhances CD8+ T Cells Infiltration and Overcomes Immunotherapy Resistance in Esophageal Squamous Cell Carcinoma by Regulating the CXCL8–CXCR2 Axis
by Hong Luo, Liwei Wang, Hui Gao, Daijun Zhou, Yu Qiu, Lijia Yang, Jing Li, Dan Du, Xiaoli Huang, Yu Zhao, Zhongchun Qi, Yue Zhang, Xuemei Huang, Lihan Sun, Tao Xu and Dong Li
Biomedicines 2025, 13(7), 1667; https://doi.org/10.3390/biomedicines13071667 - 8 Jul 2025
Viewed by 469
Abstract
Background: Esophageal squamous cell carcinoma (ESCC) is a fatal malignant tumor. Several studies have demonstrated that immune checkpoint inhibitors can provide clinical benefits to patients with ESCC. However, the single-agent efficacy of these agents remains limited. Although combination therapies (e.g., radiotherapy, chemotherapy) can [...] Read more.
Background: Esophageal squamous cell carcinoma (ESCC) is a fatal malignant tumor. Several studies have demonstrated that immune checkpoint inhibitors can provide clinical benefits to patients with ESCC. However, the single-agent efficacy of these agents remains limited. Although combination therapies (e.g., radiotherapy, chemotherapy) can help to overcome immunotherapy resistance in ESCC, their severe side effects limit clinical application. This study aimed to explore new resistance mechanisms to immunotherapy in ESCC and identify novel molecular targets to overcome immunotherapy resistance. Methods: We employed immunohistochemistry staining to examine the p-FGFR1Y654 in tumor samples obtained from 103 patients with ESCC, in addition to evaluating CD8+ T cell infiltration. In vitro expression, western blotting, CCK-8, 5-bromo-2′-deoxyuridine incorporation assays, and migration assays were used to confirm the impact of AZD4547 on p-FGFR1Y654 expression and the proliferation and migration in ESCC cell lines. Through RNA sequencing analysis, databases such as the Cancer Genome Atlas (TCGA) and Gene Set Cancer Analysis (GSCA), and the reconstruction of transgenic mice using the humanized immune system, we validated the correlation between the expression of p-FGFR1Y654 and CD8+ T cell infiltration. We also explored how p-FGFR1Y654 recruits myeloid-derived suppressor cells (MDSCs) through the CXCL8–CXCR2 axis to suppress the therapeutic efficacy of immunotherapy in ESCC. Finally, the tumor-suppressive effects of AZD4547 combined with immunotherapy were confirmed in vivo in tumor-bearing mice with a humanized immune system. Results: We found that the inhibition of p-FGFR1Y654 expression in ESCC can enhance CD8+ T cell infiltration by suppressing the CXCL8-–XCR2 recruitment of MDSCs. AZD4547, combined with immunotherapy, further promotes immunotherapeutic efficacy in ESCC. Conclusions: In conclusion, our study presents a promising model for combination therapy in ESCC immunotherapy. Full article
(This article belongs to the Section Immunology and Immunotherapy)
Show Figures

Figure 1

15 pages, 2142 KiB  
Article
DNA Damage Response Regulation Alleviates Neuroinflammation in a Mouse Model of α-Synucleinopathy
by Sazzad Khan, Himanshi Singh, Jianfeng Xiao and Mohammad Moshahid Khan
Biomolecules 2025, 15(7), 907; https://doi.org/10.3390/biom15070907 - 20 Jun 2025
Cited by 1 | Viewed by 586
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder marked by the degeneration of dopaminergic neurons in the substantia nigra, leading to decreased dopamine levels in the striatum and causing a range of motor and non-motor impairments. Although the molecular mechanisms driving PD progression [...] Read more.
Parkinson’s disease (PD) is a progressive neurodegenerative disorder marked by the degeneration of dopaminergic neurons in the substantia nigra, leading to decreased dopamine levels in the striatum and causing a range of motor and non-motor impairments. Although the molecular mechanisms driving PD progression remain incompletely understood, emerging evidence suggests that the buildup of nuclear DNA damage, especially DNA double-strand breaks (DDSBs), plays a key role in contributing neurodegeneration, promoting senescence and neuroinflammation. Despite the pathogenic role for DDSB in neurodegenerative disease, targeting DNA repair mechanisms in PD is largely unexplored as a therapeutic approach. Ataxia telangiectasia mutated (ATM), a key kinase in the DNA damage response (DDR), plays a crucial role in neurodegeneration. In this study, we evaluated the therapeutic potential of AZD1390, a highly selective and brain-penetrant ATM inhibitor, in reducing neuroinflammation and improving behavioral outcomes in a mouse model of α-synucleinopathy. Four-month-old C57BL/6J mice were unilaterally injected with either an empty AAV1/2 vector (control) or AAV1/2 expressing human A53T α-synuclein to the substantia nigra, followed by daily AZD1390 treatment for six weeks. In AZD1390-treated α-synuclein mice, we observed a significant reduction in the protein level of γ-H2AX, a DDSB marker, along with downregulation of senescence-associated markers, such as p53, Cdkn1a, and NF-κB, suggesting improved genomic integrity and attenuation of cellular senescence, indicating enhanced genomic stability and reduced cellular aging. AZD1390 also significantly dampened neuroinflammatory responses, evidenced by decreased expression of key pro-inflammatory cytokines and chemokines. Interestingly, mice treated with AZD1390 showed significant improvements in behavioral asymmetry and motor deficits, indicating functional recovery. Overall, these results suggest that targeting the DDR via ATM inhibition reduces genotoxic stress, suppresses neuroinflammation, and improves behavioral outcomes in a mouse model of α-synucleinopathy. These findings underscore the therapeutic potential of DDR modulation in PD and related synucleinopathy. Full article
Show Figures

Figure 1

10 pages, 1037 KiB  
Article
Antitumor Effect of mTOR1/2 Dual Inhibitor AZD8055 in Canine Pulmonary Carcinoma
by Tomokazu Nagashima, Kazuhiko Ochiai, Yuka Takizawa, Youta Koike, Takahiro Saito, Asumi Muramatsu, Daigo Azakami, Yukino Machida, Makoto Bonkobara, Toshiyuki Ishiwata and Masaki Michishita
Cancers 2025, 17(12), 1991; https://doi.org/10.3390/cancers17121991 - 14 Jun 2025
Viewed by 867
Abstract
Background/Objectives: Primary pulmonary carcinoma (PC) is a malignant neoplasm that occurs in humans, dogs, and other species. In canine PC, palliative care remains the most practical approach for dogs with inoperable PC. Methods: We investigated the effectiveness of mammalian target of rapamycin (mTOR) [...] Read more.
Background/Objectives: Primary pulmonary carcinoma (PC) is a malignant neoplasm that occurs in humans, dogs, and other species. In canine PC, palliative care remains the most practical approach for dogs with inoperable PC. Methods: We investigated the effectiveness of mammalian target of rapamycin (mTOR) inhibitors in canine lung cancer upon PI3K/AKT/mTOR activation. Three canine PC cell lines (AZACL1, AZACL2, and cPAC-1) were treated with three mTOR inhibitors (AZD8055, temsirolimus, and everolimus). In vitro, sensitivity assays were conducted to evaluate proliferation and Western blotting was used to examine pathway activation and phosphorylation of mTOR-related protein. Results: AZD8055 had a stronger inhibitory effect on cell proliferation than temsirolimus and everolimus in all three PC cell lines. The IC50 for AZD8055 in the AZACL1, AZACL2, and cPAC-1 cell lines were 23.8 μM, 95.8 nM, and 237 nM, for temsirolimus they were 34.6 μM, 11.5 μM, and 11.2 μM, and for everolims they were 36.6 μM, 33.4 μM, and 33.0 μM, respectively. Western blotting revealed PI3K/AKT/mTOR pathway activation and differential phosphorylation of mTOR signal-related proteins across the three PC cell lines. In xenograft mice injected with the AZACL1 and AZACL2 cell lines we showed that the AZD8055-treated group exhibited a significant reduction in tumor volume via the inhibition of tumor growth compared to the control group. Conclusions: These findings reveal that the PI3K/AKT/mTOR pathway plays a key role in canine PC and that AZD8055 may be a novel therapeutic agent for PC-bearing dogs. Full article
(This article belongs to the Special Issue Pulmonary Nodule and Lung Cancer: Diagnosis and Clinical Treatment)
Show Figures

Figure 1

24 pages, 2661 KiB  
Review
Oral Small-Molecule GLP-1 Receptor Agonists: Mechanistic Insights and Emerging Therapeutic Strategies
by Héctor Iván Saldívar-Cerón, Jorge Arturo Vargas-Camacho, Sonia León-Cabrera, Paola Briseño-Díaz, Ari Evelyn Castañeda-Ramírez, Axel Eduardo Muciño-Galicia and María Regina Díaz-Domínguez
Sci. Pharm. 2025, 93(2), 26; https://doi.org/10.3390/scipharm93020026 - 11 Jun 2025
Viewed by 3256
Abstract
Small-molecule glucagon-like peptide-1 receptor agonists (GLP-1RAs) represent an innovative advancement in oral therapeutics, addressing key limitations associated with injectable peptide-based incretin therapies. These nonpeptidic agents exert their actions primarily through non-canonical binding orthosteric sites within the GLP-1 receptor transmembrane domain, enabling selective G [...] Read more.
Small-molecule glucagon-like peptide-1 receptor agonists (GLP-1RAs) represent an innovative advancement in oral therapeutics, addressing key limitations associated with injectable peptide-based incretin therapies. These nonpeptidic agents exert their actions primarily through non-canonical binding orthosteric sites within the GLP-1 receptor transmembrane domain, enabling selective G protein (Gs)-biased signaling with reduced β-arrestin-mediated adverse effects. Orforglipron has notably advanced through Phase 3 clinical development, demonstrating significant reductions in hemoglobin A1c and body weight (up to 7.9%) with favorable tolerability. Conversely, promising candidates such as danuglipron and lotiglipron were discontinued due to hepatotoxicity, underscoring critical safety concerns intrinsic to small-molecule GLP-1RA development. Current clinical candidates, including GSBR-1290, CT-996, and ECC5004, continue to offer substantial potential due to their oral bioavailability, simplified dosing regimens, and favorable gastrointestinal tolerability. Nevertheless, challenges persist regarding hepatic safety, pharmacodynamic variability, and limited long-term outcome data. This review integrates current structural, pharmacological, and clinical evidence, highlights key mechanistic innovations—including biased agonism, covalent binding strategies, and allosteric modulation—and discusses future directions for this rapidly evolving therapeutic class in metabolic disease management. Full article
(This article belongs to the Topic Research in Pharmacological Therapies, 2nd Edition)
Show Figures

Graphical abstract

29 pages, 3643 KiB  
Article
Transcriptomic Analyses of Ovarian Clear Cell Carcinoma Spheroids Reveal Distinct Proliferative Phenotypes and Therapeutic Vulnerabilities
by Bart Kolendowski, Sylvia Cheng, Yudith Ramos Valdes, Trevor G. Shepherd and Gabriel E. DiMattia
Cells 2025, 14(11), 785; https://doi.org/10.3390/cells14110785 - 27 May 2025
Cited by 1 | Viewed by 797
Abstract
Cancer cell spheroids autonomously form in the ascites fluid and are considered a conduit for epithelial ovarian cancer metastasis within the peritoneal cavity. Spheroids are homotypic, avascular 3D structures that acquire resistance to anoikis to remain viable after cellular detachment. We used in [...] Read more.
Cancer cell spheroids autonomously form in the ascites fluid and are considered a conduit for epithelial ovarian cancer metastasis within the peritoneal cavity. Spheroids are homotypic, avascular 3D structures that acquire resistance to anoikis to remain viable after cellular detachment. We used in vitro spheroid model systems to interrogate pathways critical for spheroid cell proliferation, distinct from those driving monolayer cancer cell proliferation. Using the 105C and KOC-7c human ovarian clear cell carcinoma (OCCC) cell lines, which have distinct proliferative phenotypes as spheroids but the same prototypical OCCC gene mutation profile of constitutively activated AKT signaling with the loss of ARID1A, we revealed therapeutic targets that efficiently kill cells in spheroids. RNA-seq analyses compared the transcriptome of 3-day monolayer and spheroid cells from these lines and identified the characteristics of dormant spheroid cell survival, which included the G2/M checkpoint, autophagy, and other stress pathways induced in 105C spheroids, in sharp contrast to the proliferating spheroid cells of the KOC-7c cell line. Next, we assessed levels of various G2/M checkpoint regulators and found a consistent reduction in steady-state levels of checkpoint regulators in dormant spheroid cells, but not proliferative spheroids. Our studies showed that proliferative spheroid cells were sensitive to Wee1 inhibition by AZD1775, but the dormant spheroid cells showed a degree of resistance to AZD1775, both in terms of EC50 values and spheroid reattachment abilities. Thus, we identified biomarkers of dormant spheroids, including the G2/M checkpoint regulators Wee1, Cdc25c, and PLK1, and showed that, when compared to proliferating spheroid cells, the transcriptome of dormant OCCC spheroids is a source of therapeutic targets. Full article
Show Figures

Graphical abstract

15 pages, 612 KiB  
Article
Rates of SARS-CoV-2 Breakthrough Infection or Severe COVID-19 and Associated Risk Factors After Primary and Booster Vaccination Against COVID-19 in the Netherlands
by Jesse M. van den Berg, Marieke T. Blom, Jetty A. Overbeek, Sharon Remmelzwaal, Ron M. C. Herings and Petra J. M. Elders
Vaccines 2025, 13(6), 564; https://doi.org/10.3390/vaccines13060564 - 26 May 2025
Viewed by 785
Abstract
Background: The effectiveness of COVID-19 vaccines appears to decline rapidly over time due to waning immunity and immune evasion by emerging variants of concern, and may be reduced in high-risk populations. We aimed to evaluate the rates of SARS-CoV-2 breakthrough infection or severe [...] Read more.
Background: The effectiveness of COVID-19 vaccines appears to decline rapidly over time due to waning immunity and immune evasion by emerging variants of concern, and may be reduced in high-risk populations. We aimed to evaluate the rates of SARS-CoV-2 breakthrough infection or severe COVID-19, both in individuals who had completed their primary COVID-19 vaccination, and in those who had received their first booster vaccination. Specifically, we aimed to evaluate whether persons with certain risk factors, such as age, gender, socioeconomic status (SES), and specified comorbidities have an increased risk of either breakthrough infection or severe COVID-19, compared to those without the respective risk factors. Methods: Data on COVID-19 vaccinations, infections, hospitalizations, and deaths were collected from the PHARMO Data Network, consisting of health records from Dutch residents. Two cohorts were established: (1) all persons who have completed their primary COVID-19 vaccination regimen, and (2) those who have received their first booster vaccination. The outcomes were SARS-CoV-2 breakthrough infection, and severe COVID-19, defined as either hospitalization or death following SARS-CoV-2 infection. Incidence rates of these outcomes were calculated in both cohorts. The adjusted incidence rate ratios of these outcomes in persons with certain risk factors were calculated, using generalized linear models with a Poisson distribution. Results: In 2021, a total of 1,090,567 individuals received either two doses of BNT162b2, AZD1222, or mRNA-1273, or one dose of Ad26.COV2.S and were included in the primary vaccination cohort, of which 344,153 (31.6%) received a booster vaccination. Overall incidence rates of SARS-CoV-2 breakthrough infection and severe COVID-19 after primary vaccination were 29.9 and 3.1 per 1000 person-years, respectively, and after booster vaccination were 256.4 and 2.3, respectively. Male gender, older age, lower SES, history of COVID-19, and recent hospitalization were factors associated with a lower risk of breakthrough infection after primary vaccination, and a higher risk of severe COVID-19. The risk of severe COVID-19 after primary vaccination was increased in persons with several comorbidities, compared to those without, and remained elevated after booster vaccination in persons with diabetes or lung disease. Conclusions: Our study emphasizes the crucial role of boosters in reducing breakthrough infections, particularly in high-risk populations. The varied impact on severe outcomes in individuals with comorbidities underscores the need for ongoing surveillance and tailored vaccination strategies. Full article
Show Figures

Figure 1

24 pages, 2444 KiB  
Review
The CXCL1-CXCR2 Axis as a Component of Therapy Resistance, a Source of Side Effects in Cancer Treatment, and a Therapeutic Target
by Jan Korbecki, Mateusz Bosiacki, Maciej Pilarczyk, Marcin Kot, Piotr Defort, Ireneusz Walaszek, Dariusz Chlubek and Irena Baranowska-Bosiacka
Cancers 2025, 17(10), 1674; https://doi.org/10.3390/cancers17101674 - 15 May 2025
Viewed by 1177
Abstract
CXCL1 (Gro-α, MGSA) is a chemokine functionally similar to CXCL8/IL-8, as both activate the same receptor, CXCR2. CXCL1 levels are frequently elevated in tumors compared to healthy tissue, where they play a key role in promoting cancer cell migration, angiogenesis, and neutrophil recruitment. [...] Read more.
CXCL1 (Gro-α, MGSA) is a chemokine functionally similar to CXCL8/IL-8, as both activate the same receptor, CXCR2. CXCL1 levels are frequently elevated in tumors compared to healthy tissue, where they play a key role in promoting cancer cell migration, angiogenesis, and neutrophil recruitment. While the involvement of CXCL1 in tumor progression is well established, its relevance to cancer therapy remains underexplored. This review examines the therapeutic potential of targeting CXCL1 and its receptor, CXCR2, in cancer treatment. It discusses anti-CXCL1 antibodies and CXCR2 antagonists, including AZD5069, SB225002, SCH-479833, navarixin/SCH-527123, ladarixin/DF2156A, and reparixin, as well as strategies to enhance CXCR2 expression in lymphocytes during adoptive cell therapy to improve immunotherapy outcomes. Particular attention is given to the role of CXCL1 in treatment resistance, including resistance to chemotherapy, radiotherapy, and anti-angiogenic therapy. Cancer therapies often upregulate CXCL1 expression, which in turn drives treatment resistance. Additionally, this review explores the contribution of CXCL1 to therapy-induced side effects, such as chemotherapy-induced metastasis, neuropathy, nephrotoxicity, diarrhea, and cardiotoxicity. CXCR2 inhibitors are well tolerated by patients in clinical trials. However, the limited number of studies evaluating these agents in combination with standard chemotherapy precludes any definitive conclusions. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

20 pages, 4025 KiB  
Article
Deciphering Nicotine-Driven Oncogenesis in Head and Neck Cancer: Integrative Transcriptomics and Drug Repurposing Insights
by Guo-Rung You, Daniel Yu Chang, Hung-Han Huang, Yin-Ju Chen, Joseph T. Chang and Ann-Joy Cheng
Cancers 2025, 17(9), 1430; https://doi.org/10.3390/cancers17091430 - 24 Apr 2025
Viewed by 594
Abstract
Background: Chronic nicotine exposure drives head and neck cancer (HNC) progression, yet its molecular mechanisms remain underexplored. This study examines nicotine-induced transcriptomic changes and potential therapies via drug repurposing. Methods: HNC cell lines (OECM1, SAS, and CGHNC9) were exposed to an [...] Read more.
Background: Chronic nicotine exposure drives head and neck cancer (HNC) progression, yet its molecular mechanisms remain underexplored. This study examines nicotine-induced transcriptomic changes and potential therapies via drug repurposing. Methods: HNC cell lines (OECM1, SAS, and CGHNC9) were exposed to an IC30 nicotine dose for three months to model chronic exposure in habitual smokers. Transcriptomic profiling of these sublines was integrated with TCGA-HNSC patient data. Differentially expressed genes (DEGs) underwent functional pathway enrichment analysis. Drug repurposing was conducted using gene–drug correlation analysis across GDSC, CTRP, and PRISM databases. Results: Transcriptomic analysis identified 1223 DEGs in nicotine-exposed HNC cells, and integration with TCGA-HNSC data defined a Nic-HNC gene set of 168 genes: 149 oncogenes and 19 tumor suppressors, with 36 oncogenes overexpressed in heavy smokers. Pathway analysis revealed the upregulation of oncogenic signaling, such as PI3K-AKT, alongside the suppression of immune regulation and metabolic reprogramming. Drug repurposing identified five compounds—AZD1332, JAK-8517, NU7441, BRD-K30748066, and neopeltolide—with the first two exhibiting the strongest inverse correlations with nicotine-induced oncogenes in heavy smokers, highlighting their potential as targeted therapies for tobacco-associated HNC. Conclusions: This study comprehensively characterizes nicotine-driven molecular dysregulation in HNC and proposes AZD1332 and JAK-8517 as promising therapeutic candidates through drug repurposing. These insights advance our understanding of nicotine’s oncogenic role and provide a foundation for translational research to develop targeted interventions for tobacco-associated HNC. Full article
(This article belongs to the Special Issue Advances in Drug Repurposing to Overcome Cancers)
Show Figures

Figure 1

32 pages, 16024 KiB  
Article
Modulating NPC1L1 to Potentiate PARP Inhibitor-Induced Ferroptosis and Immune Response in Triple-Negative Breast Cancer
by Ge Li, Yuxia Yuan, Xinhua Wu and Lixian Wu
Pharmaceutics 2025, 17(5), 554; https://doi.org/10.3390/pharmaceutics17050554 - 24 Apr 2025
Viewed by 688
Abstract
Background/Objectives: Poly (ADP-ribose) polymerase (PARP) inhibitors have shown significant efficacy in treating BRCA-mutated cancers; however, a significant proportion of patients fail to respond. Emerging evidence highlights the role of PARP in lipid metabolism, suggest-ing its modulation as a novel strategy to regulate tumor [...] Read more.
Background/Objectives: Poly (ADP-ribose) polymerase (PARP) inhibitors have shown significant efficacy in treating BRCA-mutated cancers; however, a significant proportion of patients fail to respond. Emerging evidence highlights the role of PARP in lipid metabolism, suggest-ing its modulation as a novel strategy to regulate tumor progression. Methods: In this study, lipidomics and transcriptomics analyses were conducted to elucidate the mechanisms underlying PARP inhibitor-induced ferroptosis and immune modulation in triple-negative breast cancer (TNBC). Results: We demonstrated that the PARP inhibitor Niraparib significantly reprograms lipid metabolism in TNBC cells, marked by elevated phosphatidylethanolamine (PE) and cholesterol ester (ChE) levels. This metabolic shift was mechanistically linked to upregulation of the cholesterol transporter NPC1L1 via the PARP1-RELA-NPC1L1 signaling axis, which subsequently activated the AKT pathway. Combinatorial treatment with Niraparib and either Ezetimibe (an NPC1L1 inhibitor) or AZD5363 (an AKT inhibitor) synergistically enhanced TNBC cell death by promoting ferroptosis through glutathione depletion and lipid peroxidation. Furthermore, NPC1L1 inhibition amplified PARP inhibitor-induced immune responses, increasing CD8+ T cell infiltration and cytotoxicity in tumors. Conclusions: In conclusion, our findings establish NPC1L1 as a critical mediator of PARP inhibitor efficacy and propose dual targeting of lipid metabolism, providing a new therapeutic approach for the combination treatment of TNBC. Full article
Show Figures

Graphical abstract

21 pages, 8546 KiB  
Article
Identification of KIFC3 as a Colorectal Cancer Biomarker and Its Regulatory Mechanism in the Immune Microenvironment Based on Integrated Analysis of Multi-Omics Databases
by Fen Wang, Xinxin Zeng, Jielun Wen, Kexin Xian, Feng Jin, Sunfang Jiang and Liyue Sun
Biomedicines 2025, 13(4), 859; https://doi.org/10.3390/biomedicines13040859 - 2 Apr 2025
Viewed by 639
Abstract
Background: Studies suggest that kinesin family (KIF) members can promote the occurrence of colorectal cancer (CRC). However, the mechanism of action has not yet been elucidated. The aim of this study was to identify CRC biomarkers associated with KIF members and to investigate [...] Read more.
Background: Studies suggest that kinesin family (KIF) members can promote the occurrence of colorectal cancer (CRC). However, the mechanism of action has not yet been elucidated. The aim of this study was to identify CRC biomarkers associated with KIF members and to investigate their biological mechanisms in the treatment of colorectal cancer by analyzing multi-omics data. Methods: CRC-related datasets and KIF member-related genes (KIFRGs) were used. First, differentially expressed genes (DEGs) and differentially expressed methylation genes (DEMGs) in the TCGA-CRC were identified separately using different expression analyses (CRC vs. control). The intersecting genes were selected by overlapping the DEGs, DEMGs, and KIFRGs. Candidate genes were identified using survival analysis (p < 0.05). Subsequently, based on the candidate genes, biomarkers were selected by gene expression validation and survival analysis. Subsequently, functional enrichment, immune cell infiltration, and drug sensitivity analyses were performed. Single-cell analysis was utilized to perform cell annotation, and then function enrichment and pseudo-temporal analyses were performed. Results: The 12 intersecting genes were identified by overlapping 12,479 DEGs, 11,319 DEMGs, and 43 KIFRGs. The survival analysis showed that Kinesin Family Member C2 (KIFC2) and Kinesin Family Member C3 (KIFC3) had significant differences in survival (p < 0.05). Moreover, KIFC3 passed the gene expression validation and survival analysis validation (p < 0.05); thus, KIFC3 was deemed a biomarker. Subsequently, the pathways involved in KIFC3 were detected, such as the Ecm receptor intersection and chemokine signaling pathway. In addition, we found that KIFC3 was significantly positively correlated with natural killer (NK) cells (r = 0.455, p < 0.05) and NK T cells (r = 0.411, p < 0.05). Moreover, in the drug sensitivity of the CRC, the potential therapeutic benefits of AZD.2281, nilotinib, PD.173074, and shikonin were detected. Furthermore, using single-cell analysis, 16 cell clusters were annotated, and epithelial cells and M2-like macrophages were enriched in “rheumatoid arthritis”. Additionally, we observed that most M1-like macrophages were present in the early stages of differentiation, whereas M2-like macrophages were predominant in the later stages of differentiation. Conclusions: This study identifies KIFC3 as a CRC biomarker through multi-omics analysis, highlighting its unique expression, survival association, immune correlations, and drug sensitivity for potential diagnostic and therapeutic applications. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

24 pages, 3059 KiB  
Review
Oral and Non-Oral Cholesterol-Lowering Drugs with PCSK9 and Other Biomolecules as Targets: Present Status and Future Prospects
by Ajoy Basak
Biomolecules 2025, 15(4), 468; https://doi.org/10.3390/biom15040468 - 22 Mar 2025
Viewed by 1851
Abstract
The accumulation of high levels of cholesterol associated with low-density lipoprotein (LDL) in the bloodstream is the key risk factor for stroke and cardiovascular diseases. Therefore, reducing the concentration of LDL-cholesterol in the blood and maintaining it at an optimum level are vital [...] Read more.
The accumulation of high levels of cholesterol associated with low-density lipoprotein (LDL) in the bloodstream is the key risk factor for stroke and cardiovascular diseases. Therefore, reducing the concentration of LDL-cholesterol in the blood and maintaining it at an optimum level are vital especially for hypercholesterolemic individuals and cardiovascular patients. Thus, the study of cholesterol management and regulation in the physiological system has drawn significant attention from researchers across the globe. This led to the discovery of several cholesterol-lowering drugs which have been approved for administration either via oral or non-oral routes. Owing to the high comfort level, reduced or no pain, and fewer side effects with oral administration, more focus has been directed towards the development of oral-based cholesterol-lowering drugs. The other modes of administration such as intravenous or intramuscular injections are complex and sometimes painful and less tolerable. Therefore, there was a significant interest to develop oral drugs targeting PCSK9. In fact, some progress has been accomplished in recent years. This review provides an overview of the existing cholesterol-lowering drugs, and the progress made so far with oral-based PCSK9 drugs for lowering LDL-cholesterol. The review is presented in several sections highlighting the molecular targets, the individual drugs, and the modes of administration, with a focus on the oral route. Full article
(This article belongs to the Section Chemical Biology)
Show Figures

Graphical abstract

30 pages, 4516 KiB  
Article
Mutational Scanning and Binding Free Energy Computations of the SARS-CoV-2 Spike Complexes with Distinct Groups of Neutralizing Antibodies: Energetic Drivers of Convergent Evolution of Binding Affinity and Immune Escape Hotspots
by Mohammed Alshahrani, Vedant Parikh, Brandon Foley, Nishank Raisinghani and Gennady Verkhivker
Int. J. Mol. Sci. 2025, 26(4), 1507; https://doi.org/10.3390/ijms26041507 - 11 Feb 2025
Cited by 2 | Viewed by 1294
Abstract
The rapid evolution of SARS-CoV-2 has led to the emergence of variants with increased immune evasion capabilities, posing significant challenges to antibody-based therapeutics and vaccines. In this study, we conducted a comprehensive structural and energetic analysis of SARS-CoV-2 spike receptor-binding domain (RBD) complexes [...] Read more.
The rapid evolution of SARS-CoV-2 has led to the emergence of variants with increased immune evasion capabilities, posing significant challenges to antibody-based therapeutics and vaccines. In this study, we conducted a comprehensive structural and energetic analysis of SARS-CoV-2 spike receptor-binding domain (RBD) complexes with neutralizing antibodies from four distinct groups (A–D), including group A LY-CoV016, group B AZD8895 and REGN10933, group C LY-CoV555, and group D antibodies AZD1061, REGN10987, and LY-CoV1404. Using coarse-grained simplified simulation models, rapid energy-based mutational scanning, and rigorous MM-GBSA binding free energy calculations, we elucidated the molecular mechanisms of antibody binding and escape mechanisms, identified key binding hotspots, and explored the evolutionary strategies employed by the virus to evade neutralization. The residue-based decomposition analysis revealed energetic mechanisms and thermodynamic factors underlying the effect of mutations on antibody binding. The results demonstrate excellent qualitative agreement between the predicted binding hotspots and the latest experiments on antibody escape. These findings provide valuable insights into the molecular determinants of antibody binding and viral escape, highlighting the importance of targeting conserved epitopes and leveraging combination therapies to mitigate the risk of immune evasion. Full article
(This article belongs to the Collection Feature Papers in Molecular Biophysics)
Show Figures

Figure 1

15 pages, 2239 KiB  
Article
Profile of Humoral Immunity and B Cell Pool in Infection with the SARS-CoV-2 Prototype Strain and AZD1222 (ChAdOx nCoV-19) Vaccination
by Débora Familiar-Macedo, Elzinandes Leal de Azeredo, Elba Regina Sampaio de Lemos, Paulo Vieira Damasco and Luzia Maria de-Oliveira-Pinto
Vaccines 2025, 13(2), 101; https://doi.org/10.3390/vaccines13020101 - 21 Jan 2025
Viewed by 1168
Abstract
Background/Objectives: Understanding the behavior of B cells during infection and vaccination is important for determining protective humoral immunity. We evaluated the profile of humoral immunity and B cell pool in individuals who were acutely infected with SARS-CoV-2, recovered from COVID-19, or received two [...] Read more.
Background/Objectives: Understanding the behavior of B cells during infection and vaccination is important for determining protective humoral immunity. We evaluated the profile of humoral immunity and B cell pool in individuals who were acutely infected with SARS-CoV-2, recovered from COVID-19, or received two doses of the AZD1222 vaccine. Methods: Peripheral blood mononuclear cells (PBMCs) from these individuals were subjected to in vitro stimulation to promote the differentiation of B cells into antibody-secreting cells (ASCs), and the ELISpot evaluated the abundance of pan and SARS-CoV-2 Spike S1-reactive IgG+ ASC. Stimulated PBMCs were characterized using flow cytometry. Culture supernatants were assessed for soluble B-cell-activating factors. The IgA and IgG for the S1 were evaluated through ELISA. Results: The recovered individuals displayed a robust S1 ASC compared to acute and vaccinated individuals. Although the frequency of total B cells or B cell subsets did not vary among the groups, plasmablast cells were increased in naïve and double-negative B cells in the acute, recovered, and vaccinated individuals. Similar IgA and IgG production appeared to be present in the acute and recovered individuals. During vaccination, more IgG is produced than IgA. In acute patients, BAFF levels were positively correlated with total B cells and IgG+ plasmablast cells but negatively correlated with IgA+ plasmablast cells. Conclusions: Vaccination and natural infection with COVID-19 induce a differential profile and functionality of B cells. We suggest that new vaccines against COVID-19 incorporate molecular adjuvants that regulate B lymphocyte functionality and consider the beneficial aspects of the IgA response in addition to IgG. Full article
Show Figures

Figure 1

9 pages, 3534 KiB  
Article
Radiolabeling and Preliminary In Vivo Evaluation of the Candidate CCR2 Targeting PET Radioligand [11C]AZD2423
by Kenneth Dahl, Peter Johnström, Miklós Tóth, Martin Bolin, Katarina Varnäs, Ryuji Nakao, Akihiro Takano, Yasir Khani Meynaq, Malken Bayrakdarian, Zsolt Cselényi, Christer Halldin, Lars Farde and Magnus Schou
Pharmaceuticals 2025, 18(2), 135; https://doi.org/10.3390/ph18020135 - 21 Jan 2025
Viewed by 1011
Abstract
Background: AZD2423 is a high-affinity and selective negative allosteric modulator of the chemokine receptor type 2 (CCR2). This receptor plays important roles in the extravasation and transmigration of monocytes under inflammatory conditions. The aims of the current positron emission tomography (PET) study were [...] Read more.
Background: AZD2423 is a high-affinity and selective negative allosteric modulator of the chemokine receptor type 2 (CCR2). This receptor plays important roles in the extravasation and transmigration of monocytes under inflammatory conditions. The aims of the current positron emission tomography (PET) study were as follows: (i) to develop an efficient synthetic method for labeling AZD2423 with carbon-11 (11C, t1/2 = 20.4 min) and (ii) to evaluate its potential to visualize CCR2 binding in the non-human primate (NHP) brain. Methods: [11C]AZD2423 was synthesized using a novel two-step, two-pot [11C]carbon monoxide carbonylation procedure. PET imaging studies in NHPs (n = 2) were conducted to assess its brain penetration and in vivo distribution. Results: Radiolabeling of [11C]AZD2423 was accomplished with good yield (7.4 ± 0.6%, n = 4) and high radiochemical purity (>99%) using [11C]carbon monoxide. Preliminary PET imaging in NHPs revealed low [11C]AZD2423 brain exposure under both baseline and pretreatment conditions (SUVpeak = 0.4, n = 2). However, high concentrations of radioactivity were observed in organs outside the brain at baseline, e.g., the thyroid gland (SUVpeak = 3.3, n = 2), parotid gland (SUVpeak = 3.4, n = 2), and submandibular gland (SUVpeak = 4.4, n = 2). This radioactivity was markedly reduced following pretreatment with AZD2423 (3.0 mg/kg), indicating specific binding of [11C]AZD2423 to CCR2 in vivo. The presence of specific CCR2 binding was further validated using two-tissue compartment modeling, which demonstrated a 59–63% reduction in the total volume of distribution values in the analyzed peripheral tissues. Conclusions: Altogether, [11C]AZD2423 shows potential as a PET radioligand for the in vivo visualization of CCR2 expression in tissues outside the brain and may also serve as a lead compound for the further development of a CCR2 PET radioligand suitable for brain imaging. Full article
Show Figures

Graphical abstract

Back to TopTop