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

Article Types

Countries / Regions

Search Results (54)

Search Parameters:
Keywords = gene–chemo combination

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 5121 KiB  
Article
Cholesterol-Conjugated Polyion Complex Nanoparticles for Combination Delivery of Hydrophobic Paclitaxel and Hydrophilic miR-34a for Colon Cancer Therapy
by Arjaree Jobdeedamrong, Hye Jin Yoo, Hosun Jung, Chiravoot Pechyen, Sitakan Natphopsuk, Peerapat Thongnuek, Seok Jeong, Junghan Lee and Su-Geun Yang
Int. J. Mol. Sci. 2025, 26(16), 7965; https://doi.org/10.3390/ijms26167965 - 18 Aug 2025
Viewed by 262
Abstract
In recent years, combination chemotherapy with therapeutic nucleic acids has emerged as a promising strategy to enhance the effectiveness of cancer therapy. However, developing an effective co-delivery system to simultaneously transport both chemotherapeutic drugs and nucleic acids remains challenging. Herein, we fabricated cholesterol-conjugated [...] Read more.
In recent years, combination chemotherapy with therapeutic nucleic acids has emerged as a promising strategy to enhance the effectiveness of cancer therapy. However, developing an effective co-delivery system to simultaneously transport both chemotherapeutic drugs and nucleic acids remains challenging. Herein, we fabricated cholesterol-conjugated polyion complex nanoparticles (PCNs) for combination delivery of hydrophobic paclitaxel (PTX) and hydrophilic miR-34a. Cholesterol was conjugated to polyethylenimine (PEI) and hyaluronic acid (HA), producing C–PEI and C–HA, respectively. PTX was initially encapsulated within the hydrophobic core formed by the self-assembly of C–HA and C–PEI, yielding polyion complex nanoparticles (PTX@C–HA/C–PEI PCNs). Subsequently, the negatively charged miR-34a was electrostatically complexed with the cationic C–PEI moieties to generate miR-34a/PTX@C–HA/C–PEI PCNs. These PCNs exhibited a nanoscale structure with a uniform size distribution and demonstrated low cytotoxicity in colon cancer cells. Fluorescence microscopy confirmed efficient cytosolic delivery of C–HA/C–PEI PCNs in colon carcinoma cells. Furthermore, combination delivery of PTX and miR-34a using C–HA/C–PEI PCNs exhibited significantly enhanced transfection efficiency and cellular uptake for human colon cancer cells. Notably, PTX/miR-34a@C–HA/C–PEI PCNs effectively downregulated critical oncogenic targets, including Notch1, Snail1, and BCL-2, resulting in reduced cancer cell migration and proliferation. These findings indicate that PTX/miR-34a@C–HA/C–PEI PCNs hold significant potential as an innovative combination delivery platform, offering improved therapeutic efficacy for colon cancer therapy. Full article
(This article belongs to the Special Issue Fundamental and Translational Insights into Colorectal Cancer)
Show Figures

Graphical abstract

18 pages, 1553 KiB  
Article
Prognostic Impact of KRAS-TP53 Co-Mutations in Patients with Early-Stage Non-Small Cell Lung Cancer: A Single-Center Retrospective Study
by Lucia Motta, Francesca Molinari, Jana Pankovics, Benjamin Pedrazzini, Alexandra Valera, Samantha Epistolio, Luca Giudici, Stefania Freguia, Miriam Patella, Martina Imbimbo, Giovanna Schiavone, Milo Frattini and Patrizia Froesch
J. Clin. Med. 2025, 14(14), 5135; https://doi.org/10.3390/jcm14145135 - 19 Jul 2025
Viewed by 621
Abstract
Background/Objectives: The clinical value of KRAS mutations in lung adenocarcinoma, alone or in combination with other mutations, has been assessed especially in advanced stages. This study evaluates how KRAS and the presence of co-mutations could affect survival in early-stage lung. Methods: [...] Read more.
Background/Objectives: The clinical value of KRAS mutations in lung adenocarcinoma, alone or in combination with other mutations, has been assessed especially in advanced stages. This study evaluates how KRAS and the presence of co-mutations could affect survival in early-stage lung. Methods: We analyzed a real-world cohort including all staged NSCLC patients diagnosed and treated from 2018 to 2022 at our Institute with availability of NGS molecular data. Statistical analyses were made using log-rank test, the two-tailed Fisher’s exact test and Kaplan-Meier survival curves. Results: KRAS mutations were observed in 179/464 cases (38.6%). The majority of KRAS co-mutations were in TP53 (74%) and STK11 (14.3%) genes. KRAS+TP53 co-mutations were more frequent compared to KRAS-only tumors in stage IV NSCLC (p = 0.01). In early stage and locally advanced cases (stage I-III), better prognosis was associated to KRAS-only mutated NSCLC and to KRAS+STK11 mutated cases compared to KRAS+TP53 (p = 0.008). In particular, patients carrying KRAS+TP53 in stage I and II displayed a shorter survival, similar to patients diagnosed at stage III. Conclusions: Routine NGS provides important information for potential actionable mutations but also for the prognostic and predictive role of the presence of co-occurring mutations. In particular, the presence of KRAS+TP53 in stage I and II NSCLC may be considered an unfavorable prognostic marker possibly leading to adapt the perioperative chemo-immunotherapy. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

24 pages, 10329 KiB  
Article
Exploring the Molecular Mechanism of 1,25(OH)2D3 Reversal of Sorafenib Resistance in Hepatocellular Carcinoma Based on Network Pharmacology and Experimental Validation
by Zhiyan Long, Xiangyi Wu, Tianxin Luo, Xiaomei Chen, Jian Huang and Shu Zhang
Curr. Issues Mol. Biol. 2025, 47(5), 319; https://doi.org/10.3390/cimb47050319 - 29 Apr 2025
Viewed by 745
Abstract
Sorafenib is currently the first-line therapeutic agent for advanced hepatocellular carcinoma (HCC). However, sorafenib resistance remains a major clinical challenge. Studies have reported that 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) can synergize with multiple chemotherapeutic drugs to enhance their antitumor efficacy, but [...] Read more.
Sorafenib is currently the first-line therapeutic agent for advanced hepatocellular carcinoma (HCC). However, sorafenib resistance remains a major clinical challenge. Studies have reported that 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) can synergize with multiple chemotherapeutic drugs to enhance their antitumor efficacy, but the combinatorial effect between 1,25(OH)2D3 and sorafenib has not yet been investigated. This study aimed to investigate the potential molecular mechanism by which 1,25(OH)2D3 reverses sorafenib resistance in hepatocellular carcinoma using network pharmacology, molecular docking, and experimental validation. We predicted a web-based pharmacological approach to predict potential targets of 1,25(OH)2D3 and its derivatives, as well as sorafenib resistance genes in hepatocellular carcinoma from public databases. We then constructed 1,25(OH)2D3 chemo-sensitizing expression profiles through intersection analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were employed to predict the potential pathways involved in 1,25(OH)2D3 chemosensitization, followed by molecular docking analysis and analysis of molecular dynamics simulations. Finally, experimental validation were conducted to elucidate the potential mechanisms by which 1,25(OH)2D3 enhances the sensitivity of HCC to sorafenib. Compound and target screening identified 730 predicted targets of 1,25(OH)2D3 and its derivatives, 1144 genes associated with sorafenib resistance in hepatocellular carcinoma, and 56 potential chemosensitization targets from the intersection analysis. KEGG analysis suggested that the chemosensitization effect of 1,25(OH)2D3 might be mediated by the FoxO signaling pathway. Molecular docking showed that both 1,25(OH)2D3 and its derivatives could stably bind to FOXO3A, a key gene in the FoxO family, and molecular dynamics simulation analysis further indicated that the two bind well together. In vitro experiments demonstrated the synergistic effects of 1,25(OH)2D3 and sorafenib, significantly inhibiting the viability and colony formation rate of sorafenib-resistant hepatocellular carcinoma cells. Additionally, the combination treatment promoted apoptosis and inhibited autophagy. Furthermore, the combination modulated the FOXO3A/FOXM1 signaling axis. This study reveals that 1,25(OH)2D3 enhances the chemosensitivity of hepatocellular carcinoma (HCC) to sorafenib, with underlying mechanisms potentially involving the targeted modulation of the FOXO3A/FOXM1 signaling axis and the reversal of sorafenib-resistant phenotypes through the regulation of apoptotic and autophagic pathways. Full article
(This article belongs to the Special Issue Molecular Biology in Drug Design and Precision Therapy)
Show Figures

Figure 1

14 pages, 4444 KiB  
Communication
A pH-Responsive Dendritic-DNA-Based Nanohydrogel for Dual Drug Delivery
by Jing Zhao, Jingyuan Wu, Yiqi Fan, Chao Yu, Le Yu and Fangwei Shao
Biomolecules 2025, 15(4), 537; https://doi.org/10.3390/biom15040537 - 6 Apr 2025
Cited by 4 | Viewed by 873
Abstract
The rational design of multifunctional drug delivery systems capable of achieving precise drug release remains a huge challenge. Herein, we designed a stimuli-responsive dendritic-DNA-based nanohydrogel as a nanocarrier to achieve the co-delivery of doxorubicin and HMGN5 mRNA-targeting antisense oligonucleotides, thus achieving dual therapeutic [...] Read more.
The rational design of multifunctional drug delivery systems capable of achieving precise drug release remains a huge challenge. Herein, we designed a stimuli-responsive dendritic-DNA-based nanohydrogel as a nanocarrier to achieve the co-delivery of doxorubicin and HMGN5 mRNA-targeting antisense oligonucleotides, thus achieving dual therapeutic effects. The nanocarrier, constructed from dendritic DNA with three crosslinking branches and one loading branch, formed biocompatible and programmable DNA nanohydrogels. The C-rich sequences in the crosslinking branches conferred pH sensitivity, while the loading strand enabled efficient incorporation of a shielding DNA/ASO complex. DOX encapsulation yielded a chemo–gene co-delivery platform. Upon cellular uptake by cancer cells, the nanocarrier disassembled in the acidic tumor microenvironment, releasing DOX for chemotherapy and ASOs via toehold-mediated strand displacement (TMSD) for targeted gene silencing. Cellular studies demonstrated significantly enhanced cancer cell inhibition compared to single-agent treatments, highlighting strong combined effects. This study provides a novel strategy for tumor-microenvironment-responsive co-delivery, enabling precise, on-demand release of therapeutic agents to enhance combined chemo–gene therapy. Full article
(This article belongs to the Section Bio-Engineered Materials)
Show Figures

Graphical abstract

20 pages, 4091 KiB  
Article
Multi-Omics Analysis Decodes Biosynthesis of Specialized Metabolites Constituting the Therapeutic Terrains of Magnolia obovata
by Megha Rai, Amit Rai, Towa Yokosaka, Tetsuya Mori, Ryo Nakabayashi, Michimi Nakamura, Hideyuki Suzuki, Kazuki Saito and Mami Yamazaki
Int. J. Mol. Sci. 2025, 26(3), 1068; https://doi.org/10.3390/ijms26031068 - 26 Jan 2025
Cited by 1 | Viewed by 1306
Abstract
Magnolia obovata is renowned for its unique bioactive constituents with medicinal properties traditionally used to treat digestive disorders, anxiety, and respiratory conditions. This study aimed to establish a comprehensive omics resource through untargeted metabolome and transcriptome profiling to explore biosynthesis of pharmacologically active [...] Read more.
Magnolia obovata is renowned for its unique bioactive constituents with medicinal properties traditionally used to treat digestive disorders, anxiety, and respiratory conditions. This study aimed to establish a comprehensive omics resource through untargeted metabolome and transcriptome profiling to explore biosynthesis of pharmacologically active compounds of M. obovata using seven tissues: young leaf, mature leaf, stem, bark, central cylinder, floral bud, and pistil. Untargeted metabolomic analysis identified 6733 mass features across seven tissues and captured chemo-diversity and its tissue-specificity in M. obovata. Through a combination of cheminformatics and manual screening approach, we confirmed the identities of 105 metabolites, including neolignans, such as honokiol and magnolol, which were found to be spatially accumulated in the bark tissue. RNA sequencing generated a comprehensive transcriptome resource, and expression analysis revealed significant tissue-specific expression patterns. Omics dataset integration identified T12 transcript module from WGCNA being correlated with the biosynthesis of magnolol and honokiol in M. obovata. Notably, phylogenetic analysis using transcripts from T12 module identified two laccase (Mo_LAC1 and Mo_LAC2) and three dirigent proteins from the DIR-b/d subfamily as potential candidate genes involved in neolignan biosynthesis. This research established omics resources of M. obovata and laid the groundwork for future studies aimed at optimizing and further understanding the biosynthesis of metabolites of therapeutic potential. Full article
(This article belongs to the Special Issue Advances in Plant Genomics and Genetics: 2nd Edition)
Show Figures

Figure 1

21 pages, 4352 KiB  
Article
PDCD10 Is a Key Player in TMZ-Resistance and Tumor Cell Regrowth: Insights into Its Underlying Mechanism in Glioblastoma Cells
by Yuan Zhu, Su Na Kim, Zhong-Rong Chen, Rainer Will, Rong-De Zhong, Philipp Dammann and Ulrich Sure
Cells 2024, 13(17), 1442; https://doi.org/10.3390/cells13171442 - 28 Aug 2024
Cited by 2 | Viewed by 1606
Abstract
Overcoming temozolomide (TMZ)-resistance is a major challenge in glioblastoma therapy. Therefore, identifying the key molecular player in chemo-resistance becomes urgent. We previously reported the downregulation of PDCD10 in primary glioblastoma patients and its tumor suppressor-like function in glioblastoma cells. Here, we demonstrate that [...] Read more.
Overcoming temozolomide (TMZ)-resistance is a major challenge in glioblastoma therapy. Therefore, identifying the key molecular player in chemo-resistance becomes urgent. We previously reported the downregulation of PDCD10 in primary glioblastoma patients and its tumor suppressor-like function in glioblastoma cells. Here, we demonstrate that the loss of PDCD10 causes a significant TMZ-resistance during treatment and promotes a rapid regrowth of tumor cells after treatment. PDCD10 knockdown upregulated MGMT, a key enzyme mediating chemo-resistance in glioblastoma, accompanied by increased expression of DNA mismatch repair genes, and enabled tumor cells to evade TMZ-induced cell-cycle arrest. These findings were confirmed in independent models of PDCD10 overexpressing cells. Furthermore, PDCD10 downregulation led to the dedifferentiation of glioblastoma cells, as evidenced by increased clonogenic growth, the upregulation of glioblastoma stem cell (GSC) markers, and enhanced neurosphere formation capacity. GSCs derived from PDCD10 knockdown cells displayed stronger TMZ-resistance and regrowth potency, compared to their parental counterparts, indicating that PDCD10-induced stemness may independently contribute to tumor malignancy. These data provide evidence for a dual role of PDCD10 in tumor suppression by controlling both chemo-resistance and dedifferentiation, and highlight PDCD10 as a potential prognostic marker and target for combination therapy with TMZ in glioblastoma. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Glioblastoma III)
Show Figures

Figure 1

16 pages, 2671 KiB  
Review
Hybrid Nanogel Drug Delivery Systems: Transforming the Tumor Microenvironment through Tumor Tissue Editing
by Theodora Katopodi, Savvas Petanidis, George Floros, Konstantinos Porpodis and Christoforos Kosmidis
Cells 2024, 13(11), 908; https://doi.org/10.3390/cells13110908 - 24 May 2024
Cited by 5 | Viewed by 2437
Abstract
The future of drug delivery offers immense potential for the creation of nanoplatforms based on nanogels. Nanogels present a significant possibility for pharmaceutical advancements because of their excellent stability and effective drug-loading capability for both hydrophobic and hydrophilic agents. As multifunctional systems, composite [...] Read more.
The future of drug delivery offers immense potential for the creation of nanoplatforms based on nanogels. Nanogels present a significant possibility for pharmaceutical advancements because of their excellent stability and effective drug-loading capability for both hydrophobic and hydrophilic agents. As multifunctional systems, composite nanogels demonstrate the capacity to carry genes, drugs, and diagnostic agents while offering a perfect platform for theranostic multimodal applications. Nanogels can achieve diverse responsiveness and enable the stimuli-responsive release of chemo-/immunotherapy drugs and thus reprogramming cells within the TME in order to inhibit tumor proliferation, progression, and metastasis. In order to achieve active targeting and boost drug accumulation at target sites, particular ligands can be added to nanogels to improve the therapeutic outcomes and enhance the precision of cancer therapy. Modern “immune-specific” nanogels also have extra sophisticated tumor tissue-editing properties. Consequently, the introduction of a multifunctional nanogel-based drug delivery system improves the targeted distribution of immunotherapy drugs and combinational therapeutic treatments, thereby increasing the effectiveness of tumor therapy. Full article
Show Figures

Figure 1

15 pages, 3264 KiB  
Article
PBMCs as Tool for Identification of Novel Immunotherapy Biomarkers in Lung Cancer
by Caterina De Rosa, Francesca Iommelli, Viviana De Rosa, Giuseppe Ercolano, Federica Sodano, Concetta Tuccillo, Luisa Amato, Virginia Tirino, Annalisa Ariano, Flora Cimmino, Gaetano di Guida, Gennaro Filosa, Alessandra di Liello, Davide Ciardiello, Erika Martinelli, Teresa Troiani, Stefania Napolitano, Giulia Martini, Fortunato Ciardiello, Federica Papaccio, Floriana Morgillo and Carminia Maria Della Corteadd Show full author list remove Hide full author list
Biomedicines 2024, 12(4), 809; https://doi.org/10.3390/biomedicines12040809 - 5 Apr 2024
Cited by 7 | Viewed by 5701
Abstract
Background: Lung cancer (LC), including both non-small (NSCLC) and small (SCLC) subtypes, is currently treated with a combination of chemo- and immunotherapy. However, predictive biomarkers to identify high-risk patients are needed. Here, we explore the role of peripheral blood mononuclear cells (PBMCs) as [...] Read more.
Background: Lung cancer (LC), including both non-small (NSCLC) and small (SCLC) subtypes, is currently treated with a combination of chemo- and immunotherapy. However, predictive biomarkers to identify high-risk patients are needed. Here, we explore the role of peripheral blood mononuclear cells (PBMCs) as a tool for novel biomarkers searching. Methods: We analyzed the expression of the cGAS-STING pathway, a key DNA sensor that activates during chemotherapy, in PBMCs from LC patients divided into best responders (BR), responders (R) and non-responders (NR). The PBMCs were whole exome sequenced (WES). Results: PBMCs from BR and R patients of LC cohorts showed the highest levels of STING (p < 0.0001) and CXCL10 (p < 0.0001). From WES, each subject had at least 1 germline/somatic alteration in a DDR gene and the presence of more DDR gene mutations correlated with clinical responses, suggesting novel biomarker implications. Thus, we tested the effect of the pharmacological DDR inhibitor (DDRi) in PBMCs and in three-dimensional spheroid co-culture of PBMCs and LC cell lines; we found that DDRi strongly increased cGAS-STING expression and tumor infiltration ability of immune cells in NR and R patients. Furthermore, we performed FACS analysis of PBMCs derived from LC patients from the BR, R and NR cohorts and we found that cytotoxic T cell subpopulations displayed the highest STING expression. Conclusions: cGAS-STING signaling activation in PBMCs may be a novel potential predictive biomarker for the response to immunotherapy and high levels are correlated with a better response to treatment along with an overall increased antitumor immune injury. Full article
(This article belongs to the Special Issue State-of-the-Art Cancer Biology and Therapeutics in Italy)
Show Figures

Figure 1

22 pages, 5602 KiB  
Review
Tissue-Based Diagnostic Biomarkers of Aggressive Variant Prostate Cancer: A Narrative Review
by Olga Kouroukli, Vasiliki Bravou, Konstantinos Giannitsas and Vasiliki Tzelepi
Cancers 2024, 16(4), 805; https://doi.org/10.3390/cancers16040805 - 16 Feb 2024
Cited by 1 | Viewed by 2709
Abstract
Prostate cancer (PC) is a common malignancy among elderly men, characterized by great heterogeneity in its clinical course, ranging from an indolent to a highly aggressive disease. The aggressive variant of prostate cancer (AVPC) clinically shows an atypical pattern of disease progression, similar [...] Read more.
Prostate cancer (PC) is a common malignancy among elderly men, characterized by great heterogeneity in its clinical course, ranging from an indolent to a highly aggressive disease. The aggressive variant of prostate cancer (AVPC) clinically shows an atypical pattern of disease progression, similar to that of small cell PC (SCPC), and also shares the chemo-responsiveness of SCPC. The term AVPC does not describe a specific histologic subtype of PC but rather the group of tumors that, irrespective of morphology, show an aggressive clinical course, dictated by androgen receptor (AR) indifference. AR indifference represents an adaptive response to androgen deprivation therapy (ADT), driven by epithelial plasticity, an inherent ability of tumor cells to adapt to their environment by changing their phenotypic characteristics in a bi-directional way. The molecular profile of AVPC entails combined alterations in the tumor suppressor genes retinoblastoma protein 1 (RB1), tumor protein 53 (TP53), and phosphatase and tensin homolog (PTEN). The understanding of the biologic heterogeneity of castration-resistant PC (CRPC) and the need to identify the subset of patients that would potentially benefit from specific therapies necessitate the development of prognostic and predictive biomarkers. This review aims to discuss the possible pathophysiologic mechanisms of AVPC development and the potential use of emerging tissue-based biomarkers in clinical practice. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

14 pages, 1753 KiB  
Systematic Review
Antiangiogenics in Malignant Granular Cell Tumors: Review of the Literature
by Carlos Torrado, Melisa Camaño, Nadia Hindi, Justo Ortega, Alberto R. Sevillano, Gema Civantos, David S. Moura, Alessandra Dimino and Javier Martín-Broto
Cancers 2023, 15(21), 5187; https://doi.org/10.3390/cancers15215187 - 28 Oct 2023
Cited by 3 | Viewed by 3182
Abstract
Granular cell tumors (GCT) represent 0.5% of all soft tissue sarcomas (STS), and when metastatic, they exhibit aggressive behavior and determine limited survival. Metastatic GCTs are relatively chemo-resistant; however, there is growing evidence of the benefit of using pazopanib and other targeted therapies [...] Read more.
Granular cell tumors (GCT) represent 0.5% of all soft tissue sarcomas (STS), and when metastatic, they exhibit aggressive behavior and determine limited survival. Metastatic GCTs are relatively chemo-resistant; however, there is growing evidence of the benefit of using pazopanib and other targeted therapies in this histology. This is a review of the role of pazopanib and other targeted therapies in the treatment of GCTs, along with some insights on pathology and molecular biology described in GCTs. From 256 articles found in our search, 10 case-report articles met the inclusion criteria. Pazopanib was the most employed systemic therapy. The median reported time on therapy with pazopanib was seven months. Eight out of ten patients (80%) experienced disease control with pazopanib, while four out of ten (40%) patients achieved an objective RECIST response. Molecular studies suggested that antitumoral effects of pazopanib in GCT might be due to a loss-of-function of ATP6AP1/2 genes which consequently enhance signaling through several molecular pathways, such as SFKs, STAT5a/b, and PDGFR-β. Other reported targeted therapies for malignant GCTs included pazopanib in combination with crizotinib, which showed disease control for four months in one patient, and a PI3K inhibitor which achieved disease control for nine months in another patient. Dasatinib and megestrol were ineffective in two other different patients. Pazopanib has been demonstrated to be active in advanced GCTs and may be considered as a preferable treatment option. Full article
(This article belongs to the Special Issue Targeted Therapy in Sarcoma)
Show Figures

Figure 1

18 pages, 4318 KiB  
Article
Revisiting Concurrent Radiation Therapy, Temozolomide, and the Histone Deacetylase Inhibitor Valproic Acid for Patients with Glioblastoma—Proteomic Alteration and Comparison Analysis with the Standard-of-Care Chemoirradiation
by Andra V. Krauze, Yingdong Zhao, Ming-Chung Li, Joanna Shih, Will Jiang, Erdal Tasci, Theresa Cooley Zgela, Mary Sproull, Megan Mackey, Uma Shankavaram, Philip Tofilon and Kevin Camphausen
Biomolecules 2023, 13(10), 1499; https://doi.org/10.3390/biom13101499 - 10 Oct 2023
Cited by 9 | Viewed by 3337
Abstract
Background: Glioblastoma (GBM) is the most common brain tumor with an overall survival (OS) of less than 30% at two years. Valproic acid (VPA) demonstrated survival benefits documented in retrospective and prospective trials, when used in combination with chemo-radiotherapy (CRT). Purpose: The primary [...] Read more.
Background: Glioblastoma (GBM) is the most common brain tumor with an overall survival (OS) of less than 30% at two years. Valproic acid (VPA) demonstrated survival benefits documented in retrospective and prospective trials, when used in combination with chemo-radiotherapy (CRT). Purpose: The primary goal of this study was to examine if the differential alteration in proteomic expression pre vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA as compared to standard-of-care CRT. The second goal was to explore the associations between the proteomic alterations in response to VPA/RT/TMZ correlated to patient outcomes. The third goal was to use the proteomic profile to determine the mechanism of action of VPA in this setting. Materials and Methods: Serum obtained pre- and post-CRT was analyzed using an aptamer-based SOMAScan® proteomic assay. Twenty-nine patients received CRT plus VPA, and 53 patients received CRT alone. Clinical data were obtained via a database and chart review. Tests for differences in protein expression changes between radiation therapy (RT) with or without VPA were conducted for individual proteins using two-sided t-tests, considering p-values of <0.05 as significant. Adjustment for age, sex, and other clinical covariates and hierarchical clustering of significant differentially expressed proteins was carried out, and Gene Set Enrichment analyses were performed using the Hallmark gene sets. Univariate Cox proportional hazards models were used to test the individual protein expression changes for an association with survival. The lasso Cox regression method and 10-fold cross-validation were employed to test the combinations of expression changes of proteins that could predict survival. Predictiveness curves were plotted for significant proteins for VPA response (p-value < 0.005) to show the survival probability vs. the protein expression percentiles. Results: A total of 124 proteins were identified pre- vs. post-CRT that were differentially expressed between the cohorts who received CRT plus VPA and those who received CRT alone. Clinical factors did not confound the results, and distinct proteomic clustering in the VPA-treated population was identified. Time-dependent ROC curves for OS and PFS for landmark times of 20 months and 6 months, respectively, revealed AUC of 0.531, 0.756, 0.774 for OS and 0.535, 0.723, 0.806 for PFS for protein expression, clinical factors, and the combination of protein expression and clinical factors, respectively, indicating that the proteome can provide additional survival risk discrimination to that already provided by the standard clinical factors with a greater impact on PFS. Several proteins of interest were identified. Alterations in GALNT14 (increased) and CCL17 (decreased) (p = 0.003 and 0.003, respectively, FDR 0.198 for both) were associated with an improvement in both OS and PFS. The pre-CRT protein expression revealed 480 proteins predictive for OS and 212 for PFS (p < 0.05), of which 112 overlapped between OS and PFS. However, FDR-adjusted p values were high, with OS (the smallest p value of 0.586) and PFS (the smallest p value of 0.998). The protein PLCD3 had the lowest p-value (p = 0.002 and 0.0004 for OS and PFS, respectively), and its elevation prior to CRT predicted superior OS and PFS with VPA administration. Cancer hallmark genesets associated with proteomic alteration observed with the administration of VPA aligned with known signal transduction pathways of this agent in malignancy and non-malignancy settings, and GBM signaling, and included epithelial–mesenchymal transition, hedgehog signaling, Il6/JAK/STAT3, coagulation, NOTCH, apical junction, xenobiotic metabolism, and complement signaling. Conclusions: Differential alteration in proteomic expression pre- vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA. Using pre- vs. post-data, prognostic proteins emerged in the analysis. Using pre-CRT data, potentially predictive proteins were identified. The protein signals and hallmark gene sets associated with the alteration in the proteome identified between patients who received VPA and those who did not, align with known biological mechanisms of action of VPA and may allow for the identification of novel biomarkers associated with outcomes that can help advance the study of VPA in future prospective trials. Full article
Show Figures

Figure 1

18 pages, 6365 KiB  
Article
Synergistic Effects of New Curcumin Analog (PAC) and Cisplatin on Oral Cancer Therapy
by Abdelhabib Semlali, Sarra Beji, Ikram Ajala, Mohammed Al-Zharani and Mahmoud Rouabhia
Curr. Issues Mol. Biol. 2023, 45(6), 5018-5035; https://doi.org/10.3390/cimb45060319 - 8 Jun 2023
Cited by 21 | Viewed by 2999
Abstract
Oral cancer has traditionally been treated with surgery, radiotherapy, chemotherapy, or a combination of these therapies. Although cisplatin, a chemotherapy drug, can effectively kill oral cancer cells by forming DNA adducts, its clinical use is limited due to adverse effects and chemo-resistance. Therefore, [...] Read more.
Oral cancer has traditionally been treated with surgery, radiotherapy, chemotherapy, or a combination of these therapies. Although cisplatin, a chemotherapy drug, can effectively kill oral cancer cells by forming DNA adducts, its clinical use is limited due to adverse effects and chemo-resistance. Therefore, there is a need to develop new, targeted anticancer drugs to complement chemotherapy, allowing for reduced cisplatin doses and minimizing adverse effects. Recent studies have shown that 3,5-Bis (4-hydroxy-3-methoxybenzylidene)-N-methyl-4-piperidine (PAC), a new curcumin analog, possesses anticancer properties and could be considered a complementary or alternative therapy. In this study, we aimed to assess the potential complementary effects of PAC in combination with cisplatin for treating oral cancer. We conducted experiments using oral cancer cell lines (Ca9-22) treated with different concentrations of cisplatin (ranging from 0.1 μM to 1 μM), either alone or in conjunction with PAC (2.5 and 5 μM). Cell growth was measured using the MTT assay, while cell cytotoxicity was evaluated using an LDH assay. Propidium iodide and annexin V staining were employed to examine the impact on cell apoptosis. Flow cytometry was used to investigate the effects of the PAC/cisplatin combination on cancer cell autophagy, oxidative stress, and DNA damage. Additionally, a Western Blot analysis was performed to assess the influence of this combination on pro-carcinogenic proteins involved in various signaling pathways. The results demonstrated that PAC enhanced the efficacy of cisplatin in a dose-dependent manner, leading to a significant inhibition of oral cancer cell proliferation. Importantly, treatment with PAC (5 μM) alongside different concentrations of cisplatin reduced the IC50 of cisplatin tenfold. Combining these two agents increased apoptosis by further inducing caspase activity. In addition, the concomitant use of PAC and cisplatin enhances oral cancer cell autophagy, ROS, and MitoSOX production. However, combined PAC with cisplatin inhibits the mitochondrial membrane potential (ΔΨm), which is a marker for cell viability. Finally, this combination further enhances the inhibition of oral cancer cell migration via the inhibition of epithelial-to-mesenchymal transition genes, such as E-cadherin. We demonstrated that the combination of PAC and cisplatin markedly enhanced oral cancer cell death by inducing apoptosis, autophagy, and oxidative stress. The data presented indicate that PAC has the potential to serve as a powerful complementary agent to cisplatin in the treatment of gingival squamous cell carcinomas. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

29 pages, 5089 KiB  
Article
Sequential Treatment with Temozolomide Plus Naturally Derived AT101 as an Alternative Therapeutic Strategy: Insights into Chemoresistance Mechanisms of Surviving Glioblastoma Cells
by Dana Hellmold, Carolin Kubelt, Tina Daunke, Silje Beckinger, Ottmar Janssen, Margarethe Hauck, Fabian Schütt, Rainer Adelung, Ralph Lucius, Jochen Haag, Susanne Sebens, Michael Synowitz and Janka Held-Feindt
Int. J. Mol. Sci. 2023, 24(10), 9075; https://doi.org/10.3390/ijms24109075 - 22 May 2023
Cited by 2 | Viewed by 2446
Abstract
Glioblastoma (GBM) is a poorly treatable disease due to the fast development of tumor recurrences and high resistance to chemo- and radiotherapy. To overcome the highly adaptive behavior of GBMs, especially multimodal therapeutic approaches also including natural adjuvants have been investigated. However, despite [...] Read more.
Glioblastoma (GBM) is a poorly treatable disease due to the fast development of tumor recurrences and high resistance to chemo- and radiotherapy. To overcome the highly adaptive behavior of GBMs, especially multimodal therapeutic approaches also including natural adjuvants have been investigated. However, despite increased efficiency, some GBM cells are still able to survive these advanced treatment regimens. Given this, the present study evaluates representative chemoresistance mechanisms of surviving human GBM primary cells in a complex in vitro co-culture model upon sequential application of temozolomide (TMZ) combined with AT101, the R(-) enantiomer of the naturally occurring cottonseed-derived gossypol. Treatment with TMZ+AT101/AT101, although highly efficient, yielded a predominance of phosphatidylserine-positive GBM cells over time. Analysis of the intracellular effects revealed phosphorylation of AKT, mTOR, and GSK3ß, resulting in the induction of various pro-tumorigenic genes in surviving GBM cells. A Torin2-mediated mTOR inhibition combined with TMZ+AT101/AT101 partly counteracted the observed TMZ+AT101/AT101-associated effects. Interestingly, treatment with TMZ+AT101/AT101 concomitantly changed the amount and composition of extracellular vesicles released from surviving GBM cells. Taken together, our analyses revealed that even when chemotherapeutic agents with different effector mechanisms are combined, a variety of chemoresistance mechanisms of surviving GBM cells must be taken into account. Full article
Show Figures

Figure 1

19 pages, 2533 KiB  
Article
RYK Gene Expression Associated with Drug Response Variation of Temozolomide and Clinical Outcomes in Glioma Patients
by Ricardo D. Gonzalez, George W. Small, Adrian J. Green, Farida S. Akhtari, Tammy M. Havener, Julia C. F. Quintanilha, Amber B. Cipriani, David M. Reif, Howard L. McLeod, Alison A. Motsinger-Reif and Tim Wiltshire
Pharmaceuticals 2023, 16(5), 726; https://doi.org/10.3390/ph16050726 - 10 May 2023
Cited by 2 | Viewed by 2844
Abstract
Temozolomide (TMZ) chemotherapy is an important tool in the treatment of glioma brain tumors. However, variable patient response and chemo-resistance remain exceptionally challenging. Our previous genome-wide association study (GWAS) identified a suggestively significant association of SNP rs4470517 in the RYK (receptor-like kinase) gene [...] Read more.
Temozolomide (TMZ) chemotherapy is an important tool in the treatment of glioma brain tumors. However, variable patient response and chemo-resistance remain exceptionally challenging. Our previous genome-wide association study (GWAS) identified a suggestively significant association of SNP rs4470517 in the RYK (receptor-like kinase) gene with TMZ drug response. Functional validation of RYK using lymphocytes and glioma cell lines resulted in gene expression analysis indicating differences in expression status between genotypes of the cell lines and TMZ dose response. We conducted univariate and multivariate Cox regression analyses using publicly available TCGA and GEO datasets to investigate the impact of RYK gene expression status on glioma patient overall (OS) and progression-free survival (PFS). Our results indicated that in IDH mutant gliomas, RYK expression and tumor grade were significant predictors of survival. In IDH wildtype glioblastomas (GBM), MGMT status was the only significant predictor. Despite this result, we revealed a potential benefit of RYK expression in IDH wildtype GBM patients. We found that a combination of RYK expression and MGMT status could serve as an additional biomarker for improved survival. Overall, our findings suggest that RYK expression may serve as an important prognostic or predictor of TMZ response and survival for glioma patients. Full article
(This article belongs to the Topic Pharmacogenetics: A Tool in Cancer Therapy)
Show Figures

Figure 1

17 pages, 47207 KiB  
Article
Mir-153-3p Modulates the Breast Cancer Cells’ Chemosensitivity to Doxorubicin by Targeting KIF20A
by Khalil Ur Rahman, Shuo Yang, Nasir Azam, Zhen Yuan, Jiawen Yu, Chunhui Zhao and Bin Feng
Cancers 2023, 15(6), 1724; https://doi.org/10.3390/cancers15061724 - 11 Mar 2023
Cited by 5 | Viewed by 2511
Abstract
Breast cancer is considered the solid tumor most sensitive to chemotherapy. However, it can become resistant to various chemotherapeutic drugs, including doxorubicin, which triggers cell death by intercalation between DNA bases, free radical formation, and topoisomerase II inhibition. When drug resistance develops, several [...] Read more.
Breast cancer is considered the solid tumor most sensitive to chemotherapy. However, it can become resistant to various chemotherapeutic drugs, including doxorubicin, which triggers cell death by intercalation between DNA bases, free radical formation, and topoisomerase II inhibition. When drug resistance develops, several miRNAs are dysregulated, suggesting that miRNAs may play a significant role in resistance formation. In the current study, we investigated how doxorubicin sensitivity of breast cancer cells is affected by miR-153-3p and its target gene. The MTT method was used to determine the chemo-sensitizing effect of miR-153-3p on doxorubicin in MCF-7 and MDA-MB-231 cell lines. Results of Western blot and dual luciferase confirmed that miR-153-3p targets KIF20A and decreases its expression. Transwell and flow cytometry experiments showed that miR-153-3p and doxorubicin together had higher effects on MCF-7 and MDA-MB-231 cell proliferation, migration, and invasion, as well as increasing apoptosis and arresting cells in the G1 phase. Proteins related to apoptosis and the cell cycle exhibited the same tendency. Intracellular vesicle formation was inhibited and RAB26 was also downregulated by treatment with miR-153-3p alone or in combination with doxorubicin. Doxorubicin’s ability to suppress tumors may be enhanced by miR-153-3p, according to in vivo studies. According to our findings, miR-153-3p has a direct effect on KIF20A and may regulate the formation of intracellular vesicles, which in turn makes breast cancer cells more susceptible to doxorubicin. Full article
(This article belongs to the Special Issue New Challenges in Breast Cancer Diagnosis and Management)
Show Figures

Figure 1

Back to TopTop