Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.754
Filter
1.
Sci Rep ; 14(1): 23387, 2024 10 08.
Article in English | MEDLINE | ID: mdl-39379402

ABSTRACT

5-Fluorouracil (5-FU) is the leading chemotherapeutic drug used to treat hepatocellular carcinoma, one of the major cancer diseases after atherosclerosis. Because of chemo-resistance, the success rate of treatment declines with time due to continuous drug exposure. Though autophagy induction is majorly responsible for acquired resistance, the exact role of this evolutionary conserved mechanism is unknown in cancer cell survival and suppression. The usual practice involves the combinatorial use of chemotherapeutic drugs with autophagy inhibitors like Chloroquine and Bafilomycin A, while neglecting the side effects caused by autophagy impairment in healthy cells. Starvation is a well-known physiological inducer of autophagy. In this study, by caloric modulation, we tried to circumvent the resistance imposed by prolonged drug exposure and investigated the effect of 5-FU in nutrient-sufficient and deficient conditions. Our findings show a substantial correlation between autophagy and increased cancer cell death in the presence of 5-FU, with negligible effects on normal cells. Experimental data revealed that nutritional deprivation augmented cell death in the presence of 5-FU through mitochondrial membrane damage and excessive reactive oxygen species (ROS) production, initiating apoptosis. Lipidation study also unveiled that under such combinatorial treatment cellular metabolism shifts from glucose to lipid biosynthesis. Overall, our experimental findings suggest that nutritional deprivation in combination with chemotherapeutic medication can be a new effective strategy to control hepatocellular carcinoma.


Subject(s)
Apoptosis , Autophagy , Carcinoma, Hepatocellular , Fluorouracil , Liver Neoplasms , Mitochondria , Reactive Oxygen Species , Fluorouracil/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Apoptosis/drug effects , Humans , Autophagy/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Antimetabolites, Antineoplastic/pharmacology
2.
Cell Death Dis ; 15(10): 720, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353904

ABSTRACT

Nucleotide-binding oligomerization domain 2 (NOD2) is an immune sensor crucial for eliciting the innate immune responses. Nevertheless, discrepancies exist regarding the effect of NOD2 on different types of cancer. This study aimed to investigate these function of NOD2 in melanoma and its underlying mechanisms. We have validated the tumor suppressor effect of NOD2 in melanoma. NOD2 inhibited the proliferation of melanoma cells, hindering their migration and invasion while promoting the onset of apoptosis. Our study showed that NOD2 expression is closely related to pyrimidine and folate metabolism. NOD2 inhibits thymidylate synthase (TYMS) expression by promoting K48-type ubiquitination modification of TYMS, thereby decreasing the resistance of melanoma cells to 5-fluorouracil (5-FU) and capecitabine (CAP). TYMS was identified to form a complex with Polo-like Kinase 1 (PLK1) and activate the PLK1 signaling pathway. Furthermore, we revealed that the combination of the PLK1 inhibitor volasertib (BI6727) with 5-FU or CAP had a synergistic effect repressing the proliferation, migration, and autophagy of melanoma cells. Overall, our research highlights the protective role of NOD2 in melanoma and suggests that targeting NOD2 and the TYMS/PLK1 signaling axis is a high-profile therapy that could be a prospect for melanoma treatment.


Subject(s)
Cell Cycle Proteins , Cell Proliferation , Drug Resistance, Neoplasm , Melanoma , Nod2 Signaling Adaptor Protein , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Signal Transduction , Thymidylate Synthase , Humans , Protein Serine-Threonine Kinases/metabolism , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Signal Transduction/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Thymidylate Synthase/metabolism , Thymidylate Synthase/genetics , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Cell Proliferation/drug effects , Cell Movement/drug effects , Fluorouracil/pharmacology , Apoptosis/drug effects , Pteridines/pharmacology , Animals , Mice , Ubiquitination/drug effects , Autophagy/drug effects
3.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273560

ABSTRACT

Melanoma is a malignant skin cancer associated with high mortality rates and drug resistance, posing a significant threat to human health. The combination of chemotherapy and photodynamic therapy (PDT) represents a promising strategy to enhance antitumor efficacy through synergistic anti-cancer effects. Topical delivery of chemotherapeutic drugs and photosensitizers (PS) offers a non-invasive and safe way to treat melanoma. However, the effectiveness of these treatments is often hindered by challenges such as limited skin permeability and instability of the PS. In this study, transfersomes (TFS) were designed to facilitate transdermal delivery of the chemotherapeutic drug 5-Fluorouracil (5-FU) and the PS Imperatorin (IMP) for combined chemo-photodynamic therapy for melanoma. The cytotoxic and phototoxic effects of TFS-mediated PDT (TFS-UVA) were investigated in A375 cells and nude mice. The study also demonstrated that TFS-UVA generated intracellular ROS, induced G2/ M phase cell cycle arrest, and promoted cell apoptosis. In conclusion, this study indicated that 5-FU/ IMP-TFS serves as an effective transdermal therapeutic strategy for chemo-PDT in treating melanoma.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Fluorouracil , Melanoma , Photochemotherapy , Photosensitizing Agents , Photochemotherapy/methods , Animals , Humans , Apoptosis/drug effects , Melanoma/drug therapy , Melanoma/pathology , Mice , Fluorouracil/pharmacology , Fluorouracil/administration & dosage , Cell Line, Tumor , Cell Cycle Checkpoints/drug effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Mice, Nude , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , Administration, Topical , Furocoumarins/pharmacology , Furocoumarins/administration & dosage , Furocoumarins/chemistry
4.
Molecules ; 29(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39274898

ABSTRACT

Multifunctional targeted drug delivery systems have been explored as a novel cancer treatment strategy to overcome limitations of traditional chemotherapy. The combination of photodynamic therapy and chemotherapy has been shown to enhance efficacy, but the phototoxicity of traditional photosensitizers is a challenge. In this study, we prepared a multi-sensitive composite hydrogel containing gold nanoclusters (Au NCs) and the temperature-sensitive antitumor drug 5-fluorourac il (5-FU) using carboxymethyl cellulose (Carr) as a dual-functional template. Au NCs were synthesized using sodium borohydride as a reducing agent and potassium as a promoter. The resulting Au NCs were embedded in the Carr hydrogel, which was then conjugated with lactobionic acid (LA) as a targeting ligand. The resulting Au NCs/5-FU@Carr-LA composite hydrogel was used for synergistic photodynamic therapy (PDT), photothermal therapy (PTT), and chemotherapy. Au NCs/5-FU@Carr-LA releases the drug faster at pH 5.0 due to the acid sensitivity of the Carr polymer chain. In addition, at 50 °C, the release rate of Au NCs/5-FU@Carr-LA is 78.2%, indicating that the higher temperature generated by the photothermal effect is conducive to the degradation of Carr polymer chains. The Carr hydrogel stabilized the Au NCs and acted as a matrix for drug loading, and the LA ligand facilitated targeted delivery to tumor cells. The composite hydrogel exhibited excellent biocompatibility and synergistic antitumor efficacy, as demonstrated by in vitro and in vivo experiments. In addition, the hydrogel had thermal imaging capabilities, making it a promising multifunctional platform for targeted cancer therapy.


Subject(s)
Fluorouracil , Gold , Hydrogels , Metal Nanoparticles , Photochemotherapy , Gold/chemistry , Hydrogels/chemistry , Fluorouracil/pharmacology , Fluorouracil/chemistry , Animals , Metal Nanoparticles/chemistry , Mice , Humans , Photochemotherapy/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Delivery Systems , Cell Line, Tumor , Carboxymethylcellulose Sodium/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Drug Liberation , Photothermal Therapy/methods
5.
PLoS One ; 19(9): e0309014, 2024.
Article in English | MEDLINE | ID: mdl-39241034

ABSTRACT

5-Fluorouracil (5-FU) is widely used in the treatment of gastric cancer, and the emergence of drug resistance and toxic effects has limited its application. Therefore, there is an urgent need for safe and effective novel drugs or new therapies. ß-Ionone (BI) is found in vegetables and fruits and possesses an inhibitory proliferation of tumor cells in vitro and in vivo. In this study, we investigated whether BI could enhance the inhibitory effects of 5-FU on the proliferation of gastric adenocarcinoma cells and the growth of gastric cancer cell xenografts in a mouse model. The effects of BI and 5-FU alone or their combination on the cell viability, apoptosis, and mitochondrial membrane potential, the cell cycle, and its related proteins-Cyclin D1, and CDK4 as well as PCNA and GSK-3ß were evaluated in SGC-7901 cells and MKN45 cells by MTT, MB, flow cytometry and Western blot. In addition, the effects of BI and 5-FU alone or their combination on the growth of SGC-7901 cell xenografts in nude mice were investigated. The results showed that BI significantly enhanced the sensitivity of gastric adenocarcinoma cells to 5-FU in vitro and in vivo, i.e. proliferation inhibited, apoptosis induced and GSK-3ß protein activated. Therefore, our results suggest that BI increases the antitumor effect of 5-FU on gastric adenocarcinoma cells, at least partly from an activated GSK-3ß signaling pathway.


Subject(s)
Adenocarcinoma , Apoptosis , Cell Proliferation , Fluorouracil , Glycogen Synthase Kinase 3 beta , Mice, Nude , Norisoprenoids , Signal Transduction , Stomach Neoplasms , Animals , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Fluorouracil/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Cell Proliferation/drug effects , Signal Transduction/drug effects , Cell Line, Tumor , Norisoprenoids/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Mice , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Drug Synergism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Glycogen Synthase Kinase 3/metabolism , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/metabolism
6.
PLoS One ; 19(9): e0307414, 2024.
Article in English | MEDLINE | ID: mdl-39226257

ABSTRACT

Cancer continues to pose a significant global health challenge, with gastrointestinal (GI) cancers among the most prevalent and deadly forms. These cancers often lead to high mortality rates and demand the use of potent cytotoxic chemotherapeutics. For example, 5-fluorouracil (5-FU) forms the backbone of chemotherapy regimens for various GI cancers, including colorectal cancer. While these chemotherapeutics efficiently kill cancer cells, they frequently cause off-target effects such as chemotherapy-induced mucositis (CIM), characterized by debilitating symptoms like pain, nausea, and diarrhoea, necessitating medical intervention. In this study, we elucidated the potential of melatonin and misoprostol to reduce 5-FU-induced small intestinal mucositis. Morphological and cellular changes in the jejunum, along with colonic faecal water content were quantified in rats as markers for CIM. Additionally, the effects of melatonin were investigated in vitro on 5-FU treated murine intestinal organoids. The results showed that melatonin prevented villus atrophy in the rat jejunal mucosa and upheld cell viability in murine intestinal organoids. In contrast, misoprostol alone or in combination with melatonin did not significantly affect CIM caused by 5-FU. These in vivo and in vitro experiments provided promising insights that melatonin may be used as a preventive and/or adjuvant combination therapy to prevent and reduce CIM, holding the potential to enhance cancer treatment outcomes and improve patient quality-of-life.


Subject(s)
Fluorouracil , Intestine, Small , Melatonin , Mucositis , Organoids , Animals , Melatonin/pharmacology , Rats , Organoids/drug effects , Fluorouracil/adverse effects , Fluorouracil/pharmacology , Mice , Intestine, Small/drug effects , Intestine, Small/pathology , Mucositis/chemically induced , Mucositis/pathology , Mucositis/prevention & control , Mucositis/drug therapy , Male , Atrophy/chemically induced , Atrophy/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology
7.
Eur J Pharmacol ; 983: 176979, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39241942

ABSTRACT

Colon cancer is a significant health problem worldwide as it is one of the most common and deadliest cancers. The standard approach for the treatment of colon cancer is 5-fluorouracil (5-FU) based chemotherapy, which is limited by the development of resistance to this drug. Therefore, our study aimed to establish 5-FU resistance in SW-480 and HT-29 colon cancer cells and to precisely determine the molecular mechanisms and biomarkers that contribute to its development, both after short-term exposure and in cells with already developed resistance (SW-480-5FUR and HT-29-5FUR). The expression of various molecules involved in the different mechanisms of resistance development was monitored at the gene (qPCR) and protein (immunocytochemistry) levels. Based on the obtained results, alterations in the 5-FU anabolic pathway, biotransformation, drug efflux, mismatch repair, and apoptosis process together contributed to the development of 5-FU resistance in SW-480 and HT-29 colon cancer cells. In addition, UMPS, ABCC1, ABCC5, and MLH1, as well as the disturbed ratio of pro-apoptotic BAX and anti-apoptotic BCL2, should be taken into consideration as potential targets for the discovery of 5-FU resistance-related biomarkers in colon cancer cells. We suggest that future investigations focus on further validation of these findings by additional in vitro and in vivo testing, which is a limitation of our study.


Subject(s)
Colonic Neoplasms , Drug Resistance, Neoplasm , Fluorouracil , Fluorouracil/pharmacology , Humans , Drug Resistance, Neoplasm/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , HT29 Cells , Antimetabolites, Antineoplastic/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , DNA Mismatch Repair/drug effects , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism
8.
J Gastrointest Cancer ; 55(4): 1607-1619, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39240276

ABSTRACT

BACKGROUND: Drug resistance in colorectal cancer (CRC) is modulated by multiple molecular factors, which can be ascertained through genetic investigation. Single nucleotide polymorphisms (SNPs) within key genes have the potential to impair the efficacy of chemotherapeutic agents such as 5-fluorouracil (5-FU). Therefore, the identification of SNPs linked to drug resistance can significantly contribute to the advancement of tailored therapeutic approaches and the enhancement of treatment outcomes in patients with CRC. MATERIAL AND METHOD: To identify dysregulated genes in 5-FU-based chemotherapy responder or non-responder CRC patients, a meta-analysis was performed. Next, the protein-protein interaction (PPI) network of the identified genes was analyzed using the STRING database. The most significant module was chosen for further analysis. In addition, a literature review was conducted to identify drug resistance-related genes. Enrichment analysis was conducted to validate the main module genes and the genes identified from the literature review. The associations between SNPs and drug resistance were investigated, and the consequences of missense variants were assessed using in silico tools. RESULT: The meta-analysis identified 796 dysregulated genes. Then, to conduct PPI analysis and enrichment analysis, we were able to discover 23 genes that are intricately involved in the cell cycle pathway. Consequently, these 23 genes were chosen for SNP analysis. By using the dbSNP database and ANNOVAR, we successfully detected and labeled SNPs in these specific genes. Additionally, after careful exclusion of SNPs with allele frequencies below 0.01, we evaluated 6 SNPs from the HDAC1, MCM2, CDK1, BUB1B, CDC14B, and CCNE1 genes using 8 bioinformatics tools. Therefore, these SNPs were identified as potentially harmful by multiple computational tools. Specifically, rs199958833 in CDK1 (Val124Gly) was predicted to be damaging by all tools used. Our analysis strongly indicates that this specific SNP could negatively affect the stability and functionality of the CDK1 protein. CONCLUSION: Based on our current understanding, the evaluation of CDK1 polymorphisms in the context of drug resistance in CRC has yet to be undertaken. In this investigation, we showed that rs199958833 variant in the CDK1 gene may favor resistance to 5-FU-based chemotherapy. However, these findings need validation in an independent cohort of patients.


Subject(s)
Colorectal Neoplasms , Computational Biology , Drug Resistance, Neoplasm , Fluorouracil , Polymorphism, Single Nucleotide , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Fluorouracil/therapeutic use , Fluorouracil/pharmacology , Computational Biology/methods , Protein Interaction Maps/genetics , Biomarkers, Tumor/genetics , Antimetabolites, Antineoplastic/therapeutic use , Gene Expression Regulation, Neoplastic
9.
Yeast ; 41(10): 629-640, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39345013

ABSTRACT

Saccharomyces cerevisiae is an excellent model to study the effect of external cues on cell division and stress response. 5-Fluorocuracil (5-FU) has been used to treat solid tumors since several decades. The drug was initially designed to interfere with DNA replication but was later found to exert its antiproliferative effect also via RNA-dependent processes. Since 5-FU inhibits the activity of the 3'-5'-exoribonuclease Rrp6 in yeast and mammals, earlier work has compared the effect of 5-FU treatment and RRP6 deletion at the transcriptome level in diploid synchronized yeast cells. To facilitate interpreting the expression data we have developed an improved 5-Fluorouracil RNA (5-FUR) expression viewer. Users can access information via genome coordinates and systematic or standard names for mRNAs and Xrn1-dependent-, stable-, cryptic-, and meiotic unannotated transcripts (XUTs, SUTs, CUTs, and MUTs). Normalized log2-transformed or linear data can be displayed as filled diagrams, line graphs or color-coded heatmaps. The expression data are useful for researchers interested in processes such as cell cycle regulation, mitotic repression of meiotic genes, the effect of 5-FU treatment and Rrp6 deficiency on the transcriptome and expression profiles of sense/antisense loci that encode overlapping transcripts. The viewer is accessible at http://5fur.genouest.org.


Subject(s)
Fluorouracil , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Fluorouracil/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Gene Expression Regulation, Fungal/drug effects , Gene Deletion , Transcriptome , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Gene Expression Profiling , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Biochem Biophys Res Commun ; 733: 150665, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39244847

ABSTRACT

Immunogenic cell death (ICD) enhances immunogenicity and activates antitumor immune responses. ICD induction by anticancer drugs may be effective against microsatellite-stable colorectal cancers (CRCs) that are less responsive to immune checkpoint inhibitors. Calreticulin (CRT) is crucial in ICD, promoting dendritic cell phagocytosis and initiating antitumor immunity. This study investigated CRT exposure mechanisms in four CRC cell lines and three human CRC organoids. Flow cytometry and immunofluorescence showed that oxaliplatin and 5-fluorouracil caused CRT exposure in all models. Despite CRT's association with endoplasmic reticulum stress, Western blot analysis showed no increase in this stress. These findings suggest alternative pathways. RNA sequencing identified enrichment of p53 signaling pathway genes, including TP53I3, TP53INP1, and YPEL3, which were confirmed by RT-qPCR. These results suggest that the p53 signaling pathway plays an important role in CRT exposure induced by anticancer drugs.


Subject(s)
Antineoplastic Agents , Calreticulin , Colorectal Neoplasms , Signal Transduction , Tumor Suppressor Protein p53 , Humans , Calreticulin/metabolism , Calreticulin/genetics , Tumor Suppressor Protein p53/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Oxaliplatin/pharmacology , Fluorouracil/pharmacology , Endoplasmic Reticulum Stress/drug effects , Immunogenic Cell Death/drug effects
11.
Biosensors (Basel) ; 14(9)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39329810

ABSTRACT

The liver has many important functions, including the biotransformation of drugs and detoxification of the human organism. As such, it is also exposed to many harmful substances, which leads to disorders and diseases such as cirrhosis. For these reasons, it seems important to consider liver metabolism and the direct effects on the liver when evaluating the efficacy of new drugs. Accordingly, we have developed an advanced in vitro liver model using an organ-on-a-chip approach that replicates many of the morphological and functional features of the liver in vivo. The model we created can metabolize drugs, which we demonstrated using two widely used anticancer drugs, 5-fluorouracil (5FU) and capecitabine (CAP). In addition, to the best of our knowledge, we are the first who evaluate the direct effects of these drugs not only on the viability of liver model-building cells but on their functions, such as cytochrome P450 activity and albumin production. Our study brings new hope to properly evaluating drug efficacy at the in vitro level.


Subject(s)
Fluorouracil , Liver , Humans , Liver/metabolism , Fluorouracil/pharmacology , Lab-On-A-Chip Devices , Capecitabine , Cytochrome P-450 Enzyme System/metabolism , Models, Biological
12.
Theranostics ; 14(15): 5745-5761, 2024.
Article in English | MEDLINE | ID: mdl-39346545

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an invasive and rapidly progressive malignancy. A major challenge in patient management is the lack of a reliable imaging tool to monitor tumor response to treatment. Tumor-associated fibrosis characterized by high type I collagen is a hallmark of PDAC, and fibrosis further increases in response to neoadjuvant chemoradiotherapy (CRT). We hypothesized that molecular positron emission tomography (PET) using a type I collagen-specific imaging probe, 68Ga-CBP8 can detect and measure changes in tumor fibrosis in response to standard treatment in mouse models and patients with PDAC. Methods: We evaluated the specificity of 68Ga-CBP8 PET to tumor collagen and its ability to differentiate responders from non-responders based on the dynamic changes of fibrosis in nude mouse models of human PDAC including FOLFIRNOX-sensitive (PANC-1 and PDAC6) and FOLFIRINOX-resistant (SU.86.86). Next, we demonstrated the specificity and sensitivity of 68Ga-CBP8 to the deposited collagen in resected human PDAC and pancreas tissues. Eight male participant (49-65 y) with newly diagnosed PDAC underwent dynamic 68Ga-CBP8 PET/MRI, and five underwent follow up 68Ga-CBP8 PET/MRI after completing standard CRT. PET parameters were correlated with tumor collagen content and markers of response on histology. Results: 68Ga-CBP8 showed specific binding to PDAC compared to non-binding 68Ga-CNBP probe in two mouse models of PDAC using PET imaging and to resected human PDAC using autoradiography (P < 0.05 for all comparisons). 68Ga-CBP8 PET showed 2-fold higher tumor signal in mouse models following FOLFIRINOX treatment in PANC-1 and PDAC6 models (P < 0.01), but no significant increase after treatment in FOLFIRINOX resistant SU.86.86 model. 68Ga-CBP8 binding to resected human PDAC was significantly higher (P < 0.0001) in treated versus untreated tissue. PET/MRI of PDAC patients prior to CRT showed significantly higher 68Ga-CBP8 uptake in tumor compared to pancreas (SUVmean: 2.35 ± 0.36 vs. 1.99 ± 0.25, P = 0.036, n = 8). PET tumor values significantly increased following CRT compared to untreated tumors (SUVmean: 2.83 ± 0.30 vs. 2.25 ± 0.41, P = 0.01, n = 5). Collagen deposition significantly increased in response to CRT (59 ± 9% vs. 30 ± 9%, P=0.0005 in treated vs. untreated tumors). Tumor and pancreas collagen content showed a positive direct correlation with SUVmean (R2 = 0.54, P = 0.0007). Conclusions: This study demonstrates the specificity of 68Ga-CBP8 PET to tumor type I collagen and its ability to differentiate responders from non-responders based on the dynamic changes of fibrosis in PDAC. The results highlight the potential use of collagen PET as a non-invasive tool for monitoring response to treatment in patients with PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Collagen Type I , Gallium Radioisotopes , Irinotecan , Magnetic Resonance Imaging , Pancreatic Neoplasms , Positron-Emission Tomography , Animals , Humans , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Positron-Emission Tomography/methods , Mice , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Middle Aged , Male , Magnetic Resonance Imaging/methods , Collagen Type I/metabolism , Aged , Irinotecan/therapeutic use , Irinotecan/pharmacology , Mice, Nude , Fluorouracil/therapeutic use , Fluorouracil/pharmacology , Oxaliplatin/therapeutic use , Oxaliplatin/pharmacology , Cell Line, Tumor , Leucovorin/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Translational Research, Biomedical , Treatment Outcome , Fibrosis/diagnostic imaging , Radiopharmaceuticals
13.
Traffic ; 25(9): e12956, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39313937

ABSTRACT

Limited nutrient availability in the tumor microenvironment can cause the rewiring of signaling and metabolic networks to confer cancer cells with survival advantages. We show here that the limitation of glucose, glutamine and serum from the culture medium resulted in the survival of a population of cancer cells with high viability and capacity to form tumors in vivo. These cells also displayed a remarkable increase in the abundance and size of lysosomes. Moreover, lysosomes were located mainly in the perinuclear region in nutrient-limited cells; this translocation was mediated by a rapid post-transcriptional increase in the key endolysosomal trafficking protein Rab7a. The acidic lysosomes in nutrient-limited cells could trap weakly basic drugs such as doxorubicin, mediating resistance of the cells to the drug, which could be partially reversed with the lysosomal inhibitor bafilomycin A1. An in vivo chorioallantoic membrane (CAM) assay indicated a remarkable decrease in microtumor volume when nutrient-limited cells were treated with 5-Fluorouracil (5-FU) and bafilomycin A1 compared to cells treated with either agent alone. Overall, our data indicate the activation of complementary pathways with nutrient limitation that can enable cancer cells to survive, proliferate and acquire drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Lysosomes , Macrolides , rab7 GTP-Binding Proteins , Humans , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Lysosomes/metabolism , Macrolides/pharmacology , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/drug therapy , Nutrients/metabolism , rab7 GTP-Binding Proteins/metabolism
14.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39273239

ABSTRACT

5-fluorouracil (5-FU), a chemotherapeutic agent against oral squamous cell carcinoma (OSCC), is limited by poor pharmacokinetics and toxicity. The pH-sensitive zeolite imidazolate framework-8 (ZIF-8) may increase the selectivity and length of 5-FU released into the acidic tumor microenvironment. This study examined the in vitro 5-FU absorption and release profiles of ZIF-8, and then progressed to cytotoxicity assays using the OSCC primary cell line SCC7. The 5-FU loading capacity of ZIF-8 was calculated with UV-vis spectroscopy (λ = 260 nm). 5-FU release was quantified by submerging 5-FU@ZIF-8 in pH 7.4 and 5.5 acetate buffer over 48 h. For the cytotoxicity assays, 5-FU, ZIF-8, and 5-FU@ZIF-8 were added to SCC7 cultures at 25, 50, and 100 µg/mL. Cell viability was assessed through toluidine blue staining and further quantified through transcriptomic RNA sequencing. ZIF-8 stabilized at a maximum absorption of 2.71 ± 0.22 mg 5-FU, and released 0.66 mg more 5-FU at pH 5.5 than 7.4 for at least 72 h. The cytotoxicity assays showed that 5-FU@ZIF-8 had a synergistic inhibitory effect at 50 µg/mL. The RNA sequencing analysis further revealed the molecular targets of 5-FU@ZIF-8 in SCC7. 5-FU@ZIF-8 may release 5-FU based on the pH of the surrounding microenvironments and synergistically inhibit OSCC.


Subject(s)
Carcinoma, Squamous Cell , Fluorouracil , Mouth Neoplasms , Fluorouracil/pharmacology , Humans , Hydrogen-Ion Concentration , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Cell Line, Tumor , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Survival/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Zeolites/chemistry , Tumor Microenvironment/drug effects , Drug Liberation , Drug Carriers/chemistry , Drug Delivery Systems/methods , Nanoparticles/chemistry , Imidazoles
15.
Nutrients ; 16(18)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39339648

ABSTRACT

Colorectal cancer (CRC) is one of the top 10 most common cancers worldwide and caused approximately 10 million deaths in 2022. CRC mortality has increased by 10% since 2020 and 52.000 deaths will occur in 2024, highlighting the limitations of current treatments due to ineffectiveness, toxicity, or non-adherence. The widely used chemotherapeutic agent, 5-fluorouracil (5-FU), is associated with several adverse effects, including renal, cardiac, and hepatic toxicity; mucositis; and resistance. Taurine (TAU), an essential ß-amino acid with potent antioxidant, antimutagenic, and anti-inflammatory properties, has demonstrated protective effects against tissue toxicity from chemotherapeutic agents like doxorubicin and cisplatin. Taurine deficiency is linked to aging and cancers such as breast and colon cancer. This study hypothesized that TAU may mitigate the adverse effects of 5-fluorouracil (5-FU). Carcinogenesis was chemically induced in rats using 1,2-dimethylhydrazine (DMH). Following five months of cancer progression, taurine (100 mg/kg) was administered orally for 8 days, and colon tissues were analyzed. The results showed 80% of adenocarcinoma (AC) in DMH-induced control animals. Notably, the efficacy of 5-FU showed 70% AC and TAU 50% while, in the 5-FU + TAU group, no adenocarcinoma was observed. No differences were observed in the inflammatory infiltrate or the expression of genes such as K-ras, p53, and Ki-67 among the cancer-induced groups whereas APC/ß-catenin expression was increased in the 5FU + TAU-treated group. The mitotic index and dysplasia were increased in the induced 5-FU group and when associated with TAU, the levels returned to normal. These data suggest that 5-FU exhibits a synergic anticancer effect when combined with taurine.


Subject(s)
Colonic Neoplasms , Drug Synergism , Fluorouracil , Taurine , Taurine/pharmacology , Animals , Fluorouracil/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Rats , Male , Disease Models, Animal , Adenocarcinoma/drug therapy , 1,2-Dimethylhydrazine , Rats, Wistar , Antineoplastic Combined Chemotherapy Protocols/pharmacology
16.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337668

ABSTRACT

Ocular adnexal sebaceous carcinoma (SebCA) represents one of the most clinically problematic periocular tumors, often requiring aggressive surgical resection. The pathobiology of this tumor remains poorly understood, and few models exist that are suitable for preclinical testing. The aim of this study was to establish new cell lines to serve as models for pathobiological and drug testing. With patient consent, freshly resected tumor tissue was cultured using conditional reprogramming cell conditions. Standard techniques were used to characterize the cell lines in terms of overall growth, clonogenicity, apoptosis, and differentiation in vitro. Additional analyses including Western blotting, short tandem repeat (STR) profiling, and next-generation sequencing (NGS) were performed. Drug screening using mitomycin-C (MMC), 5-fluorouricil (5-FU), and 6-Diazo-5-oxo-L-norleucine (DON) were performed. JHH-SebCA01, JHH-SebCA02, and JHH-SebCA03 cell lines were established from two women and one man undergoing surgical resection of eyelid tumors. At passage 15, they each showed a doubling time of two to three days, and all could form colonies in anchorage-dependent conditions, but not in soft agar. The cells contained cytoplasmic vacuoles consistent with sebaceous differentiation, and adipophilin protein was present in all three lines. STR profiling confirmed that all lines were derived from their respective patients. NGS of the primary tumors and their matched cell lines identified numerous shared mutations, including alterations similar to those previously described in SebCA. Treatment with MMC or 5-FU resulted in dose-dependent growth inhibition and the induction of both apoptosis and differentiation. MYC protein was abundant in all three lines, and the glutamine metabolism inhibitor DON, previously shown to target high MYC tumors, slowed the growth of all our SebCA models. Ocular adnexal SebCA cell lines can be established using conditional reprogramming cell conditions, and our three new models are useful for testing therapies and interrogating the functional role of MYC and other possible molecular drivers. Current topical chemotherapies promote both apoptosis and differentiation in SebCA cells, and these tumors appear sensitive to inhibition or MYC-associated metabolic changes.


Subject(s)
Sebaceous Gland Neoplasms , Humans , Sebaceous Gland Neoplasms/pathology , Sebaceous Gland Neoplasms/metabolism , Sebaceous Gland Neoplasms/genetics , Female , Cell Line, Tumor , Male , Cell Proliferation/drug effects , Adenocarcinoma, Sebaceous/pathology , Adenocarcinoma, Sebaceous/metabolism , Adenocarcinoma, Sebaceous/genetics , Apoptosis , Mitomycin/pharmacology , Cell Differentiation , Aged , Eye Neoplasms/pathology , Eye Neoplasms/metabolism , Eye Neoplasms/genetics , Fluorouracil/pharmacology , Middle Aged
17.
ESMO Open ; 9(9): 103703, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39260162

ABSTRACT

BACKGROUND: Neoadjuvant chemotherapy is an option for patients with locally advanced rectal cancer at low risk for local recurrence. This randomized phase II trial investigated whether the addition of aflibercept to modified FOLFOX6 (mFOLFOX6) could improve the rates of centrally confirmed pathological complete remissions (pCR) and (disease-free) survival in magnetic resonance imaging (MRI)-staged cT3 rectal cancer. PATIENTS AND METHODS: Patients with rectal cancer fulfilling the following criteria were included: lower border of tumor >5 cm and <16 cm from anal verge; circumferential resection margin >2 mm and T3-tumor with a maximum infiltration of 10 mm, as determined by MRI. Patients were randomized 1 : 2 to six cycles mFOLFOX6 ± aflibercept. Surgery was scheduled 4 weeks after chemotherapy. Primary endpoint was the rate of centrally confirmed pCR. The study was designed to detect an improvement of pCR from 10% to 27% (power 80%, type I error 20%). RESULTS: A total of 119 randomized patients started treatment (39 patients mFOLFOX6, arm A, and 80 mFOLFOX + aflibercept, arm B). The incidence of all grade adverse events was similar in both arms, however, adverse events grade ≥3 were more than twice as high in the experimental arm due to hypertension. Surgical complications were comparable. Aflibercept did not improve the pCR rate (arm A 26% versus arm B 19%, P = 0.47) and more patients in arm B had node positivity. With a median follow-up of 40.1 months, the 4-year disease-free survival was 83% in arm A and 85% in arm B (P = 0.82). Only two patients in arm A and one patient in arm B developed local recurrence. CONCLUSIONS: In patients with locally advanced rectal cancer and MRI-defined low risk of local recurrence, neoadjuvant mFOLFOX6 + aflibercept was feasible and did not compromise surgery. Survival data were favorable in both arms, but pCR rates were not increased by the addition of aflibercept.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Fluorouracil , Leucovorin , Magnetic Resonance Imaging , Neoadjuvant Therapy , Organoplatinum Compounds , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins , Rectal Neoplasms , Humans , Male , Rectal Neoplasms/drug therapy , Rectal Neoplasms/diagnostic imaging , Female , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorouracil/therapeutic use , Fluorouracil/pharmacology , Leucovorin/therapeutic use , Middle Aged , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/administration & dosage , Neoadjuvant Therapy/methods , Aged , Organoplatinum Compounds/therapeutic use , Magnetic Resonance Imaging/methods , Adult , Neoplasm Staging , Disease-Free Survival
18.
J Appl Biomater Funct Mater ; 22: 22808000241284439, 2024.
Article in English | MEDLINE | ID: mdl-39323034

ABSTRACT

Nanofibrous scaffolds have emerged as promising candidates for localized drug delivery systems in the treatment of cutaneous cancers. In this study, we prepared an electrospun nanofibrous scaffold incorporating 5-fluorouracil (5-FU) and etoposide (ETP) for chemotherapy targeting melanoma cutaneous cancer. The scaffold was composed of polyvinyl alcohol (PVA) and chitosan (CS), prepared via the electrospinning process and loaded with the chemotherapeutic agents. We conducted relevant physicochemical characterizations, assessed cytotoxicity, and evaluated apoptosis against melanoma A375 cells. The prepared 5-FU/ETP co-loaded PVA/CS scaffold exhibited nanofibers (NFs) with an average diameter of 321 ± 61 nm, defect-free and homogenous morphology. FTIR spectroscopy confirmed successful incorporation of chemotherapeutics into the scaffold. Additionally, the scaffold demonstrated a hydrophilic surface, proper mechanical strength, high porosity, and efficient liquid absorption capacity. Notably, sustained and controlled drug release was observed from the nanofibrous scaffold. Furthermore, the scaffold significantly increased cytotoxicity (95%) and apoptosis (74%) in A375 melanoma cells. Consequently, the prepared 5-FU/ETP co-loaded PVA/CS nanofibrous scaffold holds promise as a valuable system for localized eradication of cutaneous melanoma tumors and mitigation of adverse drug reactions associated with chemotherapy.


Subject(s)
Etoposide , Fluorouracil , Melanoma , Nanofibers , Polyvinyl Alcohol , Fluorouracil/chemistry , Fluorouracil/pharmacology , Fluorouracil/administration & dosage , Humans , Nanofibers/chemistry , Melanoma/drug therapy , Melanoma/pathology , Cell Line, Tumor , Etoposide/chemistry , Etoposide/administration & dosage , Etoposide/pharmacology , Etoposide/pharmacokinetics , Polyvinyl Alcohol/chemistry , Tissue Scaffolds/chemistry , Chitosan/chemistry , Apoptosis/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Drug Liberation , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage
19.
J Immunother Cancer ; 12(9)2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39317456

ABSTRACT

BACKGROUND: Tumor-associated macrophages participate in the complex network of support that favors tumor growth. Among the various strategies that have been developed to target these cells, the blockade of the colony-stimulating factor 1 receptor (CSF-1R) receptor is one of the most promising ones. Here, we characterize the resulting state of human macrophages exposed to a CSF-1R kinase inhibitor. METHODS: Using RNA sequencing and metabolomics approach, we characterize the reprogramming of human monocyte-derived macrophages under CSF-1R targeting. RESULTS: We find that CSF-1R receptor inhibition in human macrophages is able to impair cholesterol synthesis, fatty acid metabolism and hypoxia-driven expression of dihydropyrimidine dehydrogenase, an enzyme responsible for the 5-fluorouracil macrophage-mediated chemoresistance. We show that this inhibition of the CSF-1R receptor leads to a downregulation of the expression of sterol regulatory element-binding protein 2, a transcription factor that controls cholesterol and fatty acid synthesis. We also show that the inhibition of extracellular signal-regulated kinase 1/2 phosphorylation resulting from targeting the CSF-1R receptor destabilizes the expression of hypoxic induced factor 2 alpha in hypoxia resulting in the downregulation of dihydropyrimidine dehydrogenase expression restoring the sensitivity to 5-fluorouracil in colorectal cancer. CONCLUSIONS: These results reveal the unexpected metabolic rewiring resulting from the CSF-1R receptor targeting of human macrophages and its potential to reverse macrophage-mediated chemoresistance in colorectal tumors.


Subject(s)
Colorectal Neoplasms , Tumor-Associated Macrophages , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Metabolic Reprogramming
20.
Cancer Med ; 13(17): e70173, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39285636

ABSTRACT

Esophageal cancer is a poor prognosis cancer characterized by intrinsic or acquired resistance to chemotherapeutic agents. The primary determinants of treatment failure are unknown. Expression of an anti-viral protein, myxovirus resistance protein A (MxA) is de-regulated in many cancers, including esophageal cancer, and its activity has been linked to apoptosis. This study has assessed whether MxA expression can influence the response of esophageal cancer cells to the chemotherapeutic agents 5-fluorouracil (5-FU) or oxaliplatin. MxA protein was differentially expressed in a panel of five esophageal cancer cell lines. KYSE450 and KYSE140 cells did not express MxA and were apoptosis incompetent. FLO-1, KYSE270, and OE21 cells expressed MxA, were more drug-sensitive and were apoptosis competent. MxA was artificially overexpressed in cell lines with no endogenous expression (KYSE450 and KYSE140). This increased the resistance of KYSE450 but not KYSE140 cells. Both cell lines remained apoptosis incompetent. We then evaluated siRNA knockdown of MxA in FLO-1 cells and CRISPR knockout in OE21 cells. Knockdown of MxA significantly increased drug sensitivity and caspase-3 activation in FLO-1 cells. OE21-MX1KO cells were also more drug-sensitive, but in contrast to FLO-1 cells, caspase-3 activation was reduced. Collectively these data indicate that MxA can promote resistance to chemotherapy, but this does not always correspond with effects on apoptosis. Effects on apoptosis are cell line specific, suggesting that other co-operating pathways determine the overall impact of MxA. Importantly, in cancer cells that overexpress the protein, drug sensitivity can be improved by interfering with MxA.


Subject(s)
Antineoplastic Agents , Apoptosis , Drug Resistance, Neoplasm , Esophageal Neoplasms , Fluorouracil , Myxovirus Resistance Proteins , Humans , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Oxaliplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL