Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.936
Filter
1.
BMC Cancer ; 24(1): 1088, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223610

ABSTRACT

BACKGROUND: The log odds of positive lymph nodes (LODDS) was considered a superior staging system to N stage in colon cancer, yet its value in determining the optimal duration of adjuvant chemotherapy for stage III colon cancer patients has not been evaluated. This study aims to assess the prognostic value of a model that combines LODDS with clinicopathological information for stage III colon cancer patients and aims to stratify these patients using the model, identifying individuals who could benefit from varying durations of adjuvant chemotherapy. METHOD: A total of 663 consecutive patients diagnosed with stage III colon cancer, who underwent colon tumor resection between November 2007 and June 2020 at Sun Yat-sen University Cancer Center and Longyan First Affiliated Hospital of Fujian Medical University, were enrolled in this study. Survival outcomes were analyzed using Kaplan-Meier, Cox regression. Nomograms were developed to forecast patient DFS, with the Area Under the Curve (AUC) values of time-dependent Receiver Operating Characteristic (timeROC) and calibration plots utilized to assess the accuracy and reliability of the nomograms. RESULTS: Multivariate analysis revealed that perineural invasion (HR = 1.776, 95% CI: 1.052-3.003, P = 0.032), poor tumor differentiation (HR = 1.638, 95% CI: 1.084-2.475, P = 0.019), and LODDS groupings of 2 and 1 (HR = 1.920, 95% CI: 1.297-2.842, P = 0.001) were independent predictors of disease-free survival (DFS) in the training cohort. Nomograms constructed from LODDS, perineural invasion, and poor tumor differentiation demonstrated robust predictive performance for 3-year and 5-year DFS in both training (3-year AUC = 0.706, 5-year AUC = 0.678) and validation cohorts (3-year AUC = 0.744, 5-year AUC = 0.762). Stratification according to this model showed that patients in the high-risk group derived significant benefit from completing 8 cycles of chemotherapy (training cohort, 82.97% vs 67.17%, P = 0.013; validation cohort, 89.49% vs 63.97%, P = 0.030). CONCLUSION: The prognostic model, integrating LODDS, pathological differentiation, and neural invasion, demonstrates strong predictive accuracy for stage III colon cancer prognosis. Moreover, stratification via this model offers valuable insights into optimal durations of postoperative adjuvant chemotherapy.


Subject(s)
Colonic Neoplasms , Neoplasm Staging , Nomograms , Humans , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/mortality , Colonic Neoplasms/surgery , Chemotherapy, Adjuvant/methods , Female , Male , Retrospective Studies , Middle Aged , Prognosis , Aged , Lymph Nodes/pathology , Lymphatic Metastasis , Adult
2.
Front Immunol ; 15: 1455907, 2024.
Article in English | MEDLINE | ID: mdl-39247185

ABSTRACT

Pembrolizumab and other immunotherapies have become central in treating metastatic colon cancer, particularly effective in patients with mismatch repair deficiencies. We report a case involving a man who initially underwent radical surgery for sigmoid colon cancer on April 27, 2011, followed by hepatic tumor resection on September 21, 2017. Post-surgery, he received eight cycles of adjuvant chemotherapy with the CAPEOX regimen and was regularly monitored through CT and MRI scans. On August 24, 2022, liver metastases were detected, and he was diagnosed with Lynch syndrome (LS) due to germline mutation in the MSH2 and EPCAM genes. He commenced treatment with 200mg of pembrolizumab intravenously every three weeks on September 2, 2022, and demonstrated a sustained response. However, after 17 cycles, he developed a treatment related adverse event (TRAE) of pancreatic endocrine dysfunction, leading to type 1 diabetes, managed with subcutaneous insulin injections. After 30 cycles of treatment, no evidence of disease was observed. This case underscores the significant clinical benefits of first-line pembrolizumab in managing hepatic metastasis in colonic carcinoma associated with LS, despite the occurrence of TRAEs. It raises critical questions regarding the optimal duration of immunotherapy following a complete or partial response and whether treatment should be discontinued upon the emergency of TRAEs. Continued research and forthcoming clinical trials with checkpoint inhibitors are expected to refine treatment protocols for LS-associated carcinoma.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Colorectal Neoplasms, Hereditary Nonpolyposis , Liver Neoplasms , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Male , Colorectal Neoplasms, Hereditary Nonpolyposis/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/drug therapy , Antineoplastic Agents, Immunological/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Middle Aged , Treatment Outcome , MutS Homolog 2 Protein/genetics , Epithelial Cell Adhesion Molecule/genetics
3.
Ann Clin Lab Sci ; 54(4): 457-465, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39293833

ABSTRACT

OBJECTIVE: Finding methods that can interfere with Wnt/ß-catenin signaling has become an important research direction in inhibiting colon cancer metastasis. Mesoporous silica nanoparticles can efficiently carry and release drugs. Therefore, combining ligustrazine, miR-570, and mesoporous silica nanoparticles as carriers will provide a theoretical basis for development of new therapeutic strategies and drugs. METHODS: We herein prepared mesoporous silica-loaded ligustrazine nanoparticles and used them to culture HT-29 cells; we observed biological behavior of HT-29 and explored the levels of miR-570 and Wnt2/ß-catenin. RESULTS: Mesoporous silica nanoparticles loaded with Ligustrazine were successfully prepared. Ligustrazine inhibited metastasis of HT-29 cells. Mesoporous silica nanoparticles carrying ligustrazine increased the expression of miR-570 and reduced Wnt/ß-catenin in HT-29 cells. Moreover, overexpression of miR-570 inhibited HT- 29 cancer cell metastasis and Wnt/ß-catenin inhibition led to inhibition of HT-29 cell metastasis, while inhibiting miR-570 expression reversed the effect of mesoporous silica nanoparticles carrying ligustrazine, thereby accelerating HT-29 cell metastasis. CONCLUSION: miR-570 can inhibit Wnt/ß-catenin expression. Mesoporous silica nanoparticles carrying ligustrazine can promote miR-570 to inhibit Wnt/ß-catenin expression, leading to inhibition of HT029cell metastasis.


Subject(s)
Colonic Neoplasms , MicroRNAs , Nanoparticles , Pyrazines , Silicon Dioxide , Humans , Silicon Dioxide/chemistry , Pyrazines/pharmacology , Colonic Neoplasms/pathology , Colonic Neoplasms/drug therapy , Nanoparticles/chemistry , HT29 Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Porosity , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Neoplasm Metastasis , Drug Carriers/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects
4.
J Nanobiotechnology ; 22(1): 542, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39238020

ABSTRACT

Phototherapy is a promising antitumor modality, which consists of photothermal therapy (PTT) and photodynamic therapy (PDT). However, the efficacy of phototherapy is dramatically hampered by local hypoxia in tumors, overexpression of indoleamine 2,3-dioxygenase (IDO) and programmed cell death ligand-1 (PD-L1) on tumor cells. To address these issues, self-assembled multifunctional polymeric micelles (RIMNA) were developed to co-deliver photosensitizer indocyanine green (ICG), oxygenator MnO2, IDO inhibitor NLG919, and toll-like receptor 4 agonist monophosphoryl lipid A (MPLA). It is worth noting that RIMNA polymeric micelles had good stability, uniform morphology, superior biocompatibility, and intensified PTT/PDT effect. What's more, RIMNA-mediated IDO inhibition combined with programmed death receptor-1 (PD-1)/PD-L1 blockade considerably improved immunosuppression and promoted immune activation. RIMNA-based photoimmunotherapy synergized with PD-1 antibody could remarkably inhibit primary tumor proliferation, as well as stimulate the immunity to greatly suppress lung metastasis and distant tumor growth. This study offers an efficient method to reinforce the efficacy of phototherapy and alleviate immunosuppression, thereby bringing clinical benefits to cancer treatment.


Subject(s)
Colonic Neoplasms , Immunotherapy , Micelles , Phototherapy , Polymers , Programmed Cell Death 1 Receptor , Animals , Colonic Neoplasms/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Mice , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Polymers/chemistry , Cell Line, Tumor , Phototherapy/methods , Indocyanine Green/chemistry , Indocyanine Green/therapeutic use , Indocyanine Green/pharmacology , Mice, Inbred BALB C , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photochemotherapy/methods , Female , Humans , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Lipid A/analogs & derivatives
5.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225102

ABSTRACT

Tumor reliance on glycolysis is a hallmark of cancer. Immunotherapy is more effective in controlling glycolysis-low tumors lacking lactate dehydrogenase (LDH) due to reduced tumor lactate efflux and enhanced glucose availability within the tumor microenvironment (TME). LDH inhibitors (LDHi) reduce glucose uptake and tumor growth in preclinical models, but their impact on tumor-infiltrating T cells is not fully elucidated. Tumor cells have higher basal LDH expression and glycolysis levels compared with infiltrating T cells, creating a therapeutic opportunity for tumor-specific targeting of glycolysis. We demonstrate that LDHi treatment (a) decreases tumor cell glucose uptake, expression of the glucose transporter GLUT1, and tumor cell proliferation while (b) increasing glucose uptake, GLUT1 expression, and proliferation of tumor-infiltrating T cells. Accordingly, increasing glucose availability in the microenvironment via LDH inhibition leads to improved tumor-killing T cell function and impaired Treg immunosuppressive activity in vitro. Moreover, combining LDH inhibition with immune checkpoint blockade therapy effectively controls murine melanoma and colon cancer progression by promoting effector T cell infiltration and activation while destabilizing Tregs. Our results establish LDH inhibition as an effective strategy for rebalancing glucose availability for T cells within the TME, which can enhance T cell function and antitumor immunity.


Subject(s)
Glucose , L-Lactate Dehydrogenase , Tumor Microenvironment , Animals , Mice , Glucose/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/immunology , Humans , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/immunology , Glucose Transporter Type 1/genetics , Cell Line, Tumor , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Glycolysis/drug effects , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Immunotherapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
6.
Immunopharmacol Immunotoxicol ; 46(5): 715-726, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39219032

ABSTRACT

BACKGROUND: Ginseng polysaccharide (GPS) is an ingredient of ginseng with documented anti-tumor properties. However, its effect on colon cancer and the underlying molecular mechanisms have not been investigated clearly. METHODS: Cell viability of HT29 and CT26 cells treated with different concentrations of GPS was assessed using the Cell Counting Kit-8 (CCK-8) assay. Western blot assay was used to detect the expression of apoptotic proteins, while the mRNA levels were assessed by real-time quantitative polymerase chain reaction (RT-qPCR). Transwell migration assays were used to examine the migration and invasion of cells. RESULTS: The results revealed that GPS effectively suppressed the proliferation of HT29 and CT26 cells. We demonstrated an upregulation of apoptotic proteins in GPS-treated cells, including Bax, cleaved Caspase-3, and p-p53. GPS treatment also increased the mRNA levels of cytochrome C and Bax. Furthermore, the results showed that GPS treatment concurrently promoted the activation of nucleotide-binding domain leucine-rich family pyrin-containing 3 (NLRP3) inflammasome. Transwell migration assays showed that GPS inhibited the migratory and invasive abilities of colon cancer cells. As expected, inhibition of NLRP3 expression using INF39 attenuated the inhibitory effect of GPS on migration and invasion. Upon NLRP3 inhibition, GPS-induced apoptosis was dramatically alleviated, accompanied by a reduction in the expression of apoptotic proteins. CONCLUSION: In conclusion, this research provides compelling evidence that the GPS-induced NLRP3 signaling pathway plays a pivotal role in apoptosis of colon cells, suggesting potential clinical implications for the therapeutic intervention of colon cancer. Thus, GPS might be a promising anti-tumor drug for the treatment of colorectal cancer.


Subject(s)
Apoptosis , Colonic Neoplasms , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Panax , Polysaccharides , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Panax/chemistry , Apoptosis/drug effects , Polysaccharides/pharmacology , Inflammasomes/drug effects , Inflammasomes/metabolism , HT29 Cells , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor
7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(4): 482-489, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39223012

ABSTRACT

Objective To investigate the effects of pterostilbene on human colon cancer LoVo cells and study the regulatory mechanism of nuclear factor E2-related factor 2 (Nrf2) in the process of pterostilbene acting on LoVo cells. Methods LoVo cells were treated with different concentrations (5,10,20,40,60,80,100 µmol/L) of pterostilbene.Cell viability,migration,invasion,and apoptosis were examined by CCK-8,scratch,Transwell,and TUNEL assays,respectively.The mitochondrial membrane potential was measured by the mitochondrial membrane potential assay kit with JC-1.The reactive oxygen species level was measured by 2',7'-dichlorofluorescein diacetate.The protein levels of Nrf2,phosphorylated Nrf2,heme oxygenase 1,and apoptotic proteins (Bcl2 and Bax) were determined by Western blotting.In addition,cell viability,Nrf2 expression,and apoptosis rate were determined after co-application of the Nrf2-specific agonist sulforaphane. Results Compared with the control group,40,60,80,100 µmol/L pterostilbene reduced the viability of LoVo cells (P=0.014,P<0.001,P<0.001,P<0.001).Pterostilbene at 5,10,20 µmol/L did not show effects on cell viability but inhibited cell migration (P=0.008,P<0.001,P<0.001) and invasion (all P<0.001).Pterostilbene at 40,60,80 µmol/L increased apoptosis (P=0.014,P<0.001,P<0.001),promoted mitochondrial membrane potential depolarization (P=0.026,P<0.001,P<0.001) and reactive oxygen species accumulation (all P<0.001),and down-regulated the expression of phosphorylated Nrf2 (P=0.030,P<0.001,P<0.001),heme oxygenase 1 (P=0.015,P<0.001,P<0.001),and Bcl2 (P=0.039,P<0.001,P<0.001) in LoVo cells.Pterostilbene at 60,80 µmol/L down-regulated Nrf2 expression (P=0.001,P<0.001) and up-regulated Bax expression (both P<0.001).The application of sulforaphane reversed the effects of pterostilbene on cell viability (P<0.001),apoptosis (P<0.001),and Nrf2 expression (P=0.022). Conclusion Pterostilbene is a compound that can effectively inhibit colon cancer cells by inhibiting the Nrf2 pathway.


Subject(s)
Apoptosis , Colonic Neoplasms , NF-E2-Related Factor 2 , Stilbenes , Humans , Stilbenes/pharmacology , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/drug therapy , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
8.
Biomed Pharmacother ; 179: 117412, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39255734

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is an immune checkpoint that degrades L-tryptophan to kynurenine (Kyn) and enhance immunosuppression, which can be an attractive target for treating colon cancer. IDO1 inhibitors have limited efficacy when used as monotherapies, and their combination approach has been shown to provide synergistic benefits. Many studies have shown that targeting chemokines can promote the efficacy of immune checkpoint inhibitors. Therefore, this study explored the use of IDO1 inhibitors with multiple chemokines to develop a new combination regimen for IDO1 inhibitors. We found that IDO1 inhibitors reduce the secretion of C-X-C motif ligand 10(CXCL10) in cancer cells, and CXCL10 supplementation significantly improved the anticancer effect of IDO1 inhibitors. The combination of the IDO1 inhibitor with CXCL10 or its agonist axitinib had a synergistic inhibitory effect on the growth of colon cancer cells and transplanted CT26 tumors. This synergistic effect may be achieved by inhibiting cancer cell proliferation, promoting cancer cell apoptosis, promoting CD8+T cell differentiation and decreasing Tregs. Two downstream pathways of IDO1 affect CXCL10 secretion. One being the Kyn-aryl hydrocarbon receptor (AHR) pathway, the other is the general control nonderepressible 2(GCN2). Our study provides a new reference for combination regimens of IDO1 inhibitors.


Subject(s)
Cell Proliferation , Chemokine CXCL10 , Colonic Neoplasms , Drug Synergism , Indoleamine-Pyrrole 2,3,-Dioxygenase , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Chemokine CXCL10/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Animals , Cell Proliferation/drug effects , Humans , Mice , Cell Line, Tumor , Mice, Inbred BALB C , Apoptosis/drug effects , Axitinib/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD8-Positive T-Lymphocytes/drug effects , T-Lymphocytes, Regulatory/drug effects
9.
Biomolecules ; 14(9)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39334877

ABSTRACT

From the perspective of circular economy, it is extremely useful to recycle waste products for human health applications. Among the health-beneficial properties of bioactive phyto-compounds, grape pomace represents a precious source of bioactive molecules with potential antitumor properties. Here, we describe the effects of a Sicilian grape pomace hydroalcoholic extract (HE) in colon and breast cancer cells. The characterization of HE composition revealed the predominance of anthoxanthins and phenolic acids. HE treatment was more effective in reducing the viability of colon cancer cells, while breast cancer cells appeared more resistant. Indeed, while colon cancer cells underwent apoptosis, as shown by DNA fragmentation, caspase-3 activation, and PARP1 degradation, breast cancer cells seemed to not undergo apoptosis. To elucidate the underlying mechanisms, reactive oxygen species (ROS) were evaluated. Interestingly, ROS increased in both cell lines but, while in colon cancer, cells' ROS rapidly increased and progressively diminished over time, in breast cancer, cells' ROS increase was persistent up to 24 h. This effect was correlated with the induction of pro-survival autophagy, demonstrated by autophagosomes formation, autophagic markers increase, and protection by the antioxidant NAC. The autophagy inhibitor bafilomycin A1 significantly increased the HE effects in breast cancer cells but not in colon cancer cells. Overall, our data provide evidence that HE efficacy in tumor cells depends on a balance between ROS-mediated autophagy and apoptosis. Therefore, inhibiting pro-survival autophagy may be a tool to target those cells that appear more resistant to the effect of HE.


Subject(s)
Apoptosis , Autophagy , Plant Extracts , Reactive Oxygen Species , Vitis , Reactive Oxygen Species/metabolism , Humans , Apoptosis/drug effects , Autophagy/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Vitis/chemistry , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology
10.
Toxins (Basel) ; 16(9)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39330861

ABSTRACT

The human colonic commensal enterotoxigenic Bacteroides fragilis (ETBF) is associated with chronic colitis and colon cancer. ETBF colonization induces colitis via the Bacteroides fragilis toxin (BFT). BFT secreted by ETBF cause colon inflammation via E-cadherin cleavage/NF-κB signaling. ETBF promotes colon tumorigenesis via interleukin 17A (IL-17A)/CXCL-dependent inflammation, but its bioactive therapeutics in ETBF-promoted tumorigenesis remain unexplored. In the current study, we investigated the caffeic acid phenethyl ester (CAPE) in the murine model of ETBF colitis and tumorigenesis. In this study, we observed that CAPE treatment mitigated inflammation induced by ETBF in mice. Additionally, our findings indicate that CAPE treatment offers protective effects against ETBF-enhanced colon tumorigenesis in a mouse model of colitis-associated colon cancer induced by azoxymethane (AOM) and dextran sulfate sodium. Notably, the decrease in colon tumorigenesis following CAPE administration correlates with a reduction in the expression of IL-17A and CXCL1 in the gastrointestinal tract. The molecular mechanism for CAPE-induced protection against ETBF-mediated tumorigenesis is mediated by IL-17A/CXCL1, and by NF-κB activity in intestinal epithelial cells. Our findings indicate that CAPE may serve as a preventive agent against the development of ETBF-induced colitis and colorectal cancer (CRC).


Subject(s)
Bacteroides fragilis , Caffeic Acids , Colitis , Phenylethyl Alcohol , Animals , Caffeic Acids/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Bacteroides fragilis/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Mice, Inbred C57BL , Interleukin-17/metabolism , Mice , Carcinogenesis/drug effects , Chemokine CXCL1/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Colonic Neoplasms/pathology , Colonic Neoplasms/microbiology , Male , Colon/drug effects , Colon/pathology , Colon/microbiology , Colon/metabolism , Bacterial Toxins/toxicity , Disease Models, Animal , Azoxymethane/toxicity , Dextran Sulfate , Metalloendopeptidases/metabolism
11.
BMC Cancer ; 24(1): 1164, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300412

ABSTRACT

The intricate interplay of cancer stem cell plasticity, along with the bidirectional transformation between epithelial-mesenchymal states, introduces further intricacy to offer insights into newer therapeutic approaches. Differentiation therapy, while successful in targeting leukemic stem cells, has shown limited overall success, with only a few promising instances. Using colon carcinoma cell strains with sequential p53/p73 knockdowns, our study underscores the association between p53/p73 and the maintenance of cellular plasticity. Morphological alterations corresponding with cell surface marker expressions, transcriptome analysis and functional assays were performed to access stemness and EMT (Epithelial-Mesenchymal Transition) characteristics in the spectrum of cells exhibiting sequential p53 and p73 knockdowns. Notably, our investigation explores the effectiveness of esculetin in reversing the shift from an epithelial to a mesenchymal phenotype, characterized by stem cell-like traits. Esculetin significantly induces enterocyte differentiation and promotes epithelial cell polarity by altering Wnt axes in Cancer Stem Cell-like cells characterized by high mesenchymal features. These results align with our previous findings in leukemic blast cells, establishing esculetin as an effective differentiating agent in both Acute Myeloid Leukemia (AML) and solid tumor cells.


Subject(s)
Cell Differentiation , Cell Plasticity , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Neoplastic Stem Cells , Tumor Protein p73 , Tumor Suppressor Protein p53 , Umbelliferones , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Umbelliferones/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Cell Differentiation/drug effects , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , Cell Plasticity/drug effects , Cell Line, Tumor , Phenotype , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism
12.
ACS Appl Mater Interfaces ; 16(37): 49003-49012, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39226043

ABSTRACT

Due to the limited efficacy and evident side effects of traditional chemotherapy drugs attributed to their lack of specificity and selectivity, novel strategies are essential for improving cancer treatment outcomes. Here, we successfully engineered Fe3O4 magnetic nanoparticles coated with zeolitic imidazolate framework-8 (ZIF-8). The resulting nanocomposite (Fe3O4@ZIF-8) demonstrates efficient adsorption of a substantial amount of doxorubicin (DOX) due to the porous nature of ZIF-8. The drug-loaded nanoparticles, Fe3O4@ZIF-8/DOX, exhibit significant accumulation at the tumor site in SW620 colon-cancer-bearing mice when guided by an external magnetic field. Within the acidic microenvironment of the tumor, the ZIF-8 framework collapses, releasing DOX and effectively inducing tumor cell death, thereby inhibiting cancer progression while not causing undesired side effects, as confirmed by a variety of in vitro and in vivo characterizations. In comparison to free DOX, Fe3O4@ZIF-8/DOX nanoparticles show superior efficacy in colon cancer treatment. Our findings suggest that Fe3O4@ZIF-8 holds promise as a carrier for small-molecule drug adsorption and its ferromagnetic properties provide drug targeting capabilities, thereby enhancing therapeutic effects on tumors at the same drug dosage. With excellent biocompatibility, Fe3O4@ZIF-8 demonstrates potential as a drug carrier in targeted cancer chemotherapy. Our work suggests that a combination of magnetic targeting and acid-responsiveness holds great promise for advancing targeted cancer therapy in precision nanomedicine.


Subject(s)
Colonic Neoplasms , Doxorubicin , Magnetite Nanoparticles , Metal-Organic Frameworks , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Animals , Metal-Organic Frameworks/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Mice , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Drug Carriers/chemistry , Cell Line, Tumor , Zeolites/chemistry , Mice, Inbred BALB C , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Imidazoles
13.
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
14.
ACS Nano ; 18(39): 26666-26689, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39300799

ABSTRACT

The biomimetic nanoparticles (NPs) possessing abilities of tumor targeting and multimodal therapy show great potential for efficient combat of colon cancer. Herein, we developed a multifunctional biomimetic nanoplatform (Fe3O4@PDA@CaCO3-ICG@CM) based on CaCO3-modified magnetic polydopamine (PDA) loaded with indocyanine green (ICG), which was encapsulated by a mouse lymphoma cell (EL4) membrane (CM) expressing functional proteins (i.e., lymphocyte function-associated antigen 1, LFA-1; transforming growth factor-ß receptor, TGF-ßR; programmed cell death protein 1, PD-1; and factor related apoptosis ligand, FasL). Under magnetic attraction and LFA-1/PD-1-mediated endocytosis, Fe3O4@PDA@CaCO3-ICG@CM efficiently targeted CT26 colon tumor cells. The released calcium ion (Ca2+) from the NPs triggered by acidic tumor microenvironment, the enhanced photothermal effect contributed by the combination of PDA and ICG, and FasL's direct killing effect together induced tumor cells apoptosis. Moreover, the apoptosis of CT26 cells induced immunogenic cell death (ICD) to promote the maturation of dendritic cells (DCs) to activate CD4+/CD8+ T cells, thereby fighting against tumor cells, which could further be boosted by programmed death-ligand 1 (PD-L1) blockage and transforming growth factor-ß (TGF-ß) scavenging by Fe3O4@PDA@CaCO3-ICG@CM. As a result, in vivo satisfactory therapeutic effect was observed for CT26 tumor bearing-mice treated with Fe3O4@PDA@CaCO3-ICG@CM under laser irradiation and magnetic attraction, which could eradicate primary tumors and restrain distant tumors through dual tumor targeting-assisted multimodal therapy and eliciting adaptive antitumor immune response, generating the immune memory for inhibiting tumor metastasis and recurrence. Taken together, the multifunctional biomimetic nanoplatform exhibits superior antitumor effects, providing an insightful strategy for the field of nanomaterial-based treatment of cancer.


Subject(s)
Colonic Neoplasms , Indocyanine Green , Animals , Mice , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Colonic Neoplasms/drug therapy , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Indocyanine Green/therapeutic use , Indoles/chemistry , Indoles/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Apoptosis/drug effects , Polymers/chemistry , Polymers/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Calcium Carbonate/chemistry , Calcium Carbonate/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Humans , Cell Proliferation/drug effects , Multifunctional Nanoparticles/chemistry
15.
Lipids Health Dis ; 23(1): 318, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334257

ABSTRACT

BACKGROUND: Bufalin (BA) is a potent traditional Chinese medicine derived from toad venom. It has shown significant antitumor activity, but its use is limited by cardiotoxicity, which necessitates innovative delivery methods, such as rod-shaped mesoporous silica nanoparticles (rMSNs). rMSNs have been extensively employed for reducing drug toxicity and for controlled or targeted drug delivery in tumor therapy. However, their potential in delivering BA has not been completely elucidated. Therefore, in this study, BA-loaded rMSNs (BA-rMSNs) were developed to investigate their potential and mechanism in impairing colon cancer cells. METHODS: rMSNs were developed via the sol‒gel method. Drug encapsulation efficiency and loading capacity were determined to investigate the advantages of the rMSN in loading BA. The antiproliferative activities of the BA-rMSNs were investigated via 5-ethynyl-2'-deoxyuridine and CCK-8. To evaluate cell death, Annexin V-APC/PI apoptotic and calcein-AM/PI double staining were performed. Western blotting, oil red O staining, and Nile red solution were employed to determine the ability of BA-rMSNs to regulate lipophagy. RESULTS: The diameter of the BA-rMSNs was approximately 60 nm. In vitro studies demonstrated that BA-rMSNs markedly inhibited HCT 116 and HT-29 cell proliferation and induced cell death. In vivo studies revealed that BA-rMSNs reduced BA-mediated cardiotoxicity and enhanced BA tumor targeting. Mechanistic studies revealed that BA-rMSNs blocked lipophagy. CONCLUSIONS: rMSNs reduced BA-mediated cardiotoxicity and impaired the growth of colon cancer cells. Mechanistically, antitumor activity depends on lipophagy.


Subject(s)
Bufanolides , Cardiotoxicity , Colonic Neoplasms , Nanoparticles , Silicon Dioxide , Humans , Bufanolides/pharmacology , Bufanolides/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Silicon Dioxide/chemistry , Animals , Nanoparticles/chemistry , Cardiotoxicity/prevention & control , Mice , Apoptosis/drug effects , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , HCT116 Cells , HT29 Cells , Drug Carriers/chemistry , Xenograft Model Antitumor Assays
16.
Medicina (Kaunas) ; 60(8)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39202652

ABSTRACT

Background and Objectives: Colorectal cancer (CRC) poses a major global health challenge, with high incidence rates and ongoing treatment debates. Adjuvant chemotherapy benefits for high-risk subgroups, particularly stage II disease, remain controversial. This study seeks to clarify this issue by specifically examining the impact of adjuvant chemotherapy on disease-free survival (DFS) and overall survival (OS) in patients diagnosed with T4 colon cancer. Materials and Methods: This retrospective study analyzed patients undergoing radical surgery for T4 colon cancer between 2002 and 2023. Results: Our study of 184 pT4 pN0 colon cancer patients revealed that 79.3% received adjuvant chemotherapy. Multivariate analysis demonstrated significant DFS improvement: a 60% reduction in risk for those who received adjuvant therapy (0.40 95% CI: 0.25-0.62, p < 0.001). Lymphovascular invasion (LVI) and adjuvant treatment were also significantly associated with OS. Adjuvant treatment reduced mortality by 60% (HR: 0.40, 95% CI: 0.23-0.68, p = 0.001). Patients with LVI had a 1.9-fold increase in mortality (HR: 1.94, 95% CI: 1.17-3.20, p = 0.011). These findings underscore the potential value of adjuvant chemotherapy and highlight the importance of treatment completion in managing T4 colon cancer. Conclusions: Our study identifies LVI and adjuvant chemotherapy as key prognostic factors in T4 colon cancer patients. These results support the consideration of adjuvant chemotherapy in this patient population.


Subject(s)
Colonic Neoplasms , Humans , Male , Female , Chemotherapy, Adjuvant/methods , Retrospective Studies , Middle Aged , Colonic Neoplasms/drug therapy , Colonic Neoplasms/mortality , Colonic Neoplasms/surgery , Colonic Neoplasms/pathology , Aged , Neoplasm Staging , Disease-Free Survival , Adult , Aged, 80 and over , Survival Analysis
17.
Asian Pac J Cancer Prev ; 25(8): 2869-2876, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205585

ABSTRACT

Colorectal Cancer (CRC) significantly contributes to global cancer-related mortality and morbidity. Callyspongia siphonella (Callyspongia sp.), a Red Sea sponge, has shown promising activity as an anticancer extract and a source of anticancer-active compounds. This study sought to determine the effects of Callyspongia siphonella and its metabolites on HCT-116 colon cancer cells. Cell viability assays showed that Callyspongia sp. inhibited in a dose-dependent manner, the growth of HCT-116 cell lines with IC50 values of 64.8±17 ug/ml on 2D culture and 141.1±6.8 ug/ml on 3D culture. The purified compounds Sipholenol-A and Sipholenone-A have an IC50 of 48.9±2.2 uM and 47.1±1.2 uM respectively. Following Callyspongia sp. treatment of HCT-116, cell cycle analysis showed arrest at G2/M.flow cytometry analysis showed an increase in total apoptosis due to Callyspongia sp treatment. Moreover, mitochondria membrane potential has been reported to be depolarized due to Callyspongia sp. which is an extra sign of apoptosis. Further investigations are needed to explain the particular underlying mechanisms of Callyspongia sp. extract and its metabolites Sipholenol-A and Sipholenone-A to explore their therapeutic potential in treating colon cancer.


Subject(s)
Apoptosis , Callyspongia , Cell Proliferation , Colonic Neoplasms , Humans , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Callyspongia/chemistry , Antineoplastic Agents/pharmacology , Tumor Cells, Cultured , Membrane Potential, Mitochondrial/drug effects , HCT116 Cells , Cell Cycle/drug effects , Indian Ocean , Porifera/chemistry
18.
Thromb Res ; 241: 109111, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098189

ABSTRACT

Thrombosis is the primary cause of death in patients with cancer. Resveratrol inhibits platelet activation, a crucial pathophysiological basis of thrombosis, in healthy individuals. However, its effects and mechanisms of action in patients with colon cancer remain unknown. Here, we investigated the effect of resveratrol on platelet adhesion and aggregation in patients with colon cancer. Through numerous in vitro and in vivo analyses, including flow cytometry, western blotting, ELISA, and immunofluorescence and colon cancer rat models, we demonstrated that resveratrol reduced thrombosis in patients with colon cancer by inhibiting the phosphorylation of the MAPK and activating the cyclic-GMP/vasodilator-stimulated phosphoprotein pathway. These findings demonstrate the potential of resveratrol in reducing thrombosis in patients with colon cancer and could be used to develop novel therapeutic strategies for this condition.


Subject(s)
Cell Adhesion Molecules , Colonic Neoplasms , Cyclic GMP , Phosphoproteins , Platelet Activation , Resveratrol , Thrombosis , Resveratrol/pharmacology , Resveratrol/therapeutic use , Thrombosis/drug therapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/complications , Humans , Animals , Platelet Activation/drug effects , Male , Rats , Cyclic GMP/metabolism , Cell Adhesion Molecules/metabolism , Phosphoproteins/metabolism , Female , Stilbenes/pharmacology , Stilbenes/therapeutic use , Rats, Sprague-Dawley , Middle Aged , Signal Transduction/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Aggregation/drug effects , Microfilament Proteins
19.
Nanomedicine (Lond) ; 19(15): 1347-1368, 2024 06 20.
Article in English | MEDLINE | ID: mdl-39105753

ABSTRACT

The nanostructured drug-delivery systems for colon-targeted drug delivery are a promising field of research for localized diseases particularly influencing the colonic region, in other words, ulcerative colitis, Crohn's disease, and colorectal cancer. There are various drug-delivery approaches designed for effective colonic disease treatment, including stimulus-based formulations (enzyme-triggered systems, pH-sensitive systems) and magnetically driven drug-delivery systems. In addition, targeted drug delivery by means of overexpressed receptors also offers site specificity and reduces drug resistance. It also covers GI tract-triggered emulsifying systems, nontoxic plant-derived nanoformulations as advanced drug-delivery techniques as well as nanotechnology-based clinical trials toward colonic diseases. This review gives insight into advancements in colon-targeted drug delivery to meet site specificity or targeted drug-delivery requirements.


[Box: see text].


Subject(s)
Colon , Colonic Neoplasms , Drug Delivery Systems , Inflammatory Bowel Diseases , Nanomedicine , Humans , Nanomedicine/methods , Inflammatory Bowel Diseases/drug therapy , Drug Delivery Systems/methods , Colonic Neoplasms/drug therapy , Colon/drug effects , Colon/metabolism , Colon/pathology , Animals
20.
J Biochem Mol Toxicol ; 38(8): e23796, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39087923

ABSTRACT

5-fluorouracil (5-FU) is an inexpensive treatment for colon cancer; however, its efficacy is limited by chemoresistance. This study investigates the combination therapy approach of 5-FU with Sitagliptin (Sita), a diabetic drug with potential cancer-modulating effects. The combination was evaluated in vitro and in silico, focusing on the effects of Sita and 5-FU on colon cancer cells. The results showed that the addition of Sita significantly decreased the IC50 of 5-FU compared to 5-Fu monotherapy. The study also found that Sita and 5-FU interact synergistically, with a combination index below 1. Sita successfully lowered the 5-FU dosage reduction index, decreasing the expression of MDR1 mRNA and p-AKT and NFκB2 subunits p100/p52 protein. Molecular docking analyses confirmed Sita's antagonistic action on MDR1 and thymidylate synthase proteins. The study concludes that sitagliptin can target MDR1, increase apoptosis, and significantly reduce the expression of p-AKT and NFκB2 cell-survival proteins. These effects sensitize colon cancer cells to 5-FU. Repurposing sitagliptin may enhance the anticancer effects of 5-FU at lower dosages.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Colonic Neoplasms , Drug Synergism , Fluorouracil , Proto-Oncogene Proteins c-akt , Sitagliptin Phosphate , Humans , Sitagliptin Phosphate/pharmacology , Fluorouracil/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Down-Regulation/drug effects , Cell Line, Tumor , Molecular Docking Simulation
SELECTION OF CITATIONS
SEARCH DETAIL