Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21.025
Filter
1.
J Ethnopharmacol ; 336: 118754, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39208999

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tubeimoside-I (TBM) promotes various cancer cell death by increasing the reactive oxygen species (ROS) production. However, the specific molecular mechanisms of TBM and its impact on oxaliplatin-mediated anti-CRC activity are not yet fully understood. AIM OF THE STUDY: To elucidate the therapeutic effect and underlying molecular mechanism of TBM on oxaliplatin-mediated anti-CRC activity. MATERIALS AND METHODS: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation, wound healing assays and flow cytometry were conducted to investigate the changes in cell phenotypes and ROS generation. Real-time quantitative PCR (qRT-PCR) and western blotting were performed to detect the expressions of related mRNA and proteins. Finally, mouse xenograft models demonstrated that synergistic anti-tumor effects of combined treatment with TBM and oxaliplatin. RESULTS: The synergistic enhancement of the anti-tumor effects of oxaliplatin in colon cancer cells by TBM involved in the regulation of ROS-mediated endoplasmic reticulum (ER) stress, C-jun-amino-terminal kinase (JNK), and p38 MAPK signaling pathways. Mechanistically, TBM increased ROS generation in colon cancer cells by inhibiting heat shock protein 60 (HSPD1) expression. Knocking down HSPD1 increased TBM-induced antitumor activity and ROS generation in colon cancer cells. The mouse xenograft tumor models further validated that the combination therapy exhibited stronger anti-tumor effects than monotherapy alone. CONCLUSIONS: Combined therapy with TBM and oxaliplatin might be an effective therapeutic strategy for some CRC patients.


Subject(s)
Colorectal Neoplasms , Drug Synergism , Endoplasmic Reticulum Stress , Oxaliplatin , Reactive Oxygen Species , Saponins , Triterpenes , Animals , Humans , Male , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Endoplasmic Reticulum Stress/drug effects , HCT116 Cells , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Mice, Nude , Oxaliplatin/pharmacology , Reactive Oxygen Species/metabolism , Saponins/pharmacology , Triterpenes/pharmacology , Xenograft Model Antitumor Assays
2.
Eur J Cancer ; 211: 114314, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39316995

ABSTRACT

INTRODUCTION: Circulating tumor (ctDNA) can be used to detect residual disease after cancer treatment. Detecting low-level ctDNA is challenging, due to the limited number of recoverable ctDNA fragments at any target loci. In response, we applied tumor-informed whole-genome sequencing (WGS), leveraging thousands of mutations for ctDNA detection. METHODS: Performance was evaluated in serial plasma samples (n = 1283) from 144 stage III colorectal cancer patients. Tumor/normal WGS was used to establish a patient-specific mutational fingerprint, which was searched for in 20x WGS plasma profiles. For reproducibility, paired aliquots of 172 plasma samples were analyzed in two independent laboratories. De novo variant calling was performed for serial plasma samples with a ctDNA level > 10 % (n = 17) to explore genomic evolution. RESULTS: WGS-based ctDNA detection was prognostic of recurrence: post-operation (Hazard ratio [HR] 6.75, 95 %CI 3.18-14.3, p < 0.001), post-adjuvant chemotherapy (HR 28.9, 95 %CI 10.1-82.8; p < 0.001), and during surveillance (HR 22.8, 95 %CI 13.7-37.9, p < 0.0001). The 3-year cumulative incidence of ctDNA detection in recurrence patients was 95 %. ctDNA was detected a median of 8.7 months before radiological recurrence. The independently analyzed plasma aliquots showed excellent agreement (Cohens Kappa=0.9, r = 0.99). Genomic characterization of serial plasma revealed significant evolution in mutations and copy number alterations, and the timing of mutational processes, such as 5-fluorouracil-induced mutations. CONCLUSION: Our study supports the use of WGS for sensitive ctDNA detection and demonstrates that post-treatment ctDNA detection is highly prognostic of recurrence. Furthermore, plasma WGS can identify genomic differences distinguishing the primary tumor and relapsing metastasis, and monitor treatment-induced genomic changes.


Subject(s)
Biomarkers, Tumor , Circulating Tumor DNA , Colorectal Neoplasms , Neoplasm Recurrence, Local , Whole Genome Sequencing , Humans , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Male , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/blood , Female , Aged , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Mutation , Neoplasm Staging , Prognosis , Adult , Aged, 80 and over
3.
Int J Nanomedicine ; 19: 9689-9705, 2024.
Article in English | MEDLINE | ID: mdl-39309187

ABSTRACT

Background: Epidermal growth factor receptor (EGFR) is a major target for the treatment of colorectal cancer. Thus, anti-EGFR antibody conjugated lipid-polymer hybrid nanoparticles can offer a potential means of enhancing the efficacy of chemotherapeutics in EGFR overexpressing cancers. In addition, the combination of chemotherapy and photothermal therapy is a promising strategy for cancer treatment. Hence, it is highly desirable to develop a safe and effective delivery system for colorectal tumor therapy. Methods: In this study, EGFR-targeted and NIR-triggered lipid-polymer hybrid nanoparticles (abbreviated as Cet-Iri-NPs) were prepared with copolymer PPG-PEG, lipids DSPE-PEG-Mal and lecithin as carriers, CPT-11 as an anticancer chemotherapeutic agent, indocyanine green (ICG) as a photothermal agent, and cetuximab as a surface-targeting ligand. Results: In vitro analyses revealed that Cet-Iri-NPs were spherical with size of 99.88 nm, charge of 29.17 mV, drug entrapment efficiency of 51.72%, and antibody conjugation efficiency of 41.70%. Meanwhile, Cet-Iri-NPs exhibited a remarkable photothermal effect, and pH/NIR-triggered faster release of CPT-11 with near infrared (NIR) laser irradiation, which induced enhanced cytotoxicity against SW480 cells. Furthermore, the promoted tumor-growth suppression effect of Cet-Iri-NPs on SW480 tumor xenograft nude mice was achieved under NIR laser irradiation. Conclusion: These results indicate that the well-defined Cet-Iri-NPs are a promising platform for targeted colorectal cancer treatment with chemo-photothermal therapy.


Subject(s)
Cetuximab , Colorectal Neoplasms , ErbB Receptors , Indocyanine Green , Irinotecan , Nanoparticles , Photothermal Therapy , ErbB Receptors/metabolism , Colorectal Neoplasms/therapy , Colorectal Neoplasms/drug therapy , Animals , Humans , Irinotecan/pharmacology , Irinotecan/chemistry , Irinotecan/pharmacokinetics , Irinotecan/administration & dosage , Cell Line, Tumor , Nanoparticles/chemistry , Cetuximab/chemistry , Cetuximab/pharmacology , Cetuximab/pharmacokinetics , Photothermal Therapy/methods , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Indocyanine Green/pharmacology , Indocyanine Green/administration & dosage , Mice , Mice, Nude , Polyethylene Glycols/chemistry , Mice, Inbred BALB C , Camptothecin/chemistry , Camptothecin/pharmacology , Camptothecin/pharmacokinetics , Camptothecin/administration & dosage , Drug Carriers/chemistry , Polymers/chemistry , Infrared Rays , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Lecithins/chemistry , Xenograft Model Antitumor Assays , Lipids/chemistry
4.
Theranostics ; 14(14): 5443-5460, 2024.
Article in English | MEDLINE | ID: mdl-39310106

ABSTRACT

Rationale : the proto-oncogene KRAS is frequently mutated in colorectal cancer (CRC), leading to inherent resistance against monoclonal antibodies targeting the epidermal growth factor receptor (EGFR), such as cetuximab. Therefore, addressing the primary resistance and expanding the indications for target therapy have become critical challenges. Methods : the screening of a natural product library against KRAS mutant CRC cells was conducted, leading to the discovery of a small molecule compound that sensitive to the KRASG13D mutation site. The anti-tumor activity of this small molecule compound in combination with cetuximab was evaluated using the KRASG13D mutant CRC models both in vivo and in vitro. This evaluation includes an examination of its effects on cell proliferation, viability, apoptosis, cell cycle progression, and tumor growth. Furthermore, RNA sequencing, western blot analysis, immunofluorescence, real-time quantitative PCR, and pull-down assays were employed to explore the molecular mechanisms underlying the synergistic anti-tumor effect of this small molecule compound in combination with cetuximab. Results : our study screened 882 compounds in KRAS mutant CRC cells and identified honokiol, a small molecule compound that exhibits specific sensitivity to KRASG13D mutant CRC cells. Furthermore, we revealed that the synergistic augmentation of cetuximab's sensitivity in vivo and in vitro models of KRASG13D mutant CRC in combination with honokiol. Mechanistically, honokiol suppresses SNX3-retromer mediated trafficking, thereby impeding lysosomal proteolytic capacity and inhibiting autophagy and macropinocytosis fluxes. Moreover, honokiol inhibits the conversion of RAS GDP to RAS GTP, heightening the susceptibility of KRASG13D CRC mutant cells to cetuximab. Conclusions : honokiol enhances the sensitivity of cetuximab by destroying SNX3 retromer in KRASG13D mutant CRC preclinical model. These findings present a promising strategy for expanding the indications of target therapy in KRAS mutant colorectal cancer patients.


Subject(s)
Apoptosis , Biphenyl Compounds , Cell Proliferation , Cetuximab , Colorectal Neoplasms , Lignans , Mutation , Proto-Oncogene Proteins p21(ras) , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Lignans/pharmacology , Lignans/therapeutic use , Cell Line, Tumor , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Mice , Cell Proliferation/drug effects , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Proto-Oncogene Mas , Drug Synergism , Mice, Nude , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Mice, Inbred BALB C , Allyl Compounds , Phenols
5.
Korean J Gastroenterol ; 84(3): 123-127, 2024 Sep 25.
Article in Korean | MEDLINE | ID: mdl-39319433

ABSTRACT

Colorectal cancer remains a significant health burden in South Korea, being the third most diagnosed cancer in the country. Despite advances in treatment, patients with metastatic colorectal cancer still face limited survival rates, with resection often deemed impossible for the majority. This review discusses the current state of chemotherapy in colorectal cancer treatment, focusing on both adjuvant chemotherapy post-surgery and palliative chemotherapy for metastatic cases. The article highlights recent updates in treatment guidelines, including the use of immunotherapy and the role of circulating tumor DNA (ctDNA) in personalized medicine. The integration of these novel approaches aims to enhance treatment efficacy, improve patient survival, and reduce recurrence rates, paving the way for more tailored and effective therapeutic strategies in colorectal cancer management.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/diagnosis , Chemotherapy, Adjuvant , Immunotherapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Circulating Tumor DNA/blood , Antineoplastic Agents/therapeutic use
6.
Medicina (Kaunas) ; 60(9)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39336420

ABSTRACT

Background and Objectives: Colorectal cancer (CRC) remains a major global health issue. Although chemotherapy is the first-line treatment, its effectiveness is limited due to drug resistance developed in CRC. To overcome resistance and improve the prognosis of CRC patients, investigating new therapeutic approaches is necessary. Materials and Methods: Using human colorectal adenocarcinoma (HT29) and metastatic CRC (SW620) cell lines, the potential anticancer properties of a newly synthesized compound 1-(Isobutyl)-3-(4-methylbenzyl) benzimidazolium chloride (IMBZC) were evaluated by performing MTT cytotoxicity, cell migration, and colony formation assays, as well as by monitoring apoptosis-related protein and gene expression using Western blot and reverse transcription-quantitative polymerase chain reaction technologies. Results: Tested at various concentrations, the half-maximal inhibitory concentrations (IC50) of IMBZC on HT29 and SW620 cell growth were determined to be 22.13 µM (6.97 µg/mL) and 15.53 µM (4.89 µg/mL), respectively. IMBZC did not alter the cell growth of normal HEK293 cell lines. In addition, IMBZC inhibited cell migration and significantly decreased colony formation, suggesting its promising role in suppressing cancer metastasis. Mechanistic analyses revealed that IMBZC treatment increased the expression of pro-apoptotic proteins p53 and Bax, while decreasing the expression of anti-apoptotic proteins Bcl-2 and Bcl-xL, thus indicating the induction of apoptosis in IMBZC-treated CRC cells, compared to untreated cells. Additionally, the addition of IMBZC to conventional chemotherapeutic drugs (i.e., 5-fluorouracil, irinotecan, and oxaliplatin) resulted in an increase in the cytotoxic potential of the drugs. Conclusions: This study suggests that IMBZC has substantial anticancer effects against CRC cells through its ability to induce apoptosis, inhibit cancer cell migration and colony formation, and enhance the cytotoxic effects of conventional chemotherapeutic drugs. These findings indicate that IMBZC could be a promising chemotherapeutic drug for the treatment of CRC. Further research should be conducted using in vivo models to confirm the anti-CRC activities of IMBZC.


Subject(s)
Antineoplastic Agents , Apoptosis , Benzimidazoles , Cell Proliferation , Colorectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Apoptosis/drug effects , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Proliferation/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Methane/pharmacology , Methane/analogs & derivatives , Methane/therapeutic use
7.
Redox Biol ; 76: 103349, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39260061

ABSTRACT

Chemoresistance remains a principal culprit for the treatment failure in colorectal cancer (CRC), especially for patients with recurrent or metastatic disease. Deciphering the molecular basis of chemoresistance may lead to novel therapeutic strategies for this fatal disease. Here, UBR5, an E3 ubiquitin ligase frequently overexpressed in human CRC, is demonstrated to mediate chemoresistance principally by inhibiting ferroptosis. Paradoxically, UBR5 shields oxaliplatin-activated Smad3 from proteasome-dependent degradation via Lys 11-linked polyubiquitination. This novel chemical modification of Smad3 facilitates the transcriptional repression of ATF3, induction of SLC7A11 and inhibition of ferroptosis, contributing to chemoresistance. Consequently, targeting UBR5 in combination with a ferroptosis inducer synergistically sensitizes CRC to oxaliplatin-induced cell death and control of tumor growth. This study reveals, for the first time, a major clinically relevant chemoresistance mechanism in CRC mediated by UBR5 in sustaining TGFß-Smad3 signaling and tuning ferroptosis, unveiling its potential as a viable therapeutic target for chemosensitization.


Subject(s)
Amino Acid Transport System y+ , Colorectal Neoplasms , Drug Resistance, Neoplasm , Ferroptosis , Signal Transduction , Smad3 Protein , Ubiquitin-Protein Ligases , Ferroptosis/drug effects , Ferroptosis/genetics , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Smad3 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Drug Resistance, Neoplasm/genetics , Signal Transduction/drug effects , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Mice , Animals , Cell Line, Tumor , Ubiquitination , Oxaliplatin/pharmacology , Ubiquitin/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Lysine/metabolism
8.
Integr Cancer Ther ; 23: 15347354241280390, 2024.
Article in English | MEDLINE | ID: mdl-39313964

ABSTRACT

BACKGROUND: In patients with metastatic colorectal cancer (mCRC), Quxie Cap-sule (QX)-a combination of conventional therapy (including chemotherapy, targeted therapy or supportive care)-has shown a significant overall survival benefit compared with placebo and might have the property of dual effects of antitumor and immunity enhancement, both mediated by the microbiome. In preclinical models, QX has also shown activity against colorectal cancer. This study aimed to describe how the aforementioned effects of QX look after when focusing on the patients in third or above line setting. METHODS: A Simon's Minimax two-stage phase II design was used in this study, which enrolled mCRC patients who progressed after second-line treatment. Patients received conventional therapy plus QX until disease progression or unacceptable toxicity. Before and after 1-month intervention, we collected patients' stool samples for microbiome analysis by 16s rRNA sequencing approaches. And the microbiome analysis before and after 1-month intervention was done through bioinformation analysis platform. RESULTS: Fifteen patients were enrolled and gut microbiome were analyzed from 7 of 10 patients that with PFS over 3.7 months. Microbiome community analysis on genus level showed that the proportion of Lachnospiraceae_UCG-001 (0.04% vs 1.06%, P = .02249) significantly increased after conventional therapy plus QX while the proportion of Alistipes (2.96% vs 1.35%, P = .03461), Flavonifractor (0.04% vs 0.02%, P = .02249), Bifidobacterium (6.11% vs 1.14%, P = .02249) and Butyricimonas (0.24% vs 0.11%, P = .03603) significantly decreased after intervention . LEfSe analysis showed that after intervention, samples were highly related with unclassified-f-lachnospiraceae, Eubacterium and Lachnospiraceae_UCG-001. CONCLUSIONS: Decrease of gut bacteria with potential roles in carcinogenesis of colorectal cancer and increase in the abundance of gut anticancer bacteria such as Lachnospiraceae may partly explain how conventional therapy combined with QX can influence carcinogenesis and tumor progression in colon cancer. TRIAL REGISTRATION: Chinese Clinical Trial Registry (ChiCTR2100053874).


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Male , Female , Middle Aged , Aged , Adult , Neoplasm Metastasis , Capsules , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Feces/microbiology
9.
Asian Pac J Cancer Prev ; 25(9): 3029-3037, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39342580

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a complex malignancy requiring multimodal treatment strategies, including neoadjuvant chemoradiation therapy (Neo-CRT), to improve patient outcomes. However, the response to Neo-CRT varies among individuals, which necessitates the development of reliable predictors of treatment response. The present study aimed to investigate the role of intravoxel incoherent motion (IVIM) and dynamic contrast-enhanced (DCE) perfusion in predicting treatment response in CRC patients after Neo-CRT. METHODS: This study was conducted on patients diagnosed with locally advanced CRC who received Neo-CRT. IVIM and DCE perfusion imaging were performed before and after CRT. Quantitative parameters, including perfusion fraction (f), diffusion coefficient (D), and transfer constant (Ktrans), were calculated from the imaging data. Treatment response was assessed based on the pathological response after surgery. Statistical data were analyzed in SPSS v. 26 using the t-test and the chi-square method. RESULTS: A total of 51 patients (female: male = 22:29, mean age = 58.14±3.49) participated in the study. Among all the patients, 15 (29.4%) cases had good responses, while 36 (70.58%) cases did not respond to treatment. All DCE parameters showed higher sensitivity and specificity than IVIM D*. Ve, Kep, and DCE Ktrans indicated significant predictive power for treatment response. Ktrans was the most accurate parameter for predicting response to treatment. Overall sensitivity and specificity of DCE were 88.8% [95% CI: 80-95.6], and 80 % [95% CI: 65-90], and those of IVIM were 65.5% and 81%, respectively. Sensitivity and specificity for DCE + IVIM were 79.5% and 93.5%, and those of DCE + IVIM + standard magnetic resonance imaging were 80.2% and 86%, respectively. CONCLUSION: IVIM and DCE perfusion imaging could serve as promising tools for predicting treatment response in CRC patients after Neo-CRT.


Subject(s)
Colorectal Neoplasms , Contrast Media , Neoadjuvant Therapy , Humans , Female , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/diagnostic imaging , Male , Middle Aged , Neoadjuvant Therapy/methods , Prognosis , Follow-Up Studies , Perfusion Imaging/methods , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Motion , Magnetic Resonance Imaging/methods
10.
J Immunother Cancer ; 12(9)2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39343509

ABSTRACT

BACKGROUND: The development and progression of colorectal cancer (CRC) are influenced by the gut environment, much of which is modulated by microbial-derived metabolites. Although some research has been conducted on the gut microbiota, there have been limited empirical investigations on the role of the microbial-derived metabolites in CRC. METHODS: In this study, we used LC-MS and 16S rRNA sequencing to identify gut microbiome-associated fecal metabolites in patients with CRC and healthy controls. Moreover, we examined the effects of Faecalibacterium prausnitzii and tyrosol on CRC by establishing orthotopic and subcutaneous tumor mouse models. Additionally, we conducted in vitro experiments to investigate the mechanism through which tyrosol inhibits tumor cell growth. RESULTS: Our study revealed changes in the gut microbiome and metabolome that are linked to CRC. We observed that Faecalibacterium prausnitzii, a bacterium known for its multiple anti-CRC properties, is significantly more abundant in the intestines of healthy individuals than in those of individuals with CRC. In mouse tumor models, our study illustrated that Faecalibacterium prausnitzii has the ability to inhibit tumor growth by reducing inflammatory responses and enhancing tumor immunity. Additionally, research investigating the relationship between CRC-associated features and microbe-metabolite interactions revealed a correlation between Faecalibacterium prausnitzii and tyrosol, both of which are less abundant in the intestines of tumor patients. Tyrosol demonstrated antitumor activity in vivo and specifically targeted CRC cells without affecting intestinal epithelial cells in cell experiments. Moreover, tyrosol treatment effectively reduced the levels of reactive oxygen species (ROS) and inflammatory cytokines in MC38 cells. Western blot analysis further revealed that tyrosol inhibited the activation of the NF-κB and HIF-1 signaling pathways. CONCLUSIONS: This study investigated the relationship between CRC development and changes in the gut microbiota and microbial-derived metabolites. Specifically, the intestinal metabolite tyrosol exhibits antitumor effects by inhibiting HIF-1α/NF-κB signaling pathway activation, leading to a reduction in the levels of ROS and inflammatory factors. These findings indicate that manipulating the gut microbiota and its metabolites could be a promising approach for preventing and treating CRC and could provide insights for the development of anticancer drugs.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Hypoxia-Inducible Factor 1, alpha Subunit , NF-kappa B , Phenylethyl Alcohol , Signal Transduction , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Gastrointestinal Microbiome/drug effects , Humans , Animals , Mice , Signal Transduction/drug effects , NF-kappa B/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Female , Cell Line, Tumor
11.
J Nanobiotechnology ; 22(1): 592, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39343911

ABSTRACT

Immunotherapy has shown marked progress in promoting systemic anti-colorectal cancer (CRC) clinical effects. For further effectively sensitizing CRC to immunotherapy, we have engineered a pH-sensitive zeolitic imidazolate framework-8 (CS/NPs), capable of efficient cGAS-STING pathway activation and immune checkpoint blockade, by encapsulating the chemotherapeutic mitoxantrone (MTX) and immunomodulator thymus pentapeptide (TP5) and tailoring with tumor-targeting chondroitin sulfate (CS). In this nanoframework, CS endows CS/NPs with specific tumor-targeting activity and reduced systemic toxicity. Of note, the coordinated Zn2+ disrupts glycolytic processes and downregulates the expression of glucose transporter type 1 (GLUT1), thus depriving the cancer cells of their energy. Zn2+ further initiates the adenosine 5'-monophosphate activated protein kinase (AMPK) pathway, which leads to PD-L1 protein degradation and sensitizes CRC cells to immunotherapy. Moreover, the damaged double-stranded DNA during MTX treatment activates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which works together with TP5 induced the proliferation and differentiation of T lymphocytes and dendritic cells to further enhance the anti-CRC immune response. Therefore, CS/NPs efficiently sensitize cells to chemotherapy and stimulate systemic antitumor immune responses both in vitro and in vivo, representing a promising strategy to increase the feasibility of CRC immunotherapy.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Immunotherapy , Membrane Proteins , Metal-Organic Frameworks , Mitoxantrone , Nucleotidyltransferases , Colorectal Neoplasms/drug therapy , Immunotherapy/methods , Animals , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Mice , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Mitoxantrone/pharmacology , Mitoxantrone/chemistry , Immune Checkpoint Inhibitors/pharmacology , Cell Line, Tumor , Signal Transduction/drug effects , Mice, Inbred BALB C , B7-H1 Antigen/metabolism , Female , Imidazoles
12.
J Evid Based Med ; 17(3): 667-674, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39327543

ABSTRACT

BACKGROUND: This study aims to compare the effectiveness and safety of the combination of raltitrexed, S-1 (RS), and fruquintinib with the combination of RS and bevacizumab in patients with refractory metastatic colorectal cancer (mCRC). METHODS: This retrospective cohort included mCRC patients who received the RS plus fruquintinib or regorafenib as the third-line therapy from May 2019 to April 2023. A propensity score matching (PSM) analysis was used to balance the baseline characteristics of all patients. Overall survival (OS), progression-free survival (PFS), tumor response, and safety of the two regimens were evaluated. RESULTS: Of the 153 patients enrolled, 123 patients received the RS plus bevacizumab and 30 patients received the RS plus fruquintinib. After PSM, 30 pairs were analyzed. Patients treated with RS plus fruquintinib had a longer PFS than those treated with RS plus bevacizumab before PSM (5.0 months vs. 4.3 months, p = 0.008) and after PSM (5.0 months vs. 4.4 months, p = 0.012). A longer OS was also observed in RS plus fruquintinib group before PSM and after PSM, but there was no statistic difference between two groups after PSM. Both objective response rate and disease control rate were higher in the RS plus fruquintinib cohort than those in the RS plus bevacizumab cohort before PSM, and the difference in values between the two groups reduced after PSM. The adverse effects (AEs) of both groups were well tolerated. CONCLUSION: In patients with refractory mCRC, RS plus fruquintinib demonstrated a superior OS, PFS than RS plus bevacizumab and had manageable AEs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Bevacizumab , Colorectal Neoplasms , Drug Combinations , Propensity Score , Quinazolines , Tegafur , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Bevacizumab/administration & dosage , Bevacizumab/therapeutic use , Bevacizumab/adverse effects , Male , Female , Retrospective Studies , Middle Aged , Quinazolines/therapeutic use , Quinazolines/adverse effects , Quinazolines/administration & dosage , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Tegafur/administration & dosage , Tegafur/therapeutic use , Tegafur/adverse effects , Oxonic Acid/administration & dosage , Oxonic Acid/therapeutic use , Oxonic Acid/adverse effects , Benzofurans/therapeutic use , Benzofurans/administration & dosage , Benzofurans/adverse effects , Adult , Progression-Free Survival , Thiophenes
13.
Pathol Res Pract ; 262: 155520, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39217771

ABSTRACT

Carcinoma of the colon and rectum, also known as colorectal cancer, ranks as the third most frequently diagnosed malignancy globally. Sorafenib exhibits broad-spectrum antitumor activity against Raf, VEGF, and PDGF pathways in hepatocellular, thyroid, and renal cancers, but faces resistance in colorectal malignancies. 6-Shogaol, a prominent natural compound found in Zingiberaceae, exhibits antioxidant, anti-inflammatory, anticancer, and antiemetic properties. We investigated the influence of 6-shogaol on sorafenib's cytotoxic profile against colorectal cancer cell lines (HT-29, HCT-116, CaCo-2, and LS174T) through its effects on cellular accumulation and metabolism. Cytotoxicity was assessed using the sulpharodamine B assay, caspase-3 and c-PARP cleavage, cell cycle distribution analysis, and P-gp efflux activity. 6-Shogoal showed considerable cytotoxicity with decreased IC50 in colorectal cancer cell lines. Combining sorafenib and 6-shogaol increased c-PARP and pro-caspase-3 concentrations in HCT-116 cells compared to sorafenib alone. In combination, pro-caspase-3 concentrations were decreased in CaCo-2 cells compared to alone. Sorafenib combinations with 6-shogaol showed a significant drop in cell cycle distribution from 16.96±1.10 % to 9.16±1.85 %, respectively. At 100 µM, sorafenib and 6-shogaol showed potent and significant activity with intra-cellular rhodamine concentration on P-gp efflux activity in CRC cell lines. In conclusion, 6-shogaol substantially improved the cytotoxic profile of sorafenib by affecting its cellular uptake and metabolism. Future research should focus on dosage optimization and formulation and evaluate the efficacy and safety of the combination in animal models with colorectal cancer.


Subject(s)
Catechols , Colorectal Neoplasms , Sorafenib , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Sorafenib/pharmacology , Catechols/pharmacology , Antineoplastic Agents/pharmacology , Drug Synergism , Apoptosis/drug effects , Cell Line, Tumor , Caco-2 Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
14.
Sci Rep ; 14(1): 20323, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39223184

ABSTRACT

Metal oxide nanoparticles(NPs) contain unique properties which have made them attractive agents in cancer treatment. The CuO nanoparticles were green synthesized using walnut shell powder in different calcination temperatures (400°, 500°, 700°, and 900 °C). The CuO nanoparticles are characterized by FTIR, XRD, BET, SEM and DLS analyses. SEM and DLS analyses showed that by increasing the required calcination temperature for synthesizing the NPs, their size was increased. DPPH analysis displayed no significant anti-oxidative properties of the CuO NPs. The MTT analysis showed that all synthesized CuO NPs exhibited cytotoxic effects on MCF-7, HCT-116, and HEK-293 cell lines. Among the CuO NPs, the CuO-900 NPs showed the least cytotoxic effect on the HEK-293 cell line (IC50 = 330.8 µg/ml). Hoechst staining and real-time analysis suggested that the CuO-900 NPs induced apoptosis by elevation of p53 and Bax genes expression levels. Also, the CuO-900 NPs increased the Nrf-2 gene expression level in MCF-7 cells, despite the HCT-116 cells. As can be concluded from the results, the CuO-900 NPs exerted promising cytotoxic effects on breast and colon cancer cells.


Subject(s)
Antineoplastic Agents , Apoptosis , Breast Neoplasms , Colorectal Neoplasms , Copper , Green Chemistry Technology , Juglans , Metal Nanoparticles , Humans , Copper/chemistry , Copper/pharmacology , Juglans/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Metal Nanoparticles/chemistry , MCF-7 Cells , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , HCT116 Cells , Female , HEK293 Cells , Particle Size , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Cell Survival/drug effects
15.
Cancer Med ; 13(17): e70171, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39254067

ABSTRACT

PURPOSE: Therapeutic management of colorectal cancer (CRC) does not yet yield promising long-term results. Therefore, there is a need for further investigation of possible therapeutic options. Various experiments have studied the effects of apigenin on CRC and have shown conflicting results. This systematic review and meta-analysis investigates the currently existing evidence on the effect of apigenin on CRC. METHODS: Medline, Embase, Scopus, and Web of Science databases were searched for articles related to apigenin and its effect on CRC in the preclinical setting. Cell viability, growth inhibition, apoptosis, and cell cycle arrest for in-vitro, and body weight, tumor size, and mortality in in-vivo studies were extracted as outcomes. RESULTS: Thirty-nine articles investigating colorectal adenocarcinoma were included in this meta-analysis. Thirty-seven of these studies had data for in vitro experiments, with eight studies having data for in vivo experiments. Six articles had both in vitro and in vivo assessments. Our analysis showed apigenin reduces cell viability and induces growth inhibition, apoptosis, and cell cycle arrest in in vitro studies. The few in vivo studies indicate that apigenin decreases tumor size while showing no effects on the body weight of animal colorectal adenocarcinoma models. CONCLUSION: Our results demonstrated that apigenin, through reducing cell viability, inducing growth inhibition, apoptosis, and cell cycle arrest, and also by decreasing the tumor size, can be considered as a possible adjuvant agent in the management of colorectal adenocarcinoma. However, further in vivo studies are needed before any efforts to translate the current evidence into clinical studies.


Subject(s)
Adenocarcinoma , Apigenin , Colorectal Neoplasms , Animals , Humans , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Apigenin/pharmacology , Apigenin/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology
16.
Lancet ; 404(10458): 1107-1118, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39306468

ABSTRACT

BACKGROUND: Despite the increasing efficacy of chemotherapy, permanently unresectable colorectal liver metastases are associated with poor long-term survival. We aimed to assess whether liver transplantation plus chemotherapy could improve overall survival. METHODS: TransMet was a multicentre, open-label, prospective, randomised controlled trial done in 20 tertiary centres in Europe. Patients aged 18-65 years, with Eastern Cooperative Oncology Group performance score 0-1, permanently unresectable colorectal liver metastases from resected BRAF-non-mutated colorectal cancer responsive to systemic chemotherapy (≥3 months, ≤3 lines), and no extrahepatic disease, were eligible for enrolment. Patients were randomised (1:1) to liver transplantation plus chemotherapy or chemotherapy alone, using block randomisation. The liver transplantation plus chemotherapy group underwent liver transplantation for 2 months or less after the last chemotherapy cycle. At randomisation, the liver transplantation plus chemotherapy group received a median of 21·0 chemotherapy cycles (IQR 18·0-29·0) versus 17·0 cycles (12·0-24·0) in the chemotherapy alone group, in up to three lines of chemotherapy. During first-line chemotherapy, 64 (68%) of 94 patients had received doublet chemotherapy and 30 (32%) of 94 patients had received triplet regimens; 76 (80%) of 94 patients had targeted therapy. Transplanted patients received tailored immunosuppression (methylprednisolone 10 mg/kg intravenously on day 0; tacrolimus 0·1 mg/kg via gastric tube on day 0, 6-10 ng/mL days 1-14; mycophenolate mofetil 10 mg/kg intravenously day 0 to <2 months and switch to everolimus 5-8 ng/mL), and postoperative chemotherapy, and the chemotherapy group had continued chemotherapy. The primary endpoint was 5-year overall survival analysed in the intention to treat and per-protocol population. Safety events were assessed in the as-treated population. The study is registered with ClinicalTrials.gov (NCT02597348), and accrual is complete. FINDINGS: Between Feb 18, 2016, and July 5, 2021, 94 patients were randomly assigned and included in the intention-to-treat population, with 47 in the liver transplantation plus chemotherapy group and 47 in the chemotherapy alone group. 11 patients in the liver transplantation plus chemotherapy group and nine patients in the chemotherapy alone group did not receive the assigned treatment; 36 patients and 38 patients in each group, respectively, were included in the per-protocol analysis. Patients had a median age of 54·0 years (IQR 47·0-59·0), and 55 (59%) of 94 patients were male and 39 (41%) were female. Median follow-up was 59·3 months (IQR 42·4-60·2). In the intention-to-treat population, 5-year overall survival was 56·6% (95% CI 43·2-74·1) for liver transplantation plus chemotherapy and 12·6% (5·2-30·1) for chemotherapy alone (HR 0·37 [95% CI 0·21-0·65]; p=0·0003) and 73·3% (95% CI 59·6-90·0) and 9·3% (3·2-26·8), respectively, for the per-protocol population. Serious adverse events occurred in 32 (80%) of 40 patients who underwent liver transplantation (from either group), and 69 serious adverse events were observed in 45 (83%) of 54 patients treated with chemotherapy alone. Three patients in the liver transplantation plus chemotherapy group were retransplanted, one of whom died postoperatively of multi-organ failure. INTERPRETATION: In selected patients with permanently unresectable colorectal liver metastases, liver transplantation plus chemotherapy with organ allocation priority significantly improved survival versus chemotherapy alone. These results support the validation of liver transplantation as a new standard option for patients with permanently unresectable liver-only metastases. FUNDING: French National Cancer Institute and Assistance Publique-Hôpitaux de Paris.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms , Liver Neoplasms , Liver Transplantation , Female , Humans , Male , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Combined Modality Therapy , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/administration & dosage , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Prospective Studies , Treatment Outcome
17.
Biomed Pharmacother ; 179: 117325, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39226729

ABSTRACT

Direct-acting antivirals ledipasvir (LDV) and daclatasvir (DCV) are widely used as part of combination therapies to treat Hepatitis C infections. Here we show that these compounds inhibit the proliferation, invasion, and colony formation of triple-negative MDA-MB-231 breast cancer cells, SRC-transduced SW620 colon cancer cells and SRC- transduced NIH3T3 fibroblasts. DCV also inhibits the expression of PDL-1, which is responsible for resistance to immunotherapy in breast cancer cells. The demonstrated low toxicity in many Hepatitis C patients suggests LDV and DCV could be used in combination therapies for cancer patients. At the molecular level, these direct-acting antivirals inhibit the phosphorylation of Akt and the ephrin type A receptor 2 (EPHA2) by destabilizing a Src-EPHA2 complex, although they do not affect the general kinase activity of Src. Thus, LDV and DCV could be effective drugs for Src-associated cancers without the inherent toxicity of classical Src inhibitors.


Subject(s)
Antiviral Agents , Benzimidazoles , Carbamates , Colorectal Neoplasms , Down-Regulation , Fluorenes , Imidazoles , Proto-Oncogene Proteins c-akt , Pyrrolidines , Signal Transduction , Triple Negative Breast Neoplasms , Valine , src-Family Kinases , Humans , Benzimidazoles/pharmacology , Animals , Pyrrolidines/pharmacology , Imidazoles/pharmacology , Mice , Proto-Oncogene Proteins c-akt/metabolism , src-Family Kinases/metabolism , Fluorenes/pharmacology , Cell Line, Tumor , Antiviral Agents/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Carbamates/pharmacology , Down-Regulation/drug effects , Valine/analogs & derivatives , Valine/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Signal Transduction/drug effects , NIH 3T3 Cells , Female , Cell Proliferation/drug effects , United States Food and Drug Administration , Drug Approval , United States
18.
Biomed Pharmacother ; 179: 117363, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236476

ABSTRACT

HER2 amplification is one of the mechanisms that induce drug resistance to anti-EGFR therapy in colorectal cancer. In recent years, data from several randomized clinical trials show that anti-HER2 therapies improved the prognosis of patients with HER2-positive colorectal cancer. These results indicate that HER2 is a promising therapeutic target in advanced colorectal cancer. Despite the anti-HER2 therapies including monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates improving the outcomes, less than 30 % of the patients achieve objective response and eventually have drug resistance. It is necessary to explore the primary and secondary mechanisms for the resistance to anti-HER2 therapies, which will pave the way to overcome the drug resistance. Several studies have reported the potential mechanisms for the resistance to anti-HER2 therapies. In this review, we present a comprehensive overview of the recent advances in clinical research, mechanisms of treatment resistance, and strategies for reversing resistance in HER2-positive colorectal cancer patients.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Molecular Targeted Therapy , Receptor, ErbB-2 , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Animals
19.
Nutrients ; 16(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39275334

ABSTRACT

In this study, we discovered the mechanisms underlying parecoxib and resveratrol combination's anti-cancer characteristics against human colorectal cancer DLD-1 cells. We studied its anti-proliferation and apoptosis-provoking effect by utilizing cell viability 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, fluorescence microscope, gene overexpression, Western blot, and flow cytometry analyses. Parecoxib enhanced the ability of resveratrol to inhibit cell viability and increase apoptosis. Parecoxib in combination with resveratrol strongly enhanced apoptosis by inhibiting the expression of thioredoxin domain containing 5 (TXNDC5) and Akt phosphorylation. Parecoxib enhanced resveratrol-provoked c-Jun N-terminal kinase (JNK) and p38 phosphorylation. Overexpression of TXNDC5 and repression of JNK and p38 pathways significantly reversed the inhibition of cell viability and stimulation of apoptosis by the parecoxib/resveratrol combination. This study presents evidence that parecoxib enhances the anti-cancer power of resveratrol in DLD-1 colorectal cancer cells via the inhibition of TXNDC5 and Akt signaling and enhancement of JNK/p38 MAPK pathways. Parecoxib may be provided as an efficient drug to sensitize colorectal cancer by resveratrol.


Subject(s)
Apoptosis , Cell Survival , Colorectal Neoplasms , Isoxazoles , Proto-Oncogene Proteins c-akt , Resveratrol , Humans , Resveratrol/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Isoxazoles/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Phosphorylation/drug effects , MAP Kinase Signaling System/drug effects , Drug Synergism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Molecules ; 29(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39274965

ABSTRACT

The endolichenic fungus Aspergillus sp. was isolated from the lichen Xanthoparmelia conspersa harvested in France. Aspergillus sp. was grown on a solid culture medium to ensure the large-scale production of the fungus with a sufficient mass of secondary metabolites. The molecular network analysis of extracts and subfractions enabled the annotation of 22 molecules, guiding the purification process. The EtOAc extract displayed an antiproliferative activity of 3.2 ± 0.4 µg/mL at 48 h against human colorectal cancer cells (HT-29) and no toxicity at 30 µg/mL against human triple-negative breast cancer (TNBC) cells (MDA-MB-231) and human embryonic kidney (HEK293) non-cancerous cells. Among the five prenylated compounds isolated, of which four are echinulin derivatives, compounds 1 and 2 showed the most important activity, with IC50 values of 1.73 µM and 8.8 µM, respectively, against HT-29 cells.


Subject(s)
Antineoplastic Agents , Aspergillus , Cell Proliferation , Colorectal Neoplasms , Humans , Aspergillus/chemistry , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , HT29 Cells , HEK293 Cells , Cell Line, Tumor , Lichens/chemistry , Lichens/microbiology , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL