Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.615
Filter
1.
Cancer Med ; 13(11): e7326, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38826114

ABSTRACT

BACKGROUND: Optimal adjuvant chemotherapy after laparoscopic surgery in gastric cancer (GC) patients is still undefined. We aimed to evaluate the efficacy of S-1 plus oxaliplatin (SOX) and capecitabine plus oxaliplatin (CAPOX) in patients with GC after laparoscopic gastrectomy. METHODS: A non-inferiority randomized controlled clinical trial was performed in China. Patients with advanced GC who underwent laparoscopic D2 gastrectomy were randomly assigned to receive SOX and CAPOX regimens. RESULTS: In total, 191 patients were screened between May 2018 and June 2019, and 140 (73.3%) were included in the modified intent-to-treat analysis (mITT), of whom 69 and 71 were assigned to the SOX and CAPOX groups, respectively. The SOX group had similar 3-year overall survival (OS) and disease-free survival to the CAPOX group. Subgroup analysis revealed significantly better OS in the SOX group for male patients ([HR] = 0.395; 95% [CI], 0.153-1.019; p = 0.045), age >60 (HR = 0.219; 95% [CI], 0.064-0.753; p = 0.016), tumors in the gastric antrum (HR = 0.273; 95% [CI], 0.076-0.981; p = 0.047), and moderately differentiated tumors (HR = 0.338; 95% [CI], 0.110-1.041; p = 0.041). There were no significant differences observed in terms of adverse events and recurrence patterns between the two groups. CONCLUSION: Adjuvant SOX was non-inferior to CAPOX treatments for patients with GC who underwent curative laparoscopic D2 gastrectomy. For male patients, aged >60 years, tumors in the gastric antrum, and moderately differentiated tumors, adjuvant SOX may achieve an improvement compared with CAPOX.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Capecitabine , Drug Combinations , Gastrectomy , Laparoscopy , Oxaliplatin , Oxonic Acid , Stomach Neoplasms , Tegafur , Humans , Stomach Neoplasms/surgery , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Male , Gastrectomy/methods , Female , Middle Aged , Laparoscopy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Oxaliplatin/therapeutic use , Oxaliplatin/administration & dosage , Tegafur/therapeutic use , Tegafur/administration & dosage , Oxonic Acid/therapeutic use , Oxonic Acid/administration & dosage , Chemotherapy, Adjuvant/methods , Capecitabine/administration & dosage , Capecitabine/therapeutic use , Aged , Adult
2.
BJS Open ; 8(3)2024 May 08.
Article in English | MEDLINE | ID: mdl-38722737

ABSTRACT

BACKGROUND: This study evaluated the efficacy of hyperthermic intraperitoneal chemotherapy (HIPEC) in colorectal cancer with peritoneal metastases (pmCRC) in a large international data set of patients. PATIENTS AND METHODS: Patients with pmCRC from 39 centres who underwent cytoreductive surgery with HIPEC between 1991 and 2018 were selected and compared for the HIPEC protocols received-oxaliplatin-HIPEC versus mitomycin-HIPEC. Following analysis of crude data, propensity-score matching (PSM) and Cox-proportional hazard modelling were performed. Outcomes of interest were overall survival (OS), recurrence-free survival (RFS) and the HIPEC dose-response effects (high versus low dose, dose intensification and double drug protocols) on OS, RFS and 90-day morbidity. Furthermore, the impact of the treatment time period was assessed. RESULTS: Of 2760 patients, 2093 patients were included. Median OS was 43 months (95% c.i. 41 to 46 months) with a median RFS of 12 months (95% c.i. 12 to 13 months). The oxaliplatin-HIPEC group had an OS of 47 months (95% c.i. 42 to 53 months) versus 39 months (95% c.i. 36 to 43 months) in the mitomycin-HIPEC group (P = 0.002), aHR 0.77, 95% c.i. 0.67 to 0.90, P < 0.001. The OS benefit persisted after PSM of the oxaliplatin-HIPEC group and mitomycin-HIPEC group (48 months (95% c.i. 42 to 59 months) versus 40 months (95% c.i. 37 to 44 months)), P < 0.001, aHR 0.78 (95% c.i. 0.65 to 0.94), P = 0.009. Similarly, matched RFS was significantly higher for oxaliplatin-HIPEC versus others (13 months (95% c.i. 12 to 15 months) versus 11 months (95% c.i. 10 to 12 months, P = 0.02)). High-dose mitomycin-HIPEC protocols had similar OS compared to oxaliplatin-HIPEC. HIPEC dose intensification within each protocol resulted in improved survival. Oxaliplatin + irinotecan-HIPEC resulted in the most improved OS (61 months (95% c.i. 51 to 101 months)). Ninety-day mortality in both crude and PSM analysis was worse for mitomycin-HIPEC. There was no change in treatment effect depending on the analysed time period. CONCLUSIONS: Oxaliplatin-based HIPEC provided better outcomes compared to mitomycin-based HIPEC. High-dose mitomycin-HIPEC was similar to oxaliplatin-HIPEC. The 90-day mortality difference favours the oxaliplatin-HIPEC group. A trend for dose-response between low- and high-dose HIPEC was reported.


Subject(s)
Colorectal Neoplasms , Cytoreduction Surgical Procedures , Hyperthermic Intraperitoneal Chemotherapy , Mitomycin , Oxaliplatin , Peritoneal Neoplasms , Humans , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/mortality , Male , Female , Middle Aged , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/mortality , Mitomycin/administration & dosage , Mitomycin/therapeutic use , Aged , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Retrospective Studies , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Propensity Score , Disease-Free Survival , Treatment Outcome , Proportional Hazards Models
3.
Acta Oncol ; 63: 322-329, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745482

ABSTRACT

BACKGROUND AND PURPOSE: Perioperative 5-FU, leucovorin, oxaliplatin, and docetaxel (FLOT) is recommended in resectable esophagogastric adenocarcinoma based on randomised trials. However, the effectiveness of FLOT in routine clinical practice remains unknown as randomised trials are subject to selection bias limiting their generalisability. The aim of this study was to evaluate the implementation of FLOT in real-world patients. METHODS: Retrospectively collected data were analysed in consecutive patients treated before or after the implementation of FLOT. The primary endpoint was complete pathological response (pCR) and secondary endpoints were margin-free resection (R0), overall survival (OS), relapse-free survival (RFS) tolerability of chemotherapy and surgical complications. RESULTS: Mean follow-up time for patients treated with FLOT (n = 205) was 37.7 versus 47.0 months for epirubicin, cis- or oxaliplatin, and capecitabine (ECX/EOX, n = 186). Surgical resection was performed in 88.0% versus 92.0%; pCR were observed in 3.8% versus 2.4%; and R0 resections were achieved in 78.0% versus 86.0% (p = 0.03) in the ECX/EOX and FLOT cohorts, respectively. Survival analysis indicated no significant difference in RFS (p = 0.17) or OS (p = 0.37) between the cohorts with a trend towards increased OS in performance status 0 (hazard ratio [HR] = 0.73, 95% confidence interval [CI]: 0.50-1.04). More patients treated with ECX/EOX completed chemotherapy (39% vs. 28%, p = 0.02). Febrile neutropenia was more common in the FLOT cohort (3.8% vs. 11%, p = 0.0086). 90-days mortality (1.2% vs. 0%) and frequency of anastomotic leakage (8% vs. 6%) were equal and low. INTERPRETATION: Patients receiving FLOT did not demonstrate improved pCR, RFS or OS. However, R0 rate was improved and patients in good PS trended towards improved OS.


Subject(s)
Adenocarcinoma , Antineoplastic Combined Chemotherapy Protocols , Capecitabine , Docetaxel , Esophageal Neoplasms , Fluorouracil , Leucovorin , Oxaliplatin , Stomach Neoplasms , Humans , Male , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adenocarcinoma/mortality , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/surgery , Female , Middle Aged , Aged , Oxaliplatin/therapeutic use , Oxaliplatin/administration & dosage , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Capecitabine/administration & dosage , Docetaxel/administration & dosage , Docetaxel/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Leucovorin/therapeutic use , Leucovorin/administration & dosage , Epirubicin/administration & dosage , Adult , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Aged, 80 and over , Perioperative Care/methods , Esophagogastric Junction/pathology
4.
Bull Exp Biol Med ; 176(5): 703-708, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38724815

ABSTRACT

The activity of known modulators of the Nrf2 signaling pathway (bardoxolone and brusatol) was studied on cultures of tumor organoids of metastatic colorectal cancer previously obtained from three patients. The effect of modulators was studied both as monotherapy and in combination with standard chemotherapy drugs used to treat colorectal cancer. The Nrf2 inhibitor brusatol and the Nrf2 activator bardoxolone have antitumor activity. Moreover, bardoxolone and brusatol also significantly enhance the effect of the chemotherapy drugs 5-fluorouracil, oxaliplatin, and irinotecan metabolite SN-38. Thus, bardoxolone and brusatol can be considered promising candidates for further preclinical and clinical studies in the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms , Fluorouracil , Irinotecan , NF-E2-Related Factor 2 , Organoids , Oxaliplatin , Quassins , Signal Transduction , NF-E2-Related Factor 2/metabolism , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Quassins/pharmacology , Quassins/therapeutic use , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Signal Transduction/drug effects , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Irinotecan/pharmacology , Irinotecan/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Synergism , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Camptothecin/therapeutic use
5.
J Nanobiotechnology ; 22(1): 257, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755645

ABSTRACT

Imperceptible examination and unideal treatment effect are still intractable difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). At present, despite 5-fluorouracil (5-FU), as a clinical first-line FOLFIRINOX chemo-drug, has achieved significant therapeutic effects. Nevertheless, these unavoidable factors such as low solubility, lack of biological specificity and easy to induce immunosuppressive surroundings formation, severely limit their treatment in PDAC. As an important source of energy for many tumor cells, tryptophan (Trp), is easily degraded to kynurenine (Kyn) by indolamine 2,3- dioxygenase 1 (IDO1), which activates the axis of Kyn-AHR to form special suppressive immune microenvironment that promotes tumor growth and metastasis. However, our research findings that 5-FU can induce effectively immunogenic cell death (ICD) to further treat tumor by activating immune systems, while the secretion of interferon-γ (IFN-γ) re-induce the Kyn-AHR axis activation, leading to poor treatment efficiency. Therefore, a metal matrix protease-2 (MMP-2) and endogenous GSH dual-responsive liposomal-based nanovesicle, co-loading with 5-FU (anti-cancer drug) and NLG919 (IDO1 inhibitor), was constructed (named as ENP919@5-FU). The multifunctional ENP919@5-FU can effectively reshape the tumor immunosuppression microenvironment to enhance the effect of chemoimmunotherapy, thereby effectively inhibiting cancer growth. Mechanistically, PDAC with high expression of MMP-2 will propel the as-prepared nanovesicle to dwell in tumor region via shedding PEG on the nanovesicle surface, effectively enhancing tumor uptake. Subsequently, the S-S bond containing nanovesicle was cut via high endogenous GSH, leading to the continued release of 5-FU and NLG919, thereby enabling circulating chemoimmunotherapy to effectively cause tumor ablation. Moreover, the combination of ENP919@5-FU and PD-L1 antibody (αPD-L1) showed a synergistic anti-tumor effect on the PDAC model with abdominal cavity metastasis. Collectively, ENP919@5-FU nanovesicle, as a PDAC treatment strategy, showed excellent antitumor efficacy by remodeling tumor microenvironment to circulate tumor chemoimmunotherapy amplification, which has promising potential in a precision medicine approach.


Subject(s)
Carcinoma, Pancreatic Ductal , Fluorouracil , Immunotherapy , Tumor Microenvironment , Tumor Microenvironment/drug effects , Animals , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Mice , Humans , Immunotherapy/methods , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Matrix Metalloproteinase 2/metabolism , Liposomes/chemistry , Kynurenine/metabolism , Interferon-gamma/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use
6.
Zhonghua Wei Chang Wai Ke Za Zhi ; 27(5): 486-494, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38778688

ABSTRACT

Objective: To investigate the sensitivity of tumor organoids derived from samples of colorectal cancer to lobaplatin and oxaliplatin hyperthermic perfusion in vitro and to assist clinical development of hyperthermic intraperitoneal chemotherapy. Method: Tumor samples and relevant clinical data were collected from patients with pathologically confirmed colorectal cancer in the Sixth Affiliated Hospital of Sun Yat-sen University from July 2021 to December 2022. Organoids were cultured and tumor tissue were passaged. In vitro hyperthermic perfusion experiments were performed on organoids with good viability. Firstly, 10 organoids were treated with oxaliplatin and lobaplatin at the following six concentrations: 1 000, 250, 62.5, 15.6, 3.9, and 0.98 µmol/L. The organoids were exposed to oxaliplatin at 42℃ for 30 minutes and to lobaplatin at 42℃ for 60 minutes. Dose-response curves of responses to in vitro hyperthermic perfusion with these two drugs were constructed and evaluated. Clinical doses of oxaliplatin and lobaplatin were further tested on 30 organoids. This testing revealed oxaliplatin was effective at 579 µmol/L at a hyperthermic perfusion temperature of 42℃ for 30 min and lobaplatin was effective at 240 µmol/L at a hyperthermic perfusion temperature of 42℃ for 60 minutes. Result: Thirty-two tumor organoids were cultured from samples of colorectal cancer. The median concentration required for oxaliplatin to eliminate 50% of tumor cells (IC50) was 577.45 µmol/L (IQR: 1846.09 µmol/L). The median IC50 for lobaplatin was 85.04 µmol/L (IQR: 305.01 µmol/L).The difference between the two groups was not statistically significant (Z=1.784, P=0.084). In seven of 10 organoids, lobaplatin showed a greater IC50 after in vitro hyperthermic perfusion than did oxaliplatin. Testing of 30 organoids with clinical doses of oxaliplatin and lobaplatin revealed that oxaliplatin achieved an average inhibition rate of 39.6% (95%CI: 32.1%‒47.0%), whereas the average rate of inhibition for lobaplatin was 89.7% (95%CI: 87.0%‒92.3%): this difference is statistically significant (t=‒15.282, P<0.001). Conclusion: The rate of inhibition achieved by hyperthermic perfusion of lobaplatin in vitro is better than that achieved by hyperthermic perfusion with oxaliplatin. Lobaplatin is more effective than oxaliplatin when administered by hyperthermic intraperitoneal perfusion and therefore has the potential to replace oxaliplatin in this setting.


Subject(s)
Colorectal Neoplasms , Cyclobutanes , Hyperthermic Intraperitoneal Chemotherapy , Organoids , Organoplatinum Compounds , Oxaliplatin , Humans , Cyclobutanes/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Oxaliplatin/pharmacology , Hyperthermia, Induced/methods , Female , Male , Antineoplastic Agents/administration & dosage
7.
BMC Cancer ; 24(1): 634, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783256

ABSTRACT

BACKGROUND: Adenocarcinoma of the ampulla of Vater (AoV) is one of the rare periampullary cancers, and due to its anatomical location, it is categorized into various histologic subtypes. Its rarity and diversity pose challenges in treatment decision-making for patients with advanced AoV carcinoma. This study investigated the efficacy and safety of the combined regimen of capecitabine and oxaliplatin (CAPOX) in a real-world clinical setting. METHODS: This investigation encompassed patients with advanced AoV carcinoma who underwent CAPOX treatment. Histologic phenotypes were identified through a combination of histopathological analysis and protein expression markers, including MUC1, CDX2, CK20, and MUC2. The correlation between histopathological determinants and survival outcomes was explored, in addition to an evaluation of the safety profile of CAPOX therapy. RESULTS: From January 2010 to June 2023, 42 patients received CAPOX. Of these, 14 patients (33.3%) had not received any prior palliative chemotherapy, while 28 patients (66.7%) had undergone one prior line of chemotherapy. At a median follow up of 9.0 months, the median progression-free survival (PFS) was 4.38 months (95% CI, 2.78-5.69) and the median overall survival (OS) was 9.57 months (95% CI 7.56-11.6). The objective response and disease control rates were 38.1% and 61.9%, respectively. Patients who received CAPOX as a second-line treatment had poorer PFS (HR = 2.62; 95% CI, 1.49-4.90, p = 0.003) and OS (HR = 2.82, 95% CI, 1.47-5.38, p = 0.001) compared to those who received CAPOX as a first-line chemotherapy. There were no statistically significant differences in PFS (p = 0.185) and OS (p = 0.097) between groups based on histologic subtypes. Neutropenia (14.3%) emerged as the predominant grade 3-4 toxicity. Notably, treatment cessation occurred in select instances owing to grade 3 fatigue (9.5%) and peripheral neuropathy (9.5%). CONCLUSIONS: This study confirmed the therapeutic efficacy and safety of CAPOX in a real-world setting, consistent with prior phase II trial results. While CAPOX proved feasible for advanced AoV carcinoma regardless of histologic subtype, its reduced effectiveness in second-line settings necessitates further research to determine its optimal palliative use.


Subject(s)
Adenocarcinoma , Ampulla of Vater , Antineoplastic Combined Chemotherapy Protocols , Capecitabine , Common Bile Duct Neoplasms , Oxaliplatin , Humans , Capecitabine/therapeutic use , Capecitabine/administration & dosage , Capecitabine/adverse effects , Male , Oxaliplatin/therapeutic use , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Ampulla of Vater/pathology , Female , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aged , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/mortality , Adult , Common Bile Duct Neoplasms/drug therapy , Common Bile Duct Neoplasms/pathology , Common Bile Duct Neoplasms/mortality , Retrospective Studies , Progression-Free Survival , Treatment Outcome
8.
J Sep Sci ; 47(11): e2400164, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38819794

ABSTRACT

Oxaliplatin (L-OHP), a third-generation platinum-based anti-tumor drug, finds widespread application in the first-line treatment of metastatic colorectal cancer. Despite its efficacy, the drug's usage is curtailed by a litany of side effects, with L-OHP-induced peripheral neuropathy (OIPN) being the most debilitating. This condition can be classified into varying degrees of severity. Employing serum metabolomics, a high-sensitivity, high-throughput technique, holds promise as a method to identify biomarkers for clinical assessment and monitoring of OIPN patients across different severity levels. In our study, we analyzed serum metabolites in patients with different OIPN levels using ultra-performance liquid chromatography-high resolution mass spectrometry. By employing statistical analyses and pathway enrichment studies, we aimed to identify potential biomarkers and metabolic pathways. Our findings characterized the serum metabolic profiles of patients with varying OIPN levels. Notably, pathway analysis revealed a significant correlation with lipid metabolism, amino acid metabolism, and energy metabolism. Multivariate statistical analysis and receiver operator characteristic curve evaluation pointed to anhalamine and glycochenodeoxycholic acid as potential biomarkers for OIPN C and A, which suggest that serum metabolomics may serve as a potent tool for exploring the metabolic status of patients suffering from diverse diseases and for discovering novel biomarkers.


Subject(s)
Metabolomics , Oxaliplatin , Humans , Male , Female , Middle Aged , Antineoplastic Agents/blood , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/blood , Peripheral Nervous System Diseases/metabolism , Chromatography, High Pressure Liquid , Aged , Biomarkers/blood , Neurotoxicity Syndromes/blood , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/diagnosis
9.
Acta Biomater ; 181: 402-414, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734282

ABSTRACT

Tumor hypoxia, high oxidative stress, and low immunogenic create a deep-rooted immunosuppressive microenvironment, posing a major challenge to the therapeutic efficiency of cancer immunotherapy for solid tumor. Herein, an intelligent nanoplatform responsive to the tumor microenvironment (TME) capable of hypoxia relief and immune stimulation has been engineered for efficient solid tumor immunotherapy. The MnO2@OxA@OMV nanoreactor, enclosing bacterial-derived outer membrane vesicles (OMVs)-wrapped MnO2 nanoenzyme and the immunogenic cell death inducer oxaliplatin (OxA), demonstrated intrinsic catalase-like activity within the TME, which effectively catalyzed the endogenous H2O2 into O2 to enable a prolonged oxygen supply, thereby alleviating the tumor's oxidative stress and hypoxic TME, and expediting OxA release. The combinational action of OxA-caused ICD effect and Mn2+ from nanoreactor enabled the motivation of the cGAS-STING pathway to significantly improve the activation of STING and dendritic cells (DCs) maturation, resulting in metalloimmunotherapy. Furthermore, the immunostimulant OMVs played a crucial role in promoting the infiltration of activated CD8+T cells into the solid tumor. Overall, the nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy. STATEMENT OF SIGNIFICANCE: A tailor-made nanoreactor was fabricated by enclosing bacterial-derived outer membrane vesicles (OMVs) onto MnO2 nanoenzyme and loading with immunogenic cell death inducer oxaliplatin (OxA) for tumor metalloimmunotherapy. The nanoreactor possesses intrinsic catalase-like activity within the tumor microenvironment, which effectively enabled a prolonged oxygen supply by catalyzing the conversion of endogenous H2O2 into O2, thereby alleviating tumor hypoxia and expediting OxA release. Furthermore, the TME-responsive release of nutritional Mn2+ sensitized the cGAS-STING pathway and collaborated with OxA-induced immunogenic cell death (ICD). Combing with immunostimulatory OMVs enhances the uptake of nanoreactors by DCs and promotes the infiltration of activated CD8+T cells. This nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy.


Subject(s)
Immunotherapy , Tumor Microenvironment , Animals , Immunotherapy/methods , Mice , Tumor Microenvironment/drug effects , Cell Line, Tumor , Tumor Hypoxia/drug effects , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Oxides/chemistry , Oxides/pharmacology , Manganese/chemistry , Manganese/pharmacology , Humans , Female , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/immunology , Neoplasms/drug therapy , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/immunology , Mice, Inbred C57BL
10.
J Cancer Res Clin Oncol ; 150(5): 283, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806870

ABSTRACT

OBJECTIVE: The aim of this study is to assess the clinical efficacy of a 5 mg dosage of olanzapine in preventing chemotherapy-induced nausea and vomiting (CINV) associated with moderately emetogenic chemotherapy (MEC) among female patients diagnosed with gastrointestinal tract tumors. METHODS: Patients undergoing the oxaliplatin/irinotecan chemotherapy regimen were enrolled in this prospective controlled study. The olanzapine group received a 5 mg dosage of olanzapine along with palonosetron and dexamethasone, while the control group received a standard two-combination regimen consisting of dexamethasone and palonosetron. The primary endpoints included the total protection (TP) rates for the entire age group and the subgroup aged 60 years and above. Secondary endpoints encompassed the total protection rates during the acute and delayed phases within the two age brackets, as well as the total control (TC) rates and complete remission (CR) rates across all three phases (total, acute, and delayed). Additionally, the study involved the assessment of quality of life and the collection of adverse events associated with the interventions. RESULTS: 1) Regarding the primary endpoint, the total phase TP rates within both the entire age group and the age group exceeding 60 years demonstrated superiority in the olanzapine group when compared to the control group (66.7% vs 37.25%, P = 0.003; 68.8% vs 44.4%, P = 0.044). 2) In terms of secondary endpoints, the olanzapine group exhibited superior acute phase TP rates in both age brackets when compared to the control group (P < 0.05). The olanzapine group also demonstrated higher delayed-phase TP rates, TC rates across all three phases, and CR rates within the two age brackets, although the differences were not statistically significant (P > 0.05). Furthermore, the quality of life in the olanzapine group surpassed that of the control group for both age brackets (P < 0.05), characterized by enhanced appetite and a higher incidence of drowsiness in the patients treated with olanzapine when compared to those in the control group (P < 0.05). CONCLUSION: Olanzapine can enhance CINV induced by MEC regimen in female patients across all age groups, including the elderly, and therefore improve the quality of life for these patients. CLINICAL TRIAL REGISTRATION: https://www.chictr.org.cn/index.html , identifier: ChiCTR20000368269, 25/08/2020.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Irinotecan , Nausea , Olanzapine , Oxaliplatin , Vomiting , Humans , Olanzapine/administration & dosage , Olanzapine/therapeutic use , Olanzapine/adverse effects , Female , Middle Aged , Nausea/chemically induced , Nausea/prevention & control , Vomiting/chemically induced , Vomiting/prevention & control , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Prospective Studies , Oxaliplatin/adverse effects , Oxaliplatin/administration & dosage , Irinotecan/adverse effects , Irinotecan/administration & dosage , Aged , Adult , Antiemetics/administration & dosage , Antiemetics/therapeutic use , Gastrointestinal Neoplasms/drug therapy , Palonosetron/administration & dosage , Palonosetron/therapeutic use , Quality of Life , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use
11.
Anal Methods ; 16(19): 3125-3130, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38700061

ABSTRACT

A new fluorescence sensing approach has been proposed for the precise determination of the anti-cancer drug oxaliplatin (Oxal-Pt). This method entails synthesizing blue-emitting copper nanoclusters (CuNCs) functionalized with bovine serum albumin (BSA) as the stabilizing agent. Upon excitation at 360 nm, the resultant probe exhibits emission at 460 nm. Notably, the fluorescence response of BSA@CuNCs substantially increases upon incubation with Oxal-Pt due to multiple binding interactions between the drug and the fluorescent probe. These interactions involve hydrogen bonding, hydrophobic interaction, and the high affinity between the SH groups (cysteine residues of BSA) and platinum (in Oxal-Pt). Consequently, this interaction induces aggregation-induced emission enhancement (AIEE) of BSA@CuNCs. The probe demonstrates a broad response range from 0.08 to 140.0 µM, along with a low detection limit of 20.0 nM, determined based on a signal-to-noise ratio of 3. Furthermore, the probe effectively detects Oxal-Pt in injections, human serum, and urine samples, yielding acceptable results. This study represents a significant advancement in the development of a straightforward and efficient sensor for monitoring platinum-containing anti-cancer drugs during chemotherapy.


Subject(s)
Antineoplastic Agents , Copper , Drug Monitoring , Fluorescent Dyes , Oxaliplatin , Serum Albumin, Bovine , Spectrometry, Fluorescence , Oxaliplatin/chemistry , Serum Albumin, Bovine/chemistry , Copper/chemistry , Humans , Antineoplastic Agents/chemistry , Drug Monitoring/methods , Spectrometry, Fluorescence/methods , Fluorescent Dyes/chemistry , Metal Nanoparticles/chemistry , Animals , Limit of Detection , Neoplasms/drug therapy , Cattle
12.
BMC Cancer ; 24(1): 587, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38741073

ABSTRACT

YAP and TAZ, the Hippo pathway terminal transcriptional activators, are frequently upregulated in cancers. In tumor cells, they have been mainly associated with increased tumorigenesis controlling different aspects from cell cycle regulation, stemness, or resistance to chemotherapies. In fewer cases, they have also been shown to oppose cancer progression, including by promoting cell death through the action of the p73/YAP transcriptional complex, in particular after chemotherapeutic drug exposure. Using HCT116 cells, we show here that oxaliplatin treatment led to core Hippo pathway down-regulation and nuclear accumulation of TAZ. We further show that TAZ was required for the increased sensitivity of HCT116 cells to oxaliplatin, an effect that appeared independent of p73, but which required the nuclear relocalization of TAZ. Accordingly, Verteporfin and CA3, two drugs affecting the activity of YAP and TAZ, showed antagonistic effects with oxaliplatin in co-treatments. Importantly, using several colorectal cell lines, we show that the sensitizing action of TAZ to oxaliplatin is dependent on the p53 status of the cells. Our results support thus an early action of TAZ to sensitize cells to oxaliplatin, consistent with a model in which nuclear TAZ in the context of DNA damage and p53 activity pushes cells towards apoptosis.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Hippo Signaling Pathway , Organoplatinum Compounds , Oxaliplatin , Protein Serine-Threonine Kinases , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Suppressor Protein p53 , Humans , Oxaliplatin/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , HCT116 Cells , Signal Transduction/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Antineoplastic Agents/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Drug Resistance, Neoplasm/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Verteporfin/pharmacology , Verteporfin/therapeutic use , Cell Line, Tumor , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , YAP-Signaling Proteins/metabolism , Porphyrins/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Apoptosis/drug effects
13.
Sci Rep ; 14(1): 10745, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38730240

ABSTRACT

Gastric cancer is one of the most common malignant tumors, and chemotherapy is the main treatment for advanced gastric cancer. However, chemotherapy resistance leads to treatment failure and poor prognosis in patients with gastric cancer. Multidrug resistance (MDR) is a major challenge that needs to be overcome in chemotherapy. According to recent research, ferroptosis activation is crucial for tumor therapeutic strategies. In this work, we explored the solution to chemoresistance in gastric cancer by investigating the effects of the Chinese medicine monomer baicalin on ferroptosis. Baicalin with different concentrations was used to treat the parent HGC27 and drug-resistant HGC27/L cells of gastric cancer. Cell viability was measured by CCK8, and synergistic effects of baicalin combined with oxaliplatin were evaluated using Synergy Finder software. The effects of baicalin on organelles and cell morphology were investigated using projective electron microscopy. Iron concentration, MDA production and GSH inhibition rate were measured by colorimetry. ROS accumulation was detected by flow cytometry. The ferroptosis-related genes (IREB2, TfR, GPX4, FTH1), P53, and SLC7A11 were analysed by Western blot, and the expression differences of the above proteins between pretreatment and pretreatment of different concentrations of baicalin, were assayed in both parental HGC27 cells and Oxaliplatin-resistant HGC27/L cells. Mechanically, Baicalin disrupted iron homeostasis and inhibits antioxidant defense, resulting in iron accumulation, lipid peroxide aggregation, and specifically targeted and activated ferroptosis by upregulating the expression of tumor suppressor gene p53, thereby activating the SLC7A11/GPX4/ROS pathway mediated by it. Baicalin activates ferroptosis through multiple pathways and targets, thereby inhibiting the viability of oxaliplatin-resistant gastric cancer HGC27/L cells and enhancing the sensitivity to oxaliplatin chemotherapy.


Subject(s)
Drug Resistance, Neoplasm , Ferroptosis , Flavonoids , Oxaliplatin , Stomach Neoplasms , Tumor Suppressor Protein p53 , Ferroptosis/drug effects , Humans , Flavonoids/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Oxaliplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Drug Synergism , Reactive Oxygen Species/metabolism , Gene Expression Regulation, Neoplastic/drug effects
14.
J Med Chem ; 67(10): 8296-8308, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38739678

ABSTRACT

Platinum-drug-based chemotherapy in clinics has achieved great success in clinical malignancy therapy. However, unpredictable off-target toxicity and the resulting severe side effects in the treatment are still unsolved problems. Although metabolic glycan labeling-mediated tumor-targeted therapy has been widely reported, less selective metabolic labeling in vivo limited its wide application. Herein, a novel probe of B-Ac3ManNAz that is regulated by reactive oxygen species in tumor cells is introduced to enhance the recognition and cytotoxicity of DBCO-modified oxaliplatin(IV) via bioorthogonal chemistry. B-Ac3ManNAz was synthesized from Ac4ManNAz by incorporation with 4-(hydroxymethyl) benzeneboronic acid pinacol ester (HBAPE) at the anomeric position, which is confirmed to be regulated by ROS and could robustly label glycans on the cell surface. Moreover, N3-treated tumor cells could enhance the tumor accumulation of DBCO-modified oxaliplatin(IV) via click chemistry meanwhile reduce the off-target distribution in normal tissue. Our strategy provides an effective metabolic precursor for tumor-specific labeling and targeted cancer therapies.


Subject(s)
Antineoplastic Agents , Oxaliplatin , Polysaccharides , Prodrugs , Reactive Oxygen Species , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Humans , Reactive Oxygen Species/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude
15.
Anticancer Res ; 44(6): 2587-2595, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821580

ABSTRACT

BACKGROUND/AIM: Apoptosis resistance in cancer cells adapted to acidic microenvironments poses a challenge for effective treatment. This study investigated the potential use of caffeic acid as an adjunct therapy to overcome drug resistance in colorectal cancer cells under acidic conditions. MATERIALS AND METHODS: Long-term exposure to low-pH conditions induced resistance in HCT116 colorectal cancer cells. The effects of caffeic acid on proliferation, clonogenicity, and apoptosis induction were assessed alone and in combination with oxaliplatin and 5-Fluorouracil. The signaling pathways involved in drug resistance were examined by assessing the activities of PI3K/Akt and ERK1/2. RESULTS: Caffeic acid inhibited the proliferation and clonogenicity of acid-adapted cancer cells, and enhanced apoptosis when combined with anticancer drugs. Mechanistically, caffeic acid attenuated the hyperactivation of the PI3K/Akt and ERK1/2 signaling pathways associated with drug resistance. CONCLUSION: Caffeic acid is a promising therapeutic agent for targeting resistant cancer cells in acidic microenvironments. Its ability to inhibit proliferation, sensitize cells to apoptosis, and modulate signaling pathways highlights its potential for overcoming drug resistance in cancer therapy.


Subject(s)
Apoptosis , Caffeic Acids , Cell Proliferation , Colonic Neoplasms , Drug Resistance, Neoplasm , Fluorouracil , Humans , Caffeic Acids/pharmacology , Apoptosis/drug effects , HCT116 Cells , Cell Proliferation/drug effects , Fluorouracil/pharmacology , Drug Resistance, Neoplasm/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/pharmacology , Oxaliplatin/pharmacology , Signal Transduction/drug effects , Hydrogen-Ion Concentration , Drug Synergism , Phosphatidylinositol 3-Kinases/metabolism , Organoplatinum Compounds/pharmacology , Tumor Microenvironment/drug effects
16.
Clin Colorectal Cancer ; 23(2): 118-127.e6, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762348

ABSTRACT

BACKGROUND: The phase 1b KEYNOTE-651 study evaluated pembrolizumab plus chemotherapy in microsatellite stable or mismatch repair-proficient metastatic colorectal cancer. PATIENTS AND METHODS: Patients with microsatellite stable or mismatch repair-proficient metastatic colorectal cancer received pembrolizumab 200 mg every 3 weeks plus 5-fluorouracil, leucovorin, oxaliplatin (previously untreated; cohort B) or 5-fluorouracil, leucovorin, irinotecan (previously treated with fluoropyrimidine plus oxaliplatin; cohort D) every 2 weeks. Primary end point was safety; investigator-assessed objective response rate per RECIST v1.1 was secondary and biomarker analysis was exploratory. RESULTS: Thirty-one patients were enrolled in cohort B and 32 in cohort D; median follow-up was 30.2 and 33.5 months, respectively. One dose-limiting toxicity (grade 3 small intestine obstruction) occurred in cohort D. In cohort B, grade 3 or 4 treatment-related adverse events (AEs) occurred in 18 patients (58%), most commonly neutropenia and decreased neutrophil count (n = 5 each). In cohort D, grade 3 or 4 treatment-related AEs occurred in 17 patients (53%), most commonly neutropenia (n = 7). No grade 5 treatment-related AEs occurred. Objective response rate was 61% in cohort B (KRAS wildtype: 71%; KRAS mutant: 53%) and 25% in cohort D (KRAS wildtype: 47%; KRAS mutant: 6%). In both cohorts, PD-L1 combined positive score and T-cell-inflamed gene expression profiles were higher and HER2 expression was lower in responders than nonresponders. No association between tumor mutational burden and response was observed. CONCLUSION: Pembrolizumab plus 5-fluorouracil, leucovorin, oxaliplatin/5-fluorouracil, leucovorin, irinotecan demonstrated an acceptable AE profile. Efficacy data appeared comparable with current standard of care (including by KRAS mutation status). Biomarker analyses were hypothesis-generating, warranting further exploration. GOV IDENTIFIER: ClinicalTrials.gov; NCT03374254.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Camptothecin , Colorectal Neoplasms , Fluorouracil , Leucovorin , Organoplatinum Compounds , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leucovorin/administration & dosage , Leucovorin/adverse effects , Leucovorin/therapeutic use , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Female , Male , Middle Aged , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Aged , Camptothecin/analogs & derivatives , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Camptothecin/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Adult , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Microsatellite Instability/drug effects , DNA Mismatch Repair , Irinotecan/administration & dosage , Irinotecan/adverse effects , Oxaliplatin/administration & dosage , Aged, 80 and over
17.
Biomed Pharmacother ; 175: 116743, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759290

ABSTRACT

Pancreatic cancer is the third leading cause of cancer-related death in the United States, with a 5-year survival rate of only 12%. The poor prognosis of pancreatic cancer is primarily attributed to the lack of early detection, the aggressiveness of the disease, and its resistance to conventional chemotherapeutics. The use of combination chemotherapy targeting different key pathways has emerged as a potential strategy to minimize drug resistance while improving therapeutic outcomes. Here, we evaluated a novel approach to treating pancreatic cancer using entinostat (ENT), a selective class I and IV HDAC inhibitor, and oxaliplatin (OXP) administered at considerably lower dosages. Combination therapy exhibited strong synergistic interaction against human (PANC-1) and murine (KPC) pancreatic cancer cells. As expected, ENT treatment enhanced acetylated histone H3 and H4 expression in treated cells, which was even augmented in the presence of OXP. Similarly, cells treated with a combination therapy showed higher expression of cleaved caspase 3 and increased apoptosis compared to monotherapy. To further improve the efficacy of the combination treatment, we encapsulated OXP and ENT into bovine serum albumin and poly(lactic-co-glycolic) acid nanoparticles. Both nanocarriers showed suitable physicochemical properties with respect to size, charge, polydispersity index, and loading. Besides, the combination of OXP and ENT nanoparticles showed similar or even better synergistic effects compared to free drugs during in vitro cytotoxicity and colony formation assays towards pancreatic cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Apoptosis , Benzamides , Drug Carriers , Nanoparticles , Oxaliplatin , Pancreatic Neoplasms , Pyridines , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pyridines/pharmacology , Pyridines/administration & dosage , Humans , Oxaliplatin/pharmacology , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Benzamides/pharmacology , Benzamides/administration & dosage , Animals , Cell Line, Tumor , Mice , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Carriers/chemistry , Apoptosis/drug effects , Drug Synergism
18.
J Cancer Res Clin Oncol ; 150(5): 244, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717526

ABSTRACT

PURPOSE: Cystatin SA (CST2) belongs to the superfamily of cysteine protease inhibitors. Emerging research indicates that CST2 is often dysregulated across various cancers. Its role and molecular mechanisms in gastric cancer remain underexplored. This study aims to explore the expression and function of CST2 in gastric cancer. METHODS: CST2 expression was analyzed and validated through Western blot. CST2 overexpression was induced by lentivirus in GC cells, and the correlation between CST2 expression levels and downstream signaling pathways was assessed. In addition, multiple assays, including cell proliferation, colony formation, wound-healing, and transwell migration/invasion, were considered to ascertain the influence of CST2 overexpression on gastric cancer. The cell cycle and apoptosis were detected by flow cytometry. RESULTS: CST2 expression at the protein level was decreased to be reduced in both gastric cancer tissues and cell lines, and CST2 expression attenuate gastric cancer growth, an effect restricted to gastric cancer cells and absent in gastric epithelial GES-1 cells. Furthermore, CST2 was demonstrated to improve chemosensitivity to Oxaliplatin in gastric cancer cells through the PI3K/AKT signaling pathway. CONCLUSION: These findings indicate that CST2 is downregulated at the protein level in gastric cancer tissues and cell lines. Additionally, CST2 was found to attenuate the growth of gastric cancer cells and to enhance sensitivity to Oxaliplatin through the PI3K/AKT signaling pathway, specific to gastric cancer cell lines. CST2 may serve as a tumor suppressor gene increasing sensitivity to Oxaliplatin in gastric cancer.


Subject(s)
Cell Proliferation , Oxaliplatin , Salivary Cystatins , Stomach Neoplasms , Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Oxaliplatin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Salivary Cystatins/metabolism , Salivary Cystatins/genetics , Signal Transduction/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics
19.
Phytomedicine ; 129: 155647, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38703660

ABSTRACT

BACKGROUND: Oxaliplatin is the most common chemotherapeutic agent for patients with colorectal cancer. However, its anti-cancer efficacy is restricted by drug resistance occurring through several mechanisms, including autophagy. Liensinine exerts a considerable anti-tumor effect and can regulate autophagy. Inhibition of autophagy is a strategy to reverse resistance to oxaliplatin. The aim of this study was to check if liensinine can enhance the therapeutic efficacy of oxaliplatin in colorectal cancer and if so, elucidate its mechanism. METHODS: Two colorectal cancer cell lines, HCT116 and LoVo, and one normal intestinal epithelial cell, NCM-460 were used for in vitro experiments. Cell Counting Kit-8 (CCK-8), colony formation, and flow cytometry assays were used to evaluate the cytotoxicity of liensinine and oxaliplatin. Network pharmacology analysis and Human XL Oncology Array were used to screen targets of liensinine. Transfections and autophagy regulators were used to confirm these targets. The relationship between the target and clinical effect of oxaliplatin was analyzed. Patient-derived xenograft (PDX) models were used to validate the effects of liensinine and oxaliplatin. RESULTS: CCK-8 and colony formation assays both showed that the combination treatment of liensinine and oxaliplatin exerted synergistic effects. Results of the network pharmacology analysis and Human XL Oncology Array suggested that liensinine can inhibit autophagy by targeting HIF-1α/eNOS. HIF-1α was identified as the key factor modulated by liensinine in autophagy and induces resistance to oxaliplatin. HIF-1α levels in tumor cells and prognosis for FOLFOX were negatively correlated in clinical data. The results from three PDX models with different HIF-1α levels showed their association with intrinsic and acquired resistance to oxaliplatin in these models, which could be reversed by liensinine. CONCLUSIONS: Research on the relationship between HIF-1α levels and the clinical effect of oxaliplatin is lacking, and whether liensinine regulates HIF-1α is unknown. Our findings suggest that liensinine overcomes the resistance of colorectal cancer cells to oxaliplatin by suppressing HIF-1α levels to inhibit autophagy. Our findings can contribute to improving prognosis following colorectal cancer therapy.


Subject(s)
Autophagy , Colorectal Neoplasms , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit , Oxaliplatin , Humans , Oxaliplatin/pharmacology , Autophagy/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Colorectal Neoplasms/drug therapy , Animals , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Mice , Mice, Nude , HCT116 Cells , Xenograft Model Antitumor Assays , Drug Synergism , Isoquinolines , Phenols
SELECTION OF CITATIONS
SEARCH DETAIL
...