Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.755
Filter
1.
Oncol Res ; 32(7): 1163-1172, 2024.
Article in English | MEDLINE | ID: mdl-38948019

ABSTRACT

Background: Osteosarcoma is the most common malignant primary bone tumor. The prognosis for patients with disseminated disease remains very poor despite recent advancements in chemotherapy. Moreover, current treatment regimens bear a significant risk of serious side effects. Thus, there is an unmet clinical need for effective therapies with improved safety profiles. Taurolidine is an antibacterial agent that has been shown to induce cell death in different types of cancer cell lines. Methods: In this study, we examined both the antineoplastic and antiangiogenic effects of taurolidine in animal models of osteosarcoma. K7M2 murine osteosarcoma cells were injected, both intramuscular and intraperitoneal, into 60 BALB/c mice on day zero. Animals were then randomized to receive treatment with taurolidine 2% (800 mg/kg), taurolidine 1% (400 mg/kg), or NaCl 0.9% control for seven days by intravenous or intraperitoneal administration. Results: After 35 days, mice were euthanized, and the tumors were harvested for analysis. Eighteen mice were excluded from the analysis due to complications. Body weight was significantly lower in the 2% taurolidine intraperitoneal treatment group from day 9 to 21, consistent with elevated mortality in this group. Intraperitoneal tumor weight was significantly lower in the 1% (p = 0.003) and 2% (p = 0.006) intraperitoneal taurolidine treatment groups compared to the control. No antineoplastic effects were observed on intramuscular tumors or for intravenous administration of taurolidine. There were no significant differences in microvessel density or mitotic rate between treatment groups. Reduced body weight and elevated mortality in the 2% taurolidine intraperitoneal group suggest that the lower 1% dose is preferable. Conclusions: In conclusion, there is no evidence of antiangiogenic activity, and the antitumor effects of taurolidine on osteosarcoma observed in this study are limited. Moreover, its toxic profile grants further evaluation. Given these observations, further research is necessary to refine the use of taurolidine in osteosarcoma treatment.


Subject(s)
Bone Neoplasms , Disease Models, Animal , Osteosarcoma , Taurine , Thiadiazines , Tumor Burden , Animals , Taurine/analogs & derivatives , Taurine/pharmacology , Taurine/therapeutic use , Thiadiazines/pharmacology , Thiadiazines/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/blood supply , Mice , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Tumor Burden/drug effects , Microvascular Density/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Humans , Neovascularization, Pathologic/drug therapy
2.
BMC Cancer ; 24(1): 895, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39054508

ABSTRACT

BACKGROUND: The metabolic tumour area (MTA) was found to be a promising predictor of prostate cancer. However, the role of MTA based on 18F-FDG PET/CT in diffuse large B-cell lymphoma (DLBCL) prognosis remains unclear. This study aimed to elucidate the prognostic significance of MTA and evaluate its incremental value to the National Comprehensive Cancer Network International Prognostic Index (NCCN-IPI) for DLBCL patients treated with first-line R-CHOP regimens. METHODS: A total of 280 consecutive patients with newly diagnosed DLBCL and baseline 18F-FDG PET/CT data were retrospectively evaluated. Lesions were delineated via a semiautomated segmentation method based on a 41% SUVmax threshold to estimate semiquantitative metabolic parameters such as total metabolic tumour volume (TMTV) and MTA. Receiver operating characteristic (ROC) curve analysis was used to determine the optimal cut-off values. Progression-free survival (PFS) and overall survival (OS) were the endpoints that were used to evaluate the prognosis. PFS and OS were estimated via Kaplan‒Meier curves and compared via the log-rank test. RESULTS: Univariate analysis revealed that patients with high MTA, high TMTV and NCCN-IPI ≥ 4 were associated with inferior PFS and OS (P < 0.0001 for all). Multivariate analysis indicated that MTA remained an independent predictor of PFS and OS [hazard ratio (HR), 2.506; 95% confidence interval (CI), 1.337-4.696; P = 0.004; and HR, 1.823; 95% CI, 1.005-3.310; P = 0.048], whereas TMTV was not. Further analysis using the NCCN-IPI model as a covariate revealed that MTA and NCCN-IPI were still independent predictors of PFS (HR, 2.617; 95% CI, 1.494-4.586; P = 0.001; and HR, 2.633; 95% CI, 1.650-4.203; P < 0.0001) and OS (HR, 2.021; 95% CI, 1.201-3.401; P = 0.008; and HR, 3.869; 95% CI, 1.959-7.640; P < 0.0001; respectively). Furthermore, MTA was used to separate patients with high NCCN-IPI risk scores into two groups with significantly different outcomes. CONCLUSIONS: Pre-treatment MTA based on 18F-FDG PET/CT and NCCN-IPI were independent predictor of PFS and OS in DLBCL patients treated with R-CHOP. MTA has additional predictive value for the prognosis of patients with DLBCL, especially in high-risk patients with NCCN-IPI ≥ 4. In addition, the combination of MTA and NCCN-IPI may be helpful in further improving risk stratification and guiding individualised treatment options. TRIAL REGISTRATION: This research was retrospectively registered with the Ethics Committee of the Third Affiliated Hospital of Soochow University, and the registration number was approval No. 155 (approved date: 31 May 2022).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cyclophosphamide , Doxorubicin , Fluorodeoxyglucose F18 , Lymphoma, Large B-Cell, Diffuse , Positron Emission Tomography Computed Tomography , Prednisone , Rituximab , Vincristine , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Positron Emission Tomography Computed Tomography/methods , Vincristine/therapeutic use , Vincristine/administration & dosage , Cyclophosphamide/therapeutic use , Cyclophosphamide/administration & dosage , Middle Aged , Female , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prednisone/administration & dosage , Prednisone/therapeutic use , Doxorubicin/therapeutic use , Doxorubicin/administration & dosage , Prognosis , Aged , Retrospective Studies , Adult , Rituximab/therapeutic use , Aged, 80 and over , Young Adult , Tumor Burden/drug effects , ROC Curve , Radiopharmaceuticals , Adolescent
3.
Clinics (Sao Paulo) ; 79: 100434, 2024.
Article in English | MEDLINE | ID: mdl-38959634

ABSTRACT

OBJECTIVES: To retrospectively investigate the impact of pre-treatment Extracellular Volume Fraction (ECV) measured by Computed Tomography (CT) on the response of primary lesions to preoperative chemotherapy in abdominal neuroblastoma. METHODS: A total of seventy-five patients with abdominal neuroblastoma were retrospectively included in the study. The regions of interest for the primary lesion and aorta were determined on unenhanced and equilibrium phase CT images before treatment, and their average CT values were measured. Based on patient hematocrit and average CT values, the ECV was calculated. The correlation between ECV and the reduction in primary lesion volume was examined. A receiver operating characteristic curve was generated to assess the predictive performance of ECV for a very good partial response of the primary lesion. RESULTS: There was a negative correlation between primary lesion volume reduction and ECV (r = -0.351, p = 0.002), and primary lesions with very good partial response had lower ECV (p < 0.001). The area under the curve for ECV in predicting the very good partial response of primary lesion was 0.742 (p < 0.001), with a 95 % Confidence Interval of 0.628 to 0.836. The optimal cut-off value was 0.28, and the sensitivity and specificity were 62.07 % and 84.78 %, respectively. CONCLUSIONS: The measurement of pre-treatment ECV on CT images demonstrates a significant correlation with the response of the primary lesion to preoperative chemotherapy in abdominal neuroblastoma.


Subject(s)
Abdominal Neoplasms , Neuroblastoma , Tomography, X-Ray Computed , Humans , Neuroblastoma/diagnostic imaging , Neuroblastoma/drug therapy , Neuroblastoma/surgery , Neuroblastoma/pathology , Male , Female , Retrospective Studies , Tomography, X-Ray Computed/methods , Child, Preschool , Child , Infant , Abdominal Neoplasms/diagnostic imaging , Abdominal Neoplasms/drug therapy , Abdominal Neoplasms/pathology , Abdominal Neoplasms/surgery , Treatment Outcome , ROC Curve , Predictive Value of Tests , Adolescent , Tumor Burden/drug effects , Sensitivity and Specificity , Reference Values , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Reproducibility of Results
4.
Cancer Lett ; 597: 217011, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38849011

ABSTRACT

BACKGROUND: Improving survival for patients diagnosed with metastatic disease and overcoming chemoresistance remain significant clinical challenges in treating breast cancer. Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by a lack of therapeutically targetable receptors (ER/PR/HER2). TNBC therapy includes a combination of cytotoxic chemotherapies, including microtubule-targeting agents (MTAs) like paclitaxel (taxane class) or eribulin (vinca class); however, there are currently no FDA-approved MTAs that bind to the colchicine-binding site. Approximately 70 % of patients who initially respond to paclitaxel will develop taxane resistance (TxR). We previously reported that an orally bioavailable colchicine-binding site inhibitor (CBSI), VERU-111, inhibits TNBC tumor growth and treats pre-established metastatic disease. To further improve the potency and metabolic stability of VERU-111, we created next-generation derivatives of its scaffold, including 60c. RESULTS: 60c shows improved in vitro potency compared to VERU-111 for taxane-sensitive and TxR TNBC models, and suppress TxR primary tumor growth without gross toxicity. 60c also suppressed the expansion of axillary lymph node metastases existing prior to treatment. Comparative analysis of excised organs for metastasis between 60c and VERU-111 suggested that 60c has unique anti-metastatic tropism. 60c completely suppressed metastases to the spleen and was more potent to reduce metastatic burden in the leg bones and kidney. In contrast, VERU-111 preferentially inhibited liver metastases and lung metastasis repression was similar. Together, these results position 60c as an additional promising CBSI for TNBC therapy, particularly for patients with TxR disease.


Subject(s)
Colchicine , Triple Negative Breast Neoplasms , Xenograft Model Antitumor Assays , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Humans , Female , Animals , Colchicine/pharmacology , Cell Line, Tumor , Binding Sites , Tubulin Modulators/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Pyridines/pharmacology , Mice, Nude , Mice , Tumor Burden/drug effects
5.
J Radiat Res ; 65(4): 467-473, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38842109

ABSTRACT

Radioresistance is increasingly developed in esophageal cancer. Increasing radiation sensitivity can reduce the mortality of esophageal cancer. To investigate the effect and mechanism of ozone on the radiotherapy sensitization of esophageal carcinoma. KYSE150 cells were xenografted subcutaneously into nude mice and irradiated with 8 Gy radiation according to different subgroups (sham, radiation, ozone and radiation+ozone group (n = 10 per group)). Half of the mice were used to determine the body weight, tumor size and tumor weight. Half of the mice were used to collect peripheral blood. The serum was centrifuged to detect circulating cell-free DNA (cf-DNA), interleukin-6 (IL-6), interferon-γ (IFN-γ), myeloperoxidase (MPO)-DNA complexes, tumor necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9) and hypoxia-inducible factor-1α (HIF-1α) using commercial kits. The levels of phosphorylation AMP-activated protein kinase (p-AMPK) and scavenger receptor-A (SR-A) were measured by immunocytochemistry and Western blotting in the tumor tissues of mice. Ozone alone or combined with radiation therapy significantly reduced the body weight, tumor volume and tumor weight of esophageal cancer compared to the sham group. The ELISA results showed that the levels of cf-DNA, IFN-γ, MPO-DNA complexes, TNF-α, IL-6, HIF-1α and MMP-9 in the peripheral blood of mice treated with ozone combined with radiation were significantly lower compared with the radiation group. Ozone, synergistically with radiation, significantly increased the protein expression of p-AMPK and SR-A. Ozone may increase the radiosensitivity of esophageal cancer by inhibiting neutrophil extracellular traps.


Subject(s)
Esophageal Neoplasms , Ozone , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/drug therapy , Ozone/therapeutic use , Ozone/pharmacology , Animals , Humans , Cell Line, Tumor , Mice, Nude , Matrix Metalloproteinase 9/metabolism , Mice , AMP-Activated Protein Kinases/metabolism , Mice, Inbred BALB C , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell-Free Nucleic Acids/blood , Peroxidase/metabolism , Interleukin-6/metabolism , Interleukin-6/blood , Xenograft Model Antitumor Assays , Interferon-gamma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Burden/radiation effects , Tumor Burden/drug effects
6.
PLoS One ; 19(5): e0300171, 2024.
Article in English | MEDLINE | ID: mdl-38701062

ABSTRACT

PURPOSE: To investigate the treatment efficacy of intra-arterial (IA) trastuzumab treatment using multiparametric magnetic resonance imaging (MRI) in a human breast cancer xenograft model. MATERIALS AND METHODS: Human breast cancer cells (BT474) were stereotaxically injected into the brains of nude mice to obtain a xenograft model. The mice were divided into four groups and subjected to different treatments (IA treatment [IA-T], intravenous treatment [IV-T], IA saline injection [IA-S], and the sham control group). MRI was performed before and at 7 and 14 d after treatment to assess the efficacy of the treatment. The tumor volume, apparent diffusion coefficient (ADC), and dynamic contrast-enhanced (DCE) MRI parameters (Ktrans, Kep, Ve, and Vp) were measured. RESULTS: Tumor volumes in the IA-T group at 14 d after treatment were significantly lower than those in the IV-T group (13.1 mm3 [interquartile range 8.48-16.05] vs. 25.69 mm3 [IQR 20.39-30.29], p = 0.005), control group (IA-S, 33.83 mm3 [IQR 32.00-36.30], p<0.01), and sham control (39.71 mm3 [IQR 26.60-48.26], p <0.001). The ADC value in the IA-T group was higher than that in the control groups (IA-T, 7.62 [IQR 7.23-8.20] vs. IA-S, 6.77 [IQR 6.48-6.87], p = 0.044 and vs. sham control, 6.89 [IQR 4.93-7.48], p = 0.004). Ktrans was significantly decreased following the treatment compared to that in the control groups (p = 0.002 and p<0.001 for vs. IA-S and sham control, respectively). Tumor growth was decreased in the IV-T group compared to that in the sham control group (25.69 mm3 [IQR 20.39-30.29] vs. 39.71 mm3 [IQR 26.60-48.26], p = 0.27); there was no significant change in the MRI parameters. CONCLUSION: IA treatment with trastuzumab potentially affects the early response to treatment, including decreased tumor growth and decrease of Ktrans, in a preclinical brain tumor model.


Subject(s)
Breast Neoplasms , Injections, Intra-Arterial , Mice, Nude , Trastuzumab , Xenograft Model Antitumor Assays , Trastuzumab/administration & dosage , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Animals , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Female , Mice , Cell Line, Tumor , Multiparametric Magnetic Resonance Imaging/methods , Tumor Burden/drug effects , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Mice, Inbred BALB C
7.
Anticancer Res ; 44(6): 2425-2436, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821607

ABSTRACT

BACKGROUND/AIM: Despite the advances in oncology and cancer treatment over the past decades, cancer remains one of the deadliest diseases. This study focuses on further understanding the complex nature of cancer by using mathematical tumor modeling to understand, capture as best as possible, and describe its complex dynamics under chemotherapy treatment. MATERIALS AND METHODS: Focusing on autoregressive with exogenous inputs, i.e., ARX, and adaptive neuro-fuzzy inference system, i.e., ANFIS, models, this work investigates tumor growth dynamics under both single and combination anticancer agent chemotherapy treatments using chemotherapy treatment data on xenografted mice. RESULTS: Four ARX and ANFIS models for tumor growth inhibition were developed, estimated, and evaluated, demonstrating a strong correlation with tumor weight data, with ANFIS models showing superior performance in handling the multi-agent tumor growth complexities. These findings suggest potential clinical applications of the ANFIS models through further testing. Both types of models were also tested for their prediction capabilities across different chemotherapy schedules, with accurate forecasting of tumor growth up to five days in advance. The use of adaptive prediction and sliding (moving) data window techniques allowed for continuous model updating, ensuring more robust predictive capabilities. However, long-term forecasting remains a challenge, with accuracy declining over longer prediction horizons. CONCLUSION: While ANFIS models showed greater reliability in predictions, the simplicity and rapid deployment of ARX models offer advantages in situations requiring immediate approximations. Future research with larger, more diverse datasets and by exploring varying model complexities is recommended to improve the models' reliability and applicability in clinical decision-making, thereby aiding the development of personalized chemotherapy regimens.


Subject(s)
Neoplasms , Animals , Mice , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Xenograft Model Antitumor Assays , Fuzzy Logic , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Tumor Burden/drug effects
8.
In Vivo ; 38(3): 1058-1063, 2024.
Article in English | MEDLINE | ID: mdl-38688611

ABSTRACT

BACKGROUND/AIM: Colorectal cancer (CRC) is the third-leading cause of death in the world. Although the prognosis has improved due to improvement of chemotherapy, metastatic CRC is still a recalcitrant disease, with a 5-year survival of only 13%. Irinotecan (IRN) is used as first-line chemotherapy for patients with unresectable CRC. However, there are severe side effects, such as neutropenia and diarrhea, which are dose-limiting. We have previously shown that methionine restriction (MR), effected by recombinant methioninase (rMETase), lowered the effective dose of IRN of colon-cancer cells in vitro. The aim of the present study was to evaluate the efficacy of the combination of low-dose IRN and MR on colon-cancer in nude mice. MATERIALS AND METHODS: HCT-116 colon-cancer cells were cultured and subcutaneously injected into the flank of nude mice. After the tumor size reached approximately 100 mm3, 18 mice were randomized into three groups; Group 1: untreated control on a normal diet; Group 2: high-dose IRN on a normal diet (2 mg/kg, i.p.); Group 3: low-dose IRN (1 mg/kg i.p.) on MR effected by a methionine-depleted diet. RESULTS: There was no significant difference between the control mice and the mice treated with high-dose IRN, without MR. However, low-dose IRN combined with MR was significantly more effective than the control and arrested colon-cancer growth (p=0.03). Body weight loss was reversible in the mice treated by low-dose IRN combined with MR. CONCLUSION: The combination of low-dose IRN and MR acted synergistically in arresting HCT-116 colon-cancer grown in nude mice. The present study indicates the MR has the potential to reduce the effective dose of IRN in the clinic.


Subject(s)
Carbon-Sulfur Lyases , Colonic Neoplasms , Irinotecan , Methionine , Mice, Nude , Xenograft Model Antitumor Assays , Animals , Irinotecan/administration & dosage , Irinotecan/pharmacology , Methionine/administration & dosage , Humans , Mice , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Disease Models, Animal , HCT116 Cells , Cell Line, Tumor , Tumor Burden/drug effects
9.
J Vasc Interv Radiol ; 35(7): 1033-1042.e11, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38513753

ABSTRACT

PURPOSE: To investigate immuno-ethanol ablation using an ethanol and immune adjuvant formulation as a potent immunoablation approach that can achieve an enhanced anticancer effect in the treatment of hepatocellular carcinoma (HCC). MATERIALS AND METHODS: Ethanol concentration- and exposure time-dependent cellular responses were investigated. Curcumin was combined with ethanol as an immunoablation agent. Cellular uptake of curcumin, cancer cell killing, and inflammatory markers of ethanol-curcumin treatment were characterized. To evaluate the potential in vivo anticancer immunity of ethanol-curcumin treatment, each right and left lobe of rat liver was concurrently inoculated with N1S1 HCC cells and a mixture of treated N1S1 cells (ethanol only or ethanol-curcumin) in Sprague Dawley rats (each group: 5 rats; control: nontreated N1S1 cells). Tumor growth and immune response were characterized with 7T magnetic resonance (MR) imaging, flow cytometry analysis, and immunohistology. RESULTS: An optimized ethanol-curcumin (10% ethanol and 0.5% weight/volume (w/v) curcumin solution) treatment contributed to an enhanced cellular uptake of curcumin, increased cancer cell killing, and decreased inflammatory reaction. Ethanol-curcumin-treated N1S1 cell implantation in the rat liver demonstrated N1S1 HCC tumor rejection. The secondary tumor growth by nontreated N1S1 cell inoculation was significantly suppressed at the same time. Activated anticancer immunity was evidenced by significantly increased CD8+ T cell infiltration (3.5-fold) and CD8+-to-regulatory T cell ratio (4.5-fold) in the experimental group compared with those in the control group. CONCLUSIONS: Enhanced anticancer effect of immuno-ethanol ablation could be achieved with ethanol-curcumin agent. The results underscore the importance of optimized immunoablation therapeutic procedures for enhanced therapeutic outcomes.


Subject(s)
Carcinoma, Hepatocellular , Curcumin , Ethanol , Rats, Sprague-Dawley , Animals , Ethanol/pharmacology , Ethanol/administration & dosage , Curcumin/pharmacology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Rats , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Male , Ablation Techniques , Antineoplastic Agents/pharmacology , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/drug therapy , Immunotherapy/methods , Dose-Response Relationship, Drug , Tumor Burden/drug effects , Time Factors
10.
J Complement Integr Med ; 21(2): 205-214, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38515384

ABSTRACT

OBJECTIVES: Sesamum indicum L. seeds; rich in zinc and lignans are endowed with antioxidant and immunomodulatory properties which attract research on their anticancer potential. Although many studies have reported the in vitro antitumor potential of S. indicum and its phytoconstituents, much is yet to be known about its in vivo effects. To fill this gap, the effects of dietary supplementation with seeds of S. indicum in 7,12-dimethylbenz(a)anthracene-exposed rats was assessed. METHODS: 42 rats aged 30-35 days were randomized into six groups (n=6) as follows: the normal (NOR) and negative (DMBA) control groups were fed with standard diet; the positive control group (DMBA + Zinc) was fed with standard diet supplemented with commercial zinc (0.01 %); the test groups were fed with standard diet supplemented with S. indicum seeds in different proportions (6.25 , 12.5 and 25 %). Breast cancer was induced by a single administration of DMBA (50 mg/kg BW, s.c.) diluted in corn oil. The experiment lasted 20 weeks and afterward, tumor incidence; tumor burden, tumor volume, tumor micro-architecture and some biochemical parameters were evaluated. RESULTS: As salient result, 100 % of rats in the DMBA group developed tumors, while rats feed with rat chow supplemented with S. indicum seeds (25 %) had a reduced incidence of tumors (33.3 %) and tumor volume (2.71 cm3 in sesame 25 % vs. 4.69 cm3 in the DMBA group, p˂0.01). The seeds (25 %) also slowed DMBA-induced neoplasm expansion in mammary ducts as compared to rats of DMBA group. CONCLUSIONS: In summary, supplementation with S. indicum seeds slowed breast tumorigenesis via its antioxidant capacity.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene , Dietary Supplements , Seeds , Sesamum , Animals , Sesamum/chemistry , Seeds/chemistry , Female , Rats , Plant Extracts/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/prevention & control , Tumor Burden/drug effects , Antioxidants/pharmacology , Breast Neoplasms/chemically induced , Breast Neoplasms/prevention & control
11.
Int J Surg ; 110(7): 4074-4082, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38537071

ABSTRACT

BACKGROUND: There is no standardized assessment for evaluating response although neoadjuvant chemotherapy (NAT) is widely accepted for borderline resectable or locally advanced pancreatic cancer (BRPC or LAPC). This study was aimed to evaluate NAT response using positron emission tomography (PET) with 2-deoxy-2-[fluorine-18]fluoro-D-glucose ( 18 F-FDG-PET/CT) parameters alongside carbohydrate antigen (CA) 19-9 levels. METHODS: Patients who underwent surgery after NAT for BRPC and LAPC between 2017 and 2021 were identified. The study assessed the prognostic value of PET-derived parameters after NAT, determining cutoff values using the K-adaptive partitioning method. It created four groups based on the elevation or normalization of PET parameters and CA19-9 levels, comparing survival between these groups. RESULTS: Of 200 eligible patients, FOLFIRINOX and gemcitabine-based NAT was administered in 166 and 34 patients, respectively (mean NAT cycles, 8.3). In a multivariate analysis, metabolic tumor volume (MTV) demonstrated the most robust performance in assessing response [hazard ratio (HR) 3.11, 95% confidence interval (CI) 1.73-5.58, P <0.001] based on cutoff value of 2.4. Patients with decreased MTV had significantly better survival than those with elevated MTV among individuals with CA19-9 levels less than 37 IU/l (median survival; 35.5 vs. 20.9 months, P <0.001) and CA19-9 levels at least 37 IU/l (median survival; 34.3 vs. 17.8 months, P =0.03). In patients suspected to be Lewis antigen negative, the predictive performance of MTV was found to be limited ( P =0.84). CONCLUSION: Elevated MTV is an influential prognostic factor for worse survival, regardless of post-NAT CA19-9 levels. These results could be helpful in identifying patients with a poor prognosis despite normalization of CA19-9 levels after NAT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Neoadjuvant Therapy , Pancreatic Neoplasms , Positron Emission Tomography Computed Tomography , Tumor Burden , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/mortality , Male , Female , Middle Aged , Aged , Prognosis , Tumor Burden/drug effects , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorodeoxyglucose F18 , CA-19-9 Antigen/blood , CA-19-9 Antigen/metabolism , Fluorouracil/administration & dosage , Adult , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Irinotecan
12.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 5265-5274, 2024 07.
Article in English | MEDLINE | ID: mdl-38270618

ABSTRACT

Melanoma, an aggressive and potentially fatal skin cancer, is constrained by immunosuppression, resistance, and high toxicity in its treatment. Consequently, there is an urgent need for innovative antineoplastic agents. Therefore, this study investigated the antimelanoma potential of guttiferone E (GE). In an allogeneic murine B16 melanoma model, GE was administered subcutaneously and intraperitoneally. Antitumor evaluation included tumor volume/weight measurements and histopathological and immunohistochemical analysis. Furthermore, the toxicity of the treatments was evaluated through body/organ weights, biochemical parameters, and genotoxicity. Subcutaneous administration of 20 mg/kg of GE resulted in a significant reduction in both tumor volume and weight, effectively suppressing melanoma cell proliferation as evidenced by a decrease in mitotic figures. The tumor growth inhibition rate was equivalent to 54%. This treatment upregulated cleaved caspase-3, indicating apoptosis induction. On the other hand, intraperitoneal administration of GE showed no antimelanoma effect. Remarkably, GE treatments exhibited no toxicity, evidenced by non-significant differences in body weight gain, as well as organ weight, biochemical parameters of nephrotoxicity and hepatotoxicity, and genotoxic damage. This study revealed, for the first time, the efficacy of subcutaneous administration of GE in reducing melanoma, in the absence of toxicity. Furthermore, it was observed that the apoptotic signaling pathway is involved in the antimelanoma property of GE. These findings offer valuable insights for further exploring GE's therapeutic applications in melanoma treatment.


Subject(s)
Melanoma, Experimental , Mice, Inbred C57BL , Animals , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Apoptosis/drug effects , Mice , Male , Antineoplastic Agents/toxicity , Antineoplastic Agents/administration & dosage , Benzophenones/pharmacology , Benzophenones/administration & dosage , Benzophenones/toxicity , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Cell Proliferation/drug effects , Tumor Burden/drug effects , Cell Line, Tumor , Injections, Subcutaneous , Female
13.
Proc Natl Acad Sci U S A ; 119(32): e2116289119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35917342

ABSTRACT

Glioblastoma (GBM) is an aggressive malignant primary brain tumor with limited therapeutic options. We show that the angiotensin II (AngII) type 2 receptor (AT2R) is a therapeutic target for GBM and that AngII, endogenously produced in GBM cells, promotes proliferation through AT2R. We repurposed EMA401, an AT2R antagonist originally developed as a peripherally restricted analgesic, for GBM and showed that it inhibits the proliferation of AT2R-expressing GBM spheroids and blocks their invasiveness and angiogenic capacity. The crystal structure of AT2R bound to EMA401 was determined and revealed the receptor to be in an active-like conformation with helix-VIII blocking G-protein or ß-arrestin recruitment. The architecture and interactions of EMA401 in AT2R differ drastically from complexes of AT2R with other relevant compounds. To enhance central nervous system (CNS) penetration of EMA401, we exploited the crystal structure to design an angiopep-2-tethered EMA401 derivative, A3E. A3E exhibited enhanced CNS penetration, leading to reduced tumor volume, inhibition of proliferation, and increased levels of apoptosis in an orthotopic xenograft model of GBM.


Subject(s)
Angiotensin II Type 2 Receptor Blockers , Benzhydryl Compounds , Brain Neoplasms , Drug Repositioning , Glioblastoma , Isoquinolines , Receptor, Angiotensin, Type 2 , Analgesics/pharmacology , Angiotensin II/chemistry , Angiotensin II/pharmacology , Angiotensin II Type 2 Receptor Blockers/therapeutic use , Apoptosis , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Protein Conformation, alpha-Helical , Receptor, Angiotensin, Type 2/chemistry , Receptor, Angiotensin, Type 2/metabolism , Tumor Burden/drug effects
14.
Anticancer Res ; 42(3): 1313-1325, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220222

ABSTRACT

BACKGROUND/AIM: Chloride intracellular channel protein 1 (CLIC1) is known as a promoter of cancer progression, metastasis, and angiogenesis. Thus, CLIC1 could be a future therapeutic target. This study aimed to evaluate the effect of anti-CLIC1 antibodies on tumour cells and vessels of human renal cell carcinoma (RCC) in rabbit cornea and chick embryo chorioallantoic membrane (CAM) models. MATERIALS AND METHODS: Human cc-RCC xenografts on rabbit cornea and CAM surface were performed. Anti-CLIC1 antibodies were applied for 5 consecutive days on both tumor models. We comparatively evaluated treated and untreated tumors by combining ultrasonography with microscopic techniques. RESULTS: RCC implants rapidly recruited blood vessels and had an exponential growth rate on both tumor models. Anti-CLIC1 antibodies suppressed tumor growth by inducing tumor cell necrosis. Tumor vessels regressed rapidly but not completely during anti-CLIC1 antibodies based therapy. CONCLUSION: Anti-CLIC1 antibodies induced tumor necrosis and tumor vasculature regression in human cc-RCC xenografts in both in vivo experimental models.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Chloride Channels/antagonists & inhibitors , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Neovascularization, Pathologic , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Chick Embryo , Chloride Channels/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Molecular Targeted Therapy , Necrosis , Rabbits , Signal Transduction , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Drug Deliv ; 29(1): 506-518, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35147065

ABSTRACT

The field of biomedical research has recently been interested in nanoplatforms with various functionalities, such as cancer drug carriers and MRI and optical imaging, as well as thermal treatment, among other things. As a result of the present investigation, a unique multifunctional liposome (MFL) was established in this investigation. Using radiofrequency-induced imaging and drug release based on magnetic field impact, a dual drug delivery targeted with tumor multi-mechanism treatment was made more effective. The C60 (fullerene) surface was coated with iron nanocomposites to establish the proposed nanosystems, and PEGylation was used (Fe3O4-C60-PEG2000). For fullerene radiofrequency-triggered drug release, thermosensitive DPPC liposomes with folate-DSPE-PEG2000 enveloped the binary nanosystems and doxorubicin (DOX). The in vitro cytotoxicity of the nanocomposites was confirmed by the liver metastasis in HT-29 colon cancer cells using radiofrequency. The flow cytometry analysis confirmed the apoptosis cell death mechanism. The thermal treatment combined chemotherapeutic MFL nano framework transformed radiofrequency radiation from thermoresponsive liposomes, which was noticed both in vivo and in vitro. Due to their superior active tumor targeting and magnetic targeting characteristics, the MFL could also selectively destroy cancerous liver cells in highly co-localized targets.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Liposomes/chemistry , Liver Neoplasms/pathology , Radiofrequency Ablation/methods , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Apoptosis/drug effects , Chemistry, Pharmaceutical , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Liberation , Fullerenes/chemistry , Liver Neoplasms/secondary , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Particle Size , Polyethylene Glycols/chemistry , Surface Properties , Tumor Burden/drug effects
16.
Nat Commun ; 13(1): 703, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121738

ABSTRACT

Rho family mechano-signaling through the actin cytoskeleton positively regulates physiological TEAD/YAP transcription, while the evolutionarily conserved Hippo tumor suppressor pathway antagonizes this transcription through YAP cytoplasmic localization/degradation. The mechanisms responsible for oncogenic dysregulation of these pathways, their prevalence in tumors, as well as how such dysregulation can be therapeutically targeted are not resolved. We demonstrate that p53 DNA contact mutants in human tumors, indirectly hyperactivate RhoA/ROCK1/actomyosin signaling, which is both necessary and sufficient to drive oncogenic TEAD/YAP transcription. Moreover, we demonstrate that recurrent lesions in the Hippo pathway depend on physiological levels of ROCK1/actomyosin signaling for oncogenic TEAD/YAP transcription. Finally, we show that ROCK inhibitors selectively antagonize proliferation and motility of human tumors with either mechanism. Thus, we identify a cancer driver paradigm and a precision medicine approach for selective targeting of human malignancies driven by TEAD/YAP transcription through mechanisms that either upregulate or depend on homeostatic RhoA mechano-signaling.


Subject(s)
Cell Cycle Proteins/genetics , Neoplasms/genetics , Signal Transduction/genetics , TEA Domain Transcription Factors/genetics , Transcription Factors/genetics , rho-Associated Kinases/genetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/genetics , Humans , Mice, SCID , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , TEA Domain Transcription Factors/metabolism , Transcription Factors/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays/methods , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
Sci Rep ; 12(1): 1701, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35105915

ABSTRACT

Interferon-gamma (IFN-γ) is shown to stimulate melanoma development and progression. However, the underlying mechanism has not been completely defined. Our study aimed to determine the role of neuronal nitric oxide synthase (nNOS)-mediated signaling in IFN-γ-stimulated melanoma progression and the anti-melanoma effects of novel nNOS inhibitors. Our study shows that IFN-γ markedly induced the expression levels of nNOS in melanoma cells associated with increased intracellular nitric oxide (NO) levels. Co-treatment with novel nNOS inhibitors effectively alleviated IFN-γ-activated STAT1/3. Further, reverse phase protein array (RPPA) analysis demonstrated that IFN-γ induced the expression of HIF1α, c-Myc, and programmed death-ligand 1 (PD-L1), in contrast to IFN-α. Blocking the nNOS-mediated signaling pathway using nNOS-selective inhibitors was shown to effectively diminish IFN-γ-induced PD-L1 expression in melanoma cells. Using a human melanoma xenograft mouse model, the in vivo studies revealed that IFN-γ increased tumor growth compared to control, which was inhibited by the co-administration of nNOS inhibitor MAC-3-190. Another nNOS inhibitor, HH044, was shown to effectively inhibit in vivo tumor growth and was associated with reduced PD-L1 expression levels in melanoma xenografts. Our study demonstrates the important role of nNOS-mediated NO signaling in IFN-γ-stimulated melanoma progression. Targeting nNOS using highly selective small molecular inhibitors is a unique and effective strategy to improve melanoma treatment.


Subject(s)
Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Disease Progression , Enzyme Inhibitors/administration & dosage , Interferon-gamma/administration & dosage , Melanoma/drug therapy , Melanoma/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Animals , B7-H1 Antigen/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Interferon-alpha/pharmacology , Melanoma/pathology , Mice , Mice, Nude , Nitric Oxide Synthase Type I/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Skin Neoplasms/pathology , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Carbohydr Polym ; 282: 119087, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35123755

ABSTRACT

The efficient triggering of prodrug release has become a challengeable task for stimuli-responsive nanomedicine utilized in cancer therapy due to the subtle differences between normal and tumor tissues and heterogeneity. In this work, a dual ROS-responsive nanocarriers with the ability to self-regulate the ROS level was constructed, which could gradually respond to the endogenous ROS to achieve effective, hierarchical and specific drug release in cancer cells. In brief, DOX was conjugated with MSNs via thioketal bonds and loaded with ß-Lapachone. TPP modified chitosan was then coated to fabricate nanocarriers for mitochondria-specific delivery. The resultant nanocarriers respond to the endogenous ROS and release Lap specifically in cancer cells. Subsequently, the released Lap self-regulated the ROS level, resulting in the specific DOX release and mitochondrial damage in situ, enhancing synergistic oxidation-chemotherapy. The tumor inhibition Ratio was achieved to 78.49%. The multi-functional platform provides a novel remote drug delivery system in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Naphthoquinones/administration & dosage , Neoplasms/drug therapy , Oxidative Stress , Prodrugs/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Chitosan/administration & dosage , Chitosan/chemistry , Chitosan/pharmacokinetics , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Mitochondria/physiology , Nanoparticles/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacokinetics , Neoplasms/metabolism , Neoplasms/pathology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Oxidation-Reduction , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Reactive Oxygen Species/metabolism , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Tumor Burden/drug effects
19.
Pharm Res ; 39(2): 353-367, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35166995

ABSTRACT

PURPOSE: The invention and application of new immunotherapeutic methods can compensate for the inefficiency of conventional cancer treatment approaches, partly due to the inhibitory microenvironment of the tumor. In this study, we tried to inhibit the growth of cancer cells and induce anti-tumor immune responses by silencing the expression of the ß-catenin in the tumor microenvironment and transmitting interleukin (IL)-15 cytokine to provide optimal conditions for the dendritic cell (DC) vaccine. METHODS: For this purpose, we used folic acid (FA)-conjugated SPION-carboxymethyl dextran (CMD) chitosan (C) nanoparticles (NPs) to deliver anti-ß-catenin siRNA and IL-15 to cancer cells. RESULTS: The results showed that the codelivery of ß-catenin siRNA and IL-15 significantly reduced the growth of cancer cells and increased the immune response. The treatment also considerably stimulated the performance of the DC vaccine in triggering anti-tumor immunity, which inhibited tumor development and increased survival in mice in two different cancer models. CONCLUSIONS: These findings suggest that the use of new nanocarriers such as SPION-C-CMD-FA could be an effective way to use as a novel combination therapy consisting of ß-catenin siRNA, IL-15, and DC vaccine to treat cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cancer Vaccines/administration & dosage , Dendritic Cells/transplantation , Drug Carriers , Interleukin-15/administration & dosage , Magnetic Iron Oxide Nanoparticles , Melanoma, Experimental/therapy , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Skin Neoplasms/therapy , beta Catenin/genetics , Animals , Antineoplastic Agents/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Dendritic Cells/immunology , Drug Compounding , Female , Gene Expression Regulation, Neoplastic , Interleukin-15/chemistry , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred BALB C , RNA, Small Interfering/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Burden/drug effects , Tumor Microenvironment
20.
Eur J Pharmacol ; 918: 174773, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35065044

ABSTRACT

Emerging evidence points to a strong association between the bile acid (BA)-gut microbiota (GM) axis, and the risk of colorectal cancer (CRC). Kaempferol, a common polyphenol in the daily diet, shows various pharmacological activities. However, it remains unclear about the effect of kaempferol on the CRC development and the BA-GM homeostasis. Here, we found kaempferol effectively reduced tumor burden, restored the damaged intestinal barrier and downregulated antigen Ki67 and leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) expressions in ApcMin/+ mice. For BA metabolism, kaempferol reversed the decreasing trend in chenodesoxycholic acid (CDCA) and 12α-hydroxylated BAs by increasing the sterol 27-hydroxylase (CYP27A1) and sterol 12α-hydroxylase (CYP8B1) expressions, and upregulated FXR expression. Importantly, molecular docking analysis revealed a direct interaction between kaempferol and farnesoid X receptor (FXR), the mater regulator of BA signaling. For GM analysis, we found higher abundances of species with anticancer properties and lower abundances of species associated with inflammation, obesity, and metabolic disorders in kaempferol-treated groups. Moreover, the gut of kaempferol-treated mice was predominantly colonized by short-chain fatty acid (SCFA) and lactic acid producing bacteria. Based on the PICRUSt-predicted pathways of our GM dataset, we demonstrated that kaempferol downregulated secondary BA synthesis pathways, increased G protein-coupled receptor activity and decreased NOD-like receptor activity, affecting cell differentiation, proliferation, survival, and apoptosis. Collectively, these results reveal that kaempferol effectively attenuates the tumor burden in ApcMin/+ mice by modulating the BA signaling and GM homeostasis.


Subject(s)
Bile Acids and Salts/metabolism , Colorectal Neoplasms , Gastrointestinal Microbiome , Kaempferols/pharmacology , Ki-67 Antigen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Gene Expression Regulation, Neoplastic/drug effects , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL