228 research outputs found

    Optimizing Druggability through Liposomal Formulations: New Approaches to an Old Concept

    Get PDF
    Developing innovative delivery strategies remains an ongoing task to improve both efficacy and safety of drug-based therapy. Nanomedicine is now a promising field of investigation, rising high expectancies for treating various diseases such as malignancies. Putting drugs into liposome is an old story that started in the late 1960s. Because of the near-total biocompatibility of their lipidic bilayer, liposomes are less concerned with the safety issue related to the possible long-term accumulation in the body of most nanoobjects currently developed in nanomedicine. Additionally, novel techniques and recent efforts to achieve better stability (e.g., through sheddable coating), combined with a higher selectivity towards target cells (e.g., by anchoring monoclonal antibodies or incorporating phage fusion protein), make new liposomal drugs an attractive and challenging opportunity to improve clinical outcome in a variety of disease. This review covers the physicochemistry of liposomes and the recent technical improvements in the preparation of liposome-encapsulated drugs in regard to the scientific and medical stakes

    Antiproliferative Effect of Ascorbic Acid Is Associated with the Inhibition of Genes Necessary to Cell Cycle Progression

    Get PDF
    BACKGROUND: Ascorbic acid (AA), or Vitamin C, is most well known as a nutritional supplement with antioxidant properties. Recently, we demonstrated that high concentrations of AA act on PMP22 gene expression and partially correct the Charcot-Marie-Tooth disease phenotype in a mouse model. This is due to the capacity of AA, but not other antioxidants, to down-modulate cAMP intracellular concentration by a competitive inhibition of the adenylate cyclase enzymatic activity. Because of the critical role of cAMP in intracellular signalling, we decided to explore the possibility that ascorbic acid could modulate the expression of other genes. METHODS AND FINDINGS: Using human pangenomic microarrays, we found that AA inhibited the expression of two categories of genes necessary for cell cycle progression, tRNA synthetases and translation initiation factor subunits. In in vitro assays, we demonstrated that AA induced the S-phase arrest of proliferative normal and tumor cells. Highest concentrations of AA leaded to necrotic cell death. However, quiescent cells were not susceptible to AA toxicity, suggesting the blockage of protein synthesis was mainly detrimental in metabolically-active cells. Using animal models, we found that high concentrations of AA inhibited tumor progression in nude mice grafted with HT29 cells (derived from human colon carcinoma). Consistently, expression of tRNA synthetases and ieF2 appeared to be specifically decreased in tumors upon AA treatment. CONCLUSIONS: AA has an antiproliferative activity, at elevated concentration that could be obtained using IV injection. This activity has been observed in vitro as well in vivo and likely results from the inhibition of expression of genes involved in protein synthesis. Implications for a clinical use in anticancer therapies will be discussed

    Pharmacodynamic therapeutic drug monitoring for cancer: challenges, advances, and future opportunities

    Get PDF
    In the modern era of cancer treatment, with targeted agents superseding more traditional cytotoxic chemotherapeutics, it is becoming increasingly important to use stratified medicine approaches to ensure that patients receive the most appropriate drugs and treatment schedules. In this context, there is significant potential for the use of pharmacodynamic biomarkers to provide pharmacological information, which could be used in a therapeutic drug monitoring setting. This review focuses on discussing some of the challenges faced to date in translating preclinical pharmacodynamic biomarker approaches to a clinical setting. Recent advances in important areas including circulating biomarkers and pharmacokinetic/pharmacodynamic modeling approaches are discussed, and selected examples of anticancer drugs where there is existing evidence to potentially advance pharmacodynamic therapeutic drug monitoring approaches to deliver more effective treatment are discussed. Although we may not yet be in a position to systematically implement therapeutic drug monitoring approaches based on pharmacodynamic information in a cancer patient setting, such approaches are likely to become more commonplace in the coming years. Based on ever-increasing levels of pharmacodynamic information being generated on newer anticancer drugs, facilitated by increasingly advanced and accessible experimental approaches available to researchers to collect these data, we can now look forward optimistically to significant advances being made in this area

    A reduced Gompertz model for predicting tumor age using a population approach

    Get PDF
    Tumor growth curves are classically modeled by ordinary differential equations. In analyzing the Gompertz model several studies have reported a striking correlation between the two parameters of the model. We analyzed tumor growth kinetics within the statistical framework of nonlinear mixed-effects (population approach). This allowed for the simultaneous modeling of tumor dynamics and inter-animal variability. Experimental data comprised three animal models of breast and lung cancers, with 843 measurements in 94 animals. Candidate models of tumor growth included the Exponential, Logistic and Gompertz. The Exponential and-more notably-Logistic models failed to describe the experimental data whereas the Gompertz model generated very good fits. The population-level correlation between the Gompertz parameters was further confirmed in our analysis (R 2 > 0.96 in all groups). Combining this structural correlation with rigorous population parameter estimation, we propose a novel reduced Gompertz function consisting of a single individual parameter. Leveraging the population approach using bayesian inference, we estimated the time of tumor initiation using three late measurement timepoints. The reduced Gompertz model was found to exhibit the best results, with drastic improvements when using bayesian inference as compared to likelihood maximization alone, for both accuracy and precision. Specifically, mean accuracy was 12.1% versus 74.1% and mean precision was 15.2 days versus 186 days, for the breast cancer cell line. These results offer promising clinical perspectives for the personalized prediction of tumor age from limited data at diagnosis. In turn, such predictions could be helpful for assessing the extent of invisible metastasis at the time of diagnosis. Author summary Mathematical models for tumor growth kinetics have been widely used since several decades but mostly fitted to individual or average growth curves. Here we compared three classical models (Exponential, Logistic and Gompertz) using a population approach, which accounts for inter-animal variability. The Exponential and the Logistic models failed to fit the experimental data while the Gompertz model showed excellent descriptive power. Moreover, the strong correlation between the two parameters of the Gompertz equation motivated a simplification of the model, the reduced Gompertz model, with a single individual parameter and equal descriptive power. Combining the mixed-effects approach with Bayesian inference, we predicted the age of individual tumors with only few late measurements. Thanks to its simplicity, the reduced Gompertz model showed superior predictive power. Although our method remains to be extended to clinical data, these results are promising for the personalized estimation of the age of a tumor from limited measurements at diagnosis. Such predictions could contribute to the development of computational models for metastasis

    A reduced Gompertz model for predicting tumor age using a population approach

    Get PDF
    Tumor growth curves are classically modeled by ordinary differential equations. In analyzing the Gompertz model several studies have reported a striking correlation between the two parameters of the model. We analyzed tumor growth kinetics within the statistical framework of nonlinear mixed-effects (population approach). This allowed for the simultaneous modeling of tumor dynamics and inter-animal variability. Experimental data comprised three animal models of breast and lung cancers, with 843 measurements in 94 animals. Candidate models of tumor growth included the Exponential, Logistic and Gompertz. The Exponential and-more notably-Logistic models failed to describe the experimental data whereas the Gompertz model generated very good fits. The population-level correlation between the Gompertz parameters was further confirmed in our analysis (R 2 > 0.96 in all groups). Combining this structural correlation with rigorous population parameter estimation, we propose a novel reduced Gompertz function consisting of a single individual parameter. Leveraging the population approach using bayesian inference, we estimated the time of tumor initiation using three late measurement timepoints. The reduced Gompertz model was found to exhibit the best results, with drastic improvements when using bayesian inference as compared to likelihood maximization alone, for both accuracy and precision. Specifically, mean accuracy was 12.1% versus 74.1% and mean precision was 15.2 days versus 186 days, for the breast cancer cell line. These results offer promising clinical perspectives for the personalized prediction of tumor age from limited data at diagnosis. In turn, such predictions could be helpful for assessing the extent of invisible metastasis at the time of diagnosis. Author summary Mathematical models for tumor growth kinetics have been widely used since several decades but mostly fitted to individual or average growth curves. Here we compared three classical models (Exponential, Logistic and Gompertz) using a population approach, which accounts for inter-animal variability. The Exponential and the Logistic models failed to fit the experimental data while the Gompertz model showed excellent descriptive power. Moreover, the strong correlation between the two parameters of the Gompertz equation motivated a simplification of the model, the reduced Gompertz model, with a single individual parameter and equal descriptive power. Combining the mixed-effects approach with Bayesian inference, we predicted the age of individual tumors with only few late measurements. Thanks to its simplicity, the reduced Gompertz model showed superior predictive power. Although our method remains to be extended to clinical data, these results are promising for the personalized estimation of the age of a tumor from limited measurements at diagnosis. Such predictions could contribute to the development of computational models for metastasis

    Monitoring of mAbs: is PK actionable?

    No full text
    International audienc
    corecore