17 research outputs found

    Lifestyle and Biological Risk Factors for Liver Fibrosis in the Miami Adult Studies on HIV (MASH) Cohort: An HIV Infected and HIV/HCV Co-infected Population

    Get PDF
    Liver disease is now a leading cause of non-AIDS related morbidity and mortality in people living with HIV (PLWH). The present study investigated the interplay between adverse lifestyle factors that are prevalent in PLWH, biological mediators of liver pathogenesis, and a non-invasive measure of liver fibrosis (FIB-4 index) in HIV mono- and HIV/HCV co-infected individuals. The results of this investigation in the Miami Adult Studies of HIV (MASH) cohort show that the odds of liver fibrosis progression significantly increased over two years for HIV mono-infected participants who drank alcohol hazardously (OR 3.038, P=0.048), and had BMI ≥ 28kg/m2 (OR 2.934, P=0.027). Cocaine use reduced the odds of advancing one stage of liver fibrosis (OR 0.228, P=0.038), but an interaction between high BMI and cocaine use slightly raised the odds by 4.8% of liver fibrosis progression (P=0.072). HIV/HCV co-infected participants showed interactions between cocaine use and high BMI with increased FIB-4 stage (OR 4.985, P= 0.034), however no lifestyle factors could independently predict FIB-4 stage in this group. Biological mediators previously associated with liver pathogenesis were associated with higher FIB-4 index over 2 years in a subset of (n=65) HIV mono-infected participants. Plasma measures of oxidative stress (% oxidized glutathione: OR 4.342, P= 0.046), hepatocyte-specific apoptosis (Cytokeratin-18 (CK-18): OR 1.008, P=0.021), and microbial endotoxin (lipopolysaccharide (LPS): OR 1.098, P= 0.097) were associated with having higher odds of progressing at least one stage of FIB-4 over 2 years. The same biological mediators were also associated with liver fibrosis within HIV infected people who also had a harmful lifestyle characteristic. FIB-4 index was significantly associated with % oxidized glutathione in obese subjects (β=0.563, P=0.018), TGF-β1 in cocaine users (β=0.858, P=0.027), and CK-18 in HIV infected individuals without any adverse lifestyle factors (β=0.435, P=0.015). Taken together, the findings of these studies describe interrelationships between HIV disease status, lifestyle, and biological mediators of liver fibrosis. The results show interactions between lifestyle conditions and the mediators of liver fibrosis may account for higher rates of liver disease in HIV infection. Research is warranted to develop personalized therapeutics for PLWH to curb the burden of liver disease

    Physics considerations in targeted anticancer drug delivery by magnetoelectric nanoparticles

    Get PDF
    In regard to cancer therapy, magnetoelectric nanoparticles (MENs) have proven to be in a class of its own when compared to any other nanoparticle type. Like conventional magnetic nanoparticles, they can be used for externally controlled drug delivery via application of a magnetic field gradient and image-guided delivery. However, unlike conventional nanoparticles, due to the presence of a non-zero magnetoelectric effect, MENs provide a unique mix of important properties to address key challenges in modern cancer therapy: (i) a targeting mechanism driven by a physical force rather than antibody matching, (ii) a high-specificity delivery to enhance the cellular uptake of therapeutic drugs across the cancer cell membranes only, while sparing normal cells, (iii) an externally controlled mechanism to release drugs on demand, and (iv) a capability for image guided precision medicine. These properties separate MEN-based targeted delivery from traditional biotechnology approaches and lay a foundation for the complementary approach of technobiology. The biotechnology approach stems from the underlying biology and exploits bioinformatics to find the right therapy. In contrast, the technobiology approach is geared towards using the physics of molecular-level interactions between cells and nanoparticles to treat cancer at the most fundamental level and thus can be extended to all the cancers. This paper gives an overview of the current state of the art and presents an ab initio model to describe the underlying mechanisms of cancer treatment with MENs from the perspective of basic physics

    Targeted and controlled anticancer drug delivery and release with magnetoelectric nanoparticles

    Get PDF
    It is a challenge to eradicate tumor cells while sparing normal cells. We used magnetoelectric nanoparticles (MENs) to control drug delivery and release. The physics is due to electric-field interactions (i) between MENs and a drug and (ii) between drug-loaded MENs and cells. MENs distinguish cancer cells from normal cells through the membrane’s electric properties; cancer cells have a significantly smaller threshold field to induce electroporation. In vitro and in vivo studies (nude mice with SKOV-3 xenografts) showed that (i) drug (paclitaxel (PTX)) could be attached to MENs (30-nm CoFe2O4@BaTiO3 nanostructures) through surface functionalization to avoid its premature release, (ii) drug-loaded MENs could be delivered into cancer cells via application of a d.c. field (~100 Oe), and (iii) the drug could be released off MENs on demand via application of an a.c. field (~50 Oe, 100 Hz). The cell lysate content was measured with scanning probe microscopy and spectrophotometry. MENs and control ferromagnetic and polymer nanoparticles conjugated with HER2-neu antibodies, all loaded with PTX were weekly administrated intravenously. Only the mice treated with PTX-loaded MENs (15/200 μg) in a field for three months were completely cured, as confirmed through infrared imaging and post-euthanasia histology studies via energy-dispersive spectroscopy and immunohistochemistry

    Survival analysis for white non-Hispanic female breast cancer patients

    No full text
    Background: Race and ethnicity are significant factors in predicting survival time of breast cancer patients. In this study, we applied advanced statistical methods to predict the survival of White non-Hispanic female breast cancer patients, who were diagnosed between the years 1973 and 2009 in the United States (U.S.). Materials and Methods: Demographic data from the Surveillance Epidemiology and End Results (SEER) database were used for the purpose of this study. Nine states were randomly selected from 12 U.S. cancer registries. A stratified random sampling method was used to select 2,000 female breast cancer patients from these nine states. We compared four types of advanced statistical probability models to identify the best-fit model for the White nonHispanic female breast cancer survival data. Three model building criterion were used to measure and compare goodness of fit of the models. These include Akaike Information Criteria (AIC), Bayesian Information Criteria (BIC), and Deviance Information Criteria (DIC). In addition, we used a novel Bayesian method and the Markov Chain Monte Carlo technique to determine the posterior density function of the parameters. After evaluating the model parameters, we selected the model having the lowest DIC value. Using this Bayesian method, we derived the predictive survival density for future survival time and its related inferences. Results: The analytical sample of White non-Hispanic women included 2,000 breast cancer cases from the SEER database (1973-2009). The majority of cases were married (55.2%), the mean age of diagnosis was 63.61 years (SD = 14.24) and the mean survival time was 84 months (SD = 35.01). After comparing the four statistical models, results suggested that the exponentiated Weibull model (DIC= 19818.220) was a better fit for White non-Hispanic females’ breast cancer survival data. This model predicted the survival times (in months) for White non-Hispanic women after implementation of precise estimates of the model parameters. Conclusions: By using modern model building criteria, we determined that the data best fit the exponentiated Weibull model. We incorporated precise estimates of the parameter into the predictive model and evaluated the survival inference for the White non-Hispanic female population. This method of analysis will assist researchers in making scientific and clinical conclusions when assessing survival time of breast cancer patients

    Abstract 2204: Targeted, controlled anticancer drug delivery and release with magnetoelectric nanoparticles

    No full text
    Abstract Most cancer therapy is nonspecific, aimed at all dividing cells, resulting in untoward side effects. Our study demonstrates the effectiveness of magnetoelectric nanoparticles (MENs), which have been developed to address the critical issue of normal cell off-targeting in cancer treatment, in both in-vitro and in-vivo studies, as well as characterizes their biodistribution and clearance. Exploiting the difference in electric properties between normal and cancer cell membranes, MENs are able to enter cancerous cells carrying a therapeutic payload and release the payload intracellularly with the application of an external magnetic field, while not affecting normal cells. SKOV-3 human ovarian carcinoma cells were used as a model to showcase the unique cancer targeting capabilities of these CoFe2O4@BaTiO3 nanostructures coated with the mitotic inhibitor Paclitaxel (PTX). The MENs-PTX bond was characterized in the lysate of treated cells using spectroscopic analysis and scanning probe microscopy. SKOV-3 xenografted athymic nude mice were treated via subcutaneous or IV injection on a weekly basis with a MEN, conventional ferromagnetic nanoparticle (MN), or polymer nanoparticle (PLGA) formulation. Biodistribution and clearance of MENs is one of the most important open questions addressed in this study. Our approach is to investigate the key parameters that affect the therapeutic index, i.e. the maximum tolerated dose, blood circulation half-life and biodistribution due organ accumulation. The approach is to study factors such as the size and shape of MENs, chemical composition, targeting ligand functionalization, MENs’ biodegradability, and microenvironment and other biological barriers. Besides using conventional fluorescent markers, a novel nanoparticle distribution approach based on energy-dispersion spectroscopy (EDS) is exploited. In-vitro studies on the cell lysate of MENs treated SKOV-3 cells determined reliable entry into the cells by MENs with the application of a small magnetic field (∼100 Oe) and reliable payload release with the application of an a.c. magnetic field (∼50 Oe, 100 Hz). In-vivo studies demonstrated that the MENs-PTX formulation in combination with an externally applied magnetic field reduces tumor growth rate when injected subcutaneously, and fully cures the cancer when delivered via IV-injection. The MENs formulation was more successful in treating the tumor than both MN and PLGA formulations. EDS confirmed the presence of MENs in tumor tissues. MENs provide a novel mechanism by which cancer cells are targeted (using the difference in the cancer electric cell membrane properties compared to normal cells) and a drug payload is released (externally triggered with the application of an a.c. magnetic field) reliably. The underlying physics of the electric field interactions involved in the MENs drug delivery system was demonstrated here using ovarian cancer, but can be applied to virtually any cancer. Citation Format: Alexandra Rodzinski, Rakesh Guduru, Emmanuel Stimphil, Tiffanie Stewart, Ping Liang, Carolyn Runowicz, Sakhrat Khizroev. Targeted, controlled anticancer drug delivery and release with magnetoelectric nanoparticles. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2204

    Abstract B47: A novel mechanism for field-controlled high-specificity targeted anticancer drug delivery and on-demand release using magnetoelectric nanoparticles

    No full text
    Abstract Background: An important challenge in chemotherapy is targeted drug delivery to eradicate tumor cells while sparing normal cells. The circulatory system can deliver a drug to almost every cell in the body; however, delivering the drug specifically into the tumor cell and then releasing it on demand remains a formidable task. Nanoparticles posses unique properties to address this issue. Despite their great potential, a significant problem remains to ensure that the drug is not prematurely released in the plasma or interstitial space but is released at an appropriate rate once at the target site. Recently, we discovered a new class of “smart” multifunctional nanostructures known as magnetoelectric nanoparticles (MENs) that enables a high-efficacy “communication” between intrinsic electric fields at the intra-cellular level, which are inherent to the cellular membrane nature, and external magnetic fields, to control targeted drug delivery and release into specific tumor cells on demand. Herein, the results of a comprehensive in vitro study and an in vivo study on using MENs to treat ovarian cancer (OC) are presented. Methods: A specific combination of d.c. and a.c.-magnetic fields is used to externally control and separate delivery and release functions, respectively. MENs in a wide diameter range, 5-1000nm, are made of coreshell CoFe2O4@BaTiO3 nanostructures. The novel approach is compared to current state-of-the-art nanotechnology deliveries including (i) active immunochemotherapeutic approaches using polymer nanoparticles conjugated with monoclonal antibodies (mAbs) and (ii) passive enhanced permeability and retention (EPR)-based approach using polymer nanoparticles without any immunoactive reagents. Mitotic inhibitor paclitaxel (PTX)–loaded MENs are administrated through systemic IV injection into a lateral tail vein or through localized subcutaneous injection directly into the tumor site. The tumor progression is monitored through infrared (IR) imaging witth mAb-conjugated fluorescent agent Her2Sense 645. Post euthanasia, the cell morphology and the tumor presence in different organs are further studied with H&E stain and Her2Sense agent, respectively. The biodistribution of the nanoparticles in the tumor sites and different organs of mice treated under different field conditions are studied through the energy-dispersive spectroscopy (EDS) mode of high-resolution scanning electron microscopy (SEM). Finally, after the completion of the treatment, the cured mice are monitored for a period of three months before being sacrificed for further immunohistochemical and particle biodistribution studies. Results: Using MENs loaded with PTX, the in vivo study on nude mice bearing SKOV-3 human ovarian carcinoma xenografts shows that intravenously administrated MENs enable high-specificity delivery and release via application of d.c. and a.c. magnetic fields, respectively. MENs distinguish cancer cells from the normal counterparts by the difference in the membrane’s electric properties. The control mice are treated with PTX loaded on ferromagnetic and polymer nanoparticles conjugated with HER2-neu antibodies. Only the mice which are weekly treated for three months with PTX-loaded 30-nm MENs (15/200 µg) in a 100-Oe local field are completely cured, as confirmed through infrared imaging and post-euthanasia immunohistochemical analysis. A comparison between systemic IV and localized subcutaneous injections of PTX-loaded MENs show that although both delivery approaches significantly slow down the progression of the tumor, the IV administration is more efficient and, unlike the subcutaneous administration, can completely eradicate the tumor during the three-month treatment period. An important observation from this study is the strong dependence of the amount of MENs in the tumor site and all the organs of treated and control mice on the external magnetic field control. The same conclusion was drawn also from the comprehensive in vitro study, in which the detailed cell lysate content was measured using direct AFM/MFM imaging of MENs and spectrophotometry detection of the drug under the investigated field conditions. Conclusion: In summary, the study directly shows that the drug-loaded MENs provide a novel way to deliver the drug specifically into the tumor site via application of a d.c. field and then, when at the site, the drug is released directly inside the cancer cells via application of an a.c. field. Citation Format: Alexandra Rodzinski, Ali Hadjikhani, Tiffanie Stewart, Emmanuel Stimphil, Rakesh Guduru, Ping Liang, Carolyn Runowicz, Sakhrat Khizroev. A novel mechanism for field-controlled high-specificity targeted anticancer drug delivery and on-demand release using magnetoelectric nanoparticles. [abstract]. In: Proceedings of the Fourth AACR International Conference on Frontiers in Basic Cancer Research; 2015 Oct 23-26; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2016;76(3 Suppl):Abstract nr B47

    Abstract 2199: Magnetoelectric nanoparticles for high-specificity treatment of cancer

    No full text
    Abstract Introduction: Delivering a drug specifically into the tumor cell past its membrane and then releasing the drug into the tumor cells without affecting the normal cells remains a formidable challenge. Unlike any other nanoparticles, magnetoelectric nanoparticles (MENs) display a non-zero magnetoelectric (ME) effect and thus present a unique capability to use external magnetic fields to control intrinsic electric fields associated with cell membranes and the interaction between MENs and therapeutic loads. Because cancer and normal cells of the same type have different electric properties, MENs is used for high-specificity targeted delivery. An a.c. magnetic field is used to trigger drug release off the nanoparticles. Brief Methods: 30-nm CoFe2O4-BaTiO3 core shell MENs, with a magnetization of 1 emu/g, a coercivity of 300 Oe, and a ME coefficient of 10-100 mV cm-1 Oe-1 were prepared with a coprecipitation process. MENs were coated with fluorescein isothiocyanate to monitor their intra-cellular transport through a high-contrast confocal microscopy. Three cancer cell lines including Skov-3 (Ovarian adenocarcinoma), U87-MG (Glioblastoma), and MCF-7A (Breast adenocarcinoma), and two normal cell lines including brain endothelial cells (Brain EC) and ovarian cells HOMEC were cultured at 37°C. The transport of MENs loaded with drugs, peptides, and RNAs through the cell membranes and the consequent release of the load under different d.c. and a.c. magnetic fields were studied through confocal microscopy and photoabsorption spectroscopy, respectively. Trypan-blue viability count was used to assess cell growth inhibition under different study conditions. Atomic force microscopy of the cell membranes was conducted to understand the interaction between the nanoparticles and the cells. Summary of new data: Comparison of MENs with purely magnetic iron oxide nanoparticles showed that the penetration through the cancer cell membrane could be achieved only with MENs. It took d.c. fields of 100 Oe and over 1000 Oe to nanoelectroporate the membranes of SKOV-3 and HOMEC cell lines, respectively. An a.c. magnetic field with a strength of 50 Oe and a near-d.c. frequency of 100 Hz was sufficient to enable release of a therapeutic load off MENs. All the cancer cell lines under study showed membrane penetration threshold d.c. fields at least a factor of ten smaller compared to their normal counterparts. Conclusion: MENs displayed unique capabilities for externally controlled high-specificity targeted anticancer drug delivery and release on demand via application of d.c. and a.c. magnetic fields, respectively. Because MENs rely on a physical mechanism rather than antibody-mediated delivery, they can be used for high specificity delivery and high-efficacy controlled release of a broad range of therapeutic loads including drugs, peptides, and RNAs to treat many different cancers. Citation Format: Emmanuel Stimphil, Abhi Nagesetti, Tiffanie S. Stewart, Alexa Rodzinski, Rakesh Guduru, Ping Liang, Carolyn Runowicz, Sakhrat Khizroev. Magnetoelectric nanoparticles for high-specificity treatment of cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2199. doi:10.1158/1538-7445.AM2017-219

    Targeted and controlled anticancer drug delivery and release with magnetoelectric nanoparticles

    No full text
    It is a challenge to eradicate tumor cells while sparing normal cells. We used magnetoelectric nanoparticles (MENs) to control drug delivery and release. The physics is due to electric-field interactions (i) between MENs and a drug and (ii) between drug-loaded MENs and cells. MENs distinguish cancer cells from normal cells through the membrane’s electric properties; cancer cells have a significantly smaller threshold field to induce electroporation. In vitro and in vivo studies (nude mice with SKOV-3 xenografts) showed that (i) drug (paclitaxel (PTX)) could be attached to MENs (30-nm CoFe2O4@BaTiO3 nanostructures) through surface functionalization to avoid its premature release, (ii) drug-loaded MENs could be delivered into cancer cells via application of a d.c. field (~100 Oe), and (iii) the drug could be released off MENs on demand via application of an a.c. field (~50 Oe, 100 Hz). The cell lysate content was measured with scanning probe microscopy and spectrophotometry. MENs and control ferromagnetic and polymer nanoparticles conjugated with HER2-neu antibodies, all loaded with PTX were weekly administrated intravenously. Only the mice treated with PTX-loaded MENs (15/200 μg) in a field for three months were completely cured, as confirmed through infrared imaging and post-euthanasia histology studies via energy-dispersive spectroscopy and immunohistochemistry
    corecore