2,610 research outputs found

    Irreversible Electroporation (IRE): Preclinical Assessment

    Get PDF
    Cancer Systems Imaginghttps://openworks.mdanderson.org/sumexp22/1079/thumbnail.jp

    Multimodality imaging in vivo for preclinical assessment of tumor-targeted doxorubicin nanoparticles.

    Get PDF
    This study presents a new multimodal imaging approach that includes high-frequency ultrasound, fluorescence intensity, confocal, and spectral imaging to improve the preclinical evaluation of new therapeutics in vivo. Here we use this approach to assess in vivo the therapeutic efficacy of the novel chemotherapy construct, HerDox during and after treatment. HerDox is comprised of doxorubicin non-covalently assembled in a viral-like particle targeted to HER2+ tumor cells, causing tumor cell death at over 10-fold lower dose compared to the untargeted drug, while sparing the heart. Whereas our initial proof-of-principle studies on HerDox used tumor growth/shrinkage rates as a measure of therapeutic efficacy, here we show that multimodal imaging deployed during and after treatment can supplement traditional modes of tumor monitoring to further characterize the particle in tissues of treated mice. Specifically, we show here that tumor cell apoptosis elicited by HerDox can be monitored in vivo during treatment using high frequency ultrasound imaging, while in situ confocal imaging of excised tumors shows that HerDox indeed penetrated tumor tissue and can be detected at the subcellular level, including in the nucleus, via Dox fluorescence. In addition, ratiometric spectral imaging of the same tumor tissue enables quantitative discrimination of HerDox fluorescence from autofluorescence in situ. In contrast to standard approaches of preclinical assessment, this new method provides multiple/complementary information that may shorten the time required for initial evaluation of in vivo efficacy, thus potentially reducing the time and cost for translating new drug molecules into the clinic

    Preclinical Assessment of HIV Vaccines and Microbicides by Repeated Low-Dose Virus Challenges

    Get PDF
    BACKGROUND: Trials in macaque models play an essential role in the evaluation of biomedical interventions that aim to prevent HIV infection, such as vaccines, microbicides, and systemic chemoprophylaxis. These trials are usually conducted with very high virus challenge doses that result in infection with certainty. However, these high challenge doses do not realistically reflect the low probability of HIV transmission in humans, and thus may rule out preventive interventions that could protect against “real life” exposures. The belief that experiments involving realistically low challenge doses require large numbers of animals has so far prevented the development of alternatives to using high challenge doses. METHODS AND FINDINGS: Using statistical power analysis, we investigate how many animals would be needed to conduct preclinical trials using low virus challenge doses. We show that experimental designs in which animals are repeatedly challenged with low doses do not require unfeasibly large numbers of animals to assess vaccine or microbicide success. CONCLUSION: Preclinical trials using repeated low-dose challenges represent a promising alternative approach to identify potential preventive interventions

    Determining the relationship between nanoparticle characteristics and immunotoxicity: key challenges and approaches

    Get PDF
    The growing wealth of information regarding the influence that physicochemical characteristics play on nanoparticle biocompatibility and safety is allowing improved design and rationale for their development and preclinical assessment. Accurate and appropriate measurement of these characteristics accompanied by informed toxicological assessment is a necessity for the development of safe and effective nanomedicines. While particle type, formulation and mode of administration dictate the individual causes for concern through development, the benefits of nanoformulation for treatment of the diseased state are great. Here we have proposed certain considerations and suggestions, which could lead to better-informed preclinical assessment of nanomaterials for nanomedicine, as well as how this information can and should be extrapolated to the physiological state of the end user

    Cross-species gene expression analysis of species specific differences in the preclinical assessment of pharmaceutical compounds

    Get PDF
    Animals are frequently used as model systems for determination of safety and efficacy in pharmaceutical research and development. However, significant quantitative and qualitative differences exist between humans and the animal models used in research. This is as a result of genetic variation between human and the laboratory animal. Therefore the development of a system that would allow the assessment of all molecular differences between species after drug exposure would have a significant impact on drug evaluation for toxicity and efficacy. Here we describe a cross-species microarray methodology that identifies and selects orthologous probes after cross-species sequence comparison to develop an orthologous cross-species gene expression analysis tool. The assumptions made by the use of this orthologous gene expression strategy for cross-species extrapolation is that; conserved changes in gene expression equate to conserved pharmacodynamic endpoints. This assumption is supported by the fact that evolution and selection have maintained the structure and function of many biochemical pathways over time, resulting in the conservation of many important processes. We demonstrate this cross-species methodology by investigating species specific differences of the peroxisome proliferatoractivator receptor (PPAR) a response in rat and human

    The transcription factor ATF5: role in cellular differentiation, stress responses, and cancer.

    Get PDF
    Activating transcription factor 5 (ATF5) is a cellular prosurvival transcription factor within the basic leucine zipper (bZip) family that is involved in cellular differentiation and promotes cellular adaptation to stress. Recent studies have characterized the oncogenic role of ATF5 in the development of several different types of cancer, notably glioblastoma. Preclinical assessment of a systemically deliverable dominant-negative ATF5 (dnATF5) biologic has found that targeting ATF5 results in tumor regression and tumor growth inhibition of glioblastoma xenografts in mouse models. In this review, we comprehensively and critically detail the current scientific literature on ATF5 in the context of cellular differentiation, survival, and response to stressors in normal tissues. Furthermore, we will discuss how the prosurvival role of ATF5 aides in cancer development, followed by current advances in targeting ATF5 using dominant-negative biologics, and perspectives on future research

    Clinical responses to adoptive T-cell transfer can be modeled in an autologous immune-humanized mouse model

    Get PDF
    Combining different types of immune therapies might benefit certain patients. Here, the authors develop an autologous immune-humanized melanoma mouse model that allows the preclinical assessment of cancer cell–T cell interactions from each individual patient and the benefits of immunotherapies combinations

    Multispectral imaging for preclinical assessment of rheumatoid arthritis models

    Get PDF
    Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune condition affecting multiple body systems. Murine models of RA are vital in progressing understanding of the disease. The severity of arthritis symptoms is currently assessed in vivo by observations and subjective scoring which are time-consuming and prone to bias and inaccuracy. The main aim of this thesis is to determine whether multispectral imaging of murine arthritis models has the potential to assess the severity of arthritis symptoms in vivo in an objective manner. Given that pathology can influence the optical properties of a tissue, changes may be detectable in the spectral response. Monte Carlo modelling of reflectance and transmittance for varying levels of blood volume fraction, blood oxygen saturation, and water percentage in the mouse paw tissue demonstrated spectral changes consistent with the reported/published physiological markers of arthritis. Subsequent reflectance and transmittance in vivo spectroscopy of the hind paw successfully detected significant spectral differences between normal and arthritic mice. Using a novel non-contact imaging system, multispectral reflectance and transmittance images were simultaneously collected, enabling investigation of arthritis symptoms at different anatomical paw locations. In a blind experiment, Principal Component (PC) analysis of four regions of the paw was successful in identifying all 6 arthritic mice in a total sample of 10. The first PC scores for the TNF dARE arthritis model were found to correlate significantly with bone erosion ratio results from microCT, histology scoring, and the manual scoring method. In a longitudinal study at 5, 7 and 9 weeks the PC scores identified changes in spectral responses at an early stage in arthritis development for the TNF dARE model, before clinical signs were manifest. Comparison of the multispectral image data with the Monte Carlo simulations suggest that in this study decreased oxygen saturation is likely to be the most significant factor differentiating arthritic mice from their normal littermates. The results of the experiments are indicative that multispectral imaging performs well as an assessor of arthritis for RA models and may outperform existing techniques. This has implications for better assessment of preclinical arthritis and hence for better experimental outcomes and improvement of animal welfare

    Preclinical assessment of ulixertinib, a novel ERK1/2 inhibitor

    Get PDF
    Ulixertinib (BVD-523) is a novel and selective reversible inhibitor of ERK1/ERK2. In xenograft studies it inhibited tumor growth in BRAF-mutant melanoma and colorectal xenografts as well as KRAS-mutant colorectal and pancreatic models. Ulixertinib is currently in Phase I clinical development for the treatment of advance solid tumors. The objective of the study is to assess the metabolic stability (in various pre-clinical and human liver microsomes/hepatocytes), permeability, protein binding, CYP inhibition, CYP induction and CYP phenotyping of ulixertinib. We have also studied the oral and intravenous pharmacokinetics of ulixertinib in mice, rats and dogs. Ulixertinib was found to be moderately to highly stable in various liver microsomes/hepatocytes tested. It is a medium permeable (2.67 x 10-6 cm /sec) drug and a substrate for efflux (efflux ratio: 3.02) in Caco-2 model. Ulixertinib was highly bound to plasma proteins. CYPs involved in its limited metabolism and it is CYP inhibition IC50 ranged between 10-20 μM. Post oral administration ulixertinib exhibited early Tmax (0.50-0.75 h) in mice and rats indicating absorption was rapid, however in dogs Tmax attained at 2 h. The half-life (t½) of ulixertinib by intravenous and oral routes ranged between 1.0-2.5 h across the species. Clearance and volume of distribution by intravenous route for ulixertinib were found to be 6.24 mL/min/kg and 0.56 L/kg; 1.67 mL/min/kg and 0.36 L/kg and 15.5 mL/min/kg and 1.61 L/kg in mice, rats and dogs, respectively. Absolute oral bioavailability in mice and rats was > 92 %, however in dogs it was 34 %

    Novel approaches towards cancer-directed immune checkpoint inhibition

    Get PDF
    Blockade of the PD-1/PD-L1 interaction using immune checkpoint-inhibiting antibodies has yielded unprecedented clinical responses, albeit only in a small subgroup of cancer types. Moreover, treatment with these antibodies is frequently associated with serious auto-immune-related side effects due to 'on-target/off-tumor' binding to PD-L1 present on normal cells. Additionally, cancer cells are known to excrete large amounts of small extracellular vesicles called ‘tumor exosomes’ (TEX), which can expose large amounts of PD-L1. Notably, TEX-exposed immune checkpoint molecules are largely resistant to current clinically used checkpoint-inhibiting antibodies. Taken together, there is an unmet clinical need for novel checkpoint inhibitors that can selectively block PD-L1 and alternate immune checkpoints both on cancer cells and TEX. This thesis describes the construction and preclinical assessment of a novel series of bispecific antibodies (bsAbs) designed to selectively inhibit both cancer cell- and EV-exposed immune checkpoints (e.g. PD-L1, CD73, or CD47). Additionally, we describe the synthesis and subsequent preclinical assessment of a novel light-activatable small-molecule inhibitor (SMI) of the CD73 immune checkpoint
    corecore