4,641 research outputs found

    Review of precision cancer medicine: Evolution of the treatment paradigm.

    Get PDF
    In recent years, biotechnological breakthroughs have led to identification of complex and unique biologic features associated with carcinogenesis. Tumor and cell-free DNA profiling, immune markers, and proteomic and RNA analyses are used to identify these characteristics for optimization of anticancer therapy in individual patients. Consequently, clinical trials have evolved, shifting from tumor type-centered to gene-directed, histology-agnostic, with innovative adaptive design tailored to biomarker profiling with the goal to improve treatment outcomes. A plethora of precision medicine trials have been conducted. The majority of these trials demonstrated that matched therapy is associated with superior outcomes compared to non-matched therapy across tumor types and in specific cancers. To improve the implementation of precision medicine, this approach should be used early in the course of the disease, and patients should have complete tumor profiling and access to effective matched therapy. To overcome the complexity of tumor biology, clinical trials with combinations of gene-targeted therapy with immune-targeted approaches (e.g., checkpoint blockade, personalized vaccines and/or chimeric antigen receptor T-cells), hormonal therapy, chemotherapy and/or novel agents should be considered. These studies should target dynamic changes in tumor biologic abnormalities, eliminating minimal residual disease, and eradicating significant subclones that confer resistance to treatment. Mining and expansion of real-world data, facilitated by the use of advanced computer data processing capabilities, may contribute to validation of information to predict new applications for medicines. In this review, we summarize the clinical trials and discuss challenges and opportunities to accelerate the implementation of precision oncology

    Robust Individual Circadian Parameter Estimation for Biosignal-based Personalisation of Cancer Chronotherapy

    Full text link
    In cancer treatment, chemotherapy is administered according a constant schedule. The chronotherapy approach, considering chronobiological drug delivery, adapts the chemotherapy profile to the circadian rhythms of the human organism. This reduces toxicity effects and at the same time enhances efficiency of chemotherapy. To personalize cancer treatment, chemotherapy profiles have to be further adapted to individual patients. Therefore, we present a new model to represent cycle phenomena in circadian rhythms. The model enables a more precise modelling of the underlying circadian rhythms. In comparison with the standard model, our model delivers better results in all defined quality indices. The new model can be used to adapt the chemotherapy profile efficiently to individual patients. The adaption to individual patients contributes to the aim of personalizing cancer therapy.Comment: Conference Biosig 2016, Berli

    Systems approaches and algorithms for discovery of combinatorial therapies

    Full text link
    Effective therapy of complex diseases requires control of highly non-linear complex networks that remain incompletely characterized. In particular, drug intervention can be seen as control of signaling in cellular networks. Identification of control parameters presents an extreme challenge due to the combinatorial explosion of control possibilities in combination therapy and to the incomplete knowledge of the systems biology of cells. In this review paper we describe the main current and proposed approaches to the design of combinatorial therapies, including the empirical methods used now by clinicians and alternative approaches suggested recently by several authors. New approaches for designing combinations arising from systems biology are described. We discuss in special detail the design of algorithms that identify optimal control parameters in cellular networks based on a quantitative characterization of control landscapes, maximizing utilization of incomplete knowledge of the state and structure of intracellular networks. The use of new technology for high-throughput measurements is key to these new approaches to combination therapy and essential for the characterization of control landscapes and implementation of the algorithms. Combinatorial optimization in medical therapy is also compared with the combinatorial optimization of engineering and materials science and similarities and differences are delineated.Comment: 25 page

    Xenograft Models for Preclinical Assessment of Anticancer Therapies: A Comprehensive Review

    Get PDF
    Introduction: Xenograft models play a pivotal role in preclinical studies for assessing the efficacy of anticancer medications. In this comprehensive review, we present an overview of current advancements and future prospects in xenograft research, focusing on their significance in guiding drug development and clinical translation. Aim: Our aim is to conduct an in-depth review of xenograft models, their utility in evaluating anticancer drug effectiveness and ultimately improve patient outcomes. Methods We conducted an in-depth literature search using databases such as ScienceDirect, Google Scholar and PubMed with keywords including "xenograft model, cancer CDX PDX." We then reviewed and analyzed relevant studies that utilized xenograft models in order to highlight key findings and contributions made through such models. Results: Our analysis showcases the essential role of xenograft models in assessing the efficacy of anticancer drugs. We discuss the benefits and limitations of these models, emphasizing their importance in guiding drug development and clinical decision-making. Conclusion: Xenograft models remain invaluable tools in preclinical cancer research despite their inherent limitations, with researchers continually striving to refine and enhance these models to ensure their reliability in an ever-evolving field of cancer therapeutics. Utilizing xenograft models allows researchers to evaluate anticancer drug activity more accurately while striving for improved patient outcomes

    Systems chronotherapeutics

    Get PDF
    Chronotherapeutics aim at treating illnesses according to the endogenous biologic rhythms, which moderate xenobiotic metabolism and cellular drug response. The molecular clocks present in individual cells involve approximately fifteen clock genes interconnected in regulatory feedback loops. They are coordinated by the suprachiasmatic nuclei, a hypothalamic pacemaker, which also adjusts the circadian rhythms to environmental cycles. As a result, many mechanisms of diseases and drug effects are controlled by the circadian timing system. Thus, the tolerability of nearly 500 medications varies by up to fivefold according to circadian scheduling, both in experimental models and/or patients. Moreover, treatment itself disrupted, maintained, or improved the circadian timing system as a function of drug timing. Improved patient outcomes on circadian-based treatments (chronotherapy) have been demonstrated in randomized clinical trials, especially for cancer and inflammatory diseases. However, recent technological advances have highlighted large interpatient differences in circadian functions resulting in significant variability in chronotherapy response. Such findings advocate for the advancement of personalized chronotherapeutics through interdisciplinary systems approaches. Thus, the combination of mathematical, statistical, technological, experimental, and clinical expertise is now shaping the development of dedicated devices and diagnostic and delivery algorithms enabling treatment individualization. In particular, multiscale systems chronopharmacology approaches currently combine mathematical modeling based on cellular and whole-body physiology to preclinical and clinical investigations toward the design of patient-tailored chronotherapies. We review recent systems research works aiming to the individualization of disease treatment, with emphasis on both cancer management and circadian timing system–resetting strategies for improving chronic disease control and patient outcomes

    Sex and gender differences in anticancer treatment toxicity - a call for revisiting drug dosing in oncology.

    Get PDF
    The practice of oncology has dramatically changed in the last decade with the introduction of molecular tumor profiling into routine tumor diagnostics and the extraordinary progress in immunotherapies. However, there remains an unmet need to explore personalized dosing strategies that take into account the patient's sex to optimize the balance between efficacy and toxicity for each individual patient. In this mini-review, we summarize the evidence on sex differences in toxicity of anticancer therapies and present data on dose reduction and dose discontinuation rates for selected chemotherapies and targeted therapies. Finally, we propose the investigation of body composition (specifically fat free muscle mass) as a viable approach for personalized treatment dosage

    Head and neck cancer: metronomic chemotherapy

    Get PDF
    In the era of personalized medicine, head and neck squamous cell carcinoma (HNSCC) represents a critical oncologic topic. Conventional chemotherapy regimens consist of drugs administration in cycles near or at the maximum tolerated dose (MDT), followed by a long drug-free period to permit the patient to recover from acute toxicities. Despite this strategy is successful in controlling the cancer process at the beginning, a significant number of HNSCC patients tend to recurred or progress, especially those patients with locally advanced or metastatic disease. The repertoire of drugs directed against tumor cells has greatly increased and metronomic chemotherapy (MC) could be an effective treatment option.It is the purpose of this article to review the concept of MC and describe its potential use in HNSCC. We provide an update of ongoing progress and current challenges related to this issue

    An implantable microdevice to perform high-throughput in vivo drug sensitivity testing in tumors

    Get PDF
    Current anticancer chemotherapy relies on a limited set of in vitro or indirect prognostic markers of tumor response to available drugs. A more accurate analysis of drug sensitivity would involve studying tumor response in vivo. To this end, we have developed an implantable device that can perform drug sensitivity testing of several anticancer agents simultaneously inside the living tumor. The device contained reservoirs that released microdoses of single agents or drug combinations into spatially distinct regions of the tumor. The local drug concentrations were chosen to be representative of concentrations achieved during systemic treatment. Local efficacy and drug concentration profiles were evaluated for each drug or drug combination on the device, and the local efficacy was confirmed to be a predictor of systemic efficacy in vivo for multiple drugs and tumor models. Currently, up to 16 individual drugs or combinations can be assessed independently, without systemic drug exposure, through minimally invasive biopsy of a small region of a single tumor. This assay takes into consideration physiologic effects that contribute to drug response by allowing drugs to interact with the living tumor in its native microenvironment. Because these effects are crucial to predicting drug response, we envision that these devices will help identify optimal drug therapy before systemic treatment is initiated and could improve drug response prediction beyond the biomarkers and in vitro and ex vivo studies used today. These devices may also be used in clinical drug development to safely gather efficacy data on new compounds before pharmacological optimization.National Cancer Institute (U.S.) (Innovative Molecular Analysis Technologies Program R21-CA177391)Kibur Medical, Inc
    • …
    corecore