227 research outputs found

    Optimisation of time-scheduled regimen for anti-cancer drug infusion

    Get PDF
    The chronotherapy concept takes advantage of the circadian rhythm of cells physiology in maximising a treatment efficacy on its target while minimising its toxicity on healthy organs. The object of the present paper is to investigate mathematically and numerically optimal strategies in cancer chronotherapy. To this end a mathematical model describing the time evolution of efficiency and toxicity of an oxaliplatin anti-tumour treatment has been derived. We then applied an optimal control technique to search for the best drug infusion laws. The mathematical model is a set of six coupled differential equations governing the time evolution of both the tumour cell population (cells of Glasgow osteosarcoma, a mouse tumour) and the mature jejunal enterocyte population, to be shielded from unwanted side effects during a treatment by oxaliplatin. Starting from known tumour and villi populations, and a time dependent free platinum Pt (the active drug) infusion law being given, the mathematical model allows to compute the time evolution of both tumour and villi populations. The tumour population growth is based on Gompertz law and the Pt anti-tumour efficacy takes into account the circadian rhythm. Similarly the enterocyte population is subject to a circadian toxicity rhythm. The model has been derived using, as far as possible, experimental data. We examine two different optimisation problems. The eradication problem consists in finding the drug infusion law able to minimise the number of tumour cells while preserving a minimal level for the villi population. On the other hand, the containment problem searches for a quasi periodic treatment able to maintain the tumour population at the lowest possible level, while preserving the villi cells. The originality of these approaches is that the objective and constraint functions we use are L∞ criteria. We are able to derive their gradients with respect to the infusion rate and then to implement efficient optimisation algorithms

    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

    Dosing-time makes the poison : circadian regulation and pharmacotherapy

    Get PDF
    Daily rhythms in physiology significantly modulate drug pharmacokinetics and pharmacodynamics according to the time-of-day, a finding that has led to the concept of chronopharmacology. The importance of biological clocks for xenobiotic metabolism has gained increased attention with the discovery of the molecular circadian clockwork. Mechanistic understanding of the cell-autonomous molecular circadian oscillator and the circadian timing system as a whole has opened new conceptual and methodological lines of investigation to understand first, the clock's impact on a specific drug's daily variations or the effects/side effects of environmental substances, and second, how clock-controlled pathways are coordinated within a given tissue or organism. Today, there is an increased understanding of the circadian modulation of drug effects. Moreover, several molecular strategies are being developed to treat disease-dependent and drug-induced clock disruptions in humans

    Clock gene Per2 as a controller of liver carcinogenesis

    Get PDF
    Environmental disruption of molecular clocks promoted liver carcinogenesis and accelerated cancer progression in rodents. We investigated the specific role of clock gene Period 2 (Per2) for liver carcinogenesis and clock-controlled cellular proliferation, genomic instability and inflammation. We assessed liver histopathology, and determined molecular and physiology circadian patterns in mice on chronic diethylnitrosamine (DEN) exposure according to constitutive Per2 mutation. First, we found that Per2m/m liver displayed profound alterations in proliferation gene expression, including c-Myc derepression, phase-advanced Wee1, and arrhythmic Ccnb1 and K-ras mRNA expressions, as well as deregulated inflammation, through arrhythmic liver IL-6 protein concentration, in the absence of any DEN exposure. These changes could then make Per2m/m mice more prone to subsequently develop liver cancers on DEN. Indeed, primary liver cancers were nearly fourfold as frequent in Per2m/m mice as compared to wild-type (WT), 4 months after DEN exposure. The liver molecular clock was severely disrupted throughout the whole carcinogenesis process, including the initiation stage, i.e. within the initial 17 days on DEN. Per2m/m further exhibited increased c-Myc and Ccnb1 mean 24h expressions, lack of P53 response, and arrhythmic ATM, Wee1 and Ccnb1 expressions. DEN-induced tumor related inflammation was further promoted through increased protein concentrations of liver IL-6 and TNF-α as compared to WT during carcinogenesis initiation. Per2 mutation severely deregulated liver gene or protein expressions related to three cancer hallmarks, including uncontrolled proliferation, genomic instability, and tumor promoting inflammation, and accelerated liver carcinogenesis several-fold. Clock gene Per2 acted here as a liver tumor suppressor from initiation to progression

    Predictability of individual circadian phase during daily routine for medical applications of circadian clocks

    Get PDF
    Background: Circadian timing of treatments can largely improve tolerability and efficacy in patients. Thus, drug metabolism and cell cycle are controlled by molecular clocks in each cell, and coordinated by the core body temperature 24-hour rhythm, which is generated by the hypothalamic pacemaker. Individual circadian phase is currently estimated with questionnaire-based chronotype, center-of-rest time, dim light melatonin onset (DLMO), or timing of CBT maximum (acrophase) or minimum (bathyphase). Methods: We aimed at circadian phase determination and read-out during daily routine in volunteers stratified by sex and age. We measured (i) chronotype; (ii) q1min CBT using two electronic pills swallowed 24-hours apart; (iii) DLMO through hourly salivary samples from 18:00 to bedtime; (iv) q1min accelerations and surface temperature at anterior chest level for seven days, using a tele-transmitting sensor. Circadian phases were computed using cosinor and Hidden-Markov modelling. Multivariate regression identified the combination of biomarkers that best predicted core temperature circadian bathyphase. Results: Amongst the 33 participants, individual circadian phases were spread over 5h10min (DLMO), 7h (CBT bathyphase) and 9h10 min (surface temperature acrophase). CBT bathyphase was accurately predicted, i.e. with an error <1h for 78.8% of the subjects, using a new digital health algorithm (INTime), combining time-invariant sex and chronotype score, with computed center-of-rest time and surface temperature bathyphase (adjusted R-squared = 0.637). Conclusion: INTime provided a continuous and reliable circadian phase estimate in real time. This model helps integrate circadian clocks into precision medicine and will enable treatment timing personalisation following further validation

    Optimal drug infusion strategies for cancer chronotherapy

    Get PDF
    The chronotherapy concept takes advantage of the circadian rhythm of cells physiology in maximising a treatment efficacy on its target while minimising its toxicity on healthy organs. The object of the present paper is to investigate mathematically and numerically optimal strategies in cancer chronotherapy. To this end a mathematical model describing the time evolution of efficiency and toxicity of an oxaliplatin anti-tumour treatment has been derived. We then applied an optimal control technique to search for the best drug infusion laws. The mathematical model is a set of six coupled differential equations governing the time evolution of both the tumour cell population (cells of a Glasgow osteosarcoma, a mouse tumour) and the mature jejunal enterocyte population, to be shielded from unwanted side effects during a treatment by oxaliplatin. Starting from known tumour and villi populations, and a time dependent free platinum Pt (the active drug) infusion law being given, the mathematical model allows to compute the time evolution of both tumour and villi populations. The tumour population growth is based on Gompertz law and the Pt anti-tumour efficacy takes into account the circadian rhythm. Similarly the enterocyte population is subject to a circadian toxicity rhythm. The model has been derived using as far as possible experimental data. We examine two different optimisation problems. The eradication problem consists in finding the drug infusion law able to minimise the number of tumour cells while preserving a minimal level for the villi population. On the other hand, the containment problem searches for a quasi periodic treatment able to maintain the tumour population at the lowest possible level, while preserving the villi cells. The originality of these approaches is that the objective and constraint functions we use are LL^\infty criteria. We are able to derive their gradients with respect to the infusion rate and then to implement efficient optimisation algorithms

    Relevance of internal time and circadian robustness for cancer patients

    Get PDF
    International audienceAbstractBackgroundAdequate circadian timing of cancer treatment schedules (chronotherapy) can enhance tolerance and efficacy several-fold in experimental and clinical situations. However, the optimal timing varies according to sex, genetic background and lifestyle. Here, we compute the individual phase of the Circadian Timing System to decipher the internal timing of each patient and find the optimal treatment timing.MethodsTwenty-four patients (11 male; 13 female), aged 36 to 77 years, with advanced or metastatic gastro-intestinal cancer were recruited. Inner wrist surface Temperature, arm Activity and Position (TAP) were recorded every 10 min for 12 days, divided into three 4-day spans before, during and after a course of a set chronotherapy schedule. Pertinent indexes, I < O and a new biomarker, DI (degree of temporal internal order maintenance), were computed for each patient and period.ResultsThree circadian rhythms and the TAP rhythm grew less stable and more fragmented in response to treatment. Furthermore, large inter- and intra-individual changes were found for T, A, P and TAP patterns, with phase differences of up to 12 hours among patients. A moderate perturbation of temporal internal order was observed, but the administration of fixed chronomodulated chemotherapy partially resynchronized temperature and activity rhythms by the end of the study.ConclusionsThe integrated variable TAP, together with the asynchrony among rhythms revealed by the new biomarker DI, would help in the personalization of cancer chronotherapy, taking into account individual circadian phase markers

    A combined experimental and mathematical approach for molecular-based optimization of irinotecan circadian delivery

    Get PDF
    Circadian timing largely modifies efficacy and toxicity of many anticancer drugs. Recent findings suggest that optimal circadian delivery patterns depend on the patient genetic background. We present here a combined experimental and mathematical approach for the design of chronomodulated administration schedules tailored to the patient molecular profile. As a proof of concept we optimized exposure of Caco-2 colon cancer cells to irinotecan (CPT11), a cytotoxic drug approved for the treatment of colorectal cancer. CPT11 was bioactivated into SN38 and its efflux was mediated by ATP-Binding-Cassette (ABC) transporters in Caco-2 cells. After cell synchronization with a serum shock defining Circadian Time (CT) 0, circadian rhythms with a period of 26 h 50 (SD 63 min) were observed in the mRNA expression of clock genes REV-ERBα, PER2, BMAL1, the drug target topoisomerase 1 (TOP1), the activation enzyme carboxylesterase 2 (CES2), the deactivation enzyme UDP-glucuronosyltransferase 1, polypeptide A1 (UGT1A1), and efflux transporters ABCB1, ABCC1, ABCC2 and ABCG2. DNA-bound TOP1 protein amount in presence of CPT11, a marker of the drug PD, also displayed circadian variations. A mathematical model of CPT11 molecular pharmacokinetics-pharmacodynamics (PK-PD) was designed and fitted to experimental data. It predicted that CPT11 bioactivation was the main determinant of CPT11 PD circadian rhythm. We then adopted the therapeutics strategy of maximizing efficacy in non-synchronized cells, considered as cancer cells, under a constraint of maximum toxicity in synchronized cells, representing healthy ones. We considered exposure schemes in the form of an initial concentration of CPT11 given at a particular CT, over a duration ranging from 1 to 27 h. For any dose of CPT11, optimal exposure durations varied from 3h40 to 7h10. Optimal schemes started between CT2h10 and CT2h30, a time interval corresponding to 1h30 to 1h50 before the nadir of CPT11 bioactivation rhythm in healthy cells

    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

    Subjective sleep and overall survival in chemotherapy-naïve patients with metastatic colorectal cancer

    Get PDF
    Backround: Sleep disorders are prevalent in patients with advanced cancer. Their impact on clinical outcomes is not well understood. Methods: A post-hoc analysis was conducted in 361 chemo-naïve patients with metastatic colorectal cancer completing twice the EORTC QLQ-C30 questionnaire within a randomized international phase III trial. The study assessed the effect on overall survival (OS) of subjective sleep complaint, used as a normal or a time-dependent covariate (TDC), using a multivariate Cox proportional hazard model. Prognostic analysis was conducted on the whole study population and separately in each treatment arm (conventional FOLFOX2, or chronomodulated chronoFLO4). Results: Sleep problems were reported by 202 patients (56%) at baseline and by 188 (52%) on treatment. Sleep problems at baseline were independently associated with a higher risk of earlier death (HR: 1.36; p = 0.011), progression (HR: 1.43; p = 0.002) and poor treatment response (RR: 0.58; p = 0.016). TDC analysis confirmed the independent prognostic effect of sleep problems on OS (HR: 1.37; p = 0.008), while on treatment this effect was only observed using univariate analysis. The negative prognostic value of sleep problems on OS at baseline, on treatment, and as a TDC was greatest on chronoFLO4 compared to FOLFOX2. Conclusions: Subjective sleep problems are associated with poor clinical outcomes in metastatic colorectal cancer patients and affect chronotherapy effectiveness. There is a need for a well-tuned circadian timing system in order to increase chronotherapy activity. Prospective studies are needed for determining the impact of therapeutic approaches on sleep disorders upon quality of life and survival of cancer patients
    corecore