Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Comput Methods Programs Biomed ; 205: 106087, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33873075

RESUMO

INTRODUCTION: Medtronic has developed a virtual patient simulator for modeling and predicting insulin therapy outcomes for people with type 1 diabetes (T1D). An enhanced simulator was created to estimate outcomes when using the MiniMedTM 670G system with standard NovoLog® (EU: NovoRapid, US: NovoLog) versus Fiasp ® by using clinical data. METHODS: Sixty-seven participants' PK profiles were generated per type of insulin (Total of 134 PK profiles). 7,485 virtual patients' PK measurements was matched with one of the 67 NovoLog® PK Tmax values. These 7,485 virtual patients were then simulated using the Medtronic MiniMed™ 670G algorithm following an in-silico protocol of 90 days: 14 days in open loop (Manual Mode) followed by 76 days in closed loop (Auto Mode). Simulation study was repeated with each NovoLog® PK profile being replaced by its corresponding Fiasp® PK profile in the same virtual patient. To validate our in-silico analysis, we compared the results of "actual" 19 "real life" patients from a clinical study RESULTS: Simulated overall and postprandial glycemic outcomes improved in all age groups with Fiasp®. The percentage of time in the euglycemic range increased by about ~2.2% with Fiasp®, in all age groups (p < 0.01). The percentage of time spent at <70 mg/dL was reduced by about ~0.6% with insulin Fiasp® (p < 0.01) and the mean glucose was reduced by about 1.3 mg/dL (p < 0.01), excluding those age <7 years. The simulated mean postprandial SG was reduced by ~5 mg/dL, a significant difference for all age groups. A clinical study results showed similar improvements with MiniMedTM 670G system when switching from NovoLog® to Fiasp®. CONCLUSIONS: The simulation studies indicate that using Fiasp® in place of NovoLog® with the MiniMedTM 670G system will significantly improve the time spent in the healthy, euglycemic range and reduce exposure to hyperglycemia and hypoglycemia in most patients.


Assuntos
Diabetes Mellitus Tipo 1 , Insulina Aspart , Glicemia , Automonitorização da Glicemia , Criança , Diabetes Mellitus Tipo 1/tratamento farmacológico , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Insulina Aspart/uso terapêutico , Sistemas de Infusão de Insulina
2.
J Diabetes Sci Technol ; 11(2): 308-314, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28264192

RESUMO

Advances in insulin pump and continuous glucose monitoring technology have primarily focused on optimizing glycemic control for people with type 1 diabetes. There remains a need to identify ways to minimize the physical burden of this technology. A unified platform with closely positioned or colocalized interstitial fluid glucose sensing and hormone delivery components is a potential solution. Present challenges to combining these components are interference of glucose sensing from proximate insulin delivery and the large discrepancy between the life span of current insulin infusion sets and glucose sensors. Addressing these concerns is of importance given that the future physical burden of this technology is likely to be even greater with the ongoing development of the artificial pancreas, potentially incorporating multiple hormone delivery, glucose sensing redundancy, and sensing of other clinically relevant nonglucose biochemical inputs.


Assuntos
Diabetes Mellitus Tipo 1 , Pâncreas Artificial , Automonitorização da Glicemia/instrumentação , Automonitorização da Glicemia/tendências , Humanos , Sistemas de Infusão de Insulina/tendências , Pâncreas Artificial/tendências
3.
J Diabetes Sci Technol ; 10(3): 708-13, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26880389

RESUMO

BACKGROUND: Efficacy and safety of the Medtronic Hybrid Closed-Loop (HCL) system were tested in subjects with type 1 diabetes in a supervised outpatient setting. METHODS: The HCL system is a prototype research platform that includes a sensor-augmented insulin pump in communication with a control algorithm housed on an Android-based cellular device. Nine subjects with type 1 diabetes (5 female, mean age 53.3 years, mean A1C 7.2%) underwent 9 studies totaling 571 hours of closed-loop control using either default or personalized parameters. The system required meal announcements with estimates of carbohydrate (CHO) intake that were based on metabolic kitchen quantification (MK), dietician estimates (D), or subject estimates (Control). Postprandial glycemia was compared for MK, D, and Control meals. RESULTS: The overall sensor glucose mean was 145 ± 43, the overall percentage time in the range 70-180 mg/dL was 80%, the overall percentage time <70 mg/dL was 0.79%. Compared to intervals of default parameter use (225 hours), intervals of personalized parameter use (346 hours), sensor glucose mean was 158 ± 49 and 137 ± 37 mg/dL (P < .001), respectively, and included more time in range (87% vs 68%) and less time below range (0.54% vs 1.18%). Most subjects underestimated the CHO content of meals, but postprandial glycemia was not significantly different between MK and matched Control meals (P = .16) or between D and matched Control meals (P = .76). There were no episodes of severe hypoglycemia. CONCLUSIONS: The HCL system was efficacious and safe during this study. Personally adapted HCL parameters were associated with more time in range and less time below range than default parameters. Accurate estimates of meal CHO did not contribute to improved postprandial glycemia.


Assuntos
Automonitorização da Glicemia/instrumentação , Glicemia/análise , Diabetes Mellitus Tipo 1/tratamento farmacológico , Sistemas de Infusão de Insulina , Pâncreas Artificial , Adulto , Algoritmos , Telefone Celular , Diabetes Mellitus Tipo 1/sangue , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Pacientes Ambulatoriais , Telemedicina
4.
J Diabetes Sci Technol ; 7(2): 381-8, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23566996

RESUMO

BACKGROUND: Subcutaneously infused insulin may interfere with the function of nearby glucose-sensing electrodes and vice versa. The prototype of the Combo-Set device (Medtronic) incorporates a subcutaneous insulin delivery catheter and continuous glucose monitoring (CGM) sensor assembled on the same platform and separated by 11 mm. We aim to evaluate Combo-Set's insulin delivery and glucose-sensing functions. METHODS: Ten subjects with type 1 diabetes wore a Combo-Set and a Sof-Sensor inserted subcutaneously in contralateral abdominal areas connected to iPro recorders (Medtronic) for 53.25 ± 0.75 h (mean ± standard deviation). The Combo-Set delivered insulin diluent except during meal tests on days 1 and 3 when insulin lispro was delivered as a meal bolus and postmeal basal. Venous plasma samples were collected at the following time points from meal start: 0, 30, 60, 120, and 180 min for insulin measurements. The accuracy of the Combo-Set sensors was evaluated and compared with that of the Sof-Sensor, with each referenced against capillary glucose values (Contour Link Meter, Bayer). RESULTS: Accuracy of the Combo-Set sensor was comparable to that of the Sof-Sensor. Clarke error grid analysis showed that 97% of Combo-Set and 93% of Sof-Sensor values were in the A+B regions (p = .20, not significant). The Combo-Set showed the expected postbolus peak insulin time (67 ± 9 min, mean ± standard error). One "no delivery" alarm occurred during the 21 patient days of use. CONCLUSION: A device providing for simultaneous adjacent placement of an insulin infusion catheter and a CGM sensor is feasible and functions within acceptable limits. The low "no delivery" alarm rate was similar to that of other infusion sets.


Assuntos
Automonitorização da Glicemia/instrumentação , Glicemia/análise , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/tratamento farmacológico , Sistemas de Infusão de Insulina , Adulto , Automonitorização da Glicemia/métodos , Estudos de Viabilidade , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Insulina Lispro/administração & dosagem , Masculino , Pessoa de Meia-Idade
5.
J Diabetes Sci Technol ; 7(2): 465-77, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23567006

RESUMO

BACKGROUND: In insulin pump therapy, optimization of bolus and basal insulin dose settings is a challenge. We introduce a new algorithm that provides individualized basal rates and new carbohydrate ratio and correction factor recommendations. The algorithm utilizes a mathematical model of blood glucose (BG) as a function of carbohydrate intake and delivered insulin, which includes individualized parameters derived from sensor BG and insulin delivery data downloaded from a patient's pump. METHODS: A mathematical model of BG as a function of carbohydrate intake and delivered insulin was developed. The model includes fixed parameters and several individualized parameters derived from the subject's BG measurements and pump data. Performance of the new algorithm was assessed using n = 4 diabetic canine experiments over a 32 h duration. In addition, 10 in silico adults from the University of Virginia/Padova type 1 diabetes mellitus metabolic simulator were tested. RESULTS: The percentage of time in glucose range 80-180 mg/dl was 86%, 85%, 61%, and 30% using model-based therapy and [78%, 100%] (brackets denote multiple experiments conducted under the same therapy and animal model), [75%, 67%], 47%, and 86% for the control experiments for dogs 1 to 4, respectively. The BG measurements obtained in the simulation using our individualized algorithm were in 61-231 mg/dl min-max envelope, whereas use of the simulator's default treatment resulted in BG measurements 90-210 mg/dl min-max envelope. CONCLUSIONS: The study results demonstrate the potential of this method, which could serve as a platform for improving, facilitating, and standardizing insulin pump therapy based on a single download of data.


Assuntos
Algoritmos , Técnicas Biossensoriais , Sistemas de Infusão de Insulina , Insulina/administração & dosagem , Modelos Teóricos , Animais , Técnicas Biossensoriais/instrumentação , Técnicas Biossensoriais/métodos , Glicemia/análise , Automonitorização da Glicemia/instrumentação , Automonitorização da Glicemia/métodos , Simulação por Computador , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/veterinária , Doenças do Cão/sangue , Cães , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacocinética , Insulina/farmacocinética
6.
J Diabetes Sci Technol ; 6(5): 1123-30, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23063039

RESUMO

BACKGROUND: Closed-loop (CL) insulin delivery systems utilizing proportional-integral-derivative (PID) controllers have demonstrated susceptibility to late postprandial hypoglycemia because of delays between insulin delivery and blood glucose (BG) response. An insulin feedback (IFB) modification to the PID algorithm has been introduced to mitigate this risk. We examined the effect of IFB on CL BG control. METHODS: Using the Medtronic ePID CL system, four subjects were studied for 24 h on PID control and 24 h during a separate admission with the IFB modification (PID + IFB). Target glucose was 120 mg/dl; meals were served at 8:00 AM, 1:00 PM, and 6:00 PM and were identical for both admissions. No premeal manual boluses were given. Reference BG excursions, defined as incremental glucose rise from premeal to peak, and postprandial BG area under the curve (AUC; 0-5 h) were compared. Results are reported as mean ± standard deviation. RESULTS: The PID + IFB control resulted in higher mean BG levels compared with PID alone (153 ± 54 versus 133 ± 56 mg/dl; p < .0001). Postmeal BG excursions (114 ± 28 versus 114 ± 47 mg/dl) and AUCs (285 ± 102 versus 255 ± 129 mg/dl/h) were similar under both conditions. Total insulin delivery averaged 57 ± 20 U with PID versus 45 ± 13 U with PID + IFB (p = .18). Notably, eight hypoglycemic events (BG < 60 mg/dl) occurred during PID control versus none during PID + IFB. CONCLUSIONS: Addition of IFB to the PID controller markedly reduced the occurrence of hypoglycemia without increasing meal-related glucose excursions. Higher average BG levels may be attributable to differences in the determination of system gain (Kp) in this study. The prevention of postprandial hypoglycemia suggests that the PID + IFB algorithm may allow for lower target glucose selection and improved overall glycemic control.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Retroalimentação Fisiológica/efeitos dos fármacos , Sistemas de Infusão de Insulina , Insulina/administração & dosagem , Insulina/farmacologia , Administração Metronômica , Adolescente , Adulto , Algoritmos , Glicemia/análise , Glicemia/efeitos dos fármacos , Automonitorização da Glicemia/instrumentação , Estudos Cross-Over , Diabetes Mellitus Tipo 1/sangue , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacologia , Masculino , Adulto Jovem
7.
Diabetes Care ; 35(10): 1994-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22815298

RESUMO

OBJECTIVE: Even under closed-loop (CL) conditions, meal-related blood glucose (BG) excursions frequently exceed target levels as a result of delays in absorption of insulin from the subcutaneous site of infusion. We hypothesized that delaying gastric emptying with preprandial injections of pramlintide would improve postprandial glycemia by allowing a better match between carbohydrate and insulin absorptions. RESEARCH DESIGN AND METHODS: Eight subjects (4 female; age, 15-28 years; A1C, 7.5 ± 0.7%) were studied for 48 h on a CL insulin-delivery system with a proportional integral derivative algorithm with insulin feedback: 24 h on CL control alone (CL) and 24 h on CL control plus 30-µg premeal injections of pramlintide (CLP). Target glucose was set at 120 mg/dL; timing and contents of meals were identical on both study days. No premeal manual boluses were given. Differences in reference BG excursions, defined as the incremental glucose rise from premeal to peak, were compared between conditions for each meal. RESULTS: CLP was associated with overall delayed time to peak BG (2.5 ± 0.9 vs. 1.5 ± 0.5 h; P < 0.0001) and reduced magnitude of glycemic excursion (88 ± 42 vs. 113 ± 32 mg/dL; P = 0.006) compared with CL alone. Pramlintide effects on glycemic excursions were particularly evident at lunch and dinner, in association with higher premeal insulin concentrations at those mealtimes. CONCLUSIONS: Pramlintide delayed the time to peak postprandial BG and reduced the magnitude of prandial BG excursions. Beneficial effects of pramlintide on CL may in part be related to higher premeal insulin levels at lunch and dinner compared with breakfast.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 1/sangue , Hipoglicemiantes/uso terapêutico , Polipeptídeo Amiloide das Ilhotas Pancreáticas/uso terapêutico , Adolescente , Adulto , Diabetes Mellitus Tipo 1/tratamento farmacológico , Feminino , Humanos , Insulina/sangue , Masculino , Refeições , Pâncreas Artificial , Período Pós-Prandial
8.
J Clin Endocrinol Metab ; 96(5): 1402-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21367930

RESUMO

CONTEXT: Initial studies of closed-loop proportional integral derivative control in individuals with type 1 diabetes showed good overnight performance, but with breakfast meal being the hardest to control and requiring supplemental carbohydrate to prevent hypoglycemia. OBJECTIVE: The aim of this study was to assess the ability of insulin feedback to improve the breakfast-meal profile. DESIGN AND SETTING: We performed a single center study with closed-loop control over approximately 30 h at an inpatient clinical research facility. PATIENTS: Eight adult subjects with previously diagnosed type 1 diabetes participated. INTERVENTION: Subjects received closed-loop insulin delivery with supplemental carbohydrate as needed. MAIN OUTCOME MEASURES: Outcome measures were plasma insulin concentration, model-predicted plasma insulin concentration, 2-h postprandial and 3- to 4-h glucose rate-of-change following breakfast after 1 d of closed-loop control, and the need for supplemental carbohydrate in response to nadir hypoglycemia. RESULTS: Plasma insulin levels during closed loop were well correlated with model predictions (R = 0.86). Fasting glucose after 1 d of closed loop was not different from nighttime target (118 ± 9 vs. 110 mg/dl; P = 0.38). Two-hour postbreakfast glucose was 132 ± 16 mg/dl with stable values 3-4 h after the meal (0.03792 ± 0.0884 mg/dl · min, not different from 0; P = 0.68) and at target (97 ± 6 mg/dl, not different from 90; P = 0.28). Three subjects required supplemental carbohydrates after breakfast on d 2 of closed loop. CONCLUSIONS/INTERPRETATION: Insulin feedback can be implemented using a model estimate of concentration. Proportional integral derivative control with insulin feedback can achieve a desired breakfast response but still requires supplemental carbohydrate to be delivered in some instances. Studies assessing more optimal control configurations and safeguards need to be conducted.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Retroalimentação Fisiológica/fisiologia , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Adulto , Algoritmos , Técnicas Biossensoriais , Calibragem , Carboidratos da Dieta/uso terapêutico , Feminino , Humanos , Hipoglicemia/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Período Pós-Prandial/fisiologia , Resultado do Tratamento , Adulto Jovem
9.
J Diabetes Sci Technol ; 5(6): 1342-51, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22226251

RESUMO

BACKGROUND: We have previously used insulin feedback (IFB) as a component of a closed-loop algorithm emulating the ß cell. This was based on the observation that insulin secretion is inhibited by insulin concentration. We show here that the effect of IFB is to make a closed-loop system behave as if delays in the insulin pharmacokinetic (PK)/pharmacodynamic (PD) response are reduced. We examine whether the mechanism can be used to compensate for delays in the subcutaneous PK/PD insulin response. METHOD: Closed-loop insulin delivery was performed in seven diabetic dogs using a proportional-integral-derivative model of the ß cell modified by model-predicted IFB. The level of IFB was set using pole placement. Meal responses were obtained on three occasions: without IFB (NONE), reference IFB (REF), and 2xREF, with experiments performed in random order. The ability of the insulin model to predict insulin concentration was evaluated by correlation with the measured profile and results reported as R(2). The ability of IFB to improve the meal response was evaluated by comparing peak and nadir postprandial glucose and area under the curve (AUC; repeated measures analysis of variance with post hoc test for linear trend). RESULTS: Insulin concentration was well predicted by the model (median R(2) = 0.87, 0.79, and 0.90 for NONE, REF, and 2xREF, respectively). Peak postprandial glucose (294 ± 15, 243 ± 21, and 247 ± 16 mg/dl) and AUC (518.2 ± 36.13, 353.5 ± 45.04, and 280.3 ± 39.37 mg/dl · min) decreased with increasing IFB (p < .05, linear trend). Nadir glucose was not affected by IFB (76 ± 5.4, 68 ± 7.3, and 72 ± 4.3 mg/dl; p = .63). CONCLUSIONS: Insulin feedback provides an effective mechanism to compensate for delay in the insulin PK/PD profile.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Hipoglicemiantes/farmacocinética , Insulina/farmacocinética , Pâncreas Artificial , Algoritmos , Animais , Área Sob a Curva , Glicemia/análise , Diabetes Mellitus Tipo 1/sangue , Cães , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/sangue , Insulina/administração & dosagem , Insulina/sangue , Sistemas de Infusão de Insulina
10.
Diabetes Care ; 32(2): 240-4, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19017777

RESUMO

OBJECTIVE: The purpose of this study was to examine the effect of type of insulin analog and age of insertion site on the pharmacodynamic characteristics of a standard insulin bolus in youth with type 1 diabetes receiving insulin pump therapy. RESEARCH DESIGN AND METHODS: Seventeen insulin pump-treated adolescents with type 1 diabetes underwent two euglycemic clamp procedures after a 0.2 unit/kg bolus of either insulin aspart or lispro on day 1 and day 4 of insulin pump site insertion. The glucose infusion rate (GIR) required to maintain euglycemia was the primary pharmacodynamic measure. RESULTS: There were no statistically significant differences in any of the pharmacodynamic parameters between aspart and lispro during day 1 and day 4. However, when the two groups were combined, time to discontinuation of exogenous glucose infusion, and time to half-maximal onset and offset of insulin action were observed significantly earlier during day 4 compared with day 1 (P = 0.03-0.0004), but the overall area under the GIR curve was similar on day 1 and day 4. CONCLUSIONS: With both insulin aspart and lispro, there is an earlier peak and shorter duration of action with increasing duration of infusion site use, but overall insulin action is not affected.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Insulina/análogos & derivados , Adolescente , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Criança , Desenho de Equipamento , Feminino , Técnica Clamp de Glucose , Hemoglobinas Glicadas/metabolismo , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/uso terapêutico , Insulina/administração & dosagem , Insulina/uso terapêutico , Insulina Aspart , Sistemas de Infusão de Insulina , Insulina Lispro , Cinética , Masculino , Seleção de Pacientes
11.
J Diabetes Sci Technol ; 3(5): 1047-57, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20144418

RESUMO

BACKGROUND: Algorithms for closed-loop insulin delivery can be designed and tuned empirically; however, a metabolic model that is predictive of clinical study results can potentially accelerate the process. METHODS: Using data from a previously conducted closed-loop insulin delivery study, existing models of meal carbohydrate appearance, insulin pharmacokinetics, and the effect on glucose metabolism were identified for each of the 10 subjects studied. Insulin's effects to increase glucose uptake and decrease endogenous glucose production were described by the Bergman minimal model, and compartmental models were used to describe the pharmacokinetics of subcutaneous insulin absorption and glucose appearance following meals. The composite model, comprised of only five equations and eight parameters, was identified with and without intraday variance in insulin sensitivity (S(I)), glucose effectiveness at zero insulin (GEZI), and endogenous glucose production (EGP) at zero insulin. RESULTS: Substantial intraday variation in SI, GEZI and EGP was observed in 7 of 10 subjects (root mean square error in model fit greater than 25 mg/dl with fixed parameters and nadir and/or peak glucose levels differing more than 25 mg/dl from model predictions). With intraday variation in these three parameters, plasma glucose and insulin were well fit by the model (R(2) = 0.933 +/- 0.00971 [mean +/- standard error of the mean] ranging from 0.879-0.974 for glucose; R(2) = 0.879 +/- 0.0151, range 0.819-0.972 for insulin). Once subject parameters were identified, the original study could be reconstructed using only the initial glucose value and basal insulin rate at the time closed loop was initiated together with meal carbohydrate information (glucose, R(2) = 0.900 +/- 0.015; insulin delivery, R(2) = 0.640 +/- 0.034; and insulin concentration, R(2) = 0.717 +/- 0.041). CONCLUSION: Metabolic models used in developing and comparing closed-loop insulin delivery algorithms will need to explicitly describe intraday variation in metabolic parameters, but the model itself need not be comprised by a large number of compartments or differential equations.


Assuntos
Automonitorização da Glicemia , Glicemia/efeitos dos fármacos , Ritmo Circadiano , Diabetes Mellitus Tipo 1/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Sistemas de Infusão de Insulina , Insulina/administração & dosagem , Pâncreas Artificial , Adulto , Algoritmos , Simulação por Computador , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Carboidratos da Dieta/administração & dosagem , Carboidratos da Dieta/metabolismo , Feminino , Humanos , Hipoglicemiantes/farmacocinética , Insulina/farmacocinética , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Modelos Estatísticos , Valor Preditivo dos Testes , Resultado do Tratamento
12.
J Diabetes Sci Technol ; 3(5): 1207-14, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20144438

RESUMO

Through the use of enzymatic sensors-inserted subcutaneously in the abdomen or ex vivo by means of microdialysis fluid extraction-real-time minimally invasive continuous glucose monitoring (CGM) devices estimate blood glucose by measuring a patient's interstitial fluid (ISF) glucose concentration. Signals acquired from the interstitial space are subsequently calibrated with capillary blood glucose samples, a method that has raised certain questions regarding the effects of physiological time lags and of the duration of processing delays built into these devices. The time delay between a blood glucose reading and the value displayed by a continuous glucose monitor consists of the sum of the time lag between ISF and plasma glucose, in addition to the inherent electrochemical sensor delay due to the reaction process and any front-end signal processing delays required to produce smooth traces. Presented is a review of commercially available, minimally invasive continuous glucose monitors with manufacturer reported device delays. The data acquisition process for the Medtronic MiniMed (Northridge, CA) continuous glucose monitoring system-CGMS Gold-and the Guardian RT monitor is described with associated delays incurred for each processing step. Filter responses for each algorithm are examined using in vitro hypoglycemic and hyperglycemic clamps, as well as with an analysis of fast glucose excursions from a typical meal response. Results demonstrate that the digital filters used by each algorithm do not cause adverse effects to fast physiologic glucose excursions, although nonphysiologic signal characteristics can produce greater delays.


Assuntos
Automonitorização da Glicemia/instrumentação , Glicemia/metabolismo , Equipamentos para Diagnóstico , Algoritmos , Artefatos , Diagnóstico por Computador , Carboidratos da Dieta/administração & dosagem , Carboidratos da Dieta/metabolismo , Técnicas Eletroquímicas/instrumentação , Desenho de Equipamento , Líquido Extracelular/metabolismo , Humanos , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Processamento de Sinais Assistido por Computador , Fatores de Tempo
14.
J Diabetes Sci Technol ; 1(5): 639-44, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19885132

RESUMO

BACKGROUND: Since the advent of subcutaneous glucose sensors, there has been intense focus on characterizing the delay in the interstitial fluid (ISF) glucose response and the effect of insulin to alter the plasma-to-ISF glucose gradient. The Medtronic MiniMed continuous glucose monitoring system (CGMS) has often been used for this purpose; however, many of the studies have used experimental conditions that fall outside its intended use, for example, studies that have assessed the delay during rapid glucose excursions brought about by intravenous infusion of glucose or insulin. Under these conditions, it is possible that the rate of glucose change may exceed that allowed by CGMS filtering routines. If so, the estimated delay may be because of the filter rather than the ISF. Also, sensor characteristics, such as nonspecific offset current or stability, may have been inadvertently attributed to changes in the plasma-to-ISF gradient. The potential for these issues to have confounded the understanding of ISF glucose delay and gradient is investigated. METHODS: An in vitro preparation in which no delay or gradient exists between sensor and measurement solution was used to recreate a rapidly changing glucose profile from a previously published in vivo study. The CGMS system (N = 6 sensors) was then used to estimate any artifactual delay and gradient introduced by the system per se. RESULTS: One-point calibration resulted in an apparent change in gradient as glucose was lowered from approximately 100 to 50 mg/dl. After a two-point calibration, sensor glucose followed the glucose profile as it was decreased slowly from approximately 100 to approximately 60 mg/dl; however, when the glucose level was subsequently increased rapidly to approximately 150 mg/dl, CGMS filtering routines limited the rate of change of sensor glucose and introduced a delay similar to that previously attributed to ISF glucose equilibration delay. CONCLUSIONS: Studies that have previously used the Medtronic MiniMed CGMS system to assess changes in the plasma-to-ISF glucose gradient may need to be reassessed to ensure that the offset current was estimated accurately. Studies that have used the system to assess ISF glucose delay during rapid, unphysiologic changes in glucose and did not remove the CGMS smoothing filters may have attributed CGMS filter delay to ISF glucose equilibration.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...