Curso Insulin Therapy

download Curso Insulin Therapy

of 31

Transcript of Curso Insulin Therapy

  • 8/13/2019 Curso Insulin Therapy

    1/31

    Insulin therapy in type 2 diabetes

    Trent Davis, MD, Steven V. Edelman, MD*

    Section of Diabetes/Metabolism, Veterans Affairs San Diego HealthCare System,

    3350 La Jolla Village Drive 111G, San Diego, CA 92161, USA

    Diabetes mellitus affects approximately 18 million people in the United

    States, which is approximately 6% of the overall population, and over

    800,000 new cases are diagnosed annually [1]. Diabetes may actually be

    more endemic than these figures indicate because there are no symptoms in

    the early stages of the disease, and potentially one undiagnosed individual

    exists for every one that is identified [2]. Of the total diabetic population,

    85% to 90% of individuals have type 2 diabetes whereas 10% to 15% have

    type 1 diabetes[3].

    Type 2 diabetes leads to a tremendous amount of death and disabilityand uses a large portion of the health care dollar [4]. Although diabetes is

    associated with multiple disorders with distinct pathologic mechanisms,

    insulin resistance is the common denominator and is associated with several

    comorbidities, including obesity, hypertension, and vascular disease. The

    natural history of the disease is often complicated by various microvascular

    and macrovascular sequelae that can lead to blindness, end-stage renal

    disease, lower-extremity amputation, and atherosclerosis resulting in heart

    attack or stroke [3,5]. Although most of the human suffering is caused by

    end-stage microvascular disease, 80% of diabetics die of macrovascularcardiovascular disease. There is now clear evidence from the United

    Kingdom Prospective Diabetes Study (UKPDS) and the Kumamoto study

    that improved glycemic control through intensive diabetes management

    delays the onset and significantly retards the progression of microvascular

    complications in patients with type 2 diabetes mellitus[6,7]. The results from

    the UKPDS are reassuring in that, although intensive treatment with insulin

    was associated with increased weight gain and hypoglycemia, there is no

    evidence of any harmful effect of insulin on cardiovascular outcomes, which

    has been a controversial issue. An epidemiologic analysis of the UKPDS

    * Corresponding author.

    E-mail address:[email protected](S.V. Edelman).

    0025-7125/04/$ - see front matter 2004 Elsevier Inc. All rights reserved.

    doi:10.1016/j.mcna.2004.04.005

    Med Clin N Am 88 (2004) 865895

    mailto:[email protected]:[email protected]
  • 8/13/2019 Curso Insulin Therapy

    2/31

    data shows a continuous association between the risk of cardiovascular

    complications and glycemia, such that for every percentage point of decrease

    in HbA1c(eg, from 9% to 8%), there is a 25% reduction in diabetes-relateddeaths, a 7% reduction in all-cause mortality, and an 18% reduction in

    combined fatal and nonfatal myocardial infarction [6].

    To achieve glycemic goals in patients with type 2 diabetes, we now have

    multiple pharmacologic agents with different mechanisms of action, in-

    cluding sulfonylureas, meglitinides, metformin, a-glucosidase inhibitors,

    thiazolidinediones, and insulin. It must be emphasized, however, that unlike

    patients with type 1 diabetes, who have no significant insulin secretion and

    hence require insulin therapy from the onset of their disease, a prominent

    feature in the early stages of the disease for patients with type 2 diabetesis insulin resistance with hyperinsulinemia. Therefore, improving insulin

    sensitivity be means of caloric restriction, exercise, and weight management

    early in the disease process will benefit type 2 diabetics. When these measures

    fail, glycemic goals can often be achieved with oral agents used alone or

    in combination with each other. When patients are diagnosed late in the

    natural history, however, there is progressive loss of pancreatic beta-cell

    function and endogenous insulin secretion, making diurnal glycemic control

    difficult. At this late stage, most patients require exogenous insulin therapy

    to achieve optimal glucose control. The American Diabetes Association(ADA) now recommends that the glycemic objective for patients with type 2

    diabetes to normalize glycemia and glycosylated hemoglobin concentrations

    should be similar to that for type 1 diabetes.

    Pathogenesis and natural history of type 2 diabetes

    Of the Americans diagnosed with type 2 diabetes, 80% to 90% are obese,

    and the remainder are lean [8]. The genesis of hyperglycemia in type 2diabetes involves a triad of abnormalities: excessive hepatic glucose pro-

    duction, impaired pancreatic insulin secretion, and peripheral resistance

    to insulin action, occurring principally in liver and muscle tissue [9]. The

    severity of these abnormalities and their contribution to the degree of

    hyperglycemia can vary considerably, causing heterogeneity in the metabolic

    expression of the diabetic state. Such differences are best exemplified by the

    lean and obese varieties of type 2 diabetes, which have the same underlying

    pathophysiologic basis but differ in the extent to which each abnormality

    contributes to the development of the hyperglycemic state. Of these ab-normalities, peripheral insulin resistance to insulin action and impaired

    pancreatic beta-cell secretion are early and primary abnormalities, whereas

    increased hepatic glucose production is a late and secondary manifestation.

    Early in their disease, patients with type 2 diabetes compensate for increased

    insulin resistance at the tissue level by increasing pancreatic beta-cell insulin

    secretion[10]. When this compensation is no longer adequate to overcome

    866 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    3/31

    the insulin resistance, blood glucose levels begin to rise. Over the course

    of the disease, endogenous insulin levels slowly begin to decrease and,

    ultimately, many patients with type 2 diabetes are unable to achieve optimalglycemic control with oral agents [11].

    In subjects with type 2 diabetes who are lean, impaired insulin secretion is

    the predominant defect, and insulin resistance tends to be less severe than in

    the obese variety [12]. On the other hand, insulin resistance and hyper-

    insulinemia are the classical abnormalities of obese persons with type

    2 diabetes [12]. In type 2 diabetes, insulin secretion is often excessive

    compared with the nondiabetic situation but is still insufficient to overcome

    the insulin resistance that is present. It is important to understand and

    appreciate these fundamental differences when considering insulin therapyin type 2 diabetes. Based on this knowledge, lean type 2 diabetic subjects

    usually fail oral agents faster and will require considerably less insulin to

    control their hyperglycemia than their obese counterparts. In contrast, large

    doses of exogenous insulin are the rule in the obese form of this disorder

    when euglycemia is desired[13].

    The need for large amounts of exogenous insulin in obese type 2 diabetes

    also raises the question of the most appropriate methods of insulin delivery.

    Under normal circumstances, insulin is secreted from the pancreas into the

    portal vein, going directly to the liver in which a large first-pass extractionof portal insulin occurs [14]. When insulin is injected subcutaneously,

    absorption occurs directly into the peripheral circulation, without the initial

    effects of hepatic extraction. Therefore, the tissues are exposed to greater

    levels of insulin than if insulin was provided by the portal route. Because the

    primary target of exogenous insulin is the liver, type 2 diabetes may be

    uniquely suited to delivery of insulin through the portal vein. Such a

    situation occurs when insulin is delivered intraperitoneally, and the majority

    of insulin is absorbed into the portal circulation[15].Intraperitoneal insulin

    delivery systems will not be discussed in this section, however, this methodholds considerable promise in type 2 diabetes because of the more

    physiologic delivery of insulin and because of selective and effective

    inhibition of hepatic glucose output, with less peripheral insulinemia than

    occurs with subcutaneous insulin injections[16].

    Intensive insulin therapy

    Successful insulin management requires an educated and motivatedpatient, as well as the participation of a multidisciplinary health care team.

    Intensive insulin therapy requires a substantial input of physician and

    support staff time, which has a significant economic impact on the health

    care system [17]. Although long-term data on costs are not yet available,

    projections suggest that substantial savings from the high costs of end-

    stage disease could be achieved by following ADA guidelines [18].

    867T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    4/31

    Furthermore, as discussed later, we now have the opportunity to use

    combinations of insulin and a variety of oral antidiabetic agents with

    differing mechanisms of action. The use of these potent combinationspermit us to safely and effectively lower blood glucose levels and to

    achieve ADA target glycemic levels with relatively low risk for hypogly-

    cemia or weight gain.

    In addition to the natural history of type 2 diabetes, there is heterogeneity

    in the pathophysiology of type 2 diabetes mellitus that may influence when

    patients require insulin. Some patients who have been diagnosed with type 2

    diabetes may actually have a condition more closely related to insulin-

    dependent or type 1 diabetes, with severe insulinopenia. Many of these pa-

    tients have been shown to have islet cell antibody positivity or antibodies toglutamic acid decarboxylase, with a decreased C-peptide response to

    glucagon stimulation and a propensity for primary oral medication failure

    [19]. These individuals are now labeled with the condition latent autoimmune

    diabetes in adults [20]. There are also wide geographic and racial differences

    that may influence the need for insulin therapy. For example, Asian patients

    with type 2 diabetes tend to be thinner, are diagnosed with diabetes at an

    earlier age, fail oral hypoglycemic agents much sooner, and are more

    sensitive to insulin therapy than the classic centrally obese patient in the

    United States and some parts of Europe[21].The goals of therapy should be tailored to individual patients. Candidates

    for intensive management should be motivated, compliant, and educable,

    and be without other medical conditions and physical limitations that

    preclude accurate and reliable home glucose monitoring (HGM) and insulin

    administration; caution is advised in patients who are aged or have hypo-

    glycemic unawareness. Other limitations to achieving normoglycemia

    may include high titers of insulin antibodies, especially in patients with a

    history of intermittent use of insulin of animal origin. The site of insulin

    injection also may change the pharmacokinetics, and absorption can behighly variable, especially if lipohypertrophy is present. The periumbilical

    area has been shown to be one of the most desirable areas to inject insulin

    because of the rapid and consistent absorption kinetics observed at this

    location; however, rotating the injection site is usually advised[22]. It is also

    advisable to inject in the same body location for a certain meal time (ie,

    triceps fat pad for breakfast, abdomen for lunch, and upper thighs for

    dinner)[23].

    In summary, before starting insulin therapy, the patient should be well

    educated in the techniques of HGM, proper insulin administration, and self-adjustment of the insulin dose, if appropriate, as well as knowledgeable

    about dietary and exercise strategies, including carbohydrate counting. The

    patient and family members also need to be informed about hypoglycemia

    prevention, recognition, and treatment. Initial and ongoing education by a

    diabetes management team, including a certified diabetes educator, is crucial

    for long-term success and safety.

    868 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    5/31

    Insulin treatment strategies

    Combination therapy

    Combination therapy usually refers to the use of daytime oral antidia-

    betic agents together with a single injection of intermediate or long-acting

    insulin at bedtime. Several studies [2436] have looked at the safety and

    efficacy of combination therapy. For many of the reasons mentioned earlier,

    the analysis of studies to evaluate the efficacy and safety of combination

    therapy is difficult. Several review articles using meta-analysis conclude that

    combination therapy results in only modest improvements in glucose con-

    trol and contribute to increased medical costs of diabetes management

    compared with insulin therapy alone. These earlier studies, however, wereconducted when sulfonylureas were the only available type of oral agent.

    Because of heterogeneity in type 2 diabetes together with variability in the

    design and clinical situations of previous studies, however, the use of meta-

    analysis may be inappropriate for making generalized statements regarding

    this form of therapy [27,37]. Based on several recent reports, the use of

    combination therapy has been quite successful in selected patients, especially

    with the newer oral agents used alone or together with insulin [26

    29,35,36,3841].

    For a number of practical reasons, combination therapy may bebeneficial. The patient does not need to learn how to mix different types

    of insulin, and patient compliance and acceptance are better with a single

    injection than with multiple injections of insulin. Combination therapy also

    requires a lower total dose of exogenous insulin than regimens of two or

    three injections per day. Combination therapy also contributes to less

    weight gain and peripheral hyperinsulinemia. Last, combination therapy is

    ideally suited to suppress excessive hepatic glucose production overnight.

    The rationale for combination therapy with insulin and sulfonylureas is

    based on the assumption that, if evening insulin lowers the fasting glucoseconcentration to normal, then daytime oral agents will be more effective

    in controlling postprandial hyperglycemia and maintaining euglycemia

    throughout the day. Metabolic profiles of patients who have type 2 diabetes

    have demonstrated that fasting blood glucose contributes significantly to

    daytime hyperglycemia[42]. In addition, the fasting blood glucose concen-

    tration is highly correlated with the degree of hepatic glucose production

    during the early morning hours [13]. Hepatic glucose output is directly

    decreased by insulin[43]and indirectly inhibited by the ability of insulin to

    reduce adipose tissue lipolysis, with lower concentrations of free fatty acidsand gluconeogenesis [41]. Also, the peak of bedtime intermediate-acting

    insulin coincides with the onset of the dawn phenomenon (early morning

    resistance to insulin caused by diurnal variations in growth hormone

    and possibly in levels of norepinephrine), which usually occurs between

    3 and 7 AM. Bedtime insulin also increases the morning serum insulin

    869T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    6/31

    concentration and may assist in reducing the post-breakfast glucose rise in

    addition to the fasting value.

    Combination of insulin and sulfonylurea agents

    In one of the first large studies demonstrating the efficacy of insulin/

    sulfonylurea combination therapy, Yki-Jarvinen et al [38] compared com-

    bination therapy with regimens of two and four insulin injections per day

    in patients with type 2 diabetes. These patients were on submaximal doses

    of glyburide (12.5 mg/d), glipizide (20 mg/d), and metformin (1.4 g/d),

    with fasting blood glucose concentrations at approximately 225 mg/dL and

    mean fasting serum C-peptide values of 0.66 nmol/L. After 3 months, alltreatment groups had similar reductions in mean diurnal glucose con-

    centrations and glycosylated hemoglobin levels (1.6%1.9%) compared

    with the control group, who were taking oral agents alone. The group

    treated with a combination of oral agents and bedtime neutral protamine

    Hagedorn (NPH) insulin, however, had the least weight gain (1.2 0.5 kg)

    of any group and a 50% to 65% lower increment in mean diurnal serum-free

    insulin concentrations. There was no evidence of severe hypoglycemia with

    combination therapy, and patient acceptance was excellent.

    Several other recent publications [26,28,29,31,34,40] also support theadditional efficacy and safety of combination therapy in patients who are

    inadequately controlled by oral hypoglycemic agents alone. A recent study

    conducted by Riddle and Schneider[36]demonstrates the efficacy and safety

    of a combination consisting of 70% NPH insulin and 30% regular insulin

    (70/30) insulin at dinnertime and sulfonylurea therapy. In this study, 145

    type 2 diabetics with uncontrolled hyperglycemia (fasting plasma glucose

    level [FPG] 180300 mg/dL), on maximum sulfonylurea therapy (glimepir-

    ide, 8 mg orally, twice daily) were randomized to placebo plus insulin or

    glimepiride plus insulin for 6 months. The dose of 70/30 insulin atdinnertime was titrated to keep fasting fingerstick capillary blood glucose

    to less than 120 mg/dL. At 24 weeks, HbA1c levels decreased significantly

    and similarly in both groups (9.9%7.6%). The combination therapy

    group, however, needed nearly 35% less insulin than the insulin-only group

    (49 versus 78 units) and achieved glycemic control faster, with fewer

    dropouts (3% versus 15%,P\ 0.01). Surprisingly, weight gain was similar

    (4.0 kg) in both groups.

    Insulin and metformin

    Weight gain is a constant occurrence in most clinical trials in which

    insulin or sulfonylureas, or both agents, are used to treat type 2 diabetes.

    Although it was attenuated with combination therapy with sulfonylureas in

    the study by Yki-Jarvinen[38],weight gain remains a problem because it can

    exacerbate insulin resistance and hyperinsulinemia. The use of metformin in

    870 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    7/31

    this situation may prove advantageous because its use is associated with

    reduced weight gain.

    The safety and efficacy of metformin in combination with insulin hasbeen demonstrated in a recent multicenter study by Yki-Jarvinen et al [35].

    In this placebo-controlled study, 96 type 2 diabetics who were poorly

    controlled with oral sulfonylurea therapy (mean glycosylated hemoglobin

    value 9.9% 0.2%; mean fasting plasma glucose level 214 5 mg/dL) were

    randomized to 1 year of treatment with bedtime intermediate-acting insulin

    plus either glyburide (10.5 mg), metformin (2 g), glyburide and metformin,

    or a second injection of intermediate-acting insulin in the morning. Patients

    were taught to adjust the bedtime insulin dose on the basis of fasting glucose

    measurements. At 1 year, body weight remained unchanged in patientsreceiving bedtime insulin plus metformin (mean change 0.9 1.2 kg) but

    increased by 3.9 0.7 kg, 3.6 1.2 kg, and 4.6 1.0 kg, respectively, in

    patients receiving bedtime insulin plus glyburide, bedtime insulin plus both

    oral drugs, and bedtime and morning insulin. In addition, the greatest

    decrease in the glycosylated hemoglobin value was observed in the bedtime

    insulin and metformin group (from 9.7 0.4% to 7.2 0.2%, a difference

    of 2.5 0.4 percentage points) at 1 year (P 0.001 compared with

    baseline and P 0.05 compared with other groups). This group also had

    significantly fewer symptomatic and biochemical cases of hypoglycemia(P 0.05) than the other groups. The authors conclude that combination

    therapy with bedtime insulin plus metformin not only prevents weight gain

    but also seems superior to other bedtime insulin regimens, with respect to

    improvements in glycemic control and frequency of hypoglycemia.

    In a more recent study [44] of approximately 390 type 2 diabetics, the

    combination of insulin and metformin led to a significant improvement in

    glycemic control that was greater than with insulin alone. The mean daily

    glucose level decreased from 141 34 to 137 31 mg/dL in the insulin-only

    group (mean decrease 0.16; 95% confidence interval [CI]; 104 mg/dL)and from 141 40 to 140 31 mg/dL in the metformin group (P= 0.006

    versus placebo; mean decrease 1.04; 95% CI; 27 to 9 mg/dL). The

    mean daily glucose level decreased by 13 mg/dL more in the metformin

    group compared with the placebo groupFig 1.

    Insulin and thiazolidinediones

    The glitazones are potent insulin sensitizers and are, therefore, well suited

    for use in insulin-resistant patients with type 2 diabetes. In several earlystudies, troglitazone was documented to not only improve glycemic control

    but also to reduce exogenous insulin requirements in obese patients with

    type 2 diabetes [45,46]; however, troglitazone was withdrawn from the

    US market as a result of an increased risk of severe idiosyncratic liver

    damage. Presently there are two glitazones available, rosiglitazone and

    pioglitazone, for clinical use in the US, and several more are in development.

    871T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    8/31

    In one 16-week study, Rubin et al[47]demonstrated that the daily additionof 15 and 30 mg of pioglitazone to the regimen of patients receiving a median

    dose of 61 units of insulin resulted in mean FPG reductions of 36 and 49 mg/

    dL and HbA1c reductions of 0.7% and 1.0%, respectively, compared with

    placebo. The insulin-sparing properties of rosiglitazone were shown in a 6-

    month study conducted by Raskin et al [48]. They demonstrated that the

    addition of 2 and 4 mg orally twice daily of rosiglitazone improved HbA1c

    Fig. 1. (A) Blood glucose levels measured at home. (B) Change in blood glucose levels

    measured at home. Data are means with SD error bars. For each time point indicated, the first

    and the second bars show values at baseline and the third and the fourth show values at

    16 weeks. Blood glucose levels in the metformin group compared with the placebo group are all

    significantly lower at 16 weeks (P 0.05). The change in glucose values is also significantly

    greater in the metformin than in the placebo group at all times during the day (P 0.05). (From

    Wulffele MG, Kooy K, Lehert P. Bets D Ogterop JC, Van Der Burg BB, Donker AJM,

    Stehouwer CDA. Combination of insulin and metformin in the treatment of type 2 diabetes.

    Diabetes Care 2002;25(12):213340; with permission.)

    872 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    9/31

    levels by 0.6% and 1.2%, respectively, compared with placebo, in 312

    patients with type 2 diabetes who were uncontrolled on approximately 70

    units of insulin daily (baseline HbA1c 9%). Moreover, insulin requirementswere also reduced by approximately 5 and 10 units, respectively, in the two

    groups treated with rosiglitazone, in keeping with the insulin sensitizing

    effects of the glitazones. In summary, both rosiglitazone and pioglitazone

    improve glucose control in poorly controlled, insulin-treated patients with

    type 2 diabetes mellitus. There have been no reports of insulin added to

    subjects treated with glitazones alone.

    Insulin and a-glucosidase inhibitors

    The addition of acarbose to insulin therapy may be an option in patients

    who have pronounced postprandial hyperglycemia. The first long-term

    controlled study to demonstrate a beneficial effect of acarbose in patients

    on insulin therapy was reported by Chiasson et al[49]. Of the total number

    of patients in this study, 91 were receiving insulin and had glycosylated

    hemoglobin values greater than 7%. Postprandial plasma glucose levels at

    90 minutes were significantly reduced to 282 mg/dL with the addition of

    acarbose, compared with 331 mg/dL seen with insulin alone. Glycosylated

    hemoglobin values decreased by 0.4% in the acarbose group, but, as ex-

    pected, no significant decreases in fasting plasma glucose levels were seen.

    Acarbose may be initiated in patients on insulin treatment by starting with

    a low dose of 25 mg with breakfast and titrating up by 25 mg weekly to 50

    to 100 mg three times daily with meals (100 mg three times daily for

    patients 60 kg body weight), depending on gastrointestinal tolerance and

    efficacy.

    Insulin glargine and oral agents

    The long-acting analog insulin glargine was studied in comparison with

    NPH insulin in 756 patients with type 2 diabetes in an open-label, 24-week,

    multicenter study [50]. In this study, patients who were inadequately

    controlled on oral agents including sulfonylurea, metformin, and glitazones

    were randomized to receive either bedtime insulin glargine or NPH insulin,

    and the doses were adjusted to obtain a target fasting glucose level of less

    than 100 mg/dL (5.6 mmol/L). At the conclusion of the trial, the median

    daily dose of insulin was approximately 0.45 IU/kg of body weight in bothgroups. The two forms of insulin produced a similar improvement in HbA1c(6.96 versus 6.97%) and similar reductions in fasting glucose levels (117

    versus 120 mg/dL); however, the incidence of mild nocturnal hypoglycemia

    was significantly lower among patients treated with insulin glargine than in

    the group treated with NPH insulin (P\ 0.001)[50]. There was a reduction

    of approximately 45% of nocturnal hypoglycemia with glargine compared

    873T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    10/31

    with NPH[50]. Treatment with NPH or glargine in addition to oral therapy

    in type 2 diabetic patients resulted in a decrease of fasting glucose in both

    groups, reaching a plateau by 12 weeks. HbA1c declined at a predictablyslower rate, stabilizing after 18 weeks (Fig. 2) [50].

    Fig. 2. (A) FPG and (B) HbA1cduring the study. Values in both figures are means; error bars

    indicate SE. (FromRiddle MC, Rosenstock J, Gerich J. The Treat-to-Target Trial: Randomized

    addition of glargine or human NPH insulin to oral therapy of type 2 diabetic patients. Diabetes

    Care 2003;26(11):30806; with permission.)

    874 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    11/31

    Selection of patients most likely to succeed on combination treatment

    The most common type of patient in whom combination therapy will

    succeed is the one who is failing oral treatment without significant glucose

    toxicity and has some evidence of responsiveness to oral agents. Patients

    have a higher likelihood of success using combination therapy if they are

    obese, have had overt diabetes for less than 10 to 15 years, are diagnosed

    with type 2 diabetes after the age of 35, do not have fasting blood glucose

    values consistently over 250 to 300 mg/dL, and have evidence of endogenous

    insulin secretory ability. Although standard measurement conditions and C-

    peptide concentrations have not been established for this clinical situation,

    a fasting C-peptide concentration (0.2 nmol/L) or glucagon-stimulated

    level (0.40 nmol/L) indicates some degree of endogenous insulin secretory

    ability[51,52]. Patients with type 2 diabetes diagnosed before the age of 35

    more often have atypical forms of diabetes. Patients who have had diabetes

    for more than 10 to 15 years tend to have a greater chance of beta-cell

    exhaustion and, thus, tend to be less responsive to oral hypoglycemic agents

    and combination therapy. Thin patients are more likely to be hypoinsuli-

    nemic and often respond inadequately to oral agents, which lead to

    combination therapy failure. In addition, markedly elevated fasting glucose

    concentration is often associated with a concomitant decrease in endoge-

    nous insulin secretory ability, which renders oral agents ineffective. The

    actual number of patients who might respond favorably to combination

    therapy is unknown but is estimated to be between 20% and 40%.

    Initiating combination therapy

    Calculation of the initial bedtime dose of intermediate-acting insulin can

    be based on clinical judgment or various formulas based on the fasting

    blood glucose concentration or body weight. For example, the average

    fasting blood glucose (mg/dL) can be divided by 18 or body weight (kg) canbe divided by 10 to calculate the initial dose of NPH or insulin glargine to be

    started at bedtime[43]. Also, 5 to 10 units of insulin can be safely started for

    thin patients, and 10 to 15 units can be started for obese patients at bedtime,

    as an initial estimated dose. In either case, the dose is increased in

    increments of 2 to 5 units every 3 to 4 days until the morning fasting blood

    glucose concentration is consistently in the range of 70 to 120 mg/dL [53].

    The best time to give the evening injection of intermediate-acting insulin

    is between 10 PM and midnight. Insulin glargine has been shown to be

    effective when taken either in the morning or evening. Many reliable pa-tients can make their own adjustments using HGM.

    Based on the results of HGM, combination therapy can be altered to

    reduce hyperglycemia at identified times during the day. For example, a

    common situation seen with daytime oral agents and bedtime intermediate-

    acting insulin therapy is an improvement in the fasting, pre-lunch, and pre-

    dinner blood sugar values, although the post-dinner blood glucose

    875T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    12/31

    concentration remains excessively high (200 mg/dL). In this clinical

    situation, an injection of premixed insulin (70/30 or 75/25 mix) can be

    given before dinner instead of a bedtime dose of intermediate- or long-actinginsulin. This regimen will often improve the post-dinner blood glucose

    values because the premixed insulin contains rapid-acting analogs yet allows

    overnight glucose control secondary to the intermediate-acting component.

    With this regimen, however, one must be more cautious about early

    morning hypoglycemia because the intermediate-acting insulin given before

    dinner will exert its peak effect earlier. In the experience of these authors,

    this has not been a major clinical problem in obese patients with type 2

    diabetes compared with those with type 1 diabetes mellitus. Normally the

    dose of bedtime intermediate- or long-acting insulin can be converted to thedose of premixed insulin, dose per dose, and adjustments can be made

    through HGM.

    Dose adjustment

    Once the fasting blood glucose concentrations are consistently in a desir-

    able range, the pre-lunch, pre-dinner, and bedtime blood sugar values must

    be monitored to determine if the oral hypoglycemic agents are maintaining

    daylong glycemia. It is recommended that after the addition of evening

    insulin patients continue to take the maximal dose of the oral sulfonylurea

    agent. If the daytime blood glucose concentrations become excessively low,

    the dose of oral medication must be reduced. The morning dose of sul-

    fonylurea should be reduced or discontinued first. This situation is common

    because glucose toxicity may be reduced because of improved glucose

    control, leading to enhanced sensitivity to both oral agents and insulin. If

    the pre-lunch and pre-dinner blood glucose concentrations remain exces-

    sively high on combination therapy, it is likely that the oral agents are not

    contributing significantly to glycemic control throughout the day. In this

    situation, a more conventional or intensive regimen of two injections perday is indicated.

    Multiple-injection regimens

    One of the most common insulin regimens used in type 2 diabetes mellitus

    is the split-mixed regimen consisting of a pre-breakfast and pre-dinner dose

    of intermediate- and fast-acting insulin. This split-mixed regimen of two

    injections per day is often inadequate for patients with type 1 or leanpatients with type 2 diabetes and can result in persistent early morning

    hypoglycemia and fasting hyperglycemia. Such problems do not appear to

    occur as frequently in obese type 2 diabetes. This is likely caused by

    pathophysiologic differences, particularly in endogenous insulin secretory

    ability, insulin resistance, and counter-regulatory mechanisms in type 1 and

    type 2 diabetes.

    876 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    13/31

    In a landmark trial with type 2 diabetes by Henry et al [54], daylong gly-

    cemia and glycosylated hemoglobin were essentially normalized by 6 months

    of intensive treatment with a split-mixed insulin regimen. In this study,14 typical obese patients with type 2 diabetes mellitus (age 59 2 years;

    duration of diabetes 7 2 years; body mass index 31 2 kg/m2; fasting

    blood glucose concentration 283 13 mg/dL) failing therapy with oral

    antidiabetic agents were intensively managed with pre-breakfast and pre-

    dinner NPH and regular insulin over a 6-month period. The insulin dose

    was adjusted based on HGM results of four injections per day. Glycemic

    control was rapidly achieved within 1 month and was maintained for the

    duration of the study.

    The average total insulin dose needed to maintain glycemic controlapproached 100 units per day, with approximately 50% of the total dose

    required before breakfast and 50% before dinner. The ratio of NPH to

    regular insulin was approximately 75%:25%. There was a very low incidence

    of mild hypoglycemic reactions, which decreased as the study progressed,

    and no reactions were severe or required assistance. In addition, patient

    compliance and sense of well being were excellent. Near-normalization of

    the glycosylated hemoglobin, however, led to some adverse effects in these

    patients. The mean serum insulin concentration obtained during 24-hour

    metabolic profile studies increased from 308 80 pmol/L at baseline to510 102 pmol/L (P 0.05) at completion of the 6-month study. The

    exacerbation of hyperinsulinemia by exogenous insulin therapy was strongly

    correlated with weight gain throughout the study. Despite biweekly visits

    with the study dietitian and instructions to reduce the daily caloric intake,

    a mean weight gain of approximately 9 kg or 18.8 pounds occurred.

    Interestingly, the total daily insulin dose was 86 13 units at 1 month and

    100 24 units at 6 months, despite minimal additional improvement in

    glycemic control during that period. Most of the improvement in glycemic

    control was caused by the suppression of basal hepatic glucose production(from 628 44 to 350 17 lmol/m2/min,P 0.001), with a more modest

    but significant improvement in peripheral glucose uptake (from 1418 156

    to 1657 128 lmol/m2/min,P 0.05), as determined by the glucose clamp

    technique.

    This study emphasizes a number of important aspects of intensive glucose

    control with insulin in obese subjects with type 2 diabetes. First, the average

    daily dose of insulin needed to control such patients approximates 1 unit per

    kilogram of body weight. Second, the total daily insulin requirement can be

    split equally between the pre-breakfast and pre-dinner injections. Third, thesplit-mixed regimen in patients with type 2 diabetes is usually devoid of the

    common problems seen with this regimen in type 1 diabetes, particularly

    early morning hypoglycemia and fasting (pre-prandial) hyperglycemia.

    Fourth, both severe and mild hypoglycemic events are much less fre-

    quent in patients with type 2 compared with patients with type 1 diabetes

    undergoing intensive insulin therapy. And finally, weight gain with

    877T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    14/31

    peripheral hyperinsulinemia occurs, which may contribute to metabolic and

    vascular complications.

    A similar but larger 3-month clinical trial [38] compared a split-mixedcombination with a multiple-injection regimen consisting of pre-meal

    regular and bedtime NPH insulin injections. Both the split-mixed and

    multiple-injection regimen treatment groups achieved equivalent and near-

    normal glycosylated hemoglobin values. These therapies, however, were

    associated with weight gain of 0.8 0.05 and 2.9 0.05 kg, a 39% and 36%

    increase in mean diurnal serum-free insulin levels, and a total daily insulin

    dose of 43 and 45 units, respectively. The authors demonstrated that the

    change in body weight was negatively correlated with the change in

    glycosylated hemoglobin values and positively correlated with the meandiurnal serum-free insulin values. The differences between these two studies

    with regard to total insulin requirements, mean insulin concentrations, and

    weight gain are primarily the result of differences in patient characteristics.

    Patients in the latter study were leaner (body mass index 29 versus 31 kg/

    m2), had lower baseline fasting blood glucose values (225 versus 283 mg/dL)

    and reduced baseline mean diurnal serum-free insulin values (138 versus 308

    pmol), and were previously treated with submaximal doses of sulfonylureas,

    compared with the patients in the former study. In addition, the latter study

    was conducted over a shorter period of time (3 months versus 6 months).Another long-term (5-year) clinical trial using a split-mixed regimen of two

    injections per day in 102 nonobese type 2 diabetic patients demonstrated

    that excellent glycemic control could be achieved with intensive split-dose

    insulin without significant hypoglycemia but at the expense of progressive

    weight gain[55]. All these studies clearly demonstrate the efficacy of various

    insulin regimens and the adverse consequences of such therapy.

    Premixed insulin approach: rapid-acting insulin analogs

    Rapid-acting insulin analogs are also available as manufactured, pre-

    mixed insulin formulations. One such insulin preparation is Humalog Mix

    75/25, which is a fixed-ratio mixture of 25% rapid-acting insulin lispro and

    75% novel protamine-based intermediate-acting insulin called neutral

    protamine lispro (NPL). NPL was developed to solve the problem of

    instability with prolonged storage that occurs with NPH combined with

    insulin. Studies of the pharmacokinetic and pharmacodynamic profiles of

    NPL show they are comparable to those of NPH insulin[56].Humalog Mix 75/25 was studied in comparison to premixed human

    insulin 70/30 in 89 patients with type 2 diabetes in a 6-month randomized,

    open-label, two-period crossover study[57]. All patients had been previously

    treated with mixed insulin therapies, including short- or rapid-acting and an

    intermediate- or long-acting insulin, twice daily for at least 30 days before

    enrollment. During a 2 to 4 week lead-in period, patients were treated with

    878 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    15/31

    human insulin 70/30. The patients were randomized to receive one of two

    treatment sequences: therapy twice per day with Humalog Mix 75/25 in-

    jected before morning and evening meals for 3 months, after which theywere crossed over to receive human insulin 70/30 using the same dosing

    frequency for an additional 3 months, or the alternate treatment sequence.

    Patients performed self-monitoring blood glucose (SMBG) at scheduled

    intervals during the study period (preprandial, 2-h postprandial, and

    occasional 3 AM readings) and recorded this information along with any

    hypoglycemic episodes in a study diary. Mean insulin doses were similar or

    identical between treatments. Blood glucose values after the morning meal

    were significantly lower during treatment with Humalog Mix 75/25

    (Humalog Mix 75/25 8.95 2.17 versus human insulin 70/30 10.00 2.28 mmol/L, P= 0.017). Treatment with Humalog Mix 75/25 produced

    similar significant blood glucose results 2 hours after the evening meal as

    well (Humalog Mix 75/25 9.28 2.15 versus human insulin 70/30 10.27

    2.76 mmol/L,P = 0.014). Blood glucose results at other time points, HbA1clevels, daytime hypoglycemia, and nocturnal hypoglycemia were not sig-

    nificantly different between treatments. Compared with human insulin 70/

    30, twice-daily injections of Humalog Mix 75/25 in patients with type 2

    diabetes resulted in improved postprandial glycemic control after the

    morning and evening meals, similar overall glycemic control, and the addedconvenience of administration immediately before meals.

    Insulin aspart, another rapid-acting insulin analog, is available in a

    premixed formulation with a protamine-retarded insulin aspart called

    Novolog Mix 70/30 (70% insulin aspart protamine suspension and 30%

    insulin aspart). A comparison study [58] of the pharmacokinetic and

    pharmacodynamic parameters of the Novolog Mix 70/30 and human

    insulin 70/30 in healthy patients showed that the faster onset and greater

    peak action of insulin aspart was preserved in the aspart mixture.

    Another study [59] compared premixed aspart mixture 70/30 withpremixed human insulin 70/30 administered twice daily in a randomized

    12-week open-label trial in 294 patients with type 1 and type 2 diabetes.

    Patients were instructed to inject the human insulin 70/30 30 minutes before

    morning and evening meals and the premixed aspart mixture 10 minutes

    before morning and evening meals. SMBG levels and hypoglycemia

    incidence were recorded in diaries. Patients required a small increase in

    the total daily aspart mixture dose compared with human insulin 70/30

    (mean difference at 12 weeks [95% CI 0.01; 0.05]), P 0.01; 0.03 U/kg.

    There was no significant difference in HbA1c between groups, yet the meanblood glucose values after treatment with the aspart mixture showed

    statistically significant treatment differences after breakfast, before lunch,

    after dinner, and at bedtime. Blood glucose values were approximately

    1.0 mmol/L lower compared with the human insulin 70/30 group at each

    time point (P 0.05). The incidence of hypoglycemia was not found to be

    different between the two groups, and weight gain was not significant during

    879T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    16/31

    the study period with either type of insulin. Treatment with twice-daily

    premixed aspart mixture 70/30 resulted in similar overall glycemic control;

    yet postprandial control improved without additional hypoglycemia andwith injections immediately before meals compared with premixed human

    insulin 70/30 given 30 minutes before the meal.

    In a more recent study that focused on changes in lipid levels, Schwartz

    et al [60] compared insulin 70/30 mix taken twice per day plus metformin

    versus triple oral therapy (secretagogues, metformin, and thiazolidine-

    diones) and clearly demonstrated that insulin plus metformin are superior

    in lowering total cholesterol and triglycerides levels. The baseline values for

    total cholesterol, low-density lipoprotein, high-density lipoprotein, and

    triglycerides indicated no differences between the triple OHA and insulin/metformin groups. By the end of the study (week 24) significant decreases in

    total cholesterol and triglycerides were evident in the insulin plus metformin

    group (P= 0.038 and 0.033, respectively, compared with the triple oral

    therapy group). Subjects in the triple oral therapy group showed a small

    increase in cholesterol and less of a decrease in triglyceride levels [60]. For

    glucose control, both groups had similar FPG values at the beginning of the

    study. After 24 weeks of treatment, the changes from baseline mean FPG

    values were 55 and 65 mg/dL for the triple oral therapy and insulin plus

    metformin, respectively[60]. Baseline HbA1c values were 9.62 1.25% forsubjects in the triple oral therapy and 9.65 1.62% in the insulin group.

    HbA1cvalues at weeks 2 and 6 demonstrated the efficacy of both treatments;

    however, insulin plus metformin treatment achieved improvements in

    HbA1c values at weeks 2 and 6 (9.03 1.35% and 8.11 1.20%,

    respectively) that were significantly greater than the response to triple oral

    therapy (P= 0.001 and 0.001, respectively). At weeks 12 and 24, no

    statistically significant difference in HbA1c between the two groups were

    observed (final values at week 24 were 7.59 1.4% for triple oral therapy

    and 7.59 1.25% for insulin plus metformin [P= 0.772]) (Fig. 3)[60].Along with SMBG, the use of rapid-acting premixed insulin analogs is

    convenient and can be beneficial in reducing postprandial hyperglycemia

    and in helping patients achieve glycemic control without the increased

    incidence of hypoglycemia. In addition, protocols are currently underway to

    assess the efficacy of using rapid-acting premixed insulin analogs three times

    per day before breakfast lunch and dinner, based on HGM data. This

    regimen is more related to a basal bolus strategy discussed below.

    Basalbolus strategy

    The basalbolus insulin strategy, which can be used in patients with

    either type 1 or type 2 diabetes, incorporates the concept of providing

    continuous basal insulin levels throughout the day and night with brief

    increases in insulin levels at the time of meal ingestion by bolus doses.

    880 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    17/31

    The use of pre-meal regular insulin with bedtime NPH as the basal insulin

    has been a common strategy for intensive insulin therapy in the United

    States, but because regular insulin should be administered 20 to 40 minutes

    before meals, a risk of hypoglycemia exists if the meal is delayed. If regular

    Fig. 3. (A) Mean FPG values at screening and weeks 12 and 24 by treatment group. No

    statistically significant changes were observed between the triple oral therapy () and insulin

    plus metformin (). (B) Mean SEM changes for the total cholesterol, HDL, LDL, andtriglycerides at week 24. * Statistically significant (P 0.05) reduction in total cholesterol and

    triglyceride levels in the insulin plus metformin group compared with the triple oral therapy

    group. HDL, high-density lipoprotein; LDL, low-density lipoprotein; OHA, XXX. (From

    Schwartz S, Sievers R, Strange P Lyness W, Hollander P. Insulin 70/30 mix plus metformin

    versus triple oral therapy in the treatment of type 2 diabetes after failure of two oral drugs:

    efficacy, safety, and cost analysis. Diabetes Care 2003;26(8):2598603; with permission.)

    881T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    18/31

    insulin is given just before a meal, high postprandial glucose levels and

    delayed hypoglycemia may result. A strategy that provides for some

    flexibility in the mealtime administration of insulin with the use of rapid-acting insulin analogs, lispro or aspart, administered immediately before

    meals, and long-acting insulin, such as glargine, ultralente, lente, or NPH as

    the basal insulin. These regimens that use multiple doses of intermediate-

    acting insulin such as NPH can be associated with unpredictable nocturnal

    hypoglycemia and day-to-day instability of blood glucose patterns in part

    because of intrapatient variability of the effect of subcutaneous injected

    insulin and the patients peak action profile[61]. NPH, which exhibits peak

    action 5 to 7 hours after administration, has also been used in combination

    with rapid-acting insulin analogs, commonly given at least twice daily,although the disadvantages of NPH used in this manner are similar to those

    associated with Ultralente [62]. Because of its time to peak action, NPH

    should be given every 6 hours or 4 times per day to be effective as a basal

    insulin, in many patients [63].

    Improved mealtime glucose control with the rapid-acting analogs has

    exposed the gaps in basal insulin coverage provided by therapy with the

    traditional intermediate- and long-acting insulin preparations. Taking

    a basal insulin analog with a relatively constant and flat pharmacokinetic

    profile such as insulin glargine once per day will result in a morephysiologic pattern of basal insulin replacement. Insulin glargine in

    combination with a rapid-acting insulin analog has demonstrated effective

    glycemic control and a lower incidence of nocturnal hypoglycemia[64]than

    other insulin preparations currently used for basal insulin supplementation

    [6468].

    Patients on multiple-injection basalbolus regiments should use carbo-

    hydrate counting to estimate their pre-meal bolus dose of a rapid-acting

    analog. In addition, a correction factor should be determined by HGM

    before and after rapid-acting insulin boluses. For example, a typicalinsulin-resistant subject with type 2 diabetes may need 1 unit of lispro or

    aspart for every 8 g of carbohydrate compared with a 1:15 ratio for a lean

    insulin-sensitive person with type 1 diabetes. A typical correction factor

    would be 1 unit of lispro or aspart to bring down the blood glucose value

    to 25 mg/dL compared with a person with type 1 diabetes whose cor-

    rection factor is 1 unit of lispro or aspart to bring down the blood glucose

    value to 50 mg/dL. The carbohydrate to insulin ratio and correction factor

    may be different depending on the time of the day and degree of

    hyperglycemia.The availability of mealtime and basal insulin analogs, combination

    therapy with oral agents, and the use of insulin regimens comprising

    basal and mealtime (bolus) insulin components that better simulate

    normal insulin secretion represent important advances in insulin therapy.

    All of these approaches can have a significant impact on treatment

    outcomes.

    882 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    19/31

    External insulin pump therapy

    External insulin pump therapy or continuous subcutaneous insulin

    infusion (CSII) has been traditionally used mainly in people with type 1

    diabetes. However, insulin pump therapy is extremely valuable in patients

    with type 2 diabetes who require insulin but who have not achieved glycemic

    control with subcutaneous injections or who are seeking for a more flexible

    lifestyle. All of the benefits that are enjoyed by patients with type 1 diabetes

    are shared with people with type 2 diabetes. Many experts believe that

    because of the more physiologic delivery of insulin, glucose control is

    achieved with less insulin than was needed with the subcutaneous insulin

    regimen. This may be caused by a reduction in glucose toxicity and im-

    provement of insulin resistance and beta-cell secretory function as a result of

    improved glycemic control with pump therapy. Weight gain is less of an

    issue because the patient is using less insulin than was used before insulin

    pump therapy. In addition, with the reduction of hypoglycemic events there

    is less overeating to compensate for excessive insulin. Last, it is possible that

    pump therapy may result in less strain placed on the pancreatic beta-cells of

    patients with type 2 diabetes, and this may help with overall glycemic

    control because a functioning beta-cell can also autoregulate against hyper-

    and hypoglycemia, as seen in non-diabetic individuals.

    Many older patients with the diagnosis of insulin-requiring type 2

    diabetes have acute, true, late-onset type 1 diabetes. The literature

    documents large groups of patients with insulin-requiring type 2 diabetes

    who were tested for anti-glutamic acid decarboxylase antibodies with a

    positivity rate of approximately 5% to 8%. These individuals are thinner at

    the time of diagnosis, generally do not respond well to oral agents, and

    require insulin, although they do not present in severe diabetic ketoacidosis.

    These patients generally should be put on an intensive insulin injection

    regimen, and insulin pump therapy should be considered.

    Insulin pump therapy allows for increased flexibility in meal timing and

    amounts, increased flexibility in the time and intensity of exercise, improved

    glucose control while traveling across time zones or with variable working

    schedules, and quality of life in terms of self-reliance and control.

    Because pumps use only regular and fast-acting insulin, there is no

    peaking of injected intermediate- and long-acting insulins, which do not

    provide as constant a basal rate caused by variable absorption and

    pharmacokinetics. Insulin glargine is an exception in that it serves as

    excellent basal insulin. Variable insulin absorption and pharmacokinetics

    are probably responsible for up to 50% to 60% of the day-to-day

    fluctuation in blood glucose values in individuals using multiple-injection

    regimens with various insulin types. Insulin pump therapy allows for more

    regular insulin absorption and pharmacokinetic profile, resulting in im-

    proved reproducibility in insulin availability and reduced fluctuations in

    glycemic control [62].

    883T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    20/31

    Presently, there is a paucity of clinical trials using insulin pumps in type 2

    diabetes, but pump therapy is a viable option in insulin-requiring patients

    with type 2 diabetes who are unable to achieve adequate glycemic controlwith multiple-injection regimens. Although some studies demonstrate

    metabolic benefits of pump therapy in type 2 diabetes, all are limited by

    a relatively short period of evaluation and a small number of heterogeneous

    subjects. Interpretation of these studies is further confounded by the

    random assignment of subjects to dissimilar conventional insulin regimens,

    making comparison between studies difficult.

    Garvey et al[69]studied the effect of intensive insulin therapy on insulin

    secretion and insulin action before and after 3 weeks of CSII therapy in 14

    patients with type 2 diabetes (age 50 3 years, duration of diabetes 7.8 2.1 years, and 119% ideal body weight). In 3 weeks of therapy, the mean

    fasting plasma blood glucose and glycosylated hemoglobin values fell 46%

    and 38%, respectively. The mean daily insulin dose stabilized at approxi-

    mately 110 units/d, and there was a 74% increase in the insulin-stimulated

    glucose disposal rate and a 45% reduction in hepatic glucose output to

    mean levels similar to those of normal subjects. In addition, there were

    significant improvements in both endogenous insulin and C-peptide

    secretion. This study demonstrated that pump therapy was feasible and ef-

    fective at improving metabolic control and reversing glucose toxicity inthese poorly controlled subjects with type 2 diabetes.

    Jennings et al[70]randomized 20 type 2 diabetic subjects (median age 61

    years, duration of diabetes 6 years, and percentage of ideal body weight

    120%) to either CSII or twice-daily injections of regular and NPH insulin for

    4 months. Glycemic control improved in both groups, although there was

    a 30% reduction in the glycosylated hemoglobin in the CSII-treated group

    and only a 17% reduction in the twice-daily injection-treated group. There

    were no significant differences between the two groups in median daily

    insulin requirement (0.58 versus 0.65 units/kg), weight gained (4.5 versus4.2 kg), prevalence of mild hypoglycemic reactions, or patient acceptance. In

    addition, in the CSII group 58% of the total daily insulin requirement was

    given as a basal infusion, with the remainder as pre-meal bolus injections

    using insulin algorithms. This ratio of basal to bolus insulin requirements are

    similar to the rates commonly used in type 1 diabetes, but there are

    characteristics of pump therapy that are very different in type 2 diabetes.

    In a more recent study, Pouwels et al[71]prospectively studied 8 patients

    with poorly controlled (HbA1c 12.0 1.7%) type 2 diabetes with high

    insulin requirements (1.92 0.66 U/kg/d). The subjects where aggressivelytreated with intravenous (IV) insulin for approximately 1 month followed by

    12 months of CSII therapy. Insulin sensitivity and secretion were measured

    before and after the IV insulin treatment phase.

    Euglycemia was achieved after 12 days of IV therapy, and the insulin

    requirements eventually reduced from 1.7 0.09 to 1.1 0.06 U/kg/d (P

    0.005) during the IV treatment phase of the protocol. Whole-body glucose

    884 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    21/31

    uptake increased from 12.7 5.7 to 22.4 8.8 lmol/kg/min (P 0.0005), and

    in 1 month of intensive therapy, the HbA1cdropped to 8.9% 1.2% with no

    significant change in the patients body weight. After 6 and 12 months of CSIItherapy, the mean HbA1c values were 7.1 0.6% and 8.3 1.4%, respectively

    (P 0.001 versus pretreatment values for all time points)[71].

    In another recent study [72], 132 CSII nave type 2 diabetics were

    randomized to the pump or multiple daily injections (MDI). This study

    showed that pump therapy provided efficacy and safety equivalent to MDI

    therapy. Lower 8-point blood glucose values were shown by the CSII group at

    most time points (values were only significant 90 min after breakfast; 167

    47.5 mg/dL versus 192 65.0 mg/dL for CSII and MDI, respectively;

    P= 0.019) (Fig. 4).In summary, insulin pump therapy has not been fully evaluated in

    patients with type 2 diabetes. From published studies, however, it is

    apparent that CSII therapy can safely improve glycemic control while

    limiting hypoglycemia. CSII may be particularly useful in treating patients

    with type 2 diabetes who do not respond satisfactorily to more conventional

    insulin treatment strategies.

    Alternative insulin delivery systems

    Inhaled insulin is currently under development by several pharmaceutical

    companies for use in people with type 1 and type 2 diabetes. The insulin is

    Fig. 4. Baseline and end-of-study 8-point blood glucose profiles (mean SEM) for the intent-

    to-treat population. Dashed lines represent baseline profiles; solid lines represent end-of-studyprofiles; , means for CSII; n, means for MDI therapy. Number of patients at each time point:

    CSII, 5663; MDI, 5459. *P, 0.02. BB, before breakfast; B90, 90 minutes after breakfast;

    BL, before lunch;L90, 90 minutes after lunch; BD, before dinner;D90, 90 minutes after dinner;

    BE, at bedtime. (From Raskin P, Bode BW, Marks JB, Hirsh IB, Weinstein RL, McGill

    JB, Peterson GE, Mudaliar SR, Reinhardt RR. Continuous subcutaneous insulin infusion and

    multiple daily injection are equally effective in type 2 diabetes: a randomized, parallel-group,

    24-week study. Diabetes Care 2003;26(9):2598603; with permission.)

    885T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    22/31

    contained in a pellet and is vaporized in an inhaler, which aerosolizes the

    liquid insulin. Inhaled insulin can also be delivered to the pulmonary

    microvasculature as a dry powder system and inhaled through a mouthpiece.It provides the obvious incentive for diabetic patients to use insulin without

    the need for injections.

    Cefalu et al[73]conducted a randomized, open-label, 3-month study with

    26 patients (16 men, 10 women; average age, 51.1 years) with type 2 diabetes

    (average duration of diabetes, 11.2 years). Patients received inhaled insulin

    before each meal plus a bedtime injection of ultralente insulin, performed

    HGM, and adjusted their insulin dose weekly. The target level for

    preprandial plasma glucose was 100 to 160 mg/dL. At the end of 3 months,

    inhaled insulin treatment significantly improved glycemic control comparedwith baseline, and mean HbA1c levels decreased by 0.07%. Hypoglycemic

    events were mild, and patients showed no significant weight gain or change

    in pulmonary function compared with baseline. Thus in this study, pul-

    monary delivery of insulin in type 2 diabetic patients who require insulin

    improved glycemic control was well tolerated and demonstrated no short-

    term adverse pulmonary effects [74].

    A new Aerodose insulin inhaler proved to be comparable to sub-

    cutaneous injections through overlapping dose-response curves, with con-

    sistent relative bioavailability and relative biopotency. The inhaler delivereda pharmacologically predictable insulin dose to type 2 diabetics, similar

    to that with subcutaneous insulin injections[75].The Aerodose inhaler used

    regular insulin (Humulin R), U-500, whereas Humulin R, U-100 was used

    for subcutaneous injections. Serum insulin levels before exogenous insulin

    administrations were similar between inhaled and subcutaneously inject

    insulin (t, baseline,P = 0.12). At the end of dosing (t, 0 min), serum insulin

    levels were significantly higher for inhalation treatments than for sub-

    cutaneously injected treatments, indicating rapid, systemic insulin absorp-

    tion following inhalation. The area under the curve (AUC)0-8h andmaximum serum insulin concentration demonstrated a clear dose-response

    relationship for the three doses of inhaled insulin and the three doses of

    subcutaneously injected insulin (Fig. 5)[75].

    Insulin can also be taken orally by capsules, enterocoated with a soybean

    trypsin inhibitor that prevents insulin degradation. This approach has

    clinical potential, but large clinical trials have not been carried out.

    Chemically modified human insulin, called hexyl insulin, using proprietary

    conjugation technology to improve its stability and oral absorption has

    shown promise. Preliminary results reported that in healthy humanvolunteers, hexyl insulin caused dose-dependent glucose lowering, was safe,

    and was well tolerated. In a small clinical study, oral insulin illustrates the

    similarities and differences among hexyl insulin monoconjugate (HIM)2 oral

    insulin, subcutaneous insulin, and placebo. All three curves are indistin-

    guishable from each other during the first hour postdose. The placebo curve

    then separates from the other two curves, displaying a significantly higher

    886 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    23/31

    peak excursion. The HIM2 and subcutaneous insulin curves remain nearly

    indistinguishable for at least another hour. During the fourth hour

    postdose, the HIM2 curve clearly separates from the subcutaneous insulincurve, becoming nearly identical to the placebo curve, as the glucose

    excursion values in all three groups decline toward baseline. Peripheral

    plasma insulin revealed an initial peak in peripheral plasma insulin

    concentrations following administration of HIM2; however, this initial

    insulin peak was caused by one patient who had a rapid peak of insulin. The

    median insulin Cmax values, following administration of HIM2 and sub-

    cutaneous regular insulin, were nearly identical (Fig. 6)[76].

    Amylin analog: a novel injectable peptide that compliments the action

    of insulin

    Destruction and dysfunction of pancreatic beta-cells, resulting in

    absolute and relative insulin deficiency, represent key abnormalities in the

    pathogenesis of type 1 and type 2 diabetes, respectively [77]. Following the

    Fig. 5. Glucose infusion rate (GIR) registered following administration of inhaled insulin (,

    80 units; n,160 units; , 240 units) and subcutaneous injection (}, 8 units; , 16 units; , 24

    units) in patients with type 2 diabetes. GIRs have been averaged over 30-minute periods. Datapoints are means SE (n = 16) at each time point for low, medium, and high doses for inhaled

    and injected insulin. (From Kim D, Mudaliar S, Chinnapongse S, Chu N, Boies SM, Davis T,

    Perera AD, Fishman RS, Shapiro DA, Henry R. Dose-response relationships of inhaled insulin

    delivered via the Aerodose insulin inhaler and subcutaneously injected insulin in patients with

    type 2 diabetes. Diabetes Care 2003;26(10):28427; with permission.)

    887T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    24/31

    888 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    25/31

    discovery of amylin in 1987, a second beta-cell 37-amino acid hormone that

    is co-secreted with insulin in response to nutrient stimuli, it was realized that

    diabetes represents a state of bi-hormonal beta-cell deficiency and that a lackof amylin action may contribute to abnormal glucose homeostasis.

    Experimental studies show that amylin acts as a neuroendocrine hormone

    that complements the effects of insulin in postprandial glucose regulation

    through several centrally mediated effects. These include a suppression of

    postprandial glucagon secretion and a vagus mediated regulation of gastric

    emptying, thereby helping to control the inflow of endogenous and

    exogenous glucose, respectively. In animal studies, amylin also reduces

    food intake and body weight, consistent with an early satiety effect[78].

    Insulin is the major hormonal regulator of glucose disposal. Preclinicaland clinical studies indicate that amylin complements the effects of insulin

    by regulating the rate of glucose inflow to the bloodstream, suppressing

    glucagons secretion and inducing satiety.

    Pramlintide is a soluble, nonaggregating, injectable, synthetic analog of

    human amylin currently under development for the treatment of type 1 and

    insulin-using type 2 diabetes. Long-term clinical studies have consistently

    demonstrated that prandial subcutaneous. injections of pramlintide, in

    addition to the current insulin regimen, reduce HbA1c and body weight in

    type 1 and type 2 diabetic patients, without an increase in insulin use or inthe incidence of severe hypoglycemia[79].

    Treatment with 120 lg twice per day of pramlintide in subjects with type

    2 diabetes led to a sustained reduction from baseline in HbA1c (0.68 and

    0.62% at weeks 26 and 52, respectively) that was significantly greater than

    that in the placebo group (P\ 0.05) (Fig. 7)[80]. The greater reduction of

    HbA1c observed with pramlintide was not accompanied by an increase in

    body weight. Instead, patients in both pramlintide treatment groups

    experienced a sustained reduction in body weight that was significantly

    different from placebo at week 26 (both P 0.05) [80]. In the group ofsubjects who took 120 lg twice daily, the reduction in body weight was

    sustained to week 52 (P 0.05 versus placebo).

    The most commonly observed side effects were gastrointestinal-related,

    mainly mild nausea, which typically occurred on initiation of treatment and

    Fig. 6. 0.5 mg/kg and 1.0 mg/kg HIM2 dose groups; pooled data. (A) Mean plasma glucose

    excursion versus time profiles and (B) mean plasma insulin concentration versus time profiles.

    At time 0, patients received 0.5 or 1.0 mg/kg oral HIM2, 8 units subcutaneous regular insulin or

    oral placebo. At 30 min, patients began ingesting the standardized meal (Boost Plus). Patientsingested the entire meal over a 10-minute period. Postprandial plasma glucose excursions and

    insulin concentrations were determined from blood samples collected a the time points

    indicated. Data are expressed as means SE (n = 12 patients). , oral HIM2 (0.5) and 1.0 mg/kg

    dose groups combined); , 8 units of subcutaneous regular insulin; , oral placebo. (From Kipnes

    M, Dandona P, Tripathy D, Still JG, Kosutic G. Control of postprandial plasma glucose by an

    oral insulin product (hexyl-insulin monoconjugate 2 [HIM2]) in patients with type 2 diabetes.

    Diabetes Care 2003;26(2):4216; with permission.)

    =

    889T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    26/31

    resolved within days or weeks. Amylin replacement with pramlintide as an

    adjunct to insulin therapy is a novel physiological approach toward

    improved long-term glycemic and weight control in patients with type 1

    and type 2 diabetes.

    Summary

    Type 2 diabetes is a common disorder often accompanied by numerous

    metabolic abnormalities leading to elevated rates of cardiovascular morbid-

    ity and mortality. Improved glycemia will delay or prevent the development

    of microvascular disease and reduce many or all of the acute and subacute

    complications that worsen the quality of daily life. Exogenous insulin is

    Fig. 7. Change from baseline in mean HbA1c (A) and weight (B) (intent-to-treat population).

    *P 0.05 for treatment arm versus placebo. , placebo; n, 90 lg twice daily; , 120 lg. (FromHollander PA, Levy P, Fineman MS, Maggs DG, Shen LZ, Strobel SA, et al. Pramlintide as

    an adjunct to insulin therapy improves long-term glycemic and weight control in patients with

    type 2 diabetes: a 1-year randomized controlled trial. Diabetes Care 2003;26(3):78490; withpermission.)

    890 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    27/31

    usually the last line of treatment used to normalize glycosylated hemoglobin

    in patients with type 2 diabetes who have failed other therapeutic modalities.

    Not all patients are candidates for aggressive insulin management; therefore,the goals of therapy should be tailored to the individual. Candidates for

    intensive management should be motivated, compliant, educable, and

    without other medical conditions and physical limitations that would

    preclude accurate and reliable HGM and insulin administration.

    In selected patients, combination therapy with insulin and oral antidia-

    betic medications can be an effective method for normalizing glycemia

    without the need for rigorous insulin regimens. The most common clinical

    situation in which combination therapy can be successful occurs in patients

    who are failing daytime oral agents therapy and still show some evidence ofresponsiveness to the medications. Bedtime intermediate- and long acting-

    insulin are administered and progressively increased until the fasting blood

    glucose concentration is normalized. Additional benefits of combination

    therapy include ease of administration, excellent patient compliance and

    safety, and lower exogenous insulin requirements with less peripheral

    hyperinsulinemia and weight gain. If combination therapy is not successful,

    a split-mixed regimen of an intermediate- and a fast-acting insulin equally

    divided between the pre-breakfast and pre-dinner periods can be effective

    especially in obese patients.For patients who do not achieve glucose control on combination or split-

    mixed regimens, an intensive basal bolus multiple-injection regimen is

    indicated. Continuous subcutaneous insulin infusion pumps can be partic-

    ularly useful in treating patients with type 2 diabetes mellitus who do not

    respond satisfactorily to more conventional treatment strategies. The use of

    fast-acting insulin analogs should be used in the majority of insulin-requiring

    diabetics because of its more physiologic pharmacokinesis. Inhaled insulin

    and the amylin analog pramlintide also hold promise to intensively control

    glycemia in patients with insulin-requiring type 2 diabetes.The glycemic objectives for patients with type 2 diabetes should be similar

    to those for patients with type 1 diabetes, namely, to normalize glycemia and

    glycosylated hemoglobin without causing undue weight gain or hypoglyce-

    mia or adversely affecting the quality of daily life. This is best achieved in

    a multidisciplinary setting using complementary therapeutic modalities that

    include a combination of diet, exercise, and pharmacologic therapy.

    Emphasis should be placed on diet and exercise initially, and throughout

    the course of management as well, since even modest success with these

    therapies will enhance the glycemic response to both oral antidiabetic agentsand insulin.

    References

    [1] American Podiatric Medical Association. E-foot FAQS [frequently asked questions]. 2002.

    Available at:http://www.apma.org/faqsdiab.html. Accessed March 12, 2003.

    891T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

    http://www.apma.org/faqsdiab.htmlhttp://www.apma.org/faqsdiab.html
  • 8/13/2019 Curso Insulin Therapy

    28/31

    [2] Bennett PH, Rewers MJ, Knowler WC. Epidemiology of diabetes mellitus. In: Porte DJ,

    Sherwin RS, editors. Ellenberg and Rifkins diabetes mellitus. Stamford: Appleton &

    Lange; 1997. p. 37397.

    [3] Sherwin RS. Diabetes mellitus. In: Goldman L, Bennett JC, editors. Cecil textbook of

    medicine. Philadelphia: WB Saunders; 2000. p. 126385.

    [4] American Academy of Family Physicians. Number of visits to family physicians by the 45

    most frequent primary diagnoses. 2002. National Ambulatory Medical Care Survey 2000.

    Department of Health and Human Services, Centers for Disease Control and Preven-

    tion. National Center for Health Statistics. Available at: http://www.aafp.org/practice/

    icd9code.html. Accessed March 12, 2003.

    [5] Hiatt WR. Atherosclerotic peripheral arterial disease. In: Goldman L, Bennett JC, editors.

    Cecil textbook of medicine. Philadelphia: WB Saunders; 2000. p. 35762.

    [6] Prospective Diabetes Study UK (UKPDS) Group. Intensive blood-glucose control with

    sulphonylureas or insulin compared with conventional treatment and risk of complications

    in patients with type 2 diabetes (UKPDS 33). Lancet 1998;352:83753.

    [7] Shichiri M, Kishikawa H, Ohkubo Y, Wake N. Long-term results of the Kumamoto Study

    on optimal diabetes control in type 2 diabetic patients. Diabetes Care 2000;23(Suppl 2):

    B219.

    [8] Galloway JA. Treatment of NIDDM with insulin agonists or substitutes. Diabetes Care

    1990;13:1209.

    [9] DeFronzo RA. Insulin resistance, hyperinsulinemia, and coronary artery disease: A com-

    plex metabolic web. J Cardiovasc Pharm 1992;20(Suppl 11):S116.

    [10] Weyer C, Bogardus C, Mott DM, Pratlet RE. The natural history of insulin secretory

    dysfunction and insulin resistance in the pathogenesis of type 2 diabetes mellitus. J Clin

    Invest 1999;104:78794.[11] American Diabetes Association. The pharmacologic treatment of hyperglycemia in

    NIDDM. Diabetes Care 1995;18:15108.

    [12] Caro JF. Insulin resistance in obese and nonobese man (Clinical Review 26). J Clin

    Endocrinol Metab 1991;73:691.

    [13] Genuth S. Insulin use in NIDDM. Diabetes Care 1990;13:1240.

    [14] Duckworth WC, Saudek CD, Henry RR, Veterans Affairs Study Group. Perspectives in

    diabetes: why intraperitoneal delivery of insulin with implantable pumps in NIDDM?

    Diabetes 1992;41:657.

    [15] Saudek CD, Duckworth WC, Veterans Affairs Study Group. The Department of Veterans

    Affairs implanted insulin pump study. Diabetes Care 1992;15:567.

    [16] Department of Veterans Affairs Implantable Insulin Pump Study Group. Implantableinsulin pump vs multiple-dose insulin for non-insulin-dependent diabetes mellitus. JAMA

    1996;276:13227.

    [17] American Diabetes Association. Clinical practice recommendations american diabetes

    association 1999. Diabetes Care 1999;22(Suppl 1):2.

    [18] Colwell JA. Controlling type 2 diabetes: are the benefits worth the costs? JAMA 1997;

    278(20):1700.

    [19] Tuomi T, Groop LC, Zimmet PZ, Rowley MJ, Knowles W, Mackay IR. Antibodies to

    glutamic acid decarboxy - lase reveal latent autoimmune diabetes mellitus in adults with

    a non-insulin-dependent onset of disease. Diabetes 1993;42:359.

    [20] American Diabetes Association. Diabetes Dictionary. 2004. Available at: http://www.

    diabetes.org/diabetesdictionary.jsp. Accessed on October 4, 2003.[21] Yu A, Wu PS, Edelman SV. The natural history of non-insulin dependent diabetes mellitus

    in a Filipino migrant population. Presented at the 3rd International Diabetes Federation,

    Western Pacific Regional Congress: Hong Kong; September 2528, 1996.

    [22] Vora JP, Burch A, Peters JR, Owens DR. Relationship between absorption of radiolabeled

    soluble insulin, subcutaneous blood flow, and anthropometry. Diabetes Care 1992;15:

    148493.

    892 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

    http://www.aafp.org/practice/icd9code.htmlhttp://www.aafp.org/practice/icd9code.htmlhttp://www.diabetes.org/diabetesdictionary.jsphttp://www.diabetes.org/diabetesdictionary.jsphttp://www.diabetes.org/diabetesdictionary.jsphttp://www.diabetes.org/diabetesdictionary.jsphttp://www.aafp.org/practice/icd9code.htmlhttp://www.aafp.org/practice/icd9code.html
  • 8/13/2019 Curso Insulin Therapy

    29/31

    [23] Edelman SV. Taking control of your diabetes. Caddo (OK): Professional Communica-

    tions, Inc., 2000.

    [24] Bailey TS, Mezitis NHE. Combination therapy with insulin and sulfonylureas for type 2

    diabetes. Diabetes Care 1990;13:687.

    [25] Lebovitz HE, Pasmantier R. Combination insulin-sulfonylurea therapy. Diabetes Care

    1990;12:667.

    [26] Groop LC, Widen E, Ekstrand A, Saloranta C, Franssila-Kallunki A, Schalin-Jantti C,

    et al. Morning or bedtime NPH insulin combined with sulfonylurea in treatment of

    NIDDM. Diabetes Care 1992;15:831.

    [27] Pugh JA, Wagner ML, Sawyer J, Ramirez G, Tuley M, Friedberg SJ. Is combination

    sulfonylurea and insulin therapy useful in NIDDM patients? A metaanalysis. Diabetes

    Care 1992;15:953.

    [28] Riddle MC. Evening insulin strategy. Diabetes Care 1990;13:676.

    [29] Taskinen M-R, Sane T, Helve E, Karonen SL, Nikkila EA, Yki-Jarvinen H. Bedtime

    insulin for suppression of overnight free-fatty acid, blood glucose, and glucose production

    in NIDDM. Diabetes 1989;38:580.

    [30] Karlander SG, Gutniak MKM, Efendic S. Effects of combination therapy with glyburide

    and insulin on serum lipid levels in NIDDM patients with secondary sulfonylurea failure.

    Diabetes Care 1991;14:963.

    [31] Mezitis NHE, Heshka S, Saitas V, Bailey TS, Costa R, Pi-Sunyer FX. Combination therapy

    for NIDDM with biosynthetic human insulin and glyburide. Diabetes Care 1992;15:265.

    [32] Vigneri R, Trischitta V, Italia S, Mazzarino S, Rabuazzo MA, Squatrito S. Treatment of

    NIDDM patients with secondary failure to glyburide: Comparison of the addition of either

    metformin or bedtime NPH insulin to glyburide. Diabetes Metab 1991;17:232.

    [33] Raskin P. Combination therapy in NIDDM. N Engl J Med 1992;327:1453.[34] Marks JB. Combination glyburide and insulin treatment in secondary sulfonylurea failure.

    Clinical Diabetes 1994;12:21.

    [35] Yki-Jarvinen H, Ryysy L, Nikkila K, Tulokas T, Vanamo R, Heikkila M. Comparison

    of bedtime insulin regimens in patients with type 2 diabetes mellitus. A randomized,

    controlled trial. Ann Int Med 1999;130(5):38996.

    [36] Riddle MC, Schneider J. Beginning insulin treatment of obese patients with evening 70/30

    insulin plus glimepiride versus insulin alone. Glimepiride Combination Group. Diabetes

    Care 1998;21(7):10527.

    [37] Johnson JL, Wolf SL, Kabadi UM. Efficacy of insulin and sulfonylurea combination

    therapy in type II diabetes. A meta-analysis of the randomized placebo-controlled trials.

    Arch Intern Med 1996;156:25964.[38] Yki-Jarvinen H, Kauppila M, Kujansuu E, Lahti J, Marjanen T, Niskanen L, et al.

    Comparison of insulin regimens in patients with non-insulin-dependent diabetes mellitus.

    N Engl J Med 1992;327:1426.

    [39] Riddle MC, Hart J, Bingham P, Garrison C, McDaniel P. Combined therapy for obese type 2

    diabetes: suppertime mixed insulin withdaytime sulfonylurea. Am J Med Sci 1992;303:1516.

    [40] Shank ML, Del Prato S, DeFronzo RA. Bedtime insulin/daytime glipizide: effective

    therapy for sulfonylurea failures in NIDDM. Diabetes 1995;44:16572.

    [41] Koivisto VA. Insulin therapy in type 2 diabetes. Diabetes Care 1993;16(Suppl 3):S29.

    [42] Riddle MC. Evening insulin strategy. Diabetes Care 1990;13:676.

    [43] Turner RC, Holman RR. Insulin use in NIDDM. Rationale based on pathophysiology

    of disease. Diabetes Care 1990;13:1011.[44] Wulffele MG, Kooy K, Lehert P, Bets D, Ogterop JC, Van Der Burg BB, et al.

    Combination of insulin and metformin in the treatment of type 2 diabetes. Diabetes Care

    2002;25(12):213340.

    [45] Buse JB, Gumbiner B, Mathias NP, Nelson DM, Faja BW, Whitcomb RW, and the

    Troglitazone Insulin Study Group. Troglitazone use in insulin-treated type 2 diabetic

    patients. Diabetes Care 1998;21(9):145561.

    893T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    30/31

    [46] Schwartz S, Raskin P, Fonseca V, and the Troglitazone Exogenous Insulin Study Group.

    Effects of troglitazone in insulin-treated patients with type 2 diabetes. Ann Intern Med

    1998;338:8616.

    [47] Rubin C, Egan J, Schneider R, et al. Combination therapy with pioglitazone and

    insulin in patients with type 2 diabetes [abstract 0474]. Diabetes 1999;48(Suppl 1):A110.

    [48] Raskin P, Dole JF, Rappaport EB. Rosiglitazone improves glucose control in poorly

    controlled, insulin treated patients with type 2 diabetes mellitus [abstract 0404]. Diabetes

    1999;48(Suppl 1):A95.

    [49] Chiasson J-L, Josse RG, Hunt JA, Palmason C, Rodger NW, Ross SA, et al. The efficacy

    of acarbose in the treatment of patients with noninsulin-dependent diabetes mellitus:

    a multicenter controlled clinical trial. Ann Intern Med 1994;121:92835.

    [50] Riddle MC, Rosenstock J, Gerich J. The Treat-to-Target Trial: Randomized addition of

    glargine or human NPH insulin to oral therapy of type 2 diabetic patients. Diabetes Care

    2003;26(11):30806.

    [51] Polonsky KS, Rubenstein AH. C-peptide as a measure of the secretion and hepatic

    extraction of insulin. Pitfalls and limitations. Diabetes 1984;33:486.

    [52] Greco AC, Caputo S, Bertoli A, Ghirlanda G. The beta cell function in NIDDM patients

    with secondary failure: A three year follow-up of combined oral hypoglycemic and insulin

    therapy. Horm Metab Res 1992;24:280.

    [53] Edelman SV, Henry RR. Diagnosis and management of type 2 diabetes. Caddo (OK):

    Professional Communications, Inc.; 2002. p. 132.

    [54] Henry RR, Gumbiner B, Ditzler T, Wallace P, Lyon R, Glauber HS. Intensive

    conventional insulin therapy for type II diabetes. Metabolic effects during a 6-mo out-

    patient trial. Diabetes Care 1993;16(1):2131.

    [55] Kudlacek S, Schernthaner G. The effect of insulin treatment on HbAlc, body weight andlipids in type 2 diabetic patients with secondary failure to sulfonylureas. A five year follow-

    up study. Horm Metab Res 1992;24:478.

    [56] Roach P, Woodworth JR. Clincal pharmacokinetics and pharmacodynamics of insulin

    lispro mixtures. Clini Pharmacokinet 2002;41(13):104357.

    [57] Roach P, Yue L, Arora V for the Humalog Mix25 Study Group. Improved postprandial

    glycemic control during treatment with Humalog Mix25, a novel protamine-based insulin

    lispro formulation. Diabetes Care 1999;22(8):125861.

    [58] Jacobsen LV, Sogaard B, Riis A. Pharmacokinetics and pharmacodynamics of a premixed

    formulation of soluble and protamine-retarded insulin aspart. Eur J Clin Pharmacol 2000;

    56(5):399403.

    [59] Boehm BO, Home PD, Brehend C, Kamp NM, Lindholm A. Premixed insulin aspart 30vs. premixed human insulin 30/70 twice daily: a random mixed trial in type 1 and type 2

    diabetic patients. Diabet Med 2002;19:3939.

    [60] Schwartz S, Sievers R, Strange P, Lyness W, Hollander P. Insulin 70/30 mix plus

    metformin versus triple oral therapy in the treatment of type 2 diabetes after failure of two

    oral drugs: efficacy, safety, and cost analysis. Diabetes Care 2003;26(8):223843.

    [61] Leahy JT. Intensive insulin therapy in type 1 diabetes mellitus. In: Leahy JT, Cefalu WT,

    editors. Insulin therapy. New York: Marcel Dekker, Inc.; 2002. p. 87112.

    [62] Lepore M, Pampanelli S, Fanelli C, et al. Pharmacokinetics and pharmacodynamics of

    subcutaneous injection of long-acting human insulin analog glargine, NPH insulin, and

    ultralente human insulin and continuous subcutaneous infusion of insulin lispro. Diabetes

    2000;49:21428.[63] Rossetti P, Pampanelli S, Fanelli C, Porcellati F, Costa E, Torlone E, et al. Intensive

    replacement of basal insulin in patients with type 1 diabetes given rapid-acting insulin

    analog at mealtime. Diabetes Care 2003;26(5):14907.

    [64] Yki-Ja rvinen H, Dressler A, Ziemen M for the HOE 901/3002 Study Group. Less

    nocturnal hypoglycemia and better post-dinner glucose control with bedtime insulin

    894 T. Davis, S.V. Edelman / Med Clin N Am 88 (2004) 865895

  • 8/13/2019 Curso Insulin Therapy

    31/31

    glargine compared with bedtime NPH insulin during insulin combination therapy in type 2

    diabetes. Diabetes Care 2000;23:11306.

    [65] Raskin P, Klaff L, Bergenstal R, Halle JP, Donley D, Mecca T. A 16-week comparison of

    the novel i