Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Aug 28.
Published in final edited form as: Expert Opin Drug Deliv. 2013 Nov 12;10(12):1699–1709. doi: 10.1517/17425247.2013.856877

Intranasal Insulin Therapy for Cognitive Impairment and Neurodegeneration: Current State of the Art

Suzanne M de la Monte 1
PMCID: PMC4551402  NIHMSID: NIHMS717108  PMID: 24215447

Abstract

Introduction

Growing evidence supports the concept that insulin resistance plays an important role in the pathogenesis of cognitive impairment and neurodegeneration, including in Alzheimer's disease (AD). The metabolic hypothesis has led to the development and utilization of insulin- and insulin agonist-based treatments. Therapeutic challenges faced include the ability to provide effective treatments that do not require repeated injections and also minimize potentially hazardous off-target effects.

Areas covered

This review covers the role of intra-nasal insulin therapy for cognitive impairment and neurodegeneration, particularly Alzheimer's disease. The literature reviewed focuses on data published within the past 5 years as this field is evolving rapidly. The author provides evidence that brain insulin resistance is an important and early abnormality in Alzheimer's disease, and that increasing brain supply and utilization of insulin improves cognition and memory. Emphasis was placed on discussing outcomes of clinical trials and interpreting discordant results to clarify the benefits and limitations of intranasal insulin therapy.

Expert Opinion

Intranasal insulin therapy can efficiently and directly target the brain to support energy metabolism, myelin maintenance, cell survival, and neuronal plasticity, which begin to fail in the early stages of neurodegeneration. Efforts must continue toward increasing the safety, efficacy, and specificity of intranasal insulin therapy.

Keywords: Alzheimer's disease, diabetes mellitus, insulin-like growth factor, insulin resistance, insulin sensitizers, intranasal insulin, mild cognitive impairment, obesity, PPAR agonists

1. Introduction

a) Presence and Trafficking of Insulin in the Brain

Insulin polypeptide and insulin receptor are expressed in the mature and developing brains 17. Insulin receptor expression is widely distributed throughout the brain 1, but most abundant in cortical-limbic structures, including the olfactory bulb, hypothalamus, hippocampus, amygdala, and cerebral cortex (orbitofrontal and cingulate regions) 3, 8, 9, as well as in the cerebellum 10, 11. Although debate continues concerning insulin synthesis in the brain, independent investigators have shown that the messenger RNA is expressed in neurons within the same regions that have high level receptor expression 2, 3. At the same time there is excellent evidence that insulin is taken up into the brain from the peripheral circulation 1, and following intranasal administration 12, 13. Although it is widely accepted that the most abundant source of insulin that traverses the blood-brain barrier originates from the pancreas, recent evidence indicates that insulin may be 14 and insulin-like growth factors (IGFs) 15, 16 are synthesized in nasal epithelium or serous glands. Insulin produced in the nasal cavities enters the central nervous system (CNS) directly through the cribriform plate, and is transported along the olfactory nerves into the brain parenchyma, and principally ventromedial limbic structures 14. Alternatively, extra-CNS insulin traverses the blood-brain barrier via specific receptors to activate insulin-dependent functions and networks 14.

b) Insulin and Insulin Receptor Functions in the Brain

Within the past 8 to 10 years, the roles of insulin in the brain have undergone intense scrutiny both in humans and experimental models. Insulin is a pleotrophic hormone that has diverse functions in nearly every cell, including those of CNS origin. In the brain, insulin has critical roles in regulating neuronal functions such as growth, metabolism, plasticity, survival, and cholinergic function, which are needed for learning and memory 9, 1719. Moreover, increased levels of CNS insulin enhance learning and memory by improving hippocampal function 2022. Like insulin, leptin is another important regulator of food intake and metabolic processes such as lipid homeostasis, glucose utilization, and energy expenditure in the brain 23. Due to insulin's important roles in mediating neurocognitive function and energy balance, investigators focusing on the contributions of either peripheral insulin resistance or CNS insulin resistance have independently helped to shape the current body of literature and our present understanding of brain insulin resistance and its consequences.

c) Brain Insulin Resistance and Alzheimer's Disease

It is now well established that defects in insulin receptor signals correlate with dementia, particularly in AD 2, 3, 2426. In addition, although cerebrospinal fluid (CSF) insulin levels can be normal or even elevated at different points in disease 27, 28, evidence suggests that CSF insulin declines with AD progression 26, 29. Postmortem studies demonstrated that in AD, brain insulin resistance is associated with reduced receptor expression, binding, tyrosine phosphorylation, and activation of the intrinsic receptor tyrosine kinase 2, 3. Correspondingly, downstream signaling networks that promote neuronal survival, plasticity, growth, and cholinergic function are inhibited 2, 3, 7, 3033. Correspondingly, molecular, neuropathological, biochemical, and neurobehavioral abnormalities of the types seen in AD were produced in experimental animals by intracerebral administration of the pro-diabetes toxin, Streptozotocin 17, 34, 35, genetic depletion of brain insulin receptors or downstream effectors 36, 37, or molecular silencing of brain insulin and IGF receptors 38. The results of these studies indicate that brain insulin resistance is sufficient to cause brain metabolic dysfunction, cognitive impairment, and neurodegeneration. Furthermore, while it is still possible that brain insulin resistance represents and adaptive or protective response to hyperinsulinemia, it is unlikely that this concept has major relevance to the CNS abnormalities in MCI or AD because : 1) most individuals with AD and brain insulin resistance do not have hyperinsulinemia 2, 3; 2) experimental models of brain insulin resistance lead to neurodegeneration 17, 38, 39; and 3) administration of insulin improves cognitive function in the early stages of AD when brain insulin resistance already exists 13, 40, 41.

d) Experimental Models of Brain Insulin Resistance

Experimental data showed that siRNA silencing of the brain insulin receptor gene produces similar molecular and biochemical effects as occur in AD-type neurodegeneration 38, and that the administration of a pro-diabetes toxin leads to loss of brain insulin receptor functions and attendant deficits in spatial learning and memory 17. The pivotal role of impaired insulin receptor function in relation to neurodegeneration was confirmed by reversal of the AD phenotype following treatment with insulin sensitizer drugs 42. Another important point is that aging-associated declines in CNS insulin levels most likely contribute to cognitive impairment and neurodegeneration 43, as aging is the strongest risk factor for developing AD. Besides the direct consequences of brain metabolic dysfunction, insulin resistance reinforces the neurodegeneration cascade in exacerbating amyloid accumulation, neuronal loss, and synaptic disconnection 7. Altogether, the aggregate data support the hypothesis that Alzheimer's is a metabolic disorder in which brain insulin deficiency and resistance are at the core, and therefore shares molecular, biochemical, and pathophysiological features with diseases such as Type I and Type II diabetes mellitus 4, 6, 33, 44, 45. Consequently a role for insulin therapy is justified in correcting the metabolic dysfunction.

e) Brain Insulin Resistance in Obesity

The second group of investigators focused heavily on the importance of insulin signaling in the brain are those interested in regulation of energy balance, satiety, and feedback loops between the brain and gut. There is now convincing evidence that obesity is linked to brain insulin resistance, cognitive impairment, and neurodegeneration 5, 18. In normal physiological states, intranasal insulin exerts anorexic effects that negatively regulate food intake 46. However, insulin resistance adversely affects the ability to achieve satiety and the regulation of caloric consumption 47. Another factor possibly contributing to obesity pertains to the finding that insulin treatment lowers plasma and saliva cortisol levels in response to social stress 48. In states of insulin resistance, insulin is less effective in attenuating stress-induced activation of the hypothalamic-pituitary-adrenal (HPA) axis 47, which could lead to stress-related disruption of energy balance due to excessive calorie consumption 49, 50.

f) Potential contributions of Type 2 Diabetes and Hyperglycemia

Since the recognition that AD is associated with brain insulin resistance, a considerable body of literature has addressed the potential role of diabetes mellitus as a causal or co-factor in neurodegeneration. In a recent prospective longitudinal study, no significant relationship between peripheral glucose intolerance and insulin resistance could be demonstrated with respect to AD pathology 51. However, since the at risk subjects were treated for diabetes, the full impact of diabetes on neurodegeneration was not adequately assessed. To this end, another recent study demonstrated significantly increased risk of dementia among diabetics and hyperglycemia in general 52. Therefore, in the pre-diagnostic phase of peripheral insulin resistance and diabetes, the CNS may be vulnerable to the consequences of hyperglycemia and/or hyperinsulinemia, or sustain progressive injury and degeneration due to simultaneous development of brain insulin resistance. These concepts are supported by the findings of impaired brain glucose utilization and deficits in memory functions in individuals with sub-clinical diabetes mellitus 53. Furthermore, the finding that patients with AD+diabetes exhibited slower cognitive declines than those with AD alone 54 could be interpreted as evidence that systemic treatment for diabetes/peripheral insulin resistance is neuroprotective.

2. Intranasal Insulin Therapy

a) Therapeutic Approach Rationale

Insulin can be administered through subcutaneous, transdermal, oral, sub-lingual, buccal, rectal, vaginal, intramuscular, intraperitoneal, and intranasal routes 55. Intranasal delivery of insulin, as well as other compounds, is enabled by the direct neuroanatomical connections between the olfactory nerves and brain. This portal of access to the brain is advantageous because structural modifications of existing therapeutic compounds may not be required, particularly when they are small molecules. Another point is that drugs which could be used to treat neurological and psychiatric diseases may have limited therapeutic responses due to poor penetrance across the blood-brain barrier and therefore restricted ability to access the CNS. Intranasal therapy circumvents these problems while simultaneously reducing unwanted systemic responses. The discovery that nasal mucosa expresses and can deliver insulin to the brain suggests that nasal-to-brain transfer of insulin is physiological. Altogether, intranasal therapy provides a practical and noninvasive approach for treating cerebrovascular, behavioral, neoplastic, neurophysiologic, and diseases, and may also enable rapid and safe delivery of neuroprotective agents. Since normal neuroanatomical connections between the olfactory and limbic systems mediate memory, cognitive function, and behavior 56, intranasal therapy could be used to target neurodegenerative and psychiatric diseases.

b) Insulin Supports Memory and Cognitive Function

Alzheimer's disease (AD) brains exhibit progressive insulin resistance and declines in insulin levels with severity of neurodegeneration 2, 20. Neuro-imaging studies linked in vivo brain insulin resistance to neurodegeneration by demonstrating significant perturbations in insulin regulated metabolism, and correlations between increased brain insulin resistance and reduced cerebral volume, impaired responses to insulin, and hippocampal atrophy in AD, Type 2 diabetes, and obesity 13. Increasing brain insulin levels improves verbal and hippocampal declarative memory 12, 57, and in AD, insulin improves cognitive function and slows cognitive decline 8, 44. Together, these observations support the concept of insulin-based therapy for AD 58, including prevention and treatment 13.

Intranasal insulin therapy provides an excellent means of supplying the brain with insulin to overcome deficits in production, transport, and responsiveness, reproducing the effects of systemic insulin but with avoidance of adverse effects. Intranasal insulin improves cognitive function with respect to attention, and verbal and hippocampal declarative memory 12, 57, 59. Clinical trials data have shown that in subjects with MCI or AD, intranasal insulin improves memory and metabolic function 13. However, sub-group analysis revealed that following intranasal insulin administration, subjects who had an apolipoprotein E-ε4 (APOE-ε4) negative genotype exhibited significant improvements in verbal memory, whereas those who were APOE–ε4 positive had poor responses on recall tasks 60, 61. The therapeutic efficacy of intranasal insulin has now been confirmed by a number of independent studies 26. The consistent findings were that, among individuals who had brain insulin resistance associated with MCI, early AD, or Type 2 diabetes mellitus, significant improvements in cognition, memory, attention and metabolic function occurred within a few months of treatment in the clinical trials 26, 41, 62, 63. Mechanistically, in addition to the stimulatory effects on energy metabolism, insulin increases amyloid clearance from the brain 64 and it supports neuronal cytoskeletal and cholinergic functions 6, 9, 11. Furthermore, enhanced insulin signaling inhibits the activity of kinases that promote tau hyperphosphorylation, and activates pathways that mediate synapse formation and neuronal plasticity 65. Although increasing availability of insulin is expected to restore insulin's actions in the brain, the insulin resistant neurons and glial cells may not be responsive. Instead, the connecting neurons that are insulin responsive but dependent upon activity-dependent connections from insulin resistant cells might be preserved. In addition, as in type 2 diabetes, providing higher levels of insulin to insulin resistant cells could also be an important means of maintaining function.

c) Intranasal Insulin and Reversal of Obesity

Overall, CNS energy balance is mediated by hypothalamic neuropeptide pathways that enable peripheral adipose tissue to signal body fat content information to the brain via insulin and leptin 47, 66, 67 These processes are distinct from those involved in hippocampal function, verbal memory, and attention 8, 68. Besides increasing cognitive performance, chronic intranasal insulin administration reduces adiposity in normal individuals 69. Functional magnetic resonance imaging studies showed that in healthy individuals, intranasal insulin reduces brain activity in response to food picture stimuli 70, whereas in obese subjects, insulin-stimulated visual evoked (food modulated) responses are reduced 71. In another study of obese individuals, insulin was observed to improve memory without significantly affecting body fat 72. In addition, intranasal insulin was demonstrated to alter activity in the hypothalamus, which regulates homeostatic and reward-related functions 73. These findings suggest that brain insulin resistance can differentially affect cognitive and metabolic signaling pathways.

Further studies demonstrated gender-specific differences in response to intranasal insulin. Men were found to be more sensitive than women to appetite/anorexogenic and metabolic effects of intranasal insulin69, 74, while both pre- and postmenopausal women were found to be more sensitive to insulin stimulation of hippocampus-dependent memory functions 74, 75. In a recent all-women study, insulin responsiveness to body mass index was demonstrated to be localized in the prefrontal and anterior cingulate cortex 76. These observations suggest that insulin regulates food intake by modifying reward activity in the brain, and that insulin resistance contributes to obesity by short-circuiting this pathway. This suggests that, under normal circumstances, insulin promotes satiety and thereby helps regulate food intake.

d) Intranasal Insulin for Treatment in Individuals with Diabetes Mellitus

The main emphasis of this review is to consider the advantages and complications of intranasal insulin therapy for treating mild cognitive impairment and AD. However, it is important to recognize that the same approach is utilized to treat systemic insulin resistance and deficiency, raising questions about specificity and potential off-target effects of therapy designed to treat neurodegeneration. One limitation of using intranasal insulin to treat diabetes mellitus is that sufficient peripheral blood levels must be achieved to restore glucose homeostasis. On the other hand, in the setting of diabetes mellitus, intranasal insulin could be used to prevent secondary effects of disease, including cognitive impairment. For example, in a murine model of type I diabetes produced by Streptozotocin administration, progressive cognitive impairment and white matter atrophy were correlated with brain insulin resistance. Treatment with intranasal insulin prevented the brain morphological abnormalities, white matter loss, and impairments in signaling through survival and synaptic plasticity pathways 77. In another animal model that more closely mimics human disease, mucosal administration of autoantigens prevented type 1 diabetes 78. However, attempts to use intranasal insulin to prevent type I diabetes shortly after detection of autoantibodies proved unsuccessful 79.

e) Intranasal Insulin-Like Growth Factor, Type 1 (IGF-1) Treatment

Insulin and IGF-1 activate very similar signaling pathways, utilizing their own receptors, and achieving distinct but related outcomes. Like insulin, IGF-1 and IGF-1 receptors are abundantly expressed in the brain 9. IGF-1 signaling mediates neuronal growth, migration, regeneration, and repair 9. In an experimental model of spinal cerebellar atrophy, intranasal IGF-I therapy helped improve motor performance and partial recovery of Purkinje cell protein expression and function 80. In another study, investigators used intranasal IGF-I to treat middle cerebral artery occlusion models of stroke. The findings were significant reductions in infarct volumes even when administered between two or four hours after vascular occlusion. On a histologic level, the reduction in infarct volume is associated with decreased apoptosis and tissue injury and functionally there was significantly greater recovery of motor and sensory systems compared with vehicle 81. Subsequent studies demonstrated effectiveness of IGF-I for reducing infarct volume or hypoxic – ischemic injury in the brain 82, 83. In addition the neuroprotective actions of IGF-I and its ability to support neurogenesis suggests a potential therapeutic application in human stroke 83.

3. Strategies for Improving the Intranasal Insulin Therapy

a) Overview of Advantages and Challenges

Intranasal insulin administration results in direct transport of insulin from the nasal cavity to the CNS via intra-neuronal and extra-neuronal pathways that bypass the blood-brain barrier and minimize systemic side effects 12. Therefore, a major advantage of intranasal delivery is that it offers the potential for unrestricted transfer of therapeutic compounds to the brain or CSF, enabling higher local levels to be achieved for disease-specific targeting 84. For example, intranasal insulin administration increases CSF levels of insulin 58, 70, is safe in humans 58, 85, and avoids repeated injections. Nonetheless, challenges exist with respect to the eventual broadened use of intranasal therapy. The major ones include: 1) inaccurate delivery and dosing by older adults; 2) the need to improve drug stabilization and time- and dose-regulated release; 3) transport and penetration across the nasal mucosa; 4) nasal transport “resistance”; and 5) CNS versus systemic targeting.

b) Challenges pertaining to accuracy of delivery and dosing

Delivery of intranasal insulin can prove challenging for older adults. One concern is that the dosing may be inaccurate due to failure to deliver the insulin to the olfactory system and cerebrospinal fluid rather than the respiratory or gastrointestinal tract. Therefore, there is a need to optimize intranasal insulin delivery systems to produce more consistent therapeutic effects. To this end, preclinical and human studies examined the effects of incorporating insulin into hydroxypropyl gel and installing it into the nasal cavity with a bio-adhesive compound. This approach proved successful for the release of insulin but was associated with sharp declines in blood sugar 86. Continued modification of related technologies designed to achieve consistent and predictable effects on hyperglycemia holds promise for the treatment of diabetes mellitus. However, the studies did not provide data on CNS delivery of insulin.

c) Drug Stability and Regulated Release

Challenges pertaining to stability and time course of drug release are being addressed through the use of polysaccharide nanoparticles and hybrid polysaccharide/oligosaccharide nanoparticles as carriers for macromolecules. For example, insulin-loaded chitosan (CS) and cyclodextrin (CD) derivative-based nanoparticles were shown to effectively reduce plasma glucose levels 87. Another approach includes the use of oligo-arginine–linked polymer backbones to insulin to enhance penetration across the nasal mucosa and more effectively achieve therapeutic reductions in plasma glucose 88. The use of lyophilized nasal insert formulations can extend nasal residence and thereby enable prolonged delivery and avoid the need for repeated use of nasal spray solutions 89.

d) Transport and Penetration Across Nasal Mucosa

An important challenge posed by intranasal delivery of insulin is that insulin is not well absorbed through the nasal mucosa due to its large size, hydrophilic nature, and low permeability across membranes. This problem exists for macromolecular therapeutics in general 90. One approach has been to use nanoparticles 91, but research along these lines has not yet reached fruition. Another approach involves the administration of tetradecyl-beta-D-maltoside (TDM), which has been shown experimentally to improve recovery of nasal permeability barrier following repeated intranasal administrations of peptides 92. Further research and development is needed to: 1) improve intranasal absorption of compounds; 2) optimize bio-adhesive delivery systems for sustained and controlled drug delivery; and 3) expand the use of water soluble powders to extend shelf-life and transportability of compounds intended for intranasal therapy 93.

e) Nasal Transport “Resistance”

Another challenge confronting the chronic use of intranasal insulin and other drug deliveries is the development of time-dependent reductions in nasal permeability. This form of nasal transport resistance is caused by alterations in nasal morphology due to loss of cell-to-cell junctions 92. Therefore, specific strategies are needed to facilitate transport of various compounds across the nasal epithelium, and thereby maintain the effectiveness of intranasal therapy in chronic use settings. To this end, investigators examined the effectiveness of cyclopentadecalactone (CPE-215), a compound that enhances absorption of molecules across mucous membranes, for intranasal insulin delivery (Nasulin, CPEX Pharmaceuticals). In a clinical trial, the investigators demonstrated that a nasal spray containing recombinant human insulin with CPE-215 could effectively lower blood glucose in diabetics, such that the dose-response pharmacodynamics were proportional and linear 94, 95.

f) CNS Versus Systemic Targeting-Resolving Therapeutic Objectives

Confusion over the therapeutic applications of intranasal insulin exists because the objectives differ among groups. Investigators fall into two dominant groups: those intending to treat diabetes mellitus by producing consistent therapeutic levels of plasma insulin, and those interested in restoring insulin responsiveness in states of brain insulin resistance/deficiency associated with MCI, AD, or obesity 96. For example, in a clinical study of healthy individuals, delivery of intranasal insulin using CPE–215 technology produced peak plasma levels of insulin 10 to 20 min. later, and persistence of elevated plasma insulin for approximately one hour. Correspondingly, plasma glucose levels declined in an insulin dose-dependent manner, but self-corrected within 1.5 to 2 hours of treatment 97. The results of similarly focused studies advocate the use of intranasal delivery systems to control hyperglycemia safely.

In contrast, in a separate publication, the investigators emphasized insulin-stimulated alterations in hypothalamic activity that regulates homeostatic and reward-related functions, and pointed out that changes in peripheral insulin resistance were short-term (30–120 min later) following intranasal administration 73. Furthermore, intranasal insulin was deemed attractive for treating aging populations with cognitive impairment or early AD because insulin is directed to the brain and CSF and has minimal systemic absorption. Moreover, clinical studies have shown that intranasal insulin improves memory 98. In essence, at the core of the problem is the fact that using essentially the same tools, the brain-focused studies boast of the robust CNS and limited systemic responses, while the diabetes-focused studies omit data about CNS effects. Going forward, more attention should be paid to the pharmacodynamics of intranasal therapy and perhaps the approaches could be tailored to favor CNS or systemic delivery. One example of success along these lines pertains to the use of the rapidly acting insulin analog, insulin Aspart, which when administered intranasal, enhances memory without altering plasma insulin and glucose levels 99. However, further research is needed to determine the degree to which different formulations of insulin designed for intranasal delivery are free of systemic actions.

4. Adverse Effects

Intranasal insulin therapy can produce unwanted systemic effects including hypoglycemia and elevated blood pressure. In one clinical study, repeated intranasal administration of insulin, every 10 minutes for 2 hours, caused immediate increases in diastolic, systolic, and mean arterial blood pressure 100, which, in the acute setting, could be concerning for individuals with hypertension. However, since these elevated blood pressure responses are not prolonged, chronic intranasal insulin administration should not be regarded as unsafe 100. Another potential adverse effect of intranasal insulin is excessive delivery with production of high local CNS levels. Since high levels of insulin can be injurious and promote tissue degeneration 101105, strategies must be taken to ensure that the dosing rates and levels are optimized (see Expert Opinion below). Finally, as insulin has growth promoting properties, prolonged use may “encourage” proliferation of neoplastic cells 106.

5. Alternative Approaches

Alternative approaches previously considered for treating or preventing AD include the use of insulin sensitizer such as thiazolidinediones and sulfonylurea. Thiazolidinediones function by enhancing insulin sensitivity whereas sulfonylurea increases peripheral insulin concentrations 98. In a retrospective case-control study, no significant alterations in risk for developing AD were associated with long-term treatment of diabetes mellitus with sulfonylureas, thiazolidinediones, or insulin 107. However, the data are difficult to interpret because of the non-random approaches to treatment and different cross-sectional outcomes of diabetes.

Initial small-scale, short-term Phase II clinical trials suggested that treatment with rosiglitazone, a gamma peroxisome-proliferator activated receptor (PPAR) agonist, could effectively delay progression of cognitive impairment by preserving attention and memory functions and maintaining amyloid-beta clearance rates in subjects with MCI or early AD 108. Others found similar results but the benefits were mainly observed in individuals who had APOE-E4-negative genotypes 109.110. Similarly, the efficacy of rosiglitazone for treating AD-type neurodegeneration was also demonstrated in standard mouse models 111, 112. However, in Phase III placebo-controlled, randomized-clinical trials that were stratified with respect to APOE-genotype, no significant long-term benefit was observed for rosiglitazone monotherapy versus placebo 113, 114, or when combined with an acetylcholine esterase inhibitor 115. Small scale studies with pioglitazone, another gamma PPAR agonist drug, have also not produced convincing therapeutic responses 116, 117.

Results from the most rigorously conducted clinical and pre-clinical studies suggest that insulin sensitizer drugs, including PPAR agonists, produce no significant long-term therapeutic effects and do not halt progression of dementia in mild or moderate AD. Yet, in the short-term studies or early phases of long-term studies, positive effects of rosiglitazone were measured. In addition, in an experimental model of AD produced by treatment with a pro-diabetes toxin, both delta and gamma PPAR agonists were effective in preserving learning, memory, and hippocampal structure 42. In light of the full discussion included in this work, particularly the need to better target CNS insulin resistance by intranasal delivery, future drug designs should entertain the concept that brain insulin resistance disease states might be effectively treated with CNS-specific PPAR agonists or other classes of insulin sensitizers delivered via the intranasal route.

6. Conclusions

Intranasal insulin therapy holds promise for targeting brain insulin resistance associated with cognitive impairment and neurodegeneration, including AD, due to its ability to enhance and preserve memory and relatively high safety index. Since intranasal insulin also can be used to treat systemic insulin resistance diseases, mechanisms to ensure CNS versus systemic delivery/transfer are needed. Compounds other than insulin may be delivered via the intranasal route to treat a range of CNS diseases Technical barriers pertaining to stability, rate of delivery, and transport across nasal epithelium must be overcome to optimize intranasal insulin therapy for chronic diseases

7. Expert Opinion

While intranasal therapy is a very promising approach for the treatment of CNS diseases due to better targeting and ability to by-pass the blood-brain barrier, the methods and reagents employed from study to study are not identical. Standardized approaches that preferentially result in delivery of compounds to the CNS rather than periphery, and vice versa are needed. As the field moves forward, consideration should be given to developing compounds that regulate the rate, timing, and levels of insulin release, perhaps through the use of nanotechnology. The need to refine the parameters of intranasal insulin delivery is particularly relevant to its long-term use as the therapeutic efficacy may vary with the patient's age, stage of cognitive impairment, and time span of treatment. Elderly patients may require tight regulation of insulin release to prevent surges and attendant side effects. At the same time, mechanisms to provide as-needed pulsatile delivery of insulin could be advantageous for accommodating the so-called `bad days' of dementia and helping to quickly restore mental clarity, particularly when important decisions are needed. With long-term use of intranasal insulin, adjustments to dosing and timing will be needed. Therefore, delivery agents should be improved to accommodate changes in treatment requirements.

There is a need to monitor the efficiency and distribution of CNS insulin and other drugs delivered via the intranasal route. In particular, more sensitive approaches for in vivo imaging of compounds, their metabolites, and effects should be developed. Currently, studies designed to examine the effectiveness of intranasal therapy rely in neurocognitive testing or neuro-imaging techniques such as fMRI or PET scanning. However, besides evaluating the consequences of intranasal insulin delivery, more information is needed about CNS localizations of the compounds, their half-lives in different brain regions, and the changes in those parameters over time. Since dementia is not a static process and over time, neurodegeneration worsens and gradually affects a broader range of brain structures, determining if the insulin or other treatments reach the additional targets will be important for evaluating efficacy. Discordances between delivery and metabolism would raise concerns about brain region-specific emergence of insulin resistance which would have to be overcome, possibly by alternative therapeutic measures.

As it is evident that neurodegeneration, including Alzheimer's disease, is mediated by a multi-step and multi-pronged cascade, most likely effective treatment will require more than a single agent and will likely change with disease progression. Therefore, the scope of therapeutic compounds that could be delivered effectively to the brain via the intranasal route should be broadened. Besides improving means of measuring uptake and delivery to the brain as discussed above, further research should determine if the administration of more than a single compound is feasible and the extents to which interferences or facilitated deliveries occur. In addition, the development of in vivo imaging approaches to assess pharmacokinetics of multiple compounds will be needed. Similar approaches could be utilized for CNS targeted treatment of primary brain neoplasms, epilepsy, and infarcts (strokes).

As with many receptor-mediated therapeutic agents, over time, cellular adaptations can lead to relative resistance and reduced efficacy of the compounds. Long-term studies are needed to assess tolerance to intranasal insulin therapy, and further the understand mechanisms for over-coming this problem, as well as predicting host susceptibility to this adaptive response. Alternatively, receptor resistance could reflect disease progression. The ability to distinguish adaptive from disease related receptor resistance is important because the corrective measures could differ. For adaptive receptor resistance, efforts should be made to develop approaches to “refresh” receptor or prevent their “exhaustion” by devising other means of drug delivery, perhaps via alternate receptors or receptor-independent mechanisms. For disease-mediated receptor resistance, which could be caused by deterioration of downstream signal transduction pathways, alternative therapeutic measures would have to be implemented to overcome the fundamental problems caused by impaired insulin signaling. For examples, effective insulin sensitizer agents that function at the level of the nucleus can drive metabolic functions, yet bypass the need for upstream activation of insulin receptors.

Since Alzheimer's and other neurodegenerative diseases are progressive, their pathologic and pathophysiologic features change over time, producing additional potential targets for therapy. As is the case for most malignancies, which are seldom cured with a single drug, with the advancement of neurodegeneration, multi-pronged therapeutic approaches will be needed to extend the benefits of intranasal insulin. The types of treatments that are needed are directly related to the consequences of insulin resistance, e.g. oxidative and endoplasmic reticulum stress, metabolic dysfunction, neuro-inflammation, impaired neuronal plasticity, and cell death. With regard to neuronal plasticity, it should be possible to supply neurotrophins via the intranasal route, but their efficacy for treating cognitive impairment and neurodegeneration requires further study. Intranasal delivery of peptides to the brain is preferred because of their high likelihood of degrading after oral administration, and inefficiently entering the CNS after parenteral administration. In contrast, treatment of other aspects of neurodegeneration may be accomplished by administering the drugs though other routes, particularly if they have not yet been adapted for intranasal delivery. The challenge will be to identify which classes of additional and alternative therapeutic compounds are needed and once administered, determining their specific effectiveness. This concept rests on developing newer approaches for assessing pathologic consequences of brain insulin deficiency and resistance that mediate progressive neurodegeneration.

Article highlights.

  • In the normal brain, insulin signaling has divers and critical functions including neuronal survival, metabolism, and plasticity.

  • Brain insulin resistance mediates cognitive impairment in neurodegeneration, including Alzheimer's disease, as well as in obesity and diabetes.

  • Intranasal insulin is as effective as systemic insulin treatment, but avoids the need for repeated injections, and with respect to the brain, the approach can lead to preferential targeting of CNS diseases with minimal unwanted systemic responses.

  • Intranasal insulin has proven effects in slowing progression of cognitive impairment and Alzheimer's disease.

  • Intranasal insulin has positive therapeutic effects on brain functions pertaining to food intake and satiety, and therefore could be optimized for use in treatment programs for obesity.

  • Alternative approaches for treating CNS insulin resistance, including the use of insulin sensitizers, were met with initial positive results in short-term studies, but failed to show significant benefits in long-term studies. Although this approach has fallen out of favor, the problems encountered in later clinical trials could have been due to inadequate brain targeting with attendant systemic side-effects, and use of selected compounds that were not brain-specific.

Acknowledgments

Supported by AA-11431 and AA-12908 from the National Institutes of Health

References

  • 1.Hallschmid M, Schultes B. Central nervous insulin resistance: a promising target in the treatment of metabolic and cognitive disorders? Diabetologia. 2009 Nov;52:2264–9. doi: 10.1007/s00125-009-1501-x. [DOI] [PubMed] [Google Scholar]
  • 2.Rivera EJ, Goldin A, Fulmer N, et al. Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer's disease: link to brain reductions in acetylcholine. Journal of Alzheimer's disease : JAD. 2005 Dec;8:247–68. doi: 10.3233/jad-2005-8304. [DOI] [PubMed] [Google Scholar]
  • 3.Steen E, Terry BM, Rivera EJ, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease--is this type 3 diabetes? Journal of Alzheimer's disease : JAD. 2005 Feb;7:63–80. doi: 10.3233/jad-2005-7107. [DOI] [PubMed] [Google Scholar]; ** First demonstration of brain insulin and IGF resistance in humans using molecular approaches to characterize deficits in insulin signaling in postmortem brain tissue from proven cases of Alzheimer's disease.
  • 4.de la Monte SM. Insulin resistance and Alzheimer's disease. BMB Rep. 2009 Aug 31;42:475–81. doi: 10.5483/bmbrep.2009.42.8.475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.de la Monte SM, Longato L, Tong M, et al. Insulin resistance and neurodegeneration: roles of obesity, type 2 diabetes mellitus and non-alcoholic steatohepatitis. Curr Opin Investig Drugs. 2009 Oct;10:1049–60. [PMC free article] [PubMed] [Google Scholar]
  • 6.de la Monte SM, Tong M, Wands JR. Insulin resistance, cognitive impairment, and neurodegeneration: roles of nitrosamine exposure, diet, and lifestyles. In: Monte DSDL, editor. Alzheimer's Disease Pathogenesis: Core Concepts, Shifting Paradigms and Therapeutic Targets: InTech. 2011. pp. 1–39. [Google Scholar]
  • 7.de la Monte SM. Contributions of brain insulin resistance and deficiency in amyloid-related neurodegeneration in Alzheimer's disease. Drugs. 2012 Jan 1;72:49–66. doi: 10.2165/11597760-000000000-00000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Derakhshan F, Toth C. Insulin and the brain. Current diabetes reviews. 2013 Mar 1;9:102–16. [PubMed] [Google Scholar]
  • 9.de la Monte SM, Wands JR. Review of insulin and insulin-like growth factor expression, signaling, and malfunction in the central nervous system: relevance to Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2005 Feb;7:45–61. doi: 10.3233/jad-2005-7106. [DOI] [PubMed] [Google Scholar]
  • 10.Xu J, Yeon JE, Chang H, et al. Ethanol impairs insulin-stimulated neuronal survival in the developing brain: role of PTEN phosphatase. J Biol Chem. 2003 Jul 18;278:26929–37. doi: 10.1074/jbc.M300401200. [DOI] [PubMed] [Google Scholar]
  • 11.Soscia SJ, Tong M, Xu XJ, et al. Chronic gestational exposure to ethanol causes insulin and IGF resistance and impairs acetylcholine homeostasis in the brain. Cell Mol Life Sci. 2006 Sep;63:2039–56. doi: 10.1007/s00018-006-6208-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Benedict C, Hallschmid M, Schultes B, et al. Intranasal insulin to improve memory function in humans. Neuroendocrinology. 2007;86:136–42. doi: 10.1159/000106378. [DOI] [PubMed] [Google Scholar]
  • 13.Freiherr J, Hallschmid M, Frey WH, 2nd, et al. Intranasal insulin as a treatment for Alzheimer's disease: a review of basic research and clinical evidence. CNS drugs. 2013 Jul;27:505–14. doi: 10.1007/s40263-013-0076-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Henkin RI. Intranasal insulin: from nose to brain. Nutrition. 2010 Jun;26:624–33. doi: 10.1016/j.nut.2009.08.003. [DOI] [PubMed] [Google Scholar]
  • 15.Suzuki Y, Takeda M. Expression of insulin-like growth factor family in the rat olfactory epithelium. Anatomy and embryology. 2002 Oct;205:401–5. doi: 10.1007/s00429-002-0266-5. [DOI] [PubMed] [Google Scholar]
  • 16.Ferguson MW, Sharpe PM, Thomas BL, et al. Differential expression of insulin-like growth factors I and II (IGF I and II), mRNA, peptide and binding protein 1 during mouse palate development: comparison with TGF beta peptide distribution. Journal of anatomy. 1992 Oct;181(Pt 2):219–38. [PMC free article] [PubMed] [Google Scholar]
  • 17.Lester-Coll N, Rivera EJ, Soscia SJ, et al. Intracerebral streptozotocin model of type 3 diabetes: relevance to sporadic Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2006 Mar;9:13–33. doi: 10.3233/jad-2006-9102. [DOI] [PubMed] [Google Scholar]
  • 18.Moroz N, Tong M, Longato L, et al. Limited Alzheimer-type neurodegeneration in experimental obesity and type 2 diabetes mellitus. Journal of Alzheimer's disease : JAD. 2008 Sep;15:29–44. doi: 10.3233/jad-2008-15103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.de la Monte SM, Wands JR. Insulin resistance and neurodegeneration: Type 2 versus Type 3 diabetes mellitus. Emerging Drugs and Targets for AD. 2009;1:235–54. [Google Scholar]
  • 20.Talbot K, Wang HY, Kazi H, et al. Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. The Journal of clinical investigation. 2012 Apr 2;122:1316–38. doi: 10.1172/JCI59903. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This article confirms the findings by Steen, et al, but takes the work to an important new height by demonstrating impairments in insulin signaling through the use of ex vivo cultures of fresh frozen human postmortem brain tissue.
  • 21.Sun X, Yao H, Douglas RM, et al. Insulin/PI3K signaling protects dentate neurons from oxygen-glucose deprivation in organotypic slice cultures. J Neurochem. 2010 Jan;112:377–88. doi: 10.1111/j.1471-4159.2009.06450.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.McClean PL, Gault VA, Harriott P, et al. Glucagon-like peptide-1 analogues enhance synaptic plasticity in the brain: a link between diabetes and Alzheimer's disease. European journal of pharmacology. 2010 Mar 25;630:158–62. doi: 10.1016/j.ejphar.2009.12.023. [DOI] [PubMed] [Google Scholar]
  • 23.Scherer T, Lehnert H, Hallschmid M. Brain insulin and leptin signaling in metabolic control: from animal research to clinical application. Endocrinology and metabolism clinics of North America. 2013 Mar;42:109–25. doi: 10.1016/j.ecl.2012.11.002. [DOI] [PubMed] [Google Scholar]
  • 24.Wada A, Yokoo H, Yanagita T, et al. New twist on neuronal insulin receptor signaling in health, disease, and therapeutics. Journal of pharmacological sciences. 2005 Oct;99:128–43. doi: 10.1254/jphs.crj05006x. [DOI] [PubMed] [Google Scholar]
  • 25.Ketterer C, Tschritter O, Preissl H, et al. Insulin sensitivity of the human brain. Diabetes research and clinical practice. 2011 Aug;93(Suppl 1):S47–51. doi: 10.1016/S0168-8227(11)70013-4. [DOI] [PubMed] [Google Scholar]
  • 26.Schioth HB, Frey WH, Brooks SJ, et al. Insulin to treat Alzheimer's disease: just follow your nose? Expert review of clinical pharmacology. 2012 Jan;5:17–20. doi: 10.1586/ecp.11.70. [DOI] [PubMed] [Google Scholar]
  • 27.Molina JA, Jimenez-Jimenez FJ, Vargas C, et al. Cerebrospinal fluid levels of insulin in patients with Alzheimer's disease. Acta Neurol Scand. 2002 Dec;106:347–50. doi: 10.1034/j.1600-0404.2002.01326.x. [DOI] [PubMed] [Google Scholar]
  • 28.Fujisawa Y, Sasaki K, Akiyama K. Increased insulin levels after OGTT load in peripheral blood and cerebrospinal fluid of patients with dementia of Alzheimer type. Biol Psychiatry. 1991 Dec 15;30:1219–28. doi: 10.1016/0006-3223(91)90158-i. [DOI] [PubMed] [Google Scholar]
  • 29.Craft S, Peskind E, Schwartz MW, et al. Cerebrospinal fluid and plasma insulin levels in Alzheimer's disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology. 1998 Jan;50:164–8. doi: 10.1212/wnl.50.1.164. [DOI] [PubMed] [Google Scholar]
  • 30.de la Monte SM. Triangulated Mal-Signaling in Alzheimer's Disease: Roles of Neurotoxic Ceramides, ER Stress, and Insulin Resistance Reviewed. Journal of Alzheimer's disease : JAD. 2012 Jan 1;30:S231–49. doi: 10.3233/JAD-2012-111727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.de la Monte SM. Brain insulin resistance and deficiency as therapeutic targets in Alzheimer's disease. Curr Alzheimer Res. 2012 Jan;9:35–66. doi: 10.2174/156720512799015037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.de la Monte SM. Therapeutic targets of brain insulin resistance in sporadic Alzheimer's disease. Front Biosci (Elite Ed) 2012;4:1582–605. doi: 10.2741/482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.de la Monte SM. Metabolic derangements mediate cognitive impairment and Alzheimer's disease: Role of peripheral insulin resistance diseases. Panminerva Medica. 2012;54:171–8. [PMC free article] [PubMed] [Google Scholar]
  • 34.Hoyer S, Muller D, Plaschke K. Desensitization of brain insulin receptor. Effect on glucose/energy and related metabolism. J Neural Transm Suppl. 1994;44:259–68. doi: 10.1007/978-3-7091-9350-1_20. [DOI] [PubMed] [Google Scholar]
  • 35.Hoyer S, Lannert H, Noldner M, et al. Damaged neuronal energy metabolism and behavior are improved by Ginkgo biloba extract (EGb 761) J Neural Transm. 1999;106:1171–88. doi: 10.1007/s007020050232. [DOI] [PubMed] [Google Scholar]
  • 36.Schubert M, Brazil DP, Burks DJ, et al. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2003 Aug 6;23:7084–92. doi: 10.1523/JNEUROSCI.23-18-07084.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schubert M, Gautam D, Surjo D, et al. Role for neuronal insulin resistance in neurodegenerative diseases. Proceedings of the National Academy of Sciences of the United States of America. 2004 Mar 2;101:3100–5. doi: 10.1073/pnas.0308724101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.de la Monte SM, Tong M, Bowling N, et al. si-RNA inhibition of brain insulin or insulin-like growth factor receptors causes developmental cerebellar abnormalities: relevance to fetal alcohol spectrum disorder. Mol Brain. 2011;4:13. doi: 10.1186/1756-6606-4-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hoyer S. The brain insulin signal transduction system and sporadic (type II) Alzheimer disease: an update. J Neural Transm. 2002 Mar;109:341–60. doi: 10.1007/s007020200028. [DOI] [PubMed] [Google Scholar]
  • 40.Bomfim TR, Forny-Germano L, Sathler LB, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer's disease- associated Abeta oligomers. The Journal of clinical investigation. 2012 Apr 2;122:1339–53. doi: 10.1172/JCI57256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Craft S, Baker LD, Montine TJ, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Archives of neurology. 2012 Jan;69:29–38. doi: 10.1001/archneurol.2011.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.de la Monte SM, Tong M, Lester-Coll N, et al. Therapeutic rescue of neurodegeneration in experimental type 3 diabetes: relevance to Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2006 Sep;10:89–109. doi: 10.3233/jad-2006-10113. [DOI] [PubMed] [Google Scholar]
  • 43.Laron Z. Insulin and the brain. Archives of physiology and biochemistry. 2009 May;115:112–6. doi: 10.1080/13813450902949012. [DOI] [PubMed] [Google Scholar]
  • 44.Morris JK, Burns JM. Insulin: an emerging treatment for Alzheimer's disease dementia? Current neurology and neuroscience reports. 2012 Oct;12:520–7. doi: 10.1007/s11910-012-0297-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.de la Monte SM, Wands JR. Alzheimer's disease is type 3 diabetes: Evidence reviewed. J Diabetes Sci Tech. 2008;2:1101–13. doi: 10.1177/193229680800200619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Jauch-Chara K, Friedrich A, Rezmer M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes. 2012 Sep;61:2261–8. doi: 10.2337/db12-0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Posey KA, Clegg DJ, Printz RL, et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. American journal of physiology Endocrinology and metabolism. 2009 May;296:E1003–12. doi: 10.1152/ajpendo.90377.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bohringer A, Schwabe L, Richter S, et al. Intranasal insulin attenuates the hypothalamic-pituitary-adrenal axis response to psychosocial stress. Psychoneuroendocrinology. 2008 Nov;33:1394–400. doi: 10.1016/j.psyneuen.2008.08.002. [DOI] [PubMed] [Google Scholar]
  • 49.Bale TL, Anderson KR, Roberts AJ, et al. Corticotropin-releasing factor receptor-2-deficient mice display abnormal homeostatic responses to challenges of increased dietary fat and cold. Endocrinology. 2003 Jun;144:2580–7. doi: 10.1210/en.2002-0091. [DOI] [PubMed] [Google Scholar]
  • 50.Kyrou I, Chrousos GP, Tsigos C. Stress, visceral obesity, and metabolic complications. Annals of the New York Academy of Sciences. 2006 Nov;1083:77–110. doi: 10.1196/annals.1367.008. [DOI] [PubMed] [Google Scholar]
  • 51.Thambisetty M, Metter EJ, Yang A, et al. Glucose Intolerance, Insulin Resistance, and Pathological Features of Alzheimer Disease in the Baltimore Longitudinal Study of Aging. JAMA neurology. 2013 Jul 29; doi: 10.1001/jamaneurol.2013.284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Crane PK, Walker R, Hubbard RA, et al. Glucose levels and risk of dementia. The New England journal of medicine. 2013 Aug 8;369:540–8. doi: 10.1056/NEJMoa1215740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Baker LD, Cross DJ, Minoshima S, et al. Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes. Archives of neurology. 2011 Jan;68:51–7. doi: 10.1001/archneurol.2010.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dominguez RO, Marschoff ER, Gonzalez SE, et al. Type 2 diabetes and/or its treatment leads to less cognitive impairment in Alzheimer's disease patients. Diabetes research and clinical practice. 2012 Oct;98:68–74. doi: 10.1016/j.diabres.2012.05.013. [DOI] [PubMed] [Google Scholar]
  • 55.Henkin RI. Inhaled insulin-intrapulmonary, intranasal, and other routes of administration: mechanisms of action. Nutrition. 2010 Jan;26:33–9. doi: 10.1016/j.nut.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • 56.Hanson LR, Frey WH., 2nd Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC neuroscience. 2008;9(Suppl 3):S5. doi: 10.1186/1471-2202-9-S3-S5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Benedict C, Frey WH, 2nd, Schioth HB, et al. Intranasal insulin as a therapeutic option in the treatment of cognitive impairments. Experimental gerontology. 2011 Feb-Mar;46:112–5. doi: 10.1016/j.exger.2010.08.026. [DOI] [PubMed] [Google Scholar]
  • 58.Chapman CD, Frey WH, 2nd, Craft S, et al. Intranasal Treatment of Central Nervous System Dysfunction in Humans. Pharmaceutical research. 2012 Nov 8; doi: 10.1007/s11095-012-0915-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Messier C, Teutenberg K. The role of insulin, insulin growth factor, and insulin-degrading enzyme in brain aging and Alzheimer's disease. Neural plasticity. 2005;12:311–28. doi: 10.1155/NP.2005.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Reger MA, Watson GS, Frey WH, 2nd, et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiology of aging. 2006 Mar;27:451–8. doi: 10.1016/j.neurobiolaging.2005.03.016. [DOI] [PubMed] [Google Scholar]; ** This is one of several articles from this group in which the authors demonstrate the utility of intranasal insulin for treating cognitive impairment and Alzheimer's disease. The work also illustrates that particular subtypes of AD are not equally sensitive to intranasal insulin, justifying the need to catalogue AD subtypes and risk factors.
  • 61.Reger MA, Watson GS, Green PS, et al. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. Journal of Alzheimer's disease : JAD. 2008 Apr;13:323–31. doi: 10.3233/jad-2008-13309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ott V, Benedict C, Schultes B, et al. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes, obesity & metabolism. 2012 Mar;14:214–21. doi: 10.1111/j.1463-1326.2011.01490.x. [DOI] [PubMed] [Google Scholar]
  • 63.Shemesh E, Rudich A, Harman-Boehm I, et al. Effect of intranasal insulin on cognitive function: a systematic review. The Journal of clinical endocrinology and metabolism. 2012 Feb;97:366–76. doi: 10.1210/jc.2011-1802. [DOI] [PubMed] [Google Scholar]
  • 64.Reger MA, Watson GS, Green PS, et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology. 2008 Feb 5;70:440–8. doi: 10.1212/01.WNL.0000265401.62434.36. [DOI] [PubMed] [Google Scholar]
  • 65.Schioth HB, Craft S, Brooks SJ, et al. Brain insulin signaling and Alzheimer's disease: current evidence and future directions. Molecular neurobiology. 2012 Aug;46:4–10. doi: 10.1007/s12035-011-8229-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hallschmid M, Benedict C, Schultes B, et al. Towards the therapeutic use of intranasal neuropeptide administration in metabolic and cognitive disorders. Regulatory peptides. 2008 Aug 7;149:79–83. doi: 10.1016/j.regpep.2007.06.012. [DOI] [PubMed] [Google Scholar]; * This reference is important for highlighting the role and effectiveness of intranasal therapy for delivering insulin and other neuropeptides to the brain. Some of the key concepts pertaining to the effectiveness of intranasal insulin are discussed along with therapeutic responses.
  • 67.Ryan KK, Woods SC, Seeley RJ. Central nervous system mechanisms linking the consumption of palatable high-fat diets to the defense of greater adiposity. Cell metabolism. 2012 Feb 8;15:137–49. doi: 10.1016/j.cmet.2011.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Reagan LP. Insulin signaling effects on memory and mood. Current opinion in pharmacology. 2007 Dec;7:633–7. doi: 10.1016/j.coph.2007.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hallschmid M, Benedict C, Schultes B, et al. Intranasal insulin reduces body fat in men but not in women. Diabetes. 2004 Nov;53:3024–9. doi: 10.2337/diabetes.53.11.3024. [DOI] [PubMed] [Google Scholar]
  • 70.Guthoff M, Grichisch Y, Canova C, et al. Insulin modulates food-related activity in the central nervous system. The Journal of clinical endocrinology and metabolism. 2010 Feb;95:748–55. doi: 10.1210/jc.2009-1677. [DOI] [PubMed] [Google Scholar]
  • 71.Guthoff M, Stingl KT, Tschritter O, et al. The insulin-mediated modulation of visually evoked magnetic fields is reduced in obese subjects. PloS one. 2011;6:e19482. doi: 10.1371/journal.pone.0019482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hallschmid M, Benedict C, Schultes B, et al. Obese men respond to cognitive but not to catabolic brain insulin signaling. Int J Obes (Lond) 2008 Feb;32:275–82. doi: 10.1038/sj.ijo.0803722. [DOI] [PubMed] [Google Scholar]
  • 73.Heni M, Kullmann S, Ketterer C, et al. Nasal insulin changes peripheral insulin sensitivity simultaneously with altered activity in homeostatic and reward-related human brain regions. Diabetologia. 2012 Jun;55:1773–82. doi: 10.1007/s00125-012-2528-y. [DOI] [PubMed] [Google Scholar]
  • 74.Benedict C, Kern W, Schultes B, et al. Differential sensitivity of men and women to anorexigenic and memory-improving effects of intranasal insulin. The Journal of clinical endocrinology and metabolism. 2008 Apr;93:1339–44. doi: 10.1210/jc.2007-2606. [DOI] [PubMed] [Google Scholar]
  • 75.Krug R, Benedict C, Born J, et al. Comparable sensitivity of postmenopausal and young women to the effects of intranasal insulin on food intake and working memory. The Journal of clinical endocrinology and metabolism. 2010 Dec;95:E468–72. doi: 10.1210/jc.2010-0744. [DOI] [PubMed] [Google Scholar]
  • 76.Kullmann S, Frank S, Heni M, et al. Intranasal insulin modulates intrinsic reward and prefrontal circuitry of the human brain in lean women. Neuroendocrinology. 2013;97:176–82. doi: 10.1159/000341406. [DOI] [PubMed] [Google Scholar]
  • 77.Francis GJ, Martinez JA, Liu WQ, et al. Intranasal insulin prevents cognitive decline, cerebral atrophy and white matter changes in murine type I diabetic encephalopathy. Brain : a journal of neurology. 2008 Dec;131:3311–34. doi: 10.1093/brain/awn288. [DOI] [PubMed] [Google Scholar]
  • 78.Bonifacio E, Achenbach P, Pan L, et al. Mucosal insulin vaccination for type 1 diabetes prevention. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association. 2008 Sep;116(Suppl 1):S26–9. doi: 10.1055/s-2008-1081484. [DOI] [PubMed] [Google Scholar]
  • 79.Nanto-Salonen K, Kupila A, Simell S, et al. Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial. Lancet. 2008 Nov 15;372:1746–55. doi: 10.1016/S0140-6736(08)61309-4. [DOI] [PubMed] [Google Scholar]
  • 80.Vig PJ, Subramony SH, D'Souza DR, et al. Intranasal administration of IGF-I improves behavior and Purkinje cell pathology in SCA1 mice. Brain research bulletin. 2006 May 15;69:573–9. doi: 10.1016/j.brainresbull.2006.02.020. [DOI] [PubMed] [Google Scholar]
  • 81.Liu XF, Fawcett JR, Hanson LR, et al. The window of opportunity for treatment of focal cerebral ischemic damage with noninvasive intranasal insulin-like growth factor-I in rats. Journal of stroke and cerebrovascular diseases : the official journal of National Stroke Association. 2004 Jan-Feb;13:16–23. doi: 10.1016/j.jstrokecerebrovasdis.2004.01.005. [DOI] [PubMed] [Google Scholar]
  • 82.Lin S, Fan LW, Rhodes PG, et al. Intranasal administration of IGF-1 attenuates hypoxic-ischemic brain injury in neonatal rats. Experimental neurology. 2009 Jun;217:361–70. doi: 10.1016/j.expneurol.2009.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kooijman R, Sarre S, Michotte Y, et al. Insulin-like growth factor I: a potential neuroprotective compound for the treatment of acute ischemic stroke? Stroke; a journal of cerebral circulation. 2009 Apr;40:e83–8. doi: 10.1161/STROKEAHA.108.528356. [DOI] [PubMed] [Google Scholar]
  • 84.Patel MM, Goyal BR, Bhadada SV, et al. Getting into the brain: approaches to enhance brain drug delivery. CNS drugs. 2009;23:35–58. doi: 10.2165/0023210-200923010-00003. [DOI] [PubMed] [Google Scholar]
  • 85.Reger MA, Craft S. Intranasal insulin administration: a method for dissociating central and peripheral effects of insulin. Drugs Today (Barc) 2006 Nov;42:729–39. doi: 10.1358/dot.2006.42.11.1007675. [DOI] [PubMed] [Google Scholar]
  • 86.D'Souza R, Mutalik S, Venkatesh M, et al. Insulin gel as an alternate to parenteral insulin: formulation, preclinical, and clinical studies. AAPS PharmSciTech. 2005;6:E184–9. doi: 10.1208/pt060227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Teijeiro-Osorio D, Remunan-Lopez C, Alonso MJ. New generation of hybrid poly/oligosaccharide nanoparticles as carriers for the nasal delivery of macromolecules. Biomacromolecules. 2009 Feb 9;10:243–9. doi: 10.1021/bm800975j. [DOI] [PubMed] [Google Scholar]
  • 88.Sakuma S, Suita M, Masaoka Y, et al. Oligoarginine-linked polymers as a new class of penetration enhancers. Journal of controlled release : official journal of the Controlled Release Society. 2010 Dec 1;148:187–96. doi: 10.1016/j.jconrel.2010.08.022. [DOI] [PubMed] [Google Scholar]
  • 89.McInnes FJ, O'Mahony B, Lindsay B, et al. Nasal residence of insulin containing lyophilised nasal insert formulations, using gamma scintigraphy. European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences. 2007 May;31:25–31. doi: 10.1016/j.ejps.2007.02.002. [DOI] [PubMed] [Google Scholar]
  • 90.Chung SW, Hil-lal TA, Byun Y. Strategies for non-invasive delivery of biologics. Journal of drug targeting. 2012 Jul;20:481–501. doi: 10.3109/1061186X.2012.693499. [DOI] [PubMed] [Google Scholar]
  • 91.Illum L. Nanoparticulate systems for nasal delivery of drugs: a real improvement over simple systems? Journal of pharmaceutical sciences. 2007 Mar;96:473–83. doi: 10.1002/jps.20718. [DOI] [PubMed] [Google Scholar]
  • 92.Arnold JJ, Fyrberg MD, Meezan E, et al. Reestablishment of the nasal permeability barrier to several peptides following exposure to the absorption enhancer tetradecyl-beta-D-maltoside. Journal of pharmaceutical sciences. 2010 Apr;99:1912–20. doi: 10.1002/jps.21977. [DOI] [PubMed] [Google Scholar]
  • 93.Duan X, Mao S. New strategies to improve the intranasal absorption of insulin. Drug discovery today. 2010 Jun;15:416–27. doi: 10.1016/j.drudis.2010.03.011. [DOI] [PubMed] [Google Scholar]
  • 94.Stote R, Marbury T, Shi L, et al. Comparison pharmacokinetics of two concentrations (0.7% and 10%) of Nasulin, an ultra-rapid-acting intranasal insulin formulation. Journal of diabetes science and technology. 2010 May;4:603–9. doi: 10.1177/193229681000400314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Stote R, Miller M, Marbury T, et al. Enhanced absorption of Nasulin, an ultrarapid-acting intranasal insulin formulation, using single nostril administration in normal subjects. Journal of diabetes science and technology. 2011 Jan;5:113–9. doi: 10.1177/193229681100500116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Krishnankutty RK, Mathew A, Sedimbi SK, et al. Alternative routes of insulin delivery. Zhong nan da xue xue bao Yi xue ban = Journal of Central South University Medical sciences. 2009 Oct;34:933–48. [PubMed] [Google Scholar]
  • 97.Leary AC, Stote RM, Breedt HJ, et al. Pharmacokinetics and pharmacodynamics of intranasal insulin administered to healthy subjects in escalating doses. Diabetes technology & therapeutics. 2005 Feb;7:124–30. doi: 10.1089/dia.2005.7.124. [DOI] [PubMed] [Google Scholar]
  • 98.Strachan MW. Insulin and cognitive function in humans: experimental data and therapeutic considerations. Biochemical Society transactions. 2005 Nov;33:1037–40. doi: 10.1042/BST20051037. [DOI] [PubMed] [Google Scholar]
  • 99.Benedict C, Hallschmid M, Schmitz K, et al. Intranasal insulin improves memory in humans: superiority of insulin aspart. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2007 Jan;32:239–43. doi: 10.1038/sj.npp.1301193. [DOI] [PubMed] [Google Scholar]; ** This reference demonstrates the advances in preserving memory by supporting insulin actions in the brain. The importance of intranasal insulin as a therapeutic approach that minimizes systemic side-effects is shown.
  • 100.Benedict C, Dodt C, Hallschmid M, et al. Immediate but not long-term intranasal administration of insulin raises blood pressure in human beings. Metabolism: clinical and experimental. 2005 Oct;54:1356–61. doi: 10.1016/j.metabol.2005.04.026. [DOI] [PubMed] [Google Scholar]
  • 101.Hotta O, Taguma Y, Chiba S, et al. Possible relationship between hyperinsulinemia and glomerular hypertrophy in nephrosclerosis. Ren Fail. 1996 Mar;18:271–8. doi: 10.3109/08860229609052797. [DOI] [PubMed] [Google Scholar]
  • 102.Luchsinger JA, Tang MX, Shea S, et al. Hyperinsulinemia and risk of Alzheimer disease. Neurology. 2004 Oct 12;63:1187–92. doi: 10.1212/01.wnl.0000140292.04932.87. [DOI] [PubMed] [Google Scholar]
  • 103.Fishel MA, Watson GS, Montine TJ, et al. Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults. Archives of neurology. 2005 Oct;62:1539–44. doi: 10.1001/archneur.62.10.noc50112. [DOI] [PubMed] [Google Scholar]
  • 104.Neumann KF, Rojo L, Navarrete LP, et al. Insulin resistance and Alzheimer's disease: molecular links & clinical implications. Curr Alzheimer Res. 2008 Oct;5:438–47. doi: 10.2174/156720508785908919. [DOI] [PubMed] [Google Scholar]
  • 105.Roriz-Filho SJ, Sa-Roriz TM, Rosset I, et al. (Pre)diabetes, brain aging, and cognition. Biochim Biophys Acta. 2009 May;1792:432–43. doi: 10.1016/j.bbadis.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 106.Lahousse SA, Carter JJ, Xu XJ, et al. Differential growth factor regulation of aspartyl-(asparaginyl)-beta-hydroxylase family genes in SH-Sy5y human neuroblastoma cells. BMC cell biology. 2006;7:41. doi: 10.1186/1471-2121-7-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Imfeld P, Bodmer M, Jick SS, et al. Metformin, other antidiabetic drugs, and risk of Alzheimer's disease: a population-based case-control study. Journal of the American Geriatrics Society. 2012 May;60:916–21. doi: 10.1111/j.1532-5415.2012.03916.x. [DOI] [PubMed] [Google Scholar]
  • 108.Watson GS, Cholerton BA, Reger MA, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. The American journal of geriatric psychiatry : official journal of the American Association for Geriatric Psychiatry. 2005 Nov;13:950–8. doi: 10.1176/appi.ajgp.13.11.950. [DOI] [PubMed] [Google Scholar]
  • 109.Risner ME, Saunders AM, Altman JF, et al. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer's disease. The pharmacogenomics journal. 2006 Jul-Aug;6:246–54. doi: 10.1038/sj.tpj.6500369. [DOI] [PubMed] [Google Scholar]
  • 110.Roses AD, Saunders AM, Huang Y, et al. Complex disease-associated pharmacogenetics: drug efficacy, drug safety, and confirmation of a pathogenetic hypothesis (Alzheimer's disease) The pharmacogenomics journal. 2007 Feb;7:10–28. doi: 10.1038/sj.tpj.6500397. [DOI] [PubMed] [Google Scholar]
  • 111.Escribano L, Simon AM, Gimeno E, et al. Rosiglitazone rescues memory impairment in Alzheimer's transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2010 Jun;35:1593–604. doi: 10.1038/npp.2010.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Escribano L, Simon AM, Perez-Mediavilla A, et al. Rosiglitazone reverses memory decline and hippocampal glucocorticoid receptor down-regulation in an Alzheimer's disease mouse model. Biochemical and biophysical research communications. 2009 Feb 6;379:406–10. doi: 10.1016/j.bbrc.2008.12.071. [DOI] [PubMed] [Google Scholar]
  • 113.Gold M, Alderton C, Zvartau-Hind M, et al. Rosiglitazone monotherapy in mild-to-moderate Alzheimer's disease: results from a randomized, double-blind, placebo-controlled phase III study. Dementia and geriatric cognitive disorders. 2010;30:131–46. doi: 10.1159/000318845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Tzimopoulou S, Cunningham VJ, Nichols TE, et al. A multi-center randomized proof-of-concept clinical trial applying [(1)(8)F]FDG-PET for evaluation of metabolic therapy with rosiglitazone XR in mild to moderate Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2010;22:1241–56. doi: 10.3233/JAD-2010-100939. [DOI] [PubMed] [Google Scholar]
  • 115.Harrington C, Sawchak S, Chiang C, et al. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimer's disease: two phase 3 studies. Curr Alzheimer Res. 2011 Aug;8:592–606. doi: 10.2174/156720511796391935. [DOI] [PubMed] [Google Scholar]
  • 116.Miller BW, Willett KC, Desilets AR. Rosiglitazone and pioglitazone for the treatment of Alzheimer's disease. The Annals of pharmacotherapy. 2011 Nov;45:1416–24. doi: 10.1345/aph.1Q238. [DOI] [PubMed] [Google Scholar]
  • 117.Galea E, Feinstein DL, Lacombe P. Pioglitazone does not increase cerebral glucose utilisation in a murine model of Alzheimer's disease and decreases it in wild-type mice. Diabetologia. 2006 Sep;49:2153–61. doi: 10.1007/s00125-006-0326-0. [DOI] [PubMed] [Google Scholar]

RESOURCES