Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2944 2022-04-12 12:25:23 |
2 update references and layout -34 word(s) 2910 2022-04-13 04:02:48 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Cacabelos, R.; Cacabelos, R.; , .; Martínez Iglesias, O.; Corzo, L.; Carril, J.C. Personalized Management and Treatment of Alzheimer’s Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/21643 (accessed on 29 March 2024).
Cacabelos R, Cacabelos R,  , Martínez Iglesias O, Corzo L, Carril JC. Personalized Management and Treatment of Alzheimer’s Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/21643. Accessed March 29, 2024.
Cacabelos, Ramon, Ramón Cacabelos,  , Olaia Martínez Iglesias, Lola Corzo, Juan C Carril. "Personalized Management and Treatment of Alzheimer’s Disease" Encyclopedia, https://encyclopedia.pub/entry/21643 (accessed March 29, 2024).
Cacabelos, R., Cacabelos, R., , ., Martínez Iglesias, O., Corzo, L., & Carril, J.C. (2022, April 12). Personalized Management and Treatment of Alzheimer’s Disease. In Encyclopedia. https://encyclopedia.pub/entry/21643
Cacabelos, Ramon, et al. "Personalized Management and Treatment of Alzheimer’s Disease." Encyclopedia. Web. 12 April, 2022.
Personalized Management and Treatment of Alzheimer’s Disease
Edit

Alzheimer’s disease (AD) is a priority health problem with a high cost to society and a large consumption of medical and social resources. The management of AD patients is complex and multidisciplinary. Over 90% of patients suffer from concomitant diseases and require personalized therapeutic regimens to reduce adverse drug reactions (ADRs), drug–drug interactions (DDIs), and unnecessary costs. Men and women show substantial differences in their AD-related phenotypes. Genomic, epigenetic, neuroimaging, and biochemical biomarkers are useful for predictive and differential diagnosis. The most frequent concomitant diseases include hypertension (>25%), obesity (>70%), diabetes mellitus type 2 (>25%), hypercholesterolemia (40%), hypertriglyceridemia (20%), metabolic syndrome (20%), hepatobiliary disorder (15%), endocrine/metabolic disorders (>20%), cardiovascular disorder (40%), cerebrovascular disorder (60–90%), neuropsychiatric disorders (60–90%), and cancer (10%). Over 90% of AD patients require multifactorial treatments with risk of ADRs and DDIs. The implementation of pharmacogenetics in clinical practice can help optimize the limited therapeutic resources available to treat AD and personalize the use of anti-dementia drugs, in combination with other medications, for the treatment of concomitant disorders. 

Alzheimer’s disease anti-dementia drugs biomarkers concomitant disorders neurodegenerative genomics pathogenic genes pharmacogenomics

1. Phenotypic Features

In a cohort of randomly-selected patients diagnosed with Alzheimer’s disease (AD) (DSM-V and NINCDS-ADRDA criteria) (n = 2701; age: 67.63 ± 0.19 years; range: 50–96 years) of both sexes (1491 Females (55%); age: 68.26 ± 0.27 years; range: 50–96 years; 1210 Males (45%); age: 66.86 ± 0.28; p < 0.001), retrospectively recruited from the CIBE database (period: 2000–2020), it was investigated sex-related common phenotypes, including biochemistry, hematology, metabolism, hormones, neurotransmitters, cardiovascular and cerebrovascular function, cognition, mood, behavior, and genomic and pharmacogenomic profiles.
In terms of anthropometric parameters, substantial differences are observed between females and males in terms of height, weight, and body-mass index (BMI). Only 28% of patients show normal weight (29% F; 20% M), with obesity in >25% and only 1% with underweight.
Significant differences between females and males are present across many biological parameters, including (i) biochemical (glucose, total-cholesterol, HDL-cholesterol, LDL-cholesterol, triglycerides, urea, creatinine, uric acid, calcium, phosphorous, liver transaminases (ALAT, GGT), alkaline phosphatase, bilirubin, CPK, LDH, ions, iron, ferritin, folate, and vitamin B12), (ii) hormonal (TSH, PRL, ACTH, FSH, LH, estrogen, and testosterone), (iii) hematological (RCB, HCT, Hb, MCV, MCH, WBC, lymphocytes, monocytes, eosinophils, basophils, and platelets), (iv) cognitive, and (v) emotional markers. No differences are found in tumor markers.
Cognitive markers (MMSE, ADAS) indicate than females show worse cognitive performance than males. Cognitive impairment and depression are the first symptoms, which appear in over 90% of LOAD, and 80% and 9%, respectively, in EOAD [1].
Late-life depression is associated with cognitive impairment, and depression is linked with an increased risk for AD. Some overlapping pathogenic substrates (for example, stress, cortisol levels, brain hypoperfusion, neuroinflammation, neurotrophic dysfunction, Aβ accumulation, tauopathic connections, epigenetic factors, and gut microbiota–brain axis) may explain the comorbidity of both clinical entities [2]. Mood disorders are more frequent in women than in men. Over 60% of AD patients show depressive symptoms, which are more severe in women than in men. Likewise, anxiety is also more frequent in females than in males. About 50% of men do not show anxiety, whereas only 30% of women with dementia are free of symptoms in early stages of the disease. Both anxiety and depression fluctuate with the clinical course of the disease [3][4][5][6][7]. Behavioral disorders and psychotic symptoms are also frequent (20–90%) in patients with AD along the clinical course of the disease [3][8][9].
Cardiovascular evaluation (ECG) is abnormal in 40% of the patients (38% F; 43% M). A normal ECG is found more frequently in females (52%) than males (43%; p < 0.01), and a borderline ECG appears more frequently in males (12%) than in females (9%; p < 0.01) (Figure 1, upper panel); however, these differences are unrelated to apolipoprotein E (APOE) variants in both sexes (Figure 1, lower panel). Despite this global perception, and considering the small number of the APOE-2/2 and APOE-4/4 genotypes, APOE-4/4 carriers show abnormal ECGs (52%) more frequently than APOE-3/3 carriers, especially in males (36%) as compared to females (10%); in contrast, homozygous APOE-2/2 carriers exhibit the lowest frequency of abnormal ECG in the whole sample (25%), with twice the number of men (50%) showing abnormal ECG than APOE-2/2 females (25%) (Figure 1, lower panel).
Life 12 00460 g001 550
Figure 1. ECG (upper panel) and APOE-related ECG (lower panel) in patients with Alzheimer’s disease.

2. Biomarkers

In addition to conventional clinical markers, which was allowed to make a differential diagnosis and assess the possibility of concomitant diseases, the most useful biomarkers for a predictive diagnosis or diagnostic confirmation of antemortem AD are genomic markers, epigenetic biomarkers, neurotransmitters, and levels of Aβ/tau in the brain (PET Scan) and/or in body fluids [10][11][12][13][14][15][16][17][18][19].

2.1. Genomic Markers

Over 600 human genes are associated with AD [20][21][22]. Mutations in the amyloid precursor protein (APP) (>50 different mutations), presenilin 1 (PSEN1 > 300 mutations), and presenilin-2 (PSEN2 > 40 mutations) genes are present in a number of AD (5–10%), and induce brain amyloidopathy. Microtubule-associated protein tau (MAPT) gene mutations (>100), also present in some patients with AD, may cause brain tauopathies (for example, frontotemporal dementia, Pick’s disease) [23][24][25]. Both conditions (amyloidopathy and tauopathy) are the two dominant pathogenic hypotheses in AD [26][27].
APP mutations can cause EOAD, with increased Aβ levels or Aβ fibrillogenesis, while some coding variants (APP A673T) may be protective with reduced Aβ levels [25]. EOAD dominant mutations tend to occur in the APP coding region or in the presenilin-related catalytic site of γ-secretase whose protease dysfunction is responsible for the abnormal process of APP cleavage and consequent accumulation of Aβ in senile plaques and vessels. In >40% AD, the presence of the apolipoprotein E4 (APOE-4) allele is the most important risk factor, involved in the impairment of Aβ clearance from brain tissue, in atherosclerosis and in hypoperfusion. Most immunotherapeutic interventions with Aβ antibodies (aducanumab, solanezumab, and crenezumab) attempt to halt the amyloidogenic process and slow-down cognitive deterioration in mild-AD or in presymptomatic ones with demonstrated genomic dysfunction [26]. In addition to these primary pathogenic genes, many other genes have been associated with AD in next-generation sequencing (NGS) and genome-wide association studies (GWAS) in different populations [21][22][28][29][30][31][32].
Recently, it was showed that multiple genetic defects can accumulate in the same of AD, conditioning its phenotypic characteristics. In a genomic panel of 18 AD-related pathogenic genes and genotypes related to cerebrovascular disorders (Figure 2), it can be verified that >60% of patients are carriers of more than 10 pathogenic variants.
Life 12 00460 g002 550
Figure 2. Pathogenic gene variants (upper panel) and cerebrovascular risk gene variants (lower panel) associated with Alzheimer’s disease. 
The genes that most frequently accumulate pathogenic variants (>50%) in cases of AD are the following: PRNP (80.70%), ACE (78.94%), PSEN1 (77.19%), CLU (63.15%), CPZ (63.15%), MS4A6A (63.15%), BIN1 (57.89%), A2M (54.38%), PICALM (54.38%), LHFPL6 (52.63%), and MS4A4E (50.87%) (Figure 3).
Life 12 00460 g003 550
Figure 3. Accumulation of defective pathogenic gene variants in patients with Alzheimer’s disease.
The pathogenic load of APOE-4 affects 35–40%, with significant phenotypic consequences. APOE-4/4 carriers tend to show an earlier age-at-onset in >80%; lower peripheral ApoE, nitric oxide, histamine, Aβ, HDL-cholesterol, and triglyceride levels; higher levels of total cholesterol and LDL-cholesterol; more pronounced brain atrophy and slower brain bioelectrical activity; more severe brain hemodynamic dysfunction represented by hypoperfusion, reduced brain blood flow velocity and increased pulsatility and resistance indices; increased lymphocyte apoptosis; faster cognitive deterioration; more frequent metabolic disorders, cardiovascular disorders, hypertension, atherosclerosis, liver metabolism dysfunction, behavioral disturbances, and alterations in circadian rhythm patterns; and a poor response to conventional treatments [20][22][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49].

2.2. Epigenetic Markers

Various epigenetic aberrations are associated with AD pathogenesis, including hypomethylation/hypermethylation in the promoters of pathogenic genes, alterations in histones, and changes in the linear and three-dimensional structure of nuclear chromatin, as well as profound alterations in microRNAs (miRNAs) that regulate gene expression in the cellular cytoplasm. Some of these epigenetic alterations have been proposed as potential biomarkers of AD [50][51][52][53][54][55][56][57].
The main problems observed with the use of epigenetic biomarkers in AD are their variability and lack of specificity. Changes in global DNA methylation are very sensitive and appear diminished in a multitude of central nervous system (CNS) diseases, such as AD (p < 0.001), Parkinson’s disease (p < 0.004), cerebrovascular disorders and stroke (p < 0.01), major depression (p < 0.05), migraine (p < 0.03), epilepsy (p < 0.05), and intellectual organic disability (OID) (p < 0.001), and to a lesser extent in schizophrenia (Figure 4, upper panel). These values are very sensitive to therapeutic interventions but unreliable as predictive or diagnostic values. The low diagnostic value of DNA methylation is compensated for by the exquisite sensitivity of this biomarker that responds in a highly sensitive manner to the therapeutic response of each patient. In addition, what appears to be important in AD is that global DNA methylation shows an APOE-dependent pattern. APOE-4 carriers tend to show a more severe DNA hypomethylation pattern than patients carrying the APOE-3 allele, which is aggravated in parallel with the degree of cognitive impairment (Figure 4, lower panel).
Life 12 00460 g004 550
Figure 4. Global DNA methylation in patients with central nervous system disorders (upper panel) and APOE-related DNA methylation in patients with Alzheimer’s disease (lower panel). C: Control; AD: Alzheimer’s disease; PD: Parkinson’s disease; CVD: Cerebrovascular disorder; MD: Major Depression; SCZ: Schizophrenia and psychotic syndromes; MIG: Migraine; EPI: Epilepsy; OID: Organic Intellectual Disability.
Epigenetic biomarkers can also help in the personalization of anti-AD treatments (Pharmacoepigenetics) [58][59][60][61] and serve as a guide in the search for epigenetic drugs with prophylactic and/or therapeutic action in the treatment of AD [50][59][60][61].

2.3. Neurotransmitters

Conventionally, the main neurotransmitter affected in AD is acetylcholine. However, premature neuronal death alters the levels of many other essential neurotransmitters for the normal functioning of the CNS [62][63][64].
Deficits in noradrenaline, dopamine, serotonin, histamine, GABA, glutamate, and various neuropeptides (GRF, CRF, somatostatin, and vasopressin) are particularly relevant, the alteration of which can lead to AD-related neuropsychiatric disorders. However, none of these biomarkers are sufficiently sensitive or specific to AD (Figure 5), although their quantification in CSF or blood is useful for monitoring brain damage and/or the efficacy or ineffectiveness of the treatments the patient receives.
Life 12 00460 g005 550
Figure 5. Noradrenaline (upper panel) and serotonin levels (lower panel) in central nervous system disorders. C: Control; ANS: Anxiety; STR: Stroke; BTU: Brain tumors; ATX: Ataxia; SCZ; Schizophrenia and psychosis; MIG: Migraine; EPI: Epilepsy; AD: Alzheimer’s disease; DEP: Depression; ALS: Amyotrophic Lateral Sclerosis; VEN: Vascular encephalopathy; MS: Multiple Sclerosis; PD: Parkinson’s disease; OID: Organic Intellectual Disability; XES: Xenoestrogenic syndrome; and BTR: Brain trauma.
Noradrenaline levels in the blood increase significantly in most neurodegenerative diseases, including ataxic syndromes (p < 0.002), AD (p < 0.001), amyotrophic lateral sclerosis (ALS) (p < 0.007), and Parkinson’s disease (p < 0.003), as well as in vascular encephalopathies (p < 0.001) and in OID (Figure 5, upper panel). In contrast, serotonin levels tend to decrease in anxiety (p < 0.02), AD (p < 0.05), depression (p < 0.05), and ALS (p < 0.05), and show high levels in ataxic syndromes (p < 0.002), OID (p < 0.05), and in xenoestrogenic syndrome (p < 0.001), a novel clinical entity present in women with chronic use of contraceptives or hormone replacement therapy (HRT) (Figure 5, lower panel).

2.4. Aβ/Tau Levels

The most popular biomarkers for AD in body fluids (CSF, plasma) are the quantification of amyloid-β (Aβ42), total tau (T-tau), and phosphorylated tau (P-tau) in the CSF (AD CSF profile: decreased Aβ42 levels together with increased T-tau and P-tau levels). Other CSF markers (synaptotagmin, rab3a, SNAP-25, and neurogranin) are also altered [13][14]. However, the heterogeneity of AD and the inconvenience of having to perform a lumbar puncture to obtain CSF do not allow these biomarkers to reach high quotas of sensitivity and specificity, nor generalized use.

3. Concomitant Disorders and Phenotype-Modifying Treatments

Most patients with dementia (>90%) require multifactorial treatments for the management of concomitant disorders and/or neuropsychiatric symptoms associated with dementia. The chronic administration of drugs from different categories increases the risk of ADRs and DDIs [65][66][67][68].
The most frequent concomitant disorders in AD are the following: systolic hypertension (21%), diastolic hypertension (28%), obesity (>70%), diabetes mellitus type 2 (26%), hypercholesterolemia (40%), hypertriglyceridemia (20%), hyperuricemia (6%), metabolic syndrome (20%), transaminitis (11%), hyperbilirubinemia (15%), endocrine disorders (5%), iron deficiency anemia (7%), folate deficit (17%), vitamin B12 deficit (10%), cardiovascular disorder (40%), cerebrovascular disorder (>90% in patients over 80 years of age), anxiety (60%), depression (65%), behavioral disorders (20–90%), and cancer (10%).
Cardiovascular risk factors (hypertension, hypercholesterolemia, and dyslipidemia) and ECG abnormalities are more frequent in males than in females. Hypertension is present in 21%. Systolic blood pressure is similar in females and males, but diastolic blood pressure is significantly higher in males than in females (p < 0.001). Cholesterol levels (Total, LDL) are higher in men and HDL-cholesterol and triglyceride levels are more elevated in females. Brain damage and increased cognitive deterioration are currently associated with cardiovascular disorders and blood pressure changes in AD [69][70]. APOE-4 carriers with dementia also exhibit cardiovascular disorders, atherosclerosis, and cerebrovascular damage [16][17][18][20][34][38][39][40][41][42][43][44][71]. Lipid metabolism disorders contribute to the cerebrovascular component of AD. Abnormalities in cholesterol metabolism and dysfunction of lipid rafts in cell membranes and arteriosclerosis are pathogenically relevant for cerebral ischemia and hypoperfusion, and accelerate premature neuronal death in patients who are predisposed to AD [17][72][73][74][75]. In contrast, the epidemiological link between diabetes and AD appears to be circumstantial, with no apparent pathogenic implications beyond the deleterious effects of hyperglycemia on brain function [76][77][78].
As a consequence of all these concomitant pathologies, patients with dementia consume a wide variety of drugs whose side-effects contribute to accelerating the degenerative process and cognitive decline. Of special importance are cardiovascular agents, statins, antidiabetics, antihypertensive drugs, analgesics, diuretics, bronchodilators, antirheumatics, and various categories of psychotropic drugs (neuroleptics, antidepressants, anxiolytics, hypnotics, and sedatives). The correct administration of these drugs requires a personalized therapeutic intervention, together with conventional anti-dementia treatments [65][79][80].
Combination treatments applied under pharmacogenetic guidance indicate that alterations in biochemical, hematological, and metabolic parameters affect drug efficacy and safety. Concerning cognitive function and neuropsychiatric disorders treated with multifactorial regimes, females and males respond differentially to treatment, showing a moderate improvement in cognition during the first year of treatment and significant improvements in anxiety and depression.
Several pharmacogenetic ones agree that APOE-4 carriers respond poorly to conventional treatments, while APOE-3 carriers tend to respond better to different therapeutic regimens. Similarly, normal metabolizers (NMs) and intermediate metabolizers (IMs) associated with the different genotypes of the CYP2D6, CYP2C9, and CYP2C19 genes show a better therapeutic response than poor metabolizers (PMs) and ultra-rapid metabolizers (UMs) to treatments with anti-dementia drugs and psychotropic drugs to regulate aberrant behaviors [3][38][39][40][41][42][43][44][45].

4. Alzheimer’s Disease Therapeutics and Drug Development

For the past 50 years, the major focus of pharmacological development in AD has been cognitive enhancers. The introduction of acetylcholinesterase inhibitors (AChEIs) in the early 1990s was the first option to restore cholinergic neurotransmission after the identification of a selective cholinergic deficit in the basal forebrain and neuronal loss in neocortex and hippocampus. Tacrine (9-amino-1,2,3,4-tetrahydroacridine) was the first AChEI introduced in 1993 for the treatment of AD. The Chinese product Huperzine A was approved in 1994.
A new generation of AChEIs (donepezil, galantamine, and rivastigmine) was introduced years later. In 2003, the FDA approved memantine, an N-methyl-D-aspartate (NMDA) glutamate receptor partial inhibitor. Since then, no new FDA-approved drugs for AD were reported until the recent approval of the antibody aducanumab in 2021 [80][81].
The main categories of drugs known during the last two decades as candidates for the treatment of AD were heterogeneous and include the following: neurotransmitter enhancers (11.38%), anti-amyloid agents (13.30%), anti-tau agents (2.03%), multi-target drugs (8.11%), novel synthetic drugs (8.13%), neuroprotective peptides (1.25%), old repository drugs (11.77%), anti-inflammatory drugs (1.20%), and a large number of natural products and derivatives (25.58%). Novel categories of therapeutic intervention (stem cell therapy, nanocarriers/nanotherapeutics) and combination treatments have also been challenged [81]. Over 2000 new AChEIs, some of them with dual inhibitory activity on acetylcholinesterase and butyrylcholinesterase, have been identified, as well as over 150 multi-target drugs [81][82]. Approximately 15% of pharmacological ones searching for anti-AD drugs focused on anti-amyloidogenic strategies (immunotherapy, APP modulators, α-secretase modulators, β-secretase (BACE) inhibitors, ϒ-secretase modulators, Aβ aggregation inhibitors, Notch inhibitors, β-sheet breakers, and Aβ scavengers), with notorious failures [81]. For the past two years, an increase in the number of disease-modifying agents targeting non-amyloid or tau pathogenic cascades has been observed [81][82][83][84].

4.1. Immunotherapy

Since the early 2000s, several categories of vaccines and immunotherapeutic procedures have been developed for the treatment of AD, following the pioneering ones of Schenk and coworkers in 1999 [85]. For two decades, millions of dollars have been invested in passive and active immunotherapy in experimental AD models and in clinical trials. About 1000 papers on AD immunotherapy were published (85% anti-Aβ and 15% anti-tau) prior to the FDA approval of aducanumab as an immunotherapeutic strategy in mild-AD [83][86].
Active immunization ones demonstrate that presymptomatic immunization of PDAPP transgenic mice, overexpressing human mutant APP (Phe717Val), prevents Aβ-plaque formation, astrogliosis, and neuritic dystrophy. Immunization with Aβ42 reduces the extent and progression of AD-related neurodegeneration and improves cognition in the TgCRND8 murine model of AD [85][86][87][88]. Over 400 experimental ones with different immunization procedures were able to replicate these results. However, clinical ones did not show the same efficacy and initial clinical trials with the Aβ vaccine AN1792 had to be discontinued. This was because of severe undesirable effects, such as acute meningoencephalitis or microhemorrhagic lesions in the brain of some immunized patients. Some of these ADRs were attributed to a T-cell-mediated pro-inflammatory response; other mechanistic causes of brain damage in patients treated with active vaccines remain elusive. Improvements in immunization processes and vaccine preparations in recent years have made it possible to obviate many of these drawbacks and avoid adverse effects with the second-generation Aβ-active immunotherapies, anti-tau immunotherapies, and anti-Aβ monoclonal antibodies targeting Aβ epitopes [89][90][91][92][93][94]. Other modalities of immunization, such as dual vaccines (EB101) [91], Aβ3-10-KLH vaccine [92], or active full-length DNA-Aβ42 trimer immunization [93], also reduce amyloidogenic and tauopathic markers in transgenic animals [94].
On 7 June 2021, aducanumab was formally approved by the FDA as a putative disease-modifying antibody for the treatment of AD [95][96][97][98][99][100]. Aducanumab penetrates into the brain, binds parenchymal Aβ, selectively targets aggregated Aβ, and dose-dependently reduces Aβ levels, with parallel slowing of cognitive decline [96][97][98][99]. Vasogenic edema is one of the major ADRs of aducanumab and other injectable antibodies (BAN2401), especially in APOE-4 carriers [100][101].

References

  1. Bature, F.; Guinn, B.A.; Pang, D.; Pappas, Y. Signs and symptoms preceding the diagnosis of Alzheimer’s disease: A systematic scoping review of literature from 1937 to 2016. BMJ Open 2017, 7, e015746.
  2. Linnemann, C.; Lang, U.E. Pathways Connecting Late-Life Depression and Dementia. Front. Pharmacol. 2020, 11, 279.
  3. Cacabelos, R.; Carril, J.C.; Corzo, L.; Fernández-Novoa, L.; Pego, R.; Cacabelos, N.; Cacabelos, P.; Alcaraz, M.; Tellado, I.; Naidoo, V. Influence of pathogenic and metabolic genes on the pharmacogenetics of mood disorders in Alzheimer’s disease. Pharmaceuticals 2021, 14, 366.
  4. Bennett, S.; Thomas, A.J. Depression and dementia: Cause, consequence or coincidence? Maturitas 2014, 79, 184–190.
  5. Gutzmann, H.; Qazi, A. Depression associated with dementia. Z. Gerontol. Geriatr. 2015, 48, 305–311.
  6. Baruch, N.; Burgess, J.; Pillai, M.; Allan, C.L. Treatment for depression comorbid with dementia. Evid. Based Ment. Health 2019, 22, 167–171.
  7. Bingham, K.S.; Flint, A.J.; Mulsant, B.H. Management of Late-Life Depression in the Context of Cognitive Impairment: A Review of the Recent Literature. Curr. Psychiatry Rep. 2019, 21, 74.
  8. Kratz, T. The Diagnosis and Treatment of Behavioral Disorders in Dementia. Dtsch. Arztebl. Int. 2017, 114, 447–454.
  9. Aarsland, D. Epidemiology and Pathophysiology of Dementia-Related Psychosis. J. Clin. Psychiatry 2020, 81, AD19038BR1C.
  10. Cacabelos, R. Pharmacogenomic Biomarkers in Neuropsychiatry: The Path to Personalized Medicine in Mental Disorders. In The Handbook of Neuropsychiatric Biomarkers, Endophenotypes and Genes. Molecular Genetic and Genomic Markers; Ritsner, M.S., Ed.; Springer: Amsterdam, The Netherlands, 2009; Volume 4, pp. 3–63.
  11. Caroli, A.; Frisoni, G.B.; Alzheimer’s Disease Neuroimaging Initiative. The dynamics of Alzheimer’s disease biomarkers in the Alzheimer’s Disease Neuroimaging Initiative cohort. Neurobiol. Aging 2010, 31, 1263–1274.
  12. Jack, C.R., Jr.; Knopman, D.S.; Jagust, W.J.; Peterson, R.C.; Weiner, M.W.; Aisen, P.S.; Shaw, L.M.; Vemuri, P.; Wiste, H.J.; Wigand, S.D.; et al. Tracking pathophysiological processes in Alzheimer’s disease: An updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013, 12, 207–216.
  13. Blennow, K.; Zetterberg, H. Biomarkers for Alzheimer’s disease: Current status and prospects for the future. J. Intern. Med. 2018, 284, 643–663.
  14. Zetterberg, H.; Bendlin, B.B. Biomarkers for Alzheimer’s disease: Preparing for a new era of disease-modifying therapies. Mol. Psychiatry 2021, 26, 296–308.
  15. Márquez, F.; Yassa, M.A. Neuroimaging Biomarkers for Alzheimer’s Disease. Mol. Neurodegener. 2019, 14, 21.
  16. Cacabelos, R. Genomic characterization of Alzheimer’s disease and genotype-related phenotypic analysis of biological markers in dementia. Pharmacogenomics 2004, 5, 1049–1105.
  17. Cacabelos, R.; Fernández-Novoa, L.; Corzo, L.; Amado, L.; Pichel, V.; Lombardi, V.; Kubota, Y. Phenotypic profiles and functional genomics in Alzheimer’s disease and in dementia with a vascular component. Neurol. Res. 2004, 26, 459–480.
  18. Cacabelos, R.; Fernández-Novoa, L.; Corzo, L.; Pichel, V.; Lombardi, V.; Kubota, Y. Genomics and phenotypic profiles in dementia: Implications for pharmacological treatment. Methods Find. Exp. Clin. Pharmacol. 2004, 26, 421–444.
  19. Cacabelos, R.; Lombardi, V.; Fernández-Novoa, L.; Kubota, Y.; Corzo, L.; Pichel, V.; Takeda, M. A functional genomics approach to the analysis of biological markers in Alzheimer disease. In Molecular Neurobiology of Alzheimer’s Disease and Related Disorders; Takeda, M., Tanaka, T., Cacabelos, R., Eds.; Karger: Basel, Switzerland, 2004; pp. 236–285.
  20. Cacabelos, R.; Fernández-Novoa, L.; Lombardi, V.; Kubota, Y.; Takeda, M. Molecular genetics of Alzheimer’s disease and aging. Methods Find. Exp. Clin. Pharmacol. 2005, 27, 1–573.
  21. Cacabelos, R.; Martínez-Bouza, R.; Carril, J.C.; Fernández-Novoa, L.; Lombardi, V.; Carrera, I.; Corzo, L.; McKay, A. Genomics and pharmacogenomics of brain disorders. Curr. Pharm. Biotechnol. 2012, 13, 674–725.
  22. Cacabelos, R.; Martínez-Bouza, R. Genomics and pharmacogenomics of dementia. CNS Neurosc. Ther. 2011, 17, 566–576.
  23. Cai, Y.; An, S.S.; Kim, S. Mutations in presenilin 2 and its implications in Alzheimer’s disease and other dementia-associated disorders. Clin. Interv. Aging 2015, 10, 1163–1172.
  24. Kelleher, R.J.; Shen, J. Presenilin-1 mutations and Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2017, 114, 629–631.
  25. Tcw, J.; Goate, A.M. Genetics of β-Amyloid Precursor Protein in Alzheimer’s Disease. Cold Spring Harb. Perspect. Med. 2017, 7, a024539.
  26. Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608.
  27. Tolar, M.; Abushakra, S.; Sabbagh, M. The path forward in Alzheimer’s disease therapeutics: Reevaluating the amyloid cascade hypothesis. Alzheimers Dement. 2020, 16, 1553–1560.
  28. Giau, V.V.; Bagyinszky, E.; Yang, Y.; Youn, Y.C.; An, S.S.A.; Kim, S.Y. Genetic analyses of early-onset Alzheimer’s disease using next generation sequencing. Sci. Rep. 2019, 9, 8368.
  29. Van Cauwenberghe, C.; van Broeckhoven, C.; Sleegers, K. The genetic landscape of Alzheimer disease: Clinical implications and perspectives. Genet. Med. 2016, 18, 421–430.
  30. Bertram, L. Next Generation Sequencing in Alzheimer’s Disease. Syst. Biol. Alzheimers Dis. 2016, 1303, 281–297.
  31. Zhu, J.B.; Tan, C.C.; Tan, L.; Yu, J.T. State of Play in Alzheimer’s Disease Genetics. J. Alzheimers Dis. 2017, 58, 631–659.
  32. Karch, C.M.; Goate, A.M. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol. Psychiatry 2015, 77, 43–51.
  33. Cacabelos, R.; Cacabelos, P.; Torrellas, C.; Tellado, I.; Carril, J.C. Pharmacogenomics of Alzheimer’s disease: Novel therapeutic strategies for drug development. Methods Mol. Biol. 2014, 1175, 323–556.
  34. Cacabelos, R.; Carril, J.C.; Cacabelos, P.; Teijido, O.; Goldgaber, D. Pharmacogenomics of Alzheimer’s Disease: Genetic determinants of phenotypic variation and therapeutic outcome. J. Genom. Med. Pharmacogenom. 2016, 1, 151–209.
  35. Cacabelos, R.; Carril, J.C.; Cacabelos, N.; Kazantsev, A.G.; Vostrov, A.V.; Corzo, L.; Cacabelos, P.; Goldgaber, D. Sirtuins in Alzheimer’s Disease: SIRT2-Related GenoPhenotypes and Implications for PharmacoEpiGenetics. Int. J. Mol. Sci. 2019, 20, 1249.
  36. Cacabelos, R.; Goldgaber, D.; Vostrov, A.; Matsuki, H.; Torrellas, C.; Corzo, L.; Carril, J.C.; Roses, A.D. APOE-TOMM40 in the Pharmacogenomics of demetia. J. Pharmacogen. Pharmacoproteom. 2014, 5, 135.
  37. Cacabelos, R.; Torrellas, C.; Teijido, O.; Carril, J.C. Pharmacogenetic considerations in the treatment of Alzheimer’s disease. Pharmacogenomics 2016, 17, 1041–1074.
  38. Cacabelos, R.; Meyyazhagan, A.; Carril, J.C.; Cacabelos, P.; Teijido, O. Pharmacogenetics of vascular risk factors in Alzheimer’s Disease. J. Pers. Med. 2018, 8, 3.
  39. Cacabelos, R. Pharmacogenomics of central nervous system (CNS) drugs. Drug Dev. Res. 2012, 73, 461–476.
  40. Cacabelos, R.; Takeda, M. Pharmacogenomics, nutrigenomics and future therapeutics in Alzheimer’s disease. Drugs Future 2006, 3, 5–146.
  41. Cacabelos, R. Psychogeriatric research: A conceptual introduction to aging and geriatric neuroscience. Psychogeriatrics 2001, 1, 158–188.
  42. Cacabelos, R. The application of functional genomics to Alzheimer’s disease. Pharmacogenomics 2003, 4, 597–621.
  43. Cacabelos, R. Pharmacogenomics in Alzheimer’s disease. Pharm. Drug Discov. Dev. 2008, 448, 213–357.
  44. Cacabelos, R.; Fernández-Novoa, L.; Martínez-Bouza, R.; McKay, A.; Carril, J.C.; Lombardi, V.; Corzo, L.; Carrera, I.; Tellado, I.; Nebril, L.; et al. Future trends in the pharmacogenomics of brain disorders and dementia: Influence of APOE and CYP2D6 variants. Pharmaceuticals 2010, 3, 3040–3100.
  45. Cacabelos, R. Molecular pathology and pharmacogenomics in Alzheimer’s disease: Polygenic-related effects of multifactorial treatments on cognition, anxiety, and depression. Methods Find. Expert Clin. Pharmacol. 2007, 29, 1–91.
  46. Sabbagh, M.N.; Malek-Ahmadi, M.; Dugger, B.N.; Lee, K.; Sue, L.I.; Serrano, G.; Walker, D.G.; Davis, K.; Jacobson, S.A.; Beach, T.G. The influence of Apolipoprotein E genotype on regional pathology in Alzheimer’s disease. BMC Neurol. 2013, 13, 44.
  47. Kennedy, R.E.; Cutter, G.R.; Schneider, L.S. Effect of APOE genotype status on targeted clinical trials outcomes and efficiency in dementia and mild cognitive impairment resulting from Alzheimer’s disease. Alzheimers Dement. 2014, 10, 349–359.
  48. Reiman, E.M.; Arboleda-Velasquez, J.F.; Quiroz, Y.T.; Huentelman, M.J.; Beach, T.G.; Caselli, R.J.; Chen, Y.; Su, Y.; Myers, A.J.; Hardy, J.; et al. Exceptionally low likelihood of Alzheimer’s dementia in APOE2 homozygotes from a 5000-person neuropathological study. Nat. Commun. 2020, 11, 667.
  49. Cacabelos, R.; Torrellas, C.; López-Muñoz, F. Epigenomics of Alzheimer’s disease. J. Exp. Med. 2014, 6, 75–82.
  50. Cacabelos, R.; Torrellas, C. Epigenetic drug discovery for Alzheimer’s disease. Expert Opin. Drug Discov. 2014, 9, 1059–1086.
  51. Cacabelos, R. (Ed.) Pathoepigenetics: The role of epigenetic biomarkers in disease pathogenesis. In Pharmacoepigenetics, 1st ed.; Academic Press: Oxford, UK, 2019; pp. 139–189.
  52. Qazi, T.J.; Quan, Z.; Mir, A.; Qing, H. Epigenetics in Alzheimer’s Disease: Perspective of DNA Methylation. Mol. Neurobiol. 2018, 55, 1026–1044.
  53. Ciceri, F.; Rotllant, D.; Maes, T. Understanding Epigenetic Alterations in Alzheimer’s and Parkinson’s Disease: Towards Targeted Biomarkers and Therapies. Curr. Pharm. Des. 2017, 23, 839–857.
  54. Swarbrick, S.; Wragg, N.; Ghosh, S.; Stolzing, A. Systematic Review of miRNA as Biomarkers in Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 6156–6167.
  55. Gupta, P.; Bhattacharjee, S.; Sharma, A.R.; Sharma, G.; Lee, S.S.; Chakraborty, C. miRNAs in Alzheimer Disease—A Therapeutic Perspective. Curr. Alzheimer Res. 2017, 14, 1198–1206.
  56. Silvestro, S.; Bramanti, P.; Mazzon, E. Role of miRNAs in Alzheimer’s Disease and Possible Fields of Application. Int. J. Mol. Sci. 2019, 20, 3979.
  57. Zetterberg, H.; Burnham, S.C. Blood-based molecular biomarkers for Alzheimer’s disease. Mol. Brain 2019, 12, 26.
  58. Nunomura, A.; Perry, G. RNA and Oxidative Stress in Alzheimer’s Disease: Focus on microRNAs. Oxidative Med. Cell. Longev. 2020, 2020, 2638130.
  59. Cacabelos, R.; Carril, J.C.; Sanmartín, A.; Cacabelos, P. Pharmacoepigenetic processors: Epigenetic drugs, Drug resistance, Toxicoepigenetics, and Nutriepigenetics. In Pharmacoepigenetics, 1st ed.; Cacabelos, R., Ed.; Academic Press: Oxford, UK, 2019; pp. 191–424.
  60. Cacabelos, R.; Cacabelos, P.; Carril, J.C. Epigenetics and pharmacoepigenetics of age-related neurodegenerative disorders. In Pharmacoepigenetics, 1st ed.; Cacabelos, R., Ed.; Academic Press: Oxford, UK, 2019; pp. 903–950.
  61. Cacabelos, R. Epigenomic networking in drug development: From pathogenic mechanisms to pharmacogenomics. Drug Dev. Res. 2014, 75, 348–365.
  62. Kandimalla, R.; Reddy, P.H. Therapeutics of Neurotransmitters in Alzheimer’s Disease. J. Alzheimers Dis. 2017, 57, 1049–1069.
  63. Chu, L.W. Alzheimer’s disease: Early diagnosis and treatment. Hong Kong Med. J. 2012, 18, 228–237.
  64. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115.
  65. Cacabelos, R.; Cacabelos, N.; Carril, J.C. The role of pharmacogenomics in adverse drug reactions. Expert Rev. Clin. Pharmacol. 2019, 12, 407–442.
  66. Barbe, C.; Jolly, D.; Morrone, I.; Wolak-Thierry, A.; Dramé, M.; Novella, J.L.; Mahmoudi, R. Factors associated with quality of life in patients with Alzheimer’s disease. BMC Geriatr. 2018, 18, 159.
  67. Ettcheto, M.; Olloquequi, J.; Sánchez-López, E.; Busquets, O.; Cano, A.; Manzine, P.R.; Beas-Zarate, C.; Castro-Torres, R.D.; García, M.L.; Bulló, M.; et al. Benzodiazepines and Related Drugs as a Risk Factor in Alzheimer’s Disease Dementia. Front. Aging Neurosci. 2020, 11, 344.
  68. Vidoni, E.D.; Kamat, A.; Gahan, W.P.; Ourso, V.; Woodard, K.; Kerwin, D.R.; Binder, E.F.; Burns, J.M.; Cullum, M.; Hydan, L.S.; et al. Baseline Prevalence of Polypharmacy in Older Hypertensive Study Subjects with Elevated Dementia Risk: Findings from the Risk Reduction for Alzheimer’s Disease Study (rrAD). J. Alzheimers Dis. 2020, 77, 175–182.
  69. Andin, U.; Passant, U.; Gustafson, L.; Englund, E. Alzheimer’s disease (AD) with and without white matter pathology-clinical identification of concurrent cardiovascular disorders. Arch. Gerontol. Geriatr. 2007, 44, 277–286.
  70. Ibrahim, B.; Suppiah, S.; Piersson, A.D.; Razali, R.M.; Mohamad, M.; Hassan, H.A.; Ibrahim, N. Cardiovascular risk factors of Alzheimer’s disease and other neurocognitive disorders in Malaysia. Med. J. Malays. 2021, 76, 291–297.
  71. Contois, J.H.; Anamani, D.E.; Tsongalis, G.J. The underlying molecular mechanism of apolipoprotein E polymorphism: Relationships to lipid disorders, cardiovascular disease, and Alzheimer’s disease. Clin. Lab. Med. 1996, 16, 105–123.
  72. Cacabelos, R.; Fernández-Novoa, L.; Lombardi, V.; Corzo, L.; Pichel, V.; Kubota, Y. Cerebrovascular risk factors in Alzheimer’s disease: Brain hemodynamics and pharmacogenomic implications. Neurol. Res. 2003, 25, 567–580.
  73. Loera-Valencia, R.; Goikolea, J.; Parrado-Fernandez, C.; Merino-Serrais, P.; Maioli, S. Alterations in cholesterol metabolism as a risk factor for developing Alzheimer’s disease: Potential novel targets for treatment. J. Steroid Biochem. Mol. Biol. 2019, 190, 104–114.
  74. Jeong, W.; Lee, H.; Cho, S.; Seo, J. ApoE4-Induced Cholesterol Dysregulation and Its Brain Cell Type-Specific Implications in the Pathogenesis of Alzheimer’s Disease. Mol. Cells 2019, 42, 739–746.
  75. Di Paolo, G.; Kim, T.W. Linking lipids to Alzheimer’s disease: Cholesterol and beyond. Nat. Rev. Neurosci. 2011, 12, 284–296.
  76. Pugazhenthi, S.; Qin, L.; Reddy, P.H. Common neurodegenerative pathways in obesity, diabetes, and Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1037–1045.
  77. Chornenkyy, Y.; Wang, W.X.; Wei, A.; Nelson, P.T. Alzheimer’s disease and type 2 diabetes mellitus are distinct diseases with potential overlapping metabolic dysfunction upstream of observed cognitive decline. Brain Pathol. 2019, 29, 3–17.
  78. Baglietto-Vargas, D.; Shi, J.; Yaeger, D.M.; Ager, R.; LaFerla, F.M. Diabetes and Alzheimer’s disease crosstalk. Neurosci. Biobehav. Rev. 2016, 64, 272–287.
  79. Cacabelos, R. Pharmacogenomics of Cognitive Dysfunction and Neuropsychiatric Disorders in Dementia. Int. J. Mol. Sci. 2020, 21, 3059.
  80. Cacabelos, R. Pharmacogenetic considerations when prescribing cholinesterase inhibitors for the treatment of Alzheimer’s disease. Expert Opin. Drug Metab. Toxicol. 2020, 16, 673–701.
  81. Cacabelos, R. Have there been improvement in Alzheimer’s disease drug discovery over the past 5 years? Exp. Opin. Drug Discov. 2018, 13, 523–538.
  82. Maramai, S.; Benchekroun, M.; Gabr, M.T.; Yahiaoui, S. Multitarget Therapeutic Strategies for Alzheimer’s Disease: Review on Emerging Target Combinations. Biomed Res. Int. 2020, 2020, 5120230.
  83. Cacabelos, R. How plausible is an Alzheimer’s disease vaccine? Expert Opin. Drug Discov. 2020, 15, 1–6.
  84. Cummings, J.; Lee, G.; Ritter, A.; Sabbagh, M.; Zhong, K. Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement. 2020, 6, e12050.
  85. Schenk, D.; Barbour, R.; Dunn, W.; Gordon, G.; Grajeda, H.; Guido, T.; Hu, K.; Huang, J.; Johnson-Wood, K.; Hkan, K.; et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999, 400, 173–177.
  86. Foroutan, N.; Hopkins, R.B.; Tarride, J.E.; Florez, I.D.; Levine, M. Safety and efficacy of active and passive immunotherapy in mild-to-moderate Alzheimer’s disease: A systematic review and network meta-analysis. Clin. Investig. Med. 2019, 42, 53–65.
  87. Janus, C.; Pearson, J.; McLaurin, J.; Mathews, P.M.; Jiang, Y.; Schmidt, S.D.; Chishti, M.A.; Horne, P.; Heslin, D.; French, J.; et al. A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer’s disease. Nature 2000, 408, 979–982.
  88. Herline, K.; Drummond, E.; Wisniewski, T. Recent advancements toward therapeutic vaccines against Alzheimer’s disease. Expert Rev. Vaccines 2018, 17, 707–721.
  89. Novak, P.; Zilka, N.; Zilkova, M.; Kovacech, B.; Shrabana, R.; Ondrus, M.; Fialova, L.; Kontsekova, E.; Otto, M.; Novak, M. AADvac1, an Active Immunotherapy for Alzheimer’s Disease and Non Alzheimer Tauopathies: An Overview of Preclinical and Clinical Development. J. Prev. Alzheimers Dis. 2019, 6, 63–69.
  90. Hoskin, J.L.; Sabbagh, M.N.; Al-Hasan, Y.; Decourt, B. Tau immunotherapies for Alzheimer’s disease. Expert Opin. Investig. Drugs 2019, 28, 545–554.
  91. Carrera, I.; Fernandez-Novoa, L.; Aliev, G.; Vigo, C.; Cacabelos, R. Validating Immunotherapy in Alzheimer’s Disease: The EB101 Vaccine. Curr. Pharm. Des. 2016, 22, 849–858.
  92. Zhang, H.Y.; Zhu, K.; Meng, Y.; Ding, L.; Wang, J.C.; Yin, W.C.; Yan, Y.; Cao, Y.P. Reduction of amyloid beta by Aβ3-10-KLH vaccine also decreases tau pathology in 3×Tg-AD mice. Brain Res. Bull. 2018, 142, 233–240.
  93. Rosenberg, R.N.; Fu, M.; Lambracht-Washington, D. Active full-length DNA Aβ42 immunization in 3xTg-AD mice reduces not only amyloid deposition but also tau pathology. Alzheimers Res. Ther. 2018, 10, 115.
  94. Oddo, S.; Billings, L.; Kesslak, J.P.; Cribbs, D.H.; LaFerla, F.M. Abeta immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron 2004, 43, 321–332.
  95. Lalli, G.; Schott, J.M.; Hardy, J.; De Strooper, B. Aducanumab: A new phase in therapeutic development for Alzheimer’s disease? EMBO Mol. Med. 2021, 13, e14781.
  96. Mukhopadhyay, S.; Banerjee, D. A Primer on the Evolution of Aducanumab: The First Antibody Approved for Treatment of Alzheimer’s Disease. J. Alzheimers Dis. 2021, 83, 1537–1552.
  97. Knopman, D.S.; Jones, D.T.; Greicius, M.D. Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE trials as reported by Biogen, December 2019. Alzheimers Dement. 2021, 17, 696–701.
  98. Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016, 537, 50–56.
  99. Ferrero, J.; Williams, L.; Stella, H.; Leitermann, K.; Mikulskis, A.; O’Gorman, J.; Sevigny, J. First-in-human, double-blind, placebo-controlled, single-dose escalation study of aducanumab (BIIB037) in mild-to-moderate Alzheimer’s disease. Alzheimers Dement. 2016, 2, 169–176.
  100. Tolar, M.; Abushakra, S.; Hey, J.A.; Porsteinsson, A.; Sabbagh, M. Aducanumab, gantenerumab, BAN2401, and ALZ-801-the first wave of amyloid-targeting drugs for Alzheimer’s disease with potential for near term approval. Alzheimers Res. Ther. 2020, 12, 95.
  101. Van de Vrede, L.; Gibbs, D.M.; Koestler, M.; La Joie, R.; Ljubenkov, P.A.; Provost, K.; Soleimani-Meiggoni, D.; Strom, A.; Tsoy, E.; Rabinovici, G.D.; et al. Symptomatic amyloid-related imaging abnormalities in an APOE ε4/ε4 patient treated with aducanumab. Alzheimers Dement. 2020, 12, e12101.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 208
Revisions: 2 times (View History)
Update Date: 13 Apr 2022
1000/1000