Beta Amyloid Imaging with Positron Emission Tomography (PET) for Alzheimer's Disease - CAM 60155

Description:
Alzheimer disease (AD) is a fatal neurodegenerative disease that causes progressive loss in memory, language, and thinking, with the eventual loss of ability to perform social and functional activities in daily life. Because clinical diagnosis can be difficult, particularly early in the course of the disease or with atypical dementia, there has been considerable interest in developing biomarkers for AD that can be imaged through positron emission tomography (PET).

Three radioactive tracers (florbetapir fluorine 18, florbetaben fluorine 18, flutemetamol fluorine 18) that bind to amyloid beta and can be detected in vivo with PET have been approved by the U.S. Food and Drug Administration (FDA) for amyloid beta imaging in patients who are being evaluated for cognitive decline.. Amyloid beta plaque PET imaging is proposed as an adjunct to the clinical diagnosis of AD and as a component of identifying patients for amyloid beta plaque-targeting
therapy.

 Fluorine 18 fluorodeoxyglucose PET (FDG-PET) quantifies brain function by measuring glucose levels. FDG-PET is proposed as a method to distinguish AD from other dementias through identifying distinct regions of hypometabolism. 

Additional Information
This policy does not currently include tau PET imaging.

See Policy 50138 for a detailed review of aducanamab for Alzheimer disease.

FDG-PET for individuals with suspected AD, previously included in Policy 60106, was added to this policy in October 2021.

Background  
ALZHEIMER DISEASE
Diagnosis
Alzheimer disease (AD) is a fatal neurodegenerative disease that causes progressive loss in memory, language, and thinking, with the eventual loss of ability to perform social and functional activities in daily life. Survival after a diagnosis of dementia due to AD generally ranges between 4 and 8 years; however, life expectancy can be influenced by other factors, such as comorbid medical conditions. It is estimated that 6.2 million Americans aged 65 and older are currently living with AD dementia, and the number is projected to reach over 12 million by 2050.1

The pathologic hallmarks of AD are extracellular deposits of amyloid beta, referred to as amyloid plaques, and intracellular aggregates of hyperphosphorylated tau in the form of neurofibrillary tangles. There are different forms of amyloid such as plaques, oligomers, and monomers, and the roles of these different forms and how specifically they are pathophysiologically associated with AD is not well understood. Generally referred to as “amyloid hypothesis,” it is believed that aggregation of amyloid beta oligomers in the brain leads to amyloid plaques and is thought to be the primary driver of the disease process. These changes in the brain result in widespread neurodegeneration and cell death, and ultimately cause the clinical signs and symptoms of dementia.2,3

Role of Positron Emission Tomography
Because clinical diagnosis can be difficult, particularly early in the course of the disease or with atypical dementia, there has been considerable interest in developing biomarkers for AD that can be imaged through positron emission tomography (PET). These biomarkers include amyloid beta plaque and glucose metabolism in the brain. PET images biochemical and physiologic functions by measuring concentrations of radioactive chemicals that have been partially metabolized in a particular region of the body. Radiopharmaceuticals used for PET imaging may be generated in a cyclotron or nuclear generator and introduced into the body by intravenous injection.

Demonstration of amyloid beta plaque is a requirement for the diagnosis of definite AD, but may also be present in individuals without dementia, in patients with mild or subjective cognitive impairment who may or may not progress to dementia, and in patients with other types of dementia. Conversely, it may be absent in a substantial proportion of patients with clinical features of AD.4,5,6

18-F FDG PET quantifies brain function by measuring glucose levels. Through identifying distinct regions of hypometabolism, FDG-PET is proposed as a method to distinguish AD from other dementias, especially in patients with atypical presentations (e.g., younger age).7

PET imaging in patients with mild cognitive impairment (MCI) or dementia is intended to provide a more accurate diagnosis earlier in the disease course than clinical diagnosis alone, resulting in earlier, appropriately targeted treatment and other management approaches.

Treatment Options
Current treatment goals for patients with AD are often directed to maintain quality of life, treat cognitive symptoms, and manage behavioral and psychological symptoms of dementia. Treatment remains largely supportive, including creation and implementation of individualized dementia care plans, caregiver education and support, care navigation, care coordination, and referral to community-based organizations for services (e.g., adult day care, caregiver training, etc).8 Nonpharmacologic
treatments include physical activity,9,10 as well as behavioral strategies to ameliorate neuropsychiatric symptoms (e.g., agitation, delusions,
disinhibition), and problem behaviors (e.g., resistance to care, hoarding, obsessive-compulsive behaviors).11

FDA-approved drugs for AD symptoms include the cholinesterase inhibitors donepezil, rivastigmine, and galantamine and the N‐methyl‐D‐aspartate antagonist,
memantine. These drugs, either alone or in combination, focus on managing cognitive and functional symptoms of the disease and have not been shown to alter
disease trajectory. The evidence for efficacy is limited and these agents are associated with significant side effects.11,12

In June 2021, aducanumab (Aduhelm; Biogen) was approved by the FDA for treatment of AD. This indication was approved under accelerated approval based on
reduction in amyloid beta plaques observed in patients treated with aducanumab. Continued approval for this indication is contingent upon verification of clinical
benefit in confirmatory trial(s).The FDA, under the accelerated approval regulations (21 CFR 601.41), requires that Biogen conduct a randomized, controlled trial to
evaluate the efficacy of aducanumab compared to an appropriate control for the treatment of AD. The trial should be of sufficient duration to observe changes on an acceptable endpoint in the patient population enrolled in the trial. The expected date of trial completion is August 2029 and final report submission to the FDA by February 2030.

In July 2021, FDA amended the approved label to emphasize the disease stages studied in the clinical trials. The amended label states, "Treatment with
aducanumab should be initiated in patients with MCI or mild dementia stage of disease, the population in which treatment was initiated in clinical trials. There are no safety or effectiveness data on initiating treatment at earlier or later stages of the disease than were studied."

Regulatory Status
Radiopharmaceuticals for PET Imaging
PET radiopharmaceuticals have been evaluated and approved as drugs by the FDA for use as diagnostic imaging agents. These radiopharmaceuticals are approved
for specific conditions.

Amyvid™, Vizamyl™, and Neuraceq™ (Table 1) are approved by the FDA "for PET imaging of the brain to estimate amyloid beta neuritic plaque density in adult
patients with cognitive impairment who are being evaluated for AD and other causes of cognitive decline."13,14,15

In 1994, the fludeoxyglucose (FDG) F18 radiotracer was originally approved by the FDA through the New Drug Application (NDA) process (NDA20306). The original
indication was for "the identification of regions of abnormal glucose metabolism associated with foci of epileptic seizures." Added indications in 2000 were for
"Assessment of glucose metabolism to assist in the evaluation of malignancy …" and "Assessment of patients with coronary artery disease and left ventricular
dysfunction…." FDA approval of FDG does not include the evaluation of patients with cognitive decline. Multiple manufacturers have approved NDAs for FDG.

The prescribing information for all 3 agents used for amyloid beta imaging states:

  • The objective of amyloid beta image interpretation "is to estimate beta-amyloid neuritic plaque density in brain gray matter, not to make a clinical diagnosis."

  • A positive amyloid beta scan "does not establish the diagnosis of AD or other cognitive disorder."

  • A negative amyloid beta scan "indicates sparse to no neuritic plaques, and is inconsistent with a neuropathologic diagnosis of AD at the time of image acquisition; a negative scan result reduces the likelihood that a patient's cognitive impairment is due to AD."

  • Florbetapir, florbetaben, and flutemetamol are not intended for use in "predicting development of dementia or other neurological condition" or for "monitoring responses to therapies."

Table 1. Radioactive Tracers Approved by the FDA for Amyloid Beta PET Imaging in Patents With Cognitive Impairment

Agent

Trade Name

Manufacturer

NDA

Approved

florbetapir F18

Amyvid™

Avid Radiopharmaceuticals (subsidiary of Eli Lilly)

202008

2012

flutemetamol F18

Vizamyl™

GE Healthcare

203137

2013

florbetaben F18

Neuraceq™

Piramal Life Sciences

204677

2014

NDA: new drug application.

Related Policies
20414 Biochemical Markers of Alzheimer’s Disease
60106 Miscellaneous (Noncardiac, Nononcologic) Applications of Positron Emission Tomography
60154 Dopamine Transporter Imaging with Single-Photon Emission Computed Tomography (DAT-SPECT)

Policy:
Amyloid beta imaging with positron emission tomography (PET) to predict conversion to Alzheimer disease is investigational/unproven therefore is considered NOT MEDICALLY NECESSARY.

Amyloid beta imaging with PET as an adjunct to clinical diagnosis in patients with dementia is investigational/unproven therefore is considered NOT MEDICALLY NECESSARY.

Amyloid beta imaging with PET to select patients with mild cognitive impairment or mild dementia due to Alzheimer disease for amyloid beta targeting plaquetherapy
is investigational/unproven therefore is considered NOT MEDICALLY NECESSARY.

Amyloid beta imaging with PET to evaluate patients with mild cognitive impairment or mild dementia due to Alzheimer disease for continuation of amyloid beta
plaque-targeting therapy is investigational/unproven therefore is considered NOT MEDICALLY NECESSARY.

PET Imaging with fluorine 18 fluorodeoxyglucose (FDG-PET) as an adjunct to clinical diagnosis in patients with dementia is investigational/unproven therefore is considered NOT MEDICALLY NECESSARY.

All other uses of amyloid beta imaging with PET are investigational/unproven therefore is considered NOT MEDICALLY NECESSARY

Policy Guidelines
None    

Benefit Application
BlueCard/National Account Issues
State or federal mandates (e.g., FEP) may dictate that certain U.S. Food and Drug Administration (FDA)-approved devices, drugs or biologics may not be considered investigational, and thus these devices may be assessed only on the basis of their medical necessity.

Rationale
Evidence reviews assess whether a medical test is clinically useful. A useful test provides information to make a clinical management decision that improves the net health outcome. That is, the balance of benefits and harms is better when the test is used to manage the condition than when another test or no test is used to manage the condition.

The first step in assessing a medical test is to formulate the clinical context and purpose of the test. The test must be technically reliable, clinically valid, and clinically useful for that purpose. Evidence reviews assess the evidence on whether a test is clinically valid and clinically useful. Technical reliability is outside the scope of these reviews, and credible information on technical reliability is available from other sources.

Amyloid Beta Imaging With Positron Emission Tomography to Predict Conversion to Alzheimer Disease in Patients with Mild Cognitive Impairment
Clinical Context and Test Purpose

The purpose of amyloid beta imaging with positron emission tomography (PET) in patients who have mild cognitive impairment (MCI) is to determine the amyloid beta burden and the likelihood of developing Alzheimer disease (AD).

The question addressed in this evidence review is: Does amyloid beta PET imaging improve the net health outcome in patients with MCI?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with MCI.

Interventions
The intervention of interest is amyloid beta imaging using a commercially available PET tracer (florbetapir F18, florbetaben F18, or flutemetamol F18).

Comparators
The criterion standard for the development of AD is postmortem neuropathologic examination. In the absence of comparisons with the criterion standard, a clinical follow-up to determine conversion to probable AD may be used to evaluate the diagnostic performance of amyloid beta imaging with PET.

Outcomes
The general outcomes of interest are test validity, symptoms, change in disease status, functional outcomes, health status measures, and quality of life.

Beneficial outcomes resulting from a true test result: The current clinical purpose of testing for amyloid beta plaque density would be to improve the prediction of conversion to AD.

Harmful outcomes resulting from a false test result: a false-positive test may result in failure to undergo additional testing for other causes of cognitive decline such as depression, obstructive sleep apnea, or drug-induced cognitive impairment; a false-negative test may lead to additional unnecessary tests (e.g., polysomnography) to evaluate these other potential causes of cognitive impairment.

Direct harms of the test: although generally well tolerated, there is a chance of adverse reactions to the radioligand.

Diagnostic accuracy can only be confirmed at autopsy or after several years of follow-up to monitor progression (or lack of progression) of disease. Conversion of MCI to AD has been shown to occur at a rate of 5% to 10% per year with conversion to any dementia at a rate of about 20% per year. Conversion of MCI to AD typically occurs in 2 to 3 years but may be as long as 8 years. Direct evidence of an improvement in health outcomes would be observed in years.

Study Selection Criteria
For the evaluation of the clinical validity of amyloid beta imaging, studies that meet the following eligibility criteria were considered:

  • Reported on the accuracy of the marketed version of the technology
  • Included a suitable reference standard (conversion to probable AD)
  • Patient/sample clinical characteristics were described
  • Patient/sample selection criteria were described

Studies were excluded from the evaluation of the clinical validity of the amyloid beta test if they did not use the marketed version of the test, did not include information needed to calculate performance characteristics, did not use an appropriate reference standard or the reference standard was unclear, did not adequately describe the patient characteristics, or did not adequately describe patient selection criteria.

Clinically Valid
A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Review of Evidence
Systematic Reviews

Martinez et al. conducted 3 Cochrane systematic reviews of the diagnostic accuracy of PET scan using florbetapir, florbetaben, and flutemetamol to detect people with MCI who will clinically progress to AD or other forms of dementia at follow-up (Table 2). The reviews included 1 study of florbetaben,18 2 studies of flutemetamol ,19,20 and 3 studies of florbetapir.21,22,23

Study characteristics, results, and methodological limitations are summarized in Table 2. The reviewers concluded that due to limited data available, varying sensitivity and specificity, and risk of bias limiting confidence in the conclusions, routine use of the technology could not be recommended.

Table 2. Systematic Reviews of the Diagnostic Accuracy of PET Imaging to Predict Progression to AD

Study Literature Search Dates Populations Interventions Studies Included
(N )
Study Designs Included Reference Standard Follow-up Duration Results- Progression from MCI to AD Methodological Limitations of Included Studies
                By Visual Assessment By SUVR > 1.45  
Martinez et al. (2017a) ]24 1946 to May 2017 Participants recruited and clinically classified as having MCI at time of performing the test. Diagnosis of MCI established using the Petersen criteria or revised Petersen criteria. 18F PET with florbetaben 1 (N = 45) Longitudinal studies with prospectively defined cohorts with any accepted definition of MCI at time of performing the scan and a reference standard Progression to the target conditions evaluated by a physician with expertise in the dementia field 2 to 4 years Sensitivity 100% (95% CI, 84% to 100%)

Specificity 83% (95% CI, 63% to 95%)
Sensitivity 100% (95% CI, 84% to 100%)

Specificity 88% (95% CI, 68% to 97%)
High risk of bias: Lack of information about participant selection; reference standard was made with knowledge of the medical studies and medical records; conflicts of interest
Martinez et al. (2017b) 25 Same as above Same as above 18F PET with flutemetamol 2 (N = 243) Same as above Same as above 3 years Sensitivity 64% (95% CI, 53 to 75)

Specificity 69% (95% CI, 60 to 76)
Sensitivity 89% (95% CI, 52% to 100%)

Specificity 80% (95% CI, 44% to 97%)
Uncertainty about the clinical diagnosis of AD; not a clear definition of a positive index test in one study; the reference standard in one study was not explicitly described; potential conflict of interest with the company that produced the tracer in both studies
Martinez et al. (2017c)26 Same as above Same as above 18F PET with florbetapir 2 (N = 448) Same as above Same as above 1.6 years and 3 years Follow-up from 2 to < 4 years:
Sensitivity 67% (95% CI, 30% to 93%)

Specificity 71% (95% CI, 54% to 85%)

Follow-up from 1 to < 2 years:
Sensitivity 89% (95% CI, 78% to 95%)

Specificity 58% (95% CI, 53% to 64%)
Follow-up from 1 to < 2 years (n = 401, 1 study):
Sensitivity 87% (95% CI, 76% to 94%)

Specificity of 51% (95% CI, 45% to 56%)

 
Uncertainty about the clinical diagnosis of AD; lack of information regarding the selection of participants; not clear if the reference standard interpretation was made without knowledge of the PET scan results in 2 studies; potential conflict of interest with the company that produced the tracer

AD: Alzheimer disease; CI: confidence interval; MCI: mild cognitive impairment; PET: positron emission tomography; SUVR: standardized uptake value ratio.

Nonrandomized Studies
Additional studies evaluating conversion from MCI to probable AD have been published following the Cochrane systematic reviews.27,28,29,30 The largest prospective study was reported by Wolk et al. (2018) (Tables 3 and 4).31, The hazard ratio for conversion to probable AD at 3 years in patients with a baseline positive amyloid beta PET scan was 2.51 (95% confidence interval [CI], 1.57 to 3.99; p < .001), increasing to 8.45 when low hippocampal volume and poorer cognitive status was added to the model.

Table 3. Study Characteristics for Patients With MCI

Study Study Population Design Reference Standard Threshold for Positive Index Test Timing of Reference and Index Tests Blinding of Assessors
Wolk et al. (2018)31 232 patients ≥ 55 years of age with MCI and no vascular, traumatic, or inflammatory causes Prospective Independent clinical adjudication committee Visual rating as amyloid beta+ (n = 98) Every 6 months for 3 years Yes

amyloid beta+: positive amyloid beta PET scan; MCI: mild cognitive impairment.

Table 4. Clinical Validity for Patients With MCI

Study Initial N Final N Conversion of amyloid beta+ to probable AD Conversion of amyloid beta- to probable AD HR (95% CI); p Clinical Validity, %
            Sensitivity Specificity PPV NPV
Wolk et al. (2018)31 232 224 54% of 97 23% of 127 2.51 (1.57 to 3.99); p < .001 64 69 54 77

AD: Alzheimer disease; amyloid beta+: positive amyloid beta PET scan; amyloid beta-: negative amyloid beta PET scan; CI: confidence interval; HR: hazard ratio; NPV: negative predictive value; PPV: positive predictive value.

Study limitations are summarized in Tables 5 and 6.

Table 5. Study Relevance Limitations

Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-Upe
Wolk et al. (2018)31   2. Used a majority rating of 5 readers instead of a single reviewer      

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.
a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.
b Intervention key: 1. Classification thresholds not defined; 2. Version used unclear; 3. Not intervention of interest.
c Comparator key: 1. Classification thresholds not defined; 2. Not compared to credible reference standard; 3. Not compared to other tests in use for same purpose.
d Outcomes key: 1. Study does not directly assess a key health outcome; 2. Evidence chain or decision model not explicated; 3. Key clinical validity outcomes not reported (sensitivity, specificity and predictive values); 4. Reclassification of diagnostic or risk categories not reported; 5. Adverse events of the test not described (excluding minor discomforts and inconvenience of venipuncture or noninvasive tests).
e Follow-Up key: 1. Follow-up duration not sufficient with respect to natural history of disease (true-positives, true-negatives, false-positives, false-negatives cannot be determined).

Table 6. Study Design and Conduct Limitations

Study Selectiona Blindingb Delivery of Testc Selective Reportingd Data Completenesse Statisticalf
Wolk et al. (2018)31           1. CIs not reported

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.
CI: confidence interval. 
a Selection key: 1. Selection not described; 2. Selection not random or consecutive (i.e., convenience).
b Blinding key: 1. Not blinded to results of reference or other comparator tests.
c Test Delivery key: 1. Timing of delivery of index or reference test not described; 2. Timing of index and comparator tests not same; 3. Procedure for interpreting tests not described; 4. Expertise of evaluators not described.
d Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.
e Data Completeness key: 1. Inadequate description of indeterminate and missing samples; 2. High number of samples excluded; 3. High loss to follow-up or missing data.
f Statistical key: 1. Confidence intervals and/or p values not reported; 2. Comparison with other tests not reported.

Clinically Useful
A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Review of Evidence
Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from randomized controlled trials (RCTs).

A multicenter RCT by Pontecorvo et al. (2017) randomized 342 patients with MCI and 276 patients with AD and greater than 15% uncertainty in the diagnosis to immediate or delayed reporting of amyloid beta PET results to their physicians (Table 7).32 Changes in diagnosis and patient management are shown in Table 8. Health outcomes were evaluated at 1 year, but there were no statistical differences between groups for cognitive performance, function, or quality of life. However, due to the exploratory nature of the analysis and lack of power, it remains uncertain whether the changes in management affected health outcomes (Tables 9 and 10). The progression of cognitive change did not differ between patients with MCI who had a positive amyloid beta PET scan or a negative amyloid beta PET scan (p = .568) over the year of the study.

Table 7. Summary of Key RCT Characteristics

Study Countries Sites Dates Participants Interventions
          Active Comparator
Pontecorvo et al. (2017)32 U.S., EU 60 2012 – 2015 618 patients 50 – 90 years of age with MCI (n = 342) or dementia (n = 276) Physicians had immediate access to amyloid beta PET results (n = 308) Physicians had delayed (12 months) access to amyloid beta PET results (n = 310)

EU: European Union; MCI: mild cognitive impairment; PET: positron emission tomography; RCT: randomized controlled trial.

Table 8. Summary of Key RCT Results

Study Countries Sites Dates Participants Interventions
          Active Comparator
Pontecorvo et al. (2017)32 U.S., EU 60 2012 – 2015 618 patients 50 – 90 years of age with MCI (n = 342) or dementia (n = 276) Physicians had immediate access to amyloid beta PET results (n = 308) Physicians had delayed (12 months) access to amyloid beta PET results (n = 310)

CI: confidence interval; Diff: difference; NNT: number needed to treat; NR: not reported; OR: odds ratio; RCT: randomized controlled trial.

Notable limitations identified in each study are shown in Tables 9 and 10.

Table 9. Study Relevance Limitations

Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-Upe
Pontecorvo et al. (2017)32 1. Results did not distinguish between patients with MCI or AD     1. Health outcomes were exploratory

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.
AD: Alzheimer disease; MCI: mild cognitive impairment.
a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.
b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.
c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.
d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.
e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.

Table 10. Study Design and Conduct Limitations

Study Allocationa Blindingb Selective Reportingc Data Completenessd Powere Statisticalf
Pontecorvo et al. (2017)32   1, 2. Not blinded to treatment or outcome assessment   6. Not intention-to-treat and number of unclear PET scans is not reported 3. Not powered for health outcomes 3. CIs and p values not reported for health outcomes

Chain of Evidence
Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

Because the clinical validity of amyloid beta PET has not been established, a chain of evidence supporting its clinical utility for this indication cannot be constructed.

Section Summary: Amyloid Beta Imaging With Positron Emission Tomography to Predict Conversion to Alzheimer Disease in Patients with Mild Cognitive Impairment
One proposed use for amyloid beta imaging is to determine which patients with MCI have a likelihood of converting to AD. Studies have been conducted to evaluate the diagnostic accuracy of amyloid beta PET in patients with MCI, using conversion to probable AD as a reference standard. Systematic reviews of these studies have concluded that limited data, varying sensitivity and specificity, and risk of bias limited confidence in conclusions. In a more recent prospective study of 224 individuals with MCI, the hazard ratio for conversion to probable AD at 3 years in patients with a baseline positive amyloid beta PET scan was 2.51 (95% CI, 1.57 to 3.99; p < .001), with a negative predictive value of 77%. The clinical utility of this is uncertain. Direct evidence on clinical utility is limited. One RCT reported on changes in diagnosis and management but did not find evidence that health outcomes (cognition, function, quality of life) were improved by testing. A major limitation of this study is that the evaluation of health outcomes was exploratory and not sufficiently powered. No trials have been identified that reported whether changes in diagnosis are more accurate.

Amyloid Beta Imaging With Positron Emission Tomography as an Adjunct to Clinical Assessment to Diagnose Alzheimer Disease in Patients with Dementia
Clinical Context and Test Purpose

One proposed use of amyloid beta PET imaging in patients with dementia is to determine the amyloid beta burden to aid a differential diagnosis between AD and non-AD causes of cognitive impairment and guide appropriate treatment and/or further testing. Amyloid PET may be positive in cognitively normal subjects who do not develop AD and in patients with other forms of non-AD dementia; therefore, the value of beta PET imaging would be to rule out a diagnosis of AD in patients with dementia. A negative amyloid beta PET scan could lead to further diagnostic testing to determine the etiology of dementia and/or avoidance of anti-Alzheimer medications that would be unnecessary. U.S. Food and Drug Administration (FDA)-approved drugs for AD symptoms include cholinesterase inhibitors donepezil, rivastigmine, and galantamine, the N‐methyl‐D‐aspartate antagonist, memantine, and the amyloid beta targeting therapy, aducanumab. Cholinesterase inhibitors are indicated in mild, moderate, and severe AD, while memantine is approved for moderate-to-severe AD. These drugs, either alone or in combination, focus on managing cognitive and functional symptoms of the disease and have not been shown to alter disease trajectory. The evidence for efficacy is limited and these agents are associated with significant side effects.

The question addressed in this evidence review is: Does amyloid beta PET imaging improve the net health outcome in patients with dementia being assessed for AD?

The following PICO was used to select literature to inform this review.

Populations
The population of interest is patients with dementia.

Interventions
The intervention of interest is amyloid beta imaging using a commercially available PET tracer (florbetapir F18, florbetaben F18, or flutemetamol F18).

Comparators
The criterion standard for the diagnosis of AD is postmortem histopathologic examination. In the absence of comparisons with the criterion standard, long-term clinical follow-up may be used to evaluate the diagnostic performance of amyloid beta PET imaging.

Outcomes
The general outcomes of interest are test validity, symptoms, change in disease status, functional outcomes, health status measures, and quality of life.

Beneficial outcomes resulting from a true test result: improvement in cognition from acetylcholinesterase inhibitors or avoiding side effects from unnecessary treatment with acetylcholinesterase inhibitors; identification and appropriate treatment of non-AD causes of dementia.

Harmful outcomes resulting from a false test result: side effects of incorrect or unnecessary treatment; not receiving correct treatment or failing to undergo additional testing such as formal neuropsychological testing and functional neuroimaging studies (e.g., single-photon emission computed tomography [SPECT], perfusion magnetic resonance imaging, or fluorine 18 fluorodeoxyglucose [FDG] PET) that evaluate areas of low metabolism or hypoperfusion and can help to distinguish AD from other causes of dementia.

Direct harms of the test: although generally well tolerated, there is a chance of adverse reactions to the radioligand.

Diagnostic accuracy can only be confirmed at autopsy or after a minimum of 3 years to monitor progression (or lack of progression) of disease. Direct evidence of an immediate effect of therapy is observable after 2 months of treatment with acetylcholinesterase inhibitors or memantine.

Study Selection Criteria
For the evaluation of the clinical validity of amyloid beta imaging for suspected AD, studies that meet the following eligibility criteria were considered:

  • Reported on the accuracy of the marketed version of the technology
  • Included a suitable reference standard (postmortem histopathologic confirmation or clinical follow-up)
  • Patient/sample clinical characteristics were described
  • Patient/sample selection criteria were described

Studies were excluded from the evaluation of the clinical validity of the test if they did not use the marketed version of the test, did not include information needed to calculate performance characteristics, did not use an appropriate reference standard or the reference standard was unclear, did not adequately describe the patient characteristics, or did not adequately describe patient selection criteria.

Clinically Valid
A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Review of Evidence
Nonrandomized Trials

A number of studies have demonstrated the reliability of florbetapir, florbetaben, and flutemetamol to detect amyloid beta in patients with an established diagnosis of AD compared with non-AD dementia or non-affected individuals.33,34,35,36,37,38,39 In some studies, autopsy results were available to confirm the accuracy of the tracers to determine amyloid beta levels (Table 11). These studies did not correlate amyloid beta PET scan results with a histopathologic diagnosis of AD. Further, these studies do not establish clinical validity in the intended use population, that is patients with suspected AD with an unclear or atypical presentation.

Table 11. Trial Results Using Amyloid Beta Plaque on Postmortem Histology as the Reference Standard

Study n Clinical Diagnosis Interval From Imaging Readers Sensitivity (95% CI or Range), % Specificity (95% CI or Range), %
Sabri et al. (2015)36 florbetaben 74
  • AD
  • Non-AD dementia
  • Dementia with Lewy body
  • No evidence of dementia
11 monthsa 3 readers 89 (81 to 98) 92 (82 to 100)
Curtis et al. (2015)37; Salloway et al. (2017)38 flutemetamol 106 End-of-life cohort 7.5 monthsa Majority of 5 readers 86 to 92b 86 to 100 b
Clark et al. (2011, 2012)33,34 florbetapir 59 End-of-life cohort ≤ 24 months Majority of 5 readers 92 (78 to 98) 100 (80 to 100)
Summary     7.5 to 24 months 3 to 5 readers 86 to 93 86 to 100

AD: Alzheimer disease; CI: confidence interval.
a Mean.
b Varied by criteria amyloid beta threshold.

Bao et al. (2021) reported on a study of PET amyloid imaging in 109 consecutive patients referred to a memory clinic in Hong Kong.40 Subjects underwent clinical assessment and the local version of the Montreal Cognitive Assessment. The mean (standard deviation [SD]) composite standardized uptake value ratio (SUVR) values for patients with a diagnosis of subjective cognitive decline, MCI, AD, and non-AD dementia were 0.50 (0.80), 0.53 (0.16), 0.76 (0.10), and 0.56 (0.16), respectively. With adjustment for age and sex, AD had significantly higher global amyloid beta retention than subjective cognitive decline (p < .0001), MCI (p < .0001), and other dementias (p < .001), while the remaining 3 groups showed no significant difference. Based on the established threshold (SUVR of 0.62) used for differentiating positive and negative scans in global binding, approximately 28% of MCI subjects had a positive global amyloid beta burden, while 91% of AD and 31% of other dementia subjects had a positive PET scan. The authors concluded that quantitative global and regional amyloid beta binding by 18F-flutemetamol PET could be used to discriminate between AD and MCI with 100% sensitivity, 69% specificity, and 79% accuracy.

Clinically Useful
A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Review of Evidence
Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

In the trial by Pontecorvo et al. (2017; discussed above), 342 patients with MCI and 276 patients with dementia were randomized to immediate or delayed reporting of amyloid beta PET results to their physicians (Table 7).32 Changes in diagnosis and patient management are shown in Table 8. Prescription of acetylcholinesterase inhibitors decreased by 8%. The progression of cognitive change did not differ between positive amyloid beta and negative amyloid beta patients with suspected AD (p = .763) during the year of follow-up. Due to the lack of power, it remains uncertain whether the changes in management improved health outcomes (Tables 9 and 10).

A number of multicenter studies have reported changes in diagnosis and patient management following amyloid beta PET imaging.41,42,43,44,45,46 The largest prospective study to date is the Imaging Dementia — Evidence for Amyloid Scanning study, which assessed the association between amyloid beta PET imaging and subsequent changes in management among 11,409 Medicare recipients.46 The primary endpoint was change in management between the pre- and post-PET visits, as assessed by a composite outcome that included AD drug therapy, other drug therapy, and counseling about safety and future planning. Changes between the pre-PET and post-PET composite management endpoint occurred in 60.2% (95% CI, 59.1% to 61.4%) of patients with MCI and 63.5% (95% CI, 62.1% to 64.9%) of patients with dementia. Physicians reported that PET results contributed substantially to the post-PET management plan in 85.2% of instances in which a change was made, and their diagnostic confidence in the uncertain range decreased from 72.4% to 16.2% at the post-PET visit. One limitation of this study is that participants were mainly non-Hispanic White patients, and thus were not reflective of the general population of Medicare beneficiaries or the US population.

In another recent study reported by Leuzy et al. (2019), 207 patients with an initial diagnosis of MCI (63%), AD (20%), or subjective cognitive decline (2%) received amyloid beta PET imaging due to diagnostic uncertainty.45 Overall, amyloid beta PET led to a significant change in diagnosis (92 patients; 44%). The highest percentage change in diagnosis was observed in those with MCI (67 patients; 51%). The outcome of imaging led to more patients receiving treatment with cholinesterase inhibitors, from 34 patients prior to imaging to 109 after imaging. Of the 109 patients receiving cholinesterase inhibitor treatment, 93 (85%) were amyloid-positive. Treatment was discontinued following imaging in 1 amyloid-negative patient with MCI due to cholinergic side effects.

One potential use of amyloid beta PET imaging is to rule out AD, but studies have reported that the most common management change in response to an imaging result was to increase the use of medications to treat symptoms of AD. There were very few instances reported in which medication was discontinued or other interventions were avoided based on a negative PET result. Additionally, none of the studies evaluated whether changes in management improved patient health outcomes. It cannot be determined from these studies whether the revised diagnoses were correct, and without longer follow-up, the effect of the management changes on health outcomes is uncertain.

Section Summary: Amyloid Beta Imaging With Positron Emission Tomography as an Adjunct to Clinical Assessment to Diagnose Alzheimer Disease in Patients with Dementia
Amyloid beta PET is proposed as a way to rule out AD in patients with an early or otherwise atypical presentation of dementia. Amyloid beta plaque is only one of several markers of AD on histopathology but is necessary for a diagnosis of AD. A negative amyloid beta PET scan would, therefore, in theory, be associated with a lower likelihood of AD. Most studies evaluating the diagnostic accuracy of amyloid beta PET in patients with dementia have been conducted in patients at the end of life. Additional, well-designed studies in patients with possible AD are needed. Direct evidence on clinical utility (i.e., improvement in net health outcomes resulting from testing) is lacking. Studies have reported a change in diagnosis and change in management, but there is no evidence of an effect of amyloid beta PET on health outcomes. One potential use of amyloid beta PET imaging is to rule out AD, however, there were very few instances reported in which medication was discontinued or other interventions were avoided based on a negative PET result. The single RCT identified had insufficient power to determine the effect of amyloid beta imaging on health outcomes (i.e., quality of life, symptoms, function).

Amyloid Beta Imaging With Positron Emission Tomography to Select Patients for Targeting Therapy
Clinical Context and Test Purpose

The purpose of amyloid beta imaging with PET in individuals with a clinical diagnosis of MCI or mild dementia due to AD is to guide a decision about initiation of amyloid beta plaque-targeting therapy. The test is intended to exclude patients with clinically diagnosed MCI/AD that are not amyloid positive, and to select for treatment those amyloid positive subjects that are potentially able to benefit from treatment.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with a clinical diagnosis of MCI or mild dementia who are being considered for an FDA-approved amyloid beta plaque-targeting therapy.

The pathophysiological changes and clinical manifestations of AD are progressive and occur along a continuum, and accumulation of amyloid beta may begin 20 years or more before symptoms arise.47 The National Institute on Aging and the Alzheimer’s Association have created a “numeric clinical staging scheme” (Table 12) that avoids traditional syndromal labels and is applicable for only those in the Alzheimer continuum. This staging scheme reflects the sequential evolution of AD from an initial stage characterized by the appearance of abnormal AD biomarkers in asymptomatic individuals. As biomarker abnormalities progress, the earliest subtle symptoms become detectable. Further progression of biomarker abnormalities is accompanied by progressive worsening of cognitive symptoms, culminating in dementia. This numeric cognitive staging scheme is not designed to be used in a clinical setting but to be used for interventional trials such as those of aducanumab. The phase 3 RCTs for aducanumab were stratified to include 80% of stage 3 patients and 20% of stage 4 patients. This numeric staging scheme is very similar to the categorical system for staging AD outlined in the FDA guidance for industry pertaining to developing drugs for treatment of early AD.48

Table 12. National Institute on Aging-Alzheimer’s Association Numerical Clinical Staging for Individuals in the Alzheimer Continuuma

Stage Stage 1 Stage 2 Stage 3 Stage 4 Stage 5 Stage 6
Severity Pre-clinical Pre-clinical MCI due to AD Mild Dementia Moderate Dementia Severe Dementia
Clinical Features
  • Performance within expected range on objective cognitive tests.
  • No evidence of recent cognitive decline or new neurobehavioral symptoms.
  • Normal performance within expected range on objective cognitive tests.
  • Transitional cognitive decline (change from individual baseline within past 1 to 3 years, and persistent for at least 6 months).
  • Mild neurobehavioral changes may coexist or may be the primary complaint rather than cognitive.
  • No functional impact on daily life activities.
  • Performance in the impaired/abnormal range on objective cognitive tests.
  • Evidence of decline from baseline.
  • Performs daily life activities independently, but cognitive difficulty may result in detectable but mild functional impact on the more complex activities of daily life.
  • Substantial progressive cognitive impairment affecting several domains, and/or neurobehavioral disturbance.
  • Clearly evident functional impact on daily life, affecting mainly instrumental activities.
  • No longer fully independent/requires occasional assistance with daily life activities.
  • Progressive cognitive impairment or neurobehavioral changes.
  • Extensive functional impact on daily life with impairment in basic activities.
  • No longer independent and requires frequent assistance with daily life activities.
  • Progressive cognitive impairment or neurobehavioral changes.
  • Clinical interview may not be possible.
  • Complete dependency due to severe functional impact on daily life with impairment in basic activities, including basic self-care.

Adapted from Table 6, Jack et al. (2018)49
a Applicable only to individuals in the Alzheimer continuum that fall into 1 of the 4 biomarker groups: 1) A+T+N+ 2) A+T-N- 3) A+T+N- 4) A+T-N+ where A: Aggregated amyloid beta or associated pathologic state (CSF amyloid beta42, or amyloid beta42/amyloid beta40 ratio or Amyloid PET), T: Aggregated tau (neurofibrillary tangles) or associated pathologic state (CSF phosphorylated tau or Tau PET) and N: Neurodegeneration or neuronal injury (anatomic MRI, FDG PET or CSF total tau)
For stages 1 to 6: Cognitive test performance may be compared to normative data of the investigators choice, with or without adjustment (choice of the investigators) for age, sex, education, etc. 
For stages 2 to 6: Although cognition is the core feature, neurobehavioral changes — for example, changes in mood, anxiety, or motivation — may coexist. 
For stages 3 to 6: Cognitive impairment may be characterized by presentations that are not primarily amnestic.
AD: Alzheimer disease; CSF: cerebrospinal fluid; FDG: fluorodeoxyglucose; MCI: mild cognitive impairment; MRI: magnetic resonance imaging; PET: positron emission tomography.

Interventions
The intervention of interest is amyloid beta imaging using a commercially available PET tracer (florbetapir F18, florbetaben F18, or flutemetamol F18).

Comparators
The comparator of interest is standard clinical management without amyloid beta imaging. A definitive diagnosis of AD requires histopathologic examination of brain tissue obtained by biopsy or autopsy. In practice, clinical criteria based on clinical examination, neurologic and neuropsychological examinations, and interviews with informants (e.g., family members or caregivers) are used to diagnose AD by excluding other diseases that can cause similar symptoms and distinguish AD from other forms of dementia.

Outcomes
The general outcomes of interest are disease-specific survival, overall survival, test validity, symptoms, change in disease status, functional outcomes, health status measures, and quality of life. Follow-up at 2 to 5 years is of interest to monitor outcomes.

Amyloid beta PET is intended to identify patients with the required plaques that are targeted by the therapy. Therefore, response to therapy is the outcome of interest. Important outcomes to measure response include cognitive, functional, and quality of life outcomes.

As per the FDA 2018 draft guidance for developing drugs for treatment of early AD, treatment for mild to moderate AD dementia (corresponding to stages 4 and 5) would be considered substantially effective if there is improvement on a core symptom (e.g., a measure of cognition) and a global clinical measure (e.g., a clinician’s judgement of change) or a functional measure (e.g., activities of daily living).48 For studies including prodromal patients with MCI (corresponding to Stage 3 in the FDA 2018 draft guidance), the FDA requires only a statistically significant change on a prespecified composite measure that includes cognition and daily function combined, as a demonstration of substantial effectiveness. In the 2013 draft guidance, the agency specifically recommended the Clinical Dementia Rating Sum of Boxes (CDR-SB) as a composite measure that had shown validity and reliability for this purpose. No quantified minimum differences were specified but the rationale was that such a composite measure serves as an indicator of change in both the core or cognitive outcome.50 Meeting minimal clinically important difference (MCID) thresholds, however, are not requisites for the FDA to conclude a trial shows substantial effectiveness or to authorize marketing approval.51

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Clinical Validity

The clinical validity of amyloid beta PET in patients with suspected AD is addressed above in this review. Most studies evaluating the diagnostic accuracy of amyloid beta PET in patients with dementia have been conducted in patients at the end of life. Studies in patients with possible AD are needed.

Clinical Utility
Evidence about the clinical utility of amyloid beta PET imaging to select patients for treatment with amyloid beta targeting-therapy is available from 4 studies conducted as part of the clinical development program for aducanumab (Table 13). PRIME was a multicenter, randomized, double-blind, placebo-controlled, dose-ranging, staggered study conducted in the United States with the primary objectives of safety and tolerability. The phase 3 studies were multicenter, global, randomized, double-blind, placebo-controlled studies of identical design with the primary objective of efficacy and safety. In all 3 studies, the diagnosis of AD was confirmed by presence of amyloid pathology measured by 18-florbetapir PET imaging. The pivotal trials ensured enrollment of patients at an earlier stage of their disease (MCI due to AD or mild AD dementia based on an entry criteria).52

This section briefly summarizes these studies. For a more detailed discussion of the evidence see Policy 50138 — Aducanumab for Alzheimer Disease.

Table 13. Summary of the Clinical Development Program for Aducanumab

Trial NCT Phase Description N Design Status
PRIME (Study 3) NCT01677572 1 Evaluate safety and tolerability of multiple doses of aducanumab in prodromal or mild AD 196 DB RCT Completed and published
ENGAGE (Study 301) NCT02477800 3 Evaluate safety and tolerability of aducanumab in early AD 1647 DB RCT Completed and unpublished
EMERGE (Study 302) NCT02484547 3 Evaluate safety and tolerability of aducanumab in early AD 1638 DB RCT Completed and unpublished
EMBARK NCT04241068 3 Evaluate long-term safety and tolerability of aducanumab in participants enrolled in previous trials of aducanumab (EMERGE, ENGAGE, the LTE of the PRIME study, and EVOLVE) 2400 Open-label Ongoing

AD: Alzheimer disease; DB: double-blind; LTE: long-term extension; RCT: randomized controlled trial.

The phase 3 studies randomized patients to aducanumab low dose (3 or 6 mg/kg for ApoE ε4 carriers and noncarriers, respectively), aducanumab high dose (10 mg/kg), or placebo every 4 weeks for 18 months, followed by an optional, dose-blind, long-term extension period. Due to early termination and consequent administrative censoring, data were missing for up to 45% of patients randomized in the 2 trials. Approximately, 60% of patients had the opportunity to complete week 78 of the trial before the trials were terminated for futility.52

Study 302 (N = 1638 randomized patients) met the primary endpoint in patients treated with high-dose aducanumab (10 mg/kg) with an absolute difference of -0.39 in favor of aducanumab on the 18-point CDR-SB scale (a relative 22% less decline in high dose aducanumab group compared to placebo, p = .0120). The reported MCID is generally considered to be 1 to 2 points on a scale from 0 to 18.53, Results of a responder analysis describing the proportion of individuals who achieved a predefined level of improvement were not reported. Results in the low-dose aducanumab (3 or 6 mg/kg for ApoE ε4 carriers and noncarriers, respectively) group were not statistically significant compared with placebo (absolute difference, ‐0.26 ; relative difference, ‐15% ; p = .0901) and therefore no statistically valid conclusions can be made for any of the secondary endpoints for either treatment arm.

Study 301 (N = 1647 randomized patients) did not meet its primary end point of a reduction relative to placebo in the CDR-SB score. For the high-dose arm, an absolute difference of 0.03 and a relative difference of 2% favored placebo (p = .8330). For the low-dose arm, an absolute difference of -0.18 and a relative difference of 12% favored aducanumab (p = .8330). Because of the pre-specified plans to control for type I error for multiple comparisons, no statistically valid conclusions can therefore be made for any of the secondary endpoints.52

Change in brain amyloid signal was measured by florbetapir fluorine 18 PET and quantified by a composite SUVR in a subset of sites and patients (n = 488) at week 78. In study 302, adjusted mean change from baseline to week 78 relative to placebo showed a dose-dependent reduction in amyloid beta by -0.179 and -0.278 in the low- and high-dose arms, respectively. In study 301, adjusted mean change from baseline to week 78 relative to placebo showed a dose-dependent reduction in amyloid beta by -0.167 and -0.232 in the low- and high-dose arms, respectively. While aducanumab showed statistically significant dose dependent changes from baseline in amyloid beta plaques, there are no satisfactory data clearly establishing that individual changes in amyloid correlate with or predict long term cognitive and functional changes as measured by CDR-SB. The FDA statistical review54 reported no correlation in study 302 between reduction in amyloid plaque and long term clinical change among the high-dose cohort or full 10 mg/kg dosed subgroup. In the absence of clinical data convincingly demonstrating a clinical effect, it cannot be concluded that observed reduction in amyloid will translate into a clinical benefit to patients.

Data with limited follow-up are available to analyze safety because the phase 3 trials were stopped prematurely due to futility. Pooled safety data from the 2 phase 3 clinical trials showed that about 35% (compared to 3% in the placebo arm) of patients on aducanumab experienced amyloid-related imaging abnormalities (ARIA), whose clinical effects can range from asymptomatic to severe. Although the majority of patients were asymptomatic or had symptoms such as headache, confusion, or dizziness that resolved with temporary stoppage of the drug, 6.2% of participants receiving the high dose of aducanumab discontinued the drug due to ARIA.55

An increase in falling adverse events was observed in the high-dose group as compared to placebo across the 2 phase 3 studies (15% vs. 12%, respectively). FDA statistical review54 reported a hazard ratio of 1.33 (p = .016) suggesting a 33% relative increase in hazard of falling for 10 mg/kg compared to placebo.

Section Summary: Amyloid Beta Imaging With Positron Emission Tomography to Select Patients for Targeting Therapy
For individuals with a clinical diagnosis of MCI or mild dementia due to AD who are being considered for an FDA-approved amyloid beta plaque-targeting therapy, the evidence includes 2 RCTs and 1 dose-finding and proof of concept phase I trial. ENGAGE (study 301) and EMERGE (study 302) were identical randomized, double-blind, placebo-controlled studies that enrolled patients with early AD. Both trials were terminated early following a prespecified interim analysis for futility. In study 301, there was no treatment benefit observed in either the high- or low-dose arms at week 78. In study 302, a statistically significant difference in change from baseline in CDR-SB was observed in the high-dose arm (difference vs. placebo, -0.39 [95% CI, -0.69 to -0.09]) but not the low-dose arm at week 78. The observed change of 0.39 was well below the range of 1 to 2 points reported as the MCID in published literature. Approval by the FDA was based on the reduction in amyloid plaques, which was observed in both trials and at all doses. However, there are no satisfactory data clearly establishing that individual changes in amyloid correlate with or predict long term cognitive and functional changes. In the absence of clinical data convincingly demonstrating cognitive and functional effects, it cannot be concluded that the observed reduction in amyloid will translate into a clinical benefit to patients. Pooled safety data showed that about 35% of patients on aducanumab experienced ARIA; the risk of falling was also increased with aducanumab. A confirmatory, prospective, and adequately powered trial is necessary to assess the net health benefit of aducanumab in patients with early AD.

Amyloid Beta Imaging With Positron Emission Tomography to Evaluate Patients Receiving Targeting Therapy for Continuation of Treatment
Clinical Context and Test Purpose

The purpose of amyloid beta imaging with PET is to guide decisions about continuation or discontinuation of amyloid beta plaque-targeting therapy.

The question addressed in this evidence review is: Does amyloid beta imaging with PET improve the net health outcome in patients who are receiving disease targeting therapy for AD?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with MCI or early AD who are being treated with amyloid beta plaque-targeting therapy.

Interventions
The intervention of interest is amyloid beta imaging using a commercially available PET tracer (florbetapir F18, florbetaben F18, or flutemetamol F18).

Comparators
The comparator of interest is standard clinical management without amyloid beta imaging. The decision to continue or discontinue treatment would be based on clinical factors.

Outcomes
The general outcomes of interest are disease-specific survival, overall survival, test validity, symptoms, change in disease status, functional outcomes, health status measures, and quality of life. Follow-up at 2 to 5 years is of interest to monitor outcomes.

Amyloid beta PET is intended to identify patients with the required plaques that are targeted by the therapy. Therefore, response to therapy is the outcome of interest. Important outcomes to measure response include cognitive, functional, and quality of life outcomes.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
No studies addressing the use of amyloid beta PET imaging to monitor response to amyloid beta targeting-therapy were identified. The aducanumab product label recommends monitoring for ARIAs using magnetic resonance imaging (MRI), but does not address monitoring amyloid beta using PET.

Section Summary: Amyloid Beta Imaging With Positron Emission Tomography to Evaluate Patients Receiving Targeting Therapy for Continuation of Treatment
No evidence was identified.

Fluorine 18 Fluorodeoxyglucose Positron Emission Tomography to Confirm a Diagnosis of Alzheimer Disease
Clinical Context and Test Purpose

The purpose of fluorine 18 fluorodeoxyglucose PET (FDG-PET) in patients with suspected AD is to confirm a diagnosis of AD.

The question addressed in this evidence review is: Does the use of FDG-PET improve the net health outcome in individuals with suspected AD?

The following PICO was used to select literature to inform this review.

Populations
The population of interest is patients with suspected AD.

A definitive diagnosis of AD requires histopathologic examination of brain tissue obtained by biopsy or autopsy. In practice, clinical criteria based on clinical examination, neurologic and neuropsychological examinations, and interviews with informants (e.g., family members or caregivers) are used to diagnose AD by excluding other diseases that can cause similar symptoms and distinguish AD from other forms of dementia.

Interventions
The intervention of interest is FDG-PET. FDG-PET quantifies brain function by measuring glucose levels. Through identifying distinct regions of hypometabolism, FDG-PET is proposed as a method to distinguish AD from other dementias, especially in patients with atypical presentations such as younger age.

For patients with suspected AD, FDG-PET would be performed following inconclusive clinical examinations and standard radiographs.

Comparators
Clinical diagnosis without FDG-PET is currently being used for suspected AD.

Outcomes
For patients with suspected AD, the main outcomes of interest are test validity, symptoms, change in disease status, functional outcomes, health status measures, and quality of life.

Study Selection Criteria
For the evaluation of the clinical validity of the tests, studies that meet the following eligibility criteria were considered:

  • Reported on the accuracy of the marketed version of the technology (including any algorithms used to calculate scores)
  • Included a suitable reference standard
  • Patient/sample clinical characteristics were described
  • Patient/sample selection criteria were described
  • Included a validation cohort separate from development cohort

Clinically Valid
A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Review of Evidence
Systematic Reviews

Summaries of the characteristics and results of several meta-analyses of the early diagnosis of AD in people with cognitive impairment or for differentiating between potential causes of dementia are shown in Tables 14 and 15, and are briefly described below.

Table 14. Characteristics of Systematic Reviews on FDG-PET for Diagnosing AD and Dementia

Study Dates Studies N (Range) Design Outcomes
Zhu et al. (2022)56 Up to 2020 16 NR OBS Diagnostic accuracy for predicting conversion from MCI to AD
Smailagic et al. (2015)57 1999 –2013 16 697 (19 – 94) OBS Diagnostic accuracy for predicting conversion from MCI to AD
Davison et al. (2014)58 Up to 2013 9 NR OBS Diagnostic accuracy for diagnosis of AD, differential diagnosis in dementia, predicting conversion from MCI to AD
Bloudek et al. (2011)59 1990 –2010 119 NR OBS Diagnostic accuracy for diagnosis of AD, differential diagnosis in dementia
Yuan et al. (2009)60 2001 – 2005 6 280 (17 –128) OBS Diagnostic accuracy for predicting conversion from MCI to AD
Matchar et al. (2001)61 1995 –2001 18 1018 (10 –138) OBS Diagnostic accuracy for distinguishing AD from healthy controls and for differential diagnosis in dementia

AD: Alzheimer disease; FDG-PET: fluorine 18 fluorodeoxyglucose positron emission tomography; MCI: mild cognitive impairment; NR: not reported; OBS: observational.

Zhu et al. (2022) conducted a meta-analysis of cerebral perfusion imaging methods (FDG-PET, SPECT, and MRI) in the assessment of MCI conversion to AD. A total of 16 studies were included (5 with FDG-PET).56 The authors found significantly higher sensitivity, specificity, and positive likelihood ratio with FDG-PET than SPECT or MRI. The studies for FDG-PET were determined to have low risk of bias.

Smailagic et al. (2015) conducted a Cochrane review to assess the diagnostic accuracy of FDG-PET for detecting people who clinically convert to AD or other forms of dementia at follow-up.57 Included studies evaluated the diagnostic accuracy of FDG-PET to determine the conversion from MCI to AD or to other forms of dementia. Sixteen studies (N = 697 participants) were included in the qualitative review and 14 studies (n = 421 participants) were included in the analysis. Because there are no accepted thresholds to define positive findings based on PET scans and studies used mixed thresholds for diagnosis, reviewers used a hierarchical summary receiver operating characteristic curve to derive pooled estimates of performance characteristics at fixed values. Results are shown in Table 15. Five studies evaluated the accuracy of FDG-PET for all types of dementia. The sensitivities ranged between 46% and 95% while the specificities ranged between 29% and 100%; however, a meta-analysis could not be conducted because of the small study sample sizes. Reviewers indicated that most studies were poorly reported and had an unclear risk of bias, mainly for the reference standard and participant selection domains.

In a systematic review (quality assessment of included studies was not reported), Davison et al. (2014) reported on studies on the diagnostic performance of FDG-PET and SPECT identified through PubMed.58 Three studies (197 patients) used histopathology as the reference standard. In patients with or without a clinical diagnosis of AD, sensitivity was 84% and specificity was 74%. In patients with memory loss or dementia, sensitivity was 94% and specificity was approximately 70%. In patients undergoing evaluation for dementia, sensitivity was 94% and specificity was 73%. Precision estimates were not given. In 3 different studies (271 participants), the sensitivities and specificities of FDG-PET for distinguishing AD from Lewy body dementia ranged from 83% to 99% and from 71% to 93%, respectively. In 2 studies (183 participants), for predicting conversion from MCI to AD, sensitivity and specificity of PET ranged from 57% to 82% and from 67% to 78%, respectively.

Bloudek et al. (2011) assessed diagnostic strategies for AD in a meta-analysis.59 Reviewers included 119 studies of diagnostic performance characteristics published from 1990 to 2010. Studies were identified through a search of PubMed and included imaging, biomarkers, and clinical diagnostic strategies. Twenty studies included performance characteristics of FDG-PET for diagnosing AD compared with normal, nondemented controls. Thirteen studies described characteristics of FDG-PET for diagnosing AD compared with demented controls. FDG-PET demonstrated the highest area under the receiver operating characteristic curve, sensitivity, and specificity among all of the diagnostic methods for distinguishing AD from normal controls, but one of the lowest receiver operating characteristic curves comparing AD with non-AD demented controls (excluding MCI), due primarily to the low specificity in this group. Results are shown in Table 15.

In a meta-analysis, Yuan et al. (2009) compared the prognostic capacity of FDG-PET, SPECT, and structural MRI to predict patients' conversion from MCI to AD.60 Using 24 articles (N = 1112 patients) published between 1990 to 2008 (6 studies with 280 patients on FDG-PET, published between 2001 – 2005), reviewers found no statistically significant difference among the 3 modalities in pooled sensitivity, pooled specificity, or negative likelihood ratio. Results are shown in Table 15. There was strong evidence of between-study heterogeneity and marked asymmetry in the funnel plot (with studies missing from the bottom left quadrant), indicating possible publication bias of studies with null results. Efforts to identify sources of heterogeneity (e.g., publication year, age, male-female ratio, follow-up interval, years of education, mean Mini-Mental State Examination score at baseline) yielded no significant results.

Using decision-analysis modeling, Matchar et al. (2001) performed a technology assessment for the Agency for Healthcare Research and Quality to examine whether the use of FDG-PET would improve health outcomes for diagnosis of AD in 3 clinical populations: patients with dementia, patients with MCI, and subjects with no symptoms, but with a first-degree relative with AD.61 For the review, a search was performed using PubMed, CINAHL, and the HealthSTAR databases. Eighteen articles (N = 1018 participants) were included. The reference standard used in the studies was either histopathology or clinical diagnosis. Studies reported on various cutoffs for PET positivity, and, therefore, an unweighted summary receiver operating characteristic method was used to calculate the pooled area under the curve. Results are summarized in Table 15. Reviewers concluded that outcomes for all 3 groups were better if all patients were treated with agents such as cholinesterase inhibitors rather than limiting treatment to patients based on FDG-PET results. The rationale was that the complications of treatment were relatively mild, and that treatment was considered to have some degree of efficacy in delaying the progression of AD.

Table 15. Results of Systematic Review on Use Assessing FDG-PET for AD and Dementia

Study Studies N Outcomes Estimate (95% CI)
Zhu et al. (2022)56 5a NR Diagnostic accuracy
  • Sensitivity: 87.2% (81.3% to 92.1%)
  • Specificity: 89.35% (77.6% to 91.8%)
  • PLR: 5.973 (3.15 to 6.72)
  • NLR: 0.132 (0.05 to 0.49)
Smailagic et al. (2015)57 14 421 Diagnostic accuracy
  • Sensitivity range: 25% to 100%
  • Specificity range: 15% to 100%
  • PLR: 4.03 (2.97 to 5.47)
  • NLR: 0.34 (0.15 to 0.75)
Davison et al. (2014)58 3 197 Diagnostic accuracy
  • Sensitivity: 84%
  • Specificity: 74%
  2 183 Diagnostic accuracy, predicting conversion from MCI to AD
  • Sensitivity range: 57% to 82%
  • Specificity range: 67% to 78%
  5 292 Diagnostic accuracy, differentiating AD and LBD
  • Sensitivity range: 83% to 92%
  • Specificity range: 67% to 93%
Bloudek et al. (2011)59 20 NR Diagnostic accuracy
  • Sensitivity: 90% (84% to 94%)
  • Specificity: 89% (81% to 94%)
  13 NR Diagnostic accuracy, AD vs. other dementia
  • Sensitivity: 92% (84% to 96%)
  • Specificity: 78% (69% to 85%)
Yuan et al. (2009)60 6 280 Diagnostic accuracy
  • Sensitivity: 89% (92% to 94%)
  • Specificity: 85% (78% to 90%)
  • PLR: 4.6 (3.2 to 6.7)
  • NLR: 0.15 (0.05 to 0.48)
Matchar et al. (2001)61 15 729 Diagnostic accuracy
  • Sensitivity: 88% (79% to 94%)
  • Specificity: 87% (77% to 93%)
  3 289 Diagnostic accuracy, distinguishing AD from non-AD dementia
  • Sensitivity range: 86% to 95%
  • Specificity range: 61% to 74%

AD: Alzheimer disease; CI: confidence interval; FDG-PET: fluorine 18 fluorodeoxyglucose positron emission tomography; LBD: Lewy body dementia; MCI: mild cognitive impairment; NLR: negative likelihood ratio; NR; not reported; PLR: positive likelihood ratio.
a Includes only the 5 studies with FDG-PET.

Clinically Useful
A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Review of Evidence
Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

Motara et al. (2017) assessed the accuracy of dual-trained radiologists and nuclear medicine physicians to diagnose the type of cognitive impairment based on FDG-PET/computed tomography (CT) images. Records of patients who had undergone FDG-PET/CT because of cognitive impairment (AD, frontotemporal dementia, mixed dementia, and dementia with Lewy bodies) following a negative CT or MRI scans were reviewed (N = 136).62 Questionnaires were sent to the referring physicians to gather information on the final clinical diagnosis, usefulness of the PET/CT report, and whether the report impacted clinical management. The response rate was 72% (98/136) and mean patient follow-up was 471 days. For the diagnosis of AD, using the final clinical diagnosis as the reference standard, the sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were 87%, 97%, 93%, and 91%, respectively. Questionnaires received from the 98 physicians indicated that PET/CT: was useful (78%); had an impact on clinical management (81%); added confidence to the pretest clinical diagnosis (43%); reduced the need for further investigations (42%); changed the pretest clinical diagnosis (35%); and led to a change in therapy (32%).

Section Summary: Fluorine 18 Fluorodeoxyglucose Positron Emission Tomography to Confirm a Diagnosis of Alzheimer Disease
Several systematic reviews offer evidence on FDG-PET for diagnosing AD in people with cognitive impairment and for differentiating between AD and other dementias. Studies included in these reviews were generally poor quality. There is no standard cutoff for positive amyloid findings on PET scanning for diagnosing AD, and many studies did not include postmortem confirmation of AD as the reference standard. These limitations lead to uncertainty about estimates of performance characteristics. Although it appears that FDG-PET has high sensitivity and specificity, the evidence does not compare the performance characteristics of clinical diagnosis with PET to clinical diagnosis without PET, so the incremental value of adding PET to the standard clinical diagnosis is unclear. No studies reported on clinical outcomes of patients diagnosed with versus without FDG-PET. A single study was identified that surveyed physicians on the clinical utility of FDG-PET/CT in managing patients with cognitive impairment. In general, the physicians found the FDG-PET/CT helpful but no clinical outcomes of patients were reported.

Summary of Evidence
For individuals who have MCI who receive amyloid beta imaging with PET to predict conversion to AD, the evidence includes studies on diagnostic accuracy and a RCT that evaluated changes in diagnosis and management. Relevant outcomes are test validity, symptoms (cognitive and/or behavioral), change in disease status, functional outcomes, health status measures, and quality of life. Studies have been conducted to evaluate the diagnostic accuracy of amyloid beta PET in patients with MCI, using conversion to probable AD as a reference standard. Systematic reviews of these studies have concluded that limited data, varying sensitivity and specificity, and risk of bias limited confidence in conclusions. In a more recent prospective study of 224 individuals with MCI, the hazard ratio for conversion to probable AD at 3 years in patients with a baseline positive amyloid beta PET scan was 2.51 (95% CI, 1.57 to 3.99; p < .001), with a NPV of 77%. Direct evidence of improved health outcomes with this technology is lacking. A RCT tested immediate versus delayed reporting of amyloid beta test results for patients with MCI and AD. No differences between the groups were found for health outcomes, although the study was not powered for these outcome measures. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals who have dementia who receive amyloid beta imaging with PET as an adjunct to clinical diagnosis, the evidence includes studies on diagnostic accuracy and a RCT that evaluated changes in diagnosis and management. Relevant outcomes are test validity, symptoms (cognitive and/or behavioral), change in disease status, functional outcomes, health status measures, and quality of life. One possible use of amyloid beta testing is as an adjunct to clinical diagnosis to rule out AD ; this could lead to further diagnostic testing to determine the etiology of dementia, and potentially facilitate avoidance of inappropriate presumptive medication use and/or appropriate use of medications for other types of dementia. The pivotal trials showed a sensitivity of 86% to 93% and a specificity of 86% to 100% compared with the criterion standard of amyloid beta plaque density on postmortem histology. However, the patients in these studies were at the end of life and not representative of the population of patients with suspected AD who present earlier in the course of the disease. Due to the lack of a criterion standard in living patients and limited follow-up, the sensitivity and specificity of amyloid beta PET in patients with suspected AD are unknown. Direct evidence of improved health outcomes with this technology is lacking. A RCT that tested immediate versus delayed reporting of amyloid beta test results for patients with MCI and AD found changes in diagnosis and management, but the effect of these changes on health outcomes such as quality of life, cognitive and behavioral symptoms, and functional outcomes is uncertain. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with a clinical diagnosis of MCI or mild dementia due to AD who are being considered for an FDA-approved amyloid beta plaque-targeting therapy, the evidence includes 2 RCTs and 1 dose-finding and proof of concept phase I trial of aducanumab. Relevant outcomes are test validity, symptoms (cognitive and/or behavioral), change in disease status, functional outcomes, health status measures, quality of life, disease-specific survival, and overall survival. ENGAGE (study 301) and EMERGE (study 302) were identical randomized, double-blind, placebo-controlled studies that enrolled patients with early AD. Both trials were terminated early following a prespecified interim analysis for futility. In study 301, there was no treatment benefit observed in either the high- or low-dose arms at week 78. In study 302, a statistically significant difference in change from baseline in CDR-SB was observed in the high-dose arm (difference vs. placebo, -0.39 [95% CI, -0.69 to -0.09]) but not the low-dose arm at week 78. The observed change of 0.39 was well below the range of 1 to 2 points considered the MCID. Approval by the FDA was based on reduction in amyloid plaques, which was observed in both trials and at all doses. However, there are no satisfactory data clearly establishing that individual changes in amyloid beta plaque correlate with or predict long-term cognitive and functional changes. In the absence of clinical data demonstrating cognitive and functional effects, it cannot be concluded that the observed reduction in amyloid will translate into a clinical benefit to patients. Pooled safety data showed that about 35% of patients on aducanumab experienced ARIA; an increased risk of falling was also observed. A confirmatory, prospective, and adequately powered trial is necessary to assess the net health benefit of aducanumab in patients with early AD. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with early AD (MCI or mild dementia due to AD) who are being treated with amyloid beta plaque-targeting therapy and are being evaluated for continuation of therapy, no evidence was identified on the role of subsequent or repeat amyloid beta PET imaging or its correlation with clinical assessment of disease status. Relevant outcomes are test validity, symptoms (cognitive and/or behavioral), change in disease status, functional outcomes, health status measures, quality of life, disease-specific survival, and overall survival. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals who have suspected AD who receive FDG-PET to diagnose AD, the evidence includes systematic reviews of nonrandomized studies. Relevant outcomes are test validity, symptoms (cognitive and/or behavioral), change in disease status, functional outcomes, health status measures, and quality of life. The studies included in the reviews were generally of poor quality. There is no standard cutoff for FDG-PET positivity for diagnosing AD, and many studies have not included postmortem confirmation of AD as the reference standard, leading to uncertainty about estimates of performance characteristics. FDG-PET may have high sensitivity and specificity for diagnosing AD, but there is little evidence comparing the performance characteristics of clinical diagnosis using FDG-PET with the clinical diagnosis not using FDG-PET. Therefore, the incremental value of adding FDG-PET to the standard clinical diagnosis is unclear. No studies have reported on clinical outcomes of patients diagnosed with and without FDG-PET. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

The purpose of the following information is to provide reference material. Inclusion does not imply endorsement or alignment with the evidence review conclusions.

Practice Guidelines and Position Statements
Guidelines or position statements will be considered for inclusion in "Supplemental Information" if they were issued by, or jointly by, a U.S. professional society, an international society with U.S. representation, or National Institute for Health and Care Excellence (NICE). Priority will be given to guidelines that are informed by a systematic review, include strength of evidence ratings, and include a description of management of conflict of interest.

American College of Radiology
The American College of Radiology appropriateness criteria for dementia, revised in 2019, state that amyloid positron emission tomography (PET) and fluorine 18 fluorodeoxyglucose positron emission tomography (FDG-PET) may be appropriate for initial imaging of patients with cognitive decline and suspected Alzheimer disease (AD).63,

Society of Nuclear Medicine and Molecular Imaging and Alzheimer's Association
The Appropriate Use Criteria (2013) for amyloid PET were developed jointly by the Society of Nuclear Medicine and Molecular Imaging and the Alzheimer's Association.64 They recommended that amyloid imaging is appropriate for individuals with all of the following characteristics:

"(i) a cognitive complaint with objectively confirmed impairment; (ii) AD [Alzheimer disease] as a possible diagnosis, but when the diagnosis is uncertain after a comprehensive evaluation by a dementia expert; and (iii) when knowledge of the presence or absence of AD pathology is expected to increase diagnostic certainty and alter management."

Appropriate candidates include:

  1. Patients with unexplained persistent or progressive MCI [mild cognitive impairment].
  2. Patients satisfying core clinical criteria for possible AD, but are unusual in the clinical presentation.
  3. Patients with progressive dementia and atypically early age of onset (e.g., 65 years of age or less).

Amyloid imaging is inappropriate in the following situations:

  1. "Patients with core clinical criteria for probable AD with typical age of onset
  2. To determine dementia severity
  3. Based solely on a positive family history of dementia or presence of apolipoprotein E (APOE) ε4
  4. Patients with a cognitive complaint that is unconfirmed on clinical examination
  5. In lieu of genotyping for suspected autosomal mutation carriers
  6. In asymptomatic individuals
  7. Nonmedical use (e.g., legal, insurance coverage, or employment screening)."

U.S. Preventive Services Task Force Recommendations
In 2020, the USPSTF concluded that the current evidence is insufficient to assess the balance of benefits and harms of screening for cognitive impairment in older adults (I statement).65

Ongoing and Unpublished Clinical Trials
Some currently ongoing and unpublished trials that might influence this review are listed in Table 16.

Table 16. Summary of Key Trials

NCT No. Trial Name Planned Enrollment Completion Date
Ongoing      
NCT05108922a A Phase 3, Open-Label, Parallel-Group, 2-Arm Study to Investigate Amyloid Plaque Clearance With Donanemab Compared With Aducanumab-avwa in Participants With Early Symptomatic Alzheimer's Disease (TRAILBLAZER-ALZ 4) 200 July 2024
NCT05457998 BioFINDER-Brown: Examination of Alzheimer's Disease Biomarkers 200 Sept. 2027
NCT05508789a Global Study to Investigate Safety and Efficacy of Donanemab in Early Symptomatic Alzheimer's Disease (TRAILBLAZER-ALZ 5) 800 July 2025
NCT02008357a Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4 Study) 1150 Dec. 2022
NCT03444870a Phase III, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Efficacy, and Safety Study of Gantenerumab in Patients With Early (Prodromal to Mild) Alzheimer's Disease 1016 Oct. 2026
NCT04437511a Assessment of Safety, Tolerability, and Efficacy of Donanemab in Early Symptomatic Alzheimer's Disease (TRAILBLAZER-ALZ 2) 1800 Aug. 2025
NCT04468659a AHEAD 3-45 Study: A Placebo-Controlled, Double-Blind, Parallel-Treatment Arm, 216 Week Study to Evaluate Efficacy and Safety of Treatment With BAN2401 in Subjects With Preclinical Alzheimer's Disease and Elevated Amyloid (A45 Trial) and in Subjects With Early Preclinical Alzheimer's Disease and Intermediate Amyloid (A3 Trial) 1400 Oct. 2027
NCT03860857 MRI and PET Biomarkers for Cognitive Decline in Older Adults 200 Dec. 2024
NCT04669028a A Phase 3, Double Blind, Randomized, Placebo Controlled, Parallel Group, Multicenter Study of NE3107 in Subjects Who Have Mild to Moderate Alzheimer's Disease 316 Jan. 2023
NCT03887455a A Placebo-Controlled, Double-Blind, Parallel-Group, 18-Month Study With an Open-Label Extension Phase to Confirm Safety and Efficacy of BAN2401 in Subjects With Early Alzheimer's Disease 1906 Sept. 2025
NCT04241068a A Study to Evaluate Safety and Tolerability of Aducanumab in Participants With Alzheimer's Disease Who Had Previously Participated in the Aducanumab Studies 221AD103, 221AD301, 221AD302 and 221AD205 1696 Feb. 2025
NCT04426539 New IDEAS: Imaging Dementia-Evidence for Amyloid Scanning Study - A Study to Improve Precision in Amyloid PET Coverage and Patient Care 7000 Dec. 2024
Unpublished      
NCT02781220 Implications for Management of PET Amyloid Classification Technology in the Imaging Dementia (IDEAS) Trial 69 July 2021 (last update Jan. 2020)

NCT: national clinical trial.
a Denotes industry-sponsored or cosponsored trial.

References: 

  1. 2022 Alzheimer's disease facts and figures. Alzheimers Dement. Apr 2022; 18(4): 700-789. PMID 35289055
  2. Alzheimer's Association. 2022 Alzheimer's disease facts and figures. Published 2022. Available at https://www.alz.org/media/Documents/alzheimers-facts-and-figures.pdf. Accessed September 2, 2022.
  3. Roberts RO, Aakre JA, Kremers WK, et al. Prevalence and Outcomes of Amyloid Positivity Among Persons Without Dementia in a Longitudinal, Population-Based Setting. JAMA Neurol. Aug 01 2018; 75(8): 970-979. PMID 29710225
  4. National Institute on Aging. Data shows racial disparities in Alzheimers disease diagnosis between Black and white research study participants. 2021. https://www.nia.nih.gov/news/data-shows-racial-disparities-alzheimers-disease-diagnosis-between-black-and-white-research. Accessed August 31, 2022.
  5. Lu ZK, Xiong X, Wang X, et al. Gender Disparities in Anti-dementia Medication Use among Older Adults: Health Equity Considerations and Management of Alzheimer's Disease and Related Dementias. Front Pharmacol. 2021; 12: 706762. PMID 34512340
  6. Vallabhajosula S. Positron emission tomography radiopharmaceuticals for imaging brain Beta-amyloid. Semin Nucl Med. Jul 2011; 41(4): 283-99. PMID 21624562
  7. Ossenkoppele R, Jansen WJ, Rabinovici GD, et al. Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. JAMA. May 19 2015; 313(19): 1939-49. PMID 25988463
  8. Jansen WJ, Ossenkoppele R, Knol DL, et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA. May 19 2015; 313(19): 1924-38. PMID 25988462
  9. Wolk DA & DeKosky ST. Clinical features and diagnosis of Alzheimer disease. In: UpToDate, DeKosky ST (Ed), UpToDate, Waltham, MA. https://www.uptodate.com/contents/clinical-features-and-diagnosis-of-alzheimer-disease. Accessed August 31, 2022.
  10. Reuben DB, Tan ZS, Romero T, et al. Patient and Caregiver Benefit From a Comprehensive Dementia Care Program: 1-Year Results From the UCLA Alzheimer's and Dementia Care Program. J Am Geriatr Soc. Nov 2019; 67(11): 2267-2273. PMID 31355423
  11. Gronek P, Balko S, Gronek J, et al. Physical Activity and Alzheimer's Disease: A Narrative Review. Aging Dis. Dec 2019; 10(6): 1282-1292. PMID 31788339
  12. Du Z, Li Y, Li J, et al. Physical activity can improve cognition in patients with Alzheimer's disease: a systematic review and meta-analysis of randomized controlled trials. Clin Interv Aging. 2018; 13: 1593-1603. PMID 30233156
  13. Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. Nov 21 2012; 308(19): 2020-9. PMID 23168825
  14. Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt HP, et al. Cholinesterase inhibitors for patients with Alzheimer's disease: systematic review of randomised clinical trials. BMJ. Aug 06 2005; 331(7512): 321-7. PMID 16081444
  15. Eli Lilly and Company. Amyvid (florbetapir F18 injection) for intravenous use prescribing information, December 2019. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=bb5a5043-0f51-11df-8a39-0800200c9a66. Accessed August 31, 2022.
  16. GE Healthcare. Vizamyl (flutemetamol F18) injection prescribing information. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=b3558f16-8f9a-4e55-8d9c-836427ebaa57. Accessed August 31, 2022.
  17. Piramal Imaging. Neuraceq (florbetaben F 18 injection) for intravenous use prescribing information. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=b0915068-cfd4-4d72-b9f8-7e31fe83cd1e. Accessed August 31, 2022.
  18. Ong KT, Villemagne VL, Bahar-Fuchs A, et al. A imaging with 18F-florbetaben in prodromal Alzheimer's disease: a prospective outcome study. J Neurol Neurosurg Psychiatry. Apr 2015; 86(4): 431-6. PMID 24970906
  19. Thurfjell L, Lotjonen J, Lundqvist R, et al. Combination of biomarkers: PET [18F]flutemetamol imaging and structural MRI in dementia and mild cognitive impairment. Neurodegener Dis. 2012; 10(1-4): 246-9. PMID 22301718
  20. Pichet Binette A, Palmqvist S, Bali D, et al. Combining plasma phospho-tau and accessible measures to evaluate progression to Alzheimer's dementia in mild cognitive impairment patients. Alzheimers Res Ther. Mar 29 2022; 14(1): 46. PMID 35351181
  21. Schreiber S, Landau SM, Fero A, et al. Comparison of Visual and Quantitative Florbetapir F 18 Positron Emission Tomography Analysis in Predicting Mild Cognitive Impairment Outcomes. JAMA Neurol. Oct 2015; 72(10): 1183-90. PMID 26280102
  22. Doraiswamy PM, Sperling RA, Johnson K, et al. Florbetapir F 18 amyloid PET and 36-month cognitive decline: a prospective multicenter study. Mol Psychiatry. Sep 2014; 19(9): 1044-51. PMID 24614494
  23. Kawas CH, Greenia DE, Bullain SS, et al. Amyloid imaging and cognitive decline in nondemented oldest-old: the 90+ Study. Alzheimers Dement. Mar 2013; 9(2): 199-203. PMID 23164550
  24. Martinez G, Vernooij RW, Fuentes Padilla P, et al. 18F PET with florbetaben for the early diagnosis of Alzheimer's disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. Nov 22 2017; 11: CD012883. PMID 29164600
  25. Martinez G, Vernooij RW, Fuentes Padilla P, et al. 18F PET with flutemetamol for the early diagnosis of Alzheimer's disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. Nov 22 2017; 11: CD012884. PMID 29164602
  26. Martinez G, Vernooij RW, Fuentes Padilla P, et al. 18F PET with florbetapir for the early diagnosis of Alzheimer's disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. Nov 22 2017; 11: CD012216. PMID 29164603
  27. Johnson KA, Sperling RA, Gidicsin CM, et al. Florbetapir (F18-AV-45) PET to assess amyloid burden in Alzheimer's disease dementia, mild cognitive impairment, and normal aging. Alzheimers Dement. Oct 2013; 9(5 Suppl): S72-83. PMID 23375563
  28. Ben Bouallegue F, Mariano-Goulart D, Payoux P. Joint Assessment of Quantitative 18F-Florbetapir and 18F-FDG Regional Uptake Using Baseline Data from the ADNI. J Alzheimers Dis. 2018; 62(1): 399-408. PMID 29439345
  29. Ottoy J, Niemantsverdriet E, Verhaeghe J, et al. Association of short-term cognitive decline and MCI-to-AD dementia conversion with CSF, MRI, amyloid- and 18 F-FDG-PET imaging. Neuroimage Clin. 2019; 22: 101771. PMID 30927601
  30. Jun S, Kim H, Kim BS, et al. Quantitative Brain Amyloid Measures Predict Time-to-Progression from Amnestic Mild Cognitive Impairment to Alzheimer's Disease. J Alzheimers Dis. 2019; 70(2): 477-486. PMID 31256127
  31. Wolk DA, Sadowsky C, Safirstein B, et al. Use of Flutemetamol F 18-Labeled Positron Emission Tomography and Other Biomarkers to Assess Risk of Clinical Progression in Patients With Amnestic Mild Cognitive Impairment. JAMA Neurol. Sep 01 2018; 75(9): 1114-1123. PMID 29799984
  32. Pontecorvo MJ, Siderowf A, Dubois B, et al. Effectiveness of Florbetapir PET Imaging in Changing Patient Management. Dement Geriatr Cogn Disord. 2017; 44(3-4): 129-143. PMID 28787712
  33. Clark CM, Schneider JA, Bedell BJ, et al. Use of florbetapir-PET for imaging beta-amyloid pathology. JAMA. Jan 19 2011; 305(3): 275-83. PMID 21245183
  34. Clark CM, Pontecorvo MJ, Beach TG, et al. Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid- plaques: a prospective cohort study. Lancet Neurol. Aug 2012; 11(8): 669-78. PMID 22749065
  35. U.S. Food and Drug Administration. Vizamyl (flutemetamol F 18) summary review. 2013; http://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/203137_vizamyl_toc.cfm. Accessed August 31, 2022.
  36. Sabri O, Sabbagh MN, Seibyl J, et al. Florbetaben PET imaging to detect amyloid beta plaques in Alzheimer's disease: phase 3 study. Alzheimers Dement. Aug 2015; 11(8): 964-74. PMID 25824567
  37. Curtis C, Gamez JE, Singh U, et al. Phase 3 trial of flutemetamol labeled with radioactive fluorine 18 imaging and neuritic plaque density. JAMA Neurol. Mar 2015; 72(3): 287-94. PMID 25622185
  38. Salloway S, Gamez JE, Singh U, et al. Performance of [ 18 F]flutemetamol amyloid imaging against the neuritic plaque component of CERAD and the current (2012) NIA-AA recommendations for the neuropathologic diagnosis of Alzheimer's disease. Alzheimers Dement (Amst). 2017; 9: 25-34. PMID 28795133
  39. Ossenkoppele R, Rabinovici GD, Smith R, et al. Discriminative Accuracy of [18F]flortaucipir Positron Emission Tomography for Alzheimer Disease vs Other Neurodegenerative Disorders. JAMA. Sep 18 2018; 320(11): 1151-1162. PMID 30326496
  40. Bao YW, Chau ACM, Chiu PK, et al. Heterogeneity of Amyloid Binding in Cognitively Impaired Patients Consecutively Recruited from a Memory Clinic: Evaluating the Utility of Quantitative 18F-Flutemetamol PET-CT in Discrimination of Mild Cognitive Impairment from Alzheimer's Disease and Other Dementias. J Alzheimers Dis. 2021; 79(2): 819-832. PMID 33361593
  41. Grundman M, Pontecorvo MJ, Salloway SP, et al. Potential impact of amyloid imaging on diagnosis and intended management in patients with progressive cognitive decline. Alzheimer Dis Assoc Disord. Jan-Mar 2013; 27(1): 4-15. PMID 23203162
  42. Boccardi M, Altomare D, Ferrari C, et al. Assessment of the Incremental Diagnostic Value of Florbetapir F 18 Imaging in Patients With Cognitive Impairment: The Incremental Diagnostic Value of Amyloid PET With [18F]-Florbetapir (INDIA-FBP) Study. JAMA Neurol. Dec 01 2016; 73(12): 1417-1424. PMID 27802513
  43. Zwan MD, Bouwman FH, Konijnenberg E, et al. Diagnostic impact of [ 18 F]flutemetamol PET in early-onset dementia. Alzheimers Res Ther. Jan 17 2017; 9(1): 2. PMID 28093088
  44. Ceccaldi M, Jonveaux T, Verger A, et al. Added value of 18 F-florbetaben amyloid PET in the diagnostic workup of most complex patients with dementia in France: A naturalistic study. Alzheimers Dement. Mar 2018; 14(3): 293-305. PMID 29107051
  45. Leuzy A, Savitcheva I, Chiotis K, et al. Clinical impact of [ 18 F]flutemetamol PET among memory clinic patients with an unclear diagnosis. Eur J Nucl Med Mol Imaging. Jun 2019; 46(6): 1276-1286. PMID 30915522
  46. Rabinovici GD, Gatsonis C, Apgar C, et al. Association of Amyloid Positron Emission Tomography With Subsequent Change in Clinical Management Among Medicare Beneficiaries With Mild Cognitive Impairment or Dementia. JAMA. Apr 02 2019; 321(13): 1286-1294. PMID 30938796
  47. Vermunt L, Sikkes SAM, van den Hout A, et al. Duration of preclinical, prodromal, and dementia stages of Alzheimer's disease in relation to age, sex, and APOE genotype. Alzheimers Dement. Jul 2019; 15(7): 888-898. PMID 31164314
  48. US Food and Drug Administration. Early Alzheimers disease: developing drugs for treatment guidance for industry. Draft Guidance. Published online Feb 29, 2018. Available at https://www.fda.gov/media/110903/download. Accessed on September 2, 2022.
  49. Jack CR, Bennett DA, Blennow K, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. Apr 2018; 14(4): 535-562. PMID 29653606
  50. US Food and Drug Administration. Draft guidance for industry on Alzheimers disease: developing drugs for the treatment of early stage disease. Published online March 28, 2013. Available at https://isctm.org/public_access/FDAGuidance_AD_Developing_Drugs_Early_Stage_Treatment.pdf. Accessed September 2, 2022.
  51. Liu KY, Schneider LS, Howard R. The need to show minimum clinically important differences in Alzheimer's disease trials. Lancet Psychiatry. Nov 2021; 8(11): 1013-1016. PMID 34087114
  52. Combined FDA and Applicant PCNS Drugs Advisory Committee Briefing Document: Peripheral and Central Nervous System (PCNS) Drugs Advisory Committee Meeting- November 6, 2020. Available at https://www.fda.gov/media/143502/download. Accessed September 2, 2022.
  53. Andrews JS, Desai U, Kirson NY, et al. Disease severity and minimal clinically important differences in clinical outcome assessments for Alzheimer's disease clinical trials. Alzheimers Dement (N Y). 2019; 5: 354-363. PMID 31417957
  54. FDA Pre-Recorded Presentation Slides for the November 6, 2020: Meeting of the Peripheral and Central Nervous System Drugs Advisory Committee. Available at https://www.fda.gov/media/143504/download. Accessed September 2, 2022.
  55. Prescribing Label: ADUHELM (aducanumab-avwa) injection, for intravenous use. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=41706573-546f-6774-6872-5374726f6e67. Accessed September 2, 2022.
  56. Zhu L, Zhao W, Chen J, et al. Systematic review and meta-analysis of diagnostic test accuracy (DTA) studies: the role of cerebral perfusion imaging in prognosis evaluation of mild cognitive impairment. Ann Palliat Med. Feb 2022; 11(2): 673-683. PMID 35249345
  57. Smailagic N, Vacante M, Hyde C, et al. F-FDG PET for the early diagnosis of Alzheimer's disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. Jan 28 2015; 1: CD010632. PMID 25629415
  58. Davison CM, O'Brien JT. A comparison of FDG-PET and blood flow SPECT in the diagnosis of neurodegenerative dementias: a systematic review. Int J Geriatr Psychiatry. Jun 2014; 29(6): 551-61. PMID 24123413
  59. Bloudek LM, Spackman DE, Blankenburg M, et al. Review and meta-analysis of biomarkers and diagnostic imaging in Alzheimer's disease. J Alzheimers Dis. 2011; 26(4): 627-45. PMID 21694448
  60. Yuan Y, Gu ZX, Wei WS. Fluorodeoxyglucose-positron-emission tomography, single-photon emission tomography, and structural MR imaging for prediction of rapid conversion to Alzheimer disease in patients with mild cognitive impairment: a meta-analysis. AJNR Am J Neuroradiol. Feb 2009; 30(2): 404-10. PMID 19001534
  61. Matchar DB, Kulasingam SL, McCrory DC, et al. Use of Positron Emission Tomography and Other Neuroimaging Techniques in the Diagnosis and Management of Alzheimer's Disease and Dementia. Rockville, MD: Agency for Healthcare Research and Quality; 2001.
  62. Motara H, Olusoga T, Russell G, et al. Clinical impact and diagnostic accuracy of 2-[ 18 F]-fluoro-2-deoxy-d-glucose positron-emission tomography/computed tomography (PET/CT) brain imaging in patients with cognitive impairment: a tertiary centre experience in the UK. Clin Radiol. Jan 2017; 72(1): 63-73. PMID 27637430
  63. Moonis G, Subramaniam RM, Trofimova A, et al. ACR Appropriateness Criteria(R) Dementia. J Am Coll Radiol. May 2020; 17(5S): S100-S112. PMID 32370954
  64. Johnson KA, Minoshima S, Bohnen NI, et al. Appropriate use criteria for amyloid PET: a report of the Amyloid Imaging Task Force, the Society of Nuclear Medicine and Molecular Imaging, and the Alzheimer's Association. J Nucl Med. Mar 2013; 54(3): 476-90. PMID 23359661
  65. Owens DK, Davidson KW, Krist AH, et al. Screening for Cognitive Impairment in Older Adults: US Preventive Services Task Force Recommendation Statement. JAMA. Feb 25 2020; 323(8): 757-763. PMID 32096858
  66. Centers for Medicare & Medicaid Services. National Coverage Determination (NCD) for beta amyloid positron tomography in dementia and neurodegenerative disease (220.6.20). 2013; https://www.cms.gov/medicare-coverage-database/details/ncd-details.aspx?NCDId=356&ncdver=1&NCAId=265&bc=AAAAAAAAQAAA&. Accessed September 2, 2022.
  67. Centers for Medicare & Medicaid Services (CMS). Decision Memo for Positron Emission Tomography (FDG) and Other Neuroimaging Devices for Suspected Dementia (CAG-00088R). 2004; https://www.cms.gov/medicare-coverage-database/details/nca-decision-memo.aspx?NCAId=104&NcaName=Positron+Emission+Tomography+(FDG)+and+Other+Neuroimaging+Devices+for+Suspected+Dementia+(1st+Recon)&bc=AiAAAAAAEAAA&. Accessed August 31, 2022.
  68. Centers for Medicare & Medicaid Services (CMS). National Coverage Determination (NCD) for FDG PET for Dementia and Neurodegenerative Diseases (220.6.13). 2009; https://www.cms.gov/medicare-coverage-database/view/ncd.aspx?ncdid=288&ncdver=3&keyword=fdg%20pet&keywordType=starts&areaId=all&docType=NCD&contractOption=all&sortBy=relevance&bc=1. Accessed August 31, 2022.    

Coding Section

Codes

Number

Description

CPT 78608 Brain imaging, positron emission tomography (PET); metabolic evaluation
  78609 Brain imaging, positron emission tomography (PET); perfusion evaluation
  78811 Positron emission tomography (PET) imaging; limited area (e.g., chest, head/neck)
  78814 Positron emission tomography (PET) with concurrently acquired computed tomography (CT) for attenuation correction and anatomical localization imaging; limited area (e.g., chest, head/neck)
HCPCS A9586 Florbetapir F18, diagnostic, per study dose, up to 10 millicuries
  A9599 Radiopharmaceutical, diagnostic, for beta-amyloid positron emission tomography (PET) imaging, per study dose, not otherwise specified
  Q9982 Flutemetamol F18, diagnostic, per study dose, up to 5 millicuries
  Q9983 Florbetaben F18, diagnostic, per study dose, up to 8.1 millicuries
ICD-10-CM   Investigational for relevant diagnoses
  F01.50-F03.C4 Dementia due to known physiological conditions, code range (Revised F01.50, deleted F01.51, Added F01.511-F01.C4; Revised F02.80, deleted F02.81, Added F02.811-F02.C4, Revised F03.90, deleted F03.91, Added F03.911-F03.C4 (eff 10/01/2022)
  G30.0-G30.9 Alzheimer disease, code range
ICD-10-PCS   ICD-10-PCS codes are only used for inpatient services
  C030YZZ Nuclear medicine, central nervous system, positron emission tomography (PET) imaging, brain, other radionuclide
  C030KZZ Nuclear medicine, central nervous system, positron emission tomography (PET) imaging, brain, Fluorine 18
Type of service Radiology  
Place of service Outpatient/inpatient

Procedure and diagnosis codes on Medical Policy documents are included only as a general reference tool for each policy. They may not be all-inclusive.

This medical policy was developed through consideration of peer-reviewed medical literature generally recognized by the relevant medical community, U.S. FDA approval status, nationally accepted standards of medical practice and accepted standards of medical practice in this community, Blue Cross Blue Shield Association technology assessment program (TEC) and other nonaffiliated technology evaluation centers, reference to federal regulations, other plan medical policies, and accredited national guidelines.

"Current Procedural Terminology © American Medical Association. All Rights Reserved" 

History From 2014 Forward     

07/01/2023 Annual review, no change to policy intent. Updating coding, rationale and references.

07/19/2022

Annual review, expanding policy verbiage to additional diagnoses/sympoms. Also updating background, description, rationale and references.

07/01/2021 

Annual review, no change to policy intent. Updating rationale and references. 

07/23/2020 

Annual review, no change to policy intent. Updating description, regulatory status, guidelines, coding, rationale and references. 

07/01/2019 

Annual review, no change to policy intent. Updating background, rationale and references. 

07/16/2018 

Annual review, no change to policy intent. Updating background, rationale and references. 

07/24/2017 

Annual review, no change to policy intent. Updating background, description, rationale and references. 

07/27/2016 

Annual review, no change to policy intent. 

07/23/2015 

Annual review, no change to policy intent. Updated background, description, rationale and references. Added guidelines, benefit application and coding.

07/29/2014

Annual review. Added related policies. Updated background, description, regulatory status, rationale and references. No change to policy intent.

Complementary Content
${loading}