Coverage Policy Manual
Policy #: 2005010
Category: Radiology
Initiated: February 2005
Last Review: December 2023
  Cardiac and Coronary Artery Computed Tomography, CT Derived Fractional Flow Reserve and CT Coronary Calcium Scoring

Description:
Computed tomographic angiography or CTA is a noninvasive imaging test that requires the use of intravenously administered contrast material and high-resolution, high-speed CT machinery to obtain detailed volumetric images of blood vessels. CTA can be applied to image blood vessels throughout the body; however, to apply CTA in the coronary arteries, several technical challenges must be overcome to obtain high-quality diagnostic images. First, very short image acquisition times are necessary to avoid blurring artifacts from the rapid motion of the beating heart. In some cases, premedication with beta-blocking agents is used to slow down the heart rate below about 60–65 beats per minute to facilitate adequate scanning, and electrocardiographic triggering or retrospective gating is used to obtain images during diastole when motion is reduced. Second, rapid scanning is also helpful so that the volume of cardiac images can be obtained during breath-holding. Third, very thin sections (<1 mm) are important to provide adequate spatial resolution and high-quality 3D reconstruction images.
 
Volumetric imaging permits multiplanar reconstruction (MPR) of cross-sectional images to display the coronary arteries. Curved MPR and thin-slab maximum intensity projections (MIPs) provide an overview of the coronary arteries, and volume-rendering techniques (VRT) provide a 3D anatomical display of the exterior of the heart. Quantification of coronary artery stenosis may be difficult given current techniques, though improvements in image reconstruction algorithms such as automatic vessel tracking are being developed.
 
Two different CT technologies can achieve high-speed CT imaging. Electron-beam CT (EBCT, also known as ultrafast CT) uses an electron gun rather than a standard x-ray tube to generate x-rays, thus permitting very rapid scanning, on the order of 50–100 milliseconds per image. Helical CT scanning (also referred to as spiral CT scanning) also creates images at greater speed than conventional CT by continuously rotating a standard x-ray tube around the patient so that data are gathered in a continuous spiral or helix rather than individual slices. Helical CT is able to achieve scan times of 500 milliseconds or less per image and use of partial ring scanning or post-processing algorithms may reduce the effective scan time even further.
 
Multidetector row helical CT scanning (MDCT) or multislice CT (MSCT) is a technological evolution of helical CT, which uses CT machines equipped with an array of multiple x-ray detectors that can simultaneously image multiple sections of the patient during a rapid volumetric image acquisition. Currently available MDCT machines may have 64 or more detectors. Diffusion of MDCT machines into the medical community has been occurring over the past several years.
 
Coronary CTA has been proposed as a noninvasive alternative to invasive coronary angiography. Applications of CTA include (but are not limited to) evaluation of:
    • obstructive coronary artery disease (CAD),
    • coronary artery bypass graft patency
    • coronary artery aneurysm, and
    • congenital coronary artery anomaly.
 
Evaluation of obstructive CAD involves quantifying arterial stenoses to determine whether hemodynamically significant stenosis is present. Symptomatic lesions with greater than 50%–75% diameter stenosis are generally considered significant and often result in revascularization procedures when viable myocardium is present. It has been suggested that CTA may be helpful to rule out the presence of CAD and to avoid invasive coronary angiography in patients with a low clinical likelihood of significant CAD. Also of note is the increasing interest in exploring the role of nonsignificant plaques (i.e., those associated with less than 50% stenosis) because it is postulated that these plaques may undergo rupture or erosion and lead to acute myocardial infarction. Cross-sectional angiographic imaging may visualize the presence and composition of these plaques and quantify the plaque burden better than conventional angiography, which only visualizes the vascular lumen.
 
The information sought from angiography after coronary artery bypass graft surgery may depend on the length of time since surgery. Bypass graft occlusion may occur during the early postoperative period; whereas, over the long term, recurrence of obstructive CAD may occur in the bypass graft, which requires a similar evaluation as CAD in native vessels.
 
Congenital coronary arterial anomalies (i.e., abnormal origination or course of a coronary artery) that lead to clinically significant problems are relatively rare lesions. Symptomatic manifestations may include ischemia or syncope. Clinical presentation of anomalous coronary arteries is hard to distinguish from other more common causes of cardiac disease; however, anomalous coronary artery is an important diagnosis to exclude, particularly in young patients who present with unexplained symptoms (e.g., syncope). There is no specific clinical presentation to suggest a coronary artery aneurysm.
 
CTA has several important limitations. The presence of dense arterial calcification or an intracoronary stent can produce significant beam-hardening artifacts and may preclude a satisfactory study. The presence of an uncontrolled rapid heart rate or arrhythmia hinders the ability to obtain diagnostically satisfactory images. EBCT is able to achieve satisfactory images of the proximal and mid-segment coronary vessels without significant motion artifact in 92% of cases; whereas, MDCT was successful in only 73% of cases (p<0.001). Evaluation of the distal coronary arteries is generally more difficult than visualization of the proximal and mid-segment coronary arteries due to greater cardiac motion and the smaller caliber of coronary vessels in distal locations.
 
It is important to consider the radiation dose associated with CTA. In comparison, 4-row MDCT delivers approximately 8 – 12 mSv; EBCT delivers approximately 1.5 to 2.0 mSv, and conventional invasive coronary angiography delivers about 4 – 8 mSv.  Higher row MDCT with modulation of the x-ray beam to avoid exposure during nonimaging phases of the cardiac cycle may reduce x-ray dosage.
 
Multidetector computed tomography of the heart (MDCT) and coronary computed tomographic angiography (CCTA) for coronary artery evaluation studies are reported with new Category III CPT Codes, CPT 0144T, 0145T, 0146T, 0147T, 0148T, 0149T, 0150T, and 0151T prior to 1/1/2010. Effective 1/1/2010, new CPT codes 75571-75574 should be reported.  Prior to 2006, these procedures were reported with CPT 76497 (unlisted CT procedure), and HCPCS codes S8092 and S8093.
 
With the introduction of the 64 slice, sub-millimeter thin slice detectors CT scanners, imaging of the cardiac structure and morphology is possible, and the negative predictive value for coronary artery studies approaches 99%.  (Hoffmann, Martin H, et. al.)
 
The use of electron-beam CT or helical CT to detect coronary artery calcification is addressed in a separate policy No. 1997061, Coronary Artery Quantitative Calcium Scoring Using Electron Beam Computed Tomography or Multidetector Helical or Spiral Computed Tomography
 
CPT code 75571 (Computed tomography, heart, without contrast material, with quantitative evaluation of coronary calcium) is the appropriate code to bill when the study is done prior to CT coronary angiography to determine if there is too much calcium to do CT coronary angiography.  The code is billed only when the CTA cannot be completed.
 
CAD Risk Assessment and Pre-test Probability
The following references (not an inclusive list) provide information on assessing CAD risk and pre-test probability.
 
Kane SP. ASCVD Risk Calculator: 10-Year Risk of First Cardiovascular Event Using Pooled Cohort Equations. ClinCalc: https://clincalc.com/Cardiology/ASCVD/PooledCohort.aspx. Updated August 6, 2020. Accessed at:  https://clincalc.com/Cardiology/ASCVD/PooledCohort.aspx
 
Pretest Probability of Coronary Artery Disease by Age, Gender, and Symptoms (Table 2) in
Gibbons RJ, Balady GJ, Beasley JW, et al. ACC/AHA Guidelines for Exercise Testing: Executive Summary. Circulation. 1997;96:345-354. Accessed at:  https://www.ahajournals.org/doi/full/10.1161/01.cir.96.1.345
 
 
CHART (eff April 09, 2023): Pretest Probability (%) of Coronary Artery Disease by Age, Gender, and Symptoms
 
Definitions:
Cardiac chest pain is centrally located, provoked by stress (exercise or emotional), and relieved by rest
Possible cardiac chest pain has two of the three characteristics associated with cardiac chest pain
Non-cardiac chest pain has one (or none) of the three characteristics associated with cardiac chest pain
 
30-39 years old
        • Cardiac
            • Male 3%
            • Female 5%
        • Possible cardiac
            • Male 4%
            • Female 3%
        • Noncardiac
            • Male 1%
            • Female 1%
        • Dyspnea without chest pain
            • Male 0%
            • Female 3%
40-49 years old
        • Cardiac
            • Male 22%
            • Female 10%
        • Possible cardiac
            • Male 10%
            • Female 6%
        • Noncardiac
            • Male 3%
            • Female 2%
        • Dyspnea without chest pain
            • Male 12%
            • Female 3%
50-59 years old
        • Cardiac
            • Male 32%
            • Female 13%
        • Possible cardiac
            • Male 17%
            • Female 6%
        • Noncardiac
            • Male 11%
            • Female 3%
        • Dyspnea without chest pain
            • Male 20%
            • Female 9%
60-79 years old
        • Cardiac
            • Male 44%
            • Female 16%
        • Possible cardiac
            • Male 26%
            • Female 11%
        • Noncardiac
            • Male 22%
            • Female 6%
        • Dyspnea without chest pain
            • Male 27%
            • Female 14%
70+ years old
        • Cardiac
            • Male 52%
            • Female 27%
        • Possible cardiac
            • Male 34%
            • Female 19%
        • Noncardiac
            • Male 24%
            • Female 10%
        • Dyspnea without chest pain
            • Male 32%
            • Female 12%
 
Adapted from Knuuti J, Wijns W, Saraste A, et al. 2019 ESC guidelines for the diagnosis and management of chronic coronary syndromes. Eur Heart J. 2020;41: 407–477.

Policy/
Coverage:
To Be Effective April 14, 2024
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Coronary CT Angiography (CCTA)
(See Description section for information on assessment of pre-test probability and CAD risk)
 
The following indications meet primary coverage criteria for Coronary CT Angiography (CCTA).
 
A_Suspected CAD in symptomatic patients who have not had evaluation for CAD within the preceding 60 days
    • Chest pain with or without other symptoms of myocardial ischemia
        • With pretest probability of CAD > 15% (See chart in Description)
    • Patients without chest pain whose predominant symptom is dyspnea
        • With pretest probability of CAD > 15% (See chart in Description)
    • Patients with any cardiac symptom who have diseases/conditions with which CAD commonly coexists, such as ANY of the following:
        • Abdominal aortic aneurysm
        • Established and symptomatic peripheral vascular disease
        • Prior history of stroke, transient ischemic attack (TIA), carotid endarterectomy (CEA), or high-grade carotid stenosis (> 70%)
        • Chronic kidney disease
B_Established flow-limiting CAD in patients who have new or worsening symptoms
    • Patients whose symptoms persist despite maximal anti-ischemic medical therapy or contraindication thereto
        • Patients with established CAD and typical angina pectoris despite maximal anti-ischemic therapy may be better served with invasive coronary angiography
C_Established or suspected CAD
    • Patients who have undergone cardiac transplantation
        • With new or worsening cardiac symptoms
        • With new or worsening physical examination abnormalities
        • Clinically stable patients who have not had evaluation for CAD in the preceding year
    • Patients (symptomatic or asymptomatic) with new onset arrhythmias who have not had evaluation for CAD since the arrhythmia was recognized
        • Patients with sustained (lasting more than 30 seconds) or nonsustained (more than 3 beats but terminating within 30 seconds) ventricular tachycardia
        • Patients with atrial fibrillation or flutter and high or intermediate risk of CAD (using ASCVD Pooled Cohort Equations)
        • Patients with atrial fibrillation or flutter and established CAD
        • Patients who have frequent premature ventricular contractions (PVC) defined as more than 30 PVCs per hour on ambulatory EKG (Holter) monitoring
            • CCTA is not clinically indicated for evaluation of infrequent premature atrial or ventricular depolarizations
    • Patients (symptomatic or asymptomatic) with new onset congestive heart failure (CHF) or recently recognized LV systolic dysfunction who have not had evaluation for CAD since the onset of LV dysfunction/CHF
        • For patients in this category with established CAD, or those with suspected CAD whose CAD risk (using ASCVD Pooled Cohort Equations) is high, coronary angiography may be more appropriate than noninvasive evaluation.
    • Abnormal resting EKG
        • Patients with ANY of the following newly recognized and not previously evaluated resting EKG changes:
            • Left bundle branch block
            • ST depression > or = 1 mm
            • Left ventricular (LV) hypertrophy with repolarization abnormality
        • Patients who would otherwise undergo exercise EKG testing (without imaging) but have ANY of the following resting EKG findings that would render the interpretation of an exercise EKG test difficult or impossible:
            • Left bundle branch block
            • Ventricular paced rhythm
            • Left ventricular hypertrophy with repolarization abnormality
            • Digoxin effect
            • ST depression > or = 1 mm on a recent EKG (within the past 30 days)
            • Pre-excitation syndromes (e.g., Wolff-Parkinson-White syndrome)
    • Patients with abnormal exercise treadmill test (performed without imaging) who have not undergone evaluation for CAD since the treadmill test
        • Abnormal findings on an exercise treadmill test (includes chest pain, ST segment change, abnormal blood pressure response, or complex ventricular arrhythmias)
    • Patients who have undergone recent (within the past 60 days) stress testing with adjunctive imaging (MPI, SE, perfusion PET, stress MRI)
        • When the stress imaging test is technically suboptimal, technically limited, inconclusive, indeterminate, or equivocal, such that myocardial ischemia cannot be adequately excluded
            • A stress imaging test is deemed to be abnormal when there are abnormalities on the imaging portion of the test. Electrocardiographic abnormalities without imaging evidence of ischemia do not render a stress imaging test abnormal.
        • When the stress imaging test is abnormal and ALL of the following apply:
            • The stress test demonstrates moderate or severe ischemia
            • CCTA is requested to exclude left main CAD
            • In the absence of left main CAD GDMT will be instituted
            • Invasive coronary angiography will be reserved for persistent symptoms on GDMT Guideline-directed medical therapy (GDMT)
    • Preoperative cardiac evaluation of patients undergoing non-cardiac surgery (includes surveillance for CAD in patients awaiting solid organ transplant)
It is assumed that those who require emergency surgery will undergo inpatient preoperative evaluation.
For patients with active cardiac conditions such as unstable coronary syndromes (unstable angina), decompensated heart failure (NYHA class IV, worsening or new onset heart failure), significant arrhythmias (third degree AV block Mobitz II AV block, uncontrolled supraventricular arrhythmia, symptomatic ventricular arrhythmias, ventricular tachycardia), symptomatic bradycardia or severe stenotic valvular lesions, it is recommended that these conditions be evaluated and managed per ACC/AHA guidelines prior to considering elective surgery. That evaluation may include CCTA.
        • Low-risk surgery (endoscopic procedures, superficial procedures, cataract surgery, breast surgery, ambulatory surgery)
            • Provided that there are no active cardiac conditions (as outlined above), CCTA prior to low-risk surgery is considered not medically necessary
        • Intermediate-risk surgery (including but not limited to intraperitoneal and intrathoracic surgery, carotid endarterectomy, head and neck surgery, orthopedic surgery, prostate surgery, gastric bypass surgery) or high-risk surgery (including but not limited to aortic and other major vascular surgery, peripheral vascular surgery) when BOTH of the following apply:
            • Patient has not had a negative evaluation for CAD or a coronary revascularization procedure within the previous one (1) year
            • At least ONE of the following applies:
                • Patient has established CAD (prior MI, prior PCI or CABG) or presumed CAD (Q waves on EKG, abnormal MPI, SE, or cardiac PET)
                • Patient has compensated heart failure or prior history of CHF
                • Patient has diabetes mellitus
                • Patient has chronic kidney disease
                • Patient has a history of cerebrovascular disease (TIA, stroke, or documented carotid stenosis requiring carotid endarterectomy)
                • Patient is unable to walk on a treadmill for reasons other than obesity
        • Patients awaiting solid organ transplant
            • Asymptomatic patients who have not undergone evaluation for CAD within the preceding one (1) year
            • Patients with symptoms consistent with myocardial ischemia
    • Preoperative evaluation for patients undergoing non-coronary cardiac surgery
        • Patients undergoing evaluation for transcatheter aortic valve implantation/replacement (TAVI or TAVR) at low risk for CAD (using ASCVD Pooled Cohort Equations) to avoid invasive angiography, where all the necessary preoperative information can be obtained using cardiac CT
        • Patients undergoing evaluation for valve surgery (not including TAVR) at low or intermediate risk for CAD (using ASCVD Pooled Cohort Equations)
D_Miscellaneous indications for CCTA
    • Inability to perform exercise EKG test
        • Patients who would otherwise undergo exercise EKG testing (without imaging) but are unable (for reasons other than obesity) to perform exercise to a degree that would yield a diagnostic test. This provision includes patients with musculoskeletal, neurological or pulmonary limitation.
    • Kawasaki disease
        • Periodic surveillance up to one year following diagnosis of Kawasaki disease when previous imaging study reveals ANY of the following:
            • Coronary abnormalities
            • Left ventricular dysfunction
            • Pericardial effusion
            • Valvular regurgitation (other than trace or trivial regurgitation)
            • Aortic dilation
        • Annual evaluation in patients with an established diagnosis of Kawasaki disease who have small or medium-sized coronary artery aneurysms
        • Semiannual evaluation (every 6 months) in patients with an established diagnosis of Kawasaki disease who have large or giant coronary artery aneurysms, or coronary artery obstruction
    • Congenital coronary artery anomalies
        • For evaluation of suspected congenital anomalies of the coronary arteries in ANY of the following scenarios:
            • Exertional syncope
            • History of anomalous coronary artery in a first -degree relative
            • Following coronary angiography which failed to adequately define the origin or course of a coronary artery
            • Coronary ostia appear to be abnormally positioned on echocardiography
 
Fractional Flow Reserve (FFR-CT) (now a separate section)
 
The following indications meet primary coverage criteria for FFR-CT when ALL of the following criteria are met:
    • The patient has symptoms consistent with myocardial ischemia
    • Symptoms persist despite maximal GDMT
    • CCTA has been performed in the preceding 90 days
    • There is at least one 40%-90% coronary stenosis located in the proximal or middle segment of a major native coronary artery or a named branch thereof
 
CT Coronary Calcium Scoring
 
Quantitative coronary artery calcium scoring meets primary coverage criteria of effectiveness when performed:
    • In a patient who meets criteria for CT coronary angiography to determine if there is too much calcium present to proceed with CT coronary angiography.  If CT coronary angiography is performed, the CT calcium scoring is considered part of the CT coronary angiogram; OR
    • As part of a pre-operative evaluation for orthotopic liver transplantation.
 
CT for Cardiac Structure
 
The following indications meet primary coverage criteria for CT for Cardiac Structure:
 
A_Congenital heart disease
      • Evaluation of suspected or established congenital heart disease in patients whose echocardiogram is technically limited or non-diagnostic; OR
      • Further evaluation of patients whose echocardiogram suggests a new diagnosis of complex congenital heart disease; OR
      • Evaluation of complex congenital heart disease in patients who are less than one year post-surgical correction; OR
      • Evaluation of complex congenital heart disease in patients who have new or worsening symptoms and/or a change in physical examination; OR
      • Assist in surgical planning for patients with complex congenital heart disease; OR
      • Surveillance in asymptomatic patients with complex congenital heart disease who have not had cardiac MRI or cardiac CT within the preceding year.
B_Cardiomyopathy
      • Evaluation of patients with suspected arrhythmogenic right ventricular dysplasia  (ARVD) who have ANY of the following:   
          • Severe right ventricular dysfunction on another cardiac imaging study
          • Precordial T wave inversion not associated with RBBB
          • First-degree relative with established ARVD or unexplained sudden cardiac death at age younger than 35 years
          • Ventricular tachycardia or frequent PVCs (> 500 in 24 hours or > 30 per hour); OR
      • To assess LV function in patients with suspected or established cardiomyopathy when all other non-invasive imaging is not feasible or technically suboptimal; OR
      • To assess RV function in patients with suspected RV dysfunction when all other non-invasive imaging is not feasible or technically suboptimal
C_Valvular heart disease
      • Evaluation of suspected dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
      • Evaluation of established dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
D_Evaluation of patients with established CAD
      • Non-invasive localization of coronary bypass grafts or potential grafts (including internal mammary artery) and/or evaluation of retrosternal anatomy in patients undergoing repeat surgical revascularization
E_Intra-cardiac and para-cardiac masses and tumors:
      • Patients with a suspected cardiac or para-cardiac mass (thrombus, tumor, etc.) suggested by transthoracic echocardiography, transesophageal echocardiography, blood pool imaging or contrast ventriculography who have not undergone cardiac CT or cardiac MRI within the preceding 60 days; OR
      • Patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically unstable; OR
      • In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically stable and have not undergone cardiac CT or cardiac MRI within the preceding year; OR
      • Patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who have undergone treatment (chemotherapy, radiation therapy, thrombolysis, anticoagulation or surgery) within the preceding year and have not had cardiac CT or cardiac MRI within the preceding 60 days
F_ Left atrial appendage closure device
      • For evaluation of cardiac anatomy prior to implantation of a left atrial appendage closure device; OR
      • Following placement of a left atrial appendage closure device, a single study may be performed as an alternative to TEE to assess for intracardiac thrombus
G_Cardiac aneurysm and pseudoaneurysm
 
H_Evaluation of pericardial conditions (pericardial effusion, constrictive pericarditis, or congenital pericardial diseases):
      • Patients with suspected pericardial constriction; OR
      • Patients with suspected congenital pericardial disease; OR
      • Patients with suspected pericardial effusion who have undergone echocardiography deemed to be technically suboptimal in evaluation of the effusion; OR
      • Patients whose echocardiogram shows a complex pericardial effusion (loculated, containing solid material)
I_Evaluation of cardiac venous anatomy
      • For localization of the pulmonary veins in patients with chronic or paroxysmal atrial fibrillation/flutter who are being considered for ablation; OR
      • Coronary venous localization prior to implantation of a biventricular pacemaker
J_Evaluation of the thoracic aorta
      • Patients with suspected thoracic aortic aneurysm/dilation who have not undergone CT or MRI of the thoracic aorta within the preceding 60 days; OR
      • Patients with confirmed thoracic aortic aneurysm / dilation with new or worsening signs/symptoms; OR
      • Ongoing surveillance of stable patients with confirmed thoracic aortic aneurysm/dilation who have not undergone surgical repair and have not had imaging of the thoracic aorta within the preceding six months; OR
      • Patients with suspected aortic dissection; OR
      • Patients with confirmed aortic dissection who have new or worsening symptoms; OR
      • Patients with confirmed aortic dissection in whom surgical repair is anticipated (to assist in preoperative planning); OR
      • Ongoing surveillance of stable patients with confirmed aortic dissection who have not undergone imaging of the thoracic aorta within the preceding year; OR
      • Patients with confirmed aortic dissection or thoracic aortic aneurysm / dilation who have undergone surgical repair within the preceding year and have not undergone imaging of the thoracic aorta within the preceding six months; OR
      • Patients who have sustained blunt chest trauma, penetrating aortic trauma or iatrogenic trauma as a result of aortic instrumentation; OR
      • Patients being evaluated for potential transcatheter aortic valve implantation/replacement (TAVI or TAVR) provided that the patient has not undergone cardiac CT or cardiac MRI within the preceding 60 days
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
For Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed as covered above including screening, does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes.  
 
For members with contracts without primary coverage criteria, Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed as covered above including screening, is considered investigational. Investigational services are Plan exclusions.
 
Effective April 09, 2023 - April 13, 2024
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Coronary CT Angiography (CCTA)
(See Description section for information on assessment of pre-test probability and CAD risk)
 
The following indications meet primary coverage criteria for Coronary CT Angiography (CCTA).
 
A_Suspected CAD in symptomatic patients who have not had evaluation for CAD within the preceding 60 days
    • Chest pain with or without other symptoms of myocardial ischemia
        • With pretest probability of CAD > 15% (See chart in Description)
    • Patients without chest pain whose predominant symptom is dyspnea
        • With pretest probability of CAD > 15% (See chart in Description)
    • Patients with any cardiac symptom who have diseases/conditions with which CAD commonly coexists, such as ANY of the following:
        • Abdominal aortic aneurysm
        • Established and symptomatic peripheral vascular disease
        • Prior history of stroke, transient ischemic attack (TIA), carotid endarterectomy (CEA), or high-grade carotid stenosis (> 70%)
        • Chronic kidney disease
B_Established flow-limiting CAD in patients who have new or worsening symptoms
    • Patients whose symptoms persist despite maximal anti-ischemic medical therapy or contraindication thereto
        • Patients with established CAD and typical angina pectoris despite maximal anti-ischemic therapy may be better served with invasive coronary angiography
C_Established or suspected CAD
    • Patients who have undergone cardiac transplantation
        • With new or worsening cardiac symptoms
        • With new or worsening physical examination abnormalities
        • Clinically stable patients who have not had evaluation for CAD in the preceding year
    • Patients (symptomatic or asymptomatic) with new onset arrhythmias who have not had evaluation for CAD since the arrhythmia was recognized
        • Patients with sustained (lasting more than 30 seconds) or nonsustained (more than 3 beats but terminating within 30 seconds) ventricular tachycardia
        • Patients with atrial fibrillation or flutter and high or intermediate risk of CAD (using ASCVD Pooled Cohort Equations)
        • Patients with atrial fibrillation or flutter and established CAD
        • Patients who have frequent premature ventricular contractions (PVC) defined as more than 30 PVCs per hour on ambulatory EKG (Holter) monitoring
            • CCTA is not clinically indicated for evaluation of infrequent premature atrial or ventricular depolarizations
    • Patients (symptomatic or asymptomatic) with new onset congestive heart failure (CHF) or recently recognized LV systolic dysfunction who have not had evaluation for CAD since the onset of LV dysfunction/CHF
        • For patients in this category with established CAD, or those with suspected CAD whose CAD risk (using ASCVD Pooled Cohort Equations) is high, coronary angiography may be more appropriate than noninvasive evaluation.
    • Abnormal resting EKG
        • Patients with ANY of the following newly recognized and not previously evaluated resting EKG changes:
            • Left bundle branch block
            • ST depression > or = 1 mm
            • Left ventricular (LV) hypertrophy with repolarization abnormality
        • Patients who would otherwise undergo exercise EKG testing (without imaging) but have ANY of the following resting EKG findings that would render the interpretation of an exercise EKG test difficult or impossible:
            • Left bundle branch block
            • Ventricular paced rhythm
            • Left ventricular hypertrophy with repolarization abnormality
            • Digoxin effect
            • ST depression > or = 1 mm on a recent EKG (within the past 30 days)
            • Pre-excitation syndromes (e.g., Wolff-Parkinson-White syndrome)
    • Patients with abnormal exercise treadmill test (performed without imaging) who have not undergone evaluation for CAD since the treadmill test
        • Abnormal findings on an exercise treadmill test (includes chest pain, ST segment change, abnormal blood pressure response, or complex ventricular arrhythmias)
    • Patients who have undergone recent (within the past 60 days) stress testing with adjunctive imaging (MPI, SE, perfusion PET, stress MRI)
        • When the stress imaging test is technically suboptimal, technically limited, inconclusive, indeterminate, or equivocal, such that myocardial ischemia cannot be adequately excluded
            • A stress imaging test is deemed to be abnormal when there are abnormalities on the imaging portion of the test. Electrocardiographic abnormalities without imaging evidence of ischemia do not render a stress imaging test abnormal.
        • When the stress imaging test is abnormal and ALL of the following apply:
            • The stress test demonstrates moderate or severe ischemia
            • CCTA is requested to exclude left main CAD
            • In the absence of left main CAD GDMT will be instituted
            • Invasive coronary angiography will be reserved for persistent symptoms on GDMT Guideline-directed medical therapy (GDMT)
    • Preoperative cardiac evaluation of patients undergoing non-cardiac surgery (includes surveillance for CAD in patients awaiting solid organ transplant)
It is assumed that those who require emergency surgery will undergo inpatient preoperative evaluation.
For patients with active cardiac conditions such as unstable coronary syndromes (unstable angina), decompensated heart failure (NYHA class IV, worsening or new onset heart failure), significant arrhythmias (third degree AV block Mobitz II AV block, uncontrolled supraventricular arrhythmia, symptomatic ventricular arrhythmias, ventricular tachycardia), symptomatic bradycardia or severe stenotic valvular lesions, it is recommended that these conditions be evaluated and managed per ACC/AHA guidelines prior to considering elective surgery. That evaluation may include CCTA.
        • Low-risk surgery (endoscopic procedures, superficial procedures, cataract surgery, breast surgery, ambulatory surgery)
            • Provided that there are no active cardiac conditions (as outlined above), CCTA prior to low-risk surgery is considered not medically necessary
        • Intermediate-risk surgery (including but not limited to intraperitoneal and intrathoracic surgery, carotid endarterectomy, head and neck surgery, orthopedic surgery, prostate surgery, gastric bypass surgery) or high-risk surgery (including but not limited to aortic and other major vascular surgery, peripheral vascular surgery) when BOTH of the following apply:
            • Patient has not had a negative evaluation for CAD or a coronary revascularization procedure within the previous one (1) year
            • At least ONE of the following applies:
                • Patient has established CAD (prior MI, prior PCI or CABG) or presumed CAD (Q waves on EKG, abnormal MPI, SE, or cardiac PET)
                • Patient has compensated heart failure or prior history of CHF
                • Patient has diabetes mellitus
                • Patient has chronic kidney disease
                • Patient has a history of cerebrovascular disease (TIA, stroke, or documented carotid stenosis requiring carotid endarterectomy)
                • Patient is unable to walk on a treadmill for reasons other than obesity
        • Patients awaiting solid organ transplant
            • Asymptomatic patients who have not undergone evaluation for CAD within the preceding one (1) year
            • Patients with symptoms consistent with myocardial ischemia
    • Preoperative evaluation for patients undergoing non-coronary cardiac surgery
        • Patients undergoing evaluation for transcatheter aortic valve implantation/replacement (TAVI or TAVR) at low risk for CAD (using ASCVD Pooled Cohort Equations) to avoid invasive angiography, where all the necessary preoperative information can be obtained using cardiac CT
        • Patients undergoing evaluation for valve surgery (not including TAVR) at low or intermediate risk for CAD (using ASCVD Pooled Cohort Equations)
D_Miscellaneous indications for CCTA
    • Inability to perform exercise EKG test
        • Patients who would otherwise undergo exercise EKG testing (without imaging) but are unable (for reasons other than obesity) to perform exercise to a degree that would yield a diagnostic test. This provision includes patients with musculoskeletal, neurological or pulmonary limitation.
    • Kawasaki disease
        • Periodic surveillance up to one year following diagnosis of Kawasaki disease when previous imaging study reveals ANY of the following:
            • Coronary abnormalities
            • Left ventricular dysfunction
            • Pericardial effusion
            • Valvular regurgitation (other than trace or trivial regurgitation)
            • Aortic dilation
        • Annual evaluation in patients with an established diagnosis of Kawasaki disease who have small or medium-sized coronary artery aneurysms
        • Semiannual evaluation (every 6 months) in patients with an established diagnosis of Kawasaki disease who have large or giant coronary artery aneurysms, or coronary artery obstruction
    • Congenital coronary artery anomalies
        • For evaluation of suspected congenital anomalies of the coronary arteries in ANY of the following scenarios:
            • Exertional syncope
            • History of anomalous coronary artery in a first -degree relative
            • Following coronary angiography which failed to adequately define the origin or course of a coronary artery
            • Coronary ostia appear to be abnormally positioned on echocardiography
 
Fractional Flow Reserve (FFR-CT) (now a separate section)
 
The following indications meet primary coverage criteria for FFR-CT when ALL of the following criteria are met:
    • The patient has symptoms consistent with myocardial ischemia
    • Symptoms persist despite maximal GDMT
    • CCTA has been performed in the preceding 90 days
    • There is at least one 40%-90% coronary stenosis located in the proximal or middle segment of a major native coronary artery or a named branch thereof
 
CT Coronary Calcium Scoring
 
Quantitative coronary artery calcium scoring meets primary coverage criteria of effectiveness when performed
 
    • In a patient who meets criteria for CT coronary angiography to determine if there is too much calcium present to proceed with CT coronary angiography.  If CT coronary angiography is performed, the CT calcium scoring is considered part of the CT coronary angiogram; OR
    • As part of a pre-operative evaluation for orthotopic liver transplantation.
 
CT for Cardiac Structure
 
The following indications meet primary coverage criteria for CT for Cardiac Structure:
 
A_Congenital heart disease
      • Evaluation of suspected or established congenital heart disease in patients whose echocardiogram is technically limited or non-diagnostic; OR
      • Further evaluation of patients whose echocardiogram suggests a new diagnosis of complex congenital heart disease; OR
      • Evaluation of complex congenital heart disease in patients who are less than one year post-surgical correction; OR
      • Evaluation of complex congenital heart disease in patients who have new or worsening symptoms and/or a change in physical examination; OR
      • Assist in surgical planning for patients with complex congenital heart disease; OR
      • Surveillance in asymptomatic patients with complex congenital heart disease who have not had cardiac MRI or cardiac CT within the preceding year.
B_Cardiomyopathy
      • Evaluation of patients with suspected arrhythmogenic right ventricular dysplasia; OR
      • To assess LV function in patients with suspected or established cardiomyopathy when all other non-invasive imaging is not feasible or technically suboptimal; OR
      • To assess RV function in patients with suspected RV dysfunction when all other non-invasive imaging is not feasible or technically suboptimal
C_Valvular heart disease
      • Evaluation of suspected dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
      • Evaluation of established dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
D_Evaluation of patients with established CAD
      • Non-invasive localization of coronary bypass grafts or potential grafts (including internal mammary artery) and/or evaluation of retrosternal anatomy in patients undergoing repeat surgical revascularization
E_Intra-cardiac and para-cardiac masses and tumors:
      • Patients with a suspected cardiac or para-cardiac mass (thrombus, tumor, etc.) suggested by transthoracic echocardiography, transesophageal echocardiography, blood pool imaging or contrast ventriculography who have not undergone cardiac CT or cardiac MRI within the preceding 60 days; OR
      • Patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically unstable; OR
      • In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically stable and have not undergone cardiac CT or cardiac MRI within the preceding year; OR
      • Patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who have undergone treatment (chemotherapy, radiation therapy, thrombolysis, anticoagulation or surgery) within the preceding year and have not had cardiac CT or cardiac MRI within the preceding 60 days
F_ Left atrial appendage closure device
      • For evaluation of cardiac anatomy prior to implantation of a left atrial appendage closure device; OR
      • Following placement of a left atrial appendage closure device, a single study may be performed as an alternative to TEE to assess for intracardiac thrombus
G_Cardiac aneurysm and pseudoaneurysm
 
H_Evaluation of pericardial conditions (pericardial effusion, constrictive pericarditis, or congenital pericardial diseases):
      • Patients with suspected pericardial constriction; OR
      • Patients with suspected congenital pericardial disease; OR
      • Patients with suspected pericardial effusion who have undergone echocardiography deemed to be technically suboptimal in evaluation of the effusion; OR
      • Patients whose echocardiogram shows a complex pericardial effusion (loculated, containing solid material)
I_Evaluation of cardiac venous anatomy
      • For localization of the pulmonary veins in patients with chronic or paroxysmal atrial fibrillation/flutter who are being considered for ablation; OR
      • Coronary venous localization prior to implantation of a biventricular pacemaker
J_Evaluation of the thoracic aorta
      • Patients with suspected thoracic aortic aneurysm/dilation who have not undergone CT or MRI of the thoracic aorta within the preceding 60 days; OR
      • Patients with confirmed thoracic aortic aneurysm / dilation with new or worsening signs/symptoms; OR
      • Ongoing surveillance of stable patients with confirmed thoracic aortic aneurysm/dilation who have not undergone surgical repair and have not had imaging of the thoracic aorta within the preceding six months; OR
      • Patients with suspected aortic dissection; OR
      • Patients with confirmed aortic dissection who have new or worsening symptoms; OR
      • Patients with confirmed aortic dissection in whom surgical repair is anticipated (to assist in preoperative planning); OR
      • Ongoing surveillance of stable patients with confirmed aortic dissection who have not undergone imaging of the thoracic aorta within the preceding year; OR
      • Patients with confirmed aortic dissection or thoracic aortic aneurysm / dilation who have undergone surgical repair within the preceding year and have not undergone imaging of the thoracic aorta within the preceding six months; OR
      • Patients who have sustained blunt chest trauma, penetrating aortic trauma or iatrogenic trauma as a result of aortic instrumentation; OR
      • Patients being evaluated for potential transcatheter aortic valve implantation/replacement (TAVI or TAVR) provided that the patient has not undergone cardiac CT or cardiac MRI within the preceding 60 days
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
For Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed as covered above including screening, does not meet primary coverage criteria.
 
For contracts without primary coverage criteria, for Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed as covered above including screening, is considered investigational. Investigational services are specific contract exclusions.
 
Effective March 13, 2022 to April 08, 2023
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Coronary CT Angiography and FFR
(See Description section for information on CAD risk assessment of pre-test probability)
 
The following indications meet primary coverage criteria for Coronary CT Angiography (CCTA). For some of these indications, FFR-CT may also meet primary coverage criteria.  For some indications, coronary CT meets coverage criteria only if the imaging facility has the capability to perform FFR.
 
1.Coronary CT Angiography only:
    • For evaluation of suspected congenital anomalies of the coronary arteries
 
2.Coronary CT Angiography, with FFR when necessary.    
FFR measurement by CT may be considered medically necessary for the following indications if needed to further assess coronary artery disease of uncertain physiological significance seen on the coronary CT angiogram:
      • Congestive Heart Failure/cardiomyopathy/left ventricular dysfunction:
          • For exclusion of CAD in patients with left ventricular ejection fraction < 55% and low to moderate coronary heart disease risk (using a standard method of risk assessment, such as the SCORE or ACC/AHA model) in whom CAD has not been excluded as the etiology of the cardiomyopathy.
      • For preoperative evaluation of patients undergoing non-coronary artery cardiac surgery:
          • Patients undergoing evaluation for transcatheter aortic valve implantation/replacement (TAVI or TAVR) at low risk for CAD (using an ASCVD Pooled Cohort Equation for risk assessment such as ACC/AHA model) to avoid invasive angiography, where all the necessary preoperative information can be obtained using cardiac CT
          • Patients undergoing evaluation for valve surgery (not including TAVR) at low or intermediate risk for CAD (using an ASCVD Pooled Cohort Equation for risk assessment such as ACC/AHA model)
      • Suspected CAD in patients who have had abnormal exercise EKG test (performed without imaging) within the past 60 days when BOTH of the following criteria are met:
          • Patient is symptomatic*
          • During testing the patient had exercise-induced chest pain, ST segment change, abnormal BP response or complex ventricular arrhythmias
      • Suspected CAD in patients who have and an equivocal myocardial perfusion imaging or stress echo within the past 60 days, when BOTH of the following criteria are met:
          • Patient is symptomatic*
          • The imaging portion of the study is neither clearly normal nor clearly abnormal.
      • Suspected CAD in patients who have had abnormal myocardial perfusion imaging or stress echo within the past 60 days when BOTH of the following criteria are met:
          • Patient is symptomatic*
          • The imaging portion of the study is abnormal
 
3.Coronary CT Angiography, with FFR when necessary. For these indications, the imaging center MUST have the capability to perform FFR-CT if indicated.
FFR measurement by CT may be considered medically necessary for the following indications if needed to further assess coronary artery disease of uncertain physiological significance seen on the coronary CT angiogram:   
        • Suspected CAD in symptomatic* patients who have an abnormal resting EKG:  
            • When resting EKG abnormalities (paced ventricular rhythm, left ventricular hypertrophy with repolarization abnormalities, resting ST depression of 1 mm or more, digoxin effect, or pre-excitation syndrome) would render an exercise treadmill test (without imaging) uninterpretable
        • Suspected CAD in symptomatic* patients who have not had recent CAD evaluation:
            • When no CAD imaging evaluation has been performed within the preceding 60 days  
Notes:
*For purposes of this policy, a patient is considered to be “symptomatic” when one of the following applies:
      • Chest pain
          • With intermediate or high pretest probability of CAD; OR
          • With low or very low pretest probability of CAD and high risk of CAD (using an ASCVD Pooled Cohort Equation for risk assessment such as ACC/AHA model)
      • Atypical symptoms: shortness of breath (dyspnea), neck, jaw, arm, epigastric or back pain, sweating (diaphoresis), or exercise-induced syncope
          • With moderate or high risk of CAD (based on a standard method of risk assessment, such as the SCORE or ACC/AHA model)
      • Other symptoms: palpitation, nausea, vomiting, anxiety, weakness, fatigue, or exercise-induced dizziness, lightheadedness or near syncope, etc.
          • With high risk of CAD (using an ASCVD Pooled Cohort Equation for risk assessment such as ACC/AHA model)
      • Patients with any cardiac symptom who have diseases/conditions with which CAD commonly coexists, such as:
          • Abdominal aortic aneurysm; OR
          • Chronic renal insufficiency or renal failure; OR
          • Diabetes mellitus; OR
          • Established and symptomatic peripheral vascular disease; OR
          • Prior history of cerebrovascular accident
*Performance of multiple non-invasive coronary artery or myocardial perfusion imaging studies on the same patient should be rarely required and will be subject to monitoring.
 
CT Coronary Calcium Scoring
 
Quantitative coronary artery calcium scoring meets primary coverage criteria of effectiveness when:
    • Performed in a patient who meets criteria for CT coronary angiography to determine if there is too much calcium present to proceed with CT coronary angiography.  If CT coronary angiography is performed, the CT calcium scoring is considered part of the CT coronary angiogram; OR
    • Performed as part of a pre-operative evaluation for orthotopic liver transplantation.
 
CT for Cardiac Structure
 
The following indications meet primary coverage criteria for CT for Cardiac Structure:  
A.Congenital heart disease
      • For evaluation of suspected or established congenital heart disease in patients whose echocardiogram is technically limited or non-diagnostic; OR
      • For further evaluation of patients whose echocardiogram suggests a new diagnosis of complex congenital heart disease; OR
      • For evaluation of complex congenital heart disease in patients who are less than one year post-surgical correction; OR
      • For evaluation of complex congenital heart disease in patients who have new or worsening symptoms and/or a change in physical examination; OR
      • To assist in surgical planning for patients with complex congenital heart disease; OR
      • For surveillance in asymptomatic patients with complex congenital heart disease who have not had cardiac MRI or cardiac CT within the preceding year.
B.Cardiomyopathy
      • Evaluation of patients with suspected arrhythmogenic right ventricular dysplasia; OR
      • To assess LV function in patients with suspected or established cardiomyopathy when all other non-invasive imaging is not feasible or technically suboptimal; OR
      • To assess RV function in patients with suspected RV dysfunction when all other non-invasive imaging is not feasible or technically suboptimal
C.Valvular heart disease
      • Evaluation of suspected dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
      • Evaluation of established dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
D.Evaluation of patients with established coronary artery disease
      • Non-invasive localization of coronary bypass grafts or potential grafts (including internal mammary artery) and/or evaluation of retrosternal anatomy in patients undergoing repeat surgical revascularization
E.Intra-cardiac and para-cardiac masses and tumors:
      • In patients with a suspected cardiac or para-cardiac mass (thrombus, tumor, etc.) suggested by transthoracic echocardiography, transesophageal echocardiography, blood pool imaging or contrast ventriculography who have not undergone cardiac CT or cardiac MRI within the preceding 60 days; OR
      • In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically unstable; OR
      • In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically stable and have not undergone cardiac CT or cardiac MRI within the preceding year; OR
      • In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who have undergone treatment (chemotherapy, radiation therapy, thrombolysis, anticoagulation or surgery) within the preceding year and have not had cardiac CT or cardiac MRI within the preceding 60 days
F.Cardiac aneurysm and pseudoaneurysm
 
G.Evaluation of pericardial conditions (pericardial effusion, constrictive pericarditis, or congenital pericardial diseases):
      • In patients with suspected pericardial constriction; OR
      • In patients with suspected congenital pericardial disease; OR
      • In patients with suspected pericardial effusion who have undergone echocardiography deemed to be technically suboptimal in evaluation of the effusion; OR
      • In patients whose echocardiogram shows a complex pericardial effusion (loculated, containing solid material)
H.Evaluation of cardiac venous anatomy
      • For localization of the pulmonary veins in patients with chronic or paroxysmal atrial fibrillation/flutter who are being considered for ablation; OR
      • Coronary venous localization prior to implantation of a biventricular pacemaker
I.Evaluation of the thoracic aorta
      • In patients with suspected thoracic aortic aneurysm / dilation who have not undergone CT or MRI of the thoracic aorta within the preceding 60 days; OR
      • In patients with confirmed thoracic aortic aneurysm / dilation with new or worsening signs/symptoms; OR
      • For ongoing surveillance of stable patients with confirmed thoracic aortic aneurysm / dilation who have not undergone surgical repair and have not had imaging of the thoracic aorta within the preceding six months; OR
      • In patients with suspected aortic dissection; OR
      • In patients with confirmed aortic dissection who have new or worsening symptoms; OR
      • In patients with confirmed aortic dissection in whom surgical repair is anticipated (to assist in preoperative planning); OR
      • For ongoing surveillance of stable patients with confirmed aortic dissection who have not undergone imaging of the thoracic aorta within the preceding year; OR
      • In patients with confirmed aortic dissection or thoracic aortic aneurysm / dilation who have undergone surgical repair within the preceding year and have not undergone imaging of the thoracic aorta within the preceding six months; OR
      • In patients who have sustained blunt chest trauma, penetrating aortic trauma or iatrogenic trauma as a result of aortic instrumentation; OR
      • In patients being evaluated for potential transcatheter aortic valve implantation/replacement (TAVI or TAVR) provided that the patient has not undergone cardiac CT or cardiac MRI within the preceding 60 days  
J. Left atrial appendage closure device (effective November 06, 2022)
      • For evaluation of cardiac anatomy prior to implantation of a left atrial appendage closure device
      • Following placement of a left atrial appendage closure device, a single study may be performed as an alternative to TEE to assess for intracardiac thrombus
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
For Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed above including screening, does not meet primary coverage criteria.
 
For contracts without primary coverage criteria, for Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed above including screening, is considered investigational. Investigational services are specific contract exclusions.
 
Effective Prior to March 13, 2022
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Coronary CT Angiography and FFR
 
The following indications meet primary coverage criteria for Coronary CT Angiography (CCTA). For some of these indications, FFR-CT may also meet primary coverage criteria.  For some indications, coronary CT meets coverage criteria only if the imaging facility has the capability to perform FFR.
 
1.Coronary CT Angiography only:
· For evaluation of suspected congenital anomalies of the coronary arteries
 
2.Coronary CT Angiography, with FFR when necessary.    
FFR measurement by CT meets primary coverage criteria for the following indications if needed to further assess coronary artery disease of uncertain physiological significance seen on the coronary CT angiogram:
· Congestive Heart Failure/cardiomyopathy/left ventricular dysfunction:
o For exclusion of CAD in patients with left ventricular ejection fraction < 55% and low to moderate coronary heart disease risk (using a standard method of risk assessment, such as the SCORE or ACC/AHA model) in whom CAD has not been excluded as the etiology of the cardiomyopathy.
· For preoperative evaluation of patients undergoing non-coronary cardiac surgery:
o For evaluation of symptomatic* or asymptomatic patients at moderate coronary heart disease risk (using a standard method of risk assessment, such as the SCORE or ACC/AHA model) to avoid an invasive angiogram, where all the necessary preoperative information can be obtained using cardiac CT (Procedures include open and percutaneous valvular procedures or ascending aortic surgery).
· Suspected CAD in patients who have had abnormal exercise EKG test (performed without imaging) within the past 60 days when BOTH of the following criteria are met:
o Patient is symptomatic*
o During testing the patient had exercise-induced chest pain, ST segment change, abnormal BP response or complex ventricular arrhythmias
· Suspected CAD in patients who have and an equivocal myocardial perfusion imaging or stress echo within the past 60 days, when BOTH of the following criteria are met:
o Patient is symptomatic*
o The imaging portion of the study is neither clearly normal nor clearly abnormal.
· Suspected CAD in patients who have had abnormal myocardial perfusion imaging or stress echo within the past 60 days when BOTH of the following criteria are met:
o Patient is symptomatic*
o The imaging portion of the study is abnormal
 
3.Coronary CT Angiography, with FFR when necessary. For these indications, the imaging center MUST have the capability to perform FFR-CT if indicated.
FFR measurement by CT meets primary coverage criteria for the following indications if needed to further assess coronary artery disease of uncertain physiological significance seen on the coronary CT angiogram:    
· Suspected CAD in symptomatic* patients who have an abnormal resting EKG:  
o When resting EKG abnormalities (paced ventricular rhythm, left ventricular hypertrophy with repolarization abnormalities, resting ST depression of 1 mm or more, digoxin effect, or pre-excitation syndrome) would render an exercise treadmill test (without imaging) uninterpretable
· Suspected CAD in symptomatic* patients who have not had recent CAD evaluation:
o When no CAD imaging evaluation has been performed within the preceding 60 days  
Notes:
*For purposes of this policy, a patient is considered to be “symptomatic” when one of the following applies:
· Chest pain
o With intermediate or high pretest probability of CAD; OR
o With low or very low pretest probability of CAD and high risk of CAD (based on a standard method of risk assessment, such as the SCORE or ACC/AHA model)
· Atypical symptoms: shortness of breath (dyspnea), neck, jaw, arm, epigastric or back pain, sweating (diaphoresis), or exercise-induced syncope
o With moderate or high risk of CAD (based on a standard method of risk assessment, such as the SCORE or ACC/AHA model)
· Other symptoms: palpitation, nausea, vomiting, anxiety, weakness, fatigue, or exercise-induced dizziness, lightheadedness or near syncope, etc.
o With high risk of CAD (based on a standard method of risk assessment, such as the SCORE or ACC/AHA model)
· Patients with any cardiac symptom who have diseases/conditions with which CAD commonly coexists, such as:
o Abdominal aortic aneurysm; OR
o Chronic renal insufficiency or renal failure; OR
o Diabetes mellitus; OR
o Established and symptomatic peripheral vascular disease; OR
o Prior history of cerebrovascular accident  
*Performance of multiple non-invasive coronary artery or myocardial perfusion imaging studies on the same patient should be rarely required and will be subject to monitoring.
 
CT Coronary Calcium Scoring
 
Quantitative coronary artery calcium scoring meets primary coverage criteria of effectiveness when:
· Performed to determine if there is too much calcium present to proceed with CT coronary angiography OR
· Performed as part of a pre-operative evaluation for orthotopic liver transplantation.
 
CT for Cardiac Structure
 
The following indications meet primary coverage criteria for CT for Cardiac Structure:
A.Congenital heart disease
· For evaluation of suspected or established congenital heart disease in patients whose echocardiogram is technically limited or non-diagnostic; OR
· For further evaluation of patients whose echocardiogram suggests a new diagnosis of complex congenital heart disease; OR
· For evaluation of complex congenital heart disease in patients who are less than one year post-surgical correction; OR
· For evaluation of complex congenital heart disease in patients who have new or worsening symptoms and/or a change in physical examination; OR
· To assist in surgical planning for patients with complex congenital heart disease; OR
· For surveillance in asymptomatic patients with complex congenital heart disease who have not had cardiac MRI or cardiac CT within the preceding year.
B.Cardiomyopathy
· Evaluation of patients with suspected arrhythmogenic right ventricular dysplasia; OR
· To assess LV function in patients with suspected or established cardiomyopathy when all other non-invasive imaging is not feasible or technically suboptimal; OR
· To assess RV function in patients with suspected RV dysfunction when all other non-invasive imaging is not feasible or technically suboptimal  
C.Valvular heart disease
· Evaluation of suspected dysfunction of native or prosthetic cardiac valves when all other cardiac imaging options are not feasible or technically suboptimal;
D.Evaluation of patients with established coronary artery disease
· Non-invasive localization of coronary bypass grafts or potential grafts (including internal mammary artery) and/or evaluation of retrosternal anatomy in patients undergoing repeat surgical revascularization
E.Intra-cardiac and para-cardiac masses and tumors:
· In patients with a suspected cardiac or para-cardiac mass (thrombus, tumor, etc.) suggested by transthoracic echocardiography, transesophageal echocardiography, blood pool imaging or contrast ventriculography who have not undergone cardiac CT or cardiac MRI within the preceding 60 days; OR
· In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically unstable; OR
· In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who are clinically stable and have not undergone cardiac CT or cardiac MRI within the preceding year; OR
· In patients with established cardiac or para-cardiac mass (thrombus, tumor, etc.) who have undergone treatment (chemotherapy, radiation therapy, thrombolysis, anticoagulation or surgery) within the preceding year and have not had cardiac CT or cardiac MRI within the preceding 60 days
F.Cardiac aneurysm and pseudoaneurysm
 
G.Evaluation of pericardial conditions (pericardial effusion, constrictive pericarditis, or congenital pericardial diseases):
· In patients with suspected pericardial constriction; OR
· In patients with suspected congenital pericardial disease; OR
· In patients with suspected pericardial effusion who have undergone echocardiography deemed to be technically suboptimal in evaluation of the effusion; OR
· In patients whose echocardiogram shows a complex pericardial effusion (loculated, containing solid material)
H.Evaluation of cardiac venous anatomy
· For localization of the pulmonary veins in patients with chronic or paroxysmal atrial fibrillation/flutter who are being considered for ablation; OR
· Coronary venous localization prior to implantation of a biventricular pacemaker
I.Evaluation of the thoracic aorta
· In patients with suspected thoracic aortic aneurysm / dilation who have not undergone CT or MRI of the thoracic aorta within the preceding 60 days; OR
· In patients with confirmed thoracic aortic aneurysm / dilation with new or worsening signs/symptoms; OR
· For ongoing surveillance of stable patients with confirmed thoracic aortic aneurysm / dilation who have not undergone surgical repair and have not had imaging of the thoracic aorta within the preceding six months; OR
· In patients with suspected aortic dissection; OR
· In patients with confirmed aortic dissection who have new or worsening symptoms; OR
· In patients with confirmed aortic dissection in whom surgical repair is anticipated (to assist in preoperative planning); OR
· For ongoing surveillance of stable patients with confirmed aortic dissection who have not undergone imaging of the thoracic aorta within the preceding year; OR
· In patients with confirmed aortic dissection or thoracic aortic aneurysm / dilation who have undergone surgical repair within the preceding year and have not undergone imaging of the thoracic aorta within the preceding six months; OR
· In patients who have sustained blunt chest trauma, penetrating aortic trauma or iatrogenic trauma as a result of aortic instrumentation; OR
· In patients being evaluated for potential transcatheter aortic valve implantation/replacement (TAVI or TAVR) provided that the patient has not undergone cardiac CT or cardiac MRI within the preceding 60 days
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
For Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed above including screening, does not meet primary coverage criteria.
 
For contracts without primary coverage criteria, for Coronary CT Angiography and FFR, coronary artery calcium, and CT for Cardiac Structure, any other indication not listed above including screening, is considered investigational. Investigational services are specific contract exclusions.
 
Effective Prior to February 14, 2021
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Multidetector computed tomography (MDCT) provides advanced spatial and temporal resolution of the heart and allows imaging of the major vessels of the chest, including the coronary arteries. This new technology lacks evidence based indications, but indirect evidence, using diagnostic performance data, decision models, and an expert consensus approach validates the following current indications. Future revisions to these indications will occur as evidence based studies become available.
 
Multidetector computed tomography (MDCT) and coronary computed tomographic angiography (CCTA), using 32 or more detectors and sub millimeter slices can reliably evaluate cardiac structure and morphology, native and anomalous coronary arteries and bypass grafts, congenital heart disease, left and right ventricular function, ejections fractions, and segmental wall motion. The following indications meet primary coverage criteria of effectiveness:
    • To evaluate the coronary arteries in a patient with a low or intermediate probability of obstructive CAD, but who has an equivocal or indeterminate noninvasive diagnostic test (exercise stress test, stress echo, stress SPECT, myocardial perfusion imaging) performed to assess suspected ischemic or obstructive coronary artery disease
    • To evaluate the coronary arteries in a patient with a low or intermediate probability of obstructive CAD, as an alternative to other testing modalities in a patient who would otherwise meet criteria for a stress echo or SPECT stress test.  
    • To evaluate suspected congenital anomalies of the coronary circulation.  
    • To evaluate acute chest pain in the emergency room in order to quickly triage patients and in order to rule out coronary artery disease as a possible cause of symptoms.
    • To evaluate the coronary and or pulmonary venous anatomy, primarily for pre surgical planning, prior to pacemaker placement or pulmonary vein catheter ablation of abnormal electrical activity for treatment of atrial fibrillation.
    • To pre-operatively evaluate cardiac and coronary anatomy prior to non-coronary artery cardiac surgery (e.g., valve or ascending aortic surgery.  
    • For noninvasive coronary arterial mapping, including internal mammary artery prior to repeat cardiac surgical revascularization.  
    • For evaluation of cardiac mass (suspected tumor or thrombus) and for evaluation of pericardial conditions (pericardial mass, constrictive pericarditis, or complications of cardiac surgery)
    • Follow-up of coronary anatomy post cardiac transplantation
    • Evaluation of coronary anatomy in patients with unexplained systolic heart failure (HFrEF) and low risk for obstructive coronary artery disease.
    • Evaluation of graft patency when grafts have not been identifiable by coronary angiography
 
Quantitative coronary artery calcium scoring meets primary coverage criteria of effectiveness when performed to determine if there is too much calcium present to proceed with CT coronary angiography.
 
 *NOTE: Performance of multiple non-invasive coronary artery or myocardial perfusion imaging studies on the same patient should be rarely required, and will be subject to monitoring.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
The following indications do not meet member benefit primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes:
 
Screening:
    • Screening tests (examinations in men or women in the absence of signs, symptoms, or disease) are exclusions in most member benefit certificates of coverage except for coverage based on the Patient Protection and Affordable Care Act (PPACA) screening recommendations for non-grandfathered plans and those contracts with wellness benefits (which like PPACA, covers specific screening procedures). Screening with CT coronary angiography is not recommended by the USPSTF and is not covered in wellness contracts. The ACC/AHA (Taylor, 2010) recommends against screening with CT coronary angiography.
    • Quantitative coronary artery calcium scoring (75571), (unless performed prior to CT coronary angiography to determine if there is too much calcium present to precede with CT coronary angiography.)
 
Low Risk Patients:
    • Patients with non anginal chest pain in whom the history, physical exam, and appropriate diagnostic tests demonstrate non cardiac causes of chest pain.  
    • Patients with low risk of coronary artery disease based on clinical information and any other normal noninvasive coronary anatomic test within the past six months.
 
High Risk Patients who meet the ACC/AHA Guidelines for Coronary Angiography:
 
    • Patients with high pretest likelihood of coronary artery disease (by age, gender, and symptoms) in whom coronary angiography is indicated and or has been scheduled.  
    • Patients with high pretest likelihood of coronary artery disease in whom coronary angiography is indicated based on Class III or Class IV symptoms (CCSC classification of Angina Pectoris). [Class III: marked limitations of ordinary physical activity – angina occurs on walking 1-2 blocks on the level and climbing 1 flight of stairs in normal condition and at a normal pace; class IV: inability to carry on any physical activity without discomfort; anginal symptoms at rest].  
    • Patients with abnormal noninvasive tests which indicate high likelihood or high risk of adverse outcomes from coronary artery disease in whom coronary angiography is recommended.
    • Patients with suspected abrupt closure or subacute stent thrombosis after percutaneous revascularization, and patients with recurrent angina or high risk criteria on noninvasive evaluation within nine months of percutaneous revascularization. In both scenarios, coronary angiography is recommended.
    • Patients who are NOT candidates for revascularization procedures because of concomitant medical issues, severe left ventricular dysfunction, or who refuse revascularization surgery.  
    • To evaluate the cause of chest pain syndrome in patients with prior bypass surgery or intracoronary stent placement.  
    • To detect coronary artery disease in asymptomatic patients who are status post revascularization procedures – (i.e., bypass grafts or intracoronary stents in asymptomatic patients).
    • For cardiac evaluation of a patient where there is a pre-test knowledge of sufficiently extensive calcification of the coronary segment in question that would diminish the interpretive value.
    • Evaluation of coronary artery disease in patients with left bundle branch block.
 
For contracts without primary coverage criteria, the indications above listed as not meeting primary
coverage criteria, are considered investigational. Investigational services are specific contract
exclusions.
 
Due to the detail of the policy statement, the document containing coverage statements for dates prior to August 2019 are not online.  If you would like a hardcopy print, please email: codespecificinquiry@arkbluecross.com

Rationale:
This policy was  developed  based on a literature search conducted on MEDLINE via PubMed through January, 2006.
 
Evaluation of Obstructive Coronary Artery Disease (CAD):
For evaluation of CAD, the reference standard comparison for CTA is with conventional invasive coronary angiography, and CTA is proposed as a noninvasive alternative to invasive angiography. In patients with a relatively low clinical likelihood of coronary artery disease but adequate suspicion to warrant further evaluation, CT coronary artery angiography may provide a high enough negative predictive value to avoid invasive coronary angiography.
 
Summary:
MDCT studies performed on scanners with sub millimeter slice thickness and at least 16 detectors /rotation yield useful diagnostic information about cardiac structure and morphology, function, ejection fraction, and wall motion.  CCTA studies performed on scanners with sub millimeter slice thickness and at least 32 detectors/rotation yield useful diagnostic information about native and anomalous coronary arteries and coronary bypass grafts.  MDCT and CCTA are noninvasive tools which may be used to evaluate patients with suspected coronary artery disease, and may provide a negative predictive value which is sufficient to avoid invasive coronary angiography.  These studies also yield valuable clinical information in patients with suspected congenital cardiac anomalies.
 
2012 Update
This policy is being updated with a search of the MEDLINE database.  There was no new literature identified that would prompt a change in the coverage statement.  There remains a lack of scientific evidence that computed tomographic angiography improves health outcomes when used for screening, for low-risk patients or for high-risk patients who meet ACC/AHA guidelines for angiography.
 
The use for screening a low-risk population was recently evaluated in 1,000 patients undergoing coronary CTA compared to a control group of 1,000 similar patients (McEvoy, 2011). Findings were abnormal in 215 screened patients. Over 18 months’ follow-up, screening was associated with more invasive testing, statin use, but without difference in cardiac event rates.
 
Appropriate use criteria (Taylor, 2010a) (Taylor, 2010b) (Taylor 2010c) and expert consensus documents (Mark, 2010a) (Mark, 2010b) Mark 2010c) have been published jointly by ACCF/ACR/AHA/NASCI/SAIP/SCAI/SCCT, but U.S. guidelines have not been developed. The authors of these publications state that the evidence base for CTA is not yet sufficiently robust to support clinical guideline development. The following are statements from the consensus document:
 
The “…overall sensitivity and specificity on a per-patient basis are both high, and the number of indeterminate studies due to inability to image important coronary segments in the select cohorts represented is less than 5%. In most circumstances, a negative coronary CT angiogram rules out significant obstructive coronary disease with a very high degree of confidence, based on the post-test probabilities obtained in cohorts with a wide range of pretest probabilities. However, post-test probabilities following a positive coronary CT angiogram are more variable, due in part to the tendency to overestimate disease severity, particularly in smaller and more distal coronary segments or in segments with artifacts caused by calcification in the arterial walls. At present, data on the prognostic value of coronary CTA using 64-channel or greater systems remain quite limited. Furthermore, no large-scale studies have yet made a direct comparison of long-term outcomes following conventional diagnostic imaging strategies versus strategies involving coronary CTA.”
 
“In the context of the emergency department evaluation of patients with acute chest discomfort, currently available data suggest that coronary CTA may be useful in the evaluation of patients presenting with an acute coronary syndrome (ACS) who do not have either acute electrocardiogram (ECG) changes or positive cardiac markers. However, existing data are limited, and large multicenter trials comparing CTA with conventional evaluation strategies are needed to help define the role of this technology in this category of patients.”
 
2013 Update
A literature search conducted through October 2013 using the MEDLINE database did not reveal any new information that would prompt a change in the coverage statement. One prospective study and one guideline are summarized as follows:
 
The Study of Myocardial Perfusion and Coronary Anatomy Imaging Roles in Coronary Artery Disease (SPARC), is a prospective multicenter registry study of imaging modalities (Hachamovitch, 2012). From 1703 patients with no history of CAD, angiography was more frequent within 90 days following coronary CTA (13.2%) compared with SPECT (4.3%) or PET (11.1%). Although study results vary and all are observational with the attendant potential for selection bias (in effect confounding by test selection), angiography rates appear higher following coronary CTA.
 
ACCF/AHA/ACP/AATS/PCNA/SCAI/STS joint guidelines for management of patients with stable ischemic heart disease were published in 2012 (Fihn, 1012a; Fihn, 2012b; Fihn, 2012c). Guideline statements for use of coronary CTA were divided whether used in patients without diagnosed disease or those with known disease and a patient’s ability to exercise:
 
Diagnosis Unknown
 
Able To Exercise
Class IIb
“CCTA might be reasonable for patients with an intermediate pretest probability of IHD who have at least moderate physical functioning or no disabling comorbidity.” (Level of Evidence: B)
 
Unable to Exercise
Class IIa
“CCTA is reasonable for patients with a low to intermediate pretest probability of IHD who are incapable of at least moderate physical functioning or have disabling comorbidity.” (Level of Evidence: B)
 
“CCTA is reasonable for patients with an intermediate pretest probability of IHD who a) have continued symptoms with prior normal test findings, or b) have inconclusive results from prior exercise or pharmacological stress testing, or c) are unable to undergo stress with nuclear MPI or echocardiography.” (Level of Evidence: C)
 
For Patients With Known Coronary Disease:
 
Able To Exercise
Class IIb
“CCTA may be reasonable for risk assessment in patients with SIHD (stabile ischemic heart disease) who are able to exercise to an adequate workload but have an uninterpretable ECG.” (Level of Evidence: B)
 
Class III: No Benefit
“Pharmacological stress imaging (nuclear MPI, echocardiography, or CMR) or CCTA is not recommended for risk assessment in patients with SIHD who are able to exercise to an adequate workload and have an interpretable ECG.” (Level of Evidence: C)
 
Unable to Exercise
Class IIa
“Pharmacological stress CMR is reasonable for risk assessment in patients with SIHD who are unable to exercise to an adequate workload regardless of interpretability of ECG.” (Level of Evidence: B)
 
“CCTA can be useful as a first-line test for risk assessment in patients with SIHD who are unable to exercise to an adequate workload regardless of interpretability of ECG.” (Level of Evidence: C)
 
Regardless of Patients’ Ability to Exercise
Class IIb
“CCTA might be considered for risk assessment in patients with SIHD unable to undergo stress imaging or as an alternative to invasive coronary angiography when functional testing indicates a moderate- to high-risk result and knowledge of angiographic coronary anatomy is unknown.” (Level of Evidence: C)
 
Class III: No Benefit
“A request to perform either a) more than 1 stress imaging study or b) a stress imaging study and a CCTA at the same time is not recommended for risk assessment in patients with SIHD.” (Level of Evidence: C)
 
2017 Update
A literature search conducted through December 2016 using the MEDLINE database did not reveal any new information that would prompt a change in the coverage statement.
 
In 2016, the Agency for Healthcare Research and Quality (AHRQ) published a comparative effectiveness review on noninvasive testing for coronary artery disease (CAD) (Skelly et al, 2016).  The review found that:
· After coronary computed tomography angiography (CCTA), clinical outcomes for patients with an intermediate pretest risk
o were similar when compared with usual care or functional testing (low-to-moderate strength of evidence).
o were similar when compared with single-photon emission computed tomography (SPECT) (low strength of evidence).
· After CCTA, referral for invasive coronary angiography (ICA) and revascularization
o was more common than after functional testing (high strength of evidence)
o was similar compared with SPECT and usual care (low strength of evidence).
· After CCTA, additional testing in the emergency department (ED) setting
o was less common compared with usual care (moderate strength of evidence).
o was more common than after SPECT (high strength of evidence)
· After CCTA, hospitalization
o was less common compared to usual care in the ED setting (moderate to low strength of evidence)
o was similar to functional testing in the outpatient setting (moderate strength of evidence).
 
Overall, the AHRQ review found no clear differences between strategies for clinical or management outcomes, although CCTA may lead to a higher frequency of referral for ICA and revascularization.
 
A 2014 RCT (CT-COMPARE) by Hamilton-Craig et al (2014) assessed length of stay and patient costs in 562 patients presenting to the ED with low-to-intermediate risk chest pain who received CCTA or exercise stress testing.17 Costs within 30 days of presentation were significantly lower in the CCTA group (mean, $2193) than in the exercise testing group (mean, $2704; p<0.001). Length of stay was significantly reduced in the CCTA patients compared with the exercise testing patients. Clinical outcomes at 30 days and at 12 months did not differ.
 
Linde et al (2013 & 2015) reported long-term follow-up from the CATCH trial. This trial randomized 600 patients to a CCTA-guided strategy or to standard of care (SOC). For the CCTA-guided strategy, referral for ICA required coronary stenosis greater than 70%. This trial differed in design from the other trials, because patients had been discharged from the ED, and if there was intermediate stenosis (50%-70%) on CCTA, a stress test was used. The referral rate for ICA was 17% for the CCTA strategy versus 12% with SOC (p=NS). At a median 18.7-month follow-up, a major cardiac event was observed in 5 patients in the CCTA-strategy arm compared to 14 in the SOC group (hazard ratio [HR], 0.36; 95% confidence interval [CI], 0.16 to 0.95; p=0.04). Three other follow-up studies reported no cardiac events after a negative CCTA in the ED after 12 (N=481), 24 (N=368), or 47 months (N=506) (Hollander et al, 2009; Schlett et al, 2011; Nasis et al, 2014).
 
2018 Update
Annual policy review completed with a literature search using the MEDLINE database through January 2018. No new literature was identified that would prompt a change in the coverage statement. Three RCTs for patients at risk of CAD comparing net health outcome after a CCTA strategy with outcomes from other noninvasive testing strategies were identified and are summarized below.  
 
 
The PROMISE trial randomized 10,003 patients to CCTA or exercise electrocardiography, nuclear stress testing, or stress echocardiography (as determined by physician preference) as the initial diagnostic evaluation (Douglas, 2015). For the composite end point of death, MI, hospitalization for unstable angina, or major procedural complication, the outcome rates between the 2 groups showed no statistically significant difference (HR=1.04; 95% CI, 0.83 to 1.29). CCTA also did not meet prespecified noninferiority criteria compared with alternative testing. Some clinical outcomes assessed at 12 months favored CCTA, but the differences were nonsignificant. Coronary catheterization rates and revascularization rates were higher in the CCTA group. In further prespecified analysis of PROMISE trial data, Hoffmann et al (2017) found that there was no difference in event rates (death, MI, or angina) between the groups at a median of 26 months follow-up (Hoffmann, 2017). However, CCTA had better discriminatory ability than functional testing to predict events (eg, in categories of normal, mildly abnormal, moderately abnormal, and severely abnormal) in patients who had nonobstructive CAD (p=0.04). When the Framingham Risk Score was added to functional testing results, there was no significant difference in prognostic capability between the approaches (p=0.29).
 
In the SCOT-HEART trial, 4146 patients were randomized to CCTA plus SOC or SOC alone. The primary end point was the change in the proportion of patients with a more certain diagnosis (presence or absence) of angina pectoris (Scot-Heart Investigators, 2015). Secondary outcomes included death, MI, revascularization procedures, and hospitalizations for chest pain. Analysis of the primary outcome showed that patients who underwent CCTA had an increase in the certainty of their diagnosis relative to those in usual care (relative risk, 1.79; 95% CI, 1.62 to 1.96). Regarding health outcomes, the rates of heart disease death and MI were lower with CCTA (1.3% vs 2.0%; HR=0.62; p=0.053), but results were of marginal statistical significance. In 2017, Williams et al reported on symptoms and quality of life for participants in the SCOT-HEART trial (Williams, 2017). Symptoms improved in both groups; however, improvements in symptoms and quality of life at 6 months were lower in patients in the CCTA arm than the functional testing arm. This outcome was due primarily to patients who were diagnosed with moderate CAD or had a new prescription of preventative therapy compared with patients diagnosed with normal coronary arteries or who had their preventative therapy discontinued.
 
 The CAPP trial (2015) randomized 500 patients with stable chest pain to CCTA or exercise stress testing (McKavanagh, 2015). The primary outcome was the change difference in scores of Seattle Angina Questionnaire domains at 3 months. Patients were also followed for further diagnostic tests and management. In the CCTA arm, 15.2% of subjects underwent revascularization. In the exercise stress testing arm, 7.7% underwent revascularization. For the primary outcome, angina stability and quality of life showed significantly greater improvement in the CCTA arm than in the exercise stress testing arm.
 
A number of studies have evaluated the diagnostic accuracy of CCTA for diagnosing CAD in an outpatient population. In general, these studies have reported high sensitivity and specificity, although there is some variability in these parameters across studies. Meta-analyses of these studies have shown that, for detection of anatomic disease, CCTA has a sensitivity greater than 95%, which is superior to all other functional noninvasive tests. Specificity is at least as good as other noninvasive tests. However, the
link between improved diagnosis and health outcomes is not as clear, and thus outcome studies are necessary to demonstrate the clinical utility of CCTA.
 
Direct clinical trial evidence comparing CCTA and other strategies in the diagnostic management of stable patients with suspected CAD has not demonstrated the superiority of CCTA in any of the single clinical trials. Clinical trials have demonstrated greater utilization of ICA and subsequent revascularization procedures after CCTA. An important problem when interpreting the clinical trials is that the comparator strategies differ: in the PROMISE and the CAPP trials, CCTA was compared with an alternative non-invasive test; in other studies, CCTA supplemented usual care (which may or may not have included a noninvasive test). These trial design differences are likely to reflect how CCTA is used in clinical practice¾either as a substitute for another noninvasive test or as an adjunct to other noninvasive tests. The PROMISE trial explicitly compared CCTA with an alternative functional test as the initial diagnostic test. Although the trial did not show the superiority of CCTA and did not meet prespecified criteria for noninferiority, examination of some secondary clinical outcomes supports a conclusion of “at least” noninferiority. The results of the other randomized trials are consistent with the noninferiority of CCTA compared with other established noninvasive tests. Thus, the randomized studies indicate that outcomes of patients are likely to be similar with CCTA vs other noninvasive tests.
 
2019 Update
A literature search was conducted through December 2018.  There was no new information identified that would prompt a change in the coverage statement.  The key identified literature is summarized below.
 
Systematic Reviews
Gongora et al published a meta-analysis of 10 RCTs (total N=6285 patients) comparing CCTA with the standard of care (SOC) in patients with acute chest pain in an ED setting or an inpatient setting (Gongora, 2018). Pooled results suggested that CCTA results in more frequent revascularization and ICA without reducing the risk of adverse cardiac events. Among the limitations of the review were the heterogeneity of SOC across assessed studies, the possibility of publication bias due to the small number of trials available, and the presence of only a few studies that prespecified downstream testing criteria following CCTA results.
 
Levsky et al published an RCT: in the CCTA arm, 39 (19%) patients were hospitalized, compared with 22 (11%) patients of the stress echocardiography arm, resulting in a difference of 8% (95% CI, 1% to 15%; p=0.026) (Levsky, 2018). Median length of stay in the hospital was longer for the CCTA arm (58 hours vs 34 hours; p=0.002, respectively). There was no significant difference between the CCTA and stress echocardiography arms in terms of major adverse cardiac events (MACE; including death): respectively, MACE occurred in 11 CCTA patients and 7 stress echocardiography patients (p=0.47) over a median follow-up of 24 months. The median complete initial work-up radiation exposure for the CCTA arm was 6.4 mSv (interquartile range, 5.3-7.8 mSv), significantly more than that of stress echocardiography (0 mSv; p<0.001). The trial had a number of limitations, including the single-center design and omission of high sensitivity troponin assays.
 
Nonrandomized Studies
Durand et al compared the diagnostic performance of dobutamine-stress echocardiography (DSE) with CCTA in 217 adults (Durand, 2017). Patients had normal measurements of troponin I or T, and electrocardiograph results. All patients received DSE and CCTA, with only 75 (34.6%) patients receiving ICA, which served as the reference test. The primary end point was the diagnostic accuracy of the tests for detecting coronary stenosis greater than 50%. Forty-nine (22.6%) patients had a positive CCTA while 33 (15.2%) patients had a positive DSE. A negative CCTA result was reported in 144 (66.4%) patients, and 146 (67.3%) had a negative DSE result. Overall, CCTA was more sensitive than DSE in detecting CAD, while specificity was similar between tests. At 6 months, no patients had died or received a diagnosis of MI, but 1 patient presented with acute coronary syndrome whose diagnosed was initially missed. No limitations were identified.
 
Case Series
Sandstedt et al published a case series evaluating 1205 patients with suspected CAD who underwent CCTA at a single center (Sandstedt, 2018) Most patients had normal findings (n=668 [55.4%]). Of the 218 patients who underwent ICA, 149 patients had obstructive stenosis, 49 patients had nonobstructive stenosis, and 20 patients did not have evidence of stenosis. The study had several limitations, including a high number of exclusions because of poor image quality, a single-reader clinical evaluation, and limitations inherent of a single-center study.
 
Systematic Reviews
Foy et al conducted a systematic review comparing CCTA with functional stress testing for patients with suspected CAD and stable or acute chest pain (Foy, 2017). In the CCTA arm, there were 10,315 patients, and in the functional stress testing arm, there were 9777 patients; both CCTA and functional stress testing strategies varied among the 13 trials. Overall mortality and cardiac hospitalization did not differ between CCTA and functional stress testing groups. There were fewer cases of MI in the CCTA group than in the functional stress testing group; however, the incidence of ICA and revascularization were higher in the CCTA group. CCTA was associated with an increase in new diagnoses of CAD as well as increased prescription of aspirin and statin therapy. All trials reported a lack of blinding, both of patients and personnel, and overall quality of evidence was moderate, despite a high risk of bias in several studies included. Additional limitations included the lack of available patient-level data, the absence of assessment of time to hospital discharge, and differences in radiation exposure.
 
January 2020 Update
A literature search was conducted through December 2019.  There was no new information identified that would prompt a change in the coverage statement.  
 
November 2020 Update
A literature search was conducted through October 2019. Following is a summary of the new literature to date.
 
Coronary CT Angiography and FFR
Nørgaard et al (2017) reported on results from symptomatic patients referred for CCTA at a single-center in Denmark from May 2014 to April 2015. All data were obtained from medical records and registries; the study was described as a "review" of diagnostic evaluations and was apparently retrospectively conducted. Follow-up through 6 to 18 months was ascertained. From 1248 referred patients, 1173 underwent CCTA; 858 received medical therapy, 82 underwent ICA, 44 MPI, and 189 FFR-CT (185 [98%] obtained successfully). Of the 185 individuals who successfully obtained FFR-CT, FFR-CT demonstrated values of 0.80 or less in 1 or more vessels in 57 (31%) patients, and 49 (86%) went on to ICA; whereas of the 128 with higher FFR-CT values, only 5 (4%) went on to ICA. Assuming ICA was planned for all patients undergoing FFR-CT, these results are consistent with FFR-CT being able to decrease the rate of ICA. However, implications are limited by the retrospective design, performance at a singlecenter, and lack of a comparator arm including one for CCTA alone. Lu et al (2017) Retrospective Cohort
 
Lu et al (2017) retrospectively examined a subgroup referred to ICA48, from the completed PROspective Multicenter Imaging Study for Evaluation of Chest Pain (PROMISE) trial. PROMISE was a pragmatic trial comparing CCTA with functional testing for the initial evaluation of patients with suspected SIHD. Of 550 participants referred to ICA within 90 days, 279 were not considered for the analyses due to CCTA performed without nitroglycerin (n=139), CCTA not meeting slice thickness guidelines (n=90), or nondiagnostic studies (n=50). Of the remaining 271 patients, 90 scans were inadequate to obtain FFR-CT, leaving 181 (33%) of those referred to ICA for analysis. Compared with those excluded, patients in the analytic sample were less often obese, hypertensive, diabetic, minority, or reported a CAD equivalent symptom. The two 2 groups had similar pretest probabilities of disease, revascularization rates, and MACE, but the distribution of stenoses in the analytic sample tended to be milder (p=0.06). FFR-CT studies were performed in a blinded manner and not available during the conduct of PROMISE for decision making.
 
Severe stenosis (> or = 70%) or left main disease (> or = 50%) were present in 110 (66%) patients by CCTA result and in 54% by ICA. Over a 29-month median follow-up, MACE (death, nonfatal MI, hospitalization for unstable angina) or revascularization occurred in 51% of patients (9% MACE, 49% revascularization). A majority (72%) of the sample had at least 1 vessel with an FFR-CT of 0.80 or less, which was also associated with a higher risk of revascularization but with a wide CI (hazard ratio=5.1; 95% CI, 2.6 to 11.5). If reserved for patients with an FFR-CT of 0.80 or less, ICAs might have been avoided in 50 patients (ie, reduced by 28%) and the rate of ICA without 50% or more stenosis from 27% (calculated 95% CI, 21% to 34%) to 15% (calculated 95% CI, 10% to 23%). If the 90 patients whose images were sent for FFR-CT but were unsatisfactory proceeded to ICA—as would have occurred in practice—the rate of ICA might have decreased by 18% and ICA without significant stenosis from 31% to 25%.
 
The authors suggested that when CCTA is used as the initial evaluation for patients with suspected SIHD, adding FFR-CT could have decreased the referral rate to ICA in PROMISE from 12.2% to 9.5%, or close to the 8.1% rate observed in the PROMISE functional testing arm. They also noted the similarity of their findings to PLATFORM and concluded, "In this hypothesis-generating study of patients with stable chest pain referred to ICA after [C]CTA, we found that adding FFRCT may improve the efficiency of referral to ICA, addressing a major concern of an anatomic [C]CTA strategy. FFRCT has incremental value over anatomic [C]CTA in predicting revascularization or major adverse cardiovascular events."
 
This retrospective observational subgroup analysis from PROMISE would suggest that when CCTA is the initial noninvasive test for the evaluation of suspected SIHD, FFRCT before ICA has the potential to reduce unnecessary ICAs and increase the diagnostic yield. However, study limitations and potential generalizability are important to consider. First, analyses included only a third of CCTA patients referred to ICA, and some characteristics of the excluded group differed from the analytic sample. Second, conclusions assume that an FFR-CT greater than 0.80 will always dissuade a physician from recommending ICA and even in the presence of severe stenosis (eg, 70% in any vessel or 50% in the left main), or almost half (46%) of patients with an FFR-CT greater than 0.80. Finally, estimates including patients with either nondiagnostic CCTA studies (n=50) or studies inadequate for calculating FFR-CT (n=90) are more appropriate because in practice most likely those patients would most likely proceed in practice to ICA. Accordingly, the estimates are appropriately considered upper bounds for what might be seen in practice. It is also important to note that in strata of the PLATFORM trial enrolling patients for initial noninvasive testing (not planned ICA), ICA was more common following CCTA and contingent FFR-CT than following usual care (18.3% vs. 12.0%) and ICA, with no obstructive disease more frequent in the FFR-CT arm (12.5% vs. 6.0%).
 
Newby et al on behalf of the SCOT-HEART Investigators (2018) published the results of an open-label, multicenter, parallel-group trial (NCT01149590) that studied and reported 5-year clinical outcomes from patients with stable chest pain who used CTA in the diagnosis and assessment of their condition.  In this trial, 4146 patients with stable chest pain who had been referred to a cardiology clinic for evaluation were randomly assigned to standard care plus CTA (2073 patients) or to standard care alone (2073 patients). Investigations, treatments, and clinical outcomes were assessed over 3 to 7 years of follow-up. The primary end point was death from coronary heart disease or nonfatal myocardial infarction at 5 years. The 5-year rate of the primary end point was lower in the CTA group than in the standard-care group (2.3% [48 patients] vs. 3.9% [81 patients]; hazard ratio, 0.59; 95% confidence interval [CI], 0.41 to 0.84; P=0.004). Although the rates of invasive coronary angiography and coronary revascularization were higher in the CTA group than in the standard-care group in the first few months of follow-up, overall rates were similar at 5 years. more preventive therapies were initiated in patients in the CTA group. There were no significant differences in death rate between the two groups (cardiovascular, non-cardiovascular, or deaths from any cause). The researchers concluded that the use of CTA in addition to standard-care versus standard-care alone resulted in a significantly lower rate of death from coronary heart disease or nonfatal myocardial infarction at 5 years in patients with stable chest pain. Also, the addition of CTA did not increase invasive interventions (coronary angiography or coronary revascularization) over the 5-year follow up.
 
CT Coronary Calcium Scoring
In 2017, Ferencik et al evaluated whether the distribution of CAC in individual coronary arteries and segments, as well as CAC in the proximal dominant coronary artery, as detected by cardiac CT predicts incident major CHD events independent of traditional CAC score in 1268 asymptomatic subjects without prevalent major CHD from the offspring and third generation cohorts of the Framingham Heart Study (Ferencik, 2017). Results revealed a total of 42 major CHD events occurring during a median follow-up period of 7.4 years. Both the number of coronary arteries with CAC (hazard ratio [HR], 1.68 per artery, 95% CI, 1.10 to 2.57; p=0.02) and the presence of CAC in the proximal dominant coronary artery (HR, 2.59; 95% CI, 1.15 to 5.83; p=0.02) were associated with major CHD events after multivariable adjustment.
 
Erbel et al (2010) assessed NRI and risk prediction based on CAC scoring in comparison with traditional risk factors in 4129 subjects without overt CAD at baseline in the Heinz Nixdorf Recall study (Erbel, 2010). Results revealed that 93 coronary deaths and nonfatal MIs occurred after 5 years of follow-up (cumulative risk 2.3%; 95% CI, 1.8% to 2.8%). Reclassifying intermediate risk subjects with CAC <100 to the low risk category and with CAC 400 to the high-risk category yielded a NRI of 21.7% (p=0.0002) and 30.6% (p<0.0001) for the FRS, respectively. Adding CAC scores to the FRS and National Cholesterol Education Panel ATP III categories improved the AUC from 0.681 to 0.749 (p<0.003) and from 0.653 to 0.755 (p=0.001), respectively. The authors concluded that limiting CAC scoring to intermediate risk subjects assists in correctly identifying a high proportion of individuals at highest risk and may contribute to reducing the number of coronary events in the general population; however, clinicians need to be aware that this may not be applicable across the board, particularly for patients in a low risk category. In 2018, Lehmann et al published additional 10-year follow-up data from Heinz Nixdorf and concluded that CAC progression is associated with coronary and CV event rates, but only weakly adds to risk prediction (Lehmann, 2018). The authors stated that what counts is the most recent CAC value and risk factor assessment.
 
Gupta et al performed a systematic review and meta-analysis evaluating the odds of initiating or continuing pharmacological (ie, aspirin, lipid-lowering, and blood pressure lowering medications) and lifestyle preventive therapies in asymptomatic CAD patients with nonzero versus 0 CAC scores as detected on cardiac CT (Gupta, 2017). Results revealed that the odds of aspirin, lipid-lowering, and blood pressure lowering medication initiation, lipid-lowering medication continuation, an increase in exercise, and dietary changes were significantly higher in patients with nonzero CAC versus 0 CAC scores. However, the odds of aspirin or blood pressure-lowering medication continuation were not significantly increased in the nonzero CAC group. Statistical heterogeneity was present across studies for many of the outcomes; potential sources of heterogeneity included variations in sample size and the proportion of patients with 0 versus nonzero CAC, whether patients were shown their CAC scan, and differences in clinical characteristics of study populations.
 
The American College of Cardiology and American Heart Association (2019) Guideline on the Primary Prevention of Cardiovascular Disease is in line with the blood cholesterol guideline stating that adults (40 to 75 years of age) who are being evaluated for cardiovascular disease prevention should initially undergo 10-year atherosclerotic cardiovascular disease (ASCVD) risk estimation with a clinician-patient risk discussion before starting pharmacological therapy (Arnett, 2019). The guideline also notes that assessing for other risk-enhancing factors can help guide decision making "about preventive interventions in select individuals, as can CAC scanning." The guideline specifically states the following recommendation regarding assessment of cardiovascular risk and CAC:
 
In adults at intermediate risk (7.5% to < 20% 10-year ASCVD risk) or selected adults at borderline risk (5% to <7.5% 10-year ASCVD risk), if risk-based decisions for preventive interventions remain uncertain, it is reasonable to measure a CAC score to guide clinician-patient risk discussion [Class (Strength) of Recommendation: IIa; Level (Quality) of Evidence: B-NR]. A IIa class of recommendation is of moderate strength based on moderate quality nonrandomized studies.
 
Kong et al (2015) published a study that evaluated the utility of preoperative coronary calcium
scores for predicting early postoperative cardiovascular complications in liver transplant recipients. This was an observational study that retrospectively analyzed the outcomes of 443 liver transplant recipients between 2010 and 2012.  Preoperative CV assessments, including coronary CT, were performed. A positive finding was defined as a coronary calcium score of > 400. Other predictive factors of early postoperative cardiovascular complications were also evaluated. Major cardiovascular complications occurring during a period of 1 month after transplant were noted. Of the 443 patients, 38 (8.6%) experienced one or more cardiovascular complications. Positive coronary CT findings were seen in 11 (2.5%) patients. The authors concluded that A preoperative coronary calcium score of >400 predicted cardiovascular complications occurring 1 month after LT, suggesting that preoperative evaluation of coronary calcium scores could help predict early postoperative cardiovascular complications in liver transplant recipients. Kong et al (2015b) also published a retrospective observational study that evaluated the incidence and cardiovascular risk factors of a coronary calcium score >400 in 548 liver transplant recipients between 2013-2014 that found similar conclusions.
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2021. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Cainzos-Achirica et al assessed whether use of CAC improved appropriate aspirin use for primary prevention compared with other risk calculators (Cainzos-Achirica, 2020). In multivariable regression analysis, a CAC score 100 was independently associated with an increased risk of CVD events compared with those with a CAC score of 0 (hazard ratio [HR], 3.9; 95% CI, 2.5 to 6.1]. The pooled cohort equations and an estimated cardiovascular risk threshold of >20% failed to identify optimal candidates for aspirin; however, a CAC score of at least 100 was able to identify subgroups of patients where aspirin would yield benefit.
 
2021 Update
A literature search was conducted through September 2021. Following is a summary of the new literature to date.
 
In 2020, the ACC/AHA published a Guideline for the Management of Patients With Valvular Heart Disease with recommendations for the diagnosis and management of valvular heart disease (Otto et al, 2021). ACC/AHA made the following recommendations related to evaluation of a patient with valvular heart disease:
    • Disease stages in patients with valvular heart disease should be classified (Stages A, B, C, and D) on the basis of symptoms, valve anatomy, the severity of valve dysfunction, and the response of the ventricle and pulmonary circulation.
    • In the evaluation of a patient with valvular heart disease, history and physical examination findings should be correlated with the results of noninvasive testing (ie, ECG, chest x-ray, transthoracic echocardiogram). If there is discordance between the physical examination and initial noninvasive testing, consider further noninvasive (computed tomography, cardiac magnetic resonance imaging, stress testing) or invasive (transesophageal echocardiography, cardiac catheterization) testing to determine optimal treatment strategy.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through November 2022. No new literature was identified that would prompt a change in the coverage statement.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through November  2023. No new literature was identified that would prompt a change in the coverage statement.
 
CCTA for CAD
In 2022, SCCT published an expert consensus document on use of CCTA for patients presenting to the emergency department with acute chest pain (Maroules et al, 2022). Relevant recommendations from the consensus document are listed below.
    • Scenario: Patient with no known CAD
        • Recommendation: None
    • Scenario: ECG diagnostic for STEMI
        • Recommendation: CCTA is usually not appropriate (door-to-balloon time <90 minutes should be prioritized).
    • Scenario: NSTE-ACS is leading diagnosis (evidence of myocardial ischemia on ECG without ST-segment elevation, elevated troponin)
        • Recommendation: CCTA may be appropriate (eg, to determine if invasive evaluation is appropriate).
    • Scenario: High risk for ACS (no definite evidence of myocardial ischemia on ECG, normal or equivocal troponin)
        • Recommendation: CCTA may be appropriate as an alternative to functional testing or invasive evaluation.  
    • Scenario: Low to intermediate risk for ACS (no definite evidence of myocardial ischemia on ECG, normal or equivocal troponin, and/or inadequate or mildly abnormal functional testing during index ED visit or within previous year)
        • Recommendation: CCTA is appropriate and is most effective to rule out ACS.
    • Scenario: Very low risk for ACS (no definite evidence of myocardial ischemia on ECG, normal or equivocal troponin, and/or non-cardiac chest pain is leading diagnosis)
        • Recommendation: CCTA may be appropriate (eg, to confidently exclude CAD and provide risk stratification).
    • Scenario: Patient with documented CAD, post-revascularization
        • Recommendation: None
    • Scenario: Prior PCI with stent >3 mm within a proximal coronary segment (no definite evidence of myocardial ischemia on ECG, normal or equivocal troponin)
        • Recommendation: CCTA is appropriate for early triage.
    • Scenario: Prior CABG (no definite evidence of myocardial ischemia on ECG, normal or equivocal troponin)
        • Recommendation: CCTA is appropriate, particularly for evaluating graft patency.
 
CCTA with Selective Noninvasive FFR
An et al (2023) conducted a meta-analysis of machine learning-based methods of determining fractional flow reserve compared to invasive methods. A total of 13 studies in patients with suspected or confirmed CAD were combined for the analysis. Characteristics of the studies were not provided, including the potential for bias, but the authors stated that none of the studies were "large sample size diagnostic performance studies". Machine learning fractional flow reserve had a lower sensitivity and higher specificity than invasive fractional flow determination (0.80 vs. 0.87; p<.01 and 0.86 vs. 0.35; p<.01, respectively). Heterogeneity for all assessments was high (I2, 57.12% to94.52%) and the authors noted that machine learning methods differed among studies.
 
Wang et al (2019) conducted a single-center prospective cohort study of the diagnostic accuracy of the Deep Vessel FFR platform. In 63patients who underwent CCTA, the deep learning software was compared to wire-based (invasive) FFR. Deep Vessel FFR had a higher diagnostic performance as assessed by area under the receiver-operation characteristics curve (0.928) compared to wire-based FFR (0.664).Deep Vessel FFR had a sensitivity, specificity, positive predictive value, and negative predictive value of 97.14%, 75%, 82.93%, and 95.45%,respectively.
 
Qiao et al (2022) conducted a prospective, single-center, nonrandomized cohort study in patients with suspected CAD. Patients received either CCTA alone (n=567) or fractional flow reserve measurement using CCTA (n=566). The primary outcome of interest, ICA that showed nonobstructive disease at 90 days, occurred in 33.3% of the CCTA alone group and 19.8% of the fractional flow reserve group (risk difference, 13.5%; 95% CI, 8.4% to 18.6%; p=.03). ICA was utilized more frequently in the CCTA alone group than the fractional flow reserve group (27.5% vs. 20.3%; p=.003). At 1 year, MACE was more common in the CCTA alone group compared to the fractional flow reserve group (6.7% vs. 3.9%; hazard ratio [HR], 1.73; 95% CI, 1.01 to 2.95; p=.04).
 

CPT/HCPCS:
0501TNoninvasive estimated coronary fractional flow reserve (FFR) derived from coronary computed tomography angiography data using computation fluid dynamics physiologic simulation software analysis of functional data to assess the severity of coronary artery disease; data preparation and transmission, analysis of fluid dynamics and simulated maximal coronary hyperemia, generation of estimated FFR model, with anatomical data review in comparison with estimated FFR model to reconcile discordant data, interpretation and report
0502TNoninvasive estimated coronary fractional flow reserve (FFR) derived from coronary computed tomography angiography data using computation fluid dynamics physiologic simulation software analysis of functional data to assess the severity of coronary artery disease; data preparation and transmission
0503TNoninvasive estimated coronary fractional flow reserve (FFR) derived from coronary computed tomography angiography data using computation fluid dynamics physiologic simulation software analysis of functional data to assess the severity of coronary artery disease; analysis of fluid dynamics and simulated maximal coronary hyperemia, and generation of estimated FFR model
0504TNoninvasive estimated coronary fractional flow reserve (FFR) derived from coronary computed tomography angiography data using computation fluid dynamics physiologic simulation software analysis of functional data to assess the severity of coronary artery disease; anatomical data review in comparison with estimated FFR model to reconcile discordant data, interpretation and report
75571Computed tomography, heart, without contrast material, with quantitative evaluation of coronary calcium
75572Computed tomography, heart, with contrast material, for evaluation of cardiac structure and morphology (including 3D image postprocessing, assessment of cardiac function, and evaluation of venous structures, if performed)
75573Computed tomography, heart, with contrast material, for evaluation of cardiac structure and morphology in the setting of congenital heart disease (including 3D image postprocessing, assessment of LV cardiac function, RV structure and function and evaluation of vascular structures, if performed)
75574Computed tomographic angiography, heart, coronary arteries and bypass grafts (when present), with contrast material, including 3D image postprocessing (including evaluation of cardiac structure and morphology, assessment of cardiac function, and evaluation of venous structures, if performed)
75580Noninvasive estimate of coronary fractional flow reserve (FFR) derived from augmentative software analysis of the data set from a coronary computed tomography angiography, with interpretation and report by a physician or other qualified health care professional
S8092Electron beam computed tomography (also known as ultrafast ct, cine ct)

References: Achenbach S, Moshage W, Ropers D, et al.(1997) Noninvasive, three-dimensional visualization of coronary artery bypass grafts by electron beam tomography. Am J Cardiol 1997; 79:856-61.

Arnett DK, Blumenthal RS, Albert MA, et al.(2019) 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. Sep 10 2019; 140(11): e596-e646. PMID 30879355

Barriales-Villa R, Moris C.(2001) Usefulness of helical computed tomography in the identification of the initial course of coronary anomalies. Am J Cardiol 2001; 88:719.

Bateman TM, Gray RJ, Whiting JS, et al.(1987) Prospective evaluation of ultrafast cardiac computed tomography for determination of coronary bypass graft patency. Circulation 1987; 75:1018-24.

Blaha MJ, Cainzos-Achirica M, Greenland P, et al.(2016) Role of coronary artery calcium score of zero and other negative risk markers for cardiovascular disease: the Multi-Ethnic Study of Atherosclerosis (MESA). Circulation. Mar 1 2016;133(9):849-858. PMID 26801055

Bonow, RO.(2009) Should coronary calcium screening be used in cardiovascular prevention strategies? N Engl J Med 2009;361:990-997.

Braunwald E, Antman EM, Beasley JW, et al.(2002) Guideline Update for the Management of Patients With Unstable Angina and Non-ST-Segment Elevation Myocardial Infarction. ACC/AHA 2002; 1-93.

Brindis RG, Douglas PS, Hendel RC, et al.(2005) Appropriateness Criteria for Single-Photon Emission Computed Tomography Myocardial Perfusion Imaging (SPECT MPI). JACC 2005; 46:81587-605.

Budoff MJ, Achenbach S, Blumenthal RS et al.(2011) Assessment of coronary artery disease by cardiac computed tomography: a scientific statement from the American Heart Association Committee on Cardiovascular Imaging and Intervention, Council on Cardiovascular Radiology and Intervention, and Committee on Cardiac Imaging, Council on Clinical Cardiology. Circulation 2006; 114(16):1761-91. Available online at http //circ.ahajournals.org/cgi/content/full/114/16/61. Last accessed July 2011.

Budoff MJ, Achenbach S, Duerinckx A.(2003) Clinical utility of computed tomography and magnetic resonance techniques for noninvasive coronary angiography. J Am Coll Cardiol 2003; 42:1867-78.

Budoff MJ, Cohen MC, et al.(2005) ACCF/AHA Clinical Competence Statement on cardiac imaging with computed tomography and magnetic resonance. J Am Coll Cardiol 2005; 46:383-402.

Budoff MJ, Mohlenkamp S, McClelland R et al.(2013) A comparison of outcomes with coronary artery calcium scanning in unselected populations: the Multi-Ethnic Study of Atherosclerosis (MESA) and Heinz Nixdorf RECALL study (HNR). J Cardiovasc Comput Tomogr 2013; 7(3):182-91.

Budoff MJ, Young R, Burke G, et al.(2018) Ten-year association of coronary artery calcium with atherosclerotic cardiovascular disease (ASCVD) events: the multi-ethnic study of atherosclerosis (MESA). Eur. Heart J., 2018 Apr 25;39(25). PMID 29688297.

Cainzos-Achirica M, Miedema MD, McEvoy JW, et al.(2020) Coronary Artery Calcium for Personalized Allocation of Aspirin in Primary Prevention of Cardiovascular Disease in 2019: The MESA Study (Multi-Ethnic Study of Atherosclerosis). Circulation. May 12 2020; 141(19): 1541-1553. PMID 32233663

Caussin C, Ohanessian A, et all.(2003) Coronary plaque burden detected by multislice computed tomography after acute myocardial infarction with near-normal coronary arteries by angiography. Am J Cardiol 2003; 92:849-52.

Chaikriangkrai K, Palamaner Subash Shantha G, Jhun HY, et al.(2016) Prognostic value of coronary artery calcium score in acute chest pain patients without known coronary artery disease: systematic review and meta-analysis. Ann Emerg Med. 2016;68(6):659-670.

Chaikriangkrai K, Velankar P, Schutt R, et al.(2015) Additive prognostic value of coronary artery calcium score over coronary computed tomographic angiography stenosis assessment in symptomatic patients without known coronary artery disease. Am J Cardiol. Mar 15 2015;115(6):738-744. PMID 25604930

Chandrashekhar Y, for the JACC: Cardiovascular Imaging Editors(2019) What Is of Recent Interest in Cardiac CT J Am Coll Cardiol 2019 Jul, 73 (25) 3352-3355

Chang SM, Nabi F, Xu J, et al.(2015) Value of CACS compared with ETT and myocardial perfusion imaging for predicting long-term cardiac outcome in asymptomatic and symptomatic patients at low risk for coronary disease: clinical implications in a multimodality imaging world. JACC Cardiovasc Imaging. Feb 2015;8(2):134-144.

Choi AD, Leifer ES, Yu J, et al.(2016) Prospective evaluation of the influence of iterative reconstruction on the reproducibility of coronary calcium quantification in reduced radiation dose 320 detector row CT. Journal of cardiovascular computed tomography. 2016;10(5):359-363.

Davies JE, Cook CM(2017) Is FFRCT Ready to Assume the Crown Jewels of Invasive FFR? J Am Coll Cardiol Img 2017 Apr, 10 (4) 434-436

deFeyter PJ, Nieman K.(2004) Noninvasive multi-slice computed tomography coronary angiography (editorial). J Am Coll Cardiol 2004; 44:1238-40.

Dewey M, Teige F, et al.(2006) Noninvasive detection of coronary artery stenoses with multislice computed tomography or magnetic resonance imaging. Ann Int Med, 2006; 145:407-15.

Dharampal AS, Rossi A, Dedic A et al.(2013) Restriction of the referral of patients with stable angina for CT coronary angiography by clinical evaluation and calcium score: impact on clinical decision making. Eur Radiol 2013; 23(10):2676-86.

Douglas PS, Hoffmann U, Patel MR, et al.(2015) Outcomes of anatomical versus functional testing for coronary artery disease. N Engl J Med. Apr 2 2015;372(14):1291-1300. PMID 25773919

Durand E, Bauer F, Mansencal N, et al.(2017) Head-to-head comparison of the diagnostic performance of coronary computed tomography angiography and dobutamine-stress echocardiography in the evaluation of acute chest pain with normal ECG findings and negative troponin tests: A prospective multicenter study. Int J Cardiol. Aug 15 2017;241:463-469. PMID 28325613

Erbel R, Mohlenkamp S, Moebus S, et al.(2010) Coronary risk stratification, discrimination, and reclassification improvement based on quantification of subclinical coronary atherosclerosis: the Heinz Nixdorf Recall study. J Am Coll Cardiol. Oct 19 2010; 56(17): 1397-406. PMID 20946997

Excellence TNIfHaC.(2016) Chest pain of recent onset: assessment and diagnosis. 2016 https://www.nice.org.uk/guidance/cg95/chapter/Recommendations. Accessed 09/19/2017, 2017.

Ferencik M, Pencina KM, Liu T, et al.(2017) Coronary Artery Calcium Distribution Is an Independent Predictor of Incident Major Coronary Heart Disease Events: Results From the Framingham Heart Study. Circ Cardiovasc Imaging. Oct 2017; 10(10). PMID 28956774

Ferket BS, Genders TS, Colkesen EB et al.(2011) Systematic review of guidelines on imaging of asymptomatic coronary artery disease. J Am Coll Cardiol 2011; 57(15):1591-600.

Fihn S, Blankenship J, Alexander K, et al.(2014) ACC/AHA/AATS/PCNA/PCNA/SCAI/STS focused update of the guideline for the diagnosis and management of patients with stable ischemic heart disease. J Am Coll Cardiol. 2014;64(18):1929-1949.

Fihn SD, Gardin JM, Abrams J et al.(2012) 2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS Guideline for the diagnosis and management of patients with stable ischemic heart disease: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines, and the American College of Physicians, American Association for Thoracic Surgery, Preventive Cardiovascular Nurses Association, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons. J Am Coll Cardiol 2012

Foy AJ, Dhruva SS, Peterson B, et al.(2017) Coronary computed tomography angiography vs functional stress testing for patients with suspected coronary artery disease: a systematic review and meta-analysis. JAMA Intern Med. Nov 1 2017;177(11):1623-1631. PMID 28973101

Gepner AD, Young R, Delaney JA, et al.(2017) Comparison of carotid plaque score and coronary artery calcium score for predicting cardiovascular disease events: the multi-ethnic study of atherosclerosis. J Am Heart Assoc. 2017;6(2).

Gerber BL, Coche E, et al.(2005) Coronary artery stenosis: direct comparison of four-section multi-detectro row CT abd 3D navigator MR imaging for detection--initial results. Radiology 2005; 234:98-108.

Ghostine S, Caussin C, et al.(2006) Non-invasive detection of coronary artery disease in patients with left bundle branch block using 64-slice computed tomography. J Am Coll Cardiol, 2006; 48:1929-34.

Gibbons RJ, Abrams J, Chatterjee K, et al.(2003) Management of Patients with Chronic Stable Angina. J am Coll Cardiol 2003; 41:159-68.

Gibbons RJ, Balady GJ, Bricker JT, et al.(2002) Guideline Update for Exercise Testing. ACC/AHA Practice Guidelines 2002; 1-57.

Gibbons RJ, Bricker JT, Fletcher GF, et al.(2002) Guideline Update for Exercise Testing: Summary Article. Circulation 2002; 1883-1892.

Gilard M, Cornily JC, et al.(2005) Noninvasive assessment of left main coronary stent patency with 16-slice computed tomography. Am J Cardiol 2005; 95:110-2.

Gongora CA, Bavishi C, Uretsky S, et al.(2018) Acute chest pain evaluation using coronary computed tomography angiography compared with standard of care: a meta-analysis of randomised clinical trials. Heart. Feb 2018;104(3):215-221. PMID 28855273

Gottlieb I, Miller JM, Arbab-Zadeh A, et al.(2010) The absence of coronary calcification does not exclude obstructive coronary artery disease or the need for revascularization in patients referred for conventional coronary angiography. J Am Coll Cardiol. 2010;55(7):627-634.

Greenland P, Alpert JS, Beller GA et al.(2010) 2010 ACCF/AHA guideline for assessment of cardiovascular risk in asymptomatic adults: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2010; 56(25):e50-103.

Greenland P.(2006) Who is a candidate for noninvasive coronary angiography. Ann Int Med, 2006; 145:466-7.

Grundy SM, Stone NJ, Bailey AL, et al.(2018) 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation, 2018 Dec 28;139(25). PMID 30586774

Gupta A, Lau E, Varshney R, et al.(2017) The Identification of Calcified Coronary Plaque Is Associated With Initiation and Continuation of Pharmacological and Lifestyle Preventive Therapies: A Systematic Review and Meta-Analysis. JACC Cardiovasc Imaging. Aug 2017; 10(8): 833-842. PMID 28797402

Haberl R, Tittus J, et al.(2005) Multislice spiral computed tomographic angiography of coronary arteries in patients with suspected coronary artery disease: an effective filter before catheter angiography. Am Heart J 2005; 149:1112-9.

Hachamovitch R, Nutter B, Hlatky MA et al.(2012) Patient management after noninvasive cardiac imaging results from SPARC (Study of myocardial perfusion and coronary anatomy imaging roles in coronary artery disease). J Am Coll Cardiol 2012; 59(5):462-74.

Hague C, Andrews G, Forster B.(2004) MDCT of a malignant anomalous right coronary artery. Am J Roentgenol 2004; 182(3):617-8.

Hamilton-Craig C, Fifoot A, Hansen M, et al.(2014) Diagnostic performance and cost of CT angiography versus stress ECG--a randomized prospective study of suspected acute coronary syndrome chest pain in the emergency department (CT-COMPARE). Int J Cardiol. Dec 20 2014;177(3):867-873. PMID 25466568.

Han D, Hartaigh BO, Gransar H, et al.(2018) Incremental prognostic value of coronary computed tomography angiography over coronary calcium scoring for major adverse cardiac events in elderly asymptomatic individuals. Eur Heart J Cardiovasc Imaging. Jun 1 2018;19(6):675-683. PMID 28977374

Hecht HS.(2005) Computed tomographic angiography coming of age. Am J Cardiol 2005; 95:838-42.

Helfand M, Buckley D, Freeman M et al.(2009) Emerging risk factors for coronary heart disease: a summary of systematic reviews conducted for the U.S. Preventive Services Task Force. Ann Intern Med. 2009 Oct 6;151(7):496-507.

Hendel RC, Kramer CM, et al.(2006) ACCF/ACR/SCCT/SCMR/ASNC/NASCI/SCAI/SIR appropriateness criteria for cardiac computed tomography and cardiac magnetic resonance imaging. J Am Coll Cardiol 2006; 48:epub.

Hoffmann U, Ferencik M, Udelson JE, et al.(2017) Prognostic Value of Noninvasive Cardiovascular Testing in Patients With Stable Chest Pain: Insights From the PROMISE Trial (Prospective Multicenter Imaging Study for Evaluation of Chest Pain). Circulation. Jun 13 2017;135(24):2320-2332. PMID 28389572

Hollander JE, Chang AM, Shofer FS, et al.(2009) One-year outcomes following coronary computerized tomographic angiography for evaluation of emergency department patients with potential acute coronary syndrome. Acad Emerg Med. Aug 2009;16(8):693-698. PMID 19594460.

Ioannidis PP, Tzoulaki I.(2010) What makes a good predictor? JAMA. 2010 Apr 28;303(16):1646-1647.

Iskandrian AE.(2006) Detecting coronary artery disease in left bundle branch block. J Am Coll Cardiol, 2006; 48:1935-7.

Jacobs PC, Gondrie MJ, van der Graaf Y, et al.(2012) Coronary artery calcium can predict all-cause mortality and cardiovascular events on low-dose CT screening for lung cancer. AJR American journal of roentgenology. 2012;198(3):505-511.

Johnson JE, Gulanick M, Penckofer S et al.(2014) Does Knowledge of Coronary Artery Calcium Affect Cardiovascular Risk Perception, Likelihood of Taking Action, and Health-Promoting Behavior Change? J Cardiovasc Nurs 2014.

Johnson JE, Gulanick M, Penckofer S, Kouba J.(2015) Does knowledge of coronary artery calcium affect cardiovascular risk perception, likelihood of taking action, and health-promoting behavior change? The Journal of cardiovascular nursing. 2015;30(1):15-25.

Kaiser C, Bremerich J, et al.(2005) Limited diagnostic yield of non-invasive coronary angiography by 16-slice multidetector spiral computed tomography in routine patients referred for evaluation of coronary artery disease. Eur Heart J 2005; (Epub ahead print).

Klocke FJ, Baird MG, Bateman TM, et al.(2003) Guidelines for the Clinical Use of Cardiac Radionuclide Imaging. ACC/AHA Practice Guidelines 2003; 2-60.

Kong Y, Ha T, Kang J, Hwang S, Lee S, Kim Y. (2015) Incidence and Predictors of Increased Coronary Calcium Scores in Liver Transplant Recipients. Transplant Proc 2015;47:1933–1938.

Kong YG, Kang JW, Kim YK et al. (2015) Preoperative coronary calcium score is predictive of early postoperative cardiovascular complications in liver transplant recipients. Br J Anaesth 2015;114:437–443.

Korley FK, George RT, Jaffe AS, et al.(2015) Low high-sensitivity troponin I and zero coronary artery calcium score identifies coronary CT angiography candidates in whom further testing could be avoided Acad Radiol. Aug 2015;22(8):1060-1067. PMID 26049777

Kuettner A, Trabold T, et al.(2004) Noninvasive detection of coronary lesions using 16-detector multislice spiral computed tomography technology: initial clinical resutls. J Am Coll Cardiol 2004; 44:1230-7.

Laudon DA, Behrenbeck TR, Wood CM et al.(2010) Computed tomographic coronary artery calcium assessment for evaluating chest pain in the emergency department: long-term outcome of a prospective blind study. Mayo Clin Proc 2010; 85(4):314-22.

Leber AW, Knez A, et al.(2005) Quantification of obstructive and nonobstructive coronary lesions by 64-slice computed tomography. J Am Coll Cardiol 2005; 46:147-54.

Lee KY, Hwang BH, Kim TH, et al.(2017) Computed tomography angiography images of coronary artery stenosis provide a better prediction of risk than traditional risk factors in asymptomatic individuals with type 2 diabetes: a long-term study of clinical outcomes. Diabetes Care. Sep 2017;40(9):1241-1248. PMID 28663384

Lehmann N, Erbel R, Mahabadi AA, et al.(2018) Value of Progression of Coronary Artery Calcification for Risk Prediction of Coronary and Cardiovascular Events: Result of the HNR Study (Heinz Nixdorf Recall). Circulation. Feb 13 2018; 137(7): 665-679. PMID 29142010

Levsky JM, Haramati LB, Spevack DM, et al.(2018) Coronary Computed Tomography Angiography Versus Stress Echocardiography in Acute Chest Pain: A Randomized Controlled Trial. JACC Cardiovasc Imaging. Jun 8 2018. PMID 29909113

Lin JS. Nontraditional risk factors in cardiovascular disease risk assessment:(2018) Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. Jul 17 2018;320(3):281-297. PMID 29998301

Linde JJ, Hove JD, Sorgaard M, et al.(2015) Long-term clinical impact of coronary CT angiography in patients with recent acute-onset chest pain: the randomized controlled CATCH Trial. JACC Cardiovasc Imaging. Dec 2015;8(12):1404-1413. PMID 26577263

Linde JJ, Kofoed KF, Sorgaard M, et al.(2013) Cardiac computed tomography guided treatment strategy in patients with recent acute-onset chest pain: results from the randomised, controlled trial: CArdiac cT in the treatment of acute CHest pain (CATCH). Int J Cardiol. Oct 15 2013;168(6):5257-5262. PMID 23998546

Lloyd-Jones DM, Braun LT, Ndumele CE, et al.(2018) Use of Risk Assessment Tools to Guide Decision-Making in the Primary Prevention of Atherosclerotic Cardiovascular Disease: A Special Report From the American Heart Association and American College of Cardiology. Circulation, 2018 Dec 28;139(25). PMID 30586766

Lo-Kioeng-Shioe MS, Rijlaarsdam-Hermsen D, van Domburg RT, et al.(2019) Prognostic value of coronary artery calcium score in symptomatic individuals: A meta-analysis of 34,000 subjects. Int. J. Cardiol., 2019 Jun 24. PMID 31229262

Lu MT, Ferencik M, Roberts RS et al(2017) Noninvasive FFR Derived From Coronary CT Angiography J Am Coll Cardiol Img 2017 Nov, 10 (11) 1350-1358

Mamudu HM, Paul TK, Veeranki SP, et al.(2014) The effects of coronary artery calcium screening on behavioral modification, risk perception, and medication adherence among asymptomatic adults: a systematic review. Atherosclerosis. Oct 2014;236(2):338-350. PMID 25128971

Mark DB, Berman DS, Budoff MJ et al.(2010) ACCF/ACR/AHA/NASCI/SAIP/SCAI/SCCT 2010 expert consensus document on coronary computed tomographic angiography: a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents. Circulation 2010; 121(22):2509-43.

Marquering HA, Dijkstra J, et al.(2005) Towards quantitative analysis of coronary CTA. Int J Cardiovasc Imaging 2005; 21:73-84.

McEvoy JW, Blaha MJ, Nasir K et al.(2011) Impact of coronary computed tomographic angiography results on patient and physician behavior in a low-risk population. Arch Intern Med 2011; 171(14):1260-8.

McKavanagh P, Lusk L, Ball PA, et al.(2015) A comparison of cardiac computerized tomography and exercise stress electrocardiogram test for the investigation of stable chest pain: the clinical results of the CAPP randomized prospective trial. Eur Heart J Cardiovasc Imaging. Apr 2015;16(4):441-448. PMID 25473041

Mollet NR, Cademartiri F, et al.(2005) Improved diagnostic accuracy with 16-row multi-slice computed tomography coronary angiography. J Am Coll Cardiol 2005; 45:128-32.

Mozaffarian D.(2005) Electron Beam Computed Tomography of Coronary Calcium. JAMA 2005; 294(22):2897-2900.

Multislice spiral computed tomography for detection and evaluation of coronary artery disease. Hayes Technology Assessment. July 2004.

Murtagh J, Foerster V, et al.(2006) Clinical and cost effectiveness of CT and MRI for selected clinical disorders: results of two systematic reviews. Canadian Agency for Drugs and Technologies in Health; 2006.

Nakanishi R, Li D, Blaha MJ, et al.(2016) All-cause mortality by age and gender based on coronary artery calcium scores. European heart journal cardiovascular Imaging. 2016;17(11):1305-1314.

Nasis A, Meredith IT, Sud PS, et al.(2014) Long-term outcome after CT angiography in patients with possible acute coronary syndrome. Radiology. Sep 2014;272(3):674-682. PMID 24738614.

National Institute for Health and Care Excellence.(2018) Chest pain of recent onset: assessment and diagnosis [CG95]. 2016; https://www.nice.org.uk/guidance/cg95/chapter/Recommendations. Accessed August 27, 2018.

Nieman K, Cademartiri F, Lemos PA, et al.(2002) Reliable noninvasive coronary angiography with fast submillimeter multislice spiral computed tomography. Circulation 2002; 106:2051-4.

Norgaard BL, Hjort J, Gaur S, et al(2017) Clinical Use of Coronary CTA-Derived FFR for Decision-Making in Stable CAD JACC Cardiovasc Imaging May 2017; 10(5): 541-550 PMID 27085447

Nørgaard BL, Terkelsen CJ, Mathiassen ON et al(2018) Coronary CT Angiographic and Flow Reserve-Guided Management of Patients With Stable Ischemic Heart Disease J Am Coll Cardiol 2018 Oct, 72 (18) 2123-2134

Norgan-Hughes GJ, Roobottom CA, et al.(2005) Highly accurate coronary angiography with submillimetre, 16 slice computed tomography. Heart 2005; 91:308-13.

O’Rourke RA, Brundage BH, Froelicher VF, et al.(2000) American College of Cardiology/American Heart Association expert consensus document on electronbeam computed tomography for the diagnosis and prognosis of coronary artery disease. JACC 2000; 36(1):326-40.

Otto CM, Nishimura RA, Bonow RO, et al.(2021) 2020 ACC/AHA Guideline for the management of patients with valvular heart disease: A report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Journal of the American College of Cardiology. 2021;77(4):e25-e197

Polonsky TS, McClelland RL, Jorgensen NW, et al.(2010) Coronary artery calcium score and risk classification for coronary heart disease prediction. JAMA. 2010 Apr 28;303(16):1610-6.

Poon M, Lesser JR, Biga C et al(2020) Current Evidence and Recommendations for Coronary CTA First in Evaluation of Stable Coronary Artery Disease J Am Coll Cardiol 2020 Sep, 76 (11) 1358-1362

Pursnani A, Chou ET, Zakroysky P, et al.(2015) Use of coronary artery calcium scanning beyond coronary computed tomographic angiography in the emergency department evaluation for acute chest pain: the ROMICAT II trial. Circ Cardiovasc Imaging. Mar 2015;8(3).

Raggi P, Callister TQ, Cooil B, et al.(2000) Identification of patients at increased risk of first unheralded acute myocardial infarction by electron-beam computed tomography. Circ 2000; 101(8):850-5.

Ropers D, Baum U, Pohle K, et al.(2003) Detection of coronary artery stenoses with thin-slice multi-detector row spiral computed tomography and multiplanar reconstruction. Circulation 2003;107:664-6.

Ropers D, Moshage W, Daniel WG, et al.(2001) Visualization of coronary artery anomalies and their anatomic course by contrast-enhanced electron beam tomography and three-dimensional reconstruction. Am J Cardiol 2001; 87:193-7.

Rourke RA, Brundage BH, Froelicher VF, et al.(2000) American College of Cardiology/American Heart Association expert consensus document on electronbeam computed tomography for the diagnosis and prognosis of coronary artery disease. JACC 2000; 36:326-40.

Rozanski A, Gransar H, Shaw LJ et al.(2011) Impact of coronary artery calcium scanning on coronary risk factors and downstream testing. J Am Coll Cardiol 2011; 57(15):1622-32.

Sabour S, Rutten A, van der Schouw YT, et al.(2007) Inter-scan reproducibility of coronary calcium measurement using Multi Detector-Row Computed Tomography (MDCT) Eur J Epidemiol. 2007;22(4):235-243.

Sand NPR, Veien KT, Nielsen SS et al(2018) Prospective Comparison of FFR Derived From Coronary CT Angiography With SPECT Perfusion Imaging in Stable Coronary Artery Disease J Am Coll Cardiol Img 2018 Nov, 11 (11) 1640-1650

Sandstedt M, De Geer J, Henriksson L, et al.(2018) Long-term prognostic value of coronary computed tomography angiography in chest pain patients. Acta Radiol. Jan 1 2018:284185118773551. PMID 29742921

Sarwar A, Shaw LJ, Shapiro MD, et al.(2009) Diagnostic and prognostic value of absence of coronary artery calcification. JACC Cardiovascular imaging. 2009;2(6):675-688.

Scanlon PJ, Faxon DP, Audet AM, et al.(1999) ACC/AHA Guidelines for Coronary Angiography. JACC 1999; 33:6(1756-824.

Schlett CL, Banerji D, Siegel E, et al.(2011) Prognostic value of CT angiography for major adverse cardiac events in patients with acute chest pain from the emergency department: 2-year outcomes of the ROMICAT trial. JACC Cardiovasc Imaging. May 2011;4(5):481-491. PMID 21565735

Schlosser T, Konorza T, et al.(2004) Noninvasive visualization fo coronary artery bypass grafts using 16-detector row computed tomography. J Am Coll Cardiol 2004; 44:1238-40.

Schuijf JD, Bax JJ, et al.(2005) Noninvasive coronary imaging and assessment of left ventricular function using 16-slice computed tomography. Am J Cardiol 2005; 95:571-4.

SCOT-HEART Investigators, Newby DE, Adamson PD, Berry C, Boon NA et al. (2018) Coronary CT Angiography and 5-Year Risk of Myocardial Infarction N Engl J Med. 2018 Sep 6;379(10):924-933. doi: 10.1056/NEJMoa1805971. Epub 2018 Aug 25. PMID: 30145934

Scot-Heart Investigators.(2015) CT coronary angiography in patients with suspected angina due to coronary heart disease (SCOT-HEART): an open-label, parallel-group, multicentre trial. Lancet. Jun 13 2015;385(9985):2383-2391. PMID 25788230

Shreibati JB, Baker LC, Hlatky MA.(2011) Association of coronary CT angiography or stress testing with subsequent utilization and spending among Medicare beneficiaries. JAMA 2011; 306(19):2128-36.

Shreibati JB, Baker LC, McConnell MV, et al.(2014) Outcomes after coronary artery calcium and other cardiovascular biomarker testing among asymptomatic medicare beneficiaries. Circ Cardiovasc Imaging. Jul 2014;7(4):655-662. PMID 24777939

Silverman MG, Blaha MJ, Krumholz HM, et al.(2014) Impact of coronary artery calcium on coronary heart disease events in individuals at the extremes of traditional risk factor burden: the Multi-Ethnic Study of Atherosclerosis. Eur Heart J. Sep 1 2014;35(33):2232-2241. PMID 24366919

Skelly AC, Hashimoto R, Buckley DI, et al.(2016) Noninvasive testing for coronary artery disease (Comparative Effectiveness Review No. 171). Rockville (MD): Agency for Healthcare Research and Quality; 2016.

Smeeth L, Skinner JS, Ashcroft J, Hemingway H, Timmis A(2010) Chest Pain Guideline Development G. NICE clinical guideline: chest pain of recent onset. Br J Gen Pract. 2010;60(577):607-610.

Takamura K, Fujimoto S, Kondo T, et al.(2017) Incremental prognostic value of coronary computed tomography angiography: high-risk plaque characteristics in asymptomatic patients. J Atheroscler Thromb. Nov 1 2017;24(11):1174-1185. PMID 28674321

Taylor AJ, Cerqueira M, Hodgson JM et al.(2010) ACCF/SCCT/ACR/AHA/ASE/ASNC/NASCI/SCAI/SCMR 2010 appropriate use criteria for cardiac computed tomography. A report of the American College of Cardiology Foundation Appropriate Use Criteria Task Force, the Society of Cardiovascular Computed Tomography, the American College of Radiology, the American Heart Association, the American Society of Echocardiography, the American Society of Nuclear Cardiology, the North American Society for Cardiovascular Imaging, the Society for Cardiovasc

TEC Contrast-Enhanced Cardiac Computed Tomographic Angiography for Coronary Artery Evaluation. 2005 Blue Cross Blue Shield Association Technology Evaluation Center Assessment.

ten Kate GJ, Caliskan K, Dedic A et al.(2013) Computed tomography coronary imaging as a gatekeeper for invasive coronary angiography in patients with newly diagnosed heart failure of unknown aetiology. Eur J Heart Fail 2013; 15(9):1028-34.

Trikalinos TA, Terasawa T, Raman G, et al.(2010) A systemic review of loss-of-heterozygosity based topographic genotyping with PathfinderTG. AHRQ Technology Assesment.

U.S. Preventive Services Task Force.(2009) Using nontraditional risk factors in coronary heart disease risk assessment: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2009; 151(7):474-82.

U.S. Preventive Services Task Force.(2018) Cardiovascular Disease: Risk Assessment With Nontraditional Risk Factors. 2018; https://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/cardiovascular-diseasescreening-using-nontraditional-risk-assessment. Accessed August 29, 2018.

Wayhs R, Zelinger A, Raggi P.(2002) High coronary artery calcium scores pose an extremely elevated risk for hard events. JACC 2002; 39(2):225-30.

Williams MC, Golay SK, Hunter A, et al.(2015) Observer variability in the assessment of CT coronary angiography and coronary artery calcium score: substudy of the Scottish COmputed Tomography of the HEART (SCOT-HEART) trial. Open Heart. 2015;2(1):e000234.

Williams MC, Hunter A, Shah A, et al.(2017) Symptoms and quality of life in patients with suspected angina undergoing CT coronary angiography: a randomised controlled trial. Heart. Jul 2017;103(13):995-1001. PMID 28246175

Won KB, Chang HJ, Niinuma H, et al(2015) Evaluation of the predictive value of coronary artery calcium score for obstructive coronary artery disease in asymptomatic Korean patients with type 2 diabetes mellitus. Coron Artery Dis. 2015;26(2):150-156.

Wong ND, Hsu JC, Detrano RC, et al.(2000) Coronary artery calcium evaluation by electron beam computed tomography and its relation to new cardiovascular events. Am J Cardiol 2000; 86(5) 495-8.

Xie X, Zhao Y, de Bock GH, et al.(2013) Validation and prognosis of coronary artery calcium scoring in non-triggered thoracic computed tomography: systematic review and meta-analysis Circulation Cardiovascular imaging. 2013;6(4):514-521.

Yerramasu A, Lahiri A, Venuraju S, et al.(2014) Diagnostic role of coronary calcium scoring in the rapid access chest pain clinic: prospective evaluation of NICE guidance. European heart journal cardiovascular Imaging. 2014;15(8):886-892.

Yoon HC, Goldin JG, Greaser LE, et al.(2000) Interscan variation in coronary artery calcium quantification in a large asymptomatic patient population. Am J Rad 2000; 174:803.

Yoon YE, Chang SA, Choi SI, et al.(2012) The absence of coronary artery calcification does not rule out the presence of significant coronary artery disease in Asian patients with acute chest pain. The international journal of cardiovascular imaging. 2012;28(2):389-398.


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.