Pathophysiology Of Heart Disease Leonard Lilly Pdf Creator

Pathophysiology of heart disease: a collaborative project of medical students and faculty. Internet Archive Language. Sabatine, and Leonard S. Lilly - Valvular heart disease / Mia M. Edwards, Patrick T. O'Gara, and Leonard S. Lilly - Heart failure / Ravi Vikram Shah and Michael A. Fifer - The cardiomyopathies / Marc N. Why Your Heart Is Not A Pump (& What Most Doctors Don’t Know About The True Cause Of Heart Disease). Both drawn to the art of healing and repelled by the way medicine was―and continues to be―practiced in the United States, Cowan returned from Swaziland, went to medical school, and established a practice in New Hampshire and, later, San Francisco. Pathophysiology of Heart Disease: A Collaborative Project of Medical Students and Faculty PDF Tags. Pathophysiology of Heart Disease: A Collaborative Project of Medical Students and Faculty pdf download, Pathophysiology of Heart Disease: A Collaborative Project of Medical Students and Faculty pdf, Pathophysiology.

  1. Pathophysiology Of Cardiovascular Disease Pdf
  2. Pathophysiology Of Heart Disease Pdf
Myocardial infarction
Other namesAcute myocardial infarction (AMI), heart attack
Diagram showing the blood supply to the heart by the two major blood vessels, the left and right coronary arteries (labelled LCA and RCA). A myocardial infarction (2) has occurred with blockage of a branch of the left coronary artery (1).
SpecialtyCardiology, emergency medicine
SymptomsChest pain, shortness of breath, nausea, feeling faint, cold sweat, feeling tired; arm, neck, back, jaw, or stomach pain[1][2]
ComplicationsHeart failure, irregular heartbeat, cardiogenic shock, cardiac arrest[3][4]
CausesUsually coronary artery disease[3]
Risk factorsHigh blood pressure, smoking, diabetes, lack of exercise, obesity, high blood cholesterol[5][6]
Diagnostic methodElectrocardiograms (ECGs), blood tests, coronary angiography[7]
TreatmentPercutaneous coronary intervention, thrombolysis[8]
MedicationAspirin, nitroglycerin, heparin[8][9]
PrognosisSTEMI 10% risk of death (developed world)[8]
Frequency15.9 million (2015)[10]

Myocardial infarction (MI), also known as a heart attack, occurs when blood flow decreases or stops to a part of the heart, causing damage to the heart muscle.[1] The most common symptom is chest pain or discomfort which may travel into the shoulder, arm, back, neck or jaw.[1] Often it occurs in the center or left side of the chest and lasts for more than a few minutes.[1] The discomfort may occasionally feel like heartburn.[1] Other symptoms may include shortness of breath, nausea, feeling faint, a cold sweat or feeling tired.[1] About 30% of people have atypical symptoms.[8] Women more often present without chest pain and instead have neck pain, arm pain or feel tired.[11] Among those over 75 years old, about 5% have had an MI with little or no history of symptoms.[12] An MI may cause heart failure, an irregular heartbeat, cardiogenic shock or cardiac arrest.[3][4]

Most MIs occur due to coronary artery disease.[3] Risk factors include high blood pressure, smoking, diabetes, lack of exercise, obesity, high blood cholesterol, poor diet and excessive alcohol intake, among others.[5][6] The complete blockage of a coronary artery caused by a rupture of an atherosclerotic plaque is usually the underlying mechanism of an MI.[3] MIs are less commonly caused by coronary artery spasms, which may be due to cocaine, significant emotional stress and extreme cold, among others.[13][14] A number of tests are useful to help with diagnosis, including electrocardiograms (ECGs), blood tests and coronary angiography.[7] An ECG, which is a recording of the heart's electrical activity, may confirm an ST elevation MI (STEMI), if ST elevation is present.[8][15] Commonly used blood tests include troponin and less often creatine kinase MB.[7]

Treatment of an MI is time-critical.[16]Aspirin is an appropriate immediate treatment for a suspected MI.[9]Nitroglycerin or opioids may be used to help with chest pain; however, they do not improve overall outcomes.[8][9]Supplemental oxygen is recommended in those with low oxygen levels or shortness of breath.[9] In a STEMI, treatments attempt to restore blood flow to the heart and include percutaneous coronary intervention (PCI), where the arteries are pushed open and may be stented, or thrombolysis, where the blockage is removed using medications.[8] People who have a non-ST elevation myocardial infarction (NSTEMI) are often managed with the blood thinner heparin, with the additional use of PCI in those at high risk.[9] In people with blockages of multiple coronary arteries and diabetes, coronary artery bypass surgery (CABG) may be recommended rather than angioplasty.[17] After an MI, lifestyle modifications, along with long term treatment with aspirin, beta blockers and statins, are typically recommended.[8]

Worldwide, about 15.9 million myocardial infarctions occurred in 2015.[10] More than 3 million people had an ST elevation MI, and more than 4 million had an NSTEMI.[18] STEMIs occur about twice as often in men as women.[19] About one million people have an MI each year in the United States.[3] In the developed world, the risk of death in those who have had an STEMI is about 10%.[8] Rates of MI for a given age have decreased globally between 1990 and 2010.[20] In 2011, a MI was one of the top five most expensive conditions during inpatient hospitalizations in the US, with a cost of about $11.5 billion for 612,000 hospital stays.[21]

  • 2Signs and symptoms
  • 3Causes
  • 4Mechanism
  • 5Diagnosis
  • 6Management
  • 7Prevention
    • 7.1Primary prevention
    • 7.2Secondary prevention
  • 8Prognosis
  • 10Society and culture

Terminology[edit]

Myocardial infarction (MI) refers to tissue death (infarction) of the heart muscle (myocardium). It is a type of acute coronary syndrome, which describes a sudden or short-term change in symptoms related to blood flow to the heart.[22] Unlike other causes of acute coronary syndromes, such as unstable angina, a myocardial infarction occurs when there is cell death, as measured by a blood test for biomarkers (the cardiac protein troponin or the cardiac enzyme CK-MB).[16] When there is evidence of an MI, it may be classified as an ST elevation myocardial infarction (STEMI) or Non-ST elevation myocardial infarction (NSTEMI) based on the results of an ECG.[23]

The phrase 'heart attack' is often used non-specifically to refer to a myocardial infarction and to sudden cardiac death. An MI is different from—but can cause—cardiac arrest, where the heart is not contracting at all or so poorly that all vital organs cease to function, thus causing death. It is also distinct from heart failure, in which the pumping action of the heart is impaired. However, an MI may lead to heart failure.[24]

Signs and symptoms[edit]

Areas where pain is experienced in myocardial infarction, showing common (dark red) and less common (light red) areas on the chest and back.

Pain[edit]

Chest pain is the most common symptom of acute myocardial infarction and is often described as a sensation of tightness, pressure, or squeezing. Pain radiates most often to the left arm, but may also radiate to the lower jaw, neck, right arm, back, and upper abdomen.[25] The pain most suggestive of an acute MI, with the highest likelihood ratio, is pain radiating to the right arm and shoulder.[26] Similarly, chest pain similar to a previous heart attack is also suggestive.[27] The pain associated with MI is usually diffuse, does not change with position, and lasts for more than 20 minutes.[23]Levine's sign, in which a person localizes the chest pain by clenching one or both fists over their sternum, has classically been thought to be predictive of cardiac chest pain, although a prospective observational study showed it had a poor positive predictive value.[28] Pain that responds to nitroglycerin does not indicate the presence or absence of a myocardial infarction.[9]

Other symptoms[edit]

Chest pain may be accompanied by sweating, nausea or vomiting, and fainting,[23][26] and these symptoms may also occur without any pain at all.[25] In women, the most common symptoms of myocardial infarction include shortness of breath, weakness, and fatigue.[29]Shortness of breath is a common, and sometimes the only symptom, occurring when damage to the heart limits the output of the left ventricle, with breathlessness arising either from low oxygen in the blood, or pulmonary edema.[25][30] Other less common symptoms include weakness, light-headedness, palpitations, and abnormalities in heart rate or blood pressure.[16] These symptoms are likely induced by a massive surge of catecholamines from the sympathetic nervous system, which occurs in response to pain and, where present, low blood pressure.[31]Loss of consciousness due to inadequate blood flow to the brain and cardiogenic shock, and sudden death, frequently due to the development of ventricular fibrillation, can occur in myocardial infarctions.[24] Cardiac arrest, and atypical symptoms such as palpitations, occur more frequently in women, the elderly, those with diabetes, in people who have just had surgery, and in critically ill patients.[23]

'Silent' myocardial infarctions can happen without any symptoms at all.[12] These cases can be discovered later on electrocardiograms, using blood enzyme tests, or at autopsy after a person has died. Such silent myocardial infarctions represent between 22 and 64% of all infarctions,[12] and are more common in the elderly,[12] in those with diabetes mellitus[16] and after heart transplantation. In people with diabetes, differences in pain threshold, autonomic neuropathy, and psychological factors have been cited as possible explanations for the lack of symptoms.[32] In heart transplantation, the donor heart is not fully innervated by the nervous system of the recipient.[33]

Women[edit]

Range of myocardial infarction symptoms in women

In women, myocardial infarctions can present with different symptoms. The classic presentation of chest pain occurs in about 50% of women. Women can also commonly experience back or neck pain, indigestion, heartburn, lightheadedness, shortness of breath, fatigue, nausea, or pain in the back of the jaw. These symptoms are often overlooked or mistaken for another condition.[34][35]

Causes[edit]

The most prominent risk factors for myocardial infarction are older age, actively smoking, high blood pressure, diabetes mellitus, and total cholesterol and high-density lipoprotein levels.[19] Many risk factors of myocardial infarction are shared with coronary artery disease, the primary cause of myocardial infarction,[16] with other risk factors including male sex, low levels of physical activity, a past family history, obesity, and alcohol use.[16] Risk factors for myocardial disease are often included in risk factor stratification scores, such as the Framingham Risk Score.[19] At any given age, men are more at risk than women for the development of cardiovascular disease.[36]High levels of blood cholesterol is a known risk factor, particularly high low-density lipoprotein, low high-density lipoprotein, and high triglycerides.[37]

Many risk factors for myocardial infarction are potentially modifiable, with the most important being tobacco smoking (including secondhand smoke).[16] Smoking appears to be the cause of about 36% and obesity the cause of 20% of coronary artery disease.[38] Lack of physical activity has been linked to 7–12% of cases.[38][39] Less common causes include stress-related causes such as job stress, which accounts for about 3% of cases,[38] and chronic high stress levels.[40]

Diet[edit]

There is varying evidence about the importance of saturated fat in the development of myocardial infarctions. Eating polyunsaturated fat instead of saturated fats has been shown in studies to be associated with a decreased risk of myocardial infarction,[41] while other studies find little evidence that reducing dietary saturated fat or increasing polyunsaturated fat intake affects heart attack risk.[42][43] Dietary cholesterol does not appear to have a significant effect on blood cholesterol and thus recommendations about its consumption may not be needed.[44]Trans fats do appear to increase risk.[42] Acute and prolonged intake of high quantities of alcoholic drinks (3–4 or more daily) increases the risk of a heart attack.[45]

Genetics[edit]

Family history of ischemic heart disease or MI, particularly if one has a male first-degree relative (father, brother) who had a myocardial infarction before age 55 years, or a female first-degree relative (mother, sister) less than age 65 increases a person's risk of MI.[36]

Genome-wide association studies have found 27 genetic variants that are associated with an increased risk of myocardial infarction.[46] The strongest association of MI has been found with chromosome 9 on the short arm p at locus 21, which contains genes CDKN2A and 2B, although the single nucleotide polymorphisms that are implicated are within a non-coding region.[46] The majority of these variants are in regions that have not been previously implicated in coronary artery disease. The following genes have an association with MI: PCSK9, SORT1, MIA3, WDR12, MRAS, PHACTR1, LPA, TCF21, MTHFDSL, ZC3HC1, CDKN2A, 2B, ABO, PDGF0, APOA5, MNF1ASM283, COL4A1, HHIPC1, SMAD3, ADAMTS7, RAS1, SMG6, SNF8, LDLR, SLC5A3, MRPS6, KCNE2.[46]

Other[edit]

The risk of having a myocardial infarction increases with older age, low physical activity, and low socioeconomic status.[36] Heart attacks appear to occur more commonly in the morning hours, especially between 6AM and noon.[47] Evidence suggests that heart attacks are at least three times more likely to occur in the morning than in the late evening.[48]Shift work is also associated with a higher risk of MI.[49] And one analysis has found an increase in heart attacks immediately following the start of daylight saving time.[50]

Women who use combined oral contraceptive pills have a modestly increased risk of myocardial infarction, especially in the presence of other risk factors.[51] The use of non-steroidal anti inflammatory drugs (NSAIDs), even for as short as a week, increases risk.[52]

Endometriosis in women under the age of 40 is an identified risk factor.[53]

Air pollution is also an important modifiable risk. Short-term exposure to air pollution such as carbon monoxide, nitrogen dioxide, and sulfur dioxide (but not ozone) have been associated with MI and other acute cardiovascular events.[54] For sudden cardiac deaths, every increment of 30 units in Pollutant Standards Index correlated with an 8% increased risk of out-of-hospital cardiac arrest on the day of exposure.[55] Extremes of temperature are also associated.[56]

A number of acute and chronic infections including Chlamydophila pneumoniae, influenza, Helicobacter pylori, and Porphyromonas gingivalis among others have been linked to atherosclerosis and myocardial infarction.[57] As of 2013, there is no evidence of benefit from antibiotics or vaccination, however, calling the association into question.[57][58] Myocardial infarction can also occur as a late consequence of Kawasaki disease.[59]

Calcium deposits in the coronary arteries can be detected with CT scans. Calcium seen in coronary arteries can provide predictive information beyond that of classical risk factors.[60]High blood levels of the amino acid homocysteine is associated with premature atherosclerosis;[61] whether elevated homocysteine in the normal range is causal is controversial.[62]

In people without evident coronary artery disease, possible causes for the myocardial infarction are coronary spasm or coronary dissection.[63]

Mechanism[edit]

Atherosclerosis[edit]

The animation shows plaque buildup or a coronary artery spasm can lead to a heart attack and how blocked blood flow in a coronary artery can lead to a heart attack.
A myocardial infarction occurs when an atheroscleroticplaque slowly builds up in the inner lining of a coronary artery and then suddenly ruptures, causing catastrophic thrombus formation, totally occluding the artery and preventing blood flow downstream.

The most common cause of a myocardial infarction is the rupture of an atherosclerotic plaque on an artery supplying heart muscle.[24][64] Plaques can become unstable, rupture, and additionally promote the formation of a blood clot that blocks the artery; this can occur in minutes. Blockage of an artery can lead to tissue death in tissue being supplied by that artery.[65] Atherosclerotic plaques are often present for decades before they result in symptoms.[65]

The gradual buildup of cholesterol and fibrous tissue in plaques in the wall of the coronary arteries or other arteries, typically over decades, is termed atherosclerosis.[66] Atherosclerosis is characterized by progressive inflammation of the walls of the arteries.[65] Inflammatory cells, particularly macrophages, move into affected arterial walls. Over time, they become laden with cholesterol products, particularly LDL, and become foam cells. A cholesterol core forms as foam cells die. In response to growth factors secreted by macrophages, smooth muscle and other cells move into the plaque and act to stabilize it. A stable plaque may have a thick fibrous cap with calcification. If there is ongoing inflammation, the cap may be thin or ulcerate. Exposed to the pressure associated with blood flow, plaques, especially those with a thin lining, may rupture and trigger the formation of a blood clot (thrombus).[65] The cholesterol crystals have been associated with plaque rupture through mechanical injury and inflammation.[67]

Other causes[edit]

Atherosclerotic disease is not the only cause of myocardial infarction, and it may exacerbate or contribute to other causes. A myocardial infarction may result from a heart with a limited blood supply subject to increased oxygen demands, such as in fever, a fast heart rate, hyperthyroidism, too few red blood cells in the bloodstream, or low blood pressure. Damage or failure of procedures such as percutaneous coronary intervention or coronary artery bypass grafts may cause a myocardial infarction. Spasm of coronary arteries, such as Prinzmetal's angina may cause blockage.[23][25]

Tissue death[edit]

Drawing of the heart showing anterior left ventricle wall infarction

If impaired blood flow to the heart lasts long enough, it triggers a process called the ischemic cascade; the heart cells in the territory of the blocked coronary artery die (infarction), chiefly through necrosis, and do not grow back. A collagenscar forms in their place.[65] When an artery is blocked, cells lack oxygen, needed to produce ATP in mitochondria. ATP is required for the maintenance of electrolyte balance, particularly through the Na/K ATPase. This leads to an ischemic cascade of intracellular changes, necrosis and apoptosis of affected cells.[68]

Cells in the area with the worst blood supply, just below the inner surface of the heart (endocardium), are most susceptible to damage.[69] Ischemia first affects this region, the subendocardial region, and tissue begins to die within 15–30 minutes of loss of blood supply.[70] The dead tissue is surrounded by a zone of potentially reversible ischemia that progresses to become a full-thickness transmural infarct.[68][70] The initial 'wave' of infarction can take place over 3–4 hours.[65][68] These changes are seen on gross pathology and cannot be predicted by the presence or absence of Q waves on an ECG.[69] The position, size and extent of an infarct depends on the affected artery, totality of the blockage, duration of the blockage, the presence of collateral blood vessels, oxygen demand, and success of interventional procedures.[25][64]

Tissue death and myocardial scarring alter the normal conduction pathways of the heart, and weaken affected areas. The size and location puts a person at risk of abnormal heart rhythms (arrhythmias) or heart block, aneurysm of the heart ventricles, inflammation of the heart wall following infarction, and rupture of the heart wall that can have catastrophic consequences.[64][71]

Diagnosis[edit]

Criteria[edit]

An acute myocardial infarction, according to current consensus, is defined by elevated cardiac biomarkers with a rising or falling trend and at least one of the following:[23][72]

  • Symptoms relating to ischemia
  • Changes on an electrocardiogram (ECG), such as ST segment changes, new left bundle branch block, or Q waves
  • Changes in the motion of the heart wall on imaging
  • Demonstration of a thrombus on angiogram or at autopsy.

Types[edit]

Myocardial infarctions are generally clinically classified into ST elevation MI (STEMI) and non-ST elevation MI (NSTEMI). These are based on changes to an ECG.[23] STEMIs make up about 25 – 40% of myocardial infarctions.[19] A more explicit classification system, based on international consensus in 2012, also exists. This classifies myocardial infarctions into five types:[23]

  1. Spontaneous MI related to plaque erosion and/or rupture, fissuring, or dissection
  2. MI related to ischemia, such as from increased oxygen demand or decreased supply, e.g. coronary artery spasm, coronary embolism, anemia, arrhythmias, high blood pressure or low blood pressure
  3. Sudden unexpected cardiac death, including cardiac arrest, where symptoms may suggest MI, an ECG may be taken with suggestive changes, or a blood clot is found in a coronary artery by angiography and/or at autopsy, but where blood samples could not be obtained, or at a time before the appearance of cardiac biomarkers in the blood
  4. Associated with coronary angioplasty or stents
    • Associated with percutaneous coronary intervention (PCI)
    • Associated with stent thrombosis as documented by angiography or at autopsy
  5. Associated with CABG
  6. Associated with spontaneous coronary artery dissection in young, fit women

Cardiac biomarkers[edit]

There are a number of different biomarkers used to determine the presence of cardiac muscle damage. Troponins, measured through a blood test, are considered to be the best,[19] and are preferred because they have greater sensitivity and specificity for measuring injury to the heart muscle than other tests.[64] A rise in troponin occurs within 2–3 hours of injury to the heart muscle, and peaks within 1–2 days. The level of the troponin, as well as a change over time, are useful in measuring and diagnosing or excluding myocardial infarctions, and the diagnostic accuracy of troponin testing is improving over time.[64] One high-sensitivity cardiac troponin is able to rule out a heart attack as long as the ECG is normal.[73][74]

Other tests, such as CK-MB or myoglobin, are discouraged.[75] CK-MB is not as specific as troponins for acute myocardial injury, and may be elevated with past cardiac surgery, inflammation or electrical cardioversion; it rises within 4–8 hours and returns to normal within 2–3 days.[25]Copeptin may be useful to rule out MI rapidly when used along with troponin.[76]

Electrocardiogram[edit]

A 12-lead ECG showing a STEMI. Elevation of the ST segment can be seen in some leads.

Electrocardiograms (ECGs) are a series of leads placed on a person's chest that measure electrical activity associated with contraction of heart muscle.[77] The taking of an ECG is an important part in the workup of an AMI,[23] and ECGs are often not just taken once, but may be repeated over minutes to hours, or in response to changes in signs or symptoms.[23]

ECG readouts product a waveform with different labelled features.[77] In addition to a rise in biomarkers, a rise in the ST segment, changes in the shape or flipping of T waves, new Q waves, or a new left bundle branch block can be used to diagnose an AMI.[23] In addition, ST elevation can be used to diagnose an ST segment myocardial infarction (STEMI). A rise must be new in V2 and V3 ≥2 mm (0,2 mV) for males or ≥1.5 mm (0.15 mV) for females or ≥1 mm (0.1 mV) in two other adjacent chest or limb leads.[19][23] ST elevation is associated with infarction, and may be preceded by changes indicating ischemia, such as ST depression or inversion of the T waves.[77] Abnormalities can help differentiate the location of an infarct, based on the leads that are affected by changes.[16] Early STEMIs may be preceded by peaked T waves.[19] Other ECG abnormalities relating to complications of acute myocardial infarctions may also be evident, such as atrial or ventricular fibrillation.[78]

Imaging[edit]

Noninvasive imaging plays an important role in the diagnosis and characterisation of myocardial infarction.[23] Tests such as chest X-rays can be used to explore and exclude alternate causes of a person's symptoms.[23] Tests such as stress echocardiography and myocardial perfusion imaging can confirm a diagnosis when a person's history, physical examination (including cardiac examination) ECG, and cardiac biomarkers suggest the likelihood of a problem.[79]

Echocardiography, an ultrasound scan of the heart, is able to visualize the heart, its size, shape, and any abnormal motion of the heart walls as they beat that may indicate a myocardial infarction. The flow of blood can be imaged, and contrast dyes may be given to improve image.[23] Other scans using radioactive contrast include SPECTCT-scans using thallium, sestamibi (MIBI scans) or tetrofosmin; or a PET scan using Fludeoxyglucose or rubidium-82.[23] These nuclear medicine scans can visualize the perfusion of heart muscle.[23] SPECT may also be used to determine viability of tissue, and whether areas of ischemia are inducible.[23][80]

Pathophysiology Of Heart Disease Leonard Lilly Pdf Creator

Medical societies and professional guidelines recommend that the physician confirm a person is at high risk for myocardial infarction before conducting imaging tests to make a diagnosis,[79][81] as such tests are unlikely to change management and result in increased costs.[79] Patients who have a normal ECG and who are able to exercise, for example, do not merit routine imaging.[79]

  • Poor movement of the heart due to an MI as seen on ultrasound[82]

  • Pulmonary edema due to an MI as seen on ultrasound[82]

Differential diagnosis[edit]

There are many causes of chest pain, which can originate from the heart, lungs, gastrointestinal tract, aorta, and other muscles, bones and nerves surrounding the chest.[83] In addition to myocardial infarction, other causes include angina, insufficient blood supply (ischemia) to the heart muscles without evidence of cell death, gastroesophageal reflux disease; pulmonary embolism, tumors of the lungs, pneumonia, rib fracture, costochondritis, heart failure and other musculoskeletal injuries.[83][84] Rarer severe differential diagnoses include aortic dissection, esophageal rupture, tension pneumothorax, and pericardial effusion causing cardiac tamponade.[85] The chest pain in an MI may mimic heartburn.[24] Causes of sudden-onset breathlessness generally involve the lungs or heart – including pulmonary edema, pneumonia, allergic reactions and asthma, and pulmonary embolus, acute respiratory distress syndrome and metabolic acidosis.[83] There are many different causes of fatigue, and myocardial infarction is not a common cause.[86]

Management[edit]

Pathophysiology Of Cardiovascular Disease Pdf

A myocardial infarction requires immediate medical attention. Treatment aims to preserve as much heart muscle as possible, and to prevent further complications.[25] Treatment depends on whether the myocardial infarction is a STEMI or NSTEMI.[64] Treatment in general aims to unblock blood vessels, reduce blot clot enlargement, reduce ischemia, and modify risk factors with the aim of preventing future MIs.[25] In addition, the main treatment for myocardial infarctions with ECG evidence of ST elevation (STEMI) include thrombolysis or percutaneous coronary intervention, although PCI is also ideally conducted within 1–3 days for NSTEMI.[64] In addition to clinical judgement, risk stratification may be used to guide treatment, such as with the TIMI and GRACE scoring systems.[16][64][87]

Pain[edit]

The pain associated with myocardial infarction may be treated with nitroglycerin or morphine.[25] Nitroglycerin (given under the tongue or intravenously) may improve the blood supply to the heart, and decrease the work the heart must do.[25] It is an important part of therapy for its pain relief, despite there being no benefit to overall mortality.[25][88] Morphine may also be used, and is effective for the pain associated with STEMI.[25] The evidence for benefit from morphine on overall outcomes, however, is poor and there is some evidence of potential harm.[89][90]

Anticoagulation[edit]

Aspirin, an antiplateletanticoagulant, is given as a loading dose with the goal of reducing the clot size and reduce further clotting in the affected artery.[25][64] It is known to decrease mortality associated with acute myocardial infarction by at least 50%.[64]P2Y12 inhibitors such as clopidogrel, prasugrel and ticagrelor are given concurrently, also as a loading dose, with the dose depending on whether further surgical management or fibrinolysis is planned.[64] Prasugrel and ticagrelor are recommended in European and American guidelines, as they are active more quickly and consistently than clopidogrel.[64] P2Y12 inhibitors are recommended in both NSTEMI and STEMI, including in PCI, with evidence also to suggest improved mortality.[64]Heparins, particularly in the unfractionated form, act at several points in the clotting cascade, help to prevent the enlargement of a clot, and are also given in myocardial infarction, owing to evidence suggesting improved mortality rates.[64] In very high-risk scenarios, inhibitors of the platelet glycoprotein αIIbβ3a receptor such as eptifibatide or tirofiban may be used.[64]

There is varying evidence on the mortality benefits in NSTEMI. A 2014 review of P2Y12 inhibitors such as clopidogrel found they do not change the risk of death when given to people with a suspected NSTEMI prior to PCI,[91] nor do heparins change the risk of death.[92] They do decrease the risk of having a further myocardial infarction.[64][92]

Angiogram[edit]

Primary percutaneous coronary intervention (PCI) is the treatment of choice for STEMI if it can be performed in a timely manner, ideally within 90–120 minutes of contact with a medical provider.[64][93] Some recommend it is also done in NSTEMI within 1–3 days, particularly when considered high-risk.[64] A 2017 review, however, did not find a difference between early versus later PCI in NSTEMI.[94]

PCI involves small probes, inserted through peripheral blood vessels such as the femoral artery or radial artery into the blood vessels of the heart. The probes are then used to identify and clear blockages using small balloons, which are dragged through the blocked segment, dragging away the clot, or the insertion of stents.[25][64]Coronary artery bypass grafting is only considered when the affected area of heart muscle large, and PCI is unsuitable, for example with difficult cardiac anatomy.[95] After PCI, people are generally placed on aspirin indefinitely and on dual antiplatelet therapy (generally aspirin and clopidogrel) for at least a year.[19][64][96]

Fibrinolysis[edit]

If PCI cannot be performed within 90 to 120 minutes in STEMI then fibrinolysis, preferably within 30 minutes of arrival to hospital, is recommended.[64][97] If a person has had symptoms for 12 to 24 hours evidence for effectiveness of thrombolysis is less and if they have had symptoms for more than 24 hours it is not recommended.[98] Thrombolysis involves the administration of medication that activates the enzymes that normally dissolve blood clots. These medications include tissue plasminogen activator, reteplase, streptokinase, and tenecteplase.[25] Thrombolysis is not recommended in a number of situations, particularly when associated with a high risk of bleeding or the potential for problematic bleeding, such as active bleeding, past strokes or bleeds into the brain, or severe hypertension. Situations in which thrombolysis may be considered, but with caution, include recent surgery, use of anticoagulants, pregnancy, and proclivity to bleeding.[25] Major risks of thrombolysis are major bleeding and intracranial bleeding.[25] Pre-hospital thrombolysis reduces time to thrombolytic treatment, based on studies conducted in higher income countries, however it is unclear whether this has an impact on mortality rates.[99]

Other[edit]

In the past, high flow oxygen was recommended for everyone with a possible myocardial infarction.[75] More recently, no evidence was found for routine use in those with normal oxygen levels and there is potential harm from the intervention.[100][101][102][103] Therefore, oxygen is currently only recommended if oxygen levels are found to be low or if someone is in respiratory distress.[25][75]

If despite thrombolysis there is significant cardiogenic shock, continued severe chest pain, or less than a 50% improvement in ST elevation on the ECG recording after 90 minutes, then rescue PCI is indicated emergently.[104][105]

Those who have had cardiac arrest may benefit from targeted temperature management with evaluation for implementation of hypothermia protocols. Furthermore, those with cardiac arrest, and ST elevation at any time, should usually have angiography.[19]Aldosterone antagonists appear to be useful in people who have had an STEMI and do not have heart failure.[106]

Rehabilitation[edit]

Cardiac rehabilitation benefits many who have experienced myocardial infarction,[64] even if there has been substantial heart damage and resultant left ventricular failure. It should start soon after discharge from the hospital. The program may include lifestyle advice, exercise, social support, as well as recommendations about driving, flying, sport participation, stress management, and sexual intercourse.[107]

Prevention[edit]

There is a large crossover between the lifestyle and activity recommendations to prevent a myocardial infarction, and those that may be adopted as secondary prevention after an initial myocardial infarction,[64] because of shared risk factors and an aim to reduce atherosclerosis affecting heart vessels.[25]

Primary prevention[edit]

Lifestyle[edit]

Physical activity can reduce the risk of cardiovascular disease, and people at risk are advised to engage in 150 minutes of moderate or 75 minutes of vigorous intensity aerobic exercise a week.[108] Keeping a healthy weight, drinking alcohol within the recommended limits, and quitting smoking reduce the risk of cardiovascular disease.[108]

Substituting polyunsaturated fats such as olive oil and rapeseed oil instead of saturated fats may reduce the risk of myocardial infarction,[41] although there is not universal agreement.[42] Dietary modifications are recommended by some national authorities, with recommendations including increasing the intake of wholegrain starch, reducing sugar intake (particularly of refined sugar), consuming five portions of fruit and vegetables daily, consuming two or more portions of fish per week, and consuming 4–5 portions of unsalted nuts, seeds, or legumes per week.[108] The dietary pattern with the greatest support is the Mediterranean diet.[109]Vitamins and mineral supplements are of no proven benefit,[110] and neither are plant stanols or sterols.[108]

Public health measures may also act at a population level to reduce the risk of myocardial infarction, for example by reduce unhealthy diets (excessive salt, saturated fat and trans fat) including food labeling and marketing requirements as well as requirements for catering and restaurants, and stimulating physical activity. This may be part of regional cardiovascular disease prevention programs, or through the health impact assessment of regional and local plans and policies.[111]

Most guidelines recommend combining different preventive strategies. A 2015 Cochrane Review found some evidence that such an approach might help with blood pressure, body mass index and waist circumference. However, there was insufficient evidence to show an effect on mortality or actual cardio-vascular events.[112]

Medication[edit]

Statins, drugs that act to lower blood cholesterol, decrease the incidence and mortality rates of myocardial infarctions.[113] They are often recommended in those at an elevated risk of cardiovascular diseases.[108]

Aspirin has been studied extensively in people considered at increased risk of myocardial infarction. Based on numerous studies in different groups (e.g. people with or without diabetes), there does not appear to be a benefit strong enough to outweigh the risk of excessive bleeding.[114][115] Nevertheless, many clinical practice guidelines continue to recommend aspirin for primary prevention,[116] and some researchers feel that those with very high cardiovascular risk but low risk of bleeding should continue to receive aspirin.[117]

Secondary prevention[edit]

There is a large crossover between the lifestyle and activity recommendations to prevent a myocardial infarction, and those that may be adopted as secondary prevention after an initial myocardial infarct.[64] Recommendations include stopping smoking, a gradual return to exercise, eating a healthy diet, low in saturated fat and low in cholesterol, and drinking alcohol within recommended limits, exercising, and trying to achieve a healthy weight.[64][107] Exercise is both safe and effective even if people have had stents or heart failure,[118] and is recommended to start gradually after 1–2 weeks.[64] Counselling should be provided relating to medications used, and for warning signs of depression.[64] Previous studies suggested a benefit from omega-3 fatty acid supplementation but this has not been confirmed.[107]

Medications[edit]

Following a heart attack, nitrates, when taken for two days, and ACE-inhibitors decrease the risk of death.[119] Other medications include:

Aspirin is continued indefinitely, as well as another antiplatelet agent such as clopidogrel or ticagrelor ('dual antiplatelet therapy' or DAPT) for up to twelve months.[107] If someone has another medical condition that requires anticoagulation (e.g. with warfarin) this may need to be adjusted based on risk of further cardiac events as well as bleeding risk.[107] In those who have had a stent, more than 12 months of clopidogrel plus aspirin does not affect the risk of death.[120]

Beta blocker therapy such as metoprolol or carvedilol is recommended to be started within 24 hours, provided there is no acute heart failure or heart block.[19][75] The dose should be increased to the highest tolerated.[107] Contrary to what was long believed, the use of beta blockers does not appear to affect the risk of death, possibly because other treatments for MI have improved.[121] When beta blocker medication is given within the first 24–72 hours of a STEMI no lives are saved. However, 1 in 200 people were prevented from a repeat heart attack, and another 1 in 200 from having an abnormal heart rhythm. Additionally, for 1 in 91 the medication causes a temporary decrease in the heart's ability to pump blood.[122]

ACE inhibitor therapy should be started within 24 hours, and continued indefinitely at the highest tolerated dose. This is provided there is no evidence of worsening kidney failure, high potassium, low blood pressure, or known narrowing of the renal arteries.[64] Those who cannot tolerate ACE inhibitors may be treated with an angiotensin II receptor antagonist.[107]

Statin therapy has been shown to reduce mortality and subsequent cardiac events, and should be commenced with the aim of lowering LDL cholesterol. Other medications, such as ezetimibe, may also be added with this goal in mind.[64]

Aldosterone antagonists (spironolactone or eplerenone) may be used if there is evidence of left ventricular dysfunction after an MI, ideally after beginning treatment with an ACE inhibitor.[107][123]

Other[edit]

A defibrillator, an electric device connected to the heart and surgically inserted under the skin, may be recommended. This is particularly if there are any ongoing signs of heart failure, with a low left ventricular ejection fraction and a New York Heart Association grade II or III after 40 days of the infarction.[64] Defibrillators detect potentially fatal arrhythmia and deliver an electrical shock to the person to depolarize a critical mass of the heart muscle.[124]

Prognosis[edit]

The prognosis after myocardial infarction varies greatly depending on the extent and location of the affected heart muscle, and the development and management of complications.[16] Prognosis is worse with older age, and social isolation.[16] Anterior infarcts, persistent ventricular tachycardia or fibrillation, development of heart blocks, and left ventricular impairment are all associated with poorer prognosis.[16] Without treatment, about a quarter of those affected by MI die within minutes, and about forty percent within the first month.[16] Morbidity and mortality from myocardial infarction has however improved over the years due to earlier and better treatment:[26] in those who have an STEMI in the United States, between 5 and 6 percent die before leaving the hospital and 7 to 18 percent die within a year.[19]

It is unusual for babies to experience a myocardial infarction, but when they do, about half die.[125] In the short-term, neonatal survivors seem to have a normal quality of life.[125]

Complications[edit]

Complications may occur immediately following the myocardial infarction or may take time to develop. Disturbances of heart rhythms, including atrial fibrillation, ventricular tachycardia and fibrillation and heart block can arise as a result of ischemia, cardiac scarring, and infarct location.[16][64]Stroke is also a risk, either as a result of clots transmitted from the heart during PCI, as a result of bleeding following anticoagulation, or as a result of disturbances in the heart's ability to pump effectively as a result of the infarction.[64]Regurgitation of blood through the mitral valve is possible, particularly if the infarction causes dysfunction of the papillary muscle.[64]Cardiogenic shock as a result of the heart being unable to adequately pump blood may develop, dependent on infarct size, and is most likely to occur within the days following an acute myocardial infarction. Cardiogenic shock is the largest cause of in-hospital mortality.[26][64] Rupture of the ventricular dividing wall or left ventricular wall may occur within the initial weeks.[64]Dressler's syndrome, a reaction following larger infarcts and a cause of pericarditis is also possible.[64]

Heart failure may develop as a long-term consequence, with an impaired ability of heart muscle to pump, scarring, and increase in size of the existing muscle. Aneurysm of the left ventricle myocardium develops in about 10% of MI and is itself a risk factor for heart failure, ventricular arrhythmia and the development of clots.[16]

Risk factors for complications and death include age, hemodynamic parameters (such as heart failure, cardiac arrest on admission, systolicblood pressure, or Killip class of two or greater), ST-segment deviation, diabetes, serum creatinine, peripheral vascular disease, and elevation of cardiac markers.[126][127][128]

Epidemiology[edit]

Myocardial infarction is a common presentation of coronary artery disease. The World Health Organization estimated in 2004, that 12.2% of worldwide deaths were from ischemic heart disease;[129] with it being the leading cause of death in high- or middle-income countries and second only to lower respiratory infections in lower-income countries.[129] Worldwide, more than 3 million people have STEMIs and 4 million have NSTEMIs a year.[18] STEMIs occur about twice as often in men as women.[19]

Rates of death from ischemic heart disease (IHD) have slowed or declined in most high-income countries, although cardiovascular disease still accounted for one in three of all deaths in the US in 2008.[130] For example, rates of death from cardiovascular disease have decreased almost a third between 2001 and 2011 in the United States.[131]

In contrast, IHD is becoming a more common cause of death in the developing world. For example, in India, IHD had become the leading cause of death by 2004, accounting for 1.46 million deaths (14% of total deaths) and deaths due to IHD were expected to double during 1985–2015.[132] Globally, disability adjusted life years (DALYs) lost to ischemic heart disease are predicted to account for 5.5% of total DALYs in 2030, making it the second-most-important cause of disability (after unipolar depressive disorder), as well as the leading cause of death by this date.[129]

Society and culture[edit]

Depictions of heart attacks in popular media often include collapsing or loss of consciousness which are not common symptoms; these depictions contribute to widespread misunderstanding about the symptoms of myocardial infarctions, which in turn contributes to people not getting care when they should.[133]

Legal implications[edit]

At common law, in general, a myocardial infarction is a disease, but may sometimes be an injury. This can create coverage issues in the administration of no-fault insurance schemes such as workers' compensation. In general, a heart attack is not covered;[134] however, it may be a work-related injury if it results, for example, from unusual emotional stress or unusual exertion.[135] In addition, in some jurisdictions, heart attacks suffered by persons in particular occupations such as police officers may be classified as line-of-duty injuries by statute or policy. In some countries or states, a person having suffered from an MI may be prevented from participating in activity that puts other people's lives at risk, for example driving a car or flying an airplane.[136]

References[edit]

  1. ^ abcdef'What Are the Signs and Symptoms of Coronary Heart Disease?'. www.nhlbi.nih.gov. September 29, 2014. Archived from the original on 24 February 2015. Retrieved 23 February 2015.
  2. ^'Heart Attack Symptoms in Women'. American Heart Association.
  3. ^ abcdef'What Is a Heart Attack?'. www.nhlbi.nih.gov. December 17, 2013. Archived from the original on 19 February 2015. Retrieved 24 February 2015.
  4. ^ ab'Heart Attack or Sudden Cardiac Arrest: How Are They Different?'. www.heart.org. Jul 30, 2014. Archived from the original on 24 February 2015. Retrieved 24 February 2015.
  5. ^ abMehta PK, Wei J, Wenger NK (February 2015). 'Ischemic heart disease in women: a focus on risk factors'. Trends in Cardiovascular Medicine. 25 (2): 140–51. doi:10.1016/j.tcm.2014.10.005. PMC4336825. PMID25453985.
  6. ^ abMendis, Shanthi; Puska, Pekka; Norrving, Bo (2011). Global atlas on cardiovascular disease prevention and control(PDF) (1st ed.). Geneva: World Health Organization in collaboration with the World Heart Federation and the World Stroke Organization. pp. 3–18. ISBN978-92-4-156437-3. Archived(PDF) from the original on 2014-08-17.
  7. ^ abc'How Is a Heart Attack Diagnosed?'. www.nhlbi.nih.gov. December 17, 2013. Archived from the original on 24 February 2015. Retrieved 24 February 2015.
  8. ^ abcdefghiSteg PG, James SK, Atar D, Badano LP, Blömstrom-Lundqvist C, Borger MA, Di Mario C, Dickstein K, Ducrocq G, Fernandez-Aviles F, Gershlick AH, Giannuzzi P, Halvorsen S, Huber K, Juni P, Kastrati A, Knuuti J, Lenzen MJ, Mahaffey KW, Valgimigli M, van 't Hof A, Widimsky P, Zahger D (October 2012). 'ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation'. European Heart Journal. 33 (20): 2569–619. doi:10.1093/eurheartj/ehs215. PMID22922416.
  9. ^ abcdefO'Connor RE, Brady W, Brooks SC, Diercks D, Egan J, Ghaemmaghami C, Menon V, O'Neil BJ, Travers AH, Yannopoulos D (November 2010). 'Part 10: acute coronary syndromes: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care'. Circulation. 122 (18 Suppl 3): S787–817. doi:10.1161/CIRCULATIONAHA.110.971028. PMID20956226.
  10. ^ abGBD 2015 Disease Injury Incidence Prevalence Collaborators (October 2016). 'Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015'. Lancet. 388 (10053): 1545–1602. doi:10.1016/S0140-6736(16)31678-6. PMC5055577. PMID27733282.
  11. ^Coventry LL, Finn J, Bremner AP (2011). 'Sex differences in symptom presentation in acute myocardial infarction: a systematic review and meta-analysis'. Heart & Lung. 40 (6): 477–91. doi:10.1016/j.hrtlng.2011.05.001. PMID22000678.
  12. ^ abcdValensi P, Lorgis L, Cottin Y (March 2011). 'Prevalence, incidence, predictive factors and prognosis of silent myocardial infarction: a review of the literature'. Archives of Cardiovascular Diseases. 104 (3): 178–88. doi:10.1016/j.acvd.2010.11.013. PMID21497307.
  13. ^'What Causes a Heart Attack?'. www.nhlbi.nih.gov. December 17, 2013. Archived from the original on 18 February 2015. Retrieved 24 February 2015.
  14. ^Devlin RJ, Henry JA (2008). 'Clinical review: Major consequences of illicit drug consumption'. Critical Care. 12 (1): 202. doi:10.1186/cc6166. PMC2374627. PMID18279535.
  15. ^'Electrocardiogram – NHLBI, NIH'. www.nhlbi.nih.gov. 9 December 2016. Archived from the original on 11 April 2017. Retrieved 10 April 2017.
  16. ^ abcdefghijklmnoBritton, the editors Nicki R. Colledge, Brian R. Walker, Stuart H. Ralston; illustrated by Robert (2010). Davidson's principles and practice of medicine (21st ed.). Edinburgh: Churchill Livingstone/Elsevier. pp. 588–599. ISBN978-0-7020-3085-7.
  17. ^Hamm CW, Bassand JP, Agewall S, Bax J, Boersma E, Bueno H, Caso P, Dudek D, Gielen S, Huber K, Ohman M, Petrie MC, Sonntag F, Uva MS, Storey RF, Wijns W, Zahger D (December 2011). 'ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: The Task Force for the management of acute coronary syndromes (ACS) in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC)'. European Heart Journal. 32 (23): 2999–3054. doi:10.1093/eurheartj/ehr236. PMID21873419.
  18. ^ abWhite HD, Chew DP (August 2008). 'Acute myocardial infarction'. Lancet. 372 (9638): 570–84. doi:10.1016/S0140-6736(08)61237-4. PMC1931354. PMID18707987.
  19. ^ abcdefghijklO'Gara PT, Kushner FG, Ascheim DD, Casey DE, Chung MK, de Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, Granger CB, Krumholz HM, Linderbaum JA, Morrow DA, Newby LK, Ornato JP, Ou N, Radford MJ, Tamis-Holland JE, Tommaso CL, Tracy CM, Woo YJ, Zhao DX, Anderson JL, Jacobs AK, Halperin JL, Albert NM, Brindis RG, Creager MA, DeMets D, Guyton RA, Hochman JS, Kovacs RJ, Kushner FG, Ohman EM, Stevenson WG, Yancy CW (January 2013). '2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines'. Circulation. 127 (4): e362–425. doi:10.1161/CIR.0b013e3182742cf6. PMID23247304.
  20. ^Moran AE, Forouzanfar MH, Roth GA, Mensah GA, Ezzati M, Flaxman A, Murray CJ, Naghavi M (April 2014). 'The global burden of ischemic heart disease in 1990 and 2010: the Global Burden of Disease 2010 study'. Circulation. 129 (14): 1493–501. doi:10.1161/circulationaha.113.004046. PMC4181601. PMID24573351.
  21. ^Torio, Celeste (August 2013). 'National Inpatient Hospital Costs: The Most Expensive Conditions by Payer, 2011'. HCUP. Archived from the original on 14 March 2017. Retrieved 1 May 2017.
  22. ^Acute Coronary Syndrome. American Heart Association. Retrieved November 25, 2006. Archived September 25, 2006, at the Wayback Machine
  23. ^ abcdefghijklmnopqrThygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD, Katus HA, Lindahl B, Morrow DA, Clemmensen PM, Johanson P, Hod H, Underwood R, Bax JJ, Bonow RO, Pinto F, Gibbons RJ, Fox KA, Atar D, Newby LK, Galvani M, Hamm CW, Uretsky BF, Steg PG, Wijns W, Bassand JP, Menasché P, Ravkilde J, Ohman EM, Antman EM, Wallentin LC, Armstrong PW, Simoons ML, Januzzi JL, Nieminen MS, Gheorghiade M, Filippatos G, Luepker RV, Fortmann SP, Rosamond WD, Levy D, Wood D, Smith SC, Hu D, Lopez-Sendon JL, Robertson RM, Weaver D, Tendera M, Bove AA, Parkhomenko AN, Vasilieva EJ, Mendis S (October 2012). 'Third universal definition of myocardial infarction'. Circulation. 126 (16): 2020–35. doi:10.1161/CIR.0b013e31826e1058. PMID22923432.
  24. ^ abcdVan de Werf F, Bax J, Betriu A, Blomstrom-Lundqvist C, Crea F, Falk V, Filippatos G, Fox K, Huber K, Kastrati A, Rosengren A, Steg PG, Tubaro M, Verheugt F, Weidinger F, Weis M (December 2008). 'Management of acute myocardial infarction in patients presenting with persistent ST-segment elevation: the Task Force on the Management of ST-Segment Elevation Acute Myocardial Infarction of the European Society of Cardiology'. European Heart Journal. 29 (23): 2909–45. doi:10.1093/eurheartj/ehn416. PMID19004841.
  25. ^ abcdefghijklmnopqrsKasper, Dennis L.; Fauci, Anthony S.; Hauser, Stephen L.; Longo, Dan L.; Jameson, J. Larry; Loscalzo, Joseph (2015). Harrison's principles of internal medicine. McGraw Hill Education. pp. 1593–1610. ISBN978-0-07-180215-4. OCLC923181481.
  26. ^ abcdKasper, Dennis L.; Fauci, Anthony S.; Hauser, Stephen L.; Longo, Dan L.; Jameson, J. Larry; Loscalzo, Joseph (2015). Harrison's Principles of Internal Medicine. McGraw Hill Education. pp. 98–99. ISBN978-0-07-180215-4. OCLC923181481.
  27. ^Gupta R, Munoz R (August 2016). 'Evaluation and Management of Chest Pain in the Elderly'. Emergency Medicine Clinics of North America. 34 (3): 523–42. doi:10.1016/j.emc.2016.04.006. PMID27475013.
  28. ^Marcus GM, Cohen J, Varosy PD, Vessey J, Rose E, Massie BM, Chatterjee K, Waters D (January 2007). 'The utility of gestures in patients with chest discomfort'. The American Journal of Medicine. 120 (1): 83–9. doi:10.1016/j.amjmed.2006.05.045. PMID17208083.
  29. ^Canto JG, Goldberg RJ, Hand MM, Bonow RO, Sopko G, Pepine CJ, Long T (December 2007). 'Symptom presentation of women with acute coronary syndromes: myth vs reality'. Archives of Internal Medicine. 167 (22): 2405–13. doi:10.1001/archinte.167.22.2405. PMID18071161.
  30. ^Ashton, Rendell; Raman, Dileep. 'Dyspnea'. www.clevelandclinicmeded.com. Cleveland Clinic. Archived from the original on 11 July 2017. Retrieved 24 May 2017.
  31. ^Lilly, Leonard S. (2012). Pathophysiology of Heart Disease: A Collaborative Project of Medical Students and Faculty. Lippincott Williams & Wilkins. p. 172. ISBN9781469816685. Archived from the original on 2017-07-28.
  32. ^Davis TM, Fortun P, Mulder J, Davis WA, Bruce DG (March 2004). 'Silent myocardial infarction and its prognosis in a community-based cohort of Type 2 diabetic patients: the Fremantle Diabetes Study'. Diabetologia. 47 (3): 395–399. doi:10.1007/s00125-004-1344-4. PMID14963648.
  33. ^Rubin, Emanuel; Gorstein, Fred; Rubin, Raphael; Schwarting, Roland; Strayer, David (2001). Rubin's Pathology — Clinicopathological Foundations of Medicine. Maryland: Lippincott Williams & Wilkins. p. 549. ISBN978-0-7817-4733-2.
  34. ^'Heart Attack Symptoms in Women'. www.heart.org. Retrieved 2018-11-07.
  35. ^'Signs of a heart attack in women'. womenshealth.gov. Retrieved 2018-11-07.
  36. ^ abcPerk J, De Backer G, Gohlke H, Graham I, Reiner Z, Verschuren M, Albus C, Benlian P, Boysen G, Cifkova R, Deaton C, Ebrahim S, Fisher M, Germano G, Hobbs R, Hoes A, Karadeniz S, Mezzani A, Prescott E, Ryden L, Scherer M, Syvänne M, Scholte op Reimer WJ, Vrints C, Wood D, Zamorano JL, Zannad F (July 2012). 'European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts)'. European Heart Journal. 33 (13): 1635–701. doi:10.1093/eurheartj/ehs092. PMID22555213.
  37. ^Smith SC, Allen J, Blair SN, Bonow RO, Brass LM, Fonarow GC, Grundy SM, Hiratzka L, Jones D, Krumholz HM, Mosca L, Pearson T, Pfeffer MA, Taubert KA (May 2006). 'AHA/ACC guidelines for secondary prevention for patients with coronary and other atherosclerotic vascular disease: 2006 update endorsed by the National Heart, Lung, and Blood Institute'. Journal of the American College of Cardiology. 47 (10): 2130–9. doi:10.1016/j.jacc.2006.04.026. PMID16697342.
  38. ^ abcKivimäki M, Nyberg ST, Batty GD, Fransson EI, Heikkilä K, Alfredsson L, Bjorner JB, Borritz M, Burr H, Casini A, Clays E, De Bacquer D, Dragano N, Ferrie JE, Geuskens GA, Goldberg M, Hamer M, Hooftman WE, Houtman IL, Joensuu M, Jokela M, Kittel F, Knutsson A, Koskenvuo M, Koskinen A, Kouvonen A, Kumari M, Madsen IE, Marmot MG, Nielsen ML, Nordin M, Oksanen T, Pentti J, Rugulies R, Salo P, Siegrist J, Singh-Manoux A, Suominen SB, Väänänen A, Vahtera J, Virtanen M, Westerholm PJ, Westerlund H, Zins M, Steptoe A, Theorell T (October 2012). 'Job strain as a risk factor for coronary heart disease: a collaborative meta-analysis of individual participant data'. Lancet. 380 (9852): 1491–7. doi:10.1016/S0140-6736(12)60994-5. PMC3486012. PMID22981903.
  39. ^Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT (July 2012). 'Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy'. Lancet. 380 (9838): 219–29. doi:10.1016/S0140-6736(12)61031-9. PMC3645500. PMID22818936.
  40. ^Steptoe A, Kivimäki M (April 2012). 'Stress and cardiovascular disease'. Nature Reviews. Cardiology. 9 (6): 360–70. doi:10.1038/nrcardio.2012.45. PMID22473079.
  41. ^ abHooper L, Martin N, Abdelhamid A, Davey Smith G (June 2015). 'Reduction in saturated fat intake for cardiovascular disease'. The Cochrane Database of Systematic Reviews. 6 (6): CD011737. doi:10.1002/14651858.CD011737. PMID26068959.
  42. ^ abcChowdhury R, Warnakula S, Kunutsor S, Crowe F, Ward HA, Johnson L, Franco OH, Butterworth AS, Forouhi NG, Thompson SG, Khaw KT, Mozaffarian D, Danesh J, Di Angelantonio E (March 2014). 'Association of dietary, circulating, and supplement fatty acids with coronary risk: a systematic review and meta-analysis'. Annals of Internal Medicine. 160 (6): 398–406. doi:10.7326/M13-1788. PMID24723079.
  43. ^de Souza RJ, Mente A, Maroleanu A, Cozma AI, Ha V, Kishibe T, Uleryk E, Budylowski P, Schünemann H, Beyene J, Anand SS (August 2015). 'Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: systematic review and meta-analysis of observational studies'. BMJ. 351: h3978. doi:10.1136/bmj.h3978. PMC4532752. PMID26268692.
  44. ^'Scientific Report of the 2015 Dietary Guidelines Advisory COmmittee'(PDF). health.gov. Feb 2015. p. 17. Archived(PDF) from the original on 2016-05-03.
  45. ^Krenz M, Korthuis RJ (January 2012). 'Moderate ethanol ingestion and cardiovascular protection: from epidemiologic associations to cellular mechanisms'. Journal of Molecular and Cellular Cardiology. 52 (1): 93–104. doi:10.1016/j.yjmcc.2011.10.011. PMC3246046. PMID22041278.
  46. ^ abcO'Donnell CJ, Nabel EG (December 2011). 'Genomics of cardiovascular disease'. The New England Journal of Medicine. 365 (22): 2098–109. doi:10.1056/NEJMra1105239. PMID22129254.
  47. ^Culić V (April 2007). 'Acute risk factors for myocardial infarction'. International Journal of Cardiology. 117 (2): 260–9. doi:10.1016/j.ijcard.2006.05.011. PMID16860887.
  48. ^Shaw E, Tofler GH (July 2009). 'Circadian rhythm and cardiovascular disease'. Current Atherosclerosis Reports. 11 (4): 289–95. doi:10.1007/s11883-009-0044-4. PMID19500492.
  49. ^Vyas MV, Garg AX, Iansavichus AV, Costella J, Donner A, Laugsand LE, Janszky I, Mrkobrada M, Parraga G, Hackam DG (July 2012). 'Shift work and vascular events: systematic review and meta-analysis'. BMJ. 345: e4800. doi:10.1136/bmj.e4800. PMC3406223. PMID22835925.
  50. ^Janszky, Imre; Ljung, Rickard (2008-10-30). 'Shifts to and from Daylight Saving Time and Incidence of Myocardial Infarction'. New England Journal of Medicine. 359 (18): 1966–1968. doi:10.1056/NEJMc0807104. ISSN0028-4793. PMID18971502.
  51. ^Roach RE, Helmerhorst FM, Lijfering WM, Stijnen T, Algra A, Dekkers OM (August 2015). 'Combined oral contraceptives: the risk of myocardial infarction and ischemic stroke'. The Cochrane Database of Systematic Reviews (8): CD011054. doi:10.1002/14651858.CD011054.pub2. PMC6494192. PMID26310586.
  52. ^Bally M, Dendukuri N, Rich B, Nadeau L, Helin-Salmivaara A, Garbe E, Brophy JM (May 2017). 'Risk of acute myocardial infarction with NSAIDs in real world use: bayesian meta-analysis of individual patient data'. BMJ. 357: j1909. doi:10.1136/bmj.j1909. PMC5423546. PMID28487435.
  53. ^Mu, Fan; Rich-Edwards, Janet; Rimm, Eric B.; Spiegelman, Donna; Missmer, Stacey A. (29 March 2016). 'Endometriosis and Risk of Coronary Heart Disease'. Circulation: Cardiovascular Quality and Outcomes. 9 (3): 257–264. doi:10.1161/CIRCOUTCOMES.115.002224. PMC4940126. PMID27025928.
  54. ^Mustafic H, Jabre P, Caussin C, Murad MH, Escolano S, Tafflet M, Périer MC, Marijon E, Vernerey D, Empana JP, Jouven X (February 2012). 'Main air pollutants and myocardial infarction: a systematic review and meta-analysis'. JAMA. 307 (7): 713–21. doi:10.1001/jama.2012.126. PMID22337682.
  55. ^Ho, Andrew Fu Wah; Wah, Win; Earnest, Arul; Ng, Yih Yng; Xie, Zhenjia; Shahidah, Nur; Yap, Susan; Pek, Pin Pin; Liu, Nan (2018-11-15). 'Health impacts of the Southeast Asian haze problem - A time-stratified case crossover study of the relationship between ambient air pollution and sudden cardiac deaths in Singapore'. International Journal of Cardiology. 271: 352–358. doi:10.1016/j.ijcard.2018.04.070. ISSN1874-1754. PMID30223374.
  56. ^Sun, Z; Chen, C; Xu, D; Li, T (October 2018). 'Effects of ambient temperature on myocardial infarction: A systematic review and meta-analysis'. Environmental Pollution (Barking, Essex : 1987). 241: 1106–1114. doi:10.1016/j.envpol.2018.06.045. PMID30029319.
  57. ^ abChatzidimitriou D, Kirmizis D, Gavriilaki E, Chatzidimitriou M, Malisiovas N (October 2012). 'Atherosclerosis and infection: is the jury still not in?'. Future Microbiology. 7 (10): 1217–30. doi:10.2217/fmb.12.87. PMID23030426.
  58. ^Charakida M, Tousoulis D (2013). 'Infections and atheromatous plaque: current therapeutic implications'. Current Pharmaceutical Design. 19 (9): 1638–50. doi:10.2174/138161213805219658. PMID23016720.
  59. ^Sánchez-Manubens J, Bou R, Anton J (February 2014). 'Diagnosis and classification of Kawasaki disease'. Journal of Autoimmunity. 48-49: 113–7. doi:10.1016/j.jaut.2014.01.010. PMID24485156.
  60. ^Hulten EA, Carbonaro S, Petrillo SP, Mitchell JD, Villines TC (March 2011). 'Prognostic value of cardiac computed tomography angiography: a systematic review and meta-analysis'. Journal of the American College of Cardiology. 57 (10): 1237–47. doi:10.1016/j.jacc.2010.10.011. PMID21145688.
  61. ^Clarke R, Halsey J, Bennett D, Lewington S (February 2011). 'Homocysteine and vascular disease: review of published results of the homocysteine-lowering trials'. Journal of Inherited Metabolic Disease. 34 (1): 83–91. doi:10.1007/s10545-010-9235-y. PMID21069462.
  62. ^Lonn E (September 2007). 'Homocysteine in the prevention of ischemic heart disease, stroke and venous thromboembolism: therapeutic target or just another distraction?'. Current Opinion in Hematology. 14 (5): 481–7. doi:10.1097/MOH.0b013e3282c48bd8. PMID17934354.
  63. ^Agewall S, Beltrame JF, Reynolds HR, Niessner A, Rosano G, Caforio AL, De Caterina R, Zimarino M, Roffi M, Kjeldsen K, Atar D, Kaski JC, Sechtem U, Tornvall P (January 2017). 'ESC working group position paper on myocardial infarction with non-obstructive coronary arteries'. European Heart Journal. 38 (3): 143–153. doi:10.1093/eurheartj/ehw149. PMID28158518.
  64. ^ abcdefghijklmnopqrstuvwxyzaaabacadaeafagahaiajReed, Grant W; Rossi, Jeffrey E; Cannon, Christopher P (January 2017). 'Acute myocardial infarction'. The Lancet. 389 (10065): 197–210. doi:10.1016/S0140-6736(16)30677-8. PMID27502078.
  65. ^ abcdefBritton, the editors Nicki R. Colledge, Brian R. Walker, Stuart H. Ralston; illustrated by Robert (2010). Davidson's principles and practice of medicine (21st ed.). Edinburgh: Churchill Livingstone/Elsevier. pp. 577–9. ISBN978-0-7020-3085-7.
  66. ^Woollard KJ, Geissmann F (February 2010). 'Monocytes in atherosclerosis: subsets and functions'. Nature Reviews. Cardiology. 7 (2): 77–86. doi:10.1038/nrcardio.2009.228. PMC2813241. PMID20065951.
  67. ^Janoudi A, Shamoun FE, Kalavakunta JK, Abela GS (July 2016). 'Cholesterol crystal induced arterial inflammation and destabilization of atherosclerotic plaque'. European Heart Journal. 37 (25): 1959–67. doi:10.1093/eurheartj/ehv653. PMID26705388.
  68. ^ abcBuja LM (July 2005). 'Myocardial ischemia and reperfusion injury'. Cardiovascular Pathology. 14 (4): 170–5. doi:10.1016/j.carpath.2005.03.006. PMID16009313.
  69. ^ abBolooki, H. Michael; Askari, Arman (August 2010). 'Acute Myocardial Infarction'. www.clevelandclinicmeded.com. Archived from the original on 28 April 2017. Retrieved 24 May 2017.
  70. ^ abConnolly, Philip I. Aaronson, Jeremy P.T. Ward, Michelle J. (2013). The cardiovascular system at a glance (4th ed.). Chichester, West Sussex: Wiley-Blackwell. pp. 88–89. ISBN978-0-470-65594-8.
  71. ^Kutty RS, Jones N, Moorjani N (November 2013). 'Mechanical complications of acute myocardial infarction'. Cardiology Clinics (Review). 31 (4): 519–31, vii–viii. doi:10.1016/j.ccl.2013.07.004. PMID24188218.
  72. ^Thygesen, Kristian; Alpert, Joseph S (25 August 2018). 'Fourth universal definition of myocardial infarction (2018)'. European Heart Journal. 40 (3): 237–269. doi:10.1093/eurheartj/ehy462. PMID30165617.
  73. ^Pickering JW, Than MP, Cullen L, Aldous S, Ter Avest E, Body R, Carlton EW, Collinson P, Dupuy AM, Ekelund U, Eggers KM, Florkowski CM, Freund Y, George P, Goodacre S, Greenslade JH, Jaffe AS, Lord SJ, Mokhtari A, Mueller C, Munro A, Mustapha S, Parsonage W, Peacock WF, Pemberton C, Richards AM, Sanchis J, Staub LP, Troughton R, Twerenbold R, Wildi K, Young J (May 2017). 'Rapid Rule-out of Acute Myocardial Infarction With a Single High-Sensitivity Cardiac Troponin T Measurement Below the Limit of Detection: A Collaborative Meta-analysis'. Annals of Internal Medicine. 166 (10): 715–724. doi:10.7326/M16-2562. PMID28418520.
  74. ^Chapman AR, Lee KK, McAllister DA, Cullen L, Greenslade JH, Parsonage W, et al. (November 2017). 'Association of High-Sensitivity Cardiac Troponin I Concentration With Cardiac Outcomes in Patients With Suspected Acute Coronary Syndrome'. JAMA. 318 (19): 1913–1924. doi:10.1001/jama.2017.17488. PMC5710293. PMID29127948.
  75. ^ abcdAmsterdam EA, Wenger NK, Brindis RG, Casey DE, Ganiats TG, Holmes DR, Jaffe AS, Jneid H, Kelly RF, Kontos MC, Levine GN, Liebson PR, Mukherjee D, Peterson ED, Sabatine MS, Smalling RW, Zieman SJ (December 2014). '2014 AHA/ACC guideline for the management of patients with non-ST-elevation acute coronary syndromes: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines'. Circulation. 130 (25): e344–426. doi:10.1161/CIR.0000000000000134. PMID25249585.
  76. ^Lipinski MJ, Escárcega RO, D'Ascenzo F, Magalhães MA, Baker NC, Torguson R, Chen F, Epstein SE, Miró O, Llorens P, Giannitsis E, Lotze U, Lefebvre S, Sebbane M, Cristol JP, Chenevier-Gobeaux C, Meune C, Eggers KM, Charpentier S, Twerenbold R, Mueller C, Biondi-Zoccai G, Waksman R (May 2014). 'A systematic review and collaborative meta-analysis to determine the incremental value of copeptin for rapid rule-out of acute myocardial infarction'. The American Journal of Cardiology. 113 (9): 1581–91. doi:10.1016/j.amjcard.2014.01.436. PMID24731654.
  77. ^ abcBritton, the editors Nicki R. Colledge, Brian R. Walker, Stuart H. Ralston; illustrated by Robert (2010). Davidson's principles and practice of medicine (21st ed.). Edinburgh: Churchill Livingstone/Elsevier. pp. 529–30. ISBN978-0-7020-3085-7.
  78. ^Kasper, Dennis L.; Fauci, Anthony S.; Hauser, Stephen L.; Longo, Dan L.; Jameson, J. Larry; Loscalzo, Joseph (2015). Harrison's principles of internal medicine. McGraw Hill Education. p. 1457. ISBN978-0-07-180215-4. OCLC923181481.
  79. ^ abcd'American College of Cardiology'. www.choosingwisely.org. Choosing Wisely. 28 February 2017. Archived from the original on 28 July 2017. Retrieved 24 May 2017.
  80. ^Schinkel AF, Valkema R, Geleijnse ML, Sijbrands EJ, Poldermans D (May 2010). 'Single-photon emission computed tomography for assessment of myocardial viability'. EuroIntervention. 6 (Supplement G): G115–22. PMID20542817.
  81. ^National Institute for Health and Clinical Excellence. Clinical guideline cg94: Unstable angina and NSTEMI. London, 2010.
  82. ^ ab'UOTW #36 - Ultrasound of the Week'. Ultrasound of the Week. 5 February 2015. Archived from the original on 9 May 2017. Retrieved 27 May 2017.
  83. ^ abcBritton, the editors Nicki R. Colledge, Brian R. Walker, Stuart H. Ralston; illustrated by Robert (2010). Davidson's principles and practice of medicine (21st ed.). Edinburgh: Churchill Livingstone/Elsevier. pp. 535, 539. ISBN978-0-7020-3085-7.
  84. ^Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD, Katus HA, Lindahl B, Morrow DA, Clemmensen PM, Johanson P, Hod H, Underwood R, Bax JJ, Bonow RO, Pinto F, Gibbons RJ, Fox KA, Atar D, Newby LK, Galvani M, Hamm CW, Uretsky BF, Steg PG, Wijns W, Bassand JP, Menasché P, Ravkilde J, Ohman EM, Antman EM, Wallentin LC, Armstrong PW, Simoons ML, Januzzi JL, Nieminen MS, Gheorghiade M, Filippatos G, Luepker RV, Fortmann SP, Rosamond WD, Levy D, Wood D, Smith SC, Hu D, Lopez-Sendon JL, Robertson RM, Weaver D, Tendera M, Bove AA, Parkhomenko AN, Vasilieva EJ, Mendis S (October 2012). 'Third universal definition of myocardial infarction'. Circulation. 126 (16): 2020–35. doi:10.1161/CIR.0b013e31826e1058. PMID22923432.
  85. ^Boie ET (November 2005). 'Initial evaluation of chest pain'. Emergency Medicine Clinics of North America. 23 (4): 937–57. doi:10.1016/j.emc.2005.07.007. PMID16199332.
  86. ^'Assessment of fatigue'. BMJ Best Practice. 17 August 2016. Retrieved 6 June 2017.
  87. ^Hess EP, Agarwal D, Chandra S, Murad MH, Erwin PJ, Hollander JE, Montori VM, Stiell IG (July 2010). 'Diagnostic accuracy of the TIMI risk score in patients with chest pain in the emergency department: a meta-analysis'. Canadian Medical Association Journal. 182 (10): 1039–44. doi:10.1503/cmaj.092119. PMC2900327. PMID20530163.
  88. ^Reeder, Guy (27 December 2016). 'Nitrates in the management of acute coronary syndrome'. www.uptodate.com. Archived from the original on 28 July 2017. Retrieved 24 May 2017.
  89. ^Yadlapati A, Gajjar M, Schimmel DR, Ricciardi MJ, Flaherty JD (December 2016). 'Contemporary management of ST-segment elevation myocardial infarction'. Internal and Emergency Medicine. 11 (8): 1107–1113. doi:10.1007/s11739-016-1550-3. PMID27714584.
  90. ^McCarthy CP, Mullins KV, Sidhu SS, Schulman SP, McEvoy JW (June 2016). 'The on- and off-target effects of morphine in acute coronary syndrome: A narrative review'. American Heart Journal. 176: 114–21. doi:10.1016/j.ahj.2016.04.004. PMID27264228.
  91. ^Bellemain-Appaix A, Kerneis M, O'Connor SA, Silvain J, Cucherat M, Beygui F, Barthélémy O, Collet JP, Jacq L, Bernasconi F, Montalescot G (October 2014). 'Reappraisal of thienopyridine pretreatment in patients with non-ST elevation acute coronary syndrome: a systematic review and meta-analysis'. BMJ. 349: g6269. doi:10.1136/bmj.g6269. PMC4208629. PMID25954988.
  92. ^ abAndrade-Castellanos CA, Colunga-Lozano LE, Delgado-Figueroa N, Magee K (June 2014). 'Heparin versus placebo for non-ST elevation acute coronary syndromes'. The Cochrane Database of Systematic Reviews. 6 (6): CD003462. doi:10.1002/14651858.CD003462.pub3. PMID24972265.
  93. ^Bagai A, Dangas GD, Stone GW, Granger CB (June 2014). 'Reperfusion strategies in acute coronary syndromes'. Circulation Research. 114 (12): 1918–28. doi:10.1161/CIRCRESAHA.114.302744. PMID24902975.
  94. ^Jobs A, Mehta SR, Montalescot G, Vicaut E, van't Hof AW, Badings EA, Neumann FJ, Kastrati A, Sciahbasi A, Reuter PG, Lapostolle F, Milosevic A, Stankovic G, Milasinovic D, Vonthein R, Desch S, Thiele H (August 2017). 'Optimal timing of an invasive strategy in patients with non-ST-elevation acute coronary syndrome: a meta-analysis of randomised trials'. Lancet. 390 (10096): 737–746. doi:10.1016/S0140-6736(17)31490-3. PMID28778541.
  95. ^Wijns W, Kolh P, Danchin N, Di Mario C, Falk V, Folliguet T, Garg S, Huber K, James S, Knuuti J, Lopez-Sendon J, Marco J, Menicanti L, Ostojic M, Piepoli MF, Pirlet C, Pomar JL, Reifart N, Ribichini FL, Schalij MJ, Sergeant P, Serruys PW, Silber S, Sousa Uva M, Taggart D (October 2010). 'Guidelines on myocardial revascularization'. European Heart Journal. 31 (20): 2501–55. doi:10.1093/eurheartj/ehq277. PMID20802248.
  96. ^Dalal F, Dalal HM, Voukalis C, Gandhi MM (July 2017). 'Management of patients after primary percutaneous coronary intervention for myocardial infarction'. BMJ. 358: j3237. doi:10.1136/bmj.j3237. PMID28729460.
  97. ^Lassen JF, Bøtker HE, Terkelsen CJ (January 2013). 'Timely and optimal treatment of patients with STEMI'. Nature Reviews. Cardiology. 1. 10 (1): 41–8. doi:10.1038/nrcardio.2012.156. PMID23165072.
  98. ^Neumar RW, Shuster M, Callaway CW, Gent LM, Atkins DL, Bhanji F, Brooks SC, de Caen AR, Donnino MW, Ferrer JM, Kleinman ME, Kronick SL, Lavonas EJ, Link MS, Mancini ME, Morrison LJ, O'Connor RE, Samson RA, Schexnayder SM, Singletary EM, Sinz EH, Travers AH, Wyckoff MH, Hazinski MF (November 2015). 'Part 1: Executive Summary: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care'. Circulation. 132 (18 Suppl 2): S315–67. doi:10.1161/cir.0000000000000252. PMID26472989.
  99. ^McCaul M, Lourens A, Kredo T (September 2014). 'Pre-hospital versus in-hospital thrombolysis for ST-elevation myocardial infarction'. The Cochrane Database of Systematic Reviews. 9 (9): CD010191. doi:10.1002/14651858.CD010191.pub2. PMID25208209.
  100. ^Cabello JB, Burls A, Emparanza JI, Bayliss SE, Quinn T (December 2016). 'Oxygen therapy for acute myocardial infarction'. The Cochrane Database of Systematic Reviews. 12: CD007160. doi:10.1002/14651858.CD007160.pub4. PMC6463792. PMID27991651.
  101. ^Hofmann R, James SK, Jernberg T, Lindahl B, Erlinge D, Witt N, Arefalk G, Frick M, Alfredsson J, Nilsson L, Ravn-Fischer A, Omerovic E, Kellerth T, Sparv D, Ekelund U, Linder R, Ekström M, Lauermann J, Haaga U, Pernow J, Östlund O, Herlitz J, Svensson L (September 2017). 'Oxygen Therapy in Suspected Acute Myocardial Infarction'. The New England Journal of Medicine. 377 (13): 1240–1249. doi:10.1056/nejmoa1706222. PMID28844200.
  102. ^Abuzaid A, Fabrizio C, Felpel K, Al Ashry HS, Ranjan P, Elbadawi A, Mohamed AH, Barssoum K, Elgendy IY (March 2018). 'Oxygen Therapy in Patients with Acute Myocardial Infarction: A Systemic Review and Meta-Analysis'. The American Journal of Medicine. 131 (6): 693–701. doi:10.1016/j.amjmed.2017.12.027. PMID29355510.
  103. ^Sepehrvand, N; James, SK; Stub, D; Khoshnood, A; Ezekowitz, JA; Hofmann, R (29 March 2018). 'Effects of supplemental oxygen therapy in patients with suspected acute myocardial infarction: a meta-analysis of randomised clinical trials'. Heart. 104 (20): heartjnl–2018–313089. doi:10.1136/heartjnl-2018-313089. PMID29599378.
  104. ^Ardehali R, Perez M, Wang P (2011). A practical approach to cardiovascular medicine. Chichester, West Sussex, UK: Wiley-Blackwell. p. 57. ISBN978-1-4443-9387-3.
  105. ^Jindal SK, ed. (2011). Textbook of pulmonary and critical care medicine. New Delhi: Jaypee Brothers Medical Publishers. p. 1758. ISBN978-93-5025-073-0.
  106. ^Dahal, K; Hendrani, A; Sharma, SP; Singireddy, S; Mina, G; Reddy, P; Dominic, P; Modi, K (1 July 2018). 'Aldosterone Antagonist Therapy and Mortality in Patients With ST-Segment Elevation Myocardial Infarction Without Heart Failure: A Systematic Review and Meta-analysis'. JAMA Internal Medicine. 178 (7): 913–920. doi:10.1001/jamainternmed.2018.0850. PMC6145720. PMID29799995.
  107. ^ abcdefghNational Institute for Health and Clinical Excellence. Clinical guideline 172: Secondary prevention in primary and secondary care for patients following a myocardial infarction. London, 2013.
  108. ^ abcdeNational Institute for Health and Clinical Excellence. Clinical guideline 181: Lipid modification: cardiovascular risk assessment and the modification of blood lipids for the primary and secondary prevention of cardiovascular disease. London, 2014.
  109. ^Stradling C, Hamid M, Taheri S, Thomas GN (2014). 'A review of dietary influences on cardiovascular health: part 2: dietary patterns'. Cardiovascular & Hematological Disorders Drug Targets. 14 (1): 50–63. doi:10.2174/1871529x14666140701095426. PMID24993125.
  110. ^Fortmann SP, Burda BU, Senger CA, Lin JS, Whitlock EP (December 2013). 'Vitamin and mineral supplements in the primary prevention of cardiovascular disease and cancer: An updated systematic evidence review for the U.S. Preventive Services Task Force'. Annals of Internal Medicine. 159 (12): 824–34. doi:10.7326/0003-4819-159-12-201312170-00729. PMID24217421.
  111. ^McPherson K, et al. (June 2010). 'Prevention of cardiovascular disease – NICE public health guidance 25'. London: National Institute for Health and Care Excellence. Archived from the original on 2014-03-29.
  112. ^Ebrahim, Shah; Taylor, Fiona; Ward, Kirsten; Beswick, Andrew; Burke, Margaret; Davey Smith, George (2011-01-19). 'Multiple risk factor interventions for primary prevention of coronary heart disease'. Cochrane Database of Systematic Reviews (1): CD001561. doi:10.1002/14651858.cd001561.pub3. PMC4160097. PMID21249647.
  113. ^Taylor F, Huffman MD, Macedo AF, Moore TH, Burke M, Davey Smith G, Ward K, Ebrahim S (January 2013). 'Statins for the primary prevention of cardiovascular disease'. The Cochrane Database of Systematic Reviews. 1 (1): CD004816. doi:10.1002/14651858.CD004816.pub5. PMC4164175. PMID23440795.
  114. ^Baigent C, Blackwell L, Collins R, Emberson J, Godwin J, Peto R, Buring J, Hennekens C, Kearney P, Meade T, Patrono C, Roncaglioni MC, Zanchetti A (May 2009). 'Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials'. Lancet. 373 (9678): 1849–60. doi:10.1016/S0140-6736(09)60503-1. PMC2715005. PMID19482214.
  115. ^Sutcliffe P, Connock M, Gurung T, Freeman K, Johnson S, Kandala NB, Grove A, Gurung B, Morrow S, Clarke A (September 2013). 'Aspirin for prophylactic use in the primary prevention of cardiovascular disease and cancer: a systematic review and overview of reviews'. Health Technology Assessment. 17 (43): 1–253. doi:10.3310/hta17430. PMC4781046. PMID24074752.
  116. ^Matthys F, De Backer T, De Backer G, Stichele RV (March 2014). 'Review of guidelines on primary prevention of cardiovascular disease with aspirin: how much evidence is needed to turn a tanker?'. European Journal of Preventive Cardiology. 21 (3): 354–65. doi:10.1177/2047487312472077. PMID23610452.
  117. ^Hodis HN, Mack WJ (July 2014). 'Hormone replacement therapy and the association with coronary heart disease and overall mortality: clinical application of the timing hypothesis'. The Journal of Steroid Biochemistry and Molecular Biology. 142: 68–75. doi:10.1016/j.jsbmb.2013.06.011. PMID23851166.
  118. ^Anderson L, Taylor RS (December 2014). 'Cardiac rehabilitation for people with heart disease: an overview of Cochrane systematic reviews'. The Cochrane Database of Systematic Reviews. 12 (12): CD011273. doi:10.1002/14651858.CD011273.pub2. hdl:10871/19152. PMID25503364.
  119. ^Perez MI, Musini VM, Wright JM (October 2009). 'Effect of early treatment with anti-hypertensive drugs on short and long-term mortality in patients with an acute cardiovascular event'. The Cochrane Database of Systematic Reviews (4): CD006743. doi:10.1002/14651858.CD006743.pub2. PMID19821384.
  120. ^Elmariah, Sammy; Mauri, Laura; Doros, Gheorghe; Galper, Benjamin Z; O'Neill, Kelly E; Steg, Philippe Gabriel; Kereiakes, Dean J; Yeh, Robert W (November 2014). 'Extended duration dual antiplatelet therapy and mortality: a systematic review and meta-analysis'. The Lancet. 385 (9970): 792–798. doi:10.1016/S0140-6736(14)62052-3. PMC4386690. PMID25467565.
  121. ^Bangalore S, Makani H, Radford M, Thakur K, Toklu B, Katz SD, DiNicolantonio JJ, Devereaux PJ, Alexander KP, Wetterslev J, Messerli FH (October 2014). 'Clinical outcomes with β-blockers for myocardial infarction: a meta-analysis of randomized trials'. The American Journal of Medicine. 127 (10): 939–53. doi:10.1016/j.amjmed.2014.05.032. PMID24927909.
  122. ^Newman, David (19 August 2010). 'Beta Blockers for Acute Heart Attack (Myocardial Infarction)'. TheNNT.com. Archived from the original on 22 December 2015. Retrieved 11 December 2015.
  123. ^Le HH, El-Khatib C, Mombled M, Guitarian F, Al-Gobari M, Fall M, Janiaud P, Marchant I, Cucherat M, Bejan-Angoulvant T, Gueyffier F (2016). 'Impact of Aldosterone Antagonists on Sudden Cardiac Death Prevention in Heart Failure and Post-Myocardial Infarction Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials'. PLoS ONE. 11 (2): e0145958. Bibcode:2016PLoSO..1145958L. doi:10.1371/journal.pone.0145958. PMC4758660. PMID26891235.
  124. ^Hazinski MF, Nolan JP, Aickin R, Bhanji F, Billi JE, Callaway CW, Castren M, de Caen AR, Ferrer JM, Finn JC, Gent LM, Griffin RE, Iverson S, Lang E, Lim SH, Maconochie IK, Montgomery WH, Morley PT, Nadkarni VM, Neumar RW, Nikolaou NI, Perkins GD, Perlman JM, Singletary EM, Soar J, Travers AH, Welsford M, Wyllie J, Zideman DA (October 2015). 'Part 1: Executive Summary: 2015 International Consensus on Cardiopulmonary Resuscitation and Emergency Cardiovascular Care Science With Treatment Recommendations'. Circulation (Review). 132 (16 Suppl 1): S2–39. doi:10.1161/CIR.0000000000000270. PMID26472854.
  125. ^ abPapneja, Koyelle; Chan, Anthony K.; Mondal, Tapas K.; Paes, Bosco (March 2017). 'Myocardial Infarction in Neonates: A Review of an Entity with Significant Morbidity and Mortality'. Pediatric Cardiology. 38 (3): 427–441. doi:10.1007/s00246-016-1556-7. ISSN1432-1971. PMID28238152.
  126. ^López de Sá E, López-Sendón J, Anguera I, Bethencourt A, Bosch X (November 2002). 'Prognostic value of clinical variables at presentation in patients with non-ST-segment elevation acute coronary syndromes: results of the Proyecto de Estudio del Pronóstico de la Angina (PEPA)'. Medicine. 81 (6): 434–42. doi:10.1097/00005792-200211000-00004. PMID12441900.
  127. ^Fox KA, Dabbous OH, Goldberg RJ, Pieper KS, Eagle KA, Van de Werf F, Avezum A, Goodman SG, Flather MD, Anderson FA, Granger CB (November 2006). 'Prediction of risk of death and myocardial infarction in the six months after presentation with acute coronary syndrome: prospective multinational observational study (GRACE)'. BMJ. 333 (7578): 1091. doi:10.1136/bmj.38985.646481.55. PMC1661748. PMID17032691. Archived from the original on 2009-03-09.
  128. ^Weir RA, McMurray JJ, Velazquez EJ (May 2006). 'Epidemiology of heart failure and left ventricular systolic dysfunction after acute myocardial infarction: prevalence, clinical characteristics, and prognostic importance'. The American Journal of Cardiology. 97 (10A): 13F–25F. doi:10.1016/j.amjcard.2006.03.005. PMID16698331.
  129. ^ abcWorld Health Organization (2008). The Global Burden of Disease: 2004 Update. Geneva: World Health Organization. ISBN978-92-4-156371-0.
  130. ^American Heart Association Statistics Committee and Stroke Statistics, Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, et al. (January 2012). 'Executive summary: heart disease and stroke statistics--2012 update: a report from the American Heart Association'. Circulation. 125 (1): 188–97. doi:10.1161/CIR.0b013e3182456d46. PMID22215894.
  131. ^Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, et al. (January 2015). 'Heart disease and stroke statistics--2015 update: a report from the American Heart Association'. Circulation. 131 (4): e29–322. doi:10.1161/cir.0000000000000152. PMID25520374. From 2001 to 2011, death rates attributable to CVD declined 30.8%.
  132. ^Gupta R, Joshi P, Mohan V, Reddy KS, Yusuf S (January 2008). 'Epidemiology and causation of coronary heart disease and stroke in India'. Heart. 94 (1): 16–26. doi:10.1136/hrt.2007.132951. PMID18083949.
  133. ^Perry K, Petrie KJ, Ellis CJ, Horne R, Moss-Morris R (July 2001). 'Symptom expectations and delay in acute myocardial infarction patients'. Heart. 86 (1): 91–3. doi:10.1136/heart.86.1.91. PMC1729795. PMID11410572.
  134. ^Workers' Compensation FAQArchived 2007-07-11 at the Wayback Machine. Prairie View A&M University. Retrieved November 22, 2006.
  135. ^SIGNIFICANT DECISIONS Subject IndexArchived 2006-12-06 at the Wayback Machine. Board of Industrial Insurance Appeals. Retrieved November 22, 2006.
  136. ^'Classification of Drivers' Licenses Regulations'. Nova Scotia Registry of Regulations. May 24, 2000. Archived from the original on April 20, 2007. Retrieved April 22, 2007.

Further reading[edit]

  • Levine GN, Bates ER, Blankenship JC, Bailey SR, Bittl JA, Cercek B, et al. (March 2016). '2015 ACC/AHA/SCAI Focused Update on Primary Percutaneous Coronary Intervention for Patients With ST-Elevation Myocardial Infarction: An Update of the 2011 ACCF/AHA/SCAI Guideline for Percutaneous Coronary Intervention and the 2013 ACCF/AHA Guideline for the Management of ST-Elevation Myocardial Infarction: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Society for Cardiovascular Angiography and Interventions'. Circulation. 133 (11): 1135–47. doi:10.1161/CIR.0000000000000336. PMID26490017.

External links[edit]

Classification
  • ICD-10: I21-I22
  • ICD-9-CM: 410
  • MeSH: D009203
  • DiseasesDB: 8664
External resources
  • MedlinePlus: 000195
  • eMedicine: med/1567emerg/327ped/2520
  • Patient UK:Myocardial infarction
  • Myocardial infarction at Curlie
  • American Heart Association's Heart Attack web site — Information and resources for preventing, recognizing and treating a heart attack.
  • TIMI Score for UA/NSTEMI and STEMI
Retrieved from 'https://en.wikipedia.org/w/index.php?title=Myocardial_infarction&oldid=911038565'
Circ Res. Author manuscript; available in PMC 2013 Mar 12.
Published in final edited form as:
doi: 10.1161/CIRCRESAHA.111.252981
NIHMSID: NIHMS361108
The publisher's final edited version of this article is available at Circ Res
See other articles in PMC that cite the published article.

Abstract

Congestive heart failure accounts for half a million deaths per year in the US. Despite its place among the leading causes of morbidity, pharmcalogical and mechanic remedies have been able to slow the progression of the disease, today’s science has yet to provide a cure and there are few therapeutic modalities available for patients with advanced heart failure. There is a critical need to explore new therapeutic approaches in heart failure and gene therapy has emerged as a viable alternative. Recent advances in understanding of the molecular basis of myocardial dysfunction, together with the evolution of increasingly efficient gene transfer technology, has placed heart failure within reach of gene-based therapy. The recent successful and safe completion of a phase 2 trial targeting the sarcoplasmic reticulum calcium ATPase pump (SERCA2a) along with the start of more recent phase 1 trials opens a new era for gene therapy for the treatment of heart failure.

Keywords: Gene therapy, heart failure, adeno-associated vectors

Introduction

Ongoing preclinical studies are providing a sound scientific basis for the evaluation of gene therapy strategies. Furthermore, the anatomical compartmentalization of the heart and its accessibility by surgical and percutaneous approaches, render the myocardium a highly amenable target system for gene therapy.

Over the last decade, novel molecular mechanisms associated with heart failure have been discovered creating new targets for therapeutic interventions. A number of these promising targets cannot be manipulated through pharmacological means but are ideal for gene therapy approaches.

In this review, we will highlight new strategies for the treatment of heart failure by gene transfer, focusing on the vectors, targets, and delivery methods along with the recent clinical results from early clinical trials.

Gene Delivery

Gene delivery vehicles fall into one of two categories: non-viral or viral gene delivery vectors. The most attractive features of non-viral gene delivery vectors are their safety profile and minimal immunogenicity. The size of the transgene in viruses is limited by the packaging constraints while with plasmid there are no packaging limitations that restricts the transgene size. However, despite extensive efforts to improve transfection efficiencies by combining “naked” DNA with delivery agents such as cationic lipids or by using plasmid DNA in combination with physical methods to enhance gene transfer, insufficient transfection efficiencies remain the Achilles heel of non-viral gene delivery methods.

Given these limitations of non-viral delivery vehicles, intense research has taken place over the past years aimed at harnessing the inherent capacity of viruses to deliver genes to cells. Today, the virus most widely used for cardiovascular gene transfer is adenovirus with a total of 54 trials, 24 of which are ongoing. Other viral vectors that have reached the clinical trial phase are derived from retroviruses, Sendai virus and adeno-associated virus. Given the non-proliferative nature of cardiomyocytes, lentiviruses could be promising vehicles for cardiovascular gene therapy. In contrast to AAV, lenti- and retroviral vectors integrate their genomes into the host genome. Whereas pseudo-random vector genome integration raises the risk of oncogenic transformation, genome integration is essential for stem cell therapy because stem cell therapy, by its very nature, requires an expansion of the stem cells to be effective. This expansion requires multiple rounds of cell division, during which the vector DNA of non-integrating vectors will be lost due to “dilution” of the vector genomes. In regards, to the potential for oncogenic transformation by γ-retro- and lentiviral vectors, it should be pointed that while both γ–retroviral vectors and lentiviral vectors preferentially integrate their genome into transcriptional units, lentiviral vector genome integration is, in contrast to γ-retroviral genome integration, usually more distant from transcription start sites. Together with the development of self-inactivating (SIN) lentiviral vectors, the resulting improved safety profile likely contributed to the approval of clinical trials using lentiviral vectors.

As can be seen from Table 1, each of the vector systems has its unique properties and vector specific advantages and limitations. One of the potential limitations of adenoviral vectors is that they can only trigger short term gene expression, although it can be argued that for certain applications—e.g., the expression of factors that stimulate cell division—this might be advantageous. The far bigger problem of adenoviral vectors is that they are highly immunogenic. While in cardiovascular gene therapy trials no adverse effect as devastating as the tragic death of Jesse Gelsinger in a trial for the treatment of ornithine transcarbamylase deficiency with an adenoviral vector encoding the deficient enzyme has been observed, the inflammation caused by adenoviral vectors is of significant concern and probably one of the main reasons for the declining the use of this vector system in clinical gene transfer for cardiovascular diseases.

Table 1

Vectors Systems Used in Cardiovascular Gene Transfer

VectorGenomePersistencyPackaging
Capacity
DividingNon-
dividing
AdvantagesDisadvantagesOpen Trials
a
Total
Trials
a
NonviralDNAVariablen/ayesYesLow cost,
nonpathogenic,
limited immune
response
Low transfection
efficiency, no
cardiotropism
4084
AdDNAnoUp to 30kbYesYesHigh
transduction
efficiency, fast
expression
kinetics,
efficient
cardiomyocyte
transduction
Immunogenicity,
transient
expression, no
cardiotropism
2454
AAVDNAyes~5kbYesYesNon-
pathogenic,
efficient
cardiomyocyte
transduction,
persistent gene
expression, low
immunogenicity
Limited
packaging
capacity
33
RVRNAyes8kbYesNoHigh
transduction
efficiencies.
Stable
transduction.
Risk of
oncogenic
transformation,
transduces only
dividing cells, no
cardiotropism
33
LVRNAyes8kbYesYesHigh
transduction
efficiencies.
Stable
transduction.
Risk of
oncogenic
transformation,
potential CTL
response, no
cardiotropism
00
SendaiRNAno15YesYesNon-pathogenic
in humans, no
obvious path
for genome
integration
Transient
expression, no
cardiotropism
22

NV: non-viral; Ad: adenovirus; AAV: adeno-associated virus; RV: retrovirus; LV: lentivirus; CTL: cytotoxic T lymphocyte. a as of June 2011

AAV Vectors in Cardiovascular Gene Transfer

AAV is a small, non-enveloped virus with a linear, single-stranded DNA that belongs to the family Parvoviridae, the subfamily Parvovirinae and the genus Dependovirus. As its name indicates, AAV depends on a helper virus such as adenovirus or herpes virus for productive replication. Contributing to its beneficial safety profile, AAV is not known to be associated with any human disease. Furthermore, the only cis-elements required for AAV vectors are the inverted terminal repeats (ITRs) that flank the (recombinant) viral genome. As a consequence of this, transduction with AAV vectors does not result in the expression of any viral genes, which likely contributes to their low immunogenicity when compared to most other viral vectors (see below).

As for all viruses, infection/transduction starts with the binding of the virus to its receptor(s) (Fig. 1). The receptors for some of the AAV serotypes have been described in the literature (see for a description of the AAV receptors) Receptor-binding is then followed by endocytosis, which for AAV2 is via the so-called CLIC/GEEC (Clathrin Independent Carriers/GPI-anchored protein Enriched Endocytic Compartment) pathway whereas for AAV5 it has been reported that endocytosis can occur both by clathrin coated vesicles or caveolae.

AAV entry

1: receptor binding & endocytosis; 2: escape into cytoplasm; 3: nuclear import; 4: capsid disassembly; 5: double strand synthesis

At least for the prototypical AAV serotype, AAV2, and AAV5 endocytosis is followed by transport to the Golgi (Fig. 1). Evidence suggests that once in the cytoplasm AAV is imported into the nucleus as an intact particle, presumably through the nuclear pore (Fig. 1). Import of viral particle is then followed by capsid uncoating and the release of the viral genome (Fig. 1). For transcription of the transgene, the single-stranded AAV genome must then first be converted into a double strand genome (Fig. 1).

Tropism of AAV Serotypes

To date, 13 AAV serotypes and more than 100 AAV variants have been described in the literature, although most gene transfer experiments are performed with vectors based on AAV1-9. Together, the AAV serotypes display broad, yet serotype-specific tissue and cell tropism (Table 2), which is likely, at least to a certain extent, a result of their diverse receptors (for a compilation see). Nonetheless—species differences notwithstanding—overall, AAV1, AAV6, AAV8 and AAV9 show strong cardiac transduction, with AAV9 appearing to be the most cardiotropic, at least in rodents. In light of the observed species differences it is unfortunate that our knowledge of the cardiotropism of the AAV serotypes in non-human primates is very limited, although it has been shown that AAV2 is able to transduce cardiomyocytes after intramyocardial injection and in Rhesus Macaques AAV6 is superior to AAV8 and AAV9 in transducing cardiac tissue when delivered by percutaneous, transendocardial injection. Importantly, the results from the recently completed phase II of the so-called CUPID (Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease) indicate that AAV1 can successfully transduce human cardiac tissue.

Table 2

Tropism of AAV Serotypes in Animal Models of Cardiac Diseases

AnimalTissueSerotype Sequence
Mous
e
Heartiv: AAV9>AAV6>AAV8>AAV4>AAV7>AAV1>AAV3>AAV2132
iv: AAV8>AAV1>AAV2133
iv: AAV9>AAV8>AAV1134
iv: AAV9>>AAV8135
iv: AAV9>AAV6136
iv: AAV1≈AAV4≈AAV5≈AAV6>AAV219
ic: AAV6>AAV9>AAV2>AAV1>AAV4>AAV8>AAV7>AAV3>AAV5137
imc: AAV1>AAV2>AAV3≈AAV4≈AAV5138
ipc: AAV9>AAV1≈AAV7>AAV6≈AAV877
ip: AAV8≈AAV7>AAV1≈AAV6>AAV2≈AAV5133
Liveriv: AAV9≈AAV7>AAV6>AAV2>AAV5>AAV1>AAV4132
iv: AAV9>AAV8135
iv: AAV9>AAV615
iv: AAV1≈AAV4≈AAV5≈AAV6>AAV219
ic: AAV7>AAV9>AAV8>AAV2>AAV6>AAV1>AAV4>AAV3>AAV5137
ipc: AAV8>AAV7≈AAV9>AAV1≈AAV677
Skelet
al Muscle
iv: AAV6≈AAV7≈AAV8≈AAV9>AAV3≈AAV1>AAV2≈AAV4≈AAV5132
iv: AAV8>AAV1>AAV2133
iv: AAV9>AAV8134
iv: AAV9>AAV8135
iv: AAV5≈AAV6>AAV1≈AAV4 (AAV2 no expression)19
ic: AAV4≈AAV7≈AAV8>AAV1≈AAV3≈AAV5≈AAV6>AAV2>AAV9137
ip: AAV1≈AAV6>AAV8≈AAV7>AAV2≈AAV5133
RatHeartiv: AAV8>AAV1>AAV6>AAV7>AAV4≈AAV5 (AAV2, AAV3 no expression)139
imc: AAV9>AAV8>AAV7>AAV177
Cani
ne
Heartimc: AAV6>>AAV9>AAV877
AAV9:140
Liverimc: AAV9>AAV8>AAV677
cv: AAV8>>AAV2141
ipv: AAV8>AAV2133
AAV2:142; AAV8:133, 141,
Skelet
al Muscle
imc: AAV6>AAV9>AAV877
AAV2:143-145; AAV6:143, 145-147; AAV8:148; AAV9:149
Swin
e
Heartimc: AAV1>AAV2150
AAV1:104, 111, 151; AAV2:152, 153

Transductional Targeting

Despite the substantial cardiotropism of AAV1, AAV6 and AAV9, all of these serotypes transduce additional tissues, at least to some extent. It is, therefore, not surprising that substantial efforts have been made to develop AAV variants with further enhanced cardiac tropism.

One general approach to alter AAV tropism, including the isolation of AAV variants with cardiotropism, that recently has gained considerable attention is so-called directed evolution (Figure 2). All studies using this method are based on the initial creation of AAV libraries with diverse AAV capsids. One method uses the insertion of a (semi)-random peptide sequence into the AAV capsid. A variation of this method replaces a short segment of an AAV capsid protein, usually located in the major capsid protein VP3, with a (semi)random peptide sequence.

Generation of mutant AAV Library and Directed Evolution to Identify Cardiotropic AAVs. A) Creation of a library of AAVs through DNA shuffling. B) Selection of cardiotropic AAVs through directed evolution.

In a different approach to create AAV libraries composed of viruses with diverse capsids, viruses with chimeric capsid proteins, i.e. capsid proteins with stretches of amino acids from different serotypes within the same capsid protein, are generated. The method for the generation of these so-called shuffled capsid libraries is outlined in Fig. 2A.

Regardless on how the AAV libraries are generated, in the next step the most cardiotropic variants are then selected by either in vitro or in vivo evolution. Whereas in vitro selection might be technically simpler, it fails to select for variants that can efficiently overcome barriers, such as escape from the vasculature, that virions only encounter in an in vivo setting. A typical in vivo selection procedure is shown schematically in Fig. 2B. Briefly, the viral library is injected into the tail vein of mice, 24-72 hours later the heart is harvested and the capsid DNA amplified by PCR. After subcloning into a wild-type AAV backbone, a secondary viral library is created and the selection procedure repeated. After one or more additional rounds of selection, individual clones are isolated and the capsid genes sequenced. Recombinant viruses encoding reporter genes are then produced and used for a detailed characterization of the new cardiotropic AAV variants. While the method just described resulted in the successful identification of AAV variants with enhanced cardiac tropism, it enriches AAV variants that successfully deliver their DNA to the heart, regardless whether they successfully infect cardiac tissue. To overcome this potential limitation, Kleinschmidt and colleagues, after the harvesting of the heart, kept heart slices in organotypic culture and super-infected these slices with adenovirus. This should allow the replication and enrichment of AAV variants that successfully infected cardiac tissue. AAV variants that, for instance, only delivered their genomes to the cytoplasm of cardiomyocytes, on the other hand, will not be enriched.

Another successful way to enhance cardiac tropism has been used by Samulski and colleagues . This group replaced a hexapeptide sequence in the receptor-binding region of the AAV2 capsid with the corresponding peptides of other AAV serotypes and variants. One of the novel AAV2 variants, AAV2i8, which is a chimera between the AAV2 capsid and the AAV8 hexapeptide, showed significant skeletal and cardiac muscle tropism with significantly reduced transduction efficiencies of the liver. Clearly, these novel AAV variants are promising candidates for cardiac gene transfer. It has to be pointed out, however, that in cases where transduction of cardiomyocytes were compared to transduction by AAV9 the latter always showed the highest transduction efficiency.

Transcriptional Targeting

Ideally, transductional targeting alone is sufficient to drive the expression of the transgene exclusively in the heart. Unfortunately, however, AAV vectors that do not transduce non-cardiac tissues to some extent are thus far unavailable. An alternative and complementary approach to restrict transgene expression to the heart is transcriptional targeting, i.e. the use of cardiac specific promoters. An additional advantage of cardiac specific promoters is that, in contrast to the most commonly used promoter, the CMV promoter, their expression is not expected to be downregulated and, hence, should result in long-term gene expression.

One promoter that was used in both adenoviral and AAV vectors is the ventricle-specific Myosin Light Chain-2 (MLC-2v) promoter. While this promoter is more than 10-fold less efficient than the already weak RSV promoter, this inadequacy can be partially over come by the inclusion of 4 copies of the 250bp enhancer fragment of the MLC-v2 gene. This promoter construct drives expression that is only 3.8-fold lower than expression from the strong CMV promoter. For AAV vectors this comes, however, at the price of reducing the allowable size of the transgene by 1,000bp, which is not insignificant in a vector system that has a packaging capacity of ~5kb. The Myosin Heavy Chain Promoter, which has been used extensively in the generation of transgenic mice that express transgenes specifically in cardiomyocytes, is not suitable for AAV vectors because it is 5.5kb long. Unfortunately, expression from a minimal version of the same promoter is much lower than expression from RSV or CMV promoters. Maybe the most promising promoter for cardiac-specific expression that is suitable for AAV vectors is a 418 bp fragment from the chicken cardiac Troponin T (cTnT) promoter. The strength of this promoter is only approximately 2-fold lower when compared to the CMV promoter.

Regulation of Transgene Expression

Both the viral and heart-specific promoters mentioned in the previous section are constitutively active, i.e. the expression levels of the transgene cannot be regulated. For certain applications, however, it is important that the transgene is either expressed only under certain physiological conditions or that expression levels or the time-frame of expression can be regulated pharmacologically. The most commonly systems that allow the manipulation of expression levels with small molecules are the so-called Tet-on and Tet-off and the rapamycin/FK506 inducible systems. Both these systems as well as a mifepristone (RU486)-inducible system have been used in context of AAV vectors.

In addition it can be useful to express the transgene under specific pathologic conditions. One system that allows the induction of transgene expression under hypoxic conditions is based on the inclusion of a hypoxia response element into the promoter region. Hypoxia inducible system have been based on the oxygen-dependent degradation (ODD) domain of hypoxia inducible factor 1α (HIF1α).

Gene Silencing with shRNAs and Artificial MicroRNAs

For many disorders, including cardiac diseases, it is necessary to overexpress a protein because the endogenous protein is either non-functional or expressed at insufficient levels. It is equally possible, however, that a disorder is caused by a mutated protein that has dominant effects or that an endogenous protein is expressed at pathologically high levels. Furthermore, in certain instances, it is therapeutically beneficial to suppress protein expression of protein that is expressed at physiological levels. A little more than a decade ago, Mello and colleagues made the seminal discovery that in Caenorhabditis elegans double-stranded RNA could prompt the degradation of complementary mRNA. Analysis of the mechanism of this process (reviewed in) revealed that the double-stranded RNA is converted by the RNAse Dicer into small 21-25 nucleotide-long RNA fragments. These fragments are then exported from nucleus in an exportin 5-dependent manner into the cytoplasm where the RNA strand that is complementary to the mRNA, the guide strand, is loaded into the RNA Induced Silencing Complex (RISC). A second class of small RNA molecules termed microRNAs (miRNAs) is encoded by long double-stranded regions usually found in introns of primary transcripts from polymerase II promoters. Whereas the process that leads to the production of miRNAs shares important aspects with the production of siRNAs, their mode of action is different. Presumably because miRNAs contain mismatches to their targets they do not trigger the degradation of the target mRNA but rather they suppress translation of the transcript. It was also shown that RNA interference also occurs in mammalian cells. This discovery revolutionized how protein expression can be silenced in cells grown in culture. It became rapidly clear, however, that small interfering RNAs (siRNAs) are poor drugs. Not only is siRNA delivery in vivo inefficient but the siRNAs are also unstable resulting in a limited and transient suppression of gene expression.

More recently, it was shown that siRNAs can also be generated by expression of so-called short hairpin RNAs (shRNAs) from plasmids. Transcription of shRNAs is in general driven by pol III promoters. After an export to the cytoplasm they are converted into siRNAs by Dicer and the guide strand is loaded into the RISC, which then results in the degradation of the target mRNA. shRNA expression cassettes can also be incorporated into viral vectors, including AAV vectors. Owing to their ability to drive long-term expression, AAV vector-based shRNA mediated gene silencing results in a lasting suppression of target gene expression. It became soon clear, however, that AAV vector-based gene silencing with shRNAs is not without potential complications. Mark Kay’s group used the strongly hepatotropic AAV serotype 8 to deliver shRNAs directed against luciferase, human α1-antitrypsin or the Hepatitis B Virus (HBV) genome. The large number of diverse shRNAs made off-target effects unlikely and further analysis revealed that the toxicity is most likely due to saturation of shared components of the shRNA/miRNA pathway, in particular exportin 5. Importantly, however, the toxicity was both dependent on viral dose and the precise nature of the shRNA sequence suggesting that a careful selection of the shRNA sequence, promoter strength and vector dose should allow to minimize toxicity, at least for certain applications.

Immune Responses Against the Viral Vector and Transgenes

Adenoviral Vectors

As mentioned earlier, adenoviral vectors are highly immunogenic, which is likely a major reason for their declining use in human cardiovascular gene therapy. This is especially pronounced in first- and second generation adenoviral vectors that express several viral genes as well the transgene. This immunogenicity is partly alleviated with helper-dependent, so- called gutless, adenoviral vectors because they do not express any viral genes. Nonetheless, even though the adaptive, especially cellular, immune response with gutless adenoviral vectors is attenuated, they still elicit a strong innate immune response. That an innate immune response against adenoviral vectors can have catastrophic consequences was demonstrated in a trial with a second generation adenoviral vector that resulted in the death of a trial participant. An additional complication for the use of adenoviral vectors is the high prevalence of neutralizing antibodies against adenoviruses. Approximately 97% of the population harbor neutralizing antibodies against type C adenoviruses, which include the commonly used Ad2 and Ad5.

Furthermore, although helper-dependent adenoviral vectors show reduced myocardial inflammation after intramyocardial injection when compared to first generation adenoviral vectors, the adenoviral capsid also seems to modulate the immune response against the transgene. Thus, while AAV-mediated GFP expression is long-lasting, both after intramyocardial and intrapericardial injection, expression from both first generation and helper-dependent, i.e. gutless, adenoviral vectors was transient, . In the future, it might be possible to devise strategies to overcome immune responses against either the adenoviral capsid or transgene but, at present, these immune reactions clearly pose a serious obstacle to the use of adenoviral vectors in cardiovascular gene therapy.

Lentiviral Vectors

Due to their ability to transduce non-dividing cells and their potential to trigger long-term transgene expression lentiviral vectors represent a possibly attractive gene delivery system for cardiovascular gene therapy. In contrast to adenoviral and AAV-based vectors, neutralizing antibodies against lentiviral vectors are usually not of concern. However, depending on the origin of the envelope protein used to create the pseudotyped lentiviral vectors, as well as the route of vector administration, a strong cytotoxic immune response can be generated. In general, this immune response is directed against the transgene, although a potential immune reaction against the envelope proteins can not be dismissed. The CTL-mediated destruction of transduced cells is likely due to fact that lentiviral vectors efficiently transduce antigen presenting cells such as Kupffer cell, splenic macrophages and dendritic cells. While in one report pseudotyping a feline immunodeficiency viral vector with the baculovirus envelope protein gp64 has been shown to overcome this limitation resulting in long-term transgene expression, in another mouse strain gene transfer with a gp64-psudotyped HIV-derived vector resulted only in transient (6 months) transgene expression. Whereas the use of tissue-specific promoters can mitigate the expression of the transgene in antigen presenting cells, at least under certain circumstances, these promoters appear not to be sufficiently tissue specific to prevent transgene expression in all antigen presenting cells. A clever approach to overcome this problem was developed by Naldini colleagues, . In this system, several copies of an miRNA target sequence that is perfectly complementary to an miRNA that is highly and specifically expressed in hematopoietic cells are included into the 3′-untranslated region of the transgene mRNA. . These miRNA target sequences are recognized by the endogenous miRNA in antigen presenting cells resulting in the degradation of the transgene mRNA in these cells but not in target cells. That this is a promising approach has been shown in a mouse model of factor IX deficiency. In this system, a lentiviral vector triggered long-term factor IX expression, .

AAV Vectors: Pre-Existing Neutralizing Antibodies

Regarding the effect of the immune system on transduction by AAV vectors, the comparatively high incidence of pre-existing neutralizing antibodies in the general population is probably the most concerning. While the prevalence of neutralizing antibodies varies with serotypes and geographic region, most studies report the presence of neutralizing titers of ≥1:20 in 20-40% of the population-. The cutoff of titers ≥1:20, however, is not only arbitrary it is also likely clinically insufficient as an exclusion criteria. Both studies in humans, , and animals- demonstrated that titers as low as 1:4 or even 1:2 can significantly influence or eliminate transgene expression or affect clinical results. For instance, in a clinical trial aimed at treating factor IX deficiency, in the high dose group with a liver-directed AAV vector encoding factor IX one individual with a neutralizing titer of 1:2 showed factor IX expression, albeit it only transiently. A second patient with a titer of 1:17, on the other hand, showed no factor IX expression. In the CUPID trial one exclusion criterion was neutralizing titers of >1:2, . In this trial, many patients in the high-dose group of AAV1.SERCA2a showed clinical improvements. However, two individuals in the high-dose group that were originally judged to harbor no neutralizing antibodies later tested positive for neutralizing antibodies, and these patients did not show any improvement, . Importantly, because patients with antibody titers as low as 1:4 were excluded from the CUPID trial, nearly half the prospective patients could not be enrolled because their antibody titers were too high, . This illustrates that future work is needed to allow the inclusion of most if not all potential patients into clinical trials based on AAV vectors.

Innate Immune Response

In contrast to adenoviral vectors, AAV does not induce a strong immune response, although systemic injection can give rise to a transient increase in inflammatory cytokines. Furthermore, it has been demonstrated that AAV can interact with factors of the complement cascade and that these interactions are essential for the endocytosis of AAV by macrophages. Nonetheless, while it is clear that the innate immune system can play a role in the host response to AAV infection, this response is not nearly as strong as the innate immune response against adenoviral vectors.

Cellular Immune Response Against the Transgene and the AAV Capsid

In gene therapy applications where a missing protein is replaced a significant risk for an immune response against the transgene exists. Fortunately, however, in cardiovascular gene transfer the transgenes to be expressed are almost exclusively proteins that are also expressed under physiological conditions but the level of expression is up- or down-regulated during disease progression. Because of this, the host developed tolerance against these proteins and an immune response against the transgene is highly unlikely.

A priori, a cellular immune response against the AAV capsid would seem unlikely because AAV vectors do not express any viral genes. However, it became clear, in an otherwise successful clinical trial to treat hemophilia B, that cellular immune responses against the AAV capsid can occur and that such cellular immune responses can result in the destruction of transduced cells, . More recently, however, it was demonstrated that such an immune response can be successfully suppressed by a short course of prednisolone. Furthermore, for cardiovascular gene therapy it is important to point out, however, that in the only cardiovascular gene therapy trial using AAV vectors, the CUPID trial, such a cytolytic T–cell response was not observed, . In this trial, only one patient showed a transient positive signal in an ELISPOT assay indicating a cellular immune response against the AAV1 capsid and this was concurrent with an influenza infection. Nonetheless, it seems advisable to monitor such responses carefully in cardiovascular or other gene therapy trials with AAV vectors.

Gene Delivery

Antegrade Arterial Infusion

Percutaneous coronary artery catheterization is a minimally invasive and well-established procedure that allows homogenous gene delivery to each territory of the heart. The major advantages of this approach are that it is minimally invasive and relatively safe. Thus, it is especially attractive for patients with end-stage heart failure. However, gene delivery can be impeded in patients with severe coronary artery disease. More importantly, there is some variability regarding the efficiency of antegrade coronary gene transfer, partly related to the relatively fast transit of the vector through the vasculature. In a large animal model of volume-overload induced heart failure, antegrade coronary gene transfer was shown to significantly restore cardiac function (Figure 3a). Infusion through the lumen of an inflated angioplasty catheter with temporary occlusion of the coronary artery may increase myocardial gene expression but remains controversial (Figure 3b). In a study by Boekstegers et al. ischemia during coronary artery infusion did not significantly increase myocardial transduction . In contrast, infusion with temporary coronary occlusion at a considerably higher flow rate and consequently elevated coronary pressures resulted in higher gene expression, but was also associated with more myocardial injury .

Antegrade coronary artery infusion a) Coronary artery infusion: The vector is injected through a catheter without interruption of the coronary flow b) Coronary artery infusion with occlusion of a coronary artery: The vector is injected through the lumen of an inflated angioplasty catheter c) Coronary artery infusion with simultaneous blocking of a coronary artery and a coronary vein: The vector is injected through an inflated angioplasty catheter and resides in the coronary circulation until both balloons are deflated.

An approach that focuses on enhancing the vector residence time in the coronary circulation is coronary venous blockade. Antegrade coronary infusion with a short occlusion of both a coronary artery and a coronary vein enhanced myocardial gene expression, (Figure 3c). This method preserved left ventricular function and inhibited ventricular remodeling in a large animal model of heart failure . However, even a short ischemia period should be considered as carrying a risk in patients with advanced heart failure.

Pathophysiology

To maximize the duration of vector exposure to the endothelium and at the same time minimize systemic distribution, Kaye et al. developed an extra-corporal device that drains blood from the coronary sinus using an occlusion catheter and returns the oxygenated coronary venous blood to the left main coronary artery via a peristaltic pump (V-Focus, Osprey Medical Inc, St Paul, MN, USA) , . In an ovine model of tachycardia induced heart failure, the closed loop recirculation method was more efficient in the transduction of cardiomyocytes than antegrade coronary infusion, which also translated into a greater improvement of left ventricular function, . Most of the approaches to improve the efficiency of coronary artery infusion are based on increasing the exposure time of the vector to the endothelium. However, transduction efficacy correlates with coronary flow as well as exposure time and vector concentration . Based on the former, antegrade coronary artery infusion supported by means of an increased coronary flow, for example an intra-aortic balloon pump, might further enhance cardiac gene transfer 63. Antegrade coronary artery infusion might be the safest percutaneous gene delivery method, but at the cost of efficiency relative to other techniques. However, to date, all optimization strategies implicate potential damage to the myocardium, limiting their clinical application.

Retrograde Venous Infusion

The coronary venous system provides an alternate route for the percutaneous delivery of therapeutic agents to the myocardium. In a clinical setting, this approach is attractive for patients with impaired coronary artery circulation and limited potential for revascularization. Access to the myocardium can be achieved with this approach regardless of atherosclerosis severity or coronary artery obstruction.

Numerous studies showed that retrograde coronary venous infusion is an alternate myocardial delivery method for cardioprotective drugs, . Shortly thereafter, retrograde venous infusion was also explored as a method for gene transfer. The rationale for a high transduction efficiency is based on controlling the exposure the time of vector to the endothelium, and on increasing the pressure gradient of capillary filtration, .

Indeed, studies in large animal models demonstrated that an efficient and homogenous myocardial transduction can be achieved by retroinfusion into the coronary venous system, , . Boeksteger et al. showed that gene expression after pressure-regulated retrograde venous infusion was significantly higher than after antegrade coronary delivery if the retroinfusion was accompanied by simultaneous induced ischemia (Figure 4b). Compared to percutaneous or surgical direct myocardial injection, retrograde venous infusion also achieved a more homogeneous and efficient reporter gene expression.

V-Focus system and retrograde coronary venous infusion a) Recirculating antegrade coronary artery infusion: The vector is injected into a coronary artery, collected from the coronary sinus and after oxygenation readministered into the coronary artery b) Retrograde coronary venous infusion with simultaneous blocking of a coronary artery and a coronary vein: The vector is injected into a coronary vein and resides in the coronary circulation until both balloons are deflated.

Similar to antegrade coronary artery infusion, a closed loop recirculation retrograde venous infusion approach is also feasible. Recently, Dr. Charles Bridges group demonstrated an extremely high transduction efficiency in the majority of cardiac cardiomyocytes in sheep while minimizing collateral organ exposure, using a retrograde recirculation method during cardio-pulmonary bypass (CPB) surgery .

Retrograde coronary infusion has been shown to be safe, when performed by a trained and experienced surgeon. Trauma to the coronary veins, a potential complication, could be minimized with modifications and improvements in catheter design. However, the procedure is not completely without risks. Highly elevated coronary venous pressure can result in myocardial edema or hemorrhage. Careful pressure monitoring should always be used to avoid this complication. Though with the growing demand for cardiac resynchronization therapy, cannulation of the coronary veins has become a routine procedure, monitoring of the pressure regulation adds to the complexity of this procedure. More importantly, the ischemia during the infusion might not be well tolerated in patients with advanced heart failure.

In summary, percutaneous retrograde coronary infusion is an effective, minimally invasive procedure, but relatively complex approach to gene delivery specifically to the myocardium. This approach requires some expertise and meticulous pressure monitoring to be performed safely.

Aortic cross clamping

In rodents, the vector is injected into the aortic root while the aorta and the pulmonary artery are cross-clamped for a few heart beats . While cross clamping the beating heart is only relatively simple and routine in rodents, a modified version of this technique has been developed for large animals. In comparison to rodent models, large animals studies using aortic cross clamping are complex surgical procedures that require cardiac arrest and a complete surgical isolation of the heart in situ with CPB, .

Virus injection into the aortic root during CPB with a cross-clamped aorta resulted in selective myocardial uptake and expression in piglets . To further increase the efficacy of this approach, Bridges et al. developed and refined a CBP platform with selective retrograde coronary sinus infusion and recirculation of the vector through the heart. This technique yielded extensive transgene expression in a significant percentage of cardiac myocytes, .

Notwithstanding the very high efficiency of the advanced aortic cross clamping techniques in large animals, their highly invasive nature and the associated potential morbidity limits a possible translation into clinical studies to patients undergoing cardiac surgery for other reasons.

Intravenous infusion

Intravenous injection is the simplest and less invasive method among current available methods of cardiac gene delivery. In rodents, injection into the tail vein results in successful cardiac gene expression. However, as of today, application of this method is feasible only in small animals, mostly due to limited cardiac specificity. While dilution by the systemic blood circulation compromises the vector concentration in the cardiac circulation, uptake by other organs such as liver, lung, and spleen before the vectors reach the heart is another issue. Bypassing the liver and the spleen by injecting the vectors into the pulmonary artery did not improve the efficacy. Development of highly cardiotropic vectors without any harmful effects on non-targeted organs, together with methods to augment cardiac uptake, such as ultrasound targeted microbubble destruction, may expand the application of intravenous injections to larger animals and eventually to human.

Direct intramyocardial injection

Intramyocardial injection is one of the most widely used gene transfer methods, ranging from small animal studies to clinical trials focusing on cardiac angiogenesis. The vectors are injected either epicardially (Figure 5a) or endocardially (Figure 5b) into the target area with a small gauge needle. The primary advantage of this method is that vector delivery bypasses the endothelial barrier. This results in a high local concentration at the injection site. In addition, by avoiding exposure to the blood, deactivation of the vectors by circulating DNases or neutralizing antibodies can be prevented. Furthermore, there is minimal exposure of the vector to off target organs, although local administration cannot completely avoid some systemic vector distribution, .

Direct myocardial injection and pericardial injection a) Percutaneous myocardial injection: The vector is injected with an injection catheter via an endocardial approach b) Surgical myocardial injection: The vector is injected via an epicardial approach c) Percutaneous pericardial injection: The vector is injected via a substernal approach.

The simplest approach, however invasive, is the injection during the thoracotomy. Surgical delivery offers direct visual confirmation, which allows precise control of the injection site, including an infarct border.

Another method of intramyocardial injection is based on a composite catheter system (TransAccess) . This delivery technique utilizes an intravascular ultrasound device for catheter guidance to the injection site before the vector is injected transvenously with an extendable needle. However, coronary sinus accessibility limits the cardiac regions impacted by this approach.

The endocardial approach requires a catheter with a retractable injection needle, and imaging guidance modality for determining the injection site. This includes electrical mapping systems , fluoroscopy , echocardiography , and magnetic resonance imaging . Currently, the NOGA electromechanical mapping system is the most commonly used guiding system that is also used in clinical angiogenesis trials.

Intramyocardial injection is attractive for local gene delivery. However, the application of this method for heart failure, where cardiomyocytes are globally impaired might be limited by a circumscribed target area and inhomogeneous expression profiles, .

Pericardial injection

Intrapericardial delivery is performed surgically in rodents , whereas for larger animals a percutaneous approach is available as well (Figure 5c). The percutaneous pericardial puncture has been proven to be feasible and safe, when guided by imaging techniques like fluoroscopy and intravascular ultrasound. The percutaneous access to the pericardial space can be achieved minimally invasive via a substernal/xhiphoidal puncture.

The pericardial space faces most of the cardiac wall except the septum. In heart failure, where widespread cardiac gene transfer with little systemic distribution is desired, this large-scale interface combined with the concept of a closed compartment can be a major advantage. These features potentially enable prolonged vector persistence, a slow release over time, a high vector concentration and minimal leakage to non-target organs. However, tightly joined pericardial cells restrict transfection to superficial myocardial layers-. This limitation can be partly overcome by the co-administration of various pharmacological agents. Proteolytic enzymes and polyethyleneimine have been shown to increase the penetration depth of the vectors and to allow progressive release, often at the cost of cardiac toxicity, . Although vectors are injected into a closed space, some studies reported extra-cardiac gene expression, probably due to the rapid turnover of the pericardium fluid through the lymphatic absorption.

Other delivery methods

Most cardiac gene transfer studies focus on the left ventricle. In contrast, a recently developed surgical gene “painting” method achieved transmural gene transfer in both atria without affecting ventricular cardiomyocytes . Although potentially interesting for targeting common atrial diseases like atrial fibrillation, this approach might be difficult to translate into a less invasive delivery method.

TARGETS

The last twenty years witnessed significant evolution in our understanding of the pathophysiology of heart failure in its molecular and cellular dimensions which broadened the scope of interventions available for gene therapy. We will discuss in this part some of the most important systems targeted to restore the function of failing cardiomyocytes. For targets to be validated, it is important that they rescue function in animal models when heart failure has been already established, that the rescue is not associated with arrhythmogenesis and that a gene-dose effect is established, ie with increasing expression of the gene of interest there is a concomitant improvement in function. Excitation-contraction coupling is dysregulated at multiple levels in the development of heart failure. For this reason, the various channels, transporters, and critical proteins have been targeted pharmacologically and by genetic editing to restore contractile function. In Figure 5, the various targets in excitation-contraction coupling are presented.

1. Targeting the β-adrenergic system

The β-adrenergic signaling is adversely affected by multiple changes which lead to β-ARs downregulation and de-sensitization. Up-regulation of the critical protein GRK2 seems to precipitate the abnormalities in β-AR signaling abnormalities. Several gene-based experiments tested the hypothesis that genetic manipulation of the myocardial β-AR system can enhance cardiac function.

a. Overexpression of β-AR

Overexpression of β-AR was initially tested as a simple way to overcome β-AR downregulation. Transgenic mice overexpressing the human β1-ARs suffered from severe cardiomyopathy . In contrast, mice with cardiac overexpression of β2-AR demonstrated increased basal myocardial adenylyl cyclase activity with increased left ventricular function . Both direct and intracoronary myocardial delivery of Adenovirus containing the human β2-AR transgene has resulted in enhanced cardiac performance in rodents and mammalian models ,.

b. Inhibition of G protein-coupled receptor kinases (GRKs)

The interaction between activated β-ARs and G proteins is regulated by kinases that modulate the receptor activity by phosphorylation of its carboxyl terminus. Agonist-dependent desensitization is mediated by a family of GRKs which phosphorylate the agonist-occupied receptors resulting in functional uncoupling. GRK2 binds to the Gbg subunit of activated G proteins phosphorylating β-ARs which then attach to an inhibitory protein β–arrestin. GRK2 is the most expressed GRK in the heart. It has been implicated in the pathogenesis of dysfunctional cardiac β-AR signaling accounting for a deleterious activity in the failing heart . Studies in mice in which HF was induced by a myocardial infarction, showed that selective GRK2 ablation 10 days postinfarction resulted in increased survival, halted ventricular remodeling and enhanced cardiac contractile performance . A peptide termed βARKct capable of inhibiting GRK2 mediated β-AR desensitization has been evaluated in vivo in animals. Using intracoronary adenovirus-mediated βARKct transgene delivery to rabbits 3 weeks after induced myocardial infarction demonstrated a marked reversal of ventricular dysfunction . More recent studies have focused on overexpressing βARKct in large animal models .

c. Activation of cardiac AC expression

Although detrimental outcomes were demonstrated with multiple elements of the β-adrenergic system used to improve the expression of cAMP, activation of AC type VI (AC VI) seems to have a unique favorable profile. Overexpression of AC VI in transgenic mice resulted in improved cardiac function in response to adrenergic stimulation along with increased cAMP production in isolated cardiac myocytes. Importantly, AC VI had a neutral effect on basal heart function and was not associated with any structural heart abnormalities . In a pacing model of HF in pigs, intracoronary delivery of adenovirus encoding AC VI resulted in improved LV function and remodeling, associated with increased cAMP generating capacity . The favorable effects of AC VI in preclinical studies are encouraging and this approach is currently under investigation for initiation of clinical trials in patients with HF .

2. Targeting Ca2+ cycling proteins

HF is characterized by multiple defects in Ca2-handling proteins involved in excitation-contraction coupling (Figure 6). Reversal of those defects by gene therapy techniques has shown very promising results. We will review the main aspects of those novel therapies in this section.

Excitation-Contraction Coupling in cardiac myocytes provides multiple targets for gene therapy.

a. Overexpression of SERCA2a

More than twenty years ago, Gwathmey et al first reported that calcium cycling is abnormal in human heart failure and was found to be partially due to decreased SERCA2a activity regardless of the etiology of the heart failure -,. Improvement in cardiac contractility after gene transfer of SERCA2a has been demonstrated in a large number of experimental models of heart failure ,. More importantly, long-term overexpression of SERCA2a by intracoronary delivery of AAV carrying SERCA2a has been associated with preserved systolic function and improved ventricular remodeling in a swine volume-overload model of HF . Beyond their effects on enhancing contractility, SERCA2a gene transfer has been shown to restore the energetics state of the heart , both in terms of energy supply and utilization, decrease ventricular arrhythmias , , and enhance coronary flow through activation of eNOS in endothelial cells .

b. PLN inhibition
Pathophysiology

Another approach to improve Ca2- handling involves inhibition of PLN. Decreasing PLN in human cardiac myocytes showed an improvement in contraction and relaxation velocities similar to the benefit seen with gene transfer of SERCA2a . Silencing of PLN expression in a sheep HF model resulted in improved SERCA activity along with improved systolic and diastolic LV function . In addition to the above conventional gene therapy strategies, RNAi therapy was used for the first time in a model of cardiac disease, specifically in rats with HF, in an attempt to suppress phospholamban expression. A AAV9-RNAi vector generated stable cardiac production of a regulatory RNA sequence, which in turn suppressed phospholamban expression. SERCA2a protein was subsequently increased accompanied by restoration of systolic and diastolic cardiac function .

c. Active I-1 and Inhibition of PP1

HF is associated with elevated PP1 activity in humans resulting in dephosphorylation of PLN. Overexpression of PP1 or ablation of I-1 in murine hearts has been associated with decreased β-AR-mediated contractile responses, depressed cardiac function and premature death consistent with HF -. Expression of a constitutively active I-1 in transgenic mice led to PP1 inhibition with increased phosphorylation of PLN and improved cardiac contractility. A recent study on transgenic mice expressing active I-1 confirmed the relationship between phosphorylation of PLN and SERCA2a activity. I-1 expression ameliorated ischemia/reperfusion-induced injury by reducing the infarct size and improving contractile recovery in addition to decreasing biomarkers of apoptosis and ER stress response -.

d. S100A1

S100 is part of a family of Ca2-modulated proteins implicated in intracellular regulatory activities. S100A1 is the most abundant S100 protein isoform in the heart. It promotes cardiac contractile and relaxation function through enhancing the activity of both RYRs and SERCA2a . In a rat model of HF, AAV6-mediated long-term expression of S100A1 resulted in a sustained in vivo reversal of LV dysfunction and remodeling , . More recently AAV9 gene transfer of S100A1 in a pre-clinical model of ischemic cardiomyopathy induced dramatic improvements in contractile function reinforcing the rationale that a clinical trial of S100A1 gene therapy for human heart failure should be forthcoming.

e. SUMO1

Recently Kho et al reported that the levels and activity of SERCA2a in cardiomyocytes are modulated in parallel with the levels of a cytoplasmic protein, small ubiquitin-like modifier type 1 (SUMO1) . SUMOs are a family of peptides that alter the function of other proteins in cells through a post-translational modification described as sumoylation. Sumoylation is involved in the modulation of various intracellular processes.

Kho et al. found that sumoylation enhanced the stability of SERCA2a in the cell as well as increase its activity. SUMO1 levels were reduced in murine and pig models of heart failure and in failing human ventricles. Increasing SUMO1 levels by AAV9 gene transfer led to a restoration of SERCA2a levels, improved hemodynamic performance, and reduced mortality among the animals with heart failure.

3. Homing of stem cells

The SDF1/CXCR4 complex has emerged as a therapeutic target in ischemic heart failure due to the ability of the SDF-1-CXCR4 system to promote the homing of stem cells to infracted myocardium. A clinical trial is underway to investigate the therapeutic benefit of SDF-1 overexpression in ischemic cardiomyopathy. In parallel, existing literature highlights the direct effects of CXCR4 on the myocardium and the cardiac myocyte. SDF-1 was shown to decrease myocardial contractility ex vivo and on cardiac myocytes. One recent report has shown increased ischemia-reperfusion injury in rat hearts overexpressing CXCR4, while another report investigated the modulation of beta-adrenergic receptor signaling by SDF-1 and CXCR4, raising interrogations over the potential complex interaction between these chemokines and the cardiovascular system. Pim-1 kinase has also been shown to enhance survival, proliferation, trafficking, lineage commitment, and functional engraftment of cardiac progenitor cells , . Pim-1 is unique as it mediates not only proliferation, but also lineage commitment, and functional engraftment in hearts , .

4.Targeting cell death

Apoptosis is a process of programmed cell death that is involved in normal organ development. In models of acute and subacute ischemia/reperfusion, overexpression of the anti-apoptotic protein Bcl-2, Akt or PI3 kinase reduces the rate of cardiomyocyte apoptosis and improved heart function . In ischemia/reperfusion injury where apoptosis plays an important part of myocardial damage gene therapy with pro-survival factors appears to be amenable to intervention, it is less clear if other forms of cardiac injury such as hypertrophy and HF can benefit from anti-apoptotic strategies. For these situations, sophisticated promoters with oxygen sensing and modified HIF1α promoters have been designed to induce survival factors in the setting of ischemia .

Clinical trials

Despite early failures, gene therapy trials for various diseases, most notably inherited blindness (whereby gene transfer by AAV vectors partially restored vision in a pediatric patient with Leber’s Congenital Amaurocis, a major cause of congenital blindness ), cancer, infectious diseases, monogenic diseases, and cardiovascular diseases are underway.

In heart failure, there are currently a number of trials ongoing or in the planning stages targeting various pathways for rescuing the failing myocardium. The targets that have been taken forward towards clinical trials include SERCA2a, adenylyl cyclase type 6, and SDF-1.

The first clinical trial of gene therapy in patients with HF was launched in the United States in 2007 , . CUPID was a multicenter trial designed to evaluate the safety profile and the biological effects of gene transfer of the SERCA2a cDNA by delivering a recombinant AAV1 (AAV1.SERCA2a) in patients with advanced HF. Participants in this trial were administered a single intracoronary infusion of AAV1.SERCA2a in an open-label approach , . Twelve month follow-up of these patients showed an acceptable safety profile , . Improvement was detected in several patients, reflected by symptomatic (5 patients), functional (4 patients), biomarker (2 patients) and LV function/remodeling (6 patients) parameters. In the phase 2 trial, 39 patients with advanced HF were randomized to receive intracoronary adeno-associated virus 1 (AAV1) mediated SERCA2a gene delivery (in one of 3 doses (low dose - 6 × 1011 DRP, middle dose - 3 × 1012 DRP and high dose - 1 × 1013 DRP) versus placebo. Patient’s symptoms (NYHA class, Minnesota Living With Heart Failure Questionnaire [MLWHFQ]), functional status (6 minute walk test [6MWT] and VO2 max), NT-proBNP levels and echocardiographic measures were evaluated over 6 months. Treatment success was determined by examining concordant trends in the above endpoints for group- and patient-based comparisons, as well as clinical outcomes. The AAV1.SERCA2a high-dose group met the pre-specified criteria for success at the group and individual patient levels. AAV1.SERCA2a treated patients, versus placebo, demonstrated improvement or stabilization in NYHA class, MLWHFQ, 6MWT, VO2 max, NT-proBNP levels, and LV end-systolic volumes. Significant increases in time to adjudicated CV events, and a decreased frequency of CV events per patient were observed in all patients receiving AAV1.SERCA2a. No increases in adverse events, disease-related events, or laboratory abnormalities were observed in AAV1.SERCA2a treated patients compared to placebo. Arrhythmias have been a concern with the overexpression of SERCA2a however in the CUPID trials there were no increases in arrhythmias as measured by ICD. Two other clinical trials targeting SERCA2a are currently enrolling patients. The first trial is in patients with advanced heart failure having received left ventricular assist devices at least one month prior to treatment and who will receive either AAV1.SERCA2a or saline. This trial is being conducted in the United Kingdom. A second Phase 2 monocenter double blind randomized placebo-controled, parallel study will be held in the Institut of Cardiology Pitié-Salpêtrière, Paris, France with the primary objective to investigate the impact of AAV1.SERCA2a on cardiac remodeling parameters in patients with severe heart failure.

In a separate clinical study adenovirus-5 encoding human adenylyl cyclase type 6 (is being delivered through intracoronary injection to patients with congestive heart failure. Intracoronary delivery of Ad5.hAC6 or PBS in 3:1 randomization with dose escalation, The patients will be randomized in a dose dependent fashion starting at 3.2 × 109 viral particles to 3.2 × 10^12 viral particles in 6 dose groups using a 3:1 randomization fashion with PBS (buffered saline being used for control). The trial is currently enrolling patients.

An additional trial is examining the effects of injecting SDF-1 directly into the myocardium of patients with ischemic heart disease. An open label dose escalation study to evaluate the safety of a single escalating dose of SDF-1 administered by endomyocardial injection to cohorts of adults with ischemic heart failure is currently enrolling patients. SDF-1 naked DNA will be injected directly into the myocardium at multiple sites through a percutaneous, left ventricular approach.

Conclusion

With a better understanding of the molecular mechanisms associated with heart failure and improved vectors with cardiotropic properties, gene therapy can now be considered as a viable adjuntcive treatment to mechanical and pharmacological therapies for heart failure. In the coming years, more targets will emerge that are amenable to genetic manipulations along with more advanced vector systems which will undoubtedly lead to safer and more effective clinical trials in gene therapy for heart failure.

Acknowledgments

Sources of Funding

This work is supported by NIH R01 HL093183, HL088434, and P20HL100396.

Non Standard Abreviations

AAVAdeno-associated vector
β-ARBeta Adrenergic Receptor
cAMPcyclic adenosine monophosphate
CMVCytoMegalo Virus
HFHeart Failure
I-1Protein Phosphatase Inhibitor-1
ICDImplantable cardiac defibrillators
LVADLeft Ventricular Assist Device
NAbNeutralizing Antibodies
PLNPhospholamban
SERCA2aSarcoplasmic Reticulum Ca2+ ATPase
SRSarcoplasmic Reticulum
Vgviral genomes

Footnotes

Disclosures

Roger Hajjar is a scientific founder of Celladon Co. which plans to commercilaize AAV1.SERCA2a for the treatment of heart failure.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

1. Raake PW, Tscheschner H, Reinkober J, Ritterhoff J, Katus HA, Koch WJ, Most P. Gene therapy targets in heart failure: the path to translation. Clin Pharmacol Ther. 2011;90(4):542–553.[PMC free article] [PubMed] [Google Scholar]
2. Simmen RC, Pabona JM, Velarde MC, Simmons C, Rahal O, Simmen FA. The emerging role of Kruppel-like factors in endocrine-responsive cancers of female reproductive tissues. J Endocrinol. 2010;204(3):223–231.[PMC free article] [PubMed] [Google Scholar]
3. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, Asnafi V, MacIntyre E, Dal Cortivo L, Radford I, Brousse N, Sigaux F, Moshous D, Hauer J, Borkhardt A, Belohradsky BH, Wintergerst U, Velez MC, Leiva L, Sorensen R, Wulffraat N, Blanche S, Bushman FD, Fischer A, Cavazzana-Calvo M. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118(9):3132–3142.[PMC free article] [PubMed] [Google Scholar]
4. Matrai J, Chuah MK, VandenDriessche T. Recent advances in lentiviral vector development and applications. Mol Ther. 2010;18(3):477–490.[PMC free article] [PubMed] [Google Scholar]
5. Raper SE, Chirmule N, Lee FS, Wivel NA, Bagg A, Gao GP, Wilson JM, Batshaw ML. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab. 2003;80(1-2):148–158. [PubMed] [Google Scholar]
6. Nonnenmacher M, Weber T. Intracellular Transport of Recombinant Adeno-Associated Virus Vectors. Gene Ther. 2012 in press. [PMC free article] [PubMed] [Google Scholar]
7. Nonnenmacher M, Weber T. Adeno-associated virus 2 infection requires endocytosis through the CLIC/GEEC pathway. Cell Host Microbe. 2011;10(6):563–576.[PMC free article] [PubMed] [Google Scholar]
8. Bantel-Schaal U, Braspenning-Wesch I, Kartenbeck J. Adeno-associated virus type 5 exploits two different entry pathways in human embryo fibroblasts. J Gen Virol. 2009;90(Pt 2):317–322. [PubMed] [Google Scholar]
9. Bantel-Schaal U, Hub B, Kartenbeck J. Endocytosis of adeno-associated virus type 5 leads to accumulation of virus particles in the Golgi compartment. J Virol. 2002;76(5):2340–2349.[PMC free article] [PubMed] [Google Scholar]
10. McTiernan CF, Mathier MA, Zhu X, Xiao X, Klein E, Swan CH, Mehdi H, Gibson G, Trichel AM, Glorioso JC, Feldman AM, McCurry KR, London B. Myocarditis following adeno-associated viral gene expression of human soluble TNF receptor (TNFRII-Fc) in baboon hearts. Gene Ther. 2007;14(23):1613–1622. [PubMed] [Google Scholar]
11. Gao G, Bish LT, Sleeper MM, Mu X, Sun L, Lou Y, Duan J, Hu C, Wang L, Sweeney HL. Transendocardial Delivery of AAV6 Results in Highly Efficient and Global Cardiac Gene Transfer in Rhesus Macaques. Hum Gene Ther. 2011;22(8):979–984. [PubMed] [Google Scholar]
12. Jessup M, Greenberg B, Mancini D, Cappola T, Pauly DF, Jaski B, Yaroshinsky A, Zsebo KM, Dittrich H, Hajjar RJ. Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID): A Phase 2 Trial of Intracoronary Gene Therapy of Sarcoplasmic Reticulum Ca2+-ATPase in Patients With Advanced Heart Failure. Circulation. 2011;124(3):304–313.[PMC free article] [PubMed] [Google Scholar]
13. Koerber JT, Klimczak R, Jang JH, Dalkara D, Flannery JG, Schaffer DV. Molecular evolution of adeno-associated virus for enhanced glial gene delivery. Mol Ther. 2009;17(12):2088–2095.[PMC free article] [PubMed] [Google Scholar]
14. Michelfelder S, Trepel M. Adeno-associated viral vectors and their redirection to cell-type specific receptors. Adv Genet. 2009;67:29–60. [PubMed] [Google Scholar]
15. Yang F, Green JJ, Dinio T, Keung L, Cho SW, Park H, Langer R, Anderson DG. Gene delivery to human adult and embryonic cell-derived stem cells using biodegradable nanoparticulate polymeric vectors. Gene Ther. 2009;16(4):533–546.[PMC free article] [PubMed] [Google Scholar]
16. Ying Y, Müller OJ, Goehringer C, Leuchs B, Trepel M, Katus HA, Kleinschmidt JA. Heart-targeted adeno-associated viral vectors selected by in vivo biopanning of a random viral display peptide library. Gene Ther. 2010;17(8):980–990. [PubMed] [Google Scholar]
17. Asokan A, Conway JC, Phillips JL, Li C, Hegge J, Sinnott R, Yadav S, DiPrimio N, Nam H-J, Agbandje-McKenna M, McPhee S, Wolff J, Samulski RJ. Reengineering a receptor footprint of adeno-associated virus enables selective and systemic gene transfer to muscle. Nat Biotechnol. 2010;28(1):79–82.[PMC free article] [PubMed] [Google Scholar]
18. Boecker W, Bernecker OY, Wu JC, Zhu X, Sawa T, Grazette L, Rosenzweig A, del Monte F, Schmidt U, Hajjar RJ. Cardiac-specific gene expression facilitated by an enhanced myosin light chain promoter. Mol Imaging. 2004;3(2):69–75. [PubMed] [Google Scholar]
19. Muller OJ, Leuchs B, Pleger ST, Grimm D, Franz WM, Katus HA, Kleinschmidt JA. Improved cardiac gene transfer by transcriptional and transductional targeting of adeno-associated viral vectors. Cardiovasc Res. 2006;70(1):70–78. [PubMed] [Google Scholar]
20. Heine HL, Leong HS, Rossi FM, McManus BM, Podor TJ. Strategies of conditional gene expression in myocardium: an overview. Methods Mol Med. 2005;112:109–154. [PubMed] [Google Scholar]
21. Franz WM, Rothmann T, Frey N, Katus HA. Analysis of tissue-specific gene delivery by recombinant adenoviruses containing cardiac-specific promoters. Cardiovasc Res. 1997;35(3):560–566. [PubMed] [Google Scholar]
22. Pacak CA, Sakai Y, Thattaliyath BD, Mah CS, Byrne BJ. Tissue specific promoters improve specificity of AAV9 mediated transgene expression following intra-vascular gene delivery in neonatal mice. Genet Vaccines Ther. 2008;6:13.[PMC free article] [PubMed] [Google Scholar]
23. Prasad KM, Xu Y, Yang Z, Acton ST, French BA. Robust cardiomyocyte-specific gene expression following systemic injection of AAV: in vivo gene delivery follows a Poisson distribution. Gene Ther. 2011;18(1):43–52.[PMC free article] [PubMed] [Google Scholar]
24. Bohl D, Salvetti A, Moullier P, Heard JM. Control of erythropoietin delivery by doxycycline in mice after intramuscular injection of adeno-associated vector. Blood. 1998;92(5):1512–1517. [PubMed] [Google Scholar]
25. Rendahl KG, Leff SE, Otten GR, Spratt SK, Bohl D, Van Roey M, Donahue BA, Cohen LK, Mandel RJ, Danos O, Snyder RO. Regulation of gene expression in vivo following transduction by two separate rAAV vectors. Nat Biotechnol. 1998;16(8):757–761. [PubMed] [Google Scholar]
26. Ye X, Rivera VM, Zoltick P, Cerasoli F, Schnell MA, Gao G, Hughes JV, Gilman M, Wilson JM. Regulated delivery of therapeutic proteins after in vivo somatic cell gene transfer. Science. 1999;283(5398):88–91. [PubMed] [Google Scholar]
27. Szymanski P, Kretschmer PJ, Bauzon M, Jin F, Qian HS, Rubanyi GM, Harkins RN, Hermiston TW. Development and validation of a robust and versatile one-plasmid regulated gene expression system. Mol Ther. 2007;15(7):1340–1347. [PubMed] [Google Scholar]
28. Tang YL, Tang Y, Zhang YC, Agarwal A, Kasahara H, Qian K, Shen L, Phillips MI. A hypoxia-inducible vigilant vector system for activating therapeutic genes in ischemia. Gene Ther. 2005;12(15):1163–1170. [PubMed] [Google Scholar]
29. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806–811. [PubMed] [Google Scholar]
30. Meister G, Tuschl T. Mechanisms of gene silencing by double-stranded RNA. Nature. 2004;431(7006):343–349. [PubMed] [Google Scholar]
31. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411(6836):494–498. [PubMed] [Google Scholar]
32. Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs in mammalian cells. Science. 2002;296(5567):550–553. [PubMed] [Google Scholar]
33. Xia H, Mao Q, Eliason SL, Harper SQ, Martins IH, Orr HT, Paulson HL, Yang L, Kotin RM, Davidson BL. RNAi suppresses polyglutamine-induced neurodegeneration in a model of spinocerebellar ataxia. Nat Med. 2004;10(8):816–820. [PubMed] [Google Scholar]
34. Grimm D, Streetz KL, Jopling CL, Storm TA, Pandey K, Davis CR, Marion P, Salazar F, Kay MA. Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature. 2006;441(7092):537–541. [PubMed] [Google Scholar]
35. Alba R, Bosch A, Chillon M. Gutless adenovirus: last-generation adenovirus for gene therapy. Gene Ther. 2005;12(Suppl 1):S18–27. [PubMed] [Google Scholar]
36. Nayak S, Herzog RW. Progress and prospects: immune responses to viral vectors. Gene Ther. 2010;17(3):295–304.[PMC free article] [PubMed] [Google Scholar]
37. Vassalli G, Bueler H, Dudler J, von Segesser LK, Kappenberger L. Adeno-associated virus (AAV) vectors achieve prolonged transgene expression in mouse myocardium and arteries in vivo: a comparative study with adenovirus vectors. Int J Cardiol. 2003;90(2-3):229–238. [PubMed] [Google Scholar]
38. Fleury S, Driscoll R, Simeoni E, Dudler J, von Segesser LK, Kappenberger L, Vassalli G. Helper-dependent adenovirus vectors devoid of all viral genes cause less myocardial inflammation compared with first-generation adenovirus vectors. Basic Res Cardiol. 2004;99(4):247–256. [PubMed] [Google Scholar]
39. Sinn PL, Burnight ER, Hickey MA, Blissard GW, McCray PB., Jr. Persistent gene expression in mouse nasal epithelia following feline immunodeficiency virus-based vector gene transfer. J Virol. 2005;79(20):12818–12827.[PMC free article] [PubMed] [Google Scholar]
40. Kremer KL, Dunning KR, Parsons DW, Anson DS. Gene delivery to airway epithelial cells in vivo: a direct comparison of apical and basolateral transduction strategies using pseudotyped lentivirus vectors. J Gene Med. 2007;9(5):362–368. [PubMed] [Google Scholar]
41. Annoni A, Brown BD, Cantore A, Sergi LS, Naldini L, Roncarolo MG. In vivo delivery of a microRNA-regulated transgene induces antigen-specific regulatory T cells and promotes immunologic tolerance. Blood. 2009;114(25):5152–5161.[PMC free article] [PubMed] [Google Scholar]
42. Brown BD, Cantore A, Annoni A, Sergi LS, Lombardo A, Della Valle P, D’Angelo A, Naldini L. A microRNA-regulated lentiviral vector mediates stable correction of hemophilia B mice. Blood. 2007;110(13):4144–4152. [PubMed] [Google Scholar]
43. Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, Masurier C. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther. 2010;21(6):704–712. [PubMed] [Google Scholar]
44. Calcedo R, Vandenberghe LH, Gao G, Lin J, Wilson JM. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J Infect Dis. 2009;199(3):381–390. [PubMed] [Google Scholar]
45. van der Marel S, Comijn EM, Verspaget HW, van Deventer S, van den Brink GR, Petry H, Hommes DW, Ferreira V. Neutralizing antibodies against adeno-associated viruses in inflammatory bowel disease patients: Implications for gene therapy. Inflamm Bowel Dis. 2011 [PubMed] [Google Scholar]
46. Jaski BE, Jessup ML, Mancini DM, Cappola TP, Pauly DF, Greenberg B, Borow K, Dittrich H, Zsebo KM, Hajjar RJ. Calcium upregulation by percutaneous administration of gene therapy in cardiac disease (CUPID Trial), a first-in-human phase 1/2 clinical trial. J Card Fail. 2009;15(3):171–181.[PMC free article] [PubMed] [Google Scholar]
47. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, Ozelo MC, Hoots K, Blatt P, Konkle B, Dake M, Kaye R, Razavi M, Zajko A, Zehnder J, Rustagi PK, Nakai H, Chew A, Leonard D, Wright JF, Lessard RR, Sommer JM, Tigges M, Sabatino D, Luk A, Jiang H, Mingozzi F, Couto L, Ertl HC, High KA, Kay MA. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12(3):342–347. [PubMed] [Google Scholar]
48. Li H, Lin SW, Giles-Davis W, Li Y, Zhou D, Xiang ZQ, High KA, Ertl HC. A preclinical animal model to assess the effect of pre-existing immunity on AAV-mediated gene transfer. Mol Ther. 2009;17(7):1215–1224.[PMC free article] [PubMed] [Google Scholar]
49. Petry H, Brooks A, Orme A, Wang P, Liu P, Xie J, Kretschmer P, Qian HS, Hermiston TW, Harkins RN. Effect of viral dose on neutralizing antibody response and transgene expression after AAV1 vector re-administration in mice. Gene Ther. 2008;15(1):54–60. [PubMed] [Google Scholar]
50. Rapti K, Louis-Jeune V, Kohlbrenner E, Ishikawa K, Ladage D, Zolotukhin S, Hajjar RJ, Weber T. Neutralizing Antibodies Against AAV Serotypes 1, 2, 6, and 9 in Sera of Commonly Used Animal Models. Mol Ther. 2012;20(1):73–83.[PMC free article] [PubMed] [Google Scholar]
51. Scallan CD, Jiang H, Liu T, Patarroyo-White S, Sommer JM, Zhou S, Couto LB, Pierce GF. Human immunoglobulin inhibits liver transduction by AAV vectors at low AAV2 neutralizing titers in SCID mice. Blood. 2006;107(5):1810–1817. [PubMed] [Google Scholar]
52. Zaiss A-K, Liu Q, Bowen GP, Wong NCW, Bartlett JS, Muruve DA. Differential activation of innate immune responses by adenovirus and adeno-associated virus vectors. J Virol. 2002;76(9):4580–4590.[PMC free article] [PubMed] [Google Scholar]
53. Zaiss AK, Cotter MJ, White LR, Clark SA, Wong NCW, Holers VM, Bartlett JS, Muruve DA. Complement is an essential component of the immune response to adeno-associated virus vectors. J Virol. 2008;82(6):2727–2740.[PMC free article] [PubMed] [Google Scholar]
54. Mingozzi F, Hasbrouck NC, Basner-Tschakarjan E, Edmonson SA, Hui DJ, Sabatino DE, Zhou S, Wright JF, Jiang H, Pierce GF, Arruda VR, High KA. Modulation of tolerance to the transgene product in a nonhuman primate model of AAV-mediated gene transfer to liver. Blood. 2007;110(7):2334–2341.[PMC free article] [PubMed] [Google Scholar]
55. Nathwani AC, Tuddenham EG, Rangarajan S, Rosales C, McIntosh J, Linch DC, Chowdary P, Riddell A, Pie AJ, Harrington C, O’Beirne J, Smith K, Pasi J, Glader B, Rustagi P, Ng CY, Kay MA, Zhou J, Spence Y, Morton CL, Allay J, Coleman J, Sleep S, Cunningham JM, Srivastava D, Basner-Tschakarjan E, Mingozzi F, High KA, Gray JT, Reiss UM, Nienhuis AW, Davidoff AM. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med. 2011;365(25):2357–2365.[PMC free article] [PubMed] [Google Scholar]
56. Boekstegers P, von Degenfeld G, Giehrl W, Heinrich D, Hullin R, Kupatt C, Steinbeck G, Baretton G, Middeler G, Katus H, Franz WM. Myocardial gene transfer by selective pressure-regulated retroinfusion of coronary veins. Gene Ther. 2000;7(3):232–240. [PubMed] [Google Scholar]
57. Shah AS, White DC, Emani S, Kypson AP, Lilly RE, Wilson K, Glower DD, Lefkowitz RJ, Koch WJ. In vivo ventricular gene delivery of a beta-adrenergic receptor kinase inhibitor to the failing heart reverses cardiac dysfunction. Circulation. 2001;103(9):1311–1316. [PubMed] [Google Scholar]
58. Hayase M, Del Monte F, Kawase Y, Macneill BD, McGregor J, Yoneyama R, Hoshino K, Tsuji T, De Grand AM, Gwathmey JK, Frangioni JV, Hajjar RJ. Catheter-based antegrade intracoronary viral gene delivery with coronary venous blockade. Am J Physiol Heart Circ Physiol. 2005;288(6):H2995–3000.[PMC free article] [PubMed] [Google Scholar]
59. Beeri R, Chaput M, Guerrero JL, Kawase Y, Yosefy C, Abedat S, Karakikes I, Morel C, Tisosky A, Sullivan S, Handschumacher MD, Gilon D, Vlahakes GJ, Hajjar RJ, Levine RA. Gene delivery of sarcoplasmic reticulum calcium ATPase inhibits ventricular remodeling in ischemic mitral regurgitation. Circ Heart Fail. 2010;3(5):627–634.[PMC free article] [PubMed] [Google Scholar]
60. Byrne CD, Olufadi R, Bruce KD, Cagampang FR, Ahmed MH. Metabolic disturbances in non-alcoholic fatty liver disease. Clin Sci (Lond) 2009;116(7):539–564. [PubMed] [Google Scholar]
61. Kaye DM, Preovolos A, Marshall T, Byrne M, Hoshijima M, Hajjar R, Mariani JA, Pepe S, Chien KR, Power JM. Percutaneous cardiac recirculation-mediated gene transfer of an inhibitory phospholamban peptide reverses advanced heart failure in large animals. J Am Coll Cardiol. 2007;50(3):253–260. [PubMed] [Google Scholar]
62. Donahue JK, Kikkawa K, Johns DC, Marban E, Lawrence JH. Ultrarapid, highly efficient viral gene transfer to the heart. Proc Natl Acad Sci U S A. 1997;94(9):4664–4668.[PMC free article] [PubMed] [Google Scholar]
63. Ladage D, Tilemann L, Ishikawa K, Kho C, Lee A, Hajjar R, Kawase Y. Increasing the efficacy of myocardial gene therapy with an intra-aortic balloon pump in a large animal model of heart failure. Eur Heart J. 2011;32(Abstract Supplement):814.[Google Scholar]
64. Ryden L, Tadokoro H, Sjoquist PO, Regardh C, Kobayashi S, Corday E, Drury JK. Pharmacokinetic analysis of coronary venous retroinfusion: a comparison with anterograde coronary artery drug administration using metoprolol as a tracer. J Am Coll Cardiol. 1991;18(2):603–612. [PubMed] [Google Scholar]
65. Karagueuzian HS, Ohta M, Drury JK, Fishbein MC, Meerbaum S, Corday E, Mandel WJ, Peter T. Coronary venous retroinfusion of procainamide: a new approach for the management of spontaneous and inducible sustained ventricular tachycardia during myocardial infarction. J Am Coll Cardiol. 1986;7(3):551–563. [PubMed] [Google Scholar]
66. Su LT, Gopal K, Wang Z, Yin X, Nelson A, Kozyak BW, Burkman JM, Mitchell MA, Low DW, Bridges CR, Stedman HH. Uniform scale-independent gene transfer to striated muscle after transvenular extravasation of vector. Circulation. 2005;112(12):1780–1788. [PubMed] [Google Scholar]
67. Raake P, von Degenfeld G, Hinkel R, Vachenauer R, Sandner T, Beller S, Andrees M, Kupatt C, Schuler G, Boekstegers P. Myocardial gene transfer by selective pressure-regulated retroinfusion of coronary veins: comparison with surgical and percutaneous intramyocardial gene delivery. J Am Coll Cardiol. 2004;44(5):1124–1129. [PubMed] [Google Scholar]
68. Hou D, Maclaughlin F, Thiesse M, Panchal VR, Bekkers BC, Wilson EA, Rogers PI, Coleman MC, March KL. Widespread regional myocardial transfection by plasmid encoding Del-1 following retrograde coronary venous delivery. Catheter Cardiovasc Interv. 2003;58(2):207–211. [PubMed] [Google Scholar]
69. White JD, Thesier DM, Swain JB, Katz MG, Tomasulo C, Henderson A, Wang L, Yarnall C, Fargnoli A, Sumaroka M, Isidro A, Petrov M, Holt D, Nolen-Walston R, Koch WJ, Stedman HH, Rabinowitz J, Bridges CR. Myocardial gene delivery using molecular cardiac surgery with recombinant adeno-associated virus vectors in vivo. Gene Ther. 2011;18(6):546–552.[PMC free article] [PubMed] [Google Scholar]
70. Hajjar RJ, Schmidt U, Matsui T, Guerrero JL, Lee KH, Gwathmey JK, Dec GW, Semigran MJ, Rosenzweig A. Modulation of ventricular function through gene transfer in vivo. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(9):5251–5256.[PMC free article] [PubMed] [Google Scholar]
71. Davidson MJ, Jones JM, Emani SM, Wilson KH, Jaggers J, Koch WJ, Milano CA. Cardiac gene delivery with cardiopulmonary bypass. Circulation. 2001;104(2):131–133. [PubMed] [Google Scholar]
72. Bridges CR, Gopal K, Holt DE, Yarnall C, Cole S, Anderson RB, Yin X, Nelson A, Kozyak BW, Wang Z, Lesniewski J, Su LT, Thesier DM, Sundar H, Stedman HH. Efficient myocyte gene delivery with complete cardiac surgical isolation in situ. J Thorac Cardiovasc Surg. 2005;130(5):1364. [PubMed] [Google Scholar]
73. Swain JD, Katz MG, White JD, Thesier DM, Henderson A, Stedman HH, Bridges CR. A translatable, closed recirculation system for AAV6 vector-mediated myocardial gene delivery in the large animal. Methods Mol Biol. 2011;709:331–354. [PubMed] [Google Scholar]
74. Chen ZY, Liang K, Qiu RX, Luo LP. Ultrasound- and liposome microbubble-mediated targeted gene transfer to cardiomyocytes in vivo accompanied by polyethylenimine. J Ultrasound Med. 2011;30(9):1247–1258. [PubMed] [Google Scholar]
75. Fujii H, Li SH, Wu J, Miyagi Y, Yau TM, Rakowski H, Egashira K, Guo J, Weisel RD, Li RK. Repeated and targeted transfer of angiogenic plasmids into the infarcted rat heart via ultrasound targeted microbubble destruction enhances cardiac repair. Eur Heart J. 2011 [PubMed] [Google Scholar]
76. Grossman PM, Han Z, Palasis M, Barry JJ, Lederman RJ. Incomplete retention after direct myocardial injection. Catheter Cardiovasc Interv. 2002;55(3):392–397. [PubMed] [Google Scholar]
77. Bish LT, Morine K, Sleeper MM, Sanmiguel J, Wu D, Gao G, Wilson JM, Sweeney HL. Adeno-associated virus (AAV) serotype 9 provides global cardiac gene transfer superior to AAV1, AAV6, AAV7, and AAV8 in the mouse and rat. Hum Gene Ther. 2008;19(12):1359–1368.[PMC free article] [PubMed] [Google Scholar]
78. Thompson CA, Nasseri BA, Makower J, Houser S, McGarry M, Lamson T, Pomerantseva I, Chang JY, Gold HK, Vacanti JP, Oesterle SN. Percutaneous transvenous cellular cardiomyoplasty. A novel nonsurgical approach for myocardial cell transplantation. J Am Coll Cardiol. 2003;41(11):1964–1971. [PubMed] [Google Scholar]
79. Gepstein L, Hayam G, Ben-Haim SA. A novel method for nonfluoroscopic catheter-based electroanatomical mapping of the heart. In vitro and in vivo accuracy results. Circulation. 1997;95(6):1611–1622. [PubMed] [Google Scholar]
80. Lederman RJ, Guttman MA, Peters DC, Thompson RB, Sorger JM, Dick AJ, Raman VK, McVeigh ER. Catheter-based endomyocardial injection with real-time magnetic resonance imaging. Circulation. 2002;105(11):1282–1284.[PMC free article] [PubMed] [Google Scholar]
81. French BA, Mazur W, Geske RS, Bolli R. Direct in vivo gene transfer into porcine myocardium using replication-deficient adenoviral vectors. Circulation. 1994;90(5):2414–2424. [PubMed] [Google Scholar]
82. Fromes Y, Salmon A, Wang X, Collin H, Rouche A, Hagege A, Schwartz K, Fiszman MY. Gene delivery to the myocardium by intrapericardial injection. Gene Ther. 1999;6(4):683–688. [PubMed] [Google Scholar]
83. Lamping KG, Rios CD, Chun JA, Ooboshi H, Davidson BL, Heistad DD. Intrapericardial administration of adenovirus for gene transfer. Am J Physiol. 1997;272(1 Pt 2):H310–317. [PubMed] [Google Scholar]
84. Ladage D, Turnbull IC, Ishikawa K, Takewa Y, Rapti K, Morel C, Karakikes I, Hadri L, Muller-Ehmsen J, Costa KD, Hajjar RJ, Kawase Y. Delivery of gelfoam-enabled cells and vectors into the pericardial space using a percutaneous approach in a porcine model. Gene Ther. 2011[PMC free article] [PubMed] [Google Scholar]
85. Roques C, Salmon A, Fiszman MY, Fattal E, Fromes Y. Intrapericardial administration of novel DNA formulations based on thermosensitive Poloxamer 407 gel. Int J Pharm. 2007;331(2):220–223. [PubMed] [Google Scholar]
86. Kikuchi K, McDonald AD, Sasano T, Donahue JK. Targeted modification of atrial electrophysiology by homogeneous transmural atrial gene transfer. Circulation. 2005;111(3):264–270. [PubMed] [Google Scholar]
87. Engelhardt S, Hein L, Dyachenkow V, Kranias EG, Isenberg G, Lohse MJ. Altered calcium handling is critically involved in the cardiotoxic effects of chronic beta-adrenergic stimulation. Circulation. 2004;109(9):1154–1160. [PubMed] [Google Scholar]
88. Milano CA, Allen LF, Rockman HA, Dolber PC, McMinn TR, Chien KR, Johnson TD, Bond RA, Lefkowitz RJ. Enhanced myocardial function in transgenic mice overexpressing the beta 2-adrenergic receptor. Science. 1994;264(5158):582–586. [PubMed] [Google Scholar]
89. Maurice JP, Hata JA, Shah AS, White DC, McDonald PH, Dolber PC, Wilson KH, Lefkowitz RJ, Glower DD, Koch WJ. Enhancement of cardiac function after adenoviral-mediated in vivo intracoronary beta2-adrenergic receptor gene delivery. J Clin Invest. 1999;104(1):21–29.[PMC free article] [PubMed] [Google Scholar]
90. Shah AS, Lilly RE, Kypson AP, Tai O, Hata JA, Pippen A, Silvestry SC, Lefkowitz RJ, Glower DD, Koch WJ. Intracoronary adenovirus-mediated delivery and overexpression of the beta(2)-adrenergic receptor in the heart : prospects for molecular ventricular assistance. Circulation. 2000;101(4):408–414. [PubMed] [Google Scholar]
91. Hata JA, Williams ML, Koch WJ. Genetic manipulation of myocardial beta-adrenergic receptor activation and desensitization. J Mol Cell Cardiol. 2004;37(1):11–21. [PubMed] [Google Scholar]
92. Raake PW, Vinge LE, Gao E, Boucher M, Rengo G, Chen X, DeGeorge BR, Jr., Matkovich S, Houser SR, Most P, Eckhart AD, Dorn GW, 2nd, Koch WJ. G protein-coupled receptor kinase 2 ablation in cardiac myocytes before or after myocardial infarction prevents heart failure. Circ Res. 2008;103(4):413–422.[PMC free article] [PubMed] [Google Scholar]
93. Katz MG, Fargnoli AS, Swain JD, Tomasulo CE, Ciccarelli M, Huang ZM, Rabinowitz JE, Bridges CR. AAV6-betaARKct gene delivery mediated by molecular cardiac surgery with recirculating delivery (MCARD) in sheep results in robust gene expression and increased adrenergic reserve. J Thorac Cardiovasc Surg. 2011[PMC free article] [PubMed] [Google Scholar]
94. Gao MH, Lai NC, Roth DM, Zhou J, Zhu J, Anzai T, Dalton N, Hammond HK. Adenylylcyclase increases responsiveness to catecholamine stimulation in transgenic mice. Circulation. 1999;99(12):1618–1622. [PubMed] [Google Scholar]
95. Lai NC, Roth DM, Gao MH, Tang T, Dalton N, Lai YY, Spellman M, Clopton P, Hammond HK. Intracoronary adenovirus encoding adenylyl cyclase VI increases left ventricular function in heart failure. Circulation. 2004;110(3):330–336. [PubMed] [Google Scholar]
96. Lai NC, Roth DM, Gao MH, Fine S, Head BP, Zhu J, McKirnan MD, Kwong C, Dalton N, Urasawa K, Roth DA, Hammond HK. Intracoronary delivery of adenovirus encoding adenylyl cyclase VI increases left ventricular function and cAMP-generating capacity. Circulation. 2000;102(19):2396–2401. [PubMed] [Google Scholar]
97. Gwathmey JK, Copelas L, MacKinnon R, Schoen FJ, Feldman MD, Grossman W, Morgan JP. Abnormal intracellular calcium handling in myocardium from patients with end-stage heart failure. Circulation Research. 1987;61(1):70–76. [PubMed] [Google Scholar]
98. Hasenfuss G, Reinecke H, Studer R, Meyer M, Pieske B, Holtz J, Holubarsch C, Posival H, Just H, Drexler H. Relation between myocardial function and expression of sarcoplasmic reticulum Ca(2+)-ATPase in failing and nonfailing human myocardium. Circulation Research. 1994;75(3):434–442. [PubMed] [Google Scholar]
99. Meyer M, Schillinger W, Pieske B, Holubarsch C, Heilmann C, Posival H, Kuwajima G, Mikoshiba K, Just H, Hasenfuss G. Alterations of sarcoplasmic reticulum proteins in failing human dilated cardiomyopathy. Circulation. 1995;92(4):778–784. [PubMed] [Google Scholar]
100. Schmidt U, Hajjar RJ, Kim CS, Lebeche D, Doye AA, Gwathmey JK. Human heart failure: cAMP stimulation of SR Ca(2+)-ATPase activity and phosphorylation level of phospholamban. Am J Physiol. 1999;277(2 Pt 2):H474–480. [PubMed] [Google Scholar]
101. Lompre AM, Lambert F, Lakatta EG, Schwartz K. Expression of sarcoplasmic reticulum Ca(2+)-ATPase and calsequestrin genes in rat heart during ontogenic development and aging. Circ Res. 1991;69(5):1380–1388. [PubMed] [Google Scholar]
102. Miyamoto MI, del Monte F, Schmidt U, DiSalvo TS, Kang ZB, Matsui T, Guerrero JL, Gwathmey JK, Rosenzweig A, Hajjar RJ. Adenoviral gene transfer of SERCA2a improves left-ventricular function in aortic-banded rats in transition to heart failure. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(2):793–798.[PMC free article] [PubMed] [Google Scholar]
103. del Monte F, Harding SE, Schmidt U, Matsui T, Kang ZB, Dec GW, Gwathmey JK, Rosenzweig A, Hajjar RJ. Restoration of contractile function in isolated cardiomyocytes from failing human hearts by gene transfer of SERCA2a. Circulation. 1999;100(23):2308–2311.[PMC free article] [PubMed] [Google Scholar]
104. Kawase Y, Ly HQ, Prunier F, Lebeche D, Shi Y, Jin H, Hadri L, Yoneyama R, Hoshino K, Takewa Y, Sakata S, Peluso R, Zsebo K, Gwathmey JK, Tardif JC, Tanguay JF, Hajjar RJ. Reversal of cardiac dysfunction after long-term expression of SERCA2a by gene transfer in a pre-clinical model of heart failure. J Am Coll Cardiol. 2008;51(11):1112–1119. [PubMed] [Google Scholar]
105. Sakata S, Lebeche D, Sakata N, Sakata Y, Chemaly ER, Liang LF, Tsuji T, Takewa Y, Del Monte F, Peluso R, Zsebo K, Jeong D, Park WJ, Kawase Y, Hajjar RJ. Restoration of mechanical and energetic function in failing aortic-banded rat hearts by gene transfer of calcium cycling proteins. J Mol Cell Cardiol. 2007;42(4):852–861.[PMC free article] [PubMed] [Google Scholar]
106. Sakata S, Lebeche D, Sakata Y, Sakata N, Chemaly ER, Liang L, Nakajima-Takenaka C, Tsuji T, Konishi N, del Monte F, Hajjar RJ, Takaki M. Transcoronary gene transfer of SERCA2a increases coronary blood flow and decreases cardiomyocyte size in a type 2 diabetic rat model. Am J Physiol Heart Circ Physiol. 2007;292(2):H1204–1207. [PubMed] [Google Scholar]
107. Lyon AR, Bannister ML, Collins T, Pearce E, Sepehripour AH, Dubb SS, Garcia E, O’Gara P, Liang L, Kohlbrenner E, Hajjar RJ, Peters NS, Poole-Wilson PA, Macleod KT, Harding SE. SERCA2a Gene Transfer Decreases SR Calcium Leak and Reduces Ventricular Arrhythmias in a Model of Chronic Heart Failure. Circ Arrhythm Electrophysiol. 2011[PMC free article] [PubMed] [Google Scholar]
108. Cutler MJ, Wan X, Laurita KR, Hajjar RJ, Rosenbaum DS. Targeted SERCA2a gene expression identifies molecular mechanism and therapeutic target for arrhythmogenic cardiac alternans. Circ Arrhythm Electrophysiol. 2009;2(6):686–694.[PMC free article] [PubMed] [Google Scholar]
109. del Monte F, Lebeche D, Guerrero JL, Tsuji T, Doye AA, Gwathmey JK, Hajjar RJ. Abrogation of ventricular arrhythmias in a model of ischemia and reperfusion by targeting myocardial calcium cycling. Proc Natl Acad Sci U S A. 2004;101(15):5622–5627.[PMC free article] [PubMed] [Google Scholar]
110. Prunier F, Kawase Y, Gianni D, Scapin C, Danik SB, Ellinor PT, Hajjar RJ, Del Monte F. Prevention of ventricular arrhythmias with sarcoplasmic reticulum Ca2+ ATPase pump overexpression in a porcine model of ischemia reperfusion. Circulation. 2008;118(6):614–624. [PubMed] [Google Scholar]
111. Hadri L, Bobe R, Kawase Y, Ladage D, Ishikawa K, Atassi F, Lebeche D, Kranias EG, Leopold JA, Lompré A-M, Lipskaia L, Hajjar RJ. SERCA2a Gene Transfer Enhances eNOS Expression and Activity in Endothelial Cells. Molecular Therapy. 2010[PMC free article] [PubMed] [Google Scholar]
112. del Monte F, Harding SE, Dec GW, Gwathmey JK, Hajjar RJ. Targeting phospholamban by gene transfer in human heart failure. Circulation. 2002;105(8):904–907.[PMC free article] [PubMed] [Google Scholar]
113. Suckau L, Fechner H, Chemaly E, Krohn S, Hadri L, Kockskamper J, Westermann D, Bisping E, Ly H, Wang X, Kawase Y, Chen J, Liang L, Sipo I, Vetter R, Weger S, Kurreck J, Erdmann V, Tschope C, Pieske B, Lebeche D, Schultheiss HP, Hajjar RJ, Poller WC. Long-term cardiac-targeted RNA interference for the treatment of heart failure restores cardiac function and reduces pathological hypertrophy. Circulation. 2009;119(9):1241–1252.[PMC free article] [PubMed] [Google Scholar]

Pathophysiology Of Heart Disease Pdf

114. Nicolaou P, Rodriguez P, Ren X, Zhou X, Qian J, Sadayappan S, Mitton B, Pathak A, Robbins J, Hajjar RJ, Jones K, Kranias EG. Inducible Expression of Active Protein Phosphatase-1 Inhibitor-1 Enhances Basal Cardiac Function and Protects Against Ischemia/Reperfusion Injury. Circ Res. 2009[PMC free article] [PubMed] [Google Scholar]
115. Pathak A, Baldwin B, Kranias EG. Key protein alterations associated with hyperdynamic cardiac function: insights based on proteomic analysis of the protein phosphatase 1 inhibitor-1 overexpressing hearts. Hellenic J Cardiol. 2007;48(1):30–36. [PubMed] [Google Scholar]
116. Pathak A, del Monte F, Zhao W, Schultz JE, Lorenz JN, Bodi I, Weiser D, Hahn H, Carr AN, Syed F, Mavila N, Jha L, Qian J, Marreez Y, Chen G, McGraw DW, Heist EK, Guerrero JL, DePaoli-Roach AA, Hajjar RJ, Kranias EG. Enhancement of cardiac function and suppression of heart failure progression by inhibition of protein phosphatase 1. Circ Res. 2005;96(7):756–766. [PubMed] [Google Scholar]
117. Most P, Pleger ST, Völkers M, Heidt B, Boerries M, Weichenhan D, Löffler M, Janssen PML, Eckhart AD, Martini J, Williams ML, Katus H, Remppis A, Koch WJ. Cardiac adenoviral S100A1 gene delivery rescues failing myocardium. Journal of Clinical Investigation. 2004;114:1550–1563.[PMC free article] [PubMed] [Google Scholar]
118. Pleger ST, Most P, Boucher M, Soltys S, Chuprun JK, Pleger W, Gao E, Dasgupta A, Rengo G, Remppis A, Katus HA, Eckhart AD, Rabinowitz JE, Koch WJ. Stable myocardial-specific AAV6-S100A1 gene therapy results in chronic functional heart failure rescue. Circulation. 2007;115(19):2506–2515. [PubMed] [Google Scholar]
119. Most P, Koch WJ. S100A1: a calcium-modulating inotropic prototype for future clinical heart failure therapy. Future Cardiol. 2007;3(1):5–11. [PubMed] [Google Scholar]
120. Kho C, Lee A, Jeong D, Oh JG, Chaanine AH, Kizana E, Park WJ, Hajjar RJ. SUMO1-dependent modulation of SERCA2a in heart failure. Nature. 2011;477(7366):601–605.[PMC free article] [PubMed] [Google Scholar]
121. Ghadge SK, Muhlstedt S, Ozcelik C, Bader M. SDF-1alpha as a therapeutic stem cell homing factor in myocardial infarction. Pharmacol Ther. 2011;129(1):97–108. [PubMed] [Google Scholar]
122. Kawase Y, Ladage D, Hajjar RJ. Rescuing the failing heart by targeted gene transfer. J Am Coll Cardiol. 2011;57(10):1169–1180.[PMC free article] [PubMed] [Google Scholar]
123. Pyo RT, Sui J, Dhume A, Palomeque J, Blaxall BC, Diaz G, Tunstead J, Logothetis DE, Hajjar RJ, Schecter AD. CXCR4 modulates contractility in adult cardiac myocytes. J Mol Cell Cardiol. 2006;41(5):834–844.[PMC free article] [PubMed] [Google Scholar]
124. Chen J, Chemaly E, Liang L, Kho C, Lee A, Park J, Altman P, Schecter AD, Hajjar RJ, Tarzami ST. Effects of CXCR4 gene transfer on cardiac function after ischemia-reperfusion injury. Am J Pathol. 2010;176(4):1705–1715.[PMC free article] [PubMed] [Google Scholar]
125. LaRocca TJ, Schwarzkopf M, Altman P, Zhang S, Gupta A, Gomes I, Alvin Z, Champion HC, Haddad G, Hajjar RJ, Devi LA, Schecter AD, Tarzami ST. beta2-Adrenergic receptor signaling in the cardiac myocyte is modulated by interactions with CXCR4. J Cardiovasc Pharmacol. 2010;56(5):548–559.[PMC free article] [PubMed] [Google Scholar]
126. Cottage CT, Bailey B, Fischer KM, Avitable D, Collins B, Tuck S, Quijada P, Gude N, Alvarez R, Muraski J, Sussman MA. Cardiac progenitor cell cycling stimulated by pim-1 kinase. Circ Res. 2010;106(5):891–901.[PMC free article] [PubMed] [Google Scholar]
127. Fischer KM, Cottage CT, Wu W, Din S, Gude NA, Avitabile D, Quijada P, Collins BL, Fransioli J, Sussman MA. Enhancement of myocardial regeneration through genetic engineering of cardiac progenitor cells expressing Pim-1 kinase. Circulation. 2009;120(21):2077–2087.[PMC free article] [PubMed] [Google Scholar]
128. Matsui T, Tao J, del Monte F, Lee KH, Li L, Picard M, Force TL, Franke TF, Hajjar RJ, Rosenzweig A. Akt activation preserves cardiac function and prevents injury after transient cardiac ischemia in vivo. Circulation. 2001;104(3):330–335. [PubMed] [Google Scholar]
129. Fomicheva EV, Turner II, Edwards TG, Hoff J, Arden E, D’Alecy LG, Metzger JM. Double oxygen-sensing vector system for robust hypoxia/ischemia-regulated gene induction in cardiac muscle in vitro and in vivo. Mol Ther. 2008;16(9):1594–1601.[PMC free article] [PubMed] [Google Scholar]
130. Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, Viswanathan A, Holder GE, Stockman A, Tyler N, Petersen-Jones S, Bhattacharya SS, Thrasher AJ, Fitzke FW, Carter BJ, Rubin GS, Moore AT, Ali RR. Effect of gene therapy on visual function in Leber’s congenital amaurosis. N Engl J Med. 2008;358(21):2231–2239. [PubMed] [Google Scholar]
131. Hajjar RJ, Zsebo K, Deckelbaum L, Thompson C, Rudy J, Yaroshinsky A, Ly H, Kawase Y, Wagner K, Borow K, Jaski B, London B, Greenberg B, Pauly DF, Patten R, Starling R, Mancini D, Jessup M. Design of a phase 1/2 trial of intracoronary administration of AAV1/SERCA2a in patients with heart failure. J Card Fail. 2008;14(5):355–367. [PubMed] [Google Scholar]
132. Zincarelli C, Soltys S, Rengo G, Rabinowitz JE. Analysis of AAV Serotypes 1-9 Mediated Gene Expression and Tropism in Mice After Systemic Injection. Mol Ther. 2008 [PubMed] [Google Scholar]
133. Wang Z, Zhu T, Qiao C, Zhou L, Wang B, Zhang J, Chen C, Li J, Xiao X. Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nat Biotechnol. 2005;23(3):321–328. [PubMed] [Google Scholar]
134. Pacak CA, Mah CS, Thattaliyath BD, Conlon TJ, Lewis MA, Cloutier DE, Zolotukhin I, Tarantal AF, Byrne BJ. Recombinant adeno-associated virus serotype 9 leads to preferential cardiac transduction in vivo. Circ Res. 2006;99(4):e3–9. [PubMed] [Google Scholar]
135. Inagaki K, Fuess S, Storm TA, Gibson GA, McTiernan CF, Kay MA, Nakai H. Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Mol Ther. 2006;14(1):45–53.[PMC free article] [PubMed] [Google Scholar]
136. Yang L, Jiang J, Drouin LM, Agbandje-McKenna M, Chen C, Qiao C, Pu D, Hu X, Wang DZ, Li J, Xiao X. A myocardium tropic adeno-associated virus (AAV) evolved by DNA shuffling and in vivo selection. Proc Natl Acad Sci U S A. 2009;106(10):3946–3951.[PMC free article] [PubMed] [Google Scholar]
137. Zincarelli C, Soltys S, Rengo G, Koch WJ, Rabinowitz JE. Comparative cardiac gene delivery of adeno-associated virus serotypes 1-9 reveals that AAV6 mediates the most efficient transduction in mouse heart. Clin Transl Sci. 2010;3(3):81–89.[PMC free article] [PubMed] [Google Scholar]
138. Du L, Kido M, Lee DV, Rabinowitz JE, Samulski RJ, Jamieson SW, Weitzman MD, Thistlethwaite PA. Differential myocardial gene delivery by recombinant serotype-specific adeno-associated viral vectors. Mol Ther. 2004;10(3):604–608. [PubMed] [Google Scholar]
139. Palomeque J, Chemaly ER, Colosi P, Wellman JA, Zhou S, Del Monte F, Hajjar RJ. Efficiency of eight different AAV serotypes in transducing rat myocardium in vivo. Gene Ther. 2007;14(13):989–997. [PubMed] [Google Scholar]
140. Pepe M, Mamdani M, Zentilin L, Csiszar A, Qanud K, Zacchigna S, Ungvari Z, Puligadda U, Moimas S, Xu X, Edwards JG, Hintze TH, Giacca M, Recchia FA. Intramyocardial VEGF-B167 gene delivery delays the progression towards congestive failure in dogs with pacing-induced dilated cardiomyopathy. Circ Res. 2010;106(12):1893–1903.[PMC free article] [PubMed] [Google Scholar]
141. Bell CL, Vandenberghe LH, Bell P, Limberis MP, Gao GP, Van Vliet K, Agbandje-McKenna M, Wilson JM. The AAV9 receptor and its modification to improve in vivo lung gene transfer in mice. J Clin Invest. 2011;121(6):2427–2435.[PMC free article] [PubMed] [Google Scholar]
142. Niemeyer GP, Herzog RW, Mount J, Arruda VR, Tillson DM, Hathcock J, van Ginkel FW, High KA, Lothrop CD., Jr. Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood. 2009;113(4):797–806.[PMC free article] [PubMed] [Google Scholar]
143. Wang Z, Kuhr CS, Allen JM, Blankinship M, Gregorevic P, Chamberlain JS, Tapscott SJ, Storb R. Sustained AAV-mediated dystrophin expression in a canine model of Duchenne muscular dystrophy with a brief course of immunosuppression. Mol Ther. 2007;15(6):1160–1166. [PubMed] [Google Scholar]
144. Sipo I, Fechner H, Pinkert S, Suckau L, Wang X, Weger S, Poller W. Differential internalization and nuclear uncoating of self-complementary adeno-associated virus pseudotype vectors as determinants of cardiac cell transduction. Gene Ther. 2007;14(18):1319–1329. [PubMed] [Google Scholar]
145. Arruda VR, Stedman HH, Haurigot V, Buchlis G, Baila S, Favaro P, Chen Y, Franck HG, Zhou S, Wright JF, Couto LB, Jiang H, Pierce GF, Bellinger DA, Mingozzi F, Nichols TC, High KA. Peripheral transvenular delivery of adeno-associated viral vectors to skeletal muscle as a novel therapy for hemophilia B. Blood. 2010;115(23):4678–4688.[PMC free article] [PubMed] [Google Scholar]
146. Wang T, Hou LB, Liu ZJ, Wang Y, Chen CL, Xiao X, Wang DW. Intramuscular delivery of rAAV-mediated kallikrein gene reduces hypertension and prevents cardiovascular injuries in model rats. Acta Pharmacol Sin. 2007;28(12):1898–1906. [PubMed] [Google Scholar]
147. Wang J, Faust SM, Rabinowitz JE. The next step in gene delivery: Molecular engineering of adeno-associated virus serotypes. J Mol Cell Cardiol. 2010 [PubMed] [Google Scholar]
148. Qiao C, Li J, Jiang J, Zhu X, Wang B, Xiao X. Myostatin propeptide gene delivery by adeno-associated virus serotype 8 vectors enhances muscle growth and ameliorates dystrophic phenotypes in mdx mice. Hum Gene Ther. 2008;19(3):241–254. [PubMed] [Google Scholar]
149. Yue Y, Ghosh A, Long C, Bostick B, Smith BF, Kornegay JN, Duan D. A single intravenous injection of adeno-associated virus serotype-9 leads to whole body skeletal muscle transduction in dogs. Mol Ther. 2008;16(12):1944–1952.[PMC free article] [PubMed] [Google Scholar]
150. Su H, Huang Y, Takagawa J, Barcena A, Arakawa-Hoyt J, Ye J, Grossman W, Kan YW. AAV serotype-1 mediates early onset of gene expression in mouse hearts and results in better therapeutic effect. Gene Ther. 2006;13(21):1495–1502. [PubMed] [Google Scholar]
151. Tao Z, Chen B, Tan X, Zhao Y, Wang L, Zhu T, Cao K, Yang Z, Kan YW, Su H. Coexpression of VEGF and angiopoietin-1 promotes angiogenesis and cardiomyocyte proliferation reduces apoptosis in porcine myocardial infarction (MI) heart. Proc Natl Acad Sci U S A. 2011;108(5):2064–2069.[PMC free article] [PubMed] [Google Scholar]
152. Jacquier A, Higgins CB, Martin AJ, Do L, Saloner D, Saeed M. Injection of adeno-associated viral vector encoding vascular endothelial growth factor gene in infarcted swine myocardium: MR measurements of left ventricular function and strain. Radiology. 2007;245(1):196–205. [PubMed] [Google Scholar]
153. Kaspar BK, Roth DM, Lai NC, Drumm JD, Erickson DA, McKirnan MD. Hammond HK. Myocardial gene transfer and long-term expression following intracoronary delivery of adeno-associated virus. J Gene Med. 2005;7(3):316–324. [PubMed] [Google Scholar]
Posted :