Skip to main content

Resistant hypertension: consensus document from the Korean society of hypertension

Abstract

Although reports vary, the prevalence of true resistant hypertension and apparent treatment-resistant hypertension (aTRH) has been reported to be 10.3% and 14.7%, respectively. As there is a rapid increase in the prevalence of obesity, chronic kidney disease, and diabetes mellitus, factors that are associated with resistant hypertension, the prevalence of resistant hypertension is expected to rise as well. Frequently, patients with aTRH have pseudoresistant hypertension [aTRH due to white-coat uncontrolled hypertension (WUCH), drug underdosing, poor adherence, and inaccurate office blood pressure (BP) measurements]. As the prevalence of WUCH is high among patients with aTRH, the use of out-of-office BP measurements, both ambulatory blood pressure monitoring (ABPM) and home blood pressure monitoring (HBPM), is essential to exclude WUCH. Non-adherence is especially problematic, and methods to assess adherence remain limited and often not clinically feasible. Therefore, the use of HBPM and higher utilization of single-pill fixed-dose combination treatments should be emphasized to improve drug adherence. In addition, primary aldosteronism and symptomatic obstructive sleep apnea are quite common in patients with hypertension and more so in patients with resistant hypertension. Screening for these diseases is essential, as the treatment of these secondary causes may help control BP in patients who are otherwise difficult to treat. Finally, a proper drug regimen combined with lifestyle modifications is essential to control BP in these patients.

Graphical Abstract

Introduction

High blood pressure (BP) is associated with increased cardiovascular complications, regardless of the type of prescribed antihypertensive medications [1]. BP reduction at any level has shown a 10% reduction in cardiovascular events per 5 mmHg reduction in systolic BP. In addition to population-based strategies for identifying patients with untreated hypertension and introducing antihypertensive therapy, BP reduction to recommended levels is important for reducing cardiovascular risk and optimizing antihypertension treatment.

In Korea, the overall control rate of hypertension is less than half of the hypertensive population, and the control rate in patients treated for hypertension is approximately 70% [2]. Patients with resistant hypertension (RH) are particularly challenging in terms of the reasons for their reduced response to multiple antihypertensive medications, clinical presentation, specific etiology, prognosis, and management. Understanding the underlying etiology and pathophysiology of uncontrolled hypertension treated with multiple antihypertensive medications will help achieve a breakthrough in the stagnant BP control rate [3]. Accordingly, the Korean Society of Hypertension has published an expert consensus on the definition, epidemiology, etiology, diagnosis, and management of RH.

Definition of resistant hypertension

RH is defined as the failure to achieve the target BP despite the use of ≥3 antihypertensive drugs, commonly including dihydropyridine calcium channel blockers(CCBs), renin-angiotensin system (RAS) inhibitors, and diuretics, or the need for treatment with ≥4 antihypertensive medications to achieve the target BP [4, 5]. Patients who fail to achieve the target BP despite the use of five or more antihypertensive medications, ideally including thiazide-like diuretics and spironolactone, are defined as having treatment-refractory hypertension [6, 7]. The target BP should be in accordance with the current guidelines. For example, the threshold for diagnosis is 130/80 mmHg according to the American College of Cardiology/American Heart Association guidelines [4]. For Korean patients, the threshold may differ based on underlying risk factors [5, 8]. Previous studies included patients who were diagnosed with hypertension based on office BP as well as patients who would be categorized as having pseudoresistant hypertension. Therefore, the term “apparent treatment RH” (aTRH) should be used for patients in whom pseudoresistant hypertension has not been ruled out. The causes of pseudoresistant hypertension include white-coat uncontrolled hypertension (WUCH), non-adherence, poorly measured office BP, or undertreatment [4]. For an accurate diagnosis of RH, out-of-office BP monitoring, such as 24-hour ambulatory blood pressure monitoring (ABPM) and/or home blood pressure monitoring (HBPM), should be performed to rule out WUCH. This is essential because WUCH impacts as many as 1/3 of patients with aTRH [9]. Moreover, the possibility of non-adherence needs to be ruled out, as it is common, especially with polypharmacy and a higher number of anti- hypertensive medications [10]. Finally, antihypertensive medications, including thiazide-like or thiazide-type diuretics, must be titrated to maximally tolerated doses before making a diagnosis.

Epidemiology of resistant hypertension

The reported prevalence of RH varies significantly according to specific studies. In the 2018 AHA scientific statement, the prevalence of RH among all patients with hypertension was 12-15% based on population studies and 15-18% for clinic based reports [4]. In a pooled analysis of 3,207,911 patients with hypertension, the prevalence rates of true and aTRH were 10.3% and 14.7%, respectively [11]. Age, higher baseline BP, obesity, excessive salt ingestion, chronic kidney disease (CKD) and diabetes mellitus (DM) were associated with a higher risk of RH [12]. The prevalence of RH increases with age. Several factors contribute to the higher risk of RH in older adults [13, 14]. They include age-related vascular changes (i.e., vascular stiffness), neurohormone imbalances, multiple comorbidities (including kidney disease, obesity, and diabetes), poor adherence to medication or polypharmacy, and insufficient lifestyle modifications (especially high-salt diets) [13, 14].

Noubiap et al. analyzed the prevalence of aTRH by region [11]. They found that the prevalence of aTRH did not differ by region, with the prevalence of RH in the Americas, Asia, and Europe being 14.8%, 14.7%, and 14.8%, respectively. The prevalence of treatment RH in cohorts that underwent out-of-office BP monitoring tended to be lower than that reported previously. In a Korean study that recruited 3088 patients with essential hypertension from 247 primary care physicians, the prevalence of RH was 7.9% [15]. In another study involving 16,284 Korean patients with hypertension who underwent ABPM at a single tertiary referral hospital, the overall prevalence of RH was 10.1%. Within this group, the prevalence of RH (excluding refractory hypertension) was 9.2%, and refractory hypertension accounted for 0.9% of the cases [16]. The prevalence of RH defined by home BP was recently reported in an analysis of the Japan Morning Surge Home BP (J-HOP) study. Using the average of home BP threshold of ≥135/85 mmHg and ≥130/80 mmHg, the prevalence of RH was 10.7% and 12.5%, respectively [17].

RH is associated with an adverse prognosis compared with patients without RH. In a retrospective cohort of 205,750 patients with incident hypertension over a median follow-up of 3.8 years, RH was associated with an approximately 50% higher risk of cardiovascular events[hazard ration (HR):1.47, 95% confidence interval (CI):1.33-1.62] [18]. In a post hoc analysis of 14,684 patients from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT), 1870 patients were defined as having aTRH. The results showed that aTRH was associated with an increased risk of coronary heart disease(HR:1.47, 95% CI:1.26-1.71), cardiovascular disease(HR:1.46, 95% CI:1.29-1.64) and end stage renal disease(HR:1.95, 95% CI:1.26-1.71) [19]. In a retrospective analysis of 16,284 Korean patients who underwent ABPM, RH was associated with a 62% higher risk of cardiovascular mortality(HR:1.62, 95% CI:1.16-2.26) [16]. Similarly, RH based on home BP measurement is associated with an adverse prognosis. In an analysis of 4,261 patients with hypertension from the J-HOP study with a median follow-up of 6.2 years, the adjusted HRs of uncontrolled treatment RH were 2.02(1.38-2.94) and 1.81(1.23-2.65) when using a home BP threshold of 135/85 mmHg and 130/80 mmHg, respectively [17]. Use of out-of-office BP monitoring, such as ABPM or HBPM, for the diagnosis and treatment of RH is important, as it can better predict cardiovascular outcomes in patients with RH [20].

Evaluation of etiology

Diagnosis of resistant hypertension

RH can be diagnosed by ruling out pseudoresistance, which is defined as a lack of BP control despite appropriate treatment in patients who do not have RH. Several factors, such as improper BP measurement, the “white-coat effect,” medication non-adherence, and treatment inertia, contribute to pseudoresistance. According to current guidelines, accurate BP measurement is the cornerstone of RH diagnosis [21, 22]. The “white-coat effect” should be excluded when using ABPM or HBPM [23]. Medication non-adherence is common in patients with aTRH [24]. Treatment inertia is one reason why a large subset of patients do not achieve their BP target. Between 2007 and 2010, less than 50% of patients were prescribed an optimal antihypertensive regimen in the United States [23]. Therefore, a careful evaluation to exclude these factors should be performed before diagnosing RH. It is also important to rule out the possibility of pseudoresistance due to the white-coat effect in the diagnosis of RH in older adults, as this phenomenon is more common [25].

Lifestyle factors contributing resistant hypertension (Table 1)

Table 1 Factors causing high blood pressure in resistant hypertension

Obesity

Increased adiposity is associated with elevated BP [26]. Obesity-related hypertension is associated with increased salt sensitivity, increased sympathetic nervous system activity, activation of the renin-angiotensin-aldosterone system(RAS), and aldosterone secretion by adipose tissue [26, 27]. In NHANES (National Health and Nutrition Examination Survey), body mass index ≥30 kg/m2 doubled the risk to have aTRH [28]. However, no current guidelines recommend the use of specific drugs in patients with obesity.

Dietary sodium

Dietary sodium increases BP, particularly in certain groups that are more salt-sensitive or have CKD [29]. Despite large person-to-person variations in salt sensitivity, vascular dysfunction, arterial stiffness, sympathetic activation, impaired renin-angiotensin axis suppression, mineralocorticoid receptor activation, and immune cell modulation are suggested as possible mechanisms [30, 31]. One study showed that reducing dietary sodium from 250 mmol (5.75g)/day to 50 mmol (1.15g)/day for 1-week lowered office systolic BP by 22.7 mmHg (P=0.008) in patients with RH [32]. Patients should be advised to adhere to the DASH (Dietary Approaches to Stop Hypertension) diet with less than 2 g of sodium/day (5 g of table salt) to effectively lower BP [29].

Physical inactivity

Physical inactivity is independently associated with BP elevation and risk of hypertension [33]. However, it is uncertain whether sedentary life itself is independently related to RH because of a paucity of data [34]. Recently, a small randomized trial (n=50) showed that a thrice-weekly treadmill exercise program for 8–12 weeks lowered daytime systolic ambulatory BP by 5.9 mmHg (P=0.03) in patients with RH [34]. Patients with RH should engage in at least 150 min/week of moderate-intensity aerobic exercise or 75 min/week of vigorous-intensity aerobic exercise.

Alcohol

Regular alcohol consumption is associated with a BP elevation of 1 mmHg for every 10 g of alcohol consumed (≈1 standard drink) [35]. Heavy alcohol use may be associated with aTRH, since alcohol may interfere with the pharmacologic action of antihypertensive agents [36].

Smoking

Nicotine from cigarettes, vaping fluids, and smokeless tobacco increases BP [37]. To reduce cardiovascular events, smoking cessation is recommended for patients with RH.

Medication-related resistant hypertension

Some classes of medications can increase BP, including anticonvulsants, antidepressants, antipsychotics, antiemetics, antineoplastic agent, calcineurin inhibitors, contraceptives, erythropoietin, glucocorticoids, mineralocorticoids, nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, estrogen-containing oral contraceptives and hormone replacement, sympathomimetic amines (pseudoephedrine, ephedrine, cocaine, amphetamine), vascular endothelial growth factor inhibitors, erythropoietin-stimulating agents, immunosuppressive agents (cyclosporine, tacrolimus), anticancer drugs(vascular endothelial growth factor inhibitors, tyrosine kinase inhibitors and proteasome inhibitors), serotonin-norepinephrine reuptake inhibitors, and dietary supplements (ginseng, licorice) [4, 38, 39]. The effect of these drugs on increasing BP varies from person to person, ranging from little to no effect to severely elevated BP. As some medications such as NSAIDs, cold medications, and oral contraceptives can be purchased and taken by patients without a prescription, medication reconciliation is an important process in the management of patients with hypertension [40].

Pseudopheochromocytoma

After true pheochromocytoma has been ruled out as the cause of the sudden increase in BP, pseudopheochromocytoma should be considered. Pseudopheochromocytoma is a pheochromocytoma-like syndrome characterized by paroxysmal hypertension, abrupt elevation of BP, and the absence of panic at the onset of attack [41]. Many patients with pseudopheochromocytoma suffer from sleep disorders such as poor sleep quality due to activation of the sympathetic nervous system and RAS [42]. Because the quality and quantity of sleep are critical issues for BP elevation in patients with RH, clinicians must consider these issues. Clinical evidence suggests that rapid heart rate that is disproportionate to patients’ BP is also associated with sleep quality [43].

Secondary hypertension

Secondary hypertension should be evaluated to identify its cause and appropriate treatment in patients with RH (Tables 2 and 3).

Table 2 Secondary causes of hypertension
Table 3 Laboratory examination of secondary hypertension

Chronic kidney disease

CKD develops into hypertension via various pathogenesis. The upregulated RAS, increased salt and fluid retention, endothelial dysfunction, and increased sympathetic nervous system activity have been suggested as mechanisms of hypertension in CKD [44]. In a previous cohort study, more than 85% of patients with chronic renal disease had hypertension at baseline [45]. Underlying renal parenchymal diseases, such as glomerulonephritis, can also cause hypertension through a similar pathogenesis [46]. Hypertension is a common manifestation of focal segmental glomerulosclerosis, membranous nephropathy, immunoglobulin A nephropathy and membranoproliferative glomerulonephritis [47]. Therefore, kidney disease should be evaluated and considered a risk factor for RH.

Primary aldosteronism

Excess aldosterone secretion in primary aldosteronism leads to salt and water retention and renal potassium wasting, resulting in hypertension [48]. Primary aldosteronism is more common than expected, and its prevalence varies from 8 to 30% [49]. Plasma aldosterone-to-renin ratio is the most commonly used screening test [50]. Although there is the potential for false-positive and false-negative results depending on the situation (medications interfering with the RAS, the cutoff values used, the time of testing, and the body positioning at the time of testing), further investigation is required if the aldosterone-to-renin ratio exceeds 20 [48].

Obstructive sleep apnea

Obstructive sleep apnea (OSA) is very common, with prevalence rates of 70 - 90% in patients with RH [51]. The high prevalence rates have been attributed to endothelial reactivity, inflammation, oxidative stress, and increased sympathetic and renin-angiotensin-aldosterone system activities, which ultimately lead to increased vascular tone and hypertension [4]. In addition, excess aldosterone and high dietary sodium intake can increase fluid retention and upper airway edema, accompanied by increased airway resistance, thereby worsening OSA [52, 53]. Shifting of fluid from the lower extremities to the neck during supine sleep has been shown to contribute to upper airway edema [54]. This effect is blunted by use of spironolactone [55]. Although routine surveillance by polysomnography is not indicated for all patients with RH, they should be thoroughly screened if OSA is present [56].

Renovascular hypertension

Renovascular hypertension (RVH) is a syndrome in which BP is elevated due to renal artery stenosis, leading to renal ischemia. RVH is one of the most common causes of RH, particularly in the elderly [57], with up to 35% of cases being a secondary cause of hypertension [58]. Most cases are caused by atherosclerosis of the renal arteries; however, RVH syndrome can also result from less common obstructive lesions, such as fibromuscular dysplasia, renal artery infarct or dissection, Takayasu arteritis, radiation fibrosis, and renal artery obstruction by an aortic endovascular stent graft [4]. Renal artery stenosis is diagnosed using duplex ultrasonography, computed tomography angiography, or magnetic resonance angiography.

Catecholamine-secreting tumors

Catecholamine-secreting tumors, such as pheochromocytomas and paragangliomas, are rare in hypertensive populations accounting for 0.01–0.2% [4]. The prevalence rate is up to 4% in patients referred for RH [59]. Although the prevalence of catecholamine-secreting tumors is low, the mortality and morbidity are high when untreated [60]. Therefore, the diagnoses of patients referred for RH must be considered [61]. Current Endocrine Society guidelines recommend screening by measuring either plasma free (sensitivity, 96-100%; specificity, 89-98%) or 24-hour urine fractionated (sensitivity, 96-100%; specificity, 89-98%) metanephrines. Imaging techniques, such as computed tomography, magnetic resonance imaging, and metaiodobenzylguanidine scanning, should be performed only after biochemical evidence of pheochromocytoma has been obtained [62].

Cushing disease or syndrome

Cushing syndrome is a relatively uncommon cause of RH caused by hypercortisolism due to glucocorticoid excess. Cushing syndrome is a constellation of classic symptoms (mood disorders, menstrual changes, and muscle wasting) and signs (acne, osteoporosis, hirsutism, weight gain, moon facies, and supraclavicular fat). However, since 26.5% of patients with Cushing syndrome with biochemical evidence of hypercortisolism do not have overt symptoms [63], clinicians should consider screening tests in patients with RH, even if they do not have the classic syndrome. Current guidelines recommend screening by measuring the 24-hour urine cortisol level, late-night salivary cortisol level, or low-dose dexamethasone suppression test.

Other endocrinopathies

Less common endocrine disorders that contribute to RH include thyroid and parathyroid glands [4]. Thyroid-stimulating hormone (TSH) levels should be assessed in patients with difficult-to-control hypertension. Testing for primary hyperparathyroidism should be considered in patients presenting with hypercalcemia.

Blood pressure measurements

Pseudo-resistant hypertension

Many cases of uncontrolled hypertension are not true RH. Despite the use of ≥3 antihypertensive drugs, BP levels may be misrepresented as uncontrolled hypertension—this is known as pseudo-resistant hypertension [4]. The prevalence of pseudoresistant hypertension among patients with aTRH has been estimated to be as high as 50% [64]. The most common causes of pseudoresistance are poor BP measurement, the white-coat effect (WUCH), medication non-adherence, and under-treatment [4]. Thus, physicians should consider excluding pseudo-resistant hypertension before confirming true RH [4].

Improper blood pressure measurement technique

Accurate BP measurements can exclude RH. Common errors during routine office BP measurements include not resting for 5 minutes before BP measurement in a quiet room, not using validated devices, not using the correctly sized cuff, not placing the cuff at the heart level, not obtaining a minimum of two readings 1 min apart, deflating the cuff too fast or too slow, observer bias, and auscultatory gap in the auscultation technique [4, 63]. Bhatt et al. reported that improper BP techniques overestimated the prevalence of uncontrolled RH in approximately 33% of patients [64]. In patients with advanced age and atherosclerotic disease, cuff pressure is elevated compared with true intra-arterial measurements; this so-called pseudo-hypertension has been described as a cause of RH. A positive Osler sign, which is a palpable radial pulse while the BP cuffs are inflated above the systolic BP, can be used as a simple screening tool for pseudohypertension. A definitive diagnosis is made by comparing the intra-arterial pressure with the cuff pressure [4].

Ambulatory Blood Pressure Monitoring (ABPM)

Out-of-office BP measurements, including ABPM and HBPM, are recommended to exclude true RH [4]. ABPM has been indicated as an essential tool for RH in the following clinical conditions: (1) confirmation of the diagnosis of RH, (2) detection of WUCH, (3) assessment of non-dipping patterns, (4) estimation of cardiovascular prognosis, and (5) assessment of treatment effectiveness in RH [4]. The Spanish ABPM Registry reported that RH was present in 12% of the treated hypertensive population, but more than one-third had normal ambulatory BP (WUCH) [4, 9]. Since non-dipping patterns are commonly observed in patients with RH and nighttime BP levels and dipping patterns provide prognostic value, information on nocturnal BP is important for the diagnosis and monitoring of RH [4]. Claudia et. al. reported that the mean cumulative ambulatory BPs during follow-up compared with the baseline BPs was the best prognostic marker of adverse cardiovascular outcomes and mortality in patients with RH. Therefore, serial ABPM examinations are widely used in RH management [20].

Home Blood Pressure Monitoring (HBPM)

HBPM is the best method for diagnosing WUCH and is more convenient, less disruptive, and more cost-effective than ABPM [4]. Some patients are sensitive to ABPM and may experience increased BP readings due to the test itself. Furthermore, ABPM has been linked to insomnia and decreased compliance in some individuals [4]. By contrast, HBPM provides more accurate readings than BP measured in a medical setting and has similar accuracy to ABPM in diagnosing RH [4]. However, HBPM may have limitations in measuring nighttime BP. With recent technological advancements, it is becoming increasingly possible to accurately measure night-time BP at home, and the role of HBPM in the diagnosis of RH is expected to grow [4]. HBPM is a reliable alternative to ABPM for diagnosing RH.

White Coat Uncontrolled Hypertension (WUCH)

WUCH is a term used to describe a condition in which a patient’s BP is elevated when measured in a medical setting (such as a doctor’s office) but is normal when measured outside of that setting (such as at home or during daily activities) despite taking antihypertensive medications [8]. WUCH is relatively common, and its prevalence has been reported to be 30-40% in Western countries [9, 65] and 13.5% in Korea [66]. Since WUCH is a major cause of pseudo-resistant hypertension and the white-coat effect is exaggerated in patients with RH [9, 65], its identification is a key factor in the proper diagnosis and planning of the treatment of RH. In the Spanish ABPM Registry, among 8,295 patients diagnosed with RH based on office BP, only 62.5% were classified as having true RH, and the remaining 37.5% had WUCH [9, 65]. Approximately one-third of patients with suspected RH have WUCH [67, 68]. Cardiovascular risk does not increase in patients with WUCH [69]. If WUCH is not excluded and a patient is diagnosed with RH based only on elevated BP measurements in the medical setting, it may lead to unnecessary treatment of high BP with medication, which can have side effects. An accurate diagnosis of WUCH requires out-of-office BP measurements like HBPM or ABPM [4].

Evaluation of resistant hypertension

Medical history

A detailed and comprehensive medical history is essential for evaluating patients with RH as it can provide valuable information for the selection of appropriate treatment strategies. When RH is suspected, the first step is to confirm proper antihypertensive medication adherence by determining the appropriate dose and frequency of administration. Medication adherence can be assessed by checking how often antihypertensive medications are skipped or using self-reported medication adherence assessment tools. If drug discontinuation is confirmed, it is important to identify the reasons for discontinuation, such as side effects and ineffectiveness [70]. Lifestyle factors that may also contribute to elevated BP and should be assessed. These include physical inactivity, high salt intake, smoking, alcohol consumption, and OSA [4]. Healthcare providers should ask about any history of snoring, daytime sleepiness, or witnessed apneas, as OSA is a common comorbidity in patients with RH [51, 71]. Recent changes in weight, stress levels, or sleep patterns should be noted. It is also vital to consider medications that may elevate BP, such as NSAIDs, contraceptives, corticosteroids, and immunosuppressants like cyclosporine. Table 1 summarizes the key items to be checked during the medical examinations of patients with RH. A thorough evaluation is necessary to identify the underlying causes of secondary hypertension in patients with RH (Table 2) [4]. The likelihood of a secondary cause related to RH is higher among older adults. Physicians should conduct a comprehensive assessment to identify underlying causes or contributing factors, such as sleep apnea, renal parenchymal disease, renal artery stenosis, or primary aldosteronism [72].

Physical examination

During the physical examination of patients with RH, identifying evidence of target organ damage and secondary hypertension is important [4]. BP measurement must be standard and accurate. Coarctation of the aorta is suspected if the difference in BP between the upper and lower extremities is > 10 mm Hg. Evidence of vascular damage can be verified through fundus examination and stenosis of the carotid, and abdominal arteries can be assessed by auscultation of the bruits. Stenotic lesions of blood vessels can also be estimated by palpating the pulsations of the limb arteries. Cushing syndrome can be suspected if there is a moon face or abdominal obesity. If the limbs are enlarged, the lower jaw is lengthened, and the bridge of the nose shows characteristic findings, acromegaly should be suspected, and further examination should be performed.

Out-of-office blood pressure monitoring

White coat effect may be suspected when BP measurements taken in the clinic are higher than those taken at home, when there is no evidence of hypertension-mediated organ damage despite sustained high BP, and when hypotensive symptoms occur frequently with drug use. The most effective way to rule out the white-coat effect is to conduct out-of-office BP monitoring using ABPM and HBPM. If the white-coat effect is confirmed, out-of-office BP monitoring is recommended for future hypertension treatment [73]. HBPM is particularly useful for diagnosing and managing RH, as it is easy to measure, can be repeated over a long period, and facilitates medication adjustment and improved adherence [74].

Laboratory examination and imaging

Laboratory examinations for the evaluation of RH should include basic blood chemistry (serum sodium, potassium, chloride, bicarbonate, glucose, blood urea nitrogen, and creatinine with glomerular filtration rate(GFR and urinalysis. Plasma aldosterone and plasma renin activity should be evaluated to screen for primary aldosteronism. Interpretation of the aldosterone-to-renin ratio is difficult in patients who take certain antihypertensive medications, such as mineralocorticoid receptor antagonists, which increase aldosterone levels, or direct renin inhibitors and beta-blockers, which lower renin levels [75, 76]. The elevated aldosterone-to-renin ratio(ARR) show low specificity because low-renin states are common (eg, volume expansion, dietary salt excess or sensitivity). A high ratio (> 20) when the serum aldosterone is >16 ng/dL and PRA is <0.6 ng/mL per hour is suggestive of primary aldosteronism, particularly in a patient taking an ACE inhibitor or ARB, but further assessments are required to confirm this diagnosis. Measurement of plasma metanephrines or 24-hour urinary metanephrines is an effective screening test for patients with suspected paraganglioma or pheochromocytoma [77]. Thyroid function tests, such as free T4, T3, and TSH are useful for screening for hyperthyroidism and hypothyroidism [2].

Imaging tests for screening renovascular hypertension are performed in young patients with a likelihood of fibromuscular dysplasia and in older patients with a history of smoking or vascular disease who have an increased risk for atherosclerosis [2]. Screening tools for renovascular hypertension include a captopril renal scan, Doppler ultrasound, computed tomography, or magnetic resonance angiography. Patients with hypokalemia without an apparent etiology or an incidentally diagnosed adrenal mass should be evaluated for hyperaldosteronism. Paroxysmal and/or refractory hypertension with hyperadrenergic symptoms is indicative of a strong possibility of pheochromocytoma. Therefore, after the measurement of aldosterone, catecholamine, and cortisol levels in the plasma and/or 24-h urine, abdominal imaging of the adrenal glands by high-resolution computerized tomography, magnetic resonance imaging, or radioisotope imaging (I-131 metaiodobenzylguanidine) is indicated if there is biochemical evidence of hormonally active tumors [5].

Sleep apnea syndrome has been suggested to be a prevalent cause of secondary hypertension among obese patients or those with resistant hypertension. Although there is still no evidence of its benefits, screening for sleep apnea using a self-questionnaire or diagnosis using sleep polysomnography may be useful for the evaluation of RH [10].

Management of resistant hypertension

Confirming aTRH and screening diagnostic issues like non-adherence, accuracy of BP measurement, and the exclusion of temporary factors such as lifestyle factors, related drugs or herbal medication is critical when deciding upon referral to a specialist.

Adherence intervention

Non-adherence to antihypertensive medication is a key contributor to uncontrolled BP; previous studies have indicated that over 30% of adults taking antihypertensive medication have low adherence in the year following treatment initiation [78]. Meta-analysis of the prevalence of non-adherence among adults with aTRH in 42 studies with 71,353 participants showed an average prevalence of 37%, with wide variation across the studies ranging from 3–86% [79].

Antihypertensive medications can be assessed using both indirect and direct methods [80]. Indirect methods include questionnaires, diaries, pill counts, and prescription refill rates. Direct methods include directly observing therapy (i.e., watching people take each dose of medication) and measuring metabolite levels or biological markers [81]. Generally, the prevalence of non-adherence is substantially lower when adherence is ascertained using indirect methods (20%) compared with direct methods (46%) [79]. Practically, a single-pill combination of antihypertensive medications and statins is helpful for serially checking adherence by evaluating cholesterol levels. The direct method provides an accurate assessment of medication taken recently but does not capture long-term adherence and is not available in every center.

Several factors, including socioeconomic status, demographics, and environmental factors, are associated with suboptimal adherence. More importantly, the rapport between the patient and clinician, the communication style of the clinician, and the degree of shared decisions have all been shown to affect adherence [80]. Access to and cost of medications are clearly important for adherence, although the nearly full coverage of the national health insurance system in Korea has an advantage over other countries. Complex medication regimens, including polypharmacy and multiple daily doses, reduce adherence. Finally, adults with hypertension often have multiple chronic conditions, including depression and cognitive dysfunction, which can adversely affect their adherence to medications and healthy lifestyles.

Drug adherence is an important but often neglected factor in the evaluation of patients with RH. According to the American Heart Association, confirmation of RH requires the assurance of antihypertensive medication adherence [4]. This is due to the high prevalence of medication non-adherence in RH, which has been reported to be as high as 50%. In a study by Lawson et al., 300 patients with uncontrolled hypertension underwent a urine antihypertensive drug assay using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The study showed that 55% of the patients were nonadherent to the prescribed medications [82]. In a study of 76 patients with uncontrolled RH, 40 (53%) were found to be non-adherent [83]. The prevalence of non-adherence is similarly high in patients with refractory hypertension. In a study by Siddiqui et al., 54 patients with apparent refractory hypertension underwent a urine antihypertensive drug metabolite assay using high-performance liquid chromatography-tandem mass spectrometry. The results showed that of the 40 fully evaluated patients, 16(40.0%) were fully adherent, 18(45.0%) were partially adherent, and six (15.0%) were non-adherent [84]. Despite the importance of medication adherence, it is difficult to assess in clinical practice. The current limitation in assessing medication adherence is the lack of a standardized, clinically available method or test to accurately assess medication adherence. Pill counting, the most widely used method in clinical trials, is associated with an overestimation of drug adherence. Electronic monitoring and drug metabolite assays, either in the blood or urine, are investigational methods that are not widely available [85, 86]. Efforts to improve medication adherence are important. First, patient education to improve self-awareness of medical conditions and the need for proper ingestion of antihypertensive medications is important. Physician efforts to lower pill counts are also important, as studies have shown a significant inverse association between antihypertensive pill counts and medication adherence [87]. Several trials designed to reduce non-adherence to antihypertensive medications have been conducted, however, their effects vary. Although a trial of simple reminder pill bottles did not show an improvement in adherence, using electronic pill devices that trigger text message reminders in conjunction with missed doses shows promise in improving adherence [88]. Morawski et al. randomized patients with poorly controlled hypertension to usual care or a smartphone app that provided reminder alerts, adherence reports, and optional peer support and found that patients receiving the intervention had improved self-reported adherence, but the intervention did not affect BP control [89]. Lastly, simplifying treatment regimens by consolidating doses using fixed-dose combinations or extended-release formulations is reportedly effective in improving adherence [90]. Switching a patient prescriptions to a once-daily regimens using fixed-dose combinations to reduce the total pill count may help improve medication adherence in patients with RH.

Lifestyle factor intervention

Healthy lifestyles, including low sodium intake, regular exercise, ideal body weight, moderate alcohol intake, and non-smoking, are important for promoting public health and preventing the development of hypertension and cardiovascular events [22, 91]. All physicians managing RH should review patients’ lifestyles and recommend maintaining the following healthy lifestyles.

Diet

Dietary sodium intake is closely associated with BP [29]. Experimental and epidemiological studies have shown that increased salt consumption increases BP, resulting in incident hypertension and cardiovascular complications [92, 93]. Reducing sodium intake lowers BP [29]. The American Heart Association defines sodium sensitivity as a characteristic that affects BP according to changes in salt intake [94]. BP in sodium-sensitive individuals responds more sensitively to sodium intake than that in sodium-resistant individuals. Sodium sensitivity is higher in hypertensive populations than in general populations without hypertension, and higher in resistant hypertensive patients than in simple hypertensive individuals [94, 95]. Older adults and people with a higher level of BP or comorbidities, such as CKD and diabetes, tend to be more sensitive to sodium intake [22]. A randomized crossover trial reported that low-salt diet decreased systolic and diastolic BP by 22.7 and 9.1 mmHg, respectively, in patients with RH [32]. To control BP and prevent cardiovascular complications, salt and sodium restrictions are recommended for patients with RH.

Dietary patterns significantly affect the incidence of chronic diseases, including hypertension and diabetes. The Dietary Approaches to Stop Hypertension (DASH) trials emphasized that hypertensive people consume more fruits, vegetables, and low-fat dairy foods and less red meat, saturated or total fat, and sugar-containing beverages to reduce their BPs [96]. The DASH diet reduced systolic and diastolic BP by 5.5 and 3.0 mmHg, respectively [96].

For patients with RH, adopting a DASH diet and reducing salt (6 g/day) and sodium (2.4 gram/day) intake are recommended to control BP [8].

Physical activity

Low physical activity and fitness levels were independent risk factors for hypertension. Physical activity is inversely associated with the development of hypertension [97]. Physical activity reduces BP in both hypertensive and normotensive populations, independent of weight loss [98]. In a meta-analysis, BP lowered by approximately 8.3 mmHg and 5.2 mmHg in systolic and diastolic BP over several endurance (aerobic) exercise programs in hypertensive population [99]. Resistance (muscle strengthening) exercises also reduce BP in patients with an elevated BP [99]. Regular physical activity is inversely associated with cardiovascular and/or all-cause mortality in patients with elevated BP [100]. Thus, physical activity is beneficial for reducing mortality in hypertensive populations in addition to BP reduction. Clinical practice guidelines for hypertension from Europe, America, and Asia commonly recommend regular physical activity [8, 22, 91]. However, there are few studies for patients with RH to follow current exercise recommendations. In patients with RH, regular aerobic exercise based on a treadmill and heated water pools reduces systolic and diastolic BP and improves physical performance [34, 101]. Another study demonstrated that a 12-week aerobic exercise training improved central BP, BP variability, and cardiovascular biomarkers such as interferon-gamma and angiotensin II in resistant hypertensive patients [102]. Despite the lack of evidence showing an association between exercise and BP in patients with RH, the current recommendations developed by experts are appropriate. In general, it is recommended that patients with RH engage in at least 150 min of moderate-intensity or 75 min of vigorous-intensity aerobic exercise per week [8, 22, 91]. A gradual increase in aerobic exercise to 300 min of moderate-intensity or 150 min of vigorous-intensity aerobic exercise is advised [103]. Muscle-strengthening exercises involving all major muscle groups for 2 or more days per week and balance training may also be advised [104].

Weight control

Body mass index is positively correlated with increased BP [26]. Patients with increased adiposity usually have insulin resistance, high sympathetic nervous system activity, an activated RAS, and increased salt sensitivity [28]. Elevated insulin levels stimulate sympathetic tone and the renin-angiotensin-aldosterone system. In obese individuals, sodium is reabsorbed by the kidneys via renovascular mechanisms [105]. Of these complicated pathophysiological mechanisms, obese patients are likely to develop RH [26, 28]. A long-term excessive energy intake of as little as 50-100 kcal per day, even in small increments of sugars, refined grains, processed foods, and alcoholic beverages when consumed for a long time, causes weight gain [106, 107]. These common risk factors for obesity and hypertension can also cause BP elevation and treatment-RH. Thus, weight reduction through a healthier lifestyle is helpful for improving BP control. Systematic reviews consistently demonstrated that 1 kg of weight reduction decreased systolic BP by approximately 1 mmHg during a follow-up of 2-3 years even although this inverse association was attenuated in longer-term follow-up [108,109,110]. Guidelines strongly recommend that overweight or obese adults with elevated BP or hypertension should lose body weight to the ideal body weight for BP reduction [8, 22, 91].

Alcohol consumption

Although modest alcohol intake increases high-density lipoprotein cholesterol levels and is inversely associated with a lower risk of coronary heart disease than abstinence [111, 112], excess alcohol intake has harmful effects on BP and cardiovascular events. Excessive alcohol consumption increases BP and accounts for approximately 10% of the population with hypertension. Patients with hypertension who consume heavy alcohol are more likely to be resistant to antihypertensive management, resulting in RH. According to a meta-analysis of randomized controlled trials, reduced alcohol consumption significantly decreased the mean systolic and diastolic BP in a dose-dependent manner [113, 114]. Adults with elevated BP or hypertension who currently consume alcohol should be advised to drink 2 and 1 or less standard drinks per day in men and women, respectively [8, 113,114,115]. One standard drink contained approximately 14 g of pure ethanol [116].

Tobacco smoking

Tobacco smoking and secondhand smoke are modifiable risk factors for hypertension and cardiovascular events, in addition to various malignant neoplasms and chronic obstructive pulmonary disease (COPD). Tobacco smoking causes an immediate but prolonged pressor effect, which may increase daytime ambulatory BP and lead to inaccurate results [117, 118]. However, several studies have reported that BP is lower in current smokers than in non-smokers or former smokers, and smoking cessation is significantly related to hypertension risk [119]. Transient elevation in BP after smoking cessation may have been caused by weight gain. The health benefits of smoking cessation outweigh the harmful effects, such as transient BP elevation through weight gain [120]. Tobacco smoking threatens public health, accounting for nearly 6.3 million deaths and 6.3% of global disability-adjusted life years (DALYs) [121]. To ameliorate the public health burden of tobacco smoking, all patients with hypertension who smoke tobacco should be advised to stop smoking.

Pharmacologic management of resistant hypertension

For pharmacological management of RH, a combination of optimal or maximally tolerated doses of long-acting RAS inhibitors, calcium channel blockers, and thiazide-type or thiazide-like diuretics should be prescribed [4, 22, 91]. If a patient has uncontrolled BP despite maximum tolerated triple combination consider switching RAS inhibitors and calcium channel blockers to the most potent, longest acting drugs in each class and to switch thiazide type diuretics to thiazide like diuretics such as chlorthalidone and/or indapamide [4]. Thiazide-like diuretics have more potent BP-lowering effects than hydrochlorothiazides [122]. Recommendations have suggested that in patients with CKD with an estimated GFR < 45 ml/min/m2 thiazide diuretics may not be effective and loop diuretics should be used instead. However, increasing evidence suggests that thiazide like diuretics can effectively reduce BP in patients with low eGFR [123]. The CLICK trial demonstrated that chlorthalidone is effective in lowering BP in patients with advanced CKD and uncontrolled hypertension [124]. This is important, considering the high prevalence of RH in patients with CKD [125]. Because it is unavoidable that patients with RH are prescribed a higher number of antihypertensive medications, RH may be associated with poor drug adherence [10]. Therefore, medications should be minimized by considering a fixed-dose combination at the beginning and, if not, by considering switching the medications to a fixed-dose combination when stabilized. In patients with CKD stage 4 or stage 5(eGFR < 30 ml/min/1.73 m2) and either resistant edema or uncontrolled BP, loop diuretics should be considered and can be used with existing thiazide diuretics [126]. Furosemide, which has a short half-life, is administered twice daily, whereas torsemide may be administered once daily.

When BP is not controlled by the maximally tolerated triple combination of RAS inhibitors, CCBs, and diuretics, spironolactone is considered the drug of choice. In the PATHWAY 2 trial, spironolactone demonstrated significantly better efficacy than doxazosin and bisoprolol in reducing the primary endpoint of the home systolic BP in patients with RH [127]. In cases where spironolactone is not tolerated, potassium-sparing diuretics, such as amiloride, may be considered [128]. However, no randomized clinical trials have compared the efficacy of spironolactone and amiloride in patients with RH. The major limitation of potassium-sparing diuretics is the relatively high incidence of hyperkalemia. This is even more problematic in patients with resistant hypertension owing to the high prevalence of CKD. In the AMBER trial, the use of spironolactone in patients with CKD and RH treatment, with or without the concomitant use of a potassium binder, was 64.2% and 35.4%, respectively [129, 130]. This study showed that using a potassium binder (patiromer) in advanced CKD patient with RH can allow for more patients to be maintained on spironolactone. Non-steroidal aldosterone antagonists, which are associated with a lower incidence of hyperkalemia than spironolactone, appear promising [130]. If BP is not controlled with spironolactone or if spironolactone cannot be used, the sequential addition of beta-blockers, alpha-blockers, and vasodilators(hydralazine, minoxidil) should be considered [4]. This treatment regimen should be sufficient to control BP in most patients with RH. However, less than 1% of patients with hypertension (10% of patients with RH) are refractory to medical treatment, including thiazide-like diuretics and spironolactone and are considered to have refractory hypertension.

Managing RH in older adults requires a comprehensive approach. Older adults are more likely to take medications that increase their BP. Therefore, efforts should be made to identify medications, such as NSAIDs, steroids, or herbal medications, that may contribute to uncontrolled hypertension. Other treatment strategies may involve the optimization of medication regimens by adjusting dosages, adding medications from different drug classes, or using fixed-dose combinations. Lifestyle modifications, including weight loss, regular exercise, and a low-sodium diet are also important in older patients. However, strict lifestyle modifications can lead to dehydration, sarcopenia, or fall-related injuries; thus, individualized recommendations should be provided to older patients with hypertension [131].

Non-pharmacologic (Device Based) management of resistant hypertension

The pivotal role of the sympathetic nervous system in the development and progression of hypertension is well-established [132]. Before the availability of antihypertensive medications, surgical sympathectomy was used [133]. It has since been largely phased out owing to undesirable side effects such as orthostatic hypotension and the growing availability of antihypertensive medications. Despite its discontinuation, significant strides made in BP control following surgical sympathectomy have served as a foundation for the development of device-based treatments for RH.

Renal denervation

Renal denervation (RDN) has emerged as a promising novel intervention for RH management [4]. The procedure targets the renal sympathetic nerves, which play a pivotal role in the regulation of BP, by delivering radiofrequency, ultrasound energy, or ethanol to interrupt the efferent and afferent nerves in the renal arteries.

The early promise shown in a proof-of-concept study [134] (SYMPLICITY HTN-1) and a subsequent randomized open-label study [135] (SYMPLICITY HTN-2) was tempered by the results of the SYMPLICITY HTN-3 trial [136], which failed to meet its primary efficacy endpoint in the RH population. However, the methodological concerns arising from SYMPLICITY HTN-3, including alterations in patient behavior in adherence to their multidrug pharmacological regimens and inadequate denervation resulting from technical failure of the catheter and/or the interventional proceduralist have led to several new catheters and new trial designs to evaluate the efficacy of more extensive renal denervation, and subsequent studies have presented more favorable results.

Several recent sham-controlled clinical trials [137,138,139] (SPYRAL HTN-OFF MED, RADIANCE-HTN SOLO, RADIANCE II trial) in patients with mild to moderate HTN, although not studies in patients with RH, have shown significant reductions in BP in RDN group. Based on these results, RDN has emerged as an evidence-based option for treating hypertension by complementing lifestyle modifications and antihypertensive medications [140].

More recent randomized controlled trials [141,142,143] (DENERHTN, SPYRAL HTN-ON MED, and RADIANCE-HTN TRIO trials) have demonstrated significant BP-lowering effects of RDN in uncontrolled hypertension, renewing optimism about the therapeutic potential of this approach. In the SPYRAL HTN-ON MED trial, 80 hypertensive patients uncontrolled with one to three antihypertensive medications were randomized to the Symplicity Spyral multielectrode catheter ablation or sham control group. The mean adjusted difference for the primary endpoint of 24 h systolic BP at 6 month was -7.0 mmHg(95% CI: -12.0 to -2.1, P=0.0059) [142]. In the long term follow up study of the SPYRAL HTN-ON MED trial, the adjusted difference for the 24 hour systolic BP at 36 months was -10.0 mmHg(95% CI: -16.6 to -3.3, P= 0.0039) [144]. The results demonstrate that the efficacy of renal denervation may be maintained in the long-term. In the RADIANCE-HTN TRIO trial, 136 patients with RH were randomized to receive renal denervation with an endovascular ultrasound device or a sham procedure, with the primary endpoint being the change in daytime ambulatory systolic BP at 2 months. The results showed a significant difference in the daytime systolic BP [median difference 4.5 mmHg(95% CI, -8.5 to -0.3, P=0.022)] [143], demonstrating the efficacy of renal denervation in treatment RH. However, despite these promising results, further long-term studies are needed to fully elucidate the role of RDN in the management of RH and identify the patient subgroups most likely to benefit. Considering the modest reduction in BP, more data are needed regarding the cost-benefit of renal denervation in patients with RH. A comparative study of aldosterone antagonists, their efficacy in refractory hypertension, and their efficacy in patients with CKD should be conducted. In the 2023 European Society of Hypertension guidelines for the management of arterial hypertension, renal denervation was given a class II recommendation for the treatment of RH with an eGFR > 40 ml/min/1.73 m2 [145].

Baroreflex activation therapy

The carotid baroreceptor reflex regulates the BP. Carotid baroreceptor activation therapy (BAT) involves the implantation of a device near the carotid sinus that modulates autonomic outflow. This modulation leads to a reduction in sympathetic activity and an increase in parasympathetic activity, resulting in decreased BP [146]. Endovascular baroreflex amplification is a novel technique used to lower BP in RH. The BAT is a crucial aspect of baroreflex amplification that specifically targets the carotid sinuses. BAT can be categorized into electrical and mechanical types.

Currently, two generations of electrical BAT have been developed and are supported by evidence from clinical trials. The first-generation Rheos System, which was evaluated in the Rheos Pivotal trial [147], did not receive FDA approval because of safety concerns. In contrast, the second-generation Barostim Neo System, assessed in the Barostim Neo Trial [148] received FDA approval for the treatment of heart failure. A recent cohort study with the Barostim Neo device demonstrated its efficacy over a period of 2 years, with 25 of 50 patients with RH achieving controlled office systolic BP levels below 140 mmHg [149]. Further randomized controlled trials are required to validate the effects of BAT on RH. However, this novel device has some unmet needs, including invasiveness during the procedure, concerns about battery replacement, and potential side effects such as syncope, arrhythmias, and unadjusted BP levels.

The less invasive Endovascular Baroreflex Amplification (EVBA) Mobius HD device implanted within the carotid sinus mechanically modulates the baroreceptors by enhancing wall stretch. This modulation activates the carotid baroreceptor and reduces BP levels [150]. The first clinical study was conducted in 2013 and showed a notable reduction in office BP at six months, with systolic and diastolic BP decreasing by 24 mmHg and 12 mmHg, respectively [151]. However, some patients experience hypotension, worsening hypertension, and infections. After a 3-year follow-up period, a significant reduction of 30 mmHg in SBP was observed, demonstrating the efficacy of EVBA in reducing BP with an allowable safety profile [152]. The CALM-2 trial is an ongoing, randomized, double-blind, sham-controlled trial evaluating EVBA using a MobiusHD device. This study enrolled 300 participants and assessed the change in mean 24-hour ambulatory systolic BP at the six-month as the primary endpoint, providing valuable insights into the effectiveness of the therapy [153]. Additional randomized, sham-controlled trials are required to confirm the long-term efficacy and safety of EVBA devices. Close collaboration between interventionists and clinicians plays a pivotal role in determining the success of device therapy in patients with RH.

Continuous Positive Airway Pressure (CPAP)

OSA is associated with an increase in nocturnal BP, a non-dipping pattern in diurnal BP variability, and an increase in morning surge, which may contribute to the difficulty of treating hypertension and increase the risk of cardiovascular events [154, 155]. OSA is very common in patients with RH, with the reported prevalence of up to 70 to 90% [4]. This may be due to an increase in fluid retention and upper airway edema in RH, which increases upper airway resistance and predisposes these patients to severe OSA [4, 54, 156]. Overall, the reported effect of continuous positive airway pressure (CPAP) on BP reduction in patients with RH is modest. In a study of 194 patients with RH who were randomized to CPAP or no therapy, CPAP was associated with a significantly greater decrease in the 24-hour mean BP [3.1 mmHg (95% CI, 0.6 to 5.6), P = 0.02] and 24-hour DBP [3.2 mmHg (95% CI, 1.0 to 5.4 mmHg), P = 0.005] at 12th week of treatment. However, this and other recent clinical trials using CPAP enrolled asymptomatic patients with OSA for obvious ethical reasons, while excluding subjects with symptomatic OSA [157]. This may underestimate the efficacy of CPAP as symptomatic OSA patients with RH may have a more robust response to effective CPAP therapy. In a randomized study of 60 symptomatic patients with moderate to severe OSA who were randomized to effective CPAP versus subtherapeutic CPAP for 9 weeks, the mean arterial BP decreased by 9.9±11.4 mmHg in patients undergoing effective CPAP therapy [158]. Therefore, a careful patient history that assesses the possible symptoms of OSA, such as daytime somnolence and morning headache, should be ascertained in patients with RH. In patients with typical symptoms, testing for OSA is highly recommended, because CPAP may be effective in improving BP control. Moreover, a prospective observational study on treatment-RH with OSA revealed that BP reduction by CPAP was stronger in patients with nocturnal hypertension evaluated by ABPM compared to that in patients with nocturnal normotension [159]. Therefore, the assessment of the presence of nocturnal hypertension using ABPM is also recommended, especially in patients with treatment-RH complicated by OSA.

Refractory hypertension

Refractory hypertension is defined as hypertension that cannot be controlled below the target BP with the use of 5 or more antihypertensive agents. A stricter definition is the failure to maintain BP below the target BP with the use of five or more antihypertensive agents, including thiazide-like diuretics(chlorthalidone or indapamide) and spironolactone [160]. In an analysis of 14,809 participants receiving antihypertensive agents from the Reasons for Geographic and Racial Differences in Stroke(REGARDS) Study, 2144(14.5%) had RH and 78(0.5%) had refractory hypertension [6]. Black race, male sex, higher body mass index(BMI), living in the stroke area of the United States(Southern States), lower heart rate, reduced estimated GFR, albuminuria, diabetes mellitus, history of stroke, and history of coronary heart disease were significantly associated with refractory hypertension [6]. In a prospective cohort study of 1,726 patients with RH, the reported prevalence of refractory hypertension among subjects with RH was 8.6% [161]. In the same cohort, patients with refractory hypertension were younger, more obese, and had a higher prevalence of smoking and CKD. Surprisingly, the prevalence of DM was also lower. Patients with refractory hypertension had a significantly higher heart rate than those without refractory hypertension, despite the higher use of beta-blockers [161]. Previous studies have also demonstrated that patients with refractory hypertension are younger, with evidence of higher sympathetic nervous system activation, and no significant difference from non-refractory hypertension in terms of volume excess parameters [160, 162]. This may explain the lack of response to diuretics and aldosterone antagonists.

Refractory hypertension is associated with increased risk of adverse cardiovascular events overall and have increased risk compared to non-refractory RH. In an analysis of a prospective cohort of 1576 patients with RH, refractory hypertension was associated with significant increase in the risk of total cardiovascular events(HR:1.44[1.01-2.07]), major adverse cardiovascular events(HR:1.68[1.16-2.45]), cardiovascular mortality(HR:1.85[1.18-2.90]) and stroke(HR:2.14[1.17-3.93]) [161] compared with subjects with non-refractory RH [161]. In a recent Korean study, 150(0.9%) of 16,284 patients who underwent ABPM had refractory hypertension. Similar to previous reports, patients with refractory hypertension had a higher prevalence of obesity, DM, CKD, heart failure, myocardial infarction, and stroke. For cardiovascular mortality, the adjusted HR of refractory hypertension was 5.22 compared to non-RH [16]. As the treatment options for these subsets of patients are currently limited, the development of treatment options for refractory hypertension is an unmet need in the field of hypertension that warrants further research [163].

Conclusion

The prevalence of the aTRH is approximately 10-15%. Among these patients, specific aspects, such as pseudo-resistance, WUCH, secondary hypertension, non-adherence, and inappropriate treatment regimens, must be ruled out to properly diagnose true RH. Comprehensive treatment approaches, including lifestyle interventions, pharmacological approaches, and systematic approaches to improve adherence, are needed to control RH. Device-based therapies aimed at sympathetic nervous system activity are currently used in patients with RH. Among device-based therapies, RDNs have been shown to be effective in some patients with RH; however, more research is needed to apply them to a larger number of patients with RH. Further studies are required to determine the long-term efficacy and safety of these devices. Refractory hypertension, defined as the failure to control BP below the target BP with the use of five or more antihypertensive agents, including thiazide-like diuretics (chlorthalidone or indapamide) and spironolactone, is associated with a poor prognosis. Further research is warranted to develop better treatment options for refractory hypertension.

Availability of data and materials

“Not applicable”

Abbreviations

BP:

Blood pressure

RH:

Resistant hypertension

RAS:

Renin-angiotensin system

ABPM:

Ambulatory blood pressure monitoring

HBPM:

Home BP monitoring

CKD:

Chronic kidney disease

DM:

Diabetes mellitus,

ALLHAT:

The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial

aTRH:

Apparent treatment resistant hypertension

DASH:

Dietary Approaches to Stop Hypertension

NSAIDs:

Nonsteroidal anti-inflammatory drugs

OSA:

Obstructive sleep apnea

RVH:

Renovascular hypertension

WUCH:

White-coat uncontrolled hypertension

COPD:

Chronic obstructive pulmonary disease

DALYs:

Disability-adjusted life years,

EVABA:

Endovascular Baroreflex Amplification

CPAP:

Continuous positive airway pressure

REGARDS:

Reasons for Geographic and Racial Differences in Stroke

ACE:

Angiotensin converting enzyme

ARB:

Angiotensin II receptor blocker

TSH:

Thyroid stimulating hormone

CCBs:

Calcium channel blockers

References

  1. Blood Pressure Lowering Treatment Trialists C. Pharmacological blood pressure lowering for primary and secondary prevention of cardiovascular disease across different levels of blood pressure: an individual participant-level data meta-analysis. Lancet. 2021;397(10285):1625-36.

  2. Kim HC, Ihm SH, Kim GH, Kim JH, Kim KI, Lee HY, et al. 2018 Korean Society of Hypertension guidelines for the management of hypertension: part I-epidemiology of hypertension. Clin Hypertens. 2019;25:16.

    PubMed  PubMed Central  Google Scholar 

  3. Korean Society H, Hypertension Epidemiology Research Working G, Kim HC, Cho MC. Korea hypertension fact sheet 2018. Clin Hypertens. 2018;24:13.

  4. Carey RM, Calhoun DA, Bakris GL, Brook RD, Daugherty SL, Dennison-Himmelfarb CR, et al. Resistant hypertension: detection, evaluation, and management: a scientific statement from the american heart association. Hypertension. 2018;72(5):e53–90.

    CAS  PubMed  Google Scholar 

  5. Lee HY, Shin J, Kim GH, Park S, Ihm SH, Kim HC, et al. 2018 Korean Society of Hypertension Guidelines for the management of hypertension: part II-diagnosis and treatment of hypertension. Clin Hypertens. 2019;25:20.

    PubMed  PubMed Central  Google Scholar 

  6. Calhoun DA, Booth JN 3rd, Oparil S, Irvin MR, Shimbo D, Lackland DT, et al. Refractory hypertension: determination of prevalence, risk factors, and comorbidities in a large, population-based cohort. Hypertension. 2014;63(3):451–8.

    CAS  PubMed  Google Scholar 

  7. Calhoun DA. Refractory and resistant hypertension: antihypertensive treatment failure versus treatment resistance. Korean Circ J. 2016;46(5):593–600.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Kim HL, Lee EM, Ahn SY, Kim KI, Kim HC, Kim JH, et al. The 2022 focused update of the 2018 Korean Hypertension Society Guidelines for the management of hypertension. Clin Hypertens. 2023;29(1):11.

    PubMed  PubMed Central  Google Scholar 

  9. de la Sierra A, Segura J, Banegas JR, Gorostidi M, de la Cruz JJ, Armario P, et al. Clinical features of 8295 patients with resistant hypertension classified on the basis of ambulatory blood pressure monitoring. Hypertension. 2011;57(5):898–902.

    PubMed  Google Scholar 

  10. Gupta P, Patel P, Strauch B, Lai FY, Akbarov A, Gulsin GS, et al. Biochemical screening for nonadherence is associated with blood pressure reduction and improvement in adherence. Hypertension. 2017;70(5):1042–8.

    CAS  PubMed  Google Scholar 

  11. Noubiap JJ, Nansseu JR, Nyaga UF, Sime PS, Francis I, Bigna JJ. Global prevalence of resistant hypertension: a meta-analysis of data from 3.2 million patients. Heart. 2019;105(2):98–105.

    PubMed  Google Scholar 

  12. Calhoun DA, Jones D, Textor S, Goff DC, Murphy TP, Toto RD, et al. Resistant hypertension: diagnosis, evaluation, and treatment. A scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research. Hypertension. 2008;51(6):1403–19.

    CAS  PubMed  Google Scholar 

  13. Weitzman D, Chodick G, Shalev V, Grossman C, Grossman E. Prevalence and factors associated with resistant hypertension in a large health maintenance organization in Israel. Hypertension. 2014;64(3):501–7.

    CAS  PubMed  Google Scholar 

  14. Choi SW, Kim MK, Han SW, Han SH, Lee BK, Lee SU, et al. Apparent treatment-resistant hypertension among elderly Korean hypertensives: an insight from the HIT registry. J Hum Hypertens. 2014;28(3):201–5.

    CAS  PubMed  Google Scholar 

  15. Lee KN, Na JO, Choi CU, Lim HE, Kim JW, Kim EJ, et al. Prevalence and characteristics of resistant hypertension at primary clinics in Korea: a nationwide cross-sectional study. Clin Hypertens. 2015;22:4.

    PubMed  Google Scholar 

  16. Yoon M, You SC, Oh J, Lee CJ, Lee SH, Kang SM, et al. Prevalence and prognosis of refractory hypertension diagnosed using ambulatory blood pressure measurements. Hypertens Res. 2022;45(8):1353–62.

    CAS  PubMed  Google Scholar 

  17. Narita K, Hoshide S, Kario K. Association of treatment-resistant hypertension defined by home blood pressure monitoring with cardiovascular outcome. Hypertens Res. 2022;45(1):75–86.

    PubMed  Google Scholar 

  18. Daugherty SL, Powers JD, Magid DJ, Tavel HM, Masoudi FA, Margolis KL, et al. Incidence and prognosis of resistant hypertension in hypertensive patients. Circulation. 2012;125(13):1635–42.

    PubMed  PubMed Central  Google Scholar 

  19. Muntner P, Davis BR, Cushman WC, Bangalore S, Calhoun DA, Pressel SL, et al. Treatment-resistant hypertension and the incidence of cardiovascular disease and end-stage renal disease: results from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Hypertension. 2014;64(5):1012–21.

    CAS  PubMed  Google Scholar 

  20. Cardoso CRL, Salles GC, Salles GF. Prognostic importance of on-treatment clinic and ambulatory blood pressures in resistant hypertension: A cohort study. Hypertension. 2020;75(5):1184–94.

    CAS  PubMed  Google Scholar 

  21. Pickering TG, Hall JE, Appel LJ, Falkner BE, Graves J, Hill MN, et al. Recommendations for blood pressure measurement in humans and experimental animals: part 1: blood pressure measurement in humans: a statement for professionals from the Subcommittee of Professional and Public Education of the American Heart Association Council on High Blood Pressure Research. Circulation. 2005;111(5):697–716.

    PubMed  Google Scholar 

  22. Whelton PK, Carey RM, Aronow WS, Casey DE Jr, Collins KJ, Dennison Himmelfarb C, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension. 2018;71(6):e13–115.

    CAS  PubMed  Google Scholar 

  23. Egan BM, Zhao Y, Li J, Brzezinski WA, Todoran TM, Brook RD, et al. Prevalence of optimal treatment regimens in patients with apparent treatment-resistant hypertension based on office blood pressure in a community-based practice network. Hypertension. 2013;62(4):691–7.

    CAS  PubMed  Google Scholar 

  24. Durand H, Hayes P, Morrissey EC, Newell J, Casey M, Murphy AW, et al. Medication adherence among patients with apparent treatment-resistant hypertension: systematic review and meta-analysis. J Hypertens. 2017;35(12):2346–57.

    CAS  PubMed  Google Scholar 

  25. Franklin SS, Thijs L, Asayama K, Li Y, Hansen TW, Boggia J, et al. The cardiovascular risk of white-coat hypertension. J Am Coll Cardiol. 2016;68(19):2033–43.

    PubMed  Google Scholar 

  26. Landsberg L, Aronne LJ, Beilin LJ, Burke V, Igel LI, Lloyd-Jones D, et al. Obesity-related hypertension: pathogenesis, cardiovascular risk, and treatment: a position paper of The Obesity Society and the American Society of Hypertension. J Clin Hypertens (Greenwich). 2013;15(1):14–33.

    CAS  PubMed  Google Scholar 

  27. Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity-induced hypertension: interaction of neurohumoral and renal mechanisms. Circ Res. 2015;116(6):991-1006.

  28. Egan BM, Zhao Y, Axon RN, Brzezinski WA, Ferdinand KC. Uncontrolled and apparent treatment resistant hypertension in the United States, 1988 to 2008. Circulation. 2011;124(9):1046–58.

    PubMed  PubMed Central  Google Scholar 

  29. Sacks FM, Svetkey LP, Vollmer WM, Appel LJ, Bray GA, Harsha D, et al. Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. DASH-Sodium Collaborative Research Group. N Engl J Med. 2001;344(1):3-10.

  30. Oh YS, Appel LJ, Galis ZS, Hafler DA, He J, Hernandez AL, et al. National heart, lung, and blood institute working group report on salt in human health and sickness: building on the current scientific evidence. Hypertension. 2016;68(2):281–8.

    PubMed  Google Scholar 

  31. Blaustein MP, Leenen FH, Chen L, Golovina VA, Hamlyn JM, Pallone TL, et al. How NaCl raises blood pressure: a new paradigm for the pathogenesis of salt-dependent hypertension. Am J Physiol Heart Circ Physiol. 2012;302(5):H1031–49.

    CAS  PubMed  Google Scholar 

  32. Pimenta E, Gaddam KK, Oparil S, Aban I, Husain S, Dell’Italia LJ, et al. Effects of dietary sodium reduction on blood pressure in subjects with resistant hypertension: results from a randomized trial. Hypertension. 2009;54(3):475–81.

    CAS  PubMed  Google Scholar 

  33. Diaz KM, Shimbo D. Physical activity and the prevention of hypertension. Curr Hypertens Rep. 2013;15(6):659–68.

    PubMed  PubMed Central  Google Scholar 

  34. Dimeo F, Pagonas N, Seibert F, Arndt R, Zidek W, Westhoff TH. Aerobic exercise reduces blood pressure in resistant hypertension. Hypertension. 2012;60(3):653–8.

    CAS  PubMed  Google Scholar 

  35. Puddey IB, Beilin LJ. Alcohol is bad for blood pressure. Clin Exp Pharmacol Physiol. 2006;33(9):847–52.

    CAS  PubMed  Google Scholar 

  36. Miller PM, Anton RF, Egan BM, Basile J, Nguyen SA. Excessive alcohol consumption and hypertension: clinical implications of current research. J Clin Hypertens (Greenwich). 2005;7(6):346–51.

    PubMed  Google Scholar 

  37. Piano MR, Benowitz NL, Fitzgerald GA, Corbridge S, Heath J, Hahn E, et al. Impact of smokeless tobacco products on cardiovascular disease: implications for policy, prevention, and treatment: a policy statement from the American Heart Association. Circulation. 2010;122(15):1520–44.

    PubMed  Google Scholar 

  38. Grossman A, Messerli FH, Grossman E. Drug induced hypertension–an unappreciated cause of secondary hypertension. Eur J Pharmacol. 2015;763(Pt A):15–22.

    CAS  PubMed  Google Scholar 

  39. Cohen JB, Brown NJ, Brown SA, Dent S, van Dorst DCH, Herrmann SM, et al. Cancer therapy-related hypertension: A scientific statement From the American heart association. Hypertension. 2023;80(3):e46–57.

    CAS  PubMed  Google Scholar 

  40. Fay KS, Cohen DL. Resistant hypertension in people with CKD: a review. Am J Kidney Dis. 2021;77(1):110–21.

    CAS  PubMed  Google Scholar 

  41. Mann SJ. Severe paroxysmal hypertension (pseudopheochromocytoma): understanding the cause and treatment. Arch Intern Med. 1999;159(7):670–4.

    CAS  PubMed  Google Scholar 

  42. Jin P, Hulshof MC, de Jong R, van Hooft JE, Bel A, Alderliesten T. Quantification of respiration-induced esophageal tumor motion using fiducial markers and four-dimensional computed tomography. Radiother Oncol. 2016;118(3):492–7.

    PubMed  Google Scholar 

  43. Zhong X, Hilton HJ, Gates GJ, Jelic S, Stern Y, Bartels MN, et al. Increased sympathetic and decreased parasympathetic cardiovascular modulation in normal humans with acute sleep deprivation. J Appl Physiol (1985). 2005;98(6):2024–32.

    PubMed  Google Scholar 

  44. Huan Y, Cohen DL, Townsend RR. Pathophysiology of hypertension in chronic kidney disease. In: Kimmel PL, Rosenberg ME, editors. Chronic renal disease. Cambredge: Massachusetts. Academic Press; 2014. p. 163–9.

    Google Scholar 

  45. Muntner P, Anderson A, Charleston J, Chen Z, Ford V, Makos G, et al. Hypertension awareness, treatment, and control in adults with CKD: results from the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis. 2010;55(3):441–51.

    CAS  PubMed  Google Scholar 

  46. Ihm CG. Hypertension in chronic glomerulonephritis. Electrolyte Blood Press. 2015;13(2):41–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Orofino L, Quereda C, Lamas S, Orte L, Gonzalo A, Mampaso F, et al. Hypertension in primary chronic glomerulonephritis: analysis of 288 biopsied patients. Nephron. 1987;45(1):22–6.

    CAS  PubMed  Google Scholar 

  48. Vaidya A, Mulatero P, Baudrand R, Adler GK. The expanding spectrum of primary aldosteronism: implications for diagnosis, pathogenesis, and treatment. Endocr Rev. 2018;39(6):1057–88.

    PubMed  PubMed Central  Google Scholar 

  49. Calhoun DA, Nishizaka MK, Zaman MA, Thakkar RB, Weissmann P. Hyperaldosteronism among black and white subjects with resistant hypertension. Hypertension. 2002;40(6):892–6.

    CAS  PubMed  Google Scholar 

  50. Funder JW, Carey RM, Fardella C, Gomez-Sanchez CE, Mantero F, Stowasser M, et al. Case detection, diagnosis, and treatment of patients with primary aldosteronism: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2008;93(9):3266–81.

    PubMed  Google Scholar 

  51. Logan AG, Perlikowski SM, Mente A, Tisler A, Tkacova R, Niroumand M, et al. High prevalence of unrecognized sleep apnoea in drug-resistant hypertension. J Hypertens. 2001;19(12):2271–7.

    CAS  PubMed  Google Scholar 

  52. Pimenta E, Stowasser M, Gordon RD, Harding SM, Batlouni M, Zhang B, et al. Increased dietary sodium is related to severity of obstructive sleep apnea in patients with resistant hypertension and hyperaldosteronism. Chest. 2013;143(4):978–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Gonzaga CC, Gaddam KK, Ahmed MI, Pimenta E, Thomas SJ, Harding SM, et al. Severity of obstructive sleep apnea is related to aldosterone status in subjects with resistant hypertension. J Clin Sleep Med. 2010;6(4):363–8.

    PubMed  PubMed Central  Google Scholar 

  54. Friedman O, Bradley TD, Chan CT, Parkes R, Logan AG. Relationship between overnight rostral fluid shift and obstructive sleep apnea in drug-resistant hypertension. Hypertension. 2010;56(6):1077–82.

    CAS  PubMed  Google Scholar 

  55. Kasai T, Bradley TD, Friedman O, Logan AG. Effect of intensified diuretic therapy on overnight rostral fluid shift and obstructive sleep apnoea in patients with uncontrolled hypertension. J Hypertens. 2014;32(3):673–80.

    CAS  PubMed  Google Scholar 

  56. Chung F, Yegneswaran B, Liao P, Chung SA, Vairavanathan S, Islam S, et al. STOP questionnaire: a tool to screen patients for obstructive sleep apnea. Anesthesiology. 2008;108(5):812–21.

    PubMed  Google Scholar 

  57. Herrmann SM, Textor SC. Current concepts in the treatment of renovascular hypertension. Am J Hypertens. 2018;31(2):139–49.

    CAS  PubMed  Google Scholar 

  58. Anderson GH Jr, Blakeman N, Streeten DH. The effect of age on prevalence of secondary forms of hypertension in 4429 consecutively referred patients. J Hypertens. 1994;12(5):609–15.

    PubMed  Google Scholar 

  59. Martell N, Rodriguez-Cerrillo M, Grobbee DE, Lopez-Eady MD, Fernandez-Pinilla C, Avila M, et al. High prevalence of secondary hypertension and insulin resistance in patients with refractory hypertension. Blood Press. 2003;12(3):149–54.

    CAS  PubMed  Google Scholar 

  60. Lenders JW, Duh QY, Eisenhofer G, Gimenez-Roqueplo AP, Grebe SK, Murad MH, et al. Pheochromocytoma and paraganglioma: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014;99(6):1915–42.

    CAS  PubMed  Google Scholar 

  61. Lenders JW, Eisenhofer G, Mannelli M, Pacak K. Phaeochromocytoma. Lancet. 2005;366(9486):665–75.

    PubMed  Google Scholar 

  62. Schwartz GL. Screening for adrenal-endocrine hypertension: overview of accuracy and cost-effectiveness. Endocrinol Metab Clin North Am. 2011;40(2):279–94 vii.

    PubMed  PubMed Central  Google Scholar 

  63. Martins LC, Conceicao FL, Muxfeldt ES, Salles GF. Prevalence and associated factors of subclinical hypercortisolism in patients with resistant hypertension. J Hypertens. 2012;30(5):967–73.

    CAS  PubMed  Google Scholar 

  64. Bhatt H, Siddiqui M, Judd E, Oparil S, Calhoun D. Prevalence of pseudoresistant hypertension due to inaccurate blood pressure measurement. J Am Soc Hypertens. 2016;10(6):493–9.

    PubMed  PubMed Central  Google Scholar 

  65. de la Sierra A, Segura J, Banegas JR, Gorostidi M. Clinical features of 8295 patients with resistant hypertension classified on the basis of ambulatory blood pressure monitoring. Hypertension. 2011;57:898–902.

    PubMed  Google Scholar 

  66. Shin J, Park SH, Kim JH, Ihm SH, Kim K-I, Kim WS, et al. Discordance between ambulatory versus clinic blood pressure according to global cardiovascular risk group. Korean J Intern Med. 2015;30(5):610.

    PubMed  PubMed Central  Google Scholar 

  67. Brown MA, Buddle ML, Martin A. Is resistant hypertension really resistant? Am J Hypertens. 2001;14(12):1263–9.

    CAS  PubMed  Google Scholar 

  68. Muxfeldt ES, Bloch KV, da Rocha Nogueira A, Salles GF. True resistant hypertension: is it possible to be recognized in the office? Am J Hypertens. 2005;18(12):1534–40.

    PubMed  Google Scholar 

  69. Verdecchia P, Schillaci G, Borgioni C, Ciucci A, Porcellati C. Prognostic significance of the white coat effect. Hypertension. 1997;29(6):1218–24.

    CAS  PubMed  Google Scholar 

  70. Eskås PA, Heimark S, Eek Mariampillai J, Larstorp AC, Fadl Elmula FE, Høieggen A. Adherence to medication and drug monitoring in apparent treatment-resistant hypertension. Blood Press. 2016;25(4):199–205.

    PubMed  Google Scholar 

  71. Min HJ, Cho YJ, Kim CH, Kim DH, Kim HY, Choi JI, et al. Clinical features of obstructive sleep apnea that determine its high prevalence in resistant hypertension. Yonsei Med J. 2015;56(5):1258–65.

    PubMed  PubMed Central  Google Scholar 

  72. Yeghiazarians Y, Jneid H, Tietjens JR, Redline S, Brown DL, El-Sherif N, et al. Obstructive sleep apnea and cardiovascular disease: A scientific statement from the american heart association. Circulation. 2021;144(3):e56–67.

    CAS  PubMed  Google Scholar 

  73. Pickering TG, White WB. When and how to use self (home) and ambulatory blood pressure monitoring. J Am Soc Hypertens. 2008;2(3):119–24.

    PubMed  Google Scholar 

  74. Kim HM, Shin J. Role of home blood pressure monitoring in resistant hypertension. Clin Hypertens. 2023;29(1):2.

    PubMed  PubMed Central  Google Scholar 

  75. Schwartz GL, Turner ST. Screening for primary aldosteronism in essential hypertension: diagnostic accuracy of the ratio of plasma aldosterone concentration to plasma renin activity. Clin Chem. 2005;51(2):386–94.

    CAS  PubMed  Google Scholar 

  76. Nishizaka MK, Pratt-Ubunama M, Zaman MA, Cofield S, Calhoun DA. Validity of plasma aldosterone-to-renin activity ratio in African American and white subjects with resistant hypertension. Am J Hypertens. 2005;18(6):805–12.

    CAS  PubMed  Google Scholar 

  77. Sawka AM, Jaeschke R, Singh RJ, Young WF Jr. A comparison of biochemical tests for pheochromocytoma: measurement of fractionated plasma metanephrines compared with the combination of 24-hour urinary metanephrines and catecholamines. J Clin Endocrinol Metab. 2003;88(2):553–8.

    CAS  PubMed  Google Scholar 

  78. Choi KH, Yu YM, Ah YM, Chang MJ, Lee JY. Persistence with antihypertensives in uncomplicated treatment-naive very elderly patients: a nationwide population-based study. BMC Cardiovasc Disord. 2017;17(1):232.

    PubMed  PubMed Central  Google Scholar 

  79. Bourque G, Ilin JV, Ruzicka M, Hundemer GL, Shorr R, Hiremath S. Nonadherence is common in patients with apparent resistant hypertension: a systematic review and meta-analysis. Am J Hypertens. 2023;36(7):394–403.

    PubMed  Google Scholar 

  80. Choudhry NK, Kronish IM, Vongpatanasin W, Ferdinand KC, Pavlik VN, Egan BM, et al. Medication adherence and blood pressure control: a scientific statement from the american heart association. Hypertension. 2022;79(1):e1–14.

    CAS  PubMed  Google Scholar 

  81. Osterberg L, Blaschke T. Adherence to medication. N Engl J Med. 2005;353(5):487–97.

    CAS  PubMed  Google Scholar 

  82. Lawson AJ, Hameed MA, Brown R, Cappuccio FP, George S, Hinton T, et al. Nonadherence to antihypertensive medications is related to pill burden in apparent treatment-resistant hypertensive individuals. J Hypertens. 2020;38(6):1165–73.

    CAS  PubMed  Google Scholar 

  83. Jung O, Gechter JL, Wunder C, Paulke A, Bartel C, Geiger H, et al. Resistant hypertension? Assessment of adherence by toxicological urine analysis. J Hypertens. 2013;31(4):766–74.

    CAS  PubMed  Google Scholar 

  84. Siddiqui M, Judd EK, Dudenbostel T, Gupta P, Tomaszewski M, Patel P, et al. Antihypertensive medication adherence and confirmation of true refractory hypertension. Hypertension. 2020;75(2):510–5.

    CAS  PubMed  Google Scholar 

  85. Burnier M, Wuerzner G, Struijker-Boudier H, Urquhart J. Measuring, analyzing, and managing drug adherence in resistant hypertension. Hypertension. 2013;62(2):218–25.

    CAS  PubMed  Google Scholar 

  86. Lane D, Lawson A, Burns A, Azizi M, Burnier M, Jones DJL, et al. Nonadherence in hypertension: How to develop and implement chemical adherence testing. Hypertension. 2022;79(1):12–23.

    CAS  PubMed  Google Scholar 

  87. Gupta P, Patel P, Štrauch B, Lai FY, Akbarov A, Marešová V, et al. Risk factors for nonadherence to antihypertensive treatment. Hypertension. 2017;69(6):1113–20.

    CAS  PubMed  Google Scholar 

  88. Reese PP, Bloom RD, Trofe-Clark J, Mussell A, Leidy D, Levsky S, et al. Automated reminders and physician notification to promote immunosuppression adherence among kidney transplant recipients: a randomized trial. Am J Kidney Dis. 2017;69(3):400–9.

    PubMed  Google Scholar 

  89. Morawski K, Ghazinouri R, Krumme A, Lauffenburger JC, Lu Z, Durfee E, et al. Association of a smartphone application with medication adherence and blood pressure control: the MedISAFE-BP randomized clinical trial. JAMA Intern Med. 2018;178(6):802–9.

    PubMed  PubMed Central  Google Scholar 

  90. Jung SH, Lee OS, Kim HS, Park CS, Lee HJ, Kwon KH, et al. Medication adherence improvement by using administration timing simplification protocol (ATSP) in cardiovascular disease patients. J Atheroscler Thromb. 2017;24(8):841–52.

    PubMed  PubMed Central  Google Scholar 

  91. Williams B, Mancia G, Spiering W, Agabiti Rosei E, Azizi M, Burnier M, et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur Heart J. 2018;39(33):3021–104.

    PubMed  Google Scholar 

  92. Weinberger MH. Salt sensitivity of blood pressure in humans. Hypertension. 1996;27(3 Pt 2):481–90.

    CAS  PubMed  Google Scholar 

  93. Strazzullo P, D’Elia L, Kandala NB, Cappuccio FP. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. Bmj. 2009;339: b4567.

    PubMed  PubMed Central  Google Scholar 

  94. Elijovich F, Weinberger MH, Anderson CA, Appel LJ, Bursztyn M, Cook NR, et al. Salt sensitivity of blood pressure: a scientific statement from the american heart association. Hypertension. 2016;68(3):e7–46.

    CAS  PubMed  Google Scholar 

  95. Balafa O, Kalaitzidis RG. Salt sensitivity and hypertension. J Hum Hypertens. 2021;35(3):184–92.

    PubMed  Google Scholar 

  96. Appel LJ, Moore TJ, Obarzanek E, Vollmer WM, Svetkey LP, Sacks FM, et al. A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group. N Engl J Med. 1997;336(16):1117–24.

    CAS  PubMed  Google Scholar 

  97. Carnethon MR, Evans NS, Church TS, Lewis CE, Schreiner PJ, Jacobs DR Jr, et al. Joint associations of physical activity and aerobic fitness on the development of incident hypertension: coronary artery risk development in young adults. Hypertension. 2010;56(1):49–55.

    CAS  PubMed  Google Scholar 

  98. Arroll B, Beaglehole R. Does physical activity lower blood pressure: a critical review of the clinical trials. J Clin Epidemiol. 1992;45(5):439–47.

    CAS  PubMed  Google Scholar 

  99. Cornelissen VA, Smart NA. Exercise training for blood pressure: a systematic review and meta-analysis. J Am Heart Assoc. 2013;2(1): e004473.

    PubMed  PubMed Central  Google Scholar 

  100. Rossi A, Dikareva A, Bacon SL, Daskalopoulou SS. The impact of physical activity on mortality in patients with high blood pressure: a systematic review. J Hypertens. 2012;30(7):1277–88.

    CAS  PubMed  Google Scholar 

  101. Guimarães GV, Cruz LG, Tavares AC, Dorea EL, Fernandes-Silva MM, Bocchi EA. Effects of short-term heated water-based exercise training on systemic blood pressure in patients with resistant hypertension: a pilot study. Blood Press Monit. 2013;18(6):342–5.

    PubMed  Google Scholar 

  102. Lopes S, Mesquita-Bastos J, Garcia C, Leitão C, Ribau V, Teixeira M, et al. Aerobic exercise improves central blood pressure and blood pressure variability among patients with resistant hypertension: results of the EnRicH trial. Hyperten Res. 2023;46:1547–57.

    CAS  Google Scholar 

  103. Piepoli MF, Hoes AW, Agewall S, Albus C, Brotons C, Catapano AL, et al. 2016 European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts)Developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR). Eur Heart J. 2016;37(29):2315–81.

    PubMed  PubMed Central  Google Scholar 

  104. Yang YJ. An overview of current physical activity recommendations in primary care. Korean J Fam Med. 2019;40(3):135–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Ahmed SB, Fisher ND, Stevanovic R, Hollenberg NK. Body mass index and angiotensin-dependent control of the renal circulation in healthy humans. Hypertension. 2005;46(6):1316–20.

    CAS  PubMed  Google Scholar 

  106. Zhai F, Wang H, Wang Z, Popkin BM, Chen C. Closing the energy gap to prevent weight gain in China. Obes Rev. 2008;9(Suppl 1):107–12.

    PubMed  Google Scholar 

  107. Mozaffarian D, Hao T, Rimm EB, Willett WC, Hu FB. Changes in diet and lifestyle and long-term weight gain in women and men. N Engl J Med. 2011;364(25):2392–404.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Neter JE, Stam BE, Kok FJ, Grobbee DE, Geleijnse JM. Influence of weight reduction on blood pressure: a meta-analysis of randomized controlled trials. Hypertension. 2003;42(5):878–84.

    CAS  PubMed  Google Scholar 

  109. Aucott L, Poobalan A, Smith WC, Avenell A, Jung R, Broom J. Effects of weight loss in overweight/obese individuals and long-term hypertension outcomes: a systematic review. Hypertension. 2005;45(6):1035–41.

    CAS  PubMed  Google Scholar 

  110. Siebenhofer A, Jeitler K, Berghold A, Waltering A, Hemkens LG, Semlitsch T, et al. Long-term effects of weight-reducing diets in hypertensive patients. Cochrane Database Syst Rev. 2011(9):Cd008274.

  111. Rimm EB, Williams P, Fosher K, Criqui M, Stampfer MJ. Moderate alcohol intake and lower risk of coronary heart disease: meta-analysis of effects on lipids and haemostatic factors. Bmj. 1999;319(7224):1523–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Mukamal KJ, Chen CM, Rao SR, Breslow RA. Alcohol consumption and cardiovascular mortality among U.S. adults, 1987 to 2002. J Am Coll Cardiol. 2010;55(13):1328–35.

    PubMed  Google Scholar 

  113. Xin X, He J, Frontini MG, Ogden LG, Motsamai OI, Whelton PK. Effects of alcohol reduction on blood pressure: a meta-analysis of randomized controlled trials. Hypertension. 2001;38(5):1112–7.

    CAS  PubMed  Google Scholar 

  114. Roerecke M, Kaczorowski J, Tobe SW, Gmel G, Hasan OSM, Rehm J. The effect of a reduction in alcohol consumption on blood pressure: a systematic review and meta-analysis. Lancet Public Health. 2017;2(2):e108–20.

    PubMed  PubMed Central  Google Scholar 

  115. Dickinson HO, Mason JM, Nicolson DJ, Campbell F, Beyer FR, Cook JV, et al. Lifestyle interventions to reduce raised blood pressure: a systematic review of randomized controlled trials. J Hypertens. 2006;24(2):215–33.

    CAS  PubMed  Google Scholar 

  116. National Institute on Alcohol Abuse and Alcoholism. What Is a standard drink? 2018;2023; Available from: https://www.niaaa.nih.gov/alcohols-effects-health/overview-alcohol-consumption/what-standard-drink.

  117. Primatesta P, Falaschetti E, Gupta S, Marmot MG, Poulter NR. Association between smoking and blood pressure: evidence from the health survey for England. Hypertension. 2001;37(2):187–93.

    CAS  PubMed  Google Scholar 

  118. Cohen DL, Townsend RR. Does cigarette use modify blood pressure measurement or the effectiveness of blood pressure medications? J Clin Hypertens (Greenwich). 2009;11(11):657–8.

    PubMed  PubMed Central  Google Scholar 

  119. Li G, Wang H, Wang K, Wang W, Dong F, Qian Y, et al. The association between smoking and blood pressure in men: a cross-sectional study. BMC Public Health. 2017;17(1):797.

    PubMed  PubMed Central  Google Scholar 

  120. Sahle BW, Chen W, Rawal LB, Renzaho AMN. Weight gain after smoking cessation and risk of major chronic diseases and mortality. JAMA Netw Open. 2021;4(4): e217044.

    PubMed  PubMed Central  Google Scholar 

  121. Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, Adair-Rohani H, et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2224–60.

    PubMed  PubMed Central  Google Scholar 

  122. Roush GC, Ernst ME, Kostis JB, Tandon S, Sica DA. Head-to-head comparisons of hydrochlorothiazide with indapamide and chlorthalidone: antihypertensive and metabolic effects. Hypertension. 2015;65(5):1041–6.

    CAS  PubMed  Google Scholar 

  123. Cirillo M, Marcarelli F, Mele AA, Romano M, Lombardi C, Bilancio G. Parallel-group 8-week study on chlorthalidone effects in hypertensives with low kidney function. Hypertension. 2014;63(4):692–7.

    CAS  PubMed  Google Scholar 

  124. Agarwal R, Sinha AD, Cramer AE, Balmes-Fenwick M, Dickinson JH, Ouyang F, et al. Chlorthalidone for hypertension in advanced chronic kidney disease. N Engl J Med. 2021;385(27):2507–19.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Rossignol P, Massy ZA, Azizi M, Bakris G, Ritz E, Covic A, et al. The double challenge of resistant hypertension and chronic kidney disease. Lancet. 2015;386(10003):1588–98.

    PubMed  Google Scholar 

  126. Tamargo J, Segura J, Ruilope LM. Diuretics in the treatment of hypertension. Part 2: loop diuretics and potassium-sparing agents. Expert Opin Pharmacother. 2014;15(5):605–21.

    CAS  PubMed  Google Scholar 

  127. Williams B, MacDonald TM, Morant S, Webb DJ, Sever P, McInnes G, et al. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): a randomised, double-blind, crossover trial. Lancet. 2015;386(10008):2059–68.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Williams B, MacDonald TM, Morant SV, Webb DJ, Sever P, McInnes GT, et al. Endocrine and haemodynamic changes in resistant hypertension, and blood pressure responses to spironolactone or amiloride: the PATHWAY-2 mechanisms substudies. Lancet Diabetes Endocrinol. 2018;6(6):464–75.

    PubMed  PubMed Central  Google Scholar 

  129. Agarwal R, Rossignol P, Romero A, Garza D, Mayo MR, Warren S, et al. Patiromer versus placebo to enable spironolactone use in patients with resistant hypertension and chronic kidney disease (AMBER): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet. 2019;394(10208):1540–50.

    CAS  PubMed  Google Scholar 

  130. Agarwal R, Pitt B, Palmer BF, Kovesdy CP, Burgess E, Filippatos G, et al. A comparative post hoc analysis of finerenone and spironolactone in resistant hypertension in moderate-to-advanced chronic kidney disease. Clin Kidney J. 2023;16(2):293–302.

    CAS  PubMed  Google Scholar 

  131. Lee JH, Kim KI, Cho MC. Current status and therapeutic considerations of hypertension in the elderly. Korean J Intern Med. 2019;34(4):687–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Parati G, Esler M. The human sympathetic nervous system: its relevance in hypertension and heart failure. Eur Heart J. 2012;33(9):1058–66.

    CAS  PubMed  Google Scholar 

  133. Evelyn KA, Alexander F, Cooper SR. Effect of sympathectomy on blood pressure in hypertension; a review of 13 years’ experience of the Massachusetts General Hospital. J Am Med Assoc. 1949;140(7):592–602.

    CAS  PubMed  Google Scholar 

  134. Krum H, Schlaich MP, Sobotka PA, Böhm M, Mahfoud F, Rocha-Singh K, et al. Percutaneous renal denervation in patients with treatment-resistant hypertension: final 3-year report of the Symplicity HTN-1 study. Lancet. 2014;383(9917):622–9.

    PubMed  Google Scholar 

  135. Esler MD, Böhm M, Sievert H, Rump CL, Schmieder RE, Krum H, et al. Catheter-based renal denervation for treatment of patients with treatment-resistant hypertension: 36 month results from the SYMPLICITY HTN-2 randomized clinical trial. Eur Heart J. 2014;35(26):1752–9.

    PubMed  PubMed Central  Google Scholar 

  136. Bhatt DL, Kandzari DE, O’Neill WW, D’Agostino R, Flack JM, Katzen BT, et al. A controlled trial of renal denervation for resistant hypertension. N Engl J Med. 2014;370(15):1393–401.

    CAS  PubMed  Google Scholar 

  137. Townsend RR, Mahfoud F, Kandzari DE, Kario K, Pocock S, Weber MA, et al. Catheter-based renal denervation in patients with uncontrolled hypertension in the absence of antihypertensive medications (SPYRAL HTN-OFF MED): a randomised, sham-controlled, proof-of-concept trial. Lancet. 2017;390(10108):2160–70.

    PubMed  Google Scholar 

  138. Azizi M, Schmieder RE, Mahfoud F, Weber MA, Daemen J, Davies J, et al. Endovascular ultrasound renal denervation to treat hypertension (RADIANCE-HTN SOLO): a multicentre, international, single-blind, randomised, sham-controlled trial. Lancet. 2018;391(10137):2335–45.

    PubMed  Google Scholar 

  139. Azizi M, Saxena M, Wang Y, Jenkins JS, Devireddy C, Rader F, et al. Endovascular ultrasound renal denervation to treat hypertension: The RADIANCE II randomized clinical trial. Jama. 2023;329(8):651–61.

    PubMed  PubMed Central  Google Scholar 

  140. Schmieder RE, Mahfoud F, Mancia G, Azizi M, Böhm M, Dimitriadis K, et al. European Society of Hypertension position paper on renal denervation 2021. J Hypertens. 2021;39(9):1733–41.

    CAS  PubMed  Google Scholar 

  141. Azizi M, Sapoval M, Gosse P, Monge M, Bobrie G, Delsart P, et al. Optimum and stepped care standardised antihypertensive treatment with or without renal denervation for resistant hypertension (DENERHTN): a multicentre, open-label, randomised controlled trial. Lancet. 2015;385(9981):1957–65.

    PubMed  Google Scholar 

  142. Kandzari DE, Böhm M, Mahfoud F, Townsend RR, Weber MA, Pocock S, et al. Effect of renal denervation on blood pressure in the presence of antihypertensive drugs: 6-month efficacy and safety results from the SPYRAL HTN-ON MED proof-of-concept randomised trial. Lancet. 2018;391(10137):2346–55.

    PubMed  Google Scholar 

  143. Azizi M, Sanghvi K, Saxena M, Gosse P, Reilly JP, Levy T, et al. Ultrasound renal denervation for hypertension resistant to a triple medication pill (RADIANCE-HTN TRIO): a randomised, multicentre, single-blind, sham-controlled trial. Lancet. 2021;397(10293):2476–86.

    CAS  PubMed  Google Scholar 

  144. Mahfoud F, Kandzari DE, Kario K, Townsend RR, Weber MA, Schmieder RE, et al. Long-term efficacy and safety of renal denervation in the presence of antihypertensive drugs (SPYRAL HTN-ON MED): a randomised, sham-controlled trial. Lancet. 2022;399(10333):1401–10.

    CAS  PubMed  Google Scholar 

  145. Mancia G, Kreutz Co-Chair R, Brunström M, Burnier M, Grassi G, Januszewicz A, et al. 2023 ESH Guidelines for the management of arterial hypertension The Task Force for the management of arterial hypertension of the European Society of Hypertension Endorsed by the European Renal Association (ERA) and the International Society of Hypertension (ISH). J Hypertens. 2023.

  146. Heusser K, Tank J, Engeli S, Diedrich A, Menne J, Eckert S, et al. Carotid baroreceptor stimulation, sympathetic activity, baroreflex function, and blood pressure in hypertensive patients. Hypertension. 2010;55(3):619–26.

    CAS  PubMed  Google Scholar 

  147. Bisognano JD, Bakris G, Nadim MK, Sanchez L, Kroon AA, Schafer J, et al. Baroreflex activation therapy lowers blood pressure in patients with resistant hypertension: results from the double-blind, randomized, placebo-controlled rheos pivotal trial. J Am Coll Cardiol. 2011;58(7):765–73.

    PubMed  Google Scholar 

  148. Hoppe UC, Brandt MC, Wachter R, Beige J, Rump LC, Kroon AA, et al. Minimally invasive system for baroreflex activation therapy chronically lowers blood pressure with pacemaker-like safety profile: results from the Barostim neo trial. J Am Soc Hypertens. 2012;6(4):270–6.

    PubMed  Google Scholar 

  149. Wallbach M, Born E, Kämpfer D, Lüders S, Müller GA, Wachter R, et al. Long-term effects of baroreflex activation therapy: 2-year follow-up data of the BAT Neo system. Clin Res Cardiol. 2020;109(4):513–22.

    CAS  PubMed  Google Scholar 

  150. Peter DA, Alemu Y, Xenos M, Weisberg O, Avneri I, Eshkol M, et al. Fluid structure interaction with contact surface methodology for evaluation of endovascular carotid implants for drug-resistant hypertension treatment. J Biomech Eng. 2012;134(4): 041001.

    PubMed  Google Scholar 

  151. Spiering W, Williams B, Van der Heyden J, van Kleef M, Lo R, Versmissen J, et al. Endovascular baroreflex amplification for resistant hypertension: a safety and proof-of-principle clinical study. Lancet. 2017;390(10113):2655–61.

    PubMed  Google Scholar 

  152. van Kleef M, Devireddy CM, van der Heyden J, Bates MC, Bakris GL, Stone GW, et al. Treatment of resistant hypertension with endovascular baroreflex amplification: 3-year results from the CALM-FIM study. JACC Cardiovasc Interv. 2022;15(3):321–32.

    PubMed  Google Scholar 

  153. Mahfoud F, Azizi M, Ewen S, Pathak A, Ukena C, Blankestijn PJ, et al. Proceedings from the 3rd European clinical consensus conference for clinical trials in device-based hypertension therapies. Eur Heart J. 2020;41(16):1588–99.

    PubMed  PubMed Central  Google Scholar 

  154. Hoshide S, Kario K, Chia YC, Siddique S, Buranakitjaroen P, Tsoi K, et al. Characteristics of hypertension in obstructive sleep apnea: An Asian experience. J Clin Hypertens (Greenwich). 2021;23(3):489–95.

    PubMed  PubMed Central  Google Scholar 

  155. Parati G, Ochoa JE, Bilo G, Mattaliano P, Salvi P, Kario K, et al. Obstructive sleep apnea syndrome as a cause of resistant hypertension. Hypertens Res. 2014;37(7):601–13.

    PubMed  Google Scholar 

  156. Chiu KL, Ryan CM, Shiota S, Ruttanaumpawan P, Arzt M, Haight JS, et al. Fluid shift by lower body positive pressure increases pharyngeal resistance in healthy subjects. Am J Respir Crit Care Med. 2006;174(12):1378–83.

    PubMed  Google Scholar 

  157. Martínez-García MA, Capote F, Campos-Rodríguez F, Lloberes P, Díaz de Atauri MJ, Somoza M, et al. Effect of CPAP on blood pressure in patients with obstructive sleep apnea and resistant hypertension: the HIPARCO randomized clinical trial. Jama. 2013;310(22):2407-15.

  158. Becker HF, Jerrentrup A, Ploch T, Grote L, Penzel T, Sullivan CE, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation. 2003;107(1):68–73.

    PubMed  Google Scholar 

  159. Sapiña-Beltrán E, Benitez ID, Torres G, Fortuna-Gutiérrez AM, Ponte Márquez P, Masa JF, et al. Effect of CPAP treatment on BP in resistant hypertensive patients according to the BP dipping pattern and the presence of nocturnal hypertension. Hypertens Res. 2022;45(3):436–44.

    PubMed  Google Scholar 

  160. Dudenbostel T, Siddiqui M, Oparil S, Calhoun DA. Refractory hypertension: a novel phenotype of antihypertensive treatment failure. Hypertension. 2016;67(6):1085–92.

    CAS  PubMed  Google Scholar 

  161. Cardoso CRL, Salles GF. Refractory Hypertension and risks of adverse cardiovascular events and mortality in patients with resistant hypertension: a prospective cohort study. J Am Heart Assoc. 2020;9(17): e017634.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Velasco A, Siddiqui M, Kreps E, Kolakalapudi P, Dudenbostel T, Arora G, et al. Refractory hypertension is not attributable to intravascular fluid retention as determined by intracardiac volumes. Hypertension. 2018;72(2):343–9.

    CAS  PubMed  Google Scholar 

  163. Salvador VD, Bakris GL. Novel antihypertensive agents for resistant hypertension: what does the future hold? Hypertens Res. 2022.

Download references

Acknowledgements

None

Funding

This study was supported by a research program funded by the Korea Disease Control and Prevention Agency [grant number 2021-ER0903-02].

Author information

Authors and Affiliations

Authors

Contributions

SHI, SP, and SJ conceived the project and wrote and reviewed the manuscript; KK, HK, HCK, EML, JHL, SYA, EJC, JHK, HTK, HYL, SKL, and WHK wrote and reviewed the manuscript; JMP reviewed and revised the manuscript.

Corresponding author

Correspondence to Sang Hyun Ihm.

Ethics declarations

Ethics approval and consent to participate

“Not applicable”

Consent for Publication

“Not applicable”

Competing interests

S.P. received honoraria from Viatris, Organon, Boryoung, Hanmi, Daewoong, Donga, Celltrion, Servier, Daiichi Sankyo, Chong Kun Dang, and Daewon, and a research grant from Daiichi Sankyo. J S received grants from Sanofi and Hanmi, and honoraria from Sankyo, Menarini, Daewoong, Boryong, Daewon, Bayer, Viatris, and Dongwha; S.H.I received honoraria from Boryoung, Hanmi, Daewoong, Donga ST, Celltrion, and Daiichi Sankyo, and a research grant from Donga ST. EML received grants from Sanofi and Boryong and honoraria from Viatris, Hanmi, Chongkundang, Norvatis, Green Cross, and Bayer. JHK received honoraria from Aju, Boryoung, Chong Kun Dang, Celltrion, Donga Daewoong, Daiichi Sankyo and Yuhan. HTK received grants from JW Pharmaceuticals. HYL received grants from Sanofi, Hanmi, and Honoraria, Sankyo, Menarini, Daewoong, Boryong, Daewon, Bayer, Viatris, and Dongwha. KK, HK, HCK, JHL, SKL, and WHK have no conflicts of interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, S., Shin, J., Ihm, S.H. et al. Resistant hypertension: consensus document from the Korean society of hypertension. Clin Hypertens 29, 30 (2023). https://doi.org/10.1186/s40885-023-00255-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40885-023-00255-4

Keywords