Skip to main content
Critical Care and Resuscitation logoLink to Critical Care and Resuscitation
. 2022 Dec 5;24(4):298–308. doi: 10.51893/2022.4.R

Midodrine use in critically ill patients: a narrative review

Rahul Costa-Pinto 1,2, Daryl A Jones 1,2, Andrew A Udy 3,4, Stephen J Warrillow 1,2, Rinaldo Bellomo 1,2,4,5
PMCID: PMC10692611  PMID: 38047013

Abstract

Midodrine is a peripherally acting, oral α-agonist that is increasingly used in intensive care units despite conflicting evidence for its effectiveness. It has pharmacological effects on blood vessels as well as pupillary, cardiac, renal, gastrointestinal, genitourinary, lymphatic and skin tissue. It has approval for use as a treatment for orthostatic hypotension, but a surge in interest over the past decade has prompted its use for a growing number of off-label indications. In critically ill patients, midodrine has been used as either an adjunctive oral therapy to wean vasoplegic patients off low dose intravenous vasopressor infusions, or as an oral vasopressor agent to prevent or minimise the need for intravenous infusion. Clinical trials have mostly focused on midodrine as an intravenous vasopressor weaning agent. Early retrospective studies supported its use for this indication, but more recent randomised controlled trials have largely refuted this practice. Key questions remain on its role in managing critically ill patients before intensive care admission, during intensive care stay, and following discharge. This narrative review presents a comprehensive overview of midodrine use for the critical care physician and highlights why lingering questions around ideal patient selection, dosing, timing of initiation, and efficacy of midodrine for critically ill patients remain unanswered.


Midodrine is an oral vasopressor agent that is receiving increasing interest as a therapy to reduce intensive care unit (ICU) admission and length of stay for patients who would otherwise require intravenous vasopressor infusions and invasive monitoring. Although usage trends increase, evidence for its effectiveness is conflicting. Adequacy and frequency of dosage, timing of initiation and patient selection are important factors to consider when prescribing midodrine for critically ill patients. This narrative review explores the historical context of midodrine usage, its pharmacological properties, current trends in use both within and outside the critical care environment, evidence to support its use, and finally, future research directions.

Historical context

Midodrine was patented in 1965 by Chemie Linz AG1 in Linz, Austria, and was first described in the medical literature in the 1970s as a novel peripherally acting α-agonist with good enteral absorption, efficacy and a long duration of action.2 Animal experiments revealed that α-(2,5-dimethoxyphenyl)- ß-glycinamido-ethanol hydrochloride, or midodrine, and its active metabolite α-(2,5-dimethoxyphenyl)-ß-aminoethanol (ST-1059 or desglymidodrine) effectively increase peripheral vascular tone and stimulate α-adrenergic receptors in intestine, bladder, bronchi and pupils2 without directly affecting cerebral blood flow.3

Plasma levels of the active metabolite, desglymidodrine, were significantly correlated with pressor activity,4 and midodrine's reported venoconstrictive effect was 50-80% of noradrenaline-induced venoconstriction in vitro.5, 6 Unlike other sympathomimetic agents with pressor effects, midodrine was equally efficacious in parenteral and enteral formulations.2

Subsequent observational studies found clinical utility for midodrine's α-sympathomimetic action and ease of oral administration for conditions such as urinary stress incontinence7, 8 and ejaculation disorders,9, 10 as well as orthostatic hypotension related to neurological conditions,11 neuroleptic medications12 and idiopathic postural hypotension in paediatric and adult populations.13, 14, 15

These pilot studies typically used oral doses of 2.5–5 mg two or three times daily. An early observational study also demonstrated midodrine's safety and efficacy in increasing blood pressure in children with septic shock.16 Most of these early, small observational and double-blind studies reported minimal adverse events.

Larger clinical trials in the 1990s established midodrine as a safe and effective agent for orthostatic hypotension.17, 18, 19, 20 The first multicentre, double-blind, randomised controlled trial (RCT) evaluating the use of midodrine for moderate to severe orthostatic hypotension was conducted in the United States and published in 1993.17 This study assigned its 97 patients to either receive placebo, or midodrine at doses of 2.5 mg, 5 mg or 10 mg over a 4-week period. At 10 mg doses, midodrine increased standing systolic blood pressure by 28% and, at all doses, significantly improved symptoms of dizziness, weakness and syncope. A larger double-blind study of 171 patients, administering midodrine at 10 mg three times daily for a 4-week period found a similar increase in standing systolic blood pressure (24% mean increase) and reduction in mean symptom score for lightheadedness.19 These studies paved the way for midodrine to receive United States Food and Drug Administration (FDA) approval in 1996 for symptomatic orthostatic hypotension via its Accelerated Approval Program.

Other emerging uses for midodrine were also reported around this time. Midodrine as a pre-medication for chronic hypotension associated with haemodialysis was shown to be safe and provided extended haemodynamic and symptomatic benefit in doses ranging from 2.5 mg to 25 mg.21, 22, 23, 24 Midodrine for reversal of hepatorenal syndrome was also described25, 26 to improve renal plasma flow and glomerular filtration rate with improved one-month survival.

In 2010, however, the FDA decided to withdraw midodrine from the market due to the failure of its manufacturers to conduct any post-marketing studies to confirm clinical benefit for orthostatic hypotension.27 Health care professional appeals and consumer complaints led to this action being delayed28, 29 pending phase 4 trials. A phase 4, double-blind, placebo-controlled, randomised tilt-table study was finally published in 2016 which showed that patients receiving stable doses of midodrine for more than 3 months had a statistically significant increase in time to tilt-table-induced syncopal symptoms.30 Nevertheless, this scrutiny stimulated interest to demonstrate midodrine's efficacy across many patient groups and clinical settings, with more than half of all published literature on midodrine appearing since this time.

Pharmacology of midodrine

Midodrine is a peripherally acting α-receptor agonist available as 2.5 mg and 5 mg tablets. It does not act preferentially on either α1,- or α2-receptors,31 but its active metabolite, desglymidodrine, selectively stimulates α1-receptors.32 It causes modest increases in supine and standing blood pressure in a dose-dependent manner.2, 20 Its other pharmacodynamic effects are to increase peripheral vascular resistance, increase venous tone and release of atrial natriuretic peptide,33 and reduce circulating plasma and blood volume31 (Figure 1).

Figure 1.

Figure 1

Pharmacological effects of midodrine in various organ systems

Midodrine has poor blood-brain barrier penetration34 and, therefore, no direct central nervous system activity. It has no myocardial ß-adrenoreceptor activity but indirectly increases end-diastolic volume and stroke volume, decreases heart rate and circulating noradrenaline levels via baroreceptor stimulation,35, 36 and causes QT prolongation.37 It has no significant metabolic or endocrine effects. It has no effect on serum lipids, insulin, or uric acid levels.38 It also does not have any established effect on pulmonary, renal,34 coagulation39 or immune function.40 It has been safely administered in pregnancy.41, 42

Desglymidodrine, the active metabolite, is generated from midodrine by the enzymatic cleavage of the amino acid glycine. The oral bioavailability of desglymidodrine is 93%. The mean maximum concentration in plasma for midodrine is 20–30 minutes after oral administration and 60 minutes for desglymidodrine.43 Binding to plasma proteins is less than 30%. Midodrine is cleared from plasma after 2 hours,31 with an elimination half-life of 30 minutes.43 The elimination half-life of desglymidodrine is 3 hours.43

Midodrine undergoes extensive metabolism in various tissues including the liver (predominantly by cytochrome P450 isoforms CYP2D6 and CYP1A244), with only 4% of a single dose excreted unchanged.31 Excretion of midodrine and desglymidodrine is primarily urinary. Haemodialysis can reduce the elimination half-life of desglymidodrine to 90 minutes. In end-stage chronic kidney disease, the elimination half-life can be as long as 10 hours.45

Common adverse effects are related to midodrine's α-agonist properties. Pilomotor reactions (piloerection, scalp pruritus) are the most frequently reported adverse effects followed by gastrointestinal and genitourinary complaints (nausea, abdominal pain, urinary retention, dysuria), cardiovascular effects (supine hypertension, bradycardia) and central nervous system effects (paraesthesia, taste and smell disturbance). Although up to 80% of patients may experience one or more of these adverse effects,46 they are dose-dependent and generally mild. Singular case reports describe midodrine use associated with takotsubo cardiomyopathy,47 intracerebral haemorrhage,48 reversible cerebral vasoconstriction syndrome,49 myoclonic seizures,50 vascular ischaemia,51 and ileus.52

In critical care settings, when administered as an intravenous vasopressor weaning agent, the most common adverse effect is reflex bradycardia53 which is proportional to midodrine dose.54 Drug interactions may occur with concomitant prescription of antiarrhythmics, ß-blockers, antipsychotics, monoamine oxidase inhibitors and tricyclic antidepressants metabolised by cytochrome CYP2D655 as well as ranitidine, metformin and procainamide, which compete with desglymidodrine at acute tubular secretion sites in the kidney.56

Midodrine daily doses of up to 120 mg (in divided doses) have been reported in the literature with no adverse effects, even in patients with end-stage chronic kidney disease.53, 57, 58 Overdosage may present as severe hypertension, bradycardia, urinary retention and piloerection.59 Hypertension can be managed with nitrovasodilator or α-sympatholytic infusions (glyceryl trinitrate, sodium nitroprusside, phentolamine). Bradycardia can be managed with atropine.

Midodrine use outside critical care settings

Aside from its well established uses for orthostatic hypotension and neurocardiogenic syncope,60 midodrine has been used off-label for multiple clinical indications over the past 20 years. Case reports and case series report its use to maintain normotension in patients with a spinal cord injury61 and following acute myocardial infarction,62 to correct dysautonomia in chronic fatigue syndrome,63 to decrease left ventricular outflow tract obstruction by improving filling in hypertrophic cardiomyopathy,64 to decrease severity of shunt and hypoxemia in patients with right-to-left intracardiac shunting,65 to increase systemic vascular resistance and reduce pulmonary pressures in porto-pulmonary hypertension,66 and to decrease lymphatic flow in refractory chylothorax.67, 68

Small prospective studies have demonstrated its use to maintain perfusion pressure in acute stroke69 and following carotid endarterectomy,70 to mitigate treatment-induced hypotension in advanced heart failure,71 to enhance exercise tolerance and sexual function in patients with a spinal cord injury,72, 73, 74 to reduce severity of symptoms in refractory gastro-oesophageal reflux disease,75 and to manage orthostatic intolerance with mobilisation following total hip and knee replacement.76

Randomised trials weakly support the use of midodrine for stress urinary incontinence in women.77 There are conflicting RCT data for its efficacy in preventing paracentesis-induced circulatory dysfunction78, 79, 80, 81 and recurrence of ascites as a substitute for albumin in patients with cirrhosis.82, 83

There is a much larger number of studies and now meta-analyses and systematic reviews that support its use as a treatment for orthostatic hypotension and recurrent vasovagal syncope,84, 85 intradialytic hypotension86 and hepatorenal syndrome.87 However, even for these indications, the pooled evidence for midodrine has often been inconsistent and of low quality. Midodrine use for intradialytic hypotension is associated with higher pre-transplant rates of all-cause hospitalisation, cardiovascular hospitalisation, and death88 as well as poorer post-transplant outcomes.89, 90 It is also less effective in improving renal outcomes and survival in type 1 hepatorenal syndrome than terlipressin or noradrenaline.91, 92

Rationale and evidence for midodrine in the ICU

The use of midodrine in the ICU was first described in 2002 for a patient following an emergency multilevel laminectomy for acute thoracic spinal cord compression. Postoperatively, it appeared to be an effective noradrenaline substitute, negating the requirement for central venous access and reducing ICU length of stay.93

Midodrine use in critically ill patients, thereafter, has mostly been as either an adjunctive oral therapy to wean vasoplegic patients off low dose intravenous vasopressor infusions, or as an oral vasopressor agent to prevent or minimise the need for intravenous infusion.53 There are several reasons why these remain attractive indications. Firstly, midodrine has a reasonable safety profile and is relatively inexpensive (less than $1.00 per 5 mg tablet). Secondly, effective use of an oral vasopressor may avoid the potential complications of central line insertion and catheter-related bloodstream infections.94 Thirdly, oral vasopressors may offer an alternative for patients with comorbid conditions not suitable for ICU admission or as a palliative strategy for patients discharged from the ICU.95 Finally, shortening the duration of intravenous vasopressor support may decrease ICU and hospital length of stay, reducing cost and improving health care access.96

Clinical trials have mostly focused on midodrine as an intravenous vasopressor weaning agent (Table 1). Early retrospective studies used modal doses of 10–20 mg 8-hourly for patients requiring intravenous vasopressors for septic shock, trauma and cardiovascular diagnoses and showed that intravenous vasopressor discontinuation occurred a median of 1.2–2.9 days after midodrine initiation or, alternatively, midodrine reduced intravenous vasopressor duration by up to 25%.58, 97, 105 The most commonly weaned vasopressor infusions were phenylephrine and noradrenaline, but patients were also weaned off adrenaline, dopamine and vasopressin. These non-randomised trials showed midodrine could be safely administered in critically ill patients in doses ranging from 10 mg 8-hourly to 40 mg 8-hourly.

Table 1.

All published retrospective, prospective and randomised controlled trials of midodrine as a vasopressor weaning agent in intensive care units (ICUs)*

First author (year) Design Setting (sample size) Midodrine dose (frequency) IV VP weaned Results
Retrospective studies
Whitson58 (2016) Observational, controlled Medical ICU (n = 275) 10-40 mg (8-hourly) Noradrenaline, phenylephrine
  • Shorter duration of IV VP (mean, 2.9 v 3.8 days; P < 0.001)

  • Shorter ICU LOS (mean, 7.5 v 9.4 days; P = 0.017)

Poveromo97 (2016) Observational, controlled Mixed ICU (n = 188) 2.5-10 mg (4- to 12-hourly) Adrenaline, dopamine, noradrenaline, phenylephrine, vasopressin
  • Lower IV VP dose at 24 hours (median, reduction of 97.3%)

  • No difference in ICU LOS (median, 5.5 v 5.0 days; P = 0.29)

Rizvi53 (2018) Observational, uncontrolled Mixed ICU (n = 1119) 5-30 mg (8-hourly) Adrenaline, dopamine, noradrenaline, phenylephrine, vasopressin
  • Fewer patients on IV VP at 24 hours (663 v 344; P < 0.001)

Tremblay98 (2020) Observational, propensity matched Cardiac ICU (n = 148) Mostly 10 mg (8-hourly) Adrenaline, noradrenaline, vasopressin
  • No difference in duration of IV VP (median, 63 v 44 hours; P = 0.052)

  • Longer ICU LOS (median, 99 v 68 hours; P = 0.001)

Macielak99 (2021) Observational, uncontrolled Mixed ICU (n = 44) 5-20 mg (6-hourly) Adrenaline, dopamine, noradrenaline, phenylephrine, vasopressin
  • Lower IV VP dose at 24 hours (mean, reduction of 40%)

Prospective studies
Levine100 (2013) Observational, uncontrolled Surgical ICU (n = 20) 5-20 mg (8-hourly) Noradrenaline, phenylephrine
  • Faster decline in IV VP rate after four doses (mean, –0.62 v –2.20 μg/min; P = 0.012)

Randomised controlled trials
Santer101 (2020) Randomised, placebo controlled Mixed ICU (n = 132) 20 mg (8-hourly) Metaraminol, noradrenaline, phenylephrine
  • No difference in duration of IV VP (median, 23.5 v 22.5 hours; P = 0.62)

  • No difference in ICU LOS (median, 6 v 6 days; P = 0.46)

Lal102 (2021) Randomised, placebo controlled Medical ICU (n = 32) 10 mg (8-hourly) Adrenaline, dopamine, noradrenaline, phenylephrine, vasopressin
  • No difference in duration of IV VP (median, 14.5 v 18.8 hours; P = 0.19)

  • No difference in ICU LOS (median, 2.29 v 2.45 days; P = 0.36)

Costa-Pinto54 (2022) Randomised, open-label controlled Mixed ICU (n = 62) 10 mg (8-hourly) Metaraminol, noradrenaline
  • No difference in duration of IV VP (median, 16.5 v 19.0 hours; P = 0.22)

  • No difference in ICU LOS (median, 50 v 59 hours; P = 0.14)

Adly103 (2022) Randomised, open-label controlled Medical ICU (n = 60) 10 mg (8-hourly) Noradrenaline
  • Shorter duration of IV VP (median, 26.0 v 78.5 hours; P < 0.001)

  • No difference in ICU LOS (mean, 11.9 v 11.5 days; P = 0.876)

Ahmed Ali104 (2022) Randomised, open-label controlled Surgical ICU (n = 90) 10 mg (8-hourly) Noradrenaline
  • Shorter duration of IV VP (mean, 3.30 v 6.93 days; P < 0.001)

  • Shorter ICU LOS (mean, 5.13 v 9.03 days; P < 0.001)

IV = intravenous; LOS = length of stay; VP = vasopressor.

*

Abstracts not included.

In contrast, the largest retrospective study of midodrine use as a vasopressor weaning agent included 2070 patients (209 adjunctive midodrine patients, 1861 intravenous vasopressor-only patients) with predominantly septic shock and found a longer intravenous vasopressor duration in the midodrine group and no difference in ICU or hospital length of stay.106 This study enrolled patients who required intravenous vasopressor for more than 7 days, so it is likely they received midodrine later in their ICU stay or had more persistent refractory vasoplegia. In combination, findings from these retrospective studies did not support the use of midodrine as a weaning agent.107

Another retrospective study of 74 cardiothoracic surgery patients who received midodrine to wean intravenous vasopressor support also found no difference in length of vasopressor duration compared with a propensity score matched control group. Of concern, however, midodrine use was associated with longer ICU length of stay and higher mortality in this study.98

The first prospective study examining this indication for midodrine was an observational study of 20 surgical ICU patients who received a modal dose of 20 mg (range, 5–20 mg) 8-hourly to wean off phenylephrine or noradrenaline infusions.100 Midodrine significantly reduced the dose of intravenous vasopressors and 70% of patients were completely weaned after four doses of midodrine. Clearly, given these mixed findings across relatively small retrospective and prospective studies, large RCTs were required to answer this important clinical question.

The first double-blind RCT to investigate the efficacy of midodrine as an intravenous vasopressor weaning agent, the MIDAS study, was registered in 2012 (ClinicalTrials.gov NCT01531959) and published in 2020.101 This international, multicentre study included all hypotensive (systolic blood pressure < 90 mmHg) patients requiring single agent intravenous vasopressor support for more than 24 hours across three tertiary referral hospitals. Exclusion criteria included high dose vasopressor support (ie, noradrenaline > 8 μg/min, phenylephrine > 100 μg/min, metaraminol > 60 μg/min), patients with ongoing clinical evidence of shock, and chronic kidney, liver and heart disease. Patients were randomised to receive either 20 mg midodrine 8-hourly, or placebo until 24 hours after cessation of intravenous vasopressor.

Overall, 132 patients were randomised in a 1:1 ratio over 7 years. Midodrine use was not associated with any differences in time to intravenous vasopressor cessation (median, 23.5 [interquartile range (IQR), 10.0–54.0] v 22.5 [IQR, 10.4–40.0] hours) nor ICU or hospital length of stay when compared with placebo.

The MAVERIC study,54 a multicentre open-label RCT, used similar inclusion and exclusion criteria but utilised a lower midodrine dose (10 mg 8-hourly) and reported similar findings to the MIDAS study. The median time to discontinuation of intravenous vasopressor was 16.5 hours (IQR, 7.5–27.5 hours) in the midodrine group and 19 hours (IQR, 12.25–38.5 hours) in the control group (P = 0.32). Again, ICU and hospital length of stay were similar between groups.

In contrast to these two negative RCTs, a single centre openlabel RCT including 60 patients in Egypt103 found a striking difference in time to intravenous vasopressor cessation using midodrine 10 mg 8-hourly for patients with septic shock receiving stable low dose intravenous vasopressor for at least 24 hours at the time of randomisation. The median time to intravenous vasopressor cessation was 26 hours (IQR, 14–106 hours) in the midodrine group and 78.5 hours (IQR, 32–280 hours) in the control group. However, in-hospital mortality was very high across both groups in this study (43.3% in the midodrine group and 73.3% in the control group), which makes generalisability of these results, without adjusting for mortality, problematic.

The MIDAS and MAVERIC studies strongly question the utility of midodrine as an intravenous vasopressor weaning agent. One postulated mechanism for this lack of effect is that in patients with chronic hypotension, increased baroreceptor sensitivity (baroreceptor habituation) may limit the utility of midodrine administration. These patients have increased heart rate variability and respiratory sinus arrhythmia compared with healthy control subjects, suggesting both increased parasympathetic cardiac tone and reduced sympathetic activity.108 In turn, there is no significant increase in cardiac output following midodrine administration in this patient population.109

Other postulated mechanisms for midodrine's lack of efficacy as a vasopressor weaning agent include the multifactorial aetiology of hypotension in critically ill patients, downregulation of adrenergic receptors with chronic vasopressor infusions,110 and unpredictable oral absorption due to gastrointestinal tract oedema or intestinal vasoconstriction.111 Partial or complete interruption of cardiovascular innervation (such as that seen in tetraplegia)112 as well as central arterial stiffness113 will also affect individual responses to midodrine.

Further considerations in the critically ill patient

One limitation of all RCT evidence thus far for midodrine as a vasopressor weaning agent is protocolised fixed drug dosing.114 Published RCTs have all either utilised 10 mg or 20 mg midodrine at 8-hourly dose intervals. This differs to normal clinical practice where vasopressor support is usually titrated to a mean arterial pressure (MAP) target in real time. An adaptive midodrine dose-titration protocol in an RCT design may help answer this question. However, such a trial would have significant implementation challenges in a critically ill cohort of patients where drug half-life and oral bioavailability may be unpredictable compared with rapidly titratable intravenous therapies.115

An alternative approach could involve increased dosing frequency of midodrine given its 3-hour half-life. A retrospective study of 23 patients receiving midodrine at 6-hourly intervals to wean off intravenous vasopressors99 found this regimen to be safe. Prospective trials are required to test this hypothesis.

Bradycardia appears to be a major limitation for trialling higher doses of midodrine in the ICU. No patients receiving 10 mg midodrine 8-hourly in the MAVERIC study had an episode of severe bradycardia (heart rate < 40 beats per minute). In contrast, 7.6% of patients receiving midodrine 20 mg 8-hourly had an episode of severe bradycardia in the MIDAS study, suggesting a dose-dependent response. Severe bradycardia may prevent ICU discharge regardless of intravenous vasopressor requirements and limit the usefulness of midodrine for this indication.

In both the MIDAS and MAVERIC RCTs, intravenous vasopressors were ceased within 24 hours in the placebo group questioning whether patients who are receiving stable low doses of intravenous vasopressors for more than 24 hours are the group most likely to benefit from adjunctive midodrine. A recently completed multicentre, pilot, feasibility double-blinded RCT of patients with sepsis of less than 24 hours duration102 suggests a larger clinical trial is warranted to explore earlier initiation of midodrine. In this study, 32 patients were randomised to receive three doses of midodrine 10 mg at 8-hourly intervals or placebo. The intervention occurred at a median time of 13 hours following admission to the ICU. There was no significant difference in duration of intravenous vasopressors or ICU length of stay and no adverse events reported.

RCTs have thus far included a heterogeneous population of critically ill patients. This may be problematic as the underlying mechanisms of hypotension in the ICU are varied and may include sepsis-driven cytokine release, adrenal insufficiency, medication or anaesthesia-related vasoplegia, hypovolaemia or inadequate cardiac output. Interestingly, a post hoc subgroup analysis in the MIDAS study found that the 31 patients with epidural analgesia had a significantly shorter duration of intravenous vasopressor therapy when administered midodrine compared with placebo (–18.4-hour difference; 95% CI, –33.5 to –3.3 hours; P = 0.045).101 This homogenous group of postoperative patients with neurogenic vasoplegia may be one such patient cohort to benefit from midodrine and should be studied further. Further evidence of this effect was seen in a recent single centre RCT in an Egyptian trauma ICU,104 which found the addition of midodrine halved the duration of intravenous vasopressor support in 30 patients with spinal cord injury and neurogenic shock (3.3 ± 1.32 days for adjunctive midodrine; 6.93 ± 2.32 days for intravenous vasopressor alone). However, this was an open label study and a lower MAP was achieved in the midodrine group, which may have affected the results.

Future directions and research priorities

Even though almost all prospective trials have failed to demonstrate clinical benefit thus far, interest remains in definitively establishing whether oral midodrine can wean ICU patients from intravenous vasopressor support more rapidly (Figure 2). The LIBERATE study116 is a Canadian multicentre, blinded RCT aiming to recruit 350 patients receiving stable intravenous vasopressor support to assess if midodrine 10 mg 8-hourly can shorten ICU length of stay.

Figure 2.

Figure 2

Research priorities for the use of midodrine in critically ill patients

ICU = intensive care unit; IV = intravenous; VP = vasopressor.

Trials investigating the "upstream" use of midodrine are currently lacking and would be of significant interest. A large retrospective, single-centre study of 1119 hypotensive patients who were administered midodrine in the ICU found that 41% were not receiving an intravenous vasopressor infusion at the time, and of these, 90% avoided the need for intravenous vasopressor after commencing midodrine.53 Prospective, randomised studies examining the role of midodrine before intravenous vasopressor infusions as either an alternative or adjunctive agent for patients in the emergency department, ward and intensive care settings would be of great value.

Finally, it is important to note that up to two-thirds of ICU patients who commence midodrine are discharged from the ICU on midodrine, and between one-third and half of all patients discharged from hospital remain on the medication.111, 117 Discharge from hospital on midodrine was associated with a 1.6-fold higher risk of one-year mortality.111 Weaning protocols were utilised in both the MIDAS and MAVERIC studies, and only 6.2% of patients in the MAVERIC study continued midodrine beyond the study period,54 suggesting such protocols may reduce the ongoing prescription of midodrine outside the ICU. Safety and compliance with midodrine weaning protocols merits further investigation.

Conclusions

This narrative review presents a comprehensive overview of midodrine use for the critical care physician coursing its early utilisation as a novel oral vasopressor for a range of outpatient indications through to its incremental use in ICUs around the world. Research interest has been piqued and will help shed light on the lingering questions around ideal patient selection, dosing, timing of initiation, and efficacy of midodrine for critically ill patients.

Competing interests

All authors declare that they do not have any potential conflict of interest in relation to this manuscript.

References

  • 1.K. Wismayr. AT 241435; Eidem, US Patent 3,340,298 (1965, 1967 both to Chemie Linz Ag).
  • 2.Pittner H., Stormann H., Enzenhofer R. Pharmacodynamic actions of midodrine, a new alpha-adrenergic stimulating agent, and its main metabolite, ST 1059. Arzneimittelforschung. 1976;26:2145–2154. [PubMed] [Google Scholar]
  • 3.Tsuchida K., Yamazaki R., Kaneko K., Aihara H. Effects of midodrine on blood flow in dog vascular beds. Arzneimittelforschung. 1986;36:1745–1748. [PubMed] [Google Scholar]
  • 4.Kolassa N., Schützenberger W.G., Wiener H. Krivanek P Plasma level of the prodrug midodrine and its active metabolite in comparison with the alpha-mimetic action in dogs. Arch Int Pharmacodyn Ther. 1979;238:96–104. [PubMed] [Google Scholar]
  • 5.Thulesius O., Gjöres J.E., Berlin E. Vasoconstrictor effect of midodrine, ST 1059, noradrenaline, etilefrine and dihydroergotamine on isolated human veins. Eur J Clin Pharmacol. 1979;16:423–424. doi: 10.1007/BF00568204. [DOI] [PubMed] [Google Scholar]
  • 6.Pittner H. Vasoconstrictor effects of midodrine, ST 1059, noradrenaline, etilefrine and norfenefrine on isolated dog femoral arteries and veins. Gen Pharmacol. 1983;14:107–109. doi: 10.1016/0306-3623(83)90075-7. [DOI] [PubMed] [Google Scholar]
  • 7.Jonas D. Treatment of female stress incontinence with midodrine: preliminary report. J Urol. 1977;118:980–982. doi: 10.1016/s0022-5347(17)58270-1. [DOI] [PubMed] [Google Scholar]
  • 8.Riccabona M. [The conservative treatment of stress incontinence in women with midodrine] [German] Wien Klin Wochenschr. 1981;93:163–165. [PubMed] [Google Scholar]
  • 9.Jonas D., Linzbach P., Weber W. The use of midodrin in the treatment of ejaculation disorders following retroperitoneal lymphadenectomy. Eur Urol. 1979;5:184–187. doi: 10.1159/000473102. [DOI] [PubMed] [Google Scholar]
  • 10.Riley A.J., Riley E.J. Partial ejaculatory incompetence: the therapeutic effect of midodrine, an orally active selective alpha-adrenoceptor agonist. Eur Urol. 1982;8:155–160. doi: 10.1159/000473504. [DOI] [PubMed] [Google Scholar]
  • 11.Schirger A., Sheps S.G., Thomas J.E., Fealey R.D., Midodrine A new agent in the management of idiopathic orthostatic hypotension and Shy–Drager syndrome. Mayo Clin Proc. 1981;56:429–433. [PubMed] [Google Scholar]
  • 12.Hebenstreit G. [Treatment of hypotension caused by psychopharmacological drugs] [German] Wien Med Wochenschr. 1981;131:109–112. [PubMed] [Google Scholar]
  • 13.Sazovsky H., Pittner H. [Diagnosis and therapy of hypotensive circulatory disorders in general practice. Experiences with Gutron using the Thulesius-diagram for diagnosis and supervision of therapy] [German] Fortschr Med. 1979;97:733–736. [PubMed] [Google Scholar]
  • 14.Hammerer I., Gassner I., Schwingshackl A. [The use of midodrin in the treatment of the orthostatic syndrome] [German] Padiatr Padol. 1981;16:59–68. [PubMed] [Google Scholar]
  • 15.Scholing W.E. [Studies on the effect of the alpha-receptor stimulant, gutron, in the orthostatic syndrome] [German] Wien Klin Wochenschr. 1981;93:429–434. [PubMed] [Google Scholar]
  • 16.Weippl G. [Infectious toxic hypotension — effect and dosage of midodrine] [German] Padiatr Padol. 1979;14:211–216. [PubMed] [Google Scholar]
  • 17.Jankovic J., Gilden J.L., Hiner B.C., et al. Neurogenic orthostatic hypotension: a double-blind, placebo-controlled study with midodrine. Am J Med. 1993;95:38–48. doi: 10.1016/0002-9343(93)90230-m. [DOI] [PubMed] [Google Scholar]
  • 18.Fouad-Tarazi F.M., Okabe M., Goren H. Alpha sympathomimetic treatment of autonomic insufficiency with orthostatic hypotension. Am J Med. 1995;99:604–610. doi: 10.1016/s0002-9343(99)80246-0. [DOI] [PubMed] [Google Scholar]
  • 19.Low P.A., Gilden J.L., Freeman R., et al. Efficacy of midodrine vs placebo in neurogenic orthostatic hypotension. A randomized, double-blind multicenter study. Midodrine Study Group. JAMA. 1997;277:1046–1051. [PubMed] [Google Scholar]
  • 20.Wright R.A., Kaufmann H.C., Perera R., et al. A double-blind, dose-response study of midodrine in neurogenic orthostatic hypotension. Neurology. 1998;51:120–124. doi: 10.1212/wnl.51.1.120. [DOI] [PubMed] [Google Scholar]
  • 21.Fang J.T., Huang C.C. Midodrine hydrochloride in patients on hemodialysis with chronic hypotension. Ren Fail. 1996;18:253–260. doi: 10.3109/08860229609052795. [DOI] [PubMed] [Google Scholar]
  • 22.Flynn J.J., Mitchell M.C., Caruso F.S., McElligott M.A. Midodrine treatment for patients with hemodialysis hypotension. Clin Nephrol. 1996;45:261–267. [PubMed] [Google Scholar]
  • 23.Lim P.S., Yang C.C., Li H.P., et al. Midodrine for the treatment of intradialytic hypotension. Nephron. 1997;77:279–283. doi: 10.1159/000190288. [DOI] [PubMed] [Google Scholar]
  • 24.Cruz D.N., Mahnensmith R.L., Brickel H.M., Perazella M.A. Midodrine is effective and safe therapy for intradialytic hypotension over 8 months of follow-up. Clin Nephrol. 1998;50:101–107. [PubMed] [Google Scholar]
  • 25.Angeli P., Volpin R., Piovan D., et al. Acute effects of the oral administration of midodrine, an alpha-adrenergic agonist, on renal hemodynamics and renal function in cirrhotic patients with ascites. Hepatology. 1998;28:937–943. doi: 10.1002/hep.510280407. [DOI] [PubMed] [Google Scholar]
  • 26.Angeli P., Volpin R., Gerunda G., et al. Reversal of type 1 hepatorenal syndrome with the administration of midodrine and octreotide. Hepatology. 1999;29:1690–1697. doi: 10.1002/hep.510290629. [DOI] [PubMed] [Google Scholar]
  • 27.Fairman K.A., Curtiss F.R. Regulatory actions on the off-label use of prescription drugs: ongoing controversy and contradiction in 2009 and 2010. J Manag Care Pharm. 2010;16:629–639. doi: 10.18553/jmcp.2010.16.8.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Somberg J.C. The midodrine withdrawal. Am J Ther. 2010;17:445. doi: 10.1097/MJT.0b013e3181f7e4ea. [DOI] [PubMed] [Google Scholar]
  • 29.Dhruva S.S., Redberg R.F. Accelerated approval and possible withdrawal of midodrine. JAMA. 2010;304:2172–2173. doi: 10.1001/jama.2010.1695. [DOI] [PubMed] [Google Scholar]
  • 30.Smith W., Wan H., Much D., et al. Clinical benefit of midodrine hydrochloride in symptomatic orthostatic hypotension: a phase 4, double-blind, placebo-controlled, randomized, tilt-table study. Clin Auton Res. 2016;26:269–277. doi: 10.1007/s10286-016-0363-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.McTavish D., Goa K.L., Midodrine A review of its pharmacological properties and therapeutic use in orthostatic hypotension and secondary hypotensive disorders. Drugs. 1989;38:757–777. doi: 10.2165/00003495-198938050-00004. [DOI] [PubMed] [Google Scholar]
  • 32.Yamazaki R., Tsuchida K., Aihara H. Effects of alpha-adrenoceptor agonists on cardiac output and blood pressure in spinally anesthetized ganglion-blocked dogs. Arch Int Pharmacodyn Ther. 1988;295:80–93. [PubMed] [Google Scholar]
  • 33.Lamarre-Cliche M., Souich P.D., Champlain J.D., Larochelle P. Pharmacokinetic and pharmacodynamic effects of midodrine on blood pressure, the autonomic nervous system, and plasma natriuretic peptides: a prospective, randomized, single-blind, two-period, crossover, placebo-controlled study. Clin Ther. 2008;30:1629–1638. doi: 10.1016/j.clinthera.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • 34.Zachariah P.K., Bloedow D.C., Moyer T.P., et al. Pharmacodynamics of midodrine, an antihypotensive agent. Clin Pharmacol Ther. 1986;39:586–591. doi: 10.1038/clpt.1986.101. [DOI] [PubMed] [Google Scholar]
  • 35.Dominiak P., Kees F., Welzel D., Grobecker H. Cardiovascular parameters and catecholamines in volunteers during passive orthostasis. Influence of antihypotensive drugs. Arzneimittelforschung. 1992;42:637–642. [PubMed] [Google Scholar]
  • 36.Iwase S., Mano T., Saito M., Ishida G. Long-acting alpha 1-adrenoceptive sympathomimetic agent suppresses sympathetic outflow to muscles in humans. J Auton Nerv Syst. 1991;36:193–199. doi: 10.1016/0165-1838(91)90043-3. [DOI] [PubMed] [Google Scholar]
  • 37.Iribarren C., Round A.D., Peng J.A., et al. Validation of a population-based method to assess drug-induced alterations in the QT interval: a self-controlled crossover study. Pharmacoepidemiol Drug Saf. 2013;22(1):222–232. doi: 10.1002/pds.3479. [DOI] [PubMed] [Google Scholar]
  • 38.Brändle J., Lageder H., Irsigler K. [Investigations of the effect of midodrine on carbohydrate and fat metabolism with particular reference to the diabetic metabolic state] [German] Wien Klin Wochenschr. 1977;89:164–167. [PubMed] [Google Scholar]
  • 39.Puchmayer V., Herdovà J., Krejcová H., Masopust J. Midodrine, a new therapeutic agent: recent experience. Int Angiol. 1993;12:113–118. [PubMed] [Google Scholar]
  • 40.Felsner P., Hofer D., Rinner I., et al. Continuous in vivo treatment with catecholamines suppresses in vitro reactivity of rat peripheral blood T-lymphocytes via -mediated mechanisms. J Neuroimmunol. 1992;37:47–57. doi: 10.1016/0165-5728(92)90154-d. [DOI] [PubMed] [Google Scholar]
  • 41.Glatter K.A., Tuteja D., Chiamvimonvat N., et al. Pregnancy in postural orthostatic tachycardia syndrome. Pacing Clin Electrophysiol. 2005;28:591–593. doi: 10.1111/j.1540-8159.2005.50026.x. [DOI] [PubMed] [Google Scholar]
  • 42.Al-Ghamdi B. Midodrine in pregnancy: a case report and literature review. Cardiol Pharmacol. 2015;4:144. [Google Scholar]
  • 43.Grobecker H., Kees F., Linden M., et al. [The bioavailability of midodrin and alpha-2,5-dimethoxyphenyl-beta-aminoethanol hydrochloride] [German] Arzneimittelforschung. 1987;37:447–450. [PubMed] [Google Scholar]
  • 44.Akimoto M., Iida I., Itoga H., et al. The in vitro metabolism of desglymidodrine, an active metabolite of prodrug midodrine by human liver microsomes. Eur J Drug Metab Pharmacokinet. 2004;29:179–186. doi: 10.1007/BF03190595. [DOI] [PubMed] [Google Scholar]
  • 45.Blowey D.L., Balfe J.W., Gupta I., et al. Midodrine efficacy and pharmacokinetics in a patient with recurrent intradialytic hypotension. Am J Kidney Dis. 1996;28:132–136. doi: 10.1016/s0272-6386(96)90142-0. [DOI] [PubMed] [Google Scholar]
  • 46.Pathak A., Raoul V., Montastruc J.L., Senard J.M. Adverse drug reactions related to drugs used in orthostatic hypotension: a prospective and systematic pharmacovigilance study in France. Eur J Clin Pharmacol. 2005;61:471–474. doi: 10.1007/s00228-005-0941-6. [DOI] [PubMed] [Google Scholar]
  • 47.Ramanath V.S., Andrus B.W., Szot C.R., et al. Takotsubo cardiomyopathy after midodrine therapy. Tex Heart Inst J. 2012;39:158–159. [PMC free article] [PubMed] [Google Scholar]
  • 48.Sandroni P., Benarroch E.E., Wijdicks E.F. Caudate hemorrhage as a possible complication of midodrine-induced supine hypertension. Mayo Clin Proc. 2001;76:1275. doi: 10.4065/76.12.1275. [DOI] [PubMed] [Google Scholar]
  • 49.Shankar Kikkeri N., Nagarajan E., Premkumar K., Nattanamai P. Reversible cerebral vasoconstriction syndrome due to midodrine in a patient with autonomic dysreflexia. Cureus. 2019;11 doi: 10.7759/cureus.4285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ye X., Ling B., Wu J., et al. Case report: severe myoclonus associated with oral midodrine treatment for hypotension. Medicine (Baltimore) 2020;99 doi: 10.1097/MD.0000000000021533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rubinstein S., Haimov M., Ross M.J. Midodrine-induced vascular ischemia in a hemodialysis patient: a case report and literature review. Ren Fail. 2008;30:808–812. doi: 10.1080/08860220802249025. [DOI] [PubMed] [Google Scholar]
  • 52.Pathak A., Debats P., Galinier M., et al. Intestinal obstruction associated with oral midodrine. Clin Auton Res. 2004;14:202–203. doi: 10.1007/s10286-004-0176-0. [DOI] [PubMed] [Google Scholar]
  • 53.Rizvi M.S., Trivedi V., Nasim F., et al. Trends in use of midodrine in the ICU: a single-center retrospective case series. Crit Care Med. 2018;46:e628–e633. doi: 10.1097/CCM.0000000000003121. [DOI] [PubMed] [Google Scholar]
  • 54.Costa-Pinto R., Yong Z.T., Yanase F., et al. A pilot, feasibility, randomised controlled trial of midodrine as adjunctive vasopressor for low-dose vasopressor-dependent hypotension in intensive care patients: the MAVERIC study. J Crit Care. 2022;67:166–171. doi: 10.1016/j.jcrc.2021.11.004. [DOI] [PubMed] [Google Scholar]
  • 55.Castrioto A., Tambasco N., Rossi A. Calabresi P Acute dystonia induced by the combination of midodrine and perphenazine. J Neurol. 2008;255:767–768. doi: 10.1007/s00415-008-0805-y. [DOI] [PubMed] [Google Scholar]
  • 56.Ali A., Farid S., Amin M., et al. Comparative clinical pharmacokinetics of midodrine and its active metabolite desglymidodrine in cirrhotic patients with tense ascites versus healthy volunteers. Clin Drug Investig. 2016;36:147–155. doi: 10.1007/s40261-015-0359-6. [DOI] [PubMed] [Google Scholar]
  • 57.Drambarean B., Bielnicka P., Alobaidi A. Midodrine treatment in a patient with treprostinil-induced hypotension receiving hemodialysis. Am J Health Syst Pharm. 2019;76:13–16. doi: 10.1093/ajhp/zxy001. [DOI] [PubMed] [Google Scholar]
  • 58.Whitson M.R., Mo E., Nabi T., et al. Feasibility, utility, and safety of midodrine during recovery phase from septic shock. Chest. 2016;149:1380–1383. doi: 10.1016/j.chest.2016.02.657. [DOI] [PubMed] [Google Scholar]
  • 59.Wong L.Y., Wong A., Robertson T., et al. Severe hypertension and bradycardia secondary to midodrine overdose. J Med Toxicol. 2017;13:88–90. doi: 10.1007/s13181-016-0574-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Perez-Lugones A., Schweikert R., Pavia S., et al. Usefulness of midodrine in patients with severely symptomatic neurocardiogenic syncope: a randomized control study. J Cardiovasc Electrophysiol. 2001;12:935–938. doi: 10.1046/j.1540-8167.2001.00935.x. [DOI] [PubMed] [Google Scholar]
  • 61.Barber D.B., Rogers S.J., Fredrickson M.D., Able A.C. Midodrine hydrochloride and the treatment of orthostatic hypotension in tetraplegia: two cases and a review of the literature. Spinal Cord. 2000;38:109–111. doi: 10.1038/sj.sc.3100959. [DOI] [PubMed] [Google Scholar]
  • 62.Sharma S., Bhambi B. Successful treatment of hypotension associated with stunned myocardium with oral midodrine therapy. J Cardiovasc Pharmacol Ther. 2005;10:77–79. doi: 10.1177/107424840501000109. [DOI] [PubMed] [Google Scholar]
  • 63.Naschitz J., Dreyfuss D., Yeshurun D., Rosner I. Midodrine treatment for chronic fatigue syndrome. Postgrad Med J. 2004;80:230–232. doi: 10.1136/pgmj.2003.011429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lafitte S., Peyrou J., Reynaud A., et al. Midodrine hydrochloride and unexpected improvement in hypertrophic cardiomyopathy symptoms. Arch Cardiovasc Dis. 2016;109:223–225. doi: 10.1016/j.acvd.2015.11.005. [DOI] [PubMed] [Google Scholar]
  • 65.Chidambaram M., Mink S., Sharma S. Atrial septal aneurysm with right-to-left interatrial shunting. Tex Heart Inst J. 2003;30:68–70. [PMC free article] [PubMed] [Google Scholar]
  • 66.Rajaram P., Spivey J., Fisher M. Novel role of midodrine in pulmonary hypertension in liver transplant candidates. Liver Transpl. 2016;22:1034–1036. doi: 10.1002/lt.24467. [DOI] [PubMed] [Google Scholar]
  • 67.Liou D.Z., Warren H., Maher D.P., et al. Midodrine: a novel therapeutic for refractory chylothorax. Chest. 2013;144:1055–1057. doi: 10.1378/chest.12-3081. [DOI] [PubMed] [Google Scholar]
  • 68.Sivakumar P., Ahmed L. Use of an alpha-1 adrenoreceptor agonist in the management of recurrent refractory idiopathic chylothorax. Chest. 2018;154:e1–e4. doi: 10.1016/j.chest.2018.02.005. [DOI] [PubMed] [Google Scholar]
  • 69.Hillis A.E., Ulatowski J.A., Barker P.B., et al. A pilot randomized trial of induced blood pressure elevation: effects on function and focal perfusion in acute and subacute stroke. Cerebrovasc Dis. 2003;16:236–246. doi: 10.1159/000071122. [DOI] [PubMed] [Google Scholar]
  • 70.Sharma S., Lardizabal J.A., Bhambi B. Oral midodrine is effective for the treatment of hypotension associated with carotid artery stenting. J Cardiovasc Pharmacol Ther. 2008;13:94–97. doi: 10.1177/1074248408317709. [DOI] [PubMed] [Google Scholar]
  • 71.Zakir R.M., Folefack A., Saric M., Berkowitz R.L. The use of midodrine in patients with advanced heart failure. Congest Heart Fail. 2009;15:108–111. doi: 10.1111/j.1751-7133.2008.00042.x. [DOI] [PubMed] [Google Scholar]
  • 72.Nieshoff E.C., Birk T.J., Birk C.A., et al. Double-blinded, placebocontrolled trial of midodrine for exercise performance enhancement in tetraplegia: a pilot study. J Spinal Cord Med. 2004;27:219–225. doi: 10.1080/10790268.2004.11753752. [DOI] [PubMed] [Google Scholar]
  • 73.Soler J.M., Previnaire J.G., Plante P., et al. Midodrine improves ejaculation in spinal cord injured men. J Urol. 2007;178:2082–2086. doi: 10.1016/j.juro.2007.07.047. [DOI] [PubMed] [Google Scholar]
  • 74.Soler J.M., Previnaire J.G., Plante P., et al. Midodrine improves orgasm in spinal cord-injured men: the effects of autonomic stimulation. J Sex Med. 2008;5:2935–2941. doi: 10.1111/j.1743-6109.2008.00844.x. [DOI] [PubMed] [Google Scholar]
  • 75.Bagheri Lankarani K., Sivandzadeh G.R., Zare M., et al. A preliminary report on the use of midodrine in treating refractory gastroesophageal disease: randomized double-blind controlled trial. Acta Biomed. 2020;91:70–78. doi: 10.23750/abm.v91i1.8486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Smits M., Lin S., Rahme J., et al. Blood pressure and early mobilization after total hip and knee replacements: a pilot study on the impact of midodrine hydrochloride. JB JS Open Access. 2019;4 doi: 10.2106/JBJS.OA.18.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Alhasso A., Glazener C.M., Pickard R., N'Dow J. Adrenergic drugs for urinary incontinence in adults. Cochrane Database Syst Rev. 2003:CD001842. doi: 10.1002/14651858.CD001842. (2) [DOI] [PubMed] [Google Scholar]
  • 78.Appenrodt B., Wolf A., Grünhage F., et al. Prevention of paracentesis-induced circulatory dysfunction: midodrine vs albumin. A randomized pilot study. Liver Int. 2008;28:1019–1025. doi: 10.1111/j.1478-3231.2008.01734.x. [DOI] [PubMed] [Google Scholar]
  • 79.Singh V., Dheerendra P.C., Singh B., et al. Midodrine versus albumin in the prevention of paracentesis-induced circulatory dysfunction in cirrhotics: a randomized pilot study. Am J Gastroenterol. 2008;103:1399–1405. doi: 10.1111/j.1572-0241.2008.01787.x. [DOI] [PubMed] [Google Scholar]
  • 80.Hamdy H., ElBaz A.A., Hassan A., Hassanin O. Comparison of midodrine and albumin in the prevention of paracentesis-induced circulatory dysfunction in cirrhotic patients: a randomized pilot study. J Clin Gastroenterol. 2014;48:184–188. doi: 10.1097/MCG.0b013e31829ae376. [DOI] [PubMed] [Google Scholar]
  • 81.Yosry A., Soliman Z.A., Eletreby R., et al. Oral midodrine is comparable to albumin infusion in cirrhotic patients with refractory ascites undergoing large-volume paracentesis: results of a pilot study. Eur J Gastroenterol Hepatol. 2019;31:345–351. doi: 10.1097/MEG.0000000000001277. [DOI] [PubMed] [Google Scholar]
  • 82.Singh V., Dhungana S.P., Singh B., et al. Midodrine in patients with cirrhosis and refractory or recurrent ascites: a randomized pilot study. J Hepatol. 2012;56:348–354. doi: 10.1016/j.jhep.2011.04.027. [DOI] [PubMed] [Google Scholar]
  • 83.Bari K., Minano C., Shea M., et al. The combination of octreotide and midodrine is not superior to albumin in preventing recurrence of ascites after large-volume paracentesis. Clin Gastroenterol Hepatol. 2012;10:1169–1175. doi: 10.1016/j.cgh.2012.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Parsaik A.K., Singh B., Altayar O., et al. Midodrine for orthostatic hypotension: a systematic review and meta-analysis of clinical trials. J Gen Intern Med. 2013;28:1496–1503. doi: 10.1007/s11606-013-2520-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Izcovich A., González Malla C., Manzotti M., et al. Midodrine for orthostatic hypotension and recurrent reflex syncope: A systematic review. Neurology. 2014;83:1170–1177. doi: 10.1212/WNL.0000000000000815. [DOI] [PubMed] [Google Scholar]
  • 86.Prakash S., Garg A.X., Heidenheim A.P., House A.A. Midodrine appears to be safe and effective for dialysis-induced hypotension: a systematic review. Nephrol Dial Transplant. 2004;19:2553–2558. doi: 10.1093/ndt/gfh420. [DOI] [PubMed] [Google Scholar]
  • 87.Nanda A., Reddy R., Safraz H., et al. Pharmacological therapies for hepatorenal syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 2018;52:360–367. doi: 10.1097/MCG.0000000000000913. [DOI] [PubMed] [Google Scholar]
  • 88.Brunelli S.M., Cohen D.E., Marlowe G., Van Wyck D. The impact of midodrine on outcomes in patients with intradialytic hypotension. Am J Nephrol. 2018;48:381–388. doi: 10.1159/000494806. [DOI] [PubMed] [Google Scholar]
  • 89.Alhamad T., Brennan D.C., Brifkani Z., et al. Pretransplant midodrine use: a newly identified risk marker for complications after kidney transplantation. Transplantation. 2016;100:1086–1093. doi: 10.1097/TP.0000000000001113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pottebaum A.A., Hagopian J.C., Brennan D.C., et al. Influence of pretransplant midodrine use on outcomes after kidney transplantation. Clin Transplant. 2018;32 doi: 10.1111/ctr.13366. [DOI] [PubMed] [Google Scholar]
  • 91.Arab J.P., Claro J.C., Arancibia J.P., et al. Therapeutic alternatives for the treatment of type 1 hepatorenal syndrome: a Delphi technique-based consensus. World J Hepatol. 2016;8:1075–1086. doi: 10.4254/wjh.v8.i25.1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Best L.M., Freeman S.C., Sutton A.J., et al. Treatment for hepatorenal syndrome in people with decompensated liver cirrhosis: a network meta-analysis. Cochrane Database Syst Rev. 2019:CD013103. doi: 10.1002/14651858.CD013103.pub2. (9) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.O'Donnell B., Synnott A. Midodrine, an alternative to intravenous vasopressor therapy after spinal surgery. Eur J Anaesthesiol. 2002;19:841–842. doi: 10.1017/s0265021502251352. [DOI] [PubMed] [Google Scholar]
  • 94.Gutman L.B., Wilson B.J. The role of midodrine for hypotension outside of the intensive care unit. J Popul Ther Clin Pharmacol. 2017;24:e45–e50. doi: 10.22374/1710-6222.24.3.4. [DOI] [PubMed] [Google Scholar]
  • 95.Gonzalez-Cordero A., Ortiz-Troche S., Nieves-Rivera J., et al. Midodrine in end-stage heart failure. BMJ Support Palliat Care. 2020 doi: 10.1136/bmjspcare-2020-002369. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 96.Anstey M.H., Wibrow B., Thevathasan T., et al. Midodrine as adjunctive support for treatment of refractory hypotension in the intensive care unit: a multicenter, randomized, placebo controlled trial (the MIDAS trial) BMC Anesthesiol. 2017;17:47. doi: 10.1186/s12871-017-0339-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Poveromo L.B., Michalets E.L., Sutherland S.E. Midodrine for the weaning of vasopressor infusions. J Clin Pharm Ther. 2016;41:260–265. doi: 10.1111/jcpt.12375. [DOI] [PubMed] [Google Scholar]
  • 98.Tremblay J.A., Laramée P., Lamarche Y., et al. Potential risks in using midodrine for persistent hypotension after cardiac surgery: a comparative cohort study. Ann Intensive Care. 2020;10:121. doi: 10.1186/s13613-020-00737-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Macielak S.A., Vollmer N.J., Haddad N.A., et al. Hemodynamic effects of an increased midodrine dosing frequency. Crit Care Explor. 2021;3 doi: 10.1097/CCE.0000000000000405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Levine A.R., Meyer M.J., Bittner E.A., et al. Oral midodrine treatment accelerates the liberation of intensive care unit patients from intravenous vasopressor infusions. J Crit Care. 2013;28:756–762. doi: 10.1016/j.jcrc.2013.05.021. [DOI] [PubMed] [Google Scholar]
  • 101.Santer P., Anstey M.H., Patrocinio M.D., et al. Effect of midodrine versus placebo on time to vasopressor discontinuation in patients with persistent hypotension in the intensive care unit (MIDAS): an international randomised clinical trial. Intensive Care Med. 2020;46:1884–1893. doi: 10.1007/s00134-020-06216-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lal A., Trivedi V., Rizvi M.S., et al. Oral midodrine administration during the first 24 hours of sepsis to reduce the need of vasoactive agents: placebo-controlled feasibility clinical trial. Crit Care Explor. 2021;3 doi: 10.1097/CCE.0000000000000382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Adly D.H.E., Bazan N.S., El Borolossy R.M., et al. Midodrine improves clinical and economic outcomes in patients with septic shock: a randomized controlled clinical trial. Ir J Med Sci. 2022 doi: 10.1007/s11845-021-02903-w. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 104.Ahmed Ali A.T., Abd El-Aziz M.A., Mohamed Abdelhafez A., Ahmed Thabet A.M. Effect of oral vasopressors used for liberation from intravenous vasopressors in intensive care unit patients recovering from spinal shock: a randomized controlled trial. Crit Care Res Pract. 2022:6448504. doi: 10.1155/2022/6448504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Liu M., Luka B., Kolli R., et al. Use of oral midodrine in weaning off intravenous vasopressors in patients with septic shock. J Pharm Pract. 2010;23:284. [Google Scholar]
  • 106.Roach E., Adie S., Gowan M., et al. 200: impact of oral midodrine on duration of intravenous vasopressor therapy. Crit Care Med. 2018;46:82. [Google Scholar]
  • 107.Hammond D.A., Smith M.N., Peksa G.D., et al. Midodrine as an adjuvant to intravenous vasopressor agents in adults with resolving shock: systematic review and meta-analysis. J Intensive Care Med. 2020;35:1209–1215. doi: 10.1177/0885066619843279. [DOI] [PubMed] [Google Scholar]
  • 108.Duschek S., Heiss H., Werner N., Reyes del Paso G.A. Modulations of autonomic cardiovascular control following acute alpha-adrenergic treatment in chronic hypotension. Hypertens Res. 2009;32:938–943. doi: 10.1038/hr.2009.115. [DOI] [PubMed] [Google Scholar]
  • 109.Duschek S., Heiss H., Buechner B., et al. Hemodynamic determinants of chronic hypotension and their modification through vasopressor application. J Physiol Sci. 2009;59:105–112. doi: 10.1007/s12576-008-0015-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Pichot C., Geloen A., Ghignone M., Quintin L. Alpha-2 agonists to reduce vasopressor requirements in septic shock? Med Hypotheses. 2010;75:652–656. doi: 10.1016/j.mehy.2010.08.010. [DOI] [PubMed] [Google Scholar]
  • 111.Rizvi M.S., Nei A.M., Gajic O., et al. Continuation of newly initiated midodrine therapy after intensive care and hospital discharge: a single-center retrospective study. Crit Care Med. 2019;47:e648–e653. doi: 10.1097/CCM.0000000000003814. [DOI] [PubMed] [Google Scholar]
  • 112.Wecht J.M., Rosado-Rivera D., Handrakis J.P., et al. Effects of midodrine hydrochloride on blood pressure and cerebral blood flow during orthostasis in persons with chronic tetraplegia. Arch Phys Med Rehabil. 2010;91:1429–1435. doi: 10.1016/j.apmr.2010.06.017. [DOI] [PubMed] [Google Scholar]
  • 113.Phillips A.A., Krassioukov A.V., Ainslie P.N., et al. Increased central arterial stiffness explains baroreflex dysfunction in spinal cord injury. J Neurotrauma. 2014;31:1122–1128. doi: 10.1089/neu.2013.3280. [DOI] [PubMed] [Google Scholar]
  • 114.Riker R.R., Gagnon D.J. Letter to the Editor: "Midodrine to liberate ICU patients from intravenous vasopressors: another negative fixed-dose trial". J Crit Care. 2022;69 doi: 10.1016/j.jcrc.2022.153995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Costa-Pinto R., Bellomo R. Randomised-control trials do not support midodrine as an intravenous vasopressor weaning strategy. J Crit Care. 2022;153996 doi: 10.1016/j.jcrc.2022.153996. [DOI] [PubMed] [Google Scholar]
  • 116.Opgenorth D., Baig N., Fiest K., et al. LIBERATE: a study protocol for midodrine for the early liberation from vasopressor support in the intensive care unit (LIBERATE): protocol for a randomized controlled trial. Trials. 2022;23:194. doi: 10.1186/s13063-022-06115-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Cardenas-Garcia J.L., Withson M., Healy L., et al. Safety of oral midodrine as a method of weaning from intravenous vasoactive medication in the medical intensive care unit. Chest. 2014;146:224A. [Google Scholar]

Articles from Critical Care and Resuscitation are provided here courtesy of Elsevier

RESOURCES