Fact-checked by Grok 2 weeks ago

Droxidopa

Droxidopa, also known by the brand name Northera, is an orally administered synthetic precursor to norepinephrine used to treat symptomatic neurogenic (nOH), a condition involving a sudden drop in upon standing that causes , , or fainting in adults with underlying disorders. It functions as a that is decarboxylated to norepinephrine, which then stimulates alpha- and beta-adrenergic receptors to constrict blood vessels and elevate without crossing the , thereby addressing peripheral hypotension while minimizing effects. Approved by the U.S. (FDA) on February 18, 2014, for the treatment of associated with primary autonomic failure—such as , , and —as well as deficiency, or non-diabetic , droxidopa represents the first new chemical entity approved for this indication in decades. It is available in capsule form (100 mg, 200 mg, and 300 mg) and is typically initiated at 100 mg three times daily, titrated up to a maximum of 600 mg three times daily based on symptom response and monitoring to avoid when lying down. Originally developed and approved in in for use in and other conditions like , it received designation in the U.S. prior to approval and has since been studied in clinical trials demonstrating improvements in orthostatic symptoms and daily activities; generic versions were approved by the FDA in 2021. Common side effects include , , , and , with a for requiring regular checks; use with nonselective inhibitors should be avoided due to the risk of . Pharmacologically, droxidopa exhibits high oral (approximately 90%), a of 2 to 3 hours, and is primarily excreted unchanged in the after decarboxylation by . No clinically significant has been reported, and it is not recommended for pediatric use due to lack of studies.

Clinical Use

Indications

Droxidopa is primarily approved by the U.S. Food and Drug Administration (FDA) for the treatment of orthostatic dizziness, lightheadedness, or the sensation of impending syncope in adult patients with symptomatic neurogenic orthostatic hypotension (nOH) caused by primary autonomic failure (including Parkinson's disease, multiple system atrophy, and pure autonomic failure), dopamine beta-hydroxylase deficiency, or non-diabetic autonomic neuropathy. This approval targets symptoms arising from underlying neurodegenerative conditions that impair autonomic nervous system function, leading to inadequate blood pressure regulation upon standing. In , droxidopa has been approved since 1989 for additional indications, including and dizziness on standing associated with , , and , as well as intradialytic hypotension in patients undergoing . It is also indicated there for freezing of gait in . By acting as a precursor to norepinephrine, droxidopa alleviates symptoms through an increase in standing systolic , with a mean change from of 11.5 mmHg compared to 4.8 mmHg with , which helps mitigate , , and fall risk in affected individuals. It is intended for adults with stemming from neurodegenerative or autonomic disorders and is not indicated for or of non-neurogenic origin.

Dosage and Administration

Droxidopa is administered orally in capsule form, with a recommended starting dose of 100 mg three times daily, taken upon arising in the morning, at midday, and in the late afternoon at least 3 hours prior to bedtime to minimize the risk of supine hypertension during sleep. Capsules should be swallowed whole and may be taken consistently with or without food. Titration begins at the starting dose and proceeds in increments of 100 mg three times daily every 24 to 48 hours, guided by the patient's symptomatic response, up to a maximum dose of 600 mg three times daily (total daily dose of 1,800 mg). must be monitored prior to initiating and following each dose increase to assess for and ensure safety. If a dose is missed, the next scheduled dose should be taken without doubling up. Special considerations include elevating the head of the bed while resting or sleeping to reduce hypertension risk, and patients should be educated on recognizing symptoms such as , , or associated with elevated . Home monitoring is recommended during initial and dose escalation to track hypertension. The last daily dose should not be taken later than early evening, such as 5 p.m., assuming typical hours, to further mitigate nighttime hypertension. Effectiveness beyond 2 weeks of treatment has not been established, and periodic reassessment is advised; if no symptomatic improvement occurs after reaching the maximum tolerated dose, discontinuation may be considered following clinical evaluation.

Safety Profile

Side Effects

In placebo-controlled clinical trials involving a total of 485 patients, adverse event rates were similar between droxidopa and placebo. Common side effects include (up to 13%), (up to 10%), (up to 9%), (up to 7%), and (up to 7%). Supine hypertension is a notable risk associated with droxidopa, occurring in up to 8% of patients (SBP >180 mmHg) in clinical trials, and if unmanaged, it can lead to serious cardiovascular events such as stroke. In a long-term open-label study, common adverse events included falls (24%), urinary tract infections (15%), headache (13%), syncope (13%), and dizziness (10%). Additional adverse effects observed at lower frequencies in short-term trials include urinary tract infections (up to 6%), nasopharyngitis (up to 5%), and syncope (up to 3%), which are typically mild and transient in nature. Postmarketing reports include cardiac disorders (e.g., ), nervous system disorders (e.g., cerebrovascular accident), and psychiatric disorders (e.g., ). Management of side effects involves dose reduction or adjustment for , with discontinuation recommended if severe reactions occur; no data on long-term carcinogenicity are available from clinical studies.

Contraindications and Precautions

Droxidopa is contraindicated in patients with a known history of to the drug or any of its components, as this may lead to serious allergic reactions including . Use with caution in patients with uncontrolled , as the medication's conversion to norepinephrine may exacerbate supine . No specific guidance exists for due to lack of data.

Drug Interactions

Coadministration of droxidopa with sympathomimetic agents, such as or norepinephrine, may result in additive pressor effects, leading to excessive increases in ; caution and close monitoring are advised. When used concurrently with levodopa or other catecholamine precursors, droxidopa may potentiate norepinephrine levels, necessitating monitoring to avoid .

Precautions

Patients treated with droxidopa require regular monitoring for supine hypertension, particularly the elderly or those with renal impairment, as this can elevate the risk of or other cardiovascular complications; strategies include elevating the head of the bed and dose reduction if needed. Caution is warranted in individuals with hepatic impairment, where dose adjustment may be necessary due to limited data and potential for altered metabolism; no specific guidelines exist for severe cases. Droxidopa has not been studied in pediatric patients and is not approved for use in this population. Limited data are available on use in pregnant women; animal studies showed embryofetal at doses similar to , and it should be used only if the potential benefit justifies the risk to the . is not recommended during droxidopa due to the absence of data on its presence in and potential adverse effects on the infant. Abrupt discontinuation may lead to worsening or rebound symptoms, so tapering is advised under medical supervision.

Overdose Management

Overdose with droxidopa primarily manifests as severe , with possible non-specific symptoms such as or ; there is no specific , and management involves supportive care, including monitoring and symptomatic treatment.

Pharmacology

Pharmacodynamics

Droxidopa is a synthetic precursor that functions as a to norepinephrine, exerting its effects through enzymatic conversion rather than direct receptor interaction. It is decarboxylated by (AADC), an enzyme widely distributed in neural and nonneural tissues, to form L-norepinephrine, which mediates the drug's sympathomimetic actions. This conversion bypasses the (DBH) step in norepinephrine biosynthesis, which is often deficient in neurogenic (), thereby restoring norepinephrine availability despite impaired endogenous synthesis. The resulting increase in peripheral norepinephrine levels enhances of arteries and veins, leading to elevated without direct of adrenergic receptors by droxidopa itself. Preclinical studies suggest that droxidopa can cross the blood-brain barrier due to its structural similarity to , allowing potential central metabolism to norepinephrine. However, the primary therapeutic effects are mediated by peripheral norepinephrine, which enhances and elevates . This peripheral augmentation of norepinephrine contributes to the drug's therapeutic profile in conditions involving sympathetic insufficiency. Droxidopa's specificity as a norepinephrine precursor is attributed to its L-threo stereoisomer, the only biologically active form that undergoes efficient to L-norepinephrine; the other stereoisomers, including the D-form, do not contribute significantly to this pathway and may even inhibit it. Consequently, there is no substantial accumulation of or other intermediates, as the molecule is designed to directly yield norepinephrine without relying on the -to-norepinephrine conversion. The pharmacodynamic effects peak within 1 to 4 hours after dosing, with plasma norepinephrine levels rising transiently and sustaining improvements in standing for approximately 4 to 6 hours.

Pharmacokinetics

Droxidopa is rapidly absorbed after , achieving peak concentrations (Cmax) within 1 to 4 hours, with a time to peak (Tmax) of approximately 2 hours in healthy volunteers. Its is approximately 90%, and is nearly dose-proportional over the clinical dose of 100 to 600 . Although a high-fat moderately reduces Cmax by about 35% and the area under the concentration-time curve () by 20% while delaying Tmax by roughly 2 hours, no dosage adjustment is recommended with . The apparent of droxidopa is approximately 200 L, corresponding to roughly 2-3 L/kg in adults. It exhibits concentration-dependent , with 75% binding at plasma concentrations of 100 ng/mL and decreasing to 26% at 10,000 ng/mL. Preclinical and indicate that droxidopa crosses the blood-brain barrier, facilitated by its structural similarity to large neutral , which enables transport via amino acid carriers such as the L-type transporter. Metabolism of droxidopa occurs primarily via the catecholamine pathway and is not mediated by enzymes. The main biotransformation involves by (AADC) to norepinephrine, predominantly within noradrenergic neurons. A secondary pathway includes O-methylation by (COMT) to form 3-O-methyldroxidopa (3-OM-DOPS), a inactive . Other routes produce protocatechualdehyde via DOPS aldolase and subsequent oxidation products. The terminal elimination of droxidopa is approximately 2.5 hours, with total clearance around 400 mL/hour after a 300 dose. Pharmacological effects may extend beyond this due to the persistence of the norepinephrine. Elimination is predominantly renal, with about 75% of the administered dose recovered in within 24 hours, mainly as metabolites; fecal is minimal. No significant accumulation occurs with repeated dosing every 4 hours. Droxidopa has no clinically relevant involvement with enzymes, resulting in low potential for metabolic drug-drug interactions. Population pharmacokinetic analyses show that mild hepatic impairment does not significantly alter exposure, as parameters such as , , , and levels do not influence droxidopa ; no dosage adjustment is needed. For renal function, no adjustments are required in mild to moderate impairment ( >30 mL/min), though data are limited in severe impairment ( <30 mL/min) and caution is advised. Age, sex, and also have no meaningful effects on .

Chemistry

Chemical Structure

Droxidopa, chemically known as (2S,3R)-2-amino-3-(3,4-dihydroxyphenyl)-3-hydroxypropanoic acid, is the L-threo of 3,4-dihydroxyphenylserine. Its molecular formula is C₉H₁₁NO₅, with a molecular weight of 213.19 g/mol. The molecule features a catecholamine backbone, characterized by a 3,4-dihydroxyphenyl ring attached to a serine-like chain, making it a β-hydroxy analog of with an additional hydroxyl group at the β-position and a threo configuration at the α- and β-carbons. This structure positions droxidopa as a synthetic precursor to norepinephrine, sharing similarities with natural catecholamine pathways. Regarding stereochemistry, the L-threo (2S,3R) exhibits pharmacological activity as a , while the other stereoisomers—L-erythro, D-threo, and D-erythro—are significantly less potent or inactive in increasing noradrenergic activity due to poorer enzymatic conversion. Physically, droxidopa appears as a white to off-white, odorless powder. It is slightly soluble in (approximately 15 mg/mL), with solubility increasing in acidic conditions such as dilute , and it is practically insoluble in common organic solvents like , , acetone, and . The compound demonstrates stability under acidic environments, supporting its formulation and storage requirements.

Synthesis and Properties

Droxidopa, chemically known as L-threo-3-(3,4-dihydroxyphenyl)serine, is synthesized through several established routes that emphasize to obtain the active (2S,3R) . One common approach involves the enzymatic of racemic dihydroxyphenylserine (DOPS) using L-amino acylase to selectively hydrolyze the L-, followed by isolation and deprotection. Alternatively, proceeds via the aldol-type condensation of protected serine or with 3,4-dihydroxybenzaldehyde, yielding a β-hydroxy intermediate that undergoes stereoselective reduction, often employing chiral catalysts like complexes or enzymatic systems to favor the threo . These methods ensure high enantiomeric excess, typically exceeding 99%, while minimizing diastereomeric impurities. In the initial development of droxidopa, enantiopure production leveraged microbial enzymes, such as thermostabilized L-threonine aldolase derived from coelicolor, to catalyze the direct condensation of and 3,4-dihydroxybenzaldehyde, achieving yields up to 2.0 mg/mL over extended batch reactions. Modern manufacturing processes further refine this by incorporating two-step enantioselective sequences, including enzyme-catalyzed aldol reactions followed by silicon-mediated amidation, reducing steps from classical resolutions and improving overall efficiency. Physicochemical properties of droxidopa include a of 232–235°C (with ), reflecting its thermal stability as a crystalline solid. It exhibits pKa values of approximately 2.0 for the group and 8.7–9.7 for the phenolic hydroxyl groups, influencing its and profile; the compound is slightly soluble in (about 15 mg/mL) and hydrophilic with a logP of -1.7 to -2.4. As an odorless, white to off-white crystalline powder, droxidopa is prone to oxidation due to its moiety, necessitating protective measures during handling and storage. For formulation, droxidopa is available in 100 mg, 200 mg, and 300 mg oral capsules containing , , and as excipients, with capsule shells incorporating and colorants for identification. Stability is maintained by storing at controlled (20–25°C, excursions to 15–30°C permitted), often in light-protected containers to prevent oxidative , as to or air can lead to discoloration and potency loss. Manufacturing specifications limit diastereomeric impurities to less than 0.5% and ensure chiral purity greater than 99% enantiomeric excess, verified through HPLC analysis to meet pharmaceutical standards.

History and Development

Early Development

Droxidopa (L-threo-dihydroxyphenylserine; L-threo-DOPS), first synthesized in , was investigated by researchers in the early as a potential precursor to norepinephrine for the of autonomic disorders. Initial investigations during this period, however, yielded negative results, concluding that threo-DOPS exhibited low norepinephrine-increasing activity in the brain and displayed weak pharmacological effects, leading to doubts about its therapeutic potential as a norepinephrine precursor. The preclinical rationale for droxidopa centered on its ability to serve as a that could be decarboxylated by L-aromatic decarboxylase to produce natural L-norepinephrine, thereby addressing deficiencies in activity seen in conditions such as and models of neurogenic . In the latter half of the , renewed efforts by groups shifted focus to the specific enantiomers; and colleagues at established that L-threo-DOPS was the effective L-norepinephrine precursor among the four DOPS stereoisomers, demonstrating key pharmacological properties including slow-onset, long-lasting pressor effects and inhibition of harmaline-induced . Animal studies in the early 1970s, involving Sumitomo Pharmaceuticals, revealed that droxidopa elevated in models using rats and dogs, without inducing cardiac toxicity, supporting its role in restoring norepinephrine levels in deficient states. Additional preclinical work by groups at (Hayashi and Suzuki) and Juntendo University (Narabayashi) confirmed benefits in norepinephrine-deficient animal models mimicking and advanced , including improvements in orthostatic symptoms and freezing-like behaviors. Intellectual property for droxidopa's synthesis and use in treating was secured through filed in the , including Japanese unexamined applications that informed later filings by Sumitomo Pharmaceuticals. During its , the compound was referred to as L-threo-DOPS or by the SM-5688.

Regulatory Approvals and Clinical Trials

Droxidopa received its initial regulatory approval in in 1989 from the Ministry of Health, Labour and Welfare for the treatment of , syncope, and upon standing in patients with , (Shy-Drager syndrome), and . The approval was supported by registration studies involving 1,255 subjects and post-marketing surveillance of 1,856 patients, which demonstrated improvements in and symptoms associated with these conditions. In 2000, the indication was expanded to include alleviation of vertigo, staggering, on standing, lassitude, and weakness in hemodialysis patients experiencing , with typical maintenance dosing of 300–600 mg daily (up to 900 mg maximum). In the United States, the (FDA) approved droxidopa (marketed as Northera) on February 18, 2014, under the accelerated approval pathway for the treatment of symptomatic neurogenic in adults with primary autonomic failure (including and ), deficiency, or non-diabetic . In February 2021, the FDA approved the first generic version of droxidopa. This approval was based on three 3 clinical trials evaluating symptom improvement and hemodynamic effects. Pivotal evidence came from Study 301, a multicenter, double-blind, -controlled trial with 162 patients randomized after an open-label ; droxidopa significantly improved the primary of the Questionnaire (OHQ) composite score by 0.90 units compared to (p=0.003), with the symptom subscore improving by 0.73 units (p=0.010) and standing systolic increasing by approximately 7 mmHg (p<0.001). Supporting data from Studies 302 and 303, including a randomized design in Study 302, further confirmed short-term symptom relief, though some endpoints showed variability across sites. A 2018 Bayesian of six randomized trials (four for droxidopa and two for , involving 783 patients) indicated similar between droxidopa and in increasing standing systolic for neurogenic , with droxidopa yielding a mean increase of 6.2 mmHg (95% : 2.4–10 mmHg) versus 17 mmHg for . However, droxidopa demonstrated better tolerability, as it did not significantly elevate the risk of supine hypertension (risk ratio 1.4, 95% : 0.7–2.7) compared to (risk ratio 5.1, 95% : 1.6–24). Long-term extension studies, such as an with mean exposure of 363 days (range: 2–1,133 days, up to approximately 3 years), confirmed the durability of these benefits, with only 7% of patients discontinuing due to lack of and sustained improvements in symptoms and daily activities. Droxidopa has not received marketing authorization in the ; it held orphan designation for in since 2007 but was withdrawn from the Union Register in January 2022 at the sponsor's request, with no approval granted. In post-marketing , the FDA updated the Northera on September 18, 2023, maintaining that and effectiveness in pediatric patients have not been established while noting no major issues leading to product withdrawals.

Society and Culture

Names and Availability

Droxidopa is the generic name and (INN) for the compound L-threo-dihydroxyphenylserine. In the United States, it is marketed under the brand name Northera by . In Japan, the brand name is Dops, produced by . Droxidopa is formulated as oral capsules in strengths of 100 mg, 200 mg, and 300 mg. In the United States, Northera received FDA approval in 2014, with generic versions entering the market in 2021 after the expiration of market exclusivity on February 18, 2021. Internationally, droxidopa has been widely available in since its approval there in 1989 for indications including . In , it received designation in 2007 for but the status was withdrawn in 2022, leaving it with limited availability and primarily investigational use. The drug is manufactured by and licensed partners, such as for Northera in select markets, with additional production by generic manufacturers like Lupin and Hikma following U.S. exclusivity expiration. Droxidopa is available exclusively by prescription and has no over-the-counter status worldwide. Droxidopa is classified as a prescription-only medication in the United States, with no applicable scheduling under the , as it does not possess significant abuse potential. It received designation from the FDA on January 17, 2007, for the treatment of neurogenic (nOH), a rare condition affecting fewer than 200,000 patients annually, which upon approval in 2014 granted the sponsor seven years of market exclusivity to encourage development for indications. This exclusivity period expired on February 18, 2021, allowing generic entry and subsequent FDA approvals for generic droxidopa capsules in 2021. The patent landscape for droxidopa has transitioned following the loss of exclusivity, with multiple generic manufacturers, including Lupin and Hikma, receiving FDA approvals for equivalent formulations since early 2021, reflecting the expiration of key intellectual property protections. While formulation-specific patents have been referenced in development processes, no major ongoing litigation regarding core droxidopa patents was identified in recent regulatory filings, facilitating broader market competition. Prior to widespread generic availability, droxidopa treatment in the carried a substantial annual , estimated at $50,000 to $60,000 per based on wholesale . As of 2025, with the launch of low- generics such as CivicaScript's (offered at $132 wholesale per 90-capsule of 300 mg in April 2025), annual costs have decreased significantly to approximately $2,000–$10,000 depending on dosage and , though actual out-of-pocket expenses vary with and assistance programs. The for droxidopa reached approximately $670 million in 2024, driven by increasing demand amid an aging prone to , and is projected to expand to $1.2 billion by 2033 at a of 7.2%, supported by enhanced awareness and diagnostic rates. Access to droxidopa in the is facilitated through coverage under , with 86% of plans including the drug on their formularies as of 2025, often subject to for nOH indications. , the current sponsor of the branded product Northera, offers patient assistance programs including a commercial copay assistance initiative that can reduce costs to as low as $0 for eligible commercially insured s, alongside broader support for uninsured or underinsured individuals meeting income criteria. Supply shortages have been rare, though isolated reports of intermittent availability occurred in 2022 due to manufacturing adjustments by generic suppliers. Globally, droxidopa's economics differ markedly by region; in , where it has been approved since 1989 under the brand name DOPS, annual treatment costs have historically been substantially lower (around $5,000 per patient as reported in 2007), reflecting long-term market maturity and generic availability. In the , droxidopa lacks marketing authorization, with its orphan medicinal product designation withdrawn in January 2022 at the sponsor's request, resulting in no availability and limited access outside contexts.

Research

Ongoing Investigations

Recent clinical investigations into droxidopa continue to explore its applications in () and related , particularly in rare diseases. A notable , NCT04977388, initiated in 2021 and completed in 2025, evaluated the safety and efficacy of droxidopa in adult survivors of experiencing , focusing on orthostatic symptom management in this rare . Results pending publication as of November 2025. This phase 2 study addresses a critical gap in treatments for long-term complications of , where autonomic dysfunction persists despite early interventions. In critical care settings, droxidopa has shown promise for facilitating vasopressor weaning in (ICU) patients with persistent . A multicenter pilot study involving 30 patients reported a time to intravenous vasopressor discontinuation of 70 hours after droxidopa initiation, with norepinephrine equivalents significantly decreasing (from 0.08 to 0.02 mcg/kg/min, p < 0.001), achieving approximately 70% success in weaning within 72 hours for those with lower baseline requirements. These findings suggest droxidopa as a safe oral adjunct, even via feeding tubes, for transitioning patients from intravenous support without major adverse events. Emerging research highlights droxidopa's potential in Parkinson's disease beyond standard nOH management. A small 2022 trial (with follow-up analyses in 2024 literature) involving 9 patients with Parkinson's and orthostatic hypotension demonstrated a 25% reduction in postural sway after droxidopa treatment, improving balance and gait stability during orthostatic challenges. Additionally, 2025 market analyses project sustained growth in droxidopa use for nOH associated with multiple system atrophy (MSA), driven by increasing diagnosis rates and limited alternatives, with the neurogenic orthostatic hypotension market expected to expand at a compound annual growth rate (CAGR) of around 7.2% through 2032. Exploratory efforts have examined droxidopa in other conditions, including attention-deficit/hyperactivity disorder (ADHD), though progress remains preliminary. Phase 2 data from earlier trials indicated potential improvements in attention and prefrontal activity. For acute , a 2024 case series described successful oral droxidopa use in weaning vasopressors for refractory cases, such as post-spinal cord injury or dialysis-related instability, highlighting its role in bridging to enteral therapy. Despite these advances, challenges persist in droxidopa research, particularly recruitment difficulties in rare diseases like and Menkes, where patient heterogeneity and low prolong trial timelines. As of 2025, the FDA has not granted fast-track designation for new indications beyond approved uses. Looking ahead, future applications may integrate droxidopa with digital monitoring technologies for real-time orthostatic assessment and personalized dosing, supporting a projected 11% CAGR in the broader market through 2035 amid rising autonomic disorder .

Discontinued or Exploratory Uses

Development for droxidopa in (ADHD) was explored in the 2010s through phase 2 clinical trials, including a pilot study combining droxidopa with carbidopa that reported improvements in ADHD symptoms during open-label treatment but required further validation in double-blind phases. However, these efforts were halted due to insufficient efficacy in demonstrating consistent benefits beyond initial observations, with no progression to phase 3 trials. Similarly, investigations into droxidopa for chronic fatigue syndrome involved a phase 2 open-label study that was terminated early, likely owing to challenges in establishing clear symptomatic relief amid the complex etiology of the condition. In fibromyalgia, a phase 2 randomized trial assessed droxidopa alone and with carbidopa, yielding a mean change in Fibromyalgia Impact Questionnaire scores of -9.72 points for droxidopa monotherapy compared to -4.74 points for placebo; these differences did not achieve statistical significance, leading to discontinuation of development around 2011 without advancement. Trials for this indication were not revived by 2022, as droxidopa failed to differentiate from placebo in reducing pain or fatigue, highlighting limitations in its norepinephrine-enhancing mechanism for central sensitization disorders. Exploratory applications in the included intradialytic among patients, where a phase 2 placebo-controlled study of 85 participants showed droxidopa improved nadir systolic by 12 mmHg versus 2 mmHg for , suggesting potential to mitigate dialysis-related complications. Despite this, U.S. development was abandoned post-2014 approval for neurogenic , shifting focus to the primary indication, though it remains approved for this use in . Limited observational data have also examined droxidopa in variants of , but these remain unapproved and exploratory, with no controlled trials establishing efficacy beyond standard neurogenic management. Discontinuation across these indications often stemmed from droxidopa's side effect profile—including supine hypertension, headache, and fatigue—outweighing marginal benefits, particularly when alternatives like midodrine offered comparable vasoconstrictive effects with established use in orthostatic contexts. A 2018 integrated analysis underscored limited evidence for droxidopa's utility outside neurogenic orthostatic hypotension, reinforcing the absence of revival for non-approved uses as of 2025. These experiences highlighted the importance of patient stratification in autonomic disorder trials to identify responders, informing more targeted future investigations.