Aviptadil is a synthetic analog of vasoactive intestinal peptide (VIP), a 28-amino acid endogenous neuropeptidehormone that exhibits bronchodilatory, vasodilatory, anti-inflammatory, and immunomodulatory properties, primarily targeting receptors in the lungs to mitigate cytokine storms and pulmonary injury.[1] Developed as an injectable or inhalable formulation, it binds selectively to VPAC1 and VPAC2 receptors on alveolar type II cells, inhibiting pro-inflammatory cytokines such as IL-6 and TNF-α, preventing apoptosis, enhancing surfactant production, and restoring endothelial barrier function to improve oxygenation in acute lung conditions.[2][3]First identified in the 1970s as a gastrointestinal hormone, VIP's synthetic counterpart aviptadil has been explored since the 1980s for therapeutic applications beyond its natural roles in vasodilation and smooth muscle relaxation.[3] In 2006 and 2007, the European Medicines Agency granted orphan designations for its use in acute lung injury and sarcoidosis, respectively, recognizing its potential to reduce inflammation in lung tissues.[3] By 2015, aviptadil received approval in Sweden and several other European countries under the brand name Invicorp, in combination with phentolamine, for treating erectile dysfunction unresponsive to oral phosphodiesterase-5 inhibitors, leveraging its vasodilatory effects.[3]In the context of respiratory failure, aviptadil has garnered significant attention for acute respiratory distress syndrome (ARDS), earning U.S. Food and Drug Administration (FDA) orphan drug designations for ARDS (2001) and sarcoidosis (2020).[4][5] During the COVID-19 pandemic, it received FDA Fast Track designation for ARDS associated with the disease, with phase 2/3 clinical trials (e.g., NCT04311697 and NCT04360096) evaluating intravenous and inhaled formulations in critically ill patients.[6][7] However, results from these trials, including a 2023 Lancet Respiratory Medicine study, indicated no significant improvement in clinical outcomes or survival up to 90 days compared to placebo, leading the FDA to decline emergency use authorization and breakthrough therapy designation requests.[8] As of 2025, aviptadil remains investigational for pulmonary indications in the U.S., with ongoing research into its role in post-COVID lung damage and other inflammatory lung diseases.[9]
Vasoactive intestinal peptide (VIP)
Structure and synthesis
Vasoactive intestinal peptide (VIP) is a 28-amino acid neuropeptide belonging to the glucagon/secretin superfamily of peptides. Its primary structure consists of the sequence His-Ser-Asp-Ala-Val-Phe-Thr-Asp-Asn-Tyr-Thr-Arg-Leu-Arg-Lys-Gln-Met-Ala-Val-Lys-Lys-Tyr-Leu-Asn-Ser-Ile-Leu-Asn-NH₂, with the C-terminal asparagine residue amidated, a modification essential for its biological activity.[10][11] This amidation occurs post-translationally via the action of peptidylglycine alpha-amidating monooxygenase (PAM) enzymes on the glycine-extended precursor form.[12]VIP is synthesized from a precursor protein known as prepro-VIP, encoded by the VIP gene located on human chromosome 6q25.2. The gene spans approximately 10 kb and consists of seven exons, with the mature VIP sequence derived from exons 5 and 6. Biosynthesis begins with transcription of the VIP gene in neural crest-derived cells, particularly neurons of the enteric nervous system, central nervous system, and other neuroendocrine tissues, followed by translation into the 170-amino acid prepro-VIP polypeptide. Proteolytic processing in the endoplasmic reticulum and Golgi apparatus cleaves the signal peptide and generates the mature peptide, often co-produced with peptide histidine-methionine (PHM) from the same precursor.[13][14][15]The structure of VIP exhibits high evolutionary conservation across mammals, reflecting its fundamental roles in physiological regulation, with the peptide sequence showing near-identity between human, porcine, and rodent forms. As a member of the glucagon/secretin family, VIP shares structural homology, including a conserved N-terminal histidine and an alpha-helical C-terminal domain critical for receptor binding, underscoring its ancient origins traceable to early chordate evolution through gene duplication events.[16][17]
Receptors and signaling pathways
Vasoactive intestinal peptide (VIP) primarily exerts its effects through two closely related G protein-coupled receptors (GPCRs), known as VPAC1 and VPAC2. These receptors exhibit high affinity for VIP, with both VPAC1 and VPAC2 binding VIP and pituitary adenylate cyclase-activating polypeptide (PACAP) with comparable affinity. VPAC1 is widely distributed in the central nervous system (CNS) and lungs, whereas VPAC2 is prominent in smooth muscle and immune cells.[18][19][20]Structurally, both VPAC1 and VPAC2 belong to class B GPCRs, characterized by seven transmembrane α-helical domains and a large extracellular N-terminal domain that facilitates ligand binding. This N-terminal region, exceeding 120 amino acids, contains conserved cysteine residues forming disulfide bridges essential for receptor stability and function. Upon VIP binding, these receptors predominantly couple to the stimulatory G protein (Gs), activating adenylyl cyclase and thereby elevating intracellular cyclic adenosine monophosphate (cAMP) levels, which in turn activates protein kinase A (PKA).[18][21]The primary signaling cascade initiated by VPAC1 and VPAC2 activation leads to diverse physiological outcomes. Increased cAMP and PKA activity promote bronchodilation through relaxation of airway smooth muscle cells and vasodilation by inhibiting vascular smooth muscle contraction. Additionally, this pathway exerts anti-inflammatory effects by suppressing nuclear factor kappa B (NF-κB) activity, thereby reducing the production of pro-inflammatory cytokines in immune cells. While VPAC1 and VPAC2 share these core mechanisms, subtle differences exist; for instance, VPAC1 is notably expressed in the liver and T-lymphocytes, whereas VPAC2 predominates in the gastrointestinal tract and suprachiasmatic nucleus.[19][18]
Physiological roles and expression
Vasoactive intestinal peptide (VIP) is widely expressed throughout the central and peripheral nervous systems, with the highest levels found in the enteric nervous system, where it constitutes up to 1% of total neuropeptides in the gut.[22] It is also prominently localized in pulmonary nerves, which innervate airway smooth muscle and epithelium, as well as in the adrenal medulla, where it co-localizes with catecholamines in chromaffin cells.[22] Additional sites of dense expression include the gastrointestinal tract, particularly the myenteric and submucosal plexuses, the lungs for bronchodilatory effects, and reproductive tissues such as the uterine cervix and vagina, where it modulates smooth muscle tone.[22] VIP expression is regulated by neuronal activity, which stimulates its release from nerve terminals, and by hormones such as glucocorticoids, which can enhance or suppress VIP gene transcription in a tissue-specific manner.[23]In the central nervous system, VIP functions as a neurotransmitter, particularly in the suprachiasmatic nucleus, where it plays a critical role in synchronizing circadian rhythms by coordinating neuronal firing and transmitting photic signals to regulate glucocorticoid release via the hypothalamic-pituitary-adrenal axis.[24] In the cardiovascular system, VIP acts as a potent vasodilator, promoting relaxation of vascular smooth muscle and increasing cardiac output through direct effects on coronary and peripheral vessels.[25] As an immunomodulator, VIP suppresses pro-inflammatory cytokines such as TNF-α and IL-6 by acting on immune cells like T lymphocytes and macrophages, thereby dampening inflammatory responses; these effects are mediated through signaling via VPAC receptors.[22]VIP's localization in the gastrointestinal tract enables it to regulate motility by relaxing smooth muscle and stimulating epithelial secretion of water and electrolytes, while in the lungs, it induces bronchodilation and inhibits mucus hypersecretion to maintain airway patency.[26] In reproductive tissues, it controls smooth muscle relaxation, facilitating processes like cervical dilation during labor.[27]Pathophysiologically, decreased VIP levels are observed in inflammatory conditions, such as asthma, where reduced peptide concentrations in bronchoalveolar lavage fluid and plasma correlate with heightened airway hyperresponsiveness and eosinophil recruitment.[28] Similarly, in sepsis models, VIP levels decline in certain tissues like the coronary artery, contributing to exacerbated inflammation and vascular dysfunction.[29]
Aviptadil pharmacology
Chemical properties and formulations
Aviptadil is a synthetic 28-amino acidpeptide analog of vasoactive intestinal peptide (VIP), possessing an identical amino acid sequence to the endogenous human VIP but manufactured through solid-phase peptide synthesis for therapeutic purposes.[30]This structure confers vasoactive properties while allowing for pharmaceutical optimization. The molecule has a molecular formula of C_{147}H_{237}N_{43}O_{43}S and a molecular weight of approximately 3,327 Da.[10]As a hydrophilic peptide, aviptadil demonstrates high solubility in aqueous solutions, typically up to 1 mg/mL in water, which facilitates its preparation for administration.[31][32]It is commonly provided in lyophilized powder form within sterile vials, such as borosilicate glass, to preserve integrity during storage and enable reconstitution with sterile water or saline immediately prior to use.[33][34]Although aviptadil shares VIP's susceptibility to rapid enzymatic degradation by proteases such as neutral endopeptidase in biological environments, its synthetic production and specialized formulations mitigate chemical instability during manufacturing, storage, and delivery, achieving shelf stability of up to 12 months or more under controlled conditions.[35][36]Key pharmaceutical formulations of aviptadil include RLF-100, a proprietary acetatesalt version developed by Relief Therapeutics as an inhaled nebulized solution or intravenous preparation in saline, demonstrating high purity and stability across temperatures suitable for global distribution.[37][38]ZYESAMI, licensed by NRx Pharmaceuticals (in collaboration with Relief Therapeutics), is an intravenous acetate formulation of aviptadil designed for acute respiratory applications.[39][40]For erectile dysfunction, aviptadil is formulated as Invicorp, a combination intracavernosal injection containing 25 micrograms of aviptadil and 2 mg of phentolamine mesilate in a sodium chloride solution (pH 3.4–3.8), buffered with phosphate and stabilized with edetate disodium for direct penile administration.[41]Aviptadil's development originated in the 1990s from foundational research on VIP by Sami I. Said at Stony Brook University, with initial focus on vasodilatory applications; Relief Therapeutics holds key patents, including US8178489B2, covering stable formulations optimized for pulmonary delivery via inhalation or intravenous routes.[42][41]
Pharmacokinetics and administration routes
Aviptadil is administered through multiple routes tailored to specific therapeutic indications. Inhalation via nebulization is the preferred method for targeting pulmonary conditions, enabling direct delivery to the alveoli with minimal systemic exposure and reduced risk of pharmacokinetic interactions. Intravenous infusion provides rapid systemic distribution and is commonly used in critical care settings for conditions requiring broad physiological effects. For erectile dysfunction, intracavernosal injection—often combined with phentolamine mesilate—allows localized action within the corpora cavernosa.Following inhalation, aviptadil is absorbed primarily through the pulmonary epithelium, bypassing first-pass hepatic metabolism and achieving high concentrations at the site of action in the lung tissue. Intravenous administration results in instantaneous entry into the plasma, facilitating quick onset of effects. Intracavernosal delivery ensures direct, localized absorption with limited dissemination beyond the injection site.Aviptadil undergoes rapid enzymatic degradation, similar to its endogenous counterpart vasoactive intestinal peptide (VIP). It is primarily metabolized by dipeptidyl peptidase IV (DPP-IV, also known as CD26), which cleaves the N-terminal dipeptide, and neutral endopeptidase (NEP, EC 3.4.24.11), which hydrolyzes internal peptide bonds, particularly in the lungs and vasculature. The plasma half-life after intravenous administration is short, approximately 1–2 minutes, due to this swift proteolytic breakdown. In contrast, inhaled administration extends the effective local half-life in the pulmonary compartment by limiting systemic circulation and degradation.The apparent volume of distribution for aviptadil is approximately 14 mL/kg, reflecting its affinity for VPAC receptors in the lungs, vascular endothelium, and other tissues expressing these G protein-coupled receptors. Excretion occurs mainly through the kidneys as inactive metabolites, with about 35% eliminated within the first 4 hours and up to 90% within 24 hours post-administration.
Mechanism of action
Aviptadil, a synthetic analog of vasoactive intestinal peptide (VIP), exerts its primary effects by binding to VPAC1 and VPAC2 receptors, which are class B G-protein-coupled receptors predominantly expressed in lungtissue and immune cells.[43] This binding activates adenylate cyclase, leading to elevated intracellular levels of cyclic adenosine monophosphate (cAMP).[44] The subsequent activation of protein kinase A (PKA) by cAMP initiates downstream signaling that modulates various cellular processes, including inflammation and smooth muscle tone.[45]Through this cAMP-PKA pathway, aviptadil inhibits the production of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), thereby mitigating cytokine storm-like responses without broadly suppressing immune function.[43] This anti-inflammatory action involves suppression of the mitogen-activated protein kinase (MAPK) pathway, which reduces phosphorylation of key components like c-Jun and extracellular signal-regulated kinase (ERK), ultimately blocking nuclear factor-kappa B (NF-κB) activation and chemokine release from innate immune cells.[46][47]In the lungs, aviptadil promotes alveolar repair by protecting type II alveolar epithelial cells from apoptosis and restoring barrier integrity at the endothelial-alveolar interface, which reduces vascular permeability and prevents edema formation.[7] It also induces surfactant production by upregulating choline phosphate cytidylyltransferase in type II cells, supporting alveolar stability and gas exchange without inducing systemic immunosuppression, as it enhances both innate and adaptive immune responses.[48][49]Aviptadil's vasodilatory effects arise from PKA-mediated relaxation of vascular and bronchial smooth muscle, achieved through phosphorylation and inhibition of voltage-gated calcium channels, which decreases intracellular calcium influx and improves pulmonary blood flow.[50][51] Compared to natural VIP, aviptadil demonstrates enhanced potency in inflamed tissues due to its formulation stability, which resists rapid peptidase degradation and minimizes receptor desensitization (tachyphylaxis) for more sustained therapeutic activity.[52]
Clinical applications
Acute respiratory distress syndrome (ARDS)
Aviptadil has been explored as a therapeutic agent for acute respiratory distress syndrome (ARDS), a severe form of lunginjury characterized by cytokine-mediated damage to the alveolar epithelium and endothelium, resulting in increased vascular permeability, pulmonary edema, and profound hypoxemia. The rationale for its use stems from its ability to counteract these pathological processes through stabilization of the pulmonary endothelium and reduction of edema formation. By binding to VPAC receptors on alveolar type II cells and vascular endothelial cells, aviptadil promotes endothelial barrier integrity, inhibits excessive fluid leakage into the alveolar space, and enhances surfactant production to maintain lung compliance.[53][54]Additionally, aviptadil modulates the inflammatory response central to ARDS progression by suppressing the cytokine storm, including reduced production of pro-inflammatory cytokines such as TNF-α and IL-6, while fostering anti-inflammatory signaling. This dual action on inflammation and vascular stability addresses the core mechanisms of alveolar damage in ARDS, potentially preventing further deterioration and supporting recovery of gas exchange function. Preclinical and early human studies have demonstrated these protective effects, positioning aviptadil as a targeted intervention for cytokine-driven lung injury.[53][48]Key clinical investigations include a phase 1 trial in the early 2000s involving eight patients with severe sepsis-related ARDS on mechanical ventilation, where escalating intravenous doses of VIP (aviptadil's parent compound) led to substantial clinical improvement in seven patients, with six successfully weaned from ventilation. These findings highlight aviptadil's potential to lower mortality by 20-30% in critical ARDS cases through intravenous administration.[55][56]In trials, aviptadil is typically administered intravenously as a continuous infusion over three consecutive days with escalating doses to optimize efficacy while minimizing side effects: 0.166 mcg/kg/hour for 10-12 hours on day 1, 0.332 mcg/kg/hour on day 2, and 0.498 mcg/kg/hour on day 3. For a standard adult patient (e.g., 70 kg), this approximates 12-35 mcg/hour, though fixed doses around 100-200 mcg/hour have been referenced in some protocols for practical administration. Inhaled formulations, delivering equivalent lung-targeted exposure, have been tested at 67 mcg three times daily via nebulization.[53][57]Despite promising early data, aviptadil's clinical application in ARDS faces limitations, primarily due to its short plasmahalf-life of about 2 minutes, necessitating continuous intravenous infusion for sustained therapeutic levels. As of November 2025, aviptadil lacks regulatory approval for routine use in non-COVID-19 ARDS, with ongoing needs for larger phase 3 trials to confirm efficacy across diverse etiologies such as sepsis or trauma-induced lung injury.[48][56]
Erectile dysfunction (ED)
Aviptadil, a synthetic analog of vasoactive intestinal peptide (VIP), is administered via intracavernosal injection to treat erectile dysfunction (ED) by promoting relaxation of the corpus cavernosum smooth muscle. This action enhances the veno-occlusive mechanism essential for maintaining erection, operating through a nitric oxide-independent pathway that increases cyclic AMP levels to facilitate smooth muscle dilation.[58] When combined with phentolamine, an alpha-adrenergic antagonist, aviptadil complements arterial inflow promotion without significantly impacting veno-occlusion, resulting in synergistic vasodilation localized to the penile vasculature.[58]The combination of aviptadil and phentolamine was initially formulated as Invicorp, a product containing 25 micrograms of aviptadil and 0.5–2 mg of phentolamine mesylate, which received marketing authorization in several countries including the UK and Denmark before being discontinued due to commercial reasons.[59] Recent formulations, often referred to as AvP (aviptadil/phentolamine), have been evaluated in phase 3 and retrospective trials during the 2020s for refractory ED cases, particularly in patients who failed phosphodiesterase-5 (PDE5) inhibitors. In a 2025 retrospective study of 308 men with refractory ED, AvP achieved efficacy rates of 59% overall for enabling penetrative sexual activity, rising to 76% among those previously experiencing pain with alprostadil injections and 36% in non-responders to alprostadil.[60] Earlier clinical data reported success rates of 60–80% in PDE5-inhibitor failures, highlighting its role as a second- or third-line option.[61][58]Administration involves direct intracavernosal injection of 25–50 micrograms of aviptadil combined with 0.5–2 mg of phentolamine, typically self-administered after initial training by healthcare professionals. Onset of erection occurs within 5–10 minutes, with duration lasting 1–2 hours, and use is limited to once daily or three times weekly to minimize risks such as priapism.[62][63]Patient monitoring, including follow-up at three months, ensures safety and efficacy.[60]This therapy offers advantages for patients with cardiovascular contraindications to systemic ED treatments like PDE5 inhibitors, as its localized action minimizes systemic absorption and hemodynamic effects. Additionally, it demonstrates lower rates of penile pain (none reported in some cohorts) and reduced priapism incidence compared to prostaglandin analogs such as alprostadil, improving tolerability in refractory cases.[64][61][58]
Post-COVID lung injury
Post-COVID lung injury, characterized by persistent fibrosis and dyspnea arising from unresolved inflammation, represents a significant sequela in recovered COVID-19 patients, often involving residual alveolar dysfunction that impairs gas exchange and epithelial integrity.[65] Inhaled aviptadil, a synthetic analog of vasoactive intestinal peptide (VIP), targets this alveolar dysfunction by promoting repair of type II epithelial cells and mitigating fibrotic remodeling in the post-acute phase.[66]A key 2025 Phase 2 multicenter, double-blind, placebo-controlled trial evaluated inhaled aviptadil in 80 hospitalized adults with COVID-19pneumonia requiring supplemental oxygen, randomizing them 1:1 to receive the drug or placebo alongside standard care, with follow-up assessments up to 180 days.[66] The study demonstrated accelerated radiological improvement, with statistically significant reductions in CTlungdamage scores at day 28 (p=0.028) compared to placebo, alongside earlier dyspnea relief as measured by the Modified Borg Scale at day 7 (p=0.033), though the latter difference was not sustained at day 28 (p=0.144).[66] These findings suggest aviptadil's potential to hasten recovery from persistent pulmonary sequelae, with shorter hospital discharge times (7.8 vs. 10 days, p=0.049) and lower mortality rates (5.1% vs. 12.2%) observed in the treatment arm.[66] Building on mechanisms explored in acute respiratory distress syndrome (ARDS), aviptadil's adaptation to the chronic post-COVID context highlights its role in addressing prolonged inflammatory cascades.[65]In this setting, aviptadil enhances epithelial repair and reduces fibrosis by inhibiting TGF-β signaling, a key driver of extracellular matrix deposition; as a VIP analog, it suppresses TGF-β1 production in macrophages, thereby attenuating epithelial-mesenchymal transition and fibrotic progression in alveolar tissues.[67] This mechanism aligns with VIP's cytoprotective and anti-inflammatory properties, which counteract the unresolved inflammation contributing to post-COVID dyspnea and fibrosis.[65]These initiatives aim to bridge gaps in managing persistent lung injury, with Phase 3 trials recommended to confirm long-term benefits.[66]
Development and regulatory status
Pre-clinical and early trials
Pre-clinical studies in the 1990s established vasoactive intestinal peptide (VIP), the active component of aviptadil, as protective in animal models of endotoxin-induced lung injury and sepsis. In rat models of endotoxic shock, VIP administration reduced histopathological alterations in the lungs and intestines, down-regulated pro-inflammatory cytokines such as TNF-α and IL-6, and improved survival rates by modulating immune responses. For instance, VIP treatment in lipopolysaccharide (LPS)-challenged rats decreased vascular permeability and inflammatory cell infiltration in the pulmonary vasculature, demonstrating up to a 40% reduction in permeability in some experiments. These findings highlighted VIP's anti-inflammatory effects through cAMP-dependent inhibition of macrophagecytokine production and NF-κB activation.[68][69][70]Early human trials in the 2000s focused on safety and proof-of-concept for aviptadil's anti-inflammatory and vasodilatory properties. A phase 1 open-label study conducted by Sami Said in 2005 evaluated intravenous aviptadil in eight patients with severe sepsis-related acute respiratory distress syndrome (ARDS) on mechanical ventilation, using escalating doses of 50–100 pmol/kg/hr over 12 hours. The trial confirmed tolerability with no serious drug-related adverse events, no immunogenicity, and doses up to 1 mg IV deemed safe; seven of eight patients were successfully weaned from ventilation and discharged from the ICU, with reduced TNF-α levels in responders.[71][72]Initial indications explored included sepsis and pulmonary hypertension, where aviptadil showed proof-of-concept for anti-inflammatory effects. Phase 2 trials in the 2000s demonstrated safety of inhaled aviptadil up to 300 μg daily in patients with pulmonary hypertension and sarcoidosis, with reductions in pulmonary vascular resistance and improved lung function without significant hypotension or immunogenicity. These studies supported aviptadil's role in modulating inflammation in septic and hypertensive conditions.[73][56]Key milestones included the development of synthetic VIP analogs, with a 1996 publication describing hybrid antagonists that informed later formulations for therapeutic use. Early exploration of erectile dysfunction (ED) began in the late 1990s, culminating in 2001 studies showing aviptadil, combined with phentolamine, induced erections in over 80% of men with ED via intracavernosal injection, offering a tolerable alternative with fewer side effects than existing therapies; approval followed in select markets like New Zealand and the United Kingdom by 2000.[74]
COVID-19 and ARDS trials
In response to the COVID-19 pandemic, aviptadil received an Investigational New Drug (IND) application from the FDA in 2020, enabling compassionate use for patients with critical COVID-19-associated acute respiratory distress syndrome (ARDS).[6] This authorization facilitated early access under emergency conditions, particularly for intubated patients facing high mortality risks, prior to formal trial initiation.[75]A pivotal Phase 2b randomized controlled trial (NCT04311697) evaluated intravenous aviptadil (ZYESAMI) in 260 enrolled patients with critical COVID-19respiratory failure, with 196 analyzed (131 aviptadil, 65 placebo).[6] The trial, conducted across multiple U.S. sites starting in April 2020, demonstrated a 2.0-fold increased odds of survival at day 60 (odds ratio [OR] 2.0; 95% CI, 1.1–3.9; p=0.035), alongside improved recovery from respiratory failure.[56] In the remdesivir-treated subgroup, survival odds were further enhanced, reaching a 4.0-fold increase at day 60 (OR 4.0; p=0.006).[75] Overall 60-day mortality was reduced from 46.2% in the placebo group to 35.1% with aviptadil.[56]Subgroup analyses highlighted greater benefits among intubated patients, where aviptadil yielded a 10.4-fold increased odds of survival (p=0.031), and in those with elevated inflammatory cytokines, as evidenced by attenuated rises in IL-6 levels (p=0.024).[56] These findings underscored aviptadil's potential to mitigate cytokine-driven lunginflammation in severe cases. The trial met its primary endpoint of time to recovery from respiratory failure at days 28 (p=0.014) and 60 (p=0.015).[38]Parallel efforts explored inhaled aviptadil (RLF-100) in a Phase 2 randomized, placebo-controlled trial (NCT04844580) involving 80 hospitalized COVID-19 patients at high risk for ARDS, completed in 2022.[76] Administered via nebulizer alongside standard care, inhaled aviptadil significantly shortened hospital stays (mean 7.8 days vs. 10 days; p=0.049) and improved lung damage scores on CT scans by day 28 (p=0.028).[66] It also reduced dyspnea on the modified Borg scale by day 7 (mean 1.8 vs. 2.7; p=0.033), indicating enhanced oxygenation and respiratory comfort, though mortality differences were not statistically significant (5.1% vs. 12.2%; p=0.433).[66]Regulatory challenges emerged in 2022 when the FDA denied Emergency Use Authorization (EUA) for ZYESAMI in critical COVID-19 patients, citing insufficient evidence from post-hoc analyses and conflicting data from the ACTIV-3/TESICO trial, which showed no overall benefit.[77] A prior EUA denial in November 2021 similarly pointed to inadequate safety and efficacydata from limited patient reviews.[78] By 2023, aviptadil had not received full FDA approval for COVID-19 or ARDS indications, halting further EUA pursuits amid evolving pandemic dynamics.[79]
Current status and future prospects
As of November 2025, aviptadil remains unapproved by the FDA and the centralized EMA for any indication and is investigational for pulmonary uses globally, though it is approved in select countries for erectile dysfunction in combination with phentolamine. The U.S. FDA's last denial of emergency use authorization for ZYESAMI (aviptadil acetate) occurred in 2022 for patients with critical COVID-19 at immediate risk of death from respiratory failure despite standard therapies, including remdesivir.[80] This follows prior denials of breakthrough therapy designation and earlier EUA requests. In the European Union, aviptadil holds orphan drug designation for acute respiratory distress syndrome (ARDS), originally granted for its potential in rare pulmonary conditions, alongside U.S. orphan status for ARDS and sarcoidosis.[73][81]Despite the lack of full approval, aviptadil is accessible in the United States through expanded access programs for critical cases of respiratory failure, particularly in COVID-19 patients who have exhausted approved treatments, as outlined in ongoing protocols from NRx Pharmaceuticals and collaborators.[82] Relief Therapeutics continues to advance an inhaled formulation (RLF-100) for post-COVID lung injury, with recent stability data supporting its viability and early clinical evidence suggesting benefits in radiological recovery and dyspnea reduction. A phase II study published in January 2025 on inhaled aviptadil for post-COVID lung injury showed reduced hospital stays (7.8 vs. 10 days; p=0.049), improved CT lung damage scores (p=0.028), and lower dyspnea scores (p=0.033) compared to placebo, though mortality differences were not significant.[37][83][66]Looking ahead, development focuses on broader pulmonary applications beyond COVID-19, including Phase I trials for general ARDS (potentially sepsis-induced) and planned Phase III studies for inhaled aviptadil in long COVID-related lung damage to confirm efficacy and safety.[84][66] Exploratory combinations with antivirals, such as remdesivir, have been tested in prior ARDS trials, though results were inconclusive, prompting further investigation in non-viral contexts.[8] If approved for pulmonary indications, aviptadil could capture a share of the ARDS market, projected at approximately $1.41 billion in 2025 and growing to $2.17 billion by 2029, driven by unmet needs in critical care.[85]Key challenges include scaling peptide manufacturing for consistent purity and yield, as aviptadil's synthetic vasoactive intestinal peptide structure requires specialized processes to ensure stability for clinical and commercial use.[86] Additionally, competition from established biologics like tocilizumab, which targets IL-6 in inflammatory ARDS, may limit adoption unless aviptadil demonstrates superior lung-specific benefits in upcoming trials.[87]