Biotech
124584-08-3
Acute Decompensated Heart Failure (ADHF)
Nesiritide is a synthetically produced, purified preparation of human B-type natriuretic peptide (hBNP), a hormone endogenously produced by the ventricular myocardium in response to increased wall stress and volume overload.[1] It is classified as a biotech drug, manufactured using recombinant DNA technology with an
Escherichia coli host system.[2] As a therapeutic agent, Nesiritide is identical in its amino acid composition to the native human peptide, designed to replicate its physiological functions in a clinical setting.[2]
The peptide consists of a single chain of 32 amino acids.[1] Its structure is characterized by a 17-amino acid ring formed by an intramolecular disulfide bond between the cysteine residues at positions 10 and 26.[6] This ring structure is a conserved feature across the family of natriuretic peptides and is crucial for receptor binding and biological activity. The full amino acid sequence of Nesiritide is: Ser-Pro-Lys-Met-Val-Gln-Gly-Ser-Gly-Cys-Phe-Gly-Arg-Lys-Met-Asp-Arg-Ile-Ser-Ser-Ser-Ser-Gly-Leu-Gly-Cys-Lys-Val-Leu-Arg-Arg-His.[5]
For clinical use, Nesiritide is formulated as a citrate salt and supplied as a sterile, white to off-white lyophilized powder that requires reconstitution before intravenous administration.[2] Its fundamental chemical and physical properties are well-defined, with a chemical formula of
C143H244N50O42S4 and a molecular weight of approximately 3464.0 g/mol.[1] The drug has been assigned numerous identifiers across various chemical and pharmacological databases, ensuring its unambiguous identification for research, clinical, and regulatory purposes.
Table 1: Key Identifiers and Properties of Nesiritide
Attribute | Value | Source(s) |
---|---|---|
Generic Name | Nesiritide | 2 |
Brand Name | Natrecor | 1 |
Type / Modality | Biotech / Protein-Based Therapy | 5 |
Drug Class | Vasodilator, Natriuretic Peptide | 5 |
CAS Number | 124584-08-3 | 1 |
DrugBank ID | DB04899 | 1 |
FDA UNII | P7WI8UL647 | 1 |
ATC Code | C01DX19 (Other vasodilators used in cardiac diseases) | 1 |
Chemical Formula | C143H244N50O42S4 | 1 |
Molar Mass | 3464.07 g·mol−1 | 1 |
Amino Acid Sequence | SPKMVQGSGCFGRKMDRISSSSGLGCKVLRRH | 5 |
Formulation | Lyophilized powder (citrate salt) for IV administration | 2 |
The therapeutic rationale for Nesiritide is rooted in its ability to mimic the actions of endogenous hBNP, a key component of the body's counter-regulatory system against the neurohormonal over-activation that characterizes heart failure.[4] Its mechanism of action is initiated by binding to specific natriuretic peptide receptors (NPRs) located on the surface of target cells.[5] The primary signaling receptor is the natriuretic peptide receptor-A (NPR-A), a particulate guanylate cyclase-linked receptor highly expressed in vascular smooth muscle and endothelial cells.[3] Nesiritide also interacts with the clearance receptor, NPR-C, which is involved in its metabolism.[5]
Activation of NPR-A stimulates its intrinsic guanylate cyclase activity, leading to the conversion of guanosine triphosphate (GTP) to the intracellular second messenger, 3',5'-cyclic guanosine monophosphate (cGMP).[1] The subsequent elevation of intracellular cGMP concentrations orchestrates a cascade of physiological effects that are beneficial in the setting of acutely decompensated heart failure (ADHF).[1]
These effects include:
These cellular and systemic actions translate into a distinct and measurable pharmacodynamic profile. In clinical studies with heart failure patients, Nesiritide administration produces rapid, dose-dependent hemodynamic improvements.[3] Within 15 minutes of administration, significant reductions in pulmonary capillary wedge pressure (PCWP)—a critical measure of left ventricular filling pressure and pulmonary congestion—are observed.[3] The therapy also leads to reductions in right atrial pressure and systemic vascular resistance, along with an increase in cardiac index and stroke volume.[7]
A key differentiating feature of Nesiritide's pharmacodynamic profile, particularly when compared to traditional inotropic agents like dobutamine, is that these hemodynamic benefits are achieved without directly affecting myocardial contractility (inotropism) or significantly increasing heart rate (chronotropism).[3] This was perceived as a major advantage, as it suggested that Nesiritide could unload the failing heart without increasing its oxygen demand or provoking arrhythmias. This compelling biological rationale, portraying Nesiritide as a "physiologic" therapy that restores a natural hormonal balance, created a powerful narrative that drove its initial adoption and set high expectations for its clinical performance.[4]
Nesiritide is administered exclusively by the intravenous (IV) route, typically as an initial weight-based bolus dose followed by a continuous infusion to maintain steady-state plasma concentrations.[1]
Upon entering the circulation, Nesiritide exhibits a biphasic disposition from the plasma.[2] The pharmacokinetics are characterized by a very rapid initial elimination phase with a half-life of approximately 2 minutes, followed by a mean terminal elimination half-life (
t1/2) of approximately 18 minutes.[2] The mean volume of distribution of the central compartment (
Vc) is estimated at 0.073 L/kg, with a steady-state volume of distribution (Vss) of 0.19 L/kg.[2] The mean clearance (CL) is approximately 9.2 mL/min/kg.[2]
Despite its short pharmacokinetic half-life, the pharmacodynamic effects of Nesiritide are more prolonged.[3] For example, after discontinuation of the infusion in patients who develop hypotension, the recovery of systolic blood pressure can take an hour or longer, indicating that the biological effects mediated by cGMP persist well after the drug has been substantially cleared from the plasma.[3]
The clearance of Nesiritide from the body is a multi-faceted process involving three independent mechanisms, listed in order of decreasing importance [3]:
Population pharmacokinetic analyses have shown that Nesiritide clearance is proportional to body weight, providing the rationale for the standard weight-adjusted dosing regimen (e.g., mcg/kg/min).[2] These analyses also indicated that clearance was not significantly influenced by patient demographics such as age, gender, or race, nor by the severity of heart failure or concomitant administration of angiotensin-converting enzyme (ACE) inhibitors.[2]
Notably, early clinical data suggested that dose adjustment was not required for patients with renal impairment.[2] The hemodynamic effects on PCWP and blood pressure were reported to be similar in patients with chronic renal insufficiency (baseline serum creatinine up to 4.3 mg/dL) and those with normal renal function.[2] This initial finding created a perception of safety in a patient population where renal dysfunction is common and often a limiting factor for other cardiovascular therapies. This perception, however, would later be directly challenged by post-marketing analyses, foreshadowing a central controversy in the drug's history and highlighting a potential disconnect between controlled pharmacokinetic studies and real-world safety outcomes in a broader, more complex patient population.
Nesiritide entered the clinical arena as the first new parenteral agent for heart failure in over a decade, representing a novel therapeutic approach grounded in physiological hormone replacement.[4] After an initial rejection and a request for additional data from the U.S. Food and Drug Administration (FDA) in 1999, Scios Inc. secured approval for Natrecor (Nesiritide) on August 10, 2001.[1] The approved indication was narrow and specific: "for the intravenous treatment of patients with acutely decompensated congestive heart failure who have dyspnea at rest or with minimal activity".[15]
The approval was granted not on the basis of improved mortality or rehospitalization rates, but on the drug's demonstrated ability to improve hemodynamic surrogate endpoints. The pivotal trial supporting the application was the Vasodilation in the Management of Acute CHF (VMAC) study.[4] In this randomized, double-blind trial, Nesiritide was compared to both placebo and standard-of-care intravenous nitroglycerin.[26] The results showed that after 3 hours of infusion, Nesiritide produced a statistically significant reduction in pulmonary capillary wedge pressure (PCWP) compared to both placebo and nitroglycerin.[26] Furthermore, patients receiving Nesiritide reported a modest but statistically significant improvement in dyspnea compared to placebo, although this symptomatic benefit was not superior to that seen with nitroglycerin.[26]
The early perception of Nesiritide was overwhelmingly positive. It was seen as an elegant solution to a complex problem, offering potent vasodilation and hemodynamic unloading without the deleterious proarrhythmic and chronotropic effects associated with inotropic agents like dobutamine.[16] This promising profile, supported by the surrogate endpoint data from VMAC, led to its rapid adoption in clinical practice.
The trajectory of Nesiritide was irrevocably altered by safety concerns that emerged in 2005 (detailed in Section 3.2). These concerns, coupled with the lack of definitive outcomes data, prompted the design and execution of the Acute Study of Clinical Effectiveness of Nesiritide in Decompensated Heart Failure (ASCEND-HF).[1] This landmark trial was intended to provide a final, unambiguous verdict on the drug's clinical value.
ASCEND-HF was a massive, international, multicenter, randomized, double-blind, placebo-controlled trial that enrolled 7,141 patients hospitalized for ADHF.[31] Patients were randomized to receive either Nesiritide (an optional 2 mcg/kg bolus followed by a 0.01 mcg/kg/min continuous infusion) or a matching placebo, in addition to standard care, for a period of 24 to 168 hours.[32] The study was rigorously designed to evaluate clinically meaningful, patient-centered outcomes.
The trial failed to meet both of its co-primary endpoints, delivering a profoundly negative result for the drug:
Secondary endpoints, such as death prior to discharge or days alive and out of the hospital, also showed no benefit for Nesiritide.[33] Crucially, the trial provided definitive safety data. It did not find a statistically significant increase in 30-day mortality or in the primary renal endpoint (worsening renal function, defined as a >25% decrease in estimated glomerular filtration rate).[30] However, it did confirm a known risk, showing a significantly higher rate of both symptomatic hypotension (7.2% vs. 4.0%) and asymptomatic hypotension (21.4% vs. 12.4%) in the Nesiritide arm.[33]
Table 2: Summary of Primary and Key Secondary Outcomes of the ASCEND-HF Trial
Endpoint | Nesiritide Group | Placebo Group | Statistical Comparison | Clinical Interpretation |
---|---|---|---|---|
Co-Primary: Dyspnea Improvement (Mod/Marked @ 24h) | 68.2% | 66.1% | p = 0.007 | Statistically significant but not clinically meaningful; did not meet predefined significance level. |
Co-Primary: Death or HF Rehospitalization @ 30 days | 9.4% | 10.1% | HR 0.93 (95% CI 0.84-1.03) | No significant difference; trial failed to meet this endpoint. |
Component: All-Cause Death @ 30 days | 3.6% | 4.0% | HR 0.90 (95% CI 0.73-1.11) | No significant difference in mortality. |
Key Safety: Symptomatic Hypotension | 7.2% | 4.0% | p < 0.001 (NNH = 32) | Significantly higher risk of symptomatic hypotension with Nesiritide. |
Key Safety: Worsening Renal Function (>25% eGFR drop) | 31.4% | 29.5% | p = 0.11 | No statistically significant difference in the primary renal endpoint. |
Sources:.[1] NNH = Number Needed to Harm.
The clinical story of Nesiritide is one of a stark disconnect between physiological effect and patient benefit. The drug reliably and effectively improves hemodynamic surrogate markers, such as PCWP, which was the basis for its initial approval and widespread adoption.[19] This created a compelling narrative that improving the numbers would inevitably lead to improved patient outcomes.
However, the ASCEND-HF trial, the largest and most definitive study ever conducted on the drug, shattered this assumption. It demonstrated unequivocally that the hemodynamic improvements did not translate into patients feeling substantially better, staying out of the hospital, or living longer.[34] The trial's failure to show a meaningful benefit on any hard clinical endpoint, coupled with the confirmed risk of hypotension, effectively erased any remaining rationale for its routine use. This journey serves as a powerful and frequently cited cautionary tale in modern medicine about the "surrogate endpoint trap": the danger of approving and adopting therapies based on their ability to change a laboratory or physiological marker, without first proving that this change leads to tangible, patient-centered clinical improvement.[35] The experience with Nesiritide reinforced a paradigm shift in cardiovascular clinical research, solidifying the principle that hard outcomes like mortality and morbidity, not just physiological measurements, must be the ultimate arbiters of a drug's value.
The safety profile of Nesiritide is dominated by its primary pharmacodynamic effect: vasodilation. The most common and clinically significant adverse event is hypotension, which is dose-dependent and serves as the principal dose-limiting toxicity.[1] Clinical trials have reported rates of hypotension ranging from 4% to as high as 35%, depending on the dose and patient population.[20] The landmark ASCEND-HF trial provided the most robust data, confirming a significantly increased risk of both symptomatic hypotension (7.2% for Nesiritide vs. 4.0% for placebo) and asymptomatic hypotension (21.4% vs. 12.4%).[33] Management of hypotension requires close blood pressure monitoring and may necessitate a reduction in the infusion rate or temporary discontinuation of the drug.[2]
Other commonly reported adverse effects (occurring in 1-10% of patients) include headache, nausea, back pain, dizziness, insomnia, and anxiety.[1] While Nesiritide was shown to be less arrhythmogenic than dobutamine, ventricular arrhythmias, including ventricular tachycardia and premature ventricular beats, have been reported.[20]
Serious adverse events, although less common, include severe allergic or hypersensitivity reactions, manifesting as hives, rash, pruritus, or angioedema.[2] Infusion site extravasation has also been noted in postmarketing reports.[2]
Beyond the expected side effect of hypotension, the history of Nesiritide is defined by a major safety controversy that erupted in 2005, years after its approval. This debate fundamentally altered the drug's perception and clinical use long before the definitive ASCEND-HF trial was completed.
In March and April of 2005, two meta-analyses of existing randomized controlled trials were published by Dr. Jonathan Sackner-Bernstein and colleagues in the high-impact journals Circulation and JAMA.[1] These analyses raised two alarming safety signals:
The publication of these findings sent shockwaves through the cardiology community and triggered an immediate and forceful response. The FDA collaborated with the manufacturer, Scios, to revise the drug's prescribing information to include warnings about these potential risks.[1] Prominent medical centers, such as the Cleveland Clinic, voted to severely restrict or ban the drug's use within their institutions.[1] An expert panel of cardiologists, convened at the manufacturer's request, issued recommendations to strictly limit Nesiritide's use to its narrow FDA-approved indication (hospitalized ADHF patients with dyspnea at rest) and explicitly advised against its off-label use to improve renal function or enhance diuresis.[18] The impact on clinical practice was dramatic and swift, with studies documenting a phenomenon of "rapid de-adoption" as prescriptions plummeted.[22]
The ASCEND-HF trial was designed in large part to provide a definitive answer to this controversy.[29] The trial's results appeared to acquit Nesiritide of the most serious charges: it found no statistically significant difference in 30-day mortality or in its primary endpoint for worsening renal function.[33] However, the story was more nuanced. A subsequent sub-analysis of the ASCEND-HF data revealed that the definition of renal injury was critical. While the primary endpoint (a relative >25% drop in eGFR) was not met, an alternative endpoint (an absolute increase in serum creatinine of ≥0.5 mg/dL) did show a statistically significant increase in the odds of renal injury with Nesiritide.[30]
This sequence of events illustrates a critical principle in pharmacovigilance: a strong signal of harm, even if generated from less-than-definitive data like a meta-analysis of small trials, can create a perception of risk that is nearly impossible to erase. The medical community, operating under the principle of "first, do no harm," reacted to the plausible threat. By the time the larger, more definitive ASCEND-HF trial provided a more reassuring (though still nuanced) safety profile, it was too late. The drug had already been proven to lack meaningful efficacy, and therefore, even a small lingering suspicion of harm was enough to render its risk/benefit profile unacceptable.
The clinical use of Nesiritide is governed by specific contraindications and a need for caution regarding drug interactions.
Contraindications:
Drug Interactions:
To fully understand the clinical role and ultimate failure of Nesiritide, it is essential to evaluate it in the context of the primary alternative intravenous therapies for ADHF: the vasodilator nitroglycerin and the inotropic agent dobutamine. Nesiritide was positioned to fill a perceived gap between these two established agents.
The comparison with nitroglycerin centers on efficacy, tolerability, and cost. Both are vasodilators, but they operate through different mechanisms—Nesiritide via the cGMP pathway and nitroglycerin via nitric oxide donation.
Ultimately, against nitroglycerin, Nesiritide offered a marginal improvement in a surrogate hemodynamic marker and a better side effect profile regarding headaches, but at a vastly higher cost and with no demonstrable superiority in improving patient symptoms.
The comparison with dobutamine, a synthetic catecholamine with primary β1-agonist activity, highlights a different set of trade-offs, focusing on safety versus inotropic support.[42]
This comparative analysis reveals how Nesiritide's clinical utility was constructed from a fragmented rationale. It was not definitively superior to either competitor on all fronts. Instead, it was positioned as a "safer" choice than dobutamine for patients at risk of arrhythmia and a "more potent and convenient" choice than nitroglycerin for hemodynamic unloading. This allowed it to carve out a significant market niche. However, the entire premise of its value, particularly its cost-effectiveness, hinged on assumptions about improved outcomes and reduced length of stay that were never validated in a large, prospective trial. When the definitive ASCEND-HF trial showed no impact on hard clinical outcomes, the economic argument collapsed, leaving a very expensive drug with no proven benefit over cheaper, established alternatives.
Table 3: Comparative Profile of Nesiritide, Nitroglycerin, and Dobutamine in ADHF
Feature | Nesiritide | Nitroglycerin | Dobutamine |
---|---|---|---|
Mechanism of Action | NPR-A agonist, ↑ cGMP | Nitric oxide donor, ↑ cGMP | β1-adrenergic agonist, ↑ cAMP |
Primary Hemodynamic Effect | Balanced arterial/venous vasodilation | Predominantly venous vasodilation | Increased inotropy and chronotropy |
Effect on Heart Rate | Neutral | Can cause reflex tachycardia | Increases heart rate |
Effect on Arrhythmias | Neutral or suppressive | Neutral | Proarrhythmic |
Key Efficacy Evidence | Superior PCWP reduction vs. low-dose NTG; no superior symptom relief 27 | Symptom relief similar to Nesiritide 27 | Symptom relief similar to Nesiritide 37 |
Major Side Effects | Hypotension, renal concerns | Headache, hypotension, tachycardia | Tachycardia, arrhythmias, ischemia |
Dosing Complexity | Simple (bolus + fixed infusion) | Requires frequent titration | Requires titration and monitoring |
Approximate Cost (mid-2000s) | Very High (~$530/day) | Low | Low |
Sources:.[15]
The story of Nesiritide cannot be fully understood through clinical data alone. Its rise and fall were profoundly influenced by a complex interplay of regulatory decisions, aggressive marketing practices that led to legal action, and the subsequent reaction of the medical community.
The two-decade journey of Nesiritide from promising innovation to discontinued product is marked by several pivotal moments that shaped its trajectory.
Table 4: Chronological Timeline of Key Events in the History of Nesiritide
Year | Event | Significance |
---|---|---|
1998 | Scios Inc. files New Drug Application (NDA) with the FDA. | Marks the beginning of the formal regulatory review process. 22 |
1999 | FDA issues an action letter requesting more data. | The initial application is deemed insufficient, delaying approval. 22 |
2001 | FDA approves Nesiritide (Natrecor) for ADHF with dyspnea at rest. | Approval is granted based on hemodynamic surrogate endpoint data. 1 |
2002 | VMAC and PRECEDENT trial results are published. | Provides key comparative data vs. nitroglycerin and dobutamine, supporting early adoption. 22 |
2005 | Sackner-Bernstein meta-analyses are published. | Two papers in high-impact journals suggest increased risks of renal dysfunction and mortality, sparking major controversy. 1 |
2005 | Braunwald Panel convenes and issues recommendations. | An expert panel advises restricting use to the approved indication and warns against off-label use for renal benefit or outpatient infusions. 1 |
2005-2006 | Rapid de-adoption of Nesiritide in clinical practice begins. | Use of the drug plummets following the publication of safety concerns. 22 |
2011 | Scios Inc. pleads guilty to misbranding Natrecor. | The company pays an $85 million criminal fine for illegal off-label promotion. 48 |
2011 | ASCEND-HF trial results are published in the NEJM. | The definitive trial shows no benefit on hard clinical outcomes, effectively ending any rationale for the drug's routine use. 1 |
2018 | Janssen Pharmaceuticals discontinues manufacturing of Nesiritide. | The drug is officially withdrawn from the market, marking the end of its clinical life. 35 |
A defining chapter in the Nesiritide saga was the U.S. Department of Justice (DOJ) investigation into the marketing practices of its manufacturer, Scios Inc., a subsidiary of Johnson & Johnson.[1] The investigation culminated in 2011 when Scios pleaded guilty to a misdemeanor violation of the Food, Drug and Cosmetic Act (FDCA) for the criminal misbranding of Natrecor.[48]
The core of the case was the allegation of illegal off-label promotion. While Natrecor was only approved for short-term, inpatient use for ADHF with dyspnea at rest, the DOJ found that Scios had intentionally promoted the drug for an unapproved use: scheduled, serial, intermittent infusions for patients with chronic (non-acute) heart failure, often administered in outpatient clinics.[49] This practice, sometimes referred to as a "tune-up" infusion, lacked any supporting evidence of benefit and was explicitly discouraged by expert panels.[18]
As part of its plea agreement, Scios admitted that it intended Natrecor to be used in this unapproved manner and that its FDA-approved labeling lacked adequate directions for this use, which constitutes misbranding under the FDCA. The company was sentenced to pay an $85 million criminal fine.[49] In addition to the criminal plea, a related civil lawsuit was filed under the False Claims Act, alleging that the company's promotion of this unproven use caused false claims to be submitted to Medicare and other federal healthcare programs.[49]
This case reveals a damaging feedback loop. The drug's approval on weak surrogate endpoint data left it with an uncertain clinical value proposition. To maximize sales and achieve a return on a significant investment, the manufacturer engaged in aggressive, and ultimately illegal, promotion far beyond the drug's narrow evidence base. This widespread use, in turn, attracted the very academic and regulatory scrutiny that would uncover the safety signals and lead to the drug's eventual downfall.
The decline of Nesiritide was not a gradual process but a rapid collapse triggered by the 2005 safety publications. Data from large hospital databases show that use of the drug peaked in early 2005 and then fell precipitously in the months immediately following the publication of the Sackner-Bernstein meta-analyses.[22] This "rapid de-adoption" occurred years before the definitive ASCEND-HF trial results were available, demonstrating the profound impact of a credible safety signal on clinical practice, especially for a drug with marginal proven benefits.
The publication of the neutral ASCEND-HF results in 2011 served as the final confirmation that there was no compelling reason to use the drug. It had a tarnished safety record, a proven lack of efficacy on hard endpoints, a history of illegal marketing, and was far more expensive than its competitors. Its clinical utility was, as the ASCEND-HF trial investigators concluded, likely to be minimal.[34] The market for the drug evaporated. Finally, in 2018, Janssen Pharmaceuticals, which had absorbed Scios, officially announced the discontinuation of Nesiritide manufacturing, bringing its tumultuous history to a close.[35]
The comprehensive history of Nesiritide—from its elegant physiological conception to its controversial clinical life and ultimate withdrawal—offers several enduring lessons for pharmacology, clinical medicine, and the pharmaceutical industry. Its story is not merely about a single failed drug but serves as a powerful case study on the complexities of modern drug development and evaluation.
The single most important lesson from the Nesiritide saga is the profound danger of relying on surrogate endpoints to judge a drug's clinical worth. Nesiritide was a resounding success at improving its target surrogate marker: it reliably and potently lowered pulmonary capillary wedge pressure.[19] The medical community and regulators initially accepted this physiological improvement as a proxy for clinical benefit, leading to the drug's approval and rapid uptake.[23]
The fundamental assumption was that because high PCWP is bad, a drug that lowers it must be good. The ASCEND-HF trial definitively proved this assumption to be false.[34] The improvement in the hemodynamic number did not translate into patients feeling substantially better, staying out of the hospital, or living longer. This stark disconnect has become a quintessential example used to teach the importance of skepticism toward surrogate markers. The experience has reinforced a now widely held principle in clinical trial design and drug regulation: therapies must demonstrate a tangible benefit on hard, patient-centered outcomes (e.g., mortality, morbidity, quality of life), not just on a physiological measurement.[30]
The story of Nesiritide is a vivid illustration of the dynamic and often contentious relationship between independent science, commercial interests, and regulatory oversight.
Nesiritide failed not because it was proven to be exceptionally dangerous—the most dire fears from the 2005 meta-analyses were not confirmed in ASCEND-HF—but because it was ultimately proven to be clinically ineffective.[34] It offered no meaningful benefit on outcomes that matter to patients, and this lack of efficacy could not justify its high cost and its known risk of hypotension.
Its legacy within cardiology and pharmacology is significant and multifaceted. It stands as a monument to several critical principles of modern, evidence-based medicine:
For a generation of cardiologists and clinical trialists, the story of Natrecor is not just a historical footnote but a formative experience that has shaped clinical skepticism, research priorities, and the standards by which new cardiovascular drugs are judged.[35]
Published at: August 7, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.