MedPath

Hydroxyethyl Starch Advanced Drug Monograph

Published:Aug 1, 2025

Brand Names

Hespan, Hextend, Volulyte, Voluven

Drug Type

Small Molecule

CAS Number

9005-27-0

Associated Conditions

Hypovolaemia

Hydroxyethyl Starch (DB09106): A Comprehensive Monograph on a Controversial Plasma Volume Expander

Abstract

Hydroxyethyl Starch (HES) is a class of synthetic colloids, chemically derived from plant amylopectin, that was developed and widely adopted for intravascular volume expansion. Its primary indication has been the treatment and prevention of hypovolemia resulting from acute blood loss in settings such as surgery and trauma. The mechanism of action is based on the principles of colloid osmosis; the large polysaccharide molecules are retained within the vasculature, exerting oncotic pressure that draws fluid from the interstitial space to expand plasma volume. However, the clinical history of HES is dominated by a significant controversy that has reshaped fluid resuscitation practices globally. A compelling body of evidence, most notably from the landmark randomized controlled trials VISEP, 6S, and CHEST, systematically demonstrated a clear and consistent association between HES administration and severe adverse outcomes, particularly in critically ill and septic patient populations. These trials revealed an increased risk of mortality, a significantly higher incidence of acute kidney injury (AKI) necessitating renal replacement therapy (RRT), and clinically relevant coagulopathy leading to increased bleeding and transfusion requirements. These findings were not confined to older, high-molecular-weight formulations but were also demonstrated with "modern," lower-molecular-weight tetrastarches, refuting the hypothesis that newer agents were safer. The irrefutable evidence of harm prompted decisive regulatory actions, including a Black Box Warning from the U.S. Food and Drug Administration (FDA) and progressive restrictions culminating in a market suspension recommendation from the European Medicines Agency (EMA). Consequently, HES has been largely relegated from clinical practice, replaced by safer and often less expensive alternatives such as balanced crystalloids and, in specific circumstances, human albumin. The trajectory of HES serves as a profound cautionary tale in pharmacovigilance, illustrating the perils of relying on surrogate physiological endpoints and underscoring the indispensable role of large-scale, independent clinical trials in establishing true patient-centered safety and efficacy.

I. Introduction: The Rationale and Rise of a Synthetic Colloid

The Clinical Challenge of Hypovolemia

Hypovolemia, a state of decreased intravascular volume, represents a fundamental threat to physiological homeostasis and is a common pathway for clinical deterioration in a multitude of acute care settings.[1] Conditions such as major trauma, hemorrhagic episodes during surgery, severe burns, and the distributive shock seen in sepsis can lead to a rapid and critical loss of circulating blood volume.[2] The physiological sequelae of untreated hypovolemia are profound, leading to reduced venous return, decreased cardiac output, and ultimately, inadequate tissue perfusion and oxygen delivery. This state of shock, if not promptly reversed, results in cellular hypoxia, organ dysfunction, and death.[2] Consequently, the rapid and effective restoration of intravascular volume is a cornerstone of resuscitation and a primary objective in emergency and critical care medicine.[5]

The Colloid vs. Crystalloid Debate

The therapeutic approach to volume replacement has historically been centered on a debate between two classes of intravenous fluids: crystalloids and colloids.[7] Crystalloids are aqueous solutions of low-molecular-weight ions (e.g., sodium chloride, lactate) that distribute freely throughout the total extracellular fluid compartment. While inexpensive and readily available, a large proportion of infused crystalloid volume rapidly extravasates into the interstitial space, meaning that significantly larger volumes are required to achieve a desired level of plasma expansion.[7]

Colloids, in contrast, are solutions containing large-molecular-weight substances that are retained within the intravascular space for a more extended period due to their limited ability to cross the capillary membrane.[7] This property allows them to exert colloid osmotic (oncotic) pressure, which retains and draws fluid into the circulation. The theoretical advantage of colloids has always been their greater efficiency in plasma volume expansion; a smaller infused volume of a colloid is required to achieve the same hemodynamic effect as a much larger volume of crystalloid.[7] This "volume-sparing" effect was believed to offer faster hemodynamic stabilization and potentially reduce the risk of tissue edema associated with large-volume crystalloid resuscitation.[10]

Emergence of Hydroxyethyl Starch

Within the class of colloids, a distinction exists between natural products, such as human albumin, and synthetic alternatives. Hydroxyethyl Starch (HES) emerged from this landscape as a prominent synthetic colloid, developed between 1944 and 1962.[6] Derived from naturally occurring amylopectin—a branched glucose polymer from plant sources like corn or potatoes—HES was engineered to serve as a plasma volume expander.[2] Its rise to prominence was fueled by several perceived advantages. HES was significantly less expensive to produce than human albumin, was readily available without reliance on blood donors, and was believed to be a highly effective plasma volume expander.[7]

The initial clinical adoption of HES was predicated on a compelling but ultimately narrow physiological rationale. The focus remained on its demonstrated ability to efficiently expand plasma volume—a surrogate endpoint—with the implicit assumption that this would translate directly to improved patient-relevant outcomes like reduced morbidity and mortality. This premise, combined with its economic advantages, propelled HES to become one of the most frequently used colloid solutions for fluid resuscitation worldwide, particularly in Europe.[21] However, this widespread use was established long before the advent of large, rigorous, independent clinical trials designed to assess its long-term safety across diverse and critically ill patient populations—an oversight that would eventually lead to the dramatic reversal of its clinical standing.[6]

II. Physicochemical Properties and Formulations: A Spectrum of Starches

Chemical Structure and Synthesis

Hydroxyethyl Starch is not a single molecular entity but a class of nonionic starch derivatives.[27] The parent molecule is amylopectin, a highly branched polymer of glucose sourced from plants such as waxy corn or potatoes.[23] The basic structure consists of anhydroglucose units linked primarily by

α−(1,4)-glycosidic bonds, with branching chains connected via α−(1,6)-glycosidic links.[24]

To create HES, the native starch is first partially hydrolyzed to reduce its molecular weight, and then it undergoes a chemical modification process known as hydroxyethylation. In this reaction, which typically occurs in the presence of sodium hydroxide, ethylene oxide reacts with the hydroxyl groups on the glucose units of the starch polymer, substituting them with hydroxyethyl groups (−CH2​CH2​OH).[23] This modification is crucial, as it increases the molecule's water solubility and, most importantly, confers resistance to degradation by endogenous

α-amylase, thereby prolonging its intravascular half-life.[32] The general molecular formula is often represented as

(C6​H10​O5​)m​(C2​H5​O)n​.[2]

Key Defining Parameters

The term "HES" encompasses a heterogeneous group of compounds, and different formulations are defined by specific physicochemical characteristics that were once believed to critically influence their pharmacokinetic and safety profiles.[27] The main parameters are:

  • Average Molecular Weight (MW): This refers to the mean molecular weight of the HES polymers in a solution, measured in kilodaltons (kDa). HES products have been historically classified based on their MW:
  • High-MW (Hetastarch): Typically 450 kDa to 670 kDa.[2]
  • Medium-MW (Pentastarch): Around 200-260 kDa.[23]
  • Low-MW (Tetrastarch): Around 130 kDa.[2]
  • Molar Substitution (MS): This is a dimensionless ratio representing the average number of hydroxyethyl groups per glucose unit in the starch polymer.[27] The MS is a primary determinant of the molecule's susceptibility to enzymatic degradation by α-amylase. A higher MS (e.g., 0.7) confers greater resistance to breakdown, leading to a longer intravascular persistence, whereas a lower MS (e.g., 0.4-0.5) results in more rapid metabolism and clearance.[30]
  • C2/C6 Ratio: This ratio describes the preferential site of hydroxyethyl substitution on the glucose ring, comparing substitution at the C2 position to the C6 position. A higher C2/C6 ratio (e.g., ~9:1 for HES 130/0.4) also hinders degradation by α-amylase, further influencing the pharmacokinetic profile of the molecule.[27]

Common Formulations and Synonyms

Based on these parameters, various HES products have been marketed over the years. They are often designated by their MW and MS (e.g., HES 130/0.4). Common synonyms include Hetastarch, Pentastarch, and Tetrastarch, which correspond to high, medium, and low degrees of molar substitution, respectively.[28] Widely known brand names that have featured prominently in clinical research and practice include Hespan, Hextend, Voluven, and Tetraspan.[27]

Physical Properties

In its raw form, HES is a white to off-white, odorless, and tasteless powder.[23] For clinical use, it is formulated as a sterile solution for intravenous infusion, typically at a concentration of 6% or 10%.[27] The solution is usually prepared in an isotonic carrier fluid, such as 0.9% sodium chloride or a balanced electrolyte solution like Ringer's acetate.[27] HES is freely soluble in water but practically insoluble in ethanol.[23]

Table 1: Physicochemical Characteristics of Key HES Formulations

HES ClassificationAverage Molecular Weight (kDa)Molar Substitution (MS)C2/C6 RatioCommon Brand Name(s)Key Clinical Trial(s) where evaluated
Hetastarch450 - 670~0.7-0.75Not specifiedHespan, HextendN/A (Older generation)
Pentastarch200 - 260~0.5Not specifiedPentaspanVISEP
Tetrastarch1300.4 - 0.42~9:1Voluven, Tetraspan6S, CHEST, CRYSTMAS

Table sources: [2]

III. Core Pharmacology: Mechanism, Kinetics, and Dynamics

A. Mechanism of Action

Primary Mechanism: Colloid Osmotic Pressure

The fundamental therapeutic action of Hydroxyethyl Starch is derived from its properties as a colloidal solution.[33] When administered intravenously, the large HES polysaccharide molecules are too large to readily pass through the semipermeable capillary endothelium.[32] Their retention within the intravascular space increases the colloid osmotic pressure (or oncotic pressure) of the plasma.[5] This oncotic gradient opposes the hydrostatic pressure that drives fluid out of the capillaries and actively pulls fluid from the surrounding interstitial and extravascular spaces into the bloodstream.[31] The net result is a rapid and sustained expansion of the circulating plasma volume, which is the intended effect for treating or preventing hypovolemic shock following trauma, surgery, or other causes of acute fluid loss.[2]

Secondary and Postulated Mechanisms

Beyond its primary oncotic effect, other bioactivities for HES have been proposed, forming part of the early rationale for its widespread clinical use. Some studies suggested that HES might reduce capillary leakage, thereby mitigating the "leaky-capillary syndrome" often seen in critical illness.[4] Other purported benefits included anti-inflammatory properties, such as inhibiting the release of inflammatory mediators, and improvements in microcirculatory blood flow.[22]

However, further investigation into the interaction of HES with the vascular endothelium has revealed a more complex and ultimately unfavorable picture. The endothelial surface is coated with a negatively charged layer known as the glycocalyx, which is critical for maintaining vascular barrier integrity, modulating coagulation, and regulating inflammatory responses.[6] While natural colloids like albumin can interact favorably with and help maintain the glycocalyx, HES molecules also possess a net negative charge. This results in electrostatic repulsion from the glycocalyx, preventing HES from contributing to endothelial integrity and potentially disrupting the normal physiological functions of this vital layer.[6]

B. Pharmacokinetics and Pharmacodynamics

The pharmacokinetic profile of HES is complex and highly dependent on the specific formulation's molecular weight and molar substitution, which govern its metabolism and elimination.[27]

Administration and Distribution

HES is administered exclusively by intravenous infusion.[29] Following administration, it primarily distributes within the plasma, consistent with its role as a volume expander. The volume of distribution for HES 130/0.4 is relatively small, approximately 5.9 liters, reflecting its confinement to the intravascular compartment.[28] The pharmacodynamic effect—plasma volume expansion—is maintained for at least 6 hours following a standard 500 mL dose of a 6% solution.[28]

Metabolism

The metabolism of HES is a critical determinant of its duration of action and safety profile. HES polymers are broken down in the plasma by the enzyme α-amylase.[6] This enzyme cleaves the glycosidic bonds within the polysaccharide chain, degrading larger molecules into smaller fragments.[30] The rate of this metabolism is inversely proportional to the degree of hydroxyethylation; higher molar substitution (MS) and a higher C2/C6 ratio protect the molecule from amylase activity, leading to slower degradation and a longer intravascular half-life.[29]

Elimination

Elimination of HES occurs primarily via the kidneys.[6] The process is size-dependent. HES molecules and fragments smaller than the renal filtration threshold (approximately 60-70 kDa) are readily excreted in the urine.[6] Larger molecules must first be metabolized by plasma

α-amylase into smaller fragments before they can be renally cleared.[6] For HES 130/0.4, approximately 62-70% of an administered dose is excreted in the urine within 72 hours.[28] However, a notable portion of the drug is not immediately accounted for by urinary excretion, leading to concerns about long-term tissue accumulation, particularly with higher-MW and higher-MS formulations.[6]

Half-Life and Duration of Effect

The plasma half-life of HES is biphasic and formulation-dependent. For the widely studied HES 130/0.4 formulation, the elimination half-life is approximately 12 hours.[28] Plasma concentrations decrease rapidly at first, remaining at 75% of peak concentration 30 minutes post-infusion, but then decline more slowly, falling to 14% at 6 hours and returning to baseline by 24 hours.[43] Despite the relatively short plasma half-life of the bulk material, the issue of tissue storage means that the biological effects, particularly adverse ones, can persist for much longer.[27]

Impact of Renal Impairment

The pharmacokinetic profile of HES is dramatically altered in patients with renal dysfunction. In patients with a creatinine clearance below 50 mL/min, the clearance of HES 130/0.4 is reduced by 42%, and the area under the curve (AUC) is increased by 73% compared to patients with normal renal function.[38] This impaired elimination leads to drug accumulation, prolonging exposure to high concentrations of HES and significantly increasing the risk of dose-dependent toxicities, most notably further kidney injury.[38]

Table 2: Summary of Key Pharmacokinetic Parameters for HES 130/0.4

ParameterValue / DescriptionSource(s)
AdministrationIntravenous infusion only40
Peak Concentration (Cmax​)4.34 mg/mL28
Volume of Distribution (Vd​)5.9 L28
Metabolism PathwayEnzymatic degradation by plasma α-amylase28
Route of EliminationPrimarily renal excretion6
Elimination Half-Life (t1/2​)12 hours28
Clearance31.4 mL/min28
Effect of Renal ImpairmentClearance decreased by 42%; AUC increased by 73% (in Clcr <50 mL/min)38

IV. The Turning Point: Evidence from Landmark Randomized Controlled Trials

The trajectory of Hydroxyethyl Starch in clinical medicine was irrevocably altered by a series of large, high-quality randomized controlled trials (RCTs) conducted in the late 2000s and early 2010s. These studies systematically investigated the safety and efficacy of HES in critically ill patients and, in doing so, provided irrefutable evidence of significant harm that ultimately led to its clinical and regulatory downfall. The narrative progression across these trials is particularly powerful, as it demonstrates the scientific process of hypothesis testing and falsification in action.

A. The VISEP Trial: Early Warnings with High-Molecular-Weight HES

The VISEP (Efficacy of Volume Substitution and Insulin Therapy in Severe Sepsis) trial, published in 2008, was a multicenter, 2x2 factorial study that served as one of the first major warnings about the dangers of HES in critical illness.[37] One arm of the trial randomized patients with severe sepsis to receive fluid resuscitation with either 10% pentastarch (HES 200/0.5), a medium-to-high molecular weight formulation, or modified Ringer's lactate, a crystalloid.[21]

The trial was stopped prematurely for safety reasons after enrolling 537 evaluable patients.[37] The results were stark: HES therapy was associated with significantly higher rates of acute renal failure and a greater need for renal-replacement therapy compared to Ringer's lactate. The authors concluded unequivocally that HES, as used in the study, was harmful and that its toxicity appeared to be dose-dependent.[37] VISEP provided the first robust, high-quality evidence from a large RCT that directly linked HES administration to nephrotoxicity in the septic patient population, challenging the prevailing assumptions about its safety profile.

B. The 6S Trial: Definitive Harm with "Modern" HES in Severe Sepsis

Following the VISEP trial, a prevailing hypothesis emerged in the medical community: perhaps the harm observed was specific to the older, higher-molecular-weight HES formulations, and that newer, "modern" tetrastarches with lower molecular weight and molar substitution would be safer. The 6S (Scandinavian Starch for Severe Sepsis/Septic Shock) trial, published in 2012, was designed to test this very hypothesis.[39]

This investigator-initiated, blinded, multicenter RCT randomized 800 patients with severe sepsis to receive fluid resuscitation with either 6% HES 130/0.42 (Tetraspan), a modern tetrastarch, or Ringer's acetate, a balanced crystalloid.[39] The findings were devastating for the "safer formulation" hypothesis.

  • Primary Outcome: The composite primary outcome of death or end-stage kidney failure (dependence on dialysis) at 90 days was significantly higher in the HES group (51%) compared to the Ringer's acetate group (43%), with a relative risk (RR) of 1.17 (95% CI 1.01-1.36; p=0.03). This difference was driven almost entirely by a higher mortality rate in the HES arm, as only one patient in each group was dialysis-dependent at 90 days.[39]
  • Secondary Outcomes: The use of renal replacement therapy at any point during the trial was significantly more frequent in the HES group (22% vs. 16%; RR 1.35, 95% CI 1.01-1.80; p=0.04). There was also a non-significant trend toward more severe bleeding in the HES group (10% vs. 6%).[39]

The 6S trial was a watershed moment. It demonstrated conclusively that even the supposedly safer, modern tetrastarch formulation was not only devoid of benefit but was actively harmful, causing a statistically significant increase in mortality when used for fluid resuscitation in patients with severe sepsis.[39]

C. The CHEST Trial: Corroborating Harm in a General ICU Population

Published concurrently with the 6S trial, the CHEST (Crystalloid versus Hydroxyethyl Starch Trial) was the largest of the three landmark studies, providing a broader perspective on the safety of modern HES in a general intensive care unit (ICU) population.[41] This multicenter, double-blind RCT randomized 7000 heterogeneous ICU patients (including those with sepsis, trauma, and post-operative needs) to receive either 6% HES 130/0.4 (Voluven) in saline or 0.9% saline alone for all fluid resuscitation.[41]

  • Primary Outcome: The trial found no significant difference in the primary outcome of 90-day mortality between the two groups. The mortality rate was 18.0% in the HES group and 17.0% in the saline group (RR 1.06; 95% CI 0.96-1.18; p=0.26).[41]
  • Secondary Outcomes: Despite the neutral mortality finding, the signal of renal harm was once again clear and statistically significant. The use of renal replacement therapy was significantly higher in the HES group (7.0%) compared to the saline group (5.8%), with an RR of 1.21 (95% CI 1.00-1.45; p=0.04). Furthermore, the HES group had a higher overall rate of adverse events (5.3% vs. 2.8%), required more blood product transfusions, and experienced more pruritus.[26]

The CHEST trial powerfully corroborated the findings of 6S. While it did not show a mortality difference in its broader, less severely ill population, it confirmed the consistent signal of renal toxicity and demonstrated a complete lack of any patient-relevant clinical benefit to justify this risk. The increased need for RRT and blood products also effectively dismantled any remaining arguments for the cost-effectiveness of HES.

The convergence of evidence from VISEP, 6S, and CHEST created an irrefutable scientific narrative. The initial alarms raised by VISEP with older HES formulations were not only confirmed but amplified by 6S and CHEST, which showed that the newer, supposedly safer formulations carried the same, if not worse, risk profile in critically ill patients. This progression demonstrated that the toxicity was likely a class effect of synthetic starches, not an issue with a single formulation, and provided the definitive evidence base that would compel regulators worldwide to act.

Table 3: Summary of Landmark Clinical Trials (VISEP, 6S, CHEST)

Trial Acronym (Year)Patient PopulationHES FormulationComparatorNPrimary OutcomeKey Secondary OutcomesAuthor's Conclusion
VISEP (2008)Severe Sepsis10% Pentastarch (HES 200/0.5)Ringer's Lactate53728-day mortality (no difference)Increased rate of acute renal failure and need for RRTHES was harmful, and its toxicity increased with accumulating doses.
6S (2012)Severe Sepsis6% Tetrastarch (HES 130/0.42)Ringer's Acetate800Increased risk of death or dialysis dependence at 90 days (RR 1.17)Increased need for RRT (RR 1.35); Trend toward more severe bleedingHES increased the risk of death at 90 days and the need for RRT.
CHEST (2012)General ICU6% Tetrastarch (HES 130/0.4)0.9% Saline7000No difference in 90-day mortalityIncreased need for RRT (RR 1.21); More adverse events; Increased need for blood productsNo clinical benefit with HES, but an increased rate of RRT.

Table sources: [37]

V. The Spectrum of Harm: A Profile of Adverse Effects

The evidence from landmark trials and subsequent meta-analyses has illuminated a consistent and multifaceted pattern of harm associated with Hydroxyethyl Starch administration, particularly in vulnerable patient populations. The adverse effects span multiple organ systems, with the most severe being nephrotoxicity, coagulopathy, and increased mortality.

A. Acute Kidney Injury and Nephrotoxicity

The most consistently demonstrated and clinically significant adverse effect of HES is acute kidney injury (AKI).[27] The VISEP, 6S, and CHEST trials all reported a statistically significant increase in the need for renal replacement therapy (RRT) in patients receiving HES compared to those receiving crystalloids.[27] This risk has been confirmed in numerous meta-analyses and observational studies, leading regulatory agencies to issue strong warnings about its nephrotoxic potential.[26] The risk appears highest in critically ill patients, especially those with sepsis, but has also been observed in surgical and trauma patients.[27]

The underlying pathophysiology of HES-induced nephrotoxicity is thought to be multifactorial. A primary mechanism involves the renal handling of HES molecules. After filtration by the glomerulus, HES polymers are taken up by proximal tubular epithelial cells via pinocytosis. The accumulation of these large, osmotically active molecules within the lysosomes of these cells can lead to swelling and vacuolization, a condition described as osmotic nephrosis-like lesions.[6] This intracellular storage is believed to impair tubular cell function and viability, ultimately precipitating acute tubular necrosis and renal failure.[6] The long-term deposition of HES in renal tissue may explain why the need for RRT has been reported up to 90 days after administration.[27]

B. HES-Induced Coagulopathy and Bleeding

Another major safety concern is the detrimental effect of HES on the coagulation system, leading to an increased risk of bleeding.[27] Clinical trials have documented that HES administration is associated with a higher need for blood product transfusions and an increased incidence of bleeding events, particularly in surgical settings like cardiopulmonary bypass.[26] The mechanisms behind this HES-induced coagulopathy are complex and extend beyond simple hemodilution.[30] Key contributing factors include:

  1. Acquired von Willebrand Syndrome: HES has a direct, non-dilutional effect on the von Willebrand factor (vWF)-Factor VIII complex. It has been shown to decrease the plasma concentration and activity of both Factor VIII and vWF, likely through mechanisms such as precipitation or accelerated clearance of the complex.[23] This acquired deficiency impairs primary hemostasis and prolongs the activated partial thromboplastin time (aPTT).[27]
  2. Platelet Dysfunction: HES molecules can physically coat the surface of platelets. This coating interferes with platelet function by reducing the availability of critical surface receptors, most notably glycoprotein IIb-IIIa, which is essential for platelet aggregation and adhesion to fibrinogen.[27] This leads to impaired platelet plug formation.
  3. Impaired Fibrin Polymerization: Beyond its effects on coagulation factors and platelets, HES has been shown to directly interfere with the final stage of clot formation. The presence of HES macromolecules impairs the polymerization of fibrin monomers into a stable, cross-linked fibrin clot, resulting in a weaker and less effective clot structure.[30]
  4. Dilutional Coagulopathy: In addition to these specific mechanisms, the infusion of large volumes of any colloid will cause a dilutional reduction in the concentration of all coagulation factors and platelets, further contributing to the overall coagulopathic state.[27]

C. Increased Mortality in Critical Illness

The most severe consequence documented with HES use is an increased risk of death in certain patient populations. The 6S trial provided the most definitive evidence, demonstrating a statistically significant increase in 90-day mortality in patients with severe sepsis treated with HES 130/0.42 compared to Ringer's acetate.[39] This finding of increased mortality in critically ill and septic patients has been supported by numerous meta-analyses and observational studies, forming the primary basis for the strong contraindications issued by regulatory bodies worldwide.[26] The increased mortality is likely a downstream consequence of the constellation of harms caused by HES, including renal failure and bleeding complications.

D. Anaphylactoid Reactions and Tissue Deposition

While less common than nephrotoxicity or coagulopathy, HES can induce other significant adverse effects.

  • Anaphylactoid Reactions: HES administration carries a risk of hypersensitivity reactions. These can range in severity from mild, influenza-like symptoms, urticaria, and pruritus to life-threatening anaphylactoid reactions involving bronchospasm, cardiovascular collapse, and non-cardiogenic pulmonary edema.[4] The incidence is estimated to be low (e.g., 0.08% in one study) but requires vigilant monitoring during infusion.[63]
  • Pruritus and Tissue Storage: A frequent and often distressing side effect of HES is severe, refractory itching (pruritus).[27] This symptom is not an acute allergic reaction but rather a consequence of the long-term accumulation and deposition of HES molecules in various body tissues, particularly the skin and peripheral nerves.[26] These deposits can persist for months or even years after the last administration of the drug, leading to chronic and difficult-to-treat pruritus.[27] The onset is often delayed by weeks, making the causal link to HES difficult to identify.[27]

Table 4: Major Adverse Effects of HES and Postulated Mechanisms

Adverse EffectClinical ManifestationPostulated Pathophysiological Mechanism
Acute Kidney Injury (AKI)Increased serum creatinine, oliguria, need for renal replacement therapy (RRT)Accumulation of HES molecules in renal tubules causing osmotic nephrosis-like lesions and tubular cell injury.
CoagulopathyIncreased bleeding, prolonged aPTT, increased need for blood product transfusionsAcquired von Willebrand syndrome (decreased FVIII/vWF); platelet dysfunction (impaired GP IIb-IIIa availability); impaired fibrin polymerization; hemodilution.
Increased MortalityHigher death rates in critically ill and septic patientsDownstream consequence of AKI, coagulopathy, and other systemic toxicities.
Anaphylactoid ReactionUrticaria, bronchospasm, hypotension, shockNon-IgE mediated mast cell and basophil degranulation (histamine release).
Pruritus (Itching)Severe, refractory itching, often with delayed onsetLong-term deposition and storage of HES molecules in skin and peripheral nerve tissues.

Table sources: [6]

VI. The Regulatory Reckoning: Global Agency Actions and Warnings

The overwhelming and consistent evidence of harm from high-quality clinical trials precipitated a cascade of decisive regulatory actions from leading health authorities around the world. These actions, escalating over a decade, effectively dismantled the clinical role of Hydroxyethyl Starch and serve as a powerful example of evidence-based pharmacovigilance.

A. U.S. Food and Drug Administration (FDA) Response

The FDA's response to the mounting safety concerns was progressive and definitive, culminating in severe restrictions on the use of HES products.

  • The Black Box Warning (2013): In June 2013, following a comprehensive review of the evidence from trials like 6S and CHEST, the FDA mandated the addition of a Black Box Warning—its most stringent safety alert—to the prescribing information for all HES products.[27] This warning explicitly highlighted the increased risk of mortality and severe renal injury. The key recommendations included [27]:
  • Do not use HES solutions in critically ill adult patients, including those with sepsis.
  • Avoid use in patients with pre-existing renal dysfunction.
  • Discontinue use at the first sign of renal injury.
  • Monitor renal function for at least 90 days in all hospitalized patients receiving HES, as the need for RRT has been reported long after administration.
  • Avoid use in patients undergoing open heart surgery in association with cardiopulmonary bypass due to the risk of excess bleeding.
  • Discontinue use at the first sign of coagulopathy.
  • 2021 Labeling Changes and Market Impact: The regulatory pressure continued to mount. In July 2021, the FDA required further safety labeling changes, expanding the warnings to include the risk of mortality and excess bleeding in surgical and blunt trauma patients.[27] Crucially, the new labeling included a statement that HES products should not be used unless adequate alternative treatment is unavailable.[55] This effectively relegated HES to a drug of last resort. The increasingly restrictive environment and liability concerns led the major manufacturers of HES to de-register their global brands for the U.S. market, significantly curtailing its availability.[27]

B. European Medicines Agency (EMA) and European Response

The response in Europe followed a similar, and in some ways more aggressive, trajectory, led by the EMA's Pharmacovigilance Risk Assessment Committee (PRAC).

  • Initial Restrictions (2013): In October 2013, following the PRAC's recommendation, European authorities implemented major restrictions on HES use.[57] HES was formally contraindicated in several high-risk populations, including patients with sepsis, burn injuries, critically ill patients (typically in the ICU), and those with any degree of renal impairment or on RRT.[27] Its use was narrowed to the treatment of hypovolemia from acute blood loss only when crystalloids were considered insufficient, and even then, with strict monitoring requirements.[57]
  • Escalation to Market Suspension Recommendation: Despite these stringent restrictions, subsequent drug utilization studies conducted in Europe revealed that HES was still being used "off-label" in contraindicated populations, exposing patients to ongoing and unnecessary risk.[72] This failure of risk minimization measures to ensure safe use led the PRAC to conclude that the risks of HES solutions continued to outweigh their benefits. Consequently, in 2018, the PRAC recommended the suspension of the marketing authorizations for all HES solutions for infusion across the European Union.[66] This recommendation was reaffirmed in 2022, leading the European Commission to confirm the suspension, effectively removing HES from the EU market, with provisions for member states to delay the suspension for up to 18 months only if deemed necessary for public health reasons.[27]

Table 5: Timeline of Key Regulatory Actions by the FDA and EMA

Date (Year)Regulatory BodyActionKey Patient Populations Affected
June 2013FDAIssues Black Box WarningCritically ill adults, sepsis patients, patients with renal dysfunction
October 2013EMAImplements major restrictions and contraindicationsCritically ill, sepsis, burn, and renal impairment patients
January 2018EMA (PRAC)Recommends suspension of marketing authorizationsAll patient populations
July 2021FDARequires further labeling changes; limits use to last resortSurgical and blunt trauma patients added to warnings
May 2022European CommissionConfirms suspension of marketing authorizationsAll patient populations in the EU

Table sources: [27]

VII. HES in Clinical Context: A Comparative and Economic Analysis

The downfall of Hydroxyethyl Starch can only be fully understood by placing it in the context of its therapeutic alternatives. The robust evidence of its harm, coupled with a re-evaluation of its benefits and costs, has led to a fundamental shift in clinical practice guidelines and a reordering of the fluid resuscitation armamentarium.

A. HES vs. Crystalloids: The New Standard of Care

The central debate in fluid resuscitation has largely been resolved in favor of crystalloids as the first-line therapy for most patients. While HES solutions are more efficient at expanding plasma volume on a per-milliliter basis, this "volume-sparing" effect has been shown to be a surrogate endpoint with no translation to improved patient-relevant outcomes.[7] The evidence from the 6S and CHEST trials, along with numerous meta-analyses, demonstrated that this physiological advantage is completely overshadowed by the significant risks of mortality, AKI, and bleeding.[7]

As a result, major international clinical practice guidelines have moved decisively away from HES. The Surviving Sepsis Campaign, a global authority on sepsis management, now provides a strong recommendation for using crystalloid fluids as the initial fluid of choice for resuscitation in patients with sepsis and septic shock.[74] The guidelines explicitly recommend against the use of HES solutions in this population due to the clear evidence of harm.[77] This shift establishes crystalloids as the undisputed standard of care for initial volume replacement in the critically ill.

B. HES vs. Albumin: Synthetic vs. Natural Colloid

When a colloid is considered necessary—for instance, in patients who require substantial volumes of crystalloids—the choice is now clearly in favor of human albumin over synthetic colloids like HES.[7] Systematic reviews and meta-analyses comparing HES directly to albumin have consistently shown that HES is associated with worse outcomes. Specifically, in patients with sepsis, HES use led to an increased need for RRT and red blood cell transfusions compared to albumin, with no mortality benefit.[54] In cardiac surgery, HES increased the risk of postoperative bleeding and reoperation compared to albumin.[59] While albumin is a more expensive natural colloid, its superior safety profile makes it the preferred colloid agent when one is indicated.[7]

C. Cost-Effectiveness Analysis: The Fallacy of Acquisition Cost

For many years, a primary justification for the use of HES was its lower direct acquisition cost compared to human albumin.[7] However, this narrow view of cost has been thoroughly discredited by a more comprehensive understanding of the total cost of care. A true cost-effectiveness analysis must account for the downstream economic consequences of the iatrogenic complications caused by a therapy.

The use of HES is associated with significant additional healthcare expenditures. The increased incidence of AKI necessitates the use of renal replacement therapy, which is an extremely costly intervention.[25] Similarly, the HES-induced coagulopathy leads to a greater need for transfusions of blood products, which also carry substantial costs.[26] When these downstream costs are factored into the economic equation, any initial savings from the lower unit price of HES are quickly erased and likely reversed. Decision-analysis models have concluded that when total medical costs and iatrogenic morbidities are considered, albumin is a more cost-effective treatment than HES in septic patients.[25] This complete inversion of the cost argument underscores the principle that a therapy that causes harm cannot be considered cost-effective, regardless of its initial price tag.

The initial perception of HES was that of a high-benefit, acceptable-risk, and low-cost agent. The cumulative evidence has systematically inverted this risk-benefit-cost equation. The benefit is now understood to be limited to a surrogate endpoint with no improvement in patient outcomes. The risk is unacceptably high, encompassing mortality, renal failure, and bleeding. The cost, when properly accounting for the management of these complications, is also high. This dramatic reframing of a drug's value proposition illustrates the power of rigorous clinical science to supplant flawed assumptions and guide rational therapeutic decision-making.

Table 6: Comparative Profile of Fluid Resuscitation Agents

AttributeIsotonic Crystalloids (e.g., Saline, Lactated Ringer's)Albumin (5% and 25%)Hydroxyethyl Starch (HES)
ClassCrystalloidNatural ColloidSynthetic Colloid
MechanismVolume expansion via distribution throughout extracellular fluidOncotic pressure from large protein moleculesOncotic pressure from large polysaccharide molecules
Key AdvantagesInexpensive, readily available, safe, long shelf lifeGood safety profile, remains intravascular, may have other beneficial properties (e.g., antioxidant)Efficient volume expansion (volume-sparing), cheaper than albumin
Key Disadvantages/RisksRequires large volumes, potential for edema, electrolyte disturbances (e.g., hyperchloremic acidosis with saline)Expensive, derived from human plasma (theoretical risk of pathogen transmission)Increased mortality, acute kidney injury, coagulopathy, bleeding, pruritus, anaphylactoid reactions
Place in TherapyFirst-line agent for most fluid resuscitation (e.g., sepsis)Second-line agent, considered when large volumes of crystalloid are neededContraindicated in critically ill, sepsis, burns, renal impairment; use severely restricted to last resort
Relative CostLowHighModerate (unit cost), High (total cost of care)

Table sources: [7]

VIII. Conclusion: The Legacy of Hydroxyethyl Starch

The clinical and regulatory history of Hydroxyethyl Starch represents a compelling and cautionary narrative in modern medicine. HES ascended to become one of the world's most frequently used plasma volume expanders based on a sound physiological principle—its ability to efficiently expand intravascular volume through colloid osmosis. For decades, it was considered a cost-effective and readily available alternative to natural colloids like albumin for the treatment of hypovolemia in acute care settings.

However, this initial promise was built on a foundation that prioritized surrogate physiological endpoints over hard, patient-relevant outcomes. The subsequent emergence of high-quality, independent, large-scale randomized controlled trials, notably VISEP, 6S, and CHEST, systematically and irrefutably dismantled the safety profile of HES. This body of evidence demonstrated with remarkable consistency that HES administration, across various formulations and patient populations, was associated with a clear pattern of harm. The risks were not trivial; they included a statistically significant increase in the incidence of acute kidney injury requiring renal replacement therapy, a clinically relevant coagulopathy leading to increased bleeding and transfusion needs, and, most alarmingly, an increased risk of death in critically ill and septic patients.

The weight of this evidence proved overwhelming, leaving no doubt that for the vast majority of its intended indications, the risks of HES therapy far outweighed any potential benefits. In response, regulatory agencies worldwide, including the U.S. FDA and the European EMA, took decisive action, issuing stringent warnings, contraindications, and ultimately, recommending the suspension of its marketing authorizations.

Consequently, the current place of Hydroxyethyl Starch in clinical therapy is, and should be, exceptionally limited. Based on the global scientific and regulatory consensus, HES has no role in the routine fluid resuscitation of critically ill patients, particularly those with sepsis, burns, trauma, or any pre-existing renal dysfunction. Its use has been correctly superseded by crystalloid solutions as the first-line agent of choice, with human albumin reserved for specific indications where a colloid is deemed necessary.

The legacy of Hydroxyethyl Starch is therefore twofold. It is a story of a therapeutic class that, despite its logical premise, was ultimately found to cause significant iatrogenic harm. More importantly, it serves as a seminal case study in pharmacovigilance and evidence-based medicine. The HES saga highlights the profound dangers of relying on surrogate endpoints for clinical decision-making, the critical importance of large-scale, pragmatic, post-marketing safety trials to evaluate real-world outcomes, and the power of independent, academic-led research to challenge established dogma and protect public health. The crucial lessons learned from the rise and fall of Hydroxyethyl Starch have fundamentally improved our understanding of fluid resuscitation and reinforced the principle that patient safety must always be the paramount consideration in therapeutic innovation.

Works cited

  1. [Hydroxyethyl starch solutions] - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/15947530/
  2. Hydroxyethyl starch - 200/0.5 | 9005-27-0 | FH29681 - Biosynth, accessed August 1, 2025, https://www.biosynth.com/p/FH29681/9005-27-0-hydroxyethyl-starch-20005
  3. go.drugbank.com, accessed August 1, 2025, https://go.drugbank.com/drugs/DB09106#:~:text=Pharmacology&text=An%20intravenous%20solution%20of%20hydroxyethyl,%2C%20surgery%2C%20or%20other%20issues.
  4. Hydroxyethyl-starch - Mechanism, Indication, Contraindications, Dosing, Adverse Effect, Interaction, Hepatic Dose | Drug Index | Pediatric Oncall, accessed August 1, 2025, https://www.pediatriconcall.com/drugs/hydroxyethyl-starch/651
  5. What is Hydroxyethyl Starch used for? - Patsnap Synapse, accessed August 1, 2025, https://synapse.patsnap.com/article/what-is-hydroxyethyl-starch-used-for
  6. Clinical use of hydroxyethyl starch and serious adverse effects: Need for awareness amongst the medical fraternity, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4213895/
  7. Colloids vs. crystalloids as resuscitation fluids - Deranged Physiology, accessed August 1, 2025, https://derangedphysiology.com/main/required-reading/electrolyte-disorders/Chapter-225/colloids-vs-crystalloids-resuscitation-fluids
  8. Colloids versus crystalloids for fluid resuscitation in critically ill people - PubMed Central, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6513027/
  9. Colloids versus crystalloids for fluid resuscitation in critically ill people - Lewis, SR - 2018 | Cochrane Library, accessed August 1, 2025, https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD000567.pub7/full
  10. Colloid fluids (Chapter 3) - Clinical Fluid Therapy in the Perioperative Setting, accessed August 1, 2025, https://www.cambridge.org/core/books/clinical-fluid-therapy-in-the-perioperative-setting/colloid-fluids/CD9761CE3D7359DDAE3A5EA38B932009
  11. Crystalloid and Colloid Compositions and Their Impact - Frontiers, accessed August 1, 2025, https://www.frontiersin.org/journals/veterinary-science/articles/10.3389/fvets.2021.639848/full
  12. Colloids versus crystalloids for fluid resuscitation in critically ill patients (Review) - LSHTM Research Online, accessed August 1, 2025, https://researchonline.lshtm.ac.uk/id/eprint/8509/1/8509.pdf
  13. Colloids versus crystalloids for fluid resuscitation in critically ill patients (Cochrane Review) | Request PDF - ResearchGate, accessed August 1, 2025, https://www.researchgate.net/publication/12520124_Colloids_versus_crystalloids_for_fluid_resuscitation_in_critically_ill_patients_Cochrane_Review
  14. Colloids versus crystalloids for fluid resuscitation in critically ill people - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/30073665/
  15. Colloids versus crystalloids for fluid resuscitation in critically ill patients - Perel, P - 2011, accessed August 1, 2025, https://www.cochranelibrary.com/doi/10.1002/14651858.CD000567.pub4/abstract
  16. Colloids versus crystalloids for fluid resuscitation in critically ill patients - Perel, P - 2013, accessed August 1, 2025, https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD000567.pub6/abstract/en
  17. Colloids versus crystalloids for fluid resuscitation in critically ill patients - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/23450531/
  18. Colloids versus crystalloids for fluid resuscitation in critically ill patients (Review) - LSHTM Research Online, accessed August 1, 2025, https://researchonline.lshtm.ac.uk/id/eprint/21092/1/Perel_et_al-2012-The_Cochrane_library.pdf
  19. Crystalloid Fluids - StatPearls - NCBI Bookshelf, accessed August 1, 2025, https://www.ncbi.nlm.nih.gov/books/NBK537326/
  20. Section 7: Questions and Controversies in Fluid Therapy - AAHA, accessed August 1, 2025, https://www.aaha.org/resources/2024-aaha-fluid-therapy-guidelines-for-dogs-and-cats/section-7-questions-and-controversies-in-fluid-therapy/
  21. Crystalloids and colloids in critical patient resuscitation - Medicina Intensiva, accessed August 1, 2025, https://www.medintensiva.org/en-crystalloids-colloids-in-critical-patient-articulo-S2173572715000351
  22. The Safety of Hydroxyethyl Starch (HES) - Products Now in Doubt, accessed August 1, 2025, https://www.apsf.org/article/the-safety-of-hydroxyethyl-starch-hes-products-now-in-doubt/
  23. Hydroxyethyl starch | 9005-27-0 - ChemicalBook, accessed August 1, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB6376714.htm
  24. Structural Aspect of Hydroxyethyl-Starch–Anticancer-Drug-Conjugates as State-of-the-Art Drug Carriers - MDPI, accessed August 1, 2025, https://www.mdpi.com/2218-0532/91/3/32
  25. Choice of Fluids in Severe Septic Patients - A Cost-effectiveness Analysis Informed by Recent Clinical Trials, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4112378/
  26. Fluid Replacement With Hydroxyethyl Starch in Critical Care—a Reassessment - PMC, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3719450/
  27. Hydroxyethyl starch - Wikipedia, accessed August 1, 2025, https://en.wikipedia.org/wiki/Hydroxyethyl_starch
  28. Hydroxyethyl Starch: Uses, Interactions, Mechanism of Action ..., accessed August 1, 2025, https://go.drugbank.com/drugs/DB09106
  29. Hydroxyethyl Starch in Sodium Chloride: Package Insert / Prescribing Info - Drugs.com, accessed August 1, 2025, https://www.drugs.com/pro/hydroxyethyl-starch-in-sodium-chloride.html
  30. Effects of Hydroxyethyl Starch Solutions on ... - Medic's Corner, accessed August 1, 2025, http://www.jvsmedicscorner.com/ICU-Blood_Transfusions_files/Effects%20of%20Hydroxyethyl%20Starch%20on%20coagulation%20review.pdf
  31. CAS 9005-27-0 Hydroxyethyl starch - BOC Sciences, accessed August 1, 2025, https://www.bocsci.com/product/hydroxyethyl-starch-cas-9005-27-0-79244.html
  32. What is the mechanism of Hydroxyethyl Starch? - Patsnap Synapse, accessed August 1, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-hydroxyethyl-starch
  33. www.mims.com, accessed August 1, 2025, https://www.mims.com/philippines/drug/info/hydroxyethyl-starch#:~:text=Mechanism%20of%20Action%3A%20Hydroxyethyl%20starch,%CE%B1%2Damylase%20in%20the%20body.
  34. Hydroxyethyl starch - 130/0.4 | 9005-27-0 | FH35059 - Biosynth, accessed August 1, 2025, https://www.biosynth.com/p/FH35059/9005-27-0-hydroxyethyl-starch-13004
  35. Coagulopathy Induced by Hydroxyethyl Starch - Portal de Periódicos da CAPES, accessed August 1, 2025, https://www.periodicos.capes.gov.br/index.php/acervo/buscador.html?task=detalhes&id=W2066488022
  36. Pharmacokinetics of Hydroxyethyl Starch (70/0.5) following Repeated Infusions, accessed August 1, 2025, https://karger.com/tmh/article-pdf/30/2/72/3568118/000070547.pdf
  37. Intensive insulin therapy and pentastarch resuscitation in severe ..., accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/18184958/
  38. Pharmacokinetics of hydroxyethyl starch - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/15966753/
  39. 6S – The Bottom Line, accessed August 1, 2025, https://www.thebottomline.org.uk/summaries/icm/6s/
  40. VOLUVEN - hydroxyethyl starch 130/0.4 injection, solution, accessed August 1, 2025, https://labeling.pfizer.com/ShowLabeling.aspx?id=7382
  41. CHEST – The Bottom Line, accessed August 1, 2025, https://www.thebottomline.org.uk/summaries/icm/chest/
  42. Evidence-based Colloid Use in the Critically Ill | American Thoracic Society Consensus Statement | American Journal of Respiratory and Critical Care Medicine, accessed August 1, 2025, https://www.atsjournals.org/doi/full/10.1164/rccm.200208-909ST
  43. Hydroxyethyl Starch 130/0.4 Monograph for Professionals - Drugs.com, accessed August 1, 2025, https://www.drugs.com/monograph/hydroxyethyl-starch-130-0-4.html
  44. What are the side effects of Hydroxyethyl Starch? - Patsnap Synapse, accessed August 1, 2025, https://synapse-patsnap-com.libproxy1.nus.edu.sg/article/what-are-the-side-effects-of-hydroxyethyl-starch
  45. Comparing the effect of hydroxyethyl starch 130/0.4 with balanced crystalloid solution on mortality and kidney failure in patients with severe sepsis (6S - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/21269526/
  46. Hydroxyethyl starch 130/0.42 versus Ringer's acetate in severe sepsis - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/22738085/
  47. 6S Trial | ICU REACH, accessed August 1, 2025, https://www.icureach.com/criticalcaretrials/6s-trial
  48. Hydroxyethyl starch 130/0.38-0.45 versus crystalloid or albumin in patients with sepsis: systematic review with meta-analysis and trial sequential analysis | The BMJ, accessed August 1, 2025, https://www.bmj.com/content/346/bmj.f839
  49. CHEST Trial | ICU REACH, accessed August 1, 2025, https://www.icureach.com/criticalcaretrials/chest-trial
  50. Hydroxyethyl starch or saline for fluid resuscitation in intensive care - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/23075127/
  51. Hydroxyethyl Starch or Saline for Fluid Resuscitation in Intensive Care - Oregon State University Libraries and Press, accessed August 1, 2025, http://search.library.oregonstate.edu/discovery/fulldisplay?docid=cdi_unpaywall_primary_10_1056_nejmoa1209759&context=PC&vid=01ALLIANCE_OSU:OSU&lang=en&search_scope=OSU_Everything_Profile&adaptor=Primo%20Central&tab=Everything&query=creator%2Cequals%2C%20van%20Haren%2C%20Frank%20%2CAND&mode=advanced&offset=0
  52. Study Details | Crystalloid Versus Hydroxyethyl Starch Trials | ClinicalTrials.gov, accessed August 1, 2025, https://clinicaltrials.gov/study/NCT00935168
  53. Hydroxyethyl starch solutions (Voluven, Volulyte 6%) associated with increased risk of mortality and renal impairment - Medsafe, accessed August 1, 2025, https://www.medsafe.govt.nz/safety/ews/2013/hydroxyethylstarch-mortality.asp
  54. (PDF) Hydroxyethyl starch 130/0.38-0.45 versus crystalloid or albumin in patients with sepsis: Systematic review with meta-analysis and trial sequential analysis - ResearchGate, accessed August 1, 2025, https://www.researchgate.net/publication/235650415_Hydroxyethyl_starch_130038-045_versus_crystalloid_or_albumin_in_patients_with_sepsis_Systematic_review_with_meta-analysis_and_trial_sequential_analysis
  55. Labeling Changes on mortality, kidney injury, and excess bleeding with hydroxyethyl starch products | FDA, accessed August 1, 2025, https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/labeling-changes-mortality-kidney-injury-and-excess-bleeding-hydroxyethyl-starch-products
  56. US Food and Drug Administration adds black boxed warning to Hydroxyethyl starch (HES), accessed August 1, 2025, https://www.apheresis.org/news/269049/US-Food-and-Drug-Administration-adds-black-boxed-warning-to-Hydroxyethyl-starch-HES.htm
  57. Hydroxyethyl starch solutions for infusion - referral | European Medicines Agency (EMA), accessed August 1, 2025, https://www.ema.europa.eu/en/medicines/human/referrals/hydroxyethyl-starch-solutions-infusion-0
  58. 1 HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights do not include all the information needed to use Voluven® safely and - FDA, accessed August 1, 2025, https://www.fda.gov/media/72764/download
  59. Hydroxyethyl starch for perioperative fluid management: a critical appraisal - SciELO, accessed August 1, 2025, https://www.scielo.br/j/bja/a/r4TcQMMhTTgFBn3MKTLSLxg/?lang=en
  60. Mechanisms of hydroxyethyl starch-induced dilutional coagulopathy - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/19422451/
  61. CONTRA: Hydroxyethyl starch solutions are unsafe in critically ill patients - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/19533093/
  62. Hydroxyethyl starch - CMDh position Annex III - EMA, accessed August 1, 2025, https://www.ema.europa.eu/en/documents/referral/hydroxyethyl-starch-article-107i-procedure-annex-iii_en.pdf
  63. Anaphylactoid Reactions Due to Hydroxyethyl Starch Infusion - Karger Publishers, accessed August 1, 2025, https://karger.com/esr/article-pdf/8/5/389/2741616/000127883.pdf
  64. Anaphylactic reaction with hydroxyethyl starch during anesthesia - A case report - - PMC, accessed August 1, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7713807/
  65. Anaphylactic reaction with hydroxyethyl starch during anesthesia: A case report - KoreaMed, accessed August 1, 2025, https://www.koreamed.org/SearchBasic.php?RID=2465338
  66. EMA calls for hydroxyethyl starch solutions to be taken off market - The BMJ, accessed August 1, 2025, https://www.bmj.com/content/360/bmj.k225
  67. Veterinarians concerned about FDA's hydroxyethyl starch warning - News - VIN, accessed August 1, 2025, https://news.vin.com/doc/?id=5881510
  68. Search - FDA, accessed August 1, 2025, https://www.fda.gov/search?s=hydroxyethyl+starch&sort_bef_combine=rel_DESC
  69. EMA confirms that hydroxyethyl starch solutions should not be used in critically ill, sepsis, or burns patients | The BMJ, accessed August 1, 2025, https://www.bmj.com/content/347/bmj.f6197
  70. Hydroxyethyl-starch solutions (HES) should no longer be used in patients with sepsis or burn injuries or in critically ill patients – CMDh endorses PRAC recommendations - EMA, accessed August 1, 2025, https://www.ema.europa.eu/en/news/hydroxyethyl-starch-solutions-hes-should-no-longer-be-used-patients-sepsis-or-burn-injuries-or-critically-ill-patients-cmdh-endorses-prac-recommendations
  71. Hydroxyethyl starch (HES) - European Commission, accessed August 1, 2025, https://ec.europa.eu/health/documents/community-register/2013/20131219127286/anx_127286_en.pdf
  72. Hydroxyethyl-starch solutions for infusion recommended for suspension from the market | European Medicines Agency (EMA), accessed August 1, 2025, https://www.ema.europa.eu/en/news/hydroxyethyl-starch-solutions-infusion-recommended-suspension-market
  73. EMA committee maintains recommendation to suspend hydroxyethyl-starch solutions for infusion - The Pharmaceutical Journal, accessed August 1, 2025, https://pharmaceutical-journal.com/article/news/ema-committee-maintains-recommendation-to-suspend-hydroxyethyl-starch-solutions-for-infusion
  74. sccm.org, accessed August 1, 2025, https://sccm.org/survivingsepsiscampaign/guidelines-and-resources/surviving-sepsis-campaign-adult-guidelines#:~:text=Sepsis%20and%20septic%20shock%20are,treatment%20and%20resuscitation%20begin%20immediately.&text=For%20patients%20with%20sepsis%2Dinduced,first%203%20hours%20of%20resuscitation.
  75. Fluid resuscitation in sepsis: the great 30 mL per kg hoax - Journal of Thoracic Disease, accessed August 1, 2025, https://jtd.amegroups.org/article/view/34911/html
  76. Surviving Sepsis Campaign 2021 Adult Guidelines - SCCM.org, accessed August 1, 2025, https://sccm.org/survivingsepsiscampaign/guidelines-and-resources/surviving-sepsis-campaign-adult-guidelines
  77. Surviving Sepsis: Updated Guidelines From the Society of Critical Care Medicine - AAFP, accessed August 1, 2025, https://www.aafp.org/pubs/afp/issues/2022/1100/practice-guidelines-sepsis.html
  78. Hydroxyethyl starch 130/0.38-0.45 versus crystalloid or albumin in patients with sepsis: systematic review with meta-analysis and trial sequential analysis - PubMed, accessed August 1, 2025, https://pubmed.ncbi.nlm.nih.gov/23418281/
  79. Hydroxyethyl starch 130/0.38-0.45 versus crystalloid or albumin in patients with sepsis - NCBI, accessed August 1, 2025, https://www.ncbi.nlm.nih.gov/books/NBK121327/
  80. Comparison of the effects of albumin and crystalloid on mortality among patients with septic shock: systematic review with meta-analysis - SciELO, accessed August 1, 2025, https://www.scielo.br/j/spmj/a/dgwRpV6XqjT7TL5738qJQ3v/?format=pdf&lang=en

Published at: August 1, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.