Fact-checked by Grok 2 weeks ago

Macrogol

Macrogol, also known as (), is a synthetic, water-soluble used primarily as an osmotic to treat by drawing water into the intestines to soften stool and promote . It is also widely used as an in pharmaceutical formulations and in for biopharmaceuticals. Available in various formulations such as macrogol 3350 and macrogol 4000, it is indicated for the of both occasional and chronic in adults, children aged 6 months and older, and elderly patients. As a minimally absorbed agent, macrogol exerts its effects locally in the without significant electrolyte disturbances, making it a preferred first-line over or saline laxatives for long-term use. Beyond constipation management, macrogol is commonly incorporated into bowel cleansing regimens for diagnostic procedures like , where it facilitates effective preparation with good tolerability. Common administration involves dissolving powder in water for oral intake, with typical adult doses of 10–20 grams daily, and occurring within 1–3 days. Adverse effects are generally mild, including , , and , and it has a favorable safety profile even in vulnerable populations.

Chemistry

Structure and nomenclature

Macrogol is the (INN) assigned by the for (), a synthetic polyether polymer used in pharmaceutical formulations. It shares synonyms such as poly(ethylene oxide) (PEO), though PEO typically denotes higher molecular weight variants of the same compound. The general is \mathrm{H-(OCH_2CH_2)_n-OH}, where n indicates the average , which determines the polymer's average molecular weight; for instance, n \approx 76 corresponds to an average molecular weight of 3350 . The nomenclature "macrogol" was first proposed in WHO's Proposed List 34 of International Nonproprietary Names in 1975 to standardize naming for pharmaceutical applications, ensuring clarity and consistency in medicinal products. This distinguishes pharmaceutical-grade macrogol, which adheres to strict pharmacopeial purity standards (e.g., ), from industrial , which may include impurities unsuitable for medical use. Specific designations like macrogol 3350, macrogol 4000, and macrogol 6000 denote variants with average molecular weights of approximately 3350, 4000, and 6000 , respectively, tailored for different pharmaceutical roles based on their chain length and properties.

Physical and chemical properties

Macrogol, known scientifically as (), is a hygroscopic characterized by its high , which arises from the ability of its oxygen atoms to form bonds with molecules. This property makes it versatile for pharmaceutical applications, where it remains fully miscible in across a wide range of molecular weights, though decreases slightly with increasing chain length. The physical properties of macrogol vary significantly with molecular weight. For instance, macrogol 3350, a commonly used grade, exhibits a of 53–57°C and a of approximately 1.12–1.18 g/cm³. Viscosity also increases with chain length; low-molecular-weight variants like macrogol 200 have a kinematic of about 4.1 mm²/s at 99°C, while higher-weight forms such as macrogol 20,000 reach up to 14,000 mm²/s under similar conditions. Chemically, macrogol is inert and non-toxic at pharmaceutical grades, demonstrating stability under physiological conditions without significant degradation. In its native form, it exhibits low , contributing to its in medical formulations. Pharmaceutical-grade macrogol must meet stringent purity standards, containing not less than 97.0% and not more than 103.0% of the labeled on an basis, with limited to not more than 1.0%. It is also required to be free of residues, with a limit of not more than 1 μg/g as specified by the ().

Pharmacology

Mechanism of action

Macrogol, also known as (), primarily functions as an osmotic by acting as a non-absorbable, high-molecular-weight that remains in the without significant systemic , thereby minimizing metabolic involvement. It exerts its osmotic effect by drawing and retaining into the intestinal through , which hydrates and softens the while increasing its volume and stimulating bowel via distension of the colon wall. This process typically begins within 24 to 72 hours after administration, promoting without direct stimulation of intestinal nerves or muscles.

Pharmacokinetics

Macrogol, a high-molecular-weight of , demonstrates negligible systemic after , with concentrations remaining low and detectable in less than 0.3% of the dose. Peak levels occur between 2 and 4 hours post-ingestion, declining to non-quantifiable amounts within 18 hours, reflecting its limited uptake from the . This poor absorption is primarily due to the polymer's high molecular weight, which restricts paracellular and transcellular passage across the ; for example, macrogol 3350 (average molecular weight 3350 Da) exhibits significantly lower absorption than lower-molecular-weight polyethylene glycols. Distribution of macrogol is largely confined to the gastrointestinal , with no substantial penetration into systemic tissues or organs, as evidenced by the absence of meaningful accumulation even after multiple doses. The reported exceeds 48,000 L, consistent with its osmotic retention of fluid within the gut rather than widespread body distribution. Macrogol undergoes no metabolism in the body, remaining chemically unchanged throughout its transit; it is not subject to intestinal enzymatic degradation or microbial breakdown in the gut. Excretion occurs predominantly via the fecal route, with over 93% of the administered dose recovered unchanged in feces and only 0.19% to 0.25% appearing in urine, indicating minimal renal involvement. The elimination half-life is approximately 4 to 6 hours, with nearly complete fecal clearance achieved within 48 hours. Pharmacokinetic parameters of macrogol show minimal variation across factors such as , , or mild renal impairment, attributable to its negligible and primary fecal elimination pathway. Molecular influences gastrointestinal , with higher weights like macrogol 3350 associated with slower transit times compared to lower-molecular- variants, which can accelerate gut motility.

Medical uses

As a laxative

Macrogol, also known as (), is widely used as an for the treatment of various forms of . It is indicated for chronic idiopathic in adults and children, opioid-induced , and in elderly patients, where it helps restore normal bowel function by retaining water in the intestinal . Additionally, macrogol is employed for bowel preparation prior to diagnostic procedures such as , particularly in patients with underlying chronic to enhance cleansing efficacy. Clinical trials demonstrate macrogol's superior efficacy compared to , with response rates of 70–80% in improving stool frequency and consistency, often achieving significant relief within 1–3 days of initiation. A 2018 systematic review and of randomized controlled trials (RCTs) confirmed that macrogol increases bowel movement frequency by approximately 1.5–2 stools per week more than , while reducing straining and associated symptoms. Long-term use is supported by evidence from RCTs showing sustained efficacy and tolerability for up to 6 months without loss of response. Standard dosing for adults with chronic constipation is 17 g of PEG 3350 daily, dissolved in 240 mL of liquid, with adjustments based on response; pediatric dosing starts at 0.4–0.8 g/kg/day, titrated to achieve soft . from RCTs, including a 2018 , supports this regimen's ability to reduce straining and improve stool form in both adults and children. The osmotic mechanism, which draws water into the colon to soften stool, underpins its rapid and reliable action. In specific conditions, macrogol is effective for constipation-predominant (IBS-C), where RCTs show it relieves constipation symptoms superior to while improving scores. For , a randomized -controlled demonstrated significant increases in stool frequency and reductions in laxative use with macrogol therapy. The 2023 American Gastroenterological Association () guidelines strongly recommend macrogol over stimulant laxatives as first-line therapy for chronic , including in IBS-C and opioid-induced cases, based on moderate-quality evidence from meta-analyses of RCTs.

As an excipient

Macrogol, commonly known as (), functions as a non-active in pharmaceutical formulations, enhancing the physical properties and manufacturability of without exerting therapeutic effects. It serves primarily as a for hydrophobic drugs, a to improve the flexibility and processability of tablets and creams, a to reduce friction during tablet compression and capsule filling, and an osmotic agent in injectables to help maintain isotonicity and stability. For instance, in the (Spikevax), PEG 2000 dimyristoyl is incorporated as a within the nanoparticles, aiding in the protection and delivery of the mRNA payload. The and non- profile of macrogol make it advantageous for diverse applications, as it minimizes while improving drug solubility, , and formulation stability. These properties stem from its hydrophilic nature and tunable molecular weights, allowing customization for specific needs such as reducing protein adsorption on surfaces or enhancing in topical preparations. is frequently employed in oral solid , where it contributes to and functions, supporting the production of robust and elegant tablets. Regulatory bodies, including the FDA, classify macrogol as (GRAS) for pharmaceutical use, with established safety records in oral, topical, and parenteral routes. Typical concentrations range from 1% to 20% w/w, varying by and PEG grade—for example, up to 30% v/v in parenteral solutions for low-molecular-weight variants like PEG , while higher weights are used at lower levels in solids. Historically, PEG has been utilized in pharmaceuticals and since the as a and in dermatological bases, with its application expanding post-2020 to include stabilizers in mRNA vaccines. Its water further facilitates the design of aqueous formulations, ensuring compatibility with active ingredients.

In PEGylation

PEGylation is a bioconjugation technique that involves the covalent attachment of macrogol (, ) chains to proteins, peptides, or other biologics to improve their pharmacokinetic properties. This process typically employs reactive linkers, such as (NHS) esters, to form stable amide bonds with primary amine groups (e.g., residues or the ) on the target molecule, enabling precise control over the site and extent of modification. PEG chain lengths for such conjugations commonly range from 5 to 40 kDa, selected based on the desired balance between steric shielding and retained biologic activity. The attachment of chains confers several key benefits, including a substantial extension of the therapeutic's —often by 20- to 100-fold—through increased hydrodynamic volume and reduced renal clearance via glomerular . This also minimizes proteolytic degradation and while enhancing . A prominent example is (Neulasta), a PEGylated approved by the FDA in 2002 for preventing chemotherapy-induced , where the 20 kDa PEG chain extends the from approximately 3.5 hours to 15–80 hours. Clinically approved PEGylated drugs leveraging macrogol conjugation span multiple therapeutic areas. (Pegasys), modified with a 40 kDa branched PEG, was approved in 2002 for treating chronic hepatitis C, allowing weekly dosing compared to thrice-weekly for the unmodified form. Similarly, (Krystexxa), a PEGylated uricase with multiple 10 kDa PEG attachments, received FDA approval in 2010 for refractory chronic , where it sustains reduction over two weeks versus less than 24 hours for the native enzyme. Despite these advantages, can elicit immune responses, including the formation of anti-PEG antibodies, with pre-existing or treatment-induced incidence reaching approximately 25% in some patient populations and up to 70% overall. These antibodies may accelerate clearance of PEGylated therapeutics, diminish efficacy, or provoke reactions, necessitating monitoring in clinical use.

Safety profile

Contraindications

Macrogol, also known as (), is absolutely contraindicated in patients with intestinal obstruction, , bowel , , , or known to PEG or any formulation components, as these conditions can lead to severe complications such as worsening obstruction or rupture. Relative contraindications include severe , such as active or , and recent gastrointestinal surgery, where use may exacerbate inflammation or postoperative . In special populations, use with caution in neonates and infants under 6 months, as data on tolerance is limited, and only under medical supervision; macrogol should be avoided in neonates with underdeveloped gastrointestinal function due to risks of inadequate tolerance, and caution is advised in patients with renal failure, despite minimal systemic absorption, particularly with formulations containing electrolytes that may alter sodium or levels. Hypersensitivity reactions to are rare, with a estimated at less than 0.1% in the general , though has increased following reports linked to vaccines containing .

Adverse effects

, also known as (), is generally well-tolerated when used as a or , with most adverse effects being mild and gastrointestinal in nature. Common side effects, reported in more than 1% of patients, include (), , , and . These effects are often dose-dependent and can typically be managed by adjusting the dosage or taking the medication with food, leading to resolution in most cases. Rare adverse effects, occurring in less than 0.1% of users, encompass imbalances and seizures, particularly in cases of overdose. Allergic reactions, including , are also uncommon but have been documented, especially with PEGylated drugs where manifests as urticaria, , , or . Post-marketing surveillance from 2021 to 2025, including reports related to PEG-containing mRNA , has highlighted rare instances of such reactions. As of 2025, continued emphasizes screening for PEG in patients with prior to PEG-containing products, including . Long-term use of macrogol does not lead to dependence or alterations in colonic mucosa, as it is non-toxic and passes through the unchanged. A 2023 multicenter study confirmed its safety in pediatric patients with , showing normalization of bowel movements without serious adverse events during maintenance therapy. Effective monitoring involves ensuring adequate hydration to mitigate risks like from , with particular caution advised for individuals prone to allergies as per guidelines.

Drug interactions

Macrogol, a non-absorbable osmotic , exhibits minimal systemic pharmacokinetic interactions due to its lack of absorption into the bloodstream, thereby avoiding metabolic pathways such as (CYP450) enzymes. This results in a low overall potential for drug-drug interactions, as confirmed by clinical databases evaluating polyethylene glycol-based laxatives. However, its osmotic effects in the can influence the absorption of co-administered oral medications by accelerating transit time or altering intestinal resorption. A key pharmacokinetic interaction occurs with , where co-administration of macrogol 4000 leads to reduced ; in a randomized crossover study of healthy volunteers, the area under the curve () for digoxin decreased by 30%, and maximum concentration (Cmax) by 40%, likely due to diminished intestinal without affecting elimination . Similar reductions in have been observed with other oral agents, such as certain antiepileptics (e.g., and ), antibiotics, iron supplements, and , necessitating separation of doses by at least 1–2 hours to mitigate potential therapeutic inefficacy. Pharmacodynamically, macrogol can potentiate the effects of other laxatives, leading to additive and increased risk of or disturbances. Caution is advised when combining macrogol with diuretics, particularly non-potassium-sparing types, as concurrent use has been associated with a twofold increase in cardiovascular mortality risk, attributed to exacerbated imbalances such as . For instance, the severity of adverse effects may heighten with , another stool softener, due to enhanced osmotic activity. In the context of PEGylated therapeutics (e.g., peginterferon or ), interactions with macrogol laxatives are rare and primarily involve theoretical immune modulation from cumulative exposure, though no significant clinical reports document direct pharmacokinetic or pharmacodynamic alterations. Overall, these interactions underscore the importance of timing administration and monitoring in scenarios.

Formulations

Dosage forms

Macrogol, also known as (PEG), is most commonly available in oral for use, primarily as powders intended for reconstitution into solutions. These powders typically contain macrogol 3350, the most prevalent molecular weight variant, in single-dose sachets of 17 grams each, which are dissolved in approximately 240 milliliters of or other beverages prior to ingestion. Macrogol 4000 is similarly formulated as powders in 10-gram sachets for oral solution, often without electrolytes for chronic management. For bowel preparation prior to , macrogol products are supplied as larger powder kits for oral solution, frequently combined with electrolytes to maintain hydration balance. Examples include formulations like HalfLytely, which provides a reduced-volume 2-liter powder mix of macrogol 3350 (approximately 210 grams) with , , and , reconstituted for split-dose administration. Another recent development is Suflave, a sulfate-based macrogol 3350 powder (178.7 grams per bottle) with , , and , approved by the FDA in June 2023; it requires reconstitution of two bottles with water for a complete split-dose regimen. Beyond oral administration, macrogol serves as an in topical such as creams and ointments, where lower molecular weight variants like enhance solubility and provide a non-greasy base for in dermatological applications. In PEGylated therapeutics, macrogol is covalently attached to active pharmaceutical ingredients, resulting in injectable including subcutaneous or intravenous solutions; for instance, is administered as a 6-milligram single-dose injection to stimulate production.

Brand names and availability

Macrogol, also known as (PEG), is marketed under various brand names primarily for its use as a , with additional applications in PEGylated pharmaceutical products. In the United States, the leading brand for relief is MiraLAX, an over-the-counter (OTC) powder formulation of PEG 3350 that is widely available in pharmacies, supermarkets, and online retailers. In the and other European countries, Movicol is a prominent prescription and OTC brand, often containing PEG 3350 combined with electrolytes for treating chronic and bowel preparation. In , Forlax is a commonly prescribed osmotic featuring macrogol 4000, available in sachets for adults and children over 8 years. For PEGylated therapeutics, notable brands include Neulasta (), used to reduce risk in patients, and Adagen (pegademase bovine), an enzyme replacement therapy for disease. Availability of macrogol-based laxatives varies by region and formulation. In the and , low-dose PEG 3350 products like MiraLAX are readily accessible OTC without a prescription for occasional . In the , higher-dose preparations such as Movicol are typically available by prescription for chronic use or bowel cleansing, though some lower-strength versions can be obtained OTC in pharmacies. Global distribution includes brands like Laxido and CosmoCol in the UK and , and equivalents in (e.g., APOHEALTH Macrogol) and (e.g., Molaxole), often as powders for oral solution. Regulatory approvals underscore macrogol's established safety profile. The US Food and Drug Administration (FDA) has recognized compounds as (GRAS) for food and pharmaceutical uses since the 1970s, with specific approval for OTC use in 1999. The (EMA) has authorized various macrogol formulations, including combinations for oral use, through national procedures and periodic safety updates. A recent example is the FDA's 2023 approval of Suflave, a low-volume bowel preparation containing PEG 3350 and electrolytes, designed for prep with improved taste. Since the mid-2000s, generic versions of macrogol laxatives have become widely available, enhancing affordability and access; for instance, generic 3350 entered the market around 2007. In 2023, 3350 ranked among the top prescribed medications in the , at approximately 196th place with over 2 million prescriptions, reflecting its broad clinical utility. The global market for macrogol and PEG-based products, including laxatives and therapeutics, was valued at around USD 5.3 billion in 2024, driven by increasing demand for osmotic laxatives and applications.

Research directions

Advanced PEGylation

Recent innovations in have focused on branched and releasable PEG linkers to minimize while preserving therapeutic efficacy. Branched PEG structures provide enhanced surface coverage on proteins, forming an "umbrella-like" configuration that reduces immune recognition compared to linear , as demonstrated in preclinical studies showing lower anti- responses. Releasable linkers, which allow controlled detachment of PEG under physiological conditions, further mitigate long-term by enabling the protein to regain full activity post-circulation, with customized designs improving stability in nanocarriers. Site-specific conjugation techniques represent another key advancement, targeting precise residues to avoid random modifications that can impair protein function. Enzymatic methods, such as sortase-mediated and ligase, enable precise attachment at non-canonical sites, as highlighted in recent reviews of therapeutic . These approaches have advanced to clinical evaluation, with 2024 trials exploring site-directed for biologics like soluble HLA-G2 homodimers, enhancing stability without compromising immunosuppressive effects. Such innovations build on established by offering greater control over conjugate homogeneity and bioactivity. In next-generation biologics, advanced supports targeted therapies, exemplified by pegylated interleukin-2 (PEG-IL-2) variants like bempegaldesleukin (BEMPEG), which showed promising objective response rates in phase II trials for advanced solid tumors when combined with checkpoint inhibitors but failed to meet primary endpoints in the phase III IO-001 trial, leading to discontinuation of development in 2022. For nanoparticle-based drug delivery, PEG coatings on poly(lactic-co-glycolic acid) () nanoparticles improve properties, enabling prolonged blood retention and controlled release in cancer applications, as evidenced by 2025 studies on enhanced tumor penetration. Challenges in advanced PEGylation include addressing anti-PEG syndrome, characterized by accelerated clearance due to pre-existing or induced antibodies. Mitigation strategies involve hydrophilic alternatives like polysarcosine and zwitterionic polymers, which provide similar effects with reduced , as shown in 2023–2025 preclinical models evading binding. Recent studies from 2023–2025 report significant extensions—often 2- to 5-fold in PEGylated proteins—through these optimized designs, though exact gains vary by conjugate and target. Ongoing trials underscore these advancements, such as expanded evaluations of , a PEGylated inhibitor for (PNH). Approved in 2021, phase 3 extensions in 2024 confirmed sustained improvements and reduced transfusion needs over three years, with real-world data from the COMPLETE study further validating long-term efficacy in diverse cohorts.

Neurological applications

Macrogol, also known as (), has been investigated for its fusogenic properties in repairing damaged neuronal membranes, particularly in injured axons where it promotes rapid to restore and prevent secondary degeneration. This involves PEG stabilizing and merging bilayers at injury sites, thereby reestablishing axonal integrity and electrophysiological function without eliciting immune responses. In spinal cord models, PEG acts as a to bridge transected tissues, facilitating immediate restoration of conduction and supporting regeneration by creating a permissive environment for axonal regrowth. Preclinical evidence from animal models demonstrates PEG's efficacy in traumatic nerve injuries. In rat models of sciatic nerve severance, PEG fusion has restored axonal continuity, reorganized sensory terminals in the spinal cord, and led to significant functional recovery. Similar results have been observed in canine models of spinal cord transection, where PEG-mediated fusion enhanced sensorimotor recovery by preventing Wallerian degeneration and promoting tissue bridging. These findings highlight PEG's potential in both peripheral and central nervous system injuries, with applications extending to preclinical neuroprotective strategies in stroke models, where PEG-3350 has mitigated neuronal damage post-oxygen-glucose deprivation by preserving membrane integrity. Early human translation includes phase I and II trials for peripheral nerve repair. A 2024 randomized involving digital nerve injuries showed that PEG fusion accelerated sensory recovery and improved patient-reported outcomes compared to standard repairs, with no significant adverse events. As of 2025, comprehensive reviews affirm PEG's promise in neurological repair, bolstered by the FDA's 2024 orphan drug designation for PEG-3350 in treating peripheral nerve injuries requiring repair, paving the way for advanced clinical development in sealants.

Oncology applications

Macrogol, or (PEG), plays a significant role in through PEGylation, which enhances the , , and tumor targeting of chemotherapeutic agents while minimizing systemic toxicity. A prominent example is Doxil, a PEGylated liposomal formulation of approved by the FDA in 1995 for treating AIDS-related and later for ovarian and breast cancers. This formulation leverages the stealth properties of PEG to prolong circulation time, improve tumor accumulation via the , and reduce compared to free . Clinical studies have demonstrated that Doxil achieves higher tumor localization and lower cardiac exposure, making it a standard in platinum-resistant therapy. In , high-molecular-weight has exhibited chemopreventive effects in animal models of , primarily through mechanisms such as downregulation of () and inhibition of aberrant crypt foci formation. In azoxymethane-induced rat models, dietary supplementation reduced the incidence of colon tumors from 70% to 10%, representing an approximately 86% decrease, and lowered multiplicity in surviving animals. More recent analyses of precursor lesions show achieving a 43% reduction in aberrant crypt foci compared to controls. These effects are attributed to PEG's ability to modulate cellular and , though its precise role in this context remains under investigation; limited human studies, including a small 2018 randomized showing reduced aberrant crypt foci in patients with prior adenomas, have been conducted, but larger confirmatory trials are needed. Emerging applications include PEG-stabilized nanoparticles (LNPs) for mRNA-based cancer , which deliver tumor to stimulate immune responses. These platforms, incorporating PEG- for and stability, have shown promise in preclinical models by enhancing and T-cell activation against solid tumors like and . Additionally, PEG-containing antibody-drug conjugates (PEG-ADCs) are advancing, with several candidates featuring PEG linkers entering phase III trials for solid tumors as of 2025, aiming to improve payload and efficacy in breast and lung cancers. Recent 2024 preclinical data further highlight PEG micelles for , where PEG-phosphatidylethanolamine (PEG-PE) transforms tumor extracellular vesicles into micelle-like structures, facilitating cytoplasmic and boosting antitumor immunity in mouse models.

Other emerging uses

In tissue engineering, polyethylene glycol (PEG) hydrogels serve as versatile scaffolds due to their , tunable mechanical properties, and ability to mimic the , facilitating and proliferation for regeneration. Recent studies have demonstrated that PEG-based hydrogels, often combined with natural polymers like , exhibit high elasticity and support differentiation, with preclinical models showing improved repair outcomes. For instance, enzymatically crosslinked PEG hydrogels have been shown to enhance chondrogenic function , while 3D-printed PEG-PLA/gelatin composites promote viability and tissue integration in models of defects. Although clinical translation remains preclinical, 2023-2024 reviews highlight ongoing advancements toward potential trials for articular defects. In , PEG acts as a enhancer by stabilizing complexes and improving cellular uptake, thereby boosting efficiency in non-viral vectors. When conjugated to or adeno-associated viruses, PEG modifications reduce toxicity and enhance endosomal escape, leading to higher expression in preclinical models. Additionally, PEGylated (PEG-IFN) has emerged in antiviral applications, particularly for , where post-2023 research confirmed that a single subcutaneous dose of PEG-IFN lambda accelerates viral clearance and reduces hospitalization risk in vaccinated outpatients, with relative risk reductions up to 62% against variants. These findings underscore PEG's role in extending the and efficacy of therapeutic proteins in infectious disease management. Environmentally, PEG derivatives contribute to the development of biodegradable plastics by serving as plasticizers in blends with polymers like (PBAT) or (PLA), enabling melt-processible materials that degrade under composting conditions. Preclinical studies from 2023 have shown that PEG-plasticized cyclic depsipeptide blends achieve over 90% in within months, offering a sustainable to conventional petroleum-based plastics while maintaining mechanical strength for packaging applications. This approach leverages PEG's hydrophilicity to accelerate , addressing without compromising functionality. Emerging applications also include , where PEG-modified dressings promote moist environments and antimicrobial activity to accelerate epithelialization. Polyurethane foam dressings functionalized with PEG and triethoxysilane have demonstrated superior and reduced inflammation in preclinical diabetic models. Broad 2024-2025 reviews emphasize these diverse uses, noting PEG's versatility in addressing unmet needs across regenerative and sustainable technologies.