top of page

Technical Review Article | Open Access | Published 8th October 2025

A Comprehensive Review on the Stability and Degradation of Polysorbates in Biopharmaceuticals


Krupal Morker¹, Ravi Patel², Dipen Purohit³ | EJPPS | 303 (2025) https://doi.org/10.37521/ejpps30301 Back to Journals |  Article |Abstract| References | Authors

Abstract 

Polysorbates (PS), particularly polysorbate 20 (PS20) and polysorbate 80 (PS80), are widely utilized as non-ionic surfactants in biopharmaceutical formulations. Their primary role is to stabilize therapeutic proteins against interfacial stresses encountered during manufacturing, handling, and storage. Despite their widespread use, PS are complex and heterogeneous mixtures prone to degradation. The two primary degradation pathways are hydrolysis and oxidation. Hydrolytic degradation is often enzyme-mediated, commonly by residual host cell proteins such as lipases and phospholipases. Oxidative degradation can be initiated by light, temperature, metal ions, or peroxides present as impurities in raw materials. PS degradation yields various products, including free fatty acids (FFA), short-chain organic acids, aldehydes, ketones, and peroxides. The complexity of PS and their degradation products necessitates the use of sophisticated analytical methods for comprehensive characterization and monitoring. Techniques such as liquid chromatography coupled with mass spectrometry (LC-MS), charged aerosol detection (CAD), or evaporative light scattering detection (ELSD), as well as specific assays for FFA and peroxides, are employed. A significant concern arising from PS degradation is the formation of visible and subvisible particles, often composed of poorly soluble FFAs. While PS degradation and particle formation may not always impact protein quality attributes such as aggregation or biological activity under certain conditions, their potential consequences on product quality, safety, and compliance require careful consideration. Effective mitigation and control strategies involve stringent raw material qualification, optimization of manufacturing processes, robust formulation development, and comprehensive stability testing with appropriate analytical methods.


Figure 1. Graphical Abstract
Figure 1. Graphical Abstract

Keywords: Polysorbate 20, Polysorbate 80, Degradation, Hydrolysis, Oxidation, Free Fatty Acids, Particles, Therapeutic Proteins, Biopharmaceuticals, Analytical Methods, Control Strategy



Introduction


The successful development and commercialization of biopharmaceutical products, particularly complex protein-based therapeutics, hinges critically on their ability to maintain stability, efficacy, and safety throughout their extended shelf life. In this context, non-ionic surfactants, predominantly polysorbate 20 (PS20) and polysorbate 80 (PS80), are indispensable excipients². These amphiphilic molecules are widely integrated into biopharmaceutical formulations due to their multifaceted protective roles. Their primary mechanism of action involves preferential adsorption to various interfaces—such as air-liquid, silicone oil-liquid, and protein-liquid—thereby effectively lowering interfacial tension³. This reduction in interfacial energy is crucial for preventing critical quality attribute (CQA) issues, including protein adsorption to container surfaces, subsequent denaturation, and irreversible aggregation, which can lead to loss of biological activity and potential immunogenicity. This vital stabilizing function is imperative not only during rigorous manufacturing processes, including mixing, filtration, and aseptic filling, but also throughout the product's lifecycle, encompassing transportation, storage, and patient administration, all of which may involve various mechanical stresses and thermal fluctuations, as encountered in freeze-thaw cycles. The inherent efficacy of polysorbates, even at remarkably low concentrations, is attributed to their optimal hydrophilic-lipophilic balance (HLB) and low critical micelle concentrations (CMC), enabling them to effectively shield therapeutic proteins¹⁻⁶.

However, the perceived simplicity of polysorbates belies their inherent complexity. Commercially available polysorbates are not discrete chemical entities but rather intricate and heterogeneous mixtures of partial fatty acid esters of sorbitol and its anhydrides, ethoxylated with varying numbers of oxyethylene units. This structural heterogeneity arises directly from their synthetic routes, primarily the ethoxylation of sorbitan esters. This intrinsic variability, coupled with their documented susceptibility to diverse degradation pathways, poses formidable challenges in consistently maintaining the long-term stability of biopharmaceutical formulations. The degradation of polysorbates can severely compromise their critical stabilizing function, leading to a cascade of undesirable events. These include the formation of problematic degradation products, such as free fatty acids (e.g., oleic acid from PS80, lauric acid from PS20) and reactive oxygen species, which can directly or indirectly induce protein aggregation and particle formation⁴. The generation of visible and subvisible particles, a direct consequence of surfactant degradation or its inability to stabilize the protein, is a significant concern for product safety and regulatory compliance. Ultimately, these issues can profoundly impact the overall quality, safety profile, and regulatory acceptability of the final biopharmaceutical product⁷.

Addressing the multifaceted challenges associated with polysorbate stability necessitates a comprehensive and interdisciplinary understanding. This includes a detailed elucidation of their complex compositional profiles and intricate degradation pathways, a thorough assessment of the impact of polysorbate degradation products on both the biopharmaceutical formulation matrix and the therapeutic protein itself, and the strategic application of advanced analytical methodologies and robust control strategies. This review aims to consolidate and critically evaluate recent advancements and findings concerning polysorbate heterogeneity and the nuanced mechanisms driving their degradation. Furthermore, it will highlight state-of-the-art analytical techniques, including advanced chromatographic and spectroscopic methods, crucial for their comprehensive characterization and continuous monitoring throughout the product lifecycle. The review will also critically discuss the profound potential consequences of polysorbate degradation on biopharmaceutical quality attributes and summarize current and emerging approaches for mitigating degradation and implementing effective control strategies to ensure product integrity and patient safety.⁸


Figure 2. Polysorbate Degradation and Its Impact on Biopharmaceuticals
Figure 2. Polysorbate Degradation and Its Impact on Biopharmaceuticals

2. Polysorbate Structure and Heterogeneity

Polysorbates, broadly classified as polyoxyethylene (POE) derivatives of sorbitan esters, constitute a cornerstone of modern biopharmaceutical formulation due to their amphiphilic nature. The archetypal structure of a polysorbate molecule comprises a sorbitan head group, which serves as the lipophilic anchor, esterified with one or more fatty acyl chains. This core is subsequently rendered hydrophilic through the attachment of multiple polyoxyethylene (POE) units, typically an average of 20, to the available hydroxyl groups of the sorbitan ring. The specific fatty acid moiety primarily defines the polysorbate type: polysorbate 20 (PS20) is predominantly based on lauric acid (C12), while polysorbate 80 (PS80) is characterized by its primary fatty acid, oleic acid (C18:1).

However, the commercial production of polysorbates, involving the ethoxylation of sorbitan esters, results in products that deviate significantly from this idealized, uniform chemical structure. Consequently, commercial polysorbates exhibit a considerable degree of inherent heterogeneity, which can be attributed to several critical factors⁹,¹⁰:


Fatty Acid Composition:

The fatty acid feedstocks utilized for esterification are rarely chemically pure, contributing substantially to the heterogeneity of the final polysorbate product. Compendial monographs, such as those established by the United States Pharmacopeia (USP), European Pharmacopoeia (Ph. Eur.), and Japanese Pharmacopoeia (JP), define the acceptable ranges for the constituent fatty acid profiles. For instance, compendial PS80 is stipulated to contain a minimum of 58% oleic acid, yet it can encompass notable proportions of other fatty acids, including palmitic (up to 16%), stearic (up to 6%), linoleic (up to 18%), linolenic (up to 4%), myristic (up to 5%), and palmitoleic (up to 8%) acids. Similarly, PS20 is required to contain 40-60% lauric acid but can also include significant quantities of myristic (14-25%), palmitic (7-15%), capric (up to 10%), caprylic (up to 10%), stearic (up to 7%), and linoleic (up to 3%) acids. It is important to note that more stringent compositional requirements may be imposed for specific high-purity grades; for example, the Chinese Pharmacopoeia (ChP) mandates greater than or equivalent to 98% oleic acid for PS80 designated for injection¹¹.


Degree of Esterification:

The polyhydroxylic nature of sorbitan and its derivatives allows for varying degrees of esterification. Consequently, commercial polysorbates are complex mixtures containing not only monoesters but also di-, tri-, and even tetra-esters, where multiple fatty acid chains are attached either directly to the sorbitan ring or to the terminal hydroxyl groups of the POE moieties. Crucially, these multiple esterifications are not necessarily confined to a single fatty acid species, further compounding the molecular diversity¹².


POE Chain Length Distribution:

The process of ethoxylation leads to a statistical distribution in the number of oxyethylene units attached to each hydroxyl group. While the average total sum of POE units is typically cited as approximately 20 (represented as W+X+Y+Z = 20 in the general chemical structure), the actual chain lengths of individual POE segments can vary considerably, ranging from approximately 10 to 35 units. This polydispersity in POE chain length directly impacts the hydrophilic character and micellar properties of individual polysorbate molecules¹³.


Core Structure Variations:

The hydrophilic core of commercial polysorbates is rarely exclusively sorbitan. It often exists as a heterogeneous mixture encompassing sorbitan and its dehydrated derivatives, primarily isosorbide species. Isosorbide, being a bicyclic ether, presents a reduced number of hydroxyl positions (typically two) available for esterification compared to sorbitan, leading to different esterification patterns and overall molecular architectures. Furthermore, the manufacturing process can introduce non-esterified sorbitan/isosorbide-PEG species and residual polyethylene glycol (PEG) oligomers as process-related impurities or by-products, further contributing to the complexity¹⁴.

This cumulative structural variability means that commercially available polysorbates are not single compounds but rather highly complex, polydisperse mixtures comprising numerous related molecular species. This intrinsic complexity presents significant analytical challenges for their comprehensive characterization and quantification, particularly when monitoring changes indicative of degradation. To address these challenges and to minimize the impact of certain impurities, "super-refined" or "ultra-pure" grades of polysorbates have been developed. These specialized grades aim to significantly reduce the levels of specific detrimental impurities, such as peroxides and other degradation by-products, thereby offering enhanced quality and improved stability profiles for sensitive biopharmaceutical formulations¹⁵.


3. Polysorbate Degradation Pathways and Root Causes

The structural complexity and amphiphilic nature of polysorbates, while essential for their stabilizing function, also render them susceptible to various degradation pathways. These processes can occur throughout the lifecycle of a biopharmaceutical product, from the manufacturing and storage of raw materials to the long-term stability of the final drug product. Under pharmaceutically relevant conditions, the two predominant degradation mechanisms observed are hydrolysis and oxidation, each contributing uniquely to polysorbate breakdown and potential impacts on formulation integrity³,¹⁶,¹⁷.


3.1. Hydrolytic Degradation

Hydrolysis, in the context of polysorbates, involves the nucleophilic attack on the ester bond that links the fatty acid moiety to the sorbitan-polyoxyethylene (POE) or isosorbide-POE core. This scission event yields two primary degradation products: free fatty acids (FFAs) and the corresponding unesterified sorbitan/isosorbide-PEG species (i.e., PEGylated sorbitans and isosorbides). The accumulation of FFAs is particularly concerning as they possess lower solubility than intact polysorbates and can precipitate, leading to visible and subvisible particle formation, and in some cases, induce protein aggregation or phase separation.

While chemical hydrolysis, catalysed by extreme pH conditions (highly acidic or basic), is generally considered negligible or very slow under typical biopharmaceutical formulation conditions (pH 4-8), enzymatic hydrolysis has emerged as a critically important and increasingly recognized degradation pathway. This pathway is predominantly catalysed by trace levels of residual host cell proteins (HCPs) carried over from the upstream protein manufacturing process, particularly in mammalian cell culture systems (e.g., Chinese Hamster Ovary, CHO cells). Specific classes of enzymes, such as lipases and phospholipases, possess the esterase activity required to cleave the ester bonds of polysorbates. Notably, Lysosomal Phospholipase A2 (LPLA2) and Putative Phospholipase B-like 2 (PLBL2) have been explicitly identified and implicated in PS degradation in therapeutic protein formulations derived from CHO cell lines. The catalytic efficiency of these enzymes is influenced by several factors, including their concentration, the incubation temperature, and the pH of the formulation, with optimal activity typically observed under conditions relevant to drug product storage. Enzyme-mediated hydrolysis of PS20, in particular, has been frequently associated with the rapid formation of insoluble FFA particles, which can manifest as turbidity or visible precipitates within the formulation¹⁸,¹⁹.


Figure 3. Degradation Pathways of Polysorbates in Biopharmaceuticals
Figure 3. Degradation Pathways of Polysorbates in Biopharmaceuticals

3.2. Oxidative Degradation

Oxidative degradation represents another significant pathway for polysorbate breakdown, proceeding primarily via a free-radical chain mechanism. This process is often initiated and accelerated by exposure to molecular oxygen (O₂), light (especially UV and visible light), trace amounts of transition metal ions (e.g., Fe²⁺, Cu²⁺), or elevated temperatures.


The autoxidation process typically involves three key stages:

Initiation: Formation of a free radical (R•) from polysorbate, often facilitated by trace metal ions or light-induced homolytic cleavage. Pre-existing peroxides (e.g., hydroperoxides from raw material or prior oxidation) can also initiate the chain by homolytic cleavage.

Propagation: The polysorbate radical (R•) rapidly reacts with molecular oxygen to form a peroxy radical (ROO•). This peroxy radical can then abstract a hydrogen atom from another polysorbate molecule (RH), forming a hydroperoxide (ROOH) and generating a new polysorbate radical (R•), thereby propagating the chain reaction.

Termination: Two radicals combine to form a stable, non-radical product, terminating the chain reaction.

Oxidation can occur at several vulnerable sites within the polysorbate molecular structure. The polyoxyethylene (POE) chain is susceptible to oxidation at the α-carbon adjacent to the ether oxygen, leading to the formation of hydroperoxides and subsequent scission of the POE chain. The ester bond itself can also undergo oxidative cleavage. However, the most labile site for oxidation, particularly in polysorbate 80 (PS80), is the carbon-carbon double bond within the unsaturated oleic acid moiety. This double bond makes PS80 generally more susceptible to oxidative degradation compared to PS20, which is primarily composed of saturated lauric acid.

Oxidative degradation yields a complex and diverse mixture of primary and secondary degradation products. Primary products include hydroperoxides, which are often unstable and can further decompose. Secondary products arise from the decomposition of hydroperoxides and scission of the polymer chains, encompassing a broad range of compounds such as aldehydes (e.g., formaldehyde, acetaldehyde), ketones, short-chain organic acids (e.g., formic acid, acetic acid), and various oxidized fatty acids (e.g., azelaic acid, nonanoic acid from oleic acid). It is also important to note that peroxides can be present as pre-existing impurities in polysorbate raw materials, originating from their synthesis or improper storage, which can readily initiate or accelerate further oxidative degradation within the final formulation.

Both hydrolytic and oxidative pathways can concurrently contribute to the overall degradation profile observed in a biopharmaceutical formulation. Identifying the predominant degradation mechanism(s) and their root causes is paramount for designing and implementing effective mitigation strategies, optimizing formulation composition, and ensuring the long-term stability and quality of the therapeutic protein²⁰⁻²².


4. Analytical Characterization of Polysorbates and Degradation Products

The accurate and comprehensive characterization of polysorbate content, composition, and their myriad degradation products within complex biopharmaceutical formulations represents a formidable analytical challenge. This complexity stems from several inherent factors: the broad molecular heterogeneity of commercial polysorbates (as discussed in Section 2), the potential for significant matrix interference from high concentrations of therapeutic proteins and other excipients, and the often-limited availability of universal reference standards for all the diverse polysorbate species and their degradants. Consequently, a holistic analytical toolbox is indispensable, necessitating the strategic deployment of complementary methodologies for both routine quality control monitoring and in-depth mechanistic characterization²³,²⁴.


4.1. Sample Preparation

Prior to analysis, a crucial sample preparation step is frequently required to isolate polysorbate-related components from the bulk therapeutic protein and other high-concentration excipients, and in some cases, to achieve an adequate concentration for detection. Common techniques employed include solid-phase extraction (SPE), liquid-liquid extraction (LLE), or protein precipitation using organic solvents (e.g., methanol, acetonitrile, or combinations thereof). SPE, leveraging various stationary phases (e.g., reversed-phase, mixed-mode), is often favoured for its ability to selectively enrich polysorbates while removing protein. However, each sample preparation strategy must be meticulously optimized and validated to minimize potential pitfalls, such as the irreversible loss of polysorbate molecules or specific degradation products due to adsorption onto surfaces or incomplete extraction, as well as to avoid the introduction of analytical artifacts or changes in the degradation profile. In some cases, direct injection methods, particularly for high-sensitivity mass spectrometry, may be explored to circumvent these sample preparation complexities, though matrix effects can become a significant consideration.


4.2. Chromatographic and Detection Methods

The inherent complexity of polysorbate mixtures and their degradation profiles necessitates the use of high-resolution separation techniques coupled with highly selective and sensitive detectors. Various hyphenated techniques coupling advanced chromatographic separation with complementary detection methods are widely utilized:


4.2.1. Liquid Chromatography-Mass Spectrometry (LC-MS)

LC-MS stands as a gold standard for the comprehensive characterization of polysorbate composition and the definitive identification of individual degradation products due to its unparalleled ability to resolve complex mixtures and provide structural information. Separation is typically achieved using reversed-phase (RP-LC) or mixed-mode (MM-LC) chromatography, which can resolve polysorbate species based on their fatty acid chain length, degree of esterification, and POE chain length distribution. RP-LC is particularly effective for separating molecules with varying lipophilicity, while MM-LC columns, possessing both hydrophobic and ion-exchange properties, offer unique selectivity for different polysorbate components and their degradants.

Mass spectrometry, especially high-resolution accurate-mass (HRAM) systems such as Quadrupole Time-of-Flight (QTOF) or Orbitrap, provides precise mass-to-charge (m/z) ratios, enabling confident identification of intact polysorbate species, their various isoforms, and a wide array of degradation products, including free fatty acids (FFAs), oxidized polysorbates, and fragmented POE chains. Electrospray ionization (ESI) in positive mode is commonly employed for comprehensive characterization of intact polysorbates and their polyethoxylated fragments, yielding protonated or ammoniated adducts. Negative mode ESI is particularly advantageous for the sensitive detection and characterization of negatively charged species like FFAs. While LC-MS excels at relative quantification of different species, achieving accurate absolute quantification remains challenging due to varying ionization efficiencies and the lack of readily available standards for all components. The use of isotopically labelled internal standards can partially address this for targeted analytes. Untargeted metabolomics-like approaches using advanced data processing software are increasingly applied for comprehensive profiling of degradation²⁵,²⁶.


4.2.2. High-Performance Liquid Chromatography coupled with Charged Aerosol Detection (HPLC-CAD) / Evaporative Light Scattering Detection (ELSD)

CAD and ELSD are widely employed for the quantitative analysis of polysorbates and for monitoring changes in their overall concentration or distribution over time. These universal detectors respond to non-volatile analytes after nebulization and subsequent evaporation of the mobile phase, making them suitable for polysorbates which lack significant UV chromophores. MM-HPLC coupled with CAD (MM-HPLC-CAD) has gained significant traction for the quantification of total polysorbate content and for providing stability-indicating profiles, as it can differentiate intact polysorbate from some degraded components. HPLC-CAD generally offers improved linearity and sensitivity over ELSD due to its more consistent response across varying analyte concentrations. While these methods are powerful for quantitative assessment and routine quality control, they provide limited structural information compared to MS-based techniques, requiring complementary methods for specific identification of degradation products²⁷,²⁸.


4.2.3. Gas Chromatography-Mass Spectrometry (GC-MS)

GC-MS is a highly valuable technique, particularly for the analysis of volatile or semi-volatile degradation products. It is extensively used for the precise identification and quantification of free fatty acids liberated from polysorbate hydrolysis, often after derivatization (e.g., methylation to fatty acid methyl esters, FAMEs) to enhance volatility and chromatographic behaviour. GC-MS is also critical for detecting other low molecular weight oxidative degradation products such as aldehydes (e.g., formaldehyde, acetaldehyde) and ketones, which can have significant toxicological implications²⁹,³⁰.


4.2.4. Size Exclusion Chromatography (SEC)

While not directly characterizing polysorbate structure, SEC (also known as Gel Permeation Chromatography, GPC) can be employed to assess the size distribution of polysorbate components or, more commonly, to monitor protein aggregation induced or mediated by polysorbate degradation. Changes in polysorbate micelle size or the presence of non-micellar polysorbate species can also be investigated³¹.


4.3. Functional and Specific Assays

In addition to chromatographic methods, several functional and specific assays provide valuable insights into polysorbate integrity and the presence of critical degradation products:


4.3.1. Fluorescence Micelle Assay (FMA)

FMA is a relatively simple and rapid flow injection assay designed to quantify the total functional polysorbate content based on its ability to form micelles above the critical micelle concentration (CMC). It utilizes a fluorescent dye, such as N-phenyl-1-naphthylamine (NPN), whose fluorescence quantum yield significantly increases upon partitioning into the hydrophobic core of polysorbate micelles. While useful for assessing the functional capacity of the surfactant (i.e., its ability to form micelles), FMA may not distinguish between intact polysorbate and certain surface-active degradation products if these products retain micelle-forming capability. Furthermore, FMA can be susceptible to interference from other hydrophobic components within the formulation or from the therapeutic protein itself, which may interact with the dye or alter micelle formation. Despite these limitations, FMA has demonstrated utility in monitoring overall changes in total polysorbate content, particularly under stress conditions such as light exposure³².


4.3.2. Free Fatty Acid (FFA) Analysis

The quantification of free fatty acids is a direct and highly sensitive indicator of hydrolytic degradation of polysorbates. Specific analytical methods are employed for this purpose, often involving an initial solid-phase extraction step to separate FFAs from the complex sample matrix. This is typically followed by fluorescent labelling (e.g., using 9-anthryldiazomethane (ADAM) or 4-(N,N-dimethylamino) phenylphenacyl bromide (PDAM)) to enhance detection sensitivity, and subsequent separation by reversed-phase ultra-performance liquid chromatography (RP-UPLC) coupled with fluorescence detection (FLR) or RP-HPLC with UV/fluorescence detection. These methods provide quantitative data on individual FFA species, allowing for precise tracking of hydrolytic pathways³³,³⁴.


4.3.3. Peroxide Assays

Peroxide assays, such as colorimetric methods (e.g., FOX2 (ferrous oxidation-xylenol orange) or Amplex Red), are used to monitor the total peroxide content in polysorbate raw materials or in-formulation. These assays detect hydroperoxides and organic hydroperoxides, serving as indicators of the early stages of oxidative degradation. However, it is crucial to recognize that peroxides are intermediate products in the oxidative cascade and can be further consumed as the reaction proceeds. Therefore, peroxide levels may not always directly correlate with the total extent of oxidative degradation, and declining peroxide levels do not necessarily indicate improved stability if secondary degradation products are accumulating. Electrochemical detection methods can offer alternative approaches for peroxide monitoring¹⁶.


4.4. Other Advanced Techniques

Beyond the core chromatographic and specific assays, several other advanced analytical techniques contribute significantly to the comprehensive understanding of polysorbate stability and degradation:


4.4.1. Nuclear Magnetic Resonance (NMR) Spectroscopy

NMR spectroscopy, particularly proton (¹H-NMR) and carbon (¹³C-NMR), provides detailed structural information on polysorbate constituents and can directly monitor degradation. ¹H-NMR can quantify different fatty acid components, determine the average POE chain length, and detect the formation of specific degradation products (e.g., aldehydes from oxidation, changes in ester vs. free hydroxyl protons from hydrolysis). It is also amenable to direct analysis in complex matrices without extensive sample preparation³⁵,³⁶.


4.4.2. Particle Analysis Techniques

Given that polysorbate degradation often leads to particle formation, techniques designed for particle quantification and characterization are critical. These include:

Light Obscuration (LO): A compendial method for subvisible particle counting.


Microfluidic Imaging (MFI) / Flow Imaging Microscopy: Provides morphological data and counts for subvisible particles, differentiating between protein aggregates, silicone oil droplets, and potential polysorbate-derived particles (e.g., FFA particles).


Dynamic Light Scattering (DLS): Measures hydrodynamic size distributions of micelles and larger aggregates.

Nanoparticle Tracking Analysis (NTA): For quantifying and sizing sub-micron particles³⁷.


4.4.3. Spectroscopic Identification of Particles

Once particles are detected, spectroscopic methods can help identify their chemical nature:

Fourier-transform Infrared (FTIR) Microscopy / Raman Spectroscopy: These techniques can provide chemical fingerprints of individual particles, allowing for differentiation between proteinaceous aggregates, silicone oil droplets, and fatty acid-derived particles (e.g., from polysorbate hydrolysis ³⁸,³⁹).


4.4.4. Surface Tension Measurement

As a functional assay, surface tension measurement can indirectly monitor the overall surfactant activity of the formulation. A significant increase in surface tension over time can indicate a loss of functional polysorbate due to degradation, even if the total mass concentration of polysorbate is seemingly unchanged (e.g., if degradation products are still surface-active but less effective).

The effective analytical characterization of polysorbates and their degradation products requires a multi-pronged approach, integrating orthogonal techniques to provide comprehensive information on chemical composition, functional integrity, and the presence of critical impurities. This integrated analytical strategy is essential for understanding degradation mechanisms, ensuring product quality, and supporting regulatory submissions⁴⁰.


5. Impact of Polysorbate Degradation on Biopharmaceutical Quality

The insidious degradation of polysorbates, often subtle in its initial stages, can precipitate a cascade of detrimental effects on the critical quality attributes (CQAs) and overall safety profile of therapeutic protein formulations. The consequences extend beyond the excipient itself, directly impinging upon the stability, efficacy, and immunogenic potential of the active pharmaceutical ingredient (API)⁴¹.


5.1. Loss of Surfactant Function and Protein Stability

The primary role of polysorbates in biopharmaceutical formulations is to mitigate interfacial stresses and prevent protein adsorption, denaturation, and subsequent aggregation. Degradation, primarily through hydrolysis and oxidation, directly diminishes the concentration of intact and functionally active polysorbate molecules. If the effective concentration of functional surfactant drops below a critical threshold—which may or may not align with the critical micelle concentration (CMC) of the original mixture due to changes in overall surface activity—the formulation's protective capacity against various stresses becomes compromised. This vulnerability is particularly evident during mechanical agitation (e.g., shaking, pumping, shipping), freeze-thaw cycles, and exposure to air-liquid or silicone oil-liquid interfaces. The reduced interfacial coverage by functional polysorbate can lead to increased protein adsorption to container surfaces, accelerated protein unfolding, and subsequent aggregation, ultimately manifesting as visible particulate matter, subvisible aggregates, or loss of protein monomer.

It is crucial to note that some polysorbate degradation products, particularly certain partial esters resulting from incomplete hydrolysis or specific oxidized species, may retain a degree of surface activity. Their contribution to the overall stabilization effect, however, is often diminished and can be highly variable depending on their specific chemical structure and concentration. Therefore, assessing the true impact of polysorbate degradation necessitates not just chemical quantification but also comprehensive functional stability studies, such as agitation stress tests, freeze-thaw challenges, and forced degradation studies designed to mimic relevant stress conditions encountered during the product's shelf-life and administration. These studies evaluate the biopharmaceutical product's ability to withstand stress in the presence of degraded polysorbates⁴⁰.


5.2. Protein Oxidation Induced by Polysorbate Degradants

A significant and often underappreciated consequence of polysorbate oxidative degradation is the generation of reactive oxygen species (ROS) and other electrophilic oxidative products. These species, which include hydroperoxides, aldehydes, ketones, and radical intermediates, can readily diffuse within the aqueous formulation. Critically, they can then directly react with susceptible amino acid residues within the therapeutic protein, leading to post-translational modifications. Methionine and tryptophan residues are particularly prone to oxidation due to their sulphur-containing (methionine) and indole ring (tryptophan) structures, but other residues like histidine, cysteine, and tyrosine can also be targeted.


Protein oxidation can induce profound alterations in the protein's primary, secondary, and tertiary structures. This can lead to misfolding, covalent and non-covalent aggregation pathways, fragmentation, and ultimately, a detrimental impact on the protein's biological activity, binding affinity, and overall therapeutic efficacy. Such oxidative damage can occur even in placebo formulations containing polysorbate alone, but it is frequently exacerbated in the presence of the therapeutic protein, as the protein itself can act as a sacrificial scavenger for oxidative species, further consuming polysorbate and accelerating its degradation. The presence of trace metal ions (e.g., iron, copper) that catalyse the decomposition of polysorbate-derived hydroperoxides into highly reactive hydroxyl radicals exacerbates this protein oxidative damage⁴².


5.3. Particle Formation and Opalescence

One of the most visually apparent and therapeutically concerning consequences of polysorbate degradation, particularly hydrolysis, is the ubiquitous formation of visible and subvisible particles, and in some cases, opalescence or turbidity. These particles are predominantly composed of free fatty acids (FFAs) released from the polysorbate ester bond cleavage. FFAs, especially those with longer hydrocarbon chains (e.g., myristic (C14), palmitic (C16), stearic (C18), and oleic (C18:1) acids), possess intrinsically limited aqueous solubility. This solubility is further diminished at low temperatures (typically 2-8°C, common for biopharmaceutical storage) and at specific pH values that promote their protonated, less soluble form. The critical micelle concentration (CMC) of intact polysorbate plays a role in solubilizing FFAs; however, as intact polysorbate degrades and its concentration drops, the micellar capacity to solubilize FFAs is compromised, leading to their supersaturation and subsequent precipitation. The kinetics of FFA particle formation involve nucleation and growth phases, which are influenced by the degree of supersaturation, presence of seed particles, and formulation excipients.

The presence of particulate matter in injectable biopharmaceuticals is a major quality concern and a significant patient safety risk, as it can potentially lead to adverse immune responses or local inflammatory reactions upon administration. Regulatory bodies (e.g., FDA, EMA) have stringent limits on particulate matter in parenteral products. Furthermore, these particles are not always solely composed of FFAs; they can co-precipitate with or adsorb protein or incorporate other impurities such as silicone oil microdroplets or subvisible protein aggregates, complicating their characterization. The composition, size distribution, and morphology of these particles are critical indicators of formulation stability⁴³.


5.4. Biological Activity and Immunogenicity

While numerous studies report no significant adverse impact of polysorbate degradation, including FFA particle formation, on the in vitro biological activity of the therapeutic protein itself for certain products, concerns regarding potential in vivo safety implications and long-term efficacy persist. The rationale behind these concerns is multi-faceted:


Altered Protein Conformation/Aggregation: Polysorbate degradation can lead to protein aggregation (as discussed in 5.1 and 5.2), and protein aggregates are well-established as a major risk factor for enhanced immunogenicity. Even if the direct biological activity remains intact, the presence of aggregates can trigger an unwanted immune response.


Adjuvant-like Effects: Polysorbate degradation products, particularly FFAs and their associated particles, have been speculated to act as "danger signals" or adjuvants. These molecules, or particles formed from them, might interact with immune cells (e.g., macrophages, dendritic cells) in a manner similar to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs), thereby activating the innate immune system and potentially breaking immunological tolerance to the therapeutic protein.


Hapten Formation: Although less commonly reported, specific polysorbate degradation products could theoretically act as haptens, binding to the protein or other macromolecules to form neoantigens, thereby eliciting an adaptive immune response.


Direct Interaction with Protein: The presence of FFAs can alter the protein's hydrophobic interactions, potentially leading to partial unfolding and aggregation.

The cumulative effect of compromised protein stability, the presence of potentially reactive degradation products, and the formation of visible/subvisible particles elevates the risk of eliciting an unwanted anti-drug antibody (ADA) response, which can compromise drug efficacy and patient safety. Therefore, a thorough understanding of the relationship between polysorbate degradation and immunogenicity is paramount throughout biopharmaceutical development⁴⁴.


6. Mitigation and Control Strategies

Effectively managing polysorbate-related risks within biopharmaceutical development and manufacturing necessitates a robust and holistic control strategy. This strategy must encompass careful considerations spanning raw material sourcing and handling, sophisticated manufacturing process design, rational formulation development, and comprehensive analytical testing and specification setting. The aim is to minimize polysorbate degradation and its downstream impact on drug product quality, safety, and efficacy⁴⁵.


6.1. Raw Material Control and Handling

The quality of incoming polysorbate raw material is foundational to mitigating future degradation issues.


Supplier and Batch Qualification: Implementing stringent qualification programmes for polysorbate suppliers is paramount. This involves not only ensuring adherence to compendial standards (e.g., USP, Ph. Eur., JP) but also establishing stricter internal specifications for critical impurities. Key parameters to monitor include peroxide value (PV), initial free fatty acid (FFA) content, aldehyde levels, and comprehensive fatty acid composition (to assess variations in feedstock purity). A risk-based assessment of suppliers and their manufacturing processes can help identify and qualify sources providing "fit-for-purpose" grades, such as low-peroxide or ultra-pure polysorbates, which are specifically manufactured to minimize pro-oxidant impurities. Monitoring batch-to-batch variability through Certificates of Analysis (CoAs) and in-house testing is essential.


Storage and Handling: Proper storage conditions for neat polysorbate raw materials are crucial to minimize oxidative degradation. Polysorbates, particularly PS80 due to its unsaturated fatty acid content, are susceptible to autoxidation upon exposure to oxygen, light, and elevated temperatures. Therefore, storage at refrigerated temperatures (e.g., 2-8°C), protection from direct light exposure (e.g., using amber containers or light-protected storage areas), and storage under an inert atmosphere (e.g., nitrogen or argon overlay, especially for bulk containers) are critical measures. The practice of avoiding long-term storage of diluted polysorbate stock solutions and instead preparing fresh solutions immediately prior to manufacturing is highly recommended to limit degradation kinetics in the aqueous phase.


Stabilization of Raw Material: In some instances, the incorporation of specific antioxidants (e.g., butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), or tocopherols) directly into the polysorbate raw material by the supplier has been explored. These antioxidants can act as radical scavengers, thereby inhibiting the initiation and propagation of oxidative degradation cascades and subsequently reducing the liberation of FFAs from hydrolysis linked to oxidative stress.


6.2. Process Development and Control

Manufacturing processes can significantly influence polysorbate stability and protein product quality.


Host Cell Protein (HCP) Reduction: If enzymatic hydrolysis is identified as a primary root cause of polysorbate degradation, a key mitigation strategy involves optimizing the downstream purification process. This includes selecting and optimizing chromatography steps (e.g., ion-exchange, hydrophobic interaction chromatography) and ultrafiltration/diafiltration (UF/DF) steps to effectively remove residual lipases, phospholipases, and other relevant esterase-active HCPs. Advanced analytical methods such as mass spectrometry-based proteomics can be employed to identify and quantify specific problematic HCPs, guiding targeted removal strategies.


Minimize Adsorption Losses: Polysorbates can adsorb to various surfaces within the manufacturing train, including filters, tubing, storage bags, and bioreactors, leading to a reduction in their effective concentration. This "loss" can render the formulation under-surfactant and susceptible to interfacial stresses. Mitigation involves careful selection of process equipment materials (e.g., specific grades of stainless steel, single-use plastics), optimizing equipment sizing, implementing effective flushing protocols, and controlling in-process hold times. Monitoring polysorbate content at critical manufacturing steps through mass balance studies is vital to ensure its adequate concentration throughout the process.


Control of Process Parameters: Manufacturing process parameters, such as pH, temperature, and agitation rates during mixing, filling, and filtration, must be tightly controlled. Suboptimal conditions can accelerate the degradation of polysorbate or protein aggregation. For example, excessive mechanical stress can disrupt polysorbate micelles, exposing proteins to interfaces, while localized pH excursions can influence enzymatic hydrolysis⁴⁶,⁴⁷.


6.3. Formulation Design

Strategic formulation development offers substantial opportunities to enhance polysorbate stability and overall product integrity.


Polysorbate Selection and Concentration: The choice of polysorbate type (PS20 vs. PS80) and its optimal concentration is critical. PS80 is generally preferred for highly concentrated antibody formulations due to its superior solubilizing capacity and protective effects, while PS20 may be considered where oxidative stability is paramount, given its predominantly saturated fatty acid profile. The optimal concentration must be determined through comprehensive experimental studies, including accelerated stability studies, agitation stress tests, and forced degradation studies, to ensure adequate protection against protein aggregation without introducing new risks. While using an excess of polysorbate might intuitively seem beneficial to compensate for degradation, excessively high concentrations can sometimes lead to other issues, such as increased micelle size, enhanced foaming, or even direct interactions with certain proteins. Design of Experiments (DoE) approaches are valuable for optimizing these parameters.


Excipient Compatibility: Careful consideration of potential interactions between polysorbate and other excipients is essential. Certain buffer components (e.g., histidine) or trace metal ions from excipients can catalyse polysorbate degradation. The selection of chelating agents (e.g., EDTA) can mitigate metal-catalysed oxidation. Maintaining an optimal pH range is crucial, as both very low and very high pH can influence polysorbate degradation rates. Ionic strength can also affect micelle formation and stability.


Antioxidant Inclusion: Incorporating appropriate antioxidants directly into the final formulation is a common strategy to mitigate oxidative degradation of both the polysorbate and the therapeutic protein. Common antioxidants include methionine (which can act as a sacrificial scavenger for ROS), histidine, and potent radical scavengers. The choice and concentration of antioxidant must be carefully evaluated to ensure compatibility with the therapeutic protein, efficacy in the specific formulation matrix, and to avoid any unintended interactions or direct impact on the protein's stability or activity.


pH and Ionic Strength Optimization: Fine-tuning the formulation's pH and ionic strength can significantly impact the kinetics of both hydrolytic and oxidative polysorbate degradation, as well as the solubility of potential FFA degradation products. For example, maintaining a pH where FFAs are more ionized can improve their solubility, delaying precipitation⁴⁸.


6.4. Testing and Specifications

Rigorous analytical testing throughout development and lifecycle management is fundamental to controlling polysorbate-related risks.


Stability Monitoring: Comprehensive stability studies under both accelerated and real-time conditions are imperative. This includes routine monitoring of intact polysorbate content and its degradation products (e.g., FFA levels for hydrolysis, peroxide value, and specific oxidized species for oxidation) using sensitive and stability-indicating analytical methods. The analytical toolbox described in Section 4 is crucial for this. Forced degradation studies are also vital to elucidate degradation pathways and confirm the stability-indicating nature of the assays.


Impact Assessment: The analytical data on polysorbate degradation must be directly correlated with critical protein quality attributes. This involves evaluating the impact on protein aggregation (soluble and particulate), protein oxidation (e.g., methionine oxidation, tryptophan degradation), protein fragmentation, charge variants, and biological activity. If particles are observed, their nature (e.g., FFA, proteinaceous, silicone) must be thoroughly characterized. Multi-variate statistical analysis can help identify correlations between polysorbate degradation and changes in protein attributes.


Control Strategy Definition: Based on a thorough understanding of polysorbate stability, its degradation pathways, and its impact on drug product quality, a robust control strategy must be defined in accordance with regulatory guidelines (e.g., ICH Q8 for Pharmaceutical Development, Q9 for Quality Risk Management, Q10 for Pharmaceutical Quality System). This strategy may involve establishing:


Raw Material Specifications: Strict release specifications for incoming polysorbate, including limits for peroxides and FFAs.


In-Process Controls (IPCs): Monitoring polysorbate concentration and critical process parameters during manufacturing.


Finished Product Specifications: Setting acceptance criteria for polysorbate content, limits for specific degradation products (e.g., FFAs), and protein quality attributes at release and throughout the shelf-life.

Risk Mitigation Plans: Implementing specific strategies (e.g., improved raw material storage, HCP removal, antioxidant addition) where polysorbate degradation poses a significant risk to product quality.

A proactive and integrated approach to polysorbate management, encompassing careful raw material selection, process optimization, rational formulation design, and robust analytical control, is essential to ensure the long-term stability, safety, and efficacy of biopharmaceutical products¹,⁴⁷,⁴⁹.


Table 1 Comparative Analysis of Polysorbates and Promising Alternative Excipients for Therapeutic Protein Stabilization ⁵,⁶

Excipient Class (Examples)

Primary Mechanism of Protein Stabilization

Key Advantages over Polysorbates (PS)

Key Limitations & Further Research Needed

Polysorbates (PS 20, PS 80)

Protects proteins by preferential adsorption to interfaces

(air–water, solid–water, oil–water), competing with proteins for adsorption sites. Thought to protect against interfacial damage.

Established history of safe parenteral use and widespread adoption in biopharmaceutical formulations. Most widely used surfactants, found in approximately 70% of marketed therapeutic antibodies.

Chemically diverse mixtures prone to degradation by oxidation and hydrolysis, producing peroxides and fatty acids. Degradation products can induce protein oxidation, aggregation, and insoluble particle formation. Increasing concerns about degradation impact on safety and quality. Immunogenicity concerns related to degradation products and the POE moiety. Heterogeneous mixtures with batch-to-batch variability.

Hydroxypropyl Beta-Cyclodextrins (HPβCD) (KLEPTOSE® HP, HPB, HPB-LB)

Reduces protein–protein interactions by shielding hydrophobic interactions. Displaces proteins from interfaces due to its weak surface activity. Can stabilize through direct interaction with the protein, especially those with substantial solvent exposure to hydrophobic amino-acid residues.

Exhibits greater chemical stability under various stress conditions, making it a more stable excipient. More effective in protecting monoclonal antibodies from light, agitation, and thermal stresses. May be considered an effective alternative to polysorbates for protein stabilization. Can also reduce PS enzymatic degradation when used in combination with PS20.

While HPβCD has weak surface activity compared to classical surfactants, it can displace protein from interfaces. Further studies are warranted to fully elucidate its stabilization mechanism on proteins. Potential susceptibility to enzymatic degradation by glycoside hydrolase family 13 enzymes (mostly bacterial origin). May also be susceptible to oxidative cleavage. When combined with PSs, it might disturb established analytical methods for PSs.

Sucrose Fatty Acid Esters (SEs), Sugar Monoesters

Surface activity; expected to protect protein integrity through direct interaction and competitive interface adsorption.

Biodegradable and biocompatible due to cleavable ester bond. Less susceptible to oxidative degradation compared to PSs. Lack of POE moiety decreases risk of immunogenic/hypersensitivity reactions.

Susceptible to chemical and enzymatic cleavage of the ester bond and enzymatic degradation of the sugar moiety. Safety for intravenous administration in humans requires more profound toxicological studies. Minimum required concentration for protein stabilization needs investigation. The issue of hydrolysis remains. Current literature does not provide direct comparative data on protein stabilization efficiency against PS.

PEG Stearates & PEG Fatty Esters (e.g., Kolliphor® HS 15, EL)

Acts through interfacial adsorption and direct interaction with proteins. Kolliphor® HS 15 has efficient solubilization properties.

Biodegradable due to the ester bond. Kolliphor® HS 15 has shown a superior safety and toxicity profile compared to PS80 in animal models.

Susceptible to hydrolysis of the ester bond and oxidation of the POE moiety. The rate of degradation and the potential influence of degradation products on protein stability needs more investigation. Potential interactions with primary packaging are yet to be fully investigated. PEG-mediated immunogenicity and hypersensitivity are general concerns for PEG-containing excipients.

Block Polyethylene-Propylene Glycols (Poloxamers® P188, P407)

Protects proteins through direct interaction and competitive interfacial adsorption.

Not susceptible to Host Cell Protein (HCP)-mediated degradation due to the absence of an ester bond. P188 has shown superior performance in maintaining syringe functionality.

Block Polyethylene-Propylene Glycols (Poloxamers® P188, P407) Protects proteins through direct interaction and competitive interfacial adsorption. Not susceptible to Host Cell Protein (HCP)-mediated degradation due to the absence of an ester bond. P188 has shown superior performance in maintaining syringe functionality. P188 can degrade in the solid state at high temperatures (starting ~40°C) due to auto-oxidation and chain cleavage, resulting in aldehydes, organic acids, and smaller polymers. P188 has lower stability in liquid formulations compared to PS surfactants. Can lead to the formation of molecule-specific visible and sub-visible protein–polydimethylsiloxane (PDMS) particles in liquid mAb formulations when stored with siliconized stoppers. Chemical stability in solution has not been thoroughly investigated. Immunological concerns from the POE moiety persist.

Polyoxyethylene Fatty Ethers (Brij® 35, 58, 92)

Provides protein stabilization, likely through interfacial adsorption and direct interaction.

Absence of an ester bond makes them less susceptible to enzymatic hydrolysis. Brij® 58 showed better inherent stability in protein formulations compared to PS20 and PS80.

Susceptible to metal- and photo-induced oxidation (ether moieties), similar to PSs. Safety of POE ethers for parenteral administration in humans needs further investigation. Stability in protein biological formulations and the influence of degradation products need profound research. Immunological concerns originating from the POE moiety must be addressed. Parenteral approval for Brij® is still needed.

Non-Ester Sugar-Based Surfactants (e.g., n-dodecyl-β-D-maltoside)

Reduces the surface tension of water. Acts as a cryoprotectant.

Non-mutagenic, non-toxic, non-irritating, biodegradable, and biocompatible. No ester bond, so resistant to esterase-mediated hydrolysis.

Susceptible to enzymatic degradation of the sugar moiety. Its use in parenteral protein therapeutics remains underexploited. The stability and potential degradation pathways of these surfactants in protein formulations need further investigation.

N-alkyl Amino Acid Polyether Amides (e.g., N-myristoyl phenylalanine Jeffamine M1000 diamide (FM1000))

Designed to combine amino acid stabilization with non-ester surfactant properties. Resistant to HCP-mediated hydrolysis due to amide bonds. Facilitates stabilization of air/water and air/silicon interfaces.

Resistant to HCP-mediated hydrolysis. Low batch-to-batch variability due to chemically defined structure. Highly effective at lower concentrations (CMC 10 times lower than PS80). Demonstrated superior or comparable IgG protection from agitation-mediated stress compared to PS80 and PS20. Significantly slowed thermal-induced aggregation (3-fold more reduction of protein particle formation compared to PS).

The amide bond is potentially susceptible to hydrolysis, though to a significantly lower extent compared to the ester bond in PS surfactants. Its industrial translation requires more in-depth investigations regarding potential pharmacological and toxicological side effects. Current available stability studies are from the manufacturer, which may imply potential for bias.

Disaccharides (Sucrose, Trehalose)

Act as osmolytes, altering solvent properties and reducing protein–solvent interactions. Surround protein molecules to reduce protein–protein interactions. Well-known cryo- and lyoprotectants.

Safe, non-toxic, and readily available. GRAS (Generally Recognized as Safe) status. Can provide a basis for chemical modification to enhance surface-active properties.

Inadequate protein protection against interface-induced stresses when used alone. Susceptible to acid hydrolysis, creating reducing sugars, which can damage proteins (necessitating pH control). Often used in combination with PS surfactants. Reducing sugars like lactose or maltose should be avoided due to Maillard reaction risk.

Sugar Alcohols (Glycerol, Mannitol, Sorbitol)

Primarily act as osmolytes, protecting protein integrity by altering protein–solvent interactions. Replace water molecules during lyophilization. Glycerol inhibits protein unfolding and aggregation by stabilizing aggregation-prone intermediates and arranging at the interface of hydrophobic protein regions and polar solvent.

Safe and have appropriate regulatory status (GRAS, FDA-approved). Widely employed in various pharmaceutical formulations.

Often cannot provide adequate protein protection from all sources of stress, particularly interface-induced stresses, necessitating combination with other excipients. Mannitol crystallization can detrimentally impact protein stability. Sorbitol also prone to re-crystallization over time. Higher doses can lead to side effects (e.g., hyperglycaemia, renal failure, haemolysis for glycerol; hypersensitivity for mannitol).

Amino Acids (e.g., Arginine, Histidine, Lysine)

Inhibit protein aggregation and promote protein refolding. Act as neutral osmo-protective molecules, altering protein–solvent interactions and serving as molecular crowders. Arginine reduces viscosity in high concentration solutions.

Safe and FDA-approved for parenteral administration. Not prone to degradation under typical formulation and storage conditions. Arginine can significantly decrease viscosity of high concentration protein solutions.

The effect of amino acids upon protein stability is protein-dependent. The complexity of their effect upon different proteins requires individual optimization of type and concentration for each specific protein formulation.

Natural Polyamines (e.g., Spermidine, Spermine, Putrescine)

Low molecular weight aliphatic polycations that prevent heat-induced aggregation and inactivation of proteins. Provide a hydrophobic environment that interferes with protein hydrophobic association, preventing aggregation.

Naturally occurring molecules. Can effectively prevent protein aggregation and inactivation under thermal stress.

Stability in high concentration liquid formulations (HCLF) needs further investigation. No specific degradation profile in formulations reported. Ability to prevent protein inactivation and particle formation in HCLF needs further investigation. Safety and tolerability for intravenous administration requires more detailed investigation due to reported hypotension in animal studies.

Albumin

The most prevalent plasma protein effectively stabilizes other proteins by competitively inhibiting protein adsorption to interfaces. Chemical structure enables protection through hydrophobic and electrostatic interactions and intermolecular structure formation. Also has antioxidant properties.

Natural and well-established excipient. Has antioxidant properties. Recombinant albumin addresses the risk of blood-transmitted diseases. Safe for parenteral use; non-toxic and non-irritant properties.

Potentially prone to forming aggregates with itself or other proteins, especially in IFN formulations, which can lead to immunogenicity concerns. Concerns about blood-transmitted diseases for human/bovine serum albumin. Exact formulation composition of commercially available recombinant albumin is rarely disclosed.

Synthetic Amphiphilic Polymers




Polyether Polyols (PEGs, Polypropylene Glycols (PPGs))

PEGs: Reduce protein aggregation possibly through steric shielding of superficial hydrophobic patches. Short-chain PEGs can protect intracorporeally by interacting with protein surface or extending into solvent. PPGs: Protein stabilization hypothesized to be a function of direct interaction with protein molecules. PPGs possess higher surface activity than PEGs.

Modifiable chemical structure, molecular weight, and functionality. PPGs have higher surface activity than PEGs.

Potentially susceptible to auto-oxidation. PEGs and PEG-containing molecules are known to induce mild to life-threatening immunogenic and hypersensitivity reactions. Cross-reactivity between anti-PEG antibodies and PPGs has been observed. Efficiency of free PEGs to reduce monomer loss or particle generation is inferior to PS surfactants. PPGs' stability and immunogenicity require thorough investigation.

Polyampholytes (Sulfobetaine-based)

A subclass of zwitterionic materials that contain opposite charges on different monomer subunits. Suppresses thermal aggregation by suppressing aggregation-induced collision within misfolded protein hydrophobic domains.

Zwitterionic nature provides unique properties for protein stabilization. Shows promising protein particle formation inhibition at elevated temperatures.

Chemical stability is largely unknown. Safety and suitability for parenteral biotherapeutic formulations remain largely uncovered. Their chemical stability, safety, and suitability for parenteral biotherapeutic formulations are largely unknown and need to be investigated.

Ionic Liquids (ILs)

Organic salts with highly tunable properties. Can improve protein extraction, separation, crystallization, solubilization, and stabilization. Can induce refolding and renaturation of chemically unfolded proteins.

Properties are highly tunable based on anion/cation combinations. Can be used as co-solvents in aqueous solutions. Generally recognized for their high stability.

Their toxicity and biocompatibility in humans are under-investigated. Toxicity depends on the type of IL, cell line, length of the alkyl chain, and size of the anion. No FDA approval for parenteral protein formulations currently. Controversial effects (stabilizing or destabilizing) depending on protein type, IL concentration, anion charge density, and alkyl chain length. Requires extensive screening of potential protein–ionic liquid pairs. High concentrations may lead to hypertonicity issues for parenteral administration.

7. Stability Challenges of Other Advanced Biologic Therapeutics:

While the preceding sections have focused extensively on the critical role and stability challenges of polysorbates primarily within therapeutic protein formulations, particularly monoclonal antibodies (mAbs) and fusion proteins, it is imperative to acknowledge that the broader landscape of advanced biologic therapeutics presents a diverse array of unique and significant stability hurdles. Understanding these challenges provides a richer context for the pervasive nature of formulation science in biopharmaceuticals and highlights shared principles, even if the direct application of polysorbates varies across these platforms.


7.1. Gene Therapy (Viral Vectors)

Viral vectors, such as adeno-associated viruses (AAV) and adenoviruses (AdV), are fundamental to gene delivery. Their stability is multifaceted and critical for maintaining infectivity and transduction efficiency. Challenges include:


Physical Instability: Adsorption to surfaces (e.g., glassware, plastic containers) and subsequent aggregation of viral particles, which can reduce infectivity and potentially elicit immunogenicity.

Chemical Degradation: Deamidation of capsid proteins, oxidation of amino acid residues, and hydrolysis of viral DNA/RNA within the capsid, all of which can compromise structural integrity and functional efficacy.

Shear Stress: Vulnerability to mechanical forces during manufacturing (e.g., filtration, pumping) leading to capsid damage. Stabilization strategies often involve optimizing excipient profiles, including specific sugars (e.g., sucrose, trehalose) and polyols (e.g., mannitol, sorbitol) for cryo- and lyophilization protection, and optimizing pH and ionic strength. While polysorbates are sometimes included in viral vector formulations, their role often focuses on mitigating adsorption to surfaces and preventing aggregation, similar to their function with proteins⁵⁰,⁵¹.


7.2. Gene Therapy (Non-Viral Methods, e.g., Lipid Nanoparticles - LNPs)

Non-viral gene delivery systems, predominantly lipid nanoparticles (LNPs) for nucleic acid (e.g., mRNA, siRNA) encapsulation, face distinct stability issues crucial for effective cargo delivery and in vivo performance. Key challenges include:


Physical Stability: Colloidal instability, leading to particle aggregation, fusion, or fission, which can alter particle size distribution and subsequent biodistribution.

Chemical Stability: Oxidation of unsaturated lipids within the LNP structure, leading to lipid degradation products that can compromise membrane integrity and cargo leakage. Hydrolysis of ionizable lipids can also occur.

Cargo Integrity: Protection of the encapsulated nucleic acid from enzymatic degradation (e.g., by nucleases) and chemical hydrolysis within the LNP. Formulation strategies focus on optimizing lipid composition (e.g., choice of ionizable lipid, helper lipids, cholesterol, and PEGylated lipids), manufacturing processes (e.g., microfluidic mixing for controlled self-assembly), and storage conditions (e.g., lyophilization) to enhance thermal and colloidal stability⁵²,⁵³.


7.3. Cell Therapy

Maintaining the viability, functionality, and genomic integrity of living cells is the paramount stability challenge throughout the complex cell therapy workflow, from cell culture and genetic manipulation to final formulation, storage, and patient delivery. Key considerations include:


Cell Viability and Potency: Susceptibility to shear stress, osmotic shock, temperature fluctuations, and metabolic stress during processing and storage.

Genomic and Phenotypic Stability: Risk of genomic instability (e.g., mutations) or unintended changes in cell phenotype and differentiation state during prolonged culture or storage.


Batch-to-Batch Variability: Significant inherent variability in cellular products due to biological complexity. Cryopreservation, utilizing cryoprotectants like dimethyl sulfoxide (DMSO) or trehalose, is a common strategy to extend shelf-life. However, cryoprotectant toxicity and cell recovery remain challenges. Rigorous process controls, advanced process analytical technologies (PAT), and comprehensive quality testing (e.g., cell viability, purity, potency assays) are essential⁵⁴,⁵⁵.


7.4. mRNA-Based Therapies

Messenger RNA (mRNA) molecules are intrinsically fragile due to their phosphodiester backbone and complex secondary/tertiary structures.

Inherent mRNA Instability: Susceptibility to enzymatic degradation by ubiquitous ribonucleases (RNases) and chemical hydrolysis of phosphodiester bonds.


LNP-Mediated Stability: For mRNA-LNP formulations, challenges include maintaining the colloidal stability of the LNPs themselves (as described above for gene therapy LNPs) and ensuring the sustained protection of the mRNA cargo from degradation both within the LNP and during in vivo delivery. Mitigation strategies involve structural and chemical modifications of the mRNA (e.g., 5' cap analogues, poly(A) tail optimization, pseudouridine incorporation to enhance stability and translation, codon optimization), and precise optimization of LNP composition and manufacturing (e.g., microfluidic mixing for controlled particle size, lyophilization to enhance thermal stability)⁵⁶,⁵⁷.


7.5. Monoclonal Antibodies (mAbs)

As the most mature class of biopharmaceuticals, mAbs still present diverse stability challenges, for which polysorbates are a critical excipient.


Conformational Instability: Unfolding or misfolding of the protein, often initiated by interfacial stresses (e.g., air-liquid, silicone oil-liquid), leading to exposure of hydrophobic regions.


Colloidal Instability: Self-association and irreversible aggregation (soluble and particulate), driven by non-covalent interactions (e.g., hydrophobic, electrostatic) or covalent cross-linking (e.g., disulphide scrambling).


Chemical Degradation: Specific chemical modifications including deamidation (especially at asparagine and glutamine residues), oxidation (primarily methionine, tryptophan, histidine), isomerization (aspartate), disulphide bond cleavage or scrambling, and glycation. Factors such as pH, temperature, ionic strength, and mechanical stress profoundly influence these degradation pathways. Polysorbates are specifically incorporated to mitigate conformational and colloidal instability by adsorbing to interfaces, thereby preventing protein adsorption and subsequent aggregation, highlighting their direct relevance to these challenges. Other excipients, such as buffers, salts, and viscosity modifiers, also play crucial roles⁵⁸,⁵⁹.


7.6. Antibody-Drug Conjugates (ADCs)

ADCs represent a sophisticated class of biologics that combine the specificity of mAbs with the potency of small molecule cytotoxic payloads via chemical linkers. Their stability concerns are complex and multi-layered, encompassing those of the mAb component, the linker, and the payload.


mAb Component Stability: Similar challenges to unconjugated mAbs, including aggregation, deamidation, and oxidation.

Linker Stability: Premature release of the highly potent payload due to linker instability (e.g., hydrolysis, enzymatic cleavage by non-target enzymes) is a major concern for safety and efficacy.

Payload Stability: Degradation (e.g., oxidation, hydrolysis) and aggregation of the small molecule payload, which can be highly hydrophobic and prone to self-association.

Conjugation Site and Drug-to-Antibody Ratio (DAR): These factors significantly influence overall ADC heterogeneity, stability, and pharmacokinetic/pharmacodynamic profiles. Mitigation strategies involve careful optimization of conjugation chemistry, designing highly stable and selectively cleavable linkers (e.g., peptide-based, disulphide-based, non-cleavable), employing site-specific conjugation methods to control DAR and heterogeneity, and optimizing formulation to enhance solubility and prevent aggregation of the hydrophobic payload⁶⁰,⁶¹.


7.7. Fusion Proteins

These engineered proteins, which combine distinct protein domains (e.g., an antibody Fc region fused to a receptor or enzyme), share structural similarities with mAbs but often possess increased complexity and unique interfaces. Consequently, they often face comparable stability issues, including aggregation, fragmentation, and structural breakdown, sometimes exacerbated by the novel interfaces created by the fusion event. Like mAbs, their stabilization heavily relies on comparable formulation strategies and the strategic use of excipients, including polysorbates, to combat interfacial stress-induced degradation.

In conclusion, while the specific chemical and physical vulnerabilities differ across these diverse advanced biologic therapeutics, the fundamental imperative to understand and control their stability remains universal. The principles of formulation optimization, rigorous analytical characterization, and robust process controls are consistently applied. Moreover, the role of excipients, including the critical function of surfactants like polysorbates in mitigating interfacial-stress induced degradation, remains a cornerstone of ensuring the long-term quality, safety, and efficacy of a wide spectrum of biopharmaceutical products⁶²,⁶³.


Conclusion

Polysorbates, encompassing PS20 and PS80, are unequivocally indispensable excipients in the formulation of a vast array of therapeutic protein products. Their amphiphilic nature and ability to adsorb to interfaces provide critical protection against myriad mechanical and interfacial stresses, thereby safeguarding protein integrity, mitigating aggregation, and preserving biological activity throughout the product lifecycle. However, as this comprehensive review has underscored, the inherent molecular heterogeneity of commercially available polysorbates, coupled with their susceptibility to diverse degradation pathways—predominantly hydrolysis and oxidation—presents complex and persistent quality challenges that demand rigorous attention. The formation of free fatty acids (FFAs), along with other oxidative and hydrolytic degradation products, can lead to a demonstrable loss of polysorbate functional capacity, induce direct oxidative modifications to the therapeutic protein, and, notably, trigger the formation of undesirable visible and subvisible particles, each with significant implications for product quality, safety, and regulatory compliance.

The accurate and comprehensive analytical characterization of polysorbate content, composition, and their resulting degradation products in complex biopharmaceutical matrices is paramount for a thorough understanding of the extent and underlying mechanisms of polysorbate degradation. As elaborated, a sophisticated and multifaceted analytical toolbox is available, ranging from highly sensitive, targeted assays for specific degradation markers (e.g., FFAs, peroxides, aldehydes) to advanced hyphenated chromatographic and mass spectrometric techniques (e.g., LC-MS, HPLC-CAD/ELSD, GC-MS, NMR) for detailed compositional profiling and structural elucidation. Despite these analytical advancements, challenges persist in standardizing quantitative methodologies across different laboratories and, crucially, in developing robust functional assays that directly and reliably correlate with polysorbate performance in situ within the final drug product. The dynamic interplay between intact polysorbate, its degradation products, and the therapeutic protein necessitates a nuanced understanding of their combined impact on surface activity and protein stability.

Effective control strategies for mitigating polysorbate-related risks must therefore be systematically integrated across every stage of the product lifecycle, from early development to commercial manufacturing and post-market surveillance. This demands rigorous qualification and appropriate handling protocols for polysorbate raw materials, emphasizing the importance of low-peroxide and low-aldehyde grades and controlled storage conditions. Furthermore, manufacturing processes must be meticulously optimized to minimize critical contaminants (e.g., host cell protein lipases and esterases) and to prevent polysorbate losses due to adsorption. Formulation design necessitates careful consideration of excipient compatibility, pH, and ionic strength, along with the judicious inclusion of antioxidants where oxidative degradation is a concern. Finally, comprehensive stability testing, employing a suite of sensitive and stability-indicating analytical methods, is indispensable for monitoring polysorbate integrity and its impact on critical quality attributes of the drug product. The ultimate level of control, including the establishment of formal specifications for polysorbate content and degradation products, must be predicated upon a thorough, risk-based assessment of their impact on product quality and patient safety.

While this review has predominantly focused on the polysorbate challenges within protein formulations, it is crucial to recognize that the broader landscape of advanced biologic therapeutics—including gene therapy viral vectors, lipid nanoparticles for nucleic acid delivery, cell therapies, mRNA-based therapies, antibody-drug conjugates, and fusion proteins—also confronts complex and diverse stability challenges. These often necessitate tailored stabilization and control strategies, yet frequently share fundamental principles of formulation science, including the critical role of interfacial stabilization.

Looking forward, continued research is indispensable to advance our understanding and control of polysorbate stability. Specific areas for future investigation include:

Deepening the understanding of polysorbate heterogeneity: A more comprehensive characterization of individual isomers and their specific degradation kinetics and impact.

Mechanistic insights: Further elucidation of the precise molecular mechanisms by which polysorbate degradation products (e.g., specific aldehydes, short-chain FFAs, altered POE fragments) interact with and destabilize therapeutic proteins.

Advanced Analytical Tools: Development of novel, high-throughput, and directly functional analytical methods that can truly predict polysorbate performance in vivo.

Predictive Modelling: Leveraging computational and machine learning approaches to predict polysorbate stability and protein-polysorbate interactions based on structural characteristics and formulation parameters.

Alternative Stabilizers: Exploration and development of novel, more stable, and well-defined alternative excipients that can provide equivalent or superior stabilization with reduced degradation liabilities.

Ultimately, ensuring the consistent stability and functional integrity of polysorbates is not merely an excipient-centric concern; it is a critical determinant for maintaining the consistent quality, established safety, and robust efficacy of a wide and ever-expanding range of biopharmaceutical products, directly impacting patient outcomes worldwide.


References


1. A. Martos, W. Koch, W. Jiskoot, K. Wuchner, G. Winter, W. Friess, A. Hawe, Trends on Analytical Characterization of Polysorbates and Their Degradation Products in Biopharmaceutical Formulations, J Pharm Sci 106 (2017). https://doi.org/10.1016/j.xphs.2017.03.001.

2. M.T. Jones, H.C. Mahler, S. Yadav, D. Bindra, V. Corvari, R.M. Fesinmeyer, K. Gupta, A.M. Harmon, K.D. Hinds, A. Koulov, W. Liu, K. Maloney, J. Wang, P.Y. Yeh, S.K. Singh, Considerations for the Use of Polysorbates in Biopharmaceuticals, Pharm Res 35 (2018). https://doi.org/10.1007/s11095-018-2430-5.

3. M. Dwivedi, M. Blech, I. Presser, P. Garidel, Polysorbate degradation in biotherapeutic formulations: Identification and discussion of current root causes, Int J Pharm 552 (2018). https://doi.org/10.1016/j.ijpharm.2018.10.008.

4. F. Bensaid, C. Dagallier, J.R. Authelin, H. Audat, V. Filipe, C. Narradon, P. Guibal, S. Clavier, P. Wils, Mechanistic understanding of metal-catalyzed oxidation of polysorbate 80 and monoclonal antibody in biotherapeutic formulations, Int J Pharm 615 (2022). https://doi.org/10.1016/j.ijpharm.2022.121496.

5. A.J. Castañeda Ruiz, M.A. Shetab Boushehri, T. Phan, S. Carle, P. Garidel, J. Buske, A. Lamprecht, Alternative Excipients for Protein Stabilization in Protein Therapeutics: Overcoming the Limitations of Polysorbates, Pharmaceutics 14 (2022). https://doi.org/10.3390/pharmaceutics14122575.

6. H. Zhang, S. Hong, S.S.K. Tan, T. Peng, L.Y.H. Goh, K.H. Lam, K.T. Chow, R. Gokhale, Polysorbates versus Hydroxypropyl Beta-Cyclodextrin (HPβCD): Comparative Study on Excipient Stability and Stabilization Benefits on Monoclonal Antibodies, Molecules 27 (2022). https://doi.org/10.3390/molecules27196497.

7. E.V. Brovč, J. Mravljak, R. Šink, S. Pajk, Degradation of polysorbates 20 and 80 catalysed by histidine chloride buffer, European Journal of Pharmaceutics and Biopharmaceutics 154 (2020). https://doi.org/10.1016/j.ejpb.2020.07.010.

8. B. Kozuch, J. Weber, J. Buske, K. Mäder, P. Garidel, T. Diederichs, Comparative Stability Study of Polysorbate 20 and Polysorbate 80 Related to Oxidative Degradation, Pharmaceutics 15 (2023). https://doi.org/10.3390/pharmaceutics15092332.

9. J. Penfold, R.K. Thomas, P.X. Li, J.T. Petkov, I. Tucker, J.R.P. Webster, A.E. Terry, Adsorption at Air-Water and Oil-Water Interfaces and Self-Assembly in Aqueous Solution of Ethoxylated Polysorbate Nonionic Surfactants, Langmuir 31 (2015). https://doi.org/10.1021/acs.langmuir.5b00151.

10. S. Deechongkit, J. Wen, L.O. Narhi, Y. Jiang, S.S. Park, J. Kim, B.A. Kerwin, Physical and biophysical effects of polysorbate 20 and 80 on darbepoetin alfa, J Pharm Sci 98 (2009). https://doi.org/10.1002/jps.21740.

11. M.A. Pegues, K. Szczepanek, F. Sheikh, S.G. Thacker, B. Aryal, M.K. Ghorab, S. Wolfgang, R.P. Donnelly, D. Verthelyi, V.A. Rao, Effect of Fatty Acid Composition in Polysorbate 80 on the Stability of Therapeutic Protein Formulations, Pharm Res 38 (2021). https://doi.org/10.1007/s11095-021-03125-6.

12. M. De Pra, D.A. Ispan, S. Meding, T. Müllner, K.S. Lovejoy, S. Grosse, K. Cook, S. Carillo, F. Steiner, J. Bones, Degradation of polysorbate investigated by a high-performance liquid chromatography multi-detector system with charged aerosol and mass detection, J Chromatogr A 1710 (2023). https://doi.org/10.1016/j.chroma.2023.464405.

13. T. Okada, Automated analyses of non-ionic surfactants in terms of hydrophobic and polyoxyethylated chain lengths, J Chromatogr A 609 (1992). https://doi.org/10.1016/0021-9673(92)80165-Q.

14. A. V. Nagornyi, M. V. Avdeev, O.I. Ivankov, Y.Y. Shlapa, S.O. Solopan, T. V. Nagorna, A. V. Shulenina, Y.L. Zabulonov, A.G. Belous, L.A. Bulavin, Structural Stability of Dispersions of Magnetic Nanoparticles in Aqueous Solutions of Polysorbate-80, Journal of Surface Investigation 15 (2021). https://doi.org/10.1134/S1027451021040339.

15. Y. Guttormsen, A. Moldes-Anaya, R. Fjellaksel, A. Oteiza, M. Martin-Armas, M. Lindemann, J. Bihler, B.H. Hoff, S. Jakobsen, E. Sundby, R. Sundset, M. Kranz, Development of a carbon-11 labeled thienopyrimidine based radioligand for EGFR-tyrosine-kinase-inhibitor (TKI) specific PET imaging of glioblastoma, Journal of Nuclear Medicine 62 (2021).

16. X. Zheng, A.T. Sutton, R.S. Yang, D. V. Miller, B. Pagels, R.R. Rustandi, J. Welch, A. Payne, M. Haverick, Extensive Characterization of Polysorbate 80 Oxidative Degradation Under Stainless Steel Conditions, J Pharm Sci 112 (2023). https://doi.org/10.1016/j.xphs.2022.10.012.

17. M. Dwivedi, J. Buske, F. Haemmerling, M. Blech, P. Garidel, Acidic and alkaline hydrolysis of polysorbates under aqueous conditions: Towards understanding polysorbate degradation in biopharmaceutical formulations, European Journal of Pharmaceutical Sciences 144 (2020). https://doi.org/10.1016/j.ejps.2019.105211.

18. A.C. Bhargava, K. Mains, A. Siu, J. Gu, J. Zarzar, L. Yi, I.H. Yuk, High-Throughput, Fluorescence-Based Esterase Activity Assay for Assessing Polysorbate Degradation Risk during Biopharmaceutical Development, Pharm Res 38 (2021). https://doi.org/10.1007/s11095-021-03011-1.

19. D. Kovner, I.H. Yuk, A. Shen, H. Li, T. Graf, S. Gupta, W. Liu, A. Tomlinson, Characterization of Recombinantly-Expressed Hydrolytic Enzymes from Chinese Hamster Ovary Cells: Identification of Host Cell Proteins that Degrade Polysorbate, J Pharm Sci 112 (2023). https://doi.org/10.1016/j.xphs.2023.01.003.

20. S. Dahotre, A. Tomlinson, B. Lin, S. Yadav, Novel markers to track oxidative polysorbate degradation in pharmaceutical formulations, J Pharm Biomed Anal 157 (2018). https://doi.org/10.1016/j.jpba.2018.05.031.

21. A. Schmidt, A. Koulov, J. Huwyler, H.C. Mahler, M. Jahn, Stabilizing Polysorbate 20 and 80 Against Oxidative Degradation, J Pharm Sci 109 (2020). https://doi.org/10.1016/j.xphs.2020.03.003.

22. J. Weber, J. Buske, K. Mäder, P. Garidel, T. Diederichs, Oxidation of polysorbates – An underestimated degradation pathway?, Int J Pharm X 6 (2023). https://doi.org/10.1016/j.ijpx.2023.100202.

23. K.S.K. Ravuri, Polysorbate degradation and quality, in: AAPS Advances in the Pharmaceutical Sciences Series, 2018. https://doi.org/10.1007/978-3-319-90603-4_2.

24. E.V. Brovč, S. Pajk, R. Šink, J. Mravljak, Comparison of the NMR and the acid value determination methods for quality control of input polysorbates, Acta Chim Slov 66 (2019). https://doi.org/10.17344/acsi.2019.5144.

25. O. V. Borisov, J.A. Ji, Y.J. Wang, F. Vega, V.T. Ling, Toward understanding molecular heterogeneity of polysorbates by application of liquid chromatography-mass spectrometry with computer-aided data analysis, Anal Chem 83 (2011). https://doi.org/10.1021/ac2005789.

26. S. Hoelterhoff, J. Wendler, L. Arackal, B. Felkel, C.H. Bell, A. Bathke, High throughput FAMS – A fatty acid mass spectrometry method for monitoring polysorbate hydrolysis in QC, J Chromatogr B Analyt Technol Biomed Life Sci 1220 (2023). https://doi.org/10.1016/j.jchromb.2023.123614.

27. S.S. Zhi Chen, A Highly Sensitive Method for the Quantitation of Polysorbate 20 and 80 to Study the Compatibility between Polysorbates and m-Cresol in the Peptide Formulation, J Anal Bioanal Tech 06 (2015). https://doi.org/10.4172/2155-9872.1000245.

28. K.A. Carnes, L.D. Oliver, T.A. Brown, R.F. Delgadillo, M.S. Ward, S. Mortazavi, M.J. Morris, J.W. Shearer, J.S. Fuller, A Platform analytical method for intact polysorbates in protein-containing biopharmaceutical products via HPLC-CAD, J Liq Chromatogr Relat Technol 45 (2022). https://doi.org/10.1080/10826076.2023.2207024.

29. N. Shende, A. Karale, S. Bhagade, A. Gulhane, P. Bore, P. Marathe, S. Chakraborty, A.D. Mallya, R.M. Dhere, Evaluation of a sensitive GC–MS method to detect polysorbate 80 in vaccine preparation, J Pharm Biomed Anal 183 (2020). https://doi.org/10.1016/j.jpba.2020.113126.

30. R.S.K. Kishore, A. Pappenberger, I.B. Dauphin, A. Ross, B. Buergi, A. Staempfli, H.C. Mahler, Degradation of polysorbates 20 and 80: Studies on thermal autoxidation and hydrolysis, J Pharm Sci 100 (2011). https://doi.org/10.1002/jps.22290.

31. R. Zhang, Y. Wang, L. Tan, H.Y. Zhang, M. Yang, Analysis of polysorbate 80 and its related compounds by RP-HPLC with ELSD and MS detection, J Chromatogr Sci 50 (2012). https://doi.org/10.1093/chromsci/bms035.

32. S. Lippold, S.H.S. Koshari, R. Kopf, R. Schuller, T. Buckel, I.E. Zarraga, H. Koehn, Impact of mono- and poly-ester fractions on polysorbate quantitation using mixed-mode HPLC-CAD/ELSD and the fluorescence micelle assay, J Pharm Biomed Anal 132 (2017). https://doi.org/10.1016/j.jpba.2016.09.033.

33. K. Morker, D. Purohit, R. Patel, PLGA based nanomaterials in Drug Delivery for cancer treatment: Innovations and Emerging Applications, Int J of Pharm Sci, 3 (2025). https://doi.org/10.5281/zenodo.15064506

34. C.C. Siska, C.J. Pierini, H.R. Lau, R.F. Latypov, R.M. Fesinmeyer, J.R. Litowski, Free fatty acid particles in protein formulations, Part 2: Contribution of polysorbate raw material, J Pharm Sci 104 (2015). https://doi.org/10.1002/jps.24144.

35. M. Khossravi, Y.H. Kao, R.J. Mrsny, T.D. Sweeney, Analysis methods of polysorbate 20: A new method to assess the stability of polysorbate 20 and established methods that may overlook degraded polysorbate 20, Pharm Res 19 (2002). https://doi.org/10.1023/A:1015306112979.

36. H.L. Svilenov, K.T. Kopp, A.P. Golovanov, G. Winter, M. Zalar, Insights into the Stabilization of Interferon Alpha by Two Surfactants Revealed by STD-NMR Spectroscopy, J Pharm Sci 112 (2023). https://doi.org/10.1016/j.xphs.2022.10.013.

37. H.C. Mahler, R. Müller, W. Frieß, A. Delille, S. Matheus, Induction and analysis of aggregates in a liquid IgG1-antibody formulation, European Journal of Pharmaceutics and Biopharmaceutics 59 (2005). https://doi.org/10.1016/j.ejpb.2004.12.004.

38. N. Sinha, A.S. Joshi, A.K. Thakur, Analytical validation of an ATR-FTIR based method for quantifying the amount of polysorbate 80 adsorbed on PLGA nanoparticles, Analytical Methods 12 (2020). https://doi.org/10.1039/d0ay01685c.

39. A.S. Joshi, A. Gahane, A.K. Thakur, Deciphering the mechanism and structural features of polysorbate 80 during adsorption on PLGA nanoparticles by attenuated total reflectance-Fourier transform infrared spectroscopy, RSC Adv 6 (2016). https://doi.org/10.1039/c6ra07699h.

40. F. Begum, S. Amin, Investigating the Influence of Polysorbate 20/80 and Polaxomer P188 on the Surface & Interfacial Properties of Bovine Serum Albumin and Lysozyme, Pharm Res 36 (2019). https://doi.org/10.1007/s11095-019-2631-6.

41. X. Li, F. Wang, H. Li, D.D. Richardson, D.J. Roush, The measurement and control of high-risk host cell proteins for polysorbate degradation in biologics formulation, Antib Ther 5 (2022). https://doi.org/10.1093/abt/tbac002.

42. H. Liu, Y. Jin, R. Menon, E. Laskowich, L. Bareford, P. de Vilmorin, D. Kolwyck, B. Yeung, L. Yi, Characterization of Polysorbate 80 by Liquid Chromatography-Mass Spectrometry to Understand Its Susceptibility to Degradation and Its Oxidative Degradation Pathway, J Pharm Sci 111 (2022). https://doi.org/10.1016/j.xphs.2021.08.017.

43. N. Glücklich, S. Carle, T. Diederichs, J. Buske, K. Mäder, P. Garidel, How enzymatic hydrolysis of polysorbate 20 influences colloidal protein stability, European Journal of Pharmaceutical Sciences 191 (2023). https://doi.org/10.1016/j.ejps.2023.106597.

44. C. L. Bennett, B. Chen, T. Hermanson, M. D. Wyatt, R. M. Schulz, P. Georgantopoulos, S. Kessler, D. W. Raisch, Z. P. Qureshi, Z. K. Lu, B. L. Love, V. Noxon, L. Bobolts, M. Armitage, J. Bian, P. Ray, R. J. Ablin, W. J. Hrushesky, I. C. Macdougall, O. Sartor, J. O. Armitage, Regulatory and clinical considerations for biosimilar oncology drugs, The Lancet Oncology 15 (2014). https://doi.org/10.1016/S1470-2045(14)70365-1.

45. T. Tsukidate, A.Q. Stiving, S. Mengisen, W.S. McKechnie, R. Carrillo, X. Li, Heat Inactivation of Host Cell–Derived Enzymes as a Control Strategy for Polysorbate Degradation, J Pharm Sci 113 (2024). https://doi.org/10.1016/j.xphs.2023.10.038.

46. M.N. Nassar, V.N. Nesarikar, R. Lozano, W.L. Parker, Y. Huang, V. Palaniswamy, W. Xu, N. Khaselev, Influence of formaldehyde impurity in polysorbate 80 and PEG-300 on the stability of a parenteral formulation of BMS-204352: Identification and control of the degradation product, Pharm Dev Technol 9 (2004). https://doi.org/10.1081/PDT-120030249.

47. K. Wuchner, L. Yi, C. Chery, F. Nikels, F. Junge, G. Crotts, G. Rinaldi, J.A. Starkey, K. Bechtold-Peters, M. Shuman, M. Leiss, M. Jahn, P. Garidel, R. de Ruiter, S.M. Richer, S. Cao, S. Peuker, S. Huille, T. Wang, V. Le Brun, Industry Perspective on the Use and Characterization of Polysorbates for Biopharmaceutical Products Part 2: Survey Report on Control Strategy Preparing for the Future, J Pharm Sci 111 (2022). https://doi.org/10.1016/j.xphs.2022.08.021.

48. J. Zhang, J. He, K.J. Smith, Fatty Acids Can Induce the Formation of Proteinaceous Particles in Monoclonal Antibody Formulations, J Pharm Sci 111 (2022). https://doi.org/10.1016/j.xphs.2021.10.008.

49. M.N. Honemann, J. Wendler, T. Graf, A. Bathke, C.H. Bell, Monitoring polysorbate hydrolysis in biopharmaceuticals using a QC-ready free fatty acid quantification method, J Chromatogr B Analyt Technol Biomed Life Sci 1116 (2019). https://doi.org/10.1016/j.jchromb.2019.03.030.

50. M.A. Croyle, X. Cheng, J.M. Wilson, Development of formulations that enhance physical stability of viral vectors for gene therapy, Gene Ther 8 (2001). https://doi.org/10.1038/sj.gt.3301527.

51. R. Mohammad, Key considerations in formulation development for gene therapy products, Drug Discov Today 27 (2022). https://doi.org/10.1016/j.drudis.2021.08.013.

52. A.M. Carvalho, R.A. Cordeiro, H. Faneca, Silica-based gene delivery systems: From design to therapeutic applications, Pharmaceutics 12 (2020). https://doi.org/10.3390/pharmaceutics12070649.

53. R. Rai, S. Alwani, I. Badea, Polymeric nanoparticles in gene therapy: New avenues of design and optimization for delivery applications, Polymers (Basel) 11 (2019). https://doi.org/10.3390/polym11040745.

54. J.L. Dashnau, Q. Xue, M. Nelson, E. Law, L. Cao, D. Hei, A risk-based approach for cell line development, manufacturing and characterization of genetically engineered, induced pluripotent stem cell–derived allogeneic cell therapies, Cytotherapy 25 (2023). https://doi.org/10.1016/j.jcyt.2022.08.001.

55. J.K. Jadlowsky, R. Leskowitz, S. McKenna, J. Karar, Y. Ma, A. Dai, G. Plesa, F. Chen, K. Alexander, J. Petrella, N. Gong, W.T. Hwang, O. Farrelly, J. Barber-Rotenberg, S. Christensen, V.E. Gonzalez, A. Chew, J.A. Fraietta, C.H. June, Long-term stability of clinical-grade lentiviral vectors for cell therapy, Mol Ther Methods Clin Dev 32 (2024). https://doi.org/10.1016/j.omtm.2024.101186.

56. R.G. Ingle, W.J. Fang, An Overview of the Stability and Delivery Challenges of Commercial Nucleic Acid Therapeutics, Pharmaceutics 15 (2023). https://doi.org/10.3390/pharmaceutics15041158.

57. Y. Weng, C. Li, T. Yang, B. Hu, M. Zhang, S. Guo, H. Xiao, X.J. Liang, Y. Huang, The challenge and prospect of mRNA therapeutics landscape, Biotechnol Adv 40 (2020). https://doi.org/10.1016/j.biotechadv.2020.107534.

58. Y. Le Basle, P. Chennell, N. Tokhadze, A. Astier, V. Sautou, Physicochemical Stability of Monoclonal Antibodies: A Review, J Pharm Sci 109 (2020). https://doi.org/10.1016/j.xphs.2019.08.009.

59. R. Wada, M. Matsui, N. Kawasaki, Influence of N-glycosylation on effector functions and thermal stability of glycoengineered IgG1 monoclonal antibody with homogeneous glycoforms, MAbs 11 (2019). https://doi.org/10.1080/19420862.2018.1551044.

60. C.M. Mckertish, V. Kayser, Advances and limitations of antibody drug conjugates for cancer, Biomedicines 9 (2021). https://doi.org/10.3390/biomedicines9080872.

61. J. He, Q. Duan, C. Ran, T. Fu, Y. Liu, W. Tan, Recent progress of aptamer‒drug conjugates in cancer therapy, Acta Pharm Sin B 13 (2023). https://doi.org/10.1016/j.apsb.2023.01.017.

62. G. Carta, A. Jungbauer, Protein Chromatography, 2020. https://doi.org/10.1002/9783527824045.

63. E. Morimoto, K. Tsuboyama, Y. Tomari, Fusion with heat-resistant obscure (Hero) proteins have the potential to improve the molecular property of recombinant proteins, PLoS One 17 (2022). https://doi.org/10.1371/journal.pone.0270097.

Author Information


Authors: Krupal Morker¹, Ravi Patel², Dipen Purohit³


¹Frontage Laboratories Inc.

²Thermofisher Scientific, 5900 M.L.K. Jr. Hwy, Greenville, NC 27834, USA

³Navinta LLC, 1499 Lower Ferry Road Ewing, NJ 08618, USA


Corresponding Author:

Krupal Morker, Frontage Laboratories Inc.

 
 
 
bottom of page