Atomfair Brainwave Hub: Nanomaterial Science and Research Primer / Characterization Techniques for Nanomaterials / Dynamic light scattering for nanoparticle sizing
Dynamic light scattering (DLS) has become an indispensable tool in the pharmaceutical industry for characterizing nanoscale drug delivery systems. The technique provides critical information about particle size distribution, aggregation behavior, and stability of colloidal systems, making it particularly valuable for liposomes, polymeric nanoparticles, and protein aggregates used in therapeutic applications. Its non-invasive nature, rapid analysis time, and ability to measure samples in their native state have established DLS as a standard method for quality control and formulation development.

In liposome characterization, DLS measures the hydrodynamic diameter of vesicles and detects the presence of aggregates or structural changes. Liposomal formulations require precise control over size distribution as it directly impacts drug loading efficiency, circulation time, and biodistribution. For example, DLS data demonstrated that Doxil, the first FDA-approved liposomal drug, maintains a consistent size range of 80-100 nm, which is essential for its enhanced permeability and retention effect in tumor tissues. Batch-to-batch consistency monitoring with DLS ensures that liposomes meet the narrow size specifications required for clinical efficacy. Stability studies using DLS track size increases over time, providing early warning signs of vesicle fusion or drug leakage.

Polymeric nanoparticle formulations benefit from DLS analysis during both development and manufacturing stages. The technique identifies subtle changes in particle size that may indicate polymer degradation, drug crystallization, or surface modification issues. In the case of Abraxane, an albumin-bound paclitaxel nanoparticle formulation, DLS confirmed the maintenance of sub-200 nm particle size during scale-up processes. The technology also plays a crucial role in monitoring the stability of polymeric micelles, where small changes in hydrodynamic diameter can signal premature drug release or micelle dissociation. Accelerated stability testing protocols frequently incorporate DLS measurements to predict shelf life by correlating particle growth rates with temperature-dependent degradation kinetics.

Protein-based therapeutics present unique characterization challenges that DLS effectively addresses. For monoclonal antibody formulations and protein-drug conjugates, DLS detects early stages of aggregation that could lead to reduced efficacy or immunogenic responses. The analysis of Adynovate, a PEGylated factor VIII product, included DLS measurements to verify the absence of protein aggregates in the final formulation. In biosimilar development, DLS provides comparative data between originator and biosimilar products, ensuring equivalent higher-order structure and aggregation profiles.

The pharmaceutical industry relies on DLS for stability-indicating studies that form the basis of shelf-life determinations. By measuring the z-average diameter and polydispersity index at regular intervals under various storage conditions, researchers can model degradation kinetics and establish expiration dates. Real-time stability studies of Onpattro, a lipid nanoparticle-based RNA therapeutic, incorporated DLS data to demonstrate maintenance of nanoparticle integrity over 24 months at recommended storage temperatures. These studies often employ Arrhenius kinetics, where DLS data collected at elevated temperatures predicts long-term behavior under normal storage conditions.

Batch release testing represents another critical application of DLS in pharmaceutical manufacturing. Regulatory agencies require demonstration of particle size consistency across production batches, with tight specifications typically set within ±10% of the target diameter. For Vyxeos, a liposomal daunorubicin and cytarabine combination, DLS analysis forms part of the quality control protocol to ensure uniform drug encapsulation and vesicle size distribution. The technique's high throughput capability allows for rapid screening of multiple batches during production scale-up.

Case studies of FDA-approved nanomedicines highlight DLS's role in product development and regulatory approval. In the development of Marqibo, a vincristine sulfate liposome injection, DLS data supported the demonstration of bioequivalence between clinical trial materials and commercial-scale products. Similarly, for Copaxone, a complex mixture of polypeptide nanoparticles, DLS measurements contributed to the physicochemical characterization required for regulatory filings. The technology's ability to detect subtle formulation differences proved crucial during the development of generic versions of nanomedicines, where DLS data helped establish equivalence in critical quality attributes.

Advanced DLS applications in pharmaceutical development include the characterization of complex nanostructures such as hybrid nanoparticles and multicomponent systems. For combination products containing both small molecule drugs and biologics, DLS monitors the integrity of each component and their interactions. The analysis of drug-loaded exosomes or extracellular vesicles represents an emerging application where DLS provides essential quality control parameters. These measurements become particularly important when scaling up production from laboratory to clinical or commercial scales.

The selection of appropriate measurement parameters significantly impacts DLS data quality in pharmaceutical applications. Sample preparation techniques, including filtration methods and dilution protocols, require optimization for each formulation type. Temperature control during measurements proves critical for temperature-sensitive formulations, while proper viscosity corrections ensure accurate size determinations for high-concentration protein solutions. The development of standardized protocols for specific nanomedicine classes continues to improve inter-laboratory reproducibility of DLS measurements.

Recent technological advancements in DLS instrumentation have expanded its pharmaceutical applications. Multi-angle detection systems improve accuracy for polydisperse samples, while automated plate readers enable high-throughput screening of formulation libraries. The integration of DLS with other characterization techniques, such as electrophoretic light scattering for zeta potential measurements, provides comprehensive nanoparticle characterization in a single platform. These developments support the growing complexity of nanomedicine formulations entering clinical development.

The pharmaceutical industry's increasing adoption of continuous manufacturing processes creates new opportunities for DLS implementation. In-line or at-line DLS systems enable real-time monitoring of nanoparticle characteristics during production, facilitating immediate process adjustments. This approach aligns with quality-by-design principles and supports the development of more robust manufacturing processes. For sensitive biologics, such monitoring can detect early signs of aggregation or particle formation that might compromise product quality.

Regulatory expectations for nanoparticle characterization continue to evolve, with DLS remaining a core analytical technique in submission packages. The technology's ability to provide quantitative data on critical quality attributes supports the demonstration of product consistency and manufacturing control. As nanomedicine pipelines expand to include increasingly sophisticated delivery systems, DLS will maintain its essential role in ensuring the quality, safety, and efficacy of these complex pharmaceutical products. The continued refinement of DLS methodologies and data analysis protocols will further enhance its value in nanomedicine development and commercialization.
Back to Dynamic light scattering for nanoparticle sizing