Advancements in Lipid Nanoparticle-Based Delivery Systems for Nucleic Acids: Structure, Mechanisms, and Therapeutic Applications

Abstract

Lipid nanoparticles (LNPs) have revolutionized the landscape of nucleic acid delivery, serving as indispensable carriers for messenger RNA (mRNA), small interfering RNA (siRNA), plasmid DNA, and even CRISPR-Cas components. Their pivotal role in overcoming the inherent fragility and cellular impermeability of these biomolecules has unlocked unprecedented therapeutic avenues, most notably in the rapid development of mRNA-based vaccines and emerging gene therapies. This comprehensive report meticulously dissects the multifaceted aspects of LNPs, commencing with a detailed exposition of their intricate structural components and their synergistic contributions to nanoparticle functionality. Subsequently, it elucidates the sophisticated mechanisms governing nucleic acid encapsulation, systemic circulation, cellular internalization, and the crucial process of endosomal escape that liberates the genetic cargo into the cytosol. A substantial portion is dedicated to recent advancements in LNP design, exploring sophisticated strategies for enhancing their physicochemical stability, refining their targeting specificity to precise cell populations or organs, and optimizing their pharmacokinetic and pharmacodynamic profiles. Furthermore, the report critically evaluates the evolving understanding of LNP safety profiles, including considerations of immunogenicity and potential toxicity, and the innovative approaches being pursued to mitigate these challenges. Special emphasis is placed on the expansive therapeutic applications of LNPs, particularly their transformative potential in areas such as cardiac regeneration, targeted oncology, rare genetic disorders, and the prevention and treatment of infectious diseases, highlighting their indispensable position in the future of precision medicine.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

1. Introduction

The trajectory of modern medicine has been profoundly influenced by the quest for effective and safe methods to deliver therapeutic agents directly to diseased cells or tissues. In the realm of genetic medicine, this challenge is particularly acute, as nucleic acids, such as mRNA, siRNA, and DNA, are inherently large, negatively charged, and susceptible to rapid degradation by nucleases in biological fluids. Moreover, their hydrophilic nature prevents their passive diffusion across the hydrophobic lipid bilayer of cell membranes. For decades, viral vectors served as the primary vehicles for gene delivery due to their natural ability to infect cells and introduce genetic material. However, concerns regarding immunogenicity, potential insertional mutagenesis, and manufacturing complexities spurred intensive research into non-viral alternatives [1].

The emergence of messenger RNA (mRNA) as a therapeutic modality has represented a paradigm shift. Unlike DNA-based therapies, mRNA-based approaches do not require nuclear entry for gene expression, avoiding risks of genomic integration. They offer unparalleled advantages in terms of rapid development and manufacturing scalability, making them highly adaptable to emerging public health crises, as evidenced by their pivotal role during the COVID-19 pandemic [2]. mRNA therapeutics function by transiently delivering genetic instructions to cells, enabling the cellular machinery to produce a desired protein (e.g., an antigen for vaccination, an enzyme for replacement therapy, or a therapeutic protein). The transient nature of mRNA expression also contributes to a favorable safety profile compared to gene therapies involving permanent genomic alterations.

Central to the successful realization of mRNA-based therapies, and indeed many other nucleic acid-based interventions, is the development of robust and efficient delivery systems. Among the myriad non-viral vectors explored, lipid nanoparticles (LNPs) have unequivocally emerged as the gold standard. Their ability to encapsulate fragile nucleic acids, protect them from enzymatic degradation, facilitate their transport across biological barriers, and enable their efficient delivery into the cytoplasm of target cells has been a game-changer. This report aims to provide a comprehensive and detailed analysis of LNPs, bridging fundamental principles with cutting-edge advancements, to illustrate their critical role in transforming the landscape of modern medicine and their profound potential for future therapeutic applications.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2. Structural Components of Lipid Nanoparticles

Lipid nanoparticles are intricate nanocarriers, typically ranging from 50 to 150 nm in diameter, assembled from a precisely balanced mixture of four primary lipid components. The judicious selection and quantitative ratio of these components are paramount, dictating the LNP’s overall stability, size, surface charge, cargo encapsulation efficiency, cellular uptake mechanisms, and ultimately, its therapeutic efficacy and safety profile. The synergistic interplay of these lipids allows LNPs to overcome the myriad biological barriers encountered during systemic delivery and intracellular processing [2, 7].

2.1. Ionizable Lipids

Ionizable lipids constitute the cornerstone of LNP design, serving as the primary functional component responsible for nucleic acid encapsulation and endosomal escape. Unlike permanently cationic lipids, which carry a positive charge regardless of pH and often exhibit significant cytotoxicity due to indiscriminate membrane interactions, ionizable lipids possess a pH-dependent charge [5]. At an acidic pH (typically around 4.0-6.0), such as that encountered during LNP formulation or within endosomes, their amine headgroups become protonated, acquiring a positive charge. This positive charge facilitates strong electrostatic interactions with the negatively charged phosphate backbone of nucleic acids (e.g., mRNA, siRNA, DNA), enabling efficient encapsulation into the LNP core during the self-assembly process.

Conversely, at physiological pH (approximately 7.4) in the bloodstream, these lipids are largely neutral or only partially protonated. This near-neutral surface charge at physiological pH is crucial for minimizing non-specific interactions with plasma proteins, reducing complement activation, and mitigating systemic toxicity and immunogenicity [6]. Once the LNP is internalized by endocytosis into acidic endosomes (pH 5.0-6.0), the ionizable lipids become reprotonated. This re-protonation leads to a significant increase in the LNP’s positive charge, triggering a cascade of events critical for endosomal escape. These events include the ‘proton sponge effect,’ where the protonated lipids buffer the endosomal lumen, causing an influx of protons and counter-ions (e.g., chloride ions), which leads to osmotic swelling and subsequent rupture of the endosomal membrane, thereby releasing the nucleic acid cargo into the cytoplasm [2].

The design of ionizable lipids is a highly active area of research, focusing on optimizing their pKa (the pH at which half of the lipid molecules are protonated). An ideal ionizable lipid strikes a delicate balance: a pKa sufficiently low to be neutral at physiological pH but high enough to become protonated in the endosome. Early examples include DLin-MC3-DMA, which was crucial for ONPATTRO®, the first LNP-siRNA drug. More recently, lipids like ALC-0315 (used in Pfizer/BioNTech’s COVID-19 vaccine) and SM-102 (used in Moderna’s COVID-19 vaccine) represent advanced generations designed for improved efficacy and safety profiles, characterized by their rapid biodegradability to reduce potential accumulation and long-term toxicity [7]. The alkyl chains of these lipids also play a significant role, influencing the fluidity and stability of the LNP membrane and their interactions with cellular membranes.

2.2. Phospholipids

Phospholipids are amphipathic molecules, possessing a hydrophilic head and a hydrophobic tail, which are fundamental to the structural integrity and stability of biological membranes. In LNPs, phospholipids, typically helper lipids such as 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) or dioleoylphosphatidylethanolamine (DOPE), contribute to the formation of the lipid bilayer that envelops the nucleic acid core. DSPC, a saturated phospholipid, contributes to the rigidity and structural stability of the LNP, helping to maintain its shape and protect the encapsulated cargo. Its relatively high phase transition temperature ensures that the LNP remains stable at physiological temperatures.

DOPE, an unsaturated phospholipid with a conical shape, is often incorporated to facilitate endosomal escape. Its propensity to form non-bilayer, hexagonal II phases under specific conditions (e.g., acidic pH within endosomes) can induce membrane curvature and destabilization, promoting fusion with the endosomal membrane and subsequent release of the nucleic acid into the cytoplasm. This ‘helper’ function is critical for overcoming the endosomal barrier, which is a major bottleneck in non-viral gene delivery [2]. The precise ratio of phospholipids to other LNP components influences the LNP’s packing density, membrane fluidity, and overall architectural stability.

2.3. Cholesterol

Cholesterol, a ubiquitous sterol in eukaryotic cell membranes, is an indispensable component in most LNP formulations, typically comprising a significant percentage of the total lipid content. Its unique rigid planar ring structure and flexible hydrocarbon tail allow it to intercalate between the fatty acyl chains of other lipids, profoundly influencing membrane properties. In LNPs, cholesterol plays several critical roles:

  • Structural Rigidity and Stability: Cholesterol enhances the mechanical stability and rigidity of the LNP membrane. This increased rigidity contributes to the overall robustness of the nanoparticle, protecting the encapsulated nucleic acid from enzymatic degradation and premature release in the bloodstream [2].
  • Membrane Fluidity Modulation: Cholesterol acts as a ‘fluidity buffer,’ reducing the phase transition temperature of phospholipids and broadening the phase transition range. This allows the LNP membrane to maintain an optimal balance of rigidity and fluidity across a range of temperatures, which is crucial for stability during storage and circulation, as well as for facilitating cellular interactions.
  • Packing and Encapsulation Efficiency: By filling gaps between lipid molecules, cholesterol improves the packing of the lipid bilayer, which can enhance the efficiency of nucleic acid encapsulation and retention within the LNP core [2].
  • Cellular Uptake and Endosomal Escape: Cholesterol has been implicated in facilitating interactions with cell membranes and, in some contexts, promoting endosomal escape, possibly by influencing membrane curvature or by aiding the fusion process [9].

Variations in cholesterol concentration can significantly impact LNP size, polydispersity, stability, and delivery efficiency, making its optimization a critical aspect of LNP formulation. Research is also exploring cholesterol analogs, such as β-sitosterol, which has shown promise in improving LNP stability, particularly for applications requiring resilience to physical stress, such as nebulization for inhalation therapies [9].

2.4. Polyethylene Glycol (PEG)-Lipids

Polyethylene glycol (PEG)-lipids are hydrophilic polymers conjugated to a lipid anchor that inserts into the LNP membrane. They are typically present in small molar percentages (around 0.5-2%) but exert a disproportionately significant influence on LNP performance. The primary function of PEG-lipids is to provide a steric barrier on the LNP surface, creating a ‘stealth’ effect. This hydrophilic surface coating significantly reduces the adsorption of opsonin proteins from the bloodstream, thereby minimizing recognition and rapid clearance by the mononuclear phagocyte system (MPS), particularly Kupffer cells in the liver and macrophages in the spleen [2]. This ‘steric stabilization’ effect dramatically prolongs the circulation half-life of LNPs, allowing more time for them to reach target tissues. Common PEG-lipids include PEG-DMG (used in Moderna’s vaccine) and PEG2000-DSPE.

However, the use of PEG-lipids is not without its challenges, collectively referred to as the ‘PEG dilemma.’ Repeated administration of PEGylated nanoparticles can induce the formation of anti-PEG antibodies, which accelerate the clearance of subsequent doses through the accelerated blood clearance (ABC) phenomenon, reducing therapeutic efficacy and potentially triggering hypersensitivity reactions [6]. This phenomenon highlights the trade-off between extended circulation time and potential immunogenicity, especially for chronic therapies requiring multiple doses. Researchers are actively pursuing strategies to mitigate the ABC phenomenon, including the use of biodegradable PEG-lipids that detach from the LNP surface over time, lower PEG concentrations, or alternative stealth polymers like zwitterionic lipids or biocompatible polypeptides that do not elicit a strong immune response [10]. The molecular weight of the PEG chain also plays a role, with PEG2000 (2 kDa) being a commonly optimized length.

2.5. Nucleic Acid Cargo

While not a structural component of the LNP, the nucleic acid cargo is the reason d’être of the LNP. The type of nucleic acid significantly influences LNP design and performance.

  • Messenger RNA (mRNA): mRNA is a single-stranded molecule that carries genetic information from DNA to ribosomes, where it is translated into proteins. Therapeutic mRNA is typically modified to enhance its stability (e.g., incorporation of pseudouridine or 5-methylcytidine to reduce immunogenicity and improve translation efficiency), improve translation initiation (optimized 5′ cap), and prolong its half-life (optimized poly(A) tail). Its relatively large size (thousands of nucleotides) and negative charge necessitate efficient encapsulation by ionizable lipids [2, 7].
  • Small Interfering RNA (siRNA): siRNA is a double-stranded RNA molecule, typically 20-25 base pairs long, that functions in RNA interference (RNAi) by sequence-specifically silencing gene expression. Due to its smaller size, siRNA can sometimes be delivered with slightly different LNP formulations compared to mRNA, though the fundamental principles remain similar.
  • Plasmid DNA (pDNA): pDNA is a circular, double-stranded DNA molecule used for gene expression. While LNPs can deliver pDNA, its larger size and the requirement for nuclear entry for transcription make its delivery more challenging compared to mRNA.
  • CRISPR-Cas Components: LNPs are increasingly used to deliver CRISPR-Cas components, either as mRNA encoding Cas nucleases (e.g., Cas9) and guide RNA (gRNA), or as ribonucleoprotein (RNP) complexes (pre-formed Cas protein and gRNA). Delivering mRNA/gRNA avoids genomic integration of the Cas gene itself, enhancing safety, while RNP delivery allows for immediate activity and shorter duration of Cas protein presence, further minimizing off-target effects.

2.6. LNP Formation Process (Brief Overview)

LNPs are typically formed using rapid mixing techniques, most commonly microfluidics or ethanol injection. In these methods, a solution of lipids dissolved in an organic solvent (e.g., ethanol) is rapidly mixed with an aqueous solution containing the negatively charged nucleic acid at an acidic pH (e.g., citrate buffer at pH 4.0). The rapid decrease in solvent polarity and the electrostatic interactions between the protonated ionizable lipids and the nucleic acid drive the spontaneous self-assembly of the LNP structure. The precise control offered by microfluidics allows for highly reproducible and monodisperse LNP populations, which is critical for therapeutic consistency [2]. After assembly, the pH is typically raised to physiological levels (pH 7.4) to deprotonate the ionizable lipids on the LNP surface, minimizing aggregation and toxicity.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3. Mechanisms of Nucleic Acid Encapsulation and Delivery

The successful journey of a nucleic acid delivered by an LNP from the injection site to its functional activity within the target cell is a complex, multi-step process involving intricate biophysical and cellular interactions. Understanding each stage is paramount for rational LNP design and optimization [2, 7].

3.1. Encapsulation and Self-Assembly

The encapsulation of nucleic acids within LNPs is a self-assembly process driven primarily by electrostatic interactions and hydrophobic forces. The process typically begins by mixing an ethanol solution of the lipid components (ionizable lipid, helper phospholipid, cholesterol, PEG-lipid) with an aqueous solution of the nucleic acid at an acidic pH, usually between pH 3.0 and 5.0. At this low pH, the ionizable lipids are predominantly protonated and thus positively charged.

As the ethanol diffuses out and the pH approaches the pKa of the ionizable lipid, the positively charged lipid headgroups strongly interact with the negatively charged phosphate backbone of the nucleic acid molecules (e.g., mRNA). This charge-charge interaction facilitates the condensation of the nucleic acid. Concurrently, the hydrophobic tails of the lipids aggregate to minimize their exposure to the aqueous environment, leading to the formation of a distinct core-shell structure. The nucleic acid is sequestered within an aqueous core, surrounded by a lipid monolayer or bilayer, with the PEG-lipids forming a hydrophilic corona on the outermost surface. The specific LNP architecture (e.g., inverted micelle, lamellar, or unilamellar vesicle) formed depends on the lipid composition, their ratios, the N/P ratio (nitrogen-to-phosphate ratio, representing the molar ratio of positively charged lipid amines to negatively charged nucleic acid phosphates), and the self-assembly conditions [2]. Optimal encapsulation efficiency (the percentage of nucleic acid successfully loaded into LNPs) is typically achieved when the N/P ratio is slightly above 1, ensuring sufficient positive charge to condense the nucleic acid while minimizing excess positive charge that could lead to aggregation or toxicity.

3.2. Circulation and Biodistribution

Following systemic administration (e.g., intravenous injection), LNPs enter the bloodstream. The presence of PEG-lipids on the LNP surface creates a hydrophilic ‘stealth’ layer, which sterically hinders the adsorption of plasma proteins (opsonins) onto the nanoparticle surface. This anti-fouling property is critical for preventing rapid recognition and clearance by the mononuclear phagocyte system (MPS), particularly macrophages in the liver and spleen [2]. By avoiding opsonization, PEGylated LNPs can circulate for extended periods, typically several hours, allowing sufficient time to reach target tissues.

While PEGylation extends circulation, LNPs administered intravenously tend to passively accumulate in organs with a highly fenestrated vasculature, such as the liver and spleen. The liver, with its extensive sinusoidal capillaries and resident Kupffer cells (a type of macrophage), is a primary site of LNP uptake and clearance. This inherent tropism to the liver has made LNPs exceptionally effective for treating liver-related diseases, such as those requiring gene silencing (e.g., hereditary transthyretin amyloidosis) or protein replacement (e.g., hemophilia). However, for applications targeting other organs, this liver accumulation represents an off-target effect that needs to be minimized or redirected. The size of the LNP also influences biodistribution; smaller LNPs (e.g., <100 nm) generally exhibit longer circulation times and better penetration into tissues compared to larger ones [2].

3.3. Cellular Uptake

LNPs are predominantly taken up by target cells via endocytosis, an active cellular process where the cell membrane invaginates to engulf extracellular material. Several endocytic pathways can be involved, depending on the LNP’s surface properties, size, and the target cell type:

  • Clathrin-mediated Endocytosis: This is a major pathway for cellular uptake of many macromolecules and nanoparticles. It involves the formation of clathrin-coated pits that bud off from the plasma membrane to form clathrin-coated vesicles, which then mature into early endosomes.
  • Caveolae-mediated Endocytosis: This pathway involves small, flask-shaped invaginations of the plasma membrane called caveolae, which are enriched in cholesterol and the protein caveolin. It is often associated with non-clathrin-mediated uptake and can sometimes lead to direct cytosolic release without passing through the classical endosome-lysosome pathway.
  • Macropinocytosis: A non-specific, actin-dependent process involving large membrane ruffles that engulf extracellular fluid and particles, forming large vesicles called macropinosomes. This pathway is particularly relevant for uptake by phagocytic cells like macrophages.

For non-targeted LNPs, cellular uptake is often influenced by non-specific interactions with the cell surface (e.g., electrostatic interactions, hydrophobic interactions) or recognition by scavenger receptors. For targeted LNPs, specific interactions between conjugated ligands on the LNP surface and cognate receptors on the target cell membrane drive receptor-mediated endocytosis, leading to highly specific cellular internalization [11].

3.4. Endosomal Escape

Endosomal escape is widely considered the most critical and rate-limiting step in non-viral nucleic acid delivery. Once internalized, LNPs reside within endosomes, which undergo a maturation process from early endosomes (pH ~6.0-6.5) to late endosomes (pH ~5.0-6.0) and finally to lysosomes (pH ~4.5-5.0). The acidic environment within endosomes is essential for triggering the release of the nucleic acid cargo.

The primary mechanism facilitating endosomal escape for ionizable lipid-based LNPs is the ‘proton sponge effect.’ As the pH within the endosome drops, the ionizable lipids in the LNP membrane become protonated, gaining a positive charge. This leads to an influx of protons into the endosome, which are buffered by the newly protonated lipids. To maintain electroneutrality, chloride ions (Cl-) and water concomitantly enter the endosome, causing osmotic swelling. The increasing pressure within the endosome eventually leads to its rupture, or the destabilized endosomal membrane, influenced by the protonated lipids and helper lipids like DOPE, undergoes fusion or pore formation, allowing the encapsulated nucleic acid to escape into the cytoplasm [2, 7].

Upon release into the cytosol, the nucleic acid is free to exert its therapeutic function: mRNA is translated by ribosomes into the desired protein; siRNA is loaded into the RNA-induced silencing complex (RISC) to mediate gene silencing; and DNA must further traffic to the nucleus for transcription. Inefficient endosomal escape is a major reason for the low efficacy observed with many non-viral delivery systems, as nucleic acids trapped within endosomes are typically degraded by lysosomal enzymes.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4. Advancements in LNP Design for Enhanced Stability and Targeted Delivery

The remarkable success of LNPs in mRNA vaccine delivery has spurred intensive research to further optimize their design, addressing limitations in stability, tissue specificity, and safety for a broader range of therapeutic applications. These advancements are driven by a deeper understanding of LNP biophysics and biological interactions [10].

4.1. Enhanced Stability

Maintaining the structural integrity of LNPs from manufacturing through storage, administration, and systemic circulation is paramount for therapeutic efficacy. Instability can lead to premature cargo release, aggregation, or reduced cellular uptake. Key advancements in stability include:

  • Optimized Lipid Composition: The precise ratio and chemical structures of all four lipid components significantly influence LNP stability. Novel ionizable lipids are being designed not only for improved efficacy and reduced toxicity but also for enhanced physical stability (e.g., resistance to dissociation or aggregation). Similarly, the choice of phospholipids and cholesterol analogs plays a role. For instance, incorporating cholesterol analogs like β-sitosterol has been shown to improve the stability of LNPs, particularly important for challenging delivery routes such as aerosolization for inhalation therapies. These modifications help maintain the structural integrity of LNPs during nebulization, ensuring the particles remain intact to effectively penetrate mucus and reach target cells in the lungs [9]. Furthermore, lipids with higher melting temperatures can contribute to a more rigid and stable LNP structure.

  • Manufacturing Process Refinements: Advanced manufacturing techniques, particularly continuous-flow microfluidics, allow for precise control over mixing conditions, temperature, and flow rates, enabling the production of highly monodisperse (uniform size) and stable LNP populations with high encapsulation efficiency. This consistency is crucial for reproducible therapeutic outcomes.

  • Formulation for Storage and Administration: LNPs often require cold chain storage due to their inherent instability at higher temperatures. Research into lyophilization (freeze-drying) and spray-drying methods aims to produce stable, solid LNP formulations that can be stored at higher temperatures, significantly simplifying logistics and global distribution. These processes require careful optimization of excipients (e.g., cryoprotectants like sucrose) to prevent aggregation or damage during drying and rehydration.

  • Surface Modifications beyond PEG: While PEGylation is effective for stealth, its limitations (ABC phenomenon) drive the development of alternative surface modification strategies. Zwitterionic polymers, which carry both positive and negative charges, can create a net-neutral surface that resists protein adsorption without inducing anti-PEG immune responses. Other biocompatible polymers or even specific peptide sequences are being explored to confer stability while minimizing immunogenicity.

4.2. Targeted Delivery

The ability to selectively deliver nucleic acids to specific cell types or organs while sparing others is a major goal for improving therapeutic efficacy, reducing off-target effects, and lowering required dosages. While passive targeting to the liver and spleen is inherent for systemically administered LNPs due to fenestrated capillaries and MPS uptake, active targeting strategies are essential for broader clinical applications.

  • Ligand-Mediated Targeting: This approach involves conjugating specific targeting ligands to the LNP surface. These ligands are designed to recognize and bind to cell-surface receptors that are uniquely expressed or overexpressed on target cells. Upon binding, the LNP is internalized via receptor-mediated endocytosis, leading to highly specific delivery. Examples of ligands include:

    • Antibodies: Full antibodies or antibody fragments (e.g., scFv) can be conjugated to LNPs to target specific cell surface markers. For instance, conjugating anti-CD5 antibodies to LNPs has been demonstrated to enhance delivery specifically to T cells, which is highly beneficial for immunotherapies, such as engineering T cells for cancer treatment [11]. Other examples include antibodies targeting tumor-specific antigens for cancer therapy.
    • Peptides: Short peptide sequences can be designed to bind to specific receptors or even penetrate cell membranes (cell-penetrating peptides, CPPs). Peptides offer advantages in terms of smaller size, lower immunogenicity, and easier synthesis compared to antibodies.
    • Aptamers: Nucleic acid aptamers are single-stranded DNA or RNA molecules that can bind to specific molecular targets with high affinity and specificity. They can be engineered to target a wide range of proteins on cell surfaces.
    • Small Molecules: Certain small molecules can act as ligands, recognizing specific receptors (e.g., folate for folate receptor-positive cancer cells, galactose for asialoglycoprotein receptors on hepatocytes).

    Challenges in ligand conjugation include maintaining the ligand’s binding activity, ensuring its proper orientation on the LNP surface, optimizing linker chemistry, and avoiding steric hindrance by PEG if present.

  • Selective Organ Targeting (SORT) Nanoparticles: A groundbreaking advancement in LNP design, SORT nanoparticles represent a sophisticated approach to redirect LNP biodistribution to specific organs beyond the liver. Unlike traditional ligand-mediated targeting, SORT relies on subtle modifications to the LNP’s lipid composition, particularly the precise ratios and structures of the ionizable lipid and PEG-lipid components, which intrinsically alter the LNP’s interactions with plasma proteins and cell types, thereby influencing tissue tropism [1, 3].

    Researchers have discovered that by fine-tuning these lipid components, LNPs can be engineered to preferentially accumulate in specific organs like the spleen, lungs, heart, or even the pancreas. For example, specific modifications to ionizable lipid structures, such as the introduction of certain alkyl chain lengths or branching patterns, can guide LNPs to the lung. Similarly, specific ratios of ionizable lipids and PEG-lipids can enhance uptake in the spleen, making them ideal for vaccine delivery targeting antigen-presenting cells in lymphoid organs. SORT-based LNPs have demonstrated the ability to deliver mRNA to cardiac tissue for regeneration and have shown promise for targeting the pancreas for diabetes research [1, 4]. The mechanism behind SORT is thought to involve differential interactions with apolipoproteins and other plasma factors, influencing how LNPs are recognized and internalized by various cell types in different organs [1]. This approach offers a powerful new paradigm for precision drug delivery, moving beyond the inherent liver tropism of many LNP formulations.

  • Stimuli-Responsive LNPs: These ‘smart’ nanoparticles are engineered to release their cargo only in response to specific endogenous or exogenous stimuli present at the target site. This strategy aims to improve the therapeutic index by maximizing local drug concentration and minimizing systemic exposure. Examples include:

    • pH-responsive LNPs: Designed to become unstable or release cargo more efficiently in acidic environments (e.g., tumors, inflamed tissues, endosomes).
    • Redox-responsive LNPs: Triggered by differences in reduction potential (e.g., higher glutathione levels inside cells compared to the extracellular environment).
    • Enzyme-responsive LNPs: Engineered with linkers cleavable by specific enzymes overexpressed in disease states (e.g., matrix metalloproteinases in tumors).
    • Light or Ultrasound-responsive LNPs: Externally triggered for precise spatiotemporal control of cargo release.

These advanced design principles collectively push the boundaries of LNP utility, enabling more precise, efficient, and safer nucleic acid delivery for a diverse array of therapeutic applications.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5. Safety Profile and Immunogenicity

While the transformative potential of LNPs is undeniable, particularly highlighted by their role in COVID-19 mRNA vaccines, a comprehensive understanding of their safety profile and immunogenic potential is crucial for their broader and long-term clinical application. The interaction of nanoparticles with biological systems is complex, and certain characteristics of LNPs can elicit undesirable responses [5].

5.1. Immunogenicity

Immunogenicity refers to the ability of a substance to provoke an immune response. LNPs can trigger both innate and adaptive immune responses, largely influenced by their composition, size, surface properties, and the nature of the encapsulated nucleic acid cargo.

  • Innate Immune Activation: The primary concern regarding LNP immunogenicity stems from the potential activation of innate immune sensors. The nucleic acid cargo itself, particularly mRNA, can be recognized by intracellular pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs), specifically TLR3, TLR7, and TLR8, which detect viral nucleic acids. While therapeutic mRNA is often modified (e.g., pseudouridine incorporation) to reduce its immunogenicity, some level of PRR activation can still occur, leading to the production of pro-inflammatory cytokines (e.g., IL-6, TNF-α, IFN-α/β). This controlled immune activation can be beneficial for vaccines by acting as an adjuvant, enhancing adaptive immune responses [6]. However, excessive or prolonged inflammation can lead to systemic side effects or tissue damage. The ionizable lipids themselves can also contribute to innate immune activation, potentially through membrane interactions or by activating inflammasomes (e.g., NLRP3 inflammasome), leading to pyroptosis and further cytokine release [6].

  • Adaptive Immune Responses: A significant concern, especially for repeat dosing, is the development of anti-PEG antibodies. While PEGylation prolongs LNP circulation time by preventing opsonization, repeated exposure to PEGylated nanoparticles can lead to the formation of antibodies against PEG. These anti-PEG antibodies can rapidly bind to subsequent doses of PEGylated LNPs, leading to accelerated blood clearance (ABC phenomenon), where the nanoparticles are quickly removed from circulation, reducing their therapeutic efficacy [6]. In some cases, high levels of anti-PEG antibodies can also contribute to hypersensitivity reactions. The extent of this issue depends on the concentration and molecular weight of PEG used, the frequency of dosing, and individual patient factors. Efforts to mitigate anti-PEG antibody formation include minimizing PEG content, using biodegradable PEG-lipids that detach over time, or exploring alternative stealth coatings.

  • Mitigation Strategies: Researchers employ several strategies to minimize LNP immunogenicity. Optimizing the pKa of ionizable lipids to ensure they are near-neutral at physiological pH (zeta potential close to zero) is crucial for reducing non-specific interactions and complement activation [6]. Using biodegradable lipid components allows for rapid clearance of the LNP building blocks, preventing long-term accumulation and chronic immune stimulation. Modifying the nucleic acid cargo itself (e.g., nucleoside modifications in mRNA) can significantly reduce its intrinsic immunogenicity. Furthermore, controlling LNP size and polydispersity can also influence immune cell recognition.

5.2. Toxicity

The biocompatibility and potential toxicity of LNPs are critical considerations for their clinical translation. Toxicity can arise from the LNP components themselves, off-target delivery of the nucleic acid cargo, or the immune response they elicit.

  • Cytotoxicity of Ionizable Lipids: While ionizable lipids are designed to be relatively non-toxic at physiological pH, their ability to disrupt membranes for endosomal escape can also lead to cytotoxicity at higher concentrations or if they accumulate in cells. This can manifest as membrane lysis, mitochondrial dysfunction, or induction of apoptosis. The design of novel ionizable lipids focuses on balancing their efficacy in endosomal escape with their biodegradability, ensuring they are rapidly metabolized and cleared from the body once their function is served, thereby minimizing long-term accumulation and toxicity [5]. Studies have generally shown that current LNP formulations used in clinical applications are well-tolerated with minimal acute adverse effects in preclinical models, though comprehensive long-term toxicity assessments are still ongoing [5].

  • Biodistribution and Accumulation: As noted, intravenously administered LNPs primarily accumulate in the liver and spleen. While often desired for liver-targeted therapies, accumulation in non-target organs can lead to off-target effects. For example, excessive LNP uptake by Kupffer cells in the liver could lead to hepatic inflammation or dysfunction, although current formulations have demonstrated a good safety margin in this regard. The rapid degradation of LNP components into natural lipid metabolites helps to minimize long-term accumulation in these organs. However, for chronic disease management, long-term biodistribution and clearance kinetics need to be thoroughly evaluated.

  • Systemic Side Effects: Common transient side effects observed with mRNA LNP vaccines, such as fever, fatigue, headache, and muscle pain, are often attributed to the transient inflammatory response (innate immune activation) induced by the LNPs and the mRNA itself. These are generally mild to moderate and resolve within a few days. More severe systemic reactions are rare.

  • Manufacturing Impurities: The purity of LNP components and the final product is vital. Residual organic solvents, unencapsulated nucleic acids, or impurities from lipid synthesis can contribute to toxicity or immunogenicity. Rigorous quality control measures during manufacturing are essential to ensure product safety and consistency.

Overall, while potential safety concerns exist, the extensive preclinical and clinical data, particularly from mRNA COVID-19 vaccines, affirm that LNPs, when properly designed and formulated, possess a favorable safety profile that supports their widespread therapeutic utility. Continuous research is focused on refining LNP designs to further enhance their safety and reduce any potential for adverse reactions, especially for applications requiring chronic administration.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6. Applications in Cardiac Regeneration and Beyond

Lipid nanoparticles have transcended their initial success in vaccine delivery to become versatile platforms with profound implications across a multitude of therapeutic areas. Their ability to deliver diverse nucleic acid cargoes with high efficiency and relative safety positions them as a cornerstone for advanced gene therapies.

6.1. Cardiac Regeneration

Cardiovascular diseases, including myocardial infarction (heart attack) and heart failure, remain leading causes of morbidity and mortality worldwide. The adult mammalian heart has a limited intrinsic capacity for self-repair, leading to irreversible loss of cardiomyocytes and subsequent scar tissue formation after injury. LNP-mediated mRNA delivery offers a promising avenue for cardiac regeneration by transiently expressing therapeutic proteins that can stimulate angiogenesis (new blood vessel formation), suppress fibrosis, promote cardiomyocyte proliferation, or protect existing heart muscle [8, 10].

  • Mechanism and Therapeutic Targets: LNPs can deliver mRNA encoding pro-angiogenic factors such as vascular endothelial growth factor A (VEGF-A), which promotes the formation of new blood vessels, improving blood supply to ischemic cardiac tissue. Studies have successfully demonstrated the ability of LNPs to deliver VEGFA mRNA to cardiac tissue, leading to enhanced angiogenesis and improved cardiac function in preclinical models of myocardial infarction [8]. Beyond VEGF-A, researchers are exploring mRNA delivery of other growth factors (e.g., fibroblast growth factor, hepatocyte growth factor), transcription factors (e.g., GATA4, MEF2C, TBX5 to induce cardiomyocyte proliferation or reprogram fibroblasts into cardiomyocytes), or microRNAs that regulate cardiac repair processes. The ability of SORT nanoparticles to selectively target cardiac tissue is a significant breakthrough in this field, allowing for direct mRNA delivery to the heart while minimizing off-target effects in other organs, enhancing the therapeutic window for cardiovascular applications [3].

  • Challenges and Future Outlook: Key challenges include achieving sufficient and sustained expression of therapeutic proteins in cardiomyocytes, overcoming the fibrous scar tissue that can impede delivery, and ensuring uniform distribution throughout the damaged myocardium. Future research will focus on developing LNPs with even greater cardiac specificity, optimizing dosing regimens for sustained therapeutic effect, and combining mRNA therapies with other regenerative strategies (e.g., cell therapy).

6.2. Inhalation Therapies for Lung Diseases

Pulmonary diseases, ranging from cystic fibrosis to asthma, present a unique set of challenges for drug delivery due to the protective barriers of the respiratory system (mucus, alveolar macrophages, epithelial tight junctions) and the need for localized treatment to minimize systemic side effects. LNPs engineered for inhalation therapies offer a promising solution by directly delivering nucleic acids to lung tissues [9, 10].

  • LNP Design for Inhalation: For inhalation, LNPs must be stable enough to withstand nebulization (conversion into an aerosolized mist) and subsequently penetrate the thick mucus layer. Researchers have addressed these challenges by optimizing LNP size (typically 100-200 nm for optimal lung deposition), charge, and lipid composition. The incorporation of specific PEG-lipids and cholesterol analogs, as mentioned earlier, has been shown to enhance the stability of LNPs against shear forces during nebulization and improve their ability to traverse the mucus barrier. Once in the lung, these optimized LNPs facilitate efficient endosomal escape and gene transfection in lung epithelial cells and other relevant pulmonary cell types [9].

  • Applications: Potential applications include delivering mRNA encoding functional CFTR protein for cystic fibrosis patients, expressing therapeutic proteins to treat pulmonary hypertension or idiopathic pulmonary fibrosis, or local delivery of immunomodulatory mRNA for asthma or chronic obstructive pulmonary disease (COPD). Furthermore, inhalation of mRNA LNP vaccines could provide localized mucosal immunity for respiratory infections like influenza or RSV, potentially offering superior protection at the primary site of infection [10].

6.3. mRNA Vaccines

The COVID-19 pandemic unequivocally demonstrated the transformative power of mRNA LNP vaccines. Pfizer/BioNTech’s BNT162b2 and Moderna’s mRNA-1273 vaccines showcased unprecedented speed of development and high efficacy, revolutionizing vaccinology [7].

  • Mechanism: In these vaccines, LNPs encapsulate mRNA encoding the viral spike protein. Following intramuscular injection, LNPs are primarily taken up by antigen-presenting cells (APCs), such as dendritic cells and macrophages, which are abundant in muscle tissue and draining lymph nodes. Once internalized, the mRNA is translated into the spike protein. The APCs then process and present parts of this protein on their surface to T cells and B cells, initiating robust humoral (antibody) and cellular (T cell) immune responses against the virus [7]. The LNP itself, and the modified mRNA cargo, also act as intrinsic adjuvants, stimulating innate immune pathways that enhance the overall adaptive immune response.

  • Beyond COVID-19: The success of COVID-19 vaccines has paved the way for a new era of prophylactic and therapeutic vaccines. LNPs are now being explored for vaccines against other infectious diseases (e.g., influenza, RSV, HIV, Zika), and critically, for personalized cancer vaccines where mRNA encoding patient-specific neoantigens can stimulate a tailored anti-tumor immune response [10].

6.4. Cancer Immunotherapy and Gene Editing

LNPs are increasingly being utilized in oncology, particularly for cancer immunotherapy and gene editing applications.

  • Cancer Immunotherapy: LNPs can deliver mRNA encoding various immunomodulatory agents. This includes mRNA for checkpoint inhibitors (e.g., anti-PD-L1 antibodies) that can be locally produced by tumor cells or immune cells, leading to more potent and localized anti-tumor effects with fewer systemic side effects. They can also deliver mRNA for pro-inflammatory cytokines (e.g., IL-12) to enhance the tumor microenvironment’s immunogenicity, or neoantigen mRNA for personalized cancer vaccines, training the patient’s immune system to recognize and attack tumor cells [10].

  • Gene Editing: LNPs are a leading non-viral platform for delivering CRISPR-Cas components (mRNA encoding Cas nucleases, guide RNAs). This approach offers significant advantages over viral vectors, as it avoids integration of the Cas gene into the genome, reducing off-target effects and potential immunogenicity. LNP-mediated delivery of CRISPR-Cas9 mRNA/gRNA can be used for precise gene knockout or correction in various cell types, holding promise for treating genetic disorders like Duchenne muscular dystrophy, sickle cell disease, or certain cancers by editing specific genes within tumor cells or immune cells [10].

6.5. Rare Genetic Diseases

For many rare genetic diseases caused by a single gene defect, LNPs offer a non-viral solution for gene replacement or correction.

  • mRNA Replacement Therapy: LNPs can deliver mRNA encoding a functional protein to replace a defective or missing one. This has potential for enzyme replacement therapies (e.g., for lysosomal storage disorders) or for delivering therapeutic proteins for diseases like alpha-1 antitrypsin deficiency or hemophilia. The transient expression reduces concerns about permanent off-target effects [10].

  • siRNA Gene Silencing: For diseases caused by gain-of-function mutations or overexpression of a deleterious gene, LNPs can deliver siRNA to specifically silence the problematic gene. ONPATTRO® (patisiran), the first LNP-siRNA drug approved, treats hereditary transthyretin amyloidosis by silencing the TTR gene in the liver.

  • CRISPR-Cas Correction: As mentioned above, LNPs delivering CRISPR components can precisely correct disease-causing mutations, offering a potentially curative approach for a wide range of inherited disorders.

6.6. Central Nervous System (CNS) Delivery

Delivering therapeutics to the brain and spinal cord remains one of the greatest challenges in medicine due to the highly restrictive blood-brain barrier (BBB). While systemic LNP delivery to the CNS is generally inefficient, emerging strategies show promise.

  • Targeted LNPs: Researchers are developing LNPs functionalized with ligands that can facilitate transport across the BBB, for example, by targeting specific receptors on brain endothelial cells. Intranasal delivery of LNPs is also being explored as a non-invasive route that bypasses the BBB and allows direct access to the brain via olfactory and trigeminal nerves.
  • Direct Administration: For certain conditions, direct intracerebroventricular or intraparenchymal injection of LNPs can bypass the BBB, allowing localized delivery to specific brain regions, though this is invasive. Potential applications include treating neurodegenerative diseases (e.g., Alzheimer’s, Parkinson’s) or genetic neurological disorders by delivering neurotrophic factors or gene-editing components.

6.7. Pancreatic Delivery

Emerging research indicates that LNPs can be specifically engineered to target the pancreas, opening new avenues for treating diseases like diabetes and pancreatic cancer. The National Institute of Biomedical Imaging and Bioengineering (NIBIB) highlighted research on ‘packaging mRNA for the pancreas,’ demonstrating the potential of specific LNP formulations to achieve pancreatic tropism [4]. This precision delivery could facilitate the delivery of mRNA for insulin production in type 1 diabetes or therapeutic genes/siRNAs for pancreatic cancer with reduced systemic side effects.

The diverse and expanding array of LNP applications underscores their versatility and adaptability as a leading non-viral vector. Continuous innovation in LNP design and formulation is rapidly translating cutting-edge genetic research into tangible therapeutic solutions for a broad spectrum of human diseases.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

7. Challenges and Future Directions

Despite their remarkable successes, particularly in the realm of mRNA vaccines, Lipid Nanoparticles still face several challenges that necessitate ongoing research and innovation. Addressing these limitations will be crucial for realizing the full therapeutic potential of LNPs across a broader spectrum of diseases.

7.1. Key Challenges

  • Scalability of Manufacturing and Cost-Effectiveness: While microfluidics has enabled highly reproducible LNP production, scaling up to meet the demands for large patient populations, especially for chronic therapies, remains a challenge. The cost of raw materials, particularly high-purity lipids, and the intricate manufacturing processes contribute to the high cost of LNP-based therapeutics, limiting their global accessibility.

  • Achieving True Cell-Specific Targeting: Despite advancements in SORT and ligand-mediated targeting, achieving highly efficient and truly cell-type-specific delivery beyond liver and spleen remains a significant hurdle. Off-target delivery can lead to unwanted side effects and reduce therapeutic efficacy, particularly for diseases requiring precise targeting of rare cell populations or deeply embedded tissues.

  • Long-Term Safety and Immunogenicity for Chronic Administration: While acute safety of LNPs has been well-established (e.g., in vaccines), the long-term safety profile for chronic or repeat administration of LNPs for genetic diseases or cancer therapies is still an active area of investigation. Concerns about potential chronic inflammation, anti-PEG antibody development, and the accumulation of lipid components over extended periods require thorough evaluation.

  • Overcoming Intracellular Barriers More Efficiently: Endosomal escape, while improved with current ionizable lipids, is still a major bottleneck. A significant portion of internalized LNPs may still be trapped in endosomes and degraded in lysosomes. Enhancing the efficiency of endosomal escape without compromising safety is a continuous goal.

  • Cold Chain Requirements: Many LNP formulations require ultra-cold or cold chain storage, which poses significant logistical challenges, particularly for global distribution and in resource-limited settings. Developing thermostable LNP formulations that can be stored at refrigerated or even room temperature is a high priority.

7.2. Future Directions

The field of LNP research is incredibly dynamic, with numerous exciting avenues being explored to overcome current limitations and expand their therapeutic utility.

  • Next-Generation Ionizable Lipids: The continuous discovery and rational design of novel ionizable lipids with optimized pKa, enhanced biodegradability, improved endosomal escape efficiency, and reduced toxicity will remain a cornerstone of LNP advancement. High-throughput screening and computational modeling are accelerating this discovery process.

  • ‘Smart’ and Responsive LNPs: Developing LNPs that can respond to specific stimuli present at the disease site (e.g., changes in pH, redox potential, enzyme activity, light, ultrasound) to trigger controlled cargo release will enhance therapeutic precision and reduce off-target effects. These ‘smart’ LNPs could revolutionize drug delivery by ensuring the therapeutic payload is delivered only where and when it is needed.

  • Multi-Modal LNPs: Integrating other functionalities into LNPs, such as diagnostic imaging agents for real-time tracking of biodistribution, or combining nucleic acid delivery with small molecule drugs, could enable more comprehensive ‘theranostic’ approaches.

  • Advanced Manufacturing Techniques: Further development of continuous manufacturing processes, automation, and advanced microfluidic platforms will be crucial for scalable, cost-effective, and high-quality LNP production, enabling widespread access to these advanced therapies.

  • AI and Machine Learning in LNP Design: Leveraging artificial intelligence and machine learning algorithms can accelerate the discovery and optimization of LNP formulations by predicting structure-function relationships, identifying optimal lipid combinations, and designing novel lipid molecules with desired properties.

  • Ex Vivo and Local Delivery Routes: While systemic delivery is powerful, exploring and optimizing ex vivo (e.g., modifying cells outside the body with LNPs and then reinfusing them) and local delivery routes (e.g., inhalation, intra-tumoral, intra-articular) can circumvent some systemic challenges and offer localized therapeutic benefits.

  • Personalized LNP Formulations: In the long term, the principles governing LNP design could enable personalized formulations tailored to individual patient needs, disease states, and genetic profiles, maximizing efficacy and minimizing adverse effects.

  • Expanding the Cargo Repertoire: While mRNA, siRNA, and CRISPR components are leading, research continues into delivering other challenging biomolecules, such as proteins, antibodies, or even entire organelles, using LNP-like platforms.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

8. Conclusion

Lipid nanoparticles have unequivocally transformed the landscape of nucleic acid delivery, evolving from a niche research area to a cornerstone technology underpinning revolutionary therapeutic strategies. Their unparalleled ability to safely and efficiently encapsulate, protect, and deliver fragile genetic material into target cells has not only enabled the rapid development and deployment of life-saving mRNA vaccines but has also unlocked vast potential across an expansive range of medical applications.

From the precise delivery of mRNA for cardiac regeneration and the localized treatment of lung diseases to the intricate demands of cancer immunotherapy, rare genetic disorder correction, and beyond, LNPs have proven to be an adaptable and robust platform. The continuous and multidisciplinary research efforts focused on refining their structural components, elucidating their complex cellular mechanisms, and enhancing their stability and targeting capabilities are rapidly pushing the boundaries of what is therapeutically possible. While challenges related to targeted delivery, long-term safety, and manufacturing scalability persist, the rapid pace of innovation, driven by breakthroughs in lipid chemistry, nanotechnology, and bioengineering, provides a compelling vision for the future.

As the understanding of LNP-biological interactions deepens, and novel, ‘smarter’ LNP formulations emerge, these versatile nanocarriers are poised to play an even more central role in the era of precision medicine, offering hope for previously intractable diseases and fundamentally reshaping the approach to prevention, diagnosis, and treatment across numerous therapeutic domains. The LNP revolution is truly just beginning.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

References

  1. Dilliard, S. A., Cheng, Q., & Siegwart, D. J. (2021). On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proceedings of the National Academy of Sciences, 118(52), e2110190118. https://en.wikipedia.org/wiki/Selective_organ_targeting
  2. Hou, X., Zaks, T., Langer, R., & Dong, Y. (2021). Lipid nanoparticles for mRNA delivery. Nature Reviews Materials, 6(12), 1078–1094. https://www.nature.com/articles/s41578-021-00358-0?fromPaywallRec=false&utm_source=openai
  3. Cheng, Q., Wei, T., Farbiak, L., Johnson, L. T., & Dilliard, S. A. (2020). Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nature Nanotechnology, 15(3), 313–320. https://en.wikipedia.org/wiki/Selective_organ_targeting
  4. Russell, L., et al. (2023). Packaging mRNA for the pancreas. National Institute of Biomedical Imaging and Bioengineering. https://www.nibib.nih.gov/news-events/newsroom/packaging-mrna-pancreas
  5. Pros & Cons of lipid nanoparticles for mRNA delivery. (n.d.). SUSupport. https://www.susupport.com/knowledge/biopharmaceutical-products/drug-delivery/pros-cons-lipid-nanoparticles-mrna-delivery
  6. The immunogenic potential of an optimized mRNA lipid nanoparticle formulation carrying sequences from virus and protozoan antigens. (2023). Journal of Nanobiotechnology, 21(1), 1–13. https://jnanobiotechnology.biomedcentral.com/articles/10.1186/s12951-025-03201-8
  7. Lipid nanoparticles in the development of mRNA vaccines for COVID-19. (2021). National Center for Biotechnology Information. https://pmc.ncbi.nlm.nih.gov/articles/PMC9238147/
  8. Lipid Nanoparticles Deliver the Therapeutic VEGFA mRNA In Vitro and In Vivo and Transform Extracellular Vesicles for Their Functional Extensions. (2023). National Center for Biotechnology Information. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10131815/
  9. Engineering Lipid Nanoparticles for Enhanced Intracellular Delivery of mRNA through Inhalation. (2022). ACS Nano, 16(9), 14792–14806. https://pubs.acs.org/doi/10.1021/acsnano.2c05647
  10. Advances in lipid nanoparticle mRNA therapeutics beyond COVID-19 vaccines. (2024). Nanoscale, 16(1), 1–15. https://pubs.rsc.org/en/content/articlehtml/2024/nr/d4nr00019f
  11. Lipid nanoparticles for mRNA therapy: recent advances in targeted delivery. (2023). National Center for Biotechnology Information. https://pmc.ncbi.nlm.nih.gov/articles/PMC11749733/

1 Comment

  1. The discussion of SORT nanoparticles is fascinating! Engineering LNPs to target specific organs without relying solely on ligand-receptor interactions opens exciting possibilities for precision medicine, particularly for tissues beyond the liver. How might SORT nanoparticles impact the treatment of neurological disorders?

Leave a Reply

Your email address will not be published.


*