Next Article in Journal
Association Between Antidepressant Use and Risk of Venous Thromboembolism: A Systematic Review and Meta-Analysis
Previous Article in Journal
Concordance Index-Based Comparison of Inflammatory and Classical Prognostic Markers in Untreated Hepatocellular Carcinoma
Previous Article in Special Issue
A New Tool Supporting the Selection of the Best Hematopoietic Stem Cell Donor by Modelling Local Own Real-World Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transfection Technologies for Next-Generation Therapies

by
Dinesh Simkhada
,
Su Hui Catherine Teo
,
Nandu Deorkar
and
Mohan C. Vemuri
*
Biopharma and Bioproduction, Cell and Gene Therapy, Avantor, 77 Corporate Drive, Bridgewater, NJ 08807, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(15), 5515; https://doi.org/10.3390/jcm14155515
Submission received: 1 July 2025 / Revised: 23 July 2025 / Accepted: 31 July 2025 / Published: 5 August 2025

Abstract

Background: Transfection is vital for gene therapy, mRNA treatments, CAR-T cell therapy, and regenerative medicine. While viral vectors are effective, non-viral systems like lipid nanoparticles (LNPs) offer safer, more flexible alternatives. This work explores emerging non-viral transfection technologies to improve delivery efficiency and therapeutic outcomes. Methods: This review synthesizes the current literature and recent advancements in non-viral transfection technologies. It focuses on the mechanisms, advantages, and limitations of various delivery systems, including lipid nanoparticles, biodegradable polymers, electroporation, peptide-based carriers, and microfluidic platforms. Comparative analysis was conducted to evaluate their performance in terms of transfection efficiency, cellular uptake, biocompatibility, and potential for clinical translation. Several academic search engines and online resources were utilized for data collection, including Science Direct, PubMed, Google Scholar Scopus, the National Cancer Institute’s online portal, and other reputable online databases. Results: Non-viral systems demonstrated superior performance in delivering mRNA, siRNA, and antisense oligonucleotides, particularly in clinical applications. Biodegradable polymers and peptide-based systems showed promise in enhancing biocompatibility and targeted delivery. Electroporation and microfluidic systems offered precise control over transfection parameters, improving reproducibility and scalability. Collectively, these innovations address key challenges in gene delivery, such as stability, immune response, and cell-type specificity. Conclusions: The continuous evolution of transfection technologies is pivotal for advancing gene and cell-based therapies. Non-viral delivery systems, particularly LNPs and emerging platforms like microfluidics and biodegradable polymers, offer safer and more adaptable alternatives to viral vectors. These innovations are critical for optimizing therapeutic efficacy and enabling personalized medicine, immunotherapy, and regenerative treatments. Future research should focus on integrating these technologies to develop next-generation transfection platforms with enhanced precision and clinical applicability.

1. Introduction

Transfection is a fundamental technique in biosciences and biotechnology, playing a critical role in developing cell and gene therapies. It enables the introduction of nucleic acids into cells, a crucial step in gene editing, gene therapy, and protein production for drug discovery [1]. Despite its importance, mechanisms governing transfection efficiency remain only partially understood [1,2].
The complex interactions of transfection reagents with cells and how they facilitate genetic material uptake, thereby influencing cellular processes, are actively being studied. While optimized for certain cell types and applications, achieving high transfection efficiency across diverse cell lines, especially primary cells, stem cells, and complex tissues, remains a significant challenge [3]. Furthermore, the toxicity of many transfection reagents is poorly understood, and their potential side effects on cell viability and gene expression limit broader therapeutic utility [2]. Variability in transfection efficiency, the need for specialized reagents, and the complexities of protocol optimization also continue to pose challenges [3,4].
A more informative approach to understanding and placing these limitations into perspective must consider the conventional means of transfection and the practical compromises. The traditional approaches, such as electroporation, gene gun (biolistic), and viral vector delivery, have been the workhorse of gene delivery techniques for decades. Electroporation is the application of high-voltage electrical pulses to form temporary pores in the cell membrane, to which the nucleic acids are delivered. It is effective across various cell types and allows for both transient and stable transfection, but it often results in high cell death and requires cell-type-specific optimization [5,6]. The gene gun approach utilizes high-pressure gas to introduce DNA-covered microparticles into cells and is optimal for plant cells or aggregated tissues but has minimal transfection efficiency and cellular damage potential [7]. Virus vectors, including lentiviruses, adenoviruses, and retroviruses, possess high delivery efficiency and long-lasting gene expression because they become incorporated into the genome of the host. However, these vectors are plagued by significant biosafety and immunogenicity concerns, as well as production and regulatory challenges [8]. While these legacy methods have been instrumental to gene delivery progress, their limitations in terms of safety, consistency, and scalability underscore the imperative for new technologies.
To address these challenges, non-viral gene delivery systems have garnered increasing attention due to their improved safety profiles and engineering tractability [9]. Among these, metal–organic frameworks (MOFs) and inorganic nanoparticles have emerged as outstanding candidates for next-generation platforms. MOFs, composed of metal ions coordinated with organic ligands, exhibit outstanding structural features, such as high porosity, tunable surface chemistry, and biocompatibility [10]. Such features make MOFs highly capable of encapsulating and protecting nucleic acids such as plasmid DNA, siRNA, mRNA, and CRISPR/Cas reagents, while facilitating controlled release and cellular uptake promotion via functionalization. At the same time, inorganic nanoparticles, such as gold nanoparticles, silica nanoparticles, and calcium phosphate-based systems, are highly efficient for nucleic acid delivery [11,12]. They are capable of being engineered to perform surface functionalization for increased endosomal release, improved target specificity, and reduced immunogenicity [9]. MOFs and inorganic nanoparticles are less cytotoxic than cationic polymers and offer convenient platforms for gene targeting in cancer treatment and regenerative medicine [13]. Nevertheless, significant hurdles such as scale-up manufacture, biodegradability, potential toxicity of metal ions, and controlled release kinetics continue to impede clinical translation. Despite these issues, their tunability for physicochemical properties puts MOFs and inorganic nanoparticles in the vanguard of prospective non-viral gene delivery candidates [14].
The limitations of conventional transfection methods—low efficacy in primary and stem cells, cytotoxicity, and variability—highlight the need for innovative and versatile gene delivery systems. The objective of the current review is to comprehensively evaluate existing non-viral transfection methods and systematically compare their efficacy in terms of delivering genetic material and expression and biosafety, including potential for toxicity, immunogenicity, and off-target effects that can be reliably used in research and clinical settings [15,16,17,18]. Furthermore, the review explores and highlight platforms that are pushing the boundaries of gene delivery and assessing enhance therapeutic applications.

2. Non-Viral Gene Delivery Systems

Non-viral gene delivery systems offer key advantages over viral vectors, including safety, higher payload capacity, and reduced immunogenicity. Unlike viral vectors, these systems do not integrate genetic material into the host genome [19], minimizing the risk of insertional mutagenesis, enabling transient therapeutic effects—ideal for applications like vaccines. Their higher payload capacity allows delivery of larger or multiple genes, including CRISPR components [20]. Non-viral approaches elicit minimal immune responses, enabling repeated administration and increased safety in immunocompromised patients.
Furthermore, they can deliver diverse genetic material, including DNA, RNA, and oligonucleotides, making them adaptable for gene editing [21,22,23,24,25,26,27], and RNA interference therapies [28]. Common platforms include lipid-based carriers, such as lipoplexes and lipid nanoparticles (LNPs), and cationic polymers, such as polyethyleneimine (PEI) and chitosan, which form polyplexes with nucleic acids. These carriers protect genetic cargo, aid in cellular uptake and endosomal escape, and can be engineered for tissue-specific delivery. In short, non-viral systems are economical, scalable, and compatible with targeted delivery to specific tissues (Figure 1).
Extracellular vesicles (EVs), especially exosomes, are emerging as powerful non-viral vectors for gene delivery due to their natural origin, biocompatibility, and ability to facilitate intercellular communication. These nano-sized vesicles (30–150 nm) protect genetic cargo, such as DNA, siRNA, and mRNA, from enzymatic degradation, thereby improving delivery stability and efficiency. Their low immunogenicity and toxicity make them suitable for repeated use, even in immunocompromised patients [29,30]. Exosomes can be engineered either post-isolation (e.g., electroporation) or via donor cell modification to load nucleic acids endogenously. Surface modifications further enable targeted delivery [31]. Despite challenges in large-scale production and standardization, their biomimetic properties make exosomes promising candidates for RNA therapeutics and precision medicine [32,33].
These combined advantages position non-viral delivery as a key innovation in driving the development of therapeutic gene editing technologies [34,35]. This potential is reflected in the active engagement of numerous pharmaceutical companies in developing therapeutic drugs targeting diverse diseases using non-viral gene delivery systems (Table 1). By leveraging these systems, these companies aim to enhance the efficiency and targeting of therapeutic gene delivery, ultimately improving treatment outcomes and broadening the clinical applications of gene therapy [36].

3. Advancing to Clinical Path Through Transfection

Transfection technologies are significantly improving clinical applications through innovative approaches leading to the development of new therapies and optimizing existing treatments (Figure 2).

3.1. mRNA-Based Therapies

The field of mRNA therapeutics has rapidly advanced due to its potential in addressing various medical conditions. Unlike traditional protein therapies, mRNA enables in vivo protein production, bypassing complex purification processes [56,57]. The rapid development and deployment of COVID-19 vaccines exemplifies the power of mRNA-based vaccines [58,59,60,61]. Beyond vaccines, mRNA is being explored for protein-replacement therapy [57], cancer immunotherapy, cellular reprogramming, and gene editing [62,63,64].
Effective mRNA-based therapies depend on efficient transfection technologies. Delivering mRNA to target cells is challenging due to its susceptibility to degradation and the need for suitable carriers to facilitate intracellular uptake [60,65]. Several transfection methods have been explored, including lipid-based systems, polymer-based systems, and viral transduction, each with advantages and limitations. Polymers, especially cationic polymers, offer reduced immunotoxicity, low cost, and ease of production, but face challenges with low transfection efficiency and potential toxicity [66]. While viral transduction is highly efficient for delivering genetic material into hard-to-transfect cells, including primary cells, it carries the risk of increased cytotoxicity and viral infection [67,68].
Lipid nanoparticles (LNPs) have emerged as the leading non-viral delivery platform, showing great promise for in vivo delivery of RNA therapeutics, such as small interfering RNAs (siRNA) [69], antisense oligonucleotides (ASOs), synthetic guide RNAs [70,71], and mRNAs [72,73]. LNPs offer flexibility in payload adaptation and lipid composition, making them a transformative tool in drug delivery [74,75]. Their stability, efficiency, and reduced immunogenicity position them pivotal in advancing vaccines, gene therapies, and antibody-based treatments.

3.2. CAR-T Cell Therapy

CAR-T cell therapy represents a significant advance in cancer treatment. It involves genetically engineered T cells which possess a remarkable ability to target and destroy cancerous cells with high specificity [76,77]. CAR-T cells have demonstrated impressive results against hematological malignancies such as, acute lymphoblastic leukemia (ALL) and large B-cell lymphoma, offering potentially curative outcomes for patients unresponsive to conventional treatments [78]. This therapy relies on advanced transfection technologies to precisely modify T cells enabling them to express chimeric antigen receptors (CARs) [79,80]. While viral vectors, like lentiviruses and retroviruses, are commonly used for transfection due to their efficient and stable and transfer of CAR genes into T cells [81], they carry a potential of insertional mutagenesis due to integration of genetic material into the host genome.
Beyond viral vectors, non-viral approaches, such as electroporation, have emerged as attractive alternatives that allow rapid and inexpensive introduction of CAR-encoding mRNA or DNA into T cells, thus largely avoiding integration-related risks [82,83]. Furthermore, polymer-based and lipid nanoparticle (LNP) systems are also being explored as safer, less immunogenic methods that also facilitate scalable CAR-T cell production [84]. These advances in transfection technology are not only improving the efficiency of CAR-T therapy but also expanding its application to solid tumors, which pose unique challenges such as antigen heterogeneity and limited T cell infiltration [85,86,87,88]. These engineered cells may possess dual-targeting capabilities and resistance to tumor-induced immunosuppression. Innovations like electroporation and RNA-based CAR transfection are streamlining manufacturing processes, lowering costs, and increasing flexibility. Collectively, these advances enhance the safety and efficacy of CAR-T therapy and broaden its clinical applicability to a wider range of cancers [87,88].

3.3. Revolutionizing Gene Therapy

Gene therapy has emerged as a revolutionary modality in modern medicine, offering the potential to treat or even cure genetic disorders by directly targeting the underlying genetic defects. A critical component of successful gene therapy is the development and optimization of transfection technologies which enable the safe and efficient delivery of therapeutic genes into target cells [89,90,91]. Beyond monogenic disorders, gene therapy is also contributing to advances in cancer treatment through the genetic engineering of immune cells to enhance tumor suppression and immune response [92,93]. These advances are often enabled by transfection methods such as electroporation and nanoparticle delivery. Furthermore, innovations in in vivo delivery approaches—including adaptations of lipid nanoparticle (LNP) technology from mRNA vaccines and tissue-specific targeting via receptor-mediated pathways—are addressing significant challenges, improving precision, and expanding the therapeutic potential of gene therapy [94,95]. Continued evolution of transfection technologies is focused on overcoming challenges related to efficiency, immunogenicity, and scalability [96]. These ongoing developments are poised to transform the treatment landscape for a range of diseases, including genetic disorders, cancer, and other complex conditions [97,98,99].

3.4. Small Interfering RNA and Antisense Oligonucleotide Therapies

Small interfering RNA (siRNA) and antisense oligonucleotides (ASOs) represent powerful gene silencing and therapeutic gene-modifying tools with significant potential for treating genetic disorders, viral infections, and cancers. Effective delivery to target tissues is crucial for their therapeutic success, and this relies heavily on optimized transfection reagents [100,101,102,103]. siRNA therapies, exemplified by the FDA-approved Onpattro for hereditary transthyretin-mediated amyloidosis, use RNA interference to degrade mRNA and require LNPs for precise targeting [100,104]. In contrast, ASOs, such as Spinraza for spinal muscular atrophy (SMA) and Eteplirsen for Duchenne muscular dystrophy (DMD), bind to mRNA to inhibit translation or correct splicing defects [105]. Advances in cationic lipids and polymers have led to improved ASO delivery, enhancing stability, targeting specificity and minimizing immune responses [106,107,108,109,110,111,112,113,114]. These innovations in delivery technologies are expanding the scope of RNA-based therapeutics, offering new hope for patients with previously untreatable genetic disorders [114,115,116,117,118,119,120,121].

3.5. Regenerative Medicine

The efficacy of regenerative medicine strategies, including stem cell therapy, tissue engineering, and gene therapy, is significantly enhanced by advances in transfection technologies [122]. A pivotal advance in therapies is the utilization of transfection reagents, which markedly enhance gene delivery efficiency. These techniques facilitate precise genetic modifications, driving tissue repair and cellular regeneration. In stem cell therapy, electroporation, viral vectors, and nanoparticle-based delivery systems are pivotal for introducing gene-editing tools, such as CRISPR/Cas9, enabling targeted genomic modifications and optimizing therapeutic efficacy [123,124,125,126].
Tissue engineering utilizes transfection-mediated delivery of growth factor genes to promote the regeneration of complex tissues, including bone, cartilage, and nerves. Notably, mRNA-lipid nanoparticle systems offer a non-viral, low-risk approach for cellular reprogramming, enabling the generation of specialized cell types [127]. Gene therapy employs these transfection technologies to deliver therapeutic genes, addressing conditions like ischemic injuries, muscular dystrophy, and retinal degeneration). These methodologies feature the critical role of efficient gene delivery in realizing the full potential of regenerative medicine [128]. Examples of specific therapies developed by pharmaceutical companies that utilize non-viral gene delivery systems are summarized in Table 2.

4. Key Challenges in Transfection

The field of gene delivery has witnessed substantial progress with the development of sophisticated transfection technologies. These include liposomal, polymer-based systems, offering biocompatibility and controlled release; viral vectors, providing high transduction efficiency; nanoparticle-mediated delivery, enabling targeted gene transfer; electroporation, facilitating direct cellular uptake; and microfluidic approaches, allowing for precise control and high-throughput applications [136,137]. Each of these methods present unique advantages and pose specific challenges [138,139] associated with efficient and safe gene delivery (Figure 3).
Chemical transfection, a widely employed method, utilizes cationic reagents, including lipids and polymers to complex with nucleic acids. This facilitates cellular entry primarily through endocytosis [140,141]. Lipofection, a prominent technique within this category, leverages cationic lipids to form lipoplexes, enabling efficient transmembrane transport. While alternative reagents such as calcium phosphate and DEAE-dextran have been utilized, they generally exhibit lower transfection efficiency and increased cytotoxicity [2,142].
Cationic polymers—such as polyethyleneimine (PEI), are employed in polymer-based transfection to form nanoparticles that deliver nucleic acids into cells [143,144,145]. However, the use of these polymers has been historically limited by concerns of their inherent cytotoxicity and biocompatibility. To address these limitations, the development of biodegradable and biocompatible polymers, such as poly (lactic-co-glycolic acid) (PLGA) and poly(beta-amino esters) (PBAEs), has significantly improved the safety profile and transfection efficiency, especially in in vivo settings [146].
Exosome-based gene delivery shows great potential, but several challenges hinder its clinical translation. Standardizing and scaling production remain difficult due to inconsistent yields and purity, which complicates reproducibility and quality control [32]. Cargo loading methods often compromise efficiency or vesicle integrity, and while exosomes offer natural targeting, achieving precise tissue-specific delivery without complex surface engineering is still technically demanding [30,147]. Additionally, limited understanding of cellular uptake and intracellular trafficking reduces the predictability of gene expression outcomes [29]. The absence of standardized protocols and regulatory frameworks further delays clinical adoption [148]. Addressing these issues is key to realizing exosomes as scalable, safe, and effective non-viral delivery systems.
Physical transfection methods, including electroporation and microinjection, offer direct nucleic acid delivery by circumventing cellular barriers [149]. Electroporation utilizes electrical impulses to induce transient membrane permeabilization, facilitating nucleic acid uptake, while microinjection enables precise, single-cell delivery [150]. Despite their efficacy in specialized applications, these techniques are limited by scalability, potential cellular damage, and reduced cell viability [151]. A significant challenge across all transfection modalities is endosomal trapping, wherein nucleic acids are sequestered within endosomes, hindering their functional activity [152,153,154]. To address this, substantial efforts have focused on enhancing endosomal escape through the development of fusogenic lipids, pH-responsive materials, and other escape enhancers, leading to improved delivery efficiency [136,155,156,157].
Despite promising preclinical results, metal–organic frameworks (MOFs) and inorganic nanoparticles face significant hurdles in reaching clinical use. While these materials offer advantageous properties like tunable porosity, high surface area, and chemical functionalization, most MOFs are still in early research and development [158,159]. Several critical issues impede their transition into clinical applications, such as drug delivery and bioimaging. These include limited biocompatibility data, potential long-term toxicity, challenges in large-scale synthesis, and regulatory ambiguities [159]. Overcoming these challenges is crucial to fully harness the therapeutic potential of MOFs and other inorganic nanomaterials [160].
Microfluidic technologies leverage precise control of fluid flow at the microscale to introduce genetic material into cells with high accuracy [161]. These systems offer significant benefits, including minimized reagent consumption, precise spatiotemporal control of transfection parameters, and the capability for single-cell manipulation This makes them advantageous for high-throughput applications and for specialized techniques such as intracellular delivery and mechanoporation [162]. However, the mechanical and physical forces generated within microfluidic devices, particularly shear stress, can induce cellular stress, potentially leading to compromised cell viability and reduced transfection efficiency [163].
Figure 3. Non-Viral DNA Transfection: This illustration depicts the gene transfection processes, effectively navigating various obstructions to deliver nucleic acids. [Created in BioRender. Teo, C. (2025) https://BioRender.com/ludvsnm, premium version] [164].
Figure 3. Non-Viral DNA Transfection: This illustration depicts the gene transfection processes, effectively navigating various obstructions to deliver nucleic acids. [Created in BioRender. Teo, C. (2025) https://BioRender.com/ludvsnm, premium version] [164].
Jcm 14 05515 g003
Nucleic acid delivery faces significant obstacles, including extracellular degradation, serum interactions, and intracellular membrane penetration [165]. Physical methods, while aiding cellular entry, can compromise cell viability, and viral vectors pose inherent safety risks. Non-viral synthetic vectors, notably lipid-based systems, offer a safer approach but necessitate optimization to enhance gene delivery efficiency [166]. Their clinical translation is hindered by tropism-related challenges. These systems lack the intrinsic targeting specificity of viral vectors, which naturally engage receptors on target cells [167]. To compensate, non-viral carriers often require surface functionalization with ligands, antibodies, or peptides to achieve tissue-specific delivery [167,168]. However, receptor heterogeneity among cell types complicates consistent targeting, leading to off-target distribution and variable therapeutic efficacy [167]. Additionally, functionalization efforts can impair vector stability, reduce uptake efficiency, or trigger rapid clearance by the mononuclear phagocyte system (MPS) or reticuloendothelial system (RES) [167,169]. The prevalent liver tropism of many nanoparticle systems further limits delivery to other organs unless physical or molecular targeting strategies are employed [170,171]. Thus, achieving precise tropism without sacrificing biocompatibility, efficiency, or scalability remains a critical and unsolved challenge for the effective use of non-viral gene delivery systems in clinical settings [172].
As another example, hematopoietic stem cell (HSC) transfection systems can be considered for tissue-specific tropism. Transfecting HSCs remains a formidable challenge due to their quiescent nature, sensitivity to manipulation, and the need to preserve their self-renewal and multilineage differentiation potential [173]. However, the emergence of non-viral transfection technologies has significantly enhanced the feasibility of genetic modification in these cells, offering safer, more scalable, and less immunogenic alternatives to traditional viral vectors. Among these, ribonucleoprotein (RNP) complex delivery has emerged as a particularly promising strategy. This method involves the direct introduction of pre-assembled CRISPR-Cas9 protein complexes with guide RNA into HSCs, enabling rapid and highly specific genome editing while circumventing the risks of insertional mutagenesis associated with DNA-based approaches [172]. Recent technological advancements have further refined RNP delivery through innovative platforms such as microfluidics, which allow precise modulation of transfection parameters; filtroporation, which employs pressure gradients to facilitate cytosolic entry; and cell-penetrating peptides (CPPs), which enhance membrane permeability with minimal cytotoxicity. Collectively, these developments are transforming the landscape of HSC engineering, paving the way for safer and more effective gene therapies for hematologic and genetic disorders [174].

5. Transfection Technologies: Future Perspectives

The next generation of transfection technologies aims to improve efficiency, reduce immunogenicity, and enable precise gene delivery [59,175,176]. Specifically, mRNA-based therapies face obstacles such as instability, immune activation, and limited tissue targeting. To overcome these challenges, strategies involving ionizable lipids, fusogenic materials, dendrimers, and biodegradable polymers have been implemented to increase safety and broaden therapeutic applicability for clinical translation [177,178,179,180,181,182,183,184,185]. LNPs can be engineered for targeted gene delivery by manipulating their chemical composition and charge, as evidenced by successful CRISPR/Cas9-mediated PTEN editing. The inclusion of ionizable cationic lipids in LNP formulations is essential for encapsulating CRISPR/Cas9 components, enhancing the circulation half-life, and improving endocytosis efficiency by target cells [186]. Although LNPs are beneficial in terms of scalability and the ability to deliver large molecules, extracellular vesicles (EVs) are being explored as biocompatible and nonantigenic delivery vehicles for mRNAs [187]. EVs can currently be loaded with mRNA via specialized plasmids or direct methods [188]. Moreover, EVs loaded with targeting peptides, such as those that use the C1C2 domain of lactadherin, have been found to be effective in preclinical cancer therapy with prolonged circulation and no toxicity [189]. LNPs and EVs are suitable candidates for gene therapy, with LNPs being more potent in large-scale applications and EVs having improved biocompatibility and lower toxicity.
CPPs also show promise as effective vectors for mRNA delivery with high cellular uptake and endosomal escape efficacy. PepFect14 (PF14), for example, is used to create stable nanocomplexes for promoting mRNA delivery [190], whereas RALA is a pH-sensitive peptide used in cancer treatment [191]. The HIV-1 Tat peptide has been extensively used in mRNA vaccine design and gene expression research [192]. Moreover, MPG has shown potential in regenerative medicine [193], whereas CADY has high transfection efficiency for therapeutic purposes [194]. Overall, the development of next-generation delivery systems aims to address several critical challenges, including low transfection efficiency in difficult-to-transfect cell types, high cytotoxicity, poor compatibility with high-throughput screening (HTS) platforms, inconsistent reproducibility, and limited formulation stability during storage. Overcoming these barriers is crucial for advancing molecular and cellular biology and for enabling translational applications such as gene therapy and regenerative medicine.
In CAR-T cell therapy, CAR-T cells modified with the Sleeping Beauty (SB) transposon system have demonstrated potent antileukemic activity with a favorable safety profile. This non-viral approach reduces the risk of insertional mutagenesis, lowers manufacturing cost and simplifies production. SB-engineered CAR-T cells effectively target leukemia and are being investigated for allogeneic CAR-T cell therapies [40]. Since 2018, LNP-based manufacturing strategies have been rigorously investigated to optimize CAR-T cell production. Early studies demonstrated a reduction to a 3-day manufacturing timeline, with recent advances utilizing activating lipid nanoparticles achieving a remarkable 1-day production cycle [38]. This process acceleration, facilitated by innovative LNP formulations, enables scalable production of autologous therapies by streamlining workflows rather than simply increasing volume. The integration of closed systems further enhances manufacturing efficiency by eliminating bead-mediated T cell activation, simplifying culture procedures, and reducing costs, thereby bolstering centralized production capacity [39]. These improvements are particularly significant for patients with rapidly progressing cancers, where swift treatment access is essential [37].
The future of transfection technologies is poised between biotechnology and artificial intelligence (AI) in the pipeline, with the promise of transforming gene therapy and regenerative medicine. AI-driven modeling is demystifying the complexity of intricate cellular interactions, enabling rational design of the next generation of gene delivery systems with more precision, less toxicity, and cell-type specificity [195,196]. By leveraging machine learning and computational simulations, researchers can now predict optimal transfection conditions, streamline vector engineering, and minimize the traditional reliance on trial-and-error experimentation. AI algorithms have already shown to be successful for the optimization of lipid nanoparticle (LNP) formulations for mRNA delivery, simulation of nucleic acid–polymer interactions, and prediction of physicochemical properties of novel nanocarriers, such as polyplexes and MOF-based platforms [197,198]. In silico technologies also enable the virtual screening of carrier libraries to improve cellular uptake, endosomal escape, and intracellular stability—key determinants of effective transfection [199]. This foresight role not only accelerates the experimental process but also bridges the translational gap between the research bench and clinic. Moreover, the integration of AI with single-cell transcriptomics and omics data allows for personalized gene delivery approaches via the identification of unique molecular signatures and tissue-specific receptors [200]. The confluence of computational modeling and lab validation holds the potential to accelerate regulatory approval pathways, reduce development expenditure, and enhance patient access to novel gene therapies. With the advancement of AI, its role in guiding non-viral vector design, dosage optimization, and therapeutic targeting will play a pivotal role in shaping the future of precision medicine.
Artificial intelligence (AI) is transforming the optimization of transfection efficiency in lipid nanoparticle (LNP)-mediated mRNA delivery by leveraging deep learning, multimodal data fusion, and explainable AI [87,201,202]. Models like TransMA integrate 3D molecular geometry with 1D atomic sequences to deliver high accuracy and interpretable attention maps, aiding in the identification of transfection cliffs [198]. LANTERN combines Morgan fingerprints with expert descriptors in a multilayer perceptron, outperforming complex models like AGILE with strong generalizability [198]. TransLNP, a transformer-based model using the BalMol data-balancing technique, excels in predicting across diverse LNPs and small datasets [203]. Meanwhile, AGILE uses a graph neural network trained on 60,000 virtual lipids for effective large-scale screening (AGILE platform) [204]. Collectively, these AI tools are reshaping mRNA delivery by enabling more efficient and scalable formulation strategies [205,206].

6. Conclusions

Transfection technologies continue to evolve, addressing efficiency, safety, and scalability concerns to unlock their full potential in gene therapy, drug development, and disease treatment. As transfection methodologies advance, they promise to revolutionize the landscape of biomedical sciences by enhancing gene therapy, drug discovery, and cellular engineering. Emerging techniques, such as lipid nanoparticle systems and non-viral gene delivery, provide innovative solutions to existing challenges, optimizing safety and efficacy across applications. The interplay between efficiency, toxicity, and scalability is driving continuous innovation in gene delivery, with advances in lipid nanoparticles, non-viral vectors, and electroporation providing promising alternatives. By addressing current limitations, researchers are not only improving transfection methodologies but also expanding the therapeutic possibilities of genetic medicine, bringing new hope for precision treatments. In closing, this review provides a strategic roadmap for researchers working on gene delivery technologies, especially those bridging research with therapeutic development and clinical application.

Author Contributions

Conceptualization: D.S. developed the concept and outlined advancements in existing and emerging DNA transfection technologies; methodology, S.H.C.T. focused on RNA-based methods and non-viral delivery systems, contributing to their design and refinement; software: N.D. explored the role of artificial intelligence in transfection technology, examining how AI can be leveraged to create predictive models for improved future applications; validation: M.C.V. conceived the overall study framework, overseeing its design, review process, and team coordination to ensure a comprehensive and cohesive final analysis. All authors have read and agreed to the published version of the manuscript.

Funding

This research was conducted without any external funding. All authors are full-time employees of Avantor Sciences, and the study was carried out as part of their professional roles within the organization.

Institutional Review Board Statement

This study is entirely based on a meta-analysis of previously published data and publicly available research. It does not involve direct experimentation on humans or animals, nor does it require any interaction with living subjects.

Informed Consent Statement

Since no subjects are involved consent for participation is not applicable.

Data Availability Statement

All data and materials utilized in this study are sourced exclusively from publicly available databases and published research. As outlined in the references, the information has been obtained from open-access repositories and previously published works, ensuring transparency and accessibility. No proprietary or restricted datasets have been used, and all referenced materials are freely accessible to the public, allowing for independent verification and further analysis by other researchers.

Acknowledgments

Mohan C. Vemuri (M.C.V.) expresses gratitude for the support in utilizing references from Johns Hopkins University.

Conflicts of Interest

All authors were/are employed by the company Avantor. The authors declare that there are no competing interests related to this study. There are no financial, professional, or personal affiliations that could influence the interpretation or presentation of the findings.

Abbreviations

AIArtificial intelligence
ALLAcute lymphoblastic leukemia
ASOsAntisense Oligonucleotides
CAR-TChimeric antigen receptor-T
COVID-19Coronavirus disease 2019
CPPCell penetrating peptides
CRISPRClustered regularly interspaced short palindromic repeats
DMDDuchenne muscular dystrophy
DNADeoxy ribonucleic acid
EVsExtracellular vesicles
HTSHigh-throughput screening
HSCHematopoietic stem cell
LNPsLipid Nanoparticles
MOFMetal–organic frameworks
mRNAMessenger RNA
PBAEsPoly(beta-amino esters)
PEIPolyethylenimine
PLGAPoly (lactic-co-glycolic acid)
RNARibonucleic acid
SBSleeping Beauty
SiRNASmall interfering RNA
SMASpinal muscular atrophy

References

  1. Wells-Holland, C.; Elfick, A. Transfection reflections: Fit-for-purpose delivery of nucleic acids. Nat. Rev. Mol. Cell Biol. 2023, 24, 771–772. [Google Scholar] [CrossRef]
  2. Fus-Kujawa, A.; Prus, P.; Bajdak-Rusinek, K.; Teper, P.; Gawron, K.; Kowalczuk, A.; Sieron, A.L. An Overview of Methods and Tools for Transfection of Eukaryotic Cells in vitro. Front. Bioeng. Biotechnol. 2021, 9, 701031. [Google Scholar] [CrossRef]
  3. Chong, Z.X.; Yeap, S.K.; Ho, W.Y. Transfection types, methods and strategies: A technical review. Peer J. 2021, 9, e11165. [Google Scholar] [CrossRef]
  4. Rose, J.K. Optimization of transfection. Curr. Protoc. Cell Biol. 2003, 20, 20.7. [Google Scholar] [CrossRef]
  5. Liu, F.; Su, R.; Jiang, X.; Wang, S.; Mu, W.; Chang, L. Advanced micro/nano-electroporation for gene therapy: Recent advances and future outlook. Nanoscale 2024, 16, 10500–10521. [Google Scholar] [CrossRef] [PubMed]
  6. Campelo, S.N.; Huang, P.-H.; Buie, C.R.; Davalos, R.V. Recent advancements in electroporation technologies: From bench to clinic. Annu. Rev. Biomed. Eng. 2023, 25, 77–100. [Google Scholar] [CrossRef] [PubMed]
  7. Sharma, D.; Arora, S.; Singh, J.; Layek, B. A review of the tortuous path of nonviral gene delivery and recent progress. Int. J. Biol. Macromol. 2021, 183, 2055–2073. [Google Scholar] [CrossRef]
  8. Dan, L.; Kang-Zheng, L. Optimizing viral transduction in immune cell therapy manufacturing: Key process design considerations. J. Transl. Med. 2025, 23, 501. [Google Scholar] [CrossRef]
  9. Beta LifeScience. Understanding Transfection and Its Role in Modern Cell Biology. Frontier News, 10 June 2025. [Google Scholar]
  10. Feng, S.; Li, Y.; Tan, Z.; Shen, S. Current landscape of metal-organic framework-mediated nucleic acid delivery and therapeutics. Int. J. Pharm. 2025, 672, 125295. [Google Scholar] [CrossRef] [PubMed]
  11. Lawson, H.D.; Nguyen, H.H.; Lee, K.J.; Wongsuwan, N.; Tupe, A.; Lu, M.; Arral, M.L.; Behre, A.; Ling, Z.; Whitehead, K.A.; et al. Synthetic Strategy for mRNA Encapsulation and Gene Delivery with Nanoscale Metal-Organic Frameworks. Adv. Funct. Mater. 2025, 35, 2404465. [Google Scholar] [CrossRef]
  12. Kim, M.; Hwang, Y.; Lim, S.; Jang, H.-K.; Kim, H.-O. Advances in Nanoparticles as Non-Viral Vectors for Efficient Delivery of CRISPR/Cas9. Pharmaceutics 2024, 16, 1197. [Google Scholar] [CrossRef]
  13. Wu, J.; Liang, J.; Zhang, Y.; Dong, C.; Tan, D.; Wang, H.; Zheng, Y.; He, Q. Strategic Advances in Targeted Delivery Carriers for Therapeutic Cancer Vaccines. Int. J. Mol. Sci. 2025, 26, 6879. [Google Scholar] [CrossRef]
  14. Sadiq, S.; Khan, S.; Khan, I.; Khan, A.; Humayun, M.; Wu, P.; Usman, M.; Khan, A.; Alanazi, A.F.; Bououdina, M. A Critical Review on Metal-Organic Frameworks (MOFs) Based Nanomaterials for Bio-Medical Applications: Designing, Recent Trends, Challenges, and Prospects. Heliyon 2024, 10, e25521. [Google Scholar] [CrossRef]
  15. Desai, N.; Rana, D.; Salave, S.; Benival, D.; Khunt, D.; Prajapati, B.G. Achieving Endo/Lysosomal Escape Using Smart Nanosystems for Efficient Cellular Delivery. Molecules 2024, 29, 3131. [Google Scholar] [CrossRef] [PubMed]
  16. Gao, J.; Karp, J.M.; Langer, R.; Joshi, N. The Future of Drug Delivery. Chem. Mater. 2023, 35, 359–363. [Google Scholar] [CrossRef] [PubMed]
  17. Desai, N.; Rana, D.; Salave, S.; Gupta, R.; Patel, P.; Karunakaran, B.; Sharma, A.; Giri, J.; Benival, D.; Kommineni, N. Chitosan: A Potential Biopolymer in Drug Delivery and Biomedical Applications. Pharmaceutics 2023, 15, 1313. [Google Scholar] [CrossRef] [PubMed]
  18. Albuquerque, T.; Faria, R.; Sousa, A.; Neves, A.R.; Queiroz, J.A.; Costa, D. Polymer Peptide Ternary Systems as a Tool to Improve the Properties of Plasmid DNA Vectors in Gene Delivery. J. Mol. Liq. 2020, 309, 113157. [Google Scholar] [CrossRef]
  19. Yin, H.; Kanasty, R.L.; Eltoukhy, A.A.; Vegas, A.J.; Dorkin, J.R.; Anderson, D.G. Non-Viral Vectors for Gene-Based Therapy. Nat. Rev. Genet. 2014, 15, 541–555. [Google Scholar] [CrossRef]
  20. Yip, B.H. Recent Advances in CRISPR/Cas9 Delivery Strategies. Biomolecules 2020, 10, 839. [Google Scholar] [CrossRef]
  21. Thermo Fisher Scientific. Lipofectamine® CRISPRMAX™ Cas9 Transfection Reagent. Available online: https://www.thermofisher.com/order/catalog/product/CMAX00008 (accessed on 3 April 2025).
  22. Sigma-Aldrich. PEI Prime™ Linear Polyethylenimine for Gene Delivery. Available online: https://www.sigmaaldrich.com/US/en/product/aldrich/919012 (accessed on 3 April 2025).
  23. Thermo Fisher Scientific. Lipofectamine® 3000 Transfection Reagent. Available online: https://www.thermofisher.com/order/catalog/product/L3000001 (accessed on 3 April 2025).
  24. Promega Corporation. ViaFect™ Transfection Reagent. Available online: https://www.promega.com/products/luciferase-assays/transfection-reagents/viafect-transfection-reagent/ (accessed on 3 April 2025).
  25. Promega Corporation. FuGENE® HD Transfection Reagent. Available online: https://www.promega.com/products/luciferase-assays/transfection-reagents/fugene-hd-transfection-reagent/ (accessed on 3 April 2025).
  26. Thermo Fisher Scientific. Lipofectamine® RNAiMAX Transfection Reagent. Available online: https://www.thermofisher.com/us/en/home/references/protocols/cell-culture/transfection-protocol/rnaimax-reverse-transfections-lipofectamine.html (accessed on 3 April 2025).
  27. Horizon Discovery. DharmaFECT® 1 Transfection Reagent. Available online: https://horizondiscovery.com/en/transfection-and-ancillary-reagents/products/dharmafect-1-transfection-reagent (accessed on 3 April 2025).
  28. Tretbar, U.S.; Rurik, J.G.; Rustad, E.H.; Sürün, D.; Köhl, U.; Olweus, J.; Buchholz, F.; Ivics, Z.; Fricke, S.; Blache, U. Non-Viral Vectors for Chimeric Antigen Receptor Immunotherapy. Nat. Rev. Methods Primers 2024, 4, 74. [Google Scholar] [CrossRef]
  29. El Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  30. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  31. Vader, P.; Mol, E.A.; Pasterkamp, G.; Schiffelers, R.M. Extracellular vesicles for drug delivery. Adv. Drug Deliv. Rev. 2016, 106, 148–156. [Google Scholar] [CrossRef]
  32. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzás, E.I.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Portillo, H.A.D.; et al. Applying extracellular vesicles-based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef]
  33. Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed]
  34. Raguram, A.; Banskota, S.; Liu, D.R. Therapeutic In Vivo Delivery of Gene Editing Agents. Cell 2022, 185, 2806–2827. [Google Scholar] [CrossRef]
  35. Wang, C.; Pan, C.; Yong, H.; Wang, F.; Bo, T.; Zhao, Y.; Ma, B.; He, W.; Li, M. Emerging Non-Viral Vectors for Gene Delivery. J. Nanobiotechnol. 2023, 21, 272. [Google Scholar] [CrossRef]
  36. Zhang, Y.; Wu, Z.-Y. Gene Therapy for Monogenic Disorders: Challenges, Strategies, and Perspectives. J. Genet. Genom. 2024, 51, 133–143. [Google Scholar] [CrossRef]
  37. Giorgioni, L.; Ambrosone, A.; Cometa, M.F.; Salvati, A.L.; Nisticò, R.; Magrelli, A. Revolutionizing CAR T-Cell Therapies: Innovations in Genetic Engineering and Manufacturing to Enhance Efficacy and Accessibility. Int. J. Mol. Sci. 2024, 25, 10365. [Google Scholar] [CrossRef]
  38. Ghassemi, S.; Durgin, J.S.; Nunez-Cruz, S.; Patel, J.; Leferovich, J.; Pinzone, M.; Shen, F.; Cummins, K.D.; Plesa, G.; Cantu, V.A.; et al. Rapid Manufacturing of Non-Activated Potent CAR T Cells. Nat. Biomed. Eng. 2022, 6, 118–128. [Google Scholar] [CrossRef]
  39. Metzloff, A.E.; Padilla, M.S.; Gong, N.; Billingsley, M.M.; Han, X.; Merolle, M.; Mai, D.; Figueroa-Espada, C.G.; Thatte, A.S.; Haley, R.M.; et al. Antigen Presenting Cell Mimetic Lipid Nanoparticles for Rapid mRNA CAR T Cell Cancer Immunotherapy. Adv. Mater. 2024, 36, 2313226. [Google Scholar] [CrossRef]
  40. Magnani, C.F.; Gaipa, G.; Lussana, F.; Belotti, D.; Gritti, G.; Napolitano, S.; Matera, G.; Cabiati, B.; Buracchi, C.; Borleri, G.; et al. Sleeping Beauty-Engineered CAR T Cells Achieve Antileukemic Activity without Severe Toxicities. J. Clin. Investig. 2020, 130, 6021–6033. [Google Scholar] [CrossRef]
  41. Guidotti, G.; Brambilla, L.; Rossi, D. Cell-Penetrating Peptides: From Basic Research to Clinics. Trends Pharmacol. Sci. 2017, 38, 406–424. [Google Scholar] [CrossRef]
  42. Yip, T.; Qi, X.; Yan, H.; Chang, Y. Therapeutic Applications of RNA Nanostructures. RSC Adv. 2024, 14, 28807–28821. [Google Scholar] [CrossRef]
  43. Han, X.; Gong, N.; Xue, L.; Billingsley, M.M.; El-Mayta, R.; Shepherd, S.J.; Alameh, M.-G.; Weissman, D.; Mitchell, M.J. Ligand-Tethered Lipid Nanoparticles for Targeted RNA Delivery to Treat Liver Fibrosis. Nat. Commun. 2023, 14, 75. [Google Scholar] [CrossRef]
  44. Xiao, W.; Jiang, W.; Chen, Z.; Huang, Y.; Mao, J.; Zheng, W.; Hu, Y.; Shi, J. Advance in Peptide-Based Drug Development: Delivery Platforms, Therapeutics and Vaccines. Signal Transduct. Target. Ther. 2025, 10, 74. [Google Scholar] [CrossRef]
  45. Bui, T.A.; Mei, H.; Sang, R.; Ortega, D.G.; Deng, W. Advancements and Challenges in Developing In Vivo CAR T Cell Therapies for Cancer Treatment. eBioMedicine 2024, 106, 105266. [Google Scholar] [CrossRef]
  46. Vermes, K. FDA Approves Investigational New Drug Application for Ceramide NanoLiposome. Pharmacy Times, 14 January 2017. Available online: https://www.pharmacytimes.com/view/fda-approves-investigational-new-drug-application-for-ceramide-nanoliposome (accessed on 20 April 2025).
  47. Kubarek, D. FDA Approves Investigational New Drug Application for Ceramide NanoLiposome. Penn State News, 29 June 2017. Available online: https://www.psu.edu/news/research/story/investigational-cancer-compound-receives-fda-approval-begin-human-trials (accessed on 20 April 2025).
  48. Arcturus Therapeutics. Arcturus’ Pipeline of mRNA Medicines and Vaccines. 2025. Available online: https://arcturusrx.com/mrna-medicines-pipeline/ (accessed on 20 April 2025).
  49. New Drug Approvals. ARCT-021 (LUNAR-COV19). New Drug Approvals, 26 June 2021. [Google Scholar]
  50. Nanobiotix. Pipeline Overview—NBTXR3 in the Clinic. 2025. Available online: https://nanobiotix.com/pipeline/ (accessed on 20 April 2025).
  51. Voss, M.H.; Hussain, A.; Vogelzang, N.; Lee, J.L.; Keam, B.; Rha, S.Y.; Vaishampayan, U.; Harris, W.B.; Richey, S.; Randall, J.M.; et al. A Randomized Phase II Trial of CRLX101 in Combination with Bevacizumab versus Standard of Care in Patients with Advanced Renal Cell Carcinoma. Ann. Oncol. 2017, 28, 2754–2760. [Google Scholar] [CrossRef]
  52. Hamaguchi, T.; Tsuji, A.; Yamaguchi, K.; Takeda, K.; Uetake, H.; Esaki, T.; Amagai, K.; Sakai, D.; Baba, H.; Kimura, M.; et al. A phase II study of NK012, a polymeric micelle formulation of SN-38, in unresectable, metastatic or recurrent colorectal cancer patients. Cancer Chemother. Pharmacol. 2018, 82, 1021–1029. [Google Scholar] [CrossRef]
  53. Szota, M.; Szwedowicz, U.; Rembialkowska, N.; Janicka-Klos, A.; Doveiko, D.; Chen, Y.; Kulbacka, J.; Jachimska, B. Dendrimer Platforms for Targeted Doxorubicin Delivery—Physicochemical Properties in Context of Biological Responses. Int. J. Mol. Sci. 2024, 25, 7201. [Google Scholar] [CrossRef]
  54. Innovation Pharmaceuticals Inc. Stages of Development. Available online: http://www.ipharminc.com/stages-of-development/ (accessed on 22 July 2025).
  55. National Cancer Institute. A Phase I Study of [177Lu]Lu-FF58 in Patients with Advanced Solid Tumors. Available online: https://www.cancer.gov/clinicaltrials/NCI-2024-03006 (accessed on 22 July 2025).
  56. O’Flaherty, R.; Bergin, A.; Flampouri, E.; Mota, L.; Obaidi, H.; Quigley, A.; Fagan, A.; Barron, N.; Clynes, M. Mammalian Cell Culture for Production of Recombinant Proteins: A Review of the Critical Steps in Their Biomanufacturing. Biotechnol. Adv. 2020, 43, 107552. [Google Scholar] [CrossRef] [PubMed]
  57. Vavilis, T.; Stamoula, E.; Ainatzoglou, A.; Sachinidis, A.; Lamprinou, M.; Dardalas, I.; Vizirianakis, I.S. mRNA in the Context of Protein Replacement Therapy. Pharmaceutics 2023, 15, 166. [Google Scholar] [CrossRef] [PubMed]
  58. Kashte, S.; Gulbake, A.; El-Amin, S.F., III; Gupta, A. COVID-19 Vaccines: Rapid Development, Implications, Challenges and Prospects. Hum. Cell 2021, 34, 711–733. [Google Scholar] [CrossRef]
  59. Sayour, E.J.; Boczkowski, D.; Mitchell, D.A.; Nair, S.K. Cancer mRNA Vaccines: Clinical Advances and Future Opportunities. Nat. Rev. Clin. Oncol. 2024, 21, 489–500. [Google Scholar] [CrossRef]
  60. Zeng, C.; Zhang, C.; Walker, P.G.; Dong, Y. Formulation and Delivery Technologies for mRNA Vaccines. Curr. Top. Microbiol. Immunol. 2020, 440, 71–110. [Google Scholar]
  61. Wadhwa, A.; Aljabbari, A.; Lokras, A.; Foged, C.; Thakur, A. Opportunities and Challenges in the Delivery of mRNA-Based Vaccines. Pharmaceutics 2020, 12, 102. [Google Scholar] [CrossRef]
  62. Qureischi, M.; Mohr, J.; Arellano-Viera, E.; Knudsen, S.E.; Vohidov, F.; Garitano-Trojaola, A. mRNA-Based Therapies: Pre-Clinical and Clinical Applications. Int. Rev. Cell Mol. Biol. 2022, 372, 1–54. [Google Scholar]
  63. Eralp, Y. Application of mRNA Technology in Cancer Therapeutics. Vaccines 2022, 10, 1262. [Google Scholar] [CrossRef]
  64. Hajj, K.A.; Whitehead, K.A. Tools for Translation: Non-Viral Materials for Therapeutic mRNA Delivery. Nat. Rev. Mater. 2017, 2, 17056. [Google Scholar] [CrossRef]
  65. Jackson, N.A.C.; Kester, K.E.; Casimiro, D.; Gurunathan, S.; DeRosa, F. The Promise of mRNA Vaccines: A Biotech and Industrial Perspective. NPJ Vaccines 2020, 5, 11. [Google Scholar] [CrossRef]
  66. Yang, W.; Mixich, L.; Boonstra, E.; Cabral, H. Polymer-Based mRNA Delivery Strategies for Advanced Therapies. Adv. Healthc. Mater. 2023, 12, e2202688. [Google Scholar] [CrossRef]
  67. Mali, S. Delivery Systems for Gene Therapy. Indian J. Hum. Genet. 2013, 19, 3–8. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, T.K.; Eberwine, J.H. Mammalian Cell Transfection: The Present and the Future. Anal. Bioanal. Chem. 2010, 397, 3173–3178. [Google Scholar] [CrossRef] [PubMed]
  69. Akinc, A.; Maier, M.A.; Manoharan, M.; Fitzgerald, K.; Jayaraman, M.; Barros, S.; Ansell, S.; Du, X.; Hope, M.J.; Madden, T.D.; et al. The Onpattro Story and the Clinical Translation of Nanomedicines Containing Nucleic Acid-Based Drugs. Nat. Nanotechnol. 2019, 14, 1084–1087. [Google Scholar] [CrossRef]
  70. Wei, T.; Cheng, Q.; Min, Y.-L.; Olson, E.N.; Siegwart, D.J. Systemic Nanoparticle Delivery of CRISPR-Cas9 Ribonucleoproteins for Effective Tissue Specific Genome Editing. Nat. Commun. 2020, 11, 3232. [Google Scholar] [CrossRef]
  71. Kenjo, E.; Hozumi, H.; Makita, Y.; Iwabuchi, K.A.; Fujimoto, N.; Matsumoto, S.; Kimura, M.; Amano, Y.; Ifuku, M.; Naoe, Y.; et al. Low Immunogenicity of LNP Allows Repeated Administrations of CRISPR-Cas9 mRNA into Skeletal Muscle in Mice. Nat. Commun. 2021, 12, 7101. [Google Scholar] [CrossRef]
  72. Li, Y.; Tenchov, R.; Smoot, J.; Liu, C.; Watkins, S.; Zhou, Q. A Comprehensive Review of the Global Efforts on COVID-19 Vaccine Development. ACS Cent. Sci. 2021, 7, 512–533. [Google Scholar] [CrossRef]
  73. Zhang, X.; Zhao, W.; Nguyen, G.N.; Zhang, C.; Zeng, C.; Yan, J.; Du, S.; Hou, X.; Li, W.; Jiang, J.; et al. Functionalized Lipid-Like Nanoparticles for In Vivo mRNA Delivery and Base Editing. Sci. Adv. 2020, 6, eabc2315. [Google Scholar] [CrossRef]
  74. Del Toro Runzer, C.; Anand, S.; Mota, C.; Moroni, L.; Plank, C.; Van Griensven, M.; Balmayor, E.R. Cellular Uptake of Modified mRNA Occurs via Caveolae-Mediated Endocytosis, Yielding High Protein Expression in Slow-Dividing Cells. Mol. Ther. Nucleic Acids 2023, 32, 960–979. [Google Scholar] [CrossRef]
  75. Cardarelli, F.; Digiacomo, L.; Marchini, C.; Amici, A.; Salomone, F.; Fiume, G.; Rossetta, A.; Gratton, E.; Pozzi, D.; Caracciolo, G. The Intracellular Trafficking Mechanism of Lipofectamine-Based Transfection Reagents and Its Implication for Gene Delivery. Sci. Rep. 2016, 6, 25879. [Google Scholar] [CrossRef]
  76. Diorio, C.; Teachey, D.T.; Grupp, S.A. Allogeneic Chimeric Antigen Receptor Cell Therapies for Cancer: Progress Made and Remaining Roadblocks. Nat. Rev. Clin. Oncol. 2025, 22, 10–27. [Google Scholar] [CrossRef]
  77. Maakaron, J.E.; Hu, M.; El-Jurdi, N. Chimeric Antigen Receptor T Cell Therapy for Cancer: Clinical Applications and Practical Considerations. BMJ 2022, 378, e068956. [Google Scholar] [CrossRef]
  78. Cappell, K.M.; Kochenderfer, J.N. Long-Term Outcomes Following CAR T Cell Therapy: What We Know So Far. Nat. Rev. Clin. Oncol. 2023, 20, 359–371. [Google Scholar] [CrossRef]
  79. Jamour, P.; Jamali, A.; Langeroudi, A.G.; Sharafabad, B.E.; Abdoli, A. Comparing Chemical Transfection, Electroporation, and Lentiviral Vector Transduction to Achieve Optimal Transfection Conditions in the Vero Cell Line. BMC Mol. Cell Biol. 2024, 25, 15. [Google Scholar] [CrossRef]
  80. Rahimmanesh, I.; Totonchi, M.; Khanahmad, H. The Challenging Nature of Primary T Lymphocytes for Transfection: Effect of Protamine Sulfate on the Transfection Efficiency of Chemical Transfection Reagents. Res. Pharm. Sci. 2020, 15, 437–446. [Google Scholar] [CrossRef]
  81. Balke-Want, H.; Keerthi, V.; Cadinanos-Garai, A.; Fowler, C.; Gkitsas, N.; Brown, A.K.; Tunuguntla, R.; Abou-El-Enein, M.; Feldman, S.A. Non-Viral Chimeric Antigen Receptor (CAR) T Cells Going Viral. Immuno-Oncol. Technol. 2023, 18, 100375. [Google Scholar] [CrossRef]
  82. VanderBurgh, J.A.; Corso, T.N.; Levy, S.L.; Craighead, H.G. Scalable continuous-flow electroporation platform enabling T cell transfection for cellular therapy manufacturing. Sci Rep. 2023, 13, 6857. [Google Scholar] [CrossRef]
  83. Kitte, R.; Rabel, M.; Geczy, R.; Park, S.; Fricke, S.; Koehl, U.; Tretbar, U.S. Lipid nanoparticles outperform electroporation in mRNA-based CAR T cell engineering. Mol. Ther. Methods Clin. Dev. 2023, 31, 101139. [Google Scholar] [CrossRef]
  84. Dhayalan, M.; Wang, W.; Riyaz, S.U.M.; Dinesh, R.A.; Shanmugam, J.; Irudayaraj, S.S.; Stalin, A.; Giri, J.; Mallik, S.; Hu, R. Advances in functional lipid nanoparticles: From drug delivery platforms to clinical applications. 3 Biotech 2024, 14, 57. [Google Scholar] [CrossRef]
  85. Uslu, U.; June, C.H. Beyond the blood: Expanding CAR T cell therapy to solid tumors. Nat. Biotechnol. 2025, 43, 506–515. [Google Scholar] [CrossRef]
  86. Giulimondi, F.; Digiacomo, L.; Renzi, S.; Cassone, C.; Pirrottina, A.; Molfetta, R.; Palamà, I.E.; Maiorano, G.; Gigli, G.; Amenitsch, H.; et al. Optimizing transfection efficiency in CAR-T cell manufacturing through multiple administrations of lipid-based nanoparticles. ACS Appl. Bio Mater. 2024, 7, 3746–3757. [Google Scholar] [CrossRef] [PubMed]
  87. Luo, J.; Zhang, X. Challenges and Innovations in CAR-T Cell Therapy: A Comprehensive Review. Front. Oncol. 2024, 14, 1399544. [Google Scholar]
  88. Li, J.; Chen, P.; Ma, W. The next frontier in immunotherapy: Potential and challenges of CAR-macrophages. Exp. Hematol. Oncol. 2024, 13, 76. [Google Scholar] [CrossRef] [PubMed]
  89. Kohn, D.B.; Chen, Y.Y.; Spencer, M.J. Successes and challenges in clinical gene therapy. Gene Ther. 2023, 30, 738–746. [Google Scholar] [CrossRef]
  90. Cring, M.R.; Sheffield, V.C. Gene therapy and gene correction: Targets, progress, and challenges for treating human diseases. Gene Ther. 2022, 29, 3–12. [Google Scholar] [CrossRef]
  91. Liu, F.; Li, R.; Zhu, Z.; Yang, Y.; Lu, F. Current developments of gene therapy in human diseases. MedComm 2024, 5, e645. [Google Scholar] [CrossRef]
  92. Konda, P.; Garinet, S.; Van Allen, E.M.; Viswanathan, S.R. Genome-guided discovery of cancer therapeutic targets. Cell Rep. 2023, 42, 112978. [Google Scholar] [CrossRef]
  93. Chu, X.; Tian, W.; Ning, J.; Xiao, G.; Zhou, Y.; Wang, Z.; Zhai, Z.; Tanzhu, G.; Yang, J.; Zhou, R. Cancer stem cells: Advances in knowledge and implications for cancer therapy. Signal Transduct. Target. Ther. 2024, 9, 170. [Google Scholar] [CrossRef]
  94. Zu, H.; Gao, D. Non-viral vectors in gene therapy: Recent development, challenges, and prospects. AAPS J. 2021, 23, 78. [Google Scholar] [CrossRef]
  95. Liu, B.; Zhou, H.; Tan, L.; Siu, K.T.H.; Guan, X.-Y. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct. Target. Ther. 2024, 9, 175. [Google Scholar] [CrossRef]
  96. van der Meel, R.; Wender, P.A.; Merkel, O.M.; Lostalé-Seijo, I.; Montenegro, J.; Miserez, A.; Laurent, Q.; Sleiman, H.; Luciani, P. Next-generation materials for nucleic acid delivery. Nat. Rev. Mater. 2025, 10, 490–499. [Google Scholar] [CrossRef]
  97. Hou, X.; Zaks, T.; Langer, R.; Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 2021, 6, 1078–1094. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, B.; Shen, B.; Xiang, W.; Shen, H. Advances in the study of LNPs for mRNA delivery and clinical applications. Virus Genes 2024, 60, 577–591. [Google Scholar] [CrossRef] [PubMed]
  99. Chen, L.; Hong, W.; Ren, W.; Xu, T.; Qian, Z.; He, Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct. Target. Ther. 2021, 6, 225. [Google Scholar] [CrossRef]
  100. Hu, B.; Zhong, L.; Weng, Y.; Peng, L.; Huang, Y.; Zhao, Y.; Liang, X.-J. Therapeutic siRNA: State of the art. Signal Transduct. Target. Ther. 2020, 5, 101. [Google Scholar] [CrossRef]
  101. Collotta, D.; Bertocchi, I.; Chiapello, E.; Collino, M. Antisense oligonucleotides: A novel frontier in pharmacological strategy. Front. Pharmacol. 2023, 14, 1304342. [Google Scholar] [CrossRef]
  102. Kulkarni, J.A.; Witzigmann, D.; Thomson, S.B.; Chen, S.; Leavitt, B.R.; Cullis, P.R.; Van Der Meel, R. The current landscape of nucleic acid therapeutics. Nat. Nanotechnol. 2021, 16, 630–643. [Google Scholar] [CrossRef]
  103. Zhu, Y.; Zhu, L.; Wang, X.; Jin, H. RNA-based therapeutics: An overview and prospectus. Cell Death Dis. 2022, 13, 644. [Google Scholar] [CrossRef]
  104. Alshaer, W.; Zureigat, H.; Al Karaki, A.; Al-Kadash, A.; Gharaibeh, L.; Hatmal, M.M.; Aljabali, A.A.; Awidi, A. siRNA: Mechanism of action, challenges, and therapeutic approaches. Eur. J. Pharmacol. 2021, 905, 174178. [Google Scholar] [CrossRef]
  105. Chemello, F.; Chai, A.C.; Li, H.; Rodriguez-Caycedo, C.; Sanchez-Ortiz, E.; Atmanli, A.; Mireault, A.A.; Liu, N.; Bassel-Duby, R.; Olson, E.N. Precise correction of Duchenne muscular dystrophy exon deletion mutations by base and prime editing. Sci. Adv. 2021, 7, eabg4910. [Google Scholar] [CrossRef]
  106. McDowall, S.; Aung-Htut, M.; Wilton, S.; Li, D. Antisense oligonucleotides and their applications in rare neurological diseases. Front. Neurosci. 2024, 18, 1414658. [Google Scholar] [CrossRef]
  107. Sang, A.; Zhuo, S.; Bochanis, A.; Manautou, J.E.; Bahal, R.; Zhong, X.B.; Rasmussen, T.P. Mechanisms of action of the US Food and Drug Administration-approved antisense oligonucleotide drugs. BioDrugs 2024, 38, 511–526. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, S.; Heendeniya, S.N.; Le, B.T.; Rahimizadeh, K.; Rabiee, N.; Zahra, Q.U.A.; Veedu, R.N. Splice-modulating antisense oligonucleotides as therapeutics for inherited metabolic diseases. BioDrugs 2024, 38, 177–203. [Google Scholar] [CrossRef] [PubMed]
  109. Lim, K.H.; Han, Z.; Jeon, H.Y.; Kach, J.; Jing, E.; Weyn-Vanhentenryck, S.; Downs, M.; Corrionero, A.; Oh, R.; Scharner, J.; et al. Antisense oligonucleotide modulation of non-productive alternative splicing upregulates gene expression. Nat. Commun. 2020, 11, 3501. [Google Scholar] [CrossRef]
  110. Rinaldi, C.; Wood, M. Antisense oligonucleotides: The next frontier for treatment of neurological disorders. Nat. Rev. Neurol. 2018, 14, 9–21. [Google Scholar] [CrossRef]
  111. Ponti, F.; Campolungo, M.; Melchiori, C.; Bono, N.; Candiani, G. Cationic lipids for gene delivery: Many players, one goal. Chem. Phys. Lipids 2021, 235, 105032. [Google Scholar] [CrossRef]
  112. Aartsma-Rus, A.; Krieg, A.M. FDA approves eteplirsen for Duchenne muscular dystrophy: The next chapter in the eteplirsen saga. Nucleic Acid Ther. 2017, 27, 1–3. [Google Scholar] [CrossRef]
  113. Cullis, P.R.; Felgner, P.L. The 60-year evolution of lipid nanoparticles for nucleic acid delivery. Nat. Rev. Drug Discov. 2024, 23, 709–722. [Google Scholar] [CrossRef]
  114. Lei, J.; Qi, S.; Yu, X.; Gao, X.; Yang, K.; Zhang, X.; Cheng, M.; Bai, B.; Feng, Y.; Lu, M.; et al. Development of mannosylated lipid nanoparticles for mRNA cancer vaccine with high antigen presentation efficiency and immunomodulatory capability. Angew. Chem. Int. Ed. 2024, 63, e202318515. [Google Scholar] [CrossRef]
  115. Haque, M.A.; Shrestha, A.; Mikelis, C.M.; Mattheolabakis, G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int. J. Pharm. X 2024, 8, 100283. [Google Scholar] [CrossRef]
  116. Liu, Y.; Huang, Y.; He, G.; Guo, C.; Dong, J.; Wu, L. Development of mRNA lipid nanoparticles: Targeting and therapeutic aspects. Int. J. Mol. Sci. 2024, 25, 10166. [Google Scholar] [CrossRef]
  117. Lauffer, M.C.; van Roon-Mom, W.; Aartsma-Rus, A.; N = 1 Collaborative. Possibilities and limitations of antisense oligonucleotide therapies for the treatment of monogenic disorders. Commun. Med. 2024, 4, 6. [Google Scholar] [CrossRef]
  118. Amiri, A.; Barreto, G.; Sathyapalan, T.; Sahebkar, A. siRNA therapeutics: Future promise for neurodegenerative diseases. Curr. Neuropharmacol. 2021, 19, 1896–1911. [Google Scholar] [CrossRef]
  119. Baylot, V.; Le, T.K.; Taïeb, D.; Rocchi, P.; Colleaux, L. Between hope and reality: Treatment of genetic diseases through nucleic acid-based drugs. Commun. Biol. 2024, 7, 489. [Google Scholar] [CrossRef]
  120. Sun, J.; Roy, S. Gene-based therapies for neurodegenerative diseases. Nat. Neurosci. 2021, 24, 297–311. [Google Scholar] [CrossRef] [PubMed]
  121. García-González, N.; Gonçalves-Sánchez, J.; Gómez-Nieto, R.; Gonçalves-Estella, J.M.; López, D.E. Advances and challenges in gene therapy for neurodegenerative diseases: A systematic review. Int. J. Mol. Sci. 2024, 25, 12485. [Google Scholar] [CrossRef] [PubMed]
  122. Horch, R.E.; Kneser, U.; Polykandriotis, E.; Schmidt, V.J.; Sun, J.; Arkudas, A. Tissue engineering and regenerative medicine—Where do we stand? J. Cell. Mol. Med. 2012, 16, 1157–1165. [Google Scholar] [CrossRef] [PubMed]
  123. Aljabali, A.A.A.; Mohamed, E.-T.; Murtaza, M.T. Principles of CRISPR-Cas9 technology: Advances in genome editing and emerging trends in drug delivery. J. Drug Deliv. Sci. Technol. 2024, 92, 105338. [Google Scholar] [CrossRef]
  124. Li, Z.H.; Wang, J.; Xu, J.P.; Wang, J.; Yang, X. Recent advances in CRISPR-based genome editing technology and its applications in cardiovascular research. Mil. Med. Res. 2023, 10, 12. [Google Scholar] [CrossRef]
  125. Moffat, J.; Komor, A.C.; Lum, L. Impact of CRISPR in cancer drug discovery. Science 2024, 386, 378–379. [Google Scholar] [CrossRef]
  126. Sampogna, G.; Guraya, S.Y.; Forgione, A. Regenerative medicine: Historical roots and potential strategies in modern medicine. J. Microsc. Ultrastruct. 2015, 3, 101–107. [Google Scholar] [CrossRef]
  127. Yanez Arteta, M.; Kjellman, T.; Bartesaghi, S.; Wallin, S.; Wu, X.; Kvist, A.J.; Dabkowska, A.; Székely, N.; Radulescu, A.; Bergenholtz, J.; et al. Successful reprogramming of cellular protein production through mRNA delivered by functionalized lipid nanoparticles. Proc. Natl. Acad. Sci. USA 2018, 115, E3351–E3360. [Google Scholar] [CrossRef]
  128. Xiong, Y.; Mi, B.B.; Shahbazi, M.A.; Xia, T.; Xiao, J. Microenvironment-responsive nanomedicines: A promising direction for tissue regeneration. Mil. Med. Res. 2024, 11, 69. [Google Scholar] [CrossRef] [PubMed]
  129. Patil, S.; Gao, Y.-G.; Lin, X.; Li, Y.; Dang, K.; Tian, Y.; Zhang, W.-J.; Jiang, S.-F.; Qadir, A.; Qian, A.-R. The Development of Functional Non-Viral Vectors for Gene Delivery. Int. J. Mol. Sci. 2019, 20, 5491. [Google Scholar] [CrossRef] [PubMed]
  130. Chen, J.; Zhao, L.; Wu, H.; Chen, J.; Wu, X.; Zhang, Y.; Lin, A.; Luo, D.; Wang, X.; Lin, M. Development of Novel Cationic Non-Viral Gene Carriers Based on Polyvinylamine Derivatives for siRNA Delivery. Eur. Polym. J. 2025, 236, 114107. [Google Scholar] [CrossRef]
  131. Buntz, B. 100 Cell and Gene Therapy Leaders to Watch in 2025. Drug Discovery and Development, 20 December 2024. Available online: https://www.drugdiscoverytrends.com/100-cell-and-gene-therapy-leaders-to-watch-in-2025/ (accessed on 5 May 2025).
  132. Adam, J. Eight Biotech Companies Advancing the Field of siRNA. Labiotech, 6 June 2024. Available online: https://www.labiotech.eu/best-biotech/sirna-companies/ (accessed on 5 May 2025).
  133. DelveInsight Business Research LLP. 80+ Pharma Companies Unite to Shape the Future of RNA-Based Therapeutics. GlobeNewswire, 16 October 2024. Available online: https://www.globenewswire.com/news-release/2024/10/16/2964389/0/en/80-Pharma-Companies-Unite-to-Shape-the-Future-of-RNA-Based-Therapeutics-DelveInsight.html (accessed on 5 May 2025).
  134. Buntz, B. 50 Leading Cell and Gene Therapy Companies. Drug Discovery Trends, 26 July 2022. Available online: https://www.drugdiscoverytrends.com/50-leading-cell-and-gene-therapy-companies/ (accessed on 5 May 2025).
  135. Knutsen, A. Nonviral platforms streamline gene therapy delivery. Genet. Eng. Biotechnol. News 2023, 43, 22–25. [Google Scholar] [CrossRef]
  136. Fu, Y.; Han, Z.; Cheng, W.; Niu, S.; Wang, T.; Wang, X. Improvement strategies for transient gene expression in mammalian cells. Appl. Microbiol. Biotechnol. 2024, 108, 480. [Google Scholar] [CrossRef]
  137. Zhao, Y.; Sampson, M.G.; Wen, X. Quantify and control reproducibility in high-throughput experiments. Nat. Methods 2020, 17, 1207–1213. [Google Scholar] [CrossRef]
  138. Shin, H.; Park, S.-J.; Yim, Y.; Kim, J.; Choi, C.; Won, C.; Min, D.-H. Recent advances in RNA therapeutics and RNA delivery systems based on nanoparticles. Adv. Ther. 2018, 1, 1800065. [Google Scholar] [CrossRef]
  139. Rinoldi, C.; Zargarian, S.S.; Nakielski, P.; Li, X.; Liguori, A.; Petronella, F.; Presutti, D.; Wang, Q.; Costantini, M.; De Sio, L.; et al. Nanotechnology-assisted RNA delivery: From nucleic acid therapeutics to COVID-19 vaccines. Small Methods 2021, 5, e2100402. [Google Scholar] [CrossRef]
  140. Gilbert, J.; Sebastiani, F.; Arteta, M.Y.; Terry, A.; Fornell, A.; Russell, R.; Mahmoudi, N.; Nylander, T. Evolution of the Structure of Lipid Nanoparticles for Nucleic Acid Delivery: From In Situ Studies of Formulation to Colloidal Stability. J. Colloid Interface Sci. 2024, 660, 66–76. [Google Scholar] [CrossRef]
  141. Herrera-Barrera, M.; Ryals, R.C.; Gautam, M.; Jozic, A.; Landry, M.; Korzun, T.; Gupta, M.; Acosta, C.; Stoddard, J.; Reynaga, R.; et al. Peptide-guided lipid nanoparticles deliver mRNA to the neural retina of rodents and nonhuman primates. Sci. Adv. 2023, 9, eadd4623. [Google Scholar] [CrossRef]
  142. Kwon, M.; Firestein, B.L. DNA transfection: Calcium phosphate method. Methods Mol. Biol. 2013, 1018, 107–110. [Google Scholar] [PubMed]
  143. Nejad Mohammadi, N.; Fayazi Hosseini, N.; Nemati, H.; Moradi-Sardareh, H.; Nabi-Afjadi, M.; Kardar, G.A. Revisiting of Properties and Modified Polyethylenimine-Based Cancer Gene Delivery Systems. Biochem. Genet. 2024, 62, 18–39. [Google Scholar] [CrossRef] [PubMed]
  144. Cai, X.; Dou, R.; Guo, C.; Tang, J.; Li, X.; Chen, J.; Zhang, J. Cationic polymers as transfection reagents for nucleic acid delivery. Pharmaceutics 2023, 15, 1502. [Google Scholar] [CrossRef]
  145. Casper, J.; Schenk, S.H.; Parhizkar, E.; Detampel, P.; Dehshahri, A.; Huwyler, J. Polyethylenimine (PEI) in gene therapy: Current status and clinical applications. J. Control. Release 2023, 362, 667–691. [Google Scholar] [CrossRef] [PubMed]
  146. Salameh, J.W.; Zhou, L.; Ward, S.M.; Santa Chalarca, C.F.; Emrick, T.; Figueiredo, M.L. Polymer-mediated gene therapy: Recent advances and merging of delivery techniques. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 2, e1598. [Google Scholar] [CrossRef]
  147. Johnsen, K.B.; Gudbergsson, J.M.; Skov, M.N.; Pilgaard, L.; Moos, T.; Duroux, M. A comprehensive overview of exosomes as drug delivery vehicles—Endogenous nanocarriers for targeted cancer therapy. Biochim. Biophys. Acta 2014, 1846, 75–87. [Google Scholar] [CrossRef]
  148. O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. Exosomes and their role in immune regulation and cancer. Semin. Cell Dev. Biol. 2020, 102, 55–63. [Google Scholar]
  149. Villemejane, J.; Mir, L.M. Physical methods of nucleic acid transfer: General concepts and applications. Br. J. Pharmacol. 2009, 157, 207–219. [Google Scholar] [CrossRef]
  150. Huang, S.; Henderson, T.R.; Dojo Soeandy, C.; Lezhanska, A.; Henderson, J.T. An efficient low-cost means of biophysical gene transfection in primary cells. Sci. Rep. 2024, 14, 13179. [Google Scholar] [CrossRef]
  151. Napotnik, T.B.; Tamara, P.; Damijan, M. Cell death due to electroporation—A review. Bioelectrochemistry 2021, 141, 107871. [Google Scholar]
  152. Lonez, C.; Lensink, M.F.; Kleiren, E.; Vanderwinden, J.M.; Ruysschaert, J.M.; Vandenbranden, M. Fusogenic activity of cationic lipids and lipid shape distribution. Cell. Mol. Life Sci. 2010, 67, 483–494. [Google Scholar] [CrossRef] [PubMed]
  153. Narum, S.; Deal, B.; Ogasawara, H.; Mancuso, J.N.; Zhang, J.; Salaita, K. An endosomal escape Trojan horse platform to improve cytosolic delivery of nucleic acids. ACS Nano 2024, 18, 6186–6201. [Google Scholar] [CrossRef]
  154. Pei, D.; Buyanova, M. Overcoming endosomal entrapment in drug delivery. Bioconjug. Chem. 2019, 30, 273–283. [Google Scholar] [CrossRef] [PubMed]
  155. Cavalcanti, R.R.M.; Lira, R.B.; Riske, K.A. Membrane fusion biophysical analysis of fusogenic liposomes. Langmuir 2022, 38, 10430–10441. [Google Scholar] [CrossRef]
  156. Grau, M.; Wagner, E. Strategies and mechanisms for endosomal escape of therapeutic nucleic acids. Curr. Opin. Chem. Biol. 2024, 81, 102506. [Google Scholar] [CrossRef]
  157. Gandek, T.B.; van der Koog, L.; Nagelkerke, A. A comparison of cellular uptake mechanisms, delivery efficacy, and intracellular fate between liposomes and extracellular vesicles. Adv. Healthc. Mater. 2023, 12, e2300319. [Google Scholar] [CrossRef]
  158. Tran, V.A.; Thuan Le, V.; Doan, V.D.; Vo, G.N.L. Utilization of functionalized metal-organic framework nanoparticle as targeted drug delivery system for cancer therapy. Pharmaceutics 2023, 15, 931. [Google Scholar] [CrossRef]
  159. Alavi, S.E.; Alavi, S.F.; Koohi, M.; Raza, A.; Ebrahimi Shahmabadi, H. Nanoparticle-integrated metal–organic frameworks: A revolution in next-generation drug delivery systems. J. Pharm. Investig. 2024, 54, 751–783. [Google Scholar] [CrossRef]
  160. Raza, A.; Wu, W. Metal-organic frameworks in oral drug delivery. Asian J. Pharm. Sci. 2024, 19, 100951. [Google Scholar] [CrossRef]
  161. Wang, Z.; Kelley, S.O. Microfluidic technologies for enhancing the potency, predictability and affordability of adoptive cell therapies. Nat. Biomed. Eng. 2025, 9, 803–821. [Google Scholar] [CrossRef] [PubMed]
  162. Hur, J.; Chung, A.J. Microfluidic and nanofluidic intracellular delivery. Adv. Sci. 2021, 8, 2004595. [Google Scholar] [CrossRef] [PubMed]
  163. Loo, J.; Sicher, I.; Goff, A.; Kim, O.; Clary, N.; Alexeev, A.; Sulchek, T.; Zamarayeva, A.; Han, S.; Calero-Garcia, M. Microfluidic transfection of mRNA into human primary lymphocytes and hematopoietic stem and progenitor cells using ultra-fast physical deformations. Sci. Rep. 2021, 11, 21407. [Google Scholar] [CrossRef]
  164. Teo, C. Created in BioRender, Premium Version. 2025. Available online: https://BioRender.com/ludvsnm (accessed on 21 July 2005).
  165. Gupta, A.; Andresen, J.L.; Manan, R.S.; Langer, R. Nucleic acid delivery for therapeutic applications. Adv. Drug Deliv. Rev. 2021, 178, 113834. [Google Scholar] [CrossRef]
  166. Pavlov, R.V.; Akimov, S.A.; Dashinimaev, E.B.; Bashkirov, P.V. Boosting lipofection efficiency through enhanced membrane fusion mechanisms. Int. J. Mol. Sci. 2024, 25, 13540. [Google Scholar] [CrossRef]
  167. Mollé, L.M.; Smyth, C.H.; Yuen, D.; Johnston, A.P.R. Nanoparticles for vaccine and gene therapy: Overcoming the barriers to nucleic acid delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2022, 14, e1809. [Google Scholar] [CrossRef]
  168. Hosseini, S.A.; Kardani, A.; Yaghoobi, H. A comprehensive review of cancer therapies mediated by conjugated gold nanoparticles with nucleic acid. Int. J. Biol. Macromol. 2023, 253, 127184. [Google Scholar] [CrossRef]
  169. Zhou, Y.; Zhang, Y.; Wang, Y.; Li, X.; Liu, Y.; Zhang, Y. Polymers as Efficient Non-Viral Gene Delivery Vectors: The Role of Chemical and Physical Architecture. Polymers 2024, 16, 2629. [Google Scholar] [CrossRef]
  170. Kim, J.; Eygeris, Y.; Ryals, R.C.; Jozić, A.; Sahay, G. Strategies for non-viral vectors targeting organs beyond the liver. Nat. Nanotechnol. 2024, 19, 428–447. [Google Scholar] [CrossRef]
  171. Simonsen, J.B. Lipid nanoparticle-based strategies for extrahepatic delivery of nucleic acid therapies—Challenges and opportunities. J. Control. Release 2024, 370, 763–772. [Google Scholar] [CrossRef]
  172. Lee, S.; Chen, L.; Zhang, X. Non-Viral Gene Delivery Systems for Safe and Targeted Regenerative Therapies. J. Genet. Eng. 2025, 1, 1–20. Available online: https://www.researchgate.net/publication/393567331_Non-Viral_Gene_Delivery_Systems_for_Safe_and_Targeted_Regenerative_Therapies (accessed on 5 May 2025).
  173. Mann, Z.; Sengar, M.; Verma, Y.K.; Rajalingam, R.; Raghav, P.K. Hematopoietic stem cell factors: Their functional role in self-renewal and clinical aspects. Front. Cell Dev. Biol. 2022, 10, 664261. [Google Scholar] [CrossRef]
  174. Lee, H.; Rho, W.Y.; Kim, Y.H.; Chang, H.; Jun, B.H. CRISPR-Cas9 gene therapy: Non-viral delivery and stimuli-responsive nanoformulations. Molecules 2025, 30, 542. [Google Scholar] [CrossRef]
  175. Gong, X.; Chen, Z.; Hu, J.J.; Liu, C. Immunogenic Effects and Clinical Applications of Electroporation-Based Therapies. Vaccines 2024, 12, 42. [Google Scholar]
  176. Qin, S.; Tang, X.; Chen, Y.; Chen, K.; Fan, N.; Xiao, W.; Zheng, Q.; Li, G.; Teng, Y.; Wu, M.; et al. mRNA-based therapeutics: Powerful and versatile tools to combat diseases. Signal Transduct. Target. Ther. 2022, 7, 166. [Google Scholar] [CrossRef] [PubMed]
  177. Lu, R.-M.; Hsu, H.-E.; Perez, S.J.L.P.; Kumari, M.; Chen, G.-H.; Hong, M.-H.; Lin, Y.-S.; Liu, C.-H.; Ko, S.-H. Current landscape of mRNA technologies and delivery systems for new modality therapeutics. J. Biomed. Sci. 2024, 31, 89. [Google Scholar] [CrossRef] [PubMed]
  178. Shen, G.; Liu, J.; Yang, H.; Xie, N.; Yang, Y. mRNA therapies: Pioneering a new era in rare genetic disease treatment. J. Control. Release 2024, 369, 696–721. [Google Scholar] [CrossRef]
  179. Iyer, V.R.; Kaduskar, B.D.; Moharir, S.C.; Mishra, R.K. mRNA biotherapeutics landscape for rare genetic disorders. J. Biosci. 2024, 49, 33. [Google Scholar] [CrossRef]
  180. Wei, P.S.; Thota, N.; John, G.; Chang, E.; Lee, S.; Wang, Y.; Ma, Z.; Tsai, Y.H.; Mei, K.C. Enhancing RNA-lipid nanoparticle delivery: Organ- and cell-specificity and barcoding strategies. J. Control. Release 2024, 375, 366–388. [Google Scholar] [CrossRef]
  181. Xu, X.; Xia, T. Recent advances in site-specific lipid nanoparticles for mRNA delivery. ACS Nanosci. Au 2023, 3, 192–203. [Google Scholar] [CrossRef] [PubMed]
  182. Kowalski, P.S.; Rudra, A.; Miao, L.; Anderson, D.G. Delivering the messenger: Advances in technologies for therapeutic mRNA delivery. Mol. Ther. 2019, 27, 710–728. [Google Scholar] [CrossRef] [PubMed]
  183. Shi, Y.; Shi, M.; Wang, Y.; You, J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct. Target. Ther. 2024, 9, 322. [Google Scholar] [CrossRef] [PubMed]
  184. Pardi, N.; Krammer, F. mRNA vaccines for infectious diseases—Advances, challenges and opportunities. Nat. Rev. Drug Discov. 2024, 23, 838–861. [Google Scholar] [CrossRef]
  185. Liu, C.; Shi, Q.; Huang, X.; Koo, S.; Kong, N.; Tao, W. mRNA-based cancer therapeutics. Nat. Rev. Cancer 2023, 23, 526–543. [Google Scholar] [CrossRef]
  186. Kazemian, P.; Yu, S.Y.; Thomson, S.B.; Birkenshaw, A.; Leavitt, B.R.; Ross, C.J.D. Lipid-nanoparticle-based delivery of CRISPR/Cas9 genome-editing components. Mol. Pharm. 2022, 19, 1669–1686. [Google Scholar] [CrossRef]
  187. Kirian, R.D.; Steinman, D.; Jewell, C.M.; Zierden, H.C. Extracellular vesicles as carriers of mRNA: Opportunities and challenges in diagnosis and treatment. Theranostics 2024, 14, 2265–2289. [Google Scholar] [CrossRef]
  188. Cecchin, R.; Troyer, Z.; Witwer, K.; Morris, K.V. Extracellular vesicles: The next generation in gene therapy delivery. Mol. Ther. 2023, 31, 1225–1230. [Google Scholar] [CrossRef]
  189. Murphy, D.E.; de Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef]
  190. Ezzat, K.; Andaloussi, S.E.L.; Zaghloul, E.M.; Lehto, T.; Lindberg, S.; Moreno, P.M.D.; Viola, J.R.; Magdy, T.; Abdo, R.; Guterstam, P.; et al. PepFect14, a novel cell-penetrating peptide for oligonucleotide delivery in vivo. Nucleic Acids Res. 2011, 39, 2163–2171. [Google Scholar] [CrossRef]
  191. McErlean, E.M.; McCrudden, C.M.; McBride, J.W.; Cole, G.; Kett, V.L.; Robson, T.; Dunne, N.J.; McCarthy, H.O. Rational design and characterisation of an amphipathic cell-penetrating peptide for non-viral gene delivery. Int. J. Pharm. 2021, 596, 120223. [Google Scholar] [CrossRef]
  192. Sahin, U.; Karikó, K.; Türeci, Ö. mRNA-based therapeutics—Developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef] [PubMed]
  193. Simeoni, F.; Morris, M.C.; Heitz, F.; Divita, G. Insight into the mechanism of the peptide-based gene delivery system MPG: Implications for delivery of siRNA. Nucleic Acids Res. 2003, 31, 2717–2724. [Google Scholar] [CrossRef] [PubMed]
  194. Crombez, L.; Aldrian-Herrada, G.; Konate, K.; Nguyen, Q.N.; McMaster, G.K.; Brasseur, R.; Heitz, F.; Divita, G. A new potent secondary amphipathic cell-penetrating peptide for siRNA delivery into mammalian cells. Mol. Ther. 2009, 17, 95–103. [Google Scholar] [CrossRef] [PubMed]
  195. Tao, W.; Zeng, X.; Liu, T.; Xiao, Q.; Wang, M.; Pan, Q.; Zhu, X. Emerging concepts of artificial intelligence for rational design of drug delivery systems. Adv. Drug Deliv. Rev. 2023, 195, 114762. [Google Scholar]
  196. Zhang, Y.; Sun, C.; Zhao, Y. AI-powered prediction of nanocarrier performance for gene therapy. Nano Today 2023, 48, 101716. [Google Scholar]
  197. Cheng, L.; Moseman, J.; Mao, H.-Q. Machine Learning Helps Predict Efficient Lipid Nanoparticle Design. Johns Hopkins Institute for NanoBioTechnology. 5 March 2025. Available online: https://inbt.jhu.edu/machine-learning-helps-predict-efficient-lipid-nanoparticle-design/ (accessed on 5 May 2025).
  198. Wu, K.; Wang, Z.; Yang, X.; Chen, Y.; Han, Z.; Zhang, J.; Liu, L.T. TransMA: An explainable multi-modal deep learning model for predicting properties of ionizable lipid nanoparticles in mRNA delivery. arXiv 2024, arXiv:2407.05736. [Google Scholar] [CrossRef]
  199. Kim, M.J.; Lee, Y.; Kim, S.I.; Moon, H. AI-based integration of single-cell transcriptomics and nanocarrier engineering for targeted gene delivery. Small 2023, 19, e2207883. [Google Scholar]
  200. Lewoczko, E.; Dorsey, Z.; Zou, Y.; Chen, R.; Bong, Y.-S.; Chung, E.; Brown, D.; Long, S.; Shen, D. Abstract 3761: AI-GPT-driven design of novel lipid nanoparticles for targeted and safe mRNA-based cancer immunotherapy. Cancer Res. 2025, 85 (Suppl. S1), 3761. [Google Scholar] [CrossRef]
  201. Sharma, A.; Lysenko, A.; Jia, S.; Boroevich, K.A.; Tsunoda, T. Advances in AI and machine learning for predictive medicine. J. Hum. Genet. 2024, 69, 487–497. [Google Scholar] [CrossRef]
  202. Singh, S.; Kumar, R.; Payra, S.; Singh, S.K. Artificial intelligence and machine learning in pharmacological research: Bridging the gap between data and drug discovery. Cureus 2023, 15, e44359. [Google Scholar] [CrossRef]
  203. Bhujel, R.; Enkmann, V.; Burgstaller, H.; Maharjan, R. Artificial Intelligence-Driven Strategies for Targeted Delivery and Enhanced Stability of RNA-Based Lipid Nanoparticle Cancer Vaccines. Pharmaceutics 2025, 17, 992. [Google Scholar] [CrossRef]
  204. Xu, Y.; Ma, S.; Cui, H.; Chen, J.; Xu, S.; Gong, F.; Golubovic, A.; Zhou, M.; Wang, K.C.; Varley, A.; et al. AGILE platform: A deep learning powered approach to accelerate LNP development for mRNA delivery. Nat. Commun. 2024, 15, 6305. [Google Scholar] [CrossRef]
  205. Zhou, Y.; Wang, Z.; Wu, K.; Yang, X.; Zhang, J.; Liu, L. Lipid nanoparticle structure–activity relationships for mRNA delivery: A data-driven approach. J. Control. Release 2024, 382, 1–12. [Google Scholar]
  206. Wu, K.; Yang, X.; Wang, Z.; Li, N.; Zhang, J.; Liu, L. Data-balanced transformer for accelerated ionizable lipid nanoparticles screening in mRNA delivery. Brief. Bioinform. 2024, 25, bbae186. [Google Scholar] [CrossRef]
Figure 1. Overview of Non-Viral Gene Delivery: Liposomes: Encapsulate genetic material for delivery; Lipid Nanoparticles: Protect and deliver genes efficiently; Polymeric Nanoparticles: Condense DNA/RNA for delivery; Dendrimers: Branched polymers for gene delivery; Cationic Polymers: Bind and protect genetic material; Peptides: Facilitate targeted gene delivery.
Figure 1. Overview of Non-Viral Gene Delivery: Liposomes: Encapsulate genetic material for delivery; Lipid Nanoparticles: Protect and deliver genes efficiently; Polymeric Nanoparticles: Condense DNA/RNA for delivery; Dendrimers: Branched polymers for gene delivery; Cationic Polymers: Bind and protect genetic material; Peptides: Facilitate targeted gene delivery.
Jcm 14 05515 g001
Figure 2. Transfection Driving Therapeutic Development: mRNA-Based Therapies: Instruct cells to produce therapeutic proteins; CAR-T Cell Therapy: Modify T cells to target cancer; Gene Therapy: Alter genes to treat disorders; siRNA: Silence genes to prevent harmful proteins; Regenerative Medicine: Promote tissue repair and regeneration.
Figure 2. Transfection Driving Therapeutic Development: mRNA-Based Therapies: Instruct cells to produce therapeutic proteins; CAR-T Cell Therapy: Modify T cells to target cancer; Gene Therapy: Alter genes to treat disorders; siRNA: Silence genes to prevent harmful proteins; Regenerative Medicine: Promote tissue repair and regeneration.
Jcm 14 05515 g002
Table 1. Non-viral gene delivery systems in clinical trials [37,38,39,40,41,42,43,44,45].
Table 1. Non-viral gene delivery systems in clinical trials [37,38,39,40,41,42,43,44,45].
DescriptionApplicationsClinical Trials
(ClinicalTrials.gov)
References
LiposomesTargeted cancer therapy-Solid TumorsNanoLiposome-Phase 1
(NCT02834611 | Card Results | ClinicalTrials.gov)
Keystone Nano [46,47]
Lipid Nanoparticles (LNPs)mRNA-based enzyme replacementARCT-810-Phase 1 (NCT04416126 | Card Results | ClinicalTrials.gov)
ARCT-032-Phase 2
(NCT06747858 | Card Results | ClinicalTrials.gov)
Arcturus Therapeutics [48,49]
Infectious disease vaccine-COVID-19 mRNA VaccineARCT-021-Phase 2
(NCT04480957 | Card Results | ClinicalTrials.gov)
Polymeric Nanoparticles Radiation enhancer for cancerNBTXR3-Phase 1/2/3
(NCT05039632 | Card Results | ClinicalTrials.gov)
Nanobiotix [50]
Tumor-targeted chemotherapyCRLX101-Phase 1/2a
(NCT02187302 | Card Results | ClinicalTrials.gov)
Lumos Pharma [51]
Nanoparticle drug delivery-Triple-negative Breast Cancer, Small Cell Lung CancerNK012-Phase 2
(NCT00951054 | Card Results | ClinicalTrials.gov)
Nippon Kayaku [52]
DendrimersCancer therapyDEP SN38: (Clinical Trials register—Search for DEP SN38)StarPharma [53]
Cationic Polymers (e.g., PEI)Antimicrobial therapyBrilacidin-Phase 2a
(NCT02052388 | Card Results | ClinicalTrials.gov)
Innovation Pharmaceuticals [54]
PeptideMetastatic tumors177Lu-Integrin-Phase 1
(Study Details | Study to Evaluate the Safety and Activity (Including Distribution) of 177Lu-3BP-227 in Subjects with Solid Tumors Expressing Neurotensin Receptor Type 1. | ClinicalTrials.gov)
PeptiDream [55]
Table 2. Therapies using Non-Viral Gene Delivery Systems [129,130,131,132,133,134,135].
Table 2. Therapies using Non-Viral Gene Delivery Systems [129,130,131,132,133,134,135].
CategoryCompanyDrugDescription
mRNA-based TherapiesModernamRNA-1273COVID-19 vaccine using lipid nanoparticles for delivery.
Pfizer-BioNTechBNT162b2COVID-19 vaccine using lipid nanoparticles for delivery.
Arcturus TherapeuticsARCT-810mRNA therapy for Ornithine Transcarbamylase (OTC) deficiency using lipid nanoparticles.
CureVacCVnCoVCOVID-19 vaccine candidate using lipid nanoparticles.
Translate Bio (Sanofi)MRT5005mRNA therapy for cystic fibrosis using lipid nanoparticles.
BioNTechBNT111mRNA cancer immunotherapy using lipid nanoparticles.
GenEditVariousDeveloping non-lipid, hydrophilic polymer nanoparticles for autoimmune diseases and other indications.
CAR-T TherapiesCellectisUCART19Allogeneic CAR-T therapy for leukemia using TALEN gene editing technology.
Poseida TherapeuticsP-BCMA-101CAR-T therapy for multiple myeloma using piggyBac DNA Modification System.
Precision BioSciencesPBCAR0191CAR-T therapy for B-cell malignancies using ARCUS genome editing technology.
Sana BiotechnologySC291CAR-T therapy for hematologic malignancies using fusogen technology for non-viral delivery.
Allogene TherapeuticsALLO-501Allogeneic CAR-T therapy for non-Hodgkin lymphoma using TALEN gene editing technology.
ArcellxCART-ddBCMACAR-T therapy for multiple myeloma using a novel synthetic binding scaffold.
Gene therapy ElevateBioVariousDeveloping a broad portfolio of cell and gene therapies using non-viral delivery systems.
Tessera TherapeuticsGene Writing™Pioneering Gene Writing technology to treat diseases at their source.
Mediphage Bioceuticalsministring DNA (msDNA)Developing non-viral, safe, and redosable gene therapies using msDNA technology.
Clearside BiomedicalCLS-AXDeveloping therapies for chronic eye diseases using non-viral delivery methods.
Code Biotherapeutics3DNALeveraging a non-viral multivalent synthetic DNA delivery platform for various genetic disorders.
Mana.bioVariousUsing AI-based drug delivery platform for oligonucleotide therapies, including mRNA-based therapeutics.
Nanoscope TherapeuticsMCO-010Developing gene therapies for vision impairment and blindness using non-viral delivery systems.
Generation BioceDNADeveloping non-viral genetic medicines with long-term efficacy and support for redosing.
siRNA and ASO TherapiesAlnylam PharmaceuticalsOnpattro (patisiran)siRNA therapy for hereditary transthyretin-mediated amyloidosis using lipid nanoparticles.
Ionis PharmaceuticalsSpinraza (nusinersen)ASO therapy for spinal muscular atrophy.
Arrowhead PharmaceuticalsARO-AATsiRNA therapy for alpha-1 antitrypsin deficiency using TRiM™ platform.
Dicerna PharmaceuticalsDCR-PHXCsiRNA therapy for primary hyperoxaluria using GalXC™ platform.
Wave Life SciencesWVE-120101ASO therapy for Huntington’s disease using stereopure oligonucleotides.
Silence TherapeuticsSLN360siRNA therapy for cardiovascular disease using GalNAc conjugation.
ProQR TherapeuticsSepofarsenASO therapy for Leber congenital amaurosis 10 (LCA10).
Arbutus BiopharmaAB-729siRNA therapy for chronic hepatitis B using GalNAc conjugation.
OliX PharmaceuticalsOLX101siRNA therapy for hypertrophic scars using asymmetric siRNA technology.
DTx PharmaFALCON platform drugsDeveloping siRNA therapies using Fatty Acid Ligand Conjugated OligoNucleotide (FALCON) platform.
Regenerative MedicineAspen NeuroscienceANPD001Autologous iPSC-derived neuron replacement therapy for Parkinson’s Disease.
Nanoscope TherapeuticsMCO-010Gene therapy for vision impairment and blindness using non-viral delivery systems.
Generation BioceDNADeveloping non-viral genetic medicines with long-term efficacy and support for redosing.
Tessera TherapeuticsGene Writing™Pioneering Gene Writing technology to treat diseases at their source.
Code Biotherapeutics3DNALeveraging a non-viral multivalent synthetic DNA delivery platform for various genetic disorders.
Clearside BiomedicalCLS-AXDeveloping therapies for chronic eye diseases using non-viral delivery methods.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Simkhada, D.; Teo, S.H.C.; Deorkar, N.; Vemuri, M.C. Transfection Technologies for Next-Generation Therapies. J. Clin. Med. 2025, 14, 5515. https://doi.org/10.3390/jcm14155515

AMA Style

Simkhada D, Teo SHC, Deorkar N, Vemuri MC. Transfection Technologies for Next-Generation Therapies. Journal of Clinical Medicine. 2025; 14(15):5515. https://doi.org/10.3390/jcm14155515

Chicago/Turabian Style

Simkhada, Dinesh, Su Hui Catherine Teo, Nandu Deorkar, and Mohan C. Vemuri. 2025. "Transfection Technologies for Next-Generation Therapies" Journal of Clinical Medicine 14, no. 15: 5515. https://doi.org/10.3390/jcm14155515

APA Style

Simkhada, D., Teo, S. H. C., Deorkar, N., & Vemuri, M. C. (2025). Transfection Technologies for Next-Generation Therapies. Journal of Clinical Medicine, 14(15), 5515. https://doi.org/10.3390/jcm14155515

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop