In vivo vectorization and delivery systems for gene therapies and RNA-based therapeutics in oncology

Julie Schock Vaiani a, Mans Broekgaarden a, Jean-Luc Coll a, Lucie Sancey a and Benoit Busser *abc
aUniv. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France. E-mail: bbusser@chu-grenoble.fr
bGrenoble Alpes Univ. Hospital (CHUGA), 38043 Grenoble, France
cInstitut Universitaire de France (IUF), 75005 Paris, France

Received 20th December 2024 , Accepted 27th January 2025

First published on 28th January 2025


Abstract

Gene and RNA-based therapeutics represent a promising frontier in oncology, enabling targeted modulation of tumor-associated genes and proteins. This review explores the latest advances in payload vectorization and delivery systems developed for in vivo cancer treatments. We discuss viral and non-viral organic particles, including lipid based nanoparticles and polymeric structures, for the effective transport of plasmids, siRNA, and self-amplifying RNA therapeutics. Their physicochemical properties, strategies to overcome intracellular barriers, and innovations in cell-based carriers and engineered extracellular vesicles are highlighted. Moreover, we consider oncolytic viruses, novel viral capsid modifications, and approaches that refine tumor targeting and immunomodulation. Ongoing clinical trials and regulatory frameworks guide future directions and emphasize the need for safe, scalable production. The potential convergence of these systems with combination therapies paves the way toward personalized cancer medicine.


image file: d4nr05371k-p1.tif

Julie Schock Vaiani

Dr Julie Schock Vaiani, PharmD, is currently completing a dual-degree program in pharmacy with a focus on industrial applications and biotechnology engineering through an apprenticeship at the Technical Innovation Unit of the Brain Institute in Paris. Demonstrating a strong commitment to advancing her expertise, she has deepened her knowledge in oncology through a specialised university diploma at the Gustave Roussy Institute and an internship at Goethe Universität Klinikum, where she contributed to research on cellular immunotherapies, specifically CAR-NK cells, for glioblastoma treatment. Julie will enter a PhD program at the Brain Institute in Paris.

image file: d4nr05371k-p2.tif

Mans Broekgaarden

Dr Mans Broekgaarden is a tenured principal investigator of INSERM, and works at the Grenoble-Alpes University in France. He obtained his PhD from the University of Amsterdam in 2016 on liposomal photosensitizing agents for cancer therapy. As a post-doc, he joined the group Tayyaba Hasan at the Wellman Center for Photomedicine. He joined the team of Dr Jean-Luc Coll at the Institute for Advanced Bioscience in Grenoble in 2017, where he was awarded a tenured position in 2020. His group investigates light- and radiotherapy-controlled release of chemotherapeutics with the support of an ERC Starting Grant.

image file: d4nr05371k-p3.tif

Jean-Luc Coll

Dr Jean-Luc Coll is Director of Research at INSERM and Team leader in the Institute for Advanced Biosciences in Grenoble. Dr Coll is a biologist with strong experience in oncology. He is using fluorescence, nanotechnologies, chemistry and large instruments for physics, to develop theranostic nanoparticles for the treatment of tumors and their metastases. His team is in particular focused on the use of near-infrared labeled nanoparticles to target tumors, guide surgery and enhance radiotherapy, phototherapy or innovative therapies. In addition to the developments of nanovectors, he is also involved in the generation of adapted medical devices.

image file: d4nr05371k-p4.tif

Lucie Sancey

Dr Lucie Sancey is Director of Research of the French CRNS at the Institute for Advanced Biosciences, in Grenoble, France. Her research activities focus on the development and evaluation of innovative compounds for cancer imaging and treatment. This includes the development of nano-agents for cancer diagnosis and therapy, in particular for neutron capture therapy, as well as new multimodal optical contrast agents and new imaging techniques for biological tissues including fluorophores for image-guided surgery. She is currently the vice-president of the French Society of Nanomedicine, SFNano.

image file: d4nr05371k-p5.tif

Benoit Busser

Dr Benoit Busser (PharmD, PhD) is a Full Professor of Biochemistry and Cancer Sciences at the School of Pharmacy, Grenoble Alpes University, France, and leads the “Metals and Health” Group at the Institute for Advanced Biosciences. He heads the Point-of-Care Testing Unit in the Laboratory Medicine Department at Grenoble Hospital and is a Junior Fellow of the Institut Universitaire de France. Recipient of the ESMI Young Investigator Award (2015), he directs the LIBS Tissue Elemental Imaging Facility, has mentored six PhD students, authored 50 peer-reviewed articles and six book chapters, cofounded two startups, and is a coinventor on three patents.


1. Introduction

Nucleic acids (NAs) have emerged as transformative therapeutic molecules, with their conceptual origins dating back to the 1960s. The 1990s marked significant progress, including the first successful demonstration of in vivo gene expression via mRNA injection and the advent of RNA interference (RNAi). These breakthroughs, coupled with recent innovations like mRNA vaccines, have positioned NAs as versatile tools in addressing complex diseases such as cancer. Unlike traditional therapies, which are often limited by toxicity and resistance, NAs offer sequence-specific approaches to target oncogenic drivers, restore cellular functions, and enhance immunotherapy. With cancer representing the focus of over 68% of gene therapy clinical trials,1 NAs hold immense promise in overcoming tumour heterogeneity and microenvironmental challenges.2,3 However, hurdles such as optimizing delivery, sustaining gene expression, and mitigating vector toxicity remain critical.4 Advancements in sequencing and delivery technologies are rapidly transforming NAs into pivotal components of next-generation cancer therapies. In this review, we examine the diverse payloads utilized in gene and RNA-based therapies, recent advancements in vectorization strategies, including viral and organic vectors, as well as ongoing clinical trials and future prospects for cancer treatment.

2. Gene therapy and RNA-based therapeutics in oncology

The development of biotechnological therapies has necessitated robust regulatory frameworks to ensure innovation and address evolving healthcare needs.5 In the EU, regulation No. 1394/2007, along with Directive 2009/120/CE, defines Gene Therapy Medicinal Products (GTMPs) as biological medicines, excluding vaccines such as mRNA COVID-19 vaccines. Historically, vaccines have been confined to immunological drugs for infectious diseases, excluding anti-cancer therapies. For example, Moderna's mRNA-4157/V940, combined with Keytruda to enhance tumour immunity, cannot yet be classified as a vaccine under current EU definitions. In contrast, the FDA does not require a biological origin for gene therapies and categorizes oligonucleotides like siRNA as drugs targeting RNA within cells. The key gene and RNA-based therapies currently under clinical evaluation, as well as those already approved, are presented in ESI 1. This review distinguishes between gene and RNA-based therapies, considering their unique strategies and regulatory nuances.

2.1. Principles of gene therapy in oncology

Gene therapy in oncology involves introducing DNA to modify cells—cancerous or immune—or modulate immune responses. It targets cancer-related genes or pathways by replacing faulty genes, silencing oncogenes, or enhancing immune responses (immune-gene therapy). Approaches include gene editing, addition, or silencing, delivered in vivo or ex vivo, with varying tools and vectors depending on tumour characteristics (see Fig. 1). Ex vivo therapy involves harvesting a patient's cells, modifying them genetically, and reinfusing them. CAR-T (Chimeric Antigen Receptor T-cell) therapy,6,7 widely used in leukaemia and lymphoma, relies on viral vectors for transduction, which raises production costs and risks such as B-cell depletion, Cytokine Release Syndrome (CRS), and secondary leukaemia due to random integration.8 Efforts are expanding to include Natural Killer (NK) cells and CAR-based therapies for solid cancers.9In vivo therapy administers therapeutic nucleic acids directly to the patient, eliminating the need for external cell manipulation. This approach is being explored for broader applications, including targeting solid tumours and delivering genetic material efficiently within the patient's body.
image file: d4nr05371k-f1.tif
Fig. 1 Ex vivo versus in vivo gene therapies.
2.1.1. Molecular tools for gene therapy. Molecular tools in gene therapy encompass techniques and nucleic elements (“payloads”) designed to modify the genetic material of cancerous or healthy cells for therapeutic purposes (Fig. 2).
image file: d4nr05371k-f2.tif
Fig. 2 Molecular tools for gene and RNA-based therapy.

These nucleic acids possess distinct physicochemical properties, such as a polar sugar-phosphate backbone and hydrophobic nitrogenous bases, which influence solubility and can be enhanced through chemical modifications to resist enzymatic degradation.


2.1.1.1. DNA plasmids carrying a therapeutic gene. Plasmids are small, circular DNA molecules used in gene therapy as delivery vehicles to introduce therapeutic genes into cells, encoding functional RNA. Plasmids can encode various genes, such as cytokines, suppressors, and endonucleases, offering a flexible platform for gene therapy. Episomal plasmids remain extra-chromosomal and enable transient gene expression, while integrative plasmids incorporate into the host genome, allowing stable expression regulated by host mechanisms. The choice between these types depends on treatment goals, such as genetic stability and persistence.10 Integrative plasmids, such as those utilizing transposon systems like Sleeping Beauty (SB)11 or piggyBac (PB), provide an alternative to viral vectors for stable genome integration. Transposons function via a “cut-and-paste” mechanism where transposases facilitate insertion of the gene of interest (GOI) into safe genomic sites. These systems are particularly valuable for cells like NK cells, which are poorly transduced by viral vectors, and have shown promising results in generating CAR immune cells for cancer immunotherapy.12–14 Clinical applications include a Phase II trial using SB to engineer T cells reactive against cancer neoantigens (NCT04102436).15 Future advancements, such as combining transposons with CRISPR/Cas9 or functionalized nanoparticles, aim to enhance efficiency and safety in gene delivery.16–18 Tumour-specific promoters in plasmids enable targeted gene expression by leveraging transcription factors abundant in cancer cells, while tissue-specific or inducible promoters, like the widely used CMV promoter, allow versatile regulation.19,20
2.1.1.2. Gene/genome editing with molecular scissors. Gene and genome editing, using programmable nucleases like CRISPR/Cas9 or transcription activator-like effectors (TALEs) and zinc fingers,21 has revolutionized therapeutic strategies by enabling precise modifications to DNA through double-strand breaks (DSBs) or more advanced methods like single-strand breaks (SSBs). CRISPR/Cas9 utilizes RNA-guided Cas9 enzymes to target DNA, allowing for gene disruption, insertion, or correction, with applications in both ex vivo and in vivo therapies.22 While CRISPR-based therapies, such as CASGEVY, have reached clinical application,23 challenges like off-target effects, genotoxicity, and risks associated with DSBs in TP53-deficient cancers persist.24,25 Alternative approaches, including base editing (BE) and prime editing (PE), enable precise modifications without introducing DSBs, significantly reducing risks.26 BE alters single nucleotide pairs via enzymatic deamination, while PE utilizes a Cas9-reverse transcriptase complex for targeted insertions or corrections guided by pegRNA.27–29 These advanced tools, particularly in combination with CRISPR systems, hold promise for safer and more effective therapies targeting genes like KRAS in cancer.27

2.2. Principles of RNA-based therapy in oncology

RNA-based therapeutics offer innovative strategies for modulating gene expression, with potential to treat various diseases, including cancer. Several RNA therapies, such as RNA interference drugs (e.g., inclisiran, patisiran, givosiran, lumasiran, teprasiran) and antisense oligonucleotides (e.g., nusinersen, eplontersen, tofersen), have received FDA and EMA approval, distinguishing themselves from gene therapies in manufacturing and mechanisms of action. While anti-cancer siRNAs, miRNAs, and antisense nucleotides (ASOs) are not yet commercially available, numerous candidates are in clinical development, highlighting their therapeutic promise (Fig. 2).30
2.2.1. Molecular tools for RNA-based therapy.
2.2.1.1. Coding RNA. Coding RNAs, such as mRNA, are single-stranded molecules characterized by a 5′ cap, a 3′ polyadenylated tail, and tailored untranslated regions (UTRs) that enhance stability and translation efficiency.31 Unlike plasmid DNA, mRNA functions without requiring nuclear entry or genome integration, making it a streamlined therapeutic option. Bicistronic mRNAs, capable of encoding two proteins from a single molecule, highlight advanced applications. For instance, OTX-2002, currently under clinical investigation (NCT05497453), targets the MYC oncogene in cancer through epigenetic regulation, encoding two distinct proteins that modulate gene expression pre-transcriptionally. To address the limitations of ex vivo CAR-T therapies—such as high costs and prolonged persistence—mRNA-transcribed CARs delivered via ionizable lipid nanoparticles (LNPs) present a significant advancement, bypassing the need for viral vectors and enabling transient, safer gene expression with fewer complications.32–34 Additionally, gene-editing tools like Cas9 can be delivered as mRNA, reducing immunogenicity compared to protein-based systems and facilitating precise genome editing. However, off-target and on-target effects, especially in TP53-mutated cancer cells, require careful monitoring during therapy.21,31,35,36 Another promising approach is self-amplifying mRNA (samRNA), which combines encoding a target protein with replication genes to amplify RNA messages post-transfection. Platforms such as naked RNA, DNA-launched replicons, and viral replicon particles have demonstrated efficacy in inducing anti-tumour immunity, which can be further enhanced through combination therapies like chemotherapy or immune checkpoint inhibitors. This approach simplifies production and offers a powerful tool for cancer immunotherapy.37,38
2.2.1.2. Non-Coding RNA (ncRNAs). ncRNAs play crucial roles in gene regulation and cancer therapy by modulating gene expression without encoding proteins. RNAi therapeutics, including small interfering RNAs (siRNAs)39 and microRNAs (miRNAs),40 selectively target mRNA to downregulate protein production. Synthetic miRNA mimics, such as MRX34[thin space (1/6-em)]41 and miR-193a-3p mimic,42 and anti-miRNAs, like anti-miR-21[thin space (1/6-em)]43 and anti-miR-155,44 have shown preclinical and clinical promise in restoring tumour suppressor functions and inhibiting cancer progression. siRNAs guide the RNA-induced silencing complex (RISC) to complementary mRNA, enabling targeted cleavage but requiring repeated dosing due to their transient effects. Small hairpin RNAs (shRNAs) provide gene silencing by integrating into cellular DNA, producing siRNA continuously.45 Despite stability challenges, siRNA modifications and delivery innovations, such as lipid encapsulation, have improved therapeutic potential, exemplified by patisiran, the first FDA-approved siRNA drug.46 Long non-coding RNAs (lncRNAs), including circular RNAs (circRNAs), influence transcription, epigenetic modifications, and mRNA stability through diverse interactions with nucleic acids and proteins. Therapeutics based on lncRNAs and circRNAs are emerging, with applications such as miRNA sponges or oncolytic RNA therapies targeting specific cancer pathways.47–49
2.2.1.3. Antisense oligonucleotides (ASOs). ASOs are synthetic single-stranded DNA or RNA analogues, typically 18–30 nucleotides long, designed to bind specific mRNA or non-coding RNA targets.45 Chemical modifications, such as phosphorothioate backbones and 2′-O-methoxyethyl groups, have enhanced their nuclease resistance, potency, and patient tolerance.50 ASOs have evolved through three generations, with improvements in binding affinity, cell penetration, and reduced off-target effects.51,52 They function either by recruiting RNase H to degrade mRNA or by steric blocking to alter splicing. In oncology, ASOs like anti-HSP27 (BOREALIS-2 trial)53 and anti-STAT354 show promise in treating metastatic urothelial carcinoma and myelodysplastic syndrome. However, few ASOs have reached clinical use, and none currently utilize nanoparticle-based delivery.

2.3. Strategic objectives of gene and RNA-based therapies

Gene and RNA-based therapies aim to modulate gene expression, restore normal gene functions, enhance immune responses, and induce targeted cancer cell death, offering innovative avenues for effective cancer treatment.
2.3.1. Gene negative regulation. Gene negative regulation involves silencing or reducing the expression of oncogenes or upregulated genes to counteract cancer progression. Knockout strategies completely eliminate gene activity using tools like CRISPR/Cas9, which introduces double-strand breaks repaired by the NHEJ or HDR pathway. For instance, knockout of hTERT, a component of telomerase, disrupts cancer cell immortality,55 while targeting KRAS mutations, such as c.35G > A, holds promise in resensitising tumours to tyrosine kinase inhibitors (TKIs).56,57 However, challenges include the multifaceted roles of target genes in cellular homeostasis.58Knock-in approaches introduce precise genetic modifications, such as stop codons to truncate dysfunctional proteins. In pancreatic cancer, researchers used CRISPR/Cas9 to introduce mutations in BRCA1/2, sensitizing cells to PARP inhibitors like olaparib and enhancing apoptosis.59Knockdown approaches reduce, rather than eliminate, gene expression using RNAi tools like siRNAs and ASOs. These methods enable multi-gene silencing, an advantage for network-based approaches in cancers such as breast cancer.60
2.3.2. Add/restore gene expression. Adding or restoring gene expression can counteract the loss of tumour suppressors or enhance immune responses. Cytokines such as IL-2 stimulate anti-tumour immunity by activating effector lymphocytes like memory T cells and NK cells. High-dose IL-2 (proleukin) therapies have shown efficacy but are limited by significant toxicities. Alternative delivery methods, such as plasmid DNA encoding IL-2 or IL-2-targeted mRNA, aim to minimize side effects.61–63 The SB transposon system, tested in a preclinical glioma model, enhanced IFN-γ immunotherapy by prolonging transgene expression, leading to improved survival and tumour regression compared to episomal plasmid expression.64Emerging neoantigen-based therapies, such as GRT-C901 and GRT-R902, use viral and mRNA vectors to stimulate T-cell responses against tumour-specific mutations. These strategies, combined with immune checkpoint inhibitors under the GRANITE phase II/III program (NCT05141721), show promise in advanced cancers, improving immune responses and patient survival. RNA-based antibodies, like BNT141, offer advantages in production speed and pharmacokinetics. For instance, BNT141 targets claudin-18.2, a protein overexpressed in gastric and pancreatic cancers, showing potential as a targeted therapy.65 Similarly, plasmid DNA-encoded antibodies have demonstrated efficacy in HER2-positive breast cancer models, providing long-term expression and tumour inhibition.66 Checkpoint inhibitors, such as anti-PD-L1 and anti-CTLA-4 antibodies, enhance T-cell activity against tumours. RNA-based approaches, including siRNA targeting PD-L1, offer alternative methods for delivering immune-modulating therapies, particularly in immune-privileged regions like the brain. Preclinical studies using vectors like HVJ-E demonstrated strong antitumor immunity and prolonged survival in glioblastoma models.67

Restoring tumour suppressor genes, such as TP53 and PTEN, offers another avenue.68 mRNA encoding functional versions of these genes has shown potential in preclinical models,69,70 particularly when combined with checkpoint inhibitors.71 CRISPR-based activation of PTEN using dCas9-VPR has also demonstrated success in upregulating tumour suppressor activity in cancer cells.72 TALEN-mediated targeting of HPV E6 and E7 oncogenes effectively induced apoptosis and inhibited tumorigenicity in HPV-positive cervical cancer models, ultimately restoring tumour suppressors like p53 and RB1.73

2.3.3. Gene-directed enzyme prodrug therapy (GDEPT). GDEPT delivers a gene encoding an enzyme that activates a non-toxic prodrug into its cytotoxic form within tumour cells.74 HSV-thymidine kinase (HSV-TK), delivered via viral vectors or transposon systems, converts ganciclovir into a toxic compound, inducing localized tumour cell death with bystander effects enhancing efficacy.75 Similarly, cytosine deaminase converts 5-fluorocytosine into the cytotoxic 5-fluorouracil, though clinical trials like Toca 5 have faced setbacks in demonstrating significant survival benefits.76

3. Strategies in vectorization for gene and RNA-based therapies

The effective delivery of nucleic acids, referred to as “payloads”, is a cornerstone of gene and RNA-based therapies, addressing critical challenges in therapeutic development. These payloads, including plasmid DNAs, siRNAs, and mRNAs, face substantial barriers due to their physicochemical properties, such as hydrophilicity, negative charge, and large molecular size. For example, the size of mRNA (∼400 kDa) starkly contrasts with small molecules like aspirin (0.18 kDa), creating significant hurdles in crossing cellular membranes. Furthermore, the negative charge of the sugar-phosphate backbone is repelled by the similarly charged cellular membrane, complicating cellular entry. These issues are exacerbated by serum nucleases that rapidly degrade nucleic acids, reducing their stability and therapeutic efficacy. Delivery systems, or vectors, are indispensable for overcoming these barriers, protecting nucleic acids from enzymatic degradation, extending their circulation time, and facilitating cellular uptake. Additionally, vectors can help navigate specific challenges, such as crossing the nuclear membrane or targeting solid tumours via abnormal vasculature, which are critical for achieving therapeutic success in gene and RNA therapies (Fig. 3).
image file: d4nr05371k-f3.tif
Fig. 3 Patho-physiological barriers to overcome for delivery systems.

The term “vector” refers to the carriers used to deliver nucleic acids to their target sites. No single vector is universally effective for all cancers; their selection must be tailored to the specific phenotypic profile of the cancer type, tumour microenvironment (TME), and the patient's pathophysiological state. This complexity underscores the need for careful vector design to ensure precise targeting and therapeutic efficacy. Strategies for vectorization are broadly categorized into viral and non-viral systems, such as nanoparticles, each with distinct advantages and challenges. The choice between these approaches requires careful consideration of safety, efficiency, and specificity, as well as the unique demands of the therapy and the underlying disease.

3.1. Viral transduction of nucleic acids

Viruses are natural vectors evolved to efficiently deliver their genetic material into host cells, making them ideal tools for gene therapy. Leveraging their ability to enter cells and initiate gene expression, viral vectors have become the most advanced platforms for transferring therapeutic nucleic acids.1,10 Early efforts in viral gene therapy, beginning in the 1980s with recombinant adeno-associated viruses (AAVs), demonstrated the potential for correcting monogenic diseases. Despite initial success, challenges such as random insertional mutagenesis and adverse immune responses highlighted the need for safer, more precise designs.77 Modern recombinant viral vectors are stripped of pathogenic elements and engineered to enhance specificity and safety. For cancer therapies, viruses can be tailored to target cancer cells through genetic modifications. This specificity capitalizes on unique characteristics of tumour cells, including defective interferon signalling, frequent division, and immune evasion. Oncolytic viruses, such as poxviruses encoding hNIS, exemplify these modifications, combining tumour cell targeting with imaging and antitumor efficacy.78,79 Viral vectors can be used for two main purposes: delivering therapeutic genes or as oncolytic agents, with some designs achieving both. Their versatility allows for integration into diverse therapeutic strategies, such as adoptive immune cell therapy using retroviral vectors like lentiviruses, and non-integrative vectors such as adenoviruses and AAVs (Fig. 4).80
image file: d4nr05371k-f4.tif
Fig. 4 Viral transduction with integrative or non-integrative viruses.

Complex engineering of capsids or envelopes redirects viral tropism to specific cell types, while chimeric viruses harness features of multiple viral types to optimize delivery.

Despite their advantages, viral vectors pose challenges, including high production costs and potential immunogenicity. Manufacturing under good practice standards involves co-transfection of producer cells and advanced purification methods, which are resource-intensive. Moreover, immune responses triggered by viral vectors require careful management to minimize toxicity.80,81 This chapter focuses on the role of viral vectors in delivering therapeutic genes, with or without additional oncolytic functions, highlighting their application in cancer gene therapy trials (see Table 1).

Table 1 Main viruses used in cancer gene therapy
Characteristics Integrative virus Non-integrative virus
Name Lentivirus Foamy virus Moloney Murine Leukaemia Virus Adenovirus (AdV) Herpes simplex virus (HSV1, HSV) Vaccinia
a Data sourced from ClinicalTrials.gov, last accessed September 2024. NA, not available
Number of open clinical trialsa 0 (soon) NA NA 179 38 31
Transgene expression Stable, long-term expression via genome integration Stable, long-term expression via genome integration Stable Transient, typically non-integrative (rare integration events possible) Transient Transient
Genome ssRNA dsDNA RNA with reverse transcriptase dsDNA dsDNA dsDNA
Envelope YES (glycoprotein envelope) YES (lipid envelope) YES NO YES YES
Packaging capacity or cloning capacity 8–10 kb 9–10 kb 8 kb 30 kb 20 kb 25 kb
Size (diameter) 80–100 nm 80–100 nm 100 nm 70–100 nm 150–200 nm ∼300 nm
Entry method Endocytosis + membrane fusion (via glycoprotein) Endocytosis followed by host membrane fusion Receptor mCAT-1 followed by endocytosis CD46 and coxsackie/adenovirus receptor (CAR) HVEM, Nectin 1; Nectin 2 Receptor-mediated endocytosis
Immunogenicity Low (envelope incorporates HLA-I from packaging cell membranes during budding) Low (less than other retroviruses) Medium High High High
Safety Risk of insertional mutagenesis; improved safety with self-inactivating (SIN) 3rd generation vectors Considered safe with modifications; low pathogenic potential; no human disease association from natural infection Highly mutagenic Immunogenic, inflammatory Pathogenic to humans; requires engineering. Neurotropic; complex genome complicates modifications Immunogenic, inflammatory
Elimination & biodegradability Viral debris cleared by immune system over time Degraded intracellularly post-infection. Likely degraded intracellularly post-infection, similar to other retroviruses. Not typically specified for adenoviruses, but likely degraded intracellularly post-infection Degraded intracellularly post-infection; may persist in latent forms in specific cases Degraded intracellularly post-infection; large genome and cytoplasmic replication may allow persistence depending on immune response
Most advanced clinical stage Approved for ex vivo gene therapy; preclinical stage for in vivo applications Preclinical stage Phase III (Toca 511) Approved: Oncolytic (oncorine, gendicine) Approved: Imlygic (oncolytic + GM-CSF) Phase III: Oncolytic (Pexa-Vex)
Vector-based (adstiladrin with IFN-α2b) Vector-based (GDEPT): Oncovirac/oncorine
Advantages -Stable, long-term transgene expression -Long-term stable expression -Stable genome expression -Efficient transduction of dividing and non-dividing cells. -Large genome allows for multiple transgene insertions -Replicates entirely in the cytoplasm
-Infects both dividing and non-dividing cells -Low immunogenicity -Modifiable for enhanced tropism -Non-mutagenic and replicative oncolytic properties -Efficient tumour infiltration -No risk of genomic integration
-Broad host range with VSV-G pseudotyping -Integration of transgene without unfavourable recombination -Easy to manipulate. -Easy to manipulate
-High-level gene expression -Rapid replication in infected cells
Disadvantages -Risk of insertional mutagenesis Potential for random gene insertion, increasing the risk of insertional mutagenesis. -Possibility of mutagenesis from incorrect insertion -Capsid and transgene can be highly immunogenic -Pathogenic to humans, requiring engineering for safety -Immune response to viral proteins
-Limited packaging capacity -Infects dividing cells only -Pre-existing antibodies (anti-AAV, AdV) may lead to rapid vector clearance -High immunogenicity and widespread pre-existing anti-HSV immunity, causing vector clearance -Unknown actions of many genes
-Potential for immune response activation -Low production titres -Low production titres hinder large-scale applications -Potential cytopathic effects on target cells
-Requires stringent safety measures for manipulation


3.1.1. Mechanisms and strategies in viral gene delivery. Viral vectors deliver genetic material to target cells either by integrating it into the host genome, enabling stable long-term expression, or maintaining it as episomal DNA for transient expression. This choice depends on the therapeutic objective and the nature of the vector. Oncolytic viruses, for instance, target cancer cells for lysis and stimulate immune responses through tumour antigen release and inflammatory factor secretion, while also enabling transgene delivery. Emerging designs combine oncolytic activity with transgene vectorization to enhance therapeutic outcomes.82

The specificity of transgene expression in viral vectors relies on promoters. Strong, ubiquitous promoters like CMV and CAG ensure robust gene expression but risk non-specific activity, whereas tumour-specific promoters, such as engineered AFP variants for liver cancer, enhance expression in targeted cells while minimizing off-target effects.83–85 Inducible systems, like the Ad-RTS-hIL-12 platform, further refine control by activating transgenes only in the presence of exogenous ligands, exemplifying precise regulation.86

To improve targeting, viral pseudotyping replaces envelope proteins with those from different viruses, tailoring the vector's tropism. For instance, pseudotyped Sindbis virus lentiviral vectors selectively target T cells, enabling precise CAR-T engineering in vivo.87 Similarly, adenoviral surface protein swapping can improve targeting, though this poses additional manufacturing challenges.

Pharmacokinetics and safety vary among vectors. AAVs are favoured for their low immunogenicity and stable expression but have limited payload capacity, whereas adenoviruses allow larger payloads at the cost of transient expression and stronger immune responses.88 Retroviral vectors enable long-term expression via genome integration but carry oncogenic risks due to random insertional mutagenesis. Advanced manufacturing and dosing strategies, such as particle threshold optimization in clinical trials, continue to improve the scalability, purity, and therapeutic efficacy of viral vector applications.89,90

3.1.2. Integrative viruses. Integrative viruses incorporate their genetic material into the host cell genome, enabling stable, long-term transgene expression (Table 1). Retroviruses, particularly lentiviruses, are commonly used in ex vivo applications such as CAR-T cell therapy due to their ability to efficiently integrate and express large transgenes.91,92 However, their use in vivo is limited by risks like insertional mutagenesis and uncontrolled gene expression, prompting efforts to redesign lentiviral vectors for safer applications.92 Foamy viruses (FVs), another retrovirus family, offer safer profiles due to preferential integration into non-coding genomic regions, reducing genotoxic risks.93–95 Unlike lentiviruses, FVs exhibit stability in quiescent cells, resuming activity upon cell division, and induce less clonal dominance, making them attractive for hematopoietic stem cell therapies and cancer applications.96 Challenges include the stability of larger transgenes during viral propagation, influencing therapeutic design.95

The Moloney murine leukaemia virus (MMLV), a type C retrovirus, has shown potential in clinical applications. Rexin-G, approved for all solid tumours in the Philippines, integrates a dominant-negative mutant of cyclin G1 (dnG1), inducing apoptosis and disrupting tumour vasculature.97,98 Similarly, Toca 511, encoding a yeast cytosine deaminase, converts the prodrug 5-fluorocytosine into the active chemotherapy agent 5-fluorouracil (5-FU). Although promising in preclinical models, Toca 511 faced challenges in clinical trials for glioblastoma due to inefficient transduction of tumour cells and limited stability in vivo.99,100 Integrative viruses hold significant therapeutic potential but require precise engineering to balance stable expression with safety concerns.

3.1.3. Non-integrative viruses. Non-integrative viral vectors are pivotal in gene therapy for delivering genetic material without incorporating it into the host genome, thus minimizing the risk of insertional mutagenesis. These vectors persist as episomes in the nucleus, offering a safer profile for therapeutic interventions (Table 1).
3.1.3.1. Adenoviruses. Adenoviruses (AdVs) are among the most widely utilized viral vectors in gene therapy, particularly in oncology.1 Engineered through successive generations to enhance safety and efficacy, they offer diverse therapeutic strategies. Wild-type adenoviruses (WTAd) are noted for high cloning capacity and robust immune responses.101 First-generation AdVs (FGAd) have deletions in essential genes (E1 and E3) to prevent replication, facilitating efficient gene delivery.102 Second-generation vectors (SGAd) involve further deletions (E2 and E4), reducing viral protein synthesis and immune reactions. Helper-dependent AdVs (HDAd), or “gutless” vectors, have extensive deletions, increasing safety and DNA carrying capacity while enabling long-term gene expression with minimal toxicity.103

In cancer therapy, adenoviral vectors are versatile and are therefore currently tested for numerous malignancies (Table S1). Gendicine, approved in China, delivers wild-type TP53 to induce cell cycle arrest and modulate the tumour TME.104 Oncorine, another approved therapy, is an oncolytic AdV targeting TP53-deficient cells, selectively replicating in tumour cells while sparing healthy ones.104 AdVs can also be engineered to secrete immunomodulatory factors, utilizing receptors like the coxsackievirus and adenovirus receptor on epithelial cells to enhance immune responses against tumors.105

AdVs have been instrumental in vaccine development, notably in the context of the SARS-CoV-2 pandemic, showcasing their capacity to stimulate immune responses.106 In oncology, nadofaragene firadenovec (rAd-IFN α2b/Syn3) was approved for non-muscle invasive bladder cancer unresponsive to Bacillus Calmette–Guérin therapy.107 This therapy employs a recombinant Ad5 vector to deliver the IFN-α2b gene, enhancing immune responses and inducing apoptosis in cancer cells via TNF-related apoptosis-inducing ligand (TRAIL)-related pathways.108 Another adenoviral therapy, aglatimagene besadenovec (CAN-2409), uses a replication-defective AdV encoding herpes simplex virus thymidine kinase (HSV-TK). The clinical developments of aglatimagene besadenovec are presented in Table S2, this treatment induces local activation of cytotoxic lymphocytes and proinflammatory cytokines, stimulating a robust antitumor immune response.109 DNX-2440 is an oncolytic AdV engineered to express OX40 ligand (OX40L), selectively replicating in tumour cells deficient in the retinoblastoma gene (Rb) or p16, enhancing T-cell proliferation and cytokine production.110,111

A challenge with AdVs is pre-existing immunity in humans, reducing efficacy and posing risks.112 To address this, non-human AdVs, like simian adenoviruses (e.g., ChAdOx1), are explored for their lower prevalence of pre-existing immunity.113 GRANITE and SLATE clinical studies utilize chimpanzee AdV vectors in a prime/boost immunotherapy to deliver personalized or shared neoantigen vaccines, enhancing CD8 T-cell responses in cancer patients.114


3.1.3.2. Adeno-associated viruses. Recombinant adeno-associated viruses (rAAVs) are small, non-enveloped single-stranded DNA viruses from the parvovirus family, with a packaging capacity of ∼4.7 kb. While this limits their use for complex transgenes in oncology, rAAVs have been approved for other diseases, such as Luxturna for retinal disorders and Zolgensma for spinal muscular atrophy. In cancer, rAAV-mediated interferon β (IFNβ) expression has shown promise in suppressing glioblastoma in mouse models.115 Capsid engineering enhances tumour specificity and transduction efficiency, exemplified by Her2-modified rAAVs and prostate-targeting rAAV7 and rAAV9.116,117

Clinical trials using AAV in oncology are limited. A Phase I trial (NCT02496273) involves ex vivo gene therapy for gastric cancer, where patients’ T lymphocytes are primed to target carcinoembryonic antigen (CEA)-expressing tumour cells. Another trial (NCT02602249) uses AAV2 to transduce dendritic cells with the MUC1 gene for gastric cancer treatment. AAVs can also deliver RNA interference molecules, such as shRNA, within target cells.118 However, toxicity from viral vectors and RNAi necessitates careful evaluation, including dose optimization and conditional promoters to minimize immune responses.

A novel approach involves in vivo AAV-mediated CAR-T cell therapy. An AAV vector encoding CD4CAR was used to transduce T cells within the patient's body, effectively reducing tumour size in a T-cell leukaemia model.119 This simplifies CAR-T production, potentially lowering costs and increasing accessibility.


3.1.3.3. Herpes simplex virus. Herpes simplex viruses (HSV-1 and HSV-2) are utilized in oncology as non-replicating vectors and oncolytic agents. The non-replicating HSV-1 vector NP2 was designed to express preproenkephalin (PENK) for pain modulation in cancer patients, showing safety and potential efficacy.120,121 Oncolytic HSV-1 vectors have also gained attention; talimogene laherparepvec (T-VEC, Imlygic), an HSV-1 vector encoding GM-CSF, was approved for recurrent melanoma.122 Despite innovative design, challenges like host antiviral responses and limited intratumoral spread affected efficacy. Ongoing trials investigate combinations of T-VEC with immune checkpoint inhibitors to enhance outcomes.123
3.1.3.4. Vaccinia virus. Vaccinia virus (VV), an enveloped double-stranded DNA orthopoxvirus, offers advantages like large genome capacity (∼190 kb), cytoplasmic replication (avoiding genome integration), and tumour tropism.124,125 Strains like Western Reserve (WR) and Modified Vaccinia Ankara (MVA) serve different purposes. TG6002, derived from the WR strain, selectively replicates in tumour cells and converts 5-fluorocytosine into 5-fluorouracil, showing efficacy in preclinical studies and ongoing trials for glioblastoma and gastrointestinal tumors.126 Pexa-Vec (JX-594), another WR-derived oncolytic virus expressing GM-CSF, has been explored in various cancers but did not improve outcomes in a Phase III trial for hepatocellular carcinoma.127 Research aims to enhance targeting and immune stimulation. MVA is extensively attenuated and used in cancer immunization strategies. A ‘prime-boost’ approach combining ChAdOx1-MAGE-A3-NY-ESO-1 and MVA vectors is under investigation to enhance immune responses in patients with specific tumour antigens.128,129
3.1.3.5. Chimeric orthopox virus. CF33 is a novel chimeric orthopoxvirus created by recombining multiple orthopoxvirus species, showing specificity for breast and pancreatic cancers.130 CF33-hNIS includes the human sodium iodide symporter for imaging,78 and CF33-hNIS-antiPDL1 expresses anti-PD-L1 antibodies, enhancing tumour localization and immune activation.131 Clinical trials, like the OASIS trial, evaluate CF33 variants with anti-CD19 agents for their oncolytic and immunostimulatory properties (Table S3).132

Despite advances with non-integrative viral vectors, concerns over safety, immunogenicity, and production complexity have prompted a shift toward non-viral vectors. Non-viral systems—such as lipid nanoparticles and polymers—offer advantages in safety, ease of production, and lower immunogenicity, making them suitable for repeat administrations and broader therapeutic applications.

3.2. Organic nanoparticles for nucleic acids transfection

3.2.1. Mechanisms and strategies for NP vectorisation. Organic nanoparticles (NPs) have transformed oncology by enabling stabilization and thus precise delivery of NAs for therapeutic purposes. Their unique properties, including small size, high surface area, and functionalisation potential, allow for encapsulating and protecting NAs such as DNA, RNA, siRNA, and mRNA.3,133 The size and shape, the charge, hydrophobicity, and encapsulation rate of the NP are key (Table S4). These attributes improve pharmacokinetics, enhance biodistribution, and minimise off-target effects, addressing the inherent challenges of NA therapies, such as enzymatic degradation, low cellular uptake, and rapid systemic clearance.134 Effective delivery systems must meet specific criteria: shielding NAs from degradation, maintaining stability in circulation, and ensuring efficient release within targeted cells. This is particularly crucial in solid tumours, especially the ones where high interstitial fluid pressure and a dense extracellular matrix pose significant barriers.135 Two primary mechanisms, the Enhanced Permeability and Retention (EPR) effect and Active Transport Retention (ATR), govern NP distribution. The EPR effect exploits the disorganised vasculature of tumours, which features large pores and poor lymphatic drainage (Fig. 3), enabling NPs of 100–200 nm to accumulate preferentially within tumour tissues.134,136 However, variability in the EPR effect due to patient-specific tumour biology limits its universal applicability. To enhance this mechanism, strategies such as carbon monoxide-releasing agents (e.g., SMA/CORM2 micelle) improve tumour blood flow and vascular permeability.137 In contrast, ATR involves active mechanisms like receptor-mediated interactions and the absence of intratumoral lymphatics to enhance NP retention. While ATR complements EPR, its success depends on understanding tumour-specific biology, requiring tailored approaches.138 Functionalising NP surfaces with ligands, such as antibodies, aptamers, or peptides, further enhances specificity.139,140 Tumour-specific antigens (TSAs), derived from genetic mutations or viral infections, and tumour-associated antigens (TAAs), overexpressed on cancer cells, serve as critical targets for ligand conjugation.141,142 A representative selection of diverse targeting agents is presented in Tables 2 and S5. These strategies reduce off-target effects and maximise therapeutic efficacy. Additionally, modifying the tumour microenvironment (TME) to deplete the extracellular matrix, inhibit cancer-associated fibroblasts, and reverse immunosuppression facilitates NP penetration and drug delivery.143,144
Table 2 Size and molecular characteristics of antibody-derived fragments and peptides for targeted vectors
Protein IgG(1) 150 kDa (10–15 nm) Fab’ 55 kDa (4–6 nm) Fab 50 kDa (4–6 nm) scFv 28 kDa (2–4 nm) VHH 12 kDa (camelids ab) (2–5 nm) Peptide 1–4 kDa (0.5 to 2 nm)
Schematic view image file: d4nr05371k-u1.tif image file: d4nr05371k-u2.tif image file: d4nr05371k-u3.tif image file: d4nr05371k-u4.tif image file: d4nr05371k-u5.tif image file: d4nr05371k-u6.tif


3.2.2. Advanced NP systems for NA delivery. Advancements in NP design have led to the development of systems that address stability, specificity, and controlled release (Fig. 5). Stealth NPs, coated with hydrophilic polymers like polyethylene glycol (PEG), evade immune detection by preventing opsonisation, thereby prolonging circulation time and improving tumour delivery.135 However, repeated PEGylated NP administration can elicit anti-PEG antibodies, reducing efficacy. Innovative alternatives, such as using different polymers or modifying dosing regimens, aim to mitigate this challenge.145
image file: d4nr05371k-f5.tif
Fig. 5 Evolution of lipid NPs’ design.

Stimuli-responsive NPs release their payload in response to specific triggers, such as pH, temperature, or redox potential. For example, pH-sensitive liposomes destabilise in the acidic TME, ensuring localised and precise drug activation.143 These systems enhance therapeutic outcomes while minimising systemic toxicity.

A critical aspect of these applications is overcoming intracellular barriers, particularly endosomal escape. Incorporating polyethylenimine (PEI) into NP formulations induces the proton sponge effect, causing endosomal swelling and rupture, which facilitates NA release into the cytoplasm.146 This mechanism is essential for achieving high transfection efficiency, particularly in hard-to-target cancers.

Recent advances in NP engineering have focused on improving specificity and therapeutic efficacy. NPs targeting the TME can reshape it by modulating immune responses or altering its physical and biochemical properties. For instance, targeting acidic, hypoxic, or fibrotic regions within the TME enhances NP delivery and therapeutic outcomes.144,147 Aptamers, specific oligonucleotides, can precisely target cell-specific receptors. Fluorescent aptamers now allow real-time monitoring of nucleic acid nanoparticle (NANP) formation and interactions. Advances in customizable NANPs have enabled the design of nanoassemblies that use aptamers to modulate cell signalling or activate therapeutic functions through aptamer–receptor interactions. These innovations pave the way for applications like reshaping the TME with nanoparticle decorations.148 Such strategies are crucial in overcoming the TME's role as a barrier to treatment and leveraging its unique characteristics for therapy.

3.2.3. Pharmacokinetics, biodistribution, and dose optimisation. Understanding the pharmacokinetics (PK) and biodistribution of NPs is pivotal for optimising their therapeutic potential. Upon administration, NPs interact with plasma proteins to form a “protein corona”, which significantly influences their stability, cellular uptake, and clearance. While the corona can enhance targeting by exposing functional ligands, it also marks NPs for immune clearance by macrophages, particularly Kupffer cells in the liver.89,146 Strategies to address this include engineering NPs with low immunogenicity.

The route of administration, structural properties, and interactions with biological matrices also impact NP performance. Intravenous delivery is most common for systemic cancer therapies, but variability in enzymatic degradation across organs necessitates tailored designs of NPs. For example, PEGylation extends circulation time but can trigger immune responses over time, underscoring the need for balancing stability and adverse immune reactions.145,149,150

Recent studies have identified a minimum effective dose threshold for tumour delivery. Administering 1 trillion NPs in mice saturates Kupffer cells, prolonging NP circulation and enhancing tumour accumulation. This approach has improved delivery efficiency in formulations like Caelyx/Doxil and provides a framework for dose optimisation in clinical settings.89

Tumour heterogeneity remains a significant challenge. Differences in genetic profiles, TME composition, and therapeutic responses necessitate personalised NP strategies. A complementary approach to reducing tumour progression and dissemination is to deliver NAs with NPs to cancer stem cells, or to target the epithelial–mesenchymal transition, or to block metastatic pathways.151–153 Similarly, NPs targeting circulating tumour cells (CTCs) offer a novel approach to prevent metastasis. For example, TRAIL-conjugated liposomes induce apoptosis in CTCs while sparing healthy tissues.154

Advancing NP-based therapies requires integrating pharmacological, physiological, and pathological considerations into design and application. This includes understanding tumour-specific biodistribution patterns, overcoming immune clearance, and optimising dosing regimens to achieve maximal therapeutic benefit. Standardising manufacturing and purification protocols is also critical for ensuring batch-to-batch consistency, which is essential for regulatory approval and clinical application.155,156 The DELIVER framework encapsulates the core principles for optimising nanomedicine preclinical development, emphasising design, manufacturing, clinical, and regulatory strategies to accelerate translation and maximise impact.157

3.2.4. Lipidic vectors. Lipidic vectors have been transformative in drug delivery, especially in oncology, where they have been applied for over two decades.

These vectors, including liposomes, lipoplexes, and lipid nanoparticles (LNPs) (Fig. 6), offer numerous advantages, such as extended circulation times, avoidance of the reticuloendothelial system (RES), and high biocompatibility. They are known for their ability to encapsulate and protect therapeutic agents, NAs, while facilitating targeted delivery. Among their applications, Doxil, a liposomal formulation of doxorubicin approved in 1995, exemplifies the efficacy of lipidic vectors in utilising the EPR effect for tumour targeting. These systems improve bioavailability, reduce systemic toxicity, and enhance therapeutic outcomes.158,159


image file: d4nr05371k-f6.tif
Fig. 6 Representation of selected lipidic vectors.

The roles and characteristics of lipids used in lipidic vectors formulations, such as ionizable, phospholipid, cholesterol, and PEGylated components, are critical for determining stability, delivery efficiency and compatibility with various NAs (Table S6). While liposomes have been traditionally used for delivering plasmid DNA, the emergence of lipoplexes and LNPs has enabled the delivery of various NAs, including mRNA, siRNA, and ASOs, through electrostatic interactions. Tables S7, S8, and S9 provide an overview of ongoing clinical trials exploring lipid-based vectors, with a focus on liposomes (Table S7), lipoplexes (Table S8), and LNPs-based systems (Table S9), showcasing their distinct roles in advancing nucleic acid delivery technologies. Importantly, only three NA therapies using LNPs—patisiran (siRNA), the Pfizer-BioNTech COVID-19 vaccine, and the Moderna COVID-19 vaccine—have received market approval, highlighting the clinical relevance of lipid-based delivery systems.160–162


3.2.4.1. Liposomes. Liposomes are spherical vesicles consisting of one or more phospholipid bilayers encapsulating an aqueous core. Due to their versatile structure, they have been widely used in drug delivery, including for NAs such as miRNA, siRNA, and ASOs. Their neutral lipid composition, often incorporating phospholipids (Table S6), enhances their biocompatibility but limits their ability to encapsulate highly charged molecules like DNA and RNA. To overcome this, modified liposomal systems have been developed, targeting smaller or chemically modified NAs with reduced charges.52

One significant advancement in liposomal technology is the development of EphA2-targeted siRNA delivery systems. EphA2, a receptor tyrosine kinase overexpressed in multiple cancers, plays a crucial role in tumour progression and metastasis. A liposome composed of DOPC encapsulating EphA2-specific siRNA, termed EPHARNA, demonstrated significant efficacy in preclinical models. For example, in ovarian cancer models, EPHARNA reduced tumour growth and metastasis while exhibiting minimal toxicity. Additionally, its combination with chemotherapy showed synergistic effects, and ongoing clinical trials (NCT01591356) are evaluating its safety and efficacy in human subjects.163,164

Another innovative liposomal formulation is BP1001, an antisense oligonucleotide (ASO) targeting Grb2 mRNA. Composed of a P-ethoxy backbone for nuclease resistance, BP1001 is encapsulated in DOPC liposomes, ensuring stability and extended circulation times. This formulation has demonstrated promising results in preclinical models of leukaemia and solid tumours, with clinical trials indicating safety and efficacy in refractory leukaemia (NCT02781883).165,166


3.2.4.2. Lipoplexes. Lipoplexes are complexes formed through electrostatic interactions between positively charged lipids and negatively charged NAs. Their simplicity, ease of customisation, and ability to deliver large NAs make them an attractive option for therapeutic applications. However, their reliance on electrostatic interactions necessitates optimisation to ensure stability and minimise aggregation in vivo.136,167

DOTMA, a cationic lipid widely used in lipoplex formulations, has demonstrated efficacy in gene transfer, particularly when combined with helper lipids like DOPE. The inclusion of DOPE facilitates membrane fusion and endosomal escape, critical steps for effective gene transfection. Additionally, lipoplexes have been modified with tumour-targeting peptides, such as iRGD (a 9-amino acid cyclic peptide, Table 2), to enhance their active targeting capabilities. For example, R-LP, a DOTAP-based lipoplex modified with iRGD, successfully delivered eIF3i-specific shRNA in melanoma models, significantly inhibiting tumour growth and metastasis.168 A notable clinical example is SGT53, a lipoplex encapsulating wild-type human p53 cDNA. Modified with an anti-transferrin receptor (TfR) scFv for selective targeting, SGT53 has shown efficacy in restoring apoptotic pathways in various cancers. It is currently being evaluated in clinical trials for glioblastoma (NCT02340156) and pancreatic cancer (NCT02340117), where it demonstrated improved progression-free survival in second-line treatments.169,170

Despite their potential, lipoplexes face challenges such as detachment of NAs in the bloodstream, leading to rapid aggregation. Innovations in lipid composition and manufacturing processes are addressing these limitations, enhancing the stability and efficacy of lipoplexes for clinical applications.171


3.2.4.3. Lipid Nanoparticles. LNPs are among the most advanced lipidic vectors, consisting of a lipid core surrounded by a phospholipid monolayer. Their composition typically includes ionizable or cationic lipids, cholesterol, phospholipids, and PEGylated lipids, allowing for precise tuning of their properties for specific therapeutic applications. Unlike lipoplexes, LNPs leverage pH-sensitive ionizable lipids to stabilise NAs and potentially enhance endosomal escape, making them highly efficient for intracellular delivery.161,167

A landmark in LNP technology is patisiran/Onpattro, an siRNA-based therapy encapsulated in LNPs, approved for hereditary transthyretin-mediated amyloidosis. This system exemplifies the clinical success of LNPs in overcoming biological barriers to NA delivery. Similarly, the Pfizer-BioNTech and Moderna COVID-19 vaccines utilised LNPs to deliver mRNA encoding the SARS-CoV-2 spike protein, demonstrating the scalability and efficacy of this platform.162 Cholesterol plays a pivotal role in stabilising LNP structures, reducing permeability, and enhancing circulation time. However, cholesterol can be rapidly extracted by cell membranes, compromising stability. Modified lipids, such as sphingomyelin derivatives, have been developed to address this issue. For instance, SM-CSS-Chol, a disulfide-bonded cholesterol analogue, improved the stability and delivery efficiency of siRNA-loaded LNPs in preclinical cancer models, highlighting its potential for clinical applications.172

Targeted LNPs have also been developed for immune cell transfection. For example, a CD3-targeted LNP encapsulating plasmid DNA for CAR-T therapy demonstrated selective delivery to T cells, enhancing gene expression while minimising off-target effects. This system incorporates PEGylation to improve circulation time and specific lipid combinations to optimise endosomal escape, showcasing the versatility of LNPs in advanced gene therapies.173

The toxicity and immunogenicity of LNPs are critical considerations, particularly for repeated dosing. Ionizable lipids, the cornerstone of LNP formulations, exhibit pH-sensitive properties that reduce cytotoxicity compared to permanently charged cationic lipids. Biodegradable linkages, such as ester and amide bonds, further minimise toxicity by facilitating in vivo cleavage. Nevertheless, careful evaluation of lipid composition and dosing regimens is necessary to balance efficacy and safety.174–176

3.2.5. Polymeric vectors. Polymeric vectors have emerged as a versatile platform in NAs delivery, offering a tunable framework to overcome challenges such as enzymatic degradation, limited stability, and poor cellular uptake of naked NAs. These vectors harness the diversity of polymers (Table S10), which can be tailored chemically and structurally to meet therapeutic demands, such as protecting NAs during systemic circulation, promoting cellular internalisation, and enabling controlled release at target sites.177 Among the various systems under development, polymersomes, polyplexes, polymeric nanocapsules, and dendrimers stand out for their unique attributes and therapeutic potential (Fig. 7).
image file: d4nr05371k-f7.tif
Fig. 7 Representation of selected polymeric vectors.

Polymers suitable for NA delivery include both natural and synthetic types, such as poly(ethyleneimine) (PEI), poly-L-lysine (PLL), poly(lactic-co-glycolic acid) (PLGA), poly(beta-amino ester) (PBAE), and poly(2-dimethylaminoethyl methacrylate) (PDMAEMA) (Table S10). Each offers specific advantages, including charge density for NA condensation, biodegradability, and biocompatibility. Modifications, such as PEGylation, are often incorporated to enhance systemic stability and minimise immune responses, ensuring the safe and efficient delivery of therapeutic agents.178–180

The design of polymeric vectors often involves balancing transfection efficiency and toxicity. Cationic polymers like PEI and PLL effectively bind and condense NAs through electrostatic interactions, forming compact polyplexes that protect NAs from enzymatic degradation. However, their high charge density and lack of biodegradability can lead to cytotoxicity and off-target effects, necessitating structural modifications to enhance safety profiles.181 PLGA, an FDA-approved polymer, offers an alternative with its excellent biocompatibility and biodegradability, making it a preferred choice for clinical applications, particularly for sustained-release systems.182


3.2.5.1. Polymersomes. Polymersomes are vesicular systems formed by the self-assembly of amphiphilic block copolymers into bilayer structures. These vesicles mimic liposomes but possess superior stability due to their thicker membranes, which resist mechanical stress and provide prolonged circulation times in vivo. Polymersomes can encapsulate both hydrophilic and hydrophobic NAs, offering a flexible platform for gene delivery.183 The chemical diversity of block copolymers enables the design of polymersomes with tailored properties for specific applications. For instance, polymersomes made from poly(ethylene glycol)–poly(caprolactone) (PEG–PCL) or poly(ethylene glycol)–poly(lactic acid) (PEG–PLA) demonstrate excellent biocompatibility and efficient encapsulation of plasmids and siRNA. Functionalisation with targeting ligands further enhances cellular uptake, enabling site-specific delivery in tumours.183,184

A notable clinical example involves the Local Drug EluteR device, which uses a PLGA-based polymer matrix to deliver siRNA targeting the KRAS G12D mutation in pancreatic cancer. This system has shown promising results in phase I and II trials (NCT01188785, NCT01676259), providing a minimally invasive approach to address one of the most challenging oncogenic mutations.


3.2.5.2. Polyplexes. Polyplexes are complexes formed by the electrostatic interaction between cationic polymers and negatively charged NAs. These compact, stable structures shield NAs from enzymatic degradation, enhance cellular uptake, and promote intracellular delivery. Unlike lipoplexes, polyplexes are composed of hydrophilic polymers, which improve their solubility and adaptability in aqueous environments.185,186

PEI, a widely studied polymer, forms polyplexes with high transfection efficiency due to its ability to condense NAs and facilitate endosomal escape via the proton sponge effect. However, its high molecular weight variants often exhibit cytotoxicity, prompting the development of biodegradable alternatives, such as poly(beta-amino ester) (PBAE) and PEGylated PEI, which combine high transfection efficiency with reduced toxicity.187

Cyclodextrin-based polyplexes, exemplified by CALAA-01, represent a pioneering approach in systemic siRNA delivery. Functionalised with transferrin for tumour targeting, CALAA-01 demonstrated dose-dependent gene silencing in phase I trials but faced challenges related to DLTs, attributed to the polymer components rather than the siRNA.188


3.2.5.3. Polymeric nanocapsules. Polymeric nanocapsules feature a core–shell architecture, where therapeutic agents are encapsulated within a polymeric shell, offering protection and controlled release. These structures are versatile platforms for NA delivery, capable of incorporating diverse polymer types, including PLGA, PEG-PBAE, and chitosan. Their rigid structure enhances stability, while their surface can be functionalised to improve targeting and cellular uptake.182,187

A key example of polymeric nanocapsule application is STP705, a system that utilises histidine-lysine copolymers to deliver siRNA targeting TGF-β1 and COX-2. This dual-targeted approach addresses both tumour cells and the tumour microenvironment, demonstrating high efficacy in preclinical and early clinical studies for cancers like hepatocellular carcinoma.189

Another innovative system employs PEG-PBAE nanocapsules co-encapsulating plasmids for glioblastoma treatment. These vectors achieve superior transfection efficiency and tumour penetration compared to non-PEGylated systems, highlighting the role of polymeric nanocapsules in addressing challenges such as endosomal escape and tumour heterogeneity.187


3.2.5.4. Dendrimers. Dendrimers are hyperbranched, tree-like macromolecules characterised by a central core and radially symmetric layers of branching units. Their unique architecture allows for precise control over size, shape, and surface functionality, making them highly adaptable for NA delivery. Dendrimers form stable complexes with NAs through electrostatic interactions, protecting the cargo and facilitating intracellular transport.190

Poly(amidoamine) (PAMAM) dendrimers are the most extensively studied for gene therapy. Their surface amine groups enable efficient condensation of NAs, while their branched structure allows for multifunctionalisation. Recent advancements include the modification of PAMAM G5 dendrimers with cholesteryl chloroformate and alkyl-PEG for dual delivery of TRAIL and doxorubicin, achieving synergistic anti-tumour effects in preclinical colon cancer models.191

Phosphorus dendrimers, incorporating protonated ammonium terminals, have shown efficacy in delivering plasmid DNA encoding tumour suppressor genes like TP53. These systems effectively induce cell cycle arrest and apoptosis in tumour cells, demonstrating their potential in targeted therapies.192 Carbosilane dendrimers, on the other hand, have been employed to deliver pro-apoptotic siRNA, highlighting their versatility in addressing oncogenic pathways.193

A novel application of dendrimers involves the formation of RNA triplex nanoparticles, combining dendritic architecture with RNA molecules for dual-function cancer therapy. These systems enable the simultaneous inhibition of oncogenic microRNAs and replacement of tumour suppressor microRNAs, offering a comprehensive approach to tumour management.194

3.2.6. Lipid-polymer hybrid nanoparticles. LPHNs, also named lipopolyplexes, combine lipid structures with cationic or neutral polymers, offering controlled release, stability, and protection for NAs.195 A PLGA polymer shell enhances sustained delivery of siRNA, enabling long-term gene silencing, reducing administration frequency, and minimising systemic side effects.195 Cationic polymers like PEI condense NAs into stable complexes, encapsulated by lipid bilayers of DPPC and cholesterol, with PEG and ligands like PR_b for targeted delivery. For glioblastomas, PEI-complexed miR-603 in PR_b-functionalised liposomes sensitises tumour cells to radiation by downregulating IGF1 signalling.151,196 LPNPs encapsulating CRISPR/Cas9 plasmids targeting MGMT, combined with focused ultrasound and microbubbles, effectively crossed the blood–brain barrier, enhancing glioblastoma sensitivity to temozolomide. Modifications like DSPE-PEG2000 improve tumour targeting and circulation time, enabling advanced therapies for drug-resistant glioblastomas.197
3.2.7. Extracellular vesicles (EVs). Extracellular vesicles (EVs), cellular membrane-based nanovesicles ranging from 50 to over 2000 nm, mediate intercellular communication via autocrine, paracrine, and endocrine signalling. Subtypes like exosomes, microvesicles, and apoptotic bodies carry diverse cargo, including proteins, NAs, and metabolites, reflecting their cellular origin. EVs’ ability to evade immunity and cross barriers like the blood–brain barrier makes them promising vectors for drug and gene therapy delivery.198 Mesenchymal stem/stromal cell (MSC)-derived EVs offer therapeutic potential for targeting tumours, facilitated by engineering their surfaces with specific ligands for precise delivery of NAs and CRISPR/Cas9 tools.199 Phase 1 trials, such as KRASG12D-siRNA-loaded exosomes for pancreatic cancer, highlight their clinical potential.200 Additionally, EVs encapsulating adeno-associated viruses (AAVs) bypass pre-existing immunity, improving vector safety and stability.201 Novel vexosomes, combining exosomes and viral vectors, enhance gene therapy applications.202 In hepatocellular carcinoma models, AAV6 vexosomes showed a 2.3-fold increase in tumour regression.203

3.3. Cells as vectors

The use of neural stem cells (NSCs) as carriers for oncolytic viruses is a promising strategy for treating malignant gliomas, leveraging NSCs’ natural tumour-tropism as biological “Trojan horses”. Genetic engineering enhances their specificity, as demonstrated by the FDA-approved NSC line HB1.F3.CD21, which delivers CRAd-S-pk7, a conditionally replicative adenovirus. CRAd-S-pk7 uses a survivin promoter for selective replication in glioma cells and features a polylysine sequence (pK7) to improve viral attachment, enhancing targeting of resistant glioma stem cells. Preclinical and phase I trials (NCT03072134) show improved antitumor activity and survival in glioblastoma models.204 Similarly, bone marrow-derived mesenchymal stem cells (BM-hMSCs) loaded with DNX-2401, another oncolytic adenovirus, utilize the same principle to precisely target gliomas. This approach demonstrates efficacy in recurrent high-grade gliomas in phase I trials (NCT03896568). These innovations highlight the potential of stem cell-based oncolytic virotherapy to overcome vectorization challenges, offering precise and effective treatment for resistant gliomas.

4. Conclusions

In summary, gene and RNA-based therapies represent an evolving frontier in oncological treatment, offering highly specific mechanisms to modulate gene expression, restore tumour suppressors, or enhance anti-tumour immunity. Advances in molecular tools—from CRISPR-based gene editing to antisense oligonucleotides and self-amplifying mRNAs—have provided unprecedented precision and flexibility. At the same time, the development of robust delivery systems remains central to the successful translation of these approaches. Viral vectors continue to be refined for safety, specificity, and stable integration, while non-viral carriers, such as lipidic or polymeric nanoparticles and extracellular vesicles, show great promise in overcoming barriers like enzymatic degradation, immunogenicity, and complex intratumoral environments. The integration of cell-based carriers, exemplified by stem cell “Trojan horses” further expands the therapeutic landscape.

Looking ahead, the field is poised to capitalize on emerging technologies for target identification, advanced gene editing methods, improved vector engineering, and more nuanced control over gene expression patterns. Combining these therapies with established treatments—chemotherapy, immunotherapy, radiation—and implementing personalized strategies based on tumour profiling will likely enhance both efficacy and safety. Continued interdisciplinary collaboration, rigorous clinical testing, and regulatory refinement will be pivotal in pushing these next-generation therapies into mainstream cancer care.

Author contributions

JSK screened the literature, wrote the original draft and made illustrations. BB developed the concept, structured the paper and supervised this work. All authors wrote and reviewed the manuscript.

Data availability

This article is a review and does not include original research data. All information discussed is derived from published sources, which are cited in the reference list.

All ESI data supporting this review are included in the manuscript and its ESI.

Where applicable, data supporting the findings have been obtained from publicly available sources, as detailed in the references.

Conflicts of interest

There are no conflicts to declare.

References

  1. S. L. Ginn, M. Mandwie, I. E. Alexander, M. Edelstein and M. R. Abedi, Gene therapy clinical trials worldwide to 2023—an update, J. Gen. Med., 2024, 26, e3721 CrossRef CAS PubMed.
  2. Y. Zhu, L. Zhu, X. Wang and H. Jin, RNA-based therapeutics: an overview and prospectus, Cell Death Dis., 2022, 13, 1–15 Search PubMed.
  3. P. R. Cullis and P. L. Felgner, The 60-year evolution of lipid nanoparticles for nucleic acid delivery, Nat. Rev. Drug Discovery, 2024, 23, 709–722 CrossRef CAS PubMed.
  4. B. B. Mendes, J. Conniot, A. Avital, D. Yao, X. Jiang, X. Zhou, N. Sharf-Pauker, Y. Xiao, O. Adir, H. Liang, J. Shi, A. Schroeder and J. Conde, Nanodelivery of nucleic acids, Nat. Rev. Methods Primers, 2022, 2, 1–21 CrossRef.
  5. D. Drago, B. Foss-Campbell, K. Wonnacott, D. Barrett and A. Ndu, Global regulatory progress in delivering on the promise of gene therapies for unmet medical needs, Mol. Ther. – Methods Clin. Dev., 2021, 21, 524–529 CrossRef CAS PubMed.
  6. R. C. Sterner and R. M. Sterner, CAR-T cell therapy: current limitations and potential strategies, J. Blood Cancer, 2021, 11, 1–11 CrossRef PubMed.
  7. S. Abbasi, M. A. Totmaj, M. Abbasi, S. Hajazimian, P. Goleij, J. Behroozi, B. Shademan, A. Isazadeh and B. Baradaran, Chimeric antigen receptor T (CAR-T) cells: Novel cell therapy for hematological malignancies, Cancer Med., 2023, 12, 7844–7858 CrossRef CAS PubMed.
  8. K. M. Cappell and J. N. Kochenderfer, Long-term outcomes following CAR T cell therapy: what we know so far, Nat. Rev. Clin Oncol., 2023, 20, 359–371 CrossRef CAS PubMed.
  9. N. Albinger, S. Müller, J. Kostyra, J. Kuska, S. Mertlitz, O. Penack, C. Zhang, N. Möker and E. Ullrich, Manufacturing of primary CAR-NK cells in an automated system for the treatment of acute myeloid leukemia, Bone Marrow Transplant., 2024, 59, 489–495 CrossRef CAS.
  10. C. L. Hardee, L. M. Arévalo-Soliz, B. D. Hornstein and L. Zechiedrich, Advances in Non-Viral DNA Vectors for Gene Therapy, Genes, 2017, 8, 65 CrossRef PubMed.
  11. F. Voigt, L. Wiedemann, C. Zuliani, I. Querques, A. Sebe, L. Mátés, Z. Izsvák, Z. Ivics and O. Barabas, Sleeping Beauty transposase structure allows rational design of hyperactive variants for genetic engineering, Nat. Commun., 2016, 7, 11126 CrossRef CAS PubMed.
  12. D. S. J. Allan, M. Chakraborty, G. C. Waller, M. J. Hochman, A. Poolcharoen, R. N. Reger and R. W. Childs, Systematic improvements in lentiviral transduction of primary human natural killer cells undergoing ex vivo expansion, Mol. Ther. – Methods Clin. Dev., 2021, 20, 559–571 CrossRef CAS PubMed.
  13. T. Bexte, L. Botezatu, C. Miskey, F. Gierschek, A. Moter, P. Wendel, L. M. Reindl, J. Campe, J. F. Villena-Ossa, V. Gebel, K. Stein, T. Cathomen, A. Cremer, W. S. Wels, M. Hudecek, Z. Ivics and E. Ullrich, Engineering of potent CAR NK cells using non-viral Sleeping Beauty transposition from minimalistic DNA vectors, Mol. Ther., 2024, 32, 2357–2372 CrossRef CAS PubMed.
  14. I. Querques, A. Mades, C. Zuliani, C. Miskey, M. Alb, E. Grueso, M. Machwirth, T. Rausch, H. Einsele, Z. Ivics, M. Hudecek and O. Barabas, A highly soluble Sleeping Beauty transposase improves control of gene insertion, Nat. Biotechnol., 2019, 37, 1502–1512 CrossRef CAS.
  15. National Cancer Institute (NCI), A Phase II Study Using the Administration of Autologous T-Cells Engineered Using the Sleeping Beauty Transposon/Transposase System to Express T-Cell Receptors Reactive Against Mutated Neoantigens in Patients With Metastatic Cancer, A Phase II Study Using the Administration of Autologous T-Cells Engineered Using the Sleeping Beauty Transposon/Transposase System to Express T-Cell Receptors Reactive Against Mutated Neoantigens in Patients With Metastatic Cancer, clinicaltrials.gov, 2024.
  16. K. Ma, W. Li, G. Zhu, S. Sun, H. Chi, Y. Yin, H. Diao, X.-J. Xing, Z. Guo, L. Wang, W. Xu, C. Cui and J. Xu, Functionalized PDA/DEX-PEI@HA nanoparticles combined with sleeping-beauty transposons for multistage targeted delivery of CRISPR/Cas9 gene, Biomed. Pharmacother., 2021, 142, 112061 CrossRef CAS PubMed.
  17. J. Tipanee, T. VandenDriessche and M. K. Chuah, Transposons: Moving Forward from Preclinical Studies to Clinical Trials, Hum. Gene Ther., 2017, 28, 1087–1104 CrossRef CAS PubMed.
  18. M. Wei, C.-L. Mi, C.-Q. Jing and T.-Y. Wang, Progress of Transposon Vector System for Production of Recombinant Therapeutic Proteins in Mammalian Cells, Front. Bioeng. Biotechnol., 2022, 10, 879222 CrossRef PubMed.
  19. C. Chen, D. Yue, L. Lei, H. Wang, J. Lu, Y. Zhou, S. Liu, T. Ding, M. Guo and L. Xu, Promoter-Operating Targeted Expression of Gene Therapy in Cancer: Current Stage and Prospect, Mol. Ther. – Nucleic Acids, 2018, 11, 508–514 CrossRef CAS PubMed.
  20. M. Montaño-Samaniego, D. M. Bravo-Estupiñan, O. Méndez-Guerrero, E. Alarcón-Hernández and M. Ibáñez-Hernández, Strategies for Targeting Gene Therapy in Cancer Cells With Tumor-Specific Promoters, Front. Oncol., 2020, 10, 605380 CrossRef PubMed.
  21. H.-X. Zhang, Y. Zhang and H. Yin, Genome Editing with mRNA Encoding ZFN, TALEN, and Cas9, Mol. Ther., 2019, 27, 735–746 CrossRef CAS PubMed.
  22. A. V. Anzalone, L. W. Koblan and D. R. Liu, Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors, Nat. Biotechnol., 2020, 38, 824–844 CrossRef CAS PubMed.
  23. C. Wong, UK first to approve CRISPR treatment for diseases: what you need to know, Nature, 2023, 623, 676–677 CrossRef CAS PubMed.
  24. E. Haapaniemi, S. Botla, J. Persson, B. Schmierer and J. Taipale, CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response, Nat. Med., 2018, 24, 927–930 CrossRef CAS.
  25. L. Jiang, K. Ingelshed, Y. Shen, S. V. Boddul, V. S. Iyer, Z. Kasza, S. Sedimbi, D. P. Lane and F. Wermeling, CRISPR/Cas9-Induced DNA Damage Enriches for Mutations in a p53-Linked Interactome: Implications for CRISPR-Based Therapies, Cancer Res., 2022, 82, 36–45 CrossRef CAS.
  26. A. Saber Sichani, M. Ranjbar, M. Baneshi, F. Torabizadeh and J. Fallahi, A Review on Advanced CRISPR-Based Genome-Editing Tools: Base Editing and Prime Editing, Mol. Biotechnol., 2023, 65, 849–860 CrossRef CAS.
  27. G. Jang, J. Kweon and Y. Kim, CRISPR prime editing for unconstrained correction of oncogenic KRAS variants, Commun. Biol., 2023, 6, 1–7 CrossRef.
  28. A. V. Anzalone, P. B. Randolph, J. R. Davis, A. A. Sousa, L. W. Koblan, J. M. Levy, P. J. Chen, C. Wilson, G. A. Newby, A. Raguram and D. R. Liu, Search-and-replace genome editing without double-strand breaks or donor DNA, Nature, 2019, 576, 149–157 CrossRef CAS.
  29. D. E. Yoon, H. Lee and K. Kim, Recent Research Trends in Stem Cells Using CRISPR/Cas-Based Genome Editing Methods, Int. J. Stem Cells, 2024, 17, 1–14 CrossRef CAS.
  30. J. R. Androsavich, Frameworks for transformational breakthroughs in RNA-based medicines, Nat. Rev. Drug Discovery, 2024, 1–24 Search PubMed.
  31. S. Castillo-Hair, S. Fedak, B. Wang, J. Linder, K. Havens, M. Certo and G. Seelig, Optimizing 5′UTRs for mRNA-delivered gene editing using deep learning, Nat. Commun., 2024, 15, 5284 CrossRef CAS PubMed.
  32. M. M. Billingsley, N. Singh, P. Ravikumar, R. Zhang, C. H. June and M. J. Mitchell, Ionizable Lipid Nanoparticle-Mediated mRNA Delivery for Human CAR T Cell Engineering, Nano Lett., 2020, 20, 1578–1589 CrossRef CAS PubMed.
  33. M. M. Billingsley, N. Gong, A. J. Mukalel, A. S. Thatte, R. El-Mayta, S. K. Patel, A. E. Metzloff, K. L. Swingle, X. Han, L. Xue, A. G. Hamilton, H. C. Safford, M.-G. Alameh, T. E. Papp, H. Parhiz, D. Weissman and M. J. Mitchell, In Vivo mRNA CAR T Cell Engineering via Targeted Ionizable Lipid Nanoparticles with Extrahepatic Tropism, Small, 2024, 20(11), 2304378 CrossRef CAS PubMed.
  34. L. Short, R. A. Holt, P. R. Cullis and L. Evgin, Direct in vivo CAR T cell engineering, Trends Pharmacol. Sci., 2024, 45, 406–418 CrossRef CAS PubMed.
  35. CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis | New England Journal of Medicine, CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis | New England Journal of Medicine, https://www.nejm.org/doi/full/10.1056/NEJMoa2107454, (accessed 19 November 2024).
  36. D. Javaid, S. Y. Ganie, Y. A. Hajam and M. S. Reshi, CRISPR/Cas9 system: a reliable and facile genome editing tool in modern biology, Mol. Biol. Rep., 2022, 49, 12133–12150 CrossRef CAS PubMed.
  37. C. D. Palmer, A. R. Rappaport, M. J. Davis, M. G. Hart, C. D. Scallan, S.-J. Hong, L. Gitlin, L. D. Kraemer, S. Kounlavouth, A. Yang, L. Smith, D. Schenk, M. Skoberne, K. Taquechel, M. Marrali, J. R. Jaroslavsky, C. N. Nganje, E. Maloney, R. Zhou, D. Navarro-Gomez, A. C. Greene, G. Grotenbreg, R. Greer, W. Blair, M. D. Cao, S. Chan, K. Bae, A. I. Spira, S. Roychowdhury, D. P. Carbone, B. S. Henick, C. G. Drake, B. J. Solomon, D. H. Ahn, A. Mahipal, S. B. Maron, B. Johnson, R. Rousseau, R. Yelensky, C.-Y. Liao, D. V. T. Catenacci, A. Allen, A. R. Ferguson and K. Jooss, Individualized, heterologous chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced metastatic solid tumors: phase 1 trial interim results, Nat. Med., 2022, 28, 1619–1629 CrossRef CAS PubMed.
  38. G. P. Dailey, E. J. Crosby and Z. C. Hartman, Cancer vaccine strategies using self-replicating RNA viral platforms, Cancer Gene Ther., 2023, 30, 794–802 CrossRef CAS PubMed.
  39. D. Hattab, A. M. Gazzali and A. Bakhtiar, Clinical Advances of siRNA-Based Nanotherapeutics for Cancer Treatment, Pharmaceutics, 2021, 13, 1009 CrossRef CAS PubMed.
  40. S. Boca, D. Gulei, A.-A. Zimta, A. Onaciu, L. Magdo, A. B. Tigu, C. Ionescu, A. Irimie, R. Buiga and I. Berindan-Neagoe, Nanoscale delivery systems for microRNAs in cancer therapy, Cell. Mol. Life Sci., 2020, 77, 1059–1086 CrossRef CAS PubMed.
  41. D. S. Hong, Y.-K. Kang, M. Borad, J. Sachdev, S. Ejadi, H. Y. Lim, A. J. Brenner, K. Park, J.-L. Lee, T.-Y. Kim, S. Shin, C. R. Becerra, G. Falchook, J. Stoudemire, D. Martin, K. Kelnar, H. Peltier, V. Bonato, A. G. Bader, S. Smith, S. Kim, V. O'Neill and M. S. Beg, Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours, Br. J. Cancer, 2020, 122, 1630–1637 CrossRef CAS PubMed.
  42. M. T. J. van den Bosch, S. Yahyanejad, M. F. Alemdehy, B. J. Telford, T. de Gunst, H. C. den Boer, R. M. Vos, M. Stegink, L. A. H. van Pinxteren, R. Q. J. Schaapveld and M. Janicot, Transcriptome-wide analysis reveals insight into tumor suppressor functions of 1B3, a novel synthetic miR-193a-3p mimic, Mol. Ther. – Nucleic Acids, 2021, 23, 1161–1171 CrossRef CAS PubMed.
  43. D. Bautista-Sánchez, C. Arriaga-Canon, A. Pedroza-Torres, I. A. De La Rosa-Velázquez, R. González-Barrios, L. Contreras-Espinosa, R. Montiel-Manríquez, C. Castro-Hernández, V. Fragoso-Ontiveros, R. M. Álvarez-Gómez and L. A. Herrera, The Promising Role of miR-21 as a Cancer Biomarker and Its Importance in RNA-Based Therapeutics, Mol. Ther. – Nucleic Acids, 2020, 20, 409–420 CrossRef PubMed.
  44. T. Fuertes, A. R. Ramiro and V. G. de Yebenes, miRNA-Based Therapies in B Cell Non-Hodgkin Lymphoma, Trends Immunol., 2020, 41, 932–947 CrossRef CAS PubMed.
  45. T. C. Roberts, R. Langer and M. J. A. Wood, Advances in oligonucleotide drug delivery, Nat. Rev. Drug Discovery, 2020, 19, 673–694 CrossRef CAS PubMed.
  46. I. Ahn, C. S. Kang and J. Han, Where should siRNAs go: applicable organs for siRNA drugs, Exp. Mol. Med., 2023, 55, 1283–1292 CrossRef CAS PubMed.
  47. M. Winkle, S. M. El-Daly, M. Fabbri and G. A. Calin, Noncoding RNA therapeutics—challenges and potential solutions, Nat. Rev. Drug Discovery, 2021, 20, 629–651 CrossRef CAS PubMed.
  48. A. G. Hamilton and M. J. Mitchell, An oncolytic circular RNA therapy, Nat. Cancer, 2024, 5, 5–7 CrossRef CAS PubMed.
  49. Z. Feng, X. Zhang, J. Zhou, Q. Li, L. Chu, G. Di, Z. Xu, Q. Chen, M. Wang, X. Jiang, H. Xia and X. Chen, An in vitro-transcribed circular RNA targets the mitochondrial inner membrane cardiolipin to ablate EIF4G2+/PTBP1 + pan-adenocarcinoma, Nat. Cancer, 2024, 5, 30–46 CrossRef CAS PubMed.
  50. V. Sardone, H. Zhou, F. Muntoni, A. Ferlini and M. S. Falzarano, Antisense Oligonucleotide-Based Therapy for Neuromuscular Disease, Molecules, 2017, 22, 563 CrossRef PubMed.
  51. R. Gupta, S. Salave, D. Rana, B. Karunakaran, A. Butreddy, D. Benival and N. Kommineni, Versatility of Liposomes for Antisense Oligonucleotide Delivery: A Special Focus on Various Therapeutic Areas, Pharmaceutics, 2023, 15, 1435 CrossRef CAS PubMed.
  52. M. Gagliardi and A. T. Ashizawa, The Challenges and Strategies of Antisense Oligonucleotide Drug Delivery, Biomedicines, 2021, 9, 433 CrossRef CAS PubMed.
  53. J. E. Rosenberg, N. M. Hahn, M. M. Regan, L. Werner, A. Alva, S. George, J. Picus, R. Alter, A. Balar, J. Hoffman-Censits, P. Grivas, R. Lauer, E. A. Guancial, C. Hoimes, G. Sonpavde, C. Albany, M. N. Stein, T. Breen, C. Jacobs, K. Anderson, J. Bellmunt, A.-K. A. Lalani, S. Pal and T. K. Choueiri, Apatorsen plus docetaxel versus docetaxel alone in platinum-resistant metastatic urothelial carcinoma (Borealis-2), Br. J. Cancer, 2018, 118, 1434–1441 CrossRef CAS PubMed.
  54. A. Khuu, M. Verreault, P. Colin, H. Tran and A. Idbaih, Clinical Applications of Antisense Oligonucleotides in Cancer: A Focus on Glioblastoma, Cells, 2024, 13, 1869 CrossRef CAS PubMed.
  55. L. Wen, C. Zhao, J. Song, L. Ma, J. Ruan, X. Xia, Y. E. Chen, J. Zhang, P. X. Ma and J. Xu, CRISPR/Cas9-Mediated TERT Disruption in Cancer Cells, Int. J. Mol. Sci., 2020, 21, 653 CrossRef CAS PubMed.
  56. A. D. Cox, S. W. Fesik, A. C. Kimmelman, J. Luo and C. J. Der, Drugging the undruggable RAS: Mission Possible?, Nat. Rev. Drug Discovery, 2014, 13, 828–851 CrossRef CAS PubMed.
  57. E. Lentsch, L. Li, S. Pfeffer, A. B. Ekici, L. Taher, C. Pilarsky and R. Grützmann, CRISPR/Cas9-Mediated Knock-Out of KrasG12D Mutated Pancreatic Cancer Cell Lines, Int. J. Mol. Sci., 2019, 20, 5706 CrossRef CAS PubMed.
  58. B. Vogelstein, N. Papadopoulos, V. E. Velculescu, S. Zhou, L. A. Diaz and K. W. Kinzler, Cancer genome landscapes, Science, 2013, 339, 1546–1558 CrossRef CAS PubMed.
  59. A. Witz, J. Dardare, A. Francois, M. Husson, M. Rouyer, J. Demange, J.-L. Merlin, P. Gilson and A. Harlé, CRISPR/Cas9-mediated knock-in of BRCA1/2 mutations restores response to olaparib in pancreatic cancer cell lines, Sci. Rep., 2023, 13, 18741 CrossRef CAS PubMed.
  60. V. Barresi, C. Musmeci, A. Rinaldi and D. F. Condorelli, Transcript-Targeted Therapy Based on RNA Interference and Antisense Oligonucleotides: Current Applications and Novel Molecular Targets, Int. J. Mol. Sci., 2022, 23, 8875 CrossRef CAS PubMed.
  61. R. Hernandez, J. Põder, K. M. LaPorte and T. R. Malek, Engineering IL-2 for immunotherapy of autoimmunity and cancer, Nat. Rev. Immunol., 2022, 22, 614–628 CrossRef CAS PubMed.
  62. A. Algazi, S. Bhatia, S. Agarwala, M. Molina, K. Lewis, M. Faries, L. Fong, L. P. Levine, M. Franco, A. Oglesby, C. Ballesteros-Merino, C. B. Bifulco, B. A. Fox, D. Bannavong, R. Talia, E. Browning, M. H. Le, R. H. Pierce, S. Gargosky, K. K. Tsai, C. Twitty and A. I. Daud, Intratumoral delivery of tavokinogene telseplasmid yields systemic immune responses in metastatic melanoma patients, Ann. Oncol., 2020, 31, 532–540 CrossRef CAS PubMed.
  63. M. E. Raeber, D. Sahin, U. Karakus and O. Boyman, A systematic review of interleukin-2-based immunotherapies in clinical trials for cancer and autoimmune diseases, EBioMedicine, 2023, 90, 104539 CrossRef CAS PubMed.
  64. A. Wu, S. Oh, K. Ericson, Z. L. Demorest, I. Vengco, S. Gharagozlou, W. Chen, W. C. Low and J. R. Ohlfest, Transposon-based interferon gamma gene transfer overcomes limitations of episomal plasmid for immunogene therapy of glioblastoma, Cancer Gene Ther., 2007, 14, 550–560 CrossRef CAS PubMed.
  65. K. P. Papadopoulos, M. Tehfe, A. R. Abdul Razak, S. Pant, V. Chung, D. W. Rasco, R. Jamal, J. Willis, J. Fuentes Antrás, K. Brettschneider, A. Zamorski, H. Bähr-Mahmud, S. Koseoglu, P. K. Chang, C. Lindemann, A. M. Schlitter, R. Magenheim, L. De Mattos-Arruda, Ö. Türeci and U. Sahin, A phase I/II dose escalation and expansion trial to evaluate safety and preliminary efficacy of BNT141 in patients with claudin-18.2-positive solid tumors, J. Clin. Oncol., 2023, 41, TPS2670 CrossRef.
  66. H. Kim, S. N. Danishmalik, H. Hwang, J.-I. Sin, J. Oh, Y. Cho, H. Lee, M. Jeong, S.-H. Kim and H. J. Hong, Gene therapy using plasmid DNA-encoded anti-HER2 antibody for cancers that overexpress HER2, Cancer Gene Ther., 2016, 23, 341–347 CrossRef CAS PubMed.
  67. N. Sugii, M. Matsuda, G. Okumura, A. Shibuya, E. Ishikawa, Y. Kaneda and A. Matsumura, Hemagglutinating virus of Japan-envelope containing programmed cell death-ligand 1 siRNA inhibits immunosuppressive activities and elicits antitumor immune responses in glioma, Cancer Sci., 2021, 112, 81–90 CrossRef CAS PubMed.
  68. M. Eisenstein, A visual guide to restoring the guardian of the genome, Nature, 2022, 603, S2–S3 CrossRef CAS.
  69. N. Kong, W. Tao, X. Ling, J. Wang, Y. Xiao, S. Shi, X. Ji, A. Shajii, S. T. Gan, N. Y. Kim, D. G. Duda, T. Xie, O. C. Farokhzad and J. Shi, Synthetic mRNA nanoparticle-mediated restoration of p53 tumor suppressor sensitizes p53-deficient cancers to mTOR inhibition, Sci. Transl. Med., 2019, 11, eaaw1565 CrossRef CAS PubMed.
  70. Y. Liu, D. Zhang, Y. An, Y. Sun, J. Li, M. Zheng, Y. Zou and B. Shi, Non-invasive PTEN mRNA brain delivery effectively mitigates growth of orthotopic glioblastoma, Nano Today, 2023, 49, 101790 CrossRef CAS.
  71. Y. Xiao, J. Chen, H. Zhou, X. Zeng, Z. Ruan, Z. Pu, X. Jiang, A. Matsui, L. Zhu, Z. Amoozgar, D. S. Chen, X. Han, D. G. Duda and J. Shi, Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy, Nat. Commun., 2022, 13, 758 CrossRef CAS PubMed.
  72. C. Moses, F. Nugent, C. B. Waryah, B. Garcia-Bloj, A. R. Harvey and P. Blancafort, Activating PTEN Tumor Suppressor Expression with the CRISPR/dCas9 System, Mol. Ther. – Nucleic Acids, 2019, 14, 287–300 CrossRef CAS PubMed.
  73. Z. Hu, W. Ding, D. Zhu, L. Yu, X. Jiang, X. Wang, C. Zhang, L. Wang, T. Ji, D. Liu, D. He, X. Xia, T. Zhu, J. Wei, P. Wu, C. Wang, L. Xi, Q. Gao, G. Chen, R. Liu, K. Li, S. Li, S. Wang, J. Zhou, D. Ma and H. Wang, TALEN-mediated targeting of HPV oncogenes ameliorates HPV-related cervical malignancy, J. Clin. Invest., 2015, 125, 425–436 CrossRef PubMed.
  74. D. J. Mitchell and R. F. Minchin, E. coli nitroreductase/CB1954 gene-directed enzyme prodrug therapy: role of arylamine N-acetlytransferase 2, Cancer Gene Ther., 2008, 15, 758–764 CrossRef CAS PubMed.
  75. T. Oishi, M. Ito, S. Koizumi, M. Horikawa, T. Yamamoto, S. Yamagishi, T. Yamasaki, T. Sameshima, T. Suzuki, H. Sugimura, H. Namba and K. Kurozumi, Efficacy of HSV-TK/GCV system suicide gene therapy using SHED expressing modified HSV-TK against lung cancer brain metastases, Mol. Ther. – Methods Clin. Dev., 2022, 26, 253–265 CrossRef CAS PubMed.
  76. L. A. Mitchell, F. Lopez Espinoza, D. Mendoza, Y. Kato, A. Inagaki, K. Hiraoka, N. Kasahara, H. E. Gruber, D. J. Jolly and J. M. Robbins, Toca 511 gene transfer and treatment with the prodrug, 5-fluorocytosine, promotes durable antitumor immunity in a mouse glioma model, Neurooncology, 2017, 19, 930–939 CAS.
  77. J.-H. Wang, D. J. Gessler, W. Zhan, T. L. Gallagher and G. Gao, Adeno-associated virus as a delivery vector for gene therapy of human diseases, Signal Transduction Targeted Ther., 2024, 9, 1–33 CrossRef CAS PubMed.
  78. S. Chaurasiya, H. Valencia, Z. Zhang, S.-I. Kim, A. Yang, J. Lu, Y. Woo, S. G. Warner, N. J. Ede and Y. Fong, An Oncolytic Poxvirus Encoding hNIS, Shows Antitumor Efficacy and Allows Tumor Imaging in a Liver Cancer Model, Mol. Cancer Ther., 2023, OF1–OF9 Search PubMed.
  79. A. L. de Matos, L. S. Franco and G. McFadden, Oncolytic Viruses and the Immune System: The Dynamic Duo, Mol. Ther. – Methods Clin. Dev., 2020, 17, 349–358 CrossRef PubMed.
  80. U. Bezeljak, Cancer gene therapy goes viral: viral vector platforms come of age, Radiol. Oncol., 2022, 56, 1–13 CrossRef CAS PubMed.
  81. J. L. Shirley, Y. P. de Jong, C. Terhorst and R. W. Herzog, Immune Responses to Viral Gene Therapy Vectors, Mol. Ther., 2020, 28, 709–722 CrossRef CAS PubMed.
  82. M. Zheng, J. Huang, A. Tong and H. Yang, Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances, Mol. Ther.: Oncol., 2019, 15, 234–247 Search PubMed.
  83. O. Humbert, L. Davis and N. Maizels, Targeted gene therapies: tools, applications, optimization, Crit. Rev. Biochem. Mol. Biol., 2012, 47, 264–281 CrossRef CAS PubMed.
  84. A. J. Brenner, K. B. Peters, J. Vredenburgh, F. Bokstein, D. T. Blumenthal, S. Yust-Katz, I. Peretz, B. Oberman, L. S. Freedman, B. M. Ellingson, T. F. Cloughesy, N. Sher, Y. C. Cohen, N. Lowenton-Spier, T. Rachmilewitz Minei, N. Yakov, I. Mendel, E. Breitbart and P. Y. Wen, Safety and efficacy of VB-111, an anticancer gene therapy, in patients with recurrent glioblastoma: results of a phase I/II study, Neurooncology, 2020, 22, 694–704 CAS.
  85. A.-R. Yoon, J. Hong, M. Kim and C.-O. Yun, Hepatocellular carcinoma-targeting oncolytic adenovirus overcomes hypoxic tumor microenvironment and effectively disperses through both central and peripheral tumor regions, Sci. Rep., 2018, 8, 2233 CrossRef PubMed.
  86. J. A. Barrett, H. Cai, J. Miao, P. D. Khare, P. Gonzalez, J. Dalsing-Hernandez, G. Sharma, T. Chan, L. J. N. Cooper and F. Lebel, Regulated intratumoral expression of IL-12 using a RheoSwitch Therapeutic System® (RTS®) gene switch as gene therapy for the treatment of glioma, Cancer Gene Ther., 2018, 25, 106–116 CrossRef CAS PubMed.
  87. J. T. Huckaby, E. Landoni, T. M. Jacobs, B. Savoldo, G. Dotti and S. K. Lai, Bispecific binder redirected lentiviral vector enables in vivo engineering of CAR-T cells, J. ImmunoTher. Cancer, 2021, 9, e002737 CrossRef PubMed.
  88. W.-C. Wang, E. E. Sayedahmed and S. K. Mittal, Significance of Preexisting Vector Immunity and Activation of Innate Responses for Adenoviral Vector-Based Therapy, Viruses, 2022, 14, 2727 CrossRef CAS PubMed.
  89. B. Ouyang, W. Poon, Y.-N. Zhang, Z. P. Lin, B. R. Kingston, A. J. Tavares, Y. Zhang, J. Chen, M. S. Valic, A. M. Syed, P. MacMillan, J. Couture-Senécal, G. Zheng and W. C. W. Chan, The dose threshold for nanoparticle tumour delivery, Nat. Mater., 2020, 19, 1362–1371 CrossRef CAS PubMed.
  90. S. Rangarajan, L. Walsh, W. Lester, D. Perry, B. Madan, M. Laffan, H. Yu, C. Vettermann, G. F. Pierce, W. Y. Wong and K. J. Pasi, AAV5−Factor VIII Gene Transfer in Severe Hemophilia A, N. Engl. J. Med., 2017, 377, 2519–2530 CrossRef CAS PubMed.
  91. S. Hacein-Bey-Abina, A. Garrigue, G. P. Wang, J. Soulier, A. Lim, E. Morillon, E. Clappier, L. Caccavelli, E. Delabesse, K. Beldjord, V. Asnafi, E. MacIntyre, L. Dal Cortivo, I. Radford, N. Brousse, F. Sigaux, D. Moshous, J. Hauer, A. Borkhardt, B. H. Belohradsky, U. Wintergerst, M. C. Velez, L. Leiva, R. Sorensen, N. Wulffraat, S. Blanche, F. D. Bushman, A. Fischer and M. Cavazzana-Calvo, Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1, J. Clin. Invest., 2008, 118, 3132–3142 CrossRef CAS PubMed.
  92. C. T. Nicolas, C. J. VanLith, R. D. Hickey, Z. Du, L. G. Hillin, R. M. Guthman, W. J. Cao, B. Haugo, A. Lillegard, D. Roy, A. Bhagwate, D. O'Brien, J.-P. Kocher, R. A. Kaiser, S. J. Russell and J. B. Lillegard, In vivo lentiviral vector gene therapy to cure hereditary tyrosinemia type 1 and prevent development of precancerous and cancerous lesions, Nat. Commun., 2022, 13, 5012 CrossRef CAS PubMed.
  93. D. Lindemann and A. Rethwilm, Foamy virus biology and its application for vector development, Viruses, 2011, 3, 561–585 CrossRef CAS PubMed.
  94. M. A. Goodman, P. Arumugam, D. M. Pillis, A. Loberg, M. Nasimuzzaman, D. Lynn, J. C. M. van der Loo, P. J. Dexheimer, M. Keddache, T. R. Bauer, D. D. Hickstein, D. W. Russell and P. Malik, Foamy Virus Vector Carries a Strong Insulator in Its Long Terminal Repeat Which Reduces Its Genotoxic Potential, J. Virol., 2018, 92, e01639–e01617 CrossRef PubMed.
  95. K. M. Budzik, R. A. Nace, Y. Ikeda and S. J. Russell, Evaluation of the stability and intratumoral delivery of foreign transgenes encoded by an oncolytic Foamy Virus vector, Cancer Gene Ther., 2022, 29, 1240–1251 CrossRef CAS PubMed.
  96. E. M. Everson, M. E. Olzsko, D. J. Leap, J. D. Hocum and G. D. Trobridge, A comparison of foamy and lentiviral vector genotoxicity in SCID-repopulating cells shows foamy vectors are less prone to clonal dominance, Mol. Ther. – Methods Clin. Dev., 2016, 3, 16048 CrossRef PubMed.
  97. E. M. Gordon and F. L. Hall, Rexin-G, a targeted genetic medicine for cancer, Expert Opin. Biol. Ther., 2010, 10, 819–832 CrossRef CAS PubMed.
  98. S. P. Chawla, H. Bruckner, M. A. Morse, N. Assudani, F. L. Hall and E. M. Gordon, A Phase I-II Study Using Rexin-G Tumor-Targeted Retrovector Encoding a Dominant-Negative Cyclin G1 Inhibitor for Advanced Pancreatic Cancer, Mol. Ther.: Oncol., 2019, 12, 56–67 CAS.
  99. H. Kushiya, K. Hiraoka, T. Suzuki, K. Inoko, A. Inagaki, H. Niwa, K. Sasaki, T. Nakamura, T. Tsuchikawa, T. Shichinohe, D. J. Jolly, N. Kasahara and S. Hirano, Retroviral Replicating Vector Toca 511 (Vocimagene Amiretrorepvec) for Prodrug Activator Gene Therapy of Lung Cancer, Cancers, 2022, 14, 5820 CrossRef CAS PubMed.
  100. S. Naik and L. Russell, The 13th International Oncolytic Virus Conference: Powerful payloads gain clinical momentum, Mol. Ther., 2022, 30, 1361–1363 CrossRef CAS PubMed.
  101. P. D. Trivedi, B. J. Byrne and M. Corti, Evolving Horizons: Adenovirus Vectors’ Timeless Influence on Cancer, Gene Therapy and Vaccines, Viruses, 2023, 15, 2378 CrossRef CAS PubMed.
  102. C. S. Lee, E. S. Bishop, R. Zhang, X. Yu, E. M. Farina, S. Yan, C. Zhao, Z. Zeng, Y. Shu, X. Wu, J. Lei, Y. Li, W. Zhang, C. Yang, K. Wu, Y. Wu, S. Ho, A. Athiviraham, M. J. Lee, J. M. Wolf, R. R. Reid and T.-C. He, Adenovirus-mediated gene delivery: Potential applications for gene and cell-based therapies in the new era of personalized medicine, Genes Dis., 2017, 4, 43 CrossRef PubMed.
  103. M. Salauddin, S. Saha, M. G. Hossain, K. Okuda and M. Shimada, Clinical Application of Adenovirus (AdV): A Comprehensive Review, Viruses, 2024, 16, 1094 CrossRef CAS PubMed.
  104. Y. Li, W. Guo, X. Li, J. Zhang, M. Sun, Z. Tang, W. Ran, K. Yang, G. Huang and L. Li, Expert consensus on the clinical application of recombinant adenovirus human p53 for head and neck cancers, Int. J. Oral Sci., 2021, 13, 38 CrossRef CAS PubMed.
  105. E. Ortiz-Zapater, G. Santis and M. Parsons, CAR: A key regulator of adhesion and inflammation, Int. J. Biochem. Cell Biol., 2017, 89, 1–5 CrossRef CAS PubMed.
  106. L. Coughlan, E. J. Kremer and D. M. Shayakhmetov, Adenovirus-based vaccines—a platform for pandemic preparedness against emerging viral pathogens, Mol. Ther., 2022, 30, 1822–1849 CrossRef CAS PubMed.
  107. A. Lee, Nadofaragene Firadenovec: First Approval, Drugs, 2023, 83, 353–357 CrossRef CAS PubMed.
  108. V. M. Narayan, S. A. Boorjian, M. Alemozaffar, B. R. Konety, N. D. Shore, L. G. Gomella, A. M. Kamat, T. J. Bivalacqua, J. S. Montgomery, S. P. Lerner, J. E. Busby, M. Poch, P. L. Crispen, G. D. Steinberg, A. K. Schuckman, T. M. Downs, J. Mashni, B. R. Lane, T. J. Guzzo, G. Bratslavsky, L. I. Karsh, M. E. Woods, G. Brown, D. Canter, A. Luchey, Y. Lotan, B. A. Inman, M. B. Williams, M. S. Cookson, S. S. Chang, A. I. Sankin, M. A. O'Donnell, D. Sawutz, R. Philipson, N. R. Parker, S. Yla-Herttuala, D. Rehm, J. S. Jakobsen, K. Juul and C. P. N. Dinney, Efficacy of Intravesical Nadofaragene Firadenovec for Patients With Bacillus Calmette-Guérin-Unresponsive Nonmuscle-Invasive Bladder Cancer: 5-Year Follow-Up From a Phase 3 Trial, J. Urol., 2024, 212, 74–86 CrossRef PubMed.
  109. M. S. Koch, M. Zdioruk, M. O. Nowicki, M. S. Hoetker, Z. T. Herbert, F. Barone, P. P. Tak, E. A. Chiocca, G. Tabatabai and S. E. Lawler, Uncovering transcriptomic landscape alterations of CAN-2409 in in vitro and in vivo glioma models, Front. Med., 2023, 10, 1140352 CrossRef PubMed.
  110. H. Jiang, D. H. Shin, T. T. Nguyen, J. Fueyo, X. Fan, V. Henry, C. C. Carrillo, Y. Yi, M. M. Alonso, T. L. Collier, Y. Yuan, F. F. Lang and C. Gomez-Manzano, Localized Treatment with Oncolytic Adenovirus Delta-24-RGDOX Induces Systemic Immunity against Disseminated Subcutaneous and Intracranial Melanomas, Clin. Cancer Res., 2019, 25, 6801–6814 CrossRef CAS PubMed.
  111. H. Jiang, D. H. Shin, Y. Yi, X. Fan, J. Gumin, J. He, A. G. Gillard, F. F. Lang, C. Gomez-Manzano and J. Fueyo, Adjuvant Therapy with Oncolytic Adenovirus Delta-24-RGDOX After Intratumoral Adoptive T-cell Therapy Promotes Antigen Spread to Sustain Systemic Antitumor Immunity, Cancer Res. Commun., 2023, 3, 1118–1131 CrossRef CAS PubMed.
  112. S. Atasheva and D. M. Shayakhmetov, Cytokine Responses to Adenovirus and Adenovirus Vectors, Viruses, 2022, 14, 888 CrossRef CAS PubMed.
  113. M. D. J. Dicks, A. J. Spencer, N. J. Edwards, G. Wadell, K. Bojang, S. C. Gilbert, A. V. S. Hill and M. G. Cottingham, A Novel Chimpanzee Adenovirus Vector with Low Human Seroprevalence: Improved Systems for Vector Derivation and Comparative Immunogenicity, PLoS One, 2012, 7, e40385 CrossRef CAS PubMed.
  114. A. R. Rappaport, C. Kyi, M. Lane, M. G. Hart, M. L. Johnson, B. S. Henick, C.-Y. Liao, A. Mahipal, A. Shergill, A. I. Spira, J. W. Goldman, C. D. Scallan, D. Schenk, C. D. Palmer, M. J. Davis, S. Kounlavouth, L. Kemp, A. Yang, Y. J. Li, M. Likes, A. Shen, G. R. Boucher, M. Egorova, R. L. Veres, J. A. Espinosa, J. R. Jaroslavsky, L. D. Kraemer Tardif, L. Acrebuche, C. Puccia, L. Sousa, R. Zhou, K. Bae, J. R. Hecht, D. P. Carbone, B. Johnson, A. Allen, A. R. Ferguson and K. Jooss, A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: phase 1 trial interim results, Nat. Med., 2024, 30, 1013–1022 CrossRef CAS PubMed.
  115. D. GuhaSarkar, J. Neiswender, Q. Su, G. Gao and M. Sena-Esteves, Intracranial AAV-IFN-β gene therapy eliminates invasive xenograft glioblastoma and improves survival in orthotopic syngeneic murine model, Mol. Oncol., 2017, 11, 180–193 CrossRef CAS PubMed.
  116. R. C. Münch, H. Janicki, I. Völker, A. Rasbach, M. Hallek, H. Büning and C. J. Buchholz, Displaying high-affinity ligands on adeno-associated viral vectors enables tumor cell-specific and safe gene transfer, Mol. Ther., 2013, 21, 109–118 CrossRef PubMed.
  117. J. Ai, D. Wang, Q. Wei, H. Li and G. Gao, Adeno-associated Virus Serotype Vectors Efficiently Transduce Normal Prostate Tissue and Prostate Cancer Cells, Eur. Urol., 2016, 69, 179–181 CrossRef PubMed.
  118. F. Borel, M. A. Kay and C. Mueller, Recombinant AAV as a Platform for Translating the Therapeutic Potential of RNA Interference, Mol. Ther., 2014, 22, 692–701 CrossRef CAS PubMed.
  119. W. Nawaz, B. Huang, S. Xu, Y. Li, L. Zhu, H. Yiqiao, Z. Wu and X. Wu, AAV-mediated in vivo CAR gene therapy for targeting human T-cell leukemia, J. Blood Cancer, 2021, 11, 1–12 CrossRef PubMed.
  120. W. Laurens, C. Joussain, D. Pierre and H. John, Unlocking potential: Herpes Simplex Virus Type 1-based gene therapy in functional urology, Continence, 2024, 10, 101311 CrossRef.
  121. D. J. Fink, J. Wechuck, M. Mata, J. C. Glorioso, J. Goss, D. Krisky and D. Wolfe, Gene therapy for pain: results of a phase I clinical trial, Ann. Neurol., 2011, 70, 207–212 CrossRef CAS PubMed.
  122. A. L. Epstein and S. D. Rabkin, Safety of non-replicative and oncolytic replication-selective HSV vectors, Trends Mol. Med., 2024, 30, 781–794 CrossRef CAS PubMed.
  123. J. R. Hecht, S. S. Raman, A. Chan, K. Kalinsky, J.-F. Baurain, M. M. Jimenez, M. M. Garcia, M. D. Berger, U. M. Lauer, A. Khattak, A. Carrato, Y. Zhang, K. Liu, E. Cha, A. Keegan, S. Bhatta, C. P. Strassburg and A. Roohullah, Phase Ib study of talimogene laherparepvec in combination with atezolizumab in patients with triple negative breast cancer and colorectal cancer with liver metastases, ESMO Open, 2023, 8, 100884 CrossRef CAS PubMed.
  124. C. T. Hiley, M. Yuan, N. R. Lemoine and Y. Wang, Lister strain vaccinia virus, a potential therapeutic vector targeting hypoxic tumours, Gene Ther., 2010, 17, 281–287 CrossRef CAS PubMed.
  125. H. Jin, Y.-R. Zhao, F. Huang, Z. Hong, X.-Y. Jia, H. Wang and Y.-G. Wang, Vaccinia virus-mediated oncolytic immunotherapy: Emerging strategies for gastrointestinal cancer treatment at dawn, Virology, 2025, 602, 110303 CrossRef CAS PubMed.
  126. J. Béguin, J. Foloppe, C. Maurey, E. Laloy, J. Hortelano, V. Nourtier, C. Pichon, S. Cochin, P. Cordier, H. Huet, E. Quemeneur, B. Klonjkowski and P. Erbs, Preclinical Evaluation of the Oncolytic Vaccinia Virus TG6002 by Translational Research on Canine Breast Cancer, Mol. Ther.: Oncol., 2020, 19, 57 Search PubMed.
  127. G. K. Abou-Alfa, P. R. Galle, Y. Chao, J. Erinjeri, J. Heo, M. J. Borad, A. Luca, J. Burke, A. Pelusio, D. Agathon, M. Lusky, C. Breitbach, S. Qin and E. Gane, PHOCUS: A Phase 3, Randomized, Open-Label Study of Sequential Treatment with Pexa-Vec (JX-594) and Sorafenib in Patients with Advanced Hepatocellular Carcinoma, Liver Cancer, 2024, 13, 248–264 CrossRef PubMed.
  128. F. Blackhall, L. Moliner, A. Samson, K. Franks, I. Karydis, D. Joseph-Pietras, O. Frank, H. Lavender, M. Jannoo, M. Marshall, P. Motteram, B. J. van den Eynde, C. Leung, A. Hill, M. R. Middleton, N. A. Coupe, N. Steele, J. Spicer, P. H. Shaw and S. N. Symeonides, 1502TiP A phase I/IIa trial of ChAdOx1 and MVA vaccines against MAGE-A3 and NY-ESO-1, Ann. Oncol., 2023, 34, S846 CrossRef.
  129. J. McAuliffe, H. F. Chan, L. Noblecourt, R. A. Ramirez-Valdez, V. Pereira-Almeida, Y. Zhou, E. Pollock, F. Cappuccini, I. Redchenko, A. V. Hill, C. S. K. Leung and B. J. Van den Eynde, Heterologous prime-boost vaccination targeting MAGE-type antigens promotes tumor T-cell infiltration and improves checkpoint blockade therapy, J. ImmunoTher. Cancer, 2021, 9, e003218 CrossRef PubMed.
  130. Y. Woo, Z. Zhang, A. Yang, S. Chaurasiya, A. K. Park, J. Lu, S.-I. Kim, S. G. Warner, D. Von Hoff and Y. Fong, Novel Chimeric Immuno-Oncolytic Virus CF33-hNIS-antiPDL1 for the Treatment of Pancreatic Cancer, J. Am. Coll. Surg., 2020, 230, 709–717 CrossRef PubMed.
  131. Z. Zhang, A. Yang, S. Chaurasiya, A. K. Park, S.-I. Kim, J. Lu, H. Valencia, Y. Fong and Y. Woo, Anti-Tumor Immunogenicity of the Oncolytic Virus CF33-hNIS-antiPDL1 against Ex Vivo Peritoneal Cells from Gastric Cancer Patients, Int. J. Mol. Sci., 2023, 24, 14189 CrossRef CAS PubMed.
  132. D. Li, A. Seiz, S. H. Fein, J. C. H. Byon, G. A. Wilkinson and P. P. Woodard, Combination therapy with the oncolytic virus CF33-CD19 and blinatumomab for the treatment of advanced solid tumors, J. Clin. Oncol., 2024, 42, TPS2687 CrossRef.
  133. M. Ferrari, Cancer nanotechnology: opportunities and challenges, Nat. Rev. Cancer, 2005, 5, 161–171 CrossRef CAS PubMed.
  134. Y. Nakamura, A. Mochida, P. L. Choyke and H. Kobayashi, Nanodrug Delivery: Is the Enhanced Permeability and Retention Effect Sufficient for Curing Cancer?, Bioconjugate Chem., 2016, 27, 2225–2238 CrossRef CAS PubMed.
  135. Z. Hussain, S. Khan, M. Imran, M. Sohail, S. W. A. Shah and M. de Matas, PEGylation: a promising strategy to overcome challenges to cancer-targeted nanomedicines: a review of challenges to clinical transition and promising resolution, Drug Delivery Transl. Res., 2019, 9, 721–734 CrossRef CAS PubMed.
  136. C. Liu, L. Zhang, W. Zhu, R. Guo, H. Sun, X. Chen and N. Deng, Barriers and Strategies of Cationic Liposomes for Cancer Gene Therapy, Mol. Ther. – Methods Clin. Dev., 2020, 18, 751–764 CrossRef CAS PubMed.
  137. J. Fang, R. Islam, W. Islam, H. Yin, V. Subr, T. Etrych, K. Ulbrich and H. Maeda, Augmentation of EPR Effect and Efficacy of Anticancer Nanomedicine by Carbon Monoxide Generating Agents, Pharmaceutics, 2019, 11, 343 CrossRef CAS PubMed.
  138. L. N. M. Nguyen, W. Ngo, Z. P. Lin, S. Sindhwani, P. MacMillan, S. M. Mladjenovic and W. C. W. Chan, The mechanisms of nanoparticle delivery to solid tumours, Nat. Rev. Bioeng., 2024, 2, 201–213 CrossRef CAS.
  139. Y. Chen, X. Zhu, X. Zhang, B. Liu and L. Huang, Nanoparticles Modified With Tumor-targeting scFv Deliver siRNA and miRNA for Cancer Therapy, Mol. Ther., 2010, 18, 1650–1656 CrossRef CAS PubMed.
  140. M. T. Luiz, J. A. P. Dutra, L. B. Tofani, J. T. C. de Araújo, L. D. Di Filippo, J. M. Marchetti and M. Chorilli, Targeted Liposomes: A Nonviral Gene Delivery System for Cancer Therapy, Pharmaceutics, 2022, 14, 821 CrossRef CAS PubMed.
  141. Y. Cai, R. Chen, S. Gao, W. Li, Y. Liu, G. Su, M. Song, M. Jiang, C. Jiang and X. Zhang, Artificial intelligence applied in neoantigen identification facilitates personalized cancer immunotherapy, Front. Oncol., 2022, 12, 1054231 CrossRef CAS PubMed.
  142. D. S. Nin and L.-W. Deng, Biology of Cancer-Testis Antigens and Their Therapeutic Implications in Cancer, Cells, 2023, 12, 926 CrossRef CAS PubMed.
  143. S. Chu, X. Shi, Y. Tian and F. Gao, pH-Responsive Polymer Nanomaterials for Tumor Therapy, Front. Oncol., 2022, 12, 855019 CrossRef CAS PubMed.
  144. M. Jia, D. Zhang, C. Zhang and C. Li, Nanoparticle-based delivery systems modulate the tumor microenvironment in pancreatic cancer for enhanced therapy, J. Nanobiotechnol., 2021, 19, 384 CrossRef CAS PubMed.
  145. X. Wang, T. Ishida and H. Kiwada, Anti-PEG IgM elicited by injection of liposomes is involved in the enhanced blood clearance of a subsequent dose of PEGylated liposomes, J. Controlled Release, 2007, 119, 236–244 CrossRef CAS PubMed.
  146. W.-F. Lai and W.-T. Wong, Design of Polymeric Gene Carriers for Effective Intracellular Delivery, Trends Biotechnol., 2018, 36, 713–728 CrossRef CAS PubMed.
  147. J. Shi, M.-W. Huang, Z.-D. Lu, X.-J. Du, S. Shen, C.-F. Xu and J. Wang, Delivery of mRNA for regulating functions of immune cells, J. Controlled Release, 2022, 345, 494–511 CrossRef CAS PubMed.
  148. M. Panigaj, M. B. Johnson, W. Ke, J. McMillan, E. A. Goncharova, M. Chandler and K. A. Afonin, Aptamers as Modular Components of Therapeutic Nucleic Acid Nanotechnology, ACS Nano, 2019, 13, 12301–12321 CrossRef CAS PubMed.
  149. O. Fenton, K. Kauffman, J. Kaczmarek, R. McClellan, S. Jhunjhunwala, M. Tibbitt, M. Zeng, E. Appel, J. Dorkin, F. Mir, J. Yang, M. Oberli, M. Heartlein, F. DeRosa, R. Langer and D. Anderson, Synthesis and Biological Evaluation of Ionizable Lipid Materials for the In Vivo Delivery of Messenger RNA to B Lymphocytes, Adv. Mater., 2017, 29, 1606944 CrossRef PubMed.
  150. P. Laverman, M. G. Carstens, O. C. Boerman, E. T. Dams, W. J. Oyen, N. van Rooijen, F. H. Corstens and G. Storm, Factors affecting the accelerated blood clearance of polyethylene glycol-liposomes upon repeated injection, J. Pharmacol. Exp. Ther., 2001, 298, 607–612 CrossRef CAS PubMed.
  151. A. M. Shabana, B. Xu, Z. Schneiderman, J. Ma, C. C. Chen and E. Kokkoli, Targeted Liposomes Encapsulating miR-603 Complexes Enhance Radiation Sensitivity of Patient-Derived Glioblastoma Stem-Like Cells, Pharmaceutics, 2021, 13, 1115 CrossRef CAS PubMed.
  152. G. S. R. Raju, E. Pavitra, G. L. Varaprasad, S. S. Bandaru, G. P. Nagaraju, B. Farran, Y. S. Huh and Y.-K. Han, Nanoparticles mediated tumor microenvironment modulation: current advances and applications, J. Nanobiotechnol., 2022, 20, 274 CrossRef PubMed.
  153. M. Cordani, R. Strippoli and Á. Somoza, Nanomaterials as Inhibitors of Epithelial Mesenchymal Transition in Cancer Treatment, Cancers, 2019, 12, 25 CrossRef PubMed.
  154. Z. Zhang and M. R. King, Nanomaterials for the Capture and Therapeutic Targeting of Circulating Tumor Cells, Cell. Mol. Bioeng., 2017, 10, 275–294 CrossRef CAS PubMed.
  155. S. Z. Alshawwa, A. A. Kassem, R. M. Farid, S. K. Mostafa and G. S. Labib, Nanocarrier Drug Delivery Systems: Characterization, Limitations, Future Perspectives and Implementation of Artificial Intelligence, Pharmaceutics, 2022, 14, 883 CrossRef CAS PubMed.
  156. R. X. Zhang, J. Li, T. Zhang, M. A. Amini, C. He, B. Lu, T. Ahmed, H. Lip, A. M. Rauth and X. Y. Wu, Importance of integrating nanotechnology with pharmacology and physiology for innovative drug delivery and therapy – an illustration with firsthand examples, Acta Pharmacol. Sin., 2018, 39, 825–844 CrossRef CAS PubMed.
  157. P. Joyce, C. J. Allen, M. J. Alonso, M. Ashford, M. S. Bradbury, M. Germain, M. Kavallaris, R. Langer, T. Lammers, M. T. Peracchia, A. Popat, C. A. Prestidge, C. J. F. Rijcken, B. Sarmento, R. B. Schmid, A. Schroeder, S. Subramaniam, C. R. Thorn, K. A. Whitehead, C.-X. Zhao and H. A. Santos, A translational framework to DELIVER nanomedicines to the clinic, Nat. Nanotechnol., 2024, 19, 1597–1611 CrossRef CAS PubMed.
  158. J. Ju, M.-L. Huan, N. Wan, H. Qiu, S.-Y. Zhou and B.-L. Zhang, Novel Cholesterol-Based Cationic Lipids as Transfecting Agents of DNA for Efficient Gene Delivery, Int. J. Mol. Sci., 2015, 16, 5666–5681 CrossRef CAS PubMed.
  159. C. H. Albertsen, J. A. Kulkarni, D. Witzigmann, M. Lind, K. Petersson and J. B. Simonsen, The role of lipid components in lipid nanoparticles for vaccines and gene therapy, Adv. Drug Delivery Rev., 2022, 188, 114416 CrossRef PubMed.
  160. A. Akinc, M. A. Maier, M. Manoharan, K. Fitzgerald, M. Jayaraman, S. Barros, S. Ansell, X. Du, M. J. Hope, T. D. Madden, B. L. Mui, S. C. Semple, Y. K. Tam, M. Ciufolini, D. Witzigmann, J. A. Kulkarni, R. van der Meel and P. R. Cullis, The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs, Nat. Nanotechnol., 2019, 14, 1084–1087 CrossRef CAS PubMed.
  161. M. Schlich, R. Palomba, G. Costabile, S. Mizrahy, M. Pannuzzo, D. Peer and P. Decuzzi, Cytosolic delivery of nucleic acids: The case of ionizable lipid nanoparticles, Bioeng. Transl. Med., 2021, 6, e10213 CrossRef CAS PubMed.
  162. G. Guerrini, D. Magrì, S. Gioria, D. Medaglini and L. Calzolai, Characterization of nanoparticles-based vaccines for COVID-19, Nat. Nanotechnol., 2022, 17, 570–576 CrossRef CAS PubMed.
  163. A. Naing, G. Lopez-Berestein, S. Fu, A. M. Tsimberidou, S. Pant, S. A. Piha-Paul, F. Janku, D. S. Hong, S. Sulovic, X. Meng, A. A. Jazaeri, C. S. Ng, D. D. Karp, V. Subbiah, F. Meric-Bernstam, R. Mitra, S. Wu, A. Sood and R. L. Coleman, EphA2 gene targeting using neutral liposomal small interfering RNA (EPHARNA) delivery: A phase I clinical trial, J. Clin. Oncol., 2017, 35, TPS2604 Search PubMed.
  164. M. J. Wagner, R. Mitra, M. J. McArthur, W. Baze, K. Barnhart, S. Y. Wu, C. Rodriguez-Aguayo, X. Zhang, R. L. Coleman, G. Lopez-Berestein and A. K. Sood, Preclinical Mammalian Safety Studies of EPHARNA (DOPC Nanoliposomal EphA2-Targeted siRNA), Mol. Cancer Ther., 2017, 16, 1114–1123 CrossRef CAS PubMed.
  165. A. M. Tari, Y. Gutiérrez-Puente, G. Monaco, C. Stephens, T. Sun, M. Rosenblum, J. Belmont, R. Arlinghaus and G. Lopez-Berestein, Liposome-incorporated Grb2 antisense oligodeoxynucleotide increases the survival of mice bearing bcr-abl-positive leukemia xenografts, Int. J. Oncol., 2007, 31, 1243–1250 CAS.
  166. M. Ohanian, A. T. Ashizawa, G. Garcia-Manero, N. Pemmaraju, T. Kadia, E. Jabbour, F. Ravandi, G. Borthakur, M. Andreeff, M. Konopleva, M. Lim, S. Pierce, S. O'Brien, Y. Alvarado, S. Verstovsek, W. Wierda, H. Kantarjian and J. Cortes, Liposomal Grb2 antisense oligodeoxynucleotide (BP1001) in patients with refractory or relapsed haematological malignancies: a single-centre, open-label, dose-escalation, phase 1/1b trial, Lancet Haematol., 2018, 5, e136–e146 CrossRef PubMed.
  167. D. Sun and Z.-R. Lu, Structure and Function of Cationic and Ionizable Lipids for Nucleic Acid Delivery, Pharm. Res., 2023, 40, 27–46 CrossRef CAS PubMed.
  168. W. Xiao, W. Zhang, H. Huang, Y. Xie, Y. Zhang, X. Guo, C. Jin, X. Liao, S. Yao, G. Chen and X. Song, Cancer Targeted Gene Therapy for Inhibition of Melanoma Lung Metastasis with eIF3i shRNA Loaded Liposomes, Mol. Pharmaceutics, 2020, 17, 229–238 CrossRef CAS PubMed.
  169. E. R. Camp, C. Wang, E. C. Little, P. M. Watson, K. F. Pirollo, A. Rait, D. J. Cole, E. H. Chang and D. K. Watson, Transferrin receptor targeting nanomedicine delivering wild-type p53 gene sensitizes pancreatic cancer to gemcitabine therapy, Cancer Gene Ther., 2013, 20, 222–228 CrossRef CAS PubMed.
  170. S.-S. Kim, J. B. Harford, M. Moghe, C. Doherty and E. H. Chang, A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models, Cells, 2022, 11, 3434 CrossRef CAS PubMed.
  171. K. Kubota, K. Onishi, K. Sawaki, T. Li, K. Mitsuoka, T. Sato and S. Takeoka, Effect of the nanoformulation of siRNA-lipid assemblies on their cellular uptake and immune stimulation, Int. J. Nanomed., 2017, 12, 5121–5133 CrossRef CAS PubMed.
  172. Z. Wang, W. Li, Y. Jiang, J. Park, K. M. Gonzalez, X. Wu, Q.-Y. Zhang and J. Lu, Cholesterol-modified sphingomyelin chimeric lipid bilayer for improved therapeutic delivery, Nat. Commun., 2024, 15, 2073 CrossRef CAS PubMed.
  173. J. Zhou, L. Sun, Y. Jia, Z. Wang, T. Luo, J. Tan, X. Fang, H. Zhu, J. Wang, L. Yu and Z. Yan, Lipid nanoparticles produce chimeric antigen receptor T cells with interleukin-6 knockdown in vivo, J. Controlled Release, 2022, 350, 298–307 CrossRef CAS PubMed.
  174. A. M. Jörgensen, R. Wibel and A. Bernkop-Schnürch, Biodegradable Cationic and Ionizable Cationic Lipids: A Roadmap for Safer Pharmaceutical Excipients, Small, 2023, 19, e2206968 CrossRef PubMed.
  175. H. Lv, S. Zhang, B. Wang, S. Cui and J. Yan, Toxicity of cationic lipids and cationic polymers in gene delivery, J. Controlled Release, 2006, 114, 100–109 CrossRef CAS PubMed.
  176. Y. Zou, Y. Zhen, Y. Zhao, H. Chen, R. Wang, W. Wang, P. Ma, D. Zhi, B. Ju and S. Zhang, pH-sensitive, tail-modified, ester-linked ionizable cationic lipids for gene delivery, Biomater. Adv., 2022, 139, 212984 CrossRef CAS PubMed.
  177. A. I. S. van den Berg, C.-O. Yun, R. M. Schiffelers and W. E. Hennink, Polymeric delivery systems for nucleic acid therapeutics: Approaching the clinic, J. Controlled Release, 2021, 331, 121–141 CrossRef CAS PubMed.
  178. B. D. Monnery, M. Wright, R. Cavill, R. Hoogenboom, S. Shaunak, J. H. G. Steinke and M. Thanou, Cytotoxicity of polycations: Relationship of molecular weight and the hydrolytic theory of the mechanism of toxicity, Int. J. Pharm., 2017, 521, 249–258 CrossRef CAS PubMed.
  179. J. Luten, C. F. van Nostrum, S. C. De Smedt and W. E. Hennink, Biodegradable polymers as non-viral carriers for plasmid DNA delivery, J. Controlled Release, 2008, 126, 97–110 CrossRef CAS PubMed.
  180. C.-K. Chen, P.-K. Huang, W.-C. Law, C.-H. Chu, N.-T. Chen and L.-W. Lo, Biodegradable Polymers for Gene-Delivery Applications, Int. J. Nanomed., 2020, 15, 2131–2150 CrossRef CAS PubMed.
  181. J. W. Salameh, L. Zhou, S. M. Ward, C. F. Santa Chalarca, T. Emrick and M. L. Figueiredo, Polymer-mediated gene therapy: Recent advances and merging of delivery techniques, Wiley Interdiscip. Rev.: Nanomed. Nanobiotechnol., 2020, 12, e1598 Search PubMed.
  182. H. O. Alsaab, F. D. Alharbi, A. S. Alhibs, N. B. Alanazi, B. Y. Alshehri, M. A. Saleh, F. S. Alshehri, M. A. Algarni, T. Almugaiteeb, M. N. Uddin and R. M. Alzhrani, PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases, Pharmaceutics, 2022, 14, 2728 CrossRef CAS PubMed.
  183. N. Aibani, T. N. Khan and B. Callan, Liposome mimicking polymersomes; A comparative study of the merits of polymersomes in terms of formulation and stability, Int. J. Pharm.:X, 2020, 2, 100040 CAS.
  184. T. Anajafi and S. Mallik, Polymersome-based drug-delivery strategies for cancer therapeutics, Ther. Delivery, 2015, 6, 521–534 CrossRef CAS PubMed.
  185. J. Chandra, N. Molugulu, S. Annadurai, S. Wahab, R. Karwasra, S. Singh, R. Shukla and P. Kesharwani, Hyaluronic acid-functionalized lipoplexes and polyplexes as emerging nanocarriers for receptor-targeted cancer therapy, Environ. Res., 2023, 233, 116506 CrossRef CAS PubMed.
  186. P. Evenou, J. Rossignol, G. Pembouong, A. Gothland, D. Colesnic, R. Barbeyron, S. Rudiuk, A.-G. Marcelin, M. Ménand, D. Baigl, V. Calvez, L. Bouteiller and M. Sollogoub, Bridging β-Cyclodextrin Prevents Self-Inclusion, Promotes Supramolecular Polymerization, and Promotes Cooperative Interaction with Nucleic Acids, Angew. Chem., Int. Ed., 2018, 57, 7753–7758 CrossRef CAS PubMed.
  187. J. Kim, S. K. Mondal, S. Y. Tzeng, Y. Rui, R. Al-Kharboosh, K. K. Kozielski, A. G. Bhargav, C. A. Garcia, A. Quiñones-Hinojosa and J. J. Green, Poly(ethylene glycol)-Poly(beta-amino ester)-Based Nanoparticles for Suicide Gene Therapy Enhance Brain Penetration and Extend Survival in a Preclinical Human Glioblastoma Orthotopic Xenograft Model, ACS Biomater. Sci. Eng., 2020, 6, 2943–2955 CrossRef CAS PubMed.
  188. J. E. Zuckerman, I. Gritli, A. Tolcher, J. D. Heidel, D. Lim, R. Morgan, B. Chmielowski, A. Ribas, M. E. Davis and Y. Yen, Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA, Proc. Natl. Acad. Sci. U. S. A., 2014, 111, 11449–11454 CrossRef CAS PubMed.
  189. J. A. Kemp and Y. J. Kwon, Cancer nanotechnology: current status and perspectives, Nano Convergence, 2021, 8, 34 CrossRef CAS PubMed.
  190. S. Zawadzki, Á. Martín-Serrano, E. Okła, M. Kędzierska, S. Garcia-Gallego, P. O. López, F. J. de la Mata, S. Michlewska, T. Makowski, M. Ionov, E. Pędziwiatr-Werbicka, M. Bryszewska and K. Miłowska, Synthesis and biophysical evaluation of carbosilane dendrimers as therapeutic siRNA carriers, Sci. Rep., 2024, 14, 1615 CrossRef CAS PubMed.
  191. P. Kesharwani, S. Banerjee, U. Gupta, M. C. I. Mohd Amin, S. Padhye, F. H. Sarkar and A. K. Iyer, PAMAM dendrimers as promising nanocarriers for RNAi therapeutics, Mater. Today, 2015, 18, 565–572 CrossRef CAS.
  192. L. Chen, J. Li, Y. Fan, J. Qiu, L. Cao, R. Laurent, S. Mignani, A.-M. Caminade, J.-P. Majoral and X. Shi, Revisiting Cationic Phosphorus Dendrimers as a Nonviral Vector for Optimized Gene Delivery Toward Cancer Therapy Applications, Biomacromolecules, 2020, 21, 2502–2511 CrossRef CAS PubMed.
  193. K. Białkowska, P. Komorowski, R. Gomez-Ramirez, F. J. de la Mata, M. Bryszewska and K. Miłowska, Interaction of Cationic Carbosilane Dendrimers and Their siRNA Complexes with MCF-7 Cells Cultured in 3D Spheroids, Cells, 2022, 11, 1697 CrossRef PubMed.
  194. J. Conde, N. Oliva, M. Atilano, H. S. Song and N. Artzi, Self-assembled RNA-triple-helix hydrogel scaffold for microRNA modulation in the tumour microenvironment, Nat. Mater., 2016, 15, 353–363 CrossRef CAS PubMed.
  195. J. Shi, Y. Xu, X. Xu, X. Zhu, E. Pridgen, J. Wu, A. R. Votruba, A. Swami, B. R. Zetter and O. C. Farokhzad, Hybrid lipid–polymer nanoparticles for sustained siRNA delivery and gene silencing, Nanomedicine, 2014, 10, e897–e900 CrossRef PubMed.
  196. V. Ramakrishnan, B. Xu, J. Akers, T. Nguyen, J. Ma, S. Dhawan, J. Ning, Y. Mao, W. Hua, E. Kokkoli, F. Furnari, B. S. Carter and C. C. Chen, Radiation-induced extracellular vesicle (EV) release of miR-603 promotes IGF1-mediated stem cell state in glioblastomas, EBioMedicine, 2020, 55, 102736 CrossRef CAS PubMed.
  197. Q. Yang, Y. Zhou, J. Chen, N. Huang, Z. Wang and Y. Cheng, Gene Therapy for Drug-Resistant Glioblastoma via Lipid-Polymer Hybrid Nanoparticles Combined with Focused Ultrasound, Int. J. Nanomed., 2021, 16, 185–199 CrossRef PubMed.
  198. H. M. Ramos-Zaldívar, I. Polakovicova, E. Salas-Huenuleo, A. H. Corvalán, M. J. Kogan, C. P. Yefi and M. E. Andia, Extracellular vesicles through the blood-brain barrier: a review, Fluids Barriers CNS, 2022, 19, 60 CrossRef PubMed.
  199. S. Dubey, Z. Chen, Y. J. Jiang, A. Talis, A. Molotkov, A. Ali, A. Mintz and F. Momen-Heravi, Small extracellular vesicles (sEVs)-based gene delivery platform for cell-specific CRISPR/Cas9 genome editing, Theranostics, 2024, 14, 2777–2793 CrossRef CAS PubMed.
  200. H. Jo, K. Shim and D. Jeoung, Exosomes: Diagnostic and Therapeutic Implications in Cancer, Pharmaceutics, 2023, 15, 1465 CrossRef CAS PubMed.
  201. A. Meliani, F. Boisgerault, Z. Fitzpatrick, S. Marmier, C. Leborgne, F. Collaud, M. Simon Sola, S. Charles, G. Ronzitti, A. Vignaud, L. van Wittenberghe, B. Marolleau, F. Jouen, S. Tan, O. Boyer, O. Christophe, A. R. Brisson, C. A. Maguire and F. Mingozzi, Enhanced liver gene transfer and evasion of preexisting humoral immunity with exosome-enveloped AAV vectors, Blood Adv., 2017, 1, 2019–2031 CrossRef CAS PubMed.
  202. S. E. Ahmadi, M. Soleymani, F. Shahriyary, M. R. Amirzargar, M. Ofoghi, M. D. Fattahi and M. Safa, Viral vectors and extracellular vesicles: innate delivery systems utilized in CRISPR/Cas-mediated cancer therapy, Cancer Gene Ther., 2023, 30, 936–954 CrossRef CAS PubMed.
  203. N. Khan, S. Maurya, S. Bammidi and G. R. Jayandharan, AAV6 Vexosomes Mediate Robust Suicide Gene Delivery in a Murine Model of Hepatocellular Carcinoma, Mol. Ther. – Methods Clin. Dev., 2020, 17, 497–504 CrossRef CAS PubMed.
  204. J. Fares, A. U. Ahmed, I. V. Ulasov, A. M. Sonabend, J. Miska, C. Lee-Chang, I. V. Balyasnikova, J. P. Chandler, J. Portnow, M. C. Tate, P. Kumthekar, R. V. Lukas, S. A. Grimm, A. K. Adams, C. D. Hébert, T. V. Strong, C. Amidei, V. A. Arrieta, M. Zannikou, C. Horbinski, H. Zhang, K. B. Burdett, D. T. Curiel, S. Sachdev, K. S. Aboody, R. Stupp and M. S. Lesniak, Neural stem cell delivery of an oncolytic adenovirus in newly diagnosed malignant glioma: a first-in-human, phase 1, dose-escalation trial, Lancet Oncol., 2021, 22, 1103–1114 CrossRef CAS PubMed.

Footnote

Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d4nr05371k

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.