Fact-checked by Grok 2 weeks ago

Viral vector

A viral vector is a genetically modified virus engineered to deliver exogenous genetic material into target cells, leveraging the virus's natural infection machinery while rendering it replication-incompetent to prevent disease causation. Such vectors, derived from recombinant DNA techniques first demonstrated by Paul Berg in the early 1970s through the construction of hybrid viral-bacterial DNA molecules, enable precise gene transfer for therapeutic purposes without the full pathogenic potential of wild-type viruses. Prominent types include adenoviral vectors, valued for high transduction efficiency and large payload capacity but prone to strong immune clearance; adeno-associated viral (AAV) vectors, favored for long-term gene expression in non-dividing cells with lower immunogenicity; and lentiviral vectors, capable of stable genomic integration suitable for dividing cells like hematopoietic stem cells. Viral vectors underpin approved gene therapies addressing inherited disorders, such as AAV9-based Zolgensma for and AAV2 for retinal dystrophy in Luxturna, marking milestones in correcting monogenic defects via delivery. They also power viral vector vaccines, exemplified by adenoviral platforms in the Janssen () and AstraZeneca vaccines, which elicit robust cellular and by encoding pathogen antigens without viral replication. However, empirical data from clinical trials reveal inherent risks, including acute immune reactions, , and rare following high-dose AAV , alongside historical insertional oncogenesis from integrating vectors like early retrovirals in SCID trials, underscoring the need for vector optimization to balance efficacy with causal safety profiles.

Definition and Characteristics

Core Definition and Mechanism

A viral vector is a engineered to deliver exogenous genetic material into target cells, exploiting the virus's natural ability to cells and introduce s without causing productive or in the host. This modification typically involves removing viral genes responsible for replication and , replacing portions of the viral with a therapeutic under control of appropriate promoters. Viral vectors are classified based on their type ( or ) and integration capability, but all rely on the virus's evolved machinery for efficient cellular entry and gene transfer. The engineering process begins with selecting a parental virus, such as adenovirus or , whose genome is cloned into a backbone for manipulation. Essential structural and replication genes are deleted or separated into helper constructs to prevent replication outside controlled production systems, while the transgene cassette—containing the of interest, regulatory elements, and signals—is inserted. cell lines or transient systems provide the missing components, enabling assembly of non-replicative particles at titers often exceeding 10^12 genomes per milliliter for clinical-scale production. This results in high-titer, purified vectors capable of transducing specific cell types based on the parental virus's . In terms of mechanism, the vector initiates infection by binding host cell receptors, such as coxsackievirus and adenovirus receptor (CAR) for adenoviral vectors, triggering endocytosis or fusion for internalization. Endosomal escape follows, mediated by viral proteins that disrupt the endosomal membrane, allowing the capsid to release its genome into the cytoplasm. For DNA vectors, the genetic payload traffics to the nucleus via nuclear localization signals or passive diffusion through nuclear pores during cell division; RNA vectors like retroviral types reverse transcribe to DNA for nuclear import. Once in the nucleus, the transgene may persist episomally, as in adeno-associated virus (AAV) vectors, enabling long-term expression in non-dividing cells, or integrate into the host genome via viral integrase for stable inheritance in dividing cells. Expression occurs through host machinery transcribing the transgene into mRNA, which is exported and translated into functional protein, with efficacy depending on vector dose, target tissue accessibility, and immune evasion.

Physical and Biological Properties

![Icosahedral_Adenoviruses.jpg][float-right] Viral vectors possess distinct physical properties that influence their , , and efficiency. Most derive from enveloped or non-enveloped viruses with diameters typically ranging from 20 nm to 150 nm; for example, adenoviral vectors feature non-enveloped icosahedral of 80-100 nm, composed of 252 capsomers, while lentiviral vectors are enveloped retroviral particles approximately 100-120 nm in size. These structures confer varying stability profiles, with non-enveloped vectors like adenoviruses exhibiting greater resilience to environmental stresses but sensitivity to and fluctuations, necessitating with buffers and excipients to prevent capsid disassembly or aggregation. Enveloped vectors, such as lentiviruses, are more labile due to bilayers prone to disruption by detergents or freeze-thaw cycles, though cryoprotectants enhance long-term viability. Biologically, viral vectors are replication-incompetent through genetic modifications that excise essential replication genes, ensuring safety by limiting propagation beyond initial transduction while preserving entry and gene expression mechanisms. They exhibit engineered tropism via capsid or envelope proteins targeting specific cellular receptors, such as coxsackievirus-adenovirus receptor (CAR) for adenoviruses or CD4/CCR5 for lentiviruses, enabling broad or tissue-specific delivery. Genome capacities vary: adenoviral vectors accommodate up to 36 kb of DNA as episomal elements, lentivirals integrate ~8-10 kb into host chromatin for stable expression in dividing and non-dividing cells, and adeno-associated vectors (AAVs) carry ~4.7 kb ssDNA persisting extrachromosomally. Immunogenicity arises from viral proteins triggering innate responses via pattern recognition receptors and adaptive immunity, particularly pronounced in adenoviral vectors due to pre-existing antibodies in 40-90% of populations, potentially reducing efficacy in repeat dosing but bolstering vaccine-induced T-cell responses. These properties underpin their utility, though off-target effects and insertional mutagenesis risks, mitigated by self-inactivating designs, demand rigorous preclinical assessment.

Types of Viral Vectors

Retroviral and Lentiviral Vectors

Retroviral vectors are derived from retroviruses, such as (MLV), and consist of a modified single-stranded genome that is reverse-transcribed into double-stranded DNA upon entry into host cells, followed by integration into the host via viral integrase. This integration enables stable, long-term expression of the therapeutic , theoretically requiring only a single administration for persistent effects. They offer a packaging capacity of approximately 8-9 , suitable for many therapeutic genes, and have been foundational in early efforts due to efficient in dividing cells. However, retroviral vectors are limited to transducing actively dividing cells, as their pre-integration complex cannot efficiently cross the intact nuclear membrane in non-dividing cells, restricting applications in tissues like neurons or quiescent hematopoietic stem cells. A major drawback is the risk of , where random integration near proto-oncogenes can activate them, leading to malignancies; this was evidenced in clinical trials for (SCID-X1), where two patients developed T-cell due to LMO2 proto-oncogene activation. To mitigate replication-competent production, vectors employ split-genome packaging systems separating gag-pol, env, and vector components across multiple plasmids. Lentiviral vectors, a subclass of retroviral vectors primarily based on HIV-1, address key limitations by enabling of both dividing and non-dividing cells through active nuclear import mediated by viral accessory proteins like and Vpr. They maintain a slightly larger packaging capacity of around 9 and are often pseudotyped with vesicular stomatitis virus (VSV-G) for enhanced , broad , and concentrated production yields. Unlike simple retroviruses, lentiviruses integrate preferentially into active transcription units, potentially reducing but not eliminating risks. Safety has evolved across generations: first-generation systems retained significant sequences including accessory genes, increasing recombination risks; second-generation removed vif, vpr, vpu, and nef; third-generation further split packaging into four or more plasmids, eliminating tat and incorporating self-inactivating (SIN) long terminal repeats (LTRs) that abolish promoter activity post-integration, minimizing insertional activation. These designs reduce the probability of generating replication-competent (RCL) to below detectable levels in preclinical assays, enabling over 100 clinical trials by with no RCL detections. Third-generation lentiviral vectors have thus become preferred for , as in approved treatments for beta-thalassemia and cerebral , balancing efficacy with reduced oncogenic potential compared to earlier retroviral systems.

Adenoviral Vectors

Adenoviral vectors are engineered derivatives of adenoviruses, a family of non-enveloped, icosahedral viruses characterized by a linear double-stranded DNA genome of approximately 36 kilobases. These vectors typically employ human adenovirus serotype 5 (Ad5) or chimpanzee-derived adenoviruses like ChAdOx1 to evade pre-existing human immunity. The viral capsid, composed of 252 capsomeres including hexons, pentons, and fiber proteins, facilitates broad cell tropism via receptor interactions such as coxsackievirus-adenovirus receptor (CAR) and integrins. To render them replication-deficient, essential early genes like E1 (encoding proteins for viral DNA replication) and often E3 (modulating host immune responses) are deleted, with therapeutic transgenes inserted in their place, allowing packaging of up to 8 kb of foreign DNA in first-generation vectors or over 30 kb in helper-dependent "gutless" variants lacking all viral coding sequences. Upon transduction, the vector genome persists episomally in the nucleus, enabling high-level but transient transgene expression without genomic integration, which reduces risks of insertional mutagenesis compared to integrating vectors. Production occurs at high titers (10^12-10^13 viral particles per liter) in complementing cell lines like HEK293, which supply the deleted E1 functions. Adenoviral vectors excel in delivering robust expression across dividing and non-dividing cells due to efficient nuclear entry and strong promoters, making them suitable for applications requiring potent . Their advantages include large capacity, ease of , and inherent adjuvanticity, which promotes both humoral and cellular immunity—key for platforms. However, drawbacks predominate in long-term : strong innate and adaptive immune responses, including cytotoxic T-lymphocyte activation against transduced cells and vector capsids, limit durability of expression to weeks. Pre-existing neutralizing antibodies, prevalent in 40-90% of populations for common serotypes like Ad5, reduce and necessitate rarer serotypes or shielding strategies. Additionally, potential for liver and storms at high doses has constrained systemic use. In vaccine development, adenoviral vectors have proven effective, as seen in authorized products like the AstraZeneca-Oxford ChAdOx1-S (encoding spike protein), Ad26.COV2.S, and Russia's Sputnik V (heterologous Ad5/Ad26 prime-boost), which elicited protective immunity against in billions of doses administered globally by 2023. The replication-competent Ad26.ZEBOV/MVA-BN-Filo (Ervebo) , approved in 2019, demonstrated 97.5% efficacy against virus disease in a 2019-2020 trial. For , applications are niche, primarily oncolytic vectors like China's approved H101 (replication-conditional, targeting p53-deficient tumors) for , though first-generation non-replicating vectors faced setbacks, such as the 1999 trial fatality from inflammatory response. Ongoing trials explore gutless vectors for hereditary diseases, leveraging reduced for safer, prolonged expression.

Adeno-Associated Viral Vectors

Adeno-associated viral (AAV) vectors derive from adeno-associated virus, a non-enveloped, single-stranded DNA dependovirus in the Parvoviridae family with a genome of approximately 4.7 kilobases flanked by inverted terminal repeats (ITRs). The wild-type virus requires co-infection with a helper virus, such as adenovirus or herpesvirus, for replication, but recombinant AAV (rAAV) vectors used in applications are engineered to be replication-incompetent by removing viral rep and cap genes, which are supplied in trans via helper plasmids or viruses during production. These vectors package transgenes up to about 4.7 kb efficiently, though capacities up to 6 kb are possible with reduced yields and increased genome instability. AAV exhibits low pathogenicity in humans, with no associated diseases, making it suitable for therapeutic delivery. Over 170 AAV serotypes have been identified, primarily from tissues, with variations dictating tissue and efficiency. AAV2, the first serotype isolated in 1965 as a contaminant in adenovirus preparations, transduces dividing and non-dividing cells but primarily targets muscle, liver, and ; it was used in the earliest clinical trials due to its established receptor interactions. AAV9 demonstrates broad , including efficient via blood-brain barrier crossing in neonates, while AAV8 favors hepatocytes for liver-directed therapies. engineering, such as through , has yielded variants like AAV-PHP.eB for enhanced neuronal targeting in , though translation to s remains limited by species-specific differences. In transduction, rAAV enters cells via , traffics to the , and converts its single-stranded to double-stranded DNA, which persists episomally in non-dividing cells for years, enabling stable expression without integration into the host in most cases. occurs rarely at AAVS1 on , posing minimal oncogenic risk compared to retroviral s. However, innate immune activation via and pattern recognition of vector components can trigger , while adaptive responses, including pre-existing neutralizing antibodies (NAbs) prevalent in 30-80% of adults depending on serotype exposure history, reduce and preclude redosing. AAV vectors dominate gene therapy applications due to their safety profile and durability, with eight FDA approvals as of 2023, including Luxturna (, AAV2 delivering for , approved December 19, 2017) and Zolgensma (, AAV9 delivering for type 1, approved May 24, 2019). Limitations include transgene size constraints, which exclude larger genes like for , and manufacturing scalability issues, as high-titer production requires transient of HEK293 cells with yields of 10^14-10^15 vector genomes per batch. Ongoing challenges involve optimization to evade NAbs and dual-vector strategies for oversized payloads.

Other Vectors (Herpesviral, Poxviral, and Non-Mammalian)

Herpesviral vectors, predominantly based on herpes simplex virus type 1 (HSV-1), exploit the virus's natural neurotropism and large genome to deliver transgenes, particularly for central nervous system targeting. These vectors are engineered as replication-defective forms, where essential viral genes are deleted to prevent replication while retaining the ability to infect and express foreign genes in non-dividing cells. HSV-1 vectors support insert sizes exceeding 100 kb, far surpassing many other systems, enabling delivery of large therapeutic cassettes such as whole genomic loci or multiple genes. Applications include gene therapy for neurodegenerative diseases like Parkinson's, where HSV vectors have demonstrated efficient transduction of neurons in preclinical models, and oncolytic variants for tumor lysis in cancers such as melanoma, achieving objective response rates of up to 24.6% in checkpoint-refractory cases. Limitations include potential immunogenicity and cytotoxicity from residual viral proteins, though next-generation designs minimize these by eliminating all viral gene expression. Poxviral vectors, derived from viruses like , (MVA), and , are DNA-based systems valued for their cytoplasmic replication, which avoids genomic integration risks, and capacity for inserts up to 25 . These vectors induce robust cellular and humoral immune responses, making them suitable for platforms; for instance, recombinant expressing antigens has been used since 1982 for studies. MVA, attenuated to prevent replication in mammalian cells, enhances safety and is employed in prime-boost regimens to amplify antigen , as seen in and trials. In , poxvirus vectors deliver tumor-associated antigens alongside cytokines, showing prolonged survival in preclinical models when combined with checkpoint inhibitors. Drawbacks include pre-existing immunity from historical vaccination, which can reduce efficacy, though avian poxviruses like mitigate this via lack of . Non-mammalian viral vectors, such as from hosts, provide a safe alternative for mammalian since they do not replicate in cells, eliminating risks of uncontrolled spread. (AcMNPV), the most studied, transduces a broad range of mammalian cell types via glycoproteins like GP64 binding to receptors, achieving suitable for production or short-term . With insert capacities over 100 kb, baculovirus vectors have been applied in preclinical for liver diseases and cancer, displaying low and toxicity compared to mammalian viruses. Challenges include inefficient nuclear entry leading to episomal persistence rather than integration, limiting long-term expression, though pseudotyping with vesicular virus improves efficiency. These vectors also facilitate scalable manufacturing in cells, supporting their use in approaches.

Applications

Basic and Preclinical Research

Viral vectors enable precise genetic manipulation in cellular and animal models, facilitating investigations into function, protein interactions, and signaling pathways central to . Lentiviral vectors, derived from HIV-1, integrate transgenes into the host for stable, heritable expression in proliferating cells, commonly used to generate or overexpression lines via shRNA or cDNA delivery, respectively. Adeno-associated viral (AAV) vectors, conversely, persist episomally in non-dividing cells like neurons, supporting long-term expression without insertional risks, as evidenced by their application in over 170 studies by 2019 for and circuit mapping. In , AAV serotypes such as AAV9 achieve widespread across regions in and , revealing mechanisms and connectivity complexities unattainable with chemical tracers alone. Lentiviral vectors complement this by enabling in stem cell-derived models, dissecting developmental pathways with efficiencies exceeding 90% in iPSCs. These tools have advanced understanding of mechanisms, such as in Parkinson's models where AAV-delivered alpha-synuclein aggregates recapitulate pathology. Preclinical research employs viral vectors to assess therapeutic candidates , evaluating efficacy, biodistribution, and in disease-specific animal models before human trials. AAV vectors, for example, have demonstrated sustained expression up to 10 years in , guiding hemophilia B dosing at 2×10^12 vg/kg for restoration achieving 10-30% normal levels. Lentiviral vectors in models correct expression in mdx mice, improving muscle function by 50-70% and informing scalable production needs. Such studies highlight vector variations, with AAV8 optimizing liver targeting in models, while underscoring challenges like pre-existing immunity affecting 30-50% of adults. These findings from , , and models validate causal links between and phenotypic correction, prioritizing vectors with minimal off-target effects for clinical progression.

Gene Therapy

Viral vectors serve as primary delivery vehicles in gene therapy, transporting therapeutic transgenes into patient cells to correct genetic deficiencies or express functional proteins. Adeno-associated virus (AAV) vectors predominate for in vivo applications due to their low immunogenicity, ability to transduce non-dividing cells, and capacity for long-term episomal gene expression, while lentiviral vectors excel in ex vivo modification of hematopoietic stem cells for stable integration. By 2023, over 20 FDA-approved gene therapies utilized viral vectors, targeting monogenic disorders such as spinal muscular atrophy (SMA), hemophilia, and inherited blindness. Early clinical trials in the 1990s employed retroviral vectors for gene correction in (ADA)-deficient (SCID), achieving immune reconstitution in some patients but revealing risks. Subsequent advancements shifted toward AAV vectors; Luxturna (), approved by the FDA in December 2017, uses AAV2 to deliver the gene to retinal cells, restoring vision in patients with caused by biallelic mutations, with clinical trials demonstrating improved multi-luminance mobility testing scores. For , Zolgensma (), an AAV9-based therapy approved in May 2019, delivers the gene via intravenous infusion, enabling survival and motor milestone achievement in infants with type 1, as evidenced by phase 3 trials showing 100% event-free survival at 14 months versus 26% in controls. Lentiviral vectors have succeeded in ex vivo therapies for blood disorders; Zynteglo (), approved in August 2022, integrates a functional β-globin into autologous hematopoietic cells for transfusion-dependent β-thalassemia, with pivotal trials reporting transfusion independence in 31 of 42 patients after a median 3.8 years. Hemgenix (etranacogene dezaparvovec), an AAV5 vector approved in November 2022 for hemophilia B, expresses , reducing annualized bleeding rates by 54% in phase 3 studies compared to prior prophylaxis. These outcomes underscore viral vectors' efficacy in achieving durable phenotypic correction, though scalability and vector dosing limitations persist. Ongoing trials expand applications to disorders, leveraging AAV's neurotropism; for instance, AAV9 vectors target neurons for , with preclinical data showing widespread CNS . Despite historical setbacks like the 1999 adenovirus-related death in an deficiency trial, refined vector designs have minimized acute toxicities, enabling broader adoption. As of 2024, viral vector gene therapies treat over a dozen rare diseases, with cumulative evidence from thousands of patients affirming their transformative potential when expression aligns with disease pathology.

Vaccine Development


Viral vector vaccines are engineered by inserting genetic sequences encoding pathogen antigens into replication-deficient viruses, enabling host cells to produce the antigens and trigger both humoral and cellular immune responses. This approach leverages the virus's natural ability to infect cells and stimulate immunity while minimizing disease risk through genetic modifications that prevent replication. Development begins with vector selection, such as adenoviruses or poxviruses, followed by transgene insertion, preclinical efficacy testing in animal models, and phased clinical trials assessing safety, immunogenicity, and protection.
The foundational milestone occurred in 1982 when researchers inserted the surface antigen gene into , demonstrating proof-of-concept for antigen expression . Subsequent progress focused on adenoviral vectors, with early trials in the for and , though pre-existing immunity to common serotypes like Ad5 posed challenges, leading to strategies like using rare or chimpanzee-derived adenoviruses. The Ervebo Ebola vaccine, utilizing a vesicular vector expressing Ebola , marked the first licensed in 2019, approved by the on November 1, 2019, after demonstrating 97.5% efficacy in a 2014-2016 outbreak ring trial involving 3,000 participants. The accelerated adenoviral vector deployment, with the AstraZeneca-Oxford (ChAdOx1 nCoV-19), based on a adenovirus expressing , receiving emergency authorization in the UK on December 30, 2020, following Phase III showing 70.4% against symptomatic in over 23,000 participants. Similarly, the Janssen (Ad26.COV2.S), using human adenovirus type 26, was authorized by the FDA on February 27, 2021, with 66% against moderate to severe in a of approximately 44,000 individuals. Russia's Sputnik V, combining Ad26 and Ad5 vectors in a prime-boost regimen, reported 91.6% in a 2021 Lancet-published of 19,866 participants. Advantages include potent T-cell responses mimicking natural infection, enabling protection against diverse pathogens like viruses requiring cellular immunity, unlike subunit . However, drawbacks encompass vector-specific immunity reducing efficacy in repeat dosing and complex manufacturing requiring 2 facilities. Ongoing innovations involve modifications to evade immunity and prime-boost schedules to enhance responses, as seen in and COVID regimens.

Oncolytic and Other Therapeutic Uses

Oncolytic virotherapy employs genetically modified viral vectors designed to selectively replicate within and destroy cancer cells while sparing healthy tissue, often eliciting an antitumor immune response. These vectors exploit molecular defects common in malignancies, such as impaired interferon signaling or dysregulated cell cycle control, enabling preferential tumor tropism. Attenuation mutations reduce pathogenicity in normal cells, while transgenes like granulocyte-macrophage colony-stimulating factor (GM-CSF) enhance immunogenicity by promoting dendritic cell activation and T-cell infiltration. Talimogene laherparepvec (T-VEC), an oncolytic type 1 (HSV-1) vector, exemplifies this approach. It features deletions in ICP34.5 genes to limit replication to cells with defective R (PKR) pathways, prevalent in cancers, and insertion of the human GM-CSF gene to boost local immunity. Administered intratumorally, T-VEC lyses injected lesions, releases tumor antigens, and induces abscopal effects on distant metastases via systemic T-cell responses. The phase III OPTiM trial (NCT00763608), involving 436 patients with advanced , demonstrated a 26.4% durable response rate (≥6 months) for T-VEC versus 2.1% for GM-CSF alone, leading to FDA approval on October 27, 2015, for unresectable cutaneous, subcutaneous, or nodal post-surgery. approval followed in December 2015. Real-world data from over 1,000 patients confirm a favorable safety profile, with flu-like symptoms and injection-site reactions as primary adverse events, though diminishes in visceral disease. Other oncolytic vectors include adenoviral platforms like H101 (recombinant adenovirus type 5), approved by China's in November 2005 for head and neck refractory to , following a phase III trial showing 78.8% response rate in combination with cisplatin/5-fluorouracil versus 39.6% for alone. Pexastimogene devacirepvec (JX-594, modified virus) expresses GM-CSF and for selective replication in EGFR/RAS pathway-dysregulated tumors; phase II trials reported median survival of 14.1 months in hepatocellular carcinoma patients versus 6.7 months on placebo, though phase III results remain pending. Reovirus (Reolysin) and Newcastle disease virus have advanced to phase II/III trials, often combined with checkpoint inhibitors like , yielding objective response rates up to 36% in refractory solid tumors. As of 2023, over 100 trials are registered on , predominantly phase I/II, highlighting immune evasion challenges and the need for combinatorial strategies. Beyond oncolysis, viral vectors enable suicide gene therapies, where delivered transgenes convert non-toxic prodrugs into cytotoxins, amplifying cell death in targeted tissues. For instance, retroviral or adenoviral vectors encoding thymidine kinase (HSV-TK) facilitate activation, restricting replication-competent viruses to proliferative cells like tumors or vascular lesions. Clinical applications include treatment, with phase III data showing prolonged survival when combined with radiotherapy. In cardiovascular therapy, adenovirus-mediated HSV-TK/ has inhibited intimal in vein grafts, reducing restenosis rates by 50-70% in preclinical models translated to early human trials. These approaches, distinct from stable transgene expression in , leverage transient for localized cytotoxicity, though limits repeat dosing.

Production and Manufacturing

Vector Design and Construction

Viral vectors are engineered by modifying the genetic backbone of wild-type viruses to eliminate replication capacity and pathogenicity while incorporating a therapeutic . This involves selecting a or strain suited to the application's requirements, such as tissue , payload capacity, and persistence— for instance, (AAV) for episomal expression with low , lentiviral vectors for genomic , or adenoviral vectors for high-capacity transient delivery. The core design principle is to retain essential cis-acting elements like inverted terminal repeats (ITRs) in AAV or long terminal repeats (LTRs) in lentiviruses for packaging and , while excising trans-acting genes (e.g., rep/cap in AAV, gag/pol/env in lentiviruses) that drive replication. The transgene cassette is cloned into a transfer plasmid, typically comprising a strong or tissue-specific promoter (e.g., CMV for broad expression or synapsin for neurons), the codon-optimized coding sequence, polyadenylation signals, and optional elements like woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) for enhanced mRNA stability or insulators to prevent silencing. Payload limits dictate design: AAV accommodates ~4.7 kb (or ~2.4 kb for self-complementary variants using mutated ITRs), lentiviruses up to 9-10 kb, and gutless adenoviruses ~36 kb by retaining only ITRs and packaging signals. To mitigate immunogenicity, sequences are depleted of CpG motifs, and microRNA binding sites (e.g., miR-142) are inserted to evade innate immune detection in antigen-presenting cells. Construction employs plasmid-based recombination to generate replication-deficient vectors, avoiding direct manipulation of infectious . For AAV, triple into HEK293 cells uses the transfer , a rep/cap , and an adenoviral helper providing E1/E4/VP proteins; assembles the . Lentiviral vectors utilize a four- system: transfer with self-inactivating 3' LTR, packaging (gag/pol), rev-expressing , and envelope (e.g., VSV-G pseudotype for broad ), transfected into HEK293T cells. Adenoviral vectors, particularly helper-dependent "gutless" designs, involve co- of the minimal with a helper adenovirus flanked by loxP sites, followed by excision to separate from helper particles. Advanced engineering refines targeting and safety: shuffling or creates novel (e.g., AAV2.7m8 for muscle), while fiber knob modifications in adenoviruses (e.g., RGD-4C insertion) enhance receptor binding. Dual-vector strategies split oversized transgenes for AAV, relying on ITR-mediated recombination or inteins for reconstitution. These modular approaches ensure scalability and compliance with good manufacturing practices, though challenges like pre-existing immunity necessitate switching or chimeric designs.

Scalable Production Methods

Scalable production of vectors primarily employs mammalian or cell lines cultured in to achieve high titers suitable for clinical and commercial applications. Key methods include transient with helper plasmids or viruses in suspension-adapted , such as HEK293 for adenoviral and adeno-associated (AAV) vectors, enabling scale-up from shake flasks to stirred-tank of 25–200 liters or more. Stable producer lines, incorporating integrated vector genomes and helper elements, offer reproducibility for large-scale manufacturing but require extensive validation for genomic stability. systems using baculovirus expression vectors (BEVS) provide an alternative for AAV serotypes, supporting scalability in volumes up to 1,000 liters while avoiding human immunogenicity concerns. For adenoviral vectors, optimized processes in HEK293 suspension cultures utilize perfusion or fed-batch modes in fixed-bed bioreactors like the iCELLis system, yielding up to 10^14 viral particles per batch in GMP settings. These methods incorporate design-of-experiments for media optimization, achieving functional titers exceeding 10^12 infectious units per milliliter post-harvest. Lentiviral vectors follow similar transient triple-plasmid in HEK293-derived suspension lines, with bioreactors enabling titers 10-fold higher than adherent methods through cell retention and intensified feeding strategies. AAV production has advanced with HEK293 triple-transfection protocols scaled to 50-liter bioreactors, delivering vector genome titers of 10^14–10^15 vg/L via in situ cell lysis and downstream-compatible harvests. BEVS in Sf9 cells offers higher volumetric productivity for certain capsids, with recent optimizations reaching 10^15 vg/L in wave or stirred bioreactors, though empty/full capsid ratios demand rigorous analytics. Challenges persist in yield consistency across serotypes, prompting hybrid platforms and continuous manufacturing to meet surging gene therapy demands projected at over 1 million doses annually by 2030.

Quality Control and Purification

Purification of viral vectors involves downstream processing to separate the target vector particles from host cell components, media, and production byproducts following harvest from cell cultures. Common methods include tangential flow filtration for initial clarification and concentration, followed by chromatography techniques such as anion-exchange, cation-exchange, or affinity chromatography to capture and polish the vectors based on charge, size, or specific ligands. For adeno-associated virus (AAV) vectors, affinity resins targeting capsid proteins enable high specificity, while density gradient ultracentrifugation, though effective for research-scale purity, is limited by poor scalability and is largely replaced by chromatographic approaches in manufacturing. Lentiviral and adenoviral vectors often require additional steps to remove envelope glycoproteins or non-infectious particles, with ultrafiltration/diafiltration used for buffer exchange and final formulation to achieve concentrations suitable for clinical dosing, typically exceeding 10^12 vector genomes per milliliter. These processes must minimize aggregation and loss of infectivity, with yields varying from 20-50% depending on vector type and scale. Quality control (QC) for viral vectors encompasses assays to verify identity, purity, potency, quantity, and safety, ensuring compliance with regulatory standards such as those outlined by the FDA for chemistry, manufacturing, and controls (CMC) in gene therapy products. Identity confirmation typically involves PCR-based sequencing of the transgene cassette or serotype-specific ELISA to distinguish vector variants, critical for preventing mix-ups in multi-product facilities. Purity assessment quantifies contaminants like host cell proteins (via ELISA, targeting <100 ng per dose), residual DNA (<10 ng per dose), and empty capsids (via analytical ultracentrifugation or capillary electrophoresis, aiming for >50% full capsids in AAV preparations). Quantity is measured by quantitative PCR for genome titers (vg/mL) and infectious titers (TU/mL), with ratios ideally between 10:1 and 100:1 to indicate vector functionality. Potency testing evaluates through assays in relevant cell lines, measuring expression via qPCR or reporter , while safety checks include sterility (USP <71>), endotoxin levels (<5 EU/mL by LAL assay), and adventitious agent screening via next-generation sequencing or panels for viruses, , and . For GMP production, these assays are phased, with full characterization required for master viral banks and lot release, addressing risks like immunogenic impurities that could trigger adverse immune responses in patients. Regulatory guidance emphasizes to demonstrate consistency, with analytics evolving to include for profiling and flow virometry for particle enumeration. Challenges persist in standardizing assays across vector types, as enveloped vectors like lentiviruses may co-purify extracellular vesicles, potentially confounding purity metrics.

Risks and Safety Concerns

Immunogenicity and Immune Evasion

Viral vectors provoke both innate and adaptive immune responses that can compromise therapeutic efficacy and safety. Innate immunity detects viral proteins via receptors such as Toll-like receptors (TLRs), triggering release, , and rapid vector clearance; for (AAV) vectors, TLR9 activation by single-stranded DNA genomes exacerbates this, leading to dose-dependent observed in high-dose clinical trials for hemophilia B. Adaptive responses include neutralizing antibodies (NAbs) against capsids, prevalent in 30-80% of humans due to prior natural infections, which block vector and exclude seropositive patients from AAV trials, as seen in Luxturna approvals requiring NAb screening. Cellular immunity, via CD8+ T cells targeting transduced cells, causes loss, with capsid-specific T cells correlating to liver toxicity in trials like those for using AAV9-Zolgensma. Pre-existing immunity particularly hampers adenoviral vectors, where 5 (Ad5) NAbs in over 90% of adults reduce expression by up to 100-fold in preclinical models and diminish in HIV trials, prompting shifts to rarer serotypes like Ad26 or Ad35. In lentiviral vectors, pseudotyped with envelopes like VSV-G, humoral responses are lower but T cell-mediated clearance persists, limiting redosing; from CAR-T therapies show vector-specific immunity reducing persistence. These responses not only curtail efficacy but pose risks, including acute or delayed , as in the 1999 AAV trial fatality from inflammatory cytokines. To evade immunity, strategies focus on vector engineering and adjunct therapies. Capsid modifications, such as AAV shuffling or rational to alter surface epitopes, reduce NAb binding by 10-100 fold and enhance in seropositive models; for instance, AAV2 variants evade 70% of sera NAbs. Rare serotypes (e.g., AAV8, AAV9) or nonhuman primate-derived minimize , enabling higher liver in trials despite 20-40% seroprevalence. Immune stealth approaches include peptide insertion for shielding or empty/full ratios to decoy antibodies, improving durability in nonhuman primates. Pharmacological evasion employs transient , like corticosteroids or rituximab, to suppress T cell responses during vector delivery, boosting expression 5-10 fold in preclinical hemophilia models but risking infections. For redosing, vectors (e.g., switching AAV serotypes) or non-viral chimeras mitigate adaptive memory, though clinical translation remains limited by incomplete evasion. Ongoing innovations, such as TLR antagonists or deimmunization via editing, aim to balance reduction with efficiency, informed by failures in early trials.

Insertional Mutagenesis and Oncogenic Risks

Insertional mutagenesis refers to the integration of viral vector DNA into the host cell genome, which can disrupt normal gene function or activate proto-oncogenes, potentially leading to oncogenic transformation. This risk is most pronounced with integrating vectors such as gamma-retroviral and lentiviral systems, where random insertion sites may preferentially target transcriptionally active regions like promoters or enhancers. In contrast, non-integrating vectors like adeno-associated virus (AAV) primarily persist as episomes, though rare integration events have been observed, particularly at hotspots such as the AAVS1 locus on chromosome 19. Early clinical trials of gamma-retroviral vectors for (SCID-X1) demonstrated this hazard concretely; between 2002 and 2006, five of 20 treated patients developed due to vector insertions near the LMO2 proto-oncogene, which aberrantly activated its expression and cooperated with secondary somatic mutations. Insertional activation of oncogenes like LMO2, CCND2, and BCL2 was confirmed through genomic analysis, halting the trial and prompting vector redesigns with self-inactivating (SIN) long terminal repeats to minimize enhancer-driven dysregulation. By 2008, four additional cases of insertional oncogenesis were reported in retroviral for SCID, underscoring the vectors' bias toward integrating near hematopoietic genes. Lentiviral vectors, derived from HIV-1, exhibit a safer profile, favoring bodies over promoters and reducing transformation risk by approximately 10-fold compared to gamma-retroviral vectors in preclinical models. lentiviral designs further mitigate , as evidenced by lower oncogenic potential in comparative studies. However, recent data from lentiviral-based therapies for cerebral , such as Skysona (elivaldogene autotemcel), reported seven cases of hematological malignancies as of 2025, prompting reevaluation of long-term risks despite no definitive vector causation in all instances. The Medicines Agency's 2013 reflection paper documented serious adverse events from in 12 patients across trials, emphasizing vigilant monitoring. For AAV vectors, remains theoretical and low-probability in humans, with mouse studies showing elevated incidence only at supraphysiological doses exceeding clinical relevance. Regulatory assessments, including those from 2022 onward, conclude that AAV does not clearly elevate tumorigenesis in standard applications, though high-dose hepatic trials warrant caution. strategies across vector types now include insulator elements, genome-wide insertion site mapping via high-throughput sequencing, and preclinical assays to predict and avert oncogenic hotspots. Despite advances, long-term follow-up in over 20 approved gene therapies as of 2025 reveals no new on zero , with calls for enhanced biodistribution and clonal tracking to address residual uncertainties.

Off-Target Effects and Long-Term Toxicity

Off-target effects in viral vector applications refer to unintended interactions, such as transduction of non-target cells or tissues, leading to ectopic gene expression or genomic alterations outside the intended site. In adenoviral and adeno-associated virus (AAV) vectors, off-target delivery can occur due to systemic administration, resulting in transgene expression in organs like the liver or spleen, which may provoke localized toxicity or immune activation. For gene editing payloads delivered via viral vectors, such as CRISPR-Cas9 in lentiviral systems, off-target cleavage at homologous genomic sites has been documented, potentially causing unintended mutations with frequencies varying by guide RNA design and Cas9 variant, though high-fidelity variants reduce this to below 1% in some assays. These effects are assessed through methods like GUIDE-seq or CIRCLE-seq, which detect double-strand breaks at non-canonical sites. Long-term toxicity concerns primarily stem from insertional mutagenesis in integrating vectors like lentiviral and gammaretroviral systems, where proviral DNA insertion near proto-oncogenes can disrupt tumor suppressors or activate oncogenes, as evidenced by leukemia cases in early severe combined immunodeficiency (SCID) gene therapy trials using gammaretroviral vectors between 2002 and 2006, affecting 5 of 20 patients. Self-inactivating (SIN) lentiviral vectors mitigate this risk by reducing enhancer activity, with preclinical models showing lower genotoxicity compared to gammaretrovirals; clinical follow-up of 783 patients treated with lentiviral-modified T cells over more than 2,200 patient-years reported no vector-related malignancies as of 2024. Nonetheless, theoretical risks persist due to preferred integration near transcription start sites, necessitating long-term monitoring protocols that extend up to 15 years post-treatment per FDA guidelines. For non-integrating AAV vectors, long-term risks include rare genomic integration events, estimated at less than 0.1% of transduced cells , potentially leading to delayed oncogenic , though large-scale from over 255 trials indicate primarily acute toxicities like at doses exceeding 10^13 vg/kg rather than chronic . High-dose AAV administration has been linked to fatal complement-mediated toxicities and in recent trials, such as those for cardiac diseases in 2023-2024, highlighting dose-dependent innate immune responses as a causal factor over insertional risks. Persistent episomal AAV genomes can also drive chronic overexpression, contributing to off-target toxicities like degeneration in ocular applications observed in studies. Overall, while empirical data from approved therapies like Zolgensma (AAV9 for , approved 2019) show durable efficacy with manageable risks up to 5 years post-infusion, assays and extended surveillance remain essential to quantify rare long-term events.

History and Milestones

Origins and Early Development (1970s–1980s)

The origins of viral vectors trace back to early attempts at leveraging viruses for gene transfer, predating formal recombinant DNA techniques. In 1970, Stanfield Rogers administered Shope papilloma virus to two young sisters with argininemia, a disorder caused by arginase deficiency, based on observations that the virus could induce arginase activity in infected cells, marking the first documented use of a virus as a potential therapeutic gene carrier. However, the treatment failed to produce sustained enzyme activity and drew criticism for lacking ethical oversight and preclinical validation, highlighting early risks of uncontrolled viral delivery. This experiment underscored viruses' natural capacity to introduce genetic material but did not involve genetic engineering. Advancements accelerated with recombinant DNA technology in 1972, when Paul Berg's laboratory constructed the first chimeric DNA molecule by ligating SV40 viral DNA—a simian polyomavirus known for its transforming potential—with lambda phage DNA using EcoRI restriction enzyme and DNA ligase, enabling in vitro propagation of hybrid viral genomes. This SV40-based construct demonstrated the feasibility of engineering viral DNA for foreign gene insertion, laying foundational principles for viral vectors despite concerns over oncogenicity from SV40's tumor-inducing properties. Berg's work, published in Proceedings of the National Academy of Sciences, shifted focus from natural viral infection to deliberate genetic modification, though initial applications remained limited to bacterial and viral propagation rather than mammalian gene delivery. These innovations prompted safety deliberations, culminating in the 1975 Molecules, convened by and others at the to assess biohazards, particularly from recombinant viral DNAs that could create novel pathogens or enhance transmissibility. The conference recommended a voluntary moratorium on certain experiments, including tumor virus DNA in prokaryotes or linking viral genomes to non-viral vectors, establishing physical and biological guidelines that influenced NIH policies and slowed but did not halt vector development. Attendees emphasized risk proportionality, prohibiting high-risk viral recombinants until safer protocols emerged, reflecting early recognition of and uncontrolled replication as causal threats. By the late 1970s, researchers exploited viral genomes for efficient gene transfer into cultured mammalian cells, with retroviruses gaining attention after Harold Varmus and Michael Bishop's 1970s discoveries that these viruses could stably integrate cellular oncogenes via reverse transcription, inspiring engineered vectors for heritable gene insertion. Initial retroviral constructs, derived from murine leukemia viruses, focused on packaging foreign DNA into viral particles without replication competence, though packaging cell lines were not refined until the early 1980s. Concurrently, adenovirus studies advanced, with early 1980s designs at Cold Spring Harbor Laboratory producing replication-defective vectors by deleting essential early region genes (E1), enabling transient gene expression in non-dividing cells and setting the stage for in vivo applications. These efforts prioritized empirical containment over speculative safety, prioritizing vectors' natural tropism while mitigating pathogenicity through genetic deletions.

Initial Clinical Trials and Major Setbacks (1990s–2000s)

The first approved utilizing a viral vector commenced in 1990, targeting (ADA) deficiency, a form of (SCID). This approach employed a retroviral vector to autologous T cells with the human ADA cDNA, which were then reinfused into the patient, a four-year-old girl named Ashanthi DeSilva. Initial results demonstrated partial restoration of ADA enzyme activity and immune function, though long-term efficacy required ongoing enzyme replacement therapy, marking a proof-of-concept rather than a definitive cure. Throughout the , retroviral vectors dominated early trials for monogenic disorders, with over 100 protocols approved by 1999, focusing on conditions like and hemophilia; however, efficiencies remained low, and sustained proved challenging due to vector instability and host immune responses. Adenoviral vectors emerged in trials by 1993, prized for their high efficiency and capacity for larger transgenes, but preclinical data overlooked potent in humans. A pivotal setback occurred on September 17, 1999, when 18-year-old died during a phase I trial at the testing an E1/E3-deleted adenoviral vector for (OTC) deficiency. Gelsinger, who had partial OTC function, received a high-dose (3.8 × 10^13 particles) intraportal infusion, triggering a , , and multi-organ failure attributed to vector-induced innate immune activation rather than the itself. Investigations revealed protocol lapses, including undeclared animal toxicity data and conflicts of interest involving the principal , James Wilson, leading the FDA to suspend the trial, impose institutional holds, and halt many adenoviral programs nationwide. This incident, the first confirmed fatality, eroded public trust and prompted stricter oversight, effectively stalling systemic vector delivery approaches for years. Further complications arose in early 2000s trials for X-linked SCID (SCID-X1) using gamma-retroviral vectors targeting the IL2RG gene. and British studies reported immune reconstitution in most of 20 treated infants by 2000–2002, with vector-marked T cells persisting long-term, offering apparent cures without conditioning chemotherapy. However, by 2002–2003, five patients developed linked to : the strong viral enhancer activated the adjacent LMO2 proto-oncogene, cooperating with secondary mutations. This oncogenic risk, inherent to gamma-retroviruses' preference for integrating near transcription start sites, prompted trial suspensions in 2003 and a field-wide shift away from such vectors, underscoring the need for safer integrase designs and self-inactivating configurations.

Revival, Approvals, and Expansion (2010s–Present)


Following the setbacks of the preceding decades, viral vector technologies experienced a revival in the 2010s driven by engineering advancements that enhanced safety profiles, such as capsid modifications in adeno-associated viruses (AAV) to reduce immunogenicity and improve tissue specificity. These refinements addressed prior issues like insertional mutagenesis and immune responses, enabling progression to clinical success. By the mid-2010s, AAV vectors emerged as the dominant platform for in vivo gene delivery, with lentiviral and retroviral vectors supporting ex vivo applications like CAR-T cell therapies.
The approved Glybera, an AAV1-based therapy for , in October 2012 as the first product utilizing viral vectors, though it was withdrawn in 2017 due to limited efficacy and high cost. In the United States, the FDA granted approval to Luxturna (), an AAV2 vector for RPE65-mediated , on December 19, 2017, marking the first AAV for an inherited . This was followed by Zolgensma (onasemnogene abeparvovec), an AAV9 vector for , approved on May 24, 2019, demonstrating durable efficacy in infants. Additional approvals included Hemgenix (etranacogene dezaparvovec), an AAV5 vector for hemophilia B, in November 2022, reflecting growing regulatory confidence. Viral vectors also expanded into vaccine development, with the FDA approving Ervebo (rVSV-ZEBOV), a vesicular stomatitis virus-based vector for Ebola virus disease, on December 20, 2019. The COVID-19 pandemic accelerated adenoviral vector vaccines: Sputnik V (Ad26/Ad5) received emergency approval in Russia on August 11, 2020; AstraZeneca's ChAdOx1-S was authorized in the UK on December 30, 2020; and Johnson & Johnson's Ad26.COV2.S gained FDA emergency use authorization on February 27, 2021. At least six adenoviral vector-based COVID-19 vaccines achieved regulatory approval globally, leveraging prior platforms from Ebola research to enable rapid deployment and billions of doses administered. By 2022, eight viral vector-based gene therapies had received FDA approval, with the pace increasing to four in 2022, seven in 2023, and six in 2024, signaling robust expansion amid a burgeoning clinical pipeline exceeding hundreds of trials. This growth underscores viral vectors' versatility across monogenic diseases, , and infectious diseases, though manufacturing scalability remains a bottleneck for widespread adoption.

Regulatory and Ethical Considerations

Biosafety and Risk Assessment Protocols

Biosafety protocols for viral vectors are primarily governed by the NIH Guidelines for Research Involving Recombinant or Synthetic Molecules, which classify experiments based on potential risks such as replication competence, host range, and genetic inserts. These guidelines require institutional committees (IBCs) to conduct site-specific s, evaluating factors including vector tropism, potential for recombination, and environmental release risks before approving work. The CDC and NIH's in Microbiological and Biomedical Laboratories (BMBL, 6th edition, 2020) emphasizes a protocol-driven approach, integrating agent hazards, laboratory procedures, and personnel training to determine appropriate biosafety levels (BSL-1 to BSL-4). Risk assessments distinguish between replication-deficient and replication-competent vectors, with the former typically requiring lower if they lack factors or oncogenic inserts. For lentiviral vectors, derived from HIV-1, NIH recommends BSL-2 due to their Group 2 (RG2) classification, involving work in biosafety cabinets (BSCs), (PPE) like gloves and lab coats, and procedures; higher levels apply if pseudotyped for broad or containing hazardous transgenes. Adenoviral vectors, often RG1 or RG2, mandate BSL-2 practices including access restriction, medical surveillance, and spill response protocols, as they can cause respiratory illness despite attenuation. (AAV) vectors may qualify for BSL-1 if replication-incompetent, free of helper , and without toxic inserts, but default to BSL-2 for animal or high-titer work to mitigate aerosol transmission . Protocols extend to animal studies under Animal Biosafety Levels (ABSL-1 to ABSL-3), with monitoring post-administration; for instance, ABSL-2 is required for adenoviral use in to prevent . Waste inactivation via autoclaving or chemical disinfectants, storage at -70°C or below, and annual IBC reviews ensure containment integrity. For clinical applications, WHO and FDA-aligned assessments incorporate patient-specific risks like immune responses and long-term genomic integration, mandating (GMP) facilities with validated inactivation methods. These measures address empirical evidence of rare but documented incidents, such as inadvertent exposures in early trials, prioritizing causal pathways like escape over unsubstantiated fears.

Approval Processes and Global Variations

In the United States, the (FDA) regulates viral vector-based products, including gene therapies and vaccines, as biologics under the Center for Biologics Evaluation and Research (CBER). The approval pathway begins with an (IND) application, supported by preclinical data on vector safety, biodistribution, and , followed by phased clinical trials demonstrating efficacy and managing risks like and . Full approval requires a Biologics License Application (BLA) with substantial evidence from controlled trials, chemistry, manufacturing, and controls () data ensuring vector purity and replication incompetence, and commitments to long-term follow-up for at least 15 years to monitor delayed adverse events. The first FDA-approved viral vector gene therapy, (Luxturna, an AAV2 vector for RPE65-mediated retinal dystrophy), received accelerated approval on December 19, 2017, based on phase 3 trial data showing improved vision, though confirmatory studies were required. In the European Union, the European Medicines Agency (EMA) classifies viral vector products as advanced therapy medicinal products (ATMPs), requiring centralized marketing authorization with guidelines emphasizing vector design, non-clinical toxicology in relevant species, and clinical data on durability of transgene expression. Approvals mandate comprehensive quality controls for adventitious agents and genomic integration risks, plus post-authorization safety monitoring via the Pharmacovigilance Risk Assessment Committee. The EMA's first viral vector approval was alipogene tiparvovec (Glybera, an AAV1 vector for lipoprotein lipase deficiency) on October 25, 2012, granted after phase 3 evidence of triglyceride reduction, but it was withdrawn in 2017 due to commercial viability despite conditional efficacy. Recent approvals, such as those for AAV-based therapies, incorporate immunogenicity assessments tailored to vector capsid and seroprevalence. Global variations reflect differing priorities in data rigor, timelines, and manufacturing oversight. China approved the world's first commercial gene therapy, Gendicine (an adenoviral vector expressing p53 for head and neck cancer), on October 16, 2003, via the National Medical Products Administration (NMPA), based on phase 1-3 trials showing tumor response rates up to 64%, though long-term survival data were limited compared to Western standards. In Russia, the Ministry of Health granted full approval to Sputnik V (a human adenoviral vector COVID-19 vaccine) on August 11, 2020, after phase 1-2 interim data from 76 participants demonstrating immunogenicity, preceding phase 3 completion and sparking international scrutiny over expedited processes without emergency use restrictions. Such approaches contrast with FDA and EMA requirements for larger phase 3 cohorts and manufacturing consistency, as evidenced by the World Health Organization's 2021 suspension of Sputnik V's emergency listing review due to Good Manufacturing Practice deficiencies at certain facilities. In regions like India and Brazil, conditional approvals for viral vector vaccines during the COVID-19 pandemic often relied on bridging studies to foreign data, highlighting tensions between rapid access and harmonized safety evaluations under frameworks like the International Council for Harmonisation. These divergences can accelerate deployment in urgent scenarios but raise concerns about equitable risk assessment, with Western agencies prioritizing extended follow-up to address vector-specific toxicities absent in faster tracks.

Ethical Debates on Human Application

The development of viral vectors for human gene therapy has prompted debates over informed consent, given the irreversible nature of genetic modifications and the potential for unforeseen adverse effects. In the 1999 clinical trial at the University of Pennsylvania involving an adenovirus vector for ornithine transcarbamylase deficiency, 18-year-old participant Jesse Gelsinger died from a massive immune response leading to multi-organ failure, highlighting failures in risk disclosure: prior animal studies showing deaths from similar doses were not fully conveyed to participants or regulators, and investigators had undisclosed financial ties to the sponsoring company. This incident, which prompted a temporary halt to U.S. gene therapy trials by the FDA, underscored ethical lapses in balancing therapeutic promise against empirical risks, as vectors like adenoviruses can trigger severe inflammation despite preclinical data suggesting tolerability at lower doses. Distinctions between and applications intensify ethical scrutiny, as vectors primarily target non-reproductive cells for but carry risks of off-target or unintended via recombination or cellular uptake. edits address diseases like , approved in therapies such as Zolgensma using AAV9 vectors since 2019, yet critics argue that incomplete vector containment—evidenced by rare cases of vector DNA persistence in non-target tissues—raises issues for future offspring without their input. editing, though largely prohibited internationally due to heritable alterations, evokes concerns over eugenics-like selection and , as empirical from animal models show vectors can achieve at rates up to 10-20% in some protocols, potentially enabling enhancements beyond . Proponents contend that first-in-human trials, regulated under frameworks like the NIH's Advisory Committee, mitigate these via phased risk assessments, but skeptics highlight systemic underreporting of long-term oncogenic risks in peer-reviewed . Equity in access forms another core debate, as viral vector therapies command costs exceeding $2 million per treatment—e.g., Luxturna for retinal dystrophy at $850,000 in 2017—limiting availability to affluent populations despite public funding of foundational research. This disparity, compounded by manufacturing scalability challenges for biologics like AAV vectors requiring specialized bioreactors, raises causal questions about whether such technologies exacerbate social divides rather than equitably curing monogenic diseases affecting 1 in 250 births globally. In vaccine contexts, such as adenoviral vectors in AstraZeneca's COVID-19 product authorized in 2020, ethical tensions arose over mandates and global distribution inequities, with lower-income nations receiving only 0.1 doses per capita by mid-2021 versus 20+ in high-income countries, though vector-specific risks like rare thrombosis were empirically low at 1-2 per million doses. Early precedents, including the 1975 Asilomar Conference organized by , established voluntary moratoriums on certain recombinant DNA experiments involving viral vectors to avert hypothetical biohazards like oncogenesis, influencing modern levels (BL1-BL4) that prioritize containment over outright bans. These guidelines, born from first-principles assessment of vector pathogenicity—e.g., SV40 contamination risks in early vaccines—emphasized empirical containment efficacy, yet debates persist on whether self-regulation suffices amid incentives for rapid commercialization, as evidenced by post-Asilomar accelerations in vector engineering without proportional ethical oversight reforms. Overall, while viral vectors have enabled successes like curing 90% of severe hemophilia B cases in AAV trials by 2022, ethical frameworks demand rigorous, transparent risk-benefit analyses to avoid prioritizing innovation over verifiable safety.

Recent Advances and Future Prospects

Innovations in Vector Engineering

Innovations in viral vector engineering have primarily focused on enhancing efficiency, reducing , and improving specificity, particularly for (AAV) s. Advances in AAV design integrate rational , , and artificial intelligence-driven approaches to generate novel variants with superior targeting capabilities; for instance, models trained since 2015 have enabled the creation of capsids exhibiting up to tenfold higher in specific s while evading pre-existing immunity. These engineered capsids, such as those developed for non-human models like AAV-ShD, demonstrate enhanced delivery to muscle and s, addressing limitations in wild-type AAV serotypes. Lentiviral vector engineering has emphasized safety enhancements through multi-generational refinements, including the development of second- and third-generation systems that eliminate accessory genes and incorporate self-inactivating long terminal repeats (LTRs) to minimize risks. Production protocols have been optimized to reduce replication-competent virus formation, with modifications like separated and cassettes achieving near-zero RCV incidence in clinical-grade batches as of 2020. Recent strategies further boost efficiency by leveraging hypoxia-inducible factors for improved in hard-to-transfect cells, yielding up to 5-fold increases in stable integration without compromising vector stability. Adenoviral vectors have seen progress in gutless or helper-dependent designs, which remove all viral coding sequences to curtail inflammatory responses and enable larger payloads up to 36 kb. Recombineering techniques, advanced in the , facilitate rapid construction of serotype chimeras that evade hepatic sequestration by Kupffer cells, improving systemic delivery efficiency by 20-50% in preclinical models. These modifications, combined with tropism-altering fiber protein swaps, have expanded applications in oncolytic therapies and platforms. As of the second quarter of 2025, the clinical pipeline for viral vector-based therapies encompasses over 2,154 candidates, with (AAV) vectors comprising the majority due to their favorable profile and for applications. Lentiviral vectors dominate approaches, particularly in for CAR-T cell therapies, while adenoviral vectors persist in development and certain oncolytic applications. Approximately 250 AAV-specific candidates are advancing, led by over 180 companies targeting indications such as neuromuscular disorders, hemophilia, and inherited blindness. Phase III trials predominate for hemophilia A and B, with ongoing efforts to mitigate and dose-limiting toxicities through engineering. Recent U.S. (FDA) approvals underscore pipeline maturation: Elevidys (delandistrogene moxeparvovec, AAVrh74 for ) received full approval for ambulatory patients and accelerated approval for non-ambulatory ones in 2024; Beqvez (fidanacogene elaparvovec, AAV5 for hemophilia B) was approved in April 2024; and SKYSONA (elivaldogene autotemcel, lentiviral for cerebral ) marked the second lentiviral vector approval. These follow earlier AAV successes like Roctavian ( for hemophilia A, 2023) and Hemgenix (etranacogene dezaparvovec for hemophilia B, 2022), reflecting a shift toward durable, one-time treatments despite challenges like vector shedding and hepatic toxicity. Globally, China's approval of BBM-H901 (AAV-based) in April 2025 highlights regulatory divergence, with faster pathways in accelerating pipeline diversity. Market trends indicate robust expansion driven by approval momentum and rising demand for personalized therapies. The viral vector sector, valued at $5.5 billion in 2023, is projected to reach $6.3 billion in 2025 and $18.8 billion by 2030, with a (CAGR) exceeding 19%. holds 47% market share, fueled by U.S. biopharma investments, while grows fastest at over 22% CAGR through 2030 due to contract scale-up. Key drivers include scalability improvements in bioreactors and purification, though bottlenecks in DNA supply and empty removal constrain output. Development costs remain high, averaging $1-2 billion per therapy, prompting partnerships like Coave Therapeutics' CNS-targeted launch in May 2025. financing dipped in Q2 2025 amid economic pressures, yet trial initiations rose, signaling sustained investor confidence in viral vectors' therapeutic edge over non-viral alternatives.
Vector TypeKey Pipeline FocusNotable 2023-2025 ApprovalsMarket Projection (2025 Value)
AAVIn vivo gene delivery (e.g., neuromuscular, ocular)Elevidys (2024), Beqvez (2024), Roctavian (2023)Dominant in $6.3B manufacturing segment
LentiviralEx vivo (e.g., CAR-T, hematopoietic)SKYSONA (prior, reinforced pipeline)Growing in oncology, ~20% CAGR
AdenoviralVaccines, oncolyticsLimited new; legacy in COVID-eraNiche, <10% share but scalable

Challenges and Alternative Approaches

Viral vectors face significant challenges related to , which can trigger strong immune responses that neutralize the vector and limit its efficacy, particularly in repeat administrations or in individuals with pre-existing immunity. For instance, adenoviral vectors elicit robust innate and adaptive immune reactions, reducing expression duration and complicating vaccine booster doses, as observed in clinical trials for vaccines where anti-vector antibodies diminished subsequent immune responses to the . Safety concerns include in integrating vectors like retroviruses and lentiviruses, which can disrupt host genes and promote oncogenesis; a notable example is the 1999 X-SCID trial where two children developed T-cell due to LMO2 proto-oncogene activation following retroviral near enhancer regions. Non-integrating vectors, such as (AAV), avoid this risk but suffer from limited payload capacity (typically under 5 kb) and potential at high doses, as evidenced by elevated liver enzymes and immune-mediated clearance in hemophilia trials. Manufacturing scalability poses another hurdle, with viral vector production requiring complex systems prone to low yields, batch variability, and risks from adventitious agents, often resulting in costs exceeding $1 million per gram for AAV vectors as of 2022. These issues stem from reliance on adherent cultures or transient , which limit titers to 10^13–10^14 vector genomes per liter for AAV, far below therapeutic demands for widespread applications. Alternative approaches emphasize non-viral gene delivery systems, which circumvent immunogenicity and integration risks while offering greater flexibility in payload size and chemical customization. Lipid nanoparticles (LNPs) have emerged as a leading option, enabling efficient mRNA or DNA delivery without viral components; for example, LNPs facilitated the rapid deployment of COVID-19 mRNA vaccines, achieving transfection efficiencies comparable to viral vectors in some hepatic and muscular targets but with reduced innate immune activation.00278-6/fulltext) Physical methods, such as electroporation and sonoporation, apply electrical pulses or ultrasound to transiently permeabilize cell membranes for direct nucleic acid uptake, bypassing endosomal barriers and achieving up to 80% transfection in ex vivo applications like CAR-T cell engineering, though they require specialized equipment and can cause cell damage at high intensities. Polymer-based vectors, including polyethyleneimine (PEI) and chitosan nanoparticles, provide electrostatic condensation of DNA for cellular uptake via endocytosis, with recent modifications enhancing endosomal escape and targeting specificity, as demonstrated in preclinical studies yielding sustained expression in vivo without the mutagenic potential of viruses. Despite lower transfection efficiencies (often 10–50% versus viral methods), these non-viral strategies support scalable, cost-effective production and repeated dosing, addressing key viral limitations in chronic disease therapies.

References

  1. [1]
    Viral Vectors - Gene & Cell Therapy Patient Education | ASGCT
    Jan 30, 2024 · Vectors are essentially vehicles designed to deliver therapeutic genetic material, such as a working gene, directly into a cell. There are four ...
  2. [2]
    Viral vector platforms within the gene therapy landscape - Nature
    Feb 8, 2021 · Contemporary viral vector-based gene therapy is achieved by in vivo delivery of the therapeutic gene into the patient by vectors based on ...
  3. [3]
    Paul Berg | Science History Institute
    In 1971 Berg's landmark gene-splicing experiment opened the door to the invention of recombinant DNA technology.
  4. [4]
    Personal Reflections on the Origins and Emergence of Recombinant ...
    In early 1971, the American Cancer Society approved a grant application in which Berg proposed to develop the means for transducing foreign DNA into mammalian ...Abstract · Creating Recombinant Dna In... · Figure 2
  5. [5]
    A complete guide to viral vectors | Revvity
    Nov 13, 2024 · Learn about viral vectors: what they are, how they can be applied to science, and the key attribute of AAVs, LVs, and AVs.
  6. [6]
    Viral Vectors in Gene Therapy - PMC - PubMed Central - NIH
    Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious ...
  7. [7]
    Viral vector‐based gene therapies in the clinic - Zhao - 2022
    Sep 14, 2021 · In this review, we highlight the diversity of viral vectors, review approved products, and discuss the current clinical landscape of in vivo viral vector-based ...<|separator|>
  8. [8]
    What are Adenovirus-Based Vaccines? - News-Medical
    Other adenovirus-based SARS-CoV-2 vaccines in development include the Sputnik V vaccine and the Johnson & Johnson vaccine. The Johnson & Johnson vaccine has now ...
  9. [9]
    COVID-19 viral vector-based vaccines - Canada.ca
    Jan 10, 2024 · The previously authorised Janssen Jcovden (Johnson & Johnson) COVID-19 vaccine and Astrazeneca Vaxzevria COVID-19 vaccine are viral vector-based vaccines.
  10. [10]
    Thrombotic Microangiopathy as an Emerging Complication of Viral ...
    Although studies have shown the efficacy of gene therapy, serious adverse events have also emerged, including thrombotic microangiopathy (TMA) following viral ...Aav Transduction Of Cells · Host Immune Responses To... · Gene Therapy--Associated Tma
  11. [11]
    Viral Vector Systems for Gene Therapy: A Comprehensive Literature ...
    In this review, we highlight some of the biosafety challenges associated with the current viral vector systems that have applications in gene therapy research.
  12. [12]
    Is the tide turning on viral vectors? The shifting landscape of gene ...
    Aug 6, 2025 · However, as the CGT market has expanded, concerns regarding the safety and efficiency of viral vectors have grown. Researchers have begun to ...
  13. [13]
    Viral vectors as a novel tool for clinical and neuropsychiatric ...
    Oct 25, 2018 · A viral vector is a genetically modified vector produced by genetic engineering. As pathogenic genes in the virus are completely or largely ...
  14. [14]
    Viral vector vaccines - PMC - PubMed Central - NIH
    May 25, 2022 · Viral vectors are a relatively new vaccine platform that relies on recombinant viruses to deliver selected immunogens into the host.
  15. [15]
    Viral Vector Process Development and Manufacturing
    Apr 1, 2024 · Manufacturing processes for viral vectors include a variety of approaches, predominantly based on the use of mammalian cells in either adherent or suspension- ...
  16. [16]
    Overview: Recombinant Viral Vectors as Neuroscience Tools - PMC
    Apr 1, 2020 · Recombinant viruses are highly efficient vehicles for in vivo gene delivery. Viral vectors expand the neurobiology toolbox to include direct ...Structure Of Viruses · Viral Tropism: Serotypes And... · Aav Serotypes
  17. [17]
    Adeno-Associated Virus (AAV) as a Vector for Gene Therapy - PMC
    Adeno-associated virus (AAV) is a versatile viral vector technology that can be engineered for very specific functionality in gene therapy applications. To date ...
  18. [18]
    Viral Vectors for Gene Delivery to the Central Nervous System - PMC
    At the forefront of potential vectors, viral systems have evolved to efficiently deliver their genetic material to a cell. The biology of different viruses ...
  19. [19]
    Viral Vector-Based Gene Therapy - PMC - PubMed Central
    Viral vectors that have been artificially modified to lose their pathogenicity are used widely as a delivery system, with the key advantages of their natural ...1. Introduction · Table 2 · 2. Aav Vectors<|separator|>
  20. [20]
    Construction and application of adenoviral vectors - PMC - NIH
    (B) The viral capsid of adenovirus has a diameter of 80–100 nm. The viral capsid has a typical icosahedral symmetry, consisting of 252 8–10 nm diameter capsid ...
  21. [21]
    Lentivirus and Lentiviral Vectors | Office of Research Safety
    HIV-derived vectors are highly efficient vehicles for in vivo gene delivery. Lentiviruses are approx. 120nm in diameter, enveloped, and contain a nucleocapsid ...Missing: properties | Show results with:properties
  22. [22]
    Manufacturing Challenges and Rational Formulation Development ...
    The role of buffer, pH, excipients, and impurities on the stability of viral vectors is also discussed. As such, the aim of this review is to outline the tools ...
  23. [23]
    Development of Long-Term Stability of Enveloped rVSV Viral Vector ...
    Liquid formulations are used to preserve the structural integrity of viral vectors by preventing denaturation and degradation. They contain protective ...
  24. [24]
    Viral and non-viral vectors in gene therapy: current state and clinical ...
    Jul 1, 2025 · An ideal viral vector needs to (1) be able to carry therapeutic genes of a certain size, (2) be easy to package and produce, (3) be replication- ...
  25. [25]
    Advances and Future Challenges in Adenoviral Vector ...
    Adenoviruses (Ads) are icosahedral non-enveloped DNA viruses with diameters approximately 90 to 100 nm (Fig. 1). Ads were first isolated from human adenoids in ...
  26. [26]
    Recent Advances in Lentiviral Vector Development and Applications
    Lentiviral vectors (LVs) have emerged as potent and versatile vectors for ex vivo or in vivo gene transfer into dividing and nondividing cells.
  27. [27]
    Past, Present, and Future of Viral Vector Vaccine Platforms
    May 15, 2025 · This review provides an exhaustive, in-depth examination of the historical evolution, immunobiological principles, current platforms, manufacturing ...
  28. [28]
    Biosafety Features of Lentiviral Vectors - PMC - PubMed Central - NIH
    Over the past decades, lentiviral vectors have evolved as a benchmark tool for stable gene transfer into cells with a high replicative potential.
  29. [29]
    The use of retroviral vectors for gene therapy-what ... - PubMed Central
    The most important advantage that retroviral vectors offer is their ability to transform their single stranded RNA genome into a double stranded DNA molecule ...
  30. [30]
    Retroviral Vectors and Gene Therapy - Holland-Frei Cancer Medicine
    Retroviral vectors are perhaps the best studied and most promising transducing vectors. They integrate, so they theoretically only need to be administered once.Missing: definition | Show results with:definition
  31. [31]
    Retroviral Vector Technology for Gene Therapy - ScienceDirect.com
    Oct 3, 2025 · ... vector design. The advantages of using retroviral vector systems include: 1) a relatively large packaging capacity of about 8-9 kb (e.g. ...
  32. [32]
    Retroviral vectors and transposons for stable gene therapy
    Oct 12, 2016 · For two decades, retrovirus family vectors have offered several attractive properties as stable gene-delivery vehicles. These vectors represent ...
  33. [33]
    Retroviral Vectors in Gene Therapy: Mechanism of Integration ...
    Introduction. Retroviral vectors have gained an increasing value in gene therapy because they stably deliver therapeutic genes to the host cell genome.Missing: advantages | Show results with:advantages
  34. [34]
    Viral Vectors 101: Types of viruses - Addgene Blog
    Jun 6, 2023 · Lentiviruses have slightly larger packaging capacities than gamma-retrovirus, coming in around 9 kb. Unlike gamma, lentivirus can infect both ...Missing: review | Show results with:review
  35. [35]
    Clinical use of lentiviral vectors | Leukemia - Nature
    Mar 22, 2018 · In this review, we discuss several aspects of lentiviral vectors that will be of interest to clinicians, including an overview of lentiviral ...
  36. [36]
    Improved third-generation lentiviral packaging with pLKO.1C vectors
    Mar 6, 2020 · A widely used third-generation lentiviral packaging system produces virus with enhanced biosafety by eliminating HIV accessory genes and ...
  37. [37]
    Clinical use of lentiviral vectors - PMC - PubMed Central
    First-generation lentiviral vectors contained a significant portion of the HIV genome, including the gag and pol genes, as well as several additional viral ...
  38. [38]
    The use of adenoviral vectors in gene therapy and vaccine ... - NIH
    Although adenoviral vectors are successful in gene transferring and expression, some concerns were raised. In vivo delivery of recombinant adenoviral vector ...
  39. [39]
    Four decades of adenovirus gene transfer vectors - Cell Press
    Apr 2, 2025 · Adenoviruses (Ad) are 90- to 100-nm nonenveloped viruses comprising an icosahedral protein capsid encompassing a double-stranded DNA genome.
  40. [40]
    Developing the next-generation of adenoviral vector vaccines - PMC
    Jul 1, 2025 · Adenoviral vector vaccines are typically rendered replication-incompetent by deletion of the E1 gene. In the wild-type virus, this encodes a set ...
  41. [41]
    Strategies for Modifying Adenoviral Vectors for Gene Therapy
    Nov 20, 2024 · Adenoviral vectors (AdVs) are effective vectors for gene therapy due to their broad tropism, large capacity, and high transduction ...
  42. [42]
    The Adenovirus Vector Platform: Novel Insights into Rational Vector ...
    Jan 11, 2023 · The adenovirus genome and capsids can be modified using highly efficient techniques, and vectors can be produced at high titers, which ...
  43. [43]
    Adenovirus Vectors: Excellent Tools for Vaccine Development - PMC
    These properties allow efficient induction of robust and durable cell-mediated and humoral immunity and make adenoviral vectors promising vaccine vectors.
  44. [44]
    Advantages and Drawbacks of Adenovirus Vector-mediated Gene ...
    1) Well tolerated, with post-infection viability of the host cells being almost 100%. 2) Great packaging capacity (up to 8kb). 3) Broad range of infectivity.
  45. [45]
    Clinical Application of Adenovirus (AdV): A Comprehensive Review
    Jul 8, 2024 · Adenovirus vectors have proven to be a viable option for gene therapy, effectively delivering genetic material into targeted cells to address ...
  46. [46]
    Advantages and disadvantages of adenovirus vectors. - ResearchGate
    In general, adenoviral vectors are recognized for their high immunogenicity, and their use is limited due to the widespread pre-existing immunity in the ...
  47. [47]
    Adenoviral vector vaccine platforms in the SARS-CoV-2 pandemic
    Aug 5, 2021 · The human Ad35, Ad11, and Ad26, although less immunogenic than Ad5, are still effective vectors for vaccination purposes. Additionally, Ads from ...
  48. [48]
    Adenoviral Vector-Based Vaccine Platform for COVID-19 - NIH
    The rVSV-ZEBOV vaccine is an illustration of a viral vector vaccine [24]. Imojev, a vaccine against Japanese Encephalitis, was the first viral vector-based ...
  49. [49]
    Adenovirus Vectors' Timeless Influence on Cancer, Gene Therapy ...
    Dec 3, 2023 · This comprehensive review seeks to offer a summary of adenovirus vectors, exploring their characteristics, genetic enhancements, and diverse applications.
  50. [50]
    Adenoviral Vectors for Gene Therapy of Hereditary Diseases
    Dec 16, 2024 · This review provides an overview of AdVs, their features, and clinical trials using them for gene replacement therapy in monogenic diseases and ...<|separator|>
  51. [51]
    Packaging Capacity of Adeno-Associated Virus Serotypes
    This study illustrates that AAV is capable of packaging and protecting recombinant genomes as large as 6.0 kb but the larger genome-containing virions are ...Missing: immunogenicity | Show results with:immunogenicity
  52. [52]
    Adeno-associated virus as a delivery vector for gene therapy of ...
    Apr 3, 2024 · Viral vector platforms within the gene therapy landscape. Sig ... Overview of MicroRNA biogenesis, mechanisms of actions, and circulation.
  53. [53]
    Various AAV Serotypes and Their Applications in Gene Therapy
    In this article, we present a review of AAV discovery, properties, different serotypes, and tropism, and a following detailed explanation of their uses in gene ...
  54. [54]
    Adeno-Associated Viral Vectors in Neuroscience Research - PMC
    The introduction of the adeno-associated viral vector (AAV) in neuroscience has helped advance both circuit identification and functional circuit analysis, and ...
  55. [55]
    Adeno-associated virus vector as a platform for gene therapy delivery
    Adeno-associated virus (AAV) vectors are the leading platform for gene delivery for the treatment of a variety of human diseases.
  56. [56]
    Understanding AAV vector immunogenicity: from particle to patient
    This review focuses on aspects of immunity against rAAV, mediated by anti-AAV neutralizing antibodies (NAbs) arising after natural exposure to AAVs or after ...Missing: history | Show results with:history
  57. [57]
    Immunogenicity of Recombinant Adeno-Associated Virus (AAV ...
    Mar 2, 2023 · This review discusses the contribution of the innate and adaptive immune response against AAVs, highlighting the challenges and potential strategies to ...
  58. [58]
    Approved Cellular and Gene Therapy Products - FDA
    Aug 15, 2025 · Approved Products ; LUXTURNA (voretigene neparvovec-rzyl), Spark Therapeutics, Inc. ; LYFGENIA (lovotibeglogene autotemcel [lovo-cel]), bluebird ...Zolgensma · Luxturna · Abecma · Adstiladrin
  59. [59]
    AAV Vs. Lentiviral Vectors - Life in the Lab - Thermo Fisher Scientific
    Sep 27, 2024 · Two vector types, adeno-associated (AAV) and lentiviral vectors (LV), have emerged as the popular virus types for in vivo and in vitro gene correction.
  60. [60]
    Herpesvirus Vectors in Gene Therapy - PMC - PubMed Central
    In an extensive review, Manservigi et al. elucidate the design and applications of recombinant HSV vectors, with an emphasis on the genomic HSV vectors.
  61. [61]
    Next-generation replication-defective HSV vectors for delivery of ...
    May 7, 2025 · This review explores next-generation rdHSV vectors, which do not express viral genes, offer high payload capacity, and can be engineered for safe, long-term ...
  62. [62]
    Review HSV-1 as a gene delivery platform for cancer gene therapy
    Syncytium-forming HSV-1 RP1 demonstrates 24.6% objective response rate in checkpoint-refractory melanoma, suggesting a promising oncolytic approach for cancer ...
  63. [63]
    The use of viral vectors in vaccine development - Nature
    Jul 4, 2022 · One advantage of using poxviral vectors is their cargo capacity for heterologous genes of around 25 kb which is far larger than that of other ...
  64. [64]
    Reflections on the Early Development of Poxvirus Vectors - PMC - NIH
    Poxvirus expression vectors were described in 1982 and quickly became widely used for vaccine development as well as research in numerous fields.
  65. [65]
    Enhancing poxvirus vectors vaccine immunogenicity - PMC - NIH
    In this review we describe several approaches for vaccine improvements of poxvirus vectors, with particular emphasis in the 2 candidate vaccine vectors most ...
  66. [66]
    Poxviral vectors for cancer immunotherapy - PMC - NIH
    This review discusses the clinical development of poxviral-based cancer vaccines, with a particular focus on the rationale for combining vaccines with other ...<|separator|>
  67. [67]
    Poxvirus vectors: orphaned and underappreciated - PMC - NIH
    Owing to their lack of immunological cross reactivity with vaccinia, fowlpox and other variants confer enhanced vaccine activity when combined with vaccinia.
  68. [68]
    Baculovirus Vectors for Gene Therapy - PMC - PubMed Central - NIH
    This chapter reviews the history of this research area, cells permissive to baculovirus transduction, factors influencing transduction and transgene expression ...
  69. [69]
    Baculovirus as a Tool for Gene Delivery and Gene Therapy - PMC
    Sep 19, 2018 · Baculovirus has been widely used as a gene expression tool for more than thirty years based on its ability to express high levels of proteins in insect cells.
  70. [70]
    Baculoviruses in Gene Therapy and Personalized Medicine - PMC
    Apr 28, 2021 · This review concludes that baculoviruses are an excllent candidate for gene therapy, personalized medicine and other biotherapeutic applications ...
  71. [71]
    Baculovirus vectors for gene therapy - PubMed
    This chapter reviews the history of this research area, cells permissive to baculovirus transduction, factors influencing transduction and transgene expression ...
  72. [72]
    Lentiviral Vectors and Adeno‐Associated Virus Vectors: Useful ...
    In this review, we have outlined characteristics of lentiviral and AAV vectors and their application in pain research. Lentiviral vectors can induce transgene ...
  73. [73]
    Viral vector-based tools advance knowledge of basal ganglia ...
    The use of viral vectors in basic research has revealed that BG circuitry is more complex than previously thought based on traditional neuronal tracers (Akkal ...
  74. [74]
    Gene therapy of adeno‐associated virus (AAV) vectors in preclinical ...
    Aug 8, 2023 · This review aims to provide an overview of the biological characteristics of AAV vectors and the therapeutic advancements observed in preclinical models of ...<|control11|><|separator|>
  75. [75]
    A Review of Viral Vectors in Gene Therapy | Biocompare
    Jun 5, 2025 · In this article, we highlight the leading viral vectors for cell and gene therapy, as well as considerations for early-stage research projects.
  76. [76]
    Evolution of Gene Therapy, Historical Perspective - PMC - NIH
    The history of gene therapy comprises the advance of theoretical concepts, understanding the human genome, availability of critical materials and instruments.
  77. [77]
    3 Examples of FDA approved AAVs in gene therapy
    Sep 19, 2023 · Luxturna is making history as the first FDA-approved AAV gene therapy for a genetic disease. The therapy employs a harmless AAV vector to ...
  78. [78]
    Adeno-associated virus therapies: Pioneering solutions for human ...
    In 2017, Luxturna became the first AAV-based gene therapy approved by the FDA in the United States, treating LCA[24], [25]. In a phase 3 randomized, controlled ...
  79. [79]
    Viral vector‐based gene therapies in the clinic - PMC
    ZOLGENSMA® is the only approved viral gene therapy for treating neurological disorders. It was approved by US FDA in 2019 for treating spinal muscular atrophy ...<|separator|>
  80. [80]
    Viral Vectors in Gene Therapy: Where Do We Stand in 2023?
    Mar 7, 2023 · Viral vectors have shown great promise in the treatment of chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, ...
  81. [81]
    Viral vectored vaccines: design, development, preventive and ...
    Apr 7, 2023 · Viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases.
  82. [82]
    Viral Vector Vaccine Development and Application during the ... - NIH
    Viral vector vaccines combine the safety advantages of inactivated vaccines with the immunological benefits and low cost of live vaccines, which have been ...Missing: peer | Show results with:peer
  83. [83]
    Viral vector vaccines – What they are, and what they are not | CEPI
    Apr 8, 2025 · Examples of vector viruses include adenoviruses—which, when live, are a cause of commons colds—as well as the vesicular stomatitis virus, or ...
  84. [84]
    Adenoviral vaccines - Sputnik V
    Adenoviral vectors are considered extremely safe, and are some of the easiest to engineer. Vectors are viruses that have had the gene responsible for ...
  85. [85]
    Table 1 Advantages and disadvantages of different vaccine platforms
    Viral vector vaccine. Stronger immune response; Preservation of native antigen; Mimicking natural infection. More complicated manufacturing process; Risk of ...Missing: peer | Show results with:peer
  86. [86]
    The use of viral vectors in vaccine development - PMC
    Jul 4, 2022 · One advantage of replicating vectors is their mimicking of a natural infection, resulting in the induction of cytokines and co-stimulatory ...
  87. [87]
    Viral Vector Vaccines Against ASF: Problems and Prospectives
    Mar 10, 2022 · The main disadvantages of viral vector vaccines are that they represent a more complex production process (22), they risk genomic integration, ...
  88. [88]
    Oncolytic virotherapy: basic principles, recent advances and future ...
    Apr 11, 2023 · Some OVs have shown to present high safety in both animal experiments and clinical trials, such as NDV, VSV and SVV. None of them are human- ...
  89. [89]
    Mechanism of Action for Talimogene Laherparepvec, a New ...
    On the basis of the study outcome, T-VEC was approved for the treatment of skin and lymph node melanoma accessible for local injection. Physicians will need to ...<|separator|>
  90. [90]
    Talimogene Laherparepvec (T-VEC): A Review of the Recent ... - NIH
    Jan 31, 2023 · Talimogene laherparepvec (T-VEC), a genetically modified herpes simplex virus (HSV), has shown promising therapeutic effects in the treatment of advanced ...
  91. [91]
    Talimogene Laherparepvec (T-VEC): An Intralesional Cancer ... - NIH
    Mar 18, 2021 · Results obtained in recently published clinical trials, showed an effective efficacy of this intralesional treatment on both injected and not- ...
  92. [92]
    Efficacy and tolerability of neoadjuvant therapy with Talimogene ...
    Jan 16, 2025 · We present a single-arm, phase II, neoadjuvant trial with the oncolytic virus talimogene laherparepvec (T-VEC) in 18 patients with difficult-to-resect ...
  93. [93]
    Oncolytic virus therapy: A new era of cancer treatment at dawn - NIH
    Other oncolytic viruses that are closing in on drug approval in North America and Europe include vaccinia virus JX‐594 (pexastimogene devacirepvec) for ...
  94. [94]
    Oncolytic viral vectors in the era of diversified cancer therapy
    Given that the favorable anti-tumor efficacy of the virus in clinical trials, phase 3 clinical trials in combination with PD-1 antagonists have been carried out ...Missing: approvals | Show results with:approvals
  95. [95]
    Oncolytic virus-based suicide gene therapy for cancer treatment
    Apr 10, 2023 · Success of clinical studies led to approval of numerous gene therapy products for cancer treatment (Table 2). Herein we review some of the gene ...
  96. [96]
    A systematic analysis on the clinical safety and efficacy of onco ...
    Several onco-virotherapy candidates have been developed and clinically evaluated for the treatment of cancer, and several are approved for clinical use.
  97. [97]
    The Principles of Viral Vector Design & Development - Form Bio
    Jul 11, 2023 · Viral vectors are the favored vehicle used in gene therapy development and exhibit many advantages over non-viral methods.Lentivirus (lv) · Viral Vector Type · Immunogenicity And Other...
  98. [98]
    Scalable AAV production process for gene therapy - Cytiva
    Sep 4, 2024 · See how we developed an AAV production process that successfully scaled to 25 L bioreactors. Generated high-titer AAV vector for AAV2, AAV5, ...
  99. [99]
  100. [100]
    AAV Production and Manufacturing - SK pharmteco
    Scalable Manufacturing. Support volumes from 200 to 1,000 L with reliable scalability, maintaining consistent output across bioreactors and reducing risk during ...
  101. [101]
    Development and optimization of large scale production of ...
    For large scale adenovirus production, the fixed-bed iCellis® 500 single-used bioreactor is used. . Using iCELLis®, an optimized manufacturing processes for ...<|separator|>
  102. [102]
    Accelerated and intensified manufacturing of an adenovirus ...
    We describe work to optimize adenoviral vector manufacturing for rapid response, by minimizing time to clinical trial and first large‐scale supply.
  103. [103]
    A Novel Scalable Production Platform for Gene Therapy Vectors ...
    A design-of-experiments approach has been employed to optimize transient production of lentiviral vectors with significantly higher titers than can be obtained ...
  104. [104]
    Developing a Robust and Scalable Platform for AAV8 Production
    Sep 9, 2025 · Development and implementation of a scalable AAV8 production process, from 2 to 50 liters, using stirred tank bioreactors, with in situ cell ...
  105. [105]
    [PDF] scalable-aav-vector-production-purification-expisf-expression ...
    Production of rAAV particles was scalable from shake flask to bioreactor scale, yielding high viral genome titers (vg/mL) of infectious rAAV, and high rAAV ...
  106. [106]
    Challenges in scaling up AAV-based gene therapy manufacturing
    Apr 29, 2023 · Accelerating the scale up of adeno-associated virus (AAV) manufacture is highly desirable to meet the increased demand for gene therapies.
  107. [107]
    Critical Assessment of Purification and Analytical Technologies for ...
    Jul 26, 2021 · This review provides insights on the likely presence of EVs in the final formulation of enveloped virus products and discusses the potential to further resolve ...
  108. [108]
    Overview of current scalable methods for purification of viral vectors
    In this chapter, we provide an overview of the most commonly used purification methods for viral gene therapy vectors.
  109. [109]
    [PDF] A Review of Virus-Vectored Vaccines: Current Production Methods ...
    The goal of this review is to provide a comprehensive overview of the types of viral vector vaccines, production and purification methods, current and future ...
  110. [110]
    [PDF] Challenges in Downstream Purification of Gene Therapy Viral Vectors
    While some have used this traditional method to purify clinical viral vectors, learning and adapting from protein purification methods are the focus of future ...
  111. [111]
    [PDF] Chemistry, Manufacturing, and Control Information for Human Gene ...
    This guidance is organized to follow the structure of the. FDA guidance on the Common Technical Document (CTD). Information on the CTD can be found in FDA's ...
  112. [112]
    Quality control for Adeno-associated viral vector production - PMC
    rAAV quality control consists of 4 general categories, quantification, identity confirmation, purity and functional testing. A number of assays are possible for ...
  113. [113]
    Vector characterization methods for quality control testing ... - PubMed
    This chapter focuses on QC testing, providing an overview of characterization methods appropriate for clinical vectors prepared for early phase clinical studies ...
  114. [114]
    Quality Control Testing of Viral Vectors - PathoQuest
    With NGS biotesting, PathoQuest offers characterization, quality control and release of viral vectors & plasmids across the manufacturing journey.<|separator|>
  115. [115]
    [PDF] Manufacturing of Gene Therapies: Ensuring Product Safety and ...
    Therefore, FDA has allowed phasing in of GMP and characterization requirements during clinical development, with the expectation that the manufacturer ...
  116. [116]
    Overcoming innate immune barriers that impede AAV gene therapy ...
    Jan 4, 2021 · This Review highlights what is known about innate immune responses against rAAVs and discusses potential strategies to circumvent these pathways.
  117. [117]
    Immunogenicity and toxicity of AAV gene therapy - Frontiers
    Aug 11, 2022 · This manuscript reviews the different types of immune responses that are elicited by AAVs and how they translate into lack of efficacy or even worse toxicity.
  118. [118]
    Pre-existing immunity against Ad vectors - PubMed Central - NIH
    In addition to Ad specific T and B cells, innate immune responses to Ad vectors can also dampen Ad efficacy, especially in the context of pre-existing immunity ...
  119. [119]
    Immune Responses to Viral Gene Therapy Vectors - PMC - NIH
    This article reviews immune response mechanisms against adenoviral, adeno-associated viral, and lentiviral vectors.
  120. [120]
    Engineering adeno-associated viral vectors to evade innate immune ...
    Feb 10, 2021 · The engineered vectors elicited markedly reduced innate immune and T cell responses and enhanced gene expression in clinically relevant mouse ...<|separator|>
  121. [121]
    The curious case of AAV immunology - ScienceDirect.com
    May 7, 2025 · AAV vectors are associated with low immunogenicity and lack of pathogenicity; however, the innate immune response to AAV vectors can still ...
  122. [122]
    Challenges Posed by Immune Responses to AAV Vectors - Frontiers
    This article focuses on the inherent immunogenic features of rAAV vectors and provides a perspective that the removal of pathogen associated molecular patterns ...
  123. [123]
    rAAV immunogenicity, toxicity, and durability in 255 clinical trials
    Oct 27, 2022 · Immune responses against AAV pose significant challenges for gene therapy development, impacting patient enrollment, toxicity profile, and ...
  124. [124]
    Review Evading and overcoming AAV neutralization in gene therapy
    Currently patients seropositive for AAV neutralizing antibodies (NAbs) are excluded from most clinical trials, and NAb generation following vector ...Review · Aav Gene Therapy Vectors And... · Declaration Of Interests
  125. [125]
    Stem cell gene therapy: the risks of insertional mutagenesis ... - NIH
    This review will summarize current information on the mechanisms of insertional mutagenesis in hematopoietic stem and progenitor cells due to integrating gene ...
  126. [126]
    Preventing and exploiting the oncogenic potential of integrating ... - JCI
    γ-Retroviral vectors (γRVs), which are commonly used in gene therapy, can trigger oncogenesis by insertional mutagenesis. Here, we have dissected the ...
  127. [127]
    AAV Vectors, Insertional Mutagenesis, and Cancer - Cell Press
    From my own perspective, some trials using AAV seem too risky even with the limited data in hand, such as gene therapy for MPS VII using a β-glucuronidase ...
  128. [128]
    Insertional oncogenesis in 4 patients after retrovirus-mediated gene ...
    Aug 7, 2008 · Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients.
  129. [129]
    Insertional mutagenesis combined with acquired somatic mutations ...
    Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients.
  130. [130]
    Preventing and exploiting the oncogenic potential of integrating ...
    now provide evidence that the lentiviral insertion pattern reduces the risk of cell transformation by a factor of 10, as compared with similarly designed γ- ...
  131. [131]
    Risk and Benefit Assessment of Gene Therapy with Lentiviral ...
    Sep 1, 2025 · Seven cases of hematological malignancy reported in recipients of Skysona™ (elivaldogene autotemcel) have reignited long-standing concerns ...
  132. [132]
  133. [133]
    Assessment of Insertional Mutagenesis Risk Following AAV Vector ...
    Nov 20, 2009 · To assess the risk of insertional mutagenesis in cells transduced by AAV vector, we analyzed the incidence of hepatocellular carcinoma (HCC). We ...
  134. [134]
    Integration of Gene Therapy Vectors: A Risk Factor for ...
    Nov 15, 2023 · In all, these findings suggest that AAV genome integration is not a clear risk factor for tumorigenesis. These profiles hold true for rAAV ...
  135. [135]
    Current landscape of vector safety and genotoxicity after ... - Nature
    Apr 8, 2025 · We summarize current clinical information and revisit genotoxicity risks following ex-vivo gene modification of HSC and T cells.
  136. [136]
    Evaluating the state of the science for adeno-associated virus ...
    Aug 3, 2022 · The risks of AAV vector genotoxicity and carcinogenicity from insertional mutagenesis remain an ongoing concern for health authorities even ...
  137. [137]
    Reducing off target viral delivery in ovarian cancer gene therapy ...
    However, efficient vector targeting to tumors is difficult and off-target effects can be severely detrimental.
  138. [138]
    Off-target effects in CRISPR/Cas9 gene editing - PMC - NIH
    The off-target effects occur when Cas9 acts on untargeted genomic sites and creates cleavages that may lead to adverse outcomes. The off-target sites are often ...
  139. [139]
    evaluating and mitigating off-target effects in CRISPR gene editing ...
    In this review, we summarize the different methods for off-target assessment, discuss their strengths and limitations, and highlight strategies to improve the ...<|separator|>
  140. [140]
    Risks Associated With Lentiviral Vector Exposures and Prevention ...
    Dec 30, 2016 · LVV-infected cells can become cancerous through activation of oncogenes or inactivation of tumor suppressor genes. Insertional mutagenesis ...
  141. [141]
    Long-term safety of lentiviral or gammaretroviral gene-modified T ...
    Long-term risks of gene therapy are not fully understood. In this study, we evaluated safety outcomes in 783 patients over more than 2,200 total patient-years ...
  142. [142]
    [PDF] Long Term Follow-Up After Administration of Human Gene Therapy ...
    Similar to integrating vectors, genome editing may produce undesirable changes in the genome (whether ex vivo or in vivo), with the risk of malignancies,.
  143. [143]
    In vivo applications and toxicities of AAV-based gene therapies in ...
    Jul 17, 2025 · This review summarizes clinical trials of AAV gene therapies for rare diseases, including ophthalmic diseases, nervous system disorders, hematological diseases ...
  144. [144]
    Gene Therapy–Related Mortality: Mechanistic Insights, Regulatory ...
    Jun 11, 2025 · Most recently, patient deaths in AAV-based clinical trials have reignited longstanding concerns about vector-associated toxicity, immunogenicity ...
  145. [145]
    AAV cis-regulatory sequences are correlated with ocular toxicity
    Mar 4, 2019 · Our results suggest that ocular AAV toxicity is associated with certain AAV cis-regulatory sequences and/or their activity and that retinal damage occurs due ...<|control11|><|separator|>
  146. [146]
    INDUCTION OF ARGINASE ACTIVITY WITH THE SHOPE ... - NIH
    INDUCTION OF ARGINASE ACTIVITY WITH THE SHOPE PAPILLOMA VIRUS IN TISSUE CULTURE CELLS FROM AN ARGININEMIC PATIENT · Stanfield Rogers · A Lowenthal · H G Terheggen.
  147. [147]
    Stanfield Rogers: Insights into Virus Vectors and Failure of an Early ...
    In 1971 Rogers published an intriguing paper that demonstrated that the arginase ... papilloma virus-induced arginase associated with mutation of the virus.
  148. [148]
    GENETIC AID FAILS IN TEST ON HUMANS - The New York Times
    Mar 1, 1975 · The virus, called the Shope papilloma virus, was known to produce arginase ... Stanfield Rogers, then of Oak Ridge, Tenn facility, and now a ...
  149. [149]
    Circular SV40 DNA Molecules Containing Lambda Phage ... - PNAS
    We have developed methods for covalently joining duplex DNA molecules to one another and have used these techniques to construct circular dimers of SV40 DNA.
  150. [150]
    Paul Berg and the origins of recombinant DNA - ScienceDirect.com
    Feb 29, 2024 · Paul Berg, a professor in Stanford University's biochemistry department, was a leader in the early development of recombinant DNA technology.
  151. [151]
    Asilomar and Recombinant DNA: The End of the Beginning - NCBI
    The Asilomar conference was to address the anxiety over recombinant DNA, especially cancer virus DNA, and to determine what experiments should or should not be ...THE 1973 GORDON... · THE ACADEMY'S TURN · THE ASILOMAR CONFERENCE
  152. [152]
    Summary statement of the Asilomar conference on ... - PNAS
    The conference reviewed recombinant DNA research, agreed that no methodology is risk-free, and that containment should match the risk. Most work requires more ...
  153. [153]
    Asilomar conference on DNA recombinant molecules - Nature
    (2) Animal viruses: Experiments in- volving li-nkage of viral genomes or genome segments to prokaryotic vec- tors and their propagation in pro- karyotic cells ...
  154. [154]
    A brief account of viral vectors and their promise for gene therapy
    Jan 26, 2017 · Luigi Naldini and Inder Verma were among the first to create lentiviral vectors that added the capability of gene delivery to non-dividing cells ...
  155. [155]
    An Early History of Gene Transfer and Therapy - Mary Ann Liebert, Inc.
    Viral genomes were used for the development of the first relatively efficient methods for gene transfer into mammalian cells in culture. In the late 1970s, ...
  156. [156]
    Adenovirus: After 40 years, a call to arms
    May 21, 2021 · In the early 1980s, the first adenovirus vectors were designed, grown, and studied at CSHL. At the time, researchers were trying to ...
  157. [157]
    Adenovirus: The First Effective In Vivo Gene Delivery Vector - PMC
    The adenovirus was first isolated from human adenoids in 1953. There are over 50 known serotypes (Wilson, 1996; Hackett and Crystal, 2008). In the late 1980s ...
  158. [158]
    long-term results of the first clinical gene therapy trial - PubMed
    The first human gene therapy experiment begun in September 1990 used a retroviral vector containing the human adenosine deaminase (ADA) cDNA to transduce ...
  159. [159]
    Clinical Development of Gene Therapies: The First Three Decades ...
    In 1993, clinical studies with in vivo administration of retroviral vectors were proposed. Retroviral vectors can integrate into the human genome, which enables ...
  160. [160]
    Lessons Learned From the Gene Therapy Trial for Ornithine ...
    Jesse Gelsinger died from complications of vector administration in a liver gene therapy trial of research subjects with a deficiency of ornithine ...
  161. [161]
    The Death of Jesse Gelsinger, 20 Years Later
    Jun 4, 2019 · The news that an experimental treatment had killed a basically healthy volunteer rocked the field of gene therapy and the broader world of ...
  162. [162]
    The redemption of James Wilson, gene therapy pioneer - C&EN
    Sep 12, 2019 · On Sept. 17, 1999, Jesse Gelsinger died after receiving an experimental gene therapy from James Wilson's lab at the University of Pennsylvania.
  163. [163]
    Gene Therapy for SCID-X1: Round 2 - PMC
    Nov 1, 2010 · However, the approach experienced a major setback when 5 of the 20 boys developed leukemia due to insertional mutagenesis and activation of an ...
  164. [164]
    A tragic setback | Nature
    Nov 14, 2002 · But if it, too, shows a penchant for inserting into genes, it will mean that the risks associated with SCID gene therapy are greater than was ...
  165. [165]
    Gene therapy then and now: A look back at changes in the field over ...
    May 7, 2025 · These viral vectors were tested in several early clinical trials targeting cancer and genetic diseases, with some showing initial signs of ...Review · Vectors For Gene Therapy... · Disease And Tissue Targeting...
  166. [166]
    Viral Vector Based Cell & Gene Therapies Approval - Exothera
    Timeline of viral vector-based Cell & Gene therapy approvals. With the approval of the ABECMA therapy last March, there are now 17 cell and gene therapy ...
  167. [167]
    Viral-vector therapies at scale: Today's challenges and future ...
    Mar 29, 2022 · Viral-vector gene therapies are here to stay. Keeping pace with increasing demand requires consideration of challenges, the potential for standardization, and ...
  168. [168]
    Gene Therapy at 30: Milestones, Setbacks, and a Path Forward
    Apr 14, 2025 · This was traced to insertional mutagenesis, a phenomenon in which the inserted genetic material disrupted normal cellular function, contributing ...
  169. [169]
    [PDF] NIH Guidelines for Research Involving Recombinant or Synthetic ...
    NIH OFFICE OF SCIENCE POLICY CONTACT INFORMATION:​​ These NIH Guidelines shall supersede all earlier versions until further notice.
  170. [170]
    [PDF] Biosafety in Microbiological and Biomedical Laboratories—6th Edition
    The core principle of this document is protocol-driven risk assessment; it is not possible for a single document to identify all of the possible combinations ...
  171. [171]
    [PDF] Biosafety Considerations for Research with Lentiviral Vectors
    Because the NIH Guidelines for Research. Involving Recombinant DNA Molecules (NIH Guidelines) do not explicitly address containment for research with lentiviral ...
  172. [172]
    [PDF] EHS-415 Guidelines For Working With Viral Vectors - Emory EHSO
    Sep 12, 2024 · These guidelines are based on The National Institutes of Health (NIH) Office of Science Policy. (OSP) Biosafety Considerations for Research with ...
  173. [173]
    Biosafety Containment: Adeno-Associated Virus - UCSD Blink
    Feb 13, 2024 · Adeno-associated virus and AAV vectors may be handled at Biosafety Level 1 (BSL-1) and Animal Biosafety Level 1 (ABSL-1) when they are: Made in ...
  174. [174]
    Biological Safety Manual - Chapter 08 - UNC Policies
    Animal Biosafety Containment​​ Adenoviral vector must be administered under ABSL-2 containment with the use of a BSC. Animals may shed/excrete adenovirus post- ...
  175. [175]
    [PDF] Guidance for Working with Viral Vectors - San Jose State University
    Nov 9, 2020 · Suggested biosafety containment levels and practices are provided in this document. A higher-containment level may be required in specific cases ...<|separator|>
  176. [176]
    [PDF] Risks Associated With Lentiviral Vector Exposures and Prevention ...
    In this paper, we review the potential risks associated with LVV exposure and provide recommendations for principal investigators (PIs), Institutional Biosafety ...
  177. [177]
    [PDF] Clinical Development of Gene Therapy Products - FDA
    Dec 7, 2022 · Approval of gene therapy products must be based on substantial evidence of effectiveness and sufficient evidence of safety.Missing: process | Show results with:process
  178. [178]
    Immunogenicity of Gene Therapy Products - FDA
    Apr 22, 2024 · The first two gene therapy products approved by the FDA used AAV2 and AAV9 vectors for gene delivery.
  179. [179]
    [PDF] guideline-quality-non-clinical-and-clinical-aspects-gene-therapy ...
    Mar 22, 2018 · This guideline addresses the requirement for a marketing authorisation application (MAA) of non-cell based GTMPs, which fall broadly into 3 ...
  180. [180]
    Guidelines relevant for advanced therapy medicinal products
    The European Medicines Agency develops scientific guidelines to help pharmaceutical companies and individuals to prepare marketing-authorisation applications ...
  181. [181]
    Quality, non-clinical and clinical issues relating specifically to ...
    This document aims to discuss quality, non-clinical and clinical issues that should be considered during the development of medicinal products derived from ...Missing: process | Show results with:process
  182. [182]
    The Fast Approval and Slow Rollout of Sputnik V: Why Is Russia's ...
    While Russia was the first country to approve a COVID-19 vaccine without limiting conditions—Sputnik V on 11 August 2020—the vaccine rollout in Russia has been ...
  183. [183]
    Sputnik V: WHO suspends approval process for COVID vaccine due ...
    Sep 16, 2021 · The World Health Organization (WHO) has suspended the approval process for Russia's Sputnik V COVID vaccine. A regional WHO official said ...
  184. [184]
    Global regulatory progress in delivering on the promise of gene ...
    This article summarizes key global regulatory challenges to the development of gene therapies and provides recommendations for solutions to overcome them.
  185. [185]
    Comparative analysis of GMO regulatory requirements for AAV ...
    1. Gene therapies are innovative treatments involving various technologies, such as viral vector gene delivery, genome editing, and genetically modified ...<|separator|>
  186. [186]
    Death but one unintended consequence of gene-therapy trial - PMC
    the gene's delivery vehicle — and died 4 days after receiving the injection. The major question ...
  187. [187]
    Gene Therapy Research & the Case of Jesse Gelsinger
    But he had a negative reaction to the injection, and four days later, on September 17, 1999, he died. ... However, as the investigation into Gelsinger's death ...
  188. [188]
    What are the ethical issues surrounding gene therapy? - MedlinePlus
    Feb 28, 2022 · Because gene therapy involves making changes to the body's basic building blocks (DNA), it raises many unique ethical concerns.
  189. [189]
    Ethical Issues: Germline Gene Editing | ASGCT
    Feb 3, 2025 · By discussing and understanding ethical issues, patients and their families can engage in informed, shared decision-making with the medical and ...
  190. [190]
    Bioethical issues in genome editing by CRISPR-Cas9 technology
    Germline genome editing leads to serial bioethical issues, such as the occurrence of undesirable changes in the genome, from whom and how informed consent is ...
  191. [191]
    10 Scientific and ethical considerations in human gene therapy
    ... ethical issues of human gene therapy ... therapy: Inefficient gene expression in dogs reconstituted with autologous marrow infected with retroviral vectors.
  192. [192]
    Ethical Challenges and Controversies in the Practice and ...
    Aug 24, 2022 · This review discusses in detail the current challenges surrounding the practice of gene therapy, focusing on the moral arguments and scientific claimsIntroduction · The Approach of Gene Therapy · Ethical Challenges of Gene...
  193. [193]
    COVID‐19 vaccine research and development: ethical issues - PMC
    This narrative review highlights ethical issues in COVID‐19 vaccine research and development that every stakeholder needs to be aware of and to consider.
  194. [194]
    Asilomar and recombinant DNA - NobelPrize.org
    Aug 26, 2004 · This unique conference marked the beginning of an exceptional era for science and for the public discussion of science policy.Missing: vectors | Show results with:vectors<|control11|><|separator|>
  195. [195]
    AI-Driven AAV Capsid Creation for Gene Therapy Advances
    Oct 17, 2025 · Starting at Harvard in 2015, the authors set out to overcome the limitations of current capsids by developing new machine-guided technologies to ...
  196. [196]
    Advances in AAV capsid engineering: Integrating rational design ...
    May 7, 2025 · These strategies have yielded novel capsids with improved transduction efficiency, reduced immunogenicity, and enhanced tissue targeting.
  197. [197]
    AAV Capsid Development - AAVnerGene
    Engineering novel AAV capsids, such as AAV-ShD, for use in non-human primates (NHPs) and potential human applications. These innovations are powered by our ...
  198. [198]
    Lentiviral Vector Guide - Addgene
    Lentiviral vectors can target non-dividing cells, enable stable long-term expression in a host, and exhibit low immunogenicity.Missing: properties | Show results with:properties
  199. [199]
    An Improved Protocol for the Production of Lentiviral Vectors
    Dec 18, 2020 · Importantly, this modification greatly reduced the risk of replication-competent viruses (RCVs), as packaging and envelope cassettes of the ...
  200. [200]
    Hypoxia and HIF-1 inhibition enhance lentiviral transduction efficiency
    May 10, 2025 · These advancements have established lentiviral vectors as a safe and efficient tool for stable transgene delivery [5]. Today, lentiviral vectors ...
  201. [201]
    Role of homologous recombination/recombineering on human ...
    This review summarizes recent advancements in the use of human adenoviral vectors in medicine and discusses the pivotal role of recombineering in the ...
  202. [202]
    Construction and application of adenoviral vectors - Cell Press
    Sep 7, 2023 · Currently, adenoviral vector-based vaccines have been developed to cover a wide range of infectious agents from malaria to HIV-1. As of March ...
  203. [203]
    Cell & Gene Therapy Clinical Trials: Current Dynamics & Pipeline ...
    Gene Therapy Pipeline Analysis: According to the Gene, Cell, & RNA Therapy Landscape Report mentioned in April 2025, there were 2,154 gene therapies including ...<|separator|>
  204. [204]
    AAV Vectors In Gene Therapy Pipeline Outlook 2025: Clinical Trial ...
    Aug 29, 2025 · DelveInsight's AAV Vectors in Gene Therapy pipeline report depicts a robust space with 180+ active players working to develop 250+ pipeline ...
  205. [205]
    [PDF] Gene, Cell, & RNA Therapy Landscape Report - ASGCT
    Sep 30, 2025 · Welcome to 2025's second quarterly report from ASGCT, Citeline, and Evaluate! This past quarter, three new therapeutics were approved, ...
  206. [206]
    Current clinical applications of AAV-mediated gene therapy
    May 5, 2025 · Currently, there are two FDA-approved products for congenital neuromuscular disorders: onasemnogene abeparvovec (Zolgensma, full approval in ...<|control11|><|separator|>
  207. [207]
    Gene & Cell Therapy News - ASGCT
    FDA Approves SKYSONA, Second Lentiviral Vector Gene Therapy in the U.S. The approval of SKYSONA® comes one month after the first lentiviral vector gene therapy ...
  208. [208]
    Viral and non-viral vectors in gene therapy: current state and clinical ...
    Advancements in gene therapy have achieved significant milestones in treating human diseases, offering renewed hope to patients with limited options.
  209. [209]
  210. [210]
    Viral Vector & Plasmid DNA Manufacturing - MarketsandMarkets
    Sep 26, 2025 · The global viral vector manufacturing market, valued at US$4.8 billion in 2022, stood at US$5.5 billion in 2023 and is projected to advance at a ...
  211. [211]
    Viral Vector Manufacturing Market Size, Share & Growth, Drivers ...
    Aug 28, 2025 · By geography, North America commanded 47.34% of 2024 revenue and Asia-Pacific is projected to grow at 22.56% CAGR between 2025 and 2030. Global ...
  212. [212]
    Viral Vector Development Market to Reach USD 5.00 Billion at 18.84 ...
    Aug 19, 2025 · ➢ In May 2025, Coave Therapeutics, a biotechnology company, launched a CNS-targeted gene therapy capsid developed with its proprietary platform.
  213. [213]
    Gene Therapy Studies Rose While Financing Declined In Q2 2025
    Aug 13, 2025 · In Q2 2025, progress in advanced therapeutics was marked by three notable approvals and continued growth across the gene therapy landscape.
  214. [214]
    Progress and problems with the use of viral vectors for gene therapy
    May 1, 2003 · An important obstacle to the use of adenovirus vectors has been their immunogenicity. Highly disabled gene-deleted adenovirus vectors have been ...
  215. [215]
    Restricting use of adenovirus vector-based COVID vaccines ... - NIH
    Aug 9, 2022 · While concerning, the risk of TTS is extremely low and the restriction of Ad vector-based vaccines is premature, as our understanding of COVID - ...
  216. [216]
    Viral Vectors in Gene Therapy: Advantages & Disadvantages
    Retroviral Vectors: These vectors integrate into the host genome through reverse transcriptase. They are widely used to deliver target or therapeutic genes ...Missing: definition | Show results with:definition
  217. [217]
    Progress and challenges in viral vector manufacturing - PMC - NIH
    The most compelling achievement in gene therapy for common life-threatening diseases are the dramatic clinical results obtained by several independent teams ...
  218. [218]
  219. [219]
    Emerging non-viral vectors for gene delivery
    Aug 17, 2023 · This review comprehensively outlines the novel fastest-growing and efficient non-viral gene delivery vectors, which include liposomes and lipid nanoparticles ( ...
  220. [220]
    Physical non-viral gene delivery methods for tissue engineering - NIH
    This review examines the advantages and disadvantages of physical non-viral methods (ie, microinjection, ballistic gene delivery, electroporation, sonoporation ...
  221. [221]
    Nanotechnology-based non-viral vectors for gene delivery in ...
    Jan 17, 2024 · Non-viral nanocarriers generally include polymers, liposomes, lipid carriers and inorganic materials (Gu et al., 2020). Non-viral nanocarriers ...
  222. [222]
    The Delivery Dilemma Looking Beyond Viral Vectors for Gene Therapy
    Oct 15, 2024 · They play a crucial role in packaging, protecting, stabilizing, and delivering gene-altering materials into cells (1, 2). That said, other ...
  223. [223]
    Nonviral Gene Delivery: Principle, Limitations, and Recent Progress
    The nonviral gene delivery methods, on the other hand, use synthetic or natural compounds or physical forces to deliver a piece of DNA into a cell. The ...