null
The Lenti Viral Vector System: A New Way to Deliver Genetic Therapies

The Lenti Viral Vector System: A New Way to Deliver Genetic Therapies

A new way to deliver genetic therapies is making waves in the medical world. The lenti virus vector system has been shown to be more effective than traditional methods of gene therapy delivery. This system uses a virus to carry the therapeutic genes into the cells of the patient's body. Researchers have found that this method results in fewer side effects and higher success rates. In this blog post, we will discuss the lenti virus vector system and its benefits for patients with genetic disorders.

Lentiviruses

The lentivirus genus includes retroviruses that cause persistent disease with long incubation times in people and other mammal species. The genus includes human immunodeficiency virus (HIV), that causes AIDS. Lentiviruses integrate a huge amount of viral complementary DNA into host cell's DNA, allowing them to readily infect non-dividing cells, making them one of the most efficient gene transporters. The term 'endogenous retrovirus' refers to a virus that has integrated its DNA into the host germline genome, allowing future host descendants to inherit it.

Lentivirus Life Cycle

the lentiviral life cycle involves a series of steps that enable the virus to enter target cells, deliver its genetic material, and replicate. Here's an overview of the main stages in the lentiviral life cycle:

1. Attachment and Entry: The process begins with the attachment of the lentivirus to the target cell's surface. The viral envelope glycoprotein (usually VSV-G or other modified proteins) interacts with specific receptors on the cell membrane. This attachment triggers a conformational change in the envelope protein, exposing a fusion peptide. This allows the virus to fuse its envelope with the cell membrane, releasing the viral core (containing the RNA genome and essential proteins) into the cytoplasm of the target cell.

2. Reverse Transcription: Once inside the cell, the viral core carries out reverse transcription. The viral enzyme reverse transcriptase converts the single-stranded RNA genome of the virus into a double-stranded DNA intermediate. This viral DNA, known as the proviral DNA, integrates into the host cell's chromosomal DNA with the help of the viral enzyme integrase.

3. Integration: The proviral DNA integrates into the host cell's genome at a semi-random site. This integration is catalyzed by integrase and is essential for the virus to establish a persistent infection. Integration can lead to the insertion of viral DNA near cellular genes, potentially influencing their function.

4. Transcription and Translation: Integrated proviral DNA serves as a template for transcription by the host cell's RNA polymerase II. Viral RNA transcripts are then processed and translated to produce viral structural and enzymatic proteins. These proteins play a role in the assembly of new virus particles.

5. Assembly and Budding: Newly synthesized viral RNA, along with the structural and enzymatic proteins, are transported to the cell surface. Here, they assemble at the plasma membrane to form new virus particles. The viral core is enveloped by the host cell membrane, acquiring the viral envelope proteins as it buds from the cell surface. This process allows the virus to escape from the host cell without necessarily destroying it.

6. Maturation and Release: During or shortly after budding, the newly formed virus particles undergo maturation, where protease enzymes cleave the precursor proteins into their mature forms. This step is crucial for the infectivity of the released viruses. The mature lentivirus is then released from the host cell and is free to infect other cells, starting the cycle anew.

Lentiviral Vectors

Lentiviral vectors are powerful tools in the field of gene delivery, widely used for their ability to efficiently introduce genetic material into a broad range of dividing and non-dividing cells

The components required for virus production are divided across many plasmids (3 for second-generation systems, 4 for third-generation systems) in order to boost lentivirus safety. To improve safety, first-generation HIV-1 lentiviral vectors split the vector components into three plasmids: (i) a packaging construct; (ii) an Env plasmid containing a viral glycoprotein; and (iii) a transfer vector genome construction.

To reduce the formation of RCLs (replication competent lentiviruses), the packaging and enveloping plasmids have been specifically designed without a packing signal or LTRs. The third-generation system effectively reduced the formation of RCLs and improved biosafety.

The following is a list of the components of both systems:

  • Lentiviral transfer plasmid (containing gene of your interest): The donor sequence is flanked by long terminal repeats (LTRs), which aid in the integration of the transfer plasmid DNA into the host genome. It is generally the sequences between and including the LTRs that are incorporated into the host genome following viral infection. Transfer plasmids are all replication incompetent and may contain an additional deletion in the 3'LTR, making the virus "self-inactivating" (SIN) after integration for safety reasons.
  • Packaging plasmid : This specialized plasmid contains key viral genes necessary for packaging and encapsidating the lentiviral genome into viral particles. Typically, the packaging plasmid carries genes encoding the structural proteins Gag and Pol, which are crucial for the formation of viral cores and the enzymatic processes required for reverse transcription and integration. Additionally, the packaging plasmid often contains the Rev gene, responsible for regulating the nuclear export of viral RNA transcripts. Through a carefully orchestrated interplay of these genes, the packaging plasmid ensures that the lentiviral vector's genetic cargo is accurately and efficiently assembled into new virus particles.
  • Envelope plasmid : This specialized plasmid contains the gene encoding the viral envelope glycoprotein, which determines the host cell specificity and entry mechanism of the lentivirus. By providing the necessary envelope protein, often derived from sources like vesicular stomatitis virus (VSV-G), the envelope plasmid enables the pseudotyping of lentiviral particles. Pseudotyping involves replacing the native envelope protein with the one encoded by the envelope plasmid, expanding the range of cell types the lentivirus can infect. This process is vital for achieving efficient and specific transduction of the desired target cells.

Adding gene packaging and envelope plasmids are generalized and appropriate for varied cell types and systems. When planning your experiment, the important component to consider and optimize is the transfer plasmid. 2nd generation lentiviral plasmids utilize the viral LTR promoter for gene expression, whereas 3rd-generation transfer plasmids utilize a hybrid LTR promoter. Additional or specialized promoters may also be included within a transfer plasmid: for example, the U6 promoter is included in the pSico plasmid to drive shRNA expression. Other features like fluorescent fusions or reporters can also be included in transfer plasmids.

The goal of the using three variants of Lentiviral gene transfer is to provide researchers with an alternative method for efficient production in vitro or animal models. New possibilities for human genomics research opened up in 2012 with the discovery of the lentivirus. Researchers are attempting to utilize lentiviruses to silence a specific gene using RNA interference technology in high-throughput methods, resulting in significant improvements in efficiency and cost effectiveness.

Lentiviral Gene Therapy

The lenti virus vector system is a new way to deliver genetic therapies. This system uses a lentivirus to carry the therapeutic genes into the cells of the patient's body. Lentiviruses are known for their ability to infect a wide range of cells, making them an ideal choice for gene delivery. In addition, lentiviruses are highly stable and can remain in the body for long periods of time. This makes them an excellent choice for patients with chronic diseases. This feature of lentivirus vectors to mediate potent transduction and stable expression into dividing and non-dividing cells both in vitro and in vivo is extremely beneficial.

One of the benefits of using lenti viruses as vectors is that they are very safe. They also have a low risk of causing adverse reactions in patients. In addition, lentiviruses can be easily eliminated from the body if necessary.

Lentiviral System - Applications

  • The use of a lentivirus to introduce a new gene into human or animal cells is another popular technique. For example, the model of mouse hemophilia expressing wild-type platelet-factor VIII, the defective gene in humans can be corrected using this method. Because lentiviral infection can efficiently infect dividing and non-dividing cells, it offers several advantages over other gene therapy techniques like long time expression of transgene and low immunogenicity. In addition, lenti viruses can target specific cells in the body, ensuring that the therapeutic genes reach their intended destination.
  • Lentiviruses have been also used to elicit an immune response against tumor antigens. Gammaretroviruses and lentiviruses have already been used in more than 300 clinical trials, with applications ranging from therapeutic options for various illnesses to vaccines.
  • The lenti virus vector system has several advantages for people who suffer from genetic conditions. This method is both safe and effective. In addition, lentiviruses can be utilized to deliver a variety of therapeutic genes.
  • These vectors are highly efficient in many species. DNA microinjection was completely ineffective in chickens. The foreign DNA in these vectors is limited to 8 kB. To produce a high amount of transgenic animals with a limited number of unique integrated copies, the quantity of vector used must be changed. The lentiviral vectors and the transgene are silenced together. Each integrated copy is different. The insulator 5′ HS4 reduces the silencing effect. The viral promoter typically determines the expression of lentiviral vectors. The lentiviral vectors are ideal for producing siRNAs. These vectors are commercially available, and the banks include vectors containing siRNA genes that target both human and mouse mRNAs.

Lenti viral vectors as gene transfer tool to CNS

The lentiviral vectors have been utilized as gene transfer vehicles for the brain since they transduce most cell types in the brain, resulting in robust and long-lasting gene expression.

In comparison to gamma-retroviruses, lentiviral vectors have been discovered to be less hazardous.

Several research have shown that lentiviral vectors can efficiently transduce most cell types in the CNS, including terminally differentiated neurons, dendritic cells, glial cells, astrocytes, and oligodendrocytes in vivo.

Although other varieties of lentiviral vectors, such as SIV, EIAV, and FIV, have been shown to send transgenes into the brain, HIV-1-based vectors have proven to be the most successful and efficient.

Lentiviral Gene Therapy - Potential Risks

While lentiviral vectors offer numerous benefits for gene therapy, there are also certain risks associated with their use. These risks primarily stem from the vector's viral origins and its potential interactions with the host organism. Some key risks include:

1. Insertional Mutagenesis: Lentiviral vectors integrate their genetic cargo into the host cell's genome. In some cases, this integration can disrupt normal cellular gene function, potentially leading to insertional mutagenesis and causing unintended consequences, such as oncogene activation or tumor formation.

2. Immunogenicity: Although lentiviral vectors are generally less immunogenic than some other viral vectors, they can still trigger an immune response in the host. This immune response could limit the effectiveness of the gene therapy or lead to adverse reactions.

3. Limited Control of Expression: While lentiviral vectors offer stable and long-term gene expression, controlling the level and timing of expression can be challenging. Overexpression or underexpression of the therapeutic gene could lead to unwanted effects or suboptimal treatment outcomes.

4. Off-Target Effects: Lentiviral vectors might inadvertently integrate into unintended genomic sites, potentially affecting the function of nearby genes. This could result in unforeseen biological consequences or safety issues.

5. Risk of Vector Mobilization: In some cases, residual viral elements in the lentiviral vector system could lead to the generation of replication-competent lentiviruses (RCLs), which have the potential to replicate and cause harm. Rigorous quality control and safety measures are necessary to mitigate this risk.

6. Inflammatory Responses: While lentiviral vectors are generally considered less inflammatory than other viral vectors, they can still trigger mild immune responses. These responses could lead to inflammation at the injection site or in the surrounding tissues.

7. Ethical Considerations: The use of lentiviral vectors in gene therapy raises ethical questions, especially when considering germline editing or permanent genetic changes that could be passed on to future generations.

8. Tropism Limitations: Lentiviral vectors might not efficiently transduce all cell types, which could limit their applicability for certain diseases or therapies that require specific cell targeting.

9. Manufacturing Challenges: The production of high-quality lentiviral vectors for clinical use can be complex and costly. Ensuring consistent vector production and purification processes is crucial for safety and efficacy.

10. Regulatory Considerations: The use of lentiviral vectors for gene therapy is subject to regulatory oversight, and the approval process can be stringent and time-consuming. Navigating regulatory requirements is an important aspect of the development and implementation of lentiviral-based therapies.

To mitigate these risks, thorough preclinical studies, robust safety assessments, and careful patient monitoring are essential. Researchers and clinicians must continually work to improve the safety profile of lentiviral-based gene therapies and address potential challenges to maximize their benefits while minimizing potential risks.

18th Aug 2023 Meghana Menon, Msc

Recent Posts