Pages
Products

Lentivirus Service

Lentivirus OverviewResearch-Grade ServicesCDMO ServicesContact UsFAQ

Lentivirus Overview

Lentiviruses are a subgroup of retroviruses with an RNA genome that is reverse-transcribed into DNA after infection. Their genome includes gag (structural proteins), pol (reverse transcriptase and integrase), and env (envelope glycoproteins). Lentiviruses enter host cells via membrane fusion or endocytosis, reverse-transcribe their RNA into DNA, and integrate into the host genome—preferably within transcriptionally active regions. Unlike γ-retroviruses (e.g., MLV), lentiviruses can infect non-dividing cells, making them valuable vectors for gene therapy, especially for quiescent cells such as HSCs and resting T cells. Lentiviral vector systems have evolved through three generations to improve safety.

Structure of Lentivirus.

What Are the Elements Involved in Lentiviral Vector Structure and Function?

Key Components

Accessory & Regulatory Genes

AbbreviationFull NameFunction
gagGroup-Specific AntigenEncodes structural proteins of the virus
MAMatrixViral matrix protein
CACapsidViral capsid protein
NCNucleocapsidViral nucleocapsid protein
polPolymeraseEncodes enzymes required for reverse transcription and integration
PRProteaseProteolytic cleavage of viral protein precursors
RTReverse TranscriptaseConverts RNA genome into DNA
INIntegraseIntegrates viral DNA into host genome
envEnvelopeEncodes envelope glycoproteins
VSV-GVesicular Stomatitis Virus GEnvelope protein used for pseudotyping; enables broad host tropism
LTRLong Terminal RepeatContains essential elements for transcription, reverse transcription, and integration
Ψ (psi)Packaging SignalRequired for packaging of viral RNA into the capsid
U3Unique 3′ regionPromoter/enhancer region; required for transcription and reverse transcription (initiation)
U5Unique 5′ regionRequired for reverse transcription (extension)
RRepeat regionFound at both ends; essential during reverse transcription
AbbreviationFull NameFunction
VifViral Infectivity FactorEnhances viral replication, counteracts host defenses
VprViral Protein RInvolved in nuclear import, immune evasion
VpuViral Protein UEnhances replication, downregulates CD4
RRERev Response ElementRev binding site, facilitates nuclear export of RNA
RevRegulator of VirionNuclear export of unspliced and singly-spliced viral RNAs
TatTrans-Activator of TranscriptionEnhances transcription of viral genes
polyApoly-Adenine tailImproves mRNA stability, export, and translation

Advantages of Lentiviral Vectors

  • High Transduction Efficiency: Lentiviral vectors can more effectively transduce non-proliferating or slowly proliferating cells, such as CD34+ hematopoietic stem cells (HSCs).
  • Good Safety Profile: Over years of clinical application, no serious adverse events related to the vector have been reported. Integration site analysis shows a predictable integration pattern within transcribed genes, and the distribution of integration sites remains stable.
  • Long-Term Expression: Lentiviral vector-modified T-cells can persist for over 5 years after treatment, potentially offering lifelong effects.

Clinical Applications

With the rapid development of gene therapy technologies, lentiviral vectors have found various important clinical applications due to their unique advantages. These include the correction of primary immunodeficiencies, engineered tumor-specific T-cell receptors (TCRs), and chimeric antigen receptor (CAR) T-cell therapy. Below are partial gene therapies that utilize lentiviral vectors:

CompanyTherapyIndicationClinical Progress & Efficacy
Bluebird BioZyntegloTransfusion-dependent β-thalassemiaFDA approved in 2022, patients are free from transfusion dependency, long-term follow-up shows hemoglobin near normal levels
Bluebird BioSKYSONAEarly active cerebral adrenoleukodystrophy (CALD)FDA approved in 2022, 90.6% of patients had no severe disability within 24 months
Oxford BiomedicaAxo-Lenti-PDParkinson's diseaseOngoing clinical trials, improvement in neurological function
Orchard TherapeuticsLibmeldy (OTL-200)Metachromatic leukodystrophy (MLD)EMA approved in 2020, only approved MLD therapy, significant improvements in motor and cognitive functions
Orchard TherapeuticsStrimvelisADA-SCIDEMA approved in 2016, first ex vivo autologous gene therapy
Mustang BioMB-107/MB-207X-linked severe combined immunodeficiency (XSCID)Phase 1/2 trials show significant immune reconstitution
AVROBIOAVR-RD-02Gaucher diseasePhase 1/2 trials ongoing
Rocket PharmaceuticalsRP-L201Leukocyte adhesion deficiency type I (LAD-I)Clinical trials show good safety and efficacy
Interius BioTherapeuticsINT2104B-cell malignanciesApproved for Phase I clinical trial by Australian TGA in July 2024, single-dose intravenous infusion, no lymphodepletion required
Umoja BiopharmaUB-VV111Hematologic malignanciesFDA IND approval in August 2024, Phase I trial initiation, including CAR-T-treated patients
Kelonia Therapeuticsanti-BCMA CAR iGPSMultiple myelomaPreclinical data superior to in vitro culture, good targeting capabilities

Lentivirus Research-Grade Services

As interest in gene and cell therapies accelerates, the demand for scalable, cost-effective lentiviral vector manufacturing platforms has intensified. With advancements in drug characterization and safety analysis technologies, lentiviral vector-based gene therapies are now widely used in late-stage clinical trials and therapies approved by regulatory agencies.

Creative Biogene is dedicated to propelling engineered cell and gene therapies forward through cutting-edge solutions. Our comprehensive end-to-end lentiviral workflows deliver exceptional products, services, and specialized expertise to empower the development of transformative lentiviral gene therapies.

Comprehensive Custom Cloning Services

Creative Biogene delivers expert custom cloning services into lentiviral backbones, accommodating diverse genetic elements including ORFs, shRNA, miRNA, sgRNA, lncRNA, and circRNA—all with versatile tagging options. Every lentiviral construct undergoes rigorous verification through gold-standard Sanger sequencing, guaranteeing perfect alignment with NCBI reference sequences.

Inventory Highlights:

  • Over 100,000 human, mouse, and rat ORF plasmids
  • More than 10,000 validated shRNA plasmids
  • 4,000+ miRNA overexpression/inhibition plasmids
  • 1,000+ promoter-driven plasmids

Creative Biogene Lentiviral Vector Types

Creative Biogene offers a diverse portfolio of lentiviral vector systems tailored for specific research and clinical applications:

Integrating Lentivirus (ILV)

Enables long-term gene expression through host genome integration, ideal for stable cell lines and transgenic models. Features high-titer production (>1×10⁹ TU/mL), broad host compatibility via VSV-G pseudotyping, and a complete workflow from gene synthesis to QC.

Non-Integrating Lentivirus (IDLV)

Provides transient expression without genome integration risk, achieving ultra-high titers (1×10¹⁰ TU/mL) suitable for in vivo applications while meeting FDA safety guidelines for gene therapy vectors.

Custom CAR Lentivirus

Specifically designed for CAR-T therapy applications with T cell-optimized vectors, serum-free processing for clinical-grade production, and comprehensive quality control measures for regulatory compliance.

shRNA Lentivirus

Delivers stable gene knockdown for functional genomics research using algorithm-optimized shRNA design with validated silencing efficiency ≥70% and compatibility with both in vitro and in vivo applications.

Pseudotyped Lentivirus

Expands targeting capabilities through alternative envelope proteins, offering tissue-specific targeting options and specialized controls for experimental validation.

Reporter-Embedded Lentivirus

Integrates fluorescent or secreted reporters for real-time monitoring, pathway-specific response systems, and pre-built libraries for applications requiring visual tracking or high-throughput screening.

One-Stop Lentiviral Production Solutions

Creative Biogene employs an advanced third-generation packaging system to produce lentivirus in both standardized and customized formats. Our sophisticated in-house production infrastructure combines premium reagents, streamlined quality-controlled workflows, and an expert R&D team—delivering consistently reliable, high-titer lentiviral particles for cutting-edge research and therapeutic applications.

Distinguished Advantages:

  • Exceptional Quality: Every batch undergoes mycoplasma testing and infectivity validation
  • Superior Titers: Standard concentrations exceed 1×10⁸ TU/mL, with custom options up to 1×10⁹ TU/mL
  • Minimal Toxicity: Serum-free, animal-origin-free production minimizes immunogenic risks
  • Enhanced Transduction: Optimized for challenging cell types and in vivo applications
  • Rapid Availability: Ready-to-use lentivirus shipped within 1–3 days
  • Optimized Packaging: Available in small aliquots to preserve viral integrity
  • Large Capacity: Specialized capability for packaging ORFs exceeding 4 kb

Service Process

How do We Support Your LV Production for Research?

Product SpecsDelivery StandardApplicationTurnaround TimePrice
Standard Virus Specification1×10⁸ TUFor Cell Transduction2-3 WeeksInquiry
Large Packaging Virus1×10⁹ TUFor Cell Transduction2-3 WeeksInquiry
Purified Virus1×10⁹ TUFor In Vivo Injection2-3 WeeksInquiry

Lentivirus CDMO Services

With the rise of the cell therapy industry, the demand for lentivirus has been growing yearly. Lentiviral vector production for clinical use must comply with cGMP to ensure high quality, with verified identity, purity, and potency. Producing cGMP-compliant lentiviral vectors is more challenging than retroviral vectors due to the lack of stable packaging cell lines, leading to production inconsistencies and higher costs.

Creative Biogene focuses on providing comprehensive CDMO services for cell therapy drugs. We have established an advanced GMP-grade serum-free suspension virus production platform, offering high-quality lentivirus CDMO services tailored to our client's diverse needs.

Creative Biogene's CDMO Service Flow

Preclinical

Plasmid construction
Pseudotype selection
Small-scale LVV production
In vitro poc
Basic assays

IND Filing

GLP tox batch
Assay development
IND CMC documentation
DMF reference

Clinical (Phase I–III)

Scale-up
Process optimization
QC testing
Method validation
Batch release

Commercial

Routine GMP production
CPV
Lifecycle management
Long-term supply

BLA/NDA Submission

PPQ runs
Comparability studies
CMC dossier finalization
Regulatory response

Customization

Regulatory Compliance

End-to-End

Clinical Integration

Lentiviral Vector Manufacturing

Creative Biogene's manufacturing infrastructure features GMP-compliant dedicated cell bank establishment areas, aseptic isolators for final product filling, and optimized processes that significantly reduce production and testing costs. Both the adherent and suspension platforms are supported by comprehensive documentation and traceability systems that fully satisfy regulatory requirements for clinical and commercial applications, ensuring a seamless transition from development to market approval.

We offer two robust lentiviral production systems with FDA DMF registration and clear IP traceability:

Adherent Production Platform

  • Utilizes industry-validated HEK293T cell line
  • Supports stable production at 50L scale
  • Complete timeline from cell bank construction to GMP batch delivery in just 6 months

Suspension Production Platform

  • Supports 200L scale production with 60% recovery rates
  • Achieves titers up to 10⁹ TU/ml
  • Significantly reduces scale-up production costs
  • Animal-free, serum-free culture technology enhancing safety and consistency
  • Linearly scalable to meet various production requirements
  • Employs 50L single-use bioreactors to increase efficiency and reduce contamination risks

How do We Support Your LV Production at Every Stage?

Service LevelTypical TiterProduction scaleProduction DetailsPrice
IIT Grade1×10⁹ TU2–50L
  • Customizable yield and quality standards
  • Authentic and traceable records
  • GMP-like quality management system
Inquiry
IND Grade1×10⁹ TU2–50L
  • Customizable cell bank size
  • Bank stability studies
Inquiry
Clinical Grade1×10⁹ TU2–200L
  • Full GMP facility with segregated virus/non-virus zones
  • Dedicated bank construction area
  • Comprehensive GMP quality system
  • Long-term and accelerated stability studies
  • Facility and equipment validation per clinical registration standards
Inquiry

QC Testing Items

Test ItemTest MethodUS/EU Applicable Regulations
Adventitious Viral AgentsIndicator Cell CultureUSP<1237>, EP 2.6.16, FDA Points to Consider
Replication-Competent LentivirusCo-cultivation MethodUSP<63>, EP 5.14, FDA Guidance for Industry
AppearanceVisual InspectionUSP<1790>, EP 2.2.1
SterilityMembrane FiltrationUSP<71>, EP 2.6.1, 21 CFR 610.12
MycoplasmaCulture/PCR MethodUSP<63>, EP 2.6.7, 21 CFR 610.30
pHpH MeterUSP<791>, EP 2.2.3
OsmolalityFreezing Point DepressionUSP<785>, EP 2.2.35
TCR IdentificationSequencingUSP<1120>, ICH Q6B
Host Cell ProteinELISAUSP<1132>, EP 2.6.34, ICH Q6B
Physical Titer (p24)ELISAUSP<1235>, ASTM F2888
Transduction TiterFlow CytometryUSP<1027>, ICH Q2(R1)
Bacterial EndotoxinLAL TestUSP<85>, EP 2.6.14, 21 CFR 610.9
Benzonase ResidueELISAUSP<1132>, EP 2.6.34
Residual Host DNAq-PCR MethodUSP<1130>, EP 2.6.35, ICH Q6B
E1A Transfer ResidueCo-cultivation MethodUSP<1237>, EP 5.14
SV40 Transfer ResidueCo-cultivation MethodUSP<1237>, EP 5.14, FDA Points to Consider

Clinical Integration

  • Experience in multi-product concurrent production, capable of simultaneously handling pilot samples and registration batches for different clients
  • Coverage of critical process validations such as T-cell transduction, directly addressing clinical efficacy data requirements

Contact Us

In the field of cell and gene therapy, stable production and compliant delivery of lentiviral vectors are key to the successful implementation of therapies. Drawing on over a decade of technical expertise, Creative Biogene has developed a practical and accessible lentiviral CDMO service platform that focuses on solving real challenges for clinical-stage clients—from process development to GMP production. Contact us today to learn more or request a quote.

FAQs

Q1: How to choose between ILV and IDLV?

  • ILV: Ideal for long-term expression (e.g., stable cell lines, CAR-T therapy).
  • IDLV: Suitable for transient expression or applications requiring minimal genome integration risks (e.g., gene editing, iPSC reprogramming).

Q2: How is viral titer determined?

  • We use qPCR (vector copy number) and flow cytometry (reporter gene expression) for dual-validation, ensuring accuracy.

Q3: My infection efficiency is low. What factors could be affecting this, and how can I improve it?

Several factors can impact lentiviral infection efficiency:

FactorPotential IssuesSolutions
Viral activityDecreased titer due to improper handlingThaw on ice, avoid freeze-thaw cycles, recheck titer after 6+ months
Target cellsCell type susceptibilityConsider adenovirus for difficult-to-infect cells; use centrifugation for suspension cells
MOIInsufficient viral particlesConduct MOI gradient experiments to find optimal concentration
Infection timePremature or delayed media changeChange media 24h post-infection; observe fluorescence at 48-72h
Transfection reagentPolybrene toxicityTest different polybrene concentrations (1-10 μg/mL)

For persistently low efficiency, consider:

  • For suspension cells: Use spin infection (1200g for 1 hour)
  • For difficult-to-infect cells: Try multiple rounds of infection (add fresh virus 24h after initial infection)
  • For primary cells: Higher MOI may be required
  • If using puromycin-resistant virus: Consider antibiotic selection to enrich for transduced cells

Q4: Despite successful transduction, why is my GFP fluorescence weak after lentiviral infection?

GFP protein expression typically peaks 72–120 hours post-infection in fast-dividing cells, while slower-growing cells may take longer. Fluorescence intensity is also closely linked to promoter strength, and weak promoters result in weaker GFP signals. Lentiviruses carrying gene inserts—especially long or GC-rich sequences—often show reduced downstream fluorescent protein expression compared to control viruses. Technical issues like low-pH media, aging microscope light sources, or ambient lighting can also weaken observed fluorescence.

For optimal visualization, consider increasing exposure time when imaging target gene-containing viruses compared to control viruses. This difference in fluorescence intensity is a normal phenomenon and doesn't necessarily indicate lower transduction efficiency.

Q5: I'm trying to establish stable cell lines with lentivirus. What's the best protocol for puromycin selection?

Step 1: Determine optimal puromycin concentration

Before selection, you need to find the minimum puromycin concentration that kills >90% of your untransduced cells:

1. Seed cells in a 24-well plate at 50-60% confluence

2. After 24h, add different puromycin concentrations (try a range: 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 μg/mL)

3. Observe for 48h and select the lowest concentration that kills >90% of cells

Step 2: Perform puromycin selection

1. Infect cells with your puromycin-resistant lentivirus

2. Allow 48-72h for viral expression (when infection efficiency reaches ~80%)

3. When cells reach 60-70% confluence, add the predetermined puromycin concentration

4. Maintain untransduced control cells with the same puromycin concentration

5. After ~48h (when >90% of control cells are dead), replace with fresh media without puromycin

6. For long-term maintenance, periodically re-select with puromycin

Step 3: Expand selected cells

After selection, expand cells through appropriate passages until you have sufficient numbers for:

  • Mixed clone stable population experiments, or
  • Single clone isolation procedures

Pro tip: For maintenance culture after initial selection, you can reduce puromycin concentration to 1/10 - 1/2 of the selection concentration to prevent transgene loss while minimizing stress on cells.

Q6: Why does my lentivirus show good RNA interference by qPCR but poor knockdown at the protein level?

This discrepancy between qPCR and Western blot results is frustrating but not uncommon. Proteins often have long half-lives and may persist for days after mRNA levels drop, with some requiring over 5–7 days for noticeable reduction. If infection efficiency is low, overall knockdown will be limited, even if the RNA interference tool is highly effective. Technical limitations in Western blotting—such as low antibody sensitivity, signal oversaturation, or high background—can obscure real knockdown results. Additionally, some genes have post-transcriptional regulatory mechanisms that maintain protein levels despite reduced mRNA.

Troubleshooting approaches:

  • Verify infection efficiency using co-expressed fluorescent markers
  • Extend the observation window to allow for protein turnover
  • Use puromycin selection to enrich transduced cells
  • Try different antibodies or detection methods
  • Consider alternative knockdown validation methods like immunofluorescence or functional assays

If qPCR consistently shows good knockdown while protein levels remain unchanged, also consider the biological significance - some systems have built-in redundancy mechanisms that may be biologically relevant.

Quick Inquiry