FAQ

Q: What is the maximum size of an rAAV transgene?

A: The upper limit of rAAV genome packaging is ~ 5Kb including the required 145bp ITR sequences at either end. Thus, rAAV accommodate a ~4.5Kb transgene expression cassette. An rAAV transgene expression cassette usually includes a promoter, a gene of interest, and a terminator signal. PackGene’s K104 vector has been designed to maximize gene of interest capacity by integrating the smallest available mammalian promoter region in miniCMV (180 bp) and terminator region (50 bp). PackGene’s K104 vector can thus accept a gene of interest up to 4.4kb in length.

Q: What are rAAV serotypes, and what steps should be taken to ensure that the best serotype is chosen?

A: Different AAV serotypes are defined by differences in the amino acid sequence and three-dimensional structure of their capsid proteins, and more than 200 AAV serotypes of have been discovered or designed. Serotype specific differences in rAAV capsid proteins correspond with variations in cell surface receptor recognition and binding. This, in turn, results in variations in the infection rate of rAAV serotypes across tissues and cell types.

PackGene’s expert technical team is available to help you determine the optimum serotype for your experiments based on the literature regarding rAAV serotype infection rates and our own internal testing. Nevertheless, for target cell types or tissues without substantial literature available, we may recommended the execution of pilot experiments using reporter transgenes to determine the most ideal serotype.

Q: What information will I need to provide to place a custom rAAV vector construction order?

A: Custom AAV vector construction projects can be generated in several ways, and our expert technical team is available to help in the design process. There are several questions that you may prepare answers for to expedite the design process, they are:

  • Do you have a transgene template?
    If so, please provide your gene template to us for verification.
  • Do you want to overexpress a gene without a template?
    If so, please provide the gene number, sequence map, host species, and gene length.
  • Do you want to generate rAAVs for the manipulation of gene expression using techniques such as RNAi or CRISPR?
    If so please provide us with the target gene number, host species, and gene length.
  • Is there a specific promoter sequence you would like to use?
  • Is there a specific fluorescent tag or reporter that you would like to use?
  • Will several transgenes need to be co-expressed simultaneously?
  • Is your total transgene sequence length <4.4kb?

Q: What information will I need to provide to place a custom rAAV packaging order?

A: Custom AAV packaging projects can be generated in several ways, and our expert technical team is available to help in the design process. There are several questions that you may prepare answers for to expedite the design process, they are:

  • Do you know which serotype you would like you use for your project?
    Our expert team is available to help guide your selection if you would like, but you may alternatively find your ideal serotype by looking toward the literature within your field.
  • Are you confident that your preferred serotype is capable of infecting the cells that you plan to use for your experiments?
    If not, we offer fluorescent control test kits for screening various serotypes. These can be used to define the infectivity of a given serotype in your cells, or to determine the optimal serotype for your experiments.
  • Do you plan to provide your own plasmid for packaging?
    If so, please make sure that you to provide a vector map and full sequence. Additionally, it is best to confirm ITR spacing and to make sure that the plasmid has been fully sequenced to avoid complications associated with common mutations that can be driven by the presence of ITRs.
  • What are your requirements for the amount, titer, and packaging of the final deliverable rAAV?

Q: How much plasmid do I need to provide for AAV packaging?

A: You only need to provide 1-4 µg of plasmid. We will handle the plasmid preparation necessary for AAV packaging. You don’t need to purchase an additional plasmid prep service unless you wish to receive more plasmid from us. Please note, the timeline in our quote already includes the plasmid preparation.

Q: What are the difference between research and NHP grade?

A: Research-grade AAV is actually the most common grade used for research and development, while NHP-grade is where we’ve improved the purification process and more stringent QC test, resulting in higher purity, lower endotoxin, better genome integrity and lower empty capsid rates. Since animal experiments demand higher virus quality and better consistency, we recommend using our NHP-grade AAV for large animal experiments, such as NHP, porcine, canine, etc. Of course, if you’re conducting cell experiments and desire higher purity, that’s also recommended.

Q: How do you choose the fluorescent or luminescent marker for live imaging in mice?

A: For in vivo imaging, it’s generally advised to use vectors with luciferase.

Currently, in vivo imaging primarily utilizes two techniques: bioluminescence and fluorescence. Bioluminescence involves using the luciferase gene to label cells or DNA, while fluorescence employs fluorescent proteins such as GFP, EGFP, RFP, YFP, mCherry, etc., to mark cells or proteins. Bioluminescence offers advantages like straightforward operation, sensitive response, rapid imaging, and clear visualization. However, its drawback lies in its relatively weak signal, necessitating the use of CCD lenses for detection and requiring instruments with high precision. On the contrary, fluorescence allows for the utilization of various proteins for labeling and enables multiplex labeling, making the process relatively straightforward. Nevertheless, nonspecific fluorescence imposes limitations on its sensitivity, necessitating the use of excitation lights of different wavelengths, thereby making precise in vivo quantification challenging. Bioluminescence relies on the interaction with luciferase to emit light, demonstrating high specificity. The red light emitted by luciferase penetrates tissues nearly 100 times more effectively in vivo than the green light emitted by green fluorescent protein, resulting in a higher signal-to-noise ratio. While fluorescent proteins necessitate excitation light to produce reflected light, nonspecific fluorescence from the mouse’s fur reduces the signal-to-noise ratio during the detection process. Fluorescent protein detection is more suited to ex vivo detection, whereas luciferase detection is better suited to in vivo detection. Currently, luciferase labeling is more commonly employed. There are two frequently used luciferases: Firefly Luciferase (Fluc) and Renilla Luciferase (Rluc), each utilizing different substrates—D-Luciferin for the former and Coelenterazine for the latter. They emit light of varying colors, with the former emitting light at approximately 560nm and the latter emitting light at approximately 450-480nm. The light emitted by the former penetrates tissues more effectively, while the latter undergoes faster metabolism in vivo compared to the former. Typically, the former is utilized as a reporter gene, although both can be simultaneously employed for dual labeling.

Q: What quality control tests do you conduct for your AAV?

A: Our AAV products are subjected to standard release testing procedures, including endotoxin assessment using Limulus Amebocyte Lysate (LAL) assay, purity analysis via Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis (SDS-PAGE), and titer determination using quantitative Polymerase Chain Reaction (qPCR) or droplet digital PCR(ddPCR). Moreover, we conduct restriction enzyme digestion for the Gene of Interest (GOI) plasmids utilized in packaging. Different grade AAV may include different QC tests as listed here.

In addition, we offer 40+ analytical tests to measure titer, AAV genome integrity, characterization, purity, aggregation, contamination and safty, including TEM, AUC, TCID50, Nanopore deep sequencing and may others. Please refer to our anlytical tests webpage.

Q: How is the titer of AAV determined?

A: During AAV titer measurement, our instruments are initially calibrated using the AAV standard product ATCC VR-1816™, a globally recognized reference titer verified by 16 laboratories. Subsequently, we employ SYBR Green qPCR methodology to ascertain titers, achieving values of 1E+13GC/ml or higher. This meticulous approach ensures alignment with prevailing academic standards and prevents inaccurate titration results.

Besides qPCR, we have several other methods available for AAV titer determination:

  1. Genome titer detection by ddPCR.
  2. Capsid titer detection by ELISA technology.
  3. Infectious titer detection by TCID50.

Q: What is the maximum exogenous gene capacity in rAAV as a gene delivery vector?

A: The genome capacity of rAAV is approximately 5Kb. Excluding the 145bp ITR sequences at both ends, this can accommodate an exogenous sequence of up to 4.5Kb; that is, the total length of the promoter, exogenous gene, and terminator signal must not exceed 4.5kb. To maximize the capacity of open reading frame, we recommend using these elements in our library from our piVector Desginer viral vector tool The smallest broad-spectrum promoter miniCMV (core region of CMV, 180 bp), Short Poly(A) (49bp), leaving maximum capacity of open reading frame (ORF) of approximately 4.4Kb in ssAAV Gene overexpression vector.

Q: What are the difference between scAAV and ssAAV?

A: Adeno-associated viruses (AAV) are single-stranded DNA viruses (ssAAV) that must first undergo a transition from single-stranded genome to transcriptionally active double-stranded form before expression can begin. This process limits gene transduction mediated by AAV vectors and directly affects gene expression efficiency. Self-complementary double-stranded DNA adeno-associated viruses (scAAV), on the other hand, mutate the 3′ ITR trs site, forming double-stranded DNA packaged into AAV. They do not require the transition from single-stranded to double-stranded form. In other words, after entering cells, scAAV viruses can express directly and more rapidly, with higher expression levels. The drawback of scAAV is its smaller packaging capacity and the potential to enhance immunogenicity. It is suitable for research requiring faster expression of target genes or stronger expression of genes smaller than 2.2kb.

Q: What does GC/ml means in rAAV titer, and how is it quantified?

A: GC represents genomic copies. GC/ml represents the total number of genome copies in each milliliter of virus solution. PackGene uses SYBR Green fluorescence quantitative PCR to detect AAV titer, with calibration using the ATCC standard product. The PCR primers is targeted at the ITR region. The titer provided by PackGene is above 1E+13 GC/ml for most serotypes. It also refers to the total number of gene copies in each milliliter of virus solution. Typically, dot-blotting, QC-PCR, and real-time PCR methods are used for determination. Samples are usually treated with DNaseI before testing, so this titer representation generally refers to the total number of gene copies in each milliliter of virus solution.

Q: What methods are employed for AAV purification, and what QC tests are included?

A: PackGene produces high-purity AAV, purified using iodixanol density gradient centrifugation and multiple steps of filtration and sterilization.

PackGene’s offers QC tests for our AAV product depend on the AAV-grade you choose, the detailed QC test comparison are listed here

Here are some QC tests we offer:

① Purity: Assessed by running SDS-PAGE followed by Coomassie blue stainig (research grade) or silver staining (NHP-grade) to observe purity.
② Titer: For research-grade or HT-grade AAV, PackGene calibrates using ATCC standard, then employs SYBR Green QPCR to detect titer, achieving a titer of 1E+13GC/ml or higher.For NHP-grade, we determing the titer via ddPCR with primers specifically complement to 5′ and 3′ ITR region.
③ Endotoxin detection: Endotoxin content does not exceed 10 EU/ml for research-grade and 1EU/ml for NHP-grade.You may request more stringent endotoxin threshold with additional fees.
④ TEM electron microscopy (optional): Observation of empty/full ratio through electron microscopy, with a rate below 30% for most common serotypes in research-grade AAV, or below 20% for most common serotypes in NHP-grade AAV.
⑤ HPLC based purity assessment (optional): We offer AEC-HPLC (Anion Exchange Chromatography – High Performance Liquid Chromatography) and SEC-HPLC(Size Exclusion Chromatography – High Performance Liquid Chromatography). Both methods are used for measuring the purity of Adeno-Associated Virus (AAV) preparations.
AEC-HPLC is particularly useful for separating and measuring AAV from impurities such as host cell proteins and nucleic acids, which may have a different net charge compared to the AAV particles.
SEC-HPLC separates molecules based on their size. SEC-HPLC is valuable for assessing the size distribution of AAV particles and separating AAV from aggregates, fragmented particles, or other macromolecules present in the sample.
⑥ Mass spectrometry analysis (optional): Determination of AAV serotype. We offer AAV Capsid Peptide Mapping test by HPLC-MS/MS, and Capsid Protein Molecular Weight and Ratio test by RP-HPLC-MS. AAV Capsid Peptide Mapping by HPLC-MS/MS involves analyzing the peptides within the capsid proteins of the Adeno-Associated Virus (AAV) using High-Performance Liquid Chromatography coupled with Tandem Mass Spectrometry (HPLC-MS/MS). Capsid Protein Molecular Weight and Ratio by RP-HPLC-MS” involves determining the molecular weight and ratio of the capsid proteins of the AAV using Reversed-Phase High-Performance Liquid Chromatography coupled with Mass Spectrometry (RP-HPLC-MS).
⑦ ddPCR titer detection (optional): compared to qPCR titer measurement, ddPCR offers high sensitivity, accuracy, and reproducibility.

For more additional QC test we provide, please check our AAV analytical serivce page.

Q: Do you perform any AAV QC tests to assess empty capsid rates? If so, what methodologies are used? What level of empty capsid rate do you typically achieve?

A: Typically, methods such as AUC, TEM, CyroTEM, or VG TITER/CAPSID TITER are employed. However, AUC, TEM, and CyroTEM are not suitable for QC release; they are better suited for quality research and process development. Currently, anion exchange chromatography HPLC is utilized for method development. We at PackGene can provide analysis method development services and validate them against CyroTEM and AUC test results.

Empty capsid rates vary for each serotype obtained through column chromatography.For research-grade and NHP-grade, we guarantee <30% or <20% empaty capsid rate by TEM for most common serotypes. For GMP production, we will test the empty capsid rate during procss development. For example, PackGene’s AAV9 production can be optimized through process improvements to maintain levels below 10%.

Q: Which serotypes does rAAV encompass, and how do you determine the suitable serotype?

A: As of now, nine naturally occuring serotypes of human AAV have been discovered (AAV1/2/3/4/5/6/7/8/9) and widely applied in scientific research. AAV10 and AAV11 were first discovered in non-human primates in 2004, and no cross-reactivity was observed between AAV10, AAV11, and AAV2, making them promising candidate vectors. Subsequently, researchers isolated AAV12 and AAV13 from simian adenovirus, with limited research on these serotypes currently. Based on these wild-type AAVs, researchers have developed many AAV mutants, such as AAV-DJ and the PHP series, through various modification strategies.

Due to differences in the spatial structure of capsid proteins among AAV serotypes, there are significant variations in their recognition and binding to cell surface receptors, leading to tropism of different AAV serotypes for different tissues. When selecting serotypes, experimental purposes can refer to AAV serotypes used in peer-reviewed literature. For example, AAV1 and AAV9 are more commonly used in brain research than other wild-type AAV serotypes, while AAV6 exhibits higher lymphocyte selectivity.

There are also many engineered serotypes that have been modified or engineered to enhance specific properties for gene therapy applications. These modifications can include alterations to the capsid proteins to change tissue tropism, improve transduction efficiency, evade immune responses, or increase payload capacity. Engineered AAV serotypes have been developed through various strategies such as directed evolution, rational design, or hybridization of existing serotypes. These engineered serotypes offer enhanced performance and versatility, making them valuable tools for targeted gene delivery in biomedical research and therapeutic applications.

PackGene offers nearly 100 serotypes for our packaging service to assist your research work.

Additionally, the development of AAV mutant serotypes with more tissue specificity and stronger infectivity is crucial for innovation in AAV-mediated gene delivery. PackGene provides comprehensive AAV serotype engineeringg services to offer you a one-stop solution.

However, despite the tissue tropism of wild-type AAVs to some extent, the infection of non-target tissues cannot be completely avoided. In such cases, combining tissue- or cell-specific promoters with serotypes can greatly enhance AAV specificity. PackGene offers various tissue-specific promoters, such as the muscle-specific promoter MHCK7-2 and the liver-specific promoter TBG669. Our piVector Design embed in our online ordering system offers various promoters including universal and tissue specific promoters. You may easily build your vector into our AAV backbones that have been rigorously verified for effective viral packaging.

Q: What features does AAV have comparing to other viral vectors?

A: AAV vectors stand out for their safety, low immunogenicity, ability to transduce non-dividing cells, and potential for long-term gene expression without integrating into the host genome. These features make them particularly attractive for gene therapy applications targeting diseases where long-term expression and safety are paramount.

However, the limited packaging capacity is a constraint when delivering larger genes. In contrast, vectors like adenovirus and HSV can carry larger genetic payloads but come with higher immunogenicity and safety concerns. Lentiviral and retroviral vectors offer stable, long-term expression through genome integration but carry risks associated with insertional mutagenesis.

By leveraging the unique advantages of AAV, such as tissue-specific targeting through various serotypes and a favorable safety profile, therapies can be designed for a range of genetic disorders with minimized risks. These characteristics contribute to the growing preference for AAV vectors in both research and clinical gene therapy programs.

Q: Is there a universal AAV in vivo delivery protocol?

A: As an effective tool for delivering exogenous genes, rAAV are being used by a rapidly increasing number of in vivo researchers. Nevertheless, the ideal injection site, rAAV quantity, rAAV concentration, and incubation period may vary across experiments and scientific fields. Due to this variability, it is difficult to adopt a universal AAV injection protocol that is applicable for all experiments. Clients must therefore explore each of these variables independently based on their specific goals.

Q: What are the general considerations when designing AAV iexperiment?

A: Serotype selection: If you are unsure which AAV serotype is most suitable for your experiments, we advise that you test the infection rates of 3 or more different serotypes in your experimental system with our rAAV fluorescent reporter constructs.
Gradient dilution infection: The level of transgene expression driven by rAAV may vary substantially across different genes. We therefore recommend that you perform 3-4 AAV gradient dose injections to determine the ideal gene expression level for each rAAV before performing any formal experiments.

Experimental control: We advise the use of a GFP positive control vector of the same serotype and promoter as your experimental vector.

Q: Any recommendations for AAV in vitro infection experiement?

A: Serotype selection: For cells cultured in vitro, AAV-DJ and AAV6 are the most common choices. In conventional cell culture, AAV-DJ can infect more than 80% of cells, while AAV6 has the strongest infective potency against blood-derived cells.

Gradient dilution infection: The level of transgene expression driven by rAAV may vary substantially across different genes. We therefore recommend that you perform 3-4 AAV gradient dose injections to determine the ideal gene expression level for each rAAV before performing any formal experiments and to verify that your experimental dose is not cytotoxic.

Assay time: usually 2-7 days after infection.

Q: How should I approach AAV in vivo delivery and experiment design?

A: Recombinant AAV (rAAV) is a powerful tool for delivering exogenous genes, and its use in in vivo research is growing rapidly. However, the ideal injection site, rAAV quantity, concentration, and incubation period can vary widely based on the specific experiment and scientific field. Due to this variability, a universal injection protocol for AAV does not exist, so researchers need to explore each of these variables independently, guided by their unique research goals.

Q: What are your recommendations for pre-experimental design?

A: Serotype selection: If you’re uncertain about the best AAV serotype for your experiments, we recommend testing at least three different serotypes in your system using our control rAAV with fluorescent reporter constructs.

Gradient dilution infection: Since transgene expression levels can vary by gene, we suggest performing 3-4 AAV gradient dose injections to identify the ideal expression level before starting formal experiments.

Experimental control: Use a GFP-positive control vector that matches the serotype and promoter of your experimental vector.

Q: What should I consider when using AAV for in vitro infection?

A: Serotype selection: For in vitro studies, AAV-DJ and AAV6 are common choices. AAV-DJ generally infects over 80% of cultured cells, while AAV6 is particularly effective with blood-derived cells.

Gradient dilution infection: Similar to in vivo experiments, we advise conducting 3-4 AAV gradient dose infections to find the optimal transgene expression level and to ensure the dose is not cytotoxic.

Assay time: Typically, assays should be performed 2-7 days post-infection to assess the effects accurately.

Q: What is the length range for PackGene’s mRNA production?

A: We can produce mRNA ranging from 400 bp to 10 kb. If you require mRNA beyond this length, please reach out to our technical support team, as special protocols may be needed.

Q: Do you guarantee a specific yield of mRNA?

A: Yes, we guarantee the delivery of specific mRNA quantities as per the agreed specifications.

Q: What is the concentration of your mRNA?

A: Our standard mRNA concentration is 1 mg/mL. However, upon request, we can adjust it up to 4 mg/mL at an additional cost.

Q: What buffer is used for mRNA?

A: We use RNase-free H2O as the default buffer and highly recommend it. We have tested its freeze-thaw stability for up to 30 cycles. Alternatively, you can request PBS or 1 mM Sodium Citrate (pH 6.4). Please contact technical support for more details.

Q: How should I store my mRNA?

A: mRNA can be stable for at least 2 years when stored properly at -80°C and if repeated freeze-thaw cycles are avoided. Stability can vary based on the mRNA length, but we’ve tested mRNA to remain stable through 30 freeze-thaw cycles. For long-term storage, -80°C is ideal, while -20°C works for short-term storage.

Q: How should I store and handle mRNA in LNP?

A: mRNA-LNP can be stored at -80°C for up to 2 years. However, mRNA-LNP is highly sensitive to freeze-thaw cycles, which can compromise its ability to transduce cells or tissues. To avoid this, please inform us of your preferred aliquot sizes. Our pre-aliquoted mRNA-LNP kits come in volumes of 10 µL or 50 µL.

Q: What is the concentration of mRNA in LNP?

A: Our standard mRNA concentration in LNP is 0.2 mg/mL, but we can increase this to 2 mg/mL. If higher concentrations are needed, please contact our technical support team.

Q: How does PackGene purify mRNA?

A: For RUO-grade customized mRNA, we use LiCl precipitation for quantities under 1 mg. For larger amounts (>1 mg) or off-the-shelf mRNA, we use HPLC-based Oligo dT purification to enhance RNA purity.

Q: What method does PackGene use for dsRNA removal?

A: Our standard mRNA purification protocol is optimized to reduce dsRNA to very low levels, with final products containing less than 0.1% dsRNA (verified by ELISA). If a more stringent dsRNA threshold is required, we offer additional methods to further reduce dsRNA during mRNA production. Please consult with our technical support team for more details.

Q: How does PackGene measure residual dsRNA?

A: Residual double-stranded RNA (dsRNA) can form during the in vitro transcription process used to synthesize mRNA, potentially triggering unwanted immune responses in humans. To detect and quantify trace amounts of dsRNA contaminants, PackGene uses a double-antibody sandwich ELISA.

The process starts with coating the wells of an ELISA plate with a monoclonal or polyclonal antibody specific to dsRNA. When the mRNA sample is added, any dsRNA present binds to the capture antibody. After incubation, unbound material is washed away, and a second, labeled antibody—also specific to dsRNA—is introduced. This second antibody attaches to the bound dsRNA, forming a “sandwich” complex.

The detection antibody is typically linked to an enzyme, such as horseradish peroxidase (HRP), which reacts with a substrate to produce a measurable signal (colorimetric, fluorescent, or luminescent). The intensity of the signal is proportional to the dsRNA concentration and is quantified using a microplate reader, with values compared to a standard curve of known dsRNA concentrations. This highly specific and sensitive method ensures the safety and purity of mRNA therapeutics by detecting low levels of dsRNA contamination.

Q: How is mRNA purity defined?

A: To assess size-based purity, we run the mRNA sample through a bioanalyzer. The purity is determined by calculating the ratio of the main peak (target size ±15%) relative to the total signal.

Q: What is the function of N1-methyl-pseudouridine (m1Ψ) in mRNA?

A: N1-methyl-pseudouridine (m1Ψ) is a modified nucleoside that plays several key roles when incorporated into mRNA:

  • Enhanced Stability: mRNA with m1Ψ modifications demonstrates increased stability, prolonging the half-life and allowing for longer-lasting protein expression.
  • Reduced Immunogenicity: m1Ψ reduces the immune response typically triggered by unmodified mRNA, preventing inflammation and degradation, making it more efficient for therapeutic delivery.
  • Improved Translation Efficiency: m1Ψ enhances translation efficiency by promoting ribosome binding and the initiation of protein synthesis, resulting in higher protein output.
  • Resistance to RNA Editing Enzymes: m1Ψ provides protection against RNA editing enzymes like ADAR, which can alter the mRNA sequence by editing adenosine residues. This ensures the integrity and fidelity of the mRNA sequence during protein synthesis.

Q: What modified bases does PackGene offer?

A: We offer substitution of uridine bases with N1-methyl-pseudouridine (N1-me-Ψ). If you require other modified bases, please contact our technical support team to discuss your options.

Q: What’s the function of capping?

A: The 5′ cap of mRNA is essential for several key processes in gene expression and stability:

  • Translation Initiation: The 5′ cap, typically a modified guanosine nucleotide (m7G), is recognized by translation initiation factors, facilitating ribosome binding and the start of translation.
  • Protection from Degradation: The cap shields the mRNA from exonucleases, which degrade RNA from the ends, thereby enhancing the molecule’s stability and lifespan.
  • Nuclear Export: The cap aids in exporting mRNA from the nucleus to the cytoplasm, where translation occurs, by interacting with nuclear export factors.
  • Translation Efficiency: It promotes efficient translation by ensuring correct ribosome positioning at the start codon, boosting the accuracy of protein synthesis.
  • mRNA Processing Regulation: The cap also influences mRNA maturation, affecting processes like splicing and polyadenylation, and can impact alternative splicing patterns.

Q: What 5’ cap does PackGene offer?

A: We provide Cap1 capping through co-transcriptional capping by default. Alternatively, enzymatic capping for Cap1 is available at an additional cost.

Q: How does PackGene measure capping efficiency?

A: We assess capping efficiency using LC-MS. Although not part of our standard release QC test, we typically achieve >97% capping efficiency based on internal testing. Detailed information about the capping efficiency measurement method is available here.

Q: Does PackGene provide Cap Efficiency analysis as stand-alone service?

A: Yes, we provide a capping efficiency assay as part of our mRNA QC services, using enzyme digestion and LC-MS methods. For more information, please contact our technical support team.

Q: Can I use my own UTRs for mRNA production at PackGene?

A: Yes, you can, but we will first evaluate your sequence to ensure compatibility with our IVT system. Please send your sequence to our technical support team for assessment.

Q: What is the sequence requirement for the T7 promoter?

A: For producing Cap1 mRNA using co-transcriptional capping, the sequence must include “AGG” following the T7 promoter.

Q: Does PackGene provide mRNA codon optimization?

A: Yes, we offer codon optimization services. Please contact our technical support team for further details.

Q: Can I use plasmids from previous vector construction orders for mRNA production?

A: Yes, we can subclone your ORF sequences into our proprietary vector, which includes the T7 promoter, UTR, and a 110A tail, for mRNA production.

Q: What method does PackGene use for the poly(A) length assay?

A: To measure poly(A) tail length, we enzymatically digest the mRNA sample with RNases or other hydrolyzing agents, breaking it down into smaller fragments and releasing the adenosine-rich poly(A) tails. These fragments are then quantified using LC-MS.

Q: Can PackGene perform quality testing on customer-supplied mRNA?

A: Yes, we can perform quality testing on mRNA provided by customers. Please refer to the mRNA QC tests we offer here.

Q: How much mRNA is needed to transfect cell cultures?

A: Generally, we recommend using 0.5 µg to 2.5 µg of mRNA per well in 6-well plates (about 2 mL of growth medium containing 1.0–3.5 x 10^6 cells). For transfection in 24-well plates, we typically use 0.5 µg to 1 µg of mRNA per well.

Q: How much mRNA in LNP is needed for mouse injections?

A: The amount of mRNA depends on the desired expression level and target cells or tissues. For mouse injections, we have tested 0.5 mg of FLuc or EGFP mRNA in LNP per kg of mouse body weight, yielding strong expression. For specific genes, it’s best to try a few different doses to optimize the amount.

Q: Does PackGene provide GMP-grade mRNA?

A: Yes, we can supply cGMP mRNA through our partnership with Kudo Biotechnology. For more information, please visit our GMP mRNA service webpage. Please check our GMP mRNA webpage.

Q: What template does PackGene use for mRNA manufacturing, and how is it produced?

A: We use linearized DNA plasmids as templates for mRNA production. The gene can either be designed by our team using PackGene’s proprietary vectors and tools or provided by you. Once the design is finalized, we manage the gene and mRNA manufacturing process and deliver the mRNA as part of the fee-for-service agreement.

Q: Can PackGene co-encapsulate both gene-editing mRNA and sgRNA in LNP? Which LNP formulation should be used?

A: Yes, we can co-encapsulate gene-editing mRNA and sgRNA in LNP. We have successfully co-encapsulated SpCas9 mRNA with sgRNA, achieving robust gene-editing efficiency in Huh7 cells. We recommend using the LP01 LNP formulation, and we will provide updates as we test other formulations.

Q: What are the differences between in vitro grade and in vivo grade lentivirus?

A: The purification processes for in vitro grade and in vivo grade lentivirus are different. Both grades undergo sterile filtration before release, but in vivo grade purification involves additional stringent steps such as ultracentrifugation and ultrafiltration. In vitro grade lentivirus is intended for cell culture experiments, while preclinical-grade lentivirus can be used for both cell culture and in vivo animal studies.

Q: How much plasmid do I need to provide for lentivirus packaging?

A: You only need to provide 1-4 µg of plasmid. We will handle the plasmid preparation necessary for lentivirus packaging. You don’t need to purchase an additional plasmid prep service unless you wish to receive more plasmid from us. Please note, the timeline in our quote already includes the plasmid preparation.

Q: What QC tests do you conduct on your lentivirus?

A: Lentivirus quality control (QC) encompasses various tests to ensure the integrity and safety of lentiviral vectors for research and therapeutic applications. Our release QC testing primarily focuses on post-transduction titer, which provides a true functional titer(infectious titer) to prevent overestimation.

Post-transduction qPCR involves infecting cells with lentivirus followed by quantifying viral titers using quantitative PCR (qPCR), providing essential information about lentiviral transduction efficiency.

Transduction tests are performed by infecting cells with lentivirus and diluting them to count fluorescent cells. Bright field and fluorescent microscopy images are then analyzed to evaluate transduction efficiency.

Additional QC tests include p24 ELISA to measure the core capsid protein of HIV for accurate lentivirus titration, PCR-based mycoplasma testing to ensure absence of contamination, bioburden testing to quantify live microorganisms, and endotoxin testing using the Limulus amebocyte lysate (LAL) assay to confirm absence of endotoxin contamination.

These comprehensive QC tests ensure that lentiviral vectors are of high quality, free from contaminants, and suitable for downstream research and therapeutic applications.

Q: Which packaging system do you utilize for lentivirus production?

A: We use 3rd generation, 4 plasmid system.

Q: What are the benefits of using the 3rd generation lentivirus system?

A: The 3rd generation lentivirus packaging system offers several benefits. It is inherently safer as it does not replicate and self-inactivate, ensuring enhanced biosafety. This system utilizes four plasmids, each carrying different HIV genes, including the removal of tat, which is advantageous for optimizing safety profiles. Additionally, it features a partially removed 5′ LTR followed by a strong promoter such as CMV, RSV, EF1, or U6 for RNA, enhancing transduction efficiency and reducing the risk of insertional mutagenesis. Furthermore, the removal of U3 renders the lentivirus self-inactivating (SIN), minimizing the risk of unintended integration and enhancing the safety profile of lentiviral vectors for gene delivery applications.

Q: How should lentivirus be stored, and what is their shelf life?

A: Lentivirus can be stored for up to 6 months at -80°C, although its titer may decrease after this period. To maintain optimal titer, we recommend avoiding freeze-thaw cycles and aliquoting the lentivirus into smaller portions upon the first thaw. We also offer aliquoting services. The first 10 vials aliquoting is complimentary, with a charge of $5 per additional vial.

Q: What is the genome capacity between the 5′ LTR and 3′ LTR of lentivirus?

A: The lentivirus genome size between LTRs is about 9.3kb. However, to ensure optimal yield during lentivirus production, we recommend keeping the lentivirus genome size between LTRs below 6.5kb. If the genome size exceeds this limit, we cannot guarantee the desired yield. Additionally, if the initial yield does not meet expectations, we will initiate a second batch production to ensure accuracy.

Q: What is the titer of your lentivirus?

A: We offer lentivirus with a functional titer of (infectious titer) ≥1E+8TU/mL for both research-grade and preclinical-grade vectors, with the exact titer provided in the certificate of analysis (COA) report. Please note the functional titer (infectious titer) measured by post-transduction qPCR is usually 100-1000 fold higher than physical titer, measured by p24.

Q: How is the lentivirus titer determined?

A: In contrast to the common practice among most vendors, who typically measure lentivirus titer directly using methods such as qPCR or p24 ELISA. The Traditional p24 ELISA kit is the most commonly published method for measuring lentiviral titer. The method is suitable for tittering native or purified recombinant virus. However, in crude (unpurified) lentiviral supernatant, significant concentrations of overexpressed p24 protein may be present that are not assembled into viral particles. This causes an extreme overestimation of lentiviral titer.

We employ a different approach. Our titer measurement focuses on the post-transduction titer determined by qPCR, which helps to eliminate any concerns regarding overestimation, providing infectious titers that are significantly higher (100-1000 times) than the physical titer measured by p24, ensuring greater accuracy and consistency for experimental applications.

Additionally, if the lentivirus carries a fluorescent protein, we can further validate the post-transduction titer by examining bright-field and fluorescent microscopy images obtained from serial dilutions of the virus.

Q: Is it possible to adjust the lentivirus titer according to specific requirements?

A: Since the transduction unit is determined after storage, we do not adjust the concentration to avoid additional freeze-thaw cycles that may significantly lower the titer. Customers can adjust the concentration as needed upon infection.

Q: What is the difference between physical titer and infectious titer?

A: Viral titers are typically reported in two forms: functional titer(infectious titer), expressed as transduction units (TU/mL), and physical titer, measured in viral particles (VP/mL). The physical titer indicates the total amount of virus present, often quantified by assessing levels of viral proteins like p24 or viral nucleic acids. In contrast, the functional titer(infectious titer) reflects the virus’s ability to infect cells and is usually 100 to 1000 times lower than the physical titer. Although direct measurement of functional titer(infectious titer) is more accurate for calculating the multiplicity of infection (MOI), it is often more labor-intensive. At PackGene, lentivirus scale is based on post-transduction titer, providing infectious titers that are significantly higher than the physical titer measured by p24, ensuring greater accuracy for experimental applications.

Q: What is the buffer composition of the lentivirus?

A: We utilize Opti-Medium as the buffer at a concentration of 50μl/mL for lentivirus storage.

Q: Which cell line is employed for lentivirus production?

A: We use either 293T adherent cells or PCS01 suspension cells (a single-clone suspension cell line originated from HEK293 cells).

Q: What are the disposal procedures for lentivirus and equipment or supplies that have been in contact with lentivirus?

A: All equipment or tips in contact with lentivirus or virus-infected cells should be immersed in a 10% bleach solution for 20 minutes before disposal in biohazardous waste.

Q: How much plasmid material is required for lentivirus packaging?

A: Customers only need to send us 10μg of plasmid for lentivirus production. We will amplify and QC the plasmid before use, with the plasmid prep cost and timeline included in the service fee. Typically, research-grade lentivirus requires 300ug for 5E7, 600ug for 1E8, while preclinical-grade lentivirus requires 1.5mg for 5E7, and 3mg for 1E8.

Q: Do you offer assistance design lentivirus plasmid?

A: Yes. We offer piVector Designer, an online-tool to help you design the lentivirus plasmid. The plamid backbones of various application we provided in piVector Designer is experimentally validated by us, and we provide a vast library of gene elements including promoter, reporter/marker, regulatory elements and polyA. Also, our Ph.D. level techincal support team will evaluate

Q: What is a plasmid?

A: Plasmids are self-replicating, extrachromosomal DNA molecules found in bacteria and some other organisms. These small, circular pieces of DNA exist independently of the bacterial chromosome and carry genes that often provide advantages, such as antibiotic resistance or unique metabolic functions. While they are not part of the main bacterial genome, plasmids can replicate on their own within the host cell, making them valuable tools in genetic research.

Q: What are the components of plasmid vectors?

A: Plasmid vectors are indispensable in genetics and molecular biology, serving as vehicles to insert, manipulate, and transfer genes within organisms. They typically contain key components like the origin of replication (ori), promoter regions, open reading frames (ORFs), regulatory elements, a polyadenylation (poly A) signal, multiple cloning sites (MCS), and antibiotic resistance genes. Choosing the right combination of these elements is crucial for the success of your research, as each one plays a specific role in plasmid functionality.

Q: What is the origin of replication (ori)?

A: An Origin of Replication (Ori) is a key DNA sequence in plasmids that signals the start of DNA replication within a host cell. It enables the plasmid to replicate independently. Selecting the right Ori is critical to ensure optimal plasmid functionality and stability. Here’s how to choose the appropriate Ori:

  • Understand Your Host Organism: Determine the organism (bacteria, yeast, or mammalian cells) where the plasmid will be expressed. Different organisms have distinct replication systems, so your Ori must be compatible with the host’s machinery.
  • Consider Copy Number: Ori affects plasmid copy number, which influences gene expression levels. High-copy plasmids are ideal for high gene expression, while low-copy plasmids provide stability and controlled expression. Other factors like promoter choice and growth conditions also affect protein expression.
  • Evaluate Regulatory Elements: Some Ori regions contain regulatory sequences that control replication efficiency and stability. If you need precise control over replication timing, choose an Ori with the necessary regulatory features.
  • Test and Optimize: It may be necessary to try different Ori sequences in pilot experiments to find the one that works best for your plasmid’s stability, replication, and gene expression.

Q: What is a promoter?

A: A promoter is a DNA sequence that acts as a molecular switch, controlling when and how genes are expressed. Positioned upstream of a gene, the promoter dictates the transcription of DNA into mRNA, which is then translated into proteins. Choosing the right promoter is essential for plasmid design.

Types of Promoters:

  • Constitutive Promoters: These promote continuous gene expression without regulation. Examples include the lac promoter (E. coli) and the CMV promoter (mammalian cells).
  • Inducible Promoters: These allow controlled gene expression by external inducers like chemicals or temperature changes. For instance, the lac promoter can be induced with IPTG.
  • Repressible Promoters: Gene expression can be reduced under specific conditions. The trp promoter (E. coli) is repressible by tryptophan.
  • Ubiquitous Promoters: These promote gene expression across a wide range of cells or tissues, commonly used in gene therapy.
  • Tissue-Specific Promoters: These restrict gene expression to specific cell types, useful in developmental biology and gene therapy.

Promoter Selection Considerations:

  • Expression Strength: Promoters differ in their ability to drive gene expression.
  • Host Organism: The promoter must be compatible with the host organism’s RNA polymerase system.
  • Regulation: Choose between constitutive or inducible promoters based on whether you want continuous or controlled expression.
  • RNA Polymerase Specificity: Different RNA polymerases (I, II, III) in eukaryotic cells recognize specific promoters, influencing gene transcription.
  • RNA polymerase I (RNA Pol I): Responsible for ribosomal RNA (rRNA) transcription.
  • RNA polymerase II (RNA Pol II): Transcribes eukaryotic protein-coding genes.
  • RNA polymerase III (RNA Pol III): Handles the transcription of small RNAs.

Q: What are commonly used eukaryotic promoters?

A: CMV (Cytomegalovirus): Known for strong expression in mammalian cells but may lead to cytotoxicity and gene silencing.
SV40 (Simian Virus 40): Provides moderate to strong expression in mammalian cells, though promoter silencing can occur over time.
EF-1α (Elongation Factor-1 alpha): Ensures strong, stable expression in mammalian cells, often used in stem cell research.
Tet-On/Tet-Off: Allows inducible expression in mammalian cells using tetracycline, offering precise control over gene activity.
U6: Commonly used for expressing small RNAs like shRNA or siRNA for gene knockdown studies.
PGK: Suitable for expressing transgenes in yeast and mammalian systems.
UAS (Yeast GAL1): Controlled by the GAL4 transcription factor, used for regulated gene expression in yeast.

To access and learn more promoters, please go to piVector Designer Gene elements library.

Q: What are reporter genes and tags?

A: Reporter Genes: These encode detectable proteins or enzymes, used to track gene expression or specific cellular conditions. 

Fluorescent Proteins (e.g., GFP): Emit fluorescence under specific light wavelengths, allowing real-time visualization of protein localization.
Luciferase: Produces light during chemical reactions, useful for studying gene expression and signaling pathways.
Beta-Galactosidase: Converts substrates into colored products, used for visualizing gene expression as blue stains in cells.

Reporter Tags: Small protein sequences genetically fused to target proteins, helping in purification or detection.

Myc Tag: An 11-amino acid tag used in various assays for protein detection.
FLAG Tag: An 8-amino acid tag useful for purification and detection. It can be removed by specific enzymes if needed.
HA Tag: A 9-amino acid sequence often used for detecting and purifying proteins.
GST Tag: Enhances solubility and aids in protein purification, widely used in prokaryotic systems.
His Tag: Composed of six histidine residues, it facilitates easy purification of recombinant proteins through metal affinity chromatography.

Q: What is a multiple cloning site (MCS)?

A: The Multiple Cloning Site (MCS), also known as a polylinker, is a short segment of DNA that contains several unique restriction enzyme recognition sites. These sites allow researchers to insert a gene of interest into the plasmid at a specific location. The MCS simplifies the process of cloning because it offers flexibility in choosing which restriction enzymes to use, making it easier to incorporate foreign DNA fragments into the plasmid without disrupting other essential elements. 

Today, seamless cloning methods are often preferred over using restriction enzymes in the MCS for gene insertion. However, the restriction enzyme sites within the MCS still provide a convenient location for plasmid linearization during seamless cloning.

Q: What is an open reading frame (ORF)?

A: An Open Reading Frame (ORF) is a continuous sequence of codons in DNA that starts with a start codon (usually AUG) and ends with a stop codon (such as UAA, UAG, or UGA). The ORF represents the portion of a gene that is translated into a protein. In plasmid design, the ORF usually includes the gene of interest that researchers wish to express in a host organism. Proper expression of the ORF is crucial for the successful production of the encoded protein.

Q: What are regulatory elements?

A: Regulatory elements are sequences within the plasmid that help control the expression of the gene of interest. One common regulatory element is the Woodchuck Hepatitis Virus Posttranscriptional Regulatory Element (WPRE), which enhances the stability and translation efficiency of mRNA. WPRE is often included in viral vectors or plasmids to increase gene expression, leading to higher levels of protein production in the host cells. Other regulatory elements may control transcription rates, splicing, or mRNA transport.

Q: What is a poly A signal?

A: The Polyadenylation Signal (Poly(A)) is a sequence found near the end of a gene that directs the addition of a poly(A) tail to the mRNA during transcription. This poly(A) tail plays a vital role in stabilizing mRNA and enhancing its translation into proteins. It also aids in mRNA export from the nucleus to the cytoplasm. Including a poly(A) signal in a plasmid ensures that the mRNA produced from the gene of interest will be properly processed, resulting in more stable and efficiently translated transcripts.

Q: What is an antibiotic resistance gene?

A: An Antibiotic Resistance Gene in a plasmid provides the host cell with the ability to survive in the presence of a specific antibiotic. This is used as a selection marker to ensure that only cells that have successfully taken up the plasmid (transformed cells) will grow in an antibiotic-containing environment. Common antibiotic resistance genes include AmpR (confers resistance to ampicillin) and KanR (confers resistance to kanamycin). Researchers use antibiotic selection to isolate and maintain populations of cells containing the plasmid.

Q: What bacteria strains do you use for cloning and plasmid preparation?

A: We use the strains below depend on different applications: 

1. DH5α
Genotype: F–, φ80dlacZΔM15, Δ(lacZYA-argF)U169, recA1, endA1, hsdR17(rK–, mK+), phoA, supE44, λ–, thi-1, gyrA96, relA1
Applications: Commonly used for general cloning purposes and blue/white screening. Its mutations in recA and endA enhance plasmid stability and transformation efficiency, making it a go-to strain for many cloning applications.

2. Top10
Genotype: F–, mcrA, Δ(mrr-hsdRMS-mcrBC), φ80lacZΔM15, ΔlacX74, recA1, araD139, Δ(ara-leu)7697, galU, galK, λ–, rpsL(StrR), endA1, nupG
Applications: Suitable for general cloning and blue/white screening with high transformation efficiency. It is often preferred when maximizing the number of transformants is critical.

3. Stbl3
Genotype: F–, mcrB, mrr, hsdS20(rB–, mB–), recA13, supE44, ara-14, galK2, lacY1, proA2, rpsL20(StrR), xyl-5, λ–, leu, mtl-1
Applications: Ideal for cloning vectors with repetitive elements, such as long terminal repeats (LTRs) in lentiviral plasmids. The recA mutation minimizes recombination, enhancing the stability of complex constructs.

4. XL-10
Genotype: TetR Δ(mcrA)183 Δ(mcrCB-hsdSMR-mrr)173 endA1 supE44 thi-1 recA1 gyrA96 relA1 lac Hte [F´ proAB lacIqZΔM15 Tn10 (TetR) Amy CamR]
Applications: Optimized for high-efficiency transformation, particularly for large or methylated DNA constructs. It is also suitable for blue/white screening and for cloning unstable or toxic sequences due to the recA mutation.

5. NEB Stable
Genotype: F´ proA+B+ lacIq Δ(lacZ)M15 Tn10 (TetR) endA1 recA1 hsdR17(rK– mK+) glnV44 λ– thi-1 gyrA96 relA1 spoT1
Applications: Designed to maintain unstable plasmids that might recombine or degrade in other strains. It is ideal for cloning large or recombination-prone plasmids, offering good yield and stability for long-term propagation.

Q: How is a plasmid constructed based on your design?

A: We use seamless cloning to construct most plasmids.

Seamless cloning is a method that allows precise insertion of DNA fragments into plasmid vectors without adding extra nucleotides at the junctions, a common issue with traditional restriction enzyme methods. This is crucial for protein expression, as even small additional amino acids can affect protein function. There are various approaches to seamless cloning, including techniques like overlap extension PCR or commercial kits.

Key Steps in Seamless Cloning:
Creation of Overlapping Ends: The DNA fragments are generated through PCR with ends that overlap with each other or the plasmid. These overlaps are designed into the primers used for amplification.
Annealing of Overlapping Ends: The complementary overlapping regions hybridize when mixed together.

Extension and Ligation:
A polymerase may extend the annealed fragments, filling in gaps.
DNA ligase then seals the nicks, or a commercial system may combine both enzymatic steps in a single process.

Requirements:
DNA Fragments with Overlaps: These fragments, usually created by PCR, must have complementary sequences for hybridization.
Polymerase: A polymerase is needed for filling in gaps, if required.
DNA Ligase: Ligase seals the nicks unless using a system that combines all steps.

Pros and Cons:

Pros:
Enables precise, in-frame gene insertions without unwanted nucleotides.
No need for restriction enzyme sites, giving more design flexibility.
Simplified primer design.

Cons:
Primer design requires precision for correct overlap and orientation.
Efficiency can depend on factors like fragment size and sequence complexity.
Commercial kits can be expensive.

Tips and Tricks:
Optimal Overlap Length: Overlaps of 15–25 nucleotides typically provide efficient annealing, though larger fragments may require longer overlaps.
High Purity DNA: DNA fragments should be pure, often achieved by gel purification post-PCR, for optimal results.
Control Reactions: Always include controls (like no-insert controls) to detect potential background noise or unwanted ligation.

Q: What plasmid quality control (QC) tests are performed?

A: For research-grade plasmids, we perform the QC tests listed below.

  • Appearance: A visual inspection of the plasmid solution to assess its clarity and the absence of particles or discoloration.
  • A260/280: Measures the purity of plasmid DNA by comparing absorbance at 260 nm (nucleic acids) and 280 nm (proteins). A ratio of ~1.8 indicates pure DNA.
  • Homogeneity by Agarose Gel: Assesses the uniformity of plasmid DNA by running it on an agarose gel to ensure consistent molecular weight and the ratio of supercoiled plasmids.
  • Restriction Analysis: Verifies the identity and integrity of plasmid DNA by cutting it with specific restriction enzymes and analyzing the resulting fragments via gel electrophoresis.
  • Endotoxin by LAL: Detects bacterial endotoxins in plasmid preparations using the Limulus Amebocyte Lysate (LAL) assay, ensuring plasmids are safe for sensitive applications.

 

Additionally, upon request, we offer extra QC tests, which are also included in our preclinical plasmid quality control.

  • Homogeneity by HPLC: Uses High-Performance Liquid Chromatography (HPLC) to evaluate the uniformity and purity of plasmid DNA, and measure the ratio of supercoiled plasmid DNA.
  • Residual RNA by SYBRGold: Quantifies any remaining RNA in the plasmid preparation by staining with SYBRGold and analyzing fluorescence intensity.
  • Residual E. coli DNA by qPCR: Detects and quantifies residual E. coli genomic DNA in plasmid preparations using quantitative PCR (qPCR).
  • Bioburden Testing by Direct Inoculation: Assesses the microbial contamination level of the plasmid preparation by inoculating samples in growth media and monitoring for microbial growth.
  • Sequencing by Sanger: Confirms the accuracy of the plasmid sequence by using Sanger sequencing to verify the inserted DNA or the entire plasmid.
  • Residual Host Protein by ELISA: Detects any leftover host proteins in plasmid preparations using an ELISA assay specific to E. coli proteins.
  • Mycoplasma Contamination by qPCR: Screens for mycoplasma contamination in the plasmid sample using sensitive qPCR techniques.
  • pH by Potentiometry: Measures the pH of the plasmid solution to ensure it is within the acceptable range for stability and application.
  • Residual Kanamycin by ELISA: Detects any remaining kanamycin antibiotic from the plasmid selection process using an ELISA assay.
  • Sterility: Confirms the absence of viable microorganisms in the plasmid preparation, ensuring it is sterile and suitable for sensitive applications.
  • Osmolality: Measures the osmolality (concentration of solutes) in the plasmid solution to ensure it is within acceptable limits for biological compatibility.
Q: Are pH measurements required, and is a large amount of sample wasted to carry out pH measurements?

A: Measurement of pH is a mandatory for the release of rAAV Fast Service deliverables. A micro pH electrode may be used to save sample and thus the required sample volume to perform pH measurements is only ~15uL-100uL.

Q: What is loading?

A: In accordance with the Pharmacopoeia General Rules 0942, we use the minimum filling quantity inspection method for detecting sample loading quantity.

Q: How to interpret A260/A280 value?

A: A260/A280 is the ratio of sample absorbance measured at wavelengths of 260nm and 280nm. This measure is commonly thought to represent the ratio of DNA to protein in a sample. For rAAV, A260/A280 can used as a measure of the full to empty shell rate and to identify protein contamination. Low A260/A280 levels may suggest that the empty shell rate is high. Alternatively, high A260/A280 may suggest that the sample has been contaminated with proteins that are not incorporated into the AAV capsid shell. The greatest advantages of this measure are its convenience and speed.

Q: What tests are performed to differentiate rAAV capsid proteins from specific protein impurities?

A: SDS-PAGE is used to identify rAAV capsid proteins. In addition, SDS-PAGE can be used to directly identify specific protein impurities including the presence of host proteins, BSA, or degraded AAV capsid proteins.

Q: How is rAAV infection titer measured, given that rAAV does not integrate the host genome?

A: The current standard for determining infection titer is TCID50 (Median Tissue Culture Infectious Dose). In this assay we test the infection rates of rAAV sample serial dilutions in H5 cells. This initial test is then followed by qPCR detection of the rAAV genome.

Q: If I have determined that a rAAV DNA vector reliably drives transgene expression in host cells prior to rAAV packaging, can I assume that this DNA vector will also drive transgene expression after it is packaged into an rAAV that is then used to infect cells?

A: Successful transgene expression via rAAV infection relies on several factors beyond the functionality of the DNA vector. We therefore recommend that independent validation of transgene expression is performed for all packaged rAAV.

Q: What does rcAAV refer to, and are there regulations regarding rcAAV content in GMP rAAV samples?

A: The term rcAAV stands for replication-competent AAV. In most cases rAAVs are designed to be replication incompetent. Thus, rAAV samples should not contain rcAAV and regulations require negligible replication capability for GMP rAAV samples.

Q: What is the difference between visible foreign matter and insoluble particles?

A: Visible foreign matter can be identified by visual inspection. According to pharmacopoeia general rule 0904, visible foreign matter in injections, ophthalmic liquid preparations, and sterile APIs can be visually observed under specified conditions . Visible foreign matter is usually derived from insoluble particles of a size or length is greater than 50μm.

Insoluble particles between 10μm and 50μm cannot be seen with the unaided eye and must therefore be detected by instrumentation. Insoluble particles identification is commonly carried out with an insoluble particle analyzer. According to pharmacopoeia general rule 0903, evaluation of the size and quantity of insoluble particles within materials to be delivered by intravenous injection (solution injection, sterile powder for injection, concentrated solution for injection) can be made according to the project nature.

Q: If I have successfully carried out several in vivo experiments in mice may I assume that mycoplasma and bacterial endotoxin levels are within acceptable ranges and forgo direct testing for these contaminants?

A: Reagents used for Gene Therapies should abide by all domestic regulations in the “Guiding Principles for Quality Control of Human Recombinant DNA Products” by the EDC. Under these standards Mycoplasma and endotoxin testing is required.

Q: What is the source of PackGene cell bank?

A: PackGene’s h293 cell bank is officially authorized for commercial use.

Q: What is the AAV output for a single batch fermentation?

A: PackGene offers single batch fermentation at several volumes, including: 2L, 7L, 25L, 50L, and 200L. AAV yields for each of these production volumes varies across AAV serotypes. As an example, AAV9 is a medium to high-yielding serotype, and expected yields for AAV9 are as follows:

Expected yield for AAV9
Volume Yield
2L 1E+14GC
50L 1E+16GC
100L 2E+16GC
Q: How are GC/ml and vg/ml related to one another, and how does PackGene determine GC/ml for AAV products?

A: The terms genome copies per ml (GC/ml) and viral genomes per ml (vg/ml) are interchangeable and equal in most cases. At PackGene we may test GC by both qPCR and ddPCR. Testing by qPCR involves the use of a calibration standard while ddPCR may use optional reference products. Measurements by qPCR are more likely to be influenced by inter-lab and inter-operator variables, and ddPCR generally shows lower %RSD precision. Typically, GC is determined by qPCR during process exploration phase and for intermediate products while the GC of final products is more often determined by ddPCR.

Q: Do specific primers need to be designed to determine GC/ml for a custom AAVs?

A: Genome copies per ml (GC/ml) can be determined using primers directed at common vector elements during the early development stage, and thus we use common features such polyA segments or ITRs to determine GC/ml during this stage. However, it is recommended that specific primers targeted at AAV vector transgenes are used for testing the GC of GMP products. Our final fast service titer test is designed to use transgenes specific primers.

Q: What method is used to determine the empty shell rate for AAV samples?

A: Viral empty shell rate can be determined using several techniques including anion chromatography HPLC, Analytical Ultracentrifugation (AUC), Transmission Electron Microscopy (TEM), CyroTEM, or VG Titer/Capsid titer. AUC, TEM and CyroTEM are typically not suitable for quantitative quality control determinations and thus PackGene’s standard method for empty shell rate determination is anion chromatography HPLC. PackGene can provide additional CyroTEM and AUC analytical services to serve as a secondary verification of the results derived from anion chromatography HPLC.

Q: What is the expected empty shell rate PackGene AAV Fast Services?

A: The expected empty shell rate for our AAV Fast Services varies across AAV serotypes. As one example, the expected empty shell rate for AAV9 generated through our AAV Fast Service is lower than 10%.

Q: Is verification of cell banks in accordance with the Chinese Pharmacopoeia 2015 Three General Principles “The Preparation and Verification Regulations of Animal Cell Matrix for the Verification of Biological Products”?

A: Yes.

Q: HIV, HBV, HCV testing, pyrogen-free tests, and wild virus tests are not in the standard quality control services list. Do these tests need to be applied?

A: The harvest liquid generated through PackGene’s AAV fast service will be broadly tested for exogenous viral elements as necessary following an evaluation of project characteristics. Both the original harvest liquid and final deliverables for the AAV Fast service will be devoid of such exogenous viral elements.

Q: What does PackGene consider acceptable error or deviation ranges for standard QC analysis methods?

A: The %RSD of general biochemical methods is usually 15%-25%, and the accepted standard of ddPCR is 10%. In most cases the %RSD for PackGene’s QC ddPCR verification data is lower than 5%, and the %RSD of TCID50 is lower than 10%.

Q: Are there any requirements for the number of samples sent in batches?

A: It is recommended that the sample volume of a single commissioned express delivery is not less than 50uL to avoid the effects of freezing and thawing, evaporation, and tube wall adhesion. The recommended sample delivery volume for genome titer, plasmid DNA residue, rcAAV, etc. is more than 10ul. The recommended sample delivery volume for infection titer is more than 20ul, and the sample delivery amount for empty shell rate testing is 5E+13vg.