Achieving Cell and Gene Therapy Excellence
Compendium
Published: November 6, 2024
Credit: iStock
Cell and gene therapies are transforming medicine, promising cures rather than symptom management. However, developing safe and effective therapies requires overcoming complex analytical challenges.
This compendium explores analytical advancements in cell and gene therapy, highlighting the latest capillary electrophoresis and mass spectrometry tools and strategies for ensuring therapeutic quality.
Download this compendium to explore:
- Cutting-edge analytical methods for characterizing lipids, mRNA, viral vectors and plasmid DNA
- Expert interviews providing insights into lessons learned and best practices for gene therapy
- Practical tips for implementing high-resolution solutions in analytical workflows
Breaking through
analytical boundaries
Development of next-generation
gene therapies and vaccines
Cell and Gene Therapy Compendium
Contents
03 Introduction
04 Non-viral delivery
Overview
05 Expert Q&A with Dr. Adam Crowe (Cytiva)
Workflows:
07 Characterization of lipids and related impurities
08 Streamlined lipid quality control
09 Lipid MetID and bioanalysis
10 Analytical solutions for lipids and LNPs
11 Tips and tricks with Dr. Paul Norris
using LC-MS/MS with EAD
12 Viral vectors
Overview
13 Expert Q&A with Dr. Jane Luo (SCIEX)
Workflows:
15 Intact viral protein characterization
16 Viral protein peptide mapping
17 Protein purity
18 Genome integrity and purity
19 Full-and-empty capsid ratios
20 Residual host cell DNA
21 Host cell protein ID
22 Monitoring of host cell proteins
23 Proteome profiling
24 Analytical solutions for viral vectors
26 Tips and tricks with Dr. Zhengwei Chen using LC-MS
27 Tips and tricks with Peter Holper using CE
28 Plasmid DNA
Overview
29 Expert Q&A with Dr. Emma Bjorgum (Aldevron)
Workflows:
31 pDNA topology and purity
32 pDNA linearization efficiency and sizing
33 pDNA restriction map
34 Residual host cell nucleic acids
35 Analytical solutions for plasmid DNA
36 IVT RNA
Overview
37 Expert Q&A with Dr. Jingtao Zhang and Daniel Turner
(Catalent® Pharma Solutions)
Workflows:
39 mRNA integrity and purity
40 srRNA integrity and purity
41 Circular RNA assessment
42 Encapsulation efficiency of mRNA
43 5’ capping of mRNA
44 3’ end poly(A) tail of mRNA with LC-MS
45 3’ end poly(A) tail of mRNA with CGE-UV
46 Protein expression analysis
47 Analytical solutions for IVT RNA
49 Oligonucleotide therapeutics
Overview
50 Expert Q&A with Dr. Troy Voelker
and Shane Karnik (Aliri Bioanalysis)
Workflows:
52 Characterization and impurity ID
53 Oligonucleotide MetID
54 Bioanalysis and DMPK
55 Analytical solutions for oligonucleotide
therapeutics
57 Tips and tricks with Dr. Dilip Reddy using LC-MS
58 Tips and tricks with Kaoru Karasawa using LC-MS
59 Gene editing
Overview
60 Expert Q&A with Ashley Jacobi (IDT)
and Tingting Li (SCIEX)
Workflows:
62 sgRNA and Cas9 mRNA integrity and purity
63 pegRNA purity for prime editing
64 Intact mass characterization of guide RNAs
65 Cas9 protein characterization
66 Proteome profiling for gene editing
67 Analytical solutions for CRISPR/Cas9
69 Concluding remarks
2 | SCIEX | CGT compendium SCIEX.com/biopharma-and-pharma-research
Introduction
Advancing human health has never been charged with so many possibilities before. Cell and gene
therapy (CGT) products transform lives by going beyond treating symptoms, often with the goal of
curing rare and lethal diseases.
Apart from gene therapy, oligonucleotide-based products are also used to treat diseases by
modulating endogenous messenger RNA (mRNA) expression or by activating the immune response
to fight malignant tumor cells. Furthermore, the platform capabilities of mRNA packaged in lipid
nanoparticles (LNPs) create opportunities in preventative care. This has the potential to enable
more rapid adaptation of vaccines against infectious diseases and germ mutations and allows
for more cost-effective manufacturing. These factors are key to enabling wider global access of
medicine and better preparation for future health crises.
Despite all the excitement new drug candidates can bring, no short cuts can be taken when
ensuring their quality and proving their safety and efficacy. The complexity and novelty of these
modalities require continuous learning and adaptation, out-of-the-box thinking and evolving
analytical technologies to get answers to pressing questions.
Here at SCIEX, we are a team dedicated to analytical science. We can help you break through
analytical boundaries and revolutionize medicine by providing capillary electrophoresis (CE) and
mass spectrometry (MS) solutions. Together, we can reach new heights as we move towards the
future of CGT.
3 | SCIEX | CGT compendium SCIEX.com/biopharma-and-pharma-research
Non-viral delivery
Adam Crowe (PhD)
Sr. Manager Analytical Development,
Cytiva
“LNPs pose unique analytical
challenges, in part due to the
complexity of their lipid excipients.
Consequently, the detailed structure elucidation capabilities
afforded by electron activated dissociation (EAD) in the
ZenoTOF 7600 system from SCIEX provide the analytical
chemist unparalleled capacity to identify problematic oxidative
impurities inside ionizable lipids, and thereby de-risk the
LNP therapeutic development process and expedite paths
to the clinic.”
Lipid nanoparticles (LNPs), are widely used for the delivery of
vaccines and therapeutics such as in vitro transcribed (IVT)
RNA, small interfering RNA (siRNA), antisense oligonucleotides
(ASOs) and more. 4 different classes of lipids are used within
LNPs: ionizable cationic lipids or cationic lipids, sterol lipids,
helper lipids and PEG-lipids. The flexibility of LNPs regarding the
type and size of the cargo, limited adverse effects and easier
scale-up compared to viral vectors, are factors that contribute
to the increased use of LNPs as delivery vehicles. As a result, it
is important to analyze lipid raw material quality, characterize
the lipids in the drug products and perform bioanalysis studies
to help mitigate risks to patient safety and drug efficacy.
Raw material and LNP characterization
Explore how to perform comprehensive structural
elucidation of lipid raw materials and lipids from LNPs.
MetID and bioanalysis
Overcome matrix complexity to perform identification of
metabolites (metID) from lipids and bioanalysis studies.
Lipid quantitation
Streamline the quality control of raw material and
the bioanalysis of LNPs with efficient and sensitive
analytical technology.
Non-viral delivery Overview
4 | SCIEX | CGT compendium SCIEX.com/lipid-nanoparticles
Expert Q&A: Lipid impurity analysis
with LC-MS/MS and EAD
Characterizing the lipid raw material is a crucial step towards the successful development of LNP-based
drugs. Dr. Adam Crowe explains why and provides insights into the learnings from his team based on
many years of research on a variety of ionizable lipids.
Dr. Adam Crowe manages a multi-discipline team at Cytiva, tasked with developing novel analytical assays related
to LNPs and nanomaterials for drug delivery. During his tenure, Adam has garnered broad expertise in analyzing
particle payloads, physiochemical characteristics, excipients, and in vitro potency using a variety of analytical
methodologies. Notably, he leveraged cutting-edge LC-MS technology for LNP characterization. Additionally, Adam
is the technical lead for the American Society for Testing and Materials (ASTM) guide document, outlining best
analytical practices for the LNP field.
In your opinion, what are the best analytical techniques for assessing the purity of ionizable lipids?
At Cytiva, my team and I use at least three different methodologies to look at quality. We use charged
aerosol detection (CAD) for the overall profile, the liquid chromatography coupled to tandem mass
spectrometry (LC-MS/MS) with EAD method described in my webinar, and a fluorescence-based assay.
The assessment should not be taken lightly, in my opinion. The detailed analysis of the ionizable lipid is
paramount for the success of a project. In my experience, one of the most common ways that clinical
programs based on LNPs fail is a lack of careful assessment of the raw material.
Can you detect N-oxides with CAD?
Yes and no. You will run into two problems. N-oxides tend to elute very close to the main peak of the
ionizable lipid.
While you can chromatographically separate them, the gradients required are quite long and you will need
prior expertise in what you are trying to separate. The other issue is the relative abundance. Because the
N-oxides are an intermediate product that degrades further, you never form huge amounts of it. At ~0.1%
relative abundance is when I start to get concerned about N-oxide formation. This makes it difficult for
CAD to detect N-oxides because of the method’s limited dynamic range, adding to the challenge of having
to know what to look for.
At which levels do N-oxides impact mRNA efficacy?
This is an interesting question. It seems in very, very low abundance. We’ve had the luxury of looking at
the adduct formation of 20 to 30 different ionizable lipids. Since the N-oxide itself is not reacting with
the mRNA, but presumably an aldehyde—a degradation product of the N-oxide as described by Packer
et al. in 2021—predictions are challenging. As a summary, I can say that when N-oxides are present in a
significant quantity, meaning ≥1% abundance, we see very significant adduct formation.
Do you have any thoughts on acceptable levels of N-oxides or adducts?
As I mentioned before, N-oxide levels above a 0.1% threshold is where we start to consider adduct
formation a problem. However, it is the lipidation event itself that you will need to monitor and do rate
calculations on to assess the severity. This is because the N-oxides are diagnostic, but not necessarily
predictive of the rate of adduct formation. There are cases where you can see relatively low N-oxide
amounts, but the rate of adduct formation on the RNA is quite fast.
In such cases, it is likely that the N-oxides have already degraded to another reactive species.
Non-viral delivery Expert Q&A
5 | SCIEX | CGT compendium SCIEX.com/lipid-nanoparticles
Do you use MS/MS with EAD only for raw materials, or do you also monitor N-oxides in formulated
LNPs?
You can absolutely use the LC-MS/MS with EAD method I presented in my webinar for formulated
LNPs. It’s obviously less complex to investigate a particular raw material compared to a
formulated LNP because you have less species in a sample. However, it can absolutely be done. I
recommend reaching out to your SCIEX representative as they might have further information on
that topic.
Can you elaborate on how much MS method optimization is typically required and how much time
you need to process the data? Can you efficiently transfer methods to new lipids?
It’s not a whole lot. Although EAD is very tunable, there are very discrete ranges for the type of
fragmentation we are seeking for ionizable lipids. Generally, lipids require high-energy fragmentation
for achieving relevant bond breakage—we used around 15 electron volt (eV). If you want to
determine the behavior of your specific lipids, you can set up a method with different energies
within one injection. The data obtained by EAD are fragment-rich and manual analysis can take
some time. However, SCIEX provides Molecule Profiler software as a solution, which can process
lipid EAD data and does a lot of the interpretation for you. Historically, we would spend almost a
week peering through the data and manually assigning the species that are there. Now, this is
done in a ~10-minute computational run through the software followed by a manual check, so it’s
quite convenient.
Could you give some more detailed information about the MS method setup of the ZenoTOF 7600
system? Did you use targeted, data-dependent or data-independent analysis?
The method used was data-dependent acquisition (DDA)—or information-dependent acquisition
(IDA), as some people call it in the industry—for fragmenting the top five candidates, combined
with an inclusion list. The inclusion list contained the m/z of expected impurities of the ionizable
lipid MC3, such as the addition of oxygen, demethylation, water loss, etc. More information on the
method settings can be found in this technical note. Depending on your needs, you can increase the
candidate ions and adjust the inclusion list.
Do you have recommendations for how to mitigate adduct formation between the ionizable lipid
N-oxide and the RNA?
It really comes down to the quality of your ionizable lipid. Ensuring that the amount of oxygen is
minimized and that the ionizable lipid is not heated or exposed to oxidizing agents will help reduce the
amount of N-oxides. Ensuring that the purification after synthesis is robust will help as well. Generally
speaking, you want to carefully consider your manufacturing synthesis, mechanism, route and
purification of your ionizable lipid to mitigate lipidation.
Non-viral delivery Expert Q&A
Read the entire Q&A blog >
6 | SCIEX | CGT compendium SCIEX.com/lipid-nanoparticles
Ionizable lipids are key components of LNPs, complexing the negatively charged cargo and
facilitating the cellular uptake. Their quality is critical for a stable and efficient product.
Even very low abundance N-oxide impurities can lead to a loss of function. Their structural
identification and differentiation from other impurities is an analytical challenge. Furthermore,
saturation of double bonds of the lipids could impact the structure of LNPs and affect the final
product.
Fully understand the structures of your cationic or ionizable lipid components using EAD
Differentiate between oxidated species and accurately localize double bonds or saturations
with EAD
Avoid missing relevant product excipients by leveraging a linear dynamic range >5 orders of
magnitude and signal-to-noise enhancement with the Zeno trap
Characterization of lipids
and related impurities
Stop the guess work – Determine exact locations of oxygen double bounds,
saturations and more
Figure 1: MS/MS EAD data of D-LinMC3-DMA (MC3) impurity. EAD-derived
fragments can be used to pinpoint an
oxygen incorporation to the tertiary
amine headgroup of MC3. Encircled
m/z show diagnostic fragment ions
for the identification of an N-oxide
impurity derived from MC3.
Non-viral delivery Characterization of lipids and related impurities
Figure 2: MS/MS EAD data of MC3. Figure shows a zoom in to the fragmentation data of C-C bonds for structural elucidation and specific
localization of double bonds, and saturations with EAD.
Discover more details in the technical notes for MC3 > and for ALC-0315 >
MC3 ALC-0315
SCIEX.com/lipid-nanoparticles
O
O N
505.4830 511.5212
518.4910
531.4994
530.4918
532.5072
544.5068
545.5150
546.5222 558.5222
571.5307 584.5382
585.5464
599.5617
627.6
613.6
599.6
585.5
571.5
558.5
545.5
531.5
518.5
505.5
511.5
Intensity
Mass/charge
CH
CH
CH2
CH CH
CH2
CH2
42.0083
91.0537
148.0959
50 100 150 200 250
Mass/charge
Intensity
61.0511
61 62 63
511.5234
512 514
O
N+
O
O
N-oxide
61.05 148.1
511.5
7 | SCIEX | CGT compendium
Commonly, 4 lipid classes (ionizable cationic lipids or cationic lipids, sterol lipids, helper lipids
and polyethylene glycol (PEG) lipids are mixed in defined ratios to form LNPs with desired
physical-chemical properties. To ensure quality criteria are met, lipid raw materials and LNP
batches need to be monitored. Following lipid characterization and impurity identification,
quantitative monitoring can be streamlined.
Leverage excellent sensitivity for quantitation of lipids and breakdown products in raw
materials and formulated LNPs
Rely on robustness and low %CVs with best-in-class triple quadrupole technology
Streamline data acquisition and data processing with intuitive software
Streamlined lipid quality control
Non-viral delivery Streamlined lipid quality control
Detect and quantify different lipid species with high sensitivity and precision
Figure 3: Extracted ion chromatograms for different lipid classes of a liposome with chromatographic separation. Using a normal phase
scheduled multiple reaction monitoring (MRM) approach, very good class separation was achieved to avoid isobaric interferences and
improve confidence in lipid species identification. Diacylglycerol (DAG), cholesteryl ester (CE), posphatidylcholine (PC), sphingomyelin
(SM), lysoposphatidylcholine (LPC), phosphatidylethanolamine (PE), lysophosphatidylethanolamine (LPE), phosphatidylglycerol (PG),
phosphatidylinositol (PI), phosphatidylserine (PS). Intensity (%)
Time (min)
100
0
6 12
DAG
CE
PC SM
LPC
PE LPE PG P1 PS
Discover more details in the technical note on lipid quantitation from liposomes >
8 | SCIEX | CGT compendium SCIEX.com/lipid-nanoparticles
2
3
4
5
6
7
-0.5 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5
Log (area)
Log (concentration)
1
2
3
4
5
6
7
8
Following administration, LNPs can travel to different parts of the body and undergo metabolic
changes. Frequently, the non-endogenous cationic or ionizable cationic lipids are used as surrogates
for quantitative analyses of LNPs in in vivo samples. Multiple bioanalytical end points from a single
administration with small sample volumes require analytical assays with high sensitivity. In addition,
matrix interferences and structural elucidation of metabolites need to be overcome.
Elucidate the structures of your cationic or ionizable lipid components and related metabolites
using EAD
Overcome matrix interferences and achieve outstanding quantitative results for difficult-tofragment lipids with accurate mass spectrometry and the Zeno trap
Achieve identification and quantitation of different lipid species and metabolites in parallel
Lipid MetID and bioanalysis
Non-viral delivery Lipid MetID and bioanalysis
Leverage powerful structural elucidation and quantitation
capabilities in one experiment
Figure 4: Identification of all four lipid species from an LNP. Total ion chromatogram showing the pegylated lipid
2-[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide, PEG2000, the sterol lipid cholesterol, the ionizable lipid
6-((2-hexyldecanoyl)oxy)-N-(6-((2-hexyldecanoyl)oxy)hexyl)-N-(4-hydroxybutyl)hexan-1-aminium (ALC-0315)
and PC without unsaturated carbon bonds as helper lipid with related time-of-flight (TOF) MS data.
Figure 5: Calibration curve for ALC-0315. LNPs were spiked into plasma and extracted using solid phase extraction.
Calibration curve is based on extracted ion chromatograms run in triplicates.
More questions? Contact us >
PEG2000
PC, 36:0
ALC-0315
Intensity 2.0e7
6.0e7
1.0e8
Intensity
Mass/charge
Mass/charge
Intensity
Intensity
Mass/charge
4.5 5.0 5.5 6.0 6.5 7.0 7.5 8.0 8.5 9.0 9.5 10.0 10.5
Time (min)
Intensity
Mass/Charge
789 790 791 792 793 794
Cholesterol - H20
9 | SCIEX | CGT compendium SCIEX.com/lipid-nanoparticles
Analytical solutions for lipids and LNPs
Suitable for:
In-depth structural elucidation
Simultaneous relative quantitation
High flexibility to perform a range of additional workflows
+ + +
Suitable for:
Targeted analysis and monitoring
Excellent quantitative performance
+ +
Non-viral delivery Analytical solutions for lipids and LNPs
SCIEX.com/lipid-nanoparticles
Molecule Profiler
software
ExionLC AE system ZenoTOF 7600 system SCIEX OS software
ExionLC AE system SCIEX 7500+ system SCIEX OS software
10 | SCIEX | CGT compendium
Tips and tricks from our application experts:
Lipid impurity analysis with EAD
Paul Norris (PhD), Sr. Application Support, US at SCIEX, shares his tips and tricks on lipid analysis
using LC-MS/MS with EAD.
Paul Norris specializes in the profiling and characterization of bioactive lipid mediators in the context of physiological
and pathophysiological processes. He has a wealth of experience maximizing the capabilities of triple quadrupole
and Q-TOF solutions for omics discovery and life sciences. Paul’s extensive lipidomics experience started in the lab
of Edward Dennis at UCSD where he contributed to studies as part of the LIPID MAPS consortium before joining
Brigham and Women’s Hospital to lead a lipidomics core facility, supporting numerous resolution
pharmacology projects.
Tip 1: Establish ideal sample concentrations
Start with a concentration of 20-200 ng/mL in 15:85 (v:v) water:acetonitrile for the ionizable lipid as raw
material or as part of a nanoparticle formulation. I recommend targeting a TOF MS signal of >1E5 counts
per second (cps) for the extracted ion chromatogram of the ionizable lipid on a ZenoTOF 7600 system and
adjusting the concentration accordingly. This will result in high quality EAD spectra. For identification of
impurities, I suggest preparing a 100x higher concentration. Since impurities are typically present in much
lower abundance, a higher concentration is required to produce ideal EAD spectra. Typically, 2 µg/mL works
well, however, concentration may need to be adjusted depending on the signal observed.
Tip 2: Resolve ionizable lipids and impurities
Chromatographic separation of ionizable lipids, their impurities and degradation products works
particularly well with reversed phase C18 columns with a larger pore size (e.g., 300 Å). Start with a mobile
phase consisting of 15% water and 85% (v:v) organic solvents like acetonitrile or methanol (or a mixture of
both) with 10 mM ammonium acetate and a 10 min gradient ending in 100% organic solvent. This should
resolve most ionizable lipid impurities and enable clean EAD spectra for characterization. Make sure to
include a 5–10-minute wash at 100% organic before re-equilibrating the column.
Tip 3: Attain comprehensive ionizable lipid fragmentation
Comprehensive EAD spectra can be obtained by using optimized parameters based on the lipid structure.
From my experience, an electron beam current at 5000 nA and an electron kinetic energy from 12—16 eV
work best for most ionizable lipids. These settings also work well for most natural structural lipids with a
similar size and structural composition (e.g., phosphatidylcholines). Great spectral quality with high signalto-noise can be achieved with a reaction time of 30—35 ms and an accumulation time of approximately
100 ms.
Tip 4: Set up method for impurities
Typical impurities observed for a range of ionizable lipids include N-oxides, epoxides and hydroxyl
functional groups at desaturated carbons relative to the parent structure. Additionally, saturation,
desaturation, methylation and demethylation of the parent structure can be observed. I recommend
designing MS/MS methods that incorporate these putative impurities in an inclusion list within a datadependent experiment. With that setup, you can achieve detailed MS/MS information of expected
impurities and of unknown impurities in your sample.
Non-viral delivery Tips and tricks
More questions? Contact us >
11 | SCIEX | CGT compendium SCIEX.com/lipid-nanoparticles
Viral vectors
Viruses are believed to have been around for several billion years
already. It is nature’s optimized way to deliver genetic material
into cells. The outlook of curing genetic diseases through virusenabled gene therapy, rather than treating symptoms, sparked
a mind shift in (bio)pharmaceutical research. In addition, the
ability for transient transgene expression has been investigated
for vaccine usage, with the first viral vector vaccine for human
use being approved in 2010. Apart from adenoviruses (AVs),
adeno-associated viruses (AAVs), lentiviruses (LVs) and more,
engineered viruses and synthetic virus-like particles are studied
for their potential. Fully understanding viral vector drugs is of
utmost importance to ensuring the quality and safety of future
medicines.
Viral vectors Overview
SCIEX.com/viral-vectors
Intact capsid protein characterization
Determine protein integrity and purity
of capsid proteins and achieve highlevel information on post-translational
modifications (PTMs).
Capsid protein peptide mapping
Assess protein sequences in depth
and fully understand PTMs of viral
vector proteins.
Viral protein purity
Assess protein integrity and
purity of your viral vectors to
help ensure vector potency.
Viral genome
Achieve definite answers on the integrity
and purity of your viral genomes to enable
desired vector potency, immunogenicity
and transduction efficiency.
Residual host cell nucleic acids
Determine quantities and lengths of
process-related impurities to assess the
impact on vector potency and the risk of
undesired immunogenicity.
Host cell protein (HCP) ID
Perform identification (ID) and quantitation
of proteins derived from packaging and
producer cell lines regardless of the cell
line and availability of antibodies against
host cell protein targets.
HCP monitoring
Robustly monitor hundreds of
impurities from various packaging
and producer cell lines.
Proteome profiling
See beyond the expected and determine
effects on the entire proteome in an
unbiased manner when modifying gene
expression.
Full-and-empty capsid ratios
Understand the quantity of viral vectors with
intact genomes to assess product quality.
Andrea Martorana (PhD)
Lead Scientist Analytical Development,
AviadoBio Ltd
“Understanding AAV critical quality
attributes is imperative for the
development of AAV particles usable in
gene therapy. CE plays a crucial role in
monitoring thesafety and efficacy of AAV particles and can be
adopted from early-stage discovery to manufacturing. The PA
800 Plus system from SCIEX equipped with the laser-induced
fluorescence (LIF) detector provides reproducibility while
offering the sensitivity and resolution required for analyzing AAV
assembly stoichiometry and integrity of the genome cargo.”
12 | SCIEX | CGT compendium
Expert Q&A: Comprehensive AAV
analysis with CE
AAVs are complex drugs, consisting of a protein shell—the capsid—and a single-stranded
desoxyribonucleic acid (DNA) genome, including the desired transgene. Here, Dr. Jane Luo, an expert in
molecular biology answers pressing questions on the usage of capillary gel electrophoresis (CGE) for the
characterization of critical quality attributes (CQAs) of AAVs.
Dr. Jane Luo is a senior scientist for Cell and Gene Therapy Applications in the Strategic Technical Marketing team
at SCIEX. She earned her PhD in Biochemistry from the City University of New York, received postdoctoral training
on Molecular Biology and Cell Biology at Weill Cornell Medical College and Harvard Medical School and conducted
cancer research as an assistant adjunct professor at UC Irvine. In 2002, she moved to industry to develop capillary
electrophoresis-based products and applications.
How much sample do you need to perform the comprehensive AAV analysis with CE?
For the comprehensive AAV analysis, we performed a capsid protein analysis with CE sodium dodecyl sulfate
(CE-SDS) and the genome analysis with CGE using the BioPhase 8800 system. We then determined the fulland-empty ratio based on genome and protein titers. For both assays we used 30 µL to 40 µL of the sample
with a titer in the range of ~1x1011 to ~1x1013 gene copies (GC)/mL.
What is the linear dynamic range for the genomic titer determination by CE?
The linear range for the method I worked on range from 1×1010 GC/mL to 2×1013 GC/mL. This is a linear
dynamic range of 3.3 orders of magnitude and was suitable for the samples we analyzed.
Do you need eight points for the calibration curve to determine sample titers?
The short answer is no; you do not need to have eight points. From my perspective, there are two aspects
associated with this question. One is linearity and the other one is detection range. For establishing
linearity, the ICH guidelines from the ‘International Council for Harmonisation of Technical Requirements for
Registration of Pharmaceuticals for Human Use’ (ICH) recommend using a minimum of five concentration
endpoints. For the dynamic range, the ICH guidelines recommend the range to cover the test concentration
and up to 30% above and below the test concentration. The method I presented in my webinar covers about
three orders of magnitude, which is suitable for most AAV samples.
You mentioned a viral protein (VP) variant peak. Can you explain what this peak is?
Absolutely. The VP3 variant peak, sometimes also called VP3 prime (VP3’), is a shorter version of the
VP3 protein that is derived from an alternative translation initiation site. For further information on the
identification of the VP3 prime variant, I suggest the following publication from a research group in Japan
(Human Gene Therapy. 32(21–22):1403-1416). This paper explains that the translation initiation site at
methionine (M) 203 can be skipped and an initiation at M211 led to an eight amino acid shorter protein. As
you can see from my results, the VP3 variant is very well separated from VP3, demonstrating the great
resolution of the CE method. In addition, the detection of a VP3 fragment with liquid chromatography
coupled to ultraviolet detection and MS (LC-UV-MS) was described. This fragment was linked to the
hydrolysis of the VP3 at the C-terminus, caused by the low pH of the mobile phase and the high temperature
used for the column oven during the liquid chromatography analysis, which are standard settings needed
for LC-UV-MS analysis. Scientists using liquid chromatography (LC) for analyzing capsid proteins should be
aware of this and might want to consider complementary evaluation with CE.
Viral vectors Expert Q&A
13 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Which signal-to-noise ratio did you use for determining the lower limit of quantitation (LLOQ)?
We follow the ICH guidelines. For determination of the LLOQ, the signal-to-noise ratio was ten or
slightly above ten.
Do you need specific software for calculating the full and empty ratio?
You do not need special software to calculate the full and empty ratio. It is a simple division of the
genome titer by the capsid titer. A standard calculator or Microsoft Excel will be fine.
Are you aware of full-and empty capsid assessments using ratios based on results from enzymelinked immunosorbent assays (ELISAs) and polymerase chain reaction (PCR)? Can you comment
on the differences between that method and yours?
Yes, there are publications for which a ratio calculation based on capsid titer from size exclusion
chromatography (SEC) or ELISA and the genome titer from qPCR was used for determining full
and empty ratios. These ratios rely on two vastly different methodologies, and therefore, data will
have compounded variability. That is one downside to consider. In addition, the genome titer from
qPCR methods often targets only the regions of inverted terminal repeat (ITR) sequences. This can
lead to overestimating the genome titer since capsids with partial sequences or partial genomes,
which contain the ITR but not the gene of interest, will be considered. With the presented method,
we clearly separate the intact genome from the partial genome and the small size impurities, and
therefore avoid overestimation of the genome titer.
Certain PCR workflows use DNAse or benzonase treatments before samples are analyzed. Do
samples need any pre-processing steps prior to CE analysis?
My recommendation is to do a simple extraction of the nucleic acid using commercially available
kits and heat the sample to avoid secondary structure formation prior to CE analysis. Since we
do not need to rely on amplification for CE-based genome integrity analysis while achieving high
sensitivity with fluorescent dye and laser-induced fluorescence detection, a pre-processing step
is not required. You can add a benzonase treatment step and subsequent inactivation and removal
of benzonase before nucleic acid extraction. Comparing the results of benzonase-treated to nontreated samples helps to decipher the amount and size range of nucleic acid impurities present
outside of the AAV capsid.
Can you elaborate on the time needed for different analytical techniques in comparison to your
method?
The exact time requirements for techniques, such as PCR/ELISA, electron microscopy (EM),
analytical ultracentrifugation (AUC), etc., will depend on the specific setup being used. I presented
estimates for each technique in my webinar, which are based on published literature. From the
comparison, you can see that it will take 2-3 workdays to perform comprehensive CQA analysis using
a combination of techniques and instrumentation, while these parameters can be assessed within a
typical workday using a single CE platform instead.
Read the entire Q&A blog >
Viral vectors Expert Q&A
14 | SCIEX | CGT compendium SCIEX.com/viral-vectors
With AAVs, 3 viral proteins (VPs) build the capsid. In the case of non-enveloped viruses, the
capsid is directly interacting with the host cells. Hence, integrity and PTM profiles of VPs are
important quality criteria that can affect viral uptake. Chromatographic separation of VPs can be
difficult to achieve, however, due to their similar physical properties. In addition, low- abundance
protein impurities can be missed.
Ensure the integrity of your capsid proteins with high-quality accurate mass data and
intuitive acquisition software
Set new frontiers for intact protein and impurity characterization with 3D visualization
options for deconvoluted data
Obtain relevant information on identities and quantities of proteoforms based on timeresolved deconvolution
Intact viral protein characterization
Obtain more information with increased ease using 3D deconvoluted data
Figure 6. 3D heatmap of
AAV8 capsid proteins cells
showing the intensity, the
retention time (RT) and
the molecular weight (MW)
using scan-by-scan, timeresolved deconvolution in
Biologics Explorer software.
Discover more details in the technical notes about AAV analysis using the
ZenoTOF 7600 system and the X500B QTOF system >
Viral vectors Intact protein characterization
Time (min)
Mass (Da)
Intensity
Figure 7. Results table
of identified AAV8 capsid
proteins with PTMs.
Average MW, RT and
calculated volume based
on 3D deconvolution is
shown using Biologics
Explorer software.
7600 X500B
15 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Sequence confirmation and identification of low-abundance PTMs require a deeper look into
the viral proteins. A peptide-mapping approach can provide information on product quality
attributes (PQAs) and CQAs. Low sample amounts, however, are a challenge for analytical
assays. Additional challenges include the identification of deamidation-derived isomers and
fragile PTMs that can affect the charge heterogeneity and, as a result, viral uptake.
Obtain high protein sequence and fragment coverage despite limited sample quantities
with highly sensitive, accurate mass data acquisition
Identify PTMs and their locations—including glycosylations, sulfations and
phosphorylations—with excellent spectral quality
Differentiate amino acid isomers and localizing fragile PTMs with an intuitive alternative
fragmentation technique
Viral protein peptide mapping
Achieve high MS/MS sequence coverage and fully understand challenging PTMs
Figure 8. Sequence
coverage map of AAV8
VP1. A nearly complete
sequence coverage (94.7%)
was obtained from a single
injection of a tryptic digest
of AAV8 using datadependent acquisition
(DDA) and processing with
Biologics Explorer software.
Viral vectors Peptide mapping
Figure 9. Identification of 3
deamidated species at N57
of the peptide YLGPFNGLDK
(z = 2). Top: Extracted ion
chromatograms (XICs) of
the different deamidated
species and the native
peptide. Bottom: Zoom-in
to EAD MS/MS data showing
signature fragment ions for
differentiation of aspartic
acid (Asp) and isoaspartic
acid (isoAsp) deamidated
forms of N57, for example
z5 - 57 for isoAsp and z5 - 44
for Asp.
Mass/charge Mass/charge Mass/charge
Intensity
Intensity
Intensity
Intensity
RT (min)
L-isoAsp
Asp 26.1
27.4
28.0
30.1
D-isoAsp
Native
z5+1-57
z5+1
z5+1-44
z5+1 z5+1-57
z5+1
L-isoAsp
27.4 min
Asp
28.0 min
D-isoAsp
30.1 min
100000
80000
60000
40000
20000
0
26 26.5 27 27.5 28 28.5 29 29.5 30
460 480 500 520 540
600
500
400
300
200
100
0
460 480 500 520 540
600
500
400
300
200
100
0
460 480 500 520 540
600
500
400
300
200
100
0
Learn more about AAV PTM analysis in this technical note >
16 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Viral vector characterization for vaccine and therapeutic drug development includes assessing
the viral proteins. While information on purity and protein ratios can be obtained with liquid
chromatography-based methods, an orthogonal approach with CE is advantageous to avoid
missing VP’ forms. High resolving power, throughput capabilities and reproducibility are
important factors for protein purity assays.
Determine protein-based titer with confidence and understand protein profiles and
impurities using high separation power
Reclaim your time with faster method development and the ability to run larger sample
batches
Avoid lengthy assay adjustments with a kit-based protein profiling workflow suitable
across serotypes and viral vectors
Protein purity
Viral vectors Protein purity
SCIEX.com/viral-vectors
Understand your viral proteins independently of the serotype
Figure 10. Separation of
VP proteins from different
AAV serotypes. Excellent
resolution of VP proteins
labelled with fluorescence
dye was achieved with CESDS-LIF.
Figure 11. CE-SDS-LIF of
Lentivirus proteome. 20
peaks were associated with
the lentivirus sample. Peak
9 was identified as the p24
proteins based on spike-in
experiments (not shown).
Relative fluorescence unit (RFU)
Time (min)
15 20 25 30
AAV1
AAV2
AAV8
VP3’
VP3’
VP3’
VP3
VP3
VP3
VP2
VP2
VP2
VP1
VP1
VP1
Relative fluorescence unit (RFU)
1
2
8 10 12 14 16 18 20 22 24 26 28
3
4 5
6
7
8 9
1011
12
13
14
15 16
17
18 19 20
Time (min)
p24
Discover more details in the technical notes about AAV capsid purity
assessment and lentivirus protein analysis >
AVV LENTIVIRUS
17 | SCIEX | CGT compendium
The integrity and purity of the viral genome are CQAs that impact vector potency,
immunogenicity and transduction efficiency. However, the limited sizing capabilities of some
analytical methods, can pose challenges for assessing the entire viral genome, especially
for viruses with larger genetic cargo. Furthermore, distinguishing between product-related
impurities, such as degraded genomes and intact genomes, and ensuring their accurate
quantitation can be challenging.
Confidently determine genome integrity, genome titer and impurities using high separation
power
Simplify viral vector genome analysis by avoiding lengthy assay adjustments with a
workflow suitable across serotypes and viral vectors
Run high-quality analyses smoothly and reproducibly with a kit-based turnkey solution
Streamline data management through compatibility with data management systems
Genome integrity and purity
Viral vectors Genome integrity and purity
SCIEX.com/viral-vectors
Assess genome integrity and nucleic acid impurities on one platform
Figure 12. High-resolution genome integrity analysis of AAV8 with CGE and LIF detection. The intact genome of 2.4 kb was
well separated from potential partial genomes and host cell ribonucleic acid (RNA), and DNA impurities.
5
0
25
50
75
7.5 10 12.5
Time (min)
Relative fluoresence units (RFU)
Host cell DNA
Host cell RNA
Partial genome
Intact genome
(2.4kb)
Take a deeper dive in the technical notes about AAV genome analysis and
lentivirus genome analysis >
AVV LENTIVIRUS
18 | SCIEX | CGT compendium
In addition to characterizing the genome and viral proteins, assessing the ratio of capsids with
an intact genome (full capsids) vs. partial or empty capsids is necessary for comprehensive
viral vector characterization. A variety of methods exist to determine this CQA, but the assays
can have limitations. For example, some assays are cumbersome, must be adjusted for each
serotype or require high levels of expertise, or provide limited understanding of partially filled
capsids.
Take back your time by assessing multiple CQAs with high-quality data on a single platform
with kit-based assays
Determine genome integrity, capsid proteins and full-and-empty ratios, including partial
capsids with serotype-independent workflows
Cover your compliance needs through compatibility with common data management
systems
Full-and-empty capsid ratios
Determine multiple CQAs, including full-and-empty ratios, on one platform
Viral vectors Full-and-empty capsid ratios
Figure 14. Comparison
of the percentages of full
capsids determined by
different technologies. A
good correlation between
transmission electron
microscopy and analytical
ultracentrifugation was
observed in comparison
to CE.
Discover more details in the technical note about full-and-empty AAV assessment >
Relative fluorescence units (RFU)
Relative fluorescence units (RFU)
Relative migration time (min) Relative migration time (min)
0.9 1.0 1.1
Genome titer
Corrected peak area
R2
= 0.9967 R2
= 0.9973
Corrected peak area
Capsid titer
0.8 0.9 1.0 1.1 1.2
Figure 13. Standard curves
for full-and-empty capsid
determination. Left: AAV
genome titer determination
with CGE. The linear dynamic
range (LDR) was determined
from 2.56x1010 GC/mL2.62x1013 GC/mL with
R² = 0.9967. Right: AAV
capsid titer determination
with LDR from 6.41x109
GC/
mL-2.62x1013 GC/mL with
R2
= 0.9973.
100%
80% 74.8%
83.4%
88.9%
60%
40%
20%
0%
Full capsids
Transmission electron
microscopy
Capillary
electrophoresis
Analytical
ultracentrifugation
19 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Host cell DNA (HCD) is a process-related impurity that can be present in cell culture-derived
products. Due to shearing during production, varying sizes of residual DNA might be present in a
product. Since DNA with >200 base pairs (bp) could encode for undesired proteins, reliable size
determination and simultaneous quantitation are crucial for product safety. Challenges arise for
risk assessment if only DNA quantity is determined without information on sizes.
Determine quantities and sizes of residual host cell DNA in your therapeutic or vaccine with
high resolving power and customizable size ranges
Achieve the highest sensitivity and quantitative performance when sample amounts are
limited with LIF detection
Cover your compliance needs through compatibility with common data management
systems
Residual host cell DNA
Determine risks of residual DNA using accurate size information and abundance
Viral vectors Residual host cell DNA
Figure 16. HCD fragment
size analysis of lentiviruses
by CGE-LIF. Upper panel
shows size marker with
200, 500, 1000 bp. Bottom
panel shows DNA extract of
lentivirus sample.
Figure 15.
Electropherogram showing
baseline resolution of 11
Hae III fragments of ΦX174
bacteriophage DNA with
fragment sizes shown
in bp.
0 5 10 15 20 25
Relative fluorescence unit (RFU)
Time (min)
dsDNA marker
Residual DNA in lentiviral sample
200bp
500bp
1000bp
Discover more details in the technical notes about HCD analysis of a
lentivirus sample and dsDNA analysis over an extended size range >
HCD dsDNA
12 14 16 18 20 22
Time (min)
Relative fluorescence units (RFU)
72 bp
118 bp
194 bp
234 bp
271 bp
281 bp
310 bp
603 bp
872 bp
1078 bp
1353 bp
0
20
40
60
80
Relative fluorescence units (RFU)
Time (min)
20 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Another class of process-related impurities that can impact the safety and efficacy of viral
vector products are proteins derived from packaging cells. The diverse landscape of packaging
cell lines and the desire to deliver relevant medicines to patients more quickly drive the need for
new strategies. Identifying process-related impurities with confidence requires highly adaptable
workflows that do not need months of development time for different viral vector products.
Streamline viral process development through relevant information on the identity and
quantity of HCPs
Avoid missing critical impurities using an unbiased data-independent acquisition (DIA)
approach with excellent coverage and detection depth
Take back your time for the identification and simultaneous quantitation of residual HCPs
without the need for lengthy assay development
Host cell protein identification
and quantitation
Identify HCPs with confidence using high-quality DIA
Viral vectors HCP identification
Figure 18. Comparison
of XICs of peptides in
complex matrix. Left:
XIC of precursor m/z of a
peptide shows high level of
interference affecting the
signal-to-noise and lower
limit of detection. Right:
XIC of a SWATH fragment
m/z of the same peptide
with reduced background
results in better signalto-noise and lower limit of
detection.
Figure 17. Schematic of
a SWATH window. Top:
Different precursors within
a given m/z range (SWATH
window) are present at a
given time point (colored
peaks) and selected for
fragmentation. Bottom:
The peak profiles of these
precursors and related
isotopes show slightly
different elution times,
which can be used for
deconvolution.
Time
Time (min)
Discover more details in the technical note for HCP analysis of lentivirus samples >
Time (min)
Intensity
MS1 MS/MS
analyte
0
500
1000
1500
2000
2500
3000
17.0 17.5 18.0 18.5 0
20
60
40
80
100
120
140
17.0 17.5 18.0 18.5
21 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Monitoring hundreds of protein impurities can provide valuable information about product quality
during process changes, such as upscaling, and reduce risks for the final product. While ligandbinding assays meet quantitation and throughput needs, obtaining actionable results can be a
challenge. Understanding which protein impurities have changed can provide tremendous insight
that can help streamline optimization of processes.
Understand product changes on a protein-specific basis without the need for months of
assay development
Move past bottlenecks and maintain flexibility when there are changes to packaging cell lines
and no ligand-binding assays are readily available
Achieve highly sensitive analyte detection, accuracy and precision
Confidently transfer assays to quality control (QC) with compliance-ready options and a
proven track record of supporting quantitation for good practice (GxP) environments
Monitoring of host cell proteins
Monitor and quantify hundreds of analytes with optimal quantitative performance
Viral vectors HCP monitoring
Figure 20. Quantitative
data from one surrogate
peptide transition in
biotherapeutic digest.
XIC shows LLOQ at 0.21
parts-per-million (ppm)
for chosen MRM transition.
Accuracy and precision
values achieved for 3
replicate injections of
different concentrations
of the target protein are
shown to the right.
Figure 19. XICs of peptide
surrogates for 48 target
proteins in a biotherapeutic
digest. More than 200
transitions were monitored
in a scheduled MRM with a
total run time of 8 min.
Discover more details in the technical note about quantitation of HCP peptides > Intensity
Time (min)
Time (min)
Intensity
LLOQ
0.21 ppm
22 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Viral vectors bear the risk of idiosyncratic integration into the host genome. In addition, gene
editing can affect the phenotype in various ways based on the complexity and interdependency of
protein networks. Genomic readouts cannot provide sufficient insights into the potential disruption
of gene regulators or detect changes to the proteome. Protein assays, such as Western blots, on
the other hand, are limited by antibody availability and cannot detect unexpected proteome-wide
changes.
Break through the boundaries of gene editing by seeing and identifying the unexpected
Unravel the effects of gene editing on the proteome level with DIA and dig deeper into
changes despite limited sample amounts
Achieve confident identification and simultaneous quantitation with excellent MS/MS data
quality
Proteome profiling
Monitor and quantify hundreds of analytes with optimal quantitative performance
Viral vectors Proteome profiling
Figure 22. Insertiondeletion (INDEL) errors
induced by gene editing
detected by bottom-up
proteomics using SWATH
DIA. The expectation is that
all peptides in the geneedited samples (blue) are
below the baseline signal.
However, compared to the
control (orange), only two
peptides at the N-terminus
of the were silenced based
on a shift in the initiation
of the protein transcription.
Bar thickness represents
standard deviation of the
peptides.
Figure 21. Identified and
quantified proteins using
SWATH DIA. Columns
show number of proteins
identified with false
discovery rate (FDR)
<1% for different loading
amounts of cell lysate
digest using a 60 min
gradient with 5 µL/min flow
rate. Line shows amount
of proteins with %CV<20%
used for quantitation as
percentage of total amount
of identified proteins with
FDR<1%.
Proteins with %CV<20
All identified proteins
Discover more details in the technical note about SWATH DIA for biomarker ID and quantitation >
Data courtesy of Allumiqs, Halifax, NS, Canada
23 | SCIEX | CGT compendium SCIEX.com/viral-vectors
RNA 9000 Purity
& Integrity kit
Suitable for:
Assessment of multiple CQAs of viral vectors
Rapid method development and larger sample sets
BioPhase 8800
system
RNA 9000 Purity
& Integrity kit
Viral vectors Analytical solutions
Suitable for:
Assessment of multiple CQAs of viral vectors
Analysis of host cell DNA
Smaller sample sets
PA 800 Plus
system
CE-SDS Protein
Analysis kit
BFS capillary
cartridge
SDS-MW Protein dsDNA 1000 kit
Analysis kit
SCIEX.com/viral-vectors
Analytical solutions for viral vectors
24 | SCIEX | CGT compendium
+
+
+
+
+
+
Analytical solutions for viral vectors
Viral vectors Analytical solutions
Suitable for:
Intact VP characterization
Peptide mapping including isomer differentiation and ID of challenging PTMs
High flexibility to perform a range of additional workflows
ExionLC AE
system
ZenoTOF 7600
system
SCIEX OS
software
Suitable for:
Intact VP characterization
Peptide mapping including ID of PTMs
Intuitive operation
ExionLC AE
system
X500B QTOF
system
SCIEX OS
software
Biologics Explorer
software
Biologics Explorer
software
SCIEX.com/viral-vectors
+ + +
25 | SCIEX | CGT compendium
+ + +
Tips and tricks from our application experts:
AAV analysis with LC-MS
Zhengwei Chen (PhD), Staff Applications Scientist at SCIEX, US, shares his tips and tricks on AAV
analysis using LC-MS/MS with the ZenoTOF 7600 system and EAD.
Dr. Zhengwei Chen serves as a Staff Application Scientist on the Biopharma Application Demo Team at SCIEX.
Zhengwei is an MS expert across a broad spectrum of biopharmaceutical workflows, such as intact and native mass
spectrometry, post-translational modifications, glycan analysis, proteomics and glycoproteomics. Zhengwei’s
expertise is built upon a strong academic background from Prof. Lingjun Li’s distinguished lab and multiple years of
experience at Regeneron, supporting all stages of drug development as part of an analytical chemistry group.
Tip 1: Sample handling
Utilize detergents at optimal concentrations, such as 6M urea or 8M guanidine hydrochloride, to ensure
your AAV capsids are thoroughly denatured. This step is crucial for enabling effective enzymatic digestion.
My recommendation is to use ~1 µg of the digested sample to assess the initial response when using an
analytical flow setup. Adjust the injection volume, aiming at a total ion current (TIC) signal of high E7 cps
using a ZenoTOF 7600 system.
Tip 2: Differentiation of amino acid isomers
EAD allows for effective differentiation between amino acid isomers, for instance between aspartic acid
(D) and isoaspartate (isoD), through diagnostic side chain fragments. When asparagine (N) in the capsid
proteins of AAVs undergoes deamidation, the isomers D and isoD can be formed, resulting in protein charge
variants, which can alter the physical and functional stability. I recommend employing a DDA experiment
with the EAD kinetic energy set to 7 eV. This approach will ensure extensive sequence coverage and enable
the precise identification of amino acid isomers.
Tip 3: Detailed characterization of phosphorylation
Surface-exposed tyrosine (Y), serine (S) and threonine (T) residues on AAV capsids can be phosphorylated,
followed by ubiquitination and degradation by the cell proteasome. Phosphorylation is therefore directly
linked to transduction efficiency and is an important PTM for AAV studies. EAD preserves the attachment
of the labile phosphate groups to the peptide backbone fragments and enables the precise pinpointing of
phosphorylation sites. This is particularly valuable in intricate scenarios where multiple phosphorylation
sites may exist on a single peptide. I suggest starting your analysis with a tryptic digestion followed by EAD
analysis. For more complex cases, employing enzymes like Asp-N before proceeding with EAD analysis may
enhance the confidence of identification.
Tip 4: Comprehensive glycosylation analysis
Glycosylation in AAVs is likely to impact gene delivery and expression by affecting viral tropism, entry
and infectivity. Leveraging EAD, complex glycosylation patterns can be unraveled, providing detailed
insights into the peptide backbone and pinpointing glycosylation sites precisely. Simultaneously, EAD can
distinguish glycan isomers, such as α2,3 and α2,6 sialic acids. This dual capability reveals intricate glycan
structures and enhances our understanding of the functional implications of glycosylation in viral vector
biology. I suggest starting your analysis by employing the intact glycopeptide method with the settings
outlined here.
More questions? Contact us >
Viral vectors Tips and tricks
26 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Tips and tricks from our application experts:
AAV analysis with CE
Peter Holper, Staff Applications Scientist at SCIEX, US, shares his tips and tricks on AAV analysis
using CE with the BioPhase 8800 system and the PA 800 Plus system.
Peter Holper has over 15 years of experience in biopharma, including his role as an analytical chemist at Eli Lilly and
Company where he was responsible for developing the analytical control strategy for bioproducts. He has extensive
experience in analytical method development for biologics and held various positions with increasing responsibility.
Peter currently works at SCIEX as a Staff Applications Scientist in Redwood City, California, where he is responsible for
developing and optimizing CE applications and providing customer demo sample analysis support.
Tip 1: Leverage the flexibility in injection modes
When starting out with a new viral vector product, my recommendation is to compare three different
modes of injection using UV detection. First, start with a standard electrokinetic injection, which allows
for the highest theoretical resolution. Next, use a pressure/hydrodynamic injection, which will inject the
same plug regardless of sample ionic strength and provide a quick estimate of the titer. Finally, use a fieldamplified sample stacking (FASS) injection to achieve the highest sensitivity, while understanding it is the
most sensitive injection method to the ionic strength of the matrix. Comparing these three peak profiles
can give significant insight into the optimal separation conditions for each molecule analyzed.
Tip 2: Deal with low sample amounts
During early-stage development of AAV vectors, oftentimes only a few micrograms of proteins or less are
available for analytics. However, most analytical technology is not practical for applications with low protein
concentration or small sample volumes. To improve the sensitivity of CE-SDS, my recommendation is to
use laser-induced fluorescence (LIF) detection instead of UV absorbance. Comparing the results from
the different injection types (tip 1) will help you determine if additional sensitivity and transition to LIF
detection is needed.
Tip 3: Optimize fluorescence dye labelling
Labeling procedure can pose challenges and require optimization for each product. Currently, the most
common fluorescent dye used in CE-SDS-LIF is Chromeo P503, which has a low quantum yield when not
bound to a protein and thus does not require additional cleanup after conjugation. When optimizing the
labeling procedure with Chromeo P503, I find the dye-to-protein ratio to be the most important factor. If
this ratio is not optimal, low signal or high peak tailing is often observed. I find that estimating the protein
titer by referring to the peak area achieved with pressure injection (tip 1) can be highly beneficial, since only
the genome titer may be known at this point.
More questions? Contact us >
Viral vectors Tips and tricks
27 | SCIEX | CGT compendium SCIEX.com/viral-vectors
Plasmid DNA
Plasmid DNA Overview
SCIEX.com/plasmid-DNA
pDNA restriction map
Achieve excellent resolution over a
large size range for fragment-based
ID of dsDNA.
Residual nucleic acids
Understand the sizes and amounts of
residual host cell nucleic acids.
pDNA purity
Separate plasmid isoforms with high
resolution and assess the purity and
stability of your pDNA.
Linear DNA size
Determine accurately the size of your
linearized pDNAs over a wide size range.
Roman Herzog (PhD)
Group Leader Bioanalytics (R&D), WACKER
Chemie AG
“Plasmid DNA (pDNA) is a widely used
starting material in the manufacturing
process of mRNA-based vaccines or
viral vectors. Consequently, a high pDNA
quality must be ensured. Capillary gel electrophoresis with
laser-induced fluorescence (CGE-LIF) on a PA 800 Plus system
from SCIEX offers accurate and highly sensitive pDNA analyses,
enabling a reliable assessment of pDNA quality prior to further
processing.”
Double-stranded DNA plasmids are an extremely versatile
tool frequently used for genetic engineering in biotechnology
applications. In a medical context, plasmid DNA (pDNA) can
be used directly—as vaccine or for ex vivo cell therapy for
instance—but also serve as raw or critical starting material for
the manufacturing of protein drugs, viral vectors, and mRNA.
Generally, pDNA contains several regions to enable its function
within drug manufacturing: An origin for the replication in
bacteria, the gene of interest (GOI), a promoter to enable the
expression of the GOI, antibiotic resistance genes for selection
as needed and in case of viral vector production, long terminal
repeats (LTRs). The pDNA quality directly impacts the quality of
subsequent protein, nucleic acid or viral vector products and
must therefore be ensured.
28 | SCIEX | CGT compendium
Expert Q&A: Plasmid manufacturing
Plasmid DNA serves a variety of purposes—from critical starting material for proteins, mRNA or viral
vectors to drug substance. Here, Dr. Emma Bjorgum, an expert in plasmid manufacturing provides
insights into the process and an outlook on the future.
Plasmid DNA Expert Q&A
Emma Bjorgum is the Vice President of Client Services of the DNA Business Unit at Aldevron with a focus on product
strategy and portfolio management. She has been employed in the cell and gene therapy industry for over a decade
with 9 years of experience at Aldevron. Before Aldevron, Emma worked for Millipore Sigma as a Business Development
Manager for the Viral and Gene Therapy Manufacturing business unit. She also worked for Be The Match Biotherapies
as a Business and Market Analyst. Emma obtained her BA in Biology with minors in Chemistry and Psychology from
Concordia College in Moorhead, MN.
SCIEX.com/plasmid-DNA
What applications does Aldevron manufacture plasmid DNA for?
Aldevron manufactures plasmid DNA for a variety of end applications. Much of our experience and expertise
is comprised of manufacturing plasmid DNA for cell and gene therapy applications. We also manufacture for
all phases and stages of pipeline development from early discovery to commercial applications. Aldevron is
among the first to offer plasmid DNA at full current Good Manufacturing Practice (cGMP) or clinical grade and
has pioneered a mid-grade between research grade and full cGMP, called GMP-Source. This has allowed us to
support an estimated 1500 clinical trials run by over 1000 clients. We thrive on supporting clients from the early
stages of their clinical programs through commercialization.
How does your support vary by application?
We can provide plasmid DNA for various applications. Two specific examples include support of mRNA and
AAV gene therapies and vaccines. For mRNA applications, we provide plasmid DNA as a linearized product and
can perform the linearization with a client-designated enzyme. We also screen the plasmid construct prior to
manufacturing to optimize conditions for both yield and stability of the poly(A) tail (if encoded). For AAV drugs,
we optimize conditions for scale up by evaluating different host cell lines and temperature combinations
for inverted terminal repeat (ITR) retention. A third example is our investment in next-generation plasmid
technology, Nanoplasmid vectors. Nanoplasmids are comprised of very small, efficient backbones (~500 bases).
Removal of bacterial and antibiotic resistance genes improve both safety and performance. One area where
Nanoplasmids are showing particularly strong performance is as a homology-directed repair (HDR) donor
template for CRISPR knock-in applications.
How do you ensure the quality of your plasmids at the different quality levels you offer?
Aldevron offers a comprehensive quality control testing panel of assays for release of plasmid DNA. Assays
include various methods for identity, safety, bacterial host components and bioburden/sterility. Almost all our
assays are conducted in-house, and methodologies are closely aligned for testing and release of RUO, GMPSource and GMP methods.
How have the requirements for plasmids changed over the past 5-10 years?
In the earlier days of cell and gene therapy, there were hardly any references to plasmid manufacturing
recommendations where plasmid DNA is utilized as a critical starting material or raw material. As cell and
gene therapy has continued to see additional approvals, we have seen more recent considerations from the
agency for CAR-T therapies with a recommendation to remove any unnecessary transgene in the vector such
as antibiotic resistance markers. Aldevron’s Nanoplasmid technology ameliorates this concern as it utilizes a
sucrose selection technology negating the need for any antibiotics in the manufacturing process.
29 | SCIEX | CGT compendium
Plasmid DNA Expert Q&A
SCIEX.com/plasmid-DNA
What changes do you anticipate moving forward?
Moving forward, we are likely to see additional scrutiny on vector backbones and the removal of any extraneous
sequences. We are also likely to see increased specificity on scale and how manufacturers can deliver exactly
what is needed at the point in time of clinical development. Aldevron is focused on providing the ‘right sized’
scale for manufacturing and can meet both exact quantity and batch deliverables.
What innovation is helping to drive the industry forward and how will analytics need to evolve?
Newer vector technologies, such as nanoplasmid, can help address concerns with extraneous sequences in the
plasmid backbone size since it consists of only 200 bp. Another innovation area is next-generation microbial
cell lines to improve the yield and stability of plasmid DNA, such as the REVIVER cell line. Additionally, non-viral
delivery systems are tackling challenges in the industry for payload delivery by lowering costs and delivering
products without the constraints of a viral system. Additionally, innovation around the client experience is a key
focus for us. Over the past 2 years we have been intensely focused on the client experience and have made
incredible progress streamlining the new program on-boarding process, reducing lead times and eliminating
deviations. For example, in 2023, we were able to reduce our lead time by up to 80% from construct selection
through product release.
mRNA is driving industry growth. How does Aldevron support the mRNA modality from a plasmid perspective?
Aldevron can provide linear plasmid DNA at any scale and quality level (RUO, GMP-Source and GMP). Our
processes allow for linearization with the client-selected enzyme, including a purification step post-linearization
to ensure the product is free from any remaining enzyme. We can provide analytical testing for the final
linearized product to confirm the percentage of linearized plasmid in addition to poly(A) tail length. Several of
our clients get linear plasmid DNA from us and do the IVT and other reactions internally. Increasingly, clients are
taking advantage of Aldevron’s broader RNA services, including linear plasmid, IVT and capping reactions, lipid
nanoparticle encapsulation and sterile fill-finish services. That includes all the associated analytics, such as
CGE, for instance.
What additional services are popular with those manufacturing plasmid for clinical applications?
Additional services often required to support plasmid DNA for clinical services include stability testing of
both final plasmid DNA product and master cell banks. Commercialization support services such as process
characterization and process validation are also often required in the late phases of clinical development.
Additionally, regulatory services are often utilized to support Chemistry, Manufacturing and Control (CMC)
sections of Investigational New Drug (IND) filings or Biologics License Applications (BLAs).
30 | SCIEX | CGT compendium
Plasmids can exist in three primary topological forms: covalently closed circular (ccc) often referred
to as supercoiled (sc), open-circular (oc) and linear. The sc form is desirable during plasmid
manufacturing and for subsequent protein expression, viral vector manufacturing or DNA vaccines.
Differentiating conformational isoforms and assessing the purity and stability of pDNA is crucial for
ensuring product quality, whether it is the critical starting material or drug substance.
Rely on excellent resolution for different topological variants of pDNA
Achieve high sensitivity for early-stage development samples with LIF detection
Confidently transfer assays from development to QC with excellent precision and streamline
data management through compatibility with data management systems
pDNA topology and purity
Differentiate different topological variants and determine purity with ease
Figure 24. Degradation
monitoring of pDNA
samples. Results from
pDNA with 7 kb, stressed
at 40°C for up to 12 weeks,
show a good correlation
between a decrease
in sc and increase in
degradation product (oc).
Figure 23. Determination
of different plasmid
isoforms (5 kb). Different
amounts of linear, sc and
oc forms were detected in
the different samples.
Discover more details in the technical note about plasmid stability monitoring >
Plasmid DNA Topology and purity Relative fluorescence units (RFU)
Time (min)
Sample 1
Sample 2
Sample 3
Sample 4
Linear
Supercoiled
Nicked (open circular)
0 5 10
Time (min)
Relative fluorescence units (RFU)
Supercoiled Open circular
Relative fluorescence units (RFU)
Time (min)
31 | SCIEX | CGT compendium SCIEX.com/plasmid-DNA
Linearized DNA serves as a template for mRNA and other IVT RNAs, and minimizes off-target or
elongated mRNA transcripts due to read-through transcription. The linearization efficiency of pDNA
is therefore an important quality attribute of DNA starting material. Furthermore, sizing of the
linearized plasmid and assessment of its purity can help determine the quality of linearized pDNA.
Determine linearization efficiency with excellent separation for different topological variants
of pDNA
Assess linear DNA sizes and purity confidently with ultra-high resolution over a wide size
range
Confidently transfer assays from development to QC and streamline data management
through compatibility with data management systems
pDNA linearization efficiency and sizing
Understand linearization efficiency and linear DNA sizes
Figure 26. Replicate
injections of plasmid
sample. The overlay of
18 replicate CGE-LIF
analyses shows excellent
reproducibility of the
method.
Figure 25. Confirmation of
different isoforms for a
7 kb plasmid. The untreated
plasmid shows two peaks
related to ccc and oc
forms. The linearized
plasmid shows one peak
that can be attributed to
the linear form.
Discover more details in the technical note about plasmid purity testing >
Plasmid DNA Linearization efficiency and size
Time (min)
Relative fluorescence units (RFU)
0 5 10
Time (min)
Relative fluorescence units (RFU)
Untreated plasmid
Linear
8 9 10
0
40
32 | SCIEX | CGT compendium SCIEX.com/plasmid-DNA
Several analytical techniques for plasmid identity testing exist. However, homologous regions,
such as poly(A) tails, LTR and ITR, present a challenge for sequencing-based methods. The
repetitive nature of these regions makes it difficult to obtain accurate information on their length
and composition. Tailored restriction mapping and analysis with high resolving CGE provide an
alternative that is not affected by long, homologous pDNA regions.
Achieve identity testing with excellent resolution of DNA restriction fragments over a wide
size range
Rely on results with excellent accuracy and precision
Confidently transfer assays from development to QC and streamline data management
through compatibility with data management systems
pDNA restriction map
Determine pDNA fragment sizes across a wide range
Figure 27. DNA restriction map analyzed with CGE. Electropherogram shows baseline resolution of 11 Hae III fragments of
Φ X174 bacteriophage DNA with fragment sizes shown in base pairs.
Discover more details in the technical note about dsDNA analysis
over an extended size range >
Plasmid DNA pDNA restriction map
12 14 16 18 20 22
Time (min)
Relative fluorescence units (RFU)
72 bp
118 bp
194 bp
234 bp
271 bp
281 bp
310 bp
603 bp
872 bp
1078 bp
1353 bp
0
20
40
60
80
Relative fluorescence units (RFU)
Time (min)
33 | SCIEX | CGT compendium SCIEX.com/plasmid-DNA
After transforming bacteria with the desired plasmid and selection of a clone, fermentation is used
for the expansion of pDNA. Extracted pDNA from the lysed host cells is concentrated and cleaned
up. Quality assessment therefore includes analytical testing for residual host nucleic acids as a
process-related impurity. Reliable size determination and simultaneous quantitation of residual
DNA and RNA are crucial to enable risk assessment and ensure product safety.
Determine size and amounts of residual host cell nucleic acids
Rely on results with excellent accuracy and precision
Achieve high sensitivity for early-stage development samples with LIF detection
Residual host cell nucleic acids
Assess residual nucleic acid sizes and quantities simultaneously
Figure 28 Electropherogram with 200 bp marker. The host cell nucleic acids migrating slower than the marker can be
attributed to impurities of sizes >200 bp that are considered high-risk residuals due to the potential for unwanted genes
encoding.
Plasmid DNA Residual host cell nucleic acids
Time (min)
0 2 4 6 8 10 12 14
200 bp marker
High-risk residual DNA
0
4
8
12
16
Relative fluorescence units (RFU)
SCIEX.com/plasmid-DNA
More questions? Contact us >
34 | SCIEX | CGT compendium
Analytical solutions for plasmid DNA
Plasmid DNA Analytical solutions
Suitable for:
pDNA purity analysis
Linear DNA sizing and fragment assessment
Residual nucleic acid analysis
PA 800 Plus system dsDNA 1000 kit
SCIEX.com/plasmid-DNA
+
35 | SCIEX | CGT compendium
IVT RNA
IVT RNA Overview
SCIEX.com/messenger-RNA
mRNA integrity
Separate impurities from your IVT RNA
and assess the integrity and purity of your
product.
Circular RNA
Ensure separation of circular products
from linear precursors and assess product
purities.
poly(A) assessment with MS
Leverage LC-MS for the characterization of
your mRNA 3’ ends.
5’ end cap
Achieve identification and
quantitation of capping
structures and intermediates.
Encapsulation
Determine
encapsulation
efficiencies of
mRNA in LNPs
and simultaneously
assess mRNA
integrity.
poly(A) assessment with CE
Characterize the poly(A) tail
length and distribution of your
IVT RNA with CE.
saRNA and srRNA integrity
Break through boundaries for assessing purity
and integrity of large RNA products beyond 9 kb.
Protein expression
Identify and quantify
expressed proteins and
characterize PTMs of
proteins.
Jérémie Parot (PhD)
Research Scientist, SINTEF
“Given the “one break no effect” with
mRNA, the integrity of mRNA drugs
must be assessed to ensure its
quality. Capillary gel electrophoresis
(CGE) is a key tool in this field. CGE systems, such as the
BioPhase 8800 system from SCIEX, offers high resolution and
can efficiently characterize RNA profiles, enabling accurate
quality control. Ensuring the integrity and purity of mRNA
formulations is essential for the development of effective and
safe therapeutics.”
Discovered in the 1960s, mRNA has had a long lead time,
approved regulatorily for the first time as a vaccine in 2021. For
the large and fragile cargo to enter cells effectively, the delivery
mechanisms needed to evolve amongst other factors. Today,
different in vitro transcribed RNA (IVT RNA) types, such as linear
mRNA, large self-amplifying RNA (saRNA) or self-replicating RNA
(srRNA), and circular RNA without 5’ and 3’ ends are approved
for drugs or are in clinical phases. Their usage in medicine is
highly diverse as seen by their application in vaccines, gene
editing, replacement therapy and neoantigen expression. For
any drug application, the comprehensive characterization of the
large and often heterogeneous IVT RNA is critical.
36 | SCIEX | CGT compendium
Expert Q&A: Addressing stability
challenges of mRNA-LNPs
mRNAs are fragile molecules that are not meant to be very stable. Increasing their stability requires
careful assessment of multiple factors. Here, Dr. Jingtao Zhang and Daniel Turner (Catalent® Pharma
Solutions) provide insights into their assessments using CGE.
Dr. Jingtao Zhang is the Scientific Director in the Biologics Group at Catalent® Pharma Solutions. He is responsible for
developing new technical capabilities and product solutions to solve clients’ pressing pharmaceutical problems with a special
focus on expanding mRNA-LNP within Catalent Pharma Solutions. He has more than 16 years of experience in R&D and
commercialization of small molecules, peptides, oligonucleotides, biologics and mRNA-LNP drug products in pharmaceutical
and CDMO settings. Jingtao received his PhD in Chemical Engineering from the University of Wisconsin-Madison and authored/
co-authored more than 30 articles in peer-reviewed journals.
IVT RNA Expert Q&A
Daniel Turner serves as an Associate Scientist at the Bloomington Development Center of Catalent® Pharma Solutions focused
on biologics formulation development. Daniel has gained experience in analytical and formulation development by supporting
numerous client projects. He has optimized the separation of biomolecules and established assays using size-exclusion
chromatography (SEC), capillary isoelectric focusing (cIEF) and CGE. Most recently, he has been supporting LNP formulation
development and optimizing CGE methods for mRNA-LNP purity quantification. Daniel obtained his BS in Biology with a minor in
Chemistry from Indiana University.
Can you provide details for the conditions of your pre-injection rinses?
Prior to starting a sample sequence, we perform an acid wash, a water wash and then fill the capillaries
with gel. As part of our optimized method, we use 70 psi for 2 minutes for the acid wash, followed by a water
wash at 70 psi for 2 minutes. To fill the capillaries with gel, we use 50 psi for 5 minutes. Once the capillaries
are filled with gel, we run the separation method briefly—for 2 minutes—to pull out any small impurities
before moving ahead to any samples. In addition to pre-injection rinses, we found it extremely important to
use sample loading solution (P/N 608082) to dilute samples prior to injection. It helped us improve the peak
shape and resolution of our mRNA samples and the RNA ladder.
Can you elaborate on your settings for the pressure injection of mRNA?
We used 1 psi for 5 seconds. However, you can adjust the duration depending on your needs. We have
also used electrokinetic injection in the past, which applied -1.0 kV for 6 seconds to load samples into the
capillaries. This method also worked well and required less sample, but we decided to stick with pressure
injection since it provides higher reproducibility in our experience.
Is there a size limit for mRNA analysis on the BioPhase 8800 system?
I’m not aware of a specific size limit on the BioPhase 8800 system. The ladder from the RNA 9000 Purity
& Integrity kit covers a range from 500 nt to 9,000 nt. While most mRNA we work with is within this
range, larger mRNA can be analyzed when extending the run time, with the caveat of working outside of
the calibration curve range which would impact sizing accuracy. This would be something interesting to
investigate.
Which capillaries do you use for mRNA analysis?
The capillaries we used for the BioPhase 8800 system are bare-fused silica (BFS) capillaries as part of the
BioPhase 8800 BFS capillary cartridge (P/N 5080121). The cartridge contains 8 pre-installed capillaries,
each 30 cm long, and a detection window 20 cm from the inlet. Liquid-based temperature control of the
separation temperature is incorporated with these cartridges.
37 | SCIEX | CGT compendium SCIEX.com/messenger-RNA
Read the entire Q&A blog >
IVT RNA Expert Q&A
Can you comment on the transferability of your extraction method with the Triton X-100 and temperature
settings for CGE analysis to other mRNA products?
In general, the method can be transferred to different mRNA products. However, I suggest optimizing the
surfactants and the temperature settings since different mRNA products can be more susceptible to secondary
structure formation, aggregation or formation of multimers, and may have different sensitivity towards
temperature. You really want to make sure that your surfactant concentration is suitable for your mRNA
product. In case you are getting poor recovery, you would want to investigate surfactant types or concentration.
Since different mRNA products exhibit different sensitivities to temperature, I recommend optimizing sample
incubation temperatures. For instance, a test range of 40 to 70 degrees Celsius, including different incubation
times for a given temperature, is a good starting point. I also recommend optimizing your cartridge temperature
as well to reduce the formation of secondary structures.
Did you find that the recovery of mRNA from your LNPs is dependent on the type of ionizable lipid used?
We tried several types of ionized lipids and different compositions of LNPs. For the ones we tried, we have not
experienced significant issues related to recovery using an optimized extraction method. We did observe that
non-optimized extraction methods, including sample preparation such as surfactants level and denaturants,
could affect recovery.
Are you able to comment on other approaches regarding stability, such as lyophilization?
The approach we currently take is focused on preserving the formulation through freezing. This adds
inconvenience and increases costs. During lyophilization, water is removed from the LNP system, and a product
can be stored in refrigerated conditions, potentially even at room temperature. The requirements for upholding
a suitable cold chain would be dramatically reduced. Alternative solutions for storage of mRNA products,
like lyophilization, are therefore very interesting. A lot of work still needs to be done, particularly regarding
lyophilization of LNPs.
Can you comment on how excipients affect mRNA integrity?
Various excipients—lipid and non-lipid excipients—play an especially important role in stabilizing the overall
product and can also affect the active ingredient, the mRNA. For instance, it is known that secondary structures
of mRNA are pH dependent. Excipients, such as buffer salts, that modulate the pH can therefore affect the
mRNA structure. Lipid excipients are crucial to the drug’s efficacy and tolerability. As a result of this and stability
concerns, we need to pay a lot of attention to their quality. Impurities in excipients can lead to degradation of
mRNA. A now well-known example is the reaction of mRNA with aldehydes, which can exist as impurities in
lipids. When evaluating excipients, my suggestion is to decouple the effects of the excipients from those related
to the excipients’ quality.
Is there any carrier system being used for mRNA other than LNPs?
Up until now, LNPs leveraging ionizable lipids are the most clinically validated system for mRNA delivery. LNPs
come in different flavors, usually using ionizable or cationic lipids as a key component, and can complex the
negatively charged mRNA cargo. Some LNP research focuses on the usage of biodegradable lipids to improve
the biocompatibilities while others have focused on targeted systems for enhanced efficiencies. Other systems
can be used for non-viral delivery, such as polymers or cell-penetrating peptides (CPP). In some cases,
polymeric systems can be coupled with LNPs to gain the best of both worlds. It’s an exciting field with a lot of
ongoing research.
38 | SCIEX | CGT compendium SCIEX.com/messenger-RNA
The stability of fragile RNA cargo requires careful testing during development because mRNAbased drugs can lose their efficacy when truncated. In addition, process-related nucleic acid
impurities may pose safety concerns. The integrity and purity of mRNA constructs are therefore
important aspects for product quality. High resolution separation with excellent reproducibility
enables the assessment of these product quality parameters accurately and reliably.
Break through analytical boundaries with ultra-high resolution and excellent reproducibility
Determine the integrity and purity of your nucleic acid products from 50 up to 9,000
nucleotides (nt) and beyond
Confidently transfer assays from development to QC and streamline data management
through compatibility with data management systems
mRNA integrity and purity
Confirm integrity and determine purity with excellent resolution and reproducibility
Figure 30. Reproducibility
across all 8 capillaries of
the BioPhase 8800 system.
The electropherograms of
the single-stranded ladder,
spanning from
150-9,000 nt, show full
separation and excellent
reproducibility for the
entire size range of the
ladder from the RNA 9000
Purity & Integrity kit.
Figure 29.
Electropherogram of
mRNA extracted from an
LNP analyzed using the
BioPhase 8800 system.
The mRNA of 1.929 kb was
encapsulated in an LNP
with MC3 as the ionizable
lipid.
Discover more details in the technical note about mRNA integrity and purity assessment >
IVT RNA mRNA integrity and purity
Time (min)
Relative fluorescence units (RFU)
3.0
2.0
1.0
0
12.5 15.0 17.5 20.0
SCIEX.com/messenger-RNA
Time (min)
Time (min)
Relative fluorescence units (RFU) Relative fluorescence units (RFU)
39 | SCIEX | CGT compendium
Conventional mRNA and base-modified mRNA (bmRNA), which incorporates chemically modified
nucleotides, are non-replicating IVT RNAs. SrRNA is an emerging third type that is based on an
engineered viral genome, devoid of viral structural protein genes. The self-replicating ability makes
srRNA a promising tool for new therapeutic drugs, despite challenges with its length.
Determine the integrity and purity of your srRNA even beyond 9,000 nt
Break through analytical boundaries with ultra-high resolution and excellent reproducibility
Confidently transfer assays from development to QC and streamline data management
through compatibility with data management systems
srRNA integrity and purity
Confirm integrity and monitor impurity profiles with high resolution
and reproducibility
Figure 31. Electropherogram of research-grade srRNA material (16 kb) obtained from Replicate Bioscience, San Diego, CA, USA.
IVT RNA srRNA integrity and purity
More questions? Contact us >
Time (min)
16 kb srRNA
Relative fluorescence units (RFU)
15 20 25 30
40 | SCIEX | CGT compendium SCIEX.com/messenger-RNA
Circular RNAs are next-generation IVT RNAs that provide the benefit of high resistance towards
exonucleases—without 5’ caps or poly(A) tails. To achieve circular RNAs, linear precursors are
chemically or enzymatically ligated. Understanding the purity of the circular product requires a
high-resolution separation workflow, which can separate linear precursors, degradation and high
molecular weight products from the desired circular RNA.
Take charge of your product quality and determine the efficiency of your circulation processes
Break through analytical boundaries with ultra-high resolution and excellent reproducibility
Confidently transfer assays from development to QC and streamline data management
through compatibility with data management systems
Circular RNA assessment
Fully separate linear precursors from circular RNAs with the highest reproducibility
Figure 33. Sensitivity
assessment for linear RNA
impurity. The circular RNA
product was spiked with
a serial dilution of linear
precursor. A detection
limit of 0.1% relative to
the circular product was
determined. Green trace:
ssRNA ladder from RNA
9000 Purity & Integrity kit.
Figure 32. Separation
of circular from linear
precursor RNA with CGELIF. The linear RNA product
migrates faster through
the gel matrix than the
circular RNA product due to
a smaller effective crosssection.
Discover more details in the technical note about circular RNA assessment >
IVT RNA Circular RNA assessment
14 15 16 17 18
5
10
15
13 14 15 16 17 18
0
5
10
15
20
25
Ladder Linear RNA
Circular RNA
SCIEX.com/messenger-RNA
Circular RNA
Linear RNA
Time (min)
Time (min)
Relative fluorescence units (RFU) Relative fluorescence units (RFU)
41 | SCIEX | CGT compendium
LNPs are designed to keep the fragile genetic cargo safe. Degradation and loss of function of
nucleic acids is accelerated if they are not sufficiently encapsulated. In addition, the cellular uptake
of the drug can be impeded. The encapsulation efficiency of the genetic cargo is therefore an
important quality criteria to be optimized and monitored during the development of LNP-based
drugs.
Take charge of development decisions by understanding encapsulation efficiencies of drug
substances with a reliable kit-based CE workflow
Determine free and encapsulated mRNA amounts with excellent repeatability and sensitivity
Simultaneously monitor degradation products in your samples leveraging exceptional
resolving power
Encapsulation efficiency of mRNA
Achieve it all: Excellent resolution, sensitivity and linearity with low %CVs
Figure 35. Results of
replicate injections
of different mRNA
concentrations.
Reproducible peak areas
with very low %CV and
very high accuracy were
determined based on
triplicates.
Figure 34. Serial dilution
of mRNA standard.
Electropherograms show
excellent migration time
reproducibility. Corrected
peak area vs. mRNA
concentration showed a
linear correlation with
R2
= 0.9989.
Discover more details in the technical note about mRNA encapsulation efficiency analysis with CE >
IVT RNA Encapsulation efficiency
0
10
20
30
14 16 18 20 Corrected peak area
Concentration (μg/mL)
Nominal Measured (µg/mL)
(µg/mL)
%CV Accuracy
(%)
#1 #2 #3 Mean
400
500
600
387
494
574
390
489
580
382
480
570
386
489
575
1.1
1.7
0.9
97
98
96
SCIEX.com/messenger-RNA
Time (min)
Relative fluorescence units (RFU)
42 | SCIEX | CGT compendium
The 5’ cap of IVT mRNA has a direct impact on its stability and translation efficiency and is
therefore considered a CQA. Since G cap, cap 0 and the mature cap 1 are linked to different
pharmacological efficacies, detailed characterization and simultaneous relative quantitation is
needed to ensure product quality. The differences between the different capping structures are
only 1-2 methyl groups, which requires high resolving power to be distinguished.
Characterize 5’ caps and intermediate products reliably using excellent time-of-flight (TOF)
MS data quality
Obtain relative quantitative information automatically or tailor quantitative calculations
specifically to your needs
Take back your time with intuitive acquisition and processing software
5’ capping analysis
Understand your product quality with ease using high-quality data
Figure 37. Results of
identified mRNA capping
moieties. Table shows
identified species,
associated molecular
weight and relative
quantitative information
using Molecule Profiler
software.
Figure 36. Deconvoluted
data for mRNA 5’ end
with isotopic resolution.
Different capping
intermediates and mature
cap 1 species as well as
sodium (Na) and potassium
(K) adducts were identified.
IVT RNA 5’ capping analysis % Intensity
Mass (Da)
G cap
Cap 1 Na
Cap 1
Cap 0 Cap 0 Na Cap 0 K
8330 8340 8350 8360 8370 8380 8390 8400
0%
10%
20%
30%
40%
50%
60%
70%
80%
90%
100%
SCIEX.com/messenger-RNA
More questions? Contact us >
43 | SCIEX | CGT compendium
Polyadenylation is needed to enable product stability and translation efficiency of mature mRNA
products. As a result, the length and distribution profile of the poly(A) tail during development are
highly relevant concerns, whether the tail is template-encoded, enzymatically added or applied
through a combination of these approaches. The detailed characterization of poly(A) tails requires
accurate data with high resolving power.
Leverage excellent data quality through exceptional negative ionization efficiency and
declustering of adducts with state-of-the-art source design
Uncover relevant information on poly(A) tails, such as molecular weight and distribution
profiles
Trust in your high-resolution results with great mass accuracy
3’ end poly(A) tail of mRNA with LC-MS
Dig deeper into your product quality with high-quality data
Figure 38. Poly(A) tail analysis from digested mRNA using LC-MS. Deconvoluted TOF-MS data shows the heterogeneity of the tail length of ~90 nt in
the mRNA sample. Each Δ amu of 329.2 Da contributes to the addition of 1 adenine (A) nucleotide in the polynucleotide. Data courtesy of Phenomenex,
CA, USA.
IVT RNA poly(A) tail analysis
More questions? Contact us > % Intensity
Mass, Da
A A A A
91 nt total
85 As
100%
80%
28000 29000 30000 31000 32000 33000
60%
40%
20%
0%
44 | SCIEX | CGT compendium SCIEX.com/messenger-RNA
Since the 3’-end poly(A) tail is a CQA affecting product stability and translation efficiency,
its optimization and monitoring throughout development and manufacturing is critical. The
determination of length and distribution profiles require accurate assays with high resolving power
and repeatability. To enable implementation in QC environments, intuitive and robust assays are
needed.
Take control of your IVT RNA 3’ CQA with reproducible, high-quality CE data
Dig deeper than ever before into the dispersity of your 3’ poly(A) tails with single-nucleotide
resolution
Confidently transfer assays from development to QC and streamline data management
through compatibility with data management systems
3’ end poly(A) tail of mRNA with CGE-UV
Enable yourself with single-base resolution and excellent reproducibility
IVT RNA poly(A) tail analysis
Figure 40. mRNA poly(A)
tail reproducibility analysis
with CGE-UV. The 4
replicate injections show
high reproducibility in
terms of migration time
and peak profiles.
Figure 39. Poly(A) tail
analysis from mRNA with
CGE-UV. Electropherogram
shows single-base
resolution of mRNA poly(A)
tails with most abundant
species of 121 nt in length.
Absorbance (AU)
0.0000
0.0005
0.0010
0.0015
0.0020
39 40 41 42 43 44 45 46 47 48
Time (min)
AU AU
Time (min) Time (min)
0.0000
0.0005
0.0010
0.0015
0.0020
39 40 41 42 43 44 45 46 47 48 49
Injection #1
0.0000
0.0005
0.0010
0.0015
0.0020
39 40 41 42 43 44 45 46 47 48 49
Injection #4
0.0000
0.0005
0.0010
0.0015
0.0020
39 40 41 42 43 44 45 46 47 48 49
Injection #2
0.0000
0.0005
0.0010
0.0015
0.0020
39 40 41 42 43 44 45 46 47 48 49
Injection #3
Discover more details in the technical note about poly(A) tail analysis with CGE >
Poly(A) tail
45 | SCIEX | CGT compendium SCIEX.com/messenger-RNA
Upon successful delivery of the genetic cargo, drug substances such as IVT RNA are supposed to
induce protein expression. While ELISAs and western blots are widely used to determine functional
potency, these assays are limited by the availability of antibodies with high specificity for the target
protein. Flexible approaches to determine protein expression that do not rely on antibodies can
help with adhering to timelines and fast-paced changes in development pipelines.
Break through the boundaries of complex matrices and achieve reliable identification and
excellent quantitation simultaneously
Leverage impeccable quantitative performance for decision making with high linear dynamic
range and low limits of detection and quantitation
Streamline your quantitative data processing with state-of-the art software
Protein expression analysis
Don’t let matrices hold you back—level up your MS/MS sensitivity
IVT RNA Protein expression
Figure 42. Peptide
quantitation in complex
matrix. Calibration curve
obtained for a peptide in
complex matrix using the
sum of multiple fragment
ions enhanced by the
Zeno trap. Inset shows
a comparison of peak
intensities obtained when
summing the same peptide
fragment ions with the
Zeno trap enabled (pink)
and without (blue).
Figure 41. Comparison of
matrix blanks. Same matrix
blank was injected onto a
triple quad system and the
ZenoTOF 7600 system. The
high resolution on the TOF
system resulted in a clean
blank and therefore lower
limits of quantitation for a
peptide analyte compared
to the triple quad data
obtained.
Discover more details in the technical note about peptide quantitation in matrix >
Time, min
Intensity, cps
1000 2000 3000 4000
4.2
5000
Concentration (ng/mL)
R2 = 0.99861
Area
0e0
1e5
2e5
3e5
4e5
0e0
1e4
2e4
3e4
4e4 4.135
Zeno MRMHR: y6, y7, y8, b3
MRMHR: y6, y7, y8, b3
5e5
6e5
Time (min) Time (min)
Intensity
Matrix blank
Triple quadrupole ZenoTOF 7600 system
1.8 2.0
1.961
2.1 2.2 2.3 2.4
0.0e0
2.0e5
4.0e5
6.0e5
8.0e5
1.0e6
1.2e6
1.4e6 1000
800
600
400
200
0
46 | SCIEX | CGT compendium SCIEX.com/messenger-RNA
Analytical solutions for IVT RNA
Suitable for:
High-quality separation
Rapid method development and sample analysis
BioPhase 8800
system
RNA 9000 Purity
& Integrity kit
IVT RNA Analytical solutions
BFS capillary
cartridge
Suitable for:
High-quality separation
Single nucleotide resolution for poly(A) 3’–ends
Large srRNA assessment
PA 800 Plus system RNA 9000 Purity
& Integrity kit
BFS capillary
cartridge
ssDNA 100-R kit
SCIEX.com/messenger-RNA
+ +
+ + +
47 | SCIEX | CGT compendium
Analytical solutions for IVT RNA
IVT RNA Analytical solutions
Suitable for:
5’cap and poly(A) 3’–end characterization
Robust, analytical flow setup
Molecule Profiler
software
Molecule Profiler
software
Suitable for:
5’cap and poly(A) tail characterization
Protein expression analysis
High flexibility to perform a range of additional workflows
SCIEX.com/messenger-RNA
ExionLC AE
system
ZenoTOF 7600
system
SCIEX OS
software
+ + +
ExionLC AE
system
X500B QTOF
system
SCIEX OS
software
+ + +
48 | SCIEX | CGT compendium
Oligonucleotide therapeutics
Oligonucleotide therapeutics Overview
SCIEX.com/oligonucleotides
Characterization and impurity ID
Confirm sequences, understand molecular
weights and achieve identification of
impurities.
Oligonucleotide MetID
Achieve a better understanding of your
ASOs by structural elucidation of their
metabolites.
Bioanalysis and DMPK
Overcome quantitation challenges of
oligonucleotides with highly sensitive and
robust solutions.
Shane Karnik
Sr. Laboratory Director, Aliri Bioanalysis
“You can theorize all you want, but until
you actually step into the lab and do the
work, you don’t know what the outcomes
are going to be.”
In 1998, the US Food and Drug Administration (FDA) approved
fomivirsen as the first oligonucleotide therapeutic. This
approval marked a revolution of action mechanism discovered
decades ago before finally coming to fruition. Since then,
the landscape of chemical modifications of oligonucleotides,
conjugations and formulations has evolved tremendously,
contributing to improvements in stability, efficacy and safety.
Today, more than a dozen ASO drugs, including siRNA and
aptamers are on the market, most of which are designated as
orphan drugs for treating rare genetic diseases. Alongside the
oligonucleotides, analytical solutions are required to keep up
with the ever-decreasing concentrations of analytes of more
potent drugs, pushing the boundaries of sensitivity.
49 | SCIEX | CGT compendium
Expert Q&A: Bioanalysis of oligonucleotide
therapeutics
The bioanalysis of oligonucleotides is particularly challenging, from sample preparation to method
optimization and data analysis for multiply charged analytes. Here, Shane Karnik and Dr. Troy Voelker, both
experts in the bioanalysis of oligonucleotides, share their experiences.
Dr. Troy Voelker is the Director of Laboratory Operations at Aliri Bioanalysis in Salt Lake City, Utah, leading the lab divisions
including method development, method validation, instrument operations, and production groups. He obtained his PhD in
synthetic organic chemistry and a post doctorate in medicinal drug development and drug metabolism. His bioanalytical
industry experience spans over 18 years, including triple quadrupoles and high-resolution accurate mass platforms for large
and small molecules. As Aliri’s subject matter expert for oligonucleotide bioanalysis, he has led method development and
validation for the past 10 years.
Shane Karnik serves as the Senior Laboratory Director at Aliri Bioanalysis in Colorado Springs, Colorado. His responsibilities
include leading a team of bioanalytical scientists supporting pharmaceutical drug development for non-GLP and GLP preclinical
and clinical studies. Shane earned a Master of Science in chemistry from the University of Colorado while working full-time
in R&D at a medical device company. He spent several more years in the pharmaceutical industry, developing and validating
analytical methods for new drug submissions.
Oligonucleotide therapeutics Expert Q&A
SCIEX.com/oligonucleotides
What type of column do you use for the bioanalysis of oligonucleotides to optimize peak shape,
retention and carryover effects?
Troy Voelker: This is a common question, and I hate to give a general answer to it: I do not have one column that
I go to every single time. I use the best column for the oligonucleotide that we are analyzing. Multiple vendors
have specialty columns for this type of work, and I recommend going through that suite of options. There is a
general consideration when looking for a column in terms of chemistry. You want a column that is going to be
stable above 65°C, especially when you are analyzing siRNA, since you must elevate the temperature to melt
the double strands. The high temperature can cause degradation over time for some column chemistries,
which you want to avoid picking. We look at columns from different vendors and decide based on peak shape
and carryover. You will have to do those experiments upfront to figure out which column is best for your
oligonucleotide.
What is your recommendation for oligonucleotide quantitation on the ZenoTOF 7600 system? Do you usually
use TOF-MS or MS/MS?
Troy Voelker: Both modes work for quantitation. Whether one is better suited for your project is something
you want to explore in your method development stage. There are 2 main factors to keep in mind: intensity
and signal to noise. Your MS2 data is not going to have the same intensity as your MS1 data. The main reason
for that is because you are breaking the precursor apart into multiple fragments and therefore spreading
the intensity across those fragments. On the other hand, you may achieve a better signal-to-noise ratio for
a fragment compared to the precursor. That’s often the case because of filtering out a lot of the background
when selecting a particular precursor for fragmentation. When working with samples that contain metabolites
that may start creating interferences by overlapping charge state envelopes, you may want to pick specific
fragments that would be unique to the metabolite and the parent, respectively. The fast acquisition rates of the
ZenoTOF 7600 system allow for collection of both full scan MS1 and multiple full scan MS2, while achieving the
desired amount of data points across the peak for accurate quantitation. Since you can get both sets of data,
you could still decide later which data set to use.
50 | SCIEX | CGT compendium
What internal standard are you using for LC-MS? Can you share general considerations for selecting internal
standards for oligonucleotide quantitation?
Shane Karnik: For the work I presented, a structural analog internal standard with 2 more bases than the
analyte oligonucleotide was used. Personally, I have never come across a project for which we got a stable
isotope labeled internal standard created. Usually, the internal standards were based on alternating bases,
additions or eliminations of bases, resulting in a different m/z than the analyte. As a general consideration, I
recommend picking an internal standard that very much acts the same way as your compound. You want a
standard that has the same extraction efficiency and the same ionization. Since you are going to look at peak
area ratios between your analyte and standard, changes in responses like enhancements or ion suppression
should be consistent between analyte and standard. We therefore do not use a universal standard. When we
work on a client project with several drug lead candidates, we can take another candidate and use that as the
internal standard.
Troy Voelker: I agree with Shane. A stable labeled internal standard is rare. The standard is almost always an
analog. To add onto Shane’s recommendations, if you are dealing with an oligonucleotide drug candidate with
special chemistry, you may want to keep that consistent for your analog selection. In addition, I recommend
using internal standards that are slightly higher in mass. With that potential, interferences with metabolites
can be avoided. Metabolites such as N-1, N-2 may cause interferences with an internal standard that is lower in
mass. We therefore do tend to go for a higher mass internal standard.you can get both sets of data, you could
still decide later which data set to use.
When you quantify metabolites, do you typically quantify against reference material or use the parent?
Shane Karnik: There are 2 answers to this question. When doing regulated GLP work, you must have a wellcharacterized reference material for that metabolite. However, for early discovery work, when we are just looking
for metabolites and want to get a general idea of the concentration of metabolites, we do not need a reference
material. We can use the TOF-MS data, look for expected metabolites and extract those m/z from the data.
For quantitation, we then assume a similar response as the parent or the internal standard and use the parent
quantitation curve for the metabolites. This would be only for non-regulated discovery work.
Troy Voelker: I can add that there are likely going to be differences in the ionization between the parent and the
metabolite. It is something that we generally observe. As Shane stated, the approach without reference material
gives you an idea of the concentrations, but you are certainly not going to get an absolute concentration.
Does the ZenoTOF 7600 system allow for characterization of the oligonucleotides by analyzing the
fragments? Is there oligonucleotide sequencing software available to process the data?
Shane Karnik: Yes. You can use the MS/MS data from the ZenoTOF 7600 system and process it with Molecule
Profiler software to achieve fragment matching and confirm sequences of synthetic oligonucleotides. If you are
trying to identify N-1, N-2, etc. metabolites, you can also use that approach to figure out from which end you are
losing nucleotides to form the metabolites. We do that kind of work for reference material. Sometimes we only
get UV spectra for these materials, which is not enough detail when we start development. The software helps
us with that.
Oligonucleotide therapeutics Expert Q&A
SCIEX.com/oligonucleotides
Read the entire Q&A blog >
51 | SCIEX | CGT compendium
ASOs are approximately 18–28 nt in length and are either single stranded or exist as duplexes,
such as for siRNA. Chemical modifications are common to increase target binding and stability,
while conjugations to N-acetylgalactosamine (GalNAc) or lipids are used for delivery and to increase
specificity. Shortmers—impurities derived from the stepwise synthesis of ASOs—increase the
complexity of synthetic oligonucleotide samples. The characterization of ASOs therefore requires a
reliable and high-resolution solution.
Trust your results with excellent raw data quality based on exceptional negative ionization
efficiency and declustering of adducts
Confidently confirm the sequence of your oligonucleotides and obtain relative quantitative
information of full-length product (FLP) and impurities
Characterization and impurity identification
Confirm integrity and determine purity with excellent resolution and reproducibility
Figure 44. Data
comparison obtained from
modified ASO
(18 nt) without the Zeno
trap (left) and with the
Zeno trap (right). MS/MS
spectra from precursor
m/z = 711.62 (top). XIC
from MS/MS fragment with
m/z = 393.05 (bottom).
Figure 43. Data from an
18 nt phosphorothioated
ASO with 2’-O-methoxyethyl
(MOE) modifications. TOF-MS
data shows different charge
states with inset showing
reconstructed data and
calculated mass error.
Discover more details in the technical note about oligonucleotide characterization >
Oligonucleotide therapeutics Characterization and impurity ID
Intensity, cps Intensity, cps
0e0
1e5
2e5
3e5
Time,min
0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5
Time,min
0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5
0e0
2e4
4e4
6e4
8e4
Mass/Charge, Da
400
393.0542 418.0594 654.4602 711.5232 759.1919
393.05626
402.0633
392.0648
418.0588
401.5663
576.6246
655.1270
654.4605
711.6216
759.1905
786.1256
837.1219
600 800
ASO (18 nt)
Zeno trap off Zeno trap on
Mass/Charge, Da
400 600 800
Mass / charge
Intensity
Mass (Da)
Intensity
3.0e4
2.0e4
1.0e4 0.0e0
3.0e4
6.0e4
9.0e4
1.2e5
0.0e0
3.5e4
2.5e4
1.5e4
5.0e3
700 800 900 1000 1100 1200
7125
7122.266
Mass error -1.4 ppm
7124.268
7123.264
7126.278
7128.274
7129.276
7130.276
7131.271
7132.271
7133.2770
7134.270
7130 7135
1300 1400 1500 1600 1700
52 | SCIEX | CGT compendium SCIEX.com/oligonucleotides
Modifications to the Phospho-backbone and ribose sugars, which are often introduced to increase
the stability of ASOs, also impact the metabolism profile. Depending on stabilizing modifications,
endonucleases or exonucleases can hydrolyze ASOs within the sequence or result in shortmers
from 5’ and/or 3’ ends. To determine the impact of metabolites on efficacy, toxicity and drug-drug
interactions, structural identification is of high relevance.
Achieve excellent raw data quality through exceptional negative ionization efficiency and
declustering of adducts
Confidently confirm oligonucleotide sequences of FLP and metabolites
Break through boundaries of duty cycles and obtain superior MS/MS data quality for low
abundance metabolites with the Zeno trap
Oligonucleotide MetID
Identify and quantify metabolites reliably
Figure 46. Matching MS/
MS fragments of an
18-mer ASO. Suggested
sequence matching of
detected MS/MS fragments
and associated sequence
with error calculation from
Molecule Profiler software.
Figure 45. Chromatograms
of a 20-mer ASO and
spiked in metabolites.
Representative XICs
for the FLP with
2’-O-methoxyethyl and
5-methyl modifications
and related metabolites
(shortmers of 5’ and 3’-
ends) are shown.
Discover more details in the technical note about metabolite identification >
Oligonucleotide therapeutics MetID
Time (min)
0.0e0
1.0e4
2.0e4
3.0e4
4.0e4
5.0e4
6.0e4
7.0e4
8.0e4
9.0e4
1.0e5
1.1e5
1.2e5
1.3e5
1.4e5
1.5e5
1.6e5
1.7e5
4.6 4.8 5.0 5.2 5.4 5.6 5.8 6.0 6.2 6.4 6.6 6.8 3’(n-12) 3’(n-11) 5’(n-10)5’(n-9) 5’(n-6) 5’(n-7) 5’(n-2) 5’(n-1) 5’(n-3)
FLP
(20-mer)
Intensity, cps
53 | SCIEX | CGT compendium SCIEX.com/oligonucleotides
Improved stability and specificity are factors contributing to an increase in potency
of oligonucleotide therapeutics. Quantitative studies to support drug metabolism and
pharmacokinetics (DMPK) require analytical assays with high sensitivity to detect the everdecreasing concentrations of the active pharmaceutical ingredient (API) and its metabolites.
Additionally, ion pairing agents, frequently used to enable chromatographic separation, result in a
need for high robustness.
Break through quantitation boundaries with high signal-to-noise and best-in-class
robustness
Achieve excellent negative ionization efficiency and declustering of adducts with state-ofthe-art source design
Bioanalysis and DMPK
Don’t choose between robustness and high sensitivity
Figure 48. Comparison of
XICs from a 20-mer ASO
at different concentrations
in extracted plasma. Top:
Data from the SCIEX Triple
Quad 6500+ system with
LLOQ of 1 ng/mL. Bottom:
Data obtained from the
SCIEX 7500 system with
LLOQ of 0.2 ng/mL.
Figure 47. Quantitative
performance of a 21-mer
antisense strand from
an siRNA-lipid conjugate
in development using
a SCIEX 7500 system.
Calibration curve based on
XIC adjusted using internal
standard shows LDR of 2.3
(n = 3). Inset shows XIC of
transition for blank sample
and LLOQ at 0.05 ng/mL.
Discover more details in the technical note about the quantitation of siRNA conjugates >
Oligonucleotide therapeutics Bioanalysis and DMPK Area ratio
Concentration (ng/mL)
Intensity
Time (min)
Blank
R2 = 0.98997
LDR = 2.3
0.05ng/mL
0
1000
2000
3000
4000 2.028
1.9 2.0 2.1 2.2
0
1000
0.0
0.5
1.0
1.5
2.0
2.5
2000
3000
4000
1.9
1 2 3 4 5 6 7 8 9
2.0 2.1 2.2
0.2 ng/mL 0.5 ng/mL 1 ng/mL
LOD LLOQ
LLOQ
SCIEX Triple Quad
6500+ system SCIEX 7500 system
Intensity Intensity
Time (min) Time (min) Time (min)
2000
1500
1000
500
0
2.6
3.163
2.899 2.907 2.917
3.157 3.172
2.8 3.0 3.2
3000
2000
1000
0
2.6 2.8 3.0 3.2
2000
1500
1000
500
0
2.5 3.0 3.5
3000
2000
1000
0
2.5 3.0 3.5
800
600
400
200
0
2.5 3.0 3.5
800
600
400
200
0
2.6 2.8 3.0 3.2
54 | SCIEX | CGT compendium SCIEX.com/oligonucleotides
Analytical solutions for
oligonucleotide therapeutics
Suitable for:
Molecular weight determination and sequence confirmation
Robust, analytical flow setup
Suitable for:
Molecular weight determination and sequence confirmation
High-resolution bioanalysis and MetID studies
Flexible LC setup
Oligonucleotide therapeutics Analytical solutions
SCIEX.com/oligonucleotides
Molecule Profiler
software
Molecule Profiler
software
ExionLC AE
system
ZenoTOF 7600
system
SCIEX OS
software
+
+
+
+
+
+
ExionLC AE
system
X500B QTOF
system
SCIEX OS
software
55 | SCIEX | CGT compendium
SCIEX 7500+
system
SCIEX Triple Quad
6500+ system
Analytical solutions for
oligonucleotide therapeutics
Suitable for:
Targeted bioanalysis studies
Best-in-class quantitative performance with superior robustness
ExionLC AE
system
Suitable for:
Targeted bioanalysis studies
Great quantitative performances
Oligonucleotide therapeutics Analytical solutions
SCIEX.com/oligonucleotides
ExionLC AE
system
SCIEX OS
software
+ +
SCIEX OS
software
+ +
56 | SCIEX | CGT compendium
Tips and tricks from our application experts:
LC-MS setup part I
DilipKumar Reddy Kandula (PhD), Staff Applications Scientist at SCIEX, US, shares his tips and
tricks on how to prepare your system for oligonucleotide LC-MS analysis.
Dr. Dilip Reddy has 10 years of experience as a research scientist in different pharma and biopharma companies,
where he focused on using MS for the analysis of biologics and small molecules. His work included the characterization
of complex molecules, bioanalysis and MetID studies with triple quad and high-resolution LC-MS instrumentation.
For the past 7 years, he applied his extensive knowledge within various functions at SCIEX. Dilip holds a PhD in protein
characterization using mass spectrometry from the Shri JJT University in Rajasthan, India.
The background on formation of metal adducts
It is very common for oligonucleotides to form adducts with alkali metal ions during LC-MS analysis. Adduct
formation occurs due to the electrostatic attraction between the negatively charged phosphate backbone
of the oligonucleotide and the positively charged alkali metal ions. The formation of alkali metal adducts
can negatively impact the data quality. Signal intensities of analytes are reduced due to spreading the
signal across different adducts. Additionally, identification and accurate quantitation of species is further
complicated. I summarized my tips below to help improve your oligonucleotide LC-MS analysis.
Tip 1: Choose the right consumables
Glass bottles tend to leach sodium ions, hence plastic bottles are recommended as containers for mobile
phases. Before the first use, it is recommended to soak the plastic bottles overnight in isopropanol (IPA)
containing 10% acetic acid and rinse them 10–15 times with milliQ water. Using high-quality LC-MS grade
additives (acetic acid, ion pairing agents) and solvents can help minimize adduct formation. Ensure that
a set of tubing and columns is set aside for oligonucleotide analysis usage only, such as the Phenomenex
BioZen Oligo LC column.
Tip 2: Prepare your LC system
Before starting analysis, ensure your LC system is prepared for oligonucleotide analysis with ion pairing
reversed phase liquid chromatography (IP-RP-LC). My recommendation is to flush the solvent and
autosampler lines with LC-MS grade IPA for 10 to 15 minutes. Include your tubing and electrode for all
flushing steps but use a connector piece instead of your column. Then, switch to 10% acetic acid for 1-2
hours. Doing “dummy” injections of a 10 % acetic acid solution is beneficial for cleaning the injection parts.
As a next step, switch to LC-MS grade or MilliQ water for 1-2 hours to remove the acetic acid. Then install
your oligonucleotide column and equilibrate system with mobile phases for analysis.
Tip 3: Determine an MS cleaning schedule
It is important to clean the ion source regularly to prevent contamination, reduce the adduct formation
and maintain optimal sensitivity. Follow the SCIEX guidelines for cleaning procedures and recommended
cleaning solutions and keep a specific cleaning schedule. A regular cleaning schedule of your source and
MS front-end is recommended to help preventing any contamination travelling further into the system,
especially when working with ion pairing agents for IP-RP-LC analysis.
More questions? Contact us >
Oligonucleotide therapeutics Tips and tricks
57 | SCIEX | CGT compendium SCIEX.com/oligonucleotides
Tips and tricks from our application experts:
LC-MS setup part II
Kaoru Karasawa, Professional Specialist, Application Support at SCIEX, Japan, shares her tips and
tricks on oligonucleotides analysis with LC-MS
Kaoru Karasawa has more than 25 years of mass spectrometry experience, primarily in high-resolution MS. She
leverages from extensive hands-on experience in impurity analysis, MetID and quantitation of small molecules and
oligonucleotides for pharmaceutical and omics applications. Prior to joining SCIEX, Kaoru worked as a natural product
chemist for the Roche Group, Japan. Currently, she is participating in a project for oligonucleotide development in
Japan and contributing to the development of analytical techniques. Kaoru holds a Master’s Degree in engineering.
Tip 1: Sample handling
Oligonucleotides tend to bind to pipette tips and vials. In addition, oligonucleotides can be sensitive to
the degradation by nucleases. I therefore recommend using low-binding LC vials and pipette tips and
recommend wearing gloves and using nuclease-free water for sample preparation to minimize the risk
of sample loss due to adsorption and/or degradation. Furthermore, the use of an internal standard is
beneficial. It can help with minimizing the adsorption of the target analyte and can increase quantitative
accuracy for quantitative studies.
Tip 2: Replace LC solvents
To facilitate chromatographic separation while enabling MS sensitivity, alkylamines and hexafluoro-2-
propanol (HFIP) are commonly used for IP-RP-LC-MS analysis. However, the additives in the solvents
can evaporate quickly. The change in concentration and pH can lead to changes in chromatographic
performance and decreased MS signal intensities. I recommend checking in advance how long your solvent
can be used and design experiments accordingly. If you notice a decrease in expected MS peak intensity,
prepare fresh solvents.
Tip 3: Select suitable columns
In many cases, the separation of analytes from impurities or metabolites requires high column
temperatures, and in some cases, can go up to 90°C. The analysis of double stranded analytes, such as
siRNA, require high temperatures to separate the sense from the complementary antisense strand. In
addition, the solvents used for IP-RP-LC are high in pH. I recommend carefully choosing a column that can
withstand these conditions, such as the Phenomenex BioZen Oligo LC column.
Tip 4: Optimize MS methods
The MS signal for therapeutic oligonucleotides is usually distributed across multiple charge states—mainly
-2 to -10—and the distribution varies based on LC and MS conditions, sequence composition and length.
For quantitative analyses, I recommend optimizing the collision energy for several charge states, since
the most intense charge state does not necessarily provide the most intense fragment ion with high
specificity. For qualitative analyses, such as sequencing, it is beneficial to combine the MS/MS information
from different charge states for best results.
More questions? Contact us >
Oligonucleotide therapeutics Tips and tricks
58 | SCIEX | CGT compendium SCIEX.com/oligonucleotides
Gene editing
Gene editing Overview
SCIEX.com/CRISPR-Cas9
sgRNA and Cas9 mRNA purity
Separate impurities and
determine the integrity and
purity of sgRNA and Cas9
mRNA in one experiment.
pegRNA purity
Move past secondary structures
and assess the purity of guide
RNAs despite high sequence
complementary.
Intact mass characterization
Achieve accurate intact
molecular weight information
of guide RNAs and related
impurities.
Cas9 protein characterization
Confirm protein sequences and
understand PTMs and amino acid
isomers in depth.
Proteome profiling
Understand gene editing
effects better with an unbiased
proteome-wide approach.
Hugo Gagnon (PhD)
Chief Scientific Officer, Allumiqs
“What’s the outcome in the cell? Is it
on target? Is it off target? What are
those undesired effects? Is there some
percentage of misintegration? Answering
these questions is what we focus on with our ZenoTOF 7600
system.”
Gene editing approaches, such as CRISPR/Cas9, have
tremendous potential to go beyond the treatment of genetic
diseases: A full cure for the majority of known human genetic
diseases is theoretically possible by leveraging gene editing.
Monogenetic diseases—diseases caused by mutations in one
gene—are the logical starting point. However, research on
how to target multiple gene locations using one treatment
with gene editing stacking is still ongoing. In addition, CRISPR
approaches bear the potential to target other diseases,
such as cancer. Challenges around delivery to target cells,
manufacturing costs and analytical assessments to ensure
the safety of these new approaches, need to be overcome.
59 | SCIEX | CGT compendium
Expert Q&A: Overcoming challenges to
assess pegRNA purity
Prime editing using prime editing guide RNAs (pegRNAs) is one of the latest approaches for gene editing.
Here, Ashley Jacobi (Integrated DNA Technologies, IDT) and Tingting Li (SCIEX) share their experiences on
overcoming analytical challenges linked to pegRNAs.
Ashley Jacobi is the Director of Application and Market Development at IDT. She received her Molecular Genetics & Biochemistry
degree from Cornell College. Ashley has spent the last 18 years holding several roles in research and product development
at IDT, focusing on RNAi and antisense oligo technologies, and CRISPR gene editing. Her work on CRISPR includes optimizing
the composition and delivery of synthetic RNAs complexed to recombinant CRISPR nucleases and understanding the genetic
outcomes following CRISPR gene editing.
Tingting Li is the Manager of Cell and Gene Therapy at SCIEX. Tingting has more than 10 years of experience in CE holding
different roles in Shanghai, China and Brea, US, focusing on scientific and specialist product support. Her focus includes new
workflow development and characterization of cell and gene therapies. Prior to SCIEX, Tingting served as an analytical scientist
and project leader in the pharma industry for analytical method development and method validation for several years. Tingting
holds a Master’s degree in Chemistry from Rutgers University New Brunswick.
Gene editing Expert Q&A
SCIEX.com/CRISPR-Cas9
Can you comment on the typical lifespan of the capillaries used for pegRNA analysis?
Tingting Li: The capillary used for this assay is the coated DNA capillary for the PA 800 Plus system available
in the ssDNA 100-R kit (PN 477480). It is validated for 100 runs. However, my personal experience is that more
runs can be performed. For the pegRNA analyses, I conducted >150 runs and the capillary can still be used
for further analyses. I used the same capillary to develop the method presented in the webinar and to analyze
the pegRNAs of different lengths, from different lots on different instruments and on different days. The key
to achieving a long capillary lifetime is to avoid exposing the capillary ends to air for more than 3 minutes.
Additionally, it is important to store the capillary appropriately after usage. When not in use, always rinse and
store the capillary in unused gel buffer at 2°C to 8°C with both capillary ends submerged in Tris-Borate-Urea
buffer.
Can you provide further insights into your strategy for addressing the challenge of pegRNA peak shape?
Tingting Li: Optimizing the peak shape of pegRNA was indeed a challenging task. Our hypothesis is that highly
complementary primer binding sites (PBS) and protospacer sequences in pegRNA molecules caused the
formation of hairpin and secondary structures, resulting in a very broad peak when analyzed with CE under
standard conditions. To confirm this, we synthesized 4 pegRNAs with varying lengths and their corresponding
non-complementary RNAs (NC RNAs) with the same PBS sequence but with a non-targeting protospacer.
We analyzed all 8 samples and found that the NC RNAs exhibited a singular sharp peak while we observed
broad, unresolved peaks for all pegRNAs. This further indicated that the broad, undesired peak shape observed
for pegRNAs was likely due to the formation of high-order structures caused by their highly complementary
sequence design. For the next step of method development, we focused on a robust denaturing technique
for CGE purity analysis of pegRNAs with high levels of secondary structure. We explored various denaturation
methods during the sample preparation. However, we did not achieve significant improvements of the peak
shape using these different sample preparation procedures. We then focused on optimizing the conditions
during the separation process. The breakthrough came with heat-based denaturation during separation in the
CE capillary. We maintained a capillary temperature of 50°C throughout the separation process. Based on the
peak profiles achieved, we concluded that this step prevented the formation of inter- and/or intramolecular
hydrogen bonds of pegRNA molecules. With the temperature-controlled capillary at 50°C, we can achieve a
single sharp peak for each of the pegRNA samples.
60 | SCIEX | CGT compendium
Read the entire Q&A blog >
Gene editing Expert Q&A
SCIEX.com/CRISPR-Cas9
Could you please elaborate on the temperature control feature of your CE system and its role in improving
the accuracy of pegRNA purity analysis?
Tingting Li: Certainly. The unique capillary temperature control feature of the PA 800 Plus system is a critical
aspect that ensures precise and stable temperature control during the separation process. This is achieved
by an inert liquid circulating through the cartridge around the separation capillary. Compared to air used
in other systems, the inert liquid can maintain temperature more precisely and consistently throughout
the analysis. For the analysis of oligonucleotides like pegRNA, the stable temperature control is vital as it
minimizes the variations in migration time and peak shape, resulting in highly reproducible results for purity
analyses.
How can the denaturing technique and gel matrix be adapted for other analyses with high levels of
secondary structures? Can you provide any advice?
Tingting Li: The denaturing technique presented in the webinar and the gel matrix can be adapted for the
analysis of other oligonucleotides with high levels of secondary structures. I recommend testing different
denaturation agents and temperatures for sample preparation and during the CE separation process
while considering the characteristics of your oligonucleotides, such as heat and pH stability for example.
In addition, the gel matrix, separation voltage and separation time can be optimized to achieve the needed
resolution and peak shape. I believe the flexibility of this method and system can open possibilities for
investigating various oligonucleotides and their functions in different research areas.
How do you foresee the CGE purity workflow impacting future research and advances in CRISPR
technologies?
Ashley Jacobi: High quality synthetic guide RNAs with high purity is very important for the efficiency of
your gene editing experiments. This holds true not only for prime editing but for any CRISPR technology. As
these technologies move from research towards therapeutic use, the purity of the starting material is of the
utmost importance. Having analytical technology that provides accurate purity determination is a necessity
to achieve desired outcomes and bring safe gene editing technologies to market.
Can you comment on modalities beyond pegRNAs in your pipeline?
Ashley Jacobi: We are always looking for improvement and innovation within IDT. The data shown in the
webinar was up to about 200 nucleotides. However, we are investigating even longer guide RNAs. Another
avenue we are exploring is different chemical modifications. How do those modifications affect editing
efficiency and/or the stability of these compounds? It is something we are actively looking into.
61 | SCIEX | CGT compendium
Time (min)
Impurities
Intact mRNA
Relative fluorescence Units (RFU)
12 13 14 15 16
0
1
2
3
For therapeutic purposes, the naturally occurring CRISPR RNA (crRNA) and the trans-activating
CRISPR RNA (tracrRNA) are commonly engineered into a single-guide RNA (sgRNA). While sgRNAs
consist of approximately 100 nt, Cas9-encoding mRNAs are >4000 nt in size. Confirming the
integrity and assessing the purity quantitively for both RNAs—sgRNA and Cas9 mRNA—are key for
successful gene editing.
Assess the quality of product with various sizes in one analysis without compromising on data
quality
Understand related nucleic acid impurities and determine sizes and quantities,
simultaneously
Cover your compliance needs through compatibility with common data management systems
sgRNA and Cas9 mRNA integrity and purity
Break through limitations based on secondary structure formations
Figure 50. Zoomed-in
electropherogram of Cas9
mRNA. The resolution of
the CGE method allows for
separation of impurities
from the main product and
quantitation of Cas9 mRNA
purity.
Figure 49. Overlay of
electropherograms from
a mixture of sgRNA and
Cas9 mRNA and an RNA
ladder using the BioPhase
8800 system. The mRNA
of ~4500 nt and the sgRNA
of 100 nt can be assessed
within the same analysis
(green trace). The RNA
ladder is shown in blue.
Discover more details in the technical note about sgRNA and CRISPR Cas9 mRNA analysis >
Gene editing sgRNA and mRNA purity
Time (min)
RNA size standard
50
sgRNA
(100nt)
Cas9 RNA
(-4500 nt)
150
500
1000
2000
3000
5000
7000
sgRNA and Cas9 mRNA
Relative fluorescence Units (RFU)
/.5 10 15
0
5
10
20
15
25
30
62 | SCIEX | CGT compendium SCIEX.com/CRISPR-Cas9
PE is a promising approach with increased specificity and efficiency. It consists of an sgRNA with
a reverse transcriptase template sequence and a primer binding site. With a length of ~120-250
nt, synthetic pegRNAs are prone to impurities derived from their stepwise synthesis. In addition,
their complementary bases can lead to secondary structure formation that is resistant to common
denaturation strategies, posing an additional analytical challenge to overcome.
Achieve superior resolution and repeatability for purity assessments of intermediate
products
Break through the boundaries of secondary structure with liquid-based temperature control
Streamline data management and cover your compliance needs through compatibility with
common data management systems
pegRNA purity for prime editing
Break through limitations based on secondary structure formations
Figure 52. CGE analysis
of pegRNAs with different
lengths using the
optimized method on a PA
800 Plus system. pegRNAs
from 179 nt to 229 nt were
tested with the optimized
protocol, all resulting in
sharp peaks.
Figure 51. CGE analysis
of pegRNA (blue trace)
and non-complementary
RNA standard (STD, green
trace) with similar length.
Left: Before method
optimization, the pegRNA
(blue trace) shows
extensive tailing compared
to the non-complementary
STD (green trace). Right:
After method optimization,
the pegRNA sample shows
a single sharp peak with
comparable peak width as
the non-complementary
STD.
Discover more details in the technical note about pegRNA purity analysis >
Gene editing pegRNA purity
Time (min)
Relative fluorescence Units (RFU)
pegRNA 179
pegRNA 189
pegRNA 199
pegRNA 209
pegRNA 219
pegRNA 229
5 10 15 20
0
30
60
90
120
10
0
20
40
60
80
100
0
20
40
60
80
100
15 20 25 8 12 16
Time (min)
RNA STD (128 nt)
pegRNA (129 nt)
RNA STD (128 nt)
pegRNA (129 nt)
100
10
80
60
40
20
0
100
80
60
40
20
0
15 20 25 8 12 16
Pre-optimization Post-optimization
SCIEX.com/CRISPR-Cas9
Relative fluorescence units (RFU)
Relative fluorescence units (RFU)
63 | SCIEX | CGT compendium
Mass/charge
Mass (Da)
Guide RNAs can vary in size from ~17 nt and ~65 nt in length for crRNA and tracrRNA respectively,
to ~100 nt for sgRNA and up to 250 nt for newer approaches, such as pegRNA. While the specificity
and efficiency can be increased with the latest sgRNAs, the likelihood of introducing impurities
increases during the stepwise synthesis of larger RNAs. The quality of sgRNAs is crucial for
achieving high gene editing efficiencies. An error in 1 nucleotide can result in significantly reduced
efficiency.
Leverage excellent raw data quality through exceptional negative ionization efficiency and
decluster adducts with state-of-the-art source design
Confidently confirm identities of your guide RNAs based on deconvoluted molecular weights
with great mass accuracy
Uncover relevant information on impurities and obtain relative quantitative information
Intact mass characterization of guide RNAs
Assess mass information of guide RNAs and impurities with ease
Figure 54. TOF-MS data
from pegRNA sample with
167 nt. Blue trace shows
TOF-MS raw data and
deconvoluted mass (inset)
of desired product. Pink
trace shows TOF-MS raw
data and deconvoluted
mass (inset) of unexpected
impurity with higher mass.
Figure 53. TOF-MS data
from pegRNA with 122 nt.
Raw TOF-MS data shows
charge state envelope
of pegRNA. Inset shows
deconvoluted mass with
excellent mass accuracy.
Additionally, lower intensity
peaks could be attributed
to desulfurization and
metal adduct formations.
Discover more details in the technical note about large oligonucleotide LC-MS analysis >
Gene editing Intact mass characterization
Mass/charge
SCIEX.com/CRISPR-Cas9
% Intensity % Intensity
64 | SCIEX | CGT compendium
To increase target specificity further, Cas9 fusion proteins are being studied. A deeper look into
these engineered proteins is required to confirm the target amino acid sequence and identify lowabundance
PTMs to ensure product quality. While a peptide-mapping approach can provide relevant
information,
it can be challenging to achieve high enough sequence coverage and full elucidation of PTMs to
characterize PQAs and CQAs.
Obtain high protein sequence and fragment coverage with limited sample amounts through
highly sensitive data acquisition
Differentiate amino acid isomers and determine relative quantities and exact locations of
PTMs including highly fragile PTMs with EAD
Take back your time with accurate and streamlined data processing
Cas9 protein characterization
Know your sequence and fully understand challenging PTMs
Figure 55. Fragment coverage comparison for long peptide from the Cas9 protein. EAD shows extensive fragmentation compared to CID,
providing higher confidence in the identity of the peptide.
Discover more details in the technical note about Cas9 protein analysis with EAD >
Gene editing Cas9 protein characterization
Mass/ charge
CID fragment coverage: 8.5%
Intensity
500
y1
y1 - H20
y2
b2 b3
y27
y28
1000 1500 2000 2500 3000
800
600
400
200
0
Y K V P S K K F K V L G N T D R H S I K K N L I G A L L F D S G E T A E
Mass/ charge
isoAsp
V8, c8 V8, c8
d9
z8 + 1
z8 + 1
z8 + 1 - 57 z8 + 1 - 44
Time (min)
Native
Asp
Asp
isoAsp
Mass/ charge
Intensity
Intensity
Intensity
790 800 810 820 830
4 5 6 7 8 9 10
840 850 860 870 810 815 820 825 830 835 840 845 850 855
0
100
200
300
400
0
0.5
1.0
1.5
2.0
2.5
3.0
0
500
1000
1500
2000
V L G N T D RH S I K V L G N T D RH S I K
Mass/ charge
EAD fragment coverage: 48.6%
Intensity
500
y1
Z3
Z4
C4
C5
C6
C7
a8
C8
C9
C10
C12
C13
C14
C15
C16
C17
Z20
C33
C34
C35
Z21 + 1
Z22 + 1 Z26 + 1
Z27 + 1Z28 + 1
Z30 + 1
Z31 + 1
Z2
C2
C1
1000 1500 2000 2500 3000 3500
800
600
400
200
0
Y K V P S K K F K V L G N T D R H S I K K N L I G A L L F D S G E T A E
Figure 56. Differentiation
of Asp and isoAsp by EAD.
Top: XICs of the native and
deamidated versions of the
peptide VLGNTDRHSIK from
the Cas9 protein. Bottom:
EAD MS/MS spectra
providing differentiation
between Asp and isoAsp.
65 | SCIEX | CGT compendium SCIEX.com/CRISPR-Cas9
In addition to specificity challenges resulting in off-target effects, gene editing can affect the
phenotype in various ways based on the complexity and interdependency of protein networks.
Genomic readouts cannot provide sufficient insights into the potential disruption of gene
regulators or detect changes to the proteome. Protein assays, such as western blots, on the other
hand, are limited by antibody availability and cannot detect unexpected proteome-wide changes.
Understand the effects of gene editing on the proteome level in an unbiased way with Zeno
SWATH DIA
Dig deeper into changes despite limited sample amounts with increased sensitivity using the
Zeno trap
Achieve confident identification and simultaneous quantitation with excellent MS/MS data
quality
Proteome profiling for gene editing
Dig deeper into the proteome and understand changes with amino acid resolution
Discover more details in the technical note about SWATH DIA for biomarker ID and quantitation >
Gene editing Proteome profiling
Figure 58. Identified and
quantified proteins using
SWATH DIA. Columns
show number of proteins
identified with a false
discovery rate (FDR)<1% for
different loading amounts
of cell lysate digest
using a 60 min gradient.
Line shows amount of
proteins with %CV<20%
used for quantitation as
percentage of total amount
of identified proteins with
FDR<1%.
Data courtesy of Allumiqs, Halifax, NS, Canada
# of proteins <1% FDR
Load (ng)
60-min gradient
Proteins with %CV<20 All identified proteins
Figure 57. Insertiondeletion (INDEL) errors
induced by gene editing
detected by bottom-up
proteomics using SWATH
DIA. The expectation is that
all peptides in the geneedited samples (blue) are
below the baseline signal.
However, compared to the
control (orange), only two
peptides at the N-terminus
of the protein were silenced
based on a shift in the
initiation of the protein
transcription. Bar thickness
represents standard
deviation of the peptides.
66 | SCIEX | CGT compendium SCIEX.com/CRISPR-Cas9
Analytical solutions for CRISPR/Cas9
Gene editing Analytical solutions
Suitable for:
High-quality separation of sgRNA and Cas9 mRNA
Rapid method development and sample analysis
+
Suitable for:
High-quality separation of sgRNA and Cas9 mRNA
High-resolution pegRNA analysis
Smaller sample sets
SCIEX.com/CRISPR-Cas9
BioPhase 8800
system
RNA 9000 Purity
& Integrity kit
BFS capillary
cartridge
PA 800 Plus system RNA 9000 Purity
& Integrity kit
BFS capillary
cartridge
ssDNA 100-R kit
+ +
+ + +
67 | SCIEX | CGT compendium
Analytical solutions for CRISPR/Cas9
Gene editing Analytical solutions
Suitable for:
Molecular weight determination of gRNAs, sgRNAs and
pegRNAs
Cas9 protein characterization
Robust analytical flow setup
Suitable for:
Molecular weight determination of gRNAs, sgRNAs and pegRNAs
Cas9 protein characterization including isomer differentiation
High flexibility to perform a range of additional workflows
SCIEX.com/CRISPR-Cas9
ExionLC AE
system
ZenoTOF 7600
system
SCIEX OS
software
ExionLC AE
system
SCIEX OS
software
Biologics Explorer
software
Biologics Explorer
software
+ + +
X500B QTOF
system
68 | SCIEX | CGT compendium
Concluding remarks
Concluding remarks
Dr. Joe Fox
President at SCIEX
“We are witnessing unprecedented advancements in genetic medicines,
capable of treating once-incurable diseases. This progress is closely linked
to the continuous evolution of analytical technologies. At SCIEX, we are
devoted to precise science that enables our customers to solve their most
impactful challenges through ground-breaking innovations and outstanding
reliability and support.”
Emmanuel Abate
President at Cytiva
“Advancing the next generation of therapeutics isn’t based on scientific
advancement alone. Future biomanufacturing will increase in its complexity,
as new therapeutic modalities emerge and the demand for personalized
medicine grows. Collaboration between industry, academia and government
bodies must work together to create accelerated, efficient pathways for drug
developers.”
Jennifer Meade
President at Aldevron
“At Aldevron, our vision—a world where every cure is possible—reflects our
25 years of experience and our commitment to the future of cell and gene
therapy. We are driven by a relentless pursuit to better serve therapeutic
developers working on life-altering treatments for patient populations. To
make this a reality, we will continue to see improvements in the speed, cost
and safety profile of new therapeutics, enabled by platform processes and
disruptive new technological advancements.”
Dr. Kaveh Kahen
President at Phenomenex
“Analytical chromatographic solutions are vital for generating the data
needed to make crucial therapeutic drug decisions. With the rapid
evolution of science and the emergence of personalized medicines, drug
discovery researchers rely on high quality chromatographic consumables
to accelerate the development timeline for new therapeutics. Phenomenex
provides a diverse range of solutions designed specifically for this purpose.
Our offerings include sample preparation, column chemistry, and round-theclock technical support, all of which instill confidence in the data required to
bring life-saving and life--enhancing therapies to market faster.”
69 | SCIEX | CGT compendium SCIEX.com/biopharma-and-pharma-research
The SCIEX clinical diagnostic portfolio is For In Vitro Diagnostic Use. Rx Only. Product(s) not available in all countries. For
information on availability, please contact your local sales representative or refer to www.sciex.com/diagnostics. All other
products are For Research Use Only. Not for use in Diagnostic Procedures. Trademarks and/or registered trademarks
mentioned herein, including associated logos, are the property of AB Sciex Pte. Ltd. or their respective owners in the United
States and/or certain other countries (see www.sciex.com/trademarks). AB Sciex™ is being used under license. Catalent®
is a trademark of Catalent® Pharma Solutions. GMP-Source®, Nanoplasmid® and REVIVER® are trademarks of Aldevron.
Biozen™ Oligo LC column is a trademark of Phenomenex.
MKT-31655-A © 20234DH Tech. Dev. Pte. Ltd. MKT-28611-A
Headquarters
500 Old Connecticut Path
Framingham, MA 01701
USA
Phone 508-383-7700
sciex.com
International Sales
For our office locations
please call the division
headquarters or refer to our
website at
sciex.com/offices
Brought to you by
Download the Compendium for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version