Past, Present and Future of Digital PCR
eBook
Published: October 22, 2025
Credit: Bio-Rad
Modern therapeutics and diagnostics demand unprecedented analytical precision to ensure effectiveness and safety. Yet, traditional qPCR methods introduce variability, require standard curves and struggle with low-abundance targets. These limitations create bottlenecks in vaccine development, quality control and biomarker discovery.
This eBook explores how Droplet Digital PCR (ddPCR) technology delivers absolute quantification, superior sensitivity and inhibitor tolerance to overcome these challenges across oncology, cell and gene therapy and vaccine development.
Download the eBook to learn how ddPCR can:
- Achieve absolute quantification without standard curves for reliable therapeutic development
- Detect rare ctDNA variants for personalized cancer monitoring
- Eliminate analytical bottlenecks in vaccine development and manufacturing
Past, Present and Future
of Digital PCR
ONCOLOGY
VACCINES
CELL
& GENE
THERAPY
eBOOK
Visit bio-rad.com/dropletdigitalpcr for more information.
© 2025 Bio-Rad Laboratories, Inc. 2
Past, Present and Future of Digital PCR
Table of Contents
Foreword.............................................................................................................................................3
The Evolution of Droplet Digital PCR Technology .............................................................................4
Blueprints of a Trusted Gold Standard: ddPCR Technology in Research and Development...........8
Advancing Quality Control in Cell and Gene Therapy Development With ddPCR technology........9
How ctDNA Analysis Takes Cancer From Bloodstreams to Breakthroughs ................................... 13
Droplet Digital PCR Revolutionizes Vaccine Development and Manufacturing.............................. 16
Building a Better Vaccine...................................................................................................................17
Resources .........................................................................................................................................20
© 2025 Bio-Rad Laboratories, Inc. 3
Past, Present and Future of Digital PCR
Foreword
Digital PCR (dPCR) is relied upon by researchers and clinicians across the
world for nucleic acid quantification. In particular, the rise of Droplet
Digital™ PCR (ddPCR™) has solidified dPCR as a competitive solution
providing increased sensitivity and precision.
Advances in modern medicine now offer the opportunity to treat diseases
previously thought to be incurable. From cell and gene therapy
development to cutting-edge cancer diagnostics, PCR solutions with
superior analytical capabilities are in high demand. Where quantitative
real-time PCR (qPCR) has previously been the standard method, digital
PCR has developed into a reliable way to achieve absolute quantification
of nucleic acids. In addition, the sensitivity offered by ddPCR presents a
significant advantage for detecting low-abundance targets, even in
complex sample matrices.
As a result, ddPCR has become an essential tool in many fields. By
overcoming the bias and variability concerns associated with relative
quantification, ddPCR is well-suited for quality control during therapeutics
development – ensuring the safety and efficacy of transformative cell and
gene therapies and vaccines. In cancer biomarker analysis, ddPCR
technology is a rapid and precise tool to analyze circulating tumor DNA,
harnessing the full potential of liquid biopsies.
This eBook explores the past, present and future of digital PCR, from its
humble beginnings to its establishment as a method trusted by researchers
worldwide. We discuss the analytical capabilities that make ddPCR so
versatile as well as the emerging applications of this technique in gene and
cell therapy, cancer biomarkers analysis and vaccine development.
© 2025 Bio-Rad Laboratories, Inc. 4
The Evolution of Droplet Digital
PCR Technology
The rediscovery of dPCR
Researchers use polymerase chain reaction (PCR) to
amplify specific nucleic acid sequences. Over the last 40
years, scientists have developed a wide range of novel
PCR methods, which can provide in-depth qualitative and
quantitative information. One such variation is quantitative
real-time PCR (qPCR). qPCR can determine the approximate
number of amplified molecules in a sample over time, but it
requires comparison of the sample to a standard curve. In
comparison, digital PCR (dPCR) does not require a standard
curve. Instead, an instrument subdivides each sample
into thousands of partitions, each of which function as an
individual PCR reaction. After amplification, each partition is
checked for fluorescently labeled DNA products and software
applies Poisson statistical analysis to quantify the absolute
number of target molecules in the sample.
The first description of dPCR – in which Saiki and colleagues
detected a single β-globin gene within 500,000 cells – was
published in 1988, predating qPCR.1 However, it was largely
abandoned in favor of qPCR until partitioning capabilities
improved, as dPCR was considered laborious and costly, with
limited usage (Figure 1).2 In 2006, dPCR gained new popularity
with the creation of microfluidic chips by Fluidigm, and the
creation of the first chip-based dPCR (cdPCR) platform. Then,
in 2010, Quantalife (later acquired by Bio-Rad) developed
the first Droplet Digital PCR (ddPCR) platform.3 However, it
was largely abandoned in favor of qPCR until partitioning
capabilities improved, as dPCR was considered laborious
and costly, with limited usage (Figure 1).2 In 2006, dPCR
gained new popularity with the creation of microfluidic chips
by Fluidigm, and the creation of the first chip-based dPCR
(cdPCR) platform. Then, in 2010, Quantalife (later acquired by
Bio-Rad) developed the first droplet dPCR (ddPCR) platform.3
Based on oil–water emulsions, the ddPCR instrument
partitions samples by generating 20,000 equally sized
droplets containing 0–1 template molecules. This enables
independent target amplification which the instrument
measures by passing the droplets through a fluorescence
detector. dPCR can now be used for a wide variety of
applications, including identifying copy number variants,
quantifying reference materials and analyzing liquid biopsies.
Exploring the dPCR publication
guidelines
Throughout its range of applications, reproducibility and
reliability of dPCR results is essential. To this end, Huggett
et al. published the minimum information for publication
of quantitative digital PCR experiments (dMIQE).4,5
These guidelines state the essential information that
researchers must include in their publications about their
dPCR experiments, to avoid publishing low-quality and
irreproducible data. As dPCR technology advanced and
became more popular, so too did the dMIQE guidelines, which
were then updated in 2020.
The information needed for the dMIQE falls under three
categories: sample preparation, dPCR protocol and data
analysis.
Sample preparation
Nucleic acid isolation and processing affect the accuracy
Digital PCR (dPCR) was gradually
developed over the last 35 years and
is now equipped to advance basic and
clinical research.
THE ORIGIN
OF dPCR
Future of dPCR
ຈ Cell and gene therapy development
ຈ Infection surveillance in patients
ຈ Monitoring environmental water
samples for infectious agents
1988
First published
description of dPCR1
1993
Quantitative real-time
PCR (qPCR) created3
2006
First chip-based dPCR
platform developed5
2013
dMIQE guidelines
published6
1990
dPCR first used for
target quantitation2
1999
The term digital PCR
came into use4
2010
First droplet dPCR
platform developed5
2020
dMIQE guidelines
updated7
Figure 1. Notable events in the development of dPCR.
© 2025 Bio-Rad Laboratories, Inc. 6
Past, Present and Future of Digital PCR
of dPCR experiments. Using a range of different methods,
such as enzymatic digestion and sonication, nucleic acids
can be extracted from a range of different specimens,
including clinical samples, animal models and cultured
cells. dPCR sensitivity can be improved by specific sample
processing methods such as restriction endonuclease-driven
DNA cleavage or DNA denaturing. However, the sample
processing methods must be considered during data analysis
for accurate results. For example, DNA denaturation produces
two template molecules, which can lead to an overestimation
in sequence quantity if not accounted for. If RNA is extracted,
the efficiency of RNA transcription to DNA will also affect
quantitation of target frequency. Therefore, information
about the reverse transcription step – such as RNA quantity,
reagents used and protocols employed – is essential for
reproducibility. The quality of extracted DNA must also
be assessed, as damaged nucleic acids can impair target
amplification and quantity.
dPCR protocol
Detailed information about pre-reaction mixture preparation
and the instrument used is essential for reproducibility, as
the chosen procedure can influence target amplification.
The dPCR protocol should be optimized to ensure that
primers are not dimerizing, and to find the best primer and
probe concentrations. Controls are key for reproducibility
and high-quality data, to assess if inhibitors are affecting
amplification and ensure that samples are not contaminated.
Positive controls should comprise primers, probes and
target sequence-containing DNA, while negative controls
should contain primers, probes but no template molecules.
Experimental variation should be monitored using biological
replicates and prepared using identical sample processing
procedures.
Data analysis
Finally, details about data analysis are also required for the
dMIQE. Example plots of positive and negative controls
should be provided to demonstrate assay success. In
addition, positive controls can be used to guide the setting of
appropriate thresholds to separate negative partitions from
positive partitions. Results from different biological replicates
should be compared to assess variation and estimate the
level of random error in the target frequency quantifications.
Overall, research and researchers that follow the dMIQE
guidelines ensure that their dPCR experiments are designed
correctly, their results are reproducible and their data are
comparable to other studies.
High-level precision across a
range of applications
Ensuring that dPCR studies are reproducible and robust is
essential as the technique becomes increasingly used across
a wide range of fields in both preclinical and clinical settings,
such as cell and gene therapies (CGT), vaccine development
and oncology. These contexts demand extremely high levels
of quality control and accuracy, and qPCR has typically been
the go-to method for measuring levels of genetic material.
However, dPCR offers several advantages over qPCR that are
now being explored.
ddPCR technology has been shown to give less variable
and more precise results than qPCR when quantifying viral
vector titres in CGT development.6,7
As the buffers required
for DNA purification can contain inhibitors that impair PCR
amplification, ddPCR can be more tolerant of inhibitors
than qPCR. In addition, ddPCR solutions have been shown
to have greater reproducibility and sensitivity at low copy
numbers than qPCR.8 ddPCR technology enables absolute
quantification of nucleic acids without requiring a standard
curve, even at very low levels, which is an advantage in
biomarker analysis and vaccine manufacturing.9,10
After decades of being overlooked in favour of qPCR,
dPCR – and in particular ddPCR – is now re-emerging as
a competitive, reliable technique for quantifying genetic
material. ddPCR technology provides researchers with
increased sensitivity, greater precision and enhanced inhibitor
tolerance, and it is therefore rapidly becoming an important
technique to evaluate genetic material in a variety of different
fields. However, it is essential for these techniques to be
performed and reported in line with the dMIQE guidelines to
ensure accurate, reproducible and high-quality results.11 This
eBook explores the emerging applications of ddPCR solutions,
and how it is transforming several different fields, including
CGT research, vaccine development and cancer biomarker
analysis to become a trusted method for researchers around
the world.
References
1. Morley AA. Digital PCR: A brief history. Biomol Detect Quantif. 2014;1(1):1–2.
doi: 10.1016/j.bdq.2014.06.001
2. Saiki RK, Gelfand DH, Stoffel S, et al. Primer-directed enzymatic
amplification of DNA with a thermostable DNA polymerase. Science.
1988;239(4839):487–491. doi: 10.1126/science.2448875
3. Baker M. Digital PCR hits its stride. Nat Methods. 2012;9(6):541–544. doi:
10.1038/nmeth.2027
4. Huggett JF, Foy CA, Benes V, et al. The digital MIQE guidelines: Minimum
information for publication of quantitative digital PCR experiments. Clin
Chem. 2013;59(6):892–902. doi: 10.1373/clinchem.2013.206375
5. The dMIQE Group, Huggett JF. The digital MIQE guidelines update: Minimum
information for publication of quantitative digital PCR experiments for 2020.
Clin Chem. 2020;66(8):1012–1029. doi: 10.1093/clinchem/hvaa125
6. Lin H-T, Okumura T, Yatsuda Y et al. Application of droplet digital PCR for
estimating vector copy number states in stem cell gene therapy. Hum Gene
Ther Methods. 2016;27(5):197–208. doi: 10.1089/hgtb.2016.059
7. Dobnik D, Kogovšek P, Jakomin T, et al. Accurate quantification and
characterization of adeno-associated viral vectors. Front Microbiol.
2019;10:1570. doi:10.3389/fmicb.2019.01570
8. Lou Y, Chen C, Long X, et al. Detection and quantification of chimeric
antigen receptor transgene copy number by droplet digital PCR
versus real-time PCR. J Mol Diagn. 2020;22(5):699–707. doi: 10.1016/j.
jmoldx.2020.02.007
9. Abachin E, Convers S, Falque S, Esson R, Mallet L, Nougarede N.
© 2025 Bio-Rad Laboratories, Inc. 7
Past, Present and Future of Digital PCR
Comparison of reverse-transcriptase qPCR and droplet digital PCR for the
quantification of dengue virus nucleic acid. Biologicals. 2018;52:49–50. doi:
10.1016/j.biologicals.2018.01.001
10. Gezer U, Bronkhorst AJ, Holdenrieder S. The clinical utility of droplet
digital PCR for profiling circulating tumor DNA in breast cancer patients.
Diagnostics. 2022;12(12):3042. doi: 10.3390/diagnostics12123042
11. Murphy LA, Marians RC, Miller K, et al. Digital polymerase chain reaction
strategies for accurate and precise detection of vector copy number in
chimeric antigen receptor T-cell products. Cytotherapy. 2023;25(1):94–102.
doi: 10.1016/j.jcyt.2022.09.004
Click here to discover end-to-end solutions for your workflow
Blueprints of a Trusted
Gold Standard: ddPCR
Technology in Research
and Development
After decades of being overlooked, digital PCR – particularly Droplet
Digital™ PCR (ddPCR™) – is now recognized as a robust, accurate
and reliable technique. In many areas of research, such as cell and
gene therapy, vaccines and cancer biomarkers, ddPCR technology
offers multiple advantages over other methods. Managing the safety
and efficacy of complex biopharmaceutical therapies and novel
vaccines can be a challenging and time-consuming process. In
oncology research, longitudinal biomarker monitoring requires rapid
turnarounds and minimally invasive sampling methods. In all these
fields – from initial discovery stages, through preclinical and clinical
testing, to manufacturing approved products – ddPCR technology has
emerged as a new gold standard method to respond to a myriad of
workflow challenges.
Key benefits of ddPCR technology
Precision Accuracy Inhibitor
tolerance
Multiplexing Time to results Sensitivity/
specificity
Click here to discover end-to-end solutions for your workflow
Trusted in cell and gene therapy development
From initial testing, through quality control and assessment into clinical research,
ddPCR solutions can support:
CMC submissions
Reliable assessment of purity,
efficacy and safety
Efficient and scalable process
development
Effective evaluation of therapies in
patients
Analyze
plasmid integrity
Detect mycoplasma
and residual DNA
Determine viral titer and
empty–full capsid ratio
Determine
biodistribution
Quantify transgene copy
number and expression
Evaluate
dose response
Applications
Trusted during vaccine development
From initial antigen characterization, through manufacture and into preclinical
and clinical studies, ddPCR solutions can help facilitate:
Tracking emerging variants
of evolving pathogens
Optimal cell line development
Quality control in manufacturing
Meeting regulatory guidelines
Effective evaluation of efficacy
in clinical studies
Applications
Discriminate between highly
specific strains and variants
Quantify viral load
and determine integrity
Identify stable and highly
expressing cell lines
Detect residual host DNA
contamination
Quantify transgene copy
number and expression
Evaluate
dose response
Trusted for biomarker detection and analysis
Due to its rapid turnaround, low costs and minimal, non-invasive sample
requirements, ddPCR solutions are used extensively in oncology research to
analyze circulating tumor DNA, enabling:
Detection of predefined biomarkers
Post-treatment monitoring
Prediction of treatment outcome
and relapse
More adaptive study designs
Development of personalized
medicine
Applications
Identify molecular residual
disease cells post-treatment
Assess rare genetic
variants
Determine treatment
effectiveness
© 2025 Bio-Rad Laboratories, Inc. 9
Advancing Quality Control in Cell
and Gene Therapy Development
With ddPCR technology
The landscape of modern medicine is being transformed by
the rapid advancement of cell and gene therapies (CGTs),
which offer unprecedented opportunities to treat previously
incurable diseases. With over 4,000 clinical trials and 139
clinically approved products globally, these therapies are
rapidly progressing from experimental concepts to clinical
reality.1 However, with this remarkable progress comes the
critical need for precise, accurate and reliable quality control
methods throughout the development and manufacturing
processes. Droplet Digital PCR (ddPCR) has emerged as a
pivotal technology in this space, offering superior analytical
capabilities that are essential for ensuring the safety and
efficacy of these complex therapeutic products.
Cell and gene therapy in a
nutshell
The journey of CGT began in 1928 when Frederick Griffith
discovered that cells could change their properties by taking
up exogenous genetic material.2 Since then, the field has
experienced both remarkable successes and significant
setbacks, including the tragic death of Jesse Gelsinger in
1999, which highlighted the critical importance of safety in
viral vector-based therapies.2
The therapeutic efficacy of CGT hinges on the precise
delivery and expression of nucleic acids inside the human
body. Gene therapy typically involves delivering exogenous
DNA into patient cells using vectors (often viral vectors).3 In
cell therapy, the patient’s cells or donor cells serve as the
DNA delivery mechanism. These cells are extracted, modified
ex vivo, expanded and reintroduced to the patient.4 This
approach eliminates many of the safety concerns associated
with direct viral injection while enabling mass production of
therapeutic cells.
The DNA being delivered can be manipulated through
recombinant DNA technologies, allowing for the restoration
of lost functions, the introduction of new capabilities or the
modulation of existing cellular processes. The advent of
advanced gene editing technologies, particularly CRISPR/
Cas systems, has accelerated progress in this field.5,6
Combined with improved viral vectors such as adenoassociated viruses (AAVs), which offer broad tissue
tropism with minimal immunogenicity, these technologies
have created new possibilities for treating a wide range of
conditions from hereditary genetic disorders to cancer and
cardiovascular disease.7
Yet, these therapies are complex and must be evaluated
not just for their biological activity, but also for their safety,
stability and manufacturing reproducibility.
The critical role of quality
control in CGTs
As cell and gene therapies move from laboratory bench to
clinical application, the complexity of manufacturing and
quality control has become increasingly apparent. These
therapies often work with patient or donor cells that are
© 2025 Bio-Rad Laboratories, Inc. 10
Past, Present and Future of Digital PCR
inherently heterogeneous and complex, making consistency
and reproducibility challenging. Moreover, current
manufacturing processes are largely manual, relying heavily
on operator experience and judgment, which can result in
considerable batch-to-batch variation.
Hence, quality control plays an essential role throughout
the CGT development pipeline, from initial research and
development through to clinical trials and commercial
manufacturing. Researchers must evaluate multiple
critical parameters, including viral vector integrity, purity,
concentration and the absence of contaminants. For
cell-based therapies, additional considerations include
transgene copy number, expression levels and cellular
viability. The precision and accuracy of these measurements
directly impact patient safety, dosage determination and
treatment efficacy.
Traditional quantitative real-time PCR (qPCR) has been the
standard method for many of these quality control tests.
However, qPCR relies on standard curves and reference
materials for quantification, which can introduce variability
and bias into the results. The technique is also susceptible
to inhibitors commonly used in purification buffers and other
manufacturing reagents, potentially leading to inaccurate
measurements. The advent of Droplet Digital PCR has allowed
researchers to address many of these limitations (Table
1).8,9,10,11 The ddPCR solution provides absolute quantification
without requiring standard curves or reference materials,
eliminating a major source of variability and bias. The
technique also enhances sensitivity, allowing detection of rare
targets that might be missed by qPCR. Additionally, ddPCR
technology demonstrates superior tolerance to inhibitors
commonly encountered in CGT manufacturing processes,
ensuring accurate measurements even in challenging
sample matrices.
Table 1: Comparison of qPCR and ddPCR technology characteristics.
Feature qPCR ddPCR Technology
Quantification Relative
(requires standard curve)
Absolute
(no standard curve)
Sensitivity Moderate High
(can detect single copies)
Inhibitor tolerance Low High
Reproducibility Variable High
Setup complexity Complex calibration Streamlined, fewer controls
ddPCR applications across the
CGT pipeline
The use of ddPCR solutions extend across multiple stages
of the CGT product lifecycle, offering critical analytical
capabilities from early research through commercial
manufacturing and clinical monitoring (Figure 1).
Why Scientists Across the CGT Pipeline
Trust ddPCR Technology
Stage Goals ddPCR Applications
Transgene design,
vector development
Copy number,
plasmid integrity
Scalable manufacturing,
QC assay design Viral titers, multiplex detection
Batch release contamination
control
Residual DNA,
mycoplasma detection
Biodistribution, persistence,
dose tracking
CAR copy quantification,
tissue monitoring
Absolute quantification
High sensitivity
Inhibitor tolerance
Reproducibility
Multiplexing capability
Figure 1. ddPCR technology application throughout the CGT product lifecycle.
© 2025 Bio-Rad Laboratories, Inc. 11
Past, Present and Future of Digital PCR
Research and development
In early-stage research, ddPCR solutions facilitate the
characterization of gene delivery vectors and engineered
cells. This capability is essential for optimizing transfection or
transduction protocols and ensuring consistent gene transfer
efficiency. Understanding how many copies of a transgene
are incorporated and whether they are stably expressed
is central to characterizing CGT products. Droplet Digital
PCR provides an accurate, reproducible measurement of
transgene copy number, even at low abundance or in highly
variable samples such as patient-derived cells. Researchers
have demonstrated that ddPCR technology provides greater
precision and reproducibility compared to qPCR when
quantifying vector copy numbers in transduced cells, leading
to more reliable results.10
Process development and
manufacturing
In manufacturing settings, ddPCR solutions serve multiple
quality control functions. The technology enables
manufacturers to precisely quantify vector titer, assess integrity
and confirm purity, which is critical for ensuring consistent
potency across production batches.12 ddPCR technology
excels at detecting low copy number sequences, verifying
the presence or absence of contaminants and measuring
residual DNA in final products. Moreover, unlike qPCR, which
can be affected by the various buffers and reagents used in
purification processes, ddPCR technology maintains accuracy
even in the presence of potential inhibitors.11 This reliability
is particularly important during process development, where
manufacturers need to evaluate the impact of different
purification methods on product quality.
For cell-based therapies, ddPCR solutions allow for the
precise monitoring of transgene expression levels and copy
number throughout the manufacturing process. This capability
is especially valuable for CAR-T cell therapies, where
the level of chimeric antigen receptor expression directly
impacts therapeutic efficacy. Studies have shown that ddPCR
technology can detect CAR copies at lower concentrations
than qPCR while providing superior repeatability and
reproducibility.9,13
Quality control and regulatory
compliance
The reproducibility, accuracy and speed of ddPCR solutions
make it particularly well-suited for good manufacturing
practice (GMP) settings and the stringent regulatory
expectations for CGT quality control. The technology’s ability
to provide absolute quantification without reliance on standard
curves reduces measurement uncertainty and supports more
robust regulatory submissions. Additionally, its enhanced
precision helps reduce the number of replicates required
for statistical significance, potentially streamlining analytical
workflows and reducing costs. ddPCR technology plays a
vital role in batch release, potency and purity testing. The
development of ddPCR assays for specific CGT applications,
such as the detection of mycoplasma contamination in cell
cultures, demonstrates the potential of the technique for
addressing unique quality control challenges in this field.
Preclinical and clinical studies
In clinical settings, ddPCR technology supports both safety
monitoring and efficacy assessment. ddPCR solutions enable
precise tracking of therapeutic cells in patient samples,
providing insights into biodistribution, persistence and
potential off-target effects.14 This monitoring is essential for
dose optimization and safety assessment, particularly for
novel therapies where long-term effects are not yet fully
understood. The enhanced sensitivity of ddPCR technology
also makes it valuable for detecting rare genetic variants or
low-level transgene expression.
Future directions
As CGTs move toward personalized medicine paradigms, the
demand for granular, high-fidelity molecular analysis tools will
only increase. Droplet Digital PCR is already expanding its role
from endpoint testing to integrated process analytics thanks
to the added flexibility of multiplexing. Moreover, ddPCR’s
compatibility with single-cell and low-input workflows makes
it well-suited for future advancements, including single-cell
profiling of gene-modified cells, integration with AI-driven
manufacturing systems and in-process biosensor feedback in
automated cell culture environments.15
As regulatory agencies continue to raise the bar on analytical
validation, ddPCR’s reproducibility and compliance with
standards such as dMIQE (minimum information for
publication of quantitative digital PCR experiments) position it
as a trusted cornerstone of CGT development.16
The integration of ddPCR solutions into CGT workflows
is more than just a technological upgrade; it represents
a commitment to the highest standards of quality and
safety that these life-changing therapies demand. As the
field continues to evolve, ddPCR technology will play an
increasingly important role in ensuring the safety and efficacy
of these transformative therapies, ultimately contributing to
better patient outcomes and the continued advancement of
precision medicine.
Learn more about ddPCR
applications in CGT here
© 2025 Bio-Rad Laboratories, Inc. 12
Past, Present and Future of Digital PCR
References
1. American Society of Gene and Cell Therapy, Citeline. The Gene, Cell, & RNA
Therapy Landscape report. Q1 2025 Quarterly Data Report. https://www.
asgct.org/global/documents/asgct-citeline-q1-2025-report.aspx
2. Wirth T, Parker N, Ylä-Herttuala S. History of gene therapy. Gene.
2013;525(2):162–169. doi: 10.1016/j.gene.2013.03.137
3. Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G. Viral vector platforms within the
gene therapy landscape. Sig Transduct Target Ther. 2021;6:53. doi: 10.1038/
s41392-021-00487-6
4. Bashor CJ, Hilton IB, Bandukwala H, Smith DM, Veiseh O. Engineering
the next generation of cell-based therapeutics. Nat Rev Drug Discov.
2022;21:655–675. doi: 10.1038/s41573-022-00476-6
5. Li H, Yang Y, Hong W, Huang M, Wu M, Zhao X. Applications of genome
editing technology in the targeted therapy of human diseases: mechanisms,
advances and prospects. Sig Transduct Target Ther. 2020;5:1. doi: 10.1038/
s41392-019-0089-y
6. Uddin F, Rudin CM, Sen T. CRISPR gene therapy: applications, limitations,
and implications for the future. Front Oncol. 2020;10:1387. doi: 10.3389/
fonc.2020.01387
7. Lundstrom K. Viral vectors in gene therapy. Diseases. 2018;6(2):42. DOI:
10.3390/diseases6020042
8. Baker M. Digital PCR hits its stride. Nat Methods. 2012;9(6):541–544. doi:
10.1038/nmeth.2027
9. Lou Y, Chen C, Long X, et al. Detection and quantification of chimeric
antigen receptor transgene copy number by droplet digital PCR
versus real-time PCR. J Mol Diagn. 2020;22(5):699–707. doi: 10.1016/j.
jmoldx.2020.02.007
10. Lin H-T, Okumura T, Yatsuda Y et al. Application of droplet digital PCR for
estimating vector copy number states in stem cell gene therapy. Hum Gene
Ther Methods. 2016;27(5):197–208. doi: 10.1089/hgtb.2016.059
11. Dobnik D, Kogovšek P, Jakomin T, et al. Accurate quantification and
characterization of adeno-associated viral vectors. Front Microbiol.
2019;10:1570. doi:10.3389/fmicb.2019.01570
12. Furuta-Hanawa B, Yamaguchi T, Uchida E. Two-dimensional droplet digital
PCR as a tool for titration and integrity evaluation of recombinant adenoassociated viral vectors. Hum Gene Ther Met. 2019;30(4):127–136. doi:
10.1089/hgtb.2019.031
13. Murphy LA, Marians RC, Miller K, et al. Digital polymerase chain reaction
strategies for accurate and precise detection of vector copy number in
chimeric antigen receptor T-cell products. Cytotherapy. 2023;25(1):94–102.
doi: 10.1016/j.jcyt.2022.09.004
14. Fehse B, Badbaran A, Berger C, et al. Digital PCR assays for precise
quantification of CD19-CAR-T cells after treatment with axicabtagene
ciloleucel. Mol Ther Methods Clin Dev. 2020;16:172–178. doi: 10.1016/j.
omtm.2019.12.018
15. Moutsatsou P, Ochs J, Schmitt RH, Hewitt CJ, Hanga MP. Automation in
cell and gene therapy manufacturing: from past to future. Biotechnol Lett.
2019;41:1245–1253. doi: 10.1007/s10529-019-02732-z
16. Huggett JF, Foy CA, Benes V, et al. The digital MIQE guidelines: Minimum
information for publication of quantitative digital PCR experiments. Clin
Chem. 2013;59(6):892–902. doi: 10.1373/clinchem.2013.206375
© 2025 Bio-Rad Laboratories, Inc. 13
How ctDNA Analysis Takes Cancer
From Bloodstreams to Breakthroughs
Introduction
When healthy cells break down, their DNA is released into
the bloodstream. This is termed cell-free DNA (cfDNA). In
patients with cancer, the concentration of cfDNA in blood
samples increases significantly and it does so in line with the
progression stage of the cancer. Between 0.01% and 90%
of the cfDNA in cancer patients originates from tumors. The
increase in cfDNA is due to the additional deposition of DNA
in the bloodstream released by malignant tumors, called
circulating tumor DNA (ctDNA).
ctDNA has emerged as one of the most informative
biomarkers in modern oncology research, and it can be
detected through liquid biopsies, such as a standard blood
test. This provides a non-invasive, repeatable and convenient
way to detect cancer and monitor disease progression.
Its adoption has opened new opportunities for cancer
researchers to study dynamic tumor biology without the
limitations of repeated tissue biopsies. This article explores
approaches to circulating tumor DNA (ctDNA) analysis,
recognizing the value of broad genomic profiling in early
research while emphasizing the need for highly sensitive,
quantitative tools for ongoing monitoring and clinical
investigation.
ctDNA: A real-time molecular
window into cancer
The non-invasive nature of ctDNA sampling goes hand in
hand with one of its most clinically relevant characteristics –
its short half-life, typically less than two hours. This provides
an opportunity to monitor changes in tumor behavior in
near real time. The ease of sample collection also allows
researchers to collect serial samples throughout treatment,
offering a dynamic picture of tumor response to treatment and
the detection of molecular residual disease, a key indicator
of relapse.
In contrast to traditional tissue biopsies, which are invasive
and offer only a static snapshot of the tumor, ctDNA analysis
provides a safer and more scalable method for capturing
tumor heterogeneity and tracking disease progression across
multiple time points. For instance, a plasma analysis study
identified common driver mutations in EGFR and KRAS in a
matter of days, potentially enabling the detection of EGFR
T790M drug-resistant variants that are otherwise missed in
tissue biopsy samples due to high tumor heterogeneity.1
However, ctDNA is typically present in extremely low
concentrations, especially in early-stage disease or after
treatment. Detecting and quantifying these rare fragments
requires high-performance molecular tools capable of
exceptional sensitivity, reproducibility and speed. While
various methods are suitable, digital PCR (dPCR) has
become a preferred method for addressing these demands,
enabling researchers to harness the full potential of ctDNA in
longitudinal cancer studies and biomarker-driven research.
Choosing the right method for
ctDNA detection
Next-generation sequencing (NGS) has the power to rapidly
sequence large quantities of DNA, making it a contender
© 2025 Bio-Rad Laboratories, Inc. 14
Past, Present and Future of Digital PCR
for analyzing ctDNA concentration
in blood samples. By providing a
comprehensive view of the genetic
composition of tumors, it can identify
known and unknown mutations,
including copy number variations and
structural rearrangements. For example,
a study found that NGS could identify
actionable mutations from non-small
cell lung cancer (NSCLC) plasma
samples.2 This method enabled more
effective personalized therapy in a
higher percentage of patients compared
to when mutations were identified using
NGS on tissue samples.
However, NGS is not without limitations.
The cost, time requirements and need
for specialized expertise make NGS
less practical and accessible than
alternative approaches like dPCR.
Nevertheless, NGS remains essential
in the early stages of clinical research,
particularly in the diagnostic stage, due
to its unparalleled breadth of screening.
It is especially relevant in situations
where specific mutations have not yet
been identified or when a broad survey
of the genome is needed.
On the other hand, dPCR, specifically
Droplet Digital PCR (ddPCR), addresses
the limitations of NGS and consequently
has emerged as a leading method in
ctDNA analysis. This technique is rapid,
precise and highly sensitive, ideal
for detecting rare genetic variants in
ctDNA, which may comprise only 0.01%
of cfDNA in a sample. The qualities of
this technique make it highly suitable
for a range of applications in cancer
analysis (Figure 1).
Applications of
digital PCR in ctDNA
research
A broad portfolio of analytically
validated ddPCR assays has been
established to target high-value cancer
mutations, supporting researchers in
the investigation of clinically relevant
biomarkers. The effectiveness of this
approach has been demonstrated
across multiple cancer types in clinical
research studies.
Breast cancer
Aside from initial diagnosis and
subsequent treatment, a vital part of
disease management is detecting
and addressing relapses. Molecular
residual disease (MRD) refers to the
small number of cancer cells remaining
in the body after treatment. Although
they are strong indicators of relapses
in many cancers, standard screening
methods often fail to detect them.
ddPCR technology has been shown
to accurately predict relapse in earlystage breast cancer, while ctDNA
sequencing has been used to identify
MRD-associated mutations more
effectively than sequencing the primary
tumor.3 Research has also found that
after treatment for triple-negative breast
cancer, the absence of ctDNA in ddPCR
analysis was linked to a reduced risk of
relapse and better overall prognosis.4
Non-small cell lung
cancer
Standard clinical monitoring of NSCLC
involves imaging methods that
identify gross changes in tumor size
or structure. However, single-gene
ddPCR analysis has demonstrated its
value as a cost-effective tool capable
of predicting early disease progression
and sustained responses to immune
checkpoint inhibitors during treatment.5
Melanoma
ddPCR technology has also been
applied to monitoring treatment
response in patients with melanoma.
While traditionally monitored by
computer tomography, this imaging
technique is expensive and timeconsuming. Studies exploring how
ddPCR solutions can help to predict
treatment response by measuring
ctDNA in melanomas have revealed
that ddPCR technology achieves higher
sensitivity than NGS and does so at
a faster pace and lower cost.6 The
technique has also been used to predict
survival and treatment outcomes of
melanoma patients.7
Colorectal cancer
Using current imaging and laboratory
methods, it is notoriously challenging
to detect relapses in bowel cancer
patients. However, through ddPCRDiagnosis
and
Treatment
MRD
Assessment
PostTreatment
Monitoring
Recurrence
and New
Treatment
PostTreatment
Monitoring
Recurrence
Monitoring
Cancer Timeline
Best Fit cDNA Analysis Method
Area of Clinical Research
Next Generation
Sequencing (NGS)
Figure 1. NGS remains essential in the diagnosis and treatment stage of clinical research, while ddPCR
technology is most effective in subsequent analysis.
ddPCR Technology
© 2025 Bio-Rad Laboratories, Inc. 15
Past, Present and Future of Digital PCR
based ctDNA analysis, early changes in ctDNA levels could
signal disease progression, facilitating prompt clinical
response and better outcomes.8
Advancing research through
longitudinal monitoring
One of the most impactful advantages of ddPCR technology
is its ability to support frequent, minimally invasive ctDNA
sampling throughout treatment courses. This longitudinal
approach provides insights that static tissue biopsies cannot
– revealing changes in tumor dynamics, emerging resistance
mechanisms and evolving biomarker profiles across multiple
cancer types.
As a result, ddPCR technology is not only accelerating
biomarker discovery but also enabling more adaptive and
responsive clinical study designs. In some research settings,
ctDNA levels measured by ddPCR solutions are being
evaluated as surrogate endpoints in clinical trials. These
molecular endpoints may allow for earlier evaluation of
therapeutic efficacy, potentially reducing trial duration, cost
and participant burden.
ctDNA and ddPCR technology:
A partnership for personalized
medicine
As precision oncology advances, the partnership between
ctDNA analysis and dPCR is proving essential to both
translational cancer research and the development of truly
personalized cancer care. ctDNA provides a dynamic,
minimally invasive view of tumor genetics, while dPCR
delivers the sensitivity, precision and quantification needed
to track those changes with confidence. Together, they
enable researchers to detect rare variants, monitor treatment
response and assess MRD across a range of cancer types
and study designs. This powerful combination is helping
translate real-time molecular insights into more informed
clinical decisions, bringing personalized medicine closer to
routine practice.
Conclusion
The use of Droplet Digital PCR for ctDNA analysis is
increasingly being recognized as the cornerstone of modern
liquid biopsy research and central to cancer research. While
broad genomic profiling through NGS remains important in the
early stages of cancer research, clinical settings often require
a more focused approach – one that can detect rare mutations
with high sensitivity, deliver rapid results and integrate
seamlessly into everyday workflows.
The accuracy and ease of use of dPCR have made it a trusted
method for researchers studying a wide range of cancers
and trial designs. As oncology moves toward more adaptive,
personalized treatment strategies, the need for real-time
monitoring of tumor genetics continues to grow, and dPCR is
well-positioned to meet that demand.
Learn more here
References
1. Sacher AG, Paweletz C, Dahlberg SE, et al. Prospective validation of rapid
plasma genotyping for the detection of EGFR and KRAS mutations in
advanced lung cancer. JAMA Oncol. 2016;2(8):1014–1022. doi: 10.1001/
jamaoncol.2016.0173
2. Aggarwal C, Thompson JC, Black TA, et al. Clinical implications of plasmabased genotyping with the delivery of personalized therapy in metastatic
non–small cell lung cancer. JAMA Oncol. 2019;5(2):173–180. doi: 10.1001/
jamaoncol.2018.4305
3. Garcia-Murillas I, Schiavon G, Weigelt B, et al. Mutation tracking in circulating
tumor DNA predicts relapse in early breast cancer. Sci Transl Med.
2015;7(302):302ra133. doi: 10.1126/scitranslmed.aab0021
4. Cavallone L, Aguilar-Mahecha A, Lafleur J, et al. Prognostic and predictive
value of circulating tumor DNA during neoadjuvant chemotherapy for triple
negative breast cancer. Sci Rep. 2020;10:14704. doi: 10.1038/s41598-020-
71236-y
5. van der Leest P, Hiddinga B, Miedema A, et al. Circulating tumor DNA
as a biomarker for monitoring early treatment responses of patients with
advanced lung adenocarcinoma receiving immune checkpoint inhibitors. Mol
Oncol. 2021;15(11):2910–2922. doi: 10.1002/1878-0261.13090
6. Forthun RB, Hovland R, Schuster C, et al. ctDNA detected by ddPCR reveals
changes in tumour load in metastatic malignant melanoma treated with
bevacizumab. Sci Rep. 2019;9:17471. doi: 10.1038/s41598-019-53917-5
7. Syeda MM, Wiggins JM, Corless BC, et al. Circulating tumour DNA in
patients with advanced melanoma treated with dabrafenib or dabrafenib plus
trametinib: a clinical validation study. Lancet Oncol. 2021;22(3):370–380.
doi: 10.1016/S1470-2045(20)30726-9
8. Holm M, Andersson E, Osterlund E, et al. Detection of KRAS mutations
in liquid biopsies from metastatic colorectal cancer patients using
droplet digital PCR, Idylla, and next generation sequencing. PloS One.
2020;15(11):e0239819. doi: 10.1371/journal.pone.0239819
© 2025 Bio-Rad Laboratories, Inc. 16
Droplet Digital PCR Revolutionizes
Vaccine Development and
Manufacturing
Modern vaccine development demands unprecedented
precision in measuring and characterizing antigens, immune
responses and product quality. Traditional analytical methods
often fall short of the sensitivity and precision requirements
needed, contributing to development timelines that can
span 5–10 years. Analytical bottlenecks throughout the
development pipeline create delays that ultimately impact
global health outcomes. This is particularly evident during
public health emergencies like the COVID-19 pandemic.1
Droplet Digital PCR (ddPCR) has emerged as a transformative
analytical technology that addresses these challenges across
vaccine research and manufacturing. By partitioning reactions
across thousands of droplets, ddPCR technology enables
absolute quantification without standard curves – making it the
gold standard for viral vector genome titer determination.
This article demonstrates how ddPCR technology supports
every stage of vaccine development, from initial antigen
identification through large-scale manufacturing and quality
control. By highlighting specific applications, it explores how
this technology is accelerating vaccine development timelines
while enhancing product safety and efficacy profiles.
The evolution of vaccine
analytics
Vaccine development has evolved dramatically,
encompassing diverse platforms, each of which present
unique analytical challenges. Modern vaccines include wholeorganism vaccines (both live attenuated and inactivated),
subunit vaccines containing specific antigenic components,
viral vector platforms that deliver genetic material and nucleic
acid vaccines including mRNA and DNA constructs.2,3 Each
platform requires specialized analytical approaches for
development and quality control, from characterizing complex
protein structures in subunit vaccines to ensuring precise
genetic payload delivery in viral vector systems.
Traditional analytical methods have struggled to meet
the demanding requirements of these advanced vaccine
platforms. Conventional qPCR methods rely on standard
curves for quantification, introducing variability and potential
bias that can compromise analytical accuracy. These
methods also struggle to detect low-abundance targets or
discriminate between closely related sequences. Issues with
reproducibility and precision also create regulatory approval
bottlenecks, extending development timelines and delaying
access to life-saving vaccines.
ddPCR technology represents a step forward through its
partition-based approach, which eliminates standard curve
dependency by distributing the sample across thousands
of individual reaction chambers. ddPCR solutions integrate
throughout the complete vaccine development pipeline, from
initial antigen discovery and characterization through to largescale manufacturing and quality control processes (Figure 1).
This approach enables direct counting of target molecules,
providing absolute quantification with superior precision and
significantly reduced bias. This generates more reliable data
Figure 1. ddPCR solutions in the vaccine development pipeline.
Verify Expression
• Develop a vaccine that delivers the
candidate antigen or its sequence.
• Assess antigen production and
purity in the chosen system by
western blot and SDS-PAGE.
Identify an Antigen
• Find an antigen that induces
protective immunity by
assessing interactions with
neutralizing antibodies using
the ZE5 Cell Analyzer.
• Detect antigen variants
with Droplet Digital PCR
(ddPCR) using a QX200
or QX ONE instrument.
Verify Immunity
• Determine the viral load required
to induce pathogenicity with
ddPCR systems.
• Quantitate viral titer for
vaccine challenge experiments
with ddPCR systems.
Analyze the Immune Response
• Track immune responses to
immunization by measuring
chemokines and cytokines with
Bio-Plex Immunoassays.
• Phenotype immune cells that
respond to vaccination with
the ZE5 Cell Analyzer.
Building a Better Vaccine
Throughout vaccine research and development, scientists must analyze their antigens, track the immune response
and determine viral loads. Modern instruments and assays aid vaccine developers and ensure that they receive
fast and reliable results.
© 2025 Bio-Rad Laboratories, Inc. 18
Past, Present and Future of Digital PCR
throughout the vaccine development pipeline, supporting
faster decision-making and more confident regulatory
submissions.4
ddPCR Technology enables
precision throughout vaccine
R&D
The analytical precision of Droplet Digital PCR can be applied
to multiple stages of vaccine research and development,
beginning with antigen identification and characterization.
The technology is able to detect single-nucleotide variants
and discriminate between closely related viral or bacterial
strains. This capability enables researchers to track emerging
variants within populations, which proved essential during
the COVID-19 pandemic, and it helps them make informed
decisions about vaccine formulation, ensuring continued
efficacy against evolving pathogens.5,6
Cell line development represents another application where
ddPCR solutions’precision drive manufacturing success.
Production cell lines must express target antigens consistently
over numerous passages while maintaining genetic
stability. ddPCR technology precisely measures transgene
copy numbers, providing researchers with reliable data to
assess genetic stability and identify high-expressing, stable
clones.7
Accurate copy number determination is essential for
establishing production cell lines that can reliably manufacture
vaccine antigens at a commercial scale while meeting
stringent quality requirements.
Immune response analysis and vaccine efficacy testing
also benefit significantly from ddPCR technology’s
quantification capabilities. The technology plays a role in
viral load quantification for vaccine challenge experiments,
where precise measurements are essential for determining
protective efficacy. Droplet Digital PCR enables researchers
to establish accurate viral dosages required for pathogenicity
studies. This ensures that challenge experiments provide
meaningful data while maintaining appropriate safety margins.
This precision is particularly important in preclinical studies
where dosage accuracy directly impacts the validity of
efficacy conclusions.
Viral vector vaccine development has embraced ddPCR
solutions as the gold standard for genome titer determination.8
Traditional qPCR methods often struggle with the complex
matrices and potential inhibitors present in viral vector
preparations, while ddPCR’s partition-based approach
provides more robust and accurate quantification. The
technology measures vector infectivity and payload delivery
to host cells, enabling developers to ensure efficacious,
non-toxic dosing. This has become increasingly important as
viral vector platforms gain prominence for vaccines against
infectious diseases and in gene therapy applications.
Quality control and
manufacturing excellence
Large-scale vaccine manufacturing demands rigorous quality
control measures, and ddPCR technology is an important
tool for ensuring product consistency and safety. Impurity
detection is a critical quality control application where
ddPCR solutions’ sensitivity provide significant advantages.
The technology detects minute quantities of host cell DNA
(HCD) contamination, a regulatory concern that requires
careful monitoring throughout manufacturing processes.9,10
Unlike traditional methods that may require extensive sample
preparation, ddPCR method provides direct quantification
without DNA purification steps, streamlining quality control
workflows while maintaining regulatory compliance.
Regulatory agencies worldwide have established strict
requirements for HCD reporting in biological products, and
ddPCR has become the preferred method for meeting these
standards.9 The technology reliably detects HCD levels
below regulatory thresholds, providing manufacturers with
confidence in their product purity assessments. Additionally,
ddPCR solutions provide DNA size measurements that help
characterize the nature of any detected contamination,
supporting comprehensive safety evaluations.
Potency testing for advanced vaccine platforms presents
unique challenges that ddPCR method addresses effectively.
Nucleic acid vaccines, including mRNA constructs encased
in lipid nanoparticles, require precise measurement of active
ingredient concentrations to ensure consistent dosing. Droplet
Digital PCR accurately quantifies these complex formulations,
determining both the concentration of delivery vehicles and
the integrity of genetic payloads.11 For viral vector vaccines,
the technology measures payload delivery within host cells,
ensuring that each vaccine dose contains the optimal amount
of active genetic material.
Manufacturing scalability and consistency depend on robust
analytical methods that can maintain accuracy across
different production scales and facilities. ddPCR solutions
ensure batch-to-batch consistency in large-scale production
by providing reliable, standardized measurements that
are independent of operator variability or standard curve
preparation. The technology supports real-time quality
monitoring throughout manufacturing processes, enabling
rapid detection of any deviations that could impact product
quality. This is particularly valuable for meeting stringent
regulatory requirements for vaccine approval, where
consistent analytical data across multiple manufacturing sites
may be required.
© 2025 Bio-Rad Laboratories, Inc. 19
Past, Present and Future of Digital PCR
Conclusions and future
prospects
ddPCR technology has already transformed vaccine
development and manufacturing practices by providing
the analytical precision and reliability required for modern
vaccine platforms. The technology enables faster, more
precise vaccine development timelines by eliminating
analytical bottlenecks that historically contributed to lengthy
development cycles.
The major advantages of ddPCR solutions across the vaccine
development pipeline include absolute quantification without
standard curves, superior precision and reduced bias,
enhanced sensitivity for detecting low-abundance targets
and robust performance in complex sample matrices. These
capabilities make ddPCR technology an essential tool for
antigen characterization, cell line development, efficacy
testing and manufacturing quality control. As vaccine
platforms continue to evolve and become more sophisticated,
the analytical demands will only increase, making Droplet
Digital PCR adoption increasingly critical for success.
Discover how ddPCR solutions
can accelerate your vaccine
development programs
References
1. Chaudhary N, Weissman D, Whitehead KA. mRNA vaccines for infectious
diseases: principles, delivery and clinical translation. Nat Rev Drug Discov.
2021;20(11):817–838. doi: 10.1038/s41573-021-00283-5
2. Liu MA. A comparison of plasmid DNA and mRNA as vaccine technologies.
Vaccines. 2019;7(2):37. doi: 10.3390/vaccines7020037
3. Ura T, Okuda K, Shimada M. Developments in Viral Vector-Based Vaccines.
Vaccines. 2014;2(3):624–641. doi: 10.3390/vaccines2030624
4. Vasudevan HN, Xu P, Servellita V, et al. Digital droplet PCR accurately
quantifies SARS-CoV-2 viral load from crude lysate without nucleic acid
purification. Sci Rep. 2021;11:780. doi: 10.1038/s41598-020-80715-1
5. Ciesielski M, Blackwood D, Clerkin T, et al. Assessing sensitivity and
reproducibility of RT-ddPCR and RT-qPCR for the quantification of SARSCoV-2 in wastewater. J Virol Methods. 2021;297:114230. doi: 10.1016/j.
jviromet.2021.114230
6. Liu H, Wu NC, Yuan M, et al. Cross-neutralization of a SARS-CoV-2
antibody to a functionally conserved site is mediated by avidity. Immunity.
2020;53(6):1272–1280.e5. doi: 10.1016/j.immuni.2020.10.023
7. Berman J, Heredia N, Regan J et al. Droplet digital PCR: High-resolution
copy number variation analysis; Bulletin 6475. Bio-Rad. www.bio-rad.com/
sites/default/files/webroot/web/pdf/lsr/literature/Bulletin_6475.pdf. Accessed
August 5, 2025.
8. Alliance for Regenerative Medicine, National Institute for Innovation in
Manufacturing Biopharmaceuticals. Generation of a quality target product
profile. In: Project A-Gene. 2021. https://alliancerm.org/wp-content/
uploads/2021/09/ALL-PROJECT-A-GENE-V10.pdf. Accessed August 5, 2025.
9. World Health Organization. Recommendations for the evaluation of animal
cell cultures as substrates for the manufacture of biological medicinal
products and for the characterization of cell banks, Annex 3, TRS No 978.
https://www.who.int/publications/m/item/animal-cell-culture-trs-no-978-
annex3. Published May 22, 2013. Accessed August 5, 2025.
10. Do D, Ma M, Klitgord N, Regan J, Ngo N. Direct quantification of residual host
cell DNA. Bio-Rad. www.bioradiations.com/wp-content/uploads/2015/05/
direct-quantification-of-residual-host-cell-dna.pdf. Accessed August 5,
2025.
11. Mucker EM, Karmali PP, Vega J, et al. Lipid nanoparticle formulation
increases efficiency of DNA-vectored vaccines/immunoprophylaxis in
animals including Transchromosomic Bovines. Sci Rep. 2020;10:8764. doi:
10.1038/s41598-020-65059-0
© 2025 Bio-Rad Laboratories, Inc. 20
Past, Present and Future of Digital PCR
Resources
Become a Master of ddPCR
Digital PCR is transforming the modern molecular biology lab and it’s easier to master than you might think!
With its simple technology and chemistry, the key to success lies in understanding how it works in real-world conditions.
This video series will guide you through both foundational concepts and advanced applications, helping you build the expertise
needed to master digital PCR.
Watch the videos here to unlock the full potential of digital PCR!
Brought to you by
Download the eBook 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