Advancing Cancer Treatment With Synthetic DNA
eBook
Published: June 26, 2025

Credit: Twist Bioscience
NGS and CRISPR technologies have revolutionized our understanding of cancer genetics, enabling precision diagnostics and targeted therapeutic approaches that were unimaginable a decade ago.
However, the extreme heterogeneity of cancers complicates the identification of universal biomarkers and effective treatments.
Synthetic DNA changes this. By providing highly uniform, scalable and precise genetic materials, synthetic DNA enables researchers to create more reliable models and improve the accuracy of sequencing.
This eBook explores how synthetic DNA is transforming cancer research, from uncovering genetic mechanisms to developing targeted therapies and improving disease models.
Download this eBook to discover:
- How synthetic DNA standards calibrate NGS platforms for ultra-rare mutation detection
- The role of synthetic DNA in developing novel cancer treatments, including CAR T-cell therapies
- How synthetic DNA supports cutting-edge CRISPR applications to create better disease models and therapeutic targets
SYNTHETIC DNA:
THE FABRIC OF MODERN
CANCER RESEARCH
TWIST BIOSCIENCE
2
Table of Contents
01 THE FADING ENIGMA OF CANCER�������������������������������������������������������������������������������� 3
Synthetic DNA Technology Overview������������������������������������������������������������������������������������������������������������������ 4
02 INTRODUCTION: SYNTHESIZING LIFE’S BLUEPRINTS���������������������� 4
Improving Research Models �������������������������������������������������������������������������������������������������������������������������������� 10
Identifying Therapeutic Targets �������������������������������������������������������������������������������������������������������������������������� 10
Uniformity in Genetic Engineering��������������������������������������������������������������������������������������������������������������������� 12
04 CRISPR: TINKERING WITH THE CANCER GENOME ������������������������������ 10
DNA SYNTHESIS ACROSS THE CANCER
RESEARCH CONTINUUM ��������������������������������������������������������������������������������������������������������� 19
06
07 REFERENCES�������������������������������������������������������������������������������������������������������������������������������������������� 21
2
05 ANTIBODIES AND CARs: SYNTHESIZING A NEW FORM OF HOPE.....14
Therapeutic Antibody Development������������������������������������������������������������������������������������������������������������������ 14
Cell-Based Therapy Development ��������������������������������������������������������������������������������������������������������������������� 17
Twist products are for research use only. They are not intended for the diagnosis, prevention, or treatment of a disease or condition. Twist Bioscience
assumes no liability regarding use of our products for applications in which they are not intended. The results presented are customer-specific and should
not be interpreted as indicative of performance across all institutions.
TARGET ENRICHMENT: CAPTURING THE MALIGNANT
GENETIC LANDSCAPE ������������������������������������������������������������������������������������������������������������������� 6
03
Liquid Biopsy Research������������������������������������������������������������������������������������������������������������������������������������������� 7
Methylation Detection��������������������������������������������������������������������������������������������������������������������������������������������� 8
The Importance of Uniformity ������������������������������������������������������������������������������������������������������������������������������� 9
TWIST BIOSCIENCE
3
Now is a great time to be in the cancer research space. Recent technological and scientific advances
have empowered researchers with a diverse array of tools, each of which helps us better understand
this vexing disease. Next-Generation Sequencing (NGS) is exposing the nuanced genetic landscape
of cancer, highlighting potential therapeutic and diagnostic strategies. CRISPR/Cas gene editing allows
researchers to methodically annotate pathological gene networks while also enabling the creation of
highly personalized cell and gene therapies.
Put simply, the enigma of cancer is slowly dissolving, washed away by modern technology.
Central to this process is synthetic DNA—a basic material that is critical for NGS; it forms the basis
of every CRISPR experiment and is foundational to the rise of Chimeric Antigen Receptor (CAR) and
T-cell receptor therapeutics.
In the following sections, we explore the many ways that synthetic DNA is used in modern cancer
research and therapeutic development. Along the way, we highlight products that are empowering
researchers to overcome the frustration that has long characterized cancer research.
Synthetic DNA is instrumental in:
01 The Fading
Enigma of Cancer
Discovery Research: uncovering mechanisms of disease
and building in vitro models
Detection: identifying biomarkers for disease detection
and characterization
Therapeutic Development: generating novel treatments
TWIST BIOSCIENCE
4
Synthetic DNA is indispensable in modern molecular biology. Artificial genes can be synthesized and
inserted into a bacterium’s genome, transforming it into an antibody-producing biofactory. Alternatively,
synthetic DNA can be used as the bait that captures specific DNA fragments from a sample, separating
the DNA you want to sequence from the DNA you’re not interested in. This simple step can improve the
efficiency and depth of sequencing efforts.
The advent of modern DNA synthesis platforms is one of the many important advances of the last
century that have collectively empowered researchers to explore the molecular world. Natural DNA
consists of nucleotides organized into repeating units that form a chemical chain, with each nucleotide
linked to another by the action of enzymes. Methods for creating synthetic DNA mimic this process
through various means, with the most widely used and validated approach involving solid-phase
phosphoramidite chemistry (Figure 1).
Exploring the differences between DNA synthesis platforms is beyond the scope of this ebook, but small
nuances in their approaches can have a significant impact on their accuracy, uniformity, and scalability.
Twist has become a leading DNA synthesis company through its optimized silicon-based synthesis
platform, capable of producing up to 9600 genes on a single silicon chip (Figure 2). With this technology,
Twist is supporting researchers across the cancer continuum, from basic cancer biology
to preclinical therapeutic development (Figure 3).
Figure 1: Twist’s phosphoramidite reaction cycle.
02 Introduction:
Synthesizing Life’s Blueprints
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
5
Figure 2: Twist’s silicon chip greatly expands DNA synthesis capacity compared to traditional methods.
Figure 3: Twist provides solutions to empower researchers across the continuum of cancer research.
DETECTION
• MRD test development
• Liquid biopsy research
• Tumor profiling
• Tumor mutational burden
THERAPEUTIC
DEVELOPMENT
• Target identification
• Target validation
• Biologics development
• Cancer vaccine development
DISCOVERY
• Population genomics
• Disease characterization
• Pathway analyses
• Disease modeling
THE CONTINUUM OF CANCER RESEARCH
Would you like to learn more about how Twist can help your research in the
fight against cancer? Watch this webinar about all the tools for the good fight.
TRADITIONAL METHODS
96 Oligos = 1 Gene
TWIST SILICON PLATFORM
>1 Million Oligos + 9,600 Genes
TWIST BIOSCIENCE
6
To better understand the genetic changes driving cancers, there may be no better technology than NGS.
NGS allows researchers to explore the genetic landscape of tumor cells, cataloging an ever-expanding
range of mutations in DNA and recording aberrant RNA expression.
Have a specific
set of targets in mind?
Twist can help you design custom
target enrichment panels. Our team
of bioinformaticians works with you
to rapidly develop a highly uniform
panel suited to your needs.
In an ideal world, researchers would be able to perform
multimodal sequencing, wherein entire genomes and
transcriptomes are sequenced in depth to ensure that
no mutation goes undetected. However, the realities
of sequencing make this impractical: Despite lowering
costs, whole genome sequencing can still be prohibitively
expensive for many laboratories (to say nothing of the
challenges associated with data analysis and storage).
The same holds true for unbiased whole transcriptome
sequencing. Instead, researchers must often strike a
balance between sequencing depth (which affords
greater confidence in sequencing results), sequencing
breadth (which determines the number of potential
mutations that can be detected), and the number of
samples that can be sequenced (Figure 4).
Figure 4: Traditional next-generation sequencing involves balancing trade-offs among sequencing breadth, depth, and scale.
03 Target Enrichment: Capturing the
Malignant Genetic Landscape
Trade-Offs In Next-Generation Sequencing
LEARN MORE
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
7
Such a balance is often found through the use of target capture panels. These panels use synthetic DNA
probes that have been designed to bind with a limited pool of specific target DNA sequences. Isolating
the probe-bound fragments allows superfluous DNA to be washed away, reducing the total number of
fragments that need to be sequenced while ensuring that important regions remain covered [1].
This is important for cancer research because laboratorians are often working with samples containing
a complex mixture of genetic material, or samples in which tumor DNA is vanishingly rare. In both cases,
target enrichment makes it possible to focus sequencing resources sparingly, enabling deep and
sensitive sequencing of the genetic material that most interests researchers.
Liquid Biopsy Research
The potential for increased sequencing sensitivity is perhaps best emphasized by studies involving
circulating tumor DNA (ctDNA). Following curative treatment, ctDNA may be as rare as 0.01% among the
collection of cell-free DNA fragments captured by liquid biopsies [2]. Detecting these rare bits of DNA
may be invaluable for the early detection of recurrent tumors, a primary goal of minimal residual disease
testing. However, for this to become a reality, researchers need the ability to study ctDNA with greater
sensitivity, meaning they need access to tools that enable the enrichment of ctDNA.
Enriching For Sensitive ctDNA Detection
In a 2024 study published to Oncotarget by researchers at Personalis, a tumor-informed minimal residual disease assay (MRD) was
reported to detect ctDNA in quantities as low as 1.67 parts per million (PPM), with a limit of detection at 3.45 PPM [3]. This ultrasensitive assay was developed using custom target enrichment panels (synthesized by Twist) to capture fragments of DNA containing
patient-specific mutations (identified through the sequencing of previous tumor biopsies). The sensitivity of this assay stands out in
its field, as other such assays demonstrate a limit of detection that ranges from 80 to 2,500 PPM. This boost in sensitivity has the
potential to greatly improve the speed at which tumor recurrence is detected.
While powerful tools, tumor-informed MRD assays present certain challenges to researchers. First, each patient will require a
bespoke panel whose design is optimized for their unique tumor profile. Doing so requires expertise in probe design to ensure that key
variants are captured with high specificity. Additionally, the sensitivity of such an assay also depends on uniform probe manufacturing
to ensure that lowly represented variants are accounted for. Doing all of this at scale and on a fast timeline can be a significant
challenge, which is why Twist’s team of experts are here to help. Twist’s silicon-based DNA synthesis platform can then be used to
manufacture these custom, often complex, panels at scale with high precision and accuracy.
CASE STUDY 1
Read the primary paper here.
TWIST BIOSCIENCE
8
Methylation Detection
A unique challenge facing some researchers is the need to carry out genome and epigenome
sequencing in tandem. Capturing information about the methylome, for example, can provide valuable
information about a ctDNA fragment’s tissue of origin. However, most current methods for methylation
sequencing must alter the target DNA sequence prior to target capture, converting unmethylated
cytosine bases into uracil (and ultimately thymine). This greatly reduces the unique complexity of DNA
fragments and can be challenging to anticipate when designing capture probes. Designing target
capture for methylation sequencing thus requires careful consideration and expertise.
Tapping the Epigenome for Non-Invasive Colorectal Cancer Detection Research
In 2023, researchers from the Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, reported a novel
approach to colorectal cancer detection (CRC), one that uses target-enriched methylation sequencing. Having identified CRC-specific
methylation patterns, the team of researchers designed a custom Twist target enrichment panel to enable the capture and assessment
of 149 cfDNA methylation markers in blood samples. With the Twist Custom Methylation Panel, the team was able to better detect CRC
relative to conventional blood tests, demonstrating an 85% sensitivity at a 90% specificity threshold. These results suggest that target
enrichment for methylation sequencing can be a valuable approach to sensitive CRC detection [4].
CASE STUDY 2
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
Read the primary paper here.
TWIST BIOSCIENCE
9
The Importance of Uniformity
Whether for ctDNA enrichment or methylation sequencing, the value of any target enrichment panel
depends on the quality of the synthetic DNA used to build it. If capture probes contain errors, they’re
less likely to bind to their target sequence, which leads to reduced overall coverage. Similarly, a nonuniform target capture panel may have under-represented capture probes. The targets of these rare
probes may be inadvertently filtered out during data analysis as they’re overwhelmed by the much
more numerous sequences targeted by highly-represented probes. In both cases, the end result may
be inefficient sequencing, incomplete coverage, and loss of data—data that may have revealed the
presence of a critical mutation. It is thus crucial to not only design panels well but to synthesize them
with high uniformity and accuracy.
To this end, Twist has developed several target capture panels and associated tools that aid in
cancer research:
Target Enrichment Panels
Twist Human
Comprehensive Exome Twist Exome 2.0 Twist Alliance Clinical
Research Exome
Twist MRD Rapid 500 Panel Twist Human Methylome Panel Twist Alliance Pan-cancer
Methylation Panel
Twist RNA Exome Twist Alliance CeGaT RNA
Fusion Panel Kit Twist Custom Panels
Library Preparation Kits
Twist Library Preparation
Enzymatic Fragmentation Kit 2.0
Twist cfDNA
Library Preparation Kit
Twist NGS Methylation
Detection System
Twist RNA Library
Preparation Kit
Other
Twist cfDNA Pan-Cancer
Reference Standard v2 Twist UMI Adapter System Twist Methylated UMI Adapters
TWIST BIOSCIENCE
10
Over the last decade, CRISPR/Cas has become an invaluable tool in molecular biology by enabling the
precise editing of genomic DNA. Thanks to ongoing innovation, the different types of edits that can be
made grow ever more diverse, from gene knockouts to epigenetic repression, single-base editing, gene
activation, and combinatorial perturbation [5]. Together, the breadth of the CRISPR/Cas toolkit is such that
researchers now have the ability to engineer advanced disease models, methodically interrogate gene
function, and validate potential therapeutic strategies—all of which are proving advantageous for the
study of cancer [6].
Improving Research Models
Cancer research benefits greatly from advances in CRISPR/Cas. Preclinical cancer modeling often
involves the use of immortalized cell lines, patient-derived cells, or animal models, each of which has
significant shortcomings. CRISPR/Cas is enabling the construction of improved models by allowing
researchers to modify healthy cells with mutations that are suspected factors in cancer development.
For example, Caeser et al. used CRISPR/Cas engineering to build a model of early-stage diffuse large
B cell lymphoma (DLBCL) [7]. Several complex genetic variants had been identified in DLBCL, but little
was known about how these variants contribute to disease development and progression. To change
this, Caeser et al. performed a functional genomic screen in primary human germinal center B cells
(the cell-type in which DLBCL is thought to originate). The screen proved highly informative, identifying
cooperating genetic alterations that produced malignant cellular behavior. With this data, the team was
able to fully transform healthy B cells into synthetically engineered DLBCL models, thereby creating a
highly relevant model system for researchers to use while studying this disease.
Identifying Therapeutic Targets
Functional genomic screening can similarly be used to identify potential therapeutic targets (Figure 5).
Behan et al., for example, performed a genome-scale CRISPR screen (targeting >18,000 genes) using
324 human cell lines that represented 30 different cancer types [8]. Their goal was to identify
high-priority targets worthy of therapeutic development. To perform this screen, they relied on gRNAs
generated from a Twist-synthesized oligo pool. The screen was more than successful, identifying 628
unique priority targets, including 92 genes that appeared to be valuable therapeutic targets across
a wide range of different cancer types.
04 CRISPR:
Tinkering With the Cancer Genome
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
11
Greater Scale and Dimensionality in CRISPR Screening
Recent advances have enabled researchers to begin CRISPR screening with high-dimensional phenotypic readouts. Rather than
simply measuring cell survival in response to gene perturbation, methods like direct capture perturb-seq (dcPerturb-seq) can be
used to assess complex phenotypes (such as alternative splicing dynamics, gene network expression changes, and more) with
single-cell resolution.
This approach not only makes CRISPR screening more informative but also more efficient, as fewer cells are needed for robust results.
For example, Replogle et al. leveraged dcPerturb-seq in a series of large-scale CRISPR screens [9–11]. In one such study, transcriptionbased phenotypes in over 2.5 million cells across nearly 10,000 genes were analyzed [10]. Their findings revealed new gene functions,
insights into aneuploidy, and unexpected genetic regulation of mitochondrial genes, all achieved at lower costs and higher throughput.
Figure 5: Synthetic DNA is a powerful tool for understanding the role of specific genes within human disease through screening,
target analysis, and rescue experiments.
CASE STUDY 3
Learn more about the benefits of dcPertrub-seq from Replogle in this webinar, Scalable and combinatorial
single-cell CRISPR screens by direct guide RNA capture and targeted seq.
TWIST BIOSCIENCE
12
Uniformity in Genetic Engineering
These examples represent just a few of the many different ways that CRISPR/Cas can be used
to advance our understanding of human disease. But, as with NGS, the value of any CRISPR/Cas
engineering study is highly dependent on the quality of the CRISPR components used. CRISPR
engineering requires the use of a synthetic RNA to guide the Cas enzyme’s editing, leading it to specific
genomic sequences. For most applications, this RNA is known simply as a guide RNA (gRNA). Here
again, we see a repeated theme: The accuracy and uniformity of gRNA synthesis have a direct influence
on the quality of CRISPR editing.
Mutations in synthesized oligos translate into mutations in the gRNA, which can impede nuclease activity,
cause off-target editing, and reduce on-target editing.
Non-uniform gRNA synthesis is particularly important for large-scale screening (Figure 7)
(as was done by Behan et al.). In these screens, 10s to 100s of thousands of gRNAs are needed to
systematically perturb every gene of interest. If guides are non-uniformly synthesized, it can result in a
substantial increase in experimental costs. This is because researchers will need to increase the number
of cells screened to ensure that rare gRNAs are sufficiently represented in the final dataset. Costs mount
as the number of cells needed increases—which can present practical challenges as well if using primary
or difficult-to-culture cells—and as researchers compensate for underrepresented gRNAs by increasing
the number of sequencing rounds. Therefore, improving the accuracy and uniformity of gRNA synthesis
can substantially affect the accuracy, efficiency, and cost of CRISPR screening.
To this end, Twist offers high-quality oligonucleotide synthesis to support CRISPR/Cas studies,
synthesizing gRNAs with supreme accuracy and uniformity (Figure 6).
Not all oligo pools are created equal—uniform libraries lead to more leads.
Learn more about the importance of uniformity in this white paper.
Figure 6: Twist helps teams efficiently conduct CRISPR screens with oligo sequences.
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
13
Figure 7: Example workflow of sgRNA synthesis to conduct a functional CRISPR screen and identify novel targets.
Have confidence in your CRISPR screen with Twist
Scalability
generate gRNA
libraries of any size
Reliability
highly uniform
oligo pools
Precision
maximum sequence
representation
TWIST BIOSCIENCE
14
Advances in protein engineering and synthetic biology have given rise to new and powerful classes
of anti-cancer therapeutics. Novel drug targets identified with NGS and validated through CRISPR
screening can now be targeted by a wide armamentarium of therapeutics, from antibodies to T cells
carrying chimeric antigen receptors (CARs).
Therapeutic Antibody Development
The therapeutic value of antibodies has long been recognized. However, it is only in recent years that
researchers have gained the ability to approach antibody discovery and development with the precision,
rigor, and scale that’s typical of small molecule research.
Nonetheless, the challenge before antibody developers is complex. These multimeric proteins
typically consist of two heavy-chain and two light-chain peptides, each containing multiple distinct
domains. Variations in amino acid composition, domain arrangement, and peptide combinations can
all affect antibody pharmacology in diverse ways [12–14]. Therefore, the discovery and development
process involves testing billions of antibody variants for desired properties, most often with variation
concentrated in the hyper-variable complementarity determining regions (CDRs), which play a critical
role in antigen recognition and affinity.
Practical Limitations of Antibody Variant Testing
Consider a typical antibody which has approximately 60 CDR amino acids that are critical for target binding. To home in on the most
clinically valuable combination of amino acids, a comprehensive approach would aim to test every potential amino acid combination
in each CDR. Doing so would require synthesizing and testing more unique sequences than physically possible. Instead, most
antibody libraries max out somewhere between 1010 and 1012 unique sequences. Researchers must therefore design their antibody
libraries carefully in order to focus their efforts, sampling specific portions of the sequence space in search of an optimal antibody
sequence—one that can itself be used to build and iterate towards a truly optimal antibody.
CASE STUDY 4
05 Antibodies and CARs:
Synthesizing a New Form of Hope
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
Read the primary paper here.
TWIST BIOSCIENCE
15
Developers can approach this challenge using hybridoma, B-cell screening, or display technologies
such as phage and yeast.
In vivo platforms such as hybridoma and B-cell screening leverage the natural antibody diversity found
in living organisms. The human body, for example, is believed to have at least 107
unique antibody
sequences in circulation at any given time [15, 16]. Antigen-binding immune cells will be isolated and,
ultimately, their immunoglobulin domains sequenced using NGS technology. The resulting dataset will
represent an enriched catalog of antibody variants that bind to the target antigen.
Twist’s in vitro phage and yeast display platforms leverage synthetic library technology. Library
designs may be inspired by natural repertoires, existing datasets (such as those produced by in vivo
immunizations), or in silico modeling data. In any case, genes encoding putative antigen binding
domains are synthesized and integrated into a phagemid, enabling antibody expression on the viral
surface. Subsequent screening can isolate antigen-binding phage, whose unique antibody sequence is
determined through NGS.
DNA synthesis technology is integral to antibody discovery and optimization, regardless of the initial
technology used. For phage or yeast display, synthetic DNA enables researchers to design vast libraries
of antibody variants for high-throughput screening, typically on the order of 1010 variants. Candidates
identified with each technology will often require optimization, wherein individual and combinatorial
variants are synthesized and screened for improved pharmacological and pharmacokinetic properties.
Those antibodies identified through non-humanized in vivo workflows or, in particular, through the use
of in vitro technologies, may need further optimization to reduce immunogenicity (a process known
as humanization).
To this end, Twist has developed an extensive suite of tools to support researchers in their study of
antibody development and discovery (Figure 8).
Figure 8: Twist has a robust antibody discovery and optimization pipeline.
IN VIVO DISCOVERY
Hyperimmune DiversimAbTM Mouse
License-Free Humanized Mice
VHH Alpaca Immunization
and more!
2-3 MONTHS
IN VITRO DISCOVERY
Fully Human Library of Libraries
Custom Library Generation
Immune Libraries
Phage and Yeast Display
3-4 MONTHS
SCREEN & EXPRESS
Twist Powered HT Antibody Production
and Characterization
OPTIMIZATION
Humanization, Developability Screening
ANTIGEN PRODUCTION
AI/ML Informed
Design and Lead
Selection
TWIST BIOSCIENCE
16
Biopharma Solutions: Among Twist’s many offerings is a solution for antibody discovery that includes the use of hybridoma, single
B-cell screening, phage-display, and machine learning technologies to enable rapid and effective screening for promising candidates.
High-Throughput Antibody Production and Characterization: Twist Bioscience’s High-Throughput Antibody Production is a geneto-protein workflow capable of reformatting tens to thousands of diverse antibody sequences as full-length IgGs and generating
screening-scale amounts of recombinant antibody material for validation and testing. With the ability to precisely write thousands of
genes per run, Twist’s Express Antibodies enable immediate expansion of your antibody production pipeline. To complete the maketest cycle in any antibody development workflow, explore our portfolio of characterization and developability assays that can be
complemented to your protein production projects. Simply upload the antibody sequences you need and let Twist do the rest.
Library of Libraries: Twist’s pre-constructed, fully human, and validated phage display libraries are built using precision DNA writing
technology, allowing the removal of motifs that can lead to downstream manufacturing liabilities, thus enriching the libraries for
theoretically interesting antibody sequences devoid of conventional sequence-based developability liabilities. Twist Biopharma
Solutions has designed multiple general, naïve, and target class-focused libraries with diversities ~1010, giving you a multifaceted
approach to your next discovery project. Select from libraries containing naïve human diversity, rationally designed with antibody
structural considerations, or with diversity computationally tailored to high-value targets, including carbohydrates, ion channels, and
GPCRs. They’re available in a variety of VHH, Fab, and scFv frameworks, including common light chain options for the generation of
bi- and multispecific therapeutic antibodies. These libraries can also be leveraged to develop monoclonal antibodies, multispecifics,
and chimeric antigen receptors (CARs) for CAR T-cell therapy.
Twist Antibody Optimization (TAO): Twist’s rapid, scalable DNA synthesis platform enables the rapid generation of defined synthetic
libraries. Twist converts your parental antibody to the closest germline sequence and comprehensively explores the human mutational
space with efficiency. Additionally, this system can be used for high-throughput conversion from scFv, Fab, or VHH formats to
full-length IgG or VHH-Fc format. Twist’s team of antibody experts leverage their collective 50 years of experience to streamline
development with biochemical, biophysical, and functional screening of any TAO-derived antibodies.
Twist’s Antibody Solutions
Hybridoma Workflow (3–5 months to sequence)
3–5 WEEKS
(OPTIONAL) 3–8 WEEKS 4–6 WEEKS 1–3 WEEKS 2–3 WEEKS 5–7 WEEKS
Antigen & Screening
Tool Production Mouse Immunization Hybridoma Fusions &
High-Throughput Screening Advanced Screening Hybridoma
Sequencing
Recombinant Antibody
Expression (+ downstream
engineering/humanization)
B Cell Screening (2–5 months to sequence)
3–5 WEEKS
(OPTIONAL) 3–16 WEEKS 1 DAY 2-3 WEEKS 5–8 WEEKS
Antigen & Screening
Tool Production
Mouse, Rabbit, or
Alpaca Immunization
Beacon-Based
B Cell Screening B Cell Sequencing Recombinant Antibody Expression
(+downstream engineering/humanization)
Library of Libraries (2–5 months to sequence)
3–5 WEEKS
(OPTIONAL) 2–4 WEEKS 2–3 WEEKS 5–8 WEEKS
Antigen & Screening
Tool Production
Phage & Yeast Display Panning
(cell- and bead-based selections)
Antibody Sequencing
(Sanger & NGS)
Recombinant Antibody Expression
(+ downstream engineering)
Immune Libraries (2–5 months to sequence)
3–5 WEEKS
(OPTIONAL) 3–16 WEEKS 4–6 WEEKS 2–4 WEEKS 2–3 WEEKS 5–8 WEEKS
Antigen &
Screening Tool
Production
Mouse, Rabbit,
or Alpaca
Immunization
Immune Library
Construction
Phage & Yeast Display Panning
(cell- and bead-based selections)
Antibody Sequencing
(Sanger & NGS)
Recombinant Antibody Expression
(+ downstream engineering)
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
17
Cell-Based Therapy Development
A fast-growing sector of cancer research concerns the development of cellular therapies. These living
therapeutics work by infusing an organism with enhanced immune cells, whether those are stem cells
lacking pathogenic mutations or, in the case of CAR T-cells, differentiated cells with specific and potent
pathogen-targeting abilities [17, 18].
Whereas therapeutic antibodies and small molecules are ephemeral in the body, cell therapies
developed by Twist clients have the potential to help patients by providing lasting protection against
recurrent malignancies.
Among the many different cell therapies, CAR T-cell technology has seen prominent success against
intractable cancers. Researchers looking to develop CAR T-cell therapies have many challenges
ahead of them, but these can be made easier if they have access to high-quality oligonucleotide
synthesis platforms.
Tools For Developing CARs
T cells are a type of lymphocyte whose activation
is not dependent on antibodies. Instead, T
cells rely on a unique cell surface protein—the
T-cell receptor (TCR)—which recognizes major
histocompatibility complex (MHC) proteins
displaying antigenic (Figure 9) peptides. Once
bound, T cells can direct adaptive immune
cells against specific pathogens and thus have
significant potential in therapeutic applications [19,
20]. By imbuing T cells with a form of synthetic,
modified TCR (known as a chimeric antigen
receptor, or CAR), researchers can target T cells
against cancer cells.
The development of safe and effective CARs
requires careful and extensive optimization to
ensure safety, specificity, and efficacy. Fortunately,
many of the same technologies used for antibody
development can be useful. Antibody phage
display systems, for example, enable large-scale
and high high-throughput screening of potential
antigen binding domains. Once identified as
specific for the target antigen, DNA coding
for binding domains can be synthesized and
assembled into a CAR gene.
Figure 9: Structure of a CAR T-cell.
TWIST BIOSCIENCE
18
As with antibodies, producing a clinically viable CAR requires extensive optimization due in part to the
highly context-dependent nature of CAR signaling. Both CAR-antigen binding kinetics and the resulting
cellular response can be significantly altered by transmembrane and intracellular CAR components.
Therefore, researchers will need the ability to test variations in antigen binding domains, hinge and
transmembrane domains, and costimulatory factor combinations. This will necessarily involve the
generation and expression of combinatorial variant CAR libraries using large-scale DNA synthesis [21, 22].
CAR and antibody development is just the tip of the biologics iceberg. There are many similar forms
of protein-based therapeutics, the development of which is aided by the use of combinatorial variant
libraries and optimization of antibody-like proteins. This includes TCR engineering, antibody-drug
conjugates, and CAR-NK cells, among many others. In each application, the same basic principles hold
true: Discovery and optimization of promising therapeutics are made easier and more efficient when
researchers have access to high-quality oligonucleotide synthesis platforms.
Cell Development Libraries
Combinatorial Variant Libraries: Twist’s massively parallel silicon-based DNA synthesis platform produces highly uniform and accurate
oligos, with 90% of oligos represented within a <2.5x relative abundance range. Using this platform, Twist can fabricate highly diverse
gene mutant libraries with excellent variant representation and highly specific user-defined composition with no unwanted bias or
motifs. Twist library technology enables a comprehensive interrogation of the variant sequence space.
Site Saturation Variant Libraries: Protein engineering screens using single-site variant libraries allow researchers to explore a
protein’s sequence space and investigate the relationship between sequence and protein structure and function. Twist Site Saturation
Variant Library construction leverages massively parallel oligonucleotide synthesis using Twist’s proprietary silicon-based DNA
synthesis platform. These libraries offer researchers complete control over codon usage (all 64 codons available) and allows for
screening of 1 to 20 different amino acids at each position.
Spread Out Low Diversity Libraries: Spread-Out Low Diversity (SOLD) libraries provide a quick and precise tool for mapping protein
sequences to explore the complex relationship between a protein and its environment. SOLD Libraries are a time- and money-saving
tool for researchers who want to efficiently investigate combinatorial possibilities. A library of precise combinatorial variants with
uniform amino acid distribution or ratio, balanced codon usage, and the ability to avoid unwanted restriction sites provides the ultimate
tool to explore the variant space. All SOLD Libraries are NGS-verified, cloning-ready, and created using Twist’s patented silicon-based
synthesis platform, ensuring low error rates.
TCR Libraries: Engineered T-Cell receptor (TCR) therapy is a type of cell therapy that leverages engineered T-cell receptors to target
tumor-specific antigens. TCR repertoire sequencing can be done via single cell sequencing or bulk sequencing. Each has its own
advantages. Bulk sequencing enables you to sample more of the sequence space, but information about the alpha-beta TCR pairing is
lost. Single-cell sequencing enables you to capture information on the alpha-beta chain pairing and receptor composition. However,
single-cell sequencing has a much lower throughput than bulk sequencing. Twist offers TCR libraries that complement both strategies:
• A paired pooled TCR library enables you to retain the original alpha-beta chain pairing to maximize hit validation and replicate
the TCR repertoire. This is a good complement to single-cell sequencing, where the alpha-beta TCR pairing is known.
• A combinatorial TCR library enables you to shuffle alpha and beta chain pairs to create additional diversity and explore novel
combinations beyond the identified repertoire. This is a good complement to bulk sequencing, where the alpha-beta TCR pairing
is unknown.
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
19
Despite decades of research, cancer remains
an enigmatic disease. Fortunately, advances
in recent decades have helped to demystify
the nature of malignant cells. From among
these critical advances, the rise of DNA
synthesis platforms stands out as particularly
transformative. Indeed, custom oligonucleotide
synthesis is foundational to many of the
technologies and discoveries that define modern
molecular biology.
NGS technology, for example, has blown the
doors open on the malignant genetic landscape,
highlighting the presence of cancer-defining
mutations and emphasizing the vast scale of
human genetic variation. The consequences of
this variation can be meticulously studied, and
reproduced in advanced disease models, thanks
to the evolution of CRISPR/Cas gene editing
technology. Not only can the validation of novel
targets inspire therapeutic development, but
CRISPR/Cas enables the engineering of entirely
novel therapeutic modalities, such as T cells
armed with synthetic CARs.
06 DNA Synthesis Across
the Cancer Research Continuum
Each of these applications relies on the use of DNA synthesis technology. Whether it’s RNA sequencing,
DNA sequencing, or methylation sequencing, researchers using NGS to study cancer genetics benefit
greatly from targeted sequencing panels, which allow for limited sequencing resources to be focused
where they’re needed most. Modern CRISPR/Cas editing is made possible through the use of synthetic
gRNAs that dictate where editing should occur. Finally, the development of CARs and other biologics
requires the synthesis of the protein’s component parts and the large-scale, iterative process of
designing, building, and testing different peptide combinations.
Synthetic DNA Across the Cancer
Research Continuum
Accurate, scalable DNA synthesis is essential for
developing targeted sequencing panels, synthetic
gRNAs, and biologics.
NGS reveals cancer-driving mutations and
human genetic variation, while CRISPR/Cas
allows for precise gene editing and novel
therapies, such as CAR T-cells.
DNA synthesis platforms are critical to advances
in cancer research, enabling technologies like
NGS and CRISPR/Cas.
Twist’s platform provides uniform,
sequence-perfect DNA synthesis to support
cancer research, from target capture to
antibody development.
TWIST BIOSCIENCE
20
Whether you have a question about sales, customer support, media,
or investor relations, our team is ready to answer all your questions. CONTACT US
Stated simply, having access to accurate, uniform, and scalable DNA synthesis is proving invaluable
across the cancer research continuum. This is why Twist has developed a sprawling catalog of products
to support this important work, from custom target capture panels for the study of minimal residual
disease testing to antibody development services. Twist’s sequence-perfect, highly uniform, and
scalable DNA synthesis platform empowers scientists to overcome the challenges of cancer research,
ultimately with the hope of seeing a future where cancer treatment is trivial.
This product is for research use only. This product is not intended for the diagnosis, prevention, or treatment of a disease or condition.
TWIST BIOSCIENCE
21
1. Burcu Yaldiz, et al. “Twist Exome Capture Allows for Lower Average Sequence Coverage in Clinical Exome Sequencing.”
Human Genomics, vol. 17, no. 1, 3 May 2023, https://doi.org/10.1186/s40246-023-00485-5.
2. Fiala, Clare, and Eleftherios P. Diamandis. “Utility of Circulating Tumor DNA in Cancer Diagnostics with Emphasis on Early
Detection.” BMC Medicine, vol. 16, no. 1, 2 Oct. 2018, https://doi.org/10.1186/s12916-018-1157-9.
3. Northcott, Josette, et al. “Analytical Validation of next Personal ® , an Ultra-Sensitive Personalized Circulating Tumor DNA
Assay.” Oncotarget, vol. 15, 6 Jan. 2024, pp. 200–218, https://doi.org/10.18632/oncotarget.28565.
4. Zhao, Fen, et al. “Efficacy of Cell-Free DNA Methylation-Based Blood Test for Colorectal Cancer Screening in HighRisk Population: A Prospective Cohort Study.” Molecular Cancer, vol. 22, no. 1, 28 Sept. 2023, https://doi.org/10.1186/
s12943023-01866-z.
5. Meaker, Grace A, et al. “Advances in Engineering CRISPR-Cas9 as a Molecular Swiss Army Knife.” Synthetic Biology, 24
Oct. 2020, https://doi.org/10.1093/synbio/ysaa021.
6. Chehelgerdi, Mohammad, et al. “Comprehensive Review of CRISPR-Based Gene Editing: Mechanisms, Challenges, and
Applications in Cancer Therapy.” Molecular Cancer, vol. 23, no. 1, 9 Jan. 2024, https://doi.org/10.1186/s12943-023-01925-5.
7. Caeser, Rebecca, et al. “Genetic Modification of Primary Human B Cells to Model High-Grade Lymphoma.” Nature
Communications, vol. 10, 4 Oct. 2019, https://doi.org/10.1038/s41467-01912494–x.
8. Behan, Fiona M., et al. “Prioritization of Cancer Therapeutic Targets Using CRISPR–Cas9 Screens.” Nature, vol. 568, no.
7753, Apr. 2019, pp. 511–516, https://doi.org/10.1038/s41586-019-1103-9.
9. Nadig, Ajay, et al. “Transcriptome-Wide Characterization of Genetic Perturbations.” BioRxiv (Cold Spring Harbor
Laboratory), 3 July 2024, https://doi.org/10.1101/2024.07.03.601903.
10. Replogle, Joseph M., et al. “Mapping Information-Rich Genotype-Phenotype Landscapes with Genome-Scale PerturbSeq.”
Cell, June 2022, https://doi.org/10.1016/j.cell.2022.05.013.
11. Replogle, Joseph M., et al. “Combinatorial Single-Cell CRISPR Screens by Direct Guide RNA Capture and Targeted
Sequencing.” Nature Biotechnology, vol. 38, no. 8, 30 Mar. 2020, pp. 954–961, https://doi.org/10.1038/s41587-020-0470-y.
12. Lu, Ruei-Min, et al. “Development of Therapeutic Antibodies for the Treatment of Diseases.” Journal of Biomedical
Science, vol. 27, no. 1, 2 Jan. 2020, pp. 1–30, https://doi.org/10.1186/s12929-019-0592-z.
13. Ryman, Josiah T., and Bernd Meibohm. “Pharmacokinetics of Monoclonal Antibodies.” CPT: Pharmacometrics & Systems
Pharmacology, vol. 6, no. 9, 29 July 2017, pp. 576–588, https://doi.org/10.1002/psp4.12224.
14. Janeway, Charles. Immunobiology: The Immune System in Health and Disease. 5th ed., London, Harcourt Brace &
Company, 2001.
15. Rees, Anthony R. “Understanding the Human Antibody Repertoire.” MAbs, vol. 12, no. 1, 1 Jan. 2020, p. 1729683, https://
doi.org/10.1080/19420862.2020.1729683.
16. Pedrioli, Alessandro, and Annette Oxenius. “Single B Cell Technologies for Monoclonal Antibody Discovery.” Trends in
Immunology, vol. 42, no. 12, 1 Dec. 2021, pp. 1143–1158, https://doi.org/10.1016/j.it.2021.10.008.
17. June, Carl H., et al. “CAR T Cell Immunotherapy for Human Cancer.” Science, vol. 359, no. 6382, 22 Mar. 2018, pp.
13611365, https://doi.org/10.1126/science.aar6711.
07 References
TWIST BIOSCIENCE
22
18. Finck, Amanda V., et al. “Engineered Cellular Immunotherapies in Cancer and Beyond.” Nature Medicine, vol. 28, no. 4, 1
Apr. 2022, pp. 678–689, https://doi.org/10.1038/s41591-022-01765-8.
19. Yisong, Y Wan, and Richard A. Flavell. “How Diverse—CD4 Effector T Cells and their Functions.” Journal of Molecular Cell
Biology, vol. 1, no. 1, 28 May 2009., pp. 20–36, doi:10.1093/jmcb/mjp001
20. Pennock, Nathan D., et al. “T Cell Responses: Naïve to Memory and Everything in Between.” Advances in Physiology
Education, vol. 37, no. 4, Dec. 2013, pp. 273–283, https://doi.org/10.1152/advan.00066.2013.
21. Rafiq, Sarwish, et al. “Engineering Strategies to Overcome the Current Roadblocks in CAR T Cell Therapy.” Nature
Reviews. Clinical Oncology, vol. 17, no. 3, 2020, pp. 147–167, https://doi.org/10.1038/s41571-019-0297-y.
22. Stock, Sophia, et al. “Chimeric Antigen Receptor T Cells Engineered to Recognize the P329G-Mutated Fc Part of EffectorSilenced Tumor Antigen-Targeting Human IgG1 Antibodies Enable Modular Targeting of Solid Tumors.” Journal for
ImmunoTherapy of Cancer, vol. 10, no. 7, 1 July 2022, p. e005054, https://doi.org/10.1136/jitc-2022-005054.
22
TWIST BIOSCIENCE
23
TWISTBIOSCIENCE.COM
© 2025 Twist Bioscience. All rights reserved. DOC-4009 REV1.0
Twist products are for research use only. They are not intended for the diagnosis, prevention, or treatment of a disease or condition. Twist Bioscience
assumes no liability regarding use of our products for applications in which they are not intended. The results presented are customer-specific and should
not be interpreted as indicative of performance across all institutions.
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