Redefining the Impossible With Genomics
eBook
Published: July 4, 2025

Credit: illumina
Genome sequencing isn’t just faster and cheaper than ever before, high-throughput platforms are now delivering insights at a scale and depth unimaginable just a few years ago.
From mapping single-cell dynamics to decoding complex disorders, researchers are no longer constrained by resolution, cost or data volume. However, scaling experiments, integrating multiomic layers and navigating vast datasets are still major challenges.
This eBook explores how researchers are dismantling these barriers by harnessing deeper sequencing, richer data and smarter workflows to unlock breakthroughs across population health, cancer biology and molecular diagnostics.
Download this eBook to explore:
- How high-resolution sequencing supports population-scale and rare variant research
- Benefits of integrating genomics, epigenetics and proteomics
- Strategies to overcome common roadblocks in study depth, scale and complexity
For Research Use Only. Not for use in diagnostics procedures.
Unlock the next wave
of genomic discovery
Broader, deeper, multiomic sequencing
enabled on the NovaSeq ™ X Series
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
2 | M-GL-01796 v1.0
3 Revolutionizing the pace of discovery
4 The dawn of the Genome Era
5 Broader, deeper sequencing methods
6 Bigger studies to improve statistical power
6 Large-cohort data at work
7 Increasing diversity in genomic data
8 Broader studies to access missed information
9 Value of whole genomes
10 Value of whole transcriptomes
11 Multiomic studies for a wider perspective
12 Common multiomic combinations
13 Next-level multiomics
14 Higher analytical resolution to decode complex systems
14 Single-cell sequencing
15 Spatial sequencing
15 Pushing the boundaries
16 Deeper studies to find rare genetic events
16 Liquid biopsy
17 Genome-wide tumor–normal sequencing
18 How the NovaSeq X Series accelerates genomic discovery
19 Enabling broader, deeper sequencing
21 Managing big experiments and big data
24 Example NovaSeq X Series workflows
26 Unimaginable experiments, made possible
27 Abbreviations
28 References
Table of contents
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
3 | M-GL-01796 v1.0
TABLE OF
CONTENTS BIGGER STUDIES
Revolutionizing the pace of discovery
Genomics research is expanding our understanding of biology and making personalized medicine a reality. Advancements
in next-generation sequencing (NGS) are enabling genomic visionaries to perform the studies that can answer the most
complex biological questions. Increased discovery power will come from larger studies with bigger cohorts, deeper
sequencing to identify rare genetic events, and broader sequencing methods and multiomics for a more comprehensive
view of cellular activity.
Still, this new scale of experiments has been held back by high sequencing costs and analytical complexity. Now, with the
NovaSeq X and NovaSeq X Plus Sequencing Systems, the extraordinary can become routine. Transformational throughput
and simplified informatics bring greater speed and lower costs that will be a force for democratizing sequencing and
powering the Genome Era.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
4 | M-GL-01796 v1.0
TABLE OF
CONTENTS BIGGER STUDIES
The dawn of the Genome Era
The first draft of the human genome sequence used hundreds of instruments performing capillary electrophoresis
Sanger sequencing and took over 13 years and nearly $3 billion (USD). Since the introduction of massively parallel
sequencing technologies like NGS, the throughput of genome sequencing has increased and the costs have decreased at
an incredible rate. Over the last two decades, sequencing output has grown over 10,000-fold, from less than 1 gigabase
(Gb) to 16 terabases (Tb) per run, while the number of reads has increased from millions to tens of billions. Experiments
that once required complex workflows now use simple push-button sequencing.
These incredible advancements in genome analysis are ushering in a new era of personalized medicine. Whole-genome
sequencing (WGS) has helped diagnose children with rare genetic diseases. Comprehensive genomic profiling of tumors
has been used to identify driver mutations and match them with targeted therapies. During the COVID-19 pandemic, DNA
sequencing tracked specific variants and provided the basis for rapid vaccine development. Managing future pandemics
and developing personalized therapies will depend on expanding access to high-throughput genomic sequencing.
COST-PER-GENOME
Dramatic decrease in sequencing costs
Genome Analyzer™ II
$3000/Gb
0.3 Gb/day
NovaSeq X Series
$2/Gb
6000 Gb/day
$2000 | 6 hours NY
What if this breakneck pace of advancement was
applied to air travel? A hypothetical flight from
New York City to Paris, France could decrease
Paris $1.33 | 1 Sec
2001
$100 M
$2 M
$100,000
$10,000
$1000
$600
$200
2003 2005 2007 2009 2011 2013 2015 2017 2019 2021 2023
from to
Cost (USD) per human genome
Year
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
5 | M-GL-01796 v1.0
TABLE OF
CONTENTS BIGGER STUDIES
Broader, deeper
sequencing methods
Despite this progress, there is much work left to do
to unlock the full power of the genome. To understand
genetic variation and complex disease, we will need to
sequence millions of genomes and integrate health data.
We need to represent diverse populations, ensuring that
more people can access sequencing in more places. To
diagnose rare disease in the neonatal intensive care unit
(NICU), we need to sequence genomes at record speed
for maximum impact. To use cell-free DNA to track
disease noninvasively, we need to sequence deeply. To
see the full picture of cellular physiology, we need to look
at multiple “omes” together—genomes, transcriptomes,
epigenomes, and proteomes. And we will need powerful
bioinformatics tools to turn all that data into insight.
With the decreased costs of sequencing, scientists
can leverage budgets to perform studies of greater scale
and depth, as well as new applications. Researchers
can generate more detailed data from their samples or
interrogate their samples at multiple levels for greater
insights. This eBook highlights the types of studies that
once were unimaginable, and are now possible thanks
to breakthrough advancements in high-throughput
genomics.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
6 | M-GL-01796 v1.0
TABLE OF
CONTENTS
BROADER
STUDIES
PACE OF
DISCOVERY
“A measure of the success
of our mission will be
whether we can convince
similar healthcare
systems around the world
to adopt approaches
developed within the
100,000 Genomes Project
to transform the application
of genomic medicine in
healthcare and bring
better outcomes to
patients worldwide.
Mark Caulfield, Chief Scientist,
Genomics England ”
Bigger studies to improve
statistical power
Increase statistical power from larger sample sizes
and more diverse cohorts
Even decades after the first human genomes were
sequenced, a majority of gene–disease connections
remain a mystery. To understand the significance of
human genetic variation in relation to complex disease,
genome-wide association studies (GWAS) aim to
connect genotype to phenotype. Most GWAS have
been performed using microarray genotyping. As the
cost of genome sequencing drops, researchers can
generate higher quality, more comprehensive sequence
data at population scale. A shared global knowledge base
that integrates genomic data with longitudinal health
records is needed to clearly identify key genes and
disease mechanisms.
Large-cohort data at work
The 100,000 Genomes Project in the United Kingdom
(UK), supported by Genomics England, is the archetype
for integrating genomics with a large-scale health system
and demonstrates what routine, genomic-informed
medicine might look like.1–3 The UK BioBank has also built
one of the largest biomedical databases in the world,
containing genetic, lifestyle, and health information
from 500,000 individuals.4 The population-scale WGS
data coming from these projects has yielded incredible
insights for cancer and increased diagnostic yield for
rare disease.3 For example, WGS of over 12,000 matched
tumor–normal samples revealed mutational signatures
that could help direct personalized cancer treatments.5
The UK is undertaking an even larger initiative, Our
Future Health, which aims to sequence five million more
genomes.6
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
7 | M-GL-01796 v1.0
TABLE OF
CONTENTS
BROADER
STUDIES
PACE OF
DISCOVERIES
Population genomics projects across the globe with numbers of participants
United Kingdom1
100,000
France13
1 million
Denmark16
50,000
Finland17
500,000
Estonia18
100,000
Turkey19
100,000
Israel22
100,000
Africa7
118,000+
Saudi Arabia21
100,000
United States29
1 million
United States31
1 million
Australia8
200,000
China26
100,000
China25
100,000
Japan23
100,000
Asia10,11
100,000
Hong Kong24
50,000
Mexico27
1 million Brazil28
7000
United States30
1 million
United States32
100,000
United Arab
Emirates20
1 million
United Kingdom6
5 million
Ireland14
400,000
Australia9
7000
Singapore12
100,000
Iceland15
200,000
United Kingdom4
500,000
Many of these efforts focus on increasing the diversity
in the pools of genome data to represent underserved
groups,7–12,19–27,31 because, as of 2021, 86% of genomics
studies have been conducted on individuals of European
descent.33 Additional tools are helping draw out the full
value of these large-cohort projects. The “pan genome”
reference genome includes 47 diverse references to
uplevel variant calling.34 The Primate AI-3D resource
mines data from other primate genomes (233 species)
to train artificial intelligence to use information from
natural selection to better classify variants of unknown
significance (VUS).35,36 Together, these efforts will help
us realize a more complete view of the genome for all
populations and develop ways to stop disease outbreaks,
decrease early deaths due to sudden heart attacks, and
reduce the strain of chronic diseases like Alzheimer’s
and diabetes.
Learn more
Population genomics
Increasing diversity in genomic data
Increased statistical power for these studies will also come from larger, more diverse cohorts. Expanding access to
NGS across the globe, especially in regions with underrepresented populations, will increase the diversity of our genome
data sets. Following in the footsteps of the pioneering work in the UK, population genomics projects are growing in other
countries and regions around the world.7–32
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
8 | M-GL-01796 v1.0
TABLE OF
CONTENTS
MULTIOMIC
STUDIES
BIGGER
STUDIES
Broader studies to access missed information
See the whole breadth of variation with whole-genome or whole-transcriptome sequencing
Historically, when sequencing is expensive and data analysis is burdensome, many labs have chosen targeted
sequencing approaches and narrowed their study focus. With lower costs and streamlined informatics, it becomes
more practical to query more broadly—to shift from gene panels to exomes or from exomes to whole genomes—and
gather the most data possible from each sample. The scale of broader research minimizes bias from experiments
by extending analyses beyond a few preselected targets.
* Coverage requirements vary depending on use-case, such as rarity of factor being measured.
Whole-genome sequencing Whole-exome sequencing
Sequencing region
Entire genome
Coverage
Pros Cons
~30×*
Captures all DNA variants in
the least biased way
Higher per-sample cost,
more demanding analysis
Sequencing region
Protein-coding regions
(~2% of the genome)
Coverage
Pros Cons
~50×–100×*
Reliable detection of
coding variants
Manipulation of the sample,
misses 98% of the genome
Targeted sequencing
Sequencing region
Specific genes or regions
of interest
Coverage
Pros Cons
> 500×*
Resources focused on
specific areas of interest
Limited resolution
Beyond the exome
Exome: 2% Noncoding genome: 98%
Structural variation
Repeat expansions
Copy number variants
Mitochondrial variants
Noncoding SNVs and indels
Regulatory elements
In our view, for deciphering the molecular causes of any genetic disorder,
we need the variants of the entire genome—especially when various
disorders exhibit overlapping symptoms and consequences. “
Dr. Kamran Shazand, Director,
Shriners Genomics Institute ”
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
9 | M-GL-01796 v1.0
TABLE OF
CONTENTS
MULTIOMIC
STUDIES
BIGGER
STUDIES
Value of whole genomes
When NGS is more accessible, studies can cast a
“wider net” to survey more genes. For example, labs
that routinely do exome sequencing can now afford
to switch to WGS. WGS offers multiple advantages for
finding variants. WGS examines the entire genome and
has the capability to assess variants in both coding and
noncoding regions of the genome.37–43 Noncoding variants
are frequently key biomarkers for disease. Unlike other
methods, WGS robustly captures all common variant
types.37–39,44 WGS captures copy number variants (CNVs)
with greater resolution than chromosomal microarrays
(CMAs).37,43,45,46 WGS also captures some variants in
exomes with greater accuracy than whole-exome
sequencing (WES).2,3,37,44,46–49
† Variant detection may vary depending on the particular laboratory and performance limits of validated variant types. Detection of repeat expansions by PCR is typically limited
to single-gene analysis, compared to multigene capabilities of WGS. Improvements to SV callers and increased success with long-read whole-genome approaches suggest fully
capable SV insights from WGS. PCR, polymerase chain reaction; FISH, fluorescence in situ hybridization.
WGS provides the most comprehensive analysis of genomic variant types†
Single nucleotide
variants (SNVs)37
Insertion-deletions
(Indels)
Copy numbers variants
(CNVs)37,50
Repeat
expansions39,40,51,52
Structural variants
(SVs)38,48
Mitochondria37
Paralogs41
Sanger Targeted NGS PCR FISH Karyotype CMA WES WGS
Capable Limited capabilities
In the case of rare genetic disease research, many disease-causing variants identified with WGS would have been
missed by WES, including those caused by repeat expansions or mutations in noncoding regions.2,3,39,40,47,48,51–53 Because
WGS provides better coverage and higher yield across the genome, including GC-rich regions, the European Society
for Human Genetics (ESHG) guidelines recommend use of WGS, even if only the exome or specific genes are examined
bioinformatically.28
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
10 | M-GL-01796 v1.0
TABLE OF
CONTENTS
MULTIOMIC
STUDIES
BIGGER
STUDIES
Value of whole transcriptomes
Even whole genomes don’t tell the whole story. Certain variants, like gene fusions, may not be readily apparent in the
DNA sequence and can be overlooked by WGS.53 RNA sequencing (RNA-Seq) enables scientists to identify and confirm
the presence of novel gene fusions and alternate transcript isoforms. Because RNA is dynamic between cell types and
states, RNA-Seq also offers critical information about biological activity.
Whole-transcriptome sequencing (WTS), or total RNA-Seq, delivers a high-resolution, base-by-base view of coding
and multiple forms of noncoding RNA activity. This provides a comprehensive picture of gene expression across the
full transcriptome at a specific moment in time. With total RNA-Seq, the whole transcriptome—including both known
and unknown regions—is captured.54–56 The increased affordability of high-throughput NGS, paired with push-button
analysis tools to decipher richer data sets, make WTS accessible for more routine use.
RNA-Seq for cancer research
In cancer research, RNA-Seq is a critical tool for direct measurement of the functional consequences of mutations.
Despite the average cancer containing about 46 mutations, only five to eight are necessary for initiation.57 Genomic
profiling alone is insufficient to differentiate these driver mutations from passenger mutations, or those mutations that do
not influence cancer initiation or progression. Measurement of gene expression patterns and mutation consequences
using RNA-Seq enables large-scale, unbiased differentiation of factors crucial for cancer progression, resulting in
more thorough and accurate cancer modeling.58–62 Growing evidence demonstrates the value of combining wholegenome
and transcriptome sequencing (WGTS) to collect broader information from cancer samples.63
RNA-Seq for genetic disease research
RNA-Seq offers a complementary approach to GWAS for genetic disease research that increases diagnostic yield.64–66
Measuring expression abundance in specific tissues can reveal the functional impact of pathogenic mutations and identify
which genes mediate the genotype’s effect on phenotype.64–67 RNA-Seq can also validate computational predictions of
splicing and increase confidence in the reclassification of VUS.
Many studies have shown that GWAS risk variants co-localize with genes that regulate expression.67,68 These genes,
known as expression quantitative trait loci (eQTLs), suggest that regulation is an important molecular mechanism used
by GWAS risk variants, most of which lie in noncoding regions of the genome. Polygenic risk scores based on eQTLs are
helping scientists better understand complex phenotypes.69
Learn more
Whole-genome sequencing
Whole-transcriptome sequencing
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
11 | M-GL-01796 v1.0
TABLE OF
CONTENTS
HIGH-RESOLUTION
STUDIES
BROADER
STUDIES
Multiple layers of information connect genotype to phenotype
Combining DNA, epigenetics, RNA, protein, or other molecular
measurements into a full cellular readout provides researchers
with novel scientific insights that cannot be found from single
omic methods alone.
Multiomic studies for a wider perspective
Multidimensional insights with sequencing provide a single readout from multiple omes across DNA, RNA, and protein
Biology is multilayered and complex. The central dogma outlines the intertwined relationship between DNA, RNA, and
protein. Genetic variation at the DNA level can impact RNA expression or protein function in diverse and unpredictable
ways. Environmental factors can also alter regulatory pathways and cellular metabolism to affect biology and human health.
Multiomics provides a perspective to power discovery across multiple levels of biology. This biological analysis approach
combines genomic data with data from other modalities, measuring gene expression, gene activation, and protein levels
to enable a more comprehensive understanding of molecular changes contributing to normal development, cellular
response, and disease.
Bigger picture biology through multiomics
Multiomics goes beyond the genome to unlock deeper
biological insights. Using every piece of molecular data available
can accelerate biological discoveries and transform our
understanding of human health.
Through the combined lens of multiomics, researchers can witness the complicated interplay between the molecules of
life. Integrating these complementary metrics into multiomic data sets brings a more comprehensive picture of cellular
phenotypes and helps pull more high-quality information from each sample. The transformational power in the latest
sequencing systems makes it easier to query multiple omes in parallel.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
12 | M-GL-01796 v1.0
TABLE OF
CONTENTS
HIGH-RESOLUTION
STUDIES
BROADER
STUDIES
Common multiomic combinations
Genomics + transcriptomes
GWAS have successfully identified genetic variants
associated with complex diseases. The genotype offers
information on susceptibility to the disease; however,
determining the specific genes and pathways affected
by those variants is more difficult. Incorporating
RNA-Seq can help researchers annotate and prioritize
variants uncovered in GWAS for functional analysis to
understand mechanisms of disease. Gene expression
analysis informs if and when the genes of interest are
down- or upregulated in the disease samples. This
multiomic approach to functional genomics can help
power drug target identification and biomarker discovery.
Genomics + epigenetics
The majority of human variation identified by GWAS is in
the noncoding regions of the genome, including introns,
promoters, or enhancers. Comprehensive epigenetic
profiling can reveal patterns of gene regulation to help
find the function of those variants. NGS-based epigenetic
techniques include chromatin immunoprecipitation
(ChIP-Seq), assay for transposase-accessible chromatin
(ATAC-Seq), and chromosome conformation capture (HiC).
DNA methylation patterns are also conserved and can
represent a new class of biomarkers. Multiomic approaches
that combine methylation, or other epigenetic profiling,
with genetic information can connect functional layers to
decipher complex pathways and disease mechanisms.
Epigenetics + transcriptomics
Epigenetics and transcriptomics offer complementary
information to study the details of cellular differentiation
and response. Combining epigenetic and RNA-Seq
methods allows researchers to directly measure the
ties between gene regulation and gene expression,
instead of simply inferring those connections. Integrating
epigenetics and RNA-Seq can help researchers identify
candidate genes and understand the mechanisms
controlling interesting phenotypes. This holistic, nonbiased
multiomics approach can uncover new regulatory
elements for biomarkers and therapeutic targets.
Transcriptomics + proteomics
RNA-Seq offers unparalleled discovery power to
interrogate the transcriptome without prior knowledge.
Incorporating protein detection with RNA-Seq can tie
new discoveries back to known canonical markers
and historical clinical outcomes. Antibodies tagged
with oligonucleotide barcodes enable analysis of cell
surface proteins with results read by sequencing, which
scales to a much higher number of parameters than
flow cytometry or mass cytometry. Single-stranded
nucleotide aptamers with selective protein binding
targets can be used in a similar way. Methods like
cellular indexing of transcriptomes and epitopes by
sequencing (CITE-Seq) combine single-cell RNA-Seq
with cell surface protein analysis. Bulk epitope and
nucleic acid sequencing (BEN-Seq) is performed at the
bulk level. Spatial transcriptomics interrogates RNA and
proteins in context with tissue morphology.
Genomics + proteomics
This multiomic approach directly connects genotype to
phenotype for more informed research on disease and
therapeutics development. Linking genetic variation
to protein expression at the single-cell level can reveal
the functional impact of somatic mutations on human
cancers to better understand tumor evolution and
disease progression.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
13 | M-GL-01796 v1.0
TABLE OF
CONTENTS
HIGH-RESOLUTION
STUDIES
BROADER
STUDIES
Next-level multiomics
The multiomic methods highlighted here represent a
holistic approach to understanding biology. Additional
discovery power comes from studies that measure
three or more modalities.70,71 Combining WGS, RNA-Seq,
and methylation sequencing is improving diagnostic
yield for neurodevelopmental disorders and other rare
diseases.70,72 Multiomic studies have also focused on
complex diseases. For example, integrated genome,
epigenome, and transcriptome data from populationscale
data sets helped identify genes and biological
mechanisms associated with blood pressure regulation.71
As the cost of sequencing continues to decrease and
the technology advances, multiomic assays will become
more comprehensive and better integrated. Labs will be
able to study more samples under different conditions
to reveal the dynamic properties of cells and systems.
Sequencing will support proteomic studies with oligotagged
antibodies or aptamers for hundreds to thousands
of markers. More assays will incorporate multimodal
measurements at higher resolution.
One of the biggest challenges for multiomic research
is how to integrate different molecular data sets
in a standardized way. Researchers need robust
computational strategies to extract biologically
meaningful insights from these vast amounts of data.
Sophisticated bioinformatics tools enable normalization
and integration of multimodal single-cell sequencing
experiments. Machine learning methods will also help
organize and filter complex multiomic data.
Learn more
Multiomics
CITE-Seq
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
14 | M-GL-01796 v1.0
TABLE OF
CONTENTS
DEEPER
STUDIES
MULTIOMIC
STUDIES
Higher analytical resolution to
decode complex systems
Transition to high-resolution approaches, including single-cell sequencing
and spatial analysis, for insights into complex tissues
High-resolution methods like single-cell sequencing and spatial analysis offer
another layer of fundamental detail to examine heterogeneity in complex
cell populations.73 These approaches have enabled researchers to look at
cancer, development, and infectious disease at the single-cell level within tissue
context.74–82 Studies have combined single-cell and spatial RNA-Seq to map
the architecture of skin cancer,83 characterize human intestinal development,84
and track COVID-19 pathology.74,75
Single-cell sequencing
Single-cell sequencing is a popular approach used to characterize hundreds
to millions of individual cells from a tissue. This method reveals cellular
heterogeneity and provides a more comprehensive understanding of tissue
composition. Significant advances in the area of single-cell characterization
include technologies for cell isolation and new methods and applications
for single-cell sequencing. These advances have stimulated the launch of
accessible commercial solutions for every step of the single-cell sequencing
workflow, from tissue preparation through data analysis.
Single-cell RNA sequencing (scRNA-Seq) has become a powerful tool in
immunology, cancer, and developmental biology studies. As part of the Human
Cell Atlas, a large-scale effort to map human development, researchers used
single-cell combinatorial indexing to profile the transcriptomes of ~2 million
cells derived from 61 embryos staged between 9.5 and 13.5 days of gestation
in a single experiment.85 Cell atlas studies often have implications in genetic
disease research. For cystic fibrosis, scRNA-Seq of human bronchial epithelial
cells helped uncover a rare cell type, pulmonary ionocytes, that accounts for
the majority of CFTR expression in the lungs.86
Cancer researchers use scRNA-Seq to better understand cancer biology,
as traditional bulk RNA-Seq does not address the heterogeneity within
and between tumors. scRNA-Seq has aided the development of targeted
therapy and immunotherapy treatments. Researchers often use scRNA-Seq
in conjunction with cell-surface protein expression and immune repertoire
sequencing to characterize the inflammatory response.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
15 | M-GL-01796 v1.0
TABLE OF
CONTENTS
DEEPER
STUDIES
MULTIOMIC
STUDIES
Spatial sequencing
Typical NGS methods using dissociated samples can lose key spatial information present in vivo. Traditionally,
immunohistochemistry and in situ hybridization have been the tools of choice to reveal spatial gene expression in
tissue sections. But the throughput of these procedures is limited, analyzing only a few genes at a time. By combining
high-throughput imaging and sequencing technologies, spatial RNA-Seq provides a previously inaccessible view of
the full transcriptome in morphological context. Spatial RNA-Seq methods that retain the precise location of biological
molecules in tissue samples can further our understanding of mechanisms in health and disease.
Pushing the boundaries
As single-cell and spatial technologies advance,
researchers are increasing the scale, scope, and
dimensions of their experiments. One consortium, the
BRAIN Initiative Cell Census Network, coordinated
large-scale multiomic analyses from multiple labs of
the mammalian primary motor cortex with single-cell
and spatial resolution.87 Cross-modal computational
analysis integrated data with single-cell transcriptomes,
chromatin accessibility, DNA methylation, and
phenotypic properties, across species, to establish
a framework for neuron organization.87
A team of researchers at the Howard Hughes Medical
Institute performed genome-scale Perturb-Seq (a
CRISPR-based functional genomics screen with scRNASeq
readouts) across 2.5 million human cells.88 From the
single-cell transcriptional phenotypes, the team was
able to dissect complex cellular pathways and develop a
map of gene and cellular function.88 The researchers note
ways to increase the power of future studies, including
greater numbers of cells or sequencing depth, full-length
RNA-Seq or protein-level readouts, and sampling a wider
range of time points or cell types.88
Learn more
Single-cell RNA sequencing
Spatial transcriptomics
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
16 | M-GL-01796 v1.0
TABLE OF
CONTENTS
NOVASEQ X
SERIES
HIGH-RESOLUTION
STUDIES
Deeper studies to find rare events
NGS assays can be designed to either target a large
number of genes with low sequencing depth (more
comprehensive, less sensitive) or a relatively small
number of genes with higher sequencing depth
(less comprehensive, more sensitive). In cases of
heterogenous samples such as blood or tumors, high
sequencing depth is necessary to provide the sensitivity
required to detect low-abundance variants accurately.
Scientists can take advantage of the latest advancements
in high-throughput sequencing to perform deeper
sequencing experiments, more rapidly, at production-scale.
Liquid biopsy
Liquid biopsy detects and characterizes various tumorderived
biomarkers present in the blood of an individual
with cancer. These biomarkers are absent in healthy
individuals and patients who are cancer free. Liquid
biopsy is a noninvasive approach that offers a potential
alternative to invasive tissue biopsies to detect targetable
oncogenic drivers and resistance mutations.89
Tumors release circulating tumor DNA (ctDNA) into
the bloodstream through various cellular mechanisms,
including apoptosis, necrosis, phagocytosis, and active
secretion.90 Even so, ctDNA represents a small fraction
of total cell-free DNA (cfDNA) in the blood,91 the majority
of which is released by erythrocytes, leukocytes, and
endothelial cells.90,92 The rarity of ctDNA combined with
the fact that different cancer types at different stages
shed ctDNA at different rates complicates analysis.
However, recent improvements in NGS instrumentation
provide options for sequencing samples at extremely
high depth of coverage for large portions of the genome
in a single sample. This enables analysis of hundreds
of genes with the sequencing depth required for ctDNA
analysis. Furthermore, these technologies enable
hypothesis-free interrogation of biofluid analytes,
providing discovery power to assay genes and pathways
that were not considered prior to experimental design.
Liquid biopsy combined with comprehensive
genomic profiling (CGP) can identify tumor-specific
mutations in patient samples93 and provide information
of cancer type, stage, and vascularization.94–97 Recent
studies that performed liquid biopsy paired with
corresponding tissue biopsy from tumor samples have
demonstrated that, when comprehensive assays are
used, ctDNA analysis detected a significant number of
guideline-recommended biomarkers and resistance
alterations not found in matched tissue biopsies.98,99
Furthermore, ctDNA sequencing identifies variants in
the tumor from which it originates, including both driver
and passenger mutations, respectively.90 Given the rapid
turnover of cfDNA and ctDNA in the bloodstream, liquid
biopsy can be a powerful tool for cancer research to
assay tumor burden in real time and monitor response to
therapy.100 Studies also demonstrate how liquid biopsybased
detection of molecular residual disease (MRD) can
predict recurrence with high sensitivity and specificity,
with lead times that precede standard imaging.101–103
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
17 | M-GL-01796 v1.0
TABLE OF
CONTENTS
NOVASEQ X
SERIES
HIGH-RESOLUTION
STUDIES
Next-level liquid biopsy
Circulating analytes include more than just ctDNA and can
apply to more than just cancer. As technology continues to
advance, there is the potential to apply multiomic approaches
that integrate analyses of the genome, transcriptome,
epigenome, proteome, and microbiome to liquid biopsy.104
For example, cell-free RNA (cfRNA) is indicative of earlystage
cancer and other conditions like preeclampsia.105–111
Methylation patterns in ctDNA can also recapitulate the
abnormal methylation patterns that are a hallmark of many
cancers.112–116 The ability to combine multiomics with liquid
biopsy will provide unique discovery power for deeper insights
and comprehensive answers to the mechanisms of cancer.
Genome-wide tumor–
normal sequencing
Deep sequencing of tumor exomes or genomes can provide
rapid and accurate CGP.117,118 Through tumor–normal WGS,
researchers can compare tumor mutations to a matched
normal sample. Tumor–normal comparisons are crucial for
identifying the somatic variants that act as driver mutations
in cancer progression. Tumor–normal sequencing typically
requires a minimum of 80× sequencing depth.
WGS for cancer studies offers base-pair resolution of the
unique mutations present in a tumor. Cancer genomes typically
contain unpredictable numbers of point mutations, gene
fusions, and other aberrations. As a hypothesis-free approach,
cancer WGS offers unbiased insights into a variety of genomic
aberrations, including emerging biomarkers.119,120 It enables
discovery of novel cancer-associated variants, including
single nucleotide variants (SNVs), copy number changes,
indels, and structural variants.
Learn more
Circulating tumor DNA sequencing
Comprehensive genomic profiling
Cancer whole-genome sequencing
We've known for many
years that there is tumorrelated
material in the
bloodstream, we just
didn't have the
technologies to detect
it for it to be meaningful.
Minetta C. Liu, MD, Mayo Clinic ”
“
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
18 | M-GL-01796 v1.0
TABLE OF
CONTENTS DEEPER WORKFLOWS
STUDIES
How the NovaSeq X Series accelerates
genomic discovery
The potential of broader, deeper sequencing can now be realized, thanks to the latest advancements in Illumina NGS
systems. The NovaSeq X and NovaSeq X Plus Sequencing Systems are built with breakthrough technological innovations
to transform the economics of high-throughput sequencing.
The NovaSeq X Series is powered by XLEAP-SBS™ chemistry—a faster, higher fidelity, and more robust version of proven
Illumina sequencing by synthesis (SBS) chemistry. XLEAP-SBS reagents are optimized for performance and speed, to
maximize throughput without sacrificing data quality. Ultrahigh-resolution optics were developed to match the capabilities
of the chemistry. Ultrahigh-density patterned flow cells with tens of billions of nanowells enable up to 16 Tb output (or up
to 52 billion single reads) per dual flow cell run on the NovaSeq X Plus System.
In addition to advancements in chemistry and optics, the NovaSeq X Series is built with DRAGEN™ hardware
onboard the instrument to accelerate and streamline secondary analysis and compress data by 80% without loss.
The NovaSeq X Series also sets a new standard in operational simplicity with a software ecosystem built specifically
to support the Illumina NGS workflow.
NovaSeq X Series specification sheet
High-accuracy NGS data with the NovaSeq X Series application note
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
19 | M-GL-01796 v1.0
TABLE OF
CONTENTS DEEPER WORKFLOWS
STUDIES
Enabling broader, deeper sequencing
The NovaSeq X Series will allow scientists to perform broader, more ambitious studies at a new level of scale.
Researchers can increase the scope of their studies without increasing their budgets. Lower costs and streamlined
informatics enable more analyses per sample and more data per analysis.
With the ability to sequence three-fold more samples, population genomics researchers can add statistical power with
larger cohorts—up to tens of thousands of genomes per year. Cancer or genetic disease researchers can sequence
three-fold deeper to detect low-frequency signals and rare variants. Other researchers can easily adopt multiomic
interrogation techniques with more omes and multiple simultaneous analyses. The comprehensive, high-resolution view
of biological systems will expand the discovery power of genomic scientists.
25B
350
300
450
400
250
200
150
100
50
0
10B 1.5B S4 S2 S1 SP NextSeqTM
2000 P3
EXAMPLE PROJECT
Single-cell gene expression study with fixed $50K reagent budget‡
~2×
NovaSeq X Series NovaSeq 6000 System
Single-cell samples per $50K reagent budget
‡ Analysis based on 20,000 reads per cell and 10,000 cells per sample.
All pricing is in USD, based on United States list prices.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
20 | M-GL-01796 v1.0
TABLE OF
CONTENTS DEEPER WORKFLOWS
STUDIES
The NovaSeq X Series more than doubles the throughput of the NovaSeq 6000 System, while taking up the same lab footprint—less than
one square meter of floor space. NovaSeq X and NovaSeq X Plus Systems provide massive throughput and productivity gains with the
ability to sequence ~2.5 genomes/hour and up to 128 genomes (30× coverage) per run and 96 human genomes (40× coverage) per dual
25B flow cell run.
The NovaSeq X Series decreases cost/Gb
by up to 50% and shrinks the cost gap between
WGS and WES by 2.5× to a difference of only
$183 (USD).
~2.5×
~2×
Throughput
Faster
NovaSeq 6000 System NovaSeq X Series
65 Gb–6 Tb Output 165 Gb–16 Tb
800M–20B Read number 1.68B–52B
~1 genome/hr Genomes per hour ~2.5 genomes/hr
2–48 genomes Genomes per run 4–128 genomes
WGS WES
SHRINKING THE GAP
WGS and WES
Sequencing cost
Library prep cost
$183
Setting a new bar in every sequencing metric
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
21 | M-GL-01796 v1.0
TABLE OF
CONTENTS DEEPER WORKFLOWS
STUDIES
Managing big experiments and big data
with the NovaSeq X Series
Day-to-day operational simplicity
Illumina believes that genomics should be available to the many, not the few. We have an obligation to make our
technology as affordable and accessible as possible while setting the highest standard for data quality and security.
To democratize access to the power of the genome, the NovaSeq X Series was designed to be especially easy to
operate, from run planning through analysis. The simple workflow requires fewer touchpoints and fewer steps than the
NovaSeq 6000 System, reducing the learning curve and the chance of user error.
Expanded global access to NGS
The stability of XLEAP-SBS reagents allows for
ambient temperature shipping, without ice packs or
dry ice. Eliminating the need for cold-chain transport
expands access to NGS globally, especially for hardto-
reach geographies. This key innovation will speed
adoption of high-throughput sequencing for more
diverse populations.
Flexible scalability
The choice of three flow cell types with individually
addressable lanes offers flexible scalability. The
NovaSeq X 10B and 25B flow cells enable larger
projects and deeper sequencing. The NovaSeq X
1.5B flow cell eases the transition from benchtop to
higher-throughput sequencing. The fast turnaround
times for the 1.5B flow cell are ideal for small batch
sizes and data-lite applications.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
22 | M-GL-01796 v1.0
TABLE OF
CONTENTS
Adjustable screen height with position memory
4K ultra
high-definition
touch screen
System prompts
to guide user
along workflow
Easy-to-handle flow cell with illuminated flow cell stage
Push-to-access hidden keyboard
Light-guided consumable loading
Assisted open doors and waist-height reagent drawers
Less time and effort with established
workflow applications
Reduced chance of error with intuitive
user interface with clear visualization
of run status and lane-level metrics,
and immediate view of secondary
analysis reports and QC metrics
> 90% reduction in reagent packaging
waste/weight and 50% reduction in
plastic for easier handling and reduced
disposal costs§
Designed with the user in mind
DEEPER WORKFLOWS
STUDIES
§ In comparison to the NovaSeq 6000 System.
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
23 | M-GL-01796 v1.0
TABLE OF
CONTENTS DEEPER WORKFLOWS
STUDIES
Integrated, cohesive bioinformatics ecosystem
The NovaSeq X Series streamlines genomic data management
with onboard secondary analysis and integration with a
comprehensive suite of bioinformatics solutions. The
Illumina Connected Software suite includes some of the
fastest, most accurate,94 and advanced solutions for data
analysis, interpretation, and aggregation. With flexible local
and cloud-based options for lab operations, run planning,
and data analysis, the NovaSeq X Series enables users to
run high-throughput sequencing without creating a
bioinformatics bottleneck.
Plug-and-play onboard DRAGEN workflows enable variant
calling directly on the system. Up to four DRAGEN applications
can run in parallel, speeding multiomic analysis. Integrated
DRAGEN ORA (original read archive) data compression reduces
data storage needs five-fold. The savings on server, licenses,
and cloud storage can total more than $1 million (USD) over
five years of ownership.** Access to a broader menu via the
cloud with highly configurable pipelines will accelerate larger,
more data-intensive sequencing projects.
** Assuming FASTQ files are archived 30 days after cloud upload
and throughput of 10,000 WGS/year for the NovaSeq X System and
20,000 WGS/year for the NovaSeq X Plus System.
“
“
The physical reduction in kit
size has greatly decreased
our cold storage needs
in lab and elimination of
dry ice shipping has made
unpacking larger orders
much easier.
One of the big benefits
I can see in the new
DRAGEN onboard analysis
on the NovaSeq X Plus
System is the considerable
reduction in file size now
being compressed to
approximately one-fifth of
what would be a traditional
file, which allows us to
run more projects at
population scale.
Eric Chow, Assistant Adjunct Professor
and Director, University of California
San Francisco (UCSF) Center for Advanced
Technology
Lachlan Morrison, Senior Technical Officer
Biomolecular Resource Facility (BRF),
Australian National University (ANU)
”
”
NovaSeq X Series software ecosystem technical note
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
24 | M-GL-01796 v1.0
TABLE OF
CONTENTS NOVASEQ X CONCLUSION
SERIES
Example NovaSeq X Series workflows
Illumina DNA
PCR-Free Prep
25–300 ng DNA input
~1.5-hour workflow
DRAGEN Germline
(onboard or in the cloud)
NovaSeq X 1.5B flow cell
~4 samples per flow cell
NovaSeq X 10B flow cell
~24 samples per flow cell
NovaSeq X 25B flow cell
~64 samples per flow cell
400M reads per sample
2 × 150 bp read length
Prepare libaries Sequence Analyze data
WGS
Illumina Complete Long
Read Prep, Human
50 ng DNA input
~1-day workflow
Illumina Complete
Long Read WGS App
(in the cloud)
NovaSeq X 10B flow cell
4 samples per flow cell
NovaSeq X 25B flow cell
10–11 samples per flow cell
750 Gb per sample
2 × 150 bp read length
Illumina DNA Prep with
Exome 2.0 Plus Enrichment
50–1000 ng DNA input
~6.5-hour workflow
DRAGEN Enrichment
(onboard or in the cloud)
NovaSeq X 1.5B flow cell
~41 samples per flow cell
NovaSeq X 10B flow cell
~250 samples per flow cell
NovaSeq X 25B flow cell
~750 samples per flow cell
8 Gb per sample
2 × 100 bp read length
Prepare libaries Sequence Analyze data
WES
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
25 | M-GL-01796 v1.0
TABLE OF
CONTENTS NOVASEQ X CONCLUSION
SERIES
Illumina Stranded Total
RNA Prep
1–1000 ng high-quality
DNA input
~7-hour workflow
DRAGEN RNA
(onboard or in the cloud)
NovaSeq X 1.5B flow cell
~32 samples per flow cell
NovaSeq X 10B flow cell
~200 samples per flow cell
NovaSeq X 25B flow cell
~520 samples per flow cell
50M reads per sample
2 × 100 bp read length
Prepare libaries Sequence Analyze data
Illumina Stranded
mRNA Prep
25–1000 ng high-quality
RNA input
~7-hour workflow
DRAGEN RNA
(onboard or in the cloud)
NovaSeq X 1.5B flow cell
~64 samples per flow cell
NovaSeq X 10B flow cell
~400 samples per flow cell
NovaSeq X 25B flow cell
~1040 samples per flow cell
25M reads per sample
2 × 100 bp read length
Illumina RNA Prep
with Enrichment
20 ng FFPE RNA input
~9-hour workflow
DRAGEN RNA
(onboard or in the cloud)
WTS
NovaSeq X 1.5B flow cell
~64 samples per flow cell
NovaSeq X 10B flow cell
~400 samples per flow cell
NovaSeq X 25B flow cell
~1040 samples per flow cell
25M reads per sample
2 × 100 bp read length
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
26 | M-GL-01796 v1.0
TABLE OF
CONTENTS WORKFLOWS ABBREVIATIONS
Unimaginable experiments,
made possible
The NovaSeq X Series is driven by your vision to help moonshot
projects become a reality. Increase statistical power with
broader study designs and larger sample cohorts. Maximize
read numbers and increase sequencing depth for a higher
resolution view that can detect low-frequency signals and
variants. Answer the most complex questions in human
genomics, with larger sample cohorts, deeper sequencing,
and more data-intensive methods—from whole-genome
sequencing to multiomics.
Genomics visionaries are ready to usher in the Genome Era,
and the NovaSeq X Series can help make it happen.
“[The NovaSeq X Series]
now allows us to provide
much larger scale projects
for both human health and
nonhuman scenarios in
the research world, and
to be able to translate
that into more meaningful
outcomes.
Joe Baini, CEO, Australian Genome Research
Facility (AGRF) ”
NovaSeq X and NovaSeq X Plus Sequencing Systems
NovaSeq X Series virtual tour
NovaSeq X Series overview video
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
27 | M-GL-01796 v1.0
TABLE OF
CONTENTS CONCLUSION REFERENCES
Abbreviations
ATAC-Seq: assay for transposase-accessible chromatin
BEN-Seq: bulk epitope and nucleic acid sequencing
CGP: comprehensive genomic profiling
cfDNA: cell-free DNA
ChIP-Seq: chromatin immunoprecipitation sequencing
CITE-Seq: cellular indexing of transcriptomes and
epitopes by sequencing
CMA: chromosomal microarray
CNV: copy number variant
CRISPR: clustered regularly interspaced short palindromic
repeats
ctDNA: circulating tumor DNA
eQTL: expression quantitative trait loci
FFPE: formalin-fixed, paraffin-embedded
Gb: gigabase
GWAS: genome-wide association studies
HiC: chromosome conformation capture
Indel: insertion–deletion
MRD: molecular residual disease
NGS: next-generation sequencing
NICU: neonatal intensive care unit
RNA-Seq: RNA sequencing
SBS: sequencing by synthesis
scRNA-Seq: single-cell RNA sequencing
SNV: single nucleotide variant
SV: structural variant
Tb: terabase
VUS: variant of unknown significance
WES: whole-exome sequencing
WGS: whole-genome sequencing
WGTS: combined whole-genome and transcriptome
sequencing
WTS: whole-transcriptome sequencing
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
28 | M-GL-01796 v1.0
TABLE OF
CONTENTS ABBREVIATIONS
References
1. 100,000 Genomes Project. Genomics England. genomicsengland.co.uk/
initiatives/100000-genomes-project. Accessed June 19, 2023.
2. 100,000 Genomes Project Pilot Investigators, Smedley D, Smith KR, et al. 100,000
Genomes Pilot on Rare-Disease Diagnosis in Health Care - Preliminary Report.
N Engl J Med. 2021;385(20):1868-1880. doi:10.1056/NEJMoa2035790
3. Turro E, Astle WJ, Megy K, et al. Whole-genome sequencing of patients with rare
diseases in a national health system. Nature. 2020;583(7814):96-102. doi:10.1038/
s41586-020-2434-2
4. UK Biobank. ukbiobank.ac.uk/. Accessed September 1, 2023.
5. Degasperi A, Zou X, Dias Amarante T, et al. Substitution mutational signatures
in whole-genome–sequenced cancers in the UK population. Science.
2022;376(6591):abl9283. doi:10.1126/science.abl9283
6. Our Future Health. ourfuturehealth.org.uk/. Accessed June 19, 2023.
7. H3Africa – Human Heredity & Health in Africa. h3africa.org/. Accessed June 19,
2023.
8. Care AGD of H and A. Genomics Health Futures Mission. Australian Government
Department of Health and Aged Care. health.gov.au/our-work/genomics-healthfutures-
mission. Published June 2, 2023. Accessed June 19, 2023.
9. Australia begins a big push to sequence underserved populations. illumina.com/
content/illumina-marketing/amr/en_US/company/news-center/feature-articles/
australia-begins-a-big-push-to-sequence-underserved-populations.html.
Accessed June 19, 2023.
10. GenomeAsia 100k. genomeasia100k.org/index.html. Accessed June 19, 2023.
11. Wall JD, Stawiski EW, Ratan A, et al. The GenomeAsia 100K Project enables genetic
discoveries across Asia. Nature. 2019;576(7785):106-111. doi:10.1038/s41586-019-
1793-z
12. Precision Health Research, Singapore (PRECISE). npm.sg/. Accessed June 16, 2023.
13. Lethimonnier F, Levy Y. Genomic medicine France 2025. Ann Oncol.
2018;29(4):783-784. doi:10.1093/annonc/mdy027
14. Petrone J. Ireland outlines national strategy to streamline access to genomic
medicine. GenomeWeb. genomeweb.com/research-funding/ireland-outlinesnational-
strategy-streamline-access-genomic-medicine. Published February 2,
2023, Accessed September 1, 2023.
15. Gudbjartsson DF, Helgason H, Gudjonsson SA, et al. Large-scale whole-genome
sequencing of the Icelandic population. Nat Genet. 2015;47(5):435-444.
doi:10.1038/ng.3247
16. Apol KD, Lydersen LN, Mortensen Ó, et al. FarGen - participants in the genetic
research infrastructure of the Faroe Islands. Scand J Public Health. 2022;50(7):980-
987. doi:10.1177/14034948211046817
17. Kurki MI, Karjalainen J, Palta P, et al. FinnGen provides genetic insights from a wellphenotyped
isolated population. Nature. 2023;613(7944):508-518. doi:10.1038/
s41586-022-05473-8
18. Estonian Biobank. University of Tartu. Institute of Genomics. genomics.ut.ee/en/
content/estonian-biobank. Published November 29, 2021. Accessed September 1,
2023.
19. BBMRI-ERIC. Turkish Genome Project Launched. bbmri-eric.eu/news-events/
turkish-genome-project-launched. Accessed September 1, 2023.
20. The National. UAE President launches National Genome Strategy to prioritise
Emirati health care. Published March 28, 2023. Accessed September 1, 2023.
21. Saudi Genome Program. vision2030.gov.sa/v2030/v2030-projects/saudi-genomeprogram/.
Accessed September 1, 2023.
22. Gilmore J. Israel to Sequence 100K People, Create Genomic Database to Support
'Digital Health'. GenomeWeb. genomeweb.com/sequencing/israel-sequence-100kpeople-
create-genomic-database-support-digital-health. Published December 13,
2018. Accessed September 1, 2023.
23. Takahashi Y, Mizusawa H. Initiative on Rare and Undiagnosed Disease in Japan.
JMA J. 2021;4(2):112-118. doi:10.31662/jmaj.2021-0003
24. Hong Kong Genome Institute. hkgp.org/en/. Accessed September 1, 2023.
25. Illumina and China’s Children’s Hospital of Fudan Bring Rapid WGS to the NICU.
illumina.com/content/illumina-marketing/amr/en_US/company/news-center/featurearticles/
illumina-and-children-s-hospital-of-fudan-in-china.html. Published April 22,
2019. Accessed September 1, 2023.
26. CGTN. China Genome Project: China launches 100,000 Genomes Project. Published
December 29, 2017. Accessed September 1, 2023.
27. Jaber IG. TecScience. OriGen Project seeks to solve the enigma of the Mexican
genome. ecscience.tec.mx/en/health/mexican-genome/. Published February 15,
2023. Accessed September 1, 2023.
28. Coelho AVC, Mascaro-Cordeiro B, Lucon DR, et al. The Brazilian Rare Genomes
Project: Validation of Whole Genome Sequencing for Rare Diseases Diagnosis. Front
Mol Biosci. 2022;9:821582. doi:10.3389/fmolb.2022.821582
29. All of Us Research Program. National Institutes of Health (NIH). allofus.nih.gov/.
Accessed September 1, 2023.
30. Million Veteran Program. US Department of Veteran Affairs. research.va.gov/mvp/.
Accessed September 1, 2023.
31. Precision Medicine Initiative (PMI) Cohort Program. National Institute on Minority
Health and Health Disparities. nimhd.nih.gov/programs/collab/pmi/. Accessed
September 1, 2023.
32. Murray M. CT Forum on DNA Testing and Genomics Medicine. Yale Center for
Genomic Health. Generations Project and DNA-based Health Screening. Published
January 23, 2020. Accessed September 1, 2023.
33. Fatumo S, Chikowore T, Choudhury A, Ayub M, Martin AR, Kuchenbaecker K.
A roadmap to increase diversity in genomic studies. Nat Med. 2022;28(2):243-250.
doi:10.1038/s41591-021-01672-4
34. Liao WW, Asri M, Ebler J, et al. A draft human pangenome reference. Nature.
2023;617(7960):312-324. doi:10.1038/s41586-023-05896-x
35. What our primate relatives can teach us about our own genomes. illumina.com/
content/illumina-marketing/amr/en_US/company/news-center/feature-articles/
PrimateAI-3D.html. Accessed June 21, 2023.
36. Improving genetic risk prediction and drug target discovery using primate
DNA and advanced artificial intelligence. illumina.com/content/illumina-marketing/
amr/en_US/science/genomics-research/articles/primateai-3d.html. Accessed
June 21, 2023.
37. Lionel AC, Costain G, Monfared N, et al. Improved diagnostic yield compared with
targeted gene sequencing panels suggests a role for whole-genome sequencing as
a first-tier genetic test. Genet Med Off J Am Coll Med Genet. 2018;20(4):435-443.
doi:10.1038/gim.2017.119
38. Chen X, Schulz-Trieglaff O, Shaw R, et al. Manta: rapid detection of structural
variants and indels for germline and cancer sequencing applications.
Bioinforma Oxf Engl. 2016;32(8):1220-1222. doi:10.1093/bioinformatics/btv710
39. Dolzhenko E, van Vugt JJFA, Shaw RJ, et al. Detection of long repeat expansions
from PCR-free whole-genome sequence data. Genome Res. 2017;27(11):1895-1903.
doi:10.1101/gr.225672.117
40. Alfares A, Aloraini T, Subaie LA, et al. Whole-genome sequencing offers additional
but limited clinical utility compared with reanalysis of whole-exome sequencing.
Genet Med Off J Am Coll Med Genet. 2018;20(11):1328-1333. doi:10.1038/
gim.2018.41
41. Chen X, Sanchis-Juan A, French CE, et al. Spinal muscular atrophy diagnosis
and carrier screening from genome sequencing data. Genet Med Off J Am Coll Med
Genet. 2020;22(5):945-953. doi:10.1038/s41436-020-0754-0
42. Sanghvi RV, Buhay CJ, Powell BC, et al. Characterizing reduced coverage regions
through comparison of exome and genome sequencing data across 10 centers.
Genet Med Off J Am Coll Med Genet. 2018;20(8):855-866. doi:10.1038/gim.2017.192
43. Lindstrand A, Eisfeldt J, Pettersson M, et al. From cytogenetics to cytogenomics:
whole-genome sequencing as a first-line test comprehensively captures the
diverse spectrum of disease-causing genetic variation underlying intellectual
disability. Genome Med. 2019;11(1):68. doi:10.1186/s13073-019-0675-1
44. Meienberg J, Bruggmann R, Oexle K, Matyas G. Clinical sequencing: is WGS the
better WES? Hum Genet. 2016;135(3):359-362. doi:10.1007/s00439-015-1631-9
45. Trost B, Walker S, Wang Z, et al. A Comprehensive Workflow for Read Depth-Based
Identification of Copy-Number Variation from Whole-Genome Sequence Data. Am J
Hum Genet. 2018;102(1):142-155. doi:10.1016/j.ajhg.2017.12.007
46. Stavropoulos DJ, Merico D, Jobling R, et al. Whole Genome Sequencing
Expands Diagnostic Utility and Improves Clinical Management in Pediatric
Medicine. NPJ Genomic Med. 2016;1:15012-. doi:10.1038/npjgenmed.2015.12
47. Belkadi A, Bolze A, Itan Y, et al. Whole-genome sequencing is more powerful
than whole-exome sequencing for detecting exome variants. Proc Natl Acad
Sci U S A. 2015;112(17):5473-5478. doi:10.1073/pnas.1418631112
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
29 | M-GL-01796 v1.0
TABLE OF
CONTENTS ABBREVIATIONS
48. Carss KJ, Arno G, Erwood M, et al. Comprehensive Rare Variant Analysis
via Whole-Genome Sequencing to Determine the Molecular Pathology of
Inherited Retinal Disease. Am J Hum Genet. 2017;100(1):75-90. doi:10.1016/j.
ajhg.2016.12.003
49. Souche E, Beltran S, Brosens E, et al. Recommendations for whole genome
sequencing in diagnostics for rare diseases. Eur J Hum Genet EJHG.
2022;30(9):1017-1021. doi:10.1038/s41431-022-01113-x
50. Gross AM, Ajay SS, Rajan V, et al. Copy-number variants in clinical genome
sequencing: deployment and interpretation for rare and undiagnosed disease.
Genet Med Off J Am Coll Med Genet. 2019;21(5):1121-1130. doi:10.1038/s41436-
018-0295-y
51. van Kuilenburg ABP, Tarailo-Graovac M, Richmond PA, et al. Glutaminase
Deficiency Caused by Short Tandem Repeat Expansion in GLS. N Engl J Med.
2019;380(15):1433-1441. doi:10.1056/NEJMoa1806627
52. Ibañez K, Polke J, Hagelstrom RT, et al. Whole genome sequencing for the
diagnosis of neurological repeat expansion disorders in the UK: a retrospective
diagnostic accuracy and prospective clinical validation study. Lancet Neurol.
2022;21(3):234-245. doi:10.1016/S1474-4422(21)00462-2
53. Splinter K, Adams DR, Bacino CA, et al. Effect of Genetic Diagnosis on Patients
with Previously Undiagnosed Disease. N Engl J Med. 2018;379(22):2131-2139.
doi:10.1056/NEJMoa1714458
54. Holliday M, Singer ES, Ross SB, et al. Transcriptome Sequencing of Patients
With Hypertrophic Cardiomyopathy Reveals Novel Splice-Altering Variants
in MYBPC3. Circ Genomic Precis Med. 2021;14(2):e003202. doi:10.1161/
CIRCGEN.120.00320241.
55. Zhu F, Zhang F, Hu L, Liu H, Li Y. Integrated Genome and Transcriptome Sequencing
to Solve a Neuromuscular Puzzle: Miyoshi Muscular Dystrophy and Early Onset
Primary Dystonia in Siblings of the Same Family. Front Genet. 2021;12:672906.
doi:10.3389/fgene.2021.672906
56. Karam R, Conner B, LaDuca H, et al. Assessment of Diagnostic Outcomes of RNA
Genetic Testing for Hereditary Cancer. JAMA Netw Open. 2019;2(10):e1913900.
doi:10.1001/jamanetworkopen.2019.13900
57. Pon JR, Marra MA. Driver and passenger mutations in cancer. Annu Rev Pathol.
2015;10:25-50. doi:10.1146/annurev-pathol-012414-040312
58. Golub TR, Slonim DK, Tamayo P, et al. Molecular classification of cancer:
class discovery and class prediction by gene expression monitoring. Science.
1999;286(5439):531-537. doi:10.1126/science.286.5439.531
59. Valk PJM, Verhaak RGW, Beijen MA, et al. Prognostically useful gene-expression
profiles in acute myeloid leukemia. N Engl J Med. 2004;350(16):1617-1628.
doi:10.1056/NEJMoa040465
60. Docking TR, Parker JDK, Jädersten M, et al. A clinical transcriptome approach
to patient stratification and therapy selection in acute myeloid leukemia.
Nat Commun. 2021;12(1):2474. doi:10.1038/s41467-021-22625-y
61. Lilljebjörn H, Orsmark-Pietras C, Mitelman F, Hagström-Andersson A,
Fioretos T. Transcriptomics paving the way for improved diagnostics and precision
medicine of acute leukemia. Semin Cancer Biol. 2022;84:40-49. doi:10.1016/j.
semcancer.2021.09.013
62. Hehir-Kwa JY, Koudijs MJ, Verwiel ETP, et al. Improved Gene Fusion Detection
in Childhood Cancer Diagnostics Using RNA Sequencing. JCO Precis Oncol.
2022;6:e2000504. doi:10.1200/PO.20.00504
63. Cuppen E, Elemento O, Rosenquist R, et al. Implementation of Whole-Genome
and Transcriptome Sequencing Into Clinical Cancer Care. JCO Precis Oncol.
2022;6:e2200245. doi:10.1200/PO.22.00245
64. Kremer LS, Bader DM, Mertes C, et al. Genetic diagnosis of Mendelian disorders via
RNA sequencing. Nat Commun. 2017;8:15824. doi:10.1038/ncomms15824
65. Cummings BB, Marshall JL, Tukiainen T, et al. Improving genetic diagnosis
in Mendelian disease with transcriptome sequencing. Sci Transl Med.
2017;9(386):eaal5209. doi:10.1126/scitranslmed.aal5209
66. Frésard L, Smail C, Ferraro NM, et al. Identification of rare-disease genes
using blood transcriptome sequencing and large control cohorts. Nat Med.
2019;25(6):911-919. doi:10.1038/s41591-019-0457-8
67. Doke T, Huang S, Qiu C, et al. Transcriptome-wide association analysis identifies
DACH1 as a kidney disease risk gene that contributes to fibrosis. J Clin Invest.
2021;131(10):e141801, 141801. doi:10.1172/JCI141801
68. Odhams CA, Cunninghame Graham DS, Vyse TJ. Profiling RNA-Seq at multiple
resolutions markedly increases the number of causal eQTLs in autoimmune disease.
PLoS Genet. 2017;13(10):e1007071. doi:10.1371/journal.pgen.1007071
69. Võsa U, Claringbould A, Westra HJ, et al. Large-scale cis- and trans-eQTL analyses
identify thousands of genetic loci and polygenic scores that regulate blood gene
expression. Nat Genet. 2021;53(9):1300-1310. doi:10.1038/s41588-021-00913-z
70. Colin E, Duffourd Y, Tisserant E, et al. OMIXCARE: OMICS technologies solved
about 33% of the patients with heterogeneous rare neuro-developmental disorders
and negative exome sequencing results and identified 13% additional candidate
variants. Front Cell Dev Biol. 2022;10:1021785. doi:10.3389/fcell.2022.1021785
71. Kamali Z, Keaton JM, Haghjooy Javanmard S, et al. Large-Scale Multi-Omics
Studies Provide New Insights into Blood Pressure Regulation. Int J Mol Sci.
2022;23(14):7557. doi:10.3390/ijms23147557
72. Lunke S, Bouffler SE, Patel CV, et al. Integrated multi-omics for rapid rare disease
diagnosis on a national scale. Nat Med. 2023:1-11. doi:10.1038/s41591-023-02401-9
73. Longo SK, Guo MG, Ji AL, Khavari PA. Integrating single-cell and spatial
transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet.
2021;22(10):627-644. doi:10.1038/s41576-021-00370-8
74. Delorey TM, Ziegler CGK, Heimberg G, et al. COVID-19 tissue atlases reveal
SARS-CoV-2 pathology and cellular targets. Nature. 2021;595(7865):107-113.
doi:10.1038/s41586-021-03570-8
75. Desai N, Neyaz A, Szabolcs A, et al. Temporal and spatial heterogeneity of host
response to SARS-CoV-2 pulmonary infection. Nat Commun. 2020;11(1):6319.
doi:10.1038/s41467-020-20139-7
76. Scheid JF, Barnes CO, Eraslan B, et al. B cell genomics behind crossneutralization
of SARS-CoV-2 variants and SARS-CoV. Cell. 2021;184(12):3205-
3221.e24. doi:10.1016/j.cell.2021.04.032
77. Huang N, Pérez P, Kato T, et al. SARS-CoV-2 infection of the oral cavity
and saliva. Nat Med. 2021;27(5):892-903. doi:10.1038/s41591-021-01296-8
78. Butler D, Mozsary C, Meydan C, et al. Shotgun transcriptome, spatial omics, and
isothermal profiling of SARS-CoV-2 infection reveals unique host responses,
viral diversification, and drug interactions. Nat Commun. 2021;12(1):1660.
doi:10.1038/s41467-021-21361-7
79. Zhang Y, Narayanan SP, Mannan R, et al. Single-cell analyses of renal cell
cancers reveal insights into tumor microenvironment, cell of origin, and therapy
response. Proc Natl Acad Sci U S A. 2021;118(24):e2103240118. doi:10.1073/
pnas.2103240118
80. Peng M, Wei G, Zhang Y, et al. Single-cell transcriptomic landscape reveals the
differences in cell differentiation and immune microenvironment of papillary
thyroid carcinoma between genders. Cell Biosci. 2021;11(1):39. doi:10.1186/
s13578-021-00549-w
81. Lalli MA, Avey D, Dougherty JD, Milbrandt J, Mitra RD. High-throughput singlecell
functional elucidation of neurodevelopmental disease-associated genes
reveals convergent mechanisms altering neuronal differentiation. Genome Res.
2020;30(9):1317-1331. doi:10.1101/gr.262295.120
82. Brady L, Kriner M, Coleman I, et al. Inter- and intra-tumor heterogeneity of
metastatic prostate cancer determined by digital spatial gene expression
profiling. Nat Commun. 2021;12(1):1426. doi:10.1038/s41467-021-21615-4
83. Ji AL, Rubin AJ, Thrane K, et al. Multimodal Analysis of Composition and Spatial
Architecture in Human Squamous Cell Carcinoma. Cell. 2020;182(6):1661-1662.
doi:10.1016/j.cell.2020.08.043
84. Fawkner-Corbett D, Antanaviciute A, Parikh K, et al. Spatiotemporal analysis of
human intestinal development at single-cell resolution. Cell. 2021;184(3):810-
826.e23. doi:10.1016/j.cell.2020.12.016
85. Cao J, Spielmann M, Qiu X, et al. The single-cell transcriptional landscape of
mammalian organogenesis. Nature. 2019;566(7745):496-502. doi:10.1038/
s41586-019-0969-x
86. Plasschaert LW, Žilionis R, Choo-Wing R, et al. A single-cell atlas of the
airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature.
2018;560(7718):377-381. doi:10.1038/s41586-018-0394-6
87. BRAIN Initiative Cell Census Network (BICCN). A multimodal cell census and
atlas of the mammalian primary motor cortex. Nature. 2021;598(7879):86-102.
doi:10.1038/s41586-021-03950-0
88. Replogle JM, Saunders RA, Pogson AN, et al. Mapping information-rich
genotype-phenotype landscapes with genome-scale Perturb-seq. Cell.
2022;185(14):2559-2575.e28. doi:10.1016/j.cell.2022.05.013
89. Li BT, Janku F, Jung B, et al. Ultra-deep next-generation sequencing of
plasma cell-free DNA in patients with advanced lung cancers: results from
the Actionable Genome Consortium. Ann Oncol. 2019;30(4):597-603. doi:10.1093/
annonc/mdz046
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY
30 | M-GL-01796 v1.0
TABLE OF
CONTENTS
1.800.809.4566 toll-free (US) | +1.858.202.4566 tel
techsupport@illumina.com | www.illumina.com
©2023 Illumina, Inc. All rights reserved. All trademarks are
the property of Illumina, Inc. or their respective owners.
For specific trademark information, see
www.illumina.com/company/legal.html.
90. Chin RI, Chen K, Usmani A, et al. Detection of Solid Tumor Molecular Residual
Disease (MRD) Using Circulating Tumor DNA (ctDNA). Mol Diagn Ther.
2019;23(3):311-331. doi:10.1007/s40291-019-00390-5
91. Xu J, Liao K, Yang X, Wu C, Wu W. Using single-cell sequencing technology
to detect circulating tumor cells in solid tumors. Mol Cancer. 2021;20(1):104.
doi:10.1186/s12943-021-01392-w
92. Grabuschnig S, Bronkhorst AJ, Holdenrieder S, et al. Putative Origins of Cell-Free
DNA in Humans: A Review of Active and Passive Nucleic Acid Release Mechanisms.
Int J Mol Sci. 2020;21(21):8062. doi:10.3390/ijms21218062
93. van ’t Erve I, Medina JE, Leal A, et al. Metastatic Colorectal Cancer Treatment
Response Evaluation by Ultra-Deep Sequencing of Cell-Free DNA and Matched
White Blood Cells. Clin Cancer Res. 2023;29(5):899-909. doi:10.1158/1078-0432.
CCR-22-2538
94. Hillen F, Griffioen AW. Tumour vascularization: sprouting angiogenesis and beyond.
Cancer Metastasis Rev. 2007;26(3-4):489-502. doi:10.1007/s10555-007-9094-7
95. Bonner ER, Harrington R, Eze A, et al. Circulating tumor DNA sequencing
provides comprehensive mutation profiling for pediatric central nervous system
tumors. NPJ Precis Oncol. 2022;6(1):63. doi:10.1038/s41698-022-00306-3
96. Nakamura Y, Taniguchi H, Ikeda M, et al. Clinical utility of circulating tumor
DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GISCREEN
and GOZILA studies. Nat Med. 2020;26(12):1859-1864. doi:10.1038/
s41591-020-1063-5
97. Bayle A, Belcaid L, Aldea M, et al. Clinical utility of circulating tumor DNA
sequencing with a large panel: a National Center for Precision Medicine
(PRISM) study. Ann Oncol Off J Eur Soc Med Oncol. 2023;34(4):389-396.
doi:10.1016/j.annonc.2023.01.008
98. Leighl NB, Page RD, Raymond VM, et al. Clinical Utility of Comprehensive
Cell-free DNA Analysis to Identify Genomic Biomarkers in Patients with
Newly Diagnosed Metastatic Non-small Cell Lung Cancer. Clin Cancer Res
Off J Am Assoc Cancer Res. 2019;25(15):4691-4700. doi:10.1158/1078-0432.
CCR-19-0624
99. Parikh AR, Leshchiner I, Elagina L, et al. Liquid versus tissue biopsy for
detecting acquired resistance and tumor heterogeneity in gastrointestinal
cancers. Nat Med. 2019;25(9):1415-1421. doi:10.1038/s41591-019-0561-9
100. Sanz-Garcia E, Zhao E, Bratman SV, Siu LL. Monitoring and adapting cancer
treatment using circulating tumor DNA kinetics: Current research, opportunities,
and challenges. Sci Adv. 2022;8(4):eabi8618. doi:10.1126/sciadv.abi8618
101. Tie J, Cohen JD, Lahouel K, et al. Circulating Tumor DNA Analysis Guiding
Adjuvant Therapy in Stage II Colon Cancer. N Engl J Med. 2022;386(24):2261-
2272. doi:10.1056/NEJMoa2200075
102. Kasi PM, Fehringer G, Taniguchi H, et al. Impact of Circulating Tumor DNABased
Detection of Molecular Residual Disease on the Conduct and Design of
Clinical Trials for Solid Tumors. JCO Precis Oncol. 2022;6:e2100181. doi:10.1200/
PO.21.00181
103. Kotani D, Oki E, Nakamura Y, et al. Molecular residual disease and efficacy
of adjuvant chemotherapy in patients with colorectal cancer. Nat Med.
2023;29(1):127-134. doi:10.1038/s41591-022-02115-4
104. Keup C, Suryaprakash V, Storbeck M, Hoffmann O, Kimmig R, Kasimir-Bauer S.
Longitudinal multi-parametric liquid biopsy approach identifies unique features
of circulating tumor cell, extracellular vesicle, and cell-free dna characterization
for disease monitoring in metastatic breast cancer patients. Cells. 2021;10(2):212.
doi:10.3390/cells10020212
105. Kopreski MS, Benko FA, Gocke CD. Circulating RNA as a tumor marker: detection
of 5T4 mRNA in breast and lung cancer patient serum. Ann N Y Acad Sci.
2001;945:172-178.
106. Perhavec A, Cerkovnik P, Novakovic S, Zgajnar J. The hTERT mRNA in plasma
samples of early breast cancer patients, non-cancer patients and healthy
individuals. Neoplasma. 2008;55(6):549-554.
107. Castellanos-Rizaldos E, Zhang X, Tadigotla VR, et al. Exosome-based detection of
activating and resistance EGFR mutations from plasma of non-small cell lung cancer
patients. Oncotarget. 2019;10(30):2911-2920. doi:10.18632/oncotarget.26885
108. Cheung KWE, Choi SYR, Lee LTC, et al. The potential of circulating cell free RNA as
a biomarker in cancer. Expert Rev Mol Diagn. 2019;19(7):579-590. doi:10.1080/1473
7159.2019.1633307
109. Larson MH, Pan W, Kim HJ, et al. A comprehensive characterization of the cellfree
transcriptome reveals tissue- and subtype-specific biomarkers for cancer
detection. Nat Commun. 2021;12(1):2357. doi:10.1038/s41467-021-22444-1
110. Munchel S, Rohrback S, Randise-Hinchliff C, et al. Circulating transcripts in maternal
blood reflect a molecular signature of early-onset preeclampsia. Sci Transl Med.
2020;12(550):eaaz0131. doi:10.1126/scitranslmed.aaz0131
111. Roskams-Hieter B, Kim HJ, Anur P, et al. Plasma cell-free RNA profiling
distinguishes cancers from pre-malignant conditions in solid and hematologic
malignancies. NPJ Precis Oncol. 2022;6(1):28. doi:10.1038/s41698-022-00270-y
112. Lissa D, Robles AI. Methylation analyses in liquid biopsy. Transl Lung Cancer Res.
2016;5(5):492-504. doi:10.21037/tlcr.2016.10.03
113. Laird PW. The power and the promise of DNA methylation markers. Nat Rev Cancer.
2003;3(4):253-266. doi:10.1038/nrc1045
114. Huang J, Wang L. Cell-Free DNA Methylation Profiling Analysis-Technologies and
Bioinformatics. Cancers. 2019;11(11):1741. doi:10.3390/cancers11111741
115. Luo H, Wei W, Ye Z, Zheng J, Xu RH. Liquid Biopsy of Methylation Biomarkers
in Cell-Free DNA. Trends Mol Med. 2021;27(5):482-500. doi:10.1016/j.
molmed.2020.12.011
116. Warton K, Lin V, Navin T, et al. Methylation-capture and Next-Generation
Sequencing of free circulating DNA from human plasma. BMC Genomics.
2014;15(1):476. doi:10.1186/1471-2164-15-476
117. Duncavage EJ, Bagg A, Hasserjian RP, et al. Genomic profiling for clinical
decision making in myeloid neoplasms and acute leukemia. Blood.
2022;140(21):2228-2247. doi:10.1182/blood.2022015853
118. Duncavage EJ, Schroeder MC, O’Laughlin M, et al. Genome Sequencing as
an Alternative to Cytogenetic Analysis in Myeloid Cancers. N Engl J Med.
2021;384(10):924-935. doi:10.1056/NEJMoa2024534
119. Martínez-Jiménez F, Movasati A, Brunner SR, et al. Pan-cancer wholegenome
comparison of primary and metastatic solid tumours. Nature.
2023;618(7964):333-341. doi:10.1038/s41586-023-06054-z
120. Rosenquist R, Cuppen E, Buettner R, et al. Clinical utility of whole-genome
sequencing in precision oncology. Semin Cancer Biol. 2022;84:32-39.
doi:10.1016/j.semcancer.2021.06.018
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