Novaseqx Applications Ebook M GL 01796

Download as pdf or txt
Download as pdf or txt
You are on page 1of 30

Unlock the next wave

of genomic discovery

Broader, deeper, multiomic sequencing


enabled on the NovaSeq™ X Series

For Research Use Only. Not for use in diagnostics procedures.


UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Table of contents

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

2 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

TABLE OF
BIGGER STUDIES
CONTENTS
3 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

COST-PER-GENOME
Dramatic decrease in sequencing costs

$100 M

$2 M
Cost (USD) per human genome

Genome Analyzer™ II
$100,000
$3000/Gb
0.3 Gb/day
$10,000

$1000

NovaSeq X Series
$600 $2/Gb
6000 Gb/day

$200

2001 2003 2005 2007 2009 2011 2013 2015 2017 2019 2021 2023

Year

What if this breakneck pace of advancement was from to


applied to air travel? A hypothetical flight from
$2000 | 6 hours NY Paris $1.33 | 1 Sec
New York City to Paris, France could decrease

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.

TABLE OF
BIGGER STUDIES
CONTENTS
4 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

TABLE OF
BIGGER STUDIES
CONTENTS
5 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Bigger studies to improve


statistical power


Increase statistical power from larger sample sizes
and more diverse cohorts
A measure of the success
of our mission will be
Even decades after the first human genomes were
sequenced, a majority of gene–disease connections whether we can convince
remain a mystery. To understand the significance of
human genetic variation in relation to complex disease,
similar healthcare
genome-wide association studies (GWAS) aim to systems around the world
connect genotype to phenotype. Most GWAS have
been performed using microarray genotyping. As the to adopt approaches
cost of genome sequencing drops, researchers can
generate higher quality, more comprehensive sequence
developed within the
data at population scale. A shared global knowledge base 100,000 Genomes Project
that integrates genomic data with longitudinal health
records is needed to clearly identify key genes and to transform the application
disease mechanisms.
of genomic medicine in
healthcare and bring
Large-cohort data at work


better outcomes to
The 100,000 Genomes Project in the United Kingdom patients worldwide.
(UK), supported by Genomics England, is the archetype
for integrating genomics with a large-scale health system Mark Caulfield, Chief Scientist,
and demonstrates what routine, genomic-informed Genomics England
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

TABLE OF PACE OF BROADER


CONTENTS DISCOVERY STUDIES
6 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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

Population genomics projects across the globe with numbers of participants

United Kingdom6 United Kingdom1


5 million 100,000

United Kingdom4
500,000 France13
1 million
Ireland 14
Denmark16
400,000
50,000
Finland17
Iceland15
500,000
200,000
Estonia18
100,000
Turkey19
100,000

United Arab
Emirates20
1 million

Japan23
Saudi Arabia21 100,000
100,000
Mexico 27 Asia10,11
Brazil28 100,000
1 million
7000
Israel22
100,000
United States29 Hong Kong24
1 million Africa7 Singapore12 50,000
United States31 118,000+ 100,000
1 million
United States30 Australia8 China25
1 million 200,000 100,000
United States 32
Australia9 China26
100,000
7000 100,000

Many of these efforts focus on increasing the diversity to train artificial intelligence to use information from
in the pools of genome data to represent underserved natural selection to better classify variants of unknown
groups,7–12,19–27,31 because, as of 2021, 86% of genomics significance (VUS).35,36 Together, these efforts will help
studies have been conducted on individuals of European us realize a more complete view of the genome for all
descent.33 Additional tools are helping draw out the full populations and develop ways to stop disease outbreaks,
value of these large-cohort projects. The “pan genome” decrease early deaths due to sudden heart attacks, and
reference genome includes 47 diverse references to reduce the strain of chronic diseases like Alzheimer’s
uplevel variant calling.34 The Primate AI-3D resource and diabetes.
mines data from other primate genomes (233 species)

Learn more

Population genomics
TABLE OF PACE OF BROADER
CONTENTS DISCOVERIES STUDIES
7 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

Whole-genome sequencing Whole-exome sequencing Targeted sequencing

Sequencing region Coverage Sequencing region Coverage Sequencing region Coverage


Entire genome ~30×* Protein-coding regions Specific genes or regions
~50×–100×* > 500×*
(~2% of the genome) of interest

Pros Cons Cons Cons


Pros Pros
Captures all DNA variants in Higher per-sample cost, Reliable detection of Manipulation of the sample, Resources focused on Limited resolution
the least biased way more demanding analysis coding variants misses 98% of the genome specific areas of interest

* Coverage requirements vary depending on use-case, such as rarity of factor being measured.

Beyond the exome

Structural variation Mitochondrial variants

Exome: 2% Noncoding genome: 98% Repeat expansions Noncoding SNVs and indels

Copy number variants 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

TABLE OF BIGGER MULTIOMIC


CONTENTS STUDIES STUDIES
8 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Value of whole genomes


When NGS is more accessible, studies can cast a are frequently key biomarkers for disease. Unlike other
“wider net” to survey more genes. For example, labs methods, WGS robustly captures all common variant
that routinely do exome sequencing can now afford types.37–39,44 WGS captures copy number variants (CNVs)
to switch to WGS. WGS offers multiple advantages for with greater resolution than chromosomal microarrays
finding variants. WGS examines the entire genome and (CMAs).37,43,45,46 WGS also captures some variants in
has the capability to assess variants in both coding and exomes with greater accuracy than whole-exome
noncoding regions of the genome.37–43 Noncoding variants sequencing (WES).2,3,37,44,46–49

WGS provides the most comprehensive analysis of genomic variant types†

Capable Limited capabilities

Sanger Targeted NGS PCR FISH Karyotype CMA WES WGS

Singie nucieotide
variants (SNVs)37

Insertion-deletions
(Indels)

Copy numbers variants


(CNVs)37,50

Repeat
expansions39,40,51,52

Structural variants
(SVs)38,48

Mitochondria37

Paralogs41

† 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.

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

TABLE OF BIGGER MULTIOMIC


CONTENTS STUDIES STUDIES
9 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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 whole-
genome 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

TABLE OF BIGGER MULTIOMIC


CONTENTS STUDIES STUDIES
10 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

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.

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.

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.

TABLE OF BROADER HIGH-RESOLUTION


CONTENTS STUDIES STUDIES
11 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Common multiomic combinations


Genomics + transcriptomes ties between gene regulation and gene expression,
instead of simply inferring those connections. Integrating
epigenetics and RNA-Seq can help researchers identify
GWAS have successfully identified genetic variants
candidate genes and understand the mechanisms
associated with complex diseases. The genotype offers
controlling interesting phenotypes. This holistic, non-
information on susceptibility to the disease; however,
biased multiomics approach can uncover new regulatory
determining the specific genes and pathways affected
elements for biomarkers and therapeutic targets.
by those variants is more difficult. Incorporating
RNA-Seq can help researchers annotate and prioritize
variants uncovered in GWAS for functional analysis to Transcriptomics + proteomics
understand mechanisms of disease. Gene expression
analysis informs if and when the genes of interest are RNA-Seq offers unparalleled discovery power to
down- or upregulated in the disease samples. This interrogate the transcriptome without prior knowledge.
multiomic approach to functional genomics can help Incorporating protein detection with RNA-Seq can tie
power drug target identification and biomarker discovery. new discoveries back to known canonical markers
and historical clinical outcomes. Antibodies tagged
Genomics + epigenetics with oligonucleotide barcodes enable analysis of cell
surface proteins with results read by sequencing, which
scales to a much higher number of parameters than
The majority of human variation identified by GWAS is in
flow cytometry or mass cytometry. Single-stranded
the noncoding regions of the genome, including introns,
nucleotide aptamers with selective protein binding
promoters, or enhancers. Comprehensive epigenetic
targets can be used in a similar way. Methods like
profiling can reveal patterns of gene regulation to help
cellular indexing of transcriptomes and epitopes by
find the function of those variants. NGS-based epigenetic
sequencing (CITE-Seq) combine single-cell RNA-Seq
techniques include chromatin immunoprecipitation
with cell surface protein analysis. Bulk epitope and
(ChIP-Seq), assay for transposase-accessible chromatin
nucleic acid sequencing (BEN-Seq) is performed at the
(ATAC-Seq), and chromosome conformation capture (HiC).
bulk level. Spatial transcriptomics interrogates RNA and
DNA methylation patterns are also conserved and can
proteins in context with tissue morphology.
represent a new class of biomarkers. Multiomic approaches
that combine methylation, or other epigenetic profiling,
with genetic information can connect functional layers to Genomics + proteomics
decipher complex pathways and disease mechanisms.
This multiomic approach directly connects genotype to
Epigenetics + transcriptomics phenotype for more informed research on disease and
therapeutics development. Linking genetic variation
to protein expression at the single-cell level can reveal
Epigenetics and transcriptomics offer complementary
the functional impact of somatic mutations on human
information to study the details of cellular differentiation
cancers to better understand tumor evolution and
and response. Combining epigenetic and RNA-Seq
disease progression.
methods allows researchers to directly measure the

TABLE OF BROADER HIGH-RESOLUTION


CONTENTS STUDIES STUDIES
12 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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 population-
scale 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 oligo-
tagged 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

TABLE OF BROADER HIGH-RESOLUTION


CONTENTS STUDIES STUDIES
13 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

TABLE OF MULTIOMIC DEEPER


CONTENTS STUDIES STUDIES
14 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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 scRNA-
Seq 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

TABLE OF MULTIOMIC DEEPER


CONTENTS STUDIES STUDIES
15 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Deeper studies to find rare events


NGS assays can be designed to either target a large However, recent improvements in NGS instrumentation
number of genes with low sequencing depth (more provide options for sequencing samples at extremely
comprehensive, less sensitive) or a relatively small high depth of coverage for large portions of the genome
number of genes with higher sequencing depth in a single sample. This enables analysis of hundreds
(less comprehensive, more sensitive). In cases of of genes with the sequencing depth required for ctDNA
heterogenous samples such as blood or tumors, high analysis. Furthermore, these technologies enable
sequencing depth is necessary to provide the sensitivity hypothesis-free interrogation of biofluid analytes,
required to detect low-abundance variants accurately. providing discovery power to assay genes and pathways
Scientists can take advantage of the latest advancements that were not considered prior to experimental design.
in high-throughput sequencing to perform deeper
sequencing experiments, more rapidly, at production-scale. Liquid biopsy combined with comprehensive
genomic profiling (CGP) can identify tumor-specific

Liquid biopsy mutations in patient samples93 and provide information


of cancer type, stage, and vascularization.94–97 Recent
studies that performed liquid biopsy paired with
Liquid biopsy detects and characterizes various tumor- corresponding tissue biopsy from tumor samples have
derived biomarkers present in the blood of an individual demonstrated that, when comprehensive assays are
with cancer. These biomarkers are absent in healthy used, ctDNA analysis detected a significant number of
individuals and patients who are cancer free. Liquid guideline-recommended biomarkers and resistance
biopsy is a noninvasive approach that offers a potential alterations not found in matched tissue biopsies.98,99
alternative to invasive tissue biopsies to detect targetable Furthermore, ctDNA sequencing identifies variants in
oncogenic drivers and resistance mutations.89 the tumor from which it originates, including both driver
and passenger mutations, respectively.90 Given the rapid
Tumors release circulating tumor DNA (ctDNA) into turnover of cfDNA and ctDNA in the bloodstream, liquid
the bloodstream through various cellular mechanisms, biopsy can be a powerful tool for cancer research to
including apoptosis, necrosis, phagocytosis, and active assay tumor burden in real time and monitor response to
secretion.90 Even so, ctDNA represents a small fraction therapy.100 Studies also demonstrate how liquid biopsy-
of total cell-free DNA (cfDNA) in the blood,91 the majority based detection of molecular residual disease (MRD) can
of which is released by erythrocytes, leukocytes, and predict recurrence with high sensitivity and specificity,
endothelial cells.90,92 The rarity of ctDNA combined with with lead times that precede standard imaging.101–103
the fact that different cancer types at different stages
shed ctDNA at different rates complicates analysis.

TABLE OF HIGH-RESOLUTION NOVASEQ X


CONTENTS STUDIES SERIES
16 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Next-level liquid biopsy


Circulating analytes include more than just ctDNA and can
apply to more than just cancer. As technology continues to
We've known for many
advance, there is the potential to apply multiomic approaches years that there is tumor-
that integrate analyses of the genome, transcriptome,
epigenome, proteome, and microbiome to liquid biopsy.104 related material in the
For example, cell-free RNA (cfRNA) is indicative of early-
stage cancer and other conditions like preeclampsia.105–111
bloodstream, we just
Methylation patterns in ctDNA can also recapitulate the didn't have the
abnormal methylation patterns that are a hallmark of many


cancers.112–116 The ability to combine multiomics with liquid technologies to detect
biopsy will provide unique discovery power for deeper insights
and comprehensive answers to the mechanisms of cancer.
it for it to be meaningful.
Minetta C. Liu, MD, Mayo Clinic

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

TABLE OF HIGH-RESOLUTION NOVASEQ X


CONTENTS STUDIES SERIES
17 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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

TABLE OF DEEPER
WORKFLOWS
CONTENTS STUDIES
18 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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.

EXAMPLE PROJECT
Single-cell gene expression study with fixed $50K reagent budget‡

450

400
~2×
350
Single-cell samples per $50K reagent budget

300

250

200

150

100

50

25B 10B 1.5B S4 S2 S1 SP NextSeqTM

NovaSeq X Series NovaSeq 6000 System 2000 P3

‡ A nalysis based on 20,000 reads per cell and 10,000 cells per sample.
All pricing is in USD, based on United States list prices.

TABLE OF DEEPER
WORKFLOWS
CONTENTS STUDIES
19 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Setting a new bar in every sequencing metric

NovaSeq 6000 System NovaSeq X Series

65 Gb–6 Tb Output 165 Gb–16 Tb

~2.5×
Throughput
800M–20B Read number 1.68B–52B

~1 genome/hr Genomes per hour


~2×
~2.5 genomes/hr

Faster

2–48 genomes Genomes per run 4–128 genomes

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.

$183
SHRINKING THE GAP
The NovaSeq X Series decreases cost/Gb
WGS and WES by up to 50% and shrinks the cost gap between
WGS and WES by 2.5× to a difference of only
$183 (USD).

Sequencing cost

Library prep cost


WGS WES

TABLE OF DEEPER
WORKFLOWS
CONTENTS STUDIES
20 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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 hard-
to-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.

TABLE OF DEEPER
WORKFLOWS
CONTENTS STUDIES
21 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Designed with the user in mind

4K ultra System prompts


high-definition to guide user
touch screen along workflow

Adjustable screen height with position memory

Easy-to-handle flow cell with illuminated flow cell stage

Less time and effort with established


workflow applications

Push-to-access hidden keyboard


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
Light-guided consumable loading plastic for easier handling and reduced
disposal costs§

§ In comparison to the NovaSeq 6000 System.

Assisted open doors and waist-height reagent drawers


TABLE OF DEEPER
WORKFLOWS
CONTENTS STUDIES
22 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY


Integrated, cohesive bioinformatics ecosystem The physical reduction in kit
The NovaSeq X Series streamlines genomic data management size has greatly decreased
with onboard secondary analysis and integration with a our cold storage needs
comprehensive suite of bioinformatics solutions. The
Illumina Connected Software suite includes some of the in lab and elimination of
fastest, most accurate,94 and advanced solutions for data
analysis, interpretation, and aggregation. With flexible local dry ice shipping has made


and cloud-based options for lab operations, run planning, unpacking larger orders
and data analysis, the NovaSeq X Series enables users to
run high-throughput sequencing without creating a much easier.
bioinformatics bottleneck.
Eric Chow, Assistant Adjunct Professor
and Director, University of California
Plug-and-play onboard DRAGEN workflows enable variant San Francisco (UCSF) Center for Advanced
calling directly on the system. Up to four DRAGEN applications Technology
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, One of the big benefits
more data-intensive sequencing projects.
I can see in the new
DRAGEN onboard analysis
NovaSeq X Series software ecosystem technical note on the NovaSeq X Plus
System is the considerable
reduction in file size now
** Assuming FASTQ files are archived 30 days after cloud upload
and throughput of 10,000 WGS/year for the NovaSeq X System and being compressed to
20,000 WGS/year for the NovaSeq X Plus System.
approximately one-fifth of
what would be a traditional
file, which allows us to


run more projects at
population scale.
Lachlan Morrison, Senior Technical Officer
Biomolecular Resource Facility (BRF),
Australian National University (ANU)

TABLE OF DEEPER
WORKFLOWS
CONTENTS STUDIES
23 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Example NovaSeq X Series workflows


WGS

Prepare libaries Sequence Analyze data

Illumina DNA
NovaSeq X 1.5B flow cell DRAGEN Germline
PCR-Free Prep
~4 samples per flow cell (onboard or in the cloud)
25–300 ng DNA input
NovaSeq X 10B flow cell
~1.5-hour workflow
~24 samples per flow cell

NovaSeq X 25B flow cell


~64 samples per flow cell

400M reads per sample


2 × 150 bp read length

Illumina Complete Long


Read Prep, Human NovaSeq X 10B flow cell Illumina Complete
4 samples per flow cell Long Read WGS App
50 ng DNA input (in the cloud)

NovaSeq X 25B flow cell


~1-day workflow
10–11 samples per flow cell

750 Gb per sample


2 × 150 bp read length

WES

Prepare libaries Sequence Analyze data

Illumina DNA Prep with


NovaSeq X 1.5B flow cell DRAGEN Enrichment
Exome 2.0 Plus Enrichment
~41 samples per flow cell (onboard or in the cloud)
50–1000 ng DNA input
NovaSeq X 10B flow cell
~6.5-hour workflow
~250 samples per flow cell

NovaSeq X 25B flow cell


~750 samples per flow cell

8 Gb per sample
2 × 100 bp read length

TABLE OF NOVASEQ X
CONCLUSION
CONTENTS SERIES
24 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

WTS

Prepare libaries Sequence Analyze data

Illumina Stranded Total


NovaSeq X 1.5B flow cell DRAGEN RNA
RNA Prep
~32 samples per flow cell (onboard or in the cloud)
1–1000 ng high-quality
DNA input NovaSeq X 10B flow cell
~200 samples per flow cell
~7-hour workflow
NovaSeq X 25B flow cell
~520 samples per flow cell

50M reads per sample


2 × 100 bp read length

Illumina Stranded
NovaSeq X 1.5B flow cell DRAGEN RNA
mRNA Prep
~64 samples per flow cell (onboard or in the cloud)
25–1000 ng high-quality
RNA input NovaSeq X 10B flow cell
~400 samples per flow cell
~7-hour workflow
NovaSeq X 25B flow cell
~1040 samples per flow cell

25M reads per sample


2 × 100 bp read length

Illumina RNA Prep


NovaSeq X 1.5B flow cell DRAGEN RNA
with Enrichment
~64 samples per flow cell (onboard or in the cloud)
20 ng FFPE RNA input
NovaSeq X 10B flow cell
~9-hour workflow ~400 samples per flow cell

NovaSeq X 25B flow cell


~1040 samples per flow cell

25M reads per sample


2 × 100 bp read length

TABLE OF NOVASEQ X
CONCLUSION
CONTENTS SERIES
25 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

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
“ [The NovaSeq X Series]
now allows us to provide
much larger scale projects
variants. Answer the most complex questions in human for both human health and
genomics, with larger sample cohorts, deeper sequencing,
and more data-intensive methods—from whole-genome
nonhuman scenarios in
sequencing to multiomics. the research world, and
Genomics visionaries are ready to usher in the Genome Era, to be able to translate
that into more meaningful


and the NovaSeq X Series can help make it happen.

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

TABLE OF
WORKFLOWS ABBREVIATIONS
CONTENTS
26 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

Abbreviations
ATAC-Seq: assay for transposase-accessible chromatin Indel: insertion–deletion

BEN-Seq: bulk epitope and nucleic acid sequencing MRD: molecular residual disease

CGP: comprehensive genomic profiling NGS: next-generation sequencing

cfDNA: cell-free DNA NICU: neonatal intensive care unit

ChIP-Seq: chromatin immunoprecipitation sequencing RNA-Seq: RNA sequencing

CITE-Seq: cellular indexing of transcriptomes and SBS: sequencing by synthesis


epitopes by sequencing
scRNA-Seq: single-cell RNA sequencing
CMA: chromosomal microarray
SNV: single nucleotide variant
CNV: copy number variant
SV: structural variant
CRISPR: clustered regularly interspaced short palindromic
repeats Tb: terabase

ctDNA: circulating tumor DNA VUS: variant of unknown significance

eQTL: expression quantitative trait loci WES: whole-exome sequencing

FFPE: formalin-fixed, paraffin-embedded WGS: whole-genome sequencing

Gb: gigabase WGTS: combined whole-genome and transcriptome


sequencing
GWAS: genome-wide association studies
WTS: whole-transcriptome sequencing
HiC: chromosome conformation capture

TABLE OF
CONCLUSION REFERENCES
CONTENTS
27 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

References
1. 100,000 Genomes Project. Genomics England. genomicsengland.co.uk/ 27. Jaber IG. TecScience. OriGen Project seeks to solve the enigma of the Mexican
initiatives/100000-genomes-project. Accessed June 19, 2023. genome. ecscience.tec.mx/en/health/mexican-genome/. Published February 15,
2. 100,000 Genomes Project Pilot Investigators, Smedley D, Smith KR, et al. 100,000 2023. Accessed September 1, 2023.
Genomes Pilot on Rare-Disease Diagnosis in Health Care - Preliminary Report. 28. Coelho AVC, Mascaro-Cordeiro B, Lucon DR, et al. The Brazilian Rare Genomes
N Engl J Med. 2021;385(20):1868-1880. doi:10.1056/NEJMoa2035790 Project: Validation of Whole Genome Sequencing for Rare Diseases Diagnosis. Front
3. Turro E, Astle WJ, Megy K, et al. Whole-genome sequencing of patients with rare Mol Biosci. 2022;9:821582. doi:10.3389/fmolb.2022.821582
diseases in a national health system. Nature. 2020;583(7814):96-102. doi:10.1038/ 29. All of Us Research Program. National Institutes of Health (NIH). allofus.nih.gov/.
s41586-020-2434-2 Accessed September 1, 2023.
4. UK Biobank. ukbiobank.ac.uk/. Accessed September 1, 2023. 30. Million Veteran Program. US Department of Veteran Affairs. research.va.gov/mvp/.
5. Degasperi A, Zou X, Dias Amarante T, et al. Substitution mutational signatures Accessed September 1, 2023.
in whole-genome–sequenced cancers in the UK population. Science. 31. Precision Medicine Initiative (PMI) Cohort Program. National Institute on Minority
2022;376(6591):abl9283. doi:10.1126/science.abl9283 Health and Health Disparities. nimhd.nih.gov/programs/collab/pmi/. Accessed
6. Our Future Health. ourfuturehealth.org.uk/. Accessed June 19, 2023. September 1, 2023.
7. H3Africa – Human Heredity & Health in Africa. h3africa.org/. Accessed June 19, 32. Murray M. CT Forum on DNA Testing and Genomics Medicine. Yale Center for
2023. Genomic Health. Generations Project and DNA-based Health Screening. Published
8. Care AGD of H and A. Genomics Health Futures Mission. Australian Government January 23, 2020. Accessed September 1, 2023.
Department of Health and Aged Care. health.gov.au/our-work/genomics-health- 33. Fatumo S, Chikowore T, Choudhury A, Ayub M, Martin AR, Kuchenbaecker K.
futures-mission. Published June 2, 2023. Accessed June 19, 2023. A roadmap to increase diversity in genomic studies. Nat Med. 2022;28(2):243-250.
9. Australia begins a big push to sequence underserved populations. illumina.com/ doi:10.1038/s41591-021-01672-4
content/illumina-marketing/amr/en_US/company/news-center/feature-articles/ 34. Liao WW, Asri M, Ebler J, et al. A draft human pangenome reference. Nature.
australia-begins-a-big-push-to-sequence-underserved-populations.html. 2023;617(7960):312-324. doi:10.1038/s41586-023-05896-x
Accessed June 19, 2023. 35. What our primate relatives can teach us about our own genomes. illumina.com/
10. GenomeAsia 100k. genomeasia100k.org/index.html. Accessed June 19, 2023. content/illumina-marketing/amr/en_US/company/news-center/feature-articles/
11. Wall JD, Stawiski EW, Ratan A, et al. The GenomeAsia 100K Project enables genetic PrimateAI-3D.html. Accessed June 21, 2023.
discoveries across Asia. Nature. 2019;576(7785):106-111. doi:10.1038/s41586-019- 36. Improving genetic risk prediction and drug target discovery using primate
1793-z DNA and advanced artificial intelligence. illumina.com/content/illumina-marketing/
12. Precision Health Research, Singapore (PRECISE). npm.sg/. Accessed June 16, 2023. amr/en_US/science/genomics-research/articles/primateai-3d.html. Accessed
13. Lethimonnier F, Levy Y. Genomic medicine France 2025. Ann Oncol. June 21, 2023.
2018;29(4):783-784. doi:10.1093/annonc/mdy027 37. Lionel AC, Costain G, Monfared N, et al. Improved diagnostic yield compared with
14. Petrone J. Ireland outlines national strategy to streamline access to genomic targeted gene sequencing panels suggests a role for whole-genome sequencing as
medicine. GenomeWeb. genomeweb.com/research-funding/ireland-outlines- a first-tier genetic test. Genet Med Off J Am Coll Med Genet. 2018;20(4):435-443.
national-strategy-streamline-access-genomic-medicine. Published February 2, doi:10.1038/gim.2017.119
2023, Accessed September 1, 2023. 38. Chen X, Schulz-Trieglaff O, Shaw R, et al. Manta: rapid detection of structural
15. Gudbjartsson DF, Helgason H, Gudjonsson SA, et al. Large-scale whole-genome variants and indels for germline and cancer sequencing applications.
sequencing of the Icelandic population. Nat Genet. 2015;47(5):435-444. Bioinforma Oxf Engl. 2016;32(8):1220-1222. doi:10.1093/bioinformatics/btv710
doi:10.1038/ng.3247 39. Dolzhenko E, van Vugt JJFA, Shaw RJ, et al. Detection of long repeat expansions
16. Apol KD, Lydersen LN, Mortensen Ó, et al. FarGen - participants in the genetic from PCR-free whole-genome sequence data. Genome Res. 2017;27(11):1895-1903.
research infrastructure of the Faroe Islands. Scand J Public Health. 2022;50(7):980- doi:10.1101/gr.225672.117
987. doi:10.1177/14034948211046817 40. Alfares A, Aloraini T, Subaie LA, et al. Whole-genome sequencing offers additional
17. Kurki MI, Karjalainen J, Palta P, et al. FinnGen provides genetic insights from a well- but limited clinical utility compared with reanalysis of whole-exome sequencing.
phenotyped isolated population. Nature. 2023;613(7944):508-518. doi:10.1038/ Genet Med Off J Am Coll Med Genet. 2018;20(11):1328-1333. doi:10.1038/
s41586-022-05473-8 gim.2018.41
18. Estonian Biobank. University of Tartu. Institute of Genomics. genomics.ut.ee/en/ 41. Chen X, Sanchis-Juan A, French CE, et al. Spinal muscular atrophy diagnosis
content/estonian-biobank. Published November 29, 2021. Accessed September 1, and carrier screening from genome sequencing data. Genet Med Off J Am Coll Med
2023. Genet. 2020;22(5):945-953. doi:10.1038/s41436-020-0754-0
19. BBMRI-ERIC. Turkish Genome Project Launched. bbmri-eric.eu/news-events/ 42. Sanghvi RV, Buhay CJ, Powell BC, et al. Characterizing reduced coverage regions
turkish-genome-project-launched. Accessed September 1, 2023. through comparison of exome and genome sequencing data across 10 centers.
20. The National. UAE President launches National Genome Strategy to prioritise Genet Med Off J Am Coll Med Genet. 2018;20(8):855-866. doi:10.1038/gim.2017.192
Emirati health care. Published March 28, 2023. Accessed September 1, 2023. 43. Lindstrand A, Eisfeldt J, Pettersson M, et al. From cytogenetics to cytogenomics:
21. Saudi Genome Program. vision2030.gov.sa/v2030/v2030-projects/saudi-genome- whole-genome sequencing as a first-line test comprehensively captures the
program/. Accessed September 1, 2023. diverse spectrum of disease-causing genetic variation underlying intellectual
22. Gilmore J. Israel to Sequence 100K People, Create Genomic Database to Support disability. Genome Med. 2019;11(1):68. doi:10.1186/s13073-019-0675-1
'Digital Health'. GenomeWeb. genomeweb.com/sequencing/israel-sequence-100k- 44. Meienberg J, Bruggmann R, Oexle K, Matyas G. Clinical sequencing: is WGS the
people-create-genomic-database-support-digital-health. Published December 13, better WES? Hum Genet. 2016;135(3):359-362. doi:10.1007/s00439-015-1631-9
2018. Accessed September 1, 2023. 45. Trost B, Walker S, Wang Z, et al. A Comprehensive Workflow for Read Depth-Based
23. Takahashi Y, Mizusawa H. Initiative on Rare and Undiagnosed Disease in Japan. Identification of Copy-Number Variation from Whole-Genome Sequence Data. Am J
JMA J. 2021;4(2):112-118. doi:10.31662/jmaj.2021-0003 Hum Genet. 2018;102(1):142-155. doi:10.1016/j.ajhg.2017.12.007
24. Hong Kong Genome Institute. hkgp.org/en/. Accessed September 1, 2023. 46. Stavropoulos DJ, Merico D, Jobling R, et al. Whole Genome Sequencing
25. Illumina and China’s Children’s Hospital of Fudan Bring Rapid WGS to the NICU. Expands Diagnostic Utility and Improves Clinical Management in Pediatric
illumina.com/content/illumina-marketing/amr/en_US/company/news-center/feature- Medicine. NPJ Genomic Med. 2016;1:15012-. doi:10.1038/npjgenmed.2015.12
articles/illumina-and-children-s-hospital-of-fudan-in-china.html. Published April 22, 47. Belkadi A, Bolze A, Itan Y, et al. Whole-genome sequencing is more powerful
2019. Accessed September 1, 2023. than whole-exome sequencing for detecting exome variants. Proc Natl Acad
26. CGTN. China Genome Project: China launches 100,000 Genomes Project. Published Sci U S A. 2015;112(17):5473-5478. doi:10.1073/pnas.1418631112
December 29, 2017. Accessed September 1, 2023.

TABLE OF
ABBREVIATIONS
CONTENTS
28 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

48. Carss KJ, Arno G, Erwood M, et al. Comprehensive Rare Variant Analysis 69. Võsa U, Claringbould A, Westra HJ, et al. Large-scale cis- and trans-eQTL analyses
via Whole-Genome Sequencing to Determine the Molecular Pathology of identify thousands of genetic loci and polygenic scores that regulate blood gene
Inherited Retinal Disease. Am J Hum Genet. 2017;100(1):75-90. doi:10.1016/j. expression. Nat Genet. 2021;53(9):1300-1310. doi:10.1038/s41588-021-00913-z
ajhg.2016.12.003 70. Colin E, Duffourd Y, Tisserant E, et al. OMIXCARE: OMICS technologies solved
49. Souche E, Beltran S, Brosens E, et al. Recommendations for whole genome about 33% of the patients with heterogeneous rare neuro-developmental disorders
sequencing in diagnostics for rare diseases. Eur J Hum Genet EJHG. and negative exome sequencing results and identified 13% additional candidate
2022;30(9):1017-1021. doi:10.1038/s41431-022-01113-x variants. Front Cell Dev Biol. 2022;10:1021785. doi:10.3389/fcell.2022.1021785
50. Gross AM, Ajay SS, Rajan V, et al. Copy-number variants in clinical genome 71. Kamali Z, Keaton JM, Haghjooy Javanmard S, et al. Large-Scale Multi-Omics
sequencing: deployment and interpretation for rare and undiagnosed disease. Studies Provide New Insights into Blood Pressure Regulation. Int J Mol Sci.
Genet Med Off J Am Coll Med Genet. 2019;21(5):1121-1130. doi:10.1038/s41436- 2022;23(14):7557. doi:10.3390/ijms23147557
018-0295-y 72. Lunke S, Bouffler SE, Patel CV, et al. Integrated multi-omics for rapid rare disease
51. van Kuilenburg ABP, Tarailo-Graovac M, Richmond PA, et al. Glutaminase diagnosis on a national scale. Nat Med. 2023:1-11. doi:10.1038/s41591-023-02401-9
Deficiency Caused by Short Tandem Repeat Expansion in GLS. N Engl J Med. 73. Longo SK, Guo MG, Ji AL, Khavari PA. Integrating single-cell and spatial
2019;380(15):1433-1441. doi:10.1056/NEJMoa1806627 transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet.
52. Ibañez K, Polke J, Hagelstrom RT, et al. Whole genome sequencing for the 2021;22(10):627-644. doi:10.1038/s41576-021-00370-8
diagnosis of neurological repeat expansion disorders in the UK: a retrospective 74. Delorey TM, Ziegler CGK, Heimberg G, et al. COVID-19 tissue atlases reveal
diagnostic accuracy and prospective clinical validation study. Lancet Neurol. SARS-CoV-2 pathology and cellular targets. Nature. 2021;595(7865):107-113.
2022;21(3):234-245. doi:10.1016/S1474-4422(21)00462-2 doi:10.1038/s41586-021-03570-8
53. Splinter K, Adams DR, Bacino CA, et al. Effect of Genetic Diagnosis on Patients 75. Desai N, Neyaz A, Szabolcs A, et al. Temporal and spatial heterogeneity of host
with Previously Undiagnosed Disease. N Engl J Med. 2018;379(22):2131-2139. response to SARS-CoV-2 pulmonary infection. Nat Commun. 2020;11(1):6319.
doi:10.1056/NEJMoa1714458 doi:10.1038/s41467-020-20139-7
54. Holliday M, Singer ES, Ross SB, et al. Transcriptome Sequencing of Patients 76. Scheid JF, Barnes CO, Eraslan B, et al. B cell genomics behind cross-
With Hypertrophic Cardiomyopathy Reveals Novel Splice-Altering Variants neutralization of SARS-CoV-2 variants and SARS-CoV. Cell. 2021;184(12):3205-
in MYBPC3. Circ Genomic Precis Med. 2021;14(2):e003202. doi:10.1161/ 3221.e24. doi:10.1016/j.cell.2021.04.032
CIRCGEN.120.00320241.  77. Huang N, Pérez P, Kato T, et al. SARS-CoV-2 infection of the oral cavity
55. Zhu F, Zhang F, Hu L, Liu H, Li Y. Integrated Genome and Transcriptome Sequencing and saliva. Nat Med. 2021;27(5):892-903. doi:10.1038/s41591-021-01296-8
to Solve a Neuromuscular Puzzle: Miyoshi Muscular Dystrophy and Early Onset 78. Butler D, Mozsary C, Meydan C, et al. Shotgun transcriptome, spatial omics, and
Primary Dystonia in Siblings of the Same Family. Front Genet. 2021;12:672906. isothermal profiling of SARS-CoV-2 infection reveals unique host responses,
doi:10.3389/fgene.2021.672906 viral diversification, and drug interactions. Nat Commun. 2021;12(1):1660.
56. Karam R, Conner B, LaDuca H, et al. Assessment of Diagnostic Outcomes of RNA doi:10.1038/s41467-021-21361-7
Genetic Testing for Hereditary Cancer. JAMA Netw Open. 2019;2(10):e1913900. 79. Zhang Y, Narayanan SP, Mannan R, et al. Single-cell analyses of renal cell
doi:10.1001/jamanetworkopen.2019.13900 cancers reveal insights into tumor microenvironment, cell of origin, and therapy
57. Pon JR, Marra MA. Driver and passenger mutations in cancer. Annu Rev Pathol. response. Proc Natl Acad Sci U S A. 2021;118(24):e2103240118. doi:10.1073/
2015;10:25-50. doi:10.1146/annurev-pathol-012414-040312 pnas.2103240118
58. Golub TR, Slonim DK, Tamayo P, et al. Molecular classification of cancer: 80. Peng M, Wei G, Zhang Y, et al. Single-cell transcriptomic landscape reveals the
class discovery and class prediction by gene expression monitoring. Science. differences in cell differentiation and immune microenvironment of papillary
1999;286(5439):531-537. doi:10.1126/science.286.5439.531 thyroid carcinoma between genders. Cell Biosci. 2021;11(1):39. doi:10.1186/
59. Valk PJM, Verhaak RGW, Beijen MA, et al. Prognostically useful gene-expression s13578-021-00549-w
profiles in acute myeloid leukemia. N Engl J Med. 2004;350(16):1617-1628. 81. Lalli MA, Avey D, Dougherty JD, Milbrandt J, Mitra RD. High-throughput single-
doi:10.1056/NEJMoa040465 cell functional elucidation of neurodevelopmental disease-associated genes
60. Docking TR, Parker JDK, Jädersten M, et al. A clinical transcriptome approach reveals convergent mechanisms altering neuronal differentiation. Genome Res.
to patient stratification and therapy selection in acute myeloid leukemia. 2020;30(9):1317-1331. doi:10.1101/gr.262295.120
Nat Commun. 2021;12(1):2474. doi:10.1038/s41467-021-22625-y 82. Brady L, Kriner M, Coleman I, et al. Inter- and intra-tumor heterogeneity of
61. Lilljebjörn H, Orsmark-Pietras C, Mitelman F, Hagström-Andersson A, metastatic prostate cancer determined by digital spatial gene expression
Fioretos T. Transcriptomics paving the way for improved diagnostics and precision profiling. Nat Commun. 2021;12(1):1426. doi:10.1038/s41467-021-21615-4
medicine of acute leukemia. Semin Cancer Biol. 2022;84:40-49. doi:10.1016/j. 83. Ji AL, Rubin AJ, Thrane K, et al. Multimodal Analysis of Composition and Spatial
semcancer.2021.09.013 Architecture in Human Squamous Cell Carcinoma. Cell. 2020;182(6):1661-1662.
62. Hehir-Kwa JY, Koudijs MJ, Verwiel ETP, et al. Improved Gene Fusion Detection doi:10.1016/j.cell.2020.08.043
in Childhood Cancer Diagnostics Using RNA Sequencing. JCO Precis Oncol. 84. Fawkner-Corbett D, Antanaviciute A, Parikh K, et al. Spatiotemporal analysis of
2022;6:e2000504. doi:10.1200/PO.20.00504 human intestinal development at single-cell resolution. Cell. 2021;184(3):810-
63. Cuppen E, Elemento O, Rosenquist R, et al. Implementation of Whole-Genome 826.e23. doi:10.1016/j.cell.2020.12.016
and Transcriptome Sequencing Into Clinical Cancer Care. JCO Precis Oncol. 85. Cao J, Spielmann M, Qiu X, et al. The single-cell transcriptional landscape of
2022;6:e2200245. doi:10.1200/PO.22.00245 mammalian organogenesis. Nature. 2019;566(7745):496-502. doi:10.1038/
64. Kremer LS, Bader DM, Mertes C, et al. Genetic diagnosis of Mendelian disorders via s41586-019-0969-x
RNA sequencing. Nat Commun. 2017;8:15824. doi:10.1038/ncomms15824 86. Plasschaert LW, Žilionis R, Choo-Wing R, et al. A single-cell atlas of the
65. Cummings BB, Marshall JL, Tukiainen T, et al. Improving genetic diagnosis airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature.
in Mendelian disease with transcriptome sequencing. Sci Transl Med. 2018;560(7718):377-381. doi:10.1038/s41586-018-0394-6
2017;9(386):eaal5209. doi:10.1126/scitranslmed.aal5209 87. BRAIN Initiative Cell Census Network (BICCN). A multimodal cell census and
66. Frésard L, Smail C, Ferraro NM, et al. Identification of rare-disease genes atlas of the mammalian primary motor cortex. Nature. 2021;598(7879):86-102.
using blood transcriptome sequencing and large control cohorts. Nat Med. doi:10.1038/s41586-021-03950-0
2019;25(6):911-919. doi:10.1038/s41591-019-0457-8 88. Replogle JM, Saunders RA, Pogson AN, et al. Mapping information-rich
67. Doke T, Huang S, Qiu C, et al. Transcriptome-wide association analysis identifies genotype-phenotype landscapes with genome-scale Perturb-seq. Cell.
DACH1 as a kidney disease risk gene that contributes to fibrosis. J Clin Invest. 2022;185(14):2559-2575.e28. doi:10.1016/j.cell.2022.05.013
2021;131(10):e141801, 141801. doi:10.1172/JCI141801 89. Li BT, Janku F, Jung B, et al. Ultra-deep next-generation sequencing of
68. Odhams CA, Cunninghame Graham DS, Vyse TJ. Profiling RNA-Seq at multiple plasma cell-free DNA in patients with advanced lung cancers: results from
resolutions markedly increases the number of causal eQTLs in autoimmune disease. the Actionable Genome Consortium. Ann Oncol. 2019;30(4):597-603. doi:10.1093/
PLoS Genet. 2017;13(10):e1007071. doi:10.1371/journal.pgen.1007071 annonc/mdz046

TABLE OF
ABBREVIATIONS
CONTENTS
29 | M-GL-01796 v1.0
UNLOCK THE NEXT WAVE OF GENOMIC DISCOVERY

90. Chin RI, Chen K, Usmani A, et al. Detection of Solid Tumor Molecular Residual 104. Keup C, Suryaprakash V, Storbeck M, Hoffmann O, Kimmig R, Kasimir-Bauer S.
Disease (MRD) Using Circulating Tumor DNA (ctDNA). Mol Diagn Ther. Longitudinal multi-parametric liquid biopsy approach identifies unique features
2019;23(3):311-331. doi:10.1007/s40291-019-00390-5 of circulating tumor cell, extracellular vesicle, and cell-free dna characterization
91. Xu J, Liao K, Yang X, Wu C, Wu W. Using single-cell sequencing technology for disease monitoring in metastatic breast cancer patients. Cells. 2021;10(2):212.
to detect circulating tumor cells in solid tumors. Mol Cancer. 2021;20(1):104. doi:10.3390/cells10020212
doi:10.1186/s12943-021-01392-w 105. Kopreski MS, Benko FA, Gocke CD. Circulating RNA as a tumor marker: detection
92. Grabuschnig S, Bronkhorst AJ, Holdenrieder S, et al. Putative Origins of Cell-Free of 5T4 mRNA in breast and lung cancer patient serum. Ann N Y Acad Sci.
DNA in Humans: A Review of Active and Passive Nucleic Acid Release Mechanisms. 2001;945:172-178.
Int J Mol Sci. 2020;21(21):8062. doi:10.3390/ijms21218062 106. Perhavec A, Cerkovnik P, Novakovic S, Zgajnar J. The hTERT mRNA in plasma
93. van ’t Erve I, Medina JE, Leal A, et al. Metastatic Colorectal Cancer Treatment samples of early breast cancer patients, non-cancer patients and healthy
Response Evaluation by Ultra-Deep Sequencing of Cell-Free DNA and Matched individuals. Neoplasma. 2008;55(6):549-554.
White Blood Cells. Clin Cancer Res. 2023;29(5):899-909. doi:10.1158/1078-0432. 107. Castellanos-Rizaldos E, Zhang X, Tadigotla VR, et al. Exosome-based detection of
CCR-22-2538 activating and resistance EGFR mutations from plasma of non-small cell lung cancer
94. Hillen F, Griffioen AW. Tumour vascularization: sprouting angiogenesis and beyond. patients. Oncotarget. 2019;10(30):2911-2920. doi:10.18632/oncotarget.26885
Cancer Metastasis Rev. 2007;26(3-4):489-502. doi:10.1007/s10555-007-9094-7 108. Cheung KWE, Choi SYR, Lee LTC, et al. The potential of circulating cell free RNA as
95. Bonner ER, Harrington R, Eze A, et al. Circulating tumor DNA sequencing a biomarker in cancer. Expert Rev Mol Diagn. 2019;19(7):579-590. doi:10.1080/1473
provides comprehensive mutation profiling for pediatric central nervous system 7159.2019.1633307
tumors. NPJ Precis Oncol. 2022;6(1):63. doi:10.1038/s41698-022-00306-3 109. Larson MH, Pan W, Kim HJ, et al. A comprehensive characterization of the cell-
96. Nakamura Y, Taniguchi H, Ikeda M, et al. Clinical utility of circulating tumor free transcriptome reveals tissue- and subtype-specific biomarkers for cancer
DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI- detection. Nat Commun. 2021;12(1):2357. doi:10.1038/s41467-021-22444-1
SCREEN and GOZILA studies. Nat Med. 2020;26(12):1859-1864. doi:10.1038/ 110. Munchel S, Rohrback S, Randise-Hinchliff C, et al. Circulating transcripts in maternal
s41591-020-1063-5 blood reflect a molecular signature of early-onset preeclampsia. Sci Transl Med.
97. Bayle A, Belcaid L, Aldea M, et al. Clinical utility of circulating tumor DNA 2020;12(550):eaaz0131. doi:10.1126/scitranslmed.aaz0131
sequencing with a large panel: a National Center for Precision Medicine 111. Roskams-Hieter B, Kim HJ, Anur P, et al. Plasma cell-free RNA profiling
(PRISM) study. Ann Oncol Off J Eur Soc Med Oncol. 2023;34(4):389-396. distinguishes cancers from pre-malignant conditions in solid and hematologic
doi:10.1016/j.annonc.2023.01.008 malignancies. NPJ Precis Oncol. 2022;6(1):28. doi:10.1038/s41698-022-00270-y
98. Leighl NB, Page RD, Raymond VM, et al. Clinical Utility of Comprehensive 112. Lissa D, Robles AI. Methylation analyses in liquid biopsy. Transl Lung Cancer Res.
Cell-free DNA Analysis to Identify Genomic Biomarkers in Patients with 2016;5(5):492-504. doi:10.21037/tlcr.2016.10.03
Newly Diagnosed Metastatic Non-small Cell Lung Cancer. Clin Cancer Res 113. Laird PW. The power and the promise of DNA methylation markers. Nat Rev Cancer.
Off J Am Assoc Cancer Res. 2019;25(15):4691-4700. doi:10.1158/1078-0432. 2003;3(4):253-266. doi:10.1038/nrc1045
CCR-19-0624 114. Huang J, Wang L. Cell-Free DNA Methylation Profiling Analysis-Technologies and
99. Parikh AR, Leshchiner I, Elagina L, et al. Liquid versus tissue biopsy for Bioinformatics. Cancers. 2019;11(11):1741. doi:10.3390/cancers11111741
detecting acquired resistance and tumor heterogeneity in gastrointestinal 115. Luo H, Wei W, Ye Z, Zheng J, Xu RH. Liquid Biopsy of Methylation Biomarkers
cancers. Nat Med. 2019;25(9):1415-1421. doi:10.1038/s41591-019-0561-9 in Cell-Free DNA. Trends Mol Med. 2021;27(5):482-500. doi:10.1016/j.
100. Sanz-Garcia E, Zhao E, Bratman SV, Siu LL. Monitoring and adapting cancer molmed.2020.12.011
treatment using circulating tumor DNA kinetics: Current research, opportunities, 116. Warton K, Lin V, Navin T, et al. Methylation-capture and Next-Generation
and challenges. Sci Adv. 2022;8(4):eabi8618. doi:10.1126/sciadv.abi8618 Sequencing of free circulating DNA from human plasma. BMC Genomics.
101. Tie J, Cohen JD, Lahouel K, et al. Circulating Tumor DNA Analysis Guiding 2014;15(1):476. doi:10.1186/1471-2164-15-476
Adjuvant Therapy in Stage II Colon Cancer. N Engl J Med. 2022;386(24):2261- 117. Duncavage EJ, Bagg A, Hasserjian RP, et al. Genomic profiling for clinical
2272. doi:10.1056/NEJMoa2200075 decision making in myeloid neoplasms and acute leukemia. Blood.
102. Kasi PM, Fehringer G, Taniguchi H, et al. Impact of Circulating Tumor DNA- 2022;140(21):2228-2247. doi:10.1182/blood.2022015853
Based Detection of Molecular Residual Disease on the Conduct and Design of 118. Duncavage EJ, Schroeder MC, O’Laughlin M, et al. Genome Sequencing as
Clinical Trials for Solid Tumors. JCO Precis Oncol. 2022;6:e2100181. doi:10.1200/ an Alternative to Cytogenetic Analysis in Myeloid Cancers. N Engl J Med.
PO.21.00181 2021;384(10):924-935. doi:10.1056/NEJMoa2024534
103. Kotani D, Oki E, Nakamura Y, et al. Molecular residual disease and efficacy 119. Martínez-Jiménez F, Movasati A, Brunner SR, et al. Pan-cancer whole-
of adjuvant chemotherapy in patients with colorectal cancer. Nat Med. genome comparison of primary and metastatic solid tumours. Nature.
2023;29(1):127-134. doi:10.1038/s41591-022-02115-4 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

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.

TABLE OF
CONTENTS
30 | M-GL-01796 v1.0

You might also like