Advancing Cancer Research Through Biomarkers
eBook
Published: October 23, 2024
Credit: Thermo Fisher Scientific
Biomarkers are revolutionizing cancer research, enabling earlier detection, better prognosis and more targeted treatments. Yet, with the variety of genetic and protein biomarkers available, researchers still face challenges in sample preparation and analysis.
Greater amounts of high-quality DNA and RNA are critical for delivering robust and sensitive results from advanced techniques such as real-time quantitative PCR and next-generation sequencing.
This eBook explores the synergy of genetic and protein biomarkers to provide a comprehensive view of cancer, offering new possibilities for diagnosis and therapy.
Download this eBook to discover:
- How genetic and protein biomarkers drive innovation in cancer research
- Key methods to improve sample quality and accuracy of analysis
- Emerging trends in liquid biopsy and molecular profiling
Synergetic biomarkers:
Fueling advancements in cancer research
Cancer research
Introduction
New technologies are enabling greater access
to genetic and protein biomarkers that are
helping revolutionize cancer research, leading
to significant advancements in diagnosis,
prognosis, and treatment.
Genetic biomarkers can reveal insights into
cancer predisposition, response to targeted
therapies, and immunotherapy response. Greater
amounts of high-quality DNA and RNA can help
deliver more robust and sensitive results from
advanced genetic analysis methodologies such
as real-time quantitative PCR (RT-qPCR) and
next-generation sequencing (NGS). More efficient
solutions to extracting high quality DNA and RNA
from solid tissues and liquid biopsy can both
provide improvements for biomarker identification.
Protein biomarker research and analysis can help
enable early detection of some cancers, greater
understanding of mechanisms of action, or the
development of protein monitoring strategies for
therapeutic response. For some cancers, these
insights can allow for earlier intervention or more
personalized treatment.
The combined analyses of genetic and protein
biomarkers provides a more comprehensive
picture, which can allow for potential future
advancements in cancer research. This may offer
opportunities to improve diagnostic accuracy,
refine risk stratification, or develop even
more targeted therapies based on a
patient’s unique molecular profile.
Article
Where it began: Solid tumors 2
Article
The emerging potential of liquid biopsy 4
Video
Precision oncology: Potential of liquid biopsy to enhance
tumor profiling capabilities in breast cancer management 7
Article
Advancements in cancer biomarker isolation: CTCs and exosomes 8
Early exploration in cancer research 10
Video
Applications in cancer research: Enumeration and molecular profiling of
circulating tumor cells for diagnosis and therapeutic monitoring
of metastatic cancer 10
Video
Applications in cancer research: RNA isolation from organoids and spheroids 11
Article
Innovations in blood cancer research 12
Video
Blood cancer research: Versatile solutions for human leukocyte antigen
testing with peripheral blood 15
Article
Sample integrity and purity are the keys to confidence in cancer research 16
Video
Cancer research: Advancing cancer research tools for liquid biopsy 19
Contents
Solid tumors Solid tumor classifications
There are three classifications of solid tumors: benign, pre-malignant
with potential to become malignant, and malignant.1
Benign tumors are non-cancerous solid tumors that can occur anywhere
in the body. They do not invade other organs or tissues to develop
secondary malignant growths (metastasis), but can cause damage by
compressing nearby tissues or organs. In rare cases, certain benign
tumors can transition to a malignant state.
Pre-malignant tumors have the potential to transform into
cancerous tumors, and therefore require monitoring. They exhibit
increased production of matrix-remodeling proteases and also
release of pro-angiogenic, proliferation, and survival factors in the
tumor’s microenvironment.
Malignant tumors can invade nearby tissues and also spread to
other parts of the body through the blood and lymph systems. They
are characterized by alterations in cell physiology, including increased
growth signals, intensified anti-growth signals, cell and tissue apoptosis
or necrosis, limitless replicative and proliferative potential, prolonged
angiogenesis, and metastasis.
Solid malignant tumors can be classified based on the type of
cell from which they originate:
• Invasive and metastatic carcinomas are malignancies of epithelial
origin that can develop in the skin or tissues lining internal organs.
Subtypes include adenocarcinoma, basal cell carcinoma, squamous cell
carcinoma, transitional cell carcinoma, and ductal carcinoma.
• Sarcoma and undifferentiated tumors arise from connective or
supporting tissues and can transform into soft or synovial sarcoma. They
can originate in bone, cartilage, fat, muscle, or blood vessels. Subtypes
include angiosarcoma, bone sarcoma, fibroblastic sarcoma, and
rhabdomyosarcoma.
• Lymphoma affects lymphocytes, which are cells of the immune
system. It begins in infection-fighting lymphocytes found in lymph
nodes and spleen. Examples include Hodgkin’s disease and
non-Hodgkin’s lymphomas.
Where it began: Solid tumors
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
2
• Blastoma, brain, and spinal cord cancers
are solid tumors that can arise in the brain,
central nervous system, or eyes. Blastomas
develop primarily in pediatric populations.
• Melanomas are skin malignancies that
can also occur in the eyes and, rarely, in
internal organs.
• Germ cell tumors typically originate in the
ovaries and testes but can also occur in the
brain, abdomen, or chest.
• Carcinosarcoma are rare malignant solid
tumors that can arise in different organs.
They consist of two types of cancerous cells
(biphasic) with the ability to metastasize:
carcinoma (epithelial cancer) and sarcoma
(connective or mesenchymal tissue cancer).
Carcinosarcomas are highly aggressive and
often appear to arise de novo.
Sample types and research challenges
Solid tumor samples for research may be
provided as fresh frozen (FF) tissues or formalinfixed paraffin-embedded (FFPE) tissue. Biofluid
samples such as urine, whole blood, serum, or
plasma may also be used for biomarker analysis.
Analysis of this wide variety of samples requires
References
1. Applied Biosystems, Inc.
(2023) A Step-by-Step
Guide to Molecular Profiling
of Tumors for Cancer
Researchers, accessed 21
May 2023 https://www.
thermofisher.com/blog/
life-in-the-lab/a-step-bystep-guide-to-molecularprofiling-of-tumors-forcancer-researchers/.
2. Applied Biosystems, Inc.
(2023) A convenient, solventfree deparaffinization method
for FFPE sample preparation.
[Application Note] https://
assets.thermofisher.
com/TFS-Assets/BID/
Application-Notes/solventfree-deparaffinizationmethod-ffpe-sample-prepapp-note.pdf.
3. US Occupational Safety
and Health Administration
(2021) Xylene, all isomers
(dimethylbenzene).
Occupational Chemical
Database. Updated April 6,
2021. https://www.osha.gov/
chemicaldata/228.
4. US Environmental Protection
Agency (2003) Xylenes;
CASRN 1330-20-7.
Integrated Risk Information
System (IRIS) Chemical
Assessment Summary.
https://iris.epa.gov/static/
pdfs/0270_summary.pdf.
5. Tse RT, et al. (2021) Urinary
Cell-Free DNA in Bladder
Cancer Detection. Diagnostics
(Basel). 11(2):306.
6. Christodoulou E, et al.
(2023) Combined low-pass
whole genome and targeted
sequencing in liquid biopsies
for pediatric solid tumors. NPJ
Precis Oncol 2023;7(1):21.
Figure 1. Workflow for deparaffinization and nucleic acid isolation using AutoLys M Tube system and MagMAX FFPE DNA/RNA Ultra Kit.
Place FFPE curl,
slide, or core in the
AutoLys M Tube.
Digest with Protease
Digestion Buer from
the MagMAX FFPE
DNA/RNA Ultra Kit.
Centrifuge to
separate wax from
the digested sample.
Cool and lift inner tube
containing the wax
from the outer tube
containing the lysate.
Extract DNA and/or
RNA from
the lysate.
Recover extracted
nucleic acid from the
elution plate for
downstream analysis.
Place FFPE
sample in tube Digest sample Centrifuge Recover lysate
Isolate DNA
and/or RNA
Perform downstream
analysis
Figure 2. Workflow for deparaffinization and nucleic acid isolation using AutoLys M Tubes and Caps.
a wide range of methodologies that may include
immunohistochemistry (IHC), fluorescence
in situ hybridization (FISH), RT-qPCR, or
next-generation sequencing (NGS).
With FFPE tissue researchers can investigate
and analyze excised biopsies directly. Immunohistological or molecular profiling techniques
can be used to gain a better understanding
of the proteins or study the morphology of
the tissue samples. FISH and IHC are more
traditional methods used to identify genetic
mutations or molecular changes. RT-qPCR
and NGS molecular profiling approaches
have become more prevalent in recent years
because they enable multiplexed analysis of
multiple loci, whereas, traditional methods focus
on a single target.1
Molecular applications such as qPCR and NGS
are utilized as driving forces behind molecular
profiling of solid tumors. With these applications
there is a need for high-quality nucleic acid
techniques that streamline isolation workflows
enabling sample-to-answer results quickly
and efficiently. Before extracting DNA or RNA
from FFPE samples, deparaffinization must be
conducted to dewax and de-crosslink the fixed
embedded tissue. Conventional methods of
deparaffinization can require extensive hands-on
time, may involve hazardous materials, and can
result in significant tissue loss. Solutions such
as Applied Biosystems™ AutoLys™ M Tubes and
Caps paired with Applied Biosystems™ MagMAX™
FFPE DNA/RNA Ultra Kit, deliver high-quality
DNA and RNA from FFPE tissues using safe and
convenient magnetic-bead-based isolation (Figure
1). This method saves researchers' hands-on time,
eliminates the need for hazardous chemicals, and
minimizes tissue loss due to pelleting techniques.2-6
Explore this workflow
A convenient, solvent-free deparaffinization
method for FFPE sample preparation
Keep exploring solid tumor samples
Application note: Comparison of DNA and RNA
from fresh-frozen vs. FFPE tissue samples
Application note: Mutation detection sensitivity
in matched FFPE tissue and liquid biopsy
samples
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
3
Liquid biopsy
The emerging potential of
liquid biopsy
A new opportunity for less invasive testing
Tissue biopsies remain the standard method in molecular analysis
research of genetic abnormalities linked to cancer. However, solid
tissue sample collection is invasive and occurs at a single timepoint,
providing only a single snapshot to analyze the tumor and assess tumor
heterogeneity. In contrast, liquid biopsies are taken from bodily fluids
such as urine, whole blood, serum, or plasma. Circulating biomarkers
such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and
exosomes that reflect the presence and progression of disease can be
investigated with these samples. Liquid biopsy is emerging as a promising,
less-invasive companion to solid tumor testing in cases where invasive
testing is not practical or to monitor and study cancer progression over
time. Continued advancements in the way we study these biomarkers and
the techniques we use can allow liquid biopsies to significantly impact the
future of the cancer research field.1-7
Oncology research and less invasive prenatal testing have been
revolutionized by liquid biopsy studies in recent years. As a minimally
invasive complementary or alternative approach to tissue biopsies, liquid
biopsies can be less risky, painful, and costly, and are increasingly being
used to analyze biomarkers in liquid samples. Recent studies have shown
the utility of liquid biopsies for:
• Enhancing understanding of tumorigenesis, metastasis, and
therapy resistance
• Detecting cancer at early stages when treatment may be
most successful
• Monitoring of cancer development, disease progression, and recurrence
• Tracking response or resistance during and after treatment to allow for
real-time adjustments to treatments
• Assessing fetal chromosomal anomalies
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
4
The present and future of biomarkers
Cell-free DNA (cfDNA) is the total extracellular
DNA that is released from a variety of cells in
the body, including normal cells, dying cells,
and tumor cells. It can originate from various
tissues and organs, reflecting the overall genetic
makeup of an individual.
Circulating tumor DNA (ctDNA) is the fraction
of cfDNA that originates from tumor cells. It
carries genetic alterations or mutations present
in the tumor cells, providing valuable information
about the genetic characteristics of the tumor.
ctDNA can be isolated from samples such as
plasma and serum to be used as a less invasive
biomarker for monitoring tumor dynamics,
assessing treatment response, detecting minimal
residual disease, or detecting the emergence of
resistance mutations.
Although liquid biopsy can provide valuable
access to ctDNA, cfDNA, and fetal DNA, these
molecules are often scarce within a large volume
of liquid sample, particularly compared to
normal or maternal free-floating DNA. Extracting
these rare molecules in sufficient quantities is
crucial for downstream analyses. A key step in
studying these targets is efficient isolation of
the fragmented DNA while leaving the larger
genomic DNA molecules behind. This aspect
of cfDNA enrichment ensures that the shorter,
fragmented DNA is concentrated and ready for
sensitive downstream analysis. High resolution
size-based recovery can help to increase the
concentration of ctDNA to improve detection.8
Explore an end-to-end workflow for
cell-free DNA analysis
Application note: A complete next-generation
sequencing workflow for circulating cell-free
DNA isolation and analysis
Analyzing total RNA including cell-free RNA
(cfRNA), microRNAs, and RNA in gene fusions
can also serve as valuable biomarker targets and
help to provide important insights to oncology
research.
Circulating tumor cells (CTCs): Intact tumor
cells can break free of their parent tumors
and circulate in the bloodstream. CTCs can
yield additional valuable information about the
composition and behavior of tumors. However,
CTCs are less abundant than ctDNA. Highly
sensitive methods are required to capture and
detect them. CTCs enable investigation of tumor
genetics as well as physical structure, protein
composition, and potential sites for immune
system action. In some cases, CTCs are agents
of metastasis by anchoring in a new location
where they begin to multiply. The intact cellular
structure of CTCs may help identify the original
source of metastatic cancer.9
Learn about CTC isolation
using Dynabeads™
Application note: Isolation of circulating tumor
cells using Dynabeads magnetic beads
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
5
References
1. Applied Biosystems, Inc. (2019) Mutation detection sensitivity
in matched FFPE tissue and liquid biopsy samples. [Application
Note] https://assets.thermofisher.com/TFS-Assets/BID/
Application-Notes/mutation-detection-sensitivity-ffpetissue-liquid-biopsy-samples-app-note.pdf.
2. Guo Q, et al. (2018) Heterogeneous mutation pattern in tumor
tissue and circulating tumor DNA warrants parallel NGS panel
testing. Mol Cancer 17:131.
3. Jovelet C et al. (2016) Circulating cell-free tumor DNA analysis
of 50 genes by next-generation sequencing in the prospective
MOSCATO trial. Clin Cancer Res 22:2690-2698.
4. Schwaederie M, et al. (2016) Use of liquid biopsies in clinical
oncology: pilot experience in 168 patients. Clin Cancer Res
22:5497-5505.
5. Chae YK, et al. (2016) Concordance between genomic
alterations assessed by next-generation sequencing in tumor
tissue or circulating cell-free DNA. Oncotarget 7:65364-65373.
6. Invitrogen (2023) Rapid bead-based isolation of exosomes
for multiomic research. [Application Note] https://assets.
thermofisher.com/TFS-Assets/BID/Application-Notes/
rapid-bead-based-isolation-exosomes-app-note.pdf.
7. Gleichman N. (2023) Liquid Biopsy: Guide, Applications
and Techniques. Tech Networks Diagnostics. https://
www.technologynetworks.com/diagnostics/articles/
liquid-biopsy-guide-applications-and-techniques-328957.
8. Applied Biosystems, Inc. (2015) A complete next-generation
sequencing workflow for circulating cell-free DNA isolation and
analysis. [Application Note]. https://assets.thermofisher.com/
TFS-Assets/LSG/Application-Notes/cfDNA-appnote.pdf.
9. Thermo Fisher Scientific (2023) Liquid Biopsy: The
How, What and Why. Behind the Bench. https://
www.thermofisher.com/blog/behindthebench/
liquid-biopsy-the-how-what-and-why/
10. Neurauter AA, et al. (2017) Automated pull-down of
extracellular vesicles (EVs) on the KingFisher system using
Dynabeads magnetic beads—standardizing EV capture and
analysis. [Application Note] https://assets.thermofisher.
com/TFS-Assets/LSG/brochures/automated-pull-downextracellular-vesicles-app-note.pdf.
11. Chitti SV, et al. (2022) Vesicles as Drug Targets and Delivery
Vehicles for Cancer Therapy. Pharmaceutics 14(12):2822.
Exosomes: Distinct from the nucleic acid and
cellular tumor debris, exosomes are a type of
small (30–150 nm) extracellular vesicle (EV) that
are secreted by cells both in vitro and in vivo
into many body fluids. They carry a cargo of
nucleic acid and protein that can yield valuable
biomarkers to assess the genetic, physiological,
and pathological status of their parent cells.
Although they are released from cells under both
pathological and normal conditions, cancer cells
secrete more exosomes than non-cancerous
cells. Exosomes carry signals between cells as
part of an intricate intercellular communications
network of physiological and pathological
processes that can cross biological barriers. In
cancer, they carry their cargo between primary
and secondary tumors elsewhere in the body,
potentially influencing growth, invasion, and
drug resistance. Exosomes are often more
abundant than CTCs and can be another less
invasive source of valuable information about
metastatic cancers.10,11
Driving discovery in liquid
biopsy workflows
The key to identifying tumor biomarkers and
uncovering valuable insights begins with high
quality sample preparation.
Explore resources to inspire
your cancer research
Sample preparation solutions for
cancer research
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
6
Cell-free DNA
Precision oncology:
Potential of liquid biopsy to enhance tumor profiling capabilities
in breast cancer management
Learn more about the work of research fellow Dr. Karen Page of The University of Leicester, Leicester Cancer Research Center,
using liquid biopsy with automation enablement and subsequent molecular analysis for research into the monitoring of breast cancer.
Karen Page, PhD, is a Research Fellow at the University of Leicester. She has worked in the field of cfDNA and liquid biopsy for
over 20 years, with a primary research interest in the role of liquid biopsies and their utility in breast cancer, focussing on earlystage disease, minimal residual disease, acquired resistance to endocrine therapy, and relapse. Dr. Page was a member of the
working group that was the first to describe whole genome analysis of cfDNA. This work was recommended by Faculty 1000
(F1000) as “an example of bench to bedside science that might be useful in the risk assessment and the monitoring of cancer.”
As the lead in the laboratory for next-generation sequencing, Dr. Page works closely with both clinicians and commercial
partners to organise, carry out, and deliver projects that involve testing new products, reagents, equipment, and methodologies.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
7
Figure 3. General workflow proposed for depletion to capture and remove leukocytes using
Invitrogen™ Dynabeads™ MyOne™ CD45 Leukocyte Depletion beads.
CTCs and exosomes
Figure 2. General workflow proposed for positive isolation to capture epithelial cells from whole
blood samples.
Harnessing magnetic bead technology for CTC isolation
CTCs contribute to the initiation of metastasis by detaching from the
primary tumor and circulated to distant organs via the bloodstream.
Once at a new location, they invade tissues and divide, forming
secondary colonization sites. Detecting and analyzing CTCs is crucial for
understanding disease progression and tailoring appropriate treatment
strategies. One of the groundbreaking technologies facilitating this process
is magnetic bead technology.
Invitrogen™ Dynabeads™ magnetic bead technology employs
superparamagnetic particles, that allow beads to be magnetic only in a
magnetic field but have no residual magnetism when removed from the
magnetic field. The superparamagnetic particles are coated with specific
antibodies that bind to certain targets on cells, allowing for their isolation
when a magnetic field is applied. Dynabeads magnetic beads provide an
automation-friendly tool for isolation of circulating biomarkers. Generally,
large beads are optimal for working on open platforms, while smaller
beads are optimal for microfluidics. Positive isolation can be utilized to
separate CTCs expressing cancer-specific markers, whereas depletion
can be utilized to deplete leukocytes from blood samples for markerindependent CTC enrichment, leaving the target cells untouched.
Positive isolation techniques use Dynabeads magnetic beads coupled
with antibodies that target surface markers on CTCs (Figure 2). This
method results in a high yield of pure CTCs from blood samples.
Dynabeads products used for positive CTC isolation include Invitrogen™
Dynabeads™ Epithelial Enrich (EpCAM) and Invitrogen™ CELLection™
Dynabeads Epithelial Enrich (EpCAM). These products target the epithelial
cell adhesion molecule EpCAM, a prominent epithelial marker on CTCs.
Alternatively, researchers can apply their own specified target antibody
by either using biotinylated antibodies with Invitrogen™ Dynabeads™
Streptavidin™ beads or by covalently coupling their antibodies using the
Invitrogen™ Dynabeads™ Antibody Coupling Kit.
In contrast, depletion techniques deplete leukocytes from blood samples,
enriching the CTCs in the process (Figure 3). This is achieved using
Dynabeads magnetic beads coupled to anti-CD45 antibodies, which bind
to leukocytes, allowing for their removal and hence enriching CTCs in the
T T T T T T
T T T T T T
TTT TTT
Collect sample
Capture
epithelial cells
Wash and
lyse cells Isolate mRNA Analyze using
RT-qPCR
Collect sample Capture leukocytes Remove cells Analyze cells
Advancements in
cancer biomarker isolation:
CTCs and exosomes
• Collect whole
blood sample
Collect sample
Collect sample Capture leukocytes Remove cells Analyze cells
Isolate mRNA Capture
epithelial cells
Wash and
lyse cells
Analyze using
RT-qPCR
• Capture epithelial
cells using
Dynabeads magentic
beads coupled to
anti-EpCAM Abs
• Add sample to
magnet, remove
blood, and keep cells
• Wash and lyse
epithelial cells
attached to
Dynabeads
magnetic beads
• Mix lysate with
Dynabeads
Oligo(dT)25
magnetic beads to
isolate mRNA
• Wash and elute
using a magnet
T T T T T T
T T T T T T
TTT TTT
Collect sample
Capture
epithelial cells
Wash and
lyse cells Isolate mRNA Analyze using
RT-qPCR
Collect sample Capture leukocytes Remove cells Analyze cells
• Collect whole blood
sample
• Prepare MNC sample
• Capture leukocytes
using Dynabeads
magnetic beads coupled
to anti-CD45 Abs
• Remove bead-bound
cells from sample
• Analyze remaining cells
• Analyze expression
and quantify cells
with RT-qPCR
Collect sample Capture
leukocytes Remove cells Analyze cells
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
8
sample. Both isolation techniques are efficient and yield high purity CTCs,
making them suitable for downstream analyses.
Positive isolation and depletion workflows can be automated using
Thermo Scientific™ KingFisher™ sample purification instruments. Kingfisher
instruments help provide high throughput, improved reproducibility, and
reduced manual handling errors.
In conclusion, magnetic bead technology has significantly simplified
the process of isolating CTCs. By providing a high yield of pure CTCs,
magnetic-bead-based isolation enables researchers to gain valuable
insights into cancer progression and metastasis.
Advancements in cancer biomarker isolation:
Harnessing magnetic bead technology for exosome isolation
EVs are categorized into different subtypes that include apoptotic bodies,
oncosomes, and nanovesicles due to differences in biogenesis, release
pathway, size and function.
Traditionally, the isolation of exosomes uses methods such as different
forms of ultracentrifugation that can be labor-intensive, involving
differential, cushion, density gradient ultracentrifugation, ultrafiltration,
and size exclusion chromatography (SEC). These methods can be
time-consuming and may require a large amount
of starting material but low exosome yield.
However, the rapid kinetics and ion exchange
properties of Dynabeads magnetic beads may
offer a more efficient and less labor-intensive
alternative (Figure 5). A key feature of Dynabeads
magnetic beads for any isolation protocol is the
rapid binding kinetics of the beads.
Positively charged Invitrogen™ Dynabeads™
Intact Virus Enrichment beads bind to negatively
charged exosomes, viruses, or proteins within
10 minutes (Figure 4). Following capture, the
exosomes (or other negatively charged vesicles)
can be released from the beads in 10 minutes
by adding an anion with a stronger relative
affinity than the bound vesicle. This short and
easy enrichment approach can be simplified
even further by using the KingFisher purification
system. The automated isolation method allows
larger numbers of samples to be processed in
only 10–20 minutes with high reproducibility,
reduced hands-on time, and minimal error rates.1,2
Dynabeads magnetic beads enable
researchers to directly isolate specific
exosome subpopulations through immunoaffinity capture. The process uses Dynabeads
magnetic beads along with primary antibodies
to common exosomal surface markers (CD9,
CD63, CD81, and EpCAM). In addition,
Invitrogen™ DynaGreen™ CaptureSelect™
Anti-IgG-Fc (Multi-Species) Magnetic Beads
offer a sustainable, microplastic-free smaller
bead option for capturing specific exosome
subpopulations from liquid biopsy samples
in only 40 minutes. This process can also be
automated for high throughput needs.
Learn more
Exosome isolation and monitoring from
cell culture and urine
References
1. Invitrogen (2023) Rapid bead-based isolation of exosomes
for multiomic research. [Application Note] https://assets.
thermofisher.com/TFS-Assets/BID/Application-Notes/
rapid-bead-based-isolation-exosomes-app-note.pdf.
2. Invitrogen (2023) Dynabeads magnetic beads Gentle, efficient
separation of biological materials for when it matters most.
[Application Note] https://assets.thermofisher.com/
TFS-Assets/BID/brochures/dynabeads-magnetic-beadsbrochure.pdf.
Figure 4. The proximity of the beads to the targets in the solution enables short
incubation times and therefore fast protocols for both manual (A) or automated (B)
exosome isolation workflows.
Figure 5. Isolation principle. (A) The positively
charged Dynabeads Intact Virus Enrichment beads
are near the negatively charged exosomes, enabling
rapid binding kinetics and a fast isolation protocol.
(B) For isolation of negatively charged exosomes or
viruses, positively charged Dynabeads Intact Virus
Enrichment beads protected with Cl – ions are used.
Exosomes added to the Dynabeads Intact Virus
Enrichment beads will replace the Cl–
ions and bind to
the bead surface. An anion with higher relative affinity
can subsequently be added to replace the exosomes
and thus release them into the sample.
Rapid kinetics
Short
distance
Cl– Cl–
Cl–
Cl–
Cl–Cl– Cl–
Cl– Cl– Cl–
Cl–
Cl– Cl– Cl– Cl–
Ion exchange
Reversible process
Cl– Cl–
Figure 1. Isolation principle. (A) The positively charged Dynabeads
Intact Virus Enrichment beads are near the negatively charged
exosomes, enabling rapid binding kinetics and a fast isolation protocol.
(B) For isolation of negatively charged exosomes or viruses, positively
charged Dynabeads Intact Virus Enrichment beads protected with
Cl– ions are used. Exosomes added to the Dynabeads Intact Virus
Enrichment beads will replace the Cl– ions and bind to the bead surface.
An anion with higher relative affinity can subsequently be added to
replace the exosomes and thus release them into the sample.
A
B
Manual bead-based target isolation workflow
Add Dynabeads
magnetic beads
to sample
Mix and incubate
for 10 minutes Wash bead-target
complex
Add release buer
and incubate
for 10 minutes
Automated bead-based target isolation workflow
Prepare plates with
beads and target
Load plates into
KingFisher instrument
Collect sample (target on
beads or released target)
Figure 2. Exosome isolation workflow using a manual (A) or
automated (B) method.
A
B
distance
Cl– Cl–
Cl–
Cl–
Cl–Cl– Cl–
Cl– Cl– Cl–
Cl–
Cl– Cl– Cl– Cl–
Ion exchange
Reversible process
Cl– Cl–
Figure 1. Isolation principle. (A) The positively charged Dynabeads
Intact Virus Enrichment beads are near the negatively charged
exosomes, enabling rapid binding kinetics and a fast isolation protocol.
(B) For isolation of negatively charged exosomes or viruses, positively
charged Dynabeads Intact Virus Enrichment beads protected with
Cl– ions are used. Exosomes added to the Dynabeads Intact Virus
Enrichment beads will replace the Cl– ions and bind to the bead surface.
An anion with higher relative affinity can subsequently be added to
replace the exosomes and thus release them into the sample.
B
Manual bead-based target isolation workflow
Add Dynabeads
magnetic beads
to sample
Mix and incubate
for 10 minutes Wash bead-target
complex
Add release buer
and incubate
for 10 minutes
Automated bead-based target isolation workflow
Prepare plates with
beads and target
Load plates into
KingFisher instrument
Collect sample (target on
beads or released target)
Figure 2. Exosome isolation workflow using a manual (A) or
automated (B) method.
A
B
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
9
Clinical research
Early
exploration
in cancer
research
New approaches to investigating
cancer in liquid biopsy and threedimensional tissue cell models
are providing opportunities
for researchers to gain early
insights into cancer mechanisms.
Advancements in enumeration
of CTCs may reveal new insights
into the impact of CTC burden on
early detection, monitoring, and
metastasis. Organoids can model
cancer tissues to investigate
growth, cellular structure, and
microenvironment. Spheroids
may be developed to mimic cell
behavior for drug screening and
resistance.
Applications in cancer research:
Enumeration and molecular profiling of circulating
tumor cells for diagnosis and therapeutic monitoring
of metastatic cancer
Learn more about Dr. Pravin D. Potdar’s methods for the enumeration and isolation of CTCs as well as the significance of
molecular profiling of CTCs for the study and monitoring of metastatic cancers.
Dr. Pravin Potdar is a founding member and former Vice President of the Molecular Pathology Association of India (MPAI) and
has served as Faculty and Professor of Genetics and Stem Cell Biology at Dr. APJ. Abdul Kalam Education and Research
Centre, Mumbai. Dr. Potdar is a former Head and Chief of the Department of Molecular Medicine and Biology at Jaslok
Hospital and Research Centre, Mumbai, India, where he established a Molecular Diagnostics and Stem Cell Research
laboratory and carried out research programs in the fields of cancer, neurological and genetic disorders, infectious diseases,
diabetes mellitus, and stem cells. With over 30 years of scientific research experience and more than 93 published papers in
cancer and stem cell research, Dr. Potdar’s work includes developing several mesenchymal and hematopoietic stem cell lines
from various normal and tumor tissues, adipose tissue, human placental membrane, dental pulp cells, and blood cells.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
10
Applications in cancer research:
RNA Isolation from organoids and spheroids
Gain insights from scientists who share their expertise on RNA extraction methods and workflows for 3D cell culture, with a special
focus on cancer applications.
Organoids and spheroids have become valuable tools in various research areas,
such as drug discovery, toxicology, disease modeling, and regenerative medicine,
as they provide a more accurate representation of complex biology compared to 2D
models. In this webinar, Thermo Fisher scientists Laura Chapman, Anupriya Gupta,
and Jay Bhandari share their expertise on RNA extraction methods and workflows
for 3D cell culture, with an emphasis on cancer applications.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
11
Hematologic cancer
Innovations in blood
cancer research
Types of blood cancer
Cancer arises from tissues, however, when that tissue is bone marrow,
abnormalities in the production and function of blood cells and their
components can result in hematologic cancers such as leukemia,
lymphoma, and myeloma. Hematologic cancer research is a dynamic
and rapidly evolving field focused on understanding and developing
effective treatments for blood cancers. Hematological cancers, including
leukemia, lymphoma, and multiple myeloma, arise from abnormalities in
the production and function of blood cells and their components.
There are three primary classes of hematologic malignancies:1
• Leukemia is characterized by the abnormal production of white blood
cells in the bone marrow.
• Lymphoma starts in the lymphatic system.
• Multiple myeloma is a cancer of plasma cells.
Each of these blood cancers can further be categorized into various
subtypes based on specific characteristics and cell types involved.
The importance of hematologic cancer research
Hematological cancer research aims to advance understanding of the
genetic, molecular, and cellular mechanisms underlying these diseases.
Through innovative research methods and therapeutic approaches,
researchers aim to improve patient outcomes, enhance early detection
strategies, and ultimately, find cures for these complex and challenging
malignancies. Here, we explore recent advancements that are
transforming hematological cancer research, highlighting the importance
of early detection and the innovative approaches being employed with the
potential for better treatment outcomes in the future. Tailored targeting of
the unique characteristics and mechanisms of each type of blood cancer
may allow for more effective, precise, and personalized treatment while
minimizing harm to healthy cells, ultimately helping lead to better outcomes
and reduced side effects.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
12
Early detection of cancer using
blood cells
Early detection of hematological malignancies
can play a pivotal role in helping improve patient
outcomes.2
Identification at an early stage can
enable timely intervention, which may lead
to more effective treatment strategies and
potentially improved outcomes. Research efforts
are underway to develop innovative techniques
for early detection, including molecular
profiling, liquid biopsies, and advanced
analytical technologies.
Molecular profiling and antigen testing
Molecular profiling techniques have dramatically
changed hematological cancer research with
new approaches to unraveling the genetic
complexity of these diseases. Researchers
utilize advanced genomic technologies, such
as NGS, to identify specific genetic alterations
and mutations associated with different
types of hematological cancers.3
These
genetic biomarker signatures can provide
valuable insights into disease classification,
prognostication, and potential therapeutic
targets. By advancing understanding of the
genetic abnormalities driving hematological
cancers, researchers may be able to develop
targeted therapies and personalized treatment
approaches to enable potentially improved
patient outcomes.
A comprehensive profile at the molecular level
can be pivotal in understanding and detecting
genetic alterations that may be relevant to these
cancer types or transplant needs. Molecular
profiling from hematological samples requires
high quality DNA and RNA isolated from the
origin sample. Innovative approaches to the way
researchers can obtain these nucleic acids are
coming to light. Sequential DNA/RNA isolation
with the Applied Biosystems™ MagMAX™
Sequential DNA/RNA Kit provides the ability
to isolate these analytes sequentially with a
single sample utilizing automated protocols on
KingFisher automated purification systems.
Antigen tests are important tools in the
cancer research field as they offer insights
into the growth and monitoring of cancer and,
potentially, its treatment. Human leukocyte
antigen (HLA) typing is a blood antigen test
that identifies antigens on the surface of cells
and tissues. The HLA genes are located in
the major histocompatibility complex, which
is a DNA locus that codes for cell surface
proteins that are essential in the process of
binding antigenic peptides during the immune
response.4
These genes are considered the
most polymorphic genetic system in humans,
with more than 35 thousand alleles described,
and high levels of diversity inside and between
human populations. HLA typing has hence
become an important test for stem cell and
solid organ transplantation, various disease
associations, and pharmacogenetics to screen
for drug hypersensitivity.
NGS technologies have transformed the HLA
typing field by generating high-resolution data
and resolving most allelic ambiguities without
multiple reflexive tests. High-performance NGS
HLA assays demand high-quality genomic DNA
from biological samples such as peripheral blood.
Automated DNA extraction using the Applied
Biosystems™ MagMAX™ DNA Multi-Sample Ultra
2.0 Kit on the KingFisher Apex system can offer
the consistency needed to generate high-quality
genomic DNA, suitable for downstream HLA
typing through downstream sequencing analysis.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
13
The impact of biomarkers
in liquid biopsies
The discovery and validation of protein or
genetic biomarkers within blood samples have
had a significant impact on the advancement of
hematological cancer studies. For hematological
cancers in particular, liquid biopsies to detect
biomarkers in blood and other bodily fluids
have provided a powerful, less invasive, and
real-time approach for monitoring disease
progression, treatment response, and
minimal residual disease.5
Researchers are
continuously exploring the potential of liquid
biopsies and biomarker studies as research
tools for early detection, disease monitoring,
and the identification of therapeutic targets for
hematologic diseases.
Hematological based disease modeling
and preclinical research
Disease modeling and preclinical research
are vital components of hematological cancer
investigation. Researchers mimic the biological
characteristics of hematological cancers using
cell lines, animal models, and three-dimensional
culture systems.6
These models enable the
investigation of disease mechanisms, drug
efficacy, and resistance mechanisms, helping
to provide a platform for the development
and testing of novel therapeutic strategies. By
studying the interactions between cancer cells
and the microenvironment, researchers can
uncover new insights into disease progression
and identify potential targets for intervention.
Paving the way to a cure
Research on hematological cancers is uniting
with genetic research in new ways, yielding
advancements in understanding the genetic
and molecular mechanisms underlying
these diseases. Innovations across workflow
applications may help contribute to expanding
knowledge of the intricacies of these diseases
that may offer researchers approaches to
develop targeted therapies, personalized
treatment approaches, and innovative diagnostic
methods. Through ongoing research efforts,
hematological cancer research strives to
improve patient outcomes, enhance early
detection strategies, and ultimately, find cures
for these complex malignancies.
Related resources
Application note: Genomic DNA extraction
from bone marrow aspirates and peripheral
blood mononuclear cells
Blog: Sample types in genomics and
oncology research: capability advancements
in automation
Application note: Versatile solutions for
human leukocyte antigen testing with
peripheral blood
References
1. Rodriguez-Abreu D, et al. (2007) Epidemiology of hematological
malignancies. Ann Oncol 18 Suppl 1:i3-i8.
2. Liu MC (2021) Transforming the landscape of early
cancer detection using blood tests—Commentary on
current methodologies and future prospects. Br J Cancer
124:1475–1477.
3. Fu Y, et al. (2021) Liquid biopsy technologies for hematological
diseases. Med Res Rev 41(1):246-274.
4. https://www.thermofisher.com/document-connect/
document-connect.html?url=https://assets.thermofisher.
com/TFS-Assets%2FBID%2FApplication-Notes%2Fhumanleukocyte-antigen-testing-peripheral-blood-app-note.pdf
5. Shegekar T, et al. (2023) The Emerging Role of Liquid Biopsies
in Revolutionising Cancer Diagnosis and Therapy. Cureus
15(8):e43650.
6. Georgomanoli M, et al. (2019) Modeling blood diseases
with human induced pluripotent stem cells. Dis Model Mech
12(6):dmm039321.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
14
Hematologic cancer
Blood cancer research:
Versatile solutions for human leukocyte antigen testing
with peripheral blood
Watch our on-demand webinar. Discover how next-generation sequencing (NGS) technologies are transforming HLA typing.
Next-generation sequencing (NGS) technologies transformed HLA typing research by generating high-resolution data and resolving most allelic
ambiguities without multiple reflexive tests. High-performance NGS HLA assays demand high-quality genomic DNA from biological samples such
as peripheral blood. Watch this on-demand webinar in which Thermo Fisher Scientific scientists and application specialists present the performance
of 96 DNA samples extracted from whole blood using the Applied Biosystems™ MagMAX™ magnetic bead-based kit on the Thermo Scientific™
KingFisher™ Flex system for high-resolution HLA typing with the One Lambda™ AllType™ and AllType FASTplex™ NGS kits. This webinar showcases
how high-quality DNA positively impacts the HLA typing process and highlights the crucial importance of quality checks on the NGS workflow.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
15
Sample prep
Sample integrity and purity
are the keys to confidence
in cancer research
Complete sample preparation workflow solutions
As cancer research evolves, tissues, cells, and body fluids are all
becoming valuable sources for new insights into the mechanisms of
cancer and pathways to potential treatments. Cell-free nucleic acids
(cfNAs), CTCs, and exosomes extracted from solid tumor, liquid biopsies,
or hematologic samples can hold the keys to helping us understand
cancer biology. Analyzing these target biomarkers allows researchers to
identify mutations in cancer-associated genes, explore mechanisms of
cancer cell function, investigate response to therapies, and identify cancerassociated biomarkers all with the objective of enabling early detection,
more informative monitoring, prediction of therapeutic response, and more
personalized treatments.
There is a need for streamlined workflows that offer the potential lifespan
monitoring of different cancer types, biomarker discovery, and ultimately
health status. Versatile and automated sample purification instrumentation,
along with optimized kits and reagents can help advance these objectives.
Extracting high-quality nucleic acids from FFPE samples
Although FFPE is excellent for preserving solid tumor tissue morphology,
it can be damaging to some biomolecules, diminish nucleic acid integrity,
and impede genetic analysis. Nucleic acid extraction from FFPE tissues,
which minimizes the impact of degraded samples and preserves the
quality of the tissue, can help ensure the success of downstream
molecular assays including NGS, RT-qPCR, and gene expression profiling.
The AutoLyS M Tube system and MagMAX FFPE DNA/RNA Ultra Kit
can be combined to help provide a streamlined workflow for high-yield,
high-quality nucleic acid isolation from FFPE tissues. The AutoLyS Tube
system yields cleared lysates from FFPE tissues without the need for
deparaffinization or organic solvents. The MagMAX FFPE DNA/RNA
Ultra Kit is designed for simplified, sequential or total isolation of DNA
and RNA separately from a single tissue sample. The resulting DNA
and RNA both are compatible with a broad range of genetic analysis
applications. MagMAX FFPE kits are also scalable for high-throughput or
automated needs.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
16
Isolating fragmented DNA
Efficient recovery and isolation of circulating
nucleic acid species can be fundamental to
preventing invasive sample collection from small
tumors or fragile patients. However, isolation can
be particularly challenging because peripheral
blood also contains circulating DNA derived
from normal cells. Thermo Fisher Scientific
provides a complete, highly efficient workflow to
isolate, purify, and characterize cfDNA even in
the presence of normal cell DNA. First, cfDNA is
isolated from the relevant biological fluid using
the Applied Biosystems™ MagMAX™ CellFree DNA Isolation Kit either manually with a
magnetic stand or automatically on a KingFisher
automated purification system. Isolated
fragments can ultimately be analyzed through use
of Ion Torrent™ Oncomine™ Cell-free assays
and Ion Torrent™ Ion GeneStudio™ S5 systems
to enable multiplexed targeted sequencing for
highly accurate molecular characterization.
Isolating CTCs
Circulating tumor cells (CTCs) are gaining
importance as prognostic markers and for
research into the monitoring of treatment
response. Because of the low number of
CTCs in circulation, highly sensitive methods
are necessary to capture and detect down
to single cells. Dynabeads magnetic beads
provide an automation-friendly tool for isolation
of circulating biomarkers. Positive isolation
can be utilized to separate CTCs expressing
cancer-specific markers, whereas depletion
can be utilized to deplete leukocytes from
blood samples for marker-independent CTC
enrichment, leaving the target cells untouched.
In cancer research, the depletion of CD45-
positive cells is commonly employed to
enrich for CTCs from liquid biopsy samples
such as PBMC and whole blood. This allows
for the acquisition of untouched and viable
CTCs, facilitating subsequent detection and
quantification studies. Dynabeads MyOne
CD45 Leukocyte Depletion Beads are
uniform, superparamagnetic beads with a
primary monoclonal mouse IgG2a antibody
specific for all known isoforms of CD45, a
membrane glycoprotein found on all human
leukocytes. The product can be used to deplete
CD45-positive cells easily and efficiently from
viscous samples such as whole blood, in
approximately 30 minutes.
For positive cell isolation, Dynabeads magnetic
beads coupled to monoclonal antibodies
targeting EpCAM can be used. Dynabeads
Epithelial Enrich Beads allow for high
priority isolation of viable cells. Dynabeads
CELLection Epithelial Enrich Beads provide
a positive cell isolation option that releases
cells after cell capture to yield pure and viable
cells that can be used in any downstream
application. Alternatively, there are options to
take a more customizable approach and couple
other cancer-specific biotinylated antibodies to
Dynabeads Streptavidin magnetic beads for
protein and cell capture.
Enrichment of circulating CTCs is a timeconsuming process if working with many
samples in parallel and can lead to increased
manual handling errors. By utilizing the
KingFisher Sample Purification Systems
with 96 deep-well plates, you can achieve
high-throughput recovery of untouched CTCs
in approximately 50 minutes after loading
the sample and reagents into the plates. The
resulting CTCs are viable and suitable for
culture, downstream molecular analysis, or
proteomic analysis.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
17
Isolation of extracellular vesicles
and exosomes
Ultracentrifugation is a conventional method of
exosome isolation. However, this approach
can be time-consuming and damaging to the
exosomes. A faster approach to exosome
separation from cells and heavy artifacts is to
use polymer precipitation to sequester water
molecules. This process reduces exosomes
solubility, causing their precipitation. The
exosomes are then harvested using low-speed
centrifugation. Invitrogen™ Total Exosome
Isolation reagents and kits leverage this
method to offer a simplistic and fast isolation
solution that is tailored to specific liquid biopsy
sample types.
An alternative and highly specific approach
to exosome isolation is immunomagnetic
separation using Dynabeads magnetic beads.
Immunomagnetic separation is well suited for
purifying exosome subpopulations from liquid
biopsy samples. Exosome-specific antibodies
are bound to the surface of these beads for
a more efficient purification of exosomes.
Dynabeads come pre-coupled with anti-CD9,
CD63, CD81, or EpCAM for subpopulation
isolation. Invitrogen™ Exosome-Streptavidin
Isolation/Detection Reagent is an alternative
option that can be coupled with a biotinylated
primary antibody for more flexibility. DynaGreen
CaptureSelect Anti-IgG-Fc (Multi-Species)
Magnetic Beads provide the flexibility to couple
exosome specific antibodies while enabling
high throughput, rapid, hands-off isolation
through KingFisher automated purification
protocols. For a more generic exosome isolation
approach using magnetic bead separation,
Dynabeads Intact Virus Enrichment beads also
offer a generic, yet rapid, isolation solution.
This is possible due to the size and charge
of exosomes, which is very similar to that
of viruses. This short and simple exosome
enrichment method can be further simplified
using KingFisher instruments as well.
Versatile, automated isolation
Regardless of your target analyte, KingFisher
automated purification systems coupled
with MagMAX isolation kits and Dynabeads
magnetic beads can support. Through versatile
functionality in automating the isolation of DNA,
RNA, proteins, exosomes, and cells, these
tools provide an efficient sample purification
method that helps maximize productivity in labs.
High yield and quality purified analytes can be
obtained allowing for the reproducibility needed
for cancer research workflows and downstream
translational research applications.
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
18
Liquid biopsy
Cancer research:
Advancing cancer research tools for liquid biopsy
Enhance your understanding of liquid biopsy workflows and assays essential in the sample preparation process.
Learn more about critical steps in the liquid biopsy workflow as Dr. Laure Jobert discusses the essential methods for biomarker
enrichment in the sample preparation process. Thermo Fisher Scientific is dedicated to advancing liquid biopsy-based assays,
including the use of Dynabeads magnetic beads, to enrich and analyze biomarkers such as circulating tumor cells (CTCs),
extracellular vesicles, and circulating tumor DNA (ctDNA).
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved.
19
For Research Use Only. Not for use in diagnostic procedures. © 2024 Thermo Fisher Scientific Inc. All rights reserved. All
trademarks are the property of Thermo Fisher Scientific and its subsidiaries unless otherwise specified. EXT6909
Learn more at thermofisher.com/cancerworkflows
Ready to gear up for
your cancer research?
Take our sample purification solutions quiz to uncover the
recommended sample prep solutions that fit your needs.
Your cancer research begins here. Discover end-to-end cancer
research workflows.
Learn more about liquid biopsy ultra-low mutation detection solutions
from prep to analysis: Accelerating the future of liquid biopsy ultralow mutation detection solutions from sample prep to data analysis.
Brought to you by
Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content.
Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version