SlideShare a Scribd company logo
1 of 40
Download to read offline
Contents
4 	 Cancer Research: Current Limitations and Possible Solutions
	 New tools and tech help address the limitations of current methodologies and treatments.
6 	 Custom Morphology Marker Development for Enhanced ROI
	 Selection with NanoString® GeoMx® Digital Spatial Profiler
	 Novel tech enables users to select biologically relevant ROIs based on tissue-specific
	 morphology markers.
11	 Rapid Evaluation of PBMC Counting and Viability
	 Study demonstrates use of an automated cell counter to count PBMCs isolated from whole
	 blood samples.
16 	 How Can Stem Cells Help with Cancer Treatment?
	 Regenerating cells after therapy, targeting cancer pathways, generating immune cells, and
	 being therapeutic carriers are among uses.
22	 Antibody Drug Development: Navigating Immune Response and
	 Multivariate Logistics on the Targeted Treatment Frontier
	 Studying these revolutionary drugs requires a comprehensive and precise approach to
	 understand the multivariate dynamics.
27	 GPCRs: From Signal Pathways to Targeted Drug Development
	 How to break through bottlenecks to obtain full-length GPCRs as antigens for drug development.
34	 Blockade of PD-L1, an Immune Checkpoint Molecule, in a Murine
	 Model of Melanoma
	 Study describes the cellular response to PD-L1 blockade using monoclonal antibodies in
	 melanoma models.
2
Produced with Support from
Tel: 800-637-1277
info@biocompare.com
www.biocompare.com
Biocompare
395 Oyster Point Blvd., Suite 300
South San Francisco, CA 94080
3
4
Cancer Research: Current
Limitations and Possible Solutions
New tools and tech help address the limitations of current
methodologies and treatments.
A major drawback of cancer treatments like radio-
therapy,chemotherapy,andimmunotherapyisthat
they can kill healthy cells along with the cancerous
cells. Across all treatment modalities, it has consis-
tently proven challenging to target cancerous cells
while not negatively affecting healthy ones. Much
work is being done to resolve these issues as well
as others that impact patient outcomes including
tumor recurrence and therapy resistance. Even be-
fore there is treatment, though, there need to be
robust ways to detect cancer, and current detec-
tion methods have their challenges.
Innovative cancer research is being conducted at
academic and medical institutions, as well as at
biopharmaceutical companies and in the life sci-
ence tool provider community. Within these pro-
grams, researchers and clinicians are developing
novel ways to detect, prevent, and treat cancer.The
chapters in this eBook will showcase some of the
new ideas, techniques, and tools that are helping
to advance cancer research by overcoming some of
the limitations of current methodologies.
Analytical techniques that allow for simultaneous
high-plex and spatial analysis of biological sam-
ples are in high demand as such techniques would
help in better identifying cancer. While immuno-
histochemistry provides spatial analysis, only one
biomarker at a time can be stained. Techniques
such as RNA-Seq provide high-plex gene expres-
sion analysis but do not provide spatial analysis.
A new solution is the NanoString® GeoMx® Digital
Spatial Profiler (DSP), which provides high-plex
and spatial analysis, tagging protein or RNA targets
with fluorescently labeled antibodies. The anti-
bodies act as morphology markers, with the mark-
ers being used to determine regions of interest to
analyze. While this development isexciting, a cur-
rent challenge is the lack of available morphology
markers. To this end, Canopy Biosciences® is devel-
oping a set of qualified and validated morphology
markers to address several cancer types, starting
with markers for T cells and macrophages.
The use of stem cells to treat cancer is also an area
of great interest. Sino Biological offers several
products to pair with various stem cell types, such
as hematopoietic stem cells, cancer stem cells, and
embryonic stem cells, to either treat or identify can-
cers. Radiotherapy or chemotherapy oftentimes
affects both cancerous and healthy cells, with the
latter undergoing slow cell growth or even cell
death. Hematopoietic stem cells may be combined
with colony-stimulating factors for infusion into
leukemia patients so that the hematopoietic stem
cells differentiate into new blood cells, replacing
5
healthy cells that were killed due to radiotherapy
or chemotherapy.
Surface markers such as CD105, HER2/ERBB2,
CD166, CD44, Lgr5, and EpCAM may be used to
identify cancer stem cells in tumors, with such
identification being helpful in developing strate-
gies for treating tumors. Incubation of embryonic
stem cells with NK cell or T cell-initiating cytokines,
such as stem cell factor, IL-3, IL-7, IL-15, and FMS-
like tyrosine kinase receptor-3 ligand, leads to dif-
ferentiation of immune cells.
Stem cells are not the only cell types of inter-
est in cancer research. Leukocytes are also of in-
terest since cancer often affects their numbers.
A challenge with obtaining accurate leukocyte
counts is the existence of anucleated red blood
cells and platelets in whole blood samples. Logos
Biosystems’LUNA-FX7™ Automated Cell Counter is
being used to address this challenge. The counter
may be used to identify fluorescently stained leu-
kocytes in whole blood samples and, if peripheral
blood mononuclear cells are isolated from whole
blood, the counter may identify such cells while
determining their concentration and viability. Cell
counting is accurate to an R-square value of at least
0.98, and cell counting is possible even with whole
blood dilutions as low as 1:640.
Raybiotech’s focus is on developing immunoassays
for drug antibodies and protein-based drugs that
target malignant cells. Drug antibodies, accounting
for one-fifth of all new drug approvals, may target
specific malignant tumors, be massed produced,
and operate via several pathways, including block-
ing pathways, flagging cells for cell death, and
delivering drugs via endocytosis. Protein-based
drugs typically operate by interacting directly with
the cancerous cell, recruiting other molecules to
the cancerous cell, or improving the function of
adjacent cells. Raybiotech has immunoassays for,
among other purposes, drug antibody detection,
drug response detection, and drug antibody bind-
ing site detection.
ACROBiosystems is developing G protein coupled
receptors (GPCRs) for antibody-based drug thera-
pies. GPCRs, which aid in regulating physiological
processes, are transmembrane proteins that have
been linked to a number of diseases, including
cancer. GPCRs are a highly studied drug target,
accounting for 27% of the global market share of
therapeutic drugs. In addition, antibodies that tar-
get GPCRs have decided advantages over small
molecule drugs, some of which have slower clear-
ance rate in the body, longer action time, and less
frequent administration. The company currently
has several full-length GPCRs, either launched or in
development, for use with conditions such as lym-
phoma (CCR4), pancreatic cancer (CXCR4), colorec-
tal cancer (EGFR, LGR5) and myeloma (GPRC5D).
BioLegend has done research to show the effect of
blocking PD-L1 ligand on melanoma. PD-L1 is the
ligand of the PD-1 receptor, an immune checkpoint
molecule often found on tumor cells. As immune
checkpoint molecules limit immune response,
their expression on tumor cells causes suppression
of immune response. PD-L1 binding ultimately
causes T cell death. Blocking the PD-L1 ligand from
binding to the PD-1 receptor with an anti-PD-L1
antibody (GoInVivo™ anti-PD-L1 antibody clone)
resulted in activation of T cells, as shown by ele-
vated expression of common T cell markers like
CD25 and CD69. Using BioLegend’s LEGENDplex™
multiplex platform, blocking PD-L1 binding also re-
sulted in increased production of Th cytokines and
chemokines with a simultaneous limiting of pro-in-
flammatory cytokine production. There was also
a reduction in tumor growth with PD-L1 binding
blockage, as well as increased levels of CD8+
T cells.
6
Custom Morphology Marker
Development for Enhanced ROI
Selection with NanoString®
GeoMx® Digital Spatial Profiler
Novel tech enables users to select biologically relevant ROIs based
on tissue-specific morphology markers.
Introduction
Molecular and cellular profiling of the tissue mi-
croenvironment is critical to understanding the
heterogeneity in biological samples and requires
technology capable of providing information
about the spatial context of cells. Yet, researchers
experience barriers when selecting the right tech-
nique—one that allows for both high-plex and
spatial analysis simultaneously.
Immunohistochemistry (IHC) is an important
method in basic clinical work for the exploration of
biomarkers, as it allows for confirmation of target
protein expression in the context of the microen-
vironment. Although IHC offers spatial context, it is
limited to staining or a single biomarker at a time.
Bulk molecular profiling techniques such as RNA-
Seq sacrifice spatial context but enable high-plex
gene expression analysis.
NanoString® GeoMx® Digital Spatial Profiler (DSP)
offers a solution to these issues and is capable of
high-plex spatial resolution of protein or RNA tar-
gets using fluorescently labeled antibodies as mor-
phology markers to guide the selection of regions
Figure 1. NanoString employs fluorescently labeled an-
tibodies as morphology markers to guide the selection
of ROIs. Tonsil tissue stained with DNA dye (blue), Pan-
Ck(green),andCD45(red)markers.
8
of interest (ROI). GeoMx DSP builds on the tenets
of traditional histology and enables users to select
biologically relevant ROIs based on tissue-specific
morphology markers.
Morphology Markers Enable
Biologically Relevant ROI Selection
Complex transcriptional and proteomic pathways
are responsible for creating highly organized tissue
structures, while a major hallmark of disease is the
breakdown of this organization. Therefore, identify-
ing specific structures is a critical first step in making
meaningfulanalyses.GeoMxDSPemploysmorphol-
ogy markers to select and separate ROIs into mean-
ingful groups to compare RNA profiles. Morphology
markers are fluorescently labeled antibodies target-
ing proteins that inform the presence of biological
structures, cell types, and compartments. Through
conscious selection of biologically relevant ROIs,
powerful transcriptome analyses can be performed.
However, the increased adoption of GeoMx DSP
has revealed a severe limitation to this approach—
namely, the lack of morphology markers available.
Although the Whole Transcriptome Atlas (WTA)
panel profiles the expression of >18,000 RNA tar-
gets, researchers are limited to off-the-shelf kits
targeting a select few morphology markers that
distinguish epithelial from immune cells. The Solid
Tumor TME morphology kit, for example, contains
a DNA stain and two markers, Pan-Ck and CD45,
that are commonly used to profile tumor and im-
mune compartments. Many researchers have ex-
pressed the desire for more targeted morphology
resolution—specific to additional cell types and
novel biomarkers.
To address this limitation, Canopy Biosciences® is
developing a portfolio of qualified and validated
morphology markers, focusing on critical targets
that will enable researchers to select ROIs with
greater precision. The catalog will be expanded
to cover morphology markers for a variety of can-
cer types and the most relevant markers in immu-
no-oncology. Further development will include
expansion into neuroscience applications. New
markers will be available to augment the current
off-the-shelf kits or create fully custom sets based
on study goals.
Morphology Marker Qualification
Marker development involves sourcing commer-
cially available antibodies—labeled with specific
fluorescent tags—and evaluating their suitabil-
ity for staining tissues with conditions compat-
ible with subsequent transcriptomic analysis.
Qualifying custom markers for GeoMx DSP is crit-
ical since the processing protocol for GeoMx RNA
assays includes a significant Proteinase K treatment
that can reduce the presence of specific epitopes
of certain antibody clones.
For this catalog, markers were tested following
the “qualified” and “verified” approaches in the
“Morphology Marker Guidelines” whitepaper pre-
sented by NanoString. Qualified markers demon-
strate expected staining patterns typical in a single
tissue,whileverifiedmarkershaveundergonemore
extensive testing on multiple tissues and were suc-
cessfully used in ROI segmentation. Our initial test-
ing of immuno-oncology antibodies resulted in a
mix of both qualified and verified markers.
Furthermore, target-specific positive tissue stain-
ing was verified by an experienced pathologist.
Antibody dilution was optimized to assess specific-
ity and reduce background fluorescence. Canopy
Biosciences uses a decade’s worth of expertise in
immunohistochemistry assay development and
9
histopathology knowledge in the development of
this catalog to validate markers that will augment
the off-the-shelf kits currently available.
Selecting Fluorescently
Labeled Antibodies
GeoMx DSP uses standard filter sets for fluores-
cence imaging, commonly referred to as the FITC,
Cy3, Texas Red, and Cy5 channels. Antibodies are
conjugated to a fluorophore compatible with one
of these filter sets to enable visualization of the tis-
sue structures based on target specificity. However,
off-the-shelf marker kits make use of only three
channels for the DNA stain (FITC) and two morphol-
ogy markers (Cy3 and Texas Red), leaving the Cy5
channel open for custom marker development.
Immuno-oncology Markers Are the
First Added
The initial priority of the project was the addition
of a set of qualified immuno-oncology markers,
including markers for T cells (CD3) and macro-
phages (CD68), among others. These were some of
Figure 2. Wavelengths of GeoMx DSP fluorescence imaging channels for morphology marker conjugation. (Source: ThermoFisher
FluorescenceSpectraViewer)
Figure3.SegmentationofCD68andCD3inbreastcan-
cer samples. Analysis of Cancer Transcriptome Atlas
data shows enrichment of macrophages in CD68 seg-
ments(leftpanel),TcellsintheCD3segment(rightpan-
el), and lack of both cell types in the Pan-CK segment
(centerpanel).
10
the first markers requested by researchers for cus-
tom addition to the Solid Tumor TME Morphology
Kitandarevalidatedintonsil,breastcancer,andcol-
orectal cancer tissues by Canopy Biosciences. Both
CD3 and CD68 differentiate alternate immune cell
phenotypes, enabling more accurate ROI selection
than is possible with a single tumor and immune
cell marker. Clients can now determine whether
gene expression of a signaling pathway correlates
with T cell or macrophage-rich tissue regions. The
pan-leukocyte marker (CD45) in the Solid Tumor
TME kit cannot distinguish between immune cell
subtypes, so custom marker development is nec-
essary for further cellular resolution.
Summary
Molecular and cellular profiling of the tissue micro-
environment is critical to understanding the het-
erogeneity in biological samples and requires tech-
nology capable of providing information about the
spatial context of cells. Here, we demonstrate how
the NanoString® GeoMx® Digital Spatial Profiler
(DSP) enables high-plex spatial resolution of pro-
tein or RNA targets using fluorescently labeled
antibodies as morphology markers, to assist with
ROI-guided analyses. Through accurate selection
of biologically relevant ROIs, transcriptional and
proteomic pathways can be deeply analyzed.
Although several off-the-shelf marker kits exist,
they make use of only three channels, leaving one
channel open for custom marker development.
Canopy Biosciences utilizes this open channel and
expertise in assay development and histopatholo-
gy knowledge to develop custom markers to aug-
ment the currently available kits. New morphology
markers are rigorously validated for precise local-
ization in multiple tissue types, starting with those
most relevant to current immuno-oncology re-
search. Canopy Biosciences’Validated Morphology
Marker Catalog will enable a broader audience of
researchers to take advantage of spatially resolved,
high-plex transcriptomic profiling.
Figure 4. Qualified morphology marker testing of custom markers. A) CD3 morphology marker validation on breast cancer tissue,
DNAstaininblueandCD3inred.B)CD68morphologymarkervalidationinDLBCLtissue,DNAstaininblueandCD68inyellow.
Additional Resource
ROI Selection Markers for GeoMx® Assays
11
Rapid Evaluation of PBMC
Counting and Viability
Study demonstrates use of an automated cell counter to count
PBMCs isolated from whole blood samples.
Introduction
Peripheral Blood Mononuclear Cells (PBMCs) are
a vital source material used in myriad research
applications, including single-cell sequencing to
vaccine development and toxicological studies.
Furthermore, PBMC derivatives such as T cells, B
cells, NK cells, and stem cells are based on cell ther-
apies, including CAR-T cell therapies and regener-
ative medicines. Therefore, accurately measuring
numbers and viability of PBMCs after collection,
isolation, or expansion are essential to making
experimental or manufacturing decisions about
downstream processes. Yet, directly obtaining
leukocyte counts in whole blood using traditional
counting techniques is complicated by the pres-
ence of mature, anucleated RBCs and platelets.
Advantageously, the use of nucleic acid stains like
Acridine Orange and Propidium Iodide (AO/PI) al-
low the nucleated leukocytes to be differentiat-
ed and accurately counted within a whole blood
Figure 1. Diluted whole blood and PBMC-enriched
buffy coat stained using AO/PI fluorescent dye. The mi-
croscopic overlay images of the whole blood stained
with AO/PI (A,C), and PBMCs from enriched buffy
coat stained with AO/PI (B,D) were acquired using the
CELENA® X High Content Imaging System with a 20X
fluorite objective (www.logosbio.com). The yellow ar-
rows indicate nucleated cells in AO/PI positive stained
cells, while the red arrows indicate RBCs. The scale bar
is100µm.
12
sample (Figure 1). Here, we exhibit the ability of
the new dual fluorescent LUNA-FX7™ Automated
Cell Counter to meet diverse cell counting needs
by demonstrating its use in counting leukocytes in
whole blood and the PBMCs isolated from whole
blood samples.
Material and Methods
One milliliter of the human peripheral blood sam-
ple was prepared, and the PBMC sample was ob-
tained by standard density gradient centrifuga-
tion technique using Histopaque-1083 (Sigma,
#10831)1. After final washing, the PBMCs enriched
in the buffy coat were resuspended in 100 µl PBS
or RPMI +10% FBS media. Cell counts were per-
formed in the LUNA-FX7™ with either the 2-channel
PhotonSlide™ (Cat# L12005) or LUNA™ 8-Channel
Slides (Cat# F72001) and used a modified default
protocol in the Fluorescence Cell Counting mode
(Table 1). Before loading the cells, cells were stained
at the standard ratio of the AO/PI reagent (Cat#
F23001); 18 µl of cells + 2 µl of AO/PI, and then 10
µl of the mix was loaded into a slide chamber.
Results
Evaluating the PBMC concentration and
viability with the LUNA-FX7™™
The separated PBMCs were analyzed by counting a
series of 2-fold dilutions using both the 8-channel
and 2-channel slides (Figure 2). The counts using
both slide types showed linearity with an R-square
value of 0.99 or above on the logarithmic scale of
concentrations over the serial dilutions. Not sur-
prisingly, with a greater volume of analysis, the 2-
channel slide showed slightly better consistency
than the 8-channel slide.
The easy counting of leukocytes in
whole blood
The leukocytes in whole blood were visualized by
fluorescence and also accurately enumerated with
the LUNA-FX7™. Among countless mature RBCs,
the fluorescence leukocytes were distinctively
counted from diluted whole blood (Figure 3).
Table1.TheoptimizedparametersettingsforPBMCorleukocytescountingoftheLUNA-FX7™onFluorescenceCellCountingmode
Better therapies start with better counts.
The LUNA-FX7
Fully Automated Cell Counter
TM
WBCs in whole blood PBMCs CAR-T cells
Incomparable accuracy and precision. Less than 1% CV.
Assured reliability through unique validation slides and Quality Control mode.
Higher throughput with multi-channel compatible 8-channel slides.
Fast and easy operation due to precise and reliable autofocus.
21 CFR Part 11 compliance through the CountWire™ software system.
Find out more at www.logosbio.com
14
Conclusion
The enumeration of PBMCs was evaluated ade-
quately on the LUNA-FX7™ Automatic Cell Counter
with the 2-channel PhotonSlide™ and LUNA™
8-Channel Slides. With more slide options for
the LUNA-FX7™, LUNA™ 1-Channel Slides (Cat#
L72011) may be applied for a more comprehensive
concentration range by analyzing 47 image fields,
and LUNA™ 3- Channel Slides (Cat# L72021) are
beneficial for preset of triplicate analysis as count-
ing options. Indeed, leukocyte counting is much
more straightforward by loading the diluted blood
after AO/PI fluorescence staining. So, you can use
the LUNA-FX7™ to respect the archived records of
PBMCs along with leukocyte counts without inten-
sive labor.
Figure 2. The linearity of counting in serial dilutions of PBMCs. (A) Tagged (live or dead) fluorescent and brightfield overlay of several
dilutions. (B) Bar graph showing the results of 5 serial dilutions. (C) The logarithmic scale of the counts over the dilutions using both
2-channeland8-channelslidesdowntoconcentrationslessthan4.00E+04cells/ml.Thescalebarrepresents100µm.
15
References
1.	 S Parasuraman, R Raveendran, and R Kesavan Blood
sample collection in small laboratory animals J
Pharmacol Pharmacother. 2010 Jul-Dec; 1(2): 87–93.
doi: 10.4103/0976-500X.72350
Figure 3. The Leukocyte count of whole blood on Fluorescent Cell Counting mode in the LUNA-FX7™. (A) Tagged leukocyte images of
fluorescent and brightfield overlay in serial dilutions of whole blood samples. (B) Bar graph displaying counting of 5 serial dilutions
from 1:40 to 1:640 of whole blood cells. (C) The linearity of counts appears across concentration on both 2-channel and 8-channel
slides.Thescalebarrepresents100µm.
Additional Resource
Automated Cell Evaluation for Single-Cell
RNA-seq Analysis
16
How Can Stem Cells Help with
Cancer Treatment?
Regeneratingcellsaftertherapy,targetingcancerpathways,generating
immunecells,andbeingtherapeuticcarriersareamonguses.
Cancer is considered to be one of the most chal-
lenging diseases. There are several cancer treat-
ments available, such as surgery, radiotherapy,
chemotherapy, and immunotherapy, but they
often result in suboptimal efficacy, therapy resis-
tance, and tumor recurrence.1,2
Stem cells possess
unique characteristics, including self-renewal and
high-capacity of differentiation. Stem cells have
been widely studied and can be roughly catego-
rized into several groups: Embryonic stem cells
(ESCs), induced pluripotent stem cells (iPSCs), he-
matopoietic stem cells (HSCs), mesenchymal stem
cells (MSCs), neural stem cells (NSCs), and cancer
stem cells (CSCs). Recently, stem cells were shown
to have the potential to improve cancer treatment
by regenerating cells after heavy therapy, targeting
cancer pathways, generating immune cells, and
being therapeutic carriers (Figure 1).3
Stem Cell Transplantation
Hematopoietic stem cells (HSCs), located in bone
marrow, can form all mature blood cells in the body.
High-dose radiotherapy or chemotherapy effects
both cancerous and normal cells, causing slow cell
growth or even cell death.4
HSC transplantation is
now a standard treatment used in multiple myelo-
ma, leukemia, and lymphomas targets after rounds
of therapy to help patients recover. For example, in
leukemia patients, HSCs are infused to help prolif-
erate and differentiate into new blood cells by add-
ing colony-stimulating factors, which activate in-
tracellular signaling pathways in HSCs.5
Until now,
the infusion of HSCs is the only procedure of stem
cells that was approved by the FDA. However, the
occurrence of graft-versus-host-disease (GVHD)
when using allogeneic sources of HSCs remains a
challenge, which is often treated with immunosup-
pressive drugs.6-8
Targeting CSC Pathways for
Cancer Therapy
Cancer stem cells (CSCs) were first identified in
leukemia in 1994. These cells are generated by epi-
genetic mutations in normal stem cells or in pre-
cursor/progenitor cells and found within tumor
tissues. The failure of cancer treatment could be
attributed to the characteristics of CSCs. They can
generate tumors via self-renewal and differentia-
tion into multiple cellular subtypes as normal stem
cells. Although the proportion of CSCs in tumor
tissues is very low, they contribute to multiple tu-
mor malignancies, such as recurrence, metastasis,
heterogeneity, multidrug resistance, and radiation
Advancing Cancer
Therapeutics
with One-stop Solutions
Superior Research Reagents and
CRO Services Relying on Advanced
Technology Platforms
The Global Leader in Recombinant Technology
Recombinant Proteins & Antibodies for Drug Targets,
Supporting Research on Different Hallmarks of Cancer
Premium Recombinant Cytokines Covering All
Cytokine Families
Fast & Efficient Bispecific Antibody Production
High-throughput & Scale-up Recombinant Production
Please visit our website to find more tools and solutions!
Sino Biological US Inc. (U.S.A)
Tel: +1-215-583-7898
Email: order_us@sinobiologicalus.com
www.sinobiological.com
Sino Biological Europe GmbH (Europe)
Tel: +49(0)6196 9678656
Email: order_eu@sinobiologicaleu.com
Sino Biological, Inc. (Global)
Tel: +86-400-890-9989
Email: order@sinobiological.com
18
resistance.9
Surface markers, such as CD166, CD44,
Endoglin/CD105, HER2/ERBB2, Lgr5, and EpCAM,
are often used to identify CSCs from highly het-
erogeneous cell types in tumors.10
The activities
of CSCs are regulated by pluripotent transcription
factors, many intracellular signal pathways, and ex-
tracellular factors, and these factors can be used as
drug targets for cancer treatment. Therefore, tar-
geting CSCs could provide a promise to treat var-
ious types of solid tumors.
Stem Cell Source for Production of
Immune Cells
Chimeric antigen receptor (CAR) T cells and natural
killer (NK) cells have been successfully applied for
anti-cancer immunotherapy. These cells are often
generated from the patient, but the quality and
quantity are hard to control in vivo. Outsourcing to
induced pluripotent stem cells (iPSCs) and embry-
onic stem cells (ESCs) could offer unlimited sources
and enable the expansion of this immunotherapy
to a larger number of patients.11
ESCs are isolated
from embryos, while iPSCs are induced from so-
matic cells in culture by overexpressing Yamanaka
factors: Oct3/4, Sox2, Klf4, c-Myc. Incubation of iP-
SCs and ESCs in growth medium containing NK cell
or T cell-initiating cytokines, for example, stem cell
factor, IL-3, IL-7, IL-15, and FMS-like tyrosine kinase
receptor-3 ligand promotes differentiation of im-
mune cells.
High Bio-activity Recombinant IL-15
Cell proliferation has been measured using MO7e
human megakaryocytic leukemic cells (Figure 2).
Figure1.Strategiesfortheapplicationofstemcelltherapyinthetreatmentofcancer.(Source:https://doi.org/10.3390/cells9030563)
19
Stem Cells as Potential
Therapeutic Carriers
Tumors are considered chronic wound tissue and
their microenvironment, constituted by extracellu-
lar matrix (ECM) and secreted paracrine factors, can
attract the migration of mesenchymal stem cells
(MSCs).12
MSCs are multipotent adult stem cells
present in multiple tissues, including the umbili-
cal cord, bone marrow, and fat tissue. Several im-
mune-related cytokines are chemoattractants for
MSCs to prostate cancer, osteosarcoma, multiple
myeloma, and breast cancer cells, for example,
CXCL16, SDF-1, CCL-25, TNF-α, IL-1β, and IL-6.13
Thus, MSCs can be utilized as potential carriers de-
livering therapeutic agents in treating cancers.
Featured Recombinant
TNF-αα Protein
Sino Biological has developed bioactive recombi-
nant TNF-α cytokines from various species, includ-
ing human, mouse, cynomolgus monkey, rat, fer-
ret, and canine (Figure 3).
Featured Recombinant IL-1ββ Protein
Compared to the competing product, Sino
Biological’s IL-1β protein demonstrates a higher
bioactivity as shown below (Figure 4).
MSCs can release nano-sized exosomes, a type of
extracellular vesicles (EV) that contain various bio-
logical materials, to regulate cell-cell interaction.14
Anti-tumor mRNA, siRNAs, or small molecule drugs
were successfully packed into MSCs and target tu-
mor niche to enhance anti-tumor effects or silence
tumor-related genes.15
MSCs can also be utilized
to carry anti-cancer drugs via nanoparticles (NPs).
Figure2.
Figure3.
20
MSCs-carried PLA NPs successfully targeted brain
tumors, and paclitaxel-loaded NPs inhibited lung
tumor growth in mice.16
Cancer cells could also be selectively killed by
oncolytic viruses (OVs), while the naked OVs are
easy to remove by immune cells. Stem cells, like
neural stem cells (NSCs), could be promising car-
riers to protect and deliver OVs to tumor sites.17
NSCs, originally present in the central nervous
system, can self-renew and generate new neu-
rons and glial cells. A previous study found OVs
carried by human NSCs in combination with
ionizing radiation and temozolomide could en-
hance cytotoxicity to glioma tumor cells in vitro
and increase the survival time of glioblastoma
multiforme (GBM)-bearing mice.18
Both MSCs and NSCs are easier to engineer to ex-
press different genes, which are responsible for the
conversion of a prodrug into cytotoxic metabolites
toward tumor cells.19
This gene therapy is called
“suicide gene therapy”, and two phase I clinical
trials have been completed. One used cytosine
deaminase (CD)-expressing NSCs to convert 5-flu-
orocytosine (5-FC) into tumor-toxic 5-fluorouracil
(5-FU) [NCT01172964, completed]. Another con-
verted ganciclovir from monophosphorylate to
triphosphate form, which is more cytotoxic, by
Herpes simplex virus thymidine kinase (HSV-TK)
expressing MSCs [EudraCT 2012-003741-15, com-
pleted]. However, the anti-tumor efficacy relies on
dose control, the number of stem cells localized
into the tumor microenvironment, and retention in
tumor sites.
In this mini-review, we introduced stem cell-related
cancer treatments. Although stem cell therapy has
achieved positive results, there are still some side
effects to consider, for example, transformation of
normal stem cells into cancer stem cells, the chron-
ic GVHD after allogeneic HSC transplantation, in-
creased immune response, etc. In summary, stem
cell technologies have high potential for tumor
treatments, but they still need further efforts to
overcome the challenges before advancing further.
References
1.	 Vanneman, M.; Dranoff, G. Combining
immunotherapy and targeted therapies in cancer
treatment. Nat. Rev. Cancer 2012, 12, 237–251.
Figure4.
21
2.	 Wang X, Zhang H, Chen X. Drug resistance and
combating drug resistance in cancer. Cancer Drug
Resist. 2019;2:141-160.
3.	 Chu DT, Nguyen TT, Tien NLB, et al. Recent Progress
of Stem Cell Therapy in Cancer Treatment: Molecular
Mechanisms and Potential Applications. Cells.
2020;9(3):563. Published 2020 Feb 28.
4.	 Baskar R, Dai J, Wenlong N, Yeo R, Yeoh KW. Biological
response of cancer cells to radiation treatment. Front
Mol Biosci. 2014;1:24. Published 2014 Nov 17.
5.	 Hopman RK, DiPersio JF. Advances in stem cell
mobilization. Blood Rev. 2014;28(1):31-40.
6.	 Copelan, E.A. Hematopoietic Stem-Cell Transplantation.
N. Engl. J. Med. 2006, 354, 1813–1826.
7.	 Méndez-Ferrer, S.; Michurina, T.V.; Ferraro, F.; Mazloom,
A.R.; MacArthur, B.; Lira, S.A.; Scadden, D.T.; Ma’Ayan,
A.; Enikolopov, G.N.; Frenette, P.S. Mesenchymal and
hematopoietic stem cells form a unique bone marrow
niche. Nature 2010, 466, 829–834.
8.	 Lee, R.H.; Oh, J.Y.; Choi, H.; Bazhanov, N. Therapeutic
factors secreted by mesenchymal stromal cells and
tissue repair. J. Cell. Biochem. 2011, 112, 3073–3078.
9.	 Yu Z, Pestell TG, Lisanti MP, Pestell RG. Cancer stem
cells. Int J Biochem Cell Biol. 2012;44(12):2144-2151
10.	 Codd, A.S.; Kanaseki, T.; Torigo, T.; Tabi, Z. Cancer stem
cells as targets for immunotherapy. Immunology
2017, 153, 304–314
11.	 Iriguchi, S.; Kaneko, S. Toward the development of
true“off-the-shelf”synthetic T-cell immunotherapy.
Cancer Sci. 2019, 110, 16–22.
12.	 Aravindhan, S., Ejam, S.S., Lafta, M.H. et al.
Mesenchymal stem cells and cancer therapy: insights
into targeting the tumour vasculature. Cancer Cell Int.
2021, 21, 158.
13.	 Jung, Y.; Kim, J.K.; Shiozawa, Y.; Wang, J.; Mishra,
A.; Joseph, J.; Berry, J.E.; McGee, S.; Lee, E.; Sun, H.;
et al.Recruitment of mesenchymal stem cells into
prostate tumours promotes metastasis. Nat. Commun.
2013, 4,1795
14.	 Fuhrmann, G.; Serio, A.; Mazo, M.M.; Nair, R.; Stevens,
M.M. Active loading into extracellular vesicles
significantly improves the cellular uptake and
photodynamic effect of porphyrins. J. Control. Release
2015, 205, 35–44
15.	 Kooijmans, S.A.; Schiffelers, R.M.; Zarovni, N.; Vago, R.
Modulation of tissue tropism and biological activity
of exosomes and other extracellular vesicles: New
nanotools for cancer treatment. Pharmacol. Res. 2016,
111, 487–500.
16.	 Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli,
P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia,
S.M.; et al. Paclitaxel is incorporated by mesenchymal
stromal cells and released in exosomes that inhibit in
vitro tumor growth: A new approach for drug delivery.
J. Control. Release 2014, 192, 262–270.
17.	 Marelli, G.; Howells, A.; Lemoine, N.R.; Wang, Y.
Oncolytic Viral Therapy and the Immune System: A
Double-Edged Sword against Cancer. Front. Immunol.
2018, 9, 866.
18.	 Duebgen, M.; Martinez-Quintanilla, J.; Tamura, K.;
Hingtgen, S.; Redjal, N.; Shah, K.; Wakimoto, H. Stem
Cells Loaded With Multimechanistic Oncolytic Herpes
Simplex Virus Variants for Brain Tumor Therapy. J. Natl.
Cancer Inst. 2014, 106
19.	 Sage, E.; Thakrar, R.M.; Janes, S.M. Genetically modified
mesenchymal stromal cells in cancer therapy.
Cytotherapy 2016, 18, 1435–1445.
Additional Resource
More Reagents for Cancer Research
22
Antibody Drug Development:
Navigating Immune Response
and Multivariate Logistics on the
Targeted Treatment Frontier
Studying these revolutionary drugs requires a comprehensive and
precise approach to understand the multivariate dynamics.
Tremendous efforts and ongoing studies are de-
voted to minimizing symptoms while precisely
targeting malignant cells that ultimately turn into
cancer. One of the biggest breakthroughs to date
has been the employment of monoclonal antibod-
ies (mAbs) in targeted immunotherapy.These drug
antibodies (DAs) now account for one-fifth of all
new drug approvals. Their precision targeting has
been shown to increase cure effectiveness while
minimizing collateral damage caused by other
treatments such as chemotherapy.6
Drug antibod-
ies targeted to specific tumorigenic mutations or
molecular aberrations can be mass produced in
cell culture and theoretically employed to treat any
cancer. Antibodies can be chimeric, humanized, or
fully human and operate through three different
mechanisms leading to tumor death: block a path-
way, flag a cell for destruction, or deliver a drug via
receptor-mediated endocytosis.
Similarly, protein-based drugs are usually synthet-
ic versions of proteins that are produced naturally.
They modify cellular responses by either acting di-
rectly with the targets of interest, recruiting other
molecules to the site, or improving the function
of nearby cells. For example, interleukin 2 (IL-2) is
used in kidney and skin cancer treatment to inter-
fere with cancer growth, recruit immune cells, and
stimulate the production of immune cells that can
help destroy the cancer cells.
Both DAs and drug proteins can stimulate the pro-
duction of anti-drug protein antibodies that can
decrease therapeutic efficacy.1,2,7
Therefore, the
path to implementation of DA or protein-based
therapies must be navigated expertly in order to
maximize patient survival. To identify the most ef-
fective DA, it is important to understand and char-
acterize factors that affect individual pharmacoki-
netic variability post-injection and incidences of
Mouse PBMC populations shown with CD4/IFNγ dual staining (RayBright Red and RayBright Violet
450); before & after stimulation. Stained with kit cat# 137-00007; antibody cat# 135-08029 and
135-24013.
CELLULAR STAINING
with RayBright
®
®
dyes
FACS Antibodies »
Live/Dead Markers »
Intracellular Staining Kits »
Universal Compensation Beads »
Flow Cytometry Services »
Now from your trusted protein detection experts:
affordably priced flow cytometry reagents.
RayBiotech offers validated FACS reagents including intracellular staining kits, universal compensation
beads, antibodies featuring ultra bright RayBright dyes, and more. Find out more here.
Empowering your proteomics
RayBiotech
R
Learn about
our sample
program
RAYBIOTECH.COM / INFO@RAYBIOTECH.COM / ISO 13485:2016
24
immune-related adverse events (IRAEs) associated
with foreign antibodies.1,2,7
While immunogenicity
in preclinical species cannot predict the incidence
of human ADAs, it does have utility in characteriz-
ing consequences of potential ADAs.3
Crucial data in optimizing mAB preclinical devel-
opment for precision cancer treatment include
monitoring production of neutralizing antibod-
ies, understanding factors that affect metabolism
and clearance rate, measuring the binding affinity
of each antibody to its target, finding the bound
epitope, and discovering biomarkers to indicate
treatment effectiveness or unintended down-
stream effects.5,7
It is also crucial to know how and
if any other health conditions impact any one of
these traits. For instance, pathogens such as viral
particles can bind to drug target receptors to facili-
tateinfectionorotherundesiredeffects.Identifying
receptor antagonists or other molecules that in-
hibit unwanted receptor activation is an ongoing
focus of drug discovery efforts.
When monitoring efficacy of and immunogen-
ic responses to an antibody therapeutic, the
researcher needs access to the most reliable,
Table 1. Multiple tools are in our cancer-fighting arsenal. A combination of singleplex and multiplex can be employed to unlock the
secretsoftargetedtherapy.
25
Table2.DrugsapprovedbytheFDAtargetvariouspathwaysintumorformationandgrowth.Theyareapprovedforimplementation
inspecificcancers.ImmunogenicityanddrugconcentrationlevelsarecommonlymonitoredwithELISAstudy.
efficient immunoassays from manufacturers with
expertise in protein detection. Fit-for-purpose as-
say formats corresponding to each goal are listed
in Table 1. Using a combination of these tools, re-
al-time inter-human response variation to target-
ed therapy molecules can be resolved regardless
of the pathway they act upon.
Conclusion
From the first approved mAB, Pembrolizumab,4,5
to now, targeted medicine using drug antibodies
has consistently offered promising horizons with-
in the comprehensive field of immunotherapy.
26
Some, such as Tocilizumab, are even used in com-
bination to reduce symptoms that come with more
active forms of therapy such CAR-T,8
a bio-hack to
your immune system that bolsters it to better kill
uninvited cells, seen in Table 2. Regardless of the
mechanism of action, continued study on these
revolutionary drugs requires a comprehensive
and precise approach to understand the multivar-
iate dynamics. Paramount are immunoassays to
discover biomarkers, individualized immunologic
responses, and other potentially neutralizing par-
ticles in a real-body environment.
References
1.	 Baxi, Shrujal, et al.“Immune-related adverse events
for anti-PD-1 and anti-PD-L1 drugs: systematic review
and meta-analysis.”Bmj 360 (2018).
2.	 Eigentler, Thomas K., et al.“Diagnosis, monitoring
and management of immune-related adverse drug
reactions of anti-PD-1 antibody therapy.”Cancer
treatment reviews 45 (2016): 7-18.
3.	 FDA. Guidance for Industry - Immunogenicity
Assessment for Therapeutic Protein Products 2014
[Available from: https://www.fda.gov/downloads/
drugs/guidances/ ucm338856.pdf.]
4.	 Kwok, Gerry, et al.“Pembrolizumab (keytruda).”
Human vaccines & immunotherapeutics 12.11 (2016):
2777-2789.
5.	 Longoria, Teresa C., and Krishnansu S. Tewari.
“Evaluation of the pharmacokinetics and metabolism
of pembrolizumab in the treatment of melanoma.”
Expert opinion on drug metabolism & toxicology
12.10 (2016): 1247-1253.
6.	 Lu, RM., Hwang, YC., Liu, IJ. et al. Development of
therapeutic antibodies for the treatment of diseases.
J Biomed Sci 27, 1 (2020). https://doi.org/10.1186/
s12929-019-0592-z
7.	 Ovacik, Meric, and Kedan Lin.“Tutorial on monoclonal
antibody pharmacokinetics and its considerations in
early development.”Clinical and translational science
11.6 (2018): 540-552.
8.	 Si, Stephanie, and David T. Teachey.“Spotlight on
tocilizumab in the treatment of CAR-T-cell-induced
cytokine release syndrome: clinical evidence to
date.”Therapeutics and clinical risk management 16
(2020): 705.
Additional Resource
Paths to Optimize Antibody Therapeutics
27
GPCRs: From Signal Pathways to
Targeted Drug Development
How to break through bottlenecks to obtain full-length GPCRs as anti-
gens for drug development.
Various proteins on the cell membrane are respon-
sible for cell protection, internal/external materi-
al transportation, and signal transmission. These
membrane proteins play diverse roles in cell biolo-
gy and have become a crucial target in drug devel-
opment. Over 50% of the currently approved drugs
target human membrane proteins. G protein-cou-
pled receptors (GPCRs) are the largest family of
membrane proteins.The primary function of GPCRs
is to transduce a wide and diverse array of extra-
cellular stimuli such as biogenic amines, peptides,
hormones, neurotransmitters, ions, odorants, and
photons into intracellular signals. This signaling, in
turn, regulates a myriad of physiological processes,
including cell metabolism, differentiation, growth,
neurotransmission, and sensory perception. GPCRs
are implicated in various diseases, including type
2 diabetes mellitus (T2DM), obesity, depression,
HIV, cancer, Alzheimer’s disease, etc. The crucial
role of GPCRs and the increased efforts in the drug
discovery field have led to GPCRs becoming the
most successful drug target class in the treatment
of various pathologies. This review discusses the
structure of GPCRs, signaling pathways, and the
development pattern of various stages of targeted
drugs. ACROBiosystems is focused on overcoming
the challenges of GPCR preparation and providing
full-length GPCR proteins to assist our customers,
and collaboratively researching and developing
antibody-drug and therapy strategies.
Structure of GPCRs
The common molecular structure of GPCRs con-
sists of seven transmembrane alpha helices, and
these domains divide the receptor into extracel-
lular N-terminus, intracellular C-terminus, three
extracellular loops, and three intracellular loops.
The extracellular ring contains two highly con-
served cysteine residues, which can stabilize the
spatial structure of the receptor by forming disul-
fide bonds. There is a G-protein binding site on the
intracellular loop. In the case of CCR5, the classical
GPCRs topology is shown in Figure 1.1
The GPCRs Signaling Pathways
The effectors of GPCR activation are the heterotri-
meric G-proteins Gα, Gβ, and Gγ. Activated GPCRs
act as GEFs (guanine-nucleotide-exchange fac-
tors) and exchange GDP for GTP in the Gα subunit,
which activates the protein. A GPCR is free of ligand
28
(L) in its basal state. Gα binds to GDP and is associ-
ated with Gβγ. The heterotrimeric protein complex
might associate with the receptor at this point or
remain free in the membrane as pictured, but once
it encounters a ligand-bound GPCR, downstream
signaling is initiated (Figure 2). Upon ligand bind-
ing, the GPCR becomes activated and undergoes a
conformational change. The resulting GTP-bound
Gα separates from βγ and the active heterotrimeric
proteins. Currently, the GDP-αβγ complex, with the
participation of Mg2+
, exchanges GTP in the bound
GDP with the cytosol to form a GTP-αβγ complex.
Then the G protein is activated and separated from
the receptor and simultaneously disassembles into
two parts, GTP-α and βγ, which diffuse freely along
the cell membrane and directly interact with ef-
fector proteins, such as PLC or adenylate cyclase,
which results in effector activation and initiation
of a second-messenger cascade and completes the
extracellular transmission of signals into the cell.
The GTP in Gα is then hydrolyzed to GDP through
the activity of Gα and RGS proteins (not shown),
leading to Gα inactivation and reassociation of the
heterotrimeric protein complex. This process rep-
resents a full GPCR G-protein cycle.
Progress in drug development
targeting GPCRs
Changing the competitive landscape
GPCRs are the most intensively studied drug tar-
gets comprising approximately 27% of the global
market share of therapeutic drugs, with aggregat-
ed sales for 2011–2015 of $890 billion. In 2017,
survey data showed 481 therapeutic drugs target-
ing GPCRs, which compose 34% of FDA-approved
drugs and mediate their effects through at least
107 unique GPCRs. Approximately 320 drugs are
currently in clinical trials, of which 35% of these
drugs 64 potential novel GPCRs targets.3
Figure1.SchematicdiagramoftheclassicGPCRs1
Figure2.GPCRssignalpathway2
30
Figure3.GPCRsclinicaldrugtargetdistribution3
31
In the field of drug development that targets
GPCRs, GPCRs antibodies have unique advantages
over small molecule drugs:
•	 The clearance rate in the body is lower,
action time is longer, and the corresponding
administration frequency is lower
•	 The selectivity of antibodies is significantly
higher than that of small molecules
•	 However, due to the blood-brain barrier,
antibody drugs cannot enter the central
nervous system. Therefore, for GPCRs that are
expressed in both the peripheral and central
nervous systems, only the peripheral part of
the drug needs to be designed; therapeutic
antibodies can be developed to make the
drug mainly distributed in the peripheral
area and reduce the toxic side effects on the
central nervous system.
Overview of Leading Projects
There are currently about 13 GPCRs targeted anti-
body projects under development that are active
worldwide, as shown in the table below. The target
is mainly concentrated in CCR4, CALCRL, CCR5,
GCGR, GPRC5D, GLP1R, C5AR, CB1, S1PR1, CCR8,
CCR7, GPR49, and AGTR1. The representative drug
is Leronlimab, a humanized IgG4 monoclonal an-
tibody targeting CCR5, an HIV viral entry inhibitor.
By masking CCR5, the HIV (R5) subtype is blocked
from entering healthy T cells, thereby protecting
these cells from viral infections.
ACROBiosystems assists
in the development of
targeted GPCRs antibodies
The first step in preparing antibodies is antigen im-
munity, and the first step in immunity is the prepa-
ration of antigens. Since GPCRs are seven-pass
transmembrane proteins, it is extremely difficult to
obtain biologically active soluble GPCRs antigens.
To obtain a full-length GPCRs antigen for drug devel-
opment, you need to break through two bottlenecks:
•	 How to increase the expression? Unlike
secretory proteins, the expression and
display of membrane proteins are limited
Table1.TargetedGPCRsdruginformationunderresearch(pharmacodia)
32
by the membrane area. Many membrane
proteins involve functions related to material
transport and signaling; overexpression on
the cell membrane can cause irreversible
damage to cells. These characteristics of
membrane proteins greatly limit their
expression. Therefore, to obtain enough
full-length membrane proteins for immunity
and drug development, it is necessary to
design and optimize the expression interval,
expression system, culture conditions, etc.
Even so, the cost of obtaining milligram
membrane proteins is still much higher than
that of soluble proteins.
•	 How to maintain the uniformity and
activity of membrane proteins during
expression and purification? The
transmembrane domain of membrane
proteins is highly hydrophobic, so
unprotected exposure to water will lead
to nonspecific protein aggregation and
even denaturation. Therefore, in the
process of enrichment and purification of
membrane proteins, it is necessary always
to maintain the hydrophilic properties
of the surrounding environment of
membrane proteins. The most widely used
method involves removing membrane
proteins from the cell membrane of the
phospholipid bilayer through detergent4
and
forming micelles to maintain their natural
conformation and functional activity as
much as possible. Furthermore, there are
also methods such as Nanodisc,5
virus-like
particles (VLP),6
and polymer lipid particles
(PoLiPa).7
Therefore, to meet different application needs in
the drug development process that targets GPCRs,
ACROBiosystems has specially set up platform solu-
tions with VLP, Detergent, and Nanodisc to provide
full-length GPCRs proteins such as GPRC5D, CXCR4,
CCR5, and CCR8.
Table2.GPCRsfromACROBiosystems
33
In addition to GPCRs, ACROBiosystems can pro-
vide a complete range of full-length multi-pass
transmembrane proteins, including four-pass
transmembrane protein CD20, Claudin18.2, and
five-pass transmembrane protein CD133. More
full-length proteins are under development.
References
1.	 Dong HF, Wigmore K, Carrington MN, Dean M,
Turpin JA, Howard OM. Variants of CCR5, which
are permissive for HIV-1 infection, show distinct
functional responses to CCL3, CCL4 and CCL5.
Genes Immun. 2005 Oct;6(7):609-19. doi: 10.1038/
sj.gene.6364247.
2.	 Hanlon CD, Andrew DJ. Outside-in signaling--a
brief review of GPCR signaling with a focus on
the Drosophila GPCR family. J Cell Sci. 2015 Oct
1;128(19):3533-42. doi: 10.1242/jcs.175158. Epub 2015
Sep 7..
3.	 Hauser AS, Attwood MM, Rask-Andersen M, Schiöth
HB, Gloriam DE. Trends in GPCR drug discovery: new
agents, targets and indications. Nat Rev Drug Discov.
2017 Dec;16(12):829-842. doi: 10.1038/nrd.2017.178.
4.	 Wiseman DN, Otchere A, Patel JH, Uddin R, Pollock
NL, Routledge SJ, Rothnie AJ, Slack C, Poyner DR,
Bill RM, Goddard AD. Expression and purification
of recombinant G protein-coupled receptors: A
review. Protein Expr Purif. 2020 Mar;167:105524. doi:
10.1016/j.pep.2019.105524. Epub 2019 Oct 31.
5.	 McLean MA, Gregory MC, Sligar SG. Nanodiscs: A
Controlled Bilayer Surface for the Study of Membrane
Proteins. Annu Rev Biophys. 2018 May 20;47:107-124.
doi: 10.1146/annurev-biophys-070816-033620.
6.	 Ho TT, Nguyen JT, Liu J, Stanczak P, Thompson AA,
Yan YG, Chen J, Allerston CK, Dillard CL, Xu H, Shoger
NJ, Cameron JS, Massari ME, Aertgeerts K. Method
for rapid optimization of recombinant GPCR protein
expression and stability using virus-like particles.
Protein Expr Purif. 2017 May;133:41-49. doi: 10.1016/j.
pep.2017.03.002.
7.	 Hothersall JD, Jones AY, Dafforn TR, Perrior T, Chapman
KL. Releasing the technical‘shackles’on GPCR drug
discovery: opportunities enabled by detergent-free
polymer lipid particle (PoLiPa) purification. Drug
Discov Today. 2020 Aug 21:S1359-6446(20)30337-8.
doi: 10.1016/j.drudis.2020.08.006.
Additional Resource
Discover more about GPCRs
34
Blockade of PD-L1, an Immune
Checkpoint Molecule, in a
Murine Model of Melanoma
Study describes the cellular response to PD-L1 blockade using
monoclonal antibodies in melanoma models.
Highlights
Injection of anti-PD-L1 antibody in a mouse mela-
noma model:
•	 Increases levels of activated CD4+
splenic
T cells.
•	 Stimulates production of Th cytokines and
suppresses pro-inflammatory cytokines.
•	 Increases antigen-specific T cells and reduces
tumor growth.
Summary
Immune checkpoint molecules help to regulate
and limit the immune response. As tumor cells are
able to exploit these mechanisms, new immuno-
therapy treatments target immune checkpoints.
We used a monoclonal antibody to block PD-L1
binding in a murine model of melanoma and char-
acterized T cell subsets and cytokine profiles. We
found that treatment with an anti-PD-L1 antibody
resulted in activation of T cells, increased produc-
tion of Th cytokines and chemokines, increased
levels of tumor-specific T cells and caused a reduc-
tion in tumor growth.
Introduction
Cancer treatment is classically composed of a com-
bination of surgery, radiation, and chemothera-
py. More recently, immunotherapy-based strate-
gies have emerged as another treatment option.
Immunotherapy relies on using the body’s own
immune system to recognize and mount an attack
against cancer cells. Immunotherapy can be broad-
ly categorized into cell-based therapies in which
cells are engineered ex vivo before being reintro-
duced into the patient and soluble factor-based
BioLegend products are manufactured in an ISO 13485:2016-certified facility to ensure the highest quality standards.
World-Class Quality | Superior Customer Support | Outstanding Value
For 20 years, BioLegend scientists have supported researchers by developing reagents for flow cytometry, multiomics, and
cell bioprocessing that are critical to accelerating cancer research. Along with our expanding line of reagents, we provide
educational resources that help researchers understand and decipher the complexity of cancer.
We have thousands of scientist-crafted reagents
engineered to combat cancer
• MojoSort™ magnetic cell separation to purify and enrich
target cells
• GMP antibodies and recombinant proteins to activate cells
• Cell-Vive™ media for T/NK cell expansion
• Flow cytometry antibodies to immune profile cells
• Immunoassays that quantitate cancer biomarkers
Discover the difference at: biolegend.com/cancer
Explore our latest infographic and other helpful resources.
Cancer cells evolve new escape mechanisms. Our resources
keep you apprised of developing technologies, such as CAR-T
and CAR-NK cells. Explore our newest infographic to learn
about this advanced immunotherapy. On our website, you
can find additional educational content, including webinars,
blogs, protocols, and scientific posters.
Bring new cancer solutions to light.
Scan the QR code
to download the
infographic.
36
therapies in which monoclonal antibodies or other
proteins are introduced into the patient to mount
an anti-tumor response.
One approach to developing immunothera-
py-based treatments is through the inhibition of
immune checkpoint molecules (reviewed in 1).
Immune checkpoints maintain immune homeosta-
sis and limit the immune response, in part, to pre-
vent autoimmunity. Tumor cells are able to exploit
this system to prevent an anti-tumor response by
expressing checkpoint markers typically found on
antigen presenting cells (APCs). Because immune
checkpoint molecules are often composed of re-
ceptors and ligands, they are able to be targeted
using monoclonal antibodies. Antibodies that can
block ligand-receptor binding allow T cells to re-
main in an activated state to mount an immune
response against the tumor cells.
One such immune checkpoint molecule that
has been studied for use in immunotherapies is
the PD-1 receptor and one of its ligands, PD-L1.2
Engagement of PD-L1 and PD-1 initiates down-
stream signaling pathways ultimately leading to T
cell death.3
As melanoma cells show elevated ex-
pression of PD-L1, the PD-L1 blockade may be par-
ticularly useful in the treatment of melanoma.4, 5
In
this report, we use a GoInVivo™ anti-mouse PD-L1
antibody in a murine model of melanoma and char-
acterize the T cells in the tumor microenvironment
and draining lymph nodes, study the antigen-spe-
cific T cell response, and phenotype the cytokine
profile in the serum.
Materials and Methods
In vivo injection of PD-L1 Antibody
BALB/cJ or C57BL/6 mice were injected with 106
B16F10 melanoma cells in the flank. Mice were
subsequently treated with either 100 or 200 µg of
GoInVivo™ anti-PD-L1 antibody clone 10F.9G2 (Cat.
No. 124328) or isotype control (Cat. No. 400666) ev-
ery 3 days for a total of 3 doses. Dosing schedule
was performed as indicated in the figure legend.
Tissue samples and serum were collected 14 or 24
days following initial dose. Spleen samples were
collected and made into a single cell suspension
for flow staining.
Flow cytometry staining
Cells were stained with anti-CD25 (Cat. No. 102033),
CD69 (Cat. No. 104530), CD152 (CTLA-4) (Cat. No.
106305), CD279 (PD-1) (Cat. No. 109111), CD278
(ICOS) (Cat. No. 313531), I-A/I-E (MHC II) (Cat. No.
107605), CD11c (Cat. No. 117317), CD80 (Cat. No.
104729), CD86 (Cat. No. 105011), MHC pentam-
er (ProImmune), and 7-AAD (Cat. No. 420403).
Cells were acquired with an LSR Fortessa™ (BD
Biosciences) and analyzed using FlowJo software.
Microscopy
Draining lymph nodes were isolated. Tissues were
sectioned in a microtome cryostat at 7 microme-
ters after freezing in OCT media. Sections were
stained with anti-CD4 (Cat. No. 100425), CD8 (Cat.
No. 100727), CD11c (Cat. No. 117346), and B220
(Cat. No. 103251), without fixation or antigen re-
trieval. Images were taken on an Olympus IX83 in-
verted microscope.
Cytokine and chemokine measurement
Cytokine and chemokine profiles were character-
ized using BioLegend’s LEGENDplex™ system (Cat.
Nos. 740740, 740134, 740007). A detailed protocol
has been published.6
37
For more information regarding LEGENDplex™,
please visit: biolegend.com/legendplex
All reagents are from BioLegend, unless
otherwise noted.
Results
Effective blocking of PD-L1 using a mono-
clonal antibody in a melanoma model
To examine the efficacy of a PD-L1 blocking anti-
body in the treatment of a mouse cancer model,
melanoma, we injected mice with 106
B16F10 mel-
anoma cells in the flank. Mice were subsequently
treated with a GoInVivo™ anti-PD-L1 blocking anti-
body every three days (or as indicated). Following
treatment, tissues and serum were collected for im-
munophenotyping (Figure 1).
Anti-PD-L1 treatment increases T cell
activation status
Splenic CD4+
cells were harvested 14 days af-
ter treatment with anti-PD-L1 antibody or the
corresponding isotype control and stained for
common T cell activation markers. Mice that had
been treated with anti-PD-L1 showed an activat-
ed phenotype as measured by increased levels of
CD25, CD69, ICOS, and CD279 (PD-1) as compared
to mice treated with an isotype control (Figure 2).
Importantly, CTLA-4, another immune checkpoint
molecule which negatively regulates the immune
Figure1.Experimentallayoutoftreatmentof
anti-PD-L1antibodytreatmentinamelanomamodel.
Figure2.SplenicCD4+
Tcellstreatedwithanti-PD-L1
(200µgbyI.V.injectionondays0,3,6)demonstrated
anactivatedphenotype.Cellswerecollected14days
followingimplantation.
38
response, was expressed at low levels and re-
mained unchanged in mice that had been injected
with anti-PD-L1.
Splenic Dendritic Cells Downregulate
CD80 following PD-L1 treatment
Next, we examined splenic dendritic cells to deter-
mine whether the increase in activated T cells cor-
related with alterations in the population of APCs.
For these experiments, we first gated on classical
dendritic cells that are MHC II high and CD11c high.
Dendritic cells (DCs) from animals that had been
treated with the anti-PD-L1 antibody showed lower
expression of CD80 (Figure 3). We hypothesize that
downregulation of CD80 may occur to allow for
CD86-CD28 interaction and less binding to CTLA-4.
Anti-PD-L1 treatment results in
elevated numbers of CD8+
DCs in
draining lymph node
To further characterize the APCs, we examined the
draining lymph nodes from animals that had been
injected with anti-PD-L1 and isotype controls. In
animals that received the anti-PD-L1 treatment,
there is an increased level of CD8a+
dendritic cells
in draining lymph nodes. CD8a+
dendritic cells are
APCs that have the ability to cross-present antigens
to CD8+
T cells, which is a common phenomenon
during tumor elimination.7
Anti-PD-L1 treatment in melanoma mod-
els alters the cytokine and chemokine
profile in serum
To more completely analyze the immune response
following anti-PD-L1 treatment, we used our
LEGENDplex™ multiplex platform to analyze the
chemokine and cytokine profile in serum (Figure
4). Notably, we found that animals treated with
anti-PD-L1 had increased content of cytokines
typically associated with a Th1 response, includ-
ing IFN-γ and TNF-α. Additionally, there was an in-
crease in cytokines associated with Th9 and Th22
phenotypes, IL-9 and IL-22, respectively. However,
this did not correspond with an overall increase in
soluble mediators, as they simultaneously limited
the production of other pro-inflammatory cyto-
kines including IL-12p70, IL-27, IL-33, and GM-CSF,
indicating that the cytokine response in these an-
imals was specific. Lastly, anti-PD-L1 treatment
resulted in the elevated production of import-
ant chemokines including MIP-3α, Eotaxin, MIG,
and IP-10. These chemokines may play a role in
anti-tumor immunity by mobilizing other cells.
Figure3.SplenicDCsshowdecreasedlevelsofCD80
(top).CellswerefirstgatedonclassicalDCs(MHCII
high,CD11chigh).IncreasedlevelsofCD8+
DCsin
thedraininglymphnodeofanimalstreatedwith
anti-PD-L1(bottom).TissuewasstainedwithCD4
(green),CD8(red),CD11c(cyan),andB220(blue).
39
Increase in antigen-specific CD8+
tumor
infiltrating lymphocytes (TIL) following
anti-PD-L1 treatment
To examine antigen-specific T cells during tumor
development, we implanted a B16 melanoma cell
line that expresses the peptide SIYRYYGL, which is
derived from the Nucleoprotein of Vesicular sto-
matitis virus (498-505) and examined the tumor
24 days after implantation and antibody treat-
ment. Using an MHC pentamer loaded with the
SIY peptide, we were able to track CD8+
TILs. As
expected, animals that received the anti-PD-L1
treatment had expanded pools of antigen-spe-
cific TILs as compared to those that received the
isotype control (Figure 5). Finally, as a confirma-
tion of other published models,4,5
treatment with
Figure5.Animalstreatedwithanti-PD-L1(100µgI.P.
injectionondays7,13,16followingimplantationof
melanomacellline)showincreasedantigen-specific
TILsandareductionintotaltumorarea.
Figure 4. Cytokine profile in the serum from treated mice
shows elevation of Th cytokines and chemokines. Animals
treated with PD-L1 also showed a simultaneous decrease
of other pro-inflammatory cytokines.
40
an anti-PD-L1 antibody significantly reduced the
tumor growth, as measured by tumor area, in this
melanoma model.
Conclusions
Immune checkpoint molecules make an attrac-
tive target for anti-cancer therapy, as inhibition
of these pathways results in an elevated immune
response, and they often rely on a ligand-recep-
tor interaction. Blockade of these pathways can
often be easily performed using a bioactive an-
tibody, like our GoInVivo™ functional antibodies.
As melanoma cells express PD-L1 to help evade
an anti-tumor response, we sought to under-
stand the specific cellular phenotype of the PD-
L1 blockade in a mouse melanoma model. As
expected, the blockade of PD-L1 resulted in ele-
vated activation of T cells as measured by expres-
sion of common T cell activation markers, includ-
ing CD25 and CD69. Interestingly, this activation
phenotype corresponds with a downregulation
of CD80, another T cell co-stimulatory molecule,
on APCs. We hypothesize that this downregula-
tion may occur to alter the interaction of other
immune checkpoint molecules, including reduc-
ing binding to CTLA-4 and to allow for more in-
teraction between CD86 and CD28. Using our
LEGENDplex™ multiplex platform, we surveyed
the cytokine and chemokine response in the se-
rum following anti-PD-L1 treatment. Interestingly,
anti-PD-L1 treatment increased certain cytokines
associated with a Th response; however, there was
not an overall increase in all soluble mediators, as
the treatment also caused simultaneous down-
regulation of other pro-inflammatory cytokines.
Importantly, treatment with anti-PD-L1 resulted
in increased levels of CD8+ tumor-specific T cells
which is also correlated with a reduction in tumor
size. Collectively, these results describe the cellu-
lar response to the blockade of PD-L1 in a mouse
melanoma model and suggest that anti-PD-L1
monoclonal antibodies may provide an effective
treatment for melanoma.
References
1.	 Pardoll DM. 2012. Nat. Rev. Cancer. 12: 252-264.
2.	 Zou W, et al. 2016. Sci. Transl. Med. 8: 328.
3.	 Jin HT, et al. 2011. Curr. Top. Microbiol. Immunol. 350:
17-37.
4.	 Tsai KK, et al. 2014. Hum. Vaccin. Immunother. 10
(11):3111-3116.
5.	 Hodi FS, et al. 2010. N. Engl. J. Med. 363: 711.
6.	 Lehmann, JS, et al. 2017. J. Vis. Exp. 9:129.
7.	 Fu, C. and Jiang A. 2018. Front. Immunol. 9: 3059.

More Related Content

Similar to Advances-in-cancer-research.pdf

Similar to Advances-in-cancer-research.pdf (20)

Biomarkers & Clinical Research
Biomarkers & Clinical ResearchBiomarkers & Clinical Research
Biomarkers & Clinical Research
 
Role of biotechnology in cancer control
Role of biotechnology in cancer controlRole of biotechnology in cancer control
Role of biotechnology in cancer control
 
Personalizing Oncology with Genomics
Personalizing Oncology with GenomicsPersonalizing Oncology with Genomics
Personalizing Oncology with Genomics
 
Molecular target and development models
Molecular target and development modelsMolecular target and development models
Molecular target and development models
 
KIYATEC_Advanced_IO_CoCulture
KIYATEC_Advanced_IO_CoCultureKIYATEC_Advanced_IO_CoCulture
KIYATEC_Advanced_IO_CoCulture
 
PancreaticCancerFinalPaper
PancreaticCancerFinalPaperPancreaticCancerFinalPaper
PancreaticCancerFinalPaper
 
Types of Biomarkers
Types of Biomarkers Types of Biomarkers
Types of Biomarkers
 
Lancet Oncology 2004
Lancet Oncology 2004Lancet Oncology 2004
Lancet Oncology 2004
 
Salisha ppt (1) (1)
Salisha ppt (1) (1)Salisha ppt (1) (1)
Salisha ppt (1) (1)
 
Biomed central
Biomed centralBiomed central
Biomed central
 
KIYATEC-Advanced-CoCulture-LRes
KIYATEC-Advanced-CoCulture-LResKIYATEC-Advanced-CoCulture-LRes
KIYATEC-Advanced-CoCulture-LRes
 
npjsba201634-2
npjsba201634-2npjsba201634-2
npjsba201634-2
 
GLIIFCA 22 final (1)
GLIIFCA 22 final (1)GLIIFCA 22 final (1)
GLIIFCA 22 final (1)
 
fonc-04-00078.pdf
fonc-04-00078.pdffonc-04-00078.pdf
fonc-04-00078.pdf
 
Genomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentGenomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and development
 
New stratigies for_combating_cancer
New stratigies for_combating_cancerNew stratigies for_combating_cancer
New stratigies for_combating_cancer
 
Lecaut et al 2012
Lecaut et al 2012Lecaut et al 2012
Lecaut et al 2012
 
S13148 019-0757-3
S13148 019-0757-3S13148 019-0757-3
S13148 019-0757-3
 
Vital Signs Edition #6
Vital Signs   Edition #6Vital Signs   Edition #6
Vital Signs Edition #6
 
Recent developments in p53 and nano oncology
Recent developments in p53 and nano oncologyRecent developments in p53 and nano oncology
Recent developments in p53 and nano oncology
 

Recently uploaded

Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactdawncurless
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxpboyjonauth
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxNirmalaLoungPoorunde1
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
URLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppURLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppCeline George
 
Organic Name Reactions for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions  for the students and aspirants of Chemistry12th.pptxOrganic Name Reactions  for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions for the students and aspirants of Chemistry12th.pptxVS Mahajan Coaching Centre
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfsanyamsingh5019
 
Concept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.CompdfConcept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.CompdfUmakantAnnand
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application ) Sakshi Ghasle
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTiammrhaywood
 
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...M56BOOKSTORE PRODUCT/SERVICE
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docxPoojaSen20
 
How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17Celine George
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxiammrhaywood
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 
Crayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon ACrayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon AUnboundStockton
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxSayali Powar
 

Recently uploaded (20)

Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impact
 
Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptx
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptx
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
URLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppURLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website App
 
Organic Name Reactions for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions  for the students and aspirants of Chemistry12th.pptxOrganic Name Reactions  for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions for the students and aspirants of Chemistry12th.pptx
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdf
 
Concept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.CompdfConcept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.Compdf
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application )
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
 
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docx
 
How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 
Staff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSDStaff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSD
 
Crayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon ACrayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon A
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
 

Advances-in-cancer-research.pdf

  • 1.
  • 2. Contents 4 Cancer Research: Current Limitations and Possible Solutions New tools and tech help address the limitations of current methodologies and treatments. 6 Custom Morphology Marker Development for Enhanced ROI Selection with NanoString® GeoMx® Digital Spatial Profiler Novel tech enables users to select biologically relevant ROIs based on tissue-specific morphology markers. 11 Rapid Evaluation of PBMC Counting and Viability Study demonstrates use of an automated cell counter to count PBMCs isolated from whole blood samples. 16 How Can Stem Cells Help with Cancer Treatment? Regenerating cells after therapy, targeting cancer pathways, generating immune cells, and being therapeutic carriers are among uses. 22 Antibody Drug Development: Navigating Immune Response and Multivariate Logistics on the Targeted Treatment Frontier Studying these revolutionary drugs requires a comprehensive and precise approach to understand the multivariate dynamics. 27 GPCRs: From Signal Pathways to Targeted Drug Development How to break through bottlenecks to obtain full-length GPCRs as antigens for drug development. 34 Blockade of PD-L1, an Immune Checkpoint Molecule, in a Murine Model of Melanoma Study describes the cellular response to PD-L1 blockade using monoclonal antibodies in melanoma models. 2
  • 3. Produced with Support from Tel: 800-637-1277 info@biocompare.com www.biocompare.com Biocompare 395 Oyster Point Blvd., Suite 300 South San Francisco, CA 94080 3
  • 4. 4 Cancer Research: Current Limitations and Possible Solutions New tools and tech help address the limitations of current methodologies and treatments. A major drawback of cancer treatments like radio- therapy,chemotherapy,andimmunotherapyisthat they can kill healthy cells along with the cancerous cells. Across all treatment modalities, it has consis- tently proven challenging to target cancerous cells while not negatively affecting healthy ones. Much work is being done to resolve these issues as well as others that impact patient outcomes including tumor recurrence and therapy resistance. Even be- fore there is treatment, though, there need to be robust ways to detect cancer, and current detec- tion methods have their challenges. Innovative cancer research is being conducted at academic and medical institutions, as well as at biopharmaceutical companies and in the life sci- ence tool provider community. Within these pro- grams, researchers and clinicians are developing novel ways to detect, prevent, and treat cancer.The chapters in this eBook will showcase some of the new ideas, techniques, and tools that are helping to advance cancer research by overcoming some of the limitations of current methodologies. Analytical techniques that allow for simultaneous high-plex and spatial analysis of biological sam- ples are in high demand as such techniques would help in better identifying cancer. While immuno- histochemistry provides spatial analysis, only one biomarker at a time can be stained. Techniques such as RNA-Seq provide high-plex gene expres- sion analysis but do not provide spatial analysis. A new solution is the NanoString® GeoMx® Digital Spatial Profiler (DSP), which provides high-plex and spatial analysis, tagging protein or RNA targets with fluorescently labeled antibodies. The anti- bodies act as morphology markers, with the mark- ers being used to determine regions of interest to analyze. While this development isexciting, a cur- rent challenge is the lack of available morphology markers. To this end, Canopy Biosciences® is devel- oping a set of qualified and validated morphology markers to address several cancer types, starting with markers for T cells and macrophages. The use of stem cells to treat cancer is also an area of great interest. Sino Biological offers several products to pair with various stem cell types, such as hematopoietic stem cells, cancer stem cells, and embryonic stem cells, to either treat or identify can- cers. Radiotherapy or chemotherapy oftentimes affects both cancerous and healthy cells, with the latter undergoing slow cell growth or even cell death. Hematopoietic stem cells may be combined with colony-stimulating factors for infusion into leukemia patients so that the hematopoietic stem cells differentiate into new blood cells, replacing
  • 5. 5 healthy cells that were killed due to radiotherapy or chemotherapy. Surface markers such as CD105, HER2/ERBB2, CD166, CD44, Lgr5, and EpCAM may be used to identify cancer stem cells in tumors, with such identification being helpful in developing strate- gies for treating tumors. Incubation of embryonic stem cells with NK cell or T cell-initiating cytokines, such as stem cell factor, IL-3, IL-7, IL-15, and FMS- like tyrosine kinase receptor-3 ligand, leads to dif- ferentiation of immune cells. Stem cells are not the only cell types of inter- est in cancer research. Leukocytes are also of in- terest since cancer often affects their numbers. A challenge with obtaining accurate leukocyte counts is the existence of anucleated red blood cells and platelets in whole blood samples. Logos Biosystems’LUNA-FX7™ Automated Cell Counter is being used to address this challenge. The counter may be used to identify fluorescently stained leu- kocytes in whole blood samples and, if peripheral blood mononuclear cells are isolated from whole blood, the counter may identify such cells while determining their concentration and viability. Cell counting is accurate to an R-square value of at least 0.98, and cell counting is possible even with whole blood dilutions as low as 1:640. Raybiotech’s focus is on developing immunoassays for drug antibodies and protein-based drugs that target malignant cells. Drug antibodies, accounting for one-fifth of all new drug approvals, may target specific malignant tumors, be massed produced, and operate via several pathways, including block- ing pathways, flagging cells for cell death, and delivering drugs via endocytosis. Protein-based drugs typically operate by interacting directly with the cancerous cell, recruiting other molecules to the cancerous cell, or improving the function of adjacent cells. Raybiotech has immunoassays for, among other purposes, drug antibody detection, drug response detection, and drug antibody bind- ing site detection. ACROBiosystems is developing G protein coupled receptors (GPCRs) for antibody-based drug thera- pies. GPCRs, which aid in regulating physiological processes, are transmembrane proteins that have been linked to a number of diseases, including cancer. GPCRs are a highly studied drug target, accounting for 27% of the global market share of therapeutic drugs. In addition, antibodies that tar- get GPCRs have decided advantages over small molecule drugs, some of which have slower clear- ance rate in the body, longer action time, and less frequent administration. The company currently has several full-length GPCRs, either launched or in development, for use with conditions such as lym- phoma (CCR4), pancreatic cancer (CXCR4), colorec- tal cancer (EGFR, LGR5) and myeloma (GPRC5D). BioLegend has done research to show the effect of blocking PD-L1 ligand on melanoma. PD-L1 is the ligand of the PD-1 receptor, an immune checkpoint molecule often found on tumor cells. As immune checkpoint molecules limit immune response, their expression on tumor cells causes suppression of immune response. PD-L1 binding ultimately causes T cell death. Blocking the PD-L1 ligand from binding to the PD-1 receptor with an anti-PD-L1 antibody (GoInVivo™ anti-PD-L1 antibody clone) resulted in activation of T cells, as shown by ele- vated expression of common T cell markers like CD25 and CD69. Using BioLegend’s LEGENDplex™ multiplex platform, blocking PD-L1 binding also re- sulted in increased production of Th cytokines and chemokines with a simultaneous limiting of pro-in- flammatory cytokine production. There was also a reduction in tumor growth with PD-L1 binding blockage, as well as increased levels of CD8+ T cells.
  • 6. 6 Custom Morphology Marker Development for Enhanced ROI Selection with NanoString® GeoMx® Digital Spatial Profiler Novel tech enables users to select biologically relevant ROIs based on tissue-specific morphology markers. Introduction Molecular and cellular profiling of the tissue mi- croenvironment is critical to understanding the heterogeneity in biological samples and requires technology capable of providing information about the spatial context of cells. Yet, researchers experience barriers when selecting the right tech- nique—one that allows for both high-plex and spatial analysis simultaneously. Immunohistochemistry (IHC) is an important method in basic clinical work for the exploration of biomarkers, as it allows for confirmation of target protein expression in the context of the microen- vironment. Although IHC offers spatial context, it is limited to staining or a single biomarker at a time. Bulk molecular profiling techniques such as RNA- Seq sacrifice spatial context but enable high-plex gene expression analysis. NanoString® GeoMx® Digital Spatial Profiler (DSP) offers a solution to these issues and is capable of high-plex spatial resolution of protein or RNA tar- gets using fluorescently labeled antibodies as mor- phology markers to guide the selection of regions Figure 1. NanoString employs fluorescently labeled an- tibodies as morphology markers to guide the selection of ROIs. Tonsil tissue stained with DNA dye (blue), Pan- Ck(green),andCD45(red)markers.
  • 7.
  • 8. 8 of interest (ROI). GeoMx DSP builds on the tenets of traditional histology and enables users to select biologically relevant ROIs based on tissue-specific morphology markers. Morphology Markers Enable Biologically Relevant ROI Selection Complex transcriptional and proteomic pathways are responsible for creating highly organized tissue structures, while a major hallmark of disease is the breakdown of this organization. Therefore, identify- ing specific structures is a critical first step in making meaningfulanalyses.GeoMxDSPemploysmorphol- ogy markers to select and separate ROIs into mean- ingful groups to compare RNA profiles. Morphology markers are fluorescently labeled antibodies target- ing proteins that inform the presence of biological structures, cell types, and compartments. Through conscious selection of biologically relevant ROIs, powerful transcriptome analyses can be performed. However, the increased adoption of GeoMx DSP has revealed a severe limitation to this approach— namely, the lack of morphology markers available. Although the Whole Transcriptome Atlas (WTA) panel profiles the expression of >18,000 RNA tar- gets, researchers are limited to off-the-shelf kits targeting a select few morphology markers that distinguish epithelial from immune cells. The Solid Tumor TME morphology kit, for example, contains a DNA stain and two markers, Pan-Ck and CD45, that are commonly used to profile tumor and im- mune compartments. Many researchers have ex- pressed the desire for more targeted morphology resolution—specific to additional cell types and novel biomarkers. To address this limitation, Canopy Biosciences® is developing a portfolio of qualified and validated morphology markers, focusing on critical targets that will enable researchers to select ROIs with greater precision. The catalog will be expanded to cover morphology markers for a variety of can- cer types and the most relevant markers in immu- no-oncology. Further development will include expansion into neuroscience applications. New markers will be available to augment the current off-the-shelf kits or create fully custom sets based on study goals. Morphology Marker Qualification Marker development involves sourcing commer- cially available antibodies—labeled with specific fluorescent tags—and evaluating their suitabil- ity for staining tissues with conditions compat- ible with subsequent transcriptomic analysis. Qualifying custom markers for GeoMx DSP is crit- ical since the processing protocol for GeoMx RNA assays includes a significant Proteinase K treatment that can reduce the presence of specific epitopes of certain antibody clones. For this catalog, markers were tested following the “qualified” and “verified” approaches in the “Morphology Marker Guidelines” whitepaper pre- sented by NanoString. Qualified markers demon- strate expected staining patterns typical in a single tissue,whileverifiedmarkershaveundergonemore extensive testing on multiple tissues and were suc- cessfully used in ROI segmentation. Our initial test- ing of immuno-oncology antibodies resulted in a mix of both qualified and verified markers. Furthermore, target-specific positive tissue stain- ing was verified by an experienced pathologist. Antibody dilution was optimized to assess specific- ity and reduce background fluorescence. Canopy Biosciences uses a decade’s worth of expertise in immunohistochemistry assay development and
  • 9. 9 histopathology knowledge in the development of this catalog to validate markers that will augment the off-the-shelf kits currently available. Selecting Fluorescently Labeled Antibodies GeoMx DSP uses standard filter sets for fluores- cence imaging, commonly referred to as the FITC, Cy3, Texas Red, and Cy5 channels. Antibodies are conjugated to a fluorophore compatible with one of these filter sets to enable visualization of the tis- sue structures based on target specificity. However, off-the-shelf marker kits make use of only three channels for the DNA stain (FITC) and two morphol- ogy markers (Cy3 and Texas Red), leaving the Cy5 channel open for custom marker development. Immuno-oncology Markers Are the First Added The initial priority of the project was the addition of a set of qualified immuno-oncology markers, including markers for T cells (CD3) and macro- phages (CD68), among others. These were some of Figure 2. Wavelengths of GeoMx DSP fluorescence imaging channels for morphology marker conjugation. (Source: ThermoFisher FluorescenceSpectraViewer) Figure3.SegmentationofCD68andCD3inbreastcan- cer samples. Analysis of Cancer Transcriptome Atlas data shows enrichment of macrophages in CD68 seg- ments(leftpanel),TcellsintheCD3segment(rightpan- el), and lack of both cell types in the Pan-CK segment (centerpanel).
  • 10. 10 the first markers requested by researchers for cus- tom addition to the Solid Tumor TME Morphology Kitandarevalidatedintonsil,breastcancer,andcol- orectal cancer tissues by Canopy Biosciences. Both CD3 and CD68 differentiate alternate immune cell phenotypes, enabling more accurate ROI selection than is possible with a single tumor and immune cell marker. Clients can now determine whether gene expression of a signaling pathway correlates with T cell or macrophage-rich tissue regions. The pan-leukocyte marker (CD45) in the Solid Tumor TME kit cannot distinguish between immune cell subtypes, so custom marker development is nec- essary for further cellular resolution. Summary Molecular and cellular profiling of the tissue micro- environment is critical to understanding the het- erogeneity in biological samples and requires tech- nology capable of providing information about the spatial context of cells. Here, we demonstrate how the NanoString® GeoMx® Digital Spatial Profiler (DSP) enables high-plex spatial resolution of pro- tein or RNA targets using fluorescently labeled antibodies as morphology markers, to assist with ROI-guided analyses. Through accurate selection of biologically relevant ROIs, transcriptional and proteomic pathways can be deeply analyzed. Although several off-the-shelf marker kits exist, they make use of only three channels, leaving one channel open for custom marker development. Canopy Biosciences utilizes this open channel and expertise in assay development and histopatholo- gy knowledge to develop custom markers to aug- ment the currently available kits. New morphology markers are rigorously validated for precise local- ization in multiple tissue types, starting with those most relevant to current immuno-oncology re- search. Canopy Biosciences’Validated Morphology Marker Catalog will enable a broader audience of researchers to take advantage of spatially resolved, high-plex transcriptomic profiling. Figure 4. Qualified morphology marker testing of custom markers. A) CD3 morphology marker validation on breast cancer tissue, DNAstaininblueandCD3inred.B)CD68morphologymarkervalidationinDLBCLtissue,DNAstaininblueandCD68inyellow. Additional Resource ROI Selection Markers for GeoMx® Assays
  • 11. 11 Rapid Evaluation of PBMC Counting and Viability Study demonstrates use of an automated cell counter to count PBMCs isolated from whole blood samples. Introduction Peripheral Blood Mononuclear Cells (PBMCs) are a vital source material used in myriad research applications, including single-cell sequencing to vaccine development and toxicological studies. Furthermore, PBMC derivatives such as T cells, B cells, NK cells, and stem cells are based on cell ther- apies, including CAR-T cell therapies and regener- ative medicines. Therefore, accurately measuring numbers and viability of PBMCs after collection, isolation, or expansion are essential to making experimental or manufacturing decisions about downstream processes. Yet, directly obtaining leukocyte counts in whole blood using traditional counting techniques is complicated by the pres- ence of mature, anucleated RBCs and platelets. Advantageously, the use of nucleic acid stains like Acridine Orange and Propidium Iodide (AO/PI) al- low the nucleated leukocytes to be differentiat- ed and accurately counted within a whole blood Figure 1. Diluted whole blood and PBMC-enriched buffy coat stained using AO/PI fluorescent dye. The mi- croscopic overlay images of the whole blood stained with AO/PI (A,C), and PBMCs from enriched buffy coat stained with AO/PI (B,D) were acquired using the CELENA® X High Content Imaging System with a 20X fluorite objective (www.logosbio.com). The yellow ar- rows indicate nucleated cells in AO/PI positive stained cells, while the red arrows indicate RBCs. The scale bar is100µm.
  • 12. 12 sample (Figure 1). Here, we exhibit the ability of the new dual fluorescent LUNA-FX7™ Automated Cell Counter to meet diverse cell counting needs by demonstrating its use in counting leukocytes in whole blood and the PBMCs isolated from whole blood samples. Material and Methods One milliliter of the human peripheral blood sam- ple was prepared, and the PBMC sample was ob- tained by standard density gradient centrifuga- tion technique using Histopaque-1083 (Sigma, #10831)1. After final washing, the PBMCs enriched in the buffy coat were resuspended in 100 µl PBS or RPMI +10% FBS media. Cell counts were per- formed in the LUNA-FX7™ with either the 2-channel PhotonSlide™ (Cat# L12005) or LUNA™ 8-Channel Slides (Cat# F72001) and used a modified default protocol in the Fluorescence Cell Counting mode (Table 1). Before loading the cells, cells were stained at the standard ratio of the AO/PI reagent (Cat# F23001); 18 µl of cells + 2 µl of AO/PI, and then 10 µl of the mix was loaded into a slide chamber. Results Evaluating the PBMC concentration and viability with the LUNA-FX7™™ The separated PBMCs were analyzed by counting a series of 2-fold dilutions using both the 8-channel and 2-channel slides (Figure 2). The counts using both slide types showed linearity with an R-square value of 0.99 or above on the logarithmic scale of concentrations over the serial dilutions. Not sur- prisingly, with a greater volume of analysis, the 2- channel slide showed slightly better consistency than the 8-channel slide. The easy counting of leukocytes in whole blood The leukocytes in whole blood were visualized by fluorescence and also accurately enumerated with the LUNA-FX7™. Among countless mature RBCs, the fluorescence leukocytes were distinctively counted from diluted whole blood (Figure 3). Table1.TheoptimizedparametersettingsforPBMCorleukocytescountingoftheLUNA-FX7™onFluorescenceCellCountingmode
  • 13. Better therapies start with better counts. The LUNA-FX7 Fully Automated Cell Counter TM WBCs in whole blood PBMCs CAR-T cells Incomparable accuracy and precision. Less than 1% CV. Assured reliability through unique validation slides and Quality Control mode. Higher throughput with multi-channel compatible 8-channel slides. Fast and easy operation due to precise and reliable autofocus. 21 CFR Part 11 compliance through the CountWire™ software system. Find out more at www.logosbio.com
  • 14. 14 Conclusion The enumeration of PBMCs was evaluated ade- quately on the LUNA-FX7™ Automatic Cell Counter with the 2-channel PhotonSlide™ and LUNA™ 8-Channel Slides. With more slide options for the LUNA-FX7™, LUNA™ 1-Channel Slides (Cat# L72011) may be applied for a more comprehensive concentration range by analyzing 47 image fields, and LUNA™ 3- Channel Slides (Cat# L72021) are beneficial for preset of triplicate analysis as count- ing options. Indeed, leukocyte counting is much more straightforward by loading the diluted blood after AO/PI fluorescence staining. So, you can use the LUNA-FX7™ to respect the archived records of PBMCs along with leukocyte counts without inten- sive labor. Figure 2. The linearity of counting in serial dilutions of PBMCs. (A) Tagged (live or dead) fluorescent and brightfield overlay of several dilutions. (B) Bar graph showing the results of 5 serial dilutions. (C) The logarithmic scale of the counts over the dilutions using both 2-channeland8-channelslidesdowntoconcentrationslessthan4.00E+04cells/ml.Thescalebarrepresents100µm.
  • 15. 15 References 1. S Parasuraman, R Raveendran, and R Kesavan Blood sample collection in small laboratory animals J Pharmacol Pharmacother. 2010 Jul-Dec; 1(2): 87–93. doi: 10.4103/0976-500X.72350 Figure 3. The Leukocyte count of whole blood on Fluorescent Cell Counting mode in the LUNA-FX7™. (A) Tagged leukocyte images of fluorescent and brightfield overlay in serial dilutions of whole blood samples. (B) Bar graph displaying counting of 5 serial dilutions from 1:40 to 1:640 of whole blood cells. (C) The linearity of counts appears across concentration on both 2-channel and 8-channel slides.Thescalebarrepresents100µm. Additional Resource Automated Cell Evaluation for Single-Cell RNA-seq Analysis
  • 16. 16 How Can Stem Cells Help with Cancer Treatment? Regeneratingcellsaftertherapy,targetingcancerpathways,generating immunecells,andbeingtherapeuticcarriersareamonguses. Cancer is considered to be one of the most chal- lenging diseases. There are several cancer treat- ments available, such as surgery, radiotherapy, chemotherapy, and immunotherapy, but they often result in suboptimal efficacy, therapy resis- tance, and tumor recurrence.1,2 Stem cells possess unique characteristics, including self-renewal and high-capacity of differentiation. Stem cells have been widely studied and can be roughly catego- rized into several groups: Embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), he- matopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and cancer stem cells (CSCs). Recently, stem cells were shown to have the potential to improve cancer treatment by regenerating cells after heavy therapy, targeting cancer pathways, generating immune cells, and being therapeutic carriers (Figure 1).3 Stem Cell Transplantation Hematopoietic stem cells (HSCs), located in bone marrow, can form all mature blood cells in the body. High-dose radiotherapy or chemotherapy effects both cancerous and normal cells, causing slow cell growth or even cell death.4 HSC transplantation is now a standard treatment used in multiple myelo- ma, leukemia, and lymphomas targets after rounds of therapy to help patients recover. For example, in leukemia patients, HSCs are infused to help prolif- erate and differentiate into new blood cells by add- ing colony-stimulating factors, which activate in- tracellular signaling pathways in HSCs.5 Until now, the infusion of HSCs is the only procedure of stem cells that was approved by the FDA. However, the occurrence of graft-versus-host-disease (GVHD) when using allogeneic sources of HSCs remains a challenge, which is often treated with immunosup- pressive drugs.6-8 Targeting CSC Pathways for Cancer Therapy Cancer stem cells (CSCs) were first identified in leukemia in 1994. These cells are generated by epi- genetic mutations in normal stem cells or in pre- cursor/progenitor cells and found within tumor tissues. The failure of cancer treatment could be attributed to the characteristics of CSCs. They can generate tumors via self-renewal and differentia- tion into multiple cellular subtypes as normal stem cells. Although the proportion of CSCs in tumor tissues is very low, they contribute to multiple tu- mor malignancies, such as recurrence, metastasis, heterogeneity, multidrug resistance, and radiation
  • 17. Advancing Cancer Therapeutics with One-stop Solutions Superior Research Reagents and CRO Services Relying on Advanced Technology Platforms The Global Leader in Recombinant Technology Recombinant Proteins & Antibodies for Drug Targets, Supporting Research on Different Hallmarks of Cancer Premium Recombinant Cytokines Covering All Cytokine Families Fast & Efficient Bispecific Antibody Production High-throughput & Scale-up Recombinant Production Please visit our website to find more tools and solutions! Sino Biological US Inc. (U.S.A) Tel: +1-215-583-7898 Email: order_us@sinobiologicalus.com www.sinobiological.com Sino Biological Europe GmbH (Europe) Tel: +49(0)6196 9678656 Email: order_eu@sinobiologicaleu.com Sino Biological, Inc. (Global) Tel: +86-400-890-9989 Email: order@sinobiological.com
  • 18. 18 resistance.9 Surface markers, such as CD166, CD44, Endoglin/CD105, HER2/ERBB2, Lgr5, and EpCAM, are often used to identify CSCs from highly het- erogeneous cell types in tumors.10 The activities of CSCs are regulated by pluripotent transcription factors, many intracellular signal pathways, and ex- tracellular factors, and these factors can be used as drug targets for cancer treatment. Therefore, tar- geting CSCs could provide a promise to treat var- ious types of solid tumors. Stem Cell Source for Production of Immune Cells Chimeric antigen receptor (CAR) T cells and natural killer (NK) cells have been successfully applied for anti-cancer immunotherapy. These cells are often generated from the patient, but the quality and quantity are hard to control in vivo. Outsourcing to induced pluripotent stem cells (iPSCs) and embry- onic stem cells (ESCs) could offer unlimited sources and enable the expansion of this immunotherapy to a larger number of patients.11 ESCs are isolated from embryos, while iPSCs are induced from so- matic cells in culture by overexpressing Yamanaka factors: Oct3/4, Sox2, Klf4, c-Myc. Incubation of iP- SCs and ESCs in growth medium containing NK cell or T cell-initiating cytokines, for example, stem cell factor, IL-3, IL-7, IL-15, and FMS-like tyrosine kinase receptor-3 ligand promotes differentiation of im- mune cells. High Bio-activity Recombinant IL-15 Cell proliferation has been measured using MO7e human megakaryocytic leukemic cells (Figure 2). Figure1.Strategiesfortheapplicationofstemcelltherapyinthetreatmentofcancer.(Source:https://doi.org/10.3390/cells9030563)
  • 19. 19 Stem Cells as Potential Therapeutic Carriers Tumors are considered chronic wound tissue and their microenvironment, constituted by extracellu- lar matrix (ECM) and secreted paracrine factors, can attract the migration of mesenchymal stem cells (MSCs).12 MSCs are multipotent adult stem cells present in multiple tissues, including the umbili- cal cord, bone marrow, and fat tissue. Several im- mune-related cytokines are chemoattractants for MSCs to prostate cancer, osteosarcoma, multiple myeloma, and breast cancer cells, for example, CXCL16, SDF-1, CCL-25, TNF-α, IL-1β, and IL-6.13 Thus, MSCs can be utilized as potential carriers de- livering therapeutic agents in treating cancers. Featured Recombinant TNF-αα Protein Sino Biological has developed bioactive recombi- nant TNF-α cytokines from various species, includ- ing human, mouse, cynomolgus monkey, rat, fer- ret, and canine (Figure 3). Featured Recombinant IL-1ββ Protein Compared to the competing product, Sino Biological’s IL-1β protein demonstrates a higher bioactivity as shown below (Figure 4). MSCs can release nano-sized exosomes, a type of extracellular vesicles (EV) that contain various bio- logical materials, to regulate cell-cell interaction.14 Anti-tumor mRNA, siRNAs, or small molecule drugs were successfully packed into MSCs and target tu- mor niche to enhance anti-tumor effects or silence tumor-related genes.15 MSCs can also be utilized to carry anti-cancer drugs via nanoparticles (NPs). Figure2. Figure3.
  • 20. 20 MSCs-carried PLA NPs successfully targeted brain tumors, and paclitaxel-loaded NPs inhibited lung tumor growth in mice.16 Cancer cells could also be selectively killed by oncolytic viruses (OVs), while the naked OVs are easy to remove by immune cells. Stem cells, like neural stem cells (NSCs), could be promising car- riers to protect and deliver OVs to tumor sites.17 NSCs, originally present in the central nervous system, can self-renew and generate new neu- rons and glial cells. A previous study found OVs carried by human NSCs in combination with ionizing radiation and temozolomide could en- hance cytotoxicity to glioma tumor cells in vitro and increase the survival time of glioblastoma multiforme (GBM)-bearing mice.18 Both MSCs and NSCs are easier to engineer to ex- press different genes, which are responsible for the conversion of a prodrug into cytotoxic metabolites toward tumor cells.19 This gene therapy is called “suicide gene therapy”, and two phase I clinical trials have been completed. One used cytosine deaminase (CD)-expressing NSCs to convert 5-flu- orocytosine (5-FC) into tumor-toxic 5-fluorouracil (5-FU) [NCT01172964, completed]. Another con- verted ganciclovir from monophosphorylate to triphosphate form, which is more cytotoxic, by Herpes simplex virus thymidine kinase (HSV-TK) expressing MSCs [EudraCT 2012-003741-15, com- pleted]. However, the anti-tumor efficacy relies on dose control, the number of stem cells localized into the tumor microenvironment, and retention in tumor sites. In this mini-review, we introduced stem cell-related cancer treatments. Although stem cell therapy has achieved positive results, there are still some side effects to consider, for example, transformation of normal stem cells into cancer stem cells, the chron- ic GVHD after allogeneic HSC transplantation, in- creased immune response, etc. In summary, stem cell technologies have high potential for tumor treatments, but they still need further efforts to overcome the challenges before advancing further. References 1. Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. Figure4.
  • 21. 21 2. Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019;2:141-160. 3. Chu DT, Nguyen TT, Tien NLB, et al. Recent Progress of Stem Cell Therapy in Cancer Treatment: Molecular Mechanisms and Potential Applications. Cells. 2020;9(3):563. Published 2020 Feb 28. 4. Baskar R, Dai J, Wenlong N, Yeo R, Yeoh KW. Biological response of cancer cells to radiation treatment. Front Mol Biosci. 2014;1:24. Published 2014 Nov 17. 5. Hopman RK, DiPersio JF. Advances in stem cell mobilization. Blood Rev. 2014;28(1):31-40. 6. Copelan, E.A. Hematopoietic Stem-Cell Transplantation. N. Engl. J. Med. 2006, 354, 1813–1826. 7. Méndez-Ferrer, S.; Michurina, T.V.; Ferraro, F.; Mazloom, A.R.; MacArthur, B.; Lira, S.A.; Scadden, D.T.; Ma’Ayan, A.; Enikolopov, G.N.; Frenette, P.S. Mesenchymal and hematopoietic stem cells form a unique bone marrow niche. Nature 2010, 466, 829–834. 8. Lee, R.H.; Oh, J.Y.; Choi, H.; Bazhanov, N. Therapeutic factors secreted by mesenchymal stromal cells and tissue repair. J. Cell. Biochem. 2011, 112, 3073–3078. 9. Yu Z, Pestell TG, Lisanti MP, Pestell RG. Cancer stem cells. Int J Biochem Cell Biol. 2012;44(12):2144-2151 10. Codd, A.S.; Kanaseki, T.; Torigo, T.; Tabi, Z. Cancer stem cells as targets for immunotherapy. Immunology 2017, 153, 304–314 11. Iriguchi, S.; Kaneko, S. Toward the development of true“off-the-shelf”synthetic T-cell immunotherapy. Cancer Sci. 2019, 110, 16–22. 12. Aravindhan, S., Ejam, S.S., Lafta, M.H. et al. Mesenchymal stem cells and cancer therapy: insights into targeting the tumour vasculature. Cancer Cell Int. 2021, 21, 158. 13. Jung, Y.; Kim, J.K.; Shiozawa, Y.; Wang, J.; Mishra, A.; Joseph, J.; Berry, J.E.; McGee, S.; Lee, E.; Sun, H.; et al.Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat. Commun. 2013, 4,1795 14. Fuhrmann, G.; Serio, A.; Mazo, M.M.; Nair, R.; Stevens, M.M. Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. J. Control. Release 2015, 205, 35–44 15. Kooijmans, S.A.; Schiffelers, R.M.; Zarovni, N.; Vago, R. Modulation of tissue tropism and biological activity of exosomes and other extracellular vesicles: New nanotools for cancer treatment. Pharmacol. Res. 2016, 111, 487–500. 16. Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. 17. Marelli, G.; Howells, A.; Lemoine, N.R.; Wang, Y. Oncolytic Viral Therapy and the Immune System: A Double-Edged Sword against Cancer. Front. Immunol. 2018, 9, 866. 18. Duebgen, M.; Martinez-Quintanilla, J.; Tamura, K.; Hingtgen, S.; Redjal, N.; Shah, K.; Wakimoto, H. Stem Cells Loaded With Multimechanistic Oncolytic Herpes Simplex Virus Variants for Brain Tumor Therapy. J. Natl. Cancer Inst. 2014, 106 19. Sage, E.; Thakrar, R.M.; Janes, S.M. Genetically modified mesenchymal stromal cells in cancer therapy. Cytotherapy 2016, 18, 1435–1445. Additional Resource More Reagents for Cancer Research
  • 22. 22 Antibody Drug Development: Navigating Immune Response and Multivariate Logistics on the Targeted Treatment Frontier Studying these revolutionary drugs requires a comprehensive and precise approach to understand the multivariate dynamics. Tremendous efforts and ongoing studies are de- voted to minimizing symptoms while precisely targeting malignant cells that ultimately turn into cancer. One of the biggest breakthroughs to date has been the employment of monoclonal antibod- ies (mAbs) in targeted immunotherapy.These drug antibodies (DAs) now account for one-fifth of all new drug approvals. Their precision targeting has been shown to increase cure effectiveness while minimizing collateral damage caused by other treatments such as chemotherapy.6 Drug antibod- ies targeted to specific tumorigenic mutations or molecular aberrations can be mass produced in cell culture and theoretically employed to treat any cancer. Antibodies can be chimeric, humanized, or fully human and operate through three different mechanisms leading to tumor death: block a path- way, flag a cell for destruction, or deliver a drug via receptor-mediated endocytosis. Similarly, protein-based drugs are usually synthet- ic versions of proteins that are produced naturally. They modify cellular responses by either acting di- rectly with the targets of interest, recruiting other molecules to the site, or improving the function of nearby cells. For example, interleukin 2 (IL-2) is used in kidney and skin cancer treatment to inter- fere with cancer growth, recruit immune cells, and stimulate the production of immune cells that can help destroy the cancer cells. Both DAs and drug proteins can stimulate the pro- duction of anti-drug protein antibodies that can decrease therapeutic efficacy.1,2,7 Therefore, the path to implementation of DA or protein-based therapies must be navigated expertly in order to maximize patient survival. To identify the most ef- fective DA, it is important to understand and char- acterize factors that affect individual pharmacoki- netic variability post-injection and incidences of
  • 23. Mouse PBMC populations shown with CD4/IFNγ dual staining (RayBright Red and RayBright Violet 450); before & after stimulation. Stained with kit cat# 137-00007; antibody cat# 135-08029 and 135-24013. CELLULAR STAINING with RayBright ® ® dyes FACS Antibodies » Live/Dead Markers » Intracellular Staining Kits » Universal Compensation Beads » Flow Cytometry Services » Now from your trusted protein detection experts: affordably priced flow cytometry reagents. RayBiotech offers validated FACS reagents including intracellular staining kits, universal compensation beads, antibodies featuring ultra bright RayBright dyes, and more. Find out more here. Empowering your proteomics RayBiotech R Learn about our sample program RAYBIOTECH.COM / INFO@RAYBIOTECH.COM / ISO 13485:2016
  • 24. 24 immune-related adverse events (IRAEs) associated with foreign antibodies.1,2,7 While immunogenicity in preclinical species cannot predict the incidence of human ADAs, it does have utility in characteriz- ing consequences of potential ADAs.3 Crucial data in optimizing mAB preclinical devel- opment for precision cancer treatment include monitoring production of neutralizing antibod- ies, understanding factors that affect metabolism and clearance rate, measuring the binding affinity of each antibody to its target, finding the bound epitope, and discovering biomarkers to indicate treatment effectiveness or unintended down- stream effects.5,7 It is also crucial to know how and if any other health conditions impact any one of these traits. For instance, pathogens such as viral particles can bind to drug target receptors to facili- tateinfectionorotherundesiredeffects.Identifying receptor antagonists or other molecules that in- hibit unwanted receptor activation is an ongoing focus of drug discovery efforts. When monitoring efficacy of and immunogen- ic responses to an antibody therapeutic, the researcher needs access to the most reliable, Table 1. Multiple tools are in our cancer-fighting arsenal. A combination of singleplex and multiplex can be employed to unlock the secretsoftargetedtherapy.
  • 25. 25 Table2.DrugsapprovedbytheFDAtargetvariouspathwaysintumorformationandgrowth.Theyareapprovedforimplementation inspecificcancers.ImmunogenicityanddrugconcentrationlevelsarecommonlymonitoredwithELISAstudy. efficient immunoassays from manufacturers with expertise in protein detection. Fit-for-purpose as- say formats corresponding to each goal are listed in Table 1. Using a combination of these tools, re- al-time inter-human response variation to target- ed therapy molecules can be resolved regardless of the pathway they act upon. Conclusion From the first approved mAB, Pembrolizumab,4,5 to now, targeted medicine using drug antibodies has consistently offered promising horizons with- in the comprehensive field of immunotherapy.
  • 26. 26 Some, such as Tocilizumab, are even used in com- bination to reduce symptoms that come with more active forms of therapy such CAR-T,8 a bio-hack to your immune system that bolsters it to better kill uninvited cells, seen in Table 2. Regardless of the mechanism of action, continued study on these revolutionary drugs requires a comprehensive and precise approach to understand the multivar- iate dynamics. Paramount are immunoassays to discover biomarkers, individualized immunologic responses, and other potentially neutralizing par- ticles in a real-body environment. References 1. Baxi, Shrujal, et al.“Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis.”Bmj 360 (2018). 2. Eigentler, Thomas K., et al.“Diagnosis, monitoring and management of immune-related adverse drug reactions of anti-PD-1 antibody therapy.”Cancer treatment reviews 45 (2016): 7-18. 3. FDA. Guidance for Industry - Immunogenicity Assessment for Therapeutic Protein Products 2014 [Available from: https://www.fda.gov/downloads/ drugs/guidances/ ucm338856.pdf.] 4. Kwok, Gerry, et al.“Pembrolizumab (keytruda).” Human vaccines & immunotherapeutics 12.11 (2016): 2777-2789. 5. Longoria, Teresa C., and Krishnansu S. Tewari. “Evaluation of the pharmacokinetics and metabolism of pembrolizumab in the treatment of melanoma.” Expert opinion on drug metabolism & toxicology 12.10 (2016): 1247-1253. 6. Lu, RM., Hwang, YC., Liu, IJ. et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci 27, 1 (2020). https://doi.org/10.1186/ s12929-019-0592-z 7. Ovacik, Meric, and Kedan Lin.“Tutorial on monoclonal antibody pharmacokinetics and its considerations in early development.”Clinical and translational science 11.6 (2018): 540-552. 8. Si, Stephanie, and David T. Teachey.“Spotlight on tocilizumab in the treatment of CAR-T-cell-induced cytokine release syndrome: clinical evidence to date.”Therapeutics and clinical risk management 16 (2020): 705. Additional Resource Paths to Optimize Antibody Therapeutics
  • 27. 27 GPCRs: From Signal Pathways to Targeted Drug Development How to break through bottlenecks to obtain full-length GPCRs as anti- gens for drug development. Various proteins on the cell membrane are respon- sible for cell protection, internal/external materi- al transportation, and signal transmission. These membrane proteins play diverse roles in cell biolo- gy and have become a crucial target in drug devel- opment. Over 50% of the currently approved drugs target human membrane proteins. G protein-cou- pled receptors (GPCRs) are the largest family of membrane proteins.The primary function of GPCRs is to transduce a wide and diverse array of extra- cellular stimuli such as biogenic amines, peptides, hormones, neurotransmitters, ions, odorants, and photons into intracellular signals. This signaling, in turn, regulates a myriad of physiological processes, including cell metabolism, differentiation, growth, neurotransmission, and sensory perception. GPCRs are implicated in various diseases, including type 2 diabetes mellitus (T2DM), obesity, depression, HIV, cancer, Alzheimer’s disease, etc. The crucial role of GPCRs and the increased efforts in the drug discovery field have led to GPCRs becoming the most successful drug target class in the treatment of various pathologies. This review discusses the structure of GPCRs, signaling pathways, and the development pattern of various stages of targeted drugs. ACROBiosystems is focused on overcoming the challenges of GPCR preparation and providing full-length GPCR proteins to assist our customers, and collaboratively researching and developing antibody-drug and therapy strategies. Structure of GPCRs The common molecular structure of GPCRs con- sists of seven transmembrane alpha helices, and these domains divide the receptor into extracel- lular N-terminus, intracellular C-terminus, three extracellular loops, and three intracellular loops. The extracellular ring contains two highly con- served cysteine residues, which can stabilize the spatial structure of the receptor by forming disul- fide bonds. There is a G-protein binding site on the intracellular loop. In the case of CCR5, the classical GPCRs topology is shown in Figure 1.1 The GPCRs Signaling Pathways The effectors of GPCR activation are the heterotri- meric G-proteins Gα, Gβ, and Gγ. Activated GPCRs act as GEFs (guanine-nucleotide-exchange fac- tors) and exchange GDP for GTP in the Gα subunit, which activates the protein. A GPCR is free of ligand
  • 28. 28 (L) in its basal state. Gα binds to GDP and is associ- ated with Gβγ. The heterotrimeric protein complex might associate with the receptor at this point or remain free in the membrane as pictured, but once it encounters a ligand-bound GPCR, downstream signaling is initiated (Figure 2). Upon ligand bind- ing, the GPCR becomes activated and undergoes a conformational change. The resulting GTP-bound Gα separates from βγ and the active heterotrimeric proteins. Currently, the GDP-αβγ complex, with the participation of Mg2+ , exchanges GTP in the bound GDP with the cytosol to form a GTP-αβγ complex. Then the G protein is activated and separated from the receptor and simultaneously disassembles into two parts, GTP-α and βγ, which diffuse freely along the cell membrane and directly interact with ef- fector proteins, such as PLC or adenylate cyclase, which results in effector activation and initiation of a second-messenger cascade and completes the extracellular transmission of signals into the cell. The GTP in Gα is then hydrolyzed to GDP through the activity of Gα and RGS proteins (not shown), leading to Gα inactivation and reassociation of the heterotrimeric protein complex. This process rep- resents a full GPCR G-protein cycle. Progress in drug development targeting GPCRs Changing the competitive landscape GPCRs are the most intensively studied drug tar- gets comprising approximately 27% of the global market share of therapeutic drugs, with aggregat- ed sales for 2011–2015 of $890 billion. In 2017, survey data showed 481 therapeutic drugs target- ing GPCRs, which compose 34% of FDA-approved drugs and mediate their effects through at least 107 unique GPCRs. Approximately 320 drugs are currently in clinical trials, of which 35% of these drugs 64 potential novel GPCRs targets.3 Figure1.SchematicdiagramoftheclassicGPCRs1 Figure2.GPCRssignalpathway2
  • 29.
  • 31. 31 In the field of drug development that targets GPCRs, GPCRs antibodies have unique advantages over small molecule drugs: • The clearance rate in the body is lower, action time is longer, and the corresponding administration frequency is lower • The selectivity of antibodies is significantly higher than that of small molecules • However, due to the blood-brain barrier, antibody drugs cannot enter the central nervous system. Therefore, for GPCRs that are expressed in both the peripheral and central nervous systems, only the peripheral part of the drug needs to be designed; therapeutic antibodies can be developed to make the drug mainly distributed in the peripheral area and reduce the toxic side effects on the central nervous system. Overview of Leading Projects There are currently about 13 GPCRs targeted anti- body projects under development that are active worldwide, as shown in the table below. The target is mainly concentrated in CCR4, CALCRL, CCR5, GCGR, GPRC5D, GLP1R, C5AR, CB1, S1PR1, CCR8, CCR7, GPR49, and AGTR1. The representative drug is Leronlimab, a humanized IgG4 monoclonal an- tibody targeting CCR5, an HIV viral entry inhibitor. By masking CCR5, the HIV (R5) subtype is blocked from entering healthy T cells, thereby protecting these cells from viral infections. ACROBiosystems assists in the development of targeted GPCRs antibodies The first step in preparing antibodies is antigen im- munity, and the first step in immunity is the prepa- ration of antigens. Since GPCRs are seven-pass transmembrane proteins, it is extremely difficult to obtain biologically active soluble GPCRs antigens. To obtain a full-length GPCRs antigen for drug devel- opment, you need to break through two bottlenecks: • How to increase the expression? Unlike secretory proteins, the expression and display of membrane proteins are limited Table1.TargetedGPCRsdruginformationunderresearch(pharmacodia)
  • 32. 32 by the membrane area. Many membrane proteins involve functions related to material transport and signaling; overexpression on the cell membrane can cause irreversible damage to cells. These characteristics of membrane proteins greatly limit their expression. Therefore, to obtain enough full-length membrane proteins for immunity and drug development, it is necessary to design and optimize the expression interval, expression system, culture conditions, etc. Even so, the cost of obtaining milligram membrane proteins is still much higher than that of soluble proteins. • How to maintain the uniformity and activity of membrane proteins during expression and purification? The transmembrane domain of membrane proteins is highly hydrophobic, so unprotected exposure to water will lead to nonspecific protein aggregation and even denaturation. Therefore, in the process of enrichment and purification of membrane proteins, it is necessary always to maintain the hydrophilic properties of the surrounding environment of membrane proteins. The most widely used method involves removing membrane proteins from the cell membrane of the phospholipid bilayer through detergent4 and forming micelles to maintain their natural conformation and functional activity as much as possible. Furthermore, there are also methods such as Nanodisc,5 virus-like particles (VLP),6 and polymer lipid particles (PoLiPa).7 Therefore, to meet different application needs in the drug development process that targets GPCRs, ACROBiosystems has specially set up platform solu- tions with VLP, Detergent, and Nanodisc to provide full-length GPCRs proteins such as GPRC5D, CXCR4, CCR5, and CCR8. Table2.GPCRsfromACROBiosystems
  • 33. 33 In addition to GPCRs, ACROBiosystems can pro- vide a complete range of full-length multi-pass transmembrane proteins, including four-pass transmembrane protein CD20, Claudin18.2, and five-pass transmembrane protein CD133. More full-length proteins are under development. References 1. Dong HF, Wigmore K, Carrington MN, Dean M, Turpin JA, Howard OM. Variants of CCR5, which are permissive for HIV-1 infection, show distinct functional responses to CCL3, CCL4 and CCL5. Genes Immun. 2005 Oct;6(7):609-19. doi: 10.1038/ sj.gene.6364247. 2. Hanlon CD, Andrew DJ. Outside-in signaling--a brief review of GPCR signaling with a focus on the Drosophila GPCR family. J Cell Sci. 2015 Oct 1;128(19):3533-42. doi: 10.1242/jcs.175158. Epub 2015 Sep 7.. 3. Hauser AS, Attwood MM, Rask-Andersen M, Schiöth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov. 2017 Dec;16(12):829-842. doi: 10.1038/nrd.2017.178. 4. Wiseman DN, Otchere A, Patel JH, Uddin R, Pollock NL, Routledge SJ, Rothnie AJ, Slack C, Poyner DR, Bill RM, Goddard AD. Expression and purification of recombinant G protein-coupled receptors: A review. Protein Expr Purif. 2020 Mar;167:105524. doi: 10.1016/j.pep.2019.105524. Epub 2019 Oct 31. 5. McLean MA, Gregory MC, Sligar SG. Nanodiscs: A Controlled Bilayer Surface for the Study of Membrane Proteins. Annu Rev Biophys. 2018 May 20;47:107-124. doi: 10.1146/annurev-biophys-070816-033620. 6. Ho TT, Nguyen JT, Liu J, Stanczak P, Thompson AA, Yan YG, Chen J, Allerston CK, Dillard CL, Xu H, Shoger NJ, Cameron JS, Massari ME, Aertgeerts K. Method for rapid optimization of recombinant GPCR protein expression and stability using virus-like particles. Protein Expr Purif. 2017 May;133:41-49. doi: 10.1016/j. pep.2017.03.002. 7. Hothersall JD, Jones AY, Dafforn TR, Perrior T, Chapman KL. Releasing the technical‘shackles’on GPCR drug discovery: opportunities enabled by detergent-free polymer lipid particle (PoLiPa) purification. Drug Discov Today. 2020 Aug 21:S1359-6446(20)30337-8. doi: 10.1016/j.drudis.2020.08.006. Additional Resource Discover more about GPCRs
  • 34. 34 Blockade of PD-L1, an Immune Checkpoint Molecule, in a Murine Model of Melanoma Study describes the cellular response to PD-L1 blockade using monoclonal antibodies in melanoma models. Highlights Injection of anti-PD-L1 antibody in a mouse mela- noma model: • Increases levels of activated CD4+ splenic T cells. • Stimulates production of Th cytokines and suppresses pro-inflammatory cytokines. • Increases antigen-specific T cells and reduces tumor growth. Summary Immune checkpoint molecules help to regulate and limit the immune response. As tumor cells are able to exploit these mechanisms, new immuno- therapy treatments target immune checkpoints. We used a monoclonal antibody to block PD-L1 binding in a murine model of melanoma and char- acterized T cell subsets and cytokine profiles. We found that treatment with an anti-PD-L1 antibody resulted in activation of T cells, increased produc- tion of Th cytokines and chemokines, increased levels of tumor-specific T cells and caused a reduc- tion in tumor growth. Introduction Cancer treatment is classically composed of a com- bination of surgery, radiation, and chemothera- py. More recently, immunotherapy-based strate- gies have emerged as another treatment option. Immunotherapy relies on using the body’s own immune system to recognize and mount an attack against cancer cells. Immunotherapy can be broad- ly categorized into cell-based therapies in which cells are engineered ex vivo before being reintro- duced into the patient and soluble factor-based
  • 35. BioLegend products are manufactured in an ISO 13485:2016-certified facility to ensure the highest quality standards. World-Class Quality | Superior Customer Support | Outstanding Value For 20 years, BioLegend scientists have supported researchers by developing reagents for flow cytometry, multiomics, and cell bioprocessing that are critical to accelerating cancer research. Along with our expanding line of reagents, we provide educational resources that help researchers understand and decipher the complexity of cancer. We have thousands of scientist-crafted reagents engineered to combat cancer • MojoSort™ magnetic cell separation to purify and enrich target cells • GMP antibodies and recombinant proteins to activate cells • Cell-Vive™ media for T/NK cell expansion • Flow cytometry antibodies to immune profile cells • Immunoassays that quantitate cancer biomarkers Discover the difference at: biolegend.com/cancer Explore our latest infographic and other helpful resources. Cancer cells evolve new escape mechanisms. Our resources keep you apprised of developing technologies, such as CAR-T and CAR-NK cells. Explore our newest infographic to learn about this advanced immunotherapy. On our website, you can find additional educational content, including webinars, blogs, protocols, and scientific posters. Bring new cancer solutions to light. Scan the QR code to download the infographic.
  • 36. 36 therapies in which monoclonal antibodies or other proteins are introduced into the patient to mount an anti-tumor response. One approach to developing immunothera- py-based treatments is through the inhibition of immune checkpoint molecules (reviewed in 1). Immune checkpoints maintain immune homeosta- sis and limit the immune response, in part, to pre- vent autoimmunity. Tumor cells are able to exploit this system to prevent an anti-tumor response by expressing checkpoint markers typically found on antigen presenting cells (APCs). Because immune checkpoint molecules are often composed of re- ceptors and ligands, they are able to be targeted using monoclonal antibodies. Antibodies that can block ligand-receptor binding allow T cells to re- main in an activated state to mount an immune response against the tumor cells. One such immune checkpoint molecule that has been studied for use in immunotherapies is the PD-1 receptor and one of its ligands, PD-L1.2 Engagement of PD-L1 and PD-1 initiates down- stream signaling pathways ultimately leading to T cell death.3 As melanoma cells show elevated ex- pression of PD-L1, the PD-L1 blockade may be par- ticularly useful in the treatment of melanoma.4, 5 In this report, we use a GoInVivo™ anti-mouse PD-L1 antibody in a murine model of melanoma and char- acterize the T cells in the tumor microenvironment and draining lymph nodes, study the antigen-spe- cific T cell response, and phenotype the cytokine profile in the serum. Materials and Methods In vivo injection of PD-L1 Antibody BALB/cJ or C57BL/6 mice were injected with 106 B16F10 melanoma cells in the flank. Mice were subsequently treated with either 100 or 200 µg of GoInVivo™ anti-PD-L1 antibody clone 10F.9G2 (Cat. No. 124328) or isotype control (Cat. No. 400666) ev- ery 3 days for a total of 3 doses. Dosing schedule was performed as indicated in the figure legend. Tissue samples and serum were collected 14 or 24 days following initial dose. Spleen samples were collected and made into a single cell suspension for flow staining. Flow cytometry staining Cells were stained with anti-CD25 (Cat. No. 102033), CD69 (Cat. No. 104530), CD152 (CTLA-4) (Cat. No. 106305), CD279 (PD-1) (Cat. No. 109111), CD278 (ICOS) (Cat. No. 313531), I-A/I-E (MHC II) (Cat. No. 107605), CD11c (Cat. No. 117317), CD80 (Cat. No. 104729), CD86 (Cat. No. 105011), MHC pentam- er (ProImmune), and 7-AAD (Cat. No. 420403). Cells were acquired with an LSR Fortessa™ (BD Biosciences) and analyzed using FlowJo software. Microscopy Draining lymph nodes were isolated. Tissues were sectioned in a microtome cryostat at 7 microme- ters after freezing in OCT media. Sections were stained with anti-CD4 (Cat. No. 100425), CD8 (Cat. No. 100727), CD11c (Cat. No. 117346), and B220 (Cat. No. 103251), without fixation or antigen re- trieval. Images were taken on an Olympus IX83 in- verted microscope. Cytokine and chemokine measurement Cytokine and chemokine profiles were character- ized using BioLegend’s LEGENDplex™ system (Cat. Nos. 740740, 740134, 740007). A detailed protocol has been published.6
  • 37. 37 For more information regarding LEGENDplex™, please visit: biolegend.com/legendplex All reagents are from BioLegend, unless otherwise noted. Results Effective blocking of PD-L1 using a mono- clonal antibody in a melanoma model To examine the efficacy of a PD-L1 blocking anti- body in the treatment of a mouse cancer model, melanoma, we injected mice with 106 B16F10 mel- anoma cells in the flank. Mice were subsequently treated with a GoInVivo™ anti-PD-L1 blocking anti- body every three days (or as indicated). Following treatment, tissues and serum were collected for im- munophenotyping (Figure 1). Anti-PD-L1 treatment increases T cell activation status Splenic CD4+ cells were harvested 14 days af- ter treatment with anti-PD-L1 antibody or the corresponding isotype control and stained for common T cell activation markers. Mice that had been treated with anti-PD-L1 showed an activat- ed phenotype as measured by increased levels of CD25, CD69, ICOS, and CD279 (PD-1) as compared to mice treated with an isotype control (Figure 2). Importantly, CTLA-4, another immune checkpoint molecule which negatively regulates the immune Figure1.Experimentallayoutoftreatmentof anti-PD-L1antibodytreatmentinamelanomamodel. Figure2.SplenicCD4+ Tcellstreatedwithanti-PD-L1 (200µgbyI.V.injectionondays0,3,6)demonstrated anactivatedphenotype.Cellswerecollected14days followingimplantation.
  • 38. 38 response, was expressed at low levels and re- mained unchanged in mice that had been injected with anti-PD-L1. Splenic Dendritic Cells Downregulate CD80 following PD-L1 treatment Next, we examined splenic dendritic cells to deter- mine whether the increase in activated T cells cor- related with alterations in the population of APCs. For these experiments, we first gated on classical dendritic cells that are MHC II high and CD11c high. Dendritic cells (DCs) from animals that had been treated with the anti-PD-L1 antibody showed lower expression of CD80 (Figure 3). We hypothesize that downregulation of CD80 may occur to allow for CD86-CD28 interaction and less binding to CTLA-4. Anti-PD-L1 treatment results in elevated numbers of CD8+ DCs in draining lymph node To further characterize the APCs, we examined the draining lymph nodes from animals that had been injected with anti-PD-L1 and isotype controls. In animals that received the anti-PD-L1 treatment, there is an increased level of CD8a+ dendritic cells in draining lymph nodes. CD8a+ dendritic cells are APCs that have the ability to cross-present antigens to CD8+ T cells, which is a common phenomenon during tumor elimination.7 Anti-PD-L1 treatment in melanoma mod- els alters the cytokine and chemokine profile in serum To more completely analyze the immune response following anti-PD-L1 treatment, we used our LEGENDplex™ multiplex platform to analyze the chemokine and cytokine profile in serum (Figure 4). Notably, we found that animals treated with anti-PD-L1 had increased content of cytokines typically associated with a Th1 response, includ- ing IFN-γ and TNF-α. Additionally, there was an in- crease in cytokines associated with Th9 and Th22 phenotypes, IL-9 and IL-22, respectively. However, this did not correspond with an overall increase in soluble mediators, as they simultaneously limited the production of other pro-inflammatory cyto- kines including IL-12p70, IL-27, IL-33, and GM-CSF, indicating that the cytokine response in these an- imals was specific. Lastly, anti-PD-L1 treatment resulted in the elevated production of import- ant chemokines including MIP-3α, Eotaxin, MIG, and IP-10. These chemokines may play a role in anti-tumor immunity by mobilizing other cells. Figure3.SplenicDCsshowdecreasedlevelsofCD80 (top).CellswerefirstgatedonclassicalDCs(MHCII high,CD11chigh).IncreasedlevelsofCD8+ DCsin thedraininglymphnodeofanimalstreatedwith anti-PD-L1(bottom).TissuewasstainedwithCD4 (green),CD8(red),CD11c(cyan),andB220(blue).
  • 39. 39 Increase in antigen-specific CD8+ tumor infiltrating lymphocytes (TIL) following anti-PD-L1 treatment To examine antigen-specific T cells during tumor development, we implanted a B16 melanoma cell line that expresses the peptide SIYRYYGL, which is derived from the Nucleoprotein of Vesicular sto- matitis virus (498-505) and examined the tumor 24 days after implantation and antibody treat- ment. Using an MHC pentamer loaded with the SIY peptide, we were able to track CD8+ TILs. As expected, animals that received the anti-PD-L1 treatment had expanded pools of antigen-spe- cific TILs as compared to those that received the isotype control (Figure 5). Finally, as a confirma- tion of other published models,4,5 treatment with Figure5.Animalstreatedwithanti-PD-L1(100µgI.P. injectionondays7,13,16followingimplantationof melanomacellline)showincreasedantigen-specific TILsandareductionintotaltumorarea. Figure 4. Cytokine profile in the serum from treated mice shows elevation of Th cytokines and chemokines. Animals treated with PD-L1 also showed a simultaneous decrease of other pro-inflammatory cytokines.
  • 40. 40 an anti-PD-L1 antibody significantly reduced the tumor growth, as measured by tumor area, in this melanoma model. Conclusions Immune checkpoint molecules make an attrac- tive target for anti-cancer therapy, as inhibition of these pathways results in an elevated immune response, and they often rely on a ligand-recep- tor interaction. Blockade of these pathways can often be easily performed using a bioactive an- tibody, like our GoInVivo™ functional antibodies. As melanoma cells express PD-L1 to help evade an anti-tumor response, we sought to under- stand the specific cellular phenotype of the PD- L1 blockade in a mouse melanoma model. As expected, the blockade of PD-L1 resulted in ele- vated activation of T cells as measured by expres- sion of common T cell activation markers, includ- ing CD25 and CD69. Interestingly, this activation phenotype corresponds with a downregulation of CD80, another T cell co-stimulatory molecule, on APCs. We hypothesize that this downregula- tion may occur to alter the interaction of other immune checkpoint molecules, including reduc- ing binding to CTLA-4 and to allow for more in- teraction between CD86 and CD28. Using our LEGENDplex™ multiplex platform, we surveyed the cytokine and chemokine response in the se- rum following anti-PD-L1 treatment. Interestingly, anti-PD-L1 treatment increased certain cytokines associated with a Th response; however, there was not an overall increase in all soluble mediators, as the treatment also caused simultaneous down- regulation of other pro-inflammatory cytokines. Importantly, treatment with anti-PD-L1 resulted in increased levels of CD8+ tumor-specific T cells which is also correlated with a reduction in tumor size. Collectively, these results describe the cellu- lar response to the blockade of PD-L1 in a mouse melanoma model and suggest that anti-PD-L1 monoclonal antibodies may provide an effective treatment for melanoma. References 1. Pardoll DM. 2012. Nat. Rev. Cancer. 12: 252-264. 2. Zou W, et al. 2016. Sci. Transl. Med. 8: 328. 3. Jin HT, et al. 2011. Curr. Top. Microbiol. Immunol. 350: 17-37. 4. Tsai KK, et al. 2014. Hum. Vaccin. Immunother. 10 (11):3111-3116. 5. Hodi FS, et al. 2010. N. Engl. J. Med. 363: 711. 6. Lehmann, JS, et al. 2017. J. Vis. Exp. 9:129. 7. Fu, C. and Jiang A. 2018. Front. Immunol. 9: 3059.