Infectious Diseases v05
Infectious Diseases v05
Infectious Diseases v05
Sponsored by:
Accurately detect your infectious disease model
The ZE5 Cell Analyzer can detect down to 0.2 µm in cell size, can run at 100,000 events
per second with washes and low carryover, and it has an established publication catalog in
infectious diseases. Advance your research with the ZE5 Cell Analyzer, infectious disease
antibody portfolio, and newly launched, exceptionally bright StarBright Dyes, which allow
you to separate negative from positive, even for rare and dim populations.
#ScienceForward
Contents
4 50
Introduction CD4+ T Cell Activation and Associated
Susceptibility to Hiv-1 Infection in vitro
5 Increased Following Acute Resistance
Exercise in Human Subjects
Flow Cytometry: An Overview BY ALEXANDER K. HOLBROOK, HUNTER D. PETERSON, SAMANTHA A.
BY KATHERINE M. MCKINNON BIANCHI, BRAD W. MACDONALD, ERIC C. BREDAHL, MICHAEL BELSHAN
& JACOB A. SIEDLIK
Current Protocols in Immunology
16
Physiological Reports
3
Introduction
Few technologies are more central to the investigation of and uptake of the nanoparticles, tumor tissues were processed
therapeutic development for infectious diseases than flow via flow cytometry for the intracellular concentration of
cytometry. Capable of identifying and characterizing single nanoparticle as well as the activation state of tumor-draining
cells based on cellular markers, this technology is critical to lymph nodes. They report that optimized nanovaccines
understanding immune responses to infection from pathogens were highly effective at priming of antigen-specific CD8+
and the pathology of cancer. Further, the development of T-cells with antitumor efficacy, illustrating their therapeutic
therapeutics to infections such as SARS-CoV-2 or personalized potential.
medicine approaches to cancer treatment benefit from this
By introducing readers to the advantages of flow cytometry
foundational technology. This collection of articles presents
for diagnostic development and research into infectious
a sampling of studies utilizing flow cytometry to better
disease, we hope to empower users to investigate the use of
understand infectious diseases and develop diagnostic and
this technology to address their specific research or diagnostic
therapeutic interventions.
goals. For more information and resources for flow cytometry,
First, McKinnon (2018) provides an in-depth overview of the we encourage you to visit the Bio-Rad ZE5 Cell Analyzer
flow cytometry technologies available to researchers. They product page and explore the resources provided there.
discuss the different modalities of flow cytometry as well as
By Jeremy Petravicz, PhD, Editor,
their potential applications to answering research questions.
Current Protocols
Next, Russell et al. (2020) describes the process of designing
a clinical diagnostic laboratory for SARS-CoV-2 testing at The
Francis Crick Institute’s Flow Cytometry Science Technology
Platform. This includes an accounting of factors, such as References
COVID-19 restrictions, that impacted the development of
a functional diagnostic pipeline and SARS-CoV-2 assay McKinnon, K. M. (2018). Flow cytometry: An overview.
development. Continuing, Nair and Gomes-Solecki (2020) Current Protocols in Immunology, 120, 5.1.1–5.1.11. https://
describe protocols for bacterial infection of mouse models doi.org/10.1002/cpim.40
with Leptospira to recapitulate disease progression and
Russell, E., et al (2021). Adapting to the Coronavirus
physiologically relevant transmission routes. They also discuss
Pandemic: Building and Incorporating a Diagnostic Pipeline
protocols for evaluating clinical, molecular, and histological
in a Shared Resource Laboratory. Cytometry, 99: 90-99.
scores, as well as flow cytometry based-analysis of the host-
https://doi.org/10.1002/cyto.a.24248
immune response. Moving to human studies, Holbrook et
al. (2019) used flow cytometry to examine the effects of Nair, N., & Gomes-Solecki, M. (2020). A mouse model of
acute resistance exercise on CD4+ T lymphocyte activation sublethal leptospirosis: Protocols for infection with Leptospira
and replication (susceptibility) of HIV-1. They were able to through natural transmission routes, for monitoring clinical
demonstrate that acute bouts of resistance exercise increased and molecular scores of disease, and for evaluation of the
the activation state of CD4+ T lymphocytes, lead to an increase host immune Response. Current Protocols in Microbiology, 59,
in infection susceptibility and the potential for this measure e127. https://doi.org/10.1002/cpmc.127
to quantify exercise-induced changes in immune function.
Holbrook, A. K., et al (2019). CD4+ T cell activation and
Lastly, personalized treatments such as cancer vaccines
associated susceptibility to HIV-1 infection in vitro increased
holds great therapeutic potential. One emerging technology
following acute resistance exercise in human subjects. Physiol
for this area is programmable nanoparticle vaccines.
Rep, 7 ( 18), e14234, https://doi.org/10.14814/phy2.14234
Polyethyleneimine (PEI)-based nanoparticle vaccines have
been studied for the treatment of numerous infectious diseases Nam, J., et al (2021). Modularly Programmable Nanoparticle
previously. In Nam et al. (2021), they explore the potential Vaccine Based on Polyethyleneimine for Personalized
of modularly programmed PEI-based nanoparticle vaccines Cancer Immunotherapy. Adv. Sci., 8, 2002577. https://doi.
for personalized cancer immunotherapy. To characterize the org/10.1002/advs.202002577
4
Flow Cytometry: An Overview UNIT 5.1
Katherine M. McKinnon1
1
Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda,
Maryland
sample 15-color Treg cell immunophenotyping such as peptides from a vaccine, to measure
panel is shown in Table 5.1.1. immune response.
Following protein transport inhibitor treat-
Antigen specific responses ment, cells are stained for viability markers
Antigen specific responses can be measured and cell surface markers, then fixed and per-
by stimulating immune cells with a specific meabilized for intracellular staining with anti-
antigen and then looking for cytokine pro- cytokine antibodies.
duction, proliferation, activation, memory, or
antigen recognition through MHC multimers. Proliferation analysis
MHC multimers are MHC monomers (MHC-I Cell proliferation can be measured by flow
or MHC-II) that are usually biotinylated and cytometry using several different assays and
then bound to a fluorescent streptavidin back- markers. These assays use different methods to
bone in groups of 4 (tetramer), 5 (pentamer) target proliferation-related events such as in-
or 10 (dextramer). These MHC multimers are corporation of thymidine analogs (BrdU) into
"loaded" with the antigen of choice and then replicating DNA, generational tracking of in-
used to bind to T cells that recognize the anti- heritable permanent dyes (CFSE), and expres-
gen, thus indicating the level of response to a sion of proliferation related antigens (Ki67,
specific antigen. This application is commonly PCNA).
used in vaccine studies. The flow cytometry equivalent of the
3
H thymidine proliferation assay utilizes the
Intracellular cytokine analysis thymidine analogs BrdU or EdU (ethynyl de-
Intracellular cytokine analysis is performed oxyuridine) to pulse growing cells for 2 to
by treating immune cells with a protein trans- 6 hours. Following this incubation, the cells
port inhibitor (Brefeldin A or Monensin) for are stained for surface markers (optional) and
2 to 12 hours to allow for cytokines produced then fixed and permeabilized for staining the
by the cells to accumulate within the cell, en- incorporated BrdU or EdU. The BrdU pro-
abling better detection. Cells can be stimulated cedure utilizes DNase to exposed the BrdU
Flow Cytometry: with various antigens during this incubation,
An Overview for antibody staining, but the EdU procedure
utilizes a copper catalyzed click chemistry used by the immune system to maintain the
to detect the EdU. Both methods are usually homeostasis by removing cells without trig-
counterstained with a DNA-binding dye like gering an inflammatory response. This is in
propidium iodide. In addition, both the BrdU contrast to necrosis, a type of cell death that
and EdU method are compatible with staining does trigger an inflammatory response. Apop-
for additional intracellular antigen markers. tosis is the mechanism of cell death for clon-
CFSE and other similar dyes (CellTrace Vi- ally expanded T cells following an immune
olet, FarRed, etc) cross the cellular membrane response, for self-targeting T cells, for autore-
in living cells and bind covalently and per- active B cells, and multiple other cells in the
manently to intracellular structures (usually to immune system.
lysine residues or other amines). The daughter The detection of apoptosis by flow cytom-
cells of each subsequent generation inherit the etry utilizes multiple targets along the cascade
dye allowing for long term analysis of prolif- of apoptosis-associated events. The transloca-
eration. This technique is very useful when tion of phosphatidylserine to the outer layer
following proliferation resulting from long- of the plasma membrane is detected by An-
term antigen stimulation. An example of CFSE nexin V staining, the endonuclease digestion
staining is shown in Figure 5.1.1. of DNA is detected by the TUNEL (TdT dUTP
Expression of proliferation-related anti- Nick End Labeling) assay, and the activation
gens can also be used as a marker for pro- of Caspases can be detected by antibodies
liferation. Ki67 is a protein expressed during and dyes, mitochondrial apoptosis is targeted
all phases of cell proliferation but not during by dyes that determine mitochondrial mem-
cell quiescence. Proliferating cell nuclear anti- brane potential and chromatin condensation in
gen (PCNA) is required for DNA replication. the nucleus detected by staining with Hoescht
The presence of either Ki67 or PCNA is an in- 33342.
dicator of cell proliferation. Ki67, PCNA, and Annexin V is a phospholipid binding pro-
BrdU staining in the same cells is shown in tein that binds to phosphatidylserine when it
Figure 5.1.2. is translocated to the outer layer of the cel-
lular membrane during apoptosis. A viability
Apoptosis analysis exclusion dye (like propidium iodide) should
Apoptosis, or programed cell death, is a be used when staining with Annexin V to con-
phenomenon that is frequently examined in firm that the binding is happening on the outer Immunofluore-
immunology and other fields of study. It is scence and Cell
surface of the cellular membrane. Sorting
TUNEL is a technique that utilizes the abil- protein–protein interactions. These method-
ity of terminal deoxynucleotidyl transferase ologies revolutionized the detection and iso-
(TdT) to label the ends of DNA breaks associ- lation of cells where the fluorescence is de-
ated with apoptosis with dUTP (deoxyuridine tected only in response to surrogate (Han et al.,
triphosphate) or BrdU. The dUTP or BrdU are 2014). This technology is used for multiple
labeled with a fluorchrome for detection and applications, for example in vivo tracking of
the cells are counter stained with a DNA dye transplanted cells, bacterial or viral infections,
prior to data acquisition. and gene knockout in cells to further elucidate
The caspase signaling pathway is activated gene function.
in most cases of apoptosis. This is targeted by
using intracellular staining and antibodies that Cell cycle analysis
are specific to the active form of caspase 3. Cell cycle analysis assays consist of stain-
There are additional assays that utilize fluo- ing DNA with a saturating amount of DNA
rogenic substrates that when exposed to cas- binding dye. In most cases, the cells are fixed
pase activity are cleaved and then emit fluo- with a 70% ethanol solution which permeabi-
rescence. lizes the cells and then stained with the dye
Mitochondrial apoptosis does not always (PI, 7AAD, DAPI). However, there are dyes
utilize the caspase pathway so different meth- that can enter living cells and stain DNA with-
ods are used for detection. Most of these meth- out harm to the cells such as Hoescht 33342.
ods examine mitochondria membrane poten- In this type of analysis, samples are acquired
tial such as using the dye JC-1. However, there at a low flow rate with linear amplification and
is an antibody against APO2.7 that is localized then analyzed using ploidy modeling software
on the mitochondrial membrane and only ex- to determine the cell cycle phases.
pressed during apoptosis.
Signal transduction flow cytometry
This application uses antibodies made
Molecular Biology against resting and phosphorylated signaling
molecules. The use of these reagents and spe-
Fluorescent protein analysis cialized buffers in staining panels allows for
Fluorescent proteins (GFP, mCherry, YFP, the study of signaling pathways in mixed pop-
mRuby, etc) are used as markers for protein ex- ulations of cells.
pression. Typically, cells are transfected with
a plasmid that contains a promotor sequence RNA flow cytometry
and encodes for a gene of interest along with RNA flow cytometry combines flow cy-
a fluorescent protein. The expression of the tometry with fluorescent in situ hybridiza-
fluorescent protein is used as an indicator for tion (FISH) to detect RNA expression along
the expression of the gene of interest. More re- with protein expression. This technique re-
cently, the expression of a split bi- or tri-partied quires staining panel optimization since not
Flow Cytometry: fluorescence complementation linked to other all fluorochrome conjugated antibodies will
An Overview
proteins allow detection of RNA–protein and withstand treatment at 40°C for multiple 1 hr
Immunofluore-
scence and Cell
Sorting
Emma Russell, Ana Agua-Doce, Lotte Carr, Asha Malla, Kerol Bartolovic, Dina Levi,
Carl Henderson, Debipriya Das, Hefin Rhys, Philip Hobson, Sukhveer Purewal, Andrew Riddell*
AT the beginning of February 2020, the COVID-19 outbreak Consortium that successfully developed a diagnostic polymer-
in the UK gathered pace and it seemed highly probable that ase chain reaction (PCR) pipeline (1,2). The Flow STP was
the United Kingdom would follow similar lockdown restric- engaged to support COVID-19 research at the Crick, helping
tion policies seen in other European countries. The Crick’s to develop novel flow cytometry assays utilizing SARS-CoV-2
Flow Cytometry Science Technology Platform (Flow STP) virus-specific proteins. The STP was tasked with building a
helped prepare scientists to finish current experiments and new clinical diagnostic lab and running a novel enzyme-
store experimental material during lockdown to enable an linked immunosorbent assay (ELISA) that had been devel-
efficient restart upon the eventual lifting of the restrictions. oped for detecting antibodies against the S1 spike of the
During this time safety measures were introduced into the SARS-CoV-2 virus. The key processes required are outlined
Flow STP, including social distancing, that directly reduced in Figure 1.
the number of instruments available for use. When lockdown
was announced in the United Kingdom on the 23rd March SARS-CoV-2 ASSAY DEVELOPMENT
2020 the total number of staff allowed into the Crick was
The Flow STP was involved in the development of serology
reduced to a core group of key workers. The Flow STP was
studies, comprising assays, and diagnostic tests focused on
given key worker status and operated to provide flow cyto-
antibodies present in serum. Initially, work began on three
metry to Crick scientists whose sole focus was now
assays: one cell-based, one bead-based, and an ELISA. The
COVID-19.
early development of these required many steps, beginning
In the lead-up to lockdown communications were sent
with a feasible idea through to procuring and testing reagents
to scientists to highlight the possible implications of the pan-
and methods. During protocol development testing was
demic on Flow STP operation. There were significant changes
required to ensure their precision, accuracy, and coherence.
in the way the Flow STP operated due to the challenges faced
To accommodate the assay development and pipeline a
outlined in Table 1.
restricted access containment level 2 (CL2) laboratory was
In early May 2020 the Crick prepared to support testing
created, housing a ZE5™ (Bio-Rad, Hercules, CA) and LSR
during the pandemic by establishing the Crick COVID-19
Fortessa™ (BD Biosciences, San Jose, CA), for analysis of
The Francis Crick Institute, Flow Cytometry Science and Technology Platform,
London, UK
Published online 22 November 2020 in Wiley Online Library
Received 18 August 2020; Revised 23 September 2020; Accepted 19 (wileyonlinelibrary.com)
October 2020
DOI: 10.1002/cyto.a.24248
Grant sponsor: Cancer Research UK, Grant number: FC001999; Grant sponsor:
© 2020 The Authors. Cytometry Part A published by Wiley Periodicals LLC on
Medical Research Council; Grant sponsor: Wellcome Trust
behalf of International Society for Advancement of Cytometry.
Additional Supporting Information may be found in the online version of this
This is an open access article under the terms of the Creative Commons
article.
Attribution License, which permits use, distribution and reproduction in any
*Correspondence to: Andrew Riddell, Flow Cytometry Science Technology medium, provided the original work is properly cited.
Platform Lead, The Francis Crick Institute, 1 Midland Road, London NW1 1AT,
UK. Email: andy.riddell@crick.ac.uk
16
SRL COMMUNICATION
Table 1. Table to highlight the approaches taken to overcome each challenge faced throughout the pandemic and pipeline creation
CHALLENGE APPROACH
As a consequence of social distancing staffing levels Staff predominantly worked from home unless required to attend the
had to be reduced. Crick to aid in assay development.
Other members of the team were furloughed under the government
Coronavirus Job Retention Scheme.
Employees received mandatory weekly COVID-19 swab tests to confirm
suitability to work.
A designated place was created in the lab where scientists could safely
drop off and collect samples to reduce face-to-face contact. Users
communicated with scientists via online video calling. Use of some
instruments in close proximity was restricted, there was reduced
occupancy in laboratories and extended cleaning regimes were put in
place.
Training was suspended to prevent any potential spread of infection, and
remote support was provided as required.
The strict and constantly changing timelines for the The Crick worked closely with a UKAS accredited medical laboratory to
development of both the assays and pipeline. quickly meet the governance requirements allowing swift assay
development.
Introducing the different approach required to work The team was able to draw on diagnostic expertise from existing staff
in diagnostics versus research. members within the Crick with a diagnostic background.
We consulted with qualified biomedical scientists within the Crick to
highlight where processes need adapting to conform to diagnostic
standards.
Restrictions imposed by space available to A complete overhaul of both the layout and laboratory equipment was
accommodate equipment and staff. undertaken in less than a week to meet the requirements of a CL2
diagnostic facility.
Balancing the COVID-19 pipeline with usual Users previously trained in cell sorting were required to perform their
workload responsibilities. own sorts and analysis and were encouraged to help their nontrained
colleagues to use the facility with oversight from the STP staff.
During lockdown non-essential flow cytometry work was suspended and
external users were banned.
Incorporating COVID-19 compliant practices into Use of PPE including masks and face shields.
training regimes for new users.
Development of a series of online videos to provide an alternative to face
to face contact.
serological samples. A second CL2 laboratory was created for pipeline. The pipeline is in the process of attaining this
ELISA testing. accreditation.
Other standards and recommendations include those
from the Royal College of Pathologists’ “The retention and
THE ROLE OF GOVERNANCE IN DIAGNOSTICS storage of pathological records and specimens” (5). These
outline how to store samples and records required in relation
To operate as a clinical diagnostic lab specific standards must
to each sample. In the United Kingdom (and Europe) all per-
be attained. The United Kingdom Accreditation Service
sonal data must be handled in accordance with data protec-
(UKAS) is a government-recognized body that provides certi-
tion laws and regulations (the GDPR and UK Data Protection
fied testing, inspection, and calibration services together with
Act 2018) (6) and all internal recording and sample handling
an oversight function. UKAS requires medical laboratories to
systems must be fit for purpose.
have International Organization for Standardization (ISO)
Completing adequate staff training was imperative and
15189:2012 certification for quality and competence to run a
staff who built the necessary workflows and resources of the
clinical laboratory (3, 4). To meet these standards the Flow
SARS-CoV-2 ELISA assay trained other staff to become com-
STP created new clinical-grade protocols and standard oper-
petent in all processes of the pipeline. Those members of staff
ating procedures (SOPs). The key requirements are auditabil-
trained others until all were fully trained. A training log was
ity and the ability to connect the samples to their results;
kept for each member of staff as part of the audit process and
these shaped the protocols and the design of the Crick’s new
17
SRL COMMUNICATION
Figure 1. Outline of the key processes involved in pipeline development and implementation.
updated when new assays were introduced. The team received • The temperature of fridges and freezers via a sample man-
information regarding how sample handling adhered to the agement system to give an independent secondary
Human Tissue Act 2004 in the United Kingdom and training recording.
in the following areas:
• Good Clinical Practice (GCP) and the legal framework of a Access to sample material was restricted in the interest
diagnostics lab. of safety, information security, and confidentiality. Other gov-
• Data protection laws (the GDPR and UK Data Protection ernance comes from the Crick’s institutional policy
Act 2018) (6). documents.
18
SRL COMMUNICATION
Figure 2. The flowchart outlines the steps taken along the pipeline from the moment of sample receipt to running the assay. The yellow
pathway follows the person performing the tasks and making decisions. The blue pathway indicates a second person who checks the
action of the person following the yellow pathway, as well as, tracking the sample manually along the pipeline. Rectangles denote
actions, diamonds indicate decision points and circles provide the two end points.
designing the pipeline the primary concern was to ensure the instruction before samples were processed. This removed the
sample could not be separated from its result. A two-person chance of processing a sample that was not meant to be tested
approach, as outlined in Figure 2, was used to enhance the or one that met rejection criteria which would potentially
level of security within the decision making and verification invalidate the result.
processes. SOPs were created to give clear guidelines for how As the pipeline developed, samples were received from
the process should be carried out. These documents will be multiple sources for various uses which required either diag-
extended and added to over time. nostic, research based or quality control output channels. The
Logs were created to identify the sample status across the sample reception process was evolved to accommodate this.
entire pipeline and allowed identification of potential
breakpoints along the way. For each breakpoint a contingency
plan was developed in case of errors as shown in Table 2. The
WHY NOT AUTOMATE FROM THE OUTSET?
contingencies were then graded on a traffic light system: red, The pipeline was started manually to get it working as quickly
amber, and green to indicate the level of impact on the outcome as possible and to enable a more efficient and timely digital
of the assay and dictate the level of verification required. development process. The parameters for a Laboratory Infor-
As part of this process quarantine measures were intro- mation Management System (LIMS) evolved from building
duced ensuring that unscheduled samples that arrived with- the pipeline physically from scratch. Pressure tests in the
out manifests, or samples with specific rejection criteria were form of dummy runs with batches of mock samples were
stored in a separate area. This was a location within the fridge undertaken to test the robustness of the manual pipeline.
that was separated by quarantine criteria notifying the team These steps were then used as a framework for digitization as
that the reporting body should be contacted for further shown in Table 3.
19
SRL COMMUNICATION
Table 2. Demonstrates examples of contingencies in place and an accurate serological evaluation. Other ways of testing for
their grading in case of operator error or equipment failures SARS-CoV-2-specific antibodies were developed including
throughout pipeline.
both ELISA and flow cytometry assays with cells and beads.
ISSUE CONTINGENCY GRADE The development of such tests for diagnostic purposes
required each variable to be tested and validated. There is
Aspirator breaks. Use multi-channel Green
currently no gold standard for SARS-CoV-2 serological tests
pipette to remove
so we used different commercial assays to cross validate our
wash.
internal assays. Although mostly concordant, the different
No precoated plates Masterplate stored in Green
tests showed disparity between some positive samples. The
prepared. fridge at 4 C for up
disparity may be due to biological differences or varying assay
to 72 h.
sensitivities. Importantly, the negatives remained concordant.
Barcode scanner Manually record the Amber
Unlike in research, a diagnostic pipeline requires strictly
breaks. barcode number in
defined protocols to minimize variability and ensure reliable
the paper log and on
reporting to external parties. The team identified steps in the
the 2 ml tube,
pipeline where human error could possibly be introduced and
proceed as normal.
incorporated extra checks or contingencies to prevent this.
Clumpy/hemolyzed Record this Amber
The first obvious step for introduction of human error was in
sample upon information in log
the creation of the 96-well master plate. As well as human
aliquoting. and continue.
pipetting errors there was potential to introduce mismatches
Plate reader failure. Add 50 μl of 1 M Red
between sample position and sample ID. We therefore auto-
NaOH to quench
mated this step using a robot to decant samples from their
plate at 15 min after
tubes into a recorded position on the master plate. As the
addition of substrate,
robot had not been previously used in a diagnostic pipeline
store in dark by
several tests and validations had to be done. At least five runs
wrapping in foil at
of an ELISA assay with large sample numbers (n > 30) were
room temperature
carried out comparing the reproducibility of the robot to
until problem can be
human pipetting.
resolved.
We calculated the intra and inter percentage coefficients of
If >12 h repeat assay.
variation (% CV) to measure the variability of samples both
Any steps not Terminate assay and Red
within and between each run. We found that dispensing sam-
completed as per repeat next day.
ples using the robot gave an intra-assay % CV of 13.07 ± 14.86
SOP/missed.
(mean ± SD) while human pipetting resulted in an intra-assay
% CV of 7.85 ± 4.74%. Conversely, robotic and human
pipetting resulted in interassay % CVs of 19.45 ± 7.51 and
23.50 ± 11.63 respectively as shown in Figure 3. This suggested
Once all the parameters had been defined and tested that while robotic sample dispensing increased intra-assay vari-
automating the process removed multiple sources of potential ation, believed to be due to uneven volume dispensing, it
human error. This increased not only the efficiency of the slightly decreased interassay variation. Importantly, we found
pipeline by reducing time spent performing manual steps but the interassay variation in outcomes for each sample
also freed up time spent by staff performing the tasks. (as detected, not detected, or indeterminate) to be lower for
Initially, we used a commercial electronic sample track- robot versus human pipetting as shown in Figure 4. Taken
ing system already in place at the Crick. After collaborating together and with time constraints these data indicated that the
with the Scientific Computing STP a web app-based LIMS assay uncertainty was within reasonable limits and the use of
was developed that encompassed all the specific requirements the robot for pipetting resulted in more consistent sample
and parameters the commercial option could not. This outcomes.
custom-built system relied upon barcoding and robot tech- For validation it was necessary to demonstrate specificity,
nology to electronically track the samples through the pipe- sensitivity and reproducibility within the results of the pipe-
line. After further rounds of dummy runs and the creation of line (8,9). Using the ELISA assay as an example we evaluated
new SOPs, to include the new LIMS, the pipeline was fully reproducibility by preparing multiple master plates with dif-
automated leaving the manual system in place as a contin- ferent operators. Our test batch of samples contained posi-
gency for any potential point of failure. tives (n = 45) and negatives (n = 40) as previously confirmed
by an independent laboratory through PCR and a commer-
cially available assay. The criterion set for specificity was that
VALIDATING THE PIPELINE tests should not report a negative sample as positive. The cri-
Despite the abundance of recent studies on SARS-CoV-2 rig- terion for sensitivity was that tests should not report a posi-
orous testing and knowledge of the immune response to the tive sample as negative. Finally, for reproducibility the
disease was still lacking. This posed a problem with reporting criterion was that results should be consistent among the five
20
SRL COMMUNICATION
Table 3. Breakdown of the steps involved in the pipeline, how each step is manually recorded and the proposed electronic alternative
Samples received by the Crick from Sample delivery recorded via Crick Notification system alerts
courier. usual processes
Flow team that samples have been
received
Samples collected from Crick drop off Operator signs the paper log to Notification system alerts
point and transported in the transfer confirm collection of samples
box via shortest route.
Flow team the samples are in transit to
the lab
Sample container placed in hood and Operator records if any sample Operator records if any sample
visually inspected for leakages. leakages seen on paper log leakages are seen on LIMS
Samples scanned to confirm receipt. Record made on the paper log of Notification system alerts sender
number on box samples have been received
Quarantine process initiated for any Quarantine logbook on sample fridge Fridge Log tracks what samples are in
samples that are unexpected, requires signing into the fridge with which section of quarantine and
incorrect or missing. specific location and reason for notification system alerts sender
quarantine. samples are waiting.
For each sample 250 μl of serum will One barcode is stuck to the paper log Notification system records which
be transferred into one 5 ml tube to confirm which tubes have been samples match those we are expected
and has a bar code attached. received. to receive and highlights any
“Quarantine” samples.
Serum quality logged for any Any samples that are hemolyzed or Option on the dashboard to select
hemolyzed or viscous samples. viscous are marked next to their sample is hemolyzed or viscous for
barcode on the paper log. specific samples.
Up to 40 samples are placed onto the Table filled in with sample barcodes. The Plate barcode is scanned.
robot and duplicated in a 96-well
plate (Library Plate) with one Plate
Manifest barcode.
A copy of the Plate Manifest barcode is The robot individually scans the
attached. barcode of each sample and tracks it
into whichever well the sample is
placed.
A plate manifest layout document is
created with the date and time.
40 original 5 ml tube samples tracked Date and time of the 5 ml samples FreezerPro® records date and time the
for −80 C storage and the date and recorded. 5 ml samples are being tracked.
time marked on the rack.
Sample tubes are stored in the fridge Operator signs samples into fridge N/A
(4 C) until ELISA is complete. using paper log.
ELISA process carried out. SOP checklist double signed to confirm Electronic checklist updated when each
operator has carried out each stage. section of ELISA SOP completed.
ELISA plate placed on reader. Plate barcode is recorded on paper log. Scan plate onto reader so that
notification system logs plate
barcode as “being read”.
Results generated for reporting. Print a copy of the CSV. File generated Electronic reporting process carried out
by the plate reader and store. via LIMS.
Completed samples are transferred Sign in and sign out sheet on each FreezerPro®.
from fridge to freezer. fridge detailing what samples are
where.
21
SRL COMMUNICATION
Figure 3. The variability between robot and manual pipetting. (A) Intra-assay percent coefficients of variation (% CV) for each sample
across separate subplots for each experimental run. (B) Interassay % CV for each sample, collapsed across experimental runs.
different repeats. Our results showed the tests were reproduc- because assays could be conducted at different time points.
ible, specific and sensitive as defined in Table 4. For this the same set of samples kept at 4 C were tested at
Most diagnostic assays do not include standard curves different time points over 2 weeks.
because they are designed to provide a “YES” or “NO”
answer. During the process of pipeline validation however, we
included standard curves in some tests. These standard curves
provided us with parameters such as limits of detection
THE NEW NORMAL
(20.41 pg/ml equivalent of positive control antibody with 95% In June 2020 lockdown restrictions were eased; members of
confidence interval [17.67, 24.65]) and dilution linearity as the Crick started to return to work in a phased approach and
shown in Figure 5 and defined in Table 4. Dilution linearity normal staff duties began to increase. The decision was made
demonstrated that the coating did not interfere with accurate to split the Flow STP into two groups on a rota basis to
detection or result in nonspecific binding. enable social distancing and to prevent the entire team having
While our methods covered all the parameters required to self-isolate if one member tested positive for COVID-19.
for confidence it remained necessary to evaluate each sample Careful management of staff time was required taking into
individually. It was imperative to evaluate sample stability consideration both annual and sick leave requirements.
22
SRL COMMUNICATION
Figure 4. The reproducibility between robot and manual pipetting. (A) Alluvial plots showing how the outcome for each well changes when
dispensed manually or with the robot. Separate subplots are drawn for each experimental run. Each horizontal ribbon represents a single well
and is colored by its manual pipetting outcome. (B) The same data as in A showing how the outcome for each well changes between
experimental runs. Separate subplots show manual and robot pipetting data.
Table 4. Summary of parameters used to validate the This reduction in staff numbers combined with the run-
performance and suitability of the ELISA assay ning of the clinical diagnostic pipeline had a major impact on
PARAMETER RESULT the services the Flow STP could provide. The pre-COVID
workload could not be sustained and was streamlined. Prior
1 Reproducibility. 1.18 ± 0.83% of samples had to the lockdown we had pretrained scientists who were able
discordant outcomesa to perform cell sorting both for themselves and their col-
2 Sensitivity. 87 ± 3.0%a leagues. Post lockdown this relieved a large burden on the
3 Specificity. 100%a Flow STP and enabled scientists to continue their research
4 Uncertainty. Intra and interassay % CVs of independently. The online booking system was adapted and
8.08 ± 7.23 and 14.57 ± 6.99, we implemented a more consultative approach to staff plan-
respectivelya ning and cell sorting which helped triage service requests.
5 Limits of 20.41 pg/ml equivalent of positive The Flow STP continues to aid researchers with COVID-
detection. control antibody with 95% 19 experiments. As previously discussed flow-based serologi-
confidence interval [17.67, cal assays continue to be under development internally. These
24.65]b will be added to the clinical diagnostic pipeline based on their
6 Dilution Linear relationship between log success. The reality of the “new normal” for the Flow STP at
linearity. positive control antibody the Crick is to continue and build on our work on serology
concentration and log assays as a clinical diagnostics lab as well as maintaining an
absorbanceb effective flow cytometry service to the Crick research labs.
a
Although our time has been repurposed, putting constraints
Mean ± standard deviation of five runs with 40 samples per run.
b
From six independent standard curves of positive control anti-
on our usual duties, we are delighted to contribute our newly
body from 0.8 to 200 ng/ml. adapted services to assist in this global pandemic.
23
SRL COMMUNICATION
Figure 5. Concentration of the anti-S1 spike protein antibody CR3022 (the assay positive control antibody) against absorbance at 405 nm
after performing six independent ELISAs. (A) is plotted on a natural scale and (B) is plotted on a semi-log scale. Two technical replicates
per plate are shown. Note that except for the saturating concentration the relationship between log concentration and log absorbance is
linear which indicates dilution linearity.
CONCLUSION their advice on protocols and for helping us to set up the CL2
laboratories. Secondly, Richard Byrne, Gill Adams, Mathew
The challenges faced by the Flow STP can be separated into
Sargent and all the hub team for responding to our instru-
those created by COVID-19 regarding the changes required
mentation requests so rapidly. With regards to ELISA assay
to everyday working practices and those posed by the uncer-
development we would like to thank Laura McCoy from
tainty and novelty of creating the pipeline as outlined in
UCL, George Kassiotis and his team for their help and Peter
Figure 1. The practices that have been put in place to over-
Cherepanov for developing the protein coating for the ELISA
come the first continue to be instilled in the current climate
as well as beads used in the bead assay. Also, thank you to
and are expected to ease off in line with both government
Jerome Nicod for providing us with the robots, to Theo San-
advice and updates to Crick policies. While this project would
derson for programming them and Phil Walker for setting up
have been possible without it, the team recognizes the advan-
the barcoding system. In addition we would like to acknowl-
tage of having diagnostic expertise to advise and guide the
edge the Crick COVID-19 Governance Team for their sup-
pipeline development process. Particularly to maximize effi-
port and guidance, to thank Richard Stone and Emma Nye
ciency by avoiding potential pitfalls. Should the reader be
for reviewing the pipeline and the Crick’s Scientific Comput-
looking to perform a similar process the authors recommend
ing STP for helping to set up a reporting strategy and the
consulting with diagnostic expertise to facilitate this. Incorpo-
LIMS. Many thanks to Kathleen Evans for her invaluable
rating the pipeline into our daily tasks while at reduced
input. Finally, the team would like to extend their thanks to
staffing levels remains an ongoing challenge. The Flow STP
all those who have not specifically been mentioned but who
have successfully carried out the development and implemen-
were fundamental to making this project possible. This work
tation of a novel SARs-CoV-2 ELISA pipeline with the confi-
was supported by the Francis Crick Institute which receives
dence that it is fit for purpose and ready for handover to be
its core funding from Cancer Research UK (FC001999), the
maintained outside the Flow STP when required.
Research Councils UK Medical Research Council (FC001999),
and the Wellcome Trust (FC001999).
ACKNOWLEDGMENTS
To set up a clinical diagnostics lab in such a short time frame
required a huge level of collaboration and assistance from
AUTHOR CONTRIBUTIONS
other members of the Crick. Firstly, thank you to the Safety, Emma Russell: Conceptualization; formal analysis; project
Health and Sustainability team as well as Nicola O’Reilly for administration; validation; writing-original draft; writing-
24
SRL COMMUNICATION
review and editing. Ana Agua-Doce: Conceptualization; • Sera is not HTA relevant material.
formal analysis; project administration; validation; writing- • Samples used were leftover serum samples from routine
original draft; writing-review and editing. Lotte Carr: clinical testing of individuals, not taken for a purpose
Conceptualization; formal analysis; project administration; within the remit of Research Ethics Committees.
validation; writing-original draft; writing-review and editing. • Individual samples were anonymized such that no personal
Asha Malla: Writing-original draft; writing-review and identifying information was present.
editing. Kerol Bartolovic: Conceptualization; writing-original
draft; writing-review and editing. Dina Levi: Writing-original These samples were provided with appropriate consents
draft; writing-review and editing. Carl Henderson: Writing- from the source.
original draft; writing-review and editing. Debipriya Das:
Writing-original draft; writing-review and editing. Hefin
Rhys: Conceptualization; formal analysis; project administra- LITERATURE CITED
tion; validation; writing-original draft; writing-review and 1. Houlihan CF, Vora N, Byrne, T, Lewer, D, Kelly, G, Heaney, J, Gandhi, S, Spyer, MJ,
Beale, R, & Cherepanov, P, et al. Pandemic peak SARS-CoV-2 infection and serocon-
editing. Philip Hobson: Conceptualization; formal analysis; version rates in London frontline health-care workers. Lancet 2020;396:e6–e7.
project administration; validation; writing-original draft; 2. Aitken, J, Allen, Z, Ambler, R, Ambrose, K, Ashton, E, Avola, A, Balakrishnan, S,
Barns-Jenkins, C, Barr, G, & Barrell, S , et al. Scalable and robust SARS-CoV-2 testing
writing-review and editing. Sukhveer Purewal: Conceptuali- in an academic center. Nat Biotechnol2020;38:927–931.
zation; project administration; writing-original draft; writing- 3. Medical laboratories — Requirements for quality and competence (ISO 15189:2012).
EUROPEAN COMMITTEE FOR STANDARDIZATION Ref. No. EN ISO 15189:
review and editing. Andrew Riddell: Conceptualization; 2012
formal analysis; project administration; resources; writing- 4. Burnett DA. Practical Guide to ISO 15189 in Laboratory Medicine. London, UK:
ACB Venture Publications, 2002.
original draft; writing-review and editing. 5. Royal College of Pathologists. The Retention and Storage of Pathological Records and
Specimens. 5th ed. London, UK: Royal College of Pathologists, 2005.
6. https://libraryfaqs.worc.ac.uk/faq/230277
DECLARATION 7. https://ukneqas.org.uk
8. https://www.oie.int/fileadmin/Home/eng/Health_standards/aahm/current/chapitre_
The following rationale was applied to conclude that no ethics validation_diagnostics_assays.pdf
approval was required to conduct this assay development: 9. (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4541289/)
25
A Mouse Model of Sublethal
Leptospirosis: Protocols for Infection with
Leptospira Through Natural Transmission
Routes, for Monitoring Clinical and
Molecular Scores of Disease, and for
Evaluation of the Host Immune Response
Nisha Nair1 and Maria Gomes-Solecki1,2
1
Department of Microbiology, Immunology and Biochemistry, The University of Tennessee
Health Science Center, Memphis, Tennessee
2
Corresponding author: mgomesso@uthsc.edu
Nair and
Gomes-Solecki
Figure 1 Workflow for infection of mice with pathogenic Leptospira and subsequent collection of biological
samples.
NOTE: All animal experiments require approval by the local ethical and animal handling
offices.
Materials
Pathogenic Leptospira species (Leptospira): we use Leptospira interrogans serovar
Copenhageni strain Fiocruz L1-130 (ATCC #BAA-1198)
EMJH base medium (BD, cat. no. 279410)
Leptospira enrichment EMJH (BD DifcoTM , cat. no. 279510)
DNeasy Blood & Tissue Kit (Qiagen, cat. no. 69506)
Maxima Probe/ROX qPCR 2× mix (Thermo Fisher Scientific, cat. no. K0233)
Leptospira-specific TAMRA probe and 16S rRNA primers (Eurofins Scientific, Nair and
Gomes-Solecki
Table 1)
Leptospira specific
Lepto_F CCCGCGTCCGATTAG
Lepto_R TCCATTGTGGCCGAACAC
LIC_TAMRA CTCACCAAGGCGACGATCGGTAGC
Mouse specific
β-actin _F CCACAGCTGAGAGGGAAATC
β-actin_R CCAATAGTGATGACCTGGCCG
β-actin_TAMRA GGAGATGGCCACTGCCGCATC
TNF-α_F CACACTCAGATCATCTTCTCAAAAT
TNF-α_R AAGGTACAACCCATCGGCTGGCA
TNF-α-TAMRA AGCCTGTAGCCCACGTCGTAGCAAAC
IFN-γ _F CAAGTGGCATAGATGTGGAAGAAA
IFN-γ_R CTGGCTCTGCAGGATTTTCA
IFN-γ-TAMRA GGAGGAACTGGCAAAAGGATGGTGAC
RANTES_F AGTGCTCCAATCTTGCAGTCGT
RANTES_R CTTCTTCTCTGGGTTGGCACACACT
RANTES-TAMRA TTGTCACTCGAAGGAACCG
MIP2_F TGACTTCAAGAACATCCAGAGCTT
MIP2_R CTTGAGAGTGGCTATGACTTCTGTC
MIP2-TAMRA TGACGCCCCCAGGACCCCA
KC_F CGAGGCTTGCCTTGACCCTGAA
KC_R GGGACACCTTTTAGCATCTT
KC_TAMRA CCCTTGGTTCAGAAAATTGTCCA
IL-4_F TGTACCAGGAGCCATATCCA
IL-4_R TTCTTCGTTGCTGTGAGGAC
IL-4_TAMRA ATCCATCTCCGTGCATGGCG
iNOS_F GCTGGGCTGTACAAACCTTC
iNOS_R GCATTGGAAGTGAAGCGTTTC
iNOS_TAMRA GGCAGCCTGTGAGACCTTTGAT
ColA1_F TAAGGGTACCGCTGGAGAAC
ColA1_R GTTCACCTCTCTCACCAGCA
ColA1_TAMRA AGAGCGAGGCCTTCCCGGAC
Additional reagents and equipment for euthanasia (see Current Protocols article:
Donovan & Brown, 2006a) injection (see Current Protocols article: Donovan &
Brown, 2006b), and anesthesia (see Current Protocols article: Donovan &
Brown, 2001) of rodents
Culture of Leptospira in vitro
1. Thaw 250 μl of a frozen stock and immediately add to 4.25 ml of EMJH base
medium. Add 500 μl of the supplement (BD DifcoTM Leptospira enrichment EMJH)
and add 5-fluorouracil at a final concentration of 100 μg/ml in a 14-ml round-bottom
tube.
2. Place cultures at 30°C up to 4 weeks.
5-Fluorouracil prevents contamination of the culture by inhibiting the growth of other
organisms, but does not affect the growth of Leptospira. When reviving a frozen stock,
inoculate quickly after thawing into supplemented EMJH base, and add 5-FU 3-4 days
after confirming growth under dark-field microscopy (DFM).
3. Monitor the culture for the presence of live, motile spirochetes by DFM every 3-
4 days: deposit 5 μl of culture on a glass slide and cover with a coverslip before
checking under the 40× objective of the microscope. Set aside a 100-μl aliquot of
the culture for quantification by qPCR.
Other pathogenic Leptospira species should grow under these standard conditions.
However, differences exist between species and serovars. If the above protocol proves
unsuccessful, consider trying modifications such as replacing plastic tubes with glass,
wrapping tubes in aluminum foil to provide a dark environment, and adding or omitting
supplements in the EMJH base (unpub. observ.).
15. Monitor the hamster for 15 days: record weight and signs of disease such as loss of
appetite, arched back, and prostration, and determine when the animal reaches the
endpoint criterion (>10% weight loss).
16. Euthanize the hamster with 5% isoflurane (see Current Protocols article: Donovan
& Brown, 2001), harvest the kidneys aseptically, cut the kidney into four pieces. and
place each piece in one tube containing 5 ml of EMJH base medium supplemented
with 500 μl Leptospira enrichment EMJH. Incubate the tubes at 30°C up to 4 weeks
Nair and
Gomes-Solecki to recover live virulent Leptospira.
Collection of Tissues After Euthanasia: Blood, Kidney, Lung, Liver, and Spleen
Additional Materials (also see Basic Protocol 2)
Heat-inactivated fetal bovine serum (FBS; Thermo Fisher Scientific, cat. no.
10-082-139)
Nair and Corning penicillin-streptomycin solution (Thermo Fisher Scientific, cat. no.
Gomes-Solecki MT30001CI)
Additional reagents and equipment for euthanasia of mice (see Current Protocols
article: Donovan & Brown, 2006a)
Humane endpoints and euthanasia
Mice are euthanized when weight loss reaches 20% unless they reach a depressed state
(>15% weight loss plus ruffled fur plus loss of mobility) before losing 20% of weight.
1d. Euthanize mice (see Current Protocols article: Donovan & Brown, 2006a) by CO2
asphyxiation or 5% isoflurane and exsanguination, and thoracotomy.
Collection of tissues
2d. Prepare complete RPMI 1640 medium by combining 50 ml of heat-inactivated
FBS and 5 ml of penicillin-streptomycin solution in 445 ml RPMI 1640 medium.
Aliquot 5 ml of into a sterile 15-ml tube and place it on ice.
3d. Spray the euthanized mouse with 70% ethanol, drain excess ethanol on a paper
towel, and place it on its back on a styrofoam holder (the Styrofoam holder from
a box of 50-ml conical Falcon-type tubes works well for this purpose). Pin down
its four limbs in a cross format.
4d. Using sterile surgical scissors or a scalpel, cut an incision line from the navel to the
top of the thorax, and with forceps, pull the skin and muscles aside (like opening a
book) to access the organs in the thorax (lung) and peritoneal cavity (liver, spleen,
and kidney).
Surgical equipment is sterilized by autoclaving in instant sealing sterilization pouches
prior to euthanasia.
5d. Hold an organ (e.g., kidney) with forceps and cut the blood vessels to release
it from the abdominal cavity. First, collect spleen into a tube with 5 ml RPMI
complete medium, and store on ice until processing. Place each organ in a sterile
tissue culture petri dish and cut in three or four fractions as needed (except spleen).
This work is done under aseptic conditions.
6d. Place one third of each organ (lung, liver, kidney) in a microcentrifuge tube con-
taining 0.5-1 ml (ideally 10 μl per mg tissue) of RNAlaterTM and freeze for pu-
rification of Leptospira DNA. Place one third of each organ in another microcen-
trifuge tube containing 0.5-1 ml of RNAlaterTM and freeze at −80°C to process
for mRNA purification for assessment of inflammatory mediators (e.g., cytokines,
fibrosis markers). Finally, place the remaining one third of the organ in a sterile 10-
ml tube containing 7 ml of EMJH medium supplemented with 500 μl of Leptospira
enrichment EMJH to culture Leptospira.
7d. Place the other kidney in neutral buffered formalin for histopathology staining and
for immunohistochemistry.
Nair and
Gomes-Solecki
NOTE: All animal experiments require approval by the local ethical and animal handling
offices.
Materials
Tissues collected from euthanized, Leptospira-infected mice (see Support Protocol,
steps 6d and 7d)
DNeasy Blood & Tissue Kit (Qiagen, cat. no. 69506)
NucleoSpin® Tissue Kit (Takarabio USA Inc., cat. no. 740952.250)
Leptospira-specific TAMRA probe and 16S rRNA primers (Eurofins Scientific)
RNeasy Mini Kit (Qiagen, cat. no. 74106)
RNase inhibitor (Applied BiosystemsTM, cat. no. 4374967)
RNase-free DNase (Qiagen, cat. no. 79254)
High-Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific, cat. no.
4368814)
TAMRA probe and primers: sequences in Table 1
Maxima Probe/ROX qPCR Master Mix (2×) (Thermo Fisher Scientific, cat. no.
K0233)
Neutral buffered formalin (Thermo Fisher Scientific, cat. no. 22-110-869)
Culture of >108 Leptospira/ml (see Basic Protocol 1)
10× phosphate-buffered saline (PBS; Thermo Fisher Scientific, cat. no.
BP399-500)
Nair and Modified Lowry Protein Assay Kit (Thermo Fisher Scientific, cat. no. PI23240)
Gomes-Solecki
H&E or PAS-D: Measure the glomeruli size in five fields per sample, and average
groups. Grade nephritis scores blindly on a scale of 0-5 in a longitudinal section
of the organ following previously published criteria (Chan, Madaio, & Shlomchik,
1997).
An example of PAS-D staining is shown in Figure 2.
Masson trichrome: Digitally analyze (40×, Photoshop) five randomly chosen fields
as a percentage of pixels of the total area.
An example is shown in Figure 3. Masson’s trichrome staining is very sensitive to process-
ing of the tissue before and after staining. It should only be performed between groups of
the same experiment by experienced personnel.
Figure 2 Histopathology of the kidney. (A) PAS-D staining showing shrinkage of glomeruli, infiltration of
immune cells, and loss of tubular structure in the infected group (20×); (B) diameter of glomeruli is measured
under 20× using a measurement function (e.g., CaptaVision Software). Legend: white arrow, tubules; black
arrow, infiltration of immune cells; black triangle, glomerulus (Sullivan et al., 2017).
sections per kidney are counted for each experiment. Acquire images at 200×, or
400× using a Zeiss microscope with ZEN software.
An example is shown in Figure 4.
13. Subject Leptospira to heat killing for 15 min at 95°C. Heat killing is confirmed by
checking under the DFM: no motile Leptospira are expected to be visible.
If motile Leptospira are visible under DFM, the process is repeated until no motile Lep-
tospira are observed (up to three times over 1 hr).
14. Determine the protein concentration using the Modified Lowry Protein Assay Kit
according to the manufacturer’s protocol.
Quantification of Leptospira-specific antibody (IgM and IgG) in serum
15. Coat a plate with antigen as follows. Dilute heat-killed Leptospira in 1× coating
buffer to a final concentration of 1 × 105 -108 /well or 100 μl/well of 4 mg/ml of
heat-killed Leptospira in a flat-bottom 96-well plate. Cover the plate and incubate
overnight at 4°C.
This protocol can be modified to test antibody levels against recombinant proteins as anti-
gens. Instead of heat-killed Leptospira, coat the plates with 0.1-1 μg/ml of the proteins
in coating buffer in step 15 and follow the protocol from step 16.
16. Next morning wash the plate using the ELISA plate washer and run a total of four
washes with 300 μl washing buffer.
17. To block the plate: Pour blocking buffer into a new reagent reservoir and add
250 μl of blocking buffer (1× PBST+ 1 %BSA; prepare fresh) in each well (us-
ing a multichannel pipet) and incubate for 1-2 hr at room temperature or 37°C.
If a plate washer is not available, use a multichannel pipettor to dispense 300 μl of
washing buffer per well, dump the washing buffer into a sink. and tap dry over a layer of Nair and
Gomes-Solecki
3-4 clean paper towels.
18. Wash the plate two times with 300 μl washing buffer. Tap dry over a layer of 3-4
clean paper towels.
19. Dilute the serum samples 1:100 in blocking buffer, add 100 μl of diluted serum
sample to each well of the Test plate, cover the plate, and incubate for 1.5 hr at room
temperature or 1 hr at 37°C.
20. Wash the Test plate four times; tap dry over a layer of 3-4 clean paper towels.
21. Clean the plate washer: Dip the wash head in washing buffer (1×PBST) in the blue
boat provided with the instrument and run a Wash plate twice to remove any primary
sera sticking to the wash head.
We use two 96-well plates to run the enzymatic reaction: one is labeled Test plate and
contains the serum samples subject to study; the second plate is a Wash plate used to
clean up the plate washer instrument after the washes.
22. Dilute the secondary antibody-HRP (IgM or IgG) in blocking buffer and pour this
into a new reagent reservoir. Add 100 μl to the wells of the Test plate using a multi-
channel pipettor. Cover the plate and incubate 1 hr at room temperature or 37°C for
30 min.
23. Wash the Test plate four times on the plate washer; tap dry over a layer of 3-4 clean
paper towels.
24. Clean the plate washer: Dip the wash head in 1×PBST in the blue boat provided
with the instrument and run a Wash plate with the “2 wash” program to remove any
HRP-conjugated antibody sticking onto the wash head.
25. Add 100 μl of TMB SureBlue substrate at room temperature into each well of the
Test plate using a multichannel pipettor.
26. Cover the plate and incubate for 15 min at room temperature or 37°C.
27. Add 100 μl of TMB Stop Solution into each well and read the absorbance at 450 nm
in the SpectraMax ELISA reader; save the file and export the raw data as an .xls
file for analysis.
28. Connect the washer to the MilliQ bottle and run a wash cycle with MilliQ water.
If the instrument is left in buffer, solutes form deposits and block the tubing.
Before starting the enzymatic reaction part of this protocol, aliquot the volume of TMB
SureBlue required for the ELISA in a 50-ml tube and keep it at room temperature covered
in foil (TMB substrate is light sensitive and is to be used at room temperature).
Nair and
30. Tease the spleen with frosted slides to produce single cell suspensions.
Gomes-Solecki
COMMENTARY
Background Information 2005, Werts et al., 2001). However, in mice,
Although Leptospira are not considered signaling occurs through TLR2 as well as
Gram-negative, they do produce lipopolysac- TLR4 (Nahori et al., 2005). Mice can tolerate
charide (LPS), a potent inflammatory cell wall levels of LPS endotoxin 250 times higher than
component. In human cells, Leptospira LPS humans (Copeland et al., 2005), which makes
signals through TLR2 rather than the conven- them excellent reservoir hosts for a number of
Nair and
tional TLR4 signaling system (Nahori et al., human pathogens, including Leptospira. For Gomes-Solecki
Nair and Figure 5 Susceptibility to lethal leptospirosis (LD50) in C3H-HeJ mice inoculated with PBS (Ctrl),
Gomes-Solecki
5 × 108 and 1 × 109 Leptospira interrogans ser. Copenhageni FioCruz. N = 4 mice per group.
Nair and
Gomes-Solecki
Keywords Abstract
Physical activity, T cell activation, viral
infection. Early studies in exercise immunology suggested acute bouts of exercise had an
immunosuppressive effect in human subjects. However, recent data, show
Correspondence acute bouts of combined aerobic and resistance training increase both lym-
Jacob A. Siedlik, Department of Exercise phocyte activation and proliferation. We quantified resistance exercise-induced
Science and Pre-Health Professions, changes in the activation state of CD4+ T lymphocytes via surface protein
Creighton University, 2500 California Plaza,
expression and using a medically relevant model of infection (HIV-1). Using a
Omaha, NE 68178.
Tel: +1 402 280 2474
randomized cross-over design, 10 untrained subjects completed a control and
Fax: +1 402 280 4732 exercise session. The control session consisted of 30-min seated rest while the
E-mail: jakesiedlik@creighton.edu exercise session entailed 3 sets 9 10 repetitions of back squat, leg press, and
leg extensions at 70% 1-RM with 2-min rest between each set. Venous blood
Funding information samples were obtained pre/post each session. CD4+ T lymphocytes were iso-
This work was supported by the Dr. George lated from whole blood by negative selection. Expression of activation markers
F. Haddix President’s Faculty Research Fund
(CD69 & CD25) in both nonstimulated and stimulated (costimulation
at Creighton University.
through CD3+CD28) cells were assessed by flow cytometry. Resistance exer-
Received: 24 June 2019; Revised: 22 August cised-induced effects on intracellular activation was further evaluated via
2019; Accepted: 27 August 2019 in vitro infection with HIV-1. Nonstimulated CD4+ T lymphocytes obtained
postexercise exhibited elevated CD25 expression following 24 h in culture.
doi: 10.14814/phy2.14234 Enhanced HIV-1 replication was observed in cells obtained postexercise. Our
results demonstrate that an acute bout of resistance exercise increases the acti-
Physiol Rep, 7 (18), 2019, e14234,
vation state of CD4+ T lymphocytes and results in a greater susceptibility to
https://doi.org/10.14814/phy2.14234
HIV-1 infection in vitro. These findings offer further evidence that exercise
induces activation of T lymphocytes and provides a foundation for the use of
medically relevant pathogens as indirect measures of intracellular activation.
50
Exercise Induced T Cell Activation and Infection A. K. Holbrook et al.
suppressed following an acute bout of exercise (Siedlik agent of acquired immune deficiency syndrome (AIDS),
et al. 2016). Notably, most of the research investigating is a member of the retrovirus family that preferentially
the effect of exercise on immunity have focused on infects activated CD4+ T lymphocytes. More than 36 mil-
PBMC proliferation in response to an acute bout of aero- lion persons are infected with HIV-1 worldwide
bic exercise (Walsh et al. 2011; b). In comparison, limited (UNAIDS). HIV-1 replication correlates strongly with the
research has been conducted examining the relationship activation state of T cells due to the metabolic require-
between acute bouts of resistance training and immunity ments of reverse transcription and integration into the
often with conflicting results (Dohi et al. 2001; Koch host genome (Gowda et al. 1989; Stevenson et al. 1990).
et al. 2001; Potteiger et al. 2001; Chan et al. 2003). Nie- Hence, any transient alteration in the metabolic state of a
man et al. (Nieman et al. 1995) observed no significant CD4+ T cell will change its susceptibility to HIV-1. Here,
differences in concanavalin A (ConA) stimulated lympho- we propose to use HIV-1 infection as a biological model
cyte proliferation in trained men following repeated sets to independently asses CD4+ T cell activation state. Nota-
of 10 repetition back squats at 65% of 1 repetition maxi- bly, quiescent CD4+ T cells, which are metabolically and
mum (1RM). Potteiger et al. (2001), however, observed transcriptionally silent (Yusuf and Fruman 2003; Tzacha-
reduced phytohemagglutinin (PHA) stimulated lympho- nis et al. 2004), were originally thought to be refractory
cyte proliferation in untrained females following an acute to HIV infection; however, evidence suggests that partially
bout of lower limb resistance training, but no change in activated cells can support infection, especially with sub-
proliferation for trained female participants. As seen sequent stimulation (Stevenson et al. 1990; Zack et al.
above, the training status of the participants, as well as 1990, 1992; Korin and Zack, 1998; Unutmaz et al. 1999;
the mitogens used for stimulation, may affect in vitro Manganaro et al. 2018).
proliferative responses. Moreover, the question of how Previous work from our laboratory demonstrated an
different immune subsets respond to acute resistance increase in CD25 expression and enhanced proliferative
exercise remains unanswered, preventing a full under- responses in CD4+ T cells following combined aerobic
standing of the clinical consequence of exercise prescrip- and resistance training exercise (i.e., circuit training)
tion. (Siedlik et al. 2017). In this study, we focused our efforts
Cellular activation in response to an antigen leads to to investigate whether resistance training alone altered
clonal expansion of antigen-specific T cells to facilitate CD4+ T cell activation state and response to co-stimula-
neutralization of an invading pathogen (Mueller et al. tion with CD28. In parallel we tested susceptibility to
1989). In laboratory and clinical settings, clonal expansion HIV-1 infection to independently validate any changes in
in response to either mitogenic stimulation or co-stimula- activation state resulting from exercise. To do this, we
tion through CD28 is commonly utilized as a measure of compared CD4+ T cells isolated from individuals prior to
lymphocyte functional ability (Siedlik et al. 2016; 2017). and after acute resistance training. Notably, the data were
Indeed, studies investigating the effects of exercise on the cross-compared to a nonexercise session as an additional
immune system have commonly used proliferation assays control. Changes in early markers of activation, specifi-
to quantify changes in lymphocyte function following an cally expression of CD25 and CD69, were analyzed from
acute bout of aerobic exercise. As addressed briefly above, two distinct perspectives. First, was there an exercise-in-
however, proliferative assays can produce ambiguous duced effect on CD4+ T cell activation absent stimuli in
results due to methodological variability (Siedlik et al. culture (Arm 1), and second, was there an exercise-in-
2016) and a failure to quantify specific elements of the duced effect on the ability of CD4+ T cells to respond to
activation process. Measurement of specific immune cell stimuli in culture (Arm 2). Together, this study represents
functions potentially can improve the understanding of a first attempt to quantify exercise-induced changes in
the impact of excise on immunity. CD4+ T cell function using a medically relevant viral
Exercise-induced alterations in immune function have model.
the potential to affect the interaction of pathogens with
the immune system. A change in cellular activation state
Methods
and/or subsequent alteration in proliferation of immune
cell subsets could alter the disease state in an individual.
Participants
For example, changes in CD4+ T lymphocyte activation
could enhance an immune response and provide greater Ten healthy, untrained, college-aged individuals
protection against an invading pathogen. Conversely, (Mean SD, n = 10 [5 male & 5 female], age =
heightened cell activation may create favorable conditions 21 2 year; weight = 71.5 10.1 kg; height = 171.8
for lymphotropic pathogens, such as human immunodefi- 7.1 cm) volunteered to participate. Untrained status was
ciency virus (HIV). Type 1 HIV (HIV-1), the etiologic defined as no more than 1 h of aerobic and/or resistance
2019 | Vol. 7 | Iss. 18 | e14234 ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of
The Physiological Society and the American Physiological Society.
51
A. K. Holbrook et al. Exercise Induced T Cell Activation and Infection
training per week. All participants provided written electronic devices during this time and were monitored at
informed consent and completed a Health & Exercise Sta- random intervals to ensure they remained alert.
tus Questionnaire prior to participation. At the time of
recruitment, subjects were instructed to maintain their
Physiological monitoring
normal dietary patterns prior to participating in either
session, but to refrain from exercise for 24 h prior to data Participants were fitted with a Zephyr BioHarness 3
collection. Participants reported that they had neither (Zephyr Technology, Annapolis, MD) to measure heart
recently taken nor were currently using non-steroidal rate (HR). Continuous HR measures were recorded at
anti-inflammatory drugs (NSAID), aspirin, or other anti- 1 sec intervals during the training session and down-
thrombotic over-the-counter or prescription medications. loaded using the Zephyr BioHarness Log Downloader
All participants reported being negative for HIV infection. (version 1.0.29.0). HRmax was estimated using the meth-
Participants reported no cold or flu symptoms in the ods of Tanaka et al. (2001).
2 weeks prior to data collection. The study conformed to
the standards set by the Declaration of Helsinki and the
Blood collections
procedures followed were in accordance with the protocol
approved by the Creighton University Institutional Blood samples were collected in sodium heparin vacutain-
Review Board (959210-1). ers prior to (Pre) and immediately following (Post) each
testing session using standard antecubital venipuncture
technique.
3-repetition maximum assessments
Each participant completed an initial strength assessment
Antibodies and reagents
that included 3-repetition maximum [RM] barbell high
bar parallel squat, 3-RM leg press, and 3-RM leg exten- Antibodies used for flow cytometry were purchased from
sion. The 3-RM back squat was tested on their first visit BioLegend (San Diego, CA) and include: anti-CD4-Alexa
and the leg press and leg extension were assessed at a visit Fluor 700 (RPA-T4), CD69-APC/Cy7 (FN50), and CD25-
at least 2 days after squat testing. All participants com- PE (M-A251).
pleted a 5 min self-paced warm-up on a cycle ergometer
prior to starting any warm-up sets. The 3-RM testing fol-
Cell purification and culture
lowed the protocol recommended by the National
Strength and Conditioning Association (Haff and Triplett Blood samples (80 mL) were obtained at each time point
2016). For all exercises, the first warm-up set required 5– for analyses of surface marker expression and viral repli-
10 repetitions. The second warm-up set and beyond cation. CD3+CD4+ T cell isolation from peripheral blood
required 2–5 repetitions until a 3-RM was attempted. was conducted through negative selection using a Human
Participants were allowed multiple attempts at the 3-RM CD4+ T cell enrichment kit as directed by the manufac-
to attain the highest load possible. The 3RM was recorded turer (Stemcell Technologies, Vancouver, BC, Canada).
and used for estimation of 1RM values using the follow- Purity was assessed following cell isolation by staining
ing equation: (3RM/ 0.9 = 1RM). All experiment visits with anti-CD4-Alexa Fluor 700 (RPA-T4) and all samples
took place at least 1 week after completion of repetition were >97% CD4+ by flow cytometry (Kohlmeier et al.
maximum testing. 2006; Newton and Benedict, 2014; Siedlik et al. 2017).
After purification, the cells were resuspended in warm
Immunocult-XF T cell expansion medium (Stemcell
Testing protocol
Technologies, Vancouver, BC, Canada).
Following the fitness assessments, each participant ran-
domly completed a control and exercise session, which
Surface marker expression – cell stimulation
occurred between 0700 and 0745 h. Both visits occurred
and culture
within a 7-day time frame. The exercise session included
a 5-min self-paced warm-up on a cycle ergometer fol- Cells were co-stimulated through CD3+CD28 using plate-
lowed by 3 sets of 10 repetitions of back squats at 70% of bound antibodies or no simulation as previously
estimated 1-RM, 3 sets of 10 repetitions of leg press at described (Chirathaworn et al. 2002; Kohlmeier et al.
70% 1-RM, and 3 sets of 10 repetitions of leg extension 2006; Siedlik et al. 2017). Each antibody was titrated to
at 70% 1-RM with 2-min rest between sets. The control the lowest concentration that gave maximum T cell acti-
session involved the subjects sitting quietly in a room for vation: anti-CD3 (OKT3) used at 1 lg/mL (BioLegend,
30 min. Subjects were not allowed to read or use San Diego, CA) and anti-CD28 (CD28.2) at 2 lg/mL
ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of 2019 | Vol. 7 | Iss. 18 | e14234
The Physiological Society and the American Physiological Society.
52
Exercise Induced T Cell Activation and Infection A. K. Holbrook et al.
(BioLegend, San Diego, CA). Antibodies were diluted in then pelleted, washed with dPBS, and resuspended in
sterile Dulbecco’s Phosphate Buffered Saline (dPBS) (Life 550 lL fresh culture media (RPMI-1640 Medium [GE
Technologies, Grand Island, NY) added to 96-well plates Healthcare, Piscataway, NJ] + 10% Fetal bovine serum
and incubated overnight at 4°C. Unbound antibodies [Corning, Corning, New York] + 2% Penicillin/Strepto-
were removed by washing 3x with dPBS prior to cell plat- mycin [Corning, Corning, New York] + 20 mmol/L L-
ing. CD4+ T cells were plated at 1.5 9 106 cells/mL in glutamine [Corning, Corning, New York] + 50 units/mL
200 lL of Immunocult-XF T-cell media (Stemcell Tech- recombinant Human IL-2 [R&D Systems, Minneapolis,
nologies, Vancouver, BC, Canada) directly after isolation. MN]) per well, in a new 24-well plate. Cells were stimu-
Cells were cultured at 37°C in a humidified incubator lated using plate-bound antibodies as outlined above, but
with 5% CO2. Cells were analyzed by flow cytometry in 24-well plates at 2 9 106 cells/mL in 500 lL of Immu-
using anti-CD4-Alexa Fluor 700, anti-CD69-APC/Cy7, nocult-XF T-cell media (Stemcell Technologies, Vancou-
and anti-CD25-PE antibodies immediately after CD4+ T ver, BC, Canada) culture. Stimulated CD4+ T cell HIV-1
cell isolation (0 h), 24 h, and 72 h in culture using a ZE5 infections occurred after 3 days of CD3/CD28 stimula-
Cell Analyzer (Propel Labs, Fort Collins, CO). Data analy- tion. Both unstimulated and CD3/CD28 stimulated cells
sis was performed with FlowJo software v10 (TreeStar, were cultured for 17 days. Supernatant samples were col-
Ashland, OR). Compensation was performed using single lected at 0, 3, 7, 10, 14, and 17 days post infection (dpi),
antibody positive and negative controls (OneComp clarified by centrifugation, and stored at 20°C. For the
eBeads Compensation Beads, ThermoFisher Scientific, reactivation studies, unstimulated cells were activated at
Waltham, MA) in each assay. Gates were set based on flu- 14 dpi with human CD3/CD28/CD2 T cell activator
orescence minus one controls. A representative gating beads for 3 days (Stemcell Technologies, Vancouver, BC,
strategy is shown in Figure 1. Canada) and additional RT sample collected at 17 dpi.
An overview of the cell culture experiments is shown in
Figure 2.
HIV-1 viral replication assays - cell
stimulation and culture
Reverse transcriptase assay
HIV-1 NLX virus stocks were produced through the
transfection of 293T cells with 5 lg of pNLX molecular Virus replication/production was quantified by a reverse
clone and quantified by p24 antigen ELISA as previously transcriptase (RT) assay as described previously (DeBoer
described (Siedlik et al. 2016). Both unstimulated and et al. 2018). Triplicate 10 lL of supernatant were assayed
CD3/CD28 stimulated cells were infected for each condi- per timepoint. Fresh culture media was used as a negative
tion. Unstimulated CD4+ T cells were infected and cul- control, and an NLX virus standard as a positive control
tured shortly after purification. 1 9 106 cells were seeded in each reaction plate. An RT assay mix of H20,
in three wells as per condition in a 24-well plate and 50 mmol/L Tris (pH 7.9), 75 mmol/L KCL, 2 nmol/L
incubated with HIV-1 at a multiplicity of infection DTT, 0.1875 mmol/L ATP, 5 mmol/L MgCl2, RT Primer
(MOI) of 0.1 for 4 h at 37°C with 5% CO2. Cells were (25 mg/L), 0.05% NP-40, 2 lmol/L dTTP, and 2 µCi
Figure 1. Representative gating procedures for analyzed samples. (A) Illustrates usage and placement of the live cell gate in the forward
scatter versus side scatter plot. Sample populations were all >97% CD3+CD4+ following isolation. All flow data were gated as in (A) before
further analysis. An unstained sample (not shown) was used as a guide for placement of the CD4+ gate in the fully stained sample (B).
Expression of surface markers of activation in non-stimulated and stimulated cell populations were quantified using median fluorescent intensity
(MFI) of (C) CD69 and (D) CD25. Overlays of the non-stimulated (light gray) and stimulated samples (dark gray) were used to correct for the
effect of costimulation through CD28 prior to analyzing exercise-induced alterations.
2019 | Vol. 7 | Iss. 18 | e14234 ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of
The Physiological Society and the American Physiological Society.
53
A. K. Holbrook et al. Exercise Induced T Cell Activation and Infection
Figure 2. Overview of experiment design. Each subject participated in both a control and exercise session with order randomized. Blood was
collected pre and post each visit (A) and CD3+CD4+ T cells isolated via negative selection. (B) Both unstimulated and CD3/CD28 stimulated cells
were cultured for 17 days. Stimulated CD4+ T cell HIV-1 infections occurred after 3 days of CD3/CD28 stimulation. Supernatant samples were
collected at 0, 3, 7, 10, 14, and 17 days post infection (dpi). Surface protein expression was quantified via flow cytometry at 0, 1, and 3 days.
[32P]-a-TTP was prepared and vortexed thoroughly (Sied- t-tests in R version 3.3.1 (Team 2014). Given the
lik et al. 2016). 30 lL of RT assay mix was added to each exploratory nature of this project, Bayesian paired sam-
well and the plate was incubated at 37°C for 3 h. RT mix ples t-tests were also performed using the BEST package
was added to Whatman paper in individual spots and in R (Kruschke 2013). Reported parameter estimates
allowed to dry. The paper was washed three times with from Bayesian models include the posterior mean differ-
29 saline-sodium citrate (SSC) for 5 min while rocking, ence, 95% highest density intervals (HDI), probability
washed once with 95% Ethanol, and allowed to dry. The the true mean difference is greater than 0, and, when
dried paper was then exposed to a phosphor screening relevant, threshold estimates to determine the probabil-
plate. After overnight exposure, the phosphor plates are ity the true difference of the means is greater than a
analyzed on GE Amersham Molecular Dynamics Typhoon 10% increase.
9410 Molecular Imager v5.0 (GE Healthcare, Piscataway,
NJ). The image was quantified using Molecular Dynamics
Results
ImageQuant v5.2 software (GE Healthcare, Piscataway,
NJ), setting the background to a negative control sample.
Resistance training session induced
substantial changes in heart rate
Statistical analysis
The exercise session elicited a substantial sympathetic
Power calculations were based on previously reported stimulus as demonstrated by the HR data. The mean pre-
data that investigated exercise-induced changes in T cell dicted HRmax for all subjects was 193 2 bpm. Average
activation and proliferation (Siedlik et al. 2017). The HR during the exercise trial was 137 14 bpm com-
power analysis indicated 10 subjects would exceed 80% pared with 82 10 bpm in the control subjects
power for detecting an effect size of dz = 0.99 [a large (P < 0.001). The average peak HR during the trials was
effect as outlined by Cohen (1992)] for relevant differ- 174 14 bpm and 109 11 bpm for exercise and con-
ences at an alpha < 0.05. For this investigation, quan- trol, respectively (P < 0.001). During the exercise session,
tification of the change in values from pre-to-post was subjects spent approximately half of the time
of more interest than the absolute values of the mea- (13.4 7.3 min) working at over 70% of their predicted
surements themselves; therefore, fold change scores were HRmax, whereas all subjects spent the entirety of the con-
calculated using log2 transformations: log2(Post/Pre). trol session under the 60% threshold of predicted HRmax.
Normality of data was verified using the D’Agostino- Summary of statistical results and comparative analyses
Pearson test. Data were analyzed using paired samples are presented in Table 1.
ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of 2019 | Vol. 7 | Iss. 18 | e14234
The Physiological Society and the American Physiological Society.
54
Exercise Induced T Cell Activation and Infection A. K. Holbrook et al.
Time Meandiff t df P 95% CI Post. Meandiff SDdiff 95% HDI P (Meandiff > 0)
No-Stim
CD25 0h 0.2 0.73 9 0.49 0.8, 0.41 0.2 0.89 0.84, 0.47 0.25
24 h 0.27 2.4 9 0.04 0.02, 0.52* 0.28 0.37 0.01, 0.53 0.98
72 h 0.04 0.12 9 0.91 0.7, 0.78 0.06 1.1 0.74, 0.83 0.57
CD69 0h 0.29 0.74 9 0.48 1.7, 0.6 0.23 1.3 1.2, 0.71 0.29
24 h 0.02 0.09 9 0.93 0.37, 0.41 0.02 0.58 0.41, 0.43 0.55
72 h 0.1 0.36 9 0.73 0.53, 0.74 0.11 0.93 0.56, 0.77 0.65
CD4 0h 0.04 1.41 9 0.19 0.1, 0.02 0.04 0.1 0.11, 0.03 0.13
24 h 0.02 0.32 9 0.76 0.11, 0.15 0.02 0.19 0.12, 0.16 0.62
72 h 0.05 1.09 9 0.3 0.05, 0.14 0.05 0.15 0.06, 0.15 0.82
Stim
CD25 24 h 0.58 0.75 9 0.47 2.35, 1.18 0.51 2.5 2.4, 1.3 0.27
72 h 0.95 2.08 9 0.07 0.08, 1.98 0.96 1.5 0.16, 2.1 0.96
CD69 24 h 0.36 0.8 9 0.45 1.37, 0.65 0.04 0.76 0.94, 0.57 0.43
72 h 0.22 0.76 9 0.47 0.43, 0.86 0.23 0.96 0.48, 0.9 0.76
CD4 24 h 0.06 0.12 9 0.91 1.24, 1.12 0.29 1.3 1.2, 0.75 0.26
72 h 0.15 0.2 9 0.84 1.49, 1.78 0.12 2.4 1.6, 2 0.56
HIV-1
Viral replication 0 day 0.35 1.34 9 0.21 0.25, 0.97 0.21 0.6 0.23, 0.78 0.87
3 day 0.28 2.68 9 0.03 0.04, 0.51* 0.27 0.35 0.02, 0.53 0.98
7 day 0.3 0.79 9 0.44 0.56, 1.15 0.31 1.3 0.65, 1.2 0.76
Heart rate
Average 56 13.14 9 <0.001 46, 65* 56 14 46, 66 >0.999
Peak 66 12.42 9 <0.001 54, 78* 65 17 52, 77 >0.999
2019 | Vol. 7 | Iss. 18 | e14234 ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of
The Physiological Society and the American Physiological Society.
55
A. K. Holbrook et al. Exercise Induced T Cell Activation and Infection
Figure 3. Expression of surface markers of activation increased on non-stimulated CD4+ T cell populations. Human CD4+ T cells were isolated
and cultured in non-stimulated wells for 1 and 3 days then stained for CD25 and CD69 and analyzed by flow cytometry. The median
fluorescence intensity (MFI) was determined for expression of CD25 at (A) 1 day (Control: 0.03 0.36, Exercise: 0.3 0.46) and at (B) 3 days
(Control: 0.07 0.7, Exercise: 0.11 0.59). CD69 expression at (C) 1 days (Control: 0.25 0.49, Exercise: 0.27 0.54) and at (D) 3 days
(Control: 0.06 0.52, Exercise: 0.16 0.55). Data are presented as fold change from baseline and visualized as spaghetti plots with each line
representing the change between paired samples; n = 10. * Indicates a statistically significant difference (P < 0.05) from the control session.
Table 2. Summary data for surface protein expression on nonstimulated CD4+ T lymphocytes.
0h 24 h 72 h
Control
CD4 Pre 9578.9 544.7 8966.1 1216.5 9151.3 1184.3
Post 9868.3 541.7 8975.9 1022.2 9454.5 1270.9
CD69 Pre 203.3 143.2 171.4 84.7 150.8 40.1
Post 207.0 100.2 198.4 71.4 159.2 52.6
CD25 Pre 342.8 156.9 291.8 102.1 195.3 55.8
Post 374.3 150.8 291.3 71.6 212.5 83.7
Exercise
CD4 Pre 9456.9 757.3 9196.3 2251.4 8998.7 1159.1
Post 9476.2 737.0 9269.2 1850.5 9653.7 1691.0
CD69 Pre 169.7 66.1 172.1 99.2 160.4 56.5
Post 180.5 99.5 194.2 69.7 173.3 52.6
CD25 Pre 340.9 117.0 325.4 233.3 230.2 90.6
Post 332.7 123.1 370.1 206.1 231.2 41.0
Values represent median fluorescent intensity (MFI) assessed via flow cytometry. Data are presented as Mean SD.
the credible values greater than zero. Thus, there is a to stimuli. Similar to the nonstimulated Arm, there were
strong probability that the estimated parameter is greater no significant differences in CD69 expression observed at
than zero (i.e., indicative of an exercise-induced increase either 24 h or 72 h (Fig. 4C–D). Data for the intensity of
in expression). The probability that the true mean differ- surface protein expression on stimulated cells is summa-
ence would be greater than a 10% increase in CD25 rized in Table 3. Summary data for the percent of acti-
expression was calculated at 93.3%. This suggests there is vated cells in both stimulated and nonstimulated cultures
a 93.3% chance that exercise alters CD4+ T cell response are shown in Table 4.
ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of 2019 | Vol. 7 | Iss. 18 | e14234
The Physiological Society and the American Physiological Society.
56
Exercise Induced T Cell Activation and Infection A. K. Holbrook et al.
Figure 4. Expression of surface markers of activation in stimulated CD4+ T cell populations. Human CD4+ T cells were stimulated for 1 and
3 days with anti-CD3 and anti-CD28 then stained for CD25 and CD69 and analyzed by flow cytometry. The median fluorescence intensity
(MFI) was determined for expression of CD25 at (A) 1 days (Control: 0.77 1.3, Exercise: 0.19 1.3) and at (B) 3 days (Control:
0.16 1.2, Exercise: 0.79 1.1). CD69 expression at (C) 1 days (Control: 0.41 0.91, Exercise: 0.06 0.71) and at (D) 3 days (Control:
0.03 0.61, Exercise: 0.25 0.6). Data are presented as fold change from baseline and visualized as spaghetti plots with each line
representing the change between paired samples; n = 10.
Table 3. Summary data for surface protein expression on CD4+ T Table 4. Summary data for percent of CD4+ T lymphocytes
cells costimulated through CD28. expressing both CD25 and CD69.
24 h 72 h 0h 24 h 72 h
Control No-stimulation
CD4 Pre 6092.3 2049.0 15675.9 3537.3 Control Pre 0.4 0.2 0.6 0.5 0.7 0.3
Post 6302.2 2090.4 17547.6 3360.9 Post 0.7 0.7 0.6 0.4 0.7 0.3
CD69 Pre 4072.4 2930.5 1031.2 269.9 Exercise Pre 0.4 0.2 0.5 0.2 0.8 0.5
Post 4436.9 2305.4 1084.5 339.1 Post 0.6 0.5 0.4 0.2 0.7 0.4
CD25 Pre 2850.3 2356.4 5496.5 4290.7 Stimulation through CD3+CD28
Post 3364.4 1889.7 4718.2 3469.1 Control Pre 57.9 19.4 69.4 18.3
Exercise Post 64.0 12.7 72.7 14.5
CD4 Pre 6417.7 2726.9 16004.6 5179.2 Exercise Pre 60.5 19.9 68.8 20.0
Post 6320.8 2505.9 16762.9 5133.9 Post 61.3 17.9 73.6 15.9
CD69 Pre 4422.8 2475.5 1748.0 1746.5
Post 4020.4 2026.4 1721.3 1270.1 Values represent percent of cells expressing both surface proteins.
CD25 Pre 3539.9 2154.8 5391.2 4791.3 Data are presented as Mean SD.
Post 3819.4 2739.6 7203.3 5967.8
Values represent median fluorescent intensity (MFI) assessed via 1 preferentially infects activated CD4+ cells due to the
flow cytometry. Data are presented as Mean SD. presence of increased metabolic activity. We hypothesized
that increased activation in CD4+ cells harvested postexer-
cise would lead to an increased susceptibility to HIV-1
HIV-1 infection is increased in stimulated
infection due to the increase in intracellular metabolism.
cells obtained postexercise
To test this, we assayed virus replication in CD4+ cells
As an alternative means to measure the level of activation isolated pre and post for both the exercise and control
in CD4+ T cells, infection with the CXCR4-tropic HIV-1 sessions for each participant. For each condition, we
NLX virus was chosen as a biological model system. HIV- infected both unstimulated (resting) and cells pre-
2019 | Vol. 7 | Iss. 18 | e14234 ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of
The Physiological Society and the American Physiological Society.
57
A. K. Holbrook et al. Exercise Induced T Cell Activation and Infection
Figure 5. Susceptibility to HIV-1 infection was increased in postexercise samples. (A) Example replication curve from one subject. Virus
replication is presented as RT activity measured from cell culture supernatants (arbitrary units). (B) Plots show fold changes in HIV-1 replication
levels in the activated CD4+ T cells in each subject between control (0.02 0.34) and exercise (0.29 0.38) sessions at 3 days post infection.
(C) The median fluorescence intensity (MFI) was determined for CD4 expression (Control: 0.46 1.6, Exercise: 0.61 1.5) following 3 days
stimulation with anti-CD3 and anti-CD28. Data are presented as fold change from baseline and visualized as spaghetti plots with each line
representing the change between paired samples; n = 10. *Indicates a statistically significant difference (P < 0.05) from the control session.
activated with CD3/CD28/CD2 beads. The levels of virus stimulated with various cytokines, including IL-7 or IL-13
replication were measured by quantification of RT activity (Unutmaz et al. 1999). These conditions are characterized
in cell supernatants collected at various days post infec- by increased metabolic activity and levels of RNA expres-
tion. Example data for one subject is shown in Figure 5A. sion equal to levels seen in the S phase of cell division
Overall, infection of unstimulated, quiescent CD4+ T cells (Korin and Zack 1998; Zack et al. 2013). Given that an
isolated from either the control or exercise session did acute bout of resistance exercise partially increased the
not produce any significant amount of virus replication, activation state of CD4+ T cells in the absence of stimuli,
nor were any differences between conditions observed. In we sought to investigate whether the cells isolated postex-
activated CD4+ T cells, the RT activity peaked on average ercise would support establishment of latent HIV-1 infec-
at 10 days post infection in cells from either the control tion. To assess this, the infected quiescent cells from both
or experiment sessions (Fig. 5A), then declined due to control and exercise sessions were reactivated at 14 dpi by
virus-induced cell death. The average fold change in RT treatment with CD3/CD28/CD2 activator beads and an
activity from pre-to-post exercise was calculated for each additional RT sample collected 3 days post stimulation
timepoint. At 3 days post infection, there was significantly (17 dpi) to measure for latent virus infection. No signifi-
greater levels of RT activity in the stimulated cells from cantly different levels of RT activity were observed
the postexercise condition compared to resting controls between either the control or exercise session, suggesting
(P = 0.03, Fig. 5B). Notably, there was no significant that exercise does not stimulate CD4+ T cells sufficiently
change in CD4 expression on cells isolated pre or post enough to support the establishment of HIV-1 latency in
the control and exercise session at the time of viral infec- CD4+ T cells (data not shown).
tion (P = 0.84, Fig. 5C). The Bayesian threshold estimate
indicates an 85.8% probability that the true parameter
Discussion
would be a greater than 10% increase in replication. Sum-
mary statistical data are shown in Table 1. The increased The present study examined the effect of resistance exer-
infection observed in cells obtained post exercise supports cise on CD4+ T cell activation and utilized a viral model
the hypothesis that there was increased metabolic activity to indirectly assess intracellular metabolic activity via viral
in CD4+ T cells following acute bouts of resistance exer- infection. The changes we observed in CD25 expression
cise. on non-stimulated cells suggest that resistance exercise
produces an effect on the activation state of CD4+ T cells
in previously untrained individuals. Furthermore, the
HIV-1 latent infection in quiescent CD4+ T-
Bayesian analyses suggest a similar exercise-induced
cells is not enhanced in cells isolated after
increase in CD25 expression in cells responding to stim-
an acute bout of resistance exercise
uli. Together, these findings indicate an increase in cell
HIV-1 preferentially infects activated CD4+ T cells, but it activation, which is also supported by the in vitro viral
has been shown that HIV-1 can infect quiescent cells par- model showing a significant increase in viral infection in
tially activated in the G1b phase of the cell cycle, or cells cells obtained postexercise.
ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of 2019 | Vol. 7 | Iss. 18 | e14234
The Physiological Society and the American Physiological Society.
58
Exercise Induced T Cell Activation and Infection A. K. Holbrook et al.
Previous work from our laboratory using an acute bout expression on nonstimulated cells obtained postexercise
of moderate intensity aerobic and resistance exercise independently validated an increase in cellular activation.
demonstrated increased activation and proliferation of Combined, these data support the hypothesis that exercise
CD3+ cells following stimulation by either phytohemag- induces an increase in the activation state of CD4+ T
glutinin (PHA) or co-stimulation through CD28 (Siedlik cells.
et al. 2017). Field studies using elite athletes in competi- A primary limitation of this study, and of research in
tive trials already demonstrate evidence of increased lym- exercise immunology in general, centers on the external
phocyte proliferation following bouts of aerobic exercise validity of the results themselves; namely, there is no
(Bassit et al. 2000; Tossige-Gomes et al. 2014). To date, direct translation from the in vitro assessments to an
no studies have examined the effect of an acute bout of in vivo model. This is particularly relevant for the current
resistance training on T lymphocyte activation states and project given the use of virus replication as an outcome
only a few have examined the effect of resistance training measure. A previous study by Roubenoff et al. (1999)
on lymphocyte proliferative capacity (Dohi et al. 2001; investigated alterations in viral RNA concentrations in
Koch et al. 2001; Potteiger et al. 2001). As an example, plasma from HIV-1 infected patients participating in a
Potteiger et al. (2001) observed a statistically significant single bout of aerobic exercise. In their study, untrained,
decrease in lymphocyte proliferation in untrained subjects HIV-1 positive individuals completed a 15 min, 60 cm
following an acute bout of lower-limb resistance training. vertical step test at a cadence of 1 step/sec. They found
Notably, however, this study along with the field studies no significant increase in HIV RNA postexercise but spec-
mentioned above (Bassit et al. 2000; Tossige-Gomes et al. ulated that the lack of change might have been due to the
2014), quantified proliferation from mixed lymphocyte relatively high baseline viral loads in most of the subjects
populations. Moreover, there was no assessment of either (Roubenoff et al. 1999). Similar to the replication thresh-
CD69 or CD25 expression which complicates any com- old we observed at 10 dpi in vitro, the authors (Rouben-
parison with our results. Indeed, there is substantial off et al. 1999) suspected there was a “ceiling effect,” or
ambiguity in the existing literature, which we propose is maximum threshold in replication, that limited exercise-
derived from variability in methods, including lympho- induced changes. Notably, patients with undetectable
cyte isolation protocols and the use of various mitogenic levels of HIV-1 RNA at the start of the study showed
agents [Reviewed in (Siedlik et al. 2016; Campbell and increases in plasma HIV-1 RNA levels postexercise. That
Turner, 2018)]. In this project, we attempted to clarify result is consistent with the findings of the current study,
some of the ambiguity of previous studies by focusing on despite the fact that we assayed virus replication in cells
a specific lymphocyte subset (CD4+ T cells) and utilizing from healthy, uninfected individuals.
a biological model of infection to assess exercise-induced Compounding concerns related to in vitro versus
effects. The use of viral infection and replication as an in vivo models, the genetic diversity, environmental expo-
outcome metric for immune function may seem counter- sures, and health histories of individuals generate highly
intuitive, but reverse transcription and the process by varied immune states (Tsang 2015). The innate variability
which HIV integrates into the host genome is dependent in immune responses in human subjects tends to lead to
upon the metabolic machinery of the cell, providing a less conclusive results compared to animal models, curb-
unique physiological method to assess exercise-induced ing enthusiasm for results obtained from human research
alterations in CD4+ T cell metabolic state. (Davis 2008). The present study identified small, transient
Cells isolated following exercise sessions supported effects representative, in scale, of exercise-induced changes
enhanced replication of HIV-1, but statistical significance in immune function. In an effort to embrace the observed
was observed only at the second timepoint (3 dpi). This variation (Gelman and Carlin 2017), the results of Baye-
is likely because once infection is established, HIV-1 has a sian analyses are provided to illustrate the probability of
logarithmic growth rate until the maximum threshold of parameter values given the observed data. Bayesian meth-
virus replication is achieved (Ribeiro et al. 2010). In our ods provide a probability distribution for an estimate;
experiments, the maximum threshold was typically meaning, we can assign a probability to our best estimate
achieved prior to the 10 dpi timepoint (overall of the mean difference and all the possible values the
mean = 9.575 2.0 dpi). Given a constant MOI and no parameter may take (Buchinsky and Chadha 2017). This
difference in cell surface expression of CD4 between the Bayesian estimation provides more informative results
control and exercise conditions, the exponential growth about the magnitude of parameters and associated vari-
rate of the viruses likely erased any differences in initial ance beyond that of traditional frequentist statistics
infection by 7 dpi. Nevertheless, the significant difference (Kruschke 2013). Moreover, we believe that improved
at 3 dpi demonstrated enhanced infection of the cells iso- assessment and interpretation of variation in studies of
lated postexercise. The significant increase in CD25 human immune responses is needed as medicine shifts
2019 | Vol. 7 | Iss. 18 | e14234 ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of
The Physiological Society and the American Physiological Society.
59
A. K. Holbrook et al. Exercise Induced T Cell Activation and Infection
toward personalized, precision care (Tsang 2015; Collins 2008.12.003. PubMed PMID: 19100694; PubMed Central
and Varmus 2015). PMCID: PMCPMC2905652.
Overall, these data support the hypothesis that an acute DeBoer, J., M. S. Wojtkiewicz, N. Haverland, Y. Li, E. Harwood, E.
bout of resistance exercise in untrained individuals gener- Leshen, et al. 2018. Proteomic profiling of HIV-infected T-cells
ates an increase in CD4+ T cell activation even in the by SWATH mass spectrometry. Virology 516:246–257. https://d
absence of stimulation. We also demonstrated the use of oi.org/10.1016/j.virol.2018.01.025. PubMed PMID: 29425767;
an in vitro viral model to quantify a physiological effect, PubMed Central PMCID: PMCPMC5836514.
arguing for the use of other pathogens to investigate exer- Dohi, K., A. M. Mastro, M. P. Miles, J. A. Bush, D. S. Grove,
cise-induced changes in immune cell subsets. Infectious S. K. Leach, et al. 2001. Lymphocyte proliferation in
models, when combined with other stimulation assays, response to acute heavy resistance exercise in women:
may help clarify the ambiguity in the current literature influence of muscle strength and total work. Eur. J. Appl.
regarding the effect of exercise on immunity. It is our Physiol. 85:367–373. PubMed PMID: 11560093.
hope, that future studies using these designs will help Gelman, A., and J. Carlin. 2017. Some natural solutions to the
advance translational research in exercise immunology p-value communication problem—and why they won’t
and ultimately lead to better informed prescription of work. J. Am. Stat. Assoc. 112:899–901. https://doi.org/10.
clinical exercise. 1080/01621459.2017.1311263.
Gowda, S. D., B. S. Stein, N. Mohagheghpour, C. J. Benike,
and E. G. Engleman. 1989. Evidence that T cell activation is
Conflict of Interest required for HIV-1 entry in CD4+ lymphocytes. J.
Immunol. 142:773–780. PubMed PMID: 2521508.
The authors report no conflict of interest.
Haff, G., and N. T. Triplett. 2016. Essentials of strength
References training and conditioning. 4th ed. Human Kinetics,
Champaign, IL.
Bassit, R. A., L. A. Sawada, R. F. Bacurau, F. Navarro, and L. Koch, A. J., J. A. Potteiger, M. A. Chan, S. H. Benedict, and B.
F. Costa Rosa. 2000. The effect of BCAA supplementation B. Frey. 2001. Minimal influence of carbohydrate ingestion
upon the immune response of triathletes. Med. Sci. Sports on the immune response following acute resistance exercise.
Exerc. 32:1214–1219. PubMed PMID: 10912884. Int. J. Sport Nutr. Exerc. Metab. 11:149–161. PubMed
Buchinsky, F. J., and N. K. Chadha. 2017. To P or Not to P: PMID: 11402249.
backing bayesian statistics. Otolaryngol. Head Neck Surg. Kohlmeier, J. E., M. A. Chan, and S. H. Benedict. 2006.
157:915–918. https://doi.org/10.1177/0194599817739260. Costimulation of naive human CD4 T cells through
PubMed PMID: 29192853. intercellular adhesion molecule-1 promotes differentiation to a
Campbell, J. P., and J. E. Turner. 2018. Debunking the myth memory phenotype that is not strictly the result of multiple
of exercise-induced immune suppression: redefining the rounds of cell division. Immunology 118:549–558. https://doi.
impact of exercise on immunological health across the org/10.1111/j.1365-2567.2006.02396.x. PubMed PMID:
lifespan. Front. Immunol. 9:648. https://doi.org/10.3389/f 16895560; PubMed Central PMCID: PMCPMC1782322.
immu.2018.00648. PubMed PMID: 29713319; PubMed Korin, Y. D., and J. A. Zack. 1998. Progression to the G1b
Central PMCID: PMCPMC5911985. phase of the cell cycle is required for completion of human
Chan, M. A., A. J. Koch, S. H. Benedict, and J. A. Potteiger. immunodeficiency virus type 1 reverse transcription in T
2003. Influence of carbohydrate ingestion on cytokine cells. J. Virol. 72:3161–3168. PubMed PMID: 9525642;
responses following acute resistance exercise. Int. J. Sport PubMed Central PMCID: PMCPMC109773.
Nutr. Exerc. Metab. 13:454–65. PubMed PMID: 14967869. Kruschke, J. K. 2013. Bayesian estimation supersedes the t test.
Chirathaworn, C., J. E. Kohlmeier, S. A. Tibbetts, L. M. J. Exp. Psychol. Gen. 142:573–603. https://doi.org/10.1037/
Rumsey, M. A. Chan, and S. H. Benedict. 2002. Stimulation a0029146. PubMed PMID: 22774788.
through intercellular adhesion molecule-1 provides a second Malek, T. R. 2008. The biology of interleukin-2. Annu. Rev.
signal for T cell activation. J. Immunol. 168:5530–5537. Immunol. 26:453–479. https://doi.org/10.1146/annurev.
PubMed PMID: 12023348. immunol.26.021607.090357. PubMed PMID: 18062768.
Cohen, J. 1992. A power primer. Psychol. Bull. 112:155–159. Manganaro, L., P. Hong, M. M. Hernandez, D. Argyle, L. C. F.
PubMed PMID: 19565683. Mulder, U. Potla, et al. 2018. IL-15 regulates susceptibility
Collins, F. S., and H. Varmus. 2015. A new initiative on of CD4(+) T cells to HIV infection. Proc. Natl. Acad. Sci.
precision medicine. N. Engl. J. Med. 372:793–795. https://d USA 115:E9659–E9667. https://doi.org/10.1073/pnas.
oi.org/10.1056/NEJMp1500523. PubMed PMID: 25635347; 1806695115. PubMed PMID: 30257946; PubMed Central
PubMed Central PMCID: PMCPMC5101938. PMCID: PMCPMC6187195.
Davis, M. M. 2008. A prescription for human immunology. Medzhitov, R. 2007. Recognition of microorganisms and
Immunity 29:835–838. https://doi.org/10.1016/j.immuni. activation of the immune response. Nature 449:819–826.
ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of 2019 | Vol. 7 | Iss. 18 | e14234
The Physiological Society and the American Physiological Society.
60
Exercise Induced T Cell Activation and Infection A. K. Holbrook et al.
2019 | Vol. 7 | Iss. 18 | e14234 ª 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of
The Physiological Society and the American Physiological Society.
61
FULL PAPER
www.advancedscience.com
1. Introduction
Nanoparticles (NPs) can serve as a promising vaccine delivery platform for
improving pharmacological property and codelivery of antigens and Therapeutic cancer vaccination aims to
adjuvants. However, NP-based vaccines are generally associated with activate and augment antitumor T cell
immunity by providing antigenic and
complex synthesis and postmodification procedures, which pose technical
costimulatory signals to professional
and manufacturing challenges for tailor-made vaccine production. Here, antigen-presenting cells (APCs).[1] In par-
modularly programmed, polyethyleneimine (PEI)-based NP vaccines are ticular, neoantigens, produced by genetic
reported for simple production of personalized cancer vaccines. Briefly, PEI is alterations occurring in a tumor- and
conjugated with neoantigens by facile coupling chemistry, followed by patient-specific manner, can be highly
immunogenic as neoantigens are entirely
electrostatic assembly with CpG adjuvants, leading to the self-assembly of
absent in normal cells, thus bypassing
nontoxic, sub-50 nm PEI NPs. Importantly, PEI NPs promote activation and central T cell tolerance.[2] Thus, amplifying
antigen cross-presentation of antigen-presenting cells and cross-priming of neoantigen-specific T cells using cancer
neoantigen-specific CD8+ T cells. Surprisingly, after only a single intratumoral vaccines offers a promising strategy for
injection, PEI NPs with optimal PEGylation elicit as high as ≈30% improving immunogenicity and selectivity
neoantigen-specific CD8+ T cell response in the systemic circulation and of cancer vaccines.[3] Indeed, neoantigen
vaccines based on peptides have recently
sustain elevated CD8+ T cell response over 3 weeks. PEI-based nanovaccines
generated promising clinical outcomes in
exert potent antitumor efficacy against pre-established local tumors as well as small cohorts of patients.[4] Although these
highly aggressive metastatic tumors. PEI engineering for modular initial clinical trials provide strong rationale
incorporation of neoantigens and adjuvants offers a promising strategy for for further development of neoantigen can-
rapid and facile production of personalized cancer vaccines. cer vaccines, these initial studies employing
free soluble vaccines exhibited limited ef-
ficiency at generating neoantigen-specific
T cells, potentially due to rapid clearance
of free antigens upon in vivo administration and poor codelivery
of antigens and adjuvants to APCs.[5]
Dr. J. Nam, Dr. S. Son Nanoparticle (NP)-based delivery systems have several advan-
Department of Pharmaceutical Sciences
Biointerfaces Institute tages for cancer vaccination, including improved pharmacologi-
University of Michigan cal properties, targeted delivery, and controlled and localized re-
Ann Arbor, MI 48109, USA lease of immunomodulatory agents for efficient modulation of
K. S. Park specific immune cells.[6] Various functional NPs based on lipo-
Department of Biomedical Engineering somes, polymers, lipoprotein nanodiscs, and inorganic NPs have
Biointerfaces Institute
been employed to improve innate immune stimulation and in-
University of Michigan
Ann Arbor, MI 48109, USA duction of antitumor T cell responses,[7] including personalized
Prof. J. J. Moon neoantigen cancer vaccines.[8] However, many NP-based vaccines
Department of Pharmaceutical Sciences generally involve complex synthesis steps and postmodification
Department of Biomedical Engineering of NPs, thus presenting technical and manufacturing challenges.
Biointerfaces Institute On the other hand, it is desirable to streamline the manufactur-
University of Michigan
Ann Arbor, MI 48109, USA ing process of neoantigen-based vaccines so that simple, scalable,
E-mail: moonjj@umich.edu affordable production with short turnaround time is feasible for
practical neoantigen-based cancer vaccination in the clinic.[9]
The ORCID identification number(s) for the author(s) of this article Here, we designed a programmable neoantigen cancer vac-
can be found under https://doi.org/10.1002/advs.202002577 cine that allows simple and facile modular assembly of defined
© 2021 The Authors. Advanced Science published by Wiley-VCH GmbH. antigens and adjuvants by exploiting the versatile functionality
This is an open access article under the terms of the Creative Commons of polyethyleneimine (PEI) (Figure 1). Furthermore, we sought
Attribution License, which permits use, distribution and reproduction in to perform systemic investigation on PEI-based vaccine system
any medium, provided the original work is properly cited.
for promoting cellular uptake of neoantigens, activation of APCs,
DOI: 10.1002/advs.202002577 and cross-priming of neoantigen-specific T cell responses. Our
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
62
www.advancedsciencenews.com www.advancedscience.com
Figure 1. Schematic illustration of PEI-based nanovaccine. PEI was sequentially modified with PEG and neoantigens via amide and disulfide bond,
respectively. Then, polycationic PEI conjugates were self-assembled with polyanionic CpG adjuvants through electrostatic interaction to form neoantigen
nanovaccine. Diverse types of antigens and adjuvants can be incorporated into the complex allowing flexible and modular design for personalized cancer
vaccines. The nanovaccine can increase the cellular uptake of neoantigens and adjuvants by APCs and promote activation and antigen cross-presentation
to effectively cross-prime antigen-specific T cells for robust antitumor immunity and antitumor efficacy.
vaccine consists of PEI-antigen conjugates and CpG adjuvants amount of the cross-linker and CSS-Adpgk was varied to ad-
that form compact nano-condensates through electrostatic in- just the density of Adpgk peptide, and the PEI–Adpgk conju-
teraction between polycationic PEI and polyanionic CpG. PEI- gates were analyzed by gel permeation chromatography (GPC)
antigen is composed of neoantigen peptides conjugated to PEI (Figure 2A). PEI–Adpgk conjugates displayed strong absorption
via a disulfide bond that can be readily cleaved in the highly peaks for Adpgk peptide at ≈15 min, which was absent in plain
reductive intracellular environment, thereby promoting cross- PEI (labeled as PEI–Adpgk(0)). When the conjugates were treated
presentation by APCs.[8b,d] Subsequently, PEI-antigen conjugates with dithiothreitol (DTT) reducing agent, the elution time of
are incubated with CpG to self-assemble into nano-sized parti- PEI–Adpgk conjugates was delayed by ≈0.9 min, and their peaks
cles for efficient codelivery of antigens and adjuvants to APCs coeluted with free CSS-Adpgk + DTT. These results demon-
– a prerequisite step for optimal T cell priming.[10] Our ap- strated successful conjugation of Adpgk peptide via reduction-
proach to exploit the intrinsic charge property can avoid complex sensitive bond, which would allow for the release of intact pep-
chemical and structural modifications and preserve immunolog- tides in a reducing environment. We prepared PEI–Adpgk conju-
ical activities of antigens and adjuvants to achieve maximum gates with Adpgk/PEI molar ratios of 2, 13, and 30, as determined
potency.[11] Importantly, we show polyethyleneglycol (PEG) mod- from the standard curve of CSS-Adpgk + DTT and concentration
ification as a simple yet powerful strategy to improve the PEI- of Adpgk released from DTT treatment of PEI–Adpgk (Figure S1,
based nanovaccine for cellular uptake, activation, and antigen Supporting Information). We could not obtain higher Adpgk con-
cross-presentation of APCs, while eliminating inherent cytotoxi- jugation as it caused precipitation due to poor solubility in aque-
city associated with PEI. The optimized nanovaccines elicited ro- ous medium. Since APCs are the first line of immune cells that
bust priming of antigen-specific CD8+ T cells and exerted strong vaccine formulations should engage for priming antitumor T cell
antitumor efficacy against pre-established local and metastatic tu- response, we examined PEI–Adpgk conjugates for potential cy-
mors, demonstrating their potential for personalized cancer im- totoxicity in bone marrow-derived dendritic cells (BMDCs) (Fig-
munotherapy. ure 2B). As PEI is known to be cytotoxic,[12] BMDCs incubated
with plain PEI and PEI–Adpgk conjugates exhibited similar lev-
2. Results els of cytotoxicity although we observed slightly reduced cytotox-
icity for PEI–Adpgk conjugates with Adpgk/PEI ratio ≥ 13.
2.1. PEGylation Reduces Cytotoxicity of PEI–Adpgk Conjugates We sought to reduce the cytotoxicity of PEI–Adpgk by employ-
and Produces Sub-50 nm CpG Complex ing PEGylation. PEG–PEI–Adpgk conjugates were synthesized
by unsaturated conjugation of methoxy poly(ethyleneglycol) pro-
We prepared PEI-antigen conjugates by employing an amine-to- pionic acid N-hydroxysuccinimide (methoxy-PEG-NHS) to a por-
sulfhydryl cross-linker that bridges PEI and cysteine-modified tion of the primary amine of PEI, followed by the cross-linker and
peptides through a reducible disulfide bond. As for the choice CSS-Adpgk conjugation as above. For the systemic investigation,
of antigen, we employed Adpgk peptide which is a neoantigen we varied the degree of PEGylation by adjusting the stoichiometry
identified in murine MC38 colon carcinoma.[3b] Specifically, the of PEG:PEI to 5:1, 10:1, 15:1, or 20:1. The efficiency of PEG conju-
primary amine of PEI was grafted with the cross-linker to create gation was nearly 100% for all cases as calculated from the unre-
pyridyldithiol functional groups to which CSS-Adpgk was con- acted free amine groups quantified using 2,4,6-trinitrobenzene
jugated to form PEI–Adpgk via disulfide linkage. The feeding sulfonic acid (data not shown).[13] PEG–PEI–Adpgk conjugates
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
63
www.advancedsciencenews.com www.advancedscience.com
Figure 2. Synthesis and characterization of PEI conjugates and CpG-containing nanovaccines. A–D) GPC spectra of A) PEI–Adpgk conjugates and C)
PEG–PEI–Adpgk conjugates measured before and after 10 × 10−3 m DTT treatment, and B, D) their dose-dependent cytotoxicity toward BMDCs assessed
after 24 h incubation. The number denotes number of conjugated Adpgk per PEI for PEI–Adpgk conjugates and number of grafted PEG per PEI for PEG-
PEI–Adpgk conjugates. E) Zeta potential and F) hydrodynamic size of nanovaccines formed by adding CpG to PEG–PEI–Adpgk conjugates with varying
weight ratio. G) TEM images of nanovaccines formulated at a weight ratio of 2 taken after 2% uranyl acetate staining for visualization of their morphology.
Scale bars = 200 nm. The data show mean ± s.d. (n = 5). **P < 0.01 and ****P < 0.0001, analyzed by two-way ANOVA with Bonferroni multiple
comparisons post-test.
were also confirmed using GPC spectra, which showed simi- drance of PEG. PEGylation dramatically improved biocompati-
lar ≈1 min peak shift after DTT treatment (Figure 2C), indi- bility of PEI–Adpgk conjugates with PEG/PEI ≥ 15 exhibiting
cating stable conjugation of Adpgk peptides via disulfide link- no cytotoxicity up to 100 µg mL−1 and rather promoting cel-
age. The ratio of Adpgk:PEI was calculated to be 46, 43, 37, and lular proliferation to some extent (Figure 2D). PEGylation was
33 for PEG(5)-, PEG(10)-, PEG(15)-, PEG(20)-PEI–Adpgk conju- mainly responsible for the reduced cytotoxicity although Adpgk
gates, respectively. The more PEG grafted, the smaller number conjugation also partially contributed to it (Figure S2, Supporting
of Adpgk was conjugated to PEI, probably due to the steric hin- Information).
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
64
www.advancedsciencenews.com www.advancedscience.com
We next investigated PEG–PEI–Adpgk conjugates formulated conjugates, compared with their respective free polymer form
with CpG. The cationic PEI in PEG–PEI–Adpgk can allow elec- (Figure 3B), and in particular, PEG(0)–PEI–Adpgk exhibited the
trostatic assembly and condensation of anionic CpG, confining greatest extent of decrease than others (Figure 3C). Nevertheless,
antigens and adjuvants into nanoparticles (NPs). NPs were for- compared with soluble Adpgk + CpG, the nanovaccine formu-
mulated by rapid mixing and 1 min incubation of CpG with PEG– lation markedly enhanced cellular uptake of CpG (Figure 3D),
PEI–Adpgk conjugates at weight ratios of PEG–PEI–Adpgk/CpG with 30–40-fold increase by PEG(5) NPs; 15–30-fold increase by
ranging 0.5–3. CpG had a zeta potential of −60 mV from its phos- PEG(10) and PEG(15) NPs; and 2–3-fold increase by PEG(0) and
phorothioate backbone units; as the feed amount of PEG–PEI– PEG(20) NPs (Figure 3E). Confocal microscopy images taken af-
Adpgk increased, the zeta potential of PEG–PEI–Adpgk/CpG ter 24 h incubation confirmed significant cellular uptake of both
NPs gradually increased toward more positive values (Figure 2E). PEI–Adpgk conjugates and CpG for PEG(5), PEG(10), and PEG
As CpG was added to PEG(0)–PEI–Adpgk and PEG(5)–PEI– (15) NPs (Figure 3F). In addition, we observed colocalization of
Adpgk, they underwent complete charge conversion to positive PEI–Adpgk conjugates and CpG in the endolysosomal compart-
at weight ratio > 1, while the conjugates with PEG ≥ 10 re- ments.
mained nearly neutral. These results suggest charge compensa- Having shown the robust uptake of nanovaccine, we next in-
tion of CpG by PEG–PEI–Adpgk conjugates by electrostatic as- vestigated activation and antigen presentation of BMDCs. We
sembly and passivation of their surface by the nonionic PEG examined nanovaccine-mediated activation of Toll-like receptor
layer. Complete CpG condensation appeared to occur at the PEG– (TLR)-9 using a HEK-Blue TLR-9 reporter cell line. When incu-
PEI–Adpgk/CpG weight ratio of 2, based on the zeta potential bated with HEK-Blue TLR-9 cells, PEG–PEI–Adpgk conjugates
measurement. As shown by the dynamic light scattering (DLS) induced only baseline signal, whereas CpG promoted strong ac-
measurements, the hydrodynamic (HD) size of NPs generally tivation of HEK-Blue TLR-9 cells, indicating TLR-9 activation by
did not change at PEG–PEI–Adpgk/CpG weight ratio ≥ 2, and CpG (Figure 4A, B). Whereas PEG(0) NPs showed only a baseline
at the weight ratio of 2, PEG(0)-, PEG(5)-, PEG(10)-, PEG(15)-, response, PEGylation of NPs significantly elevated TLR9 activa-
and PEG(20)-PEI–Adpgk conjugates formed NPs with HD size tion, with PEG ≥ 10 inducing stronger response than free CpG.
of 158 ± 19, 47 ± 18, 35 ± 16, 25 ± 7, and 20 ± 6 nm, respectively Next, we examined how NP formulation impacts antigen pre-
(Figure 2F). The negative correlation between PEG density and sentation by BMDCs. To study this, we employed a model anti-
HD size suggests that PEG passivation promotes formation of gen, SIINFEKL peptide, which is an immunodominant MHC-
small NPs by enhancing their colloidal stability, which is in line I epitope from ovalbumin. PEG-PEI-SIINFEKL conjugates were
with previous reports.[14] We confirmed NP formation with trans- synthesized and confirmed using GPC analysis as in Figure 2
mission electron microscopy (Figure 2G), which showed the size (Figure S3, Supporting Information). SIINFEKL nanovaccines
profiles in alignment with the DLS measurements. formulated with CpG at a weight ratio of 2 were incubated with
Overall, PEGylation significantly reduced cytotoxicity of PEI– BMDCs for 24 h, and BMDCs were analyzed for maturation
Adpgk conjugates and stabilized their CpG nanocomplex, and antigen presentation. Upregulation of CD40, CD80, CD86
thereby generating sub-50 nm NPs with a nearly neutral surface costimulatory marker on BMDCs (Figure 4C; Figure S4, Sup-
charge. The approach presented here allows the synthesis of well- porting Information) followed a similar pattern as PEG density-
defined PEI-antigen conjugates using facile conjugation chem- dependent increase in TLR-9 activation (Figure 4B), suggesting
istry. Subsequently, NPs can be readily produced in a few minutes CpG-mediated BMDC activation. In addition, PEG density also
by simple mixing and brief incubation with molecularly-defined affected antigen presentation on BMDCs, as measured by mono-
adjuvants. Thus, the PEI-based NP system offers a promising clonal antibody against SIINFEKL/H-2kb (pMHC) complex (Fig-
manufacturing strategy for on-demand production of personal- ure 4D). NPs with higher PEG density generally increased anti-
ized cancer vaccines with a quick turnaround. Based on the zeta gen presentation, with PEG(15) NPs inducing 8.4-fold higher
potential and HD size measurements, we chose NPs formed at pMHC level than soluble SIINFEKL + CpG (Figure 4D). Confo-
the PEG–PEI–Adpgk:CpG weight ratio of 2:1 for the subsequent cal microscopy also confirmed robust pMHC display on BMDCs
studies. treated with PEG(15) NPs, compared with soluble SIINFEKL +
CpG control (Figure 4E). In addition, pMHC was mainly local-
ized on the cell surface without much overlap with late endo-
2.2. PEGylation Enhances Cellular Uptake of CpG Nanocomplex somes/lysosomes stained with lysosomal associated membrane
and Promotes Activation and Antigen Presentation of BMDCs In protein 1 (LAMP-1) (Figure 4E). As PEI-antigen conjugates and
Vitro CpG were mainly localized in endo-lysosomes (Figure 3F), these
results suggest that the nanovaccine promotes intracellular de-
Next, we sought to investigate how PEGylation impacts on the in- livery of antigens and CpG and the subsequent steps of cross-
teractions between nanovaccines and BMDCs. PEG–PEI–Adpgk presentation, including the intracellular processing of peptide
conjugates and CpG were separately tagged with distinct fluo- antigen, MHC-loading of epitopes, and trafficking of pMHC to
rophores, formulated into NPs, incubated with BMDCs, and vi- the cell surface.[16] Without CpG, PEG-PEI-SIINFEKL conjugates
sualized to track cellular uptake of each components over time. in the form of free polymers exhibited decreased CD80, CD86,
The doses of PEG–PEI–Adpgk and CpG were fixed at 2 and CD40, and pMHC expression as the PEG density was increased
1 µg mL−1 , respectively. PEGylation decreased cellular uptake of (Figure S5, Supporting Information), possibly due to the de-
PEI–Adpgk conjugates (without CpG), likely due to the antifoul- creased cellular uptake. This is an opposite trend from the case of
ing and stealth feature of PEG (Figure 3A).[15] CpG-mediated nanovaccines, which suggests a unique beneficial role of PEGy-
NP complexation decreased cellular uptake of PEG–PEI–Adpgk lation for nanovaccine. Overall, PEGylation on PEI-antigen/CpG
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
65
www.advancedsciencenews.com www.advancedscience.com
Figure 3. Uptake of nanovaccines by BMDCs. A–C) Time lapse uptake of PEG–PEI–Adpgk conjugates in the form of A) free polymer or B) their nanovac-
cines formulated by adding CpG measured over 3 days, and C) corresponding fold change in the uptake of PEG–PEI–Adpgk conjugates after CpG
addition. D) Time lapse uptake of CpG and E) corresponding fold change in CpG uptake by nanovaccines, compared to soluble Adpgk + CpG. F) Con-
focal microscope images of BMDCs after 24 h incubation with soluble Adpgk + CpG or nanovaccine samples. Scale bar = 50 µm. The data show mean
± s.d. (n = 6). ***P < 0.001 and ****P < 0.0001, analyzed by two-way ANOVA with Bonferroni multiple comparisons post-test.
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
66
www.advancedsciencenews.com www.advancedscience.com
Figure 4. Induction of TLR9-mediated immune stimulation and antigen cross-presentation by nanovaccines. A,B) HEK-Blue TLR9 cells were incubated
with A) free polymer form of PEG–PEI–Adpgk conjugates or B) their nanovaccines with CpG, and induction of TLR9 signaling cascade was quantified
using 650 nm absorbance. Upregulation of C) CD40 and D) SIINFEKL/H-2Kb expression by BMDCs after 24 h incubation with SIINFEKL + CpG or
SIINFEKL nanovaccines. E) Confocal microscope images of BMDCs incubated with SIINFEKL + CpG or PEG(15) NP of SIINFEKL nanovaccine. Scale
bar = 50 µm. The data show mean ± s.d. (n = 6). *P < 0.05, ***P < 0.001, and ****P < 0.0001, analyzed by one-way ANOVA with Bonferroni
multiple comparisons post-test.
nanovaccines plays a vital role in cellular uptake, adjuvant activity, tightly organized in a confined volume, and thus provides a suit-
and antigen cross-presentation, and high PEG density are gener- able biological model for studying complex cellular interactions.
ally favored for activation of DCs. Adpgk nanovaccine was tested in a murine tumor model of MC38
colon carcinoma.[3b] We established MC38 colon carcinoma sub-
cutaneously on the right flank of C57BL/6 mice, and vaccines
2.3. PEGylation Reduces Tumor Retention of Nanovaccine but composed of Adpgk peptides and Alexa Fluor 647 (AF647)-tagged
Elicits Strong Immune Activation in Local Tumor-Draining Lymph CpG were administered directly into tumors. The fluorescence
Nodes In Vivo intensity of AF647-CpG measured ex vivo after 24 h revealed
that PEG(0) and PEG(5) NPs enhanced tissue retention of CpG
Next, we investigated PEGylation-dependent cellular uptake of (Figure 5A), probably due to positive surface charges (Figure 2E).
nanovaccines in vivo. Tumor tissue consists of a variety of cells Flow cytometry-based analysis of tumor tissues indicated that
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
67
www.advancedsciencenews.com www.advancedscience.com
Figure 5. Tumor retention of the nanovaccine and immune activation in tumor-draining LNs. A) Tumor retention of vaccines composed of various forms
of Adpgk peptides and AF647-CpG was visualized using ex vivo IVIS imaging after 24 h of intratumoral injection. Quantitative analysis of B) CpG+ cells
and C) corresponding MFI of CpG in CpG+ cells in tumors. D–K) Tumor-draining inguinal LNs were analyzed for the number and activation of D-G)
DCs and H–K) macrophages. The data show mean ± s.d. (n = 5). *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001, analyzed by one-way
ANOVA with Bonferroni multiple comparisons post-test.
PEG(5) NPs were broadly distributed in a larger population of macrophages in TDLNs (Figure 5I,J), resulting in a decreased ra-
cells, whereas cellular uptake of PEG(0) NPs was mainly re- tio of M2/M1-like macrophages (Figure 5K).[18] We observed sim-
stricted to a small subset of cells that internalized NPs to a greater ilar activation of DCs and macrophages in tumor-draining axil-
extent (Figure 5B,C). PEG(0) NPs appeared to be rapidly captured lary LNs, but not in contralateral non-tumor-draining inguinal or
by cells at the injection site with limited distribution in the tu- axillary LNs (Figures S8 and S9, Supporting Information). These
mor tissues, whereas PEG(5) NPs exhibited increased distribu- results show that a high degree of PEGylation potentiates the per-
tion within the tumor tissues, probably due to the PEG passi- formance of nanovaccines upon cellular entry despite the reduc-
vation layer. CpG was mainly internalized by tumor cells and tion in direct cellular association, which is in agreement with in
macrophages regardless of the formulations (Figure S6, Support- vitro results.
ing Information).
Tumor-draining lymph nodes (TDLNs) are critical sites where
T-cells are primed for immune activation against tumors.[17] 2.4. Antitumor Immune Response of Nanovaccine against
Therefore, we analyzed DCs and macrophages in inguinal Pre-Established Local Tumor
TDLNs after intratumoral administration of NPs. First, we con-
firmed that AF647 conjugation did not compromise the adjuvan- Having shown the robust activation of DCs and macrophages
ticity of CpG using BMDCs in vitro (Figure S7, Supporting In- in TDLNs, we next examined the potency of nanovaccines for
formation). PEG(15) and PEG(20) NPs enriched DCs in TDLNs priming antitumor T cell response. C57BL/6 mice were subcu-
and elevated their expression of CD40, CD80, and CD86 cos- taneously inoculated with MC38 cells, administered with Adpgk
timulatory markers (Figure 5D–G). In contrast, PEG(0), PEG(5), nanovaccines or soluble Adpgk + CpG on day 9 via intratu-
and PEG(10) NPs induced weaker activation of DCs in TDLNs moral injection, and analyzed for antitumor immune responses
(Figure 5D–G), suggesting that PEG density on NPs plays a cru- (Figure 6A). PEGylated nanovaccines induced robust priming of
cial role in DC activation in TDLNs. Similar PEG-dependency antigen-specific CD8+ T cells in the systemic circulation, as mea-
was observed for the number of macrophages in TDLNs (Fig- sured by Adpgk tetramer staining of peripheral blood mononu-
ure 5H), with PEG(15) and PEG(20) NPs significantly increas- clear cells (PBMCs) after 7 days of vaccination (Figure 6B). Sur-
ing macrophages compared with PEG(0) NPs. Compared with prisingly, with only a single injection, PEG(15) and PEG(20) NPs
PBS and PEG(0) NP, PEGylated NPs as well as the soluble vac- elicited potent neoantigen-specific CD8+ T cell responses against
cine group upregulated CD86 and downregulated CD206 on Adpgk, with 5–6-fold higher tetramer+ CD8+ T cells than soluble
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
68
www.advancedsciencenews.com www.advancedscience.com
Figure 6. Antitumor immune response of nanovaccine against pre-established local tumors. A) Schematic of treatment regimen. B,C) Adpgk-specific
CD8+ T cells in blood were analyzed after B) intratumoral injection of various vaccine formulations or C) administration of Adpgk + CpG versus PEG(15)
NP via different routes of vaccination. MC38 tumor-bearing mice were treated by intratumoral administration of Adpgk + CpG versus PEG(15) NP on day
9, and D) tumor growth and E) animal survival were monitored. F) Adpgk-specific CD8+ T cells in blood observed over 3 weeks after single immunization.
Tumor microenvironment analysis for the frequency of G) CD8+ T cells and H) Adpgk-specific CD8+ T cells, mean fluorescence intensity (MFI) of I)
perforin and J) granzyme in total CD8+ T cells. The data show mean ± s.d. (n = 5). *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001,
analyzed by one-way (B,C,G,H,I,J) or two-way (D,F) ANOVA with Bonferroni multiple comparisons post-test, or by E) log-rank (Mantel–Cox) test.
Adpgk + CpG (19 ± 4.5 and 17 ± 9.4% vs 3.3 ± 2.5%, P < 0.0001, and considerations, we chose intratumoral administration with
Figure 6B). In contrast, PEG(5) and PEG(10) NPs induced com- PEG(15) NPs for the subsequent antitumor efficacy studies.
parable CD8+ T cell responses with soluble Adpgk + CpG, while C57BL/6 mice were inoculated with MC38 tumor cells on day
PEG(0) NP had barely detectable response (Figure 6B). 0, and a single intratumoral injection of PEG(15) NP was given
Based on strong CD8+ T cell response induced by PEG(15) on day 9. PEG(15) NP effectively suppressed tumor growth (Fig-
NPs, we focused on PEG(15) NPs and examined how the route ure 6D) and eliminated established tumors in 60% mice, lead-
of immunization impacts T cell responses. After 9 days of MC38 ing to significant survival benefit compared with other groups
tumor inoculation, tumor-bearing mice were administered with (P < 0.01, Figure 6E). In contrast, soluble Adpgk + CpG had
PEG(15) NPs via intratumoral, subcutaneous (s.c.), or intra- only a modest effect with all treated mice succumbing to tu-
venous (i.v.) routes, which resulted in elicitation of 14 ± 3.1, 6.9 ± mors before day 50. Importantly, a single intratumoral admin-
5.6, and 5.8 ± 7.4% Adpgk-specific CD8+ T cell response, respec- istration of PEG(15) NP led to potent, systemic antitumor CD8+
tively, on day 16 (Figure 6C). In contrast, soluble Adpgk + CpG T cell response, achieving up to ≈30% Adpgk-tetramer+ CD8+ T
induced only 2–4% CD8+ T cell responses regardless of the in- cell response and sustaining elevated CD8+ T cell response over
jection routes. Intratumoral vaccination can be a promising can- 3 weeks (P < 0.05, Figure 6F), whereas the soluble vaccine group
cer immunotherapy as it can elicit strong antitumor immunity induced weak and transient CD8+ T cell response.
without overt systemic exposure of the vaccines. In fact, there Systemically activated CD8+ T cells need to migrate and infil-
are currently a number of clinical trials evaluating direct intratu- trate into the tumor bed in order to recognize and eradicate can-
moral injection of immunotherapies.[19] Based on these results cer cells.[20] To investigate tumor homing and cytotoxic activity of
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
69
www.advancedsciencenews.com www.advancedscience.com
CD8+ T cells, we analyzed the tumor microenvironment after 7 ysis of splenocytes using interferon (IFN)-𝛾𝛾 enzyme-linked im-
days of PEG(15) NP treatment. PEG(15) NPs promoted tumor in- munospot (ELISPOT) assay showed that PEI-M27 NP and PEI-
filtration of CD8+ T cells (Figure 6G), with significantly increased M27/M30 NP significantly enhanced antigen-specific T cell re-
frequency of Adpgk-specific CD8+ T cells (28 ± 9.0%), represent- sponses against MHC-I-restricted M27 and MHC-II-restricted
ing 14- and 3.2-fold increases over PBS and soluble Adpgk + M30 neoepitopes (Figure 7C,E,F). Soluble formulations induced
CpG, respectively (Figure 6H). Although soluble Adpgk + CpG markedly lower antigen-specific T cell responses. These results
slightly elevated the frequency of CD8+ T cells in the tumor mi- suggested that CD8+ T cell response against M27 neoepitope was
croenvironment, only a small subset of intratumoral CD8+ T cells largely sufficient for suppressing local B16F10 tumors, whereas
was specific to Adpgk peptide, with no statistical difference from systemic inhibition of metastasis required both antitumor CD8+
that of PBS-treated mice (Figure 6G,H). Intratumoral CD8+ T and CD4+ T cells. Interestingly, soluble-M27/M30 treatment
cells primed with PEG(15) NPs had high expression levels of per- induced splenomegaly indicative of systemic inflammation,[5a]
forin and granzyme (Figure 6I,J), indicating their cytotoxic po- whereas PEI-M27/M30 NP and all other treatments showed no
tential. On the other hand, we observed minimal activation of change, compared to the PBS control (Figure 6G).
CD4+ T cells and NK cells (Figure S10, Supporting Information). Overall, these results demonstrate that nanovaccines tailored
Taken together, these results demonstrate that the nanovaccines for eliciting a broad spectrum of T cell responses against multi-
can induce a robust and durable antitumor response by promot- ple neoepitopes could effectively treat highly aggressive local and
ing clonal expansion and tumor infiltration of antitumor CD8+ T metastatic tumors, while mitigating acute systemic side effects
cells. associated with soluble vaccine treatment.
3. Discussion
2.5. Nanovaccine against Highly Aggressive and Metastatic
Tumor Model PEI has been widely exploited as a gene transfection agent as
it can form positively charged nanoscale complex with DNA
Finally, we sought to evaluate the therapeutic potential of the or RNA oligonucleotides to promote their cellular uptake and
nanovaccines using B16F10 melanoma, which is a highly ag- expression.[21] In addition, PEI can stimulate immune activation
gressive model with poor immunogenicity. To mimic late stage, by triggering release of “danger signals” or “damage-associated
advanced cancer, we established B16F10 melanoma in both s.c. molecular patterns” as the result of cellular stress and damage
flank and lung tissues; C57BL/6 mice were inoculated with 3 × caused by its cytotoxic actions.[12,22] The ability of PEI to induce
105 B16F10 cells at s.c. flank as well as 4 × 105 B16F10 cells inherent immune stimulation and efficient cellular transfection
via i.v. administration, leading to the establishment of s.c. flank encouraged its development for vaccine applications associated
tumor and lung metastatic nodules (Figure 7A). Antitumor effi- with the delivery of protein- or DNA-based antigens. However,
cacy of nanovaccines was examined against both local tumors and previous studies mostly utilized PEI-based vaccines for treating
disseminated metastases after the vaccine formulations were ad- infectious disease with antibody response,[23] while a handful of
ministered directly into the s.c. flank tumors only. As this model cancer applications indicated sub-optimal intrinsic adjuvanticity
is highly aggressive, we vaccinated animals three times on days 7, of PEI for eliciting antitumor T cell response.[24] This has been
10, and 13. In addition, we utilized recently reported neoantigens attributed in part to type 2 T helper cell (Th2)-biased immune
identified in B16F10 cells, namely MHC I-restricted M27 and activation by PEI, which triggers inflammasome activation and
MHC II-restricted M30 neoepitopes, in order to study the effect humoral immunity rather than cellular immunity—a crucial cri-
of combining MHC-I epitope with MHC-II epitope.[3a] PEG(15)- terion for successful cancer vaccination.[23a,b] In addition, trans-
PEI-M27 and PEG(15)-PEI-M30 were synthesized following the fection of host bystander cells and subsequent cytotoxicity by PEI
established protocol and confirmed using high-performance liq- have been reported to activate T cells against self-antigens, poten-
uid chromatography (Figure S11, Supporting Information). CpG tially causing immune-related adverse events.[22b,25] In this work,
was added to PEG(15)-PEI-M27 or the mixture of PEG(15)-PEI- we sought to take advantage of the versatile functionality of PEI
M27 and PEG(15)-PEI-M30 conjugates, leading to the formation for delivery of antigens and adjuvants, while eliminating inher-
of PEI-M27 NP and PEI-M27/M30 NP, respectively. Both PEI ent cytotoxicity of PEI that has hampered cancer vaccine appli-
NPs exhibited similar HD size and zeta potential with nearly neu- cations. Here, we have shown that PEGylation of PEI formula-
tral surface charge; HD size was measured as 29 ± 8.6 nm and tions significantly decreased cytotoxicity of PEI, while also im-
27 ± 8.2 nm, and zeta potential 6.0 ± 4.7 mV and −1.7 ± 3.9 mV proving the performance of PEI to deliver exogenous Th1-favored
for PEI-M27 NP and PEI-M27/M30 NP, respectively. CpG adjuvant along with neoantigens in a spatiotemporally con-
Both PEI-M27 NP and PEI-M27/M30 NP treatment groups po- certed manner. The optimized PEI-based nanovaccines gener-
tently inhibited the growth of primary s.c. flank tumors compared ated robust antigen-specific T cells with a magnitude significantly
with PBS (P < 0.01, Figure 7B) although their antitumor effects greater than previously reported PEI-based vaccines,[23–24] sug-
were not statistically significant from the soluble vaccine group, gesting new engineering opportunities of PEI-based vaccines for
probably due to the aggressive nature of this B16F10 model. Im- personalized cancer immunotherapy.
portantly, PEI-M27/M30 NP treatment exerted potent systemic PEG conjugation completely abolished cytotoxicity of PEI at
antitumor efficacy against B16F10 metastasis, leading to a signif- stoichiometry of PEG/PEI ≥ 15, which is in line with previous
icantly decreased number of lung metastatic nodules by day 17 reports.[14a,26] In addition, PEG can serve as a uncharged spacer
(Figure 7C,D). In contrast, all other treatment groups had simi- unit that provides steric stabilization, decreases nonspecific cel-
lar number of lung metastatic nodules as the PBS control. Anal- lular uptake, and improves in vivo performance for PEI and its
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
70
www.advancedsciencenews.com www.advancedscience.com
Figure 7. Antitumor immune response of nanovaccine against highly aggressive, disseminated B16F10 melanoma. A) Schematic of treatment reg-
imen. B) Tumor growth curves of subcutaneous flank B16F10 tumors. C) Representative images of lungs and ELISPOT wells. ELISPOT assay was
performed after restimulation of splenocytes with M27 or M30. Quantitative analysis of D) lung tumor nodules and ELISPOT counts against E) M27
or F) M30 performed on day 17. G) Weight and images of spleens for assessement of splenomegaly. Scale bars = 1 cm. The data show mean ±
s.d. (n = 8). *P < 0.05, **P < 0.01, and ***P < 0.001, analyzed by D–G) one-way or B) two-way ANOVA with Bonferroni multiple comparisons
post-test.
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
71
www.advancedsciencenews.com www.advancedscience.com
Figure 8. Summary of the impact of PEGylation on PEG-PEI-Ag formulation, in vitro DC activation, and in vivo immune activation.
nano-complex.[27] PEGylation allowed for the formation of sub- We speculate that PEGylation serves multi-purposes; surface-
50 nm small NPs that significantly enhanced uptake of antigen displayed PEG reduces nonspecific cellular uptake while inner
and adjuvant by APCs. In particular, the uptake of CpG was PEG layer is thought to facilitate dissociation of the nanocom-
greatly improved by the nanovaccine formulation, which could plexes in the sub-cellular compartments. Efficient liberation of
be attributed to gaining positive charges from the PEI-antigen compactly packed nanocomplexes within target cellular compart-
conjugate; in return, this caused decreased cellular uptake of PEI- ments is a prerequisite for immune activation, serving as a cru-
antigen conjugate with the loss of positive charges. Nonetheless, cial factor that governs the efficacy of nanovaccines and antitu-
compared with non-PEGylated PEI-antigen/CpG nanocomplex, mor T cell responses.[28] Nonetheless, there exists an optimal
PEGylated PEI-antigen/CpG nanovaccines increased uptake of level of PEGylation for balancing cellular uptake and unpack-
both antigen and adjuvant, presumably due to PEG-mediated ing of nanocomplexes, as demonstrated by comparable in vitro
surface passivation and enhanced colloidal stability. More im- and in vivo immune activation and T cell responses induced by
portantly, the degree of PEGylation had a significant impact on PEG(15) NP and PEG(20) NP (Figures 4–6) despite significant
immunological activity of nanovaccines, with higher PEG gen- lower cellular uptake of PEG(20) NP (Figure 3). In contrast, cel-
erally potentiating the vaccine efficacy regardless of the extent lular uptake was directly associated with the activity of non-CpG-
of cellular uptake. In vitro, this was clearly demonstrated with complexed free PEI-antigen polymers, with PEGylation decreas-
PEG(20) NP. Compared with NP formed with lower PEG densi- ing cellular uptake and subsequent activation and antigen pre-
ties, PEG(20) NP induced the least cellular uptake of PEI-antigen sentation of BMDCs in vitro (Figure 3A; Figure S5, Supporting
and adjuvant (Figure 3); nevertheless, PEG(20) NP promoted ro- Information). Overall, these results suggest that immunologi-
bust TLR-9 signaling (Figure 4B), upregulation of costimulatory cal activity of nanovaccine is mainly limited by steric restriction
markers (Figure 4C), and antigen cross-presentation by DCs (Fig- of antigens and adjuvants, which could be improved by PEGy-
ure 4D). A similar observation was made in our in vivo studies. lation that facilitates unpacking and liberation of antigens and
Activation of DCs and macrophages in TDLNs (Figure 5D–K) was adjuvants from the nanocomplex. The impact of PEGylation on
increased with higher degree of PEGylation although PEGylation the various aspects of formulation and performance of PEI-based
reduced cellular association of NPs in the tumor tissues (Fig- vaccine is summarized in Figure 8.
ure 5A–C). PEG can not only passivate the surface of nanovac- The optimized nanovaccines allowed a greater amount of anti-
cines but also insert a charge-inert layer during the assembly gens and adjuvants to gain entry into cells than soluble vac-
of PEI-antigen and CpG that weakens electrostatic interaction. cines, suggesting that the NP formulation promotes endocytosis/
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
72
www.advancedsciencenews.com www.advancedscience.com
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
73
www.advancedsciencenews.com www.advancedscience.com
the ratio of the absorbance to the nonsample treated cells. For the cel- F4/80-APC (BioLegend, No. 123116), CD86-PE/Cy7 (BD Biosciences, No.
lular uptake study, BMDCs were incubated with PEI–Adpgk/AF488 con- 560582), CD206-APC/Cy7 (BioLegend, No. 321120) for CD11b+F4/80+
jugates or their NPs with CpG-AF647 at dose of 20 µg mL−1 PEI conju- macrophages. All flow cytometry was performed after suspending cells in
gates (10 µg mL−1 for free Adpgk) and/or 10 µg mL−1 CpG. At the in- DAPI solution for counting only DAPI-negative live and intact cells.
dicated time points, cells were collected, washed with FACS buffer (1% In Vivo Cancer Therapy: For MC38 tumor study, C57BL/6 mice were
BSA in PBS), and then subjected to flow cytometry for measuring fluo- subcutaneously inoculated with 5 × 105 MC38 cells into the right flank
rescence signals. To visualize cellular localization, BMDCs were grown and randomly sorted for treatment after 9 days. The mice were intra-
onto 12 mm glass coverslips in 24 well plates at a density of 5 × 105 tumorally administered with 50 µl PBS solution of Adpgk vaccine for-
cells/well and treated with samples as above for 24 h. Cells were fur- mulations at the dose of 30 µg PEI–Adpgk conjugates (equivalent of
ther incubated with Hoechst 33342 (5 µg mL−1 , Invitrogen) and Lyso- 10 µg for free Adpgk) and 15 µg CpG. In some cases, samples were
tracker Red DND-99 (100 nM, Invitrogen) for 30 min for the staining of administered into tail-base subcutaneous site for subcutaneous injec-
nuclei and endolysosomes, respectively. Then, cells were fixed with 4% tion or tail-vein for intravenous injection (100 µl in PBS). For anal-
formaldehyde in PBS and mounted on slide glass using ProLong™ Di- ysis of neoantigen-specific CD8+ T cells in systemic circulation, sub-
amond Antifade Mountant (Invitrogen) for confocal microscopy (Nikon mandibular bleeding was performed at the indicated time points and
A1Rsi). For TLR-9 signaling study, HEK-blue TLR-9 cells (Invivogen) were PBMCs were collected after removing red blood cells using ACK lysis
treated with PEI–Adpgk conjugates or their NPs at the dose of 2 µg mL−1 buffer. PBMCs were incubated with CD16/32 FcR blocking antibody and
PEI conjugates and 1 µg mL−1 CpG in HEK-Blue Detection medium. Af- then stained with Adpgk peptide-MHC tetramer tagged with PE (H-2Db -
ter 8 h, absorbance at 650 nm was measured using a microplate reader restricted ASMTNMELM, NIH Tetramer Core Facility) and anti-CD8-APC
to analyze induction of TLR-9 signaling in the cells, with the correction (BD Biosciences, No. 553035). For analysis of tumor infiltrating lympho-
of the sample effect by subtracting the absorbance of samples without cytes, tumor tissues were collected 7 days after sample administration,
TLR-9 cells. For the analysis of activation and antigen cross-presentation, cut into small pieces, treated with 1 mg mL−1 of collagenase type IV
BMDCs were incubated with PEI-SIINFEKL conjugates or their NPs for 24 and 0.1 mg mL−1 of DNase I in RPMI for 30 min at 37 °C, and filtered
h, with the dose at 2 µg mL−1 PEI conjugates or free SIINFEKL and 1 µg through a 70-µm strainer. Then, the cell suspension was washed with
mL−1 CpG. Cells were collected, washed with FACS buffer, incubated with FACS buffer, incubated with CD16/32 FcR blocking antibody, and stained
CD16/32 FcR blocking antibody (Invitrogen, No. 14016186) for 10 min, with the following antibody-fluorophore conjugates; Perforin-FITC (In-
and then stained with antibody-fluorophore conjugates including CD80- vitrogen, No. 11939280), Granzyme-PE/Cy7 (Invitrogen, No. 25889882),
FITC (BD Biosciences, No. 553768), CD86-PE/Cy7 (BD Biosciences, No. CD45-PerCP/Cy5.5 (Invitrogen 45045182), Adpgk peptide-MHC tetramer-
560582), CD40-APC (Invitrogen, No. 17040182) and SIINFEKL/H-2kb -PE PE, CD8-APC for CD8+ T cells, Perforin-FITC, Granzyme-PE/Cy7, CD45-
(Invitrogen, No. 12574382) for 30 min at room temperature. After was- PerCP/Cy5.5, NK1.1-PE (Invitrogen, 12594182) for NK cells, and CD45-
ing with FACS buffer, cell were analyzed using flow cytometry. To visualize PerCP/Cy5.5, Foxp3-PE/Cy7 (Invitrogen, No. 25577382) CD4-APC (Invitro-
antigen cross-presentation, BMDCs were grown onto 12 mm glass cov- gen, No. 17004282) for CD4+ T cells and CD4+ Foxp3+ Tregs. Flow cytom-
erslips in 24 well plates at a density of 5 × 105 cells/well, treated with etry was performed after suspending cells in DAPI solution and gating out
samples for 24 h, and further incubated with Hoechst 33342 for 30 min. DAPI-positive populations. For B16F10 tumor study, C57BL/6 mice were
Then, cells were incubated with CD16/32 FcR blocking antibody, permeabi- injected with 3 × 105 B16F10 cells subcutaneously into the right flank and
lized with Cytofix/Cytoperm Fixation/Permeabilization Solution (BD Bio- 4 × 105 B16F10 cells intravenously into tail vein, for locally established
sciences), and antibody-stained with SIINFEKL/H-2kb -biotin (Invitrogen, tumors and lung metastasis, respectively. The subcutaneous tumors were
No. 13574381) and LAMP1-AF488 (Invitrogen, No. 53107182). After fur- subjected to intratumoral administration of samples in 50 µl PBS, with the
ther staining with streptavidin-AF594 (Molecular Probes, No. S32356), dose at 15 µg PEI-M27 conjugate (equivalent of 3.5 µg for free M27) and
cells were washed with PBS and mounted on slide glass using ProLong™ 7.5 µg CpG for M27 vaccine and 15 µg PEI-M27 conjugate, 15 µg PEI-M30
Diamond Antifade Mountant for confocal microscopy. conjugate (equivalent of 5.5 µg for free M30), and 15 µg CpG for M27/M30
In Vivo Tumor Retention and Lymph Node Draining Studies: Animals vaccine. Animals were randomly sorted on day 7 and received samples ev-
were cared for following the federal, state, and local guidelines. The Uni- ery 3 days for total 3 times, followed by euthanization on day 17 for the
versity of Michigan, Ann Arbor is an AAALAC international accredited in- analysis of splenocyte ELISPOT and lung metastasis. For ELISPOT assay,
stitution, and all work conducted on animals was in accordance with and spleens were ground with the rubber end of a syringe, filtered through a
approved by the Institutional Animal Care and Use Committee (IACUC) 70-µm strainer, and treated with ACK lysis buffer for removing red blood
with the protocol # PRO00008587. Female C57BL/6 mice (5–6 weeks) cells. The obtained splenocytes were plated at 2 × 105 cells/well in 96-
were purchased from Jackson Laboratory (USA). C57BL/6 mice were sub- well PVDF plates pre-coated with IFN-𝛾𝛾 antibody (BD Biosciences), and
cutaneously inoculated with 5 × 105 MC38 cells into the right flank and re-stimulated overnight with 10 µg mL−1 of M27 or M30 peptide. Then,
randomly sorted for treatment after 9 days when tumor size reached ap- the wells were sequentially treated with biotinylated-secondary antibody,
proximately 5 mm. The mice were administered intratumorally with 50 µl streptavidin alkaline phosphatase, and AEC Substrate (BD Biosciences).
PBS solution of Adpgk vaccine formulations with CpG-AF647 at the dose The developed spots were counted using AID iSpot Reader (AID GmbH,
of 30 µg PEI–Adpgk conjugates (equivalent of 10 µg for free Adpgk) and Germany). For the analysis of lung metastasis, lungs were excised, fixed
15 µg CpG. For the analysis of tumor retention, tumors were excised 24 overnight in 4% formaldehyde, and then B16F10 lung tumor nodules were
h after sample administration and their fluorescence intensity was mea- enumerated manually. The sizes of locally established tumors were mea-
sured using IVIS optical imaging system (Caliper Life Sciences). For up- sured twice a week using a digital caliper, and the tumor volume was es-
take in a cellular level, tumors were cut into small pieces, incubated with 1 timated by ellipsoidal calculation as V = (width)2 × length × 1/2. The
mg mL−1 of collagenase type IV and 0.1 mg mL−1 of DNase I in RPMI for mice were euthanized when the tumors reached the maximum permitted
30 min at 37 °C, and filtered through a 70-µm strainer. The obtained sin- size (1.5 cm in any dimension) or ulcerations occurred.
gle cell suspension was washed with FACS buffer, and their fluorescence Statistical Analysis: For animal studies, the mice were randomized to
signal was measured using flow cytometry. For the analysis of DCs and match similar average volume of the locally established tumors. The data
macrophages in lymph node, inguinal and axillary lymph nodes were col- show mean ± s.d. (n = 5–8). Data were approximately normally dis-
lected 24 h after sample administration, ground with the rubber end of tributed and variance was similar between the groups. Statistical analysis
a syringe, and filtered through a 70-µm strainer. The cell suspension was was performed with Prism 8.1.0 software (GraphPad Software) by one-
washed with FACS buffer, incubated with CD16/32 FcR blocking antibody, way or two-way ANOVA with Bonferroni multiple comparisons post-test.
and stained with the following antibody-fluorophore conjugates; CD80- Statistical significance for survival curve was calculated by the log-rank
FITC (BD Biosciences, No. 553768), CD40-PE (Invitrogen, No. 12040183), (Mantel–Cox) test. All data were included for the statistical analysis with
CD86-PE/Cy7 (BD Biosciences, No. 560582), CD11c-APC (BioLegend, No. the significance indicated as *P < 0.05, **P < 0.01, ***P < 0.001, and
117309) for CD11c+ DCs, and CD11b-PE (Invitrogen, No. 12011282), ****P < 0.0001.
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
74
www.advancedsciencenews.com www.advancedscience.com
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
75
www.advancedsciencenews.com www.advancedscience.com
[12] S. M. Moghimi, P. Symonds, J. C. Murray, A. C. Hunter, G. Debska, [23] a) F. Wegmann, K. H. Gartlan, A. M. Harandi, S. A. Brinckmann, M.
A. Szewczyk, Mol. Ther. 2005, 11, 990. Coccia, W. R. Hillson, W. L. Kok, S. Cole, L.-P. Ho, T. Lambe, M. Puthia,
[13] S. L. Snyder, P. Z. Sobocinski, Anal. Biochem. 1975, 64, 284. C. Svanborg, E. M. Scherer, G. Krashias, A. Williams, J. N. Blattman,
[14] a) H. Petersen, P. M. Fechner, A. L. Martin, K. Kunath, S. Stolnik, P. D. Greenberg, R. A. Flavell, A. E. Moghaddam, N. C. Sheppard, Q.
C. J. Roberts, D. Fischer, M. C. Davies, T. Kissel, Bioconjugate Chem. J. Sattentau, Nat. Biotechnol. 2012, 30, 883; b) N. C. Sheppard, S. A.
2002, 13, 845; b) M. Ogris, G. Walker, T. Blessing, R. Kircheis, M. Brinckmann, K. H. Gartlan, M. Puthia, C. Svanborg, G. Krashias, S. C.
Wolschek, E. Wagner, J. Controlled Release 2003, 91, 173; c) M. Ogris, Eisenbarth, R. A. Flavell, Q. J. Sattentau, F. Wegmann, Int. Immunol.
S. Brunner, S. Schüller, R. Kircheis, E. Wagner, Gene Ther. 1999, 6, 2014, 26, 531; c) E. V. Grant, M. Thomas, J. Fortune, A. M. Klibanov,
595; d) M. Kursa, G. F. Walker, V. Roessler, M. Ogris, W. Roedl, R. N. L. Letvin, Eur. J. Immunol. 2012, 42, 2937; d) M. Bivas-Benita, L.
Kircheis, E. Wagner, Bioconjugate Chem. 2003, 14, 222. Bar, G. O. Gillard, D. R. Kaufman, N. L. Simmons, A.-H. Hovav, N. L.
[15] a) J. S. Suk, Q. Xu, N. Kim, J. Hanes, L. M. Ensign, Adv. Drug Delivery Letvin, J. Virol. 2010, 84, 5764; e) M. Rodrigo Garzón, P. Berraondo,
Rev. 2016, 99, 28; b) J. Nam, N. Won, J. Bang, H. Jin, J. Park, S. Jung, J. Crettaz, L. Ochoa, M. Vera, J. J. Lasarte, A. Vales, N. Van Rooijen,
S. Jung, Y. Park, S. Kim, Adv. Drug Delivery Rev. 2013, 65, 622. J. Ruiz, J. Prieto, J. Zulueta, G. González-Aseguinolaza, Vaccine 2005,
[16] F. Veglia, V. A. Tyurin, D. Mohammadyani, M. Blasi, E. K. Duper- 23, 1384; f) B.-S. Shim, S.-M. Park, J.-S. Quan, D. Jere, H. Chu, M. K.
ret, L. Donthireddy, A. Hashimoto, A. Kapralov, A. Amoscato, R. An- Song, D. W. Kim, Y.-S. Jang, M.-S. Yang, S. H. Han, Y.-H. Park, C.-S.
gelini, S. Patel, K. Alicea-Torres, D. Weiner, M. E. Murphy, J. Klein- Cho, C.-H. Yun, BMC Immunol. 2010, 11, 65.
Seetharaman, E. Celis, V. E. Kagan, D. I. Gabrilovich, Nat. Commun. [24] a) Y.-F. Ma, Y.-W. Yang, Eur. J. Pharm. Sci. 2010, 40, 75; b) Z. Sun, B.
2017, 8, 2122. Liu, X. Ruan, Q. Liu, Mol. Med. Rep. 2014, 10, 2657.
[17] L. Jeanbart, M. Ballester, A. de Titta, P. Corthésy, P. Romero, J. A. [25] a) R. N. Palumbo, X. Zhong, C. Wang, J. Controlled Release 2012, 157,
Hubbell, M. A. Swartz, Cancer Immunol. Res. 2014, 2, 436. 86; b) R. M. Steinman, M. C. Nussenzweig, Proc. Natl. Acad. Sci. USA
[18] a) S. Aras, M. R. Zaidi, Br. J. Cancer 2017, 117, 1583; b) J. M. Jaynes, R. 2002, 99, 351.
Sable, M. Ronzetti, W. Bautista, Z. Knotts, A. Abisoye-Ogunniyan, D. [26] H. Petersen, P. M. Fechner, D. Fischer, T. Kissel, Macromolecules 2002,
Li, R. Calvo, M. Dashnyam, A. Singh, T. Guerin, J. White, S. Ravichan- 35, 6867.
dran, P. Kumar, K. Talsania, V. Chen, A. Ghebremedhin, B. Karanam, [27] M. Jäger, S. Schubert, S. Ochrimenko, D. Fischer, U. S. Schubert,
A. Bin Salam, R. Amin, T. Odzorig, T. Aiken, V. Nguyen, Y. Bian, J. C. Chem. Soc. Rev. 2012, 41, 4755.
Zarif, A. E. de Groot, M. Mehta, L. Fan, X. Hu, A. Simeonov, N. Pate, [28] N. P. Gabrielson, D. W. Pack, Biomacromolecules 2006, 7, 2427.
M. Abu-Asab, M. Ferrer, N. Southall, C.-Y. Ock, Y. Zhao, H. Lopez, S. [29] C. C. Norbury, Immunology 2006, 117, 443.
Kozlov, N. de Val, C. C. Yates, B. Baljinnyam, J. Marugan, U. Rudloff, [30] a) T. Kawai, S. Akira, Immunity 2011, 34, 637; b) L. A. J. O’Neill, D.
Sci. Transl. Med. 2020, 12, eaax6337; c) T. M. Raimondo, D. J. Mooney, Golenbock, A. G. Bowie, Nat. Rev. Immunol. 2013, 13, 453.
Proc. Natl. Acad. Sci. USA 2018, 115, 10648; d) Q. Li, A. Shen, Z. [31] a) S. Yang, S. May, J. Chem. Phys. 2008, 129, 185105; b) A. Akinc,
Wang, RSC Adv. 2020, 10, 16537. M. Thomas, A. M. Klibanov, R. Langer, The J. Gene Med. 2005, 7,
[19] a) A. Marabelle, L. Tselikas, T. de Baere, R. Houot, Ann. Oncol. 2017, 657.
28, xii33; b) M. A. Aznar, N. Tinari, A. J. Rullán, A. R. Sánchez-Paulete, [32] O. P. Joffre, E. Segura, A. Savina, S. Amigorena, Nat. Rev. Immunol.
M. E. Rodriguez-Ruiz, I. Melero, J. Immunol. 2017, 198, 31. 2012, 12, 557.
[20] D. S. Chen, I. Mellman, Immunity 2013, 39, 1. [33] P. Nair-Gupta, A. Baccarini, N. Tung, F. Seyffer, O. Florey, Y.
[21] a) O. Boussif, F. Lezoualc’h, M. A. Zanta, M. D. Mergny, D. Scherman, Huang, M. Banerjee, M. Overholtzer, P. A. Roche, R. Tampé, B. D.
B. Demeneix, J. P. Behr, Proc. Natl. Acad. Sci. USA 1995, 92, 7297; Brown, D. Amsen, S. W. Whiteheart, J. M. Blander, Cell 2014, 158,
b) U. Lungwitz, M. Breunig, T. Blunk, A. Göpferich, Eur. J. Pharm. 506.
Biopharm. 2005, 60, 247. [34] S. Nierkens, M. H. den Brok, T. Roelofsen, J. A. L. Wagenaars, C. G.
[22] a) C. Shen, J. Li, Y. Zhang, Y. Li, G. Shen, J. Zhu, J. Tao, Int. J. Nanomed. Figdor, T. J. Ruers, G. J. Adema, PLoS One 2009, 4, 8368.
2017, 12, 5443; b) K. Regnström, E. G. E. Ragnarsson, M. Köping- [35] J. Nam, S. Son, J. J. Moon, Cell Mol. Bioeng. 2017, 10, 341.
Höggård, E. Torstensson, H. Nyblom, P. Artursson, Gene Ther. 2003, [36] M. B. Lutz, N. Kukutsch, A. L. J. Ogilvie, S. Rößner, F. Koch, N. Ro-
10, 1575. mani, G. Schuler, J. Immunol. Methods 1999, 223, 77.
Adv. Sci. 2021, 8, 2002577 2002577 © 2021 The Authors. Advanced Science published by Wiley-VCH GmbH
76
StarBright Dyes for flow cytometry
StarBright Dyes are unique fluorescent nanoparticles conjugated
to Bio-Rad’s highly validated immunology antibodies. Developed
specifically for flow cytometry, StarBright Dyes have narrow excitation
and emission spectra and give you exceptional brightness without the
need for a special buffer. StarBright Dyes are currently available for use
with violet, blue, and ultraviolet lasers.
#ScienceForward