Did you look at whether there was any correlation between the CD4:C...
Can you speculate on why you saw a difference in the frequency of P...
Do you think that responders are likely to have more antigen experi...
Do you think that these phenotypes would also been seen and predict...
Immunotherapy Advances, 2023, 3, 1–13
https://doi.org/10.1093/immadv/ltad001
Advance access publication 6 January 2023
Research Article
Stratification of PD-1 blockade response in melanoma
using pre- and post-treatment immunophenotyping of
peripheral blood
Natalie M.Edner
1,*,
, ElisavetNtavli
1
, LinaPetersone
1
, Chun JingWang
1
, AstridFabri
1
,
AlexandrosKogimtzis
1
, VitalijsOvcinnikovs
1
, Ellen M.Ross
1
, FrankHeuts
1
, YassinElfaki
1
,
Luke P.Houghton
1
, TobyTalbot
2
, AmnaSheri
3
, AlexandraPender
3
, DavidChao
3,
and
LucyS.K.Walker
1,,
1
Institute of Immunity & Transplantation, University College London Division of Infection & Immunity, London, UK
2
Royal Cornwall Hospitals NHS Trust, Truro, UK
3
Department of Oncology, Royal Free London NHS Foundation Trust, London, UK
These authors contributed equally to this work.
*
Correspondence: Natalie M. Edner, Institute of Immunity & Transplantation, University College London Division of Infection & Immunity, Royal Free Campus,
London, NW3 2PP, UK. Email: natalie.edner.15@ucl.ac.uk
Summary
Efficacy of checkpoint inhibitor therapies in cancer varies greatly, with some patients showing complete responses while others do not re-
spond and experience progressive disease. We aimed to identify correlates of response and progression following PD-1-directed therapy by
immunophenotyping peripheral blood samples from 20 patients with advanced malignant melanoma before and after treatment with the PD-1
blocking antibody pembrolizumab. Our data reveal that individuals responding to PD-1 blockade were characterised by increased CD8 T cell prolif-
eration following treatment, while progression was associated with an increase in CTLA-4-expressing Treg. Remarkably, unsupervised clustering
analysis of pre-treatment T cell subsets revealed differences in individuals that went on to respond to PD-1 blockade compared to individuals that
did not. These differences mapped to expression of the proliferation marker Ki67 and the costimulatory receptor CD28 as well as the inhibitory
molecules 2B4 and KLRG1. While these results require validation in larger patient cohorts, they suggest that flow cytometric analysis of a relatively
small number of T cell markers in peripheral blood could potentially allow stratification of PD-1 blockade treatment response prior to therapy initiation.
Graphical Abstract
Keywords: checkpoint inhibition, PD-1 blockade, immunotherapy, biomarker research
Received: August 17, 2022; Accepted: January 4, 2023
© The Author(s) 2023. Published by Oxford University Press on behalf of the British Society for Immunology.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/),
which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
2 N.M. Edner et al.
Abbreviations: CR: Complete responder; FCS: Foetal calf serum; MFI: Median uorescence intensity; PBMC: Peripheral blood mononuclear cell; PBS: Phosphate-
buffered saline; PCA: Principal component analysis; PD: Progressive disease; PR: Partial responder; SD: Stable disease; Tfr: Follicular regulatory T cells; TIL:
Tumour-inltrating lymphocyte; Treg: Regulatory T cells.
Introduction
Harnessing the power of the immune system in the form of
immunomodulatory drugs has revolutionised cancer therapy,
with checkpoint inhibitors targeting the coinhibitory PD-1
and CTLA-4 pathways now being the standard of care in the
treatment of many types of malignancies [1]. These therapies
aim to reinvigorate tumour-specic T cell responses and re-
lease tumour-inltrating lymphocytes (TILs) from tumour
suppression. In particular, antibodies blocking PD-1 or its li-
gand PD-L1 have caused a paradigm shift in cancer care with
an unprecedented 5-year overall survival of 34% for patients
with advanced melanoma treated with the PD-1 inhibitor
pembrolizumab [2].
Nevertheless, the clinical response following PD-1 pathway
interventions shows great heterogeneity and varies across in-
dividual patients and different types of tumours [3]. Therefore,
considerable effort has been directed towards identication of
biomarkers that stratify patient response following therapy.
For example, expression of PD-L1 on cells residing within the
tumour has been shown to have some positive correlation with
response, although patients with PD-L1-negative tumours can
still respond to PD-1 blockade [4, 5]. Furthermore, a higher
tumour mutational burden and abundance of neoantigens
expressed by tumour cells is favourable since it can facilitate
anti-tumour immune responses [6, 7]. Similarly, the presence
of TILs, particularly CD8 T cells, is associated with a better
response following PD-1 blockade [8, 9].
However, to predict patient response using these markers,
tumour biopsies, ideally taken at multiple sites, are required,
which is not always feasible, and robust biomarkers there-
fore still present an unmet clinical need in cancer immu-
notherapy. As an alternative, peripheral blood can provide
a snapshot of the systemic immune response and is easily
accessible. Tumour-specic T cells have been detected in the
circulation and T cell clones reinvigorated by PD-1 blockade
can be found in the blood as well as tumour inltrates
[1012]. Relatively few studies have investigated potential
biomarkers for response to PD-1 blockade in ex vivo periph-
eral blood lymphocytes and those that have been completed
primarily focus on CD8 T cells [1318]. Consequently, we
sought to identify immune correlates of clinical response fol-
lowing PD-1 blockade by immunophenotyping both CD4
and CD8 T cells in fresh peripheral blood samples. We
have previously performed immunophenotyping of circu-
lating lymphocytes in autoimmune settings where we have
identied biomarkers of response to immunotherapy [19,
20]. Drawing on this experience, a range of ow cytometry
panels were designed and applied to blood samples from
patients with advanced malignant melanoma before and
after anti-PD-1 therapy.
Methods and materials
Patients
Patients with advanced malignant melanoma of cutaneous
or mucosal origin were recruited at the Royal Free Hospital
NHS Foundation Trust as part of the PASIP research study
(NCT02909348). The protocol and consent document of
this trial were approved by the London - Bromley Research
Ethics Committee (16/LO/1296). All participants were above
the age of 18, provided written, informed consent and were
deidentied prior to analysis. Patient characteristics of the
study cohort are displayed in Supplementary Table S1. Patients
were treated with 2 mg/kg of pembrolizumab (humanised
monoclonal anti-PD-1, Keytruda, MSD) intravenously on
day 1 of each 21-day cycle until progression or unacceptable
toxicity developed. Blood samples were typically taken before
treatment and then 6 and 12 weeks after treatment initiation.
Assessment of patient response was performed prior to cycle
5 infusion by the treating physician according to RECIST ver-
sion 1.1 [21].
Sample preparation
Peripheral blood mononuclear cells (PBMCs) were isolated
from whole blood by density gradient centrifugation. In
brief, whole blood was diluted in phosphate-buffered saline
(PBS, Merck) and layered over Histopaque 1077 (Merck) in
LeucoSep 50 ml tubes (Greiner Bio-One). After centrifuga-
tion at 800 × g for 15 minutes with no brake, supernatant
above barrier was poured into new 50 ml Falcon tube and
centrifuged at 350 × g for 10 minutes to remove remaining
histopaque. Supernatant was discarded and pellet resuspended
in PBS + 2% foetal calf serum (FCS, Life Science Production).
Platelets were removed by centrifugation at 250 × g for 10
minutes. The resulting pellet was resuspended in PBS + 2%
FCS and 1 × 10
6
cells per ow cytometry panel were used for
subsequent ow cytometry staining.
Flow cytometry
PBMCs were surface stained with four panels of antibody
cocktails (see Supplementary Table S2) for 15 minutes at
37°C. For panel 1, cells were then incubated with streptavidin
APC for 10 minutes at 4°C. Cells were incubated with xable
viability dye eFluor 780 (Thermo Fisher Scientic) in PBS for
10 minutes at 4°C. For intracellular staining in panel 2, cells
were xed and permeabilized using the Foxp3/Transcription
Factor Staining Buffer Set (Thermo Fisher Scientic) and
incubated with an antibody cocktail containing anti-human
Ki67 Alexa Fluor 488 (clone: Ki-67, Biolegend), CTLA-4 PE
(clone: BNI3, BD Biosciences) and Foxp3 APC (clone: 236A/
E7, Thermo Fisher Scientic) for 30 minutes at 4°C. Samples
were acquired on a BD LSRFortessa (BD Biosciences) using
the BD FACSDiva software v.8.0.2 (BD Biosciences).
Data analysis
For manual analysis, ow cytometry data were analysed
using FlowJo v.10 (FlowJo LLC). For unsupervised clustering,
pregated live, singlet lymphocytes were preprocessed in R
v.4.0.2 as previously described [20] and the FlowSOM al-
gorithm as implemented in the Bioconductor package
CATALYST v.1.14.1 was used to identify CD4 and CD8 T
cell populations. FlowSOM clustering was then applied again
on these T cell populations and optimal number of clusters
was identied using delta area plots.
For tSNE projections, ow cytometry data were
downsampled to 3000 cells per sample and CATALYST was
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Immune correlates of αPD-1 response in melanoma 3
used to compute tSNE embedding. Principal component anal-
ysis (PCA) was performed on scaled and centred data. Prior to
PCA, cluster correlation was calculated using Pearson’s r and,
for cluster pairs that were correlated with an r ≥ 0.95, one
cluster was randomly removed from analysis. Statistical anal-
ysis was performed using R. A two-tailed Student’s t test was
used to compare two unpaired means. Plots were produced
using the CRAN packages ggplot2 v.3.3.5, ggpubr v.0.4.0,
ggtext v.0.1.1, cowplot v.1.1.1, scico v.1.3.0, and circlize
v.0.4.13 and the Bioconductor packages ComplexHeatmap
v.2.6.2. Data cleaning and formatting was carried out using
CRAN packages tidyr v.1.1.4, reshape2 v.1.4.4, Rmisc v.1.5,
rstatix v.0.7.0, and lubridate v.1.8.0.
Data sharing statement
Deidentied individual participant data supporting the
ndings of this study are available from the corresponding
author upon request.
Results
Incomplete PD-1 blockade on CD4 T cells in
non-responders
In order to investigate the effect of PD-1 blockade on pe-
ripheral immune cells, blood samples from 20 patients
with advanced malignant melanoma were collected in the
PASIP study. In this study, patients received treatment with
pembrolizumab every 3 weeks and blood samples were
processed at baseline and 6 and 12 weeks following treatment
initiation (Fig. 1A). Response was assessed at the pre-cycle 5
(12 weeks) timepoint according to RECIST version 1.1 [21].
In our patient cohort, four patients responded to therapy (one
complete responder [CR] and three partial responders [PR]),
six patients had an intermediate response (stable disease [SD])
and 10 patients failed to respond to therapy (progressive dis-
ease [PD]). The focus of this study was to identify immune
correlates of response and we therefore chose to primarily
compare patients who responded (CR/PR) and who failed to
respond (PD) when stratifying by clinical response. Age, sex,
and ECOG performance status were comparable across re-
sponse groups (Fig. 1A).
PBMCs isolated from collected blood samples were analysed
using ow cytometry and we rst sought to determine whether
PD-1 blockade altered the frequencies of CD4 and CD8 T
cells, as well as naïve and memory T cell subsets regardless of
response. We observed no differences in the CD4:CD8 T cell
ratio after treatment with pembrolizumab (Fig. 1B). Similarly,
frequencies of naïve and memory populations within CD4 and
CD8 T cells remained unchanged following PD-1 blockade
(Fig. 1C, Supplementary Fig. 1).
A
B
C
BL 6WK
12WK
2 mg/kg pembrolizumab every 3 weeks
Pre-cycle 5
Assessment
Sample collection
CR /
PR
SD PD
n4
61
0
age (s.d.)
70.5
(10.9)
75.0
(12.6)
72.4
(11.9)
%Male 25% 16.7% 50%
Median
ECOG PS
000
Figure 1. PASIP study cohort. Patients with advanced malignant melanoma were treated with 2 mg/kg pembrolizumab (αPD-1) every 3 weeks. Blood
samples were analysed using flow cytometry at baseline and 6 and 12 weeks following treatment initiation. (A) Left: Graphic representation of PASIP
study protocol. BL = Baseline, 6WK = 6 weeks, 12WK = 12 weeks. Right: Patient age, sex, and ECOG performance status (PS) across response
groups. CR = complete response, PR = partial response, SD = stable disease, PD = progressive disease; s.d., standard deviation; PS, performance
status. (B) Ratio of CD4:CD8 T cells at indicated time points. Shown are means + SD. Two-tailed Student’s t test; ns, not significant. (C) Distribution of
naïve (CD45RA+ CD62L+), central memory (CM, CD45RA- CD62L+), effector memory (EM, CD45RA- CD62L-) and terminally differentiated (TEMRA,
CD45RA+ CD62L-) CD4 (left) and CD8 (right) T cells. Shown are means of samples at indicated time points. BL, n = 19; 6WK, n = 20; 12WK, n = 16.
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
4 N.M. Edner et al.
Pembrolizumab precludes binding of the αPD-1 antibody
clone J105 used for ow cytometric staining [22], which
allowed us to examine occupancy of PD-1 by the thera-
peutic antibody on peripheral T cells. As expected, PD-1 on
CD4 and CD8 T cells was almost undetectable following
pembrolizumab treatment (Fig. 2A and B, Supplementary Fig.
2A). Surprisingly, when we split patients by clinical response,
we saw that more PD-1 tended to be detectable on CD4 T
cells in patients not responding to therapy, and this reached
statistical signicance at the 6-week timepoint (Fig. 2C,
Supplementary Fig. 2B). This was not the case for CD8 T cells
(Supplementary Fig. 2C). The higher residual PD-1 staining in
non-responders did not simply reect higher expression prior
to treatment, since patients with the highest expression at 6
Figure 2. More PD-1 detectable on CD4 T cells in individuals failing to respond to PD-1 blockade. PBMCs were stained with αPD-1 antibodies before
and after pembrolizumab treatment. (A) Representative flow cytometry plots for PD-1 and CD45RA staining in CD4 T cells in one CR (top) and one
PD (bottom) patient at indicated time points. (B) PD-1+ frequency in CD4 T cells. BL, n = 19; 6WK, n = 20; 12WK, n = 16. (C) PD-1+ frequency in CD4
T cells in responders (CR/PR) and non-responders (PD). CR/PR, n = 4 (all time points); PD, n = 9 (BL), n = 10 (6WK), n = 6 (12WK). (B/C) Shown are
means + SD. Two-tailed Student’s t test; ****, P < 0.0001; *, P < 0.05; ns, not significant.
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Immune correlates of αPD-1 response in melanoma 5
weeks were not necessarily those with the highest expression
pre-treatment (Supplementary Fig. 2D). This suggests the pos-
sibility that in non-responders, PD-1 blockade on CD4 T cells
may be incomplete for reasons that remain unclear.
PD-1 blockade induces divergent immune changes
in responders and non-responders
PD-1 engagement suppresses T cell responses by dampening
signals necessary for efcient T cell stimulation [23].
Accordingly, blockade of PD-1 should enhance T cell activa-
tion and proliferation. We therefore evaluated expression of the
proliferation marker Ki67 in T cells following pembrolizumab
treatment. As expected, Ki67 expression was signicantly
increased in CD8 T cells 6 weeks following pembrolizumab
treatment initiation (Fig. 3A). There was also a trend towards
increased proliferation in CD4 T cells, however, this was not
statistically signicant (Supplementary Fig. 3A). To further
characterise the proliferating CD8 T cells, expression levels of
several markers were compared between the Ki67+ and Ki67-
fractions. Ki67+ CD8 T cells expressed higher intracellular
CTLA-4 and lower CD45RA and CD127 levels than the non-
proliferating CD8 T cells (Fig. 3B). Moreover, CD28 expres-
sion was higher on Ki67+ CD8 T cells when compared with
Ki67- CD8 T cells. While PD-1 can inhibit T cell responses
by suppressing TCR signalling [24, 25], there is also evidence
that PD-1 engagement can lead to the dephosphorylation
of CD28, thereby dampening CD28 downstream signalling
[2628]. Stratication of CD28+Ki67+ and CD28-Ki67+
CD8 T cells by clinical response revealed that there was a
signicantly higher frequency of CD28+Ki67+ CD8 T cells in
responders than in non-responders at the 6-week timepoint,
with the CR patient showing the highest levels of Ki67 (Fig.
3C–E, Supplementary Fig. 3B). While a trend for increased
Ki67 expression in responders at the 6-week timepoint was
also observable when looking at the whole CD8 T cell popula-
tion this did not reach statistical signicance (Supplementary
Fig. 3C). Ki67 expression in CD4 T cells also did not differ
between clinical response groups (Supplementary Fig. 3D).
Taken together, these data suggest that response following
PD-1 blockade is marked by increased proliferation in CD28-
expressing CD8 T cells.
We were next interested in identifying changes occurring
specically in non-responders to pembrolizumab treatment.
When investigating CD4 T cell subsets, we found that in
patients that did not respond to therapy, the frequency of
regulatory T cells (Treg, CD25+CD127-Foxp3+CTLA-4+)
transiently increased 6 weeks post-treatment initiation and
then returned to baseline by the 12-week timepoint (Fig.
4A, Supplementary Fig. 4A). Interestingly, when evaluating
expression of Treg markers in non-responder samples using
median uorescence intensity (MFI) in all CD4 T cells, we
saw that only expression of CTLA-4 increased signicantly
following pembrolizumab treatment, while other markers,
including Foxp3, CD25, and CD127 remained consistent
over the treatment period (Fig. 4B). This increase was also
observed when analysing gated Treg (Supplementary Fig.
4B and C). Additionally, Treg in non-responders but not
responders showed increased expression of CXCR3 and ICOS
after therapy (Fig. 4C, Supplementary Fig. 4D) and there was
also a trend towards increased Ki67 expression in the Treg
compartment (Supplementary Fig. 4E). Therefore, an increase
in CTLA-4 expressing Treg is associated with non-response
following PD-1 blockade.
As frequencies of immune cell populations can have limited
applicability when clinically evaluating response to immuno-
therapy in individual patients, we sought to evaluate whether
assessing ratios of certain immune cell subsets would provide
a more robust approach for stratifying patient responses.
Since we saw increases in activated and proliferating Treg
(CXCR3+ICOS+ and Ki67+, respectively) in non-responders
following PD-1 blockade and conversely higher frequencies
of corresponding CD8 T cells in responders (Fig. 3;
Supplementary Fig. 5A), we investigated whether Treg:CD8
ratios would provide a way to distinguish clinical response.
Both the CXCR3+ICOS+ Treg to CXCR3+ICOS+ CD8 T
cell ratio and Ki67+ Treg to Ki67+CD28+ CD8 T cell ratio
were skewed to being signicantly higher in non-responders
6 weeks following pembrolizumab treatment initiation
(Fig. 4D and E, Supplementary Fig. 5B and C). Specically,
an average ratio of 1 CXCR3+ICOS+ CD8 T cell to 19.8
CXCR3+ICOS+ Treg was observed in non-responders while
responders showed an average ratio of 1 to 7.3. The ratio
of Ki67+CD28+ CD8 T cells to Ki67+ Treg was 1 to 14.9
in non-responders and 1 to 2.7 in responders. Importantly,
this does not simply reect the overall Treg:CD8 ratio which
was not signicantly different between responders and non-
responders at all timepoints investigated (Supplementary
Fig. 5D). Therefore, assessing the ratios of activated and
proliferating Treg and CD8 T cells may provide a way to
stratify clinical response following pembrolizumab treat-
ment. Additionally, we found that CXCR3+ICOS+ Treg and
CD8 T cells positively correlated with their Ki67 expressing
counterparts at the 6-week timepoint (Supplementary Fig.
5E and F), suggesting that CXCR3 and ICOS co-expression
could potentially be used as a surrogate marker for prolifera-
tion in these cell types.
Clinical response following PD-1 blockade can be
distinguished at baseline
Results presented thus far focus on response-specic changes
occurring in peripheral T cells following PD-1 blockade,
particularly 6 weeks post-treatment initiation. However, in
many malignancies, the therapeutic window is limited and
biomarkers to predict patient response to immunotherapy
before treatment would be highly benecial. Accordingly,
we were interested to see whether we could identify immune
signatures in baseline bleeds that could stratify patients based
on their clinical response following PD-1 blockade.
In order to maximise the information obtained from the
ow cytometry data, we decided to use the unsupervised
clustering algorithm FlowSOM [29] to identify as many im-
mune cell subsets as possible. Across the four ow cytometry
panels used for analysis, a total of 156 CD4 and CD8 T cell
clusters were detected (Supplementary Fig. 6). Following
pairwise correlation comparison, four of these clusters were
found to be highly correlated (Pearson correlation coefcient
> 0.95) to other clusters and therefore removed. This left 152
clusters which were used to conduct principal component
analysis (PCA). We reasoned that patient age may also inu-
ence clinical response and thus included this as a feature in
the analysis as well.
Remarkably, baseline bleeds of responders and non-
responders showed a clear separation along the rst prin-
cipal component (PC1), which accounts for 19.2% of the
variance in this dataset (Fig. 5A). We investigated the top 15
clusters contributing to PC1 in either direction to identify
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
6 N.M. Edner et al.
Figure 3. CD28-dependent increase in proliferating CD8 T cells indicative of response following PD-1 blockade. (A) Ki67+ frequency in CD8 T cells.
BL, n = 19; 6WK, n = 20; 12WK, n = 16. (B) MFI of indicated markers in Ki67+ or Ki67- CD8 T cells at the 6-week time point. n = 20. (C) Frequency of
Ki67+ CD28+ (top) and Ki67+ CD28- (bottom) in CD8 T cells stratified by response. CR/PR, n = 4 (all time points); SD, n = 6 (all time points); PD, n =
9 (BL), n = 10 (6WK), n = 6 (12WK). (D) tSNE projection of downsampled and pooled CD8 T cells of CR, PR, and PD samples. Colour indicates scaled
Ki67 expression. (E) tSNE projection shown in (D) of one CR and one PD sample at indicated time points. Colour indicates scaled Ki67 expression.
(A/C) Shown are means + SD. (B) Shown are box plots, with black horizontal line denoting median value, while box represents the IQRs (IQR, Q1–Q3
percentile) and whiskers show the minimum (Q1 − 1.5× IQR) and maximum (Q3 + 1.5× IQR) values. (A/B/C) Two-tailed Student’s t test; ****, P < 0.0001;
***, P < 0.001; **, P < 0.01; *, P < 0.05; ns, not significant.
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Immune correlates of αPD-1 response in melanoma 7
characteristics of the T cell populations driving this separa-
tion (Fig. 5B–D). One of the populations contributing most
to PC1 comprised CD8 T cells lacking expression of CD28
(panel2_CD8_9). Indeed, after manually gating CD28 expres-
sion, we saw that on average more than 50% of CD8 T cells in
patients that failed to respond were CD28- at baseline, while
this was only the case for about 25% of CD8 T cells in the
patients who responded to therapy (Fig. 5E, Supplementary
Figs. 3B and 7A). Conversely, one of the clusters associated
with response showed high Ki67 expression in CD28+ CD8
T cells (panel2_CD8_13). It is important to note that this
cluster was highly correlated with a Ki67 expressing CD4
conventional T cell cluster (panel2_CD4_16, Supplementary
Fig. 7B and C) which was consequently removed from the
features used for PCA. This, therefore, indicates that patients
responding to PD-1 blockade show higher baseline prolifera-
tion in CD8 as well as CD4 T cells.
Furthermore, we noted that clusters associated with non-
response to therapy predominantly exhibited an effector
memory or terminally differentiated effector memory phe-
notype, while clusters linked to response tended to be
central memory or naïve T cells. Among the latter, a
CD45RA+CD62L+ CD8 T cell cluster (panel4_CD8_14)
caught our attention, since it exhibited high CD38 expres-
sion and was associated with response following therapy.
We could conrm by manual gating that CD38hiCD62L+
CD8 T cells were indeed enriched in responders at baseline
(Supplementary Fig. 7D). Of possible interest, this population
also tended to be higher in frequency in responders compared
with non-responders at the 6- and 12-week timepoints and
displayed a remarkable increase in the patient showing a
complete response at the 6-week timepoint (Supplementary
Fig. 7D and E). Finally, two CD8 T cell clusters associated
with non-response following pembrolizumab treatment
showed high expression of the inhibitory receptors 2B4 and
KLRG1 (panel3_CD8_1 and panel3_CD8_4). Indeed, the
MFI of these two receptors in CD8 T cells was signicantly
higher in non-responders when compared with responders at
Figure 4. Ratio of activated and proliferating Treg and CD8 T cells is skewed in non-responders. (A) Frequency of Treg (CD25+ CD127- Foxp3+ CTLA-4+)
in CD4 T cells in responders and non-responders. (B) MFI of CD25, CD127, Foxp3, and intracellular CTLA-4 in CD4 T cells of non-responders (PD). (C)
CXCR3+ ICOS+ frequency in Treg (CD25+ CD127-). (D) Ratio of CXCR3+ ICOS+ frequency in Treg to CXCR3+ ICOS+ frequency in CD8 T cells. (E) Ratio
of Ki67+ frequency in Treg to Ki67+ CD28+ frequency in CD8 T cells. (A/C) Shown are means + SD. (B/D/E) Shown are box plots, with black horizontal
line denoting median value, while box represents the IQRs (IQR, Q1–Q3 percentile) and whiskers show the minimum (Q1 − 1.5× IQR) and maximum
(Q3 + 1.5× IQR) values. CR/PR, n = 4 (all time points); PD, n = 9 (BL), n = 10 (6WK), n = 6 (12WK). Two-tailed Student’s t test; *, P < 0.05; ns, not
significant.
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
8 N.M. Edner et al.
Figure 5. Clinical response following PD-1 blockade can be distinguished at baseline. FlowSOM clustering was applied to CD4 and CD8 T cells stained
with four distinct flow cytometry panels. (A) PCA on FlowSOM clusters of baseline samples of responders and non-responders. CR, n = 1; PR, n = 3;
PD, n = 7. (B) Top and bottom 15 FlowSOM clusters contributing to PC1 ordered by PC weight. (C) Heatmaps showing scaled MFI of indicated markers
in CD8 T cell clusters shown in (B). (D) Heatmaps showing scaled MFIs of indicated markers in CD4 T cell clusters shown in (B). Arrows in (C) and (D)
indicate directionality of associated PC1 weight. (E) CD28- frequency in CD8 T cells in baseline samples of responders and non-responders. (F) MFI
of 2B4 (top) and KLRG1 (bottom) in CD8 T cells normalised to MFI in naïve (CD45RA+ CCR7+) CD8 T cells. Data shown is from baseline samples of
responders and non-responders. (E/F) Shown are means + SD. CR/PR, n = 4; PD, n = 9. Two-tailed Student’s t test; **, P < 0.01; *, P < 0.05.
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Immune correlates of αPD-1 response in melanoma 9
baseline (Supplementary Fig. 8A and B, Supplementary Fig.
9). Since the MFI is subjective to the uorochrome and ow
cytometer used, we opted to normalise using the MFI of 2B4
and KLRG1 on naïve CD8 T cells, where it was equivalent
between responders and non-responders (Supplementary Fig.
8C). Using this approach, we saw a 2.5× increase in 2B4 ex-
pression and 2× increase in KLRG1 expression in all CD8
T cells compared with naïve CD8 T cells of non-responders,
while there was only a 1.4× and 1.3× increase, respectively, in
the responders (Fig. 5F, Supplementary Fig. 8D).
Taken together, these results suggest that it may be pos-
sible to gain insights into the clinical response following
pembrolizumab treatment by utilising ow cytometric anal-
ysis of CD4 and CD8 T cell subsets at baseline. Specically,
higher baseline proliferation in CD4 and CD8 T cells is as-
sociated with a response to therapy, while non-response is
associated with a CD8 T cell compartment featuring more
CD28-negative and inhibitory receptor-expressing cells.
Discussion
While checkpoint inhibitor therapies are now an estab-
lished treatment option in cancer therapy, clinical response
of patients is variable. Robust biomarkers of response fol-
lowing PD-1 blockade are still lacking and peripheral
blood immunophenotyping remains incompletely explored.
Using T cell-focussed ow cytometric analysis we have
identied a number of features that correlate with response
to pembrolizumab in patients with advanced malignant
melanoma.
We found that patients that responded to PD-1 blockade ex-
perienced a greater increase in expression of the proliferation
marker Ki67 particularly in CD28-expressing CD8 T cells.
This is in accordance with previously published data showing
an increase in CD8 T cell proliferation that correlates with
clinical response and is largely driven by CD28-expressing
CD8 T cells [13, 27]. Furthermore, we show that the fre-
quency of proliferating CD28+ CD8 T cells directly correlates
with the frequency of CXCR3- and ICOS-expressing CD8 T
cells at the 6-week timepoint, suggesting that the latter could
be used as surrogate markers for PD-1 induced proliferation,
avoiding the need for intracellular Ki67 staining. ICOS ex-
pression on CD8 T cells responding to PD-1 blockade has
previously been shown [13], while work by Chow et al.
demonstrated that CXCR3 expression is important for αPD-1
efcacy in a mouse tumour model [30].
Conversely, patients that failed to respond to PD-1 blockade
showed a transient increase in Treg 6 weeks after treatment
initiation. It has previously been shown that more Treg and
a higher frequency of proliferating Treg can be found in pe-
ripheral blood of melanoma patients when compared with
healthy controls [12, 31, 32] and presence of Treg within
the tumour is associated with a worse prognosis [33], con-
sistent with the idea that Treg suppress anti-tumour immune
responses [34]. The response of CD8 T cells to PD-1 blockade
has been extensively studied, however evidence of modulation
of Treg has only recently started to emerge. While initial data
did not show any correlation between increases in Treg and
clinical outcome following PD-1 blockade [12], later reports
showed that presence of proliferating Treg within the tumour
as well as peripheral blood following PD-1 blockade was as-
sociated with a poor prognosis [35, 36], consistent with our
ndings. The difference in observations may be attributed to
the markers used to identify Treg in these studies, with our
data showing that it is particularly Treg expressing CTLA-4
that are increased following therapy in non-responders. This
upregulation of CTLA-4 on Treg could be a result of enhanced
activation following PD-1 blockade and higher levels of
CTLA-4 are associated with increased Treg function [37].
CTLA-4 expression is also a key feature of highly suppres-
sive Treg found to be enriched in tumour tissue of breast and
lung cancer patients, which are associated with more aggres-
sive disease [3840]. Of note, recent analysis has highlighted
the potential for CTLA-4-expressing tumour-inltrating fol-
licular regulatory T cells (Tfr) to limit the effectiveness of
PD-1 inhibitors [41]. Together, these studies provide strong
biological context to our nding that non-responders are
characterised by increases in CTLA-4+ Treg. Since CTLA-4
is a key mediator of Treg suppression [42], it is possible that
inhibition of Treg function by anti-CTLA-4 antibodies might
prevent expanded Treg from limiting anti-tumour immu-
nity in these individuals, perhaps explaining why therapies
targeting both PD-1 and CTLA-4 work well together.
In our pre-treatment analysis, we identied that responders
showed a higher frequency of proliferating CD28-expressing
CD8 T cells as well as proliferating CD4 conventional T cells,
suggestive of ongoing immunity, potentially encompassing
anti-tumour immune responses. Conversely, non-responders
had higher frequencies of CD28- CD8 T cells, that are less
responsive to PD-1 therapy, and showed higher expression
of inhibitory receptors 2B4 and KLRG1 at baseline. CD28
expression on CD8 T cells is known to be downregulated by
repeated antigen stimulation and CD28- CD8 T cells accumu-
late with age [43, 44]. Both 2B4 and KLRG1 have previously
been linked to CD8 T cell exhaustion in chronic viral infec-
tion [45, 46] and targeting KLRG1 in combination with PD-1
blockade has been evaluated in an in vivo cancer model [47].
Interestingly, recent work has identied a population of CD8
T cells with immunoregulatory function that have lost CD28
and express both 2B4 and KLRG1 [48]. It is possible that the
non-responders in our cohort had higher levels of these sup-
pressive CD8 T cells prior to therapy.
While complete responses following PD-1 pathway
blockade have been observed, they only occur in a limited
number of individuals. Biomarkers to identify this patient
group would therefore be highly benecial. In our patient
cohort, only one individual showed a complete response to
pembrolizumab, precluding us from further investigating
correlates of response. However, we did observe a notable
increase in CD38hiCD62L+ CD8 T cells 6 weeks after treat-
ment initiation in this patient. This cell subset bears some re-
semblance to the circulating exhausted CD8 T cells shown
by Huang et al. to be reinvigorated following PD-1 blockade
[12] and could conceivably demark patients responding ex-
ceptionally well to therapy.
Finally, our anti-PD-1 antibody staining revealed a higher
frequency of unoccupied PD-1 on CD4 T cells at the 6-week
timepoint in non-responders. Most reports evaluate PD-1
expression using antibodies targeting the Fc region of the
relevant therapeutic antibody and there is relatively little
data evaluating PD-1 occupancy in patients following treat-
ment. Data from Das et al. showed that even when PD-1
is entirely occupied by therapeutic antibody in peripheral
blood, blockade of PD-1 on TILs is still incomplete [49].
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
10 N.M. Edner et al.
This analysis could suggest that if PD-1 blockade is not com-
plete in the periphery, blockade within the tumour may not
be sufcient either, potentially affecting treatment efcacy.
Additionally, Zappasodi et al. made use of an αPD-1 anti-
body that is not blocked by therapeutic αPD-1 antibodies
to show that PD-1hi CD4 T cells are decreased following
PD-1 blockade and that this reduction was less prevalent
in patients that failed to respond to therapy [50]. While
we cannot directly compare these PD-1hi CD4 T cells with
the PD-1+ CD4 T cells we observe after therapy in non-
responders, our data suggest that the relationship between
treatment efcacy and PD-1 receptor occupancy warrants
further investigation.
In summary, by conducting peripheral blood
immunophenotyping in patients with advanced malig-
nant melanoma treated with pembrolizumab, we identied
skewing of the peripheral immune response both at baseline
and shortly after treatment initiation that correlated with re-
sponse following PD-1 blockade. Specically, patients that
failed to respond to therapy displayed a more immune sup-
pressive phenotype with low CD28 expression on CD8 T
cells and an increase in Treg following therapy. Additionally,
we found that ratios of activated and proliferating Treg and
CD8 T cells could stratify patients by clinical response. Our
results were obtained in a limited number of patients, and it
will be important to validate these ndings in larger patient
cohorts. Nonetheless, our work suggests that T cell-directed
ow cytometric assays could provide a further tool to inform
rapid treatment decisions.
Supplementary material
Supplementary data are available at Immunotherapy
Advances online.
Figure S1. Frequencies of naïve and memory CD4 and
CD8 T cells does not change following PD-1 blockade. (A)
Frequency of naïve (top left, CD45RA+ CD62L+), central
memory (CM, top right, CD45RA- CD62L+), effector mem-
ory (EM, bottom left, CD45RA- CD62L-) and terminally
differentiated (TEMRA, bottom right, CD45RA+ CD62L-)
in CD4 T cells. (B) Frequency of naïve (top left, CD45RA+
CD62L+), central memory (CM, top right, CD45RA-
CD62L+), effector memory (EM, bottom left, CD45RA-
CD62L-) and terminally differentiated (TEMRA, bottom
right, CD45RA+ CD62L-) in CD8 T cells. Shown are means
+ s.d.. BL, n = 19; 6WK, n = 20; 12WK, n = 16. Two-tailed
Student’s t test; ns, not signicant.
Figure S2. PD-1 detection in CD8 T cells following PD-1
blockade is the same in responders and non-responders. (A)
PD-1+ frequency in CD8 T cells. BL, n = 19; 6WK, n = 20;
12WK, n = 16. (B) PD-1+ frequency in CD4 T cells strati-
ed by response. (C) PD-1+ frequency in CD8 T cells strat-
ied by response. (D) (top) PD-1+ frequency in CD4 T cells
in CR, PR, SD and PD patients. Points from same patient are
connected by lines. Colour indicates PD-1+ frequency at base-
line. (bottom) Ranking of PD-1 expression in CR, PR, SD and
PD patients at baseline and the 6-week timepoint. For P23 no
baseline bleed was available. (B/C/D) CR/PR, n = 4 (all time
points); SD, n = 6 (all time points); PD, n = 9 (BL), n = 10
(6WK), n = 6 (12WK). Shown are means + s.d.. Two-tailed
Student’s t test; ****, P < 0.0001; *, P < 0.05; ns, not signif-
icant.
Figure S3. Increase in proliferation in CD4 T cells does
not distinguish responders and non-responders. (A) Ki67+
frequency in CD4 T cells. (B) Representative ow cytometry
plots showing Ki67 and CD28 expression in CD8 T cells
in baseline, 6-week and 12-week bleeds of one PR and one
PD patient. (C) Ki67+ frequency in CD8 T cells stratied by
response. (D) Frequency of Ki67+ (left), Ki67+ CD28+ (mid-
dle) and Ki67+ CD28- (right) in CD4 T cells stratied by re-
sponse. (A) BL, n = 19; 6WK, n = 20; 12WK, n = 16. (C/D)
CR/PR, n = 4 (all time points); SD, n = 6 (all time points); PD,
n = 9 (BL), n = 10 (6WK), n = 6 (12WK). Shown are means +
s.d.. Two-tailed Student’s t test; ns, not signicant.
Figure S4. CTLA-4+ Treg are transiently increased in
non-responders following PD-1 blockade. (A) Frequency of
Treg (CD25+ CD127- Foxp3+ CTLA-4+) in CD4 T cells.
(B) MFI of intracellular CTLA-4 in Treg (CD25+ CD127-)
of non-responders. (C) Scaled histogram showing intra-
cellular CTLA-4 expression in naïve T cells (lled) or Treg
(open) from a non-responder at the indicated time points.
(D) CXCR3+ ICOS+ frequency in Treg (CD25+ CD127-). (E)
Ki67+ frequency in Treg. (A/D/E) Shown are means + s.d..
(B) Shown are box plots, with black horizontal line denoting
median value, while box represents the IQRs (IQR, Q1–Q3
percentile) and whiskers show the minimum (Q1 − 1.5× IQR)
and maximum (Q3 + 1.5× IQR) values. (A/B/D/E) CR/PR, n =
4 (all time points); SD, n = 6 (all time points); PD, n = 9 (BL),
n = 10 (6WK), n = 6 (12WK). Two-tailed Student’s t test; *, P
< 0.05; ns, not signicant.
Figure S5. CXCR3 and ICOS expression correlates with
proliferation of Treg and CD8 T cells. (A) CXCR3+ ICOS+
frequency in CD8 T cells. (B) Ratio of CXCR3+ ICOS+ fre-
quency in Treg to CXCR3+ ICOS+ frequency in CD8 T cells.
(C) Ratio of Ki67+ frequency in Treg to Ki67+ CD28+ fre-
quency in CD8 T cells. (D) Ratio of Treg frequency (CD25+
CD127- Foxp3+ CTLA-4+) to CD8 T cell frequency. (E)
Pearson correlation of Ki67+ frequency in Treg (CD25+
CD127- Foxp3+ CTLA-4+) to CXCR3+ ICOS+ frequency
in Treg (CD25+ CD127-). (F) Pearson correlation of Ki67+
frequency to CXCR3+ ICOS+ frequency in CD8 T cells.
(A) Shown are means + s.d.. (B/C/D) Shown are box plots,
with black horizontal line denoting median value, while box
represents the IQRs (IQR, Q1–Q3 percentile) and whiskers
show the minimum (Q1 − 1.5× IQR) and maximum (Q3 +
1.5× IQR) values. (A/B/C/D) CR/PR, n = 4 (all time points);
SD, n = 6 (all time points); PD, n = 9 (BL), n = 10 (6WK), n =
6 (12WK). Two-tailed Student’s t test; ***, P < 0.001; *, P <
0.05; ns, not signicant. (E/F) CR, n = 1; PR, n = 3; SD, n = 6;
PD, n = 10. Pearson’s R and associated p value are depicted on
plots. Black line only for visualisation purposes.
Figure S6. Heatmaps of maker expression in FlowSOM
clusters. FlowSOM clustering was applied to CD4 and CD8 T
cells stained with four distinct ow cytometry panels. Shown
are heatmaps of scaled MFIs of indicated markers in CD4
(left) and CD8 (right) T cells.
Figure S7. Clinical response following PD-1 blockade can
be distinguished using baseline bleeds. (A) CD28- frequency
in CD8 T cells in baseline samples. (B) Pearson correlation
of frequencies of FlowSOM clusters panel2_CD4_16 and
panel2_CD8_13. (C) Pearson correlation of manually gated
Ki67+ frequency in Tconv (non-Treg) and Ki67+ CD28+
frequency in CD8 T cells. (D) CD62L+ CD38hi frequency
in CD8 T cells. (E) Flow cytometry plots showing CD62L
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Immune correlates of αPD-1 response in melanoma 11
and CD38 expression in CD8 T cells in baseline and 6-week
bleeds of CR patient. (A/D) Shown are means + s.d.. CR/PR,
n = 4 (all time points); SD, n = 6 (all time points); PD, n =
9 (BL), n = 10 (6WK), n = 6 (12WK). Two-tailed Student’s t
test; **, P < 0.01; *, P < 0.05; ns, not signicant. (B/C) CR,
n = 1; PR, n = 3; SD, n = 6; PD, n = 7. Pearson’s R and as-
sociated p value are depicted on plots. Black line only for
visualisation purposes.
Figure S8. CD8 T cells of non-responders have higher ex-
pression of 2B4 and KLRG1 at baseline. (A) MFI of 2B4
(left) and KLRG1 (right) in CD8 T cells in baseline samples.
(B) Representative ow cytometry plots showing 2B4 and
KLRG1 expression in CD8 T cells in baseline bleed of one PR
patient and one PD patient. (C) MFI of 2B4 (left) and KLRG1
(right) in naïve (CD45RA+ CCR7+) CD8 T cells in baseline
samples. (D) MFI of 2B4 (left) and KLRG1 (right) in CD8 T
cells normalised to MFI in naïve (CD45RA+ CCR7+) CD8 T
cells. Data shown is from baseline samples. (A/C/D) Shown
are means + s.d.. CR/PR, n = 4; SD, n = 6; PD, n = 9. Two-
tailed Student’s t test; **, P < 0.01; ns, not signicant.
Figure S9. Gating strategies. Shown are representative
gating strategies for relevant CD4 and CD8 T cell populations
in indicated ow cytometry panels. For all panels, cells in rst
gate are live, singlet Lymphocytes. Grey background indicates
same parent gate.
Table S1. Patient characteristics. Shown are characteristics
of patients enrolled in the PASIP study.
Table S2. Flow cytometry surface panels. Shown are the
four ow cytometry panels of antibodies that were used for
surface staining of PBMCs in this study.
Acknowledgements
We gratefully acknowledge the contributions of Sachuda
Veerasamy, Leah Meaden, Kirsty Prout and Rebecca Sargent
in organising blood sample provision. The Editor-in-Chief,
Tim Elliott, and handling editor, Stephanie Dougan, would
like to thank the following reviewers, an anonymous reviewer
and Andreas Beilhack, for their contribution to the publica-
tion of this article.
Author contributions
N.M.E. assisted with the experiments, analysed the data, pre-
pared the gures and wrote the manuscript. E.N. performed
the experiments and edited the manuscript. L.P., C.J.W., A.F.,
A.K., V.O., E.M.R., F.H., Y.E. and L.P.H. assisted with the
experiments and edited the manuscript. T.T., A.S. and A.P.
provided clinical samples and edited the manuscript. D.C.
conceptualised and wrote the study protocol, applied for fund-
ing, supervised clinical sample provision, assessed treatment
response and edited the manuscript. L.S.K.W conceptualised
and supervised the study, designed the experiments, applied
for funding and wrote the manuscript.
Funding
Funding for this study was provided by a research grant
from the Investigator-Initiated Studies Program of Merck
Sharp & Dohme Limited. The opinions expressed in this pa-
per are those of the authors and do not necessarily repre-
sent those of Merck Sharp & Dohme Limited. This work as
also supported by CancerResearchUK grant no. C58264/
A26593 and Medical Research Council grant no. MR/
N001435/1. This project has received funding from the
European Union’s Horizon 2020 research and innovation
programme under the Marie Skłodowska-Curie grant agree-
ment no. 955321.
Conflict of interest
L.S.K.W. and D.C. received funding from Merck Sharp &
Dohme Limited for this study. T.T. has received travel grants
and honoraria from Merck Sharp & Dohme Limited.
Permission to reproduce
Not applicable to this manuscript.
Clinical trial registration
The PASIP research study is listed at ClinicalTrials.gov and
can be found under the identier NCT02909348. The proto-
col and consent document of this trial were approved by the
London - Bromley Research Ethics Committee (16/LO/1296).
ARRIVE guidelines
Not applicable to this manuscript.
Data availability
Deidentied individual participant data supporting the
ndings of this study are available from the corresponding
author upon request.
References
1. Robert C. A decade of immune-checkpoint inhibitors in cancer
therapy. Nat Commun 2020; 11(1):3801.https://doi.org/10.1038/
s41467-020-17670-y
2. Hamid O, Robert C, Daud A et al. Five-year survival outcomes
for patients with advanced melanoma treated with pembrolizumab
in KEYNOTE-001. Ann Oncol 2019; 30(4):582–8. https://doi.
org/10.1093/annonc/mdz011
3. Ribas A, Wolchok JD. Cancer immunotherapy using check-
point blockade. Science 2018; 359(6382):1350–5. https://doi.
org/10.1126/science.aar4060
4. Daud AI, Wolchok JD, Robert C et al. Programmed death-ligand 1
expression and response to the anti-programmed death 1 antibody
pembrolizumab in melanoma. J Clin Oncol 2016; 34(34):4102–9.
https://doi.org/10.1200/jco.2016.67.2477
5. Topalian SL, Hodi FS, Brahmer JR et al. Safety, activity, and
immune correlates of anti–PD-1 antibody in cancer. N Engl
J Med 2012; 366(26):2443–54. https://doi.org/10.1056/
nejmoa1200690
6. Gubin MM, Zhang X, Schuster H et al. Checkpoint blockade
cancer immunotherapy targets tumour-specic mutant antigens.
Nature 2014; 515(7528):577–81. https://doi.org/10.1038/na-
ture13988
7. Rizvi NA, Hellmann MD, Snyder A et al. Mutational landscape
determines sensitivity to PD-1 blockade in non-small cell lung
cancer. Science 2015; 348(6230):124–8. https://doi.org/10.1126/
science.aaa1348
8. Daud AI, Loo K, Pauli ML et al. Tumor immune proling predicts
response to anti-PD-1 therapy in human melanoma. J Clin Invest
2016; 126(9):3447–52. https://doi.org/10.1172/jci87324
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
12 N.M. Edner et al.
9. Tumeh PC, Harview CL, Yearley JH et al. PD-1 blockade induces
responses by inhibiting adaptive immune resistance. Nature 2014;
515(7528):568–71. https://doi.org/10.1038/nature13954
10. Gros A, Parkhurst MR, Tran E et al. Prospective identication
of neoantigen-specic lymphocytes in the peripheral blood of
melanoma patients. Nat Med 2016; 22(4):433–8. https://doi.
org/10.1038/nm.4051
11. Gros A, Tran E, Parkhurst MR et al. Recognition of human gas-
trointestinal cancer neoantigens by circulating PD-1+ lymphocytes.
J Clin Invest 2019; 129(11):4992–5004. https://doi.org/10.1172/
jci127967
12. Huang AC, Postow MA, Orlowski RJ et al. T-cell invigoration to
tumour burden ratio associated with anti-PD-1 response. Nature
2017; 545(7652):60–5. https://doi.org/10.1038/nature22079
13. Kamphorst AO, Pillai RN, Yang S et al. Proliferation of PD-1+
CD8 T cells in peripheral blood after PD-1-targeted therapy in lung
cancer patients. Proc Natl Acad Sci USA 2017; 114(19):4993–8.
https://doi.org/10.1073/pnas.1705327114
14. Mazzaschi G, Facchinetti F, Missale G et al. The circulating pool of
functionally competent NK and CD8+ cells predicts the outcome
of anti-PD1 treatment in advanced NSCLC. Lung Cancer 2019;
127:153–63. https://doi.org/10.1016/j.lungcan.2018.11.038
15. Kim KH, Cho J, Ku BM et al. The rst-week proliferative response
of peripheral blood PD-1+CD8+ T cells predicts the response
to Anti-PD-1 therapy in solid tumors. Clin Cancer Res 2019;
25(7):2144–54.https://doi.org/10.1158/1078-0432.ccr-18-1449
16. Juliá EP, Mandó P, Rizzo MM et al. Peripheral changes in immune
cell populations and soluble mediators after anti-PD-1 therapy
in non-small cell lung cancer and renal cell carcinoma patients.
Cancer Immunol Immunother 2019; 68(10):1585–96. https://doi.
org/10.1007/s00262-019-02391-z
17. Zuazo M, Arasanz H, Fernández-Hinojal G et al. Functional sys-
temic CD4 immunity is required for clinical responses to PD -L1/ PD
-1 blockade therapy. EMBO Mol Med 2019; 11(7):e10293.https://
doi.org/10.15252/emmm.201910293
18. Ferrara R, Naigeon M, Auclin E et al. Circulating T-cell
immunosenescence in patients with advanced non-small cell
lung cancer treated with single-agent PD-1/PD-L1 inhibitors or
platinum-based chemotherapy. Clin Cancer Res 2021; 27(2):492–
503. https://doi.org/10.1158/1078-0432.ccr-20-1420
19. Edner NM, Wang CJ, Petersone L et al. Predicting clinical response
to costimulation blockade in autoimmunity. Immunother Adv
2021; 1(1):ltaa003. https://doi.org/10.1093/immadv/ltaa003
20. Edner NM, Heuts F, Thomas N et al. Follicular helper T cell proles
predict response to costimulation blockade in type 1 diabetes. Nat
Immunol 2020; 21(10):1244–55. https://doi.org/10.1038/s41590-
020-0744-z
21. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evalu-
ation criteria in solid tumours: revised RECIST guideline (version
1.1). Eur J Cancer 2009; 45(2):228–47.https://doi.org/10.1016/j.
ejca.2008.10.026
22. Zelba H, Bochem J, Pawelec G et al. Accurate quantication of
T-cells expressing PD-1 in patients on anti-PD-1 immunotherapy.
Cancer Immunol Immunother 2018; 67(12):1845–51. https://doi.
org/10.1007/s00262-018-2244-7
23. Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibi-
tory pathway. Nat Rev Immunol 2018; 18(3):153–67. https://doi.
org/10.1038/nri.2017.108
24. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W et al. Pro-
grammed cell death 1 forms negative costimulatory microclusters
that directly inhibit T cell receptor signaling by recruiting phos-
phatase SHP2. J Exp Med 2012; 209(6):1201–17. https://doi.
org/10.1084/jem.20112741
25. Mizuno R, Sugiura D, Shimizu K et al. PD-1 primarily targets TCR
signal in the inhibition of functional T cell activation. Front Immunol
2019; 10:630. https://doi.org/10.3389/mmu.2019.00630
26. Hui E, Cheung J, Zhu J et al. T cell costimulatory receptor
CD28 is a primary target for PD-1-mediated inhibition. Sci-
ence 2017;355(6332):1428–33. https://doi.org/10.1126/science.
aaf1292
27. Kamphorst AO, Wieland A, Nasti T et al. Rescue of exhausted
CD8 T cells by PD-1-targeted therapies is CD28-dependent. Sci-
ence 2017; 355(6332):1423–7. https://doi.org/10.1126/science.
aaf0683
28. Walker LSK. PD-1 and CTLA-4: two checkpoints, one pathway?
Sci Immunol 2017; 2(11):eaan3864. https://doi.org/10.1126/
sciimmunol.aan3864
29. Van Gassen S, Callebaut B, Van Helden MJ et al. FlowSOM:
using self-organizing maps for visualization and interpretation of
cytometry data. Cytom Part A 2015; 87(7):636–45. https://doi.
org/10.1002/cyto.a.22625
30. Chow MT, Ozga AJ, Servis RL et al. Intratumoral activity of the
CXCR3 chemokine system is required for the efcacy of anti-PD-1
therapy. Immunity 2019; 50(6):1498–1512.e5. https://doi.
org/10.1016/j.immuni.2019.04.010
31. Jandus C, Bioley G, Speiser DE et al. Selective accumulation of
differentiated FOXP3+ CD4+ T cells in metastatic tumor lesions
from melanoma patients compared to peripheral blood. Cancer
Immunol Immunother 2008; 57(12):1795–805. https://doi.
org/10.1007/s00262-008-0507-4
32. Krieg C, Nowicka M, Guglietta S et al. High-dimensional single-
cell analysis predicts response to anti-PD-1 immunotherapy. Nat
Med 2018; 24(2):144–53. https://doi.org/10.1038/nm.4466
33. Baumgartner JM, Gonzalez R, Lewis KD et al. Increased sur-
vival from stage IV melanoma associated with fewer regulatory
T cells. J Surg Res 2009; 154(1):13–20. https://doi.org/10.1016/j.
jss.2008.04.043
34. Antony PA, Piccirillo CA, Akpinarli A et al. CD8+ T cell immu-
nity against a tumor/self-antigen is augmented by CD4+ T helper
cells and hindered by naturally occurring T regulatory cells.
J Immunol 2005; 174(5):2591–601. https://doi.org/10.4049/
jimmunol.174.5.2591
35. Huang AC, Orlowski RJ, Xu X et al. A single dose of neoadjuvant
PD-1 blockade predicts clinical outcomes in resectable melanoma.
Nat Med 2019; 25(3):454–61. https://doi.org/10.1038/s41591-
019-0357-y
36. Kagamu H, Kitano S, Yamaguchi O et al. CD4+ T-cell immunity
in the peripheral blood correlates with response to Anti-PD-1
therapy. Cancer Immunol Res 2020; 8(3):334–44. https://doi.
org/10.1158/2326-6066.cir-19-0574
37. Ovcinnikovs V, Ross EM, Petersone L et al. CTLA-4-mediated
transendocytosis of costimulatory molecules primarily targets mi-
gratory dendritic cells. Sci Immunol 2019; 4(35):eaaw0902. https://
doi.org/10.1126/sciimmunol.aaw0902
38. Alvisi G, Brummelman J, Puccio S et al. IRF4 instructs effector
Treg differentiation and immune suppression in human cancer.
J Clin Invest 2020; 130(6):3137–50. https://doi.org/10.1172/
jci130426
39. Plitas G, Konopacki C, Wu K et al. Regulatory T cells exhibit dis-
tinct features in human breast cancer. Immunity 2016; 45(5):1122–
34. https://doi.org/10.1016/j.immuni.2016.10.032
40. De Simone M, Arrigoni A, Rossetti G et al. Transcriptional land-
scape of human tissue lymphocytes unveils uniqueness of tumor-
inltrating T regulatory cells. Immunity 2016; 45(5):1135–47.
https://doi.org/10.1016/j.immuni.2016.10.021
41. Eschweiler S, Clarke J, Ramírez-Suástegui C et al. Intratumoral fol-
licular regulatory T cells curtail anti-PD-1 treatment efcacy. Nat
Immunol 2021; 22(8):1052–63. https://doi.org/10.1038/s41590-
021-00958-6
42. Walker LSK. Treg and CTLA-4: two intertwining pathways to im-
mune tolerance. J Autoimmun 2013; 45(100):49–57. https://doi.
org/10.1016/j.jaut.2013.06.006
43. Posnett DN, Sinha R, Kabak S et al. Clonal populations of T cells
in normal elderly humans: the T cell equivalent to “Benign Mono-
clonal Gammapathy.”. J Exp Med 1994; 179(2):609–618. https://
doi.org/10.1084/jem.179.2.609
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Immune correlates of αPD-1 response in melanoma 13
44. Valenzuela HF, Effros RB. Divergent telomerase and CD28 expres-
sion patterns in human CD4 and CD8 T cells following repeated
encounters with the same antigenic stimulus. Clin Immunol 2002;
105(2):117–25. https://doi.org/10.1006/clim.2002.5271
45. Blackburn SD, Shin H, Haining WN et al. Coregulation of CD8+
T cell exhaustion by multiple inhibitory receptors during chronic
viral infection. Nat Immunol 2009; 10(1):29–37. https://doi.
org/10.1038/ni.1679
46. Utzschneider DT, Charmoy M, Chennupati V et al. T cell factor
1-expressing memory-like CD8+ T cells sustain the immune re-
sponse to chronic viral infections. Immunity 2016; 45(2):415–27.
https://doi.org/10.1016/j.immuni.2016.07.021
47. Tata A, Dodard G, Fugère C et al. Combination blockade of KLRG1
and PD-1 promotes immune control of local and disseminated
cancers. Oncoimmunology 2021; 10(1):1933808. https://doi.org/
10.1080/2162402X.2021.1933808
48. Li J, Zaslavsky M, Su Y et al. KIR+CD8+ T cells suppress patho-
genic T cells and ar active in autoimmune diseases and COVID-19.
Science 2022; 376(6590):eabi9591. https://doi.org/10.1126/sci-
ence.abi9591
49. Das R, Verma R, Sznol M et al. Combination therapy with Anti–
CTLA-4 and anti–PD-1 leads to distinct immunologic changes
in vivo. J Immunol 2015; 194(3):950–9. https://doi.org/10.4049/
jimmunol.1401686
50. Zappasodi R, Budhu S, Hellmann MD et al. Non-conventional In-
hibitory CD4+Foxp3−PD-1hi T cells as a biomarker of immune
checkpoint blockade activity. Cancer Cell 2018; 33(6):1017–1032.
e7. https://doi.org/10.1016/j.ccell.2018.05.009
Downloaded from https://academic.oup.com/immunotherapyadv/article/3/1/ltad001/6972738 by Queen Mary University of London user on 01 March 2023
Do you think that responders are likely to have more antigen experienced PD1+ve stem cell like CD8 T cells, whilst non-responders have more exhausted PD1+ve CD8 T cells? Can you speculate on why you saw a difference in the frequency of PD1+ CD4 T cells between clinical response groups but not a concomitant increase in CD4 T cell proliferation? Do you think that these phenotypes would also been seen and predict response in other cancer patients or is it melanoma-speicific? Did you look at whether there was any correlation between the CD4:CD8 ratio at baseline and response to Pembrolizumab?