ArticlePDF Available

Fc-Optimized Anti-CD25 Depletes Tumor-Infiltrating Regulatory T Cells and Synergizes with PD-1 Blockade to Eradicate Established Tumors

Authors:

Abstract and Figures

CD25 is expressed at high levels on regulatory T (Treg) cells and was initially proposed as a target for cancer immunotherapy. However, anti-CD25 antibodies have displayed limited activity against established tumors. We demonstrated that CD25 expression is largely restricted to tumor-infiltrating Treg cells in mice and humans. While existing anti-CD25 antibodies were observed to deplete Treg cells in the periphery, upregulation of the inhibitory Fc gamma receptor (FcγR) IIb at the tumor site prevented intra-tumoral Treg cell depletion, which may underlie the lack of anti-tumor activity previously observed in pre-clinical models. Use of an anti-CD25 antibody with enhanced binding to activating FcγRs led to effective depletion of tumor-infiltrating Treg cells, increased effector to Treg cell ratios, and improved control of established tumors. Combination with anti-programmed cell death protein-1 antibodies promoted complete tumor rejection, demonstrating the relevance of CD25 as a therapeutic target and promising substrate for future combination approaches in immune-oncology.
Content may be subject to copyright.
Report
Fc-Optimized Anti-CD25 Depletes Tumor-Infiltrating
Regulatory T Cells and Synergizes with PD-1
Blockade to Eradicate Established Tumors
Graphical Abstract
Highlights
dCD25 expression is largely restricted to Treg cells in mice and
humans
dFcgRIIb inhibits anti-CD25-mediated depletion of intra-
tumoral Treg cells
dFc-optimized anti-CD25 efficiently depletes intra-tumoral
Treg cells
dAnti-CD25 synergizes with PD-1 blockade to reject
established tumors
Authors
Frederick Arce Vargas,
Andrew J.S. Furness,
Isabelle Solomon, ..., Teresa Marafioti,
Karl S. Peggs, Sergio A. Quezada
Correspondence
k.peggs@ucl.ac.uk (K.S.P.),
s.quezada@ucl.ac.uk (S.A.Q.)
In Brief
Anti-CD25 antibodies have displayed
only modest therapeutic activity against
established tumors. Arce Vargas et al.
demonstrate that existing anti-CD25
antibodies fail to deplete intra-tumoral
Treg cells due to upregulation of FcgRIIb
within tumors. Fc-optimized anti-CD25
mediates effective depletion of tumor-
infiltrating Treg cells and synergizes with
PD-1 blockade to promote tumor
eradication.
Arce Vargas et al., 2017, Immunity 46, 577–586
April 18, 2017 ª2017 Elsevier Inc.
http://dx.doi.org/10.1016/j.immuni.2017.03.013
Immunity
Report
Fc-Optimized Anti-CD25 Depletes Tumor-Infiltrating
Regulatory T Cells and Synergizes with PD-1
Blockade to Eradicate Established Tumors
Frederick Arce Vargas,
1,2,11
Andrew J.S. Furness,
1,2,3,11
Isabelle Solomon,
1,2
Kroopa Joshi,
1,2,3
Leila Mekkaoui,
4
Marta H. Lesko,
1,2
Enrique Miranda Rota,
4
Rony Dahan,
5
Andrew Georgiou,
1,2
Anna Sledzinska,
1,2
Assma Ben Aissa,
1,2
Dafne Franz,
1,2
Mariana Werner Sunderland,
1,2
Yien Ning Sophia Wong,
1,2
Jake Y. Henry,
1,2
Tim O’Brien,
6
David Nicol,
3
Ben Challacombe,
6
Stephen A. Beers,
7
Melanoma TRACERx Consortium, Renal TRACERx Consortium,
Lung TRACERx Consortium, Samra Turajlic,
3,8
Martin Gore,
3
James Larkin,
3
Charles Swanton,
8,9
Kerry A. Chester,
4
Martin Pule,
2
Jeffrey V. Ravetch,
5
Teresa Marafioti,
10
Karl S. Peggs,
1,2,
*and Sergio A. Quezada
1,2,12,
*
1
Cancer Immunology Unit, University College London Cancer Institute, London WC1E 6DD, UK
2
Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
3
The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
4
Research Department of Oncology, UCL Cancer Institute, London WC1E 6DD, UK
5
Leonard Wagner Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
6
Guy’s and St. Thomas’ NHS Foundation Trust, London SE1 9RT, UK
7
Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton SO17 1BJ, UK
8
The Francis Crick Institute, London NW1 1AT, UK
9
Translational Cancer Therapeutics Laboratory, UCL Cancer Institute, London WC1E 6DD, UK
10
Department of Cellular Pathology, University College London Hospital, London NW1 2BU, UK
11
These authors contributed equally
12
Lead Contact
*Correspondence: k.peggs@ucl.ac.uk (K.S.P.), s.quezada@ucl.ac.uk (S.A.Q.)
http://dx.doi.org/10.1016/j.immuni.2017.03.013
SUMMARY
CD25 is expressed at high levels on regulatory
T (Treg) cells and was initially proposed as a target
for cancer immunotherapy. However, anti-CD25 anti-
bodies have displayed limited activity against estab-
lished tumors. We demonstrated that CD25 expres-
sion is largely restricted to tumor-infiltrating Treg
cells in mice and humans. While existing anti-CD25
antibodies were observed to deplete Treg cells in
the periphery, upregulation of the inhibitory Fc
gamma receptor (FcgR) IIb at the tumor site pre-
vented intra-tumoral Treg cell depletion, which may
underlie the lack of anti-tumor activity previously
observed in pre-clinical models. Use of an anti-
CD25 antibody with enhanced binding to activating
FcgRs led to effective depletion of tumor-infiltrating
Treg cells, increased effector to Treg cell ratios,
and improved control of established tumors. Com-
bination with anti-programmed cell death protein-1
antibodies promoted complete tumor rejection,
demonstrating the relevance of CD25 as a therapeu-
tic target and promising substrate for future combi-
nation approaches in immune-oncology.
INTRODUCTION
Regulatory T (Treg) cells are generally regarded as one of the
major obstacles to the successful clinical application of tumor
immunotherapy. It has been consistently demonstrated that
Treg cells contribute to the early establishment and progression
of tumors in murine models and that their absence results in
delay of tumor progression (Elpek et al., 2007; Golgher et al.,
2002; Jones et al., 2002; Onizuka et al., 1999; Shimizu et al.,
1999). In humans, high tumor infiltration by Treg cells and,
more importantly, a low ratio of effector T (Teff) cells to Treg cells,
is associated with poor outcomes in multiple solid cancers
(Shang et al., 2015). Conversely, a high Teff/Treg cell ratio is
associated with favorable responses to immunotherapy in both
humans and mice (Hodi et al., 2008; Quezada et al., 2006). To
date, most studies support the notion that targeting Treg cells,
either by depletion or functional modulation, may offer significant
therapeutic benefit, particularly in combination with other im-
mune modulatory interventions such as vaccines and check-
point blockade (Bos et al., 2013; Goding et al., 2013; Quezada
et al., 2008; Sutmuller et al., 2001).
Defining appropriate targets for selective interference with
Treg cells is therefore a critical step in the development of effec-
tive therapies. In this regard, CD25, also known as the inter-
leukin-2 high-affinity receptor alpha chain (IL-2Ra), was the first
surface marker used to identify and isolate Treg cells (Sakaguchi
et al., 1995) prior to the discovery of their master regulator, tran-
scription factor forkhead box P3 (FoxP3). It is also the most exten-
sively studied target for mediating Treg cell depletion. Whereas
CD25 is constitutively expressed on Treg cells and absent on
naive Teff cells, transient upregulation has been described
upon activation of Teff cells, although these observations derive
largely from in vitro studies (Boyman and Sprent, 2012).
A number of pre-clinical studies in mice have used the anti-
CD25 antibody clone PC-61 (rat IgG1, l), which partially depletes
Treg cells in the blood and peripheral lymphoid organs (Setiady
Immunity 46, 577–586, April 18, 2017 ª2017 Elsevier Inc. 577
et al., 2010), inhibits tumor growth, and improves survival when
administered before or soon after tumor challenge (Golgher
et al., 2002; Jones et al., 2002; Onizuka et al., 1999; Quezada
et al., 2008; Shimizu et al., 1999). However, the use of anti-
CD25 as a therapeutic intervention against established tumors
fails to delay tumor growth or prolong survival (Golgher et al.,
2002; Jones et al., 2002; Onizuka et al., 1999; Shimizu et al.,
1999). This has been attributed to several factors, including
poor T cell infiltration of the tumor (Quezada et al., 2008) and po-
tential depletion of activated effector CD8
+
and CD4
+
T cells that
upregulate CD25 (Onizuka et al., 1999). Early-phase clinical
studies exploring the use of vaccines in combination with dacli-
zumab (a humanized IgG1 anti-human CD25 antibody) (Jacobs
et al., 2010; Rech et al., 2012) or denileukin difitox (a recombinant
fusion protein combining human IL-2 and a fragment of diptheria
toxin) (Dannull et al., 2005; Luke et al., 2016) demonstrate a var-
iable impact on the number of circulating Treg cells and vaccine-
induced immunity. However, the limited indirect data assessing
intra-tumoral FoxP3 transcript levels provide no clear evidence
that Treg cells in the tumor microenvironment are effectively
reduced and anti-tumor activity has appeared disappointing
across all studies, with no demonstrable survival benefit.
The modest therapeutic activity in pre-clinical and clinical set-
tings and concern regarding potential depletion of activated Teff
cells has contributed to limited enthusiasm for the further evalu-
ation of anti-CD25 antibodies in combination with novel immu-
notherapies. However, recent data demonstrate the contribution
of intra-tumoral Treg cell depletion to the activity of immune
modulatory antibody-based therapies and the relevance of the
antibody isotype in this setting (Bulliard et al., 2014; Coe et al.,
2010; Selby et al., 2013; Simpson et al., 2013). We therefore
re-evaluated CD25 as target for Treg cell depletion and tumor
immunotherapy in vivo. We demonstrated that the lack of ther-
apeutic activity of the widely used anti-CD25 antibody (PC-61)
against established mouse tumors results from a failure to effec-
tively deplete intra-tumoral Treg cells. Optimizing FcgR binding
and antibody-dependent cell-mediated cytotoxicity (ADCC)
resulted in superior intra-tumoral Treg cell depletion and potent
synergy when combined with programmed cell death protein-1
(PD-1) blockade. We demonstrated high levels of CD25
expression on Treg but not Teff cells in human tumors,
highlighting this receptor as a clinical target and anti-CD25 as
a promising therapeutic strategy in combination with novel
immunotherapies.
RESULTS
CD25 Is Highly Expressed on Murine Tumor-Infiltrating
Treg Cells
We sought to evaluate the relative expression of CD25 on indi-
vidual T lymphocyte subsets within tumors (TILs) and draining
lymph nodes (LNs) of mice 10 days after tumor challenge.
CD25 expression appeared consistent across multiple models
of transplantable tumor cell lines of variable immunogenicity
including MCA205 sarcoma, MC38 colon adenocarcinoma,
B16 melanoma, and CT26 colorectal carcinoma, with a higher
percentage of CD25-expressing CD4
+
FoxP3
+
Treg cells
relative to CD4
+
FoxP3
and CD8
+
Teff cells (Figure 1A). In
contrast to in vitro studies, minimal expression of CD25 on the
Teff cell compartment was observed in vivo and the per-
centage of CD25-expressing Teff cells (CD8
+
= 3.08%–8.35%,
CD4
+
FoxP3
= 14.11%–26.87%) was significantly lower than
on Treg cells (83.66%–90.23%) (p < 0.001) (Figure 1B). CD25
expression was also observed on Treg cells present in LNs
and blood (data not shown). However, the level of expression,
based on mean fluorescence intensity (MFI), was significantly
lower than that observed on tumor-infiltrating Treg cells (Fig-
ure 1C). Based on these data, CD25 appeared an attractive
target for preferential depletion of Treg cells.
Anti-CD25-Mediated Depletion of Treg Cells Is Limited
to Lymph Nodes and Blood
Based on evidence demonstrating the contribution of intra-tu-
moral Treg cell depletion to the activity of immune modulatory
antibodies (Bulliard et al., 2014; Coe et al., 2010; Selby et al.,
2013; Simpson et al., 2013), we sought to compare the impact
of anti-CD25 (clone PC-61 rat IgG1, aCD25-r1) on the frequency
of Teff and Treg cells in the blood, LNs, and TILs of mice with es-
tablished tumors. We focused our analyses on the MCA205
model because of its higher immunogenicity in order to deter-
mine any potential negative impact of aCD25 on activated Teff
cells within tumors.
As previously described (Onizuka et al., 1999; Setiady et al.,
2010), administration of 200 mgofaCD25-r1 on days 5 and 7 after
tumor challenge resulted in a reduced frequency of CD25
+
cells
in all analyzed sites (Figures 1D and 1E) and a reduction in the
frequency of CD4
+
FoxP3
+
Treg cells in blood and LN (Figure 1F).
However, aCD25-r1 failed to deplete tumor-infiltrating Treg cells,
which demonstrated a CD4
+
FoxP3
+
CD25
phenotype after
therapy. Their frequency remained comparable to that of un-
treated mice (Figure 1F), potentially explaining the lack of effi-
cacy observed against established tumors in previous studies
despite an apparent reduction in CD25
+
T cells within the tumor
(Golgher et al., 2002; Jones et al., 2002; Onizuka et al., 1999;
Quezada et al., 2008; Shimizu et al., 1999).
We next investigated whether an antibody with optimized
ADCC activity could efficiently deplete intra-tumoral Treg cells
without significant impact on Teff cells. We replaced the
constant regions of the original aCD25 obtained from clone
PC-61 with murine IgG2a and kconstant regions (aCD25-
m2a), the classical mouse isotype associated with ADCC,
and compared its activity to that of aCD25-r1 in vivo. While
both antibody variants resulted in reduced expression of
CD25 on T cells and a reduction in the number of Treg cells
in blood and LNs, only aCD25-m2a resulted in depletion of
tumor-infiltrating Treg cells to levels comparable to those
observed with anti-cytotoxic T lymphocyte associated pro-
tein-4 (aCTLA-4, clone 9H10), which is known to preferentially
deplete Treg cells in the tumor but not the periphery (Figures
1D–1F; Selby et al., 2013; Simpson et al., 2013). In keeping
with these observations, both aCD25 isotypes resulted in an
increased Teff/Treg cell ratio in circulating lymphocytes and
LN, but only aCD25-m2a increased the intra-tumoral ratio in
a similar manner to aCTLA-4 (Figure 1G). Despite a reduction
in the number of circulating and LN-resident Treg cells, no
macroscopic, microscopic, or biochemical evidence of toxicity
was observed in the skin, lungs, or liver after multiple doses of
aCD25-m2a (Figures S1A–S1C).
578 Immunity 46, 577–586, April 18, 2017
High Expression of FcgRIIb Inhibits aCD25-r1-Mediated
Treg Cell Depletion in the Tumor
Anti-CD25-r1 has been described to deplete circulating Treg
cells by FcgRIII-mediated ADCC (Setiady et al., 2010). However,
its intra-tumoral activity has not been investigated. To determine
this, we characterized the expression of Fc-gamma receptors
(FcgRs) on different leukocyte subpopulations in the blood,
spleen, LN, and tumor of mice bearing MCA205 tumors
A
B
C
D
E
F
G
Figure 1. Anti-CD25-r1-Mediated Depletion of CD25
+
Regulatory T Cells Is Restricted to Blood and Lymph Nodes
(A–C) Mouse LNs and TILs were analyzed by flow cytomery 10 days after MCA205 (n = 10), MC38 (n = 5), B16 (n = 3), or CT26 (n = 3) tumor implantation.
(A) CD25 expression on T cell subsets in representative mice. Dotted lines indicate the gate.
(B and C) Percentage (B) and MFI (C) of CD25 in each T cell subset. Error bars show standard error of the mean (SEM). p values obtained by two-way analysis of
variance (ANOVA).
(D–G) Tumor-bearing mice were injected with 200 mgofaCD25-r1, aCD25-m2a, or aCTLA-4 on days 5 and 7 after MCA205 tumor implantation. Blood, LNs, and
TILs were harvested and processed on day 9 for flow cytometry analysis.
(D) Representative plots showing expression of CD25 (detected with antibody clone 7D4) and FoxP3 in CD3
+
CD4
+
T cells. Numbers show percentage of cells in
each quadrant.
(E) MFI of CD25 in CD4
+
FoxP3
+
Treg cells.
(F) Percentage of FoxP3
+
Treg cells of total CD3
+
CD4
+
T cells.
(G) CD8
+
/Treg cell ratios (n = 10). Experiment was repeated three times.
Immunity 46, 577–586, April 18, 2017 579
(Figures 2A and S2). The percentage of FcgR-expressing cells
appeared higher on tumor-infiltrating myeloid cells (granulocytic
cells, dendritic cells, and monocyte/macrophages) relative to all
other studied organs (Figures 2A and 2B). We then analyzed the
binding affinity of the two Fc variants of aCD25 to FcgRs (Fig-
ure 2C). As previously described (Nimmerjahn and Ravetch,
2005), the mIgG2a isotype binds to all FcgR subtypes with a
high activatory to inhibitory ratio (A/I). In contrast, the rIgG1
isotype binds with a similar affinity to a single activatory FcgR,
FcgRIII, as well as the inhibitory FcgRIIb, resulting in a low A/I
ratio (<1) (Figure 2C).
To determine which specific FcgRs were involved in aCD25-
mediated Treg cell depletion, we quantified the number of tu-
mor-infiltrating Treg cells in mice lacking expression of different
FcgRs (Figures 2D–2G). Analysis of Fcer1g
/
mice, which lack
expression of activating FcgRs (I, III, and IV), demonstrated a
complete absence of Treg cell depletion. Treg cell elimination
by aCD25-r1 in the periphery and by aCD25-m2a in the periph-
ery and tumor therefore results from FcgR-mediated ADCC and
not blocking of IL-2 binding to CD25 (Figure 2D). Depletion by
aCD25-m2a was not dependent on any individual activatory
FcgR, with Treg cell elimination maintained in both Fcgr3
/
and Fcgr4
/
mice (Figures 2E and 2F). In keeping with previous
studies (Setiady et al., 2010), we confirmed that depletion of pe-
ripheral Treg cells by aCD25-r1 depends on FcgRIII (data not
shown), but it fails to deplete in the tumor despite high intra-tu-
moral expression of this receptor (Figure 2E). Intra-tumoral
Treg cell depletion was, however, effectively restored in mice
AB
CGDFE
Figure 2. FcgRIIb Inhibits aCD25-r1-Mediated Treg Cell Depletion in Tumors
(A and B) Expression of FcgRs was measured by flow cytometry in leukocytes from blood, spleen, LNs, and MCA205 tumors (TIL) 10 days after tumor im-
plantation.
(A) Expression of FcgRs on granulocytes (CD11b
+
Ly6G
+
), conventional dendritic cells (cDCs) (CD11c
hi
MHC-II
+
), and monocyte/macrophages (Mono/M4)
(CD11b
+
Ly6G
NK1.1
CD11c
lo/neg
). Dotted lines indicate the gate, numbers show the percentage of positive cells.
(B) Cumulative data of FcgR expression in cell subpopulations (n = 3). Error bars represent SEM; the experiment was repeated three times.
(C) Binding affinity of rat IgG1 and mouse IgG2a isotypes to individual mouse FcgRs as determined by surface plasmon resonance (SPR).
(D–G) Percentage of CD4
+
FoxP3
+
Treg cells of total CD4
+
T cells in TILs of wild-type (WT, n = 5–10), Fcer1g
/
(n = 10), Fcgr3
/
(n = 5), Fcgr4
/
(n = 10), or
Fcgr2b
/
(n = 5) mice treated as in Figures 1D–1G.
580 Immunity 46, 577–586, April 18, 2017
A
DCB
EF
HG
Figure 3. Synergistic Effect of Anti-CD25-m2a and Anti-PD-1 Combination Results in Eradication of Established Tumors
Tumor-bearing mice were treated with 200 mgofaCD25 on day 5 and 100 mgofaPD-1 on days 6, 9, and 12 after tumor implantation.
(A) Growth curves of individual MCA205 tumors, showing the product of three orthogonal tumor diameters. The number of tumor-free survivors is shown in
each graph.
(legend continued on next page)
Immunity 46, 577–586, April 18, 2017 581
lacking expression of the inhibitory receptor FcgRIIb. In this
setting, intra-tumoral Treg cell depletion was comparable be-
tween aCD25-r1 and aCD25-m2a (Figure 2G). Therefore, the
lack of Treg cell depletion by aCD25-r1 in the tumor is explained
by its low A/I binding ratio and high intra-tumoral expression of
FcgRIIb. FcgRIIb has been associated with modulation of
ADCC in tumors (Clynes et al., 2000), and in this case inhibits
ADCC mediated by the single activatory receptor engaged by
the aCD25-r1 isotype.
Anti-CD25-m2a Synergizes with Anti-PD-1 to Eradicate
Established Tumors
To determine whether the enhanced intra-tumoral Treg cell-
depleting activity of aCD25-m2a could improve therapeutic out-
comes, we compared the anti-tumor activity of aCD25-m2a and
-r1 against established tumors. We administered a single dose
of aCD25 5 days after subcutaneous implantation of MCA205
cells, when tumors were established with an average diameter
of 4–5 mm. Consistent with the observed lack of capacity to
deplete intra-tumoral Treg cells (Figure 1F) and previous studies
(Golgher et al., 2002; Jones et al., 2002; Onizuka et al., 1999;
Quezada et al., 2008; Shimizu et al., 1999), aCD25-r1 failed to
control tumor growth. Conversely, growth delay and long-term
survival was observed in a proportion of mice receiving
aCD25-m2a (15.4%) (Figures 3A and 3B).
Based on its role in T cell regulation within the tumor microen-
vironment and the observed clinical activity of agents targeting
the PD-1-PD-L1 axis, we hypothesized that depletion of CD25
+
Treg cells and PD-1 blockade might be synergistic in combina-
tion. In the same model, blocking anti-PD-1 antibody (aPD-1,
clone RMP1-14) at a dose of 100 mg every 3 days was ineffective
in the treatment of established MCA205 tumors when used as
monotherapy or in combination with aCD25-r1 (Figures 3A and
3B). However, a single dose of aCD25-m2a followed by aPD-1
therapy eradicated established tumors in 78.6% of the mice, re-
sulting in long-term survival of more than 100 days (Figures 3A
and 3B). This activity was significantly reduced in the absence
of CD8
+
T cells (Figures S3A and S3B), demonstrating that tumor
elimination depends on the impact of the aPD-1 and aCD25
combination on both CD8
+
and Treg cell compartments, and
that overall effector T cell responses are not negatively impacted
by a depleting aCD25 antibody.
Similar findings were observed in MC38 and CT26 tumor
models, where aCD25-m2a had a partial therapeutic effect
that synergized with aPD-1 therapy (Figures 3C and 3D). Ac-
tivity was also observed against the poorly immunogenic
B16 melanoma tumor model when aCD25-m2a and aPD-1
were combined with a granulocyte-macrophage colony stimu-
lating factor (GM-CSF)-expressing whole tumor cell vaccine
(Gvax). As previously described, in this system, Gvax alone
failed to extend survival of tumor-bearing mice (Quezada
et al., 2006; van Elsas et al., 2001). Combination therapy
with aCD25-m2a and aPD-1 translated into a modest increase
in survival, which was not observed with aCD25-r1 and aPD-1
(Figure S4).
To understand the mechanisms underpinning the observed
synergy, we evaluated the phenotype and function of TILs in
MCA205 tumors at the end of the treatment protocol, 24 hr after
the third dose of aPD-1 (Figures 3E–3H). Monotherapy with
aPD-1 did not impact upon Teff cell proliferation (Figure 3E)
nor the number infiltrating the tumor, where a persisting high fre-
quency of Treg cells was observed (data not shown), resulting in
a low Teff/Treg ratio (Figure 3F) and lack of therapeutic activity.
Conversely, intra-tumoral Treg cell depletion with aCD25-m2a
resulted in a higher proportion of proliferating and interferon-g
(IFN-g)-producing CD4
+
and CD8
+
T cells in the tumor, corre-
sponding to a high Teff/Treg cell ratio and anti-tumor activity
(Figures 3E–3H). This effect was further enhanced in combina-
tion with aPD-1, which yielded even higher proliferation and a
1.6-fold increase in the number of IFN-g-producing CD4
+
and
CD8
+
T cells compared to aCD25-m2a alone. In contrast, the
observed lack of Treg cell depletion with aCD25-r1 resulted in
no change in Teff cell proliferation or IFN-gproduction, when
used as monotherapy or in combination with aPD-1 (Figures
3E–3H). Combination of aCD25 and aPD-1 therefore appeared
highly effective at rejecting established tumors, but only when
intra-tumoral Treg cells were efficiently depleted by aCD25 of
appropriate isotype.
CD25 Expression Profiles in Human Cancers Validate Its
Use as Target for Therapeutic Treg Cell Depletion
To validate the translational value of CD25 as a target for Treg
cell depletion, we analyzed the expression of CD25 on periph-
eral blood mononuclear cells (PBMCs) and TILs in patients with
advanced melanoma, early-stage non-small cell lung carci-
noma (NSCLC), and renal cell carcinoma (RCC) by flow cy-
tometry and multiplex immunohistochemistry (IHC). Despite
heterogeneity in clinical characteristics both within and
between studied cohorts (Tables S1–S3), CD25 expression
remained largely restricted to CD4
+
FoxP3
+
Treg cells (mean
% CD25
+
= 54.8% of Treg, 7.5% of CD4
+
FoxP3
, and 1.9%
of CD8
+
; p < 0.0001) (Figures 4A and 4B). Similar to murine
models, the level of CD25 expression, as assessed by MFI,
was significantly higher on CD4
+
FoxP3
+
Treg cells relative to
CD4
+
FoxP3
and CD8
+
T cells within all studied tumor sub-
types (mean MFI Treg = 190.0, CD4
+
FoxP3
+
= 34.5 and
CD8
+
= 17.9; p < 0.0001) (Figure 4C).
We further performed longitudinal assessment of CD25
expression in the context of immune modulation. Core biopsies
were performed on the same lesion at baseline and after either
four cycles of nivolumab (3 mg/kg Q2W) or two cycles of
pembrolizumab (200 mg Q3W) in patients with advanced kidney
cancer and melanoma, respectively (Table S4). Despite systemic
immune modulation, CD25 expression remained restricted to
(B) Survival of mice shown in (A).
(C and D) Survival of mice with MC38 or CT26 tumors treated as described above (n = 10 per condition).
(E) Percentage of Ki67
+
cells in tumor-infiltrating CD4
+
FoxP3
and CD8
+
T cells.
(F) CD4
+
FoxP3
/CD4
+
FoxP3
+
and CD8
+
/CD4
+
FoxP3
+
cell ratios.
(G and H) Representative histograms (G) and percentage (H) of IFN-g-producing CD4
+
and CD8
+
TILs in MCA205 tumors determined by intracellular staining after
ex vivo re-stimulation with PMA and ionomycin. Graphs show cumulative data of two separate experiments (n = 10).
582 Immunity 46, 577–586, April 18, 2017
FoxP3
+
Treg cells, even in areas of dense CD8
+
T cell infiltrate
evaluated by multiplex immunohistochemistry (Figures 4D and
4E). These findings confirmed the translational value of the
described pre-clinical data, lending further support to the
concept of selective therapeutic targeting of Treg cells via
CD25 in human cancers.
DISCUSSION
We have demonstrated that CD25 is an attractive target
for Treg cell depletion owing to its expression profile on
tumor-infiltrating T cells in both mice and humans. Contrary
to in vitro studies, minimal expression of CD25 on the effector
AB
C
ED
Figure 4. CD25 Is Highly Expressed on Treg Cell Infiltrating Human Tumors
(A) Representative histograms demonstrating CD25 expression on circulating (PBMC) and tumor-infiltrating (TIL) CD8
+
, CD4
+
FoxP3
, and CD4
+
FoxP3
+
T cell
subsets. Dotted lines indicate the gate.
(B and C) Quantification of CD25 expression (percentage [B] and MFI [C]) on individual T cell subsets inhuman melanoma (n = 11) , NSCLC (n = 9), and RCC (n = 8).
Error bars represent SEM; p values obtained by two-way ANOVA.
(D) Longitudinal analysis of CD25 expression in human melanoma and RCC lesions prior to (‘‘Ba seline’’) and during PD-1 blockade (‘‘On therapy’’). CD8 staining is
displayed in red, FoxP3 in blue, and CD25 in brown.
(E) Percentage of CD25 expression on CD8
+
and FoxP3
+
T cells at baseline and during PD-1 blockade. Plotted values derive from analysis of 10 340 high-power
fields per patient at each time point.
Immunity 46, 577–586, April 18, 2017 583
compartment was observed in vivo. The efficacy of aCD25 as
an anti-tumor therapy depends on Treg cell depletion in the tu-
mor microenvironment, which can be achieved only by using an
antibody isotype optimized for engagement of activating
FcgRs, capable of inducing ADCC. Our results demonstrated
that the limited efficacy observed in pre-clinical studies using
the aCD25 PC-61 monoclonal antibody with a rat IgG1 isotype
relates to ineffective or suboptimal intra-tumoral Treg cell
depletion, a consequence of its low A/I binding ratio and high
intra-tumoral expression of inhibitory FcgRIIb. This may also
explain the modest results observed in early clinical trials using
the anti-human CD25 antibody daclizumab. However, the
impact of aCD25 antibodies of varying IgG subclass remains
to be evaluated in humans.
Local depletion of tumor-infiltrating Treg cells by aCD25
monotherapy mediated only partial tumor control, suggesting
that further intervention is necessary to increase the intra-
tumoral Teff/Treg cell balance and promote effector T cell
activity. These data mirror those previously demonstrated for
aCTLA-4 antibodies, where targeting solely the Treg cell
compartment was ineffective in eradicating established
tumors, while targeting both Treg and Teff cell compartments
resulted in effective therapeutic synergy (Peggs et al., 2009).
Increased regulation of Teff cell responses by co-inhibitory im-
mune checkpoints in the tumor microenvironment might also
explain the modest responses observed in early-stage clinical
trials evaluating aCD25 antibodies in cancer patients (Jacobs
et al., 2010; Rech et al., 2012). Our data suggest that such re-
sponses could be enhanced through combination with thera-
pies that address this regulation including immune checkpoint
blockade or agonistic antibodies targeting immune co-stimula-
tory receptors.
Treg cell depletion can be achieved by targeting other
molecules highly expressed on Treg cells (Bulliard et al., 2014;
Coe et al., 2010; Selby et al., 2013; Simpson et al., 2013). While
combined blocking and depleting activity of specific immune
modulatory antibodies is effective against certain target mole-
cules, such as CTLA-4, it can also be deleterious owing to
simultaneous high expression on Teff cells. Differential expres-
sion is therefore critical; for example, in addition to its expres-
sion on Treg cells, PD-1 is highly expressed on activated
CD8
+
T cells. Anti-PD-1 antibodies therefore lose anti-tumor
activity when a depleting antibody isotype is employed (Dahan
et al., 2015).
Anti-PD-1 therapy now forms a key part of the treatment
paradigm for multiple solid malignancies, with response rates
varying between 20% and 30% when used as monotherapy
(Topalian et al., 2015). However, the majority of responses
are partial. This could be explained in part by tumor infiltration
with CD25
+
FoxP3
+
Treg cells that are unaffected by non-
depleting aPD-1 antibodies. In this setting another target
molecule specific to Treg cells is required in order to achieve
potential synergy through Treg cell depletion. Combination of
aCTLA-4 and aPD-1 therapy has achieved superior response
rates to either agent alone in patients with advanced melanoma
(Larkin et al., 2015). This may be the result of the cell-intrinsic
immune modulatory activity of aCTLA-4 and aPD-1 antibodies
and concomitant depletion of Treg cells by aCTLA-4, although
this second activity has not been demonstrated in vivo. Combi-
nation therapy results in higher immune-related toxicity, under-
scoring the need for alternative combinations balancing
maximal activity with minimal toxicity. We have demonstrated
that aCD25 therapy synergizes with blocking aPD-1 therapy,
provided Treg cells are depleted locally in the tumor.
Combining aPD-1 with aCD25-depleting antibodies might
improve the therapeutic window compared to the aCTLA-4
combination, as aCD25 lacks the additional cell-intrinsic im-
mune modulatory activity of aCTLA-4. Such hypotheses are
further supported by our model, in which only transient Treg
cell depletion was required for effective synergy, with no evi-
dence of immune-related toxicity. These data support further
evaluation of Fc-optimized aCD25 as a combination partner
in clinical trials.
EXPERIMENTAL PROCEDURES
Antibodies and Antibody Production
The sequence of the variable regions of the heavy and light chains of aCD25
were resolved from the PC-61.5.3 hybridoma by rapid amplification of cDNA
ends (RACE), cloned into the constant regions of murine IgG2a and kchains
and expressed in a stable K562 cell line generated by co-transduction with
murine leukemia virus-derived retroviral vectors encoding both chains. The
antibody was initially purified from supernatants with a protein G HiTrap
MabSelect column (GE Healthcare), dialyzed in phosphate-buffered saline
(PBS), concentrated, and filter-sterilized. For subsequent experiments, anti-
body production was outsourced to Evitria AG. Anti-CD25-r1 (PC-61.5.3),
aCTLA-4 (9H10), aPD-1 (RMP1-14), and aCD8 (2.43) were supplied by
BioXcell. The binding affinity of isotype variants to FcgRs was measured
by SPR in the Ravetch laboratory as described before (Nimmerja hn and Rav-
etch, 2005).
Tumor Experiments
Details of mouse strains, cell lines and flow cytometry antibodies are shown in
Supplemental Experimental Procedures. Mice were injected subcutaneously
with 5 310
5
MCA205, MC38, or CT26 cells or 5 310
4
B16 cells re-suspended
in PBS. Therapeutic antibodies were administered intraperitoneally at the time
points and doses shown in figure legends. Cell suspensions for flow cytometry
were prepared as described previously (Simpson et al., 2013). Tumors were
measured twice weekly and mice were euthanized when any orthogonal tumor
diameter reached 150 mm.
Human Study Oversight
Human data derives from three translational studies approved by local institu-
tional review board and Research Ethics Committee (Melanoma, REC no.
11/LO/0003; NSCLC, REC no.13/LO/1546; RCC, REC no. 11/LO/1996). All
were conducted in accordance with the provisions of the Declaration of Hel-
sinki and with Good Clinical Practice guidelines as defined by the International
Conference on Harmonization. All patients (or their legal representatives) pro-
vided written informed consent before enrollment.
Analysis of Human Tissue
For flow cytometry, cell suspensions were prepared with the same protocol
employed for mouse tissues (Simpson et al., 2013). Leukocytes were enriched
by gradient centrifugation with Ficoll-paque (GE Healthcare). Isolated live cells
were frozen at 80C and stored in liquid nitrogen until analysis.
Histopathology protocols are described in Supplemental Experimental
Procedures.
Data Analysis
Flow cytometry data were analyzed with FlowJo v10.0.8 (Tree Star). Statistical
analyses were done with Prism 6 (GraphPad Software); p values were calcu-
lated using Kruskall-Wallis and Dunn’s post hoc tests, unless otherwise indi-
cated (ns = p > 0.05; *p %0.05; **p %0.01; ***p %0.001; ****p %0.0001).
Kaplan-Meier curves were analyzed with the log-rank test.
584 Immunity 46, 577–586, April 18, 2017
SUPPLEMENTAL INFORMATION
Supplemental Information includes four figures, four tables, Supplemental
Experimental Procedures, and consortia memberships and can be found
with this article online at http://dx.doi.org/10.1016/j.immuni.2017.03.013.
AUTHOR CONTRIBUTIONS
S.A.Q. and K.S.P. conceived the project. F.A.V., A.J.S.F., K.S.P., and S.A.Q.
designed the experiments, analyzed the data, and wrote the manuscript.
F.A.V. and A.J.S.F. performed the experiments. I.S., K.J., L.M., M.H.L., A.G.,
A.S., A.B.A., D.F., M.W.S., Y.N.S.W., and J.Y.H. contributed experimentally.
E.M.R., R.D., S.A.B., K.A.C., M.P., and J.V.R. provided reagents and contrib-
uted scientifically. T.M. performed the histology analyses. T.O., D.N., B.C.,
S.T., M.G., J.L., C.S., and the TRACERx consortia coordinated clinical trials
and provided patient samples.
ACKNOWLEDGMENTS
We thank Josep Linares for his technical expertise. S.A.Q. is a Cancer
Research U.K. (CRUK) Senior Fellow (C36463/A22246) and is funded by a
Cancer Research Institute Investigator Award and a CRUK Biotherapeut ic Pro-
gram Grant (C36463/A20764). K.S.P. receives funding from the NIHR BTRU for
Stem Cells and Immunotherapies (167097), of which he is the Scientific
Director. None of the animal work described was funded by NIHR. This work
was undertaken at UCL Hospitals/UCL with support from the CRUK-UCL
Centre (C416/A18088), CRUK’s Lung Cancer Centre of Excellence (C5759/
A20465), the CRUK and Engineering and Physical Sciences Research Council
at King’s College London and UCL (C1519/A16463), the Cancer Immuno-
therapy Accelerator Award (CITA-CRUK) (C33499/A20265), CRUK’s Lung
TRACERx study (led by C. Swanton) (C11496/A17786), the Sam Keen Founda-
tion/RMH NIHR Biomedical Research Centre, Bloodwise (formerly Leukaemia
and Lymphoma Research) (08022/P4664), the Department of Health, and
CRUK funding schemes for National Institute for Health Research Biomedical
Research Centres and Experimental Cancer Medicine Centres.
Received: August 27, 2016
Revised: January 26, 2017
Accepted: February 9, 2017
Published: April 11, 2017
REFERENCES
Bos, P.D., Plitas, G., Rudra, D., Lee, S.Y., and Rudensky, A.Y. (2013). Transient
regulatory T cell ablation deters oncogene-driven breast cancer and enhances
radiotherapy. J. Exp. Med. 210, 2435–2466.
Boyman, O., and Sprent, J. (2012). The role of interleukin-2 during homeosta-
sis and activation of the immune system. Nat. Rev. Immunol. 12, 180–190.
Bulliard, Y., Jolicoeur, R., Zhang, J., Dranoff, G., Wilson, N.S., and Brogdon,
J.L. (2014). OX40 engagement depletes intratumoral Tregs via activating
FcgRs, leading to antitumor efficacy. Immunol. Cell Biol. 92, 475–480.
Clynes, R.A., Towers, T.L., Presta, L.G., and Ravetch, J.V. (2000). Inhibitory
Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat. Med.
6, 443–446.
Coe, D., Begom, S., Addey, C., White, M., Dyson, J., and Chai, J.G. (2010).
Depletion of regulatory T cells by anti-GITR mAb as a novel mechanism for
cancer immunotherapy. Cancer Immunol. Immunother. 59, 1367–1377.
Dahan, R., Sega, E., Engelhardt, J., Selby, M., Korman, A.J., and Ravetch, J.V.
(2015). FcgRs modulate the anti-tumor activity of antibodies targeting the
PD-1/PD-L1 axis. Cancer Cell 28, 285–295.
Dannull, J., Su, Z., Rizzieri, D., Yang, B.K., Coleman, D., Yancey, D., Zhang, A.,
Dahm, P., Chao, N., Gilboa, E., and Vieweg, J. (2005). Enhancement of vac-
cine-mediated antitumor immunity in cancer patients after depletion of regula-
tory T cells. J. Clin. Invest. 115, 3623–3633.
Elpek, K.G., Lacelle, C., Singh, N.P., Yolcu, E.S., and Shirwan, H. (2007).
CD4+CD25+ T regulatory cells dominate multiple immune evasion mecha-
nisms in early but not late phases of tumor development in a B cell lymphoma
model. J. Immunol. 178, 6840–6848.
Goding, S.R., Wilson, K.A., Xie, Y., Harris, K.M., Baxi, A., Akpinarli, A., Fulton,
A., Tamada, K., Strome, S.E., and Antony, P.A. (2013). Restoring immune
function of tumor-specific CD4+ T cells during recurrence of melanoma.
J. Immunol. 190, 4899–4909.
Golgher, D., Jones, E., Powrie, F., Elliott, T., and Gallimore, A. (2002). Depletion
of CD25+ regulatory cells uncovers immune responses to shared murine tumor
rejection antigens. Eur. J. Immunol. 32, 3267–3275.
Hodi, F.S., Butler, M., Oble, D.A., Seiden, M.V., Haluska, F.G., Kruse, A.,
Macrae, S., Nelson, M., Canning, C., Lowy, I., et al. (2008). Immunologic and
clinical effects of antibody blockade of cytotoxic T lymphocyte-associa ted an-
tigen 4 in previously vaccinated cancer patients. Proc. Natl. Acad. Sci. USA
105, 3005–3010.
Jacobs, J.F.M., Punt, C.J.A., Lesterhuis, W.J., Sutmuller, R.P.M., Brouwer,
H.M.-L.H., Scharenborg, N.M., Klasen, I.S., Hilbrands, L.B., Figdor, C.G., de
Vries, I.J.M., and Adema, G.J. (2010). Dendritic cell vaccination in combination
with anti-CD25 monoclonal antibody treatment: a phase I/II study in metastatic
melanoma patients. Clin. Cancer Res. 16, 5067–5078.
Jones, E., Dahm-Vicker, M., Simon, A.K., Green, A., Powrie, F., Cerundolo, V.,
and Gallimore, A. (2002). Depletion of CD25+ regulatory cells results in sup-
pression of melanoma growth and induction of autoreactivity in mice.
Cancer Immun. 2,1.
Larkin, J., Chiarion-Sileni, V., Gonzalez, R., Grob, J.J., Cowey, C.L., Lao, C.D.,
Schadendorf, D., Dummer, R., Smylie, M., Rutkowski, P., et al. (2015).
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma.
N. Engl. J. Med. 373, 23–34.
Luke, J.J., Zha, Y., Matijevich, K., and Gajewski, T.F. (2016). Single dose deni-
leukin diftitox does not enhance vaccine-induced T cell responses or effec-
tively deplete Tregs in advanced melanoma: immune monitoring and clinical
results of a randomized phase II trial. J. Immunother. Cancer 4,35.
Nimmerjahn, F., and Ravetch, J.V. (2005). Divergent immunoglobulin g sub-
class activity through selective Fc receptor binding. Science 310, 1510–1512.
Onizuka, S., Tawara, I., Shimizu, J., Sakaguchi, S., Fujita, T., and Nakayama, E.
(1999). Tumor rejection by in vivo administration of anti-CD25 (interleukin-2
receptor alpha) monoclonal antibody. Cancer Res. 59, 3128–3133.
Peggs, K.S., Quezada, S.A., Chambers, C.A., Korman, A.J., and Allison, J.P.
(2009). Blockade of CTLA-4 on both effector and regulatory T cell compart-
ments contributes to the antitumor activity of anti-CTLA-4 antibodies.
J. Exp. Med. 206, 1717–1725.
Quezada, S.A., Peggs, K.S., Curran, M.A., and Allison, J.P. (2006). CTLA4
blockade and GM-CSF combination immunotherapy alters the intratumor
balance of effector and regulatory T cells. J. Clin. Invest. 116, 1935–1945.
Quezada, S.A., Peggs, K.S., Simpson, T.R., Shen, Y., Littman, D.R., and
Allison, J.P. (2008). Limited tumor infiltration by activated T effector cells re-
stricts the therapeutic activity of regulatory T cell depletion against established
melanoma. J. Exp. Med. 205, 2125–2138.
Rech, A.J., Mick, R., Martin, S., Recio, A., Aqui, N.A., Powell, D.J., Jr., Colligon,
T.A., Trosko, J.A., Leinbach, L.I., Pletcher, C.H., et al. (2012). CD25 blockade
depletes and selectively reprograms regulatory T cells in concert with immuno-
therapy in cancer patients. Sci. Transl. Med. 4, 134ra62.
Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M., and Toda, M. (1995).
Immunologic self-tolerance maintained by activated T cells expressing IL-2
receptor alpha-chains (CD25). Breakdown of a single mechanism of self-toler-
ance causes various autoimmune diseases. J. Immunol. 155, 1151–1164.
Selby, M.J., Engelhardt, J.J., Quigley, M., Henning, K.A., Chen, T., Srinivasan,
M., and Korman, A.J. (2013). Anti-CTLA-4 antibodies of IgG2a isotype
enhance antitumor activity through reduction of intratumoral regulatory
T cells. Cancer Immunol. Res. 1, 32–42.
Setiady, Y.Y., Coccia, J.A., and Park, P.U. (2010). In vivo depletion of
CD4+FOXP3+ Treg cells by the PC61 anti-CD25 monoclonal antibody is medi-
ated by FcgammaRIII+ phagocytes. Eur. J. Immunol. 40, 780–786.
Immunity 46, 577–586, April 18, 2017 585
Shang, B., Liu, Y., Jiang, S.J., and Liu, Y. (2015). Prognostic value of tumor-
infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and
meta-analysis. Sci. Rep. 5, 15179.
Shimizu, J., Yamazaki, S., and Sakaguchi, S. (1999). Induction of tumor
immunity by removing CD25+CD4+ T cells: a common basis between tumor
immunity and autoimmunity. J. Immunol. 163, 5211–5218.
Simpson, T.R., Li, F., Montalvo-Ortiz, W., Sepulveda, M.A., Bergerhoff, K.,
Arce, F., Roddie, C., Henry, J.Y., Yagita, H., Wolchok, J.D., et al. (2013).
Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the
efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 210,
1695–1710.
Sutmuller, R.P., van Duivenvoorde, L.M., van Elsas, A., Schumacher, T.N.,
Wildenberg, M.E., Allison, J.P., Toes, R.E., Offringa, R., and Melief, C.J.
(2001). Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade
and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alter-
native pathways for suppression of autoreactive cytotoxic T lymphocyte re-
sponses. J. Exp. Med. 194, 823–832.
Topalian, S.L., Drake, C.G., and Pardoll, D.M. (2015). Immune checkpoint
blockade: a common denominator approach to cancer therapy. Cancer Cell
27, 450–461.
van Elsas, A., Sutmuller, R.P., Hurwitz, A.A., Ziskin, J., Villasenor, J., Medema,
J.P., Overwijk, W.W., Restifo, N.P., Melief, C.J., Offringa, R., and Allison, J.P.
(2001). Elucidating the autoimmune and antitumor effector mechanisms of a
treatment based on cytotoxic T lymphocyte antigen-4 blockade in combina-
tion with a B16 melanoma vaccine: comparison of prophylaxis and therapy.
J. Exp. Med. 194, 481–489.
586 Immunity 46, 577–586, April 18, 2017
... These cells secrete various inhibitory cytokines and growth factors, engage in metabolic competition, and express IC ligands themselves preventing the effective use of CAR-T cell therapy and ICIs [30]. By depleting these immune suppressive cell types, researchers have been able to enhance the anti-tumour immune response along with the infiltration, and efficiency of CAR-T therapies and ICIs preventing further T-cell exhaustion [30][31][32]. ...
Article
Full-text available
Introduction: Chimeric antigen receptor (CAR)-T cell therapy represents a breakthrough in cancer treatment by harnessing the power of genetically engineered T cells to target and eliminate tumour cells. This approach involves modifying a patient’s T cells to express CARs that recognize and bind to specific cancer antigens, bypassing conventional antigen presentation mechanisms. While CAR-T therapy has achieved notable success in treating ‘blood,’ or ‘liquid’ cancers, its application to solid tumours remains problematic due to the immunosuppressive tumour microenvironment (TME), which impairs T cell function and promotes tumour escape. Methods: A comprehensive review was conducted using PubMed and Google Scholar, focusing on publications from March 2002 to March 2024. The search criteria included terms related to CAR-T therapy, solid tumours and TME modulation. Studies were selected based on their relevance to CAR-T therapy challenges, advancements in TME modulation and clinical applications. Research focusing exclusively on liquid tumours was excluded to ensure the review’s focus on solid tumour contexts. Results: Analysis of lymphocyte densities revealed that hot tumours, characterized by high densities of CD3+ and CD8+ lymphocytes at both the tumour periphery and core, demonstrated the most favorable prognostic outcomes. Checkpoint blockades targeting CTLA-4 and PD-1/PD-L1 have been shown to prevent tumour escape. Additionally, engineering CAR-T cells to express immune checkpoint (IC) inhibitors counteracts the expression of IC ligands by immunosuppressive cells. T cells engineered to express IL-12 or IL-18 cytokines also allowed for better infiltration into the TME. Discussion: Targeting inhibitory immune checkpoints and suppressive cells, as well as manipulating CAR-T cytokine expression, have all shown to be promising ways in which the TME can be modulated to improve patient outcomes. Future research may include looking into the development of strategies to ensure long-term immunological memory in CAR-T cells, particularly for chronic or recurring solid tumours. Conclusion: Modulating the TME through various molecular and cellular avenues is crucial in improving the effectiveness of CAR-T cell therapy and therefore, optimizing antitumour immunity.
... Since Tregs suppress cytotoxic CD8+ T cells, they play pivotal roles in restraining anti-tumour immune responses and are a target of immunotherapies themselves (Dannull et al., 2005;Vargas et al., 2017). ...
Preprint
Full-text available
Immunotherapy has revolutionised cancer treatment, yet few patients respond clinically, necessitating alternative strategies that can benefit these patients. Novel immune-oncology targets can achieve this through bypassing resistance mechanisms to standard therapies. To address this, we introduce MIDAS, a multimodal graph neural network system for immune-oncology target discovery that leverages gene interactions, multi-omic patient profiles, immune cell biology, antigen processing, disease associations, and phenotypic consequences of genetic perturbations. MIDAS generalises to time-sliced data, outcompetes existing methods, including OpenTargets, and distinguishes approved from prospective targets. Moreover, MIDAS recovers immunotherapy response-associated genes in unseen trials, thus capturing tumour-immune dynamics within human tumours. Interpretability analyses reveal a reliance on autoimmunity, regulatory networks, and relevant biological pathways. Functionally perturbing the OSM-OSMR axis, a proposed target, in TRACERx melanoma patient-derived explants yielded reduced dysfunctional CD8 ⁺ T cells, which associate with immunotherapy response. Our results present a machine learning framework for analysing multimodal data for immune-oncology discovery.
... The FCGR2B gene encodes the low-a nity immunoglobulin gamma Fc receptor for the Fc region of the IgG1 complex [30]. This receptor is a protein on the cell membrane [31] that recognizes and binds to IgG antibodies. Consequently, it is involved in the phagocytosis of immune complexes and in the regulation of B-cell antibody production. ...
Preprint
Full-text available
Purpose Endometriosis (EMT) affects millions of women worldwide and is closely associated with the body's response to oxidative stress. The role of oxidative stress markers in the diagnosis and treatment of endometriosis is a potentially fruitful avenue of research. Methods In this study, we employed a machine learning approach to model and screen key biomarkers, integrating five independent datasets from the Omnibus (GEO) database to construct a comprehensive training set. The identification of key genes was achieved through a process of cross-referencing with the aim of locating those that were differentially expressed and known to be involved in oxidative stress. Nine machine learning algorithms were employed for model selection, followed by the evaluation of immune infiltration and immune correlation through single sample gene set enrichment analysis (ssGSEA) and the CIBERSORT algorithm. Results After comparing the performance of different machine learning algorithms, the gradient boosting algorithm (GBM) was selected as the best model. Eventually it screened five featured genes (FOS, CFH, AOX1, FMO1, FCGR2B). The expression patterns of these genes showed diagnostic and predictive potential in the constructed nomograms and external validation. In addition, the association of these genes with pregnancy status and disease severity was explored. The results of immune infiltration analysis showed significant correlation between these key genes and the immune system. Conclusions This study identifies genes at the intersection of endometriosis and oxidative stress, thereby providing reliable molecular markers for clinical application. This offers a new avenue for subsequent diagnosis and treatment of endometriosis.
... More specifically, PD-1 expression is upregulated upon antigen recognition (Simon and Labarriere, 2017), indicating that certain T cells in the tumor microenvironment are actively engaged as tumor-specific T cells. Beyond CD8+ T cells, other immune cell types, such as myeloid-derived suppressor cells or regulatory T cells, which regulate tumor-specific T cell immunity, may also influence therapeutic outcomes (Arce Vargas et al., 2017;Krieg et al., 2018;Kumagai et al., 2020). Overall, the multicellular regulation of the tumor-immune microenvironment emphasizes the importance of profiling systemic tumor and immune cells at singlecell resolution to investigate factors associated with responses to ICI treatment. ...
Article
Full-text available
This study investigates the variability among patients with non-small cell lung cancer (NSCLC) in their responses to immune checkpoint inhibitors (ICIs). Recognizing that patients with advanced-stage NSCLC rarely qualify for surgical interventions, it becomes crucial to identify biomarkers that influence responses to ICI therapy. We conducted an analysis of single-cell transcriptomes from 33 lung cancer biopsy samples, with a particular focus on 14 core samples taken before the initiation of palliative ICI treatment. Our objective was to link tumor and immune cell profiles with patient responses to ICI. We discovered that ICI non-responders exhibited a higher presence of CD4+ regulatory T cells, resident memory T cells, and TH17 cells. This contrasts with the diverse activated CD8+ T cells found in responders. Furthermore, tumor cells in non-responders frequently showed heightened transcriptional activity in the NF-kB and STAT3 pathways, suggesting a potential inherent resistance to ICI therapy. Through the integration of immune cell profiles and tumor molecular signatures, we achieved an discriminative power (area under the curve [AUC]) exceeding 95% in identifying patient responses to ICI treatment. These results underscore the crucial importance of the interplay between tumor and immune microenvironment, including within metastatic sites, in affecting the effectiveness of ICIs in NSCLC.
... RG6292, derived from 7D4, selectively depletes Tregs via antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) while sparing CD4 + and CD8 + Teffs [69]. Moreover, studies in mouse models demonstrated that combining optimized anti-CD25 antibodies with PD-1 inhibitors significantly amplifies anti-tumor effects and improves ICB efficacy [70,71]. h7B7-15 S, a humanized anti-CD25 monoclonal antibody that does not block IL-2, further enhances Treg depletion when paired with anti-CTLA-4, leading to substantial improvements in the remodeling of the tumor immune microenvironment [72]. ...
Article
Full-text available
Regulatory T cells (Tregs), an essential component of the human immune system, are a heterogeneous group of T lymphocytes with the ability to suppress immune responses and maintain immune homeostasis. Recent evidence indicates that Tregs may impair antitumor immunity and facilitate cancer progression by weakening functions of effector T cells (Teffs). Consequently, targeting Tregs to eliminate them from tumor microenvironments to improve Teffs’ activity could emerge as an effective strategy for cancer immunotherapy. This review outlines the biology of Tregs, detailing their origins, classification, and crucial markers. Our focus lies on the complex role of Tregs in cancer’s development, progression and treatment, particularly on their suppressive role upon antitumor responses via multiple mechanisms. We delve into Tregs’ involvement in immune checkpoint blockade (ICB) therapy, their dual effect on cancer immunotherapy and their potential biomarkers for ICB therapy effectiveness. We also summarize advances in the therapies that adjust Tregs to optimize ICB therapy, which may be crucial for devising innovative cancer treatment strategies.
Article
Full-text available
Single-cell RNA sequencing (scRNAseq) of tumour-infiltrating immune cells in high-grade serous ovarian cancer (HGSOC) omental biopsies reveals potential targets that could enhance response to neo-adjuvant chemotherapy (NACT). Analysis of 64,097 cells identifies NACT-induced overexpression of stabilin-1 (clever-1) on macrophages and FOXP3 in Tregs that is confirmed at the protein level. STAB1 inhibition in vitro induces anti-tumour macrophages. FOXP3 anti-sense oligonucleotide (FOXP3-ASO), repolarises Tregs to an effector T cell phenotype. ScRNAseq on 69,781 cells from an HGSOC syngeneic mouse model recapitulates the patients’ data. Combining chemotherapy with anti-stabilin1 antibody and/or Foxp3-ASO significantly increases survival of mice with established peritoneal disease in two HGSOC syngeneic models and progression-free survival in a third model. Long-term survivors (300 days + ) are resistant to tumour rechallenge. Anti-stabilin1 antibody enriches the tumours with CXCL9+ macrophages and Foxp3-ASO increases TBET cell infiltration. Our results suggest that targeting these molecules in immune cells may improve chemotherapy response in patients.
Article
Kirsten rat sarcoma virus (KRAS)–G12C inhibition causes remodeling of the lung tumor immune microenvironment and synergistic responses to anti–PD-1 treatment, but only in T cell infiltrated tumors. To investigate mechanisms that restrain combination immunotherapy sensitivity in immune-excluded tumors, we used imaging mass cytometry to explore cellular distribution in an immune-evasive KRAS mutant lung cancer model. Cellular spatial pattern characterization revealed a community where CD4 ⁺ and CD8 ⁺ T cells and dendritic cells were gathered, suggesting localized T cell activation. KRAS-G12C inhibition led to increased PD-1 expression, proliferation, and cytotoxicity of CD8 ⁺ T cells, and CXCL9 expression by dendritic cells, indicating an effector response. However, suppressive regulatory T cells (T regs ) were also found in frequent contact with effector T cells within this community. Lung adenocarcinoma clinical samples showed similar communities. Depleting T regs led to enhanced tumor control in combination with anti–PD-1 and KRAS-G12C inhibitor. Combining T reg depletion with KRAS inhibition shows therapeutic potential for increasing antitumoral immune responses.
Article
Immunology-based therapies are emerging as an effective cancer treatment, using the body’s immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Article
Full-text available
PROteolysis TArgeting Chimeras (PROTACs) have been considered the next blockbuster therapies. However, due to their inherent limitations, the efficacy of PROTACs is frequently impaired by limited tissue penetration and particularly insufficient cellular internalization into their action sites. Herein, based on the ultra-pH-sensitive and enzyme-sensitive nanotechnology, a type of polymer PROTAC conjugated and pH/cathepsin B sequential responsive nanoparticles (PSRNs) are deliberately designed, following the construction of the PROTAC for Cyclin-dependent kinase 4 and 6 (CDK4/6). Colorectal cancer (CRC) which hardly responds to many treatments even immune checkpoint blockades was selected as the tumor model in this study. As a result, PSRNs were found to maintain nanostructure (40 nm) in circulation and efficiently accumulated in tumors via enhanced permeation and retention effect. Then, they were dissociated into unimers (<10 nm) in response to an acidic tumor microenvironment, facilitating tumor penetration and cellular internalization. Eventually, the CDK4/6 degrading PROTACs were released intracellularly following the cleavage of cathepsin B. Importantly, PSRNs led to the enhanced degradation of target protein in vitro and in vivo. The degradation of CDK4/6 also augmented the efficacy of immune checkpoint blockades, through the upregulation of programmed cell death-ligand 1 (PD-L1) expression in cancer cells and the suppression of regulatory T cells cell proliferation in tumor microenvironment. By combination with α-PD-1, an enhanced anti-tumor outcome is well achieved in CT26 tumor model. Overall, our study verifies the significance of precise intracellular delivery of PROTACs and introduces a promising therapeutic strategy for the targeted combination treatment of CRC.
Article
Full-text available
Background Depletion of CD25⁺ Tregs improves anti-tumor immunity in preclinical models. Denileukin diftitox is a recombinant fusion protein of human IL-2 and diptheria toxin fragment that also can kill CD25⁺ T cells. Prior clinical trials of denileukin diftitox suggested reduction of FoxP3⁺ Tregs and some clinical responses. Method To investigate the immunologic effects of denileukin difitox on vaccine-specific immune responses in melanoma, a randomized clinical trial of single dose denileukin diftitox prior to vaccination versus vaccination alone in subjects with HLA-A2⁺ metastatic melanoma was performed. Treatment included randomization to a 4-peptide vaccine (Melan-A, gp100, MAGE3 and NA17 with GM-CSF emulsified in Montanide) alone or after single dose of denileukin diftitox (18 mcg/kg). Vaccine was given every 2 weeks for 3 doses and, absent clinical progression, continued every 2 weeks. Blood and tumor biopsies were obtained pretreatment and after 3 vaccinations for immunologic assessments. Results In 17 treated subjects there were no drug-related G3-4 adverse events. One partial response and 8 stable disease were observed in 9 subjects (4 DD: 5 vaccine only) with no impact of denileukin diftitox on time to progression. Total peripheral Tregs were not significantly altered, and in 1 patient biopsy intra-tumoral FoxP3 transcripts were not reduced following denileukin diftitox. ELISA for IL2R-α demonstrated no impact on outcomes by soluble CD25 level. Immune monitoring suggested the development of modest vaccine-specific CD8⁺ T cell responses in the control group, however immunization efficacy was actually reduced in the denileukin diftitox group. Conclusion Our results indicate that denileukin diftitox did not effectively deplete Tregs, augment T cell responses, or improve clinical activity in melanoma. Clinicaltrials.gov ID: NCT00515528; Registered August 9, 2007.
Article
Full-text available
The prognostic value of FoxP3+ regulatory T cells (Tregs) in cancer remains controversial. We did a meta-analysis to assess the prognostic effect of FoxP3+ Treg across different types of cancer and to investigate factors associated with variations in this effect. PubMed, Embase, Cochrane CENTRAL, and Scopus were searched to identify eligible studies. In total, we analyzed 76 articles encompassing 17 types of cancer, and including 15,512 cancer cases. The overall pooled analysis including all types of cancer suggested FoxP3+ Tregs had a significant negative effect on overall survival (OS) (OR 1.46, P < 0.001), but the prognostic effect varied greatly according to tumor site. High FoxP3+ Tregs infiltration was significantly associated with shorter OS in the majority of solid tumors studied, including cervical, renal, melanomas, and breast cancers, et al; whereas, FoxP3+ Tregs were associated with improved survival in colorectal, head and neck, and oesophageal cancers. The stratified analysis suggested the molecular subtype and tumor stage significantly influenced the prognostic value of FoxP3+ Tregs in certain types of cancer. In conclusion, our meta-analysis suggests that the prognostic role of FoxP3+ Tregs was highly influenced by tumor site, and was also correlated with the molecular subtype and tumor stage.
Article
Full-text available
Antitumor activity of CTLA-4 antibody blockade is thought to be mediated by interfering with the negative regulation of T-effector cell (Teff) function resulting from CTLA-4 engagement by B7-ligands. In addition, a role for CTLA-4 on regulatory T cells (Treg), wherein CTLA-4 loss or inhibition results in reduced Treg function, may also contribute to antitumor responses by anti-CTLA-4 treatment. We have examined the role of the immunoglobulin constant region on the antitumor activity of anti-CTLA-4 to analyze in greater detail the mechanism of action of anti-CTLA-4 antibodies. Anti-CTLA-4 antibody containing the murine immunoglobulin G (IgG)2a constant region exhibits enhanced antitumor activity in subcutaneous established MC38 and CT26 colon adenocarcinoma tumor models compared with anti-CTLA-4 containing the IgG2b constant region. Interestingly, anti-CTLA-4 antibodies containing mouse IgG1 or a mutated mouse IgG1-D265A, which eliminates binding to all Fcγ receptors (FcγR), do not show antitumor activity in these models. Assessment of Teff and Treg populations at the tumor and in the periphery showed that anti-CTLA-4-IgG2a mediated a rapid and dramatic reduction of Tregs at the tumor site, whereas treatment with each of the isotypes expanded Tregs in the periphery. Expansion of CD8(+) Teffs is observed with both the IgG2a and IgG2b anti-CTLA-4 isotypes, resulting in a superior Teff to Treg ratio for the IgG2a isotype. These data suggest that anti-CTLA-4 promotes antitumor activity by a selective reduction of intratumoral Tregs along with concomitant activation of Teffs. Cancer Immunol Res; 1(1); 32-42. ©2013 AACR.
Article
Full-text available
Antibodies targeting checkpoint inhibitors or co-stimulatory receptors on T cells have shown significant antitumor efficacy in preclinical and clinical studies. In mouse tumor models, engagement of activating Fcγ receptor (FcγR)-expressing immune cells was recently shown to be required for the tumoricidal activity of antibodies recognizing the tumor necrosis factor superfamily receptor (TNFR) GITR (CD357) and CTLA-4 (CD152). In particular, activating FcγRs facilitated the selective elimination of intratumoral T-cell populations. However, it remains unclear whether FcγRs contribute to the antitumor efficacy of other immunomodulatory antibodies. Here, we explored the mechanism of antitumor activity mediated by an agonistic antibody (clone OX86) to the co-stimulatory TNFR OX40 (CD134). OX40 was highly expressed by intratumoral T cells, particularly those of the FoxP3(+) regulatory T-cell (Treg) lineage. OX86 administration resulted in the depletion of intratumoral regulatory T cells in an activating FcγR-dependent manner, which correlated with tumor regression. Together with previous data from our group and others, these findings support a mechanism whereby antibodies targeting antigens highly expressed by intratumoral T cells can mediate their elimination by FcγR-expressing immune cells, and facilitate subsequent antitumor immunity.Immunology and Cell Biology advance online publication, 15 April 2014; doi:10.1038/icb.2014.26.
Article
Full-text available
Rational combinatorial therapeutic strategies have proven beneficial for the management of cancer. Recent success of checkpoint blockade in highly immunogenic tumors has renewed interest in immunotherapy. Regulatory T (T reg) cells densely populate solid tumors, which may promote progression through suppressing anti-tumor immune responses. We investigated the role of T reg cells in murine mammary carcinogenesis using an orthotopic, polyoma middle-T antigen-driven model in Foxp3(DTR) knockin mice. T reg cell ablation resulted in significant determent of primary and metastatic tumor progression. Importantly, short-term ablation of T reg cells in advanced spontaneous tumors led to extensive apoptotic tumor cell death. This anti-tumor activity was dependent on IFN-γ and CD4(+) T cells but not on NK or CD8(+) T cells. Combination of T reg cell ablation with CTLA-4 or PD-1/PD-L1 blockade did not affect tumor growth or improve the therapeutic effect attained by T reg cell ablation alone. However, T reg cell targeting jointly with tumor irradiation significantly reduced tumor burden and improved overall survival. Together, our results demonstrate a major tumor-promoting role of T reg cells in an autochthonous model of tumorigenesis, and they reveal the potential therapeutic value of combining transient T reg cell ablation with radiotherapy for the management of poorly immunogenic, aggressive malignancies.
Article
Although it is known that the immune system can mount responses to a variety of tumors it is clear that most tumors exhibit weak or even undetectable immunogenicity. Recent findings suggest that the lack of tumor immunogenicity is partly due to a population of cells called CD4⁺CD25⁺ regulatory T cells since depletion of these cells in mice can result in tumor rejection.These cells have also been shown to inhibit the development of organ-specific autoimmune diseases suggesting that they inhibit immune responses to tissue-specific self-antigens. Such immune responses may also mediate tumor rejection. Alternatively, immune responses in mice depleted of regulatory cells may target tumor antigens that are not tissue-specific, but which are shared by tumors of diverse origins. In experiments performed to discriminate between these possibilities we found, using the murine colorectal tumor CT26, that tumor immunity stimulated in the absence of regulatory cells is not restricted to tumors of colorectal origin, but is effective against tumors of different histological types such as B cell lymphomas and a renal cell carcinoma. By comparing this to CT26-induced immunity through the use of adjuvant we show that the generation of cross-reactive tumor immunity is a specific manifestation of CD25⁺ regulatory cell depletion. The generation of CD4⁺T cells capable of mediating tumor rejection is another important feature of tumor immunity induced in the absence of CD25⁺ cells.
Article
Immune checkpoint blockade of the programmed cell death protein 1 (PD-1) pathway by monoclonal antibodies (Abs) has shown promising clinical benefit in the treatment of multiple cancer types. We elucidated the contribution of the fragment crystallizable (Fc) domains of anti-PD-1 and anti-PD-ligand 1 (L1) Abs for their optimal anti-tumor activity. We revealed that distinct Fcγ receptor (FcγRs) dependency and mechanisms account for the in vivo activity of anti-PD-1 versus anti-PD-L1 Abs. Anti-PD-1 Abs were found to be FcγR independent in vivo; the presence of FcγR-binding capacity compromises their anti-tumor activity. In contrast, the anti-PD-L1 Abs show augmented anti-tumor activity when activating FcγR binding is introduced into the molecules, altering myeloid subsets within the tumor microenvironment.
Article
Approximately 10% of peripheral CD4+ cells and less than 1% of CD8+ cells in normal unimmunized adult mice express the IL-2 receptor a-chain (CD25++) molecules. When CD4+ cell suspensions prepared from BALB/c nu/+ mice lymph nodes and spleens were depleted of CD25++ cells by specific mAb and C, and then inoculated into BALB/c athymic nude (nu/nu) mice, all recipients spontaneously developed histologically and serologically evi dent autoimmune diseases (such as thyroiditis, gastritis, insulitis, sialoadenitis, adrenalitis, oophoritis, glomerulo nephritis, and polyarthritis); some mice also developed graft-vs-host-like wasting disease. Reconstitution of CD4+ cells within a limited period after transfer of CD4+ cells prevented these autoimmune de velopments in a dose-dependent fashion, whereas the reconstitution several days later, or inoculation of an equivalent dose of CD8+ cells, was far less efficient for the prevention. When nu/nu mice were transplanted with allogeneic skins or immunized with xenogeneic proteins at the time of CD25++ cell inoculation, they showed significantly heightened immune responses to the skins or proteins, and reconstitution of CD4+ cells normalized the responses. Taken together, these results indicate that CD4+ cells contribute to maintaining self-tolerance by down-regulating immune response to self and non-self Ags in an Ag-nonspecific manner, pre sumably at the T cell activation stage; elimination/reduction of CD4+ cells relieves this general suppres sion, thereby not only enhancing immune responses to non-self Ags, but also eliciting autoimmune responses to certain self-Ags. Abnormality of this T cell-mediated mechanism of peripheral tolerance can be a possible cause of various autoimmune diseases.
Article
Background: Nivolumab (a programmed death 1 [PD-1] checkpoint inhibitor) and ipilimumab (a cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4] checkpoint inhibitor) have been shown to have complementary activity in metastatic melanoma. In this randomized, double-blind, phase 3 study, nivolumab alone or nivolumab plus ipilimumab was compared with ipilimumab alone in patients with metastatic melanoma. Methods: We assigned, in a 1:1:1 ratio, 945 previously untreated patients with unresectable stage III or IV melanoma to nivolumab alone, nivolumab plus ipilimumab, or ipilimumab alone. Progression-free survival and overall survival were coprimary end points. Results regarding progression-free survival are presented here. Results: The median progression-free survival was 11.5 months (95% confidence interval [CI], 8.9 to 16.7) with nivolumab plus ipilimumab, as compared with 2.9 months (95% CI, 2.8 to 3.4) with ipilimumab (hazard ratio for death or disease progression, 0.42; 99.5% CI, 0.31 to 0.57; P<0.001), and 6.9 months (95% CI, 4.3 to 9.5) with nivolumab (hazard ratio for the comparison with ipilimumab, 0.57; 99.5% CI, 0.43 to 0.76; P<0.001). In patients with tumors positive for the PD-1 ligand (PD-L1), the median progression-free survival was 14.0 months in the nivolumab-plus-ipilimumab group and in the nivolumab group, but in patients with PD-L1-negative tumors, progression-free survival was longer with the combination therapy than with nivolumab alone (11.2 months [95% CI, 8.0 to not reached] vs. 5.3 months [95% CI, 2.8 to 7.1]). Treatment-related adverse events of grade 3 or 4 occurred in 16.3% of the patients in the nivolumab group, 55.0% of those in the nivolumab-plus-ipilimumab group, and 27.3% of those in the ipilimumab group. Conclusions: Among previously untreated patients with metastatic melanoma, nivolumab alone or combined with ipilimumab resulted in significantly longer progression-free survival than ipilimumab alone. In patients with PD-L1-negative tumors, the combination of PD-1 and CTLA-4 blockade was more effective than either agent alone. (Funded by Bristol-Myers Squibb; CheckMate 067 ClinicalTrials.gov number, NCT01844505.).
Article
The immune system recognizes and is poised to eliminate cancer but is held in check by inhibitory receptors and ligands. These immune checkpoint pathways, which normally maintain self-tolerance and limit collateral tissue damage during anti-microbial immune responses, can be co-opted by cancer to evade immune destruction. Drugs interrupting immune checkpoints, such as anti-CTLA-4, anti-PD-1, anti-PD-L1, and others in early development, can unleash anti-tumor immunity and mediate durable cancer regressions. The complex biology of immune checkpoint pathways still contains many mysteries, and the full activity spectrum of checkpoint-blocking drugs, used alone or in combination, is currently the subject of intense study. Copyright © 2015 Elsevier Inc. All rights reserved.