You are on page 1of 8

Seminars in Cancer Biology 65 (2020) 189–196

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

Tumor intrinsic and extrinsic immune functions of CD155 T


a b b b,
Jake. S. O’Donnell , Jason Madore , Xian-Yang Li , Mark J. Smyth *
a
Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, QLD, Australia
b
Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia

A R T I C LE I N FO A B S T R A C T

Keywords: CD155 (PVR/necl5/Tage4), a member of the nectin-like family of adhesion molecules, is highly upregulated on
Cancer tumor cells across multiple cancer types and has been associated with worse patient outcomes. In addition to
CD155 well described cell-intrinsic roles promoting tumor progression and metastasis, CD155 has now been implicated
PVR in immune regulation. The role of CD155 as a potent immune ligand with diverse cell-extrinsic functions is now
Immunotherapy
being defined. CD155 signaling to immune cells is mediated through interactions with the co-stimulatory im-
mune receptor CD226 (DNAM-1) and the inhibitory checkpoint receptors TIGIT and CD96, which are differ-
entially regulated at the cell surface on T cells and NK cells. The integration of signals from CD155 cognate
receptors modifies the activity of tumor-infiltrating lymphocytes in a context-dependent manner, making CD155
an attractive target for immune-oncology. Preclinical studies suggest that targeting this axis can improve im-
mune-mediated tumor control, particularly when combined with existing anti-PD-1 checkpoint therapies. In this
review, we discuss the roles of CD155 on host and tumor cells in controlling tumor progression and discuss the
possibility of targeting CD155 for cancer therapy.

1. Introduction lymphocytes via interactions with both stimulatory and inhibitory re-
ceptors on T and NK cells [16] (Fig. 1B). In this review we will discuss
The ability of tumors to escape or locally suppress the immune evidence that CD155 expression contributes to tumor development
system is fundamental for their development [1,2]. Cancer im- through tumor-associated immune suppression, how this immune sup-
munotherapies targeting the PD-1/PD-L1 or CTLA4 immune check- pression impacts responsiveness to cancer immunotherapy, and why
points function by relieving immune suppression or promoting immune targeting CD155 and its associated axis might be beneficial for cancer
activation, and have achieved durable efficacy for some patients across treatment.
multiple tumor indications [3–5]. While more effective than che-
motherapies, and with greater ability to induce long-term durable re- 2. Cell-intrinsic biology of CD155
sponses than those seen with targeted therapies (BRAF/MEK inhibi-
tion), existing cancer immunotherapies are nevertheless associated with The Nectins and Nectin-like molecules (Necl) are a family of Ig-like
innate and acquired resistance [3–6]. As such, the identification of al- proteins that play important roles in supporting cell to cell adhesion via
ternative approaches and targets that may improve responsiveness homophilic and heterophilic interactions between family members.
when used in combination with existing immunotherapies is a current Necl5, also known as CD155 was originally defined as a receptor to
priority. One attractive target is immune regulatory functions of the cell permit poliovirus entry [8,17], however, work by several groups ulti-
adhesion molecule CD155 (Poliovirus receptor [7]). CD155 is com- mately implicated CD155 in a suite of physiological/pathophysiological
monly over-expressed by tumor cells and upregulated by tumor-asso- processes [14]. The extracellular region of CD155 contains im-
ciated myeloid cells in both membrane bound and soluble forms [8,9] munoglobulin V domains, a C1-like domain, and a C2 domain [14].
where its tumor cell-intrinsic roles, which include promoting cell to cell There are four splice isoforms designated α, β, δ, and γ. The α and δ
adhesion [10], cell motility [10–13], contact inhibition [14], pro- isoforms contain transmembrane domains, with the α isoform con-
liferation, and survival [15] have implicated it in tumor progression. taining a longer C terminus and an immunoreceptor tyrosine-based
More recently, CD155 has been shown to play important tumor cell- inhibitory motif (ITIM) that is necessary for CD155-signaling and its
extrinsic roles by modulating the function of tumor-infiltrating tumor cell-intrinsic biology (Fig. 1A). By contrast, the β and γ isoforms


Corresponding author.
E-mail address: mark.smyth@qimrberghofer.edu.au (M.J. Smyth).

https://doi.org/10.1016/j.semcancer.2019.11.013
Received 26 September 2019; Received in revised form 6 November 2019; Accepted 19 November 2019
Available online 26 December 2019
1044-579X/ © 2019 Elsevier Ltd. All rights reserved.
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

Fig. 1. The tumor cell intrinsic and extrinsic roles of CD155.


(A) The intracellular domain of CD155 contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) capable of recruiting SH2-containing tyrosine phos-
phatase-2 to initiate signal transduction by acting as a mediator of the important cellular functions of CD155 including adhesion, contact inhibition, cellular motility,
proliferation, and survival. Many of these functions are achieved through simple interactions with other members of the immunoglobulin superfamily. For example,
CD155 can interact with growth factor receptors such as platelet-derived growth factor receptor (PDGFR) where it can enhance growth-factor mediated Ras-Raf-MEK-
ERK signaling, and also with integrin αvβ3 to enhance cellular migration. (B) CD155 expressed by tumor and tumor-associated myeloid cells can interact with at least
three lymphocyte-expressed receptors to modulate their effector functions. The inhibitory checkpoint receptors TIGIT and CD96 are upregulated by T and NK cells
following activation and increasingly when exposed chronically to antigen in settings such as cancer and chronic viral infection. These receptors and compete with
the costimulatory receptor DNAM-1 for binding to CD155 and function over time to diminish T and NK cell functionality in the tumor microenvironment. In NK cells,
interactions between α1β2 integrins (LFA-1) and DNAM-1 appear to be necessary for DNAM-1-mediated NK cell activation.

lack a transmembrane domain and are secreted, however, the biological knocked-out demonstrate defects in proliferation, cell cycle arrest [25].
function of these isoforms has not been clarified [18]. The CD155α
ITIM domain can recruit important signaling intermediates such as
SH2-containing tyrosine phosphatase-2 (SHP-2) to initiate signal- 3. Cell-extrinsic biology of CD155
transduction [11,19]. The initiation of signaling by CD155 is usually
preceded by its interaction with either with other nectins, growth factor The overexpression of CD155 seen in many tumor types was ori-
receptors, or integrins [15,20,21] (Fig. 1A). Collectively, these sig- ginally hypothesized to act as an extrinsic tumor-suppressor mechanism
naling inputs enable the cell-intrinsic roles of CD155 in promoting capable of alerting the immune system that a tissue had undergone
cellular proliferation, migration, contract inhibition, and survival; im- malignant transformation [8]. This hypothesis was born-out of the
plicating tumor cell-intrinsic CD155 in promoting tumor progression, observation that the co-stimulatory receptor CD226 or DNAX Accessory
invasion, and metastasis. Molecule-1 (DNAM-1) expressed by T and NK cells interacts directly
The signaling outputs of CD155 have been proposed to have mul- with CD155 and the closely related CD112 (Nectin-1) molecules to
tiple effects in the context of cancer. For instance, an in vitro screen promote their activation [26]; the latter of which can also bind
identified that knock-down of CD155 significantly reduced tumor cell CD112R, a checkpoint receptor expressed by T cells [27]. These inter-
migration [22,23]. In one study, CD155 was shown to play an essential actions were shown to trigger NK cell- and T cell-mediated cytotoxicity
role in growth factor-mediated cell migration. This effect was found to and cytokine production [28,29]. In addition, mice genetically deficient
depend on the co-localisation of CD155 αVβ3 integrins at the leading in DNAM-1 demonstrated significantly less anti-tumor activity against
edge of migrating cells and the indication of intracellular signaling via CD155 positive tumor cells than wild type mice; with a higher in-
Rac and Cdc42, which have well-defined roles in cell migration. In cidence of carcinogen-induced tumors, accelerated tumor growth rate,
addition to migration, CD155 has been shown to promote tumor me- and increased experimental metastasis [30]. Despite these observations,
tastatic spread [23]. Over-expression of CD155 in rat glioma cells was over-expression of CD155 by tumors has been associated with dimin-
shown to increase Src and focal adhesion kinase signaling, which in ished activity of tumor-infiltrating lymphocytes and worse prognosis in
turn reduced substrate adhesion and promoted cell spreading [24]. several cancer types [31–33] which argues, at least in these tumor
Subsequently, observations made using mouse tumor models support types, against the aforementioned hypothesis and suggests that the
this conclusion, where CD155-expressing tumor cells have been shown overall effect of CD155 in the TME is in fact immunosuppressive for
to have greater metastatic potential than tumor cells in which CD155 many solid tumor indications.
has been knocked out [9]. CD155 signaling also appears to be important In tumor tissue, the activities of T and NK cells are commonly
for tumor cell proliferation. Tumor cells in which CD155 has been suppressed by interactions between checkpoint receptors and check-
point ligands expressed in the TME. Among these are the T cell

190
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

Fig. 2. Upregulation of TIGIT and CD96 diminishes DNAM-1 stimulation of tumor-infiltrating lymphocytes. (A) In tumor associated stroma, activated CD8+ T cells
are likely to express DNAM-1 and comparatively lower levels of TIGIT and CD96 due to lower TCR signaling resulting in a net positive signal generated from CD155
expressed in the stroma. (B) As tumors emerge from tumor-associated stroma into tumor tissue (parenchyma), DNAM-1 can be down-regulated, and the expression of
PD-1, CD96, and likely TIGIT can increase resulting in a net negative signal to the T cell from tumor CD155. Their interactions with CD155 expressed by tumor and
tumor-associated myeloid cells (APCs) are associated with a dysfunctional T cell phenotype.

immunoreceptor with Ig and ITIM domains (TIGIT) and CD96 immune domain of TIGIT contains an intracellular immunoreceptor tyrosine-
checkpoint receptors which both compete with DNAM-1 to bind with based inhibitory motif that is responsible for mediating its inhibitory
CD155 (Fig. 1B) [34]. This competition functions to suppress DNAM-1- signaling [45]. Single cell RNAseq analysis of human cancers has re-
mediated T- and NK cell-activation. These interactions occur with dif- vealed co-expression of TIGIT among tumor-infiltrating CD8+ T cells
ferent affinities [35]. For example, DNAM-1 has the lowest affinity for expressing other checkpoint receptors such as TIM-3, LAG-3, CTLA-4,
binding CD155, followed by CD96 which has an intermediate affinity, and PD-1 [46,47]. This was also shown in a recent series of articles
and TIGIT which binds with high affinity to CD155 [35,36]. This is defining the transcriptional regulation of CD8+ T cell exhaustion in
likely due to the unique structural interactions that each of CD96, both chronic viral infection and cancer [48–50]. Importantly, these
TIGIT, and DNAM-1 have with CD155 [35,36]. The competitive inter- effects might be cell type-dependent as expression of TIGIT appears to
actions between CD96 and TIGIT with DNAM-1 are reminiscent of the be important for the suppressive activity of Tregs in the tumor micro-
process by which the CTLA4 checkpoint receptor, competes with the co- environment. Tregs in TIGIT−/− mice have reduced suppressive ac-
stimulatory receptor CD28 for binding to B7 molecules expressed on tivity, and in mice TIGIT+ Tregs have increased suppression of Th1 and
antigen presenting cells (APCs) (Fig. 1B) [34,37]. DNAM-1 contains an Th17 immune responses when compared to TIGIT- Tregs [51,52]. Im-
intracellular immunoreceptor tyrosine tail (ITT)-like motif, upon in- proved CD8+ T cell function has been seen following blockade of TIGIT
teractions with CD155 or CD112, this intracellular domain has been alone or in combination with other immunotherapies [53–55].
proposed to signal via the adaptor protein Grb2 [38]. As is the case for Similar to TIGIT, upregulation of CD96 expression on intratumor T
other co-stimulatory molecules including CD28, DNAM-1 down-reg- and NK cells has been shown in several solid cancer types. Recent
ulation has been seen on T cells in the setting of chronic infection studies primarily in mouse tumor models have indicated a role for CD96
[39,40], and on NK cells in advanced cancers and has been associated as a T and NK cell checkpoint receptor. Initial investigations suggested
with T cell exhaustion and diminished NK cell-mediated cytotoxicity that CD96 mediated human NK cell adhesion to CD155-expressing
[28,29,41,42]. These findings might suggest that loss of co-stimulation target cells [56]. The first evidence that CD96 might act in an inhibitory
is a key feature of lymphocyte dysfunction, however, the mechanism(s) capacity was shown in CD96−/− mice, where NK cells were found to
underlying this effect is poorly understood [43]. produce greater IFNγ in response to IL12, IL18, or LPS stimulation [57].
In contrast to DNAM-1 downregulation, TIGIT is highly upregulated It was also shown that these mice were resistant to experimental lung
on both CD8+ T cells and tumor-infiltrating regulatory T cells, and has metastases and carcinogen-induced tumor development [57]. More
also been reported on tumor-infiltrating NK cells [44]. The intracellular recently, CD96 has been shown to reduce the activity of CD8+ T cells,

191
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

Fig. 3. Expression distribution and mutation frequency of CD155 across human cancers.
(A) Expression of CD155 detected by RNAseq across 32 cancer types. Each dot represents a single case, the color of the data point corresponds to whether and what
type of gene alteration is associated with CD155 in each case as described in the legend. Error bars represent SD. (B) As a percentage of total cases, the proportion of
each tumor type that contain a mutation, fusion, amplification, deep deletion, or multiple alterations in the PVR gene. Data presented are publicly available from the
TCGA and interpreted using https://www.cbioportal.org/.

and promote their acquisition of a dysfunctional phenotype in mouse factors in the tumor microenvironment [58]. Despite these observa-
tumor models [57–59]. Expression of CD96 appears to be most common tions, further studies will need to establish whether the im-
among tumor-infiltrating CD8+ T cells and NK cells [44,58,60]. Unlike munosuppressive activity of CD96 seen in mice is conserved in humans.
TIGIT, expression of CD96 is much less common among CD4+ T cells in In summary, the effect that CD155 has on tumor immune suppression
mouse tumor models. In human colorectal carcinoma, expression of PD- will depend on the relative balance of stimulatory (DNAM-1) and in-
1 and CD96 appears to be selectively upregulated by CD8+ T cells in- hibitory (TIGIT and CD96) signals produced by these receptors in
filtrating tumor tissue, but not those in tumor-associated stroma, sug- tumor-infiltrating T and NK cells. In addition to DNAM-1, TIGIT, and
gesting that its expression is regulated by antigen exposure or other CD96, a recent study utilizing a novel method for ligand-receptor

192
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

proteomics identified that the NK cell receptor KIR2DL5 is also likely to several ongoing clinical trials investigating various treatments are at-
be a ligand for CD155, however, the significance of this interaction has tempting to measure CD155 expression in tumor tissue prior, or fol-
not yet been defined [61]. In tumors, an increase in expression of the lowing the initiation of treatment in advanced solid tumors (Clin-
inhibitory receptors (TIGIT and CD96) and reduced expression of co- Trials.gov: NCT03667716), ovarian and gastric cancers, (ClinTrials.gov:
stimulatory receptors (DNAM-1), is likely responsible for the observed NCT03342417), melanoma (ClinTrials.gov: NCT03712358), bladder
immunosuppressive properties of CD155 seen in solid tumors (Fig. 2A cancer (ClinTrials.gov: NCT03789682), and prostate cancer (Clin-
and B). Trials.gov: NCT03071328). Importantly, elevated soluble CD155 has
been associated with increased tumor burden, and has been proposed as
4. Expression and function of CD155 on host immune cells a potential biomarker of disease progression [18,75].
Several findings suggest that CD155 is associated with tumor pro-
Under physiological conditions, CD155 is widely expressed at low gression: First, genetic deletion of CD155 from tumor cells in mice re-
levels on immune cells where it has been proposed to play various roles sults in a significant delay in tumor growth and metastasis [9,76].
such as co-stimulation of CD4+ T cells [62] and MHC-independent Second, expression of CD155 in transplantable tumor models in mice
positive-selection of T cells in the thymus [63]. There is also evidence genetically deficient in CD155 was found to significantly enhance pri-
that CD155 is expressed on endothelial cells where it has been shown to mary tumor growth and metastasis [9,76]. While overexpression of
attenuate CD8+ T cell responses [64]. Recent evidence indicates that CD155 might support tumor progression due to its cell-intrinsic effects,
CD155-expression can be modulated on tumor-infiltrating leukocytes recent evidence suggests tumor cell expression of CD155 meaningfully
and that this might function in suppressing the activity of anti-tumor suppresses anti-tumor immune responses. Relating to the latter ob-
immune responses [9]. To date the most compelling evidence sup- servation, using several retrospective cohorts in melanoma, a strong
porting its role in immune suppression came from studies in gene- and significant association between poor response to cancer im-
modified mice deficient in CD155 expression. In these mice significant munotherapy (either anti-PD-1 alone, or in combination with anti-
delays in primary tumor growth and experimental metastasis were seen CTLA4) was identified in melanoma [77]. This followed the demon-
in comparison to wild-type (CD155-expressing) mice [9]. Bone marrow stration that elevated CD155 expression associated with a diminished
chimera studies were used to confirm that CD155-expressing haema- IFNγ gene-expression signature, but not necessarily reduced CD8+ T
topoietic cells were responsible for suppression of T and NK cell- cell infiltration in pre-immunotherapy-treated tumor samples and this
mediated anti-tumor immune responses [9]. specifically highlighted CD155 and associated pathway members as key
CD155 expression on immune cells can be detected among tumor- regulators of CD8+ T cell function and response to cancer im-
infiltrating leukocytes in the primary tumor and tumor-draining lymph munotherapy [77]. Together, these studies suggest that CD155 over-
nodes, however, it appears to be mostly restricted to myeloid cells [9]. expression by tumor cells plays an important role in promoting tumor
In human melanoma samples, CD155 expression has been observed on progression both as a result of intrinsic oncogenic pathway activation,
CD14+CD11c− macrophages, CD14+CD11c+ myeloid cells, and and also of extrinsic suppression of anti-tumor immune responses. Im-
CD14−CD11c+ dendritic cells (DCs). Co-localization of CD155-ex- portantly, it is not yet understood whether soluble CD155 contributes to
pressing myeloid cells with an M2-like, immunosuppressive phenotype immune suppression in the tumor microenvironment, however, if the
with T cells in these tumors suggested a potentially immunosuppressive soluble CD155 isoforms can bind CD96 and TIGIT, this might be a
role [9]. possibility.
The mechanism(s) responsible for upregulation of CD155 on tumor- The overexpression of CD155 observed in cancers likely occurs as a
infiltrating myeloid cells is currently unclear. One report in patients result of a variety of mechanisms. CD155 overexpression has been as-
with sepsis revealed that CD155 and the closely related CD112 (Nectin- sociated with the cellular stress responses to reactive oxygen species
1) molecules were selectively upregulated by circulating monocytes (ROS), and also constitutive expression of the MYC oncogene with in-
[65]. In a separate study, ex vivo exposure of mouse bone marrow-de- duction of the ATM-ATR DNA damage repair pathway in human tumor
rived macrophages and B cells to LPS was shown to promote upregu- cell lines [78–82]. In addition, a single study found that in vitro cultures
lation of CD155 in an NF-κB-dependent manner [66–68]. Given that of human melanoma cells upregulated CD155 when exposed to IFNγ
toll-like receptor (TLR) ligands are common in the blood of both septic similar to the adaptive upregulation of PD-L1, however this observation
patients and in the tumor microenvironment, this is a possible stimulus has not been replicated [66,83]. For PD-L1, its expression in tumors
for myeloid cell CD155 upregulation, however, this hypothesis requires tends to be limited to regions of immune cell infiltration [84,85], except
testing. for rare instances in which PD-L1 is constitutively expressed by tumor
cells [86]. By contrast, when expressed, CD155 staining tends to be
5. Expression of CD155 in human cancers homogenous throughout tumor tissue, independent of immune cell lo-
calization [87]. This observation lends itself to a genetic rather than an
While expression of CD155 by tumor-infiltrating myeloid cells has external immunological cause as seen with PD-L1 upregulation in the
been shown to play a significant role in suppressing the anti-tumor presence of IFNγ. One likely mechanism is amplification of the PVR
immune response [9], in the TME the primary source of CD155 is gene itself which appears to be most common in uterine carcinoma (∼7
probably tumor cells [8]. Overexpression of CD155 by tumor cells was % of cases) and associates with its relatively high expression of CD155
first seen in the early 1990s by researchers attempting to develop (Fig. 3A and B). However, in tumors which show low CD155 expression
therapeutic monoclonal antibodies against proteins overexpressed by and do not show evidence for amplification or copy number gain such
tumor cells, however, at the time it was not recognized that the tumor as AML, DLBC, and Thymoma, selection against CD155 overexpression
antigen targeted was in fact CD155 [8,69]. Overexpression of CD155 may be driven by DNAM-1 expression on tumor infiltrating lympho-
has since been reported in many cancers including melanoma [70], cytes; however, for this to occur stimulatory signaling via DNAM-1
lung adenocarcinoma [71], pancreatic cancer [31], ovarian cancer would have to outweigh immunosuppressive signals from TIGIT and
[41], myeloid leukemia [28], neuroblastoma [28,69], colorectal cancer CD96. Nevertheless, these CD155 low cancers seem to be an exception;
[72], cholangiocarcinoma among others (Fig. 3A) [73]. This observa- the majority of tumors types show high levels of CD155 gene expression
tion has been associated with poor clinical measures including lymph (Fig. 3A and B). In addition to amplification of the PVR gene, mutations
node metastasis, a reduction in tumor-infiltrating lymphocytes, tumor in other genes that result in enhanced Ras-Raf-MEK-ERK activation,
histological grade, primary tumor size, and poor prognosis in retro- elevated fibroblast growth factor signaling [7,88], and also aberrant
spective studies across various solid cancer types [31,73,74]. To better activation of the hedgehog signaling pathway have been shown to re-
understand the relationship between CD155 and clinical correlates, sult in CD155 overexpression in vitro [89]. An understanding of the

193
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

mechanisms responsible for CD155 overexpression by tumor cells may experimental metastasis models including B16F10, 3 L L lung carci-
allow for the development of therapies to decrease its expression and noma, LWT1 melanoma, and RM-1 prostate carcinoma through an
enhance sensitivity to cancer immunotherapies. IFNγ- and NK cell-dependent mechanism [60]. More recently, blockade
of CD96 has also been shown to improve CD8 T cell-mediated anti-
6. Targeting the CD155 axis with therapies tumor responses in mice [58].
Blockade of TIGIT and CD96 together or in combination with other
The significant immune suppression induced by CD155 in addition conventional checkpoint inhibitors blocking PD-1 or CTLA4 appears to
to its very broad expression distribution in cancer highlights it potential be more effective than monotherapy treatment. Importantly, this has
as a therapeutic target. Supporting this idea, preclinical studies in mice been shown in models of experimental metastasis (in which tumor
genetically deficient in either CD155 or of TIGIT and CD96 have en- control is NK cell-mediated) [60,94], primary tumor growth models
hanced anti-tumor immunity [9,90]. As such, blocking this interaction (usually CD8+ T cell-mediated) [9,58], and also spontaneous metas-
with CD155 in the TME might be an effective therapy for tumor control. tasis studies following surgery to remove the primary tumor (NK cell
In experimental settings, the use of monoclonal antibodies against and T cell-mediated) [94]. However, whether the anti-tumor effect of
CD155 has been explored as a method for improving tumor control. In targeting CD96 and/or TIGIT partly or entirely depends on blocking
one study, experimental metastatic burden was reduced in im- CD96/CD155 versus TIGIT/CD155 interaction and whether redundancy
munocompetent mice following administration of anti-CD155 [91]. in exists between CD96 and TIGIT is still unclear [94]. These findings
vitro assays revealed that anti-CD155 treatment reduced tumor cell provide evidence to suggest that independent of inhibitory receptor
migration and invasion [91]. Additionally, CD155 blockade has also competition for DNAM-1, CD96 and TIGIT have intrinsic im-
been shown to improve the function of antigen presenting cells in ex- munosuppressive signaling effects. Mechanistic studies to investigate
perimental sepsis [92]. To date, clinical studies have not been carried the differential regulation and impact that CD155, DNAM-1, CD96, and
out to assess whether CD155 blockade is feasible in human cancers. One TIGIT have on the anti-tumor immune response are needed. An addi-
potential limitation of this approach could be that in addition to tional consideration for the therapeutic blockade of TIGIT and CD96 is
blocking the interactions of CD155 with TIGIT and CD96, CD155 that their upregulation has been associated in tumor-infiltrating T cells
blockade might impact stimulatory interactions between DNAM-1 and with terminal differentiation. In turn, this might suggest that blocking
CD155 (Fig. 1B). Selection of cancer indications for clinical trials tar- the interactions of TIGIT/CD96 with CD155 might do little to re-
geting CD155 should therefore be guided by CD155 expression levels on invigorate the anti-tumor immune response [48–50], however, the fact
tumor cells as well as TIGIT/CD96 and DNAM-1 expression levels on that therapeutic efficacy has been seen preclinically argues against this.
infiltrating lymphocytes. There are currently several clinical trials in various phases testing the
To avoid any issue inherent in blocking CD155-DNAM-1 interac- efficacy of anti-TIGIT monoclonal antibodies. To date, there are no
tions, targeting exclusively the inhibitory side of CD155 pathway (CD96 clinical trials assessing the efficacy of anti-CD96 therapy (Fig. 4).
and TIGIT) has already been shown to have a beneficial effect on tumor In addition to CD155 checkpoint blockade, another therapeutic
control (Fig. 4). Early studies suggested that TIGIT blockade enhances approach takes advantage of the overexpression of CD155 by tumors.
the in vitro killing of tumor cells by mouse PBMCs [93]. In addition to An oncolytic viral therapy, Poliovirus-Rhinovirus Chimera (PVSRIPO)
this, blockade of CD96 has demonstrated efficacy in treatment of which binds specifically to CD155 has been developed to promote

Fig. 4. Progression of therapies targeting the CD155 axis in cancer.


A description of the current phase of development that therapies targeting CD155 or its ligands, DNAM-1, TIGIT, and CD96.

194
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

immunogenic tumor cell death, inducing lysis of tumor cells and the [10] R. Lange, et al., The poliovirus receptor CD155 mediates cell-to-matrix contacts by
release of Danger-Associated Molecular Patterns (DAMPs) capable of specifically binding to vitronectin, Virology 285 (2) (2001) 218–227.
[11] T. Oda, S. Ohka, A. Nomoto, Ligand stimulation of CD155alpha inhibits cell ad-
inducing an anti-tumor immune responses involving T and NK cells hesion and enhances cell migration in fibroblasts, Biochem. Biophys. Res. Commun.
[95–98]. Also, CD155-expressing dendritic cells and macrophages when 319 (4) (2004) 1253–1264.
exposed to PVSRIPO where shown to engage T cells and limit viral [12] D.P. Sullivan, M.A. Seidman, W.A. Muller, Poliovirus receptor (CD155) regulates a
step in transendothelial migration between PECAM and CD99, Am. J. Pathol. 182
replication [96]. PVSRIPO is now undergoing clinical testing at various (3) (2013) 1031–1042.
phases (Fig. 4). [13] N. Reymond, et al., DNAM-1 and PVR regulate monocyte migration through en-
dothelial junctions, J. Exp. Med. 199 (10) (2004) 1331–1341.
[14] Y. Takai, et al., Nectins and nectin-like molecules: roles in contact inhibition of cell
7. Perspectives movement and proliferation, Nat. Rev. Mol. Cell Biol. 9 (8) (2008) 603–615.
[15] S. Kakunaga, et al., Enhancement of serum- and platelet-derived growth factor-in-
The findings discussed throughout this review have highlighted duced cell proliferation by Necl-5/Tage4/Poliovirus Receptor/CD155 through the
Ras-Raf-MEK-ERK signaling, J. Biol. Chem. 279 (35) (2004) 36419–36425.
some interesting questions that will allow for a more complete under-
[16] J.R. Bowers, et al., Poliovirus Receptor: more than a simple viral receptor, Virus
standing of the roles that CD155 plays in the tumor microenvironment Res. 242 (2017) 1–6.
and how best to target this axis. First, it will be important to more fully [17] C.L. Mendelsohn, E. Wimmer, V.R. Racaniello, Cellular receptor for poliovirus:
define the tumor cell-intrinsic and -extrinsic effects of CD155. Second, molecular cloning, nucleotide sequence, and expression of a new member of the
immunoglobulin superfamily, Cell 56 (5) (1989) 855–865.
we need to understand how CD155 expression is regulated by tumor [18] S. Koike, et al., The poliovirus receptor protein is produced both as membrane-
cells and by myeloid cells in the tumor microenvironment, as these bound and secreted forms, EMBO J. 9 (10) (1990) 3217–3224.
processes might be targetable and understanding will be required to [19] S.-I. Yusa, T.L. Catina, K.S. Campbell, SHP-1- and phosphotyrosine-independent
inhibitory signaling by a killer cell Ig-like receptor cytoplasmic domain in human
overcome potential issues with blocking CD155 expressed in healthy NK cells, J. Immunol. 168 (10) (2002) 5047–5057.
tissues. Third, it would be helpful to understand how TIGIT, CD96, and [20] M. Kinugasa, et al., Necl-5/poliovirus receptor interacts with VEGFR2 and regulates
DNAM-1 are differentially regulated and functionally interact in the VEGF-induced angiogenesis, Circ. Res. 110 (5) (2012) 716–726.
[21] S. Mueller, E. Wimmer, Recruitment of nectin-3 to cell-cell junctions through trans-
tumor microenvironment and whether this mechanism differs between heterophilic interaction with CD155, a vitronectin and poliovirus receptor that
different lymphocyte subsets and across different cancer types. Fourth, localizes to alpha(v)beta3 integrin-containing membrane microdomains, J. Biol.
it would be helpful to understand what effect soluble CD155 has on the Chem. 278 (33) (2003) 31251–31260.
[22] K.E. Sloan, et al., CD155/PVR plays a key role in cell motility during tumor cell
efficacy of cancer immunotherapy. Can soluble CD155 suppress lym- invasion and migration, BMC Cancer 4 (2004) 73.
phocyte function in the same way that membrane-bound CD155 does? [23] W. Ikeda, et al., Nectin-like molecule-5/Tage4 enhances cell migration in an in-
Finally, clinical trials to assess the feasibility and efficacy of blocking tegrin-dependent, Nectin-3-independent manner, J. Biol. Chem. 279 (17) (2004)
18015–18025.
CD155 interactions with TIGIT and CD96 are required. Are there par-
[24] K.E. Sloan, et al., CD155/PVR enhances glioma cell dispersal by regulating adhesion
ticular tumor types and therapeutic combinations for which greater signaling and focal adhesion dynamics, Cancer Res. 65 (23) (2005) 10930–10937.
efficacy will be seen? Answering these questions will help guide clinical [25] T. Kono, et al., The CD155/poliovirus receptor enhances the proliferation of ras-
trials and allow for the development of more effective therapies tar- mutated cells, Int. J. Cancer 122 (2) (2008) 317–324.
[26] A. Shibuya, et al., DNAM-1, a novel adhesion molecule involved in the cytolytic
geting the CD155 axis in cancer. function of T lymphocytes, Immunity 4 (6) (1996) 573–581.
[27] Y. Zhu, et al., Identification of CD112R as a novel checkpoint for human T cells, J.
Declaration of Competing Interest Exp. Med. 213 (2) (2016) 167–176.
[28] D. Pende, Analysis of the receptor-ligand interactions in the natural killer-mediated
lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the in-
M. J. Smyth has research agreements with Bristol Myers Squibb and volvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112), Blood 105 (5)
Tizona Therapeutics and is on the Scientific Advisory Board at Tizona (2005) 2066–2073.
[29] R. Castriconi, et al., Natural killer cell-mediated killing of freshly isolated neuro-
Therapeutics and Compass Therapeutics. X.Y. Li, J.S. O’Donnell, and J. blastoma cells: critical role of DNAX accessory molecule-1-poliovirus receptor in-
Madore declare no conflict of interest. teraction, Cancer Res. 64 (24) (2004) 9180–9184.
[30] A. Iguchi-Manaka, et al., Accelerated tumor growth in mice deficient in DNAM-1
receptor, J. Exp. Med. 205 (13) (2008) 2959–2964.
Acknowledgments
[31] S. Nishiwada, et al., Clinical significance of CD155 expression in human pancreatic
cancer, Anticancer Res. 35 (4) (2015) 2287–2297.
The project was funded by a National Health and Medical Research [32] J. Gao, et al., CD155, an onco-immunologic molecule in human tumors, Cancer Sci.
108 (10) (2017) 1934–1938.
Council of Australia (NH&MRC) Program Grant (1132519) and Project
[33] H. Triki, et al., CD155 expression in human breast cancer: clinical significance and
Grant (1098960). M. J. S. was supported by a Senior Principal Research relevance to natural killer cell infiltration, Life Sci. 231 (2019) 116543.
Fellowship (1078671). The authors thank Dr. Robert Johnston (Bristol [34] W.C. Dougall, et al., TIGIT and CD96: new checkpoint receptor targets for cancer
Myers Squibb) for critical reading of this manuscript. immunotherapy, Immunol. Rev. 276 (1) (2017) 112–120.
[35] X. Yu, et al., The surface protein TIGIT suppresses T cell activation by promoting the
generation of mature immunoregulatory dendritic cells, Nat. Immunol. 10 (1)
References (2009) 48–57.
[36] F.A. Deuss, et al., Structural basis for CD96 immune receptor recognition of nectin-
like Protein-5, CD155, Structure 27 (2) (2019) 219–228 e3.
[1] D.S. Thommen, T.N. Schumacher, T cell dysfunction in cancer, Cancer Cell 33 (4) [37] B. Husain, et al., A platform for extracellular interactome discovery identifies novel
(2018) 547–562. functional binding partners for the immune receptors B7-H3/CD276 and PVR/
[2] R.D. Schreiber, L.J. Old, M.J. Smyth, Cancer immunoediting: integrating im- CD155, Mol. Cell Proteomics (2019).
munity’s roles in cancer suppression and promotion, Science 331 (6024) (2011) [38] Z. Zhang, et al., DNAM-1 controls NK cell activation via an ITT-like motif, J. Exp.
1565–1570. Med. 212 (12) (2015) 2165–2182.
[3] A.M.M. Eggermont, et al., Adjuvant pembrolizumab versus placebo in resected [39] M. Cella, et al., Loss of DNAM-1 contributes to CD8+ T-cell exhaustion in chronic
stage III melanoma, N. Engl. J. Med. 378 (19) (2018) 1789–1801. HIV-1 infection, Eur. J. Immunol. 40 (4) (2010) 949–954.
[4] S.C. Wei, C.R. Duffy, J.P. Allison, Fundamental mechanisms of immune checkpoint [40] A.N. Vallejo, et al., Modulation of CD28 expression: distinct regulatory pathways
blockade therapy, Cancer Discov. 8 (9) (2018) 1069–1086. during activation and replicative senescence, J. Immunol. 162 (11) (1999)
[5] C. Robert, et al., Pembrolizumab versus ipilimumab in advanced melanoma, N. 6572–6579.
Engl. J. Med. 372 (26) (2015) 2521–2532. [41] M. Carlsten, et al., Primary human tumor cells expressing CD155 impair tumor
[6] N.P. Restifo, M.J. Smyth, A. Snyder, Acquired resistance to immunotherapy and targeting by down-regulating DNAM-1 on NK cells, J. Immunol. 183 (8) (2009)
future challenges, Nat. Rev. Cancer 16 (2) (2016) 121–126. 4921–4930.
[7] T. Hirota, et al., Transcriptional activation of the mouse Necl-5/Tage4/PVR/CD155 [42] B. Sanchez-Correa, et al., Decreased expression of DNAM-1 on NK cells from acute
gene by fibroblast growth factor or oncogenic Ras through the Raf–MEK–ERK–AP-1 myeloid leukemia patients, Immunol. Cell Biol. 90 (1) (2012) 109–115.
pathway, Oncogene 24 (13) (2005) 2229–2235. [43] S. Seth, et al., Intranodal interaction with dendritic cells dynamically regulates
[8] P. Kučan Brlić, et al., Targeting PVR (CD155) and its receptors in anti-tumor surface expression of the co-stimulatory receptor CD226 protein on murine T cells,
therapy, Cell. Mol. Immunol. 16 (1) (2019) 40–52. J. Biol. Chem. 286 (45) (2011) 39153–39163.
[9] X.-Y. Li, et al., CD155 loss enhances tumor suppression via combined host and [44] Q. Zhang, et al., Blockade of the checkpoint receptor TIGIT prevents NK cell ex-
tumor-intrinsic mechanisms, J. Clin. Invest. 128 (6) (2018) 2613–2625. haustion and elicits potent anti-tumor immunity, Nat. Immunol. 19 (7) (2018)

195
J.S. O’Donnell, et al. Seminars in Cancer Biology 65 (2020) 189–196

723–732. [73] D.-W. Huang, et al., CD155 expression and its correlation with clinicopathologic
[45] M. Li, et al., T-cell immunoglobulin and ITIM domain (TIGIT) receptor/poliovirus characteristics, angiogenesis, and prognosis in human cholangiocarcinoma, Onco.
receptor (PVR) ligand engagement suppresses interferon-gamma production of Ther. 10 (2017) 3817–3825.
natural killer cells via beta-arrestin 2-mediated negative signaling, J. Biol. Chem. [74] H. Yong, et al., CD155 expression and its prognostic value in postoperative patients
289 (25) (2014) 17647–17657. with breast cancer, Biomed. Pharmacother. 115 (2019) 108884.
[46] W. Chung, et al., Single-cell RNA-seq enables comprehensive tumour and immune [75] A. Iguchi-Manaka, et al., Increased soluble CD155 in the serum of cancer patients,
cell profiling in primary breast cancer, Nat. Commun. 8 (2017) 15081. PLoS One 11 (4) (2016) e0152982.
[47] S. Chevrier, et al., An Immune Atlas of Clear Cell Renal Cell Carcinoma, Cell 169 (4) [76] J. Gao, et al., CD155 downregulation synergizes with adriamycin to induce breast
(2017) 736–749 e18. cancer cell apoptosis, Apoptosis 23 (9–10) (2018) 512–520.
[48] F. Alfei, et al., TOX reinforces the phenotype and longevity of exhausted T cells in [77] A. Lepletier, et al., Tumor CD155 expression is associated with resistance to anti-
chronic viral infection, Nature 571 (7764) (2019) 265–269. PD1 immunotherapy in metastatic melanoma, Cancer Cell (submitted) (2019).
[49] A.C. Scott, et al., TOX is a critical regulator of tumour-specific T cell differentiation, [78] A. Soriani, et al., ATM-ATR-dependent up-regulation of DNAM-1 and NKG2D li-
Nature 571 (7764) (2019) 270–274. gands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell
[50] O. Khan, et al., TOX transcriptionally and epigenetically programs CD8 T cell ex- susceptibility and is associated with a senescent phenotype, Blood 113 (15) (2009)
haustion, Nature 571 (7764) (2019) 211–218. 3503–3511.
[51] S. Kurtulus, et al., TIGIT predominantly regulates the immune response via reg- [79] C. Fionda, et al., Nitric oxide donors increase PVR/CD155 DNAM-1 ligand ex-
ulatory T cells, J. Clin. Invest. 125 (11) (2015) 4053–4062. pression in multiple myeloma cells: role of DNA damage response activation, BMC
[52] N. Joller, et al., Treg cells expressing the coinhibitory molecule TIGIT selectively Cancer 15 (2015) 17.
inhibit proinflammatory Th1 and Th17 cell responses, Immunity 40 (4) (2014) [80] M. Ardolino, et al., DNAM-1 ligand expression on Ag-stimulated T lymphocytes is
569–581. mediated by ROS-dependent activation of DNA-damage response: relevance for NK-
[53] J.-M. Chauvin, et al., TIGIT and PD-1 impair tumor antigen-specific CD8+ T cells in T cell interaction, Blood 117 (18) (2011) 4778–4786.
melanoma patients, J. Clin. Invest. 125 (5) (2015) 2046–2058. [81] L. Vassena, et al., The human immunodeficiency virus type 1 Vpr protein upregu-
[54] R.J. Johnston, et al., The immunoreceptor TIGIT regulates antitumor and antiviral lates PVR via activation of the ATR-mediated DNA damage response pathway, J.
CD8(+) T cell effector function, Cancer Cell 26 (6) (2014) 923–937. Gen. Virol. 94 (Pt 12) (2013) 2664–2669.
[55] C. Guillerey, et al., TIGIT immune checkpoint blockade restores CD8 T-cell im- [82] J.L. Croxford, et al., ATM-dependent spontaneous regression of early Emu-myc-
munity against multiple myeloma, Blood 132 (16) (2018) 1689–1694. induced murine B-cell leukemia depends on natural killer and T cells, Blood 121
[56] A. Fuchs, et al., Cutting edge: CD96 (Tactile) promotes NK cell-target cell adhesion (13) (2013) 2512–2521.
by interacting with the poliovirus receptor (CD155), J. Immunol. 172 (7) (2004) [83] T. Inozume, et al., Melanoma cells control antimelanoma CTL responses via inter-
3994–3998. action between TIGIT and CD155 in the effector phase, J. Invest. Dermatol. 136 (1)
[57] C.J. Chan, et al., The receptors CD96 and CD226 oppose each other in the regula- (2016) 255–263.
tion of natural killer cell functions, Nat. Immunol. 15 (5) (2014) 431–438. [84] J. Madore, et al., PD-L1 expression in melanoma shows marked heterogeneity
[58] D. Mittal, et al., CD96 is an immune checkpoint that regulates CD8 T-cell antitumor within and between patients: implications for anti-PD-1/PD-L1 clinical trials,
function, Cancer Immunol. Res. 7 (4) (2019) 559–571. Pigment Cell Melanoma Res. 28 (3) (2015) 245–253.
[59] A. Lepletier, et al., The immune checkpoint CD96 defines a distinct lymphocyte [85] J.M. Taube, et al., Colocalization of inflammatory response with B7-h1 expression
phenotype and is highly expressed on tumor-infiltrating T cells, Immunol. Cell Biol. in human melanocytic lesions supports an adaptive resistance mechanism of im-
97 (2) (2019) 152–164. mune escape, Sci. Transl. Med. 4 (127) (2012) 127ra37.
[60] S.J. Blake, et al., Suppression of metastases using a new lymphocyte checkpoint [86] R.E. Vilain, et al., Dynamic changes in PD-L1 expression and immune infiltrates
target for Cancer immunotherapy, Cancer Discov. 6 (4) (2016) 446–459. early during treatment predict response to PD-1 blockade in melanoma, Clin.
[61] B. Husain, et al., A platform for extracellular interactome discovery identifies novel Cancer Res. 23 (17) (2017) 5024–5033.
functional binding partners for the immune receptors B7-H3/CD276 and PVR/ [87] K.E. Sloan, et al., CD155/PVR plays a key role in cell motility during tumor cell
CD155, Mol. Cell Proteomics 18 (11) (2019) 2310–2323. invasion and migration, BMC Cancer 4 (1) (2004).
[62] Y. Yamashita-Kanemaru, et al., CD155 (PVR/Necl5) mediates a costimulatory signal [88] W. Ikeda, et al., Tage4/Nectin-like molecule-5 heterophilically trans-interacts with
in CD4+ T cells and regulates allergic inflammation, J. Immunol. 194 (12) (2015) cell adhesion molecule Nectin-3 and enhances cell migration, J. Biol. Chem. 278
5644–5653. (30) (2003) 28167–28172.
[63] H. Georgiev, et al., CD155/CD226-interaction impacts on the generation of innate [89] D.J. Solecki, et al., Expression of the human poliovirus receptor/ CD155 gene is
CD8(+) thymocytes by regulating iNKT-cell differentiation, Eur. J. Immunol. 46 (4) activated by sonic hedgehog, J. Biol. Chem. 277 (28) (2002) 25697–25702.
(2016) 993–1003. [90] H. Harjunpää, et al., Deficiency of host CD96 and PD-1 or TIGIT enhances tumor
[64] N.K. Escalante, et al., CD155 on human vascular endothelial cells attenuates the immunity without significantly compromising immune homeostasis,
acquisition of effector functions in CD8 T cells, Arterioscler. Thromb. Vasc. Biol. 31 Oncoimmunology 7 (7) (2018) e1445949.
(5) (2011) 1177–1184. [91] B. Zhuo, et al., Overexpression of CD155 relates to metastasis and invasion in os-
[65] Z. Yan, Increased expression of CD155 and CD112 on monocytes in septic patients teosarcoma, Oncol. Lett. 15 (5) (2018) 7312–7318.
(INC6P.327), J. Immunol. 194 (Suppl. 1) (2015) 192.29. [92] Y. Meng, et al., CD155 blockade improves survival in experimental sepsis by re-
[66] N.K. Escalante, et al., CD155 on human vascular endothelial cells attenuates the versing dendritic cell dysfunction, Biochem. Biophys. Res. Commun. 490 (2) (2017)
acquisition of effector functions in CD8 T cells, Arterioscler. Thromb. Vasc. Biol. 31 283–289.
(5) (2011) 1177–1184. [93] N. Stanietsky, et al., Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction
[67] N. Kamran, et al., Toll-like receptor ligands induce expression of the costimulatory with PVR, Eur. J. Immunol. 43 (8) (2013) 2138–2150.
molecule CD155 on antigen-presenting cells, PLoS One 8 (1) (2013) e54406. [94] A.R. Aguilera, et al., CD96 targeted antibodies need not block CD96-CD155 inter-
[68] D. Pende, et al., Expression of the DNAM-1 ligands, Nectin-2 (CD112) and polio- actions to promote NK cell anti-metastatic activity, OncoImmunology 7 (5) (2018)
virus receptor (CD155), on dendritic cells: relevance for natural killer-dendritic cell e1424677.
interaction, Blood 107 (5) (2006) 2030–2036. [95] M.K. Merrill, et al., Poliovirus receptor CD155-targeted oncolysis of glioma,
[69] M. Gromeier, et al., Intergeneric poliovirus recombinants for the treatment of ma- NeuroOncology 6 (3) (2004) 208–217.
lignant glioma, Proc. Natl. Acad. Sci. U. S. A. 97 (12) (2000) 6803–6808. [96] E. Denniston, et al., The practical consideration of poliovirus as an oncolytic vir-
[70] V. Bevelacqua, et al., Nectin like-5 overexpression correlates with the malignant otherapy, Am. J. Virol. 5 (1) (2016) 1–7.
phenotype in cutaneous melanoma, Oncotarget 3 (8) (2012) 882–892. [97] E.M. Thompson, et al., Poliovirus receptor (CD155) expression in pediatric brain
[71] R. Nakai, et al., Overexpression of Necl-5 correlates with unfavorable prognosis in tumors mediates oncolysis of medulloblastoma and pleomorphic xanthoas-
patients with lung adenocarcinoma, Cancer Sci. 101 (5) (2010) 1326–1330. trocytoma, J. Neuropathol. Exp. Neurol. 77 (8) (2018) 696–702.
[72] D. Masson, et al., Overexpression of the CD155 gene in human colorectal carci- [98] P. Wang, G.-Z. Chen, Comment to "Recurrent glioblastoma treated with re-
noma, Gut 49 (2) (2001) 236–240. combinant poliovirus", Chin. Med. J. 131 (21) (2018) 2645–2646.

196

You might also like