You are on page 1of 12

REVIEWS

Inhibitory B7-family molecules in the


tumour microenvironment
Weiping Zou* and Lieping Chen‡
Abstract | The B7 family consists of activating and inhibitory co-stimulatory molecules that
positively and negatively regulate immune responses. Recent studies have shown that human
and rodent cancer cells, and stromal cells and immune cells in the cancer microenvironment
upregulate expression of inhibitory B7 molecules and that these contribute to tumour immune
evasion. In this Review, we focus on the roles of these B7 molecules in the dynamic interactions
between tumours and the host immune system, including their expression, regulation and
function in the tumour microenvironment. We also discuss novel therapeutic strategies that
target these inhibitory B7 molecules and their signalling pathways to treat human cancer.

Tumour-associated antigens The specific function of an individual’s immune system Inhibitory B7 molecules
(TAAs). Antigens that are in different physiological and pathological settings is T-cell activation and tolerance are not chance occur-
expressed by tumour cells. regulated by the actions of opposing factors or sys- rences. Many molecules form an orchestrated system to
These belong to three main tems. Common examples of this are the polarization of regulate the interactions between APCs and T cells. This
categories: tissue-
differentiation antigens, which
T helper (TH) cells into TH1-cell and TH2-cell subsets interaction is explained by the two-signal model, whereby
are also expressed by non- with opposing functions, and the balance between effec- signal one is provided to the T-cell receptor of T cells by
malignant cells; mutated or tor T‑cell activation and regulatory T‑cell activation. At the presentation of specific antigens on MHC molecules
aberrantly expressed the molecular level, co-stimulatory members of the B7 expressed by APCs, and signal two is provided by the B7
molecules; and cancer testis
family can have both inhibitory and stimulatory effects family and other co-stimulatory molecules that APCs
antigens, which are normally
expressed only by
on T‑cell activation. use to direct and/or fine-tune T‑cell responses (FIG. 1).
spermatocytes and An imbalance in immune regulation profoundly The growing B7 family now comprises seven members,
occasionally in the placenta. affects tumour-specific T‑cell immunity in the cancer which are CD80 (also known as B7.1), CD86 (also known
microenvironment and can reshape tumour progression, as B7.2), B7-DC (also known as PD‑L2 or CD273),
Myeloid-derived suppressor
cells
metastasis and immunotherapy in patients with cancer1. It B7-H1 (also known as PD‑L1 or CD274), B7-H2 (also
A population of cells that is well known that the lack of naturally induced immunity known as ICOSL), B7-H3 (also known as CD276) and
comprises mature and specific for tumour-associated antigens (TAAs) is not simply B7-H4 (also known as B7S1 or B7x)17–19. Compelling
immature myeloid cells. They a passive process whereby adaptive immunity is shielded evidence indicates that B7 molecules not only provide
are expanded and/or activated
from detecting TAAs1–9. It has been clearly shown that the crucial positive signals to stimulate and support T‑cell
during an inflammatory
immune response. Through
tumour microenvironment is comprised of dysfunctional activation, but can also offer negative signals that control
direct interactions and immune cells that are reprogrammed by active tumour- and suppress T‑cell responses17,18. These negative signals
secreted components, they mediated processes to evade tumour-specific immunity in are largely provided by the newly identified B7-family
inhibit T‑cell function. a highly effective manner. Three important mediators of members B7‑H1 and B7‑H4.
this evasion of tumour immunity that have been identi-
fied in the tumour microenvironment are: dysfunctional CD80/CD86–CTLA4. CD80 and CD86 control T-cell
*Department of Surgery, antigen-presenting cells (APCs), including dendritic cells activation by binding to and signalling through two
University of Michigan, Ann
(DCs), macrophages1,10 and myeloid-derived suppressor receptors, CD28 and cytotoxic T-lymphocyte antigen 4
Arbor, Michigan 48109, USA.

Departments of Dermatology cells11,12; regulatory T (TReg) cells13–16; and high levels of expres- (CTLA4), that are expressed by T cells (FIG. 1). CD80
and Oncology, Johns Hopkins sion of inhibitory B7 molecules by APCs, stromal cells and and CD86 are not classically considered as inhibitory
University School of Medicine, tumour cells1,17. The first two mediators have been spe- B7 molecules. However, on T‑cell activation, the expres-
Baltimore, Maryland 21231, cifically reviewed in the literature elsewhere1,10–13,15. In this sion of their inhibitory receptor, CTLA4, is induced
USA.
e-mails: wzou@med.umich.
Review, we focus on inhibitory B7 molecules, and detail on T cells, and engagement of CTLA4 by CD80 and
edu; lchen42@jhmi.edu their expression, regulation, function and therapeutic CD86 can limit and decrease T‑cell activation. The
doi:10.1038/nri2326 relevance in the tumour microenvironment. role of CTLA4 in controlling T‑cell activation and its

nature reviews | immunology volume 8 | june 2008 | 467


© 2008 Nature Publishing Group
REVIEWS

APC T cell B7‑H4. B7‑H4 was also identified by DNA sequence


homology to other B7 molecules28–30. B7‑H4 remains an
CD80 CD28 orphan ligand, although evidence indicates that a recep-
tor can be induced and could function on T cells28,30,31.
CD86 CTLA4 The currently known functions of B7‑H4 are exclusively
inhibitory and the effect of B7‑H4 might be mediated
by a single receptor. B- and T-lymphocyte attenuator
MHC TCR
(BTLA) was initially proposed to be the receptor for
B7‑H4 (Ref. 32), but recent studies show that this is not
B7-DC ? the case and that herpes virus entry mediator is the
PD-1 ligand for BTLA33–35.
B7-H1 CD80
Expression pattern
B7-H2 ICOS CD80 and CD86 have a restricted expression pattern,
being expressed mainly by professional APCs and
B7-H3 ? haematopoietic cells, but rarely by stromal cells and
non‑haematopoietic cancer cells17,18. By contrast, mRNA
B7-H4 ? encoding B7‑H1 and B7‑H4 is found in almost all tissues
and most stromal and haematopoietic cells. This distri-
bution pattern indicates unique functions for these mol-
IgV-like domain IgC-like domain
ecules in both lymphoid and non-lymphoid organs.
Figure 1 | The B7 family and antigen presentation to
T cells. Antigen-presenting cells (APCs) or APC-like cells Expression of B7‑H1. The expression of mRNA encod-
Nature
present a specific antigen on MHC Reviews |to
molecules Immunology
the T‑cell ing B7‑H1 is abundant in many tissues and organs
receptor (TCR) of T cells. Members of the B7 family and in humans20,36 and mice21. A recent study indicates
other co-stimulatory molecules are used to direct and/or that the phosphatase and tensin homologue (PTEN)–
fine-tune T‑cell responses. The newly identified B7‑H1 phosphatidylinositol-3‑kinase pathway might be
and B7‑H4 molecules provide negative signals that important in the post-transcriptional regulation of
control and suppress T‑cell responses. Human tumour
B7‑H1 cell-surface expression in tumours37. B7‑H1
cells and tumour-associated APCs express limited levels
of the stimulatory B7-family members CD80 and CD86, protein is often expressed by activated cells including
and high levels of the inhibitory B7-family members T cells, B cells, DCs, monocytes/macrophages 20,21,23,
Regulatory T (TReg) cells B7‑H1 and B7‑H4. This imbalance between the expression natural killer (NK) cells38, activated vascular endothe-
A T‑cell population that can of stimulatory and inhibitory B7 molecules might lial cells39, mesenchymal stem cells40 and cultured bone-
functionally suppress an contribute to tumour immune evasion in the tumour marrow-derived mast cells41. B7‑H1 is also found to
immune response by microenvironment. CTLA4; cytotoxic T-lymphocyte be expressed constitutively in immune-privileged sites
influencing the activity antigen 4; ICOS, inducible T-cell co-stimulator; PD-1, including the eyes and placenta42,43, which indicates
of another cell type. programmed cell death 1.
Several phenotypically that B7‑H1 might inhibit self-reactive T cells or B cells
distinct regulatory T‑cell and therefore control inflammatory responses in these
populations exist. The classic therapeutic relevance have been reviewed elsewhere19 tissues and organs.
regulatory T cells are and are not discussed in detail here. CTLA4-specific Most human cancers tested so far express high levels
CD4+CD25+FOXP3+ T cells
known as TReg cells.
antibodies have recently entered clinical trials for the of B7‑H1 protein44 (TABLE 1). However, low or rare B7‑H1
treatment of various human cancers19. expression is observed in most mouse and human
Two-signal model tumour-cell lines44. This might be owing to the lack
The concept that both the B7‑H1. B7‑H1 was first cloned on the basis of its DNA of the complete cancer microenvironment in cell lines
MHC–peptide complex
sequence homology to other B7 molecules belonging in vitro as these tumour-cell lines can upregulate B7‑H1
(signal 1) and co-stimulatory
signals delivered by B7-family to the immunoglobulin superfamily20. Programmed protein expression in response to cytokines44. Another
molecules expressed by cell death 1 (PD‑1; also known as CD279) has been possibility, albeit less likely, is that certain molecular
antigen-presenting cells subsequently identified as a counter-receptor for B7‑H1 profiles have been altered in established tumour-cell
(signal 2) are required for T‑cell (Ref. 21) (FIG. 1), and B7‑H1 is therefore also known as lines during culture. Therefore, the results obtained
activation. The absence of
signal 2 results in the induction
PD‑L1 to emphasize this receptor–ligand interaction21. from established tumour-cell lines in vitro might
of T‑cell anergy or deletion. However, experimental evidence indicates that addi- need to be interpreted carefully. In addition to tumour
tional counter-receptor(s) other than PD‑1 can mediate cells, B7‑H1 protein expression has been observed
Cytotoxic T‑lymphocyte- the functions of B7‑H1. In the absence of PD‑1 or if in human tumour-associated DCs 23,45, fibroblasts 46
antigen 4
binding to PD‑1 is blocked, B7‑H1 can have a stimula- and T cells47,48.
(CTLA4). After engagement by
CD80 or CD86 on antigen- tory effect on T‑cell immunity17,20,22–24. Moreover, CD80
presenting cells, CTLA4 is an additional counter-receptor for B7‑H1 for the Expression of B7‑H4. Similar to B7‑H1, mRNA encoding
signalling in activated T cells inhibition of T‑cell responses25. To make the situation B7‑H4 is widely distributed in peripheral tissues28,49,50.
induces cell-cycle arrest, more complicated, B7‑H1 can also function as a receptor However, although the expression of B7‑H4 cell-surface
decreases cytokine production
and inhibits T‑cell responses.
to transmit signals into T cells26 and tumour cells27. In protein was detected in normal human epithelial cells of
CD4+CD25+ TReg cells summary, B7‑H1 can act as both ligand and receptor to the female genital tract, kidney, lung and pancreas, B7‑H4
constitutively express CTLA4. execute immunoregulatory functions. protein was generally absent in other normal human

468 | june 2008 | volume 8 www.nature.com/reviews/immunol


© 2008 Nature Publishing Group
REVIEWS

Table 1 | B7‑H1 expression in human cancers and its immunological, clinical and pathological associations*
Human cancer type B7‑H1+ cases/ Immunological, clinical and pathological associations Refs
total cases
Breast cancer 24/56 Number of B7‑H1+ T cells correlates with tumour size, stage and HER2‡ expression 36,44,48
Colon cancer 16/25 ND 36,44
Gastric cancer 45/105 B7‑H1 expression correlates with increased tumour size, metastasis and poor survival 36,89
Glioma 10/10 B7‑H1 expression by tumour cells inhibits T‑cell activation in vitro 125
Leukaemia 17/30 B7‑H1 expression by leukaemia cells has no effect on T‑cell activation in vitro 81
Lung cancer 86/87 B7‑H1+ regions of the tumour contain fewer T cells in non-small cell lung cancer 36,44,86
Melanoma 22/22 ND 44
Multiple myeloma 82/82 B7‑H1+ plasma cells inhibit T‑cell activation in vitro 126
Oesophageal cancer 18/41 B7‑H1 expression is associated with poor prognosis 88
Ovarian cancer 82/93 B7‑H1 expression by tumour cells is associated with a decreased number of T cells in 23,36,44,87
the tumour and poor prognosis. B7‑H1+ tumour-associated dendritic cells inhibit T‑cell
function.
Pancreatic cancer 20/51 B7‑H1 expression by tumour cells is associated with a decreased number of tumour 127
T cells and poor prognosis.
Peripheral T‑cell lymphoma 7/11 ND 36
Renal-cell carcinoma 130/196 B7‑H1 expression by tumour cells is associated with poor prognosis 44,85,128
Thymic neoplasm 28/34 ND 36
Urothelial cancer 142/268 B7‑H1 expression by tumour cells is associated with advanced stage, recurrence and 129,130
poor survival
*The data for each tumour type are assembled from more than one report. Some data have not been included in the table if less than 10 tumour samples were
reported for a particular type of human cancer. ‡HER2 (also known as ERBB2 or Neu) is a tumour-associated antigen that is expressed in about 25% of cases of breast
cancer. Vaccines against HER2 are being tested for cancer therapy in humans. Monoclonal antibodies specific for HER2 (such as trastuzumab (Herceptin;
Genentech/Roche)) are approved for the therapy of human breast cancer. ND, not determined.

somatic tissues50. Nonetheless one study did observe Regulation of B7‑H1 expression. B7‑H1 expression can
broad cell-surface expression of B7‑H4 protein on mouse be induced or maintained by many cytokines23,36,44, of
haematopoietic cells30. The cause of this interspecies which interferon‑γ (IFNγ) is the most potent. Established
difference is unknown. human tumour-cell lines rarely express B7‑H1 protein
B7‑H4 is commonly detectable in the human cancer on the cell surface, but high levels of B7‑H1 expression
microenvironment (TABLE 2). For example, human ovar- can be induced by treatment with IFNγ in most of the
ian cancers express high levels of B7‑H4 protein31,49,51–54, cell lines tested so far44. Consistent with this, IFNγ can
and low levels of soluble B7‑H4 protein were found in induce high levels of B7‑H1 expression by normal epi-
the sera from patients with ovarian cancer52,53. In addi- thelial cells, vascular endothelial cells39, proximal tubular
tion to tumour cells, tumour-infiltrating macrophages31,55 epithelial cells59 and myeloid DCs36,44. It is assumed that a
and endothelial cells of small blood vessels56 in the can- strong TH1-cell response can induce B7‑H1 expression
cer microenvironment are also found to constitutively by APCs and other cells through IFNγ, and in turn main-
express B7‑H4. tain the threshold of T‑cell activation to avoid tissue and
Myeloid DCs
In summary, the current data reveal broad expression organ damage. In addition to IFNγ, type I IFN can also
A subset of dendritic cells (DCs)
that are lineage-negative, patterns of B7‑H1 and B7‑H4 protein in human tumours, stimulate B7‑H1 expression by hepatocytes60, monocytes,
HLA-DR+CD11c+ (in humans) in contrast to their rare expression in normal tissues. These DCs61 and tumour cells (S. Wei, I. Kryczek, L. Chen and
mononuclear cells with a data indicate that post-transcriptional regulation could W. Zou, unpublished observations). In this context, after
monocytoid appearance. have a crucial role in the control of B7‑H1 and B7‑H4 virus infection, tumour-associated plasmacytoid DCs
Human myeloid DCs might be
derived from myeloid
protein expression in normal tissues and organs, and produce large amounts of type I IFN in vitro62, which
precursors (for example, that this regulatory mechanism is aberrant in tumours. can in turn induce B7‑H1 expression23. Plasmacytoid
monocytes, macrophages and Further investigation of the regulatory mechanisms and DCs preferentially induce TH2-cell responses in certain
CD11c+ precursors). the signalling pathways leading to expression of B7‑H1 situations63. Therefore, the expression of B7‑H1 could
and B7‑H4 will generate important information for the potentially be stimulated in both TH1- and TH2-cell-
Plasmacytoid DCs
A subset of dendritic cells (DCs) understanding of tumour immune evasion and provide biased conditions, although the expression and relevance
that are lineage negative, potential molecular targets for treating human cancers. of B7‑H1 expression in TH1- or TH2-cell-associated dis-
HLA-DR+CD11c– (in humans) eases remains to be tested.
mononuclear cells with a Regulation of expression In light of its stimulatory effect on B7‑H1 expres-
microscopic appearance
similar to plasmablasts.
The tumour microenvironment contains a large number of sion, IFNγ could thus be a ‘double-edged sword’ in
Plasmacytoid DCs are the main cytokines and inflammatory mediators1,57,58. Some of these tumour immunity. Whereas IFNγ could increase anti-
producers of type I interferon. molecules can regulate the expression of B7‑H1 and B7‑H4. gen processing and presentation by upregulating the

nature reviews | immunology volume 8 | june 2008 | 469


© 2008 Nature Publishing Group
REVIEWS

Table 2 | B7‑H4 expression in human cancers and its immunological, clinical and pathological associations*
Human cancer type B7‑H4+ cases/ Immunological, clinical and pathological associations Refs
total cases
Breast cancer (primary) 165/173 B7‑H4 expression is associated with lack of expression of progesterone receptor and HER2‡ 50,54
Breast cancer 240/246 ND 50
(metastatic)
Lung cancer 35/86 B7‑H4 expression is more common in patients with lymph-node metastasis 49,131
Ovarian cancer 202/216 B7‑H4 tumour-associated macrophages inhibit T‑cell activation and predict poor survival
+
31,49,51–55
Prostate cancer 120/823 B7‑H4 expression by tumour cells is associated with disease spread, recurrence and death 90
Renal-cell carcinoma 153/259 B7‑H4 expression by tumour cells is associated with poor survival 56
Uterine endometrioid 90/90 B7‑H4 expression by tumour cells is associated with weak T‑cell infiltration and high-risk 132
adenocarcinoma tumours
*The data for each tumour type are assembled from more than one report. Some data have not been included in the table if less than 10 tumour samples were
reported for a particular type of human cancer. ‡HER2 (also known as ERBB2 or Neu) is a tumour-associated antigen that is expressed in about 25% of cases of breast
cancer. Vaccines against HER2 are being tested for cancer therapy in humans. Monoclonal antibodies specific for HER2 (such as trastuzumab (Herceptin;
Genentech/Roche)) are approved for the therapy of human breast cancer. ND, not determined.

expression of MHC molecules and components of the cells and B7‑H4 on APCs might be functionally distinct
antigen-processing machinery64, the effects of IFNγ on and be differentially regulated31,55. Collectively, B7‑H1
B7‑H1 expression might downregulate T‑cell immu- and B7‑H4 are regulated by distinct mechanisms.
nity. This could explain, at least partially, why IFNγ These differential regulatory patterns have important
has not been effective as a therapeutic agent for most implications in the generation and amplification of
human cancers. In addition, IFNγ has been shown to tumour-specific immunity.
stimulate the expression of indoleamine 2,3-dioxygenase
(IDO)65 and arginase66–68 on APCs. Such APCs could Evasion of tumour immunity
suppress anti-tumour immune responses. Therefore, The physiological functions of inhibitory B7-family
it is not surprising that an increase in the number of members are to limit, terminate and attenuate T‑cell
TAA-specific cytotoxic T lymphocytes (CTLs) does not responses, by which they prevent T‑cell hyperactivation
always translate into clinical regression of cancers69–72. and avoid tissue and organ damage during immune
In addition, IFNγ was also found to mediate CD4+ responses17,18. B7‑H1-deficient75,76 and B7‑H4‑deficient77
T‑cell loss and impair secondary anti-tumour immune mice have been generated and reported in the pub-
responses after successful initial immunotherapy in lished literature. The inhibitory B7 molecules can be
tumour-bearing mouse models73. As B7‑H1 expression induced in response to inflammation and potentially
can be induced on APCs and multiple human epithe- to broader danger signals. It is therefore possible that
Indoleamine 2,3- lial tumours, stimulation of B7‑H1 expression could be the expression of these molecules might contribute
dioxygenase a strategy used by the tumour to evade T‑cell‑mediated to de novo cancer initiation and development. At the
(IDO). An intracellular haem- tumour immunity. present time, however, there is no evidence that these
containing enzyme that molecules participate in carcinogenesis. However, these
catalyses the oxidative
catabolism of tryptophan.
Regulation of B7‑H4 expression. The regulation of inhibitory B7 molecules could suppress ongoing or
Insufficient availability of B7‑H4 expression has only been studied in the human induced tumour immunity.
tryptophan can lead to T‑cell system. Interleukin‑6 (IL‑6) and IL‑10 stimulate B7‑H4
apoptosis and anergy. expression by monocytes, macrophages and myeloid B7‑H1 expression by tumour cells. Initial studies
DCs. The DC‑differentiation cytokines, GM‑CSF showed that transgenic expression of B7‑H1 on a B7‑
Arginase
An enzyme that converts (granulocyte/macrophage colony-stimulating factor) H1‑deficient mouse P815 mastocytoma cell line did not
l‑arginine into l‑ornithine and and IL‑4, decrease B7‑H4 expression by these cells change its tumourigenicity in naive mice44. However,
urea. induced by IL‑6 and IL‑10 (Refs 31,55,74) (FIG. 2). IFNs B7‑H1+ P815 tumour cells could continue to grow in
seem to have a minimal effect on the induction of B7‑H4 the presence of adoptively transferred P815-specific
Danger signals
Agents that alert the immune
expression, in contrast to the induction of B7‑H1 expres- T cells, which are sufficient to induce the regression of
system to stress, usually by sion44. In human ovarian cancer, tumour-associated TReg wild-type P815 tumours44. This finding is consistent
interacting with Toll-like cells trigger macrophages to produce IL‑6 and IL‑10, and with the observation that transfection of B7‑H1 into
receptors and other pattern- these cytokines in turn stimulate B7‑H4 expression by a highly immunogenic P815 variant, which regresses
recognition receptors, and
APCs in an autocrine and/or paracrine manner55. High spontaneously in naive mice, led to progressive growth
thereby promote the
generation of innate and levels of IL‑6 and IL‑10, but not GM‑CSF and IL‑4, are of this line44. Similarly, in the presence of potent T‑cell
adaptive immune responses. detected in the ovarian tumour microenvironment. immunity, B7‑H1+ tumours are much more resistant
Danger signals can be Therefore, this dysfunctional cytokine network in the to T‑cell-mediated destruction in several mouse mod-
associated with microbial tumour micro­environment enables APCs to express els78–80 as well as in a human T‑cell leukaemia model81.
invaders (exogenous danger
signals) or produced by
B7‑H4. Interestingly, IL‑4, IL‑6, IL‑10 and GM‑CSF Interestingly, several B7‑H1– mouse tumours start to
damaged cells (endogenous have no regulatory effects on B7‑H4 expression on express B7‑H1 during growth in vivo. Injection of an
danger signals). tumour cells, which indicates that B7‑H4 on tumour antibody specific for B7‑H1 increased T‑cell immunity

470 | june 2008 | volume 8 www.nature.com/reviews/immunol


© 2008 Nature Publishing Group
REVIEWS

In this model, the injection of irradiated acute myeloid


leukaemia cells transduced by CXC‑chemokine ligand
10 (CXCL10) led to prophylactic immunity in mice.
APC TReg cell
Interestingly, CXCL10 stimulated B7‑H1 expression by
NK cells, and NK‑cell-associated B7‑H1 was essential
IL-6 for this prophylactic immunity38. These data indicate
and IL-10 that a stimulatory counter‑receptor for B7‑H1 is
GM-CSF and IL-4 expressed by T cells in the dormant tumour state owing
to the unique tumour microenvironment. It remains to
be determined if this mechanism is operative in other
↑ B7-H4
tumour models and in patients with cancer.

B7‑H4. Although high levels of B7‑H4 expression


Cell cycle are observed in human cancers (TABLE 2), there are no
published data showing that the expression of B7‑H4
T cell
by cancer cells can lead to accelerated tumour growth
Figure 2 | B7‑H4+ antigen-presenting cells in the or progression in immune-competent animals. The
tumour microenvironment. Nature B7‑H4+Reviews
antigen-presenting
| Immunology immunopathological relevance of B7‑H4 has only
cells (APCs), such as tumour-associated macrophages,
been studied in patients with ovarian cancer. High
induce T‑cell cycle arrest. B7‑H4 expression can be induced
by interleukin-6 (IL‑6) and IL‑10, and is inhibited by the
levels of B7‑H4 expression are found on a popula-
dendritic-cell differentiation cytokines granulocyte/ tion of tumour-associated macrophages in patients
macrophage colony-stimulating factor (GM-CSF) and IL‑4. with ovarian carcinoma. B7‑H4+ macrophages inhibit
Regulatory T (TReg) cells can trigger IL‑6 and IL‑10 TAA-specific T‑cell effector function31. These findings
production by APCs, and in turn upregulate B7‑H4 show that B7‑H4 is a negative regulator of TAA-specific
expression by APCs. APCs that have been conditioned by T‑cell immunity and is a molecular target for tumour
TReg cells inhibit T‑cell function through B7‑H4. High levels immunotherapy.
of TReg cells, and IL‑6 and IL‑10 are found in the tumour In summary, B7‑H1 and B7‑H4 are selectively
microenvironment, which helps to explain the observation expressed by various cellular components in the tumour
of B7‑H4+ APCs in this environment.
microenvironment, where they can inhibit tumour-
specific T‑cell immunity.
and induced tumour regression82. In these experimental
settings, it could not be excluded that B7‑H1 expression Cancer progression
by host cells, especially immune cells, could also have a Clinical data have documented that the expression of
role. In this regard, PD‑1-deficient mice79,83 were shown inhibitory B7 molecules correlates with poor prog-
to have increased T‑cell immunity and were more resist- nosis of various types of human cancer (TABLES 1,2).
ant to the outgrowth of transplanted murine tumours, However, all of these studies are retrospective and can-
which indicates that host-derived B7‑H1 has a negative cer tissues were evaluated for the expression of these
effect on tumour immunity. In addition, recent studies molecules at a specific time point. As it is unknown
show that the expression of B7‑H1 could also func- whether the level of expression of inhibitory B7 mol-
tion as a receptor to transmit an anti-apoptotic signal ecules varies during disease progression, the conclu-
to cancer cells as a means to resist immune-mediated sions and the implications from these analyses should
destruction27 (see later). be carefully considered. Nevertheless, these studies
represent an important step towards understanding
B7‑H1 expression by host cells. Myeloid DCs in human the prognosis of and developing novel treatments for
tumours and tumour-draining lymph nodes express patients with cancer.
high levels of B7‑H1 (Refs 23,45) . The interaction High levels of expression of B7‑H1 were initially
between B7‑H1+ myeloid DCs and tumour‑associated reported in many human tumours44. Subsequent stud-
T cells leads to the downregulation of expression of ies confirmed and extended these observations (TABLE 1).
myeloid-DC-derived IL‑12 and upregulation of expres- Frozen47 and formalin-fixed85 tumour tissues of clear-cell
sion of the immunosuppressive cytokine IL‑10 by renal carcinoma stained with a human B7‑H1-specific
myeloid DCs in a B7‑H1-dependent manner. Blockade monoclonal antibody44 showed that B7‑H1 expression
of B7‑H1 on tumour-infiltrating myeloid DCs increases is an indicator of poor prognosis for patient survival47.
IFNγ production by T cells and promotes tumour infil- In this study, however, the expression of B7‑H1 by
tration by IFNγ-producing T cells. Adoptive transfer of tumour-infiltrating lymphocytes derived from a frac-
such T cells improves the clearance of human tumours tion of patients was also significantly associated with
in xenotransplanted mice 23. These data support the poor prognosis47. Some reports have shown that B7‑H1
idea that B7‑H1 has a role in the downregulation of expression on tumour cells might also be associated with
DC‑mediated tumour immunity. decreased numbers of tumour-infiltrating lymphocytes
In the DA1-3b/C3H mouse model of acute myeloid leu- in patients with cancer86,87. Similarly, B7‑H1 expression
kaemia, dormant tumour cells resist CTL‑mediated killing was also identified as a poor prognostic factor in patients
owing to high levels of B7‑H1 expression by tumour cells84. with oesophageal, gastric88,89 and ovarian87 cancers.

nature reviews | immunology volume 8 | june 2008 | 471


© 2008 Nature Publishing Group
REVIEWS

The broad expression pattern of B7‑H1 in cancer Anergy. The role of B7‑H1 in T‑cell anergy was deter-
microenvironments and its relationship with clinical and mined with the use of a cell-culture system in which
pathological parameters in patients with cancer provide alloreactive T cells were induced by monocyte-derived
strong evidence that B7‑H1 is pathologically relevant, DCs. IL‑10-treated DCs induced unresponsive T cells
and that B7‑H1 and its signalling pathway are justified similar to anergic T cells, but these T cells could be
targets for treating human cancer. rescued by restimulation with DCs treated with an
Although the role of B7‑H4 expression by tumour antibody specific for human B7‑H1 (Ref. 94). A subse-
cells in evading tumour immunity has not been well quent study showed that resting DC‑induced tolerance
established, several human studies indicate that B7‑H4 of CD8 + T cells in mixed bone-marrow chimaeras
expression might be associated with the progression of could be prevented by the ablation of PD‑1+ T cells95.
certain types of human cancer. In patients with renal In the non-obese diabetic (NOD) mouse model, B7‑H1
carcinoma56 and prostate cancer90, B7‑H4 expression by and PD‑1, but not CTLA4 and TReg cells, were shown
tumour cells is associated with adverse clinical features. to be crucial for T‑cell tolerance and anergy96. Given
A recent study in patients with ovarian cancer showed the inducible nature of B7‑H1 expression in peripheral
that the level of B7‑H4 expression by tumour-associated tissues and of PD‑1 expression by activated T cells, it
macrophages, but not tumour cells, correlates with the was assumed that effector T cells, after priming in the
number of TReg cells in the tumour. Further, expression lymph nodes, migrate to peripheral tissues, where they
of B7‑H4 by macrophages and the number of TReg cells are induced to express PD‑1, and become functionally
are both negatively associated with patient outcome55. In anergic when engaged with B7‑H1 on peripheral organs
this context, B7‑H4 is shown to be a signature gene that through PD‑1. However, this view was challenged by
is consistently expressed in breast cancer91. two recent studies97,98. These studies have shown that the
expression of PD‑1 can be induced on T cells while they
Mechanisms of action are still in lymphoid organs during priming. Blockade of
B7‑H1-expressing cells use at least six distinct mecha- B7‑H1 or PD‑1 by neutralizing monoclonal antibodies
nisms to evade T‑cell immunity (FIG. 3): inducing apop- before T-cell egress to peripheral organs could convert
tosis, anergy or exhaustion of T cells, forming a molecular anergic T cells into fully activated effector T cells97,98.
shield to protect tumour cells from lysis, inducing pro- Therefore, the interaction of B7‑H1 with PD‑1 could
duction of the immunosuppressive cytokine IL‑10 and regulate T‑cell anergy in both priming and effector
promoting TReg-cell-mediated suppression. It is possible phases of an immune response. In the context of tumour
that these mechanisms work as a hierarchy to evade immune pathology, human tumour-associated DCs
immunity at different levels of T‑cell function to impose highly express B7‑H1 (Refs 23,45) and PD‑1+ T cells99
tight control. are found in the tumours. It remains unknown whether
the B7‑H1–PD‑1 interaction induces anergy of human
Apoptosis. Co-culture with B7‑H1+ tumour-cell lines TAA-specific naive and effector T cells.
increased the apoptosis of human TAA-specific T cells;
blocking B7‑H1 decreased this apoptosis44. Subsequent Exhaustion. This mechanism was revealed in stud-
studies showed that B7‑H1-mediated apoptosis is a ies of chronic infection. PD‑1 is highly expressed by
common physiological process used by the host to functionally exhausted antigen-specific T cells, and
maintain homeostasis in peripheral tissues. For example, B7‑H1 could be detected in infected tissues and lym-
B7‑H1-deficient mice have an accumulation of CD8+ phoid organs. Blockade of B7‑H1 or PD‑1 can rescue
T cells in the liver due to decreased apoptosis75. Liver the functionality of these exhausted T cells100–103. The
Kupffer cells92 and stellate cells93 constitutively express data indicate that PD‑1 might transmit an inhibitory
B7‑H1, and human hepatocytes express low levels signal that dominates TCR signalling and decreases
of B7‑H1 (Ref. 60). This ready availability of B7‑H1 in T‑cell responses during prolonged exposure to antigens
the liver might explain the decreased apoptosis of T cells during chronic infection.
Anergy
A state of non-responsiveness in B7‑H1-deficient mice75. B7‑H1 is also constitutively Cancer could be considered as a chronic inflam-
to antigen. Anergic T or B cells expressed in the eye. After corneal allografting, B7‑H1+ matory disease58. Not only do up to 15% of cancers
cannot respond to their corneal endothelium interacted with PD‑1+CD4+ T cells worldwide have a direct infectious origin104, but many
cognate antigens under and led to T‑cell apoptosis43. This observation could at human tumours are also related to chronic irritation and
optimal conditions of
stimulation.
least partially explain how the immune-privileged status inflammation1. Human tumour-associated DCs highly
of the cornea could be established. express B7‑H1, and blocking B7‑H1 markedly increases
Exhaustion B7‑H1 could have a role in the induction of effec- the effector function of tumour T cells23. In this context,
An ‘operational’ definition that tor T‑cell apoptosis in both a PD‑1-dependent and a recent study showed that PD‑1 is upregulated on a sig-
refers to the loss of antigen-
-independent manner. In one human T‑cell clone, B7‑H1 nificant fraction of tumour-infiltrating T cells in patients
specific T‑cell responses in vivo
after prolonged or repetitive can induce apoptosis of PD‑1– T cells44, but in most mouse with melanoma and that blockade of PD‑1 increased
stimulation with antigen. This models, PD‑1 is required for B7‑H1‑mediated apoptosis17. TAA-specific T‑cell proliferation and function99. Taken
has been best observed in a However, B7‑H1 can bind to PD‑1 (Ref. 21) and CD80 together, these data indicate that B7‑H1 and PD‑1 could
model of infection with (Ref. 25), and thereby regulate T‑cell function. The detailed result in human tumour-specific T‑cell exhaustion. This
lymphocytic choriomeningitis
virus Docile strain, for which
molecular mechanisms of apoptosis mediated by B7‑H1 notion, although it has not been formally tested in can-
the exact mechanism is still not remain to be elucidated, and could occur indirectly or cer, needs to be taken into consideration when designing
understood. through an uncharacterized direct pathway. tumour immunotherapies.

472 | june 2008 | volume 8 www.nature.com/reviews/immunol


© 2008 Nature Publishing Group
REVIEWS

acquisition of lysis resistance by tumour cells occurs


IL-10 production rapidly, within 4–16 hours, and was also observed for
allogeneic27,105 and re-directed T cells37 in addition to TAA-
PD-1 specific T cells. This effect could be interpreted in terms
Anergy of the rapid induction of suppressive effects on T cells
through interaction with PD‑1. However, further stud-
ies showed that this was due to the unique nature of the
Exhaustion Apoptosis tumour-cell surface, but not to the dysfunction of T cells,
Immune suppression
because wild-type tumour cells in the presence of B7‑H1+
tumour cells in the same culture could still be lysed by
B7-H1 T cells82. By disabling the intracellular domain of B7‑H1
on cancer cells, the ‘molecular shield’ is eliminated and
TReg cell the tumour cells become susceptible in vitro to immune‑
APC
B7-H1 mediated destruction. Importantly, tumours expressing
B7‑H1 with a truncated intracellular domain also become
Tumour cell more susceptible to T‑cell-mediated immunotherapy
in vivo compared with tumours expressing wild-type
B7‑H1. By contrast, the intracellular truncation of PD‑1
Molecular on T cells does not eliminate the ‘molecular-shield’ effect27.
shield In addition, B7‑H1 reverse signalling in cancer cells also
renders resistance to apoptosis mediated by antibodies
specific for CD95 (also known as FAS) and by staurosporin
Protected from
cytotoxic lysis toxin from Streptomyces staurospores (a broad-spectrum
protein-kinase inhibitor)27. In light of its broad expres-
sion pattern, B7‑H1 might be a ubiquitous anti-apoptotic
receptor expressed by cancer cells and might have a
CTL general role in the resistance of tumour cells to immune
Figure 3 | The inhibitory actions of B7‑H1 in tumour immune evasion. Tumour cells destruction and other apoptosis-based therapies.
and tumour-associated antigen-presenting cells (APCs) express high levels of B7‑H1. The
potential suppressive mechanisms of B7‑H1 have been addressed NatureinReviews
various| models
Immunology IL‑10 production. Selective induction of IL‑10 was found
in vitro and in vivo, and it is probable that a combination of mechanisms, rather than a initially following the stimulation of human T cells with
single mode of action, is used by B7‑H1-expressing cells. B7‑H1+ tumour cells and APCs CD3-specific antibody and B7‑H1–immuno­globulin
might induce T‑cell apoptosis, anergy, functional exhaustion or IL‑10 production. B7‑H1+
fusion protein20. Tumour-associated B7‑H1+ DCs also
tumour cells might be resistant to lysis by cytotoxic T lymphocytes (CTLs); this has been
induce T‑cell production of IL‑10 (Ref. 23). Subsequent
described as a molecular shield. Tumour-associated regulatory T (TReg) cells might express
B7‑H1 and mediate T‑cell suppression partially through the B7‑H1–PD‑1 pathway. The studies have established a correlation between the upreg-
detailed molecular mechanisms remain poorly understood. ulation of B7‑H1 expression and increased levels of IL‑10
in patients with HIV and HBV infection107,108. It remains
to be determined whether increased IL‑10 production
Molecular shield. The inclusion of neutralizing mono- has an important role in B7‑H1-mediated suppression
clonal antibodies specific for B7‑H1 or PD‑1 often of T‑cell responses in vivo.
increases TAA-specific CTL-mediated lysis or cytokine
release against murine and human tumour cells that TReg-cell-mediated suppression. It has been suggested that
express B7‑H1 (Refs 82–84,105,106). This observa- B7‑H1 is involved in the development and function of
tion indicates that endogenous levels of expression of TReg cells. B7‑H1+ vascular endothelial cells109 and gastric
B7‑H1 might be sufficient to confer resistance against epithelial cells110 induce the development of TReg cells in
CTL‑mediated lysis. The effect of neutralizing monoclonal an in vitro culture system. However, it has not been shown
antibodies specific for B7‑H1 or PD‑1 varies depend- that B7‑H1 or PD‑1 is required for TReg-cell differentia-
ing on the system, ranging from moderate to dramatic. tion in vivo. In tumour-bearing mice, TReg cells activated
For example, after transfection to express high levels of by IDO+ DCs induce B7‑H1 expression on target DCs.
B7‑H1, P815 mouse tumour cells are much more resist- Importantly, the ability of these TReg cells to suppress
ant to lysis by antigen-specific CD8+ T cells82. Similarly, T‑cell activation could be abrogated by antibody specific
the addition of a B7‑H1-specific monoclonal antibody to for B7‑H1 (Ref. 111). In support of this observation, it has
B7‑H1‑transfected B16‑F10 melanoma cells results in the been shown that human tumour-associated DCs express
increased secretion of cytokines from 2C CTLs105. This B7‑H1 and mediate T‑cell suppression in a B7‑H1-
resistance to cytotoxic lysis mediated by the expression of dependent manner23,45. By contrast, in non-Hodgkin lym-
high levels of B7‑H1 depends on the B7‑H1–PD‑1 inter- phoma, intratumoral CD4+CD25+ TReg cells can express
action and was described as being a ‘molecular shield’82. B7‑H1, and blocking B7‑H1 with a monoclonal antibody
In addition, tumour-specific T cells are more effective at partially decreases TReg-cell-mediated T‑cell inhibition112.
lysing human glioma cells that express wild-type PTEN These results indicate that B7‑H1 expression by tumour-
with low-level B7‑H1 expression than those that express associated DCs and TReg cells might contribute to tumour
mutant PTEN with high-level B7‑H1 expression37. The immune suppression.

nature reviews | immunology volume 8 | june 2008 | 473


© 2008 Nature Publishing Group
REVIEWS

B7‑H4 can also evade tumour immunity. B7‑H4 has non-haematopoietic cells is crucial for the accelerated
been studied in less detail than B7‑H1 in the context of disease120. However, in light of the mild autoimmune
tumour immune evasion, but there is evidence indicat- phenotypes in B7‑H1-deficient mice75,76, B7‑H1 block-
ing that B7‑H4 might exert its function through myeloid ade will be unlikely to result in the generation of severe
APCs and TReg cells to mediate T‑cell suppression in the autoimmune disease.
tumour microenvironment31,55,74. Tumour‑associated In addition to the effects of B7‑H1 on transplantation,
macrophages markedly outnumber other types of APC, autoimmune diseases and tumours, it has been found
such as DCs, and form an abundant population of APCs in that trophoblasts, which are located in the maternal–
solid tumours113–116. One population of ovarian‑cancer- fetal interface, express B7‑H1, and B7‑H1+ trophoblasts
associated macrophages expresses high levels of B7‑H4, might have a role in the suppression of maternal–fetal
and these B7‑H4+ macrophages induce T‑cell cycle arrest immunity42. Therefore, a potential side effect of B7‑H1
in vitro and in vivo partially through B7‑H4 (Ref. 31). blockade by antibody in pregnant women could be the
These findings indicate that B7‑H4 might contribute to termination of pregnancy. Nevertheless, B7‑H1-deficient
tumour immune evasion, and that B7‑H4 is a molecular mice produce normal size litters, which indicates that
target for tumour immunotherapy. this is also a less likely event.
In human ovarian cancer, TReg cells and B7‑H4+ Similarly, neutralizing antibodies specific for PD‑1
macrophages are co-localized and their numbers are will be an important addition to clinical trials. However,
correlated in the tumour environment16,31,55,74. TReg cells neutralizing antibodies specific for PD‑1 might block the
trigger high levels of IL‑6 and IL‑10 production by APCs, interaction of PD‑1 with B7-DC (FIG. 1), another counter-
and in turn, these cytokines stimulate the expression receptor for PD‑1 with potentially stimulatory effects. In
of B7‑H4 by APCs, which renders the APCs immuno­ addition, PD‑1-specific antibodies might increase the
suppressive55 (FIG. 2). These findings are in line with the risk of severe autoimmune diseases, as predicted from
observations that macrophages spontaneously produce the spontaneous development of autoimmune diseases
IL‑6 and IL‑10 in the ovarian tumour environment55,62. in PD‑1-deficient mice17,18. Finally, although in vitro
These data mechanistically link IL‑10, B7‑H4, TReg cells assays support the claim that certain specific monoclonal
and APCs, and provide a new cellular and molecular antibodies are antagonistic, if appropriate crosslinking is
mechanism for TReg-cell-mediated immunosuppression provided, these antibodies could be potentially agonis-
at the level of APCs13,55 (FIG. 2). tic121. Nonetheless, the significant potential benefits of
the clinical application of B7‑H1–PD‑1 blockade warrant
Implications for cancer immunotherapy extensive experimental and clinical studies in patients
Many tumour-associated APCs and tumour cells express with cancer.
B7‑H1 and B7‑H4, and these molecules can mediate
T‑cell suppression. The manipulation of B7-induced B7‑H4 blockade. Efficient neutralizing antibodies specific
immune suppression might therefore be a broadly appli- for human B7‑H4 are not yet available. Small interfering RNA
cable therapeutic modality to treat human cancers. (siRNA)54 and antisense oligonucleotides specific for B7‑H4
(Refs 31,74) have been used to block B7‑H4 expression.
B7‑H1 and PD‑1 blockade. Preclinical data have set the Blocking the expression of B7‑H4 by tumour‑associated
stage for clinical trials by blocking B7‑H1 in patients with macrophages disables their suppressive capacity, enables
Small interfering RNA cancer. In the context of its suppressive effects on T cells TAA-specific effector T‑cell function and reduces tumour
Double-stranded RNAs and anti-apoptotic effect on tumour cells, it would seem growth in human ovarian cancer xenografts31,74. Taken
(dsRNAs) with sequences that
precisely match a given gene
beneficial to block B7‑H1 using neutralizing antibodies. together with the proposed inhibitory role for B7‑H4 in
are able to ‘knock down’ the However, antibodies specific for B7‑H1 could potentially mouse systems28–30, these data justify B7‑H4 as a promis-
expression of that gene by also block the interaction of B7‑H1 with a putative co- ing new target for development of therapeutic reagents for
directing RNA-degrading stimulatory receptor. In this regard, it has been reported the treatment of human cancers.
enzymes to destroy the
that local expression of B7‑H1 can provide positive co-
encoded mRNA transcript. The
two most common forms of stimulation for naive T cells and promote organ-specific Combinatorial blockade. It is probable that multiple
dsRNAs used for gene silencing autoimmunity and transplant rejection in mice117. For suppressive mechanisms mediate immune evasion in a
are short — usually 21-bp long obvious reasons, therapeutic antibodies should be tested given tumour. Combinatorial treatments will therefore
— small interfering RNAs for their ability to block the B7‑H1–CD80 interaction in be required to reverse tumour immune-escape pathways
(siRNAs) or the plasmid-
delivered short hairpin RNAs
addition to blocking B7‑H1–PD‑1 binding. Despite the and lead to potent TAA-specific T‑cell immunity. In
(shRNAs). possible immune-stimulatory roles of B7‑H1 in some tumour-bearing mouse models, for example, blocking
mouse models of transplantation and autoimmune B7‑H1 in combination with blocking TGFβ signalling
Antisense oligonucleotides diseases, the effects of B7‑H1 are largely immunosup- using neutralizing antibodies synergistically induces
Short, gene-specific sequences
pressive on mouse and human tumour immunity. In tumour regression133. B7‑H1 expression is induced
of nucleic acids that are
of the opposite strand terms of the suppressive effects of B7‑H1, blocking on tumour-associated TReg cells in non-Hodgkin lym-
(complementary) to the B7‑H1 could possibly enhance ongoing autoimmune phoma and on target DCs in murine tumour‑draining
targeted mRNA. Classical diseases. For example, treatment with B7‑H1-specific lymph nodes. These TReg cells inhibit the function of
antisense oligonucleotides antibody moderately accelerates experimental autoim- PD‑1+ tumour-infiltrating T cells, partially through the
target specific strands of RNA
within a cell, thereby
mune encephalomyelitis in mice118. B7‑H1-deficient B7‑H1–PD‑1 pathway111,112. These studies indicate that
preventing translation of these NOD mice also develop diabetes more rapidly than wild- simultaneously blocking B7‑H1–PD‑1 and TReg cells
RNAs. type NOD mice119, and the loss of B7‑H1 expression on might be therapeutically relevant.

474 | june 2008 | volume 8 www.nature.com/reviews/immunol


© 2008 Nature Publishing Group
REVIEWS

The administration of a blocking monoclonal anti- an attractive strategy to treat human cancers. However,
body specific for B7‑H1 increases the therapeutic effects as we move forward to clinical trials, we have to bear in
of a co-stimulatory CD137-specific agonistic monoclonal mind several possibilities. First, the immune suppres-
antibody82,122 and of tumour-cell vaccination123. B7‑H1 sion mediated by inhibitory B7 molecules might not be
and PD‑1 blockade together with an HSP70 vaccine124 the only or the main immunosuppressive mechanism
can increase T‑cell immunity and decrease tumour used by certain tumour stages and/or certain types of
growth and metastasis in mouse models. In addition to tumour. Therefore, it is essential to define the nature and
these examples, blocking the B7‑H1–PD‑1 pathway in functional relevance of inhibitory B7-family members
combination with blockade of other well-defined sup- in each individual human tumour microenvironment.
pressive molecules, including CTLA4, VEGF, B7‑H4, Second, the interactions between the B7-family mem-
TGFβ, arginase or IDO1, could be possible options in bers and their receptors are complex and might reflect
preclinical and clinical settings to treat cancer. differences in temporal and spatial expression of these
molecules and their affinities for one another in the
Concluding remarks tumour microenvironment. Third, blocking inhibitory
There is now compelling evidence to show that tumours B7 molecules might result in autoimmune diseases.
escape host immunity by actively developing multiple Finally, the receptor(s) and signalling pathways have not
suppressive mechanisms in the tumour microenviron- been identified for several B7-family members. These
ment. The mechanisms that underlie the interactions limitations will probably constrain the clinical applica-
between the immune system and the tumour micro- tion of targeting inhibitory B7-family members as a
environment, particularly in humans, are a crucial and therapeutic modality. Identification of all the counter-
understudied area of cancer immunology, the under- receptors in these pathways and careful analysis of their
standing of which will have a significant impact on positive and negative regulatory functions will help us to
the success of immunotherapy strategies. The selective design more elegant strategies to maximize anti‑tumour
expression of inhibitory B7 molecules in the tumour immunity while decreasing the risk of autoimmune dis-
microenvironment has been determined as an impor- eases. In addition, targeting one pathway will be highly
tant immunosuppressive mechanism in many types unlikely to lead to reliable and consistent clinical effi-
of human tumour. Therefore, the manipulation of the cacy, and combinatorial therapeutic regimens will need
expression of and signalling through these molecules is to be developed.

1. Zou, W. Immunosuppressive networks in the tumour 16. Curiel, T. J. et al. Specific recruitment of regulatory 26. Dong, H. et al. Costimulating aberrant T cell
environment and their therapeutic relevance. Nature T cells in ovarian carcinoma fosters immune privilege responses by B7‑H1 autoantibodies in rheumatoid
Rev. Cancer 5, 263–274 (2005). and predicts reduced survival. Nature Med. 10, arthritis. J. Clin. Invest. 111, 363–370 (2003).
2. Khong, H. T. & Restifo, N. P. Natural selection of 942–949 (2004). 27. Azuma, T. et al. B7‑H1 is a ubiquitous antiapoptotic
tumor variants in the generation of “tumor escape” 17. Chen, L. Co-inhibitory molecules of the B7-CD28 receptor on cancer cells. Blood 111, 3635–3643
phenotypes. Nature Immunol. 3, 999–1005 family in the control of T‑cell immunity. Nature Rev. (2008).
(2002). Immunol. 4, 336–347 (2004). This study reports that B7‑H1 functions as a
3. Kaufman, H. L. & Disis, M. L. Immune system versus 18. Sharpe, A. H. & Freeman, G. J. The B7–CD28 receptor to transmit an anti-apoptotic signal to
tumor: shifting the balance in favor of DCs and superfamily. Nature Rev. Immunol. 2, 116–126 cancer cells as a means to resist immune-mediated
effective immunity. J. Clin. Invest. 113, 664–667 (2002). destruction.
(2004). 19. Korman, A. J., Peggs, K. S. & Allison, J. P. Checkpoint 28. Sica, G. L. et al. B7‑H4, a molecule of the B7 family,
4. Pardoll, D. Does the immune system see tumors as blockade in cancer immunotherapy. Adv. Immunol. 90, negatively regulates T cell immunity. Immunity 18,
foreign or self? Annu. Rev. Immunol. 21, 807–839 297–339 (2006). 849–861 (2003).
(2003). 20. Dong, H., Zhu, G., Tamada, K. & Chen, L. B7‑H1, a 29. Zang, X. et al. B7x: a widely expressed B7 family
5. Cerundolo, V., Hermans, I. F. & Salio, M. Dendritic third member of the B7 family, co-stimulates T‑cell member that inhibits T cell activation. Proc. Natl
cells: a journey from laboratory to clinic. Nature proliferation and interleukin‑10 secretion. Nature Acad. Sci. USA 100, 10388–10392 (2003).
Immunol. 5, 7–10 (2004). Med. 5, 1365–1369 (1999). 30. Prasad, D. V., Richards, S., Mai, X. M. & Dong, C.
6. Gilboa, E. The promise of cancer vaccines. Nature Rev. This study reported the first identification of B7S1, a novel B7 family member that negatively
Cancer 4, 401–411 (2004). B7‑H1 and indicated a negative role of B7-H1 in regulates T cell activation. Immunity 18, 863–873
7. Mellor, A. L. & Munn, D. H. IDO expression by T‑cell responses. (2003).
dendritic cells: tolerance and tryptophan catabolism. 21. Freeman, G. J. et al. Engagement of the PD‑1 References 28–30 identified B7‑H4 and
Nature Rev. Immunol. 4, 762–774 (2004). immunoinhibitory receptor by a novel B7 family described the inhibitory role of B7‑H4 in T‑cell
8. Finn, O. J. Cancer vaccines: between the idea and the member leads to negative regulation of responses.
reality. Nature Rev. Immunol. 3, 630–641 (2003). lymphocyte activation. J. Exp. Med. 192, 31. Kryczek, I. et al. B7‑H4 expression identifies a novel
9. Schreiber, H., Wu, T. H., Nachman, J. & Kast, W. M. 1027–1034 (2000). suppressive macrophage population in human ovarian
Immunodominance and tumor escape. Semin. Cancer This study showed that PD‑1 is an inhibitory carcinoma. J. Exp. Med. 203, 871–881 (2006).
Biol. 12, 25–31 (2002). receptor for B7‑H1. This study showed that human tumour-associated
10. Banchereau, J. & Palucka, A. K. Dendritic cells as 22. Wang, S. et al. Molecular modeling and functional macrophages express B7‑H4 and can suppress
therapeutic vaccines against cancer. Nature Rev. mapping of B7‑H1 and B7-DC uncouple costimulatory tumour‑antigen‑specific T‑cell responses, and it
Immunol. 5, 296–306 (2005). function from PD‑1 interaction. J. Exp. Med. 197, provided a cytokine profile for the regulation of
11. Gabrilovich, D. Mechanisms and functional 1083–1091 (2003). B7‑H4 expression.
significance of tumour-induced dendritic-cell defects. 23. Curiel, T. J. et al. Blockade of B7‑H1 improves myeloid 32. Watanabe, N. et al. BTLA is a lymphocyte inhibitory
Nature Rev. Immunol. 4, 941–952 (2004). dendritic cell-mediated antitumor immunity. Nature receptor with similarities to CTLA‑4 and PD‑1. Nature
12. Bronte, V. & Zanovello, P. Regulation of immune Med. 21, 562–567 (2003). Immunol. 4, 670–679 (2003).
responses by l‑arginine metabolism. Nature Rev. This study showed that human tumour- 33. Sedy, J. R. et al. B and T lymphocyte attenuator
Immunol. 5, 641–654 (2005). associated DCs express B7‑H1 and can inhibit regulates T cell activation through interaction with
13. Zou, W. Regulatory T cells, tumour immunity and tumour‑antigen‑specific T‑cell function through herpesvirus entry mediator. Nature Immunol.
immunotherapy. Nature Rev. Immunol. 6, 295–307 B7‑H1. 6, 90–98 (2005).
(2006). 24. Kanai, T. et al. Blockade of B7‑H1 suppresses the 34. Compaan, D. M. et al. Attenuating lymphocyte
14. Wei, S., Kryczek, I. & Zou, W. Regulatory T‑cell development of chronic intestinal inflammation. activity: the crystal structure of the BTLA-HVEM
compartmentalization and trafficking. Blood 108, J. Immunol. 171, 4156–4163 (2003). complex. J. Biol. Chem. 280, 39553–39561 (2005).
426–431 (2006). 25. Butte, M. J., Keir, M. E., Phamduy, T. B., Sharpe, A. H. 35. Gonzalez, L. C. et al. A coreceptor interaction between
15. Sakaguchi, S. Naturally arising Foxp3-expressing & Freeman, G. J. Programmed death‑1 ligand 1 the CD28 and TNF receptor family members B and T
CD25+CD4+ regulatory T cells in immunological interacts specifically with the b7–1 costimulatory lymphocyte attenuator and herpesvirus entry
tolerance to self and non-self. Nature Immunol. molecule to inhibit T cell responses. Immunity 27, mediator. Proc. Natl Acad. Sci. USA 102, 1116–1121
6, 345–352 (2005). 111–122 (2007). (2005).

nature reviews | immunology volume 8 | june 2008 | 475


© 2008 Nature Publishing Group
REVIEWS

36. Brown, J. A. et al. Blockade of programmed death‑1 59. Waeckerle-Men, Y., Starke, A. & Wuthrich, R. P. PD‑L1 83. Iwai, Y. et al. Involvement of PD‑L1 on tumor cells in
ligands on dendritic cells enhances T cell activation partially protects renal tubular epithelial cells from the the escape from host immune system and tumor
and cytokine production. J. Immunol. 170, attack of CD8+cytotoxic T cells. Nephrol. Dial. immunotherapy by PD‑L1 blockade. Proc. Natl Acad.
1257–1266 (2003). Transplant 22, 1527–1536 (2007). Sci. USA 99, 12293–12297 (2002).
37. Parsa, A. T. et al. Loss of tumor suppressor PTEN 60. Muhlbauer, M. et al. PD‑L1 is induced in hepatocytes 84. Saudemont, A. & Quesnel, B. In a model of tumor
function increases B7‑H1 expression and by viral infection and by interferon-α and -γ and dormancy, long-term persistent leukemic cells have
immunoresistance in glioma. Nature Med. 13, 84–88 mediates T cell apoptosis. J. Hepatol. 45, 520–528 increased B7‑H1 and B7.1 expression and resist CTL-
(2007). (2006). mediated lysis. Blood 104, 2124–2133 (2004).
This study showed that the increased expression at 61. Schreiner, B. et al. Interferon-β enhances monocyte 85. Thompson, R. H. et al. Tumor B7‑H1 is associated
a post-transcriptional level of B7‑H1 in human and dendritic cell expression of B7‑H1 (PD‑L1), a with poor prognosis in renal cell carcinoma patients
glioma is associated with loss of function of PTEN. strong inhibitor of autologous T‑cell activation: with long-term follow-up. Cancer Res. 66,
38. Saudemont, A., Jouy, N., Hetuin, D. & Quesnel, B. NK relevance for the immune modulatory effect in 3381–3385 (2006).
cells that are activated by CXCL10 can kill dormant multiple sclerosis. J. Neuroimmunol. 155, 172–182 86. Konishi, J. et al. B7‑H1 expression on non-small cell
tumor cells that resist CTL-mediated lysis and can (2004). lung cancer cells and its relationship with tumor-
express B7‑H1 that stimulates T cells. Blood 105, 62. Zou, W. et al. Stromal-derived factor‑1 in human infiltrating lymphocytes and their PD‑1 expression.
2428–2435 (2005). tumors recruits and alters the function of Clin. Cancer Res. 10, 5094–5100 (2004).
39. Mazanet, M. M. & Hughes, C. C. B7‑H1 is expressed plasmacytoid precursor dendritic cells. Nature Med. 87. Hamanishi, J. et al. Programmed cell death 1 ligand 1
by human endothelial cells and suppresses T cell 7, 1339–1346 (2001). and tumor-infiltrating CD8+ T lymphocytes are
cytokine synthesis. J. Immunol. 169, 3581–3588 63. Rissoan, M. C. et al. Reciprocal control of T helper cell prognostic factors of human ovarian cancer. Proc. Natl
(2002). and dendritic cell differentiation. Science 283, Acad. Sci. USA 104, 3360–3365 (2007).
40. Augello, A. et al. Bone marrow mesenchymal 1183–1186 (1999). 88. Ohigashi, Y. et al. Clinical significance of programmed
progenitor cells inhibit lymphocyte proliferation by 64. Boehm, U., Klamp, T., Groot, M. & Howard, J. C. death‑1 ligand‑1 and programmed death‑1 ligand‑2
activation of the programmed death 1 pathway. Eur. Cellular responses to interferon-γ. Annu. Rev. Immunol. expression in human esophageal cancer. Clin. Cancer
J. Immunol. 35, 1482–1490 (2005). 15, 749–795 (1997). Res. 11, 2947–2953 (2005).
41. Nakae, S. et al. Mast cells enhance T cell activation: 65. Munn, D. H. et al. Potential regulatory function of 89. Wu, C. et al. Immunohistochemical localization of
importance of mast cell costimulatory molecules and human dendritic cells expressing indoleamine 2,3- programmed death‑1 ligand‑1 (PD‑L1) in gastric
secreted TNF. J. Immunol. 176, 2238–2248 (2006). dioxygenase. Science 297, 1867–1870 (2002). carcinoma and its clinical significance. Acta Histochem.
42. Petroff, M. G., Chen, L., Phillips, T. A. & Hunt, J. S. B7 66. Mazzoni, A. et al. Myeloid suppressor lines inhibit 108, 19–24 (2006).
family molecules: novel immunomodulators at the T cell responses by an NO‑dependent mechanism. 90. Zang, X. et al. B7‑H3 and B7x are highly expressed in
maternal-fetal interface. Placenta 23 (Suppl. A), J. Immunol. 168, 689–695 (2002). human prostate cancer and associated with disease
95–101 (2002). 67. Kusmartsev, S. A., Li, Y. & Chen, S. H. Gr‑1+ myeloid spread and poor outcome. Proc. Natl Acad. Sci. USA
43. Hori, J. et al. B7‑H1‑induced apoptosis as a cells derived from tumor-bearing mice inhibit 104, 19458–19463 (2007).
mechanism of immune privilege of corneal allografts. primary T cell activation induced through CD3/ 91. Sadun, R. E. et al. Immune signatures of murine and
J. Immunol. 177, 5928–5935 (2006). CD28 costimulation. J. Immunol. 165, 779–785 human cancers reveal unique mechanisms of tumor
44. Dong, H. et al. Tumor-associated B7‑H1 promotes (2000). escape and new targets for cancer immunotherapy.
T‑cell apoptosis: A potential mechanism of immune 68. Saio, M., Radoja, S., Marino, M. & Frey, A. B. Clin. Cancer Res. 13, 4016–4025 (2007).
evasion. Nature Med. 8, 793–800 (2002). Tumor-infiltrating macrophages induce apoptosis in 92. Oikawa, T. et al. Intrahepatic expression of the co-
This study showed that tumour-associated B7‑H1 activated CD8+ T cells by a mechanism requiring cell stimulatory molecules programmed death‑1, and its
induces T‑cell apoptosis and that B7‑H1 protein is contact and mediated by both the cell-associated ligands in autoimmune liver disease. Pathol. Int. 57,
found in many types of human tumour, which could form of TNF and nitric oxide. J. Immunol. 167, 485–492 (2007).
be upregulated by IFNγ. 5583–5593 (2001). 93. Chen, C. H. et al. In vivo immune modulatory activity
45. Perrot, I. et al. Dendritic cells infiltrating human non- 69. Yee, C., Riddell, S. R. & Greenberg, P. D. Prospects for of hepatic stellate cells in mice. Hepatology 44,
small cell lung cancer are blocked at immature stage. adoptive T cell therapy. Curr. Opin. Immunol. 1171–1181 (2006).
J. Immunol. 178, 2763–2769 (2007). 9, 702–708 (1997). 94. Selenko-Gebauer, N. et al. B7‑h1 (programmed
46. Nazareth, M. R. et al. Characterization of human lung 70. Maio, M. & Parmiani, G. Melanoma immunotherapy: death‑1 ligand) on dendritic cells is involved in the
tumor-associated fibroblasts and their ability to new dreams or solid hopes? Immunol. Today 17, induction and maintenance of T cell anergy.
modulate the activation of tumor-associated T cells. 405–407 (1996). J. Immunol. 170, 3637–3644 (2003).
J. Immunol. 178, 5552–5562 (2007). 71. Maeurer, M. J., Storkus, W. J., Kirkwood, J. M. & 95. Probst, H. C., McCoy, K., Okazaki, T., Honjo, T. &
47. Thompson, R. H. et al. Costimulatory B7‑H1 in renal Lotze, M. T. New treatment options for patients with van den Broek, M. Resting dendritic cells induce
cell carcinoma patients: Indicator of tumor melanoma: review of melanoma- derived T‑cell peripheral CD8+ T cell tolerance through PD‑1 and
aggressiveness and potential therapeutic target. epitope-based peptide vaccines. Melanoma Res. CTLA‑4. Nature Immunol. 6, 280–286 (2005).
Proc. Natl Acad. Sci. USA 101, 17174–17179 6, 11–24 (1996). 96. Fife, B. T. et al. Insulin-induced remission in new-onset
(2004). 72. Romero, P. et al. Ex vivo staining of metastatic lymph NOD mice is maintained by the PD‑1–PD‑L1 pathway.
48. Ghebeh, H. et al. The B7‑H1 (PD‑L1) T lymphocyte- nodes by class I major histocompatibility complex J. Exp. Med. 203, 2737–2747 (2006).
inhibitory molecule is expressed in breast cancer tetramers reveals high numbers of antigen- 97. Tsushima, F. et al. Interaction between B7‑H1 and
patients with infiltrating ductal carcinoma: correlation experienced tumor-specific cytolytic T lymphocytes. PD‑1 determines initiation and reversal of T‑cell
with important high-risk prognostic factors. Neoplasia J. Exp. Med. 188, 1641–1650 (1998). anergy. Blood 110, 180–185 (2007).
8, 190–198 (2006). 73. Berner, V. et al. IFN-γ mediates CD4+ T‑cell loss and 98. Goldberg, M. V. et al. Role of PD‑1 and its ligand,
49. Choi, I. H. et al. Genomic organization and expression impairs secondary antitumor responses after B7‑H1, in early fate decisions of CD8 T cells. Blood
analysis of B7‑H4, an immune inhibitory molecule of successful initial immunotherapy. Nature Med. 13, 110, 186–192 (2007).
the B7 family. J. Immunol. 171, 4650–4654 (2003). 354–360 (2007). 99. Wong, R. M. et al. Programmed death‑1 blockade
50. Tringler, B. et al. B7‑h4 is highly expressed in ductal 74. Kryczek, I. et al. Cutting Edge: Induction of B7‑H4 enhances expansion and functional capacity of human
and lobular breast cancer. Clin. Cancer Res. 11, on APCs through IL‑10: novel suppressive mode for melanoma antigen-specific CTLs. Int. Immunol. 19,
1842–1848 (2005). regulatory T cells. J. Immunol. 177, 40–44 1223–1234 (2007).
51. Bignotti, E. et al. Differential gene expression profiles (2006). 100. Barber, D. L. et al. Restoring function in exhausted
between tumor biopsies and short-term primary 75. Dong, H. et al. B7‑H1 determines accumulation and CD8 T cells during chronic viral infection. Nature 439,
cultures of ovarian serous carcinomas: identification of deletion of intrahepatic CD8+ T lymphocytes. 682–687 (2006).
novel molecular biomarkers for early diagnosis and Immunity 20, 327–336 (2004). 101. Petrovas, C. et al. PD‑1 is a regulator of virus-specific
therapy. Gynecol. Oncol. 103, 405–416 (2006). 76. Latchman, Y. E. et al. PD‑L1‑deficient mice show that CD8+ T cell survival in HIV infection. J. Exp. Med.
52. Tringler, B. et al. B7‑H4 overexpression in ovarian PD‑L1 on T cells, antigen-presenting cells, and host 203, 2281–2292 (2006).
tumors. Gynecol. Oncol. 100, 44–52 (2006). tissues negatively regulates T cells. Proc. Natl Acad. 102. Day, C. L. et al. PD‑1 expression on HIV-specific T cells
53. Simon, I. et al. B7‑h4 is a novel membrane-bound Sci. USA 101, 10691–10696 (2004). is associated with T‑cell exhaustion and disease
protein and a candidate serum and tissue biomarker 77. Suh, W. K. et al. Generation and characterization of progression. Nature 443, 350–354 (2006).
for ovarian cancer. Cancer Res. 66, 1570–1575 B7‑H4/B7S1/B7x-deficient mice. Mol. Cell Biol. 26, 103. Trautmann, L. et al. Upregulation of PD‑1 expression
(2006). 6403–6411 (2006). on HIV-specific CD8+ T cells leads to reversible
54. Salceda, S. et al. The immunomodulatory protein 78. Strome, S. E. et al. B7‑H1 blockade augments immune dysfunction. Nature Med. 12, 1198–1202
B7‑H4 is overexpressed in breast and ovarian cancers adoptive T‑cell immunotherapy for squamous cell (2006).
and promotes epithelial cell transformation. Exp. Cell carcinoma. Cancer Res. 63, 6501–6505 (2003). 104. Kuper, H., Adami, H. O. & Trichopoulos, D. Infections
Res. 306, 128–141 (2005). 79. Iwai, Y., Terawaki, S. & Honjo, T. PD‑1 blockade as a major preventable cause of human cancer.
55. Kryczek, I. et al. Relationship between B7‑H4, inhibits hematogenous spread of poorly immunogenic J. Intern. Med. 248, 171–183 (2000).
Regulatory T cells, and patient outcome in human tumor cells by enhanced recruitment of effector 105. Blank, C. et al. PD‑L1/B7H‑1 inhibits the effector phase
ovarian carcinoma. Cancer Res. 67, 8900–8905 T cells. Int. Immunol. 17, 133–144 (2005). of tumor rejection by T cell receptor (TCR) transgenic
(2007). 80. Tsushima, F. et al. Predominant expression of B7‑H1 CD8+ T cells. Cancer Res. 64, 1140–1145 (2004).
56. Krambeck, A. E. et al. B7‑H4 expression in renal cell and its immunoregulatory roles in oral squamous cell 106. Blank, C. et al. Blockade of PD‑L1 (B7‑H1) augments
carcinoma and tumor vasculature: Associations with carcinoma. Oral Oncol. 42, 268–274 (2006). human tumor-specific T cell responses in vitro.
cancer progression and survival. Proc. Natl Acad. Sci. 81. Salih, H. R. et al. The role of leukemia-derived B7‑H1 Int. J. Cancer 119, 317–327 (2006).
USA 103, 10391–10396 (2006). (PD‑L1) in tumor‑T‑cell interactions in humans. Exp. 107. Geng, L. et al. B7‑H1 expression is upregulated in
57. Balkwill, F. Cancer and the chemokine network. Hematol. 34, 888–894 (2006). peripheral blood CD14+ monocytes of patients with
Nature Rev. Cancer 4, 540–550 (2004). 82. Hirano, F. et al. Blockade of B7‑H1 and PD‑1 by chronic hepatitis B virus infection, which correlates
58. Coussens, L. M. & Werb, Z. Inflammation and cancer. monoclonal antibodies potentiates cancer therapeutic with higher serum IL‑10 levels. J. Viral. Hepat. 13,
Nature 420, 860–867 (2002). immunity. Cancer Res. 65, 1089–1096 (2005). 725–733 (2006).

476 | june 2008 | volume 8 www.nature.com/reviews/immunol


© 2008 Nature Publishing Group
REVIEWS

108. Trabattoni, D. et al. B7‑H1 is up-regulated in HIV 117. Subudhi, S. K. et al. Local expression of B7‑H1 programmed death‑1 pathway in human pancreatic
infection and is a novel surrogate marker of disease promotes organ-specific autoimmunity and transplant cancer. Clin. Cancer Res. 13, 2151–2157 (2007).
progression. Blood 101, 2514–2520 (2003). rejection. J. Clin. Invest. 113, 694–700 (2004). 128. Thompson, R. H. et al. Costimulatory molecule B7‑H1
109. Krupnick, A. S. et al. Murine vascular endothelium 118. Zhu, B. et al. Differential role of programmed death- in primary and metastatic clear cell renal cell
activates and induces the generation of allogeneic ligand 1 [corrected] and programmed death-ligand 2 carcinoma. Cancer 104, 2084–2091 (2005).
CD4+25+Foxp3+ regulatory T cells. J. Immunol. 175, [corrected] in regulating the susceptibility and chronic 129. Nakanishi, J. et al. Overexpression of B7‑H1 (PD‑L1)
6265–6270 (2005). progression of experimental autoimmune significantly associates with tumor grade and
110. Beswick, E. J., Pinchuk, I. V., Das, S., Powell, D. W. & encephalomyelitis. J. Immunol. 176, 3480–3489 postoperative prognosis in human urothelial cancers.
Reyes, V. E. Expression of the programmed death (2006). Cancer Immunol. Immunother. 56, 1173–1182 (2007).
ligand 1, B7‑H1, on gastric epithelial cells after 119. Ansari, M. J. et al. The programmed death‑1 (PD‑1) 130. Inman, B. A. et al. PD‑L1 (B7‑H1) expression by
Helicobacter pylori exposure promotes development pathway regulates autoimmune diabetes in nonobese urothelial carcinoma of the bladder and BCG-induced
of CD4+ CD25+ FoxP3+ regulatory T cells. Infect. diabetic (NOD) mice. J. Exp. Med. 198, 63–69 granulomata: associations with localized stage
Immun. 75, 4334–4341 (2007). (2003). progression. Cancer 109, 1499–1505 (2007).
111. Sharma, M. D. et al. Plasmacytoid dendritic cells from 120. Keir, M. E. et al. Tissue expression of PD‑L1 mediates 131. Sun, Y. et al. B7‑H3 and B7‑H4 expression in
mouse tumor-draining lymph nodes directly activate peripheral T cell tolerance. J. Exp. Med. 203, non‑small‑cell lung cancer. Lung Cancer 53, 143–151
mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. 883–895 (2006). (2006).
Invest. 117, 2570–2582 (2007). 121. del Rio, M. L. et al. Antibody-mediated signaling 132. Miyatake, T. et al. B7‑H4 (DD‑O110) is overexpressed
This study showed that plasmacytoid DCs from through PD‑1 costimulates T cells and enhances in high risk uterine endometrioid adenocarcinomas
mouse tumour-draining lymph nodes activate CD28-dependent proliferation. Eur. J. Immunol. 35, and inversely correlated with tumor T‑cell infiltration.
regulatory T (TReg) cells through indoleamine 3545–3560 (2005). Gynecol. Oncol. 106, 119–127 (2007).
2,3-dioxygenase, these activated TReg cells induce 122. Melero, I. et al. Monoclonal antibodies against the 133. Wei S. et al. Tumor-induced immune suppression of in
B7‑H1 on target DCs and mediate tumour immune 4–1BB T‑cell activation molecule eradicate established vivo T effector cell priming is mediated by the B7-H1/
suppression in a B7‑H1-dependent manner. tumors. Nature Med. 3, 682–685 (1997). PD-1 axis and TGF-β1. Cancer Res. (in the press).
112. Yang, Z. Z., Novak, A. J., Stenson, M. J., Witzig, T. E. 123. Webster, W. S. et al. Targeting molecular and cellular
Acknowledgements
& Ansell, S. M. Intratumoral CD4+CD25+ regulatory inhibitory mechanisms for improvement of antitumor
We would like to thank our former and current trainees and
T‑cell‑mediated suppression of infiltrating CD4+ T cells memory responses reactivated by tumor cell vaccine.
collaborators for their intellectual input and hard work. The
in B‑cell non-Hodgkin lymphoma. Blood 107, J. Immunol. 179, 2860–2869 (2007).
work described in this Review was supported by grants from
3639–3646 (2006). 124. Geng, H. et al. HSP70 vaccine in combination with
the United States National Institutes of Health and the United
113. Wyckoff, J. et al. A paracrine loop between tumor cells gene therapy with plasmid DNA encoding sPD‑1
States Department of Defense.
and macrophages is required for tumor cell migration overcomes immune resistance and suppresses the
in mammary tumors. Cancer Res. 64, 7022–7029 progression of pulmonary metastatic melanoma. Int.
(2004). J. Cancer 118, 2657–2664 (2006). DATABASES
114. Pollard, J. W. Tumour-educated macrophages promote 125. Wintterle, S. et al. Expression of the B7-related Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.
tumour progression and metastasis. Nature Rev. molecule B7‑H1 by glioma cells: a potential fcgi?db=gene
Cancer 4, 71–78 (2004). mechanism of immune paralysis. Cancer Res. 63, B7-DC | B7-H1 | B7-H2 | B7-H3 | B7-H4 | CD80 | CD86
115. Vakkila, J. & Lotze, M. T. Inflammation and necrosis 7462–7467 (2003).
promote tumour growth. Nature Rev. Immunol. 126. Liu, J. et al. Plasma cells from multiple myeloma FURTHER INFORMATION
4, 641–648 (2004). patients express B7‑H1 (PD‑L1) and increase Weiping Zou’s homepage:
116. Mantovani, A., Sozzani, S., Locati, M., Allavena, P. & expression following stimulation with IFN-γ and TLR http://sitemaker.umich.edu/zou.lab/home
Sica, A. Macrophage polarization: tumor-associated ligands via a MyD88‑, TRAF6‑, and MEK-dependent Lieping Chen’s homepage:
macrophages as a paradigm for polarized M2 pathway. Blood 110, 296–304 (2007). http://gradimmunology.med.som.jhmi.edu
mononuclear phagocytes. Trends Immunol. 23, 127. Nomi, T. et al. Clinical significance and therapeutic All links are active in the online pdf
549–555 (2002). potential of the programmed death‑1 ligand/

nature reviews | immunology volume 8 | june 2008 | 477


© 2008 Nature Publishing Group

You might also like