negative regulators of t-cell activation: potential ......activation gene 3 (lag-3)⁄cd223 suggest...

21
Ó 2009 John Wiley & Sons A/S Immunological Reviews 229/2009 67 Tsvetelina Pentcheva-Hoang Emily Corse James P. Allison Negative regulators of T-cell activation: potential targets for therapeutic intervention in cancer, autoimmune disease, and persistent infections Authors’ address Tsvetelina Pentcheva-Hoang 1 , Emily Corse 1 , James P. Allison 1 1 Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA. Correspondence to: James P. Allison Department of Immunology Howard Hughes Medical Institute, Ludwig Center for Cancer Immunotherapy Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York, NY 10021, USA Tel.: +1 646 888 2332 Fax: +1 646 422 0618 e-mail: [email protected] Immunological Reviews 2009 Vol. 229: 67–87 Printed in Singapore. All rights reserved Ó 2009 John Wiley & Sons A/S Immunological Reviews 0105-2896 Summary: The generation of productive adaptive immune responses depends on the antigen-specific activation of T and B cells. The outcome of T-cell receptor engagement is influenced by signals from both positive and negative regulatory molecules that can either activate or inhibit T-cell function. CD28 and cytotoxic T-lymphocyte antigen-4 are the prototypi- cal members of an immunoglobulin domain-containing protein family that play important roles in the control of T-cell responses against infec- tion, cancer, and in autoimmune disease. Although the precise molecular details of their functions are still under active investigation, tumors and chronic pathogens seem to have exploited these pathways to achieve immune evasion. Furthermore, malfunction of the inhibitory arm of the immune response appears responsible for the development of multiple autoimmune pathologies. As a result, the negative regulators of T-cell activation have become attractive targets for therapeutic intervention in cancer, chronic infection, and autoimmune disease. The application of findings from basic research has provided insight into the manipulation of these pathways in the clinic and offers promising strategies for the treatment of disease. Keywords: CTLA-4, PD-1, BTLA, LAG-3, immunotherapy Introduction The activation of ab T cells requires the engagement of unique T-cell receptors (TCRs) by antigenic peptides bound to major histocompatibility complex (MHC) molecules on the surface of antigen-presenting cells (APCs). This interaction provides the so-called ‘signal 1’, which by itself is not only insufficient for T-cell activation but can lead to apoptosis or a state of antigen- specific non-responsiveness (anergy) (1). Optimal T-cell proliferation and acquisition of effector functions require an additional costimulatory ‘signal 2’, which is provided by surface molecules on specialized professional APCs (1). CD28 is the founding member of a growing family of costimulatory molecules and has been shown to enhance

Upload: others

Post on 27-Jan-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 67

    Tsvetelina Pentcheva-Hoang

    Emily Corse

    James P. Allison

    Negative regulators of T-cellactivation: potential targets fortherapeutic intervention in cancer,autoimmune disease, and persistentinfections

    Authors’ address

    Tsvetelina Pentcheva-Hoang1, Emily Corse1, James P. Allison1

    1Department of Immunology, Memorial Sloan-Kettering

    Cancer Center, New York, NY, USA.

    Correspondence to:

    James P. Allison

    Department of Immunology

    Howard Hughes Medical Institute, Ludwig Center for

    Cancer Immunotherapy

    Memorial Sloan-Kettering Cancer Center

    1275 York Avenue

    New York, NY 10021, USA

    Tel.: +1 646 888 2332

    Fax: +1 646 422 0618

    e-mail: [email protected]

    Immunological Reviews 2009

    Vol. 229: 67–87

    Printed in Singapore. All rights reserved

    � 2009 John Wiley & Sons A/SImmunological Reviews

    0105-2896

    Summary: The generation of productive adaptive immune responsesdepends on the antigen-specific activation of T and B cells. The outcomeof T-cell receptor engagement is influenced by signals from both positiveand negative regulatory molecules that can either activate or inhibit T-cellfunction. CD28 and cytotoxic T-lymphocyte antigen-4 are the prototypi-cal members of an immunoglobulin domain-containing protein familythat play important roles in the control of T-cell responses against infec-tion, cancer, and in autoimmune disease. Although the precise moleculardetails of their functions are still under active investigation, tumors andchronic pathogens seem to have exploited these pathways to achieveimmune evasion. Furthermore, malfunction of the inhibitory arm of theimmune response appears responsible for the development of multipleautoimmune pathologies. As a result, the negative regulators of T-cellactivation have become attractive targets for therapeutic intervention incancer, chronic infection, and autoimmune disease. The application offindings from basic research has provided insight into the manipulationof these pathways in the clinic and offers promising strategies for thetreatment of disease.

    Keywords: CTLA-4, PD-1, BTLA, LAG-3, immunotherapy

    Introduction

    The activation of ab T cells requires the engagement of unique

    T-cell receptors (TCRs) by antigenic peptides bound to major

    histocompatibility complex (MHC) molecules on the surface of

    antigen-presenting cells (APCs). This interaction provides the

    so-called ‘signal 1’, which by itself is not only insufficient for

    T-cell activation but can lead to apoptosis or a state of antigen-

    specific non-responsiveness (anergy) (1). Optimal T-cell

    proliferation and acquisition of effector functions require an

    additional costimulatory ‘signal 2’,which is provided by surface

    molecules on specialized professional APCs (1).

    CD28 is the founding member of a growing family of

    costimulatory molecules and has been shown to enhance

  • interleukin-2 (IL-2) secretion and to prevent the induction of

    anergy in both T-cell clones and primary T cells (2, 3). Upon

    binding to its ligands, B7-1 (CD80) and B7-2 (CD86), CD28

    enhances T-cell proliferation by increasing the transcription

    and the mRNA stability of IL-2 (4) as well as by upregulating

    the anti-apoptotic protein Bcl-XL (5). Although the precise

    mechanism of CD28-mediated costimulation is unknown,

    some of the possibilities include driving the formation of a

    mature immunological synapse (6), assembly of an indepen-

    dent signaling complex (7), and augmentation of the TCR

    signals, potentially via the recruitment of lipid rafts to the

    synapse (8).

    A closely related molecule, cytotoxic T-lymphocyte antigen-

    4 (CTLA-4) (CD152) (9), binds to the same ligands as CD28

    with much higher affinity (10, 11). In contrast to CD28,

    CTLA-4 restricts T-cell activation, as indicated by the fact that

    CTLA-4 ligation inhibits IL-2 production, IL-2 receptor

    expression, and cell cycle progression (12, 13) and that

    CTLA-4-deficient mice die of a lymphoproliferative disorder

    within 3–4 weeks of age (14–16).

    The more recent discovery of new homologues of

    CD28 ⁄ CTLA-4 and their ligands has gradually expanded thisprotein family to include additional costimulatory [inducible

    T-cell costimulator (ICOS) ⁄CD278 and its ligand B7h ⁄B7-RP ⁄ ICOS-L ⁄CD275 (17–19)], as well as inhibitory[programmed death-1 (PD-1) ⁄CD279 and its ligands PD-L1 ⁄B7-H1 ⁄ CD274 and PD-L2 ⁄B7-DC ⁄CD273 (20–23)] members.Furthermore, the identification of B7-H3 ⁄CD276 (24, 25)

    and B7x ⁄B7-H4 (26–28) as novel homologues of B7-1 andB7-2 that can inhibit T cells by binding to currently unknown

    receptors extends this family even further. An additional

    receptor of the immunoglobulin superfamily (IgSF) that

    negatively regulates T-cell activation is the B and T-lympho-

    cyte attenuator (BTLA) ⁄CD272 (29), which binds the herpes-virus entry mediator (HVEM) (30). HVEM has also recently

    been shown to interact with another negative regulator of T

    cells, CD160 (31). Finally, recent studies of the lymphocyte

    activation gene 3 (LAG-3) ⁄CD223 suggest that it plays animportant role in the regulation of T-cell responses (32–34).

    This review focuses on recent advances in our understanding

    of these negative regulators of T-cell activation (Fig. 1).

    CTLA-4, B7-1, and B7-2

    Protein structure

    Mouse CTLA-4 is a 223-amino acid (a.a.) type I trans-

    membrane glycoprotein that contains an unpaired cysteine

    residue at position 122 (Cys157 if the signal sequence is

    included) that is responsible for the formation of a disulfide-

    linked homodimer (9, 35). There are several CTLA-4 splice

    variants that include a soluble form (36, 37) as well as a

    ligand-independent molecule, found only in the mouse,

    which may be associated with resistance to autoimmune dis-

    ease (38, 39). The solution and crystal structures of both

    mouse and human CTLA-4, either alone or in complexes with

    B7-1 and B7-2, have been solved (40–43) and show that the

    B7-1B7-1PD-1PD-1

    LAG-3LAG-3TCRTCR

    CD160CD160

    BTLABTLA

    PD-L1PD-L1

    CTLA-4CTLA-4LAG-3LAG-3

    TCRTCRCD28CD28

    PD-1PD-1

    TCRTCR

    TCRTCR

    ??

    BTLABTLACD160CD160B7-1B7-1

    PD-1PD-1LAG-3LAG-3

    CD28CD28BTLABTLA

    CD160CD160BTLABTLAPD-1PD-1

    B7-1B7-1

    PD-L1PD-L1

    HVEMHVEMPD-L1PD-L1

    CTLA-4CTLA-4

    PD-1PD-1B7-1B7-1

    CTLA-4CTLA-4

    pMHCpMHCB7-1B7-1

    B7-1B7-1

    HVEMHVEM

    pMHCpMHCpMHCpMHC

    PD-L2PD-L2B7-2B7-2

    PD-L1PD-L1PD-L1PD-L1

    B7xB7x

    B7-H3B7-H3

    HVEMHVEMEffector T cellEffector T cellproliferationproliferation

    and/or functionand/or function

    B7-2B7-2

    HVEMHVEM

    PD-L2PD-L2

    pMHCpMHC

    PD-L1PD-L1

    DendriticDendriticcellcell

    PeripheralPeripheraltissuestissues

    RegulatoryRegulatoryT cellT cell

    TissueTissuemacrophagemacrophage

    Fig. 1. T-cell inhibitors of the immunoglobulin superfamily.

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    68 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • extracellular domain consists of a two-layer b-sandwich that

    resembles an Ig variable (IgV) domain (40, 41). The two

    ligand-binding sites lie distally to the homodimer interface,

    allowing the simultaneous binding of two B7-1 or two B7-2

    molecules to a single CTLA-4 (41–43).

    The mouse B7-1 protein is a 306 aa type I transmembrane

    glycoprotein that has an extracellular region consisting of two

    Ig-like domains: a membrane-distal IgV-like domain and a

    membrane-proximal Ig constant region (IgC)-like domain

    (44). The transmembrane segment contains two cysteine resi-

    dues that might be involved in lipid modifications or covalent

    interactions with other membrane proteins (44). Because of

    alternative splicing, there is a mouse B7-1 variant with a

    different cytoplasmic domain, which might contain distinct

    signaling modules (45). The crystal structure of human B7-1

    shows that it forms non-covalently linked homodimers, both

    when free and when bound to CTLA-4 (43, 46). Each mono-

    mer consists of two anti-parallel b-sandwich Ig domains, one

    characteristic of adhesion molecules and the other typical for

    antigen receptors (46).

    The mouse B7-2 protein is a type I transmembrane glyco-

    protein that consists of 309 aa (47). The extracellular regions

    of both mouse and human B7-2 contain highly homologous

    IgV-like and IgC-like domains, while the transmembrane

    regions and the cytoplasmic tails are much less conserved (47,

    48). The crystal structures of the IgV-like receptor-binding

    region of human B7-2 alone and in complex with CTLA-4

    have been solved (42, 49) and show that this domain is a

    two-layer b-sandwich that upon binding to CTLA-4 may form

    a non-covalently linked homodimer. Unlike B7-1, where the

    amino acids at the homodimer interface are mostly hydropho-

    bic, the residues mediating the B7-2 homodimerization are

    highly hydrophilic (49) and might account for the tendency

    of B7-2 to be a monomer in solution (50). This conclusion is

    supported by data from over-expression studies in Chinese

    hamster ovary (CHO) cells using fluorescence energy

    resonance transfer (FRET) measurements (51).

    Expression of CTLA-4, B7-1, and B7-2

    The mRNA for CTLA-4 is not detectable in resting T cells but

    is upregulated within 1 h of TCR stimulation and peaks

    between 24 and 48 h after activation (52, 53). The protein is

    detectable on the surface of T cells within 24 h of stimulation

    (53); however, the majority of the cellular CTLA-4 is present

    in intracellular compartments (54, 55). CTLA-4 has been

    observed in endosomes (56), lysosomes (57), and the trans

    Golgi network (TGN) (58). This intracellular localization

    seems to be predominantly due to the presence of a tyrosine-

    based internalization motif in the cytoplasmic tail of CTLA-4

    that binds the l2 subunit of the clathrin adapter protein-2

    (AP-2) (59–61). Phosphorylation or mutations of Tyr201 inhi-

    bit the binding of l2 and lead to increased levels of CTLA-4

    on the surface (59, 61). As the phosphorylated Tyr201 is also

    part of a Src homology 2 (SH2)-binding motif that mediates

    association with a number of signaling molecules (see below),

    there appears to be an inverse relationship between CTLA-4

    internalization and its ability to generate inhibitory signals

    into T cells.

    TCR ligation and increased intracellular calcium levels cause

    an upregulation of surface CTLA-4 expression, possibly via the

    secretion of CTLA-4-containing secretory lysosomes (56, 57).

    The same Tyr201 is also part of another intracellular trafficking

    motif that binds to the clathrin adapter protein AP-1 and

    mediates transport from the Golgi complex to lysosomes or

    possibly to the plasma membrane (62). More recently, the

    TCR-interacting molecule (TRIM) has been described as a

    chaperone that facilitates CTLA-4 transport from the TGN to

    the surface (58). The observations that the levels of TRIM

    expression correlate with the amount of surface CTLA-4

    suggest that TRIM is an important regulator of CTLA-4 export

    from the TGN to the plasma membrane (58).

    During T-cell migration, the majority of CTLA-4 is present

    in vesicles near the microtubule-organizing center (MTOC) at

    the back of the cell (63). Upon engagement of the TCR, these

    intracellular vesicles re-orient towards the APC with a fraction

    of CTLA-4 eventually localizing to the synapse (63). Interest-

    ingly, the amount of CTLA-4 that translocates to the synapse is

    proportional to the strength of the TCR signal, suggesting that

    CTLA-4 might preferentially restrict T-cell responses to strong

    TCR signals (63).

    The expression of CTLA-4 in effector T cells is under tight

    regulation at the level of synthesis and degradation: in the

    absence of new protein synthesis, the half-life of CTLA-4 is

    about 2 h (63). In contrast, T-regulatory (Treg) cells constitu-

    tively express high levels of CTLA-4 on their surface (64, 65).

    This expression could be due to the fact that these cells appear

    to carry self-reactive TCRs of high affinity (66, 67) that might

    trigger the constant synthesis of CTLA-4. However, the

    molecule appears to be more than a marker of chronic TCR

    stimulation, because the targeted deletion of CTLA-4 specifi-

    cally in mouse Treg cells leads to spontaneous systemic lym-

    phoproliferation and fatal T-cell-mediated autoimmunity (68).

    As already mentioned, CTLA-4 shares its ligands, B7-1 and

    B7-2, with CD28. Initial studies of B7-1 and B7-2 seemed to

    indicate that the molecules have redundant functions in both

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 69

  • costimulation and inhibition (69–71). B7-1 and B7-2 are

    expressed on the same types of cells, including B cells,

    activated T cells, macrophages, and dendritic cells (DCs) (72),

    although B7-2 is expressed earlier than B7-1 on B cells after

    lipopolysaccharide (LPS) stimulation (73) and appears to be

    the major costimulatory ligand on in vitro cultured DCs (74).

    Interestingly, the expression of B7-1 on in vitro generated

    immature DCs appears to be higher than that of B7-2, and this

    pattern correlates with a more tolerogenic phenotype (75).

    This association is noteworthy in light of the fact that B7-1 is

    the major ligand mediating CTLA-4 accumulation at the

    synapse (76), presumably because of the high affinity and

    avidity of this interaction (11).

    Mechanisms of CTLA-4 inhibition

    There have been numerous models proposed for the way in

    which CTLA-4 inhibits T-cell activation. These mechanisms

    can be generally divided into cell-intrinsic (affecting only the

    effector T cell expressing CTLA-4) and cell-extrinsic (affecting

    other cells besides the effectors). A summary of the suggested

    models is below.

    Competition for ligand and inhibitory lattice formation

    The basis for ligand competition as a mechanism for CTLA-4-

    mediated inhibition of T-cell activation lies with the observa-

    tion that the affinities of B7-1 and B7-2 for CTLA-4 are 10- to

    100-fold greater than their affinities for CD28 (10, 11). This

    finding suggests that when CTLA-4 is expressed at the plasma

    membrane after activation and under conditions where ligand

    expression is limiting, CTLA-4 might be able to act as a B7

    sink and effectively inhibit CD28-mediated costimulation by

    ligand sequestration. Support for this model comes from stud-

    ies showing that a CTLA-4 mutant lacking the cytoplasmic tail

    is capable of preventing the multiorgan lymphocytic infiltra-

    tion and early death observed in CTLA-4-deficient mice (77).

    However, the tailless CTLA-4 cannot correct the lymphade-

    nopathy or the accumulation of large numbers of activated

    T cells in older mutant mice, suggesting that CTLA-4 signaling

    or proper cellular trafficking are required for complete inhibi-

    tion of T-cell activation (77). In contrast, experiments using

    Jurkat cells expressing a panel of CTLA-4 mutants have shown

    that a tailless molecule, incapable of delivering a negative signal

    in these cells, is fully able to inhibit IL-2 secretion, and this

    inhibition is directly proportional to the amount of CTLA-4

    on the surface, suggesting that ligand sequestration is a major

    mechanism for CTLA-4-mediated inhibition (50). More recent

    work from our laboratory has led to additional support for the

    idea that CTLA-4 restricts T-cell activation through a CD28-B7-

    dependent mechanism (78). TCR transgenic T cells that over-

    express CTLA-4 show diminished antigen-specific responses

    in vitro; however, this decrease depends on the expression of

    CD28 by the transgenic T cells. In the absence of CD28, the

    CTLA-4-over-expressing T cells respond similarly to littermate

    controls, suggesting that in this model, CTLA-4 functions pre-

    dominantly by competing with CD28 for binding to B7-1 (78).

    A related mechanism for CTLA-4-mediated inhibition was

    suggested by the crystal structures of CTLA-4 and B7-1

    described above. As already mentioned, CTLA-4 is a covalently

    linked homodimer, and the two ligand-binding sites (consti-

    tuted by the M99YPPPY104 motif also present in CD28) were

    observed to lie distally to the homodimer interface (41–43).

    Because B7-1 has been reported to homodimerize in solution

    (50) as well as in the X-ray crystals (46), a molecular model

    has been proposed where a single CTLA-4 dimer can bind two

    B7-1 dimers that can in turn bind two more CTLA-4 dimers,

    and so on, allowing the formation of an extended protein

    lattice that could in theory assemble at the T-cell ⁄APC inter-face (the ‘immunological synapse’) and may be able to disrupt

    its function (43). One study has attempted to assess the

    importance of this putative CTLA-4 lattice for the inhibition

    of Jurkat T-cell activation by generating a ‘monomeric’ CTLA-

    4 by mutating Cys122 that mediates the interchain disulfide

    bond and both N-glycosylation sites in human CTLA-4 (79).

    This mutant molecule is able to traffic to the plasma

    membrane, concentrates at the immunological synapse, binds

    anti-CTLA-4 antibodies and B7-2-Ig, and inhibits IL-2 secre-

    tion. Although compelling, the importance of the results is

    limited by the observation that monomeric B7-2-Ig binding

    can induce the dimerization of the mutant CTLA-4 and by the

    fact that the cell lines expressing this mutant have significantly

    lower amounts of CTLA-4 (79). Consequently, it is possible

    that small numbers of mutant dimers (below the resolution of

    the biochemical experiments) are able to assemble and traffic

    to the immunological synapse, where they can bind ligand,

    form an extended protein lattice, and inhibit T-cell activation

    identically to the wildtype (WT) protein (79).

    Inhibitory signaling

    The idea that CTLA-4 delivers a negative signal in T cells

    can be traced to early experiments showing that anti-CTLA-4

    antibodies are capable of inhibiting the proliferation and IL-2

    secretion of polyclonal T cells stimulated with a combination

    of anti-CD3 and anti-CD28 antibodies in the absence of

    B7-1 or B7-2 (13, 80). In addition, B7-1 can inhibit CD28-

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    70 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • deficient mouse T cells, and anti-CTLA-4 antibodies can

    reverse this inhibition (81), similarly to their effect in the

    accelerated rejection of cardiac allografts in CD28-deficient

    mice due to CTLA-4 blockade (82). The fact that CTLA-4 can

    restrict T-cell activation in the absence of CD28 suggests that

    ligand competition cannot be its only mechanism of function.

    Furthermore, the cytoplasmic tails of mouse and human CTLA-4

    are 100% conserved at the amino acid level (83), suggesting

    that they might bind proteins critical for their function.

    The molecular basis for the CTLA-4-mediated inhibitory

    signaling appears to lie in the association of the CTLA-4

    cytoplasmic tail with the tyrosine phosphatase SH2-domain-

    containing phosphatase 2 (SHP-2) ⁄ SH2-domain-containingtyrosine phosphatase SYP that can dephosphorylate TCR proxi-

    mal signaling molecules like the tyrosine kinases fyn, lck, and

    f chain-associated protein kinase of 70 kDa (ZAP-70) (84).

    Subsequent studies have shown that CTLA-4 can associate with

    the CD3f chain of the TCR in T cells, and the over-expression

    of lck, CD3z and CTLA-4 in transfected human embryonic

    kidney 293 cells improves the association between the last two

    (85). SHP-2 also associates with the CD3f ⁄CTLA-4 complex inprimary T cells, and its over-expression in 293 cells eliminates

    the binding of CTLA-4 and CD3f, as well as the tyrosine phos-

    phorylation of CD3f (85). These data suggest a model where

    TCR engagement leads to the recruitment of lck and the phos-

    phorylation of CD3f, ZAP-70, and linker for activation of

    T cells (LAT), followed by the binding of CTLA-4 and the asso-

    ciated SHP-2, the subsequent dephosphorylation of signaling

    molecules and termination of signaling (85). One caveat of

    these experiments is that SHP-1, SHP-2, and the p85 subunit

    of phosphatidylinositol 3-kinase (PI3K) can associate not only

    with the phosphorylated Tyr201 in the cytoplasmic tail of

    CTLA-4 but also with the tyrosine-phosphorylated tail of

    CD28, making it difficult to envision a way in which the same

    proteins can lead to completely opposite outcomes when

    bound to CD28 versus CTLA-4 (60).

    Another signaling molecule that associates with both CD28

    and CTLA-4 is the serine ⁄ threonine protein phosphatase 2A(PP2A) (86, 87). This protein binds a conserved three-lysine

    motif in the membrane-proximal region of the cytoplasmic

    tail of CTLA-4 and represses its inhibitory function by a

    currently unknown mechanism (87). As PP2A also binds to

    CD28 and inhibits its function (86), it is possible to speculate

    that perhaps one way in which CTLA-4 inhibits T-cell activa-

    tion is by bringing the phosphatase in proximity to CD28,

    where it can repress CD28-mediated costimulatory signaling.

    More recent reports that CTLA-4 and CD28 have opposite

    effects on the surface expression of lipid rafts (88) have added

    yet another facet to the inhibitory signals potentially triggered

    by CTLA-4 engagement. Because the rafts are enriched in

    signaling molecules and are critical for early TCR signal

    transduction (89), small changes in their expression can have

    profound effects on T-cell activation. Furthermore, CTLA-4 is

    found in lipid rafts at the plasma membrane, and this associa-

    tion is required for its inhibitory function (90), presumably

    because it negatively regulates the amount of phosphorylated

    TCR present in these domains (91). The raft localization of

    CTLA-4 depends on its cytoplasmic tail (90), suggesting that

    the tailless mutants described earlier might have additional

    defects, besides their inability to associate with signaling

    components.

    The importance of ligand sequestration in CTLA-4 function

    has been recently questioned by the discovery of a mouse CTLA-

    4 isoform that lacks the extracellular domain and thus functions

    in a ligand-independent manner (38, 39). The reduced expres-

    sion of this isoform correlates with increased susceptibility to a

    mouse model of type I diabetes (38), and the protein itself

    appears capable of generating negative signals in T cells by bind-

    ing and dephosphorylating CD3f (39). Similarly, a CTLA-4

    mutant that lacks B7-1 and B7-2 binding has been shown to

    inhibit proliferation, cytokine production, and TCR-mediated

    activation of the extracellular signal regulated kinase (ERK) in

    T cells lacking endogenousCTLA-4 (92). Thismutant also delays

    the rampant lymphoproliferation and early death of CTLA-

    4-deficient mice, suggesting that some negative signals can

    occur in the absence of ligand binding; however, the fact that

    100% of the mice die at 14 weeks of age suggests that ligand

    binding is critical for CTLA-4 function (92).

    Reversal of the TCR-mediated STOP signal

    A more recent model for CTLA-4 function has emerged from

    in vitro migration and in vivo imaging experiments showing that

    the antibody-mediated ligation of CTLA-4 increases the

    motility of polyclonal CD4+ T cells and CTLA-4-positive TCR

    transgenic T cells fail to stop in response to antigen, unlike

    their CTLA-4-negative counterparts (93). These results suggest

    that CTLA-4 might be able to reverse the STOP signal gener-

    ated by TCR engagement and thus prevent T-cell activation.

    One caveat of the in vivo experiments is that the CTLA-4-

    positive population probably contains Treg cells that might

    have higher mobility than effector T cells, especially because

    they have been shown to inhibit stable contacts between

    effector T cells and DCs (94, 95).

    In an attempt to demonstrate that CTLA-4 expression by

    itself is sufficient for the reversal of the TCR STOP signal, the

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 71

  • same group investigated the motile behavior of a mouse

    hybridoma that was transfected with human CTLA-4 (96).

    In these experiments, the CTLA-4-positive cells had shorter

    contact times with APCs, formed smaller contact areas in the

    immunological synapse, and had reduced Ca2+ influx and IL-2

    secretion. The data also showed that CTLA-4 disrupted

    ZAP-70 microcluster formation, suggesting that CTLA-4

    might reduce the interactions between T cells and APCs as a

    way of inhibiting T-cell activation (96).

    Treg function

    The idea that CTLA-4 is important for the function of Treg

    cells has been a subject of controversy due to conflicting

    results in different model systems. Early studies showed that

    recombination-activating gene (RAG)-deficient mice reconsti-

    tuted with a mixture of CTLA-4-deficient and normal bone

    marrow cells were healthy, even though similar chimeras

    containing only CTLA-4) ⁄ ) cells died 10 weeks after reconsti-

    tution (97). These results were originally interpreted to mean

    that a factor secreted by normal T cells could compensate for

    the defect of CTLA-4-deficient cells; however, the discovery

    that Treg cells can suppress effectors lacking CTLA-4 and anti-

    CTLA-4 antibodies abrogate this suppression (64) provides an

    alternative explanation for the results. The findings suggest

    that CTLA-4-sufficient Treg cells can inhibit the proliferation

    of CTLA-4-deficient effectors and that CTLA-4 is crucial for

    this process (64). The importance of CTLA-4 for Treg func-

    tion is reinforced by a study of a mouse model of colitis,

    where Treg cells can prevent the intestinal inflammation

    mediated by the transfer of CD4+CD45RBhigh T cells, and this

    inhibition is abrogated by CTLA-4 blockade (65).

    In contrast, the significance of CTLA-4 for the function of

    Treg cells is brought into question by the observation that

    CTLA-4-deficient mice have normal Treg development and

    homeostasis and are capable of suppressing effector T cells

    in vitro (98). The fact that the CTLA-4) ⁄ ) Treg cells produce

    increased amounts of inhibitory cytokines like IL-10 and

    transforming growth factor b (TGFb) suggests that in the

    absence of CTLA-4, these cells can develop compensatory

    suppressive mechanisms (98). Similarly, in a mouse model

    of colitis, CTLA-4-deficient Treg cells are able to suppress

    inflammation, predominantly via the secretion of IL-10 (99).

    However, CTLA-4-sufficient Treg cells are able to suppress the

    colitis induced by CTLA-4-deficient CD4+CD45RBhigh cells

    from mice lacking B7-1 and B7-2, and anti-CTLA-4 antibodies

    abrogate this inhibition, suggesting that CTLA-4 is critical for

    the function of WT Treg cells (99).

    Most recently, experiments using mice that have a condi-

    tional deletion of CTLA-4 specifically in Treg cells have shown

    that the molecule is required for Treg function (68). The

    mutant mice spontaneously develop systemic lymphoprolifer-

    ation, hyperproduction of IgE, and die of T-cell-mediated

    autoimmunity 10 weeks after birth, while also exhibiting

    some tumor immunity (68). The CTLA-4) ⁄ ) Treg cells

    develop and survive normally but are highly proliferative and

    comprise up to half of the CD4+ T cells in these mice, suggest-

    ing that CTLA-4 has an important cell-autonomous inhibitory

    function in Treg cells, similarly to its function in effector

    T cells (68). The CTLA-4-deficient Treg cells are impaired in

    their ability to suppress the proliferation of effector T cells

    in vitro, potentially because they are unable to downregulate the

    expression of B7-1 and B7-2 on APCs (68). The latter finding

    provides a possible alternative mechanism for the way in

    which Treg cells can inhibit effector T-cell proliferation

    besides the induction of indoleamine 2,3-dioxygenase (IDO),

    discussed below. While compelling, the study does not address

    the possibility that CTLA-4 signaling in Treg cells might be also

    important for the suppressive function of these cells.

    Recent work from our laboratory has attempted to assess

    the relative significance of CTLA-4 on effector versus Treg

    cells through the use of antibodies that block the function of

    the molecule selectively in the two types of cells (Peggs KS,

    Quezada SA, Chambers CA, Korman AJ, JP Allison, unpub-

    lished data). Compartment-specific blockade is achieved by

    the generation of mice that express human CTLA-4 under the

    control of the mouse promoter and the use of antibodies

    specific to the mouse protein. The data show that in a mouse

    model of melanoma, the blockade of CTLA-4 on the Treg

    compartment alone is insufficient for tumor protection, while

    the blockade of CTLA-4 on effector T cells is able to protect

    about 40% of the mice; however, the simultaneous blockade

    of CTLA-4 function on both effector and Treg cells is most

    effective and cures 70% of the mice. These results suggest that

    CTLA-4 has important inhibitory roles in both effector and

    Treg cells, and successful immunotherapies should target both

    its cell-autonomous inhibition of effector T-cell activation, as

    well as its cell-extrinsic function via Treg cells.

    CTLA-4-mediated ‘reverse’ signaling, IDO, and tryptophan

    metabolism

    The idea that IDO is important for peripheral tolerance comes

    from experiments showing that treatment of pregnant mice

    with the IDO inhibitor 1-methyltryptophan (1-MT) triggers

    the rejection of allogeneic concept (100). It has been

    suggested that a similar mechanism might be responsible for

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    72 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • the survival of pancreatic islet allografts after CTLA-4-Ig treat-

    ment of diabetic mice. This hypothesis has been supported by

    experiments showing that the tolerogenic effects of CTLA-4-

    Ig administration are reversed by 1-MT treatment and that

    B7-1 and B7-2 expression on DCs is required for the success

    of the CTLA-4-Ig therapy (101). In this model, CTLA-4-Ig

    appears to bind B7-1 or B7-2 on DCs and triggers a signaling

    cascade involving signal transducer and activator of transcrip-

    tion 1 (STAT1), p38 mitogen-activated protein kinase

    (MAPK), and nuclear factor jB (NF-jB) that leads to inter-

    feron-c (IFN-c) secretion by the DCs. IFN-c then elicits the

    production of IDO, which degrades tryptophan and results in

    the inhibition of T-cell proliferation and the induction of

    apoptosis (101). The additional observation that IL-10 pre-

    vents the IFN-c-induced downregulation of IDO in human

    DCs (102) and the fact that Treg cells secrete IL-10 (103) sug-

    gest the existence of a regulatory mechanism where the

    engagement of B7-1 on DCs by CTLA-4 on Treg cells may

    trigger IDO production by tolerogenic DC and the possibility

    that this secretion may be further enhanced by Treg-generated

    IL-10. This hypothesis has been supported by a study showing

    that mouse Treg cells elicit IDO activity in DCs, and this pro-

    cess is abrogated by anti-CTLA-4 antibodies, the absence of

    B7-1 and B7-2 on the DCs, or the inability of the DCs to make

    IFN-c (104).

    The idea that CTLA-4 might be able to ‘reverse’ signal

    into APCs via B7-1 is reinforced by experiments showing

    that CTLA-4-Ig administration extends the long-term cardiac

    allograft survival in mice, and the success of the treatment

    depends on the expression of B7-1 by the allograft donor

    but not by the recipient (105, 106). Although the initial

    interpretation of these results consisted of a speculation that

    CTLA-4-Ig is an incomplete blocking agent leaving some

    unbound B7-1 in the WT donors that can engage CTLA-4

    and inhibit the effector T cells mediating the graft rejection,

    an alternative hypothesis is that CTLA-4-Ig binds B7-1 on

    the DCs in the allograft, triggers IDO secretion, and induces

    tolerance.

    Another example of reverse signaling by CTLA-4 has been

    reported by a study showing that B7-1 and B7-2 expression

    on effector T cells is required for their suppression by Treg

    cells in vitro and in vivo (107). The lentiviral expression of full

    length B7-1 or B7-2 but not mutant molecules lacking the

    transmembrane and cytoplasmic domains in B7-1) ⁄ )B7-2) ⁄ )

    effector cells restores their susceptibility to Treg-mediated

    inhibition, suggesting that signals originating from B7-1 or

    B7-2 engagement on the effectors, presumably by CTLA-4 on

    Treg cells, are responsible for this suppression (107).

    CTLA-4 involvement in tolerance, autoimmunity, and

    cancer progression

    The early demonstration that CTLA-4-Ig potently inhibits

    in vitro T-cell activation (108) suggested that it might be

    useful as a therapeutic agent in autoimmune disease and

    in transplantation. Subsequent studies have shown that

    CTLA-4-Ig can block autoantibody production in a mouse

    model of systemic lupus erythematosus and can extend

    the life span of the affected animals even when treatment

    is delayed until the most advanced stages of disease

    (109). Furthermore, CTLA-4-Ig induces long-term survival

    of xenogeneic pancreatic islet grafts in mice (110) and

    suppresses T-cell-dependent antibody responses to multiple

    antigens in vivo (111). Strikingly, CTLA-4-Ig treatment of

    CTLA-4-deficient mice from birth prevents the fatal lym-

    phoproliferation observed in these animals, even though

    disease develops shortly after cessation of therapy (112).

    These results are consistent with a study showing that the

    genetic ablation of B7-1 and B7-2 abrogates the lympho-

    proliferative disorder in CTLA-4-deficient mice and admin-

    istration of anti-CD28 antibodies to these animals initiates

    disease, despite having no effect on CTLA-4-sufficient

    mice (113). These studies suggest that the main mecha-

    nism by which CTLA-4-Ig inhibits T-cell immune

    responses is by blocking CD28-mediated costimulatory

    signals triggered by B7-1 and B7-2 engagement, although

    it is also possible that it stimulates IDO production by

    DCs in grafts, as discussed above. Most relevantly to

    patients, CTLA-4-Ig has been approved for treatment of

    moderate-to-severe rheumatoid arthritis (RA) by the United

    States Food and Drug Administration (FDA). Randomized

    clinical trials have shown that CTLA-4-Ig (abatacept) is

    well-tolerated, reduces the signs and symptoms of RA,

    slows the progression of joint damage, and improves the

    physical function of adults that have had inadequate

    responses to at least one other anti-rheumatic drug

    (reviewed in 114).

    The potent inhibitory effects that CTLA-4 exerts on T-cell

    activation render it an attractive target for disease settings

    where immune responses are limited or missing. One such

    example is the case of tumors that do not provide suffi-

    cient T-cell costimulatory signals and remain invisible to

    the immune system. Early studies from our laboratory have

    shown that treatment with anti-CTLA-4 antibodies leads to

    the rejection of established colon carcinoma and fibrosar-

    coma in mice, and this rejection creates immunity to

    subsequent challenges with tumor cells, suggesting the

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 73

  • existence of long-term immunological memory (115).

    Following these initial experiments, multiple studies have

    shown that the anti-CTLA-4 antibody therapy is effective

    for treatment of other cancers including prostatic carci-

    noma (116), ovarian carcinoma (117), and B and T lym-

    phomas (118, 119). However, CTLA-4 blockade is

    unsuccessful as a single-agent therapy in cases involving

    poorly immunogenic tumors like B16 melanoma (120)

    and SM1 mammary carcinoma (121). For effective treat-

    ment of established tumors in both cases, CTLA-4 blockade

    has to be combined with an irradiated cellular vaccine

    engineered to produce granulocyte-macrophage colony-

    stimulating factor (GM-CSF), which is thought to enhance

    antigen presentation (120, 121). A similar combination of

    a cellular vaccine and anti-CTLA-4 antibody therapy is also

    successful at preventing spontaneous prostate cancer devel-

    opment in TRAMP (transgenic adenocarcinoma of mouse

    prostate) mice (122).

    A recent analysis of the effects of the combination therapy

    of GM-CSF and anti-CTLA-4 antibodies on the effector and

    regulatory T-cell compartments in the B16 mouse model of

    melanoma has shown that the treatment acts via a cell-autono-

    mous mechanism on both kinds of T cells, allowing them to

    expand without depleting or permanently impairing the func-

    tion of Treg cells (123). The major change in the lymphocytic

    infiltration of the tumors induced by the combination therapy

    consists of an increase in the ratio of T effector-to-Treg cells,

    and this increase directly correlates with tumor rejection,

    suggesting that Treg cells restrict the anti-tumor function of

    both CD8+ and CD4+ effectors (123). The recent demonstra-

    tion that this combination therapy synergizes with prophylac-

    tic but not therapeutic Treg depletion and the finding that the

    success of the treatment is limited by the inability of the effec-

    tor cells to infiltrate the tumor, despite the generation of

    strong systemic anti-tumor immune responses, has important

    implications for the current clinical strategies used for treat-

    ment of established tumors in patients (124).

    In addition to the successful combination of anti-CTLA-4

    antibodies and GM-CSF for treatment of poorly immunogenic

    tumors, CTLA-4 blockade has been shown to synergize with

    a number of therapies that enhance antigen presentation or

    activate the innate immune system like peptide and DNA

    vaccination, chemotherapy, radiation, Toll-like receptor (TLR)

    engagement, and high-dose IL-2 (reviewed in 125). A

    detailed summary of the available data from pre-clinical

    studies and clinical trials of CTLA-4 as a monotherapy or

    in combination with other treatments has been recently

    presented and is beyond the scope of this review (125).

    PD-1, PD-L1 (B7-H1), and PD-L2 (B7-DC)

    PD-1 is a relatively new member of the extended

    CD28 ⁄ CTLA-4-family of T-cell regulatory molecules that wasoriginally identified as a gene that was highly expressed by cell

    lines undergoing programmed cell death (20). It has two

    ligands, PD-L1 (B7-H1) and PD-L2 (B7-DC), which are

    distantly related to B7-1 and B7-2 (21–23).

    Protein structure

    PD-1 is a 288 a.a. type I transmembrane glycoprotein that

    contains a single IgV-like domain in its extracellular region

    (20, 126). The cytoplasmic tail contains an immunoreceptor

    tyrosine-based inhibitory motif (ITIM) (20) and an immuno-

    receptor tyrosine-based switch motif (ITSM) (127). The

    protein lacks the membrane-proximal cysteine that mediates

    the inter-chain disulfide bond in the CD28 and CTLA-4

    homodimers (126), and as a result, PD-1 is monomeric both

    in solution and on the surface of transfected cells, as deter-

    mined by FRET (128). Multiple splice isoforms have been

    reported for human PD-1, some of which encode soluble

    PD-1 molecules that are elevated in the peripheral blood and

    synovial fluid of RA patients (129), and others may code for

    ligand-independent variants (130). X-ray crystallography has

    shown that the monomer is a two-layer b-sandwich that

    forms a structure closely resembling the IgV-like fold of

    CTLA-4, despite the fact that the sequence identity between

    the two extracellular domains is only 20% (128).

    PD-L1 is a 290 aa type I transmembrane glycoprotein that

    contains a membrane-distal IgV-like and a membrane-

    proximal IgC-like domain in its extracellular region (21, 22).

    The cytoplasmic tail is 30 aa long and has no known signaling

    motifs but is highly conserved between humans and mice

    (22). The crystal structures of human PD-L1 alone and in

    complex with mouse PD-1 have been solved (131). PD-L1

    binds to PD-1 via its IgV-like domain in a 1:1 receptor ⁄ ligandstoichiometry (131). The binding interface is different from

    that of the CTLA-4 ⁄B7-1 complexes and predominantlyinvolves the faces of the IgV-like domains, similarly to the

    antigen-binding regions of antibodies and TCRs (131).

    Mouse PD-L2 is a 248 aa type I transmembrane glycopro-

    tein that contains a membrane-distal IgV-like and a mem-

    brane-proximal IgC-like extracellular domain and a

    cytoplasmic tail of only 4 aa (23). The human PD-L2 protein

    is 26 aa longer, because it lacks the premature STOP codon

    present in the mouse (23). The crystal structures of mouse

    PD-L2 alone and in complex with mouse PD-1 have shown

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    74 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • that PD-L2 binds to PD-1 via its IgV-like domain in a 1:1

    receptor ⁄ ligand stoichiometry (132). The ‘face-to-face’ bind-ing interface is similar to the one between PD-1 and PD-L1,

    and the acute angle between PD-1 and its ligands may explain

    the need for the relatively long ‘stalk’ regions separating the Ig

    domains and the transmembrane segments relative to the

    homologous structures in CTLA-4 and B7-1 (132).

    Expression of PD-1, PD-L1, and PD-L2

    PD-1 is not detectable on naive T cells but its expression goes

    up in T cells, B cells, and myeloid cells after activation (22,

    133). PD-1 expression is also upregulated by the treatment of

    purified human T cells with the common c chain cytokines

    IL-2, IL-7, IL-15, and IL-21 in the absence of TCR ligation

    (134). PD-1 has been also found on the surface of TCR-stimu-

    lated Treg cells, as well as in an intracellular compartment in

    these cells prior to activation (135). In activated primary

    T cells, PD-1 is present at the plasma membrane and in

    Golgi-proximal intracellular vesicles that do not appear to be

    recycling endosomes or lysosomes (136). The expression of

    PD-1 is particularly high on the surface of functionally

    exhausted CD8+ effector T cells in multiple persistent viral

    infections in both mice and humans (137–140).

    The two PD-1 ligands differ significantly in their expression

    patterns. PD-L1 is broadly expressed on both hematopoietic

    and non-hematopoietic cells. It has been detected on resting

    mouse T cells, B cells, DCs, macrophages, natural killer (NK)

    cells, and bone marrow-derived mast cells, and its levels

    increase after activation (141, 142). PD-L1 is also expressed

    by cells in the heart endothelium and the small intestine, by

    syncyciotrophoblasts in the placenta, as well as by islet cells in

    the pancreas of naive mice (143). In mouse macrophages,

    PD-L1 expression is upregulated by LPS and IFN-c, as well as

    the addition of T-helper type 1 (Th1) cells in a process that

    requires TLR4 and STAT1 (144).

    Unlike PD-L1, the expression of PD-L2 is restricted to

    hematopoietic cells. Various cytokines can induce PD-L2

    on DCs, macrophages, bone marrow-derived mast cells, and

    B1 B cells (142, 143, 145, 146). In mouse macrophages,

    PD-L2 expression is upregulated by IL-4 and the addition of

    Th2 cells in a process that requires the IL-4 receptor a and

    STAT6 (144).

    Molecular mechanisms of PD-1 inhibitory signaling

    The initial indications that PD-1 may engage an inhibitory

    pathway upon binding to its ligands came from studies show-

    ing that PD-L1-Ig and PD-L2-Ig can restrict the proliferation

    and cytokine secretion of WT but not PD-1-deficient T cells

    (22, 23). The fact that the inhibition is greatest at lower

    amounts of antigen suggests that PD-1 might be more effec-

    tive at attenuating weak TCR signals rather than strong ones

    (22, 23). The formal demonstration that PD-1 might in fact

    deliver inhibitory signals in the cells that express it came from

    studies of B-cell lines transfected with chimeric molecules that

    fused the extracellular region of the IgG Fc receptor type IIB

    to the PD-1 cytoplasmic tail (147). The simultaneous ligation

    of the B-cell receptor (BCR) and the fusion protein results in

    growth inhibition, impairs Ca2+ mobilization, and reduces

    tyrosine phosphorylation of several effector molecules (147).

    Mutagenesis studies have shown that these inhibitory effects

    do not require the N-terminal tyrosine in the cytoplasmic tail

    but depend on the C-terminal tyrosine because upon

    phosphorylation, that amino acid recruits the tyrosine phos-

    phatase SHP-2 to the BCR and leads to the dephosphorylation

    of proximal signaling molecules (147).

    The second cytoplasmic tyrosine has subsequently been

    identified as part of an ITSM, while the first one appears part

    of an ITIM (127), and similarly to the results from B cells,

    only mutations of the C-terminal tyrosine reverse the PD-1-

    mediated inhibition of proliferation and cytokine secretion of

    primary human T cells (148, 149). Both SHP-1 and SHP-2

    co-immunoprecipitate with the cytoplasmic tail of PD-1 upon

    stimulation of these cells with pervanadate, but only SHP-2

    appears to associate with PD-1 in the absence of activation

    (148). Despite this constitutive interaction, the inhibition of

    T-cell functions requires the simultaneous engagement of the

    TCR and PD-1 (148). The engagement of PD-1 by PD-L1-Ig

    in conjunction with TCR ligation has been shown to decrease

    ERK and protein kinase C h (PKCh) phosphorylation and IL-2

    secretion from human T-cell blasts, as well as to inhibit the

    phosphorylation of ZAP-70 and CD3f in Jurkat cells (150).

    Sequencing of the proteins that are bound to the two

    phosphotyrosines in the cytoplasmic tail of PD-1 from Jurkat

    cell lysates has shown that lck can bind to the first but not the

    second tyrosine, SHP-1 and the c-src tyrosine kinase (Csk) can

    bind to the second but not the first, and SHP-2 can bind to

    both (150). Biochemical studies using primary human T cells

    have shown that PD-1 and CTLA-4 inhibit T-cell activation by

    distinct mechanisms. While ligation of both PD-1 and CTLA-4

    reduces the phosphorylation of the serine ⁄ threonine kinaseAkt in response to TCR ⁄CD28 stimulation, the inhibition viaCTLA-4 but not via PD-1 depends on the function of PP2A,

    because it is sensitive to okadaic acid, while PD-1 inhibits Akt

    phosphorylation by preventing the CD28-mediated activation

    of PI3K (149). In general, the data from these studies suggest

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 75

  • that PD-1 can associate with SHP-2 and perhaps more weakly

    with SHP-1, and upon ligand engagement, it can recruit these

    phosphatases to the immunological synapse (136), where

    they can interfere with T-cell activation by inhibiting PI3K

    and the subsequent phosphorylation and activation of Akt.

    The relatively recent discovery that PD-L1 can bind not only

    PD-1 but also B7-1 in both mouse and human cells (151,

    152) introduces an additional level of complexity in the

    interaction between the costimulatory molecules and the

    inhibitory pathways that they control. The affinity of the asso-

    ciation of PD-L1 and B7-1 is higher than that between B7-1

    and CD28 and lower than that between B7-1 and CTLA-4 and

    between PD-L1 and PD-1 (151, 152). The fact that upon

    engagement by the other molecule, both PD-L1 and B7-1

    appear able to deliver inhibitory signals in the T cells that

    express them (151) blurs the current definitions of receptors

    and ligands and demands the characterization of the signaling

    pathways mediated by these two proteins.

    PD-1 involvement in tolerance, autoimmunity, cancerprogression, and viral immunity

    The importance of PD-1 for the prevention of autoimmunity

    was initially indicated by the phenotype of PD-1-deficient

    mice. C57 ⁄ Bl6 mice lacking PD-1 develop lupus-like arthritisand glomerulonephritis with predominant deposition of IgG3

    antibodies (153), while PD-1-deficient BALB ⁄ c mice developautoimmune-dilated cardiomyopathy that leads to congestive

    heart failure and premature death (154). The heart disease is

    caused by the accumulation of antibodies specific for cardiac

    troponin I that causes the chronic stimulation of Ca2+ influx

    in cardiomyocytes (155). PD-1-deficient animals on the

    non-obese diabetic (NOD) background develop autoimmune

    disease with accelerated onset and increased frequency

    because of strong Th1 polarization of the T cells infiltrating

    the affected islets (156). Similarly, PD-L1-deficient animals

    display enhanced T-cell responses and are more susceptible to

    experimental autoimmune encephalomyelitis (EAE) and

    experimental autoimmune hepatitis (157, 158). In contrast,

    one study of PD-L2-deficient mice has shown that these mice

    have diminished Th1 and CTL responses (159), while another

    line of PD-L2) ⁄ ) mice displays increased T-cell activation after

    in vivo immunization and is unable to generate tolerance in

    response to an oral antigen (160). These results suggest that

    PD-1 and both its ligands are important for the establishment

    and ⁄or maintenance of peripheral tolerance.The role of PD-1 and its ligands in regulating autoimmune

    disease has been investigated in multiple mouse models. In

    EAE induced by immunization with myelin oligodendrocyte

    glycoprotein (MOG), anti-PD-1, and anti-PD-L2 but not anti-

    PD-L1 antibodies cause more severe disease as a result of

    increased frequencies of IFN-c-producing T cells, higher lev-

    els of serum anti-MOG antibodies, and elevated delayed-type

    hypersensitivity responses (161). The effects of anti-PD-1,

    anti-PD-L1, and anti-PD-L2 antibody blockade on EAE suscep-

    tibility and disease progression differ among mouse strains,

    suggesting that all three molecules can limit autoimmune

    responses, and their relative significance for disease manifesta-

    tion may be influenced by differences in the cytokine milieu

    under the various experimental conditions (162, 163).

    The importance of PD-1 and PD-L1 but not PD-L2 for the

    regulation of autoimmune diabetes in NOD mice has been

    emphasized by multiple reports. Studies using blocking anti-

    bodies to the molecules have shown that PD-1 and PD-L1 are

    required for prevention of diabetes, presumably because

    PD-L1 but not PD-L2 is expressed on inflamed islets of NOD

    mice (164). Furthermore, the expression of PD-L1 by paren-

    chymal cells rather than hematopoietic cells in the pancreas is

    required for protection against immunopathology after trans-

    plantation of syngeneic islets in diabetic recipients (165). The

    PD-L1-mediated inhibition of autoreactive T cells appears to

    involve PD-1, because PD-1-deficient ovalbumin-specific OT-I

    CD8+ T cells cause diabetes in mice expressing ovalbumin in

    the pancreas, while WT OT-I T cells do not (166). Finally, the

    importance of the PD-1 ⁄ PD-L1 inhibitory pathway is empha-sized by a report showing that insulin-coupled APCs can be

    successfully used as a therapy after diabetes onset, and anti-

    body-mediated blockade of PD-1 and PD-L1 reverses these

    therapeutic effects in a process that seems to involve the

    re-activation of pathogenic effector cells without affecting

    Treg activity (167). In contrast, the constitutive ectopic

    expression of PD-L1 on pancreatic islets leads to spontaneous

    diabetes that can be reversed by low doses of anti-CD3

    antibodies, suggesting that the disease is T-cell mediated

    (168). Further experiments are needed to determine the

    reasons for this discrepancy.

    PD-1 and its ligands have also been shown to be involved in

    fetomaternal tolerance (169), the regulation of alloimmune

    responses (170–172), graft-versus-host disease (173), asthma

    (174, 175), and experimental allergic conjunctivitis (176). In

    addition, the early observation that multiple tumor lines

    express PD-L1 (177, 178) has prompted the investigation of

    the potential role that PD-1 and its ligands might have in the

    regulation of tumor immunity. In humans, high PD-L1

    expression by tumor cells correlates with poor prognosis in

    ovarian cancer (179), renal cancer (180–182), urothelial

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    76 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • carcinoma (183, 184), pancreatic cancer (185), non-small-

    cell lung cancer (186), and gastric carcinoma (187). The

    mechanism by which tumor-associated PD-L1 might protect

    tumors from T-cell-mediated immune destruction has been

    alternatively suggested to involve the induction of T-cell death

    (177) or by making the tumor cells resistant to T-cell-medi-

    ated apoptosis (188). In contrast, PD-L2 has been suggested

    to promote tumor immunity via a PD-1-independent mecha-

    nism (189), which may potentially involve the PD-L2 engage-

    ment of a currently unknown costimulatory receptor on

    T cells that might also bind PD-L1 (21, 159, 190, 191).

    PD-1 or PD-L1 blockade has been shown to mediate some

    tumor protection in mice (178, 192–194), and a humanized

    anti-PD-1 antibody, CT-011, is currently in phase I clinical

    trials for treatment of patients with advanced hematologic

    malignancies (195). The antibody appears to be well tolerated

    and has had clinical benefit in 33% of patients with one

    complete remission (195). These results suggest that targeting

    of the PD-1 ⁄ PD-L1 pathway is a promising new strategy fortumor therapy that might be able to synergize with existing

    anti-cancer treatments, including CTLA-4 blockade, especially

    in light of the distinct inhibitory mechanisms employed by

    these molecules in T cells.

    The relatively recent discovery that PD-1 is upregulated on

    functionally exhausted CD8+ effector T cells during persistent

    viral infections renders it an attractive target for therapeutic

    vaccination in chronic infections. Treatment of mice that

    harbor chronic lymphocytic choriomeningitis virus (LCMV)

    infection with anti-PD-L1 antibodies restores the ability of the

    exhausted CD8+ T cells to proliferate, secrete cytokines, kill

    infected targets, and decrease viral load in the animals (137).

    Similarly, PD-1 is expressed at high levels on non-functional T

    cells during human immunodeficiency virus (HIV) infections,

    and anti-PD-1 or anti-PD-L1 antibodies appear able to restore

    their proliferation and effector functions, at least in vitro (138–

    140). Comparable findings have been observed during chronic

    infections with hepatitis B and C viruses (196–200), herpes

    simplex virus (201), Helicobacter pylori (202), and Mycobacterium

    tuberculosis (203). These reports suggest that PD-1 and its ligands

    are potentially attractive therapeutic targets for the treatment of

    persistent viral and bacterial infections in human patients.

    B7-H3 and B7x

    B7-H3 and B7x (B7-H4) are distantly related to B7-1 and

    B7-2 and appear to bind to a receptor on activated but not

    naive T cells (24, 26–28). B7-H3 has been shown to

    alternatively activate (24) or inhibit (25) T-cell function,

    presumably via distinct receptors, while B7x is a negative reg-

    ulator of T-cell activation (26–28). The costimulatory recep-

    tor for B7-H3 has been recently identified as the triggering

    receptor expressed on myeloid cells (TREM)-like transcript 2

    (TLT-2 ⁄TREML2), which is present constitutively on CD8+

    T cells and on activated CD4+ T cells (204). The inhibitory

    receptors that B7-H3 and B7x bind are currently not known.

    Protein structure and expression of B7-H3 and B7x

    B7-H3 is a 316 a.a. type I transmembrane glycoprotein that

    contains an IgV-like and an IgC-like domain in its extracellular

    region and a cytoplasmic tail of 45 a.a. (24, 205, 206). In

    humans, there is also an alternatively spliced 4Ig-B7-H3

    variant of 534 a.a. containing two IgV-like and two IgC-like

    domains that appear to have arisen via gene duplication

    (205–207). This isoform may protect tumor cells from NK

    cell-mediated lysis (208). B7x is 282 a.a. glycoprotein that

    contains a single IgV-like and a single IgC-like domain in its

    extracellular region and has a very short cytoplasmic tail

    (26–28) or may be attached to the membrane via a glyco-

    phosphatidylinositol (GPI) linkage (27).

    Unlike B7-1 and B7-2, B7-H3 and B7x are ubiquitously

    expressed in most organs, although the exact cell types express-

    ing the molecule in these tissues are currently unclear. B7-H3

    mRNA has been found in the prostate, testes, uterus, small and

    large intestine, placenta, heart, lung, liver, kidney, pancreas, as

    well as in the spleen, thymus, and lymph nodes (24, 205). The

    B7-H3 protein is found on monocytes and DCs and is induced

    on T cells, B cells, and NK cells (24, 207). B7x mRNA has been

    found in the lung, liver, prostate, testes, pancreas, placenta,

    uterus, brain, kidney, as well as in the thymus, and spleen

    (26–28, 209). The B7x protein has been detected on in vitro

    activated T cells, B cells, peritoneal macrophages, monocytes,

    and DCs (26–28). B7x has been also suggested to be a marker

    for a novel suppressive macrophage population in human ovar-

    ian carcinoma (210), and its expression may be induced by

    Treg-mediated IL-10 secretion (211), although this proposed

    mechanism is somewhat controversial (212).

    Analysis of B7-H3-deficient mice has shown that the protein

    has a role in limiting Th1-mediated immune responses,

    because the animals show enhanced Th1-mediated airway

    hypersensitivity, they accumulate higher concentrations of

    anti-DNA antibodies and are more susceptible to EAE (205).

    In contrast, B7x-deficient mice appear to have mildly elevated

    Th1 responses against Leishmania major leading to decreased

    parasite burden but otherwise have unaltered hypersensitivity

    reactions in the airway and the skin and mount normal T-cell

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 77

  • responses against LCMV (213). These results suggest that

    while B7x has a negative regulatory function in vivo, the

    existence of multiple inhibitory mechanisms for the control of

    T-cell activation may limit the absolute significance of each

    individual inhibitory pathway.

    B7-H3 and B7x in cancer progression

    Consistent with the inhibitory effects of B7x on T-cell activa-

    tion (26–28) and despite the conflicting descriptions of

    B7-H3 as a costimulatory or an inhibitory ligand for T-cell

    function (24, 25, 214), multiple studies have found that B7x

    and B7-H3 expression in tumors correlates with poor progno-

    sis. In non-small-cell lung cancer, high B7x and B7-H3 levels

    correlate with decreased number of tumor-infiltrating lym-

    phocytes (TILs) and increased lymph node metastasis (215).

    In prostatic carcinoma, high levels of B7x and B7-H3 are

    associated with increased risk of cancer recurrence, tumor

    progression after surgery, and cancer-related death (216,

    217). In clear cell renal cell carcinoma (RCC), B7x and B7-H3

    are expressed by the tumor or the tumor vasculature, and this

    expression is significantly correlated with increased risk of

    death from cancer (218, 219). Similarly, serum-soluble B7x is

    elevated in RCC patients and correlates with advanced stage of

    disease (220). B7x is also highly expressed in ductal and lobu-

    lar breast cancer (221), and soluble B7x is a potential biomar-

    ker for ovarian carcinoma (222). These findings suggest that

    while the molecular details of the inhibitory pathways that

    B7x and B7-H3 engage in T cells are currently not known, the

    proteins can be useful prognostic tools in human cancer

    and potential targets for antibody-mediated tumor immuno-

    therapy.

    BTLA, HVEM, and CD160

    Protein structure

    BTLA is type I membrane glycoprotein of the IgSF that was

    first identified as an expressed sequence tag (EST) in

    Th1-polarized TCR transgenic T cells (29) and independently

    as an EST expressed after activation of double-positive thymo-

    cytes (223). Crystal structures of human and mouse BTLA

    reveal a single b-sandwich Ig domain containing two b-sheets

    (224, 225). BTLA is related to IgV receptors such as CD28 and

    CTLA-4, but the structural data highlight notable differences.

    On one of the b-sheets BTLA lacks a strand present in CTLA-4,

    and thus belongs in the Ig intermediate (IgI) rather than the

    IgV subset of the IgSF (224). BTLA binds to the transmem-

    brane receptor HVEM (30, 226) via a region on the IgI

    domain that is distinct from the MYPPPY motif-containing

    surface through which CD28 and CTLA-4 bind to their B7-1

    and B7-2 ligands (42, 43, 224).

    Expression of BTLA, HVEM, and related ligands

    HVEM is a member of the tumor necrosis factor receptor super-

    family (TNFRSF), and the BTLA–HVEM interaction is the first

    demonstration of crosstalk between IgSF and TNF receptors

    (227). As its name would suggest, HVEM binding to herpes

    simplex virus glycoprotein D (HSV gD) is required for viral

    entry into host cells (228), and the HVEM binding site on

    gD is a structural mimic of that on BTLA (224). Prior to the

    discovery of its interaction with BTLA, HVEM was also known

    as a costimulatory receptor which binds to the TNF ligands

    homologous to lymphotoxins, exhibits inducible expression,

    and competes with HSV glycoprotein D for HVEM, a receptor

    expressed by T lymphocytes (LIGHT) and lymphotoxin a

    (LTa) (229), although the phenotype of HVEM-deficient mice

    is consistent with a role for HVEM in the negative regulation of

    T-cell activation (230). A complex of purified recombinant

    HVEM, BTLA, and LIGHT has been isolated chromatographi-

    cally (224), suggesting that the binding of LIGHT and BTLA to

    HVEM is not mutually exclusive (30, 231), but the functional

    relevance of such a ternary interaction is unknown.

    BTLA is constitutively expressed on most hematopoietic

    cells, including T cells, B cells, and DCs, with the highest

    levels present on B cells (29, 223, 232). The relatively low

    levels of BTLA on naive CD4+ T cells increase substantially

    after activation (232). Similarly to BTLA, HVEM is expressed

    widely throughout lymphoid compartments, with constitutive

    levels on naive T cells that decrease after activation (30, 233).

    The broad expression patterns of BTLA and HVEM have made

    it hard to define the particular cell–cell interactions for which

    BTLA–HVEM binding is relevant. The production of inducible

    cell type-specific BTLA- and HVEM-deficient mice will likely

    be necessary to gain insight into this problem.

    Initial evidence that BTLA negatively regulates T-cell activa-

    tion came from in vitro assays using BTLA-deficient mouse

    T cells and antibody engagement of BTLA during anti-CD3

    T-cell stimulation (29, 223). The addition of anti-BTLA

    antibodies to naive primary T cells has been shown to inhibit

    both division and IL-2 production (234). BTLA ligation on

    human T cells has similar consequences (226). CD4+ T cells

    were shown to be more sensitive than CD8+ T cells to inhibi-

    tion by anti-BTLA antibody engagement, which may be due

    to their faster induction of higher levels of surface BTLA after

    activation (234). Finally, CHO cells expressing HVEM and

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    78 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • MHC class II were shown to inhibit the antigen-specific divi-

    sion of WT but not BTLA-deficient T cells (30).

    BTLA inhibitory signaling

    The approximately 100 a.a. cytoplasmic tail of mouse BTLA

    contains three conserved tyrosine-based signaling motifs

    (235). The most membrane proximal motif (YDND) can

    interact with the adapter protein Grb-2 when phosphorylated

    and also binds to the p85 subunit of PI3K. The other two

    motifs, surrounding tyrosine residues 257 and 282 in human

    BTLA, are ITIM sequences that mediate BTLA binding to the

    protein tyrosine phosphatases SHP-1 and SHP-2 when phos-

    phorylated (236). Thus, TCR crosslinking, which induces

    phosphorylation of BTLA cytoplasmic tyrosines and phospha-

    tase recruitment (29, 223), could result in the dephosphoryla-

    tion of signaling intermediates and attenuation of T-cell

    activation. The functional relevance of this mechanism, which

    has also been proposed for CTLA-4 and PD-1 (84, 85, 147), is

    in question (237), and, as mentioned above, it is unclear how

    specific phosphatase binding only to phosphotyrosines on

    inhibitory receptors is regulated (60).

    A growing body of work using mouse models of disease

    supports the conclusion that BTLA and HVEM have a negative

    influence on T-cell mediated inflammatory responses. Early

    data showed that BTLA-deficient mice are somewhat more

    sensitive to MOG-peptide-induced EAE (29), and, like

    PD-1-deficient mice, take longer to resolve acute eosinophilic

    airway inflammation induced by protein sensitization and

    subsequent intranasal challenge (238). In contrast to PD-1,

    BTLA expression was induced on partially MHC-mismatched

    cardiac allografts, and BTLA-deficient recipients, but not

    PD-1-deficient recipients, rejected normally tolerated MHC

    class II mismatched grafts (239). Graft rejection was also seen

    in recipients lacking the BTLA ligand HVEM and in those that

    received anti-BTLA antibodies. Interestingly, in a more recent

    study, HVEM has been found on stimulated Tregs, and HVEM

    expression on Tregs and BTLA expression on effector T cells

    seems to be important for optimal in vitro Treg suppression as

    well as cardiac allograft acceptance (240). Finally, administra-

    tion of anti-BTLA antibodies was reported to synergize with

    CTLA-4-Ig to prolong survival of islet allografts in mice

    (241, 242).

    BTLA, HVEM, and autoimmunity

    Two recent studies indicate that negative regulation of T-cell

    responses by BTLA and HVEM is important in the prevention

    of autoimmunity. Careful examination of older (approxi-

    mately 12 months) BTLA-deficient mice revealed develop-

    ment of a spontaneous autoimmune hepatitis-like disease,

    which was characterized by infiltrates similar to those seen in

    human autoimmune hepatitis and increased serum levels of

    liver transaminases (243). The contribution of HVEM expres-

    sion on various cell types to intestinal inflammation was eval-

    uated using a model of autoimmune colitis involving the

    transfer of CD4+CD45RB+ T cells into RAG-deficient recipi-

    ents (244). Bone marrow chimeric mice in which HVEM was

    deficient only on radioresistant compartments showed rapid

    acceleration of colitis, suggesting that HVEM expression on

    tissue-specific APCs may be involved in attenuating intestinal

    inflammation caused by BTLA-expressing T cells.

    Recent data with BTLA-deficient mice indicate that the

    greater proliferation of total T-cell populations seen in the

    absence of BTLA is not a cell-intrinsic defect but is rather due

    to greater numbers of antigen-experienced (CD44hi) CD8+

    T cells in the lymphoid compartments of these mice (245),

    which explains the lack of an effect of BTLA deficiency on

    proliferation of purified CD4+ T cells (234). Interestingly,

    the same study (245) points to a T-cell-intrinsic role for BTLA

    in regulating the generation of CD8+CD44hiCD62Lhi central

    memory phenotype cells, rather than inhibiting initial prolif-

    eration per se. The authors proposed that BTLA may limit cell

    survival during the contraction phase of memory generation,

    and this idea is supported by recent data from a mouse model

    of asthma (246). Prolonged airway inflammation mediated by

    adoptively transferred BTLA-deficient T cells in response to

    inhaled antigen was correlated with enhanced survival of

    the BTLA-deficient T cells in the lung when compared with

    WT cells. Although increased apoptosis was not detected in

    primary T-cell cultures inhibited by anti-BTLA antibodies

    (234), it is likely that this type of assay does not realistically

    model the later stages of T-cell activation where BTLA may

    function, and regulation of apoptotic pathways during the

    contraction phase remains an attractive mechanism of BTLA

    regulation of effector T cells.

    Human CD160 was recently recognized to be another IgSF

    member that functions as an inhibitory HVEM ligand

    expressed on T cells (31). Like BTLA, CD160 contains a single

    Ig domain and binds to the same region of HVEM (31).

    Crosslinking antibodies to CD160 (in conjunction with anti-

    CD3 ⁄CD28 stimulation) was shown to inhibit both prolifera-tion and cytokine production of purified CD4+ T cells, as well

    as the phosphorylation of CD3f and other proteins. In contrast

    to BTLA, CD160 is GPI-anchored and so the direct recruitment

    of intermediates that interfere with TCR signaling seems

    unlikely. Further study of this newly appreciated inhibitory

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 79

  • pathway is likely to yield insight into the biology of TNFRSF–

    IgSF interactions and the negative regulation of T-cell activa-

    tion in general.

    LAG-3

    LAG-3 (CD223) is a type I membrane glycoprotein of the IgSF

    first cloned from an NK cell line and shown to be expressed in

    activated human NK and T cells (32) as well as in activated

    B cells (247). It has genomic and structural similarity to CD4,

    with four extracellular Ig-like domains and conserved struc-

    tural motifs in between these domains. Both human and

    mouse LAG-3 bind to MHC class II more strongly than CD4

    (248–250), and this interaction has been reported to modu-

    late cytokine production by APCs (251).

    The initial reports of LAG-3 knockout mice demonstrated a

    deficiency in NK killing (252), but no T-cell phenotypes were

    found. On naive CD4+ and CD8+ T cells, LAG-3 is present at

    low levels that are upregulated after antigen-specific activation

    (250). Several reports have suggested that mouse LAG-3

    negatively regulates T-cell activation (33, 253, 254) and that

    signaling mediated by the cytoplasmic domain is involved in

    this process (255). Human LAG-3 is also postulated to inhibit

    T-cell activation (256). LAG-3 was shown to be upregulated

    on adoptively transferred CD4+ T cells undergoing an abortive

    response to antigen expressed on pulmonary epithelium

    (257), but whether the non-functional LAG-3-positive CD4+

    cells correspond to bona fide induced CD4+Foxp3+ Treg cells is

    debatable. The relevance of MHC class II binding to the func-

    tion of LAG-3 in inhibition of T-cell activation and differentia-

    tion is unclear.

    Studies of adoptively transferred antigen-specific CD8+ cells

    in mice which express hemagglutinin (HA) as a self or tissue-

    specific tumor antigen provide the most direct evidence that

    LAG-3 inhibits T-cell function in a cell-intrinsic manner (34).

    Injection of anti-LAG-3 antibodies or transfer of LAG-3-

    deficient T cells results in greater tissue-specific accumulation

    and IFN-c production by the antigen-specific CD8+ T cells,

    suggesting that anti-LAG-3 antibody administration blocks

    LAG-3 function. In addition, the cells display enhanced cyto-

    lytic function after vaccination with HA when this is com-

    bined with anti-LAG-3 injection. Finally, in mice that develop

    autochthonous prostate cancer and express HA specifically in

    the prostate (proHA · TRAMP) (258), this type of vaccina-tion results in reduced tumor grade. The effects on tumor pro-

    gression, although modest, suggest that LAG-3 may be a

    potential immunotherapeutic target, and this possibility is

    supported by clinical reports of LAG-3 expression on TILs.

    Examination of TILs from RCC has shown LAG-3 expression

    on CD8+ but not CD4+ T cells in all eight patients examined

    (259). In addition, a study of 94 Hodgkin’s lymphoma

    patients demonstrated a correlation between Foxp3) TILs that

    expressed high levels of LAG-3 and decreased Epstein-Barr

    virus-specific IFN-c production (260). These data, along with

    the studies in the proHA · TRAMP mice (34), are consistentwith a role for LAG-3 in the regulation of tumor-reactive

    CD8+ T cells. Additional examination of the cell-intrinsic

    and ⁄or -extrinsic mechanisms by which LAG-3 inhibits T-cellfunction and the contextual dependence of this on MHC class

    II ligand binding is warranted and may yield insight into

    potential immunotherapeutic strategies that involve LAG-3

    blockade.

    Concluding remarks

    The importance of CD28-mediated costimulation and CTLA-

    4-mediated inhibition for the activation of T cells has been

    known for 30 years. Since their discovery in the late 1980s, it

    has become apparent that other receptors of the IgSF inhibit

    T-cell activation. Mechanistic descriptions of T-cell inhibition

    commonly invoke the recruitment of activated tyrosine phos-

    phatases to the TCR; however, the molecular details of the

    regulation of specific binding to inhibitory receptors are

    unclear and require further studies. Despite this, efforts have

    long been made to manipulate the signaling pathways down-

    stream of T-cell inhibitory receptors for treatment of both

    cancer and autoimmune disease. The fact that CTLA-4-Ig is

    currently an FDA-approved therapy for RA and that anti-

    CTLA-4 antibodies are used for treatment of melanoma, pros-

    tate cancer, colon cancer, and RCC either as a monotherapy or

    in combination with other treatments emphasize the clinical

    importance of these molecules. The more recent discovery of

    additional members of the extended CD28 ⁄CTLA-4 familyand their ligands offers potentially novel targets for diagnosis

    or therapeutic intervention in cancer, autoimmunity, and

    chronic infections. As the ligands for CD28 and CTLA-4 are

    predominantly expressed by professional APCs, the main

    impact of immune modulators of this pathway is probably

    limited to the initial priming phase of autoimmune disease

    and immune responses against cancer. In contrast, the expres-

    sion of PD-L1, B7-H3, and B7x on cells in the peripheral

    tissues suggests that these molecules may act as gatekeepers

    for lymphocytic infiltration of these organs. This putative

    function as part of a protective mechanism against autoim-

    mune destruction has probably made them subversion targets

    for tumors and pathogens to achieve immune escape and offer

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    80 � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009

  • additional opportunities for therapeutic intervention. The

    expression of BTLA and HVEM throughout hematopoietic

    compartments necessitates cell type-specific analysis of their

    function, but the available data suggest that this pathway is

    important in the regulation of T-cell responses in multiple

    tissues. The negative regulation of T-cell activation involves

    numerous receptors and ligands expressed in separate cellular

    compartments at different times in the immune response. This

    observation suggests that there may be synergy between

    blockade of the various inhibitory pathways and encourages

    the experimental testing of combinatorial strategies for

    treatment of disease.

    References

    1. Mueller DL, Jenkins MK, Schwartz RH.Clonal expansion versus functional clonal

    inactivation: a costimulatory signallingpathway determines the outcome of T cell

    antigen receptor occupancy. Annu RevImmunol 1989;7:445–480.

    2. Jenkins MK, Taylor PS, Norton SD, UrdahlKB. CD28 delivers a costimulatory signal

    involved in antigen-specific IL-2 productionby human T cells. J Immunol

    1991;147:2461–2466.3. Harding FA, McArthur JG, Gross JA, Raulet

    DH, Allison JP. CD28-mediated signalling

    co-stimulates murine T cells and preventsinduction of anergy in T-cell clones. Nature

    1992;356:607–609.4. Lindsten T, June CH, Ledbetter JA, Stella G,

    Thompson CB. Regulation of lymphokinemessenger RNA stability by a surface-

    mediated T cell activation pathway. Science1989;244:339–343.

    5. Boise LH, et al. CD28 costimulation canpromote T cell survival by enhancing the

    expression of Bcl-XL. Immunity 1995;3:87–98.

    6. Wulfing C, Sumen C, Sjaastad MD, Wu LC,Dustin ML, Davis MM. Costimulation and

    endogenous MHC ligands contribute toT cell recognition. Nat Immunol 2002;

    3:42–47.7. Rudd CE, Raab M. Independent CD28 sig-

    naling via VAV and SLP-76: a model for intrans costimulation. Immunol Rev

    2003;192:32–41.8. Viola A, Schroeder S, Sakakibara Y,

    Lanzavecchia A. T lymphocyte costimulationmediated by reorganization of membrane

    microdomains. Science 1999;283:680–682.9. Brunet JF, et al. A new member of the

    immunoglobulin superfamily–CTLA-4.Nature 1987;328:267–270.

    10. Linsley PS, Greene JL, Brady W, Bajorath J,Ledbetter JA, Peach R. Human B7-1 (CD80)

    and B7-2 (CD86) bind with similar aviditiesbut distinct kinetics to CD28 and CTLA-4

    receptors. Immunity 1994;1:793–801.

    11. Collins AV, et al. The interaction propertiesof costimulatory molecules revisited. Immu-

    nity 2002;17:201–210.12. Walunas TL, Bakker CY, Bluestone JA.

    CTLA-4 ligation blocks CD28-dependent

    T cell activation [erratum appears in J ExpMed 1996;184:301]. J Exp Med

    1996;183:2541–2550.13. Krummel MF, Allison JP. CTLA-4 engage-

    ment inhibits IL-2 accumulation and cellcycle progression upon activation of resting

    T cells. J Exp Med 1996;183:2533–2540.14. Waterhouse P, et al. Lymphoproliferative

    disorders with early lethality in micedeficient in Ctla-4. Science 1995;270:

    985–988.15. Tivol EA, Borriello F, Schweitzer AN, Lynch

    WP, Bluestone JA, Sharpe AH. Loss of

    CTLA-4 leads to massive lymphoprolifera-tion and fatal multiorgan tissue destruction,

    revealing a critical negative regulatory roleof CTLA-4. Immunity 1995;3:541–547.

    16. Chambers CA, Cado D, Truong T, Allison JP.Thymocyte development is normal in

    CTLA-4-deficient mice. Proc Natl Acad SciUSA 1997;94:9296–9301.

    17. Hutloff A, et al. ICOS is an inducible T-cellco-stimulator structurally and functionally

    related to CD28. Nature 1999;397:263–266.

    18. Swallow MM, Wallin JJ, Sha WC. B7h, anovel costimulatory homolog of B7.1 and

    B7.2, is induced by TNFalpha. Immunity1999;11:423–432.

    19. Yoshinaga SK, et al. T-cell co-stimulationthrough B7RP-1 and ICOS. Nature

    1999;402:827–832.20. Ishida Y, Agata Y, Shibahara K, Honjo T.

    Induced expression of PD-1, a novel mem-ber of the immunoglobulin gene superfam-

    ily, upon programmed cell death. EMBO J1992;11:3887–3895.

    21. Dong H, Zhu G, Tamada K, Chen L. B7-H1,a third member of the B7 family, co-stimu-

    lates T-cell proliferation and interleukin-10secretion. Nat Med 1999;5:1365–1369.

    22. Freeman GJ, et al. Engagement of the PD-1immunoinhibitory receptor by a novel B7

    family member leads to negative regulationof lymphocyte activation. J Exp Med

    2000;192:1027–1034.

    23. Latchman Y, et al. PD-L2 is a second ligandfor PD-1 and inhibits T cell activation. Nat

    Immunol 2001;2:261–268.24. Chapoval AI, et al. B7-H3: a costimulatory

    molecule for T cell activation and

    IFN-gamma production. Nat Immunol2001;2:269–274.

    25. Suh WK, et al. The B7 family memberB7-H3 preferentially down-regulates T

    helper type 1-mediated immune responses.Nat Immunol 2003;4:899–906.

    26. Zang X, Loke P, Kim J, Murphy K, Waitz R,Allison JP. B7x: a widely expressed B7

    family member that inhibits T cell activa-tion. Proc Natl Acad Sci USA 2003;100:

    10388–10392.27. Prasad DV, Richards S, Mai XM, Dong C.

    B7S1, a novel B7 family member that nega-

    tively regulates T cell activation. Immunity2003;18:863–873.

    28. Sica GL, et al. B7-H4, a molecule of the B7family, negatively regulates T cell immu-

    nity. Immunity 2003;18:849–861.29. Watanabe N, et al. BTLA is a lymphocyte

    inhibitory receptor with similarities toCTLA-4 and PD-1. Nat Immunol

    2003;4:670–679.30. Sedy JR, et al. B and T lymphocyte attenua-

    tor regulates T cell activation through inter-action with herpesvirus entry mediator. Nat

    Immunol 2005;6:90–98.31. Cai G, Anumanthan A, Brown JA, Greenfield

    EA, Zhu B, Freeman GJ. CD160 inhibits acti-vation of human CD4+ T cells through

    interaction with herpesvirus entry mediator.Nat Immunol 2008;9:176–185.

    32. Triebel F, et al. LAG-3, a novel lymphocyteactivation gene closely related to CD4. J Exp

    Med 1990;171:1393–1405.33. Workman CJ, Cauley LS, Kim IJ, Blackman

    MA, Woodland DL, Vignali DA. Lympho-cyte activation gene-3 (CD223) regulates

    the size of the expanding T cell populationfollowing antigen activation in vivo.

    J Immunol 2004;172:5450–5455.34. Grosso JF, et al. LAG-3 regulates CD8+ T

    cell accumulation and effector function inmurine self- and tumor-tolerance systems.

    J Clin Invest 2007;117:3383–3392.35. Linsley PS, et al. Binding stoichiometry of

    the cytotoxic T lymphocyte-associated

    molecule-4 (CTLA-4). A disulfide-linkedhomodimer binds two CD86 molecules.

    J Biol Chem 1995;270:15417–15424.36. Magistrelli G, et al. A soluble form of

    CTLA-4 generated by alternative splicing is

    Pentcheva-Hoang et al Æ T-cell regulation in cancer and autoimmunity

    � 2009 John Wiley & Sons A/S • Immunological Reviews 229/2009 81

  • expressed by nonstimulated human T cells.Eur J Immunol 1999;29:3596–3602.

    37. Oaks MK, Hallett KM, Penwell RT, StauberEC, Warren SJ, Tector AJ. A native soluble

    form of CTLA-4. Cell Immunol2000;201:144–153.

    38. Ueda H, et al. Association of the T-cellregulatory gene CTLA4 with susceptibility

    to autoimmune disease. Nature2003;423:506–511.

    39. Vijayakrishnan L, et al. An autoimmunedisease-associated CTLA-4 splice variant

    lacking the B7 binding domain signalsnegatively in T cells. Immunity

    2004;20:563–575.

    40. Metzler WJ, et al. Solution structure ofhuman CTLA-4 and delineation of a

    CD80 ⁄ CD86 binding site conserved inCD28. Nat Struct Biol 1997;4:527–531.

    41. Ostrov DA, Shi W, Schwartz JC, Almo SC,Nathenson SG. Structure of murine CTLA-4

    and its role in modulating T cell responsive-ness. Science 2000;290:816–819.

    42. Schwartz JC, Zhang X, Fedorov AA,Nathenson SG, Almo SC. Structural basis for

    co-stimulation by the human CTLA-4 ⁄ B7-2complex. Nature 2001;410:604–608.

    43. Stamper CC, et al. Crystal structure of theB7-1 ⁄ CTLA-4 complex that inhibits humanimmune responses. Nature 2001;410:608–611.

    44. Freeman GJ, et al. Structure, expression,and T cell costimulatory activity of the

    murine homologue of the human Blymphocyte activation antigen B7. J Exp

    Med 1991;174:625–631.45. Borriello F, Freeman GJ, Edelhoff S, Disteche

    CM, Nadler LM, Sharpe AH. Characteriza-tion of the murine B7-1 genomic locus

    reveals an additional exon encoding analternative cytoplasmic domain and a chro-

    mosomal location of chromosome 16, bandB5. J Immunol 1994;153:5038–5048.

    46. Ikemizu S, et al. Structure and dimerizationof a soluble form of B7-1. Immunity

    2000;12:51–60.47. Freeman GJ, et al. Murine B7-2, an

    alternative CTLA4 counter-receptor that

    costimulates T cell proliferation andinterleukin 2 production. J Exp Med

    1993;178:2185–2192.48. Freeman GJ, et al. Cloning of B7-2: a CTLA-

    4 counter-receptor that costi