Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity

https://doi.org/10.1016/j.coi.2011.12.009Get rights and content

Genetic alterations and epigenetic dysregulation in cancer cells create a vast array of neoepitopes potentially recognizable by the immune system. Immune checkpoint blockade has the capacity to enhance and sustain endogenous immunity against non-mutated tumor-associated antigens as well as uniquely mutant antigens, establishing durable tumor control. Recent evidence from preclinical models highlights the pivotal role of the Programmed Death-1 (PD-1) T cell co-receptor and its ligands, B7-H1/PD-L1 and B7-DC/PD-L2, in maintaining an immunosuppressive tumor microenvironment. Encouraging early clinical results using blocking agents against components of the PD-1 pathway have validated its importance as a target for cancer immunotherapy.

Highlights

► PD-1/B7-H1 are pivotal in maintaining an immunosuppressive tumor microenvironment. ► PD-1 and CTLA-4 play distinct roles in regulating immunity. ► B7-H1 upregulation on tumor cells likely reflects ‘adaptive resistance’. ► PD-1 blockade is active against NSCLC, thought to be a ‘non-immunogenic’ tumor. ► Monotherapies blocking PD-1 may be more effective in combinatorial regimens.

Introduction

Ever since it became clear that all cancer cells express tumor-specific and tumor-selective antigens derived from genetic alterations and epigenetic dysregulation, the immunology community has embraced the potential for immune-based therapies to induce targeted anti-tumor responses. However, skepticism about the clinical value of immunotherapies designed to specifically enhance T cell-mediated immunity against preselected commonly expressed tumor antigens, including cancer vaccines and adoptive T cell transfer strategies, has escalated [1]. A game changer for cancer immunotherapy has emerged in the form of monoclonal antibodies (mAbs) that block inhibitory receptors on immune effector cells or their ligands on tumor cells and antigen presenting cells (APCs)  so-called ‘immune checkpoints’. Immune checkpoint blockade has the potential to enhance and sustain endogenous immunity against non-mutated as well as uniquely mutant antigens, establishing durable tumor control. Recent evidence highlights the pivotal role of the Programmed Death-1 (PD-1) T cell co-receptor and its ligands B7-H1/PD-L1 and B7-DC/PD-L2 in maintaining an immunosuppressive tumor microenvironment. Encouraging early results from clinical blockade of this pathway have validated its potential as a target for cancer immunotherapy.

Section snippets

Receptor/ligand biology

The two checkpoint receptors that have been most actively studied in the context of clinical cancer immunotherapy, CTLA-4 and PD-1 [2, 3•, 4, 5, 6, 7, 8], play distinct roles in regulating immunity (Figure 1). CTLA-4, which regulates the amplitude of early activation of naïve and memory T cells, is transported to the T cell surface at levels that depend on the strength of the TCR signal. It acts physiologically as a signal dampener. Its importance in modulating T cell responses is demonstrated

Preclinical models

Preclinical evidence for PD-1's inhibitory role comes from several sources. Knockout mice develop late-onset, strain-specific autoimmunity; on a C57BL/6 background this manifests as sporadic glomerulonephritis [28], on a Balb/c background as an antibody-mediated cardiomyopathy [29]. These findings extend to models of organ-specific autoimmunity; on the non-obese diabetic (NOD) background, PD-1 KO mice develop accelerated insulitis as well as increased T cell production of effector cytokines [30

Clinical translation

An understanding of PD-1 pathway biology and the preclinical demonstration of its pivotal role in immunosuppression have fueled the clinical development of PD-1 pathway blockade for cancer therapy. There are currently four anti-PD-1 agents in the clinic: MDX-1106/BMS-936558/ONO-4538, CT-011, MK-3475, and AMP-224. The first three are reported to be PD-1 blocking mAbs, while the last is a B7-DC/IgG1 fusion protein (Table 1).

To date, most clinical experience with PD-1 blockade has been gained with

Conclusions

Despite early successes with monotherapies blocking PD-1 pathways, preclinical models indicate that combinatorial therapies will deliver maximum clinical impact. Several clinical trials are already planned or in progress, combining anti-PD-1 mAbs with cancer vaccines (melanoma, prostate cancer, renal cell carcinoma, AML), anti-tumor mAbs (lymphoma), or chemotherapies (pancreatic cancer, NSCLC). These synergistic treatment strategies will provide a foundation for the next generation of clinical

Disclosure statement

Consulting or advisory role: S.L.T., Bristol-Myers Squibb (uncompensated) and Amplimmune Inc. (spouse); C.G.D., Bristol-Myers Squibb and Amplimmune Inc.; D.M.P., Bristol-Myers Squibb (uncompensated) and Amplimmune Inc. Stock ownership: C.G.D., Amplimmune Inc. Research funding: S.L.T., Bristol-Myers Squibb.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

Supported by NIH grants R01 CA142779 (S.L.T., D.M.P.), R01 CA127153 (C.G.D.), and 1P50CA58236-15 (C.G.D.); a grant from the Melanoma Research Alliance (S.L.T., C.G.D., D.M.P.); the Laverna Hahn Charitable Trust and the Barney Family Foundation (S.L.T.); the Patrick C. Walsh Fund, the OneInSix Foundation, and the Prostate Cancer Foundation (C.G.D.).

References (43)

  • P.S. Linsley et al.

    T-cell antigen CD28 mediates adhesion with B cells by interacting with activation antigen B7/BB-1

    Proc Natl Acad Sci U S A

    (1990)
  • K. Wing et al.

    CTLA-4 control over Foxp3+ regulatory T cell function

    Science

    (2008)
  • K.S. Peggs et al.

    Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies

    J Exp Med

    (2009)
  • D.L. Barber et al.

    Restoring function in exhausted CD8 T cells during chronic viral infection

    Nature

    (2006)
  • H. Dong et al.

    B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion

    Nat Med

    (1999)
  • G.J. Freeman et al.

    Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation

    J Exp Med

    (2000)
  • Y. Latchman et al.

    PD-L2 is a second ligand for PD-1 and inhibits T cell activation

    Nat Immunol

    (2001)
  • S.Y. Tseng et al.

    B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells

    J Exp Med

    (2001)
  • C.M. Wilke et al.

    Dual biological effects of the cytokines interleukin-10 and interferon-γ

    Cancer Immunol Immunother

    (2011)
  • T. Shin et al.

    In vivo costimulatory role of B7-DC in tuning T helper cell 1 and cytotoxic T lymphocyte responses

    J Exp Med

    (2005)
  • M.J. Butte et al.

    Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses

    Immunity

    (2007)
  • Cited by (1124)

    View all citing articles on Scopus
    View full text