Immune surveillance: a balance between protumor and antitumor immunity

https://doi.org/10.1016/j.gde.2007.12.007Get rights and content

Precancerous and malignant cells can induce an immune response which results in the destruction of transformed and/or malignant cells, a process known as immune surveillance. However, immune surveillance is not always successful, resulting in ‘edited’ tumors that have escaped immune surveillance. Immunoediting is not simply because of the absence of antitumor immunity, but is because of protumor immunity that blocks antitumor adaptive and innate responses, and promotes conditions that favor tumor progression. Several immune protumor effector mechanisms are upregulated by chronic inflammation, leading to the hypothesis that inflammation promotes carcinogenesis and tumor growth by altering the balance between protumor and antitumor immunity, thereby preventing the immune system from rejecting malignant cells, and providing a tumor-friendly environment for progressive disease.

Introduction

The concept that the immune system can be harnessed as a therapeutic agent to treat established tumors (immunotherapy) was first proposed in the early 1900s by Paul Ehrlich. He suggested that molecules that we now know as antibodies could deliver toxins directly to cancer cells. Ehrlich's ‘magic bullet’ strategy was expanded upon in the 1950s by Burnet and Thomas. They hypothesized that the immune system may also protect against nascent cancers by destroying malignant cells before they developed into detectable tumors, a concept that has become the immune surveillance hypothesis [1, 2]. Although enthusiasm for the validity of immunotherapy and immune surveillance waned in the 1970s, subsequent studies demonstrated that the immune system can protect against tumor onset and be manipulated to reject established tumors. Revival of the immune surveillance hypothesis led to a re-working of the initial concept, to include the concept of ‘immunoediting.’ During immunoediting, the immune system destroys many precancerous and malignant cells; however, some cells escape the immune response and give rise to progressively growing tumors. Immunoediting is thought to continue throughout the life of the tumor so that the phenotype of an established tumor has been directed by the host's immune response. It has also become apparent that both innate and adaptive immunity have a ‘dark’ side and can promote tumor progression as well as mediate tumor destruction. Not surprisingly, chronic inflammation, which has long been associated with increased tumor risk, is involved in polarizing immunity toward those effectors that facilitate tumor growth. As a result, the immune system has the potential to either promote or delay tumor onset and progression, and the effectiveness of immune surveillance and the efficacy of immunotherapy depend on the balance between these diametric opposites (Figure 1). After a brief over-view of the observations supporting the concept of immune surveillance, this article will review the cells that mediate protumor and antitumor immunity including a discussion of how inflammation polarizes innate and adaptive immunity toward either a protumor or antitumor phenotype.

Section snippets

Immune surveillance and immunoediting

Rejuvenation of the concept that the immune system protects against nascent malignant cells occurred with the demonstration that mice deficient for various components of the adaptive or innate immune systems were more likely to develop some types of tumors, specifically sarcomas as opposed to carcinomas, as compared to immune competent mice, when exposed to carcinogens or transplanted with syngeneic tumor cells. Immune deficiencies included the absence of B cells and αβ or γδ T cells because of

CD4+ and CD8+ T lymphocytes

CD4+ and CD8+ T cells are the principal helper and effector cells, respectively, of adaptive cellular immunity, and many immunotherapy strategies are aimed at activating these cells to promote tumor cell destruction and long-term immune memory against recurrence of primary disease or outgrowth of metastases. Type 1 CD4+ T cells (Th1) facilitate tissue destruction and tumor rejection by providing help to cytotoxic CD8+ T cells, while Type 2 CD4+ T cells T(Th2) facilitate antibody production by B

B lymphocytes

Tumor-reactive monoclonal antibodies can have significant antitumor efficacy when passively administered to cancer patients. By contrast, most cancer vaccines or other therapies that are aimed at inducing tumor-reactive antibodies are largely ineffective in promoting tumor rejection, though there are exceptions [19]. More recent experiments indicate that activated B cells and their soluble products, presumably antibodies, can also facilitate carcinogenesis. Using a transgenic mouse model in

Macrophages

Macrophages are part of the innate immune system and play important roles in all aspects of immunity. They are an exceptionally heterogeneous population of cells. Similar to CD4+ T cells, macrophages can contribute to tumor destruction or facilitate tumor growth and metastasis, depending on their phenotype (Figure 3).

Macrophages that are ‘classically activated’ by IFNγ and bacterial lipopolysaccharides destroy tumor cells through their production of nitric oxide and Type 1 cytokines and

Natural killer (NK) cells

NK cells are components of the innate immune system that interact with adaptive immunity through their production of cytokines that modulate dendritic cell (DC) and cytotoxic T cell maturation. They are well recognized for their ability to directly lyse MHC class I-deficient tumor cells through the engagement of their activating receptors and lack of engagement of their inhibitory receptors. However, a subset of NK cells are also cytotoxic for activated CD8+ T cells [32] and DC [33], and

NKT cells

NKT cells, which express both NK and TCR, bridge the innate and adaptive immune systems. They are usually CD4+ and respond to lipid and glycolipid antigens as presented by nonclassical MHC class I CD1d molecules. Until recently there was confusion as to whether NKT cells promote tumor rejection or enhance immune surveillance. NKT cells prevent the spread of B16 melanoma metastases and promote immune surveillance in mice treated with the carcinogen 3-methyl-cholanthrene. However, CD1d knockout

Myeloid-derived suppressor cells (MDSCs)

MDSC are a morphologically and functionally heterogenous population of cells of myeloid origin that are elevated in almost all patients and experimental mice with cancer [37]. They suppress both innate and adaptive antitumor immunity by inhibiting CD8+ and CD4+ T cells, NK and NKT cells, and by blocking DC maturation [38, 39•, 40, 41]. MDSC suppress T cells through their production of arginase and/or reactive oxygen species (ROS); however, there is variability in which mediator(s) is used

Conclusions

The immune system has the capacity either to block tumor development and deter established tumors, or to promote carcinogenesis, tumor progression, and metastasis. Which of these conditions prevails depends on the balance between the protumor and antitumor mediators of both innate and adaptive immunity. Presumably, there are unifying mechanisms that orchestrate immunity towards tumor promotion versus tumor destruction. Since many of the tumor-promoting elements of the immune system are induced

References and recommended reading

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

  • • of special interest

  • •• of outstanding interest

Acknowledgements

The author's laboratory is supported by National Institute of Health grants R01CA118550 and R01CA84232, and Susan G. Komen Foundation for the Cure BCTR0503885.

References (57)

  • G. Gallina et al.

    Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells

    J Clin Invest

    (2006)
  • J.B. Swann et al.

    Immune surveillance of tumors

    J Clin Invest

    (2007)
  • G.P. Dunn et al.

    Interferons, immunity and cancer immunoediting

    Nat Rev Immunol

    (2006)
  • J. Galon et al.

    Type, density, and location of immune cells within human colorectal tumors predict clinical outcome

    Science

    (2006)
  • W. Zou

    Regulatory T cells, tumour immunity and immunotherapy

    Nat Rev Immunol

    (2006)
  • F. Baratelli et al.

    Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells

    J Immunol

    (2005)
  • S. Sharma et al.

    Cyclooxygenase 2 inhibition promotes IFN-gamma-dependent enhancement of antitumor responses

    J Immunol

    (2005)
  • N. Ralainirina et al.

    Control of NK cell functions by CD4+CD25+ regulatory T cells

    J Leukoc Biol

    (2007)
  • S. Wei et al.

    Plasmacytoid dendritic cells induce CD8+ regulatory T cells in human ovarian carcinoma

    Cancer Res

    (2005)
  • F. Castellino et al.

    Cooperation between CD4+ and CD8+ T cells: when, where, and how

    Annu Rev Immunol

    (2006)
  • L. Steinman

    A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage

    Nat Med

    (2007)
  • J.L. Langowski et al.

    IL-23 promotes tumour incidence and growth

    Nature

    (2006)
  • Y. Bi et al.

    Th17 cell induction and immune regulatory effects

    J Cell Physiol

    (2007)
  • J.L. Langowski et al.

    Swords into plowshares: IL-23 repurposes tumor immune surveillance

    Trends Immunol

    (2007)
  • M. Numasaki et al.

    Interleukin-17 promotes angiogenesis and tumor growth

    Blood

    (2003)
  • F. Benchetrit et al.

    Interleukin-17 inhibits tumor cell growth by means of a T-cell-dependent mechanism

    Blood

    (2002)
  • T.T. Tan et al.

    Humoral immunity, inflammation and cancer

    Curr Opin Immunol

    (2007)
  • Ostrand-Rosenberg S, Sinha P: Macrophages and tumor development. In Tumor-induced Immune Suppression: Mechanisms and...
  • Cited by (0)

    View full text