Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment

https://doi.org/10.1016/j.coi.2013.02.009Get rights and content

For tumor antigen-specific T cells to effectively control the growth of cancer cells in vivo, they must gain access to, and function within, the tumor microenvironment. While tumor antigen-based vaccines and T cell adoptive transfer strategies can result in clinical benefit in a subset of patients, most of the patients do not respond clinically. Even for tumor-infiltrating lymphocyte (TIL)-based adoptive transfer for patients with metastatic melanoma, which can provide tumor shrinkage in around 50% of treated individuals, many patients are not eligible, in part because there are not sufficient TIL present in the resected tumor. Thus, the denominator is in fact larger, and it has been suggested that absence of TIL may be a marker for poor efficacy of immunotherapies in general. While qualitative and/or quantitative features of the T cells are important considerations for efficacy, a major component of primary resistance likely can be attributed to the tumor microenvironment. Data are accumulating suggesting that two major categories of immune resistance within the tumor microenvironment may exist: failure of T cell trafficking due to low levels of inflammation and lack of chemokines for migration, and dominant suppression through immune inhibitory mechanisms. New therapeutic interventions are being guided by these observations, and preliminary clinical success is validating this working model.

Highlights

► A T cell-inflamed tumor microenvironment may be predictive of response to immunotherapies. ► Non-inflamed tumors may require interventions directed at promoting chemokine production and T cell recruitment into tumor sites. ► T cell-infiltrated tumors appear to escape via the dominant action of immune suppressive mechanisms. ► Strategies to interfere with PD-L1/PD-1 interactions, block IDO activity, depleted Tregs, and reverse T cell anergy have been validated in animal models and are being tested clinically. ► Anti-PD-1 and anti-PD-L1 mAbs have shown impressive clinical activity in early phase clinical trials.

Section snippets

Introduction: probing the tumor microenvironment as a predictive biomarker for clinical benefit from immunotherapy

Analysis of the tumor microenvironment in a systematic fashion in patients began with the goal of identifying a predictive biomarker associated with clinical benefit to therapeutic cancer vaccines. The motivation for this pursuit was the observation that vaccine-induced immune responses as measured in the peripheral blood did not consistently correlate with anti-tumor activity. In fact, with the most potent vaccine formulations, the majority of patients indeed develop antigen-specific T cell

Tumors lacking T cell-based inflammation may require innate immune triggers to promote T cell trafficking

A working model has emerged in which one might envision the need for a distinct set of immunotherapeutic interventions dictated by the presence or absence of an immune-permissive tumor microenvironment. Developing strategies for how to manipulate non-inflamed tumors and render them permissive should benefit from a greater understanding of how a spontaneous T cell response is able to develop naturally in a subset of patients. Theoretically, in addition to the expression of (neo-)antigens that

Tumors that support T cell trafficking appear to show high expression of immune suppressive pathways that can be targeted therapeutically

If a subset of tumors does indeed support endogenous T cell priming all the way through to migration of activated CD8+ effector cells into a chemokine-rich tumor microenvironment, then it may seem paradoxical that those tumors exist at all and are not rejected spontaneously by the host. However, several small functional studies of T cells isolated from the melanoma tumor microenvironment have suggested that those specific for tumor antigens appear to be hyporesponsive [40, 41, 42].

Conclusions and future directions

Increasing our understanding of the tumor microenvironment has provided a foundation for the rational development of immunotherapeutic approaches for the treatment of cancer. As a T cell-inflamed tumor microenvironment may be a relevant predictive biomarker for clinical benefit, novel approaches to induce this phenotype as a therapeutic strategy should be pursued. Proof-of-concept approaches in preclinical models have largely been evaluated using intratumoral injection, which will not likely be

References and recommended reading

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

  • • of special interest

  • •• of outstanding interest

Acknowledgments

Data discussed in this review were supported by R01CA161005, R01CA127475, R01CA118153, P01CA97296, and the Melanoma Research Alliance. Through the course of this work, the authors have appreciated the technical support of Michael Leung, Michelle Gao, and Glee Li, as well as infrastructure support through multiple University of Chicago Comprehensive Cancer Center shared resources (Human Immunologic Monitoring and cGMP facilities, Functional Genomics Facility, Human Tissue Resource, and Flow

References (97)

  • M. Pellegrini et al.

    Adjuvant IL-7 antagonizes multiple cellular and molecular inhibitory networks to enhance immunotherapies

    Nat Med

    (2009)
  • L. Gattinoni et al.

    Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells

    J Exp Med

    (2005)
  • J.C. Steel et al.

    Interleukin-15 biology and its therapeutic implications in cancer

    Trends Pharmacol Sci

    (2012)
  • T.M. Petrella et al.

    Interleukin-21 has activity in patients with metastatic melanoma: a phase II study

    J Clin Oncol

    (2012)
  • K. Sakuishi et al.

    Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity

    J Exp Med

    (2010)
  • A.C. Peterson et al.

    Immunization with Melan-A peptide-pulsed peripheral blood mononuclear cells plus recombinant human interleukin-12 induces clinical activity and T-cell responses in advanced melanoma

    J Clin Oncol

    (2003)
  • J. Fourcade et al.

    Immunization with analog peptide in combination with CpG and montanide expands tumor antigen-specific CD8+ T cells in melanoma patients

    J Immunother

    (2008)
  • S. Adams et al.

    Immunization of malignant melanoma patients with full-length NY-ESO-1 protein using TLR7 agonist imiquimod as vaccine adjuvant

    J Immunol

    (2008)
  • S.A. Rosenberg et al.

    Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma [see comments]

    Nat Med

    (1998)
  • T.F. Gajewski et al.

    Chemokines expressed in melanoma metastases associated with T cell infiltration

    2007 ASCO Annual Meeting Proceedings Part I

    J Clin Oncol

    (2007)
  • T.F. Gajewski et al.

    Association of gene expression profile in melanoma and survival to a dendritic cell-based vaccine

    J Clin Oncol

    (2009)
  • H. Harlin et al.

    Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment

    Cancer Res

    (2009)
  • J. Louahed et al.

    Expression of defined genes identified by pre-treatment tumor profiling: association with clinical responses to the GSK MAGE-A3 immunotherapeutic in metastatic melanoma patients

    J Clin Oncol

    (2008)
  • T.F. Gajewski et al.

    Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy

    Cancer J

    (2010)
  • O. Hamid et al.

    A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma

    J Transl Med

    (2011)
  • R.R. Ji et al.

    An immune-active tumor microenvironment favors clinical response to ipilimumab

    Cancer Immunol Immunother

    (2011)
  • T.F. Gajewski

    Molecular profiling of melanoma and the evolution of patient-specific therapy

    Semin Oncol

    (2011)
  • E.D. Pleasance et al.

    A comprehensive catalogue of somatic mutations from a human cancer genome

    Nature

    (2010)
  • M.B. Fuertes et al.

    Type I interferon response and innate immune sensing of cancer

    Trends Immunol

    (2012)
  • M.S. Diamond et al.

    Type I interferon is selectively required by dendritic cells for immune rejection of tumors

    J Exp Med

    (2011)
  • G.N. Barber

    Innate immune DNA sensing pathways: STING, AIMII and the regulation of interferon production and inflammatory responses

    Curr Opin Immunol

    (2011)
  • H. Ishikawa et al.

    STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity

    Nature

    (2009)
  • L. Unterholzner et al.

    IFI16 is an innate immune sensor for intracellular DNA

    Nat Immunol

    (2010)
  • C.K. Holm et al.

    Virus-cell fusion as a trigger of innate immunity dependent on the adaptor STING

    Nat Immunol

    (2012)
  • A. Gall et al.

    Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease

    Immunity

    (2012)
  • J. Ahn et al.

    STING manifests self DNA-dependent inflammatory disease

    Proc Natl Acad Sci U S A

    (2012)
  • V.A. Rathinam et al.

    Regulation of inflammasome signaling

    Nat Immunol

    (2012)
  • G. Guarda et al.

    Type I interferon inhibits interleukin-1 production and inflammasome activation

    Immunity

    (2011)
  • P. Yu et al.

    Priming of naive T cells inside tumors leads to eradication of established tumors

    Nat Immunol

    (2004)
  • S.S. Oh et al.

    Adenovirally delivered IFN-beta exerts antitumor effects through transient T-lymphocyte depletion and Ag-specific T-cell proliferation

    Int J Mol Med

    (2012)
  • B.C. Burnette et al.

    The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity

    Cancer Res

    (2011)
  • G.T. Motz et al.

    The parallel lives of angiogenesis and immunosuppression: cancer and other tales

    Nat Rev Immunol

    (2011)
  • S. Singh et al.

    Stroma is critical for preventing or permitting immunological destruction of antigenic cancer cells

    J Exp Med

    (1992)
  • B. Zhang et al.

    Equilibrium between host and cancer caused by effector T cells killing tumor stroma

    Cancer Res

    (2008)
  • G.L. Beatty et al.

    CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans

    Science

    (2011)
  • G. Niu et al.

    Gene therapy with dominant-negative Stat3 suppresses growth of the murine melanoma B16 tumor in vivo

    Cancer Res

    (1999)
  • L. Burdelya et al.

    Stat3 activity in melanoma cells affects migration of immune effector cells and nitric oxide-mediated antitumor effects

    J Immunol

    (2005)
  • S. Ugurel et al.

    Impact of the CCR5 gene polymorphism on the survival of metastatic melanoma patients receiving immunotherapy

    Cancer Immunol Immunother

    (2008)
  • Cited by (0)

    View full text