Elsevier

Vaccine

Volume 33, Issue 51, 16 December 2015, Pages 7415-7422
Vaccine

In situ vaccination by radiotherapy to improve responses to anti-CTLA-4 treatment

https://doi.org/10.1016/j.vaccine.2015.05.105Get rights and content

Abstract

Targeting immune checkpoint receptors has emerged as an effective strategy to induce immune-mediated cancer regression in the subset of patients who have significant pre-existing anti-tumor immunity. For the remainder, effective anti tumor responses may require vaccination. Radiotherapy, traditionally used to achieve local tumor control, has acquired a new role, that of a partner for immunotherapy. Ionizing radiation has pro-inflammatory effects that facilitate tumor rejection. Radiation alters the tumor to enhance the concentration of effector T cells via induction of chemokines, cytokines and adhesion molecules. In parallel, radiation can induce an immunogenic death of cancer cells, promoting cross-presentation of tumor-derived antigens by dendritic cells to T cells. Newly generated anti-tumor immune responses have been demonstrated post-radiation in both murine models and occasional patients, supporting the hypothesis that the irradiated tumor can become an in situ vaccine. It is in this role, that radiation can be applied to induce anti-tumor T cells in lymphocyte-poor tumors, and possibly benefit patients who would otherwise fail to respond to immune checkpoint inhibitors. This review summarizes preclinical and clinical data demonstrating that radiation acts in concert with antibodies targeting the immune checkpoint cytotoxic T-lymphocyte antigen-4 (CTLA-4), to induce therapeutically effective anti-tumor T cell responses in tumors otherwise non responsive to anti-CTLA-4 therapy.

Introduction

From the inception of carcinogenesis, the immune system detects and eliminates nascent tumors in a process described as cancer immunosurveillance. Stress-induced ligands and altered antigenicity render transformed cells susceptible to natural killers (NK) cells, γδ and conventional α/β T cells. Tissue disruption and unscheduled cell death that occur during tumor progression to invasion generate dangers signals in the form of damage-associated molecular pattern (DAMP) molecules that alert the immune system of a potential threat, activating both innate and adaptive immunity [1]. However, occasionally elimination of cancer cells is incomplete and cancer cells that have acquired the ability to evade immune control emerge, as a result of the selective pressure of the immune system. Thus, cancers rise to clinical detection after a long and complex crosstalk with the immune system, while a dominant immune suppressive tumor micro-environment has also emerged. The latter is enriched in cells with regulatory and immunosuppressive function that secrete cytokines such as transforming growth factor-β (TGFβ) and IL-10, which counteract immune-mediated rejection [2]. Noticeably, in some patients robust anti-tumor T cell responses are detectable at clinical diagnosis and their presence in the tumor specimen has been associated with a better prognosis [3], [4]. Patients who retain such anti-tumor immunity appear to benefit the most from immunotherapy, even at advanced stages of the disease [5]. For example, responses to immune checkpoint inhibitors rely on the patient's pre-existing anti-tumor T cells [6], [7]. Unfortunately, only a small fraction of cancer patients retains sufficient anti-tumor immune responses. Among solid tumors patients, melanoma carriers are most likely to respond to immune checkpoint inhibitors targeting CTLA-4 or programmed cell death-1 (PD-1) [8], [9], possibly because of their high mutational load [10].

Because responses to anti-CTLA-4 often are durable [11], [12], identifying combination treatments that can convert patients unresponsive to CTLA-4 inhibition into responders is an active area of investigation. Potential candidates include other immunotherapies, standard chemotherapy, targeted agents [13], [14], [15], and radiotherapy has earned a prominent place, due to substantial pre-clinical data [16], [17], [18], [19], [20] and rapidly accumulating clinical observations [21], [22], [23] that it can induce therapeutically effective anti-tumor immunity when combined with CTLA-4 blockade. Several clinical trials are currently ongoing to test radiotherapy in combination with the FDA-approved anti-CTLA-4 antibody ipilimumab (Yervoy®, Bristol Meyers-Squibb, New York, New York) (Table 1).

Here we review the available data that has informed the rationale for exploiting the synergy of radiation and CTLA-4 blockade.

Section snippets

Radiation-induced in situ tumor vaccination

Over the past decade, an improved understanding of the effects of local radiation on tumor-host interactions has led to the recognition that radiotherapy may have a novel role as an inducer of acute inflammation and immunogenic cell death, capable to convert a tumor into an in situ vaccine [24], [25], [26]. Pioneering work implicating T cells in determining the response to radiation was published several decades ago [27]. More recently, the demonstration that T cells mediate the abscopal effect

Cytotoxic T lymphocyte antigen-4, a negative regulator of T-cell activation

An array of co-stimulatory and co-inhibitory molecules regulates T cells activation, balancing the need to eliminate pathogens with the prevention of autoimmunity [71]. T cell activation requires two signals, the first is delivered by TCR binding to MHC-I/antigen. The second is delivered by CD28 costimulatory receptor that binds to CD80 (B7–1) and CD86 (B7–2) on the surface of antigen presenting cells (APC), resulting in abundant secretion of IL-2 and T-cell proliferation [72].

After TCR

Synergy of radiotherapy with anti-CTLA-4 antibody

Previous work in pre-clinical models of melanoma and breast cancer showed that tumors insensitive to anti-CTLA-4 treatment as monotherapy became responsive upon vaccination with modified autologous tumor cells [89], [90]. We hypothesized that in situ vaccination by radiation could also convert a poorly immunogenic tumor, unresponsive to anti-CTLA-4 into a responder. This hypothesis was confirmed in three different murine tumor models, 4T1 and TSA mammary carcinomas syngeneic to BALB/c mice and

Clinical translation

Since 2011, after the approval of ipilimumab for patients with metastatic or unresectable melanoma, a few dramatic abscopal responses have been reported after radiation of one metastasis in patients who were unresponsive or had ceased to respond to ipilimumab [21], [22], [93]. These reports have sparked several retrospective analyses of outcome in melanoma patients receiving radiation while treated with ipilimumab, with an excellent review of these studies recently published by Barker and

Conclusions

The ability of radiation to elicit anti-tumor immune responses has been unequivocally demonstrated in experimental models, and many of the mechanisms involved have been identified. However, more work is required to define the dose(s) and fractionation that optimally induce anti-tumor T cells, and identify the tumor characteristics that predict which tumors will respond to a given combination of radiation and immune checkpoint blockade. While the growing number of reports of occasional abscopal

Conflict of interest statement

The authors declare that no conflict of interest exists.

Acknowledgements

The authors are grateful to Sophia Ceder (www.ceder.graphics) for illustrating Fig. 1. SD is supported by grants from the USA Department of Defense Breast Cancer Research Program (W81XWH-11-1-0532), The Chemotherapy Foundation, Breast Cancer Alliance, and Breast Cancer Research Foundation. CV-B is supported by a Post-doctoral fellowship from the Department of Defense Breast Cancer Research Program (W81XWH-13-1-0012). SCF is supported by grants from USA Department of Defense Breast Cancer

References (98)

  • L. Deng et al.

    STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors

    Immunity

    (2014)
  • Y. Lee et al.

    Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment

    Blood

    (2009)
  • C.S. Tsai et al.

    Macrophages from irradiated tumors express higher levels of iNOS, arginase-I and COX-2, and promote tumor growth

    Int J Radiat Oncol Biol Phys

    (2007)
  • F. Klug et al.

    Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy

    Cancer Cell

    (2013)
  • D. Schaue et al.

    Maximizing tumor immunity with fractionated radiation

    Int J Radiat Oncol Biol Phys

    (2012)
  • E.F. Stamell et al.

    The abscopal effect associated with a systemic anti-melanoma immune response

    Int J Radiat Oncol Biol Phys

    (2013)
  • C.A. Barker et al.

    Combinations of radiation therapy and immunotherapy for melanoma: a review of clinical outcomes

    Int J Radiat Oncol Biol Phys

    (2014)
  • E.D. Kwon et al.

    Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial

    Lancet Oncol

    (2014)
  • M.D. Vesely et al.

    Natural innate and adaptive immunity to cancer

    Annu Rev Immunol

    (2011)
  • J. Galon et al.

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

    Science

    (2006)
  • F. Pagès et al.

    Immune infiltration in human tumors: a prognostic factor that should not be ignored

    Oncogene

    (2010)
  • E. Wang et al.

    Prospective molecular profiling of melanoma metastases suggests classifiers of immune responsiveness

    Cancer Res

    (2002)
  • D.M. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat Rev Cancer

    (2012)
  • T.F. Gajewski et al.

    Innate and adaptive immune cells in the tumor microenvironment

    Nat Immunol

    (2013)
  • S.L. Topalian et al.

    Safety, activity, and immune correlates of anti-PD-1 antibody in cancer

    N Engl J Med

    (2012)
  • F.S. Hodi et al.

    Improved survival with ipilimumab in patients with metastatic melanoma

    N Engl J Med

    (2010)
  • A. Snyder et al.

    Genetic basis for clinical response to CTLA-4 blockade in melanoma

    N Engl J Med

    (2014)
  • M.A. Postow et al.

    The antitumor immunity of ipilimumab: (T-cell) memories to last a lifetime

    Clin Cancer Res

    (2012)
  • T.J. Lynch et al.

    Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study

    J Clin Oncol

    (2012)
  • J.D. Wolchok et al.

    Nivolumab plus ipilimumab in advanced melanoma

    N Engl J Med

    (2013)
  • S. Hu-Lieskovan et al.

    Combining targeted therapy with immunotherapy in BRAF-mutant melanoma: promise and challenges

    J Clin Oncol

    (2014)
  • S. Demaria et al.

    Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer

    Clin Cancer res

    (2005)
  • M.Z. Dewan et al.

    Fractionated but not single dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody

    Clin Cancer Res

    (2009)
  • K.A. Pilones et al.

    Invariant natural killer T cells regulate breast cancer response to radiation and CTLA-4 blockade

    Clin Cancer Res

    (2009)
  • S. Matsumura et al.

    Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells

    J Immunol

    (2008)
  • M.G. Ruocco et al.

    Suppressing T cell motility induced by anti-CTLA-4 monotherapy improves anti-tumor effects

    J Clin Invest

    (2012)
  • M.A. Postow et al.

    Immunologic correlates of the abscopal effect in a patient with melanoma

    N Engl J Med

    (2012)
  • E.B. Golden et al.

    An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer

    Cancer Immunol Res

    (2013)
  • H.B. Stone et al.

    Effect of host immune capability on radiocurability and subsequent transplantability of a murine fibrosarcoma

    J Natl Cancer Inst

    (1979)
  • G. Ehlers et al.

    Abscopal effect of radiation in papillary adenocarcinoma

    Br J Radiol

    (1973)
  • D.P. Kingsley

    An interesting case of possible abscopal effect in malignant melanoma

    Br J Radiol

    (1975)
  • G.J. Rees et al.

    Abscopal regression following radiotherapy for adenocarcinoma

    Br J Radiol

    (1983)
  • G. Kroemer et al.

    Immunogenic cell death in cancer therapy

    Annu Rev Immunol

    (2013)
  • M. Obeid et al.

    Calreticulin exposure dictates the immunogenicity of cancer cell death

    Nat Med

    (2007)
  • L. Apetoh et al.

    Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy

    Nat Med

    (2007)
  • F. Ghiringhelli et al.

    Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors

    Nat Med

    (2009)
  • P. Scaffidi et al.

    Release of chromatin protein HMGB1 by necrotic cells triggers inflammation

    Nature

    (2002)
  • I.E. Dumitriu et al.

    Release of high mobility group box 1 by dendritic cells controls T cell activation via the receptor for advanced glycation end products

    J Immunol

    (2005)
  • B. Moser et al.

    Receptor for advanced glycation end products expression on T cells contributes to antigen-specific cellular expansion in vivo

    J Immunol

    (2007)
  • Cited by (135)

    • Exploring low-dose radiotherapy to overcome radio-immunotherapy resistance

      2023, Biochimica et Biophysica Acta - Molecular Basis of Disease
    • The paradox of radiation and T cells in tumors

      2022, Neoplasia (United States)
      Citation Excerpt :

      However, vaccination-based therapies have not significantly impacted clinical treatment of patients. [168] While vaccination effects by radiation have been observed, [169] a range of data suggest that this response alone is inadequate to generate sufficiently effective T cells for tumor control. We have found that tumor control by radiotherapy and checkpoint inhibitors depends on regulation of pre-existing immune responses rather than by vaccination by tumor irradiation. [66]

    View all citing articles on Scopus
    View full text