2,180
Views
15
CrossRef citations to date
0
Altmetric
RADIATION MEETS IMMUNOTHERAPY – A PERFECT MATCH?

Radiation meets immunotherapy – a perfect match in the era of combination therapy?

&
Pages 299-305 | Received 03 Jul 2014, Accepted 18 Nov 2014, Published online: 09 Feb 2015

Abstract

Purpose: This review focuses on recent advances in the field of combining radiation with immunotherapy for the treatment of malignant diseases, since various combinatorial cancer therapy approaches have lately proven highly successful.

Results: With initial case reports and anecdotes progressively converting into solid clinical data, interest in cancer immunotherapy (CIT) has risen steeply. Especially immune checkpoint blockade therapies have recently celebrated tremendous successes in the treatment of severe malignancies resistant to conventional treatment strategies. Nevertheless, the high variability of patient responses to CIT remains a major hurdle, clearly indicating an urgent need for improvement. It has been suggested that successful cancer therapy most probably involves combinatorial treatment approaches. Radiotherapy (RT) has been proposed as a powerful partner for CIT due to its broad spectrum of immune modulatory characteristics. Several preclinical studies, supported by an increasing number of clinical observations, have demonstrated synergistic interactions between RT and CIT resulting in significantly improved therapy outcomes.

Conclusions: Numerous reports have shown that radiation is capable of tipping the scales from tumor immune evasion to elimination in different tumor types. The next puzzle to be solved is the question of logistics – including types, schedule and dosage of combinatorial RT and CIT strategies.

Introduction

For several decades, the concept of cancer immunotherapy (CIT) has been struggling to establish itself as the fourth pillar of acknowledged cancer treatment strategies alongside surgery, radiation and chemotherapy. With its nomination as Science ‘Breakthrough of the Year 2013’ (CitationCouzin-Frankel 2013) and preclinical studies gradually translating into clinical data, the field of CIT has finally reached a state of acceptance among the established oncological domains. Currently, different immunotherapeutic approaches are standing their ground as powerful treatment strategies for a wide range of malignant diseases. A very prominent and recent example of an outstanding CIT success involves immune checkpoint blockade therapy by monoclonal antibodies (mAb) targeting inhibitory molecules on either immune effector T-cells or tumor cells. Interfering with co-inhibitors has been shown to unleash a powerful anti-tumor T-cell response (CitationPardoll 2012). Promising early-stage clinical trials have shown safety and impressive activity of mAb blocking activity of programmed cell death 1 (PD1), expressed on T-cells (CitationTopalian et al. 2012), or one of its ligands, programmed death-ligand 1 (PD-L1) (CitationBrahmer et al. 2012). Recently, the FDA approved lambrolizumab, a PD1-targeting mAb for treatment of advanced or unresected melanomas that no longer respond to other drugs (CitationHamid et al. 2013). Furthermore ipilimumab, a mAb against cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) on T-cells, was approved for the treatment of metastatic melanoma (CitationLipson and Drake 2011). In 2013, a combination of anti-CTLA4 and anti-PD1 mAb treatment was reported to act synergistically in increasing survival and tumor regression in advanced melanoma patients (CitationWolchok et al. 2013). This novel immunomodulatory approach exhibits great potential especially for the treatment of severe malignancies resistant to conventional therapies.

However, major obstacles to broad clinical applicability of CIT become more evident. Whereas significant improvements of overall and progression-free survival can be achieved in individual cancer patients, most CIT strategies fail to establish long-lasting tumor rejection in large patient groups – with many patients responding poorly to treatment (CitationBrahmer and Pardoll 2013, CitationFishman 2014, CitationRaval et al. 2014). The precise processes behind this high variability of therapeutic efficacy remain to be clarified, but most likely involve high heterogeneity of different tumor types as well as poor immunogenicity and evolving capability to escape immune recognition (CitationKalbasi et al. 2013, CitationKelderman et al. 2014). Based on the concept of cancer immunoediting, tumors undergo three distinct phases of interaction with the immune system: Elimination, equilibrium and ultimately escape (CitationSchreiber et al. 2011). Progressing through these steps involves tumors actively shaping the immune system in order to circumvent immune recognition and establish a state of permanent immune evasion. Successful therapy approaches counteract this active immunosuppression and empower the immune system to regain control over tumor growth. In numerous patients, CIT by itself fails at stably reversing the immunoediting process (CitationKalbasi et al. 2013). Therefore, the search has begun to look for potent partners in tipping the scales back from tumor immune escape to elimination.

Radiation therapy (RT) comes into play as a particularly attractive partner for CIT in empowering the immune system to re-engage in tumor elimination, since recent years have shown a number of mechanisms through which RT interacts with immunity. In addition to being highly effective at reducing tumor burden by inducing irreversible DNA damage causing cancer cell death, RT has been demonstrated to contribute actively to tumor immune recognition by various means as will be outlined in the following section (CitationKwilas et al. 2012).

From escape back to elimination – the immune modulatory capacities of radiotherapy

Numerous lines of evidence have initiated a paradigm shift from the traditional notion of radiation causing detrimental effects on various immune cell types towards the recognition of a potent systemic immunostimulatory impact (CitationMcBride et al. 2004, CitationFormenti and Demaria 2013). This more recent concept has moved into the center of attention due to repeated observations of so-called abscopal RT effects (CitationKalbasi et al. 2013), which describe the regression of tumor growth at sites distant from the primary field of irradiation (CitationDemaria et al. 2004). Several such case reports inspired researchers to investigate how radiation can systemically induce tumor elimination which led to the discovery of various mechanisms through which RT actively induces anti-tumor immunity (CitationBarker and Postow 2014).

Of utmost importance, radiation causes a strong increase in tumor-associated antigen (TAA) quantity and variety through induced cancer cell death (CitationCorso et al. 2011, CitationBurnette et al. 2012) as well as enhanced protein translation (CitationReits et al. 2006), permitting specialized antigen-presenting cells (APC) such as dendritic cells (DC) to effectively prime T-cells for specific recognition and efficient clearance of residual tumor cells (CitationAhmed et al. 2013, CitationFrey et al. 2014). In addition, it has been shown that radiation is capable of inducing activation of DC by the release of specific damage-associated molecular patterns (DAMP) through immunogenic tumor cell death, a process critical for enabling DC to orchestrate a potent anti-tumor immune response (CitationRoses et al. 2014). Particularly high-mobility group box 1 protein (HMGB1), released from irradiated dying tumor cells, has been demonstrated to induce sustained DC maturation through activation of the toll-like receptor (TLR) 4 pathway, which also increases efficiency of TAA processing and presentation (CitationApetoh et al. 2007). Persistent DC activation forms a critical part in generating a potent anti-tumor immune response since immature antigen-presenting DC induce anergy or even deletion of antigen-reactive T-cells – a mechanism which has been shown to be actively exploited by tumor cells as a means to escape T-cell recognition and cytotoxicity (CitationKim et al. 2006). The combined effects of radiation providing antigens along with adjuvant activating signals hinder tumors at taking the final step in the immunoediting process – escape – and have therefore led to the concept of referring to RT as an in situ anti-tumor vaccine (CitationDemaria et al. 2014, CitationFrey et al. 2014).

But the effects of RT on TAA detection and presentation go beyond increasing antigen availability: The induction of calreticulin translocation to the surface of tumor cells serves as a signal for recognition and phagocytosis by DC, thereby enhancing TAA processing and presentation (CitationObeid et al. 2007). Also trafficking of APC to regional lymph nodes, where interaction with T-cells takes place, has been described to be augmented by RT (CitationLugade et al. 2005). A similar effect has been reported regarding recruitment and cytotoxicity of CD8+ T-cells. CitationLim et al. (2014) observed the induction of type I and II interferons (IFN) through radiation, leading to enhanced intratumoral numbers and cytolytic activity of effector T-cells. Also CitationDraghiciu et al. (2014) reported enhanced recruitment of tumor-specific CD8+ cells into tumors upon low-dose radiation. In addition, the production of chemokines such as CXCL16 has been shown to be upregulated following radiation, which also attracts effector T-cells to the irradiated tumor site (CitationMatsumura and Demaria 2010). The direct recruitment of cytotoxic T-cells into the tumor provides a strong basis for the immune system to regain control over the transformed tissue.

RT has also been described to alter the phenotype of residual tumor cells surviving irradiation, mostly due to lower doses being transmitted further away from the radiation source (CitationGarnett et al. 2004, CitationGameiro et al. 2014). This alteration includes upregulated expression of various surface molecules, such as major histocompatibility complex (MHC) I, co-stimulatory T-cell signaling molecules, adhesion molecules and death receptors, thus further contributing to immune recognition and elimination by rendering tumor cells more visible to the immune system (CitationChakraborty et al. 2004, Citation2008b, CitationBernstein et al. 2014).

Furthermore, effects of RT on vascular normalization and density within tumors have been observed. As a result of excessive production of pro-angiogenic factors, tumors establish an abnormal vascular structure, which creates a hypoxic microenvironment that polarizes inflammatory immune cells towards immunosuppressive activity and hinders immune cells at effectively entering into tumor tissue (CitationHuang et al. 2013). In CIT, increasing evidence indicates that a normalized tumor vasculature substantially enhances immunotherapeutic success as it reverses the hypoxic microenvironment and enables immune effector cell infiltration (CitationHuang et al. 2013). RT was shown to induce normalization of tumor vasculature by increasing expression of chemokines CXCL9 and CXCL10, leading to vessel remodeling, as well as vascular cell adhesion protein 1 (VCAM-1), facilitating T-cell migration into tumors (CitationKershaw et al. 2013). A study by CitationGanss et al. (2002) showed that RT led to enhanced vessel density and diameter within tumors, which facilitated access of T-cells by enabling them to adhere to the endothelium, ultimately leading to tumor regression (CitationGanss et al. 2002). Effects of radiation on vessel remodeling therefore provide another promising rationale for its combination with CIT by actively contributing to tumor elimination.

Another branch of the immune system affected by RT is humoral immunity. In melanoma patients, increases in tumor-specific antibody levels have been observed following radiation (CitationPostow et al. 2012). CitationDemaria et al. (2005a) reported RT to induce production of pro-inflammatory cytokines, resulting in the activation and migration of various immune cell subsets. Nevertheless, the actual contribution of a heavily pro-inflammatory cytokine milieu on tumor progression or rejection has become a matter of debate among immunologists and has to be considered carefully in a context-dependent manner (CitationGrivennikov et al. 2010).

Nevertheless, when dissecting mechanisms of RT impacting on immunity, its effects on immunosuppression also have to be taken into account. Such mechanisms include proportionally increasing regulatory T-cell (Treg) incidence, which can be attributed to an inherently higher radioresistance of these cells (CitationFormenti and Demaria 2013), as well as induction of transforming growth factor (TGF) β secretion, which was shown to inhibit systemic immune-activating effects of RT (CitationDiamond et al. 2013). Blockade of TGFβ was proven not only to induce abscopal RT effects, but also to overcome local immunosuppression (CitationDiamond et al. 2013). In addition to these observations, expression of co-inhibitory molecules such as PD-L1 was shown to be induced in tumor cells after local high-dose irradiation (CitationDeng et al. 2014). This consequence provides a clear example of the strong rationale for combining RT with immune checkpoint blockade. Hence, it has been implicated that radiation may promote immunosuppression by different means in a dose-dependent manner (CitationKwilas et al. 2012).

The importance of gaining deeper understanding of RT-mediated effects and the resulting cellular interactions becomes apparent when taking the increased recognition of the tumor stroma into account. It has become well-established that various types of cells ranging from cancer cells themselves (including cancer stem and bulk cells), local and bone marrow-derived stromal stem and progenitor cells, endothelial cells, pericytes and cancer-associated fibroblasts to immune cells, contribute to the formation of a unique tumor microenvironment (CitationHanahan and Weinberg 2011). In order to achieve sustained therapy success, this entire tumor niche has to be considered and numerous groups currently focus on gaining deeper understanding of the predominant processes and cellular interactions.

Finding the perfect RT-CIT match – preclinical and clinical observations

Several groups have embarked on the mission of actively evaluating synergisms between individual RT-CIT combinations and first results showed robust improvements in therapy outcome. At a preclinical state, external beam RT (EBRT) in combination with adoptive tumor-specific CD8+ T-cell therapy (CitationChakraborty et al. 2003, CitationReits et al. 2006) and vaccination approaches including recombinant virus strategies and TLR ligand administration (CitationChakraborty et al. 2004, CitationDemaria et al. 2013, CitationWitek et al. 2014) was demonstrated to result in drastically enhanced tumor regression by increased CD4+ and CD8+ T-cell responses. Similar results were obtained with radiolabeled mAb (CitationChakraborty et al. 2008a) as well as brachytherapy and vaccine-mediated CIT (CitationHodge et al. 2012). As indicated earlier, combining local radiation with antibodies targeting immune checkpoint blockade molecules such as CTLA4 or PD-L1 also yielded highly synergistic effects on therapy outcome (CitationDemaria et al. 2005b, CitationDewan et al. 2009, CitationDeng et al. 2014). Another immunotherapeutic agent, which has been investigated in search of beneficial combinatorial strategies, is IL-2. Given its limitations in establishing long-term tumor rejection, relatively low response rates and association with severe side effects (CitationSiegel and Puri 1991, CitationAtkins et al. 1999, CitationSchwartz et al. 2002, CitationMcDermott 2007, CitationSeung et al. 2012a), several preclinical studies have focused on evaluating the potential of combining IL-2 with RT for improved therapy successes (CitationCameron et al. 1990, CitationSafwat et al. 2003, Citation2004). Importantly, these studies showed varying results based on RT dose and target with promising synergistic observations for the administration of focal radiation prior to IL-2 injection in a mouse model of liver metastases (CitationCameron et al. 1990). These are just few examples of the manifold preclinical investigations conducted thus far, which have been paving the way for a clinical evaluation of combination treatments.

Clinical data on the synergism of RT and CIT are not as extensive yet. Nevertheless, various case reports strengthen pre-clinical observations and in a number of early-stage clinical trials, combinations of mostly local low-dose RT with different immunotherapeutic strategies have been proven safe and well-tolerated and exhibit great potential of synergistically improving therapy outcomes, with various trials ongoing (reviewed extensively by e.g., CitationKwilas et al. 2012, CitationFormenti and Demaria 2013, CitationKalbasi et al. 2013, CitationBarker and Postow 2014, CitationDemaria et al. 2014, CitationWattenberg et al. 2014).

In a series of pilot trials, Gulley et al. evaluated the safety of combining EBRT with low- or metronomic-dose IL-2 and a recombinant vaccinia virus-based vaccination strategy in prostate and rectal cancer (CitationGulley et al. 2005, Citation2011, CitationLechleider et al. 2008). These studies revealed safety and tolerance of the therapy combinations as well as efficacy in generating tumor-specific immune responses. Another RT strategy, which is currently being evaluated in combination with recombinant vaccinia virus-based vaccination, is Samarium-153-ethylene diamine tetramethylene phosphonate (153Sm-EDTMP). In an interim analysis, 29.4% of metastatic castration-resistant prostate cancer (CRPC) patients receiving the combined therapies were found to remain progression-free after 4 months as compared to 11.8% receiving RT alone (CitationHeery et al. 2012). A more recent study, in which different regimens of stereotactic body RT (SBRT) were combined with high-dose IL-2 in metastatic melanoma and renal cell carcinoma patients, revealed a promising 71% response rate in patients treated with 1 or 2 fractions of 20 Gy as compared to a response rate of 16% for IL-2 monotherapy, with responding patients showing enhanced immune activation (CitationSeung et al. 2012a, Citation2012b).

Barker et al. and Postow et al. focus on dissecting combinatorial effects of RT and ipilimumab in melanoma patients regarding safety and preliminary efficacy. In a retrospective study, they reported a 39-month median overall survival in patients receiving RT during maintenance phase of ipilimumab administration (> 16 weeks after starting ipilimumab) as compared to 9 months in patients who received RT during the induction phase (≤ 16 weeks of starting ipilimumab), which underlines the importance of future research regarding treatment schedules. Importantly, they also found the combinatorial treatment to be as safe and feasible as each individual therapy alone, which is in accordance with numerous case reports at different study centers (CitationPostow et al. 2012, CitationBarker et al. 2013, CitationBarker and Postow 2014). As in melanoma, the combination of ipilimumab with RT for the treatment of CRPC was also found to be well-tolerated – nevertheless, it did not reveal significant improvements in therapy outcome as compared to ipilimumab administration alone, but further clinical trials are currently under way (CitationSlovin et al. 2009, Citation2013).

Further examples involve immunotherapeutic strategies aimed at DC-mediated tumor immune recognition (CitationFormenti and Demaria 2013). In patients bearing metastatic solid tumors, the combination of granulocyte macrophage colony-stimulating factor (GM-CSF) administration with local radiotherapy was shown to induce an abscopal response in 30% of patients (CitationFormenti and Demaria 2009). Limited success was achieved in a different phase I trial, in which advanced hepatocellular carcinoma patients were injected intratumorally with autologous DC following a single fraction of radiotherapy, with eight of 14 patients showing enhanced tumor-specific immune responses (CitationChi et al. 2005). Another approach involved intratumoral autologous DC injection during fractionated EBRT in soft-tissue sarcoma patients, which led to remarkable tumor-specific immune responses and one-year progression-free survival in 12 of 17 patients (CitationFinkelstein et al. 2012). Furthermore, a retrospective multivariate regression analysis by CitationDillman et al. (2011) revealed RT as one of six features correlating with survival in metastatic melanoma patients receiving vaccinations of autologous DC loaded with tumor antigens. CitationBrody et al. (2010) conducted a phase I/II trial for the combination of low-dose RT with intratumoral injection of a DC-activating TLR9 agonist in 15 patients with low-grade B cell lymphoma and reported one complete and three partial responses. Finally, CitationDohnal et al. (2007) demonstrated safety and feasibility of an autologous tumor-lysate-loaded DC therapy approach in combination with RT in pediatric sarcoma patients.

The era of combination therapy – hurdles to be taken in the future

In the final section we want to highlight the main challenges that have to be addressed in the future to exploit the full potential of RT-CIT combination. The repertoire is sheer endless – ranging from different RT strategies including EBRT, SBRT, bone-seeking radionuclides, radiolabeled antibodies, brachytherapy and proton therapy, to numerous CIT approaches such as immune checkpoint blockade, unspecific stimulation, different vaccine-based concepts, adoptive effector cell transfer or targeted immunotherapeutics like antibodies – and not to forget the matters of dosage, timing, and choosing the right patient population as well as a reasonable stage of disease.

One major challenge remaining is to identify the most promising strategies and treatment schedules which will result in maximum efficacy by taking advantage of the strengths of each single therapy. Considering the broad spectrum of mechanisms by which RT impacts on the immune system, the balance between immunosuppression and activation ultimately determines whether a certain approach will result in successful tumor elimination. This underlines the importance to evaluate the predominant immunomodulatory effects of different RT regimens. Various publications have shown that dose, mode of delivery and schedule of RT can cause substantially different effects on the tumor immune response – with the most vital question remaining ‘to fractionate or not to fractionate?’ (CitationFormenti and Demaria 2013, CitationBarker and Postow 2014). Several groups have reported low-dose irradiation (LDI) to induce immune-activating effects by altering tumor and immune cell surface molecule expression (CitationIna and Sakai 2005, CitationKwilas et al. 2012), promoting T-cell-stimulatory capacities of DC (CitationShigematsu et al. 2007), or an anti-tumor macrophage phenotype (CitationKlug et al. 2013). Simultaneously, ablative high-dose irradiation (HDI) was reported to stimulate potent anti-tumor cytotoxic T-cell responses, mediated primarily through DC activation (CitationLee et al. 2009, CitationGupta et al. 2012) whereas CitationSchaue et al. (2012) reported beneficial effects of medium-dose fractionated versus single dose radiation. A study by CitationShen et al. (1988) revealed higher natural killer cell activity and superior survival in tumor-bearing mice treated with hypofractionated RT as compared to conventionally fractionated RT. All these observations indicate that to date, no conclusive explanation could be given as to which strategy will provide the best platform for combination with CIT approaches.

Another major obstacle to precisely evaluating effects of RT and CIT combinations on tumor progression is posed by the still limited available imaging modalities especially in the clinical setting (CitationKalbasi et al. 2013). Monitoring the successful administration of immunotherapeutic agents and their ability to interact with tumor cells often requires tracking of individual cell populations and therefore asks for labeling techniques in order to distinguish specific immune effector subsets. The optimal characteristics for labeling agents include visualization in a non-invasive manner, minimal toxicity, possibility of serial imaging over longer time periods, specificity, as well as quantitative localization (CitationAkins and Dubey 2008). For this purpose, several molecular imaging agents have been developed – including radioisotopic, fluorescent, bioluminescent, and magnetic resonance imaging (MRI) agents (CitationYoun and Hong 2012). However, choosing an appropriate imaging technique for a given combinatorial treatment strategy has to take into account limitations of each technique. As an example, optical fluorescence or bioluminescence imaging offers high sensitivity, but shows poor penetration in deep tissues, which limits its clinical applicability (CitationLiu and Li 2014). MRI on the other hand has high resolution and contrast while lacking sensitivity (CitationAkins and Dubey 2008, CitationLiu and Li 2014). Positron emission tomography (PET) again offers high sensitivity as well as deep penetration, but suffers from a short half-life of labeling radioisotopes (CitationLiu and Li 2014). Furthermore, powerful combination treatments most likely induce strong inflammatory responses resulting in temporary tissue swelling which can be hard to distinguish from persistent disease if imaging techniques lack high resolution, contrast or sensitivity (CitationKalbasi et al. 2013). Taken together, the tracking of tumor-immune system interactions on cellular level remains challenging, especially in a clinical setting.

Based on the extensive number of advantages that both RT and CIT treatment strategies offer, it seems obvious why there is growing interest in finding the right design of combining these two approaches. As outlined, each individual therapy concept struggles with establishing potent and long-lasting tumor rejection in a large number of patients (CitationFishman 2014, CitationKelderman et al. 2014, CitationRaval et al. 2014). The devil seems to lie in the details of overcoming the tumor's ability to suppress and manipulate the immune system in order to maintain a state of immune evasion. The manifold pathways through which RT has now been shown to interact with immunological mechanisms provide a particularly strong rationale as to why these two forces might represent specifically powerful allies in the ongoing war on cancer.

Acknowledgements

The authors would like to acknowledge A. M. Dohnal (St Anna Children's Cancer Research Institute, Vienna, Austria) for critically reviewing the manuscript. The authors apologize to those colleagues whose studies were not cited in this review due to space limitations.

Funding

This study was supported by Department of Surgery, Massachusetts General Hospital (X.W.), NIH grant RO3 CA141086 (X.W.), the Austrian Science Funds (FWF) project P23271-B11 (K.S.) as well as the PhD program “BioToP – Biomolecular Technology of Proteins” (Austrian Science Funds, FWF project W1224) (K.S.).

Declaration of interest

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Ahmed MM, Hodge JW, Guha C, Bernhard EJ, Vikram B, Coleman CN. 2013. Harnessing the potential of radiation-induced immune modulation for cancer therapy. Cancer Immunol Res 1:280–284.
  • Akins EJ, Dubey P. 2008. Noninvasive imaging of cell-mediated therapy for treatment of cancer. J Nucl Med 49:180s–195s.
  • Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, Yang H, Amigorena S, Ryffel B, Barrat FJ, Saftig P, Levi F, Lidereau R, Nogues C, Mira JP, Chompret A, Joulin V, Clavel-Chapelon F, Bourhis J, Andre F, Delaloge S, Tursz T, Kroemer G, Zitvogel L. 2007. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 13:1050–1059.
  • Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, Abrams J, Sznol M, Parkinson D, Hawkins M, Paradise C, Kunkel L, Rosenberg SA. 1999. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 17:2105–2116.
  • Barker CA, Postow MA. 2014. Combinations of radiation therapy and immunotherapy for melanoma: A review of clinical outcomes. Int J Radiat Oncol Biol Phys 88:986–997.
  • Barker CA, Postow MA, Khan SA, Beal K, Parhar PK, Yamada Y, Lee NY, Wolchok JD. 2013. Concurrent radiotherapy and ipilimumab immunotherapy for patients with melanoma. Cancer Immunol Res 1:92–98.
  • Bernstein MB, Garnett CT, Zhang H, Velcich A, Wattenberg MM, Gameiro SR, Kalnicki S, Hodge JW, Guha C. 2014. Radiation- induced modulation of costimulatory and coinhibitory T-cell signaling molecules on human prostate carcinoma cells promotes productive antitumor immune interactions. Cancer Biother Radiopharm 29:153–161.
  • Brahmer JR, Pardoll DM. 2013. Immune checkpoint inhibitors: Making immunotherapy a reality for the treatment of lung cancer. Cancer Immunol Res 1:85–91.
  • Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM. 2012. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366: 2455–2465.
  • Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, Kim YH, Hoppe RT, Knox SJ, Shin LK, Wapnir I, Tibshirani RJ, Levy R. 2010. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: A phase I/II study. J Clin Oncol 28: 4324–4332.
  • Burnette B, Fu YX, Weichselbaum RR. 2012. The confluence of radiotherapy and immunotherapy. Front Oncol 2:143.
  • Cameron RB, Spiess PJ, Rosenberg SA. 1990. Synergistic antitumor activity of tumor-infiltrating lymphocytes, interleukin 2, and local tumor irradiation. Studies on the mechanism of action. J Exp Med 171:249–263.
  • Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. 2004. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res 64:4328–4337.
  • Chakraborty M, Abrams SI, Camphausen K, Liu K, Scott T, Coleman CN, Hodge JW. 2003. Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol 170:6338–6347.
  • Chakraborty M, Gelbard A, Carrasquillo JA, Yu S, Mamede M, Paik CH, Camphausen K, Schlom J, Hodge JW. 2008a. Use of radiolabeled monoclonal antibody to enhance vaccine-mediated antitumor effects. Cancer Immunol Immunother 57:1173–1183.
  • Chakraborty M, Wansley EK, Carrasquillo JA, Yu S, Paik CH, Camphausen K, Becker MD, Goeckeler WF, Schlom J, Hodge JW. 2008b. The use of chelated radionuclide (samarium-153- ethylenediaminetetramethylenephosphonate) to modulate phenotype of tumor cells and enhance T cell-mediated killing. Clin Cancer Res 14:4241–4249.
  • Chi KH, Liu SJ, Li CP, Kuo HP, Wang YS, Chao Y, Hsieh SL. 2005. Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory hepatoma. J Immunother 28:129–135.
  • Corso CD, Ali AN, Diaz R. 2011. Radiation-induced tumor neoantigens: Imaging and therapeutic implications. Am J Cancer Res 1:390–412.
  • Couzin-Frankel J. 2013. Breakthrough of the year 2013. Cancer immunotherapy. Science 342:1432–1433.
  • Demaria S, Bhardwaj N, McBride WH, Formenti SC. 2005a. Combining radiotherapy and immunotherapy: A revived partnership. Int J Radiat Oncol Biol Phys 63:655–666.
  • Demaria S, Vanpouille-Box C, Formenti SC, Adams S. 2013. The TLR7 agonist imiquimod as an adjuvant for radiotherapy-elicited in situ vaccination against breast cancer. Oncoimmunology 2:e25997.
  • Demaria S, Pilones KA, Vanpouille-Box C, Golden EB, Formenti SC. 2014. The optimal partnership of radiation and immunotherapy: From preclinical studies to clinical translation. Radiat Res 182: 170–181.
  • Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, Formenti SC. 2004. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys 58:862–870.
  • Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, Formenti SC. 2005b. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 11:728–734.
  • Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu YX. 2014. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 124:687–695.
  • Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, Demaria S. 2009. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res 15: 5379–5388.
  • Diamond J, Vanpouille-Box C, Barcellos-Hoff MH, Formenti SC, Demaria S. 2013. The abscopal effect of local radiotherapy is induced by TGFβ blockade. J Immunother Cancer 1:P115.
  • Dillman RO, Fogel GB, Cornforth AN, Selvan SR, Schiltz PM, DePriest C. 2011. Features associated with survival in metastatic melanoma patients treated with patient-specific dendritic cell vaccines. Cancer Biother Radiopharm 26:407–415.
  • Dohnal AM, Witt V, Hugel H, Holter W, Gadner H, Felzmann T. 2007. Phase I study of tumor Ag-loaded IL-12 secreting semi-mature DC for the treatment of pediatric cancer. Cytotherapy 9:755–770.
  • Draghiciu O, Walczak M, Hoogeboom BN, Franken KL, Melief KJ, Nijman HW, Daemen T. 2014. Therapeutic immunization and local low-dose tumor irradiation, a reinforcing combination. Int J Cancer 134:859–872.
  • Finkelstein SE, Iclozan C, Bui MM, Cotter MJ, Ramakrishnan R, Ahmed J, Noyes DR, Cheong D, Gonzalez RJ, Heysek RV, Berman C, Lenox BC, Janssen W, Zager JS, Sondak VK, Letson GD, Antonia SJ, Gabrilovich DI. 2012. Combination of external beam radiotherapy (EBRT) with intratumoral injection of dendritic cells as neo- adjuvant treatment of high-risk soft tissue sarcoma patients. Int J Radiat Oncol Biol Phys 82:924–932.
  • Fishman M. 2014. Challenges facing the development of cancer vaccines. Methods Mol Biol 1139:543–553.
  • Formenti SC, Demaria S. 2009. Systemic effects of local radiotherapy. Lancet Oncol 10:718–726.
  • Formenti SC, Demaria S. 2013. Combining radiotherapy and cancer immunotherapy: A paradigm shift. J Natl Cancer Inst 105:256–265.
  • Frey B, Rubner Y, Kulzer L, Werthmoller N, Weiss EM, Fietkau R, Gaipl US. 2014. Antitumor immune responses induced by ionizing irradiation and further immune stimulation. Cancer Immunol Immunother 63:29–36.
  • Gameiro SR, Jammeh ML, Wattenberg MM, Tsang KY, Ferrone S, Hodge JW. 2014. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 5:403–416.
  • Ganss R, Ryschich E, Klar E, Arnold B, Hammerling GJ. 2002. Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res 62:1462–1470.
  • Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. 2004. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res 64:7985–7994.
  • Grivennikov SI, Greten FR, Karin M. 2010. Immunity, inflammation, and cancer. Cell 140:883–899.
  • Gulley JL, Madan RA, Tsang KY, Arlen PM, Camphausen K, Mohebtash M, Kamrava M, Schlom J, Citrin D. 2011. A pilot safety trial investigating a vector-based vaccine targeting carcinoembryonic antigen in combination with radiotherapy in patients with gastrointestinal malignancies metastatic to the liver. Expert Opin Biol Ther 11:1409–1418.
  • Gulley JL, Arlen PM, Bastian A, Morin S, Marte J, Beetham P, Tsang KY, Yokokawa J, Hodge JW, Menard C, Camphausen K, Coleman CN, Sullivan F, Steinberg SM, Schlom J, Dahut W. 2005. Combining a recombinant cancer vaccine with standard definitive radiotherapy in patients with localized prostate cancer. Clin Cancer Res 11: 3353–3362.
  • Gupta A, Probst HC, Vuong V, Landshammer A, Muth S, Yagita H, Schwendener R, Pruschy M, Knuth A, van den Broek M. 2012. Radiotherapy promotes tumor-specific effector CD8 + T cells via dendritic cell activation. J Immunol 189:558–566.
  • Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A, Mateus C, Boasberg P, Tumeh PC, Chmielowski B, Ebbinghaus SW, Li XN, Kang SP, Ribas A. 2013. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369:134–144.
  • Hanahan D, Weinberg RA. 2011. Hallmarks of cancer: The next generation. Cell 144:646–674.
  • Heery C, Madan R, Bilusic M, Kim J, Singh N, Rauckhorst M, Chen C, Dahut W, Stadler W, Dipaola R, Stein M, Hodge J, Schlom J. 2012. Interim analysis of a phase II randomized clinical trial of samarium-153 (Sm-153) with or without PSA-TRICOM vaccine in metastatic castration-resistant prostate cancer after docetaxel. J Clin Oncol 30:Abstract 2526.
  • Hodge JW, Sharp HJ, Gameiro SR. 2012. Abscopal regression of antigen disparate tumors by antigen cascade after systemic tumor vaccination in combination with local tumor radiation. Cancer Biother Radiopharm 27:12–22.
  • Huang Y, Goel S, Duda DG, Fukumura D, Jain RK. 2013. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res 73:2943–2948.
  • Ina Y, Sakai K. 2005. Activation of immunological network by chronic low-dose-rate irradiation in wild-type mouse strains: Analysis of immune cell populations and surface molecules. Int J Radiat Biol 81:721–729.
  • Kalbasi A, June CH, Haas N, Vapiwala N. 2013. Radiation and immunotherapy: A synergistic combination. J Clin Invest 123: 2756–2763.
  • Kelderman S, Schumacher TN, Haanen JB. 2014. Acquired and intrinsic resistance in cancer immunotherapy. Mol Oncol 8: 1132–1139.
  • Kershaw MH, Devaud C, John LB, Westwood JA, Darcy PK. 2013. Enhancing immunotherapy using chemotherapy and radiation to modify the tumor microenvironment. Oncoimmunology 2:e25962.
  • Kim R, Emi M, Tanabe K. 2006. Functional roles of immature dendritic cells in impaired immunity of solid tumour and their targeted strategies for provoking tumour immunity. Clin Exp Immunol 146:189–196.
  • Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L, Klapproth K, Schakel K, Garbi N, Jager D, Weitz J, Schmitz-Winnenthal H, Hammerling GJ, Beckhove P. 2013. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24:589–602.
  • Kwilas AR, Donahue RN, Bernstein MB, Hodge JW. 2012. In the field: Exploiting the untapped potential of immunogenic modulation by radiation in combination with immunotherapy for the treatment of cancer. Front Oncol 2:104.
  • Lechleider RJ, Arlen PM, Tsang KY, Steinberg SM, Yokokawa J, Cereda V, Camphausen K, Schlom J, Dahut WL, Gulley JL. 2008. Safety and immunologic response of a viral vaccine to prostate-specific antigen in combination with radiation therapy when metronomic-dose interleukin 2 is used as an adjuvant. Clin Cancer Res 14:5284–5291.
  • Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, Beckett M, Sharma R, Chin R, Tu T, Weichselbaum RR, Fu YX. 2009. Therapeutic effects of ablative radiation on local tumor require CD8 + T cells: Changing strategies for cancer treatment. Blood 114:589–595.
  • Lim JY, Gerber SA, Murphy SP, Lord EM. 2014. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol Immunother 63: 259–271.
  • Lipson EJ, Drake CG. 2011. Ipilimumab: An anti-CTLA-4 antibody for metastatic melanoma. Clin Cancer Res 17:6958–6962.
  • Liu Z, Li Z. 2014. Molecular imaging in tracking tumor-specific Cytotoxic T Lymphocytes (CTLs). Theranostics 4:990–1001.
  • Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. 2005. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol 174:7516–7523.
  • Matsumura S, Demaria S. 2010. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat Res 173:418–425.
  • McBride WH, Chiang CS, Olson JL, Wang CC, Hong JH, Pajonk F, Dougherty GJ, Iwamoto KS, Pervan M, Liao YP. 2004. A sense of danger from radiation. Radiat Res 162:1–19.
  • McDermott DF. 2007. Update on the application of interleukin-2 in the treatment of renal cell carcinoma. Clin Cancer Res 13:716s–720s.
  • Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, Metivier D, Larochette N, van Endert P, Ciccosanti F, Piacentini M, Zitvogel L, Kroemer G. 2007. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 13:54–61.
  • Pardoll DM. 2012. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12:252–264.
  • Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, Mu Z, Rasalan T, Adamow M, Ritter E, Sedrak C, Jungbluth AA, Chua R, Yang AS, Roman RA, Rosner S, Benson B, Allison JP, Lesokhin AM, Gnjatic S, Wolchok JD. 2012. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 366:925–931.
  • Raval RR, Sharabi AB, Walker AJ, Drake CG, Sharma P. 2014. Tumor immunology and cancer immunotherapy: Summary of the 2013 SITC primer. J Immunother Cancer 2:14.
  • Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, Camphausen K, Luiten RM, de Ru AH, Neijssen J, Griekspoor A, Mesman E, Verreck FA, Spits H, Schlom J, van Veelen P, Neefjes JJ. 2006. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 203:1259–1271.
  • Roses RE, Datta J, Czerniecki BJ. 2014. Radiation as immunomodulator: Implications for dendritic cell-based immunotherapy. Radiat Res 182:211–218.
  • Safwat A, Aggerholm N, Roitt I, Overgaard J, Hokland M. 2003. Low-dose total body irradiation augments the therapeutic effect of interleukin-2 in a mouse model for metastatic malignant melanoma. J Exp Ther Oncol 3:161–168.
  • Safwat A, Aggerholm N, Roitt I, Overgaard J, Hokland M. 2004. Tumour burden and interleukin-2 dose affect the interaction between low-dose total body irradiation and interleukin 2. Eur J Cancer 40: 1412–1417.
  • Schaue D, Ratikan JA, Iwamoto KS, McBride WH. 2012. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys 83:1306–1310.
  • Schreiber RD, Old LJ, Smyth MJ. 2011. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 331:1565–1570.
  • Schwartz RN, Stover L, Dutcher J. 2002. Managing toxicities of high-dose interleukin-2. Oncology (Williston Park) 16:11–20.
  • Seung SK, Curti BD, Crittenden M, Urba W. 2012a. Radiation and immunotherapy: Renewed allies in the war on cancer. Oncoimmunology 1:1645–1647.
  • Seung SK, Curti BD, Crittenden M, Walker E, Coffey T, Siebert JC, Miller W, Payne R, Glenn L, Bageac A, Urba WJ. 2012b. Phase 1 study of stereotactic body radiotherapy and interleukin-2 – tumor and immunological responses. Sci Transl Med 4:137ra74.
  • Shen RN, Hornback NB, Shidnia H, Lu L, Montebello JF, Brahmi Z. 1988. A comparison of lung metastases and natural killer cell activity in daily fractions and weekly fractions of radiation therapy on murine B16a melanoma. Radiat Res 114:354–360.
  • Shigematsu A, Adachi Y, Koike-Kiriyama N, Suzuki Y, Iwasaki M, Koike Y, Nakano K, Mukaide H, Imamura M, Ikehara S. 2007. Effects of low-dose irradiation on enhancement of immunity by dendritic cells. J Radiat Res 48:51–55.
  • Siegel JP, Puri RK. 1991. Interleukin-2 toxicity. J Clin Oncol 9:694–704.
  • Slovin S, Beer T, Higano CS, Tejwani S, Hamid O, Picus J, Harzstark A, Scher HI, Lan Z, Lowy I. 2009. Initial phase II experience of ipilimumab (IPI) alone and in combination with radiotherapy (XRT) in patients with metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 27: Abstract 5138.
  • Slovin SF, Higano CS, Hamid O, Tejwani S, Harzstark A, Alumkal JJ, Scher HI, Chin K, Gagnier P, McHenry MB, Beer TM. 2013. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: Results from an open-label, multicenter phase I/II study. Ann Oncol 24:1813–1821.
  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. 2012. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366: 2443–2454.
  • Wattenberg MM, Fahim A, Ahmed MM, Hodge JW. 2014. Unlocking the combination: Potentiation of radiation-induced antitumor responses with immunotherapy. Radiat Res 182:126–138.
  • Witek M, Blomain ES, Magee MS, Xiang B, Waldman SA, Snook AE. 2014. Tumor radiation therapy creates therapeutic vaccine responses to the colorectal cancer antigen GUCY2C. Int J Radiat Oncol Biol Phys 88:1188–1195.
  • Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I, Inzunza HD, Feely W, Horak CE, Hong Q, Korman AJ, Wigginton JM, Gupta A, Sznol M. 2013. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369:122–133.
  • Youn H, Hong KJ. 2012. In vivo non invasive molecular imaging for immune cell tracking in small animals. Immune Netw 12:223–229.