Skip to main content

EDITORIAL article

Front. Immunol., 06 September 2019
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Combinatorial Approaches to Enhance Anti-Tumor Immunity: focus on Immune checkpoint blockade therapy View all 25 articles

Editorial: Combinatorial Approaches to Enhance Anti-tumor Immunity: Focus on Immune Checkpoint Blockade Therapy

  • 1Edwin L. Steele Laboratories, Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
  • 2Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
  • 3Medical Science Division, Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
  • 4Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany

Introduction

The advent of immunotherapy (IT), especially immune checkpoint blockers (ICBs), and its application in oncology has provided new hope for cancer patients. However, despite the rapid progress in the field of immunoncology, only a subset of patients currently benefit from these therapies. Many challenges remain to be resolved in order for IT to display optimal efficacy and good overall response rates in patients. First, many tumors have low tumor mutational burden (TMB), and therefore only produce limited antigens that can be recognized by endogenous T cells (1). Second, reduced antigen release or downregulation of antigen presentation machinery contributes to immune escape, leading to tumors with scarce numbers of infiltrating immune cells, indicating that reinvigoration of the pre-existing pool of anti-tumor T cells by ICBs may not be enough to induce tumor regression (2, 3). And third, even if the number and activity of T cells are successfully boosted by immunostimulatory therapies, the immunosuppressive tumor microenvironment (TME) restricts durable responses and contributes to treatment resistance (4). To overcome these limitations, new strategies are needed. Currently, several approaches exist where IT is combined with standard-of-care therapies, including radiotherapy (RT) and/or anti-angiogenic therapy (AAT) that are being evaluated in both preclinical and clinical settings. The aim of this article collection is to provide a comprehensive overview of recent developments and approaches in enhancing anti-tumor immunity with the focus on potential synergistic effects of RT and/or AAT with IT, ultimately supporting the rationale of combining IT with AAT and RT.

Strategy 1: Increase Antigen Production, Release and Presentation

Recent findings indicate that high TMB is positively correlated to ICB responses across different types of tumors (5). DNA damaging therapies such as standard-of-care RT can be applied to induce reactive oxygen species (ROS), leading to immunogenic cell death and antigen release (6). Another strategy involves using RT at sublethal levels to induce mutations to increase antigens, which would aid in immune recognition (7, 8). However, standard dosing of RT could be immunosuppressive by direct effects on lymphocytes and dendritic cells (DCs) (9). In contrast, recent evidence suggests that stereotactic body radiation therapy increases T cell activity and reduces inhibitory stroma in tumors (Menon et al.). It has been demonstrated that tumor-derived exosomes successfully delivered double-stranded DNA and induced IFN-mediated T cell responses more efficiently in irradiated mice (10). Furthermore, through increased recruitment and activity of DCs, owing to RT-induced expression of vascular endothelial cell adhesion protein 1 on the endothelium and CXCL16 in tumor cells (11, 12), RT could also directly promote T cell activation and priming. RT has been shown to stimulate the production of type I interferons (IFNs), leading to increased number of CD8α+ tumor infiltrating DCs and subsequent boost in antigen presentation and T cell priming (1315). Interestingly, selecting the optimal dose seems to be crucial to determine the anti-tumor response. For example, high-dose RT (20 Gy x 2) prevented beneficial production of type I IFNs by induction of Trex1, which degrades double-stranded DNA released by radiation-induced tumor cell death (16). Nevertheless, it will be important to assess the immune response to RT in individual patients/tumors, which has recently been reviewed elsewhere (17).

Additionally, the TME seems to play an important role in antigen presentation by regulating DC function. Jiang et al. found that high tumor cell-intrinsic expression of FASN (fatty acid synthase) led to increased lipid accumulation in DCs, which reduced their antigen presenting capacity in an ovarian cancer model. As reported, blocking FASN increased T cell infiltration, hence, it would be reasonable to speculate that ICBs may be rendered more effective.

Strategy 2: Vaccination

Instead of strategies aiming to increase the antigenicity of tumors by killing tumor cells or increasing their mutational load, vaccines can be utilized to take advantage of pre-existing alterations in tumors. Vaccines come in different flavors, including whole tumor cell lysates, synthesized proteins or peptides, viral vectors expressing tumor antigens, or DC-based vaccines. Utilizing pulsed mature DC-based vaccines could potentially overcome some of the immunosuppressive cues, which otherwise could reduce vaccine efficacy by limiting DC migration, maturation and antigen presentation (18). Unfortunately, only limited benefits with therapeutic vaccine monotherapy have been observed clinically. Even if the vaccines themselves successfully circumvent antigen presentation and priming of T cells, downstream obstacles of immunosuppression could still remain. Metabolic reprogramming in the TME could play an essential role in immunosuppression. For example, by depleting tryptophan and producing kynurenine, indoleamine 2,3-dioxygenase (IDO) promotes the generation of immunosuppressive regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) (19). Experimentally, Moreno et al., show that treatment of HPV+ tumors with immunometabolic adjuvants (such as IDO inhibitors) could induce a therapeutic benefit of an otherwise ineffective HPV-16 vaccine. However, as Eleftheriadis elaborates on in his opinion piece, IDO inhibitors have so far failed clinically and researchers are currently trying to understand why.

These preclinical and clinical lessons collectively suggest that combinatorial approaches could offer great clinical benefits to boost vaccines. Mougel et al. discuss the rationale for combining vaccines with AAT or ICBs to overcome tumor-employed immune escape mechanisms, with a focus on current clinical efforts. In addition, van Gulijk et al. provide an overview specifically on DC-based vaccines and strategies to combine with chemotherapy, RT and ICBs.

Strategy 3: Increase T Cell Infiltration

Tumor tissues typically display limited number and heterogeneous distribution of T cells. Leukocyte infiltration is an active process that can be facilitated or hindered by the endothelium of blood vessels. Blood vessels are critical mediators of inflammation by providing a direct interface with which immune cells interact to gain access to tissues. Upon inflammatory cues, the endothelial cells lining the inner surface of blood vessels will express adhesion molecules and soluble mediators of leukocyte trafficking. In tumors, however, the immature nature of blood vessels can cause endothelial anergy, a state of lymphocyte tolerance characterized by repression of adhesion molecules, leading to failure of leukocyte trafficking (2022). Klein provides a detailed review specifically on the tumor endothelium, with its implications for combination therapies using RT or IT. The endothelial-immune interface provides an opportunity for intervention, where AAT could be applied to increase the influx of anti-tumor immune cells. Strategies to normalize tumor vessels, with an overview on current preclinical and clinical efforts, and potential synergy with IT are discussed by Georganaki et al.. Furthermore, Amin and Hammers reviewed the clinical data of combining various AAT drugs with IT in advanced renal cell cancer patients, where AAT has shown particular benefits owing to high intrinsic VEGF-VEGFR signaling.

Alternative strategies could be employed to enhance T cell infiltration. For example, by performing gene expression analysis to look for correlations to immune profiles, Roszik et al. identified STAT3 as a promising target in cervical cancer. High STAT3 expression was inversely correlated with CD8+ T cell density, implying STAT3 as a promising target to enhance anti-tumor immunity (Roszik et al.). In fact, several clinical trials are investigating STAT3 inhibition. For example, one phase II trial specifically is testing the potential synergy of STAT3 inhibition with anti-PD1 (programmed cell death 1) in colorectal cancer patients (NCT03647839). Another promising approach is specific tumor cell-targeting by utilizing heat-shock-proteins (HSPs), which are overexpressed in various cancers and associated with aggressive phenotypes and poor prognosis (2326). Circulating levels of HSP70 could serve as prognostic markers (27). HSP70 for instance has been shown to successfully predict response after RT in advanced NSCLC (non-small cell lung cancer) and might serve as a therapeutic target to stimulate anti-tumor natural killer (NK) cell responses (2830). Indeed, Shevtsov et al. observed a robust increase in infiltrating CD8+ T cells following adoptive transfer of ex vivo HSP70-activated NK cells in lung and glioma mouse models. Interestingly, survival benefits were further enhanced by the addition of anti-PD1 therapy (Shevtsov et al.). The exact underlying mechanisms for the described phenotype remain to be determined. However, NK cells can trigger cell death by both apoptosis and necrosis (31), which can lead to activation of the cGAS-STING (cyclic GMP-AMP synthase-stimulator of interferon genes) pathway. The subsequent production of type I IFN has been shown to drive infiltration of CD4+ and CD8+ T cells into tumors (3234). Although highlighted in the context of RT, Goedegebuure et al. provide a schematic overview of the immune impact of cGAS-STING activation (Goedegebuure et al.: Figure 1). Paradoxically, RT-induced STING activation could also increase MDSCs via CCL2 production, thereby dampening CD8+ T cell activity (Darragh et al.: Figure 2). There are multiple ongoing clinical trials looking at RT and ICB therapies in NSCLC and head and neck squamous cell carcinoma patients, as reviewed by Nardone et al., which will provide important information on how to optimally design the treatment modalities with RT and IT. Interestingly, two recent phase II studies in NSCLC patients looking at adjuvant anti-PD1 therapy after RT, with or without other prior local ablative therapies, reported a promising although non-significant doubling in overall response rates (35) and an impressive increase in progression-free survival (36), thereby highlighting the potential of combining RT with IT.

Strategy 4: Alleviate Immunosuppression

Tumors are able to employ various resistance mechanisms to evade immune surveillance. The abnormal vasculature is one of the contributors of an immunosuppressive TME (37). The lack of perivascular coverage in tumor blood vessels and high interstitial fluid pressure in tumor tissues often result in malfunctioning or collapsed blood vessels. This results in tumor tissues experiencing high levels of hypoxia, which is one of the main drivers of immunosuppression (38, 39). While tumor cells can readily adapt to the low levels of oxygen, hypoxia also affects the phenotype of stromal cells and immune cells. For example, Tregs and MDSCs have been shown to gain further immunosuppressive capacity (40, 41), and macrophages polarize toward a tumor-promoting phenotype (TAMs) (42) under hypoxic conditions (Figure 6: Darragh et al.). By normalizing the vasculature using AAT, hypoxia can be reduced, and can thereby alleviate immunosuppression (43, 44). The increase in tissue perfusion and oxygenation will also increase the potential impact of RT by optimizing the generation of ROS. As reviewed by Goedegebuure et al., there is a reciprocal relationship where RT, in turn, can have a positive or negative impact on blood vessels and perfusion, depending on the dose and scheduling.

Although vessel normalization by AAT can indirectly improve the immunosuppressive TME, there are ways to directly target and reprogram the immune cells. Focusing on Tregs, Nagai et al. identified PRMT5 (protein arginine methyltransferase) as an interaction partner of FoxP3, a transcription factor important for Treg function. Pharmacological inhibition of PRMT5 led to reduced immunosuppressive activity in Tregs and inhibition of tumor growth (Nagai et al.). Another strategy to reprogram the immune compartment to an anti-tumor phenotype could be to provide IL-2, which would enhance cells such as CD8+ T cells and NK cells (45, 46). However, IL-2 therapy also stimulates Tregs (47), and has been limited by systemic toxicity (48). Mortara et al. reviewed the current efforts of using antibody-cytokine fusion proteins with IL-2 (so-called immunocytokines), designed to be tumor-targeting to overcome these previous limitations and hinder tumor progression by stimulating anti-tumor immunity. In addition to targeting the components of the adaptive immunity, several ongoing trials are investigating the therapeutic benefits of targeting cells of the innate immunity. Specifically, myeloid cells are strong contributors to immunosuppression, especially in glioma, which is the topic covered by Ding et al.. More generally, Dar et al. provided an overview of strategies to target the innate immunity to overcome resistance to RT, with a focus on the interplay between innate and adaptive immunity (see schematic summary in Dar et al.: Figure 1). Furthermore, Menon et al. focused on the stromal contributions to immune evasion and the immunomodulatory properties of RT as an important part of combinatorial treatment modalities. One debated potential effect of RT is the so-called radiation-induced “memory effect” by which prior RT is reported to enhance subsequent anti-tumor immune responses during, for example, ICB therapy. Retrospective analysis by Chen et al. in NSCLC patients suggested that previous RT improved the response to IL-2 infusion, which they attributed to a radiation-induced “memory effect”.

Strategy 5: Overcome Resistance

As with most therapeutic interventions, intrinsic or acquired resistance is the major obstacle for the success of RT, AAT, and IT. Knowledge of specific tumor-employed resistance mechanisms can offer a strong rational for combinatorial approaches. Darragh et al. discuss several TME-related resistance mechanisms upon RT. For example, RT-induced cell death leads to the release of ATP (adenosine triphosphate), which stimulates DC recruitment and activation (49). However, ATP is catabolized to adenosine by CD39/CD73, which is frequently upregulated on tumor cells and in the TME (50). In contrast to ATP, adenosine is immunosuppressive by limiting DCs and CD8+ T cells, and by simultaneously promoting Tregs and TAMs (Darragh et al.: Figure 4). The review by de Leve et al. highlights the therapeutic potential of targeting CD73/adenosine in cancer to improve RT responses.

To optimally target tumor cells, it has become clear that stem-like (so-called tumor-initiating) cells need to be specifically targeted as they represent a highly resistant population of cells (51, 52). Expression of SDF1 (CXCL12) and its receptor CXCR4 has been linked to stem cell niches where its signaling likely contributes to a stem-like phenotype (53). Hence, RT could greatly benefit from combination strategies with CXCR4-targeting approaches to eliminate resistant clones. The therapeutic potential of such combinations is reviewed by Eckert et al.. Another factor involved in stem cell renewal is TGF-β (transforming growth factor β), which also plays an important role in promoting immunosuppression and fibrosis. Blocking TGF-β by therapeutic antibodies has been shown to slow tumor progression, increase infiltration of T cells and synergize with ICB therapy (54, 55). Rossowska et al. took a different approach in which they modified MC38 tumor cells to secrete exosomes deprived of TGF-β1 (by expressing shRNA) and subsequently using those exosomes as treatment of wildtype MC38 tumors. In doing so, the authors observed a reduction in tumor progression, which was accompanied by increased anti-tumor immunity, thereby highlighting the therapeutic value of targeting TGF-β (Rossowska et al.).

As a concluding remark, antibodies targeting PD1/PD-L1 (programmed cell death ligand 1), with FDA approval in multiple indications have so far shown the most promise in patients. However, resistance is a major hurdle and we are only just beginning to understand the underlying mechanisms. Yao et al. report how anti-PD1 therapy can promote tumor cell proliferation if the tumor cells show intrinsic PD1 expression. In light of such findings, we need to carefully evaluate how to assess PD1/PD-L1 expression before stratifying patients for treatment. Ongoing clinical efforts are indicating that simultaneous targeting of several immune checkpoints, such as CTLA-4 (cytotoxic T-lymphocyte-associated antigen-4), Lag-3 and Tim-3, could offer significant advantages over single ICB therapies. Khair et al. provide an exhaustive overview on this topic.

Summary

One major concern when treating patients with ICBs, such as anti-PD1 and anti-CTLA-4 antibodies, is the high frequency of immune-related adverse events. This, along with lacking a reliable biomarker for patient stratification, underscores the need for multimodal therapy allowing for the use of lower doses and implementation of standard operating procedures to manage these side-effects without compromising efficacy. However, as is evident throughout the contributions in this article collection, several important outstanding questions remain to be fully addressed including optimal dosage, timing, and scheduling for these combinatorial approaches.

Author Contributions

Both authors have actively participated in shaping the idea for the article collection, recruiting authors, and acting as editors for several of the contributions. PA and CO wrote the editorial together and made final edits.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. (2018) 6:157. doi: 10.1186/s40425-018-0479-7

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Campoli M, Ferrone S. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. (2008) 27:5869–85. doi: 10.1038/onc.2008.273

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Iorgulescu JB, Braun D, Oliveira G, Keskin DB, Wu CJ. Acquired mechanisms of immune escape in cancer following immunotherapy. Genome Med. (2018) 10:87. doi: 10.1186/s13073-018-0598-2

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res. (2015) 21:687–92. doi: 10.1158/1078-0432.CCR-14-1860

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. (2019) 51:202–6. doi: 10.1038/s41588-018-0312-8

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Halperin EC, Perez CA, Brady LW. Perez and Brady's Principles and Practice of Radiation Oncology. 5th ed. Philadelphia, PA: Wolters Kluwer Health; Lippincott Williams & Wilkins (2008). p. xxxii, 2106.

7. Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. (2004) 64:7985–94. doi: 10.1158/0008-5472.CAN-04-1525

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. (2006) 203:1259–71. doi: 10.1084/jem.20052494

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Ebner DK, Tinganelli W, Helm A, Bisio A, Yamada S, Kamada T, et al. The immunoregulatory potential of particle radiation in cancer therapy. Front Immunol. (2017) 8:99. doi: 10.3389/fimmu.2017.00099

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Diamond JM, Vanpouille-Box C, Spada S, Rudqvist NP, Chapman JR, Ueberheide BM, et al. Exosomes shuttle TREX1-sensitive IFN-stimulatory dsDNA from irradiated cancer cells to DCs. Cancer Immunol Res. (2018) 6:910–20. doi: 10.1158/2326-6066.CIR-17-0581

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Matsumura S, Demaria S. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat Res. (2010) 173:418–25. doi: 10.1667/RR1860.1

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. (2005) 174:7516–23. doi: 10.4049/jimmunol.174.12.7516

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Burnette BC, Liang H, Lee Y, Chlewicki L, Khodarev NN, Weichselbaum RR, et al. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. (2011) 71:2488–96. doi: 10.1158/0008-5472.CAN-10-2820

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J Exp Med. (2011) 208:2005–16. doi: 10.1084/jem.20101159

CrossRef Full Text | Google Scholar

15. Lim JY, Gerber SA, Murphy SP, Lord EM. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol Immunother. (2014) 63:259–71. doi: 10.1007/s00262-013-1506-7

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. (2017) 8:15618. doi: 10.1038/ncomms15618

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Grassberger C, Ellsworth SG, Wilks MQ, Keane FK, Loeffler JS. Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol. (2019). doi: 10.1038/s41571-019-0238-9. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Gilboa E. DC-based cancer vaccines. J Clin Invest. (2007) 117:1195–203. doi: 10.1172/JCI31205

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Holmgaard RB, Zamarin D, Li Y, Gasmi B, Munn DH, Allison JP, et al. Tumor-expressed IDO recruits and activates MDSCs in a Treg-dependent manner. Cell Rep. (2015) 13:412–24. doi: 10.1016/j.celrep.2015.08.077

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D, Muthukuru M, et al. Human squamous cell carcinomas evade the immune response by down-regulation of vascular E-selectin and recruitment of regulatory T cells. J Exp Med. (2008) 205:2221–34. doi: 10.1084/jem.20071190

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Griffioen AW, Damen CA, Blijham GH, Groenewegen G. Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood. (1996) 88:667–73.

PubMed Abstract | Google Scholar

22. Piali L, Fichtel A, Terpe HJ, Imhof BA, Gisler RH. Endothelial vascular cell adhesion molecule 1 expression is suppressed by melanoma and carcinoma. J Exp Med. (1995) 181:811–6. doi: 10.1084/jem.181.2.811

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Nanbu K, Konishi I, Mandai M, Kuroda H, Hamid AA, Komatsu T, et al. Prognostic significance of heat shock proteins HSP70 and HSP90 in endometrial carcinomas. Cancer Detect Prev. (1998) 22:549–55. doi: 10.1046/j.1525-1500.1998.00069.x

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Xie K, Huang S. Regulation of cancer metastasis by stress pathways. Clin Exp Metast. (2003) 20:31–43. doi: 10.1023/A:1022590402748

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Stangl S, Tontcheva N, Sievert W, Shevtsov M, Niu M, Schmid TE, et al. Heat shock protein 70 and tumor-infiltrating NK cells as prognostic indicators for patients with squamous cell carcinoma of the head and neck after radiochemotherapy: a multicentre retrospective study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer. (2018) 142:1911–25. doi: 10.1002/ijc.31213

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Gunther S, Ostheimer C, Stangl S, Specht HM, Mozes P, Jesinghaus M, et al. Correlation of Hsp70 serum levels with gross tumor volume and composition of lymphocyte subpopulations in patients with squamous cell and adeno non-small cell lung cancer. Front Immunol. (2015) 6:556. doi: 10.3389/fimmu.2015.00556

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Bayer C, Liebhardt ME, Schmid TE, Trajkovic-Arsic M, Hube K, Specht HM, et al. Validation of heat shock protein 70 as a tumor-specific biomarker for monitoring the outcome of radiation therapy in tumor mouse models. Int J Radiat Oncol Biol Phys. (2014) 88:694–700. doi: 10.1016/j.ijrobp.2013.11.008

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Gehrmann M, Specht HM, Bayer C, Brandstetter M, Chizzali B, Duma M, et al. Hsp70–a biomarker for tumor detection and monitoring of outcome of radiation therapy in patients with squamous cell carcinoma of the head and neck. Radiat Oncol. (2014) 9:131. doi: 10.1186/1748-717X-9-131

CrossRef Full Text | Google Scholar

29. Ostheimer C, Gunther S, Bache M, Vordermark D, Multhoff G. Dynamics of heat shock protein 70 serum levels as a predictor of clinical response in non-small-cell lung cancer and correlation with the hypoxia-related marker osteopontin. Front Immunol. (2017) 8:1305. doi: 10.3389/fimmu.2017.01305

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Specht HM, Ahrens N, Blankenstein C, Duell T, Fietkau R, Gaipl US, et al. Heat shock protein 70 (Hsp70) Peptide activated natural killer (NK) cells for the treatment of patients with non-small cell lung cancer (NSCLC) after radiochemotherapy (RCTx) - From preclinical studies to a clinical phase II trial. Front Immunol. (2015) 6:162. doi: 10.3389/fimmu.2015.00162

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Backes CS, Friedmann KS, Mang S, Knörck A, Hoth M, Kummerow C. Natural killer cells induce distinct modes of cancer cell death: discrimination, quantification, and modulation of apoptosis, necrosis, and mixed forms. J Biol Chem. (2018) 293:16348–63. doi: 10.1074/jbc.RA118.004549

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Pantelidou C, Sonzogni O, De Oliveria Taveira M, Mehta AK, Kothari A, Wang D, et al. PARP inhibitor efficacy depends on CD8(+) T-cell recruitment via intratumoral STING pathway activation in BRCA-deficient models of triple-negative breast cancer. Cancer Discov. (2019) 9:722–37. doi: 10.1158/1538-7445.SABCS18-4490

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Sen T, Rodriguez BL, Chen L, Corte CMD, Morikawa N, Fujimoto J, et al. Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung cancer. Cancer Discov. (2019) 9:646–61. doi: 10.1158/2159-8290.CD-18-1020

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Wang Z, Sun K, Xiao Y, Feng B, Mikule K, Ma X, et al. Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models. Sci Rep. (2019) 9:1853. doi: 10.1038/s41598-019-38534-6

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Theelen WSME, Peulen HMU, Lalezari F, van der Noort V, de Vries JF, Aerts JGJV, et al. Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: results of the PEMBRO-RT phase 2 randomized clinical trial. JAMA Oncol. (2019). doi: 10.1001/jamaoncol.2019.1478. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Bauml JM, Mick R, Ciunci C, Aggarwal C, Davis C, Evans T, et al. Pembrolizumab after completion of locally ablative therapy for oligometastatic non-small cell lung cancer: a phase 2 trial. JAMA Oncol. (2019). doi: 10.1001/jamaoncol.2019.1449. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Kim R, Emi M, Tanabe K, Arihiro K. Tumor-driven evolution of immunosuppressive networks during malignant progression. Cancer Res. (2006) 66:5527–36. doi: 10.1158/0008-5472.CAN-05-4128

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Caldwell CC, Kojima H, Lukashev D, Armstrong J, Farber M, Apasov SG, et al. Differential effects of physiologically relevant hypoxic conditions on T lymphocyte development and effector functions. J Immunol. (2001) 167:6140–9. doi: 10.4049/jimmunol.167.11.6140

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Siemens DR, Hu N, Sheikhi AK, Chung E, Frederiksen LJ, Pross H, et al. Hypoxia increases tumor cell shedding of MHC class I chain-related molecule: role of nitric oxide. Cancer Res. (2008) 68:4746–53. doi: 10.1158/0008-5472.CAN-08-0054

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Ben-Shoshan J, Maysel-Auslender S, Mor A, Keren G, George J. Hypoxia controls CD4+CD25+ regulatory T-cell homeostasis via hypoxia-inducible factor-1alpha. Eur J Immunol. (2008) 38:2412–8. doi: 10.1002/eji.200838318

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. (2010) 207:2439–53. doi: 10.1084/jem.20100587

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Lewis C, Murdoch C. Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. Am J Pathol. (2005) 167:627–35. doi: 10.1016/S0002-9440(10)62038-X

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Huang Y, Goel S, Duda DG, Fukumura D, Jain RK. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res. (2013) 73:2943–8. doi: 10.1158/0008-5472.CAN-12-4354

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Huang Y, Yuan J, Righi E, Kamoun WS, Ancukiewicz M, Nezivar J, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA. (2012) 109:17561–6. doi: 10.1073/pnas.1215397109

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Henney CS, Kuribayashi K, Kern DE, Gillis S. Interleukin-2 augments natural killer cell activity. Nature. (1981) 291:335–8. doi: 10.1038/291335a0

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Rollings CM, Sinclair LV, Brady HJM, Cantrell DA, Ross SH. Interleukin-2 shapes the cytotoxic T cell proteome and immune environment-sensing programs. Sci Signal. (2018) 11:526. doi: 10.1126/scisignal.aap8112

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Sim GC, Martin-Orozco N, Jin L, Yang Y, Wu S, Washington E, et al. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J Clin Invest. (2014) 124:99–110. doi: 10.1172/JCI46266

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Jarkowski A, Wong MKK. A re-assessment of the safety and efficacy of interleukin-2 for the treatment of renal cell carcinoma. Clin Med Ther. (2009). doi: 10.4137/CMT.S2037

CrossRef Full Text | Google Scholar

49. Kojima S, Ohshima Y, Nakatsukasa H, Tsukimoto M. Role of ATP as a key signaling molecule mediating radiation-induced biological effects. Dose Response. (2017) 15:1559325817690638. doi: 10.1177/1559325817690638

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev. (2017) 276:121–44. doi: 10.1111/imr.12528

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Phi LTH, Sari IN, Yang YG, Lee SH, Jun N, Kim KS, et al. Cancer Stem Cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. (2018) 2018:5416923. doi: 10.1155/2018/5416923

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Miao Y, Yang H, Levorse J, Yuan S, Polak L, Sribour M, et al. Adaptive immune resistance emerges from tumor-initiating stem cells. Cell. (2019) 177:1172–86 e14. doi: 10.1016/j.cell.2019.03.025

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Trautmann F, Cojoc M, Kurth I, Melin N, Bouchez LC, Dubrovska A, et al. CXCR4 as biomarker for radioresistant cancer stem cells. Int J Radiat Biol. (2014) 90:687–99. doi: 10.3109/09553002.2014.906766

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. (2018) 554:538–43. doi: 10.1038/nature25492

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. (2018) 554:544–8. doi: 10.1038/nature25501

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cancer, immunotherapy, radiotherapy, antiangiogenic therapy, immune checkpoint blockade (ICB), cancer vaccine

Citation: Andersson P and Ostheimer C (2019) Editorial: Combinatorial Approaches to Enhance Anti-tumor Immunity: Focus on Immune Checkpoint Blockade Therapy. Front. Immunol. 10:2083. doi: 10.3389/fimmu.2019.02083

Received: 24 July 2019; Accepted: 16 August 2019;
Published: 06 September 2019.

Edited and reviewed by: Catherine Sautes-Fridman, INSERM U1138 Centre de Recherche des Cordeliers, France

Copyright © 2019 Andersson and Ostheimer. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Christian Ostheimer, christian.ostheimer@uk-halle.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.