Next Article in Journal
A Novel Digestive α-Amylase from Blue Crab (Portunus segnis) Viscera: Purification, Biochemical Characterization and Application for the Improvement of Antioxidant Potential of Oat Flour
Previous Article in Journal
Titanium Wear of Dental Implants from Placement, under Loading and Maintenance Protocols
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiation and Immunotherapy in Upper Gastrointestinal Cancers: The Current State of Play

1
Department of Surgery, School of Medicine, Trinity College Dublin, Dublin 8, Ireland
2
Trinity St James’ Cancer Institute, St James’s Hospital Dublin, Dublin 8, Ireland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(3), 1071; https://doi.org/10.3390/ijms22031071
Submission received: 17 December 2020 / Revised: 15 January 2021 / Accepted: 18 January 2021 / Published: 22 January 2021
(This article belongs to the Section Molecular Immunology)

Abstract

:
Radiotherapy remains one of the contemporary cornerstones of cancer treatment in the neoadjuvant, curative, adjuvant and palliative settings, either in isolation or as a multimodal approach. Moreover, recent advances in targeted immune checkpoint therapy have firmly established immunotherapy as the fourth pillar in cancer therapy alongside surgery, chemotherapy and notably radiotherapy. There is emerging evidence to suggest both radioresistance and reduced efficacy of immune checkpoint blockade (ICB) are potentiated by the tumour microenvironment (TME) and in fact modulating aspects of this immunosuppressive milieu is instrumental to unlocking anti-tumour immunity. The response rates of Upper Gastrointestinal (UGI) malignancies to ICB remains modest at 10–15%, compared to melanoma at 20–40%. Harnessing the effects of radiotherapy through remodelling of the TME using ICB as a radiosensitisor is an avenue showing promise. Here we explore the rationale behind combining radiotherapy with ICB, as a symbiotic relationship in shifting the balance in favour of anti-tumour immunity. We discuss the effects of radiotherapy on immunogenic cell death, the concept of the abscopal effect, the importance of the cGAS STING pathway, and their relevance in the context of the tumour microenvironment. Furthermore, dosing and timing of radiotherapy and ICB is now being evaluated for its synergistic effects on host tumour immunity, and we review the ongoing efforts and current available literature for single agent and dual agent ICB in combination multimodal therapy for both locally advanced operable and metastatic disease of the upper gastrointestinal tract.

1. Introduction

After years of effort to harness the immune system for the treatment of cancer, the advent of antibodies which target ‘immune checkpoints’, including programmed cell death protein 1 (PD-1) and cytotoxic lymphocyte antigen 4 (CTLA-4), has increased interest in immunological aspects of conventional therapies. These immune checkpoint inhibitors (ICIs), including pembrolizumab, tislelizumab, nivolumab (anti PD-1) and ipilimumab (anti CTLA-4), have led to dramatic and durable clinical responses in diverse cancers [1]. Cancers of the upper gastrointestinal tract (UGI) are common globally and account for a disproportionately high incidence of cancer-related mortality. In 2019, UGI malignancies accounted for 9% of cancer diagnoses and 13.5% of cancer related deaths worldwide [2]. The response rates for clinically approved ICIs in melanoma and non-small cell lung cancer are approximately 20–40% [2]; however, in UGI cancers, this decreases to 10–15% and ICIs are therefore largely confined to salvage treatment of advanced disease, with its use in the neoadjuvant and curative setting confined to select cases [3].
Radiotherapy has been one of the pillars for the management of neoplastic burden in cancer patients for over a century. It is used as a treatment modality in approximately 50% of cancer patients in the neoadjuvant, adjuvant, curative or palliative settings [4]. Tri-modality treatment of surgery, chemotherapy and radiotherapy is the standard of care in oesophageal adenocarcinoma (OAC) and oesophageal squamous cell carcinoma (OSCC), while in gastric cancer (GC) adjuvant chemotherapy use depends on surgical margins and the extent of lymph node dissection [5]. Radiation triggers DNA damage-induced cell death in cancer cells but can also modify the antigenicity and the adjuvanticity of tumours. This is by activating cytosolic DNA sensors, inducing immunogenic cell death, enhancing neoantigen expression and modulating the tumour microenvironment (TME) [6]. Therefore, combining immuno-oncology approaches with radiation could boost response to ICI in UGI cancers (Figure 1). In this review, we describe the mechanistic rationale for combining immunotherapy and radiotherapy, clinical trials of radio-immunotherapy in gastroesophageal malignancies and current strategies to optimise radiation regimen efficacy while minimising toxicity.

2. Mechanisms of Synergy

2.1. The Abscopal Effect

The “abscopal effect”, first described in 1953 by Mole, refers to regression of metastases outside the primary radiation field post irradiation [6]. This is not frequently observed in tumours treated with radiotherapy alone; a phase II trial of 60 patients with head and neck cancer did not report any abscopal responses as a secondary endpoint [7]. However, it appears abscopal response appears to occur more commonly when ICIs is used alongside radiotherapy, both in experimental models and clinical studies. In a mouse model of melanoma, CTLA-4 blockade alongside hypofractionated radiotherapy led to an abscopal effect [8]. Radiotherapy and PD-1 blockade has seen abscopal responses in mouse models of melanoma, renal cell carcinoma and thoracic cancers [9]. This has also been reported clinically, most prominently in melanoma patients treated with PD-1 blockade [10]. A pooled analysis of two clinical trials of ICIs in non-small cell lung cancer, found that the additional of radiotherapy increased the response rate of unirradiated lesions, and was associated with prolonged survival—suggesting that this phenomenon can confer a clinical benefit [11]. The mechanism of the abscopal effect is not clear, and relies on mostly preclinical data, but it is hypothesised that radiation induced cell death can release tumour antigens from the primary lesion. These antigens may be taken up by antigen-presenting cells (APCs), which migrate to lymph nodes to prime naïve CD4+ and CD8+ T cells [12]. The activated CD8+ cytotoxic T cells then travel to both the primary irradiated tumour and the non-irradiated metastatic site where cognate tumour antigens are recognised, and this can trigger immune-mediated elimination of malignant cells.

2.2. The Cyclic GMP-AMP Synthase-Stimulator of Interferon Genes (cGAS-STING) Pathway and Interferon Production

Ionising radiation causes DNA damage directly, as well as indirectly through the formation of reactive oxygen species (ROS) that form double-stranded DNA breaks [13]. DNA released following radiation-induced cell death can activate the cytoplasmic DNA sensing cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway [14]. DNA binds to cGAS, forming 2,3-cGAMP, which then activates STING. STING acts through two main pathways: (1) upregulation of type I interferon (IFN) release by activating IRF3 and by activating NF-κB, mainly by tumour infiltrating CD141+ dendritic cells, a subtype specialised in antigen cross-presentation [15]. (2) Type I interferons mediate recruitment and the effector function of CD8+ T cells [13]. Through these mechanisms, exogenous cGAMP and STING agonists enhance the efficacy of radiation in preclinical models.
Although triggering of type I IFN production by cGAS was well characterised, it was unknown how radiotherapy-induced this response until relatively recently. Classically it was thought that cGAS recognised cytosolic DNA, but the revised model indicates that DNA recognition occurs within cGAS containing micronuclei [16]. These micronuclei form when cells progress to mitosis, following DNA damage, and this explains the delayed onset between radiotherapy and innate immune signalling [17]. The cellular compartment responsible for mediating STING’s effects is unclear. Some models indicate that tumour cell-intrinsic STING is necessary, whereas others indicate tumour cell-derived DNA in exosomes contributes to radiotherapeutic immune responses [18,19]. The role of STING in radiation-induced immunity is controversial, as other studies indicate that STING activation may be tolerogenic by modifying the microenvironment and recruitment of myeloid-derived suppressor cells (MDSCs) [20]. Therefore, more work is needed to further elucidate the biology of the STING pathway in this context.
Type I interferons themselves have opposing effects on tumour and immune cells, stimulating the anti-tumour immune response while promoting tumour cell survival. Prolonged IFN signalling can induce a chronic immunosuppressive state, leading to the selection of radioresistant tumour clones [21]. A pre-existing high expression of interferon-stimulated genes (ISGs) in tumours is linked to resistance to radiation, chemotherapy and ICIs; potentially mediated by autocrine or paracrine tumour cell IFN signalling [22]. Across cancer types, patients with tumour infiltrating dysfunctional T cells show upregulated ISGs including T cell inhibitory ligands (e.g., PD-L1) and enzymes that inactivate granzyme B [23,24]. This suggests that combining ICI and radiation may only benefit those with low ISG signatures and could be detrimental in those with chronic interferon driven basal ISG phenotype.

2.3. Immunogenic Cell Death

Radiotherapy can augment the adjuvanticity of tumours through induction of immunogenic cell death. This promotes tumour antigen processing by the release of damage-associated molecular patterns (DAMPs) and activating necrotic or apoptotic pathways [25]. These DAMPs include calreticulin, ATP and HMGB1, and all are increased by radiotherapy. Calreticulin acts as a prophagocytic signal and opposes the survival signal of CD47 [26]. HMGB1 activates TLR4 and promotes antigen cross-presentation by blocking degradation of phagosomes [27]. ATP released into the TME binds to the P2X7 purinergic receptor on antigen-presenting cells. This activates the NLRP3 inflammasome, releasing IL-1β, which is essential for priming of cytotoxic T cells [28]. Therefore, the precise delivery of radiotherapy can convert a tumour into an in-situ vaccine, whereby neoantigens are released, and DAMPs enable efficient antigen presentation and effector immune cell function (Figure 2).

2.4. Neoantigen Generation and Expression

Elimination of tumour cells by cytotoxic T cells requires antigen presentation on MHC-I molecules. An accumulating body of evidence suggests that the tumour mutational burden and neoantigen load predicts clinical response to ICIs [29]. Radiation increases tumour cell MHC-I expression [30] and radiotherapy also expands the intracellular peptide pool, altering cellular MHC-I associated peptide profiles while upregulating presentation of existing peptides [31,32]. Radiation-induced DNA damage activates a cellular stress response, promoting transcription and expression of neoantigens [33]. In Non-Small Cell Lung Cancer (NSCLC), the KPNA2 gene, a member of the nuclear transporter family, is involved in the nucleocytoplasmic transport pathway of a variety of tumour-associated proteins. Its expression is upregulated by radiation, and peptide fragments trigger activation and IFN production in the patients CD8+ T cells, therefore radiotherapy increases presentation of existing neoantigens, encouraging a CD8+ T cell response [33].
Neoantigens can be divided into clonal neoantigens, present in all tumour cells and are potent drivers of anti-tumour immunity, and subclonal neoantigens which are only present in a subset of tumours cells and are less immunogenic [29]. There is a concern that even if radiotherapy induced DNA damage elicits an effective antigen specific response it will only kill a small subset of tumour cells leaving the bulk of the tumour cells behind. Preclinical models suggest that radiotherapy in combination with ICIs is associated with increased diversity of the TCR repertoire. However, these tumours are dominated by a small number of high frequency T cells clones, suggesting an immune response is mounted against just a few clonal neoantigens [34]. Additional clinical data are needed to determine the contribution of radiation-created neoantigens in the anti-tumour response.

2.5. Remodelling of the Tumour Microenvironment

The mass of non-malignant cells and stromal tissue surrounding cancerous cells is referred to as the tumour microenvironment (TME), which is composed of numerous cell types including cancer-associated fibroblasts, endothelial cells, pericytes and a wide range of innate and adaptive immune cells [35]. Radiotherapy promotes a chemokine milieu amenable to T cell infiltration, including the secretion of CXCL16, which binds to CXCR1 on TH1 cells and activates CD8+ T cells [36]. In murine lung models, a combination of radiation and Ataxia Telangiectasia Mutated and RAD3 (ATR) inhibition promotes transcription of CXCL10, which binds to the immunostimulatory CXCR3 receptor on T cells [30]. Radiation also upregulates ICAM-1 and NKG2D ligand RAE-1γ (encoded by Rae1g) in vivo [37]. Once T cells have infiltrated a tumour, MHC-I, ICAM-1, RAE-1γ and NKG2D promote T cell arrest, tumour cell engagement and were found to be essential for the efficacy of a combination of radiotherapy and CTLA-4 blockade in mice [37]. Radiation also increases production of CCL5, which acts to recruit pro-inflammatory CCL2+CCL5+ macrophages both inside the tumour and in the peripheral circulation [38]. Low dose radiation promotes vascular renormalisation, which could be useful in immune-excluded tumours where stromal elements prevent effector immune cells from accessing the tumour parenchyma [39]. Radiation can also promote polarisation of M2-macrophages to an M1 pro-inflammatory phenotype [39]. M1-macrophages secrete TH1 cytokines (IFN-y, IL-12) and enhance CD8+ T cell activity. These M1-macrophage express high levels of inducible nitric oxide synthase (iNOS+) allowing nitric oxide dependent vessel normalisation [40].
However, these immunostimulatory aspects of radiotherapy can be counterbalanced by suppressive signalling. Regulatory T (Treg) cells are a therapeutic target for anti-tumour immunity; naturally occurring FoxP3+ Treg cells supress immunity by direct cell to cell contact and inducible Treg cells secrete TGF-β and IL-10 which promote immune escape [41]. These immunosuppressive elements are upregulated by radiotherapy, and their presence has been linked to poor response to ICIs in diverse patient cohorts [41,42]. Myeloid-derived suppressor cells (MDSCs: Gr1+CD11b+) are potent inhibitors of cytotoxic T cell function [43]. Radiotherapy can upregulate CCL2, which binds to the CCR2 receptor to promote MDSC accumulation in the TME [20]. Radiation also dampens effector T cell responses as a result of increased PD-L1 expression mediated by IFN-y [44]. Overall, this highlights a double-edged sword in the context of radiotherapy, representing an important mechanism of radioresistance while simultaneously promoting sensitivity to checkpoint blockade. As such, radiotherapy can have a dual effect on the TME, both enhancing the accumulation of effector and suppressive T cell and myeloid cell populations (Figure 3).

2.6. Immunotherapy as a Radiosensitiser

Although the current focus of combining radiation and immunotherapy is to boost response to ICIs, emerging evidence indicates that immunotherapy itself is a potent radiosensitiser. The dysfunctional vasculature within tumours promotes radioresistance as a result of the hypoxic environment [43]. The lack of oxygen is responsible for the suppression of apoptosis during radiotherapy, as low oxygen availability limits ROS generation. Paradoxically, radiation itself can lead to the disruption of in vivo vascular systems around tumours, inducing a hypoxic response and activating hypoxia-inducible factor 1 (HIF1), thus reducing the generation of intratumoural ROS [45]. Hypoxia upregulates hypoxia inducible factor 1 (HIF-1), activating anaerobic glycolysis which produces lactate and antioxidants [46]. These antioxidants scavenge ROS, impeding radiation induced cell death, and lactate promotes immunotherapy resistance [47]. Recent data indicate that immunotherapy can normalise poorly formed, leaky hypoxia-promoting vessels in the TME. In preclinical models of breast and colon cancer, anti-PD-1 and anti-CTLA-4 therapy resulted in tumour regression, increased perfusion and reduced tumour hypoxia [48]. This vessel normalisation was mediated by IFN-γ producing CD8+ T cells. The angiostatic effects of IFN-γ may be related to reduced αVβ3 integrin dependent endothelial cell activation and survival [49], and was correlated with preclinical efficacy of anti PD-1 therapy [48,49]. Another in silico analysis found an association between immune-stimulating gene pathways (including Ackr1, Il1r1, Il6st and Socs2) and vessel normalisation related genes such as decreased expression of Vegfa and increased expression of Angpt1/Angpt2 [50]. In response to ICIs, TH1 cells produce IFN-γ to normalise vessels and reduce hypoxia through increased pericyte coverage, decreased leakiness and decreased hypoxia. This suggests that ICIs remodel the tumour vasculature, augmenting their own efficacy and potentially acting as a radiosensitiser.

3. Clinical Trials of Radiation and Immunotherapy in UGI Cancer

3.1. Single Agent Immunotherapy

Single agent ICI trials in UGI cancers have delivered modest results. The ATTRACTION-2 phase III trial found that nivolumab (anti-PD-1) improved overall survival (OS; 5.3 vs. 4.1 months in the placebo group, p < 0.0001) in heavily pretreated GC or gastroesophageal junction cancer (GEJC) regardless of PD-L1 expression [51]. The KEYNOTE-059 phase II study evaluated pembrolizumab (anti-PD-1) versus chemotherapy in previously treated GC or GEJC, with the objective response rate (ORR) of 11.6%, with a longer median duration of response in PD-L1+ patients (16.3 vs. 6.9 months) [52]. Based on these results, in 2018 the FDA granted accelerated approval to pembrolizumab in the third line treatment of recurrent GC or GEJC that overexpresses PD-L1 with a Combined Positive Score [CPS] ≥ 10), as determined by a U.S. Food and Drug Administration (FDA)-approved test, with disease progression after one or more prior lines of systemic therapy as identified in KEYNOTE-181. Clinical efficacy of ICIs in OSCC is slightly more encouraging, with KEYNOTE 181, a phase III trial reporting that as second line treatment, the median OS with pembrolizumab vs. chemotherapy was similar in the intention to treat(ITT) group (7.1 vs. 7.1 months) and longer in the SCC (8.2 vs. 7.1 months) and PD-L1 CPS ≥10 groups (9.3 vs. 6.7 months) [53]. A trend was observed favouring responses in OSCC, forming the basis for the 2019 FDA approval of pembrolizumab in the second line treatment of metastatic PD-L1+ OSCC. More recently, the ATTRACTION-3 phase III trial of second line nivolumab vs. chemotherapy confirmed this OS benefit (10.9 months vs. 8.4 months, p = 0.019), further supporting the place of PD-1 inhibition in metastatic OSCC [54]. Approved indications of immunotherapy in gastroesophageal cancer are therefore confined to the second- or third-line treatment of metastatic disease. However, more recent data from the CheckMate-649 and KEYNOTE-590 studies indicate that the addition of nivolumab (median OS: 14.4 vs. 11.1, HR 0.71, p < 0.0001) or pembrolizumab (median OS: 12.4 vs. 9.8, HR 0.73, p < 0.0001) to first line chemotherapy in metastatic UGI cancers can prolong overall survival [55,56]. This suggests that combining ICIs with more traditional treatment modalities can improve outcomes in gastroesophageal cancers.
Prospective clinical data of combining immunotherapy and radiation are in a nascent phase; phase III trials have not been published outside of prostate and non small cell lung cancer (NSCLC). The phase III PACIFIC trial investigated durvalumab (anti PD-L1) following chemoradiotherapy in locally advanced NSCLC [57]. Compared to placebo, durvalumab improved overall survival (28.3 vs. 16.2 months) and both arms had similar rates of treatment related adverse events. A phase III trial in metastatic castration resistant prostate cancer found no benefit of ipilimumab (anti CTLA-4) following a single 8 Gy dose of radiation to up to five bone metastases [58]. In gastroesophageal cancers, trials are in early stages and the majority are ongoing. Clinical investigation focuses on three settings: disease treatable by surgical resection, definitive chemoradiotherapy and palliative treatment in the metastatic setting with timing of delivery and ideal combination multimodal therapies yet to be elucidated.

3.2. Locally Advanced Disease

Locally advanced, nonmetastatic UGI cancers are optimally treated by surgical resection. This is accompanied by the CROSS neoadjuvant chemoradiotherapy regimen consisting of carboplatin, paclitaxel and 41.4 Gy external beam radiotherapy (EBRT) [59]. However, only 15% of patients display a pathologic complete response (pCR) and trials are investigating if the addition of ICIs can improve response rates and patient outcomes (Table 1). One example is phase I trial (NCT03044613) of pembrolizumab (anti PD-1) in combination with the CROSS regimen in stage II/II OAC, OSCC or GEJC. Primary endpoints are pCR rate and treatment related adverse events (TRAEs). Preliminary results for the first 10 patients show an encouraging pCR rate of 40%, with acceptable toxicity and no delays in surgery [60]. A larger (n = 28) phase II trial of pembrolizumab in OSCC has reported a pCR rate of 46.1%, with 82% of patients surviving at 12 months [61]. However, 2/28 patients did not undergo surgery and two treatment related deaths were reported due to acute lung injury emphasising the need for TRAE monitoring. Another approach is the use of adjuvant ICIs in postoperative patients to better control micro-metastatic disease. A phase II trial has evaluated durvalumab (anti PD-L1) in OAC or GEJC previously treated with external beam radiotherapy (EBRT) with residual disease following tri-modality treatment of surgery and chemoradiation (NCT02639065). Early results indicate that adjuvant durvalumab is safe with few dose limiting toxicities [62]. Relapse free survival in this single arm study was 79.2%, which compares favourably to the historical rate of 50%. These encouraging results further underscore the need for more data in an adjuvant setting.

3.3. Definitive Chemoradiotherapy

Definitive chemoradiotherapy (dCRT) is an alternative standard of care in OSCC and is also employed in localised OAC deemed unsuitable for surgery. A 50.4 Gy EBRT is delivered in 25 fractions, accompanied by 5-FU and cisplatin or FOLFOX (5FU, folinic acid and oxaliplatin). dCRT may promote immunogenic changes in tumours, priming tumours for ICI treatment to further enhance local and distant tumour control. The randomised, doubled blinded, phase III KEYNOTE-975 trial is evaluating pembrolizumab with traditional dCRT in localised but inoperable OSCC, OAC and GEJC (NCT04210115). The primary endpoints are overall survival and event free survival, and results could define a new standard of care in the dCRT setting. Other ongoing trials are evaluating dual checkpoint blockade (anti PD-1 and anti CTLA-4; NCT03437200), and sequential nivolumab and cetuximab (anti EGFR) with concomitant dCRT.

3.4. Systemic Treatment of Advanced Disease

Combining ICIs and radiation in the recurrent or metastatic setting seeks to activate the abscopal response, priming antigen specific CD8+ T cells against tumours outside the radiation field. The few radio-immunotherapy trials in metastatic UGI cancers are at an early stage and seek to investigate toxicities and mechanisms of response. One example, phase II trial of pembrolizumab and 30 Gy conventional fractionated radiotherapy is recruiting patients with metastatic gastroesophageal cancers (NCT03544736). Primary endpoints aim to quantify the abscopal response; changes in CD8+ TILs at the irradiated site, and changes in MDSCs and Tregs at peripheral metastases will be measured. Another approach is a combination of nivolumab and high dose brachytherapy to deliver 16 Gy over 2 fractions (NCT02642809), and durvalumab with concomitant chemoradiotherapy in the palliative setting (NCT03544736).

4. Optimising Radiation Parameters within Immunotherapy

4.1. Radiation Dose

Conventional fractionated radiation is delivered in small 1.8–2 Gray (Gy) daily fractions. For example, the CROSS regimen for OAC and OSCC involves 41.4 Gy given in 23 fractions of 1.8 Gy each, five days per week [59]. Recent advances in radiation technique and delivery, including intensity modulated radiotherapy (IMRT), volumetric-modulated arc therapy (VMAT) and proton beam therapy allow delivery of higher radiation doses while minimising acute and long-term toxicity. This has allowed a shift to ‘hypo-fractionated’ approaches, ranging from 5–10 Gy over three to five fractions to single doses of up to 24 Gy using stereotactic body radiotherapy (SBRT). SBRT employs advanced imaging, immobilisation techniques and real-time organ motion tracking, used to ablate oligometastatic disease. Hypofractionated radiotherapy can minimise toxicity while maintaining efficacy, favoured in oesophageal cancer patients unfit to receive definitive chemoradiation therapy [60].
It is postulated that conventional fractionation can have immunosuppressive effects in the TME by recruiting MDSCs, Treg cells and M2-macrophages, potentially mediated by TGF-β upregulation [61]. However, this conventional fractionation can also have the immunogenic effect of normalising the tumour vasculature [39]. Higher radiation doses per fraction (>6 Gy) have more profound immunological effects, including facilitating maturation of APCs, increasing T cell infiltration, enhancing MHC-I expression and tumour peptide presentation and upregulation of immunostimulatory signals like Fas and ICAM on tumour cells [61,62]. However, the immunogenic effects of high dose radiation seem to have a limit; higher ablative doses (>12–18 Gy) induce TREX1, an exonuclease that degrades cytoplasmic DNA [63]. This negatively regulates the cGAS-STING pathway that is vital in radiation-induced immunogenicity. For this reason, hypofractionated radiotherapy (e.g., 3 × 8 Gy doses) has the most potential as a favourable immunomodulator [63] (Figure 4).

4.2. Radiation Timing

The immunological effects of radiation are time-dependent. In vitro there is an increase in the MHC-I peptide pool after 8 h, lasting for 11 days [31]. Clinical samples show increased activated dendritic cells during the first week, potentially correlating with radiation-induced antigen presentation [64]. Populations of activated and proliferating T cells declined in the first week but increased after the third week of therapy, supporting radiation as an in-situ vaccine [64]. These time-dependent effects can be exploited in the clinic. Multivariable analysis in the PACIFIC trial found that ICI initiation <2 weeks following chemoradiotherapy was associated with greater overall survival [57]. This survival benefit when ICI is initiated concurrently or shortly after radiation was also found in a retrospective analysis of 750 patients [65]. This suggests that radiation may ‘prime’ the tumour for optimal immunotherapy efficacy, a principle that could be employed to maximise ICI efficacy in gastroesophageal cancers.

4.3. Radiation Adverse Effects

Radiotherapy is associated with a host of adverse effects, but radiation-induced lymphopenia (RIL) is most relevant to ICI treatment. Lymphocytes are critical for the anti-tumour immune response, and T cell-deficient mice are unable to mount abscopal responses [66]. Indeed, RIL is an independent predictor of poor overall survival in UGI cancers [67]. In addition to combination chemotherapy, the radiotherapy target volume is a key determining factor in incidence of RIL. This is due to radiation doses to sites of lymphopoiesis (bone marrow) or lymphocyte storage (spleen, lymph nodes) [61]. Advances in fractionation may ameliorate this adverse effect; in two pancreatic cancer cohorts, hypofractionated radiotherapy delivered by SBRT was associated with less RIL when compared to standard fractionation (1.8 Gy), highlighting that hypofractionated approaches may also have favourable toxicity as well as efficacy [68,69]. As well as direct depletion of lymphocytes, elective irradiation of draining lymph nodes has additional detrimental effects [70]. Compared to irradiating the primary tumour alone, elective nodal irradiation is associated with altered intratumuoral chemokine expression and CD8+ T cell trafficking, as this was correlated with poorer survival in a combination of radiation and immunotherapy [70]. Immunotherapy carries a risk of immune-related adverse events (irAEs), including a potentially life-threatening pneumonitis. Radiation-induced lung injury is defined by pneumonitis and fibrosis and is a common dose limiting toxicity of radiotherapy in UGI cancers. Therefore, a combination of radiation and immunotherapy could increase incidence and severity this adverse effect [61]. The KEYNOTE-001 reported a higher rate of ICI-related pneumonitis in those that had previously received thoracic radiation, and several case reports have been published of severe pneumonitis in patients treated with ICI and SBRT [71]. In the PACIFIC trial, both the durvalumab and placebo arms had similar incidence of pneumonitis [57], so further data are needed to achieve a balance between safety and efficacy in UGI cancers.

4.4. Future Directions

Despite the success in leveraging the combination of immunotherapy for the treatment of upper gastrointestinal cancers, several issues are outstanding. There is a lack of studies that directly compare the immunogenicity of different dosing and fractionation regimens. This is seen in both preclinical studies and clinical trials, and not specific to gastrointestinal cancers. Some studies vary the entire dose of radiation but do not ascertain whether this dose would be better delivered in a single ablative dose, a hypofractionated regimen or conventional 1.8–2 Gy fractionation. Well-controlled preclinical studies would provide more clarity on the subtle effects of different fractionation, while a multi-arm clinical trial would be useful to determine the optimal dosage regimen for clinical practice. These trials should also have translational study endpoints including abscopal response and changes in CD8+ TILs, Treg cells, and MDSCs levels in the irradiated and peripheral sites. This would be a means of dissecting the mechanisms of action and resistance to immuno-radiotherapy, and provide mechanistic data specific to upper GI cancers. Finally, there is a need for more trials in advanced disease setting. In contrast to single agent ICB, most trials of ICB and radiotherapy are in locally advanced, resectable disease where chemoradiotherapy is a standard of care. However, outcomes are worst in the refractory disease setting and response rates to single agent ICB is low [72]. This highlights an unmet need for trials in this population, which could stand to benefit most of this symbiotic combination.

5. Conclusions

The advent of immune checkpoint blockade has shifted the paradigm in the treatment of solid tumours, but the impact of ICIs on patient outcomes in UGI cancers has been limited. Radiotherapy has the potential to augment responses to ICI through cGAS-STING signalling, immunogenic cell death, upregulation of neoantigen expression and through inflammatory remodelling of the immune microenvironment. Given the extensive pre-existing use of radiation and the modest activity of single agent immunotherapy, gastroesophageal cancers are poised to greatly benefit from a combination of ICIs and radiotherapy. However, questions remain surrounding methods of optimising the radiation dose and timing while minimising toxicity. Most ongoing clinical trials employ conventional radiation fractionation, although preclinical data suggest that hypofractionated regimens are favourable in terms of toxicity and efficacy. A better understanding of variability in response to immune checkpoint blockade is also required. Future trials should incorporate correlative endpoints to identify predictive biomarkers of response as this will help to select patients likely to benefit from radiation and immunotherapy and facilitate a precision oncology approach.

Author Contributions

N.E.D. was responsible for the initial and subsequent drafting of this manuscript, R.P. was responsible for initial drafting and concept design, C.H. was responsible for figure design and illustrations, M.D. was responsible for subsequent drafting, J.V.R. was responsible for review and supervision, J.L. was responsible for review, editing and overall supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the CROSS Cancer Research Charity, Ireland. Registered Charity Number 15364.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Maharaj, A.D.; Holland, J.F.; Scarborough, R.O.; Evans, S.M.; Ioannou, L.J.; Brown, W.; Croagh, D.; Pilgrim, C.; Kench, J.; Lipton, L.; et al. The Upper Gastrointestinal Cancer Registry (UGICR): A clinical quality registry to monitor and improve care in upper gastrointestinal cancers. BMJ Open 2019, 9, e031434. [Google Scholar] [CrossRef] [PubMed]
  3. Kelly, R.J. The emerging role of immunotherapy for esophageal cancer. Curr. Opin. Gastroenterol. 2019, 35, 337–343. [Google Scholar] [CrossRef] [PubMed]
  4. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.W. Cancer and radiation therapy: Current advances and future directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Smyth, E.C.; Lagergren, J.; Fitzgerald, R.C.; Lordick, F.; Shah, M.A.; Lagergren, P.; Cunningham, D. Oesophageal cancer. Nat. Rev. Dis. Primers 2017, 3, 17048. [Google Scholar] [CrossRef] [PubMed]
  6. Zemek, R.M.; Chin, W.L.; Nowak, A.K.; Millward, M.J.; Lake, R.A.; Lesterhuis, W.J. Sensitizing the Tumor Microenvironment to Immune Checkpoint Therapy. Front. Immunol. 2020, 11, 223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. McBride, S.; Sherman, E.; Tsai, C.J.; Baxi, S.; Aghalar, J.; Eng, J.; Zhi, W.I.; McFarland, D.; Young, R.; Lefkowitz, R.; et al. Randomized Phase II Trial of Nivolumab With Stereotactic Body Radiotherapy Versus Nivolumab Alone in Metastatic Head and Neck Squamous Cell Carcinoma. J. Clin. Oncol. 2020. [Google Scholar] [CrossRef]
  8. Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef] [Green Version]
  9. Park, S.S.; Dong, H.; Liu, X.; Harrington, S.M.; Krco, C.J.; Grams, M.P.; Mansfield, A.S.; Furutani, K.M.; Olivier, K.R.; Kwon, E.D. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol. Res. 2015, 3, 610–619. [Google Scholar] [CrossRef] [Green Version]
  10. Postow, M.A.; Callahan, M.K.; Barker, C.A.; Yamada, Y.; Yuan, J.; Kitano, S.; Mu, Z.; Rasalan, T.; Adamow, M.; Ritter, E.; et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012, 366, 925–931. [Google Scholar] [CrossRef] [Green Version]
  11. Welsh, J.W.; Chen, D.; Baas, P.; Chang, J.Y.; Verma, V.; Comeaux, N.; Skoulidis, F.; Lin, S.H.; Heymach, J.; Theelen, W. Radiotherapy to augment pembrolizumab responses and outcomes in metastatic non-small cell lung cancer: Pooled analysis of two randomized trials. J. Clin. Oncol. 2020, 38 (Suppl. S15), 9548. [Google Scholar] [CrossRef]
  12. Ngwa, W.; Irabor, O.C.; Schoenfeld, J.D.; Hesser, J.; Demaria, S.; Formenti, S.C. Using immunotherapy to boost the abscopal effect. Nat. Rev. Cancer 2018, 18, 313–322. [Google Scholar] [CrossRef] [PubMed]
  13. Cannan, W.J.; Pederson, D.S. Mechanisms and Consequences of Double-Strand DNA Break Formation in Chromatin. J. Cell Physiol. 2016, 231, 3–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Woo, S.R.; Fuertes, M.B.; Corrales, L.; Spranger, S.; Furdyna, M.J.; Leung, M.Y.; Duggan, R.; Wang, Y.; Barber, G.N.; Fitzgerald, K.A.; et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 2014, 41, 830–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
  16. Harding, S.M.; Benci, J.L.; Irianto, J.; Discher, D.E.; Minn, A.J.; Greenberg, R.A. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017, 548, 466–470. [Google Scholar] [CrossRef] [Green Version]
  17. Huang, R.X.; Zhou, P.K. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal. Transduct Target. Ther. 2020, 5, 60. [Google Scholar] [CrossRef]
  18. Sen, T.; Rodriguez, B.L.; Chen, L.; Corte, C.M.D.; Morikawa, N.; Fujimoto, J.; Cristea, S.; Nguyen, T.; Diao, L.; Li, L.; et al. Targeting DNA Damage Response Promotes Antitumor Immunity through STING-Mediated T-cell Activation in Small Cell Lung Cancer. Cancer Discov. 2019, 9, 646–661. [Google Scholar] [CrossRef] [Green Version]
  19. Ablasser, A.; Schmid-Burgk, J.L.; Hemmerling, I.; Horvath, G.L.; Schmidt, T.; Latz, E.; Hornung, V. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 2013, 503, 530–534. [Google Scholar] [CrossRef] [Green Version]
  20. Liang, H.; Deng, L.; Hou, Y.; Meng, X.; Huang, X.; Rao, E.; Zheng, W.; Mauceri, H.; Mack, M.; Xu, M.; et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat. Commun. 2017, 8, 1736. [Google Scholar] [CrossRef] [Green Version]
  21. Budhwani, M.; Mazzieri, R.; Dolcetti, R. Plasticity of Type I Interferon-Mediated Responses in Cancer Therapy: From Anti-tumor Immunity to Resistance. Front. Oncol. 2018, 8, 322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Benci, J.L.; Johnson, L.R.; Choa, R.; Xu, Y.; Qiu, J.; Zhou, Z.; Xu, B.; Ye, D.; Nathanson, K.L.; June, C.H.; et al. Opposing Functions of Interferon Coordinate Adaptive and Innate Immune Responses to Cancer Immune Checkpoint Blockade. Cell 2019, 178, 933–948.e14. [Google Scholar] [CrossRef] [PubMed]
  23. Jiang, P.; Gu, S.; Pan, D.; Fu, J.; Sahu, A.; Hu, X.; Li, Z.; Traugh, N.; Bu, X.; Li, B.; et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 2018, 24, 1550–1558. [Google Scholar] [CrossRef]
  24. Benci, J.L.; Xu, B.; Qiu, Y.; Wu, T.J.; Dada, H.; Twyman-Saint Victor, C.; Cucolo, L.; Lee, D.S.M.; Pauken, K.E.; Huang, A.C.; et al. Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell 2016, 167, 1540–1554.e12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Golden, E.B.; Frances, D.; Pellicciotta, I.; Demaria, S.; Helen Barcellos-Hoff, M.; Formenti, S.C. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014, 3, e28518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Chao, M.P.; Jaiswal, S.; Weissman-Tsukamoto, R.; Alizadeh, A.A.; Gentles, A.J.; Volkmer, J.; Weiskopf, K.; Willingham, S.B.; Raveh, T.; Park, C.Y.; et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci. Transl. Med. 2010, 2, 63ra94. [Google Scholar] [CrossRef] [Green Version]
  27. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4–dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef]
  28. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef]
  29. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [Green Version]
  30. Dillon, M.T.; Bergerhoff, K.F.; Pedersen, M.; Whittock, H.; Crespo-Rodriguez, E.; Patin, E.C.; Pearson, A.; Smith, H.G.; Paget, J.T.E.; Patel, R.R.; et al. ATR Inhibition Potentiates the Radiation-induced Inflammatory Tumor Microenvironment. Clin. Cancer Res. 2019, 25, 3392–3403. [Google Scholar] [CrossRef] [Green Version]
  31. Reits, E.A.; Hodge, J.W.; Herberts, C.A.; Groothuis, T.A.; Chakraborty, M.; Wansley, E.K.; Camphausen, K.; Luiten, R.M.; de Ru, A.H.; Neijssen, J.; et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006, 203, 1259–1271. [Google Scholar] [CrossRef] [PubMed]
  32. Sharma, A.; Bode, B.; Wenger, R.H.; Lehmann, K.; Sartori, A.A.; Moch, H.; Knuth, A.; von Boehmer, L.; van den Broek, M. γ-Radiation promotes immunological recognition of cancer cells through increased expression of cancer-testis antigens in vitro and in vivo. PLoS ONE 2011, 6, e28217. [Google Scholar] [CrossRef] [PubMed]
  33. Formenti, S.C.; Rudqvist, N.P.; Golden, E.; Cooper, B.; Wennerberg, E.; Lhuillier, C.; Vanpouille-Box, C.; Friedman, K.; de Andrade, L.F.; Wucherpfennig, K.W.; et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat. Med. 2018, 24, 1845–1851. [Google Scholar] [CrossRef] [PubMed]
  34. Dovedi, S.J.; Cheadle, E.J.; Popple, A.L.; Poon, E.; Morrow, M.; Stewart, R.; Yusko, E.C.; Sanders, C.M.; Vignali, M.; Emerson, R.O.; et al. Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade. Clin. Cancer Res. 2017, 23, 5514–5526. [Google Scholar] [CrossRef] [Green Version]
  35. Arneth, B. Tumor Microenvironment. Medicina 2019, 56. [Google Scholar] [CrossRef] [Green Version]
  36. Matsumura, S.; Wang, B.; Kawashima, N.; Braunstein, S.; Badura, M.; Cameron, T.O.; Babb, J.S.; Schneider, R.J.; Formenti, S.C.; Dustin, M.L.; et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008, 181, 3099–3107. [Google Scholar] [CrossRef]
  37. Ruocco, M.G.; Pilones, K.A.; Kawashima, N.; Cammer, M.; Huang, J.; Babb, J.S.; Liu, M.; Formenti, S.C.; Dustin, M.L.; Demaria, S. Suppressing T cell motility induced by anti-CTLA-4 monotherapy improves antitumor effects. J. Clin. Investig. 2012, 122, 3718–3730. [Google Scholar] [CrossRef] [Green Version]
  38. Connolly, K.A.; Belt, B.A.; Figueroa, N.M.; Murthy, A.; Patel, A.; Kim, M.; Lord, E.M.; Linehan, D.C.; Gerber, S.A. Increasing the efficacy of radiotherapy by modulating the CCR2/CCR5 chemokine axes. Oncotarget 2016, 7, 86522–86535. [Google Scholar] [CrossRef] [Green Version]
  39. Klug, F.; Prakash, H.; Huber, P.E.; Seibel, T.; Bender, N.; Halama, N.; Pfirschke, C.; Voss, R.H.; Timke, C.; Umansky, L.; et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 2013, 24, 589–602. [Google Scholar] [CrossRef] [Green Version]
  40. Sektioglu, I.M.; Carretero, R.; Bender, N.; Bogdan, C.; Garbi, N.; Umansky, V.; Umansky, L.; Urban, K.; von Knebel-Döberitz, M.; Somasundaram, V.; et al. Macrophage-derived nitric oxide initiates T-cell diapedesis and tumor rejection. Oncoimmunology 2016, 5, e1204506. [Google Scholar] [CrossRef] [PubMed]
  41. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef] [PubMed]
  42. Mondini, M.; Loyher, P.L.; Hamon, P.; Gerbe de Thore, M.; Laviron, M.; Berthelot, K.; Clémenson, C.; Salomon, B.L.; Combadière, C.; Deutsch, E.; et al. CCR2-Dependent Recruitment of Tregs and Monocytes Following Radiotherapy Is Associated with TNFalpha-Mediated Resistance. Cancer Immunol. Res. 2019, 7, 376–387. [Google Scholar] [CrossRef] [PubMed]
  43. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Deng, L.; Liang, H.; Burnette, B.; Beckett, M.; Darga, T.; Weichselbaum, R.R.; Fu, Y.-X. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J. Clin. Investig. 2014, 124, 687–695. [Google Scholar] [CrossRef] [PubMed]
  45. Carlson, D.J.; Yenice, K.M.; Orton, C.G. Tumor hypoxia is an important mechanism of radioresistance in hypofractionated radiotherapy and must be considered in the treatment planning process. Med. Phys. 2011, 38, 6347–6350. [Google Scholar] [CrossRef]
  46. Meijer, T.W.; Kaanders, J.H.; Span, P.N.; Bussink, J. Targeting hypoxia, HIF-1, and tumor glucose metabolism to improve radiotherapy efficacy. Clin. Cancer Res. 2012, 18, 5585–5594. [Google Scholar] [CrossRef] [Green Version]
  47. Scott, K.E.; Cleveland, J.L. Lactate Wreaks Havoc on Tumor-Infiltrating T and NK Cells. Cell Metab. 2016, 24, 649–650. [Google Scholar] [CrossRef] [Green Version]
  48. Zheng, X.; Fang, Z.; Liu, X.; Deng, S.; Zhou, P.; Wang, X.; Zhang, C.; Yin, R.; Hu, H.; Chen, X.; et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J. Clin. Investig. 2018, 128, 2104–2115. [Google Scholar] [CrossRef] [Green Version]
  49. Rüegg, C.; Yilmaz, A.; Bieler, G.; Bamat, J.; Chaubert, P.; Lejeune, F.J. Evidence for the involvement of endothelial cell integrin alphaVbeta3 in the disruption of the tumor vasculature induced by TNF and IFN-gamma. Nat. Med. 1998, 4, 408–414. [Google Scholar] [CrossRef]
  50. Tian, L.; Goldstein, A.; Wang, H.; Ching Lo, H.; Sun Kim, I.; Welte, T.; Sheng, K.; Dobrolecki, L.E.; Zhang, X.; Putluri, N.; et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 2017, 544, 250–254. [Google Scholar] [CrossRef]
  51. Kang, Y.-K.; Boku, N.; Satoh, T.; Ryu, M.-H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.; Muro, K.; Kang, W.K.; et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538–12, ATTRACTION-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 2461–2471. [Google Scholar] [CrossRef]
  52. Fuchs, C.S.; Doi, T.; Jang, R.W.; Muro, K.; Satoh, T.; Machado, M.; Sun, W.; Jalal, S.I.; Shah, M.A.; Metges, J.-P..; et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients With Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 2018, 4, e180013. [Google Scholar] [CrossRef] [PubMed]
  53. Kojima, T.; Muro, K.; Francois, E.; Hsu, C.-H.; Moriwaki, T.; Kim, S.-B.; Lee, S.-H.; Bennouna, J.; Kato, K.; Lin, S.; et al. Pembrolizumab versus chemotherapy as second-line therapy for advanced esophageal cancer: Phase III KEYNOTE-181 study. J. Clin. Oncol. 2019, 37 (Suppl. S4), 2. [Google Scholar] [CrossRef]
  54. Kato, K.; Cho, B.C.; Takahashi, M.; Okada, M.; Lin, C.-Y.; Chin, K.; Kadowaki, S.; Ahn, M.; Hamamoto, Y.; Doki, Y.; et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 1506–1517. [Google Scholar] [CrossRef]
  55. Kato, K.; Sun, J.M.; Shah, M.A.; Enzinger, P.C.; Adenis, A.; Kojima, T.; Metges, J.P.; Li, Z.; Kim, S.B.; Cho, B.C.; et al. LBA8_PR Pembrolizumab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced esophageal cancer: The phase 3 KEYNOTE-590 study. Ann. Oncol. 2020, 31, S1192–S1193. [Google Scholar] [CrossRef]
  56. Moehler, M.; Shitara, K.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Bragagnoli, A.C.; Liu, T.; et al. LBA6_PR Nivolumab (nivo) plus chemotherapy (chemo) versus chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer (GC/GEJC)/esophageal adenocarcinoma (EAC): First results of the CheckMate 649 study. Ann. Oncol. 2020, 31, S1191. [Google Scholar] [CrossRef]
  57. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Overall Survival with Durvalumab after Chemoradiotherapy in Stage III NSCLC. N. Engl. J. Med. 2018, 379, 2342–2350. [Google Scholar] [CrossRef]
  58. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; van den Eertwegh, A.J.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H.; 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, 15, 700–712. [Google Scholar] [CrossRef] [Green Version]
  59. Weichselbaum, R.R.; Liang, H.; Deng, L.; Fu, Y.X. Radiotherapy and immunotherapy: A beneficial liaison? Nat. Rev. Clin. Oncol. 2017, 14, 365–379. [Google Scholar] [CrossRef]
  60. Jones, C.M.; Spencer, K.; Hitchen, C.; Pelly, T.; Wood, B.; Hatfield, P.; Crellin, A.; Sebag-Montefiore, D.; Goody, R.; Crosby, T.; et al. Hypofractionated Radiotherapy in Oesophageal Cancer for Patients Unfit for Systemic Therapy: A Retrospective Single-Centre Analysis. Clin. Oncol. 2019, 31, 356–364. [Google Scholar] [CrossRef]
  61. Spaas, M.; Lievens, Y. Is the Combination of Immunotherapy and Radiotherapy in Non-small Cell Lung Cancer a Feasible and Effective Approach? Front. Med. 2019, 6, 244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Chakraborty, M.; Abrams, S.I.; Camphausen, K.; Liu, K.; Scott, T.; Coleman, C.N.; Hodge, J.W. Irradiation of Tumor Cells Up-Regulates Fas and Enhances CTL Lytic Activity and CTL Adoptive Immunotherapy. J. Immunol. 2003, 170, 6338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef] [PubMed]
  64. Dorta-Estremera, S.; Colbert, L.E.; Nookala, S.S.; Yanamandra, A.V.; Yang, G.; Delgado, A.; Mikkelson, M.; Eifel, P.; Jhingran, A.; Lilie, L.L.; et al. Kinetics of Intratumoral Immune Cell Activation During Chemoradiation for Cervical Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 593–600. [Google Scholar] [CrossRef]
  65. Samstein, R.; Rimner, A.; Barker, C.A.; Yamada, Y. Combined Immune Checkpoint Blockade and Radiation Therapy: Timing and Dose Fractionation Associated with Greatest Survival Duration Among Over 750 Treated Patients. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, S129–S130. [Google Scholar] [CrossRef]
  66. Demaria, S.; Ng, B.; Devitt, M.L.; Babb, J.S.; Kawashima, N.; Liebes, L.; Formenti, S.C. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 862–870. [Google Scholar] [CrossRef]
  67. Venkatesulu, B.P.; Mallick, S.; Lin, S.H.; Krishnan, S. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit. Rev. Oncol. Hematol. 2018, 123, 42–51. [Google Scholar] [CrossRef]
  68. Crocenzi, T.; Cottam, B.; Newell, P.; Wolf, R.F.; Hansen, P.D.; Hammill, C.; Solhjem, M.C.; To, Y.; Greathouse, A.; Tormoen, G.; et al. A hypofractionated radiation regimen avoids the lymphopenia associated with neoadjuvant chemoradiation therapy of borderline resectable and locally advanced pancreatic adenocarcinoma. J. Immunother. Cancer 2016, 4, 45. [Google Scholar] [CrossRef] [Green Version]
  69. Wild, A.T.; Herman, J.M.; Dholakia, A.S.; Moningi, S.; Lu, Y.; Rosati, L.M.; Hacker-Prietz, A.; Assadi, R.K.; Saeed, A.M.; Pawlik, T.M.; et al. Lymphocyte-Sparing Effect of Stereotactic Body Radiation Therapy in Patients with Unresectable Pancreatic Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 571–579. [Google Scholar] [CrossRef] [Green Version]
  70. Marciscano, A.E.; Ghasemzadeh, A.; Nirschl, T.R.; Theodros, D.; Kochel, C.M.; Francica, B.J.; Muroyama, Y.; Anders, R.A.; Sharabi, A.B.; Velarde, E.; et al. Elective Nodal Irradiation Attenuates the Combinatorial Efficacy of Stereotactic Radiation Therapy and Immunotherapy. Clin. Cancer Res. 2018, 24, 5058–5071. [Google Scholar] [CrossRef] [Green Version]
  71. Louvel, G.; Bahleda, R.; Ammari, S.; Le Péchoux, C.; Levy, A.; Massard, C.; Le Pavec, J.; Champiat, S.; Deutsch, E. Immunotherapy and pulmonary toxicities: Can concomitant immune-checkpoint inhibitors with radiotherapy increase the risk of radiation pneumonitis? Eur. Respir. J. 2018, 51, 1701737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Power, R.; Lowery, M.A.; Reynolds, J.V.; Dunne, M.R. The Cancer-Immune Set Point in Oesophageal Cancer. Front. Oncol. 2020, 10, 891. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Combination strategies of ICIs in UGI cancers. Immune checkpoint inhibitors (ICIs) are under investigation in a variety of settings in upper gastrointestinal (UGI) cancers, including in conjunction with surgery, chemotherapy, radiation therapy, and multimodal combinations.
Figure 1. Combination strategies of ICIs in UGI cancers. Immune checkpoint inhibitors (ICIs) are under investigation in a variety of settings in upper gastrointestinal (UGI) cancers, including in conjunction with surgery, chemotherapy, radiation therapy, and multimodal combinations.
Ijms 22 01071 g001
Figure 2. Immunogenic effects of radiation therapy. Radiation can augment anti-tumour immunity in several ways. Damage and death of cancer cells leads to release of tumour neoantigens and damage-associated molecular patterns (DAMPs) such as calreticulin, high motility group box 1 (HMGB1) protein, and ATP which activate dendritic cells (DCs) to prime and stimulate CD8+ T cells responsible for cancer cell detection and elimination. Moreover, radiation can increase the expression and alter the array of peptides presented on major histocompatibility (MHC) class I proteins, which CD8+ T cells use to identify transformed cells. Radiation-induced DNA damage can activate the cGAS-STING pathway leading to type I interferon production which enhances CD8+ T cell activity. Beyond local effects, radiation may induce systemic anti-tumour immunity if tumour-specific CD8+ T cells migrate to metastatic lesions; regression of distant tumour cells outside the radiation field is known as the abscopal effect.
Figure 2. Immunogenic effects of radiation therapy. Radiation can augment anti-tumour immunity in several ways. Damage and death of cancer cells leads to release of tumour neoantigens and damage-associated molecular patterns (DAMPs) such as calreticulin, high motility group box 1 (HMGB1) protein, and ATP which activate dendritic cells (DCs) to prime and stimulate CD8+ T cells responsible for cancer cell detection and elimination. Moreover, radiation can increase the expression and alter the array of peptides presented on major histocompatibility (MHC) class I proteins, which CD8+ T cells use to identify transformed cells. Radiation-induced DNA damage can activate the cGAS-STING pathway leading to type I interferon production which enhances CD8+ T cell activity. Beyond local effects, radiation may induce systemic anti-tumour immunity if tumour-specific CD8+ T cells migrate to metastatic lesions; regression of distant tumour cells outside the radiation field is known as the abscopal effect.
Ijms 22 01071 g002
Figure 3. Radiation therapy can shape the immune response just as immunotherapy can shape the response to radiation therapy. Radiation induces production of a variety of chemokines that can facilitate and antagonise the anti-tumour response: T cells and M1 macrophages contribute to tumour elimination and can be recruited by CXCL16 or CXL10, and CCL5, respectively; myeloid-derived suppressor cells (MDSCs) are immunosuppressive and can be recruited by radiation-induced expression of CCL12. Immune checkpoint inhibitors (ICIs) boost anti-tumour immunity by disrupting suppressive signalling molecules such as PD-1, PD-L1 and CTLA-4 (latter not shown). By recruiting T cells (via aforementioned mechanisms) radiation therapy can augment this phenomenon. Furthermore, cytokines released by immune cells including IFN-γ from CD8+ T cells can promote tumour vessel normalisation counteracting hypoxia which promotes radioresistance: thus, by enhancing T cell responses ICIs can act as radiosensitisers.
Figure 3. Radiation therapy can shape the immune response just as immunotherapy can shape the response to radiation therapy. Radiation induces production of a variety of chemokines that can facilitate and antagonise the anti-tumour response: T cells and M1 macrophages contribute to tumour elimination and can be recruited by CXCL16 or CXL10, and CCL5, respectively; myeloid-derived suppressor cells (MDSCs) are immunosuppressive and can be recruited by radiation-induced expression of CCL12. Immune checkpoint inhibitors (ICIs) boost anti-tumour immunity by disrupting suppressive signalling molecules such as PD-1, PD-L1 and CTLA-4 (latter not shown). By recruiting T cells (via aforementioned mechanisms) radiation therapy can augment this phenomenon. Furthermore, cytokines released by immune cells including IFN-γ from CD8+ T cells can promote tumour vessel normalisation counteracting hypoxia which promotes radioresistance: thus, by enhancing T cell responses ICIs can act as radiosensitisers.
Ijms 22 01071 g003
Figure 4. Optimising radiation dose and delivery for optimal immunogenicity. Small doses of radiation delivered in conventional fractionated radiotherapy are believed to have immunosuppressive effects in the tumour microenvironment; accumulation of immunosuppressive cell types such as myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and M2 macrophages repress anti-tumour immunity. This immunosuppressive effect is somewhat counterbalanced by the normalising effect low dose radiation has on the tumour vasculature. Higher doses of radiation, such as those used in hypofractionation, can have a stimulatory immunogenic effect mediated by increased antigen presenting cell (APC) maturation, in addition to augmented T cell infiltration and enhanced expression of immunogenic proteins including MHC class I, ICAM and FAS on tumour cells. However, once the dose of radiation surpasses 12–18 Gy, immunogenicity is compromised: TREX1, an exonuclease, is induced leading to degradation of cytoplasmic DNA, thus negatively regulating the cGAS-STING pathway.
Figure 4. Optimising radiation dose and delivery for optimal immunogenicity. Small doses of radiation delivered in conventional fractionated radiotherapy are believed to have immunosuppressive effects in the tumour microenvironment; accumulation of immunosuppressive cell types such as myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and M2 macrophages repress anti-tumour immunity. This immunosuppressive effect is somewhat counterbalanced by the normalising effect low dose radiation has on the tumour vasculature. Higher doses of radiation, such as those used in hypofractionation, can have a stimulatory immunogenic effect mediated by increased antigen presenting cell (APC) maturation, in addition to augmented T cell infiltration and enhanced expression of immunogenic proteins including MHC class I, ICAM and FAS on tumour cells. However, once the dose of radiation surpasses 12–18 Gy, immunogenicity is compromised: TREX1, an exonuclease, is induced leading to degradation of cytoplasmic DNA, thus negatively regulating the cGAS-STING pathway.
Ijms 22 01071 g004
Table 1. Ongoing clinical Trials of radiation and immunotherapy in operable disease.
Table 1. Ongoing clinical Trials of radiation and immunotherapy in operable disease.
IdentifierPhaseNDisease SettingTreatmentRadiationPrimary Endpoint
NCT03792347I20Stage II/III OSCCPembrolizumab, carboplatin, and paclitaxel41.4 Gy in 23 fractionsTRAEs
NCT02844075II18Stage II/III OSCCPembrolizumab, carboplatin, and paclitaxel41.4 Gy in 23 fractionspCR rate
NCT03257163II40Stage II/III dMMR or EBV+ GCNeoadjuvant pembrolizumabConventional Fractionation RFS
Adjuvant Capecitabine and pembrolizumab
NCT03064490II38Stage II/III GC or OACPembrolizumab, carboplatin, and paclitaxel41.4 Gy in 23 fractionspCR rate
NCT02730546I/II68Stage II/III GC or GEJCPembrolizumab, carboplatin, and paclitaxel41.4 Gy in 23 fractionspCR rate
PFS
NCT03044613I25Stage II/III OAC, OSCC or GEJCNivolumab and carboplatin and paclitaxel41.4 Gy in 23 fractionsTRAEs
NCT03776487I/II30Stage II/III GC or GEJCFOLFOX and Nivolumab and ipilimumab followed by surgical resection50 Gy 25 fractionsTRAEs
NCT02962063II35Stage II/III GEJC and GCNeoadjuvant Durvalumab and mFOLFOX
Adjuvant durvalumab
50 Gy in 28 fractionsTRAEs
pCR rate
NCT04159974II56Stage II/III OAC or GEJCDurvalumab, carboplatin and paclitaxel41.4 Gy in 23 fractionspCR rate
TRAEs
NCT02639065II23Stage II/III OAC or GEJC with residual disease Durvalumab41.4 Gy in 23 fractionsTRAEs
DLTs
NCT03490292I/II24Stage II/III OSCC or OACAvelumab and Carboplatin, paclitaxel41.4 Gy in 23 fractionsDLT
pCR
Abbreviations: TRAEs, treatment related adverse effects; pCR, pathological complete response; DLT, dose limiting toxicity; OAC, oesophageal adenocarcinoma; OSCC, oesophageal squamous cell carcinoma; GEJC, gastroesophageal junction adenocarcinoma; dMMR, deficient mismatch repair; Gy, Gray; RFS, relapse free survival.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Donlon, N.E.; Power, R.; Hayes, C.; Davern, M.; Reynolds, J.V.; Lysaght, J. Radiation and Immunotherapy in Upper Gastrointestinal Cancers: The Current State of Play. Int. J. Mol. Sci. 2021, 22, 1071. https://doi.org/10.3390/ijms22031071

AMA Style

Donlon NE, Power R, Hayes C, Davern M, Reynolds JV, Lysaght J. Radiation and Immunotherapy in Upper Gastrointestinal Cancers: The Current State of Play. International Journal of Molecular Sciences. 2021; 22(3):1071. https://doi.org/10.3390/ijms22031071

Chicago/Turabian Style

Donlon, Noel E., Robert Power, Conall Hayes, Maria Davern, John V. Reynolds, and Joanne Lysaght. 2021. "Radiation and Immunotherapy in Upper Gastrointestinal Cancers: The Current State of Play" International Journal of Molecular Sciences 22, no. 3: 1071. https://doi.org/10.3390/ijms22031071

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop