2412

S Gasser et al. Tumour microenvironment 24:12 T283–T295 Thematic Review and therapeutic implications

The role of the tumour microenvironment in immunotherapy

Stephan Gasser1,2, Lina H K Lim3 and Florence S G Cheung3

1Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Roche Glycart AG, Schlieren, Switzerland Correspondence 2 Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, NUS Immunology should be addressed Programme, Centre for Life Sciences, National University of Singapore, Singapore to S Gasser or F S G Cheung 3 Department of Physiology, Yong Loo Lin School of Medicine, NUS Immunology Programme, Centre for Email Life Sciences, National University of Singapore, Singapore [email protected] or [email protected]

Abstract

Recent success in immunomodulating strategies in lung and has Key Words prompted much enthusiasm in their potential to treat other advanced solid . ff However, their applications have shown variable success and are even ineffective ff thyroid cancer against some tumours. The efficiency of immunotherapies relies on an immunogenic ff immunotherapy tumour microenvironment. The current field of has focused on ff tumour microenvironment understanding the interaction of cancer and host immune cells to break the state of ff endometrial cancer and explain how molecular patterns of cytokines and affect tumour progression. Here, we review our current knowledge of how inherent properties of tumours and their different tumour microenvironments affect therapeutic

Endocrine-Related Cancer Endocrine-Related outcome. We also discuss insights into recent multimodal therapeutic approaches that Endocrine-Related Cancer target tumour immune evasion and suppression to restore anti-tumour immunity. (2017) 24, T283–T295

Introduction

Recent advances in immunotherapy have presented prognostic significance of immune signatures that may opportunities to eliminate that were not achieved guide immunotherapies for endocrine-related cancers. with previous standard of care therapies (Delitto et al. 2016). Immune checkpoint inhibitor treatment has shown to be a tolerable therapy delivering very promising results Factors that correlate with responses to in treating many cancer types including and cancer immunotherapies lung cancer (Delitto et al. 2016, Rafei et al. 2017). However, Tumour histology recurrence of cancers and variable success among different cancers are not uncommon. Even in responsive The immune profile of a patient is influenced by cancers, the immune checkpoint inhibitor success rate intrinsic factors including genetic composition of the is often below 50% of patients (Chen & Mellman 2017). tumour and the expression of proinflammatory factors These inefficiencies and variability emphasise our lack of (Chen & Mellman 2017). In addition, extrinsic factors understanding of the immunologic events involved in such as prior or concurrent infections, age, microbiota the tumour microenvironment. In this review, we will or other environmental factors are likely important summarise our knowledge of the immune responses in determinates in the response to immunotherapies endocrine cancers, with new insights in predictive and (Chen & Mellman 2017).

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology This paper forms part of a special section on Immunotherapy and Cancer. DOI: 10.1530/ERC-17-0146 Published by Bioscientifica Ltd. The guest editors for this section were Joanne Y Y Ngeow and Laura S Ward. Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access

10.1530/ERC-17-0146 Thematic Review S Gasser et al. Tumour microenvironment 24:12 T284 and therapeutic implications

Recent clinical findings using checkpoint inhibitors tumours and many immune excluded tumours recruit suggest that the degree of infiltration in the tumour MDSCs and secrete TGF-β, which creates a reactive is an important predictor of response of patients to immunosuppressive stroma that lacks an inflammatory (Teng et al. 2015). Furthermore, milieu required for T cell infiltration and activation the phenotype of T cells and their localisation within (Hegde et al. 2016). In these types of cancers, checkpoint the tumour microenvironment are likely to impact the inhibitor immunotherapy responses are uncommon results (Teng et al. 2015, Chen & Mellman 2017). In an (Tothill et al. 2008, Calon et al. 2015, Ryner et al. attempt to categorise cancers according to their immune 2015). Non-inflamed tumours are also characterised by phenotype, three broad types of cancer were suggested cytokines and cell types that are associated with immune to exist: Inflamed tumours, immune excluded and suppression and tolerance, such as regulatory T cells, immunologically ignorant, also referred to as immune MDSCs and M2 (Chen & Mellman 2017). deserts (Hegde et al. 2016). Inflamed tumours with pre- M1 and M2 macrophages promote Th1 and Th2 responses, existing immunity are characterised by the presence of respectively, where M1 macrophages initially fight to tumour infiltrating lymphocytes (TILs) with high density sterilise a wound in the case of an infection followed by of functional IFN-γ-secreting CD8+ T cells and CD4+ T cells. M2 macrophages, which secrete growth factors to promote These T cells often express inhibitory receptors such as growth, proliferation, and the induction PD-1 and CTLA-4 and other markers indicative of an of anti-inflammatory regulatory T cells (Cao et al. 2010, exhausted phenotype. The inflamed phenotype correlates Mills 2012). Cancers with insignificant levels of specific in some cancers with genetic instability and occurrence are analogous to sterile wounds. The presence of of tumour-specific mutations in peptides that can be M2 macrophages promotes tumour growth while tumour- recognised by T cells, so-called neoantigens (Schumacher secreted factors such as TGF-β and prostaglandins inhibit & Schreiber 2015, Hegde et al. 2016). Inflamed tumours the presence of M1 macrophages and maintain wound are also characterised by the increased expression of healing M2 macrophages at tumour sites, shielding the proinflammatory cytokines including interleukin-1β tumour from T cells (Mills 2012). Other non-inflamed (IL-1), IL-2, IL-12, IL-23 and TNF-α (Chen & Mellman tumours show minimal infiltration of immune cells or 2017). Current checkpoint inhibitor therapies act by expression of immune-related genes possibly because they reinforcement of pre-existing T cell anti-tumour responses are non-immunogenic or represent early immune escape and are therefore most effective in inflamed tumours. In mutants. Tumours with immune desert phenotype are Endocrine-Related Cancer Endocrine-Related the presence of an active T cell tumour response, IFN-γ rare responders to checkpoint inhibitor therapies. produced by T cells and other immune cells upregulate Tumours with higher mutation and neoantigen the expression of PD-L1, which counters T cell receptor burden correlate with durable clinical benefit to and CD28-mediated activation signals of T cells (Hui et al. checkpoint inhibitor therapies. In addition, neoantigen 2017, Kamphorst et al. 2017). In clinical studies of PD-1 signatures for predicting therapy responses are also or PD-L1 antagonists, high T cell density and the adaptive beginning to emerge (Snyder et al. 2014, Rizvi et al. 2015, increase of PD-L1 expression on tumour and immune Van Allen et al. 2015). In agreement with these findings, cells are often used as features of an active anti-tumour patients with DNA repair pathway deficiencies, another T cell response (Chen & Mellman 2017). Other predictive determinant of mutational burden, were also associated biomarkers that correlate with active cellular immunity with better response (Le et al. 2015, Rizvi et al. 2015). include the expression of granzymes in T cells and chemokines such as C-X-C motif ligand 9 (CXCL9) and Role of tumour mutational load in cancer immunity CXCL10 (Chen et al. 2012). Although a response to PD-L1 inhibitor therapy is most common in inflamed tumours, A number of recent publications found correlations it is not assured, suggesting that immune cell infiltration between the mutational load uniquely present in cancer is necessary but may not be sufficient for an effective cells (neoantigens) and T cell infiltration of tumours anti-cancer response (Chen & Mellman 2017). Myeloid- (Brown et al. 2015, Li et al. 2016, Senbabaoglu et al. derived suppressor cells (MDSCs) are premature myeloid 2016). High-throughput epitope screening of cancer progenitor cells produced that suppress T cell function. mutanomes revealed that only a small fraction of The expansion of MDSCs is further promoted by tumour- mutated peptides in cancer cells produced T cell reactivity derived cytokines (Delitto et al. 2016). Some inflamed (Lu et al. 2014, Yadav et al. 2014, Linnemann et al. 2015,

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T285 and therapeutic implications

Schumacher & Schreiber 2015). The parameters that are derived from gene products that are expendable define immunogenic peptides are not well understood, for tumor progression, they are likely subject to loss by but may include MHC affinity and hydrophobicity in key downregulation, mutation or gene deletion (Lu et al. residues (Chowell et al. 2015, Chen & Mellman 2017). 2014). Examining the intra- (ITH) A number of overexpressed in various cancers in lung cancer and melanoma showed that tumours with that are lowly expressed in normal cells have also been both a high clonal (present in all tumour cells) neoantigen targeted for immunotherapeutic vaccines. These included burden and low neoantigen ITH had significantly longer antigenic peptides from genes such as renal tumour progression-free survival after checkpoint inhibitor antigen 1 (RAGE-1) for renal , preferentially therapy (McGranahan et al. 2016). Conversely, tumours expressed antigen in melanoma (PRAME) for melanoma with high subclonal (present in a subset) neoantigen and other cancers, erb-b2 2 fraction were poor responders (McGranahan et al. (ERBB2) for epithelial cancers and Wilms’ tumour 1 2016). The enrichment of subclonal neoantigens may (WT-1) for leukaemia (Vigneron 2015). Vaccines using be due to the immune shaping of the tumour during its translation-optimised E7 protein of HPV16 expressed in natural development or may be induced by cytotoxic cervical cancer induce significant E7-specific cytotoxic T chemotherapy exposure (McGranahan et al. 2016). It cell response in vivo (Liu et al. 2002). Most neoantigens might be necessary therefore to target multiple tumour arising from passenger mutations are not related to the antigens in cancer immunotherapies at the same time to tumourigenesis process and are therefore likely to be target subclonal neoantigens and to prevent outgrowth unique to every patient. The development of algorithms of less immunogenic tumour variants (Mittal et al. 2014). to predict candidate neoantigens and immunogenicity The use of peptides vaccines and CAR-T cell of peptides with higher precision are the topic of intense technology for the treatment of endocrine cancers might current research efforts in academia and the private sector be challenging as several independent whole-exome (Weber et al. 2009, Zhang et al. 2015). Such algorithms sequencing projects of pancreatic cancers (PC) tend to will be invaluable for the design of peptide vaccines show fewer mutations than lung cancer and melanoma, and chimeric antigen receptor (CAR) T cell technology, with one base pair per megabase for PC compared to where chimeras are generated by combining side-chain approximately 10 base pairs per megabase for lung variable fragments with the ζ subunit of CD3, CD28 cancer and melanoma (Alexandrov et al. 2013). Thus, the or CD137 to artificially link tumour surface antigen lower rate of somatic mutations may contribute to the Endocrine-Related Cancer Endocrine-Related specificity with activation signal transmission in T cells poor immunogenicity of PCs and render these cancers upon antigen binding (Delitto et al. 2016). Vaccines have resistant to checkpoint immunotherapy (Delitto et al. had poor track record, as they largely focus on antigens 2016). Recent genomic profiling found that although not specific to tumours (non-neoantigens). It is possible the predicted number of neoantigens in pancreatic that peripheral tolerance contributed to the low efficacy cancer samples is low, nearly all pancreatic ductal of non-neoantigen-based vaccines. The recent use of (PDAC) yielded potential targetable with melanoma-associated antigen neoantigen candidates (Bailey et al. 2016). It is important (MAGE-A)-specific CAR-T cells resulted in significant to note that the identification of neoantigens may change clinical responses in some patients but has also been the potential for immunotherapy in pancreatic cancers, accompanied by fatal on-target/off tumour adverse but must be accompanied by strategies to overcome events (Zajac et al. 2017). The shift of focus on targeting the immunosuppressive tumour microenvironment to neoantigens may circumvent issues of tolerance and tissue unmask these neoantigen-specific responses Bailey( et al. specificity to improve efficacy; however, the challenge 2016, Yarchoan et al. 2017). of heterogeneous antigen expression in tumours and difficulties in conducting randomised trials remain due to T cell exhaustion and response to cancer immunotherapy the personalised nature of neoantigens (Zajac et al. 2017). Similarly, most CAR-T cells have focused on antigens that Many TILs present in the tumour microenvironment are not tumour specific (CD19/CD20) Johnson( & June display an exhausted phenotype (Chen & Mellman 2017). CD19 is expressed on the majority of malignant B 2017). These cells are characterised by the expression cells, but is also expressed by normal B cells and follicular of immune checkpoint inhibitors such as PD-1, TIM3 dendritic cells (Wang et al. 2012). As most neoantigens and LAG3 (Chen & Mellman 2017). Recent evidence

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T286 and therapeutic implications

has shown that the response of PD-1/PD-L1-based Activation of stimulator of IFN genes (STING) immunotherapy depends on the presence and expansion can induce anti-tumour effects via type I interferons of T cells with a less exhausted phenotype (Im et al. 2016). in various cancers including PCs in animal models Encouragingly, such T cell populations might be enriched (Gasser et al. 2016). Administration of STING agonists in patients via different means. Inhibition of MEK and resulted in marked increase of CD8+ T cell infiltration PD-1 can impede the rate of T cell exhaustion, or in the and upregulation of PD-L1 (Fu et al. 2015). Consequently, case of PD-L1 inhibitors, reverse the exhaustion of T cells the combined use of STING agonists and PD-1 blockade expressing intermediate levels of PD-1, but not T cells enhanced anti-tumour immunity (Demaria et al. 2015) expressing high levels of PD-1 (Chen & Mellman 2017). and led to regression of tumours that were resistant to The enrichment of non-exhausted TILs in endometrial PD-1 therapy (Fu et al. 2015). cancers may hold great promise for these and other poorly PCs were found to frequently exhibit an immune immunogenic cancers. The combined use of MEK and infiltrate exclusion profile characterised by elevated levels PD-1/L1 inhibitors has yielded synergistic and sustained of TGF-β (Delitto et al. 2016). Moreover, it has recently pancreatic tumour regression in vivo (Ebert et al. 2016). been shown that the exclusion of T cells from PCs was However, combined blockage of PD-L1 and CTLA4 leads attributed to local immunosupression mediated by the to a more favourable treatment outcome when compared production of CXCL12 by activation protein to MEK inhibitors. A possible reason is that inhibition (FAP) expressing carcinoma-associated . of CTLA4 enhances priming and proliferation of naïve Depletion of FAP+ stromal cells restored the effects of T cells, whereas MEK inhibitors block T cell priming PD-L1 and CTLA4 inhibitors. Blocking of the CXCL12 (Buchbinder & Desai 2016). Hence, these data support receptor not only restored T cell infiltration, but also acted the conclusion that the continuous replenishment of synergistically with PD-L1 inhibition in the clearance TILs by naïve T cell priming is necessary to efficiently of tumour cells (Feig et al. 2013). Production of heme reject tumours (Chen & Mellman 2017). It is tempting oxygenase-1 by FAP+CD45+ M2 macrophages can further to speculate that careful sequential use of CTLA4 and drive immunosuppression in PCs (Arnold et al. 2014). MEK inhibitors may further increase the response rate The immune excluded phenotype of PCs is partially in endometrial cancers by allowing efficient priming of regulated by macrophages residing outside of the tumour tumour-specific T cells and preventing the exhaustion microenvironment. Depletion of these macrophages of such T cells. The recent discovery that epigenetic restored T cell immunotherapy efficacy (Beatty et al. 2015). Endocrine-Related Cancer Endocrine-Related changes contribute to the exhaustion of T cells may Other major contributors of the immunosuppressive yield new combination partners for PD-1/L1 inhibitors tumour microenvironment in PCs include suppressive (Chang et al. 2014). myeloid cells and regulatory T cells (Tregs) (Morse et al. 2009, Delitto et al. 2016). The administration of the attenuated Toxoplasma gondii vaccine strain CPS in Pancreatic cancer microenvironment Panc02 cancer models activated proinflammatory The median overall survival of advanced pancreatic cancer myeloid cell populations and increased the expression of patients have remained poor under current standard of care costimulatory molecules, the secretion of IL-12, and the therapies with tumours frequently developing resistance recruitment of T cells into the tumour microenvironment (Lim et al. 2015, Irigoyen et al. 2017). Unfortunately, (Sanders et al. 2015). A modified ISOCOM vaccine was immune checkpoint inhibitors have shown little effects shown to be ineffective for advanced cancers due to Treg- in pancreatic cancer patients (Brahmer et al. 2012, mediated immune suppression (Nicholaou et al. 2009). The Delitto et al. 2016). However, combination of high-dose response to ISOCOM tumour vaccines in an orthotopic radiotherapy with anti-PD-L1 significantly improved model of PC was improved by the use of low-dose CpG tumour responses in pancreatic ductal adenocarcinoma TLR9 agonist as adjuvant, which resulted in tumour (PDAC) cell allografts (Azad et al. 2017). Radiotherapy regression by breaking Treg-mediated immunosupression induced a large population of tumour cells susceptible (Jacobs et al. 2011). Such findings are yet to be validated to cytotoxic killing, and the early administration of for patients with advanced cancers in clinical studies. anti-PD-L1 prevented the growth of immunosuppressive T cells were also shown to induce dormancy or pro­ cells and increased recruitment and activation of T cells mote multistage carcinogenesis depending on cytokine (Azad et al. 2017). signals present in the tumour microenvironment.

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T287 and therapeutic implications

In a Simian virus 40, large T antigen-induced pancreatic (Ward 2014). Similar to papillary thyroid cancer (PTC), tumour mouse model, IFN-γ-producing CD4+ T cells follicular epithelial dysplasia lesions in chronic lymphocytic arrested tumour growth by preventing the activation thyroiditis (CLT) showed strong diffuse staining for HBME-

of αvβ3 , tumour- and multistage 1, cytokeratin 19, galectin-3 and cyclin-D1 compared to carcinogenesis via IFN-γ and TNFR1 signalling. However, the normal thyroid (Chui et al. 2013). Functional TLR3 in the absence of combined IFN-γ and TNFR1 signalling, is overexpressed in both Hashimoto’s thyroiditis (HT) the same tumour-protective CD4+ T cells strongly and PTC, which induces the production of immune cell enhanced angiogenesis and multistage carcinogenesis attracting cytokines and chemokines. In accordance, cell (Muller-Hermelink et al. 2008). This permanent infiltration in PTC resembles the immune cell infiltrates

growth arrest in G1/G0 required the activation of in HT (Harii et al. 2005, McCall et al. 2007). Although p16INK4a, STAT1, IFN-γ and TNFR1 signalling, which the molecular mechanisms that contribute to thyroid was essential for sustained Rb hypophosphorylation autoimmunity and cancer remain to be clarified, these (Braumuller et al. 2013). observations suggest a link between and Although IFN-γ generally exhibits anti-cancer activity, thyroid cancer. it can also induce the expression of PD-L1 and CD80 Inflammation in thyroid cancer depends on the in PCs. The binding of these two ligands to inhibitory exposure to radiation and iodine intake, and more receptors correlated with higher tumour infiltration importantly, the presence of concurrent chronic of Tregs, poorer anti-cancer responses and shortened inflammatory conditions and the balance of pro-tumour postoperative survival (Loos et al. 2008). and anti-tumour activity elicited by immune cells that MDSCs are important mediators of immune infiltrate the thyroid cancer microenvironment Ward( suppressive tumour microenvironments (Gabrilovich & 2014). Although many patients respond well to current Nagaraj 2009). In PDAC patients, the accumulation of therapy, 10–30% of thyroid cancer patients eventually -like CD13high MDSCs was found to correlate develop recurrent disease and tumour metastasis (Ward with shorter survival (Zhang et al. 2017). The suppression 2014). Early-stage metastasis is a characteristic of anti-cancer T cell responses by CD13high MDSCs was of thyroid cancer. These clinically undetectable micro- mediated via expression of arginase-1 (Zhang et al. 2017), metastasis to cervical lymph nodes are present in 45% of which depletes l-arginine leading to downregulation of PTC patients at diagnosis and are difficult to distinguish the TCR chain required for TCR signalling and T cell from inflammatory lymph nodes, hindering the curability Endocrine-Related Cancer Endocrine-Related ζ growth arrest (Munder 2009). MDSC expansion was found of the disease (Li et al. 2014). TLR3 is also implicated in to be further driven by tumour-derived cytokines such as PTC recurrence and metastasis, which provides further VEGF, G-CSF, GM-CSF, IFN-γ, TGF-β, IL-1β, IL-6, IL-10 and evidence for an inflammatory component influencing CCL12 in PCs (Delitto et al. 2016). disease progression (McCall et al. 2007, Li et al. 2014). -based combination therapies remain Ligands responsible for activating TLR3 have not been the first line of treatment for PCs (Lim et al. 2015, Irigoyen elucidated, but possibly include nucleotides from viruses et al. 2017). Interestingly, gemcitabine doses that have no or immunological cell death. direct effects on cell growth or cytotoxicity can induce The compromisation of the innate γδ T cell-mediated lysis of PC cells by upregulating the in PTC is suggested by the decrease in cytotoxic NK cell expression of ligands for the activating immune receptor infiltration with disease progression from thyroid nodular NKG2D (Miyashita et al. 2017). In contrast, high doses of goitre to PTC (Gogali et al. 2012). Different subsets of NK gemcitabine stimulate phosphorylation of Erk1/2, which cells can be found in the tumour microenvironment of reduces the expression of NKG2D ligands (Miyashita et al. PTC (Poli et al. 2009). Cytotoxic function of NK cells is 2017). Hence, low-dose gemcitabine may be useful in enriched among CD56dim cells, while CD56bright NK cells context of cancer immunotherapy of PC patients. predominately have immune regulatory functions. High expression level of CD16 on CD56dim NK cells also makes them efficient mediators of antibody-dependent cellular Thyroid cancer microenvironment cytotoxicity (Poli et al. 2009). The cytotoxic CD16+CD56dim In contrast to PC, thyroid cancers are highly inflammatory NK cell population predominates in healthy thyroid tumours (Ward 2014). Multiple similarities exist between tissue, thyroid nodular goitre and PTC. CD56bright NK thyroid cancers and thyroid autoimmunity, which may cells predominately infiltrate early-stage PTC tissue, but represent a risk factor for thyroid cancer development this population is subsequently replaced by the CD56dim

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T288 and therapeutic implications

cytotoxic subpopulation of NK cells in advanced stages of NCI-H295R ACC cells to LPS, but also reduced basal and PTC (Gogali et al. 2013). These findings suggest a specific growth factor-induced ACC cell viability (Kanczkowski et al. enrichment of CD56bright NK cells at earlier stages of PTC 2010). Hence, treatment of ACC patients with intratumoural development. However, the mechanisms that mediate the TLR4 agonist may create a more inflammatory tumour enrichment or the activity of either NK cell subset in PTC microenvironment that better responds to checkpoint will require further research in the future (Gogali et al. inhibitors or other cancer immunotherapies. 2013). Tumour-associated macrophages (TAMs) in poorly Pituitary carcinoma differentiated thyroid carcinoma are associated with capsular invasion and poor survival (Ryder et al. 2008). It has In pituitary adenomas, constitutively produced recently been shown that CXCL16/CXCR signalling drives intratumoural IL-6 contributes to the excessive hormone polarisation in the PTC microenvironment, secretion in endocrine-active tumours via autocrine and resulting in cells with a M-2-like phenotype characterised paracrine stimulation of the IL-6 receptor (Thiele et al. by colony stimulating factor-1 receptor (CSF-1R) and C-C 2003). This results in tumour expansion by concomitantly motif ligand and receptor 2 (CCL2/CCR2) stimulating tumour cell proliferation and tumour overexpression, which enhanced the PTC cell migration neovascularisation (Renner et al. 2009). In nude mice potential and reinforced angiogenesis (Ryder et al. 2013). studies, it was found that cytokines binding to the shared In anaplastic thyroid cancer, expression of CD163, a gp130 subunit of the IL-6 receptor contributed to tumour marker for M2 macrophages, correlated with CXCR4 progression (Graciarena et al. 2004, Renner et al. 2009). expression and more aggressive clinical characteristics The authors have not identified which cytokines were (Kim et al. 2016). The potentially CXCR4-mediated responsible for the effect. If future studies identify IL-6 as recruitment of macrophages could be countered by an important contributing factor in these mice, this model CXCR4 inhibitors or by ablation of macrophages from may serve as a good model for human pituitary adenomas. the tumour microenvironment via CSF-1R depleting It is not clear what signals trigger IL-6 production in human antibodies (Ries et al. 2014, Escamilla et al. 2015). Such pituitary adenomas, but an in vitro study showed that TLR4 compounds are currently in early clinical development by agonists can induce IL-6 expression in human pituitary several companies. adenoma cell lines (Tichomirowa et al. 2005). However, LPS also directly suppressed cell growth of the TLR4-positive Endocrine-Related Cancer Endocrine-Related pituitary carcinoma cell line AtT20 (Tichomirowa et al. Other endocrine tumours 2005). It is unclear whether TLR signals are triggered in pituitary adenoma and which effect of TLR4 agonists Adrenalcortical tumour would dominate in the tumour. Moreover, not all pituitary Adrenalcortical carcinoma (ACC) is a rare adenomas express TLR4 and only a subset of cells expresses with variable but generally dismal prognosis TLR4 in TLR4-positive tumours (Tichomirowa et al. 2005). (Bilimoria et al. 2008). Mitotane is the only FDA-approved A better understanding of these cellular processes may cytotoxic treatment, which has minimal effects on 5-year allow the targeting of gp130-dependent pathways for the survival rates (Aufforth & Nilubol 2014). Immunotherapy treatment of pituitary . approaches have not been successful due to the secretion of immunosuppressive glucocorticoids, the lack of known Current therapeutic approaches in ACC-tumour-specific antigens and insufficient knowledge endocrine cancer of the ACC immunosuppressive microenvironment (Papewalis et al. 2006). In addition, adrenal masses are Chemotherapy treatment in the past was generally often clinically unapparent and with few reliable markers, believed to be immunosuppressive (Delitto et al. 2016). histological differentiation of benign and malignant However, the combination of certain cytotoxic therapies lesions remains difficult Mansmann( et al. 2004). In the with immunotherapy may assist in synergistic anti- normal adrenal, TLR4 expression was detected throughout tumour immunity (Zitvogel et al. 2011). Cell death the adrenal cortex, whereas the TLR4 coreceptor CD14 induced by chemotherapy and radiation may provide expression was found only in the inner cortical zones. proinflammatory ‘danger’ signals in the otherwise No staining was observed in epithelial cells of ACCs. tolerogenic milieu of the tumour microenvironment The restoration of TLR4 and CD14 not only sensitised (Delitto et al. 2016). The subsequent immunologic response

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T289 and therapeutic implications

will depend on the type of cell death, the magnitude of with other cytotoxic agents such as capecitabine or cellular stress and the pattern of regulatory cytokines at erlotinib (Lim et al. 2015, Irigoyen et al. 2017). Gemcitabine play (Lake & Robinson 2005). Release of tumour antigens was found to have positive effects on the ability of by dying cells and damaged tissue may also boost existing intratumoural DCs to present antigens to tumour-specific tumour-specific immune responses or potentially induce T cells (McDonnell et al. 2015). Importantly, gemcitabine novel T cell responses against cancer. More importantly, primed the host for a strong anti-tumour response after current chemotherapy and immunotherapy combinations a second insult with a virus expressing the same antigen have started to harness the immunomodulation effects (Nowak et al. 2003). Together, these findings supported of chemotherapy (Lake & Robinson 2005), resulting in the rationale behind the combination of gemcitabine improved antigen presentation and cytotoxic activity with immunotherapy, especially when administered in the immunosuppressing tumour microenvironment. before a tumour vaccine or CD40 agonists (Table 1). A search in http://www.clinicaltrials.gov for clinical CD40 agonists are powerful activators of APCs and trials of endocrine cancers using immunomodulatory promote T cell and other cytotoxic immune responses agents has yielded a list of mostly on-going, clinical (Vonderheide & Glennie 2013). trials (Table 1). A partial list of pancreatic cancer A number of other clinical trials aim to enhance immunotherapies has been selected for discussion here, the anticancer by providing as additional pancreatic cancer-specific clinical studies proinflammatory cues such as TLR agonists, TNF or IL-12. have been extensively reviewed recently by Delitto and These agents primarily act on innate immune cells, which coworkers (Delitto et al. 2016). are important for the development of adaptive T cell responses against cancers.

Immune checkpoint inhibitors Vaccines CTLA4 and PD-1 are two inhibitory receptors on T cells that counter priming or activation of T cells (Buchbinder Another approach to enhance cancer-specific T cells & Desai 2016). After activation of naïve T cells, CTLA4 responses in endocrine cancers is the use of cancer is expressed on the cell surface and competes with the vaccines. Cancer vaccines consist of peptide or protein costimulatory receptor CD28 for binding of its B7 ligands antigens present in tumour cells and adjuvants that on antigen-presenting cells (APCs) and tumour cells stimulate APCs. Protein antigens are generally less prone Endocrine-Related Cancer Endocrine-Related (Sansom 2000). PD-1 expression levels increase with to degradation and require a weaker stimulant to be continuous activation of T cells. Its ligands are expressed immunogenic (Azmi et al. 2014). A number of different on APCs, immune cells, tumour-associated stromal cells peptide/protein and adjuvant combinations are currently and tumour cells. Recent reports have shown that PD-1 explored in clinical trials for endocrine cancers. suppresses intracellular signalling pathways initiated by CD28, but PD-1 may also counter T cell receptor Telomerase peptide vaccines Telomeres caps signals (Hui et al. 2017). PD-1/PD-L1 antagonists were shorten as cells divide, which eventually drives cells into found to restore the effector functions of T cells in the senescence or cell death (Muraki et al. 2012). Cancer cells periphery that have been turned off following extended frequently activate the expression of telomerase reverse antigen exposure, and are characterized by an exhausted transcriptase (hTERT) to maintain telomere ends of DNA phenotype described earlier in this review. Monoclonal (Maida & Masutomi 2015). The ability of hTERT peptides antibody-based antagonists against PD-1 (Nivolumab and to induce cancer cell-specific cytotoxic T cell activity was Pembrolizumab), PD-L1 (MEDI4726 and Atezolizumab) explored in the phase III TeloVac study. The MHC class and CTLA4 ( and tremelimumab) have II telomerase vaccine GV1001 was given with a GM-CSF already shown success in the treatment of other cancers, adjuvant and gemcitabine/capecitabine chemotherapy particularly in advanced melanoma and lung cancer (Table 1). The sequential arm of this study was based (Lipson et al. 2015, Postow et al. 2015). Due to the limited on the idea that a brief course of chemotherapy before effectiveness of monotherapies in endocrine cancers as vaccination may stimulate an endogenous anti-cancer discussed earlier, combination strategies are currently immune response, which would be further boosted by explored for the treatment of endocrine cancers (Table 1). the vaccine. However, no difference in survival was Therapies based on gemcitabine remain the first line observed in the chemotherapy group compared to the of treatment for pancreatic cancer, often in combination sequential chemotherapy/vaccine group (Middleton et al.

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T290 and therapeutic implications

Table 1 Immunotherapies under evaluation in clinical trials for endocrine cancers.

Immune therapy agent Phase; status Mechanism Endocrine cancers treated Other agents NCT number Modulators of innate and adaptive immunity Prembrolizumab II; recruiting Anti-PD1 Thyroid, adrenocortical NCT02721732 Prembrolizumab II; recruiting Anti-PD1 Thyroid, NCT02628067 neuroendocrine Prembrolizumab II; not yet recruiting Anti-PD1 Thyroid, NCT03012620 neuroendocrine Prembrolizumab I; recruiting Anti-PD1 Thyroid, pancreatic Enoblituzumab NCT02475213 (MGA271) Prembrolizumab II; not yet recruiting Anti-PD1 Thyroid Lenvatinib NCT02973997 Prembrolizumab II; not yet recruiting Anti-PD1 Thyroid NCT03072160 Prembrolizumab II; recruiting Anti-PD1 Thyroid NCT02688608 Prembrolizumab II; recruiting Anti-PD1 Adrenocortical NCT02673333 Prembrolizumab I/II; not yet recruiting Anti-PD1 Gastroenteropancreatic Somatuline Depot NCT03043664 Neuroendocrine Prembrolizumab II; recruiting Anti-PD1 Neuroendocrine NCT02939651 MEDI4736 II; not yet recruiting Anti-PDL1 Neuroendocrine Tremelimumab NCT03095274 (anti-CTLA4) MEDI4736 I; recruiting Anti-PDL1 Pancreatic Tremelimumab NCT02639026 (anti-CTLA4), radiation Ipilimumab I; recruiting Anti-CTLA4 Thyroid, pancreatic Enoblituzumab NCT02381314 (MGA271) Nivolumab II; recruiting Anti-PD1 Adrenocortical NCT02720484 Nivolumab II; recruiting Anti-PD1 Pancreatic Ipilimumab (anti- NCT02866383 CTLA4), radiation Nivolumab II; recruiting Anti-PD1 Adrenal cortex, adrenal NCT02834013 Ipilimumab Anti-CTLA4 gland pheochromocytoma, pancreatic, pancreatic neuroendocrine, parathyroid gland, thyroid, pituitary gland Endocrine-Related Cancer Endocrine-Related Nivolumab II; not yet recruiting Anti-PD1 Neuroendocrine NCT02923934 Ipilimumab Anti-CTLA4 Ibrutinib II; recruiting Btk inhibitor Pancreatic, carcinoid NCT02575300 RO7009789 I; recruiting CD40 agonist Pancreatic Nab-, NCT02588443 gemcitabine VTX-2337 I; active, not recruiting TLR8 agonist Pancreatic Cyclophosphamide, NCT02650635 pegfilgrastim CYT-6091 I, completed Recombinant TNF Adrenocortical, NCT00436410 pancreatic Recombinant I, completed Recombinant Thyroid, pancreatic, ABI-007, carboplatin, NCT00004074 interleukin-12 interleukin-12 parathyroid, trastuzumab adenocortical MGD009 I, recruiting Antibody for Thyroid, pancreatic NCT02628535 B7-H3 and CD3 Vaccines GV1001 III; completed Telomerase Pancreatic GM-CSF, gemcitabine, NCT00425360 peptide vaccine capecitabine GV1001 III; recruiting Telomerase Pancreatic Gemcitabine, NCT02854072 peptide vaccine capecitabine TG01 Mutant Ras GM-CSF, gemcitabine NCT02261714 peptide vaccine CEA RNA-pulsed DC I, completed DC cell based Pancreatic NCT00004604 cancer vaccine vaccine

(Continued)

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T291 and therapeutic implications

Table 1 Continued.

Immune therapy agent Phase; status Mechanism Endocrine cancers treated Other agents NCT number Ras peptide cancer II, completed Mutant RAS Aldesleukin (IL-2), NCT00019331 vaccine peptide vaccine sargramostim (GM-CSF), detoxPC TRICOM-CEA(6D) I, completed DC cells loaded Pancreatic NCT00027534 with TRICOM- CEA(6D) antigen GI-6207 II, recruiting Recombinant Thyroid NCT01856920 yeast-based vaccine express mutant full length human CEA PSMA/PRAME I, completed DNA vector Panreatic, thyroid, NCT00423254 (MKC1106-PP) pPRA-PSM with neuroendocrine synthetic peptides E-PRA and E-PSM Recombinant I, completed Recombinant Pancreatic, thyroid With or without NCT00028496 fowlpox-CEA(6D)/ fowlpox virus Sargramostim TRICOM vaccine vector encoding (GM-CSF) CEA/TRICOM

2014). Several reasons may have a contributed to the angiogenesis through integrin regulation (Conway et al. failure of this study including the enrolment of late- 2006). The MKC1106-PP vaccine is directly administered stage patients, limited efficacy of chemotherapy due to into lymph nodes, which may allow for more efficient stromal characteristics of pancreatic cancer, effects of the priming of T cells by directly targeting APCs present in chemotherapy on the immune response and choice of the lymph nodes (Smith et al. 2011). A phase I trial was peptide or adjuvant (Emens & Middleton 2015). able to induce PRAME- and PSMA-specific T cells in the blood in 15 out of 24 patients (Weber et al. 2011). In 7 Carcinoembryonic antigen vaccines The TRICOM- patients, the disease was controlled for at least 6 months Endocrine-Related Cancer Endocrine-Related CEA(6D) vaccine encodes the carcinoembryonic antigen (Weber et al. 2011). (CEA) and a TRIad of COstimulatory Molecules (B7-1, ICAM-1 and LFA-3) (TRICOM) (Palena et al. 2004). The vaccine is either administered via a recombinant fowlpox Conclusion and future perspectives virus vector or directly loaded onto DCs to enhance CEA The recent success of immunotherapy for cancer has presentation on APCs. CEA-TRICOM vaccines are generally generated enormous enthusiasm with much effort safe and can generate significant CEA-specific immune focusing on checkpoint inhibitors and cancer vaccines. responses (Marshall et al. 2005). When administered with Although some patients have benefited from favourable IFN-α, CEA-TRICOM can led to significantly increased responses, immunotherapies have proved ineffective overall survival of patients compared to vaccine alone for many cancer patients including endocrine cancer (Marshall et al. 2005, Duggan et al. 2016). patients. A better understanding of how to manipulate the tumour microenvironment to elicit an effective Melanoma and prostate peptide vaccines The cancer-directed immune response will be essential. Many MKC1106-PP vaccine consists of a DNA vector, pPRA-PSM, challenges remain, such as how to select and deliver encoding segments of two tumor antigens, PRAME tumor antigens and what immune suppression or evasion and PSMA (Weber et al. 2011). Both PRAME and PSMA mechanism to target in different cancer patients. Only are expressed in a range of tumours (Silver et al. 1997, a fraction of candidate neoantigens selected by current Conway et al. 2006, Lerut et al. 2015). The former facilitates neoantigen prediction algorithms trigger an anti-tumour tumour progression by inhibiting cellular differentiation response (Mardis 2017). A more detailed understanding through repression of retinoic acid receptor signalling in of antigen processing, trafficking and MHC loading cancer cells (Oehler et al. 2009), and the latter is expressed will be necessary for improving prediction algorithms in the neovasculature of solid tumours mediating (Mardis 2017). The newly established tumour neoantigen

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T292 and therapeutic implications

selection alliance is an effort to facilitate the sharing Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake of sequencing and neoantigen evaluation data among CG, Camacho LH, Kauh J, Odunsi K, et al. 2012 Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. universities, non-profit institutions and companies, in New England Journal of Medicine 366 2455–2465. (doi:10.1056/ the hope of generating better neoantigen predictions. NEJMoa1200694) Improved algorithm accuracy should increase the Braumuller H, Wieder T, Brenner E, Assmann S, Hahn M, Alkhaled M, Schilbach K, Essmann F, Kneilling M, Griessinger C, et al. 2013 likelihood to find immunogenic neoantigens in every T-helper-1-cell cytokines drive cancer into senescence. Nature 494 cancer patient. Mechanisms of immunosuppression and 361–365. (doi:10.1038/nature11824) evasion proposed from pre-clinical studies have yet to be Brown SD, Raeburn LA & Holt RA 2015 Profiling tissue-resident T cell repertoires by RNA sequencing. Genome Medicine 7 125. tested in patients, with results pending for many clinical (doi:10.1186/s13073-015-0248-x) trials. The outcomes of these studies will shed light on Buchbinder EI & Desai A 2016 CTLA-4 and PD-1 pathways: similarities, which therapies should be combined for the treatment of differences, and implications of their inhibition. American Journal of Clinical Oncology 39 98–106. (doi:10.1097/COC.0000000000000239) endocrine cancers and optimisation of novel personalised Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando- cancer immunotherapies. Momblona X, Iglesias M, Sevillano M, Palomo-Ponce S, Tauriello DV, Byrom D, et al. 2015 Stromal gene expression defines poor-prognosis subtypes in . Nature Genetics 47 320–329. Declaration of interest (doi:10.1038/ng.3225) The authors declare that there is no conflict of interest that could be Cao Q, Wang Y, Zheng D, Sun Y, Lee VW, Zheng G, Tan TK, Ince J, perceived as prejudicing the impartiality of this review. Alexander SI & Harris DC 2010 IL-10/TGF-beta-modified macrophages induce regulatory T cells and protect against adriamycin nephrosis. Journal of the American Society of Nephrology 21 933–942. (doi:10.1681/ASN.2009060592) Funding Chang JT, Wherry EJ & Goldrath AW 2014 Molecular regulation of This work did not receive any specific grant from any funding agency in effector and memory T cell differentiation. Nature Immunology 15 the public, commercial or not-for-profit sector. 1104–1115. (doi:10.1038/ni.3031) Chen DS, Irving BA & Hodi FS 2012 Molecular pathways: next- generation immunotherapy – inhibiting programmed death-ligand 1 References and programmed death-1. Clinical Cancer Research 18 6580–6587. (doi:10.1158/1078-0432.CCR-12-1362) Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin Chen DS & Mellman I 2017 Elements of cancer immunity and the AV, Bignell GR, Bolli N, Borg A, Borresen-Dale AL, et al. 2013 cancer-immune set point. Nature 541 321–330. (doi:10.1038/ Signatures of mutational processes in human cancer. Nature 500 nature21349) 415–421. (doi:10.1038/nature12477) Chowell D, Krishna S, Becker PD, Cocita C, Shu J, Tan X, Greenberg PD, Arnold JN, Magiera L, Kraman M & Fearon DT 2014 Tumoral immune Klavinskis LS, Blattman JN & Anderson KS 2015 TCR contact residue Endocrine-Related Cancer Endocrine-Related suppression by macrophages expressing fibroblast activation protein- hydrophobicity is a hallmark of immunogenic CD8+ T cell epitopes. alpha and heme oxygenase-1. Cancer Immunology Research 2 PNAS 112 E1754–E1762. (doi:10.1073/pnas.1500973112) 121–126. (doi:10.1158/2326-6066.CIR-13-0150) Chui MH, Cassol CA, Asa SL & Mete O 2013 Follicular epithelial Aufforth RD & Nilubol N 2014 Emerging therapy for adrenocortical dysplasia of the thyroid: morphological and immunohistochemical carcinoma. International Journal of Endocrine Oncology 1 173–182. characterization of a putative preneoplastic lesion to papillary (doi:10.2217/ije.14.13) thyroid carcinoma in chronic lymphocytic thyroiditis. Virchows Azad A, Yin Lim S, D’Costa Z, Jones K, Diana A, Sansom OJ, Kruger P, Archiv 462 557–563. (doi:10.1007/s00428-013-1397-1) Liu S, McKenna WG, Dushek O, et al. 2017 PD-L1 blockade Conway RE, Petrovic N, Li Z, Heston W, Wu D & Shapiro LH 2006 enhances response of pancreatic ductal adenocarcinoma to Prostate-specific membrane antigen regulates angiogenesis by radiotherapy. EMBO Molecular Medicine 9 167–180. (doi:10.15252/ modulating integrin signal transduction. Molecular and Cellular emmm.201606674) Biology 26 5310–5324. (doi:10.1128/MCB.00084-06) Azmi F, Ahmad Fuaad AA, Skwarczynski M & Toth I 2014 Recent progress Delitto D, Wallet SM & Hughes SJ 2016 Targeting tumor tolerance: a in adjuvant discovery for peptide-based subunit vaccines. Human new hope for pancreatic cancer therapy? Pharmacology and Vaccines and Immunotherapeutics 10 778–796. (doi:10.4161/hv.27332) Therapeutics 166 9–29. (doi:10.1016/j.pharmthera.2016.06.008) Bailey P, Chang DK, Forget MA, Lucas FA, Alvarez HA, Haymaker C, Demaria O, De Gassart A, Coso S, Gestermann N, Di Domizio J, Flatz L, Chattopadhyay C, Kim SH, Ekmekcioglu S, Grimm EA, et al. 2016 Gaide O, Michielin O, Hwu P, Petrova TV, et al. 2015 STING Exploiting the neoantigen landscape for immunotherapy of activation of tumor endothelial cells initiates spontaneous and pancreatic ductal adenocarcinoma. Scientific Reports 6 35848. therapeutic antitumor immunity. PNAS 112 15408–15413. (doi:10.1038/srep35848) (doi:10.1073/pnas.1512832112) Beatty GL, Winograd R, Evans RA, Long KB, Luque SL, Lee JW, Duggan MC, Jochems C, Donahue RN, Richards J, Karpa V, Foust E, Paul Clendenin C, Gladney WL, Knoblock DM, Guirnalda PD, et al. 2015 B, Brooks T, Tridandapani S, Olencki T, et al. 2016 A phase I study of Exclusion of T cells from pancreatic carcinomas in mice is regulated recombinant (r) vaccinia-CEA(6D)-TRICOM and rFowlpox-CEA(6D)- by Ly6C(low) F4/80(+) extratumoral macrophages. Gastroenterology TRICOM vaccines with GM-CSF and IFN-alpha-2b in patients with 149 201–210. (doi:10.1053/j.gastro.2015.04.010) CEA-expressing carcinomas. Cancer Immunology, Immunotherapy 65 Bilimoria KY, Shen WT, Elaraj D, Bentrem DJ, Winchester DJ, Kebebew E 1353–1364. (doi:10.1007/s00262-016-1893-7) & Sturgeon C 2008 Adrenocortical carcinoma in the United States: Ebert PJ, Cheung J, Yang Y, McNamara E, Hong R, Moskalenko M, treatment utilization and prognostic factors. Cancer 113 3130–3136. Gould SE, Maecker H, Irving BA, Kim JM, et al. 2016 MAP kinase (doi:10.1002/cncr.23886) inhibition promotes T cell and anti-tumor activity in combination

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T293 and therapeutic implications

with PD-L1 checkpoint blockade. Immunity 44 609–621. Group. European Journal of Cancer 75 73–82. (doi:10.1016/j. (doi:10.1016/j.immuni.2016.01.024) ejca.2016.12.032) Emens LA & Middleton G 2015 The interplay of immunotherapy and Jacobs C, Duewell P, Heckelsmiller K, Wei J, Bauernfeind F, Ellermeier J, chemotherapy: harnessing potential synergies. Cancer Immunology Kisser U, Bauer CA, Dauer M, Eigler A, et al. 2011 An ISCOM vaccine Research 3 436–443. (doi:10.1158/2326-6066.CIR-15-0064) combined with a TLR9 agonist breaks immune evasion mediated by Escamilla J, Schokrpur S, Liu C, Priceman SJ, Moughon D, Jiang Z, regulatory T cells in an orthotopic model of pancreatic carcinoma. Pouliot F, Magyar C, Sung JL, Xu J, et al. 2015 CSF1 receptor International Journal of Cancer 128 897–907. (doi:10.1002/ijc.25399) targeting in prostate cancer reverses macrophage-mediated resistance Johnson LA & June CH 2017 Driving gene-engineered T cell to androgen blockade therapy. Cancer Research 75 950–962. immunotherapy of cancer. Cell Research 27 38–58. (doi:10.1038/ (doi:10.1158/0008-5472.CAN-14-0992) cr.2016.154) Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, Connell Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, CM, Roberts EW, Zhao Q, Caballero OL, et al. 2013 Targeting Konieczny BT, Daugherty CZ, Koenig L, Yu K, et al. 2017 Rescue of CXCL12 from FAP-expressing carcinoma-associated fibroblasts exhausted CD8 T cells by PD-1-targeted therapies is CD28- synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. dependent. Science 355 1423–1427. (doi:10.1126/science.aaf0683) PNAS 110 20212–20217. (doi:10.1073/pnas.1320318110) Kanczkowski W, Tymoszuk P, Ehrhart-Bornstein M, Wirth MP, Zacharowski Fu J, Kanne DB, Leong M, Glickman LH, McWhirter SM, Lemmens E, K & Bornstein SR 2010 Abrogation of TLR4 and CD14 expression and Mechette K, Leong JJ, Lauer P, Liu W, et al. 2015 STING agonist signaling in human adrenocortical tumors. Journal of Clinical formulated cancer vaccines can cure established tumors resistant to Endocrinology and Metabolism 95 E421–E429. (doi:10.1210/jc.2010-1100) PD-1 blockade. Science Translational Medicine 7 283ra252. Kim DI, Kim E, Kim YA, Cho SW, Lim JA & Park YJ 2016 Macrophage (doi:10.1126/scitranslmed.aaa4306) densities correlated with CXC chemokine receptor 4 expression and Gabrilovich DI & Nagaraj S 2009 Myeloid-derived suppressor cells as related with poor survival in anaplastic thyroid cancer. Endocrinology regulators of the immune system. Nature Reviews Immunology 9 and Metabolism 31 469–475. (doi:10.3803/EnM.2016.31.3.469) 162–174. (doi:10.1038/nri2506) Lake RA & Robinson BW 2005 Immunotherapy and chemotherapy – a Gasser S, Zhang WY, Tan NY, Tripathi S, Suter MA, Chew ZH, Khatoo M, practical partnership. Nature Reviews Cancer 5 397–405. (doi:10.1038/ Ngeow J & Cheung FS 2016 Sensing of dangerous DNA. Mechanisms nrc1613) of Ageing and Development 165 33–46 (doi:10.1016/j. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora mad.2016.09.001) AD, Luber BS, Azad NS, Laheru D, et al. 2015 PD-1 blockade in Gogali F, Paterakis G, Rassidakis GZ, Kaltsas G, Liakou CI, Gousis P, tumors with mismatch-repair deficiency. New England Journal of Neonakis E, Manoussakis MN & Liapi C 2012 Phenotypical analysis Medicine 372 2509–2520. (doi:10.1056/NEJMoa1500596) of lymphocytes with suppressive and regulatory properties (Tregs) Lerut E, Van Poppel H, Joniau S, Gruselle O, Coche T & Therasse P 2015 and NK cells in the papillary carcinoma of thyroid. Journal of Rates of MAGE-A3 and PRAME expressing tumors in FFPE tissue Clinical Endocrinology and Metabolism 97 1474–1482. (doi:10.1210/ specimens from bladder cancer patients: potential targets for jc.2011-1838) antigen-specific cancer immunotherapeutics. International Journal of Gogali F, Paterakis G, Rassidakis GZ, Liakou CI & Liapi C 2013 CD3(−) Clinical and Experimental Pathology 8 9522–9532. (doi:10.1016/j. CD16(−)CD56(bright) immunoregulatory NK cells are increased in lungcan.2016.09.006) the tumor microenvironment and inversely correlate with advanced Li B, Li T, Pignon JC, Wang B, Wang J, Shukla SA, Dou R, Chen Q, Hodi stages in patients with papillary thyroid cancer. Thyroid 23 FS, Choueiri TK, et al. 2016 Landscape of tumor-infiltrating T cell Endocrine-Related Cancer Endocrine-Related 1561–1568. (doi:10.1089/thy.2012.0560) repertoire of human cancers. Nature Genetics 48 725–732. Graciarena M, Carbia-Nagashima A, Onofri C, Perez-Castro C, Giacomini (doi:10.1038/ng.3581) D, Renner U, Stalla GK & Arzt E 2004 Involvement of the gp130 Li D, Gu R, Yang X, Hu C, Li Y, Gao M & Yu Y 2014 TLR3 correlated cytokine transducer in MtT/S pituitary somatotroph tumour with cervical lymph node metastasis in patients with papillary development in an autocrine-paracrine model. European Journal of thyroid cancer. International Journal of Clinical and Experimental Endocrinology 151 595–604. (doi:10.1530/eje.0.1510595) Medicine 7 5111–5117. Harii N, Lewis CJ, Vasko V, McCall K, Benavides-Peralta U, Sun X, Ringel Lim JY, Cho JH, Lee SJ, Lee DK, Yoon DS & Cho JY 2015 Gemcitabine MD, Saji M, Giuliani C, Napolitano G, et al. 2005 Thyrocytes express combined with capecitabine compared to gemcitabine with or a functional toll-like receptor 3: overexpression can be induced by without erlotinib as first-line chemotherapy in patients with viral infection and reversed by phenylmethimazole and is associated advanced pancreatic cancer. Cancer Research and Treatment 47 with Hashimoto's autoimmune thyroiditis. Molecular Endocrinology 266–273. (doi:10.4143/crt.2013.158) 19 1231–1250. (doi:10.1210/me.2004-0100) Linnemann C, van Buuren MM, Bies L, Verdegaal EM, Schotte R, Calis Hegde PS, Karanikas V & Evers S 2016 The where, the when, and the JJ, Behjati S, Velds A, Hilkmann H, Atmioui DE, et al. 2015 High- how of immune monitoring for cancer immunotherapies in the era throughput epitope discovery reveals frequent recognition of neo- of checkpoint inhibition. Clinical Cancer Research 22 1865–1874. antigens by CD4+ T cells in human melanoma. Nature Medicine 21 (doi:10.1158/1078-0432.ccr-15-1507) 81–85. (doi:10.1038/nm.3773) Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Lipson EJ, Forde PM, Hammers HJ, Emens LA, Taube JM & Topalian SL Huang J, Kim JM, Mellman I, et al. 2017 T cell costimulatory 2015 Antagonists of PD-1 and PD-L1 in cancer treatment. Seminars receptor CD28 is a primary target for PD-1-mediated inhibition. in Oncology 42 587–600. (doi:10.1053/j.seminoncol.2015.05.013) Science 355 1428–1433. (doi:10.1126/science.aaf1292) Liu W, Gao F, Zhao KN, Zhao W, Fernando GJ, Thomas R & Frazer IH Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, 2002 Codon modified human papillomavirus type 16 E7 DNA Hale JS, Nasti TH, Sharpe AH, et al. 2016 Defining CD8+ T cells that vaccine enhances cytotoxic T-lymphocyte induction and anti- provide the proliferative burst after PD-1 therapy. Nature 537 tumour activity. Virology 301 43–52. (doi:10.1006/viro.2002.1584) 417–421. (doi:10.1038/nature19330) Loos M, Giese NA, Kleeff J, Giese T, Gaida MM, Bergmann F, Laschinger Irigoyen A, Gallego J, Guillen Ponce C, Vera R, Iranzo V, Ales I, Arevalo M, M WB & Friess H 2008 Clinical significance and regulation of the S, Pisa A, Martin M, Salud A, et al. 2017 Gemcitabine-erlotinib versus costimulatory molecule B7-H1 in pancreatic cancer. Cancer Letters gemcitabine-erlotinib-capecitabine in the first-line treatment of 268 98–109. (doi:10.1016/j.canlet.2008.03.056) patients with metastatic pancreatic cancer: Efficacy and safety results Lu YC, Yao X, Crystal JS, Li YF, El-Gamil M, Gross C, Davis L, Dudley of a phase IIb randomised study from the Spanish TTD Collaborative ME, Yang JC, Samuels Y, et al. 2014 Efficient identification of

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T294 and therapeutic implications

mutated cancer antigens recognized by T cells associated with Muraki K, Nyhan K, Han L & Murnane JP 2012 Mechanisms of telomere durable tumor regressions. Clinical Cancer Research 20 3401–3410. loss and their consequences for chromosome instability. Frontiers in (doi:10.1158/1078-0432.CCR-14-0433) Oncology 2 135. Maida Y & Masutomi K 2015 Telomerase reverse transcriptase Nicholaou T, Ebert LM, Davis ID, McArthur GA, Jackson H, Dimopoulos moonlights: therapeutic targets beyond telomerase. Cancer Science N, Tan B, Maraskovsky E, Miloradovic L, Hopkins W, et al. 2009 106 1486–1492. (doi:10.1111/cas.12806) Regulatory T-cell-mediated attenuation of T-cell responses to the Mansmann G, Lau J, Balk E, Rothberg M, Miyachi Y & Bornstein SR NY-ESO-1 ISCOMATRIX vaccine in patients with advanced 2004 The clinically inapparent adrenal mass: update in diagnosis malignant melanoma. Clinical Cancer Research 15 2166–2173. and management. Endocrine Reviews 25 309–340. (doi:10.1210/ (doi:10.1158/1078-0432.CCR-08-2484) er.2002-0031) Nowak AK, Lake RA, Marzo AL, Scott B, Heath WR, Collins EJ, Frelinger Mardis ER 2017 Neoantigen discovery in human cancers. Cancer Journal JA & Robinson BW 2003 Induction of tumor cell in vivo 23 97–101. (doi:10.1097/PPO.0000000000000248) increases tumor antigen cross-presentation, cross-priming rather Marshall JL, Gulley JL, Arlen PM, Beetham PK, Tsang KY, Slack R, Hodge than cross-tolerizing host tumor-specific CD8 T cells. Journal of JW, Doren S, Grosenbach DW, Hwang J, et al. 2005 Phase I study of Immunology 170 4905–4913. (doi:10.4049/jimmunol.170.10.4905) sequential vaccinations with fowlpox-CEA(6D)-TRICOM alone and Oehler VG, Guthrie KA, Cummings CL, Sabo K, Wood BL, Gooley T, Yang T, sequentially with vaccinia-CEA(6D)-TRICOM, with and without Epping MT, Shou Y, Pogosova-Agadjanyan E, et al. 2009 The granulocyte-macrophage colony-stimulating factor, in patients with preferentially expressed antigen in melanoma (PRAME) inhibits myeloid carcinoembryonic antigen-expressing carcinomas. Journal of Clinical differentiation in normal hematopoietic and leukemic progenitor cells. Oncology 23 720–731. (doi:10.1200/JCO.2005.10.206) Blood 114 3299–3308. (doi:10.1182/blood-2008-07-170282) McCall KD, Harii N, Lewis CJ, Malgor R, Kim WB, Saji M, Kohn AD, Palena C, Zhu M, Schlom J & Tsang KY 2004 Human B cells that Moon RT & Kohn LD 2007 High basal levels of functional toll-like hyperexpress a triad of costimulatory molecules via avipox-vector receptor 3 (TLR3) and noncanonical Wnt5a are expressed in infection: an alternative source of efficient antigen-presenting cells. papillary thyroid cancer and are coordinately decreased by Blood 104 192–199. (doi:10.1182/blood-2003-09-3211) phenylmethimazole together with cell proliferation and migration. Papewalis C, Fassnacht M, Willenberg HS, Domberg J, Fenk R, Rohr UP, Endocrinology 148 4226–4237. (doi:10.1210/en.2007-0459) Schinner S, Bornstein SR, Scherbaum WA & Schott M 2006 Dendritic McDonnell AM, Lesterhuis WJ, Khong A, Nowak AK, Lake RA, Currie AJ cells as potential adjuvant for immunotherapy in adrenocortical & Robinson BW 2015 Tumor-infiltrating dendritic cells exhibit carcinoma. Clinical Endocrinology 65 215–222. defective cross-presentation of tumor antigens, but is reversed by (doi:10.1111/j.1365-2265.2006.02576.x) chemotherapy. European Journal of Immunology 45 49–59. Poli A, Michel T, Theresine M, Andres E, Hentges F & Zimmer J 2009 (doi:10.1002/eji.201444722) CD56bright natural killer (NK) cells: an important NK cell subset. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, Immunology 126 458–465. (doi:10.1111/j.1365-2567.2008.03027.x) Jamal-Hanjani M, Wilson GA, Birkbak NJ, Hiley CT, et al. 2016 Postow MA, Callahan MK & Wolchok JD 2015 Immune checkpoint Clonal neoantigens elicit T cell immunoreactivity and sensitivity to blockade in cancer therapy. Journal of Clinical Oncology 33 immune checkpoint blockade. Science 351 1463–1469. (doi:10.1126/ 1974–1982. (doi:10.1200/JCO.2014.59.4358) science.aaf1490) Rafei H, El-Bahesh E, Finianos A, Nassereddine S & Tabbara I 2017 Middleton G, Silcocks P, Cox T, Valle J, Wadsley J, Propper D, Coxon F, Immune-based therapies for non-small cell lung cancer. Anticancer Ross P, Madhusudan S, Roques T, et al. 2014 Gemcitabine and Research 37 377–387. (doi:10.21873/anticanres.11330) Endocrine-Related Cancer Endocrine-Related capecitabine with or without telomerase peptide vaccine GV1001 in Renner U, De Santana EC, Gerez J, Frohlich B, Haedo M, Pereda MP, patients with locally advanced or metastatic pancreatic cancer Onofri C, Stalla GK & Arzt E 2009 Intrapituitary expression and (TeloVac): an open-label, randomised, phase 3 trial. Lancet Oncology regulation of the gp130 cytokine interleukin-6 and its implication in 15 829–840. (doi:10.1016/S1470-2045(14)70236-0) pituitary physiology and pathophysiology. Annals of the New York Mills CD 2012 M1 and M2 macrophages: oracles of health and disease. Academy of Sciences 1153 89–97. (doi:10.1111/j.1749-­ Critical Reviews in Immunology 32 463–488. (doi:10.1615/ 6632.2008.03970.x) CritRevImmunol.v32.i6.10) Ries CH, Cannarile MA, Hoves S, Benz J, Wartha K, Runza V, Rey-Giraud Mittal D, Gubin MM, Schreiber RD & Smyth MJ 2014 New insights into F, Pradel LP, Feuerhake F, Klaman I, et al. 2014 Targeting tumor- cancer and its three component phases – associated macrophages with anti-CSF-1R antibody reveals a strategy elimination, equilibrium and escape. Current Opinion in Immunology for cancer therapy. Cancer Cell 25 846–859. (doi:10.1016/j. 27 16–25. (doi:10.1016/j.coi.2014.01.004) ccr.2014.05.016) Miyashita T, Miki K, Kamigaki T, Makino I, Nakagawara H, Tajima H, Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Takamura H, Kitagawa H, Fushida S, Ahmed AK, et al. 2017 Low- Lee W, Yuan J, Wong P, Ho TS, et al. 2015 Cancer immunology. dose gemcitabine induces major histocompatibility complex class Mutational landscape determines sensitivity to PD-1 blockade in I-related chain A/B expression and enhances an antitumor innate non-small cell lung cancer. Science 348 124–128. (doi:10.1126/ immune response in pancreatic cancer. Clinical and Experimental science.aaa1348) Medicine 17 19–31. (doi:10.1007/s10238-015-0394-x) Ryder M, Ghossein RA, Ricarte-Filho JC, Knauf JA & Fagin JA 2008 Morse MA, Hall JR & Plate JM 2009 Countering tumor-induced Increased density of tumor-associated macrophages is associated immunosuppression during immunotherapy for pancreatic cancer. with decreased survival in advanced thyroid cancer. Endocrine-Related Expert Opinion on Biological Therapy 9 331–339. Cancer 15 1069–1074. (doi:10.1677/ERC-08-0036) (doi:10.1517/14712590802715756) Ryder M, Gild M, Hohl TM, Pamer E, Knauf J, Ghossein R, Joyce JA & Muller-Hermelink N, Braumuller H, Pichler B, Wieder T, Mailhammer R, Fagin JA 2013 Genetic and pharmacological targeting of CSF-1/ Schaak K, Ghoreschi K, Yazdi A, Haubner R, Sander CA, et al. 2008 CSF-1R inhibits tumor-associated macrophages and impairs BRAF- TNFR1 signaling and IFN-gamma signaling determine whether T induced thyroid cancer progression. PLoS ONE 8 e54302. cells induce tumor dormancy or promote multistage carcinogenesis. (doi:10.1371/journal.pone.0054302) Cancer Cell 13 507–518. (doi:10.1016/j.ccr.2008.04.001) Ryner L, Guan Y, Firestein R, Xiao Y, Choi Y, Rabe C, Lu S, Fuentes E, Munder M 2009 Arginase: an emerging key player in the mammalian Huw LY, Lackner MR, et al. 2015 Upregulation of periostin and immune system. British Journal of Pharmacology 158 638–651. reactive stroma is associated with primary chemoresistance and (doi:10.1111/j.1476-5381.2009.00291.x) predicts clinical outcomes in epithelial ovarian cancer. Clinical

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access Thematic Review S Gasser et al. Tumour microenvironment 24:12 T295 and therapeutic implications

Cancer Research 21 2941–2951. (doi:10.1158/1078-0432.CCR-14- Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, 3111) Sucker A, Hillen U, Geukes Foppen MH, Goldinger SM, et al. 2015 Sanders KL, Fox BA & Bzik DJ 2015 Attenuated toxoplasma gondii Genomic correlates of response to CTLA-4 blockade in metastatic stimulates immunity to pancreatic cancer by manipulation of melanoma. Science 350 207–211. (doi:10.1126/science.aad0095) myeloid cell populations. Cancer Immunology Research 3 891–901. Vigneron N 2015 Human tumor antigens and cancer immunotherapy. (doi:10.1158/2326-6066.CIR-14-0235) BioMed Research International 2015 948501. Sansom DM 2000 CD28, CTLA-4 and their ligands: who does what and Vonderheide RH & Glennie MJ 2013 Agonistic CD40 antibodies and to whom? Immunology 101 169–177. (doi:10.1046/j.1365-­ cancer therapy. Clinical Cancer Research 19 1035–1043. 2567.2000.00121.x) (doi:10.1158/1078-0432.CCR-12-2064) Schumacher TN & Schreiber RD 2015 Neoantigens in cancer Wang K, Wei G & Liu D 2012 CD19: a biomarker for B cell immunotherapy. Science 348 69–74. (doi:10.1126/science.aaa4971) development, diagnosis and therapy. Experimental Senbabaoglu Y, Gejman RS, Winer AG, Liu M, Van Allen EM, de Velasco Hematology and Oncology 1 36. (doi:10.1186/2162-3619-1-36) G, Miao D, Ostrovnaya I, Drill E, Luna A, et al. 2016 Tumor immune Ward LS 2014 Immune response in thyroid cancer: widening the microenvironment characterization in clear cell renal cell carcinoma boundaries. Scientifica 2014 article ID 125450. identifies prognostic and immunotherapeutically relevant messenger Weber CA, Mehta PJ, Ardito M, Moise L, Martin B & De Groot AS 2009 RNA signatures. Genome Biology 17 231. (doi:10.1186/s13059-016- T cell epitope: friend or foe? Immunogenicity of biologics in 1092-z) context. Advanced Drug Delivery Reviews 61 965–976. (doi:10.1016/j. Silver DA, Pellicer I, Fair WR, Heston WD & Cordon-Cardo C 1997 addr.2009.07.001) Prostate-specific membrane antigen expression in normal and Weber JS, Vogelzang NJ, Ernstoff MS, Goodman OB, Cranmer LD, malignant human tissues. Clinical Cancer Research 3 81–85. Marshall JL, Miles S, Rosario D, Diamond DC, Qiu Z, et al. 2011 A Smith KA, Qiu Z, Wong R, Tam VL, Tam BL, Joea DK, Quach A, Liu X, phase 1 study of a vaccine targeting preferentially expressed antigen Pold M, Malyankar UM, et al. 2011 Multivalent immunity targeting in melanoma and prostate-specific membrane antigen in patients tumor-associated antigens by intra-lymph node DNA-prime, peptide- with advanced solid tumors. Journal of Immunotherapy 34 556–567. boost vaccination. Cancer Gene Therapy 18 63–76. (doi:10.1038/ (doi:10.1097/CJI.0b013e3182280db1) cgt.2010.45) Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, S, Franci C, Cheung TK, Fritsche J, Weinschenk T, et al. 2014 Walsh LA, Postow MA, Wong P, Ho TS, et al. 2014 Genetic basis for Predicting immunogenic tumour mutations by combining mass clinical response to CTLA-4 blockade in melanoma. New England spectrometry and exome sequencing. Nature 515 572–576. Journal of Medicine 371 2189–2199. (doi:10.1056/NEJMoa1406498) (doi:10.1038/nature14001) Teng MW, Ngiow SF, Ribas A & Smyth MJ 2015 Classifying cancers Yarchoan M, Johnson BA, 3rd, Lutz ER, Laheru DA & Jaffee EM 2017 based on T-cell infiltration and PD-L1. Cancer Research 75 Targeting neoantigens to augment antitumour immunity. Nature 2139–2145. (doi:10.1158/0008-5472.CAN-15-0255) Reviews Cancer 17 209–222. (doi:10.1038/nrc.2016.154) Thiele JO, Lohrer P, Schaaf L, Feirer M, Stummer W, Losa M, Lange M, Zajac P, Schultz-Thater E, Tornillo L, Sadowski C, Trella E, Mengus C, Tichomirowa M, Arzt E, Stalla GK, et al. 2003 Functional in vitro Iezzi G & Spagnoli GC 2017 MAGE-A antigens and cancer studies on the role and regulation of interleukin-6 in human immunotherapy. Frontiers in Medicine 4 18. (doi:10.3389/ somatotroph pituitary adenomas. European Journal of Endocrinology fmed.2017.00018) 149 455–461. (doi:10.1530/eje.0.1490455) Zhang J, Xu X, Shi M, Chen Y, Yu D, Zhao C, Gu Y, Yang B, Guo S, Endocrine-Related Cancer Endocrine-Related Tichomirowa M, Theodoropoulou M, Lohrer P, Schaaf L, Losa M, Uhl E, Ding G, et al. 2017 CD13hi Neutrophil-like myeloid-derived Lange M, Arzt E, Stalla GK & Renner U 2005 Bacterial endotoxin suppressor cells exert immune suppression through Arginase 1 (lipopolysaccharide) stimulates interleukin-6 production and inhibits expression in pancreatic ductal adenocarcinoma. Oncoimmunology 6 growth of pituitary tumour cells expressing the toll-like receptor 4. e1258504. (doi:10.1080/2162402X.2016.1258504) Journal of Neuroendocrinology 17 152–160. (doi:10.1111/j.1365-­ Zhang W, Niu Y, Zou H, Luo L, Liu Q & Wu W 2015 Accurate prediction 2826.2005.01286.x) of immunogenic T-cell epitopes from epitope sequences using the Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, genetic algorithm-based ensemble learning. PLoS ONE 10 e0128194. Trivett MK, Etemadmoghadam D, Locandro B, et al. 2008 Novel (doi:10.1371/journal.pone.0128194) molecular subtypes of serous and endometrioid ovarian cancer Zitvogel L, Kepp O & Kroemer G 2011 Immune parameters affecting the linked to clinical outcome. Clinical Cancer Research 14 5198–5208. efficacy of chemotherapeutic regimens. Nature Reviews Clinical (doi:10.1158/1078-0432.CCR-08-0196) Oncology 8 151–160. (doi:10.1038/nrclinonc.2010.223)

Received in final form 25 July 2017 Accepted 27 July 2017 Accepted Preprint published online 28 July 2017

http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-17-0146 Printed in Great Britain Downloaded from Bioscientifica.com at 09/23/2021 06:28:41PM via free access