<<

(2015) 34, 6029–6039 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc

REVIEW Emerging strategies for immunoprevention

JC Roeser1, SD Leach2 and F McAllister3

The crucial role of the immune system in the formation and progression of tumors has been widely accepted. On one hand, the surveillance role of the immune system plays an important role in endogenous tumor prevention, but on the other hand, in some special circumstances such as in chronic inflammation, the immune system can actually contribute to the formation and progression of tumors. In recent years, there has been an explosion of novel targeted immunotherapies for advanced . In the present manuscript, we explore known and potential various types of cancer prevention strategies and focus on nonvaccine- based cancer preventive strategies targeting the immune system at the early stages of tumorigenesis.

Oncogene (2015) 34, 6029–6039; doi:10.1038/onc.2015.98; published online 14 September 2015

INTRODUCTION Differences between chemoprevention and immunoprevention. As cancer is a collection of multiple complex individual diseases Whereas immunoprevention focuses on directly or indirectly that happen in different sites of the body and progress in different targeting the immune system, chemopreventive strategies utilize ways, a large amount of information is required to understand drugs or natural compounds to prevent cancer development.3 how to prevent each cancer type. Existing interventions for cancer Less than a dozen chemopreventive agents have been FDA prevention focus on the use of chemopreventive agents or approved: and for fi prevention,4,5 for colorectal polyps prevention in designed and tested for prevention of speci c cancer 6,7 8,9 types. Cancer vaccines that aim to manipulate the patient’s own FAP, BCG and for bladder cancer prevention and several topical agents for prevention of in patients immune system to destroy existing cancer cells are called fl ‘therapeutic vaccines’.1 Cancer vaccines that target the immune with actinic keratosis: uorouracil, diclofenac, masoprocol and ingenol mebutate.10 system to prevent the development of cancer are called ‘prophylactic vaccines’ and could potentially be used for cancer immunoprevention. However, there are many other strategies to Differences between immunoprevention and immunotherapy prevent cancer aside from vaccines. In the present manuscript, we The term ‘immunoprevention’ refers to the manipulation of the will review existing and potential cancer prevention strategies and immune system for disease prophylaxis, while ‘immunotherapy’ is focus on nonvaccine approaches for the prevention of cancer reserved for the treatment of existing disease by targeting the targeting the immune system. immune system. ‘Immunoprevention’ or ‘immunoprophylaxis’ represent one of the most successful types of interventions ever developed. Preventive vaccines have practically eliminated infectious diseases such as polio. In contrast, immunotherapies Types of cancer interventions against chronic infectious diseases have not shown enough There are two global interventions to target cancer: preventive efficacy due to the mechanisms developed by infectious and therapeutic strategies (Table 1). Cancer prevention includes all pathogens to avoid recognition and elimination by the immune approaches that aim to reduce the risk and incidence of cancer. system. Until very recently, the same was believed about cancer There are two types of cancer-preventive strategies: primary and immunotherapies, with most of strategies failing due to the secondary. Primary cancer prevention focuses on health counsel- tolerance and escape of detection by the immune system.11 ing, education and environmental control to decrease exposure to However, in the past decade, huge advances have been made in risk factors.2 More than 30% of cancer deaths could be prevented the immunotherapy field, most of them based on a better by avoiding key risk factors, including use, an unhealthy understanding of the molecular events leading to tumor 12 diet, abuse, sexually transmitted HPV infections and so on. tolerance. Tobacco is the single most important risk factor for cancer causing 22% of global cancer deaths. Secondary cancer prevention Immunoprevention of tumors caused by infectious agents encompasses interventions leading to the diagnosis and control Two different types of tumors can be targeted by immunopreven- of cancer and precancerous lesions at the earliest stage possible, tion, tumors caused by infectious agents or the ones caused by including screening and active interventions. These active noninfectious agents. Effective vaccines are available for prevention strategies can be further divided in two types: associated with cancer development. The against the chemoprevention and immunoprevention. hepatitis B initially developed for hepatitis prevention has

1Department of . Johns Hopkins University. Maryland, MD, USA; 2The David M. Rubenstein Center for Pancreatic , Memorial Sloan Kettering Cancer Center, New York, NY, USA and 3Department of Clinical Cancer Prevention. The University of Texas MD Anderson Cancer Center, Houston, TX, USA. Correspondence: Dr F McAllister, Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 1360, Houston, TX 77030, USA. E-mail: [email protected] Received 8 December 2015; revised 25 February 2015; accepted 27 February 2015; published online 14 September 2015 Cancer immunoprevention JC Roeser et al 6030

Table 1. Modalities of cancer prevention and therapy

Cancer

Prevention Therapy

Primary Secondary Chemo Targeted Immuno therapy therapy therapy

Active Targeted Screening Therapeutic prevention immuno- vaccines activators

Chemo Immuno prevention prevention

Infectious Non infectious

Prophylactic Immunomodulators vaccines

Targeted Targeted Anti-inflammatories immuno-activators

also been shown to reduce the incidence and mortality of liver to (IFN)-γ in tumor-bearing mice or mice .13,14 In 2006, the FDA approved two vaccines lacking the IFN-γ receptor showed accelerated tumor growth developed primarily for the prevention of compared with control mice.18,19 Studies using perforin-deficient associated with HPV infections: (Merck) and mice have shown increased susceptibility to both chemically (GlaxoSmithKline, Rixensart, Belgium). Gardasil is a quadrivalent induced and spontaneous cancer models.20 Mice lacking the vaccine that protects against four HPV types: HPVs 6, 11, 16 and recombinase-activating gene (RAG)-2 cannot somatically rear- 18. Cervarix is a bivalent vaccine against HPVs 16 and 18. These range lymphocyte receptors and cannot produce 15 vaccines can reduce the incidence of cervical cancer by 70%. peripheral αβ T cells, B cells, NKT cells (NKTs) or γδ T. As RAG-2 is selectively expressed in the lymphoid system, RAG-2− / − mice Immunoprevention of tumors caused by noninfectious agents provided an appropriate model to exclusively study the effects of There are two kinds of approaches to noninfectious tumor host lymphocyte deficiency on tumor development. Following − / − immunoprevention: one that targets specific (vaccines) subcutaneous injection of a chemical RAG-2 mice or another that targets nonspecific immune components. The developed at the injection site faster and with greater 21,22 drugs developed for the latter approach will be called ‘immune- frequency than strain-matched controls. Data about the role modulators’ in the present manuscript. At this point, immunopre- of immunosurveillance in cancer prevention are not only restricted 23 ventive approaches for tumors of non-infectious cause are at the to preclinical mouse models, but also exists in humans. As an preclinical level of development. Multiple transgenic mouse example of this, approximately a fourfold increase in the incidence models harboring activated have been produced in of de novo malignant melanoma after organ transplantation has 24 the past decade and they can be used to test prevention been reported. In spite of that, tumors do develop in the strategies by targeting the immune system.16 presence of a functional immune system. The protection provided by the immune system may not be sufficient and immune- The immunosurveillance theory and implications for preventive strategies aiming to promote antitumor immune immunoprevention defenses may bring a proportional decrease in tumor incidence. In the 1950s, Thomas and Burnet developed the theory of tumor immunosurveillance in which they proposed that the immune Theory of immunoediting and implications for immunoprevention system protects against developing tumors by attacking abnormal The immunoediting theory later replaced the immunosurveillance cells before they progress to invasive malignancy. However, when theory to reflect the increasingly apparent dual role of immunity in this hypothesis was tested in CBA/H nude mice, the most not only preventing but also in sculpting the tumoral process. congenitally immunodeficient mice available in 1970s, mice were Dunn et al.25 proposed that the process of immunoediting was found to develop spontaneous tumors and methylcholanthrene- comprised by three phases and named them ‘three Es of cancer induced sarcomas in a similar rate to wild-type mice.17 The immunoediting’: elimination, equilibrium and escape. The first findings brought skepticism about the immunosurveillance phase, elimination, represents the original concept of immuno- theory. However, two issues in this experiment were found surveillance and when successful, the whole process of immu- decades later. First, nude mice are not entirely immunodeficient as noediting is completed. In the second phase, equilibrium, the host they still maintain natural killer (NK) cells and low numbers of immune system and the tumor cells that survived the elimination T cells. Second, the CBA/H strain of mice expresses a highly active step enter into a state in which the immune system imposes isoform of the enzyme that metabolizes methylcholanthrene to its selective pressure on the tumor cells that is enough to contain, carcinogenic form. Subsequent studies using neutralizing but not completely remove the tumor, resulting in a tumor

Oncogene (2015) 6029 – 6039 © 2015 Macmillan Publishers Limited Cancer immunoprevention JC Roeser et al 6031 composed of many genetically unstable and mutating tumoral IL-6-gp130-STAT3 axis was fundamentally required for the cells. In the third phase, escape, tumor cells selected by the orchestration of the inflammatory process.34 equilibrium phase now grow and become clinically detectable. IL-6 and its downstream targets are involved in the regulation of Some of the escape mechanisms include: elaboration of cell proliferation, survival and metabolism, explaining the close immunosuppressive (TGF-β), growth factors (VEGF), association between IL-6 signaling and tumorigenesis.34 IL-6 recruitment of regulatory T cells and changes in tumor cells that regulates normal and tumor stem cell self-renewal through affect their recognition by immune effector cells (loss of MHC, Notch-3 activation and upregulation of the hypoxia response development of IFN-γ insensitivity, expression of anti-apoptotic protein carbonic anhydrase IX (CA-IX), which creates a permissive signals and so on).26 The accumulation of metabolic enzymes, environment for breast cancer stem cells survival in hypoxic 35 36 indoleamine 2,3-dioxygenase and arginase, that suppress T-cell conditions. Gao et al. have shown that activated EGFR induces proliferation and activation as well as the high expression of epithelial expression of IL-6, which induces STAT3 (signal tolerance-inducing molecules (PD-1 and CTLA-4) are additional transducers and activators of transcription 3) activation in lung mechanisms that contribute to tumor escape.27 Further under- carcinoma. Lesina et al. have demonstrated that pancreatic standing of the mechanisms responsible for tumor equilibrium expression of oncogenic Kras(G12D) causes an IL-6 transsignal- and escape will be invaluable to generate better strategies for ing-dependent activation of Stat3/Socs3 required to promote 37 cancer immunoprevention. pancreatic tumorigenesis. In a model of colitis-associated cancer, IL-6 produced by hematopoietic cells enhances growth38 and its ablation reduces the number and size of colonic Advantages of Immunoprevention without vaccines .39 Toclizumab, a humanized anti–IL-6 receptor mono- A key issue in developing effective cancer preventive vaccines is clonal , globally blocks IL-6 activities since it competes for the large variability of tumor antigens (even within the same both the membrane-bound and soluble types of IL-6 receptors.40 patient tumor). Even in the event of a very common antigen This antibody was approved by the FDA in 2010 for the treatment 28 selected as the target, results have been less than optimal. of patients with rheumatoid arthritis that have failed at least one Immunopreventive strategies that target the immune system anti-TNF therapy. When targeting IL-6 with antibodies, the impact directly would have to deal with less variability and therefore, of blocking classical membrane-bound signaling and IL-6 trans- potentially could result in more effective outcomes as evidenced signaling will have to be considered. A recent Phase I clinical trial by the recently FDA-approved nonvaccine cancer immunotherapies.12 of a chimeric IL-6 monoclonal antibody, siltuximab, has reported good tolerance in cancer patients and phase II therapeutic trials 41 Risk profile of immune-preventive agents are currently ongoing. Clinical preventive strategies targeting fl Given the fact that preventive strategies are directed to healthy IL-6 would be highly promising, particularly in in ammation- patients who, even in the case of pertaining to high-risk groups, associated cancers. A proof of principle for anti-IL-6 immune prevention of in preclinical model with few may actually never develop cancer, safety is a major concern. 42 As immune-preventive agents continue to being developed, long-term side effects has been recently reported. major attention should be focused on dissecting their risk profile. As some protumoral targets may also have a role in host defense Interleukin-8 against pathogens, strategies that aim to suppress protumoral Interleukin-8 (IL-8), also known as CXCL8, is a proinflammatory targets may actually result in an increased risk for infections.29 On CXC chemokine that binds to CXCR1 and CXCR2, two cell-surface the other hand, strategies that focus on inducing antitumoral G-protein-coupled receptors.43,44 The receptors CXCR1 and CXCR2 targets may actually result in the development of autoimmune have also been found overexpressed on cancer cells and multiple conditions.30 reports have described an autocrine IL-8 signaling loop associated with cell proliferation, invasion and angiogenesis as mechanisms 45–47 Shotgun vs targeted immune-preventive approaches to induce tumorigenesis in various models. Humanized monoclonal antibodies against IL-8 have been used in murine Broad anti-inflammatory agents such as celecoxib and fi models and have shown to decrease bladder cancer and have proven bene cious as preventive agents in high-risk melanoma growth in xenografts models.45,46 A liposomal encap- populations but even these drugs have several toxicities, including sulated small interfering RNA that supressed IL-8 expression in a cardiovascular toxicities and increased of bleeding risk, when used 31 model of ovarian cancer xenografts resulted in decreased chronically. It would be expected that better molecular 48 fi microvessel density and growth retardation. Targeting IL-8 characterization of the speci c immune events surrounding expression for cancer prevention would fail to account for the fi preneoplastic lesions may theoretically result in high ef cacy with signaling effect of other CXC-chemokines. In this regard, several few side effects through the use of targeted immune-preventive CXCR1/2 receptors49–55 inhibitors are now being developed and strategies. they may also have a potential utility for immunoprevention.

Targets for cancer Immunoprevention TH17 and Interleukin-17 IL-17A, a proinflammatory produced mainly by CD4+ T cells known as TH17 (T-helper 17) cells, has a key role in chronic inflammation-related human diseases and autoimmunity.56,57 In PROTUMORAL 58 addition to TH17 cells, IL-17A can also be produced by NKTs, Interleukin-6 gamma–delta T cells59 and CD8+T cells.60 IL-17A binds the The complex of IL-6 and IL-6R does not lead to signaling but heteromeric IL-17R complex consisting of at IL17RA and instead associates with the protein gp130 inducing its dimeriza- IL17RC.61 TGF-β and IL-6 are the most important cytokines 62 tion and starting intracellular signaling via the JAK/STAT and the required for TH17 differentiation, while IL-23 is crucial for its – – 32 63 Ras Raf MAPK signaling pathways. IL-6 is the only cytokine that phenotype maintenance. The differentiation of TH17 cells also in vivo utilizes both classical membrane-bound receptor signaling requires the expression of retinoic acid receptor-related orphan and trans-signaling through its soluble receptor.33 Using mice in nuclear receptor gamma (RORγt), which is a STAT-3-dependent 64 which the portions of gp130 have been genetically modified in transcription factor. TH17 cells are increased in the tumor models of inflammatory diseases led to the conclusion that the microenvironment during tumor development65,66 and several

© 2015 Macmillan Publishers Limited Oncogene (2015) 6029 – 6039 Cancer immunoprevention JC Roeser et al 6032 manuscripts have reported the role of IL-17 in promoting constitutive STAT3 phosphorylation,95 and the ablation of STAT3 tumorigenesis.65,67–72 The mechanisms for the protumorigenic results in impairment of malignant transformation by tyrosine effects include the induction of angiogenic and anti-apoptotic kinase oncoproteins.96–98 STAT3 signaling within the tumor factors, migration and invasion and recruitment of myeloid microenvironment induces a procarcinogenic cytokine, IL-23, derived suppresors cells. Conversely, TH17 cells have been also while inhibiting a central anticarcinogenic cytokine, IL-12, thereby reported to mediate antitumor effects by promoting CTLs shifting the balance of tumor immunity toward .99 activities, MHC antigen expression and production of IFNγ. The Small molecule inhibitors of STAT3 have been developed and antitumor effects reported were mostly described in the setting of used in the preclinical setting, but they mostly target upstream established cancer73 and some of them relied on IFNγ, rather than receptor and non-receptor tyrosine kinases, resulting in low on IL-17A secretion. The FDA has approved the use of specificity of action. Sorafenib, a multikinase inhibitor, decreased ustekinumab, an anti-IL-23 antibody that would decrease TH17 STAT3 phosphorylation in by inhibiting responses, for moderate-to-severe psoriatic arthritis.74 Ixekizumab phosphoinositide 3-kinase/Akt and MEK/ERK pathways.100 WP1-66, and secukinumab, IL-17A-specific antibodies and brodalumab, a JAK2 inhibitor, has also been used to inhibit STAT3 activity in a monoclonal antibody directed against IL-17RA, have shown safety model of renal cell carcinoma.101 RNA interference approach has and efficacy in early phase trials for autoimmune and chronic also been done to knock down STAT3 and in preclinical models it inflammatory conditions75–77 and are currently being tested in supressed tumorigenesis.102 Finally, a chemically modified double- phase III trials for autoimmune diseases. RORγt small molecule stranded STAT3 oligonucleotide decoy that inhibited tumorigen- inhibitors are currently being developed and under preclinical esis in murine models has also been clinically tested in a phase 0 testing. In any case, the potential idea of targeting IL-17 producing setting and proved to be safe and effective to selectively suppress cells as a strategy for cancer immunoprevention should consider STAT3 target genes in head and neck tumors.103 Given the key their impacts in autoimmunity as well as the potential protumoral role of STAT3 in promoting prostate, colon and pancreatic effects.78–80 Potential preventive strategies targeting IL-17 would be cancer,104–106 STAT3 inhibitors could be useful for cancer ofparticularinterestforinflammation-induced cancer. prevention in patients with high risk for these diseases.

Interleukin-1 PD-1/ PD-L1 The IL-1 family encompasses three proteins: IL-1α, IL-1β and the Programmed cell death-1 (PD-1), a CD28 family receptor, has a key IL-1 (IL-1ra).81,82 There are two IL-1 receptors, role in tumor tolerance.107 The interaction between PD-1/PD-L1 the biologically active IL-1 receptor type I (IL-1RI), agonistic results in the inhibition of T-lymphocyte effector functions, with receptor ubiquitously expressed with preferential binding to IL-1a subsequent downregulation of antitumor immune responses.108 and the IL-1 receptor type II (IL-1RII or decoy IL-1R), antagonist PD-1 has two ligands, PD-L1 and PD-L2 with a different pattern of receptor expressed on B cells, neutrophils and monocytes with expression and regulation. PD-L1 is expressed in most hemato- preferential binding to IL-1β.83,84 The mechanisms responsible for poietic cells and in multiple tumors including melanoma, the IL-1- tumor growth promotion are: induction of prometastatic pancreatic, breast and lung tumors109–113 but PD-L2 expression genes (for example, matrix metalloproteinases) and stimulation of is restricted to macrophages, mast cells and dendritic cells (DCs). adjacent cells to produce angiogenic proteins and growth factors High expression of tumoral PD-L1 has been associated with poor (VEGF, IL-8, IL-6, TNFα and TGFβ).82,83,85 Cancer cells are able to prognosis in multiple studies.111,114 Preclinical studies have produce IL-1 or can induce its production from cells within the suggested that PD-1/PD-L1 signaling favors tumor immune tumor microenvironment.86 IL-1 has autocrine behavior by evasion and that blockade of the PD-1 pathway can restore inducing proliferation and invasion of the tumor cells and is also antitumor immune responses. Tumor cells that express PD-L1 a paracrine effector on the stromal cells. From a clinical have increased resistance to T-cell-mediated lysis with increased perspective, elevated IL-1 levels within the serum have been tumorigenesis and invasiveness, when compared with tumor cells associated with more aggressive tumors.87 that do not express PD-L1 and the administration of anti-PD-L1 The IL-1ra is an inhibitor of IL-1 and has been shown to antibodies reversed these effects.115 Also, PD-1-deficient mice decrease tumor growth, angiogenesis and metastases in murine presented inhibition of tumor growth, suggesting a strong xenograft models.84 IL-1ra is an approved treatment for patients antitumor immune response in the absence of PD-1/PD-L1 with rheumatoid arthritis, has shown some promising results in signaling.116 Finally, in mice with established tumors, the indolent multiple myeloma treatment88 and is now being tested administration of anti-PD-1 or anti-PD-L1 antibodies led to on other cancers. IL-1ra could represent effective novel immune- reduction in tumor size and increased survival.117 preventive agents but further studies are needed.81 In recent years, early phase studies reported clinical activity of PD-L1118 and PD-1119,120 monoclonal antibodies in patients with STAT3 advanced solid tumors. As larger therapeutic trials get conducted for efficacy validation, the utilization of these antibodies for tumor STATs are a family of transcription factors discovered in the early immunoprevention in high-risk populations may become a 1990s that regulate cell growth and differentiation through potential effective strategy, particularly if PD-1/PD-L1 biomarkers phosphorylation by receptor-associated kinases.89 STAT3 was are well defined in preneoplastic lesions and side effects are found to mediate the acute phase response observed with manageable.121 Given the success of these agents in melanoma inflammation and stress that is often mediated by interleukin-6 and non-small cell , immunoprevention targeting (IL-6) and other proinflammatory cytokines.90 Activated STAT3 is PD-1/PD-L1 might be of high relevance in patients with phosphorylated on tyrosine and forms a dimer through phospho- preneoplastic lesions associated with either disease. tyrosine/src homology 2 (SH2) domain interaction. The dimer enters the nucleus via interaction with importins and binds genes with critical role in cellular proliferation, survival, pluripotency, CTLA-4 invasion and angiogenesis, explaining its association with cancer CTLA-4 is a cell-surface protein constitutively expressed on T cells, development. Fibroblasts engineered to express an activated but upregulated by the interaction of the T-cell receptors and STAT3 formed tumors in nude mice, thus establishing STAT3 as an CD28.122 A splice variant of CTLA-4 may also be secreted by T cells oncogene.91 Increased STAT3 activity is present in 470% of in a soluble form that binds to T-cell receptors. CTLA-4 interacts solid tumors,92,93 and the inhibition of STAT3 can mediate with CD80 (B7-1) and CD86 (B7-2) and competitively competes tumor regression.94 Tyrosine kinase oncoproteins can target with CD28 for binding with these receptors, therefore, acting as an

Oncogene (2015) 6029 – 6039 © 2015 Macmillan Publishers Limited Cancer immunoprevention JC Roeser et al 6033 inhibitor of T-cell proliferation.123 The receptor CD28 is similar to Murine MDSCs are characterized by the coexpression of two CTLA4 and shares a homologous amino-acid sequence. CD28 myeloid differentiation antigens: Gr1 and CD11b.135 The normal binding causes costimulation and activation of T cells, whereas murine bone marrow can contain up to 30% cells with this CTLA-4 binding causes T-cell inhibition.122 As CTLA-4 provides a phenotype but the spleen contains only 2–4%. Human MDSCs regulatory signal that inhibits T-cell activation, it serves as an have a different phenotype, being defined by CD11b+/CD14 − immune checkpoint inhibitor to prevent over activation of expression.136 The levels of immune cells with these phenotypes T cells.124 Two antibodies have been tested against CTLA-4 have been shown to correlate with prognosis and overall – clinically: ipilimumab and tremelimumab. Ipilimumab has become survival.137 139 Multiple methods of inhibiting MDSCs are currently the standard of care for melanoma patients after a phase III trial under investigation. These methods involve (1) induction of showed survival benefit in patients with metastatic melanoma.125 MDSC maturation to nonsuppressive cells (all trans retinoic acid, Tremelimumab failed to show a survival advantage in a phase III D), (2) decrease MDSC levels (sunitinib, , 5-FU, clinical trial in melanoma126 and is currently being investigated in CDDO-Me) or (c) functional inhibition of MDSC (PDE-5 inhibitors, 140 combination with other agents. The recent success in targeting cyclooxygenase 2 inhibitors). These approaches may enhance CTLA-4, particularly in melanoma has led to increased interest in antitumor immunity through reduction in immune suppression understanding the role that CTLA4 has in immune surveillance and thus would be relevant in the immune-preventive scenario. and immunoprevention, particularly for patients with preneoplas- tic lesions. Tregs Regulatory T cells (Tregs) were initially called suppressive CD4+ Innate lymphoid cells T cells as they were found to negatively regulate tumor immunity 141 Innate lymphoid cells (ILCs) have been recently described as a leading to tumor growth in mice. The transcription factor Foxp3 group of innate immune cells that can regulate immunity, is crucial for the development and functionality of T regulatory fl cells and the IL-2 receptor α-chain, CD25, serves as the phenotypic in ammation and tissue repair in multiple anatomical compart- + ments. ILCs can regulate commensal bacterial communities, marker for CD4 T regulatory cells. Therefore, the classic regulatory contribute to resistance to bacterial pathogens, promote T cells are phenotyped as CD4+CD25+FOXP3+T cells (Tregs). inflammation and orchestrate tissue repair and wound Multiple possible suppressive mechanisms for regulatory T cells healing.127 RORγt+ ILCs that express and require the nuclear have been proposed: induction of B7-H4 expression by antigen- hormone receptor retinoic acid receptor related orphan receptor presenting cells, CTLA4-mediated suppression of T-cell activity and (RORγt), are involved in lymphoid tissue genesis and can produce direct inhibition of T-cell activation by the secreted cytokines IL-10 and TGF-b.142 The role of Tregs (CD4+CD25+Foxp3+) on cancer TH17-associated cytokines IL-17 and IL-22. On the basis of functional characteristics, three subpopulations of RORγt-depen- progression has been widely demonstrated in several preclinical models.143 Tumors trigger an increase of T-regulatory cells dent ILCs can be distinguished: (a) lymphoid tissue-inducer (LTi) (CD4+CD25+Foxp3+) numbers in lymph nodes and in the tumor cells, (b) ILCs dedicated to IL-22 production and (c) IL-17- microenvironment, leading to immunosuppression. Several producing ILCs. ILCs that produce both IL-17 and IL-22 have also reports have linked the abundance of Tregs in tumors with worse been found that may be cells derived from either IL-17- or IL-22- prognosis.144,145 producing cells.128 RORγt ILCs have recently been the target of is an alkylating agent that causes DNA increasing attention due to their key role in multiple models of crosslinking and is utilized as a chemotherapeutic agent for microbe-related tumorigenesis. In a murine model of - several types of tumors. High dose cyclophosphamide can induce induced colon cancer, innate lymphoid cells have been shown to 129 effective antitumor activity. However, low dose cyclophosphamide sustain tumorigenesis through production of interleukin-22. In can also induce improved antitumor immune responses as a result a melanoma model, the chemokine CCL21 expressed by the of Tregs depletion. This was demonstrated by the evidence that tumor cells recruited not only myeloid-derived suppressor cells cyclophosphamide treatment caused an immune-mediated that can directly suppress antitumor immune responses but also regression of a cyclophosphamide-resistant lymphoma.142 Several γ recruited ROR t+ ILCs that mediated the enhancement of tumor preclinical studies have confirmed the effect of cyclophosphamide growth as shown by the absence of effect in tumor growth in on Tregs depletion and clinical studies have also been conducted γ fi 130 ROR t-de cient mice. Further understanding of these cells with similar results.146 Studies have also shown that anti-CD25 will be required for the development of immune strategies antibodies induce tumor rejection in mice147,148 and humans. targeting them. However, this strategy is not as effective in advanced tumors, highlighting the critical role that Tregs may have in the tumor Myeloid-derived supressor cells immune-preventive setting. Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of early myeloid cells that are expanded in various Macrophages disease states with the capability of suppressing T-cell Macrophages are a major component of the inflammatory tumor 131–133 responses. In addition to their suppressive effect on stroma and macrophages that infiltrate tumors are called tumor- adaptive immune responses, MDSCs have also been reported to associated macrophages.149 Macrophages are classified in M1 regulate innate immune responses by modulating the cytokines (classically activated) and M2 (alternatively activated) phenotypes 134 secretion of macrophages. in analogy with the Th1 and Th2 classification.150,151 Macrophages Myeloid progenitor cells and immature myeloid cells (IMCs) have functional plasticity and can change their functional status in constitute the major types of MDSCs. In healthy patients, IMCs that response to changes in the microenvironment.152 When macro- are generated in the bone marrow quickly differentiate into phages are exposed to lipopolysaccharides and IFN-α, they mature granulocytes, macrophages or DCs. In contrast, during polarize to an M1 phenotype with antitumor properties and pathological states like cancer and infections, a block in the when they are exposed to Th2 cytokines (IL-4, IL-13, IL-10), they differentiation of IMCs into mature cells results in an increase of polarize to an immune suppressive M2 phenotype that promotes the immature populations. Another relevant event during these tumor growth.150,153 Tumor-associated macrophages are a major pathological states is the upregulation of arginase1 (ARG1) and source of COX-2 and the selective COX-2 inhibitor celecoxib has inducible nitric oxide on IMCs, conferring them immune been shown to change the tumor-associated macrophages suppresive properties. phenotype from M2 to M1, in parallel to a reduction in number

© 2015 Macmillan Publishers Limited Oncogene (2015) 6029 – 6039 Cancer immunoprevention JC Roeser et al 6034 of colon polyps in a murine model of colon cancer.154 A large solid malignancies.174 Peptide based vaccines with adjuvants clinical trial has shown that celecoxib reduces the incidence of containing cytokines, chemokines or costimulatory molecules adenomas, but increased the risk of cardiovascular events.155 have been developed to amplify and direct the immune response Other selective COX2 inhibitors, with potentially less side effects, but are beyond the scope of the present review. are currently being tested and could represent potential chemopreventive strategies. In a different manner, a recent CD4+ T cells (TH1) murine and human study has shown that CD40 agonists can cause TH1 CD4+ T cells are driven by IL-12 activation of Stat4 and T-bet macrophage activation with rapid stromal infiltration and 175,176 156 transcription factors on naïve T cells that induces and up- tumoricidal effect in pancreatic adenocarcinoma. This led us regulates IFNγ.177 Ectopic expression of T-bet in T 2orT 17 cells to speculate that similar agents could be used for prevention in H H results in their conversion into IFNγ-producing TH1-like cells. The patients with pancreatic preneoplastic lesions. central role of IFNγ- producing CD4+ T cells in antitumor immunity has been known for a long time.178 CD4+ T cells have an Mast cells indispensable role in the generation of vaccine-related therapeutic Tumor-infiltrating mast cells remodel the tumor microenviron- primary immune responses for the generation of long-term ment and promote tumor growth. Mast-cell infiltration and immune memory.179 In multiple human tumors, TH1 CD4+ T-cell activation in tumors is mainly mediated by tumor-derived stem infiltrates have been found associated with a better prognosis.180 cell factor and its receptor c-Kit.157 Mast cells accumulate in the Specific effector immune cells proliferate within the tumor tumor microenvironment and exacerbate immunosuppression by microenvironment but the contribution to anticancer immunity inducing MDSC and Treg infiltration in an IL-17-dependent is unclear, given the immunosuppressive factors within the tumor. manner.158 The presence of mast cells in many precancerous Vaccines targeting specific antigens should be planned in lesions and tumors is associated with a poor prognosis, suggesting combination with agents that aim to decrease the immunosup- that mast cells may promote an immunosuppressive tumor pressive environment. microenvironment and impede the development of protective Cancers with high mutational loads have a high number of antitumor immunity.159–161 tumor-specific neoantigens. Recently, Linnemann et al.181 devel- Targeting mast cells can be achieved indirectly via targeting oped a screening platform to evaluate CD4+ T-cell responses to mast cells mediators with agents like anti-TNF monoclonal tumor neoepitopes in melanoma patients and the results antibodies (infliximab) or directly through the use of mast-cell- indicated that CD4+ T-cell reactivity toward patient-specific stabilizing agents like sodium cromoglycate. Preclinical work with neoantigens is a common feature of melanoma and suggest that + infliximab in DSS-induced colitis has decreased development of therapeutic and potentially preventive strategies to boost CD4 colorectal tumors in association with decreased mast cell T-cell responses may be effective in melanoma and other cancers numbers.162 Stabilization of mast cells through sodium cromogly- with similar mutational loads. cate has also shown efficacy in preclinical studies,163 but the specificity of this compound has been questioned.164 Interven- NKT cells tions aimed at inhibiting mast cells function hold promise as NKTs are a subset of T cells that recognize self- and microbial- potential effective cancer preventive strategy, particularly because derived glycolipids such as α-galactosylceramide (α-GalCer) of the low side effects of these agents. presented by an HLA class I-like molecule, CD1d. NKTs are capable of producing key cytokines of both Th1 and Th2 T cells that ANTITUMORAL provide signals for other immune cells to start innate and adaptive responses.182 Low levels of circulating NKTs can predict poor α IFN- clinical outcome in certain human tumors183 and tumor infiltration alpha (IFNs-α) are pleiotropic cytokines from the type I by NKT is a predictor of favorable outcomes.184 Treatment of mice IFN family, initially described for their antiviral activity achieved by with α-GalCer has been shown to suppress tumor metastasis185 directly inhibiting viral replication in infected cells.165 IFN-α can yet clinical trials have not shown efficacy.186 Novel α-GalCer induce anti-tumoral effects through induction/promotion of analogs have been designed to favor Th1-biased immunity, with apoptosis and inhibition of cell growth.166 Multiple studies have greater preclinical anticancer potential and are currently being shown that IFN-α can induce the rapid differentiation of tested.187 monocytes into activated DCs that are involved in effective anti- 167 α tumor T-cell immunity. Moreover, IFN- can induce polarization Dendritic cells of T-helper (TH) cells to TH1 cells and activation of cytotoxic 168 DCs have a critical role in linking innate and adaptive immunity. T cells. Finally, IFN-α can promote differentiation, 169 They have a key role in role in capturing and presenting antigens antibodies production and Ig class switching. In sum, IFN-α in the form of MHC II receptors to T cells stimulating them to has a broad effect on immune system activation and prolonged develop into effector T cells. In this role, DCs have a significant role activation can lead to autoimmune reactions. IFN-a is used for the in the tumor microenvironment, surrounding tumors and provid- treatment of multiple malignancies including melanoma and renal ing signals to other immune cells as to stimulate or suppress T-cell cell carcinoma,170,171 but its utilization for cancer prevention has activation.188 Tumor tolerant DCs are one reason that budding not been explored yet. tumors can escape immune surveillance. IL-10 secreted from tumors can inhibit the maturation of DCs, and promote DCs + CD8 T cells apoptosis, thereby promoting tumor tolerance.188 Potent DCs are It is well known that CD8+ T cells are important for antitumoral required for a strong antitumor immune response after che- responses. The expansion of tumor-specific CD8+ T cells can motherapy or radiation. There is a great degree of variability in the trigger effective antitumoral immunity,172 and this constitutes the location and number of DCs in tumors.189 However, most basis for multiple vaccine trials. Recent reports indicate that CD8+ evidence shows that although there are some levels of DCs T cells may also be capable of protumoral activity depending on present in tumors, they are suppressed by a variety of the antigen specificity.173 There is large evidence about the value immunosuppressing cytokines and other factors from the tumor of high numbers of T cells at the tumor site with CD8 memory microenvironment. DCs can also have prognostic value in treating T cells as a prognostic factor for overall survival in patients with patients. Studies have shown that tumors with DCs expressing low

Oncogene (2015) 6029 – 6039 © 2015 Macmillan Publishers Limited Cancer immunoprevention JC Roeser et al 6035

IMMUNO-MODULATORS EFFECTIVE IMMUNO-PREVENTION ANTI-INFLAMMATORIES IMMUNO-ACTIVATORS

-+

IMMUNE RESPONSE

Pro-tumoral Anti-tumoral

CYTOKINES CELL TYPES CYTOKINES CELL TYPES AND SIGNALS Tregs AND SIGNALS CD8+T cells IL-6 Stat3 TH17 CD4+ (TH1) γ IL-8 PD-1 ROR t+ ILC IFN-a NKT IL-17 CTLA-4 MDSC DC IL-1B M0. (M2) Mast cells

Figure 1. Immune response equilibrium. Activation of protumoral signals and cytokines as well as recruitment of specific immune cells populations with protumoral activity (Red circle) are counteracted by activation of signals and cytokines with antitumoral effects as well as tumor infiltration by immune cells populations with antitumoral effect (Green circle). Effective immunopreventive strategies should focus on either opposing the protumoral signals and immune cells by the use of anti-inflammatories or by increasing the activity of the anti-tumoral signals and immune cells through the use of immune-activators.

levels of CD86 and greater levels of IL-10, generally are resistant to Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 1998; 90: therapy and have a poor outcome.190 All of these reasons make 1371–1388. DC stimulation or prevention of DC tolerance, potential good 5 Vogel VG, Costantino JP, Wickerham DL, Cronin WM, Cecchini RS, Atkins JN et al. strategies for tumor immune prevention. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 2006; 295:2727–2741. CONCLUSIONS 6 Phillips RK, Wallace MH, Lynch PM, Hawk E, Gordon GB, Saunders BP et al. A randomised, double blind, placebo controlled study of celecoxib, a selective The potential combination of agents that decrease or suppress the cyclooxygenase 2 inhibitor, on duodenal polyposis in familial adenomatous protumoral events with agents that boost the anti-tumoral polyposis. Gut 2002; 50:857–860. immunity applied to selected high risk populations may have a 7 Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB et al. The strong impact in decreasing cancer incidence (Figure 1). By effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous 342 – gaining further understanding on the cancer microenvironment, polyposis. N Eng J Med 2000; :1946 1952. 8 Steinberg G, Bahnson R, Brosman S, Middleton R, Wajsman Z, Wehle M. Efficacy in the future we might be able to not only identify appropriate fi and safety of valrubicin for the treatment of Bacillus Calmette-Guerin refractory high-risk patients who may bene t from immunoprevention, but of the bladder. The Valrubicin Study Group. The Journal of also determine the precise inflammatory mediators that place urology 2000; 163: 761–767. them at risk, so that individualized immunoprevention might 9 Sylvester RJ. Bacillus Calmette-Guerin treatment of non-muscle invasive become a reality. bladder cancer. Int J Urol 2011; 18: 113–120. 10 Lebwohl M, Swanson N, Anderson LL, Melgaard A, Xu Z, Berman B. Ingenol mebutate gel for actinic keratosis. N Eng J Med 2012; 366: 1010–1019. CONFLICT OF INTEREST 11 Pardoll DM. Spinning molecular immunology into successful immunotherapy. Nat Rev Immunol 2002; 2: 227–238. The authors declare no conflict of interest. 12 Pardoll DM. Immunology beats cancer: a blueprint for successful translation. Nat Immunol 2012; 13: 1129–1132. ACKNOWLEDGEMENTS 13 Chang MH, Chen CJ, Lai MS, Hsu HM, Wu TC, Kong MS et al. Universal hepatitis B vaccination in Taiwan and the incidence of hepatocellular carcinoma in children. This work was supported by 2014 Pancreatic Cancer Action Network-AACR Career Taiwan Childhood Hepatoma Study Group. NEngJMed1997; 336: 1855–1859. Development Award, grant 14-20-25-MCAL. 14 Gwack J, Park SK, Lee EH, Park B, Choi Y, Yoo KY. Hepatitis B vaccination and mortality reduction in Korean children and adolescents. Asian Pac J Cancer Prev 2011; 12:2205–2208. REFERENCES 15 CDC. Vaccine information statement: HPV (human papillomavirus) vaccine 1 Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF et al. http://wwwcdcgovezproxywelchjhmiedu/vaccines/pubs/vis/downloads/vis-hpv- Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Eng J gardasilpdf. Med 2010; 363:411–422. 16 Olive KP, Tuveson DA. The use of targeted mouse models for preclinical testing 2 Spratt JS. The primary and secondary prevention of cancer. J Surg Oncol 1981; of novel cancer therapeutics. Clin Cancer Res 2006; 12: 5277–5287. 18: 219–230. 17 Stutman O. Tumor development after 3-methylcholanthrene in immunologically 3 Patterson SL, Colbert Maresso K, Hawk E. Cancer chemoprevention: successes deficient athymic-nude mice. Science 1974; 183:534–536. and failures. Clin Chem 2013; 59:94–101. 18 Dighe AS, Richards E, Old LJ, Schreiber RD. Enhanced in vivo growth and 4 Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin WM resistance to rejection of tumor cells expressing dominant negative IFN gamma et al. Tamoxifen for prevention of breast cancer: report of the National Surgical receptors. Immunity 1994; 1:447–456.

© 2015 Macmillan Publishers Limited Oncogene (2015) 6029 – 6039 Cancer immunoprevention JC Roeser et al 6036 19 Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ et al. Demon- 46 Mian BM, Dinney CP, Bermejo CE, Sweeney P, Tellez C, Yang XD et al. Fully stration of an interferon gamma-dependent tumor surveillance system in human anti-interleukin 8 antibody inhibits tumor growth in orthotopic bladder immunocompetent mice. Proc Natl Acad Sci USA 1998; 95: 7556–7561. cancer xenografts via down-regulation of matrix metalloproteases and nuclear 20 van den Broek ME, Kagi D, Ossendorp F, Toes R, Vamvakas S, Lutz WK et al. factor-kappaB. Clin Cancer Res 2003; 9: 3167–3175. Decreased tumor surveillance in perforin-deficient mice. J Exp Med 1996; 184: 47 Yao C, Lin Y, Chua MS, Ye CS, Bi J, Li W et al. Interleukin-8 modulates growth and 1781–1790. invasiveness of estrogen receptor-negative breast cancer cells. Int J Cancer 2007; 21 Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ et al. IFNgamma 121: 1949–1957. and lymphocytes prevent primary tumour development and shape tumour 48 Merritt WM, Lin YG, Spannuth WA, Fletcher MS, Kamat AA, Han LY et al. Effect of immunogenicity. Nature 2001; 410: 1107–1111. interleukin-8 gene silencing with liposome-encapsulated small interfering RNA 22 Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M et al. RAG-2- on ovarian growth. J Natl Cancer Inst 2008; 100: 359–372. deficient mice lack mature lymphocytes owing to inability to initiate V(D)J 49 Dwyer MP, Yu Y, Chao J, Aki C, Chao J, Biju P et al. Discovery of 2-hydroxy-N, rearrangement. Cell 1992; 68: 855–867. N-dimethyl-3-{2-[[(R)-1-(5- methylfuran-2-yl)propyl]amino]-3,4-dioxocyclobut-1- 23 Teng MW, Swann JB, Koebel CM, Schreiber RD, Smyth MJ. Immune-mediated enylamino}benzamide (SCH 527123): a potent, orally bioavailable CXCR2/CXCR1 dormancy: an equilibrium with cancer. J Leukoc Biol 2008; 84:988–993. receptor antagonist. J Med Chem 2006; 49:7603–7606. 24 Sheil AG. Cancer after transplantation. World J Surg 1986; 10:389–396. 50 Gonsiorek W, Fan X, Hesk D, Fossetta J, Qiu H, Jakway J et al. Pharmacological 25 Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev characterization of Sch527123, a potent allosteric CXCR1/CXCR2 antagonist. Immunol 2004; 22:329–360. J Pharmacol Exp Ther 2007; 322:477–485. 26 Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from 51 Lai G, Merritt JR, He Z, Feng D, Chao J, Czarniecki MF et al. Synthesis and immunosurveillance to tumor escape. Nat Immunol 2002; 3: 991–998. structure-activity relationships of new disubstituted phenyl-containing 3,4-dia- 27 Becker JC, Andersen MH, Schrama D, Thor Straten P. Immune-suppressive mino-3-cyclobutene-1,2-diones as CXCR2 receptor antagonists. Bioorg Med Chem properties of the tumor microenvironment. Cancer Immunol Immunother 2013; Lett 2008; 18: 1864–1868. 62: 1137–1148. 52 McCleland BW, Davis RS, Palovich MR, Widdowson KL, Werner ML, Burman M 28 Rosenberg SA, Yang JC, Restifo NP. : moving beyond et al. Comparison of N,N'-diarylsquaramides and N,N'-diarylureas as antagonists current vaccines. Nat Med 2004; 10:909–915. of the CXCR2 chemokine receptor. Bioorg Med Chem Lett 2007; 17: 1713–1717. 29 Murdaca G, Spano F, Contatore M, Guastalla A, Penza E, Magnani O et al. 53 Ning Y, Labonte MJ, Zhang W, Bohanes PO, Gerger A, Yang D et al. The CXCR2 Infection risk associated with anti-TNF-alpha agents: a review. Expert opinion on antagonist, SCH-527123, shows antitumor activity and sensitizes cells to oxali- drug safety 2015; 14: 571–582. platin in preclinical colon cancer models. Mol Cancer Ther 2012; 11: 1353–1364. 30 Gogas H, Ioannovich J, Dafni U, Stavropoulou-Giokas C, Frangia K, Tsoutsos D 54 Walters I, Austin C, Austin R, Bonnert R, Cage P, Christie M et al. Evaluation of a et al. Prognostic significance of autoimmunity during treatment of melanoma series of bicyclic CXCR2 antagonists. Bioorg Med Chem Lett 2008; 18:798–803. with interferon. NEngJMed2006; 354: 709–718. 55 Winters MP, Crysler C, Subasinghe N, Ryan D, Leong L, Zhao S et al. Carboxylic 31 Solomon SD, Pfeffer MA, McMurray JJ, Fowler R, Finn P, Levin B et al. Effect of acid bioisosteres acylsulfonamides, acylsulfamides, and sulfonylureas as novel celecoxib on cardiovascular events and blood pressure in two trials for the antagonists of the CXCR2 receptor. Bioorg Med Chem Lett 2008; 18: 1926–1930. prevention of colorectal adenomas. Circulation 2006; 114: 1028–1035. 56 Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat 32 Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-Newen G, Schaper F. Rev Drug Discov 2012; 11:763–776. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. 57 Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the Biochem J 2003; 374:1–20. expanding diversity of effector lineages. Annu Rev Immunol 2007; 25: 33 Jones SA, Richards PJ, Scheller J, Rose-John S. IL-6 transsignaling: the in vivo 821–852. consequences. J Interferon Cytokine Res 2005; 25:241–253. 58 Rachitskaya AV, Hansen AM, Horai R, Li Z, Villasmil R, Luger D et al. Cutting edge: 34 Nowell MA, Williams AS, Carty SA, Scheller J, Hayes AJ, Jones GW et al. Ther- NKT cells constitutively express IL-23 receptor and RORgammat and rapidly apeutic targeting of IL-6 trans signaling counteracts STAT3 control of experi- produce IL-17 upon receptor ligation in an IL-6-independent fashion. J Immunol mental inflammatory arthritis. J Immunol 2009; 182: 613–622. 2008; 180: 5167–5171. 35 Sansone P, Storci G, Tavolari S, Guarnieri T, Giovannini C, Taffurelli M et al. IL-6 59 Do JS, Fink PJ, Li L, Spolski R, Robinson J, Leonard WJ et al. Cutting edge: triggers malignant features in mammospheres from human ductal breast car- spontaneous development of IL-17-producing gamma delta T cells in the thy- cinoma and normal mammary gland. J Clin Invest 2007; 117: 3988–4002. mus occurs via a TGF-beta 1-dependent mechanism. J Immunol 2010; 184: 36 Gao SP, Mark KG, Leslie K, Pao W, Motoi N, Gerald WL et al. Mutations in the 1675–1679. EGFR kinase domain mediate STAT3 activation via IL-6 production in human 60 Ciric B, El-behi M, Cabrera R, Zhang GX, Rostami A. IL-23 drives pathogenic IL-17- lung adenocarcinomas. J Clin Invest 2007; 117: 3846–3856. producing CD8+ T cells. J Immunol 2009; 182: 5296–5305. 37 Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Kloppel G et al. Stat3/ 61 Toy D, Kugler D, Wolfson M, Vanden Bos T, Gurgel J, Derry J et al. Cutting edge: Socs3 activation by IL-6 transsignaling promotes progression of pancreatic interleukin 17 signals through a heteromeric receptor complex. J Immunol 2006; intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell 177:36–39. 2011; 19: 456–469. 62 McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T 38 Becker C, Fantini MC, Schramm C, Lehr HA, Wirtz S, Nikolaev A et al. TGF-beta et al. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and suppresses tumor progression in colon cancer by inhibition of IL-6 trans-- restrain T(H)-17 cell-mediated pathology. Nat Immunol 2007; 8: 1390–1397. signaling. Immunity 2004; 21:491–501. 63 Aggarwal S, Ghilardi N, Xie MH, de Sauvage FJ, Gurney AL. Interleukin-23 pro- 39 Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S et al. IL-6 and motes a distinct CD4 T cell activation state characterized by the production of Stat3 are required for survival of intestinal epithelial cells and development of interleukin-17. J Biol Chem 2003; 278: 1910–1914. colitis-associated cancer. Cancer Cell 2009; 15: 103–113. 64 Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS et al. 40 Nishimoto N, Kanakura Y, Aozasa K, Johkoh T, Nakamura M, Nakano S et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. Humanized anti-interleukin-6 receptor antibody treatment of multicentric Cas- J Biol Chem 2007; 282:9358–9363. tleman disease. Blood 2005; 106: 2627–2632. 65 McAllister F. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 41 Kurzrock R, Voorhees PM, Casper C, Furman RR, Fayad L, Lonial S et al. A phase I, signaling axis in preinvasive pancreatic neoplasia. Cancer Cell 2014; 25: open-label study of siltuximab, an anti-IL-6 monoclonal antibody, in patients 621–637. with B-cell non-Hodgkin lymphoma, multiple myeloma, or Castleman disease. 66 Wagsater D, Lofgren S, Hugander A, Dimberg J. Expression of interleukin-17 in Clin Cancer Res 2013; 19: 3659–3670. human colorectal cancer. Anticancer Res 2006; 26:4213–4216. 42 Zhang Y, Yan W, Collins MA, Bednar F, Rakshit S, Zetter BR et al. Interleukin-6 is 67 Chae WJ, Gibson TF, Zelterman D, Hao L, Henegariu O, Bothwell AL. Ablation of required for pancreatic cancer progression by promoting MAPK signaling acti- IL-17A abrogates progression of spontaneous intestinal tumorigenesis. Proc Natl vation and oxidative stress resistance. Cancer Res 2013; 73: 6359–6374. Acad Sci USA 2010; 107: 5540–5544. 43 Holmes WE, Lee J, Kuang WJ, Rice GC, Wood WI. Structure and functional 68 Hyun YS, Han DS, Lee AR, Eun CS, Youn J, Kim HY. Role of IL-17A in the devel- expression of a human interleukin-8 receptor. Science 1991; 253: 1278–1280. opment of colitis-associated cancer. Carcinogenesis 2012; 33: 931–936. 44 Murphy PM, Tiffany HL. Cloning of complementary DNA encoding a functional 69 Nam JS, Terabe M, Kang MJ, Chae H, Voong N, Yang YA et al. Transforming human interleukin-8 receptor. Science 1991; 253: 1280–1283. growth factor beta subverts the immune system into directly promoting tumor 45 Huang S, Mills L, Mian B, Tellez C, McCarty M, Yang XD et al. Fully humanized growth through interleukin-17. Cancer Res 2008; 68: 3915–3923. neutralizing antibodies to interleukin-8 (ABX-IL8) inhibit angiogenesis, 70 Numasaki M, Fukushi J, Ono M, Narula SK, Zavodny PJ, Kudo T et al. tumor growth, and of human melanoma. Am J Pathol 2002; 161: Interleukin-17 promotes angiogenesis and tumor growth. Blood 2003; 101: 125–134. 2620–2627.

Oncogene (2015) 6029 – 6039 © 2015 Macmillan Publishers Limited Cancer immunoprevention JC Roeser et al 6037 71 Wang L, Yi T, Kortylewski M, Pardoll DM, Zeng D, Yu H. IL-17 can promote tumor 99 Kortylewski M, Xin H, Kujawski M, Lee H, Liu Y, Harris T et al. Regulation of the growth through an IL-6-Stat3 signaling pathway. J Exp Med 2009; 206: IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. 1457–1464. Cancer Cell 2009; 15: 114–123. 72 Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR et al. A human colonic 100 Gu FM, Li QL, Gao Q, Jiang JH, Huang XY, Pan JF et al. Sorafenib inhibits growth commensal promotes colon tumorigenesis via activation of T helper type 17 T and metastasis of hepatocellular carcinoma by blocking STAT3. World J Gastro- cell responses. Nat Med 2009; 15: 1016–1022. enterol 2011; 17: 3922–3932. 73 Muranski P, Boni A, Antony PA, Cassard L, Irvine KR, Kaiser A et al. Tumor-specific 101 Horiguchi A, Asano T, Kuroda K, Sato A, Asakuma J, Ito K et al. STAT3 inhibitor Th17-polarized cells eradicate large established melanoma. Blood 2008; 112: WP1066 as a novel therapeutic agent for renal cell carcinoma. Br J Cancer 2010; 362–373. 102: 1592–1599. 74 Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y et al. Efficacy and 102 Huang C, Yang G, Jiang T, Cao J, Huang KJ, Qiu ZJ. Down-regulation of STAT3 safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in expression by vector-based small interfering RNA inhibits pancreatic patients with psoriasis: 76-week results from a randomised, double-blind, cancer growth. World J Gastroenterol 2011; 17: 2992–3001. placebo-controlled trial (PHOENIX 1). Lancet 2008; 371: 1665–1674. 103 Sen M, Thomas SM, Kim S, Yeh JI, Ferris RL, Johnson JT et al. First-in-human trial 75 Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G et al. Effects of of a STAT3 decoy oligonucleotide in head and neck tumors: implications for AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid cancer therapy. Cancer Discov 2012; 2: 694–705. arthritis, and uveitis. Sci Transl Med 2010; 2: 52ra72. 104 Abdulghani J, Gu L, Dagvadorj A, Lutz J, Leiby B, Bonuccelli G et al. Stat3 pro- 76 Leonardi C, Matheson R, Zachariae C, Cameron G, Li L, Edson-Heredia E et al. motes metastatic progression of prostate cancer. Am J Pathol 2008; 172: Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. 1717–1728. NEngJMed2012; 366: 1190–1199. 105 Fukuda A, Wang SC, JPt Morris, Folias AE, Liou A, Kim GE et al. Stat3 and MMP7 77 Papp KA, Leonardi C, Menter A, Ortonne JP, Krueger JG, Kricorian G et al. Bro- contribute to pancreatic ductal adenocarcinoma initiation and progression. dalumab, an anti-interleukin-17-receptor antibody for psoriasis. NEngJMed Cancer Cell 2011; 19:441–455. 2012; 366: 1181–1189. 106 Kryczek I, Lin Y, Nagarsheth N, Peng D, Zhao L, Zhao E et al. IL-22(+)CD4(+) 78 Hemdan NY. Anti-cancer versus cancer-promoting effects of the interleukin-17- T cells promote colorectal cancer stemness via STAT3 transcription factor producing T helper cells. Immunol Lett 2013; 149:123–133. activation and induction of the methyltransferase DOT1L. Immunity 2014; 40: 79 Martin-Orozco N, Dong C. The IL-17/IL-23 axis of inflammation in cancer: friend 772–784. or foe? Curr Opin Investig Drugs 2009; 10:543–549. 107 Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA et al. Tissue 80 Murugaiyan G, Saha B. Protumor vs antitumor functions of IL-17. J Immunol 2009; expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med 2006; 203: 183: 4169–4175. 883–895. 81 Dinarello CA. Interleukin-1 and interleukin-1 antagonism. Blood 1991; 77:1627–1652. 108 Okazaki T, Honjo T. The PD-L pathway in immunological tolerance. Trends 82 Dinarello CA. Biologic basis for interleukin-1 in disease. Blood 1996; 87:2095–2147. Immunol 2006; 27:195–201. 83 Chen Z, Malhotra PS, Thomas GR, Ondrey FG, Duffey DC, Smith CW et al. 109 Ghebeh H, Mohammed S, Al-Omair A, Qattan A, Lehe C, Al-Qudaihi G et al. The Expression of proinflammatory and proangiogenic cytokines in patients with B7-H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer . Clin Cancer Res 1999; 5: 1369–1379. patients with infiltrating ductal carcinoma: correlation with important high-risk 84 Lewis AM, Varghese S, Xu H, Alexander HR. Interleukin-1 and cancer progression: prognostic factors. Neoplasia 2006; 8: 190–198. the emerging role of interleukin-1 receptor antagonist as a novel therapeutic 110 Harvey RD. Immunologic and clinical effects of targeting PD-1 in lung cancer. agent in cancer treatment. J Transl Med 2006; 4:48. Clin Pharmacol Ther 2014; 96: 214–223. 85 Voronov E, Shouval DS, Krelin Y, Cagnano E, Benharroch D, Iwakura Y et al. IL-1 is 111 Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, Honjo T et al. Tumor cell required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci USA 2003; expression of programmed cell death-1 ligand 1 is a prognostic factor for 100: 2645–2650. malignant melanoma. Cancer 2010; 116: 1757–1766. 86 Portier M, Zhang XG, Ursule E, Lees D, Jourdan M, Bataille R et al. Cytokine gene 112 Nomi T, Sho M, Akahori T, Hamada K, Kubo A, Kanehiro H et al. Clinical sig- expression in human multiple myeloma. Br J Haematol 1993; 85:514–520. nificance and therapeutic potential of the programmed death-1 ligand/pro- 87 Mitsunaga S, Ikeda M, Shimizu S, Ohno I, Furuse J, Inagaki M et al. Serum levels of grammed death-1 pathway in human pancreatic cancer. Clin Cancer Res 2007; IL-6 and IL-1beta can predict the efficacy of gemcitabine in patients with 13: 2151–2157. advanced pancreatic cancer. Br J Cancer 2013; 108: 2063–2069. 113 Wilcox RA, Ansell SM, Lim MS, Zou W, Chen L. The B7 homologues and their 88 Lust JA, Lacy MQ, Zeldenrust SR, Dispenzieri A, Gertz MA, Witzig TE et al. receptors in hematologic malignancies. Eur J Haematol 2012; 88:465–475. Induction of a chronic disease state in patients with smoldering or indolent 114 Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K et al. multiple myeloma by targeting interleukin 1{beta}-induced interleukin 6 Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes production and the myeloma proliferative component. Mayo Clin Proc 2009; 84: are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA 2007; 114–122. 104: 3360–3365. 89 Schindler C, Shuai K, Prezioso VR, Darnell Jr JE. Interferon-dependent tyrosine 115 Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of phosphorylation of a latent cytoplasmic transcription factor. Science 1992; 257: PD-L1 on tumor cells in the escape from host immune system and tumor 809–813. immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA 2002; 99: 90 Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T et al. Molecular cloning 12293–12297. of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor 116 Iwai Y, Terawaki S, Honjo T. PD-1 blockade inhibits hematogenous spread of involved in the gp130-mediated signaling pathway. Cell 1994; 77:63–71. poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int 91 Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C et al. Immunol 2005; 17:133–144. Stat3 as an oncogene. Cell 1999; 98: 295–303. 117 Okudaira K, Hokari R, Tsuzuki Y, Okada Y, Komoto S, Watanabe C et al. Blockade 92 Bar-Natan M, Nelson EA, Xiang M, Frank DA. STAT signaling in the pathogenesis of B7-H1 or B7-DC induces an anti-tumor effect in a mouse pancreatic and treatment of myeloid malignancies. JAKSTAT 2012; 1:55–64. cancer model. Int J Oncol 2009; 35:741–749. 93 Frank DA. STAT3 as a central mediator of neoplastic cellular transformation. 118 Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P et al. Safety and Cancer Lett 2007; 251:199–210. activity of anti-PD-L1 antibody in patients with advanced cancer. NEngJMed 94 Fagard R, Metelev V, Souissi I, Baran-Marszak F. STAT3 inhibitors for cancer 2012; 366:2455–2465. therapy: Have all roads been explored? JAKSTAT 2013; 2: e22882. 119 Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH et al. Phase I 95 Yu CL, Meyer DJ, Campbell GS, Larner AC, Carter-Su C, Schwartz J et al. Enhanced study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid DNA-binding activity of a Stat3-related protein in cells transformed by the Src tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. oncoprotein. Science 1995; 269:81–83. J Clin Oncol 2010; 28: 3167–3175. 96 Chiarle R, Simmons WJ, Cai H, Dhall G, Zamo A, Raz R et al. Stat3 is required for 120 Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF et al. ALK-mediated lymphomagenesis and provides a possible therapeutic target. Nat Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. NEngJ Med 2005; 11:623–629. Med 2012; 366: 2443–2454. 97 Gough DJ, Corlett A, Schlessinger K, Wegrzyn J, Larner AC, Levy DE. Mitochon- 121 Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. drial STAT3 supports Ras-dependent oncogenic transformation. Science 2009; Nat Rev Cancer 2012; 12:252–264. 324: 1713–1716. 122 Blank CU, Enk A. Therapeutic use of anti-CTLA-4 antibodies. Int Immunol 2014; 98 Schlessinger K, Levy DE. Malignant transformation but not normal cell growth 27:3–10. depends on signal transducer and activator of transcription 3. Cancer Res 2005; 123 Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator 65: 5828–5834. of T cell responses. Nat Rev Immunol 2011; 11: 852–863.

© 2015 Macmillan Publishers Limited Oncogene (2015) 6029 – 6039 Cancer immunoprevention JC Roeser et al 6038 124 Pentcheva-Hoang T, Simpson TR, Montalvo-Ortiz W, Allison JP. Cytotoxic T 151 Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of lymphocyte antigen-4 blockade enhances antitumor immunity by stimulating murine helper T cell clone. I Definition according to profiles of lymphokine melanoma-specific T-cell motility. Cancer Immunol Res 2014; 2: 970–980. activities and secreted proteins. J Immunol 1986; 136:2348–2357. 125 Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB et al. 152 Watkins SK, Egilmez NK, Suttles J, Stout RD. IL-12 rapidly alters the functional Improved survival with ipilimumab in patients with metastatic melanoma. NEng profile of tumor-associated and tumor-infiltrating macrophages in vitro and JMed2010; 363:711–723. in vivo. J Immunol 2007; 178: 1357–1362. 126 Ribas A, Kefford R, Marshall MA, Punt CJ, Haanen JB, Marmol M et al. Phase III 153 Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. randomized clinical trial comparing tremelimumab with standard-of-care che- Nat Rev Immunol 2008; 8: 958–969. motherapy in patients with advanced melanoma. J Clin Oncol 2013; 31:616–622. 154 Nakanishi Y, Nakatsuji M, Seno H, Ishizu S, Akitake-Kawano R, Kanda K et al. 127 Tait Wojno ED, Artis D. Innate lymphoid cells: balancing immunity, inflammation, COX-2 inhibition alters the phenotype of tumor-associated macrophages and tissue repair in the intestine. Cell Host Microbe 2012; 12: 445–457. from M2 to M1 in ApcMin/+ mouse polyps. Carcinogenesis 2011; 32: 128 Spits H, Cupedo T. Innate lymphoid cells: emerging insights in development, 1333–1339. lineage relationships, and function. Annu Rev Immunol 2012; 30:647–675. 155 Arber N, Eagle CJ, Spicak J, Racz I, Dite P, Hajer J et al. Celecoxib for the pre- 129 Kirchberger S, Royston DJ, Boulard O, Thornton E, Franchini F, Szabady RL et al. vention of colorectal adenomatous polyps. NEngJMed2006; 355:885–895. Innate lymphoid cells sustain colon cancer through production of interleukin-22 156 Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W et al. in a mouse model. J Exp Med 2013; 210: 917–931. CD40 agonists alter tumor stroma and show efficacy against pancreatic carci- 130 Shields JD, Kourtis IC, Tomei AA, Roberts JM, Swartz MA. Induction of lym- noma in mice and humans. Science 2011; 331:1612–1616. phoidlike stroma and immune escape by tumors that express the 157 Huang B, Lei Z, Zhang GM, Li D, Song C, Li B et al. SCF-mediated mast cell chemokine CCL21. Science 2010; 328:749–752. infiltration and activation exacerbate the inflammation and immunosuppression 131 Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the in tumor microenvironment. Blood 2008; 112:1269–1279. immune system. Nat Rev Immunol 2009; 9: 162–174. 158 Yang Z, Zhang B, Li D, Lv M, Huang C, Shen GX et al. Mast cells mobilize myeloid- 132 Greten TF, Manns MP, Korangy F. Myeloid derived suppressor cells in human derived suppressor cells and Treg cells in tumor microenvironment via IL-17 diseases. Int Immunopharmacol 2011; 11:802–807. pathway in murine hepatocarcinoma model. PLoS One 2010; 5: e8922. 133 Markowitz J, Wesolowski R, Papenfuss T, Brooks TR, Carson WE 3rd. Myeloid- 159 Johansson A, Rudolfsson S, Hammarsten P, Halin S, Pietras K, Jones J et al. Mast derived suppressor cells in breast cancer. Breast Cancer Res Treat 2013; 140:13–21. cells are novel independent prognostic markers in prostate cancer and represent 134 Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S. Cross-talk a target for therapy. Am J Pathol 2010; 177: 1031–1041. between myeloid-derived suppressor cells and macrophages subverts tumor 160 Rabenhorst A, Schlaak M, Heukamp LC, Forster A, Theurich S, von Bergwelt- immunity toward a type 2 response. J Immunol 2007; 179:977–983. Baildon M et al. Mast cells play a protumorigenic role in primary cutaneous 135 Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of lymphoma. Blood 2012; 120:2042–2054. CD8+ T cell response by immature myeloid cells in cancer is mediated by 161 Wasiuk A, Dalton DK, Schpero WL, Stan RV, Conejo-Garcia JR, Noelle RJ. Mast reactive oxygen species. J Immunol 2004; 172:989–999. cells impair the development of protective anti-tumor immunity. Cancer 136 Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase, prostaglandins, and Immunol Immunother 2012; 61: 2273–2282. myeloid-derived suppressor cells in renal cell carcinoma. Clin Cancer Res 2007; 162 Kim YJ, Hong KS, Chung JW, Kim JH, Hahm KB. Prevention of colitis-associated 13: 721s–726s. carcinogenesis with infliximab. Cancer Prev Res (Phila) 2010; 3: 1314–1333. 137 Filipazzi P, Huber V, Rivoltini L. Phenotype, function and clinical implications of 163 Melillo RM, Guarino V, Avilla E, Galdiero MR, Liotti F, Prevete N et al. Mast cells myeloid-derived suppressor cells in cancer patients. Cancer Immunol Immun- have a protumorigenic role in human thyroid cancer. Oncogene 2010; 29: other 2012; 61: 255–263. 6203–6215. 138 Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A 164 Oka T, Kalesnikoff J, Starkl P, Tsai M, Galli SJ. Evidence questioning cromolyn's et al. Identification of discrete tumor-induced myeloid-derived suppressor cell sub- effectiveness and selectivity as a 'mast cell stabilizer' in mice. Lab Invest 2012; 92: populations with distinct T cell-suppressive activity. Blood 2008; 111: 4233–4244. 1472–1482. 139 Mundy-Bosse BL, Young GS, Bauer T, Binkley E, Bloomston M, Bill MA et al. 165 Gresser I. Biologic effects of interferons. J Invest Dermatol 1990; 95:66S–71S. Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and 166 Nanus DM, Pfeffer LM, Bander NH, Bahri S, Albino AP. Antiproliferative and reduced interferon-alpha signaling in CD4(+) T cells from patients with GI antitumor effects of alpha-interferon in renal cell : correlation with malignancy. Cancer Immunol Immunother 2011; 60: 1269–1279. the expression of a kidney-associated differentiation glycoprotein. Cancer Res 140 Najjar YG, Finke JH. Clinical perspectives on targeting of myeloid derived sup- 1990; 50: 4190–4194. pressor cells in the treatment of cancer. Front Oncol 2013; 3:49. 167 Rizza P, Moretti F, Belardelli F. Recent advances on the immunomodulatory 141 Fujimoto S, Greene M, Sehon AH. Immunosuppressor T cells in tumor effects of IFN-alpha: implications for cancer immunotherapy and autoimmunity. bearing host. Immunol Commun 1975; 4: 201–217. Autoimmunity 2010; 43:204–209. 142 Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nature reviews 168 Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons Immunology 2006; 6:295–307. (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol 2005; 23: 143 Darrasse-Jeze G, Podsypanina K. How numbers, nature, and immune status of 307–336. foxp3 regulatory T-cells shape the early immunological events in tumor devel- 169 Jego G, Palucka AK, Blanck JP, Chalouni C, Pascual V, Banchereau J. Plasmacytoid opment. Front Immunol 2013; 4: 292. dendritic cells induce plasma cell differentiation through type I interferon and 144 Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL et al. Quantification of interleukin 6. Immunity 2003; 19: 225–234. regulatory T cells enables the identification of high-risk breast cancer patients 170 Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH. and those at risk of late relapse. J Clin Oncol 2006; 24: 5373–5380. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: 145 Ladoire S, Arnould L, Apetoh L, Coudert B, Martin F, Chauffert B et al. Pathologic the Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 1996; 14: complete response to neoadjuvant of breast carcinoma is asso- 7–17. ciated with the disappearance of tumor-infiltrating foxp3+ regulatory T cells. Clin 171 Sun M, Lughezzani G, Perrotte P, Karakiewicz PI. Treatment of metastatic renal Cancer Res 2008; 14: 2413–2420. cell carcinoma. Nat Rev Urol 2010; 7:327–338. 146 Greten TF, Ormandy LA, Fikuart A, Hochst B, Henschen S, Horning M et al. Low- 172 Kilinc MO, Gu T, Harden JL, Virtuoso LP, Egilmez NK. Central role of tumor- dose cyclophosphamide treatment impairs regulatory T cells and unmasks AFP- associated CD8+ T effector/memory cells in restoring systemic antitumor specific CD4+ T-cell responses in patients with advanced HCC. J Immunother immunity. J Immunol 2009; 182: 4217–4225. 2010; 33: 211–218. 173 Cuff S, Dolton G, Matthews RJ, Gallimore A. Antigen specificity determines the 147 Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E. Tumor pro- or antitumoral nature of CD8+ T cells. J Immunol 2010; 184:607–614. rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) 174 Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH et al. monoclonal antibody. Cancer Res 1999; 59: 3128–3133. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. 148 Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing J Clin Oncol 2011; 29: 1949–1955. CD25+CD4+ T cells: a common basis between tumor immunity and auto- 175 Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel tran- immunity. J Immunol 1999; 163: 5211–5218. scription factor, T-bet, directs Th1 lineage commitment. Cell 2000; 100: 655–669. 149 Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L. The origin and function of 176 Thierfelder WE, van Deursen JM, Yamamoto K, Tripp RA, Sarawar SR, Carson RT tumor-associated macrophages. Immunol Today 1992; 13:265–270. et al. Requirement for Stat4 in interleukin-12-mediated responses of natural killer 150 Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine and T cells. Nature 1996; 382: 171–174. system in diverse forms of macrophage activation and polarization. Trends 177 Lazarevic V, Glimcher LH, Lord GM. T-bet: a bridge between innate and adaptive Immunol 2004; 25: 677–686. immunity. Nat Rev Immunol 2013; 13: 777–789.

Oncogene (2015) 6029 – 6039 © 2015 Macmillan Publishers Limited Cancer immunoprevention JC Roeser et al 6039 178 Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The 184 Tachibana T, Onodera H, Tsuruyama T, Mori A, Nagayama S, Hiai H et al. central role of CD4(+) T cells in the antitumor immune response. J Exp Med 1998; Increased intratumor Valpha24-positive natural killer T cells: a prognostic factor 188: 2357–2368. for primary colorectal carcinomas. Clin Cancer Res 2005; 11:7322–7327. 179 Sharma RK, Yolcu ES, Srivastava AK, Shirwan H. CD4+ T cells play a critical role in 185 Fuji N, Ueda Y, Fujiwara H, Toh T, Yoshimura T, Yamagishi H. Antitumor effect of the generation of primary and memory antitumor immune responses elicited by alpha-galactosylceramide (KRN7000) on spontaneous hepatic metastases SA-4-1BBL and TAA-based vaccines in mouse tumor models. PLoS One 2013; 8: requires endogenous interleukin 12 in the liver. Clin Cancer Res 2000; 6: e73145. 3380–3387. 180 Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A et al. CD4(+) 186 Giaccone G, Punt CJ, Ando Y, Ruijter R, Nishi N, Peters M et al. A phase I study of follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest the natural killer T-cell ligand alpha-galactosylceramide (KRN7000) in patients 8 – 2013; 123: 2873–2892. with solid tumors. Clin Cancer Res 2002; : 3702 3709. 187 Chang YJ, Huang JR, Tsai YC, Hung JT, Wu D, Fujio M et al. Potent immune- 181 Linnemann C, van Buuren MM, Bies L, Verdegaal EM, Schotte R, Calis JJ et al. modulating and anticancer effects of NKT cell stimulatory glycolipids. Proc Natl High-throughput epitope discovery reveals frequent recognition of neo- Acad Sci USA 2007; 104: 10299–10304. antigens by CD4+ T cells in human melanoma. Nat Med 2015; 21:81–85. 188 Vicari AP, Caux C, Trinchieri G. Tumour escape from immune surveillance 182 Kronenberg M. Toward an understanding of NKT cell biology: progress and through dendritic cell inactivation. Semin Cancer Biol 2002; 12:33–42. 23 – paradoxes. Annu Rev Immunol 2005; :877 900. 189 Hegmans JP, Aerts JG. Immunomodulation in cancer. Curr Opin Pharmacol 2014; 183 Molling JW, Langius JA, Langendijk JA, Leemans CR, Bontkes HJ, van der Vliet HJ 17C:17–21. et al. Low levels of circulating invariant natural killer T cells predict poor clinical 190 Zeid NA, Muller HK. S100 positive dendritic cells in human lung tumors asso- outcome in patients with head and neck squamous cell carcinoma. J Clin Oncol ciated with cell differentiation and enhanced survival. Pathology 1993; 25: 2007; 25: 862–868. 338–343.

© 2015 Macmillan Publishers Limited Oncogene (2015) 6029 – 6039