interplay DC cell and tumor

37 36 0
interplay DC cell and tumor

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

interplay beetween DC and tumor,When faced with invading microbes, the immune system must quickly launch an appropriate response to eliminate invaders and restore tissue integrity and homeostasis DCs, described for the first time by Steinman and Cohn (1973), are the master regulators of the immune response dictating tolerance or immunity (Steinman, 2011). Cancer antigens could be presented to T cells by DCs either at tumor sites or in draining lymph nodes (Fig. 1). Cancer antigens, soluble and particulate, are transported to lymph nodes via lymphatic vessels (Steinman, 2011). Soluble antigens are captured by lymph noderesident DCs while tissueresident DCs capture antigen at tumor sites; tissueresident DCs can present antigens either at the tumor site (Chiodoni et al., 1999) or they migrate through lymphatic vessels to present antigen in lymph nodes (Bonaccorsi et al., 2015; Steinman, 2011). DCs display protein antigens in the context of classical MHC class I and MHC class II molecules that allow selection and priming of rare antigenspecific T lymphocytes including CD8+ T cells, CD4+ T cells as introduced above (Trombetta and Mellman, 2005). They can also present lipid antigens in the context of nonclassical CD1 molecules that allow activation of NKT cells (Bendelac et al., 2007). The priming of new T cell repertoires might be critical for clinical success of therapeutic agents aiming to unleash antigenspecific CTLs. The diversity of T cell response is in part ensured by three features of DCs that control their ability to finetune the adaptive immune response: (1) DC maturation (Mellman and Steinman, 2001); (2) DC plasticity in response to environmental cues such as those linked with antigen capture, antigenindependent signals as cytokines and other cells in their environment (Mellman and Steinman, 2001); and (3) The existence of distinct DC subsets with specific functions (Banchereau and Steinman, 1998). Cancer cellderived signals are able to exploit these features, thus having a major impact on DC functionality in the tumor microenvironment (TEM) as discussed in more detail hereunder

ARTICLE IN PRESS Interplay between dendritic cells and cancer cells Jan Martinek, Te-Chia Wu, Diana Cadena, Jacques Banchereau*, Karolina Palucka* The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States *Corresponding authors: e-mail address: jacques.banchereau@jax.org; karolina.palucka@jax.org Contents Introduction Dendritic cells in the immune response to cancer 2.1 Dendritic cell maturation as checkpoint of tolerance and immunity 2.2 Antigen capture and modulation of DC maturation 2.3 Dendritic cell subsets in cancer Dendritic cells dictate the outcome of immune response to cancer 3.1 DCs control anti-tumor immune response 3.2 The central role of type I IFN in tumor rejection 3.3 DCs in pro-tumor immunity and response to treatment 3.4 Chronic inflammation promotes immune escape via DCs Conclusions and future studies Acknowledgments References 12 15 15 16 17 20 22 25 25 Abstract Dendritic cells (DCs) orchestrate a repertoire of immune responses that bring about resistance to infection and tolerance to self Cancers can exploit DCs to evade immunity, but DCs also can generate resistance to cancer Owing to their capacity to capture, process, and present antigens to naïve T cells, thereby launching adaptive immunity, DCs are poised to play a critical role in cancer recognition and rejection As such, DCs represent a solution for the expansion and infiltration of T cells with tumor-rejecting properties Indeed, clinical responses to checkpoint blockade, such as anti-PD-1, are linked to the presence of T cell immunity to cancer-specific antigens However, only a fraction of patients has clinical benefit Unraveling the molecular pathways controlling DC-cancer interplay will therefore pave the way for identifying new targets for therapy that overcome limitations of current treatments and promote long-term cancer control International Review of Cell and Molecular Biology ISSN 1937-6448 https://doi.org/10.1016/bs.ircmb.2019.07.008 # 2019 Elsevier Inc All rights reserved ARTICLE IN PRESS Jan Martinek et al Introduction When faced with invading microbes, the immune system must quickly launch an appropriate response to eliminate invaders and restore tissue integrity and homeostasis Thereafter, the immune response must also rapidly subside to prevent unwanted tissue damage and to restore polyclonal T cell repertoire able to mount a new response to a different microbe (Paul, 2003) Thus, co-stimulatory and co-inhibitory pathways have co-evolved to control the extent of immune responses (Freeman et al., 2000; Krummel and Allison, 1995; Waterhouse et al., 1995) This led to the discovery of checkpoints such as programmed cell death (PD)-1, which control the effector function of T cells (Lesokhin et al., 2015; Sharma and Allison, 2015a) That in turn led to development of checkpoint inhibitors that could unleash T cell function and have already taken a significant place in cancer therapy (Sharma and Allison, 2015b; Topalian et al., 2015) Their clinical effect is at least partly associated with the presence of T cells specific to cancer-derived neo-antigens (Ags) (Le et al., 2017; Matsushita et al., 2012; Schumacher and Schreiber, 2015) Neo-Ags can be generated by a variety of means including genetic alterations in cancer cells (high mutation load, microsatellite instability and gene fusions) as well as epigenetic ( Jones et al., 2019) and post-translational regulation (Zarling et al., 2006) However, with a notable exception of malignant melanoma and Hodgkin’s disease, only a fraction of patients ($15%) responds clinically to this treatment modality (Haslam and Prasad, 2019) Treatment resistance (reviewed in Sharma et al., 2017) might be due in part to the low frequency of T cells specific to cancer neo-Ags, so-called “cold” tumors Conceptualization of the cancer-immunity cycle created a framework for the identification of barriers to effective cancer rejection (Chen and Mellman, 2013) and facilitated investigations into how to turn “cold” tumors with diminished T cell infiltrate into “hot” tumors Among others, provision of cancer Ag-specific T cells either via adoptive transfer (Fesnak et al., 2016; Rosenberg et al., 2008) or via their expansion in vivo (Palucka and Banchereau, 2014) represent ways of correcting the deficiency of T cell specific to cancer Ags Numerous T cell subsets contribute to and regulate the host response to cancer including: (1) CD4+ T cells with helper function, which is essential for establishing CD8+ T cell memory (Zhu and Paul, 2008) as well as generation of antibody response (Zhu and Paul, 2008); (2) CD4+ T cells with regulatory/suppressor function (Tregs), which represent a healthy homeostatic ARTICLE IN PRESS Interplay between dendritic cells and cancer cells mechanism but are amplified in cancer via numerous pathways to facilitate immune escape (Zhu and Paul, 2008); and (3) CD8+ T cells that can give rise to cytotoxic T lymphocytes (CTLs) able to reject tumors (Boon et al., 1994) Several cell types of the innate and adaptive immune system can contribute to the final fate of cancer cells and their rejection including innate effectors such as NK cells, neutrophils, and eosinophils, and adaptive effectors such as NKT cells, CD4+ T cells and antibodies (Abbas and Lichtman, 2003; Palucka and Coussens, 2016a) However, we will focus discussion on antigen-specific CD8+ T cells Desired criteria for anti-cancer CD8+ T cells include: (1) high T cell receptor (TCR) affinity (binding) and avidity (off-rate) for peptide major histocompatibility complexes (MHCs) expressed on cancer cells (Appay et al., 2008); (2) T cell trafficking into the tumor (e.g., expression of CXCR3) (Mullins et al., 2004) and persistence in the tumor site (e.g., CD103 (Le Floc’h et al., 2007) and CD49a (Sandoval et al., 2013)); (3) high expression of costimulatory molecules (e.g., CD137 (Wilcox et al., 2002)) or low expression of inhibitory molecules (e.g., PD-1 (Freeman et al., 2000)); and (4) high expression of effector molecules such as granzyme and perforin by T cells (Appay et al., 2008) Cancer cells cannot prime T cells by themselves due to usually low expression of MHC molecules and of costimulatory molecules and a high expression of inhibitory molecules and suppressive cytokines (Moussion and Mellman, 2018) Macrophages, despite being the most abundant myeloid cells in tumors, contribute minimally to priming of antigen-specific T cells because they are molecularly wired for tissue repair and antigen degradation rather than for antigen presentation (Ruffell and Coussens, 2015) In contrast, DCs have the remarkable capacity to capture antigens from their environment, migrate to draining lymph nodes and cross-present captured antigens on MHC class I for priming of CD8+ T cells and MHC class II for priming CD4+ T cells (Banchereau and Steinman, 1998; Steinman and Banchereau, 2007) Therefore, DCs are critical for generation of cancer antigen-specific T cells DCs are molecularly equipped to simultaneously deliver signals necessary for induction and expansion of antigen-specific T cells as they are able to: (1) present the cancer antigen peptides to both CD8+ and CD4+ T cells (cognate help); (2) deliver co-stimulatory signals to T cells via CD80, CD70 and 4-1BB, supporting T cell activation; and (3) deliver cytokine signals including interleukin (IL)-12, type I interferon (IFN) and IL-15 thereby supporting T cell expansion and polarization leading to secretion of type cytokines such as type II IFN (IFN-γ) (Banchereau and Steinman, 1998; Steinman and Banchereau, 2007) Here, we will review ARTICLE IN PRESS Jan Martinek et al the basics of DC biology in the context of their interactions with cancer cells, particularly how cancers hypnotize DCs to escape immune control by tilting the balance toward other myeloid cells (such as macrophages (Palucka and Coussens, 2016a) and myeloid derived suppressor cells (Marigo et al., 2008; Veglia et al., 2019)) to manipulate ensuing T cell responses and to escape immune control We will also discuss how the understanding of these interactions at the cellular and molecular level might offer novel therapeutic targets Dendritic cells in the immune response to cancer DCs, described for the first time by Steinman and Cohn (1973), are the master regulators of the immune response dictating tolerance or immunity (Steinman, 2011) Cancer antigens could be presented to T cells by DCs either at tumor sites or in draining lymph nodes (Fig 1) Cancer antigens, soluble and particulate, are transported to lymph nodes via lymphatic vessels (Steinman, 2011) Soluble antigens are captured by lymph node-resident DCs while tissue-resident DCs capture antigen at tumor sites; tissue-resident DCs can present antigens either at the tumor site (Chiodoni et al., 1999) or they migrate through lymphatic vessels to present antigen in lymph nodes (Bonaccorsi et al., 2015; Steinman, 2011) DCs display protein antigens in the context of classical MHC class I and MHC class II molecules that allow selection and priming of rare antigen-specific T lymphocytes including CD8+ T cells, CD4+ T cells as introduced above (Trombetta and Mellman, 2005) They can also present lipid antigens in the context of non-classical CD1 molecules that allow activation of NKT cells (Bendelac et al., 2007) The priming of new T cell repertoires might be critical for clinical success of therapeutic agents aiming to unleash antigenspecific CTLs The diversity of T cell response is in part ensured by three features of DCs that control their ability to fine-tune the adaptive immune response: (1) DC maturation (Mellman and Steinman, 2001); (2) DC plasticity in response to environmental cues such as those linked with antigen capture, antigen-independent signals as cytokines and other cells in their environment (Mellman and Steinman, 2001); and (3) The existence of distinct DC subsets with specific functions (Banchereau and Steinman, 1998) Cancer cell-derived signals are able to exploit these features, thus having a major impact on DC functionality in the tumor microenvironment (TEM) as discussed in more detail hereunder ARTICLE IN PRESS Interplay between dendritic cells and cancer cells Fig Dendritic cells control immune response to cancer The immune system is endowed with the ability to recognize a universe of diverse molecules called antigens, including cancer antigens, and to generate responses specific to the recognized antigens Lymphocytes (T, B, NK, and NKT cells) and their products are under the control of DCs DCs reside in peripheral tissues where they are poised to capture antigens Antigen-loaded migratory DCs travel from tissues through the afferent lymphatics into the draining lymph nodes There, they present processed protein and lipid Ags to T cells via both classical (MHC class I and class II) and non-classical (CD1 family) antigen presenting molecules The soluble antigens also reach the draining lymph nodes through lymphatics and conduits where they are captured, processed, and presented by lymph-node resident DCs Ag presentation by non-activated (immature) DCs leads to tolerance and/or development of Tregs Activated (mature), antigen-loaded DCs are geared towards the launching of antigen-specific immunity leading to the T cell proliferation and differentiation into helper and effector cells with unique functions and cytokine profiles DCs are also important in launching humoral immunity Thus, DCs are at the center of anti-cancer immunity 2.1 Dendritic cell maturation as checkpoint of tolerance and immunity One of DC vulnerabilities in the context of cancer is the direct link between their maturation and function as measured by the induction of T cell response (Steinman, 2011) To this end, in the steady state, non-activated (immature) DCs present antigens (including self-antigens) to T cells, thereby inducing tolerance either through T cell deletion or through differentiation of regulatory/suppressor T cells (Fig 1) These immature DCs can be ARTICLE IN PRESS Jan Martinek et al considered “immunological sensors,” alert for potentially dangerous microbes but also for the alterations of tissue homeostasis and sterile inflammation, and are capable of decoding and integrating such signals ( Janeway Jr and Medzhitov, 2002; Kawai and Akira, 2006; Pulendran et al., 2001) Immature DCs have special characteristics including: (1) expression of a specific set of damage sensing pathways (Banchereau et al., 2000; Steinman, 2011); (2) ability to efficiently capture Ags (Banchereau et al., 2000); (3) accumulation of MHC class II molecules in the late endosome-lysosomal compartment enabling loading of the peptide and assembly of peptide-MHC complexes that can then be transferred to cell surface (Steinman, 2011); and (4) low expression of costimulatory molecules (Steinman, 2011) These properties can be harnessed by cancers to generate Tregs rather than T cells able to reject tumors (Fehervari and Sakaguchi, 2004; Idoyaga et al., 2013; Melief, 2008; Tanchot et al., 2012; Yamazaki et al., 2006) For example, in a mouse model of melanoma the mere increase of DC infiltrate by their mobilization with FLT3L was not sufficient for tumor rejection even in the presence of PD-1 blockade (Salmon et al., 2016a) Yet, addition of DC activator such as TLR-3 ligand poly IC facilitated tumor regression (Salmon et al., 2016a) Mature Ag-loaded DCs can launch differentiation of Ag-specific T cells into effector cells (reviewed in Banchereau et al., 2000) (Fig 1) DC maturation is associated with: (1) down-regulation of Ag-capture activity (Trombetta and Mellman, 2005); (2) surface expression of CCR7 enabling migration of DCs into draining lymph nodes (Dieu-Nosjean et al., 1999; Forster et al., 1999); (3) translocation of peptide-MHC (pMHC) complexes to cell surface together with co-stimulatory molecules (Lanzavecchia and Sallusto, 2001); and (4) ability to secrete cytokines such as IL-12 (Veglia et al., 2019) and IL-15 (Waldmann and Tagaya, 1999) supporting T cell differentiation The ligation of the co-stimulatory receptor CD40 is an essential signal for the final differentiation into fully mature DCs (Banchereau et al., 1994) However, DC maturation alone does not result in a unique DC phenotype Instead, the different signals that are provided either directly or through the surrounding cells that respond to damage induce DCs to acquire distinct phenotypes that eventually contribute to different immune responses (Pulendran et al., 2001) (Fig 2) For example, γδ-T cells and NK cells release IFN-γ, mast cells release pre-formed IL-4 and tumor necrosis factor (TNF), plasmacytoid (p)DCs secrete IFN-α, stromal cells secrete IL-15 and thymic stromal lymphopoietin (TSLP) while neutrophils provide immunostimulatory DNA This plasticity in response to external signals ARTICLE IN PRESS Interplay between dendritic cells and cancer cells Cancer cell Dendritic cell Stromal cells NK cells Neutrophils pDCs Mast cells MDSCs Macrophages T regs CD8 T cells CD4 T cells Th2s Fig Interplay of dendritic cells and cancer: contribution of other cells DCs represent the link between the innate and adaptive immunity and as such they integrate signals from surrounding cells as well as are engaged in cross-talk with stromal cells other leukocytes and cancer cells Cancer cells and dendritic cells can impact each other indirectly by modulating intermediate cells, which is represented by the arrows in the figure This is driven by a variety of cell types, multiple surface bound and soluble factors too numerous to be represented here but several examples includes: secretion of stem cell factor (SCF) by numerous mouse and human cancer cell lines supported c-KIT expressing mast cells, which in turn secreted multiple cytokines such as IL-6, TNF-a VEGF, iNOS and CCL2, inducing tumor remodeling and altering DCs maturation/activation in TEM (Huang et al., 2008) Another example comes from TSLP production by cancer cells and stromal cells in breast and pancreatic cancers (De Monte et al., 2011; Kuan and Ziegler, 2018; Olkhanud et al., 2011b; Pedroza-Gonzalez et al., 2011b) TSLP drives DC maturation leading to expression among others of OX40-L, which enables priming of IL-4/IL-13 secreting Th2 T cells In turn, IL-4 and IL-13 modulate TEM by promoting development of suppressive macrophages producing EGF that supports cancer cell growth as well as directly impacting cancer cells by inhibition of apoptosis ITL7/BST2 mediated interaction between pDCs and cancer cells will suppress IFN-α and TNF-a production in pDCs (Cao et al., 2009), while cancer derived PGE2 and TGF-β synergistically leads to production of IL-6/IL-8 by pDCs (Bekeredjian-Ding et al., 2009) This in turn will have broad impact on both the innate (monocyte differentiation to macrophages and attraction of neutrophils) and adaptive (up regulation of OX40-L and ICOS-L on pDC upon maturation, resulting in T regulatory and Th2 T cells activation) immunity in TEM Last but not least, by secreting IL-12, DCs can directly support CD8+ CTL and NK cells (Mittal et al., 2017) In turn CD8+ T cells and NK cells will secrete IFN-y, which will stimulate CXCL9/10 production from DCs resulting in an influx of effector T cells to the tumor environment (Mikucki et al., 2015) Additionally, NK cells can produce FLT3L, a growth factor promoting pre-DCs differentiation and DCs survival (Barry et al., 2018) ARTICLE IN PRESS Jan Martinek et al represents another vulnerability that can be exploited by cancers to escape immune-mediated elimination Whereas some of the mechanisms dictating DC maturation are differentially regulated in distinct DC subsets, many of the principles are shared 2.2 Antigen capture and modulation of DC maturation DCs are scarce in tumor tissues when compared to other myeloid cells such as macrophages and must therefore compete for both Ag capture and T cell access Ag capture is a critical step controlling the acquisition and subsequent presentation of cancer Ags (Durand and Segura, 2015; Mellman and Steinman, 2001; Trombetta and Mellman, 2005) and is often linked with modulation of DC maturation (Fig 3) Acquisition of cancer Ags can be mediated via several pathways including phagocytosis (Guermonprez and Amigorena, 2005); receptor mediated endocytosis (as, for example, with the lectin Clec9A (Schreibelt et al., 2012)); capture of IgG-Ags immune complexes (Liu et al., 2006; Nimmerjahn and Ravetch, 2006; Rovere et al., 1998); pinocytosis enabling capture of soluble molecules (de Baey and Lanzavecchia, 2000); nibbling enabling capture of cell membrane fragments from live cells (Harshyne et al., 2003); capture of extracellular vesicles (Muller et al., 2016; Wolfers et al., 2001a); and capture of pre-formed peptides (cross-dressing) (Wakim and Bevan, 2011) Hereunder, we will expand on some of these mechanisms to illustrate the potential vulnerabilities that can be exploited by cancer cells to escape immune elimination 2.2.1 Phagocytosis Phagocytosis is an active process of ingestion of particulate Ags that is essential for: (1) the clearance of apoptotic bodies from dying cells; and (2) for the efficient uptake of pathogens and Ags from dying infected and/or cancer cells (Aderem and Underhill, 1999; Platt et al., 1998) When captured by macrophages, apoptotic bodies are degraded ( Jutras and Desjardins, 2005) However, when captured by DCs, their antigenic material can be cross-presented to T cells to elicit Ag-specific CD4+ (Inaba et al., 1998) and CD8+ T cell responses (Albert et al., 1998a; Berard et al., 2000a) Studies pioneered by Zitvogel and Kroemer labs demonstrated the links between phagocytosis and so-called immunogenic cell death in response to chemotherapy (Obeid et al., 2007) The critical mechanism involves the recognition of calreticulin translocated to the surface of apoptotic bodies from cancer cells and its availability for recognition by receptors on DCs ARTICLE IN PRESS Interplay between dendritic cells and cancer cells IL-10, CCL4, CCL2, CXCL1, CXCL5, PGE2, ATP, Arginase, TSLP,TGF-β, HMGB1 TIM-3 Phosphatidylserine TGF- R Activated TGFIntegrins E-cadherin EpCAM CD44 LFA-1 ICAM-1 Fc Receptor IL-1β, IL-6, IL-8, IL-15, IL-10, IFN-α, TNF-α DNA MHC-I Vesicles/ Exosomes RNA MHC-II Cancer antigen Dexosome Apoptotic bodies Endocytosis Fusion STING RIG-1 TLRs Fc Receptors CR1/CR2/CR3 SIRP DEC205, CD207, CD206, CD209, DCIR, AXL, TIM-3 Phagocytosis Fig Interplay of dendritic cells and cancer: direct interactions (A) Interactions between DCs and intact cancer cells driven by secreted and surface molecules: Here we illustrate with few examples surface bound receptors and ligands, as well as also secreted chemokine, cytokines, and metabolites involved in the interplay between DCs and intact cancer cells In addition to molecular pathways discussed in the text and other legends, cancer cells can secrete CCL2 and CCL20, attracting CCR2+ tumor promoting monocytes and macrophages but also tolerogenic immature DCs (Nagarsheth et al., 2017) IL-6 and IL-8 (CXCL8) secreted by DCs have been shown to support tumor growth, survival and invasiveness in multiple types of cancer (Ara et al., 2009; Araki et al., 2007; Yao et al., 2007) Through the secretion of IL-10, TGF-β and other factors, cancer cells induce T cell Ig and mucin domain (TIM-3) up regulation (Continued) ARTICLE IN PRESS 10 Jan Martinek et al (Obeid et al., 2007) This discovery showed that cancer cell apoptosis could efficiently activate an immune response if the correct combination of “eatme” signals is present on the surface of dying cells However, the putative DC calreticulin receptors that are important for the sensing of immunogenic cell death remain to be uncovered (Martins et al., 2010) Another important mechanism of cancer cell phagocytosis is mediated by CD47-SIRPα interactions (Chao et al., 2010; Jaiswal et al., 2009; Majeti et al., 2009) and has been recently shown to contribute to immune escape by delivering a “don’t’-eat-me” signal Indeed, CD47, a "don’t eat me" signal for phagocytic cells including DCs, is overexpressed on cancer cells as compared to matched adjacent normal (nontumor) tissue (Chao et al., 2010; Jaiswal et al., 2009; Majeti et al., 2009) In vitro, blockade of CD47 signaling using monoclonal antibodies enabled phagocytosis of cancer cells that were otherwise protected (Willingham et al., 2012) Furthermore, this pathway plays a role in DC maturation as SIRP-α engagement by CD47-Fc leads to immature DC phenotype, decreased cytokine production, and low IFN-γ production by T cells after priming linked with an impaired development of a T helper (Th)1 response (Hagnerud et al., 2006; Latour et al., 2001) The CD47-SIRPα axis appears to also dictate the fate of captured DNA as blocking the interaction of SIRPα with CD47 preferentially increased the sensing of captured DNA in DCs but not in macrophages (Xu et al., 2017) Fig 3—Cont’d by DCs TIM-3 senses danger signals such as tumor derived nucleic acid it can also sense phosphatidylserine (PS) PS serves as an “eat me” signal and is exported to the outer membrane layer under oxidative stress to facilitate phagocytosis (Birge et al., 2016) Cancer derived HMGB1 can interact with TIM-3 and inhibit its function (Chiba et al., 2012) ICs can be internalized by DCs and deliver Ags for presentation to T cells (Amigorena et al., 1992; Geissmann et al., 2001) (B) Release and capture of cancer antigen Here we illustrate how intact and dying cancer cells release cancer antigens for capture by DCs We show examples related to extracellular vesicles and apoptotic bodies derived from cancer cells and pathways through which DCs can capture them Cancer cells can produce extracellular vesicles by “blebbing” of the cell membrane and encapsulating parts of the cytosol or by formation of exosomes from the multivesicular endosome These vesicles contain genetic material, cancer antigens as well as immunosuppressive proteins such as PD-L1 and Fas-L (Bobrie and Thery, 2013; Sansone et al., 2017; Schuler et al., 2014; Wolfers et al., 2001b) Nucleic acids, contained within extracellular vesicles and apoptotic bodies can trigger intracellular danger associated receptors/pathways such as TLRs, STING and RIG-I in DCs ICAM-1 positive vesicles can be endocytosed after binding to DC surface via LFA-1 interaction (Chiba et al., 2012), they can also fuse with DCs membrane via fusion molecules such as flotillin or GTPases (Subra et al., 2010) ARTICLE IN PRESS 23 Interplay between dendritic cells and cancer cells A B C D E F Fig DCs role in the Yin and Yang circle of cancer immune response The Yang side of the tumor immune response: Images illustrate different stages of anti-cancer immune response leading to cancer rejection (A) the capture of cancer antigen by tumor infiltrating DCs (Cytokeratin (CK) in green and CD11c+ DCs in red) (B) Activated DCs provide co-stimulatory signal to T cells (CD3+ T cells (green) are in close contact with CD11c+ (not shown) DCs expressing the co-stimulatory molecule CD86 (red)) This process can take place at the draining lymph node or, as shown here, at the tumor site with cancer cells stained for CK (blue) (C) after Ag presentation along with adequate co-stimulation, Ag-specific T cells will kill cancer cells via the action of granzymes and perforin (cytotoxic CD3+ (not shown) CD8+ (red) T cell, killing a target cancer cell (blue) by perforin (green) secretion The Yin side of the tumor immune response: Images depicting how cancer cells can corrupt DCs, reprogramming them into launching an immune response that will support cancer growth and progression (D) Cancer cells can produce factors that will corrupt DC maturation (CK+ cancer cells (blue) present surface bound TGF-β (red) to DCs, which induces them to secrete IL-1β (green)) (E) Corrupted DCs, will secrete factors with an impact on the whole TME This is shown in E where CD11c+ DCs (green) produce and secrete IL-1β in proximity but also directly onto CK+ cancer cells (blue) (F) Corrupted immune system will then support cancer progression (CK+ cancer cells (blue), based on their KI67 staining (green), are highly proliferating despite being in close proximity with tumor infiltrating CD11c+ DCs (red)) the steady state and upon cancer challenge Thus, progress will come from basic studies and deep analysis of patient tissues linked with causative studies in pre-clinical models Next generation immunotherapies will be based on cycles of interventions designed to boost and modulate ARTICLE IN PRESS 24 Jan Martinek et al anti-cancer immunity Eventually all patients will be treated with checkpoint inhibitors, either directly or after interventions targeting inflammation, by vaccination to boost T cell repertoires, or by adoptive T cell transfer The majority of patients will subsequently develop acquired resistance followed by immune escape; this will lead to the next cycle of treatments incorporating multi-modal biomarkers Despite rapid progress in the field, much remains to be discovered and defined in terms of biomarkers The cancer-immunity cycle represents a framework enabling uncovering of mechanisms operative at each step We must fully understand the rules of T cell priming in vivo in humans and develop strategies for directing T cells to tumors Last but not least, the role of Tregs, so well established in murine cancer, will need to be redefined in humans Recent studies place one DC subset, cDC1, at the center of regulation of cancer immunity How then other DC subsets contribute to and modulate anti-cancer immunity and by what mechanisms? Are the mechanisms regulating DC-T cell interactions at the tumor shared with those operating in the draining lymph node? The studies on modulation of DCs in lymph nodes draining tumors have only begun (Binnewies et al., 2019) and this line of investigation is likely to enhance our understanding of how the new T cell repertoire can be primed in the context of cancer environment Metabolic regulation of DCs creates another layer of control by the TEM that will need to be explored (Sinclair et al., 2017; Wculek et al., 2019) Another important question is how the differences in the phenotype of human DC subsets between individuals and tissues (Alcantara-Hernandez et al., 2017) impact the launching of anticancer immunity and response to check point inhibitors By analogy to its role in autoimmune diseases, host genetic variation is likely to have a significant contribution to DC-cancer interactions (Hafler and Jager, 2005; Ye et al., 2014) Genome-wide association studies (GWAS) have identified more than three hundred susceptibility loci predisposed to the development of autoimmune diseases These studies of patients affected by severe autoimmune or immunodeficiency syndromes have led to the discovery of several causative variants (Gutierrez-Arcelus et al., 2016) Polymorphisms of Human Leukocyte Antigen (HLA) molecules have been associated with development of virally-induced tumors such as head and neck, cervical, and nasopharyngeal cancers (Brodin et al., 2015; Brodin and Davis, 2017; Mangino et al., 2017) Resolving all this will keep busy for a while! ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 25 Acknowledgments We thank patients and healthy donors for participation in our studies; our current and former lab members and collaborators; Dr Taneli Helenius for editing the manuscript, the JAX creative services and the Imaging sciences services at the Jackson Laboratory for expert assistance with this publication Due to space limitations we could cite only selected papers Supported by The Jackson Laboratory; R01 CA219880 (KP); U01 AI124297 (JB); and P30CA034196 (Research reported in this publication was partially supported by the National Cancer Institute of the National Institutes of Health under Award Number P30CA034196 The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health) References Abbas, A.K., Lichtman, A.H., 2003 Cellular and molecular immunology, fifth ed Saunders, Philadelphia, p 562 Aderem, A., Underhill, D.M., 1999 Mechanisms of phagocytosis in macrophages Annu Rev Immunol 17, 593–623 Albert, M.L., Sauter, B., Bhardwaj, N., 1998a Dendritic cells acquire antigen from apoptotic cells and induce class I- restricted CTLs Nature 392 (6671), 86–89 Albert, M.L., Darnell, J.C., Bender, A., Francisco, L.M., Bhardwaj, N., Darnell, R.B., 1998b Tumor-specific killer cells in paraneoplastic cerebellar degeneration Nat Med 4, 1321–1324 Alcantara-Hernandez, M., et al., 2017 High-dimensional phenotypic mapping of human dendritic cells reveals interindividual variation and tissue specialization Immunity 47 (6), 1037–1050 e6 Amigorena, S., et al., 1992 Tyrosine-containing motif that transduces cell activation signals also determines internalization and antigen presentation via type III receptors for IgG Nature 358 (6384), 337–341 Appay, V., Douek, D.C., Price, D.A., 2008 CD8+ T cell efficacy in vaccination and disease Nat Med 14 (6), 623–628 Ara, T., et al., 2009 Interleukin-6 in the bone marrow microenvironment promotes the growth and survival of neuroblastoma cells Cancer Res 69 (1), 329–337 Araki, S., et al., 2007 Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer Cancer Res 67 (14), 6854–6862 Aspord, C., Pedroza-Gonalez, A., Gallegos, M., Tindle, S., Burton, E.C., Su, D., Marches, F., Banchereau, J., Palucka, A.K., 2007 Breast cancer instructs dendritic cells to prime interleukin 13-secreting CD4 + T cells that facilitate tumor development J Exp Med 204 (5), 1037–1047 Bajan˜a, S., Turner, S., Paul, J., Ainsua-Enrich, E., Kovats, S., 2016 IRF4 and IRF8 Act in CD11c+ cells to regulate terminal differentiation of lung tissue dendritic cells J Immunol 196 (4), 1666–1677 Baker, K., Qiao, S.-W., Kuo, T.T., Aveson, V.G., Platzer, B., Andersen, J.-T., Sandlie, I., et al., 2011 Neonatal Fc receptor for IgG (FcRn) regulates cross-presentation of IgG immune complexes by CD8 ÀCD11b + dendritic Cells Proc Natl Acad Sci U S A 108 (24), 9927–9932 Baker, K., Rath, T., Flak, M.B., Arthur, J.C., Chen, Z., Glickman, J.N., Zlobec, I., et al., 2013 Neonatal Fc receptor expression in dendritic cells mediates protective immunity against colorectal cancer Immunity 39 (6), 1095–1107 Banchereau, J., Steinman, R.M., 1998 Dendritic cells and the control of immunity Nature 392 (6673), 245–252 ARTICLE IN PRESS 26 Jan Martinek et al Banchereau, J., et al., 1994 The CD40 antigen and its ligand Annu Rev Immunol 12, 881–922 Banchereau, J., et al., 2000 Immunobiology of dendritic cells Annu Rev Immunol 18, 767–811 Banerjee, D., Mathews, P., Matayeva, E., Kaufman, J.L., Steinman, R.M., Dhodapkar, K.M., 2008 Enhanced T-cell responses to glioma cells coated with the anti-EGF receptor antibody and targeted to activating FcgammaRs on human dendritic cells J Immunother 31, 113–120 Baran, J., Allendorf, D.J., Hong, F., Ross, G.D., 2007 Oral beta-glucan adjuvant therapy converts nonprotective Th2 response to protective Th1 cell-mediated immune response in mammary tumor-bearing mice Folia Histochem Cytobiol 45 (2), 107–114 Barry, K.C., et al., 2018 A natural killer-dendritic cell axis defines checkpoint therapyresponsive tumor microenvironments Nat Med 24 (8), 1178–1191 Becker, A., Thakur, B.K., Weiss, J.M., Kim, H.S., Peinado, H., Lyden, D., 2016 Extracellular vesicles in cancer: cell-to-cell mediators of metastasis Cancer Cell 30 (6), 836–848 Bekeredjian-Ding, I., et al., 2009 Tumour-derived prostaglandin E and transforming growth factor-beta synergize to inhibit plasmacytoid dendritic cell-derived interferon-alpha Immunology 128 (3), 439–450 Bell, D., Chromarat, P., Broyles, D., Netto, G., Harb, G.M., Lebecque, S., Valladeau, J., Davoust, J., Palucka, K.A., Banchereau, J., 1999 In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas J Exp Med 190, 1417–1426 Benci, J.L., Xu, B., Qiu, Y., Wu, T.J., Dada, H., Twyman-Saint Victor, C., Cucolo, L., DSM, L., Pauken, K.E., Huang, A.C., Gangadhar, T.C., Amaravadi, R.K., Schuchter, L.M., Feldman, M.D., Ishwaran, H., Vonderheide, R.H., Maity, A., Wherry, E.J., Minn, A.J., 2016 Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade Cell 167 (6), 1540–1554 e12 Bendelac, A., Savage, P.B., Teyton, L., 2007 The biology of NKT cells Annu Rev Immunol 25, 297–336 Berard, F., et al., 2000a Cross-priming of naive CD8 T cells against melanoma antigens using dendritic cells loaded with killed allogeneic melanoma cells J Exp Med 192, 1535–1544 Berard, F., Blanco, P., Davoust, J., Neidhart-Berard, E.-M., Nouri-Shirazi, M., Taquet, N., Rimoldi, D., Cerottini, J.C., Banchereau, J., Palucka, A.K., 2000b Cross-priming of naive cd8 T cells against melanoma antigens using dendritic cells loaded with killed allogeneic melanoma cells J Exp Med 192 (11), 1535–1544 Binnewies, M., et al., 2019 Unleashing type-2 dendritic cells to drive protective antitumor CD4(+) T cell immunity Cell 177 (3), 556–571 e16 Birge, R.B., et al., 2016 Phosphatidylserine is a global immunosuppressive signal in efferocytosis, infectious disease, and cancer Cell Death Differ 23 (6), 962–978 Blanco, P., et al., 2001 Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus Science 294 (5546), 1540–1543 Bobrie, A., Thery, C., 2013 Exosomes and communication between tumours and the immune system: are all exosomes equal? Biochem Soc Trans 41 (1), 263–267 Bonaccorsi, I., et al., 2015 Acquisition and presentation of tumor antigens by dendritic cells Crit Rev Immunol 35 (5), 349–364 Boon, T., et al., 1994 Tumor antigens recognized by T lymphocytes Annu Rev Immunol 12, 337–365 B€ ottcher, J.P., Bonavita, E., Chakravarty, P., Blees, H., Cabeza-Cabrerizo, M., Sammicheli, S., Rogers, N.C., Sahai, E., Zelenay, S., Reis e Sousa, C., 2018 NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control Cell 172 (5), 1022–1037 e14 ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 27 Bournazos, S., Wang, T.T., Ravetch, J.V., 2016 The role and function of Fcgamma receptors on myeloid cells Microbiol Spectr (6) Brisen˜o, C.G., Haldar, M., Kretzer, N.M., Wu, X., Theisen, D.J., Kc, W., Durai, V., Grajales-Reyes, G.E., Iwata, A., Bagadia, P., Murphy, T.L., Murphy, K.M., 2016 Distinct transcriptional programs control cross-priming in classical and monocytederived dendritic cells Cell Rep 15 (11), 2462–2474 Brodin, P., Davis, M.M., 2017 Human immune system variation Nat Rev Immunol 17 (1), 21–29 Brodin, P., et al., 2015 Variation in the human immune system is largely driven by non-heritable influences Cell 160 (1-2), 37–47 Broz, M., Binniwies, M., Boldajipour, B., Nelson, A., Pollock, J., Erle, D., Barczak, A., Rosenblum, M., Daud, A., Barber, D., Amigorena, S., van’t Veer, L.J., Sperling, A., Wolf, D., Krummel, M.F., 2014a Dissecting the tumor myeloid compartment reveals rare activating antigen presenting cells, critical for T cell immunity Cancer Cell 26 (5), 638–652 Broz, M.L., Binnewies, M., Boldajipour, B., Nelson, A.E., Pollack, J.L., Erle, D.J., Barczak, A., Rosenblum, M.D., Daud, A., Barber, D.L., Amigorena, S., van’tVeer, L.J., Sperling, A.I., Wolf, D.M., Krummel, M.F., 2014b Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity Cancer Cell 26, 638–652 Bruhns, P., J€ onsson, F., 2015 Mouse and human FcR effector functions Immunol Rev 268 (1), 25–51 Burnet, M., 1957 Cancer; a biological approach I The processes of control Br Med J (5022), 779–786 Cao, W., et al., 2009 Regulation of TLR7/9 responses in plasmacytoid dendritic cells by BST2 and ILT7 receptor interaction J Exp Med 206 (7), 1603–1614 Caux, C., et al., 1992 GM-CSF and TNF-alpha cooperate in the generation of dendritic Langerhans cells Nature 360 (6401), 258–261 Caux, C., et al., 1996 CD34+ hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to GM-CSF +TNF alpha J Exp Med 184 (2), 695–706 Caux, C., Massacrier, C., Vanbervliet, B., Dubois, B., Durand, I., Cella, M., Lanzavecchia, A., Banchereau, J., 1997 CD34 + hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to granulocyte-macrophage colony-stimulating factor plus tumor necrosis factor alpha: II Functional analysis Blood 90 (4), 1458–1470 Chao, M.P., et al., 2010 Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma Cell 142 (5), 699–713 Chapuis, F., et al., 1997 Differentiation of human dendritic cells from monocytes in vitro Eur J Immunol 27 (2), 431–441 Chen, D.S., Mellman, I., 2013 Oncology meets immunology: the cancer-immunity cycle Immunity 39 (1), 1–10 Chiba, S., et al., 2012 Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1 Nat Immunol 13 (9), 832–842 Chiodoni, C., et al., 1999 Dendritic cells infiltrating tumors cotransduced with granulocyte/ macrophage colony-stimulating factor (GM-CSF) and CD40 ligand genes take up and present endogenous tumor-associated antigens, and prime naive mice for a cytotoxic T lymphocyte response J Exp Med 190 (1), 125–133 Chomarat, P., Dantin, C., Bennett, L., Banchereau, J., Palucka, A.K., 2003 TNF skews monocyte differentiation from macrophages to dendritic cells J Immunol 171 (5), 2262–2269 ARTICLE IN PRESS 28 Jan Martinek et al Collin, M., Bigley, V., 2018 Human dendritic cell subsets: an update Immunology 54 (1), 3–20 Curtis, C., Shah, S.P., Chin, S.F., Turashvili, G., Rueda, O.M., Dunning, M.J., Speed, D., Lynch, A.G., Samarajiwa, S., Yuan, Y., Gr€af, S., Ha, G., Haffari, G., Bashashati, A., Russell, R., McKinney, S., METABRIC Group, Langerød, A., Green, A., Provenzano, E., Wishart, G., Pinder, S., Watson, P., Markowetz, F., Murphy, L., Ellis, I., Purushotham, A., Børresen-Dale, A.L., Brenton, J.D., Tavare, S., Caldas, C., Aparicio, S., 2012 The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups Nature 486 (7403), 346–352 Darnell, J.C., Albert, M.L., Darnell, R.B., 2000 Cdr2, a target antigen of naturally occuring human tumor immunity, is widely expressed in gynecological tumors Cancer Res 60 (8), 2136–2139 de Baey, A., Lanzavecchia, A., 2000 The role of aquaporins in dendritic cell macropinocytosis J Exp Med 191, 743–748 De Monte, L., et al., 2011 Intratumor T helper type cell infiltrate correlates with cancerassociated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer J Exp Med 208 (3), 469–478 DeNardo, D.G., Barreto, J.B., Andreu, P., Vasquez, L., Tawfik, D., Kolhatkar, N., Coussens, L.M., 2009 CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages Cancer Cell 16 (2), 91–102 Diamond, M.S., Kinder, M., Matsushita, H., Mashayekhi, M., Dunn, G.P., Archambault, J.M., Lee, H., Arthur, C.D., White, J.M., Kalinke, U., Murphy, K.M., Schreiber, R.D., 2011 Type I interferon is selectively required by dendritic cells for immune rejection of tumors J Exp Med 208 (10), 1989–2003 Dieu-Nosjean, M.C., et al., 1999 Regulation of dendritic cell trafficking: a process that involves the participation of selective chemokines J Leukoc Biol 66 (2), 252–262 Disis, M.L., Sciffman, K., 2001 Cancer vaccines targeting the HER2/neu oncogenic protein Semin Oncol 28 (6 Suppl 18), 12–20 Dougan, M., Dranoff, G., Dougan, S.K., 2019a Cancer immunotherapy: beyond checkpoint blockade Annu Rev Cancer Biol 3, 55–75 Dougan, M., Dranoff, G., Dougan, S.K., 2019b GM-CSF, IL-3, and IL-5 Family of Cytokines: Regulators of Inflammation Immunity 50 (4), 796–811 Dunn, G.P., Old, L.J., Schreiber, R.D., 2004 The three Es of cancer immunoediting Annu Rev Immunol 22, 329–360 Durand, M., Segura, E., 2015 The known unknowns of the human dendritic cell network Front Immunol 6, 129 Elaraj, D.M., Weinreich, D.M., Varghese, S., Puhlmann, M., Hewitt, S.M., Carroll, N.M., et al., 2006 The role of interleukin in growth and metastasis of human cancer xenografts Clin Cancer Res 12, 1088–1096 Enk, A.H., et al., 1997 Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma Int J Cancer 73 (3), 309–316 Fehervari, Z., Sakaguchi, S., 2004 CD4 + Tregs and immune control J Clin Invest 114 (9), 1209–1217 Fesnak, A.D., June, C.H., Levine, B.L., 2016 Engineered T cells: the promise and challenges of cancer immunotherapy Nat Rev Cancer 16 (9), 566–581 Finn, O.J., 2008 Cancer immunology N Engl J Med 358 (25), 2704–2715 Forster, R., et al., 1999 CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs Cell 99, 23–33 Freeman, G.J., et al., 2000 Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation J Exp Med 192 (7), 1027–1034 ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 29 Fuertes, M.B., Kacha, A.K., Kline, J., Woo, S.-R., Kranz, D.M., Murphy, K.M., Gajewski, T.F., 2011 Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α + dendritic cells J Exp Med 208 (10), 2005 Fuertes, M.B., Woo, S.R., Burnett, B., Fu, Y.X., Gajewski, T.F., 2013 Type I interferon response and innate immune sensing of cancer Trends Immunol 34, 67–73 Gabrilovich, D.I., et al., 1997 Decreased antigen presentation by dendritic cells in patients with breast cancer In Process Citation, Clin Cancer Res (3), 483–490 Gajewski, T.F., Zha, Y., Thurner, B., Schuler, G., 2009 Association of gene expression profile in melanoma and survival to a dendritic cell-based vaccine J Clin Oncol 27, 9002 Gall, V.A., Philips, A.V., Qiao, N., Clise-Dwyer, K., Perakis, A.A., Zhang, M., Clifton, G.T., Sukhumalchandra, P., Ma, Q., Reddy, S.M., Yu, D., Molldrem, J.J., Peoples, G.E., Alatrash, G., Mittendorf, E.A., 2017 Trastuzumab increases HER2 uptake and cross-presentation by dendritic cells Cancer Res 77 (19), 5374–5383 Galon, J., Angell, H.K., Bedognetti, D., Marincola, F.M., 2013 The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures Immunity 39 (1), 11–26 Garris, C.S., Arlauckas, S.P., Kohler, R.H., Trefny, M.P., Garren, S., Piot, C., Engblom, C., Pfirschke, C., Siwicki, M., Gungabeesoon, J., Freeman, G.J., Warren, S.E., Ong, S., Browning, E., Twitty, C.G., Pierce, R.H., Le MH, A.A.P., Daud, A.I., Pai, S.I., Zippelius, A., Weissleder, R., Pittet, M.J., 2018 Successful anti-PD-1 Cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12 Immunity 49 (6), 1148–1161 e7 Geissmann, F., et al., 2001 A subset of human dendritic cells expresses IgA Fc receptor (CD89), which mediates internalization and activation upon cross-linking by IgA complexes J Immunol 166 (1), 346–352 Goudot, C., Coillard, A., Villani, A.C., Gueguen, P., Cros, A., Sarkizova, S., Tang-Huau, T.L., Bohec, M., Baulande, S., Hacohen, N., Amigorena, S., Segura, E., 2017 Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages Immunity 47 (3), 582–596 e6 Greter, M., Helft, J., Chow, A., Hashimoto, D., Mortha, A., Agudo-Cantero, J., Bogunovic, M., Gautier, E.L., Miller, J., Leboeuf, M., Lu, G., Aloman, C., Brown, B.D., Pollard, J.W., Xiong, H., Randolph, G.J., Chipuk, J.E., Frenette, P.S., Merad, M., 2012 GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells Immunity 36 (6), 1031–1046 Gringhuis, S.I., den Dunnen, J., Litjens, M., van der Vlist, M., Wevers, B., Bruijns, S.C., Geijtenbeek, T.B., 2009 Dectin-1 directs T helper cell differentiation by controlling noncanonical NF-kappaB activation through Raf-1 and Syk Nat Immunol 10 (2), 203–213 Guermonprez, P., Amigorena, S., 2005 Pathways for antigen cross presentation Springer Semin Immunopathol 26 (3), 257–271 Guilliams, M., Bruhns, P., Saeys, Y., Hammad, H., Lambrecht, B.N., 2014 The function of Fcγ receptors in dendritic cells and macrophages Nat Rev Immunol 14 (2), 94–108 Gutierrez-Arcelus, M., Rich, S.S., Raychaudhuri, S., 2016 Autoimmune diseases— connecting risk alleles with molecular traits of the immune system Nat Rev Genet 17 (3), 160–174 Hafler, D.A., De Jager, P.L., 2005 Applying a new generation of genetic maps to understand human inflammatory disease Nat Rev Immunol (1), 83–91 Hagai, T., et al., 2018 Gene expression variability across cells and species shapes innate immunity Nature 563 (7730), 197–202 ARTICLE IN PRESS 30 Jan Martinek et al Hagnerud, S., et al., 2006 Deficit of CD47 results in a defect of marginal zone dendritic cells, blunted immune response to particulate antigen and impairment of skin dendritic cell migration J Immunol 176, 5772–5778 Hammerich, L., et al., 2019 Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination Nat Med 25 (5), 814–824 Harshyne, L.A., et al., 2003 A role for class A scavenger receptor in dendritic cell nibbling from live cells J Immunol 170 (5), 2302–2309 Haslam, A., Prasad, V., 2019 Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs JAMA Netw Open (5), e192535 Helft, J., Ginhoux, F., Bogunovic, M., Merad, M., 2010 Origin and functional heterogeneity of non-lymphoid tissue dendritic cells in mice Immunol Rev 234 (1), 55–75 Hildner, K., Edelson, B.T., Purtha, W.E., Diamond, M., Matsushita, H., Kohyama, M., Calderon, B., Schraml, B.U., Unanue, E.R., Diamond, M.S., Schreiber, R.D., Murphy, T.L., Murphy, K.M., 2008 Batf3 deficiency reveals a critical role for CD8alpha + dendritic cells in cytotoxic T cell immunity Science 322 (5904), 1097–1100 Huang, B., et al., 2008 SCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment Blood 112 (4), 1269–1279 Idoyaga, J., et al., 2013 Specialized role of migratory dendritic cells in peripheral tolerance induction J Clin Invest Inaba, K., Schuler, G., Steinman, R.M., van Furth, R., 1993 GM-CSF: a granulocyte/macrophage/dendritic cell stimulating factor In: Hemopoietic Growth Factors And Mononuclear Phagocytes S.Karger, Basel, pp 187–196 Inaba, K., et al., 1998 Efficient presentation of phagocytosed cellular fragments on the major histocompatibility complex class II products of dendritic cells J Exp Med 188 (11), 2163–2173 Iraolagoitia, X.L., Spallanzani, R.G., Torres, N.I., Araya, R.E., Ziblat, A., Domaica, C.I., Sierra, J.M., Nun˜ez, S.Y., Secchiari, F., Gajewski, T.F., Zwirner, N.W., Fuertes, M.B., 2016 NK cells restrain spontaneous antitumor CD8 + T cell priming through PD-1/PD-L1 interactions with dendritic cells J Immunol 197 (3), 953–961 Jaiswal, S., et al., 2009 CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis Cell 138 (2), 271–285 Janeway Jr., C.A., Medzhitov, R., 2002 Innate immune recognition Annu Rev Immunol 20, 197–216 Joffre, O.P., Segura, E., Savina, A., Amigorena, S., 2012 Cross-presentation by dendritic cells Nat Rev Immunol 12 (8), 557–569 Jones, P.A., et al., 2019 Epigenetic therapy in immune-oncology Nat Rev Cancer 19 (3), 151–161 Jongbloed, S.L., Kassianos, A.J., KJ, M.D., Clark, G.J., Ju, X., Angel, C.E., Chen, C.J., Dunbar, P.R., Wadley, R.B., Jeet, V., Vulink, A.J., Hart, D.N., Radford, K.J., 2010 Human CD141 + (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens J Exp Med 207 (6), 1247–1260 Jutras, I., Desjardins, M., 2005 Phagocytosis: at the crossroads of innate and adaptive immunity Annu Rev Cell Dev Biol 21, 511–527 Kaplanov, I., Carmi, Y., Kornetsky, R., Shemesh, A., Shurin, G.V., Shurin, M.R., Dinarello, C.A., Voronov, E., Apte, R.N., 2019 Blocking IL-1β reverses the immunosuppression in mouse breast cancer and synergizes with anti-PD-1 for tumor abrogation Proc Natl Acad Sci U S A 116 (4), 1361–1369 Kawai, T., Akira, S., 2006 TLR signaling Cell Death Differ 13, 816–825 ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 31 Keeble, A.H., Khan, Z., Forster, A., James, L.C., 2008 TRIM21 is an IgG receptor that is structurally, thermodynamically, and kinetically conserved Proc Natl Acad Sci 105 (16), 6045–6050 Klechevsky, E., Morita, R., Liu, M., Cao, Y., Coquery, S., Thompson-Snipes, L., Briere, F., Chaussabel, D., Zurawski, G., Palucka, A.K., Reiter, Y., Banchereau, J., Ueno, H., 2008 Functional specializations of human epidermal Langerhans cells and CD14 + dermal dendritic cells Immunity 29 (3), 497–510 Knutson, K.L., Sciffman, K., Disis, M.L., 2001 Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients J Clin Invest 107 (4), 477–484 Krummel, M.F., Allison, J.P., 1995 CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation J Exp Med 182 (2), 459–465 Kuan, E.L., Ziegler, S.F., 2018 A tumor-myeloid cell axis, mediated via the cytokines IL-1alpha and TSLP, promotes the progression of breast cancer Nat Immunol 19 (4), 366–374 Lanzavecchia, A., Sallusto, F., 2001 Regulation of T cell immunity by dendritic cells Cell 106 (3), 263–266 Latour, S., et al., 2001 Bidirectional negative regulation of human T and dendritic cells by CD47 and its cognate receptor signal-regulator protein-alpha: down-regulation of IL-12 responsiveness and inhibition of dendritic cell activation J Immunol 167, 2547–2554 Lavin, Y., Kobayashi, S., Leader, A., Amir, E.D., Elefant, N., Bigenwald, C., Remark, R., Sweeney, R., Becker, C.D., Levine, J.H., Meinhof, K., Chow, A., Kim-Shulze, S., Wolf, A., Medaglia, C., Li, H., Rytlewski, J.A., Emerson, R.O., Solovyov, A., Greenbaum, B.D., Sanders, C., Vignali, M., Beasley, M.B., Flores, R., Gnjatic, S., Pe’er, D., Rahman, A., Amit, I., Merad, M., 2017 Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses Cell 169 (4), 750-765.e17 Le Floc’h, A., et al., 2007 Alpha E beta integrin interaction with E-cadherin promotes antitumor CTL activity by triggering lytic granule polarization and exocytosis J Exp Med 204 (3), 559–570 Le, D.T., et al., 2017 Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade Science 357 (6349), 409–413 Lehmann, C.H.K., Baranska, A., Heidkamp, G.F., Heger, L., Neubert, K., L€ uhr, J.J., Hoffmann, A., Reimer, K.C., Br€ uckner, C., Beck, S., Seeling, M., Kießling, M., Soulat, D., Krug, A.B., Ravetch, J.V., JHW, L., Nimmerjahn, F., Dudziak, D., 2017 DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo J Exp Med 214 (5), 1509–1528 Lesokhin, A.M., et al., 2015 On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation Sci Transl Med (280), 280sr1 Liu, Y., et al., 2006 Regulated expression of FcgR in human dendritic cells controls cross-presentation of antigen-antibody complexes J Immunol 177, 8440–8447 Majeti, R., et al., 2009 CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells Cell 138 (2), 286–299 Mangino, M., et al., 2017 Innate and adaptive immune traits are differentially affected by genetic and environmental factors Nat Commun 8, 13850 Mantovani, A., Allavena, P., Sica, A., Balkwill, F., 2008 Cancer-related inflammation Nature 454, 436–444 Mantovani, A., Dinarello, C.A., Molgora, M., Garlanda, C., 2019a Interleukin-1 and related cytokines in the regulation of inflammation and immunity Immunity 50 (4), 778–795 Mantovani, A., et al., 2019b Interleukin-1 and related cytokines in the regulation of inflammation and immunity Immunity 50 (4), 778–795 ARTICLE IN PRESS 32 Jan Martinek et al Marigo, I., et al., 2008 Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells Immunol Rev 222, 162–179 Marigo, I., et al., 2016 T cell cancer therapy requires CD40-CD40L activation of tumor necrosis factor and inducible nitric-oxide-synthase-producing dendritic cells Cancer Cell 30 (3), 377–390 Martins, I., et al., 2010 Surface-exposed calreticulin in the interaction between dying cells and phagocytes Ann N Y Acad Sci 1209, 77–82 Matsui, T., et al., 2009 CD2 distinguishes two subsets of human plasmacytoid dendritic cells with distinct phenotype and functions J Immunol 182 (11), 6815–6823 Matsushita, H., et al., 2012 Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting Nature 482 (7385), 400–404 Melief, C.J., 2008 Cancer immunotherapy by dendritic cells Immunity 29 (3), 372–383 Mellman, I., Steinman, R.M., 2001 Dendritic cells: specialized and regulated antigen processing machines Cell 106 (3), 255–258 Menetrier-Caux, C., Montmain, G., Dieu, M.C., Bain, C., Favrot, M.C., Caux, C., Blay, J.Y., 1998 Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor Blood 92 (12), 4778–4791 Mestas, J., Hughes, C.C.W., 2004 Of mice and men: differences between mouse and human immunology J Immunol 172, 2731–2738 Michea, P., et al., 2018 Adjustment of dendritic cells to the breast-cancer microenvironment is subset specific Nat Immunol 19 (8), 885–897 Mikucki, M.E., et al., 2015 Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell trafficking across tumour vascular checkpoints Nat Commun 6, 7458 Min, L., Mohammad Isa, S.A., Shuai, W., Piang, C.B., Nih, F.W., Kotaka, M., Ruedl, C., 2010 Cutting edge: granulocyte-macrophage colony-stimulating factor is the major CD8 + T cell-derived licensing factor for dendritic cell activation J Immunol 184 (9), 4625–4629 Mittag, D., Proeitto, A.L., Loudovaris, T., Mannering, S.I., Vremec, D., Shortman, K., Wu, L., Harrison, L.C., 2011 Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status J Immunol 186 (11), 6207–6217 Mittal, D., et al., 2017 Interleukin-12 from CD103(+) Batf3-dependent dendritic cells required for NK-cell suppression of metastasis Cancer Immunol Res (12), 1098–1108 Mora, J.R., Bono, M.R., Manjunath, N., Weninger, W., Cavanagh, L.L., Rosemblatt, M., Von Andrian, U.H., 2003 Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells Nature 424 (6944), 88–93 Moussion, C., Mellman, I., 2018 The dendritic cell strikes back Immunity 49 (6), 997–999 Muller, L., et al., 2016 Tumor-derived exosomes regulate expression of immune functionrelated genes in human T cell subsets Sci Rep 6, 20254 Mullins, I.M., et al., 2004 CXC chemokine receptor expression by activated CD8+ T cells is associated with survival in melanoma patients with stage III disease Cancer Res 64 (21), 7697–7701 Nagarsheth, N., Wicha, M.S., Zou, W., 2017 Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy Nat Rev Immunol 17 (9), 559–572 Ng, P.M.L., et al., 2019 Enhancing antigen cross-presentation in human monocyte-derived dendritic cells by recruiting the intracellular Fc receptor TRIM21 J Immunol 202 (8), 2307–2319 Nimmerjahn, F., Ravetch, J.V., 2006 Fcg receptors: old friends and new family members Immunity 24, 19–28 ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 33 Obeid, M., et al., 2007 Calreticulin exposure dictates the immunogenicity of cancer cell death Nat Med 13 (1), 54–61 Olkhanud, P.B., Rochman, Y., Bodogai, M., Malchinkhuu, E., Wejksza, K., Xu, M., Gress, R.E., Hesdorffer, C., Leonard, W.J., Biragyn, A., 2011a Thymic stromal lymphopoietin is a key mediator of breast cancer progression J Immunol 186 (10), 5656–5662 Olkhanud, P.B., et al., 2011b Thymic stromal lymphopoietin is a key mediator of breast cancer progression J Immunol 186 (10), 5656–5662 Padovan, E., Spagnoli, G.C., Ferrantini, M., Heberer, M., 2002 IFN-alpha2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8 + effector T cells J Leukoc Biol 71, 669–676 Palucka, K., Banchereau, J., 2014 SnapShot: cancer vaccines Cell 157 (2), 516 e1 Palucka, A.K., Coussens, L.M., 2016a The basis of oncoimmunology Cell 164 (6), 1233–1247 Palucka, A.K., Coussens, L.M., 2016b The basis of oncoimmunology Cell 164, 1233–1247 Park, K.H., Gad, E., Goodell, V., Dang, Y., Wild, T., Higgins, D., Fintak, P., Childs, J., Dela Rosa, C., Disis, M.L., 2008 Insulin-like growth factor-binding protein-2 is a target for the immunomodulation of breast cancer Cancer Res 68 (20), 8400–8409 Pascual, V., Chaussabel, D., Banchereau, J., 2010 A genomic approach to human autoimmune diseases Annu Rev Immunol 28, 535–571 Paul, W.E (Ed.), 2003 Fundamental immunology, fifth ed Lippincott Williams & Wilkins, Philadelphia, p 1701 Pedroza-Gonzalez, A., Xu, K., Wu, T.C., Aspord, C., Tindle, S., Marches, F., Gallegos, M., Burton, E.C., Savino, D., Hori, T., Tanaka, Y., Zurawski, S., Zurawski, G., Bover, L., Liu, Y.J., Banchereau, J., Palucka, A.K., 2011a Thymic stromal lymphopoietin fosters human breast tumor growth by promoting type inflammation J Exp Med 208 (3), 479–490 Pedroza-Gonzalez, A., et al., 2011b Thymic stromal lymphopoietin fosters human breast tumor growth by promoting type inflammation J Exp Med 208 (3), 479–490 Perrot, I., et al., 2007 Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage J Immunol 178, 2763–2769 Pincetic, A., Bournazos, S., DJ, D.L., Maamary, J., Wang, T.T., Dahan, R., Fiebiger, B.M., Ravetch, J.V., 2014 Type I and type II Fc receptors regulate innate and adaptive immunity Nat Immunol 15 (8), 707–716 Pio, R., et al., 2019 Complementing the Cancer-Immunity Cycle Front Immunol 10, 774 Platt, N., da Silva, R.P., Gordon, S., 1998 Recognizing death: the phagocytosis of apoptotic cells Trends Cell Biol (9), 365–372 Platzer, B., Stout, M., Fiebiger, E., 2014 Antigen cross-presentation of immune complexes Front Immunol 5, 140 Poulin, L.F., Salio, M., Griessinger, E., Anjos-Afonso, F., Craciun, L., Chen, J.L., Keller, A.M., Joffre, O., Zelenay, S., Nye, E., Le Moine, A., Faure, F., Donckier, V., Sancho, D., Cerundolo, V., Bonnet, D., Reis e Sousa, C., 2010 Characterization of human DNGR-1 + BDCA3 + leukocytes as putative equivalents of mouse CD8alpha + dendritic cells J Exp Med 207 (6), 1261–1271 Pulendran, B., Palucka, K., Banchereau, J., 2001 Sensing pathogens and tuning immune responses Science 293 (5528), 253–256 Rosenberg, S.A., et al., 2008 Adoptive cell transfer: a clinical path to effective cancer immunotherapy Nat Rev Cancer (4), 299–308 Rovere, P., et al., 1998 Dendritic cells preferentially internalize apoptotic cells opsonized by anti-beta2-glycoprotein I antibodies J Autoimmun 11 (5), 403–411 ARTICLE IN PRESS 34 Jan Martinek et al Ruffell, B., Coussens, L.M., 2015 Macrophages and therapeutic resistance in cancer Cancer Cell 27 (4), 462–472 Rughetti, A., et al., 2005 Recombinant tumor-associated MUC1 glycoprotein impairs the differentiation and function of dendritic cells J Immunol 174 (12), 7764–7772 Ryan Kolb, L.P., Borcherding, N., Liu, Y., Yuan, F., Janowski, A.M., Xie, Q., Markan, K.R., Li, W., Potthoff, M.J., Fuentes-Mattei, E., Ellies, L.G., Michael Knudson, C., Lee, M.-H., Yeung, S.-C.J., Cassel, S.L., Sutterwala, F.S., Zhang, W., 2016 Obesity-associated NLRC4 inflammasome activation drives breast cancer progression Nat Commun 7, 13007 Salcedo, R., Cataisson, C., Hasan, U., Yuspa, S.H., Trinchieri, G., 2013 MyD88 and its divergent toll in carcinogenesis Trends Immunol 34, 379–389 Salmon, H., et al., 2016a Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition Immunity 44 (4), 924–938 Salmon, H., Idoyaga, J., Rahman, A., Leboeuf, M., Remark, R., Jordan, S., CasanovaAcebes, M., Khudoynazarova, M., Agudo, J., Tung, N., Chakarov, S., Rivera, C., Hogstad, B., Bosenberg, M., Hashimoto, D., Gnjatic, S., Bhardwaj, N., Palucka, A.K., Brown, B.D., Brody, J., Ginhoux, F., Merad, M., 2016b Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition Immunity 44 (4), 924–938 Sa´nchez-Paulete, A.R., Cueto, F.J., Martı´nez-Lo´pez, M., Labiano, S., Morales-Kastresana, A., Rodrı´guez-Ruiz, M.E., Jure-Kunkel, M., Azpilikueta, A., Aznar, M.A., Quetglas, J.I., Sancho, D., Melero, I., 2016 Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires batf3-dependent dendritic cells Cancer Discov (1), 71–79 Sancho, D., Reis e Sousa, C., 2013 Sensing of cell death by myeloid C-type lectin receptors Curr Opin Immunol 25 (1), 46–52 Sandoval, F., et al., 2013 Mucosal imprinting of vaccine-induced CD8(+) T cells is crucial to inhibit the growth of mucosal tumors Sci Transl Med (172), 172ra20 Sansone, P., et al., 2017 Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer Proc Natl Acad Sci U S A 114 (43), E9066–E9075 Santiago-Schwartz, F., et al., 1992 TNF in combination with GM-CSF enhances the differentiation of neonatal cord blood stem cells into dendritic cells and macrophages J Leukoc Biol 52, 274–281 Schneider, T., Hoffmann, H., Dienemann, H., Schnabel, P.A., Enk, A.H., Ring, S., Mahnke, K., 2011 Non-small cell lung cancer induces an immunosuppressive phenotype of dendritic cells in tumor microenvironment by upregulating B7-H3 J Thorac Oncol 6, 1162–1168 Schreibelt, G., Klinkenberg, L.J.J., Cruz, L.J., Tacken, P.J., Tel, J., Kreutz, M., Adema, G.J., Brown, G.D., Figdor, C.G., de Vries, I.J.M., 2012 The C-type lectin receptor CLEC9A mediates antigen uptake and (cross-)presentation by human blood BDCA3+ myeloid dendritic cells Blood 119 (10), 2284–2292 Schuler, P.J., et al., 2014 Human CD4+ CD39+ regulatory T cells produce adenosine upon co-expression of surface CD73 or contact with CD73+ exosomes or CD73 + cells Clin Exp Immunol 177 (2), 531–543 Schumacher, T.N., Schreiber, R.D., 2015 Neoantigens in cancer immunotherapy Science 348 (6230), 69–74 Schupp, J., Krebs, F.K., Zimmer, N., Trzeciak, E., Schuppan, D., Tuettenberg, A., 2017 Targeting myeloid cells in the tumor sustaining microenvironment Cell Immunol S0008-8749 (17), 30190–30199 ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 35 See, P., et al., 2017 Mapping the human DC lineage through the integration of highdimensional techniques Science 356 (6342), eaag3009 Shalapour, S., Karin, M., 2015 Immunity, inflammation, and cancer: an eternal fight between good and evil J Clin Invest 125 (9), 3347–3355 Sharma, P., Allison, J.P., 2015a The future of immune checkpoint therapy Science 348 (6230), 56–61 Sharma, P., Allison, J.P., 2015b Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential Cell 161 (2), 205–214 Sharma, P., et al., 2017 Primary, adaptive, and acquired resistance to cancer immunotherapy Cell 168 (4), 707–723 Siegal, F.P., Kadowaki, N., Shodell, M., Fitzgerald-Bocarsly, P.A., Shah, K., Ho, S., Antonenko, S., Liu, Y.J., 1999 The nature of the principal type interferon-producing cells in human blood Science Cell 284 (5421), 1835–1837 Sinclair, C., Bommakanti, G., Gardinassi, L., Loebbermann, J., Johnson, M.J., Hakimpour, P., Hagan, T., Benitez, L., Todor, A., Machiah, D., Oriss, T., Ray, A., Bosinger, S., Ravindran, R., Li, S., Pulendran, B., 2017 mTOR regulates metabolic adaptation of APCs in the lung and controls the outcome of allergic inflammation Science 357 (6355), 1014–1021 Spranger, S., Bao, R., Gajewski, T.F., 2015 Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity Nature 523, 231–235 Spranger, S., Dai, D., Horton, B., Gajewski, T.F., 2017 Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy Cancer Cell 31 (5), 711–723 e4 Steinbrink, K., et al., 1999 Interleukin-10-treated human dendritic cells induce a melanoma-antigen- specific anergy in CD8(+) T cells resulting in a failure to lyse tumor cells Blood 93, 1634–1642 Steinman, R.M., 2011 Decisions about dendritic cells: past, present, and future Annu Rev Immunol 30, 1–22 Steinman, R.M., Banchereau, J., 2007 Taking dendritic cells into medicine Nature 449 (7161), 419–426 Steinman, R.M., Cohn, Z.A., 1973 Identification of a novel cell type in peripheral lymphoid organs of mice I Morphology, quantitation, tissue distribution J Exp Med 137 (5), 1142–1162 Subra, C., et al., 2010 Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins J Lipid Res 51 (8), 2105–2120 Swiercz, R., Mo, M., Khare, P., Schneider, Z., Ober, R.J., Ward, E.S., 2016 Loss of expression of the recycling receptor , FcRn , promotes tumor cell growth by increasing albumin consumption Oncotarget (2), 3528–3541 Tanchot, C., et al., 2012 Tumor-infiltrating regulatory T cells: phenotype, role, mechanism of expansion in situ and clinical significance Cancer Microenviron (2), 147–157 Theisen, D.J., Davidson, J.T., Brisen˜o, C.G., Gargaro, M., Lauron, E.J., Wang, Q., Desai, P., Durai, V., Bagadia, P., Brickner, J.R., Beatty, W.L., Virgin, H.W., Gillanders, W.E., Mosammaparast, N., Diamond, M.S., Sibley, L.D., Yokoyama, W., Schreiber, R.D., Murphy, T.L., Murphy, K.M., 2018 WDFY4 is required for crosspresentation in response to viral and tumor antigens Science 362 (6415), 694–699 Thurnher, M., et al., 1996 Human renal cell carcinoma tissue contains dendritic cells Int J Cancer 67, 1–7 Tkach, M., Kowal, J., Zucchetti, A.E., Enserink, L., Jouve, M., Lankar, D., Saitakis, M., Martin-Jaular, L., Thery, C., 2017 Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes EMBO J 36, 3012–3028 Topalian, S.L., Drake, C.G., Pardoll, D.M., 2015 Immune checkpoint blockade: a common denominator approach to cancer therapy Cancer Cell 27 (4), 450–461 ARTICLE IN PRESS 36 Jan Martinek et al Trombetta, E.S., Mellman, I., 2005 Cell biology of antigen processing in vitro and in vivo Annu Rev Immunol 23, 975–1028 Ushach, I., Zlotnik, A., 2016 Biological role of granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) on cells of the myeloid lineage J Leukoc Biol 100 (3), 481–489 Veglia, F., et al., 2019 Fatty acid transport protein reprograms neutrophils in cancer Nature 569 (7754), 73–78 Villani, A.C., et al., 2017 Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors Science 356 (6335), eaah4573 Wada, H., Noguchi, Y., Marino, M.W., Dunn, A.R., Old, L.J., 1997 T cell functions in granulocyte/macrophage colony-stimulating factor deficient mice Proc Natl Acad Sci USA 94, 12557–12561 Wakim, L.M., Bevan, M.J., 2011 Cross-dressed dendritic cells drive memory CD8 + T-cell activation after viral infection Nature 471 (7340), 629–632 Waldmann, T.A., Tagaya, Y., 1999 The multifaceted regulation of interleukin-15 expression and the role of this cytokine in NK cell differentiation and host response to intracellular pathogens Annu Rev Immunol 17, 19–49 Wang, E., Miller, L.D., Ohnmacht, G.A., Mocellin, S., Perez-Diez, A., Petersen, D., Zhao, Y., Simon, R., Powell, J.I., Asaki, E., Alexander, H.R., Duray, P.H., Herlyn, M., Restifo, N.P., Liu, E.T., Rosenberg, S.A., Marincola, F.M., 2002 Prospective molecular profiling of melanoma metastases suggests classifiers of immune responsiveness Cancer Res 62 (13), 3581–3586 Wang, E., Worschech, A., Marincola, F.M., 2008 The immunologic constant of rejection Trends Immunol 29 (6), 256–262 Waterhouse, P., et al., 1995 Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4 Science 270, 985–988 Wculek, S.K., Khouili, S.C., Priego, E., Heras-Murillo, I., Sancho, D., 2019 Metabolic control of dendritic cell functions: digesting information Front Immunol 10, 775 Wilcox, R.A., et al., 2002 Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors J Clin Invest 109, 651–659 Willingham, S.B., et al., 2012 The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors Proc Natl Acad Sci U S A 109 (17), 6662–6667 Wolfers, J., et al., 2001a Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming Nat Med 7, 297–303 Wolfers, J., et al., 2001b Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming Nat Med (3), 297–303 Woo, S.R., et al., 2014 STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors Immunity 41 (5), 830–842 Wu, T.C., et al., 2014 Reprogramming tumor-infiltrating dendritic cells for CD103 + CD8 + mucosal T-cell differentiation and breast cancer rejection Cancer Immunol Res (5), 487–500 Wu, T.C., et al., 2018 IL1 Receptor Antagonist Controls Transcriptional Signature of Inflammation in Patients with Metastatic Breast Cancer Cancer Res 78 (18), 5243–5258 Xu, M.M., et al., 2017 Dendritic Cells but Not Macrophages Sense Tumor Mitochondrial DNA for Cross-priming through Signal Regulatory Protein alpha Signaling Immunity 47 (2), 363–373 e5 Yamazaki, S., et al., 2006 Dendritic cells expand antigen-specific Foxp3CD25CD4 regulatory T cells including suppressors of alloreactivity Immunol Rev 212, 314–329 ARTICLE IN PRESS Interplay between dendritic cells and cancer cells 37 Yang, Z., Deng, F., Meng, L., 2018 Effect of dendritic cell immunotherapy on distribution of dendritic cell subsets in non-small cell lung cancer Exp Ther Med 15 (6), 4856–4860 Yao, C., et al., 2007 Interleukin-8 modulates growth and invasiveness of estrogen receptornegative breast cancer cells Int J Cancer 121 (9), 1949–1957 Ye, C.J., et al., 2014 Intersection of population variation and autoimmunity genetics in human T cell activation Science 345 (6202), 1254665 YJ, L., 2005 IPC: professional type interferon-producing cells and plasmacytoid dendritic cell precursors Annu Rev Immunol 23, 275–306 Yu, C.I., Becker, C., Wang, Y., Marches, F., Helft, J., Leboeuf, M., Anguiano, E., Pourpe, S., Goller, K., Pascual, V., Banchereau, J., Merad, M., Palucka, K., 2013 Human CD1c + dendritic cells drive the differentiation of CD103 + CD8+ mucosal effector T cells via the cytokine TGF-β Immunity 38 (4), 818–830 Zarling, A.L., et al., 2006 Identification of class I MHC-associated phosphopeptides as targets for cancer immunotherapy Proc Natl Acad Sci U S A 103, 14889–14894 Zhou, L.-J., Tedder, T.F., 1996 CD14 + blood monocytes can differentiate into functionally mature CD83 + dendritic cells Proc Natl Acad Sci U S A 93, 2588–2592 Zhu, J., Paul, W.E., 2008 CD4 T cells: fates, functions, and faults Blood 112 (5), 1557–1569 Zilionis, R., et al., 2019 Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species Immunity 50 (5), 1317–1334 e10 Zitvogel, L., Kepp, O., Galluzzi, L., Kroemer, G., 2012 Inflammasomes in carcinogenesis and anticancer immune responses Nat Immunol 13, 343–351 ... PRESS Interplay between dendritic cells and cancer cells Cancer cell Dendritic cell Stromal cells NK cells Neutrophils pDCs Mast cells MDSCs Macrophages T regs CD8 T cells CD4 T cells Th2s Fig Interplay. .. example, γδ-T cells and NK cells release IFN-γ, mast cells release pre-formed IL-4 and tumor necrosis factor (TNF), plasmacytoid (p)DCs secrete IFN-α, stromal cells secrete IL-15 and thymic stromal... Other pathways of DC modulation Tumor- secreted extracellular vesicles (EVs) are critical mediators of intercellular communication between tumor cells and stromal cells in local and distant microenvironments

Ngày đăng: 13/11/2019, 13:37

Mục lục

  • Interplay between dendritic cells and cancer cells

    • Introduction

    • Dendritic cells in the immune response to cancer

      • Dendritic cell maturation as checkpoint of tolerance and immunity

      • Antigen capture and modulation of DC maturation

        • Phagocytosis

        • Capture of immune complexes

        • Other pathways of DC modulation

        • Dendritic cell subsets in cancer

        • Dendritic cells dictate the outcome of immune response to cancer

          • DCs control anti-tumor immune response

          • The central role of type I IFN in tumor rejection

          • DCs in pro-tumor immunity and response to treatment

            • Studies in the human

            • Mouse models

            • Chronic inflammation promotes immune escape via DCs

            • Conclusions and future studies

            • Acknowledgments

            • References

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan