Skip to main content

Plasmacytoid dendritic cells at the forefront of anti-cancer immunity: rewiring strategies for tumor microenvironment remodeling

Abstract

Plasmacytoid dendritic cells (pDCs) are multifaceted immune cells executing various innate immunological functions. Their first line of defence consists in type I interferons (I-IFN) production upon nucleic acids sensing through endosomal Toll-like receptor (TLR) 7- and 9-dependent signalling pathways. Type I IFNs are a class of proinflammatory cytokines that have context-dependent functions on cancer immunosurveillance and immunoediting. In the last few years, different studies have reported that pDCs are also able to sense cytosolic DNA through cGAS–STING (stimulator of interferon genes) pathway eliciting a potent I-IFN production independently of TLR7/9. Human pDCs are also endowed with direct effector functions via the upregulation of TRAIL and production of granzyme B, the latter modulated by cytokines abundant in cancer tissues. pDCs have been detected in a wide variety of human malignant neoplasms, including virus-associated cancers, recruited by chemotactic stimuli. Although the role of pDCs in cancer immune surveillance is still uncompletely understood, their spontaneous activation has been rarely documented; moreover, their presence in the tumor microenvironment (TME) has been associated with a tolerogenic phenotype induced by immunosuppressive cytokines or oncometabolites. Currently tested treatment options can lead to pDCs activation and disruption of the immunosuppressive TME, providing a relevant clinical benefit. On the contrary, the antibody–drug conjugates targeting BDCA-2 on immunosuppressive tumor-associated pDCs (TA-pDCs) could be proposed as novel immunomodulatory therapies to achieve disease control in patients with advance stage hematologic malignancies or solid tumors. This Review integrate recent evidence on the biology of pDCs and their pharmacological modulation, suggesting their relevant role at the forefront of cancer immunity.

Background

Plasmacytoid dendritic cells (pDCs) are bone marrow-derived circulating innate immune cells, identified as CD123+ (IL3R), CD303+ (BDCA-2, also known as C-type lectin CLEC4C), CD304+ (BDCA-4) cells, and negative for lineage markers and CD11c [1].

pDCs home to lymphoid organs and inflamed peripheral tissues by chemotactic stimuli [2], where they execute relevant immune effector and regulatory functions upon nucleic acids sensing through different surface and cytosolic receptors. pDCs have been detected in a wide variety of human neoplasms, including carcinomas [e.g. uroendothelial bladder cancer (UBC); colon-rectal adenocarcinomas (CAD); lung squamous cell carcinoma (LSCC); head neck squamous cell carcinoma (HNSCC)], cutaneous melanoma (CM), lymphomas, and virus-associated cancers (e.g., cervical cancers, oral cancers, nasopharyngeal carcinoma, Kaposi’s sarcoma) [3, 4]. In the last decade, studies have proposed a relevant role for pDC also in cancer immunity. Their expansion and activation in the tumor tissue by recently developed compounds might promote a fine-tuning remodelling of the tumor microenviroment (TME). The present review will summarize recent advances on the role of pDCs in cancer by focusing on their intrinsic antitumor activity and functional states in cancer tissues. Based on these findings, this review will propose combinatorial strategies to overcome cellular and molecular immune escape mechanisms targeting pDCs biology.

The multifaceted biology of pDC in cancer

pDCs are multifaceted immune cells, executing innate immunological functions [5]. Their first line of defense against viral infections and cancer consists of type I interferons (I-IFNs) production upon sensing nucleic acids through endosomal Toll-like receptor (TLR) 7- and 9-dependent signaling pathways [6, 7]. Besides TLRs, human pDCs express different nucleic acid sensors, such as C-type lectin receptors (CLRs), RIG-I-like receptors (RLRs), NOD-like receptors (NLRs), and cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) [8, 9].

Moreover, pDCs are producers of type III interferons (IFN-λ or IL-28/IL-29) and have been identified among the restricted cell subsets also expressing the IFN-λ functional receptor (IFNλR) [10]. As previously reviewed [11], III-IFNs can serve as an autocrine signal to potentiate the anti-viral and antitumor activities of pDCs by triggering IFN-α and IFN-λ production [12, 13], and it also influences the activation status of pDCs by upregulating CD80 and CD86. In addition to IFNs production, pDCs could exert direct cytotoxic functions or potentiate Natural Killer (NK) cells effector functions. By TLR7/9 engagement, activated pDCs could exert killing capacities via the upregulation of TNF-related apoptosis inducing ligand (TRAIL) [14, 15]. Moreover, granzyme B (GrB) is constitutively expressed in human pDCs, but its production and release are further induced by the cytokines abundant in cancer tissues [15,16,17].

Tumor-associated pDCs (TA-pDCs; i.e. into the tumor milieu or tumor-draining lymph nodes) are recruited at the tumor site by chemokines (e.g. CXCL12 and CCL20) released by cancer cells or by the TME. The presence of pDCs in the TME has been associated with different outcomes suggesting their dual functional/regulatory state in cancer immunity. Indeed, they can exhibit either a tolerogenic or immunogenic phenotype, depending on the immunological context. Moreover, pDCs may interact with tumor-associated viruses [e.g. Human Papilloma Virus (HPV), Epstein-Barr Virus (EBV), Human herpes virus 8 (HHV8)] that persist in tumor tissues and may contribute to tumor immunity [4, 18,19,20]. However, no direct pDC-virus interaction in cancers has been reported as yet. Further studies are necessary to understand the crosstalk between oncogenic viruses and pDCs and whether the tumor-infiltrating pDCs in HPV/EBV/HHV8 positive tumors are immunocompetent. Although the role of pDCs in cancer immunoediting/immune surveillance is still uncompletely understood, a large set of studies have documented that tumor-infiltrating pDCs often exhibit a non-activated and tolerogenic state [21,22,23]. The non-activated tolerogenic TA-pDCs state could be induced by immunosuppressive cytokines or oncometabolites. Tolerogenic pDCs are characterized by low I-IFNs production and co-stimulatory molecule expression and promote regulatory T cell (Treg) induction together with increased expression of immunomodulatory molecules (e.g. IDO). Accordingly, a high density of TA-pDCs have been frequently associated with a poor clinical outcome. On the other hand, pDCs are endowed with the ability to induce maturation of conventional DC1 (cDC1), can present tumor antigens and prime tumor-specific cytotoxic CD8+ T cells [24,25,26]; however, whether and how human pDCs cross-prime CD8+ T cells in vivo has been controversial for a long time. Based upon a recent report, cross-presenting pDCs were unable to prime CD8+ T cells efficiently, but require conventional DCs (cDCs) to achieve CD8+ T cell cross-priming in vivo [27].

Novel immunotherapeutic approaches aim to disrupt the immunosuppressive TME by reprogramming the tolerogenic phenotype of TA-pDCs and other immune cells, towards an immunogenic phenotype boosting innate and adaptive tumor-specific immune responses.

News in pDC affiliation

Decoding pDC ontogeny

The dispute over pDC ontogeny and affiliation started from their first identification by Karl Lennert as cells with plasma cell-like morphology located in lymphoid tissues described as “lymphoblasts” [28] (Fig. 1A). Over the decades, hematopathologists referred to these cells as “T-associated plasma cells” [29], “plasmacytoid T cells” [30] and “plasmacytoid T-zone cells” [31]. Subsequently, these cells were characterized in morphology and phenotype and referred as to plasmacytoid monocytes [32, 33] (Fig. 1A). It was then reported that peripheral blood plasmacytoid monocytes can differentiate into dendritic cells under the influence of stimuli, such as viruses, IL-3, and CD40L [34,35,36,37]. Through isolation and in vitro studies, pDCs were identified as Natural Interferon Producing Cells [38, 39] (Fig. 1A).

Fig. 1
figure 1

Discovery of plasmacytoid dendritic cells, decoding their ontogeny and differentiation. a The timeline clarifies the different nomenclature and origin that have been attributed to human pDCs from their first identification to the present. b pDCs share features with both myeloid and lymphoid lineage opening a debate regarding their ontogeny. Two main developmental cascades are proposed to generate fully differentiated pDCs from lymphoid or myeloid progenitors. These findings were obtained from in vitro experiments by using hematopoietic stem progenitor cells (HSPC) from human bone marrow (indicated with human icons) or by using mouse models (indicated with mouse icons). Human pDCs express some B-lineage transcription factors such as E2-2, BCL11A and FOXP1, but lacks expression of most classical transcription factors for B-cells, including PAX5, BLIMP1, BOB1, MUM1/IRF4, OCT2, and BCL6. MPP: multipotent progenitor; CLP: common lymphoid progenitor; MLP: multipotent lymphoid progenitor; GMDP: granulocyte monocyte and dendritic cell progenitor; MDP: monocyte and dendritic cell progenitor; CDP: common dendritic cell progenitor. Created with BioRender.com

Although the name of this subset can be immediately traced back to classical dendritic cells, the origin of the pDC is still under debate within the scientific community [40,41,42] and it was supported by the pDC phenotype sharing concomitant features with both myeloid and lymphoid cells [32, 33, 38, 43]. Moreover, plasmacytoid and classic DCs exhibit distinct features in terms of morphology, phenotype, and functions [5, 39, 44,45,46], raising doubts over whether they belong to the same cell lineage.

Recently, Ziegler-Heitbrock and colleagues proposed reclassifying pDCs as innate lymphocytes, based on developmental trajectory shared with B cells and the scant classic DC functional properties (i.e. antigen presentation and migration via the lymphatics), or restoring their original name, type I-Interferon producing cells as the defining functional feature of these cells [40] (Fig. 1A).

It has been reported that pDCs can express lymphoid transcripts including the pre-T cell antigen receptor alpha chain (pT-α), Spi-B, γ5, Pax5, and TdT [47,48,49], suggesting the lymphoid origin of pDC [50]. Moreover, human pDCs and their neoplastic counterparts are also marked by some B-lineage transcription factors such as BCL11A and FOXP1 [51]. However, the similarities between pDCs and early B cells in terms of molecular features might rely on their expression of E2-2 and E2A/HEB transcription factors, which are known to regulate common target genes [52]. More importantly, human pDCs lack expression of most classic transcription factors for B cells including PAX5, BLIMP1, BOB1, MUM1/IRF4, OCT2, and BCL6 [51]. Thus, these evidences are insufficient to prove the common origin of B cells and pDCs [42]. An early study identified a mouse M-CSFR/CD115 IL-7Rα FLT3+ CDP with the highest expression level of homologous E protein transcription factors (E2-2, also known as Tcf4) and superior potential to generate pDC compared to canonical Lin cKITint/lo FLT3+ M-CSFR/CD115+ CDP, under the same conditions [53], postulating a possible dual origin of pDCs. Despite the fact that it is now collectively accepted that pDCs can develop from both myeloid and lymphoid precursors [54,55,56,57] (Fig. 1B), this issue confuses and divides the scientific community regarding pDC classification [40,41,42]. The entire process of pDC development takes place in the bone marrow (BM) from hematopoietic stem progenitor cells (HSPCs) and is orchestrated by specific cytokine signals. In common with cDCs, pDC development is strongly dependent on fms-like tyrosine kinase-3 ligand (Flt3L) [55, 57, 58] and transcriptional cofactor Trim33 (also known as TIF1-γ) [59]. Flt3L KO mice have reduced levels of pDCs and cDCs [55], and the expression of Flt3L receptor (Flt3/CD135) by BM precursor is essential to produce both murine and human DCs [55, 60]. Moreover, Trim33 deletion in vivo caused rapid loss of pDCs and cDCs affecting the earliest stages of differentiation, but it also abolished mature B cells. In particular, Trim33-deficient Flt3+ progenitors failed to induce the DC differentiation program establishing Trim33 as a key transcriptional regulator of Flt3L-driven pDCs and cDCs development [59]. The pivotal role of Flt3L driving pDC development from BM progenitors, but its failure to induce lymphoid cell commitment, was one of the main reasons claimed to justify the close developmental affinity of pDC to cDC lineage [32]. Besides the dependence on FLT3L cytokine, cDC and pDC share the expression of transcription factors, thus supporting a common regulatory network [42, 52, 55, 58]. Notably, pDC development was observed in mice depleted of lymphoid precursor through estrogen therapy [61]. Moreover, the common cytokine receptor γ-chain or the transcription factor JAK3, which are required for the development of lymphocytes, are not required for pDC development [32, 50, 55], further confirming the interrelation of pDCs and cDCs in the developmental process. Finally, pDCs and cDCs share a restricted common dendritic cell progenitor (CDP) through myeloid lineage, with the ability to respond to myeloid cytokines (MCSF, GMCSF) [62]. An in vitro culture system has been used to demonstrate human DC sequential origin from increasingly restricted progenitors [63]. CDP has been proposed as the unique progenitor restricted to DC lineage in humans originated from human monocyte-dendritic progenitor (hMDP), which in turn originates from a human granulocyte-monocyte-DC progenitor (hGMDP) [63]. Subsequently, Helft et al. showed that a large proportion of multipotent lymphoid early progenitors (MLPs) in humans were more efficient in generating CD141+ cDCs than common myeloid progenitors (CMPs), and MLPs were also the only progenitors able to generate CD303+ pDCs in vitro and in vivo [64]. Recently, clonal lineage tracing of mouse pDCs has confirmed the shared origin of conventional and plasmacytoid DCs [65], strongly in contrast with previous data identifying mouse Ly6DhiCD2hi lymphoid progenitors as unique progenitors for pDCs [66]. Pathogenic drivers in neoplastic entities, such as blastic plasmacytoid dendritic cell neoplasm (BPDCN), “Mature PDC Proliferation” (MPDCP), and acute myeloid leukemia (AML)-PDC, are shared by blasts, pDCs, monocytes and cDCs, suggesting a common differentiation trajectory [67, 68] and that a significant proportion of pDC neoplastic proliferation might originate from a myeloid precursor.

In summary, the ontogeny of pDCs remains controversial and a dual myeloid/lymphoid origin of pDCs should be considered in order to justify the pDC heterogeneity and plasticity observed under different conditions. ScRNA-seq studies provided helpful information in the area of pDC ontogeny [61, 66]; however, this intricate issue requires additional studies to be thoroughly understood.

Human pDC heterogeneity

Increasing reports have demonstrated the potential heterogeneity of pDC cell populations devoted to specific immune functions [69,70,71,72,73,74,75,76]. However, the mechanisms behind the different pDC functions have not yet been fully elucidated and it is unclear whether the environmental cues, rather than specific genetic programs, contribute to pDC diversification [77,78,79].

Originally, two pDC subsets were identified in human blood and tonsils based on the expression level of CD2 surface antigen: a rare CD2high pDC subset as compared to the major fraction of CD2low pDCs [69, 80] (Fig. 2). CD2high and CD2low pDC subsets showed differentiated ability to initiate T cell immune responses associated with distinct transcriptional profiles. Moreover, upon activation, CD2high pDCs showed higher expression of costimulatory molecule CD80 and higher production of IL-12p40 compared to CD2low pDCs, accompanied by an increased capacity to trigger naive T cell expansion. Of note, CD2high pDCs unexpectedly overexpressed the lysozyme (LYZ), a typical myeloid marker, and the IFN-regulated surface tyrosine kinase receptor AXL. More recently, a further characterization of human pDCs in bone marrow, cord blood, and tonsils enabled the division of CD2high pDCs into CD5+CD81+ and CD5CD81 subsets both expressing classical pDC surface markers and pDC-defining transcription factors E2-2/TCF4 and SPIB [70, 80]. However, the CD2highCD5+CD81+ pDCs significantly diverged in their transcriptional profile, including a notable overexpression of LYZ and AXL receptor, and functional studies revealed that CD2highCD5+CD81+ pDCs showed a mature phenotype (i.e. costimulatory molecule and HLA-DR expression) and a weak ability to produce IFN-α, consistent with low expression of the Interferon Regulatory Factor 7 (IRF7) [70] (Fig. 2). Specifically, CD5+CD81+ pDCs were able to produce large quantities of proinflammatory cytokines, such as IL-12p40 and IL-6, promoting T cell proliferation and inducing their differentiation into Treg [70]. A further functional feature of CD5+CD81+ pDCs was their ability to activate B cells and induce plasma cell differentiation and antibody production dependent on pDC–B cell contact through CD70-CD27 interaction [70]. It should be noted that tissue pDCs are mostly negative for CD2 and CD5 [33, 51], as revealed by immunostaining on reactive lymph nodes and cutaneous lupus erythematosus (Fig. 3), suggesting that phenotypic diversification in different microenvironments [81] or different developmental phases might occur. This finding could be also explained by the absent/low expression of surface CCR7 on CD2+CD5+Axl+ DCs [82]. Based on single-cell transcriptomic profiling and functional studies, human blood DCs and monocytes were reclassified into six different clusters, among which cluster 6 (DC6) mapped to pDCs [71]. Of relevance, a new DC subset characterized by AXL and SIGLEC6 expression was identified and designated as cluster 5 (DC5) AS-DCs. The AS-DCs were found in blood and lymphoid organs, but are absent in peripheral tissues, such as skin [81]. Based on gene expression analysis, AS-DCs exhibited a spectrum of states ranging from a pDC-like signature (e.g., IL3RA, IGJ, NRP1, MZB1) to a cDC2-like signature (e.g., IFI30, ITGAX, LY86, GLIPR2, FGR, LYZ, ENTPD1) suggesting a relationship between these subsets. By flow cytometry analysis, AS-DCs were found within both cDC (CD123lowCD11c+) and pDC (CD123+CD11c) gates [71]. Although CD123+CD11c AS-DCs expressed pDC markers (e.g., CD123 and CD303), they are functionally distinct from pDCs. As compared to IFN-α producing pDCs, AS-DCs are more potent activators of T cells. Interestingly, the previously described CD2high pDC subset [69, 70] most likely correspond to AS-DCs in terms of phenotype (i.e. CD2, AXL, CX3CR1, LYZ and CD86 expression) and regulatory functions (i.e. naïve T cells activation and proliferation) [71] (Fig. 2). Therefore, some functions previously attributed to pDCs could be due to contaminating AS-DCs and functional studies should be performed by excluding Axl+ DCs. Finally, the identity and developmental trajectory of AS-DCs [72] is still a matter of debate and further studies will be required to determine whether Axl+ DCs constitute a distinct DC subset [71] or are circulating DC progenitors [83].

Fig. 2
figure 2

The human circulating pDC subsets. Human pDCs are defined as Lin CD11c CD123/IL3R+ BDCA-2/CD303+ BDCA-4/CD304+ cells and designated to IFN-α production. Upon stimulation with TLR7/9 ligands, three human pDC subpopulations were identified as PD-L1+CD80 pDCs (P1) specialized in I-IFN production, PD-L1CD80+ pDCs (P3) specialized in adaptive immune functions, and PD-L1+CD80+ pDCs (P2) showing both innate and adaptive immune functions. In unstimulated/basal conditions, no pDCs subsets have been detected. The CD2+ pDCs subset most likely corresponds to a new DC cluster, named AS-DC, that is identified as AXL+LYZ+SIGLEC6+ DCs exerting regulatory functions, such as inducing T cells activation and expansion. The expression of AXL and LYZ was also previously observed on the CD5+CD81+ subset among CD2+ pDCs. Created with BioRender.com

Fig. 3
figure 3

Tissue pDCs are mostly negative for CD2, CD5, and PD-L1. Sections are from reactive lymph nodes (LN) (a-f; representative images from 2 out of 5 cases analyzed) and cutaneous lupus erythematosus (LE) (g-i; representative images of 1 out of 3 cases analyzed) and stained with TCF4/E2.2, CD2, CD5, and PD-L1, as labeled. Magnification 400x; scale bar 50 µm

In recent years, pDC heterogeneity has been revealed as a wide range of diversification upon stimulation [73,74,75,76]. Three divergent human pDC subpopulations were identified upon activation in response to a single innate stimulus, based on programmed death-ligand 1 (PD-L1) and CD80 expression [73] (Fig. 2). These subpopulations showed distinct functional features associated with specific transcriptional signatures. The P1 pDCs (PD-L1+CD80) displayed plasmacytoid morphology and were specialized in I-IFN production, the P3 pDCs (PD-L1CD80+) exhibited dendritic morphology and performed regulatory immune functions (i.e. T cell activation and T helper 2 (Th2) differentiation), and the P2 pDCs (PD-L1+CD80+) represented an intermediate subset [73]. Although their proportions varied among donors, P3-pDCs appeared to be less abundant than P1-pDCs and P2-pDCs. Moreover, the P1-pDC and P3-pDC phenotypes were regulated by different kinetics, being the first and the last to appear after stimuli, respectively. However, the activated pDC populations displayed a phenotypic stability, with or without a second viral challenge, thus indicating that P1, P2, and P3 pDCs are stable cellular entities in diverse functional states. P1-pDCs were prevalent in IFN-α-mediated autoimmune diseases (e.g., lupus and psoriasis) [73], whereas P3-pDCs were increased in melanoma and negatively impacted clinical outcomes [84]. Therefore, CD80 and PD-L1 surface markers can be used as biomarkers to track pDCs with innate or adaptive functions in pathological conditions. However, it should be noted that in reactive lymph nodes and cutaneous lupus erythematosus most pDCs lack PD-L1 (Fig. 3) as well as E2-2/TCF4+ BPDCN neoplastic cells [85].

Subsequently, a single-cell analysis using a droplet-based microfluidic platform revealed that TLR-induced IFN-α production by pDCs closely depends on stochastic gene regulation and is amplified by I-IFN paracrine signals in the microenvironment [74]. Only a limited pool (1–3%) of pDCs responded promptly to TLR7/8 and TLR9 agonists (i.e. first responders) and were defined by high surface levels of PD-L1 and TRAIL. The early responders triggered the surrounding pDCs to generate robust I-IFN responses (i.e. second responders) by inducing specific factors [75]. pDC diversification and functional specialization was independent of preexisting heterogeneity within steady-state pDCs, but intercellular crosstalk might be involved in the pDC activation process [73, 74]. Furthermore, distinct and stable pDC populations have been identified in REX3 transgenic mice based on CXCL10 expression, a downstream effector of I-IFN signaling, following TLR7 stimulation. The CXCL10+ and CXCL10 pDC populations were defined by different transcriptional programs; however, no current markers are able to define CXCL10 expression potential [76].

Finally, a recent study integrating single-cell RNA sequencing and proteomic data revealed the existence of specialized clusters among human pDCs at baseline or upon activation with influenza virus [86]. Of relevance, this study clearly demonstrated that a single cluster of pDCs is accountable for the majority of induced cytokines, including I-IFNs and III-IFNs [86]; however, no surface markers could discriminate these pDC clusters.

The heterogeneity within human pDCs reflects their competence in orchestrating diverse immune functions, such as effectors of viral responses and drivers of inflammation, activation of adaptive immune cells, and immune tolerance. Nevertheless, these investigations were mainly limited to the healthy circulating pDCs and additional data are needed on human tissue pDCs as well as in the setting of autoimmune diseases and cancers.

pDCs and cancer immune surveillance

pDCs migration toward tumor sites

In homeostatic conditions, fully differentiated pDCs migrate from the bone marrow towards primary and secondary lymphoid tissues via high endothelial venules (HEV) [87]. In inflammatory diseases and cancers, human pDCs can be also found in peripheral non-lymphoid tissues [2]. Fully differentiated pDCs isolated from human blood highly express a set of chemokine receptors, including CCR7, CCR2, CCR5, CXCR4, and CXCR3, weakly express CCR1, CCR4, and CXCR2, whereas CCR6, CXCR1, and CXCR5 are absent on pDC subset [88]. The pDCs migrate in response to the stromal cell–derived factor 1 (SDF-1/CXCL12), a homeostatic chemokine expressed by HEV [88, 89], but also detected in melanoma [43, 90], oral squamous cell carcinoma (OSCC) [91, 92], ovarian cancer [93], breast cancer and metastatic lymph nodes [94, 95], underlining its role in pDC recruitment to tumor sites [87, 88, 93]. The functional role of CXCR3 in chemotactic response by pDCs is still controversial, and it has been previously reviewed [2, 96]. Of relevance, CXCR3 ligands released during inflammation (e.g., Mig/CXCL9 and ITAC/CXCL11) synergize with the constitutive chemokine CXCL12 to induce pDC recruitment. The adjacent and simultaneous co-expression of CXCL12, CXCL9 and CXCL11 could regulate the recruitment of pDCs at sites of inflammation [87].

Peripheral blood pDCs in stage I-III melanoma patients upregulated CCR6, a skin homing receptor, and were recruited to primary cutaneous melanoma lesions, in response to CCL20 produced in melanoma microenvironment [97]. The IL-3 growth factor is able to upregulate CCR6 expression on human blood pDCs [98]; however, it is so far undetermined whether IL-3 or other factors produced by cells of the TME are responsible for CCR6 upregulation by melanoma-associated pDCs. On the contrary, CCR6 expression was low on circulating pDCs from metastatic melanoma (MM) patients (stage IV, AJCC) and comparable to control pDCs [98, 99]. The small subset of blood pDCs expressing both CCR6 and CCR10 might originate from lymph nodes and represent a subset in transit to the inflamed epithelia [98]. Furthermore, CCR7 triggers pDC migration towards CCL19 and CCL21, especially upon activation with TLR ligands, contributing to the pDC homing to draining lymph nodes both at steady state and in inflammatory conditions [82]. Our group has also identified chemerin/RARRES2, the natural ligand of Chemerin Receptor 1, as a new chemotactic factor for human pDCs transiting toward lymphoid tissues and inflamed skin (i.e. lupus erythematosus and psoriasis) [100]. Chemerin is usually expressed by keratinocytes, fibroblasts and blood endothelial cells in inflamed skin and by HEV in reactive lymph nodes [2], but bioactive chemerin was also found in ascitic fluids secondary to ovarian carcinomas [101]. Despite many studies suggested a downregulation or loss of chemerin in various tumors compared to the normal tissue counterparts [102], upregulation of chemerin was found in gastric cancer [103, 104], mesothelioma [105], neuroblastoma [106] and cervical neoplastic lesions [107], suggesting that chemerin in the TME might have context-dependent effects on tumorigenesis and tumor progression.

Type I interferon production by pDCs is impaired by the tumor microenvironment

Type I IFNs are a class of proinflammatory cytokines (i.e. including 13 genes coding for IFN-α, and individual gene coding for IFN-β, IFN-ω, IFN-ɛ, and IFN-к) that have context-dependent functions on cancer immunosurveillance and immunoediting [108]. Depending on the dose and timing, and the downstream induced signatures, I-IFNs can foster or prevent tumor progression and immune evasion [109]. Specifically, robust, acute, and ultimately resolving I-IFN responses have been shown to participate in anticancer immunosurveillance and mediate prominent anticancer effects, such as direct cytostatic/cytotoxic activity on malignant cells, but also immunostimulatory functions. On the other hand, weak, sub-optimal, and chronic I-IFN signaling supports tumor progression and resistance to therapy by cancer cell-intrinsic effects (i.e. mediating cytoprotective activity, promoting epithelial-to-mesenchymal transition and stemness) and establishment of an immunologically exhausted TME [109]. pDCs are known as professional IFN-α producing cells and generate 10 to 100 times more IFN-α than other cell types upon activation of pattern-recognition receptors (PRRs). Specifically, TLR7 recognizes single-stranded RNA of viral origin and synthetic mimicking ligands [e.g. imidazoquinoline compounds, such as Imiquimod (IMQ) and Resiquimod (R848)]; whereas TLR9 recognizes microbial unmethylated CpG DNA sequences as well as CpG-rich oligodeoxynucleotides (CpG-ODNs). After TLR7/9 engagement in the early endosomes, the IFN-α response is induced through the cascade activation of the myeloid differentiation primary response protein 88 (MyD88) that interacts with the Interferon Regulatory Factor 7 (IRF7) [110, 111], as described elsewhere in detail [112, 113]. In recent years, different studies reported the capability of pDCs to sense cytosolic DNA through the cGAS–STING (stimulator of interferon genes) pathway eliciting potent I-IFNs production independently of TLR7/9 [114,115,116]. Briefly, cGAS interacts with double-stranded DNA (dsDNA) and induces conformational changes into 2’3’-cyclic GMP-AMP (cGAMP). The second messenger cGAMP then activates STING [117], which recruits the TANK binding kinase-1 (TBK1) that is phosphorylated and, in turn, phosphorylates and induces nuclear translocation of the Interferon Regulatory Factor 3 (IRF3) [118]. It should be noted that the IRF7-dependent I-IFN signaling pathway produces 100 times more cytokine than the IRF3-dependent I-IFN signaling pathway.

TA-pDCs could be functionally impaired by tumor-derived soluble factors released in the TME and then acquire an immunosuppressive profile [21]. Specifically, the pDC proficiency in producing IFN-α can be hijacked by immunosuppressive cytokines, oncometabolites, or ligands to inhibitory receptors expressed on the cell surface, thus supporting cancer progression [22] (Fig. 4). Among the large set of immunosuppressive molecules produced by tumor cells, TGF-β is one of the most studied soluble factors responsible for the functional inhibition of pDCs [119]. TGF-β has been detected in a wide range of cancers, alone [3, 119] or in combination with other immunosuppressive cytokines, such as IL-10 [120, 121], PGE2 [122] and TNF-α [121, 123]. Specifically, the synergy between these cytokines was accountable for the TLR7/9 signaling inhibition, including IRF7 downregulation, impairing the I-IFN production by pDCs [124, 125]. Accordingly, our group proposed a TGF-β-mediated functional impairment of TLR/MyD88-dependent signaling in melanoma-associated pDCs through IRF7 downregulation [126]. Wnt5a is an immunosuppressive molecule identified in the melanoma TME affecting pDC function [127]. As described in the previous section, HMGB1 produced by neoplastic keratinocytes was implicated in the tolerogenic switch of TA-pDCs and decreased the IFN-α secretion upon stimulation with TLR9 agonists [107]. A recent study has shown that tumor-derived pDCs were significantly dampened in IFN-α production upon TLR7/9 activation in HPV-negative HNSCC, but were functionally uncompromised in HPV-positive HNSCC [18]. The immunosuppressive cytokine milieu was rich in IL-10 and TNF-α in HPV-negative but not in HPV-positive HNSCC; the resulting functional impairment of tumor-infiltrating pDCs further supported the immunosuppressive TME by promoting the expansion of Tregs in the tumor tissue [18]. Moreover, virus-like particles have been found to activate tumor-infiltrating pDCs [128]. Although direct evidence is lacking, pDC-tumor cell crosstalk might differ between HPV + and HPV- tumors. Finally, KSHV can infect and activate human pDCs through TLR9 signaling, inducing the upregulation of CD83 and CD86 and IFN-α secretion [129]. However, weak human myxovirus resistance protein 1 (MxA) expression was measured on KS cases, suggesting that I-IFN production by pDCs was suppressed probably in relation to KSHV's ability to evade the immune system [130].

Fig. 4
figure 4

The tolerogenic and immunostimulatory roles of pDCs in the tumor microenvironment. Graphical illustration of the human pDCs polarization in cancer. pDCs can exert anti-tumor and immunostimulatory functions (left panel), whereas tolerogenic pDCs promote the immune evasion of tumor cells (right panel). Tolerogenic pDCs are depicted as functionally impaired (e.g. I-IFN production) cells by tumor-derived cytokines and oncometabolites. Tolerogenic pDCs are further characterized by expression of costimulatory receptors or coinhibitory molecules and generate an immunosuppressive and inflammatory TME (e.g. Treg expansion). On the contrary, properly activated pDCs (e.g. via TLR7/9 agonists or STING agonists) produce type I and type III IFNs and proinflammatory cytokines (e.g. CXCL10), eliciting the recruitment of adaptive immune cells. Human pDCs can exert direct effector functions against tumor cells, as well. The surface molecules and intracellular mechanisms involved in these functions are detailed. Created with BioRender.com

Human pDCs express a large set of activating or inhibitory receptors that regulate the amplitude of the I-IFN response and activation state. The inhibitory receptors include the BDCA-2, the immunoglobulin-like transcript 7 (ILT7, also known as LILRA4 and CD85g), the C-type lectin dendritic cell immunoreceptor (DCIR, also known as CLEC4A), and the natural cytotoxicity receptor NKp44 (also known as NCR2) [131,132,133,134]. The regulation of I-IFN secretion by these inhibitory receptors restrain autoimmunity but promote tumor growth. Tumor cells express different ligands that could engage inhibitory receptors expressed on pDCs contributing to pDCs exhaustion and poor antitumor response. The galactose-terminated asialo-oligosaccharides, a glycoprotein component also found on tumor cells, have been identified as natural ligands of BDCA-2 [135]. More recently, galactose-terminated glycans binding to BDCA-2 have been detected [136]. Consistently, the Caroline Aspord’s research group has recently demonstrated that tumor cells harbor a specific glycosylation pattern. Specifically, melanomas were enriched in glycans with galactose residues eliciting an inhibitory impact on pDC functionality (i.e. IFN-α production upon stimulation with R848) [137]. Moreover, the plasma-derived heparin has been identified as specific ligand for BDCA-2 resulting in the inhibition of TLR9-driven IFN-α production in human pDCs. In parallel, an activation-dependent soluble form of BDCA-2 has been identified in human plasma and functions as heparin antagonist leading to enhanced TLR9-driven IFN-α production in pDCs [138]. This very novel observation should be integrated in the emerging oncology chapter on the role of heparin and derived-molecules in the treatment of cancer patients [139]. Furthermore, BDCA-2 has been proposed as a novel target for therapy in autoimmune diseases (i.e. systemic lupus erythematosus) [140, 141] and monoclonal antibodies targeting BDCA-2 could represent a treatment option in patients with hematologic malignancies [140,141,142]. Moreover, the antibody–drug conjugates targeting BDCA-2 could be developed as immunomodulatory therapy to achieve disease control in solid tumors infiltrated by tolerogenic pDCs [141]. The ligand of ILT7 has been identified as bone marrow stromal cell antigen 2 (BST2, also known as CD317) widely expressed in human tissues to varying degrees and reported in several human cancer cell lines, as well as in a wide range of tumors [143,144,145]. BST2 expression has been associated with cancer progression and invasiveness [146,147,148]. The crosslinking of ILT7 and BST2 strongly inhibits the production of IFN-α and proinflammatory cytokines (e.g. TNF-α) by pDCs [143, 149, 150]; however, the role of this interaction in cancer is still unclear. pDCs have also been reported to express the vasoactive intestinal peptide (VIP) receptors VPAC1 and VPAC2, and their crosslinking inhibited IFN-α secretion after CpG stimulation [151].

Finally, cancer cells have a high metabolic activity responsible for nutrients (i.e. glucose) deprivation in the TME. The increased glycolytic rate leads to lactate accumulation and environmental acidification in the tumor milieu [152]. As thoroughly described in the following section of this review, pDC functionality has been linked to glycolysis in order to provide the high energy demand for rapid I-IFN production [153]. Our group has demonstrated that IFN-α production by melanoma-exposed pDCs was hijacked and could derive from their metabolic drift [126] (Fig. 4).

pDC effector functions in human cancers: a still unexplored field

In addition to their pivotal role in I-IFN production, pDCs could execute direct cytoytoxic functions (Fig. 4), though their role as antitumor effector is still largely unknown and controversial.

In mouse models of melanoma and breast cancer, activated pDCs can directly kill tumor cells through TRAIL and GrB expression [15, 154] and were also able to recruit and activate NK cells and cross-prime cytotoxic T lymphocytes (CTLs) [155], leading to tumor clearance; however, the direct antitumor effector functions of human pDCs should be explored. After TLR7/9 engagement, activated pDCs could exert killing capacities via the upregulation of cytotoxic molecules, such as TRAIL [14, 156]. TRAIL+ pDCs target TRAIL-receptor (TRAIL-R) TRAIL-R1/DR4 or TRAIL-R2/DR5 expressing cells [14, 157]. TRAIL has been detected at high levels in the cytoplasm of unstimulated pDCs, suggesting that stimulation with TLR7 ligands (i.e. ssRNA molecules) induced the re-localization of TRAIL to the cell membrane [158]. Human pDC cell line GEN2.2, stimulated with TLR7/9 agonists, is able to lyse tumor cells in a TRAIL-dependent manner [156]. The exposure of pDCs to ssRNA or dsRNA viruses (e.g. HIV, HCV, Dengue virus) induced IFN-α production as well as TRAIL expression [159,160,161], sustaining the possible TRAIL-mediated cytotoxic role of pDCs in response to viral infections. Significantly, cytotoxic pDCs could directly neutralize tumor cells. For instance, in patients with basal cell carcinoma, pDCs became competent in killing tumor cells through a cell-to-cell TRAIL-dependent interaction after stimulation with either IMQ, CpG, or IFN-α [14, 162]. At the same time, TA-pDCs could induce local antitumor effector responses through the activation of T cells and NK cells [155, 163], a process mainly mediated by I- and III-IFN production [7, 164]. In a neuroblastoma co-culture system, the TLR9-activated pDCs induced the activation of TRAIL+ NK cells that ultimately killed tumor cells [165]. The depletion of either pDCs or IFN-α led to a loss of the TRAIL-mediated tumor cell killing by CD14+ monocytes [166].

A second cytolytic molecule that plays a central role in the pDC effector functions is the serine protease granzyme B [15, 167, 168] (Fig. 4). GrB is a pro-apoptotic molecule constitutively expressed in human pDCs [169], but its production and release is further induced by cytokines, such as IL-3, IL-10 and IL-21 [168, 170], abundantly expressed in cancer tissues [17, 171, 172]. Surprisingly, activation of pDCs through TLR7/9 agonists and CD40 ligand negatively regulated GrB expression [17], indicating that IFN-α producing pDCs might not be able to exert cytotoxic function. On the other hand, GrB-expressing pDCs have been involved in the suppression of T cell expansion by inducing T cell apoptosis or by hijacking the T cell proliferation in a GrB-dependent and perforin-independent manner [17, 169, 173]. Specifically, GrB levels produced by pDCs inversely correlated with the proliferation of co-incubated T cells in vitro, whereas T cell proliferation was enhanced by administrating anti-GrB neutralizing antibody or a specific substrate inhibitor [169]. pDC-derived GrB activity might also regulate in vivo T cell responses. Functionally, IL-3-activated pDCs delivered GrB to T cells in a cell-contact dependent manner and degraded the T cell receptor (TCR)-zeta chain, which is a substrate for GrB proteolytic activity [169, 174]. These data suggested that GrB+ pDCs in the TME could participate in suppressing the expansion of tumor-specific T cells. In conclusion, GrB-expressing pDCs could have regulatory effects on tumor-specific T cells as well as cytotoxic potential toward virus-infected or transformed cells, implying a contribution to cancer progression or elimination, respectively. However, the expression of GrB in tumor-infiltrating pDCs has been poorly investigated so far and further studies are required to understand whether GrB-secreting pDCs play a role in the immune evasion of cancers.

Tolerogenic pDCs in human cancer

pDC antitumor functions can be hijacked by TME that rewires pDCs as tolerogenic cells, thus promoting immune evasion. In the latter case, TA-pDCs instruct an immunosuppressive TME by ligands of costimulatory receptors or coinhibitory molecules expression (i.e. IDO, ICOS-L, PD-L1) [175] and Treg induction [176, 177] (Fig. 4).

The inducible co-stimulator ligand (ICOS-L) is expressed on antigen presenting cells (APCs) and tumor cells and binds to ICOS on activated T cells. The ICOS/ICOS-L interaction generates a pro-tumorigenic response mediated by local Treg expansion and IL-10 production by Tregs and Th2 in the TME [178, 179]. An increased expression of ICOS-L by pDCs has been reported in hepatocellular carcinoma (HCC) [180] and ovarian carcinoma [181], in which the presence of TA-pDCs and ICOS+ Tregs was predictive for disease progression [182]. In breast carcinoma, the ICOS-ICOS-L interaction promoted the expansion of Tregs and subsequent IL-10 and TGF-β secretion [183, 184]. A recent study, using a co-culture system of pDCs and cervical/vulvar neoplastic keratinocytes, proposed a role for ICOS-L+ pDCs in the local Treg expansion throughout cervical cancer progression [107], a mechanism mediated by the secretion of HMGB1 by neoplastic keratinocytes. The combination of ICOS and cytotoxic T-lymphocyte antigen 4 (CTLA-4) or PD-1/PD-L1 blockade could generate antitumor effects, particularly by preventing the interaction between Tregs and ICOS-L+ pDCs [178]. In CM context, pDCs expressed ICOS-L together with tumor necrosis factor ligand superfamily member 4 (TNFSF4; also known as OX40-L), modulating T cell response and Th2 polarization [185, 186]. In contrast, TA-pDCs in HNSCC and tumor-draining lymph nodes (TDLNs) that express high levels of OX40 have been demonstrated to synergize with myeloid DCs to induce a potent tumor antigen-specific CD8+ T cell response [187].

The indoleamine 2,3-dioxygenase (IDO) is expressed by tolerogenic pDCs and is responsible for the activation of Tregs. Melanoma-associated human pDCs released IDO promoting tumor escape from immune surveillance [188]. IDO-expressing pDCs were also detected in mouse TDLNs, where they can directly activate mature Tregs through PD-L1 [189]. As documented in HCC [190] and HNSCC [191], hypoxia is an environmental factor that induced IDO upregulation by pDCs and thus their tolerogenic state. Particularly, in HCC the IDO upregulation by pDCs was mediated by HIF-1α/CCL20/STAT1 pathway, promoting tumor tolerance and metastatization [190]. Moreover, by targeting fatty acid-binding protein 5 (FABP5), a cellular chaperone of long-chain fatty acids involved in metabolism of lipids, IDO expression on TA-pDCs was reduced and impaired the generation of FOXP3+ Tregs in the TME [192].

Although the angiogenic properties of pDCs have been poorly explored as compared to cDCs, pDCs could exert a tumor-promoting role by inducing angiogenesis [193]. Despite the fact that in vitro pDCs were unable to produce vascular endothelial growth factor (VEGF) [194], TA-pDCs in ovarian carcinoma were shown to produce pro-angiogenic and pro-invasive cytokines, such as TNF-α and CXCL8, particularly after CD40L activation [195]. In addition, IL-1α produced by TA-pDCs was found to be responsible for promoting cell proliferation and angiogenesis in non-small cell lung cancer (NSCLC) [196].

The prognostic role of tumor-associated pDCs

TA-pDCs have been detected at the primary tumor sites and in TDLNs of various cancer types (Fig. 5). Moreover, the density of TA-pDCs and the frequency of peripheral blood pDCs fluctuate among different human neoplasia and during disease progression (Table 1). As previously reviewed [3], TA-pDC accumulation was associated with a poor outcome in different cancer, such as BC [183, 197], ovarian carcinomas [182, 198], HNSCC [22] and OSCC [124, 199]. TA-pDCs were found in colon and colorectal carcinomas (CRC) [200,201,202], but they were associated with a different pathological stage and prognostic role [200, 201]. High numbers of pDCs were also found within HCC [203, 204] and this was correlated with greater vascular invasion, advanced N stage, higher recurrence rate, shorter OS and with FOXP3+ Tregs infiltration [203, 204]. In NSCLC [196, 205] and gastric cancer [206, 207], the elevated frequency of circulating and TA-pDCs was correlated with poor clinical outcome. Interestingly, pDCs infiltrating NSCLC facilitated tumor growth through the production of high levels of IL-1α in an AIM2-dependent manner [196]. By contrast, in lung adenocarcinoma (LUAD) patients with a smoking history, TA-pDCs were enriched and associated with favorable clinical outcomes [208]. The DNA-damaging anti-cancer treatments (i.e. ionizing radiation and DNA-alkylating agent), inducing the accumulation of tumor-derived DNA, might be involved in the recruitment of activated pDCs to the TME, highlighting the TLR9 upregulation by pDCs in mice treated with cisplatin or ionizing radiation [208]. Moreover, in a cohort of 288 patients with follicular lymphoma, the presence of pDCs was associated with increased OS [209]. PDCs were also present in the skin lesions of Kaposi's sarcoma (KS), associated with KS herpesvirus (KSHV, also known as HHV8) infection, but they were significantly less abundant as compared to inflamed Molluscum contagiosum cases [130]. pDCs were recruited to cervical carcinomas and OSCC after chronic HPV infection during the early steps of malignant transformation, but were absent before the infection, suggesting a potential role in antiviral response against HPV and favouring the clearance of pre-neoplastic lesions [107, 199, 210]. In CM, pDCs were found in the primary tumor lesions, whereas they were close to absent in nevi and normal skin [43, 99], and their high density predict a poor prognosis [186, 211]. The frequency of circulating pDCs was reduced in CM patients compared to healthy donors (HDs) [211], but their homing to sentinel lymph nodes was preserved [90, 99]. Moreover, our group demonstrated the collapse of tumor-infiltrating as well as circulating compartment of pDCs during melanoma progression [3, 99, 212]. Interestingly, an increase in pDC recruitment was observed in BRAFV600E melanomas compared to BRAF wild-type melanomas [99, 213]. In silico analysis of a pDC signature in pan-cancer TCGA datasets showed that a high pDC content was limited to TDLNs and stage I primary tumors, whereas pDC signature was poor in CM as compared to primary carcinomas [99, 126]. Instead, the pDC-specific signature has been significantly associated to a favorable survival outcome in the EBV-associated nasopharyngeal carcinoma, suggesting the potential role of pDCs in inducing antitumor immune responses [214]. Reduced frequency of circulating pDCs has been demonstrated in bladder cancer [215], CRC [216], pancreatic cancer [217], BC [218], and ovarian carcinoma [181] patients as compared to HDs and throughout disease progression. pDC frequency is reduced in acute lymphoblastic leukemia as well, which would be gradually restored after chemotherapy [219].

Fig. 5
figure 5

PDCs infiltrate primary tumors and nodal metastasis of various human cancer types. BDCA-2 staining on sections from primary [non-small cell lung cancer (NSCLC, a), head&neck squamous cell carcinoma (HNSCC, b), high grade endometrial carcinoma (HGEC, c), primary cutaneous melanoma (PCM), d] and nodal metastasis (metLN) [breast carcinoma (BC, e), cutaneous melanoma (CM, f)]. Magnification 400x; scale bar 50 µm

Table 1 The clinical significance of tumor-associated and circulating pDCs in human cancers

In pan-cancer TCGA datasets, the signatures of pDC and I-IFNs were not correlated, suggesting that pDCs might be partly dysfunctional in I-IFNs production in the TME [126]. The above-mentioned studies are limited to immunohistochemistry or flow cytometry approach and lack functional data on the role of pDCs in the TME (Table 1). These limitations could explain the wide heterogeneity that has emerged in pDC frequency in the blood and tissues of cancer patients and its correlation with clinical outcome. These caveats could be overcome by using valuable tools for a spatial resolution of cancer tissues (i.e. multiplex imaging, spatial transcriptomics).

Metabolism and pDCs immune escape

Cell metabolism control of innate immune functions of pDCs

In recent years, the subject of cell metabolism in the regulation of innate and adaptive immune cell response, defined as immunometabolism, has assumed increasing importance [220, 221]. The main metabolic pathways involved in pDC homeostasis and functions are glycolysis, oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO).

It has recently been demonstrated that, upon TLR stimulation, DCs underwent metabolic reprogramming, which was critical for their activation (i.e. cytokines production) and innate immune functions [222, 223]. Particularly, TLR agonists (i.e. LPS, R848, CpG-B) induced a rapid increase in the glycolysis in DCs differentiated from bone marrow in the presence of the GM-CSF (GM-DCs) [224]. Interestingly, in GM-DCs, the glycolysis fueling was linked to CCR7 oligomerization to promote cellular motility and homing to draining lymph nodes [225]. This mechanism could also be applied to pDCs that rely on CCR7 expression to migrate in response to CCL19/CCL21 [82]. Moreover, rapid I-IFN production by pDCs demands high energy from glycolysis [153, 226]. An increase in hypoxia inducible factor alpha (HIF1α) expression has been observed together with enhanced glycolysis and IFN-α production [226, 227] in both activated primary human pDCs and GEN2.2 cell line. In fact, HIF1α is involved in the modulation of glycolysis by increasing the expression of the glucose transporter 1 (GLUT1) in Hep-2 human cells [228]; however, this mechanism has to be further investigated in pDCs. Moreover, the inhibition of glycolysis in pDCs reduced IFN-α production as well as the expression of HLA-DR and co-stimulatory molecules CD80 and CD86 [226]. These results underline the crucial nature of glycolysis for the antiviral function of pDCs (e.g. IFN-α production), but also for pDC-mediated T cell activation. Interestingly, the inhibition of FAO and mTOR pathway in pDCs resulted in a limited expression of CD40 and CD86 [229, 230], suggesting that appropriate cell metabolism is pivotal in the regulation of the mature state of pDCs. Finally, the autocrine/paracrine IFN-α feedback loop sustained FAO that, in turn, was able to support the OXPHOS in pDCs stimulated with CpG-A [230]. TLR7/8 agonists upregulated the OXPHOS and glutamine metabolism in pDCs, leading to a higher production of IFN-α and an increased T cell response [231]. In fact, the inhibition of both OXPHOS and glutamine metabolism prevented autophagy-dependent pDC activation [231]. The amino acid (AA) metabolism is important for sustaining mitochondrial respiration in DCs, as described in a recent review [232]. Moreover, several AA transporters were involved in the mTORC1 complex activation after TLR9 stimulation, resulting in I-IFN and chemokine production by pDCs [233].

Monocyte-derived DCs (moDCs) engage both TLRs and RLRs upon viral encounter, whereas pDCs use the RIG-I pathway only in the later stages of antiviral responses. Therefore, RIG-I-mediated I-IFNs response requires less energy as compared to TLR signaling and was supported by OXPHOS [227]. The inhibitors of mitochondrial electron transport chain also impaired IFN-α production by TLR7/9 activated pDCs, highlighting the importance of OXPHOS in the regulation of innate pDC functions [234]. In contrast with previous published data [226, 227], Hurley et al. did not find evidence of the central role of glycolysis in sustaining pDC functionality; however, glycolysis and FAO were both involved in IFN-α production by pDCs.

To date, the literature on pDC metabolism is still partially contradictory likely due to different culture methods for pDCs and the use of distinct TLR agonists or viral particles to stimulate them.

Oncometabolite and nutrient deprivation impair pDC functions

Immunometabolism has revolutionized the tumor immunology landscape by unveiling how to manipulate metabolic pathways and promote antitumor immune responses [221]. Despite the increasing interest in immunometabolism as a novel strategy for cancer immunotherapy, the metabolic alterations of pDCs in cancer have been poorly investigated. Cancer cells have a high glycolytic activity that reduces glucose availability in the TME, with a subsequent lactate accumulation and acidification of the extracellular environment, known as the Warburg effect [152, 235]. Metabolic pathways are able to regulate the innate and adaptive immune responses to activation signals, including TLR agonists [236, 237] (Fig. 6). Hence, the in vitro and in vivo extracellular acidification can subvert pDC functionality, specifically by inhibiting their IFN-α production [238]. The inhibitory process is mediated by the interaction of lactic acid with lactate monocarboxylate transporters (MCT-1/2) and cell surface G-protein coupled receptor (GPR81) expressed on the pDC surface [238]. Lactate accumulation in the TME is responsible for the tolerogenic switch of TA-pDCs, particularly by eliciting IDO-dependent Treg generation [238]. Moreover, as described in the previous section, the hypoxic TME in HCC [190] and HNSCC [191] induced IDO upregulation by pDCs and thus their tolerogenic state. In particular, the pDC recruitment to the tumor site and upregulation of IDO were mediated by HIF-1α/eADO/ADORA1 signaling [190, 204]. All these data substantiated the idea that TME negatively affects pDC metabolism (i.e. glycolysis and OXPHOS) and their innate immune functions (Fig. 6).

Fig. 6
figure 6

Tumor microenvironment negatively affects the cell metabolism and functions of human pDCs. Graphical illustration of the tumor microenvironment impact on pDCs metabolism. The IFN-α production by pDCs required high-energy demand from glycolysis. Cancer cells have a high glycolytic metabolism leading to both glucose deprivation (brown dots) and lactate accumulation (blue dots) with subsequent acidification of the extracellular milieu establishing an unfavourable environment directly hijacking the glycolytic capacity of pDCs. The inhibition of glycolysis could be also explained by the imbalance of import/export of lactic acid, mediated by the transporters MCT1/2 and the GPR81 receptor, leading to intracellular pH acidification. Moreover, oxygen deprivation results in HIF1α and GLUT1 increased expression on pDCs, supporting glycolysis rather than OXPHOS. Finally, the lactate accumulation and hypoxia modulate IDO expression by TA-pDCs and their tolerogenic switch. Solid lines indicate well-documented mechanisms; dashed lines indicate mechanisms based on limited evidences. Created with BioRender.com

Our group has previously demonstrated that CM secretome supported the collapse of circulating and TA-pDCs in advanced metastatic disease, with a potential role in lactic acidosis [99, 212]. We found that the lactate concentration was inversely associated to the concentration of glucose in melanoma-derived supernatants [212]. Mechanistically, in vitro exposure to melanoma soluble factors or lactic acidosis induced an IFN-α-defective tolerogenic state on fully differentiated pDCs [99, 126, 212]. Our recent data suggested that glucose deprivation and lactic acidosis by melanoma cells hindered glycolytic metabolism in pDCs, resulting ultimately in a detrimental effect on their IFN-α production [126, 212, 235] (Fig. 6). Accordingly, pDCs exposed to CM supernatants were characterized by metabolic drift and tolerogenic state [126]. Specifically, these data indicated a reduced ability of melanoma-conditioned pDCs to switch towards glycolysis to meet the cell energy demands [126]. In conclusion, the dysregulation of pDC metabolism in the TME could lead to defective antitumor function, including IFN-α production by pDCs.

Modulating interferon production by pDCs: where do we stand?

Immunotherapies triggering an anticancer immune response remodel the tumor-infiltrating immune cells landscape. Immune checkpoint blockades (ICBs) have revolutionized the clinical management of various cancers. However, a consistent proportion of patients is refractory to ICB monotherapy and combination of double, or triple, ICB therapies lead to considerable side effects. Various immunostimulatory adjuvants, including agonists of PRRs (e.g. TLRs) or of their adapters (e.g. STING), have been developed to enhance the anti-cancer immunity induced by ICBs [239] and to overcome the resistance to systemic ICB therapies.

Preclinical studies have demonstrated that the recruitment of pDCs into neoplastic tissues and their activation represent a promising immunotherapeutic approach for various types of cancer [15, 154, 240,241,242]. Accordingly, therapeutic strategies to exploit the ability of pDCs to modulate tumor-specific T cell responses and cytotoxic functions have achieved promising antitumor effects in phase I/II clinical trials. To this end, two cancer immunotherapeutic strategies could be employed to enhance the immunostimulatory properties of pDCs: i) directly activating pDCs in vivo by targeting PRR signaling pathways; ii) exploiting autologous pDCs from peripheral blood or allogeneic pDC cell lines as cell therapy by developing novel pDC-based antitumor vaccines. Human pDC line derived from HLA-A*0201 leukemic pDCs (designated as pDC*line) showed the ability to initiate antitumor immune responses in humanized mice and in melanoma patients [243, 244].

Multiple clinical trials investigating the safety profile and therapeutic efficacy of PRR agonists, as monotherapy or in combination with chemotherapy, radiotherapy, or ICBs, have been started in the last decade (Table 2). However, only few clinical studies have been published at this time, but some of them have posted accessible results on clinicaltrials.gov.

Table 2 Clinical Trials investigating the TLR-7/9 agonists and STING agonists in human cancers

Immunotherapy strategies based on TLR7/8 and TLR9 agonists

Preclinical data have demonstrated that administration of TLR7/8 and TLR9 agonists in combination with current anticancer regimens boosts the local and systemic antitumor immune response, promotes tumor cell killing and contributes to better outcomes, providing the rationale to use TLR agonists as adjuvants with ICBs [245,246,247,248,249]. Current efforts to use TLR agonists in clinical trials are largely focused on TLR7/8 and TLR9 agonists (Fig. 7), resulting in improved antitumor immunity mainly mediated by pDCs activation.

Fig. 7
figure 7

Synthetic TLR7/8, TLR9, and STING agonists in clinical trials. The immunostimulatory adjuvants can directly activate pDCs in vivo by targeting TLR or STING signaling pathways. Various TLR7/8, TLR9 and STING agonists are being used in multiple clinical trials, as monotherapy or in combination with ICB, to enhance anti-cancer immunity by exogenous activation of pDCs. IMQ is the first TLR7 agonist approved for the topical treatment of non-melanoma skin cancer. The TLR7/8 agonists include the second-generation (e.g. R848/resiquimod, BDB001, LHC165) and the third-generation (e.g. BDC-1001, NJH395) synthetic compounds. The unmethylated CpG oligodeoxynucleotides (CpG-ODN) are ligands of TLR9 and can be distinguished into three major classes: CpG-A (CpG ODN 2216, Vidutolimod/CMP-001), CpG-B (CpG ODN 1668, Tilsotolimod/IMO-2125, Cavrotolimod, CpG ODN 7909/PF-3512676, TAC-001), and CpG-C (SD101). Beyond natural cyclic dinucleotides (CDN), STING agonists include a variety of synthetic CDN (ADU-S100/ML RRS2 CDA, MK-1454, BMS-986301, BI 1387446, E7766, IMSA101, SB11285, SYNB189) and non-cyclic dinucleotides (non-CDN; MK-2118, GSK3745417, SNX281, TAK-676). Dashed lines indicate the potential binding of synthetic compounds to TLRs or STING. Solid lines indicate the well-known signaling pathways triggered upon TLR7/9 or STING activation. Created with BioRender.com

The Imiquimod (IMQ) was the first TLR7 agonist approved for the topical treatment of non-melanoma skin cancers (i.e. basal cell carcinoma) [250, 251]. The curative effects of IMQ have been linked to the activation of innate and adaptive antitumor immune mechanisms, in particular, by the recruitment of pDCs to the tumor site and trigger their cytotoxic functions [14, 162, 240]. These preclinical studies underlined the strong IMQ therapeutic potential. Specifically, the recruitment to and activation of pDCs in neoplastic tissues represent a promising immunotherapeutic approach for various types of cancer.

Several clinical trials have investigated topical IMQ in solid tumors (Table 2). Moreover, the combination of radiotherapy (RT) and IMQ generated an effective adaptive immune response, resulting in systemic control of metastatic breast cancer after local treatment of cutaneous metastases (NCT01421017).

Beyond IMQ, several synthetic TLR7/8 agonists showed promising immunostimulatory activity in preclinical models of solid cancers. These molecules have been further investigated for safety, tolerability, pharmacodynamics, pharmacokinetics, and efficacy profile as monotherapy or in combination with ICB, RT, or chemotherapeutics, in adults with advanced solid tumors (Table 2). However, none of these second- (e.g. R848) or third-generation (e.g. BDB001, BDC-1001, LHC165, NJH395) TLR7/8 agonists [252] (Fig. 7) has yet been approved by regulatory agencies for use in cancer patients. A novel injectable TLR7/8 dual agonist, 3 M-052, triggered innate immunity and activated systemic tumor-specific CD8+ T cell immunity, leading to the rejection of distant uninjected tumors without systemic cytokine release and toxicity [253]. However, pDCs and I-IFN were not indispensable for the antitumor activity of 3 M-052, although they are required for efficient tumor suppression [253]. NJH395 and BDC-1001 are novel immune-stimulator antibody conjugates (ISAC) combining an anti-HER2 antibody with TLR7/8 agonists aimed at delivering preferentially the immune activator into tumors and eliciting targeted immune modulation in the TME (i.e. IFN-I production), while lowering systemic exposure and minimizing immune-related toxicities [254], paving the way for combination treatment.

Among TLR9 agonists, unmethylated CpG oligodeoxynucleotides (CpG-ODN) can be divided into three major classes that differ in their molecular structure, endosomal trafficking, TLR9 downstream signaling, and pattern of immune cell activation: i) type A CpG-ODNs are mainly inducers of IFN-α production and activate pDC maturation, but weakly stimulate B cells; ii) type B CpG-ODNs strongly induce B-cell proliferation, cytokine production and have some effect on pDC maturation, but induce relatively scarce IFN-α secretion; iii) type C CpG-ODNs have intermediate immune stimulatory properties between type A and type B CpG-ODNs [255] (Fig. 7). Moreover, CpG-B and CpG-C possessed strong antitumor effects, but CpG-C was more rapid and effective than CpG-B in combination with the anti-PD-1 antibody [256].

Vidutolimod (formerly CMP-001) is a CpG-A ODN packaged within a virus-like particle composed of the bacteriophage coat protein Qβ [257]. Vidutolimod induced the production of anti-Qβ antibodies that formed immune complexes with virus-like particles, facilitating the uptake by pDCs expressing the costimulatory Fcγ receptor II [257]. Vidutolimod was specially designed to induce high levels of IFN-α, through activation of TLR9 in pDCs, which in turn induces a T helper 1 (Th1) response and has secondary effects on myeloid cells [128, 246, 257]. Additionally, in situ vaccination with CMP-001 triggered both local and abscopal antitumor immune responses [246, 249]. CMP-001 has been tested in combination with ICBs targeting CTLA-4 (i.e. ipilimumab), PD-1 (i.e. cemiplimab, nivolumab, or pembrolizumab) or PD-L1 (i.e. atezolizumab), alone or together with radiotherapy or surgery, in multiple neoplastic malignancies.

Tilsotolimod (also known as IMO-2125) is a type B CpG-ODN evaluated in co-treatment with ICB to test its efficacy in advanced solid tumors and melanoma [258]. Although, the ILLUMINATE-301 phase III study was terminated due to the lack of efficacy (NCT03445533). Interestingly, the administration CpG B-type ODN PF-3512676 (also known as CpG 7909) as immunostimulatory adjuvant therapy in stage I-III melanomas enhanced the activation of pDCs and CD1a+ mDCs, and lowered the frequency of immunosuppressive Treg, reducing the risk of metastatic spread in early-stage melanomas. In addition, the PF-3512676 administration was associated with increased I-IFN and proinflammatory cytokines, induction of activated CD86+ blood pDCs, and TRAIL+ IFN-induced monocyte-derived DC with a mature phenotype [259, 260].

Of relevance, SD-101 is a synthetic type C CpG-ODN able to stimulate human pDCs to release IFN-α, mature into efficient APCs and promote infiltration and expansion of CD8+ T cells expressing IFN-γ [261, 262], resulting in complete and long-lasting rejection of anti-PD-1 resistant tumors and effective systemic immunity against uninjected distant-site tumors [261, 262]. SD-101 has been studied in several clinical trials for the treatment of solid tumors, as monotherapy or in combination with pembrolizumab or IDO inhibitors alone or plus RT, or with anti-IL10 (Table 2).

Finally, the pDC cytotoxicity upon TLR7/8 or TLR9 agonists administration have been scarcely explored in clinical trials and future efforts should be designed accordingly.

The combination of ICB therapies with TLR7/8 and TLR9 agonists could turn “cold” tumors with an immunosuppressive microenvironment into immunologically “hot” tumors. Of note, preliminary studies suggest that TLR9 agonists might induce PD-1 surface expression on T cells and could enhance anti–PD-1 efficacy or reverse anti–PD-1 resistance [245,246,247,248,249] by enhancing systemic antitumor immune responses. For instance, PD-1 resistant melanoma patients could be eligible for this treatment [249]. Of relevance to this review, the IFN-α responsive genes in circulating leukocytes were measured as a surrogate of IFN-α production by TA-pDCs and were induced in all melanoma patients receiving i.t. SD-101 and pembrolizumab [248]. In responding patients, TA-pDCs were activated by vidutolimod to secrete IFN-α and induced Th1 antitumor immunity, including the rapid production of IFN-inducible chemokine CXCL10 and the generation of IFN-γ-secreting CD8+ T cells [249]. On the other hand, non-responding patients could have TA-pDCs resistant to TLR9 activation [5, 263], suggesting the need for early intervention during melanoma progression.

Critically, the expression of TLRs was also found on tumor cells. Particularly, TLR9 was strongly expressed on human tumor cell lines of different tissue-origin [264,265,266], including lung cancer [267], gastric carcinoma [268], cervical tumor [269], and prostate cancer [270], suggesting that TLR-signaling might modulate tumor development. However, the functional activity of TLR9 in tumor cells should be further investigated. The identification of the endogenous TLR ligands (i.e. nucleic acids released from dying cells) [271, 272] would clarify the mechanisms for tumor cell growth and potential off-target effects of TLR9 agonist administration. Moreover, one very recent report showed that NKp44+ pDCs physically interact with the platelet-derived growth factor D (PDGF-DD) expressing melanoma cells [273]. Therefore, PDGF-DD stimulation could enhance IFN-α secretion induced by the TLR9-mediated pDC response to self-DNA released by necrotic tumor cells. Finally, the expression of PDGF-DD by tumor cells should be taken into account in clinical trials using TLR9 agonists, such as CpG-ODN, as adjuvant therapy [273].

Controversial effect of STING agonists

In the TME, cancer cells are well-known to be repleted with cytosolic dsDNA derived from the rupture of micronuclear envelopes [274, 275], intrinsic DNA damage or exogenous genomic stress [275,276,277,278]. Tumor-derived DNA can activate the STING pathway at the level of cGAS in host immune cells or, alternatively, tumor-derived cGAMP can be transferred to host APCs thereby directly activating STING [279,280,281]. The activation of cGAS-STING pathway by tumor-derived dsDNA [241] has tremendous potential to improve antitumor immunity, playing an essential role in DC recognition of dying tumor cells, by generating I-IFN response and potently enhanced antitumor CTL responses [241, 242, 279, 282]. Although human pDCs could sense nucleic acids released from dying tumor cells [241, 279, 283], most TA-pDCs did not display endogenous activation. Notably, in melanoma, the endogenous activation of the cGAS-STING was limited to areas of spontaneous microscopic melanoma regression with a large number of tumor-infiltrating pDCs suggesting their activation [126].

Based on promising pre-clinical observations of host STING pathway involvement in endogenous local and systemic antitumor immune responses, the administration of STING agonists has been explored as a therapeutic strategy for the exogenous activation of pDCs resulting in prolonged survival and reduced tumor growth [242, 282, 284,285,286]. A variety of STING agonists, including natural or synthetic cyclic dinucleotides (CDN; e.g. c-di-GMP) and non-cyclic dinucleotides (non-CDN) with improved stability (Fig. 7), have been rapidly developed potentially to increase response rates to current immunotherapy approaches.

The flavonoid compound dimethyloxoxanthenyl acetic acid (DMXAA, also known as ASA404), a direct ligand for murine STING [287, 288], induced potent antitumor activity in mouse tumor models [242]. Several phase I-II trials have recently been initiated to investigate i.t. delivery of synthetic CDNs, such as ADU-S100, MK-1454, BMS-986301, BI 1387446, E7766, IMSA101, and SYNB189 (Table 2). Despite encouraging antitumor effects in preclinical models [242, 289], the phase I-II clinical trials of ADU-S100 (NCT03172936, NCT03937141, NCT02675439) and MK-1454 [290, 291] were terminated because no substantial antitumor activity was observed. The compounds BMS-986301, BI 1387446, and E7766 are currently in clinical testing in patients with advanced solid tumors that failed previous treatment, including ICB. Finally, non-cyclic dinucleotides (non-CDNs) include MK-2118, GSK3745417, SNX281, and TAK-676, that are being tested alone or in combination with pembrolizumab in patients with advanced solid tumors or lymphomas.

As previously described for TLR9 agonists, the activation of the cGAS-STING pathway is able to remodel immune-desert “cold” tumors into T cell-infiltrated “hot” tumors, by increasing antigen presentation, T cell trafficking and recruitment, and CTL functions. In addition, STING activation has been reported to induce upregulation of PD-L1. Therefore, combination therapies involving anti-PD-1/PD-L1 and STING are being evaluated in clinical trials (Table 2). However, the specific effects of STING agonists on the pDC-mediated antitumor response have been hitherto scarcely investigated.

In the oncology setting, it is also significant to consider the relevance of STING activation on cancer cells. The activation of cGAS-STING pathway in tumor cells can result in attenuated tumor growth, enhanced immunogenicity and susceptibility to lysis by tumor-infiltrating lymphocytes promoting tumor clearance. However, the selective pressure for tumor cells could decrease both cGAS and STING expression in human cancers protecting tumor cells [292]. Accordingly, defective STING expression has been reported in several melanoma or colorectal adenocarcinoma cell lines and in clinical cancer tissues, especially in advanced tumors [293,294,295,296]. On the other hand, emerging evidence suggested a pro-tumor role of the cGAS-STING pathway [297,298,299], which makes the clinical administration of STING agonists more challenging.

The combined immunotherapy of STING agonist with anti-PD-1/PD-L1 can neutralize the immunosuppressive effects of STING agonists (i.e. PD-L1 upregulation), delay tumor growth and protect against tumor rechallenge in mice [300, 301]. Based on this heterogeneity of outcomes, novel biomarkers for appropriate patient selection are mandatory to achieve a clinical response to this treatment.

pDC-based vaccines for cancer immunotherapy

Although the antigen-presentation capacity of conventional DCs (cDCs) is clearly defined, pDCs are generally attributed as having poor antigen-presentation function. The development of DC-based vaccines targeting tumor antigens that could be promptly cross-presented is a promising immunotherapeutic approach for cancer treatment. pDCs have the ability to present tumor antigens and prime tumor-specific cytotoxic CD8+ T cells [24, 25]. Human pDCs can expand antigen-specific CTLs in vitro. Depending on their activation state, pDCs can polarize proliferating T cells toward Th1, Th2, or Treg. However, we know very little about the antigen-presentation capacity of pDCs in vivo and their ability to elicit responses from T cells. Human pDCs are less efficient at presenting antigens as compared to cDCs, but whether they represent a distinct type of professional APC has yet to be clarified. Properly designed pDC-based immunotherapeutic approaches can boost tumor-specific immune responses and represent an attractive choice for treating cancer. Until now, most DC-based vaccination strategies are based on monocyte-derived DCs (moDCs) and cDC2s, with only a few clinical trials evaluating pDC-based vaccines. pDC-targeted vaccination elicited a strong cross-priming and durable CD8+ T cell response [27]. However, cross-presenting pDCs were unable to prime efficiently CD8+ T cells by themselves, requiring the cDC1s contribution [27]. Antigen transfer from pDCs to antigen-naïve cDCs for cross-priming was mediated by a unique mechanism of pDC-derived exosomes [27].

As previously reviewed [302], numerous clinical trials exploiting the antigen-presenting capacity of DCs have shown poor efficacy, probably due to immunosuppressive TME and the advanced disease stage of enrolled patients. A tolerogenic and dysfunctional immune phenotype in the TME could reduce the efficacy of DC-based vaccines.

Currently, peripheral blood autologous pDCs or allogeneic pDC cell lines derived from leukemic pDCs are tested for antitumor vaccine development [303, 304]. The first clinical trial that tested the therapeutic potential of pDC-based vaccine against malignant tumors was carried out in patients with MM expressing gp100 and tyrosinase (NCT01690377) [303]. Blood circulating pDCs were TLR-activated with FSME-IMMUN and loaded with melanoma-associated peptides before nodal injection. The activated pDCs displayed a mature phenotype and were able to migrate to lymph nodes. The upregulation of the IFN signature and tumor-specific T cell responses were also documented [303]. The vaccine was well tolerated, without evidence of severe toxicity, and improved PFS and OS in melanoma patients compared to controls receiving dacarbazine chemotherapy. A human pDC line derived from HLA-A*0201 leukemic pDCs was evaluated in a phase I clinical trial (NCT01863108) for the capacity to trigger antitumor responses in MM patients [304]. A similar approach was evaluated for lung cancer patients in a phase I/II trial (NCT03970746). A vaccine that combines the CD8+ T cell chemoattractive properties of pDCs with the superior tumor-antigen specific T cell priming capacity of cDC2s [305] was tested in patients with prostate cancer (NCT02692976), metastatic endometrial cancer (NCT04212377), castration-resistant prostate cancer (CRPC; NCT02692976), and metastatic endometrial cancer (NCT04212377). Functional antigen-specific T cells were detected in the peripheral blood of vaccinated patients and correlated with greater IFN-γ production and prolonged PFS [306]. Significantly, pDC-based cancer vaccines have been shown to increase the frequency of circulating antitumor T lymphocytes, together with the induction of I-IFN signature in patients with MM [303, 304, 307].

In some tumors, the administration of TLR agonists resulted in antitumor immune responses, combined with pDCs activation and clinical benefits suggesting that the development of pDC-based antitumor vaccines could represent an additional therapeutic option. The combination of pDC-based vaccination strategies with immune checkpoint inhibitors could further improve the clinical potential of pDCs [304].

Conclusions

pDCs have been detected in a wide variety of human malignant neoplasms with different clinical outcome. Modulation of their effector and regulatory functions represent a novel window of intervention. However, intrinsic pDCs heterogeneity in term of immune functions should be highly considered. Moreover, TA-pDCs have been associated with a tolerogenic phenotype and functional impairment induced by immunosuppressive TME. Novel combinatorial immunomodulatory therapies targeting pDC activation can boost innate and adaptive cancer immunity.

Availability of data and materials

Not applicable.

Abbreviations

2-DG:

2-Deoxyglucose

AML:

Acute myeloid leukemia

APCs:

Antigen presenting cells

BM:

Bone marrow

BPDCN:

Blastic plasmacytoid dendritic cell neoplasm

BST2:

Bone marrow stromal cell antigen 2

cDC1s:

Type 1 conventional dendritic cells

cDC2s:

Type 2 conventional dendritic cells

CDN:

Cyclic dinucleotides

CDP:

Common dendritic cell progenitor

cGAMP:

2’,3’-Cyclic GMP-AMP

cGAS:

Cyclic guanosine monophosphate-adenosine monophosphate synthase

CLP:

Common lymphoid progenitor

CLRs:

C-type lectin receptors

CM:

Cutaneous melanoma

CMPs:

Common myeloid progenitors

CpG-ODNs:

CpG-rich oligodeoxynucleotides

CRC:

Colorectal carcinomas

CTLA-4:

Cytotoxic T-lymphocyte antigen 4

CTLs:

Cytotoxic T lymphocytes

EBV:

Epstein-Barr Virus

FA:

Fatty acid

FAO:

Fatty acid oxidation

Flt3L:

Fms-like tyrosine kinase-3 ligand

GM-CSF:

Granulocyte–macrophage colony stimulating factor

GPR81:

G-protein coupled receptor

GrB:

Granzyme B

HCC:

Hepatocellular carcinoma

HD:

Healthy donors

HEV:

High endothelial venules

hGMDP:

Human granulocyte-monocyte-DC progenitor

HHV8:

Human herpes virus 8

HIF-1α:

Hypoxia inducible factor alpha

hMDP:

Human monocyte-dendritic progenitor

HNSCC:

Head and neck squamous cell carcinoma

HPV:

Human Papilloma Virus

HSPCs:

Hematopoietic stem progenitor cells

ICB:

Immune checkpoint blockade

ICOS-L:

Inducible co-stimulator ligand

IDO:

Indoleamine 2,3-dioxygenase

IFN-α:

Interferon alpha

IFN-λ:

Interferon lambda

I-IFN:

Type I interferon

III-IFN:

Type III interferon

IL-3:

Interleukin-3

ILT7:

Immunoglobulin-like transcript 7

IMQ:

Imiquimod

IRF:

Interferon Regulatory Factor

KSHV:

Kaposi's sarcoma herpesvirus

LUAD:

Lung adenocarcinoma

LYZ:

Ysozyme

M-CSF:

Macrophage colony stimulating factor

MLPs:

Multipotent lymphoid early progenitors

MM:

Metastatic melanoma

moDCs:

Monocyte-derived dendritic cells

MPDCP:

Mature PDC Proliferation

NK:

Natural Killer cells

NLRs:

NOD-like receptors

NSCLC:

Non-small cell lung cancer

OS:

Overall survival

OSCC:

Oral squamous cell carcinoma

OX40-L/TNFSF4:

Tumor necrosis factor ligand superfamily member 4

OXPHOS:

Oxidative phosphorylation

PDC:

Plasmacytoid Dendritic Cells

PDGF-DD:

Platelet-derived growth factor D

PD-L1:

Programmed death-ligand 1

PFS:

Progression free survival

R848:

Resiquimod

RLRs:

RIG-I-like receptors

RT:

Radiotherapy

STING:

Stimulator of interferon genes

TA-pDCs:

Tumor-associated pDCs

TDLN:

Tumor-draining lymph node

TLR:

Toll-like receptor

TME:

Tumor microenvironment

TRAIL:

TNF-related apoptosis inducing ligand

Th1:

Type 1 helper T cells

Th2:

Type 1 helper T cells

Treg:

Regulatory T cell

References

  1. Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol. 2000;165(11):6037–46.

    Article  CAS  PubMed  Google Scholar 

  2. Sozzani S, Vermi W, Del Prete A, Facchetti F. Trafficking properties of plasmacytoid dendritic cells in health and disease. Trends Immunol. 2010;31(7):270–7.

    Article  CAS  PubMed  Google Scholar 

  3. Monti M, Consoli F, Vescovi R, Bugatti M, Vermi W. Human plasmacytoid dendritic cells and cutaneous melanoma. Cells. 2020;9(2):417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zapatka M, Borozan I, Brewer DS, Iskar M, Grundhoff A, Alawi M, Desai N, Sültmann H, Moch H, Alawi M, et al. The landscape of viral associations in human cancers. Nat Genet. 2020;52(3):320–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Swiecki M, Colonna M. The multifaceted biology of plasmacytoid dendritic cells. Nat Rev Immunol. 2015;15(8):471–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Reizis B. Plasmacytoid dendritic cells: development, regulation, and function. Immunity. 2019;50(1):37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yin Z, Dai J, Deng J, Sheikh F, Natalia M, Shih T, Lewis-Antes A, Amrute S, Garrigues U, Doyle S, et al. Type III IFNs are produced by and stimulate human plasmacytoid dendritic cells. J Immunol (Baltimore, Md). 2012;189(6):2735.

    CAS  Google Scholar 

  8. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801.

    Article  CAS  PubMed  Google Scholar 

  9. Reis E, Sousa C, Yamasaki S, Brown GD. Myeloid C-type lectin receptors in innate immune recognition. Immunity. 2024;57(4):700–17.

    Article  Google Scholar 

  10. Yin Z, Dai J, Deng J, Sheikh F, Natalia M, Shih T, Lewis-Antes A, Amrute SB, Garrigues U, Doyle S, et al. Type III IFNs are produced by and stimulate human plasmacytoid dendritic cells. J Immunol. 2012;189(6):2735–45.

    Article  CAS  PubMed  Google Scholar 

  11. Finotti G, Tamassia N, Cassatella M. Interferon-λs and plasmacytoid dendritic cells: a close relationship. FrontImmunol. 2017;8:1015.

    Google Scholar 

  12. Finotti G, Tamassia N, Cassatella M. Synergistic production of TNFα and IFNα by human pDCs incubated with IFNλ3 and IL-3. Cytokine. 2016;86:124–31.

    Article  CAS  PubMed  Google Scholar 

  13. Finotti G, Tamassia N, Calzetti F, Fattovich G, Cassatella MA. Endogenously produced TNF-α contributes to the expression of CXCL10/IP-10 in IFN-λ3-activated plasmacytoid dendritic cells. J Leukoc Biol. 2016;99(1):107–19.

    Article  CAS  PubMed  Google Scholar 

  14. Kalb ML, Glaser A, Stary G, Koszik F, Stingl G. TRAIL(+) human plasmacytoid dendritic cells kill tumor cells in vitro: mechanisms of imiquimod- and IFN-alpha-mediated antitumor reactivity. J Immunol. 2012;188(4):1583–91.

    Article  CAS  PubMed  Google Scholar 

  15. Drobits B, Holcmann M, Amberg N, Swiecki M, Grundtner R, Hammer M, Colonna M, Sibilia M. Imiquimod clears tumors in mice independent of adaptive immunity by converting pDCs into tumor-killing effector cells. J Clin Invest. 2012;122(2):575–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tel J, Smits E, Anguille S, Joshi R, Figdor C, de Vries I. Human plasmacytoid dendritic cells are equipped with antigen-presenting and tumoricidal capacities. Blood. 2012;120(19):3936.

    Article  CAS  PubMed  Google Scholar 

  17. Jahrsdörfer B, Vollmer A, Blackwell S, Maier J, Sontheimer K, Beyer T, Mandel B, Lunov O, Tron K, Nienhaus G, et al. Granzyme B produced by human plasmacytoid dendritic cells suppresses T-cell expansion. Blood. 2010;115(6):1156.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Koucký V, Hladíková K, Táborská E, Bouček J, Grega M, Špíšek R, Fialová A. The cytokine milieu compromises functional capacity of tumor-infiltrating plasmacytoid dendritic cells in HPV-negative but not in HPV-positive HNSCC. Cancer Immunol Immunother. 2021;70(9):2545–57.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Bontkes H, Ruizendaal J, Kramer D, Meijer C, Hooijberg E. Plasmacytoid dendritic cells are present in cervical carcinoma and become activated by human papillomavirus type 16 virus-like particles. Gynecol Oncol. 2005;96(3):897–901.

    Article  PubMed  Google Scholar 

  20. Vermi W, Fisogni S, Salogni L, Schärer L, Kutzner H, Sozzani S, Lonardi S, Rossini C, Calzavara-Pinton P, Leboit PE, et al. Spontaneous regression of highly immunogenic Molluscum contagiosum Virus (MCV)-induced skin lesions is associated with plasmacytoid dendritic cells and IFN-DC infiltration. J Investig Dermatol. 2011;131(2):426–34.

    Article  CAS  PubMed  Google Scholar 

  21. Demoulin S, Herfs M, Delvenne P, Hubert P. Tumor microenvironment converts plasmacytoid dendritic cells into immunosuppressive/tolerogenic cells: insight into the molecular mechanisms. J Leukoc Biol. 2013;93(3):343–52.

    Article  CAS  PubMed  Google Scholar 

  22. Hartmann E, Wollenberg B, Rothenfusser S, Wagner M, Wellisch D, Mack B, Giese T, Gires O, Endres S, Hartmann G. Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer. Cancer Res. 2003;63(19):6478–87.

    CAS  PubMed  Google Scholar 

  23. Koucky V, Boucek J, Fialova A. Immunology of plasmacytoid dendritic cells in solid tumors: a brief review. Cancers (Basel). 2019;11(4):470.

    CAS  PubMed  Google Scholar 

  24. Hoeffel G, Ripoche A-C, Matheoud D, Nascimbeni M, Escriou N, Lebon P, Heshmati F, Guillet J-G, Gannagé M, Caillat-Zucman S, et al. Antigen crosspresentation by human plasmacytoid dendritic cells. Immunity. 2007;27(3):481–92.

    Article  CAS  PubMed  Google Scholar 

  25. Colonna M, Cella M. Crosspresentation: plasmacytoid dendritic cells are in the business. Immunity. 2007;27(3):419–21.

    Article  CAS  PubMed  Google Scholar 

  26. Lou Y, Liu C, Kim GJ, Liu Y-J, Hwu P, Wang G. Plasmacytoid dendritic cells synergize with myeloid dendritic cells in the induction of antigen-specific antitumor immune responses. J Immunol. 2007;178(3):1534–41.

    Article  CAS  PubMed  Google Scholar 

  27. Fu C, Peng P, Loschko J, Feng L, Pham P, Cui W, Lee KP, Krug AB, Jiang A. Plasmacytoid dendritic cells cross-prime naive CD8 T cells by transferring antigen to conventional dendritic cells through exosomes. Proc Natl Acad Sci. 2020;117(38):23730–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lennert K, Remmele W. Karyometric research on lymph node cells in man. I. Germinoblasts, lymphoblasts & lymphocytes. Acta Haematol. 1958;19(2):99–113.

    Article  CAS  PubMed  Google Scholar 

  29. Lennert K, Kaiserling E, Müller-Hermelink HK. T-associated plasma-cells. Lancet. 1975;305(7914):1031–2.

    Article  Google Scholar 

  30. Vollenweider R, Lennert K. Plasmacytoid T-cell clusters in non-specific lymphadenitis. Virchows Archiv B Cell Pathol Includ Mol Pathol. 1983;44(1):1–14.

    Article  CAS  Google Scholar 

  31. Harris N, Demirjian Z. Plasmacytoid T-zone cell proliferation in a patient with chronic myelomonocytic leukemia. Histologic and immunohistologic characterization. Am JSurg Pathol. 1991;15(1):87–95.

    Article  CAS  Google Scholar 

  32. Facchetti F, de Wolf-Peeters C, Mason DY, Pulford K, van den Oord JJ, Desmet VJ. Plasmacytoid T cells Immunohistochemical evidence for their monocyte/macrophage origin. AmJ Pathol. 1988;133(1):15–21.

    CAS  Google Scholar 

  33. Facchetti F, Vermi W, Mason D, Colonna M. The plasmacytoid monocyte/interferon producing cells. Virchows Arch. 2003;443(6):703–17.

    Article  PubMed  Google Scholar 

  34. O’Doherty U, Peng M, Gezelter S, Swiggard W, Betjes M, Bhardwaj N, Steinman R. Human blood contains two subsets of dendritic cells, one immunologically mature and the other immature. Immunology. 1994;82(3):487–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med. 1997;185(6):1101–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Facchetti F, Candiago E, Vermi W. Plasmacytoid monocytes express IL3-receptor alpha and differentiate into dendritic cells. Histopathology. 1999;35:88–9.

  37. Cella M, Facchetti F, Lanzavecchia A, Colonna M. Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent TH1 polarization. Nat Immunol. 2000;1(4):305–10.

    Article  CAS  PubMed  Google Scholar 

  38. Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, Ho S, Antonenko S, Liu YJ. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999;284(5421):1835–7.

    Article  CAS  PubMed  Google Scholar 

  39. Cella M, Jarrossay D, Facchetti F, Alebardi O, Nakajima H, Lanzavecchia A, Colonna M. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat Med. 1999;5(8):919–23.

    Article  CAS  PubMed  Google Scholar 

  40. Ziegler-Heitbrock L, Ohteki T, Ginhoux F, Shortman K, Spits H. Reclassifying plasmacytoid dendritic cells as innate lymphocytes. Nat Rev Immunol. 2023;23(1):1–2.

    Article  CAS  PubMed  Google Scholar 

  41. Ziegler-Heitbrock L, Ohteki T, Ginhoux F, Shortman K, Spits H. Reply to ‘Reclassification of plasmacytoid dendritic cells as innate lymphocytes is premature.’ Nat Rev Immunol. 2023;23(5):338–9.

    Article  CAS  PubMed  Google Scholar 

  42. Reizis B, Idoyaga J, Dalod M, Barrat F, Naik S, Trinchieri G, Tussiwand R, Cella M, Colonna M. Reclassification of plasmacytoid dendritic cells as innate lymphocytes is premature. Nat Rev Immunol. 2023;23(5):336–7.

    Article  CAS  PubMed  Google Scholar 

  43. Vermi W, Bonecchi R, Facchetti F, Bianchi D, Sozzani S, Festa S, Berenzi A, Cella M, Colonna M. Recruitment of immature plasmacytoid dendritic cells (plasmacytoid monocytes) and myeloid dendritic cells in primary cutaneous melanomas. J Pathol. 2003;200(2):255–68.

    Article  PubMed  Google Scholar 

  44. Swiecki M, Colonna M. Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance. Immunol Rev. 2010;234(1):142–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Karrich JJ, Jachimowski LC, Uittenbogaart CH, Blom B. The plasmacytoid dendritic cell as the Swiss army knife of the immune system: molecular regulation of its multifaceted functions. J Immunol. 2014;193(12):5772–8.

    Article  CAS  PubMed  Google Scholar 

  46. Villadangos JA, Young L. Antigen-presentation properties of plasmacytoid dendritic cells. Immunity. 2008;29(3):352–61.

    Article  CAS  PubMed  Google Scholar 

  47. Pelayo R, Hirose J, Huang J, Garrett KP, Delogu A, Busslinger M, Kincade PW. Derivation of 2 categories of plasmacytoid dendritic cells in murine bone marrow. Blood. 2005;105(11):4407–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bendriss-Vermare N, Barthélémy C, Durand I, Bruand C, Dezutter-Dambuyant C, Moulian N, Berrih-Aknin S, Caux C, Trinchieri G, Brière F. Human thymus contains IFN-α–producing CD11c–, myeloid CD11c+, and mature interdigitating dendritic cells. J Clin Investig. 2001;107(7):835–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Res P, Couwenberg F, Vyth-Dreese F, Spits H. Expression of pTalpha mRNA in a committed dendritic cell precursor in the human thymus. Blood. 1999;94(8):2647–57.

    Article  CAS  PubMed  Google Scholar 

  50. Chen YL, Chen TT, Pai LM, Wesoly J, Bluyssen HA, Lee CK. A type I IFN-Flt3 ligand axis augments plasmacytoid dendritic cell development from common lymphoid progenitors. J Exp Med. 2013;210(12):2515–22.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Marafioti T, Paterson JC, Ballabio E, Reichard KK, Tedoldi S, Hollowood K, Dictor M, Hansmann M-L, Pileri SA, Dyer MJ, et al. el markers of normal and neoplastic human plasmacytoid dendritic cells. Blood. 2008;111(7):3778–92.

    Article  CAS  PubMed  Google Scholar 

  52. Cisse B, Caton ML, Lehner M, Maeda T, Scheu S, Locksley R, Holmberg D, Zweier C, den Hollander NS, Kant SG, et al. Transcription factor E2–2 is an essential and specific regulator of plasmacytoid dendritic cell development. Cell. 2008;135(1):37–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Onai N, Kurabayashi K, Hosoi-Amaike M, Toyama-Sorimachi N, Matsushima K, Inaba K, Ohteki T. A clonogenic progenitor with prominent plasmacytoid dendritic cell developmental potential. Immunity. 2013;38(5):943–57.

    Article  CAS  PubMed  Google Scholar 

  54. Musumeci A, Lutz K, Winheim E, Krug AB. What makes a pDC: recent advances in understanding plasmacytoid DC development and heterogeneity. Front Immunol. 2019;10:1222.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Karsunky H, Merad M, Cozzio A, Weissman IL, Manz MG. Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid-committed progenitors to Flt3+ dendritic cells in vivo. J Exp Med. 2003;198(2):305–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. D’Amico A, Wu L. The early progenitors of mouse dendritic cells and plasmacytoid predendritic cells are within the bone marrow hemopoietic precursors expressing Flt3. J Exp Med. 2003;198(2):293–303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sathe P, Vremec D, Wu L, Corcoran L, Shortman K. Convergent differentiation: myeloid and lymphoid pathways to murine plasmacytoid dendritic cells. Blood. 2013;121(1):11–9.

    Article  CAS  PubMed  Google Scholar 

  58. Maraskovsky E, Daro E, Roux E, Teepe M, Maliszewski CR, Hoek J, Caron D, Lebsack ME, McKenna HJ. In vivo generation of human dendritic cell subsets by Flt3 ligand. Blood. 2000;96(3):878–84.

    Article  CAS  PubMed  Google Scholar 

  59. Tiniakou I, Hsu P, Lopez-Zepeda L, Garipler G, Esteva E, Adams N, Jang G, Soni C, Lau C, Liu F, et al. Genome-wide screening identifies Trim33 as an essential regulator of dendritic cell differentiation. Scienceimmunology. 2024;9(94):eadi1023.

    CAS  Google Scholar 

  60. Pulendran B, Banchereau J, Burkeholder S, Kraus E, Guinet E, Chalouni C, Caron D, Maliszewski C, Davoust J, Fay J, et al. Flt3-ligand and granulocyte colony-stimulating factor mobilize distinct human dendritic cell subsets in vivo. J Immunol. 2000;165(1):566–72.

    Article  CAS  PubMed  Google Scholar 

  61. Rodrigues PF, Alberti-Servera L, Eremin A, Grajales-Reyes GE, Ivanek R, Tussiwand R. Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells. Nat Immunol. 2018;19(7):711–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yamaoka K, Min B, Zhou Y-J, Paul WE, O’Shea JJ. Jak3 negatively regulates dendritic-cell cytokine production and survival. Blood. 2005;106(9):3227–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lee J, Breton G, Oliveira TY, Zhou YJ, Aljoufi A, Puhr S, Cameron MJ, Sékaly RP, Nussenzweig MC, Liu K. Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow. J Exp Med. 2015;212(3):385–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Helft J, Anjos-Afonso F, van der Veen AG, Chakravarty P, Bonnet D, Reis e Sousa C. Dendritic cell lineage potential in human early hematopoietic progenitors. Cell Rep. 2017;20(3):529–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Harman BC, Miller JP, Nikbakht N, Gerstein R, Allman D. Mouse plasmacytoid dendritic cells derive exclusively from estrogen-resistant myeloid progenitors. Blood. 2006;108(3):878–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Feng J, Pucella J, Jang G, Alcántara-Hernández M, Upadhaya S, Adams N, Khodadadi-Jamayran A, Lao CM, Stoeckius M, et al. Clonal lineage tracing reveals shared origin of conventional and plasmacytoid dendritic cells. Immunity. 2022;55(3):405–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Xiao W, Chan A, Waarts MR, Mishra T, Liu Y, Cai SF, Yao J, Gao Q, Bowman RL, Koche RP, et al. Plasmacytoid dendritic cell expansion defines a distinct subset of <i>RUNX1</i>-mutated acute myeloid leukemia. Blood. 2021;137(10):1377–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zalmaï L, Viailly P, Biichle S, Cheok M, Soret L, Angelot-Delettre F, Petrella T, Collonge-Rame M, Seilles E, Geffroy S, et al. Plasmacytoid dendritic cells proliferation associated with acute myeloid leukemia: phenotype profile and mutation landscape. Haematologica. 2021;106(12):3056–66.

    Article  PubMed  Google Scholar 

  69. Matsui T, Connolly JE, Michnevitz M, Chaussabel D, Yu CI, Glaser C, Tindle S, Pypaert M, Freitas H, Piqueras B, et al. CD2 distinguishes two subsets of human plasmacytoid dendritic cells with distinct phenotype and functions. J Immunol. 2009;182(11):6815–23.

    Article  CAS  PubMed  Google Scholar 

  70. Zhang H, Gregorio JD, Iwahori T, Zhang X, Choi O, Tolentino LL, Prestwood T, Carmi Y, Engleman EG. A distinct subset of plasmacytoid dendritic cells induces activation and differentiation of B and T lymphocytes. Proc Natl Acad Sci U S A. 2017;114(8):1988–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Villani A-C, Satija R, Reynolds G, Sarkizova S, Shekhar K, Fletcher J, Griesbeck M, Butler A, Zheng S. Lazo S et al: Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. 2017;356(6335):eaah4573.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Chen B, Zhu L, Yang S, Su W. Unraveling the heterogeneity and ontogeny of dendritic cells using single-cell RNA sequencing. Front Immunol. 2021;12:711329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Alculumbre SG, Saint-Andre V, Di Domizio J, Vargas P, Sirven P, Bost P, Maurin M, Maiuri P, Wery M, Roman MS, et al. Diversification of human plasmacytoid predendritic cells in response to a single stimulus. Nat Immunol. 2018;19(1):63–75.

    Article  CAS  PubMed  Google Scholar 

  74. Wimmers F, Subedi N, van Buuringen N, Heister D, Vivie J, Beeren-Reinieren I, Woestenenk R, Dolstra H, Piruska A, Jacobs JFM, et al. Single-cell analysis reveals that stochasticity and paracrine signaling control interferon-alpha production by plasmacytoid dendritic cells. Nat Commun. 2018;9(1):3317.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Van Eyndhoven LC, Chouri E, Subedi N, Tel J. Phenotypicaldiversification of early IFNα-producing human plasmacytoid dendritic cellsusing droplet-based microfluidics. Front Immunol. 2021;12:672729.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Marsman C, Lafouresse F, Liao Y, Baldwin TM, Mielke LA, Hu Y, Mack M, Hertzog PJ, de Graaf CA, Shi W, et al. Plasmacytoid dendritic cell heterogeneity is defined by CXCL10 expression following TLR7 stimulation. Immunol Cell Biol. 2018;96(10):1083–94.

    Article  CAS  PubMed  Google Scholar 

  77. Jego G, Palucka AK, Blanck J-P, Chalouni C, Pascual V, Banchereau J. Plasmacytoid Dendritic Cells Induce Plasma Cell Differentiation through Type I Interferon and Interleukin 6. Immunity. 2003;19(2):225–34.

    Article  CAS  PubMed  Google Scholar 

  78. García-Marquez M, Shimabukuro-Vornhagen A, Von Bergwelt-Baildon M. Complex interactions between B cells and dendritic cells. Blood. 2013;121(12):2367–8.

    Article  PubMed  Google Scholar 

  79. Shaw J, Wang Y-H, Ito T, Arima K, Liu Y-J. Plasmacytoid dendritic cells regulate B-cell growth and differentiation via CD70. Blood. 2010;115(15):3051–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Bryant C, Fromm PD, Kupresanin F, Clark G, Lee K, Clarke C, Silveira PA, Suen H, Brown R, Newman E, et al. A CD2 high-expressing stress-resistant human plasmacytoid dendritic-cell subset. Immunol Cell Biol. 2016;94(5):447–57.

    Article  CAS  PubMed  Google Scholar 

  81. Alcántara-Hernández M, Leylek R, Wagar LE, Engleman EG, Keler T, Marinkovich MP, Davis MM, Nolan GP, Idoyaga J. High-dimensional phenotypic mapping of human dendritic cells reveals interindividual variation and tissue specialization. Immunity. 2017;47(6):1037-1050.e1036.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Seth S, Oberdorfer L, Hyde R, Hoff K, Thies V, Worbs T, Schmitz S, Forster R. CCR7 essentially contributes to the homing of plasmacytoid dendritic cells to lymph nodes under steady-state as well as inflammatory conditions. J Immunol. 2011;186(6):3364–72.

    Article  CAS  PubMed  Google Scholar 

  83. See P, Dutertre C, Chen J, Günther P, McGovern N, Irac S, Gunawan M, Beyer M, Händler K, Duan K, et al. Mapping the human DC lineage through the integration of high-dimensional techniques. Science(New York, NY). 2017;356(6342):eaag3009.

    Article  Google Scholar 

  84. Sosa Cuevas E, Bendriss-Vermare N, Mouret S, De Fraipont F, Charles J, Valladeau-Guilemond J, Chaperot L, Aspord C. Diversification of circulating and tumor-infiltrating plasmacytoid DCs towards the P3 (CD80+PDL1−)-pDC subset negatively correlated with clinical outcomes in melanoma patients. Clini Transl Immunol. 2022;11(5):1382.

    Article  Google Scholar 

  85. Lorenzi L, Lonardi S, Vairo D, Bernardelli A, Tomaselli M, Bugatti M, Licini S, Arisi M, Cerroni L, Tucci A, et al. E-cadherin expression and blunted interferon response in blastic plasmacytoid dendritic cell neoplasm. Am JSurg Pathol. 2021;45(10):1428.

    Article  Google Scholar 

  86. Ghanem MH, Shih AJ, Khalili H, Werth EG, Chakrabarty JK, Brown LM, Simpfendorfer KR, Gregersen PK. Proteomicand single-cell transcriptomic dissection of human plasmacytoid dendritic cellresponse to influenza virus. Front Immunol. 2022;13:814627.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Vanbervliet B, Bendriss-Vermare N, Massacrier C, Homey B, de Bouteiller O, Briere F, Trinchieri G, Caux C. The inducible CXCR3 ligands control plasmacytoid dendritic cell responsiveness to the constitutive chemokine stromal cell-derived factor 1 (SDF-1)/CXCL12. J Exp Med. 2003;198(5):823–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Penna G, Sozzani S, Adorini L. Cutting edge: selective usage of chemokine receptors by plasmacytoid dendritic cells. J Immunol. 2001;167(4):1862–6.

    Article  CAS  PubMed  Google Scholar 

  89. Kohrgruber N, Gröger M, Meraner P, Kriehuber E, Petzelbauer P, Brandt S, Stingl G, Rot A, Maurer D. Plasmacytoid dendritic cell recruitment by immobilized CXCR3 ligands. J Immunol (Baltimore, Md : 1950). 2004;173(11):6592.

    Article  CAS  Google Scholar 

  90. Gerlini G, Urso C, Mariotti G, Di Gennaro P, Palli D, Brandani P, Salvadori A, Pimpinelli N, Reali UM, Borgognoni L. Plasmacytoid dendritic cells represent a major dendritic cell subset in sentinel lymph nodes of melanoma patients and accumulate in metastatic nodes. Clin Immunol. 2007;125(2):184–93.

    Article  CAS  PubMed  Google Scholar 

  91. Thiel A, Pries R, Jeske S, Trenkle T, Wollenberg B. Effect of head and neck cancer supernatant and CpG-oligonucleotides on migration and IFN-alpha production of plasmacytoid dendritic cells. Anticancer Res. 2009;29(8):3019–25.

    CAS  PubMed  Google Scholar 

  92. Oliveira-Neto H, Silva E, Leles C, Mendonça E, de Alencar RC, Silva T, Batista A. Involvement of CXCL12 and CXCR4 in lymph node metastases and development of oral squamous cell carcinomas. Tumour Biol J Int Soc Oncodev Biol Med. 2008;29(4):262.

    Article  CAS  Google Scholar 

  93. Zou W, Machelon V, Coulomb-L’Hermin A, Borvak J, Nome F, Isaeva T, Wei S, Krzysiek R, Durand-Gasselin I, Gordon A, et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001;7(12):1339–46.

    Article  CAS  PubMed  Google Scholar 

  94. Gadalla R, Hassan H, Ibrahim SA, Abdullah MS, Gaballah A, Greve B, El-Deeb S, El-Shinawi M, Mohamed MM. Tumor microenvironmental plasmacytoid dendritic cells contribute to breast cancer lymph node metastasis via CXCR4/SDF-1 axis. Breast Cancer Res Treat. 2019;174(3):679–91.

    Article  CAS  PubMed  Google Scholar 

  95. Mansfield AS, Heikkila P, von Smitten K, Vakkila J, Leidenius M. Metastasis to sentinel lymph nodes in breast cancer is associated with maturation arrest of dendritic cells and poor co-localization of dendritic cells and CD8+ T cells. Virchows Arch. 2011;459(4):391–8.

    Article  CAS  PubMed  Google Scholar 

  96. Mitchell D, Chintala S, Dey M. Plasmacytoid dendritic cell in immunity and cancer. J Neuroimmunol. 2018;322:63.

    Article  CAS  PubMed  Google Scholar 

  97. Charles J, Di Domizio J, Salameire D, Bendriss-Vermare N, Aspord C, Muhammad R, Lefebvre C, Plumas J, Leccia MT, Chaperot L. Characterization of circulating dendritic cells in melanoma: role of CCR6 in plasmacytoid dendritic cell recruitment to the tumor. J Invest Dermatol. 2010;130(6):1646–56.

    Article  CAS  PubMed  Google Scholar 

  98. Sisirak V, Vey N, Vanbervliet B, Duhen T, Puisieux I, Homey B, Bowman EP, Trinchieri G, Dubois B, Kaiserlian D, et al. CCR6/CCR10-mediated plasmacytoid dendritic cell recruitment to inflamed epithelia after instruction in lymphoid tissues. Blood. 2011;118(19):5130–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Vescovi R, Monti M, Moratto D, Paolini L, Consoli F, Benerini L, Melocchi L, Calza S, Chiudinelli M, Rossi G, et al. Collapse of the plasmacytoid dendritic cell compartment in advanced cutaneous melanomas by components of the tumor cell secretome. Cancer Immunol Res. 2019;7(1):12–28.

    Article  CAS  PubMed  Google Scholar 

  100. Vermi W, Riboldi E, Wittamer V, Gentili F, Luini W, Marrelli S, Vecchi A, Franssen JD, Communi D, Massardi L, et al. Role of ChemR23 in directing the migration of myeloid and plasmacytoid dendritic cells to lymphoid organs and inflamed skin. J Exp Med. 2005;201(4):509–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Wittamer VR, Franssen J-D, Vulcano M, Mirjolet J-FO, Le Poul E, Migeotte I, BréZillon SP, Tyldesley R, Blanpain CD, Detheux M, et al. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J Exp Med. 2003;198(7):977–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Shin WJ, Zabel BA, Pachynski RK. Mechanisms and functions of chemerin in cancer: potential roles in therapeutic intervention. Fronti Immunol. 2018;9:2772.

    Article  Google Scholar 

  103. Wang C, Wu W, Liu X, To K, Chen G, Yu J, Ng E. Increased serum chemerin level promotes cellular invasiveness in gastric cancer: a clinical and experimental study. Peptides. 2014;51:131–8.

    Article  CAS  PubMed  Google Scholar 

  104. Zhang J, Jin H, Zhu A, Ying R, Wei W, Zhang F. Prognostic significance of plasma chemerin levels in patients with gastric cancer. Peptides. 2014;61:7–11.

    Article  CAS  PubMed  Google Scholar 

  105. Mohr S, Bottin M, Lannes B, Neuville A, Bellocq J, Keith G, Rihn B. Microdissection, mRNA amplification and microarray: a study of pleural mesothelial and malignant mesothelioma cells. Biochimie. 2004;86(1):13–9.

    Article  CAS  PubMed  Google Scholar 

  106. Tümmler C, Snapkov I, Wickström M, Moens U, Ljungblad L, Maria Elfman LH, Winberg J-O, Kogner P, Johnsen JI, Sveinbjørnsson B. Inhibition of chemerin/CMKLR1 axis in neuroblastoma cells reduces clonogenicity and cell viability <i>in vitro</i> and impairs tumor growth <i>in vivo</i>. Oncotarget. 2017;8(56):95135–51.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Demoulin S, Herfs M, Somja J, Roncarati P, Delvenne P, Hubert P. HMGB1 secretion during cervical carcinogenesis promotes the acquisition of a tolerogenic functionality by plasmacytoid dendritic cells. Int J Cancer. 2015;137(2):345–58.

    Article  CAS  PubMed  Google Scholar 

  108. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6(11):836.

    Article  CAS  PubMed  Google Scholar 

  109. Holicek P, Guilbaud E, Klapp V, Truxova I, Spisek R, Galluzzi L, Fucikova J. Type I interferon and cancer. Immunol Rev. 2024;321(1):115–27.

    Article  CAS  PubMed  Google Scholar 

  110. Honda K, Yanai H, Mizutani T, Negishi H, Shimada N, Suzuki N, Ohba Y, Takaoka A, Yeh WC, Taniguchi T. Role of a transductional-transcriptional processor complex involving MyD88 and IRF-7 in Toll-like receptor signaling. Proc Natl Acad Sci U S A. 2004;101(43):15416–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, Shimada N, Ohba Y, Takaoka A, Yoshida N, et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature. 2005;434(7034):772–7.

    Article  CAS  PubMed  Google Scholar 

  112. Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, Taya C, Taniguchi T. Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature. 2005;434(7036):1035–40.

    Article  CAS  PubMed  Google Scholar 

  113. Bao M, Liu YJ. Regulation of TLR7/9 signaling in plasmacytoid dendritic cells. Protein Cell. 2013;4(1):40–52.

    Article  CAS  PubMed  Google Scholar 

  114. Bode C, Fox M, Tewary P, Steinhagen A, Ellerkmann RK, Klinman D, Baumgarten G, Hornung V, Steinhagen F. Human plasmacytoid dentritic cells elicit a Type I Interferon response by sensing DNA via the cGAS-STING signaling pathway. Eur J Immunol. 2016;46(7):1615–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Deb P, Dai J, Singh S, Kalyoussef E, Fitzgerald-Bocarsly P. Triggering of the cGAS–STING pathway in human plasmacytoid dendritic cells inhibits TLR9-mediated IFN production. J Immunol. 2023;205(1):223–36.

    Article  Google Scholar 

  116. Hopfner K-P, Hornung V. Molecular mechanisms and cellular functions of cGAS–STING signalling. Nat Rev Mol Cell Biol. 2020;21(9):501–21.

    Article  CAS  PubMed  Google Scholar 

  117. Kwon J, Bakhoum S. The cytosolic DNA-sensing cGAS-STING pathway in cancer. Cancer Discov. 2020;10(1):26.

    Article  CAS  PubMed  Google Scholar 

  118. Tanaka Y, Chen ZJ. STING specifies IRF3 phosphorylation by TBK1 in the cytosolic DNA signaling pathway. Sci Signal. 2012;5(214):ra20–ra20.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Terra M, Oberkampf M, Fayolle C, Rosenbaum P, Guillerey C, Dadaglio G, Leclerc C. Tumor-derived TGFbeta alters the ability of plasmacytoid dendritic cells to respond to innate immune signaling. Cancer Res. 2018;78(11):3014–26.

    Article  CAS  PubMed  Google Scholar 

  120. Bruchhage K, Heinrichs S, Wollenberg B, Pries R. IL-10 in the microenvironment of HNSCC inhibits the CpG ODN induced IFN-α secretion of pDCs. Oncol Lett. 2018;15(3):3985.

    PubMed  PubMed Central  Google Scholar 

  121. Koucký V, Hladíková K, Táborská E, Bouček J, Grega M, Špisek R, Fiolava A. The cytokine milieu compromises functional capacity of tumor-infiltrating plasmacytoid dendritic cells in HPV-negative but not in HPV-positive HNSCC. Cancer Immunol Immunother CII. 2021;70(9):2545.

    Article  PubMed  Google Scholar 

  122. Bekeredjian-Ding I, Schafer M, Hartmann E, Pries R, Parcina M, Schneider P, Giese T, Endres S, Wollenberg B, Hartmann G. Tumour-derived prostaglandin E and transforming growth factor-beta synergize to inhibit plasmacytoid dendritic cell-derived interferon-alpha. Immunology. 2009;128(3):439–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Sisirak V, Vey N, Goutagny N, Renaudineau S, Malfroy M, Thys S, Treilleux I, Labidi-Galy SI, Bachelot T, Dezutter-Dambuyant C, et al. Breast cancer-derived transforming growth factor-beta and tumor necrosis factor-alpha compromise interferon-alpha production by tumor-associated plasmacytoid dendritic cells. Int J Cancer. 2013;133(3):771–8.

    Article  CAS  PubMed  Google Scholar 

  124. Han N, Zhang Z, Jv H, Hu J, Ruan M, Zhang C. Culture supernatants of oral cancer cells induce impaired IFN-α production of pDCs partly through the down-regulation of TLR-9 expression. Arch Oral Biol. 2018;93:141.

    Article  CAS  PubMed  Google Scholar 

  125. Combes A, Camosseto V, N’Guessan P, Argüello RJ, Mussard J, Caux C, Bendriss-Vermare N, Pierre P, Gatti E. BAD-LAMP controls TLR9 trafficking and signalling in human plasmacytoid dendritic cells. Nat Commun. 2017;8(1):913.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Monti M, Ferrari G, Grosso V, Missale F, Bugatti M, Cancila V, Zini S, Segala A, La Via L, Consoli F, et al. Impaired activation of plasmacytoid dendritic cells via toll-like receptor 7/9 and STING is mediated by melanoma-derived immunosuppressive cytokines and metabolic drift. Front Immunol. 2024;14:1227648.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Hack K, Reilly L, Proby C, Fleming C, Leigh I, Foerster J. Wnt5a inhibits the CpG oligodeoxynucleotide-triggered activation of human plasmacytoid dendritic cells. Clin Exp Dermatol. 2012;37(5):557–61.

    Article  CAS  PubMed  Google Scholar 

  128. Sabree SA, Voigt AP, Blackwell SE, Vishwakarma A, Chimenti MS, Salem AK, Weiner GJ. Direct and indirect immune effects of CMP-001, a virus-like particle containing a TLR9 agonist. J Immunother Cancer. 2021;9(6): e002484.

    Article  PubMed  PubMed Central  Google Scholar 

  129. West JA, Gregory SM, Sivaraman V, Su L, Damania B. Activation of plasmacytoid dendritic cells by Kaposi’s Sarcoma-Associated Herpesvirus. J Virol. 2011;85(2):895–904.

    Article  CAS  PubMed  Google Scholar 

  130. Karouni M, Kurban M, Abbas O. Plasmacytoid dendritic cells in skin lesions of classic Kaposi’s sarcoma. Arch Dermatol Res. 2016;308(7):487–92.

    Article  CAS  PubMed  Google Scholar 

  131. Dzionek A, Sohma Y, Nagafune J, Cella M, Colonna M, Facchetti F, Gunther G, Johnston I, Lanzavecchia A, Nagasaka T, et al. BDCA-2, a novel plasmacytoid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon alpha/beta induction. J Exp Med. 2001;194(12):1823–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Cao W, Rosen DB, Ito T, Bover L, Bao M, Watanabe G, Yao Z, Zhang L, Lanier LL, Liu YJ. Plasmacytoid dendritic cell-specific receptor ILT7-Fc epsilonRI gamma inhibits Toll-like receptor-induced interferon production. J Exp Med. 2006;203(6):1399–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Meyer-Wentrup F, Benitez-Ribas D, Tacken PJ, Punt CJ, Figdor CG, de Vries IJ, Adema GJ. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production. Blood. 2008;111(8):4245–53.

    Article  CAS  PubMed  Google Scholar 

  134. Fuchs A, Cella M, Kondo T, Colonna M. Paradoxic inhibition of human natural interferon-producing cells by the activating receptor NKp44. Blood. 2005;106(6):2076–82.

    Article  CAS  PubMed  Google Scholar 

  135. Riboldi E, Daniele R, Parola C, Inforzato A, Arnold PL, Bosisio D, Fremont DH, Bastone A, Colonna M, Sozzani S. Human C-type lectin domain family 4, member C (CLEC4C/BDCA-2/CD303) is a receptor for asialo-galactosyl-oligosaccharides. J Biol Chem. 2011;286(41):35329–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Jégouzo S, Feinberg H, Dungarwalla T, Drickamer K, Weis W, Taylor M. A novel mechanism for binding of galactose-terminated glycans by the C-type carbohydrate recognition domain in blood dendritic cell antigen 2. J Biol Chem. 2015;290(27):16759.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Sosa Cuevas E, Roubinet B, Mouret S, Thépaut M, de Fraipont F, Charles J, Fieschi F, Landemarre L, Chaperot L, Aspord C. The melanoma tumor glyco-code impacts human dendritic cells’ functionality and dictates clinical outcomes. Front Immunol. 2023;14:1120434.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Venegas-Solis F, Staliunaite L, Rudolph E, Münch CC-S, Yu P, Freibert S-A, Maeda T, Zimmer CL, Möbs C, Keller C. A type I interferon regulatory network for human plasmacytoid dendritic cells based on heparin, membrane-bound and soluble BDCA-2. Proc Natl Acad Sci. 2024;121(12):e2312404121.

    Article  CAS  PubMed  Google Scholar 

  139. Atallah J, Khachfe H, Berro J, Assi H. The use of heparin and heparin-like molecules in cancer treatment: a review. Cancer Treat Res Commun. 2020;24:100192.

    Article  PubMed  Google Scholar 

  140. Furie RA, Van Vollenhoven RF, Kalunian K, Navarra S, Romero-Diaz J, Werth VP, Huang X, Clark G, Carroll H, Meyers A, et al. Trial of anti-BDCA2 antibody Litifilimab for systemic lupus erythematosus. N Engl J Med. 2022;387(10):894–904.

    Article  CAS  PubMed  Google Scholar 

  141. Li X, Zhang Y, Li B, Li J, Qiu Y, Zhu Z, Hua H. An immunomodulatory antibody-drug conjugate targeting BDCA2 strongly suppresses plasmacytoid dendritic cell function and glucocorticoid responsive genes. Rheumatology (Oxford). 2024;63(1):242–50.

    Article  PubMed  Google Scholar 

  142. Wilson N, Bover L, Konopleva M, Han L, Neelapu S, Pemmaraju N. CD303 (BDCA-2) - a potential novel target for therapy in hematologic malignancies. Leukemia Lymphoma. 2022;63(1):19.

    Article  CAS  PubMed  Google Scholar 

  143. Tsukamoto N, Okada S, Onami Y, Sasaki Y, Umezawa K, Kawakami Y. Impairment of plasmacytoid dendritic cells for IFN production by the ligand for immunoglobulin-like transcript 7 expressed on human cancer cells. Clin Cancer Res. 2009;15(18):5733–43.

    Article  CAS  PubMed  Google Scholar 

  144. Bego MG, Miguet N, Laliberté A, Aschman N, Gerard F, Merakos AA, Weissenhorn W, Cohen ÉA. Activation of the ILT7 receptor and plasmacytoid dendritic cell responses are governed by structurally-distinct BST2 determinants. J Biol Chem. 2019;294(27):10503–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Cao W, Bover L. Signaling and ligand interaction of ILT7: receptor-mediated regulatory mechanisms for plasmacytoid dendritic cells. Immunol Rev. 2010;234(1):163–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Xu X, Wang Y, Xue F, Guan E, Tian F, Xu J, Zhang H. BST2 promotes tumor growth via multiple pathways in hepatocellular carcinoma. Cancer Invest. 2020;38(5):329–37.

    Article  CAS  PubMed  Google Scholar 

  147. Thang Pham Q, Oue N, Yamamoto Y, Shigematsu Y, Sekino Y, Sakamoto N, Sentani K, Uraoka N, Tiwari M, Yasui W. The expression of BTS-2 enhances cell growth and invasiveness in renal cell carcinoma. Anticancer Res. 2017;37(6):2853.

    Google Scholar 

  148. Mahauad-Fernandez WD, Demali KA, Olivier AK, Okeoma CM. Bone marrow stromal antigen 2 expressed in cancer cells promotes mammary tumor growth and metastasis. Breast Cancer Res. 2014;16(6):493.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Tiwari R, de la Torre J, McGavern D, Nayak D. Beyond tethering the viral particles: immunomodulatory functions of tetherin ( BST-2). DNA Cell Biol. 2019;38(11):1170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Cao W, Bover L, Cho M, Wen X, Hanabuchi S, Bao M, Rosen DB, Wang YH, Shaw JL, Du Q, et al. Regulation of TLR7/9 responses in plasmacytoid dendritic cells by BST2 and ILT7 receptor interaction. J Exp Med. 2009;206(7):1603–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Fabricius D, O’Dorisio MS, Blackwell S, Jahrsdörfer B. Human plasmacytoid dendritic cell function: inhibition of IFN-alpha secretion and modulation of immune phenotype by vasoactive intestinal peptide. J Immunol. 2006;177(9):5920–7.

    Article  CAS  PubMed  Google Scholar 

  152. de la Cruz-López K, Castro-Muñoz L, Reyes-Hernández D, García-Carrancá A, Manzo-Merino J. Lactate in the regulation of tumor microenvironment and therapeutic approaches. Front Oncol. 2019;9:1143.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Saas P, Varin A, Perruche S, Ceroi A. Recent insights into the implications of metabolism in plasmacytoid dendritic cell innate functions: potential ways to control these functions. F1000Research. 2017;6:456.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Wu J, Li S, Yang Y, Zhu S, Zhang M, Qiao Y, Liu YJ, Chen J. TLR-activated plasmacytoid dendritic cells inhibit breast cancer cell growth in vitro and in vivo. Oncotarget. 2017;8(7):11708–18.

    Article  PubMed  Google Scholar 

  155. Liu C, Lou Y, Lizee G, Qin H, Liu S, Rabinovich B, Kim GJ, Wang YH, Ye Y, Sikora AG, et al. Plasmacytoid dendritic cells induce NK cell-dependent, tumor antigen-specific T cell cross-priming and tumor regression in mice. J Clin Invest. 2008;118(3):1165–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Chaperot L, Blum A, Manches O, Lui G, Angel J, Molens JP, Plumas J. Virus or TLR agonists induce TRAIL-mediated cytotoxic activity of plasmacytoid dendritic cells. J Immunol. 2006;176(1):248–55.

    Article  CAS  PubMed  Google Scholar 

  157. Blum A, Chaperot L, Molens J, Foissaud V, Plantaz D, Plumas J. Mechanisms of TRAIL-induced apoptosis in leukemic plasmacytoid dendritic cells. Exp Hematol. 2006;34(12):1655.

    Article  CAS  PubMed  Google Scholar 

  158. Barblu L, Machmach K, Gras C, Delfraissy J, Boufassa F, Leal M, Ruiz-Mateos E, Lambotte O, Herbeuval J. Plasmacytoid dendritic cells (pDCs) from HIV controllers produce interferon-α and differentiate into functional killer pDCs under HIV activation. J Infect Dis. 2012;206(5):790.

    Article  CAS  PubMed  Google Scholar 

  159. Gandini M, Gras C, Azeredo E, Pinto L, Smith N, Despres P, da Cunha R, de Souza L, Kubelka C, Herbeuval J. Dengue virus activates membrane TRAIL relocalization and IFN-α production by human plasmacytoid dendritic cells in vitro and in vivo. PLoS Negl Trop Dis. 2013;7(6):e2257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Hardy A, Graham D, Shearer G, Herbeuval J. HIV turns plasmacytoid dendritic cells (pDC) into TRAIL-expressing killer pDC and down-regulates HIV coreceptors by Toll-like receptor 7-induced IFN-alpha. Proc Natl Acad Sci USA. 2007;104(44):17453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Dominguez-Molina B, Machmach K, Perales C, Tarancon-Diez L, Gallego I, Sheldon J, Leal M, Domingo E, Ruiz-Mateos E. Toll-like receptor 7 (TLR-7) and TLR-9 agonists improve hepatitis C Virus replication and infectivity inhibition by plasmacytoid dendritic cells. J Virol. 2018;92(23):e01219.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Stary G, Bangert C, Tauber M, Strohal R, Kopp T, Stingl G. Tumoricidal activity of TLR7/8-activated inflammatory dendritic cells. J Exp Med. 2007;204(6):1441–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Tel J, Anguille S, Waterborg CEJ, Smits EL, Figdor CG, De Vries IJM. Tumoricidal activity of human dendritic cells. Trends Immunol. 2014;35(1):38–46.

    Article  CAS  PubMed  Google Scholar 

  164. Diamond M, Kinder M, Matsushita H, Mashayekhi M, Dunn G, Archambault J, Lee H, Arthur C, White J, Kalinke U, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208(10):1989.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Belounis A, Ayoub M, Cordeiro P, Lemieux W, Teira P, Haddad E, Herblot S, Duval M. Patients’ NK cell stimulation with activated plasmacytoid dendritic cells increases dinutuximab-induced neuroblastoma killing. Cancer Immunol Immunother CII. 2020;69(9):1767.

    Article  CAS  PubMed  Google Scholar 

  166. Kemp TJ, Elzey BD, Griffith TS. Plasmacytoid dendritic cell-derived IFN-alpha induces TNF-related apoptosis-inducing ligand/Apo-2L-mediated antitumor activity by human monocytes following CpG oligodeoxynucleotide stimulation. J Immunol. 2003;171(1):212–8.

    Article  CAS  PubMed  Google Scholar 

  167. Vermi W, Lonardi S, Morassi M, Rossini C, Tardanico R, Venturini M, Sala R, Tincani A, Poliani P, Calzavara-Pinton P, et al. Cutaneous distribution of plasmacytoid dendritic cells in lupus erythematosus. Selective tropism at the site of epithelial apoptotic damage. Immunobiology. 2009;214(9–10):877–86.

    Article  CAS  PubMed  Google Scholar 

  168. Salvi V, Vermi W, Cavani A, Lonardi S, Carbone T, Facchetti F, Bosisio D, Sozzani S. IL-21 May promote granzyme B-dependent NK/Plasmacytoid dendritic cell functional interaction in cutaneous lupus erythematosus. J Invest Dermatol. 2017;137(7):1493.

    Article  CAS  PubMed  Google Scholar 

  169. Fabricius D, Nußbaum B, Busch D, Panitz V, Mandel B, Vollmer A, Westhoff M, Kaltenmeier C, Lunov O, Tron K, et al. Antiviral vaccines license T cell responses by suppressing granzyme B levels in human plasmacytoid dendritic cells. J Immunol (Baltimore, Md : 1950). 2013;191(3):1144.

    Article  CAS  Google Scholar 

  170. Bratke K, Nielsen J, Manig F, Klein C, Kuepper M, Geyer S, Julius P, Lommatzsch M, Virchow J. Functional expression of granzyme B in human plasmacytoid dendritic cells: a role in allergic inflammation. Clin Exp Allergy J Brit Soc Allergy Clin Immunol. 2010;40(7):1015.

    Article  CAS  Google Scholar 

  171. Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA, Kobold S. Interleukins in cancer: from biology to therapy. Nat Rev Cancer. 2021;21(8):481–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Beckebaum S, Zhang X, Chen X, Yu Z, Frilling A, Dworacki G, Grosse-Wilde H, Broelsch CE, Gerken G, Cicinnati VR. Increased levels of interleukin-10 in serum from patients with hepatocellular carcinoma correlate with profound numerical deficiencies and immature phenotype of circulating dendritic cell subsets. Clin Cancer Res. 2004;10(21):7260–9.

    Article  CAS  PubMed  Google Scholar 

  173. Gondek DC, Lu L-F, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol. 2005;174(4):1783–6.

    Article  CAS  PubMed  Google Scholar 

  174. Wieckowski E, Wang G, Gastman B, Goldstein L, Rabinowich H. Granzyme B-mediated degradation of T-cell receptor zeta chain. Cancer Res. 2002;62(17):4884–9.

  175. Chen W, Liang X, Peterson AJ, Munn DH, Blazar BR. The indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. J Immunol. 2008;181(8):5396–404.

    Article  CAS  PubMed  Google Scholar 

  176. Huang XM, Liu XS, Lin XK, Yu H, Sun JY, Liu XK, Chen C, Jin HL, Zhang GE, Shi XX, et al. Role of plasmacytoid dendritic cells and inducible costimulator-positive regulatory T cells in the immunosuppression microenvironment of gastric cancer. Cancer Sci. 2014;105(2):150–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Araújo EFD, Medeiros DH, Galdino NADL, Condino-Neto A, Calich VLG, Loures FV. Tolerogenic Plasmacytoid Dendritic Cells Control Paracoccidioides brasiliensis Infection by Inducting Regulatory T Cells in an IDO-Dependent Manner. PLoS Pathog. 2016;12(12): e1006115.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Solinas C, Gu-Trantien C, Willard-Gallo K. The rationale behind targeting the ICOS-ICOS ligand costimulatory pathway in cancer immunotherapy. ESMO Open. 2020;5(1):e000544.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Ito T, Yang M, Wang YH, Lande R, Gregorio J, Perng OA, Qin XF, Liu YJ, Gilliet M. Plasmacytoid dendritic cells prime IL-10-producing T regulatory cells by inducible costimulator ligand. J Exp Med. 2007;204(1):105–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Pedroza-Gonzalez A, Zhou G, Vargas-Mendez E, Boor PP, Mancham S, Verhoef C, Polak WG, Grunhagen D, Pan Q, Janssen H, et al. Tumor-infiltrating plasmacytoid dendritic cells promote immunosuppression by Tr1 cells in human liver tumors. Oncoimmunology. 2015;4(6): e1008355.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Labidi-Galy SI, Sisirak V, Meeus P, Gobert M, Treilleux I, Bajard A, Combes JD, Faget J, Mithieux F, Cassignol A, et al. Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Res. 2011;71(16):5423–34.

    Article  CAS  PubMed  Google Scholar 

  182. Conrad C, Gregorio J, Wang YH, Ito T, Meller S, Hanabuchi S, Anderson S, Atkinson N, Ramirez PT, Liu YJ, et al. Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells. Cancer Res. 2012;72(20):5240–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Sisirak V, Faget J, Gobert M, Goutagny N, Vey N, Treilleux I, Renaudineau S, Poyet G, Labidi-Galy SI, Goddard-Leon S, et al. Impaired IFN-alpha production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 2012;72(20):5188–97.

    Article  CAS  PubMed  Google Scholar 

  184. Faget J, Sisirak V, Blay JY, Caux C, Bendriss-Vermare N, Menetrier-Caux C. ICOS is associated with poor prognosis in breast cancer as it promotes the amplification of immunosuppressive CD4(+) T cells by plasmacytoid dendritic cells. Oncoimmunology. 2013;2(3): e23185.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Ito T, Amakawa R, Inaba M, Hori T, Ota M, Nakamura K, Takebayashi M, Miyaji M, Yoshimura T, Inaba K, et al. Plasmacytoid dendritic cells regulate Th cell responses through OX40 ligand and type I IFNs. J Immunol. 2004;172(7):4253.

    Article  CAS  PubMed  Google Scholar 

  186. Aspord C, Leccia MT, Charles J, Plumas J. Plasmacytoid dendritic cells support melanoma progression by promoting Th2 and regulatory immunity through OX40L and ICOSL. Cancer Immunol Res. 2013;1(6):402–15.

    Article  CAS  PubMed  Google Scholar 

  187. Poropatich K, Dominguez D, Chan W, Andrade J, Zha Y, Wray B, Miska J, Qin L, Cole L, Coates S, et al. OX40+ plasmacytoid dendritic cells in the tumor microenvironment promote antitumor immunity. J Clin Invest. 2020;130(7):3528.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Gerlini G, Di Gennaro P, Mariotti G, Urso C, Chiarugi A, Pimpinelli N, Borgognoni L. Indoleamine 2,3-dioxygenase+ cells correspond to the BDCA2+ plasmacytoid dendritic cells in human melanoma sentinel nodes. J Invest Dermatol. 2010;130:898–901.

  189. Sharma MD, Baban B, Chandler P, Hou DY, Singh N, Yagita H, Azuma M, Blazar BR, Mellor AL, Munn DH. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest. 2007;117(9):2570–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Ye L, Chen W, Bai X, Xu X, Zhang Q, Xia X, Sun X, Li G, Hu Q, Fu Q, et al. Hypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76(4):818.

    Article  CAS  PubMed  Google Scholar 

  191. Fan C, Wu J, Shen Y, Hu H, Wang Q, Mao Y, Ye B, Xiang M. Hypoxia promotes the tolerogenic phenotype of plasmacytoid dendritic cells in head and neck squamous cell carcinoma. Cancer Med. 2022;11(4):922–30.

    Article  PubMed  Google Scholar 

  192. Kobayashi S, Wannakul T, Sekino K, Takahashi Y, Kagawa Y, Miyazaki H, Umaru B, Yang S, Yamamoto Y, Owada Y. Fatty acid-binding protein 5 limits the generation of Foxp3+ regulatory T cells through regulating plasmacytoid dendritic cell function in the tumor microenvironment. Int J Cancer. 2022;150(1):152.

    Article  CAS  PubMed  Google Scholar 

  193. Bosisio D, Ronca R, Salvi V, Presta M, Sozzani S. Dendritic cells in inflammatory angiogenesis and lymphangiogenesis. Curr Opin Immunol. 2018;53:180–6.

    Article  CAS  PubMed  Google Scholar 

  194. Riboldi E, Musso T, Moroni E, Urbinati C, Bernasconi S, Rusnati M, Adorini L, Presta M, Sozzani S. Cutting edge: proangiogenic properties of alternatively activated dendritic cells. J Immunol. 2005;175(5):2788–92.

    Article  CAS  PubMed  Google Scholar 

  195. Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, Wei S, Zou L, Kryczek I, Hoyle G, et al. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004;64(16):5535–8.

    Article  CAS  PubMed  Google Scholar 

  196. Sorrentino R, Terlizzi M, Di Crescenzo VG, Popolo A, Pecoraro M, Perillo G, Galderisi A, Pinto A. Human lung cancer-derived immunosuppressive plasmacytoid dendritic cells release IL-1alpha in an AIM2 inflammasome-dependent manner. Am J Pathol. 2015;185(11):3115–24.

    Article  CAS  PubMed  Google Scholar 

  197. Treilleux I, Blay JY, Bendriss-Vermare N, Ray-Coquard I, Bachelot T, Guastalla JP, Bremond A, Goddard S, Pin JJ, Barthelemy-Dubois C, et al. Dendritic cell infiltration and prognosis of early stage breast cancer. Clin Cancer Res. 2004;10(22):7466–74.

    Article  CAS  PubMed  Google Scholar 

  198. Labidi-Galy SI, Treilleux I, Goddard-Leon S, Combes JD, Blay JY, Ray-Coquard I, Caux C, Bendriss-Vermare N. Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis. Oncoimmunology. 2012;1(3):380–2.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Han N, Zhang Z, Liu S, Ow A, Ruan M, Yang W, Zhang C. Increased tumor-infiltrating plasmacytoid dendritic cells predicts poor prognosis in oral squamous cell carcinoma. Arch Oral Biol. 2017;78:129–34.

    Article  CAS  PubMed  Google Scholar 

  200. Kießler M, Plesca I, Sommer U, Wehner R, Wilczkowski F, Müller L, Tunger A, Lai X, Rentsch A, Peuker K, et al. Tumor-infiltrating plasmacytoid dendritic cells are associated with survival in human colon cancer. J Immunother Cancer. 2021;9(3): e001813.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Wu J, Cheng H, Wang H, Zang G, Qi L, Lv X, Liu C, Zhu S, Zhang M, Cui J, et al. Correlation between immune lymphoid cells and plasmacytoid dendritic cells in human colon cancer. Front Immunol. 2021;12:601611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Gai XD, Song Y, Li C, Lei YM, Yang B. Potential role of plasmacytoid dendritic cells for FOXP3+ regulatory T cell development in human colorectal cancer and tumor draining lymph node. Pathol Res Pract. 2013;209(12):774–8.

    Article  CAS  PubMed  Google Scholar 

  203. Zhou Z, Xin H, Li J, Hu Z, Luo C, Zhou S. Intratumoral plasmacytoid dendritic cells as a poor prognostic factor for hepatocellular carcinoma following curative resection. Cancer Immunol Immunother CII. 2019;68(8):1223.

    Article  PubMed  Google Scholar 

  204. Pang L, Ng K, Liu J, Yeung W, Zhu J, Chiu T, Liu H, Chen Z, Lo C, Man K. Plasmacytoid dendritic cells recruited by HIF-1α/eADO/ADORA1 signaling induce immunosuppression in hepatocellular carcinoma. Cancer Lett. 2021;522:80.

    Article  CAS  PubMed  Google Scholar 

  205. Shi W, Li X, Porter JL, Ostrodi DH, Yang B, Li J, Wang Y, Zhang J, Bai L, Jiao S. Level of plasmacytoid dendritic cells is increased in non-small cell lung carcinoma. Tumour Biol. 2014;35(3):2247–52.

    Article  CAS  PubMed  Google Scholar 

  206. Liu W, Zhao J, Li Q, Wang Q, Zhou Y, Tong Z. Gastric cancer patients have elevated plasmacytoid and CD1c+ dendritic cells in the peripheral blood. Oncol Lett. 2018;15(4):80.

    Google Scholar 

  207. Liu X, Yu H, Yan C, Mei Y, Lin C, Hong Y, Lin X, Zhang Q, Yu J. Plasmacytoid dendritic cells and ICOS+ regulatory T cells predict poor prognosis in gastric cancer: a pilot study. J Cancer. 2019;10(26):6711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Cha Y, Kim E, Choi Y, Kim C, Park M, Chang Y. Accumulation of plasmacytoid dendritic cell is associated with a treatment response to DNA-damaging treatment and favorable prognosis in lung adenocarcinoma. Front Immunol. 2023;14:1154881.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Butsch R, Lukas Waelti S, Schaerer S, Braun J, Korol D, Probst-Hensch N, Moch H, Kurrer M. Intratumoral plasmacytoid dendritic cells associate with increased survival in patients with follicular lymphoma. Leuk Lymphoma. 2011;52(7):1230–8.

    Article  CAS  PubMed  Google Scholar 

  210. Lenz P, Lowy DR, Schiller JT. Papillomavirus virus-like particles induce cytokines characteristic of innate immune responses in plasmacytoid dendritic cells. Eur J Immunol. 2005;35(5):1548–56.

    Article  CAS  PubMed  Google Scholar 

  211. Sosa Cuevas E, Ouaguia L, Mouret S, Charles J, De Fraipont F, Manches O, Valladeau-Guilemond J, Bendriss-Vermare N, Chaperot L, Aspord C. BDCA1+ cDC2s, BDCA2+ pDCs and BDCA3+ cDC1s reveal distinct pathophysiologic features and impact on clinical outcomes in melanoma patients. Clin Trans Immunol. 2020;9(11):e1190.

    Article  CAS  Google Scholar 

  212. Monti M, Vescovi R, Consoli F, Farina D, Moratto D, Berruti A, Specchia C, Vermi W. Plasmacytoid dendritic cell impairment in metastatic melanoma by lactic acidosis. Cancers. 2020;12(8):2085.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Dabrosin N, Sloth Juul K, Baehr Georgsen J, Andrup S, Schmidt H, Steiniche T, Heide Ollegaard T, Bonnelykke Behrndtz L. Innate immune cell infiltration in melanoma metastases affects survival and is associated with BRAFV600E mutation status. Melanoma Res. 2019;29(1):30–7.

    Article  CAS  PubMed  Google Scholar 

  214. Chen Y-P, Yin J-H, Li W-F, Li H-J, Chen D-P, Zhang C-J, Lv J-W, Wang Y-Q, Li X-M, Li J-Y, et al. Single-cell transcriptomics reveals regulators underlying immune cell diversity and immune subtypes associated with prognosis in nasopharyngeal carcinoma. Cell Res. 2020;30(11):1024–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Rossi R, Lichtner M, Iori F, Ermocida A, Mascia C, Mengoni F, Sauzullo I, Dini D, Mastroianni CM, Vullo V. Dendritic cells in blood and urine samples from bladder cancer patients undergoing BCG immunotherapy. Arch Ital Urol Androl. 2013;85(4):157–63.

    Article  PubMed  Google Scholar 

  216. Orsini G, Legitimo A, Failli A, Ferrari P, Nicolini A, Spisni R, Miccoli P, Consolini R. Quantification of blood dendritic cells in colorectal cancer patients during the course of disease. Pathol Oncol Res. 2014;20(2):267–76.

    Article  CAS  PubMed  Google Scholar 

  217. Tjomsland V, Sandstrom P, Spangeus A, Messmer D, Emilsson J, Falkmer U, Falkmer S, Magnusson KE, Borch K, Larsson M. Pancreatic adenocarcinoma exerts systemic effects on the peripheral blood myeloid and plasmacytoid dendritic cells: an indicator of disease severity? BMC Cancer. 2010;10:87.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Kini Bailur J, Gueckel B, Pawelec G. Prognostic impact of high levels of circulating plasmacytoid dendritic cells in breast cancer. J Transl Med. 2016;14(1):151.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Zhou Z, Lin W, Li X, Huang Y, Ren J, Gao Y, Li J. Aberrant phenotype and function of dendritic cells in adult b lineage acute lymphoblastic leukemia. Immunol Invest. 2019;48(8):781–93.

    Article  PubMed  Google Scholar 

  220. Makowski L, Chaib M, Rathmell J. Immunometabolism: from basic mechanisms to translation. Immunol Rev. 2020;295(1):5–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Patel CH, Leone RD, Horton MR, Powell JD. Targeting metabolism to regulate immune responses in autoimmunity and cancer. Nat Rev Drug Discov. 2019;18(9):669–88.

    Article  CAS  PubMed  Google Scholar 

  222. Joffre O, Nolte M, Spörri R, Reis e Sousa C. Inflammatory signals in dendritic cell activation and the induction of adaptive immunity. Immunol Rev. 2009;227(1):234.

    Article  CAS  PubMed  Google Scholar 

  223. Krawczyk C, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis R, Cross J, Jung E, Thompson C, Jones R, et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010;115(23):4742.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Everts B, Amiel E. Huang SC-C, Smith AM, Chang C-H, Lam WY, Redmann V, Freitas TC, Blagih J, Van Der Windt GJW et al: TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKɛ supports the anabolic demands of dendritic cell activation. Nat Immunol. 2014;15(4):323–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Guak H, Al Habyan S, Ma E, Aldossary H, Al-Masri M, Won S, Ying T, Fixman E, Jones RG, McCaffrey LM, et al. Glycolytic metabolism is essential for CCR7 oligomerization and dendritic cell migration. Nat Commun. 2018;9(1):2463.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Bajwa G, DeBerardinis R, Shao B, Hall B, Farrar J, Gill M. Cutting edge: critical role of Glycolysis in human Plasmacytoid Dendritic cell antiviral responses. J immunol (Baltimore, Md : 1950). 2016;196(5):2004.

    Article  CAS  Google Scholar 

  227. Fekete T, Sütö M, Bencze D, Mázló A, Szabo A, Biro T, Bacsi A, Pazmandi K. Human plasmacytoid and monocyte-derived dendritic cells display distinct metabolic profile upon RIG-I activation. Front Immunol. 2018;9:3070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Xu O, Li X, Qu Y, Liu S, An J, Wang M, Sun Q, Zhang W, Lu X, Pi L, et al. Regulation of glucose transporter protein-1 and vascular endothelial growth factor by hypoxia inducible factor 1α under hypoxic conditions in Hep-2 human cells. Mol Med Rep. 2012;6(6):1418.

    Article  CAS  PubMed  Google Scholar 

  229. Boor P, Metselaar H, Mancham S, van der Laan L, Kwekkeboom J. Rapamycin has suppressive and stimulatory effects on human plasmacytoid dendritic cell functions. Clin Exp Immunol. 2013;174(3):389–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Wu D, Sanin D, Everts B, Chen Q, Qiu J, Buck M, Patterson A, Smith A, Chang C, Liu Z, et al. Type 1 interferons induce changes in core metabolism that are critical for immune function. Immunity. 2016;44(6):1325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Basit F, Mathan T, Sancho D, de Vries I. Human dendritic cell subsets undergo distinct metabolic reprogramming for immune response. Front Immunol. 2018;9:2489.

    Article  PubMed  PubMed Central  Google Scholar 

  232. Brombacher E, Everts B. Shaping of dendritic cell function by the metabolic micro-environment. Front Endocrinol. 2020;11:555.

    Article  Google Scholar 

  233. Grzes K, Sanin D, Kabat A, Stanczak M, Edwards-Hicks J, Matsushita M, Hackl A, Hässler F, Knoke K, Zahalka S, et al. Plasmacytoid dendritic cell activation is dependent on coordinated expression of distinct amino acid transporters. Immunity. 2021;54(11):2514.

    Article  CAS  PubMed  Google Scholar 

  234. Hurley H, Dewald H, Rothkopf Z, Singh S, Jenkins F, Deb P, De S, Barnes B, Fitzgerald-Bocarsly P. Frontline science: AMPK regulates metabolic reprogramming necessary for interferon production in human plasmacytoid dendritic cells. J Leukocyte Biol. 2021;109(2):299.

    Article  CAS  PubMed  Google Scholar 

  235. Caronni N, Simoncello F, Stafetta F, Guarnaccia C, Ruiz-Moreno JS, Opitz B, Galli T, Proux-Gillardeaux V, Benvenuti F. Downregulation of membrane trafficking proteins and lactate conditioning determine loss of dendritic cell function in lung cancer. Can Res. 2018;78(7):1685–99.

    Article  CAS  Google Scholar 

  236. Malinarich F, Duan K, Hamid R, Bijin A, Lin W, Poidinger M, Fairhurst A, Connolly J. High mitochondrial respiration and glycolytic capacity represent a metabolic phenotype of human tolerogenic dendritic cells. J Immunol (Baltimore, Md : 1950). 2015;194(11):5174.

    CAS  Google Scholar 

  237. Gottfried E, Kunz-Schughart L, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006;107(5):2013.

    Article  CAS  PubMed  Google Scholar 

  238. Raychaudhuri D, Bhattacharya R, Sinha BP, Liu CSC, Ghosh AR, Rahaman O, Bandopadhyay P, Sarif J, D’Rozario R, Paul S, et al. Lactate induces pro-tumor reprogramming in intratumoral plasmacytoid dendritic cells. Front Immunol. 1878;2019:10.

    Google Scholar 

  239. Seya T, Takeda Y, Takashima K, Yoshida S, Azuma M, Matsumoto M. Adjuvant immunotherapy for cancer: both dendritic cell-priming and check-point inhibitor blockade are required for immunotherapy. Proceedings of the Japan Academy, Series B. 2018;94(3):153–60.

    Article  CAS  Google Scholar 

  240. Aspord C, Tramcourt L, Leloup C, Molens JP, Leccia MT, Charles J, Plumas J. Imiquimod inhibits melanoma development by promoting pDC cytotoxic functions and impeding tumor vascularization. J Invest Dermatol. 2014;134(10):2551–61.

    Article  CAS  PubMed  Google Scholar 

  241. Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, Li X-D, Mauceri H, Beckett M, Darga T, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41(5):843–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, Woo SR, Lemmens E, Banda T, Leong JJ, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 2015;11(7):1018–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Aspord C, Charles J, Leccia MT, Laurin D, Richard MJ, Chaperot L, Plumas J. A novel cancer vaccine strategy based on HLA-A*0201 matched allogeneic plasmacytoid dendritic cells. PLoS ONE. 2010;5(5): e10458.

    Article  PubMed  PubMed Central  Google Scholar 

  244. Aspord C, Leccia MT, Salameire D, Laurin D, Chaperot L, Charles J, Plumas J. HLA-A(*)0201(+) plasmacytoid dendritic cells provide a cell-based immunotherapy for melanoma patients. J Invest Dermatol. 2012;132(10):2395–406.

    Article  CAS  PubMed  Google Scholar 

  245. Milhem M, Zakharia Y, Davar D, Buchbinder E, Medina T, Daud A, Ribas A, Niu J, Gibney G. Margolin K et al: 304 Intratumoral injection of CMP-001, a toll-like receptor 9 (TLR9) agonist, in combination with pembrolizumab reversed programmed death receptor 1 (PD-1) blockade resistance in advanced melanoma. J Immuno Ther Cancer. 2020;8:2998.

    Google Scholar 

  246. Cheng Y, Lemke-Miltner CD, Wongpattaraworakul W, Wang Z, Chan CHF, Salem AK, Weiner GJ, Simons AL. <i>In situ</i> immunization of a TLR9 agonist virus-like particle enhances anti-PD1 therapy. J Immunother Cancer. 2020;8(2): e000940.

    Article  PubMed  PubMed Central  Google Scholar 

  247. O’Day S, Perez C, Wise-Draper T, Hanna G, Bhatia S, Kelly C, Medina T, Laux D, Daud A, Chandra S, et al. Safety and preliminary efficacy of intratumoral cavrotolimod (AST-008), a spherical nucleic acid TLR9 agonist, in combination with pembrolizumab in patients with advanced solid tumors. J ImmunoTher Cancer. 2020;8(Suppl. 3).

  248. Ribas A, Medina T, Kummar S, Amin A, Kalbasi A, Drabick JJ, Barve M, Daniels GA, Wong DJ, Schmidt EV, et al. SD-101 in combination with Pembrolizumab in advanced melanoma: results of a phase Ib. Multicenter Study Cancer Discov. 2018;8(10):1250–7.

    Article  CAS  PubMed  Google Scholar 

  249. Ribas A, Medina T, Kirkwood JM, Zakharia Y, Gonzalez R, Davar D, Chmielowski B, Campbell KM, Bao R, Kelley H, et al. Overcoming PD-1 blockade resistance with CpG-a toll-like receptor 9 agonist vidutolimod in patients with metastatic melanoma. Cancer Discov. 2021;11(12):2998–3007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Smits ELJM, Ponsaerts P, Berneman ZN, Van Tendeloo VFI. The use of TLR7 and TLR8 ligands for the enhancement of cancer immunotherapy. Oncologist. 2008;13(8):859–75.

    Article  CAS  PubMed  Google Scholar 

  251. Panelli M, Stashower M, Slade H, Smith K, Norwood C, Abati A, Fetsch P, Filie A, Walters S, Astry C, et al. Sequential gene profiling of basal cell carcinomas treated with imiquimod in a placebo-controlled study defines the requirements for tissue rejection. Genome Biol. 2007;8(1):R8.

    Article  PubMed  PubMed Central  Google Scholar 

  252. Patinote C, Karroum N, Moarbess G, Cirnat N, Kassab I, Bonnet P, Deleuze-Masquéfa C. Agonist and antagonist ligands of toll-like receptors 7 and 8: Ingenious tools for therapeutic purposes. Europ J Med Chem. 2020;193:112238.

    Article  CAS  Google Scholar 

  253. Singh M, Khong H, Dai Z, Huang X-F, Wargo JA, Cooper ZA, Vasilakos JP, Hwu P, Overwijk WW. Effective innate and adaptive antimelanoma immunity through localized TLR7/8 activation. J Immunol. 2014;193(9):4722–31.

    Article  CAS  PubMed  Google Scholar 

  254. Janku F, Han S-W, Doi T, Amatu A, Ajani JA, Kuboki Y, Cortez A, Cellitti SE, Mahling PC, Subramanian K, et al. Preclinical characterization and phase I study of an Anti–HER2-TLR7 immune-stimulator antibody conjugate in patients with HER2+ malignancies. Cancer Immunol Res. 2022;10(12):1441–61.

    Article  CAS  PubMed  Google Scholar 

  255. Vollmer J, Weeratna R, Payette P, Jurk M, Schetter C, Laucht M, Wader T, Tluk S, Liu M, Davis HL, et al. Characterization of three CpG oligodeoxynucleotide classes with distinct immunostimulatory activities. Eur J Immunol. 2004;34(1):251–62.

    Article  CAS  PubMed  Google Scholar 

  256. Li T, Hua C, Yue W, Wu J, Lv X, Wei Q, Zhu S, Zang G, Cui J, Liu Y, et al. Discrepant antitumor efficacies of three CpG oligodeoxynucleotide classes in monotherapy and co-therapy with PD-1 blockade. Pharmacol Res. 2020;161:105293.

    Article  CAS  PubMed  Google Scholar 

  257. Lemke-Miltner CD, Blackwell SE, Yin C, Krug AE, Morris AJ, Krieg AM, Weiner GJ. Antibody opsonization of a TLR9 agonist-containing virus-like particle enhances in situ immunization. J Immunol. 2020;204(5):1386–94.

    Article  CAS  PubMed  Google Scholar 

  258. Haymaker C, Johnson DH, Murthy R, Bentebibel S-E, Uemura MI, Hudgens CW, Safa H, James M, Andtbacka RHI, Johnson DB, et al. Tilsotolimod with Ipilimumab drives tumor responses in anti–PD-1 refractory melanoma. Cancer Discov. 2021;11(8):1996–2013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Molenkamp B, van Leeuwen P, Meijer S, Sluijter B, Wijnands P, Baars A, van den Eertwegh A, Scheper R, de Gruijl T. Intradermal CpG-B activates both plasmacytoid and myeloid dendritic cells in the sentinel lymph node of melanoma patients. Clin Cancer Res Off J Am Assoc Cancer Res. 2007;13(10):2961.

    Article  CAS  Google Scholar 

  260. Pashenkov M, Goess G, Wagner C, Hormann M, Jandl T, Moser A, Britten CM, Smolle J, Koller S, Mauch C, et al. Phase II trial of a toll-like receptor 9-activating oligonucleotide in patients with metastatic melanoma. J Clin Oncol. 2006;24(36):5716–24.

    Article  CAS  PubMed  Google Scholar 

  261. Wang S, Campos J, Gallotta M, Gong M, Crain C, Naik E, Coffman R, Guiducci C. Intratumoral injection of a CpG oligonucleotide reverts resistance to PD-1 blockade by expanding multifunctional CD8+ T cells. Proc Natl Acad Sci USA. 2016;113(46):E7240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Sato-Kaneko F, Yao S, Ahmadi A, Zhang SS, Hosoya T, Kaneda MM, Varner JA, Pu M, Messer KS, Guiducci C. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight. 2017;2(18):93397.

    Article  PubMed  Google Scholar 

  263. Lombardi VC, Khaiboullina SF, Rizvanov AA. Plasmacytoid dendritic cells, a role in neoplastic prevention and progression. Eur J Clin Invest. 2015;45(Suppl 1):1–8.

    Article  CAS  PubMed  Google Scholar 

  264. Huang B, Zhao J, Unkeless JC, Feng ZH, Xiong H. TLR signaling by tumor and immune cells: a double-edged sword. Oncogene. 2008;27(2):218–24.

    Article  CAS  PubMed  Google Scholar 

  265. Grimm M, Kim M, Rosenwald A, Heemann U, Germer C, Waaga-Gasser A, Gasser M. Toll-like receptor (TLR) 7 and TLR8 expression on CD133+ cells in colorectal cancer points to a specific role for inflammation-induced TLRs in tumourigenesis and tumour progression. Respir Res (Oxford, England : 1990). 2010;46(15):2849–57.

    CAS  Google Scholar 

  266. Tanaka J, Sugimoto K, Shiraki K, Tameda M, Kusagawa S, Nojiri K, Beppu T, Yoneda K, Yamamoto N, Uchida K, et al. Functional cell surface expression of toll-like receptor 9 promotes cell proliferation and survival in human hepatocellular carcinomas. Int J Oncol. 2010;37(4):805–14.

    CAS  PubMed  Google Scholar 

  267. Droemann D, Albrecht D, Gerdes J, Ulmer AJ, Branscheid D, Vollmer E, Dalhoff K, Zabel P, Goldmann T. Human lung cancer cells express functionally active Toll-like receptor 9. Respir Res. 2005;6(1):1.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Schmausser B, Andrulis M, Endrich S, Müller-Hermelink H, Eck M. Toll-like receptors TLR4, TLR5 and TLR9 on gastric carcinoma cells: an implication for interaction with Helicobacter pylori. International journal of medical microbiology : IJMM. 2005;295(3):179–85.

    Article  CAS  PubMed  Google Scholar 

  269. Lee JW, Choi JJ, Seo ES, Kim MJ, Kim WY, Choi CH, Kim TJ, Kim BG, Song SY, Bae DS. Increased toll-like receptor 9 expression in cervical neoplasia. Mol Carcinog. 2007;46(11):941–7.

    Article  CAS  PubMed  Google Scholar 

  270. Ilvesaro JM, Merrell MA, Swain TM, Davidson J, Zayzafoon M, Harris KW, Selander KS. Toll like receptor-9 agonists stimulate prostate cancer invasion in vitro. Prostate. 2007;67(7):774–81.

    Article  CAS  PubMed  Google Scholar 

  271. Boulé MW, Broughton C, Mackay F, Akira S, Marshak-Rothstein A, Rifkin IR. Toll-like Receptor 9–Dependent and –Independent Dendritic Cell Activation by Chromatin-Immunoglobulin G Complexes. J Exp Med. 2004;199(12):1631–40.

    Article  PubMed  PubMed Central  Google Scholar 

  272. Barrat FJ, Meeker T, Gregorio J, Chan JH, Uematsu S, Akira S, Chang B, Duramad O, Coffman RL. Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like receptors and may promote systemic lupus erythematosus. J Exp Med. 2005;202(8):1131–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Barrow A, Cella M, Edeling M, Khan M, Cervantes-Barragan L, Bugatti M, Schmedt C, Vermi W, Colonna M. Cutting edge: PDGF-DD binding to NKp44 costimulates TLR9 signaling and proinflammatory cytokine secretion in human plasmacytoid dendritic cells. J Immunol (Baltimore, Md : 1950). 2024;212(3):369–74.

    CAS  Google Scholar 

  274. Bakhoum SF, Ngo B, Laughney AM, Cavallo J-A, Murphy CJ, Ly P, Shah P, Sriram RK, Watkins TBK, Taunk NK, et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature. 2018;553(7689):467–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Mackenzie KJ, Carroll P, Martin C-A, Murina O, Fluteau A, Simpson DJ, Olova N, Sutcliffe H, Rainger JK, Leitch A, et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature. 2017;548(7668):461–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Mclaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, Harrington KJ. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–17.

    Article  CAS  PubMed  Google Scholar 

  277. Lohard S, Bourgeois N, Maillet L, Gautier F, Fétiveau A, Lasla H, Nguyen F, Vuillier C, Dumont A, Moreau-Aubry A, et al. STING-dependent paracriny shapes apoptotic priming of breast tumors in response to anti-mitotic treatment. Nat Commun. 2020;11(1):259.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Storozynsky Q, Hitt MM. The impact of radiation-induced DNA damage on cGAS-STING-mediated immune responses to cancer. Int J Mol Sci. 2020;21(22):8877.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Woo S-R, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MYK, Duggan R, Wang Y, Barber GN, Fitzgerald KA, et al. STING-dependentcytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41(5):830–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  280. Diamond JM, Vanpouille-Box C, Spada S, Rudqvist N-P, Chapman JR, Ueberheide BM, Pilones KA, Sarfraz Y, Formenti SC, Demaria S. Exosomes shuttle TREX1-sensitive IFN-stimulatory dsDNA from irradiated cancer cells to DCs. Cancer Immunol Res. 2018;6(8):910–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, Chen ZJ. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc Natl Acad Sci. 2017;114(7):1637–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Demaria O, De Gassart A, Coso S, Gestermann N, Di Domizio J, Flatz L, Gaide O, Michielin O, Hwu P, Petrova TV, et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc Natl Acad Sci. 2015;112(50):15408–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  283. Corrales L, Gajewski TF. Endogenous and pharmacologic targeting of the STING pathway in cancer immunotherapy. Cytokine. 2016;77:245–7.

    Article  PubMed  Google Scholar 

  284. Li T, Cheng H, Yuan H, Xu Q, Shu C, Zhang Y, Xu P, Tan J, Rui Y, Li P, et al. Antitumor activity of cGAMP via stimulation of cGAS-cGAMP-STING-IRF3 mediated innate immune response. Sci Rep. 2016;6(1):19049.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Shae D, Becker KW, Christov P, Yun DS, Lytton-Jean AKR, Sevimli S, Ascano M, Kelley M, Johnson DB, Balko JM, et al. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. Nat Nanotechnol. 2019;14(3):269–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang S-Y, Tran J-L, Moore P, Lehmann S, Eberl HC, et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018;564(7736):439–43.

    Article  CAS  PubMed  Google Scholar 

  287. Shih AY, Damm-Ganamet KL, Mirzadegan T. Dynamic structural differences between human and mouse STING lead to differing sensitivity to DMXAA. Biophys J. 2018;114(1):32–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Conlon J, Burdette DL, Sharma S, Bhat N, Thompson M, Jiang Z, Rathinam VAK, Monks B, Jin T, Xiao TS, et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J Immunol. 2013;190(10):5216–25.

    Article  CAS  PubMed  Google Scholar 

  289. Sivick KE, Desbien AL, Glickman LH, Reiner GL, Corrales L, Surh NH, Hudson TE, Vu UT, Francica BJ, Banda T, et al. Magnitude of therapeutic STING activation determines CD8+ T cell-mediated anti-tumor immunity. Cell Rep. 2018;25(11):3074-3085.e3075.

    Article  CAS  PubMed  Google Scholar 

  290. Mcintosh JA, Liu Z, Andresen BM, Marzijarani NS, Moore JC, Marshall NM, Borra-Garske M, Obligacion JV, Fier PS, Peng F, et al. A kinase-cGAS cascade to synthesize a therapeutic STING activator. Nature. 2022;603(7901):439–44.

    Article  CAS  PubMed  Google Scholar 

  291. Gogoi H, Mansouri S, Jin L. The age of cyclic dinucleotide vaccine adjuvants. Vaccines. 2020;8(3):453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Li A, Yi M, Qin S, Song Y, Chu Q, Wu K. Activating cGAS-STING pathway for the optimal effect of cancer immunotherapy. J Hematol Oncol. 2019;12(1):35.

    Article  PubMed  PubMed Central  Google Scholar 

  293. Xia T, Konno H, Ahn J, Barber GN. Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Rep. 2016;14(2):282–97.

    Article  CAS  PubMed  Google Scholar 

  294. Konno H, Yamauchi S, Berglund A, Putney RM, Mulé JJ, Barber GN. Suppression of STING signaling through epigenetic silencing and missense mutation impedes DNA damage mediated cytokine production. Oncogene. 2018;37(15):2037–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. Xia T, Konno H, Barber GN. Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis. Can Res. 2016;76(22):6747–59.

    Article  CAS  Google Scholar 

  296. He L, Xiao X, Yang X, Zhang Z, Wu L, Liu Z. STING signaling in tumorigenesis and cancer therapy: A friend or foe? Cancer Lett. 2017;402:203–12.

    Article  CAS  PubMed  Google Scholar 

  297. Song S, Peng P, Tang Z, Zhao J, Wu W, Li H, Shao M, Li L, Yang C, Duan F, et al. reased expression of STING predicts poor prognosis in patients with gastric cancer. Sci Rep. 2017;7(1):39858.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  298. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, Jacob L, Patwa R, Shah H. Xu K et al: Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016;533(7604):493–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Lemos H, Mohamed E, Huang L, Ou R, Pacholczyk G, Arbab AS, Munn D, Mellor AL. STING promotes the growth of tumors characterized by low antigenicity via IDO activation. Can Res. 2016;76(8):2076–81.

    Article  CAS  Google Scholar 

  300. Grabosch S, Bulatovic M, Zeng F, Ma T, Zhang L, Ross M, Brozick J, Fang Y, Tseng G, Kim E, et al. Cisplatin-induced immune modulation in ovarian cancer mouse models with distinct inflammation profiles. Oncogene. 2019;38(13):2380–93.

    Article  CAS  PubMed  Google Scholar 

  301. Fu J, Kanne D, Leong M, Glickman L, McWhirter S, Lemmens E, Mechette K, Leong J, Lauer P, Liu W, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7(283):283.

    Article  Google Scholar 

  302. Filin IY, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV. Recent advances in experimental dendritic cell vaccines for cancer. Fronti Oncol. 2021;11:730824.

    Article  CAS  Google Scholar 

  303. Tel J, Aarntzen EH, Baba T, Schreibelt G, Schulte BM, Benitez-Ribas D, Boerman OC, Croockewit S, Oyen WJ, van Rossum M, et al. Natural human plasmacytoid dendritic cells induce antigen-specific T-cell responses in melanoma patients. Cancer Res. 2013;73(3):1063–75.

    Article  CAS  PubMed  Google Scholar 

  304. Charles J, Chaperot L, Hannani D, Bruder Costa J, Templier I, Trabelsi S, Gil H, Moisan A, Persoons V, Hegelhofer H, et al. An innovative plasmacytoid dendritic cell line-based cancer vaccine primes and expands antitumor T-cells in melanoma patients in a first-in-human trial. OncoImmunology. 2020;9(1):1738812.

    Article  PubMed  PubMed Central  Google Scholar 

  305. Van Beek JJP, Flórez-Grau G, Gorris MAJ, Mathan TSM, Schreibelt G, Bol KF, Textor J, De Vries IJM. Human pDCs are superior to cDC2s in attracting cytolytic lymphocytes in melanoma patients receiving DC vaccination. Cell Rep. 2020;30(4):1027-1038.e1024.

    Article  PubMed  Google Scholar 

  306. Westdorp H, Creemers JHA, Van Oort IM, Schreibelt G, Gorris MAJ, Mehra N, Simons M, De Goede AL, Van Rossum MM, Croockewit AJ, et al. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer. J Immunother Cancer. 2019;7(1):302.

    Article  PubMed  PubMed Central  Google Scholar 

  307. Plumas J. Harnessing dendritic cells for innovative therapeutic cancer vaccines. Curr Opin Oncol. 2022;34(2):161.

    Article  CAS  PubMed  Google Scholar 

  308. Sakakura K, Chikamatsu K, Takahashi K, Whiteside TL, Furuya N. Maturation of circulating dendritic cells and imbalance of T-cell subsets in patients with squamous cell carcinoma of the head and neck. Cancer Immunol Immunother. 2006;55(2):151–9.

    Article  PubMed  Google Scholar 

  309. Abolhalaj M, Askmyr D, Sakellariou C, Lundberg K, Greiff L, Lindstedt M. Profiling dendritic cell subsets in head and neck squamous cell tonsillar cancer and benign tonsils. Scient Rep. 2018;8(1):8030.

    Article  Google Scholar 

Download references

Acknowledgements

The authors acknowledge “Studio Moretto Group Srl” for professional editing service.

Funding

The publication of this article was supported by grants from the Associazione Italiana per la Ricerca sul Cancro [Italian Association for Cancer Research] to WV (IG-23179)]. MM was supported by an AIRC fellowship for Italy (fellowship AIRC Professor Felice Martinelli, ID 28065).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: MM and WV; writing-original draft preparation: MM, GF and LG; writing-review and editing: MM, FF, and WV; supervision: WV; funding acquisition: WV. All authors read and approved the final manuscript.

Corresponding author

Correspondence to William Vermi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Monti, M., Ferrari, G., Gazzurelli, L. et al. Plasmacytoid dendritic cells at the forefront of anti-cancer immunity: rewiring strategies for tumor microenvironment remodeling. J Exp Clin Cancer Res 43, 196 (2024). https://doi.org/10.1186/s13046-024-03121-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13046-024-03121-9

Keywords