Skip to main content

Impaired dendritic cell functions in lung cancer: a review of recent advances and future perspectives

Abstract

Lung cancer is the leading cause of cancer mortality worldwide. Dendritic cells (DCs) are the key factors providing protective immunity against lung tumors and clinical trials have proven that DC function is reduced in lung cancer patients. It is evident that the immunoregulatory network may play a key role in the failure of the immune response to terminate tumors. Lung tumors likely employ numerous strategies to suppress DC-based anti-tumor immunity. Here, we summarize the recent advances in our understanding on lung tumor-induced immunosuppression in DCs, which affects the initiation and development of T-cell responses. We also describe which existing measures to restore DC function may be useful for clinical treatment of lung tumors. Furthering our knowledge of how lung cancer cells alter DC function to generate a tumor-supportive environment will be essential in order to guide the design of new immunotherapy strategies for clinical use.

Introduction

Lung cancer is the leading cause of cancer death around the world [1]. More than 85% of lung cancers are non-small-cell lung cancer (NSCLC) [2]. The 5-year overall survival rate for patients with lung cancer is less than 15% and that for patients with NSCLC clinically diagnosed as stage IV is less than 5% [3]. The most common treatment for lung cancer, such as chemotherapy and radiotherapy, has shown limited effectiveness in preventing tumor progression. It is believed that recurrence after surgical resection and chemotherapy is the main cause of lung cancer death [4, 5]. Therefore, improving both diagnostic and therapeutic methods is essential for improving public health with respect to such relapses.

Developing immunotherapy strategies that can induce long-term protective immune responses against tumor-associated antigens is an emerging research topic. Such therapeutic strategies are especially vital when conventional therapies become ineffective [6]. Recent advances in immunotherapy for lung cancer include targeting costimulatory blockade and immune cell-based vaccination [7,8,9]. A blockade of the immune checkpoint markers, such as programmed cell death 1 (PD-1), programmed cell death 1 ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated antigen-4 (CTLA4), resulted in a significantly prolonged survival rate, indicating a systemic anti-tumor immune deficiency in lung cancers [10,11,12]. However, the expression of these immune checkpoint markers differs from one cancer to another, limiting the general application of the approaches targeting them. For example, patients with low PD-1 expression have poor responses to anti-PD-1 treatment [12,13,14]. For this reason, other immunotherapeutic strategies must be developed to promote consistent therapeutic effects.

Dendritic cells (DCs) are crucial for the activation of antigen-specific CD8 T lymphocytes, a pivotal step in the initiation of the innate and adaptive immune responses, which are essential for tumor cell clearance. Previous studies have demonstrated that PD-1-deficient DCs had a stronger ability to induce antigen-specific CD8+ T cell proliferation than wild-type DCs in vivo [15]. As a nano-sized vesicle, exosomes derived from different cell types selectively enrich the proteins associated with specific cell functions [16, 17]. Moreover, DC-derived exosomes can be used for maintenance immunotherapy in NSCLC patients whose disease responded or were stabilized after induction chemotherapy, as previously described [18]. Thus, DC mobilization may be an effective treatment strategy for cancer [19, 20]. Anti-tumor effects of DCs can be reduced by several factors, including low DC count, low antigen presentation efficiency of tumor-infiltrating DCs, and weak ability of DC to migrate into tumor mass [21, 22]. A previous study has shown that the maturation rate of DCs in patients with lung tumors was significantly lower than that in healthy controls [23]. In addition to enhancing the antigen-presenting ability of DCs, blockade of the immunosuppression signal between lung tumor cells and DCs is also essential for the development of DC-based anti-tumor therapies. In this review, we summarized the mechanisms involved in lung cancer-induced DC inhibition and the recent advances in DC-based immunotherapy. Additionally, we addressed the potential approaches for restoring DC function in lung cancers, which is the key for designing more successful DC-based anti-tumor therapy.

Origin of DCs

Myeloid cells include different types of innate immune cells that can clear damaged cells and promote the recruitment of immune effector cells. In the tumor microenvironment (TME), tumor-infiltrating myeloid cells (TIMs) play a major role in anti-tumor response [24, 25]. TIMs mainly consist of granulocytes and mononuclear phagocytes. These cells share the ability to present tumor-associated antigens to T cells, which are closely related to tumor progression and response to immunotherapy [26]. Among all TIMs, DCs are best equipped to activate T cells.

DCs are professional antigen-presenting immune cells and are distributed throughout the body. They originate from the bone marrow, circulate in the blood, and have two ultimate fates, either enter the lymphoid nodes to act as lymphoid DCs or enter peripheral tissues to differentiate into non-lymphoid DCs [27]. DCs are generated from both lymphoid and myeloid progenitors in the bone marrow, which produce conventional DCs (cDCs) and plasmacytoid DCs (pDCs), respectively, in adoptive transfer experiments [28]. Among hematopoietic stem cells, monocyte-DC progenitors (MDPs) can give rise to common myeloid progenitors (CMPs), including a subset of CMPs that express colony stimulating factor 1 receptor (FMS)-like tyrosine kinase 3 (FLT3) [28]. FLT3 expression in CMPs is required to maintain cDC developmental potential [29]. MDPs can also give rise to common monocyte progenitors (cMoPs) that differentiate into monocytes. Ly6C+ circulating monocytes can differentiate into monocyte-derived DCs (MoDCs) under the appropriate circumstances [30, 31]. In clinical trials, MoDCs were shown to be easy to be induced from patient’s peripheral blood monocytes and were capable of inducing tumor-specific immune responses when co-cultured with the corresponding antigens [32].

Several DC subsets which can be broadly divided into cDCs and pDCs have been identified. Detailed analyses of murine and human lungs have revealed that two main subsets of cDCs are present in a steady state [33]. In murine lungs, these two subsets express CD103 and CD11b, respectively. CD103+ cDCs are regulated by interferon regulatory factor 8 (IRF8) and CD11b+ cDCs are regulated by IRF4 [34, 35]. CD103+ DCs have been shown to control the activation of local CD8+ T cell in TME as active APCs. CD11b+ DCs initiate Th2- and Th17-biased immune responses, thus making only a minor contribution to tumor clearance [36, 37]. Equivalent cDC subsets in human lungs expressing CD141 and CD1c, respectively, have similar functions [38]. On the other hand, pDCs are mainly characterized by the production of large amounts of type I interferon (IFN) [39]. The expression of toll-like receptor 7 and 9 (TLR7 and TLR9) on pDCs confers antiviral activity [40]. Relative to cDCs, pDCs show less antigen-presenting ability and seem to play an important role in maintaining self-tolerance [41]. The origin of DCs is illustrated in Fig. 1.

Fig. 1
figure 1

Origin of DCs. MDPs among hematopoietic stem cells give rise to common FLT3-expressing CMPs, which are the precursors of pre-cDCs and pDCs. Monocytes can differentiate into MODCs under the appropriate circumstances and Pre-cDCs circulate into lung tissue and differentiate into different classes of DCs. MDP: Monocyte-DC progenitors; FLT3: colony stimulating factor 1 receptor (FMS)-like tyrosine kinase 3; pDC: plasmacytoid DC; cDC: conventional DC; MODCs: monocyte-derived DCs: CMPs: common myeloid progenitors

Lung cancer-induced DC suppression can impede immune clearance

Functional DCs are excluded from lung tumor lesions

The antigen presenting ability of DCs plays an important role in the activation of anti-tumor T cell. DC metabolism controls T cell polarization in the lungs [42]. Under normal circumstances, original DCs infiltrate into the lung from the bone marrow and differentiate into two main subsets of mature DCs (CD11b+ and CD103+ DCs). Mature DCs express a higher level of co-stimulatory molecules (CD40/80/86), and an elevated cytokine levels indicate a potent T cell activating ability. The prominent expression of major histocompatibility complex (MHC) type II molecules on CD11b+ DCs facilitates the activation of CD4 T cells. In contrast, CD103+ DCs, which have been recently identified as active APCs in the TME, preferentially induce CD8+ T cell responses via MHC type I molecules [43,44,45,46]. However, compared to their counterparts in the peritumoral lung, lung tumor-infiltrating DCs were found to show increased expression of CD11b [47]. Moreover, a smaller population of CD103+ DCs was observed within lung tumors in a mouse model [47]. Furthermore, a paired single-cell transcriptional analysis of tumorous and non-tumorous human lungs also revealed two clusters of DCs: one cluster consisted of CD141+ DCs, which mainly interact with CD8+ T cells. The other cluster contained CD1c+ DCs, which have an increased potential to activate CD4+ T cells. Compared with non-tumorous lung tissues, the proportion of CD141+ DCs is significantly lower in tumorous tissues [38]. It is believed that human CD141+ DCs and CD1c+ DCs are similar to murine CD103+ DCs and CD11b+ DCs [48]. Taken together, the DC subset which could activate CD8+ T cell was reduced in tumor tissues. These studies demonstrate that lung cancers dynamically exclude functional DCs from the tumor region to support malignant progression.

Immunosuppressive pDCs are recruited into the surrounding tissues of lung tumors

The key role of pDCs in innate immunity has been clearly elucidated over the past few decades [49, 50]. Previous studies have shown that inactive pDCs are present in lung cancers associated with poor patient prognosis [51, 52]. Tumor-associated pDCs show impaired type I IFN production [53]. This IFN has been shown to participate in cytotoxic, immunosuppressive, and anti-tumor responses [53, 54]. The activation of intratumoral pDCs by TLR-9 agonist may induce melanoma regression via natural killer (NK) cell-dependent pathways in a C57BL/6 melanoma model [55]. Moreover, the secretion of C–C chemokine receptor type 5 (CCR5) by pDCs contributes to NK cell recruitment in tumors [55]. cDC cross-presentation is thought to depend on NK cell recruitment [55]. However, TLR-9 activation in lung tumor-bearing mice leads to the recruitment of regulatory T cells (Tregs), thus contributing to an immunosuppressive environment around the tumor [56]. Narusawa et al. [57] used imiquimod, a TLR-7 ligand, to activate pDCs in a Lewis lung carcinoma model. The addition of imiquimod significantly improved the antitumor phenotype in pDCs and decreased the proportion of Tregs in tumor-draining lymph nodes. An earlier study using a mouse model showed that low doses of lipopolysaccharides (LPS) may increase the lung tumor burden via pDC regulation [58].

Shi et al. [59] used flow cytometry to examine the pDC present levels in peripheral blood from 52 NSCLC patients and 52 healthy controls. Patients with higher tumor stages had higher pDC levels than those with lower stages. Rosalinda Sorrentino and colleagues [60] isolated pDCs from NSCLC tissues and found that higher percentages of immunosuppressive-phenotype pDCs were recruited in lung tumor regions with high expression of CD33 and PD-L1, which makes a contribution to the immunosuppressive tumor microenvironment. Moreover, these pDCs were able to produce high levels of interleukin 1α (IL-1α) in a melanoma 2 (AIM2)-dependent manner, which facilitated tumor cell proliferation. In many cases, the depletion of pDCs may reverse the immunosuppressive environment and decrease the lung tumor burden, thereby indicating the role of pDCs in promoting lung tumor processes. The interaction between lung tumors and pDCs is depicted in Fig. 2.

Fig. 2
figure 2

Peritumoral pDCs contribute to lung tumor progression. Impaired type I IFN secretion may faciliate tumor escape. CD33 and PD-L1 are upregulated in pDCs and contribute to immunosuppression. Lung tumors can activate AIM2 and inhibit CCR5 to construct a tumor-supportive microenvironment. pDC: plasmacytoid DC; cDC: conventional DC; NK: natural killer; IFN: interferon; CCR5: C–C chemokine receptor type 5; TLR: toll-like receptor; AIM2: melanoma 2; PD-L1: programmed cell death 1 ligand 1; IL-1α: interleukin 1α

Regulatory DCs (regDCs) are induced by lung tumors

cDCs play a fundamental role by presenting antigens during the induction of immune responses. However, the tumor cells can reprogram cDCs into Gr-1 or Gr-1+ regDCs in tumorous lung tissue [61]. regDCs may inhibit antitumor responses and support tumor growth, thus facilitating lung carcinoma development [61]. The immunosuppressive mechanisms of regDCs have been examined in various studies. Spallanzani et al. [62] showed that regDCs silence IFN-γ secretion by NK cells via reducing IL-18 secretion and by active cell-to-cell mechanisms mediated by IL-10. These mechanisms may promote the evasion of antitumor responses. In addition, Zhang et al. [63] demonstrated that regDCs can induce a higher percentage of Tregs to maintain immune tolerance during hepatocytes transplantation. However, under tumor conditions, the outcomes induced by regDCs could impede tumor clearance.

Downregulation of DC effector molecules in lung tumors

The unbalanced differentiation of DC precursors in TME has been demonstrated in many studies [64, 65]. After culturing DCs in the medium with serum from lung tumor patients, the expression of co-stimulatory molecules, such as CD40, CD80, CD86, MHC type II and IL-12, were disrupted. Moreover, these DCs were unable to present antigens to activate T cell responses [66]. Clinical research involving lung cancers indicated that the maturation rate of DCs in tumor tertiary lymphoid structures was significantly associated with long-term survival of patients [67].

Caronni et al. [68] demonstrated that DCs conditioned by lung TME exhibit down-regulation of soluble N-ethylmaleimide-sensitive factor accessory protein receptor, vesicle associated membrane protein3 (SNARE VAMP3), which is required for antigen cross-presentation and DC-mediate tumor rejection. Additionally, it was shown that lactic acid in TME inhibited TLR3 and stimulator of IFN genes (STING) signaling, thus inhibiting the secretion of IL-12 and IFN-I by DCs. Loss of antigen cross-presentation and cytokine secretion abilities may serve to limit the anti-tumor responses induced by DCs.

Both the nuclear factor kappa-B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways were suppressed in an in vitro model of DC dysfunction using NSCLC patient’s serum. Altered expression of functional cluster genes associated with DC differentiation was observed in this model [69]. This finding indicated that lung cancer cells can release soluble factors which could impair DC differentiation into the peripheral blood. As NF-κB suppression has a negative impact on DC activity, short hairpin RNA-based adjuvant targeting inhibitory proteins of NF-κB (IκBα), a suppressor of NF-κB, has been developed to activate DCs under tumor conditions [70]. Furthermore, NF-κB expression in lung tumor cells is essential for primary tumor growth. In prior studies, inhibition of the NF-κB pathway could prolong the survival rate of patients with a defined subset of lung adenocarcinomas [71]. Recent studies have also shown that blockading NF-κB signaling in the lung epithelium can prevent lung cancer development, but the efficiency of this treatment was neutralized by plasma IL-1β levels [72]. The complex mechanisms involved in NF-κB regulation have not yet been fully identified, and how this “double-edged sword” can be fully utilized in lung cancer therapies requires further investigation.

Immunosuppressive molecules secreted by lung tumor-derived DCs

Higher immunosuppression marker expression on DCs was also detected in lung cancers compared to the healthy controls. B7-H3 is a member of the programmed death ligand (PD-L) family, which is broadly expressed on lymphoid organs with the dual functions of co-inhibiting and co-stimulating T cells. Relative to normal lung tissues, B7-H3 is up-regulated in tumor-derived DCs [23]. A blockade of B7-H3 can restore the T cell stimulatory ability of NSCLC-derived DCs, thus indicating the crucial role it plays in mediating T cell suppression by DCs under tumor conditions. Radiofrequency ablation has been shown to result in T cell proliferation and a reduction of the B7-H3+ DC count in the peripheral blood, further highlighting the immunosuppressive effect of this molecule under lung cancer conditions [73]. In contrast, the overexpression of B7-H3 in lung cancer tissues mediates abnormal lipid metabolism to support the development of the tumors [74]. Some researchers are trying to develop B7-H3 as a diagnostic biomarker and apply the blockade of B7-H3 as a novel therapeutic approach for the treatment of lung cancer [75, 76].

MicroRNAs (miRNAs) belong to a family of small non-coding RNAs that are individually capable of regulating cell biological pathways, many of which are involved in immune regulation and tumor development [77]. Full transcriptome sequencing of lung tumor-derived DCs revealed a set of consistently dysregulated miRNAs, such as miR-301a and miR-31 [47]. Overexpression of miRNA-301a can suppress the IL-12 secretion in DCs, while decreased expression of miRNA-301a can result in decrease IFN-γ release from antigen-specific cytotoxic T cells and delayed lung tumor growth [78]. Expression of miRNA-31 in lung cancers is related to lower survival rate, drug resistance, lymph node metastasis, and perturbation of the cell cycle [78]. miRNA-31-3p-overexpressing DCs induce pro-invasive lung cancer shape changes indicative of increased invasive behavior [78]. These results indicate that lung tumors can escape from immune surveillance, in part, by reprogramming miRNA expression in DCs.

T-cell immunoglobulin and mucin-domain 3 (Tim3) is preferentially expressed in Th1 and Th17 cells with a negative impact on immune activation [79]. However, Tim3 is also expressed in myeloid cells, including macrophages and DCs. Previous studies have shown that DCs in the lung tumor tissues have a higher expression of Tim3 than normal DCs [80]. Activation of the Tim3 pathway in DCs can suppress nucleic acids transportation, and thereby limit the nucleic acid-mediated immune response [81].

Drivers of DC anergy in lung cancer

It is clear that lung tumors can induce various DC functional changes to promote tumor processes, but how this occurs and which specific signal or signals from the tumor can interact with DCs is still unknown. To gain information related to these phenomena, researchers have performed diverse studies on lung tumor-derived DCs. During cancer development, protein-glycan interactions can influence many processes. Galectin 1 was the first discovered protein from the glycan-binding family. Its overexpression in several tumors is related to the metastasis [82]. Recent studies have determined that higher secreted amounts of Galectin 1 by lung cancer cells can alter the phenotypes of MoDCs and impair all reactive T cell response by regulating the activity of the inhibitor of DNA binding 3 (ID3), which can induce IL-10 autocrine [83].

High mobility group box-1 protein (HMGB1) is a nuclear protein that is highly correlated with metastasis in multiple tumors) [84]. Extracellular HMGB1 can influence the recruitment and differentiation of antigen-presenting cells, thereby leading to the suppression of T cell-dependent immune response. Additionally, the upregulation of HMGB1 in NSCLC can reduce the sensitivity of tumor cells to chemotherapy regents [85,86,87,88]. Furthermore, HMGB1-mediated production of thymic stromal lymphopoietin (TSLP) by tumor cells can modulate DCs via the TSLP receptor under physiological conditions. Interaction of HMGB1 with its receptors (i.e. RAGE: receptor for advanced glycation end products, TLR2) together with the TSLP/TSLPR axis on DCs can induce the activation of Tregs and HMGB1, which can then interact with Tim3 to facilitate immunosuppression [80, 89]. In previous studies, HMGB1 inhibitors have been used for anticancer therapy and were able to reduce the percentage of Treg-activating DCs to enhance immune responses in a breast cancer model [90, 91]. How this treatment affects DC function in lung tumors, however, still requires further research. Moreover, exosomes from Lewis lung carcinomas block DC differentiation and induce cell apoptosis. They also induce the expression of PD-L1 in cDCs and play a role in DC-associated immune suppression [92].

Progress in the clinical application of DC function restoration

The first clinical trial on therapeutic cancer vaccine was carried out in 1998 on patients with melanoma [93]. Since the activation of the antigen-presenting ability of DCs is directly related with the induction of the CD8+ T cell response, researchers have focused on the modification of DCs in order to enhance the anti-tumor immune responses in vivo [94,95,96]. Many studies have demonstrated that most malignant tumors, including lung cancers, can produce a variety of factors that suppress anti-tumor immunity. Moreover, they have also shown that the maturation rate of DCs in patients with lung cancer is independently associated with survival [97, 98]. The ratio of Tregs and cancer recurrence decreases after DC vaccination combined with cytokine-induced killer (DC/CIK) immunotherapy in patients with NSCLC, indicating that this strategy is generally successful [99]. MoDCs are most widely used due to their easy acquisition from patients [100]. There are two main procedures used to activate DC-induced immunity in patients: one is to isolate DCs from patients and then feedback them after stimulation and modification, and the other is to target and activate DCs directly in vivo using specific molecules or vectors (Fig. 3).

Fig. 3
figure 3

Brief procedures for DC-based immunotherapy. For DC activation in vivo, various DC agonists were injected directly into patients. For DC vaccine construction in vitro, PBMCs were isolated from patients and polarized into DC in the presence of cytokines. After modified with antigen or DC activation factors, ex vivo-generated DCs were feedback to patients to get therapeutic effects. DC: dendritic cells; PBMC: peripheral blood mononuclear cells

Tumor-specific immune activation is the biggest advantage of immunotherapy compared to conventional strategies. In attempts to generate a successful DC vaccine, various antigens have been used to activate DCs, such as tumor cell lysates [101], exosomes [102], and tumor-associated antigens [103]. Significantly high expression of Survivin [104] and mucin1 (MUC1) [105] in lung cancers makes them suitable for pulsing DCs. In a phase I clinical trial involving 15 patients with NSCLC, Survivin, and MUC1 were used together to pulse DCs in vitro. Additionally, suppressor of cytokine signal 1 (SOCS1) [106] was also used in this system due to its essential role in negative regulation of DC differentiation and antigen presentation. After DC vaccine treatment, the expression of tumor markers was significantly reduced and the living quality of all patients was improved [107]. In another clinical trial, DCs were transduced with an adenoviral vector expressing C–C motif chemokine ligand 21 (CCL21), which attracts DCs and T cells by interacting with C–C motif chemokine receptor 7 (CCR7) and C-X-C motif chemokine receptor 3 (CXCR3) receptors, recruits antigen-stimulated DCs into T-cell zones in secondary lymphoid organs, and plays a key role in T-cell activation. This was performed before the intratumoral administration in patients with advanced NSCLC (n = 17). In response to this therapy, the treated tumors revealed enhanced CD8+ lymphocyte infiltration. Moreover, 6 of the 16 patients demonstrated tumor antigen-specific IFN-γ secretion and 3 patients revealed non-specific secretion of IFN-γ. Humoral responses to tumor-associated antigens were also detected in 4 of 8 patients. Moreover, after the injection of the CCL21-DC vaccine, the increased tumor expression of PD-1 was also detected [108]. As described previously, only 20% of patients respond to PD-1/PD-L1 inhibitors, whereas patients without PD-1/PD-L1 expression hardly benefit from the treatment of PD-1/PD-L1 inhibitors. This study suggests that using a combined checkpoint blockade with a DC vaccine may result in better clinical outcomes for patients with NSCLC.

Direct activation of DCs in vivo stands as another strategy for tumor therapy. As early as 1998, FLT3 ligands were used to induce MHC type II, CD11c+ and CD205+ DCs in mouse lymphoid and nonlymphoid tissues to generate effective anti-tumor responses [109]. Further studies proved that the FLT3 ligands administration after radiation therapy prolongs the survival rate of metastatic lung cancer in a mouse model through the enhancement of tumor antigen presentation by DCs [110]. In a preclinical melanoma model, a short hairpin RNA-based adjuvant that was able to silence IκBα expression was also shown to significantly activate the NF-κB pathway, thus promoting DC migration to lymph nodes and activating CD8+ T cells [70].

Although progress has been made in the development of DC-based immunotherapies, their therapeutic effect is still limited due to the complications presented by lung cancers. As such, employing a combination of different immunotherapy methods is currently essential in the fight against lung cancer. The immune regulatory function of lung cancers has been demonstrated in many previous studies, and further understanding on the interaction between lung cancers and DCs will be critical to prevent tumor growth in the future.

Future prospects

The immunosuppressive ability of lung cancer limits the usefulness of immune control [111]. Numerous investigations have identified the different mechanisms responsible for DC anergy in lung cancers, including the downregulation of functional markers, the induction of immunosuppressive molecular expression, and the exclusion of functional cDCs from lung-tumor lesions (Fig. 4). However, current immunotherapy strategies mainly focus on enhancing the antigen-presenting activity of the DCs rather than blocking the immunosuppression induced by lung tumors [112].

Fig. 4
figure 4

Lung tumors induce cDC anergy through different pathways. Tumors can alter the molecular expression of DC and also upregulate the immunosuppression miRNAs in DCs to inhibit the secretion of IL-12. Exosomes secreted by lung tumor cells can induce PD-L1 expression in DCs. Furthermore, different factors (HMGB1, TSLP, lactic acid) are highly expressed in lung tumors, and these may interact with different pathways within DCs to achieve a tumor-supportive microenvironment. cDC: conventional DC; HMGB1: high mobility group box-1 protein; TSLP: thymic stromal lymphopoietin; TSLPR: thymic stromal lymphopoietin receptor; IFN-γ: interferon γ; STING: stimulator of IFN genes; TLR2: toll-like receptor 2; Treg: regulatory T cells; PD-L1: programmed cell death 1 ligand 1; IL-10R: interleukin 10 receptor; TIM3: T-cell immunoglobulin and mucin-domain 3

Recently, one lung cancer patient was involved in the first-in-human study evaluating a DC targeting lentiviral vector which deliver antigen-encoding gene could activate antigen-specific T cell responses effectively in vivo [113]. This reminds us that this vector could also be used to deliver DC-activation genes in the future. It will be more convenient to enhance the overall DC activity directly in vivo to overcome the immunosuppression induced by lung cancer.

In spite of the therapeutic excitement of DC vaccines against lung cancer, significant challenges are still remain on their wide adoption in clinical use. Chiappori et al. [114] carried out a randomized-controlled phase II trial of salvage chemotherapy after immunization with a tumor protein p53-transfected DC vaccine in recurrent small cell lung cancer patients and found no survival differences between groups. As previously described, the injection of a tumor-antigen-loaded DC vaccine failed to consistently migrate into the lymphocyte node and elicit T-cell responses, and this may contribute to the immunosuppressive environment observed in patients [115,116,117]. However, there have been few clinical trials on blocking the DC-inhibition pathways in lung cancer till now. The block of common immune checkpoint PD-1/PD-L1 on DC has been partly explored. A study carried out by Chen et al. [118] demonstrated that PD-1 blockade-activated DC–CIK cells exhibit superior anti-tumor potency in several advanced solid tumors including NSCLC. Moreover, Ge et al. [119] showed that a blockade of the PD-1/PD-L1 immune checkpoint during DC vaccination induced potent protective immunity against breast cancer in hu-SCID mice. Thus, the combination of DC activation and blockage of its inhibition may generate a superior protection response in patients. It will be helpful to understand how lung cancer cells alter DC physiology and how we can generate novel immunotherapy strategies based on the powerful properties of DC. Efforts to reverse the critical elements of the immunosuppressive milieu to enhance DC vaccine potency are urgently needed in the future.

Conclusion

Lung cancer has the highest incidence among all cancers worldwide, and DC-based immunotherapy has become an important strategy to fight this disease, mainly via the direct activation of cytotoxic T cell responses. The recent development of DC vaccine has furthered our understanding of the vital roles of DCs in the control of tumor progression. However, the complex regulation between lung tumor and DC requires rational manipulation of DCs to initiate protective immunity. To this end, a sufficient understanding of the interaction between the lung tumor and DCs will accelerate the development of new immunotherapy strategies against this fatal disease.

Availability of data and materials

Not applicable.

Abbreviations

NSCLC:

non-small cell lung cancer

PD-1:

programmed cell death 1

PD-L1:

programmed cell death 1 ligand 1

CTLA4:

cytotoxic T-lymphocyte-associated antigen-4

DC:

dendritic cell

TME:

tumor microenvironment

TIM:

tumor-infiltrating myeloid cell

cDC:

conventional dendritic cell

pDC:

plasmacytoid dendritic cell

MDP:

monocyte-dendritic cell progenitor

CMP:

common myeloid progenitor

FLT3:

FMS-like tyrosine kinase 3

cMoP:

common monocyte progenitor

MoDC:

monocyte-derived dendritic cell

TLR:

toll-like receptor

IFN:

interferon

NK:

natural killer

CCR:

C–C chemokine receptor

Treg:

regulatory T cell

LPS:

lipopolysaccharides

IL:

interleukin

AIM2:

melanoma 2

regDC:

regulatory dendritic cell

miRNA:

microRNA

ID3:

inhibitor of DNA binding 3

DAMP:

damage-associated molecular pattern

TSLP:

thymic stromal lymphopoietin

SOCS1:

suppressor of cytokine signal 1

References

  1. Zhang Z, et al. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy. Front Med. 2019;13:57–68.

    Article  PubMed  Google Scholar 

  2. Malhotra J, Jabbour SK, Aisner J. Current state of immunotherapy for non-small cell lung cancer. Transl Lung Cancer Res. 2017;6(2):196–211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pandey D, et al. Mediastinal staging for non-small cell lung cancer revisited. It is being done under aegis of ICON and Lung cancer consortium Asia. Indian J Cancer. 2017;54(1):68–72.

    Article  CAS  PubMed  Google Scholar 

  4. Nishie K, et al. Anamorelin for advanced non-small-cell lung cancer with cachexia: systematic review and meta-analysis. Lung Cancer. 2017;112:25–34.

    Article  PubMed  Google Scholar 

  5. Wu HH, et al. Cytoplasmic Ape1 expression elevated by p53 aberration may predict survival and relapse in resected non-small cell lung cancer. Ann Surg Oncol. 2013;20(Suppl 3):S336–47.

    Article  PubMed  Google Scholar 

  6. Goldberg SB, et al. Early assessment of lung cancer immunotherapy response via circulating tumor DNA. Clin Cancer Res. 2018;24:1872–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hradilova N, et al. Generation of dendritic cell-based vaccine using high hydrostatic pressure for non-small cell lung cancer immunotherapy. PLoS ONE. 2017;12(2):e0171539.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Hubbeling HG, et al. Safety of combined PD-1 pathway inhibition and intracranial radiation therapy in non-small cell lung cancer. J Thorac Oncol. 2018;13:550–8.

    Article  PubMed  Google Scholar 

  9. Zeltsman M, et al. CAR T-cell therapy for lung cancer and malignant pleural mesothelioma. Transl Res. 2017;187:1–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hersom M, Jorgensen JT. Companion and complementary diagnostics-focus on PD-L1 expression assays for PD-1/PD-L1 checkpoint inhibitors in non-small cell lung cancer. Ther Drug Monit. 2018;40(1):9–16.

    CAS  PubMed  Google Scholar 

  11. Hunter KA, Socinski MA, Villaruz LC. PD-L1 testing in guiding patient selection for PD-1/PD-L1 inhibitor therapy in lung cancer. Mol Diagn Ther. 2018;22(1):1–10.

    Article  PubMed Central  CAS  Google Scholar 

  12. Chae YK, et al. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J Immunother Cancer. 2018;6(1):39.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Gonzalez-Cao M, et al. Targeting PD-1/PD-L1 in lung cancer: current perspectives. Lung Cancer (Auckl). 2015;6:55–70.

    CAS  Google Scholar 

  14. Larsen TV, Hussmann D, Nielsen AL. PD-L1 and PD-L2 expression correlated genes in non-small-cell lung cancer. Cancer Commun (Lond). 2019;39(1):30.

    Article  Google Scholar 

  15. Lim TS, et al. PD-1 expression on dendritic cells suppresses CD8(+) T cell function and antitumor immunity. Oncoimmunology. 2016;5(3):e1085146.

    Article  PubMed  CAS  Google Scholar 

  16. Li W, et al. Exosomes derived from Rab27aoverexpressing tumor cells elicit efficient induction of antitumor immunity. Mol Med Rep. 2013;8(6):1876–82.

    Article  CAS  PubMed  Google Scholar 

  17. Fatima F, Nawaz M. Stem cell-derived exosomes: roles in stromal remodeling, tumor progression, and cancer immunotherapy. Chin J Cancer. 2015;34(12):541–53.

    CAS  PubMed  Google Scholar 

  18. Besse B, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5(4):e1071008.

    Article  PubMed  CAS  Google Scholar 

  19. Upham JW, Xi Y. Dendritic cells in human lung disease: recent advances. Chest. 2017;151(3):668–73.

    Article  PubMed  Google Scholar 

  20. Son KJ, et al. Immunogenic cell death induced by ginsenoside Rg3: significance in dendritic cell-based anti-tumor immunotherapy. Immune Netw. 2016;16(1):75–84.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Chae CS, Teran-Cabanillas E, Cubillos-Ruiz JR. Dendritic cell rehab: new strategies to unleash therapeutic immunity in ovarian cancer. Cancer Immunol Immunother. 2017;66(8):969–77.

    Article  CAS  PubMed  Google Scholar 

  22. Shang N, et al. Dendritic cells based immunotherapy. Am J Cancer Res. 2017;7(10):2091–102.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Schneider Thomas, et al. Non-small cell lung cancer induces an immunosuppressive phenotype of dendritic cells in tumor microenvironment by upregulating B7-H3. J Thorac Oncol. 2011;6:1162–8.

    Article  PubMed  Google Scholar 

  24. Li J, et al. Myeloid-derived suppressor cells accumulate among myeloid cells contributing to tumor growth in matrix metalloproteinase 12 knockout mice. Cell Immunol. 2018;327:1–12.

    Article  PubMed  CAS  Google Scholar 

  25. Sass SN, et al. Tumor-associated myeloid cells promote tumorigenesis of non-tumorigenic human and murine prostatic epithelial cell lines. Cancer Immunol Immunother. 2018;67(6):873–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ohnuki H, et al. Tumor-infiltrating myeloid cells activate Dll4/Notch/TGF-beta signaling to drive malignant progression. Cancer Res. 2014;74(7):2038–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ruez R, et al. Dendritic cell migration toward CCL21 gradient requires functional Cx43. Front Physiol. 2018;9:288.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Gwin K, et al. Differential requirement for Hoxa9 in the development and differentiation of B, NK, and DC-lineage cells from Flt3+ multipotential progenitors. BMC Immunol. 2013;14:5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu K, Nussenzweig MC. Origin and development of dendritic cells. Immunol Rev. 2010;234(1):45–54.

    Article  CAS  PubMed  Google Scholar 

  30. Giordano D, et al. Nitric oxide controls an inflammatory-like Ly6C(hi)PDCA1+ DC subset that regulates Th1 immune responses. J Leukoc Biol. 2011;89(3):443–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sharma MD, et al. Activation of p53 in immature myeloid precursor cells controls differentiation into Ly6c(+)CD103(+) monocytic antigen-presenting cells in tumors. Immunity. 2018;48(1):91–106 e6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Narita M, et al. Induction of antigen-specific cytotoxic T lymphocytes by using monocyte-derived DCs transfected with in vitro-transcribed WT1 or SART1 mRNA. Med Oncol. 2009;26(4):429–36.

    Article  CAS  PubMed  Google Scholar 

  33. Hammad H, Lambrecht BN. Lung dendritic cell migration. Adv Immunol. 2007;93:265–78.

    Article  CAS  PubMed  Google Scholar 

  34. Bedoui S, Heath WR. Kruppel-ling of IRF4-dependent DCs into two functionally distinct DC subsets. Immunity. 2015;42(5):785–7.

    Article  CAS  PubMed  Google Scholar 

  35. Grajales-Reyes GE, et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8alpha(+) conventional DC clonogenic progenitor. Nat Immunol. 2015;16(7):708–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yan J, et al. FGL2 promotes tumor progression in the CNS by suppressing CD103(+) dendritic cell differentiation. Nat Commun. 2019;10(1):448.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Kenna T, et al. CD1 expression and CD1-restricted T cell activity in normal and tumour-bearing human liver. Cancer Immunol Immunother. 2007;56(4):563–72.

    Article  CAS  PubMed  Google Scholar 

  38. Lavin Y, et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell. 2017;169(4):750–65 e17.

    Article  CAS  Google Scholar 

  39. Masten BJ, et al. Characterization of myeloid and plasmacytoid dendritic cells in human lung. J Immunol. 2006;177(11):7784–93.

    Article  CAS  PubMed  Google Scholar 

  40. Faith A, et al. Plasmacytoid dendritic cells from human lung cancer draining lymph nodes induce Tc1 responses. Am J Respir Cell Mol Biol. 2007;36(3):360–7.

    Article  CAS  PubMed  Google Scholar 

  41. Jongbloed SL, et al. Plasmacytoid dendritic cells regulate breach of self-tolerance in autoimmune arthritis. J Immunol. 2009;182(2):963–8.

    Article  CAS  PubMed  Google Scholar 

  42. Alegre ML. DC metabolism controls lung T cell polarization. Am J Transplant. 2018;18(2):285.

    Article  PubMed  Google Scholar 

  43. Macri C, et al. Dendritic cell subsets. Semin Cell Dev Biol. 2017;84:11–21.

    Article  PubMed  CAS  Google Scholar 

  44. Liu Y, Cao X. Intratumoral dendritic cells in the anti-tumor immune response. Cell Mol Immunol. 2015;12(4):387–90.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Roberts EW, et al. Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR45 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell. 2016;30(2):324–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Salmon H, et al. Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pyfferoen L, et al. The transcriptome of lung tumor-infiltrating dendritic cells reveals a tumor-supporting phenotype and a microRNA signature with negative impact on clinical outcome. Oncoimmunology. 2017;6(1):e1253655.

    Article  PubMed  CAS  Google Scholar 

  48. Dutertre CA, Wang LF, Ginhoux F. Aligning bona fide dendritic cell populations across species. Cell Immunol. 2014;291(1–2):3–10.

    Article  CAS  PubMed  Google Scholar 

  49. Alculumbre S, et al. Plasmacytoid pre-dendritic cells (pDC): from molecular pathways to function and disease association. Semin Cell Dev Biol. 2018;86:24–35.

    Article  PubMed  CAS  Google Scholar 

  50. Manz MG. Plasmacytoid dendritic cells: origin matters. Nat Immunol. 2018;19:652.

    Article  CAS  PubMed  Google Scholar 

  51. Pinto A, Morello S, Sorrentino R. Lung cancer and toll-like receptors. Cancer Immunol Immunother. 2011;60(9):1211–20.

    Article  CAS  PubMed  Google Scholar 

  52. Conrad C, 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 

  53. Pedroza-Gonzalez A, 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  CAS  Google Scholar 

  54. Smirnov A, et al. Sphingosine 1-phosphate- and C–C chemokine receptor 2-dependent activation of CD4(+) plasmacytoid dendritic cells in the bone marrow contributes to signs of sepsis-induced immunosuppression. Front Immunol. 2017;8:1622.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Liu C, 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 

  56. Sorrentino R, et al. Plasmacytoid dendritic cells alter the antitumor activity of CpG-oligodeoxynucleotides in a mouse model of lung carcinoma. J Immunol. 2010;185(8):4641–50.

    Article  CAS  PubMed  Google Scholar 

  57. Narusawa M, et al. TLR7 ligand augments GM-CSF-initiated antitumor immunity through activation of plasmacytoid dendritic cells. Cancer Immunol Res. 2014;2(6):568–80.

    Article  CAS  PubMed  Google Scholar 

  58. Rega A, et al. Plasmacytoid dendritic cells play a key role in tumor progression in lipopolysaccharide-stimulated lung tumor-bearing mice. J Immunol. 2013;190(5):2391–402.

    Article  CAS  PubMed  Google Scholar 

  59. Shi W, et al. 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 

  60. Sorrentino R, et al. 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 

  61. Zhong H, et al. Origin and pharmacological modulation of tumor-associated regulatory dendritic cells. Int J Cancer. 2014;134(11):2633–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Spallanzani RG, et al. Regulatory dendritic cells restrain NK cell IFN-gamma production through mechanisms involving NKp46, IL-10, and MHC class I-specific inhibitory receptors. J Immunol. 2015;195(5):2141–8.

    Article  CAS  PubMed  Google Scholar 

  63. Zhang C, et al. The effect of semimature dendritic cell and the levels of Treg on transplantation tolerance of hepatocytes differentiated from mouse embryonic stem cell. Zhonghua Yi Xue Za Zhi. 2017;97(4):259–64.

    CAS  PubMed  Google Scholar 

  64. Spary LK, et al. Tumor stroma-derived factors skew monocyte to dendritic cell differentiation toward a suppressive CD14(+) PD-L1(+) phenotype in prostate cancer. Oncoimmunology. 2014;3(9):e955331.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Pardee AD, Shi J, Butterfield LH. Tumor-derived alpha-fetoprotein impairs the differentiation and T cell stimulatory activity of human dendritic cells. J Immunol. 2014;193(11):5723–32.

    Article  CAS  PubMed  Google Scholar 

  66. Zong J, et al. Tumor-derived factors modulating dendritic cell function. Cancer Immunol Immunother. 2016;65(7):821–33.

    Article  CAS  PubMed  Google Scholar 

  67. Goc J, et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 2014;74(3):705–15.

    Article  CAS  PubMed  Google Scholar 

  68. Caronni N, et al. Downregulation of membrane trafficking proteins and lactate conditioning determine loss of dendritic cell function in lung cancer. Cancer Res. 2018;78:1685–99.

    Article  CAS  PubMed  Google Scholar 

  69. Li R, et al. STAT3 and NF-kappaB are simultaneously suppressed in dendritic cells in lung cancer. Sci Rep. 2017;7:45395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Galvez-Cancino F, et al. A short hairpin RNA-based adjuvant targeting NF-kappaB repressor IkappaBalpha promotes migration of dermal dendritic cells to draining lymph nodes and antitumor CTL responses induced by DNA vaccination. Vaccine. 2017;35(33):4148–54.

    Article  CAS  PubMed  Google Scholar 

  71. Xue W, et al. Response and resistance to NF-kappaB inhibitors in mouse models of lung adenocarcinoma. Cancer Discov. 2011;1(3):236–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. McLoed AG, et al. Neutrophil-derived IL-1beta impairs the efficacy of NF-kappaB inhibitors against lung cancer. Cell Rep. 2016;16(1):120–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Schneider T, et al. Immune response after radiofrequency ablation and surgical resection in nonsmall cell lung cancer. Semin Thorac Cardiovasc Surg. 2016;28(2):585–92.

    Article  PubMed  Google Scholar 

  74. Luo D, et al. B7-H3 regulates lipid metabolism of lung cancer through SREBP1-mediated expression of FASN. Biochem Biophys Res Commun. 2017;482(4):1246–51.

    Article  CAS  PubMed  Google Scholar 

  75. He CS, et al. Astragaloside IV enhances cisplatin chemosensitivity in non-small cell lung cancer cells through inhibition of B7-H3. Cell Physiol Biochem. 2016;40(5):1221–9.

    Article  CAS  PubMed  Google Scholar 

  76. Zhang G, et al. Diagnosis value of serum B7-H3 expression in non-small cell lung cancer. Lung Cancer. 2009;66(2):245–9.

    Article  PubMed  Google Scholar 

  77. Fatima F, Nawaz M. Vesiculated long non-coding RNAs: offshore packages deciphering trans-regulation between cells, cancer progression and resistance to therapies. Noncoding RNA. 2017;3(1):10.

    PubMed Central  Google Scholar 

  78. Pyfferoen L, et al. Lung tumours reprogram pulmonary dendritic cell immunogenicity at the microRNA level. Int J Cancer. 2014;135(12):2868–77.

    Article  CAS  PubMed  Google Scholar 

  79. Luo LH, et al. Tim3/galectin-9 alleviates the inflammation of TAO patients via suppressing Akt/NF-kB signaling pathway. Biochem Biophys Res Commun. 2017;491(4):966–72.

    Article  CAS  PubMed  Google Scholar 

  80. Chiba S, et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat Immunol. 2012;13(9):832–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Nobumoto A, et al. Galectin-9 expands unique macrophages exhibiting plasmacytoid dendritic cell-like phenotypes that activate NK cells in tumor-bearing mice. Clin Immunol. 2009;130(3):322–30.

    Article  CAS  PubMed  Google Scholar 

  82. Cheng DE, et al. Involvement of IL10 and granulocyte colonystimulating factor in the fate of monocytes controlled by galectin1. Mol Med Rep. 2014;10(5):2389–94.

    Article  CAS  PubMed  Google Scholar 

  83. Kuo PL, et al. Lung cancer-derived galectin-1 mediates dendritic cell anergy through inhibitor of DNA binding 3/IL-10 signaling pathway. J Immunol. 2011;186(3):1521–30.

    Article  CAS  PubMed  Google Scholar 

  84. Chow MT, Moller A, Smyth MJ. Inflammation and immune surveillance in cancer. Semin Cancer Biol. 2012;22(1):23–32.

    Article  CAS  PubMed  Google Scholar 

  85. Michaud HA, et al. Tumor antigen-targeting monoclonal antibody-based immunotherapy: orchestrating combined strategies for the development of long-term antitumor immunity. Oncoimmunology. 2014;3(9):e955684.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Liu QY, et al. High mobility group box 1 protein suppresses T cell-mediated immunity via CD11c(low)CD45RB(high) dendritic cell differentiation. Cytokine. 2011;54(2):205–11.

    Article  CAS  PubMed  Google Scholar 

  87. Niki M, et al. New prognostic biomarkers and therapeutic effect of bevacizumab for patients with non-small-cell lung cancer. Lung Cancer (Auckl). 2017;8:91–9.

    CAS  Google Scholar 

  88. Ma Y, et al. Up-regulated HMGB1 in the pleural effusion of non-small cell lung cancer (NSCLC) patients reduces the chemosensitivity of NSCLC cells. Tumori. 2017;104:338–43.

    Article  Google Scholar 

  89. Aguilar-Cazares D, et al. Relationship of dendritic cell density, HMGB1 expression, and tumor-infiltrating lymphocytes in non-small cell lung carcinomas. Appl Immunohistochem Mol Morphol. 2014;22(2):105–13.

    Article  CAS  PubMed  Google Scholar 

  90. Smolarczyk R, et al. The role of Glycyrrhizin, an inhibitor of HMGB1 protein, in anticancer therapy. Arch Immunol Ther Exp (Warsz). 2012;60(5):391–9.

    Article  CAS  Google Scholar 

  91. Zhang Y, et al. Tumor-derived high-mobility group box 1 and thymic stromal lymphopoietin are involved in modulating dendritic cells to activate T regulatory cells in a mouse model. Cancer Immunol Immunother. 2017;67:353–66.

    Article  PubMed  CAS  Google Scholar 

  92. Ning Y, et al. Tumor exosomes block dendritic cells maturation to decrease the T cell immune response. Immunol Lett. 2018;199:36–43.

    Article  CAS  PubMed  Google Scholar 

  93. Nestle FO, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4(3):328–32.

    Article  CAS  PubMed  Google Scholar 

  94. Cornel AM, et al. Strategies to genetically modulate dendritic cells to potentiate anti-tumor responses in hematologic malignancies. Front Immunol. 2018;9:982.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Grees M, et al. Optimized dendritic cell vaccination induces potent CD8 T cell responses and anti-tumor effects in transgenic mouse melanoma models. Oncoimmunology. 2018;7(7):e1445457.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Dobrovolskiene N, et al. Tumor lysate-loaded Bacterial Ghosts as a tool for optimized production of therapeutic dendritic cell-based cancer vaccines. Vaccine. 2018;36(29):4171–80.

    Article  CAS  PubMed  Google Scholar 

  97. Arai R, et al. Lipid accumulation in peripheral blood dendritic cells and anticancer immunity in patients with lung cancer. J Immunol Res. 2018;2018:5708239.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Obleukhova I, et al. Use of antigen-primed dendritic cells for inducing antitumor immune responses in vitro in patients with non-small cell lung cancer. Oncol Lett. 2018;15(1):1297–306.

    PubMed  Google Scholar 

  99. Song H, et al. Increased cycles of DC/CIK immunotherapy decreases frequency of Tregs in patients with resected NSCLC. Int Immunopharmacol. 2017;52:197–202.

    Article  CAS  PubMed  Google Scholar 

  100. Geskin LJ, et al. Three antigen-loading methods in dendritic cell vaccines for metastatic melanoma. Melanoma Res. 2018;28(3):211–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Mookerjee A, Graciotti M, Kandalaft LE. Correction: a cancer vaccine with dendritic cells differentiated with GM-CSF and IFNalpha and pulsed with a squaric acid treated cell lysate improves T cell priming and tumor growth control in a mouse model. Bioimpacts. 2019;9(1):65.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Xiao L, et al. Efficacy of vaccination with tumor-exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology. 2017;6(6):e1319044.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Jiang T, et al. Immunotherapy with dendritic cells modified with tumor-associated antigen gene demonstrates enhanced antitumor effect against lung cancer. Transl Oncol. 2017;10(2):132–41.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Baspinar Y, et al. Development and characterization of nanobubbles containing paclitaxel and survivin inhibitor YM155 against lung cancer. Int J Pharm. 2019;566:149–56.

    Article  CAS  PubMed  Google Scholar 

  105. Chen ZQ, Huang LS, Zhu B. Assessment of seven clinical tumor markers in diagnosis of non-small-cell lung cancer. Dis Markers. 2018;2018:9845123.

    PubMed  PubMed Central  Google Scholar 

  106. Aldrich M, et al. SOCS1 downregulation in dendritic cells promotes memory T-cell responses. Vaccine. 2008;26(8):1128–35.

    Article  CAS  PubMed  Google Scholar 

  107. Ge C, et al. Phase I clinical trial of a novel autologous modified-DC vaccine in patients with resected NSCLC. BMC Cancer. 2017;17(1):884.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Lee JM, et al. Phase I trial of intratumoral injection of CCL21 gene-modified dendritic cells in lung cancer elicits tumor-specific immune responses and CD8(+) T-cell infiltration. Clin Cancer Res. 2017;23(16):4556–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Lynch DH. Induction of dendritic cells (DC) by Flt3 Ligand (FL) promotes the generation of tumor-specific immune responses in vivo. Crit Rev Immunol. 1998;18(1–2):99–107.

    Article  CAS  PubMed  Google Scholar 

  110. Chakravarty PK, et al. Flt3-ligand administration after radiation therapy prolongs survival in a murine model of metastatic lung cancer. Cancer Res. 1999;59(24):6028–32.

    CAS  PubMed  Google Scholar 

  111. Saito H, et al. Combination of p53-DC vaccine and rAd-p53 gene therapy induced CTLs cytotoxic against p53-deleted human prostate cancer cells in vitro. Cancer Gene Ther. 2017;24(7):289–96.

    Article  CAS  PubMed  Google Scholar 

  112. Li AW, et al. A facile approach to enhance antigen response for personalized cancer vaccination. Nat Mater. 2018;17:528.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Somaiah N, et al. First-in-class, first-in-human study evaluating LV305, a dendritic-cell tropic lentiviral vector, in sarcoma and other solid tumors expressing NY-ESO-1. Clin Cancer Res. 2019. https://doi.org/10.1158/1078-0432.CCR-19-1025.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Chiappori AA, et al. Randomized-controlled phase II trial of salvage chemotherapy after immunization with a TP53-transfected dendritic cell-based vaccine (Ad.p53-DC) in patients with recurrent small cell lung cancer. Cancer Immunol Immunother. 2019;68(3):517–27.

    Article  CAS  PubMed  Google Scholar 

  115. Nadafi R, et al. Dendritic cell migration to skin-draining lymph nodes is controlled by dermatan sulfate and determines adaptive immunity magnitude. Front Immunol. 2018;9:206.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Imai K, et al. Inhibition of dendritic cell migration by transforming growth factor-beta1 increases tumor-draining lymph node metastasis. J Exp Clin Cancer Res. 2012;31:3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Wang H, et al. Potential involvement of monocyte chemoattractant protein (MCP)-1/CCL2 in IL-4-mediated tumor immunity through inducing dendritic cell migration into the draining lymph nodes. Int Immunopharmacol. 2003;3(5):627–42.

    Article  CAS  PubMed  Google Scholar 

  118. Chen CL, et al. Safety and activity of PD-1 blockade-activated DC-CIK cells in patients with advanced solid tumors. Oncoimmunology. 2018;7(4):e1417721.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Ge Y, et al. Blockade of PD-1/PD-L1 immune checkpoint during DC vaccination induces potent protective immunity against breast cancer in hu-SCID mice. Cancer Lett. 2013;336(2):253–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81502589), the Natural Science Foundation of Guangdong (No. 408140352062 and 2014A030310481), the Science and Technology Project of Shenzhen (CXZZ20130515092016300 and JCYJ20160422142707177), and the China Postdoctoral Science Foundation (No. 2018M631046).

Author information

Authors and Affiliations

Authors

Contributions

JBW and XH wrote the manuscript, JBW prepared the figures and FRL edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Fu-Rong Li.

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.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, JB., Huang, X. & Li, FR. Impaired dendritic cell functions in lung cancer: a review of recent advances and future perspectives. Cancer Commun 39, 43 (2019). https://doi.org/10.1186/s40880-019-0387-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40880-019-0387-3

Keywords