Next Article in Journal
Formation of Unique Placental Seed Capsules in the Maturation Process of the Tomato Fruit
Next Article in Special Issue
Increased NMUR1 Expression in Mast Cells in the Synovial Membrane of Obese Osteoarthritis Patients
Previous Article in Journal
Cellular and Molecular Targets in Acute Ischemic Stroke
Previous Article in Special Issue
The Heat Shock Protein 90 (HSP90) Is Required for the IL-33-Induced Cytokine Production in Mast Cells (MCs)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mast Cells and Dendritic Cells as Cellular Immune Checkpoints in Immunotherapy of Solid Tumors

1
Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, V Uvalu 84, 150 06 Prague, Czech Republic
2
Laboratory of Immunotherapy, Institute of Microbiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(19), 11080; https://doi.org/10.3390/ijms231911080
Submission received: 22 August 2022 / Revised: 12 September 2022 / Accepted: 15 September 2022 / Published: 21 September 2022
(This article belongs to the Special Issue Mast Cells in Human Health and Diseases)

Abstract

:
The immune checkpoint inhibitors have revolutionized cancer immunotherapy. These inhibitors are game changers in many cancers and for many patients, sometimes show unprecedented therapeutic efficacy. However, their therapeutic efficacy is largely limited in many solid tumors where the tumor-controlled immune microenvironment prevents the immune system from efficiently reaching, recognizing, and eliminating cancer cells. The tumor immune microenvironment is largely orchestrated by immune cells through which tumors gain resistance against the immune system. Among these cells are mast cells and dendritic cells. Both cell types possess enormous capabilities to shape the immune microenvironment. These capabilities stage these cells as cellular checkpoints in the immune microenvironment. Regaining control over these cells in the tumor microenvironment can open new avenues for breaking the resistance of solid tumors to immunotherapy. In this review, we will discuss mast cells and dendritic cells in the context of solid tumors and how these immune cells can, alone or in cooperation, modulate the solid tumor resistance to the immune system. We will also discuss how this modulation could be used in novel immunotherapeutic modalities to weaken the solid tumor resistance to the immune system. This weakening could then help other immunotherapeutic modalities engage against these tumors more efficiently.

1. Introduction

Cancer is the second most common cause of death globally, and the number of people with the disease increases each year [1,2]. The therapy of cancer currently stands on four pillars––surgery, chemotherapy, radiotherapy, and immunotherapy. The first three pillars (surgery, chemotherapy, radiotherapy) target cancer cells directly. Immunotherapy, on the other hand, is a type of therapy that uses substances to modulate the immune system to help the body fight cancer [3]. For decades, cancer immunotherapy stood aside, not matching the success rate of the traditional treatment modalities (surgery, chemotherapy, or radiotherapy). However, this has changed with immunotherapy drugs called “immune checkpoint inhibitors” [4]. The efficacy of these inhibitors and their successors has already shown unprecedented results for some types of cancer, notably, where the traditional treatment options had already failed [5]. Currently, these inhibitors are increasingly being tested in a large number of clinical trials. For instance, the inhibitors against PD-1 and PD-L1 molecules are tested in thousands of clinical trials in combination with other treatment modalities [6,7,8]. Results of the studies are now allowing these inhibitors to be already registered and used even in first-line cancer therapy [9,10,11], something hardly imaginable a decade ago. Apart from the checkpoint inhibitors, another group of immunotherapeutic modalities showed promising results. These modalities are based on in-vitro-modified/expanded immune cells, which can induce cancer-targeted immune responses [12] or directly target and eliminate cancer cells [13]. Many of these drugs have already been FDA-approved––autologous dendritic cells (DCs) [14] or genetically-modified T cells with chimeric antigen receptors (CAR-T) [15] or T cell receptors [16].
Despite the great success of immunotherapy over the past few years, immunotherapy suffers from cancer resistance. This resistance means that most patients still do not benefit from the therapy [17,18]. Although there are multiple mechanisms of cancer resistance to immunotherapy [19], there could be basically drawn three critical causes why current immunotherapy faces resistance: a limited specificity, an inability to reach and eliminate its targets, and the immunosuppressive microenvironment of solid tumors. The limited specificity is caused by aiming at targets that are not present only on the targeted cancer cells. For instance, the inhibitory checkpoint molecule PD-1 is expressed not only on the tumor-targeting immune cells (such as on cytotoxic CD8+ T cells) but also on the tumor-protecting immune cells as regulatory CD4+ T cells [20]. Inhibition of PD-1, therefore, simultaneously unleashes both pro- and anti-tumorigenic immune responses, the combination of which may have either a curative or detrimental impact on the state of the disease. This may even happen in the same cohort of the treated patients [20]. The inability of immunotherapy to reach and eliminate its targets is also given by the inability of immune cells to reach and eliminate cancer cells in solid tumors. There are many solid tumors that entirely block their immune cell infiltration and, as such, prevent the immune system from attacking them [21]. The immunosuppressive microenvironment then adds up to the immunotherapy resistance where immunotherapy can overcome the solid tumor barriers, break in, and reach its targets. Herein, the immunosuppressive tumor microenvironment steps in by preventing immunotherapy from further acting against cancer cells efficiently.
To eliminate the resistance requires making solid tumors accessible to the immune cells, overcoming its immunosuppressive microenvironment to allow efficient cancer cell elimination and establishing lasting immune protection from tumor recurrence. These requirements are extremely challenging. However, there are two types of immune cells whose combined harnessing in the immunotherapeutic algorithms could stand up to these challenges. These cell types are mast cells (MCs) and dendritic cells (DCs).

2. MCs in Solid Tumors

MCs are tissue-resident highly granulated cells notoriously associated with the pathogenesis of allergic and autoimmune diseases [22,23]. These cells are equipped with a large number of receptors that enable them to respond to a large number of stimuli [24]. These responses include release (exocytosis) of preformed secretory granules (degranulation), migration, or de novo production of biologically and immunologically active products [24]. Their most notoriously known receptor is the high-affinity receptor for IgE, FcεRI. This receptor induces a fast degranulation of MCs, often referred to as anaphylactic degranulation since the content of the released granules are preformed mediators such as histamine, proteases, and heparin [25], which are responsible for anaphylaxis [26,27]. Another receptor that defines MCs is the stem cell factor (SCF) receptor, KIT, which is essential for MC development, survival, and migration [24]. Stimulation through this receptor can also increase or decrease the extent of FcεRI-induced MC degranulation [28,29] or, under some circumstances, even trigger this degranulation [30]. One of the MC receptors that has brought attention in the past several years is the Mas-related G protein-coupled receptor X2 (MRGPRX2) [31]. This receptor can recognize various chemical compounds, and its activation can cause pseudo-allergic reactions [31]. MCs are known to also interact with other immune cells and regulate their functions. Activated MCs can enhance T cell proliferation [32], co-stimulate T cells via a CD28-independent interaction [33], limit viral infection through interactions with γδ T cells [34], attract and affect NK cells [35], and recruit and alter functions of tumor-infiltrating macrophages, myeloid-derived suppressor cells, neutrophils, and dendritic cells [36].
Due to a broad spectrum of MC biological and immunological activities [37], these cells are also considered to be key orchestrators of the tumor immune microenvironment [38]. For that reason, these cells are increasingly scrutinized as potential targets for cancer immunotherapy [39]. However, MCs are highly complex immune cells whose function can render these cells either pro- or anti-tumorigenic. The mechanisms of the pro- or anti-tumorigenic activities of MCs are diverse and involve both the immune and non-immune compartments of the human body [39]. A few examples of these mechanisms shown in Table 1 indicate that effectors of these mechanisms are often soluble cytokines, chemokines, or growth factors. Indeed, the spectrum of these molecules is exceptionally vast and involves all critical molecules used in the regulation of innate and adaptive immunity [40]. However, in addition to the breadth of this spectrum, even one type of the MC-produced effector molecule, such as histamine or the serine protease tryptase (Table 1), was found to show either anti- or pro-tumorigenic activities in dependence on the leading impact they have in individual cancers [41,42,43,44]. The leading anti-tumorigenic impact of individual effector molecules could be the inhibition of tumor growth/tumor cell proliferation [43,45], inhibition of tumor engraftment [46], promotion of DC development and maturation [41], recruitment of CD8+ T cells and their subsequent antigen-mediated activation [47], tumor cell cytotoxicity [48], or direct tumor cell clearance [49]. The leading pro-tumorigenic impact, on the other hand, could be the enhancement of tumor cell proliferation [42], angiogenesis [44,50], immunosuppression and inflammation decrease [51,52,53], or promotion of the epithelial-to-mesenchymal transition (EMT) [54].
Such MC functional ambiguity towards tumors makes these cells controversial as to whether these cells should be therapeutically targeted for elimination or, rather, for enhancement of their numbers and activities. Whether MCs become pro- or anti-tumorigenic presumably lies in their functional plasticity, which is already programmed during their development and maturation from stem cell progenitors. Pioneering in vitro studies showed that a sustained presence of different cytokines during MC development programs their activation phenotype [28,55,56]. In dependence on the cytokine presence in the cell culture, this programming can be either reversible [55,56] or irreversible [28]. Since MC progenitors infiltrate tissues in which they then fully develop and mature, thus, acquiring the tissue-specific phenotypes, the tumor microenvironment is presumably also infiltrated with not yet fully developed/matured MCs, and their activation phenotype is then programmed as they develop and mature [57]. The specifics of the tumor microenvironment, such as hypoxia, low pH, and metabolite composition, can then imprint the developing and maturing MCs with a tumor microenvironment-specific activation phenotype, whose impact on the tumor could be hardly inferred from plain numbers of infiltrating MCs and their associations with the disease severity. In addition, mast cells in the tumor microenvironment can be phenotypically/functionally heterogeneous [58]. MCs heterogeneity was initially described based on their localization and protease content. The localization-based division recognizes connective tissue (CTMCs) and mucosal (MMCs) MCs. The protease-content-based division distinguishes MCs subtypes expressing tryptase (MCT) or both tryptase and chymase (MCTC) [37]. Based on their origin, MCs are also divided into a subgroup of constitutive or inducible MCs [59,60]. Nevertheless, regardless of the yet still enigmatic origin of MCs and their subtype characteristics, the resulting functional heterogeneity of developed and mature MCs seems to be substantially shaped by factors in the surrounding milieu of MC residence [24]. Then different MC functional phenotypes are presumably acquired in inflamed airways [61] and in the microenvironment of different tumors [62]. Indeed, single-cell RNA sequencing studies revealed significant diversity in MC development and MC residence-specific functional characteristics [63]. A recent study showed that MC phenotypic heterogeneity and their histological distribution with potentially distinct functions were observed even within an individual tumor microenvironment [58]. However, since there are currently no known specific markers of the tumor microenvironment-infiltrating MCs and their subtypes, and the MC role towards the tumor can be ambivalent, a design of MC-targeting strategies in cancer immunotherapy is, thus, highly challenging.

3. DCs in Solid Tumors

DCs are major professional antigen-presenting cells that present antigens on their surface via major histocompatibility complex I and II (MHC I and MHC II) to T cells to initiate adaptive antigen-dependent immune responses. DCs also mediate cross-talk between the innate and adaptive immune system through direct interactions with other immune cells––natural killer (NK) cells [64], NK T cells [65], neutrophils [66], and MCs [67]. DCs are derived from hematopoietic stem cell progenitors. The progenitors which infiltrate peripheral tissues differentiate into immature DCs [68]. These immature (tissue-resident) DCs have high phagocytic activity of extracellular material [69]. Without a maturation signal, such as stimulation with pathogen-associated molecular pattern (PAMPs) or damage-associated molecular pattern (DAMPs) molecules, the antigen presentation by immature DCs leads to immune tolerance to the presented antigens, including the antigens from the phagocyted material [70]. Such antigen presentation allows the establishment of peripheral immune tolerance to autoantigens [70]. However, once DCs mature, the phagocytic capacity of DC decreases, and DCs start to efficiently present antigens to induce either immune tolerance (tolerogenic DCs) or immune stimulation (immunogenic DCs) [70]. Which phenotype prevails is dependent on the signals triggering the maturation. These signals are elicited by soluble factors or interactions with surrounding cells [71]. Once mature, the phenotype is often irreversible [72].
Human DCs are divided into several subtypes based on their phenotype and ontogeny: plasmacytoid DC (pDC), classical DC (cDC) type 1 and 2 (cDC1 and cDC2), DC3, and monocyte-derived DC (mo-DC) [73]. The pDCs are present in lymphoid organs but not peripheral tissues. Activated pDCs specialize in the production of type I IFNs and are thought to be the main producers of these cytokines during infections and autoimmune diseases [74]. The cDCs are tissue-resident DCs. The cDCs type 1 are specialized in activating CD8+ and type 2 on CD4+ T cells, both of which play a role in the regulation of anti-tumor immunity [75,76]. The place of their residency then further shapes their phenotype to form multiple subsets of these cells in peripheral tissues [77]. The DC3 subtype is a novel subtype of DCs identified in the blood [78]. DC3s express a monocytic marker CD14, share common markers of cDC2, and have monocyte gene signatures [78,79]. Similar to DC3s are mo-DCs, which show overlapping phenotypes with DC3s [79]. Mo-DCs also share multiple phenotypic markers with monocyte-derived macrophages, thus, making them difficult to distinguish from each other [80]. Mo-DCs are able to in vivo cross-present antigens [81], and their ex vivo production from peripheral blood monocytes has become central to the development of DC-based cancer immunotherapy [82,83].
An important role in the DC anti-tumor activities in the tumor microenvironment is attributed to cDC1s, which play a crucial role in initiating and maintaining anti-tumor immunity by cross-presenting tumor antigens to CD8+ T cells, stimulating NK and NKT cells [84]. However, even though their role in anti-tumor immunity is necessary, it seems that it is insufficient and needs to be promoted by a cross-talk with other DC subtypes, namely the cDC2s [85]. The role of cDC2s in tumor immunity is ambivalent as these DCs can show either pro- or anti-tumorigenic activities [76]. Similar is true for mo-DCs and pDCs, both of which can adopt either pro- or anti-tumorigenic activities [86,87]. The mechanism of their pro- or anti-tumorigenic activities is driven by their ability to recruit and stimulate/inhibit cytotoxic or tolerogenic lymphocytes [88]. These abilities are allowed by the production of many chemokines, pro-/anti-inflammatory cytokines, and the expression of immune checkpoint molecules (details reviewed in [87,88]). However, regardless of the DC subtype, the functional phenotype of DCs is significantly shaped by factors in the milieu of their residence [89,90,91,92].
The tumor microenvironment also decides how DCs orchestrate pro- or anti-tumorigenic activities (Table 2). Anti-tumorigenic activities in the tumor microenvironment can be mediated by IL12-producing tumor-infiltrating DCs, which promote CD8+ T cell responses [93]; MHC-I cross-dressed DCs presenting tumor antigens and allowing cross-priming to CD8+ T cells [94]; CD1d high-expressor DCs, which increase activation of NKT, CD4+ and CD8+ T cells [95]; or by cCD2s, which can promote control of cytotoxic T cell-resistant tumors via the CD4+ T cell-mediated activation of myeloid cells [96]. Mature cDCs and pDCs can also inhibit angiogenesis through the production of IL-12, angiostatic chemokines, or IFNα [97]. On the other hand, pro-tumorigenic activities of DCs can be adopted by PD-1-expressing DCs, which accumulate in the tumor and inactivate CD8+ T cells [98]; arginase-expressing DCs, which causes arginine deprivation leading to the inhibition of CD4+ T cell proliferation [99] or reactive oxygen species (ROS)-mediated inhibition of CD8+ T cells [100]. Pro-tumorigenic activities are also elicited by indoleamine 2,3-dioxygenase(IDO)-expressing DCs, which cause both tryptophan-depletion-mediated inhibition of CD8+ T cells and tryptophan-metabolite-mediated expansion of CD4+ Treg cells [101]. Other DCs can produce TGF-β to induce immunosuppression [102], make IL10 alongside the surface expression of PD-L1 to impair CD8+ T cell activation [103], or shed soluble CD25 to deplete IL-2 and inhibit T cell proliferation [104]. Immature DCs can also promote angiogenesis [105]. Similar to MCs, whether the tumor-microenvironment-elicited DC phenotype drives these cells to pro- or anti-tumorigenic activities could also be hardly inferred from plain numbers of the infiltrating DCs and their associations with the disease severity. Even though recent technological advances allow for detailed characterization of DC subtypes infiltrating tumors [106,107], there are no known markers that would enable specific targeting of individual DC subsets in the tumor microenvironment to enhance or suppress, respectively, their pro- or anti-tumorigenic activities.

4. MC/DC Interplay

Both MCs and DCs are powerful standalone players in the tumor microenvironment. However, both cell types were also shown to impact each other. This impact can occur either via secreted factors or direct interactions. The interaction via secreted factors is less specific and driven mainly by MCs towards DCs. MC secreted factors can promote tolerogenic [52] or immunostimulatory [108] functions of DCs. On the other hand, direct interactions between these cell types are more specific and can impact both cell types. A study showed that migratory DCs in inflamed skin initially scan MCs and later come into long-lasting synaptic interactions. These interactions result in a DC-to-MC molecule transfer, including major histocompatibility complex class II (MHCII) molecules, which then enables MCs to prime T cells, therefore, promoting T-cell-driven inflammation in the skin [109]. Vice versa, activated MCs can form immunological synapses with immature DCs and transfer the MC-internalized antigen to immature DCs. The transferred antigen is then processed and presented by DCs to stimulate T cells [67]. Additionally, the tolerogenic or stimulatory outcome of the MC–DC interaction can be substantially affected by the expression of immune checkpoint molecules and their ligands on both cell types [53].
Regardless of the mechanisms that come into play upon MC–DC interactions, these mechanisms can substantially impact the balance between the tolerogenic and stimulatory activities of the immune system. Indeed, MCs activity toward DCs was shown to substantially suppress [52,110] or, vice versa, promote the immunogenic properties of DCs [111,112]. The suppression was elicited by MC-produced IL-13, which inhibited IL-12 expression in DCs with subsequent inhibition of DC-mediated induction of the TH1 response [52], or by promoting the DC tolerogenic phenotype through MC-produced large amounts of GM-CSF [110]. On the other hand, the promotion was elicited either by DC engulfment of MC-released granules, which was followed by DC migration to lymph nodes, their maturation, and boosting their T-cell priming efficiency [108], or by MC-mediated DC maturation followed by their recruitment to the site of infection [112].

5. MCs in Immunotherapy of Solid Tumors

The ambivalent role of MCs in tumors makes these cells challenging to judge how these cells should be targeted. Recent studies show that depletion of these cells or downregulation of their functions in the tumor microenvironment can help break tumor resistance [113,114,115]. Although these studies were made in mouse models, they used MC-targeting drugs that are already used in clinical practice––antihistamine and MC stabilizer ketotifen [116,117], MC stabilizer cromolyn [117], or c-KIT inhibitors [118]. Their use, or use of other clinically administered or tested MC-targeting drugs (anti-Siglec therapies [119] or other biologics [115]), could in combination with other cancer treatment modalities (i.e., chemotherapy or immunotherapy) then indeed represent a new promising treatment strategy in which MCs would be approached as inhibitory immune checkpoint cells in the tumor microenvironment [113,114]. This therapeutic approach would be presumably relevant in tumors where the tumor burden with MCs or their products are largely and convincingly associated with the disease severity and dismal prognosis [120,121], or where another therapeutic modality increases their numbers with pro-tumorigenic activities and causes a resistance to the therapy [122].
On the other hand, many studies showed that tumoral/peritumoral MCs are not only associated with a better prognosis [123,124] but that MCs can also directly eliminate cancer cells [49,125]. The mechanism of this elimination could be even unique, as demonstrated in a recent study where MCs were found to efficiently kill cancer cells with the help of cancer-cell-penetrating tunneling nanotube-like structures [48]. This killing was preceded by the recognition of cancer cells through the cancer cell antigen-specific IgE bound to the FcεRI expressed on the surface of MCs [48]. In addition, another recent study of the group showed that the IgE-sensitized MCs could easily infiltrate and eliminate masses of cancer cells both in vitro and in vivo [126,127]. This infiltration is presumably driven by MC chemotaxis towards the antigen of the FcεRI-bound IgE [128]. This antigen-driven chemotaxis and the unique killing mechanism may provide these cells with a novel advantage over other immune cells, which can also recognize and kill cancer cells [129]. Indeed, with the technological advances in ex vivo preparation of MCs from adipose tissue or CD34+ hematopoietic progenitors, a new field is already opening where MCs in combination with tumor antigen-specific IgEs are used for adoptive cell transfer (ACT)-based cancer cellular immunotherapy [130]. MCs in this field then could newly join the list of immune cells used or tested for this therapeutic modality.

6. DCs in Immunotherapy of Solid Tumors

In contrast to MCs, many cancer immunotherapeutic strategies based on DCs have already been tested in the last few decades. These strategies are mainly based on the unique ability of DCs to shape both adaptive and innate immune responses. As professional antigen-presenting cells, DCs are able to induce tumor-antigen-specific responses through the interaction with naive T cells [131]. Furthermore, mature DCs produce cytokines such as interleukin-12 or type I interferons that enable the regulation of many immune cell types that take part in anti-tumor immunity, including many innate immune cells [132,133]. For these biological features, DCs were believed to be very promising tools for ACT-based cancer cellular immunotherapy, also called––DC-based vaccines. DC-based vaccines are usually based on autologous DCs that are differentiated ex vivo from monocytes or hematopoietic stem cell progenitors, loaded with tumor antigens, and exposed to maturation stimuli. DCs prepared in this way are then transferred into the patient’s body, where they induce anti-tumor immune responses through the activation of tumor-specific lymphocytes [134].
The first DC-based cancer vaccine clinical trials date back to the 1990s of the twentieth century. These early clinical trials mainly focused on the treatment of melanoma and showed promising results and favorable safety profiles [135,136,137]. The next wave of trials focused on leukemia and other solid tumors [138,139,140]. Out of so many clinical trials, there came only one FDA-approved DC-like-based commercially available product, Sipuleucel T (Provenge), which was designated for the treatment of metastatic prostate cancer [141]. However, this product is still not widely used in clinical practice [142].
The disappointment from the early “DC era” is presumably due to the fact that many studies were not designed in combination with other immunotherapeutic interventions and patients were enrolled at late stages of the disease, often associated with advanced tumor microenvironment and the disease-elicited immune system defects [143,144,145]. However, DC-based immunotherapy still goes on through efforts to find new ways for their improvement [146]. Due to advances in genomics, many recent DC-based approaches focus on targeting neoantigens that have become considered powerful targets for cancer immunotherapy [147,148,149], including DC-based vaccines [150]. In addition, current designs of therapeutic algorithms also rely on combinations of DC-based cancer vaccines with other therapeutic modalities [151,152]. There is also a developing new field that attempts to identify new biomarkers that could predict the therapeutic efficacy of the vaccines based on the patient’s immunotype [153].

7. MC/DC Interplay as the Cellular Immune Checkpoint for Cancer Immunotherapy

The history of the use of MCs and DCs in cancer immunotherapy is, therefore, entirely different. Whereas MCs have become the center of interest in immunotherapy recently [130], DCs are not new players in the field. Three decades ago, DCs were thought to be highly promising immune cells for cancer therapy, namely, for ACT-based immunotherapy. Despite many disappointments in clinical trials, the use of these cells still showed a notable therapeutic efficacy, and their role in cancer immunotherapy is indeed still ongoing [146,154,155]. As research into immunotherapy accelerated after the immune checkpoint inhibitor had entered the field, the efficacy of DC-based immunotherapy was found to be largely restricted by the settings (immunotype) of the patient’s tumor immune microenvironment [153,156]. Moreover, there is growing evidence that the functional heterogeneity of DC populations is even more complex than previously thought [73], and, therefore, more attention needs to be paid to the way DCs are ex vivo produced for ACT [82]. This attention relates to the DC differentiation process, antigen loading strategy, and DC maturation [82,146]. In this regard, the interaction of MCs with DCs may come with an advantage in the ex vivo preparation of DCs. In our previous work, we found that a stimulated human LAD2 MC line could be a highly potent cellular adjuvant for the maturation of ex-vivo-produced human-monocyte-derived DCs for ACT [157]. This high adjuvant potency was conditioned by previous MC stimulation with thapsigargin, an inhibitor of sarco/endoplasmic reticulum Ca2+-ATPases (SERCA) [158,159], which induces a strong mobilization of intracellular calcium followed by fast and extensive degranulation of LAD2 MCs [160]. Such previously stimulated and rinsed LAD2 MCs were, unlike their non-stimulated or FcεRI-stimulated counterparts, able to mature monocyte-derived DCs, which were even refractory to the maturation with polyinosinic:polycytidylic acid (poly I:C), a toll-like receptor-3 agonist often used for the production of matured DCs for ACT-based cancer cellular immunotherapy [154,155]. Moreover, the study showed that the adjuvant effect was still notable even when the MC:DC ratio was 1:150 [157]. This maturation efficiency indicated that a very small number of MCs can promote DC maturation. Apart from maturation, the MC–DC interaction can provide a targeted loading of DCs with an extracellular antigen, which is then efficiently presented to T cells [67]. This mechanism could be presumed to come also into play upon an MC-based ACT, where MCs could be expected not to only specifically (in an antigen-dependent manner) destroy cancer cells in the tumor [126] but to also come in contact with the tumor-infiltrating DCs and restore their antitumor activity [67]. Restoration of efficient antitumor activity of otherwise defective tumor-infiltrating DCs was already demonstrated for many tumors using different approaches [98,161,162]. Whether an MC-based ACT could also participate in the DC restoration in the tumor microenvironment and, as such, promote lasting immune protection against the disease remains to be investigated.

8. Future Perspectives

Recent development in MC research and its translation to cancer immunotherapy opens new avenues in developing novel immunotherapeutic combination strategies for treating solid tumors. The opening of the avenues was enabled by novel approaches in ex vivo clinical-scale preparation of autologous MCs [130,163,164]. The opening was then secured by the findings that these cells can not only kill tumor cells in an antigen/IgE/FcεRI-dependent manner but also translocate in vivo into tumors where they can deliver a therapeutic impact [126]. This translocation into tumors, presumably based on antigen-driven chemotaxis of MCs, seems to be one of the MC attributes that could challenge solid tumor resistance to ACT-based cancer immunotherapy. In addition, it could be even enhanced after targeted radiotherapy or chemotherapy, which spikes the release of tumor antigens from dying cancer cells [165,166] (Figure 1).
Apart from the engagement of IgE-mediated mechanisms in the MC antitumor activities, MCs can also enter a new uncharted territory of advanced genetic engineering that has already been developed for years for ex vivo modifications of other immune cells [167]. Similar to the T cell chimeric receptors, CAR [168] and TCR [169], chimeric antigen receptors based on FcεRI and its downstream signaling could be one of the plausible strategies for advanced MC-based ACT (Figure 1).
As a standalone produced/engineered player for ACT, MCs would certainly suffer from the drawback apparent to other immune cells used for ACT––the single antigen-driven specificity, which could be evaded by the loss or downregulation of the expression of the targeted tumor antigen [170]. This drawback could be, however, overcome through the MC–DC interplay. This interplay can harness endogenous or even tumor-infiltrating DCs whose engagement and loading with antigens released from MC-killed cancer cells could stimulate T cells and, as such, broaden the tumor antigen repertoire through which the immune system recognizes and eliminates cancer cells [134,171,172]. This repertoire could also include neoantigens, whose targeting by T cells seems to be critical for immunotherapeutic efficacy [147]. This presumed synergistic MC–DC-interplay-based antitumor activity would be particularly important in so called “cold tumors” [21], which prevent the immune cells from the tumor infiltration and subsequent induction of broad antigen-driven antitumor immune responses. Since the initial work with the cytotoxic MCs was performed with the Her2+ breast cell line [126], which forms “cold tumors” [173], there is a likelihood that the presumed MC–DC-interplay-elicited antitumor activity could also be attained in the immunotherapeutically-difficult-to-challenge “cold tumors” [21,174] (Figure 1).
Many combination strategies in cancer therapy rely on DC ACT as enhancers of therapeutic efficacy [146]. Many of these strategies involve combinations of DC ACT with other immunotherapeutic approaches, including immune checkpoint inhibitors [174], NK cells [175,176], or CAR-T cells [177]. In cold tumors, the combination is presumed to work towards turning the “cold tumors” into “hot tumors,” thus, enhancing the tumor immune cell infiltration [178]. Combining the cold-tumor-penetrating cytotoxic MCs with DC ACT could, therefore, also synergize in promoting the cold-to-hot turning process (Figure 1). Apart from the “turning process”, both cell types could also enhance the anti-tumor activity of other immune cells, including endogenous T cells [32] or adoptively transferred CAR-T cells [177].
Using MCs in combination with DCs in cancer immunotherapy needs to be designed with respect to tumor types, ongoing therapy, and mast cell-driven co-morbidities. The tumors that are known to be more sinister when large numbers of MCs infiltrate them [179] or when immunotherapy enhances their numbers alongside the growing tumor resistance [122] are presumably situations where MCs activities need to be suppressed or the cells even eliminated. A similar approach needs to be also considered when patients suffer from diseases related to dysregulated MC numbers [180] or their activation [181] or when MCs are directly part of the malignant neoplastic disease [182].

9. Concluding Remarks

MCs are now becoming a new player in the field of cancer immunotherapy. The new advances in their in vitro preparation and the recent demonstration of MC’s potential to infiltrate solid tumors and kill cancer cells within the tumor [126] bring expectations in these cells for cancer immunotherapy [130]. Although the demonstrated performance of the cancer cell-killing MCs in the tumor needs to be further investigated, this review suggests that the therapeutic power of MCs in cancer treatment may not so much lie in their single-antigen-driven cytotoxicity, but in their potential to trigger much more robust and lasting anti-tumor immunity to those solid tumors that so far have resisted immunotherapeutic interventions. This review also suggests that the key partner necessary for unleashing the therapeutic potential of MCs are DCs and that combination of MC- and DC-based ACT could be the therapeutic algorithm for licensing MCs for efficient cancer immunotherapy in clinics.

Author Contributions

D.S. (Daniel Smrz) conceptualized and wrote the original draft. K.K. contributed to the conceptualization and writing of the article. S.S. and E.D. contributed to the article writing. K.K., S.S., E.D., P.T., D.S. (Dmitry Stakheev), L.V., J.B. and D.S. (Daniel Smrz) reviewed and edited the article. All authors have read and agreed to the published version of the manuscript.

Funding

Research in the authors’ laboratories was supported by the Ministry of Health, Czech Republic—project AZV NU22-03-00300, the Institutional grant RVO 61388971 of the Institute of Microbiology AS CR, and by the Institutional IPE2 funding of the Charles University, Second Faculty of Medicine in Prague.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

J.B. is a part-time employee and a minority shareholder of Sotio; K.K., S.S., E.D., P.T., D.S. (Dmitry Stakheev), L.V. and D.S. (Daniel Smrz) declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Schlueter, M.; Chan, K.; Lasry, R.; Price, M. The cost of cancer—A comparative analysis of the direct medical costs of cancer and other major chronic diseases in Europe. PLoS ONE 2020, 15, e0241354. [Google Scholar] [CrossRef] [PubMed]
  3. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  4. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef]
  5. Nixon, N.A.; Blais, N.; Ernst, S.; Kollmannsberger, C.; Bebb, G.; Butler, M.; Smylie, M.; Verma, S. Current landscape of immunotherapy in the treatment of solid tumours, with future opportunities and challenges. Curr. Oncol. 2018, 25, e373–e384. [Google Scholar] [CrossRef]
  6. Tang, J.; Yu, J.X.; Hubbard-Lucey, V.M.; Neftelinov, S.T.; Hodge, J.P.; Lin, Y. Trial watch: The clinical trial landscape for PD1/PDL1 immune checkpoint inhibitors. Nat. Rev. Drug. Discov. 2018, 17, 854–855. [Google Scholar] [CrossRef]
  7. Upadhaya, S.; Neftelinov, S.T.; Hodge, J.; Campbell, J. Challenges and opportunities in the PD1/PDL1 inhibitor clinical trial landscape. Nat. Rev. Drug. Discov. 2022, 21, 482–483. [Google Scholar] [CrossRef]
  8. Upadhaya, S.; Neftelino, S.T.; Hodge, J.P.; Oliva, C.; Campbell, J.R.; Yu, J.X. Combinations take centre stage in PD1/PDL1 inhibitor clinical trials. Nat. Rev. Drug. Discov. 2021, 20, 168–169. [Google Scholar] [CrossRef]
  9. Tung, I.; Sahu, A. Immune Checkpoint Inhibitor in First-Line Treatment of Metastatic Renal Cell Carcinoma: A Review of Current Evidence and Future Directions. Front. Oncol. 2021, 11, 707214. [Google Scholar] [CrossRef]
  10. Huang, Z.; Su, W.; Lu, T.; Wang, Y.; Dong, Y.; Qin, Y.; Liu, D.; Sun, L.; Jiao, W. First-Line Immune-Checkpoint Inhibitors in Non-Small Cell Lung Cancer: Current Landscape and Future Progress. Front. Pharmacol. 2020, 11, 578091. [Google Scholar] [CrossRef]
  11. Borcoman, E.; Marret, G.; Le Tourneau, C. Paradigm Change in First-Line Treatment of Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma. Cancers 2021, 13, 2573. [Google Scholar] [CrossRef]
  12. Cannon, M.J.; Block, M.S.; Morehead, L.C.; Knutson, K.L. The evolving clinical landscape for dendritic cell vaccines and cancer immunotherapy. Immunotherapy 2019, 11, 75–79. [Google Scholar] [CrossRef]
  13. Rosenberg, S.A.; Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef]
  14. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef]
  15. Sengsayadeth, S.; Savani, B.N.; Oluwole, O.; Dholaria, B. Overview of approved CAR-T therapies, ongoing clinical trials, and its impact on clinical practice. EJHaem 2022, 3 (Suppl. 1), 6–10. [Google Scholar] [CrossRef]
  16. Mullard, A. FDA approval of Immunocore’s first-in-class TCR therapeutic broadens depth of the T cell engager platform. Nat. Rev. Drug. Discov. 2022, 21, 170. [Google Scholar] [CrossRef]
  17. Carretero-Gonzalez, A.; Lora, D.; Ghanem, I.; Zugazagoitia, J.; Castellano, D.; Sepulveda, J.M.; Lopez-Martin, J.A.; Paz-Ares, L.; de Velasco, G. Analysis of response rate with ANTI PD1/PD-L1 monoclonal antibodies in advanced solid tumors: A meta-analysis of randomized clinical trials. Oncotarget 2018, 9, 8706–8715. [Google Scholar] [CrossRef]
  18. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef]
  19. Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front. Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef]
  20. Kamada, T.; Togashi, Y.; Tay, C.; Ha, D.; Sasaki, A.; Nakamura, Y.; Sato, E.; Fukuoka, S.; Tada, Y.; Tanaka, A.; et al. PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 9999–10008. [Google Scholar] [CrossRef] [Green Version]
  21. Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef]
  22. Brown, M.A.; Hatfield, J.K. Mast Cells are Important Modifiers of Autoimmune Disease: With so Much Evidence, why is There Still Controversy? Front. Immunol. 2012, 3, 147. [Google Scholar] [CrossRef]
  23. Brown, J.M.; Wilson, T.M.; Metcalfe, D.D. The mast cell and allergic diseases: Role in pathogenesis and implications for therapy. Clin. Exp. Allergy 2008, 38, 4–18. [Google Scholar] [CrossRef]
  24. Dahlin, J.S.; Maurer, M.; Metcalfe, D.D.; Pejler, G.; Sagi-Eisenberg, R.; Nilsson, G. The ingenious mast cell: Contemporary insights into mast cell behavior and function. Allergy 2022, 77, 83–99. [Google Scholar] [CrossRef]
  25. Wernersson, S.; Pejler, G. Mast cell secretory granules: Armed for battle. Nat. Rev. Immunol. 2014, 14, 478–494. [Google Scholar] [CrossRef]
  26. Peavy, R.D.; Metcalfe, D.D. Understanding the mechanisms of anaphylaxis. Curr. Opin. Allergy Clin. Immunol. 2008, 8, 310–315. [Google Scholar] [CrossRef]
  27. Reber, L.L.; Hernandez, J.D.; Galli, S.J. The pathophysiology of anaphylaxis. J. Allergy Clin. Immunol. 2017, 140, 335–348. [Google Scholar] [CrossRef]
  28. Ito, T.; Smrž, D.; Jung, M.Y.; Bandara, G.; Desai, A.; Smržová, S.; Kuehn, H.S.; Beaven, M.A.; Metcalfe, D.D.; Gilfillan, A.M. Stem Cell Factor Programs the Mast Cell Activation Phenotype. J. Immunol. 2012, 188, 5428–5437. [Google Scholar] [CrossRef]
  29. Hundley, T.R.; Gilfillan, A.M.; Tkaczyk, C.; Andrade, M.V.; Metcalfe, D.D.; Beaven, M.A. Kit and FcεRI mediate unique and convergent signals for release of inflammatory mediators from human mast cells. Blood 2004, 104, 2410–2417. [Google Scholar] [CrossRef]
  30. Smrž, D.; Bandara, G.; Beaven, M.A.; Metcalfe, D.D.; Gilfillan, A.M. Prevention of F-actin assembly switches the response to SCF from chemotaxis to degranulation in human mast cells. Eur. J. Immunol. 2013, 43, 1873–1882. [Google Scholar] [CrossRef] [Green Version]
  31. Quan, P.L.; Sabate-Bresco, M.; Guo, Y.; Martin, M.; Gastaminza, G. The Multifaceted Mas-Related G Protein-Coupled Receptor Member X2 in Allergic Diseases and Beyond. Int. J. Mol. Sci. 2021, 22, 4421. [Google Scholar] [CrossRef] [PubMed]
  32. Nakae, S.; Suto, H.; Kakurai, M.; Sedgwick, J.D.; Tsai, M.; Galli, S.J. Mast cells enhance T cell activation: Importance of mast cell-derived TNF. Proc. Natl. Acad. Sci. USA 2005, 102, 6467–6472. [Google Scholar] [CrossRef] [PubMed]
  33. Frossi, B.; Mion, F.; Pucillo, C. Deciphering new mechanisms on T-cell co-stimulation by human mast cells. Eur. J. Immunol. 2016, 46, 1105–1108. [Google Scholar] [CrossRef] [PubMed]
  34. Mantri, C.K.; John, A.L.S. Immune synapses between mast cells and gamma delta T cells limit viral infection. J. Clin. Investig. 2019, 129, 1094–1108. [Google Scholar] [CrossRef] [PubMed]
  35. Portales-Cervantes, L.; Dawod, B.; Marshall, J.S. Mast Cells and Natural Killer Cells-A Potentially Critical Interaction. Viruses 2019, 11, 514. [Google Scholar] [CrossRef] [PubMed]
  36. Lichterman, J.N.; Reddy, S.M. Mast Cells: A New Frontier for Cancer Immunotherapy. Cells 2021, 10, 1270. [Google Scholar] [CrossRef] [PubMed]
  37. Metcalfe, D.D.; Baram, D.; Mekori, Y.A. Mast cells. Physiol. Rev. 1997, 77, 1033–1079. [Google Scholar] [CrossRef]
  38. Komi, D.E.A.; Redegeld, F.A. Role of Mast Cells in Shaping the Tumor Microenvironment. Clin. Rev. Allergy Immunol. 2020, 58, 313–325. [Google Scholar] [CrossRef]
  39. Segura-Villalobos, D.; Ramirez-Moreno, I.G.; Martinez-Aguilar, M.; Ibarra-Sanchez, A.; Munoz-Bello, J.O.; Anaya-Rubio, I.; Padilla, A.; Macias-Silva, M.; Lizano, M.; Gonzalez-Espinosa, C. Mast Cell-Tumor Interactions: Molecular Mechanisms of Recruitment, Intratumoral Communication and Potential Therapeutic Targets for Tumor Growth. Cells 2022, 11, 349. [Google Scholar] [CrossRef]
  40. Mukai, K.; Tsai, M.; Saito, H.; Galli, S.J. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol. Rev. 2018, 282, 121–150. [Google Scholar] [CrossRef]
  41. Martner, A.; Wiktorin, H.G.; Lenox, B.; Ewald Sander, F.; Aydin, E.; Aurelius, J.; Thoren, F.B.; Stahlberg, A.; Hermodsson, S.; Hellstrand, K. Histamine promotes the development of monocyte-derived dendritic cells and reduces tumor growth by targeting the myeloid NADPH oxidase. J. Immunol. 2015, 194, 5014–5021. [Google Scholar] [CrossRef]
  42. Zhao, J.; Hou, Y.; Yin, C.; Hu, J.; Gao, T.; Huang, X.; Zhang, X.; Xing, J.; An, J.; Wan, S.; et al. Upregulation of histamine receptor H1 promotes tumor progression and contributes to poor prognosis in hepatocellular carcinoma. Oncogene 2020, 39, 1724–1738. [Google Scholar] [CrossRef]
  43. Rabelo Melo, F.; Santosh Martin, S.; Sommerhoff, C.P.; Pejler, G. Exosome-mediated uptake of mast cell tryptase into the nucleus of melanoma cells: A novel axis for regulating tumor cell proliferation and gene expression. Cell Death Dis. 2019, 10, 659. [Google Scholar] [CrossRef] [PubMed]
  44. Guo, X.; Zhai, L.; Xue, R.; Shi, J.; Zeng, Q.; Gao, C. Mast Cell Tryptase Contributes to Pancreatic Cancer Growth through Promoting Angiogenesis via Activation of Angiopoietin-1. Int. J. Mol. Sci. 2016, 17, 834. [Google Scholar] [CrossRef]
  45. Oldford, S.A.; Haidl, I.D.; Howatt, M.A.; Leiva, C.A.; Johnston, B.; Marshall, J.S. A critical role for mast cells and mast cell-derived IL-6 in TLR2-mediated inhibition of tumor growth. J. Immunol. 2010, 185, 7067–7076. [Google Scholar] [CrossRef]
  46. Abdul-Wahid, A.; Cydzik, M.; Prodeus, A.; Alwash, M.; Stanojcic, M.; Thompson, M.; Huang, E.H.; Shively, J.E.; Gray-Owen, S.D.; Gariepy, J. Induction of antigen-specific TH 9 immunity accompanied by mast cell activation blocks tumor cell engraftment. Int. J. Cancer 2016, 139, 841–853. [Google Scholar] [CrossRef]
  47. Bodduluri, S.R.; Mathis, S.; Maturu, P.; Krishnan, E.; Satpathy, S.R.; Chilton, P.M.; Mitchell, T.C.; Lira, S.; Locati, M.; Mantovani, A.; et al. Mast Cell-Dependent CD8(+) T-cell Recruitment Mediates Immune Surveillance of Intestinal Tumors in Apc(Min/+) Mice. Cancer Immunol. Res. 2018, 6, 332–347. [Google Scholar] [CrossRef]
  48. Ahani, E.; Fereydouni, M.; Motaghed, M.; Kepley, C.L. Identification and Characterization of Tunneling Nanotubes Involved in Human Mast Cell FcepsilonRI-Mediated Apoptosis of Cancer Cells. Cancers 2022, 14, 2944. [Google Scholar] [CrossRef]
  49. Della Rovere, F.; Granata, A.; Monaco, M.; Basile, G. Phagocytosis of cancer cells by mast cells in breast cancer. Anticancer Res. 2009, 29, 3157–3161. [Google Scholar]
  50. Sawatsubashi, M.; Yamada, T.; Fukushima, N.; Mizokami, H.; Tokunaga, O.; Shin, T. Association of vascular endothelial growth factor and mast cells with angiogenesis in laryngeal squamous cell carcinoma. Virchows Arch. Int. J. Pathol. 2000, 436, 243–248. [Google Scholar] [CrossRef]
  51. Gan, P.Y.; Summers, S.A.; Ooi, J.D.; O’Sullivan, K.M.; Tan, D.S.; Muljadi, R.C.; Odobasic, D.; Kitching, A.R.; Holdsworth, S.R. Mast cells contribute to peripheral tolerance and attenuate autoimmune vasculitis. J. Am. Soc. Nephrol. JASN 2012, 23, 1955–1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Leyva-Castillo, J.M.; Das, M.; Artru, E.; Yoon, J.; Galand, C.; Geha, R.S. Mast cell-derived IL-13 downregulates IL-12 production by skin dendritic cells to inhibit the TH1 cell response to cutaneous antigen exposure. J. Allergy Clin. Immunol. 2021, 147, 2305–2315.e3. [Google Scholar] [CrossRef] [PubMed]
  53. Rodrigues, C.P.; Ferreira, A.C.; Pinho, M.P.; de Moraes, C.J.; Bergami-Santos, P.C.; Barbuto, J.A. Tolerogenic IDO(+) Dendritic Cells Are Induced by PD-1-Expressing Mast Cells. Front. Immunol. 2016, 7, 9. [Google Scholar] [CrossRef] [PubMed]
  54. Visciano, C.; Liotti, F.; Prevete, N.; Cali, G.; Franco, R.; Collina, F.; de Paulis, A.; Marone, G.; Santoro, M.; Melillo, R.M. Mast cells induce epithelial-to-mesenchymal transition and stem cell features in human thyroid cancer cells through an IL-8-Akt-Slug pathway. Oncogene 2015, 34, 5175–5186. [Google Scholar] [CrossRef]
  55. Desai, A.; Jung, M.Y.; Olivera, A.; Gilfillan, A.M.; Prussin, C.; Kirshenbaum, A.S.; Beaven, M.A.; Metcalfe, D.D. IL-6 promotes an increase in human mast cell numbers and reactivity through suppression of suppressor of cytokine signaling 3. J. Allergy Clin. Immunol. 2016, 137, 1863–1871.e6. [Google Scholar] [CrossRef]
  56. Jung, M.Y.; Smrž, D.; Desai, A.; Bandara, G.; Ito, T.; Iwaki, S.; Kang, J.H.; Andrade, M.V.; Hilderbrand, S.C.; Brown, J.M.; et al. IL-33 Induces a Hyporesponsive Phenotype in Human and Mouse Mast Cells. J. Immunol. 2013, 190, 531–538. [Google Scholar] [CrossRef]
  57. De Souza, D.A., Jr.; Toso, V.D.; Campos, M.R.; Lara, V.S.; Oliver, C.; Jamur, M.C. Expression of mast cell proteases correlates with mast cell maturation and angiogenesis during tumor progression. PLoS ONE 2012, 7, e40790. [Google Scholar] [CrossRef]
  58. Leveque, E.; Rouch, A.; Syrykh, C.; Mazieres, J.; Brouchet, L.; Valitutti, S.; Espinosa, E.; Lafouresse, F. Phenotypic and Histological Distribution Analysis Identify Mast Cell Heterogeneity in Non-Small Cell Lung Cancer. Cancers 2022, 14, 1394. [Google Scholar] [CrossRef]
  59. Derakhshan, T.; Samuchiwal, S.K.; Hallen, N.; Bankova, L.G.; Boyce, J.A.; Barrett, N.A.; Austen, K.F.; Dwyer, D.F. Lineage-specific regulation of inducible and constitutive mast cells in allergic airway inflammation. J. Exp. Med. 2021, 218, e20200321. [Google Scholar] [CrossRef]
  60. Dwyer, D.F.; Barrett, N.A.; Austen, K.F.; Immunological Genome Project, C. Expression profiling of constitutive mast cells reveals a unique identity within the immune system. Nat. Immunol. 2016, 17, 878–887. [Google Scholar] [CrossRef]
  61. Dwyer, D.F.; Ordovas-Montanes, J.; Allon, S.J.; Buchheit, K.M.; Vukovic, M.; Derakhshan, T.; Feng, C.; Lai, J.; Hughes, T.K.; Nyquist, S.K.; et al. Human airway mast cells proliferate and acquire distinct inflammation-driven phenotypes during type 2 inflammation. Sci. Immunol. 2021, 6, eabb7221. [Google Scholar] [CrossRef]
  62. Varricchi, G.; de Paulis, A.; Marone, G.; Galli, S.J. Future Needs in Mast Cell Biology. Int. J. Mol. Sci. 2019, 20, 4397. [Google Scholar] [CrossRef]
  63. Cildir, G.; Yip, K.H.; Pant, H.; Tergaonkar, V.; Lopez, A.F.; Tumes, D.J. Understanding mast cell heterogeneity at single cell resolution. Trends Immunol. 2021, 42, 523–535. [Google Scholar] [CrossRef]
  64. Ferlazzo, G.; Morandi, B. Cross-Talks between Natural Killer Cells and Distinct Subsets of Dendritic Cells. Front. Immunol. 2014, 5, 159. [Google Scholar] [CrossRef]
  65. Keller, C.W.; Freigang, S.; Lunemann, J.D. Reciprocal Crosstalk between Dendritic Cells and Natural Killer T Cells: Mechanisms and Therapeutic Potential. Front. Immunol. 2017, 8, 570. [Google Scholar] [CrossRef]
  66. Bhattacharya, P.; Ismail, N.; Saxena, A.; Gannavaram, S.; Dey, R.; Oljuskin, T.; Akue, A.; Takeda, K.; Yu, J.; Karmakar, S.; et al. Neutrophil-dendritic cell interaction plays an important role in live attenuated Leishmania vaccine induced immunity. PLoS Negl. Trop. Dis. 2022, 16, e0010224. [Google Scholar] [CrossRef]
  67. Carroll-Portillo, A.; Cannon, J.L.; te Riet, J.; Holmes, A.; Kawakami, Y.; Kawakami, T.; Cambi, A.; Lidke, D.S. Mast cells and dendritic cells form synapses that facilitate antigen transfer for T cell activation. J. Cell Biol. 2015, 210, 851–864. [Google Scholar] [CrossRef]
  68. Haniffa, M.; Collin, M.; Ginhoux, F. Ontogeny and functional specialization of dendritic cells in human and mouse. Adv. Immunol. 2013, 120, 1–49. [Google Scholar]
  69. Foti, M.; Granucci, F.; Ricciardi-Castagnoli, P. A central role for tissue-resident dendritic cells in innate responses. Trends Immunol. 2004, 25, 650–654. [Google Scholar] [CrossRef]
  70. Fucikova, J.; Palova-Jelinkova, L.; Bartunkova, J.; Spisek, R. Induction of Tolerance and Immunity by Dendritic Cells: Mechanisms and Clinical Applications. Front. Immunol. 2019, 10, 2393. [Google Scholar] [CrossRef]
  71. Dalod, M.; Chelbi, R.; Malissen, B.; Lawrence, T. Dendritic cell maturation: Functional specialization through signaling specificity and transcriptional programming. EMBO J. 2014, 33, 1104–1116. [Google Scholar] [CrossRef]
  72. Spisek, R.; Bougras, G.; Ebstein, F.; Masse, D.; Meflah, K.; McIlroy, D.; Gregoire, M. Transient exposure of dendritic cells to maturation stimuli is sufficient to induce complete phenotypic maturation while preserving their capacity to respond to subsequent restimulation. Cancer Immunol. Immunother. CII 2003, 52, 445–454. [Google Scholar] [CrossRef]
  73. Segura, E. Human dendritic cell subsets: An updated view of their ontogeny and functional specialization. Eur. J. Immunol. 2022. [Google Scholar] [CrossRef]
  74. Ye, Y.; Gaugler, B.; Mohty, M.; Malard, F. Plasmacytoid dendritic cell biology and its role in immune-mediated diseases. Clin. Transl. Immunol. 2020, 9, e1139. [Google Scholar] [CrossRef]
  75. Zhang, S.; Chopin, M.; Nutt, S.L. Type 1 conventional dendritic cells: Ontogeny, function, and emerging roles in cancer immunotherapy. Trends Immunol. 2021, 42, 1113–1127. [Google Scholar] [CrossRef]
  76. Saito, Y.; Komori, S.; Kotani, T.; Murata, Y.; Matozaki, T. The Role of Type-2 Conventional Dendritic Cells in the Regulation of Tumor Immunity. Cancers 2022, 14, 1976. [Google Scholar] [CrossRef]
  77. Sichien, D.; Lambrecht, B.N.; Guilliams, M.; Scott, C.L. Development of conventional dendritic cells: From common bone marrow progenitors to multiple subsets in peripheral tissues. Mucosal Immunol. 2017, 10, 831–844. [Google Scholar] [CrossRef]
  78. 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, eaah4573. [Google Scholar] [CrossRef]
  79. Villar, J.; Segura, E. The More, the Merrier: DC3s Join the Human Dendritic Cell Family. Immunity 2020, 53, 233–235. [Google Scholar] [CrossRef]
  80. Coillard, A.; Segura, E. In vivo Differentiation of Human Monocytes. Front. Immunol. 2019, 10, 1907. [Google Scholar] [CrossRef]
  81. Tang-Huau, T.L.; Gueguen, P.; Goudot, C.; Durand, M.; Bohec, M.; Baulande, S.; Pasquier, B.; Amigorena, S.; Segura, E. Human in vivo-generated monocyte-derived dendritic cells and macrophages cross-present antigens through a vacuolar pathway. Nat. Commun. 2018, 9, 2570. [Google Scholar] [CrossRef] [PubMed]
  82. Han, P.; Hanlon, D.; Sobolev, O.; Chaudhury, R.; Edelson, R.L. Ex vivo dendritic cell generation—A critical comparison of current approaches. Int. Rev. Cell Mol. Biol. 2019, 349, 251–307. [Google Scholar] [PubMed]
  83. Cechim, G.; Chies, J.A.B. In vitro generation of human monocyte-derived dendritic cells methodological aspects in a comprehensive review. An. Da Acad. Bras. De Cienc. 2019, 91. [Google Scholar] [CrossRef]
  84. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef]
  85. Noubade, R.; Majri-Morrison, S.; Tarbell, K.V. Beyond cDC1: Emerging Roles of DC Crosstalk in Cancer Immunity. Front. Immunol. 2019, 10, 1014. [Google Scholar] [CrossRef]
  86. Koucky, V.; Boucek, J.; Fialova, A. Immunology of Plasmacytoid Dendritic Cells in Solid Tumors: A Brief Review. Cancers 2019, 11, 470. [Google Scholar] [CrossRef]
  87. Plesca, I.; Muller, L.; Bottcher, J.P.; Medyouf, H.; Wehner, R.; Schmitz, M. Tumor-associated human dendritic cell subsets: Phenotype, functional orientation, and clinical relevance. Eur. J. Immunol. 2022. [Google Scholar] [CrossRef]
  88. Del Prete, A.; Sozio, F.; Barbazza, I.; Salvi, V.; Tiberio, L.; Laffranchi, M.; Gismondi, A.; Bosisio, D.; Schioppa, T.; Sozzani, S. Functional Role of Dendritic Cell Subsets in Cancer Progression and Clinical Implications. Int. J. Mol. Sci. 2020, 21, 3930. [Google Scholar] [CrossRef]
  89. Brombacher, E.C.; Everts, B. Shaping of Dendritic Cell Function by the Metabolic Micro-Environment. Front. Endocrinol. 2020, 11, 555. [Google Scholar] [CrossRef]
  90. Raaijmakers, T.K.; Ansems, M. Microenvironmental derived factors modulating dendritic cell function and vaccine efficacy: The effect of prostanoid receptor and nuclear receptor ligands. Cancer Immunol. Immunother. CII 2018, 67, 1789–1796. [Google Scholar] [CrossRef]
  91. Di Blasio, S.; van Wigcheren, G.F.; Becker, A.; van Duffelen, A.; Gorris, M.; Verrijp, K.; Stefanini, I.; Bakker, G.J.; Bloemendal, M.; Halilovic, A.; et al. The tumour microenvironment shapes dendritic cell plasticity in a human organotypic melanoma culture. Nat. Commun. 2020, 11, 2749. [Google Scholar] [CrossRef]
  92. Peng, X.; He, Y.; Huang, J.; Tao, Y.; Liu, S. Metabolism of Dendritic Cells in Tumor Microenvironment: For Immunotherapy. Front. Immunol. 2021, 12, 613492. [Google Scholar] [CrossRef]
  93. Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef]
  94. MacNabb, B.W.; Tumuluru, S.; Chen, X.; Godfrey, J.; Kasal, D.N.; Yu, J.; Jongsma, M.L.M.; Spaapen, R.M.; Kline, D.E.; Kline, J. Dendritic cells can prime anti-tumor CD8(+) T cell responses through major histocompatibility complex cross-dressing. Immunity 2022, 55, 982–997.e8. [Google Scholar]
  95. Li, Y.; Zhao, C.; Liu, J.; Lu, Z.; Lu, M.; Gu, J.; Liu, R. CD1d highly expressed on DCs reduces lung tumor burden by enhancing antitumor immunity. Oncol. Rep. 2019, 41, 2679–2688. [Google Scholar] [CrossRef]
  96. Iwanowycz, S.; Ngoi, S.; Li, Y.; Hill, M.; Koivisto, C.; Parrish, M.; Guo, B.; Li, Z.; Liu, B. Type 2 dendritic cells mediate control of cytotoxic T cell resistant tumors. JCI Insight 2021, 6, e145885. [Google Scholar] [CrossRef]
  97. Bruno, A.; Pagani, A.; Pulze, L.; Albini, A.; Dallaglio, K.; Noonan, D.M.; Mortara, L. Orchestration of angiogenesis by immune cells. Front. Oncol. 2014, 4, 131. [Google Scholar]
  98. Wang, Y.M.; Qiu, J.J.; Qu, X.Y.; Peng, J.; Lu, C.; Zhang, M.; Zhang, M.X.; Qi, X.L.; Lv, B.; Guo, J.J.; et al. Accumulation of dysfunctional tumor-infiltrating PD-1+ DCs links PD-1/PD-L1 blockade immunotherapeutic response in cervical cancer. Oncoimmunology 2022, 11, 2034257. [Google Scholar] [CrossRef]
  99. Liu, Q.; Zhang, C.; Sun, A.; Zheng, Y.; Wang, L.; Cao, X. Tumor-educated CD11bhighIalow regulatory dendritic cells suppress T cell response through arginase I. J. Immunol. 2009, 182, 6207–6216. [Google Scholar]
  100. Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar]
  101. Fallarino, F.; Grohmann, U.; You, S.; McGrath, B.C.; Cavener, D.R.; Vacca, C.; Orabona, C.; Bianchi, R.; Belladonna, M.L.; Volpi, C.; et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J. Immunol. 2006, 176, 6752–6761. [Google Scholar] [CrossRef]
  102. Belladonna, M.L.; Volpi, C.; Bianchi, R.; Vacca, C.; Orabona, C.; Pallotta, M.T.; Boon, L.; Gizzi, S.; Fioretti, M.C.; Grohmann, U.; et al. Cutting edge: Autocrine TGF-beta sustains default tolerogenesis by IDO-competent dendritic cells. J. Immunol. 2008, 181, 5194–5198. [Google Scholar] [CrossRef]
  103. Llopiz, D.; Ruiz, M.; Infante, S.; Villanueva, L.; Silva, L.; Hervas-Stubbs, S.; Alignani, D.; Guruceaga, E.; Lasarte, J.J.; Sarobe, P. IL-10 expression defines an immunosuppressive dendritic cell population induced by antitumor therapeutic vaccination. Oncotarget 2017, 8, 2659–2671. [Google Scholar] [CrossRef]
  104. Von Bergwelt-Baildon, M.S.; Popov, A.; Saric, T.; Chemnitz, J.; Classen, S.; Stoffel, M.S.; Fiore, F.; Roth, U.; Beyer, M.; Debey, S.; et al. CD25 and indoleamine 2,3-dioxygenase are up-regulated by prostaglandin E2 and expressed by tumor-associated dendritic cells in vivo: Additional mechanisms of T-cell inhibition. Blood 2006, 108, 228–237. [Google Scholar] [CrossRef]
  105. Fainaru, O.; Almog, N.; Yung, C.W.; Nakai, K.; Montoya-Zavala, M.; Abdollahi, A.; D’Amato, R.; Ingber, D.E. Tumor growth and angiogenesis are dependent on the presence of immature dendritic cells. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2010, 24, 1411–1418. [Google Scholar] [CrossRef]
  106. Gupta, Y.H.; Khanom, A.; Acton, S.E. Control of Dendritic Cell Function Within the Tumour Microenvironment. Front. Immunol. 2022, 13, 733800. [Google Scholar] [CrossRef]
  107. Gerhard, G.M.; Bill, R.; Messemaker, M.; Klein, A.M.; Pittet, M.J. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J. Exp. Med. 2021, 218, e20200264. [Google Scholar] [CrossRef]
  108. Dudeck, J.; Froebel, J.; Kotrba, J.; Lehmann, C.H.K.; Dudziak, D.; Speier, S.; Nedospasov, S.A.; Schraven, B.; Dudeck, A. Engulfment of mast cell secretory granules on skin inflammation boosts dendritic cell migration and priming efficiency. J. Allergy Clin. Immunol. 2019, 143, 1849–1864.e4. [Google Scholar] [CrossRef]
  109. Dudeck, J.; Medyukhina, A.; Frobel, J.; Svensson, C.M.; Kotrba, J.; Gerlach, M.; Gradtke, A.C.; Schroder, B.; Speier, S.; Figge, M.T.; et al. Mast cells acquire MHCII from dendritic cells during skin inflammation. J. Exp. Med. 2017, 214, 3791–3811. [Google Scholar] [CrossRef]
  110. De Vries, V.C.; Pino-Lagos, K.; Nowak, E.C.; Bennett, K.A.; Oliva, C.; Noelle, R.J. Mast cells condition dendritic cells to mediate allograft tolerance. Immunity 2011, 35, 550–561. [Google Scholar] [CrossRef]
  111. Dudeck, J.; Ghouse, S.M.; Lehmann, C.H.; Hoppe, A.; Schubert, N.; Nedospasov, S.A.; Dudziak, D.; Dudeck, A. Mast-Cell-Derived TNF Amplifies CD8(+) Dendritic Cell Functionality and CD8(+) T Cell Priming. Cell Rep. 2015, 13, 399–411. [Google Scholar] [CrossRef] [PubMed]
  112. Hackler, Y.; Siebenhaar, F.; Lohning, M.; Maurer, M.; Munoz, M. Mast Cells Modulate Antigen-Specific CD8(+) T Cell Activation During LCMV Infection. Front. Immunol. 2021, 12, 688347. [Google Scholar] [CrossRef] [PubMed]
  113. Somasundaram, R.; Connelly, T.; Choi, R.; Choi, H.; Samarkina, A.; Li, L.; Gregorio, E.; Chen, Y.; Thakur, R.; Abdel-Mohsen, M.; et al. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy. Nat. Commun. 2021, 12, 346. [Google Scholar] [CrossRef] [PubMed]
  114. Panagi, M.; Mpekris, F.; Voutouri, C.; Michael, C.; Constantinidou, A.; Martin, J.D.; Stylianopoulos, T. Abstract 6382: Targeting mast cells restores T cell infiltration and sensitizes sarcomas to PD-L1 inhibition. Cancer Res. 2022, 82 (Suppl. 12), 6382. [Google Scholar] [CrossRef]
  115. Lyons, J.J.; Metcalfe, D.D. Targeting Mast Cells with Biologics. Immunol. Allergy Clin. North Am. 2020, 40, 667–685. [Google Scholar] [CrossRef]
  116. Sokol, K.C.; Amar, N.K.; Starkey, J.; Grant, J.A. Ketotifen in the management of chronic urticaria: Resurrection of an old drug. Ann. Allergy Asthma Immunol. 2013, 111, 433–436. [Google Scholar] [CrossRef] [Green Version]
  117. Cardet, J.C.; Akin, C.; Lee, M.J. Mastocytosis: Update on pharmacotherapy and future directions. Expert Opin. Pharmacother. 2013, 14, 2033–2045. [Google Scholar] [CrossRef]
  118. Abbaspour Babaei, M.; Kamalidehghan, B.; Saleem, M.; Huri, H.Z.; Ahmadipour, F. Receptor tyrosine kinase (c-Kit) inhibitors: A potential therapeutic target in cancer cells. Drug Des. Dev. Ther. 2016, 10, 2443–2459. [Google Scholar] [CrossRef]
  119. Murugesan, G.; Weigle, B.; Crocker, P.R. Siglec and anti-Siglec therapies. Curr. Opin. Chem. Biol. 2021, 62, 34–42. [Google Scholar] [CrossRef]
  120. Cao, K.; Zhang, G.; Zhang, X.; Yang, M.; Wang, Y.; He, M.; Lu, J.; Liu, H. Stromal infiltrating mast cells identify immunoevasive subtype high-grade serous ovarian cancer with poor prognosis and inferior immunotherapeutic response. Oncoimmunology 2021, 10, 1969075. [Google Scholar] [CrossRef]
  121. Hu, G.; Wang, S.; Cheng, P. Tumor-infiltrating tryptase(+) mast cells predict unfavorable clinical outcome in solid tumors. Int. J. Cancer 2018, 142, 813–821. [Google Scholar] [CrossRef]
  122. Li, J.; Peng, G.; Zhu, K.; Jie, X.; Xu, Y.; Rao, X.; Xu, Y.; Chen, Y.; Xing, B.; Wu, G.; et al. PD-1(+) mast cell enhanced by PD-1 blocking therapy associated with resistance to immunotherapy. Cancer Immunol. Immunother. CII 2022. [Google Scholar] [CrossRef]
  123. Dabiri, S.; Huntsman, D.; Makretsov, N.; Cheang, M.; Gilks, B.; Bajdik, C.; Gelmon, K.; Chia, S.; Hayes, M. The presence of stromal mast cells identifies a subset of invasive breast cancers with a favorable prognosis. Mod. Pathol. 2004, 17, 690–695. [Google Scholar] [CrossRef]
  124. Bo, X.; Wang, J.; Suo, T.; Ni, X.; Liu, H.; Shen, S.; Li, M.; Wang, Y.; Liu, H.; Xu, J. Tumor-infiltrating mast cells predict prognosis and gemcitabine-based adjuvant chemotherapeutic benefit in biliary tract cancer patients. BMC Cancer 2018, 18, 313. [Google Scholar] [CrossRef]
  125. Ozdemir, O. Evaluation of human mast cell-mediated cytotoxicity by DIOC18 target cell labeling in flow cytometry. J. Immunol. Methods 2007, 319, 98–103. [Google Scholar] [CrossRef]
  126. Fereydouni, M.; Ahani, E.; Desai, P.; Motaghed, M.; Dellinger, A.; Metcalfe, D.D.; Yin, Y.; Lee, S.H.; Kafri, T.; Bhatt, A.P.; et al. Human Tumor Targeted Cytotoxic Mast Cells for Cancer Immunotherapy. Front. Oncol. 2022, 12, 871390. [Google Scholar] [CrossRef]
  127. Nigro, E.A.; Brini, A.T.; Yenagi, V.A.; Ferreira, L.M.; Achatz-Straussberger, G.; Ambrosi, A.; Sanvito, F.; Soprana, E.; van Anken, E.; Achatz, G.; et al. Cutting Edge: IgE Plays an Active Role in Tumor Immunosurveillance in Mice. J. Immunol. 2016, 197, 2583–2588. [Google Scholar] [CrossRef]
  128. Draber, P.; Halova, I.; Polakovicova, I.; Kawakami, T. Signal transduction and chemotaxis in mast cells. Eur. J. Pharmacol. 2016, 778, 11–23. [Google Scholar] [CrossRef]
  129. Martinez-Lostao, L.; Anel, A.; Pardo, J. How Do Cytotoxic Lymphocytes Kill Cancer Cells? Clin. Cancer Res. 2015, 21, 5047–5056. [Google Scholar] [CrossRef]
  130. Fereydouni, M.; Motaghed, M.; Ahani, E.; Kafri, T.; Dellinger, K.; Metcalfe, D.D.; Kepley, C.L. Harnessing the Anti-Tumor Mediators in Mast Cells as a New Strategy for Adoptive Cell Transfer for Cancer. Front. Oncol. 2022, 12, 830199. [Google Scholar] [CrossRef]
  131. Nussenzweig, M.C.; Steinman, R.M.; Gutchinov, B.; Cohn, Z.A. Dendritic cells are accessory cells for the development of anti-trinitrophenyl cytotoxic T lymphocytes. J. Exp. Med. 1980, 152, 1070–1084. [Google Scholar] [CrossRef]
  132. Dunn, G.P.; Bruce, A.T.; Sheehan, K.C.; Shankaran, V.; Uppaluri, R.; Bui, J.D.; Diamond, M.S.; Koebel, C.M.; Arthur, C.; White, J.M.; et al. A critical function for type I interferons in cancer immunoediting. Nat. Immunol. 2005, 6, 722–729. [Google Scholar] [CrossRef]
  133. Macatonia, S.E.; Hosken, N.A.; Litton, M.; Vieira, P.; Hsieh, C.S.; Culpepper, J.A.; Wysocka, M.; Trinchieri, G.; Murphy, K.M.; O’Garra, A. Dendritic cells produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells. J. Immunol. 1995, 154, 5071–5079. [Google Scholar]
  134. Yu, J.; Sun, H.; Cao, W.; Song, Y.; Jiang, Z. Research progress on dendritic cell vaccines in cancer immunotherapy. Exp. Hematol. Oncol. 2022, 11, 3. [Google Scholar] [CrossRef]
  135. Hu, X.; Chakraborty, N.G.; Sporn, J.R.; Kurtzman, S.H.; Ergin, M.T.; Mukherji, B. Enhancement of cytolytic T lymphocyte precursor frequency in melanoma patients following immunization with the MAGE-1 peptide loaded antigen presenting cell-based vaccine. Cancer Res. 1996, 56, 2479–2483. [Google Scholar]
  136. Mukherji, B.; Chakraborty, N.G.; Yamasaki, S.; Okino, T.; Yamase, H.; Sporn, J.R.; Kurtzman, S.K.; Ergin, M.T.; Ozols, J.; Meehan, J.; et al. Induction of antigen-specific cytolytic T cells in situ in human melanoma by immunization with synthetic peptide-pulsed autologous antigen presenting cells. Proc. Natl. Acad. Sci. USA 1995, 92, 8078–8082. [Google Scholar] [CrossRef]
  137. Nestle, F.O.; Alijagic, S.; Gilliet, M.; Sun, Y.; Grabbe, S.; Dummer, R.; Burg, G.; Schadendorf, D. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat. Med. 1998, 4, 328–332. [Google Scholar] [CrossRef]
  138. Holtl, L.; Rieser, C.; Papesh, C.; Ramoner, R.; Bartsch, G.; Thurnher, M. CD83+ blood dendritic cells as a vaccine for immunotherapy of metastatic renal-cell cancer. Lancet 1998, 352, 1358. [Google Scholar] [CrossRef]
  139. Hsu, F.J.; Benike, C.; Fagnoni, F.; Liles, T.M.; Czerwinski, D.; Taidi, B.; Engleman, E.G.; Levy, R. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat. Med. 1996, 2, 52–58. [Google Scholar] [CrossRef]
  140. Tjoa, B.A.; Simmons, S.J.; Bowes, V.A.; Ragde, H.; Rogers, M.; Elgamal, A.; Kenny, G.M.; Cobb, O.E.; Ireton, R.C.; Troychak, M.J.; et al. Evaluation of phase I/II clinical trials in prostate cancer with dendritic cells and PSMA peptides. Prostate 1998, 36, 39–44. [Google Scholar] [CrossRef]
  141. Cheever, M.A.; Higano, C.S. PROVENGE (Sipuleucel-T) in prostate cancer: The first FDA-approved therapeutic cancer vaccine. Clin. Cancer Res. 2011, 17, 3520–3526. [Google Scholar] [CrossRef] [PubMed]
  142. Wei, X.X.; Kwak, L.; Hamid, A.; He, M.; Sweeney, C.; Flanders, S.C.; Harmon, M.; Choudhury, A.D. Outcomes in men with metastatic castration-resistant prostate cancer who received sipuleucel-T and no immediate subsequent therapy: Experience at Dana Farber and in the PROCEED Registry. Prostate Cancer Prostatic Dis. 2022, 25, 314–319. [Google Scholar] [CrossRef] [PubMed]
  143. Harari, A.; Graciotti, M.; Bassani-Sternberg, M.; Kandalaft, L.E. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat. Rev. Drug. Discov. 2020, 19, 635–652. [Google Scholar] [CrossRef] [PubMed]
  144. Thommen, D.S.; Schumacher, T.N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562. [Google Scholar] [CrossRef]
  145. Wu, P.; Gao, W.; Su, M.; Nice, E.C.; Zhang, W.; Lin, J.; Xie, N. Adaptive Mechanisms of Tumor Therapy Resistance Driven by Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 641469. [Google Scholar] [CrossRef]
  146. Laureano, R.S.; Sprooten, J.; Vanmeerbeerk, I.; Borras, D.M.; Govaerts, J.; Naulaerts, S.; Berneman, Z.N.; Beuselinck, B.; Bol, K.F.; Borst, J.; et al. Trial watch: Dendritic cell (DC)-based immunotherapy for cancer. Oncoimmunology 2022, 11, 2096363. [Google Scholar] [CrossRef]
  147. Zhang, Z.; Lu, M.; Qin, Y.; Gao, W.; Tao, L.; Su, W.; Zhong, J. Neoantigen: A New Breakthrough in Tumor Immunotherapy. Front. Immunol. 2021, 12, 672356. [Google Scholar] [CrossRef]
  148. Sahin, U.; Tureci, O. Personalized vaccines for cancer immunotherapy. Science 2018, 359, 1355–1360. [Google Scholar] [CrossRef] [Green Version]
  149. Wirth, T.C.; Kuhnel, F. Neoantigen Targeting-Dawn of a New Era in Cancer Immunotherapy? Front. Immunol. 2017, 8, 1848. [Google Scholar] [CrossRef]
  150. Ding, Z.; Li, Q.; Zhang, R.; Xie, L.; Shu, Y.; Gao, S.; Wang, P.; Su, X.; Qin, Y.; Wang, Y.; et al. Personalized neoantigen pulsed dendritic cell vaccine for advanced lung cancer. Signal Transduct. Target. Ther. 2021, 6, 26. [Google Scholar] [CrossRef]
  151. Dammeijer, F.; Lievense, L.A.; Kaijen-Lambers, M.E.; van Nimwegen, M.; Bezemer, K.; Hegmans, J.P.; van Hall, T.; Hendriks, R.W.; Aerts, J.G. Depletion of Tumor-Associated Macrophages with a CSF-1R Kinase Inhibitor Enhances Antitumor Immunity and Survival Induced by DC Immunotherapy. Cancer Immunol. Res. 2017, 5, 535–546. [Google Scholar] [CrossRef]
  152. Garg, A.D.; Coulie, P.G.; Van den Eynde, B.J.; Agostinis, P. Integrating Next-Generation Dendritic Cell Vaccines into the Current Cancer Immunotherapy Landscape. Trends Immunol. 2017, 38, 577–593. [Google Scholar] [CrossRef]
  153. Hensler, M.; Rakova, J.; Kasikova, L.; Lanickova, T.; Pasulka, J.; Holicek, P.; Hraska, M.; Hrnciarova, T.; Kadlecova, P.; Schoenenberger, A.; et al. Peripheral gene signatures reveal distinct cancer patient immunotypes with therapeutic implications for autologous DC-based vaccines. Oncoimmunology 2022, 11, 2101596. [Google Scholar] [CrossRef]
  154. Fucikova, J.; Podrazil, M.; Jarolim, L.; Bilkova, P.; Hensler, M.; Becht, E.; Gasova, Z.; Klouckova, J.; Kayserova, J.; Horvath, R.; et al. Phase I/II trial of dendritic cell-based active cellular immunotherapy with DCVAC/PCa in patients with rising PSA after primary prostatectomy or salvage radiotherapy for the treatment of prostate cancer. Cancer Immunol. Immunother. CII 2018, 67, 89–100. [Google Scholar] [CrossRef]
  155. Podrazil, M.; Horvath, R.; Becht, E.; Rozkova, D.; Bilkova, P.; Sochorova, K.; Hromadkova, H.; Kayserova, J.; Vavrova, K.; Lastovicka, J.; et al. Phase I/II clinical trial of dendritic-cell based immunotherapy (DCVAC/PCa) combined with chemotherapy in patients with metastatic, castration-resistant prostate cancer. Oncotarget 2015, 6, 18192–18205. [Google Scholar] [CrossRef]
  156. Fucikova, J.; Hensler, M.; Kasikova, L.; Lanickova, T.; Pasulka, J.; Rakova, J.; Drozenova, J.; Fredriksen, T.; Hraska, M.; Hrnciarova, T.; et al. An Autologous Dendritic Cell Vaccine Promotes Anticancer Immunity in Patients with Ovarian Cancer with Low Mutational Burden and Cold Tumors. Clin. Cancer Res. 2022, 28, 3053–3065. [Google Scholar] [CrossRef]
  157. Taborska, P.; Stakheev, D.; Bartunkova, J.; Smrz, D. Thapsigargin-Stimulated LAD2 Human Mast Cell Line Is a Potent Cellular Adjuvant for the Maturation of Monocyte-Derived Dendritic Cells for Adoptive Cellular Immunotherapy. Int. J. Mol. Sci. 2021, 22, 3978. [Google Scholar] [CrossRef]
  158. Kijima, Y.; Ogunbunmi, E.; Fleischer, S. Drug action of thapsigargin on the Ca2+ pump protein of sarcoplasmic reticulum. J. Biol. Chem. 1991, 266, 22912–22918. [Google Scholar] [CrossRef]
  159. Lytton, J.; Westlin, M.; Hanley, M.R. Thapsigargin inhibits the sarcoplasmic or endoplasmic reticulum Ca-ATPase family of calcium pumps. J. Biol. Chem. 1991, 266, 17067–17071. [Google Scholar] [CrossRef]
  160. Smrz, D.; Cruse, G.; Beaven, M.A.; Kirshenbaum, A.; Metcalfe, D.D.; Gilfillan, A.M. Rictor Negatively Regulates High-Affinity Receptors for IgE-Induced Mast Cell Degranulation. J. Immunol. 2014, 193, 5924–5932. [Google Scholar] [CrossRef]
  161. McDonnell, A.M.; Lesterhuis, W.J.; Khong, A.; Nowak, A.K.; Lake, R.A.; Currie, A.J.; Robinson, B.W. Tumor-infiltrating dendritic cells exhibit defective cross-presentation of tumor antigens, but is reversed by chemotherapy. Eur. J. Immunol. 2015, 45, 49–59. [Google Scholar] [CrossRef] [PubMed]
  162. Vicari, A.P.; Chiodoni, C.; Vaure, C.; Ait-Yahia, S.; Dercamp, C.; Matsos, F.; Reynard, O.; Taverne, C.; Merle, P.; Colombo, M.P.; et al. Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide and anti-interleukin 10 receptor antibody. J. Exp. Med. 2002, 196, 541–549. [Google Scholar] [CrossRef] [PubMed]
  163. Plotkin, J.D.; Elias, M.G.; Fereydouni, M.; Daniels-Wells, T.R.; Dellinger, A.L.; Penichet, M.L.; Kepley, C.L. Human Mast Cells from Adipose Tissue Target and Induce Apoptosis of Breast Cancer Cells. Front. Immunol. 2019, 10, 138. [Google Scholar] [CrossRef] [PubMed]
  164. Yin, Y.; Bai, Y.; Olivera, A.; Desai, A.; Metcalfe, D.D. An optimized protocol for the generation and functional analysis of human mast cells from CD34+ enriched cell populations. J. Immunol. Methods 2017, 448, 105–111. [Google Scholar] [CrossRef]
  165. Daguenet, E.; Louati, S.; Wozny, A.S.; Vial, N.; Gras, M.; Guy, J.B.; Vallard, A.; Rodriguez-Lafrasse, C.; Magne, N. Radiation-induced bystander and abscopal effects: Important lessons from preclinical models. Br. J. Cancer 2020, 123, 339–348. [Google Scholar] [CrossRef]
  166. Le, V.H.; Franko, J.; Paz, B.I.; Singh, G.; Fakih, M.; Chung, V. Chemotherapy-induced early transient increase and surge of CA 19–9 level in patients with pancreatic Adenocarcinoma. Cancer Treat. Res. Commun. 2021, 28, 100397. [Google Scholar] [CrossRef]
  167. Xu, X.; Li, T.; Shen, S.; Wang, J.; Abdou, P.; Gu, Z.; Mo, R. Advances in Engineering Cells for Cancer Immunotherapy. Theranostics 2019, 9, 7889–7905. [Google Scholar] [CrossRef]
  168. Andrea, A.E.; Chiron, A.; Mallah, S.; Bessoles, S.; Sarrabayrouse, G.; Hacein-Bey-Abina, S. Advances in CAR-T Cell Genetic Engineering Strategies to Overcome Hurdles in Solid Tumors Treatment. Front. Immunol. 2022, 13, 830292. [Google Scholar] [CrossRef]
  169. Wei, F.; Cheng, X.X.; Xue, J.Z.; Xue, S.A. Emerging Strategies in TCR-Engineered T Cells. Front. Immunol. 2022, 13, 850358. [Google Scholar] [CrossRef]
  170. Han, B.S.; Ji, S.; Woo, S.; Lee, J.H.; Sin, J.I. Regulation of the translation activity of antigen-specific mRNA is responsible for antigen loss and tumor immune escape in a HER2-expressing tumor model. Sci. Rep. 2019, 9, 2855. [Google Scholar] [CrossRef]
  171. Hubbe, M.L.; Jaehger, D.E.; Andresen, T.L.; Andersen, M.H. Leveraging Endogenous Dendritic Cells to Enhance the Therapeutic Efficacy of Adoptive T-Cell Therapy and Checkpoint Blockade. Front. Immunol. 2020, 11, 578349. [Google Scholar] [CrossRef]
  172. Walsh, S.R.; Simovic, B.; Chen, L.; Bastin, D.; Nguyen, A.; Stephenson, K.; Mandur, T.S.; Bramson, J.L.; Lichty, B.D.; Wan, Y. Endogenous T cells prevent tumor immune escape following adoptive T cell therapy. J. Clin. Investig. 2019, 129, 5400–5410. [Google Scholar] [CrossRef]
  173. Kahkonen, T.E.; Suominen, M.I.; Maki-Jouppila, J.H.E.; Halleen, J.M.; Tanaka, A.; Seiler, M.; Bernoulli, J. Human Immune System Increases Breast Cancer-Induced Osteoblastic Bone Growth in a Humanized Mouse Model without Affecting Normal Bone. J. Immunol. Res. 2019, 2019, 4260987. [Google Scholar] [CrossRef]
  174. Calmeiro, J.; Carrascal, M.A.; Tavares, A.R.; Ferreira, D.A.; Gomes, C.; Cruz, M.T.; Falcao, A.; Neves, B.M. Pharmacological combination of nivolumab with dendritic cell vaccines in cancer immunotherapy: An overview. Pharmacol. Res. 2021, 164, 105309. [Google Scholar] [CrossRef]
  175. Abakushina, E.V.; Popova, L.I.; Zamyatnin, A.A., Jr.; Werner, J.; Mikhailovsky, N.V.; Bazhin, A.V. The Advantages and Challenges of Anticancer Dendritic Cell Vaccines and NK Cells in Adoptive Cell Immunotherapy. Vaccines 2021, 9, 1363. [Google Scholar] [CrossRef]
  176. Jiang, N.; Qiao, G.; Wang, X.; Morse, M.A.; Gwin, W.R.; Zhou, L.; Song, Y.; Zhao, Y.; Chen, F.; Zhou, X.; et al. Dendritic Cell/Cytokine-Induced Killer Cell Immunotherapy Combined with S-1 in Patients with Advanced Pancreatic Cancer: A Prospective Study. Clin. Cancer Res. 2017, 23, 5066–5073. [Google Scholar] [CrossRef]
  177. Qin, V.M.; D’Souza, C.; Neeson, P.J.; Zhu, J.J. Chimeric Antigen Receptor beyond CAR-T Cells. Cancers 2021, 13, 404. [Google Scholar] [CrossRef]
  178. Kang, B.H.; Lee, H.K. Dendritic Cell-Based Immunotherapy in Hot and Cold Tumors. Int. J. Mol. Sci. 2022, 23, 7325. [Google Scholar] [CrossRef]
  179. Lv, Y.; Zhao, Y.; Wang, X.; Chen, N.; Mao, F.; Teng, Y.; Wang, T.; Peng, L.; Zhang, J.; Cheng, P.; et al. Increased intratumoral mast cells foster immune suppression and gastric cancer progression through TNF-alpha-PD-L1 pathway. J. Immunother. Cancer 2019, 7, 54. [Google Scholar] [CrossRef]
  180. Metcalfe, D.D. Mast cells and mastocytosis. Blood 2008, 112, 946–956. [Google Scholar] [CrossRef]
  181. Molderings, G.J.; Zienkiewicz, T.; Homann, J.; Menzen, M.; Afrin, L.B. Risk of solid cancer in patients with mast cell activation syndrome: Results from Germany and USA. F1000Research 2017, 6, 1889. [Google Scholar] [CrossRef]
  182. Ryan, R.J.; Akin, C.; Castells, M.; Wills, M.; Selig, M.K.; Nielsen, G.P.; Ferry, J.A.; Hornick, J.L. Mast cell sarcoma: A rare and potentially under-recognized diagnostic entity with specific therapeutic implications. Mod. Pathol. 2013, 26, 533–543. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The perspective of combined MC- and DC-based ACT. Autologous MCs are produced in vitro from adipose tissue or CD34+-derived stem cell progenitors. DCs are produced in vitro from peripheral blood monocytes or CD34+-derived stem cell progenitors. MCs are sensitized with a selected tumor antigen (Ag)-specific IgE or genetically modified with a chimeric FcεRI-based receptor specific to the selected tumor Ag. The IgE-sensitized or genetically modified MCs are transferred back to the patients (MC ACT). The transferred MCs are specifically chemoattracted to the tumor through the selected Ag concentration gradient. The release of the selected Ag from the tumor is spiked by targeted radiotherapy or chemotherapy. Chemoattracted MCs infiltrate the tumor mass where they eliminate cancer cells. The tumor-infiltrating MCs also interact with endogenous(endo)/tumor-infiltrating DCs to load them with the tumor Ag and restore their antitumor activity through priming tumor Ag-specific T cells. Multiple tumor Ags, including the patient-specific neoAgs, are released from the tumor and captured by endogenous DCs, which primes T cells with a broad repertoire of tumor Ag specificity. The DC-primed T cells expand and infiltrate the tumor. The infiltrated multiple Ag-specific T cells also eliminate evasive variants of cancer cells that are negative for the selected tumor Ag. To corroborate the efficacy of the MC-based ACT, autologous DCs are produced in vitro from peripheral blood monocytes or CD34+-derived stem cell progenitors. The DCs are loaded with the selected tumor antigen or other tumor-associated Ags, matured, and transferred back to the patients (DC ACT). The transferred DCs promote the expansion of the tumor-Ag-specific T cells. The previously cold tumor turns into a hot tumor by infiltrating immune cells with prevailing antitumor activities. Created with BioRender.com (agreement number: HM24B8W5CT).
Figure 1. The perspective of combined MC- and DC-based ACT. Autologous MCs are produced in vitro from adipose tissue or CD34+-derived stem cell progenitors. DCs are produced in vitro from peripheral blood monocytes or CD34+-derived stem cell progenitors. MCs are sensitized with a selected tumor antigen (Ag)-specific IgE or genetically modified with a chimeric FcεRI-based receptor specific to the selected tumor Ag. The IgE-sensitized or genetically modified MCs are transferred back to the patients (MC ACT). The transferred MCs are specifically chemoattracted to the tumor through the selected Ag concentration gradient. The release of the selected Ag from the tumor is spiked by targeted radiotherapy or chemotherapy. Chemoattracted MCs infiltrate the tumor mass where they eliminate cancer cells. The tumor-infiltrating MCs also interact with endogenous(endo)/tumor-infiltrating DCs to load them with the tumor Ag and restore their antitumor activity through priming tumor Ag-specific T cells. Multiple tumor Ags, including the patient-specific neoAgs, are released from the tumor and captured by endogenous DCs, which primes T cells with a broad repertoire of tumor Ag specificity. The DC-primed T cells expand and infiltrate the tumor. The infiltrated multiple Ag-specific T cells also eliminate evasive variants of cancer cells that are negative for the selected tumor Ag. To corroborate the efficacy of the MC-based ACT, autologous DCs are produced in vitro from peripheral blood monocytes or CD34+-derived stem cell progenitors. The DCs are loaded with the selected tumor antigen or other tumor-associated Ags, matured, and transferred back to the patients (DC ACT). The transferred DCs promote the expansion of the tumor-Ag-specific T cells. The previously cold tumor turns into a hot tumor by infiltrating immune cells with prevailing antitumor activities. Created with BioRender.com (agreement number: HM24B8W5CT).
Ijms 23 11080 g001
Table 1. The impact of MCs on the tumor microenvironment.
Table 1. The impact of MCs on the tumor microenvironment.
EffectorImpactMalignancy/ModelReference
Anti-tumorigenic MCs
TryptaseInhibition of tumor cell proliferationMelanoma[43]
HistaminePromotion of DC development and maturationLymphoma[41]
IL-6Inhibition of tumor growthMelanoma[45]
Nanotubes/TNF-αCytotoxic activityBreast cancer[48]
IL-9-mediated MC activityTumor engraftment inhibitionColon carcinoma[46]
CCR2, CCR7, Leukotriene B4CD8+ T cell recruitment and antigen-mediated activationIntestinal tumors[47]
PhagocytosisTumor cell clearanceBreast cancer[49]
Pro-tumorigenic MCs
TryptasePromotion of angiogenesisPancreatic cancer[44]
HistamineEnhanced proliferation of histamine receptor R1+ tumor cellsHepatocellular carcinoma[42]
VEGFPromotion of angiogenesisLaryngeal squamous cell carcinoma[50]
IL-10Anti-inflammatory/immunosuppressive-[51]
IL-13Anti-inflammatory/immunosuppressive-[52]
IL-8Promotion of EMTThyroid cancer[54]
PD-1Induction of IDO+ tolerogenic DCs-[53]
Table 2. The impact of DCs on the tumor microenvironment.
Table 2. The impact of DCs on the tumor microenvironment.
Effector/MarkerImpactReference
Anti-tumorigenic DCs
IL-12+ DCsPromotes CD8+ T cell responses[93]
DC cross-dressingCD8+ T cell priming[94]
CD1d++ DCsIncreased activation of NK T cells, CD4+, and CD8+ T cells[95]
cDC2Increased control of cytotoxic T-cell-resistant tumors via CD4+ T-cell-mediated activation of myeloid cells[96]
Mature cDCs and pDCAngiogenesis inhibition[97]
Pro-tumorigenic DCs
PD-1+ DCsCD8+ T cell inactivation[98]
Arginase+ DCsArginine-deprivation-mediated inhibition of CD4+ T cell proliferation
ROS-mediated inhibition of CD8+ T cells
[99]
[100]
IDO+ DCsTryptophan-depletion-mediated inhibition of CD8+ T cells
Tryptophan-metabolite-mediated expansion of CD4+ Treg cells
[101]
TGF-β+ DCsAnti-inflammatory/immunosuppressive[102]
IL-10+/PD-L1+ DCsImpaired CD8+ T cell activation[103]
sIL25+ DCsInhibition of T cell proliferation by IL-2 depletion[104]
Immature DCsAngiogenesis promotion[105]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kalkusova, K.; Smite, S.; Darras, E.; Taborska, P.; Stakheev, D.; Vannucci, L.; Bartunkova, J.; Smrz, D. Mast Cells and Dendritic Cells as Cellular Immune Checkpoints in Immunotherapy of Solid Tumors. Int. J. Mol. Sci. 2022, 23, 11080. https://doi.org/10.3390/ijms231911080

AMA Style

Kalkusova K, Smite S, Darras E, Taborska P, Stakheev D, Vannucci L, Bartunkova J, Smrz D. Mast Cells and Dendritic Cells as Cellular Immune Checkpoints in Immunotherapy of Solid Tumors. International Journal of Molecular Sciences. 2022; 23(19):11080. https://doi.org/10.3390/ijms231911080

Chicago/Turabian Style

Kalkusova, Katerina, Sindija Smite, Elea Darras, Pavla Taborska, Dmitry Stakheev, Luca Vannucci, Jirina Bartunkova, and Daniel Smrz. 2022. "Mast Cells and Dendritic Cells as Cellular Immune Checkpoints in Immunotherapy of Solid Tumors" International Journal of Molecular Sciences 23, no. 19: 11080. https://doi.org/10.3390/ijms231911080

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop