Next Article in Journal
Overcoming Resistance to Immune Checkpoint Inhibitor Therapy Using Calreticulin-Inducing Nanoparticle
Next Article in Special Issue
Cancer Vaccines: From the State of the Art to the Most Promising Frontiers in the Treatment of Colorectal Cancer
Previous Article in Journal
Doxorubicin Activity Is Modulated by Traditional Herbal Extracts in a 2D and 3D Multicellular Sphere Model of Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Frenemies in the Microenvironment: Harnessing Mast Cells for Cancer Immunotherapy

Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
*
Author to whom correspondence should be addressed.
Submission received: 5 May 2023 / Revised: 2 June 2023 / Accepted: 7 June 2023 / Published: 9 June 2023
(This article belongs to the Special Issue Immunotherapeutic Strategies in Cancer and Chronic Infection)

Abstract

:
Tumor development, progression, and resistance to therapies are influenced by the interactions between tumor cells and the surrounding microenvironment, comprising fibroblasts, immune cells, and extracellular matrix proteins. In this context, mast cells (MCs) have recently emerged as important players. Yet, their role is still controversial, as MCs can exert pro- or anti-tumor functions in different tumor types depending on their location within or around the tumor mass and their interaction with other components of the tumor microenvironment. In this review, we describe the main aspects of MC biology and the different contribution of MCs in promoting or inhibiting cancer growth. We then discuss possible therapeutic strategies aimed at targeting MCs for cancer immunotherapy, which include: (1) targeting c-Kit signaling; (2) stabilizing MC degranulation; (3) triggering activating/inhibiting receptors; (4) modulating MC recruitment; (5) harnessing MC mediators; (6) adoptive transferring of MCs. Such strategies should aim to either restrain or sustain MC activity according to specific contexts. Further investigation would allow us to better dissect the multifaceted roles of MCs in cancer and tailor novel approaches for an “MC-guided” personalized medicine to be used in combination with conventional anti-cancer therapies.

1. Introduction

Cancer development, progression, and resistance to therapy are shaped by stimuli coming from the tumor microenvironment (TME). The TME is composed of different types of cells, including fibroblasts, mesenchymal cells, immune cells, and endothelial cells, along with the presence of extracellular matrix proteins. Immune cells in the TME have the potential to attack tumor cells and therefore impede neoplastic progression. However, tumor cells can evade immune recognition and, in turn, can influence host bystander cells to support and maintain cancer growth and metastasis occurrence through a variety of different mechanisms [1]. The final outcome converges in a tumor-promoting and immune-suppressive microenvironment, as a result of constant crosstalk between cancer cells and the surrounding TME [2].
Commonly associated with allergies and autoimmune disorders, mast cells (MCs) have recently gained increasing attention in the TME, as they are endowed with both pro-angiogenic and pro-tumorigenic functions and have the ability to promote immunosuppression. However, in some instances, MCs can also actively restrain tumor growth and foster anti-tumor immunity. These opposite functions can depend on tumor type, MC peri- or intra-tumor localization, and their interaction with other immune cells.
In this review, we describe the multifaceted roles of MCs in tumor development and discuss possible therapeutic approaches that aim to target MC function for cancer immunotherapy. Still, many issues remain unresolved. The main challenge is to understand the activation state of MCs in the tumor area. The in vivo identification of different biomarkers associated with MC subtypes and activation status could better inform the development of therapeutic regimens for patients. Additionally, newly available technologies to evaluate the spatial distribution of cells within a tissue (i.e., single cell RNAseq or spatial transcriptomic) will help to deeply dissect the crosstalk between MCs and the bystander cells involved in cancer development. This comprehensive characterization would provide crucial insights into the significance of each population within specific cancer contexts.

2. MC Biology

MCs are cells of the innate immunity that originate from pluripotent hematopoietic cells of the bone marrow, and they mature when they reach vascularized tissues [3]. The stem cell factor (SCF)/c-Kit receptor pathway and IL-3 have a pivotal role in MC development, maturation, and proliferation [4]. SCF is also one of the main chemoattractants for MCs [5], together with CCL5, which binds to CCR1 and CCR4 [6].
MCs populate different areas of the body, such as epithelia, mucosa, gastrointestinal tracts, mucus-producing glands, and regions around nerves and blood vessels. Nevertheless, in some species, such as murine rodents, MCs are also present in pulmonary, peritoneal, and mesothelium tracts [3]. On their surface, MCs display several receptors that, once triggered by their ligands, can release a variety of different factors. These include preformed molecules (i.e., histamine, tryptases, proteases, and proteoglycans) and newly synthesized lipid mediators (i.e., leukotrienes and prostaglandins), cytokines (i.e., IL-4, TNFα, TGF-β, IL-1β), and chemokines (i.e., IL-8, CCL2, CCL4) [7]. Preformed mediators settle in large granules placed in MC cytoplasm. Every MC is endowed with about 50–200 granules that are delivered outside the cell in a few seconds upon proper stimulation. This process is called MC degranulation [8].
Murine MCs are categorized into two main classes depending on their location: mucosal-type MCs and connective tissue-type MCs. The former are characterized by the expression of MC protease (mMCP)-1 and -2, whereas the latter have mMCP-4, -5, -6, and the enzyme carboxypeptidase A (CPA). Moreover, human MCs are divided into three categories according to their serine proteases content: tryptase only (MCT), chymase only (MCC), and MCs expressing both tryptase and chymase (MCTC). Even though these classifications are still used, mixed phenotypes can be observed in both human and mouse MC subpopulations [9]. However, it is currently not possible to accurately determine in vivo the prevalence of each MC subpopulation in different cancer settings, and consequently, it is not possible to evaluate their specific effect on final disease outcome.
MCs are best known for their role in allergy and inflammation. The key mechanism of MC activation involves the binding of type E immunoglobulin (IgE) with its high-affinity FcεRI receptor, which triggers a huge secretory response. FcεRI is a heterotetrameric receptor formed by an IgE-binding α subunit, the membrane tetraspanin β subunit, and two disulfide-linked γ subunits that contain one immunoreceptor tyrosine-based activation motif (ITAM)—essential for the activation of the pathway. The signaling starts from the linking between IgE and FcεRI on the surface of MCs. Then, when IgE binds the cognate antigen (Ag), the aggregation of the receptor complex is induced. Consequently, Lyn is activated and phosphorylates ITAM, also activating the spleen tyrosine kinase (Syk). In turn, Lyn and Syk phosphorylate downstream proteins to induce MC activation. In addition, the FcεRI complex promotes the activation of another Src family kinase, Fyn, which phosphorylates Gab2, activating the phosphoinositide 3-kinase (PI3K) pathway [10]. This process culminates in the secretion of histamine, heparin, proteases, cytokines, and other preformed mediators present in the granules. IgE/Ag triggering also induces the de novo synthesis and release of lipid mediators, chemokines, growth factors, and cytokines [11,12].
FcεRI-dependent MC degranulation can potentially be counteracted by inhibitory receptors of the SIGLEC family, mainly SIGLEC-8 [13]. Besides IgE/FcεRI-stimulation, MC degranulation and the release of mediators can also be triggered by the MRGPRX2 receptor (MRGPRB2 in mice), almost selectively expressed by skin MCs [14]. Different compounds can activate the MRGPRX2 receptor, including somatostatin, angiopeptin, mast cell degranulating peptide (MCDP), and β-defensins [15].
MCs also have a pivotal role in innate immunity against microorganisms, as they express numerous receptors such as Toll-like receptors (TLRs; up to ten in humans and up to thirteen in mice) and nucleotide-binding oligomerization domain (NOD), which are able to identify pathogen-associated molecular patterns (PAMPs) [16]. Almost all TLRs are on the cell membrane except for TLR3, TLR7, TLR8, and TLR9, which are inside the cell. Specific TLRs can interact with others, forming heterodimers (i.e., TLR2/TLR1, TLR2/TLR6 and TLR4/TLR6), and most of them (except TLR3, TLR7, and TLR9) rely on MyD88 for their signaling pathway [17]. Notably, TLR4 stimulation can either work via MyD88 to activate NF-kB or independently to stimulate type I IFN response [18].
Other receptors present on the MC surface can lead to the release of pro-inflammatory mediators. IL-33 receptor plays a crucial role in MC biology. It belongs to the TLR/IL-1R (TIR) superfamily, which, upon stimulation, induces the secretion of several factors such as IL-1, IL-6, TNFα, CCL2, and CCL3. IL-33 signaling is also involved in the degranulation, homing, and survival of MCs [19]. Additionally, TSLPR, which triggers the release of IL-4, IL-5, IL-9, and IL-13 [20], and the receptors of the CD300 family, which can either activate (CD300c, CD300lb, CD300lh) or inhibit (CD300a, CD300f, CD300lf) cytokine production [15], represent key receptors for MC biology. MCs can also express PD-L1, a known immunocheckpoint molecule that can inhibit T cell activation [21]. Interestingly, PD-L1 blockade during the allergen sensitization phase can restrain MC degranulation [22]. As MCs are capable of releasing such a plethora of factors as a consequence of distinctive external stimuli, they are key players in several physiological and pathological mechanisms, including cancer.

3. MCs in Cancer

Since the first evaluation of MCs in human tumors by Paul Ehrilich, MCs have gained increased attention in cancer-related fields. Nevertheless, their role is equivocal since they can either promote or inhibit tumor development in different situations [23].

3.1. Pro-Tumorigenic Functions of MCs

MCs can support angiogenesis, inflammation, and homeostasis, thus supporting cancer development. In pancreatic cancer patients, accumulation of MCs within tumor lesions correlates with a dismal prognosis, whereas MCs are low or absent in normal tissues [24]. Accordingly, in the Myc-induced beta cell pancreatic cancer mouse model, tumor development and angiogenesis were reduced when MC degranulation was chemically restrained [25]. In bladder cancer, an increase in MC number is associated with high-grade lesions. Additionally, MCs can enhance bladder cancer metastasis via an ERβ/CCL2/CCR2 axis that leads to epithelial-to-mesenchymal transition (EMT) and MMP9 production by cancer cells [26]. Moreover, in thyroid cancer, MC-derived IL-8 can sustain EMT and stemness by triggering Akt phosphorylation with the consequent activation of Slug in tumor cells [27]. Additionally, the secretion of CXCL1, CXCL10, and histamine by MCs sustains thyroid cancer growth [28]. In line with these results, it has been shown that histamine also has a key role in cholangiocarcinoma. Indeed, impeding histamine secretion by MCs reduces tumor growth and neo-vessel formation [29]. Furthermore, it has been described that, in mice, either the genetic or pharmacologic inhibition of MCs induced the regression of preneoplastic polyps, thus supporting the role of MCs in colon carcinogenesis [30]. In line with these results, it was found that MCs sustain colon cancer growth, while conversely, they can help to resolve colon inflammation by fostering mucosal healing through the degradation of the alarmin IL-33 [31].

3.2. Anti-Tumorigenic Functions of MCs

By contrast, in other types of cancer, MCs appear to have an anti-tumorigenic role. This is the case with lung cancer [32,33] and diffuse large B-cell lymphoma [34], where the presence of MCs is a good prognostic marker.
Mechanistically, in a murine model of intestinal tumorigenesis, it was shown that MCs can induce apoptosis of adenoma cells [35]. It has also been reported that the TLR-2-mediated secretion of IL-6 is responsible for the anti-tumor function of MCs against melanoma and lung cancer, both in vitro and in vivo [36]. Moreover, in non-small cell lung cancer, MC infiltration and their production of TNFα confers improved survival among patients [37]. Indeed, anti-tumor activity mediated by TNFα derived from MCs and its cytotoxic activity was demonstrated by exploiting the WEHI-164 TNFα-sensitive cell line [38]. Furthermore, a recent pan-cancer single cell analysis of immune infiltrate in different solid tumors revealed that, in nasopharyngeal cancer, MCs have a protective role and correlate with good prognosis thanks to their production of TNFα and, in particular, their high TNFα/VEGF ratio [39]. Indeed, the same study revealed a correlation with a bad prognosis of TNFα-negative, VEGF-producing MCs in the other tumor types analyzed, including lung, colon, pancreas, and kidney [39].

3.3. Influence of Tumor Histotype and Localization on MC Function

Interestingly, the role of MCs can dramatically change according to different histotypes of the same tumor. This is the case with breast cancer, where MCs can alternatively promote and prevent the luminal and basal subtypes, respectively [40]. In breast cancer patients, correlation of MCs infiltration with prognosis also depends on the luminal, triple-negative, or basal-like phenotype of the tumor [41]. Similarly, in prostate cancer, we demonstrated that MCs can promote the growth of adenocarcinoma by supplying MMP9 [42] and suppressing the anti-tumor T cell response [43]; however, at the same time, they protect against aggressive neuroendocrine variants [42,44] that can emerge de novo or in resistance to hormone therapy.
Even the localization of MCs within the tumor can influence the outcome. In prostate cancer, intratumoral MCs negatively regulate angiogenesis and cancer growth, whereas peritumoral MCs support tumor development [45,46]. Indeed, intra- and peri-tumoral MCs are characterized by distinct phenotypes [47]. It has also been demonstrated that a high number of extra-tumoral MCs is associated with disease recurrence and metastasis onset after radical prostatectomy [48]. A similar association has also been observed in renal cell carcinoma patients, where peritumoral and intratumoral MCs correlate with bad and good prognosis, respectively [49,50,51]. Finally, it has been recently found that MC infiltration in tumors can be influenced by microbiome composition and that gut dysbiosis can prompt MCs towards pro-metastatic functions in a breast cancer model [52].

4. MC Interaction with Other Immune Cells in the TME

MCs can also regulate the function of other immune cells in the TME, thus influencing either local immunosuppression or anti-tumor immunity. For instance, in a murine hepatocarcinoma model, it has been shown that activated MCs can promote the infiltration of myeloid-derived suppressor cells (MDSCs) through the CCL2/CCR2 axis and their production of IL-17, which in turn recruits Tregs at the tumor site [53]. In the transgenic APCΔ468 mouse model of colon cancer, MCs can stimulate the migration of MDSCs through the production of 5-lipoxygenase, which is in itself essential to produce MC-derived leukotrienes, finally promoting intestinal polyposis [54]. We and others have also shown that MCs can increase the suppressive activity of MDSCs [55] via direct interaction through the CD40L/CD40 axis [43,56]. CD40L on MCs can also promote the expansion of IL-10-producing regulatory B cells (Breg) [57]. Moreover, by using several cancer cell lines, Huang and colleagues showed that SCF-activated MCs promote the release of adenosine that suppresses NK and effector T cells, also increasing the frequency of intratumor Treg cells [58]. Notably, it was recently demonstrated in a mouse melanoma model that MCs contribute to resistance to anti-PD-1 therapy and that their inhibition with sunitinib or imatinib efficiently synergize with anti-PD-1 toward tumor regression [59].
On the other hand, in colorectal cancer, MCs can switch the function of Tregs, which downregulate IL-10 and start to produce IL-17, thereby acquiring a pro-inflammatory phenotype [60]. Notably, MC-mediated skew of Tregs and effector T cells towards Th17 relies on the crosstalk between the OX40L/OX40 axis and the production of IL-6 [61]. However, Tregs can exploit the same OX40/OX40L axis to inhibit MC degranulation, thus making this crosstalk bidirectional [62]. Furthermore, the binding between MC-derived histamine and histamine receptor type 1 promotes Th1 polarization, whereas its signaling through histamine receptor type 2 is able to restrain both Th1 and Th2 responses [63]. Conversely, it has been found that histamine can support the immunosuppressive microenvironment by recruiting Tregs [64].
Besides the direct anti-tumor effects discussed in the previous paragraph, MC-derived TNFα is also important for T cell activation [65]. The activation and proliferation of CD8+ T cells are also fostered by the production of osteopontin and the expression of co-stimulatory molecules by MCs [66]. Moreover, MCs can influence the homing of effector CD8+ T cells toward inflammation sites via the release of leukotriene B4 [67]. In a murine melanoma model, it has also been observed that TLR2-activated MCs can recruit NK cells through the secretion of high doses of CCL3 [36]. In addition to CCL3, other factors secreted by MCs, such as IL-4, IL-12, and TNFα, are able to activate NK cells [68,69], and the stimulation of TLR3 or TLR9 in MCs induces the secretion of IFN- γ by NK cells [70].
The conflicting results described above suggest that MCs and their mediators can have different roles depending on the stage of cancer, their peri- or intra-tumor localization, and crosstalk with other cells of the TME. Therefore, approaches aimed at molding MC functions could represent effective strategies for cancer immunotherapy.

5. Therapies Aimed at Targeting MCs in Cancer

The targeting of MC functions has been widely exploited for therapeutic purposes in allergic reactions and mastocytosis, as comprehensively reviewed elsewhere [71,72]. Given the focus of this review, here we discuss strategies that only specifically address the roles of MCs in cancer.
As MCs can exert tumor-promoting or suppressive activities depending on tumor type, localization, and signals received form the surrounding microenvironment, therapeutic strategies could either be directed to abrogate or prompt MC functions in the appropriate settings. Several approaches have been proposed (Figure 1): (1) targeting c-Kit signaling; (2) stabilizing MC degranulation; (3) triggering activating/inhibiting receptors; (4) modulating MC recruitment; (5) harnessing MC mediators; (6) adoptive transferring of MCs.

5.1. Targeting c-Kit Signaling

As c-Kit is crucial for MC development, survival, and activation, tyrosine kinase inhibitors, such as imatinib, nilotinib, or dasatinib, are efficiently employed to target MCs in mastocytosis, arthritis, or allergic responses [73,74,75,76]. However, so far, these drugs have displayed limited application in the context of restraining MC tumor-promoting functions [71]. We reported that imatinib can slightly control MC number and degranulation, consequently hampering prostate adenocarcinoma [44]. However, we found that imatinib treatment in the TRAMP prostate cancer model resulted in the outgrowth of the neuroendocrine prostate tumor variant as a drawback. This warns us about the mere targeting of MCs in prostate cancer, unless specific for adenocarcinoma-promoting functions of MCs or unless coupled with strategies designed to target the neuroendocrine histotype [44]. Another study employing the 4T1 breast cancer model showed that imatinib and the MC stabilizer cromolyn similarly induced peri-tumoral blood clotting and promoted tumor growth [77]. On the contrary, in a preclinical model of melanoma, it has recently been demonstrated that MC targeting with imatinib or sunitinib increased the therapeutic efficacy of anti-PD-1 [59].
Nevertheless, it is worth highlighting that imatinib, nilotinib, or dasatinib are not specific for c-Kit, as they also target other kinase receptors such as PDGFR, Src, and Abl kinase and thus could have off-target effects. To overcome these limitations, a monoclonal antibody targeting c-Kit, barzolvolimab, has been developed but tested so far only in the context of c-Kit-positive gastrointestinal tumors [71] or in chronic urticaria [78].

5.2. Stabilizing MC Degranulation

Agents that can restrain MC degranulation, such as cromolyn sodium salt or ketotifen, have been widely utilized in the treatment of allergic responses [79]; however, so far, their administration for cancer therapy has been confined to preclinical models [42,80,81]. A huge limitation of the use of “MC stabilizers” in cancer is the fact that they are not specific for MCs but can affect the release of mediators by other types of cells. In most cases, this could be an advantage if the tumor cell itself represents the target [82,83]. Nevertheless, as immune cells are also able to secrete a plethora of different mediators, it would be a disadvantage when the inhibition of T cell function occurs as a result [84].
A more tailored strategy relies on drugs targeting the intracellular components of the IgE/FcεRI signaling pathway, including Syk, PI3K p110δ isoform, and Bruton’s tyrosine kinase (BTK). The use of inhibitors of Syk and PI3K p110δ with the purpose to target MCs has so far only been investigated in the context of allergic disease [85]. Conversely, the BTK-specific drug ibrutinib was successfully tested in a model of pancreatic cancer, where it reduced tumor growth by reducing MC-dependent fibrosis [71]. However, these signal transducers are also not exclusively expressed by MCs; thus, in vivo effects could be altered by off-target activity and/or toxicity.
Recently, we discovered a new way to prevent MC degranulation by utilizing the antiepileptic drug levetiracetam [86], which is administered to avoid and mitigate seizures in patients [87]. Levetiracetam targets the synaptic vesicle protein 2A (SV2A) [88]—mainly present in neural and endocrine cells and involved in cell exocytosis [89]—and we found that this protein is also expressed by prostate cancer-infiltrating MCs [86]. We demonstrated that levetiracetam can inhibit MC degranulation and, in particular, the release of MMP9, thus restraining the pro-adenocarcinoma activity of MCs in TRAMP mice [86]. Furthermore, we also found the expression of SV2A in neuroendocrine prostate cancer and proved that, in this preclinical model, levetiracetam was also able to prevent neuroendocrine differentiation of adenocarcinoma after hormone therapy [86].

5.3. Triggering Other Activating/Inhibiting Receptors

Besides c-Kit and FcεRI, MCs have a plethora of different receptors that can regulate their functions in the TME; thus, these receptors could be possible targets of MC-specific anti-cancer therapies. Nevertheless, for most of them, including TSLPR, MRGPRX2, CD300, and SIGLECs, therapeutic targeting has been so far addressed only in the context of allergic diseases and extensively reviewed elsewhere [71,72,90,91].
Stimulation of TLRs in MCs can lead to specific cytokine secretion and, consequently, to the recruitment and activation of immune cells, eventually inhibiting tumor growth. Indeed, TLR agonists are currently being evaluated in cancer therapy to activate the immune response [92,93,94], and a few reports have also demonstrated the efficacy of polarizing MCs to orchestrate anti-tumor immunity. In the B16.F10 mouse melanoma model, the TLR2 agonist Pam3CSK4 induced MCs to release cytokines such as IL-6 and CCL3, which mediated a direct antiproliferative effect on tumor cells and the recruitment of NK and T cells, respectively [36]. Another study in melanoma models and patients showed a correlation between an LPS-related signature and responsiveness to anti-CTLA4 immunocheckpoint therapy. Further investigation showed that LPS triggered MCs to release CXCL10 with consequent T cell recruitment, also proving the superior anti-tumor activity of adding intratumoral LPS administration to anti-CTLA4 treatment in the preclinical setting [95]. TLR agonists given as adjuvants in the context of anti-tumor vaccinations yielded greater anti-tumor immune responses. This effect was found to be directly mediated by MCs in a mouse colon carcinoma model treated with a vaccine formulated with a recombinant CEA IgV N domain and the TLR3 ligand poly I:C [96]. Another work in the B16.F10 model also demonstrated that TLR7/9 stimulation with imiquimod can trigger CCL2 release by MCs to efficiently recruit plasmacytoid DCs at the tumor site, which, in turn, can directly kill tumor cells via TRAIL and granzyme B [97]. On the same line, it was also shown that MCs (the MC/9 cell line) pre-treated with imiquimod could increase the expression of DC costimulatory and activation molecules, thus augmenting the efficacy of a DC-based vaccine when tested in vivo in the B16-OVA melanoma model [98].
Even if TLR stimulation can efficiently reprogram MCs to orchestrate anti-tumor immunity, it should be kept in mind that this strategy could also have the opposite effect. Indeed, it was demonstrated that co-stimulation with SCF and TLR4 ligands induced the expression of VEGF, PDGF, and IL-10 in MCs, thus enhancing their tumor-promoting function in vitro and in vivo [99]. Therefore, further investigation is required to best dissect the effect of TLR triggering or inhibition to mold MCs toward anti-tumor functions.
Other possible strategies could focus on other receptors shown to be essential for the interaction of MCs with immune suppressive cells, including, for example, CD40L and OX40L, which we and others have identified as essential for the crosstalk between MCs and MDSCs [43,56], Breg [57], and Treg cells [61,62]. MCs can also directly suppress CD8+ T cell activation via PD-L1 on their surface [21]. Thus, MCs could be another target of immunocheckpoint blockade therapy in those tumors in which they are enriched. Notably, the inhibition of MC-associated PD-L1 resulted in increased T cell activation and efficient blunting of tumor growth in a gastric carcinoma model [100].

5.4. Modulating MC Recruitment

According to the pro- or anti-tumor functions exerted by MCs in different contexts, one conceivable therapeutic strategy would rely on inhibiting or incrementing their recruitment, respectively, by actioning chemotactic pathways. Besides regulating MC maturation, proliferation, and degranulation, both the SCF/c-Kit and the FcεRI signaling can also mediate MC migration [101]. Additionally, different types of tumor cells are known to produce SCF and actively recruit MCs [58,102]. Therefore, [103] inhibitors of c-Kit, BTK, Syk, and PI3K could also restrain MC trafficking in the tumor [104].
Furthermore, many other different molecules produced by tumor cells or by cells of the TME can induce MC chemotaxis, including CCL2 [105], CCL5 [106], CCL11 [106], CCL15 [102], CXCL12 [107], VEGF [80], FGF2 [108], osteopontin [109], and lipid mediators [103]. The blocking of these chemoattractants could represent a therapeutic strategy to impede MC recruitment and, consequently, their support to the tumor. In a murine model, it has been shown that the hindering of MC infiltration by treatment with the CXCL12 inhibitor AMD3100 restrains pancreatic cancer growth [110]. Similarly, in the TRAMP-C2 prostate cancer transplantable model, the blocking of FGF with the NSC12 molecule inhibited tumor growth and vascularization, and the effect was correlated to a reduced number of infiltrating MCs [108]. Additionally, in a different set of prostate cancer preclinical models, the pharmacologic inhibition of protein kinase D blunted the production of SCF, CCL5, and CCL11 and led to decreased MC migration and reduced tumor growth in vivo [106].
On the other hand, in those settings where MCs exert anti-tumor activity, it would be desirable to increase their number via the local delivery of specific chemoattractants or by stimulating the TME to produce such molecules. These approaches require further investigation.

5.5. Harnessing MC Mediators

As described in the previous sections, MCs can release several molecules with tumor-promoting or tumor-inhibiting functions. Thus, the last way to shape MC functions in cancer could rely on the modulation of these mediators.
Similar to many other cells in the TME, MCs secrete VEGF to promote angiogenesis. Several approved drugs against VEGF, including both monoclonal antibodies and small molecule inhibitors, are clinically available for cancer patients, as extensively reviewed elsewhere [111]. Moreover, tryptase is endowed with well-known pro-angiogenic functions [112]. Indeed, it has been described that the pharmacologic inhibition of tryptase with nafamostat or APC366 restrained tumor growth in preclinical models of pancreatic [113] and breast cancer [114], respectively. MCs also represent a major source of MMPs, which can degrade the extracellular matrix, favoring tumor growth. Nevertheless, the promising results obtained in preclinical models were not confirmed in clinical trials, where MMP inhibition was also accompanied by unexpected severe side effects [115].
Histamine can either promote or suppress anti-tumor immunity depending on tumor type and the surrounding TME [64,65,116]. Numerous studies have investigated the administration of histamine or of antagonists of its receptors in combination with immunotherapeutic strategies, showing promising results in different cancer types, including leukemia, melanoma, kidney, colorectal, and prostate cancer (reviewed in ref. [116]).
On the other hand, IL-6 and TNFα are the main molecular mediators of the anti-tumor functions of MCs [36,37,38,39], with the latter able to directly activate either apoptosis or necroptosis pathways in tumor cells [117,118]. Nonetheless, the systemic delivery of pro-inflammatory cytokines causes high toxicity [119,120,121]; thus, it is not suitable in a clinical setting. To overcome these limitations, several strategies to reduce systemic toxicity and implement the anti-tumor efficacy of TNFα have been investigated, including local administration [122] and specific delivery via nanoparticles [123,124] or targeting antibodies/peptides [125,126]. In this regard, targeting of TNFα to tumor neo-vasculature by conjugation with a CNGRC peptide that binds the CD13 aminopeptidase N [127] revealed a strong anti-tumor efficacy in several preclinical models [128], and it is being evaluated in clinical trials in melanoma and other solid tumors [129,130]. A novel frontier for localized TNFα delivery could also potentially be implemented via the direct injection of TNFα-producing MCs into the tumor. Indeed, the adoptive transfer of MCs could represent a new type of cellular immunotherapy, as described below.

5.6. Adoptive Transferring of MCs

Adoptive cell transfer exploiting autologous T cells or CAR-T cells is now mainstream for hematologic malignancies [131] and in widespread use for solid tumors [132]. Nevertheless, many other non-T immune cells can exert anti-tumor functions and are currently being investigated for cellular immunotherapy, including NK cells [133] and macrophages [134]. In this view, the in vivo adoptive transfer with MCs (also named reconstitution) has been exploited to demonstrate MC-specific tumor-promoting or tumor-inhibiting functions by means of reconstituting MC-deficient mice with bone marrow-derived MCs either proficient or deficient for selected molecules [55,57,135,136,137,138]. Nonetheless, it is conceivable to exploit the anti-tumor properties of MCs for cellular therapy against cancer. Such an approach should take into account the need to reprogram MCs in order to release anti-tumor mediators (i.e., TNFα) only when in contact with tumor cells to avoid the systemic delivery of unwanted molecules, allergic reactions, or other side effects. A first attempt in this direction was made by Fereyoduni et al., who exploited MCs pre-sensitized with HER2/neu-specific IgE to efficiently kill HER2/neu-expressing tumor cells both in vitro and in vivo in a breast cancer model. Indeed, they showed that the encounter with the cognate antigen on tumor cells unleashed the release of TNFα by IgE-presensitized MCs, eventually inducing apoptosis in tumor cells [139].
More investigations are required to best exploit the potential of MCs for adoptive cell therapies in cancer and also to evaluate whether it is possible to engineer MCs before in vivo transfer, with the aim of increasing and/or restraining the production of anti- or pro-tumor mediators, respectively [140]. Another strategy worthy of study would rely on exploiting defined activating receptors (i.e., TLRs) on the surface of MCs instead of IgE/FcεRI activation in order to trigger the release of desired mediators when cognate ligands are expressed by tumor cells or the surrounding TME.

6. Clinical Trials Aimed at Targeting MCs in Cancer Patients

Despite the intense preclinical investigation, the number of clinical trials evaluating MCs in cancer is scarce. A search on clinicaltrials.gov using #cancer and #mast cells as keywords retrieved 130 results, which were reduced to 4 after filtering to exclude trials in patients with mastocytosis and mast cell leukemias. In particular, the NCT05076682 phase II trial will test the combination of cromolyn and anti-PD1 antibodies in order to see whether the inhibition of MCs can synergize with immunocheckpoint blockade in triple-negative breast cancer patients. This trial is still in the recruiting phase and is the only one among the few identified that aims to actively target MCs with specific therapy. Two other trials in lung cancer and basal cell carcinoma (NCT02161523 and NCT02576769) list MCs among the outcomes of the study. The first will monitor MC activation/phenotype after co-culture with patient-derived fibroblasts, whereas the second is an observational study that will analyze the number of MCs in the TME. Finally, we include in the list a phase I trial that tested a therapeutic antibody directed against the folate receptor in different solid tumors, including tumors in the kidney, endometrium, lung, breast, bladder, colon, and pancreas. As the antibody used for therapy regarding the IgE isotype, it is conceivable to hypothesize the induction of some degree of MC activation. Yet, MCs are not listed among the cells that will be evaluated as readout after therapy.

7. Limitations

This review aimed to describe the different roles of MCs in cancer and the possible ways to exploit them for cancer immunotherapy. For this reason, we did not cover additional aspects of MC biology that are already known to be relevant in other physiological or pathological settings. For example, we did not mention that, similar to neutrophils, MCs are able to protrude extracellular traps during pathogenic infections or that MCs can be involved in fibrosis and heart failure. Further investigation is required to determine whether such MC functions are also conserved in the TME.
Furthermore, whereas different subtypes of human and murine MCs can be defined by their protease expression, their actual identification within tumor tissues is biased by some technical limitations. Thus, the relevance of MC subsets in determining pro- or anti- tumor effects is underexplored. It would be highly valuable to exploit new methodologies that can discriminate these subpopulations in the TME by evaluating their chymase/tryptase levels or new biomarkers. This would provide crucial insights into the significance of each population within specific cancer contexts.

8. Conclusions and Future Directions

MCs are crucial players in the TME; thus, they are potential targets for anti-tumor immunotherapy. However, extensive investigation would be required before being able to efficiently apply MC-directed therapies for the benefit of cancer patients. Indeed, given the multifaceted roles of MCs in different contexts, approaches should aim to either inhibit or foster their activity according to the specific context, contributing to the notion of “MC-guided” personalized medicine. Moreover, tailored strategies that only target specific MC functions should be preferred to avoid off-target effects or paradoxical effects, as shown for prostate cancer, where MCs show split mechanisms to either promote or prevent different tumor histotypes. The harnessing of MC functions should also be tested in combination with other (immuno-) therapeutic approaches to evaluate possible synergisms against tumor growth. Finally, evidence collected in murine models should be extensively validated in human settings to ultimately prove the clinical relevance and effective benefit of MC-based immunotherapy in cancer patients.

Author Contributions

Writing—original draft preparation, E.J. and R.S.; writing—review and editing, E.J. and R.S.; supervision, E.J.; funding acquisition, E.J. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Grants from Italian Ministry of Health (GR-2016-02362484 to E.J. and “Ricerca Corrente Funds”) and Associazione Italiana per la Ricerca sul Cancro (AIRC; Investigator Grant 25854 to E.J.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Acknowledgments

We thank Daniele Lecis from Fondazione IRCCS Istituto Nazionale dei Tumori for revising the English language of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [Green Version]
  3. Galli, S.J.; Gaudenzio, N.; Tsai, M. Mast Cells in Inflammation and Disease: Recent Progress and Ongoing Concerns. Annu. Rev. Immunol. 2020, 38, 49–77. [Google Scholar] [CrossRef] [PubMed]
  4. Okayama, Y.; Kawakami, T. Development, migration, and survival of mast cells. Immunol. Res. 2006, 34, 97–115. [Google Scholar] [CrossRef] [PubMed]
  5. Nilsson, G.; Butterfield, J.H.; Nilsson, K.; Siegbahn, A. Stem cell factor is a chemotactic factor for human mast cells. J. Immunol. 1994, 153, 3717–3723. [Google Scholar] [CrossRef] [PubMed]
  6. Juremalm, M.; Olsson, N.; Nilsson, G. Selective CCL5/RANTES-induced mast cell migration through interactions with chemokine receptors CCR1 and CCR4. Biochem. Biophys. Res. Commun. 2002, 297, 480–485. [Google Scholar] [CrossRef]
  7. Moon, T.C.; Befus, A.D.; Kulka, M. Mast cell mediators: Their differential release and the secretory pathways involved. Front. Immunol. 2014, 5, 569. [Google Scholar] [CrossRef] [Green Version]
  8. Krystel-Whittemore, M.; Dileepan, K.N.; Wood, J.G. Mast Cell: A Multi-Functional Master Cell. Front. Immunol. 2015, 6, 620. [Google Scholar] [CrossRef] [Green Version]
  9. Welle, M. Development, significance, and heterogeneity of mast cells with particular regard to the mast cell-specific proteases chymase and tryptase. J. Leukoc. Biol. 1997, 61, 233–245. [Google Scholar] [CrossRef]
  10. Kalesnikoff, J.; Galli, S.J. New developments in mast cell biology. Nat. Immunol. 2008, 9, 1215–1223. [Google Scholar] [CrossRef] [Green Version]
  11. Galli, S.J.; Tsai, M. Mast cells in allergy and infection: Versatile effector and regulatory cells in innate and adaptive immunity. Eur. J. Immunol. 2010, 40, 1843–1851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Amin, K. The role of mast cells in allergic inflammation. Respir. Med. 2012, 106, 9–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Yokoi, H.; Choi, O.H.; Hubbard, W.; Lee, H.S.; Canning, B.J.; Lee, H.H.; Ryu, S.D.; von Gunten, S.; Bickel, C.A.; Hudson, S.A.; et al. Inhibition of FcepsilonRI-dependent mediator release and calcium flux from human mast cells by sialic acid-binding immunoglobulin-like lectin 8 engagement. J. Allergy Clin. Immunol. 2008, 121, 499–505.e491. [Google Scholar] [CrossRef]
  14. Subramanian, H.; Gupta, K.; Ali, H. Roles of Mas-related G protein-coupled receptor X2 on mast cell-mediated host defense, pseudoallergic drug reactions, and chronic inflammatory diseases. J. Allergy Clin. Immunol. 2016, 138, 700–710. [Google Scholar] [CrossRef] [Green Version]
  15. Bulfone-Paus, S.; Nilsson, G.; Draber, P.; Blank, U.; Levi-Schaffer, F. Positive and Negative Signals in Mast Cell Activation. Trends Immunol. 2017, 38, 657–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sandig, H.; Bulfone-Paus, S. TLR signaling in mast cells: Common and unique features. Front. Immunol. 2012, 3, 185. [Google Scholar] [CrossRef] [Green Version]
  17. Lee, C.C.; Avalos, A.M.; Ploegh, H.L. Accessory molecules for Toll-like receptors and their function. Nat. Rev. Immunol. 2012, 12, 168–179. [Google Scholar] [CrossRef] [Green Version]
  18. Wieland, C.W.; Florquin, S.; Maris, N.A.; Hoebe, K.; Beutler, B.; Takeda, K.; Akira, S.; van der Poll, T. The MyD88-dependent, but not the MyD88-independent, pathway of TLR4 signaling is important in clearing nontypeable haemophilus influenzae from the mouse lung. J. Immunol. 2005, 175, 6042–6049. [Google Scholar] [CrossRef] [Green Version]
  19. Liew, F.Y.; Pitman, N.I.; McInnes, I.B. Disease-associated functions of IL-33: The new kid in the IL-1 family. Nat. Rev. Immunol. 2010, 10, 103–110. [Google Scholar] [CrossRef]
  20. Sebastian, K.; Borowski, A.; Kuepper, M.; Friedrich, K. Signal transduction around thymic stromal lymphopoietin (TSLP) in atopic asthma. Cell Commun. Signal. 2008, 6, 5. [Google Scholar] [CrossRef] [Green Version]
  21. Hirano, T.; Honda, T.; Kanameishi, S.; Honda, Y.; Egawa, G.; Kitoh, A.; Nakajima, S.; Otsuka, A.; Nomura, T.; Dainichi, T.; et al. PD-L1 on mast cells suppresses effector CD8(+) T-cell activation in the skin in murine contact hypersensitivity. J. Allergy Clin. Immunol. 2021, 148, 563–573.e7. [Google Scholar] [CrossRef] [PubMed]
  22. Bonamichi-Santos, R.; Aun, M.V.; Kalil, J.; Castells, M.C.; Giavina-Bianchi, P. PD-L1 Blockade During Allergen Sensitization Inhibits the Synthesis of Specific Antibodies and Decreases Mast Cell Activation in a Murine Model of Active Cutaneous Anaphylaxis. Front. Immunol. 2021, 12, 655958. [Google Scholar] [CrossRef] [PubMed]
  23. Varricchi, G.; Galdiero, M.R.; Loffredo, S.; Marone, G.; Iannone, R.; Marone, G.; Granata, F. Are Mast Cells MASTers in Cancer? Front. Immunol. 2017, 8, 424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Strouch, M.J.; Cheon, E.C.; Salabat, M.R.; Krantz, S.B.; Gounaris, E.; Melstrom, L.G.; Dangi-Garimella, S.; Wang, E.; Munshi, H.G.; Khazaie, K.; et al. Crosstalk between mast cells and pancreatic cancer cells contributes to pancreatic tumor progression. Clin. Cancer Res. 2010, 16, 2257–2265. [Google Scholar] [CrossRef] [Green Version]
  25. Soucek, L.; Lawlor, E.R.; Soto, D.; Shchors, K.; Swigart, L.B.; Evan, G.I. Mast cells are required for angiogenesis and macroscopic expansion of Myc-induced pancreatic islet tumors. Nat. Med. 2007, 13, 1211–1218. [Google Scholar] [CrossRef] [PubMed]
  26. Rao, Q.; Chen, Y.; Yeh, C.R.; Ding, J.; Li, L.; Chang, C.; Yeh, S. Recruited mast cells in the tumor microenvironment enhance bladder cancer metastasis via modulation of ERbeta/CCL2/CCR2 EMT/MMP9 signals. Oncotarget 2016, 7, 7842–7855. [Google Scholar] [CrossRef] [Green Version]
  27. 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]
  28. Visciano, C.; Prevete, N.; Liotti, F.; Marone, G. Tumor-Associated Mast Cells in Thyroid Cancer. Int. J. Endocrinol. 2015, 2015, 705169. [Google Scholar] [CrossRef] [Green Version]
  29. Johnson, C.; Huynh, V.; Hargrove, L.; Kennedy, L.; Graf-Eaton, A.; Owens, J.; Trzeciakowski, J.P.; Hodges, K.; DeMorrow, S.; Han, Y.; et al. Inhibition of Mast Cell-Derived Histamine Decreases Human Cholangiocarcinoma Growth and Differentiation via c-Kit/Stem Cell Factor-Dependent Signaling. Am. J. Pathol. 2016, 186, 123–133. [Google Scholar] [CrossRef]
  30. Gounaris, E.; Erdman, S.E.; Restaino, C.; Gurish, M.F.; Friend, D.S.; Gounari, F.; Lee, D.M.; Zhang, G.; Glickman, J.N.; Shin, K.; et al. Mast cells are an essential hematopoietic component for polyp development. Proc. Natl. Acad. Sci. USA 2007, 104, 19977–19982. [Google Scholar] [CrossRef] [Green Version]
  31. Rigoni, A.; Bongiovanni, L.; Burocchi, A.; Sangaletti, S.; Danelli, L.; Guarnotta, C.; Lewis, A.; Rizzo, A.; Silver, A.R.; Tripodo, C.; et al. Mast Cells Infiltrating Inflamed or Transformed Gut Alternatively Sustain Mucosal Healing or Tumor Growth. Cancer Res. 2015, 75, 3760–3770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tomita, M.; Matsuzaki, Y.; Onitsuka, T. Correlation between mast cells and survival rates in patients with pulmonary adenocarcinoma. Lung Cancer 1999, 26, 103–108. [Google Scholar] [CrossRef] [PubMed]
  33. Welsh, T.J.; Green, R.H.; Richardson, D.; Waller, D.A.; O’Byrne, K.J.; Bradding, P. Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer. J. Clin. Oncol. 2005, 23, 8959–8967. [Google Scholar] [CrossRef]
  34. Hedstrom, G.; Berglund, M.; Molin, D.; Fischer, M.; Nilsson, G.; Thunberg, U.; Book, M.; Sundstrom, C.; Rosenquist, R.; Roos, G.; et al. Mast cell infiltration is a favourable prognostic factor in diffuse large B-cell lymphoma. Br. J. Haematol. 2007, 138, 68–71. [Google Scholar] [CrossRef] [PubMed]
  35. Sinnamon, M.J.; Carter, K.J.; Sims, L.P.; Lafleur, B.; Fingleton, B.; Matrisian, L.M. A protective role of mast cells in intestinal tumorigenesis. Carcinogenesis 2008, 29, 880–886. [Google Scholar] [CrossRef] [Green Version]
  36. 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] [PubMed] [Green Version]
  37. Shikotra, A.; Ohri, C.M.; Green, R.H.; Waller, D.A.; Bradding, P. Mast cell phenotype, TNFalpha expression and degranulation status in non-small cell lung cancer. Sci. Rep. 2016, 6, 38352. [Google Scholar] [CrossRef] [Green Version]
  38. Benyon, R.C.; Bissonnette, E.Y.; Befus, A.D. Tumor necrosis factor-alpha dependent cytotoxicity of human skin mast cells is enhanced by anti-IgE antibodies. J. Immunol. 1991, 147, 2253–2258. [Google Scholar] [CrossRef]
  39. Cheng, S.; Li, Z.; Gao, R.; Xing, B.; Gao, Y.; Yang, Y.; Qin, S.; Zhang, L.; Ouyang, H.; Du, P.; et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell 2021, 184, 792–809.e23. [Google Scholar] [CrossRef]
  40. Majorini, M.T.; Cancila, V.; Rigoni, A.; Botti, L.; Dugo, M.; Triulzi, T.; De Cecco, L.; Fontanella, E.; Jachetti, E.; Tagliabue, E.; et al. Infiltrating Mast Cell-Mediated Stimulation of Estrogen Receptor Activity in Breast Cancer Cells Promotes the Luminal Phenotype. Cancer Res. 2020, 80, 2311–2324. [Google Scholar] [CrossRef] [Green Version]
  41. Majorini, M.T.; Colombo, M.P.; Lecis, D. Few, but Efficient: The Role of Mast Cells in Breast Cancer and Other Solid Tumors. Cancer Res. 2022, 82, 1439–1447. [Google Scholar] [CrossRef] [PubMed]
  42. Pittoni, P.; Tripodo, C.; Piconese, S.; Mauri, G.; Parenza, M.; Rigoni, A.; Sangaletti, S.; Colombo, M.P. Mast cell targeting hampers prostate adenocarcinoma development but promotes the occurrence of highly malignant neuroendocrine cancers. Cancer Res. 2011, 71, 5987–5997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Jachetti, E.; Cancila, V.; Rigoni, A.; Bongiovanni, L.; Cappetti, B.; Belmonte, B.; Enriquez, C.; Casalini, P.; Ostano, P.; Frossi, B.; et al. Cross-Talk between Myeloid-Derived Suppressor Cells and Mast Cells Mediates Tumor-Specific Immunosuppression in Prostate Cancer. Cancer Immunol. Res. 2018, 6, 552–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jachetti, E.; Rigoni, A.; Bongiovanni, L.; Arioli, I.; Botti, L.; Parenza, M.; Cancila, V.; Chiodoni, C.; Festinese, F.; Bellone, M.; et al. Imatinib Spares cKit-Expressing Prostate Neuroendocrine Tumors, whereas Kills Seminal Vesicle Epithelial-Stromal Tumors by Targeting PDGFR-beta. Mol. Cancer Ther. 2017, 16, 365–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Johansson, A.; Rudolfsson, S.; Hammarsten, P.; Halin, S.; Pietras, K.; Jones, J.; Stattin, P.; Egevad, L.; Granfors, T.; Wikstrom, P.; et al. Mast cells are novel independent prognostic markers in prostate cancer and represent a target for therapy. Am. J. Pathol. 2010, 177, 1031–1041. [Google Scholar] [CrossRef] [PubMed]
  46. Fleischmann, A.; Schlomm, T.; Kollermann, J.; Sekulic, N.; Huland, H.; Mirlacher, M.; Sauter, G.; Simon, R.; Erbersdobler, A. Immunological microenvironment in prostate cancer: High mast cell densities are associated with favorable tumor characteristics and good prognosis. Prostate 2009, 69, 976–981. [Google Scholar] [CrossRef]
  47. Hempel Sullivan, H.; Maynard, J.P.; Heaphy, C.M.; Lu, J.; De Marzo, A.M.; Lotan, T.L.; Joshu, C.E.; Sfanos, K.S. Differential mast cell phenotypes in benign versus cancer tissues and prostate cancer oncologic outcomes. J. Pathol. 2021, 253, 415–426. [Google Scholar] [CrossRef]
  48. Hempel Sullivan, H.; Heaphy, C.M.; Kulac, I.; Cuka, N.; Lu, J.; Barber, J.R.; De Marzo, A.M.; Lotan, T.L.; Joshu, C.E.; Sfanos, K.S. High Extratumoral Mast Cell Counts Are Associated with a Higher Risk of Adverse Prostate Cancer Outcomes. Cancer Epidemiol. Biomark. Prev. 2020, 29, 668–675. [Google Scholar] [CrossRef]
  49. Watanabe, S.; Miyata, Y.; Matsuo, T.; Mochizuki, Y.; Nishikido, M.; Hayashi, T.; Sakai, H. High density of tryptase-positive mast cells in patients with renal cell carcinoma on hemodialysis: Correlation with expression of stem cell factor and protease activated receptor-2. Hum. Pathol. 2012, 43, 888–897. [Google Scholar] [CrossRef]
  50. Cherdantseva, T.M.; Bobrov, I.P.; Avdalyan, A.M.; Klimachev, V.V.; Kazartsev, A.V.; Kryuchkova, N.G.; Klimachev, I.V.; Myadelets, M.N.; Lepilov, A.V.; Lushnikova, E.L.; et al. Mast Cells in Renal Cancer: Clinical Morphological Correlations and Prognosis. Bull. Exp. Biol. Med. 2017, 163, 801–804. [Google Scholar] [CrossRef]
  51. Fu, H.; Zhu, Y.; Wang, Y.; Liu, Z.; Zhang, J.; Wang, Z.; Xie, H.; Dai, B.; Xu, J.; Ye, D. Tumor Infiltrating Mast Cells (TIMs) Confers a Marked Survival Advantage in Nonmetastatic Clear-Cell Renal Cell Carcinoma. Ann. Surg. Oncol. 2017, 24, 1435–1442. [Google Scholar] [CrossRef]
  52. Feng, T.Y.; Azar, F.N.; Dreger, S.A.; Rosean, C.B.; McGinty, M.T.; Putelo, A.M.; Kolli, S.H.; Carey, M.A.; Greenfield, S.; Fowler, W.J.; et al. Reciprocal Interactions Between the Gut Microbiome and Mammary Tissue Mast Cells Promote Metastatic Dissemination of HR+ Breast Tumors. Cancer Immunol. Res. 2022, 10, 1309–1325. [Google Scholar] [CrossRef]
  53. Yang, Z.; Zhang, B.; Li, D.; Lv, M.; Huang, C.; Shen, G.X.; Huang, B. Mast cells mobilize myeloid-derived suppressor cells and Treg cells in tumor microenvironment via IL-17 pathway in murine hepatocarcinoma model. PLoS ONE 2010, 5, e8922. [Google Scholar] [CrossRef] [PubMed]
  54. Cheon, E.C.; Khazaie, K.; Khan, M.W.; Strouch, M.J.; Krantz, S.B.; Phillips, J.; Blatner, N.R.; Hix, L.M.; Zhang, M.; Dennis, K.L.; et al. Mast cell 5-lipoxygenase activity promotes intestinal polyposis in APCDelta468 mice. Cancer Res. 2011, 71, 1627–1636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Saleem, S.J.; Martin, R.K.; Morales, J.K.; Sturgill, J.L.; Gibb, D.R.; Graham, L.; Bear, H.D.; Manjili, M.H.; Ryan, J.J.; Conrad, D.H. Cutting edge: Mast cells critically augment myeloid-derived suppressor cell activity. J. Immunol. 2012, 189, 511–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Danelli, L.; Frossi, B.; Gri, G.; Mion, F.; Guarnotta, C.; Bongiovanni, L.; Tripodo, C.; Mariuzzi, L.; Marzinotto, S.; Rigoni, A.; et al. Mast cells boost myeloid-derived suppressor cell activity and contribute to the development of tumor-favoring microenvironment. Cancer Immunol. Res. 2015, 3, 85–95. [Google Scholar] [CrossRef] [Green Version]
  57. Mion, F.; D’Inca, F.; Danelli, L.; Toffoletto, B.; Guarnotta, C.; Frossi, B.; Burocchi, A.; Rigoni, A.; Gerdes, N.; Lutgens, E.; et al. Mast cells control the expansion and differentiation of IL-10-competent B cells. J. Immunol. 2014, 193, 4568–4579. [Google Scholar] [CrossRef] [Green Version]
  58. Huang, B.; Lei, Z.; Zhang, G.M.; Li, D.; Song, C.; Li, B.; Liu, Y.; Yuan, Y.; Unkeless, J.; Xiong, H.; et al. SCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment. Blood 2008, 112, 1269–1279. [Google Scholar] [CrossRef]
  59. 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]
  60. Blatner, N.R.; Bonertz, A.; Beckhove, P.; Cheon, E.C.; Krantz, S.B.; Strouch, M.; Weitz, J.; Koch, M.; Halverson, A.L.; Bentrem, D.J.; et al. In colorectal cancer mast cells contribute to systemic regulatory T-cell dysfunction. Proc. Natl. Acad. Sci. USA 2010, 107, 6430–6435. [Google Scholar] [CrossRef] [Green Version]
  61. Piconese, S.; Gri, G.; Tripodo, C.; Musio, S.; Gorzanelli, A.; Frossi, B.; Pedotti, R.; Pucillo, C.E.; Colombo, M.P. Mast cells counteract regulatory T-cell suppression through interleukin-6 and OX40/OX40L axis toward Th17-cell differentiation. Blood 2009, 114, 2639–2648. [Google Scholar] [CrossRef] [PubMed]
  62. Gri, G.; Piconese, S.; Frossi, B.; Manfroi, V.; Merluzzi, S.; Tripodo, C.; Viola, A.; Odom, S.; Rivera, J.; Colombo, M.P.; et al. CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction. Immunity 2008, 29, 771–781. [Google Scholar] [CrossRef] [Green Version]
  63. Jutel, M.; Watanabe, T.; Klunker, S.; Akdis, M.; Thomet, O.A.; Malolepszy, J.; Zak-Nejmark, T.; Koga, R.; Kobayashi, T.; Blaser, K.; et al. Histamine regulates T-cell and antibody responses by differential expression of H1 and H2 receptors. Nature 2001, 413, 420–425. [Google Scholar] [CrossRef]
  64. Morgan, R.K.; McAllister, B.; Cross, L.; Green, D.S.; Kornfeld, H.; Center, D.M.; Cruikshank, W.W. Histamine 4 receptor activation induces recruitment of FoxP3+ T cells and inhibits allergic asthma in a murine model. J. Immunol. 2007, 178, 8081–8089. [Google Scholar] [CrossRef] [Green Version]
  65. Nakae, S.; Suto, H.; Iikura, M.; Kakurai, M.; Sedgwick, J.D.; Tsai, M.; Galli, S.J. Mast cells enhance T cell activation: Importance of mast cell costimulatory molecules and secreted TNF. J. Immunol. 2006, 176, 2238–2248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Stelekati, E.; Bahri, R.; D’Orlando, O.; Orinska, Z.; Mittrucker, H.W.; Langenhaun, R.; Glatzel, M.; Bollinger, A.; Paus, R.; Bulfone-Paus, S. Mast cell-mediated antigen presentation regulates CD8+ T cell effector functions. Immunity 2009, 31, 665–676. [Google Scholar] [CrossRef]
  67. Ott, V.L.; Cambier, J.C.; Kappler, J.; Marrack, P.; Swanson, B.J. Mast cell-dependent migration of effector CD8+ T cells through production of leukotriene B4. Nat. Immunol. 2003, 4, 974–981. [Google Scholar] [CrossRef]
  68. Morris, S.C.; Orekhova, T.; Meadows, M.J.; Heidorn, S.M.; Yang, J.; Finkelman, F.D. IL-4 induces in vivo production of IFN-gamma by NK and NKT cells. J. Immunol. 2006, 176, 5299–5305. [Google Scholar] [CrossRef] [Green Version]
  69. Orange, J.S.; Biron, C.A. Characterization of early IL-12, IFN-alphabeta, and TNF effects on antiviral state and NK cell responses during murine cytomegalovirus infection. J. Immunol. 1996, 156, 4746–4756. [Google Scholar] [CrossRef]
  70. Vosskuhl, K.; Greten, T.F.; Manns, M.P.; Korangy, F.; Wedemeyer, J. Lipopolysaccharide-mediated mast cell activation induces IFN-gamma secretion by NK cells. J. Immunol. 2010, 185, 119–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Harvima, I.T.; Levi-Schaffer, F.; Draber, P.; Friedman, S.; Polakovicova, I.; Gibbs, B.F.; Blank, U.; Nilsson, G.; Maurer, M. Molecular targets on mast cells and basophils for novel therapies. J. Allergy Clin. Immunol. 2014, 134, 530–544. [Google Scholar] [CrossRef] [PubMed]
  72. Rische, C.H.; Thames, A.N.; Krier-Burris, R.A.; O’Sullivan, J.A.; Bochner, B.S.; Scott, E.A. Drug delivery targets and strategies to address mast cell diseases. Expert. Opin. Drug Deliv. 2023, 20, 205–222. [Google Scholar] [CrossRef] [PubMed]
  73. Vega-Ruiz, A.; Cortes, J.E.; Sever, M.; Manshouri, T.; Quintas-Cardama, A.; Luthra, R.; Kantarjian, H.M.; Verstovsek, S. Phase II study of imatinib mesylate as therapy for patients with systemic mastocytosis. Leuk. Res. 2009, 33, 1481–1484. [Google Scholar] [CrossRef] [Green Version]
  74. Juurikivi, A.; Sandler, C.; Lindstedt, K.A.; Kovanen, P.T.; Juutilainen, T.; Leskinen, M.J.; Maki, T.; Eklund, K.K. Inhibition of c-kit tyrosine kinase by imatinib mesylate induces apoptosis in mast cells in rheumatoid synovia: A potential approach to the treatment of arthritis. Ann. Rheum. Dis. 2005, 64, 1126–1131. [Google Scholar] [CrossRef] [Green Version]
  75. El-Agamy, D.S. Anti-allergic effects of nilotinib on mast cell-mediated anaphylaxis like reactions. Eur. J. Pharmacol. 2012, 680, 115–121. [Google Scholar] [CrossRef] [PubMed]
  76. Min, H.K.; Kim, S.H.; Won, J.Y.; Kim, K.W.; Lee, J.Y.; Lee, S.H.; Kim, H.R. Dasatinib, a selective tyrosine kinase inhibitor, prevents joint destruction in rheumatoid arthritis animal model. Int. J. Rheum. Dis. 2023, 26, 718–726. [Google Scholar] [CrossRef]
  77. Samoszuk, M.; Corwin, M.A. Mast cell inhibitor cromolyn increases blood clotting and hypoxia in murine breast cancer. Int. J. Cancer 2003, 107, 159–163. [Google Scholar] [CrossRef] [PubMed]
  78. Terhorst-Molawi, D.; Hawro, T.; Grekowitz, E.; Kiefer, L.; Merchant, K.; Alvarado, D.; Thomas, L.J.; Hawthorne, T.; Crowley, E.; Heath-Chiozzi, M.; et al. Anti-KIT antibody, barzolvolimab, reduces skin mast cells and disease activity in chronic inducible urticaria. Allergy 2022, 78, 1269–1279. [Google Scholar] [CrossRef]
  79. Pacharn, P.; Vichyanond, P. Immunomodulators for conjunctivitis. Curr. Opin. Allergy Clin. Immunol. 2013, 13, 550–557. [Google Scholar] [CrossRef]
  80. Melillo, R.M.; Guarino, V.; Avilla, E.; Galdiero, M.R.; Liotti, F.; Prevete, N.; Rossi, F.W.; Basolo, F.; Ugolini, C.; de Paulis, A.; et al. Mast cells have a protumorigenic role in human thyroid cancer. Oncogene 2010, 29, 6203–6215. [Google Scholar] [CrossRef] [Green Version]
  81. Eissmann, M.F.; Dijkstra, C.; Jarnicki, A.; Phesse, T.; Brunnberg, J.; Poh, A.R.; Etemadi, N.; Tsantikos, E.; Thiem, S.; Huntington, N.D.; et al. IL-33-mediated mast cell activation promotes gastric cancer through macrophage mobilization. Nat. Commun. 2019, 10, 2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Khan, F.M.; Saleh, E.; Alawadhi, H.; Harati, R.; Zimmermann, W.H.; El-Awady, R. Inhibition of exosome release by ketotifen enhances sensitivity of cancer cells to doxorubicin. Cancer Biol. Ther. 2018, 19, 25–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Arumugam, T.; Ramachandran, V.; Logsdon, C.D. Effect of cromolyn on S100P interactions with RAGE and pancreatic cancer growth and invasion in mouse models. J. Natl. Cancer Inst. 2006, 98, 1806–1818. [Google Scholar] [CrossRef]
  84. Ito, J.; Asano, K.; Tryka, E.; Kanai, K.; Yamamoto, S.; Hisamitsu, T.; Suzaki, H. Suppressive effects of co-stimulatory molecule expressions on mouse splenocytes by anti-allergic agents in vitro. Mediators Inflamm. 2000, 9, 69–75. [Google Scholar] [CrossRef] [Green Version]
  85. Horak, F.; Puri, K.D.; Steiner, B.H.; Holes, L.; Xing, G.; Zieglmayer, P.; Zieglmayer, R.; Lemell, P.; Yu, A. Randomized phase 1 study of the phosphatidylinositol 3-kinase delta inhibitor idelalisib in patients with allergic rhinitis. J. Allergy Clin. Immunol. 2016, 137, 1733–1741. [Google Scholar] [CrossRef] [Green Version]
  86. Sulsenti, R.; Frossi, B.; Bongiovanni, L.; Cancila, V.; Ostano, P.; Fischetti, I.; Enriquez, C.; Guana, F.; Chiorino, G.; Tripodo, C.; et al. Repurposing of the Antiepileptic Drug Levetiracetam to Restrain Neuroendocrine Prostate Cancer and Inhibit Mast Cell Support to Adenocarcinoma. Front. Immunol. 2021, 12, 622001. [Google Scholar] [CrossRef] [PubMed]
  87. Sills, G.J.; Rogawski, M.A. Mechanisms of action of currently used antiseizure drugs. Neuropharmacology 2020, 168, 107966. [Google Scholar] [CrossRef]
  88. Lynch, B.A.; Lambeng, N.; Nocka, K.; Kensel-Hammes, P.; Bajjalieh, S.M.; Matagne, A.; Fuks, B. The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc. Natl. Acad. Sci. USA 2004, 101, 9861–9866. [Google Scholar] [CrossRef] [Green Version]
  89. Portela-Gomes, G.M.; Lukinius, A.; Grimelius, L. Synaptic vesicle protein 2, A new neuroendocrine cell marker. Am. J. Pathol. 2000, 157, 1299–1309. [Google Scholar] [CrossRef] [Green Version]
  90. Lichterman, J.N.; Reddy, S.M. Mast Cells: A New Frontier for Cancer Immunotherapy. Cells 2021, 10, 1270. [Google Scholar] [CrossRef]
  91. Kiwamoto, T.; Kawasaki, N.; Paulson, J.C.; Bochner, B.S. Siglec-8 as a drugable target to treat eosinophil and mast cell-associated conditions. Pharmacol. Ther. 2012, 135, 327–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Aznar, M.A.; Planelles, L.; Perez-Olivares, M.; Molina, C.; Garasa, S.; Etxeberria, I.; Perez, G.; Rodriguez, I.; Bolanos, E.; Lopez-Casas, P.; et al. Immunotherapeutic effects of intratumoral nanoplexed poly I:C. J. Immunother. Cancer 2019, 7, 116. [Google Scholar] [CrossRef] [Green Version]
  93. Sato-Kaneko, F.; Yao, S.; Ahmadi, A.; Zhang, S.S.; Hosoya, T.; Kaneda, M.M.; Varner, J.A.; Pu, M.; Messer, K.S.; Guiducci, C.; et al. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight 2017, 2, e93397. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, S.; Campos, J.; Gallotta, M.; Gong, M.; Crain, C.; Naik, E.; Coffman, R.L.; Guiducci, C. Intratumoral injection of a CpG oligonucleotide reverts resistance to PD-1 blockade by expanding multifunctional CD8+ T cells. Proc. Natl. Acad. Sci. USA 2016, 113, E7240–E7249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kaesler, S.; Wolbing, F.; Kempf, W.E.; Skabytska, Y.; Koberle, M.; Volz, T.; Sinnberg, T.; Amaral, T.; Mockel, S.; Yazdi, A.; et al. Targeting tumor-resident mast cells for effective anti-melanoma immune responses. JCI Insight 2019, 4, e125057. [Google Scholar] [CrossRef]
  96. 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]
  97. Drobits, B.; Holcmann, M.; Amberg, N.; Swiecki, M.; Grundtner, R.; Hammer, M.; Colonna, M.; Sibilia, M. Imiquimod clears tumors in mice independent of adaptive immunity by converting pDCs into tumor-killing effector cells. J. Clin. Investig. 2012, 122, 575–585. [Google Scholar] [CrossRef]
  98. Ren, S.; Wang, Q.; Zhang, Y.; Song, Y.; Dong, X.; Zhang, W.; Qin, X.; Liu, M.; Yu, T. Imiquimod enhances the potency of an exogenous BM-DC based vaccine against mouse melanoma. Int. Immunopharmacol. 2018, 64, 69–77. [Google Scholar] [CrossRef]
  99. Wei, J.J.; Song, C.W.; Sun, L.C.; Yuan, Y.; Li, D.; Yan, B.; Liao, S.J.; Zhu, J.H.; Wang, Q.; Zhang, G.M.; et al. SCF and TLR4 ligand cooperate to augment the tumor-promoting potential of mast cells. Cancer Immunol. Immunother. 2012, 61, 303–312. [Google Scholar] [CrossRef]
  100. 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] [Green Version]
  101. 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] [PubMed] [Green Version]
  102. Yu, Y.; Blokhuis, B.; Derks, Y.; Kumari, S.; Garssen, J.; Redegeld, F. Human mast cells promote colon cancer growth via bidirectional crosstalk: Studies in 2D and 3D coculture models. Oncoimmunology 2018, 7, e1504729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. 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]
  104. Burks, C.A.; Rhodes, S.D.; Bessler, W.K.; Chen, S.; Smith, A.; Gehlhausen, J.R.; Hawley, E.T.; Jiang, L.; Li, X.; Yuan, J.; et al. Ketotifen Modulates Mast Cell Chemotaxis to Kit-Ligand, but Does Not Impact Mast Cell Numbers, Degranulation, or Tumor Behavior in Neurofibromas of Nf1-Deficient Mice. Mol. Cancer Ther. 2019, 18, 2321–2330. [Google Scholar] [CrossRef] [Green Version]
  105. Giannou, A.D.; Marazioti, A.; Spella, M.; Kanellakis, N.I.; Apostolopoulou, H.; Psallidas, I.; Prijovich, Z.M.; Vreka, M.; Zazara, D.E.; Lilis, I.; et al. Mast cells mediate malignant pleural effusion formation. J. Clin. Investig. 2015, 125, 2317–2334. [Google Scholar] [CrossRef] [PubMed]
  106. Xu, W.; Qian, J.; Zeng, F.; Li, S.; Guo, W.; Chen, L.; Li, G.; Zhang, Z.; Wang, Q.J.; Deng, F. Protein kinase Ds promote tumor angiogenesis through mast cell recruitment and expression of angiogenic factors in prostate cancer microenvironment. J. Exp. Clin. Cancer Res. 2019, 38, 114. [Google Scholar] [CrossRef] [Green Version]
  107. Ellem, S.J.; Taylor, R.A.; Furic, L.; Larsson, O.; Frydenberg, M.; Pook, D.; Pedersen, J.; Cawsey, B.; Trotta, A.; Need, E.; et al. A pro-tumourigenic loop at the human prostate tumour interface orchestrated by oestrogen, CXCL12 and mast cell recruitment. J. Pathol. 2014, 234, 86–98. [Google Scholar] [CrossRef]
  108. Ronca, R.; Tamma, R.; Coltrini, D.; Ruggieri, S.; Presta, M.; Ribatti, D. Fibroblast growth factor modulates mast cell recruitment in a murine model of prostate cancer. Oncotarget 2017, 8, 82583–82592. [Google Scholar] [CrossRef] [Green Version]
  109. Nagasaka, A.; Matsue, H.; Matsushima, H.; Aoki, R.; Nakamura, Y.; Kambe, N.; Kon, S.; Uede, T.; Shimada, S. Osteopontin is produced by mast cells and affects IgE-mediated degranulation and migration of mast cells. Eur. J. Immunol. 2008, 38, 489–499. [Google Scholar] [CrossRef]
  110. Ma, Y.; Hwang, R.F.; Logsdon, C.D.; Ullrich, S.E. Dynamic mast cell-stromal cell interactions promote growth of pancreatic cancer. Cancer Res. 2013, 73, 3927–3937. [Google Scholar] [CrossRef] [Green Version]
  111. Cao, Y.; Langer, R.; Ferrara, N. Targeting angiogenesis in oncology, ophthalmology and beyond. Nat. Rev. Drug Discov. 2023, 22, 476–495. [Google Scholar] [CrossRef]
  112. Ammendola, M.; Leporini, C.; Marech, I.; Gadaleta, C.D.; Scognamillo, G.; Sacco, R.; Sammarco, G.; De Sarro, G.; Russo, E.; Ranieri, G. Targeting mast cells tryptase in tumor microenvironment: A potential antiangiogenetic strategy. Biomed. Res. Int. 2014, 2014, 154702. [Google Scholar] [CrossRef]
  113. 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] [Green Version]
  114. Qian, N.; Li, X.; Wang, X.; Wu, C.; Yin, L.; Zhi, X. Tryptase promotes breast cancer angiogenesis through PAR-2 mediated endothelial progenitor cell activation. Oncol. Lett. 2018, 16, 1513–1520. [Google Scholar] [CrossRef] [PubMed]
  115. Winer, A.; Adams, S.; Mignatti, P. Matrix Metalloproteinase Inhibitors in Cancer Therapy: Turning Past Failures Into Future Successes. Mol. Cancer Ther. 2018, 17, 1147–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Sarasola, M.P.; Taquez Delgado, M.A.; Nicoud, M.B.; Medina, V.A. Histamine in cancer immunology and immunotherapy. Current status and new perspectives. Pharmacol. Res. Perspect. 2021, 9, e00778. [Google Scholar] [CrossRef] [PubMed]
  117. Laster, S.M.; Wood, J.G.; Gooding, L.R. Tumor necrosis factor can induce both apoptic and necrotic forms of cell lysis. J. Immunol. 1988, 141, 2629–2634. [Google Scholar] [CrossRef] [PubMed]
  118. Kearney, C.J.; Cullen, S.P.; Tynan, G.A.; Henry, C.M.; Clancy, D.; Lavelle, E.C.; Martin, S.J. Necroptosis suppresses inflammation via termination of TNF- or LPS-induced cytokine and chemokine production. Cell Death Differ. 2015, 22, 1313–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Creaven, P.J.; Brenner, D.E.; Cowens, J.W.; Huben, R.P.; Wolf, R.M.; Takita, H.; Arbuck, S.G.; Razack, M.S.; Proefrock, A.D. A phase I clinical trial of recombinant human tumor necrosis factor given daily for five days. Cancer Chemother. Pharmacol. 1989, 23, 186–191. [Google Scholar] [CrossRef]
  120. Gamm, H.; Lindemann, A.; Mertelsmann, R.; Herrmann, F. Phase I trial of recombinant human tumour necrosis factor alpha in patients with advanced malignancy. Eur. J. Cancer 1991, 27, 856–863. [Google Scholar] [CrossRef]
  121. Lenk, H.; Tanneberger, S.; Muller, U.; Ebert, J.; Shiga, T. Phase II clinical trial of high-dose recombinant human tumor necrosis factor. Cancer Chemother. Pharmacol. 1989, 24, 391–392. [Google Scholar] [CrossRef] [PubMed]
  122. Xu, Y.; Shen, Y.; Ouahab, A.; Li, C.; Xiong, Y.; Tu, J. Antitumor activity of TNF-alpha after intratumoral injection using an in situ thermosensitive hydrogel. Drug Dev. Ind. Pharm. 2015, 41, 369–374. [Google Scholar] [CrossRef] [PubMed]
  123. Iltis, I.; Choi, J.; Vollmers, M.; Shenoi, M.; Bischof, J.; Metzger, G.J. In vivo detection of the effects of preconditioning on LNCaP tumors by a TNF-alpha nanoparticle construct using MRI. NMR Biomed. 2014, 27, 1063–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Afshari, A.R.; Sanati, M.; Mollazadeh, H.; Kesharwani, P.; Johnston, T.P.; Sahebkar, A. Nanoparticle-based drug delivery systems in cancer: A focus on inflammatory pathways. Semin. Cancer Biol. 2022, 86, 860–872. [Google Scholar] [CrossRef]
  125. Dakhel, S.; Lizak, C.; Matasci, M.; Mock, J.; Villa, A.; Neri, D.; Cazzamalli, S. An Attenuated Targeted-TNF Localizes to Tumors In Vivo and Regains Activity at the Site of Disease. Int. J. Mol. Sci. 2021, 22, 10020. [Google Scholar] [CrossRef]
  126. Hemmerle, T.; Probst, P.; Giovannoni, L.; Green, A.J.; Meyer, T.; Neri, D. The antibody-based targeted delivery of TNF in combination with doxorubicin eradicates sarcomas in mice and confers protective immunity. Br. J. Cancer 2013, 109, 1206–1213. [Google Scholar] [CrossRef]
  127. Curnis, F.; Sacchi, A.; Borgna, L.; Magni, F.; Gasparri, A.; Corti, A. Enhancement of tumor necrosis factor alpha antitumor immunotherapeutic properties by targeted delivery to aminopeptidase N (CD13). Nat. Biotechnol. 2000, 18, 1185–1190. [Google Scholar] [CrossRef]
  128. Corti, A.; Pastorino, F.; Curnis, F.; Arap, W.; Ponzoni, M.; Pasqualini, R. Targeted drug delivery and penetration into solid tumors. Med. Res. Rev. 2012, 32, 1078–1091. [Google Scholar] [CrossRef]
  129. Gregorc, V.; Santoro, A.; Bennicelli, E.; Punt, C.J.; Citterio, G.; Timmer-Bonte, J.N.; Caligaris Cappio, F.; Lambiase, A.; Bordignon, C.; van Herpen, C.M. Phase Ib study of NGR-hTNF, a selective vascular targeting agent, administered at low doses in combination with doxorubicin to patients with advanced solid tumours. Br. J. Cancer 2009, 101, 219–224. [Google Scholar] [CrossRef] [Green Version]
  130. Parmiani, G.; Pilla, L.; Corti, A.; Doglioni, C.; Cimminiello, C.; Bellone, M.; Parolini, D.; Russo, V.; Capocefalo, F.; Maccalli, C. A pilot Phase I study combining peptide-based vaccination and NGR-hTNF vessel targeting therapy in metastatic melanoma. Oncoimmunology 2014, 3, e963406. [Google Scholar] [CrossRef] [Green Version]
  131. Cappell, K.M.; Kochenderfer, J.N. Long-term outcomes following CAR T cell therapy: What we know so far. Nat. Rev. Clin. Oncol. 2023, 20, 359–371. [Google Scholar] [CrossRef] [PubMed]
  132. Maalej, K.M.; Merhi, M.; Inchakalody, V.P.; Mestiri, S.; Alam, M.; Maccalli, C.; Cherif, H.; Uddin, S.; Steinhoff, M.; Marincola, F.M.; et al. CAR-cell therapy in the era of solid tumor treatment: Current challenges and emerging therapeutic advances. Mol. Cancer 2023, 22, 20. [Google Scholar] [CrossRef] [PubMed]
  133. Berrien-Elliott, M.M.; Jacobs, M.T.; Fehniger, T.A. Allogeneic natural killer cell therapy. Blood 2023, 141, 856–868. [Google Scholar] [CrossRef] [PubMed]
  134. Allavena, P.; Anfray, C.; Ummarino, A.; Andon, F.T. Therapeutic Manipulation of Tumor-associated Macrophages: Facts and Hopes from a Clinical and Translational Perspective. Clin. Cancer Res. 2021, 27, 3291–3297. [Google Scholar] [CrossRef] [PubMed]
  135. Chang, D.Z.; Ma, Y.; Ji, B.; Wang, H.; Deng, D.; Liu, Y.; Abbruzzese, J.L.; Liu, Y.J.; Logsdon, C.D.; Hwu, P. Mast cells in tumor microenvironment promotes the in vivo growth of pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2011, 17, 7015–7023. [Google Scholar] [CrossRef] [Green Version]
  136. 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] [Green Version]
  137. Jimenez-Andrade, G.Y.; Ibarra-Sanchez, A.; Gonzalez, D.; Lamas, M.; Gonzalez-Espinosa, C. Immunoglobulin E induces VEGF production in mast cells and potentiates their pro-tumorigenic actions through a Fyn kinase-dependent mechanism. J. Hematol. Oncol. 2013, 6, 56. [Google Scholar] [CrossRef] [Green Version]
  138. Siebenhaar, F.; Metz, M.; Maurer, M. Mast cells protect from skin tumor development and limit tumor growth during cutaneous de novo carcinogenesis in a Kit-dependent mouse model. Exp. Dermatol. 2014, 23, 159–164. [Google Scholar] [CrossRef]
  139. 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]
  140. 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]
Figure 1. Strategies for MC-based immunotherapy in cancer. As described in the text, approaches to address MCs functions in cancer can rely on: (1) targeting c-Kit signaling; (2) stabilizing MC degranulation; (3) triggering activating/inhibiting receptors; (4) modulating MC recruitment; (5) harnessing MC mediators; (6) adoptive transferring of MCs. As MCs can exert tumor-promoting or suppressive activities depending on tumor type, their localization, and signals received from the surrounding microenvironment, therapeutic strategies could be either directed to abrogate or prompt MC functions according to the specific context. This picture was created with BioRender.com.
Figure 1. Strategies for MC-based immunotherapy in cancer. As described in the text, approaches to address MCs functions in cancer can rely on: (1) targeting c-Kit signaling; (2) stabilizing MC degranulation; (3) triggering activating/inhibiting receptors; (4) modulating MC recruitment; (5) harnessing MC mediators; (6) adoptive transferring of MCs. As MCs can exert tumor-promoting or suppressive activities depending on tumor type, their localization, and signals received from the surrounding microenvironment, therapeutic strategies could be either directed to abrogate or prompt MC functions according to the specific context. This picture was created with BioRender.com.
Pharmaceutics 15 01692 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sulsenti, R.; Jachetti, E. Frenemies in the Microenvironment: Harnessing Mast Cells for Cancer Immunotherapy. Pharmaceutics 2023, 15, 1692. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics15061692

AMA Style

Sulsenti R, Jachetti E. Frenemies in the Microenvironment: Harnessing Mast Cells for Cancer Immunotherapy. Pharmaceutics. 2023; 15(6):1692. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics15061692

Chicago/Turabian Style

Sulsenti, Roberta, and Elena Jachetti. 2023. "Frenemies in the Microenvironment: Harnessing Mast Cells for Cancer Immunotherapy" Pharmaceutics 15, no. 6: 1692. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics15061692

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