Next Article in Journal
The Role of SOX9 in IGF-II-Mediated Pulmonary Fibrosis
Previous Article in Journal
Mosaic Genome of a British Cider Yeast
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Innate Immunity in Cancer Biology and Therapy

1
Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
2
Shanghai Tinova Immune Therapeutics Co., Ltd., Shanghai 201413, China
3
Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(14), 11233; https://doi.org/10.3390/ijms241411233
Submission received: 21 May 2023 / Revised: 5 July 2023 / Accepted: 5 July 2023 / Published: 8 July 2023
(This article belongs to the Section Molecular Oncology)

Abstract

:
Immunotherapies including adaptive immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells, have developed the treatment of cancer in clinic, and most of them focus on activating T cell immunity. Although these strategies have obtained unprecedented clinical responses, only limited subsets of cancer patients could receive long-term benefits, highlighting the demand for identifying novel targets for the new era of tumor immunotherapy. Innate immunity has been demonstrated to play a determinative role in the tumor microenvironment (TME) and influence the clinical outcomes of tumor patients. A thorough comprehension of the innate immune cells that infiltrate tumors would allow for the development of new therapeutics. In this review, we outline the role and mechanism of innate immunity in TME. Moreover, we discuss innate immunity-based cancer immunotherapy in basic and clinical studies. Finally, we summarize the challenges in sufficiently motivating innate immune responses and the corresponding strategies and measures to improve anti-tumor efficacy. This review could aid the comprehension of innate immunity and inspire the creation of brand-new immunotherapies for the treatment of cancer.

1. Introduction

Cancer is one of the main leading causes of disease-associated death and it often results from genotoxic and non-genotoxic carcinogens [1]. Surgery, chemotherapy, radiotherapy and tumor immunotherapy are the principle treatment for tumors. Surgery is the treatment of choice for most solid tumors, but cannot be used for hematologic tumors and metastases and subclinical metastases of solid tumors. Compared to surgical treatment, chemotherapy is a systemic treatment for primary, metastatic and subclinical metastases of solid tumors as well as hematologic tumors. However, the poor selectivity of chemotherapeutic agents limits its clinical application. Radiation therapy protects tissue that is not affected by the tumor, produces less damage and does not require hospitalization, but the application of radiotherapy depends on the type, size and location of tumors. Compared with these traditional therapies, tumor immunotherapy can kill tumor cells by activating the host’s immune system [2,3]. The discipline of oncology has been undergoing an unprecedented revolution due to the massive advancement of immunotherapy in the treatment of cancer [4]. Infiltrating into the TME, immune cells assist in tuning the development of tumors [5,6]. In adaptive immunity, T cells participate in immune responses that are cell-mediated immune responses [7,8,9]. The treatment of solid tumors has been revolutionized by ICIs targeting PD-1/PD-L1. Blocking the PD-1/PD-L1 axis could enhance T cell infiltration and revitalize exhausted cytotoxic T cells, presenting impressive therapeutic efficacy in various cancers [10]. Interestingly, PD-L1 expression and sensitivity to immunotherapy of tumor cells are subject to metabolic reprogramming. For example, mitochondrial oxidative phosphorylation (OXPHOS) can regulate PD-L1 levels. Tumor-selective OXPHOS suppression nanoparticles have been reported to reactivate immunotherapy and may open a new therapeutic window for patients [11]. Furthermore, CAR T cells have shown impressive efficacy in the treatment of hematologic malignancies [12,13,14]. As the mechanisms of tumor development and treatment are further explored, many new strategies have also emerged. For instance, oxidative stress and reactive oxygen species (ROS) contribute to the development of cancer growth [15]. Myrrh, a traditional remedy, could be a possible therapeutic agent for cancer because of the powerful antioxidant activity of its extracts [15]. Pyroptosis is a form of programmed cell death, which is associated with tumor genesis, and therapy response. Inflammasomes are multimolecular complexes containing pattern recognition receptors that can recruit the adaptor protein containing apoptosis-associated speck-like protein containing a caspase recruitment domain and activate caspase-1. To date, six members of the gasdermin (GSDM) family have been identified, namely GSDMA, GSDMB, GSDMC, GSDMD, GSDME (DFNA5) and DFNB59. Inflammasomes can activate caspase-1 that can cleave GSDMD to induce pyroptosis [16]. It is reported that the virus-like particle can trigger the formation of an AIM2 inflammasome that can induce GSDMD-mediated pyroptosis, thus enhancing antitumor immunity [17]. The primary goal of the currently approved immunotherapies is to active T cell immunity, but innate immunity also has significant anti-tumor potential. The desire to identify novel targets in innate immunity has emerged in the new era of immunotherapy.
In the innate immune system, myeloid-derived suppressor cells (MDSCs), macrophages, neutrophils, natural killer (NK) cells, dendritic cells (DCs), mast cells (MCs) and helper innate lymphoid cells (ILCs) have important regulatory effects on tumor progression [18]. Some innate immune cells have the capacity for detecting and eliminating tumor cells through various mechanisms, such as intrinsic cytotoxicity of NK cells and macrophage phagocytosis. The interaction of the innate and adaptive immunity is exemplified by the uptake of tumor antigens by antigen-presenting cells (APCs), which results in cross-presentation and the priming of CD8+ T lymphocytes [19]. Innate immune cells are also involved in effector responses through antibody-dependent cellular phagocytosis (ADCP) and antibody-dependent cell cytotoxicity (ADCC) [20]. In this review, we state the roles of innate immunity in tumors, emphasizing the regulatory mechanisms by which innate immune cells recognize and respond to tumor cells and promote adaptive immune responses. We then highlight promising preclinical and clinical studies involving immunotherapies based on innate immune cells. Finally, we conclude the challenges in innate immunotherapy and propose corresponding strategies to improve the efficacy and facilitate its application in clinic.

2. Innate Immunity in TME

Composed of MDSCs, macrophages, neutrophils, NK cells, DCs, MCs and helper ILCs, the innate immunity plays a crucial role in defense against tumors (Figure 1). For example, macrophages can activate the tumor-killing activity of T cells through antigen cross-presentation. Antigen cross-presentation by macrophages can be achieved through the cytosolic and vacuolar pathways. In the cytosolic pathway, proteins are transported to the cell membrane, where they are degraded by the proteasome. Subsequently, the derived peptides are transferred via the transporter associated with antigen presentation to the endoplasmic reticulum (ER), where they are processed by aminopeptidases of the ER, or brought back to the antigen-containing endosome to be processed by insulin-regulated aminopeptidases. In this process, the peptide is loaded on the MHC-I. The other major cross-presentation pathway is the vacuolar pathway, in which proteins are processed by endosomal/lysosomal proteases and loaded on MHC-I within the endosomal/lysosomal compartments. The resulting peptide MHC-I molecular complex is recognized by CD8+ T cells expressing specific T cell receptors (TCRs). The signal from TCR recognition of antigen is transduced into CD8+ T cells, which triggers cytotoxic effects [21]. Neutrophils can promote tumor proliferation and invasion by secreting proteases, and can also inhibit tumor progression by secreting H2O2, based on the type, stage and location of the tumors [22]. NK cells achieve anti-tumor effects by mediating ADCC, and MCs can promote tumors or fight tumors by releasing cytokines, chemokines, eicosanoids, proteoglycans and biogenic amines [23,24]. A comprehensive understanding of innate immune cell function in the TME is vital for the design of effective cancer immunotherapy.

2.1. MDSCs

As a heterogeneous population of immature myeloid cells that possess potent immunosuppressive properties, MDSCs are typically classified into two major subsets based on their phenotypic, morphological and functional properties: mononuclear phagocyte-like MDSCs (M-MDSCs) and granulocyte-like MDSCs (G-MDSCs) [25,26,27]. G-MDSCs are predominantly abundant within the TME, whereas M-MDSCs primarily accumulate in the peripheral blood [28]. MDSCs play a crucial role in modulating immune responses and their dysregulation is associated with numerous diseases, including cancer, chronic infections, autoimmune disorders and inflammatory conditions [27]. In cancer, MDSCs suppress antitumor immune responses, promote tumor progression and contribute to immune evasion through a number of mechanisms, including boosting the production of Arg1, iNOS, ROS and nitrogen compounds like peroxynitrite (PNT) [29]. When Arg-1 is increased, L-arginine, an essential amino acid for T cell proliferation, could be metabolized into urea and L-ornithine [30]. When L-arginine is restricted, the up-regulation of iNOS contributes to NO production, which reacts with superoxide to produce PNT [31]. Subsequently, PNT causes the TCR nitration and nitrosylation, inhibiting the function of CD8+ T cells and promoting T cell tolerance [32,33]. Therefore, one crucial way of targeting MDSCs to reverse the immunosuppressive TME is to understand the basic mechanisms of MDSC recruitment into the TME [28].

2.2. Macrophages

Macrophages, which originate from myeloid-derived progenitor cells in the bone marrow, are the main phagocytes in tumors [34]. Macrophages can eliminate cellular debris clear pathogens and regulate inflammatory responses, which are essential for homeostasis maintenance [35]. Macrophages can be polarized into M1 phenotype and M2 phenotype [36]. M1 macrophages are characterized by producing pro-inflammatory cytokines such as interleukin (IL)-6, IL-12, and interferon (IFN)-γ, which possess anti-tumor properties. In addition, M1 macrophages recruit T helper type 1 (Th1) cells by secreting the chemokines CXCL9 and CXCL10, and promote T cell responses by upregulating the gene of antigen presentation and costimulatory molecules [35]. In contrast, M2 macrophages exhibit pro-tumor activities and produce immunosuppressive cytokines such as IL-4, IL-10, and transforming growth factor-β (TGF-β). In the TME, tumor-infiltrating macrophages, also known as tumor-associated macrophages (TAM), mainly exhibit an M2-like phenotype [37,38].

2.3. Neutrophils

Neutrophils, derived from the early committed neutrophil progenitor cells, are considered to be the most massive innate immune cells in the bone marrow and peripheral blood. These cells can respond quickly to inflammation, infection and injury. They can promote cancer progression through various mechanisms, such as angiogenesis, immunosuppression and cancer metastasis. In promoting tumor invasion, neutrophils secrete proteases that degrade structural proteins in the extracellular environment [39,40,41]. For instance, studies on breast cancer have shown that neutrophils can induce MMP-12 and MMP-13 to promote cancer metastasis [42]. Independent clinical studies have also confirmed the important role of neutrophils in tumor metastasis [43,44,45,46,47]. In addition, IL-1β promotes IL-17 production from γδT cells, leading to polarization of neutrophils in mice with mammary tumors [48]. Nonetheless, neutrophils can also exert anti-tumor by, for example, secreting H2O2 to cause an influx of Ca2+ and inducing apoptosis in tumor cells via Fas ligand/Fas interaction [22].

2.4. NK Cells

NK cells are cytotoxic lymphocytes that hold the capacity to kill cancerous cells. In general, NK cells can kill tumor cells via direct cytotoxicity and pro-inflammatory cytokines production [24]. “Missing-self” is one mechanism of direct cytotoxicity. Immunoreceptor tyrosine-based inhibitory motifs (ITIMs) can engage with the major histocompatibility complex-I (MHC-I) to stop NK cells from destroying ordinary cells, which means NK cell activation can be inhibited by the binding of MHC-I. However, tumor cells always lack or only very faintly express MHC-I in order to escape CD8+ T cell-mediated cytotoxicity. Thus, NK cells can recognize and reply to this missing-self phenotype [49]. ADCC is another crucial mechanism of NK cell-mediated direct cytotoxicity [24]. Additionally, NK cells express significant quantities of IFN-γ and TNF. They can not only enhance cytotoxic CD8+ T cell response, but also can they suppress the proliferation and angiogenesis of tumor cells while they promote the apoptosis of cancerous cells [50].

2.5. DCs

First discovered in 1973 by Ralph Steinman, DC is one of the most important APCs, which plays a significant role in launching and modulating innate and adaptive immune responses [51]. DCs comprise four main subsets: cDC1s, cDC2s, plasmacytoid DCs (pDCs) and monocyte-derived DCs (MoDCs). In the TME, DCs recognize, uptake, process and present tumor-associated antigens (TAAs) to T cells to shape T cell responses [52]. Distinct DCs have different T cell priming abilities. cDC1s excel at cross-presentation antigens, which could activate CD8+ T cells and promote Th1 cell polarization of CD4+ T cells. cDC2s are essential to inducing CD4+ T cell responses [53]. pDCs could effectively prime CD8+ T cells while showing poor priming of naive T cells [54,55,56]. MoDCs are mainly generated in inflammatory responses and are critical to the differentiation of CD4+ T cells. When perceiving appropriate cues, DCs express costimulatory molecules such as CD80, CD86, CD137L, OX40L, GITRL, CD70 and CD40 to regulate DC-mediated T cell priming to influence T cell-mediated immunotherapy in TME [51]. DCs also upregulated chemokine receptors such as cc-chemokine receptor 7 (CCR7) [57]. Studies show that CCR7 is critical for the migration of tumor-infiltrating DCs and can impact DCs recruitment into the TME. Chemokines such as CXC-chemokine ligand 9 (CXCL9) and CXCL10 are also produced by DCs to facilitate the recruitment of CD8+ T cells in the TME [58]. In addition, DCs could produce cytokines such as IL-12 and type 1 TNF; IL-12s are pivotal to the initiation of Th1 cells and CD8+ T cells, and type 1 TNFs have been used to treat patients with cancer [19,59,60,61].

2.6. MCs

MCs are derived from bone marrow precursors, which exert either pro- or anti-tumor effects by secreting cytokines, chemokines, eicosanoids, proteoglycans and biogenic amines after activation [23]. Studies demonstrate that IL-33-mediated MC activation promotes TAM infiltration and tumorigenesis by secreting CSF2, CCL3 and IL-6 in gastric cancer [62]. Eicosanoids, such as LTC4, LTB4 and PGD2, are also secreted upon MC activation. Transforming LTC4 to LTD4 by γ-Glutamyl transpeptidase 1 (GGT1) and GGT5, enzymes that hydrolyze γ-glutamyl of glutathione, results in lung cancer cell migration and survival via CysLT1 [63,64]. LTB4 has the potential to be a biomarker for predicting the effect of bestatin in colorectal cancer (CRC) [65]. PGD2 can suppress tumorigenesis and metastasis via PGD2/PTGDR2 signaling [66,67]. Mast cell-derived heparin is a kind of proteoglycan that could increase angiogenesis. In mouse EL-4 lymphoma and MC-38 colon cancer, histamine produced by MCs could target MDSCs and enhance the anti-tumor efficacy of checkpoint blocking [68].

2.7. Helper ILCs

ILCs are a subset of lymphocytes that lack antigen-specific receptors. There are five subsets of ILCs: NK cells, lymphoid tissue inducer cells, and groups 1, 2, and 3 (ILC1, ILC2, and ILC3) [69,70]. The Helper ILC family, which consists of ILC1, ILC2, and ILC3, can be utilized to induce and stimulate anti-tumor responses [71]. ILC1s have the capacity to express IFN-γ, NKp46, and T-bet [72]. Type 2 cytokines, including IL-5 and IL-13, are produced by ILC2s in response to TSLP, IL-18, IL-33, or IL-25 [73]. Tumor-infiltrating helper ILCs can either stimulate or restrict the proliferation of cancer cells through various mechanisms. ILC1s can produce IFN-γ and granzymes to suppress tumor growth, but express immune checkpoints on their surface may impair their function and promote tumor growth [72]. Upon stimulation with IL-33, ILC2s can secrete high levels of IL-5, GM-CSF, and CCL5, which can activate and recruit anti-tumorigenic eosinophils, DCs, and CD8+ T cells [74]. In contrast, the production of immune checkpoint molecules like PD-1 and the cytokines IL-4 and IL-13 dampens ILC2 activity and accelerates tumor growth [75]. Under the regulation of TGF-β signaling, ILC3s can also convert to ILC1s or regulatory ILCs [76]. According to the type of tumor, ILC3s produce cytokines including IL-17 and IL-22 that promote or inhibit tumor development [77,78].

3. Immunotherapies Targeting Innate Immune System

Current immunotherapies including ICIs, cell therapies and tumor vaccines, present potent antitumor efficiency [79,80,81]. Through disrupting the immune checkpoint pathway, ICIs have become the most widely used agents in cancer immunotherapy [82]. CAR T cell therapy was developed from adoptive T cell transfer, which typically modifies and redirects patients’ own T cells to recognize and kill tumor cells. Recently, CAR-T cell therapy has achieved notable success in the treatment of hematologic malignancies [79]. Through inducing APC cell-mediated immune responses, tumor vaccines could be designed to eradicate malignant cells [83]. Recently, immunotherapies based on innate immune cells have emerged, including innate ICIs, CAR innate immune cells and cytokine therapy. Innate ICIs achieve anti-tumor purposes by targeting checkpoints such as SIRPα, TIGIT, PVRIG, LILRB2, NKG2A, LAG-3, TIM-3, VISTA and CD32B (Table 1; Figure 2A) [84]. NK cells, NKT cells, γδT cells and macrophages engineered with CAR may be redirected effectively towards specific antigens (Table 1; Figure 2B) [85]. GM-CSF, type I IFNs, FLT3L, IL-2 and IL-15 can promote the development and proliferation of APCs as well as the activation of NK cells (Table 2; Figure 2C) [86].

3.1. Innate ICIs

Innate ICIs have the potential to both provide stimulation and disrupt inhibitory relationships between malignancies and phagocytes or NK cells. These checkpoints include Signal-regulatory protein-α (SIRPα), T-cell Ig and ITIM domain (TIGIT), Poliovirus receptor-related Ig domain containing protein(PVRIG), Leukocyte immunoglobulin-like receptor B (LILRB2), NK group 2 member A (NKG2A), T cell immunoglobulin and mucin domain 3 (TIM-3), V-domain immunoglobulin suppressor of T cell activation (VISTA), FcγRIIB (CD32B), lymphocyte activation gene 3 protein (LAG-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) [84,86].
The ligand of SIRPα is CD47, which is abundantly presented on various cancerous cells [87]. The SIRPα/CD47 interaction leads to phosphorylating the ITIMs and disrupting the cytoskeleton, and then inhibiting phagocytosis [87]. Blocking the SIRPα/CD47 axis could not only activate adaptive anti-tumor immune responses by promoting antigen cross-presentation by APCs, but also could activate innate immune responses by enhancing the cancer cell clearance by macrophages and DCs [84,88]. TTI-621 is a fusion protein consisting of the CD47 binding domain of human SIRPα and linked to the Fc region of IgG1. Studies show that TTI-621 could block the “do not eat me” signal of CD47 and boost phagocytosis and anti-tumor effect, and intra-tumoral injections of TTI-621 are used to treat patients with mycosis fungoides and/or Sézary syndrome, which result in a rapid decrease in tumor size [89,90,91].
TIGIT is a suppressible receptor presented on NK cells, CD4+ T cells, CD8+ T cells and regulatory T cells [86]. In addition, the ligands of TIGIT, CD112 and CD155, are presented on tumor cells [86]. According to preclinical research, TIGIT blockade could enhance NK cell activity and CD8+ T cell cytotoxicity [92,93,94,95]. Many monoclonal antibodies (mAbs) have been designed to prevent the inhibitory function of human TIGIT. Tiragolumab is a humanized anti-TIGIT IgG1 mAb created to prevent TIGIT from interacting with CD155. Tiragoulumab alone was used to treat 24 cancer patients who were refractory in an Ia/Ib phase experiment, and the medication resulted in stable disease. Additionally, 49 individuals received treatment with tiragolumab plus atezolizumab, which led to improvements in 3 patients, including partial responses in NSCLC and head and neck squamous cell carcinoma (HNSCC) [84]. Similar to TIGIT, PVRIG, presented on NK cells and CD8+ T cells, is a suppressible receptor that could identify CD112 on tumor cells [96]. Preclinical studies suggest that anti-PVRIG therapy could enhance CD8+ T cell cytotoxicity and NK cell anti-tumor effect [94,97].
LILRB2 is presented on innate immune cells and CD4+ T cells, which could interact with MHC-I on nucleated cells [98]. Acetylated by acetyltransferase CREB-binding protein (CBP), STAT6 could suppress M2 polarization during macrophage maturation [99]. In the presence of macrophage colony-stimulating factor M-CSF and IL-4, LILRB2 inhibitors can regulate M2 polarization by inhibiting STAT6 activation [100]. LILRB2 antagonism also could improve NF-κB and STAT1 activation and reduce the suppressive effect of macrophages on T cell proliferation. Additionally, when combined with anti-PD-L1, LILRB2 blockade could remodel the TME and provoke anti-tumor immunity [100].
NKG2A and CD94 form a suppressible receptor which is presented on NK cells and CD8+ T cells and is capable of recognizing HLA-E [86]. Both hematologic and solid tumors upregulate HLA-E expression, which has been linked to a worse prognosis and lower NK cell cytotoxicity [92,101]. In cancer patients, combining NKG2A inhibition with PD-L1 blockade may increase the anti-tumor efficacy of NK and CD8+ T cells. Monalizumab is an anti-NKG2A antibody that, when combined with PD-x axis blockade, can improve NK cell activity against multiple tumor cells and promote the CD8+ T cell effect. Monalizumab and cetuximab were used in a trial to treat patients with HNSCC, and led to a 31% objective response rate [102].
While immune checkpoints such as LAG-3, TIM-3, VISTA and CTLA-4 are primarily adaptive immunological checkpoints, they may also have innate immune effects. It has been shown that LAG-3 reduces the effector functions of NK cells, NKT cells and DCs [86]. Through inhibiting tumor nucleic acid sensing, TIM-3 suppresses tumor detection of DCs [103]. Elevated expression of VISTA was reported in macrophages, DCs and monocytes [104]. CTLA-4 binds to its ligands, CD86 and CD80 that are expressed on APCs such as DCs and macrophages, leading to the depletion of naive and memory T cells [105]. Many areas of current and future research will probably elucidate the significance of these checkpoints in tumor treatment.

3.2. Innate Immune Cells Engineered with CARs

Consisting of an extracellular region, hinge region, transmembrane domain and intracellular domain, CAR is a functional chimeric antigen receptor that typically confers an immune cell with the specificity for TAAs [106]. Usually, single-chain variable fragment in the extracellular region can specifically recognize antigens expressed on tumor cells. The intracellular domain usually includes co-stimulatory molecules and the CD3ζ domain of TCR. When the CAR recognizes the specific antigen, immunoreceptor tyrosine-based activation motifs are phosphorylated, thereby recruiting adaptor molecules, regulating downstream pathways and activating immune cells [107]. Innate immune cells such as macrophages, NK cells and NKT cells can also be used for CAR redirection despite the fact that T cells predominate in CAR-based immunotherapy [85].
CAR-M treatment is based on a 1 week manufacturing procedure that begins with a patient’s own blood. Subcutaneous G-CSF treatment mobilizes monocytes prior to leukapheresis and CD14+ monocyte selection. Following ex vivo differentiation of monocytes into macrophages, Ad5f35—which encodes the CAR transgene—is next introduced into the cells. Surprisingly, CAR-M, generated with Ad5f35, could eradicate tumor cells more efficiently than M1 macrophages. Additionally, Ad5f35 transduced macrophages did not differentiate into M2 macrophages when stimulated with IL-4, IL-10, IL-13 or a tumor-conditioned medium. Notably, CAR-M macrophages could upregulate antigen presentation pathways and exhibit increased T cell activation capability as compared to control macrophages [108].
The initial signaling domain of CAR-NK cells is frequently CD3, and the costimulatory domain is either CD28 or CD137. Due to their improved tumor-specific targeting and cytotoxicity, CAR NK cells have been employed to identify cancer cells [109]. NKT cells are recognized by the production of TCRs and NK cell lineage markers [110]. Studies have shown that NKT modified with ganglioside GD2-specific CARs has a killing effect on GD2-positive tumor cells as well as CD1d-positive M2 macrophages in vitro, and CD19-specific CAR-modified NKT cells can be used to treat B-cell lymphoma [111,112]. CAR γδT cells can recognize multiple antigens and are tumor cytotoxic [113]. CD19-directed CAR γδT cells target both CD19-positive cell lines and CD19-negative leukemia cells [114].

3.3. Cytokines

Innate immunity plays a central role in promoting T cell effector functions. To enhance this effect, many attempts have been made and the results show that cytokines, such as GM-CSF, type I IFNs, FLT3L, IL-2 and IL-15, can activate NK cells or promote the maturation and multiplication of APC [86].
GM-CSF could recruit NK cells and DCs to promote the presentation of TAAs thereby mediating anti-tumor effects. In mice, a GM-CSF injection could stimulate immunity and present long-lasting anti-tumor responses [86]. In patients with melanoma, an intra-tumoral injection of GM-CSF could contribute to increasing the number of tumor-infiltrating DCs. Talimogene laherparepvec (T-VEC) is an engineered oncolytic virus with the gene that encodes GM-CSF, and it is demonstrated that its efficacy is superior to GM-CSF in patients with advanced melanoma [115]. Studies suggest that the therapeutic effect of T-VEC is related to the increase in tumor-specific T cells and the decrease in Treg cells [115].
As a protein localized on the ER membrane, the stimulator of interferon genes (STING) plays a vital role in activating immune responses [116,117]. Cyclic GMP-AMP synthase (cGAS) is a cellular DNA sensor that could recognize pathogen-released cytoplasmic double-stranded DNA (dsDNA). Cytosolic dsDNA, originating from various sources such as chromosomal instability, acute genomic stressors and mitochondrial dysfunction, is frequently abundant in cancer cells [118]. cGAS binds to its DNA ligand and forms a dimer, in which the DNA is sandwiched between two cGAS protomers. Active cGAS binds to ATP and GTP and catalyzes the condensation reaction of these two nucleotides to produce 2′,3′-cyclic GMP–AMP (2′,3′-cGAMP). Then, cGAMP binds to STING, and results in TANK-binding kinase 1-dependent phosphorylation of interferon regulatory factor 3 (IRF3). Upon activation, IRF3 translocates into the nucleus where it initiates the transcriptional activation of type I IFNs. Additionally, the cGAS-STING signaling pathway induces the expression of genes encoding pro-inflammatory cytokines via NF-κB activation [118,119]. Type I IFNs are significant for the spontaneous activation of tumor-specific CD8+ T cells and have considerable influence on the capacity of APCs to deliver dead cell-associated antigens [120]. Moreover, type 1 INF could promote the generation of CXCL9 and CXCL10 to impact the activity and recruitment of NK cells in the TME [121]. Of note, interferon-induced tetratricopeptide repeat (IFIT) proteins could influence the translation of a diverse array of viruses. As one of the most crucial members of both the IFIT family and the interferon-stimulated gene family, IFIT3 is vital for cell proliferation, differentiation and cancer metastasis, eliciting an important role in antiviral innate immunity [122]. A study shows that the addition of IFN2a can improve the anti-tumor ability and survival of mice when using denileukin diftitox to treat ovarian cancer [123]. In preclinical trials, using antibody-cytokine conjugates to send type 1 INF to the TME has resulted in an anti-tumor effect with no obvious toxicity [124,125,126].
FLT3L is a kind of cytokine manufactured by NK cells, which can effectively enhance DCs’ survival and proliferation [127]. A study shows that injecting FLT3L into mice can increase the number of CD8+ T cells and the ratio of CD8+ T cells to Treg cells in the TME, which result in the proliferation of cross-presenting DC and enhancement of the responses to intra-tumoral TLR3 agonists [128]. In a current trial, patients with advanced stage NHL were treated with the combination of FLT3L, a TLR3 agonist and localized radiotherapy, which has yielded an encouraging result [129].
Both IL-2 and IL-15 could bind with a CD122-CD132 receptor, thus treatment with IL-2 or IL-15 can induce the activation and extension of NK cells, CD8+ T cells, NKT cells and γδT cells [86]. However, IL-2 also has a high affinity to CD25-CD122-CD132 complex that is also presented on Treg cells. Thus, the reason why treatment with IL-2 results in a significant decrease in tumor in mouse models but yields a poor result in some patients with melanoma or renal cell cancer is that IL-2 promotes the proliferation of Treg cells [86,130]. IL-15 does not bind with CD25 or induce the expansion of the Treg cell population and therefore, shows better anti-tumor activity and lower toxicity than IL-2.

4. Challenges and Future Directions of Innate Immunotherapy

Although innate immunotherapy has significantly improved the treatment of tumors by using the body’s own defense mechanisms to successfully combat a variety of cancers, there are immune-related side effects that can impact several organ systems.
ICI therapies focus on common anti-tumor mechanisms to provide generalizability to a variety of cancers. The cardiovascular system is just one of the major organ systems that can experience ICI-related adverse effects. Studies show that ICI-related cardiotoxicity includes myocarditis, atherosclerosis and arrhythmias. Among them, the most well-known and most likely fatal is myocarditis, with a mortality rate of nearly 50% [131]. These side effects could be reduced with supportive care and immunosuppression with corticosteroids. While SIRP/CD47 inhibition causes the phagocytosis of tumors, red blood cells (RBC) are also prone to phagocytosis that is dose-dependent and may result in anemia and complications due to infusions [132]. When the Fc region of the antibody binds to the Fc receptor (FcγR) on macrophages, an antibody-dependent cellular phagocytosis (ADCP) is initiated. As noted above, blockade of CD47/SIRPα, the “don’t eat me” axis, promotes phagocytosis of tumor cells. CD47 is also widely expressed in normal human cells, especially on RBC, which leads to macrophages phagocytosis of RBC via ADCP, leading to anemia. Therefore, using Fc that cannot bind to FcγR or have a weak binding effect (such as IgG2 and IgG4) is an effective strategy to reduce anemia side effects. In addition, adjusting the dose of the drug to priming dosing and reducing the affinity of the antibody to some extent may also reduce the risk of anemia [84].
Adoptive new treatment with CAR immunotherapy has made significant progress. CAR immunotherapy for solid tumors, however, is far behind. Manufacturing of cells, a dearth of tumor-specific antigens and ineffective penetration into tumor locations are the principal obstacles to CAR immunotherapy in solid tumors [109]. Although the benefits of CAR NK cell treatment are clear, there are also substantial drawbacks. The current state of CAR NK manufacturing and storage, optimization of CAR NK structure, maintaining cytotoxic effects at the immunosuppressive TME and minimization of off-target toxicity are all areas of active research. Additionally, the half-life of NK cells is less than 10 days, which limits the off-target toxicity, but it may also represent a limitation in persistence, function and the durability of the response, meaning multiple administrations could be necessary to obtain a long-lasting reaction [133]. Currently, researchers are working to enhance the persistence of CAR NK cells in vivo. MyD88/CD40 are signaling molecules for NK cell activation and proliferation. In one study, inducible MyD88/CD40 (iMC) CAR NK cells were extended to CAR NK cells and enhanced the tumor killing efficacy of CAR-NK by increasing cytotoxic function, cytokine secretion and proliferation. The cytotoxicity and persistence of NK cells could also be tuned by IL-15. Autocrine IL-15 coupled with iMC could further drive CAR-NK cell proliferation and survival in vivo, providing a novel strategy to effectively improve the persistence of CAR NK cells [134]. Only one clinical trial for CAR macrophage has been started, and there have been no findings announced as yet. CAR M enables it to target tumor cells expressing specific antigens. The recognition of tumor antigens can induce several macrophage-mediated anticancer mechanisms: (1) triggering macrophage-mediated ADCC to kill tumor cells; (2) upon binding to specific tumor antigens, CAR receptors activate intracellular signaling pathways, induce phagocytosis and rapidly present tumor antigens to activate T cell-mediated immunity; (3) inhibiting pro-tumor polarization of macrophages and activating pro-inflammatory response pathways; (4) secreting pro-inflammatory cytokines such as interferon-gamma to recruit and activate other immune cells; (5) CAR macrophages continuously kill cancer cells, and they may also stimulate adaptive immune response, thus providing persistent anti-tumor immunity; (6) increasing the expression of MHC-I and MHC-II, thus enhancing T-cell activation; and (7) promoting the intratumoral infiltration of CD4+ and CD8+ T cells, NK cells and DCs, thereby activating a systemic immune response against solid tumors [135]. In TME of solid tumors, there are usually few T cells but abundant macrophages. This may make CAR M more efficient than CAR T and CAR NK when encountering a “cold” tumor, but still needs a long time before widespread clinical use [109].
Immunostimulatory cytokines are important to immune system regulation and exhibit significant promise as immunotherapeutic drugs. However, substantial toxicities brought by systemic immune activation have restricted their clinical usage. Fusions of cytokines with mAbs or antibody fragments have been designed in order to increase efficacy and decrease systemic toxicity [136,137]. Additionally, cytokine fusions with inhibitory moieties that can be broken down by tumor-associated proteases, lowering the cytokine’s affinity to improve targeting, and variations of oligomeric cytokines that are initiated by colocalization of monomeric subunits can all be used to ensure that cytokines are only activated at tumor sites [138,139,140]. A conditionally active cytokine system has been developed for monomeric cytokines such as IL-2 to prevent the persistent off-target activity that is typically seen with systemic cytokine and immune-cytokine treatment [141]. There is controversy around the usage of GM-CSF as an immunoadjuvant because studies suggest that using it as an adjuvant could result in immunosuppression rather than immune stimulation [70,142,143]. Even though GM-CSF has a long history and a large body of clinical evidence demonstrating its safety and effectiveness as an immune adjuvant, it is important to carefully evaluate the studies previously mentioned and take into account the potential mechanisms of GM-CSF-induced immunosuppression as suggested above.

5. Conclusions

In this review, we discussed in depth the roles and mechanisms of innate immunity in the TME and immunotherapies targeting the innate immune system, which include innate ICIs, innate immune cells engineered with CAR and cytokines. Then we discussed the challenges and future strategies of innate immunotherapy. Although innate immunity brings new opportunities for oncology treatment, there are still challenges such as inadequate eradication rate, toxic side effects and the potential need for repeated dosing, which means we still need to further investigate how to adequately use these innate immune cells in cancer immunotherapy. From multiple perspectives, multi-strategy combination therapy is the way forward for the treatment of tumors.

Author Contributions

D.J., S.W. and X.Z. design and proofread the concept. Y.Z., W.X., C.X., Y.N., X.Z. and S.M. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (32200745, 82073752, 81773620 and 82002915), the Shanghai Sailing Program (21YF1401900, 20YF1402800) and the Scientific and Innovative Action Plan of Shanghai (20S11904700, 20JC1411000 and 20S11906100).

Data Availability Statement

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

Conflicts of Interest

Shuang Mei is the chief executive officer of Shanghai Tinova Immune Therapeutics Co., Ltd. The other authors declare no conflicts of interest.

References

  1. Anwar, S.; Almatroudi, A.; Alsahli, A.M.; Khan, A.M.; Khan, A.A.; Rahmani, H.A. Natural Products: Implication in Cancer Prevention and Treatment through Modulating Various Biological Activities. Anti-Cancer Agents Med. Chem. 2020, 20, 2025–2040. [Google Scholar] [CrossRef] [PubMed]
  2. Chinnadurai, R.K.; Khan, N.; Meghwanshi, G.K.; Ponne, S.; Althobiti, M.; Kumar, R. Current research status of anti-cancer peptides: Mechanism of action, production, and clinical applications. Biomed. Pharmacother. 2023, 164, 114996. [Google Scholar] [CrossRef] [PubMed]
  3. Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T.; et al. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021, 6, 201. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef]
  5. Seager, R.J.; Hajal, C.; Spill, F.; Kamm, R.D.; Zaman, M.H. Dynamic interplay between tumour, stroma and immune system can drive or prevent tumour progression. Converg. Sci. Phys. Oncol. 2017, 3, 034002. [Google Scholar] [CrossRef]
  6. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [Green Version]
  7. Vesely, M.D.; Kershaw, M.H.; Schreiber, R.D.; Smyth, M.J. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 2011, 29, 235–271. [Google Scholar] [CrossRef] [Green Version]
  8. Alderton, G.K.; Bordon, Y. Tumour immunotherapy—Leukocytes take up the fight. Nat. Rev. Immunol. 2012, 12, 237. [Google Scholar] [CrossRef]
  9. Yuen, G.J.; Demissie, E.; Pillai, S. B lymphocytes and cancer: A love-hate relationship. Trends Cancer 2016, 2, 747–757. [Google Scholar] [CrossRef] [Green Version]
  10. Ohaegbulam, K.C.; Assal, A.; Lazar-Molnar, E.; Yao, Y.; Zang, X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol. Med. 2015, 21, 24–33. [Google Scholar] [CrossRef] [Green Version]
  11. Liu, Y.; Zhou, Z.; Hou, J.; Xiong, W.; Kim, H.; Chen, J.; Zheng, C.; Jiang, X.; Yoon, J.; Shen, J. Tumor Selective Metabolic Reprogramming as a Prospective PD-L1 Depression Strategy to Reactivate Immunotherapy. Adv. Mater. 2022, 34, 2206121. [Google Scholar] [CrossRef]
  12. Lim, W.A.; June, C.H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168, 724–740. [Google Scholar] [CrossRef] [Green Version]
  13. Li, X.; Song, W.; Shao, C.; Shi, Y.; Han, W. Emerging predictors of the response to the blockade of immune checkpoints in cancer therapy. Cell. Mol. Immunol. 2019, 16, 28–39. [Google Scholar] [CrossRef] [Green Version]
  14. Wei, J.; Guo, Y.; Wang, Y.; Wu, Z.; Bo, J.; Zhang, B.; Zhu, J.; Han, W. Clinical development of CAR T cell therapy in China: 2020 update. Cell. Mol. Immunol. 2021, 18, 792–804. [Google Scholar] [CrossRef]
  15. Rahmani, A.H.; Anwar, S.; Raut, R.; Almatroudi, A.; Babiker, A.Y.; Khan, A.A.; Alsahli, M.A.; Almatroodi, S.A. Therapeutic Potential of Myrrh, a Natural Resin, in Health Management through Modulation of Oxidative Stress, Inflammation, and Advanced Glycation End Products Formation Using In Vitro and In Silico Analysis. Appl. Sci. 2022, 12, 9175. [Google Scholar] [CrossRef]
  16. Liu, S.W.; Song, W.J.; Ma, G.K.; Wang, H.; Yang, L. Pyroptosis and its role in cancer. World J. Clin. Cases 2023, 11, 2386–2395. [Google Scholar] [CrossRef]
  17. Xu, X.; Fan, H.; Yang, Y.; Yao, S.; Yu, W.; Guo, Z.; Tan, W. Virus-Like Particle-Induced cGAS-STING Activation and AIM2 Inflammasome-Mediated Pyroptosis for Robust Cancer Immunotherapy. Angew. Chem. Int. Ed. Engl. 2023, 62, e202303010. [Google Scholar] [CrossRef]
  18. Li, X.; Dai, H.; Wang, H.; Han, W. Exploring innate immunity in cancer immunotherapy: Opportunities and challenges. Cell. Mol. Immunol. 2021, 18, 1607–1609. [Google Scholar] [CrossRef]
  19. Broz, M.L.; Binnewies, M.; Boldajipour, B.; Nelson, A.E.; Pollack, J.L.; Erle, D.J.; Barczak, A.; Rosenblum, M.D.; Daud, A.; Barber, D.L.; et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014, 26, 638–652. [Google Scholar] [CrossRef] [Green Version]
  20. Matsushita, H.; Vesely, M.D.; Koboldt, D.C.; Rickert, C.G.; Uppaluri, R.; Magrini, V.J.; Arthur, C.D.; White, J.M.; Chen, Y.S.; Shea, L.K.; et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012, 482, 400–404. [Google Scholar] [CrossRef] [Green Version]
  21. Muntjewerff, E.M.; Meesters, L.D.; van den Bogaart, G. Antigen Cross-Presentation by Macrophages. Front. Immunol. 2020, 11, 1276. [Google Scholar] [CrossRef] [PubMed]
  22. Furumaya, C.; Martinez-Sanz, P.; Bouti, P.; Kuijpers, T.W.; Matlung, H.L. Plasticity in Pro- and Anti-tumor Activity of Neutrophils: Shifting the Balance. Front. Immunol. 2020, 11, 2100. [Google Scholar] [CrossRef]
  23. Chen, D.; Zhang, X.; Li, Z.; Zhu, B. Metabolic regulatory crosstalk between tumor microenvironment and tumor-associated macrophages. Theranostics 2021, 11, 1016–1030. [Google Scholar] [CrossRef] [PubMed]
  24. Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef] [PubMed]
  25. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
  26. Dolcetti, L.; Peranzoni, E.; Ugel, S.; Marigo, I.; Fernandez Gomez, A.; Mesa, C.; Geilich, M.; Winkels, G.; Traggiai, E.; Casati, A.; et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur. J. Immunol. 2010, 40, 22–35. [Google Scholar] [CrossRef]
  27. Hegde, S.; Leader, A.M.; Merad, M. MDSC: Markers, development, states, and unaddressed complexity. Immunity 2021, 54, 875–884. [Google Scholar] [CrossRef]
  28. Zhou, S.; Zhao, Z.; Zhong, H.; Ren, Z.; Li, Y.; Wang, H.; Qiu, Y. The role of myeloid-derived suppressor cells in liver cancer. Discov. Oncol. 2023, 14, 77. [Google Scholar] [CrossRef]
  29. Li, K.; Shi, H.; Zhang, B.; Ou, X.; Ma, Q.; Chen, Y.; Shu, P.; Li, D.; Wang, Y. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct. Target. Ther. 2021, 6, 362. [Google Scholar] [CrossRef]
  30. Van Vlerken-Ysla, L.; Tyurina, Y.Y.; Kagan, V.E.; Gabrilovich, D.I. Functional states of myeloid cells in cancer. Cancer Cell 2023, 41, 490–504. [Google Scholar] [CrossRef]
  31. Mohamed, A.G.; Salome, V.I.; Abdelmetalab, F.T.; Youssef, E.; Hanh, H.L.; Matthew, J.D.; Ali, H.E.-B.; Dorota, W.; Ilyes, A.B.; Luis Del, V.; et al. Targeting PARP-1 with metronomic therapy modulates MDSC suppressive function and enhances anti-PD-1 immunotherapy in colon cancer. J. Immunother. Cancer 2021, 9, e001643. [Google Scholar] [CrossRef]
  32. Francesco De, S.; Alessia, L.; Federico, B.; Chiara, M.; Simone, C.; Rosalinda, T.; Alessandra, F.; Cristina, F.; Cristina, A.; Ornella, P.; et al. Interrupting the nitrosative stress fuels tumor-specific cytotoxic T lymphocytes in pancreatic cancer. J. Immunother. Cancer 2022, 10, e003549. [Google Scholar] [CrossRef]
  33. Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef] [Green Version]
  34. Lavin, Y.; Merad, M. Macrophages: Gatekeepers of tissue integrity. Cancer Immunol. Res. 2013, 1, 201–209. [Google Scholar] [CrossRef] [Green Version]
  35. Anderson, N.R.; Minutolo, N.G.; Gill, S.; Klichinsky, M. Macrophage-Based Approaches for Cancer Immunotherapy. Cancer Res. 2021, 81, 1201–1208. [Google Scholar] [CrossRef]
  36. Zhou, J.; Tang, Z.; Gao, S.; Li, C.; Feng, Y.; Zhou, X. Tumor-Associated Macrophages: Recent Insights and Therapies. Front. Oncol. 2020, 10, 188. [Google Scholar] [CrossRef]
  37. Biswas, S.K.; Gangi, L.; Paul, S.; Schioppa, T.; Saccani, A.; Sironi, M.; Bottazzi, B.; Doni, A.; Vincenzo, B.; Pasqualini, F.; et al. A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation). Blood 2006, 107, 2112–2122. [Google Scholar] [CrossRef] [Green Version]
  38. Saccani, A.; Schioppa, T.; Porta, C.; Biswas, S.K.; Nebuloni, M.; Vago, L.; Bottazzi, B.; Colombo, M.P.; Mantovani, A.; Sica, A. p50 nuclear factor-kappaB overexpression in tumor-associated macrophages inhibits M1 inflammatory responses and antitumor resistance. Cancer Res. 2006, 66, 11432–11440. [Google Scholar] [CrossRef] [Green Version]
  39. Leach, J.; Morton, J.P.; Sansom, O.J. Neutrophils: Homing in on the myeloid mechanisms of metastasis. Mol. Immunol. 2019, 110, 69–76. [Google Scholar] [CrossRef]
  40. Dumitru, C.A.; Lang, S.; Brandau, S. Modulation of neutrophil granulocytes in the tumor microenvironment: Mechanisms and consequences for tumor progression. Semin. Cancer Biol. 2013, 23, 141–148. [Google Scholar] [CrossRef]
  41. Benson, D.D.; Meng, X.; Fullerton, D.A.; Moore, E.E.; Lee, J.H.; Ao, L.; Silliman, C.C.; Barnett, C.C., Jr. Activation state of stromal inflammatory cells in murine metastatic pancreatic adenocarcinoma. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2012, 302, R1067-75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yu, P.F.; Huang, Y.; Han, Y.Y.; Lin, L.Y.; Sun, W.H.; Rabson, A.B.; Wang, Y.; Shi, Y.F. TNFα-activated mesenchymal stromal cells promote breast cancer metastasis by recruiting CXCR2+ neutrophils. Oncogene 2017, 36, 482–490. [Google Scholar] [CrossRef] [PubMed]
  43. Noh, H.; Eomm, M.; Han, A. Usefulness of Pretreatment Neutrophil to Lymphocyte Ratio in Predicting Disease-Specific Survival in Breast Cancer Patients. J. Breast Cancer 2013, 16, 55–59. [Google Scholar] [CrossRef] [Green Version]
  44. Azab, B.; Bhatt, V.R.; Phookan, J.; Murukutla, S.; Kohn, N.; Terjanian, T.; Widmann, W.D. Usefulness of the Neutrophil-to-Lymphocyte Ratio in Predicting Short- and Long-Term Mortality in Breast Cancer Patients. Ann. Surg. Oncol. 2012, 19, 217–224. [Google Scholar] [CrossRef] [PubMed]
  45. Han, Y.; Yu, Z.; Wen, S.; Zhang, B.; Cao, X.; Wang, X. Prognostic value of chemotherapy-induced neutropenia in early-stage breast cancer. Breast Cancer Res. Treat. 2012, 131, 483–490. [Google Scholar] [CrossRef]
  46. Chen, W.-C.; Lai, Y.-H.; Chen, H.-Y.; Guo, H.-R.; Su, I.-J.; Chen, H.H.W. Interleukin-17-producing cell infiltration in the breast cancer tumour microenvironment is a poor prognostic factor. Histopathology 2013, 63, 225–233. [Google Scholar] [CrossRef]
  47. Novitskiy, S.V.; Pickup, M.W.; Gorska, A.E.; Owens, P.; Chytil, A.; Aakre, M.; Wu, H.; Shyr, Y.; Moses, H.L. TGF-β Receptor II Loss Promotes Mammary Carcinoma Progression by Th17-Dependent Mechanisms. Cancer Discov. 2011, 1, 430–441. [Google Scholar] [CrossRef] [Green Version]
  48. Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.-S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.J.A.C.; Jonkers, J.; et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef] [Green Version]
  49. Karre, K. NK cells, MHC class I molecules and the missing self. Scand. J. Immunol. 2002, 55, 221–228. [Google Scholar] [CrossRef]
  50. Castro, F.; Cardoso, A.P.; Goncalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front. Immunol. 2018, 9, 847. [Google Scholar] [CrossRef] [Green Version]
  51. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
  52. Wang, Y.; Xiang, Y.; Xin, V.W.; Wang, X.-W.; Peng, X.-C.; Liu, X.-Q.; Wang, D.; Li, N.; Cheng, J.-T.; Lyv, Y.-N.; et al. Dendritic cell biology and its role in tumor immunotherapy. J. Hematol. Oncol. 2020, 13, 107. [Google Scholar] [CrossRef]
  53. Binnewies, M.; Mujal, A.M.; Pollack, J.L.; Combes, A.J.; Hardison, E.A.; Barry, K.C.; Tsui, J.; Ruhland, M.K.; Kersten, K.; Abushawish, M.A.; et al. Unleashing Type-2 Dendritic Cells to Drive Protective Antitumor CD4(+) T Cell Immunity. Cell 2019, 177, 556–571.e516. [Google Scholar] [CrossRef]
  54. Salio, M.; Palmowski, M.J.; Atzberger, A.; Hermans, I.F.; Cerundolo, V. CpG-matured murine plasmacytoid dendritic cells are capable of in vivo priming of functional CD8 T cell responses to endogenous but not exogenous antigens. J. Exp. Med. 2004, 199, 567–579. [Google Scholar] [CrossRef] [Green Version]
  55. Tel, J.; Aarntzen, E.H.; Baba, T.; Schreibelt, G.; Schulte, B.M.; Benitez-Ribas, D.; Boerman, O.C.; Croockewit, S.; Oyen, W.J.; van Rossum, M.; et al. Natural human plasmacytoid dendritic cells induce antigen-specific T-cell responses in melanoma patients. Cancer Res. 2013, 73, 1063–1075. [Google Scholar] [CrossRef] [Green Version]
  56. Chiang, M.C.; Tullett, K.M.; Lee, Y.S.; Idris, A.; Ding, Y.; McDonald, K.J.; Kassianos, A.; Leal Rojas, I.M.; Jeet, V.; Lahoud, M.H.; et al. Differential uptake and cross-presentation of soluble and necrotic cell antigen by human DC subsets. Eur. J. Immunol. 2016, 46, 329–339. [Google Scholar] [CrossRef]
  57. Russo, E.; Teijeira, A.; Vaahtomeri, K.; Willrodt, A.-H.; Bloch, J.S.; Nitschké, M.; Santambrogio, L.; Kerjaschki, D.; Sixt, M.; Halin, C. Intralymphatic CCL21 Promotes Tissue Egress of Dendritic Cells through Afferent Lymphatic Vessels. Cell Rep. 2016, 14, 1723–1734. [Google Scholar] [CrossRef] [Green Version]
  58. Spranger, S.; Dai, D.; Horton, B.; Gajewski, T.F. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017, 31, 711–723 e714. [Google Scholar] [CrossRef] [Green Version]
  59. Martinez-Lopez, M.; Iborra, S.; Conde-Garrosa, R.; Sancho, D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur. J. Immunol. 2015, 45, 119–129. [Google Scholar] [CrossRef]
  60. 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]
  61. Parker, B.S.; Rautela, J.; Hertzog, P.J. Antitumour actions of interferons: Implications for cancer therapy. Nat. Rev. Cancer 2016, 16, 131–144. [Google Scholar] [CrossRef] [PubMed]
  62. 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]
  63. Lukic, A.; Wahlund, C.J.E.; Gómez, C.; Brodin, D.; Samuelsson, B.; Wheelock, C.E.; Gabrielsson, S.; Rådmark, O. Exosomes and cells from lung cancer pleural exudates transform LTC4 to LTD4, promoting cell migration and survival via CysLT1. Cancer Lett. 2019, 444, 1–8. [Google Scholar] [CrossRef] [PubMed]
  64. Lukic, A.; Ji, J.; Idborg, H.; Samuelsson, B.; Palmberg, L.; Gabrielsson, S.; Rådmark, O. Pulmonary epithelial cancer cells and their exosomes metabolize myeloid cell-derived leukotriene C4 to leukotriene D4. J. Lipid Res. 2016, 57, 1659–1669. [Google Scholar] [CrossRef] [Green Version]
  65. Zhao, S.; Yao, K.; Li, D.; Liu, K.; Jin, G.; Yan, M.; Wu, Q.; Chen, H.; Shin, S.H.; Bai, R.; et al. Inhibition of LTA4H by bestatin in human and mouse colorectal cancer. eBioMedicine 2019, 44, 361–374. [Google Scholar] [CrossRef]
  66. Trabanelli, S.; Chevalier, M.F.; Martinez-Usatorre, A.; Gomez-Cadena, A.; Salome, B.; Lecciso, M.; Salvestrini, V.; Verdeil, G.; Racle, J.; Papayannidis, C.; et al. Tumour-derived PGD2 and NKp30-B7H6 engagement drives an immunosuppressive ILC2-MDSC axis. Nat. Commun. 2017, 8, 593. [Google Scholar] [CrossRef] [Green Version]
  67. Zhang, B.; Bie, Q.; Wu, P.; Zhang, J.; You, B.; Shi, H.; Qian, H.; Xu, W. PGD2/PTGDR2 Signaling Restricts the Self-Renewal and Tumorigenesis of Gastric Cancer. Stem Cells 2018, 36, 990–1003. [Google Scholar] [CrossRef] [Green Version]
  68. Grauers Wiktorin, H.; Nilsson, M.S.; Kiffin, R.; Sander, F.E.; Lenox, B.; Rydstrom, A.; Hellstrand, K.; Martner, A. Histamine targets myeloid-derived suppressor cells and improves the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade. Cancer Immunol. Immunother. 2019, 68, 163–174. [Google Scholar] [CrossRef] [Green Version]
  69. Spits, H.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate lymphoid cells—A proposal for uniform nomenclature. Nat. Rev. Immunol. 2013, 13, 145–149. [Google Scholar] [CrossRef]
  70. Parmiani, G.; Castelli, C.; Pilla, L.; Santinami, M.; Colombo, M.P.; Rivoltini, L. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann. Oncol. 2007, 18, 226–232. [Google Scholar] [CrossRef]
  71. Jacquelot, N.; Seillet, C.; Vivier, E.; Belz, G.T. Innate lymphoid cells and cancer. Nat. Immunol. 2022, 23, 371–379. [Google Scholar] [CrossRef]
  72. Cuff, A.O.; Sillito, F.; Dertschnig, S.; Hall, A.; Luong, T.V.; Chakraverty, R.; Male, V. The Obese Liver Environment Mediates Conversion of NK Cells to a Less Cytotoxic ILC1-Like Phenotype. Front. Immunol. 2019, 10, 2180. [Google Scholar] [CrossRef] [Green Version]
  73. Ricardo-Gonzalez, R.R.; Van Dyken, S.J.; Schneider, C.; Lee, J.; Nussbaum, J.C.; Liang, H.-E.; Vaka, D.; Eckalbar, W.L.; Molofsky, A.B.; Erle, D.J.; et al. Tissue signals imprint ILC2 identity with anticipatory function. Nat. Immunol. 2018, 19, 1093–1099. [Google Scholar] [CrossRef]
  74. Kim, J.; Kim, W.; Moon, U.J.; Kim, H.J.; Choi, H.J.; Sin, J.I.; Park, N.H.; Cho, H.R.; Kwon, B. Intratumorally Establishing Type 2 Innate Lymphoid Cells Blocks Tumor Growth. J. Immunol. 2016, 196, 2410–2423. [Google Scholar] [CrossRef] [Green Version]
  75. Jacquelot, N.; Seillet, C.; Wang, M.; Pizzolla, A.; Liao, Y.; Hediyeh-Zadeh, S.; Grisaru-Tal, S.; Louis, C.; Huang, Q.; Schreuder, J.; et al. Blockade of the co-inhibitory molecule PD-1 unleashes ILC2-dependent antitumor immunity in melanoma. Nat. Immunol. 2021, 22, 851–864. [Google Scholar] [CrossRef]
  76. Wang, S.; Qu, Y.; Xia, P.; Chen, Y.; Zhu, X.; Zhang, J.; Wang, G.; Tian, Y.; Ying, J.; Fan, Z. Transdifferentiation of tumor infiltrating innate lymphoid cells during progression of colorectal cancer. Cell Res. 2020, 30, 610–622. [Google Scholar] [CrossRef]
  77. Kirchberger, S.; Royston, D.J.; Boulard, O.; Thornton, E.; Franchini, F.; Szabady, R.L.; Harrison, O.; Powrie, F. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J. Exp. Med. 2013, 210, 917–931. [Google Scholar] [CrossRef]
  78. Chan, I.H.; Jain, R.; Tessmer, M.S.; Gorman, D.; Mangadu, R.; Sathe, M.; Vives, F.; Moon, C.; Penaflor, E.; Turner, S.; et al. Interleukin-23 is sufficient to induce rapid de novo gut tumorigenesis, independent of carcinogens, through activation of innate lymphoid cells. Mucosal Immunol. 2014, 7, 842–856. [Google Scholar] [CrossRef]
  79. Yang, Y.H.; Liu, J.W.; Lu, C.; Wei, J.F. CAR-T Cell Therapy for Breast Cancer: From Basic Research to Clinical Application. Int. J. Biol. Sci. 2022, 18, 2609–2626. [Google Scholar] [CrossRef]
  80. Peng, M.; Mo, Y.; Wang, Y.; Wu, P.; Zhang, Y.; Xiong, F.; Guo, C.; Wu, X.; Li, Y.; Li, X.; et al. Neoantigen vaccine: An emerging tumor immunotherapy. Mol. Cancer 2019, 18, 128. [Google Scholar] [CrossRef] [Green Version]
  81. Zhou, F.; Qiao, M.; Zhou, C. The cutting-edge progress of immune-checkpoint blockade in lung cancer. Cell Mol. Immunol. 2021, 18, 279–293. [Google Scholar] [CrossRef] [PubMed]
  82. Schumacher, T.N.; Kesmir, C.; van Buuren, M.M. Biomarkers in cancer immunotherapy. Cancer Cell 2015, 27, 12–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef] [PubMed]
  84. Lentz, R.W.; Colton, M.D.; Mitra, S.S.; Messersmith, W.A. Innate Immune Checkpoint Inhibitors: The Next Breakthrough in Medical Oncology? Mol. Cancer Ther. 2021, 20, 961–974. [Google Scholar] [CrossRef] [PubMed]
  85. Lin, C.; Zhang, J. Chimeric antigen receptor engineered innate immune cells in cancer immunotherapy. Sci. China Life Sci. 2019, 62, 633–639. [Google Scholar] [CrossRef]
  86. Demaria, O.; Cornen, S.; Daëron, M.; Morel, Y.; Medzhitov, R.; Vivier, E. Harnessing innate immunity in cancer therapy. Nature 2019, 574, 45–56. [Google Scholar] [CrossRef]
  87. Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667. [Google Scholar] [CrossRef]
  88. Jia, X.; Yan, B.; Tian, X.; Liu, Q.; Jin, J.; Shi, J.; Hou, Y. CD47/SIRPalpha pathway mediates cancer immune escape and immunotherapy. Int. J. Biol. Sci. 2021, 17, 3281–3287. [Google Scholar] [CrossRef]
  89. Huang, Y.; Ma, Y.; Gao, P.; Yao, Z. Targeting CD47: The achievements and concerns of current studies on cancer immunotherapy. J. Thorac. Dis. 2017, 9, E168–E174. [Google Scholar] [CrossRef] [Green Version]
  90. Folkes, A.S.; Feng, M.; Zain, J.M.; Abdulla, F.; Rosen, S.T.; Querfeld, C. Targeting CD47 as a cancer therapeutic strategy: The cutaneous T-cell lymphoma experience. Curr. Opin. Oncol. 2018, 30, 332–337. [Google Scholar] [CrossRef]
  91. Querfeld, C.; Thompson, J.; Taylor, M.; Pillai, R.; Johnson, L.D.S.; Catalano, T.; Petrova, P.S.; Uger, R.A.; Irwin, M.; Sievers, E.L.; et al. A Single Direct Intratumoral Injection of TTI-621 (SIRPαFc) Induces Antitumor Activity in Patients with Relapsed/Refractory Mycosis Fungoides and Sézary Syndrome: Preliminary Findings Employing an Immune Checkpoint Inhibitor Blocking the CD47 “Do Not Eat” Signal. Blood 2017, 130, 4076. [Google Scholar] [CrossRef]
  92. Hodgins, J.J.; Khan, S.T.; Park, M.M.; Auer, R.C.; Ardolino, M. Killers 2.0: NK cell therapies at the forefront of cancer control. J. Clin. Investig. 2019, 129, 3499–3510. [Google Scholar] [CrossRef] [Green Version]
  93. Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018, 19, 723–732. [Google Scholar] [CrossRef]
  94. Whelan, S.; Ophir, E.; Kotturi, M.F.; Levy, O.; Ganguly, S.; Leung, L.; Vaknin, I.; Kumar, S.; Dassa, L.; Hansen, K.; et al. PVRIG and PVRL2 Are Induced in Cancer and Inhibit CD8(+) T-cell Function. Cancer Immunol. Res. 2019, 7, 257–268. [Google Scholar] [CrossRef] [Green Version]
  95. Harjunpaa, H.; Guillerey, C. TIGIT as an emerging immune checkpoint. Clin. Exp. Immunol. 2020, 200, 108–119. [Google Scholar] [CrossRef] [Green Version]
  96. Sanchez-Correa, B.; Valhondo, I.; Hassouneh, F.; Lopez-Sejas, N.; Pera, A.; Bergua, J.M.; Arcos, M.J.; Banas, H.; Casas-Aviles, I.; Duran, E.; et al. DNAM-1 and the TIGIT/PVRIG/TACTILE Axis: Novel Immune Checkpoints for Natural Killer Cell-Based Cancer Immunotherapy. Cancers 2019, 11, 877. [Google Scholar] [CrossRef] [Green Version]
  97. Xu, F.; Sunderland, A.; Zhou, Y.; Schulick, R.D.; Edil, B.H.; Zhu, Y. Blockade of CD112R and TIGIT signaling sensitizes human natural killer cell functions. Cancer Immunol. Immunother. 2017, 66, 1367–1375. [Google Scholar] [CrossRef]
  98. Feng, M.; Jiang, W.; Kim, B.Y.S.; Zhang, C.C.; Fu, Y.X.; Weissman, I.L. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 568–586. [Google Scholar] [CrossRef]
  99. Yu, T.; Gan, S.; Zhu, Q.; Dai, D.; Li, N.; Wang, H.; Chen, X.; Hou, D.; Wang, Y.; Pan, Q.; et al. Modulation of M2 macrophage polarization by the crosstalk between Stat6 and Trim24. Nat. Commun. 2019, 10, 4353. [Google Scholar] [CrossRef] [Green Version]
  100. Chen, H.M.; van der Touw, W.; Wang, Y.S.; Kang, K.; Mai, S.; Zhang, J.; Alsina-Beauchamp, D.; Duty, J.A.; Mungamuri, S.K.; Zhang, B.; et al. Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J. Clin. Investig. 2018, 128, 5647–5662. [Google Scholar] [CrossRef] [Green Version]
  101. Chiossone, L.; Dumas, P.Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef] [PubMed]
  102. Andre, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Blery, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743.e1713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid–mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Dulal, D.; Boring, A.; Terrero, D.; Johnson, T.; Tiwari, A.K.; Raman, D. Tackling of Immunorefractory Tumors by Targeting Alternative Immune Checkpoints. Cancers 2023, 15, 2774. [Google Scholar] [CrossRef]
  105. Naimi, A.; Mohammed, R.N.; Raji, A.; Chupradit, S.; Yumashev, A.V.; Suksatan, W.; Shalaby, M.N.; Thangavelu, L.; Kamrava, S.; Shomali, N.; et al. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun. Signal. 2022, 20, 44. [Google Scholar] [CrossRef]
  106. Xu, C.; Ju, D.; Zhang, X. Chimeric antigen receptor T-cell therapy: Challenges and opportunities in lung cancer. Antib. Ther. 2022, 5, 73–83. [Google Scholar] [CrossRef]
  107. Yilmaz, A.; Cui, H.; Caligiuri, M.A.; Yu, J. Chimeric antigen receptor-engineered natural killer cells for cancer immunotherapy. J. Hematol. Oncol. 2020, 13, 168. [Google Scholar] [CrossRef]
  108. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E.; et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef]
  109. Pan, K.; Farrukh, H.; Chittepu, V.; Xu, H.; Pan, C.X.; Zhu, Z. CAR race to cancer immunotherapy: From CAR T, CAR NK to CAR macrophage therapy. J. Exp. Clin. Cancer Res. 2022, 41, 119. [Google Scholar] [CrossRef]
  110. Bollino, D.; Webb, T.J. Chimeric antigen receptor-engineered natural killer and natural killer T cells for cancer immunotherapy. Transl. Res. 2017, 187, 32–43. [Google Scholar] [CrossRef]
  111. Heczey, A.; Liu, D.; Tian, G.; Courtney, A.N.; Wei, J.; Marinova, E.; Gao, X.; Guo, L.; Yvon, E.; Hicks, J.; et al. Invariant NKT cells with chimeric antigen receptor provide a novel platform for safe and effective cancer immunotherapy. Blood 2014, 124, 2824–2833. [Google Scholar] [CrossRef] [Green Version]
  112. Tian, G.; Courtney, A.N.; Jena, B.; Heczey, A.; Liu, D.; Marinova, E.; Guo, L.; Xu, X.; Torikai, H.; Mo, Q.; et al. CD62L+ NKT cells have prolonged persistence and antitumor activity in vivo. J. Clin. Investig. 2016, 126, 2341–2355. [Google Scholar] [CrossRef] [Green Version]
  113. Morandi, F.; Yazdanifar, M.; Cocco, C.; Bertaina, A.; Airoldi, I. Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on gammadelta T and NK Cells. Cells 2020, 9, 1757. [Google Scholar] [CrossRef]
  114. Rozenbaum, M.; Meir, A.; Aharony, Y.; Itzhaki, O.; Schachter, J.; Bank, I.; Jacoby, E.; Besser, M.J. Gamma-Delta CAR-T Cells Show CAR-Directed and Independent Activity Against Leukemia. Front. Immunol. 2020, 11, 1347. [Google Scholar] [CrossRef]
  115. Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
  116. Song, X.; Peng, Y.; Wang, X.; Chen, Q.; Lan, X.; Shi, F. The stimulator of interferon genes (STING) agonists for treating acute myeloid leukemia (AML): Current knowledge and future outlook. Clin. Transl. Oncol. 2023, 25, 1545–1553. [Google Scholar] [CrossRef]
  117. Pan, X.; Zhang, W.; Guo, H.; Wang, L.; Wu, H.; Ding, L.; Yang, B. Strategies involving STING pathway activation for cancer immunotherapy: Mechanism and agonists. Biochem. Pharmacol. 2023, 213, 115596. [Google Scholar] [CrossRef]
  118. Kwon, J.; Bakhoum, S.F. The Cytosolic DNA-Sensing cGAS-STING Pathway in Cancer. Cancer Discov. 2020, 10, 26–39. [Google Scholar] [CrossRef]
  119. Hopfner, K.-P.; Hornung, V. Molecular mechanisms and cellular functions of cGAS–STING signalling. Nat. Rev. Mol. Cell Biol. 2020, 21, 501–521. [Google Scholar] [CrossRef]
  120. Zitvogel, L.; Galluzzi, L.; Kepp, O.; Smyth, M.J.; Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 2015, 15, 405–414. [Google Scholar] [CrossRef]
  121. Wennerberg, E.; Kremer, V.; Childs, R.; Lundqvist, A. CXCL10-induced migration of adoptively transferred human natural killer cells toward solid tumors causes regression of tumor growth in vivo. Cancer Immunol. Immunother. 2015, 64, 225–235. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, W.; Li, Y.; Xin, S.; Yang, L.; Jiang, M.; Xin, Y.; Wang, Y.; Cao, P.; Zhang, S.; Yang, Y.; et al. The emerging roles of IFIT3 in antiviral innate immunity and cellular biology. J. Med. Virol. 2023, 95, e28259. [Google Scholar] [CrossRef] [PubMed]
  123. Thibodeaux, S.R.; Barnett, B.B.; Pandeswara, S.; Wall, S.R.; Hurez, V.; Dao, V.; Sun, L.; Daniel, B.J.; Brumlik, M.J.; Drerup, J.; et al. IFNalpha Augments Clinical Efficacy of Regulatory T-cell Depletion with Denileukin Diftitox in Ovarian Cancer. Clin. Cancer Res. 2021, 27, 3661–3673. [Google Scholar] [CrossRef] [PubMed]
  124. Huang, T.H.; Chintalacharuvu, K.R.; Morrison, S.L. Targeting IFN-alpha to B cell lymphoma by a tumor-specific antibody elicits potent antitumor activities. J. Immunol. 2007, 179, 6881–6888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Rossi, E.A.; Rossi, D.L.; Cardillo, T.M.; Stein, R.; Goldenberg, D.M.; Chang, C.H. Preclinical studies on targeted delivery of multiple IFNalpha2b to HLA-DR in diverse hematologic cancers. Blood 2011, 118, 1877–1884. [Google Scholar] [CrossRef] [Green Version]
  126. Yang, X.; Zhang, X.; Fu, M.L.; Weichselbaum, R.R.; Gajewski, T.F.; Guo, Y.; Fu, Y.X. Targeting the tumor microenvironment with interferon-beta bridges innate and adaptive immune responses. Cancer Cell 2014, 25, 37–48. [Google Scholar] [CrossRef] [Green Version]
  127. Cueto, F.J.; Sancho, D. The Flt3L/Flt3 Axis in Dendritic Cell Biology and Cancer Immunotherapy. Cancers 2021, 13, 1525. [Google Scholar] [CrossRef]
  128. Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and Activation of CD103(+) Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef] [Green Version]
  129. Hammerich, L.; Marron, T.U.; Upadhyay, R.; Svensson-Arvelund, J.; Dhainaut, M.; Hussein, S.; Zhan, Y.; Ostrowski, D.; Yellin, M.; Marsh, H.; et al. Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat. Med. 2019, 25, 814–824. [Google Scholar] [CrossRef]
  130. Sim, G.C.; Martin-Orozco, N.; Jin, L.; Yang, Y.; Wu, S.; Washington, E.; Sanders, D.; Lacey, C.; Wang, Y.; Vence, L.; et al. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J. Clin. Investig. 2014, 124, 99–110. [Google Scholar] [CrossRef] [Green Version]
  131. Stein-Merlob, A.F.; Rothberg, M.V.; Ribas, A.; Yang, E.H. Cardiotoxicities of novel cancer immunotherapies. Heart 2021, 107, 1694–1703. [Google Scholar] [CrossRef]
  132. Advani, R.; Flinn, I.; Popplewell, L.; Forero, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.; Roschewski, M.; LaCasce, A.; Collins, G.P.; et al. CD47 Blockade by Hu5F9-G4 and Rituximab in Non-Hodgkin’s Lymphoma. N. Engl. J. Med. 2018, 379, 1711–1721. [Google Scholar] [CrossRef]
  133. Zhang, Y.; Wallace, D.L.; de Lara, C.M.; Ghattas, H.; Asquith, B.; Worth, A.; Griffin, G.E.; Taylor, G.P.; Tough, D.F.; Beverley, P.C.; et al. In vivo kinetics of human natural killer cells: The effects of ageing and acute and chronic viral infection. Immunology 2007, 121, 258–265. [Google Scholar] [CrossRef]
  134. Wang, X.; Jasinski, D.L.; Medina, J.L.; Spencer, D.M.; Foster, A.E.; Bayle, J.H. Inducible MyD88/CD40 synergizes with IL-15 to enhance antitumor efficacy of CAR-NK cells. Blood Adv. 2020, 4, 1950–1964. [Google Scholar] [CrossRef]
  135. Mishra, A.K.; Malonia, S.K. Advancing cellular immunotherapy with macrophages. Life Sci. 2023, 328, 121857. [Google Scholar] [CrossRef]
  136. Hutmacher, C.; Neri, D. Antibody-cytokine fusion proteins: Biopharmaceuticals with immunomodulatory properties for cancer therapy. Adv. Drug Deliv. Rev. 2019, 141, 67–91. [Google Scholar] [CrossRef]
  137. Holder, P.G.; Lim, S.A.; Huang, C.S.; Sharma, P.; Dagdas, Y.S.; Bulutoglu, B.; Sockolosky, J.T. Engineering interferons and interleukins for cancer immunotherapy. Adv. Drug Deliv. Rev. 2022, 182, 114112. [Google Scholar] [CrossRef]
  138. Garcin, G.; Paul, F.; Staufenbiel, M.; Bordat, Y.; Van der Heyden, J.; Wilmes, S.; Cartron, G.; Apparailly, F.; De Koker, S.; Piehler, J.; et al. High efficiency cell-specific targeting of cytokine activity. Nat. Commun. 2014, 5, 3016. [Google Scholar] [CrossRef] [Green Version]
  139. Puskas, J.; Skrombolas, D.; Sedlacek, A.; Lord, E.; Sullivan, M.; Frelinger, J. Development of an attenuated interleukin-2 fusion protein that can be activated by tumour-expressed proteases. Immunology 2011, 133, 206–220. [Google Scholar] [CrossRef]
  140. Venetz, D.; Koovely, D.; Weder, B.; Neri, D. Targeted Reconstitution of Cytokine Activity upon Antigen Binding using Split Cytokine Antibody Fusion Proteins. J. Biol. Chem. 2016, 291, 18139–18147. [Google Scholar] [CrossRef] [Green Version]
  141. Quijano-Rubio, A.; Bhuiyan, A.M.; Yang, H.; Leung, I.; Bello, E.; Ali, L.R.; Zhangxu, K.; Perkins, J.; Chun, J.H.; Wang, W.; et al. A split, conditionally active mimetic of IL-2 reduces the toxicity of systemic cytokine therapy. Nat. Biotechnol. 2022, 41, 532–540. [Google Scholar] [CrossRef] [PubMed]
  142. Slingluff, C.L., Jr.; Petroni, G.R.; Olson, W.C.; Smolkin, M.E.; Ross, M.I.; Haas, N.B.; Grosh, W.W.; Boisvert, M.E.; Kirkwood, J.M.; Chianese-Bullock, K.A. Effect of granulocyte/macrophage colony-stimulating factor on circulating CD8+ and CD4+ T-cell responses to a multipeptide melanoma vaccine: Outcome of a multicenter randomized trial. Clin. Cancer Res. 2009, 15, 7036–7044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Faries, M.B.; Hsueh, E.C.; Ye, X.; Hoban, M.; Morton, D.L. Effect of granulocyte/macrophage colony-stimulating factor on vaccination with an allogeneic whole-cell melanoma vaccine. Clin. Cancer Res. 2009, 15, 7029–7035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The role of innate immune cells in tumor progression. Innate immune cells show multiple roles in tumor progression through antigen presentation, phagocytosis, secretion of cytokines, direct killing effect, etc.
Figure 1. The role of innate immune cells in tumor progression. Innate immune cells show multiple roles in tumor progression through antigen presentation, phagocytosis, secretion of cytokines, direct killing effect, etc.
Ijms 24 11233 g001
Figure 2. Innate immune cell-based immunotherapy. Innate immunotherapies include innate ICIs, CAR innate immune cells and cytokines. (A) innate immunity-based ICIs; (B) CAR innate immune cells; (C) cytokines in innate immunity.
Figure 2. Innate immune cell-based immunotherapy. Innate immunotherapies include innate ICIs, CAR innate immune cells and cytokines. (A) innate immunity-based ICIs; (B) CAR innate immune cells; (C) cytokines in innate immunity.
Ijms 24 11233 g002
Table 1. Selected clinical trials of innate immunity-based ICIs and engineered cellular immunotherapies.
Table 1. Selected clinical trials of innate immunity-based ICIs and engineered cellular immunotherapies.
TargetNCT NumberStatusPhaseCancer TypeInterventionsEnrollment
SIRPαNCT05076591RecruitingIAdvanced Breast Cancer, Advanced Gastric CancerIMM2902135
NCT05276310RecruitingIAdvanced CancerIMC-00224
NCT05192512RecruitingIAdvanced CancerTQB2928180
NCT05507541Not yet recruitingIIMultiple cancer typesTTI-622 and TTI-621 in combination with Pembrolizumab62
NCT05263271RecruitingIAML; MDSGentulizumab58
TIGIT&PVRIGNCT04570839RecruitingI/IIEndometrial Neoplasms, Ovarian Cancer, HNCCOM701 in combination with BMS-986207 and Nivolumab100
NCT03667716RecruitingIOvarian Cancer, Breast CancerCOM701 and Nivolumab140
LILRB2NCT04669899RecruitingI/IIMultiple cancer typesJTX-8064 281
NCT05054348RecruitingISolid TumorIO-108, Pembrolizumab 36
NKG2ANCT05162755RecruitingISolid TumorS095029129
NCT04914351Not yet recruitingISolid TumorHY-010232
NCT05414032Not yet recruitingIIAdvanced HNSCCMonalizumab, Cetuximab200
LAG-3NCT03252938RecruitingIPeritoneal CarcinomatosisIMP321 45
NCT05101109RecruitingIAdvanced Solid TumorABL50136
NCT05078593RecruitingISolid Tumor, LymphomaHLX2611
NCT04618393RecruitingI/IIAdvanced Solid TumorEMB-0243
NCT05577182Not yet recruitingIAdvanced MalignanciesINCA32459-101120
TIM-3NCT05357651Not yet recruitingISolid Tumor, LymphomaLB1410100
NCT02817633RecruitingIMultiple cancer typesTSR-022 369
NCT04370704RecruitingI/IIMelanomaINCAGN02385 in combination with INCAGN02390 and INCMGA00012146
NCT04931654RecruitingI/IINSCLCAZD778981
NCT05367401Not yet recruitingI/IIMDS, AMLSabatolimab, Magrolimab63
VISTANCT04475523RecruitingISolid TumorCI-899350
NCT05082610RecruitingINSCLCHMBD-002, Pembrolizumab240
CD32BNCT04219254RecruitingI/IISolid TumorBI-1206 in combination with Pembrolizumab90
NCT05555251RecruitingI/IIHER2-positive Breast Cancer, HER2-positive Gastric CancerBI-1607 in combination with Trastuzumab116
NCT03571568RecruitingI/IIIndolent B-Cell NHLBI1206 in combination with Rituximab30
CAR NKNCT05213195RecruitingIRefractory Metastatic CRCNKG2D CAR-NK cells38
NCT05507593RecruitingISCLCDLL3-CAR-NK cells18
NCT04662788Not yet recruiting IHematological MalignanciesNK cells/Combined Monoclonal Antibodies36
NCT05410717RecruitingI/IIOvarian cancer, Testis cancerClaudin6 targeting CAR-NK cells40
CAR MNCT04660929RecruitingIMultiple cancer typesCT-050818
CAR NKTNCT03294954RecruitingINeuroblastomaGD2 Specific CAR and IL-15 Expressing Autologous NKT Cells36
NCT03774654RecruitingIMultiple cancer typesCD19.CAR-aNKT cells48
NCT05487651Not yet recruitingINHL, BCL, DLBCLKUR-50236
AML: acute myelocytic leukemia; MDS: myelodysplastic syndrome; HNSCC: head and neck squamous cell carcinoma; HNC: head and neck cancer; CRC: colorectal adenocarcinoma; DLBCL: diffuse large B cell lymphomas; BCL: B cell lymphoma; HNSCLC: non-small-cell lung cancer; SCLC: small-cell lung cancer; NHL: non-hodgkin lymphoma.
Table 2. Selected clinical trials of cytokines in innate immunity.
Table 2. Selected clinical trials of cytokines in innate immunity.
TargetNCT NumberStatusPhaseCancer TypeInterventionsEnrollment
GM-CSFNCT05284214Not yet recruitingIISolid TumorSargramostim, Ipilimumab-containing therapy65
NCT05530200Not yet recruitingIIMetastatic Solid TumorPD-L1 inhibitor, GM-CSF, IL-256
NCT03866525RecruitingI/IIGastrointestinal CancerOH2300
NCT05292417RecruitingIICRCGM-CSF in combination with Sintilimab and Fruquintinib71
NCT04725331RecruitingI/IIMetastatic CancerBT-001, Pembrolizumab48
type I IFNsNCT04053673RecruitingISolid TumorRBN-2397130
NCT05127590RecruitingI/IIAdvanced NSCLCRBN-239750
NCT04544007RecruitingIIGliomaPoly ICLC20
FLT3LNCT05029999RecruitingIMetastatic Triple Negative Breast CancerPLD Chemotherapy, CDX-1140, CDX-30145
NCT04491084RecruitingI/IINSCLCCDX-301, CDX-114046
NCT03789097RecruitingI/IINHL; HNSCC; Metastatic Breast CancerPembrolizumab, CDX-301, Poly ICLC56
NCT04616248Not yet recruitingIMultiple cancer typesCDX-301, CDX-1140, Poly ICLC18
IL-2NCT05307874RecruitingI/IISolid TumorICT01 in combination with IL-275
NCT05267626RecruitingI/IIAdvanced Solid TumorAU-007 69
NCT04862767RecruitingISolid TumorTASO-001 in combination with IL-29
NCT05493566Not yet recruitingILung Cancer IL-2 in combination with Pembrolizumab15
NCT05538624Not yet recruitingI/IIMultiple cancer typesAVB-001 44
IL-15NCT04294576RecruitingIAdvanced/Metastatic Solid TumorBJ-00192
NCT05470283RecruitingIMetastatic MelanomaOBX-11530
NCT05445882Not yet recruitingIICastration-Resistant PCAN-803TET28
NCT05266612Not yet recruitingISolid TumorVG202512
NCT05359211RecruitingIMultiple cancer typesNKTR-25524
CRC: colorectal adenocarcinoma; TET: thymic epithelial tumor; NSCLC: non-small-cell lung cancer; NHL: non-hodgkin lymphoma; HNSCC: head and neck squamous cell carcinoma; PCA: prostate cancer.
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

Zhang, Y.; Xue, W.; Xu, C.; Nan, Y.; Mei, S.; Ju, D.; Wang, S.; Zhang, X. Innate Immunity in Cancer Biology and Therapy. Int. J. Mol. Sci. 2023, 24, 11233. https://doi.org/10.3390/ijms241411233

AMA Style

Zhang Y, Xue W, Xu C, Nan Y, Mei S, Ju D, Wang S, Zhang X. Innate Immunity in Cancer Biology and Therapy. International Journal of Molecular Sciences. 2023; 24(14):11233. https://doi.org/10.3390/ijms241411233

Chicago/Turabian Style

Zhang, Yuxia, Wenjing Xue, Caili Xu, Yanyang Nan, Shuang Mei, Dianwen Ju, Shaofei Wang, and Xuyao Zhang. 2023. "Innate Immunity in Cancer Biology and Therapy" International Journal of Molecular Sciences 24, no. 14: 11233. https://doi.org/10.3390/ijms241411233

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