Open access peer-reviewed chapter

Role of Immune System in Kidney Cancer

Written By

Ana Marisa Chudzinski-Tavassi, Kátia Luciano Pereira Morais, Jean Gabriel de Souza and Roger Chammas

Submitted: 19 July 2017 Reviewed: 20 April 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.77379

From the Edited Volume

Evolving Trends in Kidney Cancer

Edited by Sashi S. Kommu and Inderbir S. Gill

Chapter metrics overview

1,375 Chapter Downloads

View Full Metrics

Abstract

Almost all kidney cancers are associated to immune dysfunction. Among these, renal cell carcinoma (RCC) represents approximately 2% of malignancies that affect adults and for 90–95% of all kidney cancers. Recent evidences have collaborated to elucidate the mechanisms involved in the development of this disease. In this view, dysfunctional neutrophil migration, as well as T lymphocyte-DC (dendritic cell) cross talk, DC maturation, immune cell metabolism, and reactivity and abnormal expression of cytokines and chemokines and their receptors have been highlighted in RCC and stroma cells. A rational development of novel therapies to recover antitumor activity of immune system is closely related to the understanding of the complex interactions between immune system and tumor. Some insights have been reached and immunomodulatory molecules, such as interleukin-2 (IL-2) and IFN-α, immune checkpoint inhibitors, and chemokines antagonists have shown clinical efficacy. In this chapter, we overview the essential role of innate and adaptive immune response in RCC and discuss drugs approved or in development for its treatment.

Keywords

  • renal cell carcinoma
  • cytokines
  • chemokines
  • natural immunity
  • adaptive immunity

1. Introduction

Basically, the kidney is assigned to the urine production function. Indeed, kidney function overcomes this definition, since it is critical for the regulation of the body’s electrolytes, body’s fluid balance, body’s acid-base balance, and depuration of body waste. Thereby, diseases such as kidney cancer compromise the kidney’s ability to perform its functions and bring consequences to the whole organism.

According to the cell type where the tumor starts, kidney cancers are classified as renal cell carcinoma (RCC) subdivided in papillary RCC, chromophobe RCC, rare types of RCC, and unclassified RCC. Also, there are transitional cell carcinoma, Wilms’ tumor, which almost always occur in children and renal sarcoma [1, 2, 3, 4].

Regardless of the category, kidney tumors have been associated with immune dysfunction [1, 2, 3, 5]. RCCs are rich in immune infiltrates consisting of T cells, natural killer (NK), DCs, macrophages among others [6, 7]. Different functions are ascribed to the different subsets of leukocytes. While the function of some of these cells is still elusive, like neutrophils which are essential components of the RCC microenvironment, others have well-defined roles in tumor progression. For example, tumor-associated macrophages (TAMs) are known for their immunosuppressive action, which is associated to the secretion of inhibitory cytokines, the generation of reactive oxygen species, regulatory T cells (Treg) development, and the induction of angiogenesis [6, 8, 9]. Likewise, myeloid-derived suppressor cells (MDSCs) have been reported preventing the formation and execution of an effective antitumor immune response by the inhibition of effector T-cell function and the induction of Tregs maturation [6, 7], besides the inhibition of DC maturation and DC-induced T-cell activation and antitumor cytotoxic T lymphocyte (CTL) [10].

The role of the immune system in RCC is not only observed at the cellular level but also through inflammatory mediators, that is, through the action of cytokines and chemokines which act in tumor and stroma cells [11, 12, 13]. These mediators in RCC and stroma cells lead to survival, proliferation, and migration and favor angiogenesis and metastasis [11, 12, 13]. Thereby, the modulation of immune system effectors has shown therapeutic potential. In fact, strategies involving immune checkpoint inhibitors, immunotherapy, and antagonists of chemokine receptors have proven clinical efficacy [14, 15].

Herein, we summarize relevant information about the role of natural and adaptive immunity in the development of RCC. Additionally, we describe cytokine and chemokine intracellular signaling pathways and mention how all knowledge has been useful for identification and advancement of therapeutic approaches for RCC.

Advertisement

2. Natural and adaptive immunity in RCC

The tumor microenvironment is a complex structure composed by several mediators that are involved in the cell signaling. The activation profile of extracellular matrix, fibroblasts, immune cells, blood vessels, and endothelial cells is essential to the pathogenesis of cancer, as well as to define therapeutic approaches [16, 17, 18]. Resident nontumor cells or infiltrated cells at tumor sites may even suppress the development of RCC. However, tumor cells can avoid the immune response coordinating changes in these cells and stimulating the secretion of immunosuppressive factors for tumorigenesis, pro-inflammatory cytokines including angiogenic factors, thus ensuring the supply of nutrients by newly formed, blood vessels, which further allow for tumor growth [19, 20].

In addition, the profile of soluble factors, such as cytokines and chemokines, present in the tumor microenvironment may undergo dual polarization allowing either tumor growth (leading to progressive disease) or its suppression (leading to regressive disease). Immune cells could interact with tumor cells, especially through immunosuppression, via cell-cell contact, or by the release of factors that maintain a supportive environment for tumor growth. Through some soluble mediators, such as tumor necrosis factor-alpha (TNFα), immune cells can stimulate other cells, such as fibroblasts, to produce proteolytic enzymes for extracellular matrix remodeling and collagen, facilitating the spread of metastases. It may also stimulate the involvement of endothelial cells, which become able to form new vessels [18, 21, 22]. On the other hand, TNFα, which is mainly produced by macrophages, has also an important role in the recruitment of other immune cells to the tumor site where they will be able to assemble a response against the tumor. Thus, the constant interference of mediators may favor/inhibit an adequate environment for tumor maintenance and growth [18, 21].

Indeed, tissue repair promoted by inflammation is self-limited, and the imbalance on this process may be pathogenic, giving inflammatory cells either a beneficial and/or a detrimental role in the pathogenesis of various diseases, including chronic inflammation and neoplasia [23]. The TAMs have at least two well-described states of polarization, according to immunological competences. While M1 (classically activated) macrophages can inhibit tumor growth, M2 (alternatively activated) macrophages stimulate tumor growth [24, 25, 26]. The M1 polarization is functionally characterized by the release of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, and TNFα) and reactive nitrogen/oxygen species (RNI/ROS) acting as microbicidal and tumoricidal [24]. In the tumor context, some authors suggest that macrophages in the initial phase of tumorigenesis can naturally inhibit tumor growth, eradicate tumor cells, and stimulate the immune response. In contrast, M2 macrophages may favor neoplasia by producing anti-inflammatory cytokines that suppress the cellular response, release vascular endothelial growth factors (VEGFs), responsible for angiogenesis, and release transforming growth factors (TGF-beta) [27].

The different subtypes of T lymphocytes are also recruited into the tumor microenvironment and interact with tumor cells, which may induce tumor cell death, becoming anergic or even suppressing the immune response against the tumor [28, 29]. T CD8+ lymphocytes, also known as cytotoxic T cells, play an important role in the adaptive immunity in response to tumor-specific epitopes. Together with NK and Natural killer T (NKT) cells, T CD8+ cells induce tumor cell death by apoptosis through the secretion of cytotoxic factor, such as granzymes and perforins. Besides, when the TCD8 lymphocyte is activated, it secretes IFN-gamma and IL2, activating other TCD8 cells and M1 macrophages, skewing the inflammatory response toward a Th1 profile. However, RCC and some types of tumor-infiltrating immune cells, such as MDSCs, present in the tumor microenvironment can suppress antitumor response T cell mediated by the expression of programmed death ligand-1 (PD-L1, also known as B7-H1 or CD274) [30, 31].

CD4 T lymphocyte is endowed with great plasticity, being able to differentiate into Th1, Th2, Th3, Th17, and Treg subtypes. Treg cells play a critical role in the control of the acute phase of the inflammatory response through immunosuppression. High levels of Treg in the tumor microenvironment are considered as poor prognosis for tumors, for example, in renal and pancreatic tumor, due to the recruitment of other immunosuppressive cells and the stimulation of the angiogenic process [29, 32]. MDSCs are involved in chronic inflammation processes, which are recruited by tumor or Treg, increasing immunosuppression in the tumor microenvironment [33]. A common feature of almost all solid tumors is hypoxia that could lead to the stability of the HIF-1α transcription factor (hypoxia-inducible factor-1α). HIF-1α can bind to a hypoxia-responsive element in the PD-L1 promoter leading to PD-L1 expression, not only on tumor cells but also on MDSCs, macrophages, and DCs within the tumor microenvironment [34].

PD-L1 binds with PD-1 present on the cell surface of activated T cells leading to proliferation blockade, dysfunctional response, and T-cell death. CTLA-4 (CTL -associated antigen 4), a receptor also present in T cells, is the best studied inhibitory molecule, well known for its capacity of blocking T cell-mediated immune responses [35, 36]. CTLA-4 competes with CD28 for CD80 or CD86 binding. When the CTLA-4/CD80 or CD86 interaction occurs, the cell becomes anergic and dies. The overexpression of CTLA-4 is involved in several neoplastic, inflammatory, and autoimmune diseases [37]. The inhibition of T response allows for RCC to avoid effectors of immunologic control and facilitates invasion and metastasis. Accordingly, PD-L1 overexpression is related to worse prognosis for metastatic RCC, and it is therefore an important target for drug discovery [38, 39]. New drugs based on monoclonal antibodies, such as anti-PD-1 and anti-CTLA-4, have been strongly explored in clinical trial and proven to be efficacious to combinatorial treatments [39]. Indeed, immunotherapy is a promising therapeutic strategy for different types of cancers, but it is often not sufficient to control tumor growth [40]. Several therapeutic strategies based on innate, adaptive, humoral, or cytokine immune system responses have been studied in order to combat tumor cells in the host [41, 42, 43, 44, 45].

Studies using allogeneic DCs in metastatic Renal Cell Carcinoma (mRCC) and melanoma patients are widespread due to the ability of these cells to mediate the cell signaling between the innate and adaptive immune response. As professional antigen-presenting cells (APCs), these cells phagocytose tumor cell particles, process them, and further present their epitopes to further activate effector lymphocytes [46]. APCs play a crucial role in coordinating the immune response, where the imbalance between populations of macrophages, immature, and mature DCs significantly affects the immune response against solid tumors [47]. Both DCs and macrophages can be activated by some microbial stimulus or cytokines in an inflammatory environment. DCs and macrophages could differentiate from monocytes, according to the tissue environment, where cytokines such as IL4 and macrophage colony-stimulating factor granulocytes (GM-CSF) may induce differentiation, followed by TNFα-induced maturation [47]. In the tumor context, the inflammatory profile present at different stages of disease development may contribute to or eradicate the pathogenesis [47]. The International Society for the Biological Therapy of Cancer (iSBTc), together with the Society for Cancer Immunotherapy (SITC), has been discussing the theme with the purpose of advancing the critical understanding of the involvement of inflammation during pathogenesis and cancer treatment [48, 49]. In fact, the dynamics of tumor progression along with immunoedition has been studied for years, through the realization of which cells, molecules, and pathways of the immune system are engaged at different steps of the evolution of cancers. Nonetheless, an integrative picture of the whole process is still missing [50, 51]. Vesely et al. described several interactions between innate and adaptive immunity in cancer, suggesting a dynamic immuno-delivery model, where cells such as M2 macrophages, MDSC, Th17, Treg, and TCD8+ overexpressing CTLA-4 receptor are present in chronic inflammation and favor the processes related to tumor progression [52]. In contrast, NK, NKT, TCD8+, TCD4+, M1 macrophages, DCs, Tγδ cells and IL12 and IFN-gamma cytokines are important for equilibrium and elimination phases of tumor control [52]. Several cytokines are involved in the differentiation process of immune cells. Interferon-gamma (INF-gamma) belongs to interferon family and is a natural glycoprotein that shows antiviral, antiproliferative, and immunomodulatory properties. INF-gamma plays a key role in the tumor microenvironment, where it aids in tumor eradication. This cytokine is able to recruit and induce the proliferation of T lymphocytes in the tumor microenvironment, to activate innate immunity cells, rendering them cytotoxic, besides polarizing the Th1 response from T CD4 lymphocytes [53, 54]. IL1-beta is one of the major cytokines involved in the pro-inflammatory response, which is synthesized by several immune cells, such as monocytes, macrophages, DCs, B lymphocytes, NK cells, among others. It has similar activities as described for TNFα, favoring tumor invasion and the angiogenic process, as well as favoring vascular permeability and facilitating the recruitment of immune system cells to the tumor microenvironment [55]. However, TNFα is the main mediator of the acute inflammatory response, being secreted primarily by macrophages and T cells. TNFα causes vascular endothelial cells to increase the expression of leukocyte integrins inducing chemotaxis. In addition, TNFα also acts on phagocytic cells, which characterizes an autocrine effect, since macrophages, apart from secreting TNFα, may respond to the stimulus itself, releasing IL1-beta [56, 57]. IL12 is secreted primarily by macrophages, DCs, monocytes, and neutrophils. It has action in the activation of cytotoxic NK cells and TCD8 lymphocytes, but its main function in the antitumor activity is involved in the activation and proliferation of T lymphocytes and NK cells, which induces the production of IFN-gamma. Moreover, IL12 and INF-gamma together are able to differentiate T-helper cells into Th1 cells [58, 59]. IL6 is synthesized by mononuclear phagocytes, such as macrophages and also by some activated T cells and by other cell types that are not part of the immune system in response to microorganisms or IL1-beta and TNFα stimuli [60]. IL10 is a cytokine known to be anti-inflammatory, synthesized in the form of monomers of 18–20 kDa, being functional in the form of homodimers. This cytokine can be produced by Th2 lymphocytes, monocytes, and epithelial cells. Its main action is to suppress the synthesis of several inflammatory cytokines such as IL1-beta, TNFα, IL-6, IL-8, and IL-12, as well as hematopoietic growth factors (GM-CSF, G-CFS) and macrophage colony-stimulating factor (M-CFS). In addition, IL-10 can inhibit the synthesis of nitric oxide, gelatinase, and collagenase, avoiding tissue injury [61]. Although its role in the tumor context remains unclear, IL-17 is a pro-inflammatory cytokine secreted by Th17 lymphocytes, which regulates NFkB and MAPK activities. It is constantly involved in the acute phase of inflammatory diseases, such as autoimmune diseases, and it is associated with poor prognosis in patients with RCC [62, 63].

Taken all these studies together, the dual role of natural and adaptive immunity in RCC is evident (Figure 1).

Figure 1.

Dual role of natural and adaptive immunity in RCC. Innate immune cells (dendritic cells, macrophages, NK cells, neutrophils, and MDSCs) and adaptive immunity (B and T cells) in the tumor microenvironment may undergo dual polarization allowing either tumor growth (leading to progressive disease) or its suppression (leading to regressive disease). Dendritic cells are primed by tumor antigens, which are then presented to T and B cells for adaptive responses. On the other hand, dendritic cells can directly drive tumor angiogenesis through the release of pro-angiogenic cytokines such as TNFα and CXCL8. Similarly, neutrophils in the tumor microenvironment are able to release pro-angiogenic factors and chemokines that could contribute for cancer progression and metastasis. T CD8+ lymphocytes, also known as cytotoxic T cells, play an important role against tumor cells under response to tumor-specific epitopes. However, the PD-L1/PD-1 interaction on CD8+ T-cell surface induces cellular energy suppressing the effector response, leading CD8 T cell to death. Failure of CD8+ T cells to kill tumor cells involves signals from multiple cells including MDSC, Treg, and TAMs. NK cells are characterized by a high cytolytic capacity against transformed cancer cells by the secretion of granzyme and perforin. Tumor cells and fibroblasts also produce survival/growth-promoting chemokines. Metastatic cancer cells are facilitated by the upregulation of particular chemokine receptors (such as CXCR4) by tumor cells, which enables them to migrate to secondary tissues where the ligands are expressed.

Advertisement

3. Cytokines and chemokine intracellular signaling pathways

Physiologically, inflammation is a mechanism of tissue reaction for elimination, neutralization, and destruction of the cause of aggression, as injurious stimuli such as microbial pathogens, irritants, or toxic cellular components [64]. Cells of the immune system including monocytes, macrophages, neutrophils, basophils, DCs, mast cells, T cells, and B cells play a role in this process [64]. These events are in turn controlled by a host of extracellular molecular regulators, including members of the cytokine and chemokine families that mediate both immune cell recruitment and complex intracellular signaling control mechanisms; as a result, cells assemble and disassemble a complex array of signaling pathways as they move from inactive to dedicated roles within the inflammatory response site. Disruption of these pathways triggers inflammatory disorders that could contribute for the development of some diseases as kidney cancer and other types of cancers [65].

Key pro-inflammatory cytokines in kidney cancer include interleukin-1 beta (IL-1β), IL-6, and TNFα, all of which signal via the type I cytokine receptors that are structurally divergent from other cytokine receptor types [66, 67]. IL-1 signaling starts through its binding to its receptor composed of two subunits, interleukin 1 receptor type I (IL-1RI) and interleukin 1 receptor accessory protein (IL1RAP) [68, 69]. Signaling proceeds with TIR adaptor and MyD88 by recruitment of IL-1R-associated kinases (IRAKs), which promote TNFR-associated factor 6 (TRAF6) polyubiquitination via lysine 63 linkages. Subsequently, TRAF6 interacts with the TAK1/TAB1/TAB2 complex that allows NFκB nuclear translocation (p65/p50) resulting in pro-inflammatory gene expression [69]. Also, TAK1/TAB1/TAB2 complex triggers the activation of the mitogen-activated protein kinases (MAPKs), c-JunN-terminal kinase (JNK), and p38, which induce the expression of pro-inflammatory genes. Similarly, TNFα binding to TNFR1 results in NFκB nuclear translocation, MAPKs, JNK, and p38, but signaling is coordinated by complex I (TRADD/TRAF2/RIP) [67, 70]. Importantly, TNFα signaling following receptor internalization is thought to be pro-apoptotic, via the formation of complex II (TRADD/FADD/Pro-Caspase-8) [67]. On the other hand, IL-6 binds to the FIII domains of the IL-6R chains, unleashing its signal via the gp130 proteins [71, 72]. Consequently, Janus kinases (JAKs) are recruited to the receptor, phosphorylating it and themselves, triggering STAT3 activation and transcription of pro-inflammatory genes and intracellular adhesion molecules [64].

Currently, it is not fully understood how the signaling triggered by these cytokines contributes to tumor progression, but high serum levels of these pro-inflammatory cytokines are associated with advanced disease [73]. Some evidence has arisen, as follows. It is well known that angiogenesis is stimulated by inflammatory mediators in the tumor microenvironment, such as those expressed by TAMs [74]. Interestingly, TAMs isolated from RCC tumors express high levels of IL-1β, TNFa, and IL-6 [75]. In addition, mouse models have demonstrated that the inhibition of IL-1β signaling reduced tumor blood vessel formation [76] and IL-1β mediates metalloproteinase-dependent RCC tumor cell invasion through the activation of cytosine-cytosine-adenosine-adenosine-thymidine (CCAAT) enhancer binding protein b [67]. Regarding TNFα signaling, many studies associated it to chemokine overexpression in tumor and nontumor cells [77, 78]. Moreover, TNFα plays an important role in the progression of RCC by inducing epithelial to mesenchymal transition and CD44 expression, which may be involved in the resistance to the sunitinib treatment [66]. There is no direct correlation between IL-6R and RCC development; however, RCC cells express high levels of IL-6, and its signaling activity seems necessary for carcinogenesis, tumor progression, and tumor evasion of the immune system. STAT3 activation by IL-6 promotes tumorigenesis by preventing apoptosis while enhancing proliferation, angiogenesis, invasiveness, and immune evasion [79]. For example, activated STAT3 induces HIF-1α-mediated VEGF expression in human RCC cell [79].

Besides these pro-inflammatory cytokines, other mediators act as crucial players in RCC. Chemotactic cytokines or chemokines are responsible for the recruitment of cells from both the innate and adaptive immune systems to the site of injury or infection [64]. Chemokines induce integrin expression, such as the β2-integrin lymphocyte function-associated antigen (LFA-1), in target leukocytes, thus acting in the arrest of these cells and favoring diapedesis through the endothelium [71]. Despite this, primary chemotaxis action, chemokines, and their receptors are physiological relevant in many biological process, such as the initiation of adaptive immune responses, immune surveillance and the migration, proliferation, and survival signals in multiple cell types [64]. Chemokine signals are transduced through binding to members of the seven-transmembrane, G-protein-coupled receptor (GPCR) superfamily [80]. GPCRs exist as a heterotrimer containing three subunits: α, β, and γ. In its inactive form, the G protein is complexed in α, β, and γ, with guanosine diphosphate (GDP) fixed to the α subunit. Once stimulated by a receptor activated by its ligand, the α subunit exchanges its GDP for Guanosine-5’-triphosphate (GTP) [81]. This causes the dissociation of α which separate β and γ subunits by interacting with an effector protein or ion channel in order to stimulate or inhibit secondary intracellular messengers [81]. CXCR4 is well known for its role in the homing of progenitor cells into the bone marrow and, recently, associated with poor RCC prognosis, and it is mainly coupled to the Gαi subunit, which, after dissociation of the Gαβγ complex upon CXCR4 stimulation (Figure 2), is traditionally been regarded as the major signaling subunit, inhibits adenylyl cyclase activity, and triggers MAPK and phosphatidylinositol-3-kinase (PI3K) pathway activation [82]. The Gβγ subunits, in turn, lead to the activation of phospholipase C (PLC), causing the hydrolysis of the phospholipid membrane phosphatidylinositol 4,5-bisphosphate (PIP2) in inositol 1,4,5-trisphosphate (IP3) and 1,2-diacylglycerol (DAG) [13, 82]. IP3 can bind to channels of the endoplasmic reticulum, inducing the mobilization of Ca2+ ions [83, 84]. This could also be considered a downstream effect of Gαi activity, since the inhibition of Gαi activity by its potent inhibitor pertussis toxin has been reported to lead to a decreased Ca2+ mobilization from intracellular stores [85]. CXCR4 can also act by interaction with other Gα subunits, that is, Gαq or Gα12, each of which has been associated with different intracellular signaling cascades [86]. Indeed, chemokine receptors also activate signaling pathways independent of G proteins, including p38MAPK and JAK/Stat to regulate cellular processes such as migration and gene transcription [87, 88].

Figure 2.

Signaling induced by CXCR4 via G protein. CXCL12 binding causes the dissociation of α which separate β and γ subunits; as a result, βγ PLC is activated leading to calcium mobilization. PI3K/Akt activation by Gα triggers a transduction signal that contributes with cell survival, proliferation, and migration, which is associated with RCC disease. In contrast, the antagonist of CXCR4 blocks CXCR4 intracellular pathway.

Regarding the signals triggered by pro-inflammatory mediators in RCC, CXCL12 chemokine and its receptors CXCR4 and CXCR7 have gained prominence, since this pathway, which is associated with chronic inflammation, is upregulated in RCC [65, 89, 90]. In RCC cells and in other tumor cells, these chemokines activate the PI3K/Akt pathway; consequently, many downstream elements of the Akt pathway are regulated, leading to tumor cell survival [13, 91, 92]. Frequently, NFκB nuclear translocation is observed following transcription of various apoptosis inhibitors and cell-cycle-promoting genes [93], but can be activated through other pathways, such as PKC [94]. Other downstream targets of Akt include procaspase-9 and the pro-apoptotic Bcl-2 family member, BAD (Bcl-2/Bcl-XL-antagonist, causing cell death), both of which are inhibited upon phosphorylation. Other consequence of Akt activation is the inhibition of members of the FKHR (forkhead in rhabdomyosarcoma) family of transcription factors, which induce the transcription of numerous apoptotic genes [95, 96]. Besides, Akt signaling could induce p53 degradation and inhibition of GSK-3β (glycogen synthase kinase-3β), leading to stabilization of β-catenin, resulting in the downstream inhibition of negative regulators of cell cycle and the activation of cell-cycle-promoting genes [97]. mTOR (mammalian target of rapamycin) activation is also induced by Akt, which leads to p70S6K (p70 S6 kinase) activation and thus enhances protein translation of numerous cell-growth regulators [13]. Furthermore, intracellular events triggered by the activation of chemokines receptors lead to ERK1/2 signaling, following the inhibition of procaspase-9 and BAD [98, 99], the induction of transcription factors involved in cell-cycle regulation, and differentiation, thereby promoting cell proliferation [100]. Other MAPKs, including JNK, have also been implicated in chemokine-induced proliferation signaling [101]. Also, HIF-1α may be induced by chemokines signaling that contributes to VEGF expression, which is known to be the inducer of CXCR4 expression [101]. It is important to mention that not all of these pathways have been studied in detail in RCC cells, but there is strong evidence of their involvement in the development of this disease. For example, Rac1 was not previously reported to be involved in RCC development, but some studies have shown its role in controlling tumor cell growth and chronic kidney disease [102]. In summary, CXCL12, CXCR4, and CXCR7 release result in the activation of transcription factors involved in antiapoptotic mechanisms, cell-cycle regulation, and growth factor production, favoring tumor growth and metastasis.

Also, there are some particularities regarding CXCR7 role in RCC, which are independent of GPCRs. CXCR7 plays a role as a decoy for CXCL12, promoting some CXCL12α accumulation and triggering a differential signaling by CXCR4 [103, 104]. Besides, CXCR7 interacts and signals by β-arrestin in a ligand-dependent manner [83, 104].

Obviously, here we emphasize intracellular events in tumor cells. However, while some processes take place within the tumor cells, others would occur in the stroma: it is the synergism between these responses that contributes to the progression of the disease. RCC and other tumors interact with the surrounding tumor stroma through a variety of cytokines, chemokines, and growth factors [84]. The tumor chemotactic environment recruits inflammatory cells including neutrophils, macrophages, and lymphocytes. Although initially these cells may have a protective antitumoral role, as displayed by neutrophils, which have a higher cytotoxic activity against poorly metastatic cells, secondarily neutrophils could contribute to cancer progression. Leukocytes can produce cytokines, growth factors, and MMPs that enhance growth, proliferation, and angiogenesis, as exemplified by the TAMs, which release growth and angiogenic factors (e.g., VEGF) and basic fibroblast growth factor [105]. Thus, cellular communication by paracrine and autocrine chemokine/cytokine signaling contributes for the survival and growth of metastatic cells. In other words, stroma cells may support tumors at the same time as that tumor cells in turn modulate the microenvironment within which they reside.

Advertisement

4. Immunomodulatory molecules as new therapeutic targets for RCC

Among the standard drugs for metastatic RCC treatment, the most effective regimens include the combination of targeted therapy agents and antiangiogenic agents (tyrosine kinase inhibitors, such as sorafenib, sunitinib, pazopanib, and axitinib), or an antiangiogenic antibody routinely employed in combination with interferon alpha (bevacizumab) [106, 107], and antiproliferative agents (mTOR inhibitors, such as temsirolimus and everolimus) [106, 107]. Despite the effectiveness of these therapies, resistance to antiangiogenic therapy almost always occurs and are associated with high toxicity and, consequently, serious adverse events.

Considering all knowledge previously mentioned in this chapter, it is not surprising that agents altering the immune response in order to prevent tumor growth and metastases have gained space in this scenario [12, 14, 108]. Known as immunomodulators, these molecules have been studied for some time now [109]. Among the first representatives of this class are IL-2 and interferon-alpha (IFN-α). IL-2 is a naturally occurring cytokine, which have antitumor activity by the induction of proliferation of NK cells, lymphokine-activated killer cells (LAKs), and other cytotoxic cells [110, 111]. IFN-α is also a molecule that had its clinical effect for the treatment of RCC demonstrated years ago [109, 112]. It is a pleiotropic cytokine with immunomodulatory activities that is able to induce the differentiation of monocytes into highly activated DCs, which are particularly effective in recognizing complex antigens and inducing T- and B-cell immunity and thus participate in the generation of antitumor T-cell immunity [112]. However, cytokine immunotherapies work effectively only in a minority of RCC patients and currently are not considered for the standard treatment of RCC due to their high toxicity.

In this context, cytokines have been exploited. Combination therapy using an antihuman IL-6R antibody with interferon has been suggested as a novel therapeutic approach for the treatment of RCC. Besides, an IL-6R-neutralizing antibody, tocilizumab, in combination with sorafenib suppressed tumor growth and inhibits angiogenesis in vivo more efficiently than sorafenib alone [113, 114]. Similarly, infliximab, an anti-TNFα monoclonal antibody that prevents TNFα binding to its receptors, TNFR1 (p55 receptor) and TNFR2 (p75 receptor) and causes cell death via complement-mediated lysis through interaction with membrane-bound TNF, has been considered for RCC treatment [115]. However, treatments with anti-TNFα monoclonal antibody showed varying results in independent studies, probably due to reactions given by this cytokine to different conditions, environmental and genetic factors and/or other unknown or unexplained factors [116]. Besides anti-TNFα strategies, targeting IL-1β has also been reported as possible therapy for RCC [117].

Promising candidates for RCC treatment have also been designed to specifically target chemokines and their receptors. One of the most widely studied compounds is AMD3100 which is thought to specifically block CXCR4 signaling [118] and that acts directly in RCC tumor [91] cells as well as local antitumor immune response, by impairing Tregs function [119], know to suppress a whole range of immune cells including B cells, NK cells, NKT cells, CD4+ or CD8+ T cells, monocytes, and DCs [119]. Also, many other chemokine antagonists have also shown potential for clinical application in cancer treatment and could be useful for RCC treatment in the future. For example, anti-CXCR7-12G8 and CCX77, CXCR7 inhibitors or CTCE-9908, which is a peptide analog of CXCL12 and an active inhibitor of the ligand, has shown promising results as well as tolerated drug that stabilized disease in early clinical trials for late-stage cancer patients [12, 120].

Recently, antibodies that inhibit T-cell coinhibitory receptors have emerged as therapeutic promises not only in the treatment of RCC but also in other tumors by inhibiting T-cell regulatory activity and increasing the antitumor immune response [14, 121]. Nivolumab and pembrolizumab (anti-PD-1) [14, 122], avelumab, and durvalumab (anti-PD-L1) are in late-stage clinical development for a number of indications [14, 123], besides the first in its class, ipilimumab (anti-CTLA-4), already approved for use in a number of indications [14, 124].

Finally, the close relationship between cancer and immune system has suggested that current drug therapies used to treat inflammatory diseases or particular types of cancers could function as inhibitors of chemokine signaling and could therefore be redirected toward the treatment of other cancers [12, 116]. This hypothesis needs to be tested by further preclinical and clinical investigation, which elucidates how these drugs would act at molecular and systemic levels.

Advertisement

5. Role of immune system in kidney cancer and the future

The role of immune system in kidney cancer is becoming more clear, whereas new findings that arise from clinical trials and identification of additional predictive biomarkers increase our understanding of the tumor microenvironment. Looking to the future based on the knowledge we have today, the perspective is a better understanding of immune system in tumor stroma as well as in various steps in cancer growth and metastasis.

Regarding RCC therapy, a promising option is the combination therapy based on targeted agents (inhibition of mTOR or VEGF pathways associated with immunotherapies) or immunotherapy + immunotherapy, which would overcome tumor resistance, as well as to restore functional immune system cancer surveillance and response. Currently, there are many clinical trials investigating combination therapy: nivolumab (anti-PD-1) + ipilimumab (anti-CTLA-4) [125], pembrolizumab (anti-PD-1) + ipilimumab (anti-CTLA-4) [126], pidilizumab (anti-PD-1) + vaccine (DC/RCC fusion cell vaccine) [127], atezolizumab (anti-PD-L1) + bevacizumab (anti-VEGF) [127], nivolumab (anti-PD-1) + bevacizumab (anti-VEGF) [128], pembrolizumab (anti-PD-1) + pazopanib (TKI) [129], nivolumab (anti-PD-1) + sunitinib [130].

Also, cell-based therapies have become interesting, which includes adoptive T-cell therapies such as tumor-infiltrating lymphocytes (TILs), T-cell receptor (TCR), and chimeric antigen T-cell (CAR-T) therapy [127].

Other promising alternative of therapy for RCC is vaccine-based immunotherapy. AGS-003 is an autologous DC vaccine that is generated from host DCs in response to tumor mRNA [131], which is also to investigate in combination with sunitinib [132].

All these therapeutic investigations highlighted the importance of immune system in the future study about RCC. Information about immune system may be decisive for clinical decisions.

Advertisement

6. Conclusion

Immune system plays a role in kidney cancer, and this should be considered for both the understanding of the disease and the development of novel therapies.

Advertisement

Acknowledgments

Work in our laboratory is supported by grants provided by the São Paulo Research Foundation (CETICs 2013/07467-1 and 2015/50040-4, São Paulo Research Foundation and GlaxoSmithKline). The authors thank the National Counsel of Technological and Scientific Development (CNPq, INCTTox), Coordination of Improvement of Higher Education Personnel (CAPES), BNDES 13.2.0711.1/2013, and União Química Farmacêutica Nacional for the support.

Advertisement

Conflict of interest

The authors declare they have no conflicts of interest.

References

  1. 1. Shimodaira S, Hirabayashi K, Yanagisawa R, Higuchi Y, Sano K, Koizumi T. In: van den Heuvel-Eibrink MM, editor. Dendritic Cell-Based Cancer Immunotherapy Targeting Wilms’ Tumor 1 for Pediatric Cancer. Brisbane, Australia: Wilms Tumor; 2016
  2. 2. Hanna KS. A review of immune checkpoint inhibitors for the management of locally advanced or metastatic urothelial carcinoma. Pharmacotherapy. 2017;37(11):1391-1405. PubMed PMID: 28950037
  3. 3. Alikhan MB, Pease G, Watkin W, Grogan R, Krausz T, Antic T. Primary epithelioid sarcoma of the kidney and adrenal gland: Report of 2 cases with immunohistochemical and molecular cytogenetic studies. Human Pathology. 2017;61:158-163. PubMed PMID: 27769872
  4. 4. Kidney Cancer Update. The year in review in kidney cancer. Clinical Advances in Hematology & Oncology: H&O. 2015;13(5):327-329. PubMed PMID: 26352778
  5. 5. Florek M, Haase M, Marzesco AM, Freund D, Ehninger G, Huttner WB, et al. Prominin-1/CD133, a neural and hematopoietic stem cell marker, is expressed in adult human differentiated cells and certain types of kidney cancer. Cell and Tissue Research. 2005;319(1):15-26. PubMed PMID: 15558321
  6. 6. Santoni M, Berardi R, Amantini C, Burattini L, Santini D, Santoni G, et al. Role of natural and adaptive immunity in renal cell carcinoma response to VEGFR-TKIs and mTOR inhibitor. International Journal of Cancer [Journal International du Cancer]. 2014;134(12):2772-2777. PubMed PMID: 24114790
  7. 7. Murphy KA, James BR, Guan Y, Torry DS, Wilber A, Griffith TS. Exploiting natural anti-tumor immunity for metastatic renal cell carcinoma. Human Vaccines & Immunotherapeutics. 2015;11(7):1612-1620. PubMed PMID: 25996049. Pubmed Central PMCID: 4514306
  8. 8. Daurkin I, Eruslanov E, Stoffs T, Perrin GQ, Algood C, Gilbert SM, et al. Tumor-associated macrophages mediate immunosuppression in the renal cancer microenvironment by activating the 15-lipoxygenase-2 pathway. Cancer Research. 2011;71(20):6400-6409. PubMed PMID: 21900394
  9. 9. Hemmerlein B, Johanns U, Halbfass J, Bottcher T, Heuser M, Radzun HJ, et al. The balance between MMP-2/−9 and TIMP-1/−2 is shifted towards MMP in renal cell carcinomas and can be further disturbed by hydrogen peroxide. International Journal of Oncology. 2004;24(5):1069-1076. PubMed PMID: 15067327
  10. 10. Cabillic F, Bouet-Toussaint F, Toutirais O, Rioux-Leclercq N, Fergelot P, de la Pintiere CT, et al. Interleukin-6 and vascular endothelial growth factor release by renal cell carcinoma cells impedes lymphocyte-dendritic cell cross-talk. Clinical and Experimental Immunology. 2006;146(3):518-523. PubMed PMID: 17100773. Pubmed Central PMCID: 1810419
  11. 11. O'Hayre M, Salanga CL, Handel TM, Allen SJ. Chemokines and cancer: Migration, intracellular signalling and intercellular communication in the microenvironment. The Biochemical Journal. 2008;409(3):635-649. PubMed PMID: 18177271
  12. 12. Hembruff SL, Cheng N. Chemokine signaling in cancer: Implications on the tumor microenvironment and therapeutic targeting. Cancer Therapy. 2009;7(A):254-267. PubMed PMID: 20651940. Pubmed Central PMCID: 2907742
  13. 13. Pawig L, Klasen C, Weber C, Bernhagen J, Noels H. Diversity and Inter-connections in the CXCR4 chemokine receptor/ligand family: Molecular perspectives. Frontiers in Immunology. 2015;6:429. PubMed PMID: 26347749. Pubmed Central PMCID: 4543903
  14. 14. Ross K, Jones RJ. Immune checkpoint inhibitors in renal cell carcinoma. Clinical Science. 2017;131(21):2627-2642. PubMed PMID: 29079639
  15. 15. Chan JY, Choudhury Y, Tan MH. Predictive molecular biomarkers to guide clinical decision making in kidney cancer: Current progress and future challenges. Expert Review of Molecular Diagnostics. 2015;15(5):631-646. PubMed PMID: 25837857
  16. 16. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Integrating immunity's roles in cancer suppression and promotion. Science. 2011;331(6024):1565-1570. PubMed PMID: 21436444. Epub 2011/03/26. eng
  17. 17. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005;7(3):211-217
  18. 18. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436-444
  19. 19. Ono M. Molecular links between tumor angiogenesis and inflammation: Inflammatory stimuli of macrophages and cancer cells as targets for therapeutic strategy. Cancer Science. 2008;99(8):1501-1506. PubMed PMID: 18754859
  20. 20. Hori Y, Stern PJ, Hynes RO, Irvine DJ. Engulfing tumors with synthetic extracellular matrices for cancer immunotherapy. Biomaterials. 2009;30(35):6757-6767 PubMed PMID: 19766305. Pubmed Central PMCID: 2788234. Epub 2009/09/22. eng
  21. 21. Mantovani A. Cancer: Inflaming metastasis. Nature. 2009;457(7225):36-37
  22. 22. de Visser KE, Coussens LM. The interplay between innate and adaptive immunity regulates cancer development. Cancer Immunology, Immunotherapy. 2005;54(11):1143-1152. PubMed PMID: 15889249. Epub 2005/05/13. eng
  23. 23. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860-867. PubMed PMID: 12490959
  24. 24. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends in Immunology. 2002;23(11):549-555. PubMed PMID: 12401408
  25. 25. Sica A, Saccani A, Mantovani A. Tumor-associated macrophages: A molecular perspective. International Immunopharmacology. 2002;2(8):1045-1054. PubMed PMID: 12349942
  26. 26. Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L. The origin and function of tumor-associated macrophages. Immunology Today. 1992;13(7):265-270. PubMed PMID: 1388654
  27. 27. Gebhard B, Gnant M, Schutz G, Roka S, Weigel G, Kandioler D, et al. Different transendothelial migration behaviour pattern of blood monocytes derived from patients with benign and malignant diseases of the breast. Anticancer Research. 2000;20(6B):4599-4604. PubMed PMID: 11205309
  28. 28. Schlossman SF, Reinherz EL. Human T-cell subsets in health and disease. Springer Seminars in Immunopathology. 1984;7(1):9-18
  29. 29. Zikich D, Schachter J, Besser MJ. Predictors of tumor-infiltrating lymphocyte efficacy in melanoma. Immunotherapy. 2016;8(1):35-43. PubMed PMID: 26653685. Epub 2015/12/15. Eng
  30. 30. Thompson RH, Kuntz SM, Leibovich BC, Dong H, Lohse CM, Webster WS, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Research. 2006;66(7):3381-3385. PubMed PMID: 16585157. Epub 2006/04/06. eng
  31. 31. Zhang BY, Thompson RH, Lohse CM, Leibovich BC, Boorjian SA, Cheville JC, et al. A novel prognostic model for patients with sarcomatoid renal cell carcinoma. BJU International. 2015;115(3):405-411. PubMed PMID: 24730416. Epub 2014/04/16. eng
  32. 32. Oleinika K, Nibbs RJ, Graham GJ, Fraser AR. Suppression, subversion and escape: The role of regulatory T cells in cancer progression. Clinical and Experimental Immunology. 2013;171(1):36-45. PubMed PMID: PMC3530093
  33. 33. Ribas A, Shin DS, Zaretsky J, Frederiksen J, Cornish A, Avramis E, et al. PD-1 blockade expands intratumoral T memory cells. Cancer Immunology Research. 2016;19:2016
  34. 34. Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. The Journal of Experimental Medicine. 2014;211(5):781-790
  35. 35. Figlin RA, Thompson JA, Bukowski RM, Vogelzang NJ, Novick AC, Lange P. Multicenter, randomized, phase III trial of CD8(+) tumor-infiltrating lymphocytes in combination with recombinant interleukin-2 in metastatic renal cell carcinoma. Journal of Clinical Oncology. 1999;17(8):2521-2529
  36. 36. Thompson RH, Kwon ED, Allison JP. Inhibitors of B7-CD28 costimulation in urologic malignancies. Immunotherapy. 2009;1(1):129-139 PubMed PMID: 20445772. Pubmed Central PMCID: 2864044. Epub 2009/01/01. eng
  37. 37. McCoy KD, Gros G. The role of CTLA-4 in the regulation of T cell immune responses. Immunology and Cell Biology. 1999;77(1):1-10
  38. 38. McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, et al. Atezolizumab, an anti–programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: Long-term safety, clinical activity, and immune correlates from a phase Ia study. Journal of Clinical Oncology. 2016;34(8):833-842. PubMed PMID: 26755520
  39. 39. Philips GK, Atkins MB. New agents and new targets for renal cell carcinoma. American Society of Clinical Oncology Educational Book. 2014:e222-e227 PubMed PMID: 24857106. Epub 2014/05/27. Eng
  40. 40. Pappalardo F, Forero IM, Pennisi M, Palazon A, Melero I, Motta S. SimB16: Modeling induced immune system response against B16-melanoma. PLoS One. 2011;6(10):e26523
  41. 41. Dall'Oglio M, Srougi M, Barbuto JA. Complete response of metastatic renal cancer with dendritic cell vaccine. International Brazilian Journal of Urology. 2003;29(6):517-519. PubMed PMID: 15748305
  42. 42. Buhtoiarov IN, Neal ZC, Gan J, Buhtoiarova TN, Patankar MS, Gubbels JA, et al. Differential internalization of hu14.18-IL2 immunocytokine by NK and tumor cell: impact on conjugation, cytotoxicity, and targeting. Journal of Leukocyte Biology. 2011;89(4):625-638 PubMed PMID: 21248148. Pubmed Central PMCID: 3058817. Epub 2011/01/21. eng
  43. 43. Buhtoiarov IN, Sondel PM, Wigginton JM, Buhtoiarova TN, Yanke EM, Mahvi DA, et al. Anti-tumour synergy of cytotoxic chemotherapy and anti-CD40 plus CpG-ODN immunotherapy through repolarization of tumour-associated macrophages. Immunology. 2011;132(2):226-239. PubMed PMID: 21039467. Pubmed Central PMCID: 3050446. Epub 2010/11/03. eng
  44. 44. Gubbels JA, Gadbaw B, Buhtoiarov IN, Horibata S, Kapur AK, Patel D, et al. Ab-IL2 fusion proteins mediate NK cell immune synapse formation by polarizing CD25 to the target cell-effector cell interface. Cancer Immunology, Immunotherapy. 2011;60(12): 1789-1800. PubMed PMID: 21792658. Epub 2011/07/28. Eng
  45. 45. Johnson EE, Buhtoiarov IN, Baldeshwiler MJ, Felder MA, Van Rooijen N, Sondel PM, et al. Enhanced T-cell-independent antitumor effect of cyclophosphamide combined with anti-CD40 mAb and CpG in mice. Journal of Immunotherapy. 2011;34(1):76-84. PubMed PMID: 21150715. Pubmed Central PMCID: 3031426. Epub 2010/12/15. eng
  46. 46. Barbuto JAM, Ensina LFC, Neves AR, Bergami-Santos PC, Leite KRM, Marques R, et al. Dendritic cell–tumor cell hybrid vaccination for metastatic cancer. Cancer Immunology, Immunotherapy. 2004;53(12):1111-1118
  47. 47. Baleeiro RB, Anselmo LB, Soares FA, Pinto CA, Ramos O, Gross JL, et al. High frequency of immature dendritic cells and altered in situ production of interleukin-4 and tumor necrosis factor-alpha in lung cancer. Cancer Immunology, Immunotherapy. 2008;57(9):1335-1345. PubMed PMID: 18286287. Epub 2008/02/21. eng
  48. 48. Demaria S, Pikarsky E, Karin M, Coussens LM, Chen YC, El-Omar EM, et al. Cancer and inflammation: Promise for biologic therapy. Journal of Immunotherapy. 2010;33(4):335-351. PubMed PMID: 20386472. Pubmed Central PMCID: 2941912. Epub 2010/04/14. eng
  49. 49. Balwit J, Hwu P, Urba W, Marincola F. The iSBTc/SITC primer on tumor immunology and biological therapy of cancer: A summary of the 2010 program. Journal of Translational Medicine. 2011;9(1):18. PubMed PMID. DOI: 10.1186/1479-5876-9-18
  50. 50. Reiman JM, Kmieciak M, Manjili MH, Knutson KL. Tumor immunoediting and immunosculpting pathways to cancer progression. Seminars in Cancer Biology. 2007;17(4):275-287
  51. 51. Berezhnaya NM. Interaction between tumor and immune system: The role of tumor cell biology. Experimental Oncology. 2010;32(3):159-166. PubMed PMID: 21403611. Epub 2011/03/16. eng
  52. 52. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to Cancer. Annual Review of Immunology. 2011;29(1):235-271
  53. 53. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFN[gamma] and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410(6832):1107-1111
  54. 54. Frucht DM, Fukao T, Bogdan C, Schindler H, O'Shea JJ, Koyasu S. IFN-γ production by antigen-presenting cells: Mechanisms emerge. Trends in Immunology. 2001;22(10):556-560
  55. 55. Ulloa L, Tracey KJ. The ‘cytokine profile’: A code for sepsis. Trends in Molecular Medicine. 2005;11(2):56-63
  56. 56. Lee H, Baek S, Joe S-J, Pyo S-N. Modulation of IFN-γ production by TNF-α in macrophages from the tumor environment: Significance as an angiogenic switch. International Immunopharmacology. 2006;6(1):71-78
  57. 57. Lo SZY, Steer JH, Joyce DA. TNF-[alpha] renders macrophages resistant to a range of cancer chemotherapeutic agents through NF-[kappa]B-mediated antagonism of apoptosis signalling. Cancer Letters. 2011;307(1):80-92
  58. 58. Watford WT, Moriguchi M, Morinobu A, O'Shea JJ. The biology of IL-12: Coordinating innate and adaptive immune responses. Cytokine & Growth Factor Reviews. 2003;14(5):361-368
  59. 59. Xu M, Mizoguchi I, Morishima N, Chiba Y, Mizuguchi J, Yoshimoto T. Regulation of antitumor immune responses by the IL-12 family cytokines, IL-12, IL-23, and IL-27. Clinical & Developmental Immunology. 2010;2010:1-10. PubMed PMID: 20885915. Pubmed Central PMCID: 2946577. Epub 2010/10/05. eng
  60. 60. Heusinkveld M, de Vos van Steenwijk PJ, Goedemans R, Ramwadhdoebe TH, Gorter A, MJP W, et al. M2 macrophages induced by prostaglandin E2 and IL-6 from cervical carcinoma are switched to activated M1 macrophages by CD4+ Th1 cells. The Journal of Immunology. 2011;187(3):1157-1165
  61. 61. Ouyang W, Rutz S, Crellin NK, Valdez PA, Hymowitz SG. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annual Review of Immunology. 2011;29(1):71-109
  62. 62. Dhodapkar KM, Barbuto S, Matthews P, Kukreja A, Mazumder A, Vesole D, et al. Dendritic cells mediate the induction of polyfunctional human IL17-producing cells (Th17-1 cells) enriched in the bone marrow of patients with myeloma. Blood. 2008;112(7):2878-2885. PubMed PMID: PMC2556623
  63. 63. Inozume T, K-i H, Wang QJ, Yang JC. IL-17 secreted by tumor reactive T cells induces IL-8 release by human renal cancer cells. Journal of Immunotherapy. 2009;32(2):109-117. PubMed PMID: 00002371-200902000-00002
  64. 64. Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochimica et Biophysica Acta. 2014;1843(11):2563-2582. PubMed PMID: 24892271
  65. 65. Parihar JS, Tunuguntla HS. Role of chemokines in renal cell carcinoma. Reviews in Urology. 2014;16(3):118-121. PubMed PMID: 25337041. Pubmed Central PMCID: 4191631
  66. 66. Mikami S, Mizuno R, Kosaka T, Saya H, Oya M, Okada Y. Expression of TNF-alpha and CD44 is implicated in poor prognosis, cancer cell invasion, metastasis and resistance to the sunitinib treatment in clear cell renal cell carcinomas. International Journal of Cancer [Journal International du Cancer]. 2015;136(7):1504-1514. PubMed PMID: 25123505
  67. 67. Petrella BL, Vincenti MP. Interleukin-1beta mediates metalloproteinase-dependent renal cell carcinoma tumor cell invasion through the activation of CCAAT enhancer binding protein beta. Cancer Medicine. 2012;1(1):17-27. PubMed PMID: 23342250. Pubmed Central PMCID: 3544428
  68. 68. O'Neill LA. The interleukin-1 receptor/toll-like receptor superfamily: 10 years of progress. Immunological Reviews. 2008;226:10-18. PubMed PMID: 19161412
  69. 69. Elzinga BM, Twomey C, Powell JC, Harte F, McCarthy JV. Interleukin-1 receptor type 1 is a substrate for gamma-secretase-dependent regulated intramembrane proteolysis. The Journal of Biological Chemistry. 2009;284(3):1394-1409. PubMed PMID: 18996842
  70. 70. Baker SJ, Reddy EP. Modulation of life and death by the TNF receptor superfamily. Oncogene. 1998;17(25):3261-3270. PubMed PMID: 9916988
  71. 71. Taga T, Kishimoto T. Gp130 and the interleukin-6 family of cytokines. Annual Review of Immunology. 1997;15:797-819. PubMed PMID: 9143707
  72. 72. Goette NP, Lev PR, Heller PG, Glembotsky AC, Chazarreta CD, Salim JP, et al. Abnormal regulation of soluble and anchored IL-6 receptor in monocytes from patients with essential thrombocythemia. Experimental Hematology. 2010;38(10):868-76 e1. PubMed PMID: 20600579
  73. 73. Yoshida N, Ikemoto S, Narita K, Sugimura K, Wada S, Yasumoto R, et al. Interleukin-6, tumour necrosis factor alpha and interleukin-1beta in patients with renal cell carcinoma. British Journal of Cancer. 2002;86(9):1396-1400. PubMed PMID: 11986770. Pubmed Central PMCID: 2375361
  74. 74. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883-899 PubMed PMID: 20303878. Pubmed Central PMCID: 2866629
  75. 75. Ikemoto S, Yoshida N, Narita K, Wada S, Kishimoto T, Sugimura K, et al. Role of tumor-associated macrophages in renal cell carcinoma. Oncology Reports. 2003;10(6):1843-1849. PubMed PMID: 14534706
  76. 76. Dinarello CA. Why not treat human cancer with interleukin-1 blockade? Cancer Metastasis Reviews. 2010;29(2):317-329. PubMed PMID: 20422276. Pubmed Central PMCID: 2865633
  77. 77. Burns JM, Summers BC, Wang Y, Melikian A, Berahovich R, Miao Z, et al. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. The Journal of Experimental Medicine. 2006;203(9):2201-2213. PubMed PMID: 16940167. Pubmed Central PMCID: 2118398
  78. 78. Kulbe H, Hagemann T, Szlosarek PW, Balkwill FR, Wilson JL. The inflammatory cytokine tumor necrosis factor-alpha regulates chemokine receptor expression on ovarian cancer cells. Cancer Research. 2005;65(22):10355-10362. PubMed PMID: 16288025
  79. 79. Xin H, Zhang C, Herrmann A, Du Y, Figlin R, Yu H. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Research. 2009;69(6):2506-2513. PubMed PMID: 19244102. Pubmed Central PMCID: 2664264
  80. 80. Moser B, Wolf M, Walz A, Loetscher P. Chemokines: Multiple levels of leukocyte migration control. Trends in Immunology. 2004;25(2):75-84. PubMed PMID: 15102366
  81. 81. Palczewski K, Kumasaka T, Hori T, Behnke CA, Motoshima H, Fox BA, et al. Crystal structure of rhodopsin: A G protein-coupled receptor. Science. 2000;289(5480):739-745. PubMed PMID: 10926528
  82. 82. Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2010;16(11):2927-2931. PubMed PMID: 20484021
  83. 83. Kalatskaya I, Berchiche YA, Gravel S, Limberg BJ, Rosenbaum JS, Heveker N. AMD3100 is a CXCR7 ligand with allosteric agonist properties. Molecular Pharmacology. 2009;75(5):1240-1247. PubMed PMID: 19255243
  84. 84. McAllister SS, Weinberg RA. Tumor-host interactions: A far-reaching relationship. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2010;28(26):4022-4028. PubMed PMID: 20644094
  85. 85. Putney JW, Broad LM, Braun FJ, Lievremont JP, Bird GS. Mechanisms of capacitative calcium entry. Journal of Cell Science. 2001;114(Pt 12):2223-2229. PubMed PMID: 11493662
  86. 86. Mukherjee D, Zhao J. The role of chemokine receptor CXCR4 in breast cancer metastasis. American Journal of Cancer Research. 2013;3(1):46-57. PubMed PMID: 23359227. Pubmed Central PMCID: 3555200
  87. 87. Goda S, Inoue H, Umehara H, Miyaji M, Nagano Y, Harakawa N, et al. Matrix metalloproteinase-1 produced by human CXCL12-stimulated natural killer cells. The American Journal of Pathology. 2006;169(2):445-458. PubMed PMID: 16877347. Pubmed Central PMCID: 1698790
  88. 88. Vila-Coro AJ, Rodriguez-Frade JM, Martin De Ana A, Moreno-Ortiz MC, Martinez AC, Mellado M. The chemokine SDF-1alpha triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology. 1999;13(13):1699-1710. PubMed PMID: 10506573
  89. 89. Sun X, Cheng G, Hao M, Zheng J, Zhou X, Zhang J, et al. CXCL12/CXCR4/CXCR7 chemokine axis and cancer progression. Cancer Metastasis Reviews. 2010;29(4):709-722. PubMed PMID: 20839032. Pubmed Central PMCID: 3175097
  90. 90. Schrader AJ, Lechner O, Templin M, Dittmar KE, Machtens S, Mengel M, et al. CXCR4/CXCL12 expression and signalling in kidney cancer. British Journal of Cancer. 2002;86(8):1250-1256. PubMed PMID: 11953881. Pubmed Central PMCID: 2375348
  91. 91. Ierano C, Santagata S, Napolitano M, Guardia F, Grimaldi A, Antignani E, et al. CXCR4 and CXCR7 transduce through mTOR in human renal cancer cells. Cell Death & Disease. 2014;5:e1310. PubMed PMID: 24991762. Pubmed Central PMCID: 4123065
  92. 92. Curnock AP, Logan MK, Ward SG. Chemokine signalling: Pivoting around multiple phosphoinositide 3-kinases. Immunology. 2002;105(2):125-136. PubMed PMID: 11872087. Pubmed Central PMCID: 1782650
  93. 93. Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441(7092):431-436. PubMed PMID: 16724054
  94. 94. Ye RD. Regulation of nuclear factor kappaB activation by G-protein-coupled receptors. Journal of Leukocyte Biology. 2001;70(6):839-848. PubMed PMID: 11739545
  95. 95. Lee BC, Lee TH, Avraham S, Avraham HK. Involvement of the chemokine receptor CXCR4 and its ligand stromal cell-derived factor 1alpha in breast cancer cell migration through human brain microvascular endothelial cells. Molecular Cancer Research: MCR. 2004;2(6):327-338. PubMed PMID: 15235108
  96. 96. Johnson-Holiday C, Singh R, Johnson EL, Grizzle WE, Lillard JW Jr, Singh S. CCR9-CCL25 interactions promote cisplatin resistance in breast cancer cell through Akt activation in a PI3K-dependent and FAK-independent fashion. World Journal of Surgical Oncology. 2011;9:46. PubMed PMID: 21539750. Pubmed Central PMCID: 3110128
  97. 97. Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes & Development. 1998;12(22):3499-3511. PubMed PMID: 9832503. Pubmed Central PMCID: 317244
  98. 98. Allan LA, Morrice N, Brady S, Magee G, Pathak S, Clarke PR. Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nature Cell Biology. 2003;5(7):647-654. PubMed PMID: 12792650
  99. 99. Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 1999;286(5443):1358-1362. PubMed PMID: 10558990
  100. 100. Kyriakis JM. MAP kinases and the regulation of nuclear receptors. Science's STKE: Signal Transduction Knowledge Environment. 2000;2000(48):pe1. PubMed PMID: 11752606
  101. 101. Zagzag D, Lukyanov Y, Lan L, Ali MA, Esencay M, Mendez O, et al. Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: Implications for angiogenesis and glioma cell invasion. Laboratory Investigation; A Journal of Technical Methods and Pathology. 2006;86(12):1221-1232. PubMed PMID: 17075581
  102. 102. van Golen KL, Ying C, Sequeira L, Dubyk CW, Reisenberger T, Chinnaiyan AM, et al. CCL2 induces prostate cancer transendothelial cell migration via activation of the small GTPase Rac. Journal of Cellular Biochemistry. 2008;104(5):1587-1597. PubMed PMID: 18646053
  103. 103. Boldajipour B, Mahabaleshwar H, Kardash E, Reichman-Fried M, Blaser H, Minina S, et al. Control of chemokine-guided cell migration by ligand sequestration. Cell. 2008;132(3):463-473. PubMed PMID: 18267076
  104. 104. Rajagopal S, Kim J, Ahn S, Craig S, Lam CM, Gerard NP, et al. Beta-arrestin-but not G protein-mediated signaling by the "decoy" receptor CXCR7. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(2):628-632. PubMed PMID: 20018651. Pubmed Central PMCID: 2818968
  105. 105. Kovaleva OV, Samoilova DV, Shitova MS, Gratchev A. Tumor associated macrophages in kidney Cancer. Analytical Cellular Pathology. 2016;2016:9307549. PubMed PMID: 27807511. Pubmed Central PMCID: 5078639
  106. 106. Banumathy G, Cairns P. Signaling pathways in renal cell carcinoma. Cancer Biology & Therapy. 2010;10(7):658-664. PubMed PMID: 20814228. Pubmed Central PMCID: 3093809
  107. 107. Ghidini M, Petrelli F, Ghidini A, Tomasello G, Hahne JC, Passalacqua R, et al. Clinical development of mTor inhibitors for renal cancer. Expert Opinion on Investigational Drugs. 2017;26(11):1229-1237. PubMed PMID: 28952411
  108. 108. Song M. Recent developments in small molecule therapies for renal cell carcinoma. European Journal of Medicinal Chemistry. 2017;142:383-392. PubMed PMID: 28844802
  109. 109. Minasian LM, Motzer RJ, Gluck L, Mazumdar M, Vlamis V, Krown SE. Interferon alfa-2a in advanced renal cell carcinoma: Treatment results and survival in 159 patients with long-term follow-up. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 1993;11(7):1368-1375. PubMed PMID: 8315435
  110. 110. Yagoda A, Bander NH. Failure of cytotoxic chemotherapy, 1983-1988, and the emerging role of monoclonal antibodies for renal cancer. Urologia Internationalis. 1989;44(6):338-345. PubMed PMID: 2696193
  111. 111. Iguchi M, Matsumoto M, Hojo K, Wada T, Matsuo Y, Arimura A, et al. Antitumor efficacy of recombinant human interleukin-2 combined with sorafenib against mouse renal cell carcinoma. Japanese Journal of Clinical Oncology. 2009;39(5):303-309. PubMed PMID: 19336449
  112. 112. Rizza P, Moretti F, Belardelli F. Recent advances on the immunomodulatory effects of IFN-alpha: Implications for cancer immunotherapy and autoimmunity. Autoimmunity. 2010;43(3):204-209. PubMed PMID: 20187707
  113. 113. Ishibashi K, Haber T, Breuksch I, Gebhard S, Sugino T, Kubo H, et al. Overriding TKI resistance of renal cell carcinoma by combination therapy with IL-6 receptor blockade. Oncotarget. 2017;8(33):55230-55245. PubMed PMID: 28903416. Pubmed Central PMCID: 5589655
  114. 114. Oguro T, Ishibashi K, Sugino T, Hashimoto K, Tomita S, Takahashi N, et al. Humanised antihuman IL-6R antibody with interferon inhibits renal cell carcinoma cell growth in vitro and in vivo through suppressed SOCS3 expression. European Journal of Cancer. 2013;49(7):1715-1724. PubMed PMID: 23274199
  115. 115. Harrison ML, Obermueller E, Maisey NR, Hoare S, Edmonds K, Li NF, et al. Tumor necrosis factor alpha as a new target for renal cell carcinoma: Two sequential phase II trials of infliximab at standard and high dose. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2007;25(29):4542-4549. PubMed PMID: 17925549
  116. 116. Kobak S, Karaarslan A, Aktakka Y. Renal cell carcinoma in a patient with rheumatoid arthritis treated with adalimumab. Current Drug Safety. 2014;9(1):69-72. PubMed PMID: 24191905
  117. 117. Triozzi PL, Kim JA, Martin EW, Young DC, Benzies T, Villasmil PM. Phase I trial of escalating doses of interleukin-1 beta in combination with a fixed dose of interleukin-2. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 1995;13(2):482-489. PubMed PMID: 7844609
  118. 118. Burger JA, Burger M, Kipps TJ. Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells. Blood. 1999;94(11):3658-3667. PubMed PMID: 10572077
  119. 119. Santagata S, Napolitano M, D'Alterio C, Desicato S, Maro SD, Marinelli L, et al. Targeting CXCR4 reverts the suppressive activity of T-regulatory cells in renal cancer. Oncotarget. 2017;8(44):77110-77120. PubMed PMID: 29100374. Pubmed Central PMCID: 5652768
  120. 120. Kavsak PA, Henderson M, Moretto P, Hirte H, Evans K, Wong D, et al. Biochip arrays for the discovery of a biomarker surrogate in a phase I/II study assessing a novel anti-metastasis agent. Clinical Biochemistry. 2009;42(10-11):1162-1165. PubMed PMID: 19389390
  121. 121. Atkins MB, Clark JI, Quinn DI. Immune checkpoint inhibitors in advanced renal cell carcinoma: Experience to date and future directions. Annals of Oncology : Official Journal of the European Society for Medical Oncology. 2017;28(7):1484-1494. PubMed PMID: 28383639
  122. 122. McDermott DF, Drake CG, Sznol M, Choueiri TK, Powderly JD, Smith DC, et al. Survival, durable response, and long-term safety in patients with previously treated advanced renal cell carcinoma receiving Nivolumab. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2015;33(18):2013-2020. PubMed PMID: 25800770. Pubmed Central PMCID: 4517051
  123. 123. Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Molecular Immunology. 2015;67(2 Pt A):4-17. PubMed PMID: 25749122
  124. 124. Nivolumab Plus Ipilimumab Has Antitumor Activity in Metastatic RCC. Cancer Discovery. 2017;7(9):OF7. PubMed PMID: 28710099
  125. 125. Hao C, Tian J, Liu H, Li F, Niu H, Zhu B. Efficacy and safety of anti-PD-1 and anti-PD-1 combined with anti-CTLA-4 immunotherapy to advanced melanoma: A systematic review and meta-analysis of randomized controlled trials. Medicine. 2017;96(26):e7325. PubMed PMID: 28658143. Pubmed Central PMCID: 5500065
  126. 126. Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: Mechanism, combinations, and clinical outcome. Frontiers in Pharmacology. 2017;8:561. PubMed PMID: 28878676. Pubmed Central PMCID: 5572324
  127. 127. Weinstock M, McDermott D. Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma. Therapeutic Advances in Urology. 2015;7(6):365-377. PubMed PMID: 26622321. Pubmed Central PMCID: 4647139
  128. 128. Mazza C, Escudier B, Albiges L. Nivolumab in renal cell carcinoma: Latest evidence and clinical potential. Therapeutic Advances in Medical Oncology. 2017;9(3):171-181. PubMed PMID: 28344662. Pubmed Central PMCID: 5349425
  129. 129. Rodriguez-Vida A, Hutson TE, Bellmunt J, Strijbos MH. New treatment options for metastatic renal cell carcinoma. ESMO Open. 2017;2(2):e000185 PubMed PMID: 28761748. Pubmed Central PMCID: 5519813
  130. 130. Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nature Communications. 2016;7:12624. PubMed PMID: 27571927. Pubmed Central PMCID: 5013615 provided the study drug. Some of the authors of this manuscript are employees of Genentech/Roche. The remaining authors declare no competing financial interests
  131. 131. Gill DM, Hahn AW, Hale P, Maughan BL. Overview of current and future first-line systemic therapy for metastatic clear cell renal cell carcinoma. Current Treatment Options in Oncology. 2018;19(1):6. PubMed PMID: 29368125
  132. 132. Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, et al. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. Journal for Immunotherapy of Cancer. 2015;3:14. PubMed PMID: 25901286. Pubmed Central PMCID: 4404644

Written By

Ana Marisa Chudzinski-Tavassi, Kátia Luciano Pereira Morais, Jean Gabriel de Souza and Roger Chammas

Submitted: 19 July 2017 Reviewed: 20 April 2018 Published: 05 November 2018