The immune system plays a critical role in preventing cancer development and progression. However, the complex network of cells and soluble factor that form the tumor microenvironment (TME) can dictate the differentiation of tumor-infiltrating leukocytes and shift the antitumor immune response into promoting tumor growth. With the advent of cancer immunotherapy, there has been a reinvigorated interest in defining how the TME shapes the antitumor immune response. This interest brought to light the microbiome as a novel player in shaping cancer immunosurveillance. Indeed, accumulating evidence now suggests that the microbiome may confer susceptibility or resistance to certain cancers and may influence response to therapeutics, particularly immune checkpoint inhibitors. As we move forward into the age of precision medicine, it is vital that we define the factors that influence the interplay between the triad immune system-microbiota-cancer. This knowledge will contribute to improve the therapeutic response to current approaches and will unravel novel targets for immunotherapy.

The interplay between the immune system and cancer is unquestionable. Considered a key factor during tumorigenesis, the immune response against cancer has been extensively studied over the last decade. While it is now clear that the immune system plays crucial roles in controlling tumor growth, in some instances the activity of immune cells can also favor cancer progression [1‒3]. Owing the improved understanding on the interaction between the immune system and cancer, in the last years we have witnessed a revolution in cancer treatment with the development of immunotherapies aimed at boosting or dampening key immune pathways to promote antitumor immune status [4‒6]. This recent development in cancer treatment also brought to light key factors that modulate the immune response to cancer and therefore the efficacy of immunotherapies. One such factor is the microbiome, the complete collection of microorganisms (including bacteria, viruses, archaea, and fungi) and their genomes that colonize the different surfaces of the human body, such as the gut [7]. Indeed, recent exciting data demonstrated the critical role of the gut microbiota in modulating the antitumor immune response and the efficacy of the recently developed checkpoint blockade immunotherapies across different cancer types [8‒11]. These data suggest that modulating the gut microbiota may represent a novel adjunct approach to current anticancer therapeutic modalities.

To understand how alterations in the composition and abundance of the microbiota, including of particular species that are commonly part of the human microbiota, influence cancer immune responses and the efficacy of immunotherapies requires a deep understanding of the key factors that govern the interaction between the triad immune system-microbiota-cancer. To help in this regard, here we discuss new advances in the understanding of the mechanisms underlying the interaction between the immune system and microbiota during carcinogenesis, as well as the factors whereby the microbiota impacts the immune response against cancer. While novel research is shedding light into the role of the virome and mycobiome in host immunity, the techniques currently in use to isolate and characterize bacterial communities of the microbiota are more developed and standardized. Therefore, in this review we will focus on the bacterial communities of the microbiota and discuss their impact in in host immunity and cancer.

A plethora of mechanisms control cellular growth and death. It is the failure in these processes, mostly caused by DNA mutations, that is the main cause for the development of aberrant cells that accumulate and form tumors [12, 13]. Despite these DNA mutations being a common feature of cellular growth and differentiation, they do not always cause cancer development. Indeed, the transformation of a normal cell into a malignant phenotype triggers intrinsic cell death pathways that limit the development of tumors [14‒16]. Accordingly, one of the hallmarks of human cancers is their tendency to evade programmed cell death [13]. Additionally, neoplastic transformation can be associated with significant molecular alterations that trigger immune responses to rapidly eliminate malignant cells. Indeed, mounting evidence both from mice and from clinical epidemiology suggest that the immune system functions as a significant barrier to tumor formation and progression [17]. The hypothesis that the immune system is capable of recognizing and eliminating tumor cells was first postulated by Sir Frank Burnett and is currently known as cancer immunosurveillance [18, 19].

The immune system comprises different lineages of cells, including macrophages, neutrophils, and lymphocytes, that act in a concerted manner to maintain homeostasis by protecting the host from external and, as postulated by Burnett, internal aggressions. Indeed, it has been now clearly demonstrated that the immune system detects and eliminates malignant cells including through antigen-specific responses initiated against antigens expressed by tumor cells [20‒22]. Seminal studies from Kaplan and others show that the absence of one or several components of the immune system increased the incidence of cancer, both in mice and in humans [23‒25]. One crucial observation that unraveled the relevance of the immune system in the fight against cancer was in mice lacking perforin, a component of cytolytic granules of cytotoxic T cells. In a set of different experimental approaches, perforin-dependent cytotoxicity was demonstrated to be a crucial mechanism of resistance to injected tumor cell lines but also to viral and chemical-induced carcinogenesis [26]. Also in mice, the blockade of interferon (IFN)-γ was associated with enhanced Meth-A tumor cell growth [27]. The critical role of the immune system in limiting carcinogenic events is also highlighted by several studies associating the presence of tumor-specific T cells in the blood, bone marrow, or tumors with good prognosis [28‒34]. Despite this, preclinical studies have also shown that preconditioning of mice with IFN-γ resulted in increased B16 melanoma colonization of the lungs [35]. This enhanced colonization was attributed to the effects of IFN-γ in B16 cells, which caused a decreased sensitivity of these cells to the activity of natural killer (NK) cells [35]. In line with these data is the observation that in vitro IFN-γ induces the expression of markers that are usually associated with more aggressive phenotypes of melanoma cells [36]. When taken together, these studies suggest that the protective effect of the immune responses may be, at lease in some cases, dependent upon the type of tumor [20]. Furthermore, the presence of different types of immune cells infiltrating the tumor has been associated with tumor progression [37‒40]. To help untangling the complex interaction between the immune response with tumor cells, in this section we will discuss the involvement of the immune system in tumor control or progression (Fig. 1).

Fig. 1.

The antitumor and pro-tumor immune mechanisms generated toward cancer cells. IFN, interferon; TNF, tumor necrosis factor; IL, interleukin; TGF, transforming growth factor; MDSCs, myeloid-derived suppressor cells; Tregs, regulatory T cells.

Fig. 1.

The antitumor and pro-tumor immune mechanisms generated toward cancer cells. IFN, interferon; TNF, tumor necrosis factor; IL, interleukin; TGF, transforming growth factor; MDSCs, myeloid-derived suppressor cells; Tregs, regulatory T cells.

Close modal

The Immune Response in Tumor Control

Cancers are heterogeneous masses of different types of cells, including tumor cells, immune cells, fibroblasts, and pericytes. This niche composed by cells and their by-products constitute the tumor microenvironment (TME). Soluble factors, such as cytokines, chemokines, and growth factors, mediate the intercellular communication between the different constituents of the TME [41].

Tumor-infiltrating leukocytes are an important component of the TME. Among these leukocytes, tumor-associated macrophages (TAMs) are one of the most represented cells [42]. TAMs differentiate locally from recruited peripheral blood monocytes in response to cytokines and growth factors produced by stromal and tumor cells in the TME [42]. In responses to these factors, and as a result of their plasticity, macrophages can polarize toward an M1- or M2-like phenotype [43, 44]. Usually, M1-like macrophages are differentiated by pro-inflammatory mediators including lipopolysaccharides and IFN-γ, while M2-like macrophages are differentiated by the immunomodulatory cytokines interleukin (IL)-4, IL-10, and IL-13, and transforming growth factor (TGF)-β [44]. It is important to note that these 2 macrophage phenotypes are a simplistic view of the intricate mechanisms underlying macrophage differentiation [45]. Indeed, not only macrophage polarization is not definitive, as it can be altered in response to the microenvironment [46], but these macrophage phenotypes are not mutually exclusive and M1- and M2-like macrophages can coexist in the TME which is important to prevent dysregulated responses [45].

In the initial steps of the immune response, M1-like macrophages have been shown to play antitumor roles through the production of pro-inflammatory cytokines such as IL-12, which promotes the differentiation of T-helper type 1 (Th1) cells [42, 47]. These cells produce IFN-γ and other pro-inflammatory cytokines and chemokines that enhance antigen presentation and promote the expression of inducible nitric oxide synthase, an enzyme actively associated with cytostatic and growth inhibitory effects [48]. On the other hand, M2-like macrophages are characterized by their pro-tumor role as discussed below [49].

In addition to macrophages, neutrophils are another important cellular component of the TME that has received increasing attention over the past years. In this regard, recent data show that neutrophils can also adopt 2 distinct phenotypes in response to the cytokines of the TME [50]. As discussed above for TAMs, neutrophils can also adopt antitumor (N1-like) or pro-tumor (N2-like) phenotypes [51]. N1-like neutrophils contribute to the antitumor immune response through the production of antimicrobial molecules with cytotoxicity against cancer cells [52‒54], the inhibition of TGF-β [50], or by actively promoting the recruitment and stimulation of T cell proliferation [55] and IFN-γ secretion [56]. Indeed, recent studies using human samples of early-stage colorectal cancer (CRC) have associated the infiltration of neutrophils with good prognosis and increased overall survival [57, 58]. Furthermore, neutrophils have recently emerged as important targets of immunotherapy [59]. For example, a recent study from Zhang et al. [60] found that the ablation of TOLLIP, an innate immune-cell modulator, from neutrophils enhances the antitumor immune response in a mouse model of CRC induced by azoxymethane-dextran sulfate sodium salt. This study showed that TOLLIP-deficient mice exhibited a marked reduction in both microscopic and macroscopic polyps when compared with WT mice. This phenotype was associated with an increased expression of CD80 and a downregulation of immunosuppressive molecules by TOLLIP-deficient neutrophils, which enhanced the activation and survival of T cell [60]. While these data suggest a crucial role of neutrophils in the antitumor immune response, most studies investigating the role of these cells in cancer have relied on animal models or circulating human neutrophils. As such, only limited information is available on the roles of tumor-associated neutrophils (TANs) in cancer patients. Therefore, further studies are required to better define the mechanisms and pathways whereby neutrophils modulate tumor immunity.

NK cells are another innate cellular component of the TME [61]. This population has been shown to play critical roles in the elimination of abnormal cells [61, 62] both in mice [63, 64] and in humans [65‒67]. The effector functions of NK cells to eliminate tumor cells relies in the expression of molecules that can act either directly on tumor cells, such as perforins [68‒70] and granzymes [69, 70], or initiate the recruitment of other immune cells which contribute to tumor clearance [71‒73].

In addition to the innate cellular components, lymphocytes are another important presence in the TME. The T cell populations that infiltrate the TME include CD4+ T cells, specifically the phenotypes Th1, Th17, and regulatory T cells (Tregs), and CD8+ cytotoxic T cells. The antitumor mechanism of action of CD8+ T cells is particularly important as it involves antigen-specific cytotoxicity, in addition to IFN-γ and tumor necrosis factor (TNF)-α secretion [74]. This mechanism is supported by CD4+ T cells, particularly Th1 cells through the production of IL-2, IFN-γ, and TNF-α [74]. In some circumstances, Th2 cells have also been also showed to prevent tumor progression [75] mainly though the recruitment of eosinophils. Indeed, the production of IL-18 by eosinophils has been shown to mediate the death of Colo-205 cells through the upregulation of adhesion molecules that facilitate the interaction between effector and target cells [76]. Additionally, eosinophils can also produce cytotoxins, namely, granzyme-A, that promote tumor elimination [77]. In addition to conventional T cells, NK T cells also participate in tumor clearance [78]. Their effector mechanism is dependent upon the secretion of granzymes and perforin that lyse tumor cells [79]. Additionally, NK-T cells also secrete IFN-γ, which promotes the activation of cells CD8+ T cells as well as the generation of M1-like macrophages [74]. The relevance of these populations in tumor control is highlighted by data showing that tumor-infiltrating lymphocytes are generally associated with good prognosis [74, 80].

Together, the above data show that protection conferred by the immune system is critical to prevent cancer development. Indeed, in recent years we have witnessed an increased development of cancer treatments that target different components of the immune system, including dendritic cell [81] and checkpoint blockade therapies [82‒84]. However, the modulation that immune cells are subjected to in the TME can promote, rather than eliminate, cancer (Fig. 1).

The Immune Response in Tumor Progression

As discussed above, the immune system plays a crucial role in controlling tumor development and progression. However, tumors can develop even in the presence of a functional immune system suggesting that tumor cells and the microenvironment that they create can modulate the antitumor function of the immune response. Indeed, the ability of tumor cells to avoid immune destruction has been now considered in the updated version of the “hallmarks of cancer” [13].

The induction of immune suppression mechanisms is a complex process that requires the involvement of tumor and immune cells that act synergistically to dampen antitumor immune response. As discussed in the previous section, the differentiation of TAMs in the TME can contribute for their pro-tumor characteristics [42, 85]. In this regard, several meta-analysis have clearly demonstrated that an increase in the number of TAMs is generally associated with poor prognosis [86, 87]. During cancer progression, several mechanisms have been identified to overcome antitumor response by TAMs including the secretion of prostaglandin E2 [88], the immunosuppressive cytokine IL-10 [89], the angiogenic factors such as IL-1β, that through upregulation of HIF-1α protein increases vascular endothelial growth factor (VEGF) secretion [90], VEGF [91], endothelin-2 [92], and epidermal growth factor family ligands [93]. Additionally, TAMs may also play a role in tumor aggressiveness by releasing matrix metalloproteases (namely, MMP-2 and MMP-9) that damage the extracellular matrix and the basement membrane, facilitating tumor invasion and metastases [93‒95]. Using a mouse model of colon carcinoma, the expression of arginase and production of nitrogen monoxide by TAMs were also shown to contribute to tumor progression by inducing apoptosis of CD8+ cytotoxic T cells [96].

TANs may also adopt an immunosuppressive phenotype in the context of tumors, including by producing chemokines that promote the recruitment of Tregs [50, 97]. N2-like neutrophils also produce cathepsin G and arginase [51], further promoting immunosuppressive status. As described above, most studies on the role of neutrophils in cancer have relied on mouse models. As such, it is still not clear if the N1-/N2-like profile described in mouse models can be directly applied to humans. However, some of the genes reported to be associated with the phenotype and function of neutrophils in mice have also been reported in humans. For example, the chemokines CCL2 and CCL17 that were originally found as part of the N2-like neutrophils signature in mice were also associated with increased tumor progression in humans [98, 99]. Accordingly, patients with low CCL2 or CCL17 TAN counts had substantially better outcomes than those with higher numbers of these cells [97]. Furthermore, patients diagnosed in advance stages of cancer have been reported to present neutrophilia, which was associated with poor prognosis [100, 101]. Additionally, TANs produce several molecules that promote invasion and metastasis including MMPs and VEGF [102, 103]. Accordingly, it has been shown that neutrophils promoted lung metastases by breast cancer cells [104] and their depletion in murine models of melanoma and fibrosarcoma reverts tumor growth, angiogenesis, and metastasis [104, 105]. More studies are required to determine the impact of neutrophils, and neutrophil polarization, particularly in human cancers.

In addition to TAMs and TANs, myeloid-derived suppressor cells (MDSCs) are another population associated with the suppression of antitumor immune response [106]. The suppressive capacity of these cells appears to be enhanced as they infiltrate tumors, pointing to the crucial role of the TME in modulating their function [107]. MDSCs are characterized by their ability to suppress the immune response through different mechanisms, including the inhibition of T cell functionality associated with the expression and activity of arginase-1 [108, 109], promoting the differentiation of Tregs [110], and the differentiation of pro-tumor TAMs [111, 112]. Recent work in humans also showed that MDSCs can promote angiogenesis via MMP-9 secretion [113].

In the context of the adaptive immune response, Tregs have been identified as central suppressors of antitumor immune responses [114, 115]. The immunosuppressive function of Tregs is mediated through the secretion of IL-10 and TGF-β, which hamper the activity of effector T cells and their ability to eliminate tumor cells. Accordingly, increased accumulation of Tregs in the TME associates with poor prognosis, in multiple types of cancer [80, 116, 117]. In addition to Tregs, Th2 response has also been demonstrated to favor tumor progression through different mechanisms, including the production of IL-10 [118‒120] that inhibits antitumor immune responses as well as the production of arginase-1 by M2-like macrophages [121], differentiated in a context of a Th-2 response, that inhibits cytotoxic T cell activity.

While the data described above show that there are multiple immune mechanisms associated with the inhibition of antitumor immune responses, it is important to note that this inhibition is not only accomplished through suppressive mechanisms. Indeed, an exaggerated immune response can promote dysregulated inflammation which favors cancer development. It was Virchow that suggested the first association between inflammation and cancer, over 150 years ago [122]. After this initial observation, several pieces of evidence confirmed the association between prolonged inflammation and increased risk of cancer [123, 124]. As the inflammatory process promotes the further recruitment of inflammatory immune cells to damaged sites, the constant production of angiogenic factors, pro-inflammatory cytokines, and reactive oxygen species creates the conditions to drive normal cells into tumorigenic [125, 126]. As discussed later in this review, infection by pathogens instigates inflammation that promotes the development of cancer. Indeed, infections have been estimated to be the cause of approximately 20% of all cancers [127, 128].

After birth, microorganisms colonize all the surfaces of the human body exposed to the external environment. These communities of microorganisms (microbiota) and their collective genomes (microbiome) have long been recognized to play crucial roles in the digestion of nutrients, the production of vitamins, and other essential molecules [129] However, more recent data show that the microbiota plays a critical role in the development and function of the immune system [130‒132].

The development of new methodologies for nucleic acid sequencing unraveled the diversity and function of the microbiota, particularly bacterial communities [133]. These methodologies showed that the composition of the gut microbiota stabilizes after 3 years of age and remains constant during adolescence and adulthood [134]. Hosting >1,000 different bacterial species, the gastrointestinal tract harbors the most complex and well-studied microbiota of the human body [134]. The upper part of the gastrointestinal track, including stomach, duodenum, and jejunum, is enriched in aerobic gram-positive bacteria from the Lactobacillus and Enterococcus genera. In the ileum, the concentration of bacteria increases, with the representation of coliforms. The distal region of gastrointestinal tract (cecum and colon) harbors the most diversity with species of bacteria belonging to the genera Bacteroidetes, Proteobacteria, and Actinobacteria [134].

Although the microbiota composition remains relatively stable, changes in lifestyle, such as diet [135, 136], or consumption of antibiotics [137, 138] can lead to microbiota imbalances, a condition known as dysbiosis [139]. With the recent body of literature pointing toward the critical role of the microbiota as determinant of health or pathologic conditions, including cancer [140], in the following sections we will discuss the interaction between gut microbiota and cancer development or control.

The Microbiota in Tumor Control

The microbiota plays crucial roles in controlling tumor development and progression through the production of metabolites that fuel the immune responses or promote the development of antitumor environments (Fig. 2). Specifically, fiber-rich foods have been suggested to have health benefits through the maintenance of a healthy microbiota capable of production of short-chain fatty acids (SCFA) [141‒143]. Accordingly, recent data show that butyrate and propionate, 2 SCFA produced by different microbiota species, display antitumor capacity against CRC [144] or hepatic cancer [145]. The protective capacities of these metabolites have been shown to be mediated directly in tumor cells or indirectly by modulating the immune response and the TME. Specifically in tumor cells, several in vitro studies show that these metabolites inhibit the activity of histone deacetylases, which are critical to control of gene transcription [146‒148]. In line with these data, Zagato et al. [149] elegantly showed that the endogenous murine microbiota member Faecalibaculum rodentium and its human homolog, Holdemanella biformis, were involved in CRC prevention. This study showed that the production of SCFA, specifically butyrate, was inhibiting calcineurin-mediated NFATc3 transcription factor activation through its inhibitory effect at the histone deacetylase level [149]. These data corroborated the results from Peuker et al. [150] that identified the contribution of calcineurin activity to tumor growth and proliferation. In what regard the indirect effects of SCFA, a recent study showed that butyrate plays a key role in regulating colonic inflammation and therefore CRC development [151]. In this study, the protective role of butyrate was mediated by promoted anti-inflammatory properties in colonic macrophages and dendritic cells which promoted the differentiation of Treg cells and IL-10-producing T cells [151]. Accordingly, mice deficient in the receptor for butyrate in the colon (GPR109A encoded by Niacr1) were more susceptible to development of colon cancer. Importantly, Niacin, a pharmacological Gpr109a agonist, suppressed colitis and colon cancer [151].

Fig. 2.

The antitumor and pro-tumor impact of the microbiota in cancer.

Fig. 2.

The antitumor and pro-tumor impact of the microbiota in cancer.

Close modal

In addition to SCFA, microbiota also produce other molecules with antitumor potential. For example, the production of pyrodoxine, from the vitamin B group, by a wide group of bacteria stimulates the antitumor immune response [152, 153]. Additionally, Konishi et al. [154] showed that the production of ferricrhome by Lactobacillus casei induces the apoptosis of colon-derived tumor cells through the activation of the JNK-mediated apoptosis. Lactic acid-producing bacteria have also been shown to play key roles in controlling tumor cells growth by stimulating antitumor immune responses [155‒157]. Specifically, the supplementation of L. casei in a chemical-induced model of intestine injury was associated with increased production of lactate which decreased the production of myeloperoxidases and TNF-α [158]. These data were corroborated by another study wherein L. casei supplementation improved chronic inflammatory bowel disease by downregulating the production of pro-inflammatory cytokines such as IL-6 and IFN-γ by intestinal lamina propria mononuclear cells [159].

Another important mechanism whereby microbiota promotes regulation of inflammation, and therefore tumor control, is through the production of metabolites that are agonists of the aryl hydrocarbon receptor (AhR). This receptor is expressed at barrier sites, such as gastrointestinal tract, acting as a sensor of environmental chemicals, including dietary compounds. Recent data show that mice treated with AhR antagonists develop severe symptoms upon induction of colitis and that patients with ulcerative colitis had reduced expression of AhR [160]. Alexeev et al. [161] showed that the levels of indole-3-propionic acid, a metabolite of the microbial tryptophan catabolism recently identified as an AhR agonist, were reduced in both ulcerative colitis patients and mice. Importantly, the supplementation of mice with indole-3-propionic acid enhanced the production of IL-10 and inhibited the production of pro-inflammatory cytokines [161].

While the above data show that microbiota metabolites can regulate inflammation, the expression of AhR can be an indicator of poor prognosis in certain types of cancer [162‒165]. Accordingly, Shimba et al. [163] reported that A549 tumor cells expressing high levels of AhR had accelerated cell growth due to shortening late M to S phases of cell cycle. Additionally, Yin et al. [164] described shortened cell cycles and increased proliferation of gastric cancer cell lines due to the activity of the AhR. The activity of AhR also blocked the efficiency of tyrosine kinase inhibitors on lung adenocarcinoma, thus favoring tumor growth [166]. Together, these data show that while the activity of the AhR may be beneficial in regulating inflammatory contexts it can also enhance tumor cell growth and resistance to therapy.

The Microbiota in Tumor Progression

As discussed above, uncontrolled chronic inflammatory contexts are characterized by increased production of pro-inflammatory cytokines and other molecules that promote tumor development and progression (Fig. 2). The microbiota play a key role in these contexts as the outgrowth of certain commensal bacteria has been associated with chronic inflammation and cancer development [167‒169]. Indeed, infectious agents are currently estimated to be responsible for ∼20% of human cancers [127, 128]. While a large percentage of these occurrences are due to viral infections, such as the human papillomavirus (HPV) that causes cervical cancer, the contribution of bacteria is usually relegated to a second plane [170, 171]. However, in recent years an increasing number of studies have identified an important role of bacterial infections in carcinogenesis [167, 172]. As an example, bacterial vaginosis has been associated with the prevalence of HPV infection [173‒175]. Although the mechanisms underlying this association are not fully understood, it is likely that the disruption of the vaginal milieu by bacterial vaginosis prompts a decrease in protective species of the genus Lactobacillus and an increase in both strict and facultative anaerobic bacteria, namely, Gardnerella vaginalis [173‒175]. The presence of these bacteria in the vaginal mucosa is likely to promote the activation of downstream inflammatory pathways that influences mucosal susceptibility for HPV [176‒179].

One of the first associations between cancer and bacteria was reported in the 1980s [180]. These initial studies identified Helicobacter pylori as a causative agent of infectious gastritis, a chronic inflammation of the stomach [180]. This chronic inflammatory state promotes a niche that prompts tumor development. Accordingly, this infectious agent is now classified by the World Health Organization as a carcinogen class I [181]. Disseminated through the planet, it has been estimated that >50% of world population harbors these bacteria [182]. Owing to its ability to neutralize gastric acidity, through the expression of urease, H. pylori is able to survive and proliferate in the stomach and infect the gastric mucosa [183]. The expression of particular cytotoxins, mostly VacA and CagA, promotes the production of the 2 classical pro-inflammatory cytokines IL-8 and IL-6 by gastric epithelial cells [184‒187] and activates the NF-kB pathway [188, 189], thus enhancing and perpetuating the inflammatory response.

Another common member of the human microbiota that has been shown to enhance tumorigenesis through the production on an enterotoxin is Bacteroides fragilis [190, 191]. Haghi et al. [192] reported a higher prevalence of B. fragilis in CRC patients when compared to control group. Interestingly, the authors also showed an increased expression of the B. fragilis enterotoxin gene in advanced stages of the disease [192]. In line with these data, Purcell et al. [193] associated the presence of enterotoxigenic B. fragilis to the abundance of early stage carcinogenic lesions in colorectal tissue. Further, B. fragilis enterotoxin has been shown to induce persistent colitis in mice promoting chronic inflammatory status and cancer development [194]. This inflammation is mediated by increased IL-8 production and b-catenin signaling [195, 196]. Recently, B. fragilis toxin was also shown to modulate the Wnt pathway and to promote cell proliferation [197].

Also associated with increased CRC incidence is Fusobacterium nucleatum [198, 199]. The pathogenesis of these bacteria has been associated with the expression of FadA and Fap2 virulence factors [200, 201]. These virulence factors induce the expression of oncogenes and promote the growth of cancer cells [200]. A recent study identified a tumor-based immune evasion mechanism that is dependent on the interaction of Fap2 protein with the inhibitory receptor T-cell immunoglobulin and ITIM domain, expressed by all human NK cells and some T cell populations, inhibiting their function [202]. Additionally, the expression of pro-inflammatory cytokines, such as IL-8, TNF, and IL-6, associated with high levels of F. nucleatum may also contribute to establishing a microenvironment amenable for cancer progression [198], including the recruitment of myeloid-derived suppressor cells, and promoting the differentiation of M2-like macrophages and tumor-associated neutrophils which sustain the immunosuppressive environment [203, 204].

The diversity of cells that are part of the tumor as well as the heterogeneity of the mutations that result in malignant cell differentiation results in altered molecules and pathways that are prime targets for therapeutic approaches. The combination of different approaches is also used to further enhance the potential of cancer therapy [205, 206]. As discussed above, the immune system plays crucial roles in multiple carcinogenic scenarios. As such, cancer therapies that aim to boost the antitumor immune response, known as immunotherapies, have brought a new wave of enthusiasm in the fight against cancer [207, 208]. Immunotherapies aim to boost the function of specific subsets of cells, which is usually accomplished through reeducation, as it is the case for dendritic cell therapies, or modulation of the immune response, as it is the case of blockade antibodies or cytokines which target critical immune pathways [4].

The first immunotherapy approved was based on the administration of high doses of IL-2 to treat metastatic renal cell carcinoma and metastatic melanoma [209]. The potential of this therapy was associated with ability of IL-2 in prompting the activation and differentiation of naïve T cells into effector and memory T cells and by stimulation of activation-induced cell death pathways [210, 211]. In addition to IL-2, IFN-α has been also approved for adjuvant treatment of completely resected high-risk melanoma patients and several refractory malignancies [212, 213]. Despite the success of these therapies to activate the immune system of cancer patients, cytokine therapy has been difficult to implement in clinical settings. Indeed, while multiple cytokines are still under evaluation in preclinical setting and clinical trials (including granulocyte-macrophage colony-stimulating factor, IFN-γ, IL-7, IL-12, and IL-21), cytokines in monotherapy have not fulfilled the promise of efficacy seen in preclinical experiments. This is likely associated with their dose-limiting toxicities and pleotropic functions. However, as our knowledge in the role of specific cytokines increases and novel methodologies for the administration and engineering of these molecules are developed, we will certainly see a rise in the use of cytokines as cancer therapies [214].

One central mechanism whereby the TME contributes to the suppression of antitumor immunity is through the induction of immune checkpoint inhibitors that act as “brakes” of the acquired immune response. While immune checkpoint inhibitors play key roles in preventing autoimmunity and tissue damage during an immune response, their dysregulation during tumorigenesis can hamper the activation of immune cells and prevent their antitumor activity. It was in this context that the recent development and clinical use of monoclonal antibodies that block the function of the immune checkpoint inhibitors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), program-death 1 (PD-1), and programmed death-ligand 1 (PD-L1) revolutionized cancer therapy [215]. Both CTLA-4 and PD-1 are negative regulators of T-cell immune function [216, 217]. CTLA-4 is a CD28 homolog expressed by T cells that binds to B7 molecules in antigen presenting cells. Contrary to the stimulatory signal provided by the interaction CD28-B7, binding of CTLA-4 to B7 does not produce a stimulatory signal. As affinity of CTLA-4 for B7 is much higher than CD28, the CTLA-4-B7 binding prevent the costimulatory signal normally provided by CD28, thus preventing T cell activation and effector differentiation [216, 217]. Similar to CTLA-4 signaling, the binding of PD-1 with its ligands PD-L1 or PD-L2 also has an inhibitory effect on T cells, specifically in T cell proliferation and cytokine production [218, 219]. However, unlike CTLA-4 ligands expressed only by antigen-presenting cells, PD-1 ligands are more widely expressed. This pattern of expression enables PD-1 functions to impair T cell activity during the effector phase of the response in peripheral tissues, specifically tumors [216, 217].

Pioneering studies conducted by James Allison provided evidence that the inhibition of CTLA-4 through the use of monoclonal antibodies was associated with tumor regression in mice [220]. Subsequent studies also highlighted the antitumor potential of CTLA-4 blockade in adjunct therapies, with vaccines or chemotherapy [221, 222]. In clinical settings, ipilimumab, a commercialized form of CTLA-4 monoclonal antibody, has been proven efficient in the treatment of metastatic melanoma [223]. Additionally, a longitudinal follow-up study of 177 patients indicated a curative regression of metastatic melanoma in a small percent of patients [224]. The potential of anti-CTLA-4 in cancer treatment led to the approval of ipilimumab treatment for metastatic melanoma. In addition to the overall increase in the survival of patients treated with ipilimumab, other studies demonstrated a synergistic effect of ipilimumab and gp100 peptide vaccine, which is indicated for melanoma treatment [223]. However, these treatments were also associated with adverse effects. Therefore, future studies and clinical trials will be crucial to assess the relevance of CTLA-4 blockade in cancer treatment [223, 225].

In addition to CTLA-4, PD-1 and its ligand PD-L1 raised interest as immunotherapeutic targets. During tumor progression, cancer cells can increase their expression of PD-L1 that upon binding to PD-1 expressed by T cells inhibits their response [226]. Indeed, this pathway has been shown to play a key role in the escape of tumors from immunosurveillance as T cells express high levels of PD-1 upon tumor infiltration [227]. Accordingly, PD-L1 expression has been reported in several types of cancer including bladder, lung, colon, and melanoma. Importantly, the administration of anti-PD-1/PD-L1 monoclonal antibodies has been successful in controlling tumor progression and it has been approved for the treatment of several types of cancer [228‒232]. Moreover, it has been reported that PD-1 blockade in melanoma [233] and lung cancer [234] is more efficient in cases of highly mutated cells. It has been hypothesized that this enhanced efficiency is associated with the increased expression of neoantigens by tumor cells, which in combination with anti-PD-1 further enhance the response of T cells [234]. The efficiency of these therapies has led to the combination of CTLA-4 and PD1/PD-L1. While their combination showed increased efficiency, it also resulted in worse adverse effects [235‒239].

In the last decade, several other immune checkpoint receptors including TIM-3 and LAG-3 have been in the frontline to be validated as therapeutic targets, showing promising preclinical and clinical results [240‒243]. Nonetheless, not all the individuals respond to immunotherapy nor they are free from relapsing, which opens the door for the constant development of new strategies to cancer treatment. While heterogeneity of tumors and preexisting tumor immune response can dictate the success of immunotherapies, the impact of commensal microbiota has been under scrutiny in the last years.

Microbiota Modulation of Cancer Therapy

The impact of the microbiota in the tumor immune response is undeniable. Although its importance has recently been reinvigorated, older studies showed the therapeutic potential of controlled doses of bacteria, or bacterial products, to potentiate the immune response and achieve antitumor immune status [244‒246]. More recently, the microbiota has been shown to play a critical role in the efficacy of immunotherapy, with 2 recent preclinical studies reporting that alterations in microbiota composition dictated the efficiency of CTLA-4 and PD-1 blockade [8, 247]. Vétizou et al. [8] reported that anti-CTLA-4 therapy did not inhibit tumor growth in germ-free or antibiotic-treated mice, pointing to a key role of the microbiota in the efficacy of anti-CTLA4. Further experiments showed that Bacteroidales and Burkholderiales were crucial for the antitumor activity of anti-CTLA-4 therapy [8]. Indeed, oral transplantation of B. fragilis in combination with B. thetaiotaomicron or B. cepacia restored the efficiency of anti-CTLA-4 as did the fecal transplants from human samples with elevated levels of Bacteroides species [8]. Mechanistically, the microbiota promoted the generation of antitumor antigen-specific Th1 responses which controlled tumor growth both in mice and patients, likely by producing cross-reactive antigens and enhancing antitumor T cell responses [8].

The impact of microbiota is not specific to anti-CTLA4 therapy. Indeed, Sivan et al. [247] reported discrepancies in tumor growth and anti-PD-1 therapy when comparing genetically similar mice harboring distinct microbiota. Interestingly, fecal transplants from responsive to unresponsive mice restored the efficacy of PD-1 blockade, showing that microbiota composition plays a critical role in the anti-PD-1 therapy. Importantly, the microbiota of mice prone to respond to anti-PD-1 treatment was enriched in Bifidobacterium species and the supplementation with Bifidobacterium restored antitumor efficacy of PD-1 blockade in mice that did not have these bacteria [247]. As for the anti-CTLA4 data discussed above, augmented dendritic cell function and enhanced CD8+ T cell priming and accumulation in the TME was shown to be associated with the efficacy of PD-1 blockade [247]. The relevance of these data in clinical setting was demonstrated by a follow-up study, showing that the bacterial genera Akkermansia, Enterococcus, Bifidobacterium, and Faecalibacterium were associated with a response to PD-1 blockade [9, 248, 249]. While our understanding on the mechanisms whereby the microbiota, and particular species of the microbiota, modulate the antitumor immune response is in its infancy, these data described suggest that targeting the microbiota may promote the efficacy of antitumor therapies. In this regard, it has been shown that mice treated with antibiotics do not respond to cisplatin, oxaliplatin, or cyclophosphamide drug treatment [250‒252]. However, combining cisplatin with probiotics administration, Lactobacilli, improved mice response to therapy [250]. Accordingly, the oral administration of Lactobacillus johonsonii and Enterococcus hirae induced the differentiation of pro-inflammatory T helper cells which improved the efficacy of cyclophosphamide treatment in tumor-bearing mice [251].

In the light of the current knowledge, it is clear that the microbiota plays pivotal roles in cancer development, progression, and in the efficacy of classical and novel therapeutic approaches. As such, future studies should focus on the therapeutic potential of manipulating gut microbiota in cancer patients (by means of probiotic consumption, fecal microbial transplants, or design of specific diets) as adjunct approach to current or novel immunotherapeutic modalities.

Cancer is the second leading cause of death globally with an estimated death toll of 9.6 million people in 2018 alone. Recent developments in cancer therapy, including checkpoint blockade therapies, has brought significant improvements for patients in terms of survival and quality of life. Unfortunately, not all patients benefit for these novel approaches. As such, defining the factor that modulates cancer immune responses is key to improve current and novel therapeutic approaches. Genetics and functional studies have now demonstrated the dual role of the gut microbiome in cancer. While in some cases bacteria can overgrow during dysbiosis, triggering an inflammatory pro-cancerogenic environment, other bacteria and bacterial-derived metabolites protect the host. Adding to this knowledge is the recently identified gut microbiota species that contribute to improve the efficacy of immunotherapy in cancer immunotherapy responders. Together, these data highlight the potential of microbiota modulation as adjunct to current and novel immunotherapies. It is therefore critical to continue defining the therapeutic benefits derived from modulating the microbiota to further explore their potential as we move into the age of personalized medicine.

The authors have no conflicts of interest to disclose.

Our work is funded by national funds through the Foundation for Science and Technology (FCT) – projects PTDC/MED-ONC/28658/2017, UIDB/50026/2020, and UIDP/50026/2020 and by the projects NORTE-01-0145-FEDER-000013, and NORTE-01-0145-FEDER-000023, supported by Norte Portugal Regional Operational Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF). ET was supported by the FCT investigator grant IF/01390/2014 and AMB through the FCT PhD fellowship SFRH/BD/120371/2016.

1.
Ostrand-Rosenberg
S
.
Immune surveillance: a balance between protumor and antitumor immunity
.
Curr Opin Genet Dev
.
2008 Feb
;
18
(
1
):
11
8
.http://dx.doi.org/10.1016/j.gde.2007.12.007.
2.
Goswami
KK
,
Ghosh
T
,
Ghosh
S
,
Sarkar
M
,
Bose
A
,
Baral
R
.
Tumor promoting role of anti-tumor macrophages in tumor microenvironment
.
Cell Immunol
.
2017 Jun
;
316
:
1
10
.
3.
Munhoz
RR
,
Postow
MA
.
Recent advances in understanding antitumor immunity
.
F1000Res
.
2016 Oct
;
5
:
2545
.
4.
Esfahani
K
,
Roudaia
L
,
Buhlaiga
N
,
Del Rincon
SV
,
Papneja
N
,
Miller
WH
Jr
.
A review of cancer immunotherapy: from the past, to the present, to the future
.
Curr Oncol
.
2019 Mar
;
27
(
S2
):
87
97
.
5.
Herrscher
H
,
Robert
C
.
Immune checkpoint inhibitors in melanoma in the metastatic, neoadjuvant, and adjuvant setting
.
Curr Opin Oncol
.
2020 Mar
;
32
(
2
):
106
13
.
6.
Wei
SC
,
Anang
NAS
,
Sharma
R
,
Andrews
MC
,
Reuben
A
,
Levine
JH
,
.
Combination anti-CTLA-4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies
.
Proc Natl Acad Sci U S A
.
2019 Nov
;
116
(
45
):
22699
709
.
7.
Thursby
E
,
Juge
N
.
Introduction to the human gut microbiota
.
Biochem J
.
2017 Jun
;
474
(
11
):
1823
36
.
8.
Vétizou
M
,
Pitt
JM
,
Daillère
R
,
Lepage
P
,
Waldschmitt
N
,
Flament
C
,
.
Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota
.
Science
.
2015 Nov
;
350
(
6264
):
1079
84
.
9.
Routy
B
,
Le Chatelier
E
,
Derosa
L
,
Duong
CPM
,
Alou
MT
,
Daillère
R
,
.
Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors
.
Science
.
2018 Jan
;
359
(
6371
):
91
7
.
10.
Goldszmid
RS
,
Dzutsev
A
,
Viaud
S
,
Zitvogel
L
,
Restifo
NP
,
Trinchieri
G
.
Microbiota modulation of myeloid cells in cancer therapy
.
Cancer Immunol Res
.
2015 Feb
;
3
(
2
):
103
9
.
11.
Roy
S
,
Trinchieri
G
.
Microbiota: a key orchestrator of cancer therapy
.
Nat Rev Cancer
.
2017 May
;
17
(
5
):
271
85
.
12.
Bertram
JS
.
The molecular biology of cancer
.
Mol Aspects Med
.
2000 Dec
;
21
(
6
):
167
223
.
13.
Hanahan
D
,
Weinberg
RA
.
Hallmarks of cancer: the next generation
.
Cell
.
2011 Mar
;
144
(
5
):
646
74
.
14.
Adams
JM
,
Cory
S
.
The Bcl-2 apoptotic switch in cancer development and therapy
.
Oncogene
.
2007 Feb
;
26
(
9
):
1324
37
.
15.
Lowe
SW
,
Cepero
E
,
Evan
G
.
Intrinsic tumour suppression
.
Nature
.
2004 Nov
;
432
(
7015
):
307
15
.
16.
Evan
G
,
Littlewood
T
.
A matter of life and cell death
.
Science
.
1998 Aug
;
281
(
5381
):
1317
22
.
17.
Finn
OJ
.
A believer’s overview of cancer immunosurveillance and immunotherapy
.
J Immunol
.
2018 Jan
;
200
(
2
):
385
91
.
18.
Burnet
M
.
Cancer-A biological approach I. The processes of control
.
Br Med J
.
1957 Apr
;
1
(
5022
):
779
86
.
19.
Burnet
FM
.
The concept of immunological surveillance
.
Prog Exp Tumor Res
.
1970
;
13
:
1
27
.
20.
Dunn
GP
,
Bruce
AT
,
Ikeda
H
,
Old
LJ
,
Schreiber
RD
.
Cancer immunoediting: from immunosurveillance to tumor escape
.
Nat Immunol
.
2002 Nov
;
3
(
11
):
991
8
.
21.
Swann
JB
,
Smyth
MJ
.
Immune surveillance of tumors
.
J Clin Invest
.
2007 May
;
117
(
5
):
1137
46
.
22.
Vigneron
N
,
Van den Eynde
BJ
.
Insights into the processing of MHC class I ligands gained from the study of human tumor epitopes
.
Cell Mol Life Sci
.
2011 May
;
68
(
9
):
1503
20
.
23.
Kaplan
DH
,
Shankaran
V
,
Dighe
AS
,
Stockert
E
,
Aguet
M
,
Old
LJ
,
.
Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice
.
Proc Natl Acad Sci U S A
.
1998 Jun
;
95
(
13
):
7556
61
.
24.
Street
SE
,
Cretney
E
,
Smyth
MJ
.
Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis
.
Blood
.
2001 Jan
;
97
(
1
):
192
7
.
25.
Hadden
JW
.
Immunodeficiency and cancer: prospects for correction
.
Int Immunopharmacol
.
2003 Aug
;
3
(
8
):
1061
71
.
26.
van den Broek
ME
,
Kägi
D
,
Ossendorp
F
,
Toes
R
,
Vamvakas
S
,
Lutz
WK
,
.
Decreased tumor surveillance in perforin-deficient mice
.
J Exp Med
.
1996 Nov
;
184
(
5
):
1781
90
.
27.
Dighe
AS
,
Richards
E
,
Old
LJ
,
Schreiber
RD
.
Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors
.
Immunity
.
1994 Sep
;
1
(
6
):
447
56
.
28.
Topalian
SL
,
Solomon
D
,
Rosenberg
SA
.
Tumor-specific cytolysis by lymphocytes infiltrating human melanomas
.
J Immunol
.
1989 May
;
142
(
10
):
3714
25
..
29.
Disis
ML
,
Calenoff
E
,
McLaughlin
G
,
Murphy
AE
,
Chen
W
,
Groner
B
,
.
Existent T-cell and antibody immunity to HER-2/neu protein in patients with breast cancer
.
Cancer Res
.
1994 Jan
;
54
(
1
):
16
20
.
30.
Beckhove
P
,
Feuerer
M
,
Dolenc
M
,
Schuetz
F
,
Choi
C
,
Sommerfeldt
N
,
.
Specifically activated memory T cell subsets from cancer patients recognize and reject xenotransplanted autologous tumors
.
J Clin Invest
.
2004 Jul
;
114
(
1
):
67
76
.
31.
Clemente
CG
,
Mihm
MC
,
Bufalino
R
,
Zurrida
S
,
Collini
P
,
Cascinelli
N
.
Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma
.
Cancer
.
1996
;
77
(
7
):
1303
10
.
32.
Galon
J
,
Costes
A
,
Sanchez-Cabo
F
,
Kirilovsky
A
,
Mlecnik
B
,
Lagorce-Pagès
C
,
.
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome
.
Science
.
2006
;
313
(
5795
):
1960
4
.
33.
Valpione
S
,
Galvani
E
,
Tweedy
J
,
Mundra
PA
,
Banyard
A
,
Middlehurst
P
,
.
Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy
.
Nat Cancer
.
2020 Feb
;
1
(
2
):
210
21
.
34.
Wu
TD
,
Madireddi
S
,
de Almeida
PE
,
Banchereau
R
,
Chen
YJ
,
Chitre
AS
,
.
Peripheral T cell expansion predicts tumour infiltration and clinical response
.
Nature
.
2020 Mar
;
579
(
7798
):
274
8
.
35.
Taniguchi
K
,
Petersson
M
,
Höglund
P
,
Kiessling
R
,
Klein
G
,
Kärre
K
.
Interferon gamma induces lung colonization by intravenously inoculated B16 melanoma cells in parallel with enhanced expression of class I major histocompatibility complex antigens
.
Proc Natl Acad Sci U S A
.
1987 May
;
84
(
10
):
3405
9
.
36.
Garbe
C
,
Krasagakis
K
,
Zouboulis
CC
,
Schröder
K
,
Krüger
S
,
Stadler
R
,
.
Antitumor activities of interferon alpha, beta, and gamma and their combinations on human melanoma cells in vitro: changes of proliferation, melanin synthesis, and immunophenotype
.
J Invest Dermatol
.
1990 Dec
;
95
(
6 Suppl
):
231S
237S
.
37.
Schlecker
E
,
Stojanovic
A
,
Eisen
C
,
Quack
C
,
Falk
CS
,
Umansky
V
,
.
Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth
.
J Immunol
.
2012 Dec
;
189
(
12
):
5602
11
.
38.
Curiel
TJ
,
Coukos
G
,
Zou
L
,
Alvarez
X
,
Cheng
P
,
Mottram
P
,
.
Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival
.
Nat Med
.
2004 Sep
;
10
(
9
):
942
9
.
39.
Xu
L
,
Xu
W
,
Qiu
S
,
Xiong
S
.
Enrichment of CCR6+Foxp3+ regulatory T cells in the tumor mass correlates with impaired CD8+ T cell function and poor prognosis of breast cancer
.
Clin Immunol
.
2010 Jun
;
135
(
3
):
466
75
.
40.
Klingen
TA
,
Chen
Y
,
Aas
H
,
Wik
E
,
Akslen
LA
.
Tumor associated macrophages are strongly related to vascular invasion, non-luminal subtypes and interval breast cancer
.
Hum Pathol
.
2017 Nov
;
69
:
72
80
.
41.
Balkwill
FR
,
Capasso
M
,
Hagemann
T
.
The tumor microenvironment at a glance
.
J Cell Sci
.
2012 Dec
;
125
(
Pt 23
):
5591
6
.
42.
Yang
L
,
Zhang
Y
.
Tumor-associated macrophages: from basic research to clinical application
.
J Hematol Oncol
.
2017 Dec
;
10
(
1
):
58
.
43.
Mantovani
A
,
Sozzani
S
,
Locati
M
,
Allavena
P
,
Sica
A
.
Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes
.
Trends Immunol
.
2002 Nov
;
23
(
11
):
549
55
.
44.
Cassetta
L
,
Cassol
E
,
Poli
G
.
Macrophage polarization in health and disease
.
ScientificWorldJournal
.
2011
;
11
:
2391
402
.
45.
Najafi
M
,
Hashemi Goradel
N
,
Farhood
B
,
Salehi
E
,
Nashtaei
MS
,
Khanlarkhani
N
,
.
Macrophage polarity in cancer: a review
.
J Cell Biochem
.
2019 Mar
;
120
(
3
):
2756
65
.
46.
Zhang
W
,
Chen
L
,
Ma
K
,
Zhao
Y
,
Liu
X
,
Wang
Y
,
.
Polarization of macrophages in the tumor microenvironment is influenced by EGFR signaling within colon cancer cells
.
Oncotarget
.
2016 Nov
;
7
(
46
):
75366
78
.
47.
Hao
NB
,
MH
,
Fan
YH
,
Cao
YL
,
Zhang
ZR
,
Yang
SM
.
Macrophages in tumor microenvironments and the progression of tumors
.
Clin Dev Immunol
.
2012
;
2012
:
948098
11
.
48.
Zhang
Y
,
Choksi
S
,
Chen
K
,
Pobezinskaya
Y
,
Linnoila
I
,
Liu
ZG
.
ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of tumor-associated macrophages
.
Cell Res
.
2013 Jul
;
23
(
7
):
898
914
.
49.
Sica
A
,
Mantovani
A
.
Macrophage plasticity and polarization: in vivo veritas
.
J Clin Invest
.
2012 Mar
;
122
(
3
):
787
95
.
50.
Fridlender
ZG
,
Albelda
SM
.
Tumor-associated neutrophils: friend or foe
.
Carcinogenesis
.
2012 May
;
33
(
5
):
949
55
.
51.
Fridlender
ZG
,
Sun
J
,
Kim
S
,
Kapoor
V
,
Cheng
G
,
Ling
L
,
.
Polarization of tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN
.
Cancer Cell
.
2009 Sep
;
16
(
3
):
183
94
.
52.
Dissemond
J
,
Weimann
TK
,
Schneider
LA
,
Schneeberger
A
,
Scharffetter-Kochanek
K
,
Goos
M
,
.
Activated neutrophils exert antitumor activity against human melanoma cells: reactive oxygen species-induced mechanisms and their modulation by granulocyte-macrophage–colony-stimulating factor
.
J Invest Dermatol
.
2003 Oct
;
121
(
4
):
936
8
.
53.
Zivkovic
M
,
Poljak-Blazi
M
,
Egger
G
,
Sunjic
SB
,
Schaur
RJ
,
Zarkovic
N
.
Oxidative burst and anticancer activities of rat neutrophils
.
BioFactors
.
2005
;
24
(
1–4
):
305
12
.
54.
Granot
Z
,
Henke
E
,
Comen
EA
,
King
TA
,
Norton
L
,
Benezra
R
.
Tumor entrained neutrophils inhibit seeding in the premetastatic lung
.
Cancer Cell
.
2011 Sep
;
20
(
3
):
300
14
.
55.
Governa
V
,
Trella
E
,
Mele
V
,
Tornillo
L
,
Amicarella
F
,
Cremonesi
E
,
.
The interplay between neutrophils and CD8
.
Clin Cancer Res
.
2017 Jul
;
23
(
14
):
3847
58
.
56.
Eruslanov
EB
,
Bhojnagarwala
PS
,
Quatromoni
JG
,
Stephen
TL
,
Ranganathan
A
,
Deshpande
C
,
.
Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer
.
J Clin Invest
.
2014 Dec
;
124
(
12
):
5466
80
.
57.
Wikberg
ML
,
Ling
A
,
Li
X
,
Öberg Å
ES
,
Palmqvist
R
.
Neutrophil infiltration is a favorable prognostic factor in early stages of colon cancer
.
Hum Pathol
.
2017 Oct
;
68
:
193
202
.
58.
Berry
RS
,
Xiong
M-J
,
Greenbaum
A
,
Mortaji
P
,
Nofchissey
RA
,
Schultz
F
,
.
High levels of tumor-associated neutrophils are associated with improved overall survival in patients with stage II colorectal cancer
.
PLoS One
.
2017 Dec
;
12
(
12
):
e0188799
.
59.
Németh
T
,
Sperandio
M
,
Mócsai
A
.
Neutrophils as emerging therapeutic targets
.
Nat Rev Drug Discov
.
2020 Apr
;
19
(
4
):
253
75
.
60.
Zhang
Y
,
Lee
C
,
Geng
S
,
Li
L
.
Enhanced tumor immune surveillance through neutrophil reprogramming due to Tollip deficiency
.
JCI Insight
.
2019 Jan
;
4
(
2
):
e122939
.
61.
Shin
MH
,
Kim
J
,
Lim
SA
,
Kim
J
,
Kim
S-J
,
Lee
K-M
.
NK cell-based immunotherapies in cancer
.
Immune Netw
.
2020 Apr
;
20
(
2
):
e14
.
62.
Topham
NJ
,
Hewitt
EW
.
Natural killer cell cytotoxicity: how do they pull the trigger
.
Immunology
.
2009 Sep
;
128
(
1
):
7
15
.
63.
Smyth
MJ
,
Thia
KYT
,
Street
SEA
,
MacGregor
D
,
Godfrey
DI
,
Trapani
JA
.
Perforin-mediated cytotoxicity is critical for surveillance of spontaneous lymphoma
.
J Exp Med
.
2000 Sep
;
192
(
5
):
755
60
.
64.
Guerra
N
,
Tan
YX
,
Joncker
NT
,
Choy
A
,
Gallardo
F
,
Xiong
N
,
.
NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy
.
Immunity
.
2008 Apr
;
28
(
4
):
571
80
.
65.
Lorenzi
L
,
Tabellini
G
,
Vermi
W
,
Moratto
D
,
Porta
F
,
Notarangelo
LD
,
.
Occurrence of nodular lymphocyte-predominant hodgkin lymphoma in hermansky-pudlak type 2 syndrome is associated to natural killer and natural killer T cell defects
.
PLoS One
.
2013 Nov
;
8
(
11
):
e80131
.
66.
Imai
K
,
Matsuyama
S
,
Miyake
S
,
Suga
K
,
Nakachi
K
.
Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population
.
Lancet
.
2000 Nov
;
356
(
9244
):
1795
9
.
67.
Spinner
MA
,
Sanchez
LA
,
Hsu
AP
,
Shaw
PA
,
Zerbe
CS
,
Calvo
KR
,
.
GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity
.
Blood
.
2014 Feb
;
123
(
6
):
809
21
.
68.
Gwalani
LA
,
Orange
JS
.
Single degranulations in NK cells can mediate target cell killing
.
J Immunol
.
2018 May
;
200
(
9
):
3231
43
.
69.
Carotta
S
.
Targeting NK cells for anticancer immunotherapy: clinical and preclinical approaches
.
Front Immunol
.
2016 Apr
;
7
:
152
.
70.
Li
X-Y
,
Li
Z
,
An
G-J
,
Liu
S
,
Lai
Y-D
.
Co-expression of perforin and granzyme B genes induces apoptosis and inhibits the tumorigenicity of laryngeal cancer cell line Hep-2
.
Int J Clin Exp Pathol
.
2014
[cited 2020 Sep 11];
7
(
3
):
978
86
.
71.
Barry
KC
,
Hsu
J
,
Broz
ML
,
Cueto
FJ
,
Binnewies
M
,
Combes
AJ
,
.
A natural killer–dendritic cell axis defines checkpoint therapy–responsive tumor microenvironments
.
Nat Med
.
2018 Aug
;
24
(
8
):
1178
91
.
72.
Böttcher
JP
,
Bonavita
E
,
Chakravarty
P
,
Blees
H
,
Cabeza-Cabrerizo
M
,
Sammicheli
S
,
.
NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control
.
Cell
.
2018 Feb
;
172
(
5
):
1022
37.e14
.
73.
Morvan
MG
,
Lanier
LL
.
NK cells and cancer: you can teach innate cells new tricks
.
Nat Rev Cancer
.
2016 Jan
;
16
(
1
):
7
19
.
74.
Fridman
WH
,
Pagès
F
,
Sautès-Fridman
C
,
Galon
J
.
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
.
2012 Apr
;
12
(
4
):
298
306
.
75.
Lorvik
KB
,
Hammarström
C
,
Fauskanger
M
,
Haabeth
OAW
,
Zangani
M
,
Haraldsen
G
,
.
Adoptive transfer of tumor-specific Th2 cells eradicates tumors by triggering an in situ inflammatory immune response
.
Cancer Res
.
2016 Dec
;
76
(
23
):
6864
76
.
76.
Gatault
S
,
Delbeke
M
,
Driss
V
,
Sarazin
A
,
Dendooven
A
,
Kahn
J-E
,
.
IL-18 is involved in eosinophil-mediated tumoricidal activity against a colon carcinoma cell line by upregulating LFA-1 and ICAM-1
.
J Immunol
.
2015 Sep
;
195
(
5
):
2483
92
.
77.
Legrand
F
,
Driss
V
,
Delbeke
M
,
Loiseau
S
,
Hermann
E
,
Dombrowicz
D
,
.
Human eosinophils exert TNF-α and granzyme A-mediated tumoricidal activity toward colon carcinoma cells
.
J Immunol
.
2010 Dec
;
185
(
12
):
7443
51
.
78.
Nair
S
,
Dhodapkar
MV
.
Natural Killer T cells in cancer immunotherapy
.
Front Immunol
.
2017 Sep
;
8
:
1178
.
79.
Pandya
PH
,
Murray
ME
,
Pollok
KE
,
Renbarger
JL
.
The immune system in cancer pathogenesis: potential therapeutic approaches
.
J Immunol Res
.
2016
;
2016
:
1
13
.
80.
Ha
T-Y
.
The role of regulatory T cells in cancer
.
Immune Netw
.
2009
;
9
(
6
):
209
.
81.
Wculek
SK
,
Cueto
FJ
,
Mujal
AM
,
Melero
I
,
Krummel
MF
,
Sancho
D
.
Dendritic cells in cancer immunology and immunotherapy
.
Nat Rev Immunol
.
2020 Jan
;
20
(
1
):
7
24
.
82.
Phan
GQ
,
Yang
JC
,
Sherry
RM
,
Hwu
P
,
Topalian
SL
,
Schwartzentruber
DJ
,
.
Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma
.
Proc Natl Acad Sci U S A
.
2003 Jul
;
100
(
14
):
8372
7
.
83.
Hodi
FS
,
Mihm
MC
,
Soiffer
RJ
,
Haluska
FG
,
Butler
M
,
Seiden
MV
,
.
Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients
.
Proc Natl Acad Sci U S A
.
2003 Apr
;
100
(
8
):
4712
7
.
84.
Ramagopal
UA
,
Liu
W
,
Garrett-Thomson
SC
,
Bonanno
JB
,
Yan
Q
,
Srinivasan
M
,
.
Structural basis for cancer immunotherapy by the first-in-class checkpoint inhibitor ipilimumab
.
Proc Natl Acad Sci U S A
.
2017 May
;
114
(
21
):
E4223
32
.
85.
Kim
J
,
Bae
J-S
.
Tumor-associated macrophages and neutrophils in tumor microenvironment
.
Mediators Inflamm
.
2016
;
2016
(
4
):
1
11
.
86.
Zhao
X
,
Qu
J
,
Sun
Y
,
Wang
J
,
Liu
X
,
Wang
F
,
.
Prognostic significance of tumor-associated macrophages in breast cancer: a meta-analysis of the literature
.
Oncotarget
.
2017 May
;
8
(
18
):
30576
86
.
87.
Ding
W
,
Tan
Y
,
Qian
Y
,
Xue
W
,
Wang
Y
,
Jiang
P
,
.
Clinicopathologic and prognostic significance of tumor-associated macrophages in patients with hepatocellular carcinoma: a meta-analysis
.
PLoS One
.
2019 Oct
;
14
(
10
):
e0223971
.
88.
Elgert
KD
,
Alleva
DG
,
Mullins
DW
.
Tumor-induced immune dysfunction: the macrophage connection
.
J Leukoc Biol
.
1998 Sep
;
64
(
3
):
275
90
.
89.
Trinchieri
G
.
Interleukin-10 production by effector T cells: Th1 cells show self control
.
J Exp Med
.
2007 Feb
;
204
(
2
):
239
43
.
90.
Jung
Y-J
,
Isaacs
JS
,
Lee
S
,
Trepel
J
,
Neckers
L
.
IL-1β mediated up-regulation of HIF-lα via an NFkB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis
.
FASEB J
.
2003 Nov
;
17
(
14
):
1
22
.
91.
Lai
Y
,
Wahyuningtyas
R
,
Aui
S
,
Chang
K
.
Autocrine VEGF signalling on M2 macrophages regulates PD-L1 expression for immunomodulation of T cells
.
J Cell Mol Med
.
2019 Feb
;
23
(
2
):
1257
67
.
92.
Grimshaw
MJ
,
Wilson
JL
,
Balkwill
FR
.
Endothelin-2 is a macrophage chemoattractant: implications for macrophage distribution in tumors
.
Eur J Immunol
.
2002 Sep
;
32
(
9
):
2393
400
.
93.
Pollard
JW
.
Tumour-educated macrophages promote tumour progression and metastasis
.
Nat Rev Cancer
.
2004 Jan
;
4
(
1
):
71
8
.
94.
Giraudo
E
,
Inoue
M
,
Hanahan
D
.
An amino-bisphosphonate targets MMP-9–expressing macrophages and angiogenesis to impair cervical carcinogenesis
.
J Clin Invest
.
2004 Sep
;
114
(
5
):
623
33
.
95.
Vérollet
C
,
Charrière
GM
,
Labrousse
A
,
Cougoule
C
,
Le Cabec
V
,
Maridonneau-Parini
I
.
Extracellular proteolysis in macrophage migration: losing grip for a breakthrough
.
Eur J Immunol
.
2011 Oct
;
41
(
10
):
2805
13
.
96.
Kusmartsev
S
,
Gabrilovich
DI
.
STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion
.
J Immunol
.
2005 Apr
;
174
(
8
):
4880
91
.
97.
Zhou
S-L
,
Zhou
Z-J
,
Hu
Z-Q
,
Huang
X-W
,
Wang
Z
,
Chen
E-B
,
.
Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib
.
Gastroenterology
.
2016 Jun
;
150
(
7
):
1646
58.e17
.
98.
Mishalian
I
,
Bayuh
R
,
Eruslanov
E
,
Michaeli
J
,
Levy
L
,
Zolotarov
L
,
.
Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17-A new mechanism of impaired antitumor immunity
.
Int J Cancer
.
2014 Sep
;
135
(
5
):
1178
86
.
99.
Shaul
ME
,
Levy
L
,
Sun
J
,
Mishalian
I
,
Singhal
S
,
Kapoor
V
,
.
Tumor-associated neutrophils display a distinct N1 profile following TGFβ modulation: a transcriptomics analysis of pro- vs. antitumor TANs
.
Oncoimmunology
.
2016 Nov
;
5
(
11
):
e1232221
.
100.
Schmidt
H
,
Bastholt
L
,
Geertsen
P
,
Christensen
IJ
,
Larsen
S
,
Gehl
J
,
.
Elevated neutrophil and monocyte counts in peripheral blood are associated with poor survival in patients with metastatic melanoma: a prognostic model
.
Br J Cancer
.
2005 Aug
;
93
(
3
):
273
8
.
101.
Atzpodien
J
,
Reitz
M
.
Peripheral blood neutrophils as independent immunologic predictor of response and long-term survival upon immunotherapy in metastatic renal-cell carcinoma
.
Cancer Biother Radiopharm
.
2008 Feb
;
23
(
1
):
129
34
.
102.
Nozawa
H
,
Chiu
C
,
Hanahan
D
.
Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis
.
Proc Natl Acad Sci U S A
.
2006 Aug
;
103
(
33
):
12493
8
.
103.
Hiramatsu
S
,
Tanaka
H
,
Nishimura
J
,
Sakimura
C
,
Tamura
T
,
Toyokawa
T
,
.
Neutrophils in primary gastric tumors are correlated with neutrophil infiltration in tumor-draining lymph nodes and the systemic inflammatory response
.
BMC Immunol
.
2018 Dec
;
19
(
1
):
13
.
104.
Wculek
SK
,
Malanchi
I
.
Neutrophils support lung colonization of metastasis-initiating breast cancer cells
.
Nature
.
2015 Dec
;
528
(
7582
):
413
7
.
105.
Jablonska
J
,
Leschner
S
,
Westphal
K
,
Lienenklaus
S
,
Weiss
S
.
Neutrophils responsive to endogenous IFN-β regulate tumor angiogenesis and growth in a mouse tumor model
.
J Clin Invest
.
2010 Apr
;
120
(
4
):
1151
64
.
106.
Diaz-Montero
CM
,
Salem
ML
,
Nishimura
MI
,
Garrett-Mayer
E
,
Cole
DJ
,
Montero
AJ
.
Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin–cyclophosphamide chemotherapy
.
Cancer Immunol Immunother
.
2009 Jan
;
58
(
1
):
49
59
.
107.
Kumar
V
,
Patel
S
,
Tcyganov
E
,
Gabrilovich
DI
.
The nature of myeloid-derived suppressor cells in the tumor microenvironment
.
Trends Immunol
.
2016 Mar
;
37
(
3
):
208
20
.
108.
Dar
AA
,
Patil
RS
,
Pradhan
TN
,
Chaukar
DA
,
D’Cruz
AK
,
Chiplunkar
SV
.
Myeloid-derived suppressor cells impede T cell functionality and promote Th17 differentiation in oral squamous cell carcinoma
.
Cancer Immunol Immunother
.
2020 Jun
;
69
(
6
):
1071
86
.
109.
Bronte
V
,
Serafini
P
,
Mazzoni
A
,
Segal
DM
,
Zanovello
P
.
L-arginine metabolism in myeloid cells controls T-lymphocyte functions
.
Trends Immunol
.
2003 Jun
;
24
(
6
):
301
5
.
110.
Huang
B
,
Pan
PY
,
Li
Q
,
Sato
AI
,
Levy
DE
,
Bromberg
J
,
.
Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host
.
Cancer Res
.
2006
;
66
(
2
):
1123
31
.
111.
Sinha
P
,
Clements
VK
,
Bunt
SK
,
Albelda
SM
,
Ostrand-Rosenberg
S
.
Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response
.
J Immunol
.
2007 Jul
;
179
(
2
):
977
83
.
112.
Corzo
CA
,
Condamine
T
,
Lu
L
,
Cotter
MJ
,
Youn
JI
,
Cheng
P
,
.
HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment
.
J Exp Med
.
2010
;
207
(
11
):
2439
53
.
113.
Condamine
T
,
Ramachandran
I
,
Youn
J-I
,
Gabrilovich
DI
.
Regulation of tumor metastasis by myeloid-derived suppressor cells
.
Annu Rev Med
.
2015 Jan
;
66
(
1
):
97
110
.
114.
McNally
A
,
Hill
GR
,
Sparwasser
T
,
Thomas
R
,
Steptoe
RJ
.
CD4+CD25+ regulatory T cells control CD8+ T-cell effector differentiation by modulating IL-2 homeostasis
.
Proc Natl Acad Sci U S A
.
2011 May
;
108
(
18
):
7529
34
.
115.
Chen
M-L
,
Pittet
MJ
,
Gorelik
L
,
Flavell
RA
,
Weissleder
R
,
von Boehmer
H
,
.
Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF- signals in vivo
.
Proc Natl Acad Sci U S A
.
2005 Jan
;
102
(
2
):
419
24
.
116.
Zhang
A
,
Qian
Y
,
Zheng
S
.
Prognostic significance of regulatory T lymphocytes in patients with hepatocellular carcinoma
.
J Zhejiang Univ B
.
2016 Dec
;
17
(
12
):
984
91
.
117.
Tu
J-F
,
Ding
Y-H
,
Ying
X-H
,
Wu
F-Z
,
Zhou
X-M
,
Zhang
D-K
,
.
Regulatory T cells, especially ICOS+ FOXP3+ regulatory T cells, are increased in the hepatocellular carcinoma microenvironment and predict reduced survival
.
Sci Rep
.
2016 Dec
;
6
(
1
):
35056
.
118.
Mocellin
S
,
Marincola
FM
,
Young
HA
.
Interleukin-10 and the immune response against cancer: a counterpoint
.
J Leukoc Biol
.
2005 Nov
;
78
(
5
):
1043
51
.
119.
Salazar-Onfray
F
,
López
MN
,
Mendoza-Naranjo
A
.
Paradoxical effects of cytokines in tumor immune surveillance and tumor immune escape
.
Cytokine Growth Factor Rev
.
2007 Feb
;
18
(
1–2
):
171
82
.
120.
Sharma
S
,
Stolina
M
,
Lin
Y
,
Gardner
B
,
Miller
PW
,
Kronenberg
M
,
.
T cell-derived IL-10 promotes lung cancer growth by suppressing both T cell and APC function
.
J Immunol
.
1999 Nov
;
163
(
9
):
5020
8
.
121.
Rodriguez
PC
,
Quiceno
DG
,
Zabaleta
J
,
Ortiz
B
,
Zea
AH
,
Piazuelo
MB
,
.
Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses
.
Cancer Res
.
2004 Aug
;
64
(
16
):
5839
49
.
122.
Virchow
R
.
Cellular pathology. As based upon physiological and pathological histology. Lecture XVI: atheromatous affection of arteries. 1858
.
Nutr Rev
.
1989 Jan
;
47
(
1
):
23
5
.
123.
Balkwill
F
,
Mantovani
A
.
Inflammation and cancer: back to Virchow
.
Lancet
.
2001 Feb
;
357
(
9255
):
539
45
.
124.
Philip
M
,
Rowley
DA
,
Schreiber
H
.
Inflammation as a tumor promoter in cancer induction
.
Semin Cancer Biol
.
2004 Dec
;
14
(
6
):
433
9
.
125.
Sieweke
M
,
Thompson
N
,
Sporn
M
,
Bissell
M
.
Mediation of wound-related Rous sarcoma virus tumorigenesis by TGF-beta
.
Science
.
1990 Jun
;
248
(
4963
):
1656
60
.
126.
Hussain
SP
,
Hofseth
LJ
,
Harris
CC
.
Radical causes of cancer
.
Nat Rev Cancer
.
2003 Apr
;
3
(
4
):
276
85
.
127.
De Flora
S
,
Bonanni
P
.
The prevention of infection-associated cancers
.
Carcinogenesis
.
2011 Jun
;
32
(
6
):
787
95
.
128.
de Martel
C
,
Ferlay
J
,
Franceschi
S
,
Vignat
J
,
Bray
F
,
Forman
D
,
.
Global burden of cancers attributable to infections in 2008: a review and synthetic analysis
.
Lancet Oncol
.
2012 Jun
;
13
(
6
):
607
15
.
129.
Milani
C
,
Duranti
S
,
Bottacini
F
,
Casey
E
,
Turroni
F
,
Mahony
J
,
.
The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota
.
Microbiol Mol Biol Rev
.
2017 Dec
;
81
(
4
):
e00036
17
.
130.
Macpherson
AJ
,
Harris
NL
.
Interactions between commensal intestinal bacteria and the immune system
.
Nat Rev Immunol
.
2004 Jun
;
4
(
6
):
478
85
.
131.
Belkaid
Y
,
Hand
TW
.
Role of the microbiota in immunity and inflammation
.
Cell
.
2014 Mar
;
157
(
1
):
121
41
.
132.
Negi
S
,
Das
DK
,
Pahari
S
,
Nadeem
S
,
Agrewala
JN
.
Potential role of gut microbiota in induction and regulation of innate immune memory
.
Front Immunol
.
2019 Oct
;
10
:
2441
.
133.
Janda
JM
,
Abbott
SL
.
16S rRNA gene sequencing for bacterial identification in the diagnostic laboratory: pluses, perils, and pitfalls
.
J Clin Microbiol
.
2007 Sep
;
45
(
9
):
2761
4
.
134.
Rinninella
E
,
Raoul
P
,
Cintoni
M
,
Franceschi
F
,
Miggiano
G
,
Gasbarrini
A
,
.
What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases
.
Microorganisms
.
2019 Jan
;
7
(
1
):
14
.
135.
Kolodziejczyk
AA
,
Zheng
D
,
Elinav
E
.
Diet–microbiota interactions and personalized nutrition
.
Nat Rev Microbiol
.
2019 Dec
;
17
(
12
):
742
53
.
136.
Bibbò
S
,
Ianiro
G
,
Giorgio
V
,
Scaldaferri
F
,
Masucci
L
,
Gasbarrini
A
,
.
The role of diet on gut microbiota composition
.
Eur Rev Med Pharmacol Sci
.
2016
;
20
(
22
):
4742
9
.
137.
Ianiro
G
,
Tilg
H
,
Gasbarrini
A
.
Antibiotics as deep modulators of gut microbiota: between good and evil
.
Gut
.
2016 Nov
;
65
(
11
):
1906
15
.
138.
Zimmermann
P
,
Curtis
N
.
The effect of antibiotics on the composition of the intestinal microbiota: a systematic review
.
J Infect
.
2019 Dec
;
79
(
6
):
471
89
.
139.
Hasan
N
,
Yang
H
.
Factors affecting the composition of the gut microbiota, and its modulation
.
PeerJ
.
2019 Aug
;
7
(
8
):
e7502
.
140.
Dzutsev
A
,
Goldszmid
RS
,
Viaud
S
,
Zitvogel
L
,
Trinchieri
G
.
The role of the microbiota in inflammation, carcinogenesis, and cancer therapy
.
Eur J Immunol
.
2015 Jan
;
45
(
1
):
17
31
.
141.
Danneskiold-Samsøe
NB
,
Dias de Freitas Queiroz Barros
H
,
Santos
R
,
Bicas
JL
,
Cazarin
CBB
,
Madsen
L
,
.
Interplay between food and gut microbiota in health and disease
.
Food Res Int
.
2019 Jan
;
115
:
23
31
.
142.
Samraj
AN
,
Pearce
OMT
,
Läubli
H
,
Crittenden
AN
,
Bergfeld
AK
,
Banda
K
,
.
A red meat-derived glycan promotes inflammation and cancer progression
.
Proc Natl Acad Sci U S A
.
2015 Jan
;
112
(
2
):
542
7
.
143.
Conlon
M
,
Bird
A
.
The impact of diet and lifestyle on gut microbiota and human health
.
Nutrients
.
2014 Dec
;
7
(
1
):
17
44
.
144.
Wu
X
,
Wu
Y
,
He
L
,
Wu
L
,
Wang
X
,
Liu
Z
.
Effects of the intestinal microbial metabolite butyrate on the development of colorectal cancer
.
J Cancer
.
2018
;
9
(
14
):
2510
7
.
145.
Bindels
LB
,
Porporato
P
,
Dewulf
EM
,
Verrax
J
,
Neyrinck
AM
,
Martin
JC
,
.
Gut microbiota-derived propionate reduces cancer cell proliferation in the liver
.
Br J Cancer
.
2012 Oct
;
107
(
8
):
1337
44
.
146.
Li
M
,
van Esch
BCAM
,
Henricks
PAJ
,
Folkerts
G
,
Garssen
J
.
The anti-inflammatory effects of short chain fatty acids on lipopolysaccharide- or tumor necrosis factor α-stimulated endothelial cells via activation of GPR41/43 and inhibition of HDACs
.
Front Pharmacol
.
2018 May
;
9
:
533
.
147.
Licciardi
PV
,
Ververis
K
,
Karagiannis
TC
.
Histone deacetylase inhibition and dietary short-chain fatty acids
.
ISRN Allergy
.
2011
;
2011
:
1
8
.
148.
Yuille
S
,
Reichardt
N
,
Panda
S
,
Dunbar
H
,
Mulder
IE
.
Human gut bacteria as potent class I histone deacetylase inhibitors in vitro through production of butyric acid and valeric acid
.
PLoS One
.
2018 Jul
;
13
(
7
):
e0201073
.
149.
Zagato
E
,
Pozzi
C
,
Bertocchi
A
,
Schioppa
T
,
Saccheri
F
,
Guglietta
S
,
.
Endogenous murine microbiota member Faecalibaculum rodentium and its human homologue protect from intestinal tumour growth
.
Nat Microbiol
.
2020 Mar
;
5
(
3
):
511
24
.
150.
Peuker
K
,
Muff
S
,
Wang
J
,
Künzel
S
,
Bosse
E
,
Zeissig
Y
,
.
Epithelial calcineurin controls microbiota-dependent intestinal tumor development
.
Nat Med
.
2016 May
;
22
(
5
):
506
15
.
151.
Singh
N
,
Gurav
A
,
Sivaprakasam
S
,
Brady
E
,
Padia
R
,
Shi
H
,
.
Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis
.
Immunity
.
2014 Jan
;
40
(
1
):
128
39
.
152.
Komatsu
S
,
Yanaka
N
,
Matsubara
K
,
Kato
N
.
Antitumor effect of vitamin B6 and its mechanisms
.
Biochim Biophys Acta
.
2003 Apr
;
1647
(
1–2
):
127
30
.
153.
Yoshii
K
,
Hosomi
K
,
Sawane
K
,
Kunisawa
J
.
Metabolism of dietary and microbial vitamin B family in the regulation of host immunity
.
Front Nutr
.
2019 Apr
;
6
:
48
.
154.
Konishi
H
,
Fujiya
M
,
Tanaka
H
,
Ueno
N
,
Moriichi
K
,
Sasajima
J
,
.
Probiotic-derived ferrichrome inhibits colon cancer progression via JNK-mediated apoptosis
.
Nat Commun
.
2016 Nov
;
7
(
1
):
12365
.
155.
Nouri
Z
,
Karami
F
,
Neyazi
N
,
Modarressi
MH
,
Karimi
R
,
Khorramizadeh
MR
,
.
Dual anti-metastatic and anti-proliferative activity assessment of two probiotics on HeLa and HT-29 cell lines
.
Cell J
.
2016 Jul
;
18
(
2
):
127
34
.
156.
Tiptiri-Kourpeti
A
,
Spyridopoulou
K
,
Santarmaki
V
,
Aindelis
G
,
Tompoulidou
E
,
Lamprianidou
EE
,
.
Lactobacillus casei exerts anti-proliferative effects accompanied by apoptotic cell death and up-regulation of TRAIL in colon carcinoma cells
.
PLoS One
.
2016 Feb
;
11
(
2
):
e0147960
.
157.
Gill
HS
,
Rutherfurd
KJ
,
Prasad
J
,
Gopal
PK
.
Enhancement of natural and acquired immunity by Lactobacillus rhamnosus (HN001), Lactobacillus acidophilus (HN017) and Bifidobacterium lactis (HN019)
.
Br J Nutr
.
2000 Feb
;
83
(
2
):
167
76
.
158.
Watanabe
T
,
Nishio
H
,
Tanigawa
T
,
Yamagami
H
,
Okazaki
H
,
Watanabe
K
,
.
Probiotic Lactobacillus casei strain Shirota prevents indomethacin-induced small intestinal injury: involvement of lactic acid
.
Am J Physiol Liver Physiol
.
2009 Sep
;
297
(
3
):
G506
13
.
159.
Matsumoto
S
,
Hara
T
,
Hori
T
,
Mitsuyama
K
,
Nagaoka
M
,
Tomiyasu
N
,
.
Probiotic Lactobacillus-induced improvement in murine chronic inflammatory bowel disease is associated with the down-regulation of pro-inflammatory cytokines in lamina propria mononuclear cells
.
Clin Exp Immunol
.
2005 Jun
;
140
(
3
):
417
26
.
160.
Monteleone
I
,
Rizzo
A
,
Sarra
M
,
Sica
G
,
Sileri
P
,
Biancone
L
,
.
Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract
.
Gastroenterology
.
2011 Jul
;
141
(
1
):
237
48.e1
.
161.
Alexeev
EE
,
Lanis
JM
,
Kao
DJ
,
Campbell
EL
,
Kelly
CJ
,
Battista
KD
,
.
Microbiota-derived indole metabolites promote human and murine intestinal homeostasis through regulation of interleukin-10 receptor
.
Am J Pathol
.
2018 May
;
188
(
5
):
1183
94
.
162.
Mohamed
HT
,
Gadalla
R
,
El-Husseiny
N
,
Hassan
H
,
Wang
Z
,
Ibrahim
SA
,
.
Inflammatory breast cancer: activation of the aryl hydrocarbon receptor and its target CYP1B1 correlates closely with Wnt5a/b-β-catenin signalling, the stem cell phenotype and disease progression
.
J Adv Res
.
2019 Mar
;
16
:
75
86
.
163.
Shimba
S
,
Komiyama
K
,
Moro
I
,
Tezuka
M
.
Overexpression of the aryl hydrocarbon receptor (AhR) accelerates the cell proliferation of A549 cells
.
J Biochem
.
2002 Nov
;
132
(
5
):
795
802
.
164.
Yin
XF
,
Chen
J
,
Mao
W
,
Wang
YH
,
Chen
MH
.
Downregulation of aryl hydrocarbon receptor expression decreases gastric cancer cell growth and invasion
.
Oncol Rep
.
2013 Jul
;
30
(
1
):
364
70
.
165.
Lin
P
,
Chang
H
,
Tsai
W-T
,
Wu
M-H
,
Liao
Y-S
,
Chen
J-T
,
.
Overexpression of aryl hydrocarbon receptor in human lung carcinomas
.
Toxicol Pathol
.
2003 Jan
;
31
(
1
):
22
30
.
166.
Ye
M
,
Zhang
Y
,
Gao
H
,
Xu
Y
,
Jing
P
,
Wu
J
,
.
Activation of the aryl hydrocarbon receptor leads to resistance to EGFR TKIs in non–small cell lung cancer by activating Src-mediated bypass signaling
.
Clin Cancer Res
.
2018 Mar
;
24
(
5
):
1227
39
.
167.
Bultman
SJ
.
Emerging roles of the microbiome in cancer
.
Carcinogenesis
.
2014 Feb
;
35
(
2
):
249
55
.
168.
Arthur
JC
,
Perez-Chanona
E
,
Mühlbauer
M
,
Tomkovich
S
,
Uronis
JM
,
Fan
T-J
,
.
Intestinal inflammation targets cancer-inducing activity of the microbiota
.
Science
.
2012 Oct
;
338
(
6103
):
120
3
.
169.
Zamani
S
,
Hesam Shariati
S
,
Zali
MR
,
Asadzadeh Aghdaei
H
,
Sarabi Asiabar
A
,
Bokaie
S
,
.
Detection of enterotoxigenic Bacteroides fragilis in patients with ulcerative colitis
.
Gut Pathog
.
2017 Dec
;
9
(
1
):
53
.
170.
Liao
JB
.
Viruses and human cancer
.
Yale J Biol Med
.
2006 Dec
;
79
(
3–4
):
115
22
.
171.
White
MK
,
Pagano
JS
,
Khalili
K
.
Viruses and human cancers: a long road of discovery of molecular paradigms
.
Clin Microbiol Rev
.
2014 Jul
;
27
(
3
):
463
81
.
172.
Vivarelli
S
,
Salemi
R
,
Candido
S
,
Falzone
L
,
Santagati
M
,
Stefani
S
,
.
Gut microbiota and cancer: from pathogenesis to therapy
.
Cancers
.
2019 Jan
;
11
(
1
):
38
.
173.
Gillet
E
,
Meys
JFA
,
Verstraelen
H
,
Bosire
C
,
De Sutter
P
,
Temmerman
M
,
.
Bacterial vaginosis is associated with uterine cervical human papillomavirus infection: a meta-analysis
.
BMC Infect Dis
.
2011 Dec
;
11
(
1
):
10
.
174.
Suehiro
TT
,
Malaguti
N
,
Damke
E
,
Uchimura
NS
,
Gimenes
F
,
Souza
RP
,
.
Association of human papillomavirus and bacterial vaginosis with increased risk of high-grade squamous intraepithelial cervical lesions
.
Int J Gynecol Cancer
.
2019 Feb
;
29
(
2
):
242
9
.
175.
Łaniewski
P
,
Barnes
D
,
Goulder
A
,
Cui
H
,
Roe
DJ
,
Chase
DM
,
.
Linking cervicovaginal immune signatures, HPV and microbiota composition in cervical carcinogenesis in non-hispanic and hispanic women
.
Sci Rep
.
2018 Dec
;
8
(
1
):
7593
.
176.
Murta
EFC
,
Souza
MAHde
,
Araújo Júnior
E
,
Adad
SJ
.
Incidence of Gardnerella vaginalis, Candida sp and human papilloma virus in cytological smears
.
Sao Paulo Med J
.
2000 Jul
;
118
(
4
):
105
8
.
177.
Usyk
M
,
Zolnik
CP
,
Castle
PE
,
Porras
C
,
Herrero
R
,
Gradissimo
A
,
.
Cervicovaginal microbiome and natural history of HPV in a longitudinal study
.
PLoS Pathog
.
2020 Mar
;
16
(
3
):
e1008376
.
178.
Gheit
T
.
Mucosal and cutaneous human papillomavirus infections and cancer biology
.
Front Oncol
.
2019 May
;
9
:
355
.
179.
Herfs
M
,
Soong
T
,
Delvenne
P
,
Crum
C
.
Deciphering the multifactorial susceptibility of mucosal junction cells to HPV infection and related carcinogenesis
.
Viruses
.
2017 Apr
;
9
(
4
):
85
.
180.
Marshall
BJ
,
Warren
JR
.
Unidentified curved Bacilli in the stomach of patients with gastritis and peptidic ulceration
.
Lancet
.
1984 Jun
;
323
(
8390
):
1311
5
.
181.
Schistosomes, liver flukes and Helicobacter pylori. IARC Working Group on the evaluation of carcinogenic risks to humans. Lyon, 7–14 June 1994. IARC Monogr Eval Carcinog risks to humans. 1994;61:1–241
.
182.
Peleteiro
B
,
Bastos
A
,
Ferro
A
,
Lunet
N
.
Prevalence of Helicobacter pylori infection worldwide: a systematic review of studies with national coverage
.
Dig Dis Sci
.
2014 Aug
;
59
(
8
):
1698
709
.
183.
Dunne
C
.
Factors that mediate colonization of the human stomach by Helicobacter pylori
.
World J Gastroenterol
.
2014
;
20
(
19
):
5610
.
184.
Fazeli
Z
,
Alebouyeh
M
,
Rezaei Tavirani
M
,
Azimirad
M
,
Yadegar
A
.
Helicobacter pylori CagA induced interleukin-8 secretion in gastric epithelial cells
.
Gastroenterol Hepatol Bed Bench
.
2016 Dec
;
9
(
Suppl 1
):
S42
6
.
185.
Audibert
C
,
Janvier
B
,
Grignon
B
,
Salaüna
L
,
Burucoa
C
,
Lecron
J-C
,
.
Correlation between IL-8 induction, cagA status and vacA genotypes in 153 French Helicobacter pylori isolates
.
Res Microbiol
.
2000 Apr
;
151
(
3
):
191
200
.
186.
Lin
Q
,
Xu
H
,
Chen
X
,
Tang
G
,
Gu
L
,
Wang
Y
.
Helicobacter pylori cytotoxin.associated gene-A activates tumor necrosis factor-α and interleukin-6 in gastric epithelial cells through P300/CBP-associated factor-mediated nuclear factor-κB p65 acetylation
.
Mol Med Rep
.
2015 Oct
;
12
(
4
):
6337
45
.
187.
Nakagawa
H
,
Tamura
T
,
Mitsuda
Y
,
Goto
Y
,
Kamiya
Y
,
Kondo
T
,
.
Significant association between serum interleukin-6 and Helicobacter pylori antibody levels among H. pylori -positive Japanese adults
.
Mediators Inflamm
.
2013
;
2013
:
1
5
.
188.
Maeda
S
,
Yoshida
H
,
Ogura
K
,
Mitsuno
Y
,
Hirata
Y
,
Yamaji
Y
,
.
H. pylori activates NF-κB through a signaling pathway involving IκB kinases, NF-κB—inducing kinase, TRAF2, and TRAF6 in gastric cancer cells
.
Gastroenterology
.
2000 Jul
;
119
(
1
):
97
108
.
189.
Ferrero
RL
,
Avé
P
,
Ndiaye
D
,
Bambou
J-C
,
Huerre
MR
,
Philpott
DJ
,
.
NF-κB activation during acute Helicobacter pylori infection in mice
.
Infect Immun
.
2008 Feb
;
76
(
2
):
551
61
.
190.
Sears
CL
.
The toxins of Bacteroides fragilis
.
Toxicon
.
2001 Nov
;
39
(
11
):
1737
46
.
191.
Wu
S
,
Shin
J
,
Zhang
G
,
Cohen
M
,
Franco
A
,
Sears
CL
.
The Bacteroides fragilis toxin binds to a specific intestinal epithelial cell receptor
.
Infect Immun
.
2006 Sep
;
74
(
9
):
5382
90
.
192.
Haghi
F
,
Goli
E
,
Mirzaei
B
,
Zeighami
H
.
The association between fecal enterotoxigenic B. fragilis with colorectal cancer
.
BMC Cancer
.
2019 Dec
;
19
(
1
):
879
.
193.
Purcell
RV
,
Pearson
J
,
Aitchison
A
,
Dixon
L
,
Frizelle
FA
,
Keenan
JI
.
Colonization with enterotoxigenic Bacteroides fragilis is associated with early-stage colorectal neoplasia
.
PLoS One
.
2017 Feb
;
12
(
2
):
e0171602
.
194.
Rhee
K-J
,
Wu
S
,
Wu
X
,
Huso
DL
,
Karim
B
,
Franco
AA
,
.
Induction of persistent colitis by a human commensal, enterotoxigenic Bacteroides fragilis, in wild-type C57BL/6 mice
.
Infect Immun
.
2009 Apr
;
77
(
4
):
1708
18
.
195.
Wu
S
,
Rhee
K-J
,
Zhang
M
,
Franco
A
,
Sears
CL
.
Bacteroides fragilis toxin stimulates intestinal epithelial cell shedding and -secretase-dependent E-cadherin cleavage
.
J Cell Sci
.
2007 Jun
;
120
(
11
):
1944
52
.
196.
Hwang
S
,
Gwon
S-Y
,
Kim
MS
,
Lee
S
,
Rhee
K-J
.
Bacteroides fragilis toxin induces IL-8 secretion in HT29/C1 cells through disruption of E-cadherin junctions
.
Immune Netw
.
2013
;
13
(
5
):
213
.
197.
Wu
S
,
Morin
PJ
,
Maouyo
D
,
Sears
CL
.
Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation
.
Gastroenterology
.
2003 Feb
;
124
(
2
):
392
400
.
198.
Wu
J
,
Li
Q
,
Fu
X
.
Fusobacterium nucleatum contributes to the carcinogenesis of colorectal cancer by inducing inflammation and suppressing host immunity
.
Transl Oncol
.
2019 Jun
;
12
(
6
):
846
51
.
199.
Sun
C-H
,
Li
B-B
,
Wang
B
,
Zhao
J
,
Zhang
X-Y
,
Li
T-T
,
.
The role of Fusobacterium nucleatum in colorectal cancer: from carcinogenesis to clinical management
.
Chronic Dis Transl Med
.
2019 Sep
;
5
(
3
):
178
87
.
200.
Rubinstein
MR
,
Wang
X
,
Liu
W
,
Hao
Y
,
Cai
G
,
Han
YW
.
Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin
.
Cell Host Microbe
.
2013 Aug
;
14
(
2
):
195
206
.
201.
Abed
J
,
Emgård
JEM
,
Zamir
G
,
Faroja
M
,
Almogy
G
,
Grenov
A
,
.
Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc
.
Cell Host Microbe
.
2016 Aug
;
20
(
2
):
215
25
.
202.
Gur
C
,
Ibrahim
Y
,
Isaacson
B
,
Yamin
R
,
Abed
J
,
Gamliel
M
,
.
Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack
.
Immunity
.
2015 Feb
;
42
(
2
):
344
55
.
203.
Kostic
AD
,
Chun
E
,
Robertson
L
,
Glickman
JN
,
Gallini
CA
,
Michaud
M
,
.
Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment
.
Cell Host Microbe
.
2013 Aug
;
14
(
2
):
207
15
.
204.
Chen
T
,
Li
Q
,
Wu
J
,
Wu
Y
,
Peng
W
,
Li
H
,
.
Fusobacterium nucleatum promotes M2 polarization of macrophages in the microenvironment of colorectal tumours via a TLR4-dependent mechanism
.
Cancer Immunol Immunother
.
2018 Oct
;
67
(
10
):
1635
46
.
205.
Palmer
AC
,
Sorger
PK
.
Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy
.
Cell
.
2017 Dec
;
171
(
7
):
1678
91.e13
.
206.
Koch
G
,
Walz
A
,
Lahu
G
,
Schropp
J
.
Modeling of tumor growth and anticancer effects of combination therapy
.
J Pharmacokinet Pharmacodyn
.
2009 Apr
;
36
(
2
):
179
97
.
207.
Oiseth
SJ
,
Aziz
MS
.
Cancer immunotherapy: a brief review of the history, possibilities, and challenges ahead
.
J Cancer Metastasis Treat
.
2017 Oct
;
3
(
10
):
250
.
208.
Dobosz
P
,
Dzieciątkowski
T
.
The intriguing history of cancer immunotherapy
.
Front Immunol
.
2019 Dec
;
10
:
2965
.
209.
Lotze
MT
,
Frana
LW
,
Sharrow
SO
,
Robb
RJ
,
Rosenberg
SA
.
In vivo administration of purified human interleukin 2. I. Half-life and immunologic effects of the Jurkat cell line-derived interleukin 2
.
J Immunol
.
1985 Jan
[cited 2020 Jun 23];
134
(
1
):
157
66
.
210.
Rosenberg
SA
,
Mulé
JJ
,
Spiess
PJ
,
Reichert
CM
,
Schwarz
SL
.
Regression of established pulmonary metastases and subcutaneous tumor mediated by the systemic administration of high-dose recombinant interleukin 2
.
J Exp Med
.
1985 May
;
161
(
5
):
1169
88
.
211.
Jiang
T
,
Zhou
C
,
Ren
S
.
Role of IL-2 in cancer immunotherapy
.
Oncoimmunology
.
2016 Jun
;
5
(
6
):
e1163462
.
212.
Patel
JN
,
Walko
CM
.
Sylatron: a pegylated interferon for use in melanoma
.
Ann Pharmacother
.
2012 Jun
;
46
(
6
):
830
8
.
213.
Davar
D
,
Kirkwood
JM
.
Adjuvant therapy of melanoma
.
Cancer treatment and research
.
Kluwer Academic Publishers
;
2016
. p.
181
208
.
214.
Berraondo
P
,
Sanmamed
MF
,
Ochoa
MC
,
Etxeberria
I
,
Aznar
MA
,
Pérez-Gracia
JL
,
.
Cytokines in clinical cancer immunotherapy
.
Br J Cancer
.
2019 Jan
;
120
(
1
):
6
15
.
215.
Li
B
,
Chan
HL
,
Chen
P
.
Immune checkpoint inhibitors: basics and challenges
.
Curr Med Chem
.
2019 Aug
;
26
(
17
):
3009
25
.
216.
Buchbinder
EI
,
Desai
A
.
CTLA-4 and PD-1 pathways
.
Am J Clin Oncol
.
2016 Feb
;
39
(
1
):
98
106
.
217.
Wei
SC
,
Duffy
CR
,
Allison
JP
.
Fundamental mechanisms of immune checkpoint blockade therapy
.
Cancer Discov
.
2018 Sep
;
8
(
9
):
1069
86
.
218.
Okazaki
T
,
Honjo
T
.
The PD-1–PD-L pathway in immunological tolerance
.
Trends Immunol
.
2006 Apr
;
27
(
4
):
195
201
.
219.
Keir
ME
,
Liang
SC
,
Guleria
I
,
Latchman
YE
,
Qipo
A
,
Albacker
LA
,
.
Tissue expression of PD-L1 mediates peripheral T cell tolerance
.
J Exp Med
.
2006 Apr
;
203
(
4
):
883
95
.
220.
Leach
DR
,
Krummel
MF
,
Allison
JP
.
Enhancement of antitumor immunity by CTLA-4 blockade
.
Science
.
1996 Mar
;
271
(
5256
):
1734
6
.
221.
Ariyan
CE
,
Brady
MS
,
Siegelbaum
RH
,
Hu
J
,
Bello
DM
,
Rand
J
,
.
Robust antitumor responses result from local chemotherapy and CTLA-4 blockade
.
Cancer Immunol Res
.
2018 Feb
;
6
(
2
):
189
200
.
222.
Lesterhuis
WJ
,
Salmons
J
,
Nowak
AK
,
Rozali
EN
,
Khong
A
,
Dick
IM
,
.
Synergistic effect of CTLA-4 blockade and cancer chemotherapy in the induction of anti-tumor immunity
.
PLoS One
.
2013 Apr
;
8
(
4
):
e61895
.
223.
Hodi
FS
,
O’Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
,
.
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
.
2010 Aug
;
363
(
8
):
711
23
.
224.
Prieto
PA
,
Yang
JC
,
Sherry
RM
,
Hughes
MS
,
Kammula
US
,
White
DE
,
.
CTLA-4 blockade with Ipilimumab: long-term follow-up of 177 patients with metastatic melanoma
.
Clin Cancer Res
.
2012 Apr
;
18
(
7
):
2039
47
.
225.
Fellner
C
.
Ipilimumab (yervoy) prolongs survival in advanced melanoma: serious side effects and a hefty price tag may limit its use
.
P T
.
2012 Sep
[cited 2020 Jun 23];
37
(
9
):
503
30
.
226.
Chen
S
,
Crabill
GA
,
Pritchard
TS
,
McMiller
TL
,
Wei
P
,
Pardoll
DM
,
.
Mechanisms regulating PD-L1 expression on tumor and immune cells
.
J Immunother Cancer
.
2019 Dec
;
7
(
1
):
305
.
227.
Ahmadzadeh
M
,
Johnson
LA
,
Heemskerk
B
,
Wunderlich
JR
,
Dudley
ME
,
White
DE
,
.
Tumor antigen–specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired
.
Blood
.
2009 Aug
;
114
(
8
):
1537
44
.
228.
Opdivo (nivolumab) [prescribing information]
.
Princeton, NJ
:
Bristol-Myers Squibb
;
2018
.
229.
Keytruda (pembrolizumab) [prescribing information]
.
Whitehouse Station, NJ
:
Merck & Co, Inc
;
2017
.
230.
Tecentriq (atezolizumab) [prescribing information]
.
South San Francisco, CA
:
Genentech
;
2018
.
231.
Imfinzi (durvalumab) [prescribing information]
.
Cambridge, England
:
AstraZeneca, Inc
;
2018
.
232.
Bavencio (avelumab) [prescribing information]
.
Rockland, MA
:
EMD Serono, Inc
;
2017
.
233.
Yarchoan
M
,
Albacker
LA
,
Hopkins
AC
,
Montesion
M
,
Murugesan
K
,
Vithayathil
TT
,
.
PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers
.
JCI Insight
.
2019 Mar
;
4
(
6
):
e126908
.
234.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
,
.
Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer
.
Science
.
2015 Apr
;
348
(
6230
):
124
8
.
235.
Wu
K
,
Yi
M
,
Qin
S
,
Chu
Q
,
Zheng
X
,
Wu
K
.
The efficacy and safety of combination of PD-1 and CTLA-4 inhibitors: a meta-analysis
.
Exp Hematol Oncol
.
2019 Dec
;
8
(
1
):
26
.
236.
Rotte
A
.
Combination of CTLA-4 and PD-1 blockers for treatment of cancer
.
J Exp Clin Cancer Res
.
2019 Dec
;
38
(
1
):
255
.
237.
Johnson
DB
,
Balko
JM
,
Compton
ML
,
Chalkias
S
,
Gorham
J
,
Xu
Y
,
.
Fulminant myocarditis with combination immune checkpoint blockade
.
N Engl J Med
.
2016 Nov
;
375
(
18
):
1749
55
.
238.
Cappelli
LC
,
Gutierrez
AK
,
Baer
AN
,
Albayda
J
,
Manno
RL
,
Haque
U
,
.
Inflammatory arthritis and sicca syndrome induced by nivolumab and ipilimumab
.
Ann Rheum Dis
.
2017 Jan
;
76
(
1
):
43
50
.
239.
Cousin
S
,
Seneschal
J
,
Italiano
A
.
Toxicity profiles of immunotherapy
.
Pharmacol Ther
.
2018 Jan
;
181
:
91
100
.
240.
Brignone
C
,
Escudier
B
,
Grygar
C
,
Marcu
M
,
Triebel
F
.
A phase I pharmacokinetic and biological correlative study of IMP321, a novel MHC class II agonist, in patients with advanced renal cell carcinoma
.
Clin Cancer Res
.
2009 Oct
;
15
(
19
):
6225
31
.
241.
Liu
J
,
Zhang
S
,
Hu
Y
,
Yang
Z
,
Li
J
,
Liu
X
,
.
Targeting PD-1 and Tim-3 pathways to reverse CD8 T-cell exhaustion and enhance ex vivo T-cell responses to autologous dendritic/tumor vaccines
.
J Immunother
.
2016 May
;
39
(
4
):
171
80
.
242.
Sakuishi
K
,
Apetoh
L
,
Sullivan
JM
,
Blazar
BR
,
Kuchroo
VK
,
Anderson
AC
.
Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity
.
J Exp Med
.
2010 Sep
;
207
(
10
):
2187
94
.
243.
Ngiow
SF
,
von Scheidt
B
,
Akiba
H
,
Yagita
H
,
Teng
MWL
,
Smyth
MJ
.
Anti-TIM3 antibody promotes T cell IFN: mediated antitumor immunity and suppresses established tumors
.
Cancer Res
.
2011 May
;
71
(
10
):
3540
51
.
244.
Coley
WB
.
The treatment of inoperable sarcoma by bacterial toxins (the mixed toxins of the Streptococcus erysipelas and the Bacillus prodigiosus)
.
Proc R Soc Med
.
1910 Jun
;
3
(
Surg_Sect
):
1
48
.
245.
Old
LJ
,
Clarke
DA
,
Benacerraf
B
.
Effect of bacillus calmette-guérin infection on transplanted tumours in the mouse
.
Nature
.
1959
;
184
(
4682
):
291
2
.
246.
Mathé
G
,
Amiel
JL
,
Schwarzenberg
L
,
Schneider
M
,
Cattan
A
,
Schlumberger
JR
,
.
Active immunotherapy for acute lymphoblastic leukaemia
.
Lancet
.
1969 Apr
;
1
(
7597
):
697
9
.
247.
Sivan
A
,
Corrales
L
,
Hubert
N
,
Williams
JB
,
Aquino-Michaels
K
,
Earley
ZM
,
.
Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy
.
Science
.
2015 Nov
;
350
(
6264
):
1084
9
.
248.
Matson
V
,
Fessler
J
,
Bao
R
,
Chongsuwat
T
,
Zha
Y
,
Alegre
M-L
,
.
The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients
.
Science
.
2018 Jan
;
359
(
6371
):
104
8
.
249.
Chaput
N
,
Lepage
P
,
Coutzac
C
,
Soularue
E
,
Le Roux
K
,
Monot
C
,
.
Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab
.
Ann Oncol
.
2017 Jun
;
28
(
6
):
1368
79
.
250.
Gui
Q-F
,
Lu
H-F
,
Zhang
C-X
,
Xu
Z-R
,
Yang
Y-H
.
Well-balanced commensal microbiota contributes to anti-cancer response in a lung cancer mouse model
.
Genet Mol Res
.
2015 May
;
14
(
2
):
5642
51
.
251.
Viaud
S
,
Saccheri
F
,
Mignot
G
,
Yamazaki
T
,
Daillere
R
,
Hannani
D
,
.
The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide
.
Science
.
2013 Nov
;
342
(
6161
):
971
6
.
252.
Iida
N
,
Dzutsev
A
,
Stewart
CA
,
Smith
L
,
Bouladoux
N
,
Weingarten
RA
,
.
Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment
.
Science
.
2013 Nov
;
342
(
6161
):
967
70
.