Immunoprevention refers to a strategy of preventing pathogen-associated and spontaneous cancers through the use of vaccines, antibodies, and immune modulators. Immune modulators function by enhancing the endogenous ability of the immune system to monitor for malignancy, so-called “immunosurveillance.” There is growing evidence that many of the most promising cancer chemoprevention agents including aspirin, COX-2 inhibitors, aromatase inhibitors, and bisphosphonates mediate their effects, in part, by enhancing immunosurveillance and reversing the immune evasive mechanisms that premalignant lesions use. In the following review, we introduce critical components of the human immune surveillance system—dendritic cells, T cells, and immune suppressive cells—and discuss the emerging data suggesting that common chemoprevention agents may modulate the function of these immunologic cells. Cancer Prev Res; 6(8); 764–73. ©2013 AACR.

Cancer prevention strategies include (i) risk reduction through elimination of environmental factors (asbestos, tobacco, and alcohol); (ii) chemoprevention in high-risk populations with agents such as COX-2 inhibitors and aromatase inhibitors; and (iii) immunoprevention with vaccines, antibodies, and immune modulators. The most successful application of immunoprevention to date has been vaccination against the infectious causative agents of hepatocellular carcinoma (HCC) and cervical cancer, 2 of the most common cancers worldwide. In prevention of HCC, a nationwide hepatitis B vaccination program in Taiwan was shown to reduce the average annual incidence of HCC in children over several years from 0.7 per 100,000 children (1981–1986) to 0.57 (1986–1990) and 0.36 (1990–1994) with a decrease in corresponding rates of mortality (1). In the prevention of cervical and other gynecologic cancers related to human papilloma virus (HPV), 2 international, double blind, placebo-controlled randomized trials (FUTURE I/II) evaluated the efficacy of the quadrivalent HPV vaccine (serotypes 6, 11, 16, and 18). In lesions caused by virus corresponding to the specific serotypes included in the quadrivalent vaccine, efficacy at preventing cervical intraepithelial neoplasia grade 1 was 96% and for vulvar and vaginal intraepithelial neoplasia reached 100% (2). Vaccines designed to stop infection-associated cancers have been one of the most successful prevention strategies to date.

Most cancers, however, have not been shown to be caused by infectious agents, instead arising from genetic alterations, and the immune system may inhibit tumor growth even in this setting. Thomas and Burnet developed the “tumor surveillance” theory in the 1950s in which they hypothesized that the immune system protects against nascent cancers by destroying abnormal cells before evolution to invasive malignancy. Burnet predicted that “if there were tumor immunity, it would be invisible,” anticipating the difficulty of providing evidence in humans to support immune surveillance (3). Evidence for the role of the immune system in modulating the growth of common cancers now exists. First, many cancer patients across a variety of tumor types spontaneously develop significant levels of antibodies and/or T cells specific for antigens expressed on their tumors, which, in some cases, are associated with prognosis including the occasional spontaneous regression (4). Second, the composition of tumor-infiltrating lymphocytes (TIL) has been shown to have prognostic implications in a variety of different malignancies (5). Indeed, for colon cancer an immune scoring system based on enumerating CD8+ T cells and memory T cells can predict prognosis with greater accuracy than standard tumor-node-metastasis staging (6). Third, immunodeficiency has been associated with cancer risk. Patients with impaired immunity, for example HIV infection or the use of antirejection drugs for transplantation, have a higher risk of both virally associated and nonvirally induced tumors, suggesting a cancer protective effect via an intact immune system (7).

Standard cytotoxic chemotherapy has long been thought to work primarily by selectively causing death of rapidly proliferating tumor cells. Recently, however, many chemotherapeutic agents have been shown to stimulate tumor-specific immune responses by inducing immunogenic cell death or stimulating/activating immune effector cells which contribute to drug efficacy (8). Within the nascent field of cancer immunoprevention, similar data are emerging that many of the most promising chemoprevention agents under study may exert their effects, in part, by enhancing immune surveillance. As with cytotoxic drugs, chemoprevention agents have been shown to increase antigen processing by potent antigen-presenting cells (APC), stimulate the proliferation and antitumor capabilities of CD8+ T cells, and inhibit the function or decrease the number of immune suppressive cells. In the following review, we discuss relevant elements of cancer destructive immunity and explain how chemoprevention agents may have immunomodulatory effects.

Dendritic cells are the most effective of the APCs in presenting immunogenic proteins to T cells. Dendritic cells sample antigens in peripheral tissues and process them into small peptides as they mature and migrate to lymphoid organs. After antigen uptake, APCs must receive suitable activation or stimulatory signals that result in sufficient maturation so that they differentiate to promote immunity rather than tolerance, as most immunogenic cancer-associated proteins are self-antigens. Once activated, APCs present processed peptides to naive T cells, stimulating a cellular immune response composed of CD4+ T helper cells (TH) and cytotoxic effector CD8+ T cells that are critical for destruction of preinvasive and invasive lesions (Fig. 1A and B; ref. 9).

Figure 1.

Cancer chemoprevention agents enhance the immunologic activity of APCs. A, PGE2 can inhibit the ability of maturing dendtritic cells (DC) to produce IL-12 during priming, biasing, the resulting adaptive immune response toward a TH2 profile. COX-2 inhibitors such as aspirin and celecoxib can reverse this effect, allowing generation of TH1. B, PGE2 facilitates differentiation of monocytes into immunosuppressive MDSCs, which function to inhibit the adaptive immune response and promote Treg populations through depletion of environmental arginine, expression of nitric oxide synthase, production of reactive oxygen species, and elaboration of TH2 cytokines IL-10 and TGF-B. COX-2 inhibitors aspirin and celecoxib can reverse this effect.

Figure 1.

Cancer chemoprevention agents enhance the immunologic activity of APCs. A, PGE2 can inhibit the ability of maturing dendtritic cells (DC) to produce IL-12 during priming, biasing, the resulting adaptive immune response toward a TH2 profile. COX-2 inhibitors such as aspirin and celecoxib can reverse this effect, allowing generation of TH1. B, PGE2 facilitates differentiation of monocytes into immunosuppressive MDSCs, which function to inhibit the adaptive immune response and promote Treg populations through depletion of environmental arginine, expression of nitric oxide synthase, production of reactive oxygen species, and elaboration of TH2 cytokines IL-10 and TGF-B. COX-2 inhibitors aspirin and celecoxib can reverse this effect.

Close modal

Aspirin and the COX-2 inhibitors are well-studied chemoprevention agents. Aspirin and the COX-2 inhibitor celecoxib have both been shown to reduce the risk of colorectal cancer (10–14). In addition, systematic reviews of the results of aspirin in cardiovascular studies have suggested that low-dose aspirin reduces overall cancer incidence and mortality (15–17). Nonsteroidal anti-inflammatory drugs (NSAID) may limit carcinogenesis by preventing prostaglandin E2 (PGE2)-mediated inhibition of dendritic cells. PGE2 is a product of COX enzymes and, normally, mediates physiologic functions such as maintenance of gastric mucosal integrity and renal blood flow when constitutively expressed. However, components of the tumor microenvironment can also produce PGE2 through COX-2 expression during oncogenesis. PGE2 alters the balance and function of dendritic cells in different ways dependent on their maturation state at the time of exposure to the prostaglandin. Early in development, PGE2 has been shown to suppress differentiation of human monocytes into functional TH1-inducing APC (Fig. 1A), instead redirecting monocytes to become immunosuppressive MDSCs (Fig. 1B). TH1 are critical in mediating tumor regression (9). When monocyte-derived immature dendritic cells are matured with interleukin (IL)-1β and TNF-α in the presence of PGE2in vitro, resultant dendritic cells are phenotypically identical but have reduced capacity to produce IL-12 when stimulated with CD40L and IFN-γ (Fig. 1A). IL-12 is necessary for efficient generation of TH1 and cytotoxic T cells (type I cells). Although naive TH cells primed with PGE2–dendritic cells have similar expansion kinetics as compared to controls, the cells also have an enhanced ability to produce TH2-type cytokines, IL-4 and IL-5, and a reduced ability to produce the TH1 cytokine IFN-γ. TH2 cells can contribute to tumor growth by dampening the generation of cytotoxic T cells and support cancer proliferation. The effects of PGE2 on dendritic cells may be most important early in oncogenesis, as cervical, breast, head and neck, and colorectal precursor lesions have been shown to overexpress COX-2 (18–21). In a cervical model, COX-2 expression was inversely correlated with density of dendritic cells and ability to stimulate T cells (20). NSAIDs and COX-2 inhibitors may be particularly suited to cancer prevention by protecting the integrity of dendritic cells in mediating immunosurveillance by allowing selective induction and proliferation of type I T cells (Fig. 1).

APCs must induce protective T-cell responses through antigen recognition (9). This adaptive T-cell response is largely composed of TH1 CD4+ T cells and CD8+ T cells. TH1 cells are critical for propagation of the acute tissue destructive inflammatory response, secreting cytokines such as IFN-γ, TNF-α, and IL-2 that support CD8+ T cells and tumor destruction. This is in contrast to TH2 cells that secrete cytokines such as IL-4, IL-5, and IL-10, which limit CD8+ T-cell proliferation and promote tumor growth (Fig. 2A). CD8+ T cells are activated after binding antigen presented by MHC class I molecules on APCs and some tumor cells and can deliver cytokines and cytotoxic enzymes that result in tumor cell lysis (9). Lesions that escape cell-mediated death do so by subverting this arm of the immune system, shifting the tumor environment to a TH2 type and inhibiting proliferation of CD8+ T cells. Cancer escapes immune surveillance in a myriad of ways including down regulation of MHC class I molecules, rendering them invisible to CD8+ T cells; production of factors that inhibit CD8+ T-cell survival and expansion; and production of cytokines and chemokines that attract immune suppressive cells (22).

Figure 2.

Cancer chemoprevention agents stimulate the adaptive immune system. A, curcumin administration can enhance the TH1 and decrease TH2 immune response. B, metformin may increase MHC-I expression on tumor cells, increasing visibility to effector CD8+ T cells. C, ZA and other bisphosphonates increase phosphoantigens in PBMCs and on cancer cells themselves, resulting in activation of anti-tumor γδ T cells. D, metformin and curcumin increase effector CD8+ T-cell populations and resulting memory cells, both of which are critical for an effective adaptive immune response. *, Only preclinical data exists to support concept at this time.

Figure 2.

Cancer chemoprevention agents stimulate the adaptive immune system. A, curcumin administration can enhance the TH1 and decrease TH2 immune response. B, metformin may increase MHC-I expression on tumor cells, increasing visibility to effector CD8+ T cells. C, ZA and other bisphosphonates increase phosphoantigens in PBMCs and on cancer cells themselves, resulting in activation of anti-tumor γδ T cells. D, metformin and curcumin increase effector CD8+ T-cell populations and resulting memory cells, both of which are critical for an effective adaptive immune response. *, Only preclinical data exists to support concept at this time.

Close modal

After antigenic stimulation, CD8+ T cells undergo expansion of antigen-specific effector populations followed by persistence of long-lived central and effector TM cells. The number of tumor-infiltrating central and effector TM cells is inversely correlated with tumor invasion including vascular emboli, lymphatic invasion, and perineural invasion (23). In addition, the presence of TM cells is associated with reduced risk of tumor recurrence, suggesting that these cells are important for secondary prevention (23, 24). The antidiabetic drug metformin has been suggested as a chemoprevention agent that may enhance the generation of the TM-cell compartment (Fig. 2B). Preclinical and epidemiologic studies have suggested a cancer prevention role of metformin with a recent meta-analysis showing a 30% overall reduction in cancer incidence in diabetics on metformin compared to other diabetics (25–27). Metformin significantly decreased both aberrant crypt foci and proliferating cell nuclear antigen index over a single month compared to controls in a small, randomized pilot study (28). Metformin may enhance TM-cell numbers via modulation of fatty acid metabolism. Preclinical experiments with mice deficient in TNF receptor-associated factor 6 (TRAF-6) showed that, although mice mounted normal effector CD8+ T-cell responses to infections, they had compromised CD8+ TM-cell generation. Microarray data revealed altered expression of genes that regulate fatty acid metabolism with defective AMP-activated kinase (AMPK) activation and mitochondrial fatty acid oxidation. Metformin, which has been shown to promote AMPK activation, when administered restored the ability to generate TM cells (Fig. 2B; ref. 29). Furthermore, metformin promoted survival of CD8+ T cells in wild-type mice, resulting in enhanced generation of TM cells (Fig. 2B).

Effective CD8+ T-cell immunosurveillance is dependent on MHC-I–mediated antigen presentation. Immunohistochemical staining across a spectrum of premalignant and malignant lesions has showed an association between malignant transformation of cells and HLA class I antigen defects, suggesting that loss of class I expression is an early, critical step in selection and outgrowth of malignant lesions (30, 31). Strategies that increase MHC-I expression on transformed cells may restore immunosurveillance and prevent the development of overt malignancy (32). In a preclinical study, metformin increased MHC-I expression on cancer cells (Fig. 2C). Most cancer cells shift from oxidative phosphorylation to glycolysis to generate energy. In vitro studies with leukemic cells showed culture conditions that forced respiration also had the effect of upregulating MHC-I transcription and protein levels at the cell surface, suggesting a link between the bioenergetic signature of cancer cells and their visibility to the immune system (33). In the SKBR3 breast cancer cell line, metformin enhanced oxidative phosphorylation resulting in a 25-fold increase in cell surface–associated MHC-I protein levels (Fig. 2C; ref. 34). Because MHC-I downregulation has been well documented in premalignant lesions, metformin could serve to increase the immunogenicity of preinvasive disease. The retinoid X receptor agonist bexarotene also has the potential to enhance T-cell numbers and function. Bexarotene has been extensively studied in preclinical models and has been shown to be a potent antiproliferative agent (35). Indeed, clinical trials assessing the potential for bexarotene as a chemoprevention agent are ongoing (36). Bexarotene has also been shown to upregulate the expression of high-affinity IL-2 receptor on the surface of immune cells, when cultured together in vitro, potentially allowing an enhanced proliferation when exposed to an environment rich in IL-2 (37). In addition, the use of bexarotene may increase the lifespan of T cells as the agent has been shown to increase BCL2 expression and inhibit the development of apoptosis in T cells (Fig. 2D; ref. 38).

Curcumin is a potential cancer chemoprevention agent currently being tested in clinical trials that may promote a TH1 environment that increases the number of CD4+ and CD8+ T cells. Curcumin inhibits targets important in oncogenesis including COX-2, tumor growth factor β (TGF-β), and indoleamine 2,3-dixoygenase (IDO), which suppresses the adaptive T-cell immune response (39). Preclinical studies have showed chemoprevention effects of curcumin across multiple tumor types (39). Results of a phase IIa trial revealed that curcumin treatment significantly reduced aberrant crypt foci (40). In the ApcMin/+ mouse, curcumin administration reduced colonic tumor formation by approximately 70%. Immunohistochemistry of the mucosal lymphoid population of curcumin-treated mice revealed a 30% increase in CD4+ T cells compared to controls, although these cells were not further characterized (41). In a mouse model of mammary carcinoma, curcumin inhibited tumor growth by (i) reversing tumor-induced depletion of CD4+ and CD8+ T cells (Fig. 2B and C) and potentiating CD8+ T-cell cytotoxicity (Fig. 2C); (ii) restoring memory T-cell (TM) populations to levels comparable to controls (Fig. 2B); and (iii) shifting the cytokine signature from TH2 to TH1 (Fig. 2A; ref. 42).

γδ T cells are T lymphocytes with attributes of both the innate and adaptive immune system and account for 1% to 10% of all peripheral blood T cells. γδ T cells exhibit many qualities of the innate immune system such as the capability of being activated by nonself ligands and phosphoantigens generated by the isoprenoid pathway used by microorganisms or mevalonate pathway in infected or transformed cells. Once activated, γδ T cells can expand, exhibit cytotoxicity in both a MHC-dependent and independent fashion, and release TH1 cytokines that further support the adaptive immune system (43). In mouse models of prostate and carcinogen-induced cutaneous malignancy, mice lacking γδ T cells developed higher disease burdens and progression of premalignant lesions to overt malignancy than controls and adoptive transfer of γδ cells could abrogate this effect (44, 45). In a longitudinal case–control study of renal transplant patients, 18 patients who developed cancer 2 to 6 years after transplantation were compared to a control group of 45 transplant recipients with similar demographics. Interestingly, patients who developed cancer had significantly fewer γδ T cells (<4%) measured in blood at 6, 12, and 18 months before their diagnosis of cancer compared to control patients (46). These findings have increased interest in the potential utility of γδ T cells in cancer prevention.

Nitrogen-bisphosphonates (N-BP) such as zoledronic acid are being studied as chemoprevention agents that may stimulate the proliferation of γδ T cells (Fig. 2E; ref. 47). N-BPs are primarily used for osteoporosis therapy and to reduce skeletal-related events in patients with bone metastases in solid tumors (48). Preclinical evidence for a cancer preventative effect was seen with the bisphosphonate ibadronate, which reduced colorectal dysplasia induced in an experimental mouse model of ulcerative colitis (49). Multiple observational studies have suggested that bisphosphonates are associated with reductions in the incidence of both breast and colon cancer (50, 51). N-BPs stimulate γδ T cells indirectly by increasing concentrations of isopentenyl pyrophosphate (IPP), a precursor in the mevalonate pathway, in peripheral blood mononuclear cells, which subsequently activates γδ T-cell receptors (TCR). There is also evidence that N-BPs can increase IPP in tumor cells themselves, resulting in a chemotactic and stimulatory signal for γδ T cells (Fig. 2E; ref. 52). Activated γδ T cells release TH1 cytokines such as TNF-α, IL-6, and IFN-γ that are important in immune surveillance (53, 54). Both preclinical data and phase I studies have shown N-BPs can activate tumoricidal γδ T cells in a broad range of tumors including breast, prostate, and renal cell carcinoma (55–58).

Cancer chemoprevention agents may inhibit the function of immune suppressor cells

Foxp3 regulatory T cells (Tregs) constitute 5% of all peripheral CD4+ T cells in healthy adults and are important in the regulation of immune responses to both self and foreign antigens and maintenance of immune homeostasis. Tumor-infiltrating Tregs have been shown to correlate with poor prognosis across a spectrum of different cancers. Studies have shown that Tregs suppress both proliferation and activity of effector T cells. In cancer prevention, the strongest support for an important role of Tregs comes from preclinical rodent studies with carcinogen-induced tumors. In studies with methylcholanthrene-induced fibrosarcomas, depletion of 50% to 70% the total number of Tregs with specific antibodies prevented fibrosarcoma development compared to control mice, suggesting that Tregs interfere with immune surveillance (59, 60). Modulation of Tregs could be a means of cancer prevention.

PGE2 increases the inhibitory potential of Tregs and there is evidence that COX-2 inhibitors reverse this effect (Fig. 3A). In the cancer prevention setting, a mouse model of azoxymethane-induced colon cancer was studied to assess effects of PGE2 reduction. Mice underwent genetic deletion of mPGES-1, an inducible terminal synthase that produces PGE2, resulting in 40% reduction of premalignant aberrant crypt foci in the distant colon; 85% suppression of tumor number; and a 90% reduction in tumor load. Evaluation of colon histology of mPGES-1 knockout mice revealed presence of macroscopically inflamed mesenteric lymph nodes with markedly elevated CD4+ and CD8+ lymphocytes and 55% reduction of CD4+ Foxp3+ cells (61). Further support comes from the cancer literature where multiple studies have showed that PGE2 enhances the ability of Tregs to suppress effector T-cell proliferation and that COX-2 inhibition abrogated this effect (62, 63). Curcumin has also been shown to have similar effects, preventing cancer-induced Treg expansion and reducing TH2 cytokine release (Fig. 3B; ref. 42). These findings suggest that both COX-2 inhibition and curcumin may decrease Treg function and contribute to enhanced immune surveillance (Fig. 3A and B).

Figure 3.

Cancer chemoprevention agents inhibit the function of immune suppressor cells. A, PGE2 can increase immunosuppressive Treg, MDSC, and M2 macrophage populations. COX inhibitors aspirin, celecoxib, and meloxicam can reverse this effect. B, ZA decreases populations of M2 macrophages and may repolarize them to the antitumor M1 phenotype. C the retinoid ATRA can differentiate MDSCs into immature DCs, which may account for its ability to enhance proliferation of both effector and memory CD8+ T cells. Curcumin can inhibit MDSCs and differentiate them toward a M1-like phenotype. D bexarotene inhibits apoptosis in T cells by increasing expression of BCL2. E, both the AI letrozole and curcumin have been shown to reduce Treg populations. *, Only preclinical data exists to support concept at this time.

Figure 3.

Cancer chemoprevention agents inhibit the function of immune suppressor cells. A, PGE2 can increase immunosuppressive Treg, MDSC, and M2 macrophage populations. COX inhibitors aspirin, celecoxib, and meloxicam can reverse this effect. B, ZA decreases populations of M2 macrophages and may repolarize them to the antitumor M1 phenotype. C the retinoid ATRA can differentiate MDSCs into immature DCs, which may account for its ability to enhance proliferation of both effector and memory CD8+ T cells. Curcumin can inhibit MDSCs and differentiate them toward a M1-like phenotype. D bexarotene inhibits apoptosis in T cells by increasing expression of BCL2. E, both the AI letrozole and curcumin have been shown to reduce Treg populations. *, Only preclinical data exists to support concept at this time.

Close modal

Aromatase inhibitors are breast cancer chemoprevention agents associated with up to a 65% relative reduction in annual incidence of invasive breast cancer (64). Aromatase inhibitors may mediate this effect in part by decreasing Treg populations (Fig. 3B; ref. 47). Aromatase inhibitors function by decreasing the peripheral conversion of androgenic precursors into estrogen. Estrogen has been shown to promote a TH2 cytokine profile and expand Tregs, raising the possibility that aromatase inhibitors could shift this balance to TH1 and resolution of aberrant lesions. Consistent with this observation, in a preclinical mouse study of inflammatory arthritis, aromatase inhibitor treatment resulted in a lower percentage of splenic and lymph node Tregs and increased TH1-cytokine release in response to lymphocyte stimulation compared to untreated mice (Fig. 3B; ref. 65). In a randomized phase II trial, patients with locally advanced ER+ breast cancer received the aromatase inhibitor letrozole or letrozole plus cyclophosphamide. There was a significant reduction in Treg number for all patients after treatment with a nonsignificant trend toward the letrozole and cyclophosphamide arm and Treg number at residual histology showed a significant, inverse relationship with response (Fig. 3B; ref. 66).

MDSCs are regulatory cells that suppress tumor immune surveillance. In healthy individuals, these immature myeloid cells generally differentiate into mature cells of the myeloid lineage. In cancer patients, MDSCs accumulate and can contribute to oncogenesis through inhibition of type I immunity (67). MDSCs can inhibit IL-2 production by activated intratumoral T cells as well as activation and proliferation of CD4+ and CD8+ T cells. In addition, MDSCs have the capacity to stimulate Treg recruitment and proliferation. MDSCs act via the depletion of environmental arginine (Arg), an essential amino acid for T-cell function. MDSC stimulate the secretion of inducible nitric oxide synthase (iNOS) and the reactive oxygen species, which promote mutagenesis and inhibit T cells (Fig. 3C; ref. 67). Studies modulating MDSC through inhibition of function or selective depletion have resulted in prevention of carcinogen-mediated neoplasia and restoration of immune surveillance (68, 69).

PGE2 influences differentiation of monocytes, promoting the MDSC phenotype, and COX-2 inhibition may prevent this effect (Fig. 1B). In preclinical studies, PGE2 shifted development of monocytes from immature dendritic cells to MDSCs when added to a standard preparative regimen of GM-CSF and IL-4 (Fig. 1B; ref. 70). In addition, PGE2 exposure promoted COX-2 expression in MDSCs, suggesting that PGE2 initiates a COX-2–mediated positive feedback loop, perpetuating the immunosuppressive signal (71). PGE2 has also been shown to be responsible for chemotaxis of MDSCs to tumor sites through chemokine induction and COX-2 inhibition reversed this effect (72). In a carcinogen induced mouse model of intestinal cancer, the COX-2 inhibitor celecoxib administration delayed tumor development and reduced number of tumors at autopsy compared to controls. Coinciding with this, there was a significant reduction in tumor-infiltrating MDSCs and splenic MDSCs and a decrease in NOS and Arg mRNA levels from splenic cells (73). In a mouse model of glioma prevention, treatment with aspirin or celecoxib reduced systemic PGE2 production, MDSC number, and consequently significantly delayed glioma development (74).

Retinoids are promising chemoprevention agents that may function by redirecting development of MDSCs to immature dendritic cells (Fig. 3C). Initial studies suggested that retinoids were important in myeloid development, as vitamin A deficient mice had significant myeloid expansion in bone marrow, spleen, and peripheral blood, and this effect was reversed with introduction of vitamin A (75). In prevention, 13-cis-retinoic acid decreased leukoplakia lesion size in 67% of patients compared to 10% in placebo and reversed dysplasia in 54% of patients compared to 10% in the placebo group (76). In vitro studies using MDSCs isolated from patients with a variety of solid tumors showed that all-trans retinoic acid (ATRA) could reverse their suppressive effects on CD8+ T cells (Fig. 3C). ATRA was shown to mediate this effect by redifferentiating MDSCs into immature dendritic cells (77–79). In a therapeutic vaccine trial, a recombinant HPV protein vaccine inhibited HPV-related tumor growth by 85% when combined with ATRA compared to 42% with the vaccine alone and this coincided with a significant decrease in the number of MDSCs, an increase in mature dendritic cells, and enhanced HPV-specific CD8+ T-cell response (80). In addition to reducing MDSCs, there is evidence that ATRA can enhance observed proliferation of effector CD8+ T cells by increasing IL-2 release and can augment TM cells when given in combination with a viral vaccine (81). These studies suggest retinoids can reduce suppressive MDSCs and, as a consequence, enhance proliferation and effector function of CD8+ T cells (Fig. 3C). Curcumin has been shown to have similar effects, reducing percentages of MDSCs in peripheral tissues of mice and may actually repolarize them toward a type 1 (M1) macrophage phenotype (Fig. 3C; ref. 82).

Tumor-associated macrophages (TAM) are recruited early in dysplastic or premalignant lesions and contribute significantly to oncogenesis (83–86). M1 macrophages are tumoricidal whereas type 2 (M2) macrophages support tumorigenesis and immunosuppression. M2 macrophages express a host of tumorigenic factors including VEGF, COX-2, epidermal growth factor receptor, and matrix metalloproteinases (MMP), which support angiogenesis, tissue repair, and remodeling in tumors (Fig. 3D). M2 macrophages also induce immunosuppression through elaboration of cytokines such as IL-10 and TGF-β and inhibit T-cell proliferation through expression of Arg and IDO (Fig. 3D; ref. 87). The presence of M2 macrophages correlates with poor prognosis in a number of different tumor types (88). There is evidence that TAMs are recruited early in preneoplasia (89). In a transgenic mouse of mammary cancer (PyMT mice), mice progress through 4 stages from benign hyperplasia to overt malignancy with metastases. There was a significant correlation between low density of macrophages in primary tumors and a delay in vascular development and malignant transition. Macrophage depletion resulted in delayed progression of premalignant lesions (90).

In addition to their other antitumor effects, COX-2 inhibitors inhibit M2 macrophage accumulation (Fig. 3A). PGE2 overexpression increases M2 macrophage density in models of dysplastic and premalignant gastric, esophageal, and colon lesions, which is associated with progression to overt malignancy (83–85). In a rodent model of prevention, surgically induced duodenal reflux resulted in inflammation-induced dysplasia that progressed to squamous cell carcinoma in the forestomach. In control mice, 90% developed dysplasia and 38% of mice developed squamous cell carcinoma (SCC) at week 60 compared to 20% and 0%, respectively, in mice given the COX-2 inhibitor meloxicam. COX-2 was predominantly detected in infiltrating macrophages, suggesting that these cells mediated inflammation-induced COX-2 expression and oncogenesis (91). In another study, ApcMin/+ mice developed premalignant polyps heavily infiltrated by M2 macrophages with a TH2 cytokine profile. Celecoxib administration for 8 weeks (i) reduced the size and number of polyps compared to controls; (ii) shifted the TAM phenotype from M2 to M1 macrophage predominant infiltrate; and (iii) increased the TH1 signature in the environment (Fig. 3A; ref. 92).

Bisphosphonates have been shown to have inhibitory effects on TAMs as well, which is not unexpected given the shared lineage between macrophages and osteoclasts (Fig. 3D). Macrophages endocytose bisphosphonates and release them into the cytosol where they can induce apoptosis by inhibiting the mevalonate pathway (93). Bisphosphosphonates decrease release of protumorigenic factors such as MMP-9 by activated macrophages and decrease other proangiogenic factors such as VEGF associated with activated macrophages (Fig. 3D; ref. 93). In the transgenic BALB-neuT mouse, mice develop spontaneous mammary tumors in a stepwise manner similar to human breast cancer. In one study, mice received either control drug or cycles of zoledronic acid mimicking standard dosing schedules. Mice treated with zoledronic acid had significant extension of median tumor-free survival, delayed growth kinetics, and overall survival compared to control. Tumor stroma of control mice exhibited heavy infiltration of VEGF-expressing macrophages, whereas zoledronic acid–treated mice had a marked reduction. Interestingly, macrophages from control mice exhibited a TH2 cytokine pattern, whereas macrophages from zoledronic acid–treated mice were strongly positive for IFN-γ, suggesting that in addition to reducing total macrophage numbers, zoledronic acid repolarized M2 macrophages to M1 type (94). In a murine model expressing HPV-16 genes (HPV/E2 mouse) of cervical carcinogenesis, mice develop cervical intraepithelial neoplasia (CIN) lesions that progress to invasive SCCs. In a trial comparing effect of zoledronic acid on mice with CIN-3 lesions to controls, at 5 months, controls had SCC incidence of 85%, whereas zoledronic acid–treated mice had an incidence of 30%, suggesting a strong preventative effect. In addition to inducing apoptosis of tumor cells themselves, zoledronic acid reduced infiltrating macrophages by 10% and MMP-9 expression by 73% in remaining macrophages relative to control again suggesting a repolarizing effect of the drug (Fig. 3D; ref. 95).

The immune system is a powerful sentinel against cancer with several types of cells surveying the environment and eliminating preinvasive lesions before the development of overt malignancy. For immunocompetent individuals, a hallmark of cancer involves evolution of the mechanisms for the tumor to evade the immune system (96). In the field of immunoprevention, an emerging strategy involves enhancing immune surveillance via pharmacologic means. Tumor vaccines and antibodies hold promise in terms of bolstering the adaptive immune system for tumor prevention. However, many of the most promising chemoprevention agents may exert their effects, in part, by enhancing function of both dendritic cells and effector T cells and decreasing the functional impact of immunosuppressive cells. As the immune effects of chemoprevention agents are further delineated, the exciting possibility of using them in combination to elicit effective tumor immune surveillance will be within reach.

No potential conflicts of interest were disclosed.

Conception and design: E.A. Marzbani, M.L. Disis

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Lu, M.L. Disis

Writing, review, and/or revision of the manuscript: E.A. Marzbani, H. Lu, M.L. Disis

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C. Inatsuka, M.L. Disis

This work was supported for E. Marzbani by T32 5T32CA009515-27 and for M.L. Disis by NCI N01-CN-53300/WA#10,N01-CN-53300 and DOD W81XWH-11-1-0760.

1.
Chang
MH
,
Chen
CJ
,
Lai
MS
,
Hsu
HM
,
Wu
TC
,
Kong
MS
, et al
Universal hepatitis B vaccination in Taiwan and the incidence of hepatocellular carcinoma in children. Taiwan Childhood Hepatoma Study Group
.
N Engl J Med
1997
;
336
:
1855
9
.
2.
Group
FIIS
,
Dillner
J
,
Kjaer
SK
,
Wheeler
CM
,
Sigurdsson
K
,
Iversen
OE
, et al
Four year efficacy of prophylactic human papillomavirus quadrivalent vaccine against low grade cervical, vulvar, and vaginal intraepithelial neoplasia and anogenital warts: randomised controlled trial
.
BMJ
2010
;
341
:
c3493
.
3.
Darnell
RB
,
Posner
JB
. 
Observing the invisible: successful tumor immunity in humans
.
Nat Immunol
2003
;
4
:
201
.
4.
Dougan
M
,
Dranoff
G
. 
Immune therapy for cancer
.
Annu Rev Immunol
2009
;
27
:
83
117
.
5.
Fridman
WH
,
Pages
F
,
Sautes-Fridman
C
,
Galon
J
. 
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
2012
;
12
:
298
306
.
6.
Mlecnik
B
,
Tosolini
M
,
Kirilovsky
A
,
Berger
A
,
Bindea
G
,
Meatchi
T
, et al
Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction
.
J Clin Oncol
2011
;
29
:
610
8
.
7.
Vesely
MD
,
Kershaw
MH
,
Schreiber
RD
,
Smyth
MJ
. 
Natural innate and adaptive immunity to cancer
.
Annu Rev Immunol
2011
;
29
:
235
71
.
8.
Zitvogel
L
,
Apetoh
L
,
Ghiringhelli
F
,
Kroemer
G
. 
Immunological aspects of cancer chemotherapy
.
Nat Rev Immunol
2008
;
8
:
59
73
.
9.
Disis
ML
. 
Immune regulation of cancer
.
J Clin Oncol
2010
;
28
:
4531
8
.
10.
Bertagnolli
MM
,
Eagle
CJ
,
Zauber
AG
,
Redston
M
,
Solomon
SD
,
Kim
K
, et al
Celecoxib for the prevention of sporadic colorectal adenomas
.
N Engl J Med
2006
;
355
:
873
84
.
11.
Cuzick
J
,
Otto
F
,
Baron
JA
,
Brown
PH
,
Burn
J
,
Greenwald
P
, et al
Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: an international consensus statement
.
Lancet Oncol
2009
;
10
:
501
7
.
12.
Force USPST
. 
Routine aspirin or nonsteroidal anti-inflammatory drugs for the primary prevention of colorectal cancer: U.S. Preventive Services Task Force recommendation statement
.
Ann Intern Med
2007
;
146
:
361
4
.
13.
Steinbach
G
,
Lynch
PM
,
Phillips
RK
,
Wallace
MH
,
Hawk
E
,
Gordon
GB
, et al
The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis
.
N Engl J Med
2000
;
342
:
1946
52
.
14.
Thun
MJ
,
Jacobs
EJ
,
Patrono
C
. 
The role of aspirin in cancer prevention
.
Nat Rev Clin Oncol
2012
;
9
:
259
67
.
15.
Mills
EJ
,
Wu
P
,
Alberton
M
,
Kanters
S
,
Lanas
A
,
Lester
R
. 
Low-dose aspirin and cancer mortality: a meta-analysis of randomized trials
.
Am J Med
2012
;
125
:
560
7
.
16.
Rothwell
PM
,
Fowkes
FG
,
Belch
JF
,
Ogawa
H
,
Warlow
CP
,
Meade
TW
. 
Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials
.
Lancet
2011
;
377
:
31
41
.
17.
Rothwell
PM
,
Price
JF
,
Fowkes
FG
,
Zanchetti
A
,
Roncaglioni
MC
,
Tognoni
G
, et al
Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials
.
Lancet
2012
;
379
:
1602
12
.
18.
Eberhart
CE
,
Coffey
RJ
,
Radhika
A
,
Giardiello
FM
,
Ferrenbach
S
,
DuBois
RN
. 
Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas
.
Gastroenterology
1994
;
107
:
1183
8
.
19.
Half
E
,
Tang
XM
,
Gwyn
K
,
Sahin
A
,
Wathen
K
,
Sinicrope
FA
. 
Cyclooxygenase-2 expression in human breast cancers and adjacent ductal carcinoma in situ
.
Cancer Res
2002
;
62
:
1676
81
.
20.
Herfs
M
,
Herman
L
,
Hubert
P
,
Minner
F
,
Arafa
M
,
Roncarati
P
, et al
High expression of PGE2 enzymatic pathways in cervical (pre)neoplastic lesions and functional consequences for antigen-presenting cells
.
Cancer Immunol Immunother
2009
;
58
:
603
14
.
21.
Saba
NF
,
Choi
M
,
Muller
S
,
Shin
HJ
,
Tighiouart
M
,
Papadimitrakopoulou
VA
, et al
Role of cyclooxygenase-2 in tumor progression and survival of head and neck squamous cell carcinoma
.
Cancer Prev Res
2009
;
2
:
823
9
.
22.
Biragyn
A
,
Longo
DL
. 
Neoplastic “Black Ops”: cancer's subversive tactics in overcoming host defenses
.
Semin Cancer Biol
2012
;
22
:
50
9
.
23.
Pages
F
,
Berger
A
,
Camus
M
,
Sanchez-Cabo
F
,
Costes
A
,
Molidor
R
, et al
Effector memory T cells, early metastasis, and survival in colorectal cancer
.
N Engl J Med
2005
;
353
:
2654
66
.
24.
Galon
J
,
Costes
A
,
Sanchez-Cabo
F
,
Kirilovsky
A
,
Mlecnik
B
,
Lagorce-Pages
C
, et al
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome
.
Science
2006
;
313
:
1960
4
.
25.
Bhalla
K
,
Hwang
BJ
,
Dewi
RE
,
Twaddel
W
,
Goloubeva
OG
,
Wong
KK
, et al
Metformin prevents liver tumorigenesis by inhibiting pathways driving hepatic lipogenesis
.
Cancer Prev Res
2012
;
5
:
544
52
.
26.
Decensi
A
,
Puntoni
M
,
Goodwin
P
,
Cazzaniga
M
,
Gennari
A
,
Bonanni
B
, et al
Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis
.
Cancer Prev Res
2010
;
3
:
1451
61
.
27.
Memmott
RM
,
Mercado
JR
,
Maier
CR
,
Kawabata
S
,
Fox
SD
,
Dennis
PA
. 
Metformin prevents tobacco carcinogen-induced lung tumorigenesis
.
Cancer Prev Res
2010
;
3
:
1066
76
.
28.
Hosono
K
,
Endo
H
,
Takahashi
H
,
Sugiyama
M
,
Sakai
E
,
Uchiyama
T
, et al
Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial
.
Cancer Prev Res
2010
;
3
:
1077
83
.
29.
Pearce
EL
,
Walsh
MC
,
Cejas
PJ
,
Harms
GM
,
Shen
H
,
Wang
LS
, et al
Enhancing CD8 T-cell memory by modulating fatty acid metabolism
.
Nature
2009
;
460
:
103
7
.
30.
Chang
CC
,
Ferrone
S
. 
Immune selective pressure and HLA class I antigen defects in malignant lesions
.
Cancer Immunol Immunother
2007
;
56
:
227
36
.
31.
Cromme
FV
,
Meijer
CJ
,
Snijders
PJ
,
Uyterlinde
A
,
Kenemans
P
,
Helmerhorst
T
, et al
Analysis of MHC class I and II expression in relation to presence of HPV genotypes in premalignant and malignant cervical lesions
.
Br J Cancer
1993
;
67
:
1372
80
.
32.
Lampen
MH
,
van Hall
T
. 
Strategies to counteract MHC-I defects in tumors
.
Curr Opin Immunol
2011
;
23
:
293
8
.
33.
Charni
S
,
de Bettignies
G
,
Rathore
MG
,
Aguilo
JI
,
van den Elsen
PJ
,
Haouzi
D
, et al
Oxidative phosphorylation induces de novo expression of the MHC class I in tumor cells through the ERK5 pathway
.
J Immunol
2010
;
185
:
3498
503
.
34.
Oliveras-Ferraros
C
,
Cufi
S
,
Vazquez-Martin
A
,
Menendez
OJ
,
Bosch-Barrera
J
,
Martin-Castillo
B
, et al
Metformin rescues cell surface major histocompatibility complex class I (MHC-I) deficiency caused by oncogenic transformation
.
Cell Cycle
2012
;
11
:
865
70
.
35.
Li
Y
,
Zhang
Y
,
Hill
J
,
Kim
HT
,
Shen
Q
,
Bissonnette
RP
, et al
The rexinoid, bexarotene, prevents the development of premalignant lesions in MMTV-erbB2 mice
.
Br J Cancer
2008
;
98
:
1380
8
.
36.
Strecker
TE
,
Shen
Q
,
Zhang
Y
,
Hill
JL
,
Li
Y
,
Wang
C
, et al
Effect of lapatinib on the development of estrogen receptor-negative mammary tumors in mice
.
J Natl Cancer Inst
2009
;
101
:
107
13
.
37.
Gorgun
G
,
Foss
F
. 
Immunomodulatory effects of RXR rexinoids: modulation of high-affinity IL-2R expression enhances susceptibility to denileukin diftitox
.
Blood
2002
;
100
:
1399
403
.
38.
Rasooly
R
,
Schuster
GU
,
Gregg
JP
,
Xiao
JH
,
Chandraratna
RA
,
Stephensen
CB
. 
Retinoid x receptor agonists increase bcl2a1 expression and decrease apoptosis of naive T lymphocytes
.
J Immunol
2005
;
175
:
7916
29
.
39.
Aggarwal
BB
,
Kumar
A
,
Bharti
AC
. 
Anticancer potential of curcumin: preclinical and clinical studies
.
Anticancer Res
2003
;
23
:
363
98
.
40.
Carroll
RE
,
Benya
RV
,
Turgeon
DK
,
Vareed
S
,
Neuman
M
,
Rodriguez
L
, et al
Phase IIa clinical trial of curcumin for the prevention of colorectal neoplasia
.
Cancer Prev Res
2011
;
4
:
354
64
.
41.
Churchill
M
,
Chadburn
A
,
Bilinski
RT
,
Bertagnolli
MM
. 
Inhibition of intestinal tumors by curcumin is associated with changes in the intestinal immune cell profile
.
J Surg Res
2000
;
89
:
169
75
.
42.
Bhattacharyya
S
,
Md Sakib Hossain
D
,
Mohanty
S
,
Sankar Sen
G
,
Chattopadhyay
S
,
Banerjee
S
, et al
Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts
.
Cell Mol Immunol
2010
;
7
:
306
15
.
43.
Hannani
D
,
Ma
Y
,
Yamazaki
T
,
Dechanet-Merville
J
,
Kroemer
G
,
Zitvogel
L
. 
Harnessing gammadelta T cells in anticancer immunotherapy
.
Trends Immunol
2012
;
33
:
199
206
.
44.
Girardi
M
,
Oppenheim
DE
,
Steele
CR
,
Lewis
JM
,
Glusac
E
,
Filler
R
, et al
Regulation of cutaneous malignancy by gammadelta T cells
.
Science
2001
;
294
:
605
9
.
45.
Liu
Z
,
Eltoum
IE
,
Guo
B
,
Beck
BH
,
Cloud
GA
,
Lopez
RD
. 
Protective immunosurveillance and therapeutic antitumor activity of gammadelta T cells demonstrated in a mouse model of prostate cancer
.
J Immunol
2008
;
180
:
6044
53
.
46.
Couzi
L
,
Levaillant
Y
,
Jamai
A
,
Pitard
V
,
Lassalle
R
,
Martin
K
, et al
Cytomegalovirus-induced gammadelta T cells associate with reduced cancer risk after kidney transplantation
.
J Am Soc Nephrol
2010
;
21
:
181
8
.
47.
Cuzick
J
. 
Aromatase inhibitors for breast cancer prevention
.
J Clin Oncol
2005
;
23
:
1636
43
.
48.
Gober
HJ
,
Kistowska
M
,
Angman
L
,
Jeno
P
,
Mori
L
,
De Libero
G
. 
Human T cell receptor gammadelta cells recognize endogenous mevalonate metabolites in tumor cells
.
J Exp Med
2003
;
197
:
163
8
.
49.
Sassa
S
,
Okabe
H
,
Nemoto
N
,
Kikuchi
H
,
Kudo
H
,
Sakamoto
S
. 
Ibadronate may prevent colorectal carcinogenesis in mice with ulcerative colitis
.
Anticancer Res
2009
;
29
:
4615
9
.
50.
Rennert
G
,
Pinchev
M
,
Rennert
HS
. 
Use of bisphosphonates and risk of postmenopausal breast cancer
.
J Clin Oncol
2010
;
28
:
3577
81
.
51.
Rennert
G
,
Pinchev
M
,
Rennert
HS
,
Gruber
SB
. 
Use of bisphosphonates and reduced risk of colorectal cancer
.
J Clin Oncol
2011
;
29
:
1146
50
.
52.
Benzaid
I
,
Monkkonen
H
,
Stresing
V
,
Bonnelye
E
,
Green
J
,
Monkkonen
J
, et al
High phosphoantigen levels in bisphosphonate-treated human breast tumors promote Vgamma9Vdelta2 T-cell chemotaxis and cytotoxicity in vivo
.
Cancer Res
2011
;
71
:
4562
72
.
53.
Kunzmann
V
,
Bauer
E
,
Feurle
J
,
Weissinger
F
,
Tony
HP
,
Wilhelm
M
. 
Stimulation of gammadelta T cells by aminobisphosphonates and induction of antiplasma cell activity in multiple myeloma
.
Blood
2000
;
96
:
384
92
.
54.
Kunzmann
V
,
Bauer
E
,
Wilhelm
M
. 
Gamma/delta T-cell stimulation by pamidronate
.
N Engl J Med
1999
;
340
:
737
8
.
55.
Dieli
F
,
Gebbia
N
,
Poccia
F
,
Caccamo
N
,
Montesano
C
,
Fulfaro
F
, et al
Induction of gammadelta T-lymphocyte effector functions by bisphosphonate zoledronic acid in cancer patients in vivo
.
Blood
2003
;
102
:
2310
1
.
56.
Dieli
F
,
Vermijlen
D
,
Fulfaro
F
,
Caccamo
N
,
Meraviglia
S
,
Cicero
G
, et al
Targeting human {gamma}delta} T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer
.
Cancer Res
2007
;
67
:
7450
7
.
57.
Marten
A
,
Lilienfeld-Toal
M
,
Buchler
MW
,
Schmidt
J
. 
Zoledronic acid has direct antiproliferative and antimetastatic effect on pancreatic carcinoma cells and acts as an antigen for delta2 gamma/delta T cells
.
J Immunother
2007
;
30
:
370
7
.
58.
Meraviglia
S
,
Eberl
M
,
Vermijlen
D
,
Todaro
M
,
Buccheri
S
,
Cicero
G
, et al
In vivo manipulation of Vgamma9Vdelta2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients
.
Clin Exp Immunol
2010
;
161
:
290
7
.
59.
Betts
G
,
Twohig
J
,
Van den Broek
M
,
Sierro
S
,
Godkin
A
,
Gallimore
A
. 
The impact of regulatory T cells on carcinogen-induced sarcogenesis
.
Br J Cancer
2007
;
96
:
1849
54
.
60.
Teng
MW
,
Ngiow
SF
,
von Scheidt
B
,
McLaughlin
N
,
Sparwasser
T
,
Smyth
MJ
. 
Conditional regulatory T-cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth
.
Cancer Res
2010
;
70
:
7800
9
.
61.
Nakanishi
Y
,
Nakatsuji
M
,
Seno
H
,
Ishizu
S
,
Akitake-Kawano
R
,
Kanda
K
, et al
COX-2 inhibition alters the phenotype of tumor-associated macrophages from M2 to M1 in ApcMin/+mouse polyps
.
Carcinogenesis
2011
;
32
:
1333
9
.
62.
Baratelli
F
,
Lin
Y
,
Zhu
L
,
Yang
SC
,
Heuze-Vourc'h
N
,
Zeng
G
, et al
Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells
.
J Immunol
2005
;
175
:
1483
90
.
63.
Mahic
M
,
Yaqub
S
,
Johansson
CC
,
Tasken
K
,
Aandahl
EM
. 
FOXP3+CD4+CD25+ adaptive regulatory T cells express cyclooxygenase-2 and suppress effector T cells by a prostaglandin E2-dependent mechanism
.
J Immunol
2006
;
177
:
246
54
.
64.
Goss
PE
,
Ingle
JN
,
Ales-Martinez
JE
,
Cheung
AM
,
Chlebowski
RT
,
Wactawski-Wende
J
, et al
Exemestane for breast-cancer prevention in postmenopausal women
.
N Engl J Med
2011
;
364
:
2381
91
.
65.
Wang
J
,
Zhang
Q
,
Jin
S
,
Feng
M
,
Kang
X
,
Zhao
S
, et al
Immoderate inhibition of estrogen by anastrozole enhances the severity of experimental polyarthritis
.
Exp Gerontol
2009
;
44
:
398
405
.
66.
Generali
D
,
Bates
G
,
Berruti
A
,
Brizzi
MP
,
Campo
L
,
Bonardi
S
, et al
Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients
.
Clin Cancer Res
2009
;
15
:
1046
51
.
67.
Ostrand-Rosenberg
S
,
Sinha
P
. 
Myeloid-derived suppressor cells: linking inflammation and cancer
.
J Immunol
2009
;
182
:
4499
506
.
68.
Morales
JK
,
Kmieciak
M
,
Graham
L
,
Feldmesser
M
,
Bear
HD
,
Manjili
MH
. 
Adoptive transfer of HER2/neu-specific T cells expanded with alternating gamma chain cytokines mediate tumor regression when combined with the depletion of myeloid-derived suppressor cells
.
Cancer Immunol Immunother
2009
;
58
:
941
53
.
69.
Terabe
M
,
Matsui
S
,
Park
JM
,
Mamura
M
,
Noben-Trauth
N
,
Donaldson
DD
, et al
Transforming growth factor-beta production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence
.
J Exp Med
2003
;
198
:
1741
52
.
70.
Sinha
P
,
Clements
VK
,
Fulton
AM
,
Ostrand-Rosenberg
S
. 
Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells
.
Cancer Res
2007
;
67
:
4507
13
.
71.
Obermajer
N
,
Muthuswamy
R
,
Lesnock
J
,
Edwards
RP
,
Kalinski
P
. 
Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells
.
Blood
2011
;
118
:
5498
505
.
72.
Obermajer
N
,
Muthuswamy
R
,
Odunsi
K
,
Edwards
RP
,
Kalinski
P
. 
PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment
.
Cancer Res
2011
;
71
:
7463
70
.
73.
Talmadge
JE
,
Hood
KC
,
Zobel
LC
,
Shafer
LR
,
Coles
M
,
Toth
B
. 
Chemoprevention by cyclooxygenase-2 inhibition reduces immature myeloid suppressor cell expansion
.
Int Immunopharmacol
2007
;
7
:
140
51
.
74.
Fujita
M
,
Kohanbash
G
,
Fellows-Mayle
W
,
Hamilton
RL
,
Komohara
Y
,
Decker
SA
, et al
COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells
.
Cancer Res
2011
;
71
:
2664
74
.
75.
Kuwata
T
,
Wang
IM
,
Tamura
T
,
Ponnamperuma
RM
,
Levine
R
,
Holmes
KL
, et al
Vitamin A deficiency in mice causes a systemic expansion of myeloid cells
.
Blood
2000
;
95
:
3349
56
.
76.
Hong
WK
,
Endicott
J
,
Itri
LM
,
Doos
W
,
Batsakis
JG
,
Bell
R
, et al
13-cis-retinoic acid in the treatment of oral leukoplakia
.
N Engl J Med
1986
;
315
:
1501
5
.
77.
Kusmartsev
S
,
Su
Z
,
Heiser
A
,
Dannull
J
,
Eruslanov
E
,
Kubler
H
, et al
Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma
.
Clin Cancer Res
2008
;
14
:
8270
8
.
78.
Mirza
N
,
Fishman
M
,
Fricke
I
,
Dunn
M
,
Neuger
AM
,
Frost
TJ
, et al
All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients
.
Cancer Res
2006
;
66
:
9299
307
.
79.
Nefedova
Y
,
Fishman
M
,
Sherman
S
,
Wang
X
,
Beg
AA
,
Gabrilovich
DI
. 
Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells
.
Cancer Res
2007
;
67
:
11021
8
.
80.
Song
X
,
Ye
D
,
Liu
B
,
Cui
J
,
Zhao
X
,
Yi
L
, et al
Combination of all-trans retinoic acid and a human papillomavirus therapeutic vaccine suppresses the number and function of immature myeloid cells and enhances antitumor immunity
.
Cancer Sci
2009
;
100
:
334
40
.
81.
Tan
X
,
Sande
JL
,
Pufnock
JS
,
Blattman
JN
,
Greenberg
PD
. 
Retinoic acid as a vaccine adjuvant enhances CD8+ T cell response and mucosal protection from viral challenge
.
J Virol
2011
;
85
:
8316
27
.
82.
Tu
SP
,
Jin
H
,
Shi
JD
,
Zhu
LM
,
Suo
Y
,
Lu
G
, et al
Curcumin induces the differentiation of myeloid-derived suppressor cells and inhibits their interaction with cancer cells and related tumor growth
.
Cancer Prev Res
2012
;
5
:
205
15
.
83.
Femia
AP
,
Dolara
P
,
Luceri
C
,
Salvadori
M
,
Caderni
G
. 
Mucin-depleted foci show strong activation of inflammatory markers in 1,2-dimethylhydrazine-induced carcinogenesis and are promoted by the inflammatory agent sodium dextran sulfate
.
Int J Cancer
2009
;
125
:
541
7
.
84.
Oshima
H
,
Matsunaga
A
,
Fujimura
T
,
Tsukamoto
T
,
Taketo
MM
,
Oshima
M
. 
Carcinogenesis in mouse stomach by simultaneous activation of the Wnt signaling and prostaglandin E2 pathway
.
Gastroenterology
2006
;
131
:
1086
95
.
85.
Oshima
H
,
Oshima
M
,
Inaba
K
,
Taketo
MM
. 
Hyperplastic gastric tumors induced by activated macrophages in COX-2/mPGES-1 transgenic mice
.
EMBO J
2004
;
23
:
1669
78
.
86.
Schmid
MC
,
Avraamides
CJ
,
Foubert
P
,
Shaked
Y
,
Kang
SW
,
Kerbel
RS
, et al
Combined blockade of integrin-alpha4beta1 plus cytokines SDF-1alpha or IL-1beta potently inhibits tumor inflammation and growth
.
Cancer Res
2011
;
71
:
6965
75
.
87.
Mancino
A
,
Lawrence
T
. 
Nuclear factor-kappaB and tumor-associated macrophages
.
Clin Cancer Res
2010
;
16
:
784
9
.
88.
Lewis
CE
,
Pollard
JW
. 
Distinct role of macrophages in different tumor microenvironments
.
Cancer Res
2006
;
66
:
605
12
.
89.
Kawsar
HI
,
Weinberg
A
,
Hirsch
SA
,
Venizelos
A
,
Howell
S
,
Jiang
B
, et al
Overexpression of human beta-defensin-3 in oral dysplasia: potential role in macrophage trafficking
.
Oral Oncol
2009
;
45
:
696
702
.
90.
Lin
EY
,
Li
JF
,
Gnatovskiy
L
,
Deng
Y
,
Zhu
L
,
Grzesik
DA
, et al
Macrophages regulate the angiogenic switch in a mouse model of breast cancer
.
Cancer Res
2006
;
66
:
11238
46
.
91.
Oba
M
,
Miwa
K
,
Fujimura
T
,
Harada
S
,
Sasaki
S
,
Oyama
K
, et al
A selective cyclooxygenase-2 inhibitor prevents inflammation-related squamous cell carcinogenesis of the forestomach via duodenogastric reflux in rats
.
Cancer
2009
;
115
:
454
64
.
92.
Nakanishi
M
,
Menoret
A
,
Tanaka
T
,
Miyamoto
S
,
Montrose
DC
,
Vella
AT
, et al
Selective PGE(2) suppression inhibits colon carcinogenesis and modifies local mucosal immunity
.
Cancer Prev Res
2011
;
4
:
1198
208
.
93.
Rogers
TL
,
Holen
I
. 
Tumour macrophages as potential targets of bisphosphonates
.
J Transl Med
2011
;
9
:
177
.
94.
Coscia
M
,
Quaglino
E
,
Iezzi
M
,
Curcio
C
,
Pantaleoni
F
,
Riganti
C
, et al
Zoledronic acid repolarizes tumour-associated macrophages and inhibits mammary carcinogenesis by targeting the mevalonate pathway
.
J Cell Mol Med
2010
;
14
:
2803
15
.
95.
Giraudo
E
,
Inoue
M
,
Hanahan
D
. 
An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis
.
J Clin Invest
2004
;
114
:
623
33
.
96.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.