Skip to main content
Integrative Cancer Therapies logoLink to Integrative Cancer Therapies
. 2017 Mar 20;17(2):210–216. doi: 10.1177/1534735416681639

Potential Contributions of Antioxidants to Cancer Therapy: Immunomodulation and Radiosensitization

Anita Thyagarajan 1, Ravi P Sahu 1,
PMCID: PMC6041931  PMID: 28627256

Abstract

Antioxidants play important roles in the maintenance of cellular integrity and thus are critical in maintaining the homeostasis of the host immune system. A balance between the levels of pro-oxidants and antioxidants defines the cellular fate of genomic integrity via maintaining the redox status of the cells. An aberration in this balance modulates host immunity that affects normal cellular signaling pathways resulting in uncontrolled proliferation of cells leading to neocarcinogenesis. For decades, there have been scientific debates on the use of antioxidants for the treatment of human cancers. This review is focused on current updates on the implications of antioxidant use as adjuncts in cancer therapy with an emphasis on immunomodulation and radiosensitization.

Keywords: antioxidants, cancer therapy, immunosuppression, oxidized glycerophosphocholines, platelet-activating factor–receptor, immunomodulation, radiosensitization

Introduction

The involvement of the reactive oxygen species (ROS) is well documented in various disease processes, including cancers.1-3 Under normal circumstances, the redox status of cells is maintained by a balance between ROS production and its sequestration by antioxidants.1 While ROS is used as an innate mechanism of host immunity to fight against extracellular pathogens, including bacterial and viral infections, an exacerbated generation causes imbalance in cellular redox potential leading to alterations in signaling pathways and neocarcinogenesis.1-8 ROS release can be affected by several cellular compartments.9,10 As changes in the mitochondrial electron transport activity result in the production of ROS (mROS), heteroplasmic mutations in the mitochondrial DNA have been shown to increase the tumorigenicity of cancer cells via overproduction of mROS.8,11,12 Several cellular antioxidant systems, including superoxide dismutase and thioredoxin play crucial roles in counteracting the damaging effects of increased ROS.10,13 Cancer cells, particularly in the tumor microenvironment exhibit higher basal oxidative stress compared to normal cells and thus take advantage of the upregulated antioxidant system to circumvent ROS-mediated tumor cell damage.10,14,15

Antioxidants and Immunity

The development of cancer has been linked to an inability of the host immune system to respond appropriately to tumor antigens, which leads to tumor immune evasion.16,17 The recognition and eradication of cancer cells by the immune system are categorized as elimination, equilibrium, and escape phases, referred to as the 3Es of immunoediting, which are governed by various factors.18-21 Briefly, in elimination phase, the host immune cells via the surveillance process recognize and try to eliminate nascent tumors.21 Whereas the equilibrium phase starts when a few tumor cells become resistant enough to sustain immune surveillance mechanisms and enter into the dormant stage, where equilibrium exists between tumor cell proliferation and immune cell–mediated apoptosis.21,22 An escape phase is established when tumor cells override their destruction by the immune system, an immunosuppressive environment is maintained, which favors tumor cell proliferation, and the development of clinically detectable tumors is finally achieved.21,23 A direct link between antioxidants and modulation of cancer immunoediting has not been fully established. Nevertheless, curcumin has been shown to modulate immunoediting processes including resurrecting immune surveillance mechanisms to help eradicate cancer cells via (a) restoration of CD4+/CD8+ T cells, (b) suppression of T cell apoptosis, and (c) inhibition of immunosuppression through attenuation of immunosuppressive regulatory T cells (Tregs) (Table 1).24-28 Curcumin is a yellow-colored curcuminoid of turmeric of the family Zingiberaceae that is used as an herbal supplement, food flavoring and food coloring agent and is known to possess medicinal and anticarcinogenic properties that are attributed to its antioxidant and anti-inflammatory activities.29-31 In addition, the antioxidant ascorbic acid (vitamin C) has been demonstrated to play an important role in stimulating the immune system via attenuating chronic inflammatory responses, the persistence of which is implicated in the etiology of various diseases, including cancer.32 Of note, chronic inflammation in tumors mediated via tumor infiltrating leucocytes (TIL) has been shown to induce immunosuppression and promote cancer growth.33

Table 1.

Effect of Antioxidants on Host Antitumor Immunity: Evidences From Preclinical Studies.

Treatment Experimental Model Results References
Curcumin Swiss albino mice with Ehrlich ascites carcinoma cells Restoration of tumor-induced depletion of host CD4+/CD8+ T cell proliferation, inhibition of thymocytes and splenocytes apoptosis, expansion of central memory and effector memory T cells and inhibition of suppressive activity of Tregs 27, 28
Resveratrol BALB/c mice with 4T1 breast carcinoma cells Inactivation of Stat3, prevention of tumor-evoked regulatory B cells (tBregs) generation and function and restricting the tBreg-induced conversion of FoxP3+ Tregs 34
Resveratrol C57BL/6 and BABL/c mice with EG6 lymphoma and CT26 colon carcinoma cell lines Suppression of tumor-derived Tregs 35
Fish oil and selenium yeast BALB/cByJ mice with Line-1 and YAC-1 lymphoma cells Decreased populations of immunosuppressive Tregs and myeloid-derived suppressor cells 36

Immunosuppression (ie, inhibition of the host immune system) via diverse environmental agents (such as ultraviolet radiation) acting on the skin is classically divided into local (localized to the site of application) and systemic immunosuppression.37 Of importance, systemic immunosuppression has been implicated in human malignancies such as melanoma and glioblastoma.38-40 In most cancer models, tumor-dependent immune suppression has been recognized as one of the major events in promoting immune evasion of malignant cells from the host’s antitumor immunity.41-44 While tumor antigen-induced effector T-cells are required in inducing antitumor immunity, suppressor cells such as Tregs and myeloid-derived suppressor cells (MDSCs) promote tumor escape mechanisms.41-47 Importantly, immune checkpoint inhibitors such as cytotoxic T lymphocyte associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) have been shown to mediate Tregs-induced immunosuppression and promote cancer growth. The blockade of these immune checkpoint inhibitors via anti-CTLA-4 and anti-PD-1 antibodies attenuates tumor growth as well as augments the effectiveness of cancer therapy approaches.47-50 Notably, Tregs and MDSCs have been shown to be associated with poor prognosis and cancer treatment failure, including immunotherapy and vaccination approaches.51-53 Therefore, agents/anti-cancer drugs that target these immunosuppressive cell types are being explored in several preclinical and clinical cancer models and also in combination with known anti-cancer drugs against various malignancies.54-57 With regard to antioxidant use in attenuating the enhanced tumor growth mediated by these immunosuppressive cells, studies by Lee-Chang et al34 have shown that administration of resveratrol, a dietary polyphenol compound possessing antioxidant properties at low doses that are nontoxic to immune cells, inhibits lung metastasis of breast cancer tumor. This resveratrol-induced effect was mediated via Stat3 inactivation, resulting in an inhibition of the generation and function of tumor-evoked regulatory B cells (tBregs) and their conversion into Tregs. In support of these findings, studies by Yang et al35 have demonstrated that suppression of tumor-derived Tregs mediates resveratrol-induced inhibition of syngeneic murine EG7 lymphoma and CT26 colon carcinoma tumors in C57BL/6 and BALB/c mice models.35 Of importance, resveratrol can exert both antioxidant and pro-oxidant properties depending on its concentration and cell types used.58 Along similar lines, Wang et al36 have demonstrated that a combination of fish oil and selenium that possesses anti-inflammatory and antioxidant activities exerted synergistic effects in suppressing lung tumor growth mediated via decreasing the population of splenic Tregs and MDSCs and thus augmented host anti-tumor immunity against lung carcinoma.

Exposure to pro-oxidative stressors generating ROS, including ultraviolet B (UVB) radiation and cigarette smoke (CS), is associated with the modulation of host immunity. Studies including ours have demonstrated the beneficial effects of antioxidant administration, including vitamin C and N-acetyl cysteine (NAC) in attenuating the systemic immunosuppressive effects of various environmental pro-oxidative agents, including UVB and CS in animal models.59-65 Of significance, UVB and CS generate a class of oxidized lipid mediators nonenzymatically via free radical mediated attack on membrane phospholipid glycerophosphocholines (ox-GPCs).59-65 In contrast, enzymatic synthesis of platelet-activating factor (PAF, 1-hexadecyl-2-acetyl-glycerophosphocholine) is a tightly regulated process, which requires cytoplasmic phospholipase A2 (cPLA2) that acts on 1-alkyl glycerophosphocholines (GPC) with sn-2 long-chained unsaturated fatty acids (eg, arachidonate) forming the lyso species which then is acetylated, forming PAF.66,67 These ox-GPCs possess PAF and PAF-like agonist activities, which bind to and activate a 7-transmembrane G-protein coupled receptor, the PAF-receptor (PAF-R) which is expressed on various cell types (immune and nonimmune) including keratinocytes and tumor cells.59-68 Our group uses 2 functional assays to quantify total PAF-agonists by very specific interleukin 8 (IL-8) protein or Ca2+ mobilization using stably PAF-R expressing (KBP) and deficient (KBM) cells generated retrovirally from human epidermal KB cells.59-62,65,69-71 The activity of Ox-GPCs/PAF is regulated by PAF-acetyl hydrolases (PAF-AH; PLA2G7).66-68 Of 3 types, PAF-AHI is the major plasma isoform. PAF-R activation mediates various biological activities including early pro-inflammatory and delayed systemic immunosuppressive effects.59-64 Our research group and others have shown that ox-GPCs/PAF-R agonists mediate UVB- and CS-induced systemic immunosuppression in a PAF-R dependent manner which is measured by inhibition of contact hypersensitivity (CHS) or delayed type hypersensitivity (DTH) responses to an eliciting allergen, dinitrofluorobenzene (DNFB) or an antigen, Candida albicans.59-64 In using this methodology, for example, to measure UVB-mediated systemic immunosuppression, the shaved dorsal back skin of mice were exposed to UVB. A group of mice injected intraperitoneally with either PBS or the PAF-R agonist, CPAF served as negative and positive controls. Five days later, a 2x2 cm area of the back skin (approximately 2.5 cm distant from the UVB-radiated site) was sensitized with 0.5% DNFB topically. After 9 days, postelicitation changes in CHS were assessed by measuring changes in ear thickness. With similar methodology, the local immunosuppressive effect of UVB is assessed when dorsal skin of mice is sensitized with DNFB onto the UVB-exposed area (a model of local immunosuppression) and usually required lower UVB doses. Using this methodology we have demonstrated that PAF-R does not mediate UVB-induced local immunosuppressive effects,65 despite its effect in mediating systemic immunosuppression. This PAF-R-dependent systemic immunosuppression is mediated via upregulation of COX-2 enzyme and COX-2-generated prostanoids, immunosuppressive cytokine interleukin 10 (IL-10) and the Tregs cell type, in a process blocked by antioxidants.59-64 As both environmental and therapeutic pro-oxidative stressors generate ROS and thus ox-GPCs/PAF-R agonists, we have shown that supplementation of vitamin C and NAC in drinking water prophylactically suppressed generation of ox-GPCs/PAF-R agonists mediated by UVB, chemotherapy, and radiation therapy—as well as diminishing the augmentation of tumor growth induced by systemic immunosuppression.69-71 Several standard chemotherapeutic agents, including dacarbazine and melphalan generate PAF-R agonists from both murine and human melanoma cells in vitro and intratumorally treated melanoma tumor xenografts in vivo.70 Using a dual tumor model, we demonstrated that intratumoral melphalan (MELP) chemotherapy (of one tumor) augments the growth of secondary (untreated) B16F10 melanoma tumor in a PAF-R dependent manner.70 Systemic antioxidants, COX-2 inhibitors or depleting Treg Abs attenuated this MELP-mediated enhanced growth of secondary tumors in WT mice,70 indicating the role of oxidatively generated PAF-R agonists and downstream COX-2 and Tregs in modulating MELP efficacy. Nevertheless, antioxidant use postchemotherapy or to augment cancer therapy effectiveness in preclinical cancer models with regard to immunosuppression requires further investigation.

Antioxidants in Cancer Therapy

Despite recent advances in local and systemic treatment modalities, chemotherapy, radiation therapy and immunotherapy are widely considered either alone or in combinations for a variety of cancers. In chemotherapy, cancer cells are targeted by chemically modified agents/natural compounds with cytotoxic properties; radiation therapy uses high-energy particles/waves, including x-rays and gamma rays, to kill tumor cells; and immunotherapy treatments are designed to stimulate the host’s own immune system to attack cancer cells. One of the consequences of chemotherapy and radiation therapy is the generation of ROS which via its direct and indirect effects on tumor cells, induces DNA damage and/or affects DNA replication machinery, leading to aberrations in several cellular signaling pathways resulting in chemotherapy- or radiation therapy-induced cell death.72-74 Most of these therapies are not considered a good option as a single agent to treat advanced-stage/metastatic cancers, in part due to the development of therapy-induced innate and/or acquired tumor resistance or local/systemic toxicities leading to either reduced response, nonresponsiveness or tumor relapse after an initial antitumor response.75,76 Therefore, potentially new therapeutic approaches with agents that exhibit anticancer properties and can potentiate chemotherapy- or radiation therapy–mediated antitumor responses are required for inducing optimal and long-term benefits in cancer patients.

Several nutritional cancer chemopreventive compounds having antioxidant properties have been documented to potentiate radiation therapy–induced cytotoxic effects on cancer cells while reducing its toxicity on normal surrounding tissues.77-86 In this regard, multiple studies by Raffoul et al78 have shown that phytochemical soy isoflavones (genistein, daidzein, and glycitein), which exhibit anticarcinogenic properties in part via their antioxidant activities, could be used as potent radiosensitizers to enhance the efficacy of radiotherapy-mediated suppression of the growth and metastatic ability of cancers, including prostate cancer.78-80 A study comparing the effects of the soy isoflavone component genistein on prostate cancer demonstrated that both soy and genistein inhibited the growth of in vitro human PC-3 prostate cancer cells and in vivo orthotopic PC-3 tumors and that these effects were enhanced when soy or genistein was combined with radiotherapy.79 Mechanistically, soy isoflavones attenuated the radiation-induced increase in expression of apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) and activation of NF-kB and its DNA-binding activity, resulting in potentiating radiotherapy-mediated effects and their use as radiosensitizers.79 In addition, soy isoflavones have been shown to augment radiation-induced suppression of in vitro growth of human A549 non–small cell lung cancer cells via inducing increased DNA damage, inhibition of APE1/Ref-1 mediated DNA repair and decreased expression of the transcription factors NF-κB and HIF-1α.82 Importantly, agents possessing proteotoxic stress activities have been shown to enhance radiosensitization. In this regard, Pruitt et al83 have demonstrated that dietary polyphenols known as hydroxychalones augmented radiation-mediated death of human colorectal adenocarcinoma HT-29 and pancreatic cancer Panc 1 cells. These effects were mediated via activation of the heat shock factor 1 (Hsf1) in a process blocked by prophylactic treatment with α-napthoflavone (ANF), a specific inhibitor of cytochrome P450 1A2 (CYP1A2).83 Along similar lines, resveratrol and piperine, which possess properties including antitumor activities, have been shown to augment ionizing radiation (IR)-induced apoptosis and loss of mitochondrial membrane potential in murine colon carcinoma CT26 and melanoma B16F10 cells via enhancing IR-induced ROS generation.84 Moreover, pentoxifylline (PTX), a methylxanthine that possesses antioxidant properties is known to improve tumor tissue oxygenation in murine hypoxic tumors as well as inhibiting post radiation-induced normal tissue injury in mice.85-88 Importantly, PTX has been shown to enhance radiation-mediated effects in human breast MCF-7 and colon HT-29 carcinoma cells in vitro and murine mammary adenocarcinoma SCK tumors in A/J mice in-vivo.85-88 In in vitro studies, PTX treatment has been shown to enhance radiotherapy-mediated effects in a dose and time dependent manner in post-irradiated cells, and did not change the cellular response to radiation in pre-irradiated cells.86

Several chemotherapeutic agents used for the treatment of human malignancies generate ROS as one of the potent mechanisms to eradicate tumor cells via targeting multiple oncogenic signaling pathways. Hence, targeting ROS by antioxidants not surprisingly has yielded mixed results in the therapeutic efficacy of chemotherapy.89-91 Importantly, in a systematic review, Block et al92,93 have summarized the impact of antioxidant supplementation on the efficacy and toxicity of chemotherapeutic agents from several randomized controlled trials (RCTs). Antioxidants such as glutathione, melatonin, vitamin A and E, NAC, selenium, l-carnitine, Co-Q10, ellagic acid, an antioxidant mixture such as vitamin C and E with beta-carotene or selenium supplementation were evaluated on the efficacy of chemotherapeutic regimens, including oxaliplatin and cisplatin in combinations with agents such as etoposide, gemcitabine for several malignancies including breast, lung and gastric cancers.90 Survival of patients and tumor response were the primary outcomes of these trials.90 While a statistically significant improved survival rate either at 1 year or 5 years was associated with melatonin supplementation, vitamin A exerted mixed responses on therapy outcomes and no significant differences in the tumor response or survival were reported with other antioxidants.90 Of significance, 24 RCTs reported significantly decreased toxicities with concurrent supplementation of antioxidants (mentioned above) with chemotherapy in cancer patients compared to patients who were not on systemic antioxidants (controls).91 On the other hand, 9 RCTs reported no differences in the toxicities by antioxidants supplementation and 1 RCT with vitamin A reported increased toxicity.91 Along similar lines, a systematic review on RCTs by Yasueda et al94 summarized that it is difficult to determine whether antioxidant supplements affect treatment outcomes or ameliorate adverse effects induced by chemotherapy and radiotherapy. The authors concluded that harm caused by antioxidant supplementation remained unclear for patients during cancer therapy, except for smokers undergoing radiotherapy.94 As cancer patients experience therapy-induced adverse side effects, including weight loss due to low nutritional intake and/or loss of appetite, individualized counsel for the use of antioxidants or supplements with antioxidant properties during treatment is important to circumvent its detrimental effects and/or therapy outcome.

Conclusion

While the safety and benefits of antioxidants use during cancer treatment are limited, their indispensable role in the maintenance of immune system homeostasis cannot be overruled. A large percentage of cancer patients undergoing active treatments uses antioxidants and not all antioxidants are likely to be beneficial; as well, their mode of action on cellular systems and interaction with anticancer drugs remained largely unexplored. Therefore, further preclinical and clinical studies are needed to establish the clinical implications of antioxidant doses and timings based on treatment regimens, disease stage, and especially immune suppression status.

Acknowledgments

The authors thank Dr Jeffrey Travers for critically reading the manuscript and providing inputs.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: RPS acknowledges the support from National Institutes of Health grant K22 ES023850.

References

  • 1. Halliwell B. Reactive oxygen species in living systems: source, biochemistry, and role in human disease. Am J Med. 1991;91(3C):14S-22S. [DOI] [PubMed] [Google Scholar]
  • 2. Sreevalsan S, Safe S. Reactive oxygen species and colorectal cancer. Curr Colorectal Cancer Rep. 2013;9:350-357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Marinescu S, Anghel R, Gruia MI, Beuran M. Involvement of reactive oxygen species in the mechanisms associated with cervical cancer specific treatment. Chirurgia (Bucur). 2014;109:806-811. [PubMed] [Google Scholar]
  • 4. Paiva CN, Bozza MT. Are reactive oxygen species always detrimental to pathogens? Antioxid Redox Signal. 2014;20:1000-1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Bahia AC, Oliveira JH, Kubota MS, et al. The role of reactive oxygen species in Anopheles aquasalis response to Plasmodium vivax infection. PLoS One. 2013;8:e57014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Cui J, Chen Y, Wang HY, Wang RF. Mechanisms and pathways of innate immune activation and regulation in health and cancer. Hum Vaccin Immunother. 2014;10:3270-3285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Acharya A, Das I, Chandhok D, Saha T. Redox regulation in cancer: a double-edged sword with therapeutic potential. Oxid Med Cell Longev. 2010;3:23-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Sullivan LB, Chandel NS. Mitochondrial reactive oxygen species and cancer. Cancer Metab. 2014;2:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Panieri E, Gogvadze V, Norberg E, Venkatesh R, Orrenius S, Zhivotovsky B. Reactive oxygen species generated in different compartments induce cell death, survival, or senescence. Free Radic Biol Med. 2013;57:176-187. [DOI] [PubMed] [Google Scholar]
  • 10. Raninga PV, Trapani GD, Tonissen KF. Cross talk between two antioxidant systems, thioredoxin and DJ-1: consequences for cancer. Oncoscience. 2014;1:95-110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417:1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell. 2012;48:158-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Oyewole AO, Birch-Machin MA. Mitochondria-targeted antioxidants. FASEB J. 2015;29:4766-4771. [DOI] [PubMed] [Google Scholar]
  • 14. Bernardes SS, de Souza-Neto FP, Ramalho LN, et al. Systemic oxidative profile after tumor removal and the tumor microenvironment in melanoma patients. Cancer Lett. 2015;361:226-232. [DOI] [PubMed] [Google Scholar]
  • 15. Satoh H, Moriguchi T, Taguchi K, et al. Nrf2-deficiency creates a responsive microenvironment for metastasis to the lung. Carcinogenesis. 2010;31:1833-1843. [DOI] [PubMed] [Google Scholar]
  • 16. Yan X, Li W, Pan L, et al. Lewis lung cancer cells promote SIGNR1 (CD209b)-mediated macrophages polarization induced by IL-4 to facilitate immune evasion. J Cell Biochem. 2016;117:1158-1166. [DOI] [PubMed] [Google Scholar]
  • 17. Vences-Catalán F, Rajapaksa R, Srivastava MK, et al. Tetraspanin CD81 promotes tumor growth and metastasis by modulating the functions of T regulatory and myeloid-derived suppressor cells. Cancer Res. 2015;75:4517-4526. [DOI] [PubMed] [Google Scholar]
  • 18. Teng MW, Galon J, Fridman WH, Smyth MJ. From mice to humans: developments in cancer immunoediting. J Clin Invest. 2015;125:3338-3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Bhatia A, Kumar Y. Cancer stem cells and tumour immunoediting: putting two and two together. Expert Rev Clin Immunol. 2016;12:605-607. [DOI] [PubMed] [Google Scholar]
  • 20. Jones LM, Broz ML, Ranger JJ, et al. STAT3 establishes an immunosuppressive microenvironment during the early stages of breast carcinogenesis to promote tumor growth and metastasis. Cancer Res. 2016;76:1416-1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565-1570. [DOI] [PubMed] [Google Scholar]
  • 22. Koebel CM, Vermi W, Swann JB, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007;450:903-907. [DOI] [PubMed] [Google Scholar]
  • 23. Quintana E, Shackleton M, Foster HR, et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell. 2010;18:510-523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Bose S, Panda AK, Mukherjee S, Sa G. Curcumin and tumor immune-editing: resurrecting the immune system. Cell Div. 2015;10:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Ranjan D, Johnston TD, Wu G, Elliott L, Bondada S, Nagabhushan M. Curcumin blocks cyclosporine A–resistant CD28 costimulatory pathway of human T-cell proliferation. J Surg Res. 1998;77:174-178. [DOI] [PubMed] [Google Scholar]
  • 26. Jagetia GC, Aggarwal BB. “Spicing up” of the immune system by curcumin. J Clin Immunol. 2007;27:19-35. [DOI] [PubMed] [Google Scholar]
  • 27. Pal S, Bhattacharyya S, Choudhuri T, Datta GK, Das T, Sa G. Amelioration of immune cell number depletion and potentiation of depressed detoxification system of tumor-bearing mice by curcumin. Cancer Detect Prev. 2005;29:470-478. [DOI] [PubMed] [Google Scholar]
  • 28. Bhattacharyya S, Md Sakib Hossain D, Mohanty S, et al. Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts. Cell Mol Immunol. 2010;7:306-315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Sahu PK, Sahu PK, Sahu PL, Agarwal DD. Structure activity relationship, cytotoxicity and evaluation of antioxidant activity of curcumin derivatives. Bioorg Med Chem Lett. 2016;26:1342-1347. [DOI] [PubMed] [Google Scholar]
  • 30. Anuradha BR, Bai YD, Sailaja S, Sudhakar J, Priyanka M, Deepika V. Evaluation of anti-inflammatory effects of curcumin gel as an adjunct to scaling and root planing: a clinical study. J Int Oral Health. 2015;7:90-93. [PMC free article] [PubMed] [Google Scholar]
  • 31. Kumaravel M, Sankar P, Latha P, Benson CS, Rukkumani R. Antiproliferative effects of an analog of curcumin in Hep-2 cells: a comparative study with curcumin. Nat Prod Commun. 2013;8:183-186. [PubMed] [Google Scholar]
  • 32. Sorice A, Guerriero E, Capone F, Colonna G, Castello G, Costantini S. Ascorbic acid: its role in immune system and chronic inflammation diseases. Mini Rev Med Chem. 2014;14:444-452. [DOI] [PubMed] [Google Scholar]
  • 33. Umansky V, Sevko A. Overcoming immunosuppression in the melanoma microenvironment induced by chronic inflammation. Cancer Immunol Immunother. 2012;61:275-282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Lee-Chang C, Bodogai M, Martin-Montalvo A, et al. Inhibition of breast cancer metastasis by resveratrol-mediated inactivation of tumor-evoked regulatory B cells. J Immunol. 2013;191:4141-4151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Yang Y, Paik JH, Cho D, et al. Resveratrol induces the suppression of tumor-derived CD4+ CD25+ regulatory T cells. Int Immunopharmacol. 2008;8:542-547. [DOI] [PubMed] [Google Scholar]
  • 36. Wang H, Chan YL, Li TL, et al. Reduction of splenic immunosuppressive cells and enhancement of anti-tumor immunity by synergy of fish oil and selenium yeast. PLoS One. 2013;8:e52912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Ullrich SE. Mechanisms underlying UV-induced immune suppression. Mutat Res. 2005;571:185-205. [DOI] [PubMed] [Google Scholar]
  • 38. Finlay-Jones JJ, Hart PH. Ultraviolet irradiation, systemic immunosuppression and skin cancer: role of urocanic acid. Australas J Dermatol. 1997;38(suppl 1):S7-S12. [DOI] [PubMed] [Google Scholar]
  • 39. Schoof N, Iles MM, Bishop DT, et al. Genomel Consortium. Pathway-based analysis of a melanoma genome-wide association study: analysis of genes related to tumour-immunosuppression. PLoS One. 2011;6:e29451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Gustafson MP, Lin Y, New KC, et al. Systemic immune suppression in glioblastoma: the interplay between CD14+HLA-DRlo/neg monocytes, tumor factors, and dexamethasone. Neuro Oncol. 2010;12:631-644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol. 2008;181:5791-5802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Nagaraj S, Collazo M, Corzo CA, et al. Regulatory myeloid suppressor cells in health and disease. Cancer Res. 2009;69:7503-7506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Nagaraj S, Gabrilovich DI. Myeloid-derived suppressor cells in human cancer. Cancer J. 2010;16:348-353. [DOI] [PubMed] [Google Scholar]
  • 44. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001;166:5398-5406. [DOI] [PubMed] [Google Scholar]
  • 45. Schlecker E, Stojanovic A, Eisen C, et al. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J Immunol. 2012;189:5602-5611. [DOI] [PubMed] [Google Scholar]
  • 46. Steitz J, Brück J, Lenz J, Knop J, Tüting T. Depletion of CD25+ CD4+ T cells and treatment with tyrosinase-related protein 2-transduced dendritic cells enhance the interferon α-induced, CD8+ T-cell-dependent immune defense of B16 melanoma. Cancer Res. 2001;61:8643-8646. [PubMed] [Google Scholar]
  • 47. Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol. 2009;21:1065-1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. van Elsas A, Sutmuller RP, Hurwitz AA, et al. Elucidating the autoimmune and antitumor effector mechanisms of a treatment based on cytotoxic T lymphocyte antigen-4 blockade in combination with a B16 melanoma vaccine: comparison of prophylaxis and therapy. J Exp Med. 2001;194:481-489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer–preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol. 2012;37:430-439. [DOI] [PubMed] [Google Scholar]
  • 50. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2012;107:4275-4280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Flammiger A, Weisbach L, Huland H, et al. High tissue density of FOXP3+ T cells is associated with clinical outcome in prostate cancer. Eur J Cancer. 2013;49:1273-1279. [DOI] [PubMed] [Google Scholar]
  • 52. Ye L, Li X, Sun S, et al. A study of circulating anti-CD25 antibodies in non-small cell lung cancer. Clin Transl Oncol. 2013;15:633-637. [DOI] [PubMed] [Google Scholar]
  • 53. Zhou J, Ding T, Pan W, Zhu LY, Li L, Zheng L. Increased intratumoral regulatory T cells are related to intratumoral macrophages and poor prognosis in hepatocellular carcinoma patients. Int J Cancer. 2009;125:1640-1648. [DOI] [PubMed] [Google Scholar]
  • 54. Curiel TJ. Regulatory T cells and treatment of cancer. Curr Opin Immunol. 2008;20:241-246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Son CH, Bae JH, Shin DY, et al. Combination effect of regulatory T-cell depletion and ionizing radiation in mouse models of lung and colon cancer. Int J Radiat Oncol Biol Phys. 2015;92:390-398. [DOI] [PubMed] [Google Scholar]
  • 56. Rossowska J, Pajtasz-Piasecka E, Anger N, et al. Cyclophosphamide and IL-12-transduced DCs enhance the antitumor activity of tumor antigen-stimulated DCs and reduce Tregs and MDSCs number. J Immunother. 2014;37:427-439. [DOI] [PubMed] [Google Scholar]
  • 57. Hu M, Li K, Maskey N, et al. Decreased intratumoral Foxp3 Tregs and increased dendritic cell density by neoadjuvant chemotherapy associated with favorable prognosis in advanced gastric cancer. Int J Clin Exp Pathol. 2014;7:4685-4694. [PMC free article] [PubMed] [Google Scholar]
  • 58. de la Lastra CA, Villegas I. Resveratrol as an antioxidant and pro-oxidant agent: mechanisms and clinical implications. Biochem Soc Trans. 2007;35(pt 5):1156-1160. [DOI] [PubMed] [Google Scholar]
  • 59. Zhang Q, Yao Y, Konger RL, et al. UVB radiation-mediated inhibition of contact hypersensitivity reactions is dependent on the platelet-activating factor system. J Invest Dermatol. 2008;128:1780-1787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Konger RL, Marathe GK, Yao Y, Zhang Q, Travers JB. Oxidized glycerophosphocholines as biologically active mediators for ultraviolet radiation-mediated effects. Prostaglandins Other Lipid Mediat. 2008;87:1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Yao Y, Wolverton JE, Zhang Q, et al. Ultraviolet B radiation generated platelet-activating factor receptor agonist formation involves EGF-R-mediated reactive oxygen species. J Immunol. 2009;182:2842-2848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Sahu RP, Petrache I, Van Demark MJ, et al. Cigarette smoke exposure inhibits contact hypersensitivity via the generation of platelet activating factor agonists. J Immunol. 2013;190:2447-2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Lehr HA, Weyrich AS, Saetzler RK, et al. Vitamin C blocks inflammatory platelet-activating factor mimetics created by cigarette smoking. J Clin Invest. 1997;99:2358-2364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Ullrich SE. Mechanisms underlying UV-induced immune suppression. Mutat Res. 2005;571:185-205. [DOI] [PubMed] [Google Scholar]
  • 65. Sahu RP, Yao Y, Konger RL, Travers JB. Platelet-activating factor does not mediate UVB-induced local immune suppression. Photochem Photobiol. 2012;88:490-493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Braquet P, Touqui L, Shen TY, Vargaftig BB. Perspectives in platelet-activating factor research. Pharmacol Rev. 1987;39:97-145. [PubMed] [Google Scholar]
  • 67. Stafforini DM, McIntyre TM, Zimmerman GA, Prescott SM. Platelet-activating factor, a pleiotrophic mediator of physiological and pathological processes. Crit Rev Clin Lab Sci. 2003;40:643-672. [DOI] [PubMed] [Google Scholar]
  • 68. Travers JB, Huff JC, Rola-Pleszczynski M, Gelfand EW, Morelli JG, Murphy RC. Identification of functional platelet-activating factor receptors on human keratinocytes. J Invest Dermatol. 1995;105:816-823. [DOI] [PubMed] [Google Scholar]
  • 69. Sahu RP, Turner MJ, DaSilva SC, et al. The environmental stressor ultraviolet B radiation inhibits murine antitumor immunity through its ability to generate platelet-activating factor agonists. Carcinogenesis. 2012;33:1360-1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Sahu RP, Ocana JA, Harrison KA, et al. Chemotherapeutic agents subvert tumor immunity by generating agonists of platelet-activating factor. Cancer Res. 2014;74:7069-7078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Sahu RP, Harrison KA, Weyerbacher J, et al. Radiation therapy generates platelet-activating factor agonists. Oncotarget. 2016;7:20788-20800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Glebova K, Veiko N, Kostyuk S, Izhevskaya V, Baranova A. Oxidized extracellular DNA as a stress signal that may modify response to anticancer therapy. Cancer Lett. 2015;356:22-33. [DOI] [PubMed] [Google Scholar]
  • 73. Wang J, Yi J. Cancer cell killing via ROS: to increase or decrease that is the question. Cancer Biol Ther. 2008;7:1875-1884. [DOI] [PubMed] [Google Scholar]
  • 74. Vikram B, Coleman CN, Deye JA. Current status and future potential of advanced technologies in radiation oncology. Part 1. Challenges and resources. Oncology (Williston Park). 2009;23:279-283. [PubMed] [Google Scholar]
  • 75. Shaked Y. Balancing efficacy of and host immune responses to cancer therapy: the yin and yang effects. Nat Rev Clin Oncol. 2016;13:611-626. doi: 10.1038/nrclinonc.2016.57. [DOI] [PubMed] [Google Scholar]
  • 76. Khan S, Karmokar A, Howells L, et al. Targeting cancer stem-like cells using dietary-derived agents—where are we now? Mol Nutr Food Res. 2016;60:1295-1309. [DOI] [PubMed] [Google Scholar]
  • 77. Mut-Salud N, Álvarez PJ, Garrido JM, Carrasco E, Aránega A, Rodríguez-Serrano F. Antioxidant intake and antitumor therapy: toward nutritional recommendations for optimal results. Oxid Med Cell Longev. 2016;2016:6719534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Raffoul JJ, Sarkar FH, Hillman GG. Radiosensitization of prostate cancer by soy isoflavones. Curr Cancer Drug Targets. 2007;7:759-765. [DOI] [PubMed] [Google Scholar]
  • 79. Raffoul JJ, Banerjee S, Singh-Gupta V, et al. Down-regulation of apurinic/apyrimidinic endonuclease 1/redox factor-1 expression by soy isoflavones enhances prostate cancer radiotherapy in vitro and in vivo. Cancer Res. 2007;67:2141-2149. [DOI] [PubMed] [Google Scholar]
  • 80. Raffoul JJ, Banerjee S, Che M, et al. Soy isoflavones enhance radiotherapy in a metastatic prostate cancer model. Int J Cancer. 2007;120:2491-2498. [DOI] [PubMed] [Google Scholar]
  • 81. Ahmad IU, Forman JD, Sarkar FH, et al. Soy isoflavones in conjunction with radiation therapy in patients with prostate cancer. Nutr Cancer. 2010;62:996-1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Singh-Gupta V, Joiner MC, Runyan L, et al. Soy isoflavones augment radiation effect by inhibiting APE1/Ref-1 DNA repair activity in non–small cell lung cancer. J Thorac Oncol. 2010;6:688-698. [DOI] [PubMed] [Google Scholar]
  • 83. Pruitt R, Sasi N, Freeman ML, Sekhar KR. Radiosensitization of cancer cells by hydroxychalcones. Bioorg Med Chem Lett. 2010;20:5997-6000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Tak JK, Lee JH, Park JW. Resveratrol and piperine enhance radiosensitivity of tumor cells. BMB Rep. 2012;45:242-246. [DOI] [PubMed] [Google Scholar]
  • 85. Song CW, Hasegawa T, Kwon HC, Lyons JC, Levitt SH. Increase in tumor oxygenation and radiosensitivity caused by pentoxifylline. Radiat Res. 1992;130:205-210. [PubMed] [Google Scholar]
  • 86. Kim SH, Khil MS, Ryu S, Kim JH. Enhancement of radiation response on human carcinoma cells in culture by pentoxifylline. Int J Radiat Oncol Biol Phys. 1992;25:61-65. [DOI] [PubMed] [Google Scholar]
  • 87. Horvath B, Marton Z, Halmosi R, et al. In vitro antioxidant properties of pentoxifylline, piracetam, and vinpocetine. Clin Neuropharmacol. 2002;25:37-42. [DOI] [PubMed] [Google Scholar]
  • 88. Zhang X, Sharma RK, Agarwal A, Falcone T. Effect of pentoxifylline in reducing oxidative stress-induced embryotoxicity. J Assist Reprod Genet. 2005;22:415-417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Tong L, Chuang CC, Wu S, Zuo L. Reactive oxygen species in redox cancer therapy. Cancer Lett. 2015;367:18-25. [DOI] [PubMed] [Google Scholar]
  • 90. Glasauer A, Chandel NS. Targeting antioxidants for cancer therapy. Biochem Pharmacol. 2014;92:90-101. [DOI] [PubMed] [Google Scholar]
  • 91. Watson J. Oxidants, antioxidants and the current incurability of metastatic cancers. Open Biol. 2013;3:120144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Block KI, Koch AC, Mead MN, Tothy PK, Newman RA, Gyllenhaal C. Impact of antioxidant supplementation on chemotherapeutic efficacy: a systematic review of the evidence from randomized controlled trials. Cancer Treat Rev. 2007;33:407-418. [DOI] [PubMed] [Google Scholar]
  • 93. Block KI, Koch AC, Mead MN, Tothy PK, Newman RA, Gyllenhaal C. Impact of antioxidant supplementation on chemotherapeutic toxicity: a systematic review of the evidence from randomized controlled trials. Int J Cancer. 2008;123:1227-1239. [DOI] [PubMed] [Google Scholar]
  • 94. Yasueda A, Urushima H, Ito T. Efficacy and interaction of antioxidant supplements as adjuvant therapy in cancer treatment: a systematic review. Integr Cancer Ther. 2016;15:17-39. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Integrative Cancer Therapies are provided here courtesy of SAGE Publications

RESOURCES