Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Immunomodulatory Effects of Drugs for Effective Cancer Immunotherapy

Immunomodulatory Effects of Drugs for Effective Cancer Immunotherapy Hindawi Journal of Oncology Volume 2018, Article ID 8653489, 7 pages https://doi.org/10.1155/2018/8653489 Review Article Immunomodulatory Effects of Drugs for Effective Cancer Immunotherapy Maiko Matsushita and Mai Kawaguchi Division of Clinical Physiology and erapeutics, Keio University, Faculty of Pharmacy, --, Shiabakoen, Minato-Ku, Tokyo, Japan Correspondence should be addressed to Maiko Matsushita; matsushita-mk@pha.keio.ac.jp Received 31 August 2018; Accepted 10 October 2018; Published 25 October 2018 Guest Editor: Hailun Wang Copyright © 2018 Maiko Matsushita and Mai Kawaguchi. is Th is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Recent advances in cancer immunotherapy, including immune checkpoint inhibitors or adoptive T cell therapies, have contributed to better outcomes in cancer patients. However, there are still many cancers with no cure. Therefore, combinations of several treatment strategies are being explored, and enhancing anticancer immunity will play an important role to combat the disease. eTh re have been several reports on the immune-modulatory effects of commonly used drugs, namely, statin, metformin, and angiotensin receptor blockers (ARBs), which suggest that these drugs could enhance immunity against cancer cells. Other anticancer drugs, such as anthracyclines, thalidomides, lenalidomides, and hypomethylating drugs, could also strengthen the immune system to attack cancer cells at a relatively low dose. Hence, these drugs might contribute to better outcomes in cancer patients. 1. Introduction drugs [12, 13]. In this review, we discuss drugs that have been reported to elicit immunomodulatory effects in addition to Immune system dysfunction is related to many diseases, their original pharmacological effects. including inflammatory diseases, autoimmune diseases, We searched previously published literature for pa- infectious diseases, atherosclerosis, and cancer [1–5]. Several pers on immunomodulatory function of drugs using drugs that directly target the immune system have been PubMed (https://www.ncbi.nlm.nih.gov/pubmed/). By using developed for the treatment of these disorders. For example, “immunomodulating” and “drugs” as search words, we immune checkpoint inhibitors (ICIs) have greatly improved obtained 3433 papers (August 2018). We excluded papers the outcome of various cancers by altering patients’ immune- with monoclonal antibodies and previously approved im- suppressive status and enhancing antitumor immunity [6– munosuppressive drugs, such as corticosteroids. We classified 8]. However, even ICIs are ineffective in the treatment and the remaining drugs, obtained in the search, based on their cure of certain cancers. Therefore, new strategies to enhance effects on cytokines (Table 1), immune cells (T cells, B the efficacy of these treatments are needed. Moreover, ICIs cells, and antigen presenting cells; Table 2), and immune- and many of the other targeted anticancer therapies consist of related signaling pathways (nuclear factor-kappa B: NF-kB, monoclonal antibodies, which make them highly expensive. Signal Transducers and Activator of Transcription: STAT, On the other hand, immunomodulatory effects have Peroxisome Proliferator-Activated Receptor 𝛾 :PPAR1𝛾 , been described for several small molecular drugs, which Extracellular Signal-Regulated Kinase: ERK, mechanistic have been used for a long time to treat common diseases, target of rapamycin: mTOR, and AMP-activated protein including hyperlipidemia, diabetes, and hypertension [9–11]. kinase: AMPK; Table 3). These drugs are cost-eeff ctive and could be used safely, as their adverse reactions are well known. If immunomodula- To ascertain drugs that could be used for cancer treat- ment, we selected the drugs that have been reported to have tory effects of these previously approved drugs are clearly established, it might be possible to enhance the eeff cts of anticancer effects by enhancing immunological responses to cancer cells. Three drugs approved for treatment of conventional cancer therapy by combinatorial use of these 2 Journal of Oncology ff fl Table 1: Eects on cytokine productions by commonly used drugs. IL-1 IL-6 IL-8 TNF- IFN- IL-2 IL-4 IL-12 IL-18 TNF- IL-3 IL-5 IL-10 IL-13 H2 receptor antagonist NSAIDs Prostaglandin Noroxacin ↓↓ ↑ Linezolid ↓↓ ↓↓↓ ↑ Macrolide ↓↓or↑ ↑↓↓ ↓or↑↓or↑↓or↑↓or↑ Antifungal ↑↑ ↑ ↓↓ drugs 2adrenalin agonist D2 receptor agonist ACE inhibitor ↓ ARB ↓↓ Ca channel ↓or↑↓or↑ ↓↓↑ blocker Digoxin DPP4 inhibitor GLP-1 receptor ↓↓ agonist Biguanide Statin ↓ Journal of Oncology 3 Table 2: Eeff cts on immune cells by commonly used drugs. T celll Th1 Treg Th17 MDSC CTL B cell NK CD4 CD8+TIL H2 receptor antagonist NSAIDs ↑ Prostaglandin Norfloxacin Linezolid Macrolide ↑ Antifungal drugs 𝛽 2adrenalin agonist D2 receptor agonist ACE inhibitor ↓ ARB ↓ Ca channel blocker Digoxin ↓ DPP4 inhibitor ↑↓ Biguanide ↑ Statin ↓ ↓↑↓ ↓ Table 3: Eeff cts on gene expressions in immune-related signaling pathways by commonly used drugs. NF-𝜅BSTAT PPAR1𝛾 ERK mTOR AMPK H2 receptor antagonist NSAIDs Prostaglandin Norfloxacin Linezolid ↓ Macrolide ↓↓ Antifungal drugs 𝛽 2adrenalin agonist D2 receptor agonist ACE inhibitor ARB Ca channel ↓or↑ blocker Digoxin DPP4 inhibitor ↑ Biguanide ↓↑ Statin ↓↑ ↑ Cannabidiol 4 Journal of Oncology common diseases (statin, metformin, and angiotensin recep- angiotensin, which play a pathophysiological role in cancer tor blocker) and three categories of anticancer drugs development [32]. Several clinical studies have demonstrated that show immunomodulatory effects (thalidomide and its that taking ARBs could decrease the risk of developing derivatives, anthracyclines or other chemotherapeutic drugs, cancers [33, 34]. In vitro and in vivo studies have further and demethylating agents) are discussed further for their revealed that these anticancer effects of ARB include direct anticancer property via immunological mechanisms. suppression of cancer growth and increase of antitumor immunity [35–38]. Nakamura et al. showed that adminis- tration of ARB to colon cancer-bearing mice induced the 2. Immunomodulatory Effects of Drugs Used expansion of cancer-specific CTLs and reduced the levels of for Treating Common Diseases immune-suppressive cytokines, interleukin-6 (IL-6), IL-10, vascular endothelial cell factor, and arginase in the tumor .. Statins. Statins have been used by millions of peo- microenvironment [39]. They demonstrated that combina- ple for lowering blood lipids to prevent coronary heart tion therapy with ARB and anti PD-L1 antibody significantly disease [14]. They inhibit hydroxymethylglutaryl (HMG) reduced tumor size, suggesting that this therapy is also useful CoA reductase in the cholesterol biosynthesis pathway. In in clinical setting. addition to their cholesterol-lowering effects, many studies have shown that statins also exhibit immune-modulatory properties through mevalonate-independent and -dependent 3. Immunomodulating Effects of pathways [15]. They inhibit the activity of Ras and Rho family Anticancer Drugs GTPases, which regulate various cellular functions, such as cell death, metastasis, and immune reactions [16–18]. Statins .. alidomide, Lenalidomide, and Pomalidomide. Thalido- aec ff t the immune cells by decreasing the production of mide, and its derivatives, lenalidomide, and pomalidomide inflammatory cytokines and activating CD8 Tcells [19]. are used as key drugs in the treatment of multiple myeloma, Sarrabayrouse et al. have also reported that statin treatment which is a plasma cell neoplasm [40–42]. Thalidomide first induces the expression of CD80 and CD86 in melanoma cells made headlines for its direct tumoricidal effects on myeloma by upregulating their gene expression through transcriptional cells by causing cell cycle arrest and also its antiangiogenic factors controlled by Rho proteins [20]. In fact, clinical properties. Later, it was classified as an immunomodula- studies have confirmed the role of statins in preventing tory drug (IMiD) owing to its immunological effects [43]. several types of cancer [21–23]. These data suggest that statins IMiDs stimulate T cells and natural killer T (NKT) cells to might be useful for enhancing the immunity against cancer secrete IL-2 and interferon-𝛾 , leading to NK cell activation cells. and inhibition of regulatory T cells. As a result, myeloma- specicfi immunity is ampliefi d [44–46]. The FDA has recently approved lenalidomide for maintenance therapy of posttrans- .. Metformin. Metformin is one of the widely used antidi- plant myeloma patients, considering that lenalidomide can abetic drugs synthesized from the plant Galega officinalis, suppress the residual myeloma cells. which lowers blood glucose level by suppressing hepatic glucose production in patients with type 2 diabetes [24]. It inhibits NADH: ubiquinone oxidoreductase located in .. Anthracyclines and Other Chemotherapeutic Drugs. the mitochondrial membrane, leading to the activation of Some of conventional anticancer drugs, such as anthracy- 5 -AMP-activated protein kinase (AMPK) and suppression clines or alkylating agents, have been reported to cause of gluconeogenesis. AMPK-independent pathway also con- induction of immunomodulatory effects on various cancer tributes to its pharmacological actions [25]. In addition to cells by enhancing the cell surface expression of calreticulin its antihyperglycemic effects, there are many reports of its (CRT) followed by the release of high mobility group box role in immune response. It has been shown that metformin 1(HMGB1),ATP,annexin A1, and typeI interferon from enhances the number and function of tumor-infiltrating cancer cells [47–50]. The dendritic cells (DCs) then recognize lymphocytes (TILs) [26]. Pereira et al. have reported that CRT, HMGB1, and ATP through CD91, Toll-like receptor metformin exerts strong immunomodulatory eeff cts and 4, and P2X purinoreceptor 7, respectively, and take up the contributes to reduced lung metastasis of melanoma cells cancer cells. This series of events is called immunogenic [27]. Cha et al. have shown that metformin reduces the cell death (ICD) [51–53]. Mouse immunization experiments stability and membrane localization of programmed death- using cancer cells pretreated with chemotherapeutic drugs, ligand 1 (PD-L1) and contributes to the enhancement of such as doxorubicin or mitoxantrone, have shown eeff ctive cytotoxic T lymphocyte (CTL) activity against cancer cells cancer regression through CRT expression and HMGB1 [28]. Metformin also exerts anti-inflammatory effects [29, secretion [54–56]. These observations were further con- 30]. It was recently reported that these effects might be related firmed by clinical data, which have indicated that CRT to the alteration of gut microbiota [31]. These results suggest expression is important for the improvement of disease that metformin can be used for the treatment of cancer. outcome in cancer patients [57]. These results suggest that some of the chemotherapeutic drugs not only kill cancer .. Angiotensin Receptor Blockers (ARBs). Renin-angiotens- cells directly but also strengthen patients’ immune reactions in system mainly controls blood pressure; however, cancer against cancer cells, thereby contributing to the eradication cells and their microenvironment also express renin and of cancer cells. Moreover, other types of anticancer drugs, Journal of Oncology 5 such as epidermal growth factor receptor inhibitors, also elicit of inflammatory bowel diseases,” Acta Gastro-Enterologica Bel- gica,vol.69, no. 4,pp. 393–405, 2006. immune reactions against cancer cells through ICD [58]. ICD inducers, especially cyclophosphamide, are now expected to [2]A. Lombardi, E. Tsomos,S. S.Hammerstad, and Y.Tomer, “Interferon alpha: eTh key trigger of type 1 diabetes,” Journal of enhance the antitumor effects of other immunotherapies, Autoimmunity, pp. 30323–30328, 2018. such as cancer vaccines or immune checkpoint inhibitors [59–62]. [3] Q. H. Sam, W. S. Yew, C. J. Seneviratne, M. W. Chang, and L. Y. Chai, “Immunomodulation as Therapy for Fungal Infection: Are We Closer?” Frontiers in Microbiology,vol.9,2018. .. Azacitidine and Decitabine. Hypermethylation of tumor suppressor genes in the promoter region and subsequent [4] A. Gistera˚ and D. F. Ketelhuth, “Lipid-driven immunometabolic responses in atherosclerosis,” Current Opinion in Lipidology,pp. silencing of these genes is observed in many cancer cells. 375–380, 2018. Hence, hypomethylating agents (HMAs) are expected to [5] D. S. Chen and I. Mellman, “Elements of cancer immunity and be useful in cancer treatment by causing epigenetic mod- the cancer-immune set point,” Nature,vol.541,no.7637,pp.321– ulation of these cancer-related genes [63]. HMAs, such 330, 2017. as azacitidine or decitabine, have been approved for the [6] B.Wahid,A.Ali, S. Rafique etal., “An overview of cancer treatment of myelodysplastic syndromes (MDS) and acute immunotherapeutic strategies,” Immunotherapy,vol. 10,no.11, myeloid leukemia (AML) [64]. These HMAs are analogues pp. 999–1010, 2018. of pyrimidine nucleosides, which are incorporated into RNA [7] L. A. Emens, “Breast cancer immunotherapy: Facts and hopes,” or DNA, and they impair DNA methylation by inhibiting Clinical Cancer Research,vol. 24, no.3, pp. 511–520, 2018. DNA methyltransferase [65, 66]. At the same time, HMAs are [8] A.Ribas and J.D.Wolchok,“Cancer immunotherapy using known to enhance expression of cancer-specicfi antigens and checkpoint blockade,” Science,vol.359, no.6382,pp.1350–1355, MHC molecules, making cancer cells sensitive to killing by CTLs in vitro and in vivo [67–69]. Stone et al. have recently [9] T.Fehr, C.Kahlert,W.Fierz et al., “Statin-inducedimmunomo- reported that HMAs could also alter immunosuppressive dulatory effects on human T cells in vivo,” Atherosclerosis,vol. tumor microenvironment by activating type 1 interferon 175, no.1,pp.83–90,2004. signaling, in combination with another class of epigenetic [10] F.Ursini,E.Russo,G.Pellinoetal.,“MetforminandAutoimmu- drugs, histone deacetylase inhibitors [70]. nity: A “New Deal” of an Old Drug,” Frontiers in Immunology, vol. 9, 2018. 4. Conclusion [11] J. An,T.Nakajima, K. Kuba, and A. Kimura,“Losar- tan inhibits LPS-induced inflammatory signaling through a There have been reports on immunomodulating properties PPAR𝛾 -dependent mechanism in human THP-1 macrophages,” of several commonly used drugs, such as statins, metformin, Hypertension Research,vol. 33, no.8, pp. 831–835, 2010. and ARB, which can be used to treat or prevent cancer [12] A. Popovic, E. M. Jaee, ff and N. Zaidi, “Emerging strategies for via increased antitumor immunity. These drugs can be used combination checkpoint modulators in cancer immunother- safely because of their known adverse reactions and might apy,” e Journal of Clinical Investigation, vol.128,no. 8, pp. contribute to cancer treatment. Among anticancer drugs, 3209–3218, 2018. anthracyclines, IMiDs, and epigenetic drugs not only kill the [13] P.A.Ott,F.S.Hodi, H. L.Kaufman,J.M.Wigginton, and J.D. cancer cells directly, but also enhance the immune system to Wolchok, “Combination immunotherapy: A road map,” Journal attack the cancer cells. It will be beneficial to combine these for Immunoerapy of Cancer,vol.5,no.1,2017. drugs with conventional cancer therapies to achieve better [14] M. Banach, C. Serban, A. Sahebkar et al., “Impact of statin outcomes in cancer patients. therapy on coronary plaque composition: a systematic review and meta-analysis of virtual histology intravascular ultrasound studies,” BMC Medicine,vol.13,no. 1,2015. Conflicts of Interest [15] G. Sarrabayrouse, C. Pich, I. Teiti, and A. F. Tilkin-Mariame, “Regulatory properties of statins and rho gtpases prenylation The authors report no conifl cts of interest. inhibitiors to stimulate melanoma immunogenicity and pro- mote anti-melanoma immune response,” International Journal Acknowledgments of Cancer, vol.140,no. 4,pp. 747–755, 2017. [16] V. Ziegler, C. Henninger, I. Simiantonakis et al., “Rho inhibition This work received funding from Keio Gijuku Fukuzawa by lovastatin affects apoptosis and DSB repair of primary Memorial Fund for the Advancement of Education and human lung cells in vitro and lung tissue in vivo following Research. We are thankful to Ms. Saori Matsumoto, Mr. Sho fractionated irradiation,” Cell Death & Disease,vol. 8,no. 8,p. Kashiwazaki, and Mr. Michio Kobori for their suggestions e2978, 2017. about drugs related to immunogenic cell death. We are also [17] R. Palomino-Morales, S. Perales, C. Torres, A. Linares, and thankful to Dr. Yutaka Hattori and Dr. Daiju Ichikawa for M. J. Alejandre, “Eec ff t of HMG-CoA reductase inhibition on their valuable feedbacks. vascular smooth muscle cells extracellular matrix production: Role of RhoA,” Current Vascular Pharmacology,vol. 14, no. 4, pp. 345–352, 2016. References [18] M.Islam, S. Sharma, B.Kumar,and T.N. Teknos,“Atorvastatin [1] D. Latinne and R. Fiasse, “New insights into the cellular inhibits RhoC function and limits head and neck cancer immunology of the intestine in relation to the pathophysiology metastasis,” Oral Oncology,vol.49, no.8,pp.778–786, 2013. 6 Journal of Oncology [19] J. Cote-Daigneault, S. Mehandru, R. Ungaro, A. Atreja, and J.- Cancer,” World Journal of Surgery, vol.41, no.9, pp.2361–2370, F. Colombel, “Potential Immunomodulatory Eeff cts of Statins 2017. in Inflammatory Bowel Disease,” Inflammatory Bowel Diseases, [35] K. Oura, T. Tadokoro, S. Fujihara et al., “Telmisartan inhibits vol. 22, no. 3, pp. 724–732, 2015. hepatocellular carcinoma cell proliferation in vitro by inducing [20] G. Sarrabayrouse, C. Pich, R. Moriez et al., “Melanoma cells cell cycle arrest,” Oncology Reports, vol.38, no.5, pp. 2825–2835, treated with GGTI and IFN-𝛾 allow murine vaccination and 2017. enhance cytotoxic response against human melanoma cells,” [36] S. Fujihara, A. Morishita, K. Ogawa et al., “eTh angiotensin II PLoS ONE, vol.5,no.2,2010. type 1 receptor antagonist telmisartan inhibits cell proliferation [21] T. J. Vogel, M. T. Goodman, A. J. Li, and C. Y. Jeon, “Statin treat- and tumor growth of esophageal adenocarcinoma via the AMPKalpha/mTOR pathway in vitro and in vivo,” Oncotarget, ment is associated with survival in a nationally representative population of elderly women with epithelial ovarian cancer,” vol.8,no.5,2017. Gynecologic Oncology,vol.146,no.2,pp.340–345, 2017. [37] S. Saikawa, K. Kaji, N. Nishimura et al., “Angiotensin receptor [22] A. Wang, H. A. Wakelee, A. K. Aragaki et al., “Protective Eec ff ts blockade attenuates cholangiocarcinoma cell growth by inhibit- ing the oncogenic activity of Yes-associated protein,” Cancer of Statins in Cancer: Should eTh y Be Prescribed for High-Risk Patients?” Current Atherosclerosis Reports,vol.18, no. 12,2016. Letters, vol. 434, pp. 120–129, 2018. [38] R.Coulson, S.H. Liew,A.A.Connelly,and etal., “eTh [23] P. Desai, R. Wallace, and M. Anderson, “An analysis of the effect of statins on the risk of Non-Hodgkin’s Lymphoma in the angiotensin receptor blocker, losartan, inhibits mammary tumor development and progression to invasive carcinoma,” Women’s Health Initiative cohort,” Cancer Medicine,vol.7, no. 5, pp. 2121–2130, 2018. Oncotarget, vol. 8, pp. 18640–18656, 2017. [39] K. Nakamura, T. Yaguchi, G. Ohmura et al., “Involvement [24] R. J. Shaw, K. A. Lamia, D. Vasquez et al., “Medicine: the kinase LKB1 mediates glucose homeostasis in liver and therapeutic of local renin-angiotensin system in immunosuppression of tumor microenvironment,” Cancer Science, vol.109,no.1, pp. effects of metformin,” Science,vol. 310,no. 5754, pp.1642–1646, 2005. 54–64, 2018. [40] C. S.Chim,S.K.Kumar, R. Z. Orlowskiet al., “Management [25] G.Rena, D.G.Hardie, and E. R.Pearson, “eTh mechanisms of action of metformin,” Diabetologia,vol.60, no.9,pp.1577–1585, of relapsed and refractory multiple myeloma: Novel agents, antibodies, immunotherapies and beyond,” Leukemia,vol. 32, no. 2, pp. 252–262, 2018. [26] S.Eikawa,M.Nishida,S.Mizukami,C.Yamazaki,E.Nakayama, [41] P. Moreau, “How I treat myeloma with new agents,” Blood,vol. and H. Udono, “Immune-mediated antitumor effect by type 2 130, no. 13, pp. 1507–1513, 2017. diabetes drug, metformin,” Proceedings of the National Acadamy of Sciences of the United States of America, vol. 112, no. 6, pp. [42] B. Pan and S. Lentzsch, “The application and biology of 1809–1814, 2015. immunomodulatory drugs (IMiDs) in cancer,” Pharmacology & erapeutics,vol.136, no.1,pp. 56–68, 2012. [27] F. V. Pereira, A. C. Melo, J. S. Low et al., “Metformin exerts antitumor activity via induction of multiple death pathways in [43] J. B. Zeldis, R. Knight, M. Hussein, R. Chopra, and G. tumor cells and activation of a protective immune response,” Muller, “A review of the history, properties, and use of the Oncotarget ,vol.9, no. 40,2018. immunomodulatory compound lenalidomide,” Annals of the New York Academy of Sciences, vol. 1222, no. 1, pp. 76–82, 2011. [28] J. Talairach and P. Thournoux, “Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation [44] K. Luptakova, J. Rosenblatt, B. Glotzbecker et al., “Lenalidomide of PD-L1,” Mollecular Cell, vol.71,pp.606–620, 2018. enhances anti-myeloma cellular immunity,” Cancer Immunol- ogy, Immunotherapy,vol.62,no. 1, pp.39–49, 2013. [29] D. Di Fusco, V. Dinallo, I. Monteleone et al., “Metformin inhibits inflammatory signals in the gut by controlling AMPK and p38 [45] H. Quach, D. Ritchie, A. K. Stewart et al., “Mechanism of action MAP kinase activation,” Clinical Science,vol.132,no. 11, pp. of immunomodulatory drugs (IMiDS) in multiple myeloma,” 1155–1168, 2018. Leukemia,vol. 24, no.1, pp. 22–32,2010. [30] H. J. Son, J. Lee, S. Y. Lee, and etal., “Metformin attenuates [46] A. K. Hsu, H. Quach, T. Tai et al., “e Th immunostimulatory experimental autoimmune arthritis through reciprocal regula- eeff ct of lenalidomide on NK-cell function is profoundly inhib- tion of Th17/Treg balance and osteoclastogenesis,” Mediators of ited by concurrent dexamethasone therapy,” Blood,vol.117,no. Inflammation,vol.2014,pp.1–13, 2014. 5, pp. 1605–1613, 2011. [31] J. Rodriguez, S. Hiel, and N. M. Delzenne, “Metformin: Old [47] M. Obeid, A. Tesniere, F. Ghiringhelli et al., “Calreticulin friend, new ways of action-implication of the gut microbiome?” exposure dictates the immunogenicity of cancer cell death,” Current Opinion in Clinical Nutrition & Metabolic Care,vol.21, Nature Medicine,vol.13,no.1,pp.54–61, 2007. no.4,pp.294–301,2018. [48] J. W. Hodge, C. T. Garnett, B. Farsaci et al., “Chemotherapy- [32] A. J. George, W. G. o Th mas, and R. D. Hannan, “eTh renin- induced immunogenic modulation of tumor cells enhances angiotensin system and cancer: old dog, new tricks,” Nature killing by cytotoxic T lymphocytes and is distinct from Reviews Cancer, vol. 10, no. 11, pp. 745–759, 2010. immunogenic cell death,” International Journal of Cancer,vol. ´ 133, no.3,pp.624–636, 2013. [33] J. Busby, U. McMenamin, A. Spence, B. T. Johnston, C. Hughes, and C. R. Cardwell, “Angiotensin receptor blocker use and [49] E. Vacchelli, Y. Ma, E. E. Baracco et al., “Chemotherapy- gastro-oesophageal cancer survival: a population-based cohort induced antitumor immunity requires formyl peptide receptor study,” Alimentary Pharmacology & erapeutics,vol.47, no.2, 1,” Science,vol.350, pp.972–978,2015. pp. 279–288, 2018. [50] A. Sistigu, T. Yamazaki, E. Vacchelli et al., “Cancer cell- [34] M. Cerullo,F.Gani, S. Y. Chen, J.K. Canner,and T. M.Pawlik, autonomous contribution of type I interferon signaling to the “Impact of Angiotensin Receptor Blocker Use on Overall efficacy of chemotherapy,” Nature Medicine,vol.20, no. 11, pp. Survival Among Patients Undergoing Resection for Pancreatic 1301–1309, 2014. Journal of Oncology 7 [51] L. Galluzzi, A. Buque, ´ O. Kepp, L. Zitvogel, and G. Kroemer, [68] M. Matsushita, Y. Otsuka, N. Tsutsumida et al., “Identification “Immunogenic cell death in cancer and infectious disease,” of Novel HLA-A*24:02-Restricted Epitope Derived from a Nature Reviews Immunology, vol. 17, no. 2, pp. 97–111, 2017. Homeobox Protein Expressed in Hematological Malignancies,” PLoS ONE,vol.11,no.1,p.e0146371, 2016. [52] G. Kroemer, L. Galluzzi, O. Kepp, and L. Zitvogel, “Immuno- genic cell death in cancer therapy,” Annual Review of Immunol- [69] P. Srivastava, B. E. Paluch, J. Matsuzaki et al., “Induction of ogy,vol.31,pp. 51–72, 2013. cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monother- [53] S. Gebremeskel and B. Johnston, “Concepts and mechanisms underlying chemotherapy induced immunogenic cell death: apy,” Oncotarget ,vol.7,no.11,pp. 12840–12856,2016. Impact on clinical studies and considerations for combined [70] M. L. Stone, K. B. Chiappinelli, H. Li et al., “Epigenetic therapy therapies,” Oncotarget ,vol.6,no.39,pp.41600–41619,2015. activates type I interferon signaling in murine ovarian cancer to [54] N. Casares, M. O. Pequignot, A. Tesniere et al., “Caspase- reduce immunosuppression and tumor burden,” Proceedings of the National Acadamy of Sciences of the United States of America, dependent immunogenicity of doxorubicin-induced tumor cell death,” e Journal of Experimental Medicine,vol.202, no.12, vol. 114, no. 51, pp. E10981–E10990, 2017. pp. 1691–1701, 2005. [55] A. Tesniere, F. Schlemmer, V. Boige et al., “Immunogenic death of colon cancer cells treated with oxaliplatin,” Oncogene,vol. 29, no.4,pp.482–491, 2010. [56] X. Lu, Z.-C. Ding, Y. Cao et al., “Alkylating agent melphalan augments the efficacy of adoptive immunotherapy using tumor- specific CD4+ T cells,” e Journal of Immunology,vol. 194, no. 4, pp. 2011–2021, 2015. [57] J. Fucikova, I. Truxova, M. Hensler et al., “Calreticulin exposure by malignant blasts correlates with robust anticancer immunity and improved clinical outcome in AML patients,” Blood,vol. 128, no. 26, pp. 3113–3124, 2016. [58] C. Pozzi,A.Cuomo,I. Spadoni et al., “eE Th GFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death,” Nature Medicine,vol.22,no.6,pp. 624–631, 2016. [59] L.Bezu,L.C.Gomes-de-Silva,H.Dewitteetal.,“Combinatorial Strategies for the Induction of Immunogenic Cell Death,” Frontiers in Immunology, vol.6,2015. [60] C.Pfirschke, C.Engblom, S.Rickelt et al., “Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy,” Immunity,vol. 44, no.2, pp. 343–354,2016. [61] B. Montico, A. Nigro, V. Casolaro, and J. Dal Col, “Immuno- genic Apoptosis as a Novel Tool for Anticancer Vaccine Devel- opment,” International Journal of Molecular Sciences,vol. 19, no. 2, p. 594, 2018. [62] A. D.Garg,S.More, N. Rufo et al., “Trial watch: Immuno- genic cell death induction by anticancer chemotherapeutics,” OncoImmunology,vol.6,no. 12, p.e1386829, 2017. [63] L. Ding, MH. Bailey, and E. Porta-Pardo, “Perspective on oncogenic processes at the end of the beginning of cancer genomics,” Cell, vol.173,pp.305–320, 2018. [64] E. J. B.Derissen,J. H.Beijnen,and J. H. M. Schellens,“Concise drug review: Azacitidine and decitabine,” e Oncologist,vol.18, no.5,pp.619–624, 2013. [65] T.Schroeder, C.Rautenberg, R.Haas, U.Germing, and G. Kobbe, “Hypomethylating agents for treatment and prevention of relapse aeft r allogeneic blood stem cell transplantation,” International Journal of Hematology,vol. 107, no.2,pp.138–150, [66] Y. Chen, P. L. McCarthy, T. Hahn et al., “Methods to prevent and treat relapse aer ft hematopoietic stem cell transplantation with tyrosine kinase inhibitors, immunomodulating drugs, deacety- lase inhibitors, and hypomethylating agents,” Bone Marrow Transplantation. [67] N. Luo, M. J. Nixon, P. I. Gonzalez-Ericsson et al., “DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer,” Nature Communications, vol.9,no. 1, 2018. MEDIATORS of INFLAMMATION The Scientific Gastroenterology Journal of World Journal Research and Practice Diabetes Research Disease Markers Hindawi Hindawi Publishing Corporation Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 http://www www.hindawi.com .hindawi.com V Volume 2018 olume 2013 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 International Journal of Journal of Immunology Research Endocrinology Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Submit your manuscripts at www.hindawi.com BioMed PPAR Research Research International Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Journal of Obesity Evidence-Based Journal of Journal of Stem Cells Complementary and Ophthalmology International Alternative Medicine Oncology Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2013 Parkinson’s Disease Computational and Behavioural Mathematical Methods AIDS Oxidative Medicine and in Medicine Neurology Research and Treatment Cellular Longevity Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Journal of Oncology Hindawi Publishing Corporation

Immunomodulatory Effects of Drugs for Effective Cancer Immunotherapy

Journal of Oncology , Volume 2018: 7 – Oct 25, 2018

Loading next page...
 
/lp/hindawi-publishing-corporation/immunomodulatory-effects-of-drugs-for-effective-cancer-immunotherapy-tqtCuuZPrb

References (77)

Publisher
Hindawi Publishing Corporation
Copyright
Copyright © 2018 Maiko Matsushita and Mai Kawaguchi. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
ISSN
1687-8450
eISSN
1687-8469
DOI
10.1155/2018/8653489
Publisher site
See Article on Publisher Site

Abstract

Hindawi Journal of Oncology Volume 2018, Article ID 8653489, 7 pages https://doi.org/10.1155/2018/8653489 Review Article Immunomodulatory Effects of Drugs for Effective Cancer Immunotherapy Maiko Matsushita and Mai Kawaguchi Division of Clinical Physiology and erapeutics, Keio University, Faculty of Pharmacy, --, Shiabakoen, Minato-Ku, Tokyo, Japan Correspondence should be addressed to Maiko Matsushita; matsushita-mk@pha.keio.ac.jp Received 31 August 2018; Accepted 10 October 2018; Published 25 October 2018 Guest Editor: Hailun Wang Copyright © 2018 Maiko Matsushita and Mai Kawaguchi. is Th is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Recent advances in cancer immunotherapy, including immune checkpoint inhibitors or adoptive T cell therapies, have contributed to better outcomes in cancer patients. However, there are still many cancers with no cure. Therefore, combinations of several treatment strategies are being explored, and enhancing anticancer immunity will play an important role to combat the disease. eTh re have been several reports on the immune-modulatory effects of commonly used drugs, namely, statin, metformin, and angiotensin receptor blockers (ARBs), which suggest that these drugs could enhance immunity against cancer cells. Other anticancer drugs, such as anthracyclines, thalidomides, lenalidomides, and hypomethylating drugs, could also strengthen the immune system to attack cancer cells at a relatively low dose. Hence, these drugs might contribute to better outcomes in cancer patients. 1. Introduction drugs [12, 13]. In this review, we discuss drugs that have been reported to elicit immunomodulatory effects in addition to Immune system dysfunction is related to many diseases, their original pharmacological effects. including inflammatory diseases, autoimmune diseases, We searched previously published literature for pa- infectious diseases, atherosclerosis, and cancer [1–5]. Several pers on immunomodulatory function of drugs using drugs that directly target the immune system have been PubMed (https://www.ncbi.nlm.nih.gov/pubmed/). By using developed for the treatment of these disorders. For example, “immunomodulating” and “drugs” as search words, we immune checkpoint inhibitors (ICIs) have greatly improved obtained 3433 papers (August 2018). We excluded papers the outcome of various cancers by altering patients’ immune- with monoclonal antibodies and previously approved im- suppressive status and enhancing antitumor immunity [6– munosuppressive drugs, such as corticosteroids. We classified 8]. However, even ICIs are ineffective in the treatment and the remaining drugs, obtained in the search, based on their cure of certain cancers. Therefore, new strategies to enhance effects on cytokines (Table 1), immune cells (T cells, B the efficacy of these treatments are needed. Moreover, ICIs cells, and antigen presenting cells; Table 2), and immune- and many of the other targeted anticancer therapies consist of related signaling pathways (nuclear factor-kappa B: NF-kB, monoclonal antibodies, which make them highly expensive. Signal Transducers and Activator of Transcription: STAT, On the other hand, immunomodulatory effects have Peroxisome Proliferator-Activated Receptor 𝛾 :PPAR1𝛾 , been described for several small molecular drugs, which Extracellular Signal-Regulated Kinase: ERK, mechanistic have been used for a long time to treat common diseases, target of rapamycin: mTOR, and AMP-activated protein including hyperlipidemia, diabetes, and hypertension [9–11]. kinase: AMPK; Table 3). These drugs are cost-eeff ctive and could be used safely, as their adverse reactions are well known. If immunomodula- To ascertain drugs that could be used for cancer treat- ment, we selected the drugs that have been reported to have tory effects of these previously approved drugs are clearly established, it might be possible to enhance the eeff cts of anticancer effects by enhancing immunological responses to cancer cells. Three drugs approved for treatment of conventional cancer therapy by combinatorial use of these 2 Journal of Oncology ff fl Table 1: Eects on cytokine productions by commonly used drugs. IL-1 IL-6 IL-8 TNF- IFN- IL-2 IL-4 IL-12 IL-18 TNF- IL-3 IL-5 IL-10 IL-13 H2 receptor antagonist NSAIDs Prostaglandin Noroxacin ↓↓ ↑ Linezolid ↓↓ ↓↓↓ ↑ Macrolide ↓↓or↑ ↑↓↓ ↓or↑↓or↑↓or↑↓or↑ Antifungal ↑↑ ↑ ↓↓ drugs 2adrenalin agonist D2 receptor agonist ACE inhibitor ↓ ARB ↓↓ Ca channel ↓or↑↓or↑ ↓↓↑ blocker Digoxin DPP4 inhibitor GLP-1 receptor ↓↓ agonist Biguanide Statin ↓ Journal of Oncology 3 Table 2: Eeff cts on immune cells by commonly used drugs. T celll Th1 Treg Th17 MDSC CTL B cell NK CD4 CD8+TIL H2 receptor antagonist NSAIDs ↑ Prostaglandin Norfloxacin Linezolid Macrolide ↑ Antifungal drugs 𝛽 2adrenalin agonist D2 receptor agonist ACE inhibitor ↓ ARB ↓ Ca channel blocker Digoxin ↓ DPP4 inhibitor ↑↓ Biguanide ↑ Statin ↓ ↓↑↓ ↓ Table 3: Eeff cts on gene expressions in immune-related signaling pathways by commonly used drugs. NF-𝜅BSTAT PPAR1𝛾 ERK mTOR AMPK H2 receptor antagonist NSAIDs Prostaglandin Norfloxacin Linezolid ↓ Macrolide ↓↓ Antifungal drugs 𝛽 2adrenalin agonist D2 receptor agonist ACE inhibitor ARB Ca channel ↓or↑ blocker Digoxin DPP4 inhibitor ↑ Biguanide ↓↑ Statin ↓↑ ↑ Cannabidiol 4 Journal of Oncology common diseases (statin, metformin, and angiotensin recep- angiotensin, which play a pathophysiological role in cancer tor blocker) and three categories of anticancer drugs development [32]. Several clinical studies have demonstrated that show immunomodulatory effects (thalidomide and its that taking ARBs could decrease the risk of developing derivatives, anthracyclines or other chemotherapeutic drugs, cancers [33, 34]. In vitro and in vivo studies have further and demethylating agents) are discussed further for their revealed that these anticancer effects of ARB include direct anticancer property via immunological mechanisms. suppression of cancer growth and increase of antitumor immunity [35–38]. Nakamura et al. showed that adminis- tration of ARB to colon cancer-bearing mice induced the 2. Immunomodulatory Effects of Drugs Used expansion of cancer-specific CTLs and reduced the levels of for Treating Common Diseases immune-suppressive cytokines, interleukin-6 (IL-6), IL-10, vascular endothelial cell factor, and arginase in the tumor .. Statins. Statins have been used by millions of peo- microenvironment [39]. They demonstrated that combina- ple for lowering blood lipids to prevent coronary heart tion therapy with ARB and anti PD-L1 antibody significantly disease [14]. They inhibit hydroxymethylglutaryl (HMG) reduced tumor size, suggesting that this therapy is also useful CoA reductase in the cholesterol biosynthesis pathway. In in clinical setting. addition to their cholesterol-lowering effects, many studies have shown that statins also exhibit immune-modulatory properties through mevalonate-independent and -dependent 3. Immunomodulating Effects of pathways [15]. They inhibit the activity of Ras and Rho family Anticancer Drugs GTPases, which regulate various cellular functions, such as cell death, metastasis, and immune reactions [16–18]. Statins .. alidomide, Lenalidomide, and Pomalidomide. Thalido- aec ff t the immune cells by decreasing the production of mide, and its derivatives, lenalidomide, and pomalidomide inflammatory cytokines and activating CD8 Tcells [19]. are used as key drugs in the treatment of multiple myeloma, Sarrabayrouse et al. have also reported that statin treatment which is a plasma cell neoplasm [40–42]. Thalidomide first induces the expression of CD80 and CD86 in melanoma cells made headlines for its direct tumoricidal effects on myeloma by upregulating their gene expression through transcriptional cells by causing cell cycle arrest and also its antiangiogenic factors controlled by Rho proteins [20]. In fact, clinical properties. Later, it was classified as an immunomodula- studies have confirmed the role of statins in preventing tory drug (IMiD) owing to its immunological effects [43]. several types of cancer [21–23]. These data suggest that statins IMiDs stimulate T cells and natural killer T (NKT) cells to might be useful for enhancing the immunity against cancer secrete IL-2 and interferon-𝛾 , leading to NK cell activation cells. and inhibition of regulatory T cells. As a result, myeloma- specicfi immunity is ampliefi d [44–46]. The FDA has recently approved lenalidomide for maintenance therapy of posttrans- .. Metformin. Metformin is one of the widely used antidi- plant myeloma patients, considering that lenalidomide can abetic drugs synthesized from the plant Galega officinalis, suppress the residual myeloma cells. which lowers blood glucose level by suppressing hepatic glucose production in patients with type 2 diabetes [24]. It inhibits NADH: ubiquinone oxidoreductase located in .. Anthracyclines and Other Chemotherapeutic Drugs. the mitochondrial membrane, leading to the activation of Some of conventional anticancer drugs, such as anthracy- 5 -AMP-activated protein kinase (AMPK) and suppression clines or alkylating agents, have been reported to cause of gluconeogenesis. AMPK-independent pathway also con- induction of immunomodulatory effects on various cancer tributes to its pharmacological actions [25]. In addition to cells by enhancing the cell surface expression of calreticulin its antihyperglycemic effects, there are many reports of its (CRT) followed by the release of high mobility group box role in immune response. It has been shown that metformin 1(HMGB1),ATP,annexin A1, and typeI interferon from enhances the number and function of tumor-infiltrating cancer cells [47–50]. The dendritic cells (DCs) then recognize lymphocytes (TILs) [26]. Pereira et al. have reported that CRT, HMGB1, and ATP through CD91, Toll-like receptor metformin exerts strong immunomodulatory eeff cts and 4, and P2X purinoreceptor 7, respectively, and take up the contributes to reduced lung metastasis of melanoma cells cancer cells. This series of events is called immunogenic [27]. Cha et al. have shown that metformin reduces the cell death (ICD) [51–53]. Mouse immunization experiments stability and membrane localization of programmed death- using cancer cells pretreated with chemotherapeutic drugs, ligand 1 (PD-L1) and contributes to the enhancement of such as doxorubicin or mitoxantrone, have shown eeff ctive cytotoxic T lymphocyte (CTL) activity against cancer cells cancer regression through CRT expression and HMGB1 [28]. Metformin also exerts anti-inflammatory effects [29, secretion [54–56]. These observations were further con- 30]. It was recently reported that these effects might be related firmed by clinical data, which have indicated that CRT to the alteration of gut microbiota [31]. These results suggest expression is important for the improvement of disease that metformin can be used for the treatment of cancer. outcome in cancer patients [57]. These results suggest that some of the chemotherapeutic drugs not only kill cancer .. Angiotensin Receptor Blockers (ARBs). Renin-angiotens- cells directly but also strengthen patients’ immune reactions in system mainly controls blood pressure; however, cancer against cancer cells, thereby contributing to the eradication cells and their microenvironment also express renin and of cancer cells. Moreover, other types of anticancer drugs, Journal of Oncology 5 such as epidermal growth factor receptor inhibitors, also elicit of inflammatory bowel diseases,” Acta Gastro-Enterologica Bel- gica,vol.69, no. 4,pp. 393–405, 2006. immune reactions against cancer cells through ICD [58]. ICD inducers, especially cyclophosphamide, are now expected to [2]A. Lombardi, E. Tsomos,S. S.Hammerstad, and Y.Tomer, “Interferon alpha: eTh key trigger of type 1 diabetes,” Journal of enhance the antitumor effects of other immunotherapies, Autoimmunity, pp. 30323–30328, 2018. such as cancer vaccines or immune checkpoint inhibitors [59–62]. [3] Q. H. Sam, W. S. Yew, C. J. Seneviratne, M. W. Chang, and L. Y. Chai, “Immunomodulation as Therapy for Fungal Infection: Are We Closer?” Frontiers in Microbiology,vol.9,2018. .. Azacitidine and Decitabine. Hypermethylation of tumor suppressor genes in the promoter region and subsequent [4] A. Gistera˚ and D. F. Ketelhuth, “Lipid-driven immunometabolic responses in atherosclerosis,” Current Opinion in Lipidology,pp. silencing of these genes is observed in many cancer cells. 375–380, 2018. Hence, hypomethylating agents (HMAs) are expected to [5] D. S. Chen and I. Mellman, “Elements of cancer immunity and be useful in cancer treatment by causing epigenetic mod- the cancer-immune set point,” Nature,vol.541,no.7637,pp.321– ulation of these cancer-related genes [63]. HMAs, such 330, 2017. as azacitidine or decitabine, have been approved for the [6] B.Wahid,A.Ali, S. Rafique etal., “An overview of cancer treatment of myelodysplastic syndromes (MDS) and acute immunotherapeutic strategies,” Immunotherapy,vol. 10,no.11, myeloid leukemia (AML) [64]. These HMAs are analogues pp. 999–1010, 2018. of pyrimidine nucleosides, which are incorporated into RNA [7] L. A. Emens, “Breast cancer immunotherapy: Facts and hopes,” or DNA, and they impair DNA methylation by inhibiting Clinical Cancer Research,vol. 24, no.3, pp. 511–520, 2018. DNA methyltransferase [65, 66]. At the same time, HMAs are [8] A.Ribas and J.D.Wolchok,“Cancer immunotherapy using known to enhance expression of cancer-specicfi antigens and checkpoint blockade,” Science,vol.359, no.6382,pp.1350–1355, MHC molecules, making cancer cells sensitive to killing by CTLs in vitro and in vivo [67–69]. Stone et al. have recently [9] T.Fehr, C.Kahlert,W.Fierz et al., “Statin-inducedimmunomo- reported that HMAs could also alter immunosuppressive dulatory effects on human T cells in vivo,” Atherosclerosis,vol. tumor microenvironment by activating type 1 interferon 175, no.1,pp.83–90,2004. signaling, in combination with another class of epigenetic [10] F.Ursini,E.Russo,G.Pellinoetal.,“MetforminandAutoimmu- drugs, histone deacetylase inhibitors [70]. nity: A “New Deal” of an Old Drug,” Frontiers in Immunology, vol. 9, 2018. 4. Conclusion [11] J. An,T.Nakajima, K. Kuba, and A. Kimura,“Losar- tan inhibits LPS-induced inflammatory signaling through a There have been reports on immunomodulating properties PPAR𝛾 -dependent mechanism in human THP-1 macrophages,” of several commonly used drugs, such as statins, metformin, Hypertension Research,vol. 33, no.8, pp. 831–835, 2010. and ARB, which can be used to treat or prevent cancer [12] A. Popovic, E. M. Jaee, ff and N. Zaidi, “Emerging strategies for via increased antitumor immunity. These drugs can be used combination checkpoint modulators in cancer immunother- safely because of their known adverse reactions and might apy,” e Journal of Clinical Investigation, vol.128,no. 8, pp. contribute to cancer treatment. Among anticancer drugs, 3209–3218, 2018. anthracyclines, IMiDs, and epigenetic drugs not only kill the [13] P.A.Ott,F.S.Hodi, H. L.Kaufman,J.M.Wigginton, and J.D. cancer cells directly, but also enhance the immune system to Wolchok, “Combination immunotherapy: A road map,” Journal attack the cancer cells. It will be beneficial to combine these for Immunoerapy of Cancer,vol.5,no.1,2017. drugs with conventional cancer therapies to achieve better [14] M. Banach, C. Serban, A. Sahebkar et al., “Impact of statin outcomes in cancer patients. therapy on coronary plaque composition: a systematic review and meta-analysis of virtual histology intravascular ultrasound studies,” BMC Medicine,vol.13,no. 1,2015. Conflicts of Interest [15] G. Sarrabayrouse, C. Pich, I. Teiti, and A. F. Tilkin-Mariame, “Regulatory properties of statins and rho gtpases prenylation The authors report no conifl cts of interest. inhibitiors to stimulate melanoma immunogenicity and pro- mote anti-melanoma immune response,” International Journal Acknowledgments of Cancer, vol.140,no. 4,pp. 747–755, 2017. [16] V. Ziegler, C. Henninger, I. Simiantonakis et al., “Rho inhibition This work received funding from Keio Gijuku Fukuzawa by lovastatin affects apoptosis and DSB repair of primary Memorial Fund for the Advancement of Education and human lung cells in vitro and lung tissue in vivo following Research. We are thankful to Ms. Saori Matsumoto, Mr. Sho fractionated irradiation,” Cell Death & Disease,vol. 8,no. 8,p. Kashiwazaki, and Mr. Michio Kobori for their suggestions e2978, 2017. about drugs related to immunogenic cell death. We are also [17] R. Palomino-Morales, S. Perales, C. Torres, A. Linares, and thankful to Dr. Yutaka Hattori and Dr. Daiju Ichikawa for M. J. Alejandre, “Eec ff t of HMG-CoA reductase inhibition on their valuable feedbacks. vascular smooth muscle cells extracellular matrix production: Role of RhoA,” Current Vascular Pharmacology,vol. 14, no. 4, pp. 345–352, 2016. References [18] M.Islam, S. Sharma, B.Kumar,and T.N. Teknos,“Atorvastatin [1] D. Latinne and R. Fiasse, “New insights into the cellular inhibits RhoC function and limits head and neck cancer immunology of the intestine in relation to the pathophysiology metastasis,” Oral Oncology,vol.49, no.8,pp.778–786, 2013. 6 Journal of Oncology [19] J. Cote-Daigneault, S. Mehandru, R. Ungaro, A. Atreja, and J.- Cancer,” World Journal of Surgery, vol.41, no.9, pp.2361–2370, F. Colombel, “Potential Immunomodulatory Eeff cts of Statins 2017. in Inflammatory Bowel Disease,” Inflammatory Bowel Diseases, [35] K. Oura, T. Tadokoro, S. Fujihara et al., “Telmisartan inhibits vol. 22, no. 3, pp. 724–732, 2015. hepatocellular carcinoma cell proliferation in vitro by inducing [20] G. Sarrabayrouse, C. Pich, R. Moriez et al., “Melanoma cells cell cycle arrest,” Oncology Reports, vol.38, no.5, pp. 2825–2835, treated with GGTI and IFN-𝛾 allow murine vaccination and 2017. enhance cytotoxic response against human melanoma cells,” [36] S. Fujihara, A. Morishita, K. Ogawa et al., “eTh angiotensin II PLoS ONE, vol.5,no.2,2010. type 1 receptor antagonist telmisartan inhibits cell proliferation [21] T. J. Vogel, M. T. Goodman, A. J. Li, and C. Y. Jeon, “Statin treat- and tumor growth of esophageal adenocarcinoma via the AMPKalpha/mTOR pathway in vitro and in vivo,” Oncotarget, ment is associated with survival in a nationally representative population of elderly women with epithelial ovarian cancer,” vol.8,no.5,2017. Gynecologic Oncology,vol.146,no.2,pp.340–345, 2017. [37] S. Saikawa, K. Kaji, N. Nishimura et al., “Angiotensin receptor [22] A. Wang, H. A. Wakelee, A. K. Aragaki et al., “Protective Eec ff ts blockade attenuates cholangiocarcinoma cell growth by inhibit- ing the oncogenic activity of Yes-associated protein,” Cancer of Statins in Cancer: Should eTh y Be Prescribed for High-Risk Patients?” Current Atherosclerosis Reports,vol.18, no. 12,2016. Letters, vol. 434, pp. 120–129, 2018. [38] R.Coulson, S.H. Liew,A.A.Connelly,and etal., “eTh [23] P. Desai, R. Wallace, and M. Anderson, “An analysis of the effect of statins on the risk of Non-Hodgkin’s Lymphoma in the angiotensin receptor blocker, losartan, inhibits mammary tumor development and progression to invasive carcinoma,” Women’s Health Initiative cohort,” Cancer Medicine,vol.7, no. 5, pp. 2121–2130, 2018. Oncotarget, vol. 8, pp. 18640–18656, 2017. [39] K. Nakamura, T. Yaguchi, G. Ohmura et al., “Involvement [24] R. J. Shaw, K. A. Lamia, D. Vasquez et al., “Medicine: the kinase LKB1 mediates glucose homeostasis in liver and therapeutic of local renin-angiotensin system in immunosuppression of tumor microenvironment,” Cancer Science, vol.109,no.1, pp. effects of metformin,” Science,vol. 310,no. 5754, pp.1642–1646, 2005. 54–64, 2018. [40] C. S.Chim,S.K.Kumar, R. Z. Orlowskiet al., “Management [25] G.Rena, D.G.Hardie, and E. R.Pearson, “eTh mechanisms of action of metformin,” Diabetologia,vol.60, no.9,pp.1577–1585, of relapsed and refractory multiple myeloma: Novel agents, antibodies, immunotherapies and beyond,” Leukemia,vol. 32, no. 2, pp. 252–262, 2018. [26] S.Eikawa,M.Nishida,S.Mizukami,C.Yamazaki,E.Nakayama, [41] P. Moreau, “How I treat myeloma with new agents,” Blood,vol. and H. Udono, “Immune-mediated antitumor effect by type 2 130, no. 13, pp. 1507–1513, 2017. diabetes drug, metformin,” Proceedings of the National Acadamy of Sciences of the United States of America, vol. 112, no. 6, pp. [42] B. Pan and S. Lentzsch, “The application and biology of 1809–1814, 2015. immunomodulatory drugs (IMiDs) in cancer,” Pharmacology & erapeutics,vol.136, no.1,pp. 56–68, 2012. [27] F. V. Pereira, A. C. Melo, J. S. Low et al., “Metformin exerts antitumor activity via induction of multiple death pathways in [43] J. B. Zeldis, R. Knight, M. Hussein, R. Chopra, and G. tumor cells and activation of a protective immune response,” Muller, “A review of the history, properties, and use of the Oncotarget ,vol.9, no. 40,2018. immunomodulatory compound lenalidomide,” Annals of the New York Academy of Sciences, vol. 1222, no. 1, pp. 76–82, 2011. [28] J. Talairach and P. Thournoux, “Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation [44] K. Luptakova, J. Rosenblatt, B. Glotzbecker et al., “Lenalidomide of PD-L1,” Mollecular Cell, vol.71,pp.606–620, 2018. enhances anti-myeloma cellular immunity,” Cancer Immunol- ogy, Immunotherapy,vol.62,no. 1, pp.39–49, 2013. [29] D. Di Fusco, V. Dinallo, I. Monteleone et al., “Metformin inhibits inflammatory signals in the gut by controlling AMPK and p38 [45] H. Quach, D. Ritchie, A. K. Stewart et al., “Mechanism of action MAP kinase activation,” Clinical Science,vol.132,no. 11, pp. of immunomodulatory drugs (IMiDS) in multiple myeloma,” 1155–1168, 2018. Leukemia,vol. 24, no.1, pp. 22–32,2010. [30] H. J. Son, J. Lee, S. Y. Lee, and etal., “Metformin attenuates [46] A. K. Hsu, H. Quach, T. Tai et al., “e Th immunostimulatory experimental autoimmune arthritis through reciprocal regula- eeff ct of lenalidomide on NK-cell function is profoundly inhib- tion of Th17/Treg balance and osteoclastogenesis,” Mediators of ited by concurrent dexamethasone therapy,” Blood,vol.117,no. Inflammation,vol.2014,pp.1–13, 2014. 5, pp. 1605–1613, 2011. [31] J. Rodriguez, S. Hiel, and N. M. Delzenne, “Metformin: Old [47] M. Obeid, A. Tesniere, F. Ghiringhelli et al., “Calreticulin friend, new ways of action-implication of the gut microbiome?” exposure dictates the immunogenicity of cancer cell death,” Current Opinion in Clinical Nutrition & Metabolic Care,vol.21, Nature Medicine,vol.13,no.1,pp.54–61, 2007. no.4,pp.294–301,2018. [48] J. W. Hodge, C. T. Garnett, B. Farsaci et al., “Chemotherapy- [32] A. J. George, W. G. o Th mas, and R. D. Hannan, “eTh renin- induced immunogenic modulation of tumor cells enhances angiotensin system and cancer: old dog, new tricks,” Nature killing by cytotoxic T lymphocytes and is distinct from Reviews Cancer, vol. 10, no. 11, pp. 745–759, 2010. immunogenic cell death,” International Journal of Cancer,vol. ´ 133, no.3,pp.624–636, 2013. [33] J. Busby, U. McMenamin, A. Spence, B. T. Johnston, C. Hughes, and C. R. Cardwell, “Angiotensin receptor blocker use and [49] E. Vacchelli, Y. Ma, E. E. Baracco et al., “Chemotherapy- gastro-oesophageal cancer survival: a population-based cohort induced antitumor immunity requires formyl peptide receptor study,” Alimentary Pharmacology & erapeutics,vol.47, no.2, 1,” Science,vol.350, pp.972–978,2015. pp. 279–288, 2018. [50] A. Sistigu, T. Yamazaki, E. Vacchelli et al., “Cancer cell- [34] M. Cerullo,F.Gani, S. Y. Chen, J.K. Canner,and T. M.Pawlik, autonomous contribution of type I interferon signaling to the “Impact of Angiotensin Receptor Blocker Use on Overall efficacy of chemotherapy,” Nature Medicine,vol.20, no. 11, pp. Survival Among Patients Undergoing Resection for Pancreatic 1301–1309, 2014. Journal of Oncology 7 [51] L. Galluzzi, A. Buque, ´ O. Kepp, L. Zitvogel, and G. Kroemer, [68] M. Matsushita, Y. Otsuka, N. Tsutsumida et al., “Identification “Immunogenic cell death in cancer and infectious disease,” of Novel HLA-A*24:02-Restricted Epitope Derived from a Nature Reviews Immunology, vol. 17, no. 2, pp. 97–111, 2017. Homeobox Protein Expressed in Hematological Malignancies,” PLoS ONE,vol.11,no.1,p.e0146371, 2016. [52] G. Kroemer, L. Galluzzi, O. Kepp, and L. Zitvogel, “Immuno- genic cell death in cancer therapy,” Annual Review of Immunol- [69] P. Srivastava, B. E. Paluch, J. Matsuzaki et al., “Induction of ogy,vol.31,pp. 51–72, 2013. cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monother- [53] S. Gebremeskel and B. Johnston, “Concepts and mechanisms underlying chemotherapy induced immunogenic cell death: apy,” Oncotarget ,vol.7,no.11,pp. 12840–12856,2016. Impact on clinical studies and considerations for combined [70] M. L. Stone, K. B. Chiappinelli, H. Li et al., “Epigenetic therapy therapies,” Oncotarget ,vol.6,no.39,pp.41600–41619,2015. activates type I interferon signaling in murine ovarian cancer to [54] N. Casares, M. O. Pequignot, A. Tesniere et al., “Caspase- reduce immunosuppression and tumor burden,” Proceedings of the National Acadamy of Sciences of the United States of America, dependent immunogenicity of doxorubicin-induced tumor cell death,” e Journal of Experimental Medicine,vol.202, no.12, vol. 114, no. 51, pp. E10981–E10990, 2017. pp. 1691–1701, 2005. [55] A. Tesniere, F. Schlemmer, V. Boige et al., “Immunogenic death of colon cancer cells treated with oxaliplatin,” Oncogene,vol. 29, no.4,pp.482–491, 2010. [56] X. Lu, Z.-C. Ding, Y. Cao et al., “Alkylating agent melphalan augments the efficacy of adoptive immunotherapy using tumor- specific CD4+ T cells,” e Journal of Immunology,vol. 194, no. 4, pp. 2011–2021, 2015. [57] J. Fucikova, I. Truxova, M. Hensler et al., “Calreticulin exposure by malignant blasts correlates with robust anticancer immunity and improved clinical outcome in AML patients,” Blood,vol. 128, no. 26, pp. 3113–3124, 2016. [58] C. Pozzi,A.Cuomo,I. Spadoni et al., “eE Th GFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death,” Nature Medicine,vol.22,no.6,pp. 624–631, 2016. [59] L.Bezu,L.C.Gomes-de-Silva,H.Dewitteetal.,“Combinatorial Strategies for the Induction of Immunogenic Cell Death,” Frontiers in Immunology, vol.6,2015. [60] C.Pfirschke, C.Engblom, S.Rickelt et al., “Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy,” Immunity,vol. 44, no.2, pp. 343–354,2016. [61] B. Montico, A. Nigro, V. Casolaro, and J. Dal Col, “Immuno- genic Apoptosis as a Novel Tool for Anticancer Vaccine Devel- opment,” International Journal of Molecular Sciences,vol. 19, no. 2, p. 594, 2018. [62] A. D.Garg,S.More, N. Rufo et al., “Trial watch: Immuno- genic cell death induction by anticancer chemotherapeutics,” OncoImmunology,vol.6,no. 12, p.e1386829, 2017. [63] L. Ding, MH. Bailey, and E. Porta-Pardo, “Perspective on oncogenic processes at the end of the beginning of cancer genomics,” Cell, vol.173,pp.305–320, 2018. [64] E. J. B.Derissen,J. H.Beijnen,and J. H. M. Schellens,“Concise drug review: Azacitidine and decitabine,” e Oncologist,vol.18, no.5,pp.619–624, 2013. [65] T.Schroeder, C.Rautenberg, R.Haas, U.Germing, and G. Kobbe, “Hypomethylating agents for treatment and prevention of relapse aeft r allogeneic blood stem cell transplantation,” International Journal of Hematology,vol. 107, no.2,pp.138–150, [66] Y. Chen, P. L. McCarthy, T. Hahn et al., “Methods to prevent and treat relapse aer ft hematopoietic stem cell transplantation with tyrosine kinase inhibitors, immunomodulating drugs, deacety- lase inhibitors, and hypomethylating agents,” Bone Marrow Transplantation. [67] N. Luo, M. J. Nixon, P. I. Gonzalez-Ericsson et al., “DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer,” Nature Communications, vol.9,no. 1, 2018. MEDIATORS of INFLAMMATION The Scientific Gastroenterology Journal of World Journal Research and Practice Diabetes Research Disease Markers Hindawi Hindawi Publishing Corporation Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 http://www www.hindawi.com .hindawi.com V Volume 2018 olume 2013 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 International Journal of Journal of Immunology Research Endocrinology Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Submit your manuscripts at www.hindawi.com BioMed PPAR Research Research International Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 Journal of Obesity Evidence-Based Journal of Journal of Stem Cells Complementary and Ophthalmology International Alternative Medicine Oncology Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2013 Parkinson’s Disease Computational and Behavioural Mathematical Methods AIDS Oxidative Medicine and in Medicine Neurology Research and Treatment Cellular Longevity Hindawi Hindawi Hindawi Hindawi Hindawi www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018 www.hindawi.com Volume 2018

Journal

Journal of OncologyHindawi Publishing Corporation

Published: Oct 25, 2018

There are no references for this article.