Repurposing of Drugs for Immunotherapy
Jyothishmathi Swaminathan
Departments of Pediatrics and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
Search for more papers by this authorVidya Gopalakrishnan
Departments of Pediatrics and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
Search for more papers by this authorJyothishmathi Swaminathan
Departments of Pediatrics and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
Search for more papers by this authorVidya Gopalakrishnan
Departments of Pediatrics and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
Search for more papers by this authorLaurence J. N. Cooper MD, PhD
Search for more papers by this authorElizabeth A. Mittendorf MD, PhD
Search for more papers by this authorJudy Moyes MB, BChir, FRCP(C), FRCPCH
Search for more papers by this authorSabitha Prabhakaran PhD
Search for more papers by this authorSummary
The increasing rate of cancer diagnosis and mortality has created an augmented need for new drugs and innovative strategies to battle cancer. During the pregenomics era, drug design was geared towards promoting cytotoxicity to the tumor cells, which due to lack of precision, killed a number of surrounding, healthy cells. Then came more rationally designed, target-based drugs that relied on the identification of signaling pathways that are deregulated in a specific type of cancer. Of these, immunotherapy was viewed as one of the best strategies to combat the disease, but it has proved difficult to achieve its maximum capability. Developing new drugs is cost prohibitive, but repurposing existing ones has proven to be promising and cost effective. In this review, we discuss some of the popular existing drugs that have immunomodulatory properties and therefore the potential for repurposing as immunotherapy against cancers.
References
- Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011; 480(7378): 480–9.
- Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015; 161(2): 205–14.
- Topalian SL, Weiner GJ, Pardoll DM. Cancer immunotherapy comes of age. J Clin Oncol. 2011; 29(36): 4828–36.
- Gilham DE, Anderson J, Bridgeman JS, Hawkins RE, Exley MA, Stauss H, et al. Adoptive T cell therapy for cancer in the United kingdom: a review of activity for the British Society of Gene and Cell Therapy annual meeting 2015. Human Gene Therapy. 2015; 26(5): 276–85.
- Hinrichs CS, Rosenberg SA. Exploiting the curative potential of adoptive T cell therapy for cancer. Immunol Rev. 2014; 257(1): 56–71.
- Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. Gene therapy with human and mouse T cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009; 114(3): 535–46.
- Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011; 118(23): 6050–6.
- Maher J. Immunotherapy of malignant disease using chimeric antigen receptor engrafted T cells. ISRN Oncology. 2012; 2012: 278093.
- Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. New England Journal of Medicine. 2014; 371(16): 1507–17.
- Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014; 123(17): 2625–35.
- Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011; 29(7): 917–24.
- Pardoll DM, Topalian SL. The role of CD4+ T cell responses in antitumor immunity. Current Opinion in Immunology. 1998; 10(5): 588–94.
- Cheng M, Chen Y, Xiao W, Sun R, Tian Z. NK cell-based immunotherapy for malignant diseases. Cellular & Molecular Immunology. 2013; 10(3): 230–52.
- Webster RM. Combination therapies in oncology. Nature Reviews Drug Discovery. 2016; 15(2): 81–2.
- Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5): 646–74.
- Baylin SB, Jones PA. A decade of exploring the cancer epigenome: biological and translational implications. Nature Reviews Cancer. 2011; 11(10): 726–34.
- Kulis M, Esteller M. DNA methylation and cancer. Advances in Genetics. 2010; 70: 27–56.
- Esteller M. Epigenetics in cancer. New England Journal of Medicine. 2008; 358(11): 1148–59.
- Rodriguez-Paredes M, Esteller M. Cancer epigenetics reaches mainstream oncology. Nature Medicine. 2011; 17(3): 330–9.
- Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. International Journal of Cancer. 2008; 123(1): 8–13.
- Derissen EJ, Beijnen JH, Schellens JH. Concise drug review: azacitidine and decitabine. The Oncologist. 2013; 18(5): 619–24.
- Bubenik J. MHC class I down-regulation: tumour escape from immune surveillance? (review). International Journal of Oncology. 2004; 25(2): 487–91.
- Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, et al. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Molecular Oncology. 2011; 5(2): 164–82.
- Wang L, Amoozgar Z, Huang J, Saleh MH, Xing D, Orsulic S, et al. Decitabine enhances lymphocyte migration and function and synergizes with CTLA-4 blockade in a murine ovarian cancer model. Cancer Immunology Research. 2015; 3(9): 1030–41.
- Salmaninejad A, Zamani MR, Pourvahedi M, Golchehre Z, Hosseini Bereshneh A, Rezaei N. Cancer/testis antigens: expression, regulation, tumor invasion, and use in immunotherapy of cancers. Immunological Investigations. 2016; 45(7): 619–40.
- Atanackovic D, Luetkens T, Kloth B, Fuchs G, Cao Y, Hildebrandt Y, et al. Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. American Journal of Hematology. 2011; 86(11): 918–22.
- Maio M, Coral S, Fratta E, Altomonte M, Sigalotti L. Epigenetic targets for immune intervention in human malignancies. Oncogene. 2003; 22(42): 6484–8.
- Tomasi TB, Magner WJ, Khan AN. Epigenetic regulation of immune escape genes in cancer. Cancer Immunology, Immunotherapy: CII. 2006; 55(10): 1159–84.
- Dubovsky JA, McNeel DG, Powers JJ, Gordon J, Sotomayor EM, Pinilla-Ibarz JA. Treatment of chronic lymphocytic leukemia with a hypomethylating agent induces expression of NXF2, an immunogenic cancer testis antigen. Clinical Cancer Research. 2009; 15(10): 3406–15.
- Wu X, Tao Y, Hou J, Meng X, Shi J. Valproic acid upregulates NKG2D ligand expression through an ERK-dependent mechanism and potentially enhances NK cell-mediated lysis of myeloma. Neoplasia. 2012; 14(12): 1178–89.
- Tang KF, He CX, Zeng GL, Wu J, Song GB, Shi YS, et al. Induction of MHC class I-related chain B (MICB) by 5-aza-2'-deoxycytidine. Biochemical and Biophysical Research Communications. 2008; 370(4): 578–83.
- Fonsatti E, Nicolay HJ, Sigalotti L, Calabro L, Pezzani L, Colizzi F, et al. Functional up-regulation of human leukocyte antigen class I antigens expression by 5-aza-2'-deoxycytidine in cutaneous melanoma: immunotherapeutic implications. Clinical Cancer Research. 2007; 13(11): 3333–8.
- Adair SJ, Hogan KT. Treatment of ovarian cancer cell lines with 5-aza-2'-deoxycytidine upregulates the expression of cancer-testis antigens and class I major histocompatibility complex-encoded molecules. Cancer Immunology, Immunotherapy. 2009; 58(4): 589–601.
- Kersh EN, Fitzpatrick DR, Murali-Krishna K, Shires J, Speck SH, Boss JM, et al. Rapid demethylation of the IFN-gamma gene occurs in memory but not naive CD8 T cells. Journal of Immunology. 2006; 176(7): 4083–93.
- Frikeche J, Clavert A, Delaunay J, Brissot E, Gregoire M, Gaugler B, et al. Impact of the hypomethylating agent 5-azacytidine on dendritic cells function. Experimental Hematology. 2011; 39(11): 1056–63.
- Janson PC, Linton LB, Bergman EA, Marits P, Eberhardson M, Piehl F, et al. Profiling of CD4+ T cells with epigenetic immune lineage analysis. Journal of Immunology. 2011; 186(1): 92–102.
- Dubovsky JA, Powers JJ, Gao Y, Mariusso LF, Sotomayor EM, Pinilla-Ibarz JA. Epigenetic repolarization of T lymphocytes from chronic lymphocytic leukemia patients using 5-aza-2'-deoxycytidine. Leukemia Research. 2011; 35(9): 1193–9.
- Costantini B, Kordasti SY, Kulasekararaj AG, Jiang J, Seidl T, Abellan PP, et al. The effects of 5-azacytidine on the function and number of regulatory T cells and T-effectors in myelodysplastic syndrome. Haematologica. 2013; 98(8): 1196–205.
- Schmiedel BJ, Arelin V, Gruenebach F, Krusch M, Schmidt SM, Salih HR. Azacytidine impairs NK cell reactivity while decitabine augments NK cell responsiveness toward stimulation. International Journal of Cancer. 2011; 128(12): 2911–22.
- Chan HW, Kurago ZB, Stewart CA, Wilson MJ, Martin MP, Mace BE, et al. DNA methylation maintains allele-specific KIR gene expression in human natural killer cells. The Journal of Experimental Medicine. 2003; 197(2): 245–55.
- Daurkin I, Eruslanov E, Vieweg J, Kusmartsev S. Generation of antigen-presenting cells from tumor-infiltrated CD11b myeloid cells with DNA demethylating agent 5-aza-2'-deoxycytidine. Cancer Immunology, Immunotherapy. 2010; 59(5): 697–706.
- Triozzi PL, Aldrich W, Achberger S, Ponnazhagan S, Alcazar O, Saunthararajah Y. Differential effects of low-dose decitabine on immune effector and suppressor responses in melanoma-bearing mice. Cancer Immunology, Immunotherapy. 2012; 61(9): 1441–50.
- Fan H, Lu X, Wang X, Liu Y, Guo B, Zhang Y, et al. Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a phase I/II report. Journal of Immunology Research. 2014; 2014: 371087.
- Krishnadas DK, Shusterman S, Bai F, Diller L, Sullivan JE, Cheerva AC, et al. A phase I trial combining decitabine/dendritic cell vaccine targeting MAGE-A1, MAGE-A3 and NY-ESO-1 for children with relapsed or therapy-refractory neuroblastoma and sarcoma. Cancer Immunology, Immunotherapy. 2015; 64(10): 1251–60.
- Weintraub K. Take two: combining immunotherapy with epigenetic drugs to tackle cancer. Nature Medicine. 2016; 22(1): 8–10.
- Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. New England Journal of Medicine. 2015; 373(1): 23–34.
- Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011; 21(3): 381–95.
- Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst. 2000; 92(15): 1210–6.
- Kroesen M, Gielen P, Brok IC, Armandari I, Hoogerbrugge PM, Adema GJ. HDAC inhibitors and immunotherapy: a double edged sword? Oncotarget. 2014; 5(16): 6558–72.
- Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules. 2015; 20(3): 3898–941.
- Falkenberg KJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nature Reviews Drug Discovery. 2014; 13(9): 673–91.
- West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Invest. 2014; 124(1): 30–9.
- Akhtar MW, Raingo J, Nelson ED, Montgomery RL, Olson EN, Kavalali ET, et al. Histone deacetylases 1 and 2 form a developmental switch that controls excitatory synapse maturation and function. J Neurosci. 2009; 29(25): 8288–97.
- Graff J, Rei D, Guan JS, Wang WY, Seo J, Hennig KM, et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature. 2012; 483(7388): 222–6.
- Majdzadeh N, Morrison BE, D'Mello SR. Class IIA HDACs in the regulation of neurodegeneration. Front Biosci. 2008; 13: 1072–82.
- Montgomery RL, Hsieh J, Barbosa AC, Richardson JA, Olson EN. Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc Natl Acad Sci U S A. 2009; 106(19): 7876–81.
- Nebbioso A, Carafa V, Benedetti R, Altucci L. Trials with 'epigenetic' drugs: an update. Molecular Oncology. 2012; 6(6): 657–82.
- New M, Olzscha H, La Thangue NB. HDAC inhibitor-based therapies: can we interpret the code? Molecular Oncology. 2012; 6(6): 637–56.
- Qiu T, Zhou L, Zhu W, Wang T, Wang J, Shu Y, et al. Effects of treatment with histone deacetylase inhibitors in solid tumors: a review based on 30 clinical trials. Future Oncol. 2013; 9(2): 255–69.
- Yang S, Kittlesen D, Slingluff CL, Jr., Vervaert CE, Seigler HF, Darrow TL. Dendritic cells infected with a vaccinia vector carrying the human gp100 gene simultaneously present multiple specificities and elicit high-affinity T cells reactive to multiple epitopes and restricted by HLA-A2 and -A3. Journal of Immunology. 2000; 164(8): 4204–11.
- Lapointe R, Royal RE, Reeves ME, Altomare I, Robbins PF, Hwu P. Retrovirally transduced human dendritic cells can generate T cells recognizing multiple MHC class I and class II epitopes from the melanoma antigen glycoprotein 100. Journal of Immunology (Baltimore, Md: 1950). 2001; 167(8): 4758–64.
- Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. New England Journal of Medicine. 2011; 364(22): 2119–27.
- Li W, Joshi MD, Singhania S, Ramsey KH, Murthy AK. Peptide vaccine: progress and challenges. Vaccines (Basel). 2014; 2(3): 515–36.
- Murakami T, Sato A, Chun NA, Hara M, Naito Y, Kobayashi Y, et al. Transcriptional modulation using HDACi depsipeptide promotes immune cell-mediated tumor destruction of murine B16 melanoma. J Invest Dermatol. 2008; 128(6): 1506–16.
- Garrido F, Ruiz-Cabello F, Cabrera T, Perez-Villar JJ, Lopez-Botet M, Duggan-Keen M, et al. Implications for immunosurveillance of altered HLA class I phenotypes in human tumours. Immunol Today. 1997; 18(2): 89–95.
- Manning J, Indrova M, Lubyova B, Pribylova H, Bieblova J, Hejnar J, et al. Induction of MHC class I molecule cell surface expression and epigenetic activation of antigen-processing machinery components in a murine model for human papilloma virus 16-associated tumours. Immunology. 2008; 123(2): 218–27.
- Setiadi AF, David MD, Seipp RP, Hartikainen JA, Gopaul R, Jefferies WA. Epigenetic control of the immune escape mechanisms in malignant carcinomas. Mol Cell Biol. 2007; 27(22): 7886–94.
- Woods DM, Woan K, Cheng F, Wang H, Perez-Villarroel P, Lee C, et al. The antimelanoma activity of the histone deacetylase inhibitor panobinostat (LBH589) is mediated by direct tumor cytotoxicity and increased tumor immunogenicity. Melanoma Res. 2013; 23(5): 341–8.
- Northrop JK, Thomas RM, Wells AD, Shen H. Epigenetic remodeling of the IL-2 and IFN-gamma loci in memory CD8 T cells is influenced by CD4 T cells. Journal of Immunology. 2006; 177(2): 1062–9.
- Zhang F, Zhou X, DiSpirito JR, Wang C, Wang Y, Shen H. Epigenetic manipulation restores functions of defective CD8(+) T cells from chronic viral infection. Mol Ther. 2014; 22(9): 1698–706.
- Wang D, Iclozan C, Liu C, Xia C, Anasetti C, Yu XZ. LBH589 enhances T cell activation in vivo and accelerates graft-versus-host disease in mice. Biol Blood Marrow Transplant. 2012; 18(8): 1182–90 e1.
- Hopewell EL, Zhao W, Fulp WJ, Bronk CC, Lopez AS, Massengill M, et al. Lung tumor NF-kappaB signaling promotes T cell-mediated immune surveillance. J Clin Invest. 2013; 123(6): 2509–22.
- Pages F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. New England Journal of Medicine. 2005; 353(25): 2654–66.
- Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. New England Journal of Medicine. 2003; 348(3): 203–13.
- Zheng H, Zhao W, Yan C, Watson CC, Massengill M, Xie M, et al. HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma. Clinical Cancer Research. 2016; 22(16): 4119–32.
- Gupta P, Reid RC, Iyer A, Sweet MJ, Fairlie DP. Towards isozyme-selective HDAC inhibitors for interrogating disease. Curr Top Med Chem. 2012; 12(14): 1479–99.
- Halili MA, Andrews MR, Sweet MJ, Fairlie DP. Histone deacetylase inhibitors in inflammatory disease. Curr Top Med Chem. 2009; 9(3): 309–19.
- Lohman RJ, Iyer A, Fairlie TJ, Cotterell A, Gupta P, Reid RC, et al. Differential anti-inflammatory activity of HDAC inhibitors in human macrophages and rat arthritis. J Pharmacol Exp Ther. 2016; 356(2): 387–96.
- Wu Y, Guo SW. Suppression of IL-1beta-induced COX-2 expression by trichostatin A (TSA) in human endometrial stromal cells. Eur J Obstet Gynecol Reprod Biol. 2007; 135(1): 88–93.
- Wu Y, Guo SW. Peroxisome proliferator-activated receptor-gamma and retinoid X receptor agonists synergistically suppress proliferation of immortalized endometrial stromal cells. Fertil Steril. 2009; 91(5 Suppl): 2142–7.
- Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nature Medicine. 2007; 13(11): 1299–307.
- Avogadri F, Yuan J, Yang A, Schaer D, Wolchok JD. Modulation of CTLA-4 and GITR for cancer immunotherapy. Curr Top Microbiol Immunol. 2011; 344: 211–44.
- Lucas JL, Mirshahpanah P, Haas-Stapleton E, Asadullah K, Zollner TM, Numerof RP. Induction of Foxp3+ regulatory T cells with histone deacetylase inhibitors. Cell Immunol. 2009; 257(1–2): 97–104.
- Zhang ZY, Schluesener HJ. Oral administration of histone deacetylase inhibitor MS-275 ameliorates neuroinflammation and cerebral amyloidosis and improves behavior in a mouse model. J Neuropathol Exp Neurol. 2013; 72(3): 178–85.
- Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. New England Journal of Medicine. 2010; 363(8): 711–23.
- Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002; 99(19): 12293–7.
- Cheng F, Lienlaf M, Perez-Villarroel P, Wang HW, Lee C, Woan K, et al. Divergent roles of histone deacetylase 6 (HDAC6) and histone deacetylase 11 (HDAC11) on the transcriptional regulation of IL10 in antigen presenting cells. Molecular Immunology. 2014; 60(1): 44–53.
- Lienlaf M, Perez-Villarroel P, Knox T, Pabon M, Sahakian E, Powers J, et al. Essential role of HDAC6 in the regulation of PD-L1 in melanoma. Molecular Oncology. 2016; 10(5): 735–50.
- Wang D, DuBois RN. The role of prostaglandin E2 in tumor-associated immunosuppression. Trends in Molecular Medicine. 2016; 22(1): 1–3.
- Harris RE. Cyclooxygenase-2 (COX-2) blockade in the chemoprevention of cancers of the colon, breast, prostate, and lung. Inflammopharmacology. 2009; 17: 55–67.
- Crusz SM, Balkwill FR. Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol. 2015; 12(10): 584–96.
- Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol. 1971; 231(25): 232–5.
- Zha S, Yegnasubramanian V, Nelson WG, Isaacs WB, De Marzo AM. Cyclooxygenases in cancer: progress and perspective. Cancer Lett. 2004; 215(1): 1–20.
- Wang D, Dubois RN. Eicosanoids and cancer. Nature Reviews Cancer. 2010; 10(3): 181–93.
- Zelenay S, van der Veen AG, Bottcher JP, Snelgrove KJ, Rogers N, Acton SE, et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell. 2015; 162(6): 1257–70.
- Becker MR, Siegelin MD, Rompel R, Enk AH, Gaiser T. COX-2 expression in malignant melanoma: a novel prognostic marker? Melanoma Res. 2009; 19(1): 8–16.
- Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJ, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014; 26(5): 638–52.
- Chen JH, Perry CJ, Tsui YC, Staron MM, Parish IA, Dominguez CX, et al. Prostaglandin E2 and programmed cell death 1 signaling coordinately impair CTL function and survival during chronic viral infection. Nature Medicine. 2015; 21(4): 327–34.
- Wang D, DuBois RN. Immunosuppression associated with chronic inflammation in the tumor microenvironment. Carcinogenesis. 2015; 36(10): 1085–93.
- Kyi C, Postow MA. Checkpoint blocking antibodies in cancer immunotherapy. FEBS Letters. 2014; 588(2): 368–76.
- Balch CM, Dougherty PA, Cloud GA, Tilden AB. Prostaglandin E2-mediated suppression of cellular immunity in colon cancer patients. Surgery. 1984; 95(1): 71–7.
- Ohshika Y, Umesaki N, Sugawa T. Immunomodulating capacity of the monocyte-macrophage system in patients with uterine cervical cancer. Nihon Sanka Fujinka Gakkai Zasshi. 1988; 40(5): 601–8.
- Cameron DJ, O'Brien P. Relationship of the suppression of macrophage mediated tumor cytotoxicity in conjunction with secretion of prostaglandin from the macrophages of breast cancer patients. Int J Immunopharmacol. 1982; 4(5): 445–50.
-
Cameron DJ, Stromberg BV. The ability of macrophages from head and neck cancer patients to kill tumor cells. Effect of prostaglandin inhibitors on cytotoxicity. Cancer. 1984; 54(11): 2403–8.
10.1002/1097-0142(19841201)54:11<2403::AID-CNCR2820541116>3.0.CO;2-F PubMed Web of Science® Google Scholar
-
Baxevanis CN, Reclos GJ, Gritzapis AD, Dedousis GV, Missitzis I, Papamichail M. Elevated prostaglandin E2 production by monocytes is responsible for the depressed levels of natural killer and lymphokine-activated killer cell function in patients with breast cancer. Cancer. 1993; 72(2): 491–501.
10.1002/1097-0142(19930715)72:2<491::AID-CNCR2820720227>3.0.CO;2-1 CAS PubMed Web of Science® Google Scholar
- Kundu N, Walser TC, Ma X, Fulton AM. Cyclooxygenase inhibitors modulate NK activities that control metastatic disease. Cancer Immunology, Immunotherapy. 2005; 54(10): 981–7.
- Shparyk Ia V. [The dynamic indices of the natural killer cells in the chemotherapy of patients with digestive organ cancer]. Vrach Delo. 1990(7): 14–6.
- Tanaka N, Okamoto Y, Gotoh K, Hizuta A, Yunoki S, Orita K. Combined therapy with interleukin 2 and indomethacin in mice inoculated with MH134 hepatoma. Acta Med Okayama. 1995; 49(5): 241–5.
- Zamai L, Ponti C, Mirandola P, Gobbi G, Papa S, Galeotti L, et al. NK cells and cancer. Journal of Immunology. 2007; 178(7): 4011–6.
- Lang S, Lauffer L, Clausen C, Lohr I, Schmitt B, Holzel D, et al. Impaired monocyte function in cancer patients: restoration with a cyclooxygenase-2 inhibitor. FASEB J. 2003; 17(2): 286–8.
- Lang S, Tiwari S, Andratschke M, Loehr I, Lauffer L, Bergmann C, et al. Immune restoration in head and neck cancer patients after in vivo COX-2 inhibition. Cancer Immunology, Immunotherapy. 2007; 56(10): 1645–52.
- Schellhorn M, Haustein M, Frank M, Linnebacher M, Hinz B. Celecoxib increases lung cancer cell lysis by lymphokine-activated killer cells via upregulation of ICAM-1. Oncotarget. 2015; 6(36): 39342–56.
- Ikemoto S, Kishimoto T, Iimori H, Morikawa Y, Hayahara N, Maekawa M. Defective interleukin-1 production of monocytes in patients with bladder cancer. Br J Urol. 1990; 65(2): 181–5.
- Petrini B, Wolk G, Wasserman J, Vedin I, Strannegard O, Blomgren H, et al. Indomethacin modulation of monocyte cytokine release following pelvic irradiation for cancer. Eur J Cancer. 1991; 27(5): 591–4.
- Yin T, Wang G, Ye T, Wang Y. Sulindac, a non-steroidal anti-inflammatory drug, mediates breast cancer inhibition as an immune modulator. Sci Rep. 2016; 6: 19534.
-
Han T, Takita H. Indomethacin-mediated enhancement of lymphocyte response to mitogens in healthy subjects and lung cancer patients. Cancer. 1980; 46(11): 2416–20.
10.1002/1097-0142(19801201)46:11<2416::AID-CNCR2820461120>3.0.CO;2-G CAS PubMed Web of Science® Google Scholar
- Blomgren H, Rotstein S, Wasserman J, Petrini B, Hammarstrom S. In vitro capacity of various cyclooxygenase inhibitors to revert immune suppression caused by radiation therapy for breast cancer. Radiother Oncol. 1990; 19(4): 329–35.
- Ferrandina G, Ranelletti FO, Legge F, Salutari V, Martinelli E, Fattorossi A, et al. Celecoxib up-regulates the expression of the zeta chain of T cell receptor complex in tumor-infiltrating lymphocytes in human cervical cancer. Clinical Cancer Research. 2006; 12(7 Pt 1): 2055–60.
- Liu JY, Wu Y, Zhang XS, Yang JL, Li HL, Mao YQ, et al. Single administration of low dose cyclophosphamide augments the antitumor effect of dendritic cell vaccine. Cancer Immunology, Immunotherapy. 2007; 56(10): 1597–604.
- Walmesley AJ, Zweiri J, Christmas SE, Watson AJ. Rofecoxib has different effects on chemokine production in colorectal cancer cells and tumor immune splenocytes. J Immunother. 2007; 30(6): 614–23.
- Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nature Reviews Cancer. 2004; 4(6): 423–36.
- Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabzevari H. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood. 2005; 105(7): 2862–8.
- Ghiringhelli F, Larmonier N, Schmitt E, Parcellier A, Cathelin D, Garrido C, et al. CD4 + CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol. 2004; 34(2): 336–44.
- Tongu M, Harashima N, Yamada T, Harada T, Harada M. Immunogenic chemotherapy with cyclophosphamide and doxorubicin against established murine carcinoma. Cancer Immunology, Immunotherapy. 2010; 59(5): 769–77.
- Kaneno R, Shurin GV, Tourkova IL, Shurin MR. Chemomodulation of human dendritic cell function by antineoplastic agents in low noncytotoxic concentrations. J Transl Med. 2009; 7: 58.
- Veltman JD, Lambers ME, van Nimwegen M, de Jong S, Hendriks RW, Hoogsteden HC, et al. Low-dose cyclophosphamide synergizes with dendritic cell-based immunotherapy in antitumor activity. J Biomed Biotechnol. 2010; 2010: 798467.
- Moschella F, Proietti E, Capone I, Belardelli F. Combination strategies for enhancing the efficacy of immunotherapy in cancer patients. Ann N Y Acad Sci. 2010; 1194: 169–78.
- Radojcic V, Bezak KB, Skarica M, Pletneva MA, Yoshimura K, Schulick RD, et al. Cyclophosphamide resets dendritic cell homeostasis and enhances antitumor immunity through effects that extend beyond regulatory T cell elimination. Cancer Immunology, Immunotherapy. 2010; 59(1): 137–48.
- Nars MS, Kaneno R. Immunomodulatory effects of low dose chemotherapy and perspectives of its combination with immunotherapy. International Journal of Cancer. 2013; 132(11): 2471–8.
- Ramakrishnan R, Assudani D, Nagaraj S, Hunter T, Cho HI, Antonia S, et al. Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice. J Clin Invest. 2010; 120(4): 1111–24.
- Zhong H, Han B, Tourkova IL, Lokshin A, Rosenbloom A, Shurin MR, et al. Low-dose paclitaxel prior to intratumoral dendritic cell vaccine modulates intratumoral cytokine network and lung cancer growth. Clinical Cancer Research. 2007; 13(18 Pt 1): 5455–62.
- Ko HJ, Kim YJ, Kim YS, Chang WS, Ko SY, Chang SY, et al. A combination of chemoimmunotherapies can efficiently break self-tolerance and induce antitumor immunity in a tolerogenic murine tumor model. Cancer Res. 2007; 67(15): 7477–86.
- Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b + myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clinical Cancer Research. 2005; 11(18): 6713–21.
- He Q, Li J, Yin W, Song Z, Zhang Z, Yi T, et al. Low-dose paclitaxel enhances the anti-tumor efficacy of GM-CSF surface-modified whole-tumor-cell vaccine in mouse model of prostate cancer. Cancer Immunology, Immunotherapy. 2011; 60(5): 715–30.
- Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nature Reviews Cancer. 2016; 16(9): 566–81.
- Lisiero DN, Soto H, Everson RG, Liau LM, Prins RM. The histone deacetylase inhibitor, LBH589, promotes the systemic cytokine and effector responses of adoptively transferred CD8+ T cells. J Immunother Cancer. 2014; 2: 8.
- Song DG, Ye Q, Santoro S, Fang C, Best A, Powell DJ, Jr. Chimeric NKG2D CAR-expressing T cell-mediated attack of human ovarian cancer is enhanced by histone deacetylase inhibition. Human Gene Therapy. 2013; 24(3): 295–305.
- Salem ML, Kadima AN, El-Naggar SA, Rubinstein MP, Chen Y, Gillanders WE, et al. Defining the ability of cyclophosphamide preconditioning to enhance the antigen-specific CD8+ T-cell response to peptide vaccination: creation of a beneficial host microenvironment involving type I IFNs and myeloid cells. J Immunother. 2007; 30(1): 40–53.
- Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. Journal of Experimental Medicine. 2005; 202(7): 907–12.
- Ding ZC, Lu X, Yu M, Lemos H, Huang L, Chandler P, et al. Immunosuppressive myeloid cells induced by chemotherapy attenuate antitumor CD4+ T-cell responses through the PD-1-PD-L1 axis. Cancer Res. 2014; 74(13): 3441–53.
- Terranova-Barberio M, Thomas S, Munster PN. Epigenetic modifiers in immunotherapy: a focus on checkpoint inhibitors. Immunotherapy. 2016; 8(6): 705–19.