The Progress of Immunotherapy in Refractory Pituitary Adenomas and Pituitary Carcinomas

Congxin Dai, Siyu Liang, Bowen Sun, Jun Kang, Congxin Dai, Siyu Liang, Bowen Sun, Jun Kang

Abstract

Most pituitary adenomas (PAs) are considered benign tumors, but approximately 0.2% can present metastasis and are classified as pituitary carcinomas (PCs). Refractory PAs lie between benign adenomas and true malignant PC and are defined as aggressive-invasive PAs characterized by a high Ki-67 index, rapid growth, frequent recurrence, and resistance to conventional treatments, including temozolomide. It is notoriously difficult to manage refractory PAs and PC because of the limited therapeutic options. As a promising therapeutic approach, cancer immunotherapy has been experimentally used for the treatment of many tumors, including pituitary tumors. The purpose of this review is to report the progress of immunotherapy in pituitary tumors, including refractory PAs and PCs. The tumor immune microenvironment has been recognized as a key contributor to tumorigenesis, progression, and prognosis. One study indicated that the number of CD68+ macrophages was positively correlated with tumor size and Knosp classification grade for tumor invasiveness. The infiltration of CD4+ and CD8+ T cells was relatively scant in these adenomas, but pituitary growth hormone (GH) adenomas exhibited significantly more CD4+ and CD8+ T cells than non-GH adenomas. These results suggest an association of CD68+ macrophage infiltration with an increase in pituitary tumor size and invasiveness. Another study suggested that a lower number of CD8+ lymphocytes is associated with cavernous sinus invasion and resistance to treatment with first-generation somatostatin analogs in acromegaly patients, highlighting a potential role of the tumor immune microenvironment in determining the prognosis of somatotroph pituitary tumors. Preclinical studies have indicated that widely varying degrees of programmed death-ligand 1 (PD-L1) expression and tumor-infiltrating lymphocytes (TILs) are found among different subtypes. Functional PAs and aggressive PAs express significantly higher levels of PD-L1 and TILs than other subtypes, indicating that PD-1 blockade might be a promising alternative therapy for patients with aggressive PAs. PD-L1 transcript and protein levels were found to be significantly increased in functioning (GH and prolactin-expressing) pituitary tumors compared to nonfunctioning (null cell and silent gonadotroph) adenomas. Moreover, primary pituitary tumors harbored higher levels of PD-L1 mRNA than recurrent tumors. These findings suggest the possibility of considering checkpoint blockade immunotherapy for functioning pituitary tumors refractory to conventional management. Animal models of Cushing's disease also demonstrated PD-L1 and TIL expression in cultured tumors and murine models, as well as the effectiveness of checkpoint blockade therapy in reducing the tumor mass, decreasing hormone secretion, and increasing the survival rate. Clinical studies show that immunotherapy may be an effective treatment in patients with pituitary tumors. One corticotroph carcinoma patient showed a significant reduction in hormone levels and shrinkage of the tumor size of primary and metastatic lesions immediately after investigational treatment with ipilimumab and nivolumab. However, another patient with corticotroph adenoma progressed rapidly after four cycles of anti-PD-1 (pembrolizumab) treatment. To date, there are two registered clinical trials of immunotherapy for pituitary tumors. One of them is the phase II clinical trial of nivolumab combined with ipilimumab for patients with aggressive pituitary tumors (NCT04042753). The other one is also a phase II clinical trial of the combination of nivolumab and ipilimumab for rare tumors, including pituitary tumors (NCT02834013). Both clinical trials are in the stage of recruiting patients and have not been completed. In summary, the results from preclinical research and clinical studies indicated that immunotherapy might be a promising alternative therapy for PCs and refractory PAs resistant to conventional treatments. The combination of immunotherapy and radiotherapy or temozolomide may have synergistic effects compared to a single treatment. More preclinical and clinical studies are needed to further indicate the exact efficacy of immunotherapy in pituitary tumors.

Keywords: immunotherapy; pituitary carcinomas; programmed death-ligand 1; refractory pituitary adenomas; tumor-infiltrating lymphocytes.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2020 Dai, Liang, Sun and Kang.

Figures

Figure 1
Figure 1
Tumor immune microenvironment of pituitary tumors and of Working model of anti-PD1 and anti-anti-CTLA4. The CD4+, CD8+, and CD68+ cells were identified in pituitary tumors, and anti-PD1 and anti-anti-CTLA4 rescue T cell anti-tumor effector functions.

References

    1. Molitch ME. Diagnosis and Treatment of Pituitary Adenomas: A Review. JAMA (2017) 317(5):516–24. 10.1001/jama.2016.19699
    1. Melmed S. Pathogenesis of pituitary tumors. Nat Rev Endocrinol (2011) 7(5):257–66. 10.1038/nrendo.2011.40
    1. Hansen TM, Batra S, Lim M, Gallia GL, Burger PC, Salvatori R, et al. Invasive adenoma and pituitary carcinoma: a SEER database analysis. Neurosurg Rev (2014) 37(2):279–85; discussion 85-6. 10.1007/s10143-014-0525-y
    1. Kasuki L, Raverot G. Definition and diagnosis of aggressive pituitary tumors. Rev Endocr Metab Disord (2020) 21(2):203–8. 10.1007/s11154-019-09531-x
    1. Raverot G, Burman P, McCormack A, Heaney A, Petersenn S, Popovic V, et al. European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol (2018) 178(1):G1–G24. 10.1530/EJE-17-0796
    1. Lopes MBS. The 2017 World Health Organization classification of tumors of the pituitary gland: a summary. Acta Neuropathol (2017) 134(4):521–35. 10.1007/s00401-017-1769-8
    1. Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder M, Grossman AB. Clinical review: Diagnosis and management of pituitary carcinomas. J Clin Endocrinol Metab (2005) 90(5):3089–99. 10.1210/jc.2004-2231
    1. Daly AF, Tichomirowa MA, Beckers A. The epidemiology and genetics of pituitary adenomas. Best Pract Res Clin Endocrinol Metab (2009) 23(5):543–54. 10.1016/j.beem.2009.05.008
    1. Heaney AP. Clinical review: Pituitary carcinoma: difficult diagnosis and treatment. J Clin Endocrinol Metab (2011) 96(12):3649–60. 10.1210/jc.2011-2031
    1. Dai C, Liu X, Ma W, Wang R. The Treatment of Refractory Pituitary Adenomas. Front Endocrinol (Lausanne) (2019) 10:334. 10.3389/fendo.2019.00334
    1. Syro LV, Rotondo F, Camargo M, Ortiz LD, Serna CA, Kovacs K. Temozolomide and Pituitary Tumors: Current Understanding, Unresolved Issues, and Future Directions. Front Endocrinol (Lausanne) (2018) 9:318. 10.3389/fendo.2018.00318
    1. Ilie MD, Jouanneau E, Raverot G. Aggressive Pituitary Adenomas and Carcinomas. Endocrinol Metab Clin North Am (2020) 49(3):505–15. 10.1016/j.ecl.2020.05.008
    1. Ji Y, Vogel RI, Lou E. Temozolomide treatment of pituitary carcinomas and atypical adenomas: systematic review of case reports. Neurooncol Pract (2016) 3(3):188–95. 10.1093/nop/npv059
    1. McCormack AI, Wass JA, Grossman AB. Aggressive pituitary tumours: the role of temozolomide and the assessment of MGMT status. Eur J Clin Invest (2011) 41(10):1133–48. 10.1111/j.1365-2362.2011.02520.x
    1. Araki T, Cooper O, Fukuoka H. Editorial: Targeted Therapy for Pituitary Adenomas. Front Endocrinol (Lausanne) (2019) 10:358. 10.3389/fendo.2019.00358
    1. Dai C, Zhang B, Liu X, Ma S, Yang Y, Yao Y, et al. Inhibition of PI3K/AKT/mTOR pathway enhances temozolomide-induced cytotoxicity in pituitary adenoma cell lines in vitro and xenografted pituitary adenoma in female nude mice. Endocrinology (2013) 154(3):1247–59. 10.1210/en.2012-1908
    1. Lamb LS, Sim H-W, McCormack AI. Exploring the Role of Novel Medical Therapies for Aggressive Pituitary Tumors: A Review of the Literature-“Are We There Yet?”. Cancers (Basel) (2020) 12(2):E308. 10.3390/cancers12020308
    1. Abbott M, Ustoyev Y. Cancer and the Immune System: The History and Background of Immunotherapy. Semin Oncol Nurs (2019) 35(5):150923. 10.1016/j.soncn.2019.08.002
    1. O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol (2019) 16(3):151–67. 10.1038/s41571-018-0142-8
    1. Corrales L, Matson V, Flood B, Spranger S, Gajewski TF. Innate immune signaling and regulation in cancer immunotherapy. Cell Res (2017) 27(1):96–108. 10.1038/cr.2016.149
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science (2018) 359(6382):1350–5. 10.1126/science.aar4060
    1. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu Rev Immunol (2019) 37:457–95. 10.1146/annurev-immunol-041015-055318
    1. Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol (2016) 34:539–73. 10.1146/annurev-immunol-032414-112049
    1. Sharma P, Allison JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol (2020) 20(2):75–6. 10.1038/s41577-020-0275-8
    1. Atkins P, Ur E. Primary and Ipilimumab-induced Hypophysitis: A Single-center Case Series. Endocr Res (2020) 45(4):246–53. 10.1080/07435800.2020.1817064
    1. Faje A. Immunotherapy and hypophysitis: clinical presentation, treatment, and biologic insights. Pituitary (2016) 19(1):82–92. 10.1007/s11102-015-0671-4
    1. Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin (2020) 70(2):86–104. 10.3322/caac.21596
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med (2015) 373(1):23–34. 10.1056/NEJMoa1504030
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer (2019) 19(3):133–50. 10.1038/s41568-019-0116-x
    1. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol (2016) 17(12):e542–e51. 10.1016/S1470-2045(16)30406-5
    1. Giraldo NA, Sanchez-Salas R, Peske JD, Vano Y, Becht E, Petitprez F, et al. The clinical role of the TME in solid cancer. Br J Cancer (2019) 120(1):45–53. 10.1038/s41416-018-0327-z
    1. Clemente CG, Mihm MC, Jr., Bufalino R, Zurrida S, Collini P, Cascinelli N. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer (1996) 77(7):1303–10. 10.1002/(SICI)1097-0142(19960401)77:7<1303::AID-CNCR12>;2-5
    1. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A (2005) 102(51):18538–43. 10.1073/pnas.0509182102
    1. Smyrk TC, Watson P, Kaul K, Lynch HT. Tumor-infiltrating lymphocytes are a marker for microsatellite instability in colorectal carcinoma. Cancer (2001) 91(12):2417–22. 10.1002/1097-0142(20010615)91:12<2417::aid-cncr1276>;2-u
    1. Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, et al. CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma. Proc Natl Acad Sci U S A (2007) 104(10):3967–72. 10.1073/pnas.0611618104
    1. Zhang QW, Liu L, Gong CY, Shi HS, Zeng YH, Wang XZ, et al. Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS One (2012) 7(12):e50946. 10.1371/journal.pone.0050946
    1. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med (2018) 24(5):541–50. 10.1038/s41591-018-0014-x
    1. Ilie MD, Vasiljevic A, Raverot G, Bertolino P. The Microenvironment of Pituitary Tumors-Biological and Therapeutic Implications. Cancers (Basel) (2019) 11(10):1605. 10.3390/cancers11101605
    1. Rossi ML, Jones NR, Esiri MM, Havas L, Izzi M, Coakham HB. Mononuclear cell infiltrate and HLA-Dr expression in 28 pituitary adenomas. Tumori (1990) 76(6):543–7. 10.1177/030089169007600605
    1. Lupi I, Manetti L, Caturegli P, Menicagli M, Cosottini M, Iannelli A, et al. Tumor infiltrating lymphocytes but not serum pituitary antibodies are associated with poor clinical outcome after surgery in patients with pituitary adenoma. J Clin Endocrinol Metab (2010) 95(1):289–96. 10.1210/jc.2009-1583
    1. Heshmati HM, Kujas M, Casanova S, Wollan PC, Racadot J, Van Effenterre R, et al. Prevalence of lymphocytic infiltrate in 1400 pituitary adenomas. Endocr J (1998) 45(3):357–61. 10.1507/endocrj.45.357
    1. Lu J-Q, Adam B, Jack AS, Lam A, Broad RW, Chik CL. Immune Cell Infiltrates in Pituitary Adenomas: More Macrophages in Larger Adenomas and More T Cells in Growth Hormone Adenomas. Endocr Pathol (2015) 26(3):263–72. 10.1007/s12022-015-9383-6
    1. Mei Y, Bi WL, Greenwald NF, Du Z, Agar NYR, Kaiser UB, et al. Increased expression of programmed death ligand 1 (PD-L1) in human pituitary tumors. Oncotarget (2016) 7(47):76565–76. 10.18632/oncotarget.12088
    1. Wang P-F, Wang T-J, Yang Y-K, Yao K, Li Z, Li YM, et al. The expression profile of PD-L1 and CD8(+) lymphocyte in pituitary adenomas indicating for immunotherapy. J Neuro Oncol (2018) 139(1):89–95. 10.1007/s11060-018-2844-2
    1. Sato M, Tamura R, Tamura H, Mase T, Kosugi K, Morimoto Y, et al. Analysis of Tumor Angiogenesis and Immune Microenvironment in Non-Functional Pituitary Endocrine Tumors. J Clin Med (2019) 8(5):695. 10.3390/jcm8050695
    1. Barry S, Carlsen E, Marques P, Stiles CE, Gadaleta E, Berney DM, et al. Tumor microenvironment defines the invasive phenotype of AIP-mutation-positive pituitary tumors. Oncogene (2019) 38(27):5381–95. 10.1038/s41388-019-0779-5
    1. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol (2019) 19(6):369–82. 10.1038/s41577-019-0127-6
    1. Suteau V, Collin A, Menei P, Rodien P, Rousselet MC, Briet C. Expression of programmed death-ligand 1 (PD-L1) in human pituitary neuroendocrine tumor. Cancer Immunol Immunother (2020) 69(10):2053–61. 10.1007/s00262-020-02611-x
    1. Uraki S, Ariyasu H, Doi A, Takeshima K, Morita S, Inaba H, et al. MSH6/2 and PD-L1 Expressions Are Associated with Tumor Growth and Invasiveness in Silent Pituitary Adenoma Subtypes. Int J Mol Sci (2020) 21(8):2831. 10.3390/ijms21082831
    1. Salomon MP, Wang X, Marzese DM, Hsu SC, Nelson N, Zhang X, et al. The epigenomic landscape of pituitary adenomas reveals specific alterations and differentiates among acromegaly, Cushing’s disease and endocrine-inactive subtypes. Clin Cancer Res (2018) 24(17):4126–36. 10.1158/1078-0432
    1. Kemeny HR, Elsamadicy AA, Farber SH, Champion CD, Lorrey SJ, Chongsathidkiet P, et al. Targeting PD-L1 Initiates Effective Antitumor Immunity in a Murine Model of Cushing Disease. Clin Cancer Res (2020) 26(5):1141–51. 10.1158/1078-0432.CCR-18-3486
    1. Kim YH, Kim JH. Transcriptome Analysis Identifies an Attenuated Local Immune Response in Invasive Nonfunctioning Pituitary Adenomas. Endocrinol Metab (Seoul) (2019) 34(3):314–22. 10.3803/EnM.2019.34.3.314
    1. Srirangam Nadhamuni V, Korbonits M. Novel insights into Pituitary Tumorigenesis: Genetic and Epigenetic Mechanisms. Endocr Rev (2020) 41(6):bnaa006. 10.1210/endrev/bnaa006
    1. Elsarrag M, Patel PD, Chatrath A, Taylor D, Jane JA. Genomic and molecular characterization of pituitary adenoma pathogenesis: review and translational opportunities. Neurosurg Focus (2020) 48(6):E11. 10.3171/2020.3.FOCUS20104
    1. Alshaikh OM, Asa SL, Mete O, Ezzat S. An Institutional Experience of Tumor Progression to Pituitary Carcinoma in a 15-Year Cohort of 1055 Consecutive Pituitary Neuroendocrine Tumors. Endocr Pathol (2019) 30(2):118–27. 10.1007/s12022-019-9568-5
    1. McCormack A, Dekkers OM, Petersenn S, Popovic V, Trouillas J, Raverot G, et al. Treatment of aggressive pituitary tumours and carcinomas: results of a European Society of Endocrinology (ESE) survey 2016. Eur J Endocrinol (2018) 178(3):265–76. 10.1530/EJE-17-0933
    1. Hui E. Immune checkpoint inhibitors. J Cell Biol (2019) 218(3):740–1. 10.1083/jcb.201810035
    1. de Miguel M, Calvo E. Clinical Challenges of Immune Checkpoint Inhibitors. Cancer Cell (2020) 38(3):326–33. 10.1016/j.ccell.2020.07.004
    1. Lin AL, Jonsson P, Tabar V, Yang TJ, Cuaron J, Beal K, et al. Marked Response of a Hypermutated ACTH-Secreting Pituitary Carcinoma to Ipilimumab and Nivolumab. J Clin Endocrinol Metab (2018) 103(10):3925–30. 10.1210/jc.2018-01347
    1. Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB, et al. Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment. Clin Cancer Res (2007) 13(7):2038–45. 10.1158/1078-0432.CCR-06-2149
    1. Caccese M, Barbot M, Ceccato F, Padovan M, Gardiman MP, Fassan M, et al. Rapid disease progression in patient with mismatch-repair deficiency pituitary ACTH-secreting adenoma treated with checkpoint inhibitor pembrolizumab. Anti Cancer Drugs (2020) 31(2):199–204. 10.1097/CAD.0000000000000856
    1. Meng X, Huang Z, Teng F, Xing L, Yu J. Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy. Cancer Treat Rev (2015) 41(10):868–76. 10.1016/j.ctrv.2015.11.001
    1. Morris DG, Musat M, Czirjak S, Hanzely Z, Lillington DM, Korbonits M, et al. Differential gene expression in pituitary adenomas by oligonucleotide array analysis. Eur J Endocrinol (2005) 153(1):143–51. 10.1530/eje.1.01937
    1. Tong Y, Zheng Y, Zhou J, Oyesiku NM, Koeffler HP, Melmed S. Genomic characterization of human and rat prolactinomas. Endocrinology (2012) 153(8):3679–91. 10.1210/en.2012-1056
    1. Yu SY, Hong LC, Feng J, Wu YT, Zhang YZ. Integrative proteomics and transcriptomics identify novel invasive-related biomarkers of non-functioning pituitary adenomas. Tumour Biol (2016) 37(7):8923–30. 10.1007/s13277-015-4767-2
    1. Michaelis KA, Knox AJ, Xu M, Kiseljak-Vassiliades K, Edwards MG, Geraci M, et al. Identification of growth arrest and DNA-damage-inducible gene beta (GADD45beta) as a novel tumor suppressor in pituitary gonadotrope tumors. Endocrinology (2011) 152(10):3603–13. 10.1210/en.2011-0109
    1. Yang Q, Wang Y, Zhang S, Tang J, Li F, Yin J, et al. Biomarker Discovery for Immunotherapy of Pituitary Adenomas: Enhanced Robustness and Prediction Ability by Modern Computational Tools. Int J Mol Sci (2019) 20(1):151. 10.3390/ijms20010151
    1. Kasuki L, Wildemberg LE, Gadelha MR. MANAGEMENT OF ENDOCRINE DISEASE: Personalized medicine in the treatment of acromegaly. Eur J Endocrinol (2018) 178(3):R89–R100. 10.1530/EJE-17-1006
    1. Lines KE, Stevenson M, Thakker RV. Animal models of pituitary neoplasia. Mol Cell Endocrinol (2016) 421:68–81. 10.1016/j.mce.2015.08.024
    1. Cristina C, Luque GM, Demarchi G, Lopez Vicchi F, Zubeldia-Brenner L, Perez Millan MI, et al. Angiogenesis in pituitary adenomas: human studies and new mutant mouse models. Int J Endocrinol (2014) 2014:608497. 10.1155/2014/608497
    1. Karamboulas C, Meens J, Ailles L. Establishment and Use of Patient-Derived Xenograft Models for Drug Testing in Head and Neck Squamous Cell Carcinoma. STAR Protoc (2020) 1(1):100024. 10.1016/j.xpro.2020.100024
    1. Nivolumab and Ipilimumab in People With Aggressive Pituitary Tumors. Available at: (Accessed September 20, 2020).
    1. Nivolumab and Ipilimumab in Treating Patients With Rare Tumors. Available at: (Accessed September 20, 2020).

Source: PubMed

3
Iratkozz fel