Efficacy of pembrolizumab in patients with pituitary carcinoma: report of four cases from a phase II study

Nazanin Majd, Steven G Waguespack, Filip Janku, Siqing Fu, Marta Penas-Prado, Mingxuan Xu, Anas Alshawa, Carlos Kamiya-Matsuoka, Shaan M Raza, Ian E McCutcheon, Aung Naing, Nazanin Majd, Steven G Waguespack, Filip Janku, Siqing Fu, Marta Penas-Prado, Mingxuan Xu, Anas Alshawa, Carlos Kamiya-Matsuoka, Shaan M Raza, Ian E McCutcheon, Aung Naing

Abstract

Pituitary carcinoma is an aggressive tumor characterized by metastatic spread beyond the sellar region. Symptoms can be debilitating due to hormonal excess and survival is poor. Pituitary carcinomas recur despite conventional multimodality treatments. Given the recent advances in the use of immune checkpoint inhibitors (CPIs) to treat various solid cancers, there has been interest in exploring the role of immunotherapy for treating aggressive, refractory pituitary tumors. We treated 4 patients with pituitary carcinoma with pembrolizumab as part of a phase II clinical trial. Two patients (patients 1 and 2) with functioning corticotroph pituitary carcinomas (refractory to surgery, radiotherapy and chemotherapy) had partial radiographic (60% and 32% per Immune-Related Response Evaluation Criteria In Solid Tumors, respectively) and hormonal responses. Patient 1's response continues 42 months after initiation of pembrolizumab and his tumor tissue obtained after treatment with temozolomide demonstrated a hypermutator phenotype with MSH2 and MSH6 gene mutations. Patient 2's tumor after exposure to temozolomide was not sampled, but prior somatic mutational testing was negative. One patient with a non-functioning corticotroph tumor (patient 3) had a best response of stable disease for 4 months. One patient with a prolactin-secreting carcinoma (patient 4) had progressive disease. The latter 2 patients' tumors did not demonstrate a hypermutator phenotype after treatment with temozolomide. Programmed death-ligand 1 staining was negative in all tumors. We report 2 cases of corticotroph pituitary carcinoma responsive to pembrolizumab after prior exposure to alkylating agents. The role of CPIs in treating patients with pituitary carcinoma, the relationship between tumor subtype and response to immunotherapy and mechanisms of hypermutation in this orphan disease require further study.Trial registration number: NCT02721732.

Keywords: biomarkers; clinical trials; immunotherapy; phase II as topic; tumor.

Conflict of interest statement

Competing interests: No, there are no competing interests.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Response to pembrolizumab in patient 1. (A) T1 postcontrast MRI obtained prior to pembrolizumab treatment demonstrates a lobulated enhancing mass within the sphenoid sinus and posterior ethmoid air cells (gray arrow) with extension to the anterior and inferior aspects of the left temporal lobe (white arrow). (B) MRI of the brain after two cycles of pembrolizumab demonstrates significant improvement of the enhancing mass in the cavernous sinus and resolution of the nodule extending to the left temporal lobe. (C) The adrenocorticotropic hormone (ACTH) level became undetectable after treatment with pembrolizumab. (Reference ranges for ACTH are 0–46 pg/mL (before 4/19/18) and 7–63 pg/mL (after 04/19/18); ACTH levels are listed as ‘0’ if the reported value was less than the lower limit of detection for the assay.)
Figure 2
Figure 2
Clinical response in patient 2. (A) Adrenocorticotropic hormone (ACTH) levels became significantly elevated after starting pembrolizumab and then gradually declined but have not yet nadired as of the last evaluation. (B) Overview of the patient’s skin pigmentation changes. (i) Two and a half years before the diagnosis of pituitary carcinoma, the patient had normal skin coloration and an ACTH level of 160 pg/mL. (ii) At diagnosis of pituitary carcinoma, when the ACTH was 84 851 pg/mL, the patient had become remarkably hyperpigmented. Ten months after treatment with pembrolizumab commenced and when the ACTH was 628 pg/mL (iv), her skin pigmentation was noticeably lighter compared with pictures obtained 6 months earlier (iii), after 4 months of therapy, when her ACTH was >11 000 pg/mL. (Reference ranges for ACTH are 0–46 pg/mL (before 4/19/18) and 7–63 pg/mL (after 04/19/18).).

References

    1. Lloyd RV, Osamura RY, Klöppel G, et al. . Who classification of tumours of endocrine organs. 4th edn Lyon: IARC Press, 2017.
    1. Buurman H, Saeger W. Subclinical adenomas in postmortem pituitaries: classification and correlations to clinical data. Eur J Endocrinol 2006;154:753–8. 10.1530/eje.1.02107
    1. Daly AF, Tichomirowa MA, Beckers A. The epidemiology and genetics of pituitary adenomas. Best Pract Res Clin Endocrinol Metab 2009;23:543–54. 10.1016/j.beem.2009.05.008
    1. Raverot G, Burman P, McCormack A, et al. . European Society of endocrinology clinical practice guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol 2018;178:G1–24. 10.1530/EJE-17-0796
    1. Zacharia BE, Gulati AP, Bruce JN, et al. . High response rates and prolonged survival in patients with corticotroph pituitary tumors and refractory Cushing disease from capecitabine and temozolomide (CAPTEM): a case series. Neurosurgery 2014;74:E447–55. discussion E55. 10.1227/NEU.0000000000000251
    1. Darvin P, Toor SM, Sasidharan Nair V, et al. . Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med 2018;50:1–11. 10.1038/s12276-018-0191-1
    1. Campbell BB, Light N, Fabrizio D, et al. . Comprehensive analysis of hypermutation in human cancer. Cell 2017;171:1042–56. 10.1016/j.cell.2017.09.048
    1. Kandoth C, McLellan MD, Vandin F, et al. . Mutational landscape and significance across 12 major cancer types. Nature 2013;502:333–9. 10.1038/nature12634
    1. Kamiya-Matsuoka C, Metrus NR, Shaw KR, et al. . The natural course of hypermutator gliomas. Journal of Clinical Oncology 2018;36:2014 10.1200/JCO.2018.36.15_suppl.2014
    1. Duhamel C, Ilie MD, Salle H, et al. . Immunotherapy in corticotroph and lactotroph aggressive tumors and carcinomas: two case reports and a review of the literature. J Pers Med 2020;10. 10.3390/jpm10030088. [Epub ahead of print: 13 08 2020].
    1. Lin AL, Jonsson P, Tabar V, et al. . Marked response of a hypermutated ACTH-secreting pituitary carcinoma to ipilimumab and nivolumab. J Clin Endocrinol Metab 2018;103:3925–30. 10.1210/jc.2018-01347
    1. Naing A, Meric-Bernstam F, Stephen B, et al. . Phase 2 study of pembrolizumab in patients with advanced rare cancers. J Immunother Cancer 2020;8:e000347. 10.1136/jitc-2019-000347
    1. Nishino M, Tirumani SH, Ramaiya NH, et al. . Cancer immunotherapy and immune-related response assessment: the role of radiologists in the new arena of cancer treatment. Eur J Radiol 2015;84:1259–68. 10.1016/j.ejrad.2015.03.017
    1. Dolled-Filhart M, Locke D, Murphy T, et al. . Development of a prototype immunohistochemistry assay to measure programmed death ligand-1 expression in tumor tissue. Arch Pathol Lab Med 2016;140:1259–66. 10.5858/arpa.2015-0544-OA
    1. Marabelle A, Le DT, Ascierto PA, et al. . Efficacy of pembrolizumab in patients with Noncolorectal high microsatellite Instability/Mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol 2020;38:1–10. 10.1200/JCO.19.02105
    1. Kunkel TA, Erie DA. Eukaryotic mismatch repair in relation to DNA replication. Annu Rev Genet 2015;49:291–313. 10.1146/annurev-genet-112414-054722
    1. Le DT, Durham JN, Smith KN, et al. . Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–13. 10.1126/science.aan6733
    1. Caccese M, Barbot M, Ceccato F, et al. . Rapid disease progression in patient with mismatch-repair deficiency pituitary ACTH-secreting adenoma treated with checkpoint inhibitor pembrolizumab. Anticancer Drugs 2020;31:199–204. 10.1097/CAD.0000000000000856
    1. Hodges TR, Ott M, Xiu J, et al. . Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro Oncol 2017;19:1047–57. 10.1093/neuonc/nox026
    1. Mei Y, Bi WL, Greenwald NF, et al. . Increased expression of programmed death ligand 1 (PD-L1) in human pituitary tumors. Oncotarget 2016;7:76565–76. 10.18632/oncotarget.12088
    1. Yagnik G, Rutowski MJ, Shah SS, et al. . Stratifying nonfunctional pituitary adenomas into two groups distinguished by macrophage subtypes. Oncotarget 2019;10:2212–23. 10.18632/oncotarget.26775
    1. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 2006;124:263–6. 10.1016/j.cell.2006.01.007
    1. Lu J-Q, Adam B, Jack AS, et al. . Immune cell infiltrates in pituitary adenomas: more macrophages in larger adenomas and more T cells in growth hormone adenomas. Endocr Pathol 2015;26:263–72. 10.1007/s12022-015-9383-6
    1. Knosp E, Steiner E, Kitz K, et al. . Pituitary adenomas with invasion of the cavernous sinus space: a magnetic resonance imaging classification compared with surgical findings. Neurosurgery 1993;33:610–7. discussion 7-8. 10.1227/00006123-199310000-00008
    1. Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. . Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol 2018;4:173–82. 10.1001/jamaoncol.2017.3064
    1. Kitajima K, Ashida K, Wada N, et al. . Isolated ACTH deficiency probably induced by autoimmune-related mechanism evoked with nivolumab. Jpn J Clin Oncol 2017;47:463–6. 10.1093/jjco/hyx018
    1. Tan MH, Iyengar R, Mizokami-Stout K, et al. . Spectrum of immune checkpoint inhibitors-induced endocrinopathies in cancer patients: a scoping review of case reports. Clin Diabetes Endocrinol 2019;5:1. 10.1186/s40842-018-0073-4

Source: PubMed

3
订阅