PD-1 Blockade in Anaplastic Thyroid Carcinoma

Jaume Capdevila, Lori J Wirth, Thomas Ernst, Santiago Ponce Aix, Chia-Chi Lin, Rodryg Ramlau, Marcus O Butler, Jean-Pierre Delord, Hans Gelderblom, Paolo A Ascierto, Angelica Fasolo, Dagmar Führer, Marie Luise Hütter-Krönke, Patrick M Forde, Anna Wrona, Armando Santoro, Peter M Sadow, Sebastian Szpakowski, Hongqian Wu, Geraldine Bostel, Jason Faris, Scott Cameron, Andreea Varga, Matthew Taylor, Jaume Capdevila, Lori J Wirth, Thomas Ernst, Santiago Ponce Aix, Chia-Chi Lin, Rodryg Ramlau, Marcus O Butler, Jean-Pierre Delord, Hans Gelderblom, Paolo A Ascierto, Angelica Fasolo, Dagmar Führer, Marie Luise Hütter-Krönke, Patrick M Forde, Anna Wrona, Armando Santoro, Peter M Sadow, Sebastian Szpakowski, Hongqian Wu, Geraldine Bostel, Jason Faris, Scott Cameron, Andreea Varga, Matthew Taylor

Abstract

Purpose: Anaplastic thyroid carcinoma is an aggressive malignancy that is almost always fatal and lacks effective systemic treatment options for patients with BRAF-wild type disease. As part of a phase I/II study in patients with advanced/metastatic solid tumors, patients with anaplastic thyroid carcinoma were treated with spartalizumab, a humanized monoclonal antibody against the programmed death-1 (PD-1) receptor.

Methods: We enrolled patients with locally advanced and/or metastatic anaplastic thyroid carcinoma in a phase II cohort of the study. Patients received 400 mg spartalizumab intravenously, once every 4 weeks. The overall response rate was determined according to RECIST v1.1.

Results: Forty-two patients were enrolled. Adverse events were consistent with those previously observed with PD-1 blockade. Most common treatment-related adverse events were diarrhea (12%), pruritus (12%), fatigue (7%), and pyrexia (7%). The overall response rate was 19%, including three patients with a complete response and five with a partial response. Most patients had baseline tumor biopsies positive for PD-L1 expression (n = 28/40 evaluable), and response rates were higher in PD-L1-positive (8/28; 29%) versus PD-L1-negative (0/12; 0%) patients. The highest rate of response was observed in the subset of patients with PD-L1 ≥ 50% (6/17; 35%). Responses were seen in both BRAF-nonmutant and BRAF-mutant patients and were durable, with a 1-year survival of 52.1% in the PD-L1-positive population.

Conclusion: To our knowledge, this is the first clinical trial to show responsiveness of anaplastic thyroid carcinoma to PD-1 blockade.

Trial registration: ClinicalTrials.gov NCT02404441.

Figures

FIG 1.
FIG 1.
Duration of exposure to spartalizumab and percentage change from baseline in sum of diameters of target lesions, by programmed death-ligand 1 (PD-L1) expression at baseline. (A) Duration of exposure to spartalizumab. Mutant denotes BRAF mutation. (B) Best percentage change from baseline in sum of diameters of target lesions. Thirty-one patients were evaluable for best percentage change; 11 patients were not evaluable because of discontinuation or death before first postbaseline assessment (n = 8) or missing postbaseline assessment (n = 3). Best overall response by RECIST v1.1 is indicated. Where available, BRAF mutation status is indicated. Patient number refers to number in Data Supplement. CR, complete response; DCR, disease control rate [CR + PR + SD]; irRC, immune-related response criteria; NGS, next-generation sequencing; ORR, overall response rate [CR + PR]; PCR, polymerase chain reaction; PD, progressive disease; PR, partial response; SD, stable disease; UNK, unknown.
FIG 2.
FIG 2.
Overall survival by PD-L1 expression at baseline. NE, not estimable; PD-L1, programmed death-ligand 1.
FIG 3.
FIG 3.
Interferon γ (IFNγ) signature, by best percentage change from baseline in sum of diameters of target lesions. Best overall response by RECIST v1.1 is indicated. Data were available for 18 patients. Spearman coefficient, −0.67; 95% CI, −0.9 to −0.3. CR, complete response; PD, progressive disease; PD-L1, programmed death-ligand 1; PR, partial response; SD, stable disease.

References

    1. Haddad RI, Nasr C, Bischoff L, et al. NCCN guidelines insights: Thyroid carcinoma, version 2.2018. J Natl Compr Canc Netw. 2018;16:1429–1440.
    1. Smallridge RC, Ain KB, Asa SL, et al. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid. 2012;22:1104–1139.
    1. Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: Pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol) 2010;22:486–497.
    1. Tiedje V, Stuschke M, Weber F, et al. Anaplastic thyroid carcinoma: Review of treatment protocols. Endocr Relat Cancer. 2018;25:R153–R161.
    1. Landa I, Ibrahimpasic T, Boucai L, et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J Clin Invest. 2016;126:1052–1066.
    1. Pozdeyev N, Gay LM, Sokol ES, et al. Genetic analysis of 779 advanced differentiated and anaplastic thyroid cancers. Clin Cancer Res. 2018;24:3059–3068.
    1. Kunstman JW, Juhlin CC, Goh G, et al. Characterization of the mutational landscape of anaplastic thyroid cancer via whole-exome sequencing. Hum Mol Genet. 2015;24:2318–2329.
    1. Subbiah V, Kreitman RJ, Wainberg ZA, et al. Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid cancer. J Clin Oncol. 2018;36:7–13.
    1. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372:2521–2532.
    1. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–1639.
    1. Khunger M, Hernandez AV, Pasupuleti V, et al: Programmed cell death 1 (PD-1) ligand (PD-L1) expression in solid tumors as a predictive biomarker of benefit from PD-1/PD-L1 axis inhibitors: A systematic review and meta-analysis. JCO Precis Oncol doi: .
    1. Ahn S, Kim TH, Kim SW, et al. Comprehensive screening for PD-L1 expression in thyroid cancer. Endocr Relat Cancer. 2017;24:97–106.
    1. Zwaenepoel K, Jacobs J, De Meulenaere A, et al. CD70 and PD-L1 in anaplastic thyroid cancer - promising targets for immunotherapy. Histopathology. 2017;71:357–365.
    1. Kollipara R, Schneider B, Radovich M, et al. Exceptional response with immunotherapy in a patient with anaplastic thyroid cancer. Oncologist. 2017;22:1149–1151.
    1. Iyer PC, Dadu R, Gule-Monroe M, et al. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J Immunother Cancer. 2018;6:68.
    1. Naing A, Gainor JF, Gelderblom H, et al: A first-in-human phase 1 dose escalation study of spartalizumab (PDR001), an anti-PD-1 antibody, in patients with advanced solid tumors. J Immunother Cancer 8:e000530, 2020.
    1. Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–1031.
    1. Perucho M. Cancer of the microsatellite mutator phenotype. Biol Chem. 1996;377:675–684.
    1. Khoja L, Butler MO, Kang SP, et al. Pembrolizumab. J Immunother Cancer. 2015;3:36.
    1. Brahmer JR, Hammers H, Lipson EJ. Nivolumab: Targeting PD-1 to bolster antitumor immunity. Future Oncol. 2015;11:1307–1326.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
    1. Turajlic S, Litchfield K, Xu H, et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: A pan-cancer analysis. Lancet Oncol. 2017;18:1009–1021.
    1. Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. 2018;6:157.
    1. Kaufman HL, Russell JS, Hamid O, et al. Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after ≥1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial. J Immunother Cancer. 2018;6:7.

Source: PubMed

3
Prenumerera