Pexidartinib Long-Term Hepatic Safety Profile in Patients with Tenosynovial Giant Cell Tumors

James H Lewis, Hans Gelderblom, Michiel van de Sande, Silvia Stacchiotti, John H Healey, William D Tap, Andrew J Wagner, Antonio Lopez Pousa, Mihaela Druta, Chia-Chi Lin, Hideo A Baba, Youngsook Choi, Qiang Wang, Dale E Shuster, Sebastian Bauer, James H Lewis, Hans Gelderblom, Michiel van de Sande, Silvia Stacchiotti, John H Healey, William D Tap, Andrew J Wagner, Antonio Lopez Pousa, Mihaela Druta, Chia-Chi Lin, Hideo A Baba, Youngsook Choi, Qiang Wang, Dale E Shuster, Sebastian Bauer

Abstract

Background: Pexidartinib is approved in the U.S. for tenosynovial giant cell tumors (TGCTs). Herein, we assessed the hepatic safety profile of pexidartinib across patients with TGCTs receiving pexidartinib.

Materials, and methods: Hepatic adverse reactions (ARs) were assessed by type and magnitude of liver test abnormalities, classified as (a) isolated aminotransferase elevations (alanine [ALT] or aspartate [AST], without significant alkaline phosphatase [ALP] or bilirubin elevations), or (b) mixed or cholestatic hepatotoxicity (increase in ALP with or without ALT/AST and bilirubin elevations, based on adjudication). Median follow-up from initial pexidartinib treatment was 39 months (range, 32-82) in 140 patients with TGCTs across clinical studies NCT01004861, NCT02371369, NCT02734433, and NCT03291288.

Results: In total, 95% of patients with TGCTs (133/140) treated with pexidartinib (median duration of exposure, 19 months [range, 1-76]), experienced a hepatic AR. A total of 128 patients (91%) had reversible, low-grade dose-dependent isolated AST/ALT elevations without significant ALP elevations. Five patients (4%) experienced serious mixed or cholestatic injury. No case met Hy's law criteria. Onset of hepatic ARs was predominantly in the first 2 months. All five serious hepatic AR cases recovered 1-7 months following pexidartinib discontinuation. Five patients from the non-TGCT population (N = 658) experienced serious hepatic ARs, two irreversible cases.

Conclusion: This pooled analysis provides information to help form the basis for the treating physician's risk assessment for patients with TCGTs, a locally aggressive but typically nonmetastatic tumor. In particular, long-term treatment with pexidartinib has a predictable effect on hepatic aminotransferases and unpredictable risk of serious cholestatic or mixed liver injury.

Implications for practice: This is the first long-term pooled analysis to report on the long-term hepatic safety of pexidartinib in patients with tenosynovial giant cell tumors associated with severe morbidity or functional limitations and not amenable to improvement with surgery. These findings extend beyond what has been previously published, describing the observed instances of hepatic toxicity following pexidartinib treatment across the clinical development program. This information is highly relevant for medical oncologists and orthopedic oncologists and provides guidance for its proper use for appropriate patients within the Pexidartinib Risk Evaluation and Mitigation Safety program.

Keywords: Adverse reactions; Hepatic safety; Long-term; Pexidartinib; Tenosynovial giant cell tumor.

Conflict of interest statement

Disclosures of potential conflicts of interest may be found at the end of this article.

© 2020 The Authors. The Oncologist published by Wiley Periodicals LLC on behalf of AlphaMed Press.

Figures

Figure 1
Figure 1
Kaplan‐Meier curve of time to first occurrence of hepatic adverse reactions. Hepatic Laboratory Criteria: alanine aminotransferase >3× upper limit of normal (ULN), or aspartate aminotransferase >3× ULN, or alkaline phosphatase (ALP) >2× ULN, or total bilirubin > ULN, or direct bilirubin > ULN. If ALP >2× ULN and gamma‐glutamyl transferase (GGT) is measured on the same date, GGT must also be >2× ULN.Abbreviation: TGCT, tenosynovial giant cell tumor.
Figure 2
Figure 2
Clinical laboratory results of patients with tenosynovial giant cell tumors (TGCTs) experiencing serious hepatic adverse reactions. (A): Case no. 1. (B): Case no. 1: H&E: portal tract (black circle) without a bile duct or ductular proliferation but with few inflammatory cells. 400× magnification. (C): Case no. 1: immunohistochemical staining against CK7. Portal tract shows loss of bile duct but CK7‐positive adjacent hepatocytes indicating chronic cholestasis (arrows). 400× magnification. (D): Case no. 2. (E): Case no. 3. (F): Case no. 4. (G): Case no. 5.Abbreviations: ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; BL, baseline; CK7, cytokeratin 7; d, day; H&E, hematoxylin and eosin; LFT, liver function test; ULN, upper limit of normal.

References

    1. Mastboom MJL, Palmerini E, Verspoor FGM et al. Surgical outcomes of patients with diffuse‐type tenosynovial giant‐cell tumours: An international, retrospective, cohort study. Lancet Oncol 2019;20:877–886.
    1. de Saint Aubain Somerhausen N, van de Rijn M. Tenosynovial giant cell tumor: localized type, diffuse type. In: Fletcher C, Bridge J, Hogendoorn P, Martens F, eds. WHO Classification of Tumours of Soft Tissue and Bone. 4th ed. Lyon, France: IARC Press; 2013:100–103.
    1. Staals EL, Ferrari S, Donati DM et al. Diffuse‐type tenosynovial giant cell tumour: Current treatment concepts and future perspectives. Eur J Cancer 2016;63:34–40.
    1. Mastboom MJL, Verspoor FGM, Gelderblom H et al. Limb amputation after multiple treatments of tenosynovial giant cell tumour: Series of 4 Dutch cases. Case Rep Orthop 2017;2017:7402570.
    1. Tap WD. Multidisciplinary care in tenosynovial giant cell tumours. Lancet Oncol 2019;20:755–756.
    1. Tap WD, Wainberg ZA, Anthony SP et al. Structure‐guided blockade of CSF1R kinase in tenosynovial giant‐cell tumor. N Engl J Med 2015;373:428–437.
    1. Brahmi M, Vinceneux A, Cassier PA. Current systemic treatment options for tenosynovial giant cell tumor/pigmented villonodular synovitis: Targeting the CSF1/CSF1R axis. Curr Treat Options Oncol 2016;17:10.
    1. Cassier PA, Italiano A, Gomez‐Roca CA et al. CSF1R inhibition with emactuzumab in locally advanced diffuse‐type tenosynovial giant cell tumours of the soft tissue: A dose‐escalation and dose‐expansion phase 1 study. Lancet Oncol 2015;16:949–956.
    1. Gelderblom H, Cropet C, Chevreau C et al. Nilotinib in locally advanced pigmented villonodular synovitis: A multicentre, open‐label, single‐arm, phase 2 trial. Lancet Oncol 2018;19:639–648.
    1. Nakayama R, Jagannathan JP, Ramaiya N et al. Clinical characteristics and treatment outcomes in six cases of malignant tenosynovial giant cell tumor: Initial experience of molecularly targeted therapy. BMC Cancer 2018;18:1296.
    1. Palmerini E, Staals EL, Maki RG et al. Tenosynovial giant cell tumour/pigmented villonodular synovitis: Outcome of 294 patients before the era of kinase inhibitors. Eur J Cancer 2015;51:210–217.
    1. Ravi V, Wang WL, Lewis VO. Treatment of tenosynovial giant cell tumor and pigmented villonodular synovitis. Curr Opin Oncol 2011;23:361–366.
    1. Rebuzzi SE, Grassi M, Catalano F et al. Multiple systemic treatment options in a patient with malignant tenosynovial giant cell tumour. Anticancer Drugs 2020;31:80–84.
    1. Tap WD, Gelderblom H, Palmerini E et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): A randomised phase 3 trial. Lancet 2019;394:478‐487.
    1. Temple HT. Pigmented villonodular synovitis therapy with MSCF‐1 inhibitors. Curr Opin Oncol 2012;24:404–408.
    1. Verspoor FGM, Mastboom MJL, Hannink G et al. Long‐term efficacy of imatinib mesylate in patients with advanced tenosynovial giant cell tumor. Sci Rep 2019;9:14551.
    1. Turalio (pexidartinib) capsules, for oral use . Prescribing information. Daiichi Sankyo, Inc.; 2020.
    1. NCCN clinical practice guidelines in oncology: Soft tissue sarcoma—version 5.2019. Plymouth Meeting, PA: National Comprehensive Cancer Network; 2020.
    1. Shah RR, Morganroth J, Shah DR. Hepatotoxicity of tyrosine kinase inhibitors: Clinical and regulatory perspectives. Drug Saf 2013;36:491–503.
    1. Teo YL, Ho HK, Chan A. Risk of tyrosine kinase inhibitors‐induced hepatotoxicity in cancer patients: A meta‐analysis. Cancer Treat Rev 2013;39:199–206.
    1. Teo YL, Ho HK, Chan A. Formation of reactive metabolites and management of tyrosine kinase inhibitor‐induced hepatotoxicity: A literature review. Expert Opin Drug Metab Toxicol 2015;11:231–242.
    1. Lee JH, Chen TW, Hsu CH et al. A phase I study of pexidartinib, a colony‐stimulating factor 1 receptor inhibitor, in Asian patients with advanced solid tumors. Invest New Drugs 2020;38:99–110.
    1. U.S. Food and Drug Administration. FDA Briefing Document. Oncologic Drugs Advisory Committee Meeting May 14 , 2019: NDA 211810: Pexidartinib. Washington, DC: U.S. Food and Drug Administration; 2019
    1. Regev A, Björnsson ES. Drug‐induced liver injury: Morbidity, mortality, and Hy's law. Gastroenterology 2014;147:20–24.
    1. Danan G, Benichou C. Causality assessment of adverse reactions to drugs–I. A novel method based on the conclusions of international consensus meetings: Application to drug‐induced liver injuries. J Clin Epidemiol 1993;46:1323–1330.
    1. Piawah S, Hyland C, Umetsu SE et al. A case report of vanishing bile duct syndrome after exposure to pexidartinib (PLX3397) and paclitaxel. NPJ Breast Cancer 2019;5:17.
    1. Genovese MC, Hsia E, Belkowski SM et al. Results from a phase IIA parallel group study of JNJ‐40346527, an oral CSF‐1R inhibitor, in patients with active rheumatoid arthritis despite disease‐modifying antirheumatic drug therapy. J Rheumatol 2015;42:1752–1760.
    1. Pognan F, Couttet P, Demin I et al. Colony‐stimulating factor‐1 antibody lacnotuzumab in a phase 1 healthy volunteer study and mechanistic investigation of safety outcomes. J Pharmacol Exp Ther 2019;369:428–442.
    1. Chen M, Borlak J, Tong W. High lipophilicity and high daily dose of oral medications are associated with significant risk for drug‐induced liver injury. Hepatology 2013;58:388–396.
    1. Lammert C, Einarsson S, Saha C et al. Relationship between daily dose of oral medications and idiosyncratic drug‐induced liver injury: Search for signals. Hepatology 2008;47:2003–2009.
    1. Lewis JH. Drug‐induced liver injury, dosage, and drug disposition: Is idiosyncrasy really unpredictable? Clin Gastroenterol Hepatol 2014;12:1556–1561.
    1. Chalasani NP, Hayashi PH, Bonkovsky HL et al. ACG Clinical Guideline: The diagnosis and management of idiosyncratic drug‐induced liver injury. Am J Gastroenterol 2014;109:950‐966; quiz 967.
    1. Lewis JH. The art and science of diagnosing and managing drug‐induced liver injury in 2015 and beyond. Clin Gastroenterol Hepatol 2015;13:2173–2189.e2178.
    1. Huang J, Meng L, Yang B et al. Safety profile of epidermal growth factor receptor tyrosine kinase inhibitors: A disproportionality analysis of FDA adverse event reporting system. Sci Rep 2020;10:4803.
    1. Chalasani NP, Bonkovsky HL, Fontana RJ et al. Features and outcomes of 899 patients with drug‐induced liver injury: The DILIN prospective study. Gastroenterology 2015;148:1340–1352.
    1. LiverTox : Clinical and research information on drug‐induced liver injury. Bethesda, MD: National Institute of Diabetes and Digestive and Kidney Diseases; 2012.
    1. Kim MN, Yee J, Cho YS et al. Risk factors for erlotinib‐induced hepatotoxicity: A retrospective follow‐up study. BMC Cancer 2018;18:988.
    1. Desmet VJ. Vanishing bile duct disorders. Prog Liver Dis 1992;10:89–121.
    1. Desmet VJ, van Eyken P, Roskams T. Histopathology of vanishing bile duct diseases. Adv Clin Path 1998;2:87–99.
    1. Visentin M, Lenggenhager D, Gai Z et al. Drug‐induced bile duct injury. Biochim Biophys Acta Mol Basis Dis 2018;1864:1498–1506.

Source: PubMed

3
구독하다