Your new experience awaits. Try the new design now and help us make it even better

ORIGINAL RESEARCH article

Front. Oncol., 18 August 2025

Sec. Pharmacology of Anti-Cancer Drugs

Volume 15 - 2025 | https://doi.org/10.3389/fonc.2025.1594585

This article is part of the Research TopicOptimizing Cancer Treatment Timing: Balancing Patient Outcomes and Healthcare EfficiencyView all articles

Adverse events of pexidartinib for the treatment of TGCT: a real-world disproportionality analysis using FDA Adverse Event Reporting System database

Yu Lin&#x;Yu Lin1†Xinlei Zheng&#x;Xinlei Zheng2†Li Lin*Li Lin3*Maohua Chen*Maohua Chen2*
  • 1Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, China
  • 2Department of Pharmacy, Pingtan Comprehensive Experimental Area Hospital, Fuzhou, China
  • 3Department of Medical Oncology, Department of Medical Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China

Introduction: Pexidartinib, an oral selective colony-stimulating factor 1 receptor (CSF1R) inhibitor, is the only systemic therapy approved by the U.S. Food and Drug Administration (FDA) for tenosynovial giant cell tumor (TGCT). While clinical trials have defined its initial safety profile, their limited sample sizes and short follow-up restrict the detection of rare or delayed adverse events (AEs), underscoring the need for real-world pharmacovigilance.

Methods: Using the FDA Adverse Event Reporting System (FAERS) database, we conducted a disproportionality analysis to characterize the post-approval safety profile of pexidartinib and identify unlabeled AEs, applying reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS) methods.

Results: Among 7,168,342 FAERS reports, 668 implicated pexidartinib as the primary suspect, with AEs reported across 26 organ systems. Sixty-seven preferred terms met the criteria of all four signal detection methods, including 16 not listed in the FDA-approved label.

Discussion: The overall safety profile was largely consistent with clinical trial findings, while newly detected AEs suggest possible rare or delayed toxicities in broader patient populations. These results highlight the importance of continuous post-marketing surveillance and support the need for prospective studies to clarify causal relationships.

1 Introduction

Tenosynovial giant cell tumor (TGCT), historically termed pigmented villonodular synovitis (PVNS) or giant cell tumor of the tendon sheath, is a rare mesenchymal neoplasm originating from synovial membranes of joints and the tendon sheaths (1). For patients with symptomatic TGCT refractory to local therapies or causing significant functional impairment, systemic therapy should be considered if surgery is unlikely to achieve functional outcome improvement. The molecular pathogenesis of TGCT involves genomic alterations at the colony-stimulating factor 1 (CSF1) locus (1p13), resulting in aberrant CSF1 overexpression in a subset of tumor cells (2). This molecular pathology supports CSF1/CSF1 receptor (CSF1R) axis inhibition as a central therapeutic strategy.

Pexidartinib, an oral selective CSF1R inhibitor, represents the only approved systemic treatment for TGCT (3). Its Food and Drug Administration (FDA) approval in August 2019 introduced it as the first systemic treatment for adult patients with symptomatic TGCT associated with severe morbidity or functional limitations and not amenable to improvement with surgery (4, 5). Pre-marketing clinical trials have since validated pexidartinib’s efficacy and safety, reinforcing its pivotal role in TGCT management (6, 7).

Comprehensive monitoring of pexidartinib’s real-world safety profile remains imperative given its expanding clinical utilization following FDA approval. Drug safety assessments advocate intervention strategies, including dose control, to mitigate toxicity (8). According to FDA’s prescribing information, pexidartinib’s common adverse events (AEs) included increased lactate dehydrogenase, increased aspartate aminotransferase, hair color changes, fatigue, increased alanine aminotransferase, decreased neutrophils, increased cholesterol, increased alkaline phosphatase, decreased lymphocytes, periorbital edema, decreased hemoglobin, rash, dysgeusia, and decreased phosphate. However, patients may experience AEs during off-label use due to comorbidities, concomitant medications, or genetic predispositions. Given the limited sample sizes and short observation periods in clinical trials, extensive post-marketing safety research in real-world settings is essential. Nevertheless, comprehensive real-world pharmacovigilance studies specifically addressing its AEs remain notably lacking.

The FDA Adverse Event Reporting System (FAERS) serves as a national post-marketing surveillance tool and one of the world’s largest pharmacovigilance databases, designed to systematically capture spontaneous safety reports for therapeutic agents (9, 10). Functioning as a critical spontaneous reporting system, this continuously updated, quarterly database (11) captures diverse safety signals, including clinician-reported AEs, medication error documentation, and product quality complaints. For the past few years, many drug safety profile studies based on the FAERS database have been published, affirming the reliability of this resource (12, 13). Using disproportionality analysis based on the FAERS database, we identified post-marketing safety signals associated with pexidartinib. This study detected AEs not previously documented in FDA-approved prescribing information. Importantly, these findings provide critical insights for optimizing therapeutic monitoring and enhancing pharmacovigilance strategies in clinical practice.

2 Patients and methods

2.1 Data sources and procedures

Available data related to pexidartinib were extracted and analyzed from the FAERS database (https://fis.fda.gov/extensions/FPD-QDE-FAERS/FPD-QDE-FAERS.html). There was no need for specific ethical approval and informed consent, as no direct human intervention or human sample collection was required.

The FAERS dataset comprises seven sections containing demographic and management information, drug information, adverse drug reaction information, patient outcomes, reporting sources, start and end dates of treatment with reported drugs, indications, and deleted cases. Since pexidartinib was approved in August 2019, we included data from the third quarter of 2019 through the second quarter of 2023. Given the potential for duplicate entries in the FAERS database, we performed a rigorous deduplication process. Specifically, we manually reviewed reports to remove entries with lower PRIMARYIDs when the CASEIDs are the same. Moreover, we further eliminated records listed in the deleted case file. We then identified pexidartinib-associated cases in both the “drugname” and “prod_ai” columns using “pexidartinib” and “SUNOSI” in the “DRUG” files.

Pexidartinib-related cases were identified in both the “drugname” and “prod_ai” fields of the DRUG file using the terms “pexidartinib” and “SUNOSI”. Adverse event dates (EVENT_DT) were obtained from the DEMO file, and therapy start dates (START_DT) were obtained from the THER file. Time-to-onset (TTO) was calculated as the difference between EVENT_DT and START_DT. Records were included only if both dates were valid and correctly formatted (YYYYMMDD), and reports with incomplete dates or implausible sequences (i.e., EVENT_DT earlier than START_DT) were excluded. TTO analysis was conducted based on medians, quartiles, and the Weibull shape parameter (WSP) test. The WSP analysis was conducted using the Minitab statistical software (v20.0; Minitab LLC, State College, PA, USA).

All reported AEs were coded using the Medical Dictionary for Regulatory Activities version 26.0 (MedDRA 26.0). The MedDRA terminology is structured hierarchically into five levels: system organ class (SOC), high-level group term (HLGT), high-level term (HLT), preferred term (PT), and lowest-level term (LLT). This study focused on identifying all pexidartinib-related AEs recorded in the adverse reaction files of FAERS. Events were systematically classified and analyzed at both the SOC and PT levels to assess their distribution and severity.

In FAERS, the drug role is designated by the reporter using one of three codes: 1 means primary suspect, 2 means concomitant, and 3 means interacting. To improve analytical specificity, we included only records in which pexidartinib was designated as the primary suspect (code 1). This strategy was adopted to enhance the accuracy and reliability of the safety signal detection.

2.2 Statistical analysis

Disproportionality analysis is a crucial technique in pharmacovigilance studies, serving a vital role in identifying potential signals indicating AEs associated with a drug (14). This methodology involves a comparative assessment of the frequency of AEs linked to a specific drug relative to the occurrence of AEs related to all other medications. Fundamentally, it relies on the concept that a signal emerges during data extraction when the incidence rate of a particular AE for a given drug significantly exceeds the background occurrence rate observed across the entire database. This deviation from the norm must exceed a predetermined threshold or set of criteria to be considered statistically significant.

In our analysis, we employed both frequentist and Bayesian approaches within the framework of disproportionality analysis. This dual approach enabled us to explore the association between a drug and a specific AE. We used the reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS) algorithms to quantify the signals of pexidartinib-related AEs (15).

In this study, a signal was considered valid only when the criteria of all four algorithms were simultaneously met. For further details regarding the mathematical equations and specific threshold values for each algorithm, the readers are referred to Supplementary Table S1. All disproportionality measures were derived from a standard 2 × 2 contingency table, as shown in Supplementary Table S2.

3 Results

3.1 Characteristics of AE reports

Throughout the course of the study, a total of 7,168,342 AE reports were initially scrutinized. Following meticulous deduplication of duplicate entries, a refined dataset of 668 reports directly associated with pexidartinib remained, as depicted in Figure 1.

Figure 1
Flowchart depicting the process of analyzing adverse event reports related to pexidartinib. It starts with three datasets: DRUG, DEMO, and REAC. DEMO data has duplicates removed. Subsequently, reports are categorized into adverse event reports and PTs induced by pexidartinib. The analysis methods include reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS). Outcomes considered are indications, outcome events and incidence, onset time of events, and clinical characteristics.

Figure 1. The flow diagram of selecting pexidartinib-related AEs from FAERS database. DEMO, demographic file; DRUG, drug file; REAC, reaction file; PS, primary suspect; AEs, adverse events; FAERS, Food and Drug Administration Adverse Event Reporting System.

To characterize patients who experienced AEs related to pexidartinib, their baseline demographics and clinical features are summarized in Table 1. Of the patients who experienced AEs linked to pexidartinib collected from FAERS, the majority was female (n = 340, 60.18%), adult (n = 161, 84.29%), and patients with TGCT or PVNS (n = 468, 82.69%). Our analysis of the top five co-administered drugs in pexidartinib-related AE cases identified amlodipine, vitamin D3, famotidine, ondansetron (Zofran), and oxycodone as the most frequently reported agents. Regarding serious clinical outcomes, hospitalization was the most frequently reported (n = 64, 41.56%), followed by death (n = 13, 8.44%). The median TTO of AEs was 20 days (interquartile range: 6.5–211 days). With the exception of the first and second quarters of 2023 and the third and fourth quarters of 2019, the highest number of AE reports was recorded in 2022 (n = 190).

Table 1
www.frontiersin.org

Table 1. Clinical characteristics of reports with pexidartinib from the FAERS database.

3.2 Disproportionality analysis

The signal reports for pexidartinib at the SOC level are presented in Table 2. Adverse event occurrences linked to pexidartinib encompass a wide spectrum of 26 distinct organ systems. Among these, the most frequently reported SOCs were general disorders and administration site conditions (SOC: 10018065, n = 453), skin and subcutaneous tissue disorders (SOC: 10040785, n = 373), and injury, poisoning, and procedural complications (SOC: 10022117, n = 324). Notably, “skin and subcutaneous tissue disorders” exhibited positive signal detection across all four algorithms. In contrast, “general disorders and administration site conditions” and “injury, poisoning, and procedural complications” generated positive signals only with the PRR method, but not with the ROR, BCPNN, and MGPS methods.

Table 2
www.frontiersin.org

Table 2. Signal strength of reports of pexidartinib at the SOC level in FAERS database.

Additionally, disproportionality analysis was performed at the PT level, as presented in Table 3. A comparison was made between the detected PTs and the adverse reactions listed in the official prescribing information, with an asterisk (*) used to indicate events not mentioned in the label. The unlabeled AEs not previously documented in FDA-approved information included photosensitivity reaction, dysmenorrhea, oligomenorrhea, increased gamma-glutamyltransferase, decreased blood iron, increased blood calcium, prolonged prothrombin time, increased red cell distribution width, hepatitis A, seasonal allergy, vanishing bile duct syndrome, cholecystitis, hunger, soft feces, eye color change, and blepharospasm.

Table 3
www.frontiersin.org

Table 3. Top significant signals on the PT level.

4 Discussion

Our pharmacovigilance study provides the first systematic characterization of pexidartinib’s post-marketing safety profile using real-world evidence from the FAERS database. Analysis of AEs’ temporal trends revealed a progressive increase in reports over time, likely attributable to expanded clinical use and enhanced pharmacovigilance awareness following FDA approval. We identified a higher frequency of AE reports among adult women in FAERS, which may reflect the known higher prevalence of TGCT in women. TGCT is more common in women than men, and the mean age at diagnosis is 35–50 years (1, 16). However, due to the absence of an accurate number of patients using pexidartinib, prospective population-based studies with standardized AE ascertainment remain necessary to establish robust risk stratification models.

The results of our study underscore a clustering of common SOCs around “general disorders and administration site conditions”, “injury, poisoning, and procedural complications”, “skin and subcutaneous tissue disorders”, and investigations, as well as gastrointestinal disorders. Additionally, frequently reported PTs associated with pexidartinib include fatigue, hair color changes, product dose omission issues, nausea, pruritus, increased aspartate aminotransferase, and increased alanine aminotransferase. Notably, these AEs are in accordance with information provided in the FDA’s drug label and previous research on pexidartinib. For example, a randomized phase 3 clinical trial of pexidartinib versus placebo for advanced TGCT identified that hair color changes (67%), fatigue (54%), increased aspartate aminotransferase (39%), nausea (38%), increased alanine aminotransferase (28%), and dysgeusia (25%) were the most frequent pexidartinib-associated AEs (6). A study of long-term outcomes of pexidartinib in TGCT revealed that “hair color changes” was the most frequent AE (17). A. Vaynrub et al. concluded that the most common AEs associated with pexidartinib are mild hypopigmentation of the hair and transient aminotransferase elevation (18).

Beyond confirming established safety signals, our pharmacovigilance analysis identified 16 novel adverse drug reactions not currently documented in pexidartinib’s FDA labeling. These include photosensitivity reaction, dysmenorrhea, oligomenorrhea, increased gamma-glutamyltransferase, decreased blood iron, increased blood calcium, prolonged prothrombin time, increased red cell distribution width, hepatitis A, seasonal allergy, vanishing bile duct syndrome, cholecystitis, hunger, soft feces, eye color change, and blepharospasm. Sorbarikor Piawah et al. (19) reported a case of severe drug-induced liver injury requiring liver transplantation due to vanishing bile duct syndrome after exposure to pexidartinib. Beyond vanishing bile duct syndrome previously reported in the literature, most AEs associated with pexidartinib identified in our study represent novel safety findings. These newly detected AEs warrant further validation to ensure the safe clinical use of pexidartinib. Importantly, the mechanisms underlying most unexpected AEs remain unstudied, necessitating dedicated mechanistic investigations.

Occurrences such as therapy cessation, therapy change, tumor excision, product dose omission issue, product dose omission in error, wrong dose, no adverse event, and treatment non-compliance were not classified as pexidartinib-induced AEs. Pexidartinib was often used as therapy for TGCT preoperation and postoperation (20, 21), and a few patients have reported drug dosage reduction from the initial dose (20). Therefore, we posit that treatment discontinuation, therapy modification, and tumor resection are integral components of managing the primary disease. Additionally, intentional dose omissions and unintentional dosing errors were predominantly associated with supply chain disruptions. Recognizing these distinctions is essential for holistic patient assessment and optimized therapeutic management.

Gamma-glutamyltransferase is one of the key enzymes involved in the transformation function of hepatocytes, and it is also an important link in glutathione metabolism (22). Therefore, we postulate that elevated gamma-glutamyltransferase levels may reflect pexidartinib-induced hepatotoxicity. Notably, decreased hemoglobin is a documented adverse reaction in the drug’s prescribing information. Significantly, we identified reduced serum iron and increased red cell distribution width as previously unreported AEs potentially causally linked to hemoglobin reduction. Increased blood calcium, an off-label AE of pexidartinib, may be associated with decreased phosphate mentioned in pexidartinib’s prescribing information (23, 24). The relationship between these clinical manifestations should attract the attention of patients and physicians, and the mechanism warrants further research.

Our analysis identified dysmenorrhea as a previously unreported adverse drug reaction associated with pexidartinib. This finding holds particular significance given the established association between anticoagulant-induced platelet dysfunction and menstrual abnormalities. A multicenter, single-arm, open-label, phase 2a, proof-of-concept trial (25) revealed that menorrhagia and dysmenorrhea are adverse drug reactions to rivaroxaban. Moreover, prolonged prothrombin time is detected as a kind of AE of pexidartinib. In a number of studies, prolonged prothrombin time is a common adverse reaction to tigecycline (26, 27). Therefore, the coagulation dysfunction induced by pexidartinib is worthy of attention and needs further exploration.

While leveraging that the FAERS database enables large-scale pharmacoepidemiologic surveillance, our study design inherits limitations inherent to spontaneous reporting systems that warrant cautious interpretation. First, because of the voluntary nature of the FAERS database, it has some inherent selection bias, such as the ethnicity and geography of the reported cases, the timing of approval and market penetration of different drugs, the level of public awareness of specific adverse reactions, and the fact that not all reports of serious adverse reactions occurring are being collected (28). Second, polypharmacy, comorbidities, and underlying disease severity pose a challenge in addressing confounding factors. The absence of comprehensive clinical information, such as interventions following AEs and the health status of the reporting patient, prevents the impact of confounding factors on the determination of causality between AEs and pexidartinib from being mitigated (29). Third, due to the absence of an accurate number of patients using pexidartinib, it remains impossible to calculate the true incidence rates for each AE (30).

Notwithstanding these limitations, the FAERS database remains a cornerstone resource in global pharmacovigilance, providing critical post-marketing surveillance insights through its unparalleled scale (30). It is crucial to acknowledge that although data mining techniques cannot compensate for the inherent limitations of a spontaneous reporting system, the combined utilization of the large-scale database and case reports remains an effective approach for delving into adverse drug reactions (31). The insights gleaned from the FAERS database provide valuable preliminary information for further investigation and prospective studies. Although the findings should be interpreted with caution, they represent contributions to a broader understanding of the safety profile of pexidartinib and its potential implications in clinical practice.

5 Conclusion

Through a comprehensive and systematic pharmacoepidemiologic study based on the FAERS database, we characterized the post-marketing safety profile linked to pexidartinib. Most of the AEs we identified closely align with the information provided in the FDA’s official prescribing guidelines. Notably, our study unveiled 16 unexpected AEs, expanding upon the existing knowledge derived from pre-marketing clinical trials. Although the limitations of the FAERS database are difficult to overcome, these findings are particularly valuable for ensuring drug safety. Further prospective clinical trials are warranted to establish a definitive connection between pexidartinib and these newly identified AEs. This study demonstrates the utility of pharmacovigilance databases in complementing clinical trial data for comprehensive drug safety assessment.

Data availability statement

The raw data supporting the conclusions of this article will be made available by the authors, without undue reservation.

Author contributions

YL: Writing – original draft, Project administration, Conceptualization, Writing – review & editing. XZ: Data curation, Methodology, Writing – review & editing. LL: Writing – review & editing, Investigation, Validation. MC: Methodology, Data curation, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

Over the course of our research and writing this paper, we are thankful to all authors.

Conflict of interest

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.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2025.1594585/full#supplementary-material

References

1. Stacchiotti S, Dürr HR, Schaefer I-M, Woertler K, Haas R, Trama A, et al. Best clinical management of tenosynovial giant cell tumour (TGCT): A c onsensus paper from the community of experts. Cancer Treat Rev. (2023) 112:102491. doi: 10.1016/j.ctrv.2022.102491

PubMed Abstract | Crossref Full Text | Google Scholar

2. West RB, Rubin BP, Miller MA, Subramanian S, Kaygusuz G, Montgomery K, et al. A landscape effect in tenosynovial giant-cell tumor from activation of CSF1 expression by a translocation in a minority of tumor cells. Proc Natl Acad Sci U.S.A. (2006) 103:690–5. doi: 10.1073/pnas.0507321103

PubMed Abstract | Crossref Full Text | Google Scholar

3. Lamb YN. Pexidartinib: first approval. Drugs. (2019) 79:1805–12. doi: 10.1007/s40265-019-01210-0

PubMed Abstract | Crossref Full Text | Google Scholar

4. Roskoski R Jr. Properties of FDA-approved small molecule protein kinase inhibitors: A 2020 update. Pharmacol Res. (2020) 152:104609. doi: 10.1016/j.phrs.2019.104609

PubMed Abstract | Crossref Full Text | Google Scholar

5. Wen J, Wang S, Guo R, and Liu D. CSF1R inhibitors are emerging immunotherapeutic drugs for cancer treat ment. Eur J Med Chem. (2023) 245:114884. doi: 10.1016/j.ejmech.2022.114884

PubMed Abstract | Crossref Full Text | Google Scholar

6. Tap WD, Gelderblom H, Palmerini E, Desai J, Bauer S, Blay JY, et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): a randomised phase 3 trial. Lancet. (2019) 394:478–87. doi: 10.1016/S0140-6736(19)30764-0

PubMed Abstract | Crossref Full Text | Google Scholar

7. Van De Sande M, Tap WD, Gelhorn HL, Ye X, Speck RM, Palmerini E, et al. Pexidartinib improves physical functioning and stiffness in patients with tenosynovial giant cell tumor: results from the ENLIVEN randomized clinical trial. Acta Orthop. (2021) 92:493–9. doi: 10.1080/17453674.2021.1922161

PubMed Abstract | Crossref Full Text | Google Scholar

8. Yin O, Zahir H, French J, Polhamus D, Wang X, van de Sande M, et al. Exposure-response analysis of efficacy and safety for pexidartinib in patients with tenosynovial giant cell tumor. CPT Pharmacometrics Syst Pharmacol. (2021) 10:1422–32. doi: 10.1002/psp4.12712

PubMed Abstract | Crossref Full Text | Google Scholar

9. Sakaeda T, Tamon A, Kadoyama K, and Okuno Y. Data mining of the public version of the FDA Adverse Event Reporting S ystem. Int J Med Sci. (2013) 10:796–803. doi: 10.7150/ijms.6048

PubMed Abstract | Crossref Full Text | Google Scholar

10. Chen C, Wu B, Zhang C, and Xu T. Immune-related adverse events associated with immune checkpoint inhibi tors: An updated comprehensive disproportionality analysis of the FDA adverse event reporting system. Int Immunopharmacol. (2021) 95:107498. doi: 10.1016/j.intimp.2021.107498

PubMed Abstract | Crossref Full Text | Google Scholar

11. Khaleel MA, Khan AH, Ghadzi SMS, Adnan AS, and Abdallah QM. A standardized dataset of a spontaneous adverse event reporting system. Healthcare (Basel). (2022) 10:420. doi: 10.3390/healthcare10030420

PubMed Abstract | Crossref Full Text | Google Scholar

12. Yu RJ, Krantz MS, Phillips EJ, and Stone CA Jr. Emerging causes of drug-induced anaphylaxis: A review of anaphylaxis-A ssociated reports in the FDA adverse event reporting system (FAERS). J Allergy Clin Immunol Pract. (2021) 9:819–829.e812. doi: 10.1016/j.jaip.2020.09.021

PubMed Abstract | Crossref Full Text | Google Scholar

13. Yang Z, Lv Y, Yu M, Mei M, Xiang L, Zhao S, et al. GLP-1 receptor agonist-associated tumor adverse events: A real-world s tudy from 2004 to 2021 based on FAERS. Front Pharmacol. (2022) 13:925377. doi: 10.3389/fphar.2022.925377

PubMed Abstract | Crossref Full Text | Google Scholar

14. Michel C, Scosyrev E, Petrin M, and Schmouder R. Can disproportionality analysis of post-marketing case reports be used for comparison of drug safety profiles? Clin Drug Invest. (2017) 37:415–22. doi: 10.1007/s40261-017-0503-6

PubMed Abstract | Crossref Full Text | Google Scholar

15. Shu Y, He X, Liu Y, Wu P, and Zhang Q. A real-world disproportionality analysis of olaparib: data mining of the public version of FDA adverse event reporting system. Clin Epidemiol. (2022) 14:789–802. doi: 10.2147/CLEP.S365513

PubMed Abstract | Crossref Full Text | Google Scholar

16. Mastboom MJL, Verspoor FGM, Verschoor AJ, Uittenbogaard D, Nemeth B, Mastboom WJB, et al. Higher incidence rates than previously known in tenosynovial giant cel l tumors. Acta Orthop. (2017) 88:688–94. doi: 10.1080/17453674.2017.1361126

PubMed Abstract | Crossref Full Text | Google Scholar

17. Gelderblom H, Wagner AJ, Tap WD, Palmerini E, Wainberg ZA, Desai J, et al. Long-term outcomes of pexidartinib in tenosynovial giant cell tumors. Cancer. (2021) 127:884–93. doi: 10.1002/cncr.33312

PubMed Abstract | Crossref Full Text | Google Scholar

18. Vaynrub A, Healey JH, Tap W, and Vaynrub M. Pexidartinib in the management of advanced tenosynovial giant cell tum or: focus on patient selection and special considerations. Onco Targets Ther. (2022) 15:53–66. doi: 10.2147/OTT.S345878

PubMed Abstract | Crossref Full Text | Google Scholar

19. Piawah S, Hyland C, Umetsu SE, Esserman LJ, Rugo HS, and Chien AJ. A case report of vanishing bile duct syndrome after exposure to pexida rtinib (PLX3397) and paclitaxel. NPJ Breast Cancer. (2019) 5:17. doi: 10.1038/s41523-019-0112-z

PubMed Abstract | Crossref Full Text | Google Scholar

20. Lin F, Kwong WJ, Pan I, Ye X, Dai D, and Tap W. Real-world patient experience of pexidartinib for tenosynovial giant-C ell tumor. Oncologist. (2024) 2023:oyad282. doi: 10.1093/oncolo/oyad282

PubMed Abstract | Crossref Full Text | Google Scholar

21. Li Z, Ding Y, Liu J, Wang J, Mo F, Wang Y, et al. Depletion of tumor associated macrophages enhances local and systemic platelet-mediated anti-PD-1 delivery for post-surgery tumor recurrence treatment. Nat Commun. (2022) 13:1845. doi: 10.1038/s41467-022-29388-0

PubMed Abstract | Crossref Full Text | Google Scholar

22. Ke Q, Xiang F, Xiao C, Huang Q, Liu X, Zeng Y, et al. Exploring the clinical value of preoperative serum gamma-glutamyl tran sferase levels in the management of patients with hepatocellular carci noma receiving postoperative adjuvant transarterial chemoembolization. BMC Cancer. (2021) 21:1117. doi: 10.1186/s12885-021-08843-z

PubMed Abstract | Crossref Full Text | Google Scholar

23. Granjon D, Bonny O, and Edwards A. Coupling between phosphate and calcium homeostasis: a mathematical mod el. Am J Physiol Renal Physiol. (2017) 313:F1181–99. doi: 10.1152/ajprenal.00271.2017

PubMed Abstract | Crossref Full Text | Google Scholar

24. Trautvetter U, Ditscheid B, Jahreis G, and Glei M. Calcium and phosphate metabolism, blood lipids and intestinal sterols in human intervention studies using different sources of phosphate as supplements-pooled results and literature search. Nutrients. (2018) 10:936. doi: 10.3390/nu10070936

PubMed Abstract | Crossref Full Text | Google Scholar

25. Weishaupt C, Strölin A, Kahle B, Kreuter A, Schneider SW, Gerss J, et al. Anticoagulation with rivaroxaban for livedoid vasculopathy (RILIVA): a multicentre, single-arm, open-label, phase 2a, proof-of-concept trial. Lancet Haematol. (2016) 3:e72–79. doi: 10.1016/S2352-3026(15)00251-3

PubMed Abstract | Crossref Full Text | Google Scholar

26. Guo M, Liang J, Li D, Zhao Y, Xu W, Wang L, et al. Coagulation dysfunction events associated with tigecycline: a real-wor ld study from FDA adverse event reporting system (FAERS) database. Thromb J. (2022) 20:12. doi: 10.1186/s12959-022-00369-z

PubMed Abstract | Crossref Full Text | Google Scholar

27. Leng B, Shen C, Gao T, Zhao K, Zhao X, Guo Y, et al. Incidence, characteristics and risk factors of hypofibrinogenemia asso ciated with tigecycline: A multicenter retrospective study in China. Front Pharmacol. (2022) 13:943674. doi: 10.3389/fphar.2022.943674

PubMed Abstract | Crossref Full Text | Google Scholar

28. Zhou C, Peng S, Lin A, Jiang A, Peng Y, Gu T, et al. Psychiatric disorders associated with immune checkpoint inhibitors: a pharmacovigilance analysis of the FDA Adverse Event Reporting System (FAERS) database. EClinicalMedicine. (2023) 59:101967. doi: 10.1016/j.eclinm.2023.101967

PubMed Abstract | Crossref Full Text | Google Scholar

29. Feng Z, Li X, Tong WK, He Q, Zhu X, Xiang X, et al. Real-world safety of PCSK9 inhibitors: A pharmacovigilance study based on spontaneous reports in FAERS. Front Pharmacol. (2022) 13:894685. doi: 10.3389/fphar.2022.894685

PubMed Abstract | Crossref Full Text | Google Scholar

30. Zhou J, Wei Z, Xu B, Liu M, Xu R, and Wu X. Pharmacovigilance of triazole antifungal agents: Analysis of the FDA a dverse event reporting system (FAERS) database. Front Pharmacol. (2022) 13:1039867. doi: 10.3389/fphar.2022.1039867

PubMed Abstract | Crossref Full Text | Google Scholar

31. Shu Y, Ding Y, He X, Liu Y, Wu P, and Zhang Q. Hematological toxicities in PARP inhibitors: A real-world study using FDA adverse event reporting system (FAERS) database. Cancer Med. (2023) 12:3365–75. doi: 10.1002/cam4.5062

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: disproportionality analysis, FAERS database, pexidartinib, real-world adverse events, tenosynovial giant cell tumor

Citation: Lin Y, Zheng X, Lin L and Chen M (2025) Adverse events of pexidartinib for the treatment of TGCT: a real-world disproportionality analysis using FDA Adverse Event Reporting System database. Front. Oncol. 15:1594585. doi: 10.3389/fonc.2025.1594585

Received: 16 March 2025; Accepted: 28 July 2025;
Published: 18 August 2025.

Edited by:

Jianrong Zhang, Cancer Council Victoria, Australia

Reviewed by:

Ebru Haciosmanoglu Aldogan, Istanbul University Cerrahpasa, Türkiye
Abdul Rajper, Daiichi Sankyo, Inc., United States

Copyright © 2025 Lin, Zheng, Lin and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Maohua Chen, bWhjcHR5eUAxMjYuY29t; Li Lin, bGlubGl6enVAMTYzLmNvbQ==

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.