Next Article in Journal
3D-Printing of Capsule Devices as Compartmentalization Tools for Supported Reagents in the Search of Antiproliferative Isatins
Next Article in Special Issue
Special Issue “Rheumatic Diseases: Pathophysiology, Targeted Therapy, Focus on Vascular and Pulmonary Manifestations 2022”
Previous Article in Journal
Coupling of NIR Spectroscopy and Chemometrics for the Quantification of Dexamethasone in Pharmaceutical Formulations
Previous Article in Special Issue
Nano Methotrexate versus Methotrexate in Targeting Rheumatoid Arthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Nintedanib in Idiopathic Pulmonary Fibrosis: Tolerability and Safety in a Real Life Experience in a Single Centre in Patients also Treated with Oral Anticoagulant Therapy

1
Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
2
Biostatistics Unit, Department of Medical Sciences, University of Trieste, 34149 Trieste, Italy
3
Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester M6 8HD, UK
4
24ORE Business School, Via Monte Rosa, 91, 20149 Milano, Italy
5
Department of Radiology, Cattinara Hospital, University of Trieste, 34127 Trieste, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Pharmaceuticals 2023, 16(2), 307; https://doi.org/10.3390/ph16020307
Submission received: 18 December 2022 / Revised: 30 January 2023 / Accepted: 13 February 2023 / Published: 16 February 2023

Abstract

:
Idiopathic pulmonary fibrosis (IPF) is a rare and severe disease with a median survival of ~3 years. Nintedanib (NTD) has been shown to be useful in controlling interstitial lung disease (ILD) in IPF. Here we describe the experience of NTD use in IPF in a real-life setting. Objective. Our objective was to examine the safety profile and efficacy of nintedanib even in subjects treated with anticoagulants. Clinical data of patients with IPF treated with NTD at our center were retrospectively evaluated at baseline and at 6 and 12 months after the introduction of NTD. The following parameters were recorded: IPF clinical features, NTD tolerability, and pulmonary function tests (PFT) (i.e., Forced Vital Capacity (FVC) and carbon monoxide diffusing capacity (DLCO)). In total, 56 IPF patients (34% female and 66% male, mean onset age: 71 ± 11 years, mean age at baseline: 74 ± 9 years) treated with NTD were identified. At enrollment, HRCT showed an UIP pattern in 45 (80%) and a NSIP in 11 (20%) patients. For FVC and FEV1 we found no significant change between baseline and 6 months, but for DLCO we observed a decrease (p = 0.012). We identified a significant variation between baseline and 12 months for FEV1 (p = 0.039) and for DLCO (p = 0.018). No significant variation was observed for FVC. In the cohort, 18 (32%) individuals suspended NTD and 10 (18%) reduced the dosage. Among individuals that suspended the dosage, 14 (78%) had gastrointestinal (GI) collateral effects (i.e., diarrhea being the most common complaint (67%), followed by nausea/vomiting (17%) and weight loss (6%). Bleeding episodes have also not been reported in patients taking anticoagulant therapy. (61%). One patient died within the first 6 months and two subjects died within the first 12 months. In a real-life clinical scenario, NTD may stabilize the FVC values in IPF patients. However, GI side effects are frequent and NTD dose adjustment may be necessary to retain the drug in IPF patients. This study confirms the safety of NTD, even in patients treated with anticoagulant drugs.

1. Introduction

Idiopathic pulmonary fibrosis (IPF) is a rare and irreversible chronic interstitial pneumonia characterized by abnormal deposition of extracellular matrix in the lung parenchyma [1,2,3]. Aberrant lung repair leads to repetitive tissue scar formation, alveolar structure abnormalities with significant impairment of alveolar gas exchange, and reduced lung function (forced vital capacity (FVC) and carbon monoxide diffusing capacity (DLCO)) [2,3]. IPF is the most common of its class, being approximately 50–60% of all idiopathic interstitial pneumonia, with an estimated incidence of 3–9 cases per 100,000 individuals in Europe and North America [1,2,3]. IPF patients present a nonspecific symptomatology, which is the fundamental cause of the delay in diagnosis. It causes progressive exertional dyspnea, nonproductive cough and asthenia. These symptoms cause a reduction in daily physical activity and muscle strength; patients suffer a precarious quality of life, which can be responsible of social isolation, depressive and anxiety disorders and increased levels of dependence. The evolution of the disease is often associated with different cardiopulmonary complications. All of these elements ends in a situation that is difficult to manage for both patients and their caregivers [1,2,3,4,5,6]. Another cause for the delay in the diagnosis of IPF is that this is an entity that can be easily confused with other respiratory pathologies requiring multidisciplinary assessment by the pulmonology, radiology and pathological anatomy services, thereby using more healthcare resources [3,4,5,6,7,8,9,10,11]. Although IPF exhibits a varied nature [4], clinical-pathologic and histologic patterns allow the description of so-called habitual interstitial pneumonia (UIP) [5], defined radiologically on high-resolution computed tomography (HRCT) by a bilateral peripheral distribution of fibrosis and most pronounced at the bases of the lungs. The pathogenesis of these patterns is not entirely clear. The environment (cigarette smoke, air pollution, inhalation of wood and metal dust, GERD) plays a significant role in the development of IPF, although without an established relationship to genetics. The single nucleotide polymorphism in the mucin 5B promoter region (MUC5B) is the most relevant genetic risk factor for sporadic and familial IPF, but the mechanisms in lung epithelial cells still need to be better understood. The interaction between variable expression of genetic polymorphisms seems to be the origin of the disease, which is clearly associated also with changes related to cellular aging and exposure to environmental factors, smoking, industrial powders, chronic gastric microaspiration, viral infections and possibly alterations in the lung microbiome [1,3,12,13,14,15,16,17,18,19]. The repetitive exposures aberrantly activate the alveolar epithelial cells of genetically susceptible individuals, promoting epitelial apoptosis, recruitment of mesenchymal cells and increased vascular permeability. Unregulated epithelial/mesenchymal interaction results in the secretion of various profibrotic cytokines, metalloproteinases and procoagulant mediators, which promote uncontrolled migration and proliferation, and differentiation in fibroblasts to myofibroblasts as well as fibrosis in the extracellular matrix [20,21,22,23,24,25,26,27,28]. The main pro-inflammatory cytokines involved in fibrosis are tumour necrosis factor (TNF)-α and interleukin (IL)-1, as well as some fibrous factors such as transforming growth factor (TGF)-β and platelet-derived growth factor (PDGF) [28,29,30,31,32].
Antifibrotic therapies (pirfenidone, nintedanib) can reduce respiratory function decline and improve survival of IPF patients, but they fail to halt disease progression; therefore, prognosis remains poor, with a median survival usually 2–3 years from the time of diagnosis [6,7].
Nintedanib (ex-BIBF 1120) is an intracellular inhibitor of multiple receptor tyrosine kinases (RTKs) and non-receptor tyrosine kinases (nRTKs). It competitively binds to the adenosine triphosphate (ATP) binding pocket of its targets, which include platelet-derived growth factor (PDGFRα and PDGFRβ), fibroblast growth factor receptor (FGFR1, FGFR2, FGFR3), vascular endothelial growth factor (VEGFR1, VEGFR2, VEGFR3), colony-stimulating factor receptor 1 (CSF1R), and FMS-like tyrosine kinase three (FLT3), all of which are critical for fibroblast proliferation, migration, and differentiation [8]. As a result of this broad inhibition, pro-fibrotic and pro-angiogenic processes are reduced and, as a result, fibroblast and myofibroblast number and activity are reduced, as well as ECM (Extracellular Matrix) production, ultimately rebalancing pro-fibrotic and antifibrotic processes [9]. The current indications for the antifibrotic effect of nintedanib are idiopathic pulmonary fibrosis (IPF) from 2014, systemic sclerosis-associated interstitial lung disease (Sc-ILD) from 2019 [5], further confirmed by the SENSCIS and SENSCIS-ON clinical trials [6,7] and progressive pulmonary fibrosis (PPF) from 2020 [10]. In addition, nintedanib is also being used and studied for cancers (lung, ovary, kidney, colorectal, and liver) [11,12,13]; in this context, new modes of administration, such as inhalation [12], are being investigated. The efficacy of nintedanib′s antifibrotic properties first emerged in the phase II TOMORROW study [14] and was later confirmed by the phase III IMPULSIS-1 and IMPULSIS-2 studies [15]. Nintedanib is administered orally at a dosage of 150 mg twice daily or, in cases of poor tolerance, 100 mg twice daily. It reaches maximum plasma concentrations in about 2–4 h depending on whether or not it is taken with food, and reaches steady-state plasma concentrations within a week. The absolute bioavailability of nintedanib 100 mg is 4.7 percent. Biotransformation of nintedanib mainly involves esterase-mediated hydrolysis, followed by glucuronidation, finally yielding glucuronide BIBF 1202. A minor role is played by CYP pathways, particularly CYP 3A4. Caution should be exercised with concomitant administration of nintedanib with CYP3A4 and P-gp inhibitors (e.g., ketoconazole) with inducers (e.g., rifampin), which may result in increased or decreased exposure to nintedanib, respectively. The main route of elimination is fecal/biliary excretion (about 93% of the administered dose), while renal excretion contributes little to the total clearance [1,14,15,16,17,18]; the resulting half-life is 9.5 h. The main adverse effects include, first and foremost, diarrhea (61.5 percent of cases), but also increased liver enzymes (which should be checked before initiating therapy and then monitored), abdominal pain, nausea, vomiting, and weight loss, all of which are generally manageable by reducing the dose (200 mg/day), discontinuing treatment, and applying symptomatic measures (e.g., loperamide) [19,20,21,22,23,24,25,26,27].
The purpose of our retrospective study was to examine the safety profile and efficacy of nintedanib in patients with IPF, including in subjects treated with anticoagulants.

2. Results

In this single-center retrospective study, 56 patients with IPF were included: 19 (34%) males, mean age of onset: 71 ± 11 years, mean age at baseline: 74 ± 9 years (Table 1). The prevalence of coexisting GERD was 12 out of 56 patients, with a comparable prevalence of PPI use. Among all comorbidities, smoking was largely prevalent (34 out of 56). Other notable comorbidities were COPD (5.5%) and cancer (15%). The most frequent clinical symptoms were cough (80% of patients) and dyspnea (50%). Concomitant medications were considered: steroid use was the most frequent (40%), especially at a high steroid dosage of >15 mg (21%). A minority of patients were on immunosuppressants, specifically two patients on hydroxychloroquine, three on mycophenolate mofetil, one on azathioprine, and one on rituximab.
All 56 patients started NTD therapy at baseline, of whom 25 (45%) completed the 12-month follow-up at full dose, 10 (18%) reduced the dose and 18 (32%) discontinued the drug. In addition, two patients (3.6%) died within 12 months and one (1.8%) died within 6 months. The main reason for treatment reduction was gastrointestinal intolerance, particularly diarrhea (six patients), but also nausea (one patient), and abdominal pain (one patient). Second, liver disruption resulted in four NTD dose reductions. Regarding discontinuation, 14 of 18 discontinued treatment due to gastrointestinal intolerance, particularly diarrhea (12 patients) but also nausea/vomiting (3 patients). One patient reported weight loss and consequently discontinued the drug. No hemorrhagic events were reported in our population even in patients taking anticoagulants.
Pulmonary function test (PFT) data were included, specifically Forced Vital Capacity (FVC), Forced Expiratory Volume in 1 s (FEV1), and Diffusion Lung CO (DLCO) were recorded at baseline, 6 months, and 12 months (Table 1). FVC measurements at baseline and 6 months were available for 31 patients, with no statistically significant differences (p-value 0.6, Figure 1). FEV1 measurements at baseline and 6 months were available for 29 subjects, with no statistically significant differences (p-value > 0.9). In contrast, DLCO measurements, available at baseline and at 6 months for 25 patients, revealed a statistically significant difference (p-value 0.012).
The same evaluation was made for differences between measurements at baseline and 12 months, which for FVC was available for 22 patients, for FEV1was available for 22 patients, and for DLCO was available for 20 patients (Figure 2 and Figure 3). In this case, a statistically significant difference was detected for both FEV1 and DLCO (p-value, respectively, 0.039 and 0.018, Figure 2), while difference in FVC between baseline and 12 months expressed no statistical significance (p-value 0.2). In Figure 3, it is possible to observe the trend over the three time-point for subjects who had all the information available. The analysis restricted to male individuals gave similar results. Both in the 6-month and 12-month change, we only observed a significant decrease for DLCO (p = 0.018 and p = 0.020, respectively).
For FVC, six patients (19%) were identified as non-responders at 6 months and three (14%) as non-responders at 12 months (Table 2). No association was found with smoking status, steroid use, anti-antibody use, or PPI. For DLCO, 13 (52%) were identified as non-responders at 6 months and 5 (25%) at 12 months. Similarly, in this case no association was found with smoking status, steroid use, anti-antibody use, and PPI.
Considering individuals that took NTD for at least 4 months, life status information was available for 50 of them, with median follow-up of 17 months. The overall survival was 80.1% (95% CI [68.7, 80.1]) at one year, 60.8% (95% CI [47.0, 78.6]) at two years, and 38.4% (95% CI [24.8, 59.4]) at three years (Figure 4).

3. Discussion

This retrospective single-center study aims to evaluate the role of NTD in the treatment of 56 patients with IPF. Our study confirms the efficacy of NTD in slowing disease progression in terms of stabilizing lung function without, as already known, allowing a marked improvement in fibrotic damage. Patients with IPF may have a variable clinical course also influenced by comorbidities, smoking habits first and foremost, which affected 61% of the enrolled patients and by any NTD-associated therapies; however, the existence of a phenotype associated with a rapid decline in FVC leading rapidly to death, which occurred within a year in 5% of the cases in our cluster, should also be kept in mind [22,23,24,25]. In the studies performed in the 1990s and the early 2000s, before the era of the new American Thoracic Society (ATS)/European Respiratory Society (ERS) classification and the introduction of antifibrotic medication, the median survival of IPF patients was 2–3 years [26,27,28]. Several recent studies demonstrated that IPF mortality is increasing in Europe [26,27,28]. The Finnish IPF registry enrolled 453 patients; at the time of diagnosis, the mean age of these patients was 73.0 years and the age varied from 44 to 91 years [26]. The 5-year survival rate was 45%. The independent predictors of survival were age and lung function at diagnosis. During 2011–2017, only a quarter of the patients had received antifibrotic medication, and the transplant rate was very low. Patients who received ≥6 months of treatment had better survival compared with those who did not receive treatment [26]. In Italy, the last official data before the COVID-19 pandemic show that in healthy people with similar age of IPF patients, a 70-year-old male today has about a 33% chance of dying by age 80, a 50% chance by age 84, and a 20% chance of reaching 90. A 70-year-old woman has a more than 80% chance of making it to age 80, a 50% chance of making it to 87 and a nearly 20% chance of making it to 93 [29].
On the SENSCIS line, about half of the patients examined were on NTD monotherapy; the remainder were on concomitant treatment with OCS and only a minority were on immunosuppressants. In our study, only 13% could be classified into this subgroup. No major adverse events were reported in this group of patients treated with this combination. This finding highlights that NTD can currently be considered a single reference therapy in the treatment of IPF. We found no association between the non-responders status and smoking status, anti-antibody, PPI, and both steroid groups, at 6 and 12 months, respectively, for all spirometry parameters. Our study considered patients without distinction of age or exclusion criteria. The number of patients enrolled was lower than that of another previous study, which examined two groups of patients divided according to age above or below 80 years, with a single collection of functional data at 12 months [24]. From the point of view of pulmonary function, the most significant data in our study concerned FEV1 values at 6 months and DLCO values at 6 and 12 months, measured for a sample of about 20 follow-up patients. As is well known, DLCO is a typically altered parameter in IPF and is closely related to the development of other comorbidities, such as pulmonary hypertension. The stabilization of diffusing capacity values after the initiation of NTD therapy confirms what is expected in terms of treatment efficacy. Moreover, most of our patients had UIP pathology at baseline at HRTC (80%), which could explain the essentially stable FVC values during follow-up. In particular, the systematic review and network meta-analysis conducted by Pitre et al. [23], highlights not only the potential of NTD in reducing FVC decline (2.92%; 1.51 to 4.14), but also its efficacy in reducing mortality (RR 0.69; 0.44 to 1.1). One noteworthy element concerns the safety of NTD therapy in enrolled patients. As in the SENSCIS study, gastrointestinal adverse effects (AE) were the main problem associated with NTD. The percentage of patients who reduced drug dosage was lower (18% vs. 48% over 12 months), while only 32% of subjects discontinued treatment. Given the expected results in terms of lung function, it was our specific intention to reduce the dosage as soon as possible (due to difficult gastrointestinal tolerance, rather than in case of increased liver function test), rather than to recommend permanent discontinuation of treatment, so as not to lose the patient during the study. The safety of this drug regimen confirmed by our study is in line with evidence from the latest work on the subject. In fact, analysis of the Latin American Registry of IPF [24] showed that NTD therapy leads to AE in a third of patients, with a non-statistically significant difference in terms of the proportion of patients with AE compared with pirfenidone. Furthermore, this study confirms the safety of NTD even in patients treated with anticoagulant drugs. The main limitation of our study is a limited number of enrolled patients, as well as the even smaller number of repeated measurements. Despite this, it was possible to obtain evidence on the efficacy of NTD in terms of progression of the disease from a pulmonary function point of view as expected. Finally, it was not possible to assess whether stabilization of lung function was associated with better quality of life because of the absence of a systematic evaluation of this parameter.
In the future, the increase in the numbers of patients enrolled, and above all the availability of PTF data—in particular about the relation to the variation of FVC over time—would make it possible to analyze the trend of lung function together with main clinical factors (e.g., using a multivariate model). This could surely help to define more precisely the effective therapeutic efficacy of NTD.

4. Materials and Methods

4.1. Study Population

Patients classified as IPF according to the 2022 ATS/ERS/JRS/ALAT Clinical Practice Guideline and who started treatment with nintedanib (NTD) were identified in our center and were retrospectively reviewed.

4.2. Data Collection

Clinical and laboratory data were collected retrospectively at baseline and at 6 and 12 months after the introduction of NTD. The following clinical characteristics were collected: sex, age, duration of illness, and clinical symptoms (fever, cough, dyspnea, arthralgia/arthritis). In addition, comorbidities including hypertension, coronary artery disease, diabetes mellitus, chronic lung disease, and rheumatic diseases were recorded. All adverse events were recorded. Ethical approval and informed patient consent were obtained from our center. Laboratory results included blood cell counts (neutrophils, lymphocytes, hemoglobin, platelets); biochemical parameters (aspartate aminotransferase, AST; lactic dehydrogenase, LDH; creatine kinase, CK); erythrocyte sedimentation rate (ESR); and C-reactive protein. Antinuclear antibodies (ANA) and extractable nuclear antigens (ENA) were also collected (Table 3).

4.3. Assessment of Imaging Involvement

The lung disease presented mainly as interstitial lung disease (ILD) with a variety of radiological pictures, which included usual interstitial pneumonia (UIP), nonspecific interstitial pneumonia (NSIP), and organizational pneumonia (OP) [4]. The evaluation of ILD in our hospital was performed by radiologists with more than 10 years of experience in chest imaging, relying mainly on clinical manifestations and respective abnormalities suggestive of ILD in high-resolution computed tomography (HRCT). During the data review, any indeterminate ILD was excluded from the analysis.

4.4. Assessment of Pulmonary Involvement

Pulmonary function tests (PFT) were performed among patients with ILD, and the following indexes were recorded, including forced vital capacity (FVC), the median forced expiratory volume in 1 s (FEV1), and diffusing capacity of the lung for carbon monoxide (DLCO). FVC, FEV1, and DLCO were described as a percentage of the predicted values for the patient’s age, sex, and height [21]. Abnormalities of PFTs were defined as predicted values of forced vital capacity (FVC) < 80% and diffusing capacity of the lung for carbon monoxide (DLCO) < 70% [22].

4.5. Statistical Analysis

Continuous variables were summarized using mean (standard deviation) or median (interquartile range) as appropriate, while categorical variables were reported as percentages. The normality of continuous variables was tested with the Shapiro–Wilk test. As for spirometry measurements, values were compared at different time points using the paired t-test or the non-parametric Wilcoxon signed-rank test when assumptions for the parametric test were not satisfied. When comparing measurements at baseline with those at 6 months, we considered only those individuals who had been taking NTDs for at least 4 months. When comparing measurements at baseline and at 2 months, we considered only individuals who had been taking NTD for at least 9 months. The comparison was carried out in the restricted sample of male subjects. For FVC, we considered a non-responder subject after 6 months of NTD if FVC decreased >5% and if differences were observed during the follow-up regarding blood test evaluations.
We considered a non-responder subject after 10 months of NTD if FVC decreased >10%. Similarly, for DLCO, we considered a non-responder subject after 6 months of NTD if DLCO decreased >6% and we considered a non-responder subject after 10 months of NTD if DLCO decreased >10% We performed a survival analysis considering the onset of NTD as a baseline and after 1 year of observation. The endpoint was death from any cause. Survival function was estimated by the Kaplan–Meier method. In all the analysis, the statistical significance was set at 0.05. Analyses were performed using R software version 4.2.1.

5. Conclusions

Our real-life study, despite the small number of patients enrolled, confirms the efficacy and safety of NTD. This therapy aims to stabilize lung function in terms of FVC values, although it is unable to achieve a marked improvement in fibrosis. Our relatively small study population confirms the effect of NTD in stabilizing FVC; furthermore, it does confirm its major role in slowing DLCO decline, which is another pivotal goal of this therapy. Despite the retrospective design and its inherent limitations, this study is of importance because real-life data are expected to increase the knowledge about NTB effects.
Indeed, in this observational study, the results report some meaningful effects with stabilization of FVC in UIP patients and a safety profile that sounds better than the one reported in SENSCIS trial. Furthermore, our data supported good safety, even in patients on anticoagulants.
In the future, increasing the number of patients enrolled and the data collected may better clarify the actual therapeutic efficacy of NTD.

Author Contributions

B.R., I.G., P.C., F.S. conceived and designed the study, analyzed and interpreted the data, drafted, revised, and approved the work; R.P., S.T., C.B. collected and interpreted the data, revised and approved the work; M.H., S.L. interpreted the data, revised and approved the work; M.K., R.C., E.B. collected the data, revised and approved the work; M.C. conceived and designed the study, revised and approved the work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study protocol was approved by the referral Ethics Committee (208-2022H) and it complied with the Declaration of Helsinki The study was approved by the Local Ethics Committee of CEUR (protocol 2018-34) and conducted according to the guidelines of the Declaration of Helsinki and ICH E6 Guideline for Good Clinical Practice.

Informed Consent Statement

Each patient signed a written informed consent before enrollment.

Data Availability Statement

Deidentified participant data will be made available upon motivated request to the Corresponding Author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Serra López-Matencio, J.M.; Gómez, M.; Vicente-Rabaneda, E.F.; González-Gay, M.A.; Ancochea, J.; Castañeda, S. Pharmacological Interactions of Nintedanib and Pirfenidone in Patients with Idiopathic Pulmonary Fibrosis in Times of COVID-19 Pandemic. Pharmaceuticals 2021, 14, 819. [Google Scholar] [CrossRef]
  2. Ruaro, B.; Cerinic, M.M.; Salton, F.; Baratella, E.; Confalonieri, M.; Hughes, M. Editorial: Pulmonary Fibrosis: One Manifestation, Various Diseases. Front. Pharmacol. 2022, 13, 1027332. [Google Scholar] [CrossRef]
  3. Cameli, P.; Alonzi, V.; d’Alessandro, M.; Bergantini, L.; Pordon, E.; Guerrieri, M.; Refini, R.M.; Sestini, P.; Bargagli, E. The Effectiveness of Nintedanib in Patients with Idiopathic Pulmonary Fibrosis, Familial Pulmonary Fibrosis and Progressive Fibrosing Interstitial Lung Diseases: A Real-World Study. Biomedicines 2022, 10, 1973. [Google Scholar] [CrossRef]
  4. Hostettler, K.E.; Zhong, J.; Papakonstantinou, E.; Karakiulakis, G.; Tamm, M.; Seidel, P.; Sun, Q.; Mandal, J.; Lardinois, D.; Lambers, C.; et al. Anti-Fibrotic Effects of Nintedanib in Lung Fibroblasts Derived from Patients with Idiopathic Pulmonary Fibrosis. Respir. Res. 2014, 15, 157. [Google Scholar] [CrossRef]
  5. Distler, O.; Highland, K.B.; Gahlemann, M.; Azuma, A.; Fischer, A.; Mayes, M.D.; Raghu, G.; Sauter, W.; Girard, M.; Alves, M.; et al. Nintedanib for Systemic Sclerosis-Associated Interstitial Lung Disease. N. Engl. J. Med. 2019, 380, 2518–2528. [Google Scholar] [CrossRef]
  6. Denton, C.P.; Goh, N.S.; Humphries, S.M.; Maher, T.M.; Spiera, R.; Devaraj, A.; Ho, L.; Stock, C.; Erhardt, E.; Alves, M.; et al. Extent of Fibrosis and Lung Function Decline in Patients with Systemic Sclerosis and Interstitial Lung Disease: Data from the SENSCIS Trial. Rheumatology 2022, keac535. [Google Scholar] [CrossRef]
  7. Allanore, Y.; Vonk, M.C.; Distler, O.; Azuma, A.; Mayes, M.D.; Gahlemann, M.; James, A.; Kohlbrenner, V.; Alves, M.; Khanna, D.; et al. Continued Treatment with Nintedanib in Patients with Systemic Sclerosis-Associated Interstitial Lung Disease: Data from SENSCIS-ON. Ann. Rheum. Dis. 2022, 81, 1722–1729. [Google Scholar] [CrossRef]
  8. Hilberg, F.; Roth, G.J.; Krssak, M.; Kautschitsch, S.; Sommergruber, W.; Tontsch-Grunt, U.; Garin-Chesa, P.; Bader, G.; Zoephel, A.; Quant, J.; et al. BIBF 1120: Triple Angiokinase Inhibitor with Sustained Receptor Blockade and Good Antitumor Efficacy. Cancer Res. 2008, 68, 4774–4782. [Google Scholar] [CrossRef] [Green Version]
  9. Confalonieri, P.; Volpe, M.C.; Jacob, J.; Maiocchi, S.; Salton, F.; Ruaro, B.; Confalonieri, M.; Braga, L. Regeneration or Repair? The Role of Alveolar Epithelial Cells in the Pathogenesis of Idiopathic Pulmonary Fibrosis (IPF). Cells 2022, 11, 2095. [Google Scholar] [CrossRef]
  10. Wells, A.U.; Flaherty, K.R.; Brown, K.K.; Inoue, Y.; Devaraj, A.; Richeldi, L.; Moua, T.; Crestani, B.; Wuyts, W.A.; Stowasser, S.; et al. Nintedanib in Patients with Progressive Fibrosing Interstitial Lung Diseases-Subgroup Analyses by Interstitial Lung Disease Diagnosis in the INBUILD Trial: A Randomised, Double-Blind, Placebo-Controlled, Parallel-Group Trial. Lancet Respir. Med. 2020, 8, 453–460. [Google Scholar] [CrossRef]
  11. Zhu, H.; Xia, M.-M.; Tong, K.-H.; Duan, W.-B. Nintedanib Induces the Autophagy-Dependent Death of Gastric Cancer Cells by Inhibiting the STAT3/Beclin1 Pathway. Dig Dis. Sci. 2022. [Google Scholar] [CrossRef]
  12. Shukla, S.K.; Nguyen, V.; Goyal, M.; Gupta, V. Cationically Modified Inhalable Nintedanib Niosomes: Enhancing Therapeutic Activity against Non-Small-Cell Lung Cancer. Nanomedicine 2022, 17, 935–958. [Google Scholar] [CrossRef]
  13. Baldini, C.; Danlos, F.-X.; Varga, A.; Texier, M.; Halse, H.; Mouraud, S.; Cassard, L.; Champiat, S.; Signolle, N.; Vuagnat, P.; et al. Safety, Recommended Dose, Efficacy and Immune Correlates for Nintedanib in Combination with Pembrolizumab in Patients with Advanced Cancers. J. Exp. Clin. Cancer Res. 2022, 41, 217. [Google Scholar] [CrossRef]
  14. Richeldi, L.; Costabel, U.; Selman, M.; Kim, D.S.; Hansell, D.M.; Nicholson, A.G.; Brown, K.K.; Flaherty, K.R.; Noble, P.W.; Raghu, G.; et al. Efficacy of a Tyrosine Kinase Inhibitor in Idiopathic Pulmonary Fibrosis. N. Engl. J. Med. 2011, 365, 1079–1087. [Google Scholar] [CrossRef] [Green Version]
  15. Richeldi, L.; du Bois, R.M.; Raghu, G.; Azuma, A.; Brown, K.K.; Costabel, U.; Cottin, V.; Flaherty, K.R.; Hansell, D.M.; Inoue, Y.; et al. Efficacy and Safety of Nintedanib in Idiopathic Pulmonary Fibrosis. N. Engl. J. Med. 2014, 370, 2071–2082. [Google Scholar] [CrossRef] [Green Version]
  16. OFEV (Nintedanib) Summary of Product Characteristics. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/Ofev (accessed on 1 December 2022).
  17. Lamb, Y.N. Nintedanib: A Review in Fibrotic Interstitial Lung Diseases. Drugs 2021, 81, 575–586. [Google Scholar] [CrossRef]
  18. Chen, C.-H.; Lin, H.-C.; Wang, Y.-H.; Wang, C.-Y.; Lin, Y.S.; Lai, C.-C. The Safety of Nintedanib for the Treatment of Interstitial Lung Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. PLoS ONE 2021, 16, e0251636. [Google Scholar] [CrossRef]
  19. Crestani, B.; Huggins, J.T.; Kaye, M.; Costabel, U.; Glaspole, I.; Ogura, T.; Song, J.W.; Stansen, W.; Quaresma, M.; Stowasser, S.; et al. Long-Term Safety and Tolerability of Nintedanib in Patients with Idiopathic Pulmonary Fibrosis: Results from the Open-Label Extension Study, INPULSIS-ON. Lancet Respir. Med. 2019, 7, 60–68. [Google Scholar] [CrossRef]
  20. Noth, I.; Oelberg, D.; Kaul, M.; Conoscenti, C.S.; Raghu, G. Safety and Tolerability of Nintedanib in Patients with Idiopathic Pulmonary Fibrosis in the USA. Eur. Respir. J. 2018, 52, 1702106. [Google Scholar] [CrossRef] [Green Version]
  21. Antoniou, K.; Markopoulou, K.; Tzouvelekis, A.; Trachalaki, A.; Vasarmidi, E.; Organtzis, J.; Tzilas, V.; Bouros, E.; Kounti, G.; Rampiadou, C.; et al. Efficacy and Safety of Nintedanib in a Greek Multicentre Idiopathic Pulmonary Fibrosis Registry: A Retrospective, Observational, Cohort Study. ERJ Open Res. 2020, 6, 00172–02019. [Google Scholar] [CrossRef] [Green Version]
  22. Fournier, D.; Jouneau, S.; Bouzillé, G.; Polard, E.; Osmont, M.-N.; Scailteux, L.-M. Real-World Safety Profiles of Pirfenidone and Nintedanib in Idiopathic Pulmonary Fibrosis Patients. Pulm. Pharmacol. Ther. 2022, 76, 102149. [Google Scholar] [CrossRef]
  23. Pitre, T.; Mah, J.; Helmeczi, W.; Khalid, M.F.; Cui, S.; Zhang, M.; Husnudinov, R.; Su, J.; Banfield, L.; Guy, B.; et al. Medical Treatments for Idiopathic Pulmonary Fibrosis: A Systematic Review and Network Meta-Analysis. Thorax 2022, 77, 1243–1250. [Google Scholar] [CrossRef]
  24. Caro, F.; Buendía-Roldán, I.; Noriega-Aguirre, L.; Alberti, M.L.; Amaral, A.; Arbo, G.; Auteri, S.; Bermúdez, A.; Curbelo, P.; de Jesús Díaz Verduzco, M.; et al. Latin American Registry of Idiopathic Pulmonary Fibrosis (REFIPI): Clinical Characteristics, Evolution and Treatment. Arch. Bronconeumol. 2022, 58, 794–801. [Google Scholar] [CrossRef]
  25. Mondoni, M.; Alfano, F.; Varone, F.; Muscato, G.; Conti, C.; Saderi, L.; Chiesa, A.; Di Marco, F.; Vancheri, C.; Richeldi, L.; et al. Observational, Multicenter Study on the Efficacy, Tolerability, and Safety of Nintedanib in Patients with Idiopathic Pulmonary Fibrosis Older than 80 Years. Respir. Int. Rev. Thorac. Dis. 2022, 1, 9. [Google Scholar] [CrossRef]
  26. Kaunisto, J.; Salomaa, E.R.; Hodgson, U.; Kaarteenaho, R.; Kankaanranta, H.; Koli, K.; Vahlberg, T.; Myllärniemi, M. Demographics and survival of patients with idiopathic pulmonary fibrosis in the FinnishIPF registry. ERJ Open Res. 2019, 5, 00170–02018. [Google Scholar] [CrossRef]
  27. King, T.E., Jr.; Albera, C.; Bradford, W.Z.; Costabel, U.; du Bois, R.M.; Leff, J.A.; Nathan, S.D.; Sahn, S.A.; Valeyre, D.; Noble, P.W. All-cause mortality rate in patients with idiopathic pulmonary fibrosis. Implications for the design and execution of clinical trials. Am. J. Respir. Crit. Care Med. 2014, 189, 825–831. [Google Scholar] [CrossRef] [Green Version]
  28. Marshall, D.C.; Salciccioli, J.D.; Shea, B.S.; Akuthota, P. Trends in mortality from idiopathic pulmonary fibrosis in the European Union: An observational study of the WHO mortality database from 2001–2013. Eur. Respir. J. 2018, 51, 1701603. [Google Scholar] [CrossRef] [Green Version]
  29. Available online: https://www.istat.it/it/files/2017/12/cap01.pdf (accessed on 1 December 2022).
  30. Sulli, A.; Ruaro, B.; Cutolo, M. Evaluation of blood perfusion by laser speckle contrast analysis in different areas of hands and face in patients with systemic sclerosis. Ann Rheum Dis. 2014, 73, 2059–2061. [Google Scholar] [CrossRef]
  31. Bernero, E.; Sulli, A.; Ferrari, G.; Ravera, F.; Pizzorni, C.; Ruaro, B.; Zampogna, G.; Alessandri, E.; Cutolo, M. Prospective capillaroscopy-based study on transition from primary to secondary Raynaud’s phenomenon: Preliminary results. Reumatismo 2013, 65, 186–191. [Google Scholar] [CrossRef] [Green Version]
  32. Ruaro, B.; Confalonieri, M.; Matucci-Cerinic, M.; Salton, F.; Confalonieri, P.; Santagiuliana, M.; Citton, G.M.; Baratella, E.; Bruni, C. The Treatment of Lung Involvement in Systemic Sclerosis. Pharmaceuticals 2021, 14, 154. [Google Scholar] [CrossRef]
Figure 1. The 6-month trend for spirometry measurements. Box-plots representing levels of FVC, FEV1 and DLCO at baseline and after 6 months.
Figure 1. The 6-month trend for spirometry measurements. Box-plots representing levels of FVC, FEV1 and DLCO at baseline and after 6 months.
Pharmaceuticals 16 00307 g001
Figure 2. The 12-month trend for spirometry measurements. Box-plots representing levels of FVC, FEV1 and DLCO at baseline and after 12 months.
Figure 2. The 12-month trend for spirometry measurements. Box-plots representing levels of FVC, FEV1 and DLCO at baseline and after 12 months.
Pharmaceuticals 16 00307 g002
Figure 3. Trend for spirometry measurements between 0 and 12 months. Dots represent the median value, bars indicate the first (Q1) and the third (Q3) quartiles.
Figure 3. Trend for spirometry measurements between 0 and 12 months. Dots represent the median value, bars indicate the first (Q1) and the third (Q3) quartiles.
Pharmaceuticals 16 00307 g003
Figure 4. Overall survival. On the top, Kaplan–Meier curve for overall survival. On the bottom, number of subject at risk and number of events for each time point.
Figure 4. Overall survival. On the top, Kaplan–Meier curve for overall survival. On the bottom, number of subject at risk and number of events for each time point.
Pharmaceuticals 16 00307 g004
Table 1. Comparison of spirometric measurements after 6 months and after 12 months.
Table 1. Comparison of spirometric measurements after 6 months and after 12 months.
Baseline
[Median (IQR)]
6 Months
[Median (IQR)]
np-ValueBaseline
[Median (IQR)]
12 Months
[Median (IQR)]
np-Value
FVC
76.0 (22.0)76.0 (22.0)310.676.0 (14.2)76.0 (23.2)220.2
FEV1
77.0 (23.0)81.0 (24.0)29>0.982.5 (21.0)74.0 (16.5)200.039
DLCO
45.0 (28.0)37.0 (20.0)250.01247.0 (30.8)43.0 (30.2)200.018
Table 2. Steroid use in responders and non-responders for FVC and DLCO.
Table 2. Steroid use in responders and non-responders for FVC and DLCO.
FVC
6 months12 months
Responder (n = 25)Non-responder (n = 6)p-valueResponder (n = 19)Non-responder (n = 3)p-value
10 (40%)2 (33%)>0.97 (39%)2 (67%)0.6
DLCO
6 months12 months
Responder (n = 12)Non-responder (n = 13)p-valueResponder (n = 15)Non-responder (n = 5)p-value
4 (33%)6 (46%)0.74 (29%)4 (80%)0.1
Table 3. Clinical and demographic characteristics of our cohort of IPF patients.
Table 3. Clinical and demographic characteristics of our cohort of IPF patients.
Clinical and demographic characteristics56 patients
Females, n (%)19 (34%)
Mean onset age (years), mean ± SD71 ± 11 years
Mean age at baseline (years), mean ± SD74 ± 9 years
Smokers (active or former) 34 (62%)
Autoantibodies
Antinuclear antibodies (ANA)7 (12%)
Anti-Ro52, n (%)3 (5%)
Organ involvement
COPD (n, %)3 (5%)
Cancer (n, %)8 (15%)
Hypertension (n, %)36 (64%)
Coronary heart disease (n, %)11 (19%)
Diabetes mellitus (n, %) 22 (39%)
Rheumatic diseases (n, %)1 (2%)
Previous episodes of venous thromboembolism (n, %)11 (19%)
Cardiac comorbidities (n, %)31 (55%)
Concurrent Therapies
Mycophenolate mofetil, n (%)3 (5%)
Azathioprine, n (%)1 (2%)
Hydroxychloroquine, n (%)2 (4%)
Rituximab, n (%)1 (2%)
Steroid high dose (i.e., >15 mg/die), n (%)12 (21%)
Steroid low dose (i.e., <15 mg/die), n (%)8 (15%)
Antihypertensive drugs, n (%)36 (64%)
Pump inhibitor drugs, n (%)38 (59%)
Anticoagulant drugs, n (%)14 (25%)
Warfarin, n (%)11 (20%)
Apixaban and Rivaroxaban3 (5%)
Baseline HRCT 56 patients
 NSIP, n (%)11 (20%)
 UIP, n (%)45 (80%)
Legend. N = number; SD = standard deviation; Ab = antibodies; GI = gastro-intestinal; NSIP = non-specific interstitial pneumonia; UIP = usual interstitial pneumonia.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ruaro, B.; Gandin, I.; Pozzan, R.; Tavano, S.; Bozzi, C.; Hughes, M.; Kodric, M.; Cifaldi, R.; Lerda, S.; Confalonieri, M.; et al. Nintedanib in Idiopathic Pulmonary Fibrosis: Tolerability and Safety in a Real Life Experience in a Single Centre in Patients also Treated with Oral Anticoagulant Therapy. Pharmaceuticals 2023, 16, 307. https://doi.org/10.3390/ph16020307

AMA Style

Ruaro B, Gandin I, Pozzan R, Tavano S, Bozzi C, Hughes M, Kodric M, Cifaldi R, Lerda S, Confalonieri M, et al. Nintedanib in Idiopathic Pulmonary Fibrosis: Tolerability and Safety in a Real Life Experience in a Single Centre in Patients also Treated with Oral Anticoagulant Therapy. Pharmaceuticals. 2023; 16(2):307. https://doi.org/10.3390/ph16020307

Chicago/Turabian Style

Ruaro, Barbara, Ilaria Gandin, Riccardo Pozzan, Stefano Tavano, Chiara Bozzi, Michael Hughes, Metka Kodric, Rossella Cifaldi, Selene Lerda, Marco Confalonieri, and et al. 2023. "Nintedanib in Idiopathic Pulmonary Fibrosis: Tolerability and Safety in a Real Life Experience in a Single Centre in Patients also Treated with Oral Anticoagulant Therapy" Pharmaceuticals 16, no. 2: 307. https://doi.org/10.3390/ph16020307

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop