Skip to main content
Chinese Medical Journal logoLink to Chinese Medical Journal
. 2024 Oct 11;138(3):264–277. doi: 10.1097/CM9.0000000000003149

Cancer therapy-related interstitial lung disease

Chengzhi Zhou 1, Haiyi Deng 2, Yilin Yang 1, Fei Wang 1,3, Xinqing Lin 1, Ming Liu 1, Xiaohong Xie 1, Tao Luan 1,4, Nanshan Zhong 1,
Editor: Peifang Wei
PMCID: PMC11771665  PMID: 39402974

Abstract

With the increasing utilization of cancer therapy, the incidence of lung injury associated with these treatments continues to rise. The recognition of pulmonary toxicity related to cancer therapy has become increasingly critical, for which interstitial lung disease (ILD) is a common cause of mortality. Cancer therapy-related ILD (CT-ILD) can result from a variety of treatments including chemotherapy, targeted therapy, immune checkpoint inhibitors, antibody–drug conjugates, and radiotherapy. CT-ILD may progress rapidly and even be life-threatening; therefore, prompt diagnosis and timely treatment are crucial for effective management. This review aims to provide valuable information on the risk factors associated with CT-ILD; elucidate its underlying mechanisms; discuss its clinical features, imaging, and histological manifestations; and emphasize the clinical-related views of its diagnosis. In addition, this review provides an overview of grading, typing, and staging treatment strategies used for the management of CT-ILD.

Keywords: Cancer therapy, Interstitial lung disease, Lung toxicity

Introduction

The lungs have two distinct blood supply systems, with a large contact surface, making them vulnerable to attacks by different toxic substances. More than 400 drugs have been reported to cause lung injury, with damage to the airways, lung parenchyma, pulmonary vasculature, and pleura. Drug-induced interstitial lung disease (DILD) is the most common form of lung damage. DILD is a very heterogeneous adverse event, ranging from asymptomatic interstitial lung disease (ILD) to respiratory failure and even life-threatening conditions. Among the cases of DILD, cancer drug-related ILD is the most prevalent, accounting for 23–51% of all cases.[1] An analysis of the Japanese Adverse Drug Event Report (JADER) database found methotrexate, gemcitabine, docetaxel, gefitinib, erlotinib, everolimus, and nivolumab to be the top six drugs with the highest ILD reporting rates.[2] Tamura et al[3] found that the most common drugs that cause lung toxicity were anticancer drugs (42%), followed by antirheumatic drugs (24%). In addition, radiotherapy (RT) can cause lung toxicity, known as radiation pneumonitis (RP). In clinical practice, RT is often simultaneous or sequential with anticancer drugs, particularly in combination immunotherapy (IO), which makes it difficult to clearly distinguish DILD from RP. Additionally, radiation recall pneumonitis (RRP) is a pulmonary disease that arises as a delayed consequence of RT and is often triggered by systemic agents, such as anticancer drugs.[4] Therefore, ILD caused by cancer therapy is collectively called cancer therapy-related ILD (CT-ILD).

CT-ILD is the most common severe and lethal adverse event associated with tumors. A retrospective study analyzed 770 patients with non-small cell lung cancer (NSCLC), among whom 44 patients (6%) developed pneumonitis during anticancer treatment, resulting in a mortality rate of 36%.[5] A study of the French REISAMIC registry reported a considerably higher frequency of very serious immune-related adverse events (irAEs) in the respiratory system, with ILD being the most common fatal irAEs.[6] Furthermore, another study indicated that pneumonia was predominant in fatal adverse events related to programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors. A comprehensive analysis of 53 cohorts also revealed that ILD was the most frequent fatal toxicities associated with the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI).[7]

CT-ILD has a rapid and potentially life-threatening progression, underscoring the importance of accurate and rapid diagnosis, followed by early treatment. However, the clinical, imaging, and histological features of CT-ILD lack specificity. In addition, a patient may be receiving multiple treatments that can trigger ILD simultaneously. CT-ILD is an exclusive diagnosis that is easily confused with a lung infection, tumor progression, and pulmonary edema. This review aims to provide valuable information on the risk factors associated with CT-ILD, elucidate its underlying mechanisms, discuss its clinical features, imaging, and histological manifestations, and emphasize the clinical-related views of its diagnosis and management methods.

Epidemiology

The precise incidence of CT-ILD is difficult to estimate, as it is influenced by multiple factors, including the specific drug, the RT regimen, the dose, and the combination regimen. A study examining 19,132 cancer patients who received anticancer drugs between 2014 and 2018, reported that 120 (0.62%) patients developed severe DILD.[8] Terbuch et al[9] analyzed 2499 patients with advanced cancer participating in phase I clinical trials and found that 60 patients (2.40%) developed DILD. Furthermore, based on a summary of 470 oncology phase I trials, the incidence rates of pulmonary toxicity, total DILD, and grade 3–4 DILD were 2.70%, 0.77%, and 0.31%, respectively.[10]

CT-ILD can be broadly classified into two types: anticancer drug-related lung injury and RT-related lung injury. Various anticancer drugs may cause different degrees of ILD, including chemotherapy (e.g., bleomycin, cyclophosphamide, methotrexate, gemcitabine, and taxanes), small-molecular targeted drugs (e.g., gefitinib, erlotinib, osimertinib, crizotinib, and pralsetinib), mammalian target of rapamycin (mTOR) inhibitors (e.g., sirolimus and everolimus), immune checkpoint inhibitors (ICIs) (e.g., nivolumab, pembrolizumab, durvalumab), and antibody-drug conjugates (ADCs) (e.g., trastuzumab emtansine). Conventional chemotherapy typically carries a low incidence of ILD, while RT is associated with a higher incidence of RP, mainly grades 1–2 in severity. Newer antitumor drugs, especially ADC drugs and ICIs, appear to be more lethal and have a higher incidence of ILD, while TKI-ILD has a lower incidence [Supplementary Figure 1, http://links.lww.com/CM9/C15].

TKIs

The incidence of all-grade and high-grade ILD related to EGFR-TKI in a meta-analysis has been reported to be 1.6% (95% confidence interval [CI], 1.0–2.4%) and 0.9% (95% CI, 0.6–1.4%), respectively, with a mortality rate of 13.0% (95% CI, 7.6–21.6%).[11] Another meta-analysis showed that the overall incidence of pneumonitis associated with EGFR-TKI was 1.12% for all grades, 0.61% for high grade, and 0.20% for grade 5.[12] Furthermore, a meta-analysis by Suh et al[13] revealed that the overall pooled incidence of anaplastic lymphoma kinase (ALK) TKIs-associated pneumonitis for all grades, high grade, and grade 5 ILD was 2.14%, 1.33%, and 0.22%, respectively.

mTOR inhibitors

An analysis of phases II–III randomized controlled trials involving mTOR inhibitors reported an incidence of all-grade pulmonary toxicity at 10.4%, with high-grade incidence of 2.4%.[14] Another study revealed that the overall incidence of fatal adverse events associated with mTOR inhibitors was 1.8%.[15]

ICIs

A meta-analysis of 19 studies showed that the incidence rate of checkpoint inhibitor-related pneumonitis (CIP) at any grade or high grade is higher in the PD-1 inhibitor group than in the PD-L1 inhibitor group (3.6% vs. 1.3%, P = 0.001; 1.1% vs. 0.4%, P = 0.02; respectively).[16] Another meta-analysis revealed that the incidence of CIP in patients receiving PD-1 inhibitor monotherapy was 2.7% for all grades and 0.8% for grade ≥3 CIP.[17] Furthermore, a recent study revealed that the distribution of very severe respiratory toxicity (grades 4–5) associated with ICI was 1.1%.[6]

ADCs

A meta-analysis of drug-associated pneumonitis with ADCs found that the total incidence of all-grade and grade ≥3 pneumonitis was 5.86% and 0.68%, respectively. Specifically, the incidence of all-grade pneumonitis was 13.58%, and for grade ≥3 was 2.19% with trastuzumab deruxtecan (T-DXd), the highest rate observed in ADC therapy.[18] In one meta-analysis focusing specifically on T-DXd monotherapy-associated pneumonitis, a total ILD incidence of 15.4% and a grade 5 incidence of 2.2% were reported.[19]

Pathogenesis

The pathogenesis of CT-ILD has not yet been fully understood. Possible mechanisms may involve cytotoxicity and immune-mediated lung injury. Cancer therapy can directly or after biological transformation damage the alveolar epithelial, airway epithelial cells, or capillary endothelial cells, produce reactive oxygen species (ROS) or induce phospholipid deposition and activate immune cells as a hapten or mimic an antigen, thus causing a cascade of immune reactions [Figure 1].[20,21]

Figure 1.

Figure 1

Pathobiology of cancer therapy-related interstitial lung disease. Cancer therapy can directly or after biological transformation damage the alveolar epithelial, airway epithelial cells, or capillary endothelial cells, produce ROS, increase endothelial permeability, or induce phospholipid deposition. The drugs may act as antigens or haptens to activate immune cells or enhance the activity of T cells against cross-antigens expressed in tumors and normal tissues, causing the recruitment of inflammatory cells and the release of inflammatory factors. ROS: Reactive oxygen species; RT: Radiotherapy.

Chemotherapeutic drugs can produce ROS and nitric oxide (NO), triggering oxidative stress mechanisms that result in DNA damage and cell death.[22,23] They can also promote the release of inflammatory cytokines and chemokines such as tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), monocyte chemoattractant protein-1 (MCP-1), and transforming growth factor-beta (TGF-β).[23,24] These cytokines induce inflammation in lung tissue and may lead to capillary leakage and pulmonary edema (as observed with gemcitabine, for example).[25,26,27]

EGFR is involved in the repair of lung injury. EGFR-TKIs block EGFR phosphorylation, thus preventing the regeneration and proliferation of damaged epithelial cells, which can lead to the appearance of lung injury.[28,29] EGFR-TKIs exacerbate the progression of lung injury in TNF-overexpressing lung tissues by suppressing TNF-induced EGFR transactivation.[28] TKI-ILD is related to increased expression of interferon-induced protein 10 (IP-10) and heat shock protein 70 (HSP 70).[30,31]

ICI-induced pneumonia may be associated with the following five aspects: (1) Imbalance of T cell subsets. Treatment with ICI can activate immune function, leading to an imbalance of T cell subsets. The alveolar lavage fluid from patients with CIP showed an increase in CD8+ T and CD4+ T cells and a decrease in cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and PD-L1 positive Treg cells.[32] Another analysis of biopsy pathology in patients with CIP found elevated CD8+ T cells and activated memory CD4+ T cells, and decreased resting memory CD4+ T cells.[33] (2) Activation and increase of pre-existing autoantibodies.[34,35] Anti-CD74 autoantibody levels increase significantly in CIP patients during pneumonia compared to those before ICI treatment.[36] (3) The production and release of cytokines/chemokines. Studies have found an association between the appearance of CIP and various inflammatory cytokines, and chemokines, including C-reactive protein (CRP), interleukin-6 (IL-6), IL-10, IL-17A, IL-35, interferon-gamma (IFN-γ), and C-X-C motif chemokine ligand (CXCL)9/CXCL10/CXCL11.[37,38,39] (4) Cross-antigen reactivity: Tumor cells possess antigens on their surface that are identical to those found in normal lung tissue. Overactivated T cells mistakenly recognize antigens on the surface of normal tissue cells and initiate an attack.[40,41,42] Furthermore, the dissolution of tumor cells and disintegration of tumor tissues during anticancer therapy can lead to the diffusion of antigenic epitopes.[43] (5) Microbial modulation of host immunity may be related to the toxicity of IO.[44]

ADC-ILD can potentially be linked to multiple mechanisms, including (1) target-dependent uptake of the ADC, (2) target-independent uptake of the ADC in healthy cells, (3) bystander killing caused by the release of free payload from cancer cells, and (4) the presence of circulating free payload due to deconjugation of the ADC.[45]

Ionizing radiation induces free radicals and DNA damage, promoting oxidative stress, vascular injury, and inflammation. Persistent inflammation sustains injury to alveolar epithelial and endothelial cells, recruiting a large number of immune cells and releasing cytokines. Alveolar epithelial injury leads to reduced release of pulmonary surfactant, resulting in decreased alveolar tension. Endothelial cell damage causes changes in vascular permeability, leading to pulmonary edema. Prolonged damage to the alveoli and vascular systems results in excessive proliferation of fibroblasts and eventually leads to fibrotic changes.[46,47]

Risk Factors

The development of CT-ILD is largely unpredictable and idiosyncratic, although it may be associated with the cumulative dose and renal insufficiency. Risk factors for different types of CT-ILD may vary [Supplementary Table 1, http://links.lww.com/CM9/C15].

Age

Older patients have shown a higher susceptibility to developing CT-ILD, with a higher incidence of severe cases. A retrospective study found that patients with fatal bleomycin pulmonary toxicity were older than other patients in the study sample. For patients aged over 40 years old, especially those with renal insufficiency, the risk of fatal pulmonary toxicity was reported to exceed 10%.[48] Similarly, studies have shown that advanced age was a risk factor for the ILD associated with gemcitabine,[49] nab-paclitaxel,[50] gefitinib,[51] ALK-TKI,[52] and RT.[53] However, a study evaluating T-DXd, found the opposite result: age <65 years was associated with the occurrence of CT-ILD.[19]

Ethnicity and gene

According to a meta-analysis, the frequency of ILD among EGFR-TKI-treated patients was higher in Asian patients compared to non-Asian patients, although this difference did not reach statistical significance (2.5% vs. 0.9%, odds ratio [OR] = 2.79, 95% CI = 0.82–9.40, P = 0.11). Among Asian patients, the incidence of ILD was found to be higher in Japanese patients compared to that of other Asian countries (3.8% vs. 0.3%; OR = 12.7, 95% CI, 1.69–95.1; P = 0.0009).[54] Additionally, the incidence of all- and severe-grade EGFR-TKI-related ILDs in Japanese studies was found to be higher than in non-Japanese studies.[12] Xie et al[7] also identified that the frequency of fatal toxicity related to EGFR-TKIs in Japanese patients was higher, compared to non-Eastern Asian patients. Similarly, the incidence of ALK-TKI-related pneumonitis was significantly higher in studies from Japan compared to non-Japanese studies, both for all grade (6.25% vs. 1.14%, P <0.001) and high-grade pneumonitis (3.31% vs. 0.39%, P <0.001).[13] These findings were similarly observed in the study of T-DXd.[19]

A study investigating the association between DILD and human leukocyte antigen (HLA) alleles showed that the higher frequency of HLA-DRB1*04:05 was in the Japanese population, compared to other populations, which may partly explain the high incidence rate of CT-ILD in the Japanese population.[54] Schwaiblmair et al[55] found that in patients with advanced pancreatic cancer treated with gemcitabine and erlotinib, the combination of HLA-B*15:01 and DRB1*15:01 was associated with ILD. Previous studies have shown that gene polymorphism may also be related to DILD.[56,57,58]

Previous lung diseases

Several studies have shown that previous lung disease was a risk factor for the development of lung toxicity in patients treated with gemcitabine.[59,60] Pre-existing ILD is related to the increased incidence of docetaxel and paclitaxel-related ILD.[50,61,62] Additionally, Sakurada et al[63] found pre-existing ILD was a risk factor for chemotherapy-induced ILD. Smoking, decreased lung function, and pre-existing chronic ILD have also been identified as being associated with the occurrence of ILD during gefitinib treatment or chemotherapy.[51] Hotta et al[64] found that poor performance status (PS) scores and previous pulmonary fibrosis increased the risk of ILD associated with gefitinib and erlotinib. Poor PS score, smoking history, previous or concomitant ILD, and pleural effusion also increased the risk of crizotinib-related ILD. A recent analysis of nine phase I and II studies of T-DXd found that low oxygen saturation levels and coexisting lung comorbidities were associated with increased CT-ILD.[19] Preexisting ILD, chronic obstructive pulmonary disease (COPD), emphysema, asthma, and obstructive pneumonia are related to the appearance of CIP. Both COPD and ILD have been confirmed to be risk factors for the development of RP.[65,66]

Histological type

A meta-analysis summarizing studies related to docetaxel plus gemcitabine found that patients with lung cancer had a higher incidence of serious CT-ILD than patients with breast cancer.[67] Similarly, a retrospective analysis of patients receiving gemcitabine-based chemotherapy in the Japanese nationwide administrative database showed that lung cancer was associated with a high incidence of CT-ILD.[68] Another meta-analysis by Ma et al[69] showed that patients with NSCLC have a higher incidence of ILD related to ICI compared to other cancers. Furthermore, a retrospective study reported that the frequency of ICI-related ILD was higher in squamous cell carcinoma than in adenocarcinoma.[70] A recent study on T-DXd-induced ILD revealed that uterine carcinomatosis (26.47%) and NSCLC (24.77%) were the most common types.[71]

Combination therapy

Drug combinations can have varying effects on lung toxicity. The combination of the same type of drugs may cause similar pulmonary patterns of toxicity, while the combination of different types of drugs may worsen or alleviate it. For example, gemcitabine combined with docetaxel has been shown to increase the incidence rate of CT-ILD.[67] The proportion of ILD induced by ICIs combined with EGFR-TKI is significantly higher than that of either drug alone.[72] Combining PD-1/PD-L1 inhibitors with CTLA-4 inhibitors has been associated with the risk of CT-ILD.[73] However, a meta-analysis showed that compared to single or dual ICI treatments, ICIs plus chemotherapy reduced both all grades and high grades of CT-ILD.[74] A recent meta-analysis of randomized clinical trials showed that TKI combined with bevacizumab reduced the incidence of CT-ILD, while the combination of chemotherapy and bevacizumab did not.[75]

RT subsequent to or in combination with chemotherapy, especially with paclitaxel, can increase the risk of lung toxicity.[76] A retrospective study showed that the incidence rate of RP was 81.7% in patients receiving concurrent chemoradiotherapy, and V20 greater than V30 was identified as a risk factor.[77] Sequential chemoradiotherapy appears to have a higher risk of pulmonary toxicity compared to concurrent chemoradiotherapy.[53] Several studies have confirmed that RT combined with ICIs increased severe lung toxicity.[78] In addition, there is an increased risk of pneumonitis when TKI is combined with RT compared to TKI or RT alone.[79,80] However, other studies have reported no significant increase in pneumonitis incidence in the combination groups.[81,82] It has been suggested that the occurrence of pneumonitis may be influenced by factors such as the timing and sequence of TKI and RT overlap, as well as the specific type of TKI administered.[83,84,85]

Clinical Features

The onset time of CT-ILD can vary greatly, ranging from hours to years after initiating treatment. The clinical presentation of CT-ILD is often non-specific and CT-ILD may have an insidious onset without any clinical symptoms or may develop acutely with symptoms such as dyspnea, cough, fever, fatigue, chest pain, or hemoptysis.[86] Some cases can progress slowly or rapidly to respiratory failure and can even be life-threatening. Lung auscultation may reveal normal sounds or wet rales or “velcro-like” crackles.[25] Later stages of the disease can be accompanied by signs of right ventricular dysfunction and pulmonary hypertension, such as lower limb edema and jugular vein dilation.[87] There are no distinctive clinical characteristics specifically attributed to the causation of the drug.

Radiographic Features

CT-ILD imaging also shows no specificity, which is usually found in tumor patients during routine imaging examinations. Cleverley et al[88] revealed that only 45% of patients with DILD have consistent imaging and histology. However, imaging examination still plays a central role in the diagnosis of CT-ILD. Even in the absence of symptoms, the presence of CT-ILD patients can be detected by imaging examinations, which can also be used to assess the severity of the condition. High-resolution computed tomography (HRCT) is more accurate than conventional chest radiographs in evaluating abnormalities of the lung parenchyma and airway, pattern characteristics, and distribution of lesions, and is more sensitive in the early diagnosis of CT-ILD. For example, one study showed that 23 cases of DILD were abnormal in HRCT, but only 17 cases in chest radiographs.[89] Another study showed that HRCT showed good sensitivity and specificity in diagnosing DILD, especially in organizing pneumonia (OP) and diffuse alveolar damage (DAD).[90] Therefore, in cases of suspected CT-ILD, if the patient’s condition allows, it is recommended to undergo HRCT evaluation. HRCT is also essential in the differential diagnosis and follow-up after treatment.

There are many patterns of CT-ILD imaging, and different patterns can be caused by a single drug, or present in the same patient. Some common patterns include non-specific interstitial pneumonia (NSIP), OP, DAD, hypersensitivity pneumonitis (HP), and simple pulmonary eosinophilia (PEo) [Table 1].

Table 1.

Radiologic patterns of cancer therapy-related interstitial lung disease at HRCT.

Patterns Imaging manifestations at HRCT
NSIP GGO or patchy consolidation, often with reticular opacities, with or without tractive bronchiectasis; bilateral and symmetric, mainly involving the lower lungs and subpleural areas.
OP Multifocal patchy consolidation shadow, showing peribronchovascular and/or peripheral distribution, may present with “reversed halo sign”.
DAD The exudation phase showed extensive bilateral GGO and gas cavity consolidation, tractive bronchiectasis, and decreased lung volumes in the fibrotic phase.
HP Bilateral diffuse GGO and/or poorly defined lobular central nodules, and mosaic attenuation.
PEo Unilateral or bilateral, non-segmental consolidation or GGO, transient, and migratory.

DAD: Diffuse alveolar damage; GGO: Ground-glass opacity; HP: Hypersensitivity pneumonitis; HRCT: High resolution computed tomography; NSIP: Non-specific interstitial pneumonia; OP: Organizing pneumonia; PEo: Pulmonary eosinophilia.

Imaging findings of ILD induced by different treatments share similarities, but also present variations. The imaging characteristics of gefitinib-induced ILD can be classified into four patterns: (1) non-specific areas of ground glass attenuation, corresponding to NSIP; (2) multifocal areas of airspace consolidation, corresponding to OP; (3) patchy distribution of ground glass attenuation with thickened interlobular septa, corresponding to PEo; and (4) extensive bilateral ground-glass attenuation or airspace consolidation accompanied by traction bronchiectasis, corresponding to DAD. Among these, pattern 1 is the most frequently observed pattern. Pattern 4 exhibits a higher mortality rate compared to the other patterns.[28,91,92] A newly discovered drug-related lung disease called transient asymptomatic pulmonary opacity was observed in patients receiving EGFR-TKI treatment. This condition resolves spontaneously without the need for specific treatment while patients continue their TKI therapy.

In a retrospective study of 66 patients treated with the mTOR inhibitor everolimus, 14 patients (21%) developed drug-related pneumonitis (OP pattern in eight, NSIP pattern in five, and HP pattern in one).[93] In another study, the classification of mTOR inhibitor-associated pneumonia revealed 16 cases (70%) of OP and seven cases (30%) of NSIP.[94]

Naidoo et al[95] proposed five imaging classifications for CIP: OP, ground-glass opacities (GGOs), interstitial pneumonitis, hypersensitivity pneumonitis, and pneumonitis not otherwise specified. GGO is the most common type and is associated with a poor prognosis.[95,96] Among the 20 patients with CIP, the OP pattern was the most common (n = 13), followed by the NSIP pattern (n = 3), the HP pattern (n = 2), and the DAD pattern (n = 2).[97] The DAD pattern had the highest severity grade, followed by OP, while NSIP and HP had lower severity grades.[97] Although different types of CT-ILD present variations, one common aspect among them is that the DAD pattern has a poor prognosis. The most common types of CT-ILD among different subtypes are OP or NSIP.

Histopathology

The pathologic features of CT-ILD are also non-specific, and diagnosis requires the integration of clinical, laboratory, and imaging information. Cancer therapy may impact multiple compartments and, as a result, can cause overlapping histological patterns of lung toxicity. Several common histopathological patterns were observed including HP, OP, DAD, NSIP, eosinophilic pneumonia, pulmonary hemorrhage, and granulomatous pneumonitis.[87,98]

HP is characterized by cellular bronchiolitis and cellulate-rich inflammation in the surrounding lung tissue, along with scattered non-caseous granulomas.[87,99] OP is identified by the proliferation of fibroblasts in the alveolar cavity producing immature collagen, as well as infiltration of lymphocytes, plasma cells and histiocyte.[87,100] DAD is characterized histologically by the presence of an alveolar cavity lined with hyaluronic membrane, type II alveolar epithelial cells, and fibroblast proliferation, but it is difficult to observe abundant hyaluronic membrane in the acute or exudative phase.[98,100] NSIP histologically shows evenly distributed fibroblastic foci, which may not be obvious or absent; with infiltrations of lymphocytes and plasma cells within the alveolar septum and peribronchial spaces.[87,100,101] Eosinophilic pneumonia is characterized by a large number of eosinophils that fill the alveolar cavity, together with fibrin and some erythrocytes.[99,100]

Diagnostic Criteria and Procedure

The diagnosis of CT-ILD is particularly complicated due to the diverse range of cancer therapies and the varying occurrence time, cumulative dose, and clinical manifestations of lung injury between different patients and treatments. CT-ILD is an exclusive diagnosis, and commonly considered differential diagnoses include lung tumor progression, pulmonary edema, infectious pneumonia, and pulmonary embolism. The diagnostic criteria have been proposed in a previous study[87] and include (1) a history of exposure to drugs that can cause lung injury; (2) clinical, imaging, and histopathological patterns that are consistent with earlier observations of the same drug; (3) exclusion of other possible causes; (4) symptoms improved after withdrawal of the causative drug; and (5) symptoms worsen on rechallenge. Recently, Johkoh et al[102] proposed new diagnostic criteria: (1) new parenchymal opacities found on computed tomography (CT) or chest radiography; (2) a temporal association between the presentation and the initiation of a therapeutic agent; and (3) exclusion of other causes of ILD. However, it can sometimes be challenging to completely exclude other causes or potential co-existing factors. Furthermore, since early treatment is crucial for improved outcomes in CT-ILD, a definitive diagnosis, excluding all alternative etiologies, may not always be mandatory prior to initiating steroid treatment, particularly in severe cases.[103] Therefore, patients can be categorized as either definite, probable, or possible according to determinants.

Based on the consensus of DILD published by Kubo et al,[86] we summarize the diagnostic procedure for CT-ILD in Figure 2. It is important that patients be thoroughly evaluated before receiving cancer therapy, which has been reported to induce severe ILD, especially for baseline chest imaging findings. Additionally, high-risk patients should be more closely screened and monitored. For new-onset cough, dyspnea, fever, chest tightness, or aggravation of existing respiratory symptoms, CT-ILD should be considered and chest CT (HRCT is recommended) should be performed as soon as possible. CT may facilitate early detection of CT-ILD when it is still in the reversible stage or may help identify other potential causes that could explain the symptoms of the patient.[104]

Figure 2.

Figure 2

Flow chart of diagnosing CT-ILD. BAL: Bronchoalveolar lavage; BNP: Brain natriuretic peptide; CMV: Cytomegalovirus; CRP: C-reactive protein; CT: Computed tomography; CT-ILD: Cancer therapy-related interstitial lung disease; ECG: Electrocardiogram; HRCT: High resolution computed tomography; KL-6: Krebs von den Lungen-6; LDH: Lactate dehydrogenase; NT-proBNP: N-terminal pro b-type natriuretic peptide; SpO2: Oxygen saturation; SP-D: Surfactant protein-D.

Bronchoscopy combined with bronchoalveolar lavage (BAL) has no definite value in the diagnosis of CT-ILD, but it can help rule out an infection, alveolar hemorrhage, or tumor progression.[105,106] Solazzo et al[107] found that among 26 ILD cases, 12 were from infection and 14 were from DILD. The total number and classification count of BAL cells in different CT-ILD patterns varied but without specificity.[108] However, the American Thoracic Society states that when combined with clinical information and chest imaging, BAL cell patterns and other characteristics provide useful information for the diagnosis of patients with suspected DILD,[109] especially for lymphocytosis in HP.[110] Therefore, BAL combined with bronchoscopy is recommended if the patient’s condition allows.

Whether a patient with suspected CT-ILD should perform a lung biopsy depends on the clinical context: If the patient cannot be definitively diagnosed after multidisciplinary team (MDT) discussions on clinical, radiological, and bronchoscopic information, or if a vastly different treatment strategy is required according to the differential diagnosis, a lung biopsy may be necessary.[100] However, this decision requires full consideration of benefit-risk analysis, expected outcomes, and consultation with the patient. Evidence for the use of biopsy to diagnose CT-ILD is limited, with only small sample studies reported.[90,111] A study of 418 surgical lung biopsies (SLBs) diagnosing ILD revealed that a specific diagnosis was made in 88.0% of cases with a low incidence of postoperative complications and mortality,[112] with similar results reported in another study.[113] However, SLB should be approached with caution in patients with immunocompromise, mechanical ventilation dependence, or severe respiratory dysfunction, as such patients may have an increased mortality rate.[114] In a recent meta-analysis that included 43 studies, it was found that transbronchial lung cryobiopsy (TBLC) and SLB had diagnostic rates of 76.8% and 93.5%, respectively, for ILD. Additionally, TBLC demonstrated a lower mortality rate compared to SLB.[115] TBLC may be considered an acceptable alternative to SLB in diagnosing undetermined ILD.[116] Compared to conventional transbronchial lung biopsy (TBLB), TBLC offers a larger tissue yield but carries a higher risk of complications.[117] TBLB combined with HRCT can improve the diagnostic accuracy of ILD.[118] A retrospective study of 245 cases of ILD found that bronchoscopy caused a change in the diagnosis of 58 patients (23.7%). Compared to BAL alone, combining BAL with TBLB increased the diagnostic rate from 21.8% to 34.1% (P = 0.027).[119] In clinical practice, the combination of TBLB and BAL is more commonly used for the diagnosis and differential diagnosis of CT-ILD. For patients with peripheral lesions, ultrasound or CT-guided lung biopsy can be considered. While not essential for the diagnosis of CT-ILD, a pathological biopsy can provide valuable guidance for differential diagnosis and pathological classification. Additionally, it is also crucial in evaluating tumor progression and subsequent efficacy. However, biopsy alone is not the definitive diagnostic gold standard, but rather it should be considered within the context of MDT discussions for diagnosis.

MDT discussions involving the integration of clinical, radiological, and pathological information are recommended to enhance confidence in the diagnosis of CT-ILD. One study has shown that MDTs changed the original histological diagnosis by 30% and improved the diagnosis from probable to confident by a further 17%.[120] MDTs have higher reliability in the diagnosis of ILD than radiologists or clinicians alone. A joint statement from the American Thoracic Society (ATS) and the European Respiratory Society (ERS) states that the diagnosis of ILD recommends a dynamic diagnosis by a respiratory physician, a radiologist, and a pathologist.[121] Therefore, we recommend that the diagnosis, treatment, and management of CT-ILD require the joint participation of respiratory physicians, oncologists, radiologists, pathologists, and pharmacists to form the MDT. This approach can improve the accuracy of the diagnosis of CT-ILD and jointly develop rational treatment strategies.

Management

Grading management

CT-ILD was graded according to clinical symptoms and imaging results [Table 2].[122,123] Severity grading facilitates hierarchical management. Treatment for suspected CT-ILD should, in principle, be discontinued. Treatment of CT-ILD is primarily based on glucocorticoids [Figure 3].

Table 2.

Grading systems of CT-ILD.

Grades Symptoms Imaging
Grade 1 Asymptomatic; only clinical or diagnostic observations, no treatment required; no intervention needed. Confined to one lobe of the lung or <25% of lung parenchyma.
Grade 2 Symptomatic; medical intervention indicated; limiting instrumental ADL Involves more than one lobe of the lung or 25–50% of lung parenchyma.
Grade 3 Severe symptoms; hospitalization required; limiting self-care ADL; oxygen indicated Involves all lung lobes or >50% of lung parenchyma.
Grade 4 Life-threatening respiratory compromise; urgent intervention indicated (e.g., intubation and ventilation)

ADL: Activities of daily living; CT-ILD: Cancer therapy-related interstitial lung disease.

Figure 3.

Figure 3

The grading, typing, and phase management of cancer therapy-related ILD. ACT: Anti-cancer treatment; ILD: Interstitial lung disease.

In grade 1 patients, it is advisable to suspend the causative treatment until there is evidence of radiographic improvement, and closely monitor the patient’s condition [Figure 3]. In general, it is usually appropriate to continue the original treatment dosage. For the challenges of ADCs, it is necessary to consider the duration of resolution (same dose if ≤28 days, dosage reduction if >28 days) and the specific medication (permanent discontinuation of trastuzumab emtansine).[45,124] If there is no improvement, patients should be treated as grade 2.

In grade 2 patients, cancer therapy should be temporarily suspended, and a treatment regimen of 0.5–1 mg·kg−1·d−1 of prednisone (or equivalent) should be started [Figure 3]. In patients who have been receiving ICI, it is recommended to immediately administer 1–2 mg·kg−1·d−1 of prednisolone (or equivalent).[125] For patients who have previously received ADCs, prompt administration of prednisolone at a dose of ≥1 mg·kg−1·d−1 is recommended.[124] In most cases, cancer treatment is suspended until resolution to grade 1 or lower, but for patients with ADC-ILD, it is recommended to permanently discontinue ADC.[45,124] If there is no improvement after 48–72 h of prednisone, treat it as grade 3.

In severe cases (≥grade 3), it is recommended to permanently discontinue cancer therapy and receive prednisone (or equivalent) at a dose of 1–2 mg·kg−1·d−1 [Figure 3]. Patients who have been receiving ADCs should be promptly treated with initial pulse therapy with methylprednisolone at a dose of 0.5–1 g/day for at least 3 days, followed by prednisone treatment at a dose of 1.0 mg·kg−1·d−1.[45] Oxygen supplementation is recommended in the presence of signs or symptoms of hypoxemia. In cases where an underlying infection cannot be definitively ruled out or to prevent secondary infections, the consideration of empirical anti-infection is warranted. In corticosteroid-refractory cases who have been receiving ICI and ADCs, treatment with additional immunosuppressive agents such as infliximab, mycophenolate mofetil, cyclophosphamide, or intravenous immunoglobulin can be considered.[45,125] It should be noted that the evidence for the use of these medications in CT-ILD is limited.

Typing management

A prospective study from the French REISAMIC registry reported that CIP has the highest death rate, higher than myocarditis.[6] The lung is a unique organ directly exposed to the external environment, serving as both the site of gas exchange and a portal for microorganisms to invade. Consequently, most patients will be infected with various pathogenic microorganisms simultaneously or exhibit a secondary infection after lung injury caused by various physicochemical factors related to cancer therapy. Treatment of lung injuries may be more complex than that of other organs. There have been reported cases of multiple TKI or chemotherapy-related ILD and RP complicated with a secondary infection.[126,127,128,129] A recent study[130] has proposed the clinical types of CIP, including the mixed type, which refers to CIP complicated with infectious pneumonia, tumor progression, or RP. The authors also proposed the induced type (with different etiologies, such as RT induction, cytomegalovirus, or Epstein-Barr virus reactivation), and pure type (idiopathic, with or without autoimmune disease). We suggest that other CT-ILD may also have a mixed type, for which additional treatment should also be provided to treat comorbidities in addition to CT-ILD, such as anti-infective and anticancer therapy [Figure 3]. Regarding anticancer treatment, patients with CT-ILD may have a compromised health status and limited options for therapy. In such cases, the use of less toxic antiangiogenic agents, such as anlotinib and bevacizumab, can be considered. These medications effectively suppress vascular endothelial growth factor (VEGF), reducing vascular permeability and alleviating pulmonary exudation. In an interleukin-induced pneumonitis model, the anti-VEGF antibody demonstrated protection against vascular-to-alveolar leakage of protein and fluid caused by PD-1 signal blockade.[131] A clinical case provides evidence for the favorable therapeutic effect of combining bevacizumab with corticosteroid treatment in ALK-ILD.[132] In the IMpower150 trial, patients with pneumonitis who did not receive bevacizumab had a reported “Not recovered/Not resolved” outcome rate of 29.4%, while those who received bevacizumab had a significantly lower rate of 9.1%.[131]

RRP is a rare and special CT-ILD mainly found in patients’ previously irradiated lungs after treatment with inciting agents. Extensive research has shown that a range of cancer therapies, including chemotherapy,[133] targeted therapy,[84] and IO[4] can cause RRP. Recent studies have also suggested a possible association between RRP and vaccination[134,135] as well as coronavirus disease 2019 (COVID-19) infection.[136] Based on these findings, we propose that other CT-ILDs may also have an induced type, which is characterized by alterations in the immune microenvironment induced by drugs, viruses, or RT, and the addition of other cancer therapies, which may contribute to the development of ILD. Taken together, we recommend discontinuing potential inducers along with the culprit cancer therapy in cases of the induced type. Furthermore, antiviral therapy should be considered if virus induction is suspected [Figure 3].

Phase management

The dynamic process of lung tissue injury and repair in CT-ILD shows phased characteristics in imaging and pathology over time, owing to differences in treatment time, the severity of CT-ILD, and therapeutic effect. Previous studies have identified three stages of RP: early stage, intermediate stage (acute pneumonitis), and late stage (pulmonary fibrosis).[46,137,138] Min et al[139] proposed that TKI-LID undergoes an acute and chronic process, with an acute phase characterized by DAD. With the help of T lymphocytes and macrophages, the acute injury appears to progress to the chronic phase. A recent study has revealed that patients treated with ICI develop chronic pneumonitis in addition to acute pulmonary toxicity.[140] Recently, Zhou et al[141] proposed a clinical phase of CIP: acute, subacute, and chronic phases. Another study also demonstrated that a proportion of patients will develop chronic ICI pneumonitis.[142] Wijsenbeek et al[143] have classified ILD into acute, subacute, and chronic phases. Therefore, we suggest that other CT-ILDs may also have acute, subacute, and chronic phases, although not all patients experience all three phases. Some patients who receive timely treatment in the acute phase and recover quickly may not enter the chronic phase.

Treatment for CT-ILD in the acute phase should be dominated by glucocorticoid-based anti-inflammatories. From the mechanisms discussed earlier, it is evident that various forms of CT-ILD involve the release of cytokines, particularly during the acute phase. In patients refractory to corticosteroids during this stage, the utilization of cytokine-targeting drugs, including IL-1, IL-6, and TGF inhibitors, may represent a potential treatment approach. A study demonstrated clinical improvement in 27 out of 34 cases (79.4%) of irAEs treated with IL-6 receptor antagonist tocilizumab.[144] ROS plays a crucial role in the occurrence and progression of CT-ILD, and antioxidants and certain natural adjuvants may have a protective effect.[23]

In the chronic phase, corticosteroid treatment shows limited effectiveness for patients, and prolonged use of glucocorticoids may lead to increased adverse reactions. Fibrosis is a prominent feature during this phase, and patients in this stage may benefit from antifibrotic therapies. A small-sample prospective study indicated that pirfenidone may improve exercise capacity in patients with radiation-induced lung fibrosis.[145] There have also been case reports describing the use of pirfenidone or nintedanib used to treat ILD caused by TKIs or ICIs.[146]Table 3 summarizes the potential personalized treatments for CT-ILD patients.

Table 3.

Personalized treatment regimens for the management of CT-ILD.

Targets Proposed drugs Applicable phase and types
VEGF(R) Bevacizumab; anlotinib Acute/subacute; TKI-ILD,[132] CIP[131]
ROS Amifostine Acute; RP[148]
TNF Infliximab Acute; CIP,[149] ADC-ILD[124]
TGF-β Metformin Acute; TKI-ILD[150]
IL-6 Tocilizumab Acute; CIP[144]
Anti-fibrotic drugs Pirfenidone; nintedanib Subacute/chronic; RP,[151] TKI-ILD,[146] and CIP[147]
Immunosuppresive agents Mycophenolate mofetil; cyclophosphamide; intravenous immunoglobulin Acute/subacute; CIP,[149] ADC-ILD[124]

ADC: Antibody-drug conjugates; CIP: Checkpoint inhibitor-related pneumonitis; CT-ILD: Cancer therapy-related interstitial lung disease; IL-6: Interleukin-6; ILD: Interstitial lung disease; ROS: Reactive oxygen species; RP: Radiation pneumonitis; TGF-β: Transforming growth factor-beta; TKI: Tyrosine kinase inhibitor; TNF: Tumor necrosis factor; VEGF(R): Vascular endothelial growth factor receptor.

Conclusions

The prevalence of CT-ILD has increased with the increasing use of cancer therapies. Accurate identification and diagnosis of CT-ILD pose challenges, and the treatment options are limited. The implementation of grading management has proven insufficient to address the current concerns, prompting the proposal of a more personalized approach utilizing typing and phase. Future research on improving diagnostic methods may help in the timely detection of CT-ILD, such as radiomics-based diagnosis and home monitoring. More effective treatment may involve exploring and understanding the mechanisms of CT-ILD.

Funding

This work was supported by the grants from the Guangzhou Science and Technology Major Clinical Project (No. 2023C-DZ06) and China Primary Health Care Foundation (No. BH001496).

Conflicts of interest

None.

Supplementary Material

cm9-138-264-s001.docx (153.6KB, docx)

Footnotes

Chengzhi Zhou and Haiyi Deng contributed equally to this work.

How to cite this article: Zhou CZ, Deng HY, Yang YL, Wang F, Lin XQ, Liu M, Xie XH, Luan T, Zhong NS. Cancer therapy-related interstitial lung disease. Chin Med J 2025;138:264–277. doi: 10.1097/CM9.0000000000003149

References

  • 1.Skeoch S Weatherley N Swift AJ Oldroyd A Johns C Hayton C, et al. Drug-induced interstitial lung disease: A systematic review. J Clin Med 2018;7:356. doi: 10.3390/jcm7100356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Iwasa E Fujiyoshi Y Kubota Y Kimura R Chandler RE Taavola H, et al. Interstitial lung disease as an adverse drug reaction in Japan: Exploration of regulatory actions as a basis for high reporting. Drug Saf 2020;43:1121–1131. doi: 10.1007/s40264-020-00968-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Tamura M Saraya T Fujiwara M Hiraoka S Yokoyama T Yano K, et al. High-resolution computed tomography findings for patients with drug-induced pulmonary toxicity, with special reference to hypersensitivity pneumonitis-like patterns in gemcitabine-induced cases. Oncologist 2013;18:454–459. doi: 10.1634/theoncologist.2012-0248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cousin F, Desir C, Ben Mustapha S, Mievis C, Coucke P, Hustinx R. Incidence, risk factors, and CT characteristics of radiation recall pneumonitis induced by immune checkpoint inhibitor in lung cancer. Radiother Oncol 2021;157:47–55. doi: 10.1016/j.radonc.2021.01.001. [DOI] [PubMed] [Google Scholar]
  • 5.Fujimoto D Kato R Morimoto T Shimizu R Sato Y Kogo M, et al. Characteristics and prognostic impact of pneumonitis during systemic anti-cancer therapy in patients with advanced non-small-cell lung cancer. PLoS One 2016;11:e0168465. doi: 10.1371/journal.pone.0168465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ruste V Goldschmidt V Laparra A Messayke S Danlos FX Romano-Martin P, et al. The determinants of very severe immune-related adverse events associated with immune checkpoint inhibitors: A prospective study of the French REISAMIC registry. Eur J Cancer 2021;158:217–224. doi: 10.1016/j.ejca.2021.08.048. [DOI] [PubMed] [Google Scholar]
  • 7.Xie X Wang X Wu S Yang H Liu J Chen H, et al. Fatal toxic effects related to EGFR tyrosine kinase inhibitors based on 53 cohorts with 9,569 participants. J Thorac Dis 2020;12:4057–4069. doi: 10.21037/jtd-19-4000A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kaku S Horinouchi H Watanabe H Yonemori K Okusaka T Boku N, et al. Incidence and prognostic factors in severe drug-induced interstitial lung disease caused by antineoplastic drug therapy in the real world. J Cancer Res Clin Oncol 2022;148:1737–1746. doi: 10.1007/s00432-022-03932-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Terbuch A Tiu C Candilejo IM Scaranti M Curcean A Bar D, et al. Radiological patterns of drug-induced interstitial lung disease (DILD) in early-phase oncology clinical trials. Clin Cancer Res 2020;26:4805–4813. doi: 10.1158/1078-0432.Ccr-20-0454. [DOI] [PubMed] [Google Scholar]
  • 10.Yonemori K Hirakawa A Kawachi A Kinoshita F Okuma H Nishikawa T, et al. Drug induced interstitial lung disease in oncology phase I trials. Cancer Sci 2016;107:1830–1836. doi: 10.1111/cas.13087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Qi WX, Sun YJ, Shen Z, Yao Y. Risk of interstitial lung disease associated with EGFR-TKIs in advanced non-small-cell lung cancer: A meta-analysis of 24 phase III clinical trials. J Chemother 2015;27:40–51. doi: 10.1179/1973947814y.0000000189. [DOI] [PubMed] [Google Scholar]
  • 12.Suh CH, Park HS, Kim KW, Pyo J, Hatabu H, Nishino M. Pneumonitis in advanced non-small-cell lung cancer patients treated with EGFR tyrosine kinase inhibitor: Meta-analysis of 153 cohorts with 15,713 patients: Meta-analysis of incidence and risk factors of EGFR-TKI pneumonitis in NSCLC. Lung Cancer 2018;123:60–69. doi: 10.1016/j.lungcan.2018.06.032. [DOI] [PubMed] [Google Scholar]
  • 13.Suh CH, Kim KW, Pyo J, Hatabu H, Nishino M. The incidence of ALK inhibitor-related pneumonitis in advanced non-small-cell lung cancer patients: A systematic review and meta-analysis. Lung Cancer 2019;132:79–86. doi: 10.1016/j.lungcan.2019.04.015. [DOI] [PubMed] [Google Scholar]
  • 14.Iacovelli R, Palazzo A, Mezi S, Morano F, Naso G, Cortesi E. Incidence and risk of pulmonary toxicity in patients treated with mTOR inhibitors for malignancy. A meta-analysis of published trials. Acta Oncol 2012;51:873–879. doi: 10.3109/0284186x.2012.705019. [DOI] [PubMed] [Google Scholar]
  • 15.Qi WX, Huang YJ, Yao Y, Shen Z, Min DL. Incidence and risk of treatment-related mortality with mTOR inhibitors everolimus and temsirolimus in cancer patients: A meta-analysis. PLoS One 2013;8:e65166. doi: 10.1371/journal.pone.0065166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Khunger M Rakshit S Pasupuleti V Hernandez AV Mazzone P Stevenson J, et al. Incidence of pneumonitis with use of programmed death 1 and programmed death-ligand 1 inhibitors in non-small cell lung cancer: A systematic review and meta-analysis of trials. Chest 2017;152:271–281. doi: 10.1016/j.chest.2017.04.177. [DOI] [PubMed] [Google Scholar]
  • 17.Nishino M, Giobbie-Hurder A, Hatabu H, Ramaiya NH, Hodi FS. Incidence of programmed cell death 1 inhibitor-related pneumonitis in patients with advanced cancer: A systematic review and meta-analysis. JAMA Oncol 2016;2:1607–1616. doi: 10.1001/jamaoncol.2016.2453. [DOI] [PubMed] [Google Scholar]
  • 18.Zhu Z Shen G Li J Qiu T Fang Q Zheng Y, et al. Incidence of antibody-drug conjugates-related pneumonitis in patients with solid tumors: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2023;184:103960. doi: 10.1016/j.critrevonc.2023.103960. [DOI] [PubMed] [Google Scholar]
  • 19.Powell CA Modi S Iwata H Takahashi S Smit EF Siena S, et al. Pooled analysis of drug-related interstitial lung disease and/or pneumonitis in nine T-DXd monotherapy studies. ESMO Open 2022;7:100554. doi: 10.1016/j.esmoop.2022.100554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Matsuno O. Drug-induced interstitial lung disease: Mechanisms and best diagnostic approaches. Respir Res 2012;13:39. doi: 10.1186/1465-9921-13-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Spagnolo P, Bonniaud P, Rossi G, Sverzellati N, Cottin V. Drug-induced interstitial lung disease. Eur Respir J 2022;60:2102776. doi: 10.1183/13993003.02776-2021. [DOI] [PubMed] [Google Scholar]
  • 22.Yang H Villani RM Wang H Simpson MJ Roberts MS Tang M, et al. The role of cellular reactive oxygen species in cancer chemotherapy. J Exp Clin Cancer Res 2018;37:266. doi: 10.1186/s13046-018-0909-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Xu C, Shang Z, Najafi M. Lung pneumonitis and fibrosis in cancer therapy: A review on cellular and molecular mechanisms. Curr Drug Targets 2022;23:1505–1525. doi: 10.2174/1389450123666220907144131. [DOI] [PubMed] [Google Scholar]
  • 24.Jeong BK, Kim JH, Jung MH, Kang KM, Lee YH. Cytokine profiles of non-small cell lung cancer patients treated with concurrent chemoradiotherapy with regards to radiation pneumonitis severity. J Clin Med 2021;10:699. doi: 10.3390/jcm10040699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sadowska AM, Specenier P, Germonpre P, Peeters M. Antineoplastic therapy-induced pulmonary toxicity. Expert Rev Anticancer Ther 2013;13:997–1006. doi: 10.1586/14737140.2013.817684. [DOI] [PubMed] [Google Scholar]
  • 26.Baron D, Mayo A, Kluger Y. Gemcitabine-induced chronic systemic capillary leak syndrome: A life-threatening disease. Clin Oncol (R Coll Radiol) 2006;18:90–91. doi: 10.1016/j.clon.2005.09.003. [DOI] [PubMed] [Google Scholar]
  • 27.Chaudhary NI Roth GJ Hilberg F Müller-Quernheim J Prasse A Zissel G, et al. Inhibition of PDGF, VEGF and FGF signalling attenuates fibrosis. Eur Respir J 2007;29:976–985. doi: 10.1183/09031936.00152106. [DOI] [PubMed] [Google Scholar]
  • 28.Ohmori T Yamaoka T Ando K Kusumoto S Kishino Y Manabe R, et al. Molecular and clinical features of EGFR-TKI-associated lung injury. Int J Mol Sci 2021;22:792. doi: 10.3390/ijms22020792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Suzuki H, Aoshiba K, Yokohori N, Nagai A. Epidermal growth factor receptor tyrosine kinase inhibition augments a murine model of pulmonary fibrosis. Cancer Res 2003;63:5054–5059. [PubMed] [Google Scholar]
  • 30.Kataoka K Taniguchi H Hasegawa Y Kondoh Y Kimura T Nishiyama O, et al. Interstitial lung disease associated with gefitinib. Respir Med 2006;100:698–704. doi: 10.1016/j.rmed.2005.07.015. [DOI] [PubMed] [Google Scholar]
  • 31.Namba T, Tanaka K, Hoshino T, Azuma A, Mizushima T. Suppression of expression of heat shock protein 70 by gefitinib and its contribution to pulmonary fibrosis. PLoS One 2011;6:e27296. doi: 10.1371/journal.pone.0027296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Suresh K Naidoo J Zhong Q Xiong Y Mammen J de Flores MV, et al. The alveolar immune cell landscape is dysregulated in checkpoint inhibitor pneumonitis. J Clin Invest 2019;129:4305–4315. doi: 10.1172/jci128654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lin X Deng J Deng H Yang Y Sun N Zhou M, et al. Comprehensive analysis of the immune microenvironment in checkpoint inhibitor pneumonitis. Front Immunol 2021;12:818492. doi: 10.3389/fimmu.2021.818492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.de Moel EC Rozeman EA Kapiteijn EH Verdegaal EME Grummels A Bakker JA, et al. Autoantibody development under treatment with immune-checkpoint inhibitors. Cancer Immunol Res 2019;7:6–11. doi: 10.1158/2326-6066.Cir-18-0245. [DOI] [PubMed] [Google Scholar]
  • 35.Gowen MF Giles KM Simpson D Tchack J Zhou H Moran U, et al. Baseline antibody profiles predict toxicity in melanoma patients treated with immune checkpoint inhibitors. J Transl Med 2018;16:82. doi: 10.1186/s12967-018-1452-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tahir SA Gao J Miura Y Blando J Tidwell RSS Zhao H, et al. Autoimmune antibodies correlate with immune checkpoint therapy-induced toxicities. Proc Natl Acad Sci U S A 2019;116:22246–22251. doi: 10.1073/pnas.1908079116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Jia XH Geng LY Jiang PP Xu H Nan KJ Yao Y, et al. The biomarkers related to immune related adverse events caused by immune checkpoint inhibitors. J Exp Clin Cancer Res 2020;39:284. doi: 10.1186/s13046-020-01749-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang YN Lou DF Li DY Jiang W Dong JY Gao W, et al. Elevated levels of IL-17A and IL-35 in plasma and bronchoalveolar lavage fluid are associated with checkpoint inhibitor pneumonitis in patients with non-small cell lung cancer. Oncol Lett 2020;20:611–622. doi: 10.3892/ol.2020.11618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lin X Deng H Yang Y Wu J Qiu G Li S, et al. Peripheral blood biomarkers for early diagnosis, severity, and prognosis of checkpoint inhibitor-related pneumonitis in patients with lung cancer. Front Oncol 2021;11:698832. doi: 10.3389/fonc.2021.698832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Johnson DB Balko JM Compton ML Chalkias S Gorham J Xu Y, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med 2016;375:1749–1755. doi: 10.1056/NEJMoa1609214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Berner F Bomze D Diem S Ali OH Fässler M Ring S, et al. Association of checkpoint inhibitor-induced toxic effects with shared cancer and tissue antigens in non-small cell lung cancer. JAMA Oncol 2019;5:1043–1047. doi: 10.1001/jamaoncol.2019.0402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Miao K, Zhang L. Pathogenesis, pathological characteristics and individualized therapy for immune-related adverse effects. Chin Med J Pulm Crit Care Med 2023;1:215–222. doi:10.1016/j.pccm.2023.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kepp O, Marabelle A, Zitvogel L, Kroemer G. Oncolysis without viruses-inducing systemic anticancer immune responses with local therapies. Nat Rev Clin Oncol 2020;17:49–64. doi: 10.1038/s41571-019-0272-7. [DOI] [PubMed] [Google Scholar]
  • 44.Esfahani K Elkrief A Calabrese C Lapointe R Hudson M Routy B, et al. Moving towards personalized treatments of immune-related adverse events. Nat Rev Clin Oncol 2020;17:504–515. doi: 10.1038/s41571-020-0352-8. [DOI] [PubMed] [Google Scholar]
  • 45.Tarantino P Modi S Tolaney SM Cortés J Hamilton EP Kim SB, et al. Interstitial lung disease induced by anti-ERBB2 antibody-drug conjugates: A review. JAMA Oncol 2021;7:1873–1881. doi: 10.1001/jamaoncol.2021.3595. [DOI] [PubMed] [Google Scholar]
  • 46.Hanania AN, Mainwaring W, Ghebre YT, Hanania NA, Ludwig M. Radiation-induced lung injury: Assessment and management. Chest 2019;156:150–162. doi: 10.1016/j.chest.2019.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Arroyo-Hernández M, Maldonado F, Lozano-Ruiz F, Muñoz-Montaño W, Nuñez-Baez M, Arrieta O. Radiation-induced lung injury: Current evidence. BMC Pulm Med 2021;21:9. doi: 10.1186/s12890-020-01376-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Simpson AB, Paul J, Graham J, Kaye SB. Fatal bleomycin pulmonary toxicity in the west of Scotland 1991-95: A review of patients with germ cell tumours. Br J Cancer 1998;78:1061–1066. doi: 10.1038/bjc.1998.628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hamada T Yasunaga H Nakai Y Isayama H Matsui H Fushimi K, et al. Interstitial lung disease associated with gemcitabine: A Japanese retrospective cohort study. Respirology 2016;21:338–343. doi: 10.1111/resp.12665. [DOI] [PubMed] [Google Scholar]
  • 50.Yoshioka K Abe M Shiko Y Koshikawa K Kawasaki Y Iwasawa S, et al. Clinical characteristics and risk factors of lung injury induced by nab-paclitaxel. Drug Des Devel Ther 2022;16:759–767. doi: 10.2147/dddt.S342283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kudoh S Kato H Nishiwaki Y Fukuoka M Nakata K Ichinose Y, et al. Interstitial lung disease in Japanese patients with lung cancer: A cohort and nested case-control study. Am J Respir Crit Care Med 2008;177:1348–1357. doi: 10.1164/rccm.200710-1501OC. [DOI] [PubMed] [Google Scholar]
  • 52.Koshikawa K Terada J Abe M Iwasawa S Sakayori M Yoshioka K, et al. Clinical characteristics and risk factors of drug-induced lung injury by ALK tyrosine kinase inhibitors: A single center retrospective analysis. Thorac Cancer 2020;11:1495–1502. doi: 10.1111/1759-7714.13416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Vogelius IR, Bentzen SM. A literature-based meta-analysis of clinical risk factors for development of radiation induced pneumonitis. Acta Oncol 2012;51:975–983. doi: 10.3109/0284186x.2012.718093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Takeda M, Okamoto I, Nakagawa K. Pooled safety analysis of EGFR-TKI treatment for EGFR mutation-positive non-small cell lung cancer. Lung Cancer 2015;88:74–79. doi: 10.1016/j.lungcan.2015.01.026. [DOI] [PubMed] [Google Scholar]
  • 55.Nishimura M Toyoda M Takenaka K Imamura Y Chayahara N Kiyota N, et al. The combination of HLA-B*15:01 and DRB1*15:01 is associated with gemcitabine plus erlotinib-induced interstitial lung disease in patients with advanced pancreatic cancer. Cancer Chemother Pharmacol 2016;77:1165–1170. doi: 10.1007/s00280-016-3026-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Schwaiblmair M, Behr W, Foerg W, Berghaus T. Cytochrome P450 polymorphisms and drug-induced interstitial lung disease. Expert Opin Drug Metab Toxicol 2011;7:1547–1560. doi: 10.1517/17425255.2011.629185. [DOI] [PubMed] [Google Scholar]
  • 57.Wijnen PA Drent M Nelemans PJ Kuijpers PM Koek GH Neef C, et al. Role of cytochrome P450 polymorphisms in the development of pulmonary drug toxicity: A case-control study in the Netherlands. Drug Saf 2008;31:1125–1134. doi: 10.2165/0002018-200831120-00008. [DOI] [PubMed] [Google Scholar]
  • 58.Lei X, Du L, Yu W, Wang Y, Ma N, Qu B. GSTP1 as a novel target in radiation induced lung injury. J Transl Med 2021;19:297. doi: 10.1186/s12967-021-02978-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Umemura S Yamane H Suwaki T Katoh T Yano T Shiote Y, et al. Interstitial lung disease associated with gemcitabine treatment in patients with non-small-cell lung cancer and pancreatic cancer. J Cancer Res Clin Oncol 2011;137:1469–1475. doi: 10.1007/s00432-011-1013-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Barlési F, Villani P, Doddoli C, Gimenez C, Kleisbauer JP. Gemcitabine-induced severe pulmonary toxicity. Fundam Clin Pharmacol 2004;18:85–91. doi: 10.1046/j.0767-3981.2003.00206.x. [DOI] [PubMed] [Google Scholar]
  • 61.Tamiya A Naito T Miura S Morii S Tsuya A Nakamura Y, et al. Interstitial lung disease associated with docetaxel in patients with advanced non-small cell lung cancer. Anticancer Res 2012;32:1103–1106. [PubMed] [Google Scholar]
  • 62.Kashiwada T Saito Y Terasaki Y Hisakane K Takeuchi S Sugano T, et al. Interstitial lung disease associated with nanoparticle albumin-bound paclitaxel treatment in patients with lung cancer. Jpn J Clin Oncol 2019;49:165–173. doi: 10.1093/jjco/hyy180. [DOI] [PubMed] [Google Scholar]
  • 63.Sakurada T Kakiuchi S Tajima S Horinouchi Y Okada N Nishisako H, et al. Characteristics of and risk factors for interstitial lung disease induced by chemotherapy for lung cancer. Ann Pharmacother 2015;49:398–404. doi: 10.1177/1060028014566446. [DOI] [PubMed] [Google Scholar]
  • 64.Hotta K Kiura K Takigawa N Yoshioka H Harita S Kuyama S, et al. Comparison of the incidence and pattern of interstitial lung disease during erlotinib and gefitinib treatment in Japanese patients with non-small cell lung cancer: The Okayama Lung Cancer Study Group experience. J Thorac Oncol 2010;5:179–184. doi: 10.1097/JTO.0b013e3181ca12e0. [DOI] [PubMed] [Google Scholar]
  • 65.Takeda A Kunieda E Ohashi T Aoki Y Oku Y Enomoto T, et al. Severe COPD is correlated with mild radiation pneumonitis following stereotactic body radiotherapy. Chest 2012;141:858–866. doi: 10.1378/chest.11-1193. [DOI] [PubMed] [Google Scholar]
  • 66.Ueki N Matsuo Y Togashi Y Kubo T Shibuya K Iizuka Y, et al. Impact of pretreatment interstitial lung disease on radiation pneumonitis and survival after stereotactic body radiation therapy for lung cancer. J Thorac Oncol 2015;10:116–125. doi: 10.1097/jto.0000000000000359. [DOI] [PubMed] [Google Scholar]
  • 67.Binder D, Hübner RH, Temmesfeld-Wollbrück B, Schlattmann P. Pulmonary toxicity among cancer patients treated with a combination of docetaxel and gemcitabine: A meta-analysis of clinical trials. Cancer Chemother Pharmacol 2011;68:1575–1583. doi: 10.1007/s00280-011-1648-2. [DOI] [PubMed] [Google Scholar]
  • 68.Hamada T Yasunaga H Nakai Y Isayama H Matsui H Fushimi K, et al. Interstitial lung disease associated with gemcitabine: A Japanese retrospective cohort study. Respirology 2016;21:338–343. doi: 10.1111/resp.12665. [DOI] [PubMed] [Google Scholar]
  • 69.Ma K, Lu Y, Jiang S, Tang J, Li X, Zhang Y. The relative risk and incidence of immune checkpoint inhibitors related pneumonitis in patients with advanced cancer: A meta-analysis. Front Pharmacol 2018;9:1430. doi: 10.3389/fphar.2018.01430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Suresh K Voong KR Shankar B Forde PM Ettinger DS Marrone KA, et al. Pneumonitis in non-small cell lung cancer patients receiving immune checkpoint immunotherapy: Incidence and risk factors. J Thorac Oncol 2018;13:1930–1939. doi: 10.1016/j.jtho.2018.08.2035. [DOI] [PubMed] [Google Scholar]
  • 71.Abuhelwa Z, Alloghbi A, Alqahtani A, Nagasaka M. Trastuzumab deruxtecan-induced interstitial lung disease/pneumonitis in ERBB2-positive advanced solid malignancies: A systematic review. Drugs 2022;82:979–987. doi: 10.1007/s40265-022-01736-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Oshima Y, Tanimoto T, Yuji K, Tojo A. EGFR-TKI-associated interstitial pneumonitis in nivolumab-treated patients with non-small cell lung cancer. JAMA Oncol 2018;4:1112–1115. doi: 10.1001/jamaoncol.2017.4526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Su Q Zhu EC Wu JB Li T Hou YL Wang DY, et al. Risk of pneumonitis and pneumonia associated with immune checkpoint inhibitors for solid tumors: A systematic review and meta-analysis. Front Immunol 2019;10:108. doi: 10.3389/fimmu.2019.00108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chen X Zhang Z Hou X Zhang Y Zhou T Liu J, et al. Immune-related pneumonitis associated with immune checkpoint inhibitors in lung cancer: A network meta-analysis. J Immunother Cancer 2020;8:e001170. doi: 10.1136/jitc-2020-001170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Nozawa K Takatsuka D Endo Y Horisawa N Ozaki Y Kataoka A, et al. Association between bevacizumab with cancer drug therapies and drug-induced interstitial lung disease in patients with solid tumor: A systematic review and meta-analysis of randomized clinical trials. Crit Rev Oncol Hematol 2022;174:103703. doi: 10.1016/j.critrevonc.2022.103703. [DOI] [PubMed] [Google Scholar]
  • 76.Taghian AG Assaad SI Niemierko A Kuter I Younger J Schoenthaler R, et al. Risk of pneumonitis in breast cancer patients treated with radiation therapy and combination chemotherapy with paclitaxel. J Natl Cancer Inst 2001;93:1806–1811. doi: 10.1093/jnci/93.23.1806. [DOI] [PubMed] [Google Scholar]
  • 77.Perez EG Peloche MGB Quiroz CFA Garcia Y Portulas ED Monne JMS, et al. Prediction parameters for radiation pneumonitis in patients with stage III NSCLC. J Clin Oncol 2014;32:e18538. doi: 10.1200/jco.2014.32.15_suppl.e18538. [Google Scholar]
  • 78.Xu Z, Feng J, Weng Y, Jin Y, Peng M. Combination of immune checkpoint inhibitors and radiotherapy for advanced non-small-cell lung cancer and prostate cancer: A meta-analysis. J Oncol 2021;2021:6631643. doi: 10.1155/2021/6631643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Xu K Liang J Zhang T Zhou Z Chen D Feng Q, et al. Clinical outcomes and radiation pneumonitis after concurrent EGFR-tyrosine kinase inhibitors and radiotherapy for unresectable stage III non-small cell lung cancer. Thorac Cancer 2021;12:814–823. doi: 10.1111/1759-7714.13816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Tang X Shen Y Meng Y Hou L Zhou C Yu C, et al. Radiation-induced lung damage in patients treated with stereotactic body radiotherapy after EGFR-TKIs: Is there any difference from stereotactic body radiotherapy alone? Ann Palliat Med 2021;10:2832–2842. doi: 10.21037/apm-20-1116. [DOI] [PubMed] [Google Scholar]
  • 81.Jiang T, Min W, Li Y, Yue Z, Wu C, Zhou C. Radiotherapy plus EGFR TKIs in non-small cell lung cancer patients with brain metastases: An update meta-analysis. Cancer Med 2016;5:1055–1065. doi: 10.1002/cam4.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Li X Wang F Jia H Lian Z Ren K Yuan Z, et al. Efficacy and safety of EGFR inhibitors and radiotherapy in locally advanced non-small-cell lung cancer: A meta-analysis. Future Oncol 2022;18:3055–3065. doi: 10.2217/fon-2022-0491. [DOI] [PubMed] [Google Scholar]
  • 83.Jia W Gao Q Wang M Li J Jing W Yu J, et al. Overlap time is an independent risk factor of radiation pneumonitis for patients treated with simultaneous EGFR-TKI and thoracic radiotherapy. Radiat Oncol 2021;16:41. doi: 10.1186/s13014-021-01765-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Chiang CL, Chen YW, Wu MH, Huang HC, Tsai CM, Chiu CH. Radiation recall pneumonitis induced by epidermal growth factor receptor-tyrosine kinase inhibitor in patients with advanced nonsmall-cell lung cancer. J Chin Med Assoc 2016;79:248–255. doi: 10.1016/j.jcma.2016.01.008. [DOI] [PubMed] [Google Scholar]
  • 85.Mu F Fan B Li B Qin W Li H Wang C, et al. Comparison of the incidence rate of radiation pneumonitis observed in patients with advanced lung adenocarcinoma treated with simultaneous thoracic radiotherapy and 1G/2G/3G EGFR-TKIs. Cancer Manag Res 2023;15:351–362. doi: 10.2147/cmar.S404874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kubo K Azuma A Kanazawa M Kameda H Kusumoto M Genma A, et al. Consensus statement for the diagnosis and treatment of drug-induced lung injuries. Respir Investig 2013;51:260–277. doi: 10.1016/j.resinv.2013.09.001. [DOI] [PubMed] [Google Scholar]
  • 87.Schwaiblmair M, Behr W, Haeckel T, Märkl B, Foerg W, Berghaus T. Drug induced interstitial lung disease. Open Respir Med J 2012;6:63–74. doi: 10.2174/1874306401206010063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Cleverley JR, Screaton NJ, Hiorns MP, Flint JD, Müller NL. Drug-induced lung disease: High-resolution CT and histological findings. Clin Radiol 2002;57:292–299. doi: 10.1053/crad.2001.0792. [DOI] [PubMed] [Google Scholar]
  • 89.Padley SP, Adler B, Hansell DM, Müller NL. High-resolution computed tomography of drug-induced lung disease. Clin Radiol 1992;46:232–236. doi: 10.1016/s0009-9260(05)80161-8. [DOI] [PubMed] [Google Scholar]
  • 90.Piciucchi S Romagnoli M Chilosi M Bigliazzi C Dubini A Beomonte Zobel B, et al. Prospective evaluation of drug-induced lung toxicity with high-resolution CT and transbronchial biopsy. Radiol Med 2011;116:246–263. doi: 10.1007/s11547-010-0608-y. [DOI] [PubMed] [Google Scholar]
  • 91.Endo M, Johkoh T, Kimura K, Yamamoto N. Imaging of gefitinib-related interstitial lung disease: Multi-institutional analysis by the West Japan Thoracic Oncology Group. Lung Cancer 2006;52:135–140. doi: 10.1016/j.lungcan.2006.02.002. [DOI] [PubMed] [Google Scholar]
  • 92.Yuan MK Chang CY Chang SC Chang SJ Tang GJ Wei YF, et al. Imaging patterns and prognosis of patients with gefitinib-related interstitial lung disease. Int J Clin Pharmacol Ther 2011;49:587–593. doi: 10.5414/cp201566. [DOI] [PubMed] [Google Scholar]
  • 93.Nishino M, Brais LK, Brooks NV, Hatabu H, Kulke MH, Ramaiya NH. Drug-related pneumonitis during mammalian target of rapamycin inhibitor therapy in patients with neuroendocrine tumors: A radiographic pattern-based approach. Eur J Cancer 2016;53:163–170. doi: 10.1016/j.ejca.2015.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Nishino M, Boswell EN, Hatabu H, Ghobrial IM, Ramaiya NH. Drug-related pneumonitis during mammalian target of rapamycin inhibitor therapy: Radiographic pattern-based approach in waldenström macroglobulinemia as a paradigm. Oncologist 2015;20:1077–1083. doi: 10.1634/theoncologist.2015-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Naidoo J Wang X Woo KM Iyriboz T Halpenny D Cunningham J, et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J Clin Oncol 2017;35:709–717. doi: 10.1200/jco.2016.68.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Watanabe S Ota T Hayashi M Ishikawa H Otsubo A Shoji S, et al. Prognostic significance of the radiologic features of pneumonitis induced by anti-PD-1 therapy. Cancer Med 2020;9:3070–3077. doi: 10.1002/cam4.2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Nishino M Ramaiya NH Awad MM Sholl LM Maattala JA Taibi M, et al. PD-1 inhibitor-related pneumonitis in advanced cancer patients: Radiographic patterns and clinical course. Clin Cancer Res 2016;22:6051–6060. doi: 10.1158/1078-0432.Ccr-16-1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Flieder DB, Travis WD. Pathologic characteristics of drug-induced lung disease. Clin Chest Med 2004;25:37–45. doi: 10.1016/s0272-5231(03)00138-2. [DOI] [PubMed] [Google Scholar]
  • 99.Silva CI, Müller NL. Drug-induced lung diseases: Most common reaction patterns and corresponding high-resolution CT manifestations. Semin Ultrasound CT MR 2006;27:111–116. doi: 10.1053/j.sult.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 100.Anticancer Drug-induced Interstitial Lung Disease Management Group . Expert consensus on the diagnosis and treatment of anticancer drug-induced interstitial lung disease. Zhonghua Zhong Liu Za Zhi 2022;44:693–702. doi: 10.3760/cma.j.cn112152-20220412-00244. [DOI] [PubMed] [Google Scholar]
  • 101.Desai SR, Prosch H, Galvin JR. Plain film and HRCT diagnosis of interstitial lung disease. In: Hodler J, Kubik-Huch RA, von Schulthess GK, eds. Diseases of the chest, breast, heart and vessels 2019-2022: Diagnostic and interventional imaging. Cham (CH): Springer; 2019: 37–45. [Google Scholar]
  • 102.Johkoh T Lee KS Nishino M Travis WD Ryu JH Lee HY, et al. Chest CT diagnosis and clinical management of drug-related pneumonitis in patients receiving molecular targeting agents and immune checkpoint inhibitors: A position paper from the Fleischner Society. Chest 2021;159:1107–1125. doi: 10.1016/j.chest.2020.11.027. [DOI] [PubMed] [Google Scholar]
  • 103.Conte P Ascierto PA Patelli G Danesi R Vanzulli A Sandomenico F, et al. Drug-induced interstitial lung disease during cancer therapies: Expert opinion on diagnosis and treatment. ESMO Open 2022;7:100404. doi: 10.1016/j.esmoop.2022.100404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Taylor CR. Diagnostic imaging techniques in the evaluation of drug-induced pulmonary disease. Clin Chest Med 1990;11:87–94. doi: 10.1016/S0272-5231(21)00674-2. [PubMed] [Google Scholar]
  • 105.Hogea SP, Tudorache E, Pescaru C, Marc M, Oancea C. Bronchoalveolar lavage: Role in the evaluation of pulmonary interstitial disease. Expert Rev Respir Med 2020;14:1117–1130. doi: 10.1080/17476348.2020.1806063. [DOI] [PubMed] [Google Scholar]
  • 106.Costabel U, Uzaslan E, Guzman J. Bronchoalveolar lavage in drug-induced lung disease. Clin Chest Med 2004;25:25–35. doi: 10.1016/s0272-5231(03)00143-6. [DOI] [PubMed] [Google Scholar]
  • 107.Solazzo A, Botta C, Nava F, Baisi A, Bonucchi D, Cappelli G. Interstitial lung disease after kidney transplantation and the role of everolimus. Transplant Proc 2016;48:349–351. doi: 10.1016/j.transproceed.2015.12.039. [DOI] [PubMed] [Google Scholar]
  • 108.Gharsalli H, Mlika M, Sahnoun I, Maalej S, Douik El Gharbi L, Mezni FE. The utility of bronchoalveolar lavage in the evaluation of interstitial lung diseases: A clinicopathological perspective. Semin Diagn Pathol 2018;35:280–287. doi: 10.1053/j.semdp.2018.08.003. [DOI] [PubMed] [Google Scholar]
  • 109.Meyer KC Raghu G Baughman RP Brown KK Costabel U du Bois RM, et al. An official American Thoracic Society clinical practice guideline: The clinical utility of bronchoalveolar lavage cellular analysis in interstitial lung disease. Am J Respir Crit Care Med 2012;185:1004–1014. doi: 10.1164/rccm.201202-0320ST. [DOI] [PubMed] [Google Scholar]
  • 110.Kebbe J, Abdo T. Interstitial lung disease: The diagnostic role of bronchoscopy. J Thorac Dis 2017;9:S996–S1010. doi: 10.21037/jtd.2017.06.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Romagnoli M Bigliazzi C Casoni G Chilosi M Carloni A Dubini A, et al. The role of transbronchial lung biopsy for the diagnosis of diffuse drug-induced lung disease: A case series of 44 patients. Sarcoidosis Vasc Diffuse Lung Dis 2008;25:36–45. [PubMed] [Google Scholar]
  • 112.Zhang D, Liu Y. Surgical lung biopsies in 418 patients with suspected interstitial lung disease in China. Intern Med 2010;49:1097–1102. doi: 10.2169/internalmedicine.49.3225. [DOI] [PubMed] [Google Scholar]
  • 113.Rotolo N Imperatori A Dominioni L Facchini A Conti V Castiglioni M, et al. Efficacy and safety of surgical lung biopsy for interstitial disease. Experience of 161 consecutive patients from a single institution in Italy. Sarcoidosis Vasc Diffuse Lung Dis 2015;32:251–258. [PubMed] [Google Scholar]
  • 114.Han Q Luo Q Xie JX Wu LL Liao LY Zhang XX, et al. Diagnostic yield and postoperative mortality associated with surgical lung biopsy for evaluation of interstitial lung diseases: A systematic review and meta-analysis. J Thorac Cardiovasc Surg 2015;149:1394–1401.e1. doi: 10.1016/j.jtcvs.2014.12.057. [DOI] [PubMed] [Google Scholar]
  • 115.Rodrigues I Estêvão Gomes R Coutinho LM Rego MT Machado F Morais A, et al. Diagnostic yield and safety of transbronchial lung cryobiopsy and surgical lung biopsy in interstitial lung diseases: A systematic review and meta-analysis. Eur Respir Rev 2022;31:210280. doi: 10.1183/16000617.0280-2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Raghu G Remy-Jardin M Richeldi L Thomson CC Inoue Y Johkoh T, et al. Idiopathic pulmonary fibrosis (an Update) and progressive pulmonary fibrosis in adults: An official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med 2022;205:e18–e47. doi: 10.1164/rccm.202202-0399ST. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Korevaar DA Colella S Fally M Camuset J Colby TV Hagmeyer L, et al. European Respiratory Society guidelines on transbronchial lung cryobiopsy in the diagnosis of interstitial lung diseases. Eur Respir J 2022;60:2200425. doi: 10.1183/13993003.00425-2022. [DOI] [PubMed] [Google Scholar]
  • 118.Sheth JS Belperio JA Fishbein MC Kazerooni EA Lagstein A Murray S, et al. Utility of transbronchial vs surgical lung biopsy in the diagnosis of suspected fibrotic interstitial lung disease. Chest 2017;151:389–399. doi: 10.1016/j.chest.2016.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Adams TN Batra K Silhan L Anand V Joerns EK Moore S, et al. Utility of bronchoalveolar lavage and transbronchial biopsy in patients with interstitial lung disease. Lung 2020;198:803–810. doi: 10.1007/s00408-020-00389-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Burge PS, Reynolds J, Trotter S, Burge GA, Walters G. Histologist’s original opinion compared with multidisciplinary team in determining diagnosis in interstitial lung disease. Thorax 2017;72:280–281. doi: 10.1136/thoraxjnl-2016-208776. [DOI] [PubMed] [Google Scholar]
  • 121.American Thoracic Society; European Respiratory Society . American Thoracic Society/European Respiratory Society International Multidisciplinary Consensus Classification of the Idiopathic Interstitial Pneumonias. This joint statement of the American Thoracic Society (ATS), and the European Respiratory Society (ERS) was adopted by the ATS board of directors, June 2001 and by the ERS Executive Committee, June 2001. Am J Respir Crit Care Med 2002;165:277–304. doi: 10.1164/ajrccm.165.2.ats01. [DOI] [PubMed] [Google Scholar]
  • 122.Brahmer JR Lacchetti C Schneider BJ Atkins MB Brassil KJ Caterino JM, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018;36:1714–1768. doi: 10.1200/jco.2017.77.6385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Shah RR. Tyrosine kinase inhibitor-induced interstitial lung disease: Clinical features, diagnostic challenges, and therapeutic dilemmas. Drug Saf 2016;39:1073–1091. doi: 10.1007/s40264-016-0450-9. [DOI] [PubMed] [Google Scholar]
  • 124.Swain SM Nishino M Lancaster LH Li BT Nicholson AG Bartholmai BJ, et al. Multidisciplinary clinical guidance on trastuzumab deruxtecan (T-DXd)-related interstitial lung disease/pneumonitis-focus on proactive monitoring, diagnosis, and management. Cancer Treat Rev 2022;106:102378. doi: 10.1016/j.ctrv.2022.102378. [DOI] [PubMed] [Google Scholar]
  • 125.Puzanov I Diab A Abdallah K Bingham CO 3rd Brogdon C Dadu R, et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer 2017;5:95. doi: 10.1186/s40425-017-0300-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhang J, Zhan Y, Ouyang M, Qin Y, Zhou C, Chen R. Fatal interstitial lung disease associated with icotinib. J Thorac Dis 2014;6:E267–271. doi: 10.3978/j.issn.2072-1439.2014.10.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Kato K Noguchi S Naito K Ikushima I Hanaka T Yamasaki K, et al. Pulmonary nocardiosis caused by nocardia exalbida in a patient with lung cancer and radiation pneumonitis: A case report and literature review. Intern Med 2019;58:1605–1611. doi: 10.2169/internalmedicine.2177-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Bae K, Jeon KN, Choi HS, Song DH, Kim HC. Pulmonary tuberculosis mimicking radiation pneumonitis in a patient with neck malignancy: A case report. Medicine (Baltimore) 2019;98:e16398. doi: 10.1097/md.0000000000016398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wang GS, Yang KY, Perng RP. Life-threatening hypersensitivity pneumonitis induced by docetaxel (taxotere). Br J Cancer 2001;85:1247–1250. doi: 10.1054/bjoc.2001.2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lin X Deng H Chen L Wu D Chen X Yang Y, et al. Clinical types of checkpoint inhibitor-related pneumonitis in lung cancer patients: A multicenter experience. Transl Lung Cancer Res 2021;10:415–429. doi: 10.21037/tlcr-20-1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Iwai T, Sugimoto M, Patel H, Yorozu K, Kurasawa M, Kondoh O. Anti-VEGF antibody protects against alveolar exudate leakage caused by vascular hyperpermeability, resulting in mitigation of pneumonitis induced by immunotherapy. Mol Cancer Ther 2021;20:2519–2526. doi: 10.1158/1535-7163.Mct-21-0031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Xie X Guo B Lin X Qin Y Ouyang M Li S, et al. Successful therapy with bevacizumab combined with corticosteroids for crizotinib-induced interstitial lung disease. Angiogenesis 2019;22:477–479. doi: 10.1007/s10456-019-09673-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Ding X, Ji W, Li J, Zhang X, Wang L. Radiation recall pneumonitis induced by chemotherapy after thoracic radiotherapy for lung cancer. Radiat Oncol 2011;6:24. doi: 10.1186/1748-717x-6-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hughes NM, Hammer MM, Awad MM, Jacene HA. Radiation recall pneumonitis on FDG PET/CT triggered by COVID-19 vaccination. Clin Nucl Med 2022;47:e281–e283. doi: 10.1097/rlu.0000000000003980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Shinada K, Murakami S, Yoshida D, Saito H. Radiation recall pneumonitis after COVID-19 vaccination. Thorac Cancer 2022;13:144–145. doi: 10.1111/1759-7714.14239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kurosaki H, Utsumi N, Miura K. A case of suspected radiation recall pneumonitis after a COVID-19 infection. Cureus 2021;13:e13688. doi: 10.7759/cureus.13688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Coggle JE, Lambert BE, Moores SR. Radiation effects in the lung. Environ Health Perspect 1986;70:261–291. doi: 10.1289/ehp.8670261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Gross NJ. Pulmonary effects of radiation therapy. Ann Intern Med 1977;86:81–92. doi: 10.7326/0003-4819-86-1-81. [DOI] [PubMed] [Google Scholar]
  • 139.Min JH Lee HY Lim H Ahn MJ Park K Chung MP, et al. Drug-induced interstitial lung disease in tyrosine kinase inhibitor therapy for non-small cell lung cancer: A review on current insight. Cancer Chemother Pharmacol 2011;68:1099–1109. doi: 10.1007/s00280-011-1737-2. [DOI] [PubMed] [Google Scholar]
  • 140.Johnson DB, Nebhan CA, Moslehi JJ, Balko JM. Immune-checkpoint inhibitors: Long-term implications of toxicity. Nat Rev Clin Oncol 2022;19:254–267. doi: 10.1038/s41571-022-00600-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zhou C Yang Y Lin X Fang N Chen L Jiang J, et al. Proposed clinical phases for the improvement of personalized treatment of checkpoint inhibitor-related pneumonitis. Front Immunol 2022;13:935779. doi: 10.3389/fimmu.2022.935779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Naidoo J Cottrell TR Lipson EJ Forde PM Illei PB Yarmus LB, et al. Chronic immune checkpoint inhibitor pneumonitis. J Immunother Cancer 2020;8:e000840. doi: 10.1136/jitc-2020-000840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wijsenbeek M, Suzuki A, Maher TM. Interstitial lung diseases. Lancet 2022;400:769–786. doi: 10.1016/s0140-6736(22)01052-2. [DOI] [PubMed] [Google Scholar]
  • 144.Stroud CR Hegde A Cherry C Naqash AR Sharma N Addepalli S, et al. Tocilizumab for the management of immune mediated adverse events secondary to PD-1 blockade. J Oncol Pharm Pract 2019;25:551–557. doi: 10.1177/1078155217745144. [DOI] [PubMed] [Google Scholar]
  • 145.Dy GK, Prasad D, Kumar P, Attwood K, Adjei AA. A phase 2 randomized, double-blind, placebo-controlled study evaluating nintedanib versus placebo as prophylaxis against radiation pneumonitis in patients with unresectable NSCLC undergoing chemoradiation therapy. J Thorac Oncol 2021;16:e19–e20. doi: 10.1016/j.jtho.2020.11.019. [DOI] [PubMed] [Google Scholar]
  • 146.Fang W, Huang Y, Gan J, He B, Zhang L. Nintedanib effect in osimertinib-induced interstitial pneumonia. J Thorac Oncol 2020;15:e34–e35. doi: 10.1016/j.jtho.2019.09.086. [DOI] [PubMed] [Google Scholar]
  • 147.Xie XH Deng HY Lin XQ Wu JH Liu M Xie ZH, et al. Case report: Nintedanib for pembrolizumab-related pneumonitis in a patient with non-small cell lung cancer. Front Oncol 2021;11:673877. doi: 10.3389/fonc.2021.673877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Antonadou D Coliarakis N Synodinou M Athanassiou H Kouveli A Verigos C, et al. Randomized phase III trial of radiation treatment ± amifostine in patients with advanced-stage lung cancer. Int J Radiat Oncol Biol Phys 2001;51:915–922. doi: 10.1016/s0360-3016(01)01713-8. [DOI] [PubMed] [Google Scholar]
  • 149.Beattie J Rizvi H Fuentes P Luo J Schoenfeld A Lin IH, et al. Success and failure of additional immune modulators in steroid-refractory/resistant pneumonitis related to immune checkpoint blockade. J Immunother Cancer 2021;9:e001884. doi: 10.1136/jitc-2020-001884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Li L Huang W Li K Zhang K Lin C Han R, et al. Metformin attenuates gefitinib-induced exacerbation of pulmonary fibrosis by inhibition of TGF-β signaling pathway. Oncotarget 2015;6:43605–43619. doi: 10.18632/oncotarget.6186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Simone NL Soule BP Gerber L Augustine E Smith S Altemus RM, et al. Oral pirfenidone in patients with chronic fibrosis resulting from radiotherapy: A pilot study. Radiat Oncol 2007;2:19. doi: 10.1186/1748-717x-2-19. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Chinese Medical Journal are provided here courtesy of Wolters Kluwer Health

RESOURCES