Skip to main content
  • Educational Review
  • Open access
  • Published:

The radiologist’s role in detecting systemic anticancer therapy-related interstitial lung disease: an educational review

Abstract

Systemic anticancer therapies (SACTs) are the leading cause of drug-induced interstitial lung disease (ILD). As more novel SACTs become approved, the incidence of this potentially life-threatening adverse event (AE) may increase. Early detection of SACT-related ILD allows for prompt implementation of drug-specific management recommendations, improving the likelihood of AE resolution and, in some instances, widening the patient’s eligibility for future cancer treatment options. ILD requires a diagnosis of exclusion through collaboration with the patient’s multidisciplinary team to rule out other possible etiologies of new or worsening respiratory signs and symptoms. At Grade 1, ILD is asymptomatic, and thus the radiologist is key to detecting the AE prior to the disease severity worsening. Planned computed tomography scans should be reviewed for the presence of ILD in addition to being assessed for tumor response to treatment, and when ILD is suspected, a high-resolution computed tomography (HRCT) scan should be requested immediately. An HRCT scan, with < 2-mm slice thickness, is the most appropriate method for detecting ILD. Multiple patterns of ILD exist, which can impact patient prognosis. The four main patterns include acute interstitial pneumonia / acute respiratory distress syndrome, organizing pneumonia, hypersensitivity pneumonitis, and non-specific interstitial pneumonia; their distinct radiological features, along with rarer patterns, are discussed here. Furthermore, HRCT is essential for following the course of ILD and might help to determine the intensity of AE management and the appropriateness of re-challenging with SACT, where indicated by drug-specific prescribing information. ILD events should be monitored closely until complete resolution.

Critical relevance statement

The incidence of potentially treatment-limiting and life-threatening systemic anticancer therapy-related interstitial lung disease (SACT-related ILD) events is likely increasing as more novel regimens become approved. This review provides best-practice recommendations for the early detection of SACT-related ILD by radiologists.

Key Points

  • Radiologists are crucial in detecting asymptomatic (Grade 1) ILD before severity/prognosis worsens.

  • High-resolution computed tomography is the most appropriate method for detecting ILD.

  • Drug-induced ILD is a diagnosis of exclusion, involving a multidisciplinary team.

  • Familiarity with common HRCT patterns, described here, is key for prompt detection.

  • Physicians should highlight systemic anticancer therapies (SACTs) with a known risk for interstitial lung diseases (ILD) on scan requisitions.

Graphical Abstract

Background

Introduction

Interstitial lung disease (ILD) is a heterogeneous group of lung disorders, including pneumonitis, that manifest as inflammation and/or fibrosis of the lungs [1, 2]. ILD can have a broad range of etiologies, occurring because of hypersensitivity; exposure to specific toxic metals; or autoimmune, inflammatory, and genetic disorders; or secondary to smoking-related disorders [3]. In Europe, most cases of ILD are idiopathic or related to sarcoidosis, with between 11.5% and 38.6% of cases having no identifiable cause [4]. A subset of ILD events result from exposure to drugs [3, 5]; systemic anticancer therapies (SACTs) are the leading cause of drug-induced ILD [6]. As the number of novel anticancer agents and indications for existing anticancer agents associated with ILD increases, SACT-related ILD is expected to become more prevalent across cancer types [7]. A recently published position paper from the Fleischner Society identified and answered specific questions regarding the diagnostic criteria for and management of drug-induced ILD in patients with cancer receiving molecular targeting agents and immune checkpoint inhibitors (ICIs) [8]. In this review, the role of the radiologist in detecting SACT-related ILD is outlined, and the authors provide best-practice recommendations on the most appropriate imaging criteria and key radiological abnormalities to be vigilant for when monitoring patients.

The importance of early detection of SACT-related ILD

The severity of ILD events is typically graded from 1 to 5 using the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 [7, 9]. Grade 1 ILD is asymptomatic, and Grade ≥ 2 events are symptomatic (Table 1) [9]. Fine crackles on chest auscultation and indications of interstitial changes on chest imaging—as well as fever or new or worsening respiratory symptoms, including dyspnea, cough, and hypoxia at rest or with exertion—indicate that a patient may have ILD [10, 11].

Table 1 CTCAE version 5.0 grading of ILD, defined as focal or diffuse inflammation affecting the lung parenchyma [9]

Time to diagnosis of SACT-related ILD has clinical and therapeutic consequences. Early diagnosis allows for prompt treatment of ILD with corticosteroids as recommended in the prescribing information (PI) of some SACTs [12, 13], increasing the likelihood of complete resolution of the adverse event (AE) [7], and also providing the opportunity to rechallenge patients who are eligible for resuming their treatment upon resolution of the AE, per drug-specific PI [12, 13]. Urgent reporting of ILD to a patient’s treating physician can also prevent redosing until AE resolution, reducing the risk of escalating severity.

As Grade 1 ILD is defined as an asymptomatic event, with radiological findings only [9], the radiologist is key for the early identification of ILD via proactive monitoring for manifestations before symptom onset. Proactive communication between treating physicians and radiologists provides more opportunity for prompt detection of SACT-related ILD. Treating physicians should indicate on scan requisitions when a patient is receiving SACTs with a relevant risk for ILD. Planned computed tomography (CT) scans should be assessed for the presence of ILD in addition to treatment response [14], especially when tumor assessments and redosing are scheduled for the same day. Where ILD is suspected, radiologists should be consulted, and a joint review of scans can be considered when the patient is being treated with a drug known to increase the risk of developing ILD. During routine CT scans for tumor response, radiologists should inform the ordering physician when ILD is suspected. When diagnosing SACT-related ILD, a multidisciplinary approach should be used to rule out other causes [15]. A pulmonary consultation assessing the potential impact of suspected lung damage with a pulmonary function test is also recommended for suspected ILD, particularly in symptomatic cases [16].

Pathogenesis of SACT-related ILD

Two basic mechanisms, direct cytotoxic and immune-mediated lung injury, are commonly accepted as the likely underlying processes for SACT-related ILD, despite the exact pathogenesis not being completely understood [17, 18]. Direct cytotoxic effects may manifest via multiple pathways resulting in damage to alveolar Type I epithelial cells, airway epithelial cells, or vascular endothelial cells [17, 19]. Neutrophilia has been observed in patients with SACT-related ILD [20], and has been theorized to trigger the release of neutrophil extracellular traps, inducing pulmonary fibrosis [21]. Metabolism of SACTs in the lungs may result in the release of highly cytotoxic reactive oxygen species leading to pulmonary injuries [19]. Several theories have been posed to explain the association between antibody-drug conjugates (ADCs) and ILD, including the target-dependent uptake and catabolism of ADCs or cytotoxic effects resulting from potential uptake and catabolism in off-target lung epithelial cells [22, 23]. The bystander effect, related to the membrane-permeable tetrapeptide-based cleavable linker present in some ADCs, may also be a mechanism for cytotoxic injury [23]. As ILD has been observed in patients receiving ADCs with a range of different therapeutic targets, including human epidermal growth factor receptor 2 (HER2), folate receptor α, and trophoblast cell surface antigen 2, it is likely that the lung toxicity associated with ADCs is not related to any specific target protein [24]. Some SACTs may cause amplified autoimmune responses and subsequent inflammation and immune-mediated lung injury [19]. Immune cells may be activated by SACTs that mimic antigens through non-recognition of the drug triggering pro-inflammatory signals, or via the hapten hypothesis [17, 19], where drugs are proposed to modify cellular proteins and generate novel molecules that stimulate multiple immune pathways [25].

Risk factors for and incidence of SACT-related ILD

The risk of developing ILD has been associated with various types of SACT, with warnings in the labels of specific ADCs, anti-programmed cell death protein 1 (PD-1) drugs, cyclin-dependent kinase 4/6 inhibitors (CDK4/6i), mammalian target of rapamycin (mTOR) inhibitors, and tyrosine kinase inhibitors (TKIs) [12, 13, 26,27,28,29,30]. For example, a meta-analysis of 12876 patients with solid tumors across 23 randomized controlled trials (RCTs) found that those receiving the PD-1 inhibitors nivolumab or pembrolizumab were associated with a significantly increased risk of developing any-grade and Grade ≥ 3 ILD compared with those receiving chemotherapy (risk ratio 5.17 [95% confidence interval (CI) 2.82, 9.47], p < 0.001, and 4.14 [95% CI 1.82, 9.42], p < 0.001, respectively) [31]. Similarly, another meta-analysis investigated the incidence of ILD among patients with various types of cancer receiving the CDK4/6i abemaciclib, palbociclib, or ribociclib across 12 Phase 2/3 clinical trials (N = 16060) [32]. The study found that in patients receiving CDK4/6i, the risks of developing any-grade ILD were doubled and the risk of developing Grade ≥ 3 ILD was tripled compared with controls (Peto odds ratio 2.12 [95% CI 1.57, 2.86], p < 0.00001, and 3.22 [95% CI 1.28, 8.09], p = 0.01, respectively) [32]. A meta-analysis of results from 12 RCTs investigated the association between patients with breast or gastric cancer treated with HER2-directed ADCs, including trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), and trastuzumab duocarmazine, and the risk of developing ILD compared with controls [33,34,35,36,37,38,39,40,41,42,43,44,45]. Among patients treated with HER2-directed ADCs, there was more than double the risk of developing both any-grade and Grade ≥ 3 ILD compared with controls receiving the standard-of-care therapies lapatinib plus chemotherapy, or trastuzumab alone or in combination with pertuzumab and/or chemotherapy (Peto odds ratio 2.62 [95% CI 1.71, 4.04], p < 0.0001, and 2.82 [95% CI 1.07, 7.42], p = 0.04, respectively) [33].

Although the risk of developing SACT-related ILD may vary between therapeutic agents, patients with male sex at birth, an Eastern Cooperative Oncology Group performance status (ECOG PS) > 1, and/or those with advanced age have been found to be at greater risk of developing SACT-related ILD when treated with various SACTs than their counterparts [6, 46,47,48]. Pre-existence of interstitial lung abnormalities (ILAs), defined as both fibrotic and non-fibrotic features on CT scans, and pre-existence of lung diseases may also be risk factors for patients receiving SACTs [47,48,49,50,51]. A retrospective, blinded, single-center cohort study found that in patients with non-small cell lung cancer receiving ICIs (N = 475), the risk of developing Grade ≥ 2 SACT-related ILD was doubled among those with pre-existing ILAs compared with their peers without ILAs (odds ratio 2.2 [95% CI 1.03, 4.50], p = 0.041) [51]. Several retrospective analyses investigating patients receiving TKIs or chemotherapeutic agents have demonstrated that those with pre-existing lung diseases, including concurrent or pre-existing chronic obstructive pulmonary disease, emphysema, and ILD, were at an increased risk of developing SACT-related ILD compared with their peers [47,48,49,50]. The overall incidence of SACT-related ILD in clinical trials ranges from 0.2% to 15.4%, depending on the treatment (Table 2).

Table 2 Incidence and mortality rates of ILD in patients treated with various SACTs in clinical trials

Detection of SACT-related ILD

Recommendations for monitoring patients receiving SACTs associated with a risk of developing ILD

The role of a baseline CT examination should go beyond oncological staging, enabling assessment of lung parenchyma before SACTs are introduced. In addition to treatment evaluation during follow-up CT scans, those receiving SACTs known to be associated with ILD should be screened for any potential signs of ILD. The incidental appearance of pulmonary pathology on CT scans should be investigated by a radiologist.

When ILD is suspected, a high-resolution computed tomography (HRCT) scan should be ordered [11, 15]. HRCT is more sensitive than standard CT scans and enables the extent of lung involvement to be assessed [7, 19]. Thin slices should be used (< 2-mm slice thickness), along with high-resolution reconstruction kernel, improving spatial resolution and allowing subtle reticular and nodular changes to be distinguished that would not be observable with a conventional chest CT scan [52, 53]. HRCT scans are routinely obtained at full inspiration; however, expiratory images may be helpful for determining the cause of a mosaic attenuation pattern [53]. The highest pitch and shortest rotation time feasible should be used to reduce the time for image acquisition and minimize the likelihood of movement introducing artifacts [54], especially as patients are often dyspneic and may not be able to hold their breath for long periods of time [55].

Once SACT-related ILD has been diagnosed based on radiological findings in combination with the results of other multidisciplinary team (MDT) testing, close monitoring using HRCT scans should continue until resolution of ILD, regardless of severity; this includes after drug discontinuation [14, 15]. Frequency of repeat HRCT scans should be individualized to each patient, depending on a multitude of clinical factors, including but not limited to the causative agent, pattern of ILD, grade of the event, and baseline risk factors. In general, close follow up is recommended for all cases of ILD, with repeat scans and clinical assessments within 1–2 weeks from diagnosis for low-grade cases (Grade ≤ 2), and within a few days of diagnosis for high-grade cases (Grade ≥ 3).

Presentation patterns of SACT-related ILD

The prognosis of SACT-related ILD varies not only by grade of the AE and the associated drug a patient receives, but may also depend on the radiological pattern [7]. Though there are no specific radiological features specific to ILD [19], patterns of presentation have been defined by the joint American Thoracic Society (ATS) and European Respiratory Society (ERS) guidelines [56]. The four main patterns that commonly present for SACT-related ILD include organizing pneumonia (OP), hypersensitivity pneumonitis (HP), acute interstitial pneumonia / acute respiratory distress syndrome (AIP/ARDS), and non-specific interstitial pneumonia (NSIP) [57].

Patterns with areas of high attenuation on HRCT

OP pattern

The OP pattern is characterized by sharply demarcated, bilateral, peripheral consolidations that may be migratory, fluctuating in position over time [56, 58] (Fig. 1A). The reversed halo, also known as the ‘atoll sign’, where a dense outer rim of consolidation is situated around a focal ground-glass opacity (GGO), may also be observed with this pattern [58] (Fig. 1B).

Fig. 1
figure 1

A CT scan of a patient with breast cancer and drug-induced ILD with OP pattern, characterized by sharply demarcated, bilateral, peripheral peribronchial consolidations (diagnosed by a multidisciplinary team [MDT] consensus); B CT scan of a patient with lung cancer and MDT-diagnosed drug-induced ILD with OP pattern and atoll sign in both under lobes. Images reproduced with permission from Dinkel J. 2023. University Hospital LMU Munich, Munich, Germany

Though radiological patterns for SACT-related ILD can be highly variable for the same drug [6], some ILD patterns may occur more frequently in patients being treated with a specific drug or drug class. A pooled analysis of 90 patients with advanced cancers and ILD associated with T-DXd across two Phase 1/2 single-arm, multicenter, global clinical trials found that 72.2% of cases (n = 65/90) had the OP pattern [59,60,61]. Similarly, a Japanese nationwide post-marketing surveillance program found that among 130 patients with HER2-positive metastatic breast cancer or gastric cancer and adjudicated T-DXd-related ILD, the majority of cases (63.1%) had the OP pattern [62]. The OP pattern was also the most common type observed in a retrospective multicenter study of patients with ILD related to treatment with ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA‑4) inhibitors, PD-1 inhibitors, and programmed cell death ligand 1 (PD-L1) inhibitors (23.4%, n = 15/64) [63].

HP pattern

The HP pattern has a distinctive phenotype on HRCT [64]. Extensive GGOs can be observed, with areas of high attenuation and air trapping (low attenuation) forming a characteristic mosaic pattern, also known as ‘head cheese sign’ or the ‘three-density pattern’ [64, 65] (Fig. 2). Normal, hypodense, and hyperdense areas of lung parenchyma coexist on the HRCT scan [65]. Fibrotic changes can occur with the drug-induced HP pattern, but are rare in the context of SACT-induced toxicity.

Fig. 2
figure 2

CT scan of a patient with systemic anticancer therapy-induced interstitial lung disease with the hypersensitivity pneumonitis pattern, diagnosed by multidisciplinary team consensus. The examinations were performed with contrast to exclude the presence of pulmonary embolism. Diagnosis of the three-density pattern is therefore difficult after injection. It is recommended that the examination be repeated without contrast agent if there is any uncertainty. This CT scan depicts mosaic density with extensive ground glass opacities and exclusion of several secondary lobules. Images reproduced with permission from Dinkel J. 2023. University Hospital LMU Munich, Munich, Germany

A retrospective analysis of patients with advanced cancer enrolled into Phase 1 clinical trials in a single center found that HP was the most commonly occurring pattern among those with ILD related to treatment with phosphoinositide 3-kinase / AKT serine / threonine kinase / mTOR inhibitors, accounting for ~45.0% of all cases; this was closely followed by the NSIP pattern [66].

AIP/ARDS pattern

HRCT findings for the AIP/ARDS pattern include extensive areas of diffuse or patchy consolidation and GGOs, often with a crazy-paving appearance where areas of high attenuation resemble irregularly shaped paving stones [56, 67] (Fig. 3). Formal diagnosis requires pathological examination, if available, confirming the presence of diffuse alveolar damage (DAD) [56, 68]—a histological hallmark of the AIP/ARDS pattern [56, 68]. However, patients typically present with such a serious clinical picture that the potential benefits of bronchoscopy sampling may not outweigh the risk of complications [69, 70]. The AIP/ARDS pattern is considered an aggressive form with a high mortality rate [69, 71], progressing rapidly over the course of days or weeks [56, 69]. A single-center real-world analysis of patients with SACT-related ILD requiring hospitalization (N = 120) found that the mortality rate was higher among patients with radiological patterns indicating DAD than among those with non-DAD patterns of drug-related ILD (53.3% vs. 13.3%, respectively); the presence of radiological patterns indicating DAD was associated with an increased risk of mortality compared with cases without DAD patterns (hazard ratio 11.24 [95% CI 4.82, 26.20], p < 0.01) [71].

Fig. 3
figure 3

A, B CT scans of a patient with drug-induced interstitial lung disease showing the acute interstitial pneumonia / acute respiratory distress syndrome pattern, including bilateral consolidations and pleural effusion (diagnosed by multidisciplinary team consensus)

Patterns with a reticulation HR pattern

NSIP pattern

The predominant findings for drug-induced ILD events with an NSIP pattern are usually symmetrical GGOs with peripheral, subpleural, and/or basal reticulation [56]. Secondly, the appearance of subtle traction bronchiectasis indicates a tendency towards fibrotic changes [72] (Fig. 4).

Fig. 4
figure 4

A, B CT scans of a patient with systemic anticancer therapy-induced interstitial lung disease showing the non-specific interstitial pneumonia pattern, diagnosed by multidisciplinary team consensus

Rare forms of SACT-related ILD

Sarcoid-like reaction

Certain SACTs—including the ICIs ipilimumab, nivolumab, and pembrolizumab—have been associated with a rare clinical manifestation known as a ‘sarcoid-like reaction’, which is characterized by non-necrotic granulomas (Fig. 5) [73,74,75]. These reactions are indistinguishable from sarcoidosis, and as the pathogenesis of sarcoidosis is not known, it is unclear whether SACTs cause sarcoidosis or simply simulate the syndrome [73].

Fig. 5
figure 5

CT scans of a patient with a head and neck tumor and a sarcoid-like reaction induced by systemic anticancer therapy (diagnosed by multidisciplinary team consensus). A, B After two cycles of pembrolizumab treatment, the patient developed new mediastinal lymphadenopathy and (C, D) a new pulmonary peri-lymphatic micronodular pattern. Sarcoidosis was found in a mediastinal lymph node sample and the case was finally classified by the interstitial lung disease board as a sarcoid-like reaction

Radiation recall pneumonitis

Radiation recall pneumonitis (RRP) is a rare reaction occurring in previously irradiated pulmonary tissue following exposure to ‘triggering agents’ (Fig. 6) [76], where areas of pneumonitis, characterized by consolidation or GGOs, are limited to the prior field(s) of radiation [77]. RRP has been observed following radiation and treatment with the ADC T-DXd, the anti-PD-1 drugs nivolumab and sintilimab, epidermal growth factor receptor (EGFR) TKIs, and various classes of chemotherapeutic agents [62, 78,79,80,81]. A retrospective study reviewed the medical records of patients with advanced non-small lung cancer who had received EGFR-TKIs within 5 years of radiotherapy and found that the RRP pattern was present in 6/20 (30.0%) patients who developed SACT-related ILD [78]. A Japanese nationwide post-marketing surveillance study found that, among 130 cases adjudicated as T-DXd-related ILD/pneumonitis in patients with breast and gastric cancer, 3/130 (2.3%) cases had ‘other’ imaging patterns, including RRP pattern, non-cardiogenic pulmonary edema pattern and cases where a definitive diagnosis of ILD/pneumonitis could not be made because of very light shadows [62].

Fig. 6
figure 6

A CT scan of a patient with left-central lung cancer treated with definitive radiochemotherapy. Due to a recurrence 2 years later in the form of mediastinal lymphadenopathy, the patient was treated with monoimmunotherapy and after four cycles the patient developed recall pneumonitis; B the diagnosis of which was established by consensus within a multidisciplinary team. Images reproduced with permission from Dinkel J. 2023. University Hospital LMU Munich, Munich, Germany

Differential diagnosis of SACT-related ILD

ILD is a diagnosis of exclusion; as radiological findings may be similar between SACT-related ILD and events secondary to other etiologies, they must be analyzed in combination with investigations from across the MDT [7, 15, 82]. Additionally, as not all ILD events in patients who are receiving SACT are drug-related, the diagnostic process should include identifying the cause of any ILD events detected, ensuring that the patient receives the most appropriate treatment.

Differential diagnoses of SACT-related ILD include the progression of an underlying disease, edema, other pulmonary disorders of unknown origin, and seasonal and/or opportunistic infections in immunocompromised patients [7, 17]. Interstitial pulmonary edema may also present with rales or crackles, and thus should be investigated when SACT-related ILD is suspected based on chest auscultation findings [83]. Infectious etiologies of interstitial pneumonia may be viral (caused by pathogens such as adenovirus, cytomegalovirus, Epstein-Barr virus, influenza, respiratory syncytial virus or severe acute respiratory syndrome coronavirus 2), bacterial (caused by Haemophilus, Streptococcus, or Pseudomonas species), or fungal (caused by organisms such as Pneumocystis jirovecii [PJ]) [84,85,86,87]. Microbial and serological tests, such as polymerase chain reaction assays, may help to exclude possible infectious etiologies when SACT-related ILD is suspected [7]. Joint clinical practice guidelines from the Latin American Thoracic Association, ATS, ERS, and Japanese Respiratory Society suggest that performing bronchoalveolar lavage (BAL) cellular analysis may help distinguish the etiology of suspected ILD events [88]. Pulmonary lymphangitis carcinomatosa can have a similar appearance to ILD on HRCT, with diffuse GGOs [56, 89, 90]. BAL and transbronchial biopsies can confirm the absence of adenocarcinoma cells [89]. Guidelines developed by the British Thoracic Society in collaboration with the Thoracic Society of Australia and New Zealand and the Irish Thoracic Society recommend excluding pulmonary embolism by CT pulmonary angiography when suspected ILD presents with respiratory failure [91]. In cases where there is uncertainty in confirming a diagnosis of ILD, HRCT imagery, test results, and patient details should be presented to the ILD board [92, 93].

The possibility for co-existing conditions should be considered when diagnosing SACT-related ILD. Although each potential differential diagnosis and co-existing condition has distinct diagnostic characteristics that aid in determining their likelihood, with CT imaging particularly valuable in this regard, ascertaining the primary limiting condition can be complex, especially when there is microbiological evidence of infection. For instance, the presence of PJ in the airways may not necessarily indicate Pneumocystis pneumonia (PJP), but simply colonization [94]. Usually, patients with PJP will present with a degree of hypoxia and respiratory distress [95], whereas SACT-induced ILD cases are typically mild in severity (Table 2). Although the co-existence of drug-induced ILD and PJP is rare [96], distinguishing between the two based solely on clinical presentation, imaging, and laboratory results can be difficult, particularly in severe cases [97, 98]. Where clear differentiation is challenging or time and resources are limited, it may be necessary to treat both conditions simultaneously. For example, it is not uncommon to administer steroids for SACT-related ILD and broad-spectrum antibiotics for suspected co-existing infections [94, 97]. Following the initiation of therapy, more detailed investigations typically guide definitive diagnosis.

SACT-related ILD may develop within days to months after drug administration; however, late clinical manifestations do occur and late-occurring ILD events should not be excluded as a possible diagnosis [18, 99].

Future directions for detection of ILD

The use of artificial intelligence (AI) to detect patterns of ILD is increasingly being investigated, with existing literature demonstrating the ability of AI models to make crude classifications of suspected ILD cases, such as determining from HRCT scans whether cases are fibrosing or non-fibrosing, and classifying the pattern of ILD as ‘definitely’, ‘possibly’, or ‘inconsistent with’ usual interstitial pneumonia [100]. Before AI algorithms can be integrated into the process of detecting ILD and determining patterns of disease, they must be validated by expert radiologists [100]. Additionally, existing models rely solely on the results from HRCT scans; the development of multivariate models that can consider patient characteristics and clinical findings from MDT assessments may be the next approach investigated in the AI space [100].

There also appears to be interest in the identification of biomarkers that may help clinicians detect ILD [101, 102]. A single-center cross-sectional study of patients with ILD (n = 322) found that eNose technology demonstrated promising accuracy in distinguishing these patients from healthy controls (n = 48) through breath analysis (area under the curve 1.00), and may be a useful tool to increase diagnostic confidence in combination with the results of other clinical assessments [102]. Additionally, the use of wearable biosensors for detection of pulmonary dysfunction in high-risk individuals has also been investigated [103], and this could be further assessed in patients at high-risk of developing SACT-ILD.

Therapeutic management of SACT-related ILD

Specific management guidelines are available for some SACTs that have an associated risk of lung injury, including post-marketing recommendations for the management of ILD events occurring during treatment with abemaciclib, everolimus, irinotecan, lapatinib, pembrolizumab, T-DM1, and T-DXd [12, 13, 26,27,28,29,30]. Corticosteroid administration is recommended for Grade ≥ 2 ILD events occurring during pembrolizumab treatment, with an initial dose of 1–2 mg/kg/day prednisolone or equivalent followed by taper [12]. Pembrolizumab should be withheld for Grade 2 events, with the option to resume in patients with complete resolution or partial resolution (to Grade 1) after corticosteroid taper; for recurrent Grade 2 ILD events and those Grade ≥ 3 in severity, pembrolizumab should be permanently discontinued [12]. A strategy for detecting, monitoring and managing T-DXd-related ILD, termed the five ‘S’ rules, has been developed, and includes screening (with careful patient selection prior to treatment initiation and regular clinical assessments to exclude signs/symptoms of ILD throughout treatment), scanning (with HRCT at baseline and repeated every 6–12 weeks), synergy (educating patients and the care team to facilitate early reporting of signs and symptoms, as well as multidisciplinary management once ILD is suspected), suspending treatment, and steroid treatment [101, 104]. On the diagnosis of a Grade 1 ILD event during treatment with T-DXd, the PI recommends a treatment delay, and rechallenge with a maintained dose if the event has completely resolved within 28 days of onset, or with the dose reduced one level if the event has resolved in greater than 28 days from onset [13]. In the investigators’ brochure for clinical trials, it is recommended that treatment with T-DXd be permanently discontinued if the Grade 1 ILD event has not resolved within 126 days of onset [105]. PI recommends that corticosteroid treatment should be considered as soon as ILD is suspected (e.g., ≥ 0.5 mg/kg/day prednisolone or equivalent) [13]. In the event of Grade ≥ 2 ILD, T-DXd treatment should be permanently discontinued and corticosteroid treatment promptly initiated (e.g., ≥ 1 mg/kg/day prednisolone or equivalent); this should be continued for at least 14 days followed by a gradual taper for at least 4 weeks [13]. Hospitalization is generally required for severe cases (Grade ≥ 3) [9]. Permanent discontinuation is recommended upon diagnosis of an ILD event of any grade related to treatment with irinotecan [26], and Grade ≥ 3 events related to treatment with lapatinib [27].

Conclusion

As the cancer treatment landscape evolves, it is likely that the incidence of potentially life-threatening SACT-related ILD events will increase. Collaborative working of MDTs is imperative for accurate and timely detection, diagnosis, and management of SACT-related ILD, with evaluations by the radiologist, primary physician, nurse practitioner, pulmonologist, thoracic surgeon, pathologist, and infectious disease specialist, where appropriate, necessary to rule out other causes [7, 15].

Radiologists are at the forefront of asymptomatic, Grade 1 ILD detection. Proactive communication between treating physicians and radiologists, noting when patients are receiving SACTs with a known risk for ILD on scan requisitions and highlighting when ILD is suspected, makes early detection more feasible. The correct scan methodology and urgent reporting to the patient’s treating physician and allied team are paramount. Given its high sensitivity and specificity, HRCT with < 2-mm slice thickness is the most appropriate CT technique for the investigation of suspected ILD. A stepwise approach to ILD detection, underpinned by an awareness of high-resolution ILD patterns, which can impact patient prognosis, is crucial for accurate diagnosis. Where there is uncertainty in confirming a diagnosis of ILD, the case should be presented to the ILD board. Patients with confirmed SACT-related ILD should be monitored until complete resolution of the AE.

Early detection of SACT-related ILD allows prompt implementation of drug-specific treatment management guidelines and recommendations. Detection and diagnosis at Grade 1, before symptom onset and prior to redosing, allows time for resolution of ILD and will likely reduce the risk of progression to higher-grade lung toxicity. Additionally, identification and resolution at Grade 1 allows patients to be rechallenged with some SACTs, as per specific post-marketing recommendations, widening patients’ eligibility for future cancer treatment options.

Abbreviations

ADC:

Antibody-drug conjugate

AIP/ARDS:

Acute interstitial pneumonia / acute respiratory distress syndrome

ATS:

American Thoracic Society

BAL:

Bronchoalveolar lavage

CDK4/6i:

Cyclin-dependent kinase 4/6 inhibitor(s)

CTCAE:

Common Terminology Criteria for Adverse Events

DAD:

Diffuse alveolar damage

EGFR:

Epidermal growth factor receptor

ERS:

European Respiratory Society

GGO:

Ground-glass opacity

HER2:

Human epidermal growth factor receptor 2

HP:

Hypersensitivity pneumonitis

HRCT:

High-resolution computed tomography

ICI:

Immune checkpoint inhibitor

ILA:

Interstitial lung abnormality

ILD:

Interstitial lung disease

MDT:

Multidisciplinary team

mTOR:

Mammalian target of rapamycin

NSIP:

Non-specific interstitial pneumonia

OP:

Organizing pneumonia

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed cell death ligand 1

PI:

Prescribing information

PJ:

Pneumocystis jirovecii

PJP:

Pneumocystis pneumonia

RRP:

Radiation recall pneumonitis

SACT:

Systemic anticancer therapy

T-DM1:

Trastuzumab emtansine

T-DXd:

Trastuzumab deruxtecan

TKI:

Tyrosine kinase inhibitor

References

  1. Antoine MH, Mlika M (2023) Interstitial lung disease. In: StatPearls. StatPearls Publishing, Treasure Island, FL, USA

  2. Schraufnagel DE (2010) Breathing in America: diseases, progress, and hope, 1st Edn. American Thoracic Society, New York, NY, USA

  3. Kreuter M, Herth FJ, Wacker M et al (2015) Exploring clinical and epidemiological characteristics of interstitial lung diseases: rationale, aims, and design of a nationwide prospective registry—the EXCITING-ILD registry. Biomed Res Int 2015:123876. https://doi.org/10.1155/2015/123876

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kaul B, Cottin V, Collard HR, Valenzuela C (2021) Variability in global prevalence of interstitial lung disease. Front Med Lausanne 8:751181. https://doi.org/10.3389/fmed.2021.751181

    Article  PubMed  PubMed Central  Google Scholar 

  5. Meyer KC (2014) Diagnosis and management of interstitial lung disease. Transl Respir Med 2:4. https://doi.org/10.1186/2213-0802-2-4

    Article  PubMed  PubMed Central  Google Scholar 

  6. Skeoch S, Weatherley N, Swift AJ et al (2018) Drug-induced interstitial lung disease: a systematic review. J Clin Med 7:356. https://doi.org/10.3390/jcm7100356

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Conte P, Ascierto PA, Patelli G et al (2022) Drug-induced interstitial lung disease during cancer therapies: expert opinion on diagnosis and treatment. ESMO Open 7:100404. https://doi.org/10.1016/j.esmoop.2022.100404

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Johkoh T, Lee KS, Nishino M et al (2021) 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. Radiology 298:550–566. https://doi.org/10.1148/radiol.2021203427

    Article  PubMed  Google Scholar 

  9. U.S. Department of Health and Human Services (HSS) (2017) Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_5x7.pdf. Accessed 17 Jul 2023

  10. Moran-Mendoza O, Ritchie T, Aldhaheri S (2021) Fine crackles on chest auscultation in the early diagnosis of idiopathic pulmonary fibrosis: a prospective cohort study. BMJ Open Respir Res 8:e000815. https://doi.org/10.1136/bmjresp-2020-000815

    Article  PubMed  PubMed Central  Google Scholar 

  11. Bardia A, Harnden K, Mauro L et al (2022) Clinical practices and institutional protocols on prophylaxis, monitoring, and management of selected adverse events associated with trastuzumab deruxtecan. Oncologist 27:637–645. https://doi.org/10.1093/oncolo/oyac107

    Article  PubMed  PubMed Central  Google Scholar 

  12. U.S. Food & Drug Administration (FDA) (2024) KEYTRUDA (pembrolizumab): highlights of prescribing information (PI). https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/125514s124lbl.pdf. Accessed 15 Feb 2024

  13. U.S. Food and Drug Administration (FDA) (2024) ENHERTU (fam-trastuzumab deruxtecan-nxki): highlights of prescribing information. www.accessdata.fda.gov/drugsatfda_docs/label/2024/761139s028lbl.pdf. Accessed 19 Apr 2024

  14. Rugo HS, Bianchini G, Cortes J et al (2022) Optimizing treatment management of trastuzumab deruxtecan in clinical practice of breast cancer. ESMO Open 7:100553. https://doi.org/10.1016/j.esmoop.2022.100553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Swain SM, Nishino M, Lancaster LH et al (2022) Multidisciplinary clinical guidance on trastuzumab deruxtecan (T-DXd)-related interstitial lung disease/pneumonitis – focus on proactive monitoring, diagnosis, and management. Cancer Treat Rev 106:102378. https://doi.org/10.1016/j.ctrv.2022.102378

    Article  CAS  PubMed  Google Scholar 

  16. Tarantino P, Modi S, Tolaney SM et al (2021) Interstitial lung disease induced by anti-ERBB2 antibody-drug conjugates: a review. JAMA Oncol 7:1873–1881. https://doi.org/10.1001/jamaoncol.2021.3595

    Article  PubMed  Google Scholar 

  17. Kubo K, Azuma A, Kanazawa M et al (2013) Consensus statement for the diagnosis and treatment of drug-induced lung injuries. Respir Investig 51:260–277. https://doi.org/10.1016/j.resinv.2013.09.001

    Article  PubMed  Google Scholar 

  18. Yonemori K, Hirakawa A, Kawachi A et al (2016) Drug induced interstitial lung disease in oncology phase I trials. Cancer Sci 107:1830–1836. https://doi.org/10.1111/cas.13087

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Matsuno O (2012) Drug-induced interstitial lung disease: mechanisms and best diagnostic approaches. Respir Res 13:39. https://doi.org/10.1186/1465-9921-13-39

    Article  PubMed  PubMed Central  Google Scholar 

  20. Schwaiblmair M, Behr W, Haeckel T et al (2012) Drug induced interstitial lung disease. Open Respir Med J 6:63–74. https://doi.org/10.2174/1874306401206010063

    Article  PubMed  PubMed Central  Google Scholar 

  21. Yan S, Li M, Liu B et al (2023) Neutrophil extracellular traps and pulmonary fibrosis: an update. J Inflamm (Lond) 20:2. https://doi.org/10.1186/s12950-023-00329-y

    Article  CAS  PubMed  Google Scholar 

  22. Kumagai K, Aida T, Tsuchiya Y et al (2020) Interstitial pneumonitis related to trastuzumab deruxtecan, a human epidermal growth factor receptor 2-targeting Ab-drug conjugate, in monkeys. Cancer Sci 111:4636–4645. https://doi.org/10.1111/cas.14686

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. de Goeij BE, Lambert JM (2016) New developments for antibody-drug conjugate-based therapeutic approaches. Curr Opin Immunol 40:14–23. https://doi.org/10.1016/j.coi.2016.02.008

    Article  CAS  PubMed  Google Scholar 

  24. Tarantino P, Ricciuti B, Pradhan SM, Tolaney SM (2023) Optimizing the safety of antibody-drug conjugates for patients with solid tumours. Nat Rev Clin Oncol 20:558–576. https://doi.org/10.1038/s41571-023-00783-w

    Article  CAS  PubMed  Google Scholar 

  25. Sakamoto E, Katahira Y, Mizoguchi I et al (2023) Chemical- and drug-induced allergic, inflammatory, and autoimmune diseases via haptenation. Biol Basel 12:123. https://doi.org/10.3390/biology12010123

    Article  CAS  Google Scholar 

  26. U.S. Food & Drug Administration (FDA) (2017) ONIVYDE (irinotecan liposome injection): highlights of prescribing information (PI). https://www.ipsen.com/websites/Ipsen_Online/wp-content/uploads/sites/9/2019/01/21083350/ONIVYDE_USPI.pdf. Accessed 26 Jul 2023

  27. U.S. Food & Drug Administration (FDA) (2022) TYKERB (lapatinib): highlights of prescribing information (PI). https://www.novartis.com/us-en/sites/novartis_us/files/tykerb.pdf. Accessed 24 Jul 2023

  28. European Medicines Agency (EMA) AFINITOR: summary of product characteristics. https://www.ema.europa.eu/en/documents/product-information/afinitor-epar-product-information_en.pdf. Accessed 25 Aug 2023

  29. European Medicines Agency (EMA) VERZENIOS: summary of product characteristics. https://www.ema.europa.eu/en/documents/product-information/verzenios-epar-product-information_en.pdf. Accessed 25 Aug 2023

  30. European Medicines Agency (EMA) (2023) KADCYLA: summary of product characteristics. https://www.ema.europa.eu/en/documents/product-information/kadcyla-epar-product-information_en.pdf. Accessed 25 Aug 2023

  31. Su Q, Zhu EC, Wu J-B et al (2019) Risk of pneumonitis and pneumonia associated with immune checkpoint inhibitors for solid tumors: a systematic review and meta-analysis. Front Immunol 10:108. https://doi.org/10.3389/fimmu.2019.00108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang Y, Ma Z, Sun X et al (2022) Interstitial lung disease in patients treated with cyclin-dependent kinase 4/6 inhibitors: a systematic review and meta-analysis of randomized controlled trials. Breast 62:162–169. https://doi.org/10.1016/j.breast.2022.02.011

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ma Z, Zhang Y, Zhu M et al (2022) Interstitial lung disease associated with anti-HER2 anti-body drug conjugates: results from clinical trials and the WHO’s pharmacovigilance database. Expert Rev Clin Pharmacol 15:1351–1361. https://doi.org/10.1080/17512433.2022.2121705

    Article  CAS  PubMed  Google Scholar 

  34. ClinicalTrials.gov (2016) NCT00829166. A study of trastuzumab emtansine versus capecitabine + lapatinib in participants with HER2-positive locally advanced or metastatic breast cancer (EMILIA). Updated. https://clinicaltrials.gov/study/NCT00829166. Accessed 2 Oct 2023

  35. ClinicalTrials.gov (2024) NCT01772472. A study of trastuzumab emtansine versus trastuzumab as adjuvant therapy in patients with HER2-positive breast cancer who have residual tumor in the breast or axillary lymph nodes following preoperative therapy (KATHERINE). Updated. https://clinicaltrials.gov/study/NCT01772472. Accessed 2 Oct 2023

  36. ClinicalTrials.gov (2017) NCT01641939. A study of trastuzumab emtansine versus taxane in participants with human epidermal growth factor receptor 2 (HER2)-positive advanced gastric cancer. Updated. https://clinicaltrials.gov/study/NCT01641939. Accessed 2 Oct 2023

  37. ClinicalTrials.gov (2017) NCT02144012. A study to evaluate the efficacy and safety of trastuzumab emtansine versus the combination of trastuzumab plus docetaxel in patients with HER2-positive breast cancer. Updated. https://clinicaltrials.gov/study/NCT02144012. Accessed 2 Oct 2023

  38. ClinicalTrials.gov (2016) NCT01419197. A study of trastuzumab emtansine in comparison with treatment of physician’s choice in participants with HER2-positive breast cancer who have received at least two prior regimens of HER2-directed therapy (TH3RESA). Updated. https://clinicaltrials.gov/study/NCT01419197. Accessed 2 Oct 2023

  39. ClinicalTrials.gov (2023) NCT01853748. T-DM1 vs paclitaxel/trastuzumab for breast (ATEMPT trial). Updated. https://clinicaltrials.gov/study/NCT01853748. Accessed 2 Oct 2023

  40. ClinicalTrials.gov (2022) NCT03329690. DS-8201a in human epidermal growth factor receptor 2 (HER2)-expressing gastric cancer [DESTINY-Gastric01]. Updated. https://clinicaltrials.gov/study/NCT03329690. Accessed 2 Oct 2023

  41. ClinicalTrials.gov (2023) NCT03262935. SYD985 vs. physician’s choice in participants with HER2-positive locally advanced or metastatic breast cancer (TULIP). Updated. https://clinicaltrials.gov/study/NCT03262935. Accessed 2 Oct 2023

  42. ClinicalTrials.gov (2014) NCT00679341. A study of the efficacy and safety of trastuzumab emtansine (trastuzumab-MCC-DM1) vs. trastuzumab (Herceptin®) and docetaxel (Taxotere®) in patients with metastatic HER2-positive breast cancer who have not received prior chemotherapy for metastatic disease. Updated. https://clinicaltrials.gov/study/NCT00679341. Accessed 2 Oct 2023

  43. ClinicalTrials.gov (2017) NCT01120184. A study of trastuzumab emtansine (T-DM1) plus pertuzumab/pertuzumab placebo versus trastuzumab [Herceptin] plus a taxane in participants with metastatic breast cancer (MARIANNE). Updated. https://clinicaltrials.gov/study/NCT01120184. Accessed 2 Oct 2023

  44. ClinicalTrials.gov (2022) NCT01966471. A study of trastuzumab emtansine (Kadcyla) plus pertuzumab (Perjeta) following anthracyclines in comparison with trastuzumab (Herceptin) plus pertuzumab and a taxane following anthracyclines as adjuvant therapy in participants with operable HER2-positive primary breast cancer. Updated. https://clinicaltrials.gov/study/NCT01966471. Accessed 2 Oct 2023

  45. ClinicalTrials.gov (2019) NCT02131064. A study evaluating trastuzumab emtansine plus pertuzumab compared with chemotherapy plus trastuzumab and pertuzumab for participants with human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Updated. https://clinicaltrials.gov/study/NCT02131064. Accessed 2 Oct 2023

  46. Osawa M, Kudoh S, Sakai F et al (2015) Clinical features and risk factors of panitumumab-induced interstitial lung disease: a postmarketing all-case surveillance study. Int J Clin Oncol 20:1063–1071. https://doi.org/10.1007/s10147-015-0834-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sakurada T, Kakiuchi S, Tajima S et al (2015) Characteristics of and risk factors for interstitial lung disease induced by chemotherapy for lung cancer. Ann Pharmacother 49:398–404. https://doi.org/10.1177/1060028014566446

    Article  CAS  PubMed  Google Scholar 

  48. Tomii K, Kato T, Takahashi M et al (2017) Pemetrexed-related interstitial lung disease reported from post marketing surveillance (malignant pleural mesothelioma/non-small cell lung cancer). Jpn J Clin Oncol 47:350–356. https://doi.org/10.1093/jjco/hyx010

    Article  PubMed  Google Scholar 

  49. Gemma A, Kudoh S, Ando M et al (2014) Final safety and efficacy of erlotinib in the phase 4 POLARSTAR surveillance study of 10 708 Japanese patients with non-small-cell lung cancer. Cancer Sci 105:1584–1590. https://doi.org/10.1111/cas.12550

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Johkoh T, Sakai F, Kusumoto M et al (2014) Association between baseline pulmonary status and interstitial lung disease in patients with non-small-cell lung cancer treated with erlotinib-a cohort study. Clin Lung Cancer 15:448–454. https://doi.org/10.1016/j.cllc.2014.06.003

    Article  CAS  PubMed  Google Scholar 

  51. Petranovic M, McDermott S, Mercaldo S et al (2023) Impact of baseline interstitial lung abnormalities on pneumonitis risk in patients receiving immune checkpoint inhibitors for non-small-cell lung cancer. Clin Lung Cancer 24:682–688.e5. https://doi.org/10.1016/j.cllc.2023.08.014

    Article  CAS  PubMed  Google Scholar 

  52. Canadian Society of Thoracic Radiology (CSTR) (2020) High resolution CT of the chest recommended technique. https://car.ca/wp-content/uploads/2020/02/High-Resolution-CT-of-the-Chest-Recommended-Technique-2020.pdf. Accessed 18 Jul 2023

  53. Kazerooni EA (2001) High-resolution CT of the lungs. AJR Am J Roentgenol 177:501–519. https://doi.org/10.2214/ajr.177.3.1770501

    Article  CAS  Google Scholar 

  54. Torres PP, Rabahi MF, Moreira MA et al (2021) Importance of chest HRCT in the diagnostic evaluation of fibrosing interstitial lung diseases. J Bras Pneumol 47:e20200096. https://doi.org/10.36416/1806-3756/e20200096

    Article  PubMed  PubMed Central  Google Scholar 

  55. Studler U, Gluecker T, Bongartz G et al (2005) Image quality from high-resolution CT of the lung: comparison of axial scans and of sections reconstructed from volumetric data acquired using MDCT. AJR Am J Roentgenol 185:602–607. https://doi.org/10.2214/ajr.185.3.01850602

    Article  Google Scholar 

  56. American Thoracic Society, European Respiratory Society (2002) American Thoracic Society/European Respiratory Society international multidisciplinary consensus classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med 165:277–304. https://doi.org/10.1164/ajrccm.165.2.ats01

    Article  Google Scholar 

  57. Nishino M, Hatabu H, Hodi FS, Ramaiya NH (2017) Drug-related pneumonitis in the era of precision cancer therapy. JCO Precis Oncol 1:PO.17.00026. https://doi.org/10.1200/PO.17.00026

  58. Chandra D, Maini R, Hershberger DM (2023) Cryptogenic organizing pneumonia. In: StatPearls. StatPearls Publishing, Treasure Island, FL, USA

  59. Modi S, Saura C, Yamashita T et al (2020) Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med 382:610–621. https://doi.org/10.1056/NEJMoa1914510

    Article  CAS  PubMed  Google Scholar 

  60. Tamura K, Tsurutani J, Takahashi S et al (2019) Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive breast cancer previously treated with trastuzumab emtansine: a dose-expansion, phase 1 study. Lancet Oncol 20:816–826. https://doi.org/10.1016/S1470-2045(19)30097-X

    Article  CAS  PubMed  Google Scholar 

  61. Nishino M, Kusumoto M, Bankier AA et al (2023) Trastuzumab deruxtecan‒related interstitial lung disease/pneumonitis: computed tomography imaging patterns to guide diagnosis and management. JCO Precis Oncol 7:e2300391. https://doi.org/10.1200/PO.23.00391

    Article  PubMed  Google Scholar 

  62. Baba T, Kusumoto M, Kato T et al (2023) Clinical and imaging features of interstitial lung disease in cancer patients treated with trastuzumab deruxtecan. Int J Clin Oncol 28:1585–1596. https://doi.org/10.1007/s10147-023-02414-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Delaunay M, Cadranel J, Lusque A et al (2017) Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur Respir J 50:1700050. https://doi.org/10.1183/13993003.00050-2017

    Article  CAS  PubMed  Google Scholar 

  64. Chandra D, Cherian SV (2023) Hypersensitivity pneumonitis. In: StatPearls. StatPearls Publishing, Treasure Island, FL, USA

  65. Hamblin M, Prosch H, Vašáková M (2022) Diagnosis, course and management of hypersensitivity pneumonitis. Eur Respir Rev 31:210169. https://doi.org/10.1183/16000617.0169-2021

    Article  PubMed  PubMed Central  Google Scholar 

  66. Terbuch A, Tiu C, Candilejo IM et al (2020) Radiological patterns of drug-induced interstitial lung disease (DILD) in early-phase oncology clinical trials. Clin Cancer Res 26:4805–4813. https://doi.org/10.1158/1078-0432.CCR-20-0454

    Article  CAS  PubMed  Google Scholar 

  67. De Wever W, Meersschaert J, Coolen J et al (2011) The crazy-paving pattern: a radiological-pathological correlation. Insights Imaging 2:117–132. https://doi.org/10.1007/s13244-010-0060-5

    Article  PubMed  PubMed Central  Google Scholar 

  68. Mukhopadhyay S, Parambil JG (2012) Acute interstitial pneumonia (AIP): relationship to Hamman-Rich syndrome, diffuse alveolar damage (DAD), and acute respiratory distress syndrome (ARDS). Semin Respir Crit Care Med 33:476–485. https://doi.org/10.1055/s-0032-1325158

    Article  PubMed  Google Scholar 

  69. American Thoracic Society, European Respiratory Society (2000) Idiopathic pulmonary fibrosis: diagnosis and treatment. International consensus statement. Am J Respir Crit Care Med 161:646–664. https://doi.org/10.1164/ajrccm.161.2.ats3-00

    Article  Google Scholar 

  70. Du Rand IA, Blaikley J, Booton R et al (2013) British Thoracic Society guideline for diagnostic flexible bronchoscopy in adults: accredited by NICE. Thorax 68:i1–i44. https://doi.org/10.1136/thoraxjnl-2013-203618

    Article  PubMed  Google Scholar 

  71. Kaku S, Horinouchi H, Watanabe H et al (2022) 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 148:1737–1746. https://doi.org/10.1007/s00432-022-03932-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jain A, Shannon VR, Sheshadri A (2018) Pneumonitis after precision oncology therapies: a concise review. J Immunother Precis Oncol 1:26–37. https://doi.org/10.4103/JIPO.JIPO_9_18

    Article  Google Scholar 

  73. Gkiozos I, Kopitopoulou A, Kalkanis A et al (2018) Sarcoidosis-like reactions induced by checkpoint inhibitors. J Thorac Oncol 13:1076–1082. https://doi.org/10.1016/j.jtho.2018.04.031

    Article  CAS  PubMed  Google Scholar 

  74. Bokhari SR, Zulfiqar H, Mansur A (2023) Sarcoidosis. In: StatPearls. StatPearls Publishing, Treasure Island, FL, USA

  75. Tan QL, Leow LC, Mohideen SM, Sewa DW (2019) Sarcoid-like reaction associated with lung adenocarcinoma: a case report. Proc Singap Heal 28:68–70. https://doi.org/10.1177/2010105818766363

    Article  Google Scholar 

  76. Jan P-R, Chang JW-C, Wu C-E (2022) Radiation recall pneumonitis: a rare syndrome that should be recognized. Cancers Basel 14:4642. https://doi.org/10.3390/cancers14194642

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kalisz KR, Ramaiya NH, Laukamp KR, Gupta A (2019) Immune checkpoint inhibitor therapy-related pneumonitis: patterns and management. Radiographics 39:1923–1937. https://doi.org/10.1148/rg.2019190036

    Article  PubMed  Google Scholar 

  78. Chiang C-L, Chen Y-W, Wu M-H et al (2016) Radiation recall pneumonitis induced by epidermal growth factor receptor-tyrosine kinase inhibitor in patients with advanced non-small cell lung cancer. J Chin Med Assoc 79:248–255. https://doi.org/10.1016/j.jcma.2016.01.008

    Article  PubMed  Google Scholar 

  79. Wang M, Xu S, Zhu H (2022) Radiation recall pneumonitis induced by sintilimab: a case report and literature review. Front Immunol 13:823767. https://doi.org/10.3389/fimmu.2022.823767

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Shibaki R, Akamatsu H, Fujimoto M et al (2017) Nivolumab induced radiation recall pneumonitis after two years of radiotherapy. Ann Oncol 28:1404–1405. https://doi.org/10.1093/annonc/mdx115

    Article  CAS  PubMed  Google Scholar 

  81. Ding X, Ji W, Li J et al (2011) Radiation recall pneumonitis induced by chemotherapy after thoracic radiotherapy for lung cancer. Radiat Oncol 6:24. https://doi.org/10.1186/1748-717X-6-24

    Article  PubMed  PubMed Central  Google Scholar 

  82. Suzuki T, Tada Y, Tsushima K et al (2013) Pneumocystis pneumonia in everolimus therapy: an indistinguishable case from drug induced interstitial lung disease. Respir Med Case Rep 10:27–30. https://doi.org/10.1016/j.rmcr.2013.07.003

    Article  PubMed  PubMed Central  Google Scholar 

  83. Pirrotta F, Mazza B, Gennari L, Palazzuoli A (2021) Pulmonary congestion assessment in heart failure: traditional and new tools. Diagn Basel 11:1306. https://doi.org/10.3390/diagnostics11081306

    Article  Google Scholar 

  84. Azadeh N, Limper AH, Carmona EM, Ryu JH (2017) The role of infection in interstitial lung diseases: a review. Chest 152:842–852. https://doi.org/10.1016/j.chest.2017.03.033

    Article  PubMed  Google Scholar 

  85. Richter AG, Stockley RA, Harper L, Thickett DR (2009) Pulmonary infection in Wegener granulomatosis and idiopathic pulmonary fibrosis. Thorax 64:692–697. https://doi.org/10.1136/thx.2008.110445

    Article  CAS  PubMed  Google Scholar 

  86. Mehta P, Rosas IO, Singer M (2022) Understanding post-COVID-19 interstitial lung disease (ILD): a new fibroinflammatory disease entity. Intensive Care Med 48:1803–1806. https://doi.org/10.1007/s00134-022-06877-w

    Article  PubMed  PubMed Central  Google Scholar 

  87. Ushiki A, Yamazaki Y, Hama M et al (2014) Viral infections in patients with an acute exacerbation of idiopathic interstitial pneumonia. Respir Investig 52:65–70. https://doi.org/10.1016/j.resinv.2013.07.005

    Article  PubMed  Google Scholar 

  88. Raghu G, Remy-Jardin M, Myers JL et al (2018) Diagnosis of idiopathic pulmonary fibrosis. An official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med 198:e44–e68. https://doi.org/10.1164/rccm.201807-1255ST

    Article  PubMed  Google Scholar 

  89. Imakita T, Fujita K, Kanai O, Mio T (2022) Pulmonary lymphangitis carcinomatosa mimicking immunotherapy-related interstitial pneumonitis: a case report. Case Rep Oncol 15:732–737. https://doi.org/10.1159/000525800

    Article  PubMed  PubMed Central  Google Scholar 

  90. Moubax K, Wuyts W, Vandecaveye V, Prenen H (2012) Pulmonary lymphangitic carcinomatosis as a primary manifestation of gastric carcinoma in a young adult: a case report and review of the literature. BMC Res Notes 5:638. https://doi.org/10.1186/1756-0500-5-638

    Article  PubMed  PubMed Central  Google Scholar 

  91. Bradley B, Branley HM, Egan JJ et al (2008) Interstitial lung disease guideline: the British Thoracic Society in collaboration with the Thoracic Society of Australia and New Zealand and the Irish Thoracic Society. Thorax 63:v1–v58. https://doi.org/10.1136/thx.2008.101691

    Article  PubMed  Google Scholar 

  92. Guler SA, Berezowska SA, Christe A et al (2016) Multidisciplinary discussion for diagnosis of interstitial lung disease in real life. Swiss Med Wkly 146:w14318. https://doi.org/10.4414/smw.2016.14318

    Article  PubMed  Google Scholar 

  93. Lynch DA, Sverzellati N, Travis WD et al (2018) Diagnostic criteria for idiopathic pulmonary fibrosis: a Fleischner Society White Paper. Lancet Respir Med 6:138–153. https://doi.org/10.1016/S2213-2600(17)30433-2

    Article  PubMed  Google Scholar 

  94. Vidal S, de la Horra C, Martín J et al (2006) Pneumocystis jirovecii colonisation in patients with interstitial lung disease. Clin Microbiol Infect 12:231–235. https://doi.org/10.1111/j.1469-0691.2005.01337.x

    Article  CAS  PubMed  Google Scholar 

  95. Truong J, Ashurst JV (2023) Pneumocystis jirovecii pneumonia. In: StatPearls. StatPearls Publishing, Treasure Island, FL, USA

  96. Sun Y, Shao C, Huang H et al (2022) Prognostic analysis of Pneumocystis jirovecii pneumonia in interstitial lung disease patients: a retrospective clinical study. Diagn Basel 12:2925. https://doi.org/10.3390/diagnostics12122925

    Article  CAS  Google Scholar 

  97. Saito Y, Nagayama M, Miura Y et al (2013) A case of pneumocystis pneumonia associated with everolimus therapy for renal cell carcinoma. Jpn J Clin Oncol 43:559–562. https://doi.org/10.1093/jjco/hyt019

    Article  PubMed  Google Scholar 

  98. Oikonomou A, Prassopoulos P (2012) Mimics in chest disease: interstitial opacities. Insights Imaging 4:9–27. https://doi.org/10.1007/s13244-012-0207-7

    Article  PubMed  PubMed Central  Google Scholar 

  99. Powell CA, Modi S, Iwata H et al (2022) Pooled analysis of drug-related interstitial lung disease and/or pneumonitis in nine trastuzumab deruxtecan monotherapy studies. ESMO Open 7:100554. https://doi.org/10.1016/j.esmoop.2022.100554

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Dack E, Christe A, Fontanellaz M et al (2023) Artificial intelligence and interstitial lung disease: diagnosis and prognosis. Invest Radiol 58:602–609. https://doi.org/10.1097/RLI.0000000000000974

    Article  PubMed  PubMed Central  Google Scholar 

  101. Wekking D, Porcu M, Pellegrino B et al (2023) Multidisciplinary clinical guidelines in proactive monitoring, early diagnosis, and effective management of trastuzumab deruxtecan (T-DXd)-induced interstitial lung disease (ILD) in breast cancer patients. ESMO Open 8:102043. https://doi.org/10.1016/j.esmoop.2023.102043

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Moor CC, Oppenheimer JC, Nakshbandi G et al (2021) Exhaled breath analysis by use of eNose technology: a novel diagnostic tool for interstitial lung disease. Eur Respir J 57:2002042. https://doi.org/10.1183/13993003.02042-2020

    Article  PubMed  Google Scholar 

  103. Shahrestani S, Chou T-C, Shang K-M et al (2021) A wearable eddy current based pulmonary function sensor for continuous non-contact point-of-care monitoring during the COVID-19 pandemic. Sci Rep 11:20144. https://doi.org/10.1038/s41598-021-99682-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Tarantino P, Tolaney SM (2023) Detecting and managing T-DXd-related interstitial lung disease: the five “S” rules. JCO Oncol Pr 19:526–527. https://doi.org/10.1200/OP.23.00097

    Article  Google Scholar 

  105. AstraZeneca, Daiichi Sankyo (2023) Trastuzumab deruxtecan (T-DXd) investigator’s brochure

  106. Diéras V, Harbeck N, Budd GT et al (2014) Trastuzumab emtansine in human epidermal growth factor receptor 2-positive metastatic breast cancer: an integrated safety analysis. J Clin Oncol 32:2750–2757. https://doi.org/10.1200/JCO.2013.54.4999

    Article  CAS  PubMed  Google Scholar 

  107. Xu B-H, Jiang Z-F, Chua D et al (2011) Lapatinib plus capecitabine in treating HER2-positive advanced breast cancer: efficacy, safety, and biomarker results from Chinese patients. Chin J Cancer 30:327–335. https://doi.org/10.5732/cjc.010.10507

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Capri G, Chang J, Chen S-C et al (2010) An open-label expanded access study of lapatinib and capecitabine in patients with HER2-overexpressing locally advanced or metastatic breast cancer. Ann Oncol 21:474–480. https://doi.org/10.1093/annonc/mdp373

    Article  CAS  PubMed  Google Scholar 

  109. Blackwell KL, Pegram MD, Tan-Chiu E et al (2009) Single-agent lapatinib for HER2-overexpressing advanced or metastatic breast cancer that progressed on first- or second-line trastuzumab-containing regimens. Ann Oncol 20:1026–1031. https://doi.org/10.1093/annonc/mdn759

    Article  CAS  PubMed  Google Scholar 

  110. Tolaney SM, Wardley AM, Zambelli S et al (2020) Abemaciclib plus trastuzumab with or without fulvestrant versus trastuzumab plus standard-of-care chemotherapy in women with hormone receptor-positive, HER2-positive advanced breast cancer (monarcHER): a randomised, open-label, phase 2 trial. Lancet Oncol 21:763–775. https://doi.org/10.1016/S1470-2045(20)30112-1

    Article  CAS  PubMed  Google Scholar 

  111. Johnston SR, Harbeck N, Hegg R et al (2020) Abemaciclib combined with endocrine therapy for the adjuvant treatment of HR+, HER2-, node-positive, high-risk, early breast cancer (monarchE). J Clin Oncol 38:3987–3998. https://doi.org/10.1200/JCO.20.02514

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Mayer EL, Dueck AC, Martin M et al (2021) Palbociclib with adjuvant endocrine therapy in early breast cancer (PALLAS): interim analysis of a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 22:212–222. https://doi.org/10.1016/S1470-2045(20)30642-2

    Article  CAS  PubMed  Google Scholar 

  113. Finn RS, Rugo HS, Gelmon KA et al (2021) Long‐term pooled safety analysis of palbociclib in combination with endocrine therapy for hormone receptor‐positive/human epidermal growth factor receptor 2‐negative advanced breast cancer: updated analysis with up to 5 years of follow‐up. Oncologist 26:e749–e755. https://doi.org/10.1002/onco.13684

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Slamon DJ, Neven P, Chia S et al (2020) Overall survival with ribociclib plus fulvestrant in advanced breast cancer. N Engl J Med 382:514–524. https://doi.org/10.1056/NEJMoa1911149

    Article  CAS  PubMed  Google Scholar 

  115. Im S-A, Lu Y-S, Bardia A et al (2019) Overall survival with ribociclib plus endocrine therapy in breast cancer. N Engl J Med 381:307–316. https://doi.org/10.1056/NEJMoa1903765

    Article  CAS  PubMed  Google Scholar 

  116. Wolchok JD, Chiarion-Sileni V, Gonzalez R et al (2017) Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 377:1345–1356. https://doi.org/10.1056/NEJMoa1709684

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Antonia SJ, López-Martin JA, Bendell J et al (2016) Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 17:883–895. https://doi.org/10.1016/S1470-2045(16)30098-5

    Article  CAS  PubMed  Google Scholar 

  118. Hodi FS, Chesney J, Pavlick AC et al (2016) Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol 17:1558–1568. https://doi.org/10.1016/S1470-2045(16)30366-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Robert C, Schachter J, Long GV et al (2015) Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 372:2521–2532. https://doi.org/10.1056/NEJMoa1503093

    Article  CAS  PubMed  Google Scholar 

  120. Socinski MA, Jotte RM, Cappuzzo F et al (2023) Association of immune-related adverse events with efficacy of atezolizumab in patients with non–small cell lung cancer. JAMA Oncol 9:527–535. https://doi.org/10.1001/jamaoncol.2022.7711

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The medical writing support was provided by Francesca Codrington, BSc, Helios Medical Communications, Cheshire, UK.

Funding

Medical writing support was provided by Helios Medical Communications, Cheshire, UK, and funded by AstraZeneca in accordance with Good Publication Practice guidelines (http://www.ismpp.org/gpp-2022). The manuscript was reviewed for medical accuracy by AstraZeneca and Daiichi Sankyo; however, the authors retained full control of the content and made the final decisions for all aspects of this article.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to drafting and critically reviewing the manuscript, as well as approving the final version.

Corresponding author

Correspondence to Julien Dinkel.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

University Hospital LMU Munich guidelines do not require individual consent for publication of images, as long as they are anonymized.

Competing interests

J.D.: Speaker and/or consulting fees from AstraZeneca, Boehringer Ingelheim and Daiichi Sankyo, and participation in the ILD advisory committee for the DESTINY-Breast12 study for AstraZeneca and Daiichi Sankyo. N.K.: Speaker and/or consulting fees from AstraZeneca and Daiichi Sankyo, and participation in the ILD advisory committee for the DESTINY-Breast12 study for AstraZeneca and Daiichi Sankyo. P.T.: Research funds (to institution) and support attending meetings from AstraZeneca, and speaker and/or consulting fees from AstraZeneca, Daiichi Sankyo, Eli Lilly and Company, Genentech, Gilead, Novartis, and Roche.

Additional information

Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dinkel, J., Kneidinger, N. & Tarantino, P. The radiologist’s role in detecting systemic anticancer therapy-related interstitial lung disease: an educational review. Insights Imaging 15, 191 (2024). https://doi.org/10.1186/s13244-024-01771-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13244-024-01771-z

Keywords