Journal

Vol. 28 No. 2, 2025

Table of Contents

ORIGINAL ARTICLES

Thirteen Years’ Experience of Stereotactic Body Radiation Therapy for Ultra-Central Lung Tumours in Hong Kong

   CME

JKW Ng, MTY Kam, KKS Wong, JQ Du, ECY Wong, RMW Yeung

ORIGINAL ARTICLE    CME
 
Thirteen Years’ Experience of Stereotactic Body Radiation Therapy for Ultra-Central Lung Tumours in Hong Kong
 
JKW Ng, MTY Kam, KKS Wong, JQ Du, ECY Wong, RMW Yeung
Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China
 
Correspondence: Dr KW Ng, Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China. Email: njk540@ha.org.hk
 
Submitted: 14 August 2024; Accepted: 6 December 2024.
 
Contributors: JKWN and MTYK designed the study. JKWN acquired and analysed the data, and drafted the manuscript. All authors critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: This research was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: CIRB-2024-319-5). The requirement of patient consent was waived by the Board due to the retrospective nature of the research.
 
 
 
 
 
Abstract
 
Introduction
 
Stereotactic body radiation therapy (SBRT) for ultra-central lung tumours is controversial given the proximity of the tumours to critical organs at risk. We undertook a retrospective review of the efficacy and safety outcomes of ultra-central lung SBRT at a major cancer centre in Hong Kong.
 
Methods
 
We analysed patients with either primary or oligometastatic ultra-central lung tumours treated with SBRT from 2009 to 2022. The primary outcome was local progression-free survival. Secondary outcomes included the incidence of grade ≥2 SBRT-related toxicity and overall survival. Clinical and dosimetric factors were collected and analysed for potential associations with survival outcomes.
 
Results
 
A total of 66 patients were included. Twenty-four cases were primary lung tumours and 42 were lung metastases, with the majority of metastatic lesions being of lung origin (n = 32). Indications for SBRT for lung metastases included oligoprogression (n = 23), oligoresidual disease (n = 13), and oligorecurrence (n = 6). Most patients (86%) received 50 Gy in five fractions. Median follow-up was 54 months, and median overall survival was 59 months. The 1-year and 3-year local failure-free survival rates were 98% and 88%, respectively. Grade 3 and grade 5 toxicity rates were 4.5% and 6%, respectively. A higher dose to 4 cc of the proximal bronchial tree and tumours located within 1 cm of the mainstem bronchus were associated with grade ≥2 airway toxicity. Oesophageal mean and maximum doses, and dose to 5 cc of the oesophagus were positively associated with grade ≥2 oesophageal toxicity.
 
Conclusion
 
We demonstrated high rates of local control and acceptable toxicity outcomes with ultra-central lung SBRT. Further results from prospective studies may clarify the optimal dose fractionation and organ-at-risk constraints for this population.
 
 
Key Words: Lung neoplasms; Organs at risk; Radiosurgery
 
 
中文摘要
 
香港使用軀體立體定位放射治療超中央型肺腫瘤的十三年經驗
 
吳珈瑋、甘子揚、黃嘉誠、杜綺鈞、王晉彥、楊美雲
 
引言
由於超中央型肺腫瘤接近關鍵的危及器官,因此使用軀體立體定位放射治療(SBRT)這腫瘤具爭議性。我們在香港一所主要癌症中心進行回顧性研究,檢視超中央型肺腫瘤的治療效用及安全性。
 
方法
我們分析了於2009至2022年期間接受SBRT治療的原位或寡轉移超中央型肺腫瘤患者。主要結果為局部無惡化存活期,次要結果為二級或以上、與SBRT相關的毒性發生率及整體存活期。我們收集並分析了臨床及劑量因素,以找出與存活結果有關的潛在關聯。
 
結果
本研究共包括66名患者,24名患有原位肺腫瘤,42名出現肺轉移,大部分轉移性病變源自肺部(n = 32)。SBRT治療肺轉移的適應症包括寡進展(n = 23)、寡殘留疾病(n = 13)及寡復發(n = 6)。大部分患者(86%)分五次接受劑量為50 Gy的治療。隨訪中位數為54個月,整體存活期中位數為59個月。一年及三年局部無疾病存活率分別為98%及88%。三級及五級毒性比率分別為4.5%及6%。用於受照射的4 cc近端支氣管樹體積的較高劑量以及位於主支氣管1厘米內的腫瘤與二級或以上毒性相關。食道平均及最高劑量以及用於受照射的5 cc食道體積的劑量與二級或以上食道毒性呈正相關。
 
結論
研究結果顯示超中央型肺部SBRT的局部控制率高,而且毒性結果可接受。未來可研究釐清適用於相關患者的最佳分次劑量及危及器官的限制。
 
 
 
INTRODUCTION
 
Stereotactic body radiation therapy (SBRT) is an established treatment for medically inoperable early-stage non–small-cell lung cancer and has been increasingly utilised for treatment of oligometastatic or oligoprogressive lung metastases. In early-stage peripherally located lung tumours, SBRT confers high rates of local control, cancer specificity, and overall survival (OS) with a low incidence of severe toxicity.[1] However, the safety of SBRT to ‘ultra-central’ lesions, where the gross tumour volume (GTV) and/or planning target volume (PTV) overlaps critical mediastinal structures such as the central airway or oesophagus, remains a matter of debate.[2] This subgroup was underrepresented in the RTOG 0813 trial where ultra-central tumours comprised only 17% of the study population.[2] Alarmingly high rates of fatal airway bleeding (12%) were also reported in the phase II HILUS trial.[3]
 
SBRT for ultra-central lung tumours has been performed in our institution, a major cancer centre in Hong Kong, since its introduction in 2009. We sought to undertake a retrospective review of the efficacy and safety outcomes of ultra-central lung SBRT at our centre.
 
METHODS
 
Study Design and Patient Population
 
All consecutive cases of primary or oligometastatic ultra-central lung tumours treated with SBRT from 2009 to 2022 at Pamela Youde Nethersole Eastern Hospital were included for analysis. Patients with incomplete or missing clinical/dosimetric data were excluded. Ultra-central tumours were defined as lesions with the PTV overlapping the trachea, proximal bronchial tree (PBT) or oesophagus.
 
Procedures
 
Patients were simulated with arms above their head with arm/shoulder supports and immobilised with the BodyFIX system (Elekta, Stockholm, Sweden). Expiratory breath hold was used for lobe tumours while four-dimensional computed tomography simulation was used for upper/middle lobe tumours or for patients unable to cooperate with the breath-hold procedure. Respiratory motion was monitored using a real-time position management system (Varian; Varian Medical Systems, Palo Alto [CA], US). An additional intravenous contrast-enhanced simulation scan was performed for tumours adjacent to mediastinal structures; oral contrast was administered at the discretion of the treating physician.
 
GTV was delineated in the pulmonary window (window width = 1600 Hounsfield unit [HU] and window level = -600 HU), supplemented with images acquired in the soft tissue window (window width = 400 HU, window level = 20 HU). An internal target volume was generated from four-dimensional CT simulation scans and an isotropic margin of 5 mm expanded from the internal target volume to form the PTV. For cases undergoing breath hold, the GTV-to-PTV margin was 8 mm. No clinical target volume was used.
 
Dose fractionation schemes included 50 Gy in five fractions (57 cases), 60 Gy in eight fractions (seven cases), or 35 Gy in five fractions (two cases). Treatments were administered on alternating days.
 
Static-field dynamic intensity modulated radiotherapy and/or volumetric modulated arc therapy with 6–mega-voltage photons were used. The prescription isodose level was chosen such that 95% of the PTV received the prescribed dose and 99% of the PTV received ≥90% of the prescribed dose. The prescribed isodose ranged between 80% and 90% for all plans. Dose constraints were adapted from the RTOG 0813 protocol[2] and the American Association of Physicists in Medicine Task Group 101 report.[4] The maximum point doses (Dmax) to the trachea, the PBT and the oesophagus were limited to 105% of the prescription dose.
 
Treatment was delivered with linear accelerators (TrueBeam; Varian Medical Systems, Palo Alto [CA], US), with pretreatment cone-beam computed tomography images obtained before each treatment. Online verification and matching were performed. Systemic therapies (excluding hormonal treatments) were withheld at least 24 hours before and after SBRT.
 
All patients underwent computed tomography scans of the thorax at 6-month intervals for at least 3 years. Clinical follow-up and need for additional imaging were performed at the discretion of treating clinicians.
 
Outcomes
 
The primary outcome analysed was local progression-free survival (PFS). Secondary outcomes included the incidence of grade ≥2 SBRT-related toxicity—classified according to the Common Terminology Criteria for Adverse Events version 5.0 grading system[5]—and OS. Clinical and dosimetric factors were collected and analysed for potential associations with survival outcomes.
 
Statistical Analyses
 
Local PFS and OS rates were calculated using the Kaplan–Meier method. Local PFS was defined as the time from the date of the first SBRT fraction to either local progression or last follow-up. OS was defined as the time from SBRT to death from any cause or last follow-up.
 
Clinical and dosimetric variables were analysed using descriptive statistics. Categorical data were represented as numbers with percentages, while continuous data were reported as medians with interquartile ranges. All dosimetric parameters were converted to equivalent doses of 2-Gy fractions (alpha-beta ratio = 3, for dosimetric parameters of organs at risk only) using the linear-quadratic model for comparison across different dose fractionations.
 
Comparison of clinical and dosimetric parameters between patients with or without grade ≥2 toxicity was done with Chi squared/Fisher’s exact test for categorical variables, and the Mann-Whitney U test for continuous variables.
 
Simple Cox proportional hazards regression analyses were conducted to identify potential associations between clinical/dosimetric variables and survival outcomes. Variables with a p value < 0.1 were entered into multivariable analysis. A p value of < 0.05 was considered statistically significant.
 
Statistical analyses were performed using commercial software SPSS (Windows version 27.0; IBM Corp, Armonk [NY], US). The STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) checklist was followed in the preparation of the study.
 
RESULTS
 
Patient Population
 
A total of 66 patients were analysed. The median follow-up was 54 months (range, 4-114). Baseline patient characteristics and dosimetric parameters are detailed in Table 1. The median age was 71.5 years. Most patients (76%) had an Eastern Cooperative Oncology Group performance status score of 0 to 1.
 
Table 1. Baseline demographic, clinical and dosimetric parameters of the study population (n = 66).
 
Twenty-four cases were primary lung tumours and 42 cases were lung metastases. The histological diagnoses for primary lung and metastatic lesions are shown in Table 2. Most metastatic lesions (32/42) were of lung origin. Indications of SBRT for lung metastases included oligoprogression (n = 23), oligoresidual disease (n = 13), and oligorecurrence (n = 6).
 
Table 2. Histological subtypes of primary lung and metastatic tumours in the study population (n = 66).
 
Breath hold was used in 11 patients (17%), with the remainder utilising four-dimensional CT simulation. The median prescription isodose was 85.55%. Median GTV and PTV were 22.35 cm3 and 58.90 cm3, respectively. Tumour PTV overlapped with the PBT or trachea in 61 lesions and oesophagus in 10 lesions. The median PTV coverage by the prescription isodose was 95.15% (Table 1).
 
Local Control and Survival Outcomes
 
The 1-year and 3-year local failure-free survival rates were 98% and 88%, respectively. Mean local failure-free survival was 79 months (95% confidence interval [CI]=65-94) [Figure 1]. OS ranged from 4 to 148 months, with a median OS of 59 months (95% CI = 54-85) [Figure 2]. The 1-year and 3-year OS rates were 89.4% and 69.7%, respectively.
 
Figure 1. Kaplan–Meier curve for local failure-free survival.
 
Figure 2. Kaplan–Meier curve for overall survival.
 
Safety Outcomes
 
Grade ≥2 toxicity occurred in 18 patients (27%). Three patients (5%) had grade 3 toxicity, including oesophageal stricture, radiation pneumonitis, and lung collapse for each. Four patients (6%) had grade 5 toxicity (one case of oesophageal ulcer bleeding, two cases of airway bleeding, and one case of multifactorial respiratory failure). The median time to grade ≥3 toxicity was 4.5 months.
 
Among the 61 patients with PTV overlapping the PBT or trachea, grade ≥2 pulmonary toxicity occurred in 14 cases (23%). Airway obstruction and/or bleeding occurred in all 14 patients, and eight also had radiation pneumonitis. Among the 10 patients with PTV overlapping the oesophagus, four (40%) developed grade ≥2 oesophageal toxicity, including two with odynophagia, one with oesophageal stricture, and one with oesophageal ulcer bleeding.
 
When comparing patients with or without grade ≥2 airway toxicity (bleeding or obstruction), there was a statistically significant difference for higher Dmax (p = 0.035) and higher dose to 4 cc (D4cc) of the PBT (p = 0.002) [Table 3].
 
Table 3. Mann-Whitney U test for grade ≥2 radiotherapy-related toxicities (n = 66).
 
For grade ≥2 airway bleeding, a statistically significant difference was found with group A tumours (≤1 cm from the main bronchi and trachea)[3] [p = 0.039], while a higher D4cc of the PBT (p = 0.075) and endobronchial tumour location (p = 0.083) did not reach statistical significance. The use of anticoagulant or antiangiogenic therapy was not significantly associated with grade ≥2 bleeding (p = 0.276) [Table 4].
 
Table 4. Chi squared/Fisher’s exact test for grade ≥2 radiotherapy toxicities.
 
Baseline forced expiratory volume in 1 second, smoking status, history of chronic obstructive pulmonary disease, and the percentage of lung receiving a dose ≥20 Gy were not significantly associated with grade ≥2 radiation pneumonitis. For grade ≥2 oesophageal toxicity, statistically significant differences were found in higher mean dose (Dmean) [p < 0.001], higher Dmax (p = 0.004), and higher dose to 5 cc (D5cc) of the oesophagus (p = 0.005) [Table 3].
 
No postmortems were performed; thus, all deaths classified as grade 5 events were based on clinical grounds alone.
 
Simple and Multivariable Analyses
 
Simple Cox regression found age (hazard ratio [HR] = 0.957, 95% CI = 0.917-0.999; p = 0.047), and group A tumours (HR = 0.316, 95% CI = 0.106-0.945; p = 0.039) were predictors for local failure; however, these variables were not significant in the multivariable analysis (Table 5). Simple cox regression did not find any significant predictors for OS (Table 6).
 
Table 5. Simple and multivariable Cox regression analyses of predictors for local failure.
 
Table 6. Simple Cox regression analysis of predictors for overall survival.
 
DISCUSSION
 
Our study provides some of the longest follow-up data on the real-world outcomes of SBRT to ultra-central lung tumours. With a median follow-up of 54 months, our 3-year local control rate of 88% and grade 3 and grade 5 toxicity rates (5% and 6%, respectively) were comparable to prior studies.[6] In a recent meta-analysis of ultra-central SBRT including 1183 patients over 27 studies,[6] the pooled 2-year local control rate was 89%, while the grade 3 to 4 toxicity rate was 6% and the grade 5 toxicity rate was 4%.
 
A primary concern in ultra-central lung SBRT is radiation-induced airway bleeding. In our study, grade ≥2 airway bleeding occurred in only five patients (8%), including three fatal haemorrhages, representing 5% of the study population. A possible reason may be our institutional practice of limiting hotspots to 120%, in contrast to the HILUS trial[3] where hotspots of up to 150% were allowed.
 
Our univariate analysis revealed that grade ≥2 airway toxicity was associated with a higher D4cc of the PBT, and airway bleeding occurred more frequently in group A tumours. This is consistent with findings of prior dosimetric studies[7] [8] where the majority of fatal lung haemorrhages were observed in group A tumours, with rates of 70% to 89%.
 
Our grade 5 toxicity rate of 6% is comparable to previous studies on ultra-central lung SBRT.[6] Among these, two patients had bronchoscopy-proven endobronchial tumour involvement. Although endobronchial tumour location was more common in patients with grade ≥2 airway bleeding the association did not reach statistical significance (p = 0.083) [Table 4]. This parallels findings from Tekatli et al[9] where endobronchial tumours comprised 46% of all SBRT-related grade ≥3 lung haemorrhages.
 
In our cohort, 10 patients had the PTV overlapping the oesophagus, and grade 3 to 5 events occurred in three of them. Literature focusing specifically on oesophageal toxicity in SBRT is limited, with small sample size. In Wang et al’s retrospective study[10] of 88 patients, 23 tumours had the PTV overlapping the oesophagus. Grade ≥3 oesophageal toxicity rate was 13%, including two cases of tracheoesophageal fistulisation.[10] Univariate analysis suggested that shorter distance between the tumour and the oesophagus predicted toxicity and suggested the use of more protracted fractionation.[10]
 
Our dosimetric analysis revealed that oesophageal Dmax, D5cc, and Dmean were associated with higher rates of grade ≥2 oesophageal toxicity. However, the optimal threshold for oesophageal toxicity remains undefined in the literature. Among the 10 patients whose PTV overlapped the oesophagus, D5cc exceeded the RTOG 0813 constraint of 27.5 Gy in three patients, two of whom had grade ≥3 events. This suggests that strict adherence to a D5cc of <27.5 Gy may help to reduce severe toxicity.
 
Taken together, our results and the literature suggest that lesions close to/abutting the oesophagus carry a substantial risk of SBRT-related toxicity. Protracted fractionations and avoiding tumours with direct invasion or abutment of the oesophagus would be advisable to reduce severe toxicity. Further data are awaited to define optimal dose constraints and fractionation for these tumours.
 
Limitations
 
Our study had several limitations, including its retrospective nature and non-randomised design. Our sample size was also small, and the number of events was too limited for detailed statistical analyses and elucidation of safe dose constraints for the investigated toxicity endpoints. Our database relied on clinical records documented by treating clinicians rather than a prospective database for research purposes; thus, some toxicities may have been underreported. The lack of autopsy information on the exact cause of death also made it difficult to definitively conclude whether they were truly SBRT-related mortalities.
 
CONCLUSION
 
In our study of 66 patients undergoing ultra-central SBRT, long-term follow-up showed sustained high rates of local control and acceptable toxicity outcomes. Caution should be taken when delivering SBRT to group A lesions, and attention should be paid to dosimetric constraints such as the D4cc of the PBT and the D5cc of the oesophagus. Further studies are needed to clarify the optimal dose fractionation and organ at risk constraints to minimise toxicity.
 
REFERENCES
 
1. Tateishi Y, Takeda A, Horita N, Tsurugai Y, Eriguchi T, Kibe Y, et al. Stereotactic body radiation therapy with a high maximum dose improves local control, cancer-specific death, and overall survival in peripheral early-stage non–small cell lung cancer. Int J Radiat Oncol Biol Phys. 2021;111:143-51. Crossref
 
2. Bezjak A, Paulus R, Gaspar LE, Timmerman RD, Straube WL, Ryan WF, et al. Safety and efficacy of a five-fraction stereotactic body radiotherapy schedule for centrally located non–small-cell lung cancer: NRG Oncology/RTOG 0813 trial. J Clin Oncol. 2019;37:1316-25. Crossref
 
3. Lindberg K, Grozman V, Karlsson K, Lindberg S, Lax I, Wersäll P, et al. The HILUS-trial—a prospective Nordic multicenter phase 2 study of ultracentral lung tumors treated with stereotactic body radiotherapy. J Thorac Oncol. 2021;16:1200-10. Crossref
 
4. Benedict SH, Yenice KM, Followill D, Galvin JM, Hinson W, Kavanagh B, et al. Stereotactic body radiation therapy: the report of AAPM Task Group 101. Med Phys. 2010;37:4078-101. Crossref
 
5. United States Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. 2017 Nov 27. Available from: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_5x7.pdf. Accessed 30 Apr 2025.
 
6. Yan M, Louie AV, Kotecha R, Ashfaq Ahmed M, Zhang Z, Guckenberger M, et al. Stereotactic body radiotherapy for ultra-central lung tumors: a systematic review and meta-analysis and International Stereotactic Radiosurgery Society practice guidelines. Lung Cancer. 2023;182:107281. Crossref
 
7. Tekatli H, Giraud N, van Eekelen R, Lagerwaard FJ, Senan S. Ten years outcomes after SABR in central and ultracentral primary lung tumors. Radiother Oncol. 2023;188:109848. Crossref
 
8. Lindberg S, Grozman V, Karlsson K, Onjukka E, Lindbäck E, Jirf KA, et al. Expanded HILUS trial: a pooled analysis of risk factors for toxicity from stereotactic body radiation therapy of central and ultracentral lung tumors. Int J Radiat Oncol Biol Phys. 2023;117:1222-31. Crossref
 
9. Tekatli H, Duijm M, Oomen-de Hoop E, Verbakel W, Schillemans W, Slotman BJ, et al. Normal tissue complication probability modeling of pulmonary toxicity after stereotactic and hypofractionated radiation therapy for central lung tumors. Int J Radiat Oncol Biol Phys. 2018;100:738-47. Crossref
 
10. Wang C, Rimner A, Gelblum DY, Dick-Godfrey R, McKnight D, Torres D, et al. Analysis of pneumonitis and esophageal injury after stereotactic body radiation therapy for ultra-central lung tumors. Lung Cancer. 2020;147:45-8. Crossref
 
 
 

Magnetic Resonance Imaging–guided Biopsy of the Breast: A Ten-Year Experience

CCY Chan, EPY Fung, WP Cheung, KM Kwok, WS Mak, KM Wong, LW Lo, A Wong, D Fenn, AYH Leung

ORIGINAL ARTICLE
 
Magnetic Resonance Imaging–guided Biopsy of the Breast: A Ten-Year Experience
 
CCY Chan, EPY Fung, WP Cheung, KM Kwok, WS Mak, KM Wong, LW Lo, A Wong, D Fenn, AYH Leung
Department of Diagnostic and Interventional Radiology, Kwong Wah Hospital, Hong Kong SAR, China
 
Correspondence: Dr CCY Chan, Department of Diagnostic and Interventional Radiology, Kwong Wah Hospital, Hong Kong SAR, China. Email: chancherrycy@gmail.com
 
Submitted: 29 September 2023; Accepted: 27 August 2024.
 
Contributors: CCYC, EPYF, WPC and KMK designed the study. CCYC acquired the data. CCYC and EPYF analysed the data. CCYC drafted the manuscript. All authors critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: As an editor of the journal, CCYC was not involved in the peer review process. Other authors disclosed no conflicts of interest.
 
Funding/Support: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sector.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: This research was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: KC/KE-23-0152/ER-2). Informed patient consent was waived by the Board due to the retrospective nature of the research.
 
 
 
 
 
Abstract
 
Introduction
 
Magnetic resonance imaging (MRI) is an effective modality for high-risk patient screening, local staging, and disease monitoring in breast malignancies. There is an increasing demand for MRI-guided biopsy of lesions that are occult on mammography or ultrasound. This study summarises our 10-year experience with the procedure. Technical challenges, as well as tips and tricks to achieve procedural success are discussed.
 
Methods
 
A total of 37 consecutive cases of MRI-guided vacuum-assisted breast biopsies performed at a single centre between August 2012 and August 2023 were retrospectively reviewed. Targets were localised using 1.5-T MRI systems with a dedicated breast coil and localisation device. Biopsies were performed using 10-gauge or 9-gauge needles. Imaging characteristics, histopathological results, and subsequent management for all biopsied lesions were recorded.
 
Results
 
The mean age of patients was 51.6 years. Technical success was achieved in 35 out of 37 cases (94.6%). In 27 cases (77.1%), the biopsied breast was placed in the ipsilateral coil, and in eight cases (22.9%) it was placed in the contralateral coil for optimal imaging and biopsy access. Between 8 and 24 cuttings were taken for each target. Three cases (8.6%) developed biopsy site haematomas. Of the 35 successfully biopsied lesions, 11 (31.4%) were malignant. Among the malignant lesions, six (54.5%) presented as non-mass enhancement and five (45.5%) as mass enhancement. Four lesions (36.4%) showed restricted diffusion, while seven (63.6%) did not.
 
Conclusion
 
MRI-guided vacuum-assisted biopsy of the breast is a safe and effective procedure in the hands of experienced interventionists. It is essential for the diagnosis and management of breast lesions occult on conventional imaging.
 
 
Key Words: Breast neoplasms; Fibroadenoma; Image-guided biopsy; Magnetic resonance imaging; Mammography
 
 
中文摘要
 
磁力共振成像引導乳腺活檢檢查:十年經驗分享
 
陳卓忻、馮寶恩、張偉彬、郭勁明、麥詠詩、黃嘉敏、羅麗雲、黃皓澧、范德信、梁燕霞
 
引言
磁力共振成像是乳腺惡性腫瘤高風險患者篩檢、局部分期和病情監測的有效方法。對於乳房造影掃瞄或超聲波檢查中難以發現的病變,磁力共振成像引導下活檢的需求日益增長。本研究總結了我們十年來在該技術上的經驗,並探討其中的技術挑戰以及確保活檢成功的技巧和竅門。
 
方法
我們對2012年8月至2023年8月期間在同一中心進行的連續37例磁力共振成像引導下真空輔助乳腺活檢病例進行回顧性分析。我們使用配備專用乳腺線圈和定位裝置的1.5 T磁力共振成像系統進行標靶區定位。活檢採用10號或9號穿刺針。我們記錄了所有活檢病變的影像學特徵、組織病理學結果及後續處理。
 
結果
患者平均年齡為51.6歲。 37例患者中35例(94.6%)技術成功。活檢時27例(77.1%)乳腺置於同側線圈內,8例(22.9%)乳腺置於對側線圈內,以獲得最佳影像及活檢入路。每個目標活檢8至24個乳腺組織。三例(8.6%)出現活檢部位血腫。在35個成功活檢的病灶中,11例(31.4%)為惡性病灶。在惡性病灶中,6例(54.5%)表現為無腫塊強化,5例(45.5%)表現為腫塊強化。四例(36.4%)病灶顯示彌散受限,7例(63.6%)未顯示彌散受限。
 
結論
磁力共振成像引導下乳腺真空輔助活檢在經驗豐富的介入醫生操作下是一種安全有效的操作,對於常規影像學檢查無法確診的乳腺病變的診斷和治療至關重要。
 
 
 
INTRODUCTION
 
Magnetic resonance imaging (MRI) of the breast is an effective modality for screening high-risk patients, local staging and monitoring breast malignancies that are occult on radiography and sonography. This article summarises our centre’s experience in MRI-guided breast biopsy over the past 10 years, with the aim of reviewing the fundamentals of the procedure and emphasising tips and tricks for technically challenging cases.
 
MAGNETIC RESONANCE IMAGING–GUIDED BREAST BIOPSY
 
Indications and Contraindications
 
MRI of the breast is performed for indications as categorised by the European Society of Breast Cancer Specialists working group, including screening for high-risk patients such as those with a strong family history of breast malignancies or known genetic mutations, e.g., BRCA1 and BRCA2; characterisation of inconclusive findings on mammography or ultrasound; assessment for unknown primary breast cancer; preoperative local staging and surgical planning in patients with biopsy-proven breast malignancy (particularly those considered for breast conserving therapy); and disease monitoring in known breast malignancies, e.g., evaluating treatment response to neoadjuvant chemotherapy.[1] When a suspicious lesion occult on conventional breast imaging (i.e., mammography and ultrasound) is detected on MRI, an MRI-guided biopsy should be performed if a targeted second-look ultrasound is unyielding according to the American College of Radiology (ACR)[2] and the European Society of Breast Imaging[3] recommendations. Absolute contraindications to MRI-guided breast biopsy are the same as for any MRI scan, including the presence of MRI-incompatible metallic or magnetic implants, claustrophobia, contrast allergy, or severe renal impairment.[4] Relative contraindications include thrombocytopenia and coagulopathies.[4]
 
Magnetic Resonance Imaging Protocol
 
At our centre, MRI-guided breast biopsy is performed using one of two 1.5-T MRI systems (Achieva XR; Philips Healthcare, Best, the Netherlands, and MAGNETOM Sola; Siemens Healthcare, Erlangen, Germany) with phased-array dedicated breast coils containing four or seven channels, respectively. The MRI protocol for biopsy differs from the usual diagnostic protocol by using breast coils with fewer channels and prioritising rapid image acquisition with sequences optimised for target localisation.[5] Our protocol consists of T1-weighted pre- and post-contrast sequences acquired in 1-mm section thickness. Gadoterate meglumine is the gadolinium-based contrast medium of choice, administered at the recommended dose of 0.2 mL/kg (or 0.1 mmol/kg) at a rate of 2 mL/s via a pump injector. Dynamic T1-weighted three-dimensional fat-saturated images are acquired, with the first set of post-contrast images obtained 1.5 to 2 minutes after injection, followed by a second set at a 25-second interval, and then at 1-minute intervals up to 4 minutes, as recommended by the ACR guidelines.[2] Subtraction of the unenhanced images is performed.
 
Patient Positioning
 
Optimal patient positioning is crucial for procedural success. The patient is positioned prone with cushion support for the head and neck, and a headset for noise cancellation. Arms are positioned overhead with padding at the sternum, abdomen, and legs for comfort and stability. The targeted breast is placed hanging freely and as deeply and centrally as possible in the dedicated breast coil with the nipple pointing directly downwards.[6] The operator should ensure there are no breast folds resulting from compression at the edge of the coil, as this leads to uneven fat saturation on MRI.[7]
 
Lesion Localisation
 
Once the patient is optimally positioned, breast compression is performed using a grid paddle. Pre-contrast MRI is conducted to verify breast placement to ensure the target falls within the multichannel localisation grid.[6] A fiducial marker, either an MRI-visible fish oil capsule or a small plastic marker is affixed to skin of the breast within the grid square, close to, but not directly over, the anticipated location of the target. Contrast is administered and MRI images are acquired as per protocol. Post-processing subtraction images are generated to localise the target.
 
Following localisation, the biopsy tract is planned either manually or using the computer-assisted localisation software[4] [6] (syngo MR XA 51; Siemens, Erlangen, Germany) [Figure 1a]. In the manual approach, an MRI grid worksheet (Figure 1b) with two sets of views, namely, the patient view and the MRI view, is used to select the localisation grid channel and needle tunnel based on calculation of lesion coordinates. The patient view represents a 90° anti-clockwise rotation of the sagittal MRI image, as in reality the patient lies prone. The image section where the hypointense localisation grid contacts the skin surface is taken as the first section (Figure 1c). The number of sections from this point to the target is multiplied by section thickness to determine lesion depth. The thickness of the needle guidance cube block (2 cm) is then added for calculation of overall needle insertion depth. Distances between the fiducial marker and the target along the horizontal and vertical axes are also measured. It is important to verify correct laterality (i.e., left or right breast) and approach (i.e., lateral or medial) when selecting the worksheet as each is different; and to carefully translate the target from the MRI view to the patient view by turning anti-clockwise of the clock face or direction by 90° on the worksheet, as any mistake in these steps will result in inaccurate targeting. Alternatively, the computer localisation software automatically calculates lesion coordinates and indicates the specific square of the multichannel localisation grid, the tunnel within the needle guide cube block, and the required needle insertion depth for accurate targeting.[6]
 
Figure 1. (a) Lesion localisation using computer software. (b) Magnetic resonance imaging (MRI) grid worksheet used for biopsy planning in manual calculation with image view on the left and patient view on the right. Note that the target position is at the left lower corner (black star) in grid square B2 (black arrow) on the image view (left). On the patient view (right) there is a 90-degree rotation from the image view, hence the target is located at the right lower corner (black star) in grid square B2 (black arrow). After careful translation of the target position from image to patient view, the best suited needle insertion tunnel of the needle guidance cube block is selected, which in this case is the one in the right lower corner (black star indicated by black arrow in IV of patient view). (c) Pre-biopsy sagittal image shows the section where the hypointense localisation grid touches the breast surface. The MRI-visible obturator is inserted into the square of the grid where the biopsy trajectory is calculated (white arrow). (d) Target lesion (white arrow) on T1-weighted post-gadolinium subtraction axial image. (e) The obturator is confirmed to align with the target in (d) [white arrow]. (f) The vacuum-assisted biopsy needle is inserted into the lockable needle guidance cube block and held in place for sampling.
 
Biopsy
 
Following injection of local anaesthesia and skin incision at the expected needle position based on calculated lesion coordinates, a small lockable needle guidance cube block is inserted into the localisation grid channel (i.e., one of the many channels/boxes from the square grid; A1 to F8 in Figure 1a) over the skin incision and secured. The numerically labelled plastic introducer sheath, with its depth stop set to the calculated insertion depth, together with the inner non-ferrous metallic trocar, is inserted into one of the tunnels of the needle guidance cube block, which is best positioned over the skin incision, and the coaxial system is advanced to the calculated lesion depth. The metallic trocar is then replaced with an EnCor MRI-visible obturator (BD Inc, Franklin Lakes [NJ], US) and MRI images are acquired to confirm the alignment of the obturator with the target (Figure 1d and e). The obturator is then removed and the vacuum-assisted biopsy needle is inserted to the same calculated depth. The aperture of the biopsy needle is oriented to face in the direction of the lesion relative to the selected needle tunnel, a technique known as ‘directional sampling’.[7] Vacuum-assisted biopsy is then performed (Figure 1f) with the desired number of cuttings and the needle is removed. A post-biopsy MRI scan is acquired to confirm the correct site and adequate sampling of the target.
 
After Biopsy
 
After sampling, an MRI-compatible biopsy marker is inserted via the biopsy tract through the introducer sheath and deployed at the biopsy site. Acquisition of post-marker insertion images is optional and not routinely performed, as haematoma or gas artefacts often obscures the marker.[8] All needles are removed and haemostasis is achieved by manual compression of the breast for at least 15 minutes.
 
Complications
 
Bleeding and haematoma formation at biopsy site are the most common complications.[9] Other complications include infection and muscle injury (i.e., injury to the pectoralis muscles). Rare complications include pneumothorax and injury to mediastinal structures, which only occur when biopsy is performed using a freehand approach without a localisation grid.[9]
 
METHODS
 
Thirty-seven consecutive cases of MRI-guided vacuum-assisted breast biopsies performed between August 2012 and August 2023 in a single centre were retrospectively reviewed. Target lesions were localised using the Philips or Siemens 1.5-T MRI system with dedicated breast coils and an Invivo (Philips, Amsterdam, Netherlands) or Breast BI 7 Coil (Siemens, Höchberg, Germany) localisation device. Biopsies were performed using 10-gauge EnCor or 9-gauge Suros (Hologic, Marlborough [MA], US) needles. Imaging characteristics including size, morphology, and enhancement pattern were recorded. Histopathology of all biopsied lesions was obtained with subsequent management documented.
 
RESULTS
 
The mean age of patients was 51.6 years (range, 33-76). Technical success was achieved in 35 out of 37 cases (94.6%). In three cases, the original target was not visualised on pre-procedural MRI, resulting in cancellation of the procedure in two cases, while a nearby target was selected in the remaining case.
 
In 27 cases (77.1%), the lateral approach was adopted and the biopsied breast was placed in the ipsilateral coil. In eight cases (22.9%), the medial approach was used, and the breast was placed in the contralateral coil. A total of 34 lesions were biopsied using a 10-gauge EnCor needle and one lesion was biopsied using a 9-gauge Suros needle. Between 8 and 24 cuttings were taken for each target, with an average of 13 cuttings made. Three cases (8.6%) were complicated by biopsy-site haematomas: two were managed by prolonged manual compression for more than 30 minutes and one required aspiration of the haematoma using the vacuum-assisted biopsy device followed by manual compression. Haemostasis was successfully achieved in all three cases.
 
Table 1 shows the histology of the lesions and Table 2 shows the malignant diagnoses. Of the 11 malignant lesions, one case (9.1%) yielded a false-negative result from MRI-guided biopsy and proceeded to surgical excision after consensus was reached at the multidisciplinary meeting due to clinical and radiological-histopathological discordance. Histology from the surgical specimen revealed invasive ductal carcinoma (Table 2).
 
Table 1. Histopathological results of biopsied lesions (n = 35)
 
Table 2. Histopathological results of the biopsied lesions positive for malignancy (n = 11)
 
Six malignant lesions (54.5%) presented as non-mass enhancement and five as mass enhancement (45.5%). The size of the malignant lesions ranged from 0.5 cm to 4.3 cm. Four lesions (36.3%) showed restricted diffusion, while seven (63.6%) did not. Nine malignant lesions (81.8%) exhibited a type II enhancement curve and two (18.2%) demonstrated a type III enhancement curve. Ten malignant lesions (90.9%) were classified as BI-RADS (Breast Imaging Reporting and Data System)[10] category 4 and the remaining case (9.1%) as category 5. All but one patient underwent surgery with either mastectomy or breast conserving therapy; the remaining patient declined surgery and remained under regular clinical and radiological follow-up.
 
DISCUSSION
 
MRI-guided vacuum-assisted biopsy of the breast is a technically demanding procedure requiring specialised equipment and a skilled, well-trained team with appropriate experience. Various factors contribute to procedural success. First, patient safety in the MRI suite should be ensured for a smooth procedure. Any equipment entering the suite should be carefully examined for MRI compatibility.[2] A trolley is usually prepared for the transport of equipment in and out of the suite between image acquisition and biopsy, and all metallic devices must be removed during image acquisition. Second, efficiency is essential for successful biopsy owing to limited timeframe between lesion enhancement and contrast washout, while progressive background parenchymal enhancement further obscures targets.[7] It is also important to note that patients are often placed in an uncomfortable position and may move during prolonged procedures, resulting in lesion motion and therefore sampling failure.[7] Meticulous preprocedural planning with review of the diagnostic MRI, education and communication with the patient prior to biopsy to reduce anxiety and manage expectation, particularly regarding the importance to stay still throughout the procedure, and efficient execution of each biopsy step is therefore crucial to achieve procedural success. The following are tips and tricks accumulated over the years to address challenging cases.
 
Patient selection
 
Compression Technique
 
Controlled compression of the breast with moderate pressure is performed with a grid paddle and should be adequate for immobilisation with the breast just taut. Inadequate compression increases the risk of mistargeting due to breast and lesion motion throughout the procedure, while excessive compression increases patient discomfort and may impede blood flow to the breast, resulting in reduced or non-enhancement of the target leading to localisation failure.[9]
 
Thin Breasts
 
Thin breasts (Figure 2a) present unique challenges. Target lesions may not fall adequately into the breast coil to allow needle access. Optimising patient positioning can markedly improve procedural success, whereby chest pads can be removed from the coil or replaced by thinner pads, and the patient can be tilted into an oblique position, allowing the breast to drop further into the coil lumen.[11] After compression, breast thickness is further reduced (Figure 2b) and may be inadequate to accommodate the standard biopsy needle, therefore compression may be reduced to increase tissue thickness to allow biopsy.[12] Local anaesthesia can also be injected either anterior or posterior to the target to increase distance between skin and the target to accommodate the biopsy needle. A blunt tip needle, half-aperture size needle or petit needle (e.g., 13-gauge) can be employed to minimise chest wall injury or contralateral skin penetration risks.[11]
 
Figure 2. Biopsy techniques in thin breasts. Thin breasts are commonly encountered in the Asian population and pose difficulty due to limited tissue thickness which may not adequately fall into the breast coil lumen. (a) A patient with thin breasts without compression on diagnostic magnetic resonance imaging. (b) Further reduction in breast thickness is noted after compression during biopsy in the same patient. A half-aperture size needle is employed in this case to avoid skin injury.
 
Lesion Located at The Cross of Localisation Grid Squares
 
Occasionally, the target may fall onto the intersection of localisation grid squares after injection of local anaesthesia as shown in Figure 1a (red circle). In such cases, directional sampling and appropriate manoeuvring will significantly improve procedural success.[7] The needle guidance cube block is inserted into the A4 square and the biopsy needle is inserted into the tunnel of the cube block indicated by computer software (tunnel c3; highlighted in green), then directional sampling is performed with the aperture of the biopsy needle facing the 7 to 8 o’clock position aiming at the target, while manual pressure is applied by the operator’s finger through another grid square (B4 in this case) in attempts to push the breast tissue and hence the target towards the direction of the biopsy needle to aid sampling (Figure 1a). Niketa et al[11] also described a biopsy technique with two diagonally placed entry sites in adjacent holes paired with directional sampling technique to improve sampling success in such cases.
 
Location of Lesions in the Breast
 
Anterior Lesions
 
For anterior lesions, with large breasts, they may touch the table and distort the breast, rendering localisation difficult. Padding can be added to raise the body from the coil so that the target is more easily reached.
 
Posterior Lesions
 
For lesions close to the chest wall (Figure 3a), removing coil cushion covers brings the chest closer to the coil aperture. The arms can be placed down by the sides of the patient instead of above the head, as this relaxes the pectoralis muscles and allows the breast to sink deeper into the coil (Figure 3b). However, if the target is too close to the pectoralis muscles, the arms should be placed above the head to help retract the muscle away from the coil lumen to avoid muscle injury, which can cause excessive pain and haemorrhage. Tilting the patient into an oblique position may also help the posterior parts of the breast sink further (Figure 3c). On rare occasions, the target may lie posterior to the localisation grid even after these manoeuvres. Performing the biopsy posterior to the grid or by freehand needle insertion without breast compression and localisation grids has been described.[11] The importance of maintaining stability of the needle position in these scenarios is emphasised, as the absence of support from the localisation grid and/or immobilisation of the breast from compression increases the difficulty of targeting.
 
Figure 3. Biopsy techniques for posteriorly located lesions. (a) A posteriorly located BI-RADS (Breast Imaging Reporting and Data System) category 4 lesion (arrow). (b) The patient’s arms are placed down by her sides to relax the pectoralis muscles, allowing the posterior breast to sink deeper into the coil. (c) The obturator (arrow) is aiming at the target lesion after patient positioning was optimised. Sampling was successful in this case.
 
Medial Lesions
 
It is difficult to target medial lesions from the medial side due to the increased distance between the biopsy apparatus and the breast when the breast is placed in an ipsilateral coil. The design of the breast coil, with a downward slant from the lateral bar to the sternal bar, also aggravates the difficulty of accessing posteromedial lesions, as the further the biopsy needle travels, the more anteriorly (towards the nipple) the needle tip will be directed due to this angulation.[6] It is thus often helpful to place the targeted breast in the contralateral breast coil (i.e., the right breast in the left breast coil [Figure 4]), which shortens the distance between the biopsy apparatus and the target and reduces the downward angulation the biopsy needle must overcome. This is a less comfortable position for the patient due to the tilting, making it harder to stay still. It is therefore vital for operators to optimise patient comfort before commencement of biopsy to minimise lesion motion. Another limitation to this manoeuvre is that obese patients may not be able to fit through the bore of the MRI.[6]
 
Figure 4. Biopsy techniques for medially located lesions. The patient was initially positioned with her right breast placed in the right breast coil. She had a target lesion located in the right inner breast (a) [arrow]. However, simulation for biopsy found that the target would be difficult to approach from the medial side as the large distance between the biopsy apparatus and the target rendered positioning of the biopsy needle (b) [arrow] suboptimal. The patient was then repositioned obliquely, with her right breast placed in the left breast coil (c). The biopsy was completed smoothly with successful sampling.
 
Superficial Lesions
 
Injury to the skin is the primary concern in these lesions. If the needle aperture is not completely embedded within the breast during biopsy, air leakage and loss of vacuum effect may ensue, which further lower the rate of successful sampling.[11] This can be tackled by generous injection of local anaesthetic proximal to the target to increase tissue depth to accommodate the biopsy needle.[11] The biopsy needle can also be inserted a few millimetres beyond the target so that the target falls into the proximal part of the needle aperture. Alternatively, smaller-aperture needles may be employed.
 
Periareolar Lesions
 
Biopsy around the nipple-areolar complex carries increased risks of haemorrhage and pain as it is a highly vascularised and innervated structure. It also raises cosmetic concerns. In these cases, the nipple-areolar complex can be manually rolled away from the expected site of skin incision and biopsy needle entry to avoid injury to the complex.[11]
 
Breast Implants
 
Breast implants are increasingly common, and their presence renders biopsy difficult. The operator must exercise extra caution not to puncture the capsule, which may result in implant rupture. Adequacy of breast parenchymal thickness should be assessed according to the expected biopsy trajectory and if there is inadequate tissue depth, half-aperture needles can be employed.[7] If the target is located too close to the implant, blunt-tip needles may minimise the risk of implant puncture (Figure 5), or alternatively, fine needle aspiration can be performed instead.[12] Injection of local anaesthetic between the target and the implant also allows tissue dissection and increases the distance between the two, providing more room for tissue sampling.
 
Figure 5. Biopsy techniques in patients with breast implants. A patient with breast implants and a BI-RADS (Breast Imaging Reporting and Data System) category 4A lesion in the right outer breast (a) [arrow]. The presence of breast implants often limits tissue depth for sampling, especially when the target is located close to the implant. This can be resolved by using needles with a blunt tip or half aperture, as in this case (b).
 
Non-visualisation of the Target in Pre-biopsy Magnetic Resonance Imaging
 
Non-visualisation of the target (Figure 6) in preprocedural MRI has been reported in approximately 8% to 13% of cases.[13] Several factors should be taken into consideration before abandoning biopsy. The diagnostic MRI should be carefully reviewed to identify the sequences in which the target is best visualised. For instance, the lesion may be T2-hyperintense or shows restricted diffusion on diffusion-weighted images (Figure 7), and if it is not well delineated in the standard pre-biopsy MRI sequences, these additional sequences should be performed. This is also helpful when other non-target lesions are conspicuous in these sequences and can be identified as landmarks. Sometimes, the lesion may not be identified due to inherent differences between the breast coils used in diagnostic and pre-biopsy protocols, as the latter includes a smaller number of channels, which may lower the image quality. It is also important to bear in mind that the breast is compressed using a grid paddle in preprocedural MRI, whereas no compression is applied during diagnostic MRI. Enhancement dynamics of the target may therefore differ as blood inflow may be impeded by compression of the breast, preventing lesion enhancement and resulting in a false-negative scan.[7] In such cases, the operator should verify that compression pressure is not excessive and reduce it if necessary. Another tip is to prolong post-contract image acquisition, e.g., at 1-minute intervals for up to 5 minutes, as lesion enhancement may be delayed due to compression of the breast.[6] Non-visualisation can persist after these manoeuvres due to several factors, including fluctuation in background parenchymal enhancement related to hormonal cycles and transient infective or inflammatory process.[7] In such events, the biopsy should be cancelled. However, non-visualisation of the target during biopsy does not preclude malignancy, which has been found in approximately 3.5% of such cases upon follow-up imaging.[14] It is therefore prudent to perform a follow-up MRI within 6 months upon cancellation of biopsy according to ACR recommendations.[2]
 
Figure 6. Non-visualisation of target in pre-biopsy magnetic resonance imaging (MRI). (a) Enhancing target in the left inner breast (arrow). (b) The target was not well delineated in the pre-biopsy MRI despite adjustment of compression pressure and acquisition of delayed post-contrast images; therefore, the biopsy was abandoned. A follow-up scan performed 7 months later also shows the lesion was no longer visualised (not shown).
 
Figure 7. Non-enhancing lesions with restricted diffusion. Diffusion-weighted imaging (DWI) [a, c] with corresponding apparent diffusion coefficient maps (b, d) showing a 1.6-cm irregular area of restricted diffusion in the left inner breast (arrows) without corresponding enhancement. This lesion was graded as BI-RADS (Breast Imaging Reporting and Data System) category 4A, and magnetic resonance imaging–guided biopsy was performed based on DWI images, without the standard T1-weighted pre- and post-gadolinium injection sequences. Histopathology revealed fibrocystic change with sclerosing adenosis and ductal ectasia (benign).
 
Postprocedural Haematoma
 
Manual compression is applied to the biopsied breast for haemostasis for at least 15 minutes. A pressure dressing or tight breast wraps can be used to facilitate further compression afterwards. A sizeable biopsy site haematoma may sometimes be seen on post-biopsy MRI. In such cases, the vacuum-assisted biopsy needle can be re-inserted through the co-axial system and switched to aspiration mode for evacuation of the haematoma before deployment of the biopsy marker (Figure 8). This often reduces pain as well as minimises the risk of marker displacement. In cases of uncontrolled bleeding with suspected arterial injury, thrombin injection into the biopsy cavity may be helpful for haemostasis control.[11]
 
Figure 8. Postprocedural haematoma. (a) A large biopsy site haematoma (arrow) on a postprocedural image. (b) Another case complicated by a biopsy site haematoma. A cavity with an air-blood level (arrow) is seen as a vertical line in this prone patient. (c) Aspiration of the haematoma was performed in the case shown in (b) with the vacuum-assisted biopsy device. Post-aspiration image shows marked reduction in the size of the cavity (arrow).
 
Radiological-Histopathological Discordance
 
Unlike ultrasound-guided biopsy where there is real-time visualisation of the biopsy trajectory and lesion, or in stereotactic- or tomosynthesis-guided core biopsy where specimen radiographs confirms the presence of calcifications, there is no direct method to assess targeting accuracy in MRI-guided vacuum-assisted biopsy. Radiological-histopathological concordance is therefore of utmost importance to avoid missing any malignancies in cases of suspicious imaging findings with negative biopsy results.[15] [16] It is the operator’s responsibility to review the histology results and report any discordance to the surgical team, for which the next appropriate step of management entails a repeated or excisional biopsy.
 
CONCLUSION
 
MRI has become an indispensable component of breast imaging due to its high sensitivity in lesion detection. However, its limited specificity, with significant overlap of MRI characteristics between malignant and benign lesions, highlights the importance of radiological-histopathological correlation. It is therefore vital for breast radiologists to understand the fundamentals of MRI-guided vacuum-assisted biopsy in the face of its growing demands to achieve technical success and guide the management of breast lesions occult on mammography and ultrasound. MRI-guided vacuum-assisted biopsy of the breast is a safe, feasible, and effective procedure with high diagnostic yield in the hands of experienced interventionists.
 
REFERENCES
 
1. Sardanelli F, Boetes C, Borisch B, Decker T, Federico M, Gilbert FJ, et al. Magnetic resonance imaging of the breast: recommendations from the EUSOMA working group. Eur J Cancer. 2010;46:1296-316. Crossref
 
2. American College of Radiology. ACR Practice Parameter for the Performance of Magnetic Resonance; Image-guided Breast Interventional Procedures. Reston, VA: American College of Radiology; 2016.
 
3. Mann RM, Balleyguier C, Baltzer PA, Bick U, Colin C, Cornford E, et al. Breast MRI: EUSOBI recommendations for women’s information. Eur Radiol. 2015;25:3669-78. Crossref
 
4. Papalouka V, Kilburn-Toppin F, Gaskarth M, Gilbert F. MRI-guided breast biopsy: a review of technique, indications, and radiological-pathological correlations. Clin Radiol. 2018;73:908.e17-25. Crossref
 
5. McGrath AL, Price ER, Eby PR, Rahbar H. MRI-guided breast interventions. J Magn Reson Imaging. 2017;46:631-45. Crossref
 
6. Price ER. Magnetic resonance imaging–guided biopsy of the breast: fundamentals and finer points. Magn Reson Imaging Clin N Am. 2013;21:571-81. Crossref
 
7. Santiago L, Candelaria RP, Huang ML. MR imaging–guided breast interventions: indications, key principles, and imaging-pathology correlation. Magn Reason Imaging Clin N Am. 2018;26:235-46. Crossref
 
8. Liberman L, Bracero N, Morris E, Thornton C, Dershaw DD. MRI-guided 9-gauge vacuum-assisted breast biopsy: initial clinical experience. AJR Am J Roentgenol. 2005;185:183-93. Crossref
 
9. Heywang-Köbrunner SH, Sinnatamby R, Lebeau A, Lebrecht A, Britton PD, Schreer I, et al. Interdisciplinary consensus on the uses and technique of MR-guided vacuum-assisted breast biopsy (VAB): results of a European consensus meeting. Eur J Radiol. 2009;72:289-94. Crossref
 
10. American College of Radiology. Breast Imaging Reporting and Data System, 5th edition. Reston, VA: American College of Radiology; 2013.
 
11. Niketa C, Pang KA, Lim JW. Challenges in MRI-guided breast biopsy and some suggested strategies: case-based review. Diagnostics (Basel). 2022;12:1985. Crossref
 
12. Chesebro AL, Chikarmane SA, Ritner JA, Birdwell RL, Giess CS. Troubleshooting to overcome technical challenges in image-guided breast biopsy. Radiographics. 2017;37:705-18. Crossref
 
13. Gao P, Kong X, Song Y, Song Y, Fang Y, Ouyang H, et al. Recent progress for the techniques of MRI-guided breast interventions and their applications on surgical strategy. J Cancer. 2020;11:4671-82. Crossref
 
14. Brennan SB, Sung JS, Dershaw DD, Liberman L, Morris EA. Cancellation of MR imaging–guided breast biopsy due to lesion nonvisualization: frequency and follow-up. Radiology. 2011;261:92-9. Crossref
 
15. Meucci R, Pistolese Chiara A, Perretta T, Vanni G, Portarena I, Manenti G, et al. MR imaging–guided vacuum assisted breast biopsy: radiological-pathological correlation and underestimation rate in pre-surgical assessment. Eur J Radiol Open. 2020;7:100244. Crossref
 
16. Myers KS, Kamel IR, Macura KJ. MRI-guided breast biopsy: outcomes and effect on patient management. Clin Breast Cancer. 2015;15:143-52. Crossref
 
 
 

Outcomes of Patients with Unresectable Stage III Non–Small-Cell Lung Cancer Treated with Durvalumab After Chemoradiotherapy

SSN Leung, MY Lim, TTS Lau

ORIGINAL ARTICLE
 
Outcomes of Patients with Unresectable Stage III Non–Small-Cell Lung Cancer Treated with Durvalumab After Chemoradiotherapy
 
SSN Leung, MY Lim, TTS Lau
Department of Oncology, Princess Margaret Hospital, Hong Kong SAR, China
 
Correspondence: Dr SSN Leung, Department of Oncology, Princess Margaret Hospital, Hong Kong SAR, China. Email: sheonaleung@ha.org.hk
 
Submitted: 23 August 2024; Accepted: 25 November 2024.
 
Contributors: All authors designed the study. SSNL acquired the data. SSNL and MYL analysed the data. SSNL drafted the manuscript. All authors critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: The research was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: CIRB-2024-185-2). The requirement for informed patient consent was waived by the Board due to the retrospective nature of the research and the use of anonymised data in the research.
 
 
 
 
 
Abstract
 
Introduction
 
This study evaluated the efficacy and safety of durvalumab in unresectable stage III non–small-cell lung cancer (NSCLC) at a tertiary centre in Hong Kong.
 
Methods
 
Cases of stage III NSCLC treated with radical-intent chemoradiotherapy (CRT), with or without durvalumab, from December 2017 to June 2023 were included. Outcomes, including progression-free survival (PFS) and overall survival, were analysed using the Kaplan–Meier method. Adverse events, including any-grade pneumonitis and the Common Terminology Criteria for Adverse Events grade ≥3 immune-related adverse events, were reviewed.
 
Results
 
A total of 113 cases were analysed (51 cases of durvalumab plus CRT and 62 cases of CRT). The durvalumab plus CRT cohort demonstrated a significantly longer median PFS compared to the CRT cohort (34.9 vs. 10.5 months; p = 0.01), while median overall survival remained immature at the time of analysis. Among patients with epidermal growth factor receptor (EGFR) mutations, the estimated PFS also favoured the durvalumab plus CRT cohort. A significantly higher incidence of any-grade pneumonitis was observed in the durvalumab plus CRT cohort (31% vs. 8%; p = 0.002), with most cases occurring within the initial 3 months of durvalumab use.
 
Conclusion
 
Durvalumab following CRT significantly benefitted patients with unresectable stage III NSCLC, including those with EGFR mutations. Symptomatic pneumonitis tended to occur in the first 3 months of durvalumab therapy and was generally manageable. Close follow-up during this period is recommended to facilitate early detection and intervention. Further research is warranted to understand the complex interplay among EGFR mutation status, programmed death ligand 1 expression, and treatment outcomes with and without durvalumab in NSCLC.
 
 
Key Words: Carcinoma, non–small-cell lung; Chemoradiotherapy; ErbB receptors; Progression-free survival
 
 
中文摘要
 
無法切除的第三期非小細胞肺癌患者在同步化學放射治療後接受度伐魯單抗治療的療效分析
 
梁詩雅、林美瑩、劉芷珊
 
引言
本研究旨在評估香港一所三級醫院針對無法切除的第三期非小細胞肺癌患者採用度伐魯單抗作為鞏固治療的效益及安全性。
 
方法
本研究回溯性納入於2017年12月至2023年6月期間接受根治性同步化學放射治療(放化療)的無法切除第三期非小細胞肺癌患者,依據後續是否接受度伐魯單抗治療分組。我們使用Kaplan-Meier法分析疾病無惡化存活期及整體存活期,並系統性評估不良事件,涵蓋各級別非感染性肺炎及符合「常見不良事件評價標準」【CTCAE】第3級或以上免疫相關不良反應。
 
結果
本研究共分析了113例患者,包括51例放化療合併度伐魯單抗及62例僅接受放化療。放化療合併度伐魯單抗組的疾病無惡化存活期中位數顯著較單獨放化療組長(34.9個月與10.5個月;p = 0.01),整體存活期中位數則在分析時尚未成熟。在表皮生長因子受體(EGFR)基因突變患者中,放化療合併度伐魯單抗組也呈現較長的預估疾病無惡化存活期。安全性方面,放化療合併度伐魯單抗組在非感染性肺炎總發生率顯著較高(31%與8%;p = 0.002),且多數病例集中於治療起始3個月內發生。
 
結論
同步放化療後接續度伐魯單抗治療對於第三期非小細胞肺癌患者(包括EGFR基因突變患者)具顯著臨床效益。症狀性肺炎雖易於治療初期首3個月出現,但整體可控。我們建議在此段期間密集隨訪,以監察非感染性肺炎的早期徵狀。EGFR基因突變狀態、細胞程式死亡─配體1(PD-L1)表現量及度伐魯單抗的治療效益存在複雜相互作用,有待未來研究作進一步釐清。
 
 
 
INTRODUCTION
 
The PACIFIC trial[1] showed that 1 year of durvalumab consolidation therapy following chemoradiotherapy (CRT) significantly improves the progression-free survival (PFS) and overall survival (OS) in unresectable stage III non–small-cell lung cancer (NSCLC), with a median PFS of 16.9 months and OS of 47.5 months.[2] The PACIFIC-R study[3] substantiated these findings, suggesting that real-world outcomes align with the drug’s registration trial results.[4]
 
In Hong Kong, durvalumab has been a registered drug since October 2018 and included in the Community Care Fund Medical Assistance Programme since May 2020.[5] Given the emerging concern that patients harbouring epidermal growth factor receptor (EGFR) mutations may derive less benefit from immune checkpoint inhibitors, including maintenance durvalumab, studies have been conducted to review the outcomes in this subgroup.[6] [7] [8] Pneumonitis, a major adverse event associated with durvalumab, is of particular concern in patients who have undergone thoracic radiotherapy.
 
This study aimed to evaluate the real-world efficacy and safety of durvalumab in unresectable stage III NSCLC in a population with a high prevalence of EGFR mutations and to assess pneumonitis incidence relative to radiation dose, enabling early toxicity detection and optimising follow-up protocols to ensure that local patients achieve maximal therapeutic benefit with minimised risks.
 
METHODS
 
Inclusion Criteria and Data Collection
 
This retrospective study included patients with stage III NSCLC who were treated with chemoradiotherapy (CRT) between December 2017 and June 2023 in Princess Margaret Hospital, Hong Kong. The durvalumab cohort was drawn from the Clinical Data Analysis and Reporting System of Hospital Authority, comprising all patients who received durvalumab during the specified period. The CRT cohort—patients who received CRT only—was drawn from our department’s ARIA Oncology Information System. Each case was screened via the Electronic Patient Record system for eligibility. Inclusion criteria were adult patients aged ≥18 years, diagnosed with stage III NSCLC and treated with CRT with curative intent. All patients were restaged using the 8th edition of the American Joint Commission on Cancer TNM (tumour, node and metastasis) Classification.[9] Patients who had commenced treatment in other centres must have received at least one dose of durvalumab in our hospital to be included in the analysis. Cases of proven disease progression within 2 months of CRT completion were excluded. Patient and disease demographics, details of chemoradiotherapy treatment regimens, and response to treatment were documented. Treatment-related toxicities were graded according to the CTCAE (Common Terminology Criteria for Adverse Events) version 5.0.[10]
 
Treatment and Follow-up
 
Standard radical-intent CRT in the stage III NSCLC study population involved three-dimensional conformal radiotherapy of 60 to 66 Gy at 2 Gy per fraction, typically paired with etoposide/cisplatin for two cycles once every 3 weeks. For non-squamous cases, pemetrexed/cisplatin was an alternative, especially for patients with poor venous access or concerns about tolerance. Patients unsuitable for cisplatin (e.g., creatinine clearance <50 mL/min or congestive heart failure) received weekly paclitaxel/carboplatin. Induction chemotherapy was planned on a case-by-case basis. Optimal organs-at-risk dose constraints were: (1) the percentage of lung receiving ≥20 Gy (lung V20Gy) ≤30%; (2) lung V5Gy ≤55%; and (3) mean lung dose (MLD) ≤15 Gy.
 
Durvalumab consolidation was offered to eligible patients without progression after CRT as self-funded treatment since October 2018, or with financial assistance from the Community Care Fund for those with programmed death ligand 1 (PD-L1) expression of tumour proportion score ≥1% since May 2020.[5] Durvalumab at 10 mg/kg biweekly for up to 12 months was usually started within 42 days post-radiotherapy, though this was not mandatory. Pre-cycle chest radiographs (CXR) and laboratory tests, including complete blood count, liver/renal/thyroid function, cortisol level, and fasting glucose level were taken to monitor for adverse events. Post-treatment, patients were followed up every 4 to 6 months with CXR, and carcinoembryonic antigen was also measured in cases of adenocarcinoma. Computed tomography scans were performed subject to availability and clinical judgement.
 
Statistical Analyses
 
Baseline characteristics and dosimetric parameters of the two cohorts were compared using Chi squared or Fisher’s exact tests. PFS and OS were measured from the last day of radiotherapy to disease progression or death. The data cut-off was 15 June 2024. The Kaplan-Meier method was utilised to estimate PFS and OS. Subgroup analysis explored outcomes in EGFR-mutated (EGFRm) and EGFR–wild-type (EGFRwt) patients. Univariate logistic regression analysis was employed to evaluate any significant predictive factors (clinical or dosimetric) for the incidence of any-grade pneumonitis, with only significant univariate factors further analysed by multivariate analysis. Statistical analyses were conducted using commercial software SPSS (Windows version 29.0; IBM Corp, Armonk [NY], US), each with a significance level of 0.05. For missing data, a listwise deletion approach was employed to analyse cases with complete data only. Receiver operating characteristic (ROC) analysis was conducted as an exploratory measure to identify an optimal cut-off value for lung V20Gy associated with pneumonitis occurrence.
 
RESULTS
 
Patient Characteristics
 
This study included 113 cases, with 51 in the durvalumab plus CRT cohort and 62 in the CRT cohort (Table 1). Both cohorts had a predominance of male and smoker/ex-smoker patients. The median ages were 65 years and 66.5 years in the durvalumab plus CRT and CRT cohorts, respectively. Baseline characteristics were similar, except for a higher proportion of patients with no PD-L1 expression in the CRT cohort compared with the durvalumab plus CRT cohort. Histology was mainly adenocarcinoma (41% in the durvalumab plus CRT cohort and 48% in the CRT cohort) and squamous cell carcinoma (27% and 37%, respectively). NSCLC of no specific type was reported in 27% of the durvalumab plus CRT cohort and 6% of the CRT cohort. Approximately 70% of patients had their EGFR status tested; 7 (14%) and 15 (24%) patients in the durvalumab plus CRT and CRT cohorts, respectively, were confirmed as EGFRm. Commonly used CRT chemotherapy regimens were etoposide/platinum (37% and 48%), paclitaxel/carboplatin (35% and 37%), and pemetrexed/platinum (12% and 10%) in the durvalumab plus CRT and CRT cohorts, respectively. The median duration from CRT completion to durvalumab initiation was 45 days (range, 8-172); 59% (n = 30) of patients completed the planned 26 cycles of biweekly durvalumab (median: 13.8 months). Treatment discontinuation was attributed to disease progression, adverse events, patient decision, or death (Table 2).
 
Table 1. Baseline characteristics (n = 113).
 
Table 2. Reasons for and timing of durvalumab discontinuation (n = 51).
 
At the time of analysis, all patients in the durvalumab plus CRT cohort had either discontinued or completed 26 cycles of durvalumab consolidation treatment. 94% and 95% patients in the durvalumab plus CRT and CRT cohorts, respectively, had completed CRT, defined as either having received chemotherapy once every 3 weeks for 2 cycles or a concurrent regimen once a week for 5 cycles. All patients, except for one treated in the private sector with missing data, received a radical dose of at least 60 Gy (equivalent dose in 2 Gy fractions).
 
Efficacy Outcomes
 
The median follow-up was 25.6 months for the durvalumab plus CRT cohort and 31.0 months for the CRT cohort. The median PFS was significantly longer in the durvalumab plus CRT cohort, at 34.9 months (95% confidence interval [CI] = 17.8-52.0) compared to 10.5 months (95% CI = 7.1-14.0) in the CRT cohort (p = 0.01) [Figure 1]. The median OS was 50.8 months (95% CI = 26.6-75.0) in the durvalumab plus CRT cohort and 41.5 months (95% CI = 22.2-60.7) in the CRT cohort, which was not statistically significant (p = 0.32) [Figure 2].
 
Figure 1. Progression-free survival.
 
Figure 2. Overall survival.
 
The estimated PFS for EGFRm patients was not reached in the durvalumab plus CRT cohort, compared to 7.8 months (95% CI = 3.4-12.1) in the CRT cohort. OS analysis was not performed due to the limited number of events (one in the durvalumab plus CRT cohort and 8 in the CRT cohort). Notably, all EGFRm patients in the durvalumab plus CRT cohort had either unknown or low PD-L1 expression, while those in the CRT cohort had either unknown or negative PD-L1 expression. No EGFRm patients had high PD-L1 expression.
 
Pneumonitis
 
A significantly higher incidence of any-grade pneumonitis was observed in the durvalumab plus CRT cohort compared to the CRT cohort (31% vs. 8%; p = 0.002) [Table 3]. In total, 57% of EGFRm patients and 27% of EGFRwt/EGFR-unknown patients in the durvalumab plus CRT cohort developed pneumonitis, compared to 0% and 10%, respectively, in the CRT cohort. Of the 16 patients in the durvalumab plus CRT cohort who developed pneumonitis, the majority (87.5%) experienced their first episode during the initial six biweekly cycles (range, 2-12). Approximately 80% of cases were grade 1 to 2 and responded to appropriate management strategies including corticosteroids, except one grade 4 pneumonitis (Table 4). Overall, 12% discontinued durvalumab treatment due to pneumonitis. In the CRT cohort, five patients (8%) developed any grade of radiation pneumonitis (RP), with onset ranging from 6 to 91 days after the last day of radiotherapy. All improved clinically after a course of steroids.
 
Table 3. Occurrence of pneumonitis in the two cohorts.
 
Table 4. Detailed account of pneumonitis occurrence in the durvalumab plus chemoradiotherapy cohort.
 
The single case of grade 4 pneumonitis in the durvalumab plus CRT cohort was a patient with a history of rectal and hepatocellular carcinoma in remission, who was diagnosed with a third primary, T4N0 poorly differentiated NSCLC with focal squamous differentiation. The patient received two cycles of induction 3-weekly paclitaxel/carboplatin followed by CRT and subsequently three weekly cycles of paclitaxel/carboplatin due to neutropenia and thrombocytopenia. A computed tomography scan performed 1 day after CRT completion showed stable disease, leading to durvalumab initiation on day 22. He developed grade 2 pneumonitis before cycle 4 of durvalumab, leading to treatment suspension and initiation of a 1-month tapering course of prednisolone at 1 mg/kg. After radiological and clinical improvement, cycle 4 of durvalumab was resumed 43 days after its original planned date. Seven days later, he was admitted for respiratory failure requiring high-flow oxygen. Intravenous methylprednisolone 2 mg/kg was administered for 5 days, but there was further consolidation on CXR treated with one dose of infliximab on day 6. He was subsequently transitioned to oral prednisolone on day 54, with clinical improvement and reduced oxygen requirement. He became deconditioned 3 months later after steroid weaning, developing brain metastases and hospital-acquired pneumonia, and succumbed after 140 days of hospitalisation.
 
No significant differences were observed in lung V5Gy, lung V20Gy, MLD, or planning target volume between the two cohorts (Table 5). Among these parameters, only lung V20Gy demonstrated a significant correlation with any grade pneumonitis in univariate logistic analysis, with an odds ratio of 1.11 (95% CI = 1.013-1.213; p = 0.03), indicating that for each 1% increase in the volume of lung receiving ≥20 Gy, the odds of developing pneumonitis increased by approximately 11% (Table 6). Focusing on the durvalumab plus CRT cohort, ROC analysis identified an optimal lung V20Gy threshold of 22.76% for predicting pneumonitis, with a Youden’s index of 0.469, optimising sensitivity (0.92) and specificity (0.46). The area under the curve of the ROC analysis was 0.71, indicating moderate discriminatory power.
 
Table 5. Radiation dosimetry of radical chemoradiotherapy.
 
Table 6. Univariate analysis for predictive factors of pneumonitis.
 
Grade 3 or 4 Immunotherapy-Related Adverse Events Within the Durvalumab Cohort
 
The overall incidence of grade 3 or 4 immune-related adverse events was 13.7% (7/51). Three patients (6%) had RP, with one concurrently developing grade 3 hepatitis after 6 cycles that resolved over 2 months of corticosteroid treatment. For the remaining four patients, two (3.9%) developed grade 3 hyperglycaemia without a baseline history of diabetes, one (2%) experienced grade 3 skin rash after 17 cycles of durvalumab, and one (2%) developed grade 3 pneumonia. No patients discontinued durvalumab due to adverse events other than pneumonitis.
 
DISCUSSION
 
Our durvalumab plus CRT cohort demonstrated superior PFS to the CRT cohort, consistent with findings from the PACIFIC trial and real-world studies.[2] [11] [12] [13] The disparity in median follow-up times between the CRT cohort (31 months) and the durvalumab plus CRT cohort (25.6 months) may be attributed to delayed availability of durvalumab funding, resulting in more patients receiving CRT alone between 2018 to 2020. This complicates PFS and OS interpretation, especially with the survival curve of the durvalumab plus CRT cohort plateauing.
 
Our mean PFS durations of 34.9 months (the durvalumab plus CRT cohort) and 10.5 months (the CRT cohort) exceeded those of the PACIFIC trial results,[2] nearly doubling their reported numbers. While real-world follow-up variability might underestimate early progression, prognostic advantages in our cohort likely contributed. These included a higher proportion with Eastern Cooperative Oncology Group performance status score of 0 (88% vs. 50% in the PACIFIC trial[14]) and more never-smokers (25% vs. 9%). PD-L1 status showed dual roles: in the CRT cohort, the higher proportion of PD-L1-negative patients (~50%) aligns with its known favourable prognostic value in the pre-immunotherapy era, supported by multiple meta-analyses.[15] [16] [17] [18] Conversely, the PD-L1–enriched population in the durvalumab plus CRT cohort (~90% positive, ~50% with ≥ 50% expression) reflect its predictive value, consistent with the PACIFIC subgroup analysis showing enhanced immunotherapy benefit with higher PD-L1 expression.[2] Additionally, approximately half of the cohort received at least one cycle of induction chemotherapy, compared to only a quarter in the PACIFIC trial.[2] Any potentiation of immunotherapy with induction chemotherapy, through neoantigen release and tumour microenvironment modulation, is a theoretical consideration. Further elucidation, however, is required to determine the application of PD-L1 for risk stratification and to optimise treatment sequencing and combination, including toxicity risks.[19] [20]
 
The incidence of any-grade pneumonitis in our durvalumab plus CRT cohort (31%) was similar to the figure reported in the PACIFIC study (34%),[2] where it was the most common adverse event leading to treatment discontinuation (6.3%).[2] It was higher than in the CRT cohort (8%), though the majority (~80%) were grade 1 to 2 per the CTCAE version 5.0 criteria.[10] Differentiating between immunotherapy-induced pneumonitis (IP) and RP, especially in the early cycles, proved challenging. Radiologically, RP is more likely if the consolidative changes are seen only within the irradiated field. Observation from our study reinforced this diagnostic difficulty as the majority of events occurred within the first 3 months in both groups (87.5% in the durvalumab plus CRT cohort vs. all within 91 days in the CRT cohort). This aligns with other studies reporting median pneumonitis onset around 3 to 4 months,[21] [22] emphasising the importance of close monitoring during early durvalumab treatment.
 
Fortunately, treatment is mostly similar for both conditions with corticosteroids as the mainstay, although IP may require longer treatment. In cases of steroid-refractory IP, immunosuppressive agents such as mycophenolate mofetil or infliximab can be considered.[23] Supportive management such as symptom-relieving medications and oxygen support should always be given where clinically indicated. Vigilance for concomitant infection due to the immunosuppressive effects of the cancer treatments and high-dose steroids is also essential. The decision to rechallenge with durvalumab after resolution of low-grade pneumonitis should be made after ensuring patients are well informed of recurrent or higher-grade pneumonitis risks. Among patients in the durvalumab plus CRT cohort who developed pneumonitis, 31% experienced recurrence of events after treatment resumption. Overall, 12% discontinued durvalumab due to pneumonitis, similar to the reported 9.5% in the PACIFIC-R study.[4]
 
There is no doubt that RP could compromise patients’ outcomes and quality of life, therefore continuous efforts have been put to identify any clinical and dosimetric factors that are predictive and/or preventive. Lung V20Gy is the most representative among the commonly reported parameters. However, it is uncertain whether the traditional dose constraints used in CRT are equally applicable to patients also receiving immunotherapy. In our cohort, lung V20Gy was the only radiation dose parameter that correlated with pneumonitis, with an optimal threshold at 22.76% based on ROC analysis. However, the low specificity (0.46) suggests that lung V20Gy alone is not a strong predictor due to its high false positive rate. Of note, this threshold is lower than the commonly reported 30% for normofractionated thoracic radiotherapy in the preimmunotherapy era. Even lower thresholds, such as 18.77% in a Japanese study[21] and 15.8% in the Mayo Clinic, have been proposed for predicting grade≥2 pneumonitis.[22] All these highlight a change in regulation of immune and/or lung homeostasis after exposure to immunotherapy and radiotherapy; this could possibly lead to different lung parenchymal susceptibilities. The high incidence of any-grade (88%) and grade ≥3 pneumonitis (12%) in the Japanese study involving 91 patients,[21] and Asian predominance in pneumonitis after CRT with or without immunotherapy in a recent meta-analysis over 20,000 patients[24] and in the PACIFIC subgroup analysis[25] raise further research questions with regard to any ethnic and/or genetic contributing factors. Although direct comparison across trials to derive the optimal dose constraint is not possible due to varying radiotherapy planning techniques, chemotherapy regimens, and patient factors, efforts to reduce the lung V20Gy to as low as possible are reasonable.
 
Practically, applying more stringent lung dose constraints while maintaining target coverage in radiotherapy planning for stage III NSCLC, where tumours are often bulky, is challenging. Advanced technology, including intensity-modulated radiation therapy and proton therapy, may offer benefits over conventional techniques.[26] However, uncertainty remains regarding any interplay between low radiation exposure (e.g., lung V5Gy and MLD) and immunotherapy in modulating pneumonitis risk. Moreover, the labour-intensive nature of planning and treatment delivery warrants careful patient selection, especially in high-workload or resource-limited settings.
 
In addition to pneumonitis, our study also examined all-cause immune-related grade 3 or 4 adverse events. The incidence in our cohort (13.7% grade 3 and 3% grade 4) were higher than in the PACIFIC trial (3.4% in the durvalumab plus CRT cohort and 2.6% in the CRT group),[2] but a solid conclusion on differences in safety cannot be made due to the small sample size and variable documentation of our study. Reassuringly, a similar proportion of patients required durvalumab discontinuation due to adverse events (12% in our study vs. 15.4% in the PACIFIC trial).[2] This underpins the fact that adequate patient education together with team-based engagement remain the key to ensuring timely recognition and effective management of immune-related adverse events.
 
When focusing on EGFRm patients, the estimated PFS was not reached in the durvalumab plus CRT cohort, compared with 7.8 months in the CRT cohort, suggesting a potential benefit of adjuvant durvalumab. This contrasts with the lack of benefit in the post-hoc analysis of EGFRm subgroups in the PACIFIC trial[6] and another retrospective review involving multiple academic medical centres in the US.[7] However, caution should be exercised when interpreting these results due to the small sample size, the low treatment completion rates (15%-50%) reported in the abovementioned studies, and the short follow-up interval of our study.
 
Another notable observation from our durvalumab plus CRT cohort is the higher occurrence of pneumonitis in EGFRm patients (57%) compared to EGFRwt/unknown patients (27%), though the difference was not statistically significant. While the exact mechanism underlying this difference remains unknown, this observation carries important clinical implications as initiating EGFR tyrosine kinase inhibitors (TKIs) after CRT is a relatively common post-radical treatment for EGFRm stage III disease due to the high risk of progression. There is already growing recognition of the increased risk of pneumonitis with sequential immunotherapy followed by early TKI treatment.[27] Prior RP and IP may exacerbate this risk through increased lung tissue sensitivity, cumulative lung injuries, and/or shared mechanisms such as immune response dysregulation. Although none of our three EGFRm patients who received erlotinib immediately upon disease progression during durvalumab plus CRT treatment developed pneumonitis, this should not over-reassure clinicians given the safety alert reported in other studies[28] [29] when using immunotherapy and TKIs in close intervals. Optimal timing to guide safe use of immunotherapy and TKIs is undefined, but the premature terminations of the TATTON[28] [30] and CAURAL trials[29] [31] due to the higher incidence of interstitial lung disease—like events with osimertinib and durvalumab provided important information, leading to the consensus that concurrent use should be avoided outside clinical trials. Common practice to reduce pneumonitis risk is to defer the TKI initiation for at least 1 month, preferably 3 months for less aggressive diseases, after the last use of immunotherapy.[7] [32] Extra caution is needed with the third-generation TKI osimertinib compared to first- or second-generation TKIs, especially in patients with preexisting lung injuries.[32]
 
Limitations
 
Limitations of our study included small sample size, variable follow-up, and assessment tools, leading to inconsistent evaluations of efficacy and toxicities. The unexpectedly low EGFR mutation rate (~30%) among those tested makes it challenging to draw statistically significant conclusions about the benefits for the controversial EGFRm subgroup, despite an observed improvement in PFS. Retrospective EGFR analysis of the 37 untested cases could enhance understanding, though further EGFR population enrichment may be limited due to the expected low mutation rates based on histology[33] (70.3% squamous, 8.1% lymphoepithelioma-like carcinoma, 5.4% large cell, and 13.5% NSCLC of no specific type). The imbalance and deviation in PD-L1 expression pattern, probably due to small sample size, may also confound results. Collaborative multi-centre analysis, adoption of universal EGFR testing for non-squamous NSCLC, and increased accessibility of PD-L1 test in Hong Kong oncology centres could enhance the statistical value of future similar studies by reducing the untested population and increasing the overall sample size.
 
CONCLUSION
 
This study provides compelling evidence that durvalumab consolidation therapy following CRT improves PFS in unresectable stage III NSCLC, with manageable adverse effects. Pneumonitis, occurring mainly within the first 3 months, underscores the need for close monitoring and timely management, especially at the start of durvalumab. Lung V20Gy may predict pneumonitis and should be kept as low as possible after balancing a reasonable target coverage, but its low specificity suggests it should be used alongside other clinical factors for individual risk assessment and planning.
 
As the treatment landscape for locally advanced NSCLC is evolving, therapies effective in metastatic disease are applied earlier in the treatment pathway. The recently published LAURA study,[34] [35] which demonstrated a highly encouraging PFS benefit from 5.6 months to 39.1 months with adjuvant osimertinib in EGFRm patients, is probably just the start. With increasing evidence, both PD-L1 and EGFR status are expected to be critical in the near future to guide treatment selection. Further large-scale studies and uniform follow-up are needed to validate the roles of different biomarkers in tailoring treatments for patients with unresectable stage III NSCLC, similar to the approach in stage IV disease.
 
REFERENCES
 
1. ClinicalTrials.gov. A Global Study to Assess the Effects of MEDI4736 Following Concurrent Chemoradiation in Patients With Stage III Unresectable Non-Small Cell Lung Cancer (PACIFIC). Available from: https://clinicaltrials.gov/study/NCT02125461. Accessed 21 May 2025.
 
2. Spigel DR, Faivre-Finn C, Gray JE, Vicente D, Planchard D, Paz-Ares L, et al. Five-year survival outcomes from the PACIFIC trial: durvalumab after chemoradiotherapy in stage III non–small-cell lung cancer. J Clin Oncol. 2022;40:1301-11. Crossref
 
3. ClinicalTrials.gov. First Real-world Data on Unresectable Stage III NSCLC Patients Treated With Durvalumab After Chemoradiotherapy. Available from: https://clinicaltrials.gov/study/NCT03798535?term=NCT03798535&rank=1. Accessed 21 May 2025.
 
4. Girard N, Bar J, Garrido P, Garassino MC, McDonald F, Mornex F, et al. Treatment characteristics and real-world progression-free survival in patients with unresectable stage III NSCLC who received durvalumab after chemoradiotherapy: findings from the PACIFIC-R study. J Thorac Oncol. 2023;18:181-93. Crossref
 
5. Hong Kong SAR Government. LCQ15: Treatments and support for patients with cancers and rare diseases. 2020 Jun 24. Available from: https://www.info.gov.hk/gia/general/202006/24/P2020062400288.htm. Accessed 23 Jun 2024.
 
6. Naidoo J, Antonia S, Wu YL, Cho BC, Thiyagarajah P, Mann H, et al. Brief report: durvalumab after chemoradiotherapy in unresectable stage III EGFR-mutant NSCLC: a post hoc subgroup analysis from PACIFIC. J Thorac Oncol. 2023;18:657-63. Crossref
 
7. Aredo JV, Mambetsariev I, Hellyer JA, Amini A, Neal JW, Padda SK, et al. Durvalumab for stage III EGFR-mutated NSCLC after definitive chemoradiotherapy. J Thorac Oncol. 2021;16:1030-41. Crossref
 
8. Yang H, Zhu J, Xiao R, Liu Y, Yu F, Cai L, et al. EGFR mutation status in non–small-cell lung cancer receiving PD-1/PD-L1 inhibitors and its correlation with PD-L1 expression: a meta-analysis. Cancer Immunol Immunother. 2022;71:1001-16. Crossref
 
9. Detterbeck FC. The eighth edition TNM stage classification for lung cancer: what does it mean on main street? J Thorac Cardiovasc Surg. 2018;155:356-9. Crossref
 
10. United States Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. 2017 Nov 27. Available from: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_5x7.pdf. Accessed 4 Aug 2024.
 
11. Jung HA, Noh JM, Sun JM, Lee SH, Ahn JS, Ahn MJ, et al. Real-world data of durvalumab consolidation after chemoradiotherapy in stage III non–small-cell lung cancer. Lung Cancer. 2020;146:23-9. Crossref
 
12. Park CK, Oh HJ, Kim YC, Kim YH, Ahn SJ, Jeong WG, et al. Korean real-world data on patients with unresectable stage III NSCLC treated with durvalumab after chemoradiotherapy: PACIFIC-KR. J Thorac Oncol. 2023;18:1042-54. Crossref
 
13. Preti BT, Sanatani MS, Breadner D, Lakkunarajah S, Scott C, Esmonde-White C, et al. Real-world analysis of durvalumab after chemoradiation in stage III non–small-cell lung cancer. Curr Oncol. 2023;30:7713-21. Crossref
 
14. Kawaguchi T, Takada M, Kubo A, Matsumura A, Fukai S, Tamura A, et al. Performance status and smoking status are independent favorable prognostic factors for survival in non–small-cell lung cancer: a comprehensive analysis of 26,957 patients with NSCLC. J Thorac Oncol. 2010;5:620-30. Crossref
 
15. Tuminello S, Sikavi D, Veluswamy R, Gamarra C, Lieberman-Cribbin W, Flores R, et al. PD-L1 as a prognostic biomarker in surgically resectable non–small-cell lung cancer: a meta-analysis. Transl Lung Cancer Res. 2020;9:1343-60. Crossref
 
16. Vrankar M, Zwitter M, Kern I, Stanic K. PD-L1 expression can be regarded as prognostic factor for survival of non–small-cell lung cancer patients after chemoradiotherapy. Neoplasma. 2018;65:140-6. Crossref
 
17. Zhao Y, Shi F, Zhou Q, Li Y, Wu J, Wang R, et al. Prognostic significance of PD-L1 in advanced non–small-cell lung carcinoma. Medicine (Baltimore). 2020;99:e23172. Crossref
 
18. Zhou ZJ, Zhan P, Song Y. PD-L1 over-expression and survival in patients with non–small-cell lung cancer: a meta-analysis. Transl Lung Cancer Res. 2015;4:203-8. Crossref
 
19. Sordo-Bahamonde C, Lorenzo-Herrero S, Gonzalez-Rodriguez AP, Martínez-Pérez A, Rodrigo JP, García-Pedrero JM, et al. Chemo-immunotherapy: a new trend in cancer treatment. Cancers (Basel). 2023;15:2912. Crossref
 
20. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res. 2015;3:436-43. Crossref
 
21. Oshiro Y, Mizumoto M, Sekino Y, Maruo K, Ishida T, Sumiya T, et al. Risk factor of pneumonitis on dose-volume relationship for chemoradiotherapy with durvalumab: multi-institutional research in Japan. Clin Transl Radiat Oncol. 2021;29:54-9. Crossref
 
22. Gao RW, Day CN, Yu NY, Bush A, Amundson AC, Prodduturvar P, et al. Dosimetric predictors of pneumonitis in locally advanced non–small-cell lung cancer patients treated with chemoradiation followed by durvalumab. Lung Cancer. 2022;170:58-64. Crossref
 
23. 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. Crossref
 
24. Liu T, Li S, Ding S, Qiu J, Ren C, Chen J, et al. Comparison of post-chemoradiotherapy pneumonitis between Asian and non-Asian patients with locally advanced non–small-cell lung cancer: a systematic review and meta-analysis. EClinicalMedicine. 2023;64:102246. Crossref
 
25. Faehling M, Schulz C, Laack H, Wolff T, Rückert A, Reck M, et al. PACIFIC subgroup analysis: pneumonitis in stage III, unresectable NSCLC patients treated with durvalumab vs. placebo after CRT. Pneumologie. 2019;73(S 01):s-0039-1678247. Crossref
 
26. Chun SG, Hu C, Choy H, Komaki RU, Timmerman RD, Schild SE, et al. Impact of intensity-modulated radiation therapy technique for locally advanced non–small-cell lung cancer: a secondary analysis of the NRG oncology RTOG 0617 randomized clinical trial. J Clin Oncol. 2017;35:56-62. Crossref
 
27. Jung J, Kim HY, Kim DG, Park SY, Ko AR, Han JY, et al. Sequential treatment with an immune checkpoint inhibitor followed by a small-molecule targeted agent increases drug-induced pneumonitis. Cancer Res Treat. 2021;53:77-86. Crossref
 
28. Ahn MJ, Cho BC, Ou X, Walding A, Dymond AW, Ren S, et al. Osimertinib plus durvalumab in patients with EGFR-mutated, advanced NSCLC: a phase 1b, open-label, multicenter trial. J Thorac Oncol. 2022;17:718-23. Crossref
 
29. Yang JC, Shepherd FA, Kim DW, Lee GW, Lee JS, Chang GC, et al. Osimertinib plus durvalumab versus osimertinib monotherapy in EGFR T790M–positive NSCLC following previous EGFR TKI therapy: CAURAL brief report. J Thorac Oncol. 2019;14:933-9. Crossref
 
30. ClinicalTrials.gov. AZD9291 in Combination With Ascending Doses of Novel Therapeutics. Available from: https://clinicaltrials.gov/study/NCT02143466. Accessed 21 May 2025.
 
31. ClinicalTrials.gov. Study of AZD9291 Plus MEDI4736 Versus AZD9291 Monotherapy in NSCLC After Previous EGFR TKI Therapy in T790M Mutation Positive Tumours (CAURAL). Available from: https://clinicaltrials.gov/study/NCT02454933. Accessed 21 May 2025.
 
32. Kalra A, Rashdan S. The toxicity associated with combining immune check point inhibitors with tyrosine kinase inhibitors in patients with non–small-cell lung cancer. Front Oncol. 2023;13:1158417. Crossref
 
33. Joshi A, Zanwar S, Noronha V, Patil VM, Chougule A, Kumar R, et al. EGFR mutation in squamous cell carcinoma of the lung: does it carry the same connotation as in adenocarcinomas? Onco Targets Ther. 2017;10:1859-63. Crossref
 
34. Lu S, Kato T, Dong X, Ahn MJ, Quang LV, Soparattanapaisarn N, et al. Osimertinib after chemoradiotherapy in stage III EGFR-mutated NSCLC. N Engl J Med. 2024;391:585-97. Crossref
 
35. ClinicalTrials.gov. A Global Study to Assess the Effects of Osimertinib Following Chemoradiation in Patients With Stage III Unresectable Non-small Cell Lung Cancer (LAURA). Available from: https://clinicaltrials.gov/study/NCT03521154. Accessed 21 May 2025.
 
 
 
CASE REPORTS

Endovascular Management of Renal Arteriovenous Fistula: Three Case Reports

JK Fung, HK Chin, WKW Leung, KYK Tang, CY Chu, WK Kan

CASE REPORT
 
Endovascular Management of Renal Arteriovenous Fistula: Three Case Reports
 
JK Fung, HK Chin, WKW Leung, KYK Tang, CY Chu, WK Kan
Department of Radiology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China
 
Correspondence: Dr JK Fung, Department of Radiology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China. Email:
 
Submitted: 2 September 2023; Accepted: 18 July 2024.
 
Contributors: All authors designed the study. HKC, KYKT and CYC acquired the data. JKF, HKC, WKWL, KYKT and CYC analysed the data. JKF, HKC and WKWL drafted the manuscript. JKF, HKC, WKWL, KYKT and WKK critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: The study was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: CIRB-2024-080-5). The requirement for informed patient consent was waived by the Board due to the retrospective nature of the study.
 
 
 
 
INTRODUCTION
 
Renal arteriovenous fistula (AVF) is a rare vascular anomaly classified as traumatic or non-traumatic. There are no guidelines for endovascular treatment. Some case reports involve coil deployment[1] [2] but some require additional techniques such as vascular plugs, occlusive balloons, or stents[3] [4] to minimise the risk of coil embolisation.
 
We report three cases of renal AVF endovascular treatment, including two idiopathic AVFs, and focus on treatment considerations and technical perspectives with reference to current reported practices.
 
CASE PRESENTATIONS
 
Case 1
 
A 68-year-old female presented with haematuria. Elective computed tomography (CT) urogram demonstrated an AVF in the left kidney at the mid to lower pole. It was supplied by a hypertrophied renal artery, drained by a dilated renal vein and via the engorged inferior vena cava into the enlarged right atrium (Figure 1a and c). There were two relatively sizeable saccular aneurysms connected by a short stenotic segment (Figure 1b). The case was discussed with the vascular team and deemed unsuitable for endovascular stenting.
 
Figure 1. Case 1. Corticomedullary-phase contrast-enhanced computed tomography with maximal intensity projection in oblique sagittal (a) and axial (b, c) planes. (a) Hypertrophied supplying left renal artery (Art) and early opacification of the dilated draining renal vein (Vein). (b) Two saccular aneurysms (asterisk and hash) were connected by a stenotic segment (arrows). (c) Dilated draining renal vein (Vein) and inferior vena cava (IVC).
 
Digital subtraction angiography (DSA) confirmed the AVF was supplied by the main trunk of the left renal artery. The most proximal aneurysm was the largest at 3.5 cm (Figure 2a). Two 6-Fr guiding sheaths (Flexor Ansel; Cook Medical, Bloomington [IN], US) were advanced to the left main renal artery via a femoral approach. A 0.035-inch balloon catheter (Mustang [10 × 20 mm]; Boston Scientific, Marlborough [MA], US) was then directed to the proximal left main renal artery to control arterial inflow. Detachable coils (standard Ruby coils; Penumbra Inc, Alameda [CA], US) of various sizes and lengths were deployed into the most proximal aneurysm using the scaffold technique via a dedicated microcatheter (Excelsior XT-27; Stryker, Kalamazoo [MI], US) [Figure 2b]. The feeding left renal artery was eventually packed with detachable coils (0.035-inch Interlock; Boston Scientific, Marlborough [MA], US). Follow-up magnetic resonance (MR) renal angiogram 6 months later revealed significantly reduced vascularity in the dilated vessels and aneurysms (Figure 2c).
 
Figure 2. Case 1. Left renal digital subtraction angiography (DSA). (a) Pre-embolisation DSA shows the two aneurysms detected on computed tomography (asterisk and hash). The hypertrophied left renal artery (white arrow) measures 1 cm. (b) Application and packing with Ruby coils of various lengths into the more proximal aneurysm (asterisk). The stenotic segment allowed successful anchorage of the coils. The Mustang catheter balloon (black arrow) was inflated during coil deployment to reduce arterial inflow and prevent coil migration. (c) Check DSA confirmed significantly reduced vascularity across the arteriovenous fistula.
 
Case 2
 
A 49-year-old female presented with gross haematuria and loss of consciousness. Haemoglobin level was 7.8 g/dL on admission. CT urogram demonstrated a right AVF centred at the interpolar region, with acute blood products dilating the right renal collecting system and the right ureter (Figure 3).
 
Figure 3. Case 2. Corticomedullary-phase contrast-enhanced computed tomography with maximal intensity projection in axial (a) and coronal (b) planes. Delayed nephrogram in both projections. Tortuous vascular structures with hyperenhancement are seen at the anterior interpolar region of the right kidney (arrows). The renal pelvis (P) and the right ureter (U) were dilated by hyperdense blood products. The right renal vein and inferior vena cava were not dilated (not shown).
 
On DSA, the AVF was shown to be supplied by two branches from the anterior segmental renal artery (Figure 4a). Selective cannulation was achieved with a 2.7-Fr microcatheter (Rebar 18 reinforced microcatheter; Medtronic, Minneapolis [MN], US) [Figure 4c]. The first vessel supplying the right AVF was embolised with two detachable coils (Concerto Helix coils; Medtronic, Minneapolis [MN], US). A microvascular plug (MVP-3Q; Medtronic, Minneapolis [MN], US) was launched more proximally (Figure 4d) along the first artery to effectively address the smaller side branches (Figure 4d). The remaining artery was packed with detachable coils. The second artery supplying the AVF was first embolised with a detachable coil, followed by pushable coils (Nester microcoils; Cook Medical, Bloomington [IN], US) [Figure 4e]. Repeat DSA confirmed significant reduction in AVF vascularity [Figure 4f]. There was no recurrence of haematuria at 1-year clinical follow-up.
 
Figure 4. Case 2. Digital subtraction angiography (DSA) of right renal arteriovenous fistula. (a) Preprocedural DSA at the right main renal artery with an SHK catheter (Cordis, Miami [FL], US) confirmed supply by two branches of the anterior segmental renal artery (artery 1: white solid arrow, artery 2: hollow notched arrow). Early opacification of the inferior vena cava is noted (dashed arrow). (b) Selective cannulation showing small branches (arrowheads) supplied by artery 1. (c) Superselective angiogram with a Rebar 18 microcatheter confirmed dominant supply from one of the medial branches. (d) Deployment of an MVP-3Q microvascular plug (hollow arrowheads) proximally after distal coil embolisation. (e) Selective angiogram of artery 2 (hollow arrow) with a Rebar 18 microcatheter. Early opacification of the right renal vein is noted (dashed arrow). Note that the 4-6 mm larger coils (white solid arrow) had densely packed the proximal portion of artery 1. The MVP-3Q microvascular plug (hollow arrowheads) was located between the coils in artery 1. (f) Check DSA after coil embolisation of artery 2. Only tiny slow-flow serpiginous vessels were observed (not shown).
 
Case 3
 
A 48-year-old man was diagnosed with end-stage renal failure and managed by haemodialysis. A left renal AVF shown as a cystic area with moderate vascularity (Figure 5a and b) was incidentally detected on ultrasound and was presumed biopsy-related. Intervention was deemed indicated by nephrologists and urologists in view of the higher bleeding risk in end-stage renal failure. CT urogram confirmed an AVF centred at the lower pole of the left kidney with aneurysmal changes. Double renal arteries were seen. One of the renal arteries directly supplied the AVF and showed ostial stenosis and hypertrophy (7 mm) [Figure 5c]. The ostium measured 2.5 mm, limiting the option of sheaths and catheters. A short segment tight stenosis was seen in the dilated draining renal vein proximally near the renal hilum (Figure 5d), which minimised the migration of embolic agents.
 
Figure 5. Case 3. Doppler ultrasound (a, b) and reformatted maximal intensity projection contrast-enhanced computed tomography in corticomedullary phase (c, d). (a) The arteriovenous fistula (AVF) presenting with cystic sacs with chaotic colour flow signal. (b) The supplying renal artery is hypertrophied, with a reduced resistive index of 0.34 (normal = 0.5-0.7). (c) Ostial stenosis (solid arrow) of the supplying renal artery, which narrows to 2.5 mm. (d) Short segment stenosis (hollow arrow) of the draining renal vein near the AVF.
 
The supplying renal artery was cannulated with a 5-Fr H1 catheter (Torcon NB Advantage Catheter; Cook Medical, Bloomington [IN], US) [Figure 6a]. The most proximal venous pouch was selectively cannulated (Excelsior XT-27). Using the scaffold technique, the venous pouch was packed with coils of varying lengths and calibres (Ruby coils) [Figure 6c]. A microvascular plug (MVP-7Q; Medtronic, Minneapolis [MN], US) was launched at the supplying left renal artery [Figure 6d], followed by more proximal deployment of two detachable coils [Figure 6e]. Check DSA confirmed significant reduction in AVF vascularity and absence of collateral supply from the other renal artery (Figure 6f).
 
Figure 6. Case 3. Digital subtraction angiography (DSA) of left renal arteriovenous fistula. (a) Early-phase diagnostic DSA of the supplying renal artery (0.7 cm) with a H1 catheter. The most proximal venous pouch in the arteriovenous malformation (AVM) is opacified. (b) DSA confirms tight stenosis (solid arrow) at the distal left renal vein near the AVF. (c) Threading of Ruby coils, starting with larger coils with greater radial force. The anchor technique (dashed arrow), where the distal end was landed in a small adjacent branch, was applied to provide stability. (d) Deployment of an MVP-7Q microvascular plug (hollow arrowheads) proximal to the coil mass. Note the reflux of injected contrast along the renal artery. (e) Further proximal coil packing (hollow arrow) along the hypertrophied supplying renal artery with a Rebar 18 microcatheter (upper image). Reflux of injected contrast is again seen (lower image). (f) Check DSA of the other renal artery excluded collateral supply to the AVM.
 
DISCUSSION
 
To preserve renal function, endovascular treatment has become the mainstay treatment of renal AVF in the current literature. For non-traumatic AV shunts, Marunos et al[5] proposed corresponding treatment modalities based on three types of angioarchitecture. Type I involves single or few arteries shunting to a dilated single draining vein, while type II contains multiple arterioles shunting to a single dilated draining vein. Coils are recommended in these two types, while vascular plugs can be considered in type I shunts. For type III, where multiple connections exist between arterioles and venules, particles and liquid embolic agents are recommended. Proximal embolisation of the arterial feeder with coils in type III shunts should be avoided to prevent recruitment of collaterals. Traumatic shunts, which usually present with pseudoaneurysms, are located peripherally and have similar angioarchitecture to type I shunts. As well as coils, glue is a treatment option. These endovascular treatment modalities are considered effective and are commonly used in clinical practice.
 
Detachable coils allow precise deployment and have low risk of non-target embolisation in a high-flow setting. Particles and liquid embolic agents are time-efficient in type III profiles but carry risks of proximal and non-target embolisation. The combination of distal coil anchor and proximal vascular plug is gaining in popularity, with reported success in recanalised[6] and giant AVF,[2] although limited case numbers mean its superiority has not been validated. Plugging is an efficient alternative to coil mass, but a straight non-conical landing zone is required. The maximum sizes that the Amplatzer or the MVP Micro Vascular Plug system offer may also limit their application in enlarged feeders. For Amplatzer vascular plugs, serial deployment may be considered to achieve optimal flow control, especially for larger plugs due to their larger pore size.[7] In our experience, deployment of a single plug may be insufficient for flow control. It is therefore our preference to perform distal coil packing.
 
AVFs impose an elevated risk of distal non-target embolisation due to their high-flow nature. To provide coil stability, the double-catheter technique (Case 1) or the side-branch anchor technique (Case 3) can be performed. Flow modulation with occlusive balloons applied proximal (Case 1) and distal to the fistula also provides stability for the initial coil framework.[8] [9] The ‘pre-framing’ technique, which involves coiling the microcatheter in the designated area prior to coil deployment, has also been practised.[10] The rigidity of mechanically detachable or larger-sized coils is nonetheless technically difficult since the coils traverse through the tortuous catheter framework. It also risks catheter knotting and requires a side branch for the microcatheter to anchor upon. Alternatively, the use of covered or constrained stents for coil trapping has been successful.[3]
 
CT or MR arteriography provides an excellent roadmap for preprocedural planning and a crude estimation of the post-embolisation residual functional kidney. In Case 3 for example, ostial stenosis limited catheter sizing and subsequent choice of embolic agents. The venous outflow should also be carefully studied. A grossly dilated vein, as in Case 1, implies a high risk of distal non-target embolisation. A venous occlusive balloon is most reported to prevent distal embolisation. Suprarenal inferior vena cava filters may also be considered but their application is limited in flow-induced mega cava, as in Case 1. The use of an atrial septal defect occluder has also been reported.[11] Embolisation of the venous outflow tract is not commonly practised and not necessarily indicated when feeder obliteration is achieved. It may be considered when multifocal feeders are present, where extensive embolisation would result in lowered nephron-sparing capacity.
 
CONCLUSION
 
This case series is based on single-centre experience with a small sample size. Some cases were excluded, including those with difficult vascular anatomy and concerns about compromising renal function.
 
Endovascular treatment of three selected cases of renal AVF is illustrated. Various treatment modalities have been proven successful and may be selected according to the angioarchitecture. The combination of coil and plug is gaining popularity. The high-flow nature of AVF requires careful preprocedural planning and additional intra-procedural manoeuvres to minimise the risk of embolic agent migration. Target coiling of larger aneurysms also contributes to treatment success.
 
REFERENCES
 
1. Nagpal P, Bathla G, Saboo SS, Khandelwal A, Goyal A, Rybicki FJ, et al. Giant idiopathic renal arteriovenous fistula managed by coils and Amplatzer device: case report and literature review. World J Clin Cases. 2016;4:364-8. Crossref
 
2. Yoneda S, Madono K, Tanigawa G, Fujita K, Yazawa K, Hosomi M, et al. Case of giant renal arteriovenous fistula in a long-term hemodialysis patient [in Japanese]. Hinyokika Kiyo. 2009;55:559-62.
 
3. Resnick S, Chiang A. Transcatheter embolization of a high-flow renal arteriovenous fistula with use of a constrained wallstent to prevent coil migration. J Vasc Interv Radiol. 2006;17:363-7. Crossref
 
4. Shie RF, Su TW, Hsu MY, Chu SY, Ko PJ. Transarterial embolization of a large high-flow right renal arteriovenous fistula using stents and an across-stent wire-trapping technique. J Vasc Surg Cases Innov Tech. 2019;5:122-7. Crossref
 
5. Maruno M, Kiyosue H, Tanoue S, Hongo N, Matsumoto S, Mori H, et al. Renal arteriovenous shunts: clinical features, imaging appearance, and transcatheter embolization based on angioarchitecture. Radiographics. 2016;36:580-95. Crossref
 
6. Abdel-Aal AK, Elsabbagh A, Soliman H, Hamed M, Underwood E, Saddekni S. Percutaneous embolization of a postnephrectomy arteriovenous fistula with intervening pseudoaneurysm using the Amplatzer Vascular Plug 2. Vasc Endovascular Surg. 2014;48:516-21. Crossref
 
7. Balasubramanian K, Keshava SN, Lenin A, Mukha R. Endovascular management of a patient with massive renal arteriovenous fistula: challenges and tricks. BMJ Case Rep. 2021;14:e236358. Crossref
 
8. Idowu O, Barodawala F, Nemeth A, Trerotola SO. Dual use of an Amplatzer device in the transcatheter embolization of a large high-flow renal arteriovenous fistula. J Vasc Interv Radiol. 2007;18:671-6. Crossref
 
9. Mansueto G, D’Onofrio M, Minniti S, Ferrara RM, Procacci C. Therapeutic embolization of idiopathic renal arteriovenous fistula using the “stop-flow” technique. J Endovasc Ther. 2001;8:210-5. Crossref
 
10. Sundarakumar DK, Kroma GM, Smith CM, Lopera JE, Suri R. Embolization of a large high-flow renal arteriovenous fistula using 035” and 018” detachable coils. Indian J Radiol Imaging. 2013;23:151-4. Crossref
 
11. Chen X, Zeng Q, Ye P, Miao H, Chen Y. Embolization of high-output idiopathic renal arteriovenous fistula primarily using an atrial septal defect occluder via venous access: a case report. BMC Nephrol. 2019;20:15. Crossref
 
 
 

Aceruloplasminemia with Neurodegenerative Condition: A Case Report

CK Li, CY Lau, KH Chin, CY Chu

CASE REPORT
 
Aceruloplasminemia with Neurodegenerative Condition: A Case Report
 
CK Li, CY Lau, KH Chin, CY Chu
Department of Radiology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China
 
Correspondence: Dr CK Li, Department of Radiology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China. Email: lck340@ha.org.hk
 
Submitted: 4 May 2024; Accepted: 3 September 2024.
 
Contributors: CKL designed the study, acquired and analysed the data, and drafted the manuscript. CYL, KHC and CYC critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: The study was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: IRB-2024-245). The patient was treated in accordance with the tenets of the Declaration of Helsinki. Informed verbal consent was obtained from the patient’s first-degree relative for the publication of this case report, including the accompanying images.
 
 
 
 
CASE PRESENTATION
 
A 68-year-old Chinese woman presented to the Accident and Emergency Department of our institution in March 2023 with confusion, gait instability, and a history of falls. She had experienced a rapid decline in mobility and motivation, rendering her homebound since December 2022. Her medical history revealed repetitive behaviour spanning over a decade, alongside co-morbidities such as diabetes mellitus and mild anaemia since 2007. Neither the patient nor her relatives reported seizures or loss of consciousness. Physical examination showed no focal neurological deficits. Dementia evaluation by the Montreal Cognitive Assessment test yielded a score of 2 out of 30, indicating a high clinical suspicion.
 
Non-contrast computed tomography (CT) of the brain was unremarkable with known chronic ventriculomegaly as the only notable finding. Subsequent contrast-enhanced magnetic resonance imaging (MRI) showed extensive symmetrical blooming artefacts in various deep grey matter areas on the susceptibility-weighted imaging (SWI) sequence, including the bilateral caudate nuclei, lentiform nuclei, thalami, red nuclei, substantia nigra, and bilateral dentate nuclei of the cerebellum. Diffuse gyriform-like blooming artefacts were observed outlining the surfaces of the cerebrum and cerebellum (Figure 1). These MRI findings suggested significant mineral deposition, raising suspicion of aceruloplasminemia and other differential diagnoses such as other neurodegeneration with brain iron accumulation. In view of the suspected iron accumulation, contrast-enhanced CT of the abdomen and the pelvis, as well as MRI of the liver and the heart, were performed. The CT scan revealed diffuse hyperattenuation of the liver parenchyma, while MRI showed evidence of iron overload in both the liver parenchyma and myocardium (Table 1).
 
Figure 1. Brain magnetic resonance imaging (3T). Axial susceptibility-weighted sequence shows extensive symmetrical blooming artefacts in deep grey matter areas, including (a) bilateral caudate nuclei (yellow arrows), lentiform nuclei (white arrows), thalami (red stars); (b) red nuclei (yellow circles), substantia nigra (orange arrows); and (c) bilateral dentate nuclei of the cerebellum (red arrows). (d, e) Diffuse gyriform-like blooming artefacts outlining the surfaces of the cerebrum and cerebellum (yellow arrowheads).
 
Table 1. Calculated T2-star value of liver parenchyma and myocardium.
 
Biochemically, the patient exhibited a markedly low ceruloplasmin level of under 0.02 g/L (normal range = 0.22-0.58), an elevated ferritin level of 3270 pmol/L (normal range = 25-689), and a low iron saturation of 13.1% (Table 2). She also had a history of chronic mild anaemia for at least a decade, with haemoglobin levels ranging from 10.4 g/dL in April 2013 to 8.9 g/dL in March 2023. Genetic testing subsequently identified a pathogenic variant of the ceruloplasmin gene, confirming the diagnosis of aceruloplasminemia.
 
Table 2. Laboratory findings in our case.
 
The patient and her relative were counselled about the definitive diagnosis, and the features of the disease were explained. No specific treatment was prescribed for aceruloplasminemia due to chronic neurological symptoms and impaired cognitive function. The patient continued to receive holistic care in a residential elderly care home, with monitoring for her diabetes. Genetic testing was also offered to her first-degree relatives.
 
DISCUSSION
 
Aceruloplasminemia is a rare autosomal recessive disorder characterised by the absence or dysfunction of ceruloplasmin with consequent iron accumulation in various tissues and organs, leading to a spectrum of neurological and systemic manifestations.[1] Our case illustrates the importance of recognising the clinical and radiological features of aceruloplasminemia to facilitate accurate diagnosis and management.
 
Aceruloplasminemia was first documented in 1987 by Miyajima et al[2] in a 52-year-old woman with blepharospasm, retinal degeneration, and diabetes mellitus. The estimated prevalence is approximately 1 in 2,000,000 population among Japanese individuals born from non-consanguineous marriages.[3] Nonetheless, this estimation is region-specific and may not be applicable to other populations.[4] Clinical manifestations leading to diagnosis by neurologists include cerebellar signs such as dysarthria, trunk and limb ataxia, and involuntary movements including dystonia, chorea, and tremors. Symptoms may vary widely among individuals and may overlap with other neurological or metabolic disorders.[5]
 
To understand the pathophysiology of aceruloplasminemia, two distinct isoforms of ceruloplasmin are produced via alternative splicing in exons 19 and 20, resulting in a soluble form in plasma and a glycosylphosphatidylinositol-anchored membrane form.[6] The ferroxidase activity of the membrane-bound ceruloplasmin plays a vital role for incorporating ferric cation Fe3+ into plasma transferrin, facilitating its delivery to other cells via transferrin receptor 1. In the absence of ceruloplasmin, iron initially accumulates in astrocytes, triggering neuronal iron starvation. Consequently, neurons resort to alternative iron sources such as non–transferrin-bound iron, exacerbating toxicity (Figure 2).[1] [7]
 
Figure 2. The ferroxidase activity of the membrane-bound ceruloplasmin (CP) plays a vital role in incorporating ferric cation Fe3+ into plasma transferrin, facilitating its delivery to other cells. In the absence of CP, iron accumulates in astrocytes, triggering neuronal iron starvation. Consequently, neurons resort to alternative iron sources, such as non–transferrin-bound iron, exacerbating toxicity.1,7 Apotransferrin is the iron-free form of transferrin, indicating failure of iron incorporation. The accumulation of ferric cation Fe2+ also leads to fenton reaction with generation of highly reactive hydroxyl radicals, causing oxidative damage.
 
The hallmark radiological feature of aceruloplasminemia manifests as symmetric blooming artefacts on SWI, attributable to iron accumulation in the brain. Typically, this involves regions such as the basal ganglia and thalamus, cerebral cortex and dentate nuclei of the cerebellum.[8] Aceruloplasminemia stands out as the sole recognised disorder featuring both cerebral and systemic manifestations of iron accumulation.[9] As in our patient, cardiac and hepatic iron overload may also occur. Hepatic iron overload often presents with hyperattenuation of the liver parenchyma on CT scans and is quantitatively assessed via MRI dedicated to evaluating iron overload in the liver. Nonetheless, liver iron accumulation seldom leads to clinical manifestations such as cirrhosis or liver failure.[10] Iron deposition in other organs, including the heart, pancreas, and other endocrine glands, has been documented and can be evaluated by MRI.[7]
 
The neurological manifestations of aceruloplasminemia are heterogeneous and often progressive. In our patient, initial symptoms such as confusion, gait instability, and falls were consistent with those commonly reported in the literature. Documented neurological features included behavioural changes or psychiatric manifestations, cognitive impairment, extrapyramidal signs, cerebellar signs, and involuntary movements.[5] Another classic clinical manifestation is diabetes mellitus, typically presenting in the fourth to sixth decades of life in individuals without classic risk factors or need for insulin treatment.[11] The mechanism underlying the development of diabetes mellitus in aceruloplasminemia remains poorly understood, although iron accumulation is noted predominantly in exocrine rather than endocrine pancreatic cells.[12] Some studies suggest that the clinical triad of aceruloplasminemia may comprise neurodegeneration, diabetes mellitus, and retinal degeneration.[13] [14] Nonetheless retinopathy is less frequently observed in non-Japanese case series, and its direct association with aceruloplasminemia remains uncertain.[13] [14]
 
Biochemically, the first detectable parameters of aceruloplasminemia, as indicated by all major case series including our own, encompass mild microcytic anaemia, low transferrin saturation, and hyperserotonaemia. This biochemical triad holds crucial diagnostic significance long before other clinical manifestations emerge. Serum ceruloplasmin is typically undetectable or markedly reduced and serves as an important diagnostic parameter. Although mild microcytic anaemia often emerges as the earliest biochemical sign of aceruloplasminemia,[5] [10] it rarely leads to diagnosis at the early pre-symptomatic stage. By integrating biochemical studies with radiological and clinical manifestations, the exclusion of other differential neurodegenerative diseases becomes more manageable. As in our case, genetic testing provides definitive evidence to confirm the diagnosis of aceruloplasminemia and enables genetic counselling and family screening for at-risk individuals.
 
Treatment of aceruloplasminemia primarily involves iron-chelating agents; however, their effectiveness in reducing brain iron and alleviating neurological symptoms remains uncertain. Currently, there is no convincing evidence supporting the clinical benefits of iron removal therapy. Phlebotomy, another treatment option, is also considered suboptimal. Alternative strategies focus on preventing oxidative tissue damage, such as administering vitamin E or zinc sulphate.[10] Timely diagnosis and treatment are paramount to prevent irreversible neurological complications.[7]
 
CONCLUSION
 
Aceruloplasminemia is difficult to diagnose and requires a high level of awareness of its clinical features, biochemical parameters, and radiological findings. The biochemical triad of mild anaemia, low transferrin saturation, and hyperserotonaemia serves as a key diagnostic indicator when no alternative explanation is evident. The condition should be considered in patients who present with mild microcytic anaemia, early-onset diabetes mellitus, and unexplained liver iron overload. In later stages, adult-onset neurological dysfunction, such as behavioural changes, psychiatric disturbances, as well as cerebellar and extrapyramidal signs, become apparent. Corresponding MRI findings often reveal symmetrical hypointensity in the basal ganglia and thalamus, cerebral cortex and dentate nuclei of cerebellum in T2 and T2-star sequences, along with a pronounced blooming artifact in SWI. Prompt diagnosis is crucial to prevent irreversible neurological complications.
 
REFERENCES
 
1. Fasano A, Colosimo C, Miyajima H, Tonali PA, Re TJ, Bentivoglio AR. Aceruloplasminemia: a novel mutation in a family with marked phenotypic variability. Mov Disord. 2008;23:751-5. Crossref
 
2. Miyajima H, Nishimura Y, Mimguchi K, Sakamoto M, Shimizu T, Honda N. Familial apoceruloplasmin deficiency associated with blepharospasm and retinal degeneration. Neurology. 1987;37:761-7. Crossref
 
3. Miyajima H, Kohno S, Takahashi Y, Yonekawa O, Kanno T. Estimation of the gene frequency of aceruloplasminemia in Japan. Neurology. 1999;53:617-9. Crossref
 
4. Yamamura A, Kikukawa Y, Tokunaga K, Miyagawa E, Endo S, Miyake H, et al. Pancytopenia and myelodysplastic changes in aceruloplasminemia: a case with a novel pathogenic variant in the ceruloplasmin gene. Intern Med. 2018;57:1905-10. Crossref
 
5. Miyajima H, Hosoi Y. Aceruloplasminemia. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJ, et al, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993. Available from: http://www.ncbi.nlm.nih.gov/books/NBK1493/". Accessed 4 May 2024.
 
6. Patel BN, Dunn RJ, David S. Alternative RNA splicing generates a glycosylphosphatidylinositol-anchored form of ceruloplasmin in mammalian brain. J Biol Chem. 2000;275:4305-10. Crossref
 
7. Marchi G, Busti F, Lira Zidanes A, Castagna A, Girelli D. Aceruloplasminemia: a severe neurodegenerative disorder deserving an early diagnosis. Front Neurosci. 2019;13:325. Crossref
 
8. Grisoli M, Piperno A, Chiapparini L, Mariani R, Savoiardo M. MR imaging of cerebral cortical involvement in aceruloplasminemia. AJNR Am J Neuroradiol. 2005;26:657-61.
 
9. Touarsa F, Ali Mohamed D, Onka B, Rostoum S, Ech-Cherif El Kettani N, Fikri M, et al. Brain iron accumulation on MRI revealing aceruloplasminemia: a rare cause of simultaneous brain and systemic iron overload. BJR Case Rep. 2022;8:20220035. Crossref
 
10. Pelucchi S, Mariani R, Ravasi G, Pelloni I, Marano M, Tremolizzo L, et al. Phenotypic heterogeneity in seven Italian cases of aceruloplasminemia. Parkinsonism Relat Disord. 2018;51:36-42. Crossref
 
11. Vroegindeweij LH, Langendonk JG, Langeveld M, Hoogendoorn M, Kievit AJ, Di Raimondo D, et al. New insights in the neurological phenotype of aceruloplasminemia in Caucasian patients. Parkinsonism Relat Disord. 2017;36:33-40. Crossref
 
12. Kato T, Daimon M, Kawanami T, Ikezawa Y, Sasaki H, Maeda K. Islet changes in hereditary ceruloplasmin deficiency. Hum Pathol. 1997;28:499-502. Crossref
 
13. Miyajima H, Takahashi Y, Kono S. Aceruloplasminemia, an inherited disorder of iron metabolism. Biometals. 2003;16:205-13. Crossref
 
14. McNeill A, Pandolfo M, Kuhn J, Shang H, Miyajima H. The neurological presentation of ceruloplasmin gene mutations. Eur Neurol. 2008;60:200-5. Crossref
 
 
 
PICTORIAL ESSAYS

Imaging Features of Clavicular Pathologies and Their Articulations: A Pictorial Essay

   CME

BWT Cheng, JHM Cheng, KH Chin, CY Chu

PICTORIAL ESSAY    CME
 
Imaging Features of Clavicular Pathologies and Their Articulations: A Pictorial Essay
 
BWT Cheng, JHM Cheng, KH Chin, CY Chu
Department of Radiology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China
 
Correspondence: Dr BWT Cheng, Department of Radiology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China. Email: cwt413@ha.org.hk
 
Submitted: 10 January 2024; Accepted: 1 November 2024.
 
Contributors: BWTC and JHMC designed the study. BWTC, JHMC and KHC acquired the data. All authors analysed the data. BWTC drafted the manuscript. All authors critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: The study was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: CIRB-2023-btained from the Board due to the retrospective nature of the study.
 
 
 
 
INTRODUCTION
 
The clavicle, the acromioclavicular (AC) joint and the sternoclavicular joint can be affected by a wide range of pathologies, including infection, inflammation, degeneration, metabolic disorders, neoplasms, trauma, and congenital anomalies. This pictorial essay illustrates the radiological features of clavicular pathologies to facilitate accurate diagnosis and management, based on cases of clavicular, AC and sternoclavicular pathologies diagnosed in the Hong Kong East Cluster from April 2014 to April 2023.
 
NORMAL ANATOMY
 
A basic understanding of the anatomy of the clavicle and its articulations is essential for image interpretation. The clavicle is a horizontally oriented S-shaped bone that has a large medial metaphysis articulating with the sternum, a tubular diaphysis, and a flared lateral metaphysis that articulates with the acromion (Figure 1). The sternoclavicular joint is a synovial joint formed by the medial clavicular metaphysis, the clavicular notch of the manubrium sterni, and the cartilage of the first rib. The articular surfaces of the clavicle and manubrium are separated by a fibrocartilaginous disk.[1] The costoclavicular and sternoclavicular ligaments (thickenings of the joint capsule), and interclavicular ligament (located between the superomedial ends of the two clavicles) provide joint stability (Figure 2).[2] The AC joint is a planar diarthrodial joint located between the lateral surface of the clavicle and the medial surface of the acromion. Stabilisers of the joint include the AC joint capsule and the AC, coracoacromial, and coracoclavicular (consisting of trapezoid and conoid ligaments) ligaments. A flexible fibrocartilaginous disk is peripherally continuous with the joint capsule (Figure 3).[3] There are four types of acromion shape, namely, flat, curved, hooked, and convex. An unfused acromial ossification centre (os acromiale) is an anatomical variant.
 
Figure 1. Anatomy of the clavicle. (a) Axial-oblique T1-weighted and (b) short-tau inversion recovery magnetic resonance images of the right clavicle demonstrate the S-shaped bone with a large medial end, a tubular mid-portion, and flaring of the lateral end.
 
Figure 2. Anatomy of the sternoclavicular joint. (a) Coronal T1-weighted and (b) short-tau inversion recovery magnetic resonance imaging (MRI) of the right sternoclavicular joint with intact fibrocartilaginous disk (open arrows) and costoclavicular ligament (circle in [b]). (c) Coronal T1-weighted MRI of bilateral sternoclavicular joints shows the interclavicular ligament across the upper sternum (arrows).
 
Figure 3. Anatomy of the acromioclavicular joint. (a) Coronal proton density (PD)–weighted and (b) T2-weighted fat-suppressed magnetic resonance imaging (MRI) of the right acromioclavicular joint with normal acromioclavicular joint capsule (circles), which cannot be differentiated from the acromioclavicular ligaments on routine MRI. (c, d) Coronal PD-weighted MRI of the right shoulder demonstrates ligaments around the distal clavicle. The coracoacromial ligament is located most laterally (arrow in [c]). The trapezoid portion of the coracoclavicular ligament is located more medially and inserts onto the inferior margin of the lateral clavicle; the conoid portion (curved arrow in [d]) is the most medial and vertically oriented.
 
RADIOLOGICAL FEATURES OF PATHOLOGIES
 
Congenital Anomalies
 
Cleidocranial dysostosis is a rare autosomal dominant disease that mainly affects midline skeletal structures, with features including hypoplasia or aplasia of the clavicles, large fontanelles, multiple Wormian bones, a widened pubic symphysis, and supernumerary teeth (Figure 4). Eight cases of cleidocranial dysostosis were identified during the review period.
 
Figure 4. A case of cleidocranial dysostosis. (a) Chest X-ray demonstrates aplasia of bilateral clavicles (ellipse). (b) Pelvic X-ray demonstrates widening of pubic symphysis (arrowheads). Frontal (c) and lateral (d) skull X-rays demonstrate widened sagittal suture (open arrows in [c]) and multiple Wormian bones (arrows in [d]). (e) Orthopantomogram demonstrates supernumerary teeth.
 
Articular Infection
 
Septic arthritis of the sternoclavicular joint is uncommon and is usually monoarticular with an insidious onset.[4] Radiological features on radiographs and computed tomography (CT) include subarticular erosions, joint space widening, and fluid collections. Magnetic resonance imaging (MRI) features include bone marrow oedema, bone destruction, joint effusion, and inflammatory changes of the surrounding soft tissue. Both CT and MRI are useful for early diagnosis and assessment of complications such as associated osteomyelitis and retrosternal/chest wall abscesses that may require surgical treatment.
 
Inflammation
 
Spondyloarthropathies such as ankylosing spondylitis and psoriasis can affect the sternoclavicular joint. Radiographic and CT features include bone erosions, partial or complete fusion of the joint, and hyperostosis surrounding the joint[2] (Figures 5, 6 and 7). Rheumatoid arthritis may be accompanied by pannus formation with bony erosions on imaging.
 
Figure 5. Two cases of spondyloarthropathy of the sternoclavicular joint. (a-c) First case. (a) Coronal and (b) axial computed tomography (CT) bone window images of bilateral sternoclavicular joints in a patient with known ankylosing spondylitis. The right sternoclavicular joint demonstrates osseous fusion (circles), while the left shows hyperostosis with mild bone erosions and subchondral sclerotic changes (arrows). (c) X-ray of the cervical spine demonstrates bamboo spine. (d-f) Second case. (d) Coronal and (e) axial-oblique CT bone window images of the left sternoclavicular joint demonstrate hypertrophic change, bone erosions and sclerosis (open arrows) in a case of known psoriasis. (f) Pelvic X-ray demonstrates bony ankylosis of bilateral sacroiliac joints (arrowheads).
 
Figure 6. Two cases of the SAPHO (synovitis, acne, pustulosis, hyperostosis, and osteitis) syndrome. (a, b) First case. (a) Radiograph of the clavicles demonstrates prominent hyperostosis in both sternoclavicular joints (circle), more on the right, and sclerosis in both distal clavicles (arrows). (b) Bone scintigraphy demonstrates diffusely increased tracer uptake in both sternocostoclavicular junctions and medial clavicular ends (open arrows), associated with hyperostosis, compatible with the ‘bull’s head’ sign. (c-e) Second case. (c) X-ray demonstrates hyperostosis of both medial clavicles (circle). A Ryles tube is noted (arrowheads). (d) Coronal and (e) axial bone window computed tomography images demonstrate corresponding significant sternoclavicular hyperostosis (open arrows in [e]), marked joint space narrowing on the right with cortical irregularities (arrowheads in [d]) and complete ankylosis on the left (arrows in [d]).
 
Figure 7. A case of osteoarthritis of the right acromioclavicular joint. (a) Radiograph and (b) coronal bone window computed tomography of the acromioclavicular joint demonstrate osteoarthritis with joint space narrowing, marginal osteophytes, subchondral sclerosis and cysts (circles). (c) Coronal short-tau inversion recovery magnetic resonance imaging (MRI) of the acromioclavicular joint shows subchondral bone marrow oedema (arrowhead) and thickened superior capsule (arrows). (d) Sagittal T1-weighted MRI shows inferior osteophyte of the acromion (open arrow) and capsular hypertrophy (curved arrows) of the acromioclavicular joint mildly indenting the supraspinatus tendon.
 
The SAPHO (synovitis, acne, pustulosis, hyperostosis, and osteitis) syndrome is an inflammatory condition with aseptic osteoarticular involvement and characteristic skin lesions. In adults, it usually involves the anterior chest wall (60%-95%), particularly the sternocostoclavicular junction, followed by the axial skeleton, such as the spine and sacroiliac joints. Features of SAPHO on radiographs and CT include bony sclerosis, cortical thickening, and narrowing of the medullary canal. Adjacent changes include joint space narrowing and periarticular osteopenia, as well as ligamentous ossification with bony bridging across the joint. CT is good for detecting the osteoarticular manifestations, while MRI is sensitive in detecting early disease with bone and soft tissue oedema. On bone scintigraphy, SAPHO in the anterior chest wall typically manifests as the ‘bull’s head’ sign, with mostly symmetrical increased uptake in the sternoclavicular regions.[5] The radiological differential diagnosis includes sternoclavicular osteoarthritis, condensing osteitis of the clavicle, osteonecrosis, and septic arthritis (Figure 6). Six cases of SAPHO were identified during the review period.
 
Degenerative Joint Disease
 
Osteoarthritis is a common cause of pain at the AC and sternoclavicular joints (Figures 7, 8 and 9). Radiological features include narrowing of the joint space, marginal osteophytes, capsular hypertrophy, subchondral sclerosis, cysts, and bone marrow oedema.
 
Figure 8. Two cases of complete chronic supraspinatus tear with geyser sign. (a-d) First case. (a) Radiograph shows a soft tissue shadow (arrows) above the acromioclavicular joint with mild osteoarthritic change. Superior migration of the humeral head and subacromial acetabularisation are noted, highly suggestive of chronic supraspinatus tear. (b) Transverse ultrasound demonstrates a cystic lesion with low-level echoes above the acromioclavicular joint (white star). Corresponding coronal (c) and sagittal (d) T2-weighted fat-supressed magnetic resonance images show complete supraspinatus tendon tear (open arrows in [c]) with uncovering and superior migration of the humeral head, disrupted inferior capsule (circle in [c]), and a well-defined homogenous cystic lesion resembling the geyser sign (black stars). Incidental findings include a subcortical bone cyst (arrowhead in [c]) and enchondroma (curved arrow in [d]) at the humeral head. (e, f) Second case. (e) Axial and (f) coronal T1-weighted fat-supressed volumetric interpolated breath-hold examination magnetic resonance arthrogram images of the right shoulder demonstrate a full-thickness tear of the supraspinatus-infraspinatus interdigitation (circle in [f]) with superior migration of the humeral head. There is contrast extension via the subacromial subdeltoid bursa to the acromioclavicular joint (arrows).
 
Figure 9. Two cases of sternoclavicular joint osteoarthritis. (a) First case. Coronal bone window computed tomography of bilateral sternoclavicular joints demonstrates osteoarthritis of the left sternoclavicular joint with joint space narrowing, articular irregularity, subchondral sclerosis and cysts (circle). The right sternoclavicular joint is preserved. (b, c) Second case. (b) Axial T2-weighted fat-supressed and (c) coronal T1-weighted magnetic resonance images of the left sternoclavicular joint demonstrate capsular thickening (arrows in [b]) and marginal osteophytes (arrowhead in [c]).
 
A chronic large full-thickness supraspinatus tendon tear can lead to superior migration of the humeral head, which may erode the subacromial-subdeltoid bursa and inferior AC capsule, forming a communication between the glenohumeral and AC joints. This may lead to a sizeable fluid pouch over the AC joint, giving rise to the geyser sign (Figure 8). During the review period, six such cases were identified.
 
Trauma
 
Clavicular fractures (Figures 10, 11 and 12) are common and represent 2.6% to 5% of all fractures, with the vast majority occurring in the mid clavicle (69%-82%).[6] Apart from location, alignment of the clavicle with the AC and sternoclavicular joints should be assessed, since malalignment may signify significant ligamentous injury.
 
Figure 10. A case of right midclavicular fracture. (a) Coronal T1-weighted and (b) short-tau inversion recovery (STIR) magnetic resonance images demonstrate displaced midclavicular fracture (circles). T1-weighted hypointense, STIR hyperintense bone marrow change next to the fracture indicate posttraumatic change. (c) Coronal and (d) axial bone window computed tomography images show displaced midclavicular fracture (circles) with superior angulation and inferior displacement of the distal fragment.
 
Figure 11. Two cases of acromioclavicular joint injury. (a-d) First case. (a) Radiograph of the left acromioclavicular joint demonstrates slight superior displacement of the clavicle (arrows). (b-d) Sagittal and coronal T2-weighted fat-suppressed magnetic resonance imaging (MRI) of the left shoulder demonstrates increased signal in the coracoclavicular ligament (circle) and acromioclavicular ligament/joint capsule (open arrows), suggestive of Rockwood type II left acromioclavicular joint injury. (e-g) Second case. (e, f) Radiographs of the right acromioclavicular joint show markedly elevated clavicle with increased coracoclavicular distance, consistent with Rockwood type V right acromioclavicular joint injury. (g) Post–open reduction radiograph shows satisfactory joint alignment.
 
Figure 12. A case of posttraumatic left distal clavicle osteolysis. (a) Radiograph shows widening of the acromioclavicular joint with erosions at the lateral end of the clavicle (arrows). (b) Coronal and (c) axial bone window computed tomography demonstrates erosions over the lateral end of the clavicle (open arrows) with tiny adjacent osseous foci, and widening of the acromioclavicular joint with mild soft tissue swelling.
 
AC joint injury is a common injury, occurring in 9% to 12% of shoulder injuries. The Rockwood classification is the most widely used classification system for AC joint injuries. It is classified into six types, depending on the direction and degree of clavicular displacement, which correlates with the severity of injury and involvement of the AC and coracoclavicular ligaments, and the deltotrapezial complex (Table).
 
Table. Rockwood classification.
 
Sternoclavicular joint dislocations are classified as anterior or posterior, and posterior dislocation has potentially serious complications due to the risk of injury to mediastinal structures such as the trachea and great vessels. On non-rotated radiographs, a difference in the relative craniocaudal positions of the medial clavicles exceeding 50% of the width of the clavicular heads suggests dislocation. However, diagnosis by radiographs may be difficult due to anatomical superimposition. CT is required for definitive diagnosis and to assess potential mediastinal injury.
 
Distal Clavicle Osteolysis
 
Distal clavicle osteolysis (Figure 13) is painful bone resorption of the distal clavicle, most common in young adults with male predominance. It can be categorised into posttraumatic or overuse forms, which share identical imaging findings. Radiological features on radiographs and CT include cortical irregularity, ‘flame-shaped’ bony resorption, and subchondral cysts involving the distal clavicle. MRI is most sensitive in demonstrating clavicular marrow oedema in the early phase of the disease. Effusion and capsular oedema are other features on MRI.
 
Figure 13. Posttraumatic right distal clavicle osteolysis. (a) Radiograph shows Rockwood type II injury with bone resorption at the inferior clavicular end (arrows). (b) Coronal proton density–weighted, (c) short-tau inversion recovery (STIR), and (d) sagittal STIR magnetic resonance images show widened joint space (double-head arrow in [b]) with capsular thickening (open arrows in [c]), and bone marrow oedema over the distal clavicle (circle in [d]).
 
The differential diagnoses of distal clavicle erosion include rheumatoid arthritis, hyperparathyroidism, and scleroderma.
 
Non-Articular Infection
 
Radiological features of clavicular osteomyelitis on radiographs and CT include cortical erosion, regional osteopenia, periosteal reaction, and adjacent soft tissue swelling (Figures 14, 15 and 16). On MRI, features of osteomyelitis typically include bone marrow oedema and surrounding soft tissue inflammatory change or collection. With time, an intraosseous abscess may form, typically seen as a focal intramedullary T2-weighted hyperintensity with variable rim enhancement. Subsequently, other osteomyelitic features such as sequestrum, involucrum, and cloaca formation may also become apparent.
 
Figure 14. A case of septic arthritis of the left sternoclavicular joint. (a) Coronal T1-weighted, (b) short-tau inversion recovery, (c) coronal, and (d) axial post-contrast T1-weighted fat-supressed magnetic resonance images show enhancing soft tissues (open arrows in [c] and [d]), bone marrow oedema of the medial clavicle and adjacent manubrium (arrowheads in [b]), and bony erosion of the medial end of the clavicle (arrows in [a]).
 
Figure 15. A case of Staphylococcus aureus septic arthritis of the right sternoclavicular joint. (a, b) Ultrasound shows gross capsular thickening of the joint (arrows in [a]) with cortical erosion over the articular surface of the clavicular head (arrowhead in [b]). (c) Axial soft tissue window and (d) coronal bone window computed tomography demonstrates enhancing soft tissue and fluid (open arrows in [c]) with subarticular erosions of the medial clavicle and adjacent manubrium (curved arrows in [d]). (e, f) Gallium-67 single-photon emission computed tomography/computed tomography of the thorax shows increased gallium activity involving the right sternoclavicular joint, adjacent right upper chest wall muscles with probable extension into the right medial clavicular head (circles).
 
Figure 16. A case of tuberculous osteomyelitis of the left clavicle. (a) Coronal and (b) axial soft tissue window computed tomography (CT) images demonstrate a lytic lesion at the clavicular head with cortical destruction (circle in [a]) and adjacent soft tissue swelling (arrows in [b]). (c) Axial T1-weighted, (d) short-tau inversion recovery (STIR), (e) axial, and (f) coronal post-contrast T1-weighted fat-supressed magnetic resonance images demonstrate a corresponding intramedullary T1-weighted hypointense, STIR hyperintense lesion with peripheral enhancement at the clavicular head (open arrows), and adjacent subcutaneous soft tissue oedema and rim-enhancing collection (curved arrow in [e]). (g) CT-guided biopsy confirms mycobacterial infection.
 
Neoplasm
 
Bone tumours of the clavicle are rare, with a reported frequency of less than 1% of all bone tumours.[6] While primary bone tumour of the clavicle is uncommon, the majority are malignant and include plasmacytoma, osteosarcoma, and Ewing’s sarcoma (Figures 17, 18 and 19). Bone metastases can involve the clavicle, with breast, lung, and prostate cancer being the more common primaries. Radiological features of aggressive bone tumours on radiographs and CT include a wide zone of transition, cortical destruction, and periosteal reaction. On MRI, there is invariably marrow replacement, sometimes with bony destruction, extraosseous extension, and perilesional oedema.
 
Figure 17. Carcinosarcoma of the right clavicle in a patient with breast cancer post-radiotherapy. (a) Coronal T1-weighted, (b) short-tau inversion recovery, (c) coronal, and (d) axial post-contrast T1-weighted fat-supressed magnetic resonance imaging demonstrates a huge irregular heterogeneous enhancing T1-weighted isointense, T2-weighted hyperintense mass replacing the clavicle (arrows), with internal cystic components. (e) Ultrasound-guided biopsy confirms carcinosarcoma.
 
Figure 18. A case of myeloma at the left clavicle. (a) X-ray of the clavicle demonstrates a lytic lesion at the medial clavicle (circle). (b) Coronal T1-weighted, (c) short-tau inversion recovery (STIR), and (d) post-contrast T1-weighted fat-supressed magnetic resonance images demonstrate a corresponding T1-weighted intermediate, STIR hyperintense and enhancing nodular lesion (arrows), expansile with mild adjacent soft tissue oedema and enhancement. Small joint effusion and capsule enhancement are noted (open arrows in [c] and [d]). Myeloma was confirmed by bone marrow aspiration.
 
Figure 19. Two cases of bone metastases to the clavicle. (a-d) First case. (a) Radiograph of the left clavicle demonstrates an expansile destructive lytic lesion at the clavicular head with indistinct superior cortex (circle). (b) Axial soft tissue window computed tomography demonstrates a suspicious irregular hypoenhancing lung lesion in the lingula (open arrow), subsequently diagnosed as epidermal growth factor receptor mutation–positive lung carcinoma. (c) Axial bone window computed tomography (CT) and (d) positron emission tomography/CT demonstrate an expansile destructive lesion in the left clavicular head with pathological fracture and increased 18F-fluorodeoxyglucose uptake (arrows), compatible with bone metastasis. The right clavicular head is unremarkable. (e-g) Second case. (e, f) Axial bone window CT of bilateral clavicles of a patient with known prostate cancer demonstrate sclerotic lesions in the distal clavicle diaphyses (arrowheads). (g) Bone scan demonstrates innumerable foci of increased uptake, including the corresponding distal clavicles (circles), indicating disseminated bone metastasis.
 
CONCLUSION
 
The clavicle, AC joint, and sternoclavicular joint are important structures of the upper extremity. To make an accurate diagnosis for treatment guidance, radiologists need to be familiar with the normal anatomy as well as the radiological features of abnormalities across different imaging modalities.
 
REFERENCES
 
1. Klein MA, Miro PA, Spreitzer AM, Carrera GF. MR imaging of the normal sternoclavicular joint: spectrum of findings. AJR Am J Roentgenol. 1995;165:391-3. Crossref
 
2. Olivier T, Kasprzak K, Herteleer M, Demondion X, Jacques T, Cotten A. Anatomical study of the sternoclavicular joint using high-frequency ultrasound. Insights Imaging. 2022;13:66. Crossref
 
3. Flores DV, Goes PK, Gómez CM, Umpire DF, Pathria MN. Imaging of the acromioclavicular joint: anatomy, function, pathologic features, and treatment. Radiographics. 2020;40:1355-82. Crossref
 
4. Restrepo CS, Martinez S, Lemos DF, Washington L, McAdams HP, Vargas D, et al. Imaging appearances of the sternum and sternoclavicular joints. Radiographics. 2009;29:839-59. Crossref
 
5. Depasquale R, Kumar N, Lalam RK, Tins BJ, Tyrrell PN, Singh J, et al. SAPHO: what radiologists should know. Clin Radiol. 2012;67:196-206. Crossref
 
6. Jeray KJ. Acute midshaft clavicular fracture. J Am Acad Orthop Surg. 2007;15:239-48. Crossref
 
 
 

Clinical Applications of Amino Acid Positron Emission Tomography–Magnetic Resonance Imaging in Neuro-Oncology: A Pictorial Essay

JCY Lam, SSM Lo, DYW Siu, PW Cheng

PICTORIAL ESSAY
 
Clinical Applications of Amino Acid Positron Emission Tomography–Magnetic Resonance Imaging in Neuro-Oncology: A Pictorial Essay
 
JCY Lam1, SSM Lo2, DYW Siu2, PW Cheng2
1 Department of Radiology, Tuen Mun Hospital Neuroscience Centre, Hong Kong SAR, China
2 Scanning Department, St Teresa’s Hospital, Hong Kong SAR, China
 
Correspondence: Dr JCY Lam, Department of Radiology, Tuen Mun Hospital Neuroscience Centre, Hong Kong SAR, China. Email: ljc057@ha.org.hk
 
Submitted: 24 July 2024; Accepted: 24 July 2024.
 
Contributors: All authors designed the study, acquired the data, analysed the data, drafted the manuscript, and critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: This study was approved by St Teresa’s Hospital Research Ethics Committee, Hong Kong (Ref No.: MGT-POL-008). The patients were treated in accordance with the Declaration of Helsinki. Informed consent was obtained from patients aged 18 years or older and the carers of patients aged under 18 years for all treatments and procedures, as well as for the publication of this article and the accompanying images.
 
Acknowledgement: The authors thank the research staff at the Scanning Department of St Teresa’s Hospital for their assistance in data collection.
 
 
 
 
INTRODUCTION
 
Management of an intracranial neoplasm involves sophisticated neuroimaging investigations. Magnetic resonance imaging (MRI) is important in diagnosing primary brain tumour, though it has limitations. Gadolinium-enhanced MRI can assess the morphology but does not allow determination of tumour metabolism. It also has limitations in evaluating non-enhancing gliomas. Magnetic resonance spectroscopy (MRS) provides information on the presence of neuronal and membrane metabolites. However, it has poor spatial resolution and is prone to susceptibility artefact. 18F-fluorodeoxyglucose positron emission tomography (PET)/computed tomography can give clues on tumour metabolism, yet interpretation can be unreliable due to high background brain uptake of 18F-fluorodeoxyglucose. In past decades, metabolic imaging with amino acid tracers (e.g., 11C-methionine [11C-MET] and 18F-fluoroethyl-L-tyrosine [18F-FET]) has established its added value in the non-invasive investigation of brain tumours. The pairing of amino acid PET (AA-PET) with MRI allows evaluation of both tumour morphology and corresponding metabolic activity in a single visit to the imaging institution. This pictorial review will illustrate the clinical applications of AA-PET/MRI in neuro-oncology.
 
MECHANISM OF RADIOLABELLED AMINO ACID POSITRON EMISSION TOMOGRAPHY TRACER
 
Amino acids play an essential role in many cellular processes. In addition to passive diffusion, the majority of amino acid uptake is governed by carriers such as large amino acid transporters (LATs) and the alanine-serine-cysteine transporter (ASCT). An LAT subtype, LAT1, is present at both the luminal and abluminal sides of the endothelial cell; it plays a crucial role in transporting amino acids across the blood-brain barrier. Unlike gadolinium contrast used in MRI, an intact blood-brain barrier does not limit the uptake of amino acids into an actively proliferating neoplasm.
 
Compared with healthy brain tissue, brain tumour cells significantly overexpress LAT1 and ASCT2, a subtype of ASCT, resulting in increased amino acid uptake by tumour and increased amino acid metabolism. Normal brain tissue has low expression of these transporters, resulting in the markedly lower amino acid tracer background activity and high tumour-to-normal tissue contrast in AA-PET.
 
An increased rate of metabolism in biological processes involving deoxyribonucleic acid and protein synthesis for cell growth and proliferation results in increased uptake of methionine, which involves LAT1, ASCT and ASCT2 transporters. The major limitation of 11C-MET PET study is the short half-life of the 11C-radiotracer (20 minutes). An on-site cyclotron facility is required for its production prior to the study.
 
18F-FET, another amino acid tracer, shows similar uptake and image contrast by brain tumours compared with 11C-MET. 18F-FET is metabolically inert which facilitates kinetic analysis for distinguishing high-grade from low-grade gliomas. It is easier to produce and has a longer half-life (110 minutes), making it more convenient for clinical applications.
 
DIFFERENTIATING NEOPLASMS AND NON-NEOPLASTIC LESIONS
 
11C-MET PET imaging and 18F-FET PET imaging can be used to distinguish gliomas from non-neoplastic lesions. Early diagnosis can guide timely treatment and avoid unnecessarily invasive workups, particularly for paediatric patients and for lesions in eloquent areas.
 
Based on the 2019 European guidelines,[1] qualitative and semi-quantitative evaluations can be performed with cutoff thresholds depending on clinical questions. To differentiate neoplastic from non-neoplastic tissue, the recommended cutoff thresholds for definition of biological tumour volume are: (1) a standardised uptake value (SUV) of 11C-MET PET imaging >1.3 × the mean value of healthy brain[2]; or (2) a SUV of 18F-FET PET imaging >1.6 to 1.8 × the mean value of healthy brain.[3] For 18F-FET PET imaging, the recommended threshold to differentiate between neoplastic and non-neoplastic tissue is a maximum tumour-to-background ratio (TBR) [TBRmax] of 2.5 or a mean TBR (TBRmean) of 1.9.[1] High tracer uptake with TBRmax exceeding 2.5 was found to have a high positive predictive value for detecting neoplastic lesions.[4] A commonly used threshold for 11C-MET uptake is a TBRmax of 1.3 to 1.5.[2] [5]
 
A 15-year-old patient presented with panhypopituitarism. MRI of the pituitary gland before and after gadolinium contrast showed pituitary stalk thickening with a hypoenhancing lesion involving the pituitary gland and stalk (Figure 1). 11C-MET PET/MRI showed strong tracer activity within the gland and along the stalk, suggesting an active neoplastic process (Figure 2). The diagnosis was biopsy-proven pituitary gland germinoma. The patient underwent chemoradiation. Follow-up 11C-MET PET/MRI at 3 and 6 months showed normalisation of tracer uptake in the pituitary gland (Figure 3), suggesting complete response to treatment.
 
Figure 1. Pituitary gland germinoma of a 15-year-old patient. (a) Pituitary stalk thickening with prominent size of the pituitary gland is seen on T2-weighted sagittal magnetic resonance imaging (MRI) [arrow]. (b) Hypoenhancing lesion involves the pituitary gland and stalk on post-gadolinium T1-weighted MRI [arrow].
 
Figure 2. Pituitary gland germinoma of the same patient in Figure 1. Pituitary stalk thickening with prominent pituitary gland is seen on pre-treatment T2-weighted magnetic resonance images (upper row) [arrows]. Strong 11C-methionine tracer uptake is noted within the pituitary gland and along the pituitary stalk on pre-treatment hybrid positron emission tomography–magnetic resonance images (lower row) [arrows]. (a) Axial view. (b) Sagittal view. (c) Coronal view.
 
Figure 3. Pituitary gland germinoma of the same patient in Figure 1. Normalisation of tracer uptake in the pituitary gland (arrows) is seen on 3-month (upper row) and 6-month (lower row) follow-up 11C-methionine positron emission tomography. (a) Axial view. (b) Sagittal view. (c) Coronal view.
 
A 12-year-old patient presented with left-sided weakness. Computed tomography of the brain showed a hyperdense lesion in the right basal ganglia. MRI showed an ill-defined T2-weighted hyperintense lesion in the right posterior basal ganglia and the thalamus with enhancement and restricted diffusion. No choline peak was detected on MRS (Figure 4). 11C-MET PET/MRI showed significantly increased 11C-MET tracer activity (TBRmean = 1.80; TBRmax = 2.24) [Figure 5], suggesting an active neoplastic process. The patient was treated with chemoradiation. Follow-up PET/MRI showed decreasing T2-weighted signal and no residual 11C-MET tracer activity in the right basal ganglia and the thalamus (Figures 6 and 7), suggesting complete response to treatment. For lesions in eloquent areas, AA-PET can depict the location of highest metabolic activity to indicate the most appropriate site for biopsy and increase the chance of obtaining the best representative tumour tissue. AA-PET also has advantages in detecting foci of high-grade glioma within a background of lower-grade tumour,[6] particularly when conventional MRI fails to identify heterogeneity.
 
Figure 4. Basal ganglia germ cell tumour of a 12-year-old patient. (a) Hyperdense lesion on computed tomography (arrow). (b) Infiltrative T2-weighted hyperintense lesion in the right posterior basal ganglia and the thalamus (arrow) with mild enhancement on post-gadolinium T1-weighted magnetic resonance imaging (c) [arrow]. The corresponding lesion showed restricted diffusion on diffusion-weighted imaging (d) and apparent diffusion coefficient mapping (e) [arrows]. There is no significant elevation of choline peak on magnetic resonance spectroscopy (f).
 
Figure 5. Basal ganglia germ cell tumour of the same patient in Figure 4, which is hyperintense in the right basal ganglia and the thalamus on T2-weighted magnetic resonance imaging (upper row) [arrows]. There is increased 11C-methionine tracer activity in the right basal ganglia and the thalamus (lower row) [arrows]. (a) Axial view. (b) Sagittal view. (c) Coronal view.
 
Figure 6. Basal ganglia germ cell tumour of the same patient in Figure 4. There is increased 11C-methionine tracer activity in the right basal ganglia (arrows) in pre-treatment positron emission tomography.
 
Figure 7. Basal ganglia germ cell tumour of the same patient in Figure 4. There is no residual 11C-methionine tracer activity in the right basal ganglia and the thalamus, compared with pre-treatment positron emission tomography in Figure 6.
 
With good tumour-to-background signal contrast, AA-PET/ MRI can also be performed for spinal tumours. A 50-year-old patient presented with limb weakness and numbness. MRI of the cervical spine showed syringohydromyelia with an enhancing soft tissue nodule at the C6 to C7 vertebrae (Figure 8). 18F-FET PET/MRI showed increased tracer uptake at the corresponding site of enhancing soft tissue nodule with significantly increased TBRmean of 2.02 and TBRmax of 3.38 (Figure 9), suggesting active neoplastic growth. The wall of the syrinx showed no increased tracer activity to suggest tumoural involvement. An AA-PET/MRI study in this case depicted the exact tumour site for operation. A study showed incorporation of AA-PET imaging increased the number of complete resections, which was associated with prolonged survival.[7]
 
Figure 8. Grade 2 ependymoma of a 50-year-old patient. (a) Syringohydromyelia with an inferiorly located soft tissue nodule at the C6 to C7 vertebrae is seen on T2-weighted magnetic resonance imaging (MRI) [arrow]. (b) Enhancement of the soft tissue nodule is noted on post-gadolinium T1-weighted MRI (arrow).
 
Figure 9. Grade 2 ependymoma of the same patient in Figure 8. (a) Enhancing soft tissue nodule at the C6 to C7 vertebrae (arrows) is seen on post-gadolinium T1-weighted magnetic resonance imaging (MRI) [upper row]. (b) Increased 18F-fluoroethyl-L-tyrosine (18F-FET) tracer uptake of the enhancing soft tissue nodule is noted on FET/MRI (lower row) [arrows]. There is no tracer activity along the wall of the syrinx. Images on the left show axial view while those on the right show sagittal view.
 
TUMOUR GRADING AND PERIOPERATIVE APPLICATIONS
 
A study has shown that patients with high-grade gliomas exhibit significantly higher 18F-FET tracer uptake than patients with low-grade gliomas.[4] In addition, the diagnostic performance for grading with 18F-FET PET/MRI can be improved, given that high-grade tumours frequently show characteristic dynamic data with an early time to peak (TTP) within the first 10 to 20 minutes followed by a plateau or a descent of the time-activity curve.[8] Although a reliable differentiation of World Health Organization (WHO) grade III/ IV and grade I/II gliomas is not possible because of a high proportion of active tumours among the latter, especially in oligodendrogliomas,[1] an early finding of low invasiveness of the tumour might help the neurooncologist decide on patient management. The recommended PET parameters[1] of 18F-FET PET/MRI to differentiate WHO grade I/II versus grade III/IV glioma include a TBRmax of 2.5 to 2.7, a TBRmean of 1.9 to 2.0, a TTP <35 minutes, or TAC pattern II (an early peak followed by a plateau) or III (a decreasing TAC).[1]
 
In 2021, the WHO classification of central nervous system tumours has incorporated molecular information into the diagnosis of brain tumours.[9] The grading system has been reformed and significantly restructured, especially for diffuse gliomas. The isocitrate dehydrogenase (IDH) mutation status has important diagnostic and therapeutic roles. Preoperative reliable prediction of IDH status can facilitate preliminary diagnosis of a high-grade tumour and prompt therapeutic strategies.
 
A reliable cutoff value for TBRmax or TBRmean in conventional static 18F-FET PET/MRI to differentiate IDH status is still under debate. A study with a large patient population showed a significantly shorter median TTP in IDH-wildtype gliomas compared with IDH-mutant gliomas.[10] Therefore, a short TTP in dynamic 18F-FET PET/MRI serves as a good predictor of IDH-wildtype status, particularly in non–contrast-enhancing gliomas, with high diagnostic power.[10] Another study with smaller patient populations suggested combining TTP with TBRmax to achieve higher accuracies in predicting IDH mutation status.[11] Further studies are needed to verify the role of 18F-FET PET/MRI in early detection of IDH status in glioma.
 
A 38-year-old patient presented with epilepsy. MRI of the brain showed a left temporal lobe infiltrative non-enhancing lesion with hyperintense T2-weighted signals (Figure 10). 18F-FET PET/MRI showed a significant increase in tracer uptake (TBRmean = 1.97) in the left temporal lobe (Figure 11), suggesting an active neoplastic process. Despite classical imaging features of a low-grade glioma in conventional MRI, a significant increase in tracer activity in 18F-FET PET suggests a higher-grade lesion, which may alter clinical management.
 
Figure 10. Glioblastoma of a 38-year-old patient. (a) Left temporal lobe infiltrative hyperintense lesion is seen on T2-weighted magnetic resonance imaging (MRI) [arrow]. (b) There is no enhancement on post-gadolinium T1-weighted MRI (arrow).
 
Figure 11. Glioblastoma, isocitrate dehydrogenase–wild type and telomerase reverse transcriptase mutation of the same patient in Figure 10. Images in the upper row are T2-weighted magnetic resonance imaging. There is increased 18F-fluoroethyl-L-tyrosine tracer uptake in the left temporal lobe (arrows) on positron emission tomography–magnetic resonance imaging with a mean tumour-to-background ratio above imaging thresholds (lower row). (a) Axial view. (b) Sagittal view. (c) Coronal view.
 
TUMOUR TREATMENT RESPONSE ASSESSMENT AND DIFFERENTIATION FROM TREATMENT-RELATED PSEUDOPROGRESSION
 
Early detection of high-grade tumour recurrence can be achieved by performing AA-PET/MRI with follow-up MRIs, due to the high tumour-to-normal tissue contrast. A 63-year-old patient had a history of complete removal of a right temporal lobe glioblastoma (Figure 12). A follow-up MRI 9 months after surgery showed a new enhancing focus in the left frontal lobe subependymal region. 11C-MET PET/MRI showed increased tracer uptake within the enhancing lesion, with a TBRmean of 2.66 and a TBRmax of 2.49 (Figure 13), suggesting an active neoplastic process.
 
Figure 12. (a) Right anterior temporal lobe necrotic glioblastoma of a 63-year-old patient on post-gadolinium T1-weighted magnetic resonance imaging (MRI) before treatment (arrow). (b) Complete tumour removal on postoperative MRI.
 
Figure 13. Recurrent glioblastoma of the same patient in Figure 12. (a) New enhancing nodule is seen in the left frontal lobe subependymal region on post-gadolinium T1-weighted magnetic resonance imaging (MRI) [arrow]. (b-d) Images in the upper row are T1-weighted magnetic resonance imaging. There is increased 11C-methionine tracer uptake within the enhancing lesion (arrows) on positron emission tomography–magnetic resonance imaging (lower row). (b) Axial view. (c) Sagittal view. (d) Coronal view.
 
Conventional MRI has poor sensitivity and specificity in detecting post-therapy recurrence due to its limitations in differentiating between recurrence and radionecrosis. As viable tumour cells take up 18F-FET more avidly than inflammatory cells, AA-PET offers advantages over conventional MRI, especially in haemorrhagic lesions.
 
A 53-year-old patient had a left cerebellopontine angle meningioma resected and irradiated. Follow-up MRI showed residual meningioma with postoperative changes (Figure 14). A new rim-enhancing lesion developed in the left cerebellum with central necrosis and internal haemorrhage (Figure 15). Advanced MRI techniques (i.e., MRI perfusion and MRS) did not provide useful information in the presence of haemorrhage. 18F-FET PET/MRI showed significantly increased tracer uptake along the enhancing wall of the lesion (TBRmax = 2.26; TBRmean = 1.89) [Figure 16]. The commonly used thresholds to differentiate between true progression and pseudoprogression are a TBRmax of 2.3 for early pseudoprogression, and a TBRmax or a TBRmean of 1.9 for late pseudoprogression.[1] Therefore, it suggested a high-grade active neoplastic process.
 
Figure 14. A 53-year-old patient with a history of left cerebellopontine angle (CPA) meningioma treated with resection and radiotherapy. Residual left CPA meningioma with postoperative and post-irradiation changes (arrowhead) are seen on follow-up T2-weighted magnetic resonance imaging. There is a new lesion in the left cerebellum with internal haemorrhage (arrow).
 
Figure 15. Glioblastoma of the same patient in Figure 14. Post-gadolinium T1-weighted magnetic resonance imaging shows a rim-enhancing lesion in the left cerebellum with central necrotic area (arrow).
 
Figure 16. Glioblastoma of the same patient in Figure 14. Images in the upper row are T2-weighted fluid-attenuated inversion recovery magnetic resonance imaging. Positron emission tomography–magnetic resonance imaging shows increased 18F-fluoroethyl-L-tyrosine uptake along the enhancing wall of the left cerebellar lesion (lower row) [arrows], with increased maximum and mean tumour-to-background ratios. (a) Axial view. (b) Sagittal view. (c) Coronal view.
 
FALSE POSITIVITY OF AMINO ACID POSITRON EMISSION TOMOGRAPHY WITHOUT MAGNETIC RESONANCE IMAGING
 
Several physiological and pathological causes of increased amino acid tracer uptake have been reported, including cortical ischaemia,[12] sarcoidosis,[13] haematoma[14] and abscess.[15] Vascular lesions with amino acid tracer accumulation due to slow washout may also lead to misinterpretation.[16] Molecular PET, in combination with a multiparametric MRI, can provide both structural and functional information to reduce false positive cases that might be seen on AA-PET alone.
 
A 45-year-old patient presented with ataxia. MRI of the brain showed a heterogeneous T2-weighted hyperintense cortical right cerebellar lesion with perifocal vasogenic oedema. It showed intense solid enhancement without cystic component. 11C-MET and 18F-FET PET/MRI showed strong nodular tracer uptake in the corresponding right cerebellar lesion (Figure 17). The pathological diagnosis was haemangioblastoma.
 
Figure 17. Haemangioblastoma of a 45-year-old patient. (a) Heterogeneous T2-weighted hyperintense cortical locating right cerebellar lesion (arrow) with perifocal vasogenic oedema. (b) Intense solid enhancement without cystic component is seen on post-gadolinium T1-weighted magnetic resonance imaging (arrow). Strong nodular 11C-methionine (c) and 18F-fluoroethyl-L-tyrosine (d) tracer uptake (arrows) on positron emission tomography–magnetic resonance imaging of the lesion was pathology-proven haemangioblastoma. (e) Post-gadolinium T1-wighted magnetic resonance imaging of the brain. (c-e) Axial view (upper row) and coronal view (lower row).
 
CONCLUSION
 
AA-PET has been developed for decades yet not routinely implemented in neuro-oncology. Previously, PET was criticised for its poor spatial resolution. With technological advancement, the fusion of MRI and PET images can yield additional insight beyond either examination alone by differentiating neoplastic from non-neoplastic processes, preoperatively predicting the tumour grading according to the recommended cutoff values, as well as differentiating post-treatment changes from early tumour recurrence. The location within the tumour with the highest metabolic activity can be depicted to aid biopsy and operation. Hybrid PET/MRI is more patient-friendly and offers practical advantages; however, careful interpretation and post-processing of the images by experienced operators are crucial for the accuracy and reliability of the results. Further studies are needed to evaluate the role of AA-PET, with the emerging classification of central nervous system tumours, in predicting IDH status and other radiogenomic applications in precision cancer medicine.
 
REFERENCES
 
1. Law I, Albert NL, Arbizu J, Boellaard R, Drzezga A, Galldiks N, et al. Joint EANM/EANO/RANO practice guidelines/SNMMI procedure standards for imaging of gliomas using PET with radiolabelled amino acids and [18F]FDG: version 1.0. Eur J Nucl Med Mol Imaging. 2019;46:540-57. Crossref
 
2. Kracht LW, Miletic H, Busch S, Jacobs AH, Voges J, Hoevels M, et al. Delineation of brain tumor extent with [11C]L-methionine positron emission tomography: local comparison with stereotactic histopathology. Clin Cancer Res. 2004;10:7163-70. Crossref
 
3. Pauleit D, Floeth F, Hamacher K, Riemenschneider MJ, Reifenberger G, Müller HW, et al. O-(2-[18F]fluoroethyl)-L-tyrosine PET combined with MRI improves the diagnostic assessment of cerebral gliomas. Brain. 2005;128:678-87. Crossref
 
4. Rapp M, Heinzel A, Galldiks N, Stoffels G, Felsberg J, Ewelt C, et al. Diagnostic performance of 18F-FET PET in newly diagnosed cerebral lesions suggestive of glioma. J Nucl Med. 2013;54:229-35. Crossref
 
5. Herholz K, Hölzer T, Bauer B, Schröder R, Voges J, Ernestus RI, et al. 11C-methionine PET for differential diagnosis of low-grade gliomas. Neurology. 1998;50:1316-22. Crossref
 
6. Kunz M, Thon N, Eigenbrod S, Hartmann C, Egensperger R, Herms J, et al. Hot spots in dynamic 18FET-PET delineate malignant tumor parts within suspected WHO grade II gliomas. Neuro Oncol. 2011;13:307-16. Crossref
 
7. Pirotte BJ, Levivier M, Goldman S, Massager N, Wikler D, Dewitte O, et al. Positron emission tomography–guided volumetric resection of supratentorial high-grade gliomas: a survival analysis in 66 consecutive patients. Neurosurgery. 2009;64:471-81; discussion 481. Crossref
 
8. Pöpperl G, Kreth FW, Mehrkens JH, Herms J, Seelos K, Koch W, et al. FET PET for the evaluation of untreated gliomas: correlation of FET uptake and uptake kinetics with tumour grading. Eur J Nucl Med Mol Imaging. 2007;34:1933-42. Crossref
 
9. WHO Classification of Tumours Editorial Board. WHO Classification of Tumours, 5th Edition, Volume 6: Central Nervous System Tumours. World Health Organization: 2021.
 
10. Vettermann F, Suchorska B, Unterrainer M, Nelwan D, Forbrig R, Ruf V, et al. Non-invasive prediction of IDH-wildtype genotype in gliomas using dynamic 18F-FET PET. Eur J Nucl Med Mol Imaging. 2019;46:2581-9.Crossref
 
11. Verger A, Stoffels G, Bauer EK, Lohmann P, Blau T, Fink GR, et al. Static and dynamic 18F-FET PET for the characterization of gliomas defined by IDH and 1p/19q status. Eur J Nucl Med Mol Imaging. 2018;45:443-51. Crossref
 
12. Rottenburger C, Doostkam S, Prinz M, Meckel S, Nikkhah G, Meyer PT, et al. Interesting image. Amino acid PET tracer accumulation in cortical ischemia: an interesting case. Clin Nucl Med. 2010;35:907-8. Crossref
 
13. Pichler R, Wurm G, Nussbaumer K, Kalev O, Silye R, Weis S. Sarcoidois and radiation-induced astrogliosis causes pitfalls in neuro-oncologic positron emission tomography imaging by O-(2- [18F]fluoroethyl)-L-tyrosine. J Clin Oncol. 2010;28:e753-5. Crossref
 
14. Salber D, Stoffels G, Oros-Peusquens AM, Shah NJ, Reifenberger G, Hamacher K, et al. Comparison of O-(2-18F-fluoroethyl)-L-tyrosine and L-3H-methionine uptake in cerebral hematomas. J Nucl Med. 2010;51:790-7. Crossref
 
15. Salber D, Stoffels G, Pauleit D, Oros-Peusquens AM, Shah NJ, Klauth P, et al. Differential uptake of O-(2-18F-fluoroethyl)-Ltyrosine, L-3H-methionine, and 3H-deoxyglucose in brain abscesses. J Nucl Med. 2007;48:2056-62. Crossref
 
16. Stockhammer F, Prall F, Dunkelmann S, Plotkin M, Piek J. Stereotactic biopsy of a cerebral capillary telangiectasia after a misleading F-18-FET-PET. J Neurol Surg A Cent Eur Neurosurg. 2012;73:407-9. Crossref
 
 
 

Breast Lesions in Paediatric and Young Adults: A Pictorial Essay

EH Chan, SC Woo, CM Chau, WY Fung, TKB Lai, RLS Chan, Y Leng, C Tang, NY Pan, T Wong

PICTORIAL ESSAY
 
Breast Lesions in Paediatric and Young Adults: A Pictorial Essay
 
EH Chan, SC Woo, CM Chau, WY Fung, TKB Lai, RLS Chan, Y Leng, C Tang, NY Pan, T Wong
Department of Diagnostic and Interventional Radiology, Kowloon West Cluster, Hong Kong SAR, China
 
Correspondence: Dr EH Chan, Department of Diagnostic and Interventional Radiology, Kowloon West Cluster, Hong Kong SAR, China. Email: ceh278@ha.org.hk
 
Submitted: 10 August 2024; Accepted: 25 November 2024. This version may differ from the print version.
 
Contributors: All authors designed the study. EHC, SCW and TW acquired and analysed the data. EHC drafted the manuscript. SCW, CMC, WKF, TKBL, RLSC, YL, CT, NYP and TW critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: As an editor of the journal, TW was not involved in the peer review process. Other authors have disclosed no conflicts of interest.
 
Funding/Support: This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
 
Ethics Approval: This study was approved by the Central Institutional Review Board of Hospital Authority, Hong Kong (Ref No.: PAED-2024-029). The requirement for patient consent was waived by the Board due to the retrospective nature of the study.
 
 
 
 
INTRODUCTION
 
Breast lesions can present as palpable lumps in children and young adults, causing anxiety to patients and their caregivers. Although malignant lesions are exceedingly rare in this age-group, familiarity with the spectrum of breast lesions and the diagnostic approach is crucial to guide appropriate management. Evaluation and intervention should be tailored to minimise damage to developing breast tissue. All patients with breast abnormalities should first undergo clinical assessment.[1] When imaging is indicated, ultrasound (US) is recommended as the initial radiological examination for females under 30 years of age with palpable breast masses, according to the American College of Radiology (ACR) Appropriateness Criteria.[2] Mammography is less favoured due to the ionising radiation and reduced sensitivity in dense breast tissues of young patients. Most benign lesions in young women are not visible on mammography.[2] [3] Digital breast tomosynthesis potentially increases lesion detection in overlapping tissue in young dense breasts. Magnetic resonance imaging (MRI) is used for defining disease extent, surgical planning, and screening in high-risk females with hereditary predispositions and prior irradiation.[1] [4]
 
This pictorial essay showcases both benign and malignant breast lesions in individuals under 30 years of age on multimodality imaging, with emphasis on various MRI presentations as its use in both diagnostic and screening indications has been rapidly expanding. Guidelines on screening and risk factors for early-onset breast cancer, including hereditary predispositions and prior radiotherapy, are included. The role of radiologists in follow-up imaging and the appropriate timing for image-guided intervention, while staying aware of the risks of iatrogenic injury to developing breasts, is discussed.
 
NORMAL BREAST DEVELOPMENT AND VARIANTS
 
Neonatal Breast Development
 
Breast development occurs at prenatal and pubertal stages. At the fourth week of gestation, paired ectodermal thickenings develop on the ventral surface of the embryo and extend in a line between the axilla and inguinal regions, forming the mammary crest. This is followed by involution of the mammary crest at the tenth week of gestation except at the fourth intercostal spaces, giving rise to breast buds.[3] [5]
 
Accessory breast tissue, also known as polymastia, develops when there is incomplete regression. This can be found in up to 6% of the population, usually occurring along the mammary crest and most commonly in the axilla.[1] Imaging shows heterogeneous fibroglandular tissue with characteristics similar to normal breast parenchyma[3] [6] (Figure 1). It is crucial to recognise this variant as it could be affected by the pathological processes that occur in normal breast tissues.
 
Figure 1. A 21-year-old female presented with a painful left axillary nodule. (a) Targeted ultrasound (US) shows a heterogeneous hyperechoic lesion at the left axilla similar to glandular breast tissue. (b-d) Magnetic resonance imaging (MRI) was subsequently performed as the US findings did not account for the clinical symptoms of significant pain. Axial T1-weighted (b), T2-weighted (c), and post-contrast T1-weighted MRI (d) of the left axilla showed a subcutaneous lesion with tissue signal identical to the left breast glandular tissue on all phases (arrows), consistent with accessory breast tissue.
 
Physiological Neonatal Breast Development
 
Up to 70% of newborns experience physiological breast development under maternal oestrogen influence.[3] It can be unilateral or more commonly bilateral. This condition is transient and usually resolves spontaneously by 6 months of age. Normal breast buds may fluctuate in size and remain palpable up to 2 years of age, after which they remain quiescent until puberty.[3] US features include retroareolar hypoechoic tissue (Figure 2), or hyperechoic nodule with hypoechoic linear structures representing simple branch ducts.
 
Figure 2. A 7-month-old female infant with palpable right breast mass. (a) Right breast. (b) Left breast. Ultrasound shows asymmetrical hypoechoic tissues in both retroareolar regions, more prominent on the right. There was spontaneous resolution at follow-up, consistent with physiological breast development.
 
Thelarche
 
During puberty, female breasts develop under the influence of the secretion of oestrogen and other hormones. This is known as thelarche, which is divided into five stages on the Tanner scale[5] [7] (Figure 3). On US, stage I shows ill-defined echogenic retroareolar tissue. In stage II, a central stellate hypoechoic area appears. Stage III shows central spider-like hypoechoic projections extending out from the retroareolar region, with surrounding hyperechoic glandular tissue. Stage IV involves growth of periareolar hyperechoic fibroglandular tissue with a hypoechoic central area. Finally, stage V reveals hyperechoic fibroglandular tissue and increased subcutaneous adipose tissue with disappearance of the central hypoechoic area.
 
Figure 3. Ultrasound features of Tanner stages of normal breast development. (a) Stage I: Ill-defined echogenic retroareolar tissue. (b) Stage II: Echogenic retroareolar tissue with central stellate hypoechoic area. (c) Stage III: Central spider-like hypoechoic projections extending away from the retroareolar region, with surrounding hyperechoic glandular tissue. (d) Stage IV: The retroareolar hypoechoic central area persists, with enlargement of the periareolar hyperechoic fibroglandular tissue. (e) Stage V: Mature breast appearance with hyperechoic fibroglandular tissue and increased subcutaneous adipose tissue. The central hypoechoic area is absent.
 
Premature Thelarche
 
Premature thelarche refers to isolated early breast development in girls under 8 years without associated skeletal maturation.[5] It can be unilateral or bilateral, symmetrical or asymmetrical. Imaging features are identical to thelarche, seen as developing breast tissue without discrete lesion on US.[8]
 
Gynaecomastia
 
Gynaecomastia refers to enlargement of male breast tissue, occurring most frequently during adolescence due to physiological transient increase in oestrogen levels. It typically involutes spontaneously when androgen levels rise.[3] Secondary causes include Klinefelter syndrome; drug use (e.g., anabolic steroids, exogenous oestrogens, marijuana); and tumours such as prolactinomas.[3] [5] On mammography, a flame-shaped retroareolar density is characteristic, while it is triangular and hypoechoic on US (Figure 4).[3]
 
Figure 4. A 17-year-old male presented with a 3-month history of progressively enlarging bilateral chest wall masses. (a) Right breast. (b) Left breast. Ultrasound (US) showed triangular hypoechoic areas in the subareolar region of both breasts without increased vascularity on Doppler US (arrows), consistent with gynaecomastia.
 
NON-NEOPLASTIC LESIONS
 
Trauma or Surgery-Related
 
Haematomas should be considered in patients who present with a new-onset breast lesion after recent trauma or surgery. They can be solid, cystic, or of mixed echogenicity on US, and are commonly avascular[1] [3] (Figure 5). It is crucial to look for the presence of foreign bodies, as removal may be needed.[3]
 
Figure 5. A 6-year-old girl presented with a right anterior chest wall mass after a fall injury. (a) Ultrasound (US) shows a lobulated echogenic lesion with cystic areas (arrow) beneath the right pectoralis major and minor muscles, superficial to the ribcage. (b) Colour Doppler US shows no increased vascularity. Follow-up US (not shown) shows complete resolution of the mass, consistent with haematoma.
 
Prior breast trauma can also result in fat necrosis, which may appear as solid masses to oil cysts, depending on lesion age.[3] [5] On US, they can be hyperechoic, hypoechoic with posterior acoustic enhancement, anechoic, or of mixed echogenicity with internal cystic spaces. With a typical trauma history, follow-up US in 3 to 6 months is suggested to confirm resolution.[3]
 
Galactoceles
 
Galactoceles are milk retention cysts resulting from lactiferous duct obstruction. They are predominantly seen in pregnant or lactating women and are rare in infants and adolescents. US shows a complex cystic mass with variable internal echogenicity depending on its fat and water content. Fat-fluid levels within the lesion are considered diagnostic (Figure 6).[3] [5]
 
Figure 6. A 23-year-old lactating female presented with a palpable lump in the left breast at 10 o’clock position. (a) Targeted ultrasound (US) in transverse plane shows a hypoechoic and anechoic lesion with a fat-fluid level (arrow) and posterior acoustic enhancement. (b) Doppler US study shows no internal vascularity. Fine needle aspiration confirmed the diagnosis of galactocele.
 
Cysts
 
Cysts are uncommon in paediatric patients and are usually solitary.[1] A cyst appears as an avascular anechoic lesion with thin wall and posterior acoustic enhancement on US, indicating benignity. Infected cysts may contain internal echoes, fluid-fluid levels, thickened walls, and peripheral hypervascularity.[7]
 
Infection and Inflammation
 
Mastitis refers to infection or inflammation of breast tissue. In the first 2 months of life, mastitis neonatorum can occur due to mammary ductal obstruction or skin breaks permitting bacteria seeding.[7] Puerperal mastitis can affect pregnant or breast-feeding women. The most common pathogen is Staphylococcus aureus.[7] [8] On US, mastitis may appear as focal or diffuse ill-defined heterogeneous hypoechoic and hyperechoic areas, with overlying skin thickening. Colour Doppler may show hyperaemia with central flow (Figure 7).[1] [3] [4]
 
Granulomatous mastitis may be idiopathic or due to systemic conditions, including autoimmune diseases, diabetes, or tuberculosis. These should be excluded before diagnosing idiopathic granulomatous mastitis. Over 50% of cases showed an irregular hypoechoic parallel mass with tubular extensions on US (Figure 8).[9]
 
Figure 7. A 24-year-old female presented with fever and mastalgia for 5 days during breastfeeding. Physical examination found erythema in the lower outer quadrant of the left breast. Targeted ultrasound shows an ill-defined area of altered echotexture and loss of the normal parenchymal pattern (a). The subcutaneous fat appeared hyperechoic, while the glandular parenchyma was hypoechoic with increased central blood flow on colour Doppler (b). The overlying skin was thickened and hyperechoic. The features were suggestive of puerperal mastitis.
 
Figure 8. A 27-year-old female presented with a 1-week history of increasing left breast swelling and erythema, which persisted despite antibiotics. (a) Ultrasound showed an irregular hypoechoic mass with tubular extensions (arrows) in the subareolar region of the left breast. There was associated oedema in the adjacent fibroglandular tissue and mild overlying skin thickening. (b) The mass showed peripheral vascularity on colour Doppler, mimicking a breast abscess. Incision and drainage confirmed the diagnosis of granulomatous mastitis. Acid-fast bacilli smear and culture were negative.
 
Breast abscesses are often seen as anechoic or hypoechoic lesions with debris and posterior acoustic enhancement on US. In contrast to mastitis, abscesses show only peripheral flow.[7] [8]
 
Infantile Mammary Duct Ectasia
 
Infantile mammary duct ectasia refers to retroareolar ductal dilatation in infants and young children. The exact cause is unknown.[3] Patients may be asymptomatic or present with bloody nipple discharge.[3] US demonstrates a cluster of tubular anechoic structures with or without internal debris.[3] Associated simple or multiloculated cystic lesions may also be seen.[3] The condition typically resolves after breastfeeding ceases.[3]
 
Intramammary Lymph Nodes
 
Intramammary lymph nodes, found in the breast and axillary tail, may become reactive due to inflammation, infection, or recent vaccination. Suspicious features include eccentric cortical thickening of more than 3 mm, extracapsular extension, loss of fatty hilum, or non-hilar blood flow; all require tissue sampling.[3]
 
NEOPLASTIC LESIONS
 
Vascular and Lymphatic Tumours
 
Infantile haemangioma (IH) is the most common benign neoplasm in infants and can occur in the breast.[8] [10] It is typically absent at birth, rapidly proliferates in the first few weeks to months of life and usually reaches its maximal size by 3 months of age, then spontaneously involutes from 12 months of age, with complete regression by 4 years old in most cases.[3] US or MRI are mainly for treatment planning. During proliferation, IH appears as a well-defined solid mass with a lobulated border of mixed echogenicity on US, with marked diffuse increased vascularity (Figure 9), followed by the plateau phase where it stops enlarging. Finally, IH decreases in size and vascularity during the involution phase. Echogenic areas may be identified, suggestive of fibrofatty tissue. Other vascular tumours such as congenital haemangioma and tufted angioma are uncommon.[10]
 
Figure 9. A 4-month-old female infant was noted to have a rapidly enlarging right breast mass since 2 weeks of age. Targeted ultrasound [US] (a) shows a mixed hypoechoic and hyperechoic mass in the right breast with multiple dilated vessels on colour Doppler US (b), compatible with the clinical diagnosis of infantile haemangioma.
 
Lymphangiomas are benign developmental lymphatic tumours, most frequently in the neck or axilla, but may also affect the breast. Usually presenting before 2 years of age, they appear as avascular compressible multiseptated cystic masses on US (Figure 10) and T2-hyperintense lesions without an enhancing solid component on MRI3 (Figure 11).
 
Figure 10. A 1-month-old male infant found to have a soft fluctuant right anterior chest wall mass since birth. (a) Targeted ultrasound (US) revealed a multilocular anechoic cystic mass with lobulated margins and posterior acoustic enhancement at the subcutaneous layer with no solid component (arrows). (b) Colour Doppler US shows no internal vascularity within the mass. Features were consistent with lymphangioma.
 
Figure 11. A 14-year-old boy presented with right breast swelling for 6 months. (a) T2-weighted magnetic resonance imaging (MRI) with fat saturation. (b) Post-contrast T1-weighted MRI with fat saturation. MRI shows a multiloculated T2-hyperintense mass with thin internal enhancing septations in the subcutaneous layer of the right upper anterior chest wall (arrows). No enhancing solid component was seen. Fine needle aspiration confirmed the diagnosis of lymphangioma.
 
Fibroepithelial Lesions
 
Fibroadenoma is the most common benign fibroepithelial tumour in females under 30 years of age, arising from stromal and epithelial tissues and accounting for 54% to 94% of breast masses in children and adolescents.[5] Masses reaching 5 cm are termed giant fibroadenomas.[3] [5] Juvenile fibroadenoma is an uncommon variant with hypercellular stromal proliferation that can grow rapidly and cause skin distortion.[1] [4] [5] On US, fibroadenoma typically appears as a well-circumscribed hypoechoic parallel mass with variable posterior enhancement and sometimes a pseudocapsule. Fibroadenomas in up to one-third of young breasts are vascular7 (Figure 12a).
 
Figure 12. A 17-year-old female presented with bilateral self-palpated breast masses. Initial ultrasound (US) showed multiple hypoechoic lesions in both breasts (not shown). A follow-up US was performed 1 year later. (a) In the left breast at 6 o’clock position 2 cm from the nipple, an oval parallel circumscribed and hypoechoic lesion with mild vascularity on colour Doppler US was stable in size. (b) In the left breast at 6 o’clock position in the subareolar region, an oval parallel circumscribed and hypoechoic lesion showed interval increase in size, with mild vascularity on colour Doppler US. (c) In the right breast at 12 o’clock position, an oval parallel microlobulated and hypoechoic lesion also showed interval enlargement, without increased Doppler flow. Surgical excision was performed. Pathology showed fibroadenoma and a benign phyllodes tumour in the left breast, and pseudoangiomatous stromal hyperplasia (PASH) in the right breast, corresponding with findings on US. This highlights the occasional similar appearances of fibroadenoma, phyllodes tumour, and PASH.
 
Phyllodes tumour is another fibroepithelial tumour of cellular stroma with branching leaf-like epithelium-lined cystic spaces, typically presenting as a rapid growing mass.[3] It may look sonographically identical to fibroadenoma, appearing as an oval homogeneous hypoechoic circumscribed parallel solid mass[3] (Figure 12b). Phyllodes tumours are classified as benign, borderline or malignant subtypes; however, all types may recur and metastasize, especially to the lungs.[3] In all, 85% of phyllodes tumour in children and adolescents are benign.[5] As imaging findings and fine needle aspiration do not distinguish benign from malignant phyllodes tumour, core needle biopsy is essential.[4] [5] Wide local excision with negative margins is recommended to minimise local recurrence.[3]
 
Pseudoangiomatous Stromal Hyperplasia
 
Pseudoangiomatous stromal hyperplasia is a rare benign localised stromal overgrowth, possibly mediated by hormones.[3] [5] It is usually an incidental finding on histological analysis but can also present as a lump with variable sonographic appearance, sometimes seen as an oval circumscribed hypoechoic or heterogeneous mass (Figure 12c).[3] [4] [8] Surgery is indicated for symptomatic or enlarging masses, but recurrence may occur.[3]
 
Papillomatous Lesions
 
Intraductal papilloma arises from benign epithelial proliferation of central mammary duct, projecting into and possibly obstructing the duct, causing nipple discharge at presentation.[1] It is uncommon in children and adolescents, and rare in boys.[5] [8] Typically solitary, it may appear as a well-defined solid nodule within a dilated duct on US (Figure 13),[3] often with a vascular feeding pedicle seen on colour Doppler.[11]
 
Figure 13. A 25-year-old female presented with left nipple discharge. (a) Transverse plane. (b) Longitudinal plane. Ultrasound shows a dilated central mammary duct (arrows) in the left breast periareolar region at 4 o’clock position associated with intraductal soft tissue nodule. Subsequent biopsy confirmed intraductal papilloma.
 
Juvenile papillomatosis occurs when there is localised proliferation with multiple papillomas in the peripheral ducts. Unlike intraductal papilloma, there is no fibrovascular core.[3] Ill-defined hypoechoic masses are seen on US; the presence of multiple peripheral cystic spaces with a ‘Swiss cheese’ appearance hints at the diagnosis.[4] On MRI, they are T1 hypointense showing avid enhancement, with internal T2-hyperintense cystic spaces (Figure 14).[1] [3] Although juvenile papillomatosis is benign, up to 80.4% of patients have coexisting atypical or neoplastic lesions, and it is a marker of familial breast cancer.[3] [11] This signifies the importance of close follow-up screening given the increased lifetime breast cancer risk.[1] [3] [4] [5]
 
Figure 14. A 22-year-old female with biopsy-proven papillomatosis involving both breasts. (a) On ultrasound, the largest lesion in the left breast lower inner quadrant 2 cm from the nipple is shown to be a lobulated mass with a cystic component (arrow) and an associated dilated duct (arrowheads). Transverse plane (left) and longitudinal plane (right). (b-e) Magnetic resonance imaging shows the same lesion (arrows) to be T1-hypointense with avid contrast enhancement (d) and T2-hyperintense (c). A maximum intensity projection of the post-contrast study with subtraction (e) shows multiple avidly enhancing lesions in both breasts (arrowheads), in keeping with the diagnosis of juvenile papillomatosis.
 
Primary Breast Cancer
 
Primary breast cancer is rare in paediatrics and young adults. It accounts for approximately 0.1 case per million in females younger than 20 years old, and even less in males.[1] Approximately half of the patients under 30 years old with breast cancer harbour a germline mutation, such as BRCA1/2, TP53 for Li-Fraumeni syndrome, and PTEN for Cowden syndrome.[12] Hence, the diagnosis of breast cancer in young patients should prompt genetic testing and counselling.[1] [3] Individuals over the age of 25 years from a family with known BRCA1/2 mutation carriers should undergo genetic testing. All females with a lifetime breast cancer risk of over 20% are recommended to begin undergoing annual screening MRIs from the age of 25 years with additional annual mammography from the age of 30 years.[13]
 
In 2020, the International Guideline Harmonization Group recommends breast cancer screening in females with a history of chest radiotherapy with radiation dose of over 10 Gy, or previous upper abdominal radiotherapy, given the increased risk for breast cancer.[14] They include childhood cancer survivors such as those with supradiaphragmatic Hodgkin lymphoma who underwent chest irradiation, and haematopoietic cell transplant recipients who had total body irradiation. The elevated risk begins 8 years after treatment and remains increased beyond 40 years.[14] These cancer survivors who develop breast cancer after radiotherapy are reported to have higher mortalities than women with de novo breast cancer in the general population.[14] The National Comprehensive Cancer Network Clinical Guidelines suggest that annual breast MRI and mammography should begin 10 years after treatment, but not before age 25 years and 30 years, respectively, while the Children’s Oncology Group Guidelines (2018 version 5) recommend annual mammography and breast MRI to commence 8 years after treatment or at 25 years of age, whichever is later.[14]
 
Radiological features considered suspicious in paediatrics are no different from adults. On US, concerning features include spiculated margins, microlobulation, marked hypoechogenicity, and not being parallel to the chest wall (Figure 15). On mammography, an irregular, high-density mass with spiculated or indistinct borders; and microcalcifications with fine pleomorphic, linear, or linear branching morphology; and linear or segmental distribution are worrisome for malignancy (Figure 16). Suspicious MRI findings include an irregular mass with spiculated margins and heterogeneous enhancement; or clumped, heterogeneous, or homogeneous non-mass enhancement with linear or segmental distribution; and plateau or washout enhancement kinetics. However, there is overlap of enhancement kinetics between benign and malignant lesions, and persistent enhancement cannot exclude malignancy[15] (Figure 17).
 
Figure 15. A 26-year-old female presented with a right breast mass. Ultrasound shows two closely related microlobulated hypoechoic lesions at 8 o’clock position (arrows). Core biopsy showed atypical ductal hyperplasia. On surgical excision, the histological diagnosis was low-grade intraductal carcinoma arising in a fibroadenoma. In view of this early-onset breast cancer, genetic testing was performed and found the pathogenic PTEN variant, consistent with Cowden syndrome.
 
Figure 16. A 21-year-old female presented with a highly suspicious right breast mass. Mammography with craniocaudal (a) and mediolateral oblique views (b) show an irregular high-density mass in the upper outer quadrant, associated with fine pleomorphic microcalcifications (arrows). Ipsilateral axillary lymphadenopathy was present. The biopsy and surgical specimens yielded invasive ductal carcinoma with ipsilateral axillary nodal metastasis (arrowheads in [b]).
 
Figure 17. A 28-year-old female with known Li-Fraumeni syndrome and a family history of breast cancer. She had a history of left ovarian teratoma and right primary ovarian neuroectodermal tumour treated with salpingo-oophorectomy at 12 years and 13 years of age, respectively. A germline TP53 gene mutation was detected; therefore, she underwent magnetic resonance imaging (MRI) screening for breast cancer (a). A T1-weighted post-contrast MRI with subtraction shows linear non-mass enhancement in the left breast 11 o’clock position (arrow) which demonstrated a type I enhancement curve, new since her prior MRI 2 years ago (b). There was no mammographic or sonographic correlation. MRI-guided vacuum-assisted biopsy showed atypical glands. Surgical excision was performed and confirmed high-grade ductal carcinoma in situ, indicating that a type I–enhancing kinetic curve does not exclude a malignancy (c).
 
Other Breast Malignancies
 
Breast metastases are more common than primary breast malignancies in paediatric patients, most frequently from rhabdomyosarcoma (Figure 18), followed by neuroblastoma, haematological malignancies including lymphoma and leukaemia, and Ewing sarcoma.[1] [3] [4] Breast metastases are usually large and solitary with variable US features, which can be irregular or lobulated, heterogeneous, and hypoechoic with hyperechoic foci.[1] [8] Rhabdomyosarcoma and Ewing sarcoma can also involve the breast directly as a primary chest wall malignancy, where evaluation of disease extent with cross-sectional imaging is often helpful.[1] [3] Lymphoma, most commonly non–Hodgkin lymphoma, can affect the breast and ipsilateral axillary lymph nodes primarily, but is exceedingly rare due to the lack of lymphoid tissue in the breast.[3]
 
Figure 18. A 29-year-old female with biopsy-proven alveolar rhabdomyosarcoma in the nasopharynx (arrow) as shown on a sagittal post-contrast T1-weighted image (a). (b, c) A staging 18F-fluorodeoxyglucose positron emission tomography–computed tomography shows a hypermetabolic mass at 3 o’clock position in the left breast (arrowheads). Subsequent biopsy and left mastectomy confirmed a rhabdomyosarcoma metastasis in the left breast.
 
Next Step of Management: When to Biopsy
 
According to the ACR Appropriateness Criteria,[2] US is the most appropriate radiological procedure for initial evaluation of palpable breast masses in females under 30 years of age. Lesions with benign US features can be followed up clinically. Sonographic features of benign breast lesions include circumscribed margins, orientation parallel to the skin, and less than three gentle smooth lobulations. Short interval follow-up is recommended for probably benign lesions.[2]
 
Developing breast buds in paediatric patients are vulnerable to injury from biopsy, with potential long-term consequences including permanent disfiguration. Therefore, image-guided biopsy should be carefully considered and discussed. Biopsy should be reserved for probably benign masses smaller than 4 cm showing atypical US features or rapid enlargement, probably benign masses that are larger than 4 cm, or masses that demonstrate malignant features on US.[1] In high-risk patients with known genetic mutations, prior irradiation, or extramammary malignancies presenting with an enlarging breast mass, biopsy should be considered even if the US findings appear benign.[1] [3] Core biopsy is preferred over fine needle aspiration due to higher sensitivity, specificity, and accuracy in histological grading, while tumour receptor status can also be tested.[2] Surgical excision may be indicated for rapidly enlarging or symptomatic breast masses even if they show benign radiological features or biopsy results, as phyllodes tumours cannot be excluded.[1]
 
CONCLUSION
 
The majority of breast lesions in females under 30 years of age are benign, but malignancies do occur. Radiologists must be familiar with the diagnostic approach and able to identify lesions suitable for follow-up to minimise unnecessary intervention. Prior to biopsy, the potential long-term consequences on breast development in young patients must be considered. When early-onset breast cancer is suspected or diagnosed, it is important not only to review the patient’s medical history but also to explore possible hereditary predispositions.
 
REFERENCES
 
1. Gao Y, Saksena MA, Brachtel EF, terMeulen DC, Rafferty EA. How to approach breast lesions in children and adolescents. Eur J Radiol. 2015;84:1350-64. Crossref
 
2. Expert Panel on Breast Imaging; Moy L, Heller SL, Bailey L, D’Orsi C, DiFlorio RM, et al. ACR Appropriateness Criteria ® Palpable Breast Masses. J Am Coll Radiol. 2017;14(5S):S203-24. Crossref
 
3. Harper LK, Simmons CL, Woodard GA, Solanki MH, Bhatt AA. Pictorial review of common and uncommon pediatric breast lesions. Radiographics. 2023;43:e220117. Crossref
 
4. Kaneda HJ, Mack J, Kasales CJ, Schetter S. Pediatric and adolescent breast masses: a review of pathophysiology, imaging, diagnosis, and treatment. AJR Am J Roentgenol. 2013;200:W204-12. Crossref
 
5. Lee EJ, Chang YW, Oh JH, Hwang J, Hong SS, Kim HJ. Breast lesions in children and adolescents: diagnosis and management. Korean J Radiol. 2018;19:978-91. Crossref
 
6. DeFilippis EM, Arleo EK. The ABCs of accessory breast tissue: basic information every radiologist should know. AJR Am J Roentgenol. 2014;202:1157-62. Crossref
 
7. García CJ, Espinoza A, Dinamarca V, Navarro O, Daneman A, García H, et al. Breast US in children and adolescents. Radiographics. 2000;20:1605-12. Crossref
 
8. Chung EM, Cube R, Hall GJ, González C, Stocker JT, Glassman LM. From the archives of the AFIP: Breast masses in children and adolescents: radiologic-pathologic correlation. Radiographics. 2009;29:907-31. Crossref
 
9. Pluguez-Turull CW, Nanyes JE, Quintero CJ, Alizai H, Mais DD, Kist KA, et al. Idiopathic granulomatous mastitis: manifestations at multimodality imaging and pitfalls. Radiographics. 2018;38:330-56. Crossref
 
10. Inarejos Clemente EJ, Diaz Leyva J, Karakas SP, Duarte AM, Mas TR, Restrepo R. Radiologic and clinical features of infantile hemangioma: potential pitfalls and differential diagnosis. Radiographics. 2023;43:e230064. Crossref
 
11. Eiada R, Chong J, Kulkarni S, Goldberg F, Muradali D. Papillary lesions of the breast: MRI, ultrasound, and mammographic appearances. AJR Am J Roentgenol. 2012;198:264-71. Crossref
 
12. Cathcart-Rake EJ, Ruddy KJ, Bleyer A, Johnson RH. Breast cancer in adolescent and young adult women under the age of 40 years. JCO Oncol Pract. 2021;17:305-13. Crossref
 
13. Paluch-Shimon S, Cardoso F, Sessa C, Balmana J, Cardoso MJ, Gilbert F, et al. Prevention and screening in BRCA mutation carriers and other breast/ovarian hereditary cancer syndromes: ESMO clinical practice guidelines for cancer prevention and screening. Ann Oncol. 2016;27(suppl 5):v103-10. Crossref
 
14. Gao Y, Perez CA, Chhor C, Heller SL. Breast cancer screening in survivors of childhood cancer. Radiographics. 2023;43:e220155. Crossref
 
15. Macura KJ, Ouwerkerk R, Jacobs MA, Bluemke DA. Patterns of enhancement on breast MR images: interpretation and imaging pitfalls. Radiographics. 2006;26:1719-34. Crossref