eISSN: 2081-2841
ISSN: 1689-832X
Journal of Contemporary Brachytherapy
Current Issue Archive Supplements Articles in Press Journal Information Aims and Scope Editorial Office Editorial Board Register as Author Register as Reviewer Instructions for Authors Abstracting and indexing Subscription Advertising Information Links
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank

6/2022
vol. 14
 
Share:
Share:
Original paper

Computed tomography-guided iodine-125 radioactive seed implantation in small-cell lung cancer: A retrospective study

Mengjie Li
1
,
Peng Liu
1
,
Huan Wang
1
,
Biao Wang
1
,
Jie Zhou
1
,
Yuzhen Jin
1
,
Cuicui Wang
1
,
Jian Yang
2
,
Ping Zhang
2

1.
Department of Respiratory and Critical Care Medicine, Cangzhou Fifth Hospital (People’s Hospital of Qingxian), Cangzhou 062650, China
2.
Precision Radiotherapy Center, Cangzhou People’s Hospital, Cangzhou 061001, China
J Contemp Brachytherapy 2022; 14, 6: 536–541
Online publish date: 2022/12/30
Article file
- Computed tomography.pdf  [0.25 MB]
Get citation
 
 

Purpose

Lung cancer is the second most common cancer worldwide [1], and is generally categorized into small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC), according to the morphology of the cancer cells [2]. SCLC is an aggressive epithelial tumor characterized by a high propensity for early development, which accounts for about 15% of all lung cancers [3, 4]. Approximately, two-thirds of SCLC patients develop metastatic diseases with limited treatment options, suggesting that SCLC remains one of the most lethal malignancies [2]. Radiotherapy and chemotherapy are the cornerstones of SCLC treatment, and a combination of immune checkpoint inhibitors can also be employed to treat extensive SCLC [3, 5]. Etoposide and platinum-doublet chemotherapy is recommended as the standard first-line therapy, and combination chemotherapy with other agents, including carboplatin (paraplatin) and irinotecan (camptosar), is considered the standard second-line treatment [4, 6]. In patients with limited-stage SCLC (LS-SCLC), the combination of chemotherapy and radiotherapy may improve local control as well as overall survival (OS). However, for patients with extensive-stage SCLC (ES-SCLC), who make up the majority of SCLC patients in clinical practice, the most common initial chemotherapy regimen remains etoposide plus cisplatin (EP regimen), and no novel chemotherapy combinations have shown superior efficacy [3, 6]. Therefore, there is an urgent need for a safe and effective treatment to improve the clinical prognosis of SCLC patients.

Iodine-125 (125I) radioactive seed implantation therapy is a local treatment that has been widely used in treatment of various malignancies [7-9]. With the advantages of low doses, small adverse reactions, and minimal invasiveness, it has been increasingly applied in treating NSCLC, achieving certain efficacy [10-12]. However, this method is rarely used in SCLC treatment. Therefore, this study retrospectively analyzed data of SCLC patients treated with 125I seed implantation, and evaluated its’ efficacy and the corresponding complications.

Material and methods

Patient data

A total of 12 SCLC patients, who were admitted to the respiratory and critical care medicine department of our hospital from September 2014 to September 2018 were enrolled into this study. Inclusion criteria: (1) Patients aged ≥ 18 years; (2) Diagnosed with SCLC by fiberoptic bronchoscopy or percutaneous lung biopsy; (3) Those who received 125I radioactive seed implantation. Patients with respiratory failure, heart failure, severe arrhythmia, severe emphysema, high-risk of bulla puncture, and severe coagulation dysfunction as well as patients with incomplete data or follow-up information were not considered for this retrospective analysis. Clinical characteristics of the patients at baseline are shown in Table 1. Characteristics of each patient are presented in Tables 2 and 3, which included TNM stage, tumor location and size, prescription dose, previous treatment, and reasons for omitting standard treatment.

Table 1

Patients’ characteristics

CharacteristicsValue (n)
Gender
Male5
Female7
Age (years), mean (range)64.4 (55-80)
VALSG stage
Limited1
Extensive11
Treatment history
Chemotherapy9
Radiotherapy1
None2
Tumor size (cm), mean (range)4.5 (3-7)
Clinical presentation
Cough10
Hemoptysis2
Difficulty in feeding2
Weakness3
Chest tightness and dyspnea6
Chest pain2
Infection3
Complications
Atelectasis3
Superior vena cava syndrome2
Mediastinal lymph node enlargement and compression7
Table 2

Characteristics of patients with 125I seed implantation

Patient numberGenderAge (years)KPS scoreStageTNM stageTumor locationTumor size (cm)Prescription dose, PD (Gy)Surgical lesion numberSeed numberOS (months)
1Female5590ExtensiveT3N3M1a IVaRight lower lobe of the lung; No. 10 lymph nodes metastasis; No. 7 lymph nodes metastasis4.3 × 5.4, 4.3 × 5.2, 3.2 × 3.5110315, 39, 1624
2Male5670LimitedT3N3M0 IIIcRight lower lobe of the lung5.2 × 5.511014026
3Male6880LimitedT3N2M0 IIIbLeft upper lobe of the lung6.2 × 5.512016029
4Male6170ExtensiveT2aN2M1c IVbLeft lower lobe of the lung3.8 × 4.31001265
5Female6890LimitedT3aN0M0 IIbLeft lower lobe of the lung; No. 10 lymph nodes metastasis5.2 × 6.3, 3.7 × 4.2110254, 3571
6Female6370LimitedT3N3M0 IIIcRight upper lobe of the lung6.5 × 5.111016018
7Female8080LimitedT2aN0M0 IbLeft upper lobe of the lung3.0 × 3.6100120> 69
8Female6780ExtensiveT3N3M1a IVaRight upper lobe of the lung5.7 × 6.41101507
9Female6640ExtensiveT2bN2M1a IVaRight lower lobe of the lung; No. 1 lymph nodes metastasis4.5 × 3.6, 2.8 × 2.3100330, 26, 139
10Female7350ExtensiveT2aN3M1c IVbLeft upper lobe of the lung; No. 7 lymph nodes metastasis3.6 × 3.7, 5.7 × 6.8120228, 4012
11Male6090LimitedT4N3M0 IIIcRight upper lobe of the lung7.0 × 6.310015812
12Male5660ExtensiveT2bN2M1c IVbLeft upper lobe of the lung; Right upper lobe of the lung3.5 × 3.7, 4.3 × 3.1100224, 287

[i] OS – overall survival

Table 3

Previous treatments of patients with 125I seed implantation and reason for omitting a standard treatment

Patient numberPrevious treatment and reason for omitting a standard treatment
1The patient progressed after 6 cycles of EP regimen chemotherapy. Grade 4 myelosuppression occurred and radiotherapy was refused. Iodine-125 seeds were implanted and then metastasized into the lung.
2Two cycles of EP regimen chemotherapy were ineffective. Due to the occurrence of IV degree myelosuppression, the patient was unable to continue chemotherapy and refused radiotherapy. Iodine-125 seeds were implanted and then metastasized into the liver.
3Two cycles of EP regimen chemotherapy were ineffective. Grade 3 myelosuppression occurred in chemotherapy, and radiotherapy was refused. Iodine-125 seeds were implanted, followed by pleural metastasis and peritoneal metastasis.
4The patient progressed after 5 cycles of EP regimen chemotherapy and atelectasis occurred. Due to the occurrence of grade 4 myelosuppression, the patient refused radiotherapy. Iodine-125 seeds were implanted and then the primary lesions progressed.
5Oral Chinese medicine was ineffective, the patient refused radiotherapy and chemotherapy; the primary lesion increased. Iodine-125 seeds were implanted, regular re-examination found a dose cold area, and then the EP regimen for a total of 8 cycles of chemotherapy was applied, followed by liver metastasis.
6One cycle of EP regimen chemotherapy; the patient refused radiotherapy. Iodine-125 seeds were implanted and post-operative 6 cycles of EP regimen chemotherapy were given. Then, intra-pulmonary metastasis and supra-clavicular lymph node metastasis occurred.
7Progressed after 4 cycles of EP regimen chemotherapy, the patient refused radiotherapy because of grade 4 myelosuppression, and 125I seeds were implanted.
8Progressed after 7 cycles of IP regimen chemotherapy, and the patient refused radiotherapy due to grade 4 myelosuppression. Iodine-125 seeds were implanted, and posterior pleural metastasis and liver metastasis occurred.
9Two cycles of EP regimen chemotherapy were ineffective; the patient refused radiotherapy. KPS score during seed implantation was 40, which indicated intolerant to radiotherapy and chemotherapy, and then intra-pulmonary metastasis occurred.
10KPS score 50, intolerant to radiotherapy and chemotherapy. Iodine-125 seeds were implanted and then, posterior head skin metastasis, rib metastasis, and abdominal metastasis occurred.
11Progressed after 6 cycles of EP regimen chemotherapy and radiotherapy. Iodine-125 seeds were implanted and then, posterior pleural metastasis and liver metastasis occurred.
12Grade 4 myelosuppression occurred after the first cycle of EP regimen chemotherapy; the patient refused radiotherapy. KPS score before seed implantation was 60, which indicated intolerant to radiotherapy and chemotherapy. Iodine-125 seeds were implanted and then, hindbrain metastasis, liver metastasis, and bone metastasis occurred.

Instruments and equipment

For 125I radioactive seed implantation, 18 G implantation needles and a turntable implantation gun were obtained from HAKKO Medical Co., Ltd. (Nagano, Japan) and Zhuhai Hejia Medical Equipment Co., Ltd. (Zhuhai, China), respectively. 125I seeds were produced by Beijing ZHIBO Bio-Medical Technology Co., Ltd. (Beijing, China), and were fully enclosed in a titanium shell, with a length of 4.5 mm, a diameter of 0.8 mm, a half-life of 60.1 d, and an activity of 0.7 mCi. Seed implantation was guided by a Siemens 32-row helical computed tomography (CT) scanner (Siemens, Erlangen, Germany), with a slice thickness of 5 mm.

Pre-operative preparation

Before 125I seed implantation, patients underwent a blood routine examination, accompanied by withdrawal of antiplatelet and anticoagulant drugs. An enhanced chest CT examination was performed to clarify the lesion scope and its’ relationship with blood vessels as well as the route of needle insertion. Then, treatment planning system (TPS; Beijing Tianhang Kelin Technology Development Co., Ltd., Beijing, China) was applied to determine the number and distribution of 125I seeds. Prescribed dose was 100-120 Gy. Prior to implantation, codeine was given to relieve cough, and diazepam was applied for sedation according to a patient’s condition. Those with a high-risk of bleeding were administered hemostatic drugs, such as intra-muscular injection of hemagglutinin, and intravenous access was established.

CT-guided 125I radioactive seed implantation

Patients were placed in a supine, prone, or lateral position, depending on the lesion location. Oxygen inhalation and ECG monitoring were given during the operation, and CT scans were conducted to determine the tumor site and the needle insertion point of 125I seeds, with a line spacing of 1-1.5 cm and particle spacing of 0.5-1 cm. After routine skin disinfection with iodophor, a sterile hole towel was laid, and the puncture point area was anesthetized with local infiltration of 2% lidocaine. The syringe needles were indwelled after local anesthesia at all puncture points. Next, a CT scan was performed to examine whether the puncture point and puncture angle were correct. Then, the syringe needle was pulled out, and the implant needle was punctured at a suitable point and angle. After confirming the correct puncture location by a CT scan, 125I seeds were implanted and post-operative verification was conducted. Subsequently, the needle was removed, and the puncture point was covered and fixed with sterile gauze and tape. If there was a bleeding from the puncture point, compression was carried out.

Outcomes and assessments

Post-operative complications were closely monitored, and all patients were reviewed every 2 months. Patients were followed up until death, last visiting time, or the end of study. The efficacy was evaluated using response evaluation criteria in solid tumors (RECIST) guideline, version 1.1 [13]: (1) Complete response (CR): All target lesions disappeared and short-axis of any pathological lymph node (whether target or non-target) must be less than 10 mm; (2) Partial response (PR): A reduction of at least 30% in the sum of target lesion diameters compared with that of baseline sum diameters; (3) Progressive disease (PD): At least 20% of an increase in the sum of target lesion diameters compared with the smallest sum in the study (including the baseline sum if that was the smallest in the study). In addition to the relative increase of 20%, the absolute increase in the sum of diameters must be greater than 5 mm. The appearance of one or more new lesions was also considered progression; (4) Stable disease (SD): Neither sufficient shrinkage to qualify for PR nor enough increase to qualify for PD, the smallest sum diameters were used as a reference for the study. Overall response rate (ORR) = (CR + PR)/total number of cases × 100%. Local control rate (LCR) = (CR + PR + SD)/total number of cases × 100%. OS was defined as the time from the start of treatment to the death of a patient due to any cause. Progression-free survival (PFS) was considered the period between implantation and the date of recurrence, loco-regional progression, metastasis, or death.

Statistical analysis

Data were analyzed by SPSS version 20.0 (SPSS, Chicago, IL, USA). Continuous data were presented as mean (minimum-maximum) and categoric data were shown as absolute numbers (percentages). Median OS and PFS were estimated using Kaplan-Meier method.

Results

Characteristics of iodine-125 seed implantation

All patients were successfully implanted with 125I radioactive seeds. As depicted in Table 2, a total of 662 125I seeds were implanted into 19 tumor lesions among the 12 patients, with a range of 20-89 seeds implanted in each patient. One of the cases 9 was found to have a dose cold zone, localized, with a supplemental implantation of 13 seeds.

Efficacy

The ORR was 83.3%, 63.6%, 50%, and 40% at 2, 6, 12, and 24 months after implantation, respectively (Table 4). The LCR at 1 and 2 years was 75% (6/8) and 60% (3/5), respectively. The patient’s treatment results are shown in Figure 1. Of the 12 patients, 2 patients with superior vena cava syndrome had complete remission, and 2 of the 3 patients with atelectasis had lung recruitment.

Table 4

Efficacy evaluation of 125I radioactive seed implantation

Time after surgery (months)nCRPRSDPDORR (%)LCR (%)
212462083.3100.0
611343163.690.9
128222250.075.0
245111240.060.0

[i] CR – complete response; PR – partial response; SD – stable disease; PD – progressive disease; ORR – overall response rate; LCR – local control rate

Fig. 1

CT images showing particle distribution after 125I radioactive seed implantation in small-cell lung cancer (SCLC) patients and treatment after 3 months. A) After 125I radioactive seed implantation, a few bleeding complications appeared in the dorsal lung of the tumor. B) Three months after 125I radioactive seed implantation, the tumor gradually shrunk, and the gathered particles could be seen at the original tumor site: mild radiation pneumonia in the lung tissue around the tumor

/f/fulltexts/JCB/49843/JCB-14-49843-g001_min.jpg

Survival analysis

As shown in Figure 2, the patients who received 125I seed implantation had a median OS of 12 months (range, 5-71 months). Moreover, the OS rate at 6, 12, and 24 months after implantation was 91.67%, 66.67%, and 41.67%, respectively (Table 2). The median PFS was 8 months (range, 3-69 months).

Fig. 2

Kaplan-Meier curve showing overall survival of 12 small-cell lung cancer (SCLC) patients treated with 125I seed implantation

/f/fulltexts/JCB/49843/JCB-14-49843-g002_min.jpg

Surgical complications

No surgery-related death occurred. During the follow-up period, mild complications were observed, including 2 (16.67%) cases of cough aggravation, 1 (8.33%) case of hemoptysis, and 2 (16.67%) cases of pneumothorax.

Discussion

Iodine-125 seed implantation therapy was first used in the treatment of prostate cancer, and with the continuous innovation and improvement of medical technology, it is now widely used in treating various malignancies. Its’ efficacy has been fully verified in the treatment of NSCLC [10-12]. A meta-study on NSCLC showed significant differences in ORR, disease control rate (DCR), and OS between 125I brachytherapy in combination with chemotherapy and chemotherapy alone, confirming the clinical benefit of 125I seed implantation for NSCLC treatment [14]. Additionally, lung squamous cell carcinoma is sensitive to 125I seed irradiation, which exhibits significant therapeutic effects. However, there is little information on other pathologic lung tumor types irradiated by 125I seed irradiation in addition to NSCLC [15]. In the current study, 12 SCLC patients received 125I seed implantation, and it was found that tumor growth was inhibited and the quality of life improved. The 125I radioactive particles can continuously release low-dose γ-rays to break molecular chains and chemical bonds of biological macro-molecules, thereby destructing DNA, RNA, proteins, and various catalytic enzymes, with no or only slight damage to normal tissues. In addition, in the tumor micro-environment, 125I irradiation inhibits cell proliferation by reducing the level of Ki-67, and induces apoptosis by increasing the P21 level and reducing the surviving level in vivo [15]. Therefore, inhibition of cell proliferation and induction of apoptosis by 125I irradiation is the key mechanism for the therapeutic effect of 125I seed implantation [16].

Unlike other types of lung cancer, SCLC is initially sensitive to chemotherapy and radiotherapy. However, despite the sensitivity to radiotherapy and chemotherapy, patients with newly diagnosed extensive-stage SCLC generally have a poor prognosis, with a median PFS of 5.5 months and a median OS of 10 months [17]. According to a previous study, the low OS rate in highly chemotherapy-sensitive SCLC populations is due to the rapid development of drug resistance and the failure of second-line and follow-up treatments to affect or slow tumor growth and metastasis [18]. For decades, both ESMO [6] and NCCN [4] clinical treatment guidelines have recommended chemotherapy containing EP as the standard treatment for ES-SCLC. An earlier paper reported that the average response rate of patients treated with the EP regimen ranged from 60% to 80%, with a median OS of 8-10 months [19]. In this research, patients receiving 125I seed implantation had an ORR of 83.3%, 63.6%, 50%, and 40% at 2, 6, 12, and 24 months, respectively. Furthermore, the median PFS was 8 months, the median OS was 12 months, and the mean OS was 23.6 months, which was superior to the efficacy of standard treatment. In addition, a randomized phase 3 CREST trial published in 2015 [20] evaluating thoracic radiotherapy for ES-SCLC, revealed that 247 patients who received thoracic radiotherapy (30 Gy in ten fractions) had a 1-year OS rate of 33% and 2-year OS rate of 13% as well as a 6-month PFS rate of 24%. In this study, the incidence of OS at 12 and 24 months after the surgery was 66.67% and 41.67%, respectively, which was higher than that of ES-SCLC patients, who received general systemic treatment or chest radiotherapy after chemotherapy [20]. Nevertheless, more studies are still needed to compare thoracic radiotherapy and high-dose-rate brachytherapy (HDR-BT) with 125I seeds brachytherapy for the treatment of SCLC. However, HDR-BT lacks experience and data from large phase III clinical trials in lung cancer, since most studies have focused on NSCLC, with few data on SCLC. In addition, the quality control of HDR-BT technology is difficult to be guaranteed due to the characteristics of personal proficiency and individualized operation techniques.

Although 125I seed implantation has the advantages of minimal invasiveness, quick recovery, and few adverse reactions, there are various complications, such as worsening cough, pneumothorax, and hemoptysis. Generally, patients who cannot tolerate radiotherapy or chemotherapy and refuse radiotherapy and chemotherapy, present with tumor emergencies, and whose disease has progressed after radiotherapy and chemotherapy, could benefit from 125I seed implantation therapy. However, for SCLC, 125I seed implantation is the only local treatment, and systemic treatment is still required.

Conclusions

Iodine-125 seed implantation is a safe and effective supplementary treatment for patients with small-cell lung cancer, who cannot tolerate radiotherapy.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of Cangzhou Fifth Hospital (People’s Hospital of Qingxian) (No. 201407141), and all patients have signed written informed consent.

Funding

This work was supported by the Cangzhou Key R&D Plan Guidance Project (No: 183302131) as well as the Cangzhou Science and Technology Plan Self-funded Project (No: 204106141).

Disclosure

The authors report no conflict of interest.

References

1 

Sung H, Ferlay J, Siegel RL et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209-249.

2 

Thai A, Solomon B, Sequist L et al. Lung cancer. Lancet (London, England) 2021; 398: 535-554.

3 

Rudin C, Brambilla E, Faivre-Finn C et al. Small-cell lung cancer. Nat Rev Dis Primers 2021; 7: 3.

4 

Ganti A, Loo B, Bassetti M et al. Small cell lung cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19: 1441-1464.

5 

Hiddinga B, Raskin J, Janssens A et al. Recent developments in the treatment of small cell lung cancer. Eur Respir Rev 2021; 30: 210079.

6 

Dingemans A, Früh M, Ardizzoni A et al. Small-cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2021; 32: 839-853.

7 

Huo X, Huo B, Wang H et al. Percutaneous computed tomography-guided permanent 125I implantation as therapy for pulmonary metastasis. J Contemp Brachytherapy 2018; 10: 132-141.

8 

Chen W, Luo Y, Zhang Y et al. Ultrasound-guided implantation of radioactive I seed in radioiodine refractory differentiated thyroid carcinoma. BMC Cancer 2021; 21: 834.

9 

Li J, Zhang L, Xu W et al. Computed tomography-guided implantation of 125I seeds brachytherapy for recurrent multiple pulmonary oligometastases: initial experience and results. J Contemp Brachytherapy 2017; 9: 132-138.

10 

Huo X, Huo B, Wang H et al. Implantation of computed tomography-guided Iodine-125 seeds in combination with chemotherapy for the treatment of stage III non-small cell lung cancer. J Contemp Brachytherapy 2017; 9: 527-534.

11 

Yang D, Lin Y, Xue C et al. CT-guided percutaneous implantation of I particles in treatment of early lung cancer. J Thorac Dis 2020; 12: 5996-6009.

12 

Liu B, Wang Y, Tian S et al. Salvage treatment of NSCLC recurrence after first-line chemotherapy failure: Iodine-125 seed brachytherapy or microwave ablation? Thorac Cancer 2020; 11: 697-703.

13 

Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009; 45: 228-247.

14 

Zhang W, Li J, Li R et al. Efficacy and safety of iodine-125 radioactive seeds brachytherapy for advanced non-small cell lung cancer-A meta-analysis. Brachytherapy 2018; 17: 439-448.

15 

Jin Q, Lin C, Zhu X et al. (125)I seeds irradiation inhibits tumor growth and induces apoptosis by Ki-67, P21, survivin, livin and caspase-9 expression in lung carcinoma xenografts. Radiat Oncol 2020; 15: 238.

16 

Zhu Y, Dong M, Yang J et al. Evaluation of iodine-125 interstitial brachytherapy using micro-positron emission tomography/computed tomography with 18F-fluorodeoxyglucose in hepatocellular carcinoma HepG2 xenografts. Med Sci Monit 2019; 25: 371-380.

17 

Kalemkerian GP, Loo BW, Akerley W et al. NCCN Guidelines Insights: small cell lung cancer, Version 2.2018. J Natl Compr Canc Netw 2018; 16: 1171-1182.

18 

Lally BE, Urbanic JJ, Blackstock AW et al. Small cell lung cancer: have we made any progress over the last 25 years? Oncologist 2007; 12: 1096-1104.

19 

Kim DW, Kim HG, Kim JH et al. Randomized phase III trial of irinotecan plus cisplatin versus etoposide plus cisplatin in chemotherapy-naive korean patients with extensive-disease small cell lung cancer. Cancer Res Treat 2019; 51: 119-127.

20 

Slotman B, van Tinteren H, Praag J et al. Use of thoracic radiotherapy for extensive stage small-cell lung cancer: a phase 3 randomised controlled trial. Lancet (London, England) 2015; 385: 36-42.

Copyright: © 2022 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.