Open Access

Radionecrosis mimicking pseudo‑progression in a patient with lung cancer and brain metastasis following the combination of anti‑PD‑1 therapy and stereotactic radiosurgery: A case report

  • Authors:
    • Xiaolin Ji
    • Luxuan Wang
    • Yanli Tan
    • Yanhong Shang
    • Ran Huo
    • Chuan Fang
    • Chunhui Li
    • Lijian Zhang
  • View Affiliations

  • Published online on: July 6, 2023     https://doi.org/10.3892/ol.2023.13947
  • Article Number: 361
  • Copyright: © Ji et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Brain metastases (BMs) usually develop in patients with non‑small cell lung cancer. In addition to systemic therapy, radiation therapy and surgery, anti‑programmed cell death‑ligand 1 (PD‑L1) therapy is another promising clinical anticancer treatment modality. However, the optimal timing and drug‑drug interactions of anti‑PD‑L1 therapy with other combined treatments remain to be elucidated. Treatment with anti‑PD‑L1 therapy is associated with an increased risk of radionecrosis (RN) regardless of tumor histology. The present study described a case of RN in a patient with lung adenocarcinoma and with BM who received anti‑PD‑L1 therapy. Before anti‑PD‑L1 treatment, the patient received whole brain radiotherapy. During durvalumab treatment, the intracranial metastases regressed. The progression of intracranial lesions 9 months later prompted a second‑line of therapy with PD‑L1 inhibitor durvalumab and stereotactic radiotherapy (SRT). Despite stereotactic irradiation, the lesions progressed further, leading to surgical resection. On examination, RN was detected, but there was no evidence of metastatic lung cancer. The aim of the present study was to present the longitudinal change in magnetic resonance imaging in RN following STR and anti‑PD‑L1 combined therapy. The atypical image of RN is conditionally important for making an accurate preoperative diagnosis.

Introduction

Brain metastases (BMs) are the most common type of tumor in the central nervous system in adults, occurring in ~20% of malignant tumors (1). BMs are most common in patients with lung cancer compared with other types of cancer and lung cancer is responsible for ~50% of all BM cases worldwide, which poses a threat to the improvement and effectiveness of oncological treatment (2). In addition to traditional methods such as chemotherapy, radiation therapy, surgery and molecularly targeted therapy that have been used in the past, emerging immunotherapeutic agents, such as checkpoint inhibitors, are also demonstrating promising therapeutic results in the treatment of lung cancer BMs (3). The development of immune checkpoint inhibitor (ICI) therapy, such as anti-programmed cell death-ligand 1 (PD-L1) therapy, has been reported to be effective in numerous types of cancer, including non-small cell lung cancer and small cell lung cancer (4). In addition, ICI therapies have been evaluated in patients receiving combination therapy, especially radiotherapy. The results obtained from clinical trials provide evidence supporting the safety and efficacy of radiotherapy in combination with anti-PD-1/PD-L1 treatment, which could be more effective than monotherapy (5). Magnetic resonance imaging (MRI) is the most commonly used modality to investigate radionecrosis (RN) (6). However, the imaging features of RN and tumor recurrence overlap considerably, with both entities demonstrating a degree of contrast enhancement and perilesional edema (7,8). Accordingly, a range of clinical and imaging strategies are being developed to evaluate tumor responses and to rule out pseudo-progression or RN. An accurate differential diagnosis is required for decision-making in the management of patients.

Case report

A 61-year-old female patient with stage IV adenocarcinoma of the lung was initially admitted to Suning County People's Hospital (Cangzhou, China) with a 6-month history of a dull headache and left upper limb weakness in December 2020. In Suning County People's Hospital, the patient ordered a service to perform next-generation sequencing (NGS) and PD-L1 immunohistochemistry (Topgen-Biopharm). The NGS was performed using an OncoDrug-Seq kit (Topgen-Biopharm) for a panel of 33 tumor-targeting genes and was performed on the NextSeq500 system (Illumina, Inc.). The lung cancer samples were fixed with 10% formalin at room temperature for 24 h and 4-µm paraffin-embedded samples were used for PD-L1 immunohistochemistry. Rabbit anti-human monoclonal antibodies to PD-L1 (1:50; cat. no. ab205921; Abcam) were used. Briefly, sections were dewaxed, dehydrated with a series of alcohol (70, 80 and 95%) at room temperature (1 h for each alcohol concentration) and the tissues were then placed in toluene for 30 min at room temperature for deparaffinization. After neutralization of endogenous peroxidase with 3% H2O2 at room temperature for 15 min and microwave antigen retrieval (800 W in 0.01 M citrate buffer pH 6), slides were preincubated with 5% bovine serum albumin blocking buffer (Thermo Fisher Scientific, Inc.) for 1 h at room temperature and then incubated overnight with monoclonal antibodies at 4°C. Subsequently, the sections were serially rinsed, incubated with Goat anti-Rabbit IgG H&L (HRP) secondary antibodies (1:200; cat. no. ab97051; Abcam) and avidin-biotinylated peroxidase complex for 1 h at 37°C, and again washed for 10 min with PBS at 37°C. Nuclear counterstaining was performed with DAPI (cat. no. C1005; Beyotime Institute of Biotechnology) at room temperature for 5 min. The immunohistochemistry images were obtained using a light microscope. The results of immunohistochemistry indicated PD-L1 positivity (Fig. 1).

Based on these results, the patient decided to undergo further clinical treatment at the Department of Oncology of the Affiliated Hospital of Hebei University (Hebei, China). An initial computed tomography scan in the Affiliated Hospital of Hebei University revealed that the patient presented with a space-occupying lesion in the superior lobe of the right lung, with multiple bilateral pulmonary nodules and with masses in the mediastinal lymph nodes and liver. Brain MRI revealed a space-occupying lesion in the frontal parietal lobe (Fig. 2A). The patient received single-agent paclitaxel therapy for 2 cycles (intravenously; 135 mg/m2, 3 weeks per cycle). On routine reexamination, MRI revealed an enlarged space-occupying lesion (Fig. 2B). The patient was then treated with direct tomotherapy (planning target volume, 36 Gy/3 Gy/12 fx). Bevacizumab (intravenously, 15 mg/kg) and paclitaxel (intravenously, 175 mg/m2)-carboplatin (intravenously, 5 mg) chemotherapy was used for 6 cycles (21 days per cycle), which demonstrated regression in BM (Fig. 2C). However, new extrapulmonary metastases in the pancreas, kidney and ovaries were detected. Based on the results of the PD-L1 immunohistochemistry, the PD-L1 inhibitor, durvalumab (intravenously, 20 mg/kg), and systemic chemotherapy (camrelizumab (intravenously, 20 mg) plus anlotinib (orally, 12 mg) and gemcitabine (intravenously, 1,000 mg/m2) for 3 cycles and duvaliumab (intravenously, 10 mg/kg) plus anlotinib (orally, 12 mg) and gemcitabine (intravenously, 1,000 mg/m2) for 9 cycles (21 days per cycle) were administered to the patient (Fig. 2D). However, the progression of BM prompted stereotactic radiotherapy (SRT) with 12 Gy radiosurgical volume (Fig. 2E). Therapy with a combination of anlotinib (orally, 12 mg) and gemcitabine (intravenously, 1,000 mg/m2) was then administered to the patient for 11 cycles (21 days per cycle). Brain MRI revealed an abnormal signal (no enhancement) and intracranial nodular enlargement (Fig. 2F). After 13 cycles of treatment with anlotinib and gemcitabine, brain MRI demonstrated an enlarged nodule with strong enhancement (Fig. 2G and H). Before SRT, magnetic resonance spectroscopy (MRS) was performed. The results suggested pseudo-progression with choline/N-acetyl-aspartate ratio (Cho/NAA) 0.92, choline/creatine ratio (Cho/Cr) 1.95 and N-acetyl-aspartate/creatine ratio (NAA/Cr) 2.12. In the contralateral normal brain tissue, the metabolite ratios of Cho/NAA Cho/Cr and NAA/Cr were 0.672, 1.08 and 1.16, respectively. Three-dimensional arterial spin labeling (3DASL) of the brain revealed low perfusion in the intracranial nodule (Fig. 3A-C). Before the surgical operation, the patients had another MRS scan and the results suggested RN with Cho/NAA 1.54, Cho/Cr 1.79 and NAA/Cr 1.16. In the contralateral normal brain tissue, the metabolite ratios of Cho/NAA Cho/Cr and NAA/Cr were 0.537, 0.904 and 1.68, respectively. The 3DASL of the brain also revealed low perfusion in the intracranial nodule (Fig. 3D-F). The patient decided to undergo surgical treatment at the Department of Neurosurgery of the Affiliated Hospital of Hebei University. The progressive BM was surgically removed and subjected to neuropathological examination. The brain tumor tissue was fixed with 10% buffered formalin at 37°C for 8–10 min. Subsequently, sections (5 µm) were cut from paraffin blocks and stained with hematoxylin and eosin at room temperature for 5 min (cat. no. C0105M; Beyotime Institute of Biotechnology), and DAPI for histopathological examination under a light microscope (Leica DM4000 M; Leica Microsystems GmbH). Histopathological analysis revealed RN with no evidence of metastatic lung cancer (Fig. 4).

Discussion

The present case demonstrated the side effects of the concurrent use of radiotherapy and anti-PD-L1 inhibitors in patients with BM. The principle of anti-programmed cell death protein 1 (PD-1)/PD-L1 therapy is to block the negative regulatory process of the PD-1/PD-L1 signaling pathway on T-cell activation and proliferation by inhibiting the complex formed by PD-1 and its ligand, PD-L1. Thus, T cells gradually recover immune activity by reactivation of the recognition and necrotic function of tumor cells (9). PD-1/PD-L1 inhibitors combined with radiotherapy mediate the antitumor effect in the dynamic interaction between effector cells and regulatory cells, such as CD8-positive T cells and tumor-infiltrating Tregs (10). In a previous study, melanoma tumors were irradiated with 10 Gy radiation; after tumor radiation, two important co-stimulatory molecules, CD86 and CD70, were revealed to be substantially upregulated on dendritic cells, which serve an important role in T-cell-mediated immune responses (11). Radiotherapy can regulate the expression of immune checkpoints, affect the expression levels of cytokines and promote the antitumor effects of immune drugs. Evidence has shown that several inflammatory cytokines, including tumor necrosis factor α, interleukin 1 and interleukin 2 can be upregulated by radiation therapy, which may be caused by an acute-phase inflammatory response (12). Conversely, radiation therapy can lead to substantial increases in the immunosuppressive cytokine transforming growth factor β in in response to cell death and stress, which have important roles in dampening radiation-induced immune responses (13). Inflammatory cytokines released from the irradiated tissue and the upregulation of checkpoint ligands can prevent autoimmune responses against healthy and malignant cells. In one study, radiation-induced upregulation of PD-L1 on the surface of tumor cells was shown to be dependent on interferon γ derived from CD8 T cells (14). In contrast, PD-1/PD-L1 inhibitors can promote radiotherapeutic effects by inhibition of negative immune-regulatory cells or molecules (1517). For example, a previous study have showed that PD-1/PD-L1 monoclonal antibody could restore T-cell activity, reduce Treg numbers and increase CD8+T/Treg ratio, thus enhancing tumor cell death (18). A case-control trial with 93 patients by Trommer et al (19) suggested that the use of PD-1 inhibitors combined with radiotherapy had benefits and could improve overall survival rates. However, with the wide application of combination therapy in clinical practice, an increasing number of studies have reported adverse reactions after the use of PD-1/PD-L1 inhibitors (2022). A previous study indicated that anti-PD-1 therapy could increase the risk of RN when combined with radiotherapy (23). The present case highlights the difficulty in differentiating between RN and pseudo-progression, following sequential treatment with PD-1/PD-L1 inhibitors and radiotherapy.

In the present case, the patient also received bevacizumab. Bevacizumab, a recombinant human monoclonal antibody, binds vascular endothelial growth factor (VEGF) and prevents VEGF from binding its receptors (VEGFR-1 and kinase insert domain receptor) on the endothelial cell surface, which serves a role in pruning blood vessels, regulating vascular permeability, reducing brain edema caused by brain necrosis and treating brain necrosis (24). In 2007, Gonzalez et al (25) first reported using bevacizumab to treat radiation brain necrosis. At present, clinical studies have proven the clinical efficacy of bevacizumab. For example, Dashti et al (26) reported that a single low-dose targeted bevacizumab infusion resulted in durable clinical and imaging improvements in 80% of patients. Another randomized double-blind study with 14 patients also supported consideration of this treatment option for patients with RN (27). At present, the majority of patients respond well to bevacizumab. However, the effect of bevacizumab on RN could not be ruled out in the present case. Jeyaretna et al (28) reported an exacerbation of cerebral RN by bevacizumab, which could lead to the hypothesis that initial treatment with bevacizumab might result in a reduction in cerebral edema. However, prolonged treatment might result in the over-pruning of at-risk blood vessels within the radiation field. The underlying mechanisms of bevacizumab-induced enlargement of RN remains unclear. At present, the duration, optimal dose and dosing interval of bevacizumab, require further evaluation.

A previous study has reported that the incidence of radiation-induced brain necrosis in patients with melanoma brain metastasis treated with SRT combined with PD-L1 immunotherapy was increased in a retrospective analysis of patients with melanoma treated with SRT (29). The study by Pires da Silva et al (30) followed-up 135 patients with melanoma that received radiotherapy combined with PD-L1 immunotherapy for an average of 23.6 months and revealed reported the probability of RN was 17% along with a cumulative incidence rate of 18% in 2 years. Furthermore, it was proposed that the time of occurrence of RN-associated symptoms was similar to the time of occurrence of radiological abnormalities (30). It has been previously reported that the rate of RN was increased with the addition of concurrent systemic therapies to SRT and whole brain radiotherapy (WBRT) (Table I) (3035). A corresponding increase in RN was not reported in patients treated with concurrent therapies and SRT alone. The present case is consistent with the findings of a previous study (30), suggesting that anti-PD-1 therapy may increase the risk of RN when combined with radiotherapy. In the present case, the patient received the PD-L1 inhibitor duvaliumab and SRT. Approximately 3 months following the combined therapy, the brain MRI indicated an abnormal signal (no enhancement), and ~7 months later, the intracranial nodule was enlarged. MRS suggested RN. The 3DASL of the brain also indicated low perfusion in the intracranial nodule. Pathological examination also indicated RN. Numerous previous studies have reported that SRT combined with immunotherapy increased the risk of radiation necrosis (31,36,37). Therefore, the concern regarding the potential risk of RN following SRT combined with PD-L1 immunotherapy has increased. Numerous previous studies have reported that PD-L1 immunotherapy combined with brain radiotherapy is effective and feasible. However, due to the potential of adverse reactions, the sequence, dosage and volume should be strictly controlled during the combined treatment, and imaging should be closely monitored to reduce the occurrence of adverse reactions such as RN. In the present case, it was demonstrated that the size of the intracranial nodules gradually decreased. After SRT, brain MRI indicated an abnormal signal (without enhancement), and that the intracranial nodule was enlarged. At this stage, it was not easy to differentially diagnose RN from tumor recurrence because of their shared clinical symptoms such as symptoms of increased intracranial pressure and/or seizures, and conventional imaging and pathological biopsy are still the best methods for differential diagnosis. However, it is difficult to perform surgery in the early clinical stage. Therefore, dynamic MRI sequence monitoring is often used to confirm the diagnosis in the clinic. In the early stages, traditional imaging of RN and tumor progression may demonstrate contrast enhancement on MRI, and large edema zones are usually observed around the lesions. In the long-term follow-up, RN indicated a decrease in tumor volume, while tumor progression indicated an increase in tumor volume (3840). In these patients, the T2-weighted image margin ‘mismatched’ the contrast-enhanced T1-weighted image margin. When the lesion appears indistinct on T2, the histology usually indicated necrosis and contrast enhancement when the contrast-enhanced rim on the T1-weighted image is associated with a distinct border on T2, and the pathology was usually a recurrent tumor (39). MRI findings of RN are often described as ‘Swiss cheese’ or ‘soap bubble’ lesions (40). At present, RN, tumor pseudo-progression and tumor recurrence have different treatment strategies. Pseudo-progression is defined as a radiographic increase in enhancement and/or edema on MRI without tumor progression. This transient increase in enhancement and/or edema exhibits spontaneous recovery, which usually occurs within a few weeks or months after the onset of pseudo-progression (41). Tumor recurrence may require surgical intervention. For RN, therapy involves corticosteroids, bevacizumab or surgical intervention (42). It is difficult to distinguish between RN, tumor pseudo-progression and tumor recurrence using conventional structural MRI at an early stage. Currently, regional cerebral blood volume and amino acid positron emission computed tomography (PET) are used to differentiate the diagnosis of these conditions. The most common imaging marker of RN in conventional MRI is the ‘Swiss cheese’ pattern with diffuse enhancements at the margins between the cortex and white matter (43). In the present case, these features were not apparent on enhanced MRI. Therefore, it was suspected that the local tumor had recurred. However, bevacizumab treatment might influence the results of MRS. Pseudo-progression usually occurs 2–5 months after radiotherapy, is self-limiting and curable. Post-radiation damage occurs after a delay of >6 months from the time of radiation and consists principally of necrosis caused by blood-brain barrier (BBB) disruption and radiation-induced demyelination leading to white matter injury (44). RN involves a space-occupying necrotic lesion with a mass effect and neurological dysfunction (45). It is not a self-limiting disease and therefore requires specialized treatment (46).

Table I.

Reported rates of radiation necrosis with anti-PD-1 therapy combined with radiation therapy.

Table I.

Reported rates of radiation necrosis with anti-PD-1 therapy combined with radiation therapy.

First author, yearHistologyCases, nSystemic treatmentRadiation necrosis rate (%)(Refs.)
Pires da Silva et al, 2019Melanoma137Anti-CTLA4, anti-PD-1 and all patients received immunotherapy combined with radiotherapy27(30)
Kim et al, 2017Melanoma135Radiotherapy and anti-PD-1 therapy17(31)
Colaco et al, 2016Melanoma, lung, breast, renal and colorectal cancer42/180 received immunotherapyAnti-CTLA4, anti-PD-1 and all patients received immunotherapy combined with radiotherapy37.5 with immunotherapy only(32)
Martin et al, 2018Melanoma and lung cancer115Immune checkpoint inhibitors therapy combined with radiotherapy20(33)
Weingarten et al, 2019Melanoma, renal-cell carcinoma, lung and breast cancer57Immunotherapy combined with radiotherapy7(34)
Andring et al, 2023Melanoma and lung cancer63Anti-CTLA4, anti-PD-1 and radiotherapy22(35)

[i] CTLA4, cytotoxic T-lymphocyte-associated protein 4; PD-1, programmed cell death protein 1.

Generally, MRS could not be used to obtain an affirmative conclusion to diagnose ‘pseudo-progression’ or ‘RN’. One of the main challenges for neurosurgeons or treating clinicians is to make a differential diagnosis of either tumor recurrence, RN or pseudo-progression in clinical settings. Even with improving neuroimaging methods or different diagnostic imaging modalities, such as diffusion-weighted imaging/diffusion tensor imaging, MRS and PET/single photon emission computed tomography, it is still challenging (47). MRS is a metabolic imaging technique that could provide value in differentiating pseudo-progression from recurrent tumors by identifying specific metabolites within the tumor that are present during active tumor growth (48). Previous studies have reported increased total choline levels in recurrent disease and reduced choline levels in tumors which exhibited pseudo-progression (49,50). Tumor recurrence has been reported to show higher Cho/Cr and Cho/NAA values compared with those of RN (51,52). In the present case, bevacizumab treatment might have influenced the results of MRS. Previous studies have reported that bevacizumab treatment could impact tumor energy and membrane metabolism, which resulted in increased intracellular pH and a decrease in the ratio of phosphatidylcholine to glycerophosphocholine or Cho/NAA values (53,54). However, in the present study, this finding was confirmed using histological examination. In the present case, there was a longitudinal change in the MRI of RN following SRT and anti-PD-L1 combined therapy. Dynamic changes in RN on enhanced MRI was demonstrated. By employing longitudinal MRI, the present case revealed atypical images of RN. These treatment-associated imaging changes were necessary for clinicians to make an accurate preoperative diagnosis in this case.

However, the exact molecular mechanism of SRT-induced RN is still unclear and has not been fully elucidated. Evidence has indicated that high-dose SRT can damage the vascular endothelium by destroying the BBB on a large scale, leading to intracranial vasogenic edema and then further to ischemia of the surrounding brain tissue (55). Furthermore, this leads to an increase in the levels of hypoxia inducible factor-1A and VEGF and finally leads to infarction and necrosis of the brain parenchyma. A previous study indicated that RN is associated with abnormalities in vascular structures, including telangiectasia, hyaline thickening of vessels and fibrinoid necrosis with intravascular thromboses (56). The expression of VEGF promotes these abnormalities in newly formed vascular structures, increases the brittleness and permeability of vascular structures, and increases edema around the lesion (57). Certain scholars have proposed that radiation damage occurs through the combination of demyelination and vascular abnormalities (35,58). In the penumbra around the necrotic nucleus, astrocytes, microglia and oligodendrocytes produce factors (e.g. VEGF) that promote cytokine release and increase the permeability of the BBB (37). However, the mechanisms by which the combination of anti-PD-L1 and radiotherapy promotes RN are still unclear. The main limitation of the present case report is that the patient received multiple different agents. As well as radiotherapy and anti-PD-1 therapy, the patient also received chemotherapy; therefore, the role of chemotherapy in the formation of RN could not be ruled out. The effect of bevacizumab on RN is uncertain. In the present case, it was hypothesized that neurological symptoms and radiologically suspected radioactive brain necrosis and tumor progression may occur after 7 months of treatment with a PD-L1 inhibitor and 2 months of treatment with SRT. The side effects of WBRT in the early stage are uncertain, and, to the best of our knowledge, there is not any literature which clearly reports that the simultaneous application of anlotinib and gemcitabine can increase the probability of radioactive brain necrosis.

In conclusion, the present study reported a case of RN following sequential PD-1/PD-L1-directed immunotherapy, WBRT and SRT. RN mimicked cancer progression with enlarged intracranial nodules. For the first time, the present study demonstrated the dynamic changes in RN on enhanced MRI.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XJ, LW, YT, YS, RH, CF, CL and LZ participated in the conception, design and data acquisition for the paper. XJ and LW participated in drafting and revising the manuscript. LZ critically revised the paper. LW ensured that questions related to the integrity of any part of the work were appropriately investigated and resolved. YT, YS, RH and CF confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Written informed consent was obtained from the patient for the publication of anonymized data and any accompanying images.

Competing interests

The authors declare that they have no competing interests.

References

1 

Achrol AS, Rennert RC, Anders C, Soffietti R, Ahluwalia MS, Nayak L, Peters S, Arvold ND, Harsh GR, Steeg PS and Chang SD: Brain metastases. Nat Rev Dis Primers. 5:52019. View Article : Google Scholar : PubMed/NCBI

2 

Rybarczyk-Kasiuchnicz A, Ramlau R and Stencel K: Treatment of brain metastases of non-small cell lung carcinoma. Int J Mol Sci. 22:5932021. View Article : Google Scholar : PubMed/NCBI

3 

Yang X, Zeng Y, Tan Q, Huang Z, Jia J and Jiang G: Efficacy of PD-1/PD-L1 inhibitors versus chemotherapy in lung cancer with brain metastases: A systematic review and meta-analysis. J Immunol Res. 2022:45188982022. View Article : Google Scholar : PubMed/NCBI

4 

Horn L, Mansfield AS, Szczęsna A, Havel L, Krzakowski M, Hochmair MJ, Huemer F, Losonczy G, Johnson ML, Nishio M, et al: First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 379:2220–2229. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Takamori S, Toyokawa G, Takada K, Shoji F, Okamoto T and Maehara Y: Combination therapy of radiotherapy and anti-PD-1/PD-L1 treatment in non-small-cell lung cancer: A mini-review. Clin Lung Cancer. 19:12–16. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Vellayappan B, Tan CL, Yong C, Khor LK, Koh WY, Yeo TT, Detsky J, Lo S and Sahgal A: Diagnosis and management of radiation necrosis in patients with brain metastases. Front Oncol. 8:3952018. View Article : Google Scholar : PubMed/NCBI

7 

Barajas RF, Chang JS, Sneed PK, Segal MR, McDermott MW and Cha S: Distinguishing recurrent intra-axial metastatic tumor from radiation necrosis following gamma knife radiosurgery using dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging. AJNR Am J Neuroradiol. 30:367–372. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Forsyth PA, Kelly PJ, Cascino TL, Scheithauer BW, Shaw EG, Dinapoli RP and Atkinson EJ: Radiation necrosis or glioma recurrence: Is computer-assisted stereotactic biopsy useful? J Neurosurg. 82:436–344. 1995. View Article : Google Scholar : PubMed/NCBI

9 

Budimir N, Thomas GD, Dolina JS and Salek-Ardakani S: Reversing T-cell exhaustion in cancer: Lessons learned from PD-1/PD-L1 immune checkpoint blockade. Cancer Immunol Res. 10:146–153. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Gong X, Li X, Jiang T, Xie H, Zhu Z, Zhou F and Zhou C: Combined radiotherapy and anti-PD-L1 antibody synergistically enhances antitumor effect in non-small cell lung cancer. J Thorac Oncol. 12:1085–1097. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Gupta A, Probst HC, Vuong V, Landshammer A, Muth S, Yagita H, Schwendener R, Pruschy M, Knuth A and van den Broek M: Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. 189:558–566. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Hong JH, Chiang CS, Campbell IL, Sun JR, Withers HR and McBride WH: Induction of acute phase gene expression by brain irradiation. Int J Radiat Oncol Biol Phys. 33:619–626. 1995. View Article : Google Scholar : PubMed/NCBI

13 

Hong JH, Chiang CS, Tsao CY, Lin PY, McBride WH and Wu CJ: Rapid induction of cytokine gene expression in the lung after single and fractionated doses of radiation. Int J Radiat Biol. 75:1421–1427. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, Stratford IJ, Poon E, Morrow M, Stewart R, et al: Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 74:5458–5468. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Luke JJ, Lemons JM, Karrison TG, Pitroda SP, Melotek JM, Zha Y, Al-Hallaq HA, Arina A, Khodarev NN, Janisch L, et al: Safety and clinical activity of pembrolizumab and multisite stereotactic body radiotherapy in patients with advanced solid tumors. J Clin Oncol. 36:1611–1618. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Theelen WSME, Peulen HMU, Lalezari F, van der Noort V, de Vries JF, Aerts JGJV, Dumoulin DW, Bahce I, Niemeijer AN, de Langen AJ, et al: Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: Results of the PEMBRO-RT phase 2 randomized clinical trial. JAMA Oncol. 5:1276–1282. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Su Z, Zhou L, Xue J and Lu Y: Integration of stereotactic radiosurgery or whole brain radiation therapy with immunotherapy for treatment of brain metastases. Chin J Cancer Res. 32:448–466. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Wen L, Tong F, Zhang R, Chen L, Huang Y and Dong X: The research progress of PD-1/PD-L1 inhibitors enhancing radiotherapy efficacy. Front Oncol. 11:7999572021. View Article : Google Scholar : PubMed/NCBI

19 

Trommer M, Adams A, Celik E, Fan J, Funken D, Herter JM, Linde P, Morgenthaler J, Wegen S, Mauch C, et al: Oncologic outcome and immune responses of radiotherapy with anti-PD-1 treatment for brain metastases regarding timing and benefiting subgroups. Cancers (Basel). 14:12402022. View Article : Google Scholar : PubMed/NCBI

20 

Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KH, de Wit M, et al: Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 377:1919–1929. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Ahn JS, Ahn YC, Kim JH, Lee CG, Cho EK, Lee KC, Chen M, Kim DW, Kim HK, Min YJ, et al: Multinational randomized phase III trial with or without consolidation chemotherapy using docetaxel and cisplatin after concurrent chemoradiation in inoperable stage III non-small-cell lung cancer: KCSG-LU05-04. J Clin Oncol. 33:2660–2666. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Wozniak AJ, Moon J, Thomas CR Jr, Kelly K, Mack PC, Gaspar LE, Raben D, Fitzgerald TJ, Pandya KJ and Gandara DR: A pilot trial of cisplatin/etoposide/radiotherapy followed by consolidation docetaxel and the combination of bevacizumab (NSC-704865) in patients with inoperable locally advanced stage III non-small-cell lung cancer: SWOG S0533. Clin Lung Cancer. 16:340–347. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Mowery YM, Patel K, Chowdhary M, Rushing CN, Roy Choudhury K, Lowe JR, Olson AC, Wisdom AJ, Salama JK, Hanks BA, et al: Retrospective analysis of safety and efficacy of anti-PD-1 therapy and radiation therapy in advanced melanoma: A bi-institutional study. Radiother Oncol. 138:114–120. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Zhuang H, Shi S, Yuan Z and Chang JY: Bevacizumab treatment for radiation brain necrosis: Mechanism, efficacy and issues. Mol Cancer. 18:212019. View Article : Google Scholar : PubMed/NCBI

25 

Gonzalez J, Kumar AJ, Conrad CA and Levin VA: Effect of bevacizumab on radiation necrosis of the brain. Int J Radiat Oncol Biol Phys. 67:323–326. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Dashti SR, Kadner RJ, Folley BS, Sheehan JP, Han DY, Kryscio RJ, Carter MB, Shields LBE, Plato BM, La Rocca RV, et al: Single low-dose targeted bevacizumab infusion in adult patients with steroid-refractory radiation necrosis of the brain: A phase II open-label prospective clinical trial. J Neurosurg. 137:1676–1686. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Levin VA, Bidaut L, Hou P, Kumar AJ, Wefel JS, Bekele BN, Grewal J, Prabhu S, Loghin M, Gilbert MR and Jackson EF: Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int J Radiat Oncol Biol Phys. 79:1487–1495. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Jeyaretna DS, Curry WT Jr, Batchelor TT, Stemmer-Rachamimov A and Plotkin SR: Exacerbation of cerebral radiation necrosis by bevacizumab. J Clin Oncol. 29:e159–e162. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Fang P, Jiang W, Allen P, Glitza I, Guha N, Hwu P, Ghia A, Phan J, Mahajan A, Tawbi H and Li J: Radiation necrosis with stereotactic radiosurgery combined with CTLA-4 blockade and PD-1 inhibition for treatment of intracranial disease in metastatic melanoma. J Neurooncol. 133:595–602. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Pires da Silva I, Glitza IC, Haydu LE, Johnpulle R, Banks PD, Grass GD, Goldinger SMA, Smith JL, Everett AS, Koelblinger P, et al: Incidence, features and management of radionecrosis in melanoma patients treated with cerebral radiotherapy and anti-PD-1 antibodies. Pigment Cell Melanoma Res. 32:553–563. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Kim JM, Miller JA, Kotecha R, Xiao R, Juloori A, Ward MC, Ahluwalia MS, Mohammadi AM, Peereboom DM, Murphy ES, et al: The risk of radiation necrosis following stereotactic radiosurgery with concurrent systemic therapies. J Neurooncol. 133:357–368. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Colaco RJ, Martin P, Kluger HM, Yu JB and Chiang VL: Does immunotherapy increase the rate of radiation necrosis after radiosurgical treatment of brain metastases? J Neurosurg. 125:17–23. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Martin AM, Cagney DN, Catalano PJ, Alexander BM, Redig AJ, Schoenfeld JD and Aizer AA: Immunotherapy and symptomatic radiation necrosis in patients with brain metastases treated with stereotactic radiation. JAMA Oncol. 4:1123–1124. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Weingarten N, Kruser TJ and Bloch O: Symptomatic radiation necrosis in brain metastasis patients treated with stereotactic radiosurgery and immunotherapy. Clin Neurol Neurosurg. 179:14–18. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Andring L, Squires B, Seymour Z, Fahim D, Jacob J, Ye H, Marvin K and Grills I: Radionecrosis (RN) in patients with brain metastases treated with stereotactic radiosurgery (SRS) and immunotherapy. Int J Neurosci. 133:186–193. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Shaw E, Scott C, Souhami L, Dinapoli R, Kline R, Loeffler J and Farnan N: Single dose radiosurgical treatment of recurrent previously irradiated primary brain tumors and brain metastases: final report of RTOG protocol 90–05. Int J Radiat Oncol Biol Phys. 47:291–298. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Vaios EJ, Winter SF, Shih HA, Dietrich J, Peters KB, Floyd SR, Kirkpatrick JP and Reitman ZJ: Novel mechanisms and future opportunities for the management of radiation necrosis in patients treated for brain metastases in the era of immunotherapy. Cancers (Basel). 15:24322023. View Article : Google Scholar : PubMed/NCBI

38 

Yaman E, Buyukberber S, Benekli M, Oner Y, Coskun U, Akmansu M, Ozturk B, Kaya AO, Uncu D and Yildiz R: Radiation induced early necrosis in patients with malignant gliomas receiving temozolomide. Clin Neurol Neurosurg. 112:662–667. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Leeman JE, Clump DA, Flickinger JC, Mintz AH, Burton SA and Heron DE: Extent of perilesional edema differentiates radionecrosis from tumor recurrence following stereotactic radiosurgery for brain metastases. Neuro Oncol. 15:1732–1738. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Smith EJ, Naik A, Shaffer A, Goel M, Krist DT, Liang E, Furey CG, Miller WK, Lawton MT, Barnett DH, et al: Differentiating radiation necrosis from tumor recurrence: A systematic review and diagnostic meta-analysis comparing imaging modalities. J Neurooncol. 162:15–23. 2023. View Article : Google Scholar : PubMed/NCBI

41 

Le Fèvre C, Lhermitte B, Ahle G, Chambrelant I, Cebula H, Antoni D, Keller A, Schott R, Thiery A, Constans JM and Noël G: Pseudoprogression versus true progression in glioblastoma patients: A multiapproach literature review: Part 1-molecular, morphological and clinical features. Crit Rev Oncol Hematol. 157:1031882021. View Article : Google Scholar : PubMed/NCBI

42 

Kano H, Kondziolka D, Lobato-Polo J, Zorro O, Flickinger JC and Lunsford LD: T1/T2 matching to differentiate tumor growth from radiation effects after stereotactic radiosurgery. Neurosurgery. 66:486–491. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Kumar AJ, Leeds NE, Fuller GN, Van Tassel P, Maor MH, Sawaya RE and Levin VA: Malignant gliomas: MR imaging spectrum of radiation therapy- and chemotherapy-induced necrosis of the brain after treatment. Radiology. 217:377–384. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Parvez K, Parvez A and Zadeh G: The diagnosis and treatment of pseudoprogression, radiation necrosis and brain tumor recurrence. Int J Mol Sci. 15:11832–11846. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Zikou A, Sioka C, Alexiou GA, Fotopoulos A, Voulgaris S and Argyropoulou MI: Radiation necrosis, pseudoprogression, pseudoresponse, and tumor recurrence: Imaging challenges for the evaluation of treated gliomas. Contrast Media Mol Imaging. 2018:68283962018. View Article : Google Scholar : PubMed/NCBI

46 

Metaweh NAK, Azab AO, El Basmy AAH, Mashhour KN and El Mahdy WM: Contrast-enhanced perfusion MR imaging to differentiate between recurrent/residual brain neoplasms and radiation necrosis. Asian Pac J Cancer Prev. 19:941–948. 2018.PubMed/NCBI

47 

Miyatake SI, Nonoguchi N, Furuse M, Yoritsune E, Miyata T, Kawabata S and Kuroiwa T: Pathophysiology, diagnosis, and treatment of radiation necrosis in the brain. Neurol Med Chir (Tokyo). 55 (Suppl 1):S50–S59. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Mayo ZS, Halima A, Broughman JR, Smile TD, Tom MC, Murphy ES, Suh JH, Lo SS, Barnett GH, Wu G, et al: Radiation necrosis or tumor progression? A review of the radiographic modalities used in the diagnosis of cerebral radiation necrosis. J Neurooncol. 161:23–31. 2023. View Article : Google Scholar : PubMed/NCBI

49 

Zeng QS, Li CF, Liu H, Zhen JH and Feng DC: Distinction between recurrent glioma and radiation injury using magnetic resonance spectroscopy in combination with diffusion-weighted imaging. Int J Radiat Oncol Biol Phys. 68:151–158. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Rock JP, Scarpace L, Hearshen D, Gutierrez J, Fisher JL, Rosenblum M and Mikkelsen T: Associations among magnetic resonance spectroscopy, apparent diffusion coefficients, and image-guided histopathology with special attention to radiation necrosis. Neurosurgery. 54:1111–1119. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Kamada K, Houkin K, Abe H, Sawamura Y and Kashiwaba T: Differentiation of cerebral radiation necrosis from tumor recurrence by proton magnetic resonance spectroscopy. Neurol Med Chir (Tokyo). 37:250–256. 1997. View Article : Google Scholar : PubMed/NCBI

52 

Shah R, Vattoth S, Jacob R, Manzil FF, O'Malley JP, Borghei P, Patel BN and Curé JK: Radiation necrosis in the brain: Imaging features and differentiation from tumor recurrence. Radiographics. 32:1343–1359. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Hattingen E, Bähr O, Rieger J, Blasel S, Steinbach J and Pilatus U: Phospholipid metabolites in recurrent glioblastoma: In vivo markers detect different tumor phenotypes before and under antiangiogenic therapy. PLoS One. 8:e564392013. View Article : Google Scholar : PubMed/NCBI

54 

Hattingen E, Jurcoane A, Bähr O, Rieger J, Magerkurth J, Anti S, Steinbach JP and Pilatus U: Bevacizumab impairs oxidative energy metabolism and shows antitumoral effects in recurrent glioblastomas: A 31P/1H MRSI and quantitative magnetic resonance imaging study. Neuro Oncol. 13:1349–1363. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Turnquist C, Harris BT and Harris CC: Radiation-induced brain injury: Current concepts and therapeutic strategies targeting neuroinflammation. Neurooncol Adv. 2:vdaa0572020.PubMed/NCBI

56 

Katsura M, Sato J, Akahane M, Furuta T, Mori H and Abe O: Recognizing radiation-induced changes in the central nervous system: Where to look and what to look for. Radiographics. 41:224–248. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Nonoguchi N, Miyatake S, Fukumoto M, Furuse M, Hiramatsu R, Kawabata S, Kuroiwa T, Tsuji M, Fukumoto M and Ono K: The distribution of vascular endothelial growth factor-producing cells in clinical radiation necrosis of the brain: Pathological consideration of their potential roles. J Neurooncol. 105:423–431. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Yoshii Y: Pathological review of late cerebral radionecrosis. Brain Tumor Pathol. 25:51–58. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2023
Volume 26 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ji X, Wang L, Tan Y, Shang Y, Huo R, Fang C, Li C and Zhang L: Radionecrosis mimicking pseudo‑progression in a patient with lung cancer and brain metastasis following the combination of anti‑PD‑1 therapy and stereotactic radiosurgery: A case report. Oncol Lett 26: 361, 2023
APA
Ji, X., Wang, L., Tan, Y., Shang, Y., Huo, R., Fang, C. ... Zhang, L. (2023). Radionecrosis mimicking pseudo‑progression in a patient with lung cancer and brain metastasis following the combination of anti‑PD‑1 therapy and stereotactic radiosurgery: A case report. Oncology Letters, 26, 361. https://doi.org/10.3892/ol.2023.13947
MLA
Ji, X., Wang, L., Tan, Y., Shang, Y., Huo, R., Fang, C., Li, C., Zhang, L."Radionecrosis mimicking pseudo‑progression in a patient with lung cancer and brain metastasis following the combination of anti‑PD‑1 therapy and stereotactic radiosurgery: A case report". Oncology Letters 26.2 (2023): 361.
Chicago
Ji, X., Wang, L., Tan, Y., Shang, Y., Huo, R., Fang, C., Li, C., Zhang, L."Radionecrosis mimicking pseudo‑progression in a patient with lung cancer and brain metastasis following the combination of anti‑PD‑1 therapy and stereotactic radiosurgery: A case report". Oncology Letters 26, no. 2 (2023): 361. https://doi.org/10.3892/ol.2023.13947