18F-NaF uptake in skull-base bone as a predictor of treatment response in advanced nasopharyngeal carcinoma (2024)

Background

Nasopharyngeal carcinoma (NPC) is the most common form of head and neck squamous cell carcinoma, particularly prevalent in Southeast Asia. Annually, it accounts for approximately 129,000 new cases and 73,000 deaths globally1. Due to its distinctive anatomical location, about 70–80% of NPC patients are diagnosed at an advanced stage. The standard treatment protocol for these advanced-stage patients involves adding induction chemotherapy to concurrent platinum-based chemoradiotherapy2. Despite this approach, around 20% of patients experience treatment failure due to resistance to chemotherapy or radiotherapy, which significantly affects survival outcomes3.

The current AJCC/UICC 8th edition TNM classification for NPC categorizes patients with skull-base bone invasion (SBBI) as T3 stage, significantly influencing treatment choices, response to therapy, and prognosis4,5. Previous studies have demonstrated that SBBI is associated with reduced bone metastasis-free survival in patients with locally advanced NPC, emphasizing its role as a risk factor for bone metastasis6. Furthermore, Li et al. found that the extent of SBBI visible on anatomic images correlated with 5-year overall survival and progression-free survival7. In a comprehensive analysis of 8,834 newly diagnosed NPC patients, Du et al. proposed reclassifying T3 NPC cases with early SBBI as T2, due to their similarly favorable prognosis with contemporary treatments8. These findings indicate that SBBI is a significant risk factor for poorer outcomes in NPC patients. However, because obtaining biopsies for SBBI is impractical, current evaluations of SBBI in NPC patients rely primarily on MRI, which reflects anatomical changes but lacks information on the metabolic alterations within the SBBI.

18F-NaF PET/CT leverages the high affinity of fluoride ions for areas of active bone remodeling9 and combines this with the advanced imaging capabilities of PET/CT. This technique allows for accurate and sensitive detection and assessment of bone metastases in patients with malignancies10. Studies have shown that 18F-NaF PET/CT achieves a patient-level sensitivity of 97.1% and specificity of 94.6%, and a lesion-level sensitivity of 99.5% and specificity of 91.5%. These results surpass the performance of 18F-NaF PET alone and planar 99mTc-MDP bone scintigraphy in detecting bone metastases in patients with NPC11. Furthermore, 18F-NaF PET/CT has demonstrated efficacy in detecting SBBI and exhibits high interobserver agreement12, establishing its value as a crucial imaging modality for the initial management of NPC13. Additionally, 18F-NaF uptake in bone disease has been identified as a predictive biomarker for response to anti-tumor therapy14,15,16. SBBI may reflect significant osteoblastic activity driven by tumor invasion and represent a more aggressive tumor phenotype that potentially exhibits greater resistance to standard chemotherapy and radiotherapy. The high 18F-NaF uptake detected at the skull base in cases of SBBI may, therefore, correlate with treatment resistance. Therefore, investigating the differences in 18F-NaF uptake at the skull base and its association with treatment response in patients with advanced NPC undergoing standard treatment is warranted.

In this study, we evaluated the differences in 18F-NaF uptake at the skull base in patients with advanced NPC and investigated whether baseline 18F-NaF uptake at the skull base is associated with response to standard treatment.

Method and materials

Patients

This retrospective study received ethical clearance and the requirement to obtain informed consent was waived by the institutional review boards of Affiliated Hospital of Guilin Medical University (ID: 2023QTLL-16). The study protocol adheres to the tenets of the 1964 Declaration of Helsinki. The study cohort comprised 142 patients diagnosed with NPC, confirmed through pathological evaluation, who underwent 18F-NaF PET/CT for staging at the Affiliated Hospital of Guilin Medical University between December 2020 and December 2023. Inclusion criteria were as follows: newly diagnosed, pathologically confirmed NPC; advanced disease stage (AJCC stage III or IV) at initial diagnosis; absence of other malignancies at presentation; pre-treatment whole-body 18F-NaF PET/CT imaging; pre-treatment MRI of the head and neck regions; standardized treatment regimen followed by comprehensive clinical and imaging follow-up data for over six months (including CT, MRI, and PET/CT); completion of induction chemotherapy (IC) followed by intensity-modulated radiation therapy (IMRT) without treatment interruption for locally advanced NPC.

Treatment protocol

For locally advanced NPC, the IC regimen comprised taxane and cisplatin: docetaxel 75mg/m2 or paclitaxel 135–175mg/m2 administered intravenously on day 1, and cisplatin or nedaplatin at 80mg/m2 intravenously on day 1, every 3weeks for 2–3 cycles. Concurrent chemoradiotherapy (CCRT) was initiated within 21 to 28days after the final IC cycle.

Radiation therapy involved IMRT with 6 MV photon irradiation. The prescribed doses were: 66–70Gy for the planning target volume derived from the primary nasopharyngeal gross tumor volume, 64–70Gy for the PTV from the nodal gross tumor volume, 60–62Gy for the PTV from the high-risk clinical target volume, and 54–56Gy for the PTV from the low-risk clinical target volume, delivered in 30–33 fractions. Concurrent chemotherapy with cisplatin or nedaplatin was administered intravenously at 80mg/m2 every 3weeks on days 1, 22, and 43 during radiotherapy. The number of chemotherapy cycles was adjusted based on the patient’s physical condition.

Evaluation of treatment response

Two radiologists (X.L and Y.J.) independently assessed tumor response by comparing pre-treatment and 1–3months post-CCRT MRI scans. Clinical responses of primary tumor at 1–3months post-CCRT were categorized according to the Response Evaluation Criteria in Solid Tumors 1.1 (RECIST 1.1) as: complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD). For analysis, patients were grouped into CR and non-CR (combining PR, SD, and PD). Any discrepancies between the radiologists’ evaluations were resolved through discussion.

18F-NaF PET/CT protocol

The 18F-NaF PET/CT scans were performed following guidelines from the Society of Nuclear Medicine and the European Association of Nuclear Medicine17. Imaging was conducted using the Ingenuity TF PET/CT scanner (Philips, Amsterdam, Netherlands). Patients received an intravenous injection of 259.0MBq (interquartile range [IQR]: 233.1–299.7MBq) of 18F-NaF and rested for 74.5min (IQR: 58.3–90.8min) before image acquisition from the head to the feet.

A low-dose CT scan for attenuation correction was acquired with the following parameters: tube voltage of 120kV, tube current of 50 mAs, slice thickness of 5.0mm, and a reconstruction slice thickness of 1.0mm. Subsequently, PET imaging was performed with a duration of 0.5min per bed position, covering 20–21 bed positions, in accordance with the manufacturer’s recommendations. Image reconstruction was carried out using a fully three-dimensional maximum likelihood ordered subsets expectation–maximization algorithm.

Interpretation of 18F-NaF PET/CT images

Two experienced nuclear medicine physicians (X.M. and Z.L.) independently evaluated the 18F-NaF PET/CT images in a randomized order for each patient, blinded to all clinical information except for the NPC diagnosis. The final diagnosis was determined by consensus, with any inconsistencies resolved through the decision of an additional experienced physician (W.F.). Visual analysis was employed for image interpretation, rather than semiquantitative analysis (e.g., standardized uptake value [SUV] cutoffs).

For the 18F-NaF PET/CT scans, areas of focally increased 18F-NaF uptake at the skull-base bone were recorded as positive for SBBI, unless a benign etiology (e.g., degenerative changes or hemangioma) was identified at the same location on the corresponding CT images. This assessment included the sphenoid body, clivus, bilateral pterygoid processes, and bilateral petrous apices.

For semiquantitative analysis, regions of interest (ROI) were placed on positive findings in the 18F-NaF PET/CT images. In patients without positive skull-base bone findings, ROIs were placed on the sphenoid body with a diameter of 1 cm2. The maximum standardized uptake values (SUVmax) and mean standardized uptake values (SUVmean) at the skull-base bone were recorded for each patient. The skull-base bone-to-background ratio (SBR) was calculated by dividing the SUVmax of the SBBI or skull-base bone by the SUVmean of the occipital bone.

Reference standard

The final diagnosis was determined based on a combination of imaging results and clinical follow-up. Due to the impracticality of obtaining biopsies for all SBBI and osseous metastases lesions, the presence of skull-base bone invasion in patients was verified using pre-treatment contrast-enhanced MRI and follow-up imaging modalities (PET/CT, MRI, or CT). In MRI imaging, the diagnosis of skull base invasion was primarily based on T1WI and contrast-enhanced T1WI. If the skull base adjacent to the tumor appeared hypointense on T1WI and showed enhancement on post-contrast T1WI, skull base invasion was considered positive. Additionally, six-month follow-up imaging was utilized to confirm skull-base invasion in NPC. Skull-base invasions that persisted or increased in size on images taken after six months confirmed the existence of bone invasion. Lesions that were indeterminate or showed no significant changes during follow-up were considered benign lesions (verified negatives) in the analysis.

Statistical analysis

Statistical analyses were performed using GraphPad Prism 10.1 (GraphPad Software Inc., La Jolla, CA, USA) by X.M. Categorical variables were presented as counts and percentages. For continuous variables, normally distributed data were reported as means ± standard deviation (SD), while skewed variables were described using medians and IQR. Differences between two groups were evaluated using the Mann–Whitney U test. For comparisons among multiple groups, the Kruskal–Wallis test followed by Dunn’s multiple comparison test was used. The receiver operating characteristic (ROC) curve was utilized to assess discriminative power, with the area under the ROC curve (AUC) calculated. Univariate and multivariable logistic regression analyses were carried out to evaluate the relationship between risk factors and treatment outcomes. All statistical tests were two-sided, with a P-value < 0.05 indicating statistical significance.

Results

Baseline clinical and PET/CT characteristics

This study included 142 patients diagnosed with advanced NPC, comprising 105 males and 37 females, with a mean age of 52.2 ± 10.7years. Upon undergoing 18F-NaF PET/CT, 18 patients were found to have bone metastases, while the remaining 124 showed no evidence of such metastases (Fig.1). Table 1 presents the baseline clinical characteristics of the patients.

The flowchart of this study.

Full size image
Full size table

18F-NaF uptake analysis based on T stage

Using the AJCC staging system, patients were classified as follows: 31 (22%) T2 stage, 75 (52%) T3 stage, and 36 (25%) T4 stage. A significant increase in 18F-NaF uptake at the skull-base was observed with advancing clinical T stage (Fig.2). T4 stage patients exhibited markedly higher SUVmax and SUVmean values in the skull-base region compared to T3 and T2 stages (median SUVmax: 13.8 vs. 9.7 vs. 4.8, p < 0.0001; median SUVmean: 8.1 vs. 5.3 vs. 3.0, p < 0.0001). Similarly, the SBR on 18F-NaF PET/CT was significantly elevated in T4 stage patients compared to T3 and T2 stages (median SBR: 8.2 vs. 5.4 vs. 2.6, p < 0.0001).

Distribution of 18F-NaF Uptake at Skull Base Across Tumor Stages in Patients with advanced NPC. (A) The SUVmax for each stage. (B) The SUVmean for each stage. (C) SBR for each stage. Each data point represents an individual patient measurement, overlaid on bars indicating the median value per stage.

Full size image

Comparison of skull-base 18F-NaF uptake in patients with and without bone metastases

Among the 18 patients with bone metastases, 15 (83.3%) showed SBBI on 18F-NaF PET/CT. All 18 patients also demonstrated positive findings for distant bone metastases. A notable difference in 18F-NaF uptake was observed between patients with and without distant bone metastases (all p < 0.05; Table 2). Patients with distant bone metastases showed higher skull-base SUVmax (17.6 [7.1, 27.9]) compared to those without (7.8 [5.3, 12.8], p = 0.01). Skull-base SUVmean was also elevated in patients with distant bone metastases (11.4 [4.0, 15.4]) compared to those without (4.4 [3.1, 7.6], p = 0.01). Using occipital bone SUVmean as background, patients with distant bone metastases exhibited a higher SBR (10.7 [4.5, 17.8]) than those without (5.1 [2.9, 7.9], p = 0.02).

Full size table

18F-NaF uptake analysis based on treatment outcome

Of the 124 patients without distant bone metastases, SBBI was detected in 77 patients. 18F-NaF PET/CT confirmed positive findings for skull-base invasion in 72 of these patients (93.5%). The diagnostic performance of 18F-NaF PET/CT for SBBI detection was as follows: sensitivity 93.5%, specificity 91.5%, accuracy 92.7%, positive predictive value 94.7%, and negative predictive value 89.6%.

Baseline 18F-NaF PET/CT parameters were compared between patients achieving CR and those with non-CR after IC followed by CRRT. Patients with non-CR showed significantly higher 18F-NaF uptake than those with CR, according to RECIST 1.1 criteria (Table 3, p < 0.001). Specifically, non-CR patients exhibited higher skull-base SUVmax (11.8 vs. 6.1, p < 0.0001) and SUVmean (6.6 vs. 3.6, p < 0.001) compared to CR patients. Using occipital bone SUVmean as background, non-CR patients demonstrated a higher SBR (6.2 [4.7, 10.8]) than CR patients (3.7 [2.6, 6.7], p = 0.0002). Figures3 and 4 show representative PET/CT images of responders and non-responders after IC followed by CRRT.

Full size table

A 57-y-old man with newly diagnosed locally advanced nasopharyngeal carcinoma who underwent 18F-NaF PET/CT and MRI at pre-treatment and MRI at post-treatment. (A) Maximum-intensity projection (MIP) and Axial images from 18F-NaF PET/CT: MIP illustrates a tracer accumulation at the base of the skull (red dashed arrow). Axial images demonstrate high 18F-NaF uptake in the lesion (dashed circle), with a SUVmax of 7.9. (B) Pre-treatment MRI: Axial contrast-enhanced T1-weighted MRI reveals an enhancing mass at the nasopharyngeal mass associated with bone invasion and soft-tissue extension. (C) Post-treatment MRI: Axial contrast-enhanced T1-weighted MRI taken after therapy shows significant reduction in the size, indicating a complete response to treatment.

Full size image

A 37-year-old man with newly diagnosed locally advanced nasopharyngeal carcinoma who underwent 18F-NaF PET/CT and MRI at pre-treatment and MRI at post-treatment. (A) Maximum-intensity projection (MIP) and Axial images from 18F-NaF PET/CT: MIP image from an 18F-NaF PET/CT scan demonstrating a hypermetabolic lesion in the skull base (red dashed arrow). Axial images demonstrate high 18F-NaF uptake in the lesion (red dashed circle), with a SUVmax of 41.1. (B) Pre-treatment MRI: Axial contrast-enhanced T1-weighted MRI reveals an enhancing mass at the nasopharyngeal mass associated with bone invasion and soft-tissue extension. (C) Post-treatment MRI: Axial contrast-enhanced T1-weighted MRI taken after therapy shows no significant reduction in the size, indicating a stable disease after treatment.

Full size image

ROC curve analysis of 18F-NaF PET/CT parameters for differentiating between CR and non-CR after IC and CRRT revealed the following: SUVmax > 10.0 showed an AUC of 0.77 (95% CI: 0.68–0.83), sensitivity 78.4%, specificity 70.0% (p < 0.0001); SUVmean > 5.2 demonstrated an AUC of 0.76 (95% CI: 0.68–0.83), sensitivity 74.3%, specificity 72.0% (p < 0.0001); and SBR > 5.1 showed an AUC of 0.70 (95% CI: 0.61–0.78), sensitivity 66.2%, specificity 74.0% (p = 0.0001) for distinguishing between CR and non-CR (Fig.5).

Univariate and multivariable analyses

We investigated the association between various clinical and imaging risk factors and treatment response in locally advanced NPC patients (Table 4). Univariate analysis identified T stage, 18F-NaF PET/CT results, SUVmax, SUVmean, and SBR as significant predictors of treatment response (p < 0.05). Due to collinearity between SUVmax and SUVmean for SBBI on 18F-NaF PET/CT, SUVmean was excluded from the multivariable analysis. In the multivariable analysis, only SUVmax remained a significant predictor (p < 0.05) with an odds ratio of 7.03 (95% CI: 1.97–25.13).

Full size table

Discussion

This study assessed the potential of 18F-NaF PET/CT for predicting treatment response in patients with advanced NPC by evaluating 18F-NaF uptake at the skull-base bone. The results indicate a significant association between higher 18F-NaF uptake and more advanced T stages, the presence of bone metastasis, and poorer treatment outcomes. ROC analysis further confirmed the discriminatory power of 18F-NaF PET/CT parameters in differentiating CR from non-CR, with SUVmax > 10 and SUVmean > 5.2 showing AUC of 0.77 and 0.76, respectively. In addition, the multivariable analysis demonstrated that SUVmax is an important risk factor for treatment response, with an odds ratio of 7.03 (95% CI: 1.97–25.13). These findings underscore the potential of 18F-NaF PET/CT as a promising predictor of treatment response in patients with advanced NPC.

The role of SBBI in the staging and prognosis of NPC has been extensively documented4. Previous studies identified a correlation between SBBI and advanced T stages, with MRI findings often serving as a reference for anatomical changes associated with SBBI18. Our study expands on these findings by demonstrating that higher T-stage NPC not only correlates with greater anatomical invasion but also with increased 18F-NaF uptake at the skull base, indicating elevated bone activity. This aligns with the known association between SBBI and poor treatment response, reflecting the substantial tumor burden and anatomical complexity involved. The observed higher 18F-NaF uptake in patients with distant bone metastasis corroborates prior reports linking SBBI-positive NPC with a higher incidence of bone metastasis6. Our results support the hypothesis that increased bone activity at the skull base is indicative of metastatic potential, potentially serving as a marker for subsequent bone metastasis. Further studies are required to determine whether elevated bone activity at the skull base will lead to metachronous bone metastasis, resulting in a worse prognosis for NPC patients19.

Patients with locally advanced NPC often receive IC followed by CCRT, which generally improves prognosis20. However, despite similar treatment regimens, approximately 20% of these patients exhibit unsatisfactory outcomes due to individual differences and tumor heterogeneity21. This leads to residual masses and poorer prognoses. The anatomical location of the skull base, near critical neural and vascular structures, further complicates treatment, often resulting in inadequate delivery and a higher risk of residual disease or recurrence22. Our findings on the predictive value of 18F-NaF PET/CT parameters for treatment outcomes resonate with prior studies that employed 18F-NaF PET/CT parameters for similar purposes14,23,24. A plausible explanation is that high 18F-NaF uptake is indicative of extensive tumor invasion and burden, which may affect the delineation of radiotherapy target areas or contribute to chemotherapy resistance, leading to non-CR to standard treatment. Further studies are necessary to confirm if patients with non-CR and high SBBI bone activity have poorer prognoses compared to those with lower bone activity. The significant correlation between 18F-NaF uptake and treatment response emphasizes the metric’s potential as a non-invasive biomarker for predicting treatment outcomes. The high discriminatory power of 18F-NaF PET/CT parameters, as evidenced by the ROC analysis, reinforces its clinical applicability in predicting outcomes and guiding therapeutic decisions. While 18F-FDG PET/CT is widely recognized for its utility in assessing metabolic activity and is particularly effective for evaluating soft tissue involvement and overall disease burden, 18F-NaF PET/CT offers unique advantages in detecting bone activity, particularly in the context of SBBI, which is often missed or less well characterized by 18F-FDG PET/CT. By incorporating 18F-NaF PET/CT findings into current protocols, clinicians can better delineate radiotherapy targets and predict chemotherapy efficacy, potentially reducing the incidence of residual disease and recurrence. Additionally, early identification of patients unlikely to respond to standard IC + CRRT can facilitate timely adjustments to treatment regimens, such as intensifying therapy or exploring alternative modalities, thereby optimizing clinical outcomes.

While our study provides compelling evidence for the utility of 18F-NaF PET/CT in predicting treatment response in advanced NPC, several limitations should be acknowledged. The retrospective nature of the study and the relatively small sample size may limit the generalizability of our findings. Prospective studies with larger cohorts are warranted to validate these results and establish standardized 18F-NaF PET/CT protocols for NPC management. A key limitation of our study is the absence of a direct comparison between 18F-NaF PET/CT and conventional imaging modalities such as MRI, CT, or 18F-FDG PET/CT, which limits our ability to fully assess the added value of 18F-NaF PET/CT over these standard anatomical techniques for detecting SBBI. Future studies that incorporate direct comparisons with these modalities are necessary to validate the potential advantages of 18F-NaF PET/CT, establish its complementary role in imaging NPC patients with SBBI, and confirm its robustness in predicting treatment response. Additionally, the mechanistic underpinnings of increased 18F-NaF uptake at the skull base in relation to NPC progression and metastasis remain unclear. Investigating the biological factors driving elevated bone activity at this site could provide deeper insights into NPC pathophysiology and inform more effective therapeutic strategies.

Conclusion

In summary, this study demonstrates the significant association between 18F-NaF uptake at the skull base and disease severity, bone metastasis in advanced NPC, and treatment response in advanced NPC. These findings highlight the potential of 18F-NaF PET/CT as a valuable tool for predicting treatment outcomes, facilitating more tailored and effective therapeutic approaches.

Data availability

The datasets used and analyzed during the present study are available from the corresponding author at the reasonable request.

Abbreviations

18F-NaF:

Fluorine-18 sodium fluoride

PET/CT:

Positron emission tomography/computed tomography

SBBI:

Skull-base bone invasion

NPC:

Nasopharyngeal carcinoma

RECIST:

Response evaluation criteria in solid tumors

CR:

Complete response

ROC:

Receiver operating characteristic

SUV:

Standardized uptake value

AUC:

Area under the curve

MRI:

Magnetic resonance imaging

IC:

Induction chemotherapy

IMRT:

Intensity-modulated radiation therapy

CCRT:

Concurrent chemoradiotherapy

PR:

Partial response

SD:

Stable disease

PD:

Progressive disease

SBR:

Skull-base bone-to-background ratio

References

  1. Bray, F. et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 74, 229–263 (2024).

    Article PubMed Google Scholar

  2. Tang, L. L. et al. The Chinese Society of Clinical Oncology (CSCO) clinical guidelines for the diagnosis and treatment of nasopharyngeal carcinoma. Cancer Commun. (Lond). 41, 1195–1227 (2021).

    Article PubMed PubMed Central Google Scholar

  3. Lee, A. W., Ma, B. B., Ng, W. T. & Chan, A. T. Management of nasopharyngeal Carcinoma: Current practice and future perspective. J. Clin. Oncol. 33, 3356–3364 (2015).

    Article PubMed Google Scholar

  4. Pan, J. J. et al. Proposal for the 8th edition of the AJCC/UICC staging system for nasopharyngeal cancer in the era of intensity-modulated radiotherapy. Cancer. 122, 546–558 (2016).

    Article PubMed Google Scholar

  5. Wu, Z. et al. Characteristics of local extension based on tumor distribution in nasopharyngeal carcinoma and proposed clinical target volume delineation. Radiother. Oncol. 183, 109595 (2023).

    Article PubMed Google Scholar

  6. Wu, B., Guo, Y., Yang, H.-H., Gao, Q.-G. & Tian, Y. Predicting bone metastasis risk based on skull base invasion in locally advanced nasopharyngeal carcinoma. Front Oncol. 12, 812358 (2022).

    Article PubMed PubMed Central Google Scholar

  7. Li, S. et al. Value of skull base invasion subclassification in nasopharyngeal carcinoma: implication for prognostic stratification and use of induction chemotherapy. Eur. Radiol. 32, 7767–7777 (2022).

    Article PubMed PubMed Central Google Scholar

  8. Du, X. J. et al. Refining the 8th edition TNM classification for EBV related nasopharyngeal carcinoma. Cancer Cell. 42, 464-473.e3 (2024).

    Article CAS PubMed Google Scholar

  9. Czernin, J., Satyamurthy, N. & Schiepers, C. Molecular mechanisms of bone 18F-NaF deposition. J. Nucl. Med. 51, 1826–1829 (2010).

    Article CAS PubMed Google Scholar

  10. Bénard, F. et al. Intra-individual comparison of 18F-sodium fluoride PET-CT and 99mTc bone scintigraphy with SPECT in patients with prostate cancer or breast cancer at high risk for skeletal metastases (MITNEC-A1): A multicentre, phase 3 trial. Lancet Oncol. 23, 1499–1507 (2022).

    Article PubMed Google Scholar

  11. Wang, D. et al. Comparison of Bone Metastases between 18F-NaF PET/CT, 18F-NaF PET, and Planar 99mTc-MDP bone scintigraphy in patients with newly diagnosed nasopharyngeal carcinoma. Contrast. Media Mol. Imaging. 2022, 1–8 (2022).

    Google Scholar

  12. Xiao, J. et al. Observer agreement and accuracy of 18F-sodium fluoride PET/computed tomography in the diagnosis of skull-base bone invasion and osseous metastases in newly diagnosed nasopharyngeal carcinoma. Nucl. Med. Commun. 41, 942–949 (2020).

    Article CAS PubMed Google Scholar

  13. Wang, D., Guo, C. & Xiao, J. The potential advantages of 18F sodium fluoride positron emission tomography-computed tomography for clinical staging and management planning in patients with nasopharyngeal carcinoma. Quant. Imaging Med. Surg. 14, 3393–3404 (2024).

    Article CAS PubMed PubMed Central Google Scholar

  14. Donners, R. et al. The value of baseline 18F-sodium fluoride and 18F-choline PET activity for identifying responders to radium-223 treatment in castration-resistant prostate cancer bone metastases. Eur. Radiol. 34, 1146–1154 (2024).

    Article CAS PubMed Google Scholar

  15. Velez, E. M., Desai, B. & Jadvar, H. Treatment response assessment of skeletal metastases in prostate cancer with 18F-NaF PET/CT. Nucl. Med. Mol. Imaging. 53, 247–252 (2019).

    Article CAS PubMed PubMed Central Google Scholar

  16. Apolo, A. B. et al. Prospective study evaluating Na18F PET/CT in predicting clinical outcomes and survival in advanced prostate cancer. J. Nucl. Med. 57, 886–892 (2016).

    Article CAS PubMed Google Scholar

  17. Beheshti, M. et al. 18F-NaF PET/CT: EANM procedure guidelines for bone imaging. Eur. J. Nucl. Med. Mol. Imaging. 42, 1767–1777 (2015).

    Article CAS PubMed Google Scholar

  18. Pan, X. X. et al. A simplified T classification based on the 8th edition of the UICC/AJCC staging system for nasopharyngeal carcinoma. Cancer Manag. Res. 11, 3163–3319 (2019).

    Article ADS PubMed PubMed Central Google Scholar

  19. Luo, D. H. et al. The map of bone metastasis in nasopharyngeal carcinoma: A real-world study. Cancer Med. 12, 17660–17670 (2023).

    Article CAS PubMed PubMed Central Google Scholar

  20. Zhang, B. et al. Association of chemoradiotherapy regimens and survival among patients with nasopharyngeal carcinoma: A systematic review and meta-analysis. JAMA Netw. Open. 2, e1913619 (2019).

    Article PubMed PubMed Central Google Scholar

  21. Zhang, L. et al. Development and validation of a magnetic resonance imaging-based model for the prediction of distant metastasis before initial treatment of nasopharyngeal carcinoma: A retrospective cohort study. EBioMedicine. 40, 327–335 (2019).

    Article PubMed PubMed Central Google Scholar

  22. Tian, Y. M. et al. Impact of primary tumor volume and location on the prognosis of patients with locally recurrent nasopharyngeal carcinoma. Chin. J. Cancer. 34, 247–253 (2015).

    Article CAS PubMed Google Scholar

  23. Letellier, A. et al. Uptake of Radium-223 dichloride and Early [(18)F]NaF PET response are driven by baseline [(18)F]NaF parameters: A pilot study in castration-resistant prostate cancer patients. Mol. Imaging Biol. 20, 482–491 (2018).

    Article CAS PubMed Google Scholar

  24. Peterson, L. M. et al. Prospective study of serial (18)F-FDG PET and (18)F-Fluoride PET to predict time to skeletal-related events, time to progression, and survival in patients with bone-dominant metastatic breast cancer. J. Nucl. Med. 59, 1823–1830 (2018).

    Article CAS PubMed PubMed Central Google Scholar

Download references

Funding

This work was supported by Joint Project on Guangxi Health and Family Commission (Grant numbers Z20210771; Z-C20220815); Guilin Science and Technology Bureau (Grant numbers 20220139-2).

Author information

Author notes

  1. Xingyu Mu and Jingze Li: contributed equally to this work.

Authors and Affiliations

  1. Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People’s Republic of China

    Xingyu Mu,Jingze Li,Jingquan Huang,Zhenzhen Wang,Zuguo Li&Wei Fu

  2. Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People’s Republic of China

    Xun Li,Yu Jiang&Zhipeng Zhou

Authors

  1. Xingyu Mu

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  2. Jingze Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  3. Jingquan Huang

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  4. Zhenzhen Wang

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  5. Zuguo Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  6. Xun Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  7. Yu Jiang

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  8. Zhipeng Zhou

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  9. Wei Fu

    View author publications

    You can also search for this author in PubMedGoogle Scholar

Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by XM, JL, JH, ZW, ZL, XL and YJ. ZZ and WF coordinated the study. The first draft of the manuscript was written by XM and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript. Written informed consent was obtained from the individual(s) for the publication of any potentially identifiable images or data included in this article.

Corresponding authors

Correspondence to Zhipeng Zhou or Wei Fu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethical approval and consent to participate

This retrospective study received ethical clearance and the requirement to obtain informed consent was waived by the institutional review boards of Affiliated Hospital of Guilin Medical University (ID: 2023QTLL-16). The study protocol adheres to the tenets of the 1964 Declaration of Helsinki.

Additional information

Publisher’s note

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

Rights and permissions

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

Reprints and permissions

About this article

18F-NaF uptake in skull-base bone as a predictor of treatment response in advanced nasopharyngeal carcinoma (6)

Cite this article

Mu, X., Li, J., Huang, J. et al. 18F-NaF uptake in skull-base bone as a predictor of treatment response in advanced nasopharyngeal carcinoma. Sci Rep 14, 29501 (2024). https://doi.org/10.1038/s41598-024-81350-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41598-024-81350-w

Keywords

  • 18F-NaF
  • PET/CT
  • Skull-base bone invasion
  • Nasopharyngeal carcinoma
  • Treatment response
18F-NaF uptake in skull-base bone as a predictor of treatment response in advanced nasopharyngeal carcinoma (2024)
Top Articles
Latest Posts
Recommended Articles
Article information

Author: Laurine Ryan

Last Updated:

Views: 5855

Rating: 4.7 / 5 (57 voted)

Reviews: 88% of readers found this page helpful

Author information

Name: Laurine Ryan

Birthday: 1994-12-23

Address: Suite 751 871 Lissette Throughway, West Kittie, NH 41603

Phone: +2366831109631

Job: Sales Producer

Hobby: Creative writing, Motor sports, Do it yourself, Skateboarding, Coffee roasting, Calligraphy, Stand-up comedy

Introduction: My name is Laurine Ryan, I am a adorable, fair, graceful, spotless, gorgeous, homely, cooperative person who loves writing and wants to share my knowledge and understanding with you.