Purpose:

Determine the safety and specificity of a tumor-targeted radiotracer (89Zr-pan) in combination with 18F-FDG PET/CT to improve diagnostic accuracy in head and neck squamous cell carcinoma (HNSCC).

Experimental Design:

Adult patients with biopsy-proven HNSCC scheduled for standard-of-care surgery were enrolled in a clinical trial and underwent systemic administration of 89Zirconium-panitumumab and panitumumab-IRDye800 followed by preoperative 89Zr-pan PET/CT and intraoperative fluorescence imaging. The sensitivity, specificity, and AUC were evaluated.

Results:

A total of fourteen patients were enrolled and completed the study. Four patients (28.5%) had areas of high 18F-FDG uptake outside the head and neck region with maximum standardized uptake values (SUVmax) greater than 2.0 that were not detected on 89Zr-pan PET/CT. These four patients with incidental findings underwent further workup and had no evidence of cancer on biopsy or clinical follow-up. Forty-eight lesions (primary tumor, LNs, incidental findings) with SUVmax ranging 2.0–23.6 were visualized on 18F-FDG PET/CT; 34 lesions on 89Zr-pan PET/CT with SUVmax ranging 0.9–10.5. The combined ability of 18F-FDG PET/CT and 89Zr-pan PET/CT to detect HNSCC in the whole body was improved with higher specificity of 96.3% [confidence interval (CI), 89.2%–100%] compared to 18F-FDG PET/CT alone with specificity of 74.1% (CI, 74.1%–90.6%). One possibly related grade 1 adverse event of prolonged QTc (460 ms) was reported but resolved in follow-up.

Conclusions:

89Zr-pan PET/CT imaging is safe and may be valuable in discriminating incidental findings identified on 18F-FDG PET/CT from true positive lesions and in localizing metastatic LNs.

Translational Relevance

Current standard of care imaging in head and neck cancer relies on nontumor-specific agents to detect potential lesions, which leads to identification of incidental lesions and subsequent overdiagnosis and unnecessary procedures. In this early-phase clinical study, we studied a tumor-targeted PET radiopharmaceutical – zirconium-89 panitumumab (89Zr-pan) – in 14 patients with head and neck squamous cell carcinoma. We found that the use of 89Zr-pan PET/CT led to improved specificity and positive predictive value in detection of metastatic head and neck squamous cell carcinoma lesions compared with fluorine-18 fluorodeoxyglucose (18F-FDG) PET/CT alone. Tumor-targeted imaging using 89Zr-panitumumab PET/CT following 18F-FDG PET/CT has the potential to significantly improve preoperative cancer staging in patients with head and neck squamous cell carcinoma.

Fluorine-18 fluorodeoxyglucose PET combined with CT (18F-FDG PET/CT) is one of the most common imaging modalities for staging and surveillance of cancer. 18F-FDG is taken up by cells with high glucose metabolism, and as such, increased uptake is visualized in aggressive tumors as well as in normal physiologic states (e.g., brain cortex, genitourinary tract, bone marrow, palatine tonsils) and other states of increased metabolic activity (e.g., post-surgical changes, inflammation, and infection).

Although 18F-FDG PET/CT has high sensitivity and specificity for detection of cancer lesions for TNM (tumor, lymph node, metastasis) staging in head and neck cancer (sensitivity - T: 97%–100%, N: 82%–87%, M: 53%–100%; specificity - T: 94%, N: 79%–86%, M: 93%–98%; refs. 1–3), the prevalence of incidental findings in head and neck cancer continues to be high due to lack of tumor-specificity with 18F-FDG, especially in regional lymph nodes. Incidental lesions are identified by 18F-FDG PET/CT in 35–38% of patients with head and neck cancer, 12%–20% of whom undergo invasive procedures (e.g., biopsy and/or endoscopy) while the true positive rate for distant metastatic disease is only 5%–9% (4, 5). This results in a significant number of unnecessary invasive procedures, which may delay treatment initiation or may result in unnecessary treatment (6).

To improve whole body imaging in head and neck cancer, tumor-targeted radiotracers such as CD44v6 (a cell surface molecule; ref. 7), angiogenesis-related integrins (8), somatostatin receptors (9), and hypoxia markers (10) have been tested. Yet, heterogeneous expression of receptors, limited application to tumors with specific receptors, and immunogenicity impede their adoption. Head and neck squamous cell carcinoma (HNSCC) virtually universally overexpress epidermal growth factor receptor (EGFR). EGFR overexpression has been used as a target for tyrosine kinase inhibitors and monoclonal antibodies with modest anti-cancer effect (11–16). Moreover, using optical agents (e.g., fluorescently conjugated monoclonal antibodies) targeting EGFR in the intraoperative setting demonstrated its value for real-time tumor detection (17–21). With this successful targeting of EGFR in the intraoperative setting, we wanted to explore the value of a radiolabeled antibody for its ability to preoperatively identify tumor and potentially create a roadmap to further improve intraoperative identification of regional metastatic disease. To determine the potential of EGFR targeted whole body imaging for preoperative evaluation, we evaluated zirconium-89 panitumumab (89Zr-pan) to detect HNSCC lesions in patients with head and neck cancer.

Clinical trial design

A single center, open label, non-randomized early phase clinical study was performed to evaluate panitumumab-IRDye800 (pan800) and 89Zr-pan for dual-modality imaging in patients with HNSCC. 89Zr-pan (IND 135573) and pan800 (IND 119474) were approved as investigational drugs by the FDA, and the study was approved by the Institutional Review Board (IRB-41878) and performed in accordance with the Declaration of Helsinki, ICH-GCP guidelines, and the United States Common Rule.

An overview of the trial workflow is presented in Fig. 1. A total of 14 adult patients with recurrent or new biopsy-proven HNSCC with any T stage and any subsite and 18F-FDG PET/CT performed as standard of care who were scheduled to undergo neck dissection were enrolled sequentially after obtaining written informed consent at Stanford Hospital and Clinics between 4/2019 and 12/2020. Patients with previous bilateral neck dissection, pregnant or breastfeeding, severe renal/heart/lung disease were excluded (detailed inclusion and exclusion criteria available upon request). Patients were infused with a 30 mg (n = 9) or 50 mg (n = 5) flat dose of pan800, followed by 1 mCi (37 MBq) of 89Zr-pan 1–5 days prior to surgery. The molecular weight of panitumumab is 147 kDa, IRdye800 1.1 kDa. Trace amounts (<1 mg) of panitumumab were conjugated to 89Zirconium.

Figure 1.

Overview of imaging workflow. Enrolled patients received a systemic infusion of 89Zr-pan followed by pan800 and underwent PET/CT imaging prior to surgery. During surgery, open- and closed-field near-infrared fluorescence imaging of the primary tumor and cervical lymph nodes were performed. After surgery, closed-field fluorescence imaging and macroscopic imaging were performed on the formalin-fixed, paraffin-embedded tissue, and microscopic imaging of routine H&E and IHC stainings were performed. 89Zr-pan, 89Zirconium-panitumumab; pan800, panitumumab-IRDye800; H&E, hematoxylin & eosin.

Figure 1.

Overview of imaging workflow. Enrolled patients received a systemic infusion of 89Zr-pan followed by pan800 and underwent PET/CT imaging prior to surgery. During surgery, open- and closed-field near-infrared fluorescence imaging of the primary tumor and cervical lymph nodes were performed. After surgery, closed-field fluorescence imaging and macroscopic imaging were performed on the formalin-fixed, paraffin-embedded tissue, and microscopic imaging of routine H&E and IHC stainings were performed. 89Zr-pan, 89Zirconium-panitumumab; pan800, panitumumab-IRDye800; H&E, hematoxylin & eosin.

Close modal

All patients underwent one PET/CT scan using a SiPM-based camera with 3D and time of flight acquisition (Discovery MI, GE Healthcare, Chicago, IL, USA) 1–5 days after administration of 89Zr-pan. Five patients were enrolled in the dosimetry sub-study and underwent a total of three PET/CT scans at 1–2 hours, 24–48 hours, and 72–96 hours after administration of the study drug. Whole body images were acquired from vertex to mid-thigh, and data were reconstructed with an ordered subset expectation maximization (OSEM) algorithm using 2 iterations and 17 subsets and were corrected for decay, attenuation, scattering, dead time, and randoms. A low-dose whole-body CT scan was acquired once for attenuation correction and organ delineation.

The 89Zr-pan PET/CT and the 18F-FDG PET/CT scans were reviewed in a blinded manner by two board-certified nuclear medicine physicians (AI, HD) prior to patients undergoing their standard of care surgical resection.

Quantification and comparison of the 18F-FDG PET/CT and 89Zr-pan PET/CT uptake

The 18F-FDG PET/CT scans were evaluated first, followed by 89Zr-pan PET/CT scans. Regions of interest (ROI), such as primary tumor, lymph nodes (LNs), and other organs were outlined, and the maximum standardized uptake values (SUVmax) (22) were quantified (HD) to evaluate exploratory thresholds.

Quantification of fluorescence imaging and correlation with preoperative PET/CT scans

Intraoperative near-infrared fluorescence imaging was performed as described by Nishio and colleagues (19) LNs identified on preoperative 89Zr-pan PET/CT were correlated with the intraoperatively identified specimens based on size and location in the neck levels. Resected tissue specimens underwent routine histopathologic processing followed by EGFR immunohistochemistry for target validation as previously described (19).

Dosimetry analysis

Absorbed doses (ADs) were calculated using OLINDA/EXM software (version 1.1, Vanderbilt University, Memphis, TN) which bases the dose calculations on the MIRD model (23) in a similar manner as described by Lindenberg and colleagues (24).

Statistical analysis

Descriptive statistics are presented as means and standard deviations (SD) for normally distributed continuous characteristics, including tumor size, time from drug infusion to imaging; median and ranges for non-normally distributed continuous characteristics, including mean signal intensities (MSI), such as mean fluorescence intensities (MFI) and SUVmax, and signal-to-background ratios (SBR). Standard boxplots (median as center line, the first and third interquartile range (IQR) as hinges, the highest and lowest values within the 1.5*IQR as hinges, outlier as circle, and mean as diamond) were used to graphically represent the difference in MSIs among the different imaging modalities.

For the non-normally distributed data, the Kruskal–Wallis test was used to compare the MSIs and SBRs within each type of image (18F-FDG PET/CT, 89Zr-pan PET/CT, pan800 imaging), and post-hoc Wilcoxon rank sum test was used to evaluate pairwise comparisons. For normally distributed data, the one-way ANOVA was used to compare mean MSIs and SBRs after evaluation of key assumptions, including extreme outliers, normality, and homogeneity of variances. To compare the SUVmax between the two PET/CT scans, Wilcoxon signed rank test was performed to evaluate the paired, non-normally distributed data. Sensitivity and specificity in detecting HNSCC among the different imaging modalities were calculated using the receiver operating characteristic (ROC) curves on a per-lesion basis. The optimal cutoff values for MSI and SBR were calculated using the Youden index, which is (sensitivity + specificity) – 1. Logistic regression was performed using 18F-FDG SUVmax and 89Zr-pan SUVmax to predict the binary response variable of histopathology – whether cancer was present or not. For all analyses, a two-tailed P value of less than 0.05 was deemed statistically significant (*, P < 0.05; **, P < 0.01; ***, P < 0.001). The relationship between SUVmax and MFI was assessed using the Pearson correlation coefficient, the coefficient of determination (R2) value, and the Spearman's correlation coefficient. Repeated measures ANOVA was performed to compare mean SUVmax across different scan times of the 89Zr-pan PET/CT after confirming the following assumptions of no significant outliers, normal distribution using the Shapiro-Wilk test and plotting QQ plots, and homogeneity of the variances using Mauchly's test. Missing SUVmax data was limited (5.1% and 7.7% for 18F-FDG PET/CT and 89Zr-pan PET/CT, respectively) and was omitted for analysis.

Powe considerations were limited given this was an early Phase 1 study. However, given our expectation of 420 specimens (14 subjects and average 30 LNs per subject), we calculated the sample size based on prevalence of 7.5%, significance level of 0.05, 0.3 proportion of discordant pairs, 95% power to detect 0.1 difference in specificities, and greater than 71% power to detect 0.25 difference in sensitivity in detecting metastatic lymph nodes. However, we evaluated only those lesions that were detected on either 18F-FDG PET/CT or 89Zr-pan PET/CT with their corresponding histopathology to compare the diagnostic performance of the two radiotracers. Given the potential for underpowered analysis, interpretation based on 95% confidence intervals was recommended.

Data availability

The study protocol and the data generated in this study are available upon request from the corresponding author.

Patient characteristics

There were 14 patients enrolled and 17 neck dissection specimens since 3 patients underwent bilateral neck dissection (Table 1, Supplementary Fig. S1). The clinical N stage was evenly distributed between N0 (no nodal disease) and N+ (nodal disease). One patient underwent neoadjuvant immunochemotherapy with good clinical response prior to surgical resection with pathologic staging showing no evidence of residual cancer. All patients received both 89Zr-pan and pan800. The median time between 18F-FDG PET/CT and 89Zr-pan PET/CT was 23 days (IQR 14.8 days), and there were no clinical interventions between the two imaging studies. All patients received 1.00 ± 0.12 mCi (mean ± SD) of 89Zr-pan followed by 30 mg or 50 mg of pan800. The mean ± SD time interval from 89Zr-pan administration to 89Zr-pan PET/CT was 73.5 ± 31.6 hours, and the time interval from pan800 infusion to the first fluorescent intraoperative image was 96.1 ± 37.0 hours. Overall, a total of 37 lesions were evaluated on both 18F-FDG PET/CT and 89Zr-pan PET/CT scans and histopathology; 11/12 primary tumors were positive for disease and 10/25 regional LNs were positive for disease on histopathology. In addition, 11 incidental lesions outside the head and neck region were included for analysis, but pathology was only available for one lesion. One grade 1 adverse event of prolonged QTc (460 ms) was observed and deemed possibly related to the study on day of infusion, but this resolved in follow-up. There were no grade 2 or 3 events that were related to the study drug (Supplementary Table S1).

Table 1.

Patient demographics.

 Age (years)/SexTumor siteNeck dissection lateralityClinical StagePathologic StageMaximum Tumor Diameter (cm)Dose 89Zr-pan (mCi)Time from 89Zr-pan to PET/CT (hrs)aDose pan800 (mg)Time from pan800 infusion to FL image (hrs)b
Patient 1 56 M Oral cavity Right cT2N0 pT2N0 2.7 1.00 27.4 50 48.1 
Patient 2 69 F Cutaneous Right cTxN+ pTxN3b NA 0.90 28.5 50 50 
Patient 3 83 M Oral cavity Left cT4N0 pT4aN0 1.13 28.4 50 48.7 
Patient 4 73 M Oral cavity Right cT3N0 pT2N0 2.5 0.74 95.2 50 117.5 
Patient 5 71 M Oral cavity Right cT4N+ pT4aN2b 0.96 101.2 50 123.9 
Patient 6 68 F Oral cavity Right cT3N+ pT4aN0 0.9 0.98 87.3 30 90.7 
Patient 7 62 F Oral cavity Right cT1N0 pT1N0 5.5 1.01 92.7 30 144.5 
Patient 8 63 F Oral cavity Left cT4N0 cT4N0 0.81 95 30 121.1 
Patient 9 54 M Oral cavity Bilateral cT4aN2b No cancer detected NA 1.08 91.1 30 119.9 
Patient 10 79 M Oral cavity Bilateral cT3N0 pT3N0 3.2 1.10 20.4 30 27.2 
Patient 11 64 M Oral cavity Left cT2N2 pT2N3b 2.2 1.01 95.9 30 117.4 
Patient 12 63 M Oropharynx Right cT2N0 pT3N0 4.5 1.06 94 30 120.9 
Patient 13 72 M Cutaneous Left cT3N2 pT3N3b 1.12 76.4 30 97 
Patient 14 67 M Hypopharynx Bilateral cT2N1 pT2N1 3.5 1.11 95.4 30 118.9 
Mean ± SD 67.4 ± 8.0     4.3 ± 2.1 1.00 ± 0.12 73.5 ± 31.6 37 ± 9.9 96.1 ± 37.0 
 Age (years)/SexTumor siteNeck dissection lateralityClinical StagePathologic StageMaximum Tumor Diameter (cm)Dose 89Zr-pan (mCi)Time from 89Zr-pan to PET/CT (hrs)aDose pan800 (mg)Time from pan800 infusion to FL image (hrs)b
Patient 1 56 M Oral cavity Right cT2N0 pT2N0 2.7 1.00 27.4 50 48.1 
Patient 2 69 F Cutaneous Right cTxN+ pTxN3b NA 0.90 28.5 50 50 
Patient 3 83 M Oral cavity Left cT4N0 pT4aN0 1.13 28.4 50 48.7 
Patient 4 73 M Oral cavity Right cT3N0 pT2N0 2.5 0.74 95.2 50 117.5 
Patient 5 71 M Oral cavity Right cT4N+ pT4aN2b 0.96 101.2 50 123.9 
Patient 6 68 F Oral cavity Right cT3N+ pT4aN0 0.9 0.98 87.3 30 90.7 
Patient 7 62 F Oral cavity Right cT1N0 pT1N0 5.5 1.01 92.7 30 144.5 
Patient 8 63 F Oral cavity Left cT4N0 cT4N0 0.81 95 30 121.1 
Patient 9 54 M Oral cavity Bilateral cT4aN2b No cancer detected NA 1.08 91.1 30 119.9 
Patient 10 79 M Oral cavity Bilateral cT3N0 pT3N0 3.2 1.10 20.4 30 27.2 
Patient 11 64 M Oral cavity Left cT2N2 pT2N3b 2.2 1.01 95.9 30 117.4 
Patient 12 63 M Oropharynx Right cT2N0 pT3N0 4.5 1.06 94 30 120.9 
Patient 13 72 M Cutaneous Left cT3N2 pT3N3b 1.12 76.4 30 97 
Patient 14 67 M Hypopharynx Bilateral cT2N1 pT2N1 3.5 1.11 95.4 30 118.9 
Mean ± SD 67.4 ± 8.0     4.3 ± 2.1 1.00 ± 0.12 73.5 ± 31.6 37 ± 9.9 96.1 ± 37.0 

Abbreviations: FL, fluorescence (FL); pan800, panitumumab-IRDye800; 89Zr-pan, 89Zirconium-panitumumab.

aTime from infusion of panitumumab-IRDye800 to the first intraoperative fluorescence image in hours.

bTime from infusion of 89Zr-panitumumab to 89Zr-PET image in hours. Includes the third PET scan of subjects in the dosimetry cohort.

Isotope and optical labeling of panitumumab demonstrated similar results

All lesions that were detected on 18F-FDG PET/CT and confirmed to be HNSCC on histopathology were identified on 89Zr-pan PET/CT (Fig. 2). 18F-FDG PET/CT had higher median SUVmax compared to 89Zr-pan PET/CT for primary tumor (11.6 vs. 3.4; P < 0.001; Wilcoxon sign rank), LN with cancer (11.6 vs. 2.4; P = 0.009; Wilcoxon sign rank), and LN without cancer (4.7 vs. 2.4; P < 0.001; Wilcoxon sign rank) with the following ranges for primary tumor, LN with cancer, LN without cancer (18F-FDG: 6.5–22.6, 7.5–23.6, 2.0–21.0; 89Zr-pan: 0.94–10.5, 1.06–9.8, 1.4–2.4), respectively (Fig. 3B). Consistent with previously published studies using optically labeled panitumumab (19, 20, 25), both MFI and SBR in this study were able to distinguish benign tissue from tumor tissue with high sensitivity and specificity [MFI: 85.2% (95% confidence interval (CI), 72%–99%], 80.2% (95% CI, 74%–86%); SBR: 96.3% (95% CI, 89.2%–100%), 91.0% (95% CI, 87% – 95%), respectively) with cutoff values of 0.016 for MFI and 2.9 for SBR. SUVmax of the 89Zr-pan PET/CT and MFI of the fluorescence imaging were more highly correlated (Pearson = 0.82; R2 = 0.67; Spearman = 0.66) than the SUVmax of 18F-FDG PET/CT with MFI (Pearson = 0.18; R2 = 0.03; Spearman = 0.09) in detecting tumor lesions (Fig. 3C).

Figure 2.

89Zr-pan PET/CT detects HNSCC. 18F-FDG PET/CT and 89Zr-pan PET/CT images of the primary tumor (blue outline) of the tongue in Subject 11 (A) and hard palate in Subject 6 (B). A centrally necrotic LN (orange outline) in level II in Subject 11 (C) is visualized with higher SUVmax uptake in the periphery of the LNs on both scans. An additional LN (orange outlines) was detected on 89Zr-pan PET/CT in a patient with primary tumor in the hypopharynx (blue outline) in Subject 14 (D). LN, lymph node; SCC, squamous cell carcinoma; 18F-FDG PET/CT, 18Fluorodeoxyglucose-positron emission tomography/computed tomography; 89Zr-pan PET/CT, 89Zirconium-positron emission tomography/computed tomography.

Figure 2.

89Zr-pan PET/CT detects HNSCC. 18F-FDG PET/CT and 89Zr-pan PET/CT images of the primary tumor (blue outline) of the tongue in Subject 11 (A) and hard palate in Subject 6 (B). A centrally necrotic LN (orange outline) in level II in Subject 11 (C) is visualized with higher SUVmax uptake in the periphery of the LNs on both scans. An additional LN (orange outlines) was detected on 89Zr-pan PET/CT in a patient with primary tumor in the hypopharynx (blue outline) in Subject 14 (D). LN, lymph node; SCC, squamous cell carcinoma; 18F-FDG PET/CT, 18Fluorodeoxyglucose-positron emission tomography/computed tomography; 89Zr-pan PET/CT, 89Zirconium-positron emission tomography/computed tomography.

Close modal
Figure 3.

Tumor-specific preoperative 89Zr-pan PET/CT and intraoperative fluorescence imaging detects HNSCC. Examples of preoperative and intraoperative imaging with corresponding histopathology, including a primary tumor of the hard palate in Subject 6 (A, top row), metastatic LNs in Subject 14 (A, middle row), and no HNSCC in Subject 9 (A, bottom row) are shown. Distribution of SUVmax values for 18F-FDG PET/CT and 89Zr-pan PET/CT (B) and correlation between MFI and SUVmax (C) are shown. HNSCC, head and neck squamous cell carcinoma; SUVmax, maximum standardized uptake value; MFI, mean fluorescence intensity.

Figure 3.

Tumor-specific preoperative 89Zr-pan PET/CT and intraoperative fluorescence imaging detects HNSCC. Examples of preoperative and intraoperative imaging with corresponding histopathology, including a primary tumor of the hard palate in Subject 6 (A, top row), metastatic LNs in Subject 14 (A, middle row), and no HNSCC in Subject 9 (A, bottom row) are shown. Distribution of SUVmax values for 18F-FDG PET/CT and 89Zr-pan PET/CT (B) and correlation between MFI and SUVmax (C) are shown. HNSCC, head and neck squamous cell carcinoma; SUVmax, maximum standardized uptake value; MFI, mean fluorescence intensity.

Close modal

89Zr-pan PET/CT is highly specific for HNSCC compared to 18F-FDG PET/CT

Using the SUVmax and histopathology as the reference standard, ROC curves were evaluated for the SUVmax of the two scans for only the lesions that were evaluated on histopathology. 89Zr-pan PET/CT had higher AUC (0.82, 95% CI, 0.68–0.95) compared to 18F-FDG PET/CT (0.76, 95% CI, 0.59–0.93; Table 2). Based on the optimal SUVmax cutoff of 2.61 and 9.27 for 89Zr-pan PET/CT and 18F-FDG PET/CT using Youden's Index, respectively, 89Zr-pan PET/CT had 100% specificity compared to 81.2% (95% CI, 62.1–100%) for 18F-FDG PET/CT (Table 2). However, 89Zr-pan PET/CT had a sensitivity of 59.1% (95% CI, 38.5–79.6%) compared to 77.3% (95% CI, 59.8–94.8%) for 18F-FDG PET/CT. This suggests that the imaging modalities may have improved accuracy when performed together in the preoperative staging [sensitivity 60.9% (95% CI, 40.9–80.8%), specificity 100% (95% CI, 100–100%)].

Table 2.

Diagnostic statistics for all tissues with SUVmax signal evaluated on histopathology.

FDG-PETZr-PETCombined FDG/Zr-PETa
SUVmaxSUVmax
# LNs (total # tissue)b 24 (37) 21 (33) 24 (37) 
Sensitivity (%)c 77.3 (59.8–94.8) 59.1 (38.5–79.6) 60.9 (40.9–80.8) 
Specificity (%)c 81.2 (62.1–100) 100 (100–100) 100 (100–100) 
Positive predictive value (%)c 85.0 (69.4–100) 100 (100–100) 100 (100–100) 
Negative predictive value (%)c 72.2 (51.5–92.9) 64.0 (45.2–82.8) 64.9 (43.1–81.9) 
AUCc 0.76 (58.6–93.1) 0.82 (68.2–95.2) 0.83 (70.1–95.7) 
Optimal thresholdd 9.27 2.61 
FDG-PETZr-PETCombined FDG/Zr-PETa
SUVmaxSUVmax
# LNs (total # tissue)b 24 (37) 21 (33) 24 (37) 
Sensitivity (%)c 77.3 (59.8–94.8) 59.1 (38.5–79.6) 60.9 (40.9–80.8) 
Specificity (%)c 81.2 (62.1–100) 100 (100–100) 100 (100–100) 
Positive predictive value (%)c 85.0 (69.4–100) 100 (100–100) 100 (100–100) 
Negative predictive value (%)c 72.2 (51.5–92.9) 64.0 (45.2–82.8) 64.9 (43.1–81.9) 
AUCc 0.76 (58.6–93.1) 0.82 (68.2–95.2) 0.83 (70.1–95.7) 
Optimal thresholdd 9.27 2.61 

Abbreviations: MFI, mean fluorescence intensity; MSI, mean signal intensity; SBR, signal to background ratio; SUVmax, maximum standard uptake value; AUC, area under the curve.

aPredicted diagnostic statistics from logistic regression with combined FDG-PET and Zr-PET imaging.

bAll tissues with SUVmax signal or MFI signal, including primary tumor and LNs, were included for analysis.

c95% confidence interval in parentheses.

dOptimal threshold calculated using Youden's Index.

89Zr-pan PET/CT is highly specific for HNSCC on whole body imaging

Evaluation of whole-body PET/CT scans further reinforced the high specificity of 89Zr-pan PET/CT in preoperative detection and staging of HNSCC as shown in Fig. 4AE. Patient 2 presented with bilateral cutaneous SCC of the scalp and regional cervical adenopathy and underwent 18F-FDG PET/CT and 89Zr-pan PET/CT prior to surgery. On 18F-FDG PET/CT, the patient was noted to have numerous mediastinal LNs (Fig. 4A; green outline) and a left inguinal node (Fig. 4B; green outline), which were not visualized on 89Zr-pan PET/CT. The mediastinal nodes (biopsy) and other sites (18-month follow-up) were negative for SCC. Another patient had high SUVmax signals in a right apical pulmonary lesion and around a shoulder prosthesis on 18F-FDG PET/CT, (Fig. 4C and D; green outline), neither of which were visualized on 89Zr-pan PET/CT. In the bottom row, a patient had good clinical response with neoadjuvant immunochemotherapy (Fig. 4E). 18F-FDG PET/CT demonstrated moderate signal at the primary tumor site but resolution of the cervical LNs. However, 89Zr-pan PET/CT performed just prior to surgery was negative for any SUVmax signal, and there was no histopathological evidence of cancer in any of the specimens.

Figure 4.

Combined 18F-FDG PET/CT and 89Zr-pan PET/CT imaging improves preoperative detection of HNSCC. The following cases illustrate the high specificity of 89Zr-pan as a radiotracer for preoperative detection of HNSCC. Mediastinal LNs (A) and inguinal LN (B) in Subject 2, pulmonary nodule (C) and inflammation around a prosthetic joint (D) in Subject 14, and primary tongue SCC after systemic therapy (E) in Subject 9, which were negative for HNSCC (green outlines), were only visualized on 18F-FDG PET/CT and not on 89Zr-pan PET/CT. ROC curves were used to evaluate detection of HNSCC in the whole body (including all incidental lesions with 18F-FDG uptake) using 18F-FDG PET/CT (F), 89Zr-pan PET/CT (G), and combined PET/CTs (H). *, Final pathology of primary tumor site showed no evidence of SCC.

Figure 4.

Combined 18F-FDG PET/CT and 89Zr-pan PET/CT imaging improves preoperative detection of HNSCC. The following cases illustrate the high specificity of 89Zr-pan as a radiotracer for preoperative detection of HNSCC. Mediastinal LNs (A) and inguinal LN (B) in Subject 2, pulmonary nodule (C) and inflammation around a prosthetic joint (D) in Subject 14, and primary tongue SCC after systemic therapy (E) in Subject 9, which were negative for HNSCC (green outlines), were only visualized on 18F-FDG PET/CT and not on 89Zr-pan PET/CT. ROC curves were used to evaluate detection of HNSCC in the whole body (including all incidental lesions with 18F-FDG uptake) using 18F-FDG PET/CT (F), 89Zr-pan PET/CT (G), and combined PET/CTs (H). *, Final pathology of primary tumor site showed no evidence of SCC.

Close modal

Combined 18F-FDG PET/CT and 89Zr-pan PET/CT improves detection of HNSCC

ROC curves were evaluated using the SUVmax for tumor and incidental lesions for 18F-FDG PET/CT (Fig. 4F), 89Zr-pan PET/CT (Fig. 4G), and the combination of both scans (Fig. 4H). Forty-eight lesions (primary tumor, LNs, incidental findings) with median SUVmax 8.52 (range 2.0–23.6) were visualized on 18F-FDG PET/CT and 34 lesions with median SUVmax 2.0 (range 0.9–10.5) were identified with 89Zr-pan PET/CT (detailed flowchart of identified lesions available in Supplementary Fig. S1). The combined 18F-FDG PET/CT and 89Zr-pan PET/CT scans detected cancer lesions with a specificity of 96.3% (95% CI, 89.2%–100%) and AUC of 0.91 (95% CI, 0.84–0.99) compared with 18F-FDG PET/CT alone with specificity 74.1% (95% CI, 57.5%–90.6%) and AUC 0.79 (95% CI, 0.65–0.93).

89Zr-pan PET/CT has improved specificity for the detection of metastatic lymph nodes

There was no statistically significant difference in SUVmax between the LNs with and without cancer on pathologic assessment for 18F-FDG PET/CT (P = 0.064, one-way ANOVA). Among the 25 LNs identified as having high SUVmax signal on either PET/CT scan, 4 LNs were noted to have only 18F-FDG uptake, 20 LNs had both 18F-FDG and 89Zr uptake, and 1 LN had only 89Zr uptake, which was noted to be positive for disease on review of pathology (Fig. 5A). There was a significant difference in SUVmax between LNs with and without cancer on the 89Zr-pan PET/CT scan (P = 0.029; one-way ANOVA), which correlated with a significant difference in MFI and SBR between LNs with and without cancer on fluorescence imaging (P < 0.001; Kruskal–Wallis; Fig. 5B). The specificity was higher for 89Zr-pan PET/CT (86.7%; 95% CI, 69.5% – 100%) compared to 18F-FDG PET/CT (66.7%; 95% CI, 42.8% – 90.5%) (Fig. 5C). The combined 18F-FDG PET/CT and 89Zr-pan PET/CT scans detected lymph node lesions with a specificity of 80% (95% CI, 59.8%–100%) and AUC of 0.81 (95% CI, 63.9–98.7) compared to 18F-FDG PET/CT alone [see above for specificity; AUC 0.73 (95% CI, 50.6–96.1)].

Figure 5.

89Zr-pan PET/CT has higher specificity for distinguishing metastatic LNs compared to 18F-FDG PET/CT. All LNs identified on 18F-FDG PET/CT and 89Zr-pan PET/CT scans were mapped onto individual subject's necks with corresponding pathology results (A). The range of SUVmax for LNs detected on18F-FDG PET/CT and 89Zr-pan PET/CT and the range of MFI and SBR for LNs were evaluated using standard boxplots with median as center line and mean as the diamond. P-values were determined using the Kruskal–Wallis test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; B). ROC curves were evaluated for LNs detected on 18F-FDG PET/CT and 89Zr-pan PET/CT (C). The ROC curves for the combined PET/CTs had higher AUC and specificity compared with 18F-FDG PET/CT alone (95% CIs available in the manuscript).

Figure 5.

89Zr-pan PET/CT has higher specificity for distinguishing metastatic LNs compared to 18F-FDG PET/CT. All LNs identified on 18F-FDG PET/CT and 89Zr-pan PET/CT scans were mapped onto individual subject's necks with corresponding pathology results (A). The range of SUVmax for LNs detected on18F-FDG PET/CT and 89Zr-pan PET/CT and the range of MFI and SBR for LNs were evaluated using standard boxplots with median as center line and mean as the diamond. P-values were determined using the Kruskal–Wallis test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; B). ROC curves were evaluated for LNs detected on 18F-FDG PET/CT and 89Zr-pan PET/CT (C). The ROC curves for the combined PET/CTs had higher AUC and specificity compared with 18F-FDG PET/CT alone (95% CIs available in the manuscript).

Close modal

Dosimetry results

The liver received the highest organ dose, with a mean of 1.03 ± 0.38 mGy/MBq, and the maximum observed value was 1.58 mGy/MBq. The small intestine and the upper large intestine received a mean of 0.54 ± 0.15 and 0.53 ± 0.16 mGy/MBq respectively (maximum: 0.70 and 0.68 mGy/MBq) and kidneys and spleen received a mean dose of 0.40 ± 0.04 and 0.29 ± 0.07 mGy/MBq, respectively (maximum dose: 0.44 and 0.38 mGy/MBq). Absorbed doses in the remainder of the body were 0.14 ± 0.03 mGy/MBq (Supplementary Table S2).

This study demonstrates that 89Zr-pan is a safe and specific radiotracer that can be used to improve accuracy in preoperative detection and staging of HNSCC, particularly in combination with 18F-FDG PET/CT. Although this was a phase 1 pilot study, we show limited/no adverse effects associated with the study drug and a high specificity and positive predictive value of 89Zr-pan PET/CT to detect HNSCC lesions in the head and neck region, as well as in the whole body. The data on the combined use of 18F-FDG PET/CT and 89Zr-pan PET/CT showed high specificity compared with 18F-FDG PET/CT alone, suggesting an opportunity to improve diagnostic accuracy and reduce the use of invasive diagnostic procedures. Moreover, this method could obviate invasive procedures necessary to confirm histology in lesions detected by 18F-FDG PET/CT coupled with the obvious advantage of decreasing risk to the patient and improve cost-effectiveness.

Pan800 has previously been studied in HNSCC and has been shown to have high sensitivity and specificity for detecting HNSCC in diverse applications, including intraoperative detection of the sentinel margin, assessment of the wound bed for residual tumor, and distinguishing between benign and metastatic LNs (18–20, 26). We independently evaluated two HNSCC-specific tracers, 89Zr-pan and pan800, in our study. We found several similarities between the IRDye800- and 89Zr- labeled panitumumab: there was generally higher signal in the primary tumor compared to the metastatic LNs (Fig. 3B), and there was a high degree of correlation between each imaging modality. This correlation between pan800 and 89Zr-pan allows us to infer that 89Zr-pan and pan800 have similar discriminating ability in distinguishing cancer vs. non-cancer. Using these combined preoperative and intraoperative modalities, a unique hybrid approach could be taken in which preoperative 89Zr-pan PET/CT information could be validated in the operating room through imaging of pan800, thereby allowing the surgeon to confirm target lesion(s) seen on preoperative imaging.

We relied on the quantifiable PET signal, the SUVmax to determine the sensitivity and specificity of 89Zr-pan. Previous studies evaluating 18F-FDG PET/CT used a SUVmax cut-off of 3.5 for detecting nodal metastases from oral SCC with (sensitivity 67.9, specificity 94.6; ref. 27). For 89Zr-pan PET/CT, this cut off was found to be 2.6 (sensitivity 59.1, specificity 100) from our pilot study. The optimal scan time and dose for 89Zr-pan remains undefined. However, based on our current study, the SBR using muscle as background begins to decrease after Day 3, suggesting that the optimal scan time is prior to Day 3 (Supplementary Fig. S2B).

89Zr has been used by multiple research groups to label antibodies given its favorable half-life (28–31). The higher radiation dose was accommodated with a 1 mCi the maximum administered activity, which is a limitation. However, there are no alternative radioisotopes currently for in vivo PET imaging of antibodies. In order to improve image quality given the limited administered activity, longer scanning at each bed position, and using modern PET scanners with increased axial Field of View and SiPM detectors could result in improved image quality. Compared to the study by Lindenberg and colleagues (24), we showed decreased absorbed dose in the liver and spleen (P = 0.06, Kruskal–Wallis test; Supplementary Table S2), most likely due to a loading dose of “cold” panitumumab (pan800) in our study. These findings are in line with previous studies, in which a loading dose was demonstrated to decrease radiation exposure to organs as well as increased SUVmax values resulting in improved visualization of target lesions (31–39). Despite the loading dose, the major limitation of 89Zr-pan PET/CT is the relatively high SUVmax signal in the liver, a consequence of the liver acting as a sink due to its high EGFR expression in hepatocytes in adult patients (40), which in turn also limits the dose of 89Zr-pan that can be given and the image quality that can be achieved. Moreover, this also limits the ability to detect liver metastases. Fortunately, in HNSCC the highest incidence of distant metastases occurs in the lungs (up to 78%), bones (up to 30%) and the liver (17%; refs. 41, 42); thus, 89Zr-pan PET/CT has the opportunity to further lesions after detection of the initial lesions with the current standard of care imaging. However, 89Zr-pan may have limited application in other solid tumor with frequent metastases to the liver, such as pancreatic cancer (43). In addition, the longer half-life (compared with 18F-FDG) of 89Zr-pan requires systemic administration of the drug up to 3 days prior to imaging, which may result in logistical challenges for routine clinical use. Further studies with increased sample size to determine the optimal dose and imaging window are needed to fully elucidate the potential value added of 89Zr-pan PET/CT as a tumor imaging adjunct to the current standard of care of 18F-FDG PET/CT.

A. Iagaru reports grants from GE Healthcare, personal fees from ITM, Clarity, and Telix, and grants and personal fees from Novartis outside the submitted work. E.L. Rosenthal reports grants from NIH during the conduct of the study. No disclosures were reported by the other authors.

Y.J. Lee: Data curation, software, formal analysis, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. N.S. van den Berg: Conceptualization, data curation, formal analysis, validation, investigation, visualization, methodology, project administration, writing–review and editing. H. Duan: Data curation, formal analysis, validation, investigation, visualization, writing–review and editing. E. Azevedo: Validation, investigation, visualization, project administration. V. Ferri: Software, formal analysis, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing. M. Hom: Data curation, validation, investigation, visualization, project administration, writing–review and editing. R.C. Raymundo: Investigation, methodology, project administration, writing–review and editing. A. Valencia: Data curation, validation, investigation, methodology, project administration, writing–review and editing. J. Castillo: Validation, investigation, methodology, project administration, writing–review and editing. B. Shen: Resources, supervision, validation, investigation, visualization, project administration, writing–review and editing. Q. Zhou: Investigation, visualization, methodology, writing–review and editing. L. Freeman: Data curation, investigation, visualization, project administration, writing–review and editing. M. Koran: Investigation, methodology, writing–review and editing. M.J. Kaplan: Data curation, validation, investigation, visualization, project administration, writing–review and editing. A. Colevas: Investigation, methodology, project administration, writing–review and editing. F.M. Baik: Validation, investigation, project administration, writing–review and editing. F.T. Chin: Resources, supervision, validation, investigation, project administration, writing–review and editing. B.A. Martin: Data curation, formal analysis, validation, investigation, visualization, methodology, writing–review and editing. A. Iagaru: Resources, data curation, software, formal analysis, supervision, investigation, visualization, methodology, project administration, writing–review and editing. E.L. Rosenthal: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, investigation, methodology, project administration, writing–review and editing.

We thank Yifei Ma (Stanford University) for providing statistical consultation and Dr. Jared Grice (Vanderbilt University) for providing his expertise in medical physics. This work was supported in part by the Stanford Comprehensive Cancer Center (ELR), the Netherlands Organization for Scientific Research (NSVDB; Rubicon; 019.171LW.022), the National Institutes of Health and the National Cancer Institute (ELR; R01CA190306) and the National Institute on Deafness and Other Communication Disorders (YL; T32DC015209).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Veit-Haibach
P
,
Luczak
C
,
Wanke
I
,
Fischer
M
,
Egelhof
T
,
Beyer
T
, et al
.
TNM staging with FDG-PET/CT in patients with primary head and neck cancer
.
Eur J Nucl Med Mol Imaging
2007
;
34
:
1953
62
.
2.
Xu
G-Z
,
Guan
D-J
,
He
Z-Y
.
18FDG-PET/CT for detecting distant metastases and second primary cancers in patients with head and neck cancer. A meta-analysis
.
Oral Oncol
2011
;
47
:
560
5
.
3.
Goel
R
,
Moore
W
,
Sumer
B
,
Khan
S
,
Sher
D
,
Subramaniam
RM
.
Clinical practice in PET/CT for the management of head and neck squamous cell cancer
.
AJR Am J Roentgenol.
2017
;
209
:
289
303
.
4.
Britt
CJ
,
Maas
AM
,
Kennedy
TA
,
Hartig
GK
.
Incidental findings on FDG PET/CT in head and neck cancer
.
Otolaryngol Head Neck Surg
2018
;
158
:
484
8
.
5.
Casselden
E
,
Sheerin
F
,
Winter
SC
.
Incidental findings on 18-FDG PET-CT in head and neck cancer. A retrospective case-control study of incidental findings on 18-FDG PET-CT in patients with head and neck cancer
.
Eur Arch Otorhinolaryngol
2019
;
276
:
243
7
.
6.
Schoonbeek
RC
,
Bult
FFS
,
Plaat
BEC
,
Witjes
MJH
,
van der Hoorn
A
,
van Dijk
BAC
, et al
.
Incidental findings during the diagnostic work-up in the head and neck cancer pathway: Effects on treatment delay and survival
.
Oral Oncol
2021
;
118
:
105350
.
7.
Spiegelberg
D
,
Nilvebrant
J
.
CD44v6-Targeted Imaging of Head and Neck Squamous Cell Carcinoma: Antibody-Based Approaches
.
Contrast Media Mol Imaging
2017
;
2017
:
2709547
.
8.
Lobeek
D
,
Rijpkema
M
,
Terry
SYA
,
Molkenboer-Kuenen
JDM
,
Joosten
L
,
van Genugten
EAJ
, et al
.
Imaging angiogenesis in patients with head and neck squamous cell carcinomas by [(68)Ga]Ga-DOTA-E-[c(RGDfK)](2) PET/CT
.
Eur J Nucl Med Mol Imaging
2020
;
47
:
2647
55
.
9.
Schartinger
VH
,
Dudás
J
,
Decristoforo
C
,
Url
C
,
Schnabl
J
,
Göbel
G
, et al
.
68Ga-DOTA0-Tyr3-octreotide positron emission tomography in head and neck squamous cell carcinoma
.
Eur J Nucl Med Mol Imaging
2013
;
40
:
1365
72
.
10.
Komar
G
,
Seppänen
M
,
Eskola
O
,
Lindholm
P
,
Grönroos
TJ
,
Forsback
S
, et al
.
18F-EF5: a new PET tracer for imaging hypoxia in head and neck cancer
.
J Nucl Med
2008
;
49
:
1944
51
.
11.
Grandis
JR
,
Tweardy
DJ
.
Elevated levels of transforming growth factor alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer
.
Cancer Res
1993
;
53
:
3579
84
.
12.
Santini
J
,
Formento
JL
,
Francoual
M
,
Milano
G
,
Schneider
M
,
Dassonville
O
, et al
.
Characterization, quantification, and potential clinical value of the epidermal growth factor receptor in head and neck squamous cell carcinomas
.
Head Neck
1991
;
13
:
132
9
.
13.
Tian
Y
,
Lin
J
,
Tian
Y
,
Zhang
G
,
Zeng
X
,
Zheng
R
, et al
.
Efficacy and safety of anti-EGFR agents administered concurrently with standard therapies for patients with head and neck squamous cell carcinoma: a systematic review and meta-analysis of randomized controlled trials
.
Int J Cancer
2018
;
142
:
2198
206
.
14.
Johnson
DE
,
Burtness
B
,
Leemans
CR
,
Lui
VWY
,
Bauman
JE
,
Grandis
JR
.
Head and neck squamous cell carcinoma
.
Nat Rev Dis Primer
2020
;
6
:
92
.
15.
Xu
MJ
,
Johnson
DE
,
Grandis
JR
.
EGFR-targeted therapies in the post-genomic era
.
Cancer Metastasis Rev
2017
;
36
:
463
73
.
16.
Sundvall
M
,
Karrila
A
,
Nordberg
J
,
Grénman
R
,
Elenius
K
.
EGFR targeting drugs in the treatment of head and neck squamous cell carcinoma
.
Expert Opin Emerg Drugs
2010
;
15
:
185
201
.
17.
Gao
RW
,
Teraphongphom
NT
,
van den Berg
NS
,
Martin
BA
,
Oberhelman
NJ
,
Divi
V
, et al
.
Determination of tumor margins with surgical specimen mapping using Near-infrared fluorescence
.
Cancer Res
2018
;
78
:
5144
54
.
18.
van Keulen
S
,
Nishio
N
,
Birkeland
A
,
Fakurnejad
S
,
Martin
B
,
Forouzanfar
T
, et al
.
The sentinel margin: Intraoperative ex vivo specimen mapping using relative fluorescence intensity
.
Clin Cancer Res
2019
;
25
:
4656
62
.
19.
Nishio
N
,
van den Berg
NS
,
van Keulen
S
,
Martin
BA
,
Fakurnejad
S
,
Teraphongphom
N
, et al
.
Optical molecular imaging can differentiate metastatic from benign lymph nodes in head and neck cancer
.
Nat Commun
2019
;
10
:
5044
.
20.
Krishnan
G
,
van den Berg
NS
,
Nishio
N
,
Juniper
G
,
Pei
J
,
Zhou
Q
, et al
.
Metastatic and sentinel lymph node mapping using intravenously delivered Panitumumab-IRDye800CW
.
Theranostics
2021
;
11
:
7188
98
.
21.
Vonk
J
,
de Wit
JG
,
Voskuil
FJ
,
Tang
YH
,
Hooghiemstra
WT
,
Linssen
MD
, et al
.
Epidermal growth factor receptor targeted fluorescence molecular imaging for postoperative lymph node assessment in patients with oral cancer
.
J Nucl Med
2022
;
63
:
672
8
.
22.
Kinahan
PE
,
Fletcher
JW
.
Positron emission tomography-computed tomography standardized uptake values in clinical practice and assessing response to therapy
.
Semin Ultrasound CT MR
2010
;
31
:
496
505
.
23.
Bolch
WE
,
Eckerman
KF
,
Sgouros
G
,
Thomas
SR
.
MIRD pamphlet No. 21: a generalized schema for radiopharmaceutical dosimetry–standardization of nomenclature
.
J Nucl Med
2009
;
50
:
477
84
.
24.
Lindenberg
L
,
Adler
S
,
Turkbey
IB
,
Mertan
F
,
Ton
A
,
Do
K
, et al
.
Dosimetry and first human experience with (89)Zr-panitumumab
.
Am J Nucl Med Mol Imaging
2017
;
7
:
195
203
.
25.
Rosenthal
EL
,
Moore
LS
,
Tipirneni
K
,
de Boer
E
,
Stevens
TM
,
Hartman
YE
, et al
.
Sensitivity and specificity of cetuximab-IRDye800CW to identify regional metastatic disease in head and neck cancer
.
Clin Cancer Res
2017
;
23
:
4744
52
.
26.
Nishio
N
,
van den Berg
NS
,
van Keulen
S
,
Martin
BA
,
Fakurnejad
S
,
Zhou
Q
, et al
.
Optimal dosing strategy for fluorescence-guided surgery with panitumumab-IRDye800CW in head and neck cancer
.
Mol Imaging Biol
2020
;
22
:
156
64
.
27.
Kitajima
K
,
Suenaga
Y
,
Minamikawa
T
,
Komori
T
,
Otsuki
N
,
Nibu
K-I
, et al
.
Clinical significance of SUVmax in (18)F-FDG PET/CT scan for detecting nodal metastases in patients with oral squamous cell carcinoma
.
SpringerPlus
2015
;
4
:
718
.
28.
Bensch
F
,
Lamberts
LE
,
Smeenk
MM
,
Jorritsma-Smit
A
,
Lub-de Hooge
MN
,
van Scheltinga
AGTT
, et al
.
89Zr-Lumretuzumab PET imaging before and during HER3 antibody lumretuzumab treatment in patients with solid tumors
.
Clin Cancer Res
2017
;
23
:
6128
37
.
29.
Pandit-Taskar
N
,
Postow
MA
,
Hellmann
MD
,
Harding
JJ
,
Barker
CA
,
O'Donoghue
JA
, et al
.
First-in-humans imaging with 89Zr-Df-IAB22M2C anti-CD8 minibody in patients with solid malignancies: preliminary pharmacokinetics, biodistribution, and lesion targeting
.
J Nucl Med
2020
;
61
:
512
9
.
30.
Menke-van der Houven van Oordt
CW
,
McGeoch
A
,
Bergstrom
M
,
McSherry
I
,
Smith
DA
,
Cleveland
M
, et al
.
Immuno-PET imaging to assess target engagement: experience from 89Zr-anti-HER3 mAb (GSK2849330) in patients with solid tumors
.
J Nucl Med
2019
;
60
:
902
9
.
31.
Dijkers
EC
,
Oude Munnink
TH
,
Kosterink
JG
,
Brouwers
AH
,
Jager
PL
,
de Jong
JR
, et al
.
Biodistribution of 89Zr-trastuzumab and PET imaging of HER2-positive lesions in patients with metastatic breast cancer
.
Clin Pharmacol Ther
2010
;
87
:
586
92
.
32.
Bensch
F
,
van der Veen
EL
,
Lub-de Hooge
MN
,
Jorritsma-Smit
A
,
Boellaard
R
,
Kok
IC
, et al
.
89Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer
.
Nat Med
2018
;
24
:
1852
8
.
33.
van Loon
J
,
Even
AJG
,
Aerts
HJWL
,
Öllers
M
,
Hoebers
F
,
van Elmpt
W
, et al
.
PET imaging of zirconium-89 labelled cetuximab: a phase I trial in patients with head and neck and lung cancer
.
Radiother Oncol
2017
;
122
:
267
73
.
34.
Börjesson
PKE
,
Jauw
YWS
,
Boellaard
R
,
de Bree
R
,
Comans
EFI
,
Roos
JC
, et al
.
Performance of immuno-positron emission tomography with zirconium-89-labeled chimeric monoclonal antibody U36 in the detection of lymph node metastases in head and neck cancer patients
.
Clin Cancer Res
2006
;
12
:
2133
40
.
35.
Gaykema
SBM
,
Brouwers
AH
,
Lub-de Hooge
MN
,
Pleijhuis
RG
,
Timmer-Bosscha
H
,
Pot
L
, et al
.
89Zr-bevacizumab PET imaging in primary breast cancer
.
J Nucl Med
2013
;
54
:
1014
8
.
36.
van Asselt
SJ
,
Oosting
SF
,
Brouwers
AH
,
Bongaerts
AHH
,
de Jong
JR
,
Lub-de Hooge
MN
, et al
.
Everolimus reduces (89)Zr-Bevacizumab tumor uptake in patients with neuroendocrine tumors
.
J Nucl Med
2014
;
55
:
1087
92
.
37.
Pandit-Taskar
N
,
O'Donoghue
JA
,
Beylergil
V
,
Lyashchenko
S
,
Ruan
S
,
Solomon
SB
, et al
.
89Zr-huJ591 immuno-PET imaging in patients with advanced metastatic prostate cancer
.
Eur J Nucl Med Mol Imaging
2014
;
41
:
2093
105
.
38.
Pandit-Taskar
N
,
O'Donoghue
JA
,
Durack
JC
,
Lyashchenko
SK
,
Cheal
SM
,
Beylergil
V
, et al
.
A phase I/II study for analytic validation of 89Zr-J591 ImmunoPET as a molecular imaging agent for metastatic prostate cancer
.
Clin Cancer Res
2015
;
21
:
5277
85
.
39.
Oosting
SF
,
Brouwers
AH
,
van Es
SC
,
Nagengast
WB
,
Munnink
THO
,
Lub-de Hooge
MN
, et al
.
89Zr-bevacizumab PET visualizes heterogeneous tracer accumulation in tumor lesions of renal cell carcinoma patients and differential effects of antiangiogenic treatment
.
J Nucl Med
2015
;
56
:
63
9
.
40.
Berasain
C
,
Latasa
MU
,
Urtasun
R
,
Goñi
S
,
Elizalde
M
,
Garcia-Irigoyen
O
, et al
.
Epidermal Growth Factor Receptor (EGFR) Crosstalks in Liver Cancer
.
Cancers
2011
;
3
:
2444
61
.
41.
Duprez
F
,
Berwouts
D
,
De Neve
W
,
Bonte
K
,
Boterberg
T
,
Deron
P
, et al
.
Distant metastases in head and neck cancer
.
Head Neck
2017
;
39
:
1733
43
.
42.
Ferlito
A
,
Shaha
AR
,
Silver
CE
,
Rinaldo
A
,
Mondin
V
.
Incidence and sites of distant metastases from head and neck cancer
.
ORL J Otorhinolaryngol Relat Spec
2001
;
63
:
202
7
.
43.
Ryan
DP
,
Hong
TS
,
Bardeesy
N
.
Pancreatic adenocarcinoma
.
N Engl J Med
2014
;
371
:
1039
49
.

Supplementary data