Skip to main content

Main menu

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Journal of Nuclear Medicine

Advanced Search

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • View or Listen to JNM Podcast
  • Visit JNM on Facebook
  • Join JNM on LinkedIn
  • Follow JNM on Twitter
  • Subscribe to our RSS feeds
Research ArticleClinical Investigation

Diagnostic Accuracy of 68Ga-FAPI Versus 18F-FDG PET in Patients with Various Malignancies

Nader Hirmas, Rainer Hamacher, Miriam Sraieb, Lukas Kessler, Kim M. Pabst, Francesco Barbato, Helena Lanzafame, Stefan Kasper, Michael Nader, Claudia Kesch, Bastian von Tresckow, Hubertus Hautzel, Clemens Aigner, Martin Glas, Martin Stuschke, Sherko Kümmel, Philipp Harter, Celine Lugnier, Waldemar Uhl, Boris Hadaschik, Viktor Grünwald, Jens T. Siveke, Ken Herrmann and Wolfgang P. Fendler
Journal of Nuclear Medicine March 2024, 65 (3) 372-378; DOI: https://doi.org/10.2967/jnumed.123.266652
Nader Hirmas
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rainer Hamacher
2Department of Medical Oncology, West German Cancer Center, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Miriam Sraieb
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lukas Kessler
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kim M. Pabst
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesco Barbato
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Helena Lanzafame
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stefan Kasper
2Department of Medical Oncology, West German Cancer Center, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael Nader
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Claudia Kesch
3Department of Urology, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bastian von Tresckow
4Department of Hematology and Stem Cell Transplantation, West German Cancer Center, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hubertus Hautzel
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clemens Aigner
5Department of Thoracic Surgery and Thoracic Endoscopy, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Martin Glas
6Division of Clinical Neurooncology, Department of Neurology, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Martin Stuschke
7Department of Radiation Therapy, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sherko Kümmel
8Breast Unit, Kliniken Essen–Mitte, Essen, Germany;
9Department of Gynecology with Breast Center, Charité–Universitätsmedizin Berlin, Berlin, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Philipp Harter
10Department of Gynecology and Gynecologic Oncology, Evangelische Kliniken Essen-Mitte, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Celine Lugnier
11Department of Hematology and Oncology with Palliative Care, Ruhr University Bochum, Bochum, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Waldemar Uhl
12Department of General and Visceral Surgery, Ruhr University Bochum, Bochum, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Boris Hadaschik
3Department of Urology, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Viktor Grünwald
3Department of Urology, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jens T. Siveke
13Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany; and
14Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK partner site Essen), German Cancer Research Center, Heidelberg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ken Herrmann
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wolfgang P. Fendler
1Department of Nuclear Medicine, German Cancer Consortium–University Hospital Essen, University of Duisburg–Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • PDF
Loading

Visual Abstract

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Abstract

To assess the diagnostic accuracy of 68Ga-labeled fibroblast activation protein inhibitor (FAPI) and 18F-labeled FDG PET for the detection of various tumors, we performed a head-to-head comparison of both imaging modalities across a range of tumor entities as part of our ongoing 68Ga-FAPI PET observational trial. Methods: The study included 115 patients with 8 tumor entities who received imaging with 68Ga-FAPI for tumor staging or restaging between October 2018 and March 2022. Of those, 103 patients received concomitant imaging with 68Ga-FAPI and 18F-FDG PET and had adequate lesion validation for accuracy analysis. Each scan was evaluated for the detection of primary tumor, lymph nodes, and visceral and bone metastases. True or false positivity and negativity to detected lesions was assigned on the basis of histopathology from biopsies or surgical excision, as well as imaging validation. Results: 68Ga-FAPI PET revealed higher accuracy than 18F-FDG PET in the detection of colorectal cancer (n = 14; per-patient, 85.7% vs. 78.6%; per-region, 95.6% vs. 91.1%) and prostate cancer (n = 22; per-patient, 100% vs. 90.9%; per-region, 96.4% vs. 92.7%). 68Ga-FAPI PET and 18F-FDG PET had comparable per-patient accuracy in detecting breast cancer (n = 16, 100% for both) and head and neck cancers (n = 10, 90% for both modalities). 68Ga-FAPI PET had lower per-patient accuracy than 18F-FDG PET in cancers of the bladder (n = 12, 75% vs. 100%) and kidney (n = 10, 80% vs. 90%), as well as lymphoma (n = 9, 88.9% vs. 100%) and myeloma (n = 10, 80% vs. 90%). Conclusion: 68Ga-FAPI PET demonstrated higher diagnostic accuracy than 18F-FDG PET in the diagnosis of colorectal cancer and prostate cancer, as well as comparable diagnostic performance for cancers of the breast and head and neck. Accuracy and impact on management will be further assessed in an ongoing prospective interventional trial (NCT05160051).

  • FAPI
  • FDG
  • PET
  • oncology
  • theranostic
  • accuracy

Imaging is fundamental in the treatment of malignancies, with varying detection rates depending on the tumor entity and diagnostic modality. PET of cancer cells using 18F-FDG PET acquires additional molecular information useful for the detection of disease recurrence and metastases, response assessment, disease management, and prognostication (1–6). However, drawbacks of 18F-FDG include false-positive findings due to physiologic uptake or inflammatory responses, as well as false-negative findings due to elevated serum blood glucose levels. As such, targeting of cancer cells using alternative radioisotopes has been an area of growing interest.

Cancer-associated fibroblasts, a constituent of the tumor microenvironment, are involved in tumor growth, migration, and progression (7). Fibroblast activation protein (FAP) α is expressed by cancer-associated fibroblasts, a marker associated with protumorigenic functions (8–12) and, therefore, a suitable target for diagnostic and therapeutic purposes. Multiple preclinical and clinical studies have shown the promise of FAP-directed therapies, including radiolabeled FAP inhibitors (FAPIs), which exhibit favorable properties in cancer diagnosis and therapy (13–18). These properties include, but are not limited to, fast imaging times, high contrast in tumor lesions, and no dietary requirements with regard to imaging, as well as acceptable side effects and long tumor retention times with regard to therapy.

Because of the favorable characteristics of this imaging modality, patients were referred for clinical 68Ga-FAPI PET staging, both at initial diagnosis and for reevaluation, and were offered subsequent enrollment in our prospective observational 68Ga-FAPI registry.

In this report, we assess the diagnostic accuracy of 68Ga-FAPI compared with 18F-FDG PET separately for various tumor entities by analyzing sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and accuracy on per-patient and per-region bases.

MATERIALS AND METHODS

Study Design and Participants

Until March 2022, adult patients who underwent clinical 68Ga-FAPI PET were offered the possibility to consent to a prospective observational trial for correlation and clinical follow-up of PET findings (NCT04571086). Patients signed a written informed consent form, and evaluation of data was approved by the ethics committee of the University Duisburg–Essen (20-9485-BO and 19-8991-BO). We previously reported data on 68Ga-FAPI PET uptake and accuracy in sarcoma (n = 47 (19)), as well as 68Ga-FAPI PET uptake in mixed cohorts (n = 69 (20), n = 91 (21), and n = 324 (22)). Patients with sarcoma, pancreatic cancer, and pleural mesothelioma have been excluded from this analysis since the results have already been or will be published separately. Moreover, solid tumor entities with fewer than 10 patients per entity for 68Ga-FAPI PET accuracy assessment were excluded from this analysis.

Details of data collection (23,24), imaging and administration of radioligands (20,25,26), imaging analysis, lesion validation, follow-up (27), and statistical analysis are provided in the supplemental materials (available at http://jnm.snmjournals.org).

RESULTS

Patient Characteristics

We identified 133 patients, of whom 115 with adequate lesion validation were included in this analysis. In total, 8 tumor entities and 313 regions were analyzed; patient characteristics (n = 115) are outlined in Table 1. The median age was 63 y (interquartile range, 17 y). The most common tumor entities were prostate cancer (22/115, 19%), head and neck cancers (18/115, 16%), breast cancer (16/115, 14%), colorectal cancer (15/115, 13%), and bladder cancer (12/115, 10%). Most patients (81/115, 70%) underwent 68Ga-FAPI PET imaging for restaging purposes. A total of 103 (90%) patients underwent concomitant imaging via 68Ga-FAPI and 18F-FDG PET and had adequate lesion validation for the accuracy analysis, and this set of patients was included in the composite analysis.

View this table:
  • View inline
  • View popup
TABLE 1.

Patient Characteristics (n = 115)

Composite Analysis: Higher Diagnostic Accuracy with 68Ga-FAPI PET Than with 18F-FDG PET

68Ga-FAPI PET showed higher diagnostic accuracy than 18F-FDG PET in the diagnosis of colorectal cancer and prostate cancer as listed in Table 2.

View this table:
  • View inline
  • View popup
TABLE 2.

Comparison of Diagnostic Efficacy Between 68Ga-FAPI and 18F-FDG PET (Per-Patient and Per-Region Analysis) for Tumors in Which 68Ga-FAPI Outperformed 18F-FDG PET

At a per-patient level in colorectal cancer, 68Ga-FAPI PET was superior to 18F-FDG PET in accuracy (85.7% vs. 78.6%), sensitivity (90.9% vs. 81.8%), and NPV (66.7% vs. 50%). At a per-region level, 68Ga-FAPI PET was superior to 18F-FDG PET in accuracy (95.6% vs. 91.1%), sensitivity (94.1% vs. 88.2%), and PPV (94.1% vs. 88.2%).

Furthermore, at a per-patient level in prostate cancer, 68Ga-FAPI PET was superior to 18F-FDG PET in accuracy (100% vs. 90.9%) and sensitivity (100% vs. 90.9%). At a per-region level, 68Ga-FAPI PET was superior to 18F-FDG PET in sensitivity (94.3% vs. 88.6%) and NPV (90.9% vs. 83.3%).

Composite Analysis: Comparable Diagnostic Accuracy Between 68Ga-FAPI PET and 18F-FDG PET

68Ga-FAPI PET was comparable to 18F-FDG PET in the diagnosis of breast cancer and head and neck cancers as listed in Table 3.

View this table:
  • View inline
  • View popup
TABLE 3.

Comparison of Diagnostic Efficacy Between 68Ga-FAPI and 18F-FDG PET (Per-Patient and Per-Region Analysis) for Tumors in Which 68Ga-FAPI Was Comparable to 18F-FDG PET

At a per-patient level in breast cancer, 68Ga-FAPI PET and 18F-FDG PET showed equal accuracy, sensitivity, specificity, PPV, and NPV (all 100%). At a per-region level, 68Ga-FAPI PET showed accuracy (97.9% vs. 100%) and sensitivity (96.6% vs. 100%) comparable to those of 18F-FDG PET but lower NPV (94.7% vs. 100%).

At a per-patient level in head and neck cancers, 68Ga-FAPI PET and 18F-FDG PET showed equal accuracy (90%), sensitivity (100%), and PPV (90%). At a per-region level, 68Ga-FAPI PET showed accuracy (90.3% vs. 93.6%) and specificity (86.7% for both) comparable to those of 18F-FDG PET but lower sensitivity (93.8% vs. 100%) and NPV (92.9% vs. 100%).

Composite Analysis: Lower Diagnostic Accuracy with 68Ga-FAPI PET Than with 18F-FDG PET

68Ga-FAPI PET showed lower accuracy than 18F-FDG PET in the diagnosis of bladder and kidney cancers, lymphoma, and myeloma as shown in Table 4.

View this table:
  • View inline
  • View popup
TABLE 4.

Comparison of Diagnostic Efficacy Between 68Ga-FAPI and 18F-FDG PET (Per-Patient and Per-Region Analysis) for Tumors in Which 68Ga-FAPI Underperformed in Comparison to 18F-FDG PET

At a per-patient level in bladder cancer, 68Ga-FAPI PET showed lower accuracy (75% vs. 100%), sensitivity (72.7% vs. 100%), and NPV (25% vs. 100%) than 18F-FDG PET. At a per-region level, 68Ga-FAPI PET showed lower accuracy (89.2% vs. 94.4%), sensitivity (78.6% vs. 92.3%), and NPV (88% vs. 95.7%) than 18F-FDG PET.

At a per-patient level in kidney cancer, 68Ga-FAPI PET showed sensitivity comparable to that of 18F-FDG PET (87.5% for both) but lower accuracy (80% vs. 90%), specificity (50% vs. 100%), and PPV (87.5% vs. 100%). At a per-region level, 68Ga-FAPI PET showed accuracy (90.3% vs. 93.6%), sensitivity (92.9% for both), and NPV (93.8% vs. 94.1%) comparable to those of 68Ga-FAPI PET but lower specificity (88.2% vs. 94.1%) and PPV (86.7% vs. 92.9%).

At a per-patient level in lymphoma, 68Ga-FAPI PET showed lower accuracy (88.9% vs. 100%), sensitivity (87.5% vs. 100%), and NPV (50% vs. 100%) than 18F-FDG PET. At a per-region level, 68Ga-FAPI PET showed lower accuracy (90% vs. 96.7%), sensitivity (78.6% vs. 100%), and NPV (84.2% vs. 100%) than 18F-FDG PET.

Finally, for myeloma at per-patient and per-region levels, accuracy (80% vs. 90%) and sensitivity (75% vs. 87.5%) were lower with 68Ga-FAPI PET than with 18F-FDG PET.

Histopathology-Only Analysis

In a subgroup of 45 patients and 5 tumor entities, accuracy was assessed by histopathology validation only (Supplemental Table 1). In line with the findings of the composite analysis, 68Ga-FAPI PET demonstrated higher accuracy than 18F-FDG PET for prostate cancer, comparable accuracy for breast cancer and colorectal cancer, and lower accuracy for bladder and kidney cancers.

DISCUSSION

Here, we compare the diagnostic accuracy of 68Ga-FAPI and 18F-FDG PET for various tumors. Tumor validation by a composite reference standard revealed that the diagnostic accuracy of 68Ga-FAPI PET was higher than that of 18F-FDG PET in colorectal cancer and prostate cancer, comparable in breast cancer and head and neck cancer, and lower in bladder and kidney cancers, lymphoma, and myeloma. Histopathology-only analysis revealed that the diagnostic accuracy of 68Ga-FAPI PET was higher than that of 18F-FDG PET in prostate cancer, comparable in breast and colorectal cancers, and lower in bladder and kidney cancers.

For cancers of the abdomen and pelvis, 68Ga-FAPI uptake was low in normal parenchyma, such as bowel (SUVmax range, 0.08–3.56), liver (SUVmax range, 0.47–2.91), and spleen (SUVmax range, 0.64–2.81) (15,28,29). This improves tumor delineation, with absolute and tumor-to-liver uptakes being higher on 68Ga-FAPI PET than on 18F-FDG PET, which may lead to superior diagnostic accuracy (22). This is particularly relevant in abdominal surgery, for example, after which patients are required to take nothing by mouth until bowel recovery. Also, the prevalence of coexisting diabetes (≤15.5% in patients with colon cancer, for instance (30)) poses limitations for molecular imaging with 18F-FDG PET. 68Ga-FAPI PET in such a context has protocol advantages, given that no diet or fasting is required in preparation for imaging, and image acquisition can take place a few minutes after tracer application. 68Ga-FAPI PET, therefore, has the potential to replace 18F-FDG for abdominal staging.

Our findings are corroborated by other studies that have also shown 68Ga-FAPI PET to have diagnostic accuracy superior to that of 18F-FDG PET in breast cancer (31–33) and head and neck cancers (34–36). Moreover, reports have shown that 68Ga-FAPI PET can detect PSMA-negative prostate cancer lesions (37–39), which can aid in the diagnostic process, with potential therapeutic implications.

With regard to lymphoma and myeloma, several studies have shown that 68Ga-FAPI PET is inferior to (or at best, not superior to) 18F-FDG PET (40–43). For example, in comparison to colorectal cancer, lymphoma lesions show lower uptake with 68Ga-FAPI than with 18F-FDG PET (22,41,44), higher background uptake, and, thus, lower tumor-to-background values (e.g., median SUVmax of 7.4 vs. 22.5 and median liver tumor-to-background ratio of 6.4 vs. 10.5 for 68Ga-FAPI vs. 18F-FDG PET, respectively (22)). Taking this a step further, using systematic lesion validation and follow-up, our study revealed 68Ga-FAPI to be less accurate than 18F-FDG PET in lymphoma and myeloma.

An ongoing prospective clinical trial at our department (NCT05160051) is exploring the diagnostic accuracy of 68Ga-FAPI-46 PET and its effect on patient management and interreader reproducibility for different tumor entities. An interim analysis of findings has shown that 68Ga-FAPI PET is associated with a lower rate of false-positive findings, especially in lymph node assessments (44).

With high tumor and low organ uptakes (22), as well as diagnostic accuracy across various tumor entities, 68Ga-FAPI PET has a role as a gatekeeper for FAP-directed radioligand therapy. Feasibility of FAP radioligand therapy has been reported for 90Y- and 153Sm-labeled compounds in breast (13) and ovarian (45) cancer, as well as sarcomas and pancreatic cancers (17,46). 177Lu-labeled compounds have also been used in multiple advanced and refractory tumors, including thyroid cancer (16,47–49). In patients with intense FAP expression on 68Ga-FAPI PET, 90Y-FAPI-46 radioligand therapy led to disease control in about one third of patients with initially progressive sarcomas, pancreatic cancer, and other cancers (50), and the novel dimeric 177Lu-labeled FAPI radioligand (177Lu-DOTAGA.(SA.FAPi)2) led to disease control in almost half the patients with radioiodine-refractory differentiated thyroid cancer who had progressed on tyrosine kinase inhibitors (49). FAPI imaging therefore has the potential to enhance drug development with targeted clinical applications.

One notable example of a FAP-targeting drug that has shown clinical promise is talabostat, which has demonstrated tumor control in 21% of patients with colorectal cancer (51). Moreover, targeting FAP with chimeric antigen receptor T cells has shown promise in preclinical studies and case reports (52,53), and there is potential for combination with cancer vaccines or immune checkpoint inhibitors (such as PD-1 inhibitors), which would lead to further blockade of immunosuppressive factors (52). Another promising approach is using cancer vaccines that successfully target FAP, particularly the genome of stromal fibroblasts (54). As such, future drug development and its potential clinical applications may be enhanced through 68Ga-FAPI imaging, which aids in selecting patients whose tumors exhibit high 68Ga-FAPI uptake and who would potentially benefit from FAP-directed therapy. This theranostic approach also has the potential to improve clinical trial design.

Our study is limited by its retrospective design and the small number of patients included per tumor entity. Histopathology was not available for all patients, as tissue sampling is not routinely performed, and biopsy of metastatic lesions may be difficult because they may be small or remote. Thus, most lesion follow-up was based on correlative or follow-up imaging with known intrinsic limitations. Despite these limitations, the study provided valuable systematic information on the diagnostic efficacy of 68Ga-FAPI PET from an ongoing registry study evaluating 68Ga-FAPI and 18F-FDG PET, using a composite reference standard with adequate follow-up time (≤∼6 mo) and across a wide range of tumor entities, thereby adding to the growing pool of theranostic data.

CONCLUSION

When compared with 18F-FDG PET, 68Ga-FAPI PET demonstrated higher accuracy in the diagnosis of colorectal cancer and prostate cancer, as well as comparable diagnostic performance for cancers of the breast and head and neck. 68Ga-FAPI has the potential for improved staging or theranostic screening, particularly for these tumor entities.

DISCLOSURE

Rainer Hamacher is supported by the Clinician Scientist Program of the University Medicine Essen Clinician Scientist Academy (UMEA) sponsored by the faculty of medicine and Deutsche Forschungsgemeinschaft (DFG) and has received travel grants from Lilly, Novartis, and Pharma Mar, as well as fees from Lilly and Pharma Mar. Lukas Kessler is a consultant for AAA and BTG and received fees from Sanofi. Kim Pabst is a consultant for Novartis, has received a Junior Clinician Scientist Stipend from the University Medicine Essen Clinician Scientist Academy (UMEA) sponsored by the Faculty of Medicine at the University of Duisburg–Essen and the Deutsche Forschungsgemeinschaft (DFG), and has received research funding from Bayer outside the submitted work. Stefan Kasper reports personal fees and grants from AstraZeneca, Merck Serone, Merck Sharpe & Dohme, Amgen, Bristol Myers Squibb, Roche, Lilly, Servier, Incyte, and SanofiAventis outside the submitted work. Claudia Kesch has received consultant fees from Apogepha, research funding from AAA/Novartis and Curie Therapeutics, and compensation for travel from Janssen R&D, Amgen, and Bayer. Bastian von Tresckow is an advisor or consultant for Allogene, BMS/Celgene, Cerus, Incyte, IQVIA, Gilead Kite, Lilly, Miltenyi, Novartis, Noscendo, Pentixapharm, Roche, Amgen, Pfizer, Takeda, Merck Sharp & Dohme, and Gilead Kite; has received honoraria from AstraZeneca, BMS, Incyte, Lilly, Novartis, Roche Pharma AG, Takeda, and Merck Sharp & Dohme; reports research funding from Novartis (to the institution), Merck Sharp & Dohme (to the institution), and Takeda (to the institution); and reports travel support from AbbVie, AstraZeneca, Gilead Kite, Lilly, Merck Sharp & Dohme, Pierre Fabre, Roche, Takeda, and Novartis. Hubertus Hautzel reports research funding and travel support from PARI GmbH outside the submitted work. Martin Glas reports honoraria from Roche, Novartis, UCB, Abbvie, Daiichi Sankyo, Novocure, Bayer, Janssen-Cilag, Medac, Merck, and Kyowa Kirin; travel support from Novocure and Medac; and a research grant from Novocure. Ken Herrmann reports personal fees from Bayer, SIRTEX, Adacap, Curium, Endocyte, IPSEN, Siemens Healthineers, GE Healthcare, Amgen, Novartis, ymabs, Aktis, Oncology, and Pharma15, as well as personal and other fees from SOFIE Biosciences, nonfinancial support from ABX, and grants and personal fees from BTG, all of which are outside the submitted work. Boris Hadaschik declares grants to the institution from Novartis, BMS, and the German Research Foundation; consulting fees from ABX, Amgen, AstraZeneca, Bayer, BMS, Janssen, Lightpoint Medical, Novartis, and Pfizer; payment for lectures from Janssen and Monrol; support for travel or meeting attendance from AstraZeneca, Bayer, and Janssen; and participation on data safety monitoring boards for Janssen, all outside the submitted work. Philipp Harter reports honoraria from Amgen, AstraZeneca, GSK, Roche, Sotio, Stryker, Zai Lab, MSD, Clovis, Eisai, Mersana, and Exscientia. He is on the advisory board for Astra Zeneca, Roche, GSK, Clovis, Immunogen, MSD, Miltenyi, Novartis, and Eisai. He has received research funding (to the institution) from AstraZeneca, Roche, GSK, Genmab, DFG, European Union, DKH, Immunogen, Seagen, Clovis, and Novartis. Jens Siveke received honoraria as a consultant or for continuing medical education presentations from AstraZeneca, Bayer, Bristol-Myers Squibb, Eisbach Bio, Immunocore, Novartis, Roche/Genentech, and Servier; his institution receives research funding from Bristol-Myers Squibb, Celgene, Eisbach Bio, and Roche/Genentech; and he holds ownership and serves on the board of directors of Pharma15, all outside the submitted work. Wolfgang P. Fendler reports fees from SOFIE Biosciences (research funding), Janssen (consultant, speaker), Calyx (consultant, image review), Bayer (consultant, speaker, research funding), Novartis (speaker, consultant), Telix (speaker), GE Healthcare (speaker), Eczacıbaşı Monrol (speaker), Abx (speaker), and Amgen (speaker) outside the submitted work. No other potential conflict of interest relevant to this article was reported.

KEY POINTS

QUESTION: How does 68Ga-FAPI compare with 18F-FDG PET in the diagnosis of various malignancies?

PERTINENT FINDINGS: We compared the diagnostic accuracy of 68Ga-FAPI and 18F-FDG PET for the detection of various tumors. Tumor validation by a composite reference standard revealed higher diagnostic accuracy for 68Ga-FAPI PET in colorectal and prostate cancers, comparable diagnostic performance for cancers of the breast and head and neck, and lower diagnostic accuracy for bladder and kidney cancers, lymphoma, and myeloma.

IMPLICATIONS FOR PATIENT CARE: 68Ga-FAPI PET is particularly suited for the diagnosis of colorectal cancer, prostate cancer, and cancers of the breast and head and neck. 68Ga-FAPI PET offers theranostic screening and has the potential for more precise staging and management of patients who have these entities than is possible with 18F-FDG PET.

Footnotes

  • Published online Feb. 8, 2024.

  • © 2024 by the Society of Nuclear Medicine and Molecular Imaging.

REFERENCES

  1. 1.↵
    1. van Tinteren H,
    2. Hoekstra OS,
    3. Smit EF,
    4. et al
    . Effectiveness of positron emission tomography in the preoperative assessment of patients with suspected non-small-cell lung cancer: the PLUS multicentre randomised trial. Lancet. 2002;359:1388–1393.
    OpenUrlCrossRefPubMed
  2. 2.
    1. Ell PJ
    . The contribution of PET/CT to improved patient management. Br J Radiol. 2006;79:32–36.
    OpenUrlAbstract/FREE Full Text
  3. 3.
    1. Choi JY,
    2. Lee KH,
    3. Shim YM,
    4. et al
    . Improved detection of individual nodal involvement in squamous cell carcinoma of the esophagus by FDG PET. J Nucl Med. 2000;41:808–815.
    OpenUrlAbstract/FREE Full Text
  4. 4.
    1. Annovazzi A,
    2. Rea S,
    3. Zoccali C,
    4. et al
    . Diagnostic and clinical impact of 18F-FDG PET/CT in staging and restaging soft-tissue sarcomas of the extremities and trunk: mono-institutional retrospective study of a sarcoma referral center. J Clin Med. 2020;9:2549.
    OpenUrl
  5. 5.
    1. Grimer R,
    2. Judson I,
    3. Peake D,
    4. Seddon B
    . Guidelines for the management of soft tissue sarcomas. Sarcoma. 2010;2010:506182.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Ioannidis JPA,
    2. Lau J
    . 18F-FDG PET for the diagnosis and grading of soft-tissue sarcoma: a meta-analysis. J Nucl Med. 2003;44:717–724.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Erdogan B,
    2. Ao M,
    3. White LM,
    4. et al
    . Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 2017;216:3799–3816.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    1. Chen WT,
    2. Kelly T
    . Seprase complexes in cellular invasiveness. Cancer Metastasis Rev. 2003;22:259–269.
    OpenUrlCrossRefPubMed
  9. 9.
    1. Keane FM,
    2. Nadvi NA,
    3. Yao TW,
    4. Gorrell MD,
    5. Neuropeptide Y
    , B-type natriuretic peptide, substance P and peptide YY are novel substrates of fibroblast activation protein-alpha. FEBS J. 2011;278:1316–1332.
    OpenUrlCrossRefPubMed
  10. 10.
    1. Huang Y,
    2. Wang S,
    3. Kelly T
    . Seprase promotes rapid tumor growth and increased microvessel density in a mouse model of human breast cancer. Cancer Res. 2004;64:2712–2716.
    OpenUrlAbstract/FREE Full Text
  11. 11.
    1. Kelly T
    . Fibroblast activation protein-alpha and dipeptidyl peptidase IV (CD26): cell-surface proteases that activate cell signaling and are potential targets for cancer therapy. Drug Resist Updat. 2005;8:51–58.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Mueller SC,
    2. Ghersi G,
    3. Akiyama SK,
    4. et al
    . A novel protease-docking function of integrin at invadopodia. J Biol Chem. 1999;274:24947–24952.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Lindner T,
    2. Loktev A,
    3. Altmann A,
    4. et al
    . Development of quinoline-based theranostic ligands for the targeting of fibroblast activation protein. J Nucl Med. 2018;59:1415–1422.
    OpenUrlAbstract/FREE Full Text
  14. 14.
    1. Loktev A,
    2. Lindner T,
    3. Mier W,
    4. et al
    . A tumor-imaging method targeting cancer-associated fibroblasts. J Nucl Med. 2018;59:1423–1429.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    1. Giesel FL,
    2. Kratochwil C,
    3. Lindner T,
    4. et al
    . 68Ga-FAPI PET/CT: biodistribution and preliminary dosimetry estimate of 2 DOTA-containing FAP-targeting agents in patients with various cancers. J Nucl Med. 2019;60:386–392.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Baum RP,
    2. Schuchardt C,
    3. Singh A,
    4. et al
    . Feasibility, biodistribution, and preliminary dosimetry in peptide-targeted radionuclide therapy of diverse adenocarcinomas using 177Lu-FAP-2286: first-in-humans results. J Nucl Med. 2022;63:415–423.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    1. Ferdinandus J,
    2. Fragoso Costa P,
    3. Kessler L,
    4. et al
    . Initial clinical experience with 90Y-FAPI-46 radioligand therapy for advanced stage solid tumors: a case series of nine patients. J Nucl Med. 2022;63:727–734.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Kratochwil C,
    2. Flechsig P,
    3. Lindner T,
    4. et al
    . 68Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med. 2019;60:801–805.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. Kessler L,
    2. Ferdinandus J,
    3. Hirmas N,
    4. et al
    . 68Ga-FAPI as a diagnostic tool in sarcoma: data from the 68Ga-FAPI PET prospective observational trial. J Nucl Med. 2022;63:89–95.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Ferdinandus J,
    2. Kessler L,
    3. Hirmas N,
    4. et al
    . Equivalent tumor detection for early and late FAPI-46 PET acquisition. Eur J Nucl Med Mol Imaging. 2021;48:3221–3227.
    OpenUrl
  21. 21.↵
    1. Kessler L,
    2. Ferdinandus J,
    3. Hirmas N,
    4. et al
    . Pitfalls and common findings in 68Ga-FAPI-PET: a pictorial analysis. J Nucl Med. 2022;63:890–896.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Hirmas N,
    2. Hamacher R,
    3. Sraieb M,
    4. et al
    . Fibroblast activation protein positron emission tomography and histopathology in a single-center database of 324 patients and 21 tumor entities. J Nucl Med. 2023;64:711–716.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Harris PA,
    2. Taylor R,
    3. Minor BL,
    4. et al
    .; the REDCap consortium. Building an international community of software platform partners. J Biomed Inform. 2019;95:103208.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Harris PA,
    2. Taylor R,
    3. Thielke R,
    4. Payne J,
    5. Gonzalez N,
    6. Conde JG
    . Research electronic data capture (REDCap): a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377–381.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Lindner T,
    2. Loktev A,
    3. Giesel F,
    4. Kratochwil C,
    5. Altmann A,
    6. Haberkorn U
    . Targeting of activated fibroblasts for imaging and therapy. EJNMMI Radiopharm Chem. 2019;4:16.
    OpenUrl
  26. 26.↵
    1. Loktev A,
    2. Lindner T,
    3. Burger EM,
    4. et al
    . Development of fibroblast activation protein-targeted radiotracers with improved tumor retention. J Nucl Med. 2019;60:1421–1429.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Eisenhauer EA,
    2. Therasse P,
    3. Bogaerts J,
    4. et al
    . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–247.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Gündoğan C,
    2. Güzel Y,
    3. Can C,
    4. Kaplan İ,
    5. Kömek H
    . FAPI-04 uptake in healthy tissues of cancer patients in 68Ga-FAPI-04 PET/CT imaging. Contrast Media Mol Imaging. 2021;2021:9750080.
    OpenUrl
  29. 29.↵
    1. Giesel FL,
    2. Kratochwil C,
    3. Schlittenhardt J,
    4. et al
    . Head-to-head intra-individual comparison of biodistribution and tumor uptake of 68Ga-FAPI and 18F-FDG PET/CT in cancer patients. Eur J Nucl Med Mol Imaging. 2021;48:4377–4385.
    OpenUrl
  30. 30.↵
    1. Roderburg C,
    2. Loosen SH,
    3. Hoyer L,
    4. Luedde T,
    5. Kostev K
    . Prevalence of diabetes mellitus among 80,193 gastrointestinal cancer patients in five European and three Asian countries. J Cancer Res Clin Oncol. 2022;148:1057–1062.
    OpenUrl
  31. 31.↵
    1. Zheng S,
    2. Lin J,
    3. Zhu Y,
    4. et al
    . 68Ga-FAPI versus 18F-FDG PET/CT in evaluating newly diagnosed breast cancer patients: a head-to-head comparative study. Clin Nucl Med. 2023;48:e104–e109.
    OpenUrl
  32. 32.
    1. Kömek H,
    2. Can C,
    3. Güzel Y,
    4. et al
    . 68Ga-FAPI-04 PET/CT, a new step in breast cancer imaging: a comparative pilot study with the 18F-FDG PET/CT. Ann Nucl Med. 2021;35:744–752.
    OpenUrl
  33. 33.↵
    1. Ballal S,
    2. Yadav MP,
    3. Moon ES,
    4. et al
    . Biodistribution, pharmacokinetics, dosimetry of [68Ga]Ga-DOTA.SA.FAPi, and the head-to-head comparison with [18F]F-FDG PET/CT in patients with various cancers. Eur J Nucl Med Mol Imaging. 2021;48:1915–1931.
    OpenUrl
  34. 34.↵
    1. Chen S,
    2. Chen Z,
    3. Zou G,
    4. et al
    . Accurate preoperative staging with [68Ga]Ga-FAPI PET/CT for patients with oral squamous cell carcinoma: a comparison to 2-[18F]FDG PET/CT. Eur Radiol. 2022;32:6070–6079.
    OpenUrl
  35. 35.
    1. Fu H,
    2. Wu J,
    3. Huang J,
    4. et al
    . 68Ga fibroblast activation protein inhibitor PET/CT in the detection of metastatic thyroid cancer: comparison with 18F-FDG PET/CT. Radiology. 2022;304:397–405.
    OpenUrl
  36. 36.↵
    1. Linz C,
    2. Brands RC,
    3. Kertels O,
    4. et al
    . Targeting fibroblast activation protein in newly diagnosed squamous cell carcinoma of the oral cavity: initial experience and comparison to [18F]FDG PET/CT and MRI. Eur J Nucl Med Mol Imaging. 2021;48:3951–3960.
    OpenUrl
  37. 37.↵
    1. Khreish F,
    2. Rosar F,
    3. Kratochwil C,
    4. Giesel FL,
    5. Haberkorn U,
    6. Ezziddin S
    . Positive FAPI-PET/CT in a metastatic castration-resistant prostate cancer patient with PSMA-negative/FDG-positive disease. Eur J Nucl Med Mol Imaging. 2020;47:2040–2041.
    OpenUrl
  38. 38.
    1. Pang Y,
    2. Meng T,
    3. Xu W,
    4. Shang Q,
    5. Chen H
    . 68Ga-FAPI PET/CT detected non-PSMA/FDG-avid primary tumor in de novo metastatic prostate cancer. Clin Nucl Med. 2022;47:1108–1111.
    OpenUrl
  39. 39.↵
    1. Aryana K,
    2. Manafi-Farid R,
    3. Amini H,
    4. Divband G,
    5. Moghadam SZ
    . 68Ga-FAPI-46 PET/CT in a metastatic castration-resistant prostate cancer patient with low PSMA expression. Clin Nucl Med. 2022;47:972–973.
    OpenUrl
  40. 40.↵
    1. Lan L,
    2. Liu H,
    3. Wang Y,
    4. et al
    . The potential utility of [68Ga]Ga-DOTA-FAPI-04 as a novel broad-spectrum oncological and non-oncological imaging agent: comparison with [18F]FDG. Eur J Nucl Med Mol Imaging. 2022;49:963–979.
    OpenUrl
  41. 41.↵
    1. Chen X,
    2. Lin X,
    3. Yuan T,
    4. Wei M,
    5. Wang X
    . Head-to-head comparison between 68Ga-FAPI-04 and [18F]-FDG-PET/CT in lymphomas: a preliminary analysis [abstract]. J Nucl Med. 2022;63(suppl 2):2272.
    OpenUrl
  42. 42.
    1. Elboga U,
    2. Sahin E,
    3. Cayirli YB,
    4. et al
    . Comparison of [68Ga]-FAPI PET/CT and [18F]-FDG PET/CT in multiple myeloma: clinical experience. Tomography. 2022;8:293–302.
    OpenUrl
  43. 43.↵
    1. Jin X,
    2. Wei M,
    3. Wang S,
    4. et al
    . Detecting fibroblast activation proteins in lymphoma using 68Ga-FAPI PET/CT. J Nucl Med. 2022;63:212–217.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    1. Fendler WP,
    2. Bartel T,
    3. Pabst KM,
    4. et al
    . 68Ga-FAPI-46 PET for cancer imaging: a prospective single-arm clinical trial [abstract]. J Clin Oncol. 2023;41(suppl):3064.
    OpenUrl
  45. 45.↵
    1. Lindner T,
    2. Altmann A,
    3. Kramer S,
    4. et al
    . Design and development of 99mTc-labeled FAPI tracers for SPECT imaging and 188Re therapy. J Nucl Med. 2020;61:1507–1513.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    1. Kratochwil C,
    2. Giesel FL,
    3. Rathke H,
    4. et al
    . [153Sm]samarium-labeled FAPI-46 radioligand therapy in a patient with lung metastases of a sarcoma. Eur J Nucl Med Mol Imaging. 2021;48:3011–3013.
    OpenUrl
  47. 47.↵
    1. Kuyumcu S,
    2. Kovan B,
    3. Sanli Y,
    4. et al
    . Safety of fibroblast activation protein-targeted radionuclide therapy by a low-dose dosimetric approach using 177Lu-FAPI04. Clin Nucl Med. 2021;46:641–646.
    OpenUrl
  48. 48.
    1. Assadi M,
    2. Rekabpour SJ,
    3. Jafari E,
    4. et al
    . Feasibility and therapeutic potential of 177Lu-fibroblast activation protein inhibitor-46 for patients with relapsed or refractory cancers: a preliminary study. Clin Nucl Med. 2021;46:e523–e530.
    OpenUrl
  49. 49.↵
    1. Ballal S,
    2. Yadav MP,
    3. Moon ES,
    4. et al
    . Novel fibroblast activation protein inhibitor-based targeted theranostics for radioiodine-refractory differentiated thyroid cancer patients: a pilot study. Thyroid. 2022;32:65–77.
    OpenUrl
  50. 50.↵
    1. Fendler WP,
    2. Pabst KM,
    3. Kessler L,
    4. et al
    . Safety and efficacy of 90Y-FAPI-46 radioligand therapy in patients with advanced sarcoma and other cancer entities. Clin Cancer Res. 2022;28:4346–4353.
    OpenUrl
  51. 51.↵
    1. Narra K,
    2. Mullins SR,
    3. Lee HO,
    4. et al
    . Phase II trial of single agent Val-boroPro (talabostat) inhibiting fibroblast activation protein in patients with metastatic colorectal cancer. Cancer Biol Ther. 2007;6:1691–1699.
    OpenUrlCrossRefPubMed
  52. 52.↵
    1. Bughda R,
    2. Dimou P,
    3. D’Souza RR,
    4. Klampatsa A
    . Fibroblast activation protein (FAP)-targeted CAR-T cells: launching an attack on tumor stroma. ImmunoTargets Ther. 2021;10:313–323.
    OpenUrlCrossRef
  53. 53.↵
    1. Pircher M,
    2. Schuberth P,
    3. Gulati P,
    4. et al
    . FAP-specific re-directed T cells first in-man study in malignant pleural mesothelioma: experience of the first patient treated [abstract]. J Immunother Cancer. 2015;3(suppl 2):P120.
    OpenUrlFREE Full Text
  54. 54.↵
    1. Busek P,
    2. Mateu R,
    3. Zubal M,
    4. Kotackova L,
    5. Sedo A
    . Targeting fibroblast activation protein in cancer: prospects and caveats. Front Biosci (Landmark Ed). 2018;23:1933–1968.
    OpenUrl
  • Received for publication September 8, 2023.
  • Accepted for publication December 20, 2023.
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 65 (3)
Journal of Nuclear Medicine
Vol. 65, Issue 3
March 1, 2024
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Complete Issue (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Journal of Nuclear Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Diagnostic Accuracy of 68Ga-FAPI Versus 18F-FDG PET in Patients with Various Malignancies
(Your Name) has sent you a message from Journal of Nuclear Medicine
(Your Name) thought you would like to see the Journal of Nuclear Medicine web site.
Citation Tools
Diagnostic Accuracy of 68Ga-FAPI Versus 18F-FDG PET in Patients with Various Malignancies
Nader Hirmas, Rainer Hamacher, Miriam Sraieb, Lukas Kessler, Kim M. Pabst, Francesco Barbato, Helena Lanzafame, Stefan Kasper, Michael Nader, Claudia Kesch, Bastian von Tresckow, Hubertus Hautzel, Clemens Aigner, Martin Glas, Martin Stuschke, Sherko Kümmel, Philipp Harter, Celine Lugnier, Waldemar Uhl, Boris Hadaschik, Viktor Grünwald, Jens T. Siveke, Ken Herrmann, Wolfgang P. Fendler
Journal of Nuclear Medicine Mar 2024, 65 (3) 372-378; DOI: 10.2967/jnumed.123.266652

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Diagnostic Accuracy of 68Ga-FAPI Versus 18F-FDG PET in Patients with Various Malignancies
Nader Hirmas, Rainer Hamacher, Miriam Sraieb, Lukas Kessler, Kim M. Pabst, Francesco Barbato, Helena Lanzafame, Stefan Kasper, Michael Nader, Claudia Kesch, Bastian von Tresckow, Hubertus Hautzel, Clemens Aigner, Martin Glas, Martin Stuschke, Sherko Kümmel, Philipp Harter, Celine Lugnier, Waldemar Uhl, Boris Hadaschik, Viktor Grünwald, Jens T. Siveke, Ken Herrmann, Wolfgang P. Fendler
Journal of Nuclear Medicine Mar 2024, 65 (3) 372-378; DOI: 10.2967/jnumed.123.266652
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • Visual Abstract
    • Abstract
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • CONCLUSION
    • DISCLOSURE
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • PDF

Related Articles

  • PubMed
  • Google Scholar

Cited By...

  • Development of FAP-targeted theranostics discovered by next-generation sequencing-augmented mining of a novel immunized VNAR library
  • 1,090 Publications and 5 Years Later: Is FAP-Targeted Theranostics Really Happening?
  • Google Scholar

More in this TOC Section

  • 177Lu-PSMA-617 Consolidation Therapy After Docetaxel in Patients with Synchronous High-Volume Metastatic Hormone-Sensitive Prostate Cancer: A Randomized, Phase 2 Trial
  • Transarterial Radioembolization in the TACOME Trial: Dosimetric Analysis and Clinical Features in Predicting Response and Overall Survival
  • Retreatment of Metastatic Castration-Resistant Prostate Cancer Patients with 223Ra Therapy in Daily Practice
Show more Clinical Investigation

Similar Articles

Keywords

  • FAPI
  • FDG
  • PET
  • oncology
  • theranostic
  • accuracy
SNMMI

© 2025 SNMMI

Powered by HighWire