Skip to main content

Nuclear medicine imaging in non-seminomatous germ cell tumors: lessons learned from the past failures

Abstract

There is an unmet need for a more accurate molecular imaging radiotracer in the field of non-seminomatous germ cell tumors (NSGCT). The clinical problem is that no single imaging modality is able to differentiate teratoma from necrotic tissue in NSGCTs, which the nuclear medicine techniques are no exception. The exponential growth in the list of potentially promising radiotracers may hold promise in the future for imaging of NSGCTs. Here, we have reviewed the past efforts and potential future advances in this field.

Epidemiology

Testicular cancer is the most common malignancy in men aged 15–45 years and accounts for 1-1.8% of all male cancers [1, 2]. Of testicular malignancies, 95% are germ cell tumors (GCTs) [3]. About 50% of patients with GCT present with advanced non-seminomatous germ cell tumors (NSGCT) [4, 5].

The imaging problem in the field of GCT

Following radical orchiectomy and adjuvant cisplatin-based triplet chemotherapy for the treatment of stage IIB-III NSGCT, there is a 30–40% chance of retroperitoneal mass persistence [6,7,8,9]. In 40–51% of these cases the retroperitoneal masses represent necrotic/fibrotic tissues, while 30–47% are teratomas, and the remaining 6–17% of cases are different histopathologies simply grouped as viable GCTs [10,11,12]. Post-chemotherapy retroperitoneal lymph node dissection (PC-RPLND) aims to eradicate all the remaining viable malignant tissue in the advanced NSGCTs, however distinguishing residual viable tumor from post-therapy changes remains a major challenge when deciding if surgery is required [13].

Several studies have shown that not only the viable tumors but also those with teratoma are at increased risk of recurrence [14,15,16,17,18]. For instance, Nestler and colleagues [14] analyzed data from a multi-center cohort of 1204 non-seminomas who underwent PC-RPLND and observed a significantly increased risk of recurrence by five years in the viable GCT/teratoma subgroups compared to patients with only necrosis (81% and 59%, vs. 19%, respectively, p < .001). Moreover, teratomas should be resected due to their resistance to chemoradiation, compressive effect on adjacent organs, and their ability to undergo malignant transformation, especially in the subtype of teratoma with somatic malignancy [11, 19]. Therefore, differentiation of teratomas from necrosis/fibrosis is clinically relevant.

Retroperitoneal teratomas are usually asymptomatic, and tumor markers frequently fall within the normal range, except in cases of mixed GCT or those with mucinous or hepatoid differentiation. Therefore, its detection and follow-up are highly reliant on anatomic imaging [19, 20]. Mature teratomas, also referred to as differentiated teratomas, usually present as low attenuation retroperitoneal masses with less aggressive behavior [21]. There are some conventional imaging features that are useful for the differentiation of mature teratoma from immature or growing teratoma (Table 1). However, irrespective of their subtype, teratomas should be resected as per international guidelines [22, 23].

Table 1 Anatomic features for differentiation and prognostication of mature teratoma versus immature or growing teratoma across a prognosis range from poor to good [20, 22, 109]

Review of the guidelines

Unfortunately, no non-invasive diagnostic modality or validated risk calculator can accurately determine the nature of the residual mass (Table 2) [6, 11, 13, 24,25,26,27,28,29]. Therefore, the EAU guidelines recommends resecting the post-chemotherapy residual mass if > 1 cm in greatest diameter on contrast-enhanced CT (ceCT) whenever feasible [30]. In this context, PC-RPLND serves as both a diagnostic and a therapeutic tool [28]. This approach will over-treat almost half of the patients while leaving 25% risk of teratoma and 5% risk of viable tumor in small sub-centimetric lesions, which have an overall 6–9% risk of relapse and may be captured by subsequent imaging [3, 11, 27]. In this context, surgery is often without oncological benefit [14] and major post-surgical complication rates are non-negligible according to a systematic review [31].

Table 2 Prognosticators of post-chemotherapy retroperitoneal residual mass

Some authors have suggested that the incorporation of non-invasive imaging modalities, such as 18F-FDG PET/CT, into the management algorithm may allow better prediction of viable residual tumors and, thus better risk stratification in this setting [4]. However, the NCCN guideline [32] recommends abdominopelvic ceCT, MRI, and CXR as modalities for imaging first- and second-line chemotherapy patients and routine follow-up cases, which is also consistent with other guidelines (e.g., ESMO, SWENOTECA) [33,34,35]. The NCCN guidelines currently recommend against the routine use of 18F-FDG PET/CT while considering its possible usefulness for surveillance of patients in the post-chemotherapy status [32].

Why 18F-FDG PET/CT was not so successful?

The utility of 18F-FDG PET/CT in patients with NSGCT has been a topic of considerable debate, with views ranging from some to no benefit [36,37,38]. Viable tumors have significantly higher FDG uptake (Fig. 1) as compared to the generally low FDG uptake in necrosis, fibrosis, or teratoma [39]. Also, a negative 18F-FDG PET/CT scan has been linked with increased overall survival [40]. 18F-FDG PET/CT has also been investigated in the context of relapse following definitive NSGCT treatment. It has also been shown that the levels of tumor markers (i.e., LDH, AFP, and hCG) have a significantly positive correlation with the 18F-FDG uptake [41].

Fig. 1
figure 1

A 32-year-old male with non-seminomatous testicular cancer, initially treated with orchidectomy, presented with suspicious para-aortic lymph nodes on CT and underwent an FDG PET scan. The images reveal a subcentimetre left supraclavicular lymph node (SUVmax 3.5; images a-c, crosshairs), bilateral intensely FDG-avid retrocrural lymph nodes (SUVmax 8 on the right and 12 on the left; images d-f, red arrow), and bilateral para-aortic lymphadenopathy extending from the axial level of L1 to L2/L3 on the right (SUVmax 21) and from the axial level of L1/L2 to L2/L3 on the left (SUVmax 25; images g-I). A subsequent biopsy of the left supraclavicular node confirmed metastatic involvement. Retroperitoneal lymph node dissection also confirmed multifocal retroperitoneal nodal metastases

Contrary to the studies mentioned above, in one study, 18F-FDG PET was even inferior to CT for differentiation of necrosis/fibrosis from teratoma [42] (Fig. 2), which is considered the main disadvantage of 18F-FDG PET/CT [43,44,45,46]. Moreover, 18F-FDG PET/CT may falsely show extensive uptake in post-chemotherapy inflammatory changes [47], especially if imaged early post-treatment [12] (Fig. 3). Furthermore, it will miss small (i.e., < 5–10 mm) lesions [48, 49], leading to high relapse rates among the PET-negative patients [50]. Therefore, its application for routine staging of NSGCT is discouraged since it will not have a clear added value to the standard ceCT and will not alter the treatment management [7, 23, 51, 52]. Also, for prediction of response to chemotherapy, 18F-FDG PET/CT has not been shown to be superior to ceCT or serum tumor markers, although being a strong predictor of pathologic viable disease [37, 53, 54].

Fig. 2
figure 2

A 30-year-old male with a non-seminoma germ cell tumor, previously treated with a left orchidectomy. The PET scan demonstrates multiple low-density nodal lesions in the retroperitoneum, including at the aortocaval and left para-aortic stations (red arrows), with no increased FDG uptake. The patient subsequently underwent retroperitoneal lymph node dissection, which revealed multiple nodal metastases

Fig. 3
figure 3

A 33-year-old male with a history of non-seminoma germ cell tumor, previously treated with left orchidectomy, chemotherapy, and retroperitoneal lymph node dissection for bulky para-aortic lymph node metastases, presents for a progress assessment. The PET scan (Image A) demonstrates mild foci of uptake around the aortocaval (SUV max 3.9) and left para-aortic (SUV max 3.3) regions at the level of L3, adjacent to surgical clips (red arrows, a-c), which were reported as indeterminate (either post-surgical inflammatory changes or residual disease). Twelve months later, a follow-up PET scan (Image B) showed an interval reduction in the intensity of retroperitoneal foci of uptake (d-f), consistent with resolving post-operative inflammatory changes

In one study in NSGCT patients examined 85 residual lesions, with 32 (38%) showing increased tracer uptake, resulting in a sensitivity of 59%, specificity of 92%, NPV of 62%, and PPV of 91% [53]. Therefore, 18F-FDG PET positivity may be clinically relevant in evaluation of residual masses. In another study, quantitative 18F-FDG PET analyses indicated significant differences between mature teratoma and necrosis or scar tissue, supporting its use for evaluating residual lesions post-chemotherapy [55]. Additionally, another study found that an SUV greater than 5 is more likely to be linked to viable GCT than necrosis, fibrosis, or mature teratoma [36]. However, the overall diagnostic benefit of 18F-FDG PET/CT over traditional markers and CT scans for suspected NSGCT recurrence remains uncertain (Fig. 4), though patients with elevated tumor markers with equivocal CT findings might benefit from 18F-FDG PET/CT [56, 57].

Fig. 4
figure 4

A 23-year-old male with non-seminomatous testicular cancer, treated with orchidectomy followed by chemotherapy, presented with suspicious retroperitoneal lymphadenopathy on CT and underwent an FDG PET scan (Image A). The axial (a, d), coronal (b, e), and sagittal (c, f) views show enlarged hypoattenuating retroperitoneal lesions with no metabolic activity (red arrow). Serial CT scans demonstrated ongoing enlargement of these lesions. Ten months later, a follow-up PET scan (Image B) revealed further enlargement of the hypoattenuating retroperitoneal lesions with interval development of peripheral metabolic activity (images g-l, green arow), highly suggestive of nodal metastases. Subsequent nodal dissection confirmed the presence of nodal metastases on histopathology

In conclusion, in the era of “forget the PET” approach, 18F-FDG PET/CT may infrequently be requested for rising tumor markers with normal ceCT and those with equivocal ceCT findings [58, 59]. Kinetic analysis may improve its diagnostic performance but is not performed in routine clinical practice, and has not been fully validated in large prospective studies [55].

Teratoma imaging with other radiopharmaceuticals: a land of failures

The multi-layered embryologic origin of teratoma sometimes contains immature neuro-ectodermal elements, which may eventually trap radioiodine or radiotracers targeting the somatostatin-receptors [60, 61]. Multiple research groups have explored the added value of other PET radiopharmaceuticals, most of which were not very successful (Table 3). Perhaps the best one was imaging with radiopharmaceuticals targeting integrins. Yet, none of these radiotracers find their way into clinical practice. A more in-depth review of the experience gained by each imaging modality is discussed as follows.

Table 3 Teratoma imaging with non-18F-FDG radiotracers *1

PET tracers

11C-tyrosine PET

A study showed that 11C-tyrosine is not suited to visualize the apparently slowly proliferating NSGCT or to determine the nature of a residual mass after chemotherapy [62].

18F- fluorothymidine (18F-FLT)

In a small series of 11 patients (10 NSGCT, 1 seminoma) with metastatic NSGCTs, Pfannenberg et al. compared the diagnostic value of 18F-FLT, which measures tumor cell proliferation, with 18F-FDG PET/CT. Despite the lower incidence of false-positive results with 18F-FLT PET than with 18F-FDG PET, the low negative predictive value of 18F-FDG PET could not be improved by the application of the 18F-FLT (60% and 50%, respectively). Therefore, PET-negative residual masses after chemotherapy of metastatic NSGCT still require resection. The low sensitivity of 18F-FLT PET/CT for the detection of viable residual tumors in this study may be related to the lower tissue uptake of 18F-FLT than of 18F-FDG in GCTs. Positive results on 18F-FDG PET after chemotherapy correlated strongly with the presence of viable tumors. For prediction of response after completion of chemotherapy, the final PET/CT scan, whether performed using 18F-FDG or using 18F-FLT, cannot be replaced by early response evaluation [63].

18F-fluciclovine

The potential use of 18F-fluciclovine for molecular imaging of NSGCTs was evaluated in a small prospective study, which revealed poor sensitivity and specificity in detecting teratoma from fibrosis/necrosis in patients with residual masses undergoing PC-RPLND. Half of the negative 18F-fluciclovine PET/CT cases were found to have residual disease/teratoma following surgery. The low utility of 18F-fluciclovine PET/CT in guiding the management of NSGCT post-chemotherapy was evident, with sensitivity and specificity rates at 29% and 33%, respectively [64].

Prostate-Specific Membrane Antigen (PSMA)

Prior human tissue studies have shown the expression of PSMA in some cases of NSGCT [65]. A case report of metastatic mixed (immature teratoma and yolk sac carcinoma) testicular GCT with acceptable tumor-to-background ratio was treated with the therapeutic counterpart of PSMA, namely 177Lu-PSMA, was not successful [66].

Integrins

Since their discovery in 2006, induced pluripotent stem cells (iPSCs) have gained increasing interest in tissue regeneration and transplantation therapies. However, teratoma formation after iPSC transplantation is one of the most serious drawbacks of this procedure. In a study, it was investigated whether human iPSC-derived teratomas could be detected by an integrin-targeting agent, 99mTc-PEG4-E[PEG4-c(RGDfK)]2 (99mTc-3PRGD2). Gamma camera imaging with 99mTc-3PRGD2 may be a promising approach for the non-invasive monitoring of tumorigenicity after human iPSCs transplantation [67]. Unfortunately, these preclinical observations were never explored on human subjects.

Non-PET Tracers

SPECT/CT’s spatial resolution is challenged over PET/CT, particularly in small lesions that are not always metabolically active, including NSGCTs [68]. This gap can be bridged with advanced quantification and reconstruction techniques and multi-pinhole collimators focusing on gamma rays [69]. PET facilities are preferred over SPECT in regions where both options are available; however, SPECT remains cost-effective in specific clinical applications and resource-limited countries [70, 71]. The results of teratoma imaging utilizing SPECT are unsatisfactory [71,72,73,74,75,76,77,78,79,80]. Small case series and case reports reported non-visualization of teratocarcinoma using Tl-201 [71] or incidental detection of NSGCTs using 99mTc-MDP due to ossification and cartilage tissue in teratoma [72], and 99mTc-pertechnetate due to increased flow in tumoral tissue [73].

Gallium-67

Traditionally, 67Ga scintigraphy was considered valuable in assessing the intra-abdominal spread of malignant tumors of the testes. However, it appeared that metastatic tumors of the embryonal-cell and seminoma type, compared to teratoma, are more readily detectable by gallium-67 scanning [72]. Although its application for imaging of NSGCT were disappointing and discontinued [73, 74], its utility for staging in seminoma also became obsolete after the introduction of 18F-FDG PET/CT [75].

Radiolabeled antibody

Radioimmunodetection captures tumor-specific or tumor-associated markers by preferentially accumulating tumor-specific antisera in tumoral tissues. Murine teratocarcinomas were localized using external gamma-ray scintigraphy with 131I-labeled monoclonal antibodies. By removing background radioactivity from the control monoclonal antibody 123 of the same immunoglobulin class, detection was enhanced [76]. Javadpour et al. utilized 131I-labeled antibodies targeting tumor-associated antigens in testicular cancer to identify occult disease [77]. The limited sensitivity of this approach in identifying lesions under 2 × 2 cm and interference from background radioactivity limit its practical applicability. Epenetos et al. investigated placental alkaline phosphatase-targeting indium-111 monoclonal antibodies. Their study showed improved ovarian, cervical, and testicular cancer diagnosis. However, there are still ongoing issues regarding the pharmacokinetics and immunogenicity of antibodies, despite the positive outcomes shown [78].

Potential of imaging teratoma with novel radiotracers

Fibroblast activation protein inhibitor (FAPi)

FAP-targeting PET tracers have been extensively studied in both malignant and non-malignant entities [79, 80]. FAPi ligands may have a complementary role in detecting metastatic lymph nodes, especially if coupled with 18F-FDG PET imaging in various cancers [81]. In addition, FAPi PET imaging has been shown to be able to detect fibrotic tissue in various scenarios (e.g., post-chemotherapy fibrosis in GI malignancies, idiopathic retroperitoneal fibrosis, and various non-malignant fibrotic pathologies) [79, 82]. However, to the best of our knowledge, till today, there has been no published comprehensive paper on FAPi PET tracers in NSGCTs.

Regarding FAP application in detecting teratoma, there is insufficient data in the literature. Xi et al., in a study conducted to compare 68Ga-FAPi-04 PET/MR and 18F-FDG PET/CT in ovarian tumors in 2023, and reported that of all the included cases, two patients had teratoma (one considered benign pathology and the latter borderline) [83]. In a study comparing 68Ga-FAP-2286 PET/CT head-to-head with 18F-FDG PET/CT in various malignancies, one case of metastatic yolk sac germ cell tumor was evaluated and showed better LN detection performance of FAPi ligand over 18F-FDG with a better target-to-background ratio (separate values are not reported). Visually, in comparison to [68Ga]Ga-FAP-46, this new FAP ligand seems to have a higher uptake [84].

There are two additional papers that each report one case of GCT imaged with FAPi radiotracers. Dai et al. [85]. reported a rare case of extragonadal yolk-sac tumor in which 68Ga-FAPI PET/MR outperformed 18F-FDG PET/CT in the detection of the cranial lesion. The other case report looked at 68Ga-FAPi-04 PET/CT and 18F-FDG PET/CT in a person who had mixed testicular GCT that was 65% post-pubertal teratoma, 25% yolk sac and 10% seminoma. The retroperitoneal and lung nodules showed a slight uptake of FDG. Meanwhile, FAPi imaging revealed a mild-moderate uptake in the affected lesions (SUVmax of 3.9) [1].

CXCR4

CXCR4 is a seven transmembrane domain G protein-coupled receptor (GPCR) that contributes to chemotaxis, invasion, angiogenesis, aggressiveness, tumor progression, proliferation, and metastasis [86]. The CXCR4 ligand is frequently overexpressed in various types of cancer [87]. The CXCL12/CXCR4 pathway has a confirmed and significant role in the adult human testis microenvironment and is also expressed in gonadal and extragonadal GCTs [88, 89]. Yet, in all the publications concerning cellular studies, there is no clinical data to support this hypothesis.

Ghrelin

Testicular tumors differentially express the Ghrelin receptor, a GPCR involved in growth hormone secretion and food intake. In order to enhance in vivo stability and incorporate the Fluorine-18 isotope for PET imaging, the ghrelin ligand has recently undergone some modifications. This novel PET agent has been shown to have a high affinity for the ghrelin receptor in biochemical and preclinical studies [90, 91]. For now, there is no available clinical (in humans) data regarding Ghrelin receptor imaging in NSGCT.

Other solutions beyond nuclear medicine approaches

Magnetic Resonance Imaging

Advancements in the field of MRI in patients with testicular cancer are threefold. First, follow-up whole-body MRI may be employed in the future in lieu of ceCT due to concerns regarding radiation exposure and in the light of positive non-inferiority trials recently published in this regard [92, 93]. Second, new MRI sequences, namely T1-Dixon and T2-BLADE, have been shown to propose better performance in detecting retroperitoneal metastasis and were better than DWI-MRI in a prospective study [94]. Third, lymphotropic nanoparticle MRI (LNMRI) utilizes nanoparticles that aggregate with a mixed signal within pathological nodal tissue [95]. Its use is superior to conventional MRI, according to meta-analyses, in terms of both sensitivity (88% vs. 63%) and specificity (96% vs. 95%) [96, 97]. Harisinghani et al. [97] conducted a pilot trial of LNMRI to detect occult metastases in 18 men with testicular cancer. LNMRI had improved sensitivity (88% vs. 71%) and specificity (92% vs. 68%) compared to MRI or CT size criteria among these patients. Likewise, LNMRI was 100% sensitive in detecting positive lymph nodes less than 10 mm, that would not have been considered suspicious on conventional imaging. However, the lengthy duration (24–36 h) between nanoparticle injection and MRI, along with the requirement for an experienced radiologist to accurately interpret the images, restricts its adoption [54].

The potential role of radiomics

Literature regarding the added value of radiomics to differentiate necrosis/fibrosis from teratoma is emerging, with some studies showing encouraging results [98, 99]. As of now, these studies are yet insufficient to precisely select patients for PC-RPLND to prevent over-treatment [100, 101], inconclusive [99,100,101,102,103] and sometimes controversial [98, 100, 104,105,106]. Moreover, non-automated approaches for delineation of the regions of interest are time consuming and not repeatable, limiting its practicality in daily practice [107].

microRNAs: a potential target for imaging?

A recent study used molecular analysis to explore a non-imaging method to differentiate between teratoma, viable GCT, and necrosis post-chemotherapy [108]. This approach identified AGR2 and KRT19 as key proteins significantly overexpressed in teratoma compared to necrosis at both microRNA and protein levels.

This approach involved classifying 48 patients into three groups: those with teratoma, those with viable GCT, and those with necrosis. Using a microdissection technique, they precisely isolated representative areas of each tissue type within the lymph nodes [108].

From a nuclear medicine perspective, if these proteins are tagged with a PET tracer, it would shed light on the precise diagnosis in this gray area.

Conclusion

Imaging of NSGCT remains challenging, and while 18F-FDG PET imaging has limitations, in a few selected scenarios is still able to contribute to clinical management decisions. The experiences with non-18F-FDG radiotracers have not yet identified a compelling radiotracer for use in this clinical scenario. For now, the complementary benefits of different imaging techniques could be a reasonable approach. The introduction of miRNAs is speculated to revolutionize the field, which are great candidates for future targets to be radiolabeled for imaging NSGCT. The evolving role of radiomics, which remains inconclusive in the field of NSGCT, is still in its infancy but may eventually become a part of routine practice.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ceCT:

contrast-enhanced CT

FAPi:

Fibroblast activation protein inhibitor

GPCR:

G protein-coupled receptor

iPSCs:

induced pluripotent stem cells

LNMRI:

lymphotropic nanoparticle MRI

NSGCT:

non-seminomatous germ cell tumors

PC-RPLND:

Post-chemotherapy retroperitoneal lymph node dissection

PSMA:

Prostate-Specific Membrane Antigen

RAID:

radioimmunodetection

References

  1. Kaplan İ, Can C, Güzel Y, Alabalik U, Kömek H. 68 GA-FAPI-04 PET/CT Versus 18 F-FDG PET/CT in imaging of malignant mixed germ cell testicular tumor. Clin Nucl Med. 2023;48(4):e195–7.

    Article  PubMed  Google Scholar 

  2. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48.

    Article  PubMed  Google Scholar 

  3. Nallu A, Mannuel HD, Hussain A. Testicular germ cell tumors: biology and clinical update. Curr Opin Oncol. 2013;25(3):266–72.

    Article  PubMed  CAS  Google Scholar 

  4. Daneshmand S, Albers P, Fosså SD, Heidenreich A, Kollmannsberger C, Krege S, et al. Contemporary management of postchemotherapy testis cancer. Eur Urol. 2012;62(5):867–76.

    Article  PubMed  Google Scholar 

  5. Nauman M, Leslie SW. Nonseminomatous Testicular Tumors. StatPearls. Treasure Island (FL): StatPearls Publishing Copyright © 2024. StatPearls Publishing LLC.; 2024.

  6. Masterson TA, Tagawa ST. A 25-year review of advances in testicular cancer: Perspectives on evaluation, treatment, and future directions/challenges. Urol Oncol. 2021;39(9):561–8.

    Article  PubMed  Google Scholar 

  7. Giannopoulou CD. PET/CT Findings in Testicular Cancer. In: Andreou JA, Kosmidis PA, Gouliamos AD, editors. Artificial Intelligence in PET/CT Oncologic Imaging. Cham: Springer International Publishing; 2022. pp. 93–8.

    Chapter  Google Scholar 

  8. Wakileh GA, Ruf C, Heidenreich A, Dieckmann KP, Lisson C, Prasad V, et al. Contemporary options and future perspectives: three examples highlighting the challenges in testicular cancer imaging. World J Urol. 2022;40(2):307–15.

    Article  PubMed  CAS  Google Scholar 

  9. Oldenburg J, Berney DM, Bokemeyer C, Climent MA, Daugaard G, Gietema JA, et al. Testicular seminoma and non-seminoma: ESMO-EURACAN Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33(4):362–75.

    Article  PubMed  CAS  Google Scholar 

  10. Mano R, Becerra MF, Carver BS, Bosl GJ, Motzer RJ, Bajorin DF, et al. Clinical Outcome of Patients with Fibrosis/Necrosis at Post-Chemotherapy Retroperitoneal Lymph Node Dissection for Advanced Germ Cell Tumors. J Urol. 2017;197(2):391–7.

    Article  PubMed  Google Scholar 

  11. Dusaud M, Malavaud B, Bayoud Y, Sebe P, Hoepffner JL, Salomon L, et al. Post-chemotherapy retroperitoneal teratoma in nonseminomatous germ cell tumors: Do predictive factors exist? Results from a national multicenter study. J Surg Oncol. 2016;114(8):992–6.

    Article  PubMed  Google Scholar 

  12. Santis MD, Maj-Hes A, Bachner M. Positron Emission Tomography (PET) in Germ Cell Tumors (GCT). In: de la Rosette JJMCH, Manyak MJ, Harisinghani MG, Wijkstra H, editors. Imaging in Oncological Urology. London: Springer London; 2009. pp. 305–13.

    Chapter  Google Scholar 

  13. Conduit C, Hong W, Martin F, Thomas B, Lawrentschuk N, Goad J, et al. A meta-analysis of clinicopathologic features that predict necrosis or fibrosis at post-chemotherapy retroperitoneal lymph node dissection in individuals receiving treatment for non-seminoma germ cell tumours. Front Oncol. 2022;12:931509.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Nestler T, Paffenholz P, Pfister D, Schoch J, Nini A, Hiester A, et al. Adjunctive Surgery Is Often Without Oncological Benefit at Time of Postchemotherapy Retroperitoneal Lymph Node Dissection. J Urol. 2024;211(3):426–35.

    Article  PubMed  Google Scholar 

  15. Cotner CE, Hilton S, Mamtani R, Guzzo T, Vaughn DJ. Surveillance of postchemotherapy subcentimeter residual retroperitoneal mass in metastatic nonseminomatous germ cell tumor: Does how you measure matter? Urol Oncol. 2021;39(2):136. .e11-.e17.

    Article  Google Scholar 

  16. Antonelli L, Ardizzone D, Tachibana I, Adra N, Cary C, Hugar L, et al. Risk Factors for Relapse in Nonseminomatous Testicular Cancer After Postchemotherapy Retroperitoneal Lymph Node Dissection With Viable Residual Cancer. J Clin Oncol. 2023;41(34):5296–305.

    Article  PubMed  CAS  Google Scholar 

  17. Gilligan TD. Resection of Residual Masses After Chemotherapy for Metastatic Nonseminomatous Germ Cell Tumors in Adolescents and Adults. J Clin Oncol. 2023;41(23):3899–904.

    Article  PubMed  CAS  Google Scholar 

  18. Ravi P, Gray KP, O’Donnell EK, Sweeney CJ. A meta-analysis of patient outcomes with subcentimeter disease after chemotherapy for metastatic non-seminomatous germ cell tumor. Ann Oncol. 2014;25(2):331–8.

    Article  PubMed  CAS  Google Scholar 

  19. McAlpine K, Clark R, Spiess PE, Necchi A, Gage K, Hamilton RJ. The Importance of Repeat Imaging Prior to Treatment Decision-making in Testicular Cancer: Commentary From the Inaugural Global Society of Rare Genitourinary Tumors Summit. Clin Genitourin Cancer. 2023;21(3):418. .e1-.e6.

    Article  Google Scholar 

  20. Revels JW, Wang SS, Gangadhar K, Ali A, Ali AA, Lee JH. Multimodality Radiological Pictorial Review of Testicular Carcinoma: From Initial Staging to Restaging. Res Rep Urol. 2020;12:599–613.

    PubMed  PubMed Central  Google Scholar 

  21. Yacoub JH, Oto A, Allen BC, Coakley FV, Friedman B, Hartman MS, et al. ACR Appropriateness Criteria Staging of Testicular Malignancy. J Am Coll Radiol. 2016;13(10):1203–9.

    Article  PubMed  Google Scholar 

  22. Jiao H, Qiu Y, Huang W, Zhang Y, Chen Z, Wang A, Kang L. Alpha-fetoprotein-elevated postpubertal testicular teratoma with retroperitoneal metastasis on (18)F-FDG PET/CT: case report and literature review. Front Med (Lausanne). 2023;10:1269587.

    Article  PubMed  Google Scholar 

  23. Schieda N, Oto A, Allen BC, Akin O, Barker SJ, Fulgham PF, et al. ACR Appropriateness Criteria® Staging and Surveillance of Testicular Cancer: 2021 Update. J Am Coll Radiol. 2022;19(5s):S194–207.

    Article  PubMed  Google Scholar 

  24. Malik K, Raja A, Radhakrishnan V, Kathiresan N. A retrospective analysis of patients undergoing postchemotherapy retroperitoneal lymph node dissection and metastasectomy in advanced nonseminomatous germ cell tumors. Indian J Urol. 2020;36(2):112–6.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Murez T, Fléchon A, Branger N, Savoie PH, Rocher L, Camparo P, et al. French AFU Cancer Committee Guidelines - Update 2022–2024: testicular germ cell cancer. Progrès en Urologie. 2022;32(15):1066–101.

    Article  PubMed  CAS  Google Scholar 

  26. Steiner H, Berg B, Stöhr B, Fritzer A, Ramoner R, Aigner F et al. Prediction of Retroperitoneal Histology in Metastatic Nonseminomatous Testicular Cancer Patients after Chemotherapy Based on Clinical and Radiological Parameters. Curr Urol. 2010;4(3).

  27. Steyerberg EW, Keizer HJ, Habbema JD. Prediction models for the histology of residual masses after chemotherapy for metastatic testicular cancer. ReHiT Study Group. Int J Cancer. 1999;83(6):856–9.

    Article  PubMed  CAS  Google Scholar 

  28. Aparicio J. On behalf of the Spanish Germ Cell Cancer G. Positron emission tomography (PET) is not indicated in the postchemotherapy evaluation of advanced non-seminomatous testicular germ cell tumors. Clin Transl Oncol. 2014;16(5):509–10.

    Article  PubMed  Google Scholar 

  29. Wood MJ, Tirumani SH, Sweeney C, Ramaiya NH, Howard SA. Approach to risk stratification in testicular germ cell tumors: a primer for radiologists. Abdom Imaging. 2015;40(6):1871–86.

    Article  PubMed  Google Scholar 

  30. Patrikidou A, Cazzaniga W, Berney D, Boormans J, de Angst I, Di Nardo D, et al. European Association of Urology Guidelines on Testicular Cancer: 2023 Update. Eur Urol. 2023;84(3):289–301.

    Article  PubMed  Google Scholar 

  31. Rosenvilde JJ, Pedersen GL, Bandak M, Lauritsen J, Kreiberg M, Wagner T, et al. Oncological outcome and complications of post-chemotherapy retroperitoneal surgery in non-seminomatous germ cell tumours - a systematic review. Acta Oncol. 2021;60(6):695–703.

    Article  PubMed  CAS  Google Scholar 

  32. Timothy Gilligan DWL, Adra N, Aggarwal R, Bagrodia A, Costa D, Drakaki A, Emamekhoo H, Evans C, Feldman DR, Geynisman DM, Graham L, Humphrey P, Levine EG. Amy Luckenbaugh, Benjamin Maughan, Bradley McGregor, Paul Monk, Joel Picus, Soroush Rais-Bahrami, Zachery Reichert, Jean-Claude Rwigema, Philip Saylor, Ankeet Shah, Sumit Shah, Nirmish Singla, Kanishka Sircar, Benjamin A. Teply, David VanderWeele, David Vaughn, Kosj Yamoah, Ali Zhumkhawala. Testicular Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. National Comprehensive Cancer Network. 2024.

  33. Tandstad T, Ståhl O, Håkansson U, Wahlqvist R, Klepp O, Cavallin-Ståhl E, Cohn-Cedermark G. The SWENOTECA group: a good example of continuous binational and multidisciplinary collaboration for patients with testicular cancer in Sweden and Norway. Scandinavian J Urol. 2016;50(1):9–13.

    Article  Google Scholar 

  34. Ortiz AFH, Beaujon LJF, Villamizar SYG, López FFF. Magnetic resonance versus computed tomography for the detection of retroperitoneal lymph node metastasis due to testicular cancer: a systematic literature review. Eur J Radiol Open. 2021;8:100372.

    Article  Google Scholar 

  35. Honecker F, Aparicio J, Berney D, Beyer J, Bokemeyer C, Cathomas R, et al. ESMO Consensus Conference on testicular germ cell cancer: diagnosis, treatment and follow-up. Ann Oncol. 2018;29(8):1658–86.

    Article  PubMed  CAS  Google Scholar 

  36. Lassen U, Daugaard G, Eigtved A, Højgaard L, Damgaard K, Rørth M. Whole-body FDG-PET in patients with stage I non-seminomatous germ cell tumours. Eur J Nucl Med Mol Imaging. 2003;30:396–402.

    Article  PubMed  CAS  Google Scholar 

  37. Oechsle K, Hartmann M, Brenner W, Venz S, Weissbach L, Franzius C, et al. [18F] Fluorodeoxyglucose positron emission tomography in nonseminomatous germ cell tumors after chemotherapy: the German multicenter positron emission tomography study group. J Clin Oncol. 2008;26(36):5930–5.

    Article  PubMed  Google Scholar 

  38. Makovník M, Rejleková K, Uhrin I, Mego M, Chovanec M. Intricacies of radiographic assessment in testicular germ cell tumors. Front Oncol. 2021;10:587523.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Quak E, Kovacs I, Oyen WJ, Van der Graaf WT. FDG-PET/CT in a patient with poor-risk non-seminoma testis with mature teratoma and secondary gliosarcoma: multimodality imaging for guiding multimodality treatment. Nuclear Med Mol Imaging. 2015;49:237–40.

    Article  Google Scholar 

  40. Alongi P, Evangelista L, Caobelli F, Spallino M, Gianolli L, Midiri M, Picchio M. Diagnostic and prognostic value of 18F-FDG PET/CT in recurrent germinal tumor carcinoma. Eur J Nucl Med Mol Imaging. 2018;45:85–94.

    Article  PubMed  CAS  Google Scholar 

  41. Aydos U, Tahtaci G, Akdemir ÜÖ, Özet A. 18F-FDG PET/CT for primary staging of patients with testicular germ cell tumors: the predictors of 18F-FDG PET positivity and prognostic value of PET derived metabolic parameters. Nucl Med Commun. 2020;41(11):1199–209.

    Article  PubMed  CAS  Google Scholar 

  42. Spermon JR, De Geus-Oei LF, Kiemeney LA, Witjes JA, Oyen WJ. The role of (18)fluoro-2-deoxyglucose positron emission tomography in initial staging and re-staging after chemotherapy for testicular germ cell tumours. BJU Int. 2002;89(6):549–56.

    Article  PubMed  CAS  Google Scholar 

  43. Kollmannsberger C, Oechsle K, Dohmen BM, Pfannenberg A, Bares R, Claussen CD, et al. Prospective comparison of [18F] fluorodeoxyglucose positron emission tomography with conventional assessment by computed tomography scans and serum tumor markers for the evaluation of residual masses in patients with nonseminomatous germ cell carcinoma. Cancer: Interdisciplinary Int J Am Cancer Soc. 2002;94(9):2353–62.

    Article  Google Scholar 

  44. Karapetis C, Strickland A, Yip D, Steer C, Harper P. Use of fluorodeoxyglucose positron emission tomography scans in patients with advanced germ cell tumour following chemotherapy: single-centre experience with long‐term follow up. Intern Med J. 2003;33(9–10):427–35.

    Article  PubMed  CAS  Google Scholar 

  45. Pfannenberg AC, Oechsle K, Bokemeyer C, Kollmannsberger C, Dohmen BM, Bares R, et al. The role of [18 F] FDG-PET, CT/MRI and tumor marker kinetics in the evaluation of postchemotherapy residual masses in metastatic germ cell tumors—prospects for management. World J Urol. 2004;22:132–9.

    Article  PubMed  CAS  Google Scholar 

  46. Putra LJ, Lawrentschuk N, Ballok Z, Hannah A, Poon A, Tauro A, et al. 18F-fluorodeoxyglucose positron emission tomography in evaluation of germ cell tumor after chemotherapy. Urology. 2004;64(6):1202–7.

    Article  Google Scholar 

  47. Tsatalpas P, Beuthien-Baumann B, Kropp J, Manseck A, Tiepolt C, Hakenberg OW, et al. Diagnostic value of 18F-FDG positron emission tomography for detection and treatment control of malignant germ cell tumors. Urol Int. 2002;68(3):157–63.

    Article  PubMed  Google Scholar 

  48. De Santis M, Pont J. The role of positron emission tomography in germ cell cancer. World J Urol. 2004;22(1):41–6.

    Article  PubMed  Google Scholar 

  49. Ambrosini V, Zucchini G, Nicolini S, Berselli A, Nanni C, Allegri V, et al. 18F-FDG PET/CT impact on testicular tumours clinical management. Eur J Nucl Med Mol Imaging. 2014;41(4):668–73.

    Article  PubMed  CAS  Google Scholar 

  50. Huddart RA, O’Doherty MJ, Padhani A, Rustin GJ, Mead GM, Joffe JK, et al. 18fluorodeoxyglucose positron emission tomography in the prediction of relapse in patients with high-risk, clinical stage I nonseminomatous germ cell tumors: preliminary report of MRC Trial TE22–the NCRI Testis Tumour Clinical Study Group. J Clin Oncol. 2007;25(21):3090–5.

    Article  PubMed  Google Scholar 

  51. de Wit M, Brenner W, Hartmann M, Kotzerke J, Hellwig D, Lehmann J, et al. [18F]-FDG-PET in clinical stage I/II non-seminomatous germ cell tumours: results of the German multicentre trial. Ann Oncol. 2008;19(9):1619–23.

    Article  PubMed  Google Scholar 

  52. Chen J, Daneshmand S. Modern Management of Testicular Cancer. In: Daneshmand S, Chan KG, editors. Genitourinary Cancers. Cham: Springer International Publishing; 2018. pp. 273–308.

    Chapter  Google Scholar 

  53. Kollmannsberger C, Oechsle K, Dohmen BM, Pfannenberg A, Bares R, Claussen CD, et al. Prospective comparison of [18F]fluorodeoxyglucose positron emission tomography with conventional assessment by computed tomography scans and serum tumor markers for the evaluation of residual masses in patients with nonseminomatous germ cell carcinoma. Cancer. 2002;94(9):2353–62.

    Article  PubMed  Google Scholar 

  54. Joice GA, Rowe SP, Gorin MA, Pierorazio PM. Molecular imaging for evaluation of viable testicular cancer nodal metastases. Curr Urol Rep. 2018;19:1–7.

    Article  Google Scholar 

  55. Sugawara Y, Zasadny KR, Grossman HB, Francis IR, Clarke MF, Wahl RL. Germ Cell Tumor: Differentiation of Viable Tumor, Mature Teratoma, and Necrotic Tissue with FDG PET and Kinetic Modeling. Radiology. 1999;211(1):249–56.

    Article  PubMed  CAS  Google Scholar 

  56. Sharma P, Jain T, Parida G, Karunanithi S, Patel C, Sharma A, et al. Diagnostic accuracy of integrated 18F-FDG PET/CT for restaging patients with malignant germ cell tumours. Br J Radiol. 2014;87(1040):20140263.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Buchler T, Simonova K, Fencl P, Jarkovsky J, Abrahamova J. Clinical outcomes of patients with nonseminomatous germ cell tumours and negative postchemotherapy positron emission tomography. Cancer Invest. 2012;30(6):487–92.

    Article  PubMed  Google Scholar 

  58. Cook GJ, Sohaib A, Huddart RA, Dearnaley DP, Horwich A, Chua S. The role of 18F-FDG PET/CT in the management of testicular cancers. Nucl Med Commun. 2015;36(7):702–8.

    Article  PubMed  CAS  Google Scholar 

  59. Feldman DR. State-of-the-Art Management of Germ Cell Tumors. Am Soc Clin Oncol Educ Book. 2018;38:319–23.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Cabral FC, Krajewski KM, Rosenthal MH, Hirsch MS, Howard SA. Teratoma with malignant transformation: report of three cases and review of the literature. Clin Imaging. 2014;38(5):589–93.

    Article  PubMed  Google Scholar 

  61. Yoshida M, Tanaka M, Gomi K, Ohama Y, Kigasawa H, Iwanaka T, Tanaka Y. Malignant steroidogenic tumor arising from sacrococcygeal mature teratoma. Hum Pathol. 2011;42(10):1568–72.

    Article  PubMed  CAS  Google Scholar 

  62. Kole AC, Hoekstra HJ, Sleijfer DT, Nieweg OE, Koops HS, Vaalburg W. L-[1-carbon-11] tyrosine imaging of metastatic testicular nonseminoma germ-cell tumors. J Nucl Med. 1998;39(6):1027–9.

    PubMed  CAS  Google Scholar 

  63. Pfannenberg C, Aschoff P, Dittmann H, Mayer F, Reischl G, von Weyhern C, et al. PET/CT with 18F-FLT: does it improve the therapeutic management of metastatic germ cell tumors? J Nucl Med. 2010;51(6):845–53.

    Article  PubMed  Google Scholar 

  64. Woldu SL, Meng X, Wong D, Baky F, Margulis V, Xi Y, et al. Performance characteristics of 18F-fluciclovine positron emission tomography/computed tomography prior to retroperitoneal lymph node dissection. Can Urol Association J. 2022;16(3):E167.

    Google Scholar 

  65. Kinoshita Y, Kuratsukuri K, Landas S, Imaida K, Rovito PM, Wang CY, Haas GP. Expression of prostate-specific membrane antigen in normal and malignant human tissues. World J Surg. 2006;30:628–36.

    Article  PubMed  Google Scholar 

  66. Simsek DH, Civan C, Ekenel M, Kuyumcu S, Sanli Y. 177Lu-PSMA therapy for metastatic testicular mixed germ cell tumor. Clin Nucl Med. 2021;46(5):415–8.

    Article  Google Scholar 

  67. Li Y, Liu Z, Dong C, He P, Liu X, Zhu Z, et al. Noninvasive Detection of Human-Induced Pluripotent Stem Cell (hiPSC)-Derived Teratoma with an Integrin-Targeting Agent 99m Tc-3PRGD2. Mol imaging biology. 2013;15:58–67.

    Article  CAS  Google Scholar 

  68. Małkiewicz B, Świrkosz G, Lewandowski W, Demska K, Szczepaniak Z, Karwacki J, et al. Lymph Node Dissection in Testicular Cancer: The State of the Art and Future Perspectives. Curr Oncol Rep. 2024;26(4):318–35.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Saha GB. Physics and radiobiology of nuclear medicine. Springer Science & Business Media; 2012.

  70. Crișan G, Moldovean-Cioroianu NS, Timaru D-G, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT imaging: a literature review over the last decade. Int J Mol Sci. 2022;23(9):5023.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ward ZJ, Scott AM, Hricak H, Abdel-Wahab M, Paez D, Lette MM, et al. Estimating the impact of treatment and imaging modalities on 5-year net survival of 11 cancers in 200 countries: a simulation-based analysis. Lancet Oncol. 2020;21(8):1077–88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Sauerbrunn BJ, Andrews GA, Hübner KF. Ga-67 citrate imaging in tumors of the genito-urinary tract: report of cooperative study. J Nucl Med. 1978;19(5):470–5.

    PubMed  CAS  Google Scholar 

  73. Bekerman C, Hoffer PB, Bitran JD. The role of gallium-67 in the clinical evaluation of cancer. Semin Nucl Med. 1984;14(4):296–323.

    Article  PubMed  CAS  Google Scholar 

  74. Willan BD, Penney H, Castor WR, McGowan DG. The usefulness of gallium-67 citrate scanning in testicular seminoma. Clin Nucl Med. 1987;12(10):813–5.

    Article  PubMed  CAS  Google Scholar 

  75. Jana S, Blaufox MD. Nuclear Medicine Studies of the Prostate, Testes, and Bladder. Semin Nucl Med. 2006;36(1):51–72.

    Article  PubMed  Google Scholar 

  76. Ballou B, Levine G, Hakala TR, Solter D. Tumor location detected with radioactively labeled monoclonal antibody and external scintigraphy. Science. 1979;206(4420):844–7.

    Article  PubMed  CAS  Google Scholar 

  77. Javadpour N, Kim EE, DeLand FH, Salyer JR, Shah U, Goldenberg DM. The role of radioimmunodetection in the management of testicular cancer. JAMA. 1981;246(1):45–9.

    Article  PubMed  CAS  Google Scholar 

  78. Epenetos A, Hooker G, Durbin H, Bodmer W, Snook D, Begent R, et al. Indium-111 labelled monoclonal antibody to placental alkaline phosphatase in the detection of neoplasms of testis, ovary, and cervix. Lancet. 1985;326(8451):350–3.

    Article  Google Scholar 

  79. Bentestuen M, Al-Obaydi N, Zacho HD, editors. FAPI-avid nonmalignant PET/CT findings: an expedited systematic review. Seminars in Nuclear Medicine. Elsevier; 2023.

  80. Treglia G, Muoio B, Roustaei H, Kiamanesh Z, Aryana K, Sadeghi R. Head-to-head comparison of fibroblast activation protein inhibitors (FAPI) radiotracers versus [18F] F-FDG in oncology: a systematic review. Int J Mol Sci. 2021;22(20):11192.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Roustaei H, Kiamanesh Z, Askari E, Sadeghi R, Aryana K, Treglia G. Could fibroblast activation protein (FAP)-specific radioligands be considered as pan-tumor agents? Contrast Media & Molecular Imaging. 2022;2022.

  82. Pan Q, Luo Y, Zhang W. Idiopathic retroperitoneal fibrosis with intense uptake of 68Ga-fibroblast activation protein inhibitor and 18F-FDG. Clin Nucl Med. 2021;46(2):175–6.

    Article  PubMed  Google Scholar 

  83. Xi Y, Sun L, Che X, Huang X, Liu H, Wang Q, et al. A comparative study of [68Ga] Ga-FAPI-04 PET/MR and [18F] FDG PET/CT in the diagnostic accuracy and resectability prediction of ovarian cancer. Eur J Nucl Med Mol Imaging. 2023;50(9):2885–98.

    Article  PubMed  CAS  Google Scholar 

  84. Pang Y, Zhao L, Meng T, Xu W, Lin Q, Wu H, et al. PET imaging of fibroblast activation protein in various types of cancer using 68Ga-FAP-2286: comparison with 18F-FDG and 68Ga-FAPI-46 in a single-center, prospective study. J Nucl Med. 2023;64(3):386–94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Dai Y, Pang Y, Bao W, Cheng Y, Chen H. Pineal yolk sac tumor producing α-fetoprotein detected by 68Ga-FAPI PET/MRI. Clin Nucl Med. 2022;47(1):90–2.

    Article  PubMed  Google Scholar 

  86. Kircher M, Herhaus P, Schottelius M, Buck AK, Werner RA, Wester H-J, et al. CXCR4-directed theranostics in oncology and inflammation. Ann Nucl Med. 2018;32:503–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Zhao H, Guo L, Zhao H, Zhao J, Weng H, Zhao B. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6(7):5022.

    Article  PubMed  Google Scholar 

  88. Gilbert D, Chandler I, McIntyre A, Goddard N, Gabe R, Huddart R, Shipley J. Clinical and biological significance of CXCL12 and CXCR4 expression in adult testes and germ cell tumours of adults and adolescents. J Pathology: J Pathological Soc Great Br Irel. 2009;217(1):94–102.

    Article  CAS  Google Scholar 

  89. Mallik S, Qin G, Jia P, Zhao Z. Molecular signatures identified by integrating gene expression and methylation in non-seminoma and seminoma of testicular germ cell tumours. Epigenetics. 2021;16(2):162–76.

    Article  PubMed  Google Scholar 

  90. Gaytan F, Barreiro M, Caminos J, Chopin L, Herington A, Morales C, et al. Expression of ghrelin and its functional receptor, the type 1a growth hormone secretagogue receptor, in normal human testis and testicular tumors. J Clin Endocrinol Metabolism. 2004;89(1):400–9.

    Article  CAS  Google Scholar 

  91. Charron CL, Hou J, McFarland MS, Dhanvantari S, Kovacs MS, Luyt LG. Structure–activity study of ghrelin (1–8) resulting in high affinity fluorine-bearing ligands for the ghrelin receptor. J Med Chem. 2017;60(17):7256–66.

    Article  PubMed  CAS  Google Scholar 

  92. Larsen SKA, Løgager V, Bylov C, Nellemann H, Agerbæk M, Als AB, Pedersen EM. Can whole-body MRI replace CT in management of metastatic testicular cancer? A prospective, non-inferiority study. J Cancer Res Clin Oncol. 2023;149(3):1221–30.

    Article  PubMed  Google Scholar 

  93. Sohaib S, Koh D, Barbachano Y, Parikh J, Husband J, Dearnaley D, et al. Prospective assessment of MRI for imaging retroperitoneal metastases from testicular germ cell tumours. Clin Radiol. 2009;64(4):362–7.

    Article  PubMed  CAS  Google Scholar 

  94. Mosavi F, Laurell A, Ahlström H. Whole body MRI, including diffusion-weighted imaging in follow-up of patients with testicular cancer. Acta Oncol. 2015;54(10):1763–9.

    Article  PubMed  Google Scholar 

  95. Małkiewicz B, Świrkosz G, Lewandowski W, Demska K, Szczepaniak Z, Karwacki J et al. Lymph Node Dissection in Testicular Cancer: The State of the Art and Future Perspectives. Curr Oncol Rep. 2024:1–18.

  96. Will O, Purkayastha S, Chan C, Athanasiou T, Darzi AW, Gedroyc W, Tekkis PP. Diagnostic precision of nanoparticle-enhanced MRI for lymph-node metastases: a meta-analysis. Lancet Oncol. 2006;7(1):52–60.

    Article  PubMed  Google Scholar 

  97. Harisinghani MG, Saksena M, Ross RW, Tabatabaei S, Dahl D, McDougal S, Weissleder R. A pilot study of lymphotrophic nanoparticle-enhanced magnetic resonance imaging technique in early stage testicular cancer: a new method for noninvasive lymph node evaluation. Urology. 2005;66(5):1066–71.

    Article  PubMed  Google Scholar 

  98. Baessler B, Nestler T, Pinto dos Santos D, Paffenholz P, Zeuch V, Pfister D, et al. Radiomics allows for detection of benign and malignant histopathology in patients with metastatic testicular germ cell tumors prior to post-chemotherapy retroperitoneal lymph node dissection. Eur Radiol. 2020;30(4):2334–45.

    Article  PubMed  Google Scholar 

  99. King KG, Bhanvadia S, Ghodoussipour S, Hwang D, Varghese B, Cen SY, et al. Distinguishing fibrosis/necrosis from teratoma or viable disease in the retroperitoneum in post-chemotherapy, nonseminomatous testicular germ cell tumor using quantitative CT texture analysis. American Society of Clinical Oncology; 2018.

  100. Fournier C, Leguillette C, Leblanc E, Le Deley M-C, Carnot A, Pasquier D, et al. Diagnostic Value of the Texture Analysis Parameters of Retroperitoneal Residual Masses on Computed Tomographic Scan after Chemotherapy in Non-Seminomatous Germ Cell Tumors. Cancers. 2023;15(11):2997.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Scavuzzo A, Pasini G, Crescio E, Jimenez-Rios MA, Figueroa-Rodriguez P, Comelli A, et al. Radiomics Analyses to Predict Histopathology in Patients with Metastatic Testicular Germ Cell Tumors before Post-Chemotherapy Retroperitoneal Lymph Node Dissection. J Imaging. 2023;9(10):213.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Venishetty N, Taylor J, Xi Y, Howard JM, Ng YS, Wong D, et al. Testicular radiomics to predict pathology at time of postchemotherapy retroperitoneal lymph node dissection for nonseminomatous germ cell tumor. Clin Genitourin Cancer. 2024;22(1):33–7.

    Article  PubMed  Google Scholar 

  103. NITTA S, KOJIMA T, GIDO M, NAKAGAWA S, KAKEYA H, KANDORI S, et al. A Machine Learning Model to Predict the Histology of Retroperitoneal Lymph Node Dissection Specimens. Anticancer Res. 2024;44(5):2151–7.

    Article  PubMed  Google Scholar 

  104. King KG, Bhanvadia S, Ghodoussipour S, Hwang D, Varghese B, Cen SY, et al. Distinguishing fibrosis/necrosis from teratoma or viable disease in the retroperitoneum in post-chemotherapy, nonseminomatous testicular germ cell tumor using quantitative CT texture analysis. J Clin Oncol. 2018;36(6suppl):563.

    Article  Google Scholar 

  105. Lewin J, Dufort P, Halankar J, O’Malley M, Jewett MAS, Hamilton RJ, et al. Applying Radiomics to Predict Pathology of Postchemotherapy Retroperitoneal Nodal Masses in Germ Cell Tumors. JCO Clin Cancer Inf. 2018;2:1–12.

    Google Scholar 

  106. Nestler T, Baeßler B, Pinto dos Santos D, Pfister DJ, Maintz D, Heidenreich A. Predicting vital retroperitoneal residual tumors of metastatic testicular tumor patients after chemotherapy using radiomics. American Society of Clinical Oncology; 2019.

  107. Ding R, Li X, Liu Z. Overview of Novel Biomarkers for Management of Postchemotherapy Residual Masses in Testicular Cancer. Eur Urol Focus. 2024.

  108. Nestler T, Kremer L, von Brandenstein M, Wittersheim M, Paffenholz P, Wagener-Ryczek S, et al. Differentially expressed messenger RNA/proteins can distinguish teratoma from necrosis in postchemotherapy retroperitoneal lymph node dissection tissue. Cancer. 2023;129(4):634–42.

    Article  PubMed  CAS  Google Scholar 

  109. Katabathina VS, Vargas-Zapata D, Monge RA, Nazarullah A, Ganeshan D, Tammisetti V, Prasad SR. Testicular Germ Cell Tumors: Classification, Pathologic Features, Imaging Findings, and Management. Radiographics. 2021;41(6):1698–716.

    Article  PubMed  Google Scholar 

  110. Aide N, Briand M, Bohn P, Dutoit S, Lasnon C, Chasle J, et al. αvβ3 imaging can accurately distinguish between mature teratoma and necrosis in 18F-FDG-negative residual masses after treatment of non-seminomatous testicular cancer: a preclinical study. Eur J Nucl Med Mol Imaging. 2011;38(2):323–33.

    Article  PubMed  Google Scholar 

  111. Cao F, Li Z, Lee A, Liu Z, Chen K, Wang H, et al. Noninvasive De novo Imaging of Human Embryonic Stem Cell–Derived Teratoma Formation. Cancer Res. 2009;69(7):2709–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Bohn P, Modzelewski R, Rouvet J, Briand M, Dutoit S, Pille J-Y et al. Biodistribution and imaging of [99mTc]-HYNIC-RGD in MDA-MB-231 and NTERA-2 cancer cell xenografts. Nucl Med Commun. 2013;34(7).

  113. Lehner S, Lang C, Kaissis G, Todica A, Zacherl MJ, Boening G, et al. 124 I-PET assessment of human sodium iodide symporter reporter gene activity for highly sensitive in vivo monitoring of teratoma formation in mice. Mol imaging biology. 2015;17:874–83.

    Article  CAS  Google Scholar 

  114. Stroosma OB, Delaere KP. Carcinoid tumours of the testis. BJU Int. 2008;101(9):1101–5.

    Article  PubMed  Google Scholar 

  115. Amine MM, Mohamed B, Mourad H, Majed H, Slim C, Mehdi B, et al. Neuroendocrine Testicular Tumors: A Systematic Review and Meta-Analysis. Curr Urol. 2017;10(1):15–25.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Center UJCC. Prospective Exploratory Study of FAPi PET/CT with Histopathology Validation in Patients with Various Cancers. ClinicalTrialsgov IDNCT04459273.

Download references

Acknowledgements

Not applicable.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

NA & AMS: Conceptualization NA, EA and AMS: Methodology. EA, MF, MS, AA and HR: Data Collection and initial drafting. NA, AMS and EA: Review & Editing MF and MS: Revision NA and AMS: Supervision * NA and EA contributed equally as first authors.

Corresponding author

Correspondence to Andrew M. Scott.

Ethics declarations

Ethics approval and consent to participate

Not applicable. This is a review article.

Consent for publication

All authors have reviewed the final version and consented for publication of the study.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ayati, N., Askari, E., Fotouhi, M. et al. Nuclear medicine imaging in non-seminomatous germ cell tumors: lessons learned from the past failures. Cancer Imaging 24, 156 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40644-024-00794-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40644-024-00794-5

Keywords