Abstract
Testicular germ cell tumours (TGCTs) are the most common solid tumours in young men and have an excellent overall cure rate and prognosis. In most patients, localised disease is cured by surgery alone, and a minority of patients receive short-course adjuvant chemotherapy to reduce the risk of further relapse. Also, in about 80% of patients, metastatic disease can be cured by systemic cisplatin-based chemotherapy. Unfortunately, for a proportion of patients, the disease exhibits platinum resistance and relapse occurs. Despite further lines of systemic treatment, cure can be difficult to achieve in these patients and ultimately about 20% of them will die from disease progression. Addressing the mechanisms underpinning platinum resistance is critical to improving the survival and chances of cure for these patients. This review describes the latest advances in TGCT research, focusing on the identification of novel biomarkers, genetic characteristics and exploring novel treatments.
Keywords: testicular germ cell tumours, germ cell cancer, genetic hallmarks, biomarkers, novel treatments
Introduction
Testicular germ cell tumours (TGCTs) are the most common cancer in young adult men1,2. These tumours represent a rare neoplasm on a population scale (about 1% of all oncological diagnoses in men) and their incidence has been increasing progressively over the last three decades3,4.
TGCTs arise mostly in the testis but can develop in extra-gonadal sites, albeit rarely (2–5%). Histologically, they are classified as seminoma, non-seminoma (yolk sac, embryonal carcinoma, choriocarcinoma and teratoma) or mixed5.
In the last 40 years, excellent rates of cure and overall survival (OS) have been achieved, including in the metastatic setting, thanks to multimodality treatment but principally owing to the introduction of cisplatin-based chemotherapy regimens6. Unfortunately, in up to 30% of metastatic TGCT patients, their disease is not cured by initial systemic therapy and salvage treatment is required7. Limited therapeutic options are available for patients with platinum-refractory disease, and prognosis is dismal and cure rates are low (<5%)8.
Staging and prognosis assessment are crucial in the disease management of TGCTs. Since its publication in 1997, the International Germ Cell Cancer Collaborative Group (IGCCCG) prognostic classification has provided a valuable tool for patients with TGCTs in defining risk stratification on the basis of histology, marker levels and metastatic sites (Table 1)9.
Table 1. International Germ Cell Collaborative Group Prognostic Classification (1997).
Good | Intermediate | Poor | |
---|---|---|---|
Seminoma | Any primary site No NPVM |
Any primary site NPVM |
Not applicable |
Non- seminoma | All criteria: Gonadal/RP primary No NPVM hCG < 5000 IU/L AFP < 1000 ng/mL LDH < 1.5 ULN |
All criteria: Gonadal/RP primary No NPVM 5000 ≤ hCG ≤ 50,000 IU/L 1000 ≤ AFP ≤ 10,000 ng/mL 1.5 ≤ LDH ≤ 10 ULN |
Any criteria: Mediastinal primary NPVM hCG > 50,000 IU/L AFP > 10,000 ng/mL LDH > 10 ULN |
AFP, α-fetoprotein; hCG, human chorionic gonadotropin; LDH, lactate dehydrogenase; NPVM, non-pulmonary visceral metastasis; RP, retroperitoneal; ULN, upper limit of normal.
Notably, the IGCCCG classification in advanced non-seminoma was recently redefined, and encouraging results have arisen from patients who received treatment in more recent years (1990–2013) compared with historical data based on treatments delivered prior to the 1990s. In the updated series, progression-free survival (PFS) improved for poor-risk patients (5-year PFS of 54% versus 41%) whereas OS improved in all IGCCCG risk groups (5-year OS of 96%, 89% and 67% respectively in good, intermediate and poor prognosis). A new prognostic model that includes older age and presence of lung metastases as additional negative factors has been proposed10. Also, a lactate dehydrogenase (LDH) cut-off of 2.5 times the upper limit of normal has been proposed to refine the classification of seminoma patients without non-pulmonary visceral metastases (formerly included in the good-prognosis group) as their outcome reflects the IGCCCG intermediate-risk group11.
The PFS and OS improvements highlight progress in the management of TGCTs over the last 30 years but also confirm the need for further research focused on patients with cisplatin-refractory disease or relapse and those with late relapses (occurring 2 or more years after the completion of treatment).
This review discusses recent developments in testicular cancer biology and clinical management, focusing on the following areas:
-
-
tumour markers and biomarkers
-
-
genetic predisposition and hallmarks
-
-
platinum-resistant disease and novel treatments.
Tumour markers and biomarkers
Tumour serum protein markers (α-fetoprotein, human chorionic gonadotrophin and LDH) are widely used at diagnosis, in monitoring treatment response and in follow-up12. Additionally, their levels contribute to the risk stratification of patients with metastatic non-seminoma according to the IGCCCG prognostic classification9. However, in a significant proportion of patients, disease will be marker-negative and therefore tumour marker levels will not reflect disease burden13.
There is an unmet need for biomarker development in the following areas of clinical practice:
-
-
improving diagnostic performance at disease outset
-
-
using biomarkers to aid in identifying which stage 1 patients will relapse and should be offered adjuvant chemotherapy, sparing those who do not require it
-
-
improving early detection of relapse, particularly in those patients who are non-secretors of the traditional TGCT markers; this may allow a reduction in the radiation burden on these young patients and reduce imaging costs
-
-
identifying patients with residual active disease in post-chemotherapy masses.
Recent data suggest a promising role for circulating microRNAs (miRNAs) in addressing such questions. miRNAs are non-protein coding RNAs that regulate the expression of protein-coding genes.
There is evidence of significant overexpression in the tissue of specific miRNA clusters (miR-371-373 and miR-302) in all TGCTs, regardless of histological subtype, patient age or site of primary presentation, but not in normal tissue14. The identification of stable serum miRNAs has led to a number of studies aimed at identifying miRNAs at time of diagnosis and in response to treatment15,16.
A prospective multicentre study of 616 patients with primary diagnosed TGCTs and 258 controls has shown extremely high sensitivity (90.1%) and specificity (94%) of miRNA-371a-3p (M371 test by quantitative polymerase chain reaction) in all TGCT subgroups except teratoma17. The promising role of microRNA 371a-3p has also been investigated in a cohort of 24 low-stage chemotherapy-naïve patients undergoing retroperitoneal lymph node dissection, and results have been excellent (area under the curve [AUC] on receiver operating characteristic [ROC] analysis 0.965, sensitivity 100% and specificity 92%). However, miRNA was not predictive of pure teratoma18.
miRNA levels have also been investigated as predictors of residual viable disease at retroperitoneal lymph node dissection after chemotherapy in 82 patients with TGCTs, and results were positive (AUC 0.874)19.
Interestingly, in a recent pilot trial in 111 patients with TGCTs where miR-371a-3p expression was retrospectively and blindly analysed on prospectively obtained samples and compared against clinical events, extremely high sensitivity (96%) and specificity (100%) were demonstrated, along with a positive predictive value of 100% and a negative predictive value of 98% in predicting active TGCTs20.
A recent comprehensive review addresses differences among studies conducted to date and miRNA performance21. Circulating miRNAs are being validated in prospective randomised clinical trials (Table 2).
Table 2. MicroRNA (miRNA) evaluation in current prospective clinical trials.
Study | Type of study |
Intervention | Line | Patients | Serum miRNA | Timing | ClinicalTrials. gov Identifier |
---|---|---|---|---|---|---|---|
AGCT1531 | Phase III | Active Surveillance |
NA | Low-risk Stage I |
• Correlation of miRNA levels and stage I relapse |
• Pre orchidectomy • Every 1 month × 3 • Every 3 months for 1 year • Every 6 months for 1 year |
NCT03067181 |
Carboplatin versus cisplatin (+ etoposide, bleomycin) |
I | Standard-risk Metastatic |
• Marker decline on treatment • Identification prognostic miRNAs |
Not available | |||
UKP3BEP | Phase III randomised |
Accelerated versus standard BEP (bleomycin, etoposide and platinum) chemotherapy |
I | Intermediate/ poor-risk Metastatic/ Mediastinal primary |
• Marker decline on treatment • Identification prognostic miRNAs |
• Day 1 • Day 22 • Day 43 • End of treatment • 1 year |
NCT02582697 |
SWOG S1823 |
Prospective cohort study |
Surveillance | NA | Stage I | • Correlation of miR-371a-3p levels and stage I relapse |
• Pre-orchidectomy • Post-orchidectomy • 3 monthly (for 2 years) |
NCT04435756 |
NA, not applicable.
Although further follow-up, validation and standardisation are needed, the excellent performance of miRNAs in terms of sensitivity, specificity and short half-life in these studies shows promise for positively impacting on patient management.
Genetic predisposition and hallmarks
Compared with other solid tumours, TGCTs have a strong inherited genetic basis which accounts for almost half of the disease risk22. As far as development is concerned, TGCTs derive from reprogramming of cells in the early embryo and the germline, which dysregulates their developmental potency. A recent model identified seven types of TGCT, each harbouring unique epigenetic features23.
Genome-wide association studies have shed significant light on the factors leading to TGCT heritability, which, however, is not yet fully elucidated. First, heritability is not related to a single high-penetrance risk locus but instead is highly polygenic, and up to 49 risk loci have been identified so far24,25. Second, the role of several signalling pathways, such as KIT-KITLG signalling and DAZL and PRDM14 (both of which are involved in germ cell differentiation), has been identified as crucial to disease development and genomic integrity26.
Overall, TGCTs are characterised by a number of genetic hallmarks, which can be related to tumour development and can have an impact on treatment response and resistance.
Chromosomal abnormalities, such as increased copy number of chromosome p12 (mostly as isochromosome p12), which is a well-established pathognomonic factor in TGCTs, are common27,28.
Also, unlike the majority of solid tumours, TGCTs, owing to their embryonic origin, are known to harbour a very low mutational burden and typically exhibit a lack of recurrent somatic non-synonymous mutations (mean of 0.5 mutations per megabase)29.
The Cancer Genome Atlas Research Network analysis on 137 primary GCTs showed somatic mutations in KIT (18%), KRAS (14%), and NRAS (4%), exclusively in seminomas30.
Other identified mutations by whole-exome sequencing in 42 TGCT cases include the tumour suppressor gene CDC27 (11.9%). Copy number analysis showed amplification of the spermatocyte development gene FSIP2 (15.3%). In two patients with cisplatin-resistant disease, a missense XRCC2 mutation was also identified29.
Platinum-resistant disease and novel treatments
Platinum-resistant disease remains a management challenge in TGCTs, and there is no consensus on the optimum salvage treatment to achieve disease remission. Several platinum-based standard-dose chemotherapy regimens—VeIP (vinblastine + ifosfamide + cisplatin), VIP (etoposide + ifosfamide + cisplatin), TIP (paclitaxel + ifosfamide + cisplatin) and EP (etoposide + cisplatin)—are currently used in this setting31–33, as is high-dose chemotherapy followed by autologous bone marrow transplant34–38. The complexity and frequency of this situation mean that guidelines recommend that such treatment should be undertaken in specialist centres. Key to progress in this area is multicentre and multinational collaboration. This collaboration has been facilitated by the development of international germ cell tumour groups such as the International Global Germ Cell Tumor Collaborative Group (G3) and the Malignant Germ Cell International Consortium (MaGiC).
A key example of this is the international randomised phase 3 study (TIGER, ClinicalTrials.gov Identifier: NCT02375204) that is currently comparing conventional dose salvage treatment (TIP) with high-dose chemotherapy (two cycles of paclitaxel and ifosfamide followed by three cycles of carboplatin and etoposide). The study is actively recruiting and the results are long-awaited to inform the salvage treatment of TGCTs.
At a molecular level, platinum sensitivity and resistance have not been fully elucidated. It has been shown that platinum sensitivity depends highly on cisplatin-induced DNA damage (due to insufficient or inefficient nucleotide excision repair and double-strand break repair), intact p53 signalling39,40 and an increased chemotherapy-induced apoptotic response due to mitochondrial priming40.
p53 mutations and MDM2 amplifications have been identified in platinum-resistant TGCTs, particularly in those with adverse clinical features, and have been related to poor outcomes independent of the IGCCCG risk class41.
Recent whole-exome sequencing on platinum-resistant TGCTs, compared with platinum-sensitive tumours, has shown several hallmarks of platinum-resistant disease, including increasing copy number and structural aberrations and an increased frequency of mutations affecting KIT, p53 and WNT/CTNNB1 signalling genes as well as loss of pluripotency genes and hypermethylation42. For example, in a 2016 study40, apoptosis and pluripotency regulators NANOG and POU5F1 (also known as OCT3/4) expressed in TGCTs were not expressed in metastatic tumour deposits or mediastinal GCTs resistant to chemotherapy. However, the exact impact of these changes and the impact on mitochondrial priming are unknown.
Further research is needed to address the molecular basis of cisplatin resistance, and the exploration of novel treatments is a priority to improve outcomes in resistant disease.
A comprehensive description of targeted and novel treatments explored in TGCTs is beyond the scope of this review and has been addressed by others39,43. As summarised in Table 3, pathways explored include VEGF/PDGF-mediated angiogenesis, receptor tyrosine kinases (for example, c-KIT and MET), mechanistic target of rapamycin (mTOR) signalling, cyclin-dependent kinases, and poly (ADP-ribose) polymerase (PARP)-mediated DNA repair. Early-phase trials exploring the role of these novel agents in refractory TGCTs have shown disappointing results overall. While acknowledging that these studies often include heavily pre-treated patients, some of these studies were terminated early because of futility and because the observed overall response rates and outcomes were extremely poor. At the same time, it should be recognised that most of these studies have been in unselected patients.
Table 3. Clinical trials on novel treatments in testicular germ cell tumours.
Category | Drug | Phase | Patients enrolled (evaluable) |
Relative risk | Progression- free survival |
Overall survival |
Patient selection |
ClinicalTrials. gov Identifier |
Reference | Status |
---|---|---|---|---|---|---|---|---|---|---|
TKI | ||||||||||
PDGFR/VEGFR | Sunitinib | II | 10 | CR/PR 0% SD 50% |
- | - | Unselected | NCT00453310 | 46 | Completed |
II | 33 (32) | PR 9% SD 41% |
mPFS 2.0 months |
mOS 3.8 months |
Unselected | NCT00371553 | 45 | Completed | ||
II | 5 | - | 12-week PFS: 20.0% |
- | Unselected | NCT00912912 | 47 | Terminated (slow accrual) |
||
PDGFR/VEGFR/ FGFR/c-KIT |
Pazopanib | II | 43 | PR 4.7% SD 44.2% |
3-month PFS 12.8% mPFS 2.5 months |
1-year OS 28.5% mOS 5.3 months |
Unselected | NCT01743482 | 44 | Completed |
PDGFR/VEGFR/RAF/ c-KIT |
Sorafenib | II | 18 | CR/PR 0% SD 3/18 (>1 year) |
- | - | Unselected | NCT00772694 | 48 | Completed |
MET | Tivantinib | II | 27 (25) | CR/PR 0% SD 20.0% |
12-week PFS 21% mPFS 1 month |
mOS 6 months |
Unselected | NCT01055067 | 49 | Completed |
c-KIT, BCR-ABL, PDGFR |
Imatinib | II | 6 | CR/PR 0% | - | - | Selected | - | 50 | Terminated |
II | 7 | CR/PR/SD 0% | - | - | Selected | - | 51 | Terminated | ||
Anti CD30 | ||||||||||
Brentuximab | II | 9 | CR 11.1% PR 11.1% SD 22.2% |
3-month PFS 22.2% mPFS 1.5 months |
6-month OS 77.8% mOS 8.0 months |
Selected | NCT01851200 | 52 | Completed | |
II | 7 | CR 14.3% PR 14.3% SD 42.8% |
- | - | Selected | NCT01461538 | 53 | Completed | ||
II | 18 | - | - | - | Selected | NCT02689219 | - | Terminated | ||
mTOR inh | ||||||||||
Everolimus | II | 15 | CR/PR 0% | 12-week PFS 40.0% mPFS 1.7 months |
mOS 3.6 months |
Unselected | NCT01466231 | 54 | Terminated | |
Everolimus | II | 25 (22) | CR/PR 0% | 12-week PFS 0% mPFS 7.4 weeks |
mOS 8.3 weeks |
Unselected | NCT01242631 | 55 | Completed | |
Sirolimus + erlotinib |
II | 4 | - | - | - | Unselected | NCT01962896 | 56 | Terminated (low accrual) |
|
PARP inh | ||||||||||
Olaparib | II | 18 | CR/PR 0% SD 27.8% |
12-week PFS 27.8% |
12- month OS 27.8% |
Unselected | NCT02533765 | 57 | Active | |
Veliparib + carboplatin + gemcitabine |
II | - | - | - | - | Unselected | NCT02860819 | - | Active | |
CDK inh | ||||||||||
Palbociclib | II | 30 (29) | CR/PR 0% | 24-week PFS: 28% mPFS 11 weeks |
- | Selected | NCT01037790 | 58,59 | Completed | |
Ribociclib [teratoma] |
II R | 10 (8 ribociclib : 2 placebo) |
CR/PR 0% | 24-month PFS 71% ribociclib 0% placebo |
- | Unselected | NCT02300987 | 60 | Terminated (low accrual) | |
Ribociclib [teratoma] |
II | - | - | - | - | Selected | NCT02187783 | Completed | ||
Hypo-methylation | ||||||||||
DNMT | Guadecitabine + cisplatin |
I | 14 | ORR 23% | mPFS 1.7 months |
mOS 7.8 months |
Unselected | NCT02429466 | 61 | Completed |
DNMT | Guadecitabine + cisplatin + gemcitabine |
Ia | 2 | - | - | - | Unselected | 62 | Completed | |
Immunotherapy | ||||||||||
PDL1 | Avelumab | II | 8 | CR/PR 0% | 12-week PFS: 0% mPFS 0.9 months |
mOS: 2.7 months | Unselected | NCT03403777 | 63,64 | Terminated |
PDL1 | Atezolizumab | II | - | - | - | - | Unselected | NCT02458638 | - | Completed |
PDL1 + CTLA 4 | Durvalumab + tremelimumab |
II | - | - | - | - | Unselected | NCT03158064 | - | Recruiting |
PDL1 + CTLA 4 | Durvalumab +/− tremelimumab |
II R | 22 | - | - | - | Unselected | NCT03081923 | 65 | Recruiting Durvalumab monotherapy closed to accrual |
PD1 | Pembrolizumab | II | 12 | CR/PR 0% SD 16.7% |
- | - | Unselected | NCT02499952 | 66 | Terminated |
PD1 | Pembrolizumab | II | 12 | - | - | Unselected | NCT02721732 | 67 | Recruiting | |
PD1 + CTLA 4 | Nivolumab +/− ipilimumab |
II | - | - | - | Unselected | NCT02834013 | 68 | Recruiting | |
PD1 + CTLA 4 | Nivolumab + ipilimumab x 4 -> nivolumab maintenance |
II | 5 | CR/PR 0% SD 20.0% |
- | - | Unselected | NCT03333616 | 69 | Recruiting |
PD1 + VEGFR/MET + CTLA 4 |
Nivolumab + cabozantinib +/− ipilimumab |
I | 5 | CR/PR 0% | - | - | Unselected | NCT02496208 | 70 | Recruiting |
Other targets | ||||||||||
Claudin 6 | ASP1650 | II | - | - | - | - | Unselected | NCT03760081 | 71 | Completed |
B-RAF/MEK | Dabrafenib + trametinib |
II | - | - | - | - | Selected | NCT02034110 | 72 | Active |
ALDH | Cisplatin + disulfiram |
II | - | - | - | - | Unselected | NCT03950830 | Recruiting |
aSGI-110 with cisplatin and gemcitabine chemotherapy in patients with bladder cancer (EudraCT: 2015-004062-29). CR, complete response; mOS, median overall survival; mPFS, median progression-free survival; PFS, progression-free survival; PR, partial response; SD, stable disease.
A phase II single-arm study of pazopanib, an angiogenesis-targeted treatment, in 43 patients who had progressed after two or more platinum-based regimens yielded a 3-month PFS of 12.8% and a 1-year OS of 28.5%44. Likewise, despite promising pre-clinical activity, sunitinib showed poor clinical results in patients with cisplatin-refractory disease (partial responses ranged from 0 to 9%)45,46.
The role of KIT in genetic predisposition and the frequency of somatic mutations suggested this as an attractive target. Unfortunately, the reported responses with the tyrosine kinase inhibitors imatinib and tivantinib were disappointing49–51. This lack of activity may reflect the molecular features of TGCTs, as most identified KIT mutations are localised on exon 17 and associate with imatinib resistance41. Likewise, a phase II study of everolimus reported disappointing results55. A previous phase II study in the same setting was terminated because of futility; there were no responses in the first 15 patients who received treatment54. Similar results were reported in phase II studies addressing the cyclin-dependent kinases CDK4–658–60. The hypothesis that cisplatin-sensitive tumours may also be sensitive to PARP inhibitors has led to testing of these inhibitors but with little evidence of activity to date57.
The lack of impact of targeted therapies on the disease has prompted exploration of alternative approaches.
Some activity has been seen with the antibody conjugate brentuximab vedotin targeting the CD30 antigen. In a small cohort of heavily pre-treated patients with CD30-expressing TGCTs, 3-month PFS and 6-month OS rates were 22.2% and 77.8% respectively52. In a different case series of seven patients who received brentuximab vedotin, two patients achieved an objective response53.
PDL1 expression in a significant proportion of TGCTs has led to the exploration of checkpoint inhibitors despite the low tumour mutational burden of these cancers. However, studies of the PD1 inhibitor pembrolizumab and PDL1 inhibitors avelumab and durvalumab have failed to show clinical activity, and no responses were observed in TGCTs63–67.
Current studies are investigating PD1-directed agents alone or in combination (such as nivolumab + ipilimumab and durvalumab/tremelimumab)68,69. Cabozantinib +/− ipilimumab in a phase I study, however, showed no observed responses in the TGCT subgroup70.
Hypomethylating agents such as guadecitabine (SGI-110) have been suggested as potentially promising novel targets in view of the observed DNA hypermethylation exhibited by platinum-refractory TGCTs as opposed to platinum-sensitive tumours. In pre-clinical studies, TGCTs were extremely sensitive to low-dose decitabine, a DNA methyltransferase inhibitor, which restored sensitivity to cisplatin in cell lines73. A phase I study of guadecitabine and cisplatin in 14 patients with TGCTs showed an overall response rate of 23%61; excellent responses were also reported in two patients with platinum-refractory disease treated in a phase 1 study of cisplatin, gemcitabine and guadecitabine62.
It is still unknown to what extent the lack of activity observed with the majority of these agents is due to patient selection or to TGCT intrinsic biology. Certainly, further research on biomarkers is needed to identify novel treatments in refractory disease.
Conclusions
TGCTs are a heterogeneous group of diseases which in general have excellent cure rates that have improved over the last three decades. Treatment and cure of cisplatin-refractory disease are challenging, and the optimal treatment for these patients is not yet clear. Further research on cisplatin resistance is needed to expand the current therapeutic options and to achieve better outcomes for patients with refractory disease.
Promising novel biomarkers are being investigated and should their role be fully validated, the management of patients with TGCTs will certainly evolve.
Given the complexity of its management and multimodality treatment, referral to high-volume centres is crucial. As with any rare disease, international scientific collaboration such as the International Global Germ Cell Tumor Collaborative Group (G3) and the Malignant Germ Cell International Consortium (MaGIC) is the key to allow faster advances in research and clinical practice.
The peer reviewers who approve this article are:
Ben Tran, Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
Constantine Alifrangis, Department of Medical Oncology, University College London Hospitals & St Bartholomew's Hospital, London, UK
Funding Statement
This review represents independent research funded by the National Institute for Health Research (NIHR) Biomedical Research Centre at the Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, London. The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
References
- 1.Bosl GJ, Motzer RJ: Testicular germ-cell cancer. N Engl J Med. 1997; 337(4): 242–53. 10.1056/NEJM199707243370406 [DOI] [PubMed] [Google Scholar]
- 2.Horwich A, Shipley J, Huddart R: Testicular germ-cell cancer. Lancet. 2006; 367(9512): 754–65. 10.1016/S0140-6736(06)68305-0 [DOI] [PubMed] [Google Scholar]
- 3.Bray F, Ferlay J, Soerjomataram I, et al. : Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68(6): 394–424. 10.3322/caac.21492 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 4.Gurney JK, Florio AA, Znaor A, et al. : International Trends in the Incidence of Testicular Cancer: Lessons from 35 Years and 41 Countries. Eur Urol. 2019; 76(5): 615–23. 10.1016/j.eururo.2019.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 5.Moch H, Cubilla AL, Humphrey PA, et al. : The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur Urol. 2016; 70(1): 93–105. 10.1016/j.eururo.2016.02.029 [DOI] [PubMed] [Google Scholar]
- 6.Einhorn LH, Donohue J: Cis-diamminedichloroplatinum, vinblastine, and bleomycin combination chemotherapy in disseminated testicular cancer. Ann Intern Med. 1977; 87(3): 293–8. 10.7326/0003-4819-87-3-293 [DOI] [PubMed] [Google Scholar]
- 7.Feldman DR, Bosl GJ, Sheinfeld J, et al. : Medical treatment of advanced testicular cancer. JAMA. 2008; 299(6): 672–84. 10.1001/jama.299.6.672 [DOI] [PubMed] [Google Scholar]
- 8.Porcu P, Bhatia S, Sharma M, et al. : Results of treatment after relapse from high-dose chemotherapy in germ cell tumors. J Clin Oncol. 2000; 18(6): 1181–6. 10.1200/JCO.2000.18.6.1181 [DOI] [PubMed] [Google Scholar]
- 9.International Germ Cell Consensus Classification: A prognostic factor-based staging system for metastatic germ cell cancers. International Germ Cell Cancer Collaborative Group. J Clin Oncol. 1997; 15(2): 594–603. 10.1200/JCO.1997.15.2.594 [DOI] [PubMed] [Google Scholar]
- 10.Gillessen S, Collette L, Daugaard G, et al. : 9030 - Redefining the IGCCCG classification in advanced non-seminoma. Ann Oncol. 2019; 30(Supplement 5): v357–v358. 10.1093/annonc/mdz249.002 [DOI] [Google Scholar]
- 11.Beyer J, Collette L, Daugaard G, et al. : Prognostic factors in advanced seminoma: An analysis from the IGCCCG Update Consortium. J Clin Oncol. 2020; 38(6_suppl): 386. 10.1200/JCO.2020.38.6_suppl.386 [DOI] [Google Scholar]
- 12.Gilligan TD, Seidenfeld J, Basch EM, et al. : American Society of Clinical Oncology Clinical Practice Guideline on uses of serum tumor markers in adult males with germ cell tumors. J Clin Oncol. 2010; 28(20): 3388–404. 10.1200/JCO.2009.26.4481 [DOI] [PubMed] [Google Scholar]
- 13.Murray MJ, Huddart RA, Coleman N: The present and future of serum diagnostic tests for testicular germ cell tumours. Nat Rev Urol. 2016; 13(12): 715–25. 10.1038/nrurol.2016.170 [DOI] [PubMed] [Google Scholar]
- 14.Palmer RD, Murray MJ, Saini HK, et al. : Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Res. 2010; 70(7): 2911–23. 10.1158/0008-5472.CAN-09-3301 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Murray MJ, Halsall DJ, Hook CE, et al. : Identification of microRNAs From the miR-371~373 and miR-302 clusters as potential serum biomarkers of malignant germ cell tumors. Am J Clin Pathol. 2011; 135(1): 119–25. 10.1309/AJCPOE11KEYZCJHT [DOI] [PubMed] [Google Scholar]
- 16.Gillis AJM, Rijlaarsdam MA, Eini R, et al. : Targeted serum miRNA (TSmiR) test for diagnosis and follow-up of (testicular) germ cell cancer patients: A proof of principle. Mol Oncol. 2013; 7(6): 1083–92. 10.1016/j.molonc.2013.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dieckmann KP, Radtke A, Geczi L, et al. : Serum Levels of MicroRNA-371a-3p (M371 Test) as a New Biomarker of Testicular Germ Cell Tumors: Results of a Prospective Multicentric Study. J Clin Oncol. 2019; 37(16): 1412–23. 10.1200/JCO.18.01480 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 18.Lafin JT, Singla N, Woldu SL, et al. : Serum MicroRNA-371a-3p Levels Predict Viable Germ Cell Tumor in Chemotherapy-naïve Patients Undergoing Retroperitoneal Lymph Node Dissection. Eur Urol. 2020; 77(2): 290–2. 10.1016/j.eururo.2019.10.005 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 19.Leão R, van Agthoven T, Figueiredo A, et al. : Serum miRNA Predicts Viable Disease after Chemotherapy in Patients with Testicular Nonseminoma Germ Cell Tumor. J Urol. 2018; 200(1): 126–35. 10.1016/j.juro.2018.02.068 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 20.Nappi L, Thi M, Lum A, et al. : Developing a Highly Specific Biomarker for Germ Cell Malignancies: Plasma miR371 Expression Across the Germ Cell Malignancy Spectrum. J Clin Oncol. 2019; 37(33): 3090–8. 10.1200/JCO.18.02057 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 21.Almstrup K, Lobo J, Mørup N, et al. : Application of miRNAs in the diagnosis and monitoring of testicular germ cell tumours. Nat Rev Urol. 2020; 17(4): 201–13. 10.1038/s41585-020-0296-x [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 22.Litchfield K, Thomsen H, Mitchell JS, et al. : Quantifying the heritability of testicular germ cell tumour using both population-based and genomic approaches. Sci Rep. 2015; 5: 13889. 10.1038/srep13889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Oosterhuis JW, Looijenga LHJ: Human germ cell tumours from a developmental perspective. Nat Rev Cancer. 2019; 19(9): 522–37. 10.1038/s41568-019-0178-9 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 24.Litchfield K, Levy M, Orlando G, et al. : Identification of 19 new risk loci and potential regulatory mechanisms influencing susceptibility to testicular germ cell tumor. Nat Genet. 2017; 49(7): 1133–40. 10.1038/ng.3896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wang Z, McGlynn KA, Rajpert-De Meyts E, et al. : Meta-analysis of five genome-wide association studies identifies multiple new loci associated with testicular germ cell tumor. Nat Genet. 2017; 49(7): 1141–7. 10.1038/ng.3879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Litchfield K, Levy M, Huddart RA, et al. : The genomic landscape of testicular germ cell tumours: From susceptibility to treatment. Nat Rev Urol. 2016; 13(7): 409–19. 10.1038/nrurol.2016.107 [DOI] [PubMed] [Google Scholar]
- 27.Atkin NB, Baker MC: Specific chromosome change, i(12p), in testicular tumours? Lancet. 1982; 2(8311): 1349. 10.1016/s0140-6736(82)91557-4 [DOI] [PubMed] [Google Scholar]
- 28.Summersgill B, Osin P, Lu YJ, et al. : Chromosomal imbalances associated with carcinoma in situ and associated testicular germ cell tumours of adolescents and adults. Br J Cancer. 2001; 85(2): 213–20. 10.1054/bjoc.2001.1889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Litchfield K, Summersgill B, Yost S, et al. : Whole-exome sequencing reveals the mutational spectrum of testicular germ cell tumours. Nat Commun. 2015; 6: 5973. 10.1038/ncomms6973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Shen H, Shih J, Hollern DP, et al. : Integrated Molecular Characterization of Testicular Germ Cell Tumors. Cell Rep. 2018; 23(11): 3392–406. 10.1016/j.celrep.2018.05.039 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 31.Motzer RJ, Geller NL, Tan CCY, et al. : Salvage chemotherapy for patients with germ cell tumors. The memorial sloan-kettering cancer center experience (1979-1989). Cancer. 1991; 67(5): 1305–10. [DOI] [PubMed] [Google Scholar]
- 32.Loehrer PJ, Sr, Gonin R, Nichols CR, et al. : Vinblastine plus ifosfamide plus cisplatin as initial salvage therapy in recurrent germ cell tumor. J Clin Oncol. 1998; 16(7): 2500–4. 10.1200/JCO.1998.16.7.2500 [DOI] [PubMed] [Google Scholar]
- 33.Kondagunta GV, Bacik J, Donadio A, et al. : Combination of paclitaxel, ifosfamide, and cisplatin is an effective second-line therapy for patients with relapsed testicular germ cell tumors. J Clin Oncol. 2005; 23(27): 6549–55. 10.1200/JCO.2005.19.638 [DOI] [PubMed] [Google Scholar]
- 34.Pico JL, Rosti G, Kramar A, et al. : A randomised trial of high-dose chemotherapy in the salvage treatment of patients failing first-line platinum chemotherapy for advanced germ cell tumours. Ann Oncol. 2005; 16(7): 1152–9. 10.1093/annonc/mdi228 [DOI] [PubMed] [Google Scholar]
- 35.Einhorn LH, Williams SD, Chamness A, et al. : High-dose chemotherapy and stem-cell rescue for metastatic germ-cell tumors. N Engl J Med. 2007; 357(4): 340–8. 10.1056/NEJMoa067749 [DOI] [PubMed] [Google Scholar]
- 36.Feldman DR, Sheinfeld J, Bajorin DF, et al. : TI-CE high-dose chemotherapy for patients with previously treated germ cell tumors: Results and prognostic factor analysis. J Clin Oncol. 2010; 28(10): 1706–13. 10.1200/JCO.2009.25.1561 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 37.Lorch A, Kleinhans A, Kramar A, et al. : Sequential versus single high-dose chemotherapy in patients with relapsed or refractory germ cell tumors: Long-term results of a prospective randomized trial. J Clin Oncol. 2012; 30(8): 800–5. 10.1200/JCO.2011.38.6391 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 38.Adra N, Abonour R, Althouse SK, et al. : High-Dose Chemotherapy and Autologous Peripheral-Blood Stem-Cell Transplantation for Relapsed Metastatic Germ Cell Tumors: The Indiana University Experience. J Clin Oncol. 2017; 35(10): 1096–102. 10.1200/JCO.2016.69.5395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.de Vries G, Rosas-Plaza X, van Vugt MATM, et al. : Testicular cancer: Determinants of cisplatin sensitivity and novel therapeutic opportunities. Cancer Treat Rev. 2020; 88: 102054. 10.1016/j.ctrv.2020.102054 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 40.Taylor-Weiner A, Zack T, O'Donnell E, et al. : Genomic evolution and chemoresistance in germ-cell tumours. Nature. 2016; 540(7631): 114–8. 10.1038/nature20596 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bagrodia A, Lee BH, Lee W, et al. : Genetic Determinants of Cisplatin Resistance in Patients With Advanced Germ Cell Tumors. J Clin Oncol. 2016; 34(33): 4000–7. 10.1200/JCO.2016.68.7798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Loveday C, Litchfield K, Proszek PZ, et al. : Genomic landscape of platinum resistant and sensitive testicular cancers. Nat Commun. 2020; 11(1): 2189. 10.1038/s41467-020-15768-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Galvez-Carvajal L, Sanchez-Muñoz A, Ribelles N, et al. : Targeted treatment approaches in refractory germ cell tumors. Crit Rev Oncol Hematol. 2019; 143: 130–8. 10.1016/j.critrevonc.2019.09.005 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 44.Necchi A, Lo Vullo S, Giannatempo P, et al. : Pazopanib in advanced germ cell tumors after chemotherapy failure: Results of the open-label, single-arm, phase 2 Pazotest trial. Ann Oncol. 2017; 28(6): 1346–51. 10.1093/annonc/mdx124 [DOI] [PubMed] [Google Scholar]
- 45.Oechsle K, Honecker F, Cheng T, et al. : Preclinical and clinical activity of sunitinib in patients with cisplatin-refractory or multiply relapsed germ cell tumors: A Canadian Urologic Oncology Group/German Testicular Cancer Study Group cooperative study. Ann Oncol. 2011; 22(12): 2654–60. 10.1093/annonc/mdr026 [DOI] [PubMed] [Google Scholar]
- 46.Feldman DR, Turkula S, Ginsberg MS, et al. : Phase II trial of sunitinib in patients with relapsed or refractory germ cell tumors. Invest New Drugs. 2010; 28(4): 523–8. 10.1007/s10637-009-9280-2 [DOI] [PubMed] [Google Scholar]
- 47.Subbiah V, Meric-Bernstam F, Mills GB, et al. : Next generation sequencing analysis of platinum refractory advanced germ cell tumor sensitive to Sunitinib (Sutent®) a VEGFR2/PDGFRβ/c-kit/ FLT3/RET/CSF1R inhibitor in a phase II trial. J Hematol Oncol. 2014; 7: 52. 10.1186/s13045-014-0052-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Skoneczna IA, Natorska U, Tacikowska M, et al. : Sorafenib monotherapy in patients with inoperable/recurrent germ cell tumors (GCT) refractory to chemotherapy: Phase II study. J Clin Oncol. 2014; 32(4_suppl): 367. 10.1200/jco.2014.32.4_suppl.36724419110 [DOI] [Google Scholar]
- 49.Feldman DR, Einhorn LH, Quinn DI, et al. : A phase 2 multicenter study of tivantinib (ARQ 197) monotherapy in patients with relapsed or refractory germ cell tumors. Invest New Drugs. 2013; 31(4): 1016–22. 10.1007/s10637-013-9934-y [DOI] [PubMed] [Google Scholar]
- 50.Einhorn LH, Brames MJ, Heinrich MC, et al. : Phase II study of imatinib mesylate in chemotherapy refractory germ cell tumors expressing KIT. Am J Clin Oncol. 2006; 29(1): 12–3. 10.1097/01.coc.0000195086.47548.ef [DOI] [PubMed] [Google Scholar]
- 51.Piulats JM, Garcia del Muro X, Huddart R, et al. : Phase II multicenter study of imatinib in patients with chemorefractory germ cell tumors that express c-kit. Cancer Res. 2007; 67(9 Supplement): 2648 LP–2648. Reference Source [Google Scholar]
- 52.Necchi A, Anichini A, Raggi D, et al. : Brentuximab Vedotin in CD30-Expressing Germ Cell Tumors After Chemotherapy Failure. Clin Genitourin Cancer. 2016; 14(4): 261–264.e4. 10.1016/j.clgc.2016.03.020 [DOI] [PubMed] [Google Scholar]
- 53.Albany C, Einhorn L, Garbo L, et al. : Treatment of CD30-Expressing Germ Cell Tumors and Sex Cord Stromal Tumors with Brentuximab Vedotin: Identification and Report of Seven Cases. Oncologist. 2018; 23(3): 316–23. 10.1634/theoncologist.2017-0544 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 54.Mego M, Svetlovska D, Miskovska V, et al. : Phase II study of everolimus in refractory testicular germ cell tumors. Urol Oncol. 2016; 34(3): 122.e17–22. 10.1016/j.urolonc.2015.10.010 [DOI] [PubMed] [Google Scholar]
- 55.Fenner M, Oing C, Dieing A, et al. : Everolimus in patients with multiply relapsed or cisplatin refractory germ cell tumors: Results of a phase II, single-arm, open-label multicenter trial (RADIT) of the German Testicular Cancer Study Group. J Cancer Res Clin Oncol. 2019; 145(3): 717–23. 10.1007/s00432-018-2752-z [DOI] [PubMed] [Google Scholar]
- 56.Laetsch TW, Kumar K, Rakheja D, et al. : A phase II study of sirolimus and erlotinib in recurrent/refractory germ cell tumors. J Clin Oncol. 2014; 32(15_suppl): TPS2638. 10.1200/jco.2014.32.15_suppl.tps2638 [DOI] [Google Scholar]
- 57.de Giorgi U, Schepisi G, Gurioli G, et al. : Olaparib as salvage treatment for advanced germ cell tumors after chemotherapy failure: Results of the open-label, single-arm, IGG-02 phase II trial. J Clin Oncol. 2020; 38(15): 5058. 10.1200/JCO.2020.38.15_suppl.5058 [DOI] [Google Scholar]
- 58.Vaughn DJ, Hwang WT, Lal P, et al. : Phase 2 trial of the cyclin-dependent kinase 4/6 inhibitor palbociclib in patients with retinoblastoma protein-expressing germ cell tumors. Cancer. 2015; 121(9): 1463–8. 10.1002/cncr.29213 [DOI] [PubMed] [Google Scholar]
- 59.Narayan V, Hwang WT, Lal P, et al. : Cyclin-Dependent Kinase 4/6 Inhibition for the Treatment of Unresectable Mature Teratoma: Long-Term Follow-Up of a Phase II Study. Clin Genitourin Cancer. 2016; 14(6): 504–10. 10.1016/j.clgc.2016.03.010 [DOI] [PubMed] [Google Scholar]
- 60.Castellano DE, Quinn DI, Feldman DR, et al. : A phase II study of ribociclib in men with unresectable, incurable teratoma with recent progression. J Clin Oncol. 2019; 37(7): 517. 10.1200/JCO.2019.37.7_suppl.517 [DOI] [Google Scholar]
- 61.Albany C, Fazal Z, Singh R, et al. : A phase 1 study of combined guadecitabine and cisplatin in platinum refractory germ cell cancer. Cancer Med. 2021; 10(1): 156–63. 10.1002/cam4.3583 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 62.Crabb SJ, Huddart RA, Brown E, et al. : Response to guadecitabine (SGI-110) combined with cisplatin and gemcitabine (GCG) in platinum refractory germ cell tumors (GCTs). J Clin Oncol. 2020; 38(15_suppl): e17057. 10.1200/JCO.2020.38.15_suppl.e17057 [DOI] [Google Scholar]
- 63.Mego M, Svetlovska D, Chovanec M, et al. : Phase II study of avelumab in multiple relapsed/refractory germ cell cancer. Invest New Drugs. 2019; 37(4): 748–54. 10.1007/s10637-019-00805-4 [DOI] [PubMed] [Google Scholar]
- 64.Mego M, Svetlovska D, Chovanec M, et al. : Phase II study of avelumab in multiple relapsed/refractory testicular germ cell cancer. J Clin Oncol. 2019; 37(15_suppl): e16045. 10.1200/JCO.2019.37.15_suppl.e16045 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 65.Necchi A, Giannatempo P, Raggi D, et al. : An Open-label Randomized Phase 2 study of Durvalumab Alone or in Combination with Tremelimumab in Patients with Advanced Germ Cell Tumors (APACHE): Results from the First Planned Interim Analysis. Eur Urol. 2019; 75(1): 201–3. 10.1016/j.eururo.2018.09.010 [DOI] [PubMed] [Google Scholar]
- 66.Adra N, Einhorn LH, Althouse SK, et al. : Phase II trial of pembrolizumab in patients with platinum refractory germ-cell tumors: A Hoosier Cancer Research Network Study GU14-206. Ann Oncol. 2018; 29(1): 209–14. 10.1093/annonc/mdx680 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 67.Naing A, Meric-Bernstam F, Stephen B, et al. : Phase 2 study of pembrolizumab in patients with advanced rare cancers. J Immunother Cancer. 2020; 8(1): e000347. 10.1136/jitc-2019-000347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Patel SP, Othus M, Chae YK, et al. : SWOG 1609 (DART): A phase II basket trial of dual anti-CTLA-4 and anti-PD-1 blockade in rare tumors. J Clin Oncol. 2019; 37(15_suppl): TPS2658. 10.1200/JCO.2019.37.15_suppl.TPS2658 [DOI] [Google Scholar]
- 69.McGregor BA, Campbell MT, Xie W, et al. : Phase II study of nivolumab and ipilimumab for advanced rare genitourinary cancers. J Clin Oncol. 2020; 38(15_suppl): 5018. 10.1200/JCO.2020.38.15_suppl.5018 [DOI] [Google Scholar]
- 70.Nadal RM, Mortazavi A, Stein M, et al. : Results of phase I plus expansion cohorts of cabozantinib (Cabo) plus nivolumab (Nivo) and CaboNivo plus ipilimumab (Ipi) in patients (pts) with with metastatic urothelial carcinoma (mUC) and other genitourinary (GU) malignancies. J Clin Oncol. 2018; 36(6_suppl): 515. 10.1200/JCO.2018.36.6_suppl.51529267131 [DOI] [Google Scholar]
- 71.Adra N, Vaughn DJ, Einhorn L, et al. : A phase II study assessing the safety and efficacy of ASP1650 in male patients with incurable platinum refractory germ cell tumors. J Clin Oncol. 2020; 38(6_suppl): TPS424. 10.1200/JCO.2020.38.6_suppl.TPS424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Subbiah V, Bang YJ, Lassen UN, et al. : ROAR: A phase 2, open-label study in patients (pts) with BRAF V600E-mutated rare cancers to investigate the efficacy and safety of dabrafenib (D) and trametinib (T) combination therapy. J Clin Oncol. 2016; 34(15_suppl): TPS2604. 10.1200/JCO.2016.34.15_suppl.TPS2604 [DOI] [Google Scholar]
- 73.Beyrouthy MJ, Garner KM, Hever MP, et al. : High DNA methyltransferase 3B expression mediates 5-aza-deoxycytidine hypersensitivity in testicular germ cell tumors. Cancer Res. 2009; 69(24): 9360–6. 10.1158/0008-5472.CAN-09-1490 [DOI] [PMC free article] [PubMed] [Google Scholar]