Attention: Restrictions on use of AUA, AUAER, and UCF content in third party applications, including artificial intelligence technologies, such as large language models and generative AI.
You are prohibited from using or uploading content you accessed through this website into external applications, bots, software, or websites, including those using artificial intelligence technologies and infrastructure, including deep learning, machine learning and large language models and generative AI.

Diagnosis and Treatment of Early-Stage Testicular Cancer: AUA Guideline

Using AUA Guidelines

This AUA guideline is provided free of use to the general public for academic and research purposes. However, any person or company accessing AUA guidelines for promotional or commercial use must obtain a licensed copy. To obtain the licensable copy of this guideline, please contact Keith Price at kprice@auanet.org.

(Published 2019; Amended 2023)

To cite this guideline:

Stephenson A, Bass EB, Bixler BR, et al. Diagnosis and treatment of early-stage testicular cancer: AUA Guideline amendment 2023. J Urol. 2023;10.1097/JU.0000000000003694. https://doi.org/10.1097/JU.0000000000003694

Unabridged version of this guideline [pdf]
Amendment Summary [pdf]

Panel Members

Andrew Stephenson, MD; Scott E. Eggener, MD; David M. Chelnick, BS; Siamak Daneshmand, MD; Darren Feldman, MD; Timothy Gilligan, MD; Jose A. Karam, MD; Bradley Leibovich, MD, FACS; Stanley L. Liauw, MD; Timothy A. Masterson, MD; Joshua J. Meeks, MD, PhD; Phillip M. Pierorazio, MD; Joel Sheinfeld, MD

2023 Amendment Panel

Andrew Stephenson, MD; Siamak Daneshmand, MD; Philippe E Spiess, MD

Staff and Consultants

Brooke R. Bixler, MPH; Erin Kirkby, MS; Eric B. Bass, MD, MPH; Alexis Marianes, PhD; Ritu Sharma

SUMMARY

Purpose

Testis cancer is the most common solid malignancy in young males. Testis cancer is a relatively rare malignancy, with outcomes defined by specific cancer- and patient-related factors. The vast majority of men with testis cancer have low-stage disease (limited to the testis and retroperitoneum; clinical stages I-IIB); survival rates are high with standard therapy. A priority for those patients with low-stage disease is limiting the burden of therapy and treatment-related toxicity without compromising cancer control. Thus, surveillance has assumed an increasing role among those with cancer clinically confined to the testis.  Likewise, paradigms for management have undergone substantial changes in recent years as evidence regarding risk stratification, recurrence, survival, and treatment-related toxicity has emerged.

Methodology

The systematic review utilized to inform this guideline was conducted by a methodology team at the Johns Hopkins University Evidence-based Practice Center. Scoping of the report and review of the final systematic review to inform guideline statements was conducted in conjunction with the Testicular Cancer expert panel. The methodology team searched using PubMed®, Embase®, and the Cochrane Central Register of Controlled Trials (CENTRAL) from January 1980 through August 2018. The team developed a search strategy by using medical subject heading (MeSH) terms and key words relevant to the diagnosis and treatment of early-stage testicular cancer. The evidence review team also reviewed relevant systematic reviews and references provided by the panel to identify articles that may have been missed by the database searches. Searches were updated using the same methodological protocol to capture literature published through March 2023.

GUIDELINE STATEMENTS

Initial Management

Diagnosis and Initial Consultation

1. A solid mass in the testis identified by physical exam or imaging should be managed as a malignant neoplasm until proven otherwise. (Clinical Principle)

2. In a man with a solid mass in the testis suspicious for malignant neoplasm, serum tumor markers (AFP, hCG, and LDH) should be drawn and measured prior to any treatment, including orchiectomy. (Moderate Recommendation; Evidence Level: Grade C)

3. Prior to definitive management, patients should be counseled about the risks of hypogonadism and infertility (Moderate Recommendation; Evidence Level: Grade C) and should be offered sperm banking, when appropriate. In patients without a normal contralateral testis or with known subfertility, this should be considered prior to orchiectomy. (Clinical Principle)

4. Scrotal ultrasound with Doppler should be obtained in patients with a unilateral or bilateral scrotal mass suspicious for neoplasm. (Strong Recommendation; Evidence Level: Grade B)

5. Testicular microlithiasis in the absence of solid mass and risk factors for developing a germ cell tumor (GCT) does not confer an increased risk of malignant neoplasm and does not require further evaluation. (Moderate Recommendation; Evidence Level: Grade C)

6. Patients with normal serum tumor markers (hCG and AFP) and indeterminate findings on physical exam or testicular ultrasound for testicular neoplasm should undergo repeat imaging in six to eight weeks. (Clinical Principle)

7. Magnetic Resonance Imaging (MRI) should not be used in the initial evaluation and diagnosis of a testicular lesion suspicious for neoplasm. (Moderate Recommendation; Evidence Level: Grade C)

Orchiectomy

8. Patients with a testicular lesion suspicious for malignant neoplasm and a normal contralateral testis should undergo a radical inguinal orchiectomy; testis-sparing surgery (TSS) is not recommended. Transscrotal orchiectomy is discouraged. (Strong Recommendation; Evidence Level: Grade B)

9. Testicular prosthesis should be discussed prior to orchiectomy. (Expert Opinion)

10. Patients who have undergone scrotal orchiectomy for malignant neoplasm should be counseled regarding the increased risk of local recurrence and may rarely be considered for adjunctive therapy (excision of scrotal scar or radiotherapy) for local control. (Moderate Recommendation; Evidence Level: Grade C)

Testis-Sparing Surgery

11a. TSS through an inguinal incision may be offered as an alternative to radical inguinal orchiectomy in highly selected patients wishing to preserve gonadal function with masses <2cm and (1) equivocal ultrasound/physical exam findings and negative tumor markers (hCG and AFP), (2) congenital, acquired or functionally solitary testis, or (3) bilateral synchronous tumors. (Conditional Recommendation; Evidence Level: Grade C)

11b. Patients considering TSS should be counseled regarding (1) higher risk of local recurrence, (2) need for monitoring with physical examination and ultrasound, (3) role of adjuvant radiotherapy to the testicle to reduce local recurrence, (4) impact of radiotherapy on sperm and testosterone production, and (5) the risk of testicular atrophy and need for testosterone replacement therapy, and/or subfertility/infertility. (Moderate Recommendation; Evidence Level: Grade C)

11c. When TSS is performed, in addition to the suspicious mass, multiple biopsies of the ipsilateral testicle normal parenchyma should be obtained for evaluation by an experienced genitourinary pathologist. (Moderate Recommendation; Evidence Level: Grade C)

GCNIS Counseling and Management

12. Clinicians should inform patients with a history of GCT or GCNIS of risks of a second primary tumor while rare is significantly increased in the contralateral testis. (Moderate Recommendation; Evidence Level: Grade B)

13a. In patients with GCNIS on testis biopsy or malignant neoplasm after TSS, clinicians should inform patients of the risks/benefits of surveillance, radiation, and orchiectomy. (Moderate Recommendation; Evidence Level: Grade C)

13b. Clinicians should recommend surveillance in patients with GCNIS or malignant neoplasm after TSS who prioritize preservation of fertility and testicular androgen production. (Moderate Recommendation; Evidence Level: Grade C)

13c.  Clinicians should recommend testicular radiation (18-20 Gy) or orchiectomy in patients with GCNIS or malignant neoplasm after TSS who prioritize reduction of cancer risk taking into consideration that radiation reduces the risk of hypogonadism compared to orchiectomy. (Moderate Recommendation; Evidence Level: Grade C)

Staging

Serum Tumor Markers

14. Nadir serum tumor markers (AFP, hCG, and LDH) should be repeated at appropriate T1/2 time intervals after orchiectomy for staging and risk stratification. (Moderate Recommendation; Evidence Level: Grade B)

15. For patients with elevated AFP or hCG post-orchiectomy, clinicians should monitor serum tumor markers to establish nadir levels before treatment only if marker nadir levels would influence treatment. (Clinical Principle)

16. For patients with metastatic GCT (Stage IIC or III) requiring chemotherapy, clinicians must base chemotherapy regimen and number of cycles on the IGCCCG risk stratification. IGCCCG risk stratification is based on nadir serum tumor marker (hCG, AFP and LDH) levels obtained prior to the initiation of chemotherapy, staging imaging studies, and tumor histology following radical orchiectomy. (Strong Recommendation; Evidence Level: Grade A) Any post-pubertal male, regardless of age, should be treated according to adult treatment guidelines. (Moderate Recommendation; Evidence Level: Grade B)

17. For patients in whom serum tumor marker (AFP and hCG) levels are borderline elevated (within 3x upper limit of normal) post-orchiectomy, a rising trend should be confirmed before management decisions are made as false-positive elevations may occur. (Clinical Principle)

Imaging

18. In patients with newly diagnosed GCT, clinicians should obtain cross-sectional imaging of the abdomen and pelvis with IV contrast or MRI if CT is contraindicated. (Strong Recommendation; Evidence Level: Grade B)

19a. In patients with newly diagnosed GCT, clinicians must obtain chest imaging. (Clinical Principle)

19b. In the presence of elevated and rising post-orchiectomy markers (hCG and AFP) or evidence of metastases on abdominal/pelvic imaging, chest x-ray or physical exam, a CT chest should be obtained. (Strong Recommendation; Evidence Level: Grade C)

19c. In patients with clinical stage I seminoma, clinicians should preferentially obtain a chest x-ray over a CT scan. (Moderate Recommendation; Evidence Level: Grade B)

19d. In patients with non-seminomatous germ cell tumors (NSGCT), clinicians may preferentially obtain a CT scan of the chest over a chest x-ray and should prioritize CT chest for those patients recommended to receive adjuvant therapy. (Conditional Recommendation; Evidence Level: Grade C)

20. In patients with newly diagnosed GCT, clinicians should not obtain a positron emission tomography (PET) scan for staging. (Strong Recommendation; Evidence Level: Grade B)

21. Patients should be assigned a TNM-s category to guide management decisions. (Strong Recommendation; Evidence Level: Grade B)

Management

Principles of Management

22. Management decisions should be based on imaging obtained within the preceding 4 weeks and serum tumor markers (hCG and AFP) within the preceding 10 days. (Expert Opinion)

23. Management decisions should be made in a multidisciplinary setting involving experienced clinicians in urology, medical oncology, radiation oncology, pathology, and radiology. (Clinical Principle)

24. Expert review of pathologic specimens should be considered in clinical scenarios where treatment decisions will be impacted. (Moderate Recommendation; Evidence Level: Grade C)

25. In patients with normal serum tumor markers (hCG and AFP) and equivocal imaging findings for metastasis, clinicians may consider repeat imaging in six to eight weeks to clarify the extent of disease prior to making a treatment recommendation. (Clinical Principle)

Seminoma Management – Surveillance/RPLND/Chemotherapy/Radiation

26. Clinicians should recommend surveillance after orchiectomy for patients with stage I seminoma. Adjuvant radiotherapy and carboplatin-based chemotherapy are less preferred alternatives. (Strong Recommendation; Evidence Level: Grade B)

27a. For patients with stage IIA or IIB seminoma with a lymph node ≤3cm, clinicians should recommend RT or multi-agent cisplatin-based chemotherapy based on shared decision-making. (Moderate Recommendation; Evidence Level: Grade B)

27b. For patients with stage IIA or IIB seminoma with a lymph node ≤3cm who wish to avoid the long-term toxicities associated with chemotherapy or radiation therapy, RPLND may be offered as an appropriate and effective treatment option. (Moderate Recommendation; Evidence Level: Grade B)

27c. For patients with IIB seminoma with a lymph node >3 cm, chemotherapy is recommended. (Moderate Recommendation; Evidence Level: Grade B)

Non Seminoma Management – Surveillance/RPLND/Chemotherapy/Radiation

28. Clinicians should recommend risk-appropriate, multi-agent chemotherapy for patients with NSGCT with elevated and rising post-orchiectomy serum AFP or hCG (i.e. stage TanyN1-2S1). (Strong Recommendation; Evidence Level: Grade B)

29. Clinicians should recommend surveillance for patients with stage IA NSGCT. RPLND or one cycle of bleomycin, etoposide, and cisplatin chemotherapy are effective and appropriate alternative treatment options for patients who decline surveillance or are at risk for non-compliance. (Moderate Recommendation; Evidence Level: Grade B)

30. For patients with stage IB NSGCT, clinicians should recommend surveillance, RPLND, or one or two cycles of bleomycin, etoposide, and cisplatin chemotherapy based on shared decision-making. (Strong Recommendation; Evidence Level: Grade B) 

31. Patients with stage I NSGCT and any secondary somatic malignancy (also known as teratoma with malignant transformation) in the primary tumor at orchiectomy should undergo RPLND. (Expert Opinion)

32. Clinicians should recommend RPLND or chemotherapy for patients with stage IIA NSGCT with normal post-orchiectomy serum (S0) AFP and hCG. (Moderate Recommendation; Evidence Level: Grade B)

33. In patients with clinical stage IIB NSGCT and normal post-orchiectomy serum AFP and hCG, clinicians should recommend risk-appropriate, multi-agent chemotherapy. (Moderate Recommendation; Evidence Level: Grade B). Clinicians may offer RPLND as an alternative to chemotherapy to select patients with clinical stage IIB NSGCT with normal post-orchiectomy serum AFP and hCG. (Conditional Recommendation; Evidence Level: Grade C)

34. Among patients who are candidates for RPLND, it is recommended clinicians consider referral to an experienced surgeon at a high-volume center. (Moderate Recommendation; Evidence Level: Grade C)

35. Surgeons with experience in the management of GCT and expertise in minimally invasive surgery may offer a minimally-invasive RPLND, acknowledging the lack of long-term data on oncologic outcomes. (Expert Opinion)

36. Primary RPLND should be performed with curative intent in all patients. RPLND should be performed with adherence to the following anatomical principles, regardless of the intent to administer adjuvant chemotherapy. These principles are applied to both open and minimally-invasive approaches. (Moderate Recommendation; Evidence Level: Grade B)

  • A full bilateral template dissection should be performed in patients with suspicious lymph nodes based on CT imaging or intraoperative assessment and in those with somatic-type malignancy in the primary tumor.
  • A full bilateral template or modified template dissection may be performed in patients with clinically negative lymph nodes.
  • A right modified template dissection may omit the para-aortic lymph nodes below the inferior mesenteric artery. Omission of para-aortic lymph nodes above the inferior mesenteric artery is controversial.
  • A left modified template dissection may omit paracaval, precaval, and retrocaval lymph nodes. Omission of interaortocaval lymph nodes is controversial.
  • Nerve-sparing should be offered in select patients desiring preservation of ejaculatory function.
  • Nerve-sparing attempts should not compromise the quality of the lymph node dissection.
  • A complete retroaortic and/or retrocaval lymph node dissection with division of lumbar vessels should be performed when within the planned template.
  • The ipsilateral gonadal vessels should be removed in all patients.
  • The cephalad extent of the dissection is the crus of the diaphragm to the level of the renal arteries. The caudad extent of disease is the crossing of the ureter over the ipsilateral common iliac artery.

37.  After primary RPLND, clinicians should recommend surveillance or adjuvant chemotherapy in patients with NSGCT who have pathological stage II disease that is not pure teratoma. (Moderate Recommendation; Evidence Level: Grade B)

  • For patients with pN1 and/or pN1-3 pure teratoma, surveillance is preferred.
  • For patients with pN2-3 at RPLND, multi-agent cisplatin-based chemotherapy is preferred.

Surveillance for Stage I Testicular Cancer

38. For patients with clinical stage I seminoma choosing surveillance, clinicians should obtain a history and physical examination and perform cross-sectional imaging of the abdomen with or without the pelvis, every 6 months for the first 2 years, and then every 6-12 months in years 3-5. Routine surveillance imaging of the chest and serum tumor marker assessment can be obtained as clinically indicated. (Strong Recommendation; Evidence Level: Grade B)

39. In patients with stage I NSGCT undergoing surveillance after orchiectomy, clinicians should perform a physical examination and obtain serum tumor markers (AFP, hCG +/- LDH) every 2-3 months in year 1, every 2-4 months in year 2, every 4-6 months in year 3, and every 6-12 months for years 4 and 5. (Moderate Recommendation; Evidence Level: Grade C)

40. In patients with stage I NSGCT undergoing surveillance after orchiectomy, radiologic assessment (chest x-ray and imaging of the abdomen with or without the pelvis) should be obtained every 3-6 months in year 1 starting at 3 months, every 4-12 months in year 2, once in year 3, and once in year 4 or 5. (Moderate Recommendation; Evidence Level: Grade B) Men at higher risk of relapse (e.g., lymphovascular invasion) should be imaged with shorter intervals. (Expert Opinion)

41. Patients who relapse on surveillance should be fully restaged and treated based on their TNM-s status. (Moderate Recommendation; Evidence Level: Grade C)

42. Clinicians should inform patients with stage I GCT on surveillance of the ≤1% risk of late relapse after 5 years. (Moderate Recommendation; Evidence Level: Grade B) Annual serologic and radiographic assessment may be performed thereafter as indicated based upon clinical concerns. (Clinical Principle)

Additional Survivorship

43. Clinicians should refer patients to a survivorship clinic appreciating the long-term risks and potential sequelae of prior treatment among patients with GCT, with the integration of screening and monitoring for potential medical issues which may arise (Expert Opinion) including:

  • Monitoring for signs and symptoms of hypogonadism. If present, serum AM testosterone and luteinizing hormone (LH) levels should be measured.
  • Patients with a history of GCT whose treatment has included radiation therapy, chemotherapy, or both should be advised of the elevated risk of cardiovascular disease and should establish regular care with a primary care physician so that modifiable risk factors for cardiovascular disease (e.g., diet, exercise, smoking, serum lipid levels, blood pressure, serum glucose) can be monitored.
  • Patients with a history of GCT whose treatment has included radiation therapy, chemotherapy, or both should be advised of the elevated risk of secondary malignancy and should establish regular care with a primary care physician for appropriate health care maintenance and cancer screening as appropriate.

INTRODUCTION

Purpose

Testis cancer is the most common solid malignancy in young males. The vast majority of men with testis cancer have low-stage disease (limited to the testis and retroperitoneum; clinical stages I-IIB); survival rates are high with standard therapy. A priority for those patients with low-stage disease is limiting the burden of therapy and treatment-related toxicity without compromising cancer control. Thus, surveillance has assumed an increasing role among those with cancer clinically confined to the testis. Testis cancer is a relatively rare malignancy, with outcomes defined by specific cancer- and patient-related factors. Likewise, paradigms for management have undergone substantial change in recent years as evidence regarding risk stratification, recurrence, survival, and treatment-related toxicity has emerged.

Urologists are frequently the initial treating clinician for men with newly diagnosed testis cancer and thus play a crucial role in counseling and treatment decision making. This clinical practice guideline provides evidence-based recommendations for clinicians regarding the diagnosis, staging, treatment selection, and post-treatment surveillance of patients with clinical stages I, IIA, and IIB seminoma and nonseminomatous germ cell tumor (NSGCT). Please also refer to the associated Low-Stage Testis Cancer Treatment Algorithm.

Methodology

Panel Formation and Process

The Testicular Cancer Panel was created in 2017 by the American Urological Association Education and Research, Inc. (AUAER). The Practice Guidelines Committee (PGC) of the AUA selected the Panel Chairs who in turn appointed the additional panel members based on specific expertise in this area. The Panel included specialties from urology, oncology, and radiology. In 2023, an update review assessing abstracts from new studies published since the publication of the 2019 Guideline was completed.

Peer Review and Document Approval

An integral part of the guideline development process at the AUA is external peer review. The AUA conducted a thorough peer review process to ensure that the document was reviewed by experts in the diagnosis and treatment of testicular cancer. In addition to reviewers from the AUA PGC, Science and Quality Council (SQC), and Board of Directors (BOD), the document was reviewed by representatives from American Society of Clinical Oncology, American Society for Radiation Oncology, and Society of Urologic Oncology as well as external content experts. Additionally, a call for reviewers was placed on the AUA website from December 14-28, 2018 to allow any additional interested parties to request a copy of the document for review. The guideline was also sent to the Urology Care Foundation to open the document further to the patient perspective. The draft guideline document was distributed to 105 peer reviewers. All peer review comments were blinded and sent to the Panel for review. In total, 45 reviewers provided comments, including 30 external reviewers.  At the end of the peer review process, a total of 530 comments were received. Following comment discussion, the Panel revised the draft as needed. Once finalized, the guideline was submitted for approval to the AUA PGC, SQC and BOD for final approval.

In 2023, as a part of the amendment process, the AUA conducted a thorough peer review process. A call for peer reviewers was posted in April 2023 and the draft guideline document was distributed to 62 peer reviewers, 16 of which submitted comments. The Amendment Panel reviewed and discussed all submitted comments and revised the draft as needed. Once finalized, the guideline was submitted for approval to the original guideline panel, the PGC and SQC. It was then submitted to AUA BODs for final approval. Panel members received no renumeration for their work.

Search Strategy

The Johns Hopkins University Evidence-based Practice Center team searched PubMed®, Embase®, and the Cochrane Central Register of Controlled Trials (CENTRAL) from January 1980 through August 2018. Searches were updated using the same methodological protocol to capture literature published through March 2023. The team developed a search strategy by using medical subject headings terms and key words relevant to the diagnosis and treatment of early-stage testicular cancer. The evidence review team also reviewed relevant systematic reviews and references provided by the Panel to identify articles that may have been missed by the database searches.

Study Selection and Data Abstraction

Study selection was based on predefined eligibility criteria for the patient populations, interventions, outcomes, and study designs of interest. Two reviewers independently screened titles, abstracts, and full text for inclusion. Differences between reviewers regarding eligibility were resolved through consensus.

Reviewers extracted information on study characteristics, participants, interventions, and outcomes. One reviewer completed data abstraction, and a second reviewer checked for accuracy.

Assessment of Risk of Bias (ROB) and Data Extraction

Two reviewers independently assessed risk of bias for individual studies. The Cochrane Collaboration’s tool was used for assessing the risk of bias of randomized controlled trials (RCTs).1 For non-randomized studies of treatment interventions, the reviewers used the Cochrane Risk of Bias Assessment Tool for Non-Randomized Studies of Interventions (ACROBAT-NRSI). For diagnostic studies, reviewers used the quality assessment tool for diagnostic accuracy studies (QUADAS -2).2 Differences between reviewers were resolved through consensus. The evidence review team graded strength of evidence on outcomes by adapting the AUA’s three predefined levels of strength of evidence.

Determination of Evidence Strength

The categorization of evidence strength is conceptually distinct from the quality of individual studies. Evidence strength refers to the body of evidence available for a particular question and includes not only the quality of individual studies but consideration of study design; consistency of findings across studies; adequacy of sample sizes; and generalizability of study populations, settings, and interventions for the purposes of the guideline. The AUA categorizes body of evidence strength as Grade A (well-conducted and highly-generalizable RCTs or exceptionally strong observational studies with consistent findings), Grade B (RCTs with some weaknesses of procedure or generalizability or moderately strong observational studies with consistent findings), or Grade C (RCTs with serious deficiencies of procedure or generalizability or extremely small sample sizes or observational studies that are inconsistent, have small sample sizes, or have other problems that potentially confound interpretation of data). By definition, Grade A evidence has a high level of certainty, Grade B evidence has a moderate level of certainty, and Grade C evidence has a low level of certainty (Table 1).3

AUA Nomenclature: Linking Statement Type to Evidence Strength

The AUA nomenclature system explicitly links statement type to body of evidence strength, level of certainty, magnitude of benefit or risk/burdens, and the Panel’s judgment regarding the balance between benefits and risks/burdens (Table 2). Strong Recommendations are directive statements that an action should (benefits outweigh risks/burdens) or should not (risks/burdens outweigh benefits) be undertaken because net benefit or net harm is substantial. Moderate Recommendations are directive statements that an action should (benefits outweigh risks/burdens) or should not (risks/burdens outweigh benefits) be undertaken because net benefit or net harm is moderate. Conditional Recommendations are non-directive statements used when the evidence indicates there is no apparent net benefit or harm or when the balance between benefits and risks/burden is unclear. All three statement types may be supported by any body of evidence strength grade. Body of evidence strength Grade A in support of a Strong or Moderate Recommendation indicates the statement can be applied to most patients in most circumstances and that future research is unlikely to change confidence. Body of evidence strength Grade B in support of a Strong or Moderate Recommendation indicates the statement can be applied to most patients in most circumstances, but better evidence could change confidence. Body of evidence strength Grade C in support of a Strong or Moderate Recommendation indicates the statement can be applied to most patients in most circumstances, but better evidence is likely to change confidence. Body of evidence strength Grade C is only rarely used in support of a Strong Recommendation. Conditional Recommendations can also be supported by any evidence strength. When body of evidence strength is Grade A, the statement indicates benefits and risks/burdens appear balanced, the best action depends on patient circumstances, and future research is unlikely to change confidence. When body of evidence strength Grade B is used, benefits and risks/burdens appear balanced, the best action also depends on individual patient circumstances, and better evidence could change confidence. When body of evidence strength Grade C is used, there is uncertainty regarding the balance between benefits and risks/burdens, alternative strategies may be equally reasonable, and better evidence is likely to change confidence.

Where gaps in the evidence existed, Clinical Principles or Expert Opinions are provided via consensus of the Panel. A Clinical Principle is a statement about a component of clinical care widely agreed upon by urologists or other clinicians for which there may or may not be evidence in the medical literature. Expert Opinion refers to a statement based on members' clinical training, experience, knowledge, and judgment for which there may or may not be evidence in the medical literature.

Table 1: Strength of Evidence Definitions

TABLE 1: STRENGTH OF EVIDENCE DEFINITIONS

Table 2: AUA Nomenclature Linking Statement Type to Level of Certainty, Magnitude of Benefit or Risk/Burden, and Body of Evidence Strength

Table 2: AUA Nomenclature Linking Statement Type to Level of Certainty, Magnitude of Benefit or Risk/Burden, and Body of Evidence Strength

BACKGROUND

Epidemiology

In 2019, an estimated 9,500 men will be diagnosed with testis cancer in the United States, and 400 will die from the disease.4 Testis cancer is the most common solid malignancy among men aged 20 to 40 years. The incidence rate is highest among Caucasians, lowest among African-Americans, and most rapidly increasing in Hispanic populations.5,6  Age-adjusted incidence has nearly doubled over the last 4 decades for unknown reasons, from 3.7 per 100,000 in 1975 to 6.4 per 100,000 in 2014.5 A stage migration of GCT has been observed, presumably due to increased awareness and earlier diagnosis. Between 1973 and 2014, the percentage of tumors diagnosed at a localized stage increased from 55% to 68% in the United States. Currently, less than 15% of men present with stage III disease (to the lungs, viscera, or non-regional lymph nodes).

Risk factors for developing testis cancer include germ cell neoplasia in situ (GCNIS), history of undescended testis (UDT)/ cryptorchidism, family history, and a personal history of testis cancer. Infertility is associated with the presence of GCT, though this association is thought to arise from inherent testicular dysfunction.7,8,9 GCNIS is the precursor lesion from which the majority of GCTs arise. Among men with invasive GCT, GCNIS is found in adjacent testicular parenchyma in 80-90%. Among men with GCNIS, the risk of developing invasive GCT is approximately 50% within 5 years.10 Men with cryptorchidism have a four to six fold increased risk of developing testis cancer in the affected testicle, but the relative risk (RR) falls to two to three fold if orchiopexy is performed before puberty.11,12 Studies assessing the cancer risk of UDT in the contralateral testis are conflicting, though a meta-analysis of cryptorchidism studies showed the contralateral descended testis is also at slightly increased risk of developing cancer (RR 1.74; 95% Confidence Interval [CI], 1.01 to 2.98).13 Men with a first-degree relative with GCT are at an increased risk of developing testis cancer and at an earlier age.14 Men with a personal history of testis cancer are at a 12-fold increased risk of developing GCT in the contralateral testis, but the 15-year cumulative incidence is only 2%.15

Etiology

GCNIS arises from transformed primordial germ cells that develop in utero or early infancy that lay dormant until puberty when they are stimulated by increased serum LH and/or testosterone levels.16 The carcinogenesis of GCNIS and testis cancer is poorly understood. The increase in testis cancer incidence along with other male reproductive disorders (e.g., infertility, hypospadias, UDT) suggests that GCT may arise from ‘testicular dysgenesis,’ which results from a combination of environmental and/or lifestyle factors (possibly from exposure in utero) in combination with genetic susceptibility. The role of genetic factors is supported by the clustering of testis cancer in some families, the extreme difference in the rate of testis cancer in black and white Americans, and the finding of susceptibility loci on chromosomes 5, 6, and 12.14 Additionally, polymorphisms of certain genes, including the gene encoding c-KIT ligand, have been associated with an increased risk of testis cancer.17 Gonocytes depend on KIT ligand for survival; the gene for this protein is located on the long arm of chromosome 12, where an increased number of copies is a universal finding in adult GCT.18 Thus, a connection between mutations or polymorphisms in c-KIT ligand and GCT has biological plausibility. Inherited alterations to susceptibility genes involved in DNA repair may contribute to the development of adult GCT. A multicenter case-control gene-level enrichment analysis of germline pathogenic variants in individuals with GCT relative to cancer-free controls found 22 pathogenic germline DRG variants, one-third of which were in CHEK2. The variant CHEK2 allele was found in 9.8% of cases and associated with a four-fold increased risk of GCT.19

Histological Classification

The histological classification of post-pubertal GCT is outlined in Table 3.16 GCT are broadly classified as GCNIS-derived (germ-cell) and non-GCNIS derived (non-germ cell). The vast majority of post-pubertal GCT are GCNIS-derived. GCT are divided into seminoma and NSGCT, with relative distribution of 52-56% and 44-48%, respectively.20 NSGCT includes embryonal carcinoma, yolk sac tumor, teratoma, and choriocarcinoma subtypes, either alone as pure forms or in combination as mixed GCT with or without seminoma. Most NSGCTs are composed of two or more GCT subtypes (mixed tumors). GCTs that contain both NSGCT subtypes and seminoma are classified as NSGCT even if the NSGCT component represents a tiny proportion of the tumor.

The classification of GCT into seminoma and NSGCT has histological, biological, and practical implications. Compared to NSGCT, pure seminomas tend to develop at a later age, are of lower stage at diagnosis, and grow at a slower rate.21 The risk of occult systemic disease for stage I disease is lower for seminoma than for NSGCT. Lastly, pure seminomas are even more highly sensitive to chemotherapy relative to NSGCTs and sensitive to radiation therapy. All these differences have important treatment implications. Among NSGCT, embryonal carcinoma is the most undifferentiated cell type and has totipotential capacity to differentiate into other NSGCT cell types (yolk sac, choriocarcinoma, and teratoma) within the primary tumor and at metastatic sites.

Among NSGCT patients, the potential for teratoma to arise within the primary tumor or at metastatic sites has important management implications. Though histologically benign, teratomas contain many genetic abnormalities frequently found in malignant GCT elements.22,23 Teratoma typically grows slowly or may be indolent. However, their underlying genetic instability may lead to uncontrollable growth and invasion of surrounding structures (growing teratoma syndrome)24 or transformation into somatic-type malignancies such as sarcoma or adenocarcinoma.25 Unlike other GCT subtypes, teratoma is universally resistant to chemotherapy and only curable by surgical resection. This has important implications in treatment selection for all stages of NSGCT. 

Table 3: 2016 World Health Organization Classification of Germ Cell Tumors of the Testis16

Serum Tumor Markers

Testis cancer is one of the few malignancies with reliable serum tumor markers (alpha-fetoprotein [AFP], human chorionic gonadotropin [hCG], and lactate dehydrogenase [LDH]) that are essential for diagnosis, prognosis, clinical staging, management, response to therapy, and post-treatment surveillance. AFP is produced by yolk sac and embryonal carcinoma and is elevated in 10-40% of low-stage (clinical stages I, IIA, IIB) NSGCT.26 Choriocarcinoma and seminoma do not produce AFP. Patients with pure seminoma in the primary tumor with an elevated serum AFP are considered to have NSGCT. The half-life of AFP is five to seven days. Other malignant sources of AFP include cancers of the stomach, pancreas, biliary tract, liver, and lung. Non-malignant sources of AFP include liver disease (infectious, drug-induced, alcohol-induced, autoimmune), ataxia telangiectasia, hereditary tyrosinemia, and heterophile antibodies.27,28 Hereditary persistence of AFP (HPAFP), a congenital alteration in the hepatic nuclear factor binding site of the AFP gene leads to increased AFP transcription and is a rare cause of elevated AFP.29  Despite most laboratories considering an AFP level of >8 ng/mL to be abnormally elevated, a proportion of the population may have levels up to 15 or 25 ng/mL in the absence of any pathology.7 Treatment decisions soley based on “elevated” AFP levels that are stable and <25 ng/mL are discouraged.

hCG levels are elevated in 10-30% of low-stage NSGCT and 10-15% of seminomas.26 hCG is secreted by choriocarcinoma, embryonal carcinoma, and syncytiotrophoblastic cells found in 10-15% of seminomas. The half-life of hCG is 24-36 hours. hCG levels may be elevated in cancers of the liver, biliary tract, pancreas, stomach, lung, breast, kidney and bladder. The alpha-subunit of hCG is common to several pituitary tumors, thus immunoassays for hCG are directed at the beta-subunit. Similarly, heterophile antibodies, hypogonadism, and possibly some medications can lead to false-positive elevations of hCG.30-34

LDH levels are the least relevant and clinically applicable of the serum tumors markers and elevated in approximately 20% of low-stage GCT.26 LDH is expressed in smooth, cardiac, and skeletal muscle and can be elevated from cancerous (e.g., kidney, lymphoma, GI, breast) or non-cancerous conditions (e.g., heart failure, anemia, HIV). Of the five isoenzymes of LDH, LDH-1 is the most frequently elevated isoenzyme in GCT. The magnitude of LDH elevation correlates with bulk of disease. As a non-specific marker, its main GCT use is in the prognostic assessment at diagnosis. Treatment decisions based solely on LDH elevation in the setting of normal AFP and hCG should be discouraged.

Prognosis and Staging

Prognosis and initial management decisions are dictated by clinical stage, which is based on the pathological stage of the primary tumor, post-orchiectomy serum tumor marker levels, and staging as determined by physical examination and imaging. In 1997, an international consensus for GCT staging was developed by the American Joint Committee on Cancer (AJCC) and Union Internationale Contre le Cancer (UICC) (Table 4).35 The AJCC and UICC staging systems for GCT are unique because, for the first time, a serum tumor marker category (S) based on post-orchiectomy AFP, hCG, and LDH levels was used to supplement the prognostic stages (Table 5). Clinical stage I is defined as disease clinically confined to the testis, clinical stage II indicates regional (retroperitoneal) lymph node metastasis, and clinical stage III represents non-regional lymph node, lung and/or visceral metastasis, although post-orchiectomy serum tumor marker levels can upstage patients from clinical stage II to III.

For patients with low-stage GCT (clinical stage I, IIA, or IIB), long-term survival is 95% or better. As GCT patients are often healthy and young with long estimated life expectancy, reducing the burden of therapy and treatment-related toxicity are particularly important.

Table 4: Stages of Testicular Cancer 35

Table 5: Tumor Marker S Designation Levels35

GUIDELINE STATEMENTS

Initial Management

Diagnosis and Initial Consultation

Guideline Statement 1

A solid mass in the testis identified by physical exam or imaging should be managed as a malignant neoplasm until proven otherwise. (Clinical Principle)

Discussion


Guideline Statement 2

In a man with a solid mass in the testis suspicious for malignant neoplasm, serum tumor markers (AFP, hCG, and LDH) should be drawn and measured prior to any treatment, including orchiectomy. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 3

Prior to definitive management, patients should be counseled about the risks of hypogonadism and infertility (Moderate Recommendation; Evidence Level: Grade C) and should be offered sperm banking, when appropriate. In patients without a normal contralateral testis or with known subfertility, this should be considered prior to orchiectomy. (Clinical Principle)

Discussion


Guideline Statement 4

Scrotal ultrasound with Doppler should be obtained in patients with a unilateral or bilateral scrotal mass suspicious for neoplasm. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 5

Testicular microlithiasis in the absence of solid mass and risk factors for developing a germ cell tumor (GCT) does not confer an increased risk of malignant neoplasm and does not require further evaluation. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 6

Patients with normal serum tumor markers (hCG and AFP) and indeterminate findings on physical exam or testicular ultrasound for testicular neoplasm should undergo repeat imaging in six to eight weeks. (Clinical Principle)

Discussion


Guideline Statement 7

Magnetic Resonance Imaging (MRI) should not be used in the initial evaluation and diagnosis of a testicular lesion suspicious for neoplasm. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Orchiectomy

Guideline Statement 8

Patients with a testicular lesion suspicious for malignant neoplasm and a normal contralateral testis should undergo a radical inguinal orchiectomy; testis-sparing surgery (TSS) is not recommended. Transscrotal orchiectomy is discouraged. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 9

Testicular prosthesis should be discussed prior to orchiectomy. (Expert Opinion)

Discussion


Guideline Statement 10

Patients who have undergone scrotal orchiectomy for malignant neoplasm should be counseled regarding the increased risk of local recurrence and may rarely be considered for adjunctive therapy (excision of scrotal scar or radiotherapy) for local control. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Testis-Sparing Surgery

Guideline Statement 11

11a. TSS through an inguinal incision may be offered as an alternative to radical inguinal orchiectomy in highly selected patients wishing to preserve gonadal function with masses <2cm and (1) equivocal ultrasound/physical exam findings and negative tumor markers (hCG and AFP), (2) congenital, acquired or functionally solitary testis, or (3) bilateral synchronous tumors. (Conditional Recommendation; Evidence Level: Grade C)

11b.  Patients considering TSS should be counseled regarding (1) higher risk of local recurrence, (2) need for monitoring with physical examination and ultrasound, (3) role of adjuvant radiotherapy to the testicle to reduce local recurrence, (4) impact of radiotherapy on sperm and testosterone production, and (5) the risk of testicular atrophy and need for testosterone replacement therapy, and/or subfertility/infertility. (Moderate Recommendation; Evidence Level: Grade C)

11c. When TSS is performed, in addition to the suspicious mass, multiple biopsies of the ipsilateral testicle normal parenchyma should be obtained for evaluation by an experienced genitourinary pathologist. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


GCNIS Counseling and Management

Guideline Statement 12

Clinicians should inform patients with a history of GCT or GCNIS of risks of a second primary tumor while rare is significantly increased in the contralateral testis. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 13

13a. In patients with GCNIS on testis biopsy or malignant neoplasm after TSS, clinicians should inform patients of the risks/benefits of surveillance, radiation, and orchiectomy. (Moderate Recommendation; Evidence Level: Grade C)

13b. Clinicians should recommend surveillance in patients with GCNIS or malignant neoplasm after TSS who prioritize preservation of fertility and testicular androgen production. (Moderate Recommendation; Evidence Level: Grade C)

13c. Clinicians should recommend testicular radiation (18-20 Gy) or orchiectomy in patients with GCNIS or malignant neoplasm after TSS who prioritize reduction of cancer risk taking into consideration that radiation reduces the risk of hypogonadism compared to orchiectomy. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Staging

Serum Tumor Markers

Guideline Statement 14

Nadir serum tumor markers (AFP, hCG, and LDH) should be repeated at appropriate T1/2 time intervals after orchiectomy for staging and risk stratification. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 15

For patients with elevated AFP or hCG post-orchiectomy, clinicians should monitor serum tumor markers to establish nadir levels before treatment only if marker nadir levels would influence treatment. (Clinical Principle)

Discussion


Guideline Statement 16

For patients with metastatic GCT (Stage IIC or III) requiring chemotherapy, clinicians must base chemotherapy regimen and number of cycles on the IGCCCG risk stratification. IGCCCG risk stratification is based on nadir serum tumor marker (hCG, AFP and LDH) levels obtained prior to the initiation of chemotherapy, staging imaging studies, and tumor histology following radical orchiectomy (Strong Recommendation; Evidence Level: Grade A). Any post-pubertal male, regardless of age, should be treated according to adult treatment guidelines. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 17

For patients in whom serum tumor marker (AFP and hCG) levels are borderline elevated (within 3x upper limit of normal) post-orchiectomy, a rising trend should be confirmed before management decisions are made as false-positive elevations may occur. (Clinical Principle)

Discussion


Imaging

Guideline Statement 18

In patients with newly diagnosed GCT, clinicians should obtain cross-sectional imaging of the abdomen and pelvis with IV contrast or MRI if CT is contraindicated. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 19

19a. In patients with newly diagnosed GCT, clinicians must obtain chest imaging. (Clinical Principle)

19b. In the presence of elevated and rising post-orchiectomy markers (hCG and AFP) or evidence of metastases on abdominal/pelvic imaging, chest x-ray or physical exam, a CT chest should be obtained. (Strong Recommendation; Evidence Level: Grade C)

19c. In patients with clinical stage I seminoma, clinicians should preferentially obtain a chest x-ray over a CT scan. (Moderate Recommendation; Evidence Level: Grade B)

19d. In patients with non-seminomatous germ cell tumors (NSGCT), clinicians may preferentially obtain a CT scan of the chest over a chest x-ray and should prioritize CT chest for those patients recommended to receive adjuvant therapy. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 20

In patients with newly diagnosed GCT, clinicians should not obtain a positron emission tomography (PET) scan for staging. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 21

Patients should be assigned a TNM-s category to guide management decisions. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Management

Principles of Management

Guideline Statement 22

Management decisions should be based on imaging obtained within the preceding 4 weeks and serum tumor markers (hCG and AFP) within the preceding 10 days. (Expert Opinion)

Discussion


Guideline Statement 23

Management decisions should be made in a multidisciplinary setting involving experienced clinicians in urology, medical oncology, radiation oncology, pathology, and radiology. (Clinical Principle)

Discussion


Guideline Statement 24

Expert review of pathologic specimens should be considered in clinical scenarios where treatment decisions will be impacted. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 25

In patients with normal serum tumor markers (hCG and AFP) and equivocal imaging findings for metastasis, clinicians may consider repeat imaging in six to eight weeks to clarify the extent of disease prior to making a treatment recommendation. (Clinical Principle)

Discussion


Seminoma Management ā€“ Surveillance/RPLND/Chemotherapy/Radiation

Guideline Statement 26

Clinicians should recommend surveillance after orchiectomy for patients with stage I seminoma. Adjuvant radiotherapy and carboplatin-based chemotherapy are less preferred alternatives. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 27

27a. For patients with stage IIA or IIB seminoma with a lymph node ≤3cm, clinicians should recommend RT or multi-agent cisplatin-based chemotherapy based on shared decision-making. (Moderate Recommendation; Evidence Level: Grade B)

27b. For patients with stage IIA or IIB seminoma with a lymph node ≤3cm who wish to avoid the long-term toxicities associated with chemotherapy or radiation therapy, RPLND may be offered as an appropriate and effective treatment option. (Moderate Recommendation; Evidence Level: Grade B)

27c. For patients with IIB seminoma with a lymph node >3 cm, chemotherapy is recommended. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Non Seminoma Management ā€“ Surveillance/RPLND/Chemotherapy/Radiation

Guideline Statement 28

Clinicians should recommend risk-appropriate, multi-agent chemotherapy for patients with NSGCT with elevated and rising post-orchiectomy serum AFP or hCG (i.e. stage TanyN1-2S1). (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 29

Clinicians should recommend surveillance for patients with stage IA NSGCT. RPLND or one cycle of bleomycin, etoposide, and cisplatin chemotherapy are effective and appropriate alternative treatment options for patients who decline surveillance or are at risk for non-compliance. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 30

For patients with stage IB NSGCT, clinicians should recommend surveillance, RPLND, or one or two cycles of bleomycin, etoposide, and cisplatin chemotherapy based on shared decision-making. (Strong Recommendation; Evidence Level: Grade B) 

Discussion


Guideline Statement 31

Patients with stage I NSGCT and any secondary somatic malignancy (also known as teratoma with malignant transformation) in the primary tumor at orchiectomy should undergo RPLND. (Expert Opinion)

Discussion


Guideline Statement 32

Clinicians should recommend RPLND or chemotherapy for patients with stage IIA NSGCT with normal post-orchiectomy serum (S0) AFP and hCG. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 33

In patients with clinical stage IIB NSGCT and normal post-orchiectomy serum AFP and hCG, clinicians should recommend risk-appropriate, multi-agent chemotherapy. (Moderate Recommendation; Evidence Level: Grade B). Clinicians may offer RPLND as an alternative to chemotherapy to select patients with clinical stage IIB NSGCT with normal post-orchiectomy serum AFP and hCG. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 34

Among patients who are candidates for RPLND, it is recommended clinicians consider referral to an experienced surgeon at a high-volume center. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 35

Surgeons with experience in the management of GCT and expertise in minimally invasive surgery may offer a minimally-invasive RPLND, acknowledging the lack of long-term data on oncologic outcomes. (Expert Opinion)

Discussion


Guideline Statement 36

Primary RPLND should be performed with curative intent in all patients. RPLND should be performed with adherence to the following anatomical principles, regardless of the intent to administer adjuvant chemotherapy. These principles are applied to both open and minimally-invasive approaches. (Moderate Recommendation; Evidence Level: Grade B)

  • A full bilateral template dissection should be performed in patients with suspicious lymph nodes based on CT imaging or intraoperative assessment and in those with somatic-type malignancy in the primary tumor.
  • A full bilateral template or modified template dissection may be performed in patients with clinically negative lymph nodes.
  • A right modified template dissection may omit the para-aortic lymph nodes below the inferior mesenteric artery. Omission of para-aortic lymph nodes above the inferior mesenteric artery is controversial.
  • A left modified template dissection may omit paracaval, precaval, and retrocaval lymph nodes. Omission of interaortocaval lymph nodes is controversial.
  • Nerve-sparing should be offered in select patients desiring preservation of ejaculatory function.
  • Nerve-sparing attempts should not compromise the quality of the lymph node dissection.
  • A complete retroaortic and/or retrocaval lymph node dissection with division of lumbar vessels should be performed when within the planned template.
  • The ipsilateral gonadal vessels should be removed in all patients.
  • The cephalad extent of the dissection is the crus of the diaphragm to the level of the renal arteries. The caudad extent of disease is the crossing of the ureter over the ipsilateral common iliac artery.

Discussion


Guideline Statement 37

After primary RPLND, clinicians should recommend surveillance or adjuvant chemotherapy in patients with NSGCT who have pathological stage II disease that is not pure teratoma. (Moderate Recommendation; Evidence Level: Grade B)

  • For patients with pN1 and/or pN1-3 pure teratoma, surveillance is preferred.
  • For patients with pN2-3 at RPLND, multi-agent cisplatin-based chemotherapy is preferred.

Discussion


Surveillance for Stage I Testicular Cancer

Guideline Statement 38

For patients with clinical stage I seminoma choosing surveillance, clinicians should obtain a history and physical examination and perform cross-sectional imaging of the abdomen with or without the pelvis, every 6 months for the first 2 years, and then every 6-12 months in years 3-5. Routine surveillance imaging of the chest and serum tumor marker assessment can be obtained as clinically indicated. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 39

In patients with stage I NSGCT undergoing surveillance after orchiectomy, clinicians should perform a physical examination and obtain serum tumor markers (AFP, hCG +/- LDH) every 2-3 months in year 1, every 2-4 months in year 2, every 4-6 months in year 3, and every 6-12 months for years 4 and 5. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 40

In patients with stage I NSGCT undergoing surveillance after orchiectomy, radiologic assessment (chest x-ray and imaging of the abdomen with or without the pelvis) should be obtained every 3-6 months in year 1 starting at 3 months, every 4-12 months in year 2, once in year 3, and once in year 4 or 5. (Moderate Recommendation; Evidence Level: Grade B) Men at higher risk of relapse (e.g., lymphovascular invasion) should be imaged with shorter intervals. (Expert Opinion)

Discussion


Guideline Statement 41

Patients who relapse on surveillance should be fully restaged and treated based on their TNM-s status. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 42

Clinicians should inform patients with stage I GCT on surveillance of the ≤1% risk of late relapse after 5 years. (Moderate Recommendation; Evidence Level: Grade B) Annual serologic and radiographic assessment may be performed thereafter as indicated based upon clinical concerns. (Clinical Principle)

Discussion


Additional Survivorship

Guideline Statement 43

Clinicians should refer patients to a survivorship clinic appreciating the long-term risks and potential sequelae of prior treatment among patients with GCT, with the integration of screening and monitoring for potential medical issues which may arise (Expert Opinion) including:

  • Monitoring for signs and symptoms of hypogonadism. If present, serum AM testosterone and luteinizing hormone (LH) levels should be measured.
  • Patients with a history of GCT whose treatment has included radiation therapy, chemotherapy, or both should be advised of the elevated risk of cardiovascular disease and should establish regular care with a primary care physician so that modifiable risk factors for cardiovascular disease (e.g., diet, exercise, smoking, serum lipid levels, blood pressure, serum glucose) can be monitored.
  • Patients with a history of GCT whose treatment has included radiation therapy, chemotherapy, or both should be advised of the elevated risk of secondary malignancy and should establish regular care with a primary care physician for appropriate health care maintenance and cancer screening as appropriate.

Discussion


FUTURE DIRECTIONS

Biomarkers for Micrometastases in Low-Stage GCT

Practice patterns of patients with clinical stage I seminoma and NSGCT indicate a substantial shift towards surveillance, even among those with risk factors.229 Lymphovascular invasion is the only parameter that reliably identifies patients at risk for relapse among patients with clinical stage I NSGCT, and the risk of lymphovascular invasion varies from 35-55%.230,211 Thus, patients who relapse following surveillance are exposed to treatment intensification. Likewise, adjuvant therapy exposes a substantial proportion of patients to treatment and its associated toxicity who were otherwise cured by orchiectomy. Lastly, despite changes to surveillance protocols, patients on surveillance are subject to intensive monitoring. Circulating biomarkers that reliably identify the presence of residual disease may be helpful in selecting clinical stage I patients for adjuvant therapy, identifying which patients with residual masses after chemotherapy benefit from surgical resection, and modifying surveillance schedules.

In early clinical studies, serum microRNA (miRNA) has demonstrated substantial promise as a biomarker. miRNA are small, non-coding RNA molecules that interact with messenger RNA (mRNA) to regulate gene expression at the post-transcriptional stage. In several cancer types, miRNA plays a role in malignant transformation and exhibits aberrant expression.231,232 In malignant GCT, expression analysis has demonstrated increased expression of several miRNA clusters, specifically miR-371-373 (chromosome 19q13), and miR-302-367 (chromosome 4q25).233,234 Both of these are specific to GCT and elevated in patients with both seminoma and NSGCT. Of the miRNA clusters, miR-371a-3p has the best performance characteristics as a biomarker in terms of sensitivity, specificity, and accuracy. Serum levels of miR-371a-3p have been shown to correlate with the extent of disease, response to therapy, relapse, and presence of residual malignant GCT elements.235-237 Serum levels of miR-371a-3p are not elevated in patients with teratoma. On the basis of the accumulating evidence in support of miR-371a-3p, two large intergroup trials are under development to define the role of miR371a-3p as a circulating biomarker in low-stage and advanced GCT to guide subsequent therapy.

Primary Surgical Management of Low-Volume Metastatic Seminoma

Primary RPLND has previously not been considered in the treatment of low-stage seminoma. Favorable outcomes with primary radiotherapy and primary chemotherapy are associated with acceptable acute toxicity and have been considered the standard-of-care for decades. However, concerns about late toxicity of these modalities has stimulated renewed interest in RPLND for clinical stage IIA and IIB seminoma. Primary RPLND has been a standard option for low-stage NSGCT with proven oncological efficacy and favorable short- and long-term morbidity. In comparison with NSGCT, seminoma is well-suited to treatment by RPLND as it is more likely to spread via lymphatic routes and has lower risks of occult systemic disease. RPLND for IIA and IIB seminoma has been evaluated in small studies, and low relapse rates with surgery alone have been reported.238-240

Tools and Resources

ABBREVIATIONS

AFP

Alpha-fetoprotein

AJCC

American Joint Committee on Cancer

AUA

American Urological Association

BEP

Bleomycin, Etoposide, Cisplatin

BOD

Board of Directors

CI

Confidence Interval

CT

Computed tomography

EP

Etoposide, Cisplatin

GCNIS

Germ Cell Neoplasia In Situ

GCT

Germ Cell Tumors

hCG

Human Chorionic gonadotropin

IGCCCG

International Germ Cell Cancer Collaborative Group

LDH

Lactate Dehydrogenase

LH

Luteinizing Hormone

MRI

Magnetic Resonance Imaging

NPV

Negative Predictive Value

NSGCT

Non-seminomatous germ cell tumors

PET

Positron Emission Tomography

PVV

Positive Predictive Value

PGC

Practice Guidelines Committee

RCT

Randomized Controlled Trials

RPLND

Retroperitoneal Lymph Node Dissection

RR

Relative Risk

SQC

Science and Quality Council

SEMS

Surgery in Early Metastatic Seminoma

TSS

Testis-Sparing Surgery

UDT

Undescended Testis

UICC

Union Internationale Contre le Cancer

REFERENCES

  1. Higgins J: Assessing quality of included studies in cochrane reviews. The Cochrane Collaboration Methods Groups Newsletter 2007; 11.
  2. Whiting PF, Rutjes AW, Westwood ME et al: Quadas-2: A revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 2011; 155: 529.
  3. Faraday M, Hubbard H, Kosiak B et al: Staying at the cutting edge: A review and analysis of evidence reporting and grading; the recommendations of the american urological association. BJU Int 2009; 104: 294.
  4. Siegel RL, K.D. M and Jemal A: Cancer statistics, 2019. CA Cancer J Clin 2019; 69.
  5. Howlader N, Noone A, Krapcho M et al: Seer cancer statistics review, 1975-2014. Bethesda, MD, 2017
  6. Nigam M, Aschebrook-Kilfoy B, Shikanov S et al: Increasing incidence of testicular cancer in the united states and europe between 1992 and 2009. World J Urol 2015; 33: 623.
  7. Wymer KM, Daneshmand S, Pierorazio PM et al: Mildly elevated serum alpha-fetoprotein (afp) among patients with testicular cancer may not be associated with residual cancer or need for treatment. Ann Oncol 2017; 28: 899.
  8. Doria-Rose VP, Biggs ML and Weiss NS: Subfertility and the risk of testicular germ cell tumors (united states). Cancer Causes Control 2005; 16: 651.
  9. Hanson HA, Anderson RE, Aston KI et al: Subfertility increases risk of testicular cancer: Evidence from population-based semen samples. Fertil Steril 2016; 105: 322.
  10. Skakkebaek NE, Berthelsen JG and Muller J: Carcinoma-in-situ of the undescended testis. Urol Clin North Am 1982; 9: 377.
  11. Dieckmann KP and Pichlmeier U: Clinical epidemiology of testicular germ cell tumors. World J Urol 2004; 22: 2.
  12. Wood HM and Elder JS: Cryptorchidism and testicular cancer: Separating fact from fiction. J Urol 2009; 181: 452.
  13. Akre O, Pettersson A and Richiardi L: Risk of contralateral testicular cancer among men with unilaterally undescended testis: A meta analysis. Int J Cancer 2009; 124: 687.
  14. Mai PL, Chen BE, Tucker K et al: Younger age-at-diagnosis for familial malignant testicular germ cell tumor. Familial cancer 2009; 8: 451.
  15. Fossa SD, Chen J, Schonfeld SJ et al: Risk of contralateral testicular cancer: A population-based study of 29,515 u.S. Men. J Natl Cancer Inst 2005; 97: 1056.
  16. Williamson SR, Delahunt B, Magi-Galluzzi C et al: The world health organization 2016 classification of testicular germ cell tumours: A review and update from the international society of urological pathology testis consultation panel. Histopathology 2017; 70: 335.
  17. Sheikine Y, Genega E, Melamed J et al: Molecular genetics of testicular germ cell tumors. Am J Cancer Res 2012; 2: 153.
  18. Mayer F, Stoop H, Scheffer GL et al: Molecular determinants of treatment response in human germ cell tumors. Clin Cancer Res 2003; 9: 767.
  19. AlDubayan SH, Pyle LC, Gamulin M et al: Association of inherited pathogenic variants in checkpoint kinase 2 (chek2) with susceptibility to testicular germ cell tumors. JAMA Oncol 2019.
  20. Powles TB, Bhardwa J, Shamash J et al: The changing presentation of germ cell tumours of the testis between 1983 and 2002. BJU Int 2005; 95: 1197.
  21. Cheville JC: Classification and pathology of testicular germ cell and sex cord-stromal tumors. Urol Clin North Am 1999; 26: 595.
  22. Castedo SM, de Jong B, Oosterhuis JW et al: Chromosomal changes in human primary testicular nonseminomatous germ cell tumors. Cancer Res 1989; 49: 5696.
  23. Sella A, el Naggar A, Ro JY et al: Evidence of malignant features in histologically mature teratoma. J Urol 1991; 146: 1025.
  24. Logothetis CJ, Samuels ML, Trindade A et al: The growing teratoma syndrome. Cancer 1982; 50: 1629.
  25. Motzer RJ, Amsterdam A, Prieto V et al: Teratoma with malignant transformation: Diverse malignant histologies arising in men with germ cell tumors. J Urol 1998; 159: 133.
  26. 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: 3388.
  27. Germa JR, Llanos M, Tabernero JM et al: False elevations of alpha-fetoprotein associated with liver dysfunction in germ cell tumors. Cancer 1993; 72: 2491.
  28. Albany C and Einhorn L: Pitfalls in management of patients with germ cell tumors and slight elevation of serum alpha-fetoprotein. J Clin Oncol 2014; 32: 2114.
  29. Deshpande N, Chavan R, Bale G et al: Hereditary persistence of alpha-fetoprotein is associated with the -119g>a polymorphism in afp gene. ACG Case Rep J 2017; 4: e33.
  30. Garnick MB: Spurious rise in human chorionic gonadotropin induced by marihuana in patients with testicular cancer. N Engl J Med 1980; 303: 1177.
  31. Vladutiu AO, Sulewski JM, Pudlak KA et al: Heterophilic antibodies interfering with radioimmunoassay. A false-positive pregnancy test. Jama 1982; 248: 2489.
  32. Soares DG, Millot F, Lacroix I et al: Heterophile antibody interference led to unneeded chemotherapy in a testicular cancer patient. Urol Case Rep 2016; 9: 1.
  33. Germa JR, Arcusa A and Casamitjana R: False elevations of human chorionic gonadotropin associated to iatrogenic hypogonadism in gonadal germ cell tumors. Cancer 1987; 60: 2489.
  34. Lempiainen A, Hotakainen K, Blomqvist C et al: Increased human chorionic gonadotropin due to hypogonadism after treatment of a testicular seminoma. Clin Chem 2007; 53: 1560.
  35. Amin MB, Greene FL, Edge SB et al: The eighth edition ajcc cancer staging manual: Continuing to build a bridge from a population-based to a more "personalized" approach to cancer staging. CA Cancer J Clin 2017; 67: 93.
  36. Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin 2018; 68: 7.
  37. Bosl GJ, Vogelzang NJ, Goldman A et al: Impact of delay in diagnosis on clinical stage of testicular cancer. Lancet 1981; 2: 970.
  38. Moul JW, Paulson DF, Dodge RK et al: Delay in diagnosis and survival in testicular cancer: Impact of effective therapy and changes during 18 years. J Urol 1990; 143: 520.
  39. Javadpour N, McIntire KR and Waldmann TA: Human chorionic gonadotropin (hcg) and alpha-fetoprotein (afp) in sera and tumor cells of patients with testicular seminoma: A prospective study. Cancer 1978; 42: 2768.
  40. Williams DHt, Karpman E, Sander JC et al: Pretreatment semen parameters in men with cancer. J Urol 2009; 181: 736.
  41. Bokemeyer C, Berger CC, Kuczyk MA et al: Evaluation of long-term toxicity after chemotherapy for testicular cancer. J Clin Oncol 1996; 14: 2923.
  42. Howell SJ and Shalet SM: Spermatogenesis after cancer treatment: Damage and recovery. J Natl Cancer Inst Monogr 2005: 12.
  43. Lieng H, Chung P, Lam T et al: Testicular seminoma: Scattered radiation dose to the contralateral testis in the modern era. Pract Radiat Oncol 2018; 8: e57.
  44. Fossa SD, Horwich A, Russell JM et al: Optimal planning target volume for stage i testicular seminoma: A medical research council randomized trial. Medical research council testicular tumor working group. J Clin Oncol 1999; 17: 1146.
  45. Foster RS, McNulty A, Rubin LR et al: The fertility of patients with clinical stage i testis cancer managed by nerve sparing retroperitoneal lymph node dissection. J Urol 1994; 152: 1139.
  46. Jewett MA, Kong YS, Goldberg SD et al: Retroperitoneal lymphadenectomy for testis tumor with nerve sparing for ejaculation. J Urol 1988; 139: 1220.
  47. Narayan P, Lange PH and Fraley EE: Ejaculation and fertility after extended retroperitoneal lymph node dissection for testicular cancer. J Urol 1982; 127: 685.
  48. Haugnes HS, Bosl GJ, Boer H et al: Long-term and late effects of germ cell testicular cancer treatment and implications for follow-up. J Clin Oncol 2012; 30: 3752.
  49. Kerns SL, Fung C, Monahan PO et al: Cumulative burden of morbidity among testicular cancer survivors after standard cisplatin-based chemotherapy: A multi-institutional study. J Clin Oncol 2018; 36: 1505.
  50. Sprauten M, Brydoy M, Haugnes HS et al: Longitudinal serum testosterone, luteinizing hormone, and follicle-stimulating hormone levels in a population-based sample of long-term testicular cancer survivors. J Clin Oncol 2014; 32: 571.
  51. Dogra VS, Gottlieb RH, Oka M et al: Sonography of the scrotum. Radiology 2003; 227: 18.
  52. Rifkin MD, Kurtz AB, Pasto ME et al: Diagnostic capabilities of high-resolution scrotal ultrasonography: Prospective evaluation. J Ultrasound Med 1985; 4: 13.
  53. Kim W, Rosen MA, Langer JE et al: Us mr imaging correlation in pathologic conditions of the scrotum. Radiographics 2007; 27: 1239.
  54. Balzer BL and Ulbright TM: Spontaneous regression of testicular germ cell tumors: An analysis of 42 cases. Am J Surg Pathol 2006; 30: 858.
  55. Winter TC, Kim B, Lowrance WT et al: Testicular microlithiasis: What should you recommend? AJR Am J Roentgenol 2016; 206: 1164.
  56. Wang T, Liu L, Luo J et al: A meta-analysis of the relationship between testicular microlithiasis and incidence of testicular cancer. Urol J 2015; 12: 2057.
  57. DeCastro BJ, Peterson AC and Costabile RA: A 5-year followup study of asymptomatic men with testicular microlithiasis. J Urol 2008; 179: 1420.
  58. Tan IB, Ang KK, Ching BC et al: Testicular microlithiasis predicts concurrent testicular germ cell tumors and intratubular germ cell neoplasia of unclassified type in adults: A meta-analysis and systematic review. Cancer 2010; 116: 4520.
  59. Albers P, Albrecht W, Algaba F et al: Guidelines on testicular cancer: 2015 update. Eur Urol 2015; 68: 1054.
  60. Manganaro L, Saldari M, Pozza C et al: Dynamic contrast-enhanced and diffusion-weighted mr imaging in the characterisation of small, non-palpable solid testicular tumours. Eur Radiol 2018; 28: 554.
  61. Tsili AC, Ntorkou A, Astrakas L et al: Magnetic resonance diffusion tensor imaging of the testis: Preliminary observations. Eur J Radiol 2017; 95: 265.
  62. El Sanharawi I, Correas JM, Glas L et al: Non-palpable incidentally found testicular tumors: Differentiation between benign, malignant, and burned-out tumors using dynamic contrast-enhanced mri. Eur J Radiol 2016; 85: 2072.
  63. Algebally AM, Tantawy HI and Yousef RRH: Value of diffusion weighted magnetic resonance imaging in diagnosis and characterization of scrotal abnormalities. Egyptian Journal of Radiology and Nuclear Medicine 2014; 45: 949.
  64. Sonmez G, Sivrioglu AK, Velioglu M et al: Optimized imaging techniques for testicular masses: Fast and with high accuracy. Wien Klin Wochenschr 2012; 124: 704.
  65. Tsili AC, Argyropoulou MI, Giannakis D et al: Mri in the characterization and local staging of testicular neoplasms. AJR Am J Roentgenol 2010; 194: 682.
  66. Andipa E, Liberopoulos K and Asvestis C: Magnetic resonance imaging and ultrasound evaluation of penile and testicular masses. World J Urol 2004; 22: 382.
  67. Oyen R, Verellen S, Drochmans A et al: Value of mri in the diagnosis and staging of testicular tumors. J Belge Radiol 1993; 76: 84.
  68. Johnson JO, Mattrey RF and Phillipson J: Differentiation of seminomatous from nonseminomatous teticular tumors with mr imaging. American Journal of Roentgenology 1990; 154: 539.
  69. Pierorazio PM, Patel HD, Cheaib J et al: Staging and management of early-stage testicular cancer. Unpublished systematic review. Prepared by the johns hopkins university evidence-based practice center for the american urological association education and research, inc. 2018.
  70. Woldrich JM, Im RD, Hughes-Cassidy FM et al: Magnetic resonance imaging for intratesticular and extratesticular scrotal lesions. Can J Urol 2013; 20: 6855.
  71. Rice KR, Cary CK, Masterson TA et al: Campbell-walsh urology: Surgery of testicular tumors, 11 ed, 2016
  72. Nord C, Bjoro T, Ellingsen D et al: Gonadal hormones in long-term survivors 10 years after treatment for unilateral testicular cancer. Eur Urol 2003; 44: 322.
  73. Clifford TG, Burg ML, Hu B et al: Satisfaction with testicular prosthesis after radical orchiectomy. Urology 2018; 114: 128.
  74. Dieckmann KP, Anheuser P, Schmidt S et al: Testicular prostheses in patients with testicular cancer - acceptance rate and patient satisfaction. BMC Urol 2015; 15: 16.
  75. Nichols PE, Harris KT, Brant A et al: Patient decision-making and predictors of genital satisfaction associated with testicular prostheses after radical orchiectomy: A questionnaire-based study of men with germ cell tumors of the testicle. Urology 2018.
  76. Yossepowitch O, Aviv D, Wainchwaig L et al: Testicular prostheses for testis cancer survivors: Patient perspectives and predictors of long-term satisfaction. J Urol 2011; 186: 2249.
  77. Muller T, Gozzi C, Akkad T et al: Management of incidental impalpable intratesticular masses of < or = 5 mm in diameter. BJU Int 2006; 98: 1001.
  78. Rolle L, Tamagnone A, Destefanis P et al: Microsurgical "testis-sparing" surgery for nonpalpable hypoechoic testicular lesions. Urology 2006; 68: 381.
  79. Rowland R: Management of nonpalpable testicular tumor: Sheynkin yr, sukkarieh t, lipke m, cohen hl, schulsinger da, department of urology, health science center, state university of new york at stony brook, stony brook, ny. Urologic Oncology: Seminars and Original Investigations 2005; 23: 139.
  80. Shilo Y, Zisman A, Lindner A et al: The predominance of benign histology in small testicular masses. Urol Oncol 2012; 30: 719.
  81. Harari SE, Sassoon DJ, Priemer DS et al: Testicular cancer: The usage of central review for pathology diagnosis of orchiectomy specimens. Urol Oncol 2017; 35: 605.e9.
  82. Bojanic N, Bumbasirevic U, Bojanic G et al: Testis sparing surgery for treatment of small testicular lesions: Is it feasible even in germ cell tumors? J Surg Oncol 2017; 115: 287.
  83. Bojanic N, Bumbasirevic U, Vukovic I et al: Testis sparing surgery in the treatment of bilateral testicular germ cell tumors and solitary testicle tumors: A single institution experience. J Surg Oncol 2015; 111: 226.
  84. Leonhartsberger N, Pichler R, Stoehr B et al: Organ preservation technique without ischemia in patients with testicular tumor. Urology 2014; 83: 1107.
  85. Ferretti L, Sargos P, Gross-Goupil M et al: Testicular-sparing surgery for bilateral or monorchide testicular tumours: A multicenter study of long-term oncological and functional results. BJU Int 2014; 114: 860.
  86. Lawrentschuk N, Zuniga A, Grabowksi AC et al: Partial orchiectomy for presumed malignancy in patients with a solitary testis due to a prior germ cell tumor: A large north american experience. J Urol 2011; 185: 508.
  87. Kazem I and Danella JF: Organ preservation for the treatment of contralateral testicular seminoma. Radiother Oncol 1999; 53: 45.
  88. Weissbach L: Organ preserving surgery of malignant germ cell tumors. J Urol 1995; 153: 90.
  89. Heidenreich A, Bonfig R, Derschum W et al: A conservative approach to bilateral testicular germ cell tumors. J Urol 1995; 153: 10.
  90. Dell'Atti L: Efficacy of ultrasound-guided testicle-sparing surgery for small testicular masses. J Ultrasound 2016; 19: 29.
  91. Heidenreich A, Weissbach L, Holtl W et al: Organ sparing surgery for malignant germ cell tumor of the testis. J Urol 2001; 166: 2161.
  92. von der Maase H, Giwercman A, Muller J et al: Management of carcinoma-in-situ of the testis. Int J Androl 1987; 10: 209.
  93. Schulze C and Holstein AF: On the histology of human seminoma: Development of the solid tumor from intratubular seminoma cells. Cancer 1977; 39: 1090.
  94. Pryor JP, Cameron KM, Chilton CP et al: Carcinoma in situ in testicular biopsies from men presenting with infertility. Br J Urol 1983; 55: 780.
  95. von der Maase H, Rorth M, Walbom-Jorgensen S et al: Carcinoma in situ of contralateral testis in patients with testicular germ cell cancer: Study of 27 cases in 500 patients. Br Med J (Clin Res Ed) 1986; 293: 1398.
  96. Zequi Sde C, da Costa WH, Santana TB et al: Bilateral testicular germ cell tumours: A systematic review. BJU Int 2012; 110: 1102.
  97. Kopp RP, Chevinsky M, Bernstein M et al: Bilateral testicular germ cell tumors in the era of multimodal therapy. Urology 2017; 103: 154.
  98. Harland SJ, Cook PA, Fossa SD et al: Intratubular germ cell neoplasia of the contralateral testis in testicular cancer: Defining a high risk group. J Urol 1998; 160: 1353.
  99. Dieckmann KP and Loy V: The value of the biopsy of the contralateral testis in patients with testicular germ cell cancer: The recent german experience. APMIS 1998; 106: 13.
  100. Kier MG, Lauritsen J, Almstrup K et al: Screening for carcinoma in situ in the contralateral testicle in patients with testicular cancer: A population-based study. Ann Oncol 2015; 26: 737.
  101. Dieckmann KP, Wilken S, Loy V et al: Treatment of testicular intraepithelial neoplasia (intratubular germ cell neoplasia unspecified) with local radiotherapy or with platinum-based chemotherapy: A survey of the german testicular cancer study group. Ann Oncol 2013; 24: 1332.
  102. Petersen PM, Giwercman A, Daugaard G et al: Effect of graded testicular doses of radiotherapy in patients treated for carcinoma-in-situ in the testis. J Clin Oncol 2002; 20: 1537.
  103. Bang AK, Petersen JH, Petersen PM et al: Testosterone production is better preserved after 16 than 20 gray irradiation treatment against testicular carcinoma in situ cells. Int J Radiat Oncol Biol Phys 2009; 75: 672.
  104. Dieckmann KP and Loy V: Intratesticular effects of cisplatin-based chemotherapy. Eur Urol 1995; 28: 25.
  105. Mumperow E, Lauke H, Holstein AF et al: Further practical experiences in the recognition and management of carcinoma in situ of the testis. Urol Int 1992; 48: 162.
  106. Dieckmann KP, Besserer A and Loy V: Low-dose radiation therapy for testicular intraepithelial neoplasia. J Cancer Res Clin Oncol 1993; 119: 355.
  107. Kleinschmidt K, Dieckmann KP, Georgiew A et al: Chemotherapy is of limited efficacy in the control of contralateral testicular intraepithelial neoplasia in patients with testicular germ cell cancer. Oncology 2009; 77: 33.
  108. 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: 594.
  109. Hanna NH and Einhorn LH: Testicular cancer--discoveries and updates. N Engl J Med 2014; 371: 2005.
  110. Feldman DR, Bosl GJ, Sheinfeld J et al: Medical treatment of advanced testicular cancer. Jama 2008; 299: 672.
  111. Cheng L, Albers P, Berney DM et al: Testicular cancer. Nat Rev Dis Primers 2018; 4: 29.
  112. Gilligan T, Lin DW, Aggarwal R et al: Testicular cancer: Nccn clinical practice guidelines in oncology. 2019: 73.
  113. Frazier AL, Hale JP, Rodriguez-Galindo C et al: Revised risk classification for pediatric extracranial germ cell tumors based on 25 years of clinical trial data from the united kingdom and united states. J Clin Oncol 2015; 33: 195.
  114. Shaikh F, Cullen JW, Olson TA et al: Reduced and compressed cisplatin-based chemotherapy in children and adolescents with intermediate-risk extracranial malignant germ cell tumors: A report from the children's oncology group. J Clin Oncol 2017; 35: 1203.
  115. de Wit M, Brenner W, Hartmann M 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: 1619.
  116. Bussar-Maatz R and Weissbach L: Retroperitoneal lymph node staging of testicular tumours. Tnm study group. Br J Urol 1993; 72: 234.
  117. Pizzocaro G, Nicolai N, Salvioni R et al: Comparison between clinical and pathological staging in low stage nonseminomatous germ cell testicular tumors. J Urol 1992; 148: 76.
  118. Fernandez EB, Moul JW, Foley JP et al: Retroperitoneal imaging with third and fourth generation computed axial tomography in clinical stage i nonseminomatous germ cell tumors. Urology 1994; 44: 548.
  119. Leibovitch L, Foster RS, Kopecky KK et al: Improved accuracy of computerized tomography based clinical staging in low stage nonseminomatous germ cell cancer using size criteria of retroperitoneal lymph nodes. J Urol 1995; 154: 1759.
  120. Hilton S, Herr HW, Teitcher JB et al: Ct detection of retroperitoneal lymph node metastases in patients with clinical stage 1 testicular nonseminomatous germ cell cancer: Assessment of size and distribution criteria. American Journal of Roentgenology 1997; 169: 521.
  121. Laukka M, Mannisto S, Beule A et al: Comparison between ct and mri in detection of metastasis of the retroperitoneum in testicular germ cell tumors: A prospective trial. Acta Oncol 2020; 59: 660.
  122. Larsen SKA, Agerbæk M, Jurik AG et al: Ten years of experience with mri follow-up of testicular cancer stage i: A retrospective study and an mri protocol with dwi. Acta Oncol 2020; 59: 1374.
  123. Narine R, Osman H, Wongwaisayawan S et al: Unenhanced mri of the abdomen and pelvis for surveillance of patients with stage 1 testicular cancer post-radical orchiectomy. Abdom Radiol (NY) 2021; 46: 1157.
  124. Sohaib SA, Koh DM, Barbachano Y et al: Prospective assessment of mri for imaging retroperitoneal metastases from testicular germ cell tumours. Clin Radiol 2009; 64: 362.
  125. Chen J and Daneshmand S: Modern management of testicular cancer. Cancer Treat Res 2018; 175: 273.
  126. White PM, Adamson DJA, Howard GCW et al: Imaging of the thorax in the management of germ cell testicular tumours. Clin Radiol 1999; 54: 207.
  127. Horan G, Rafique A, Robson J et al: Ct of the chest can hinder the management of seminoma of the testis; it detects irrelevant abnormalities. Br J Cancer 2007; 96: 882.
  128. Fernandez EB, Colon E, McLeod DG et al: Efficacy of radiographic chest imaging in patients with testicular cancer. Urology 1994; 44: 243.
  129. Ambrosini V, Zucchini G, Nicolini S et al: 18f-fdg pet/ct impact on testicular tumours clinical management. Eur J Nucl Med Mol Imaging 2014; 41: 668.
  130. Hain SF, O'Doherty MJ, Timothy AR et al: Fluorodeoxyglucose pet in the initial staging of germ cell tumours. Eur J Nucl Med 2000; 27: 590.
  131. Spermon JR, De Geus-Oei LF, Kiemeney LA et al: 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: 549.
  132. Stephenson AJ and Gilligan T: Neoplasms of the testis (in cambell-walsh urology, 11th edition), p. 784, 2015
  133. Schrader AJ, Ohlmann CH, Rossmanith S et al: Impact of evidence-based interdisciplinary guidelines on testis cancer management. Cancer 2006; 106: 313.
  134. Delaney RJ, Sayers CD, Walker MA et al: The continued value of central histopathological review of testicular tumours. Histopathology 2005; 47: 166.
  135. Lee AH, Mead GM and Theaker JM: The value of central histopathological review of testicular tumours before treatment. BJU Int 1999; 84: 75.
  136. Purshouse K, Watson RA, Church DN et al: Value of supraregional multidisciplinary review for the contemporary management of testicular tumors. Clin Genitourin Cancer 2017; 15: 152.
  137. Tandstad T, Dahl O, Cohn-Cedermark G et al: Risk-adapted treatment in clinical stage i nonseminomatous germ cell testicular cancer: The swenoteca management program. J Clin Oncol 2009; 27: 2122.
  138. Kollmannsberger C, Tyldesley S, Moore C et al: Evolution in management of testicular seminoma: Population-based outcomes with selective utilization of active therapies. Ann Oncol 2011; 22: 808.
  139. Daneshmand S and Hugen C: Early-stage (stage i) seminoma. In: Evidenceā€based urology. Edited by P. Dahm and R. Dmochowski, 2018
  140. Dieckmann KP, Dralle-Filiz I, Matthies C et al: Testicular seminoma clinical stage 1: Treatment outcome on a routine care level. J Cancer Res Clin Oncol 2016; 142: 1599.
  141. Tandstad T, Smaaland R, Solberg A et al: Management of seminomatous testicular cancer: A binational prospective population-based study from the swedish norwegian testicular cancer study group. J Clin Oncol 2011; 29: 719.
  142. Soper MS, Hastings JR, Cosmatos HA et al: Observation versus adjuvant radiation or chemotherapy in the management of stage i seminoma: Clinical outcomes and prognostic factors for relapse in a large us cohort. Am J Clin Oncol 2014; 37: 356.
  143. Tandstad T, Ståhl O, Dahl O et al: Treatment of stage i seminoma, with one course of adjuvant carboplatin or surveillance, risk-adapted recommendations implementing patient autonomy: A report from the swedish and norwegian testicular cancer group (swenoteca). Annals of Oncology 2016; 27: 1299.
  144. Jones WG, Fossa SD, Mead GM et al: Randomized trial of 30 versus 20 gy in the adjuvant treatment of stage i testicular seminoma: A report on medical research council trial te18, european organisation for the research and treatment of cancer trial 30942 (isrctn18525328). J Clin Oncol 2005; 23: 1200.
  145. Oliver RT, Mason MD, Mead GM et al: Radiotherapy versus single-dose carboplatin in adjuvant treatment of stage i seminoma: A randomised trial. Lancet 2005; 366: 293.
  146. Horwich A, Fossa SD, Huddart R et al: Second cancer risk and mortality in men treated with radiotherapy for stage i seminoma. Br J Cancer 2014; 110: 256.
  147. Haugnes HS, Wethal T, Aass N et al: Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: A 20-year follow-up study. J Clin Oncol 2010; 28: 4649.
  148. Brydoy M, Fossa SD, Klepp O et al: Paternity following treatment for testicular cancer. J Natl Cancer Inst 2005; 97: 1580.
  149. Travis LB, Fossa SD, Schonfeld SJ et al: Second cancers among 40,576 testicular cancer patients: Focus on long-term survivors. J Natl Cancer Inst 2005; 97: 1354.
  150. Tabakin AL, Shinder BM, Kim S et al: Retroperitoneal lymph node dissection as primary treatment for men with testicular seminoma: Utilization and survival analysis using the national cancer data base, 2004-2014. Clin Genitourin Cancer 2020; 18: e194.
  151. Classen J, Dieckmann K, Bamberg M et al: Radiotherapy with 16 gy may fail to eradicate testicular intraepithelial neoplasia: Preliminary communication of a dose-reduction trial of the german testicular cancer study group. Br J Cancer 2003; 88: 828.
  152. Garcia-del-Muro X, Maroto P, Guma J et al: Chemotherapy as an alternative to radiotherapy in the treatment of stage iia and iib testicular seminoma: A spanish germ cell cancer group study. J Clin Oncol 2008; 26: 5416.
  153. Fossa SD, Aass N, Winderen M et al: Long-term renal function after treatment for malignant germ-cell tumours. Ann Oncol 2002; 13: 222.
  154. Rossen PB, Pedersen AF, Zachariae R et al: Health-related quality of life in long-term survivors of testicular cancer. J Clin Oncol 2009; 27: 5993.
  155. O'Sullivan JM, Huddart RA, Norman AR et al: Predicting the risk of bleomycin lung toxicity in patients with germ-cell tumours. Ann Oncol 2003; 14: 91.
  156. Hu B and Daneshmand S: Retroperitoneal lymph node dissection as primary treatment for metastatic seminoma. Adv Urol 2018; 2018: 7978958.
  157. Daneshmand S, Cary C, Masterson T et al: Surgery in early metastatic seminoma: A phase ii trial of retroperitoneal lymph node dissection for testicular seminoma with limited retroperitoneal lymphadenopathy. J Clin Oncol 2023: Jco2200624.
  158. Hiester A, Che Y, Lusch A et al: Phase 2 single-arm trial of primary retroperitoneal lymph node dissection in patients with seminomatous testicular germ cell tumors with clinical stage iia/b (primetest). Eur Urol 2022.
  159. Heidenreich A, Paffenholz P, Nestler T et al: Nerve sparing retroperitoneal lymph node dissection in clinical stage iia/b seminoma: The cotrims trial. Journal of Clinical Oncology 2022; 40: 418.
  160. Stephenson AJ, Bosl GJ, Motzer RJ et al: Retroperitoneal lymph node dissection for nonseminomatous germ cell testicular cancer: Impact of patient selection factors on outcome. J Clin Oncol 2005; 23: 2781.
  161. Saxman SB, Nichols CR, Foster RS et al: The management of patients with clinical stage i nonseminomatous testicular tumors and persistently elevated serologic markers. J Urol 1996; 155: 587.
  162. Davis BE, Herr HW, Fair WR et al: The management of patients with nonseminomatous germ cell tumors of the testis with serologic disease only after orchiectomy. J Urol 1994; 152: 111.
  163. Kollmannsberger C, Moore C, Chi KN et al: Non-risk-adapted surveillance for patients with stage i nonseminomatous testicular germ-cell tumors: Diminishing treatment-related morbidity while maintaining efficacy. Ann Oncol 2010; 21: 1296.
  164. Daugaard G, Petersen PM and Rorth M: Surveillance in stage i testicular cancer. Apmis 2003; 111: 76.
  165. Divrik RT, Akdogan B, Ozen H et al: Outcomes of surveillance protocol of clinical stage i nonseminomatous germ cell tumors-is shift to risk adapted policy justified? J Urol 2006; 176: 1424.
  166. Hermans BP, Sweeney CJ, Foster RS et al: Risk of systemic metastases in clinical stage i nonseminoma germ cell testis tumor managed by retroperitoneal lymph node dissection. J Urol 2000; 163: 1721.
  167. Sweeney CJ, Hermans BP, Heilman DK et al: Results and outcome of retroperitoneal lymph node dissection for clinical stage i embryonal carcinoma--predominant testis cancer. J Clin Oncol 2000; 18: 358.
  168. Ondrus D, Goncalves F, Kausitz J et al: The value of prognostic factors in the management of stage i nonseminomatous germ cell testicular tumors (nsgctt). Neoplasma 1996; 43: 195.
  169. Gumus M, Bilici A, Odabas H et al: Outcomes of surveillance versus adjuvant chemotherapy for patients with stage ia and ib nonseminomatous testicular germ cell tumors. World J Urol 2017; 35: 1103.
  170. Al-Ahmadie HA, Carver BS, Cronin AM et al: Primary retroperitoneal lymph node dissection in low-stage testicular germ cell tumors: A detailed pathologic study with clinical outcome analysis with special emphasis on patients who did not receive adjuvant therapy. Urology 2013; 82: 1341.
  171. Sogani PC, Perrotti M, Herr HW et al: Clinical stage i testis cancer: Long-term outcome of patients on surveillance. J Urol 1998; 159: 855.
  172. Pohar KS, Rabbani F, Bosl GJ et al: Results of retroperitoneal lymph node dissection for clinical stage i and ii pure embryonal carcinoma of the testis. J Urol 2003; 170: 1155.
  173. Sturgeon JF, Moore MJ, Kakiashvili DM et al: Non-risk-adapted surveillance in clinical stage i nonseminomatous germ cell tumors: The princess margaret hospital's experience. Eur Urol 2011; 59: 556.
  174. Tandstad T, Cohn-Cedermark G, Dahl O et al: Long-term follow-up after risk-adapted treatment in clinical stage 1 (cs1) nonseminomatous germ-cell testicular cancer (nsgct) implementing adjuvant cvb chemotherapy. A swenoteca study. Ann Oncol 2010; 21: 1858.
  175. Daugaard G, Gundgaard MG, Mortensen MS et al: Surveillance for stage i nonseminoma testicular cancer: Outcomes and long-term follow-up in a population-based cohort. J Clin Oncol 2014; 32: 3817.
  176. Oldenburg J, Aparicio J, Beyer J et al: Personalizing, not patronizing: The case for patient autonomy by unbiased presentation of management options in stage i testicular cancer. Ann Oncol 2015; 26: 833.
  177. Elwyn G, Frosch D, Thomson R et al: Shared decision making: A model for clinical practice. J Gen Intern Med 2012; 27: 1361.
  178. Scholl I, LaRussa A, Hahlweg P et al: Organizational- and system-level characteristics that influence implementation of shared decision-making and strategies to address them - a scoping review. Implement Sci 2018; 13: 40.
  179. Williams N, Fleming C and Doubleday A: Patient and provider perspectives on shared decision making: A systematic review of the peer-reviewed literature. J Comp Eff Res 2017; 6: 683.
  180. Donadio AC, Motzer RJ, Bajorin DF et al: Chemotherapy for teratoma with malignant transformation. J Clin Oncol 2003; 21: 4285.
  181. El Mesbahi O, Terrier-Lacombe MJ, Rebischung C et al: Chemotherapy in patients with teratoma with malignant transformation. Eur Urol 2007; 51: 1306.
  182. Ko JJ, Bernard B, Tran B et al: Conditional survival of patients with metastatic testicular germ cell tumors treated with first-line curative therapy. J Clin Oncol 2016; 34: 714.
  183. van Dijk MR, Steyerberg EW and Habbema JD: Survival of non-seminomatous germ cell cancer patients according to the igcc classification: An update based on meta-analysis. Eur J Cancer 2006; 42: 820.
  184. Donohue JP, Thornhill JA, Foster RS et al: Clinical stage b non-seminomatous germ cell testis cancer: The indiana university experience (1965&#x2013;1989) using routine primary retroperitoneal lymph node dissection. European Journal of Cancer 1995; 31: 1599.
  185. Richie JP and Kantoff PW: Is adjuvant chemotherapy necessary for patients with stage b1 testicular cancer? J Clin Oncol 1991; 9: 1393.
  186. Donohue JP, Thornhill JA, Foster RS et al: The role of retroperitoneal lymphadenectomy in clinical stage b testis cancer: The indiana university experience (1965 to 1989). J Urol 1995; 153: 85.
  187. Kondagunta GV and Motzer RJ: Adjuvant chemotherapy for stage ii nonseminomatous germ cell tumors. Urol Clin North Am 2007; 34: 179.
  188. Kondagunta GV and Motzer RJ: Adjuvant chemotherapy for stage ii nonseminomatous germ-cell tumors. Semin Urol Oncol 2002; 20: 239.
  189. Behnia M, Foster R, Einhorn LH et al: Adjuvant bleomycin, etoposide and cisplatin in pathological stage ii non-seminomatous testicular cancer. The indiana university experience. Eur J Cancer 2000; 36: 472.
  190. Williams SD, Stablein DM, Einhorn LH et al: Immediate adjuvant chemotherapy versus observation with treatment at relapse in pathological stage ii testicular cancer. N Engl J Med 1987; 317: 1433.
  191. Zaid MA, Gathirua-Mwangi WG, Fung C et al: Clinical and genetic risk factors for adverse metabolic outcomes in north american testicular cancer survivors. J Natl Compr Canc Netw 2018; 16: 257.
  192. Chovanec M, Abu Zaid M, Hanna N et al: Long-term toxicity of cisplatin in germ-cell tumor survivors. Ann Oncol 2017; 28: 2670.
  193. Fung C, Fossa SD, Williams A et al: Long-term morbidity of testicular cancer treatment. Urol Clin North Am 2015; 42: 393.
  194. Fung C, Sesso HD, Williams AM et al: Multi-institutional assessment of adverse health outcomes among north american testicular cancer survivors after modern cisplatin-based chemotherapy. J Clin Oncol 2017; 35: 1211.
  195. Fung C, Fossa SD, Milano MT et al: Solid tumors after chemotherapy or surgery for testicular nonseminoma: A population-based study. J Clin Oncol 2013; 31: 3807.
  196. Lowrance WT, Cookson MS, Clark PE et al: Assessing retroperitoneal lymphadenectomy experience in united states urological residency programs. J Urol 2007; 178: 500.
  197. Woldu SL, Matulay JT, Clinton TN et al: Impact of hospital case volume on testicular cancer outcomes and practice patterns. Urol Oncol 2018; 36: 14.e7.
  198. Shamash J, Ansell W, Alifrangis C et al: The impact of a supranetwork multidisciplinary team (smdt) on decision-making in testicular cancers: A 10-year overview of the anglian germ cell cancer collaborative group (agcccg). Br J Cancer 2021; 124: 368.
  199. Pearce SM, Golan S, Gorin MA et al: Safety and early oncologic effectiveness of primary robotic retroperitoneal lymph node dissection for nonseminomatous germ cell testicular cancer. Eur Urol 2017; 71: 476.
  200. Nicolai N, Tarabelloni N, Gasperoni F et al: Laparoscopic retroperitoneal lymph node dissection for clinical stage i nonseminomatous germ cell tumors of the testis: Safety and efficacy analyses at a high volume center. J Urol 2018; 199: 741.
  201. Schwen ZR, Gupta M and Pierorazio PM: A review of outcomes and technique for the robotic-assisted laparoscopic retroperitoneal lymph node dissection for testicular cancer. Adv Urol 2018; 2018: 2146080.
  202. Weissbach L, Boedefeld EA and Horstmann-Dubral B: Surgical treatment of stage-i non-seminomatous germ cell testis tumor. Final results of a prospective multicenter trial 1982-1987. Testicular tumor study group. Eur Urol 1990; 17: 97.
  203. Foster RS: Modified retroperitoneal lymphadenectomy. BJU Int 2004; 94: 941.
  204. Eggener SE, Carver BS, Sharp DS et al: Incidence of disease outside modified retroperitoneal lymph node dissection templates in clinical stage i or iia nonseminomatous germ cell testicular cancer. J Urol 2007; 177: 937.
  205. Nelson JB, Chen RN, Bishoff JT et al: Laparoscopic retroperitoneal lymph node dissection for clinical stage i nonseminomatous germ cell testicular tumors. Urology 1999; 54: 1064.
  206. Albers P, Siener R, Krege S et al: Randomized phase iii trial comparing retroperitoneal lymph node dissection with one course of bleomycin and etoposide plus cisplatin chemotherapy in the adjuvant treatment of clinical stage i nonseminomatous testicular germ cell tumors: Auo trial ah 01/94 by the german testicular cancer study group. J Clin Oncol 2008; 26: 2966.
  207. Motzer RJ, Sheinfeld J, Mazumdar M et al: Etoposide and cisplatin adjuvant therapy for patients with pathologic stage ii germ cell tumors. J Clin Oncol 1995; 13: 2700.
  208. Fraley EE, Narayan P, Vogelzang NJ et al: Surgical treatment of patients with stages i and ii nonseminomatous testicular cancer. J Urol 1985; 134: 70.
  209. Weissbach L, Bussar-Maatz R, Flechtner H et al: Rplnd or primary chemotherapy in clinical stage iia/b nonseminomatous germ cell tumors? Results of a prospective multicenter trial including quality of life assessment. Eur Urol 2000; 37: 582.
  210. McHugh DJ, Funt SA, Silber D et al: Adjuvant chemotherapy with etoposide plus cisplatin for patients with pathologic stage ii nonseminomatous germ cell tumors. J Clin Oncol 2020; 38: 1332.
  211. Kollmannsberger C, Tandstad T, Bedard PL et al: Patterns of relapse in patients with clinical stage i testicular cancer managed with active surveillance. J Clin Oncol 2015; 33: 51.
  212. Cummins S, Yau T, Huddart R et al: Surveillance in stage i seminoma patients: A long-term assessment. Eur Urol 2010; 57: 673.
  213. von der Maase H, Specht L, Jacobsen GK et al: Surveillance following orchidectomy for stage i seminoma of the testis. Eur J Cancer 1993; 29a: 1931.
  214. Choo R, Thomas G, Woo T et al: Long-term outcome of postorchiectomy surveillance for stage i testicular seminoma. Int J Radiat Oncol Biol Phys 2005; 61: 736.
  215. Joffe JK, Cafferty FH, Murphy L et al: Imaging modality and frequency in surveillance of stage i seminoma testicular cancer: Results from a randomized, phase iii, noninferiority trial (trisst). J Clin Oncol 2022; 40: 2468.
  216. Yu HY, Madison RA, Setodji CM et al: Quality of surveillance for stage i testis cancer in the community. J Clin Oncol 2009; 27: 4327.
  217. Gels ME, Hoekstra HJ, Sleijfer DT et al: Detection of recurrence in patients with clinical stage i nonseminomatous testicular germ cell tumors and consequences for further follow-up: A single-center 10-year experience. J Clin Oncol 1995; 13: 1188.
  218. Colls BM, Harvey VJ, Skelton L et al: Late results of surveillance of clinical stage i nonseminoma germ cell testicular tumours: 17 years' experience in a national study in new zealand. BJU Int 1999; 83: 76.
  219. Rustin GJ, Mead GM, Stenning SP et al: Randomized trial of two or five computed tomography scans in the surveillance of patients with stage i nonseminomatous germ cell tumors of the testis: Medical research council trial te08, isrctn56475197--the national cancer research institute testis cancer clinical studies group. J Clin Oncol 2007; 25: 1310.
  220. Azad AA, Eigl BJ, Murray RN et al: Efficacy of enzalutamide following abiraterone acetate in chemotherapy-naive metastatic castration-resistant prostate cancer patients. Eur Urol 2015; 67: 23.
  221. Nayan M, Jewett MA, Hosni A et al: Conditional risk of relapse in surveillance for clinical stage i testicular cancer. Eur Urol 2017; 71: 120.
  222. Hamilton RJ, Nayan M, Anson-Cartwright L et al: Treatment of relapse of clinical stage i nonseminomatous germ cell tumors on surveillance. J Clin Oncol 2019: Jco1801250.
  223. Huang J, Chan SC, Tin MS et al: Worldwide distribution, risk factors, and temporal trends of testicular cancer incidence and mortality: A global analysis. Eur Urol Oncol 2022; 5: 566.
  224. Fung C, Dinh PC, Fossa SD et al: Testicular cancer survivorship. J Natl Compr Canc Netw 2019; 17: 1557.
  225. Einhorn LH and Donohue J: Cis-diamminedichloroplatinum, vinblastine, and bleomycin combination chemotherapy in disseminated testicular cancer. Ann Intern Med 1977; 87: 293.
  226. Verdecchia A, Francisci S, Brenner H et al: Recent cancer survival in europe: A 2000-02 period analysis of eurocare-4 data. Lancet Oncol 2007; 8: 784.
  227. Fung C and Travis LB: Testicular cancer survivorship: Looking back to move forward. J Clin Oncol 2021; 39: 3531.
  228. Shrem NS, Wood L, Hamilton RJ et al: Testicular cancer survivorship: Long-term toxicity and management. Can Urol Assoc J 2022; 16: 257.
  229. Weiner AB, Pearce SM and Eggener SE: Management trends for men with early-stage nonseminomatous germ cell tumors of the testicle: An analysis of the national cancer database. Cancer 2017; 123: 245.
  230. Roeleveld TA, Horenblas S, Meinhardt W et al: Surveillance can be the standard of care for stage i nonseminomatous testicular tumors and even high risk patients. J Urol 2001; 166: 2166.
  231. Deng S, Calin GA, Croce CM et al: Mechanisms of microrna deregulation in human cancer. Cell Cycle 2008; 7: 2643.
  232. Hayes J, Peruzzi PP and Lawler S: Micrornas in cancer: Biomarkers, functions and therapy. Trends Mol Med 2014; 20: 460.
  233. Voorhoeve PM, le Sage C, Schrier M et al: A genetic screen implicates mirna-372 and mirna-373 as oncogenes in testicular germ cell tumors. Cell 2006; 124: 1169.
  234. 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: 2911.
  235. Dieckmann KP, Radtke A, Spiekermann M et al: Serum levels of microrna mir-371a-3p: A sensitive and specific new biomarker for germ cell tumours. Eur Urol 2017; 71: 213.
  236. Nappi L, B ON and S D: Circulating mir-371a-3p for the detection of low volume viable germ cell tumor: Expanded pilot data, clinical implications and future study. J Clin Oncol 2018.
  237. van Agthoven T, Eijkenboom WMH and Looijenga LHJ: Microrna-371a-3p as informative biomarker for the follow-up of testicular germ cell cancer patients. Cell Oncol (Dordr) 2017; 40: 379.
  238. Hu B, Shah S, Shojaei S et al: Retroperitoneal lymph node dissection as first-line treatment of node-positive seminoma. Clin Genitourin Cancer 2015; 13: e265.
  239. Mezvrishvili Z and Managadze L: Retroperitoneal lymph node dissection for high-risk stage i and stage iia seminoma. Int Urol Nephrol 2006; 38: 615.
  240. Warszawski N and Schmucking M: Relapses in early-stage testicular seminoma: Radiation therapy versus retroperitoneal lymphadenectomy. Scand J Urol Nephrol 1997; 31: 355.