<<

EUROPEAN 68 (2015) 1054–1068 available at www.sciencedirect.com journal homepage: www.europeanurology.com

Guidelines Guidelines on Testicular : 2015 Update

Peter Albers a,*, Walter Albrecht b, Ferran Algaba c, Carsten Bokemeyer d, Gabriella Cohn-Cedermark e, Karim Fizazi f, Alan Horwich g, Maria Pilar Laguna h, Nicola Nicolai i, Jan Oldenburg j a Department of Urology, Medical Faculty, Du¨sseldorf University, Du¨sseldorf, ; b Department of Urology, Mistelbach, Austria; c Department of , Fundacio Puigvert, Barcelona, Spain; d Department of , Hematology and Bone Marrow Transplantation with section Pneumology, Universita¨tskliniken Eppendorf, Hamburg, Germany; e Department of Oncology-Pathology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; f Department of Medicine, University of Paris XI, Villejuif, France; g Academic Unit of Radiotherapy and Oncology, Royal Marsden NHS Trust and The Institute of Cancer Research, Sutton, UK; h Department of Urology, Amsterdam Medical Centre, Amsterdam University, Amsterdam, The Netherlands; i Department of , Urology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; j Health Sciences, Høgskolen i Buskerud og Vestfold, Kongsberg, Norway

Article info Abstract

Article history: Context: This is an update of the previous European Association of Urology testis cancer Accepted July 18, 2015 guidelines published in 2011, which included major changes in the diagnosis and treatment of germ cell tumours. Associate Editor: Objective: To summarise latest developments in the treatment of this rare . James Catto Recommendations have been agreed within a multidisciplinary working group consist- ing of urologists, medical oncologists, and radiation oncologists. Evidence acquisition: A semi-structured literature search up to February 2015 was Keywords: performed to update the recommendations. In addition, this document was subjected to Testis cancer double-blind peer review before publication. Evidence synthesis: This publication focuses on the most important changes in treat- Germ cell tumour ment recommendations for clinical stage I disease and the updated recommendations Retroperitoneal for follow-up. dissection Conclusions: Most changes in the recommendations will lead to an overall reduction in Risk-adapted treatment treatment burden for patients with germ cell tumours. In advanced stages, treatment intensification is clearly defined to further improve overall survival rates. Patient summary: This is an update of a previously published version of the European Metastatic disease Association of Urology guidelines for testis cancer, and includes new recommendations Relapse for clinical stage I disease and revision of the follow-up recommendations. Patients Follow-up should be fully informed of all the treatment options available to them. EAU guidelines # 2015 European Association of Urology. Published by Elsevier B.V. All rights reserved.

* Corresponding author. Department of Urology, Medical Faculty, University Hospital, Heinrich- Heine University Du¨ sseldorf, Moorenstrasse 5, D-40225 Du¨ sseldorf, Germany. Tel. +49 21 18118110; Fax: +49 21 18118676. E-mail address: [email protected] (P. Albers).

1. Introduction expertise for patients with advanced disease. In the overwhelming group of patients with low-stage disease, Testicular cancer is a rare disease, with an incidence of up to the main goal of treatment is to reduce treatment-related 10 in 100 000 men. In Europe, cure rates of up to 97% for all long-term toxicities, including secondary . Thus, stages have been achieved [1]. However, cure is dependent regular updates of current treatment guidelines are on excellent diagnosis, treatment facilities, and specialised necessary to achieve the best possible treatment outcome. http://dx.doi.org/10.1016/j.eururo.2015.07.044 0302-2838/# 2015 European Association of Urology. Published by Elsevier B.V. All rights reserved. EUROPEAN UROLOGY 68 (2015) 1054–1068 1055

Current evidence suggests that the best results are 3. Evidence synthesis obtained in high-volume reference centres, not only for patients with advanced disease but also for patients with 3.1. Diagnosis of testicular cancer early-stage disease. Although early stages can be successfully treated in a nonreference centre, the relapse rate is higher It is very important to correctly classify a patient with a [2]. In poor- nonseminomatous germ-cell tumours, newly diagnosed germ-cell cancer. Ultrasound (US) of both overall survival (OS) within a clinical trial depends on the testis remains the cornerstone of primary imaging in number of patients treated at the participating centre (poorer patients with testicular tumours followed by computed if fewer than five patients are enrolled) [3]. In the same tomography (CT) of the and chest as subsequent context, the frequency of postchemotherapy residual tumour staging tools (Table 1). Correct interpretation of tumour resection is associated with perioperative mortality and OS markers before and after in conjunction with [4,5]. CT findings allows correct patient classification according to First analyses of a recently introduced internet-based TNM and Union for International Cancer Control staging second-opinion portal suggest that up to 40% of primary (Tables 2 and 3) [9]. Patients with metastatic disease should diagnoses were incorrect and treatment was changed in a be classified according to the International Germ Cell considerably high number of patients after a second opinion Cancer Collaborative Group (IGCCCG) [10] to tailor further [6]. treatment (Table 4).

2. Evidence acquisition 3.2. Diagnosis and treatment of testicular intraepithelial neoplasia (TIN) A multidisciplinary team of urologists, medical oncologists, radiation oncologists, and a pathologist were involved in Contralateral has been advocated to rule out the producing this document, which is based on a semi- presence of TIN [11]. Although systematic contralateral structured review of the literature up to February 2015. This biopsy is routine policy in some countries, the low incidence publication focuses on the most important changes and of TIN (up to 9%) and contralateral metachronous testicular relevant clinical recommendations. For unchanged recom- tumours (2.5%) [12,13], the morbidity associated with TIN mendations, the reader is referred to the previous publica- treatment, and the low stage of most metachronous tion of the guidelines in European Urology [7]. The full tumours at presentation mean that biopsy recommendation version of the 2015 guidelines is available on the European for all patients is controversial. Association of Urology (EAU) website (http://uroweb.org/ It is still difficult to reach a consensus on whether guideline/testicular-cancer/). References were assessed the existence of contralateral TIN must be identified according to their level of scientific evidence, and guideline in all cases. However, biopsy of the contralateral testis recommendations were graded according to a system should be offered to patients at high risk of contralateral modified from the Oxford Centre for Evidence-based TIN (testicular volume <12 ml, history of cryptorchidism, Medicine levels of evidence [8]. or poor spermatogenesis with Johnson score 1–3). A contralateral biopsy is not necessary in patients older than 40 yr without risk factors [14,15].Adoublebiopsy Table 1 – Recommended tests for staging at diagnosis increases sensitivity [14]. Patients should be informed that a testicular tumour may arise in spite of a negative Test Recommendation GR biopsy. Serum tumour markers a-Fetoprotein A When TIN is diagnosed, local radiotherapy (16–20 Gy in hCG fractions of 2 Gy) is the treatment of choice in the case of a solitary testis. Testicular radiotherapy in a solitary testis Abdominopelvic CT All patients A Chest CT All patients A will result in and a higher long-term risk of Leydig Testis ultrasound (bilateral) All patients A cell insufficiency [16,17]. Fertile patients who wish to father Bone scan or spinal MRI In the case of symptoms children may delay and can be followed Brain scan (CT/MRI) In the case of symptoms and patients with metastatic with regular testicular US [14]. disease with multiple lung If TIN is diagnosed and the contralateral testis is healthy, metastases and/or the options for management are orchiectomy or close high b-hCG levels observation (with a 5-yr risk of developing testicular cancer Further investigations of 50%).

Fertility investigations: B Total testosterone 3.3. Prognostic factors for progression and recurrence in Luteinising clinical stage I Follicle-stimulating hormone Semen analysis In stage I , tumour size (>4 cm) and invasion of Sperm banking Should be offered A the have been identified as predictors of relapse GR = grade of recommendation; hCG = human chorionic gonadotrophin; in a pooled analysis. However, these risk factors have not CT = computed tomography; MRI = magnetic resonance imaging. been validated in a prospective setting, although the 1056 EUROPEAN UROLOGY 68 (2015) 1054–1068

Table 2 – TNM classification of testicular cancer [9]

pT: primary tumour a pTX Primary tumour cannot be assessed pT0 No evidence of primary tumour (eg, histologic scar in testis) pTis Intratubular germ-cell neoplasia (testicular intraepithelial neoplasia) pT1 Tumour limited to testis and without vascular/lymphatic invasion; tumour may invade tunica albuginea, but not tunica vaginalis pT2 Tumour limited to testis and epididymis with vascular/lymphatic invasion, or tumour extending through into the tunica albuginea with involvement of tunica vaginalis pT3 Tumour invades with or without vascular/lymphatic invasion pT4 Tumour invades with or without vascular/lymphatic invasion N: regional lymph nodes (clinical) NX Regional lymph nodes cannot be assessed N0 No regional lymph node N1 Metastasis with a lymph node mass 2 cm IGD or multiple lymph nodes, none >2 cm IGD N2 Metastasis with a lymph node mass >2 cm but not >5 cm IGD, or multiple lymph nodes with any one mass >2 cm but not >5 cm IGD N3 Metastasis with a lymph node mass >5 cm IGD pN: pathologic lymph nodes pNX Regional lymph nodes cannot be assessed pN0 No regional lymph node metastasis pN1 Metastasis with a lymph node mass 2 cm IGD and 5 positive nodes, none >2 cm IGD pN2 Metastasis with a lymph node mass >2 cm but not >5 cm IGD; or >5 nodes positive, none >5 cm IGD; or evidence of extranodal tumour extension pN3 Metastasis with a lymph node mass >5 cm IGD M: distant metastasis MX Distant metastasis cannot be assessed M0 No distant metastasis M1 Distant metastasis M1a Nonregional lymph node(s) or lung M1b Other sites S: serum tumour markers SX Serum marker studies not available or not performed S0 Serum marker levels within normal limits S1 LDH <1.5 Â ULN and hCG <5000 mIU/ml and AFP <1000 ng/ml S2 LDH 1.5–10 Â ULN or hCG 5000–50 000 mIU/ml or AFP 1000–10 000 ng/ml S3 LDH >10 Â ULN or hCG >50 000 mIU/ml or AFP >10 000 ng/ml

IGD = in greatest dimension; LDH = lactate dehydrogenase; hCG = human chorionic gonadotrophin; AFP = a-fetoprotein; ULN = upper limit of normal range. a Except for pTis and pT4, for which radical orchiectomy is not always necessary for classification purposes, the extent of the primary tumour is classified after radical orchiectomy. In other circumstances, TX is used if no radical orchiectomy has been performed.

Table 3 – Stage grouping for cancer absence of both factors indicated a low recurrence rate (6%) [18]. Stage 0 pTis N0 M0 S0, SX Stage I a pT1–T4 N0 M0 SX For stage I nonseminoma, vascular invasion by the Stage IA pT1 N0 M0 S0 primary tumour into blood or lymphatic vessels is the most Stage IB pT2–T4 N0 M0 S0 important predictor of occult metastatic disease. The Stage IS Any patient/TX N0 M0 S1–3 Stage II Any patient/TX N1–N3 M0 SX proliferation rate and the percentage of embryonal carci- Stage IIA Any patient/TX N1 M0 S0, S1 noma are additional predictors that increase the positive Stage IIB Any patient/TX N2 M0 S0, S1 and negative predictive values, respectively, for vascular Stage IIC Any patient/TX N3 M0 S0, S1 invasion [19]. Stage III Any patient/TX Any N M1a SX Stage IIIA Any patient/TX Any N M1a S0, S1 Stage IIIB Any patient/TX N1–N3 M0 S2 3.4. Treatment of clinical stage I seminoma Any patient/TX Any N M1a S2 Stage IIIC Any patient/TX N1–N3 M0 S3 Staging procedures reveal that approximately 15–20% of Any patient/TX Any N M1a S3 Any patient/TX Any N M1b Any S stage I seminoma patients have subclinical metastatic disease, usually in the retroperitoneum, and will relapse a Stage I testicular cancer includes the following substages: after orchiectomy alone. The decision regarding adjuvant Stage IA: patients have primary tumours limited to the testis and epididymis, with no evidence of microscopic vascular or lymphatic invasion by tumour treatment should always be based on a thorough discussion cells on microscopy, no sign of metastases on clinical examination or imaging, with the patient, taking into account the advantages and and post-orchiectomy serum tumour marker levels within normal limits. disadvantages described and the patient’s individual situa- Marker decline in patients with clinical stage I disease should be assessed until tion. normalisation. Stage IB: patients have a more locally invasive primary tumour, but no sign of metastatic disease. 3.4.1. Surveillance Stage IS: patients have persistently elevated (and usually increasing) serum Several prospective nonrandomised surveillance studies tumour marker levels after orchiectomy, indicating subclinical metastatic have been conducted during the past decade. The largest disease (or possibly a second germ-cell tumour in the remaining testis). study, with >1500 patients, showed an overall relapse rate EUROPEAN UROLOGY 68 (2015) 1054–1068 1057

Table 4 – Prognostic-based staging system for metastatic germ cell cancer (International Germ Cell Cancer Collaborative Group) a

Group Type Criteria

Good prognosis Nonseminoma (56% of cases) All of the following criteria: 5-yr PFS 89%  Testis/retroperitoneal primary 5-yr survival 92%  No nonpulmonary visceral metastases  AFP <1000 ng/ml  hCG <5000 IU/l (1000 ng/ml)  LDH <1.5 Â ULN Seminoma (90% of cases) All of the following criteria: 5-yr PFS 82%  Any primary site 5-yr survival 86%  No nonpulmonary visceral metastases  Normal AFP  Any hCG  Any LDH Intermediate prognosis Nonseminoma (28% of cases) All of the following criteria: 5-yr PFS 75%  Testis/retroperitoneal primary 5-yr survival 80%  No nonpulmonary visceral metastases  AFP 1000–10 000 ng/ml or  hCG 5000–50 000 IU/l or  LDH 1.5–10 Â ULN Seminoma (10% of cases) All of the following criteria: 5-yr PFS 67%  Any primary site 5-yr survival 72%  Nonpulmonary visceral metastases  Normal AFP  Any hCG  Any LDH Poor prognosis Nonseminoma (16% of cases) Any of the following criteria: 5-yr PFS 41%  Mediastinal primary 5-yr survival 48%  Nonpulmonary visceral metastases  AFP >10 000 ng/ml or  hCG > 50 000 IU/l (10 000 ng/ml) or  LDH > 10 Â ULN Seminoma No patients classified as poor prognosis

PFS = progression-free survival; AFP = a-fetoprotein; hCG = human chorionic gonadotrophin; LDH = lactate dehydrogenase; ULN = upper limit of normal range. a Prechemotherapy serum tumour markers should be assessed immediately before administration of chemotherapy (same day).

of 16.8% in unselected patients. In patients with low risk adjuvant therapy with one course at a dosage of (tumour size 4 cm and no rete testis invasion), the AUC Â 7 is an alternative to radiotherapy or surveillance in recurrence under surveillance is as low as 6% [20]. Chemo- stage I seminoma [22–25]. Two courses of , according to the IGCCCG classification, is a possible carboplatin seem to further reduce the relapse rate to the treatment for seminoma relapse under surveillance. How- order of 1–3% [18,26]; however, additional experience and ever, 70% of patients with relapse are suitable for treatment long-term observation are needed. with radiotherapy alone because of small-volume disease at the time of recurrence. Patients who experience relapse 3.4.3. Adjuvant radiotherapy after salvage radiotherapy can be effectively treated with Adjuvant radiotherapy to a paraaortic field with moderate chemotherapy [21]. The combination of carboplatin che- doses (total 20–24 Gy) will reduce the relapse rate to 1–3% motherapy and modern radiotherapy for treatment of low- [27–29]. The main concern regarding adjuvant radiotherapy stage seminoma relapse (IIA/IIB) is under investigation. is the higher risk of radiation-induced second non–germ- The overall cancer-specific survival rate under surveil- cell [30,31]. Therefore, adjuvant radiotherapy lance performed by experienced centres is 97–100% for should no longer be used in young patients. stage I seminoma [21,22]. The main drawback of surveil- lance is the need for more intensive follow-up, especially 3.4.4. Risk-adapted treatment with repeated imaging examinations of the retroperitoneal Using tumour size >4 cm and rete testis invasion, patients lymph nodes. with stage I seminoma may be subdivided into group with low and high risk of occult metastatic disease. Patients with 3.4.2. Adjuvant chemotherapy and without both risk factors have a 32% and 12% risk of A joint trial by the Medical Research Council (MRC) and the occult disease, respectively. A prospective trial based on European Organisation for Research and Treatment these risk factors (no risk factors, surveillance; both risk of Cancer (trial MRC TE19), which compared one cycle of factors, two courses of carboplatin AUC Â 7) showed the carboplatin (dosage of area under the curve [AUC] Â 7) with feasibility of a risk-adapted approach, and patients without adjuvant radiotherapy, did not show a significant difference either risk factor had a 6.0–14.8% risk of relapse at 5 yr. This with regard to recurrence rate, time to recurrence, and is why adjuvant treatment is not recommended for patients survival after a median follow-up of 4 yr [23–25]. Therefore, with very low risk (Table 5). Patients in the high-risk group 1058 EUROPEAN UROLOGY 68 (2015) 1054–1068

Table 5 – Guidelines for the treatment of stage I seminoma

GR

Surveillance is a recommended management option (if facilities are available and the patient is compliant) A * Carboplatin-based chemotherapy (one course at AUC Â 7) is recommended A Adjuvant treatment is not recommended for patients at very low risk A Radiotherapy is not recommended as adjuvant treatment A

* Upgraded following panel consensus. GR = grade of recommendation; AUC = area under curve. treated with carboplatin experienced a relapse rate of more than 200 patients, some with a median follow-up of 1.4–3.2% at a mean follow-up of 34 mo [20,32]. nearly 7.9 yr [39], a relapse rate of only 2.7% was reported, with very little long-term toxicity. 3.5. Treatment of clinical stage I nonseminoma Two cycles of -based adjuvant chemotherapy do not seem to adversely affect fertility or sexual activity Up to 30% of patients with clinical stage I nonseminomatous [41]. However, the very long-term (>20 yr) side effects of germ cell tumour (NSGCT) have subclinical metastases and adjuvant chemotherapy in this setting are currently will relapse if surveillance alone is undertaken after unknown, especially the long-term cardiovascular effects orchiectomy. [42]. In a randomised trial, one course of adjuvant BEP in The decision regarding adjuvant treatment should patients with clinical stage I nonseminoma significantly always be based on a thorough discussion with the patient, increased the 2-yr recurrence-free survival to 99.41% taking into account the advantages and disadvantages compared to primary retroperitoneal lymph node dissec- described and the patient’s individual situation. tion (RPLND). Of 174 patients who received one course of BEP, 43% had high-risk features (>pT1) [43].Ina 3.5.1. Surveillance community-based prospective nonrandomised study, the The largest studies on a surveillance strategy indicate a relapse rate at 5 yr among 490 patients who received cumulative relapse rate of 30%, with 80% of relapses BEP Â 1 was 3.2% for patients with lymphovascular invasion occurring during the first 12 mo of follow-up, 12% during (LVI) and 1.6% for patients without LVI [44]. After a median the second year, and 6% during the third year, decreasing to follow-up of 8.1 yr, the relapse rate was 2.3% for all patients, 1% thereafter [33–36]. Approximately 35% of patients who 3.4% for those with LVI, and 1.3% for those without LVI experience relapse have normal levels of serum tumour [45]. A reduction from two to one BEP cycle considerably markers at relapse. Some 60% of relapses are in the improves the risk-benefit ratio of adjuvant chemotherapy retroperitoneum. According to overall cancer-specific (Table 6). survival data, surveillance within an experienced surveil- lance programme can be safely offered to patients with 3.5.3. Risk-adapted treatment non–risk-stratified clinical stage I nonseminoma as long as Risk-adapted treatment is an alternative to surveillance for they are compliant and informed about the expected all patients with stage I NSGCT. Risk-adapted treatment is recurrence rate and the salvage treatment [37,38]. based on vascular invasion. Similar survival rates and a final cure rate close to 100% can be achieved (Table 7 and Fig. 1) 3.5.2. Adjuvant chemotherapy [39,40,44–47]. Patients with clinical stage I NSGCT have a 14–48% risk of recurrence within 2 yr after orchiectomy. Adjuvant 3.5.4. RPLND chemotherapy with two courses of cisplatin, , In view of the high cancer-specific survival rates for and (BEP) was introduced in 1996 in a surveillance with salvage treatment in cases of relapse prospective MRC trial [39]. Subsequently, adjuvant chemo- and the low relapse rates if adjuvant chemotherapy therapy was mainly given to patients with high risk is chosen, the role of primary diagnostic RPLND has (vascular invasion present) [39,40]. In these series involving diminished. Higher rates of in-field recurrences and of

Table 6 – Guidelines for the treatment of stage I nonseminomatous germ cell tumour (NSGCT)

LE GR

Patients with stage I NSGCT should be informed about all adjuvant treatment options after orchiectomy (surveillance, adjuvant 2a A * chemotherapy, and RPLND) including treatment-specific recurrence rates and acute and long-term side effects Surveillance and risk-adapted treatment based on vascular invasion (Table 6) are the recommended treatment options 2a A * If patients are not willing to undergo surveillance, one course of BEP as adjuvant treatment was superior to RPLND in terms of 1b A * recurrence rate in a community-based study Salvage treatment of patients with recurrence during surveillance consists of three or four courses of BEP chemotherapy followed by 2a A postchemotherapy RPLND if necessary

GR = grade of recommendation; LE = level of evidence; RPLND = retroperitoneal lymph node dissection. EUROPEAN UROLOGY 68 (2015) 1054–1068 1059

Table 7 – Risk-adapted treatment for clinical stage I based on vascular invasion

Stage LE GR

Stage IA (pT1, no vascular invasion): low risk Surveillance is recommended if the patient is willing and able to comply 2a A For patients not willing (or suitable) to undergo surveillance, adjuvant chemotherapy with one course of BEP is recommended 2a A * Stage IB (pT2–pT4): high risk Primary chemotherapy with one course of BEP is recommended 2a A * Patients should be informed about the advantages and disadvantages of one course of BEP Surveillance and nerve-sparing RPLND are options for those not willing to undergo adjuvant chemotherapy; if RPLND reveals pathologic A * stage II, further chemotherapy and observation should be discussed with each patient * Upgraded following panel consensus. BEP = cisplatin, etoposide, bleomycin; GR = grade of recommendation; LE = level of evidence; RPLND = retroperitoneal lymph node dissection. [(Fig._1)TD$IG]

Fig. 1 – Risk-adapted treatment in patients with clinical stage I nonseminoma. All treatment options need to be discussed with individual patients to allow them to make an informed decision as to their further care. BEP = cisplatin, etoposide, bleomycin; CS = clinical stage; IGCCCG = International Germ Cell Cancer Collaborative Group; RLNPD = retroperitoneal lymph node dissection; VIP = etoposide, cisplatin, . complications have been reported for RPLND performed in with particular expertise in testis tumour management a multicentre setting [43,48]. Thus, nerve-sparing RPLND, [49]. if indicated, should be performed by an experienced surgeon in a specialised centre. If there is an indication to 3.6. Treatment of patients with metastatic disease perform a staging RPLND, laparoscopic or robot-assisted RPLND is feasible in expert hands. This minimally First-line treatment of metastatic germ-cell tumours depends invasive approach is not recommended as the standard on the of the primary tumour and prognostic groups approach outside of a specialised laparoscopic centre as defined by the IGCCCG (Tables 4 and 8) [10]. 1060 EUROPEAN UROLOGY 68 (2015) 1054–1068

Table 8 – Guidelines for the treatment of metastatic germ-cell tumours

LE GR

Low-volume stage IIA/B NSGCT with elevated markers should be treated similarly to advanced NSGCT with good or intermediate 2A prognosis, with three or four cycles of BEP In stage IIA/B NSGCT without marker elevation, histology can be obtained by RPLND or biopsy; staging can be repeated after 6 wk 3B of surveillance before the final decision on further treatment In metastatic NSGCT (stage IIC) with good prognosis, three courses of BEP is the primary treatment of choice 1 A In metastatic NSGCT with intermediate prognosis, the primary treatment of choice is four courses of standard BEP 1 A In metastatic NSGCT with poor prognosis, the primary treatment of choice is one cycle of BEP, followed by tumour 1A marker assessment after 3 wk; in the case of an unfavourable decline, chemotherapy intensification can be initiated; in the case of a favourable decline, BEP should be continued up to a total of four cycles Surgical resection of residual masses after chemotherapy in NSGCT is indicated in the case of visible residual masses and 2A when serum levels of tumour markers are normal or normalising Stage IIA/B seminoma can be treated initially with radiotherapy; when necessary, chemotherapy can be used as a salvage treatment 2B according to the same schedule as for the corresponding NSGCT prognostic groups In stage IIA/B seminoma, chemotherapy (3 Â BEP or 4 Â EP with good prognosis) is an alternative to radiotherapy; 3 Â BEP and 1A 4 Â EP appear to achieve similar levels of disease control Stage IIC seminoma should be treated with primary chemotherapy according to the same principles used for NSGCT 1 A

BEP = cisplatin, etoposide, bleomycin; EP = etoposide, cisplatin; GR = grade of recommendation; LE = level of evidence; NSGCT = nonseminomatous germ cell tumour; BEP = cisplatin, etoposide, bleomycin; RPLND = retroperitoneal lymph node dissection.

Table 9 – Construction of the International Germ Cell Cancer Collaborative Group-2 prognostic score [50]

Variable Points

–1 0 1 2 3

Histology Seminoma Nonseminoma Primary site Gonadal Retroperitoneal Mediastinal Response CR/PRm– PRm+/SD PD PFI >3mo 3mo AFP salvage Normal <1000 ng/ml 1000 ng/ml hCG salvage <1000 ng/ml 1000 ng/ml LBB No Yes

AFP = a-fetoprotein; CR = complete remission; hCG = human chorionic gonadotrophin; LBB = liver, bone, brain metastases; PD = progressive disease; PFI = progression-free interval; PRm– = partial remission, negative markers; PRm+ = partial remission, positive markers; SD = stable disease.

In patients with relapse, a new prognostic score seminoma has been radiotherapy, with relapse rates of including response to first-line therapy can be used to 9–24% reported [51,52]. Accumulating data on long-term estimate patient outcome following salvage chemotherapy morbidity, such as a higher risk of cardiovascular events and (Tables 9 and 10) [50]. risk of second malignancies following radiotherapy, have led to concern. Most reports refer to patients irradiated with 3.6.1. Stage IIA/B seminoma larger target volumes and higher doses, but there are also An observation period of 8 wk with a second staging is more recent studies on patients treated with more modern recommended for patients with enlarged lymph nodes radiotherapy [53]. The radiation dose delivered for stage IIA without marker elevation, unless a biopsy verifies meta- and IIB is approximately 30 and 36 Gy, respectively, and the static disease. Treatment should not be initiated unless field size is enlarged. This technique yields relapse-free metastatic disease is unequivocal, (eg, growth or positive survival of 92% for stage IIA and 90% for stage IIB, with OS of biopsy). To date, the standard treatment for stage IIA/B almost 100% [51,52]. For stage IIA/B, chemotherapy with three courses of BEP or four courses with etoposide and cisplatin (EP) is an Table 10 – Progression-free survival (PFS) and overall survival (OS) alternative to radiotherapy. There are no randomised estimates for all patients according to the International Germ Cell Cancer Collaborative Group-2 prognostic score [50] studies comparing radiotherapy and chemotherapy. Al- though more toxic than radiotherapy in the short-term, Score Patients HR 2-yr PFS 3-yr OS three courses of BEP or four courses of EP achieves a similar (n = 1435) n (%) (%) (%) level of disease control. Very low 76 (5.3) 1 75.1 77.0 Single-agent carboplatin is not an alternative to Low 257 (17.9) 2.07 52.6 69.0 standard EP or BEP chemotherapy for metastatic disease Intermediate 646 (45.0) 2.88 42.8 57.3 (Fig. 2) [54]. High 351 (24.5) 4.81 26.4 31.7 Very high 105 (7.3) 8.95 11.5 14.7 Missing 159 3.6.2. Stage IIA/B non-seminoma Patients with clinical stage II NSGCT and elevated marker HR = hazard ratio. levels should receive chemotherapy according to the EUROPEAN UROLOGY 68 (2015) 1054–1068 1061 [(Fig._2)TD$IG]

Fig. 2 – Treatment options for patients with clinical stage IIA and IIB seminoma. BEP = cisplatin, etoposide, bleomycin; EP = etoposide, cisplatin.

IGCCCG classification, followed by residual tumour bleomycin, EP Â 4 should be given. Postchemotherapy resection if indicated (Table 8). Patients without elevated masses should be managed as described in Section 3.7. tumour marker levels can be managed by primary RPLND or surveillance to clarify the clinical stage [55]. 3.6.3.2. Seminoma with intermediate prognosis. For patients with A stable or growing lesion indicates either or an intermediate-risk seminoma, four cycles of BEP or etopo- undifferentiated malignant tumour. An alternative to side, cisplatin, and ifosfamide (VIP) are recommended, surveillance for marker-negative stage IIA/B nonseminoma although no randomised trial has focused specifically on with suspicion of an undifferentiated malignant tumour is a this rare group of patients. (CT-guided) biopsy, if technically possible. There are insufficient data on positron emission tomography (PET) 3.6.3.3. Nonseminoma with good prognosis. For nonseminoma, scans in this situation. When primary chemotherapy is the primary treatment of choice for metastatic disease in contraindicated or is refused by the patient, primary nerve- patients with good prognosis according to the IGCCCG risk sparing RPLND represents a viable option. classification is three cycles of BEP combination chemo- Primary chemotherapy and primary RPLND are compa- therapy. This regimen is superior to cisplatin, , rable options in terms of outcome, but differ in their side and bleomycin (PVB) in patients with advanced disease. effects and toxicity, so the patient should be involved in While data show that a 3-d regimen of combination selecting the treatment of choice [55,56]. The cure rate for chemotherapy is equally effective as a 5-d regimen, it is either approach is close to 98% [57,58]. associated with higher toxicity when four cycles are used [61], so the 5-d BEP regimen is recommended. In selected 3.6.3. Metastatic disease (stages IIC and III) cases in which bleomycin is contraindicated, four cycles of 3.6.3.1. Seminoma with good prognosis. For metastatic semi- EP can be given [9]. Therapy should be given without dose noma, very limited data are available from randomised reductions at 21-d intervals; delaying the subsequent trials and they indicate that a cisplatin-based regimen is chemotherapy cycle is justified only in cases of fever with preferable to carboplatin chemotherapy [59]. Recent data granulocytopaenia <1000/mm3 or thrombocytopaenia indicate that EP Â 4 results in cure in almost all cases of <100 000/IU. There is no indication for prophylactic seminomatous germ-cell cancers with good prognosis administration of haematopoietic growth factors. However, [60]. Standard treatment in this setting should therefore if infectious complications occur during chemotherapy or be BEP Â 3orEPÂ 4. In the case of contraindications to the treatment interval is delayed because of myelotoxicity, 1062 EUROPEAN UROLOGY 68 (2015) 1054–1068 prophylactic administration of granulocyte-colony stimu- tumour growth. Early therapy crossover to a completely lating factor (G-CSF) is recommended in subsequent cycles. new regimen is only indicated for a documented increase in marker levels after two courses of chemotherapy. Such 3.6.3.4. Nonseminoma with intermediate prognosis. Patients with patients are usually candidates for new drug trials. intermediate IGCCCG prognosis have a 5-yr survival rate of Patients with a low-level human chorionic gonadotro- approximately 80%. The available data support four cycles phin (hCG) plateau after treatment should be observed to of BEP as standard treatment [10]. A randomised trial see whether complete normalisation occurs. In patients comparing four cycles of BEP without and with addition of with a low serum a-fetoprotein (AFP) plateau after paclitaxel (T-BEP) found no significant improvement in OS chemotherapy, surgery of residual masses is indicated, [62]. with postsurgical AFP monitoring. Salvage chemotherapy is only indicated for a documented increase in marker level. 3.6.3.5. Nonseminoma with poor prognosis. For patients with poor-prognosis nonseminoma as defined by the IGCCCG 3.7. Residual tumour resection and favourable marker decline, standard treatment consists of four cycles of BEP. Four cycles of cisplatin, etoposide, and 3.7.1. Seminoma ifosfamide (PEI) have the same effect, but the myelotoxicity A residual seminoma mass, irrespective of size, should not is higher. The 5-yr progression-free survival (PFS) is be primarily resected but should be controlled using between 45% and 50%. Three randomised trials showed imaging investigations and tumour markers. no OS advantage for high-dose chemotherapy in the overall Fluorodeoxyglucose (FDG)-PET has a high negative group of patients with poor prognosis [35,63,64]. predictive value in patients with residual masses after However, patients with a slow decline in tumour marker treatment of seminoma. False-positive results are less levels after the first or second cycle represent a prognos- frequent when scans are scheduled >2 mo after chemo- tically inferior subgroup [63]. therapy [71]. An international randomised phase 3 trial conducted in In the case of a postchemotherapy mass that is still 263 patients with IGCCCG poor-risk NSGCT demonstrated positive at reclassification FDG-PET with no volume that intensifying treatment with dose-dense chemotherapy increase, a second FDG-PET scan should be performed improves PFS but not OS in patients with an early 6 wk later. Alternatively, a biopsy should be carried out to unfavourable decline in tumour marker levels [65].On ascertain if persistent disease is present. In such cases and in the basis of results from this trial, patients with an cases with progressive disease, salvage therapy is indicated unfavourable decline in tumour marker levels after one (usually chemotherapy or radiotherapy). Patients with cycle of BEP should be switched to a more intensive persistent and progressing hCG elevation after first-line [66]. Further prospective trials and chemotherapy should immediately undergo salvage registries are planned to further validate this approach. A chemotherapy. Patients with progressing disease without matched-pair analysis comparing high-dose to convention- hCG progression should undergo histologic verification al treatment revealed a better survival rate [67], so patients (eg, biopsy or mini-invasive or open surgery) before salvage with poor prognosis should still be treated in ongoing chemotherapy is administered. When RPLND is indicated, prospective trials or registries whenever available. this should be performed in referral centres, as residual Patients meeting poor-prognosis criteria should be seminoma masses may be difficult to remove because of transferred to a reference centre, as better outcomes were extensive fibrosis. Ejaculation may be preserved in these observed for patients with intermediate or poor prognosis cases. when treated in a clinical trial in a high-volume centre [3,68]. There are no general recommendations for treatment 3.7.2. Nonseminoma modifications for patients with poor general condition Following first-line BEP chemotherapy, only 6–10% of (Karnofsky performance score <50%) or extended liver residual masses contain viable cancer, 50% contain mature infiltration (>50%), but two small studies indicate that a teratoma, and 40% contain necrotic-fibrotic tissue [72]. first cycle of dose-reduced therapy may reduce acute FDG-PET is not indicated for restaging after chemother- mortality without compromising long-term outcome apy [73]. In cases of complete remission after first-line [69,70]. However, the number of subsequent cycles of chemotherapy (no visible tumour), tumour resection is not full-dose therapy should not be reduced after a first low- indicated [74,75]. Residual tumour resection is mandatory dose induction cycle. for all patients with a residual mass >1 cm along the short axis at cross-sectional CT imaging [76]. The role of surgery 3.6.3.6. Restaging and further treatment. Restaging involves in patients with retroperitoneal residual lesions <1cmis imaging investigations and reevaluation of tumour mar- debated. There is still a risk of residual cancer or teratoma, kers. If marker levels decline and a stable or regressive although the vast majority of patients (>70%) harbour tumour is observed, chemotherapy is completed (three or fibronecrotic tissue [77]. Proponents of postchemotherapy four cycles, depending on the initial stage) [10]. If marker RPLND for all patients refer to the fact that both teratoma levels decline but metastases continue to grow, tumour and vital malignant germ-cell tumours are still found resection is obligatory after termination of induction after radiologic complete remission in lesions <10 mm [78]. therapy, other than in an emergency, according to local The alternative is to place patients with residual disease EUROPEAN UROLOGY 68 (2015) 1054–1068 1063

Table 11 – Standard PEI/VIP, TIP, and GIP chemotherapy regimens (interval 21 d)

Regimen Chemotherapy agents Dosage Cycle duration

PEI/VIP Cisplatin a 20 mg/m2 Days 1–5 Etoposide 75–100 mg/m2 Days 1–5 Ifosfamide b 1.2 g/m2 Days 1–5 TIP Paclitaxel 250 mg/m2 c 24-h continuous infusion day 1 Ifosfamide b 1.5 g/m2 Days 2–5 Cisplatin a 25 mg/m2 Days 2–5 GIP 1000 mg/m2 Days 1 and 5 Ifosfamide 1200 mg/m2 Days 1–5 Cisplatin 20 mg/m2 Days 1–5

PEI/VIP = cisplatin, etoposide, ifosfamide; TIP = paclitaxel, ifosfamide, cisplatin; GIP = gemcitabine, ifosfamide, cisplatin. a Plus hydration. b Plus mesna protection. c A Medical Research Council schedule uses paclitaxel at 175 mg/m2 in a 3-h infusion. of <1 cm on an observation protocol, for which the followed by ad hoc reconstructive surgery (eg, vascular recurrence rate is 6–9%, depending on the follow-up time interventions such as vena cava or aortic prostheses) [74,75]. Patients treated with first-line chemotherapy [88,89]. In patients with intermediate or poor risk and should be informed about a lifelong risk of recurrence of residual disease of >5 cm, the probability of vascular approximately 10% before consenting to observation of procedures is as high as 20% [90]. This intense (‘‘maximal’’) residual lesions of <1 cm. Patients after salvage chemo- surgery must therefore be referred to specialised centres therapy or high-dose chemotherapy in first or subsequent capable of interdisciplinary surgery (hepatic resections, salvage situations harbour vital tumour at a much higher vessel replacement, spinal neurosurgery, thoracic surgery). rate. Therefore, there is an indication to perform surgery in Patients treated within such centres benefit from a salvage patients even with residual disease <1cm[74,75].If significant reduction in perioperative mortality from 6% residual surgery is indicated, all areas of primary metastatic to 0.8% [4]. In addition, specialised urologic surgeons are sites must be completely resected within 2–6 wk of capable of reducing the local recurrence rate from 16% to 3% chemotherapy completion. If technically feasible, a bilateral with a higher rate of complete resections [5]. Surgery of nerve-sparing procedure should be performed. There is resectable disease even after (multiple) salvage treatments growing evidence that template resection with unilateral remains a potentially curative option with durable com- preservation of nerves in selected patients yields equivalent plete remissions in the order of 20% [91,92]. long-term results to bilateral systematic resection in all patients. Mere resection of the residual tumour (so-called 3.8. Systemic salvage treatment for relapsing or refractory lumpectomy) should not be performed [75,77,79–82]. disease Laparoscopic RPLND may yield similar outcomes to the open procedure in very selected cases with small residual Cisplatin-based combination salvage chemotherapy will disease and in experienced hands, but this strategy is not result in long-term remissions in approximately 50% of recommended outside specialised laparoscopic centres patients with relapse after first-line chemotherapy, but the with particular expertise in testis tumour management results are highly dependent on several prognostic factors. [83–85]. The regimens of choice are four cycles of a triplet regimen including cisplatin and ifosfamide plus etoposide, paclitax- 3.7.3. Surgical timing in the case of multiple sites el, or potentially gemcitabine as a third agent (Table 11) In general, surgery for residual masses should start at the [93]. No randomised trial has ever compared these regi- location with the highest volume of residual disease. The mens. Owing to their potentially lethal risk of haematologic histology may diverge in different organ sites. In cases with toxicity, these regimens should be used with G-CSF support retroperitoneal and lung residual masses, the presence of and by well-trained oncologists. At present it is impossible fibronecrotic tissue in the retroperitoneum is associated to determine whether conventional dosing for cisplatin- with a probability as high as 90% that lung masses contain based combination chemotherapy is sufficient as first- the same histology. Resection of contralateral pulmonary salvage treatment or whether early intensification of lesions is not mandatory if pathologic examination of first-salvage treatment with high-dose chemotherapy lesions from the first lung shows complete necrosis. should be used. However, there is evidence from large However, discordant histology between the two lungs retrospective analyses that there are different prognostic may occur in up to 20% of patients [86,87]. groups in the case of relapse after first-line chemotherapy [50,94,95], and the Lorch-Beyer score has resulted in five 3.7.4. Surgical quality and intensity prognostic subgroups (Table 9). A second large analysis in Postchemotherapy surgery is always demanding. Approxi- this cohort of 1600 patients showed an OS improvement of mately one third of patients may require a planned approximately 10–15% among patients from all prognostic intervention in which organs affected by the disease subgroups for high-dose salvage therapy compared to (eg, kidney, psoas muscle, or gross vessels) are removed, standard-dose therapy. If high-dose chemotherapy is used 1064 EUROPEAN UROLOGY 68 (2015) 1054–1068

Table 12 – Recommended minimum follow-up schedule in a surveillance policy for stage I nonseminoma [103]

Procedure Year

1 2 3 4–5

Physical examination Four times Four times Four times Once/yr Tumour markers Four times Four times Four times Once/yr Plain radiography of the chest Twice Twice Twice Twice Abdominopelvic computed tomography Twice (at 3 and 12 mo) Once (at 24 mo) Once (at 36 mo)

Table 13 – Recommended minimum follow-up schedule after retroperitoneal lymphadenectomy or adjuvant chemotherapy for stage I nonseminoma

Procedure Year

1 2 3 4–5 6–10

Physical examination Four times Four times Four times Once/yr Once/yr Tumour markers Four times Four times Four times Once/yr Once/yr Plain radiography of the chest Twice Twice Twice Abdominopelvic computed tomography Once Once Once Once/yr

Table 14 – Recommended minimum follow-up schedule for post-orchiectomy surveillance, radiotherapy, or chemotherapy for stage I seminoma [36]

Procedure Year

1 2 3–5

Physical examination Three times Three times Once/yr Tumour markers Three times Three times Once/yr Plain radiography of the chest Twice Twice Abdominopelvic computed tomography Twice Twice At 36 and 60 mo

Table 15 – Recommended minimum follow-up schedule in metastatic nonseminomatous germ cell tumour and seminoma

Procedure Year

1 2 3–5 Thereafter

Physical examination Four times Four times Twice/yr Once/yr Tumour markers Four times Four times Twice/yr Once/yr Plain radiography of the chest Four times Four times Twice/yr Once/yr Abdominopelvic CT a Twice Twice Once/yr As indicated Chest CT b,c Once/yr Once/yr Once/yr As indicated Brain CT d Once/yr Once/yr Once/yr As indicated

CT = computed tomography. a Abdominal CT must be performed at least annually if teratoma is found in the retroperitoneum. b If postchemotherapy evaluation in a seminoma patient shows any mass >3 cm, the appropriate CT should be repeated 2 and 4 mo later to ensure that the mass is continuing to regress. If available, fluorodeoxyglucose positron emission tomography/CT can be performed. c Chest CT is indicated if an abnormality is detected on a plain chest x-ray and after pulmonary resection. d In patients with headaches, focal neurological findings, or any central nervous system symptoms.

as a salvage treatment, sequential cycles of high-dose with or without surgery for residual tumour surgery. carboplatin and etoposide should be preferred over a single According to a pooled analysis, late relapse occurs in 1.4% high-dose regimen because the former is associated with of seminoma and 3.2% of nonseminoma cases [97,98].If fewer toxicity-related deaths [96]. It is clearly of the utmost feasible, all lesions in late-relapsing nonseminoma cases importance that these rare patients with relapse are treated should be removed by radical surgery. Patients with rapidly within clinical trials and at experienced centres. rising hCG may benefit from induction salvage chemother- apy before complete resection, but in most patients surgery 3.9. Late relapse should be performed irrespective of the level of their tumour markers to completely resect all undifferentiated Late relapse is defined as recurrence >2 yr after cure germ-cell tumour and mature teratoma with or without following chemotherapy for metastatic testicular cancer, somatic transformation [99–101]. To minimise mortality, EUROPEAN UROLOGY 68 (2015) 1054–1068 1065 late relapses should be treated only at centres experienced References in managing such patients [102]. [1] Hoffmann R, Plug I, McKee M, et al. Innovations in health care and mortality trends from five cancers in seven European countries 3.10. Follow-up schedules between 1970 and 2005. Int J Public Health 2014;59:341–50. [2] Jones A, Fergus JN, Chapman J, Houghton L. Is surveillance for stage Tables 12–15 list the recommended follow-up schedules. 1 germ cell tumours of the testis appropriate outside a specialist centre? BJU Int 1999;84:79–84. 4. Conclusions [3] Collette L, Sylvester RJ, Stenning SP, et al. Impact of the treating institution on survival of patients with ‘‘poor-prognosis’’ meta- For all stages of testicular cancer, the best cure rates are static nonseminoma. J Natl Cancer Inst 1999;91:839–46. obtained in high-volume reference centres. Patients diag- [4] Capitanio U, Jeldres C, Perrotte P, et al. Population-based study of nosed with testicular cancer should be informed about the perioperative mortality after retroperitoneal lymphadenectomy for short- and long-term side effects of radiotherapy and nonseminomatous testicular germ cell tumors. Urology 2009;74: 373–7. chemotherapy. The aim of risk-adapted treatment is to [5] Flechon A, Tavernier E, Boyle H, Meeus P, Rivoire M, Droz JP. reduce long-term treatment-related sequelae without Long-term oncological outcome after post-chemotherapy retro- compromising therapeutic efficacy. peritoneal lymph node dissection in men with metastatic non- seminomatous germ cell tumour. BJU Int 2010;106:779–85. Author contributions: Peter Albers had full access to all the data in the [6] Zengerling F, Hartmann M, Heidenreich A, et al. German second- study and takes responsibility for the integrity of the data and the opinion network for testicular cancer: sealing the leaky pipe accuracy of the data analysis. between evidence and clinical practice. Oncol Rep 2014;31: Study concept and design: Albers. 2477–81. Acquisition of data: Albers, Albrecht, Cohn-Cedermark, Algaba, Bokemeyer, [7] Albers P, Albrecht W, Algaba F, et al. EAU Guidelines on testicular Fizazi, Horwich, Laguna, Nicolai, Oldenburg. cancer: 2011 update. Eur Urol 2011;60:304–19. Analysis and interpretation of data: Albers, Albrecht, Cohn-Cedermark, [8] Howick J. Levels of evidence. Oxford, UK: Centre for Evidence- Algaba, Bokemeyer, Fizazi, Horwich, Laguna, Nicolai, Oldenburg. based Medicine; 2009,, http://www.cebm.net/oxford-centre- Drafting of the manuscript: Albers. evidence-based-medicine-levels-evidence-march-2009/. Critical revision of the manuscript for important intellectual content: Albers, [9] Sobin LH, Gospodarowicz MK, Wittekind C, editors. TNM classifi- Albrecht, Cohn-Cedermark, Algaba, Bokemeyer, Fizazi, Horwich, Laguna, cation of malignant tumors. ed. 7. Oxford, UK: Wiley-Blackwell; Nicolai, Oldenburg. 2009. p. 249–54. Statistical analysis: None. [10] Mead GM, Stenning SP. The International Germ Cell Consensus Obtaining funding: None. Classification: a new prognostic factor-based staging classification Administrative, technical, or material support: Albers. for metastatic germ cell tumours. Clin Oncol 1997;9:207–9. Supervision: Albers. [11] Dieckmann KP, Loy V. Prevalence of contralateral testicular intrae- Other: None. pithelial neoplasia in patients with testicular germ cell . J Clin Oncol 1996;14:3126–32. Financial disclosures: Peter Albers certifies that all conflicts of interest, [12] von der Maase H, Rorth M, Walbom-Jorgensen S, et al. in including specific financial interests and relationships and affiliations situ of contralateral testis in patients with testicular germ cell relevant to the subject matter or materials discussed in the manuscript cancer: study of 27 cases in 500 patients. BMJ 1986;293:1398–401. (eg, employment/affiliation, grants or funding, consultancies, honoraria, [13] Harland SJ, Cook PA, Fossa SD, et al. Intratubular germ cell neo- stock ownership or options, expert testimony, royalties, or patents filed, plasia of the contralateral testis in testicular cancer: defining a received, or pending), are the following: Peter Albers has received high risk group. J Urol 1998;160:1353–7. consultancy fees from Thieme Books, Pfizer, MSD, and Novartis; has [14] Dieckmann KP, Kulejewski M, Pichlmeier U, Loy V. Diagnosis of received speaker honoraria from Pfizer, Novartis, Sanofi-Aventis, Amgen, contralateral testicular intraepithelial neoplasia (TIN) in patients Astra Zeneca, Astellas, Hexal, and Dendreon; has participated in trials by with testicular germ cell cancer: systematic two-site are Novacea, Sanofi-Aventis, Bayer, Astra Zeneca, Novartis, and Ferring; and more sensitive than a single random biopsy. Eur Urol 2007;51: has received grants or research support from Novartis Oncology and Bayer 175–85. Oncology. Walter Albrecht has received speaker honoraria from Takeda, [15] Classen J, Dieckmann K, Bamberg M, et al. Radiotherapy with Astellas, Janssen, Amgen, and Ferring. Ferran Algaba has received a speaker 16 Gy may fail to eradicate testicular intraepithelial neoplasia: honorarium from Inibsa Laboratorios and has participated in a trial by preliminary communication of a dose-reduction trial of the Ger- Pfizer. Carsten Bokemeyer has participated in trials by Novartis and Roche; man Testicular Cancer Study Group. Br J Cancer 2003;88:828–31. has received grants or research support from Novartis, Sanofi-Aventis, [16] Petersen PM, Giwercman A, Daugaard G, et al. Effect of graded and Fresenius Biotech; is a company consultant for Merck Serono; and testicular doses of radiotherapy in patients treated for carcinoma- has received speaker honoraria from Merck Serono, Sanofi-Aventis, in-situ in the testis. J Clin Oncol 2002;20:1537–43. and Novartis. Karim Fizazi has received consultancy fees and speaker [17] Heidenreich A, Holtl W, Albrecht W, Pont J, Engelmann UH. Testis- honoraria from Amgen, Astellas, Bayer, BMS, Dendreon, Ipsen, Janssen, preserving surgery in bilateral testicular germ cell tumours. Br J Takeda, Novartis, Sanofi-Aventis; and has participated in trials by Amgen, Urol 1997;79:253–7. Sanofi-Aventis, Astra Zeneca, Janssen, Medivation, Millenium, Celgene, [18] Aparicio J, Germa JR, Garcia del Muro X, et al. Risk-adapted Oncogenex, and BMS. Alan Horwich has received honoraria and management for patients with clinical stage I seminoma: the consultation fees from Astellas. Nicola Nicolai is a consultant for Second Spanish Germ Cell Cancer Cooperative Group study. J Clin Janssen. Gabriella Cohn-Cedermark, Maria Pilar Laguna, and Jan Oldenburg Oncol 2005;23:8717–23. have nothing to disclose. [19] Albers P, Siener R, Kliesch S, et al. Risk factors for relapse in clinical Funding/Support and role of the sponsor: None. stage I nonseminomatous testicular germ cell tumors: results of 1066 EUROPEAN UROLOGY 68 (2015) 1054–1068

the German Testicular Cancer Study Group trial. J Clin Oncol [37] Kollmannsberger C, Moore C, Chi KN, et al. Non-risk-adapted 2003;21:1505–12. surveillance for patients with stage I nonseminomatous testicular [20] Aparicio J, Maroto P, Garcia del Muro X, et al. Prognostic factors for germ-cell tumors: diminishing treatment-related morbidity while relapse in stage I seminoma: a new nomogram derived from three maintaining efficacy. Ann Oncol 2010;21:1296–301. consecutive, risk-adapted studies from the Spanish Germ Cell [38] Nichols CR, Roth B, Albers P, et al. Active surveillance is the Cancer Group (SGCCG). Ann Oncol 2014;25:2173–8. preferred approach to clinical stage I testicular cancer. J Clin Oncol [21] Tandstad T, Smaaland R, Solberg A, et al. Management of semi- 2013;31:3490–3. nomatous testicular cancer: a binational prospective population- [39] Cullen MH, Stenning SP, Parkinson MC, et al. Short-course adju- based study from the Swedish Norwegian Testicular Cancer Study vant chemotherapy in high-risk stage I nonseminomatous germ Group. J Clin Oncol 2011;29:719–25. cell tumors of the testis: a Medical Research Council report. J Clin [22] Aparicio J, Garcia del Muro X, Maroto P, et al. Multicenter study Oncol 1996;14:1106–13. evaluating a dual policy of postorchiectomy surveillance and [40] Pont J, Albrecht W, Postner G, Sellner F, Angel K, Holtl W. Adjuvant selective adjuvant single-agent carboplatin for patients with clin- chemotherapy for high-risk clinical stage I nonseminomatous ical stage I seminoma. Ann Oncol 2003;14:867–72. testicular germ cell cancer: long-term results of a prospective [23] Oliver RT, Mead GM, Rustin GJ, et al. Randomized trial of carboplatin trial. J Clin Oncol 1996;14:441–8. versus radiotherapy for stage I seminoma: mature results on [41] Bohlen D, Burkhard FC, Mills R, Sonntag RW, Studer UE. Fertility relapse and contralateral testis cancer rates in MRC TE19/EORTC and sexual function following orchiectomy and 2 cycles of che- 30982 study (ISRCTN27163214). J Clin Oncol 2011;29:957–62. motherapy for stage I high risk nonseminomatous germ cell [24] Oliver RT, Mason MD, Mead GM, et al. Radiotherapy versus single- cancer. J Urol 2001;165:441–4. dose carboplatin in adjuvant treatment of stage I seminoma: a [42] Huddart RA, Norman A, Shahidi M, et al. Cardiovascular disease as randomised trial. Lancet 2005;366:293–300. a long-term complication of treatment for testicular cancer. J Clin [25] Mead GM, Fossa SD, Oliver RT, et al. Randomized trials in Oncol 2003;21:1513–23. 2466 patients with stage I seminoma: patterns of relapse and [43] Albers P, Siener R, Krege S, et al. Randomized phase III trial follow-up. J Natl Cancer Inst 2011;103:241–9. comparing retroperitoneal lymph node dissection with one course [26] Schoffski P, Ho¨hn N, Kowalski R, et al. Health-related quality of life of bleomycin and etoposide plus cisplatin chemotherapy in the (QoL) in patients with seminoma stage I treated with either adjuvant treatment of clinical stage I nonseminomatous testicular adjuvant radiotherapy (RT) or two cycles of carboplatinum che- germ cell tumors: AUO trial AH 01/94 by the German Testicular motherapy (CT): results of a randomized phase III trial of the Cancer Study Group. J Clin Oncol 2008;26:2966–72. German Interdisciplinary Working Party on Testicular Cancer. J [44] Tandstad T, Dahl O, Cohn-Cedermark G, et al. Risk-adapted treat- Clin Oncol 2007;25(18 Suppl):5050. ment in clinical stage I nonseminomatous germ cell testicular [27] Fossa SD, Horwich A, Russell JM, et al. Optimal planning target cancer: the SWENOTECA management program. J Clin Oncol volume for stage I testicular seminoma: a Medical Research 2009;27:2122–8. Council randomized trial. J Clin Oncol 1999;17:1146. [45] Tandstad T, Stahl O, Hakansson U, et al. One course of adjuvant BEP [28] Jones WGFS, Mead GM, et al. A randomized trial of two radiother- in clinical stage I nonseminoma mature and expanded results apy schedules in the adjuvant treatment of stage I seminoma from the SWENOTECA group. Ann Oncol 2014;25:2167–72. (MRC TE 18). Eur J Cancer 2001;37:572. [46] Maroto P, Garcia del Muro X, Aparicio J, et al. Multicentre risk- [29] Melchior D, Hammer P, Fimmers R, Schuller H, Albers P. Long term adapted management for stage I non-seminomatous germ cell results and morbidity of paraaortic compared with paraaortic and tumours. Ann Oncol 2005;16:1915–20. iliac adjuvant radiation in clinical stage I seminoma. Anticancer [47] Tandstad T, Cohn-Cedermark G, Dahl O, et al. Long-term follow-up Res 2001;21:2989–93. after risk-adapted treatment in clinical stage 1 (CS1) nonsemino- [30] van den Belt-Dusebout AW, de Wit R, Gietema JA, et al. Treatment- matous germ-cell testicular cancer (NSGCT) implementing adju- specific risks of second malignancies and cardiovascular disease in vant CVB chemotherapy. A SWENOTECA study. Ann Oncol 5-year survivors of testicular cancer. J Clin Oncol 2007;25:4370–8. 2010;21:1858–63. [31] Horwich A, Fossa SD, Huddart R, et al. Second cancer risk and [48] Heidenreich A, Albers P, Hartmann M, et al. Complications of mortality in men treated with radiotherapy for stage I seminoma. primary nerve sparing retroperitoneal lymph node dissection Br J Cancer 2014;110:256–63. for clinical stage I nonseminomatous germ cell tumors of the [32] Aparicio J, Maroto P, Garcia del Muro X, et al. Risk-adapted testis: experience of the German Testicular Cancer Study Group. treatment in clinical stage I testicular seminoma: the third Span- J Urol 2003;169:1710–4. ish Germ Cell Cancer Group study. J Clin Oncol 2011;29:4677–81. [49] Neyer M, Peschel R, Akkad T, et al. Long-term results of laparoscopic [33] Freedman LS, Parkinson MC, Jones WG, et al. Histopathology in the retroperitoneal lymph-node dissection for clinical stage I nonsemi- prediction of relapse of patients with stage I testicular teratoma nomatous germ-cell testicular cancer. J Endourol 2007;21:180–3. treated by orchidectomy alone. Lancet 1987;2:294–8. [50] Lorch A, Beyer J, Bascoul-Mollevi C, et al. Prognostic factors in [34] Read G, Stenning SP, Cullen MH, et al. Medical Research Council patients with metastatic germ cell tumors who experienced treat- prospective study of surveillance for stage I testicular teratoma. J ment failure with cisplatin-based first-line chemotherapy. J Clin Clin Oncol 1992;10:1762–8. Oncol 2010;28:4906–11. [35] Daugaard G, Skoneczna I, Aass N, et al. A randomized phase III [51] Classen J, Schmidberger H, Meisner C, et al. Radiotherapy for study comparing standard dose BEP with sequential high-dose stages IIA/B testicular seminoma: final report of a prospective cisplatin, etoposide, and ifosfamide (VIP) plus stem-cell support in multicenter clinical trial. J Clin Oncol 2003;21:1101–6. males with poor-prognosis germ-cell cancer. An intergroup study [52] Chung PW, Gospodarowicz MK, Panzarella T, et al. Stage II testic- of EORTC, GTCSG, and Grupo Germinal (EORTC 30974). Ann Oncol ular seminoma: patterns of recurrence and outcome of treatment. 2011;22:1054–61. Eur Urol 2004;45:754–9. [36] Kollmannsberger C, Tandstad T, Bedard PL, et al. Patterns of [53] Horwich A, Dearnaley DP, Sohaib A, Pennert K, Huddart RA. relapse in patients with clinical stage I testicular cancer managed Neoadjuvant carboplatin before radiotherapy in stage IIA and with active surveillance. J Clin Oncol 2015;33:51–7. IIB seminoma. Ann Oncol 2013;24:2104–7. EUROPEAN UROLOGY 68 (2015) 1054–1068 1067

[54] Krege S, Boergermann C, Baschek R, et al. Single agent carboplatin [68] Thibault C, Fizazi K, Barrios D, et al. Compliance with guidelines for CS IIA/B testicular seminoma. A phase II study of the German and correlation with outcome in patients with advanced germ-cell Testicular Cancer Study Group (GTCSG). Ann Oncol 2006;17: tumours. Eur J Cancer 2014;50:1284–90. 276–80. [69] Massard C, Plantade A, Gross-Goupil M, et al. Poor prognosis [55] Stephenson AJ, Bosl GJ, Motzer RJ, Bajorin DF, Stasi JP, Sheinfeld J. nonseminomatous germ-cell tumours (NSGCTs): should chemo- Nonrandomized comparison of primary chemotherapy and retro- therapy doses be reduced at first cycle to prevent acute respiratory peritoneal lymph node dissection for clinical stage IIA and IIB distress syndrome in patients with multiple lung metastases? Ann nonseminomatous germ cell testicular cancer. J Clin Oncol Oncol 2010;21:1585–8. 2007;25:5597–602. [70] Gillessen S, Powles T, Lim L, Wilson P, Shamash J. Low-dose [56] Weissbach L, Bussar-Maatz R, Flechtner H, Pichlmeier U, Hart- induction chemotherapy with Baby-BOP in patients with meta- mann M, Keller L. RPLND or primary chemotherapy in clinical static germ-cell tumours does not compromise outcome: a single- stage IIA/B nonseminomatous germ cell tumors? Results of a centre experience. Ann Oncol 2010;21:1589–93. prospective multicenter trial including quality of life assessment. [71] De Santis M, Becherer A, Bokemeyer C, et al. 2-18Fluoro-deoxy-D- Eur Urol 2000;37:582–94. glucose positron emission tomography is a reliable predictor for [57] Horwich A, Norman A, Fisher C, Hendry WF, Nicholls J, Dearnaley viable tumor in postchemotherapy seminoma: an update of the DP. Primary chemotherapy for stage II nonseminomatous germ prospective multicentric SEMPET trial. J Clin Oncol 2004;22: cell tumors of the testis. J Urol 1994;151:72–7. 1034–9. [58] Donohue JP, Thornhill JA, Foster RS, Bihrle R, Rowland RG, Einhorn [72] Carver BS, Serio AM, Bajorin D, et al. Improved clinical outcome in LH. The role of retroperitoneal lymphadenectomy in clinical stage recent years for men with metastatic nonseminomatous germ cell B testis cancer: the Indiana University experience (1965 to 1989). J tumors. J Clin Oncol 2007;25:5603–8. Urol 1995;153:85–9. [73] Oechsle K, Hartmann M, Brenner W, et al. [18F]Fluorodeoxyglucose [59] Bokemeyer C, Kollmannsberger C, Stenning S, et al. Metastatic positron emission tomography in nonseminomatous germ cell seminoma treated with either single agent carboplatin or cisplat- tumors after chemotherapy: the German multicenter positron in-based combination chemotherapy: a pooled analysis of two emission tomography study group. J Clin Oncol 2008;26:5930–5. randomised trials. Br J Cancer 2004;91:683–7. [74] Kollmannsberger C, Daneshmand S, So A, et al. Management of [60] Fizazi K, Delva R, Caty A, et al. A risk-adapted study of cisplatin and disseminated nonseminomatous germ cell tumors with risk- etoposide, with or without ifosfamide, in patients with metastatic based chemotherapy followed by response-guided postche- seminoma: results of the GETUG S99 multicenter prospective motherapy surgery. J Clin Oncol 2010;28:537–42. study. Eur Urol 2014;65:381–6. [75] Ehrlich Y, Brames MJ, Beck SD, Foster RS, Einhorn LH. Long-term [61] de Wit R, Roberts JT, Wilkinson PM, et al. Equivalence of three or follow-up of Cisplatin combination chemotherapy in patients with four cycles of bleomycin, etoposide, and cisplatin chemotherapy disseminated nonseminomatous germ cell tumors: is a postche- and of a 3- or 5-day schedule in good-prognosis germ cell cancer: a motherapy retroperitoneal lymph node dissection needed after randomized study of the European Organization for Research and complete remission? J Clin Oncol 2010;28:531–6. Treatment of Cancer Genitourinary Tract Cancer Cooperative [76] Steyerberg EW, Keizer HJ, Habbema JD. Prediction models for the Group and the Medical Research Council. J Clin Oncol 2001;19: histology of residual masses after chemotherapy for metastatic 1629–40. testicular cancer. ReHiT Study Group. Int J Cancer 1999;83:856–9. [62] de Wit R, Skoneczna I, Daugaard G, et al. Randomized phase III [77] Carver BS, Shayegan B, Serio A, Motzer RJ, Bosl GJ, Sheinfeld J. study comparing paclitaxel-bleomycin, etoposide, and cisplatin Long-term clinical outcome after postchemotherapy retroperito- (BEP) to standard BEP in intermediate-prognosis germ-cell cancer: neal lymph node dissection in men with residual teratoma. J Clin intergroup study EORTC 30983. J Clin Oncol 2012;30:792–9. Oncol 2007;25:1033–7. [63] Motzer RJ, Nichols CJ, Margolin KA, et al. Phase III randomized trial [78] Oldenburg J, Alfsen GC, Lien HH, Aass N, Waehre H, Fossa SD. of conventional-dose chemotherapy with or without high-dose Postchemotherapy retroperitoneal surgery remains necessary in chemotherapy and autologous hematopoietic stem-cell rescue as patients with nonseminomatous testicular cancer and minimal first-line treatment for patients with poor-prognosis metastatic residual tumor masses. J Clin Oncol 2003;21:3310–7. germ cell tumors. J Clin Oncol 2007;25:247–56. [79] Baniel J, Foster RS, Rowland RG, Bihrle R, Donohue JP. Complica- [64] Droz JP, Kramar A, Biron P, et al. Failure of high-dose cyclophos- tions of post-chemotherapy retroperitoneal lymph node dissec- phamide and etoposide combined with double-dose cisplatin and tion. J Urol 1995;153:976–80. bone marrow support in patients with high-volume metastatic [80] Fizazi K, Oldenburg J, Dunant A, et al. Assessing prognosis and nonseminomatous germ-cell tumours: mature results of a ran- optimizing treatment in patients with postchemotherapy viable domised trial. Eur Urol 2007;51:739–48. nonseminomatous germ-cell tumors (NSGCT): results of the sCR2 [65] Fizazi K, Pagliaro L, Laplanche A, et al. Personalised chemotherapy international study. Ann Oncol 2008;19:259–64. based on tumour marker decline in poor prognosis germ-cell [81] Heidenreich A, Pfister D, Witthuhn R, Thuer D, Albers P. Postche- tumours (GETUG 13): a phase 3, multicentre, randomised trial. motherapy retroperitoneal lymph node dissection in advanced Lancet Oncol 2014;15:1442–50. testicular cancer: radical or modified template resection. Eur Urol [66] Olofsson SE, Tandstad T, Jerkeman M, et al. Population-based 2009;55:217–26. study of treatment guided by decline in patients [82] Beck SD, Foster RS, Bihrle R, Donohue JP, Einhorn LH. Is full bilateral with metastatic nonseminomatous : a report from retroperitoneal lymph node dissection always necessary for post- the Swedish-Norwegian Testicular Cancer Group. J Clin Oncol chemotherapy residual tumor? Cancer 2007;110:1235–40. 2011;29:2032–9. [83] Calestroupat JP, Sanchez-Salas R, Cathelineau X, et al. Postche- [67] Bokemeyer C, Kollmannsberger C, Meisner C, et al. First-line high- motherapy laparoscopic retroperitoneal lymph node dissection dose chemotherapy compared with standard-dose PEB/VIP in nonseminomatous germ-cell tumor. J Endourol 2009;23: chemotherapy in patients with advanced germ cell tumors: A 645–50. multivariate and matched-pair analysis. J Clin Oncol 1999;17: [84] Busch J, Magheli A, Erber B, et al. Laparoscopic and open post- 3450–6. chemotherapy retroperitoneal lymph node dissection in patients 1068 EUROPEAN UROLOGY 68 (2015) 1054–1068

with advanced testicular cancer—a single center analysis. BMC metastatic germ-cell tumors (GCT): a prospective multicenter Urol 2012;12:15. GETUG phase II trial. Ann Oncol 2014;25:987–91. [85] Arai Y, Kaiho Y, Yamada S, et al. Extraperitoneal laparoscopic [94] Lorch A, Kollmannsberger C, Hartmann JT, et al. Single versus retroperitoneal lymph node dissection after chemotherapy for sequential high-dose chemotherapy in patients with relapsed or nonseminomatous testicular germ-cell tumor: surgical and on- refractory germ cell tumors: a prospective randomized multicen- cological outcomes. Int Urol Nephrol 2012;44:1389–95. ter trial of the German Testicular Cancer Study Group. J Clin Oncol [86] Besse B, Grunenwald D, Flechon A, et al. Nonseminomatous germ 2007;25:2778–84. cell tumors: assessing the need for postchemotherapy contralat- [95] Oechsle K, Lorch A, Honecker F, et al. Patterns of relapse after eral pulmonary resection in patients with ipsilateral complete chemotherapy in patients with high-risk non-seminomatous necrosis. J Thorac Cardiovasc Surg 2009;137:448–52. germ cell tumor. Oncology 2010;78:47–53. [87] Schirren J, Trainer S, Eberlein M, Lorch A, Beyer J, Bolukbas S. The [96] Lorch A, Kleinhans A, Kramar A, et al. Sequential versus single role of residual tumor resection in the management of nonsemi- high-dose chemotherapy in patients with relapsed or refractory nomatous germ cell cancer of testicular origin. Thorac Cardiovasc germ cell tumors: long-term results of a prospective randomized Surg 2012;60:405–12. trial. J Clin Oncol 2012;30:800–5. [88] Ehrlich Y, Kedar D, Zelikovski A, Konichezky M, Baniel J. Vena caval [97] Oldenburg J, Martin JM, Fossa SD. Late relapses of germ cell reconstruction during postchemotherapy retroperitoneal lymph malignancies: incidence, management, and prognosis. J Clin Oncol node dissection for metastatic germ cell tumor. Urology 2009;73, 2006;24:5503–11. 442.e17–9. [98] Beyer J, Albers P, Altena R, et al. Maintaining success, reducing [89] Beck SD, Foster RS, Bihrle R, Koch MO, Wahle GR, Donohue JP. treatment burden, focusing on survivorship: highlights from the Aortic replacement during post-chemotherapy retroperitoneal third European consensus conference on diagnosis and treatment lymph node dissection. J Urol 2001;165:1517–20. of germ-cell cancer. Ann Oncol 2013;24:878–88. [90] Winter C, Pfister D, Busch J, et al. Residual tumor size and IGCCCG [99] Baniel J, Foster RS, Gonin R, Messemer JE, Donohue JP, Einhorn risk classification predict additional vascular procedures in LH. Late relapse of testicular cancer. J Clin Oncol 1995;13: patients with germ cell tumors and residual tumor resection: a 1170–6. multicenter analysis of the German Testicular Cancer Study [100] George DW, Foster RS, Hromas RA, et al. Update on late relapse of Group. Eur Urol 2012;61:403–9. germ cell tumor: a clinical and molecular analysis. J Clin Oncol [91] Oechsle K, Kollmannsberger C, Honecker F, et al. Long-term sur- 2003;21:113–22. vival after treatment with gemcitabine and oxaliplatin with [101] Oldenburg J, Alfsen GC, Waehre H, Fossa SD. Late recurrences of and without paclitaxel plus secondary surgery in patients with germ cell malignancies: a population-based experience over three cisplatin-refractory and/or multiply relapsed germ cell tumors. decades. Br J Cancer 2006;94:820–7. Eur Urol 2011;60:850–5. [102] Lipphardt ME, Albers P. Late relapse of testicular cancer. World J [92] Nicolai N, Necchi A, Gianni L, et al. Long-term results of a combi- Urol 2004;22:47–54. nation of paclitaxel, cisplatin and gemcitabine for salvage therapy [103] Gels ME, Hoekstra HJ, Sleijfer DT, et al. Detection of recurrence in in male germ-cell tumours. BJU Int 2009;104:340–6. patients with clinical stage I nonseminomatous testicular germ [93] Fizazi K, Gravis G, Flechon A, et al. Combining gemcitabine, cell tumors and consequences for further follow-up: a single- cisplatin, and ifosfamide (GIP) is active in patients with relapsed center 10-year experience. J Clin Oncol 1995;13:1188–94.