Abstract
Innovations in the care of adolescent and young adult (AYA) germ cell tumors (GCT) are needed for one of the most common AYA cancers for which treatment has not significantly changed for several decades. Testicular GCTs (TGCT) are the most common cancer in 15–39-year-old men and ovarian GCTs (OvGCT) are the leading gynecologic malignancy in women under age 25 years. Excellent outcomes, even in widely metastatic disease using cisplatin-based chemotherapy have been achievable since Einhorn and Donohue’s landmark 1977 study in TGCT. However, as the severity of accompanying late effects (ototoxicity, neurotoxicity, cardiovascular disease, second malignant neoplasms, nephrotoxicity, and others) has emerged, efforts to de-intensity treatment and find alternatives to cisplatin have taken on new urgency. Current innovations include the collaborative design of clinical trials that accrue GCT tumors across all ages and both sexes, including adolescents (previously on pediatric trials), and OvGCT (previously on gynecologic-only trials). Joint trials accrue larger sample sizes at a faster rate and therefore evaluate new approaches more rapidly. These joint trials also allow for biospecimen collection to further probe GCT etiology as well as underlying mechanisms of tumor growth, thus providing new therapeutic options. This AYA approach has been fostered by The Malignant Germ Cell International Consortium (MaGIC) which includes over 115 GCT disease experts from pediatric, gynecologic and genitourinary oncology in 16 countries. Trials in development incorporate, for the first time, molecular risk stratification and precision oncology approaches based on specific GCT biology. This collaborative AYA approach pioneered successfully in GCT could serve as a model for impactful research for other AYA cancer types.
INTRODUCTION
King Pyrrhus of Greece triumphed over a Roman army in a key 279 B.C. battle, but most of his forces were destroyed, and he concluded, “Another such victory and we are undone.” Since that time, a “Pyrrhic victory” refers to one that take such a destructive toll on the victor that it is almost equivalent to defeat. Prior to cisplatin’s introduction into germ cell tumor (GCT) management in the 1970s, only approximately 20% of patients with advanced disease achieved durable remissions.1 Cisplatin’s addition to the already established regimen of vinblastine plus bleomycin increased durable remission rates to over 80% as detailed in the seminal 1977 publication by Einhorn and Donohue.2 While cisplatin has since demonstrated activity in many additional tumor types, the extent and duration of response in metastatic GCT remains unparalleled. Nonetheless, as the long-term sequelae of these highly successful treatments have become apparent,3,4 it is their prevalence and severity that has driven many of the changes in management. No new agent or regimen has emerged for these patients since the early 2000s when oxaliplatin was introduced. Thus, in the first- and second-line settings as well as post-high-dose chemotherapy, standard chemotherapy regimens have remained largely unchanged for over 20 years.
Here we provide an overview of GCT epidemiology, genomics, therapeutic approaches, ongoing clinical trials, and survivorship, citing studies representative of recent literature. GCT comprise a paradigm of innovation possible through collaborative adolescent and young adult (AYA) efforts. In fact, since the vast majority of GCTs occur in patients aged 15–39, most of what we know about GCTs and the evidence presented herein relates specifically to AYA populations with few exceptions.
EPIDEMIOLOGY AND GENOMICS
Testicular GCTs.
All GCT originate from primordial germ cells. Testicular GCTs (TGCTs) are the most common cancer in AYA males aged 15–39 year.5,6 U.S. TCGT incidence rates per 100,000 men are highest among non-Hispanic white men (7.0), followed by American Indians/Alaska Natives (4.8), Hispanics (4.2), Asians/Pacific Islanders (2.0), and black men (1.2).7 A growing body of evidence reviewed elsewhere,7 supports the hypothesis that most TGCT are connected to testicular dysgenesis and originate in utero, but that postnatal events determine in part which men will eventually develop TGCT.
The high genetic heritability of TCGT is well-documented,8 with the relative risk of developing TGCT in first-degree relatives of affected men between 6 and 10.9–11 To date, genome-wide association studies (GWAS) have identified 78 independent susceptibility loci for TGCT accounting for 44% of disease heritability.12 These loci are noteworthy for their relatively large per allele effect sizes (odds ratios: 1.4–3.0) and because they are located in genetic regions highly relevant to germ cell development. Associations with some of these variants were recently confirmed in pediatric and adolescent GCTs,13 with some variants showing similar effect sizes in both pre- and post-pubertal GCTs while others were stronger in the post-pubertal age group. Notably, a sequencing study in adult TGCT did not find evidence for rare mutations with large genetic effects.14 Thus, available data support polygenic inheritance with substantial contributions from common variants, with many not yet identified.15
Additional genomic hallmarks of GCT, observed in nearly all post-pubertal tumors, include aneuploidy and gain of chromosome 12p, most commonly as isochromosome (i[12p]).16 Despite knowledge of the ubiquitous 12p gain in GCT for over 3 decades, the specific gene(s) on 12p responsible for tumorigenesis remains unclear. In fact, one hypothesis is that 12p gain represents merely a bystander effect, reflecting dysfunctional meiosis and not a pathogenic role. Nevertheless, given its specificity, 12p gain remains a useful marker for determining whether poorly differentiated malignancies are of GCT origin.17
Ovarian GCTs.
Ovarian GCT (OvGCTs) have an overall incidence rate of 1.2 cases per 100,000 women aged 15–19.18 Incidence peaks in adolescence and young adulthood, and under age 25 years, OvGCTs are the leading gynecologic cancer.19 OvGCT prevalence as a proportion of all ovarian tumors is lower in North America (2%) and Europe (1.3%) vs. Central and South America (3.9%) and Asia (4.2%)20 although this differs between histological subtypes, as reviewed elsewhere.21 OvGCTs originate in utero,22 likely influenced by genetic alterations (e.g., cKIT mutation and amplification in dysgerminoma)23 combined with external factors.22 They are more common in individuals with gonadal dysgenesis with excess risk from 0–60% depending on the syndrome, with prophylactic gonadectomy advised in high-risk individuals, e.g., Turner’s and Swyer Syndromes.24
OvGCTs are classified by morphological appearance and specific immunohistochemical profiles, which are shared with analogous histologies at other anatomic sites. OvGCTs have low mutation rates, very few recurrent somatic mutations and stable copy number profiles23. Ovarian yolk sac tumors, like testicular counterparts, are distinguished by recurrent KRAS mutations and PIK3CA and AKT1 amplification, while KIT mutations are commonly observed in ovarian dysgerminomas and testicular seminomas23. OvGCTs also exhibit chromosome 12p gain, the notable exception being pure mature and immature teratomas (MT, IT).23 Interestingly, frequent loss of heterozygosity is observed in IT and MT25, but not in OvGCTs with mixed histologies that include teratomatous components26. In addition to indicating common clonal origins of IT and MT26, these biological features separate teratomas from other OvGCT histologies and may have relevance for clinical management.
Pediatric GCTs.
Pediatric GCTs represent 3% of childhood cancers before age 14, but incidence rises with puberty onset at ~age 8 in girls and ~age 10 in boys. In adolescence, GCTs are the most common solid tumor, besides thyroid carcinoma, representing 15% of cancer diagnoses. Incidence rates are similar in males and females diagnosed before age 5 years, but are much higher in males in the adolescent age group.27 Incidence rates differ by race and ethnicity in males vs. females in the adolescent age group27 and largely follow trends described above for adult TGCT and OvGCT. Annually in the U.S., approximately 900 GCTs are diagnosed in children (ages 0–19), with 5,700 testicular and 260 ovarian GCTs in young adults (ages 20–39).28
THERAPEUTIC APPROACHES: OVERVIEW
Management:
GCTs are largely managed by pediatric oncologists, gynecologic oncologists or surgeons, or TGCT specialists (oncologists, urologists). Etoposide and cisplatin-based chemotherapy have anchored systemic treatment for over four decades but are associated with significant toxicity.3,29,30 Safe approaches to reduce treatment-related toxicity are a current priority. Systematic investigation, however, has been hampered by challenges in obtaining research funding and creating global clinical trial networks for these cancers, which are rare compared with other adult malignancies. In particular, progress for OvGCT and pediatric GCT has lagged behind TGCT due to smaller populations. Nonetheless, there have been recent advances in OvGCT management, including changes in pathologic classification31 and emerging evidence to support different approaches for these distinct histologic subtypes.32
Cisplatin sensitivity:
A unique, defining GCT characteristic is their exquisite cisplatin sensitivity, allowing cure to be achieved even in widely disseminated disease. Therefore, long sought-after goals have been to understand the etiology of this sensitivity and mechanisms of resistance in the minority who are not cured. While this goal remains elusive, several important clues have emerged over the last three decades. Cisplatin interacts directly with DNA to form intrastrand and interstrand adducts, disrupting DNA structure, resulting in failed recognition by DNA repair proteins, inhibition of transcription and DNA replication, and ultimately apoptosis. Proposed mechanisms of cisplatin sensitivity include low levels of DNA repair proteins, including those involved in nucleoside excision repair,33 defective homologous recombination,34 and primed apoptotic response.35,36 These processes are largely dependent on intact TP53 pathways, which in contrast to most malignancies, are almost always present in GCT. TP53 mutations occur exclusively in cisplatin-resistant GCT and interestingly, are most frequent in primary mediastinal nonseminoma (~70%), which are characterized by poor response rates and unfavorable prognosis.37 Amplification of MDM2, which downregulates TP53, has also been associated with cisplatin resistance in advanced GCT.37
MicroRNAs:
An exciting development in the last decade has been the identification of circulating microRNAs in plasma or serum of testicular GCT patients; the utility in pediatric and OvGCT remains to be defined, and is part of the goal of Children’s Oncology Group (COG) AGCT1531. These small noncoding RNAs inhibit translation by binding to complementary messenger RNA sequences. Among the miRNA 302 and 371–373 clusters identified specific to GCT,38 miRNA 371a-3p has emerged as the most promising new GCT marker. Among 522 TGCT patients, the sensitivity, specificity, positive predictive value, and negative predictive value of serum miRNA 371a-3p for GCT diagnosis were 92%, 96%, 97%, and 83%, far exceeding AFP, HCG, and LDH individually and combined.39 Numerous potential applications of this novel biomarker exist include use as a diagnostic test, a prognostic factor, a means to assess treatment response, and detection of minimal residual disease, and as part of surveillance to potentially reduce the frequency of currently recommended axial imaging.
Unified approach:
What has become increasingly clear is the need for a unified approach to manage GCT. The future holds promise for the ability to molecularly stratify patients and go beyond clinical characteristics, histopathology, and serum tumor markers. Trials in development focus on biospecimen collection and incorporation of genomic alterations into risk stratification for treatment; other adaptations have incorporated all patients with post-pubertal GCT, including adolescents (traditionally treated on pediatric trials), and women with GCT (typically treated on gyn-oncology only trials), justified by cross-comparisons showing that GCT outcomes are similar when comparing by age/stage/risk group, independent of sex and primary tumor site. Moreover, this joint approach has facilitated more rapid accrual40, allowing contemporary trials to be designed with larger sample sizes, thus providing more power to detect treatment differences. This contrasts to several important TGCT trials which closed prematurely due to poor accrual.41 A unified approach also allows for biospecimen collection to continue to delve into etiology and molecularly-based treatment options; for example, 3 cross-age, cross-sex National Clinical Trials Network (NCTN) trials are either underway or recently completed (NCT03067181, NCT02582697, NCT02375204), designed by the Malignant Germ Cell International Consortium (MaGIC) that together will accrue nearly 3,000 patients. Two new concepts under review at NCTN have been co-designed and co-sponsored by pediatric, gynecologic, and genitourinary oncology.
TREATMENT
First-line:
Standard of care, first-line chemotherapy management for TGCT and post-pubertal extragonadal GCT is determined by the International Germ Cell Cancer Collaborative Group (IGCCCG) prognostic model, which divides patients into good-, intermediate- and poor-prognosis with cure rates of approximately 90%, 80%, and 55%, respectively.42,43 For good-risk patients, three cycles of bleomycin, etoposide, and cisplatin (BEP×3) or four cycles of etoposide and cisplatin (EP×4) is standard whereas for intermediate- and poor-risk patients, BEPx4 or four cycles of etoposide, ifosfamide, and cisplatin (VIP×4) (for those unable to receive bleomycin) are recommended. Efforts to decrease treatment intensity and toxicity for patients with good-risk disease, including etoposide dose reductions and substitution of carboplatin for cisplatin, led to decremental progression-free survival and in some series, overall survival.44–46 Similarly, attempts to escalate treatment intensity to improve efficacy in intermediate- and poor-risk patients failed to improve upon BEP results, but led to excess toxicity.47–49 Although risk stratification schemes have been devised for adult OvGCTs,50,51 these have not been adopted in clinical practice, and NCCN and ESMO guidelines both recommend BEP for all stages of non-dysgerminoma, regardless of histology and without specifying the optimal number of cycles.
In contrast to men with TGCT, in the UK, children and adolescents with GCT had excellent outcomes with first-line carboplatin-based regimens, notably delivered at higher dose and dose-intensity than in the TGCT trial. This observation led to agreements between COG in the U.S., Canada, and Australia (where BEP was standard) and the Children’s Cancer and Leukemia Group, UK (where carboplatin-based treatment was standard) to combine past clinical data to compare outcomes between cisplatin- and carboplatin-based regimens. This collaboration started the MaGIC consortium which now includes over 115 GCT experts in 16 countries representing numerous disciplines comprising clinical care and research (Figure 1). MaGIC demonstrated no significant difference in outcome by age or risk group in children and adolescents administered carboplatin vs. cisplatin.52 This equipoise justified the current randomized clinical trial (RCT), AGCT1531 (NCT03067181), comparing BEP to BEC (bleomycin, etoposide carboplatin) in both pre-pubertal children and AYAs up to age 25 years.
Figure 1.
The Malignant Germ Cell International Consortium, June 2023
Similarly, MaGIC showed that adolescent males over age 11 years with TGCT had outcomes similar to those expected in adult TCGT using the IGCCCG, and that adolescent females with FIGO Stage IV OvGCT had outcomes equivalent to men with IGCCCG poor-risk tumors.53 These observations rationalized the inclusion of adolescent males and females onto a clinical trial for IGCCCG intermediate- and poor-risk patients developed by the Australian New Zealand Urogenital and Prostate Clinical Trials Group (ANZUP) comparing standard BEP given every 3 weeks to compressed BEP given every 2 weeks (P3BEP, AGCT1532, NCT02582697). This study had originally been limited only to adult men with TGCT. Notably, AYA accrual from pediatric clinical trial organizations account for 30% of total enrollments to date, showing that cooperation between oncologic sub-specialties can enhance accrual.
Second- and later-line chemotherapy:
For patients progressing after first-line chemotherapy, second-line chemotherapy consists of either conventional-dose chemotherapy (CDCT) regimens such as paclitaxel, ifosfamide, and cisplatin (TIP) or vinblastine, ifosfamide, and cisplatin (VeIP), or alternatively high-dose chemotherapy with autologous stem cell transplant (HDCT/ASCT).54–57 Due to a lack of randomized trials, no optimal CDCT or HDCT/ASCT regimen has been determined. In addition, whether HDCT or CDCT is the preferred second-line strategy remains controversial with retrospective studies consistently favoring HDCT/ASCT,58,59 but the one RCT demonstrating no benefit to a single HDCT cycle over initial salvage CDCT.60 International collaboration facilitated the design and successful accrual completion of the phase III TIGER trial (A031102, E1407, NCT02375204) to definitively answer this question. Patients were randomized to receive either TIPx4 or HDCT/ASCT with the TI-CE regimen (paclitaxel plus ifosfamide followed by high-dose carboplatin and etoposide), with the primary endpoint of overall survival. Based on input from MaGIC and COG, the lower study age limit was reduced from 16 to 14 years; it opened in 13 countries across 3 continents without pharmaceutical company support, sponsored by the NCTN’s adult and pediatric cooperative groups, European Organisation for Research and Treatment of Cancer (EORTC), ANZUP, and the philanthropic organization, Movember.
Patients progressing after HDCT/ASCT are generally considered incurable with most dying from their disease. GCT death typically occurs in the 20s and 30s, and as such, accounts for the greatest average number of life-years’ lost of any non-childhood malignancy.61,62 Options in this setting are inadequate, but include gemcitabine plus oxaliplatin, gemcitabine plus paclitaxel, and oral etoposide.63–66 No new agent or regimen has emerged for these patients since the early 2000s when oxaliplatin was introduced. In summary, in the first- and second-line settings as well as post-HDCT, standard chemotherapy regimens have remained unchanged for over 20 years.
Currently, two trials in development bring molecular risk stratification into GCT treatment for the first time. A trial under Eastern Cooperative Oncology Group (ECOG) review proposes to use post-orchiectomy serum miRNA 371a-p to counsel Stage I patients regarding retroperitoneal lymph node dissection, based on a retrospective study in which miR 371a-3p was 100% sensitive and 90% specific in predicting which patients had viable GCT in resected nodes.67 A second trial under review by Alliance for men and women with IGCCCG intermediate- or poor-risk disease will use identification of TP53 pathway alterations37 as well as slower than expected serum tumor marker decline after 1 chemotherapy cycle68 to select patients for intensified therapy.
In the relapsed/refractory setting (progression post-HDCT), MaGIC has been fostering basic and translational science collaborations needed to develop novel rational agents and simultaneously working to develop a clinical trials consortium to facilitate more rapid drug study. Promising agents under development include demethylating agents, WNT inhibition, CAR-T strategies targeting claudin 6, and therapies targeting glypican-3.
SURVIVORSHIP
As reviewed by Fung,69 ototoxicity, neurotoxicity, cardiovascular disease (CVD), second malignant neoplasms (SMN), pulmonary toxicity, nephrotoxicity, infertility, hypogonadism, depression, anxiety, and cognitive impairment have emerged as important survivorship concerns for GCT survivors affecting quality of life and functional status, with the majority of data generated in AYA TGCT survivors. Long-term platinum retention may contribute in part to several long-term toxicities.70 Key research findings in recent publications from the three European cohorts of testicular cancer survivor cohorts are summarized in Table 1 and briefly described below, along with major results from the multi-institutional U.S.-based TGCT survivor study (the “Platinum Study”). The latter clinical cohort includes patients from 8 major cancer centers, including Indiana University and Memorial Sloan Kettering Cancer Center,3,4 and importantly, a small subset is non-white. The type, number, and prevalence of a wide range of adverse health outcomes (AHOs) in 952 adult TGCT survivors given cisplatin-based chemotherapy in this cohort were described in detail.3 Over one-third of TGCT survivors (median age: 37) reported ≥3 AHOs with 12.5% reporting ≥5 AHOs. The most common AHOs were tinnitus (37.1%), hearing loss (HL) (31.5%), obesity (30.9%), and peripheral neuropathy (27.0%). After EPX4 or BEPX3, types and prevalence of individual AHOs were similar (P >0.05), except for Raynaud phenomenon (11.6% v 21.4%; P <0 .01), peripheral neuropathy (29.2% v 21.4%; P =0.02), and obesity (25.5% v 33.0%; P = 0.04). Risk factors predicting higher numbers of AHOs/patient included larger cumulative bleomycin doses, increasing age, current or former smoking, lower physical activity levels, and less than college education. Self-reported health was excellent/very good in approximately 60% of TGCT survivors, but declined as AHOs increased (P <0.001). Similarly, chemotherapy with BEP and more than one line of chemotherapy (OR 2.4 to 4.7 depending on treatment intensity, P<0.001); current smoking status (OR=1.7), and Charlson comorbidity (OR=2.1) were associated with significantly increased neurotoxicity in the Danish Testicular Cancer Late Effect Treatment Cohort.71 Within the Dutch Testicular Cancer Survivor case-cohort study, cisplatin-based chemotherapy (HR=1.9); obesity (HR=4.6) tobacco use (HR=1.7); Raynaud’s phenomenon (HR=1.9); dyslipidemia (HR=2.8); and positive CVD family history (HR=2.9) were associated with significantly higher CVD risks.72
Table 1:
European Studies of Testicular Cancer Survivors (TCS)
| Study Cohort | Study Methods | Treatment Dates | Number of Survivors | Age at TC diagnosis (years) | Outcome(s) in Recent Publication | Overview: All Cohort Outcomes |
|---|---|---|---|---|---|---|
| National Norwegian Testicular Cancer Survivor Study73,86 | Longitudinal study: Identified by Norwegian Cancer Registry for survivors treated for unilateral TC at 4 Norwegian university hospitals. In 1998, eligible TCS were invited by mail to complete a survey and then invited to undergo physical examination including blood tests. Subsequently, 2 more surveys administered: 2007–2009 and 2015–2016. | 1980–1994 | First survey: 1,813 invited, 1,436 responded (79%) Second survey: 1,371 invited, 1,050 responded (77%) Third survey: 963 invited, 783 responded (81%) |
Responders for first survey: Median: 33 SD: ± 8.9 |
Cumulative overall 20-year mortality: 14% (95% CI 11.8 to 16.8) compared with expected rate: 11%. Significantly increased overall mortality was related to increasing age (HR=1.08), cisplatin-based chemotherapy with cumulative dose >630 mg (HR=1.97), major comorbidity (HR=1.93), lower socioeconomic status (HR=1.97), unhealthy lifestyle (HR=1.65), and depressive disorder (HR=1.86). Cancer-related mortality. Higher cancer-related mortality was associated with cisplatin-based chemotherapy with cumulative dose >630 mg (HR=3.65) and depressive disorder (HR=2.21). |
|
| The Danish Testicular Cancer Late Effect Treatment Cohort (DeTeCa Late)71,87 | Cross-sectional study: Identified by applying these inclusion criteria to DeTeCa database: Germ cell cancer diagnosis, Danish citizenship, follow- up and medical treatment at oncology ward in Denmark. Postal invitation sent to all eligible TCS: November 2014. Clinical data collected using hospital files, pathology reports. | 1984–2007 | 4,271 TCS invited to participate, 2,572 responded (60%). | Mean: 53 Range: 25–95 |
Neurotoxicity: Significantly increased neurotoxicity was associated with chemotherapy with BEP and more than one line of chemotherapy (OR 2.4 to 4.7 depending on treatment intensity, P < 0.001); current smoking (OR 1.7), and Charlson comorbidity score (OR 2.1). Radiotherapy and surveillance were not associated with neurotoxicity. After receiving BEP, neurotoxicity was highly associated with all indicators of worse quality of life after approximately 20 years of follow-up (P-trend:1.5 × 10−17 to 1.1 × 10−28) |
|
| Dutch Testicular Cancer Survivor Study72 | Case-cohort study: Five Dutch TC treatment centers provided access to medical records of TC patients. Those developing CVD after TC diagnosis were identified via regular follow-up and compared with 15% of base cohort in 3 of 5 treatment centers and 25% of base cohort in remaining 2 centers. Treatment data abstracted from medical records; all surviving patients in case-cohort study invited to complete risk factor survey: 2015–2017. | 1976–2007 | Entire case-cohort:Cases with CVD: 272 Base cohort: 925 Case-cohort with completed risk factor survey:Cases with CVD: 120 Base cohort: 447 |
For entire 4,748 patients: Mean: Not provided Range: 12–50 |
Cardiovascular disease risks: Significantly higher CVD risks were associated with cisplatin combination chemotherapy (HR=1.9); obesity at TC diagnosis (HR=4.6); smoking at TC diagnosis (HR=1.7); development of Raynaud’s phenomenon (HR=1.9); dyslipidemia (HR=2.8); and positive family history for CVD (HR=2.9). Quality of life: TC survivors with CVD reported inferior quality of life on physical domains vs. survivors not developing CVD. Cardiovascular disease risk factors: Among 304 testicular cancer survivors with CVD (median age at clinical evaluation=51), 86% had dyslipidemia, 50% had hypertension, and 35% had metabolic syndrome, irrespective of treatment. |
|
Abbreviations: BEP, bleomycin, etoposide, cisplatin; CVD, cardiovascular disease; HR, hazard ratio; OR odds ratio; TC, testicular cancer; TCS, testicular cancer survivors.
Refer to Table 2 in original manuscript by Kreiberg et al.87 for comprehensive outline of outcomes studied. These span physical health (e.g., neurotoxicity, fatigue, infertility before and after testicular cancer diagnosis, symptoms of testosterone deficiency and erectile dysfunction) and psychosocial health (e.g. psychological distress; anxiety; alcohol and tobacco habits; substance abuse history).
Both the frequency and severity of a wide spectrum of AHOs were combined into one overall cumulative burden of morbidity (CBM) score after 1,214 TGCT survivors were accrued to The Platinum Study.4 About one in five patients had a high (15%) or very high/severe (4.1%) CBM score (Figure 2). Significant risks factors for higher CBM scores included VIPX4 (OR=2.0 vs BEPX3) or BEPX4 (OR=1.4 vs BEPX3), older attained age (OR=1.2 per 5 years), current disability leave (OR=3.5), less-than-college education (OR=1.4), and current or former smoking status (OR=1.3). Asian race (OR=0.41) and vigorous physical activity (OR=0.68) were inversely associated with higher CBM scores (Table 2). Worse CBM scores may translate to worse overall mortality. In the Norwegian TGCT study, patients had worse cumulative overall 20-year mortality of 14% (95% CI: 11.8 to 16.8) vs. an expected rate of 11%.73 Major comorbidity (HR=1.93), increasing age (HR=1.08), cisplatin-based chemotherapy with cumulative dose >630 mg (HR=1.97), lower socioeconomic status (HR=1.97), unhealthy lifestyle (HR=1.65), and depressive disorder (HR=1.86) were associated with significantly increased overall mortality.
Figure 2.
Distribution of Cumulative Burden of Morbidity (CBM) Score Among 1,214 Participants in the Platinum Study. The above Figure is reproduced from Kerns et al.4 with permission granted by Wolters Kluwer Health, Inc. on May 1, 2023.
Table 2.
Multivariable Ordinal Logistic Regressiona of Factors Associated With CBM Score
| Odds Ratio (95% CI) | p-value | |
|---|---|---|
| Age at evaluation (per 5 years)b | 1.18 (1.10, 1.26) | <0.001 |
|
Time since chemotherapy completion, years < 2 2–5 6–9 10+ |
Ref. 0.91 (0.68, 1.23) 0.61 (0.42, 0.89) 0.55 (0.38, 0.85) |
- 0.54 0.010 0.002 |
|
Race White Black/African-American Asian Other |
Ref. 1.56 (0.49, 5.03) 0.41 (0.23, 0.72) 1.05 (0.63, 1.76) |
- 0.45 0.002 0.84 |
|
Education College or post-college graduate Less than college education |
Ref. 1.44 (1.11, 1.87) |
- 0.006 |
|
Current employment status Employed Unemployed Retired On disability leave |
Ref. 0.90 (0.55, 1.47) 1.10 (0.36, 3.39) 3.53 (1.57, 7.95) |
- 0.66 0.87 0.002 |
|
Smoking status Never Current or Former |
Ref. 1.28 (1.02, 1.63) |
- 0.037 |
|
Vigorous physical activity (≥6 METs)c No Yes |
Ref. 0.68 (0.52, 0.89) |
- 0.004 |
|
RPLNDd No Yes |
Ref. 0.88 (0.69, 1.12) |
- 0.31 |
|
Type of chemotherapye BEPX3 EPX4 BEPX4 VIPX4 |
Ref. 1.09 (0.75, 1.60) 1.44 (1.04, 1.98) 1.96 (1.04, 3.71) |
- 0.65 0.028 0.039 |
Abbreviations: BEP, bleomycin, etoposide, cisplatin; EP, etoposide, cisplatin; MET, metabolic equivalent task; RPLND, retroperitoneal lymph node dissection; VIP, etoposide, ifosfamide, cisplatin
Odds ratios and p-values are from an adjusted model that includes all other variables listed in the table, as well as enrollment center, with CBM score as the outcome (dependent) variable. The ‘very high’ and ‘severe’ categories were collapsed due to sparse data. Analysis includes 1,013 (83.4%) testicular cancer survivors with non-missing data for all variables in the model.
Age at diagnosis was not included in the model given the strong correlation with age at evaluation (r=0.81), which was included
See Table 1, footnote o and Appendix-Supplemental Methods for details on assessment of physical activity.
RPLND was retained in the multivariable model to control for potential residual confounding, given its correlation with chemotherapy regimen (P < 0.001); approximately 34%, 55%, 66%, and 44% of TCS treated with BEPX3, EPX4, BEPX4, and VIPX4, respectively, had an RPLND.
Disease stage was not significantly associated with CBM score (P = 0.48), suggesting that increased scores after BEPX4 or VIPX4 were not explained by more advanced tumor status
The above table is reproduced from Kerns et al.4 with permission granted by Wolters Kluwer Health, Inc. on May 1, 2023.
Cisplatin-related toxicities among TGCT survivors can also negatively impact functional status. In one recent report,74 243 TGCT survivors were evaluated for functional impairment due to cisplatin-related ototoxicity. Approximately one-third (35.8%) of those with HL experienced clinically significant functional impairment. Severity of both HL and tinnitus were significantly associated with worsening scores on the Hearing Handicap for Adults (HHIA) and Tinnitus Primary Function Questionnaire (TPFQ), respectively (P<.0001 each). Survivors with either greater HL or more severe tinnitus were also significantly more likely to report cognitive dysfunction (OR=5.52 and 2.56), fatigue (OR=6.18 and 4.04), depression (OR=3.93 and 3.83), and lower overall health (OR=0.39 and 0.46). These results prompted a clinical recommendation that the HHIA and TPFQ be used to risk-stratify TGCT survivors at clinical follow-up who could benefit from referrals to audiologists for evaluation and management.
In order to elucidate etiopathogenetic mechanisms of AHOs among TGCT survivors, prior investigations have identified significant associations of germline genetic mutations with ototoxicity, peripheral neuropathy, and bleomycin-induced pulmonary toxicity, as reviewed elsewhere.69 Single nucleotide polymorphisms (SNPs) in megalin (rs2075252), COMT (rs9332377), TPMT (rs12201199), ACYP2 (rs1872328), WFS1 (rs62283056), and OTOS (rs7606353) are significantly associated with platinum-related ototoxicity. Lower expression levels of RPRD1B and SNPs of both glutathione S-transferase (GST) P1 and RPRD1B are associated with cisplatin-induced peripheral neuropathy. The H63D variant of HFE is significantly associated with higher frequencies of bleomycin-induced pulmonary toxicity vs. wild-type HFE.
Late effects of treatment among children and adolescents with GCT are largely unexplored since they were not previously included in large cohort studies of childhood cancer survivors.75 Nonetheless, GCT survivors comprise the 3rd largest group of U.S. childhood cancer survivors due to the high survival rate, currently numbering close to 40,000 in the U.S. alone.76 Because primary treatment regimens for pediatric GCT include the same agents used for adult TGCT (cisplatin, etoposide, and bleomycin), similar late effects are likely to occur. However, because risk may differ with either exposures at younger ages or by sex, efforts are needed to characterize these toxicities.77,78 A cohort exclusively composed of pediatric and adolescent GCT survivors (the GCT Outcomes and Late effects Data (GOLD) Study)79 was recently established using COG registry protocols to identify/recruit patients.80 Initial efforts will focus on the most prevalent short-term toxicities, i.e., ototoxicity and neuropathy. Preliminary findings (Table 3) suggest that tinnitus (14%), hearing loss (20%), and neuropathy (9%) occurs less frequently in children vs. adult TGCT survivors, but are still noteworthy. The GOLD study will also permit future investigations of other potential late effects in pediatric GCT survivors with delayed onset, including infertility, CVD, and SMN.
Table 3.
Selected data from the GOLD Study (N=325)
| N (%) | |
|---|---|
| Male sex | 165 (51) |
| Median age at questionnaire completion | 21yrs (range 5–30) |
| Median follow-up time | 8yrs (range 4–14) |
| Tumor Location | |
| Testis | 55 (17) |
| Ovary | 86 (26) |
| Intracranial | 122 (38) |
| Extragonadal | 62 (19) |
| Treatment | |
| Chemotherapy +/− other treatments | 244 (75) |
| No chemotherapy | 81 (25) |
| Hearing lossa | 59 (24) |
| Tinnitusa | 34 (14) |
| Neuropathya | 22 (9) |
Restricted to 244 individuals who reported chemotherapy
CONCLUSIONS AND FUTURE DIRECTIONS
In a 2007 Journal of Clinical Oncology editorial entitled “The Graying of Testis Cancer Patients: What Have We Learned?”, Bajorin81 noted how the TGCT model established far more than curative chemotherapy combinations in the 1970’s and 1980s. Indeed “the concept of modeling response to chemotherapy using serum tumor markers emerged and risk-directed treatment strategies and risk-directed clinical trials soon followed, particularly to reduce toxicity in highly curable patients.” Bajorin stated that TGCT patients were “directly responsible for these great advances by having volunteered for clinical trials in such high numbers that important clinical advances were made at an unparalleled pace for a rare adult disease, and they helped establish randomized clinical trials as the gold standard in oncology research.” This monumental contribution of these early TGCT treatments to the field of oncology and, importantly, the sacrifices and dedication of the involved patients, should never be forgotten.
Similarly, in 2023, there is another tremendous opportunity for patients and researchers to impact GCT management and outcomes. First, the collaborative, inclusive approach exemplified by MaGIC allows advances to benefit all GCTs in all patient populations, spanning pediatric and AYA age groups, and including men and women. This unified approach also facilitates biospecimen collection allowing deeper interrogation of GCT etiology, biology and mechanisms of toxicity and treatment resistance, allowing for improved prognostication, risk stratification and development of novel treatments. Similar to MaGIC, the Global Germ Cell Cancer Group has fostered international collaborations, playing an integral role in development of the TIGER trial, updating the IGCCCG risk models, and conducting retrospective analyses on important clinical situations, e.g., patients with bone or brain metastases and those relapsing after carboplatin for stage I seminoma or BEP for stage I nonseminoma.42,43,82–85 Survivorship research should be built alongside these efforts. Importantly, most GCT treatments are homogeneous and allow in-depth analysis of platinating agent toxicities. Subsequently, etiopathogenetic pathways can be elucidated, which are needed to formulate risk-based, targeted prevention and intervention strategies. Optimally, patients should be administered validated baseline questionnaires at clinical trial enrollment with later surveys addressing potential AHOs, comorbidities, and lifestyle behaviors during life-long follow-up. Interactions between aging and the onset and progression of AHOs after GCT treatment should also be explored, given long patient life spans. GCT populations also provide a prime opportunity for novel interventional strategies, including incorporation of emerging technologies such as radiomics and artificial intelligence. As Bajorin81 stated in 2007, beyond its established role as a model for a curable neoplasm, TGCT also confers a unique opportunity for conducting collaborative survivorship studies with potential to impact all cancer survivors. Similarly, the synergistic efforts currently being pioneered for all AYA GCT could serve as a model for research in other malignancies. A future in which our patients’ cancer victories are no longer “Pyrrhic” may finally be within reach.
Acknowledgments of research support:
Dr. Travis was supported by 2 R01 CA157823 funded by the National Cancer Institute (NCI). Dr. Feldman was supported by National Institutes of Health (NIH) P30 Cancer Center Support Grant No. P30 CA008748. Dr. Poynter was supported by W81XWH2210184 funded by the Department of Defense and the Children’s Cancer Research Fund. Frazier: Dr. Frazier was supported by UH3CA240688 funded by the NCI, CA220465 funded by the Department of Defense, and the St. Baldrick’s and Rally Foundations.
Footnotes
Previous presentations of data: not applicable
Disclaimers: None
Conflicts of Interest:
Travis: None
Fung: on ASCO website
Lockley: None
Feldman: on ASCO website
Poynter: None
Frazier: on ASCO website (consultant for Decibel Therapeutics)
REFERENCES
- 1.Samuels ML, Johnson DE, Holoye PY: Continuous intravenous bleomycin (NSC-125066) therapy with vinblastine (NSC-49842) in stage III testicular neoplasia. Cancer Chemother Rep 59:563–70, 1975 [PubMed] [Google Scholar]
- 2.Einhorn LH, Donohue J: Cis-diamminedichloroplatinum, vinblastine, and bleomycin combination chemotherapy in disseminated testicular cancer. Ann Intern Med 87:293–8, 1977 [DOI] [PubMed] [Google Scholar]
- 3.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. Journal of Clinical Oncology 35:1211–1222, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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 36:1505–1512, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.SEER*Explorer: An interactive website for SEER cancer statistics, Surveillance Research Program, National Cancer Institute
- 6.Coccia PF: Overview of Adolescent and Young Adult Oncology. J Oncol Pract 15:235–237, 2019 [DOI] [PubMed] [Google Scholar]
- 7.McGlynn K, Rajpert-De Meyts E, Stang A: Testis Cancer, in Thun M, Linet M, Cerhan J, et al. (eds): Cancer Epidemiology and Prevention 4th Edition (ed 4th). New York, NY, Oxford University Press, 2017 [Google Scholar]
- 8.Czene K, Lichtenstein P, Hemminki K: Environmental and heritable causes of cancer among 9.6 million individuals in the Swedish Family-Cancer Database. Int J Cancer 99:260–6, 2002 [DOI] [PubMed] [Google Scholar]
- 9.Forman D, Oliver RTD, Brett AR, et al. : Familial testicular cancer: a report of the UK family register, estimation of risk and an HLA class 1 sib-pair analysis. British Journal of Cancer 65:255–262, 1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Heimdal K, Olsson H, Tretli S, et al. : Familial testicular cancer in Norway and southern Sweden. British Journal of Cancer 73:964–969, 1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Westergaard T, Olsen JH, Frisch M, et al. : Cancer risk in fathers and brothers of testicular cancer patients in Denmark. A population-based study. Int J Cancer 66:627–31, 1996 [DOI] [PubMed] [Google Scholar]
- 12.Pluta J, Pyle LC, Nead KT, et al. : Identification of 22 susceptibility loci associated with testicular germ cell tumors. Nat Commun 12:4487, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Marcotte EL, Pankratz N, Amatruda JF, et al. : Variants in BAK1, SPRY4, and GAB2 are associated with pediatric germ cell tumors: A report from the children’s oncology group. Genes Chromosomes Cancer 56:548–558, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Litchfield K, Loveday C, Levy M, et al. : Large-scale Sequencing of Testicular Germ Cell Tumour (TGCT) Cases Excludes Major TGCT Predisposition Gene. Eur Urol, 2018 [DOI] [PubMed] [Google Scholar]
- 15.Litchfield K, Thomsen H, Mitchell JS, et al. : Quantifying the heritability of testicular germ cell tumour using both population-based and genomic approaches. Sci Rep 5:13889, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sandberg AA, Meloni AM, Suijkerbuijk RF: Reviews of chromosome studies in urological tumors. III. Cytogenetics and genes in testicular tumors. J Urol 155:1531–56, 1996 [PubMed] [Google Scholar]
- 17.Motzer RJ, Rodriguez E, Reuter VE, et al. : Molecular and cytogenetic studies in the diagnosis of patients with poorly differentiated carcinomas of unknown primary site. J Clin Oncol 13:274–82, 1995 [DOI] [PubMed] [Google Scholar]
- 18.Quirk JT, Natarajan N, Mettlin CJ: Age-specific ovarian cancer incidence rate patterns in the United States. Gynecol Oncol 99:248–50, 2005 [DOI] [PubMed] [Google Scholar]
- 19.Lockley M, Stoneham SJ, Olson TA: Ovarian cancer in adolescents and young adults. Pediatr Blood Cancer 66:e27512, 2019 [DOI] [PubMed] [Google Scholar]
- 20.Matz M, Coleman MP, Sant M, et al. : The histology of ovarian cancer: worldwide distribution and implications for international survival comparisons (CONCORD-2). Gynecol Oncol 144:405–413, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Smith HO, Berwick M, Verschraegen CF, et al. : Incidence and survival rates for female malignant germ cell tumors. Obstet Gynecol 107:1075–85, 2006 [DOI] [PubMed] [Google Scholar]
- 22.Kraggerud SM, Hoei-Hansen CE, Alagaratnam S, et al. : Molecular characteristics of malignant ovarian germ cell tumors and comparison with testicular counterparts: implications for pathogenesis. Endocr Rev 34:339–76, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Van Nieuwenhuysen E, Busschaert P, Neven P, et al. : The genetic landscape of 87 ovarian germ cell tumors. Gynecol Oncol 151:61–68, 2018 [DOI] [PubMed] [Google Scholar]
- 24.Cools M, Looijenga LH, Wolffenbuttel KP, et al. : Disorders of sex development: update on the genetic background, terminology and risk for the development of germ cell tumors. World J Pediatr 5:93–102, 2009 [DOI] [PubMed] [Google Scholar]
- 25.Heskett MB, Sanborn JZ, Boniface C, et al. : Multiregion exome sequencing of ovarian immature teratomas reveals 2N near-diploid genomes, paucity of somatic mutations, and extensive allelic imbalances shared across mature, immature, and disseminated components. Mod Pathol 33:1193–1206, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Snir OL, DeJoseph M, Wong S, et al. : Frequent homozygosity in both mature and immature ovarian teratomas: a shared genetic basis of tumorigenesis. Mod Pathol 30:1467–1475, 2017 [DOI] [PubMed] [Google Scholar]
- 27.Poynter JN, Amatruda JF, Ross JA: Trends in incidence and survival of pediatric and adolescent patients with germ cell tumors in the United States, 1975 to 2006. Cancer 116:4882–91, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.United States Cancer Statistics - Mortality Data: 1999–2019, WONDER Online Database, United States Department of Health and Human Services Centers for Disease Control and Prevention, 2022. https://wonder.cdc.gov/cancer.HTML
- 29.Howard R, Gilbert E, Lynch CF, et al. : Risk of leukemia among survivors of testicular cancer: a population-based study of 42,722 patients. Ann Epidemiol 18:416–21, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Watson RA, De La Pena H, Tsakok MT, et al. : Development of a best-practice clinical guideline for the use of bleomycin in the treatment of germ cell tumours in the UK. Br J Cancer 119:1044–1051, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lokuhetty D WV, Cree IA, editors. : World Health organization classification (WHO) of tumours: female genital tumours. (ed 5th ). Lyon, IARC Press;, 2020 [Google Scholar]
- 32.Newton C, Murali K, Ahmad A, et al. : A multicentre retrospective cohort study of ovarian germ cell tumours: Evidence for chemotherapy de-escalation and alignment of paediatric and adult practice. Eur J Cancer 113:19–27, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Welsh C, Day R, McGurk C, et al. : Reduced levels of XPA, ERCC1 and XPF DNA repair proteins in testis tumor cell lines. Int J Cancer 110:352–61, 2004 [DOI] [PubMed] [Google Scholar]
- 34.Cavallo F, Graziani G, Antinozzi C, et al. : Reduced proficiency in homologous recombination underlies the high sensitivity of embryonal carcinoma testicular germ cell tumors to Cisplatin and poly (adp-ribose) polymerase inhibition. PLoS One 7:e51563, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gutekunst M, Oren M, Weilbacher A, et al. : p53 hypersensitivity is the predominant mechanism of the unique responsiveness of testicular germ cell tumor (TGCT) cells to cisplatin. PLoS One 6:e19198, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Taylor-Weiner A, Zack T, O’Donnell E, et al. : Genomic evolution and chemoresistance in germ-cell tumours. Nature 540:114–118, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bagrodia A, Lee BH, Lee W, et al. : Genetic Determinants of Cisplatin Resistance in Patients With Advanced Germ Cell Tumors. J Clin Oncol 34:4000–4007, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Murray MJ, Halsall DJ, Hook CE, et al. : Identification of microRNAs From the miR-371~373 and miR-302 clusters as potential serum biomarkers of malignant germ cell tumors. Am J Clin Pathol 135:119–25, 2011 [DOI] [PubMed] [Google Scholar]
- 39.Dieckmann KP, Radtke A, Geczi L, et al. : Serum Levels of MicroRNA-371a-3p (M371 Test) as a New Biomarker of Testicular Germ Cell Tumors: Results of a Prospective Multicentric Study. J Clin Oncol 37:1412–1423, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Williams SD, Kauderer J, Burnett AF, et al. : Adjuvant therapy of completely resected dysgerminoma with carboplatin and etoposide: a trial of the Gynecologic Oncology Group. Gynecol Oncol 95:496–9, 2004 [DOI] [PubMed] [Google Scholar]
- 41.de Wit R, Skoneczna I, Daugaard G, et al. : Randomized phase III study comparing paclitaxel-bleomycin, etoposide, and cisplatin (BEP) to standard BEP in intermediate-prognosis germ-cell cancer: intergroup study EORTC 30983. J Clin Oncol 30:792–9, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Beyer J, Collette L, Sauvé N, et al. : Survival and New Prognosticators in Metastatic Seminoma: Results From the IGCCCG-Update Consortium. J Clin Oncol 39:1553–1562, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gillessen S, Sauvé N, Collette L, et al. : Predicting Outcomes in Men With Metastatic Nonseminomatous Germ Cell Tumors (NSGCT): Results From the IGCCCG Update Consortium. Journal of Clinical Oncology 39:1563–1574, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Grimison PS, Stockler MR, Thomson DB, et al. : Comparison of two standard chemotherapy regimens for good-prognosis germ cell tumors: updated analysis of a randomized trial. J Natl Cancer Inst 102:1253–62, 2010 [DOI] [PubMed] [Google Scholar]
- 45.Bajorin DF, Sarosdy MF, Pfister DG, et al. : Randomized trial of etoposide and cisplatin versus etoposide and carboplatin in patients with good-risk germ cell tumors: a multiinstitutional study. J Clin Oncol 11:598–606, 1993 [DOI] [PubMed] [Google Scholar]
- 46.Horwich A, Sleijfer DT, Fosså SD, et al. : Randomized trial of bleomycin, etoposide, and cisplatin compared with bleomycin, etoposide, and carboplatin in good-prognosis metastatic nonseminomatous germ cell cancer: a Multiinstitutional Medical Research Council/European Organization for Research and Treatment of Cancer Trial. J Clin Oncol 15:1844–52, 1997 [DOI] [PubMed] [Google Scholar]
- 47.Motzer RJ, Nichols CJ, Margolin KA, et al. : Phase III randomized trial of conventional-dose chemotherapy with or without high-dose chemotherapy and autologous hematopoietic stem-cell rescue as first-line treatment for patients with poor-prognosis metastatic germ cell tumors. J Clin Oncol 25:247–56, 2007 [DOI] [PubMed] [Google Scholar]
- 48.Nichols CR, Williams SD, Loehrer PJ, et al. : Randomized study of cisplatin dose intensity in poor-risk germ cell tumors: a Southeastern Cancer Study Group and Southwest Oncology Group protocol. J Clin Oncol 9:1163–72, 1991 [DOI] [PubMed] [Google Scholar]
- 49.de Wit R, Skoneczna I, Daugaard G, et al. : Randomized Phase III Study Comparing Paclitaxel-Bleomycin, Etoposide, and Cisplatin (BEP) to Standard BEP in Intermediate-Prognosis Germ-Cell Cancer: Intergroup Study EORTC 30983. J Clin Oncol, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Song Z, Wang Y, Zhou Y, et al. : Nomograms to predict the prognosis in malignant ovarian germ cell tumors: a large cohort study. BMC Cancer 22:257, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu YL, Manning-Geist BL, Knezevic A, et al. : Predicting outcomes in female germ cell tumors using a modified International Germ Cell Cancer Collaborative Group classification system to guide management. Gynecol Oncol 170:93–101, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Frazier AL, Stoneham S, Rodriguez-Galindo C, et al. : Comparison of carboplatin versus cisplatin in the treatment of paediatric extracranial malignant germ cell tumours: A report of the Malignant Germ Cell International Consortium. European journal of cancer (Oxford, England : 1990) 98:30–37, 2018 [DOI] [PubMed] [Google Scholar]
- 53.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 33:195–201, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kondagunta GV, Bacik J, Donadio A, et al. : Combination of paclitaxel, ifosfamide, and cisplatin is an effective second-line therapy for patients with relapsed testicular germ cell tumors. J Clin Oncol 23:6549–55, 2005 [DOI] [PubMed] [Google Scholar]
- 55.Loehrer PJ, Gonin R, Nichols CR, et al. : Vinblastine plus ifosfamide plus cisplatin as initial salvage therapy in recurrent germ cell tumor. J Clin Oncol 16:2500–4, 1998 [DOI] [PubMed] [Google Scholar]
- 56.Feldman DR, Sheinfeld J, Bajorin DF, et al. : TI-CE high-dose chemotherapy for patients with previously treated germ cell tumors: results and prognostic factor analysis. J Clin Oncol 28:1706–13, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Adra N, Abonour R, Althouse SK, et al. : High-Dose Chemotherapy and Autologous Peripheral-Blood Stem-Cell Transplantation for Relapsed Metastatic Germ Cell Tumors: The Indiana University Experience. J Clin Oncol 35:1096–1102, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Beyer J, Stenning S, Gerl A, et al. : High-dose versus conventional-dose chemotherapy as first-salvage treatment in patients with non-seminomatous germ-cell tumors: a matched-pair analysis. Ann Oncol 13:599–605, 2002 [DOI] [PubMed] [Google Scholar]
- 59.Lorch A, Bascoul-Mollevi C, Kramar A, et al. : Conventional-dose versus high-dose chemotherapy as first salvage treatment in male patients with metastatic germ cell tumors: evidence from a large international database. J Clin Oncol 29:2178–84, 2011 [DOI] [PubMed] [Google Scholar]
- 60.Pico JL, Rosti G, Kramar A, et al. : A randomised trial of high-dose chemotherapy in the salvage treatment of patients failing first-line platinum chemotherapy for advanced germ cell tumours. Annals of oncology : official journal of the European Society for Medical Oncology 16:1152–1159, 2005 [DOI] [PubMed] [Google Scholar]
- 61.SEER cancer statistics review 1975–2004: Average years of life lost per person dying of cancer, all races, both sexes. http://seer.cancer.gov/csr/1975_2004/results_figure/sect_01_zfig.19.pdf. accessed May 22, 2023
- 62.Howlader N, Noone A, Krapcho M, et al. : SEER Cancer Statistics Review, 1975–2018, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/csr/1975_2018/, based on November 2020 SEER data submission, posted to the SEER web site, April 2021 [Google Scholar]
- 63.Kollmannsberger C, Beyer J, Liersch R, et al. : Combination chemotherapy with gemcitabine plus oxaliplatin in patients with intensively pretreated or refractory germ cell cancer: a study of the German Testicular Cancer Study Group. J Clin Oncol 22:108–14, 2004 [DOI] [PubMed] [Google Scholar]
- 64.Einhorn LH, Brames MJ, Juliar B, et al. : Phase II study of paclitaxel plus gemcitabine salvage chemotherapy for germ cell tumors after progression following high-dose chemotherapy with tandem transplant. J Clin Oncol 25:513–6, 2007 [DOI] [PubMed] [Google Scholar]
- 65.Miller JC, Einhorn LH: Phase II study of daily oral etoposide in refractory germ cell tumors. Semin Oncol 17:36–9, 1990 [PubMed] [Google Scholar]
- 66.Porcu P, Bhatia S, Sharma M, et al. : Results of treatment after relapse from high-dose chemotherapy in germ cell tumors. J Clin Oncol 18:1181–6, 2000 [DOI] [PubMed] [Google Scholar]
- 67.Lafin JT, Singla N, Woldu SL, et al. : Serum MicroRNA-371a-3p Levels Predict Viable Germ Cell Tumor in Chemotherapy-naïve Patients Undergoing Retroperitoneal Lymph Node Dissection. European Urology 77:290–292, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Fizazi K, Pagliaro L, Laplanche A, et al. : Personalised chemotherapy based on tumour marker decline in poor prognosis germ-cell tumours (GETUG 13): a phase 3, multicentre, randomised trial. Lancet Oncol 15:1442–1450, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Fung C, Dinh PC, Fossa SD, et al. : Testicular Cancer Survivorship. J Natl Compr Canc Netw 17:1557–1568, 2019 [DOI] [PubMed] [Google Scholar]
- 70.Hjelle LV, Gundersen POM, Hellesnes R, et al. : Long-term serum platinum changes and their association with cisplatin-related late effects in testicular cancer survivors. Acta Oncol 57:1392–1400, 2018 [DOI] [PubMed] [Google Scholar]
- 71.Lauritsen J, Bandak M, Kreiberg M, et al. : Long-term neurotoxicity and quality of life in testicular cancer survivors-a nationwide cohort study. J Cancer Surviv 15:509–517, 2021 [DOI] [PubMed] [Google Scholar]
- 72.Lubberts S, Groot HJ, de Wit R, et al. : Cardiovascular Disease in Testicular Cancer Survivors: Identification of Risk Factors and Impact on Quality of Life. J Clin Oncol:Jco 2201016, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Fosså SD, Dahl AA, Thorsen L, et al. : Mortality and Second Cancer Incidence After Treatment for Testicular Cancer: Psychosocial Health and Lifestyle Are Modifiable Prognostic Factors. J Clin Oncol 40:2588–2599, 2022 [DOI] [PubMed] [Google Scholar]
- 74.Sanchez VA, Shuey MM, Dinh PC Jr., et al. : Patient-Reported Functional Impairment Due to Hearing Loss and Tinnitus After Cisplatin-Based Chemotherapy. J Clin Oncol:JCO 2201456, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Armstrong GT, Chen Y, Yasui Y, et al. : Reduction in Late Mortality among 5-Year Survivors of Childhood Cancer. N Engl J Med 374:833–42, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Phillips SM, Padgett LS, Leisenring WM, et al. : Survivors of childhood cancer in the United States: prevalence and burden of morbidity. Cancer Epidemiol Biomarkers Prev 24:653–63, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ozdemir BC, Csajka C, Dotto GP, et al. : Sex Differences in Efficacy and Toxicity of Systemic Treatments: An Undervalued Issue in the Era of Precision Oncology. J Clin Oncol 36:2680–2683, 2018 [DOI] [PubMed] [Google Scholar]
- 78.van Dorp W, Haupt R, Anderson RA, et al. : Reproductive Function and Outcomes in Female Survivors of Childhood, Adolescent, and Young Adult Cancer: A Review. J Clin Oncol 36:2169–2180, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lone DW, Sadak KT, Miller BS, et al. : Growth Hormone Deficiency in Childhood Intracranial Germ Cell Tumor Survivors. J Endocrinol Metab 12:79–88, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Brown AL, Sok P, Scheurer ME, et al. : An updated assessment of 43,110 patients enrolled in the Childhood Cancer Research Network: A Children’s Oncology Group report. Cancer 128:2760–2767, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bajorin DF: The Graying of Testis Cancer Patients: What Have We Learned? Journal of Clinical Oncology 25:4341–4343, 2007 [DOI] [PubMed] [Google Scholar]
- 82.Feldman DR, Lorch A, Kramar A, et al. : Brain Metastases in Patients With Germ Cell Tumors: Prognostic Factors and Treatment Options--An Analysis From the Global Germ Cell Cancer Group. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 34:345–351, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Oing C, Oechsle K, Necchi A, et al. : Impact of primary metastatic bone disease in germ cell tumors: results of an International Global Germ Cell Tumor Collaborative Group G3 Registry Study. Ann Oncol 28:576–582, 2017 [DOI] [PubMed] [Google Scholar]
- 84.Errata. J Clin Oncol 35:812, 2017 [Google Scholar]
- 85.Fischer S, Tandstad T, Cohn-Cedermark G, et al. : Outcome of Men With Relapses After Adjuvant Bleomycin, Etoposide, and Cisplatin for Clinical Stage I Nonseminoma. J Clin Oncol 38:1322–1331, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Fossa SD, Dahl AA, Myklebust TA, et al. : Risk of positive selection bias in longitudinal surveys among cancer survivors: Lessons learnt from the national Norwegian Testicular Cancer Survivor Study. Cancer Epidemiol 67:101744, 2020 [DOI] [PubMed] [Google Scholar]
- 87.Kreiberg M, Bandak M, Lauritsen J, et al. : Cohort Profile: The Danish Testicular Cancer Late Treatment Effects Cohort (DaTeCa-LATE). Front Oncol 8:37, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]


