U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Feingold KR, Anawalt B, Blackman MR, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.

Cover of Endotext

Endotext [Internet].

Show details

Testicular Cancer: Pathogenesis, Diagnosis and Management with Focus on Endocrine Aspects

, MD, PhD, DMSc, , MD, PhD, , MD, DMSc, , MD, PhD, and , MD, PhD.

Author Information and Affiliations

Last Update: March 29, 2023.

ABSTRACT

Testicular cancer comprises different neoplasms, depending on the cell of origin and the typical age at presentation, but germ cell-derived tumors constitute the vast majority of cases. Testicular germ cell tumors (TGCT) can be diagnosed in every age group, but more than 90% of cases occur in young men. These tumors, comprising seminoma and nonseminoma, are derived from germ cell neoplasia in situ (GCNIS). Pathogenesis of TGCT associated with GCNIS partly overlaps with that of other developmental disorders of the male reproductive system within the testicular dysgenesis syndrome (TDS). Testicular somatic cell neoplasms, known as sex cord-stromal tumors, are relatively rare, but can have endocrine manifestations, such as precocious puberty or gynecomastia. In addition to its malignant features, cancer of the testis represents a developmental, endocrine, and reproductive problem. These issues are the focus of this chapter, and emphasis is given to aspects that are of interest to endocrinologists, including pediatric endocrinologists and andrologists. Management of invasive testicular tumors is largely handled by urologists and oncologists, thus only general information on surgical treatment, radiotherapy, and chemotherapy is presented. Impact of cancer treatment on the endocrine system, co-morbidities, fertility issues, and quality of life issues are also briefly reviewed. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

INTRODUCTION

Testicular cancer comprises a number of different neoplasms, depending on the cell of origin and the typical age at presentation (1, 2). Although several cell types in the testis can undergo neoplastic transformation, germ cell-derived tumors constitute the vast majority of cases of testicular neoplasms. The relative distribution depends on age: in young adult men nearly all tumors are germ cell tumors, whereas in patients aged 70 years or older, a large proportion of lymphomas and secondary carcinomas can be expected (2). In other words, as explained in detail further in the text, germ cell cancer can be diagnosed in every age group, but more than 90% of cases occur in young men, and this subgroup is the main focus of this chapter. Pathogenesis of testicular germ cell tumors (TGCT) of young adults partly overlaps with that of other developmental disorders of the male reproductive system, within the testicular dysgenesis syndrome (TDS) (3, 4, 5). Somatic cell tumors in the testis, known as sex cord-stromal neoplasms are relatively rare, but are also discussed in this chapter. These neoplasms can have endocrine manifestations due to their origin from endocrine cells.

In addition to its malignant features, cancer of the testis represents a developmental, endocrine, and reproductive problem. These issues are the focus of this chapter, and emphasis is given to aspects that are of interest to endocrinologists, including pediatric endocrinologists and andrologists. The contribution of andrologists and endocrinologists to the overall management of patients with testicular cancer is very important, especially concerning early diagnosis, fertility issues, testosterone deficiency, and the impact of treatment on the quality of life of the patients, the majority of whom are young adults. We have to emphasize that only very general information on surgical and oncological treatment options (e.g., chemotherapy) is presented here. However, urologists and oncologists, who are responsible for the clinical management of testicular tumors, would also benefit from learning about the pathogenesis and andrological aspects of testicular cancer summarized in this chapter.

GERM CELL TUMORS (GCT)

Germ cell tumors (GCT) are characterized by extreme phenotypic heterogeneity (2). They display features of pluripotency and about half of them (nonseminomas) can differentiate into virtually any somatic tissue type within the so-called teratomas that recapitulate early embryonic differentiation (6, 7, 8). Although in comparison to other solid tissue cancers, GCTs are relatively rare, they constitute the most common form of solid tissue cancer of young age. The typical localization of GCT is the testis in males and ovary in females, but GCTs may also be found outside the gonads, thus these tumors are named extragonadal GCTs (8, 9). Extragonadal GCTs occur most often in children of both genders, preferentially along the body midline (intracranial, pineal, mediastinal) but can occur also in adults (8, 9). An increased frequency of extragonadal GCTs, especially in mediastinum, has been associated with Klinefelter syndrome (10). It is important to remember that a large proportion of cases of extragonadal GCT in retroperitoneal locations are associated with pre-malignant changes in testicles, and can be assumed early metastases of testicular neoplasms, although a multi-site development of GCT cannot be completely excluded (8, 9, 11, 12). This chapter focuses on the testicular GCT. For the extragonadal GCT in children and adults, the reader should consult specialized literature.

Classification and Histopathology of Testicular Germ Cell Tumors (TGCT)

Testicular germ cell tumors (TGCT) are by far the most frequent neoplasms of the testis and comprise approximately 90-95% of cases. They may affect infants (rarely), young men (commonly) and elderly men (rarely). The TGCTs of childhood are mentioned only briefly at the end of this section. The diagnosis and management aspects of TGCT in adult men is the focus of this chapter.

The most commonly accepted and currently used classification is the WHO classification of testicular tumors. The main changes occurred in 2016 when the WHO consensus panel proposed a thorough revision of the classification, which for the first time was based on biological evidence (2). The most radical change was the new division of TGCT into two major groups according to the origin from germ cell neoplasia in situ (GCNIS), and coining the term GCNIS, replacing several previously used and confusing names (2, 13). The latest 2022 WHO classification update proposed only minor adjustments; the most important is recognition of gonadoblastoma as a precursor similar to GCNIS and proposing placing seminoma under the major category of germinomas (14).

For use by non-pathologists, a simplified division of TGCT shown in Table 1.

Table 1.

Simplified Classification of Testicular Germ Cell Tumors (TGCT), Based on the WHO Classification

GCT derived from germ cell neoplasia in situ (GCNIS)
Non-invasive lesions: GCNIS (9064/2)* and gonadoblastoma (9073/1)
Germinomas:
Seminoma, pure (9061/3)
Seminoma with syncythiotrophoblastic cells
Nonseminomatous (non-germinomatous) GCT, pure
Embryonal carcinoma (9070/3)
Yolk sac tumor, postpubertal type (9071/3)
Trophoblastic tumors, incl. choriocarcinoma (9100/3)
Teratoma, postpubertal type (9080/3), incl. teratoma with somatic-type transformation (9084/3)
Nonseminomatous (non-germinomatous) mixed germ cell tumors (9085/3)
Regressed GCTs (9080/1)
GCT unrelated to GCNIS
Spermatocytic tumor (9063/3)
Prepubertal (pediatric) tumors
Teratoma, prepubertal type (9084/0)
Dermoid cyst
Epidermoid cyst
Yolk sac tumor, prepubertal type (9071/3)
Prepubertal type testicular neuroendocrine tumor (8240/3)
Mixed prepubertal type tumors (9085/3)
*

Footnote: The codes in parentheses are from the International Classification of Diseases for Oncology (ICD-O-3).

Precursor Lesions

The TGCT of young adults originate from a common precursor, germ cell neoplasia in situ (GCNIS), initially termed carcinoma in situ (CIS) testis (13, 15). GCNIS is considered to originate from developmentally arrested immature germ cells (gonocytes) that fail to differentiate to spermatogonia (4, 16, 17). Accordingly, morphology of GCNIS cells resembles closely that of fetal gonocytes, but with more irregular chromatin in the nuclei. GCNIS cells are located inside seminiferous tubules, most frequently in a single row along the basement membrane (Figure 1).

Gonadoblastoma is a preinvasive lesion which occurs almost exclusively in individuals with disorders of sexual development (DSD). This lesion is most often found in female patients with mixed gonadal dysgenesis (45,X/46,XY) or Turner Syndrome but can also occur in males (2, 18, 19). Gonadoblastoma cells and GCNIS cells have a very similar gonocyte/oogonium-like phenotype, but the surrounding somatic cells are different; GCNIS cells are present inside seminiferous tubules, which are usually well developed but may be hypoplastic and contain immature Sertoli cells, while gonadoblastoma consists of groups of germ cells, which are nested in small stromal cells similar to granulosa cells (19, 20, 21, 22). GCNIS and gonadoblastoma can be present in the same gonad and there are also lesions with morphology in between the two entities (18, 19). The clinical course of pure gonadoblastoma may be benign, but it has a potential to transform into a malignant germ cell tumor, especially if accompanied by GCNIS and greater virilization of the patient (19, 22, 23, 24).

Figure 1. . Histology of germ cell neoplasia in situ (GCNIS).

Figure 1.

Histology of germ cell neoplasia in situ (GCNIS). The upper panel (hematoxyllin-eosin, HE staining shows a low magnification view of GCNIS in a typical pattern with only GCNIS cells and Sertoli cells present inside tubules. The tubules with neoplasia have a smaller diameter than normal seminiferous tubules. On the right side of this image a few tubules with decreased spermatogenesis are visible. The lower left image shows a fragment of a tubule with GCNIS side-by-side with a tubule with preserved spermatogenesis; note the large GCNIS nuclei. The lower right image displays GCNIS cells visualized by immunohistochemical staining for placental-like alkaline phosphatase (PLAP).

The immunohistochemical profiles of GCNIS and gonadoblastoma cells are virtually identical and resemble very closely those of primordial germ cells and fetal gonocytes (21, 25, 26). This was subsequently confirmed by comparative studies at the transcriptional level using microarrays (17, 27, 28). Among many genes highly expressed in GCNIS cells (as well as gonadoblastoma, normal fetal germ cells and several TGCTs) the following should be mentioned because of their interesting biological function (germ cell survival or maintenance of embryonic stem cell pluripotency) and usefulness in histopathological diagnosis (24): OCT4 (29), AP2-gamma/TFAP2C (27, 30), NANOG (27, 31, 32), KIT (25, 33), TP53 (34), and LIN28 (35, 36).

In addition to protein-coding genes, GCNIS cells display a specific profile of embryonic-type micro-RNAs (miRNA); miR-371-3 cluster, miR-302 and miR-367 (37, 38). This miRNA profile is also detectable in overt TGCT, except teratoma (38), see the description in the section on serum markers.

Testicular Germ Cell Tumors (TGCT) Derived From GCNIS

As mentioned in the introduction, TGCT of young adult men display very variable histology (some examples are shown in Figure 2) and are divided into seminomas (under the main GCT category of germinomas which occur in men and women) and nonseminomas (non-germinomas) (2, 14, 39).

Figure 2. . Histology of main types of testicular germ cell tumors.

Figure 2.

Histology of main types of testicular germ cell tumors. The large images show a general histology pattern of a seminoma (upper panel) and two most often seen types of nonseminoma: undifferentiated embryonal carcinoma (middle panel) and teratoma, a tumor displaying differentiation into various somatic tissues (bottom panel). Small square pictures on the right show cellular characteristics in a greater magnification. All sections are stained with hematoxylin-eosin (HE).

Seminomas are most often diagnosed in the 25- to 40-year-old age group, whereas nonseminomatous tumors occur in even younger men (adolescence to 30 years). Both types originate from GCNIS (2, 16). Seminoma resembles a mass of immature germ cells; the tumor cells are morphologically very close to GCNIS cells and proliferate as a homogeneous tumor, which retains features of germinal lineage. The gene expression profile of seminoma is similar to that of GCNIS and fetal gonocytes, and virtually identical to the female equivalent of germinoma, called dysgerminoma (26, 39).

Nonseminomatous tumors display a variety of histological forms and contain undifferentiated embryonal carcinoma and somatic components partly differentiated along embryonic lineage of any tissue type (1, 2, 8). Nonseminomas also contain extra-embryonic tissue components (yolk sac tumor and choriocarcinoma). The combined or mixed tumors contain elements of seminoma and nonseminoma but are classified and clinically treated as nonseminoma, which usually has more severe clinical course than seminoma (1, 2).

Testicular Germ Cell Tumors Not Associated with GCNIS

PREPUBERTAL (PEDIATRIC) TGCTs

These tumors occur in early childhood (between birth and approximately 5 years of age) and comprise two main histological types: yolk sac tumor of the prepubertal type and mature teratoma (including dermoid cyst) (Table 1). Prepubertal teratomas can contain secondary neuroendocrine elements, which can overgrow the tumor, hence, the latest WHO classification added this entity as the third subtype (14). The histology of the prepubertal tumors does not differ from the adult equivalents, which are components of nonseminoma (2). Likewise, these tumors display similar characteristic transcriptome and micro-RNA profiles (40, 41). In contrast to the TGCT derived from GCNIS, the tumor genome does not display isochromosome 12p. The etiology and pathogenesis of infantile TGCT remain unknown. These tumors are assumed to originate from primordial germ cells (PGC) but there is no known precursor lesion of GCNIS/gonadoblastoma type, neither are there signs of testicular dysgenesis (2, 42, 43).

SPERMATOCYTIC TUMOR

This rare tumor occurs primarily in older men (median age at diagnosis around 50-55 years, range 25-94) but occasionally can be diagnosed in men in in the third decade of life (44). Spermatocytic tumor has no extragonadal or ovarian counterpart, and occurs exclusively in post-pubertal testis (1, 2, 8). This tumor is not derived from GCNIS (45) and has the expression profile similar to spermatogonia B or early primary spermatocytes (26, 46, 47, 48). Spermatocytic tumors appear to grow from clonally expanding germ cells, which are committed to but have not yet entered meiosis. The following de novo genetic aberrations causing increased spermatogonial proliferation have been identified; amplifications in chromosome 9p encompassing DMRT1 locus (46), rare gain-of-function mutations in genes encoding FGFR2, FGFR3, HRAS, NRAS, PTPN11, and simultaneous whole chromosome gains of chromosome 9 and chromosome 20 combined with a loss of chromosome 7 (49, 50). Some of these mutations that occur spontaneously in germ cells of aging men have been depicted as “selfish mutations” because - if transmitted to next generation - can cause severe inborn skeletal abnormalities in the offspring, such as achondroplasia, hypochondroplasia, thanatophoric dysplasia or Costello syndrome (51).

Etiology and Pathogenesis of Testicular Germ Cell Cancer of Young Adults (Derived From GCNIS)

Testicular Dysgenesis Syndrome (TDS)

The association of testicular cancer with poor testicular function, cryptorchidism, hypospadias, and abnormal testicular development led to a hypothesis that poor gonadal development and testicular neoplasia are etiologically linked. A concept of TDS was proposed, in which testicular cancer is one of the symptoms, in addition to other phenotypes, including cryptorchidism, hypospadias, shortened anogenital distance (AGD), reduced Leydig cell function, and decreased spermatogenesis (3, 5). This hypothesis is supported by histological studies showing that dysgenetic features, such as undifferentiated tubules with visibly immature Sertoli cells, clusters of poorly differentiated tubules, or hyaline bodies, often seen in association with testicular cancer, are not uncommon among men referred to andrology clinics because of fertility problems (3, 5, 18, 71). It is important to underline that not all cases of genital malformations and infertility are a part of TDS. Milder phenotypes linked to impaired development of the testis in fetal life due to diverse environmental or lifestyle-related factors can be considered part of TDS (5, 77). Severe genital malformations caused by genetic disorders are clearly a part of DSD but there is a partial overlap between the two syndromes (18). However, TDS severity and prevalence are undoubtedly modulated by the genetic variability (polymorphisms) and epigenetics (5). A schematic representation of the pathogenesis and manifestations of TDS is depicted in Figure 4.

Figure 4. . Schematic illustration of aetiology and pathogenesis of disorders grouped within Testicular Dysgenesis Syndrome (TDS).

Figure 4.

Schematic illustration of aetiology and pathogenesis of disorders grouped within Testicular Dysgenesis Syndrome (TDS). The TDS concept implicates disturbed function of testicular somatic cells (Leydig- and Sertoli cells) caused by inherited genetic variation (gene polymorphisms) in combination with environmental /lifestyle factors acting during early development. Dysfunction of the somatic cells results in disturbed hormonal homeostasis and causes impaired germ cell differentiation. Depending on the severity of the impairment, multiple outcomes or phenotypes may occur, ranging from reduced anogenital distance (AGD), genital malformations to testicular cancer. Note that the most severe forms of TDS (disorders of sex development with gonadoblastoma (GDB) or GCNIS are the least frequently seen, whereas the mildest forms, such as impaired spermatogenesis are quite common. Modified from Skakkebæk et al., Hum Reprod 2001 (3) and Physiol Rev 2016 (5).

Genomics of TGCT and Predisposing Polymorphisms

Tumors derived from germ cells via GCNIS stage are characterized by the presence of a nearly universal aneuploidy (polyploidization) and amplification of chromosome 12p, often in the form of an isochromosome (8, 78). Additional secondary genomic aberrations in invasive TGCT include rare recurring chromosomal gains or losses (in particular loss of chromosome Y), and fusion transcripts (79, 80). Strikingly few oncogenic mutations have been reported in TGCTs, and the short list of affected genes includes only KIT (predominantly in seminomas) and KRAS (mainly in nonseminomas) (79, 81, 83).

Although testicular cancer in most cases is a sporadic disease, the familial risk of testicular GCT is among the highest when compared to other cancers; brothers and sons of TGCT cases have an 8-10-fold and 4-6-fold increased risk, respectively (84). A twin study from Nordic countries estimated the heritability of testicular cancer as 37%, with 24% attributed to shared environment (85). Recent studies using next generation sequencing technology confirmed the absence of specific oncogenic driver mutations that would explain a high heritable risk of testicular cancer (86). The bulk of inherited risk is instead caused by a constellation of unfavorable gene variants, which jointly account for estimated 44% of testicular cancer heritability (87). Men with a polygenic risk score in the 95th percentile have a 6.8-fold increased risk of TGCT compared to men with median scores (87). This estimation is based on numerous genome-wide association studies (GWAS) performed in recent decades, which revealed a large number of significant gene variants (markers) associated with TGCT risk (87, 88, 89, 90, 91, 92, 93). The strongest association, which was identified in all GWAS, and most interesting from the biological point of view, is with a cluster of single nucleotide polymorphisms (SNPs) within or near KIT ligand gene (KITLG) and in genes downstream of the KIT/KITLG/MAPK signaling pathway, e.g., SPRY4. This pathway is essential for germ cell migration and survival, is highly active in GCNIS, with the frequent secondary gain-of-function mutations identified in seminomas (33, 81, 82, 83, 89, 94, 95). Among other significant SNPs associated with TGCT risk, the most interesting are DMRT1, a transcription factor involved in sex differentiation and regulation of meiosis (96, 97), PRDM14 and DAZL, factors involved in primordial germ cell specification and differentiation, AMHR2, the receptor for anti-Mullerian hormone (AMH), AR, the androgen receptor, and several genes in the telomerase and DNA repair pathway (87, 90, 91, 92, 93). It is biologically important that several predisposing variants are potentially associated with germ cell differentiation and testis development, including hormone regulation, and there is an overlap with some pathways involved in TDS.

Current Views on the Environmental Etiology of TGCT

In spite of significant recent inroads into the understanding of the pathogenesis of TGCT, etiology of these tumors, and especially the reason for the rise in incidence, remains obscure. The high incidence of testicular cancer in subjects with congenital errors of gonadal development strongly implicates the involvement of intrauterine factors and perinatal factors. We believe that the neoplastic transformation of male germ cells occurs during their pre-meiotic development, and this happens preferentially in individuals with genetic susceptibility. GCNIS cells and primordial germ cells share some distinct features, such as expression of embryonic pluripotency factors, low DNA methylation and constellation of miRNAs and histone modifications (4, 5, 17, 27, 28, 37, 39, 99, 100).

The mechanisms of neoplastic transformation of early germ cell are not known. There is a growing consensus that there may be multiple mechanisms and testicular cancer is a multifactorial and polygenic disorder. A disturbance in the fetal programming of gonadal development may be a result of an intrauterine hormonal imbalance, which in turn may be caused by a genetic disorder or by an impact of an exogenous factor targeting a key pathway, e.g., androgen signaling, KIT-KITLG signaling, DMRT1 signaling and regulation of meiosis, the TGF-beta superfamily regulation (including Nodal pathway) or the WNT pathway, leading to a delay in the testis development and maturation of fetal gonocytes (3, 4, 91, 98, 94, 101, 102, 103).

As mentioned above, the rising and quickly changing incidence of testicular cancer in well-developed countries suggests a possible adverse influence of environmental or lifestyle-related causative factors. In recent decades a great number of potent natural and synthetic hormones and hormone antagonists have been identified in environment. Observations in wildlife and experiments in laboratory animals exposed to synthetic hormones and a broad range of endocrine disrupters suggest that these substances can cause a disturbance of hormonal milieu of the developing gonad and disturb differentiation of early germ cells (3, 4, 5, 104, 105, 106). Whether or not the endocrine disrupters have contributed to the rise in testicular cancer remains to be demonstrated. Human exposure studies are difficult, mainly because of the long interval between the vulnerable period of early development and manifestation of cancer development in adulthood. There is also the problem of the plethora of confounding factors, including mixtures of possible contaminating factors present in food, packaging, cosmetics, and other products of daily life. Therefore, the evidence for the role of endocrine disrupters in the etiology of TGCT remains scarce. Among few existing data one can mention a higher serum level of some persistent organic pollutants found in mothers of men with testicular cancer (107), and a greater burden of p,p′-DDE (a DDT metabolite) in serum of testicular cancer patients (108). For more information on the existing evidence and possible mechanisms the reader is referred to recent review articles on the subject (109, 110). It is clear that further studies in large cohorts of patients and controls are needed to identify the causative factors behind the observed testicular cancer epidemics. Few data exist concerning postnatal exposures and risk of TGCT. A heavy use of marijuana (cannabis) has been consistently reported as a risk factor for nonseminoma but the mechanism remains to be elucidated (111, 112).

Diagnosis and Staging of Testicular Germ Cell Neoplasms

PREINVASIVE LESIONS AND TESTICULAR BIOPSY

All attending physicians should consider a possibility of incipient germ cell neoplasia in men with infertility, especially those with small testes, a history of cryptorchidism, or previous testicular tumors, or in individuals from the high-risk group of DSD (5, 68, 69). The preinvasive stage of GCNIS is asymptomatic, so diagnosis at this early stage is sporadic. Ultrasonographic examination can be helpful in some cases. GCNIS or incipient TGCT are frequently associated with an irregular echo pattern or microcalcifications /microlithiasis (113, 114, 115, 116). However, ultrasonic microlithiasis is very common and can be present in normal men or patients with mild disorders, so it alone is not an indication for a testis biopsy. A biopsy is indicated if microlithiasis is accompanied by additional risk factors, e.g., history of cryptorchidism, a family history of testicular cancer, atrophic testis (<12 mL), or poor semen quality, especially azoospermia (117, 118). Surgical testicular biopsy is currently the only sure diagnostic procedure for GCNIS diagnosis (119). Experience in the use of needle biopsies for GCNIS diagnosis is limited and this method is not commonly used in the clinic. Regardless of the method of obtaining the testis tissue, examination for GCNIS by immunohistochemistry in a diagnostic biopsy or any leftover material after testicular sperm extraction in fertility clinics is now considered mandatory (119, 120, 121).

GCNIS is present in the testis contralateral to a testicular tumor in approximately 5% of cases, with a range of 4% to 8%, depending on the studied cohort (71, 75, 121, 122). Screening for the contralateral GCNIS to prevent the appearance of metachronous bilateral TGCT has been practiced in several countries, including nationwide in Denmark and in some centers in other European countries (Germany, Austria, the Netherlands). Most of other centers adopted a recommendation of the European Association of Urology (123) to take contralateral biopsies only in patients at risk for GCNIS presence, such as by a history of cryptorchidism, a small testis volume (<12 ml), or testicular microlithiasis (75, 76). In the USA and Canada, the contralateral biopsy is rarely practiced, and American guidelines recommend this procedure only if the contralateral testis is cryptorchid, shows marked atrophy, or suspicious ultrasonic changes, but not microlithiasis alone (124, 125). Our recommendation is to offer a contralateral biopsy to all patients at the time of surgery for the primary TGCT, perhaps with the exception of men older than 50, because of a minimal risk of GCNIS. Added benefits of a biopsy are a good assessment of the fertility potential of the other testicle, and the peace of mind for the patient concerning the possibility of bilateral cancer (126). Mild post-biopsy complications (3%) are rare and the procedure is well accepted by the patients (127). In young men with normal size testicles, two-site biopsies may be considered to decrease a chance of a false-negative biopsy (126). False-negative cases are relatively rare (128) but the procedure does not completely eliminate the risk of the second TGCT. In the Danish nationwide study, the cumulative incidence of the metachronous TGCT was 1.9% (after median 20-year follow-up) compared to 3.1% in a non-biopsied earlier cohort of patients (121). However, in a sub-cohort of Danish patients, in whom the contralateral biopsy was performed with a quality control and obligatory immunohistochemistry with GCNIS markers, the cumulative rate of the second cancer (after similar follow-up period rate) decreased to 0.95% (121).

Technical aspects of surgical biopsy are important. The tissue fragment has to be sufficiently large (approx. 3x3x3 mm) and care has to be taken to avoid damage to the specimen, which should be dropped directly to a container with a fixative solution. For morphology evaluation Bouin's or similar solutions are preferred because formalin causes shrinkage artefacts, while for immunohistochemistry buffered formalin is preferred. As for any diagnostic testis biopsy, the contralateral biopsy evaluation must include immunohistochemical staining for at least one (better two) of the known markers for GCNIS, e.g., placental-like alkaline phosphatase (PLAP), OCT-3/4 (POU5F1), PDPN/M2A/D2-40, or AP2γ-TFP2AC (120, 122, 129). An example of immunohistochemical PLAP staining for GCNIS detection is shown in Figure 1.

SEMEN ANALYSIS

Poor spermatogenesis is a good indication that the patient may be at risk of harboring GCNIS (5, 68, 69, 75, 76, 130, 131). Semen analysis cannot as yet be used alone for detection of early stages of testicular cancer. However, it has been known for a long time that GCNIS cells may be occasionally found in semen (132). Over the years, progress has been made towards establishing better quality immunocytochemical detection of GCNIS or early invasive intratubular tumor cells in semen, and an improved automated double-staining assay has been developed (133, 134, 135). Unfortunately, the sensitivity of the cytological method is not high enough for screening, but it can be used as an additional part of semen analysis in experienced andrological centers performing immunohistochemistry (135). It is also not possible to use a micro-RNA (miRNA)-based assay, especially miRNA-371a-3p, a very promising serum marker for invasive TGCT, for detection of GCNIS in seminal fluid, despite that this embryonic-type miRNA is secreted by GCNIS cells (37). Even though some patients with GCNIS can have measurable miR-371a-3p in serum (136), the presence of the same miRNAs in normal germ cells in control patients with non-malignant conditions precludes using this assay in seminal fluid (137, 138, 139).

SERUM TUMOR MARKERS, DIAGNOSIS AND MONITORING OF OVERT TGCT

In the vast majority of cases a scrotal mass is usually the first presentation of testicular cancer, with tenderness reported by very few patients. In a few percent of testicular cancer cases, the presenting symptoms are the result of metastatic disease. They are usually uncharacteristic and may include lumbar pain, palpable abdominal mass, supra-clavicular lymph node enlargement, and in rare cases pulmonary symptoms (1).

The majority of primary and metastatic germ cell tumors secrete proteins and other biochemical products that can be detected in circulating blood. These biochemical serum tumor markers are very helpful in diagnosis and monitoring of testicular cancer (38, 124, 140).

The most important serum markers established in clinical practice are human chorionic gonadotropin (HCG), alpha-fetoprotein (AFP), and lactate dehydrogenase (LDH), the first two mainly useful to detect nonseminomas (1, 140). Among nonseminomatous tumors, choriocarcinoma, which resembles a gestational trophoblast, produces large quantities of HCG, while yolk sac tumor, which is similar in morphology to the embryonic yolk sac, secretes AFP (38, 140, 141). In addition, LDH may be secreted by both seminomas and nonseminomas. LDH levels in serum tend to be higher in patients harboring tumors with an increased copy number of chromosome 12p, consistent with the genomic location of the LDHB gene (142). Increased concentrations of LDH in the absence of AFP and HCG suggests the presence of seminoma. It is important to keep in mind that TGCT rarely occur in pure histological forms, and a subtype of seminoma with syncytiotrophoblastic cells can secrete HCG, which can be measurable in patients’ blood (14, 143). In preinvasive GCNIS and in most cases of pure seminoma, none of the above-mentioned markers are detectable in serum.

The most promising currently emerging serum markers are micro-RNAs (miRNA), among which miRNA 371a-3p is the best studied and most robust marker, and has already been proven valuable for detection of seminomas and nonseminomas, except for differentiated pure teratomas, in children and adults (38, 144, 145). Several large clinical studies clearly demonstrated that miRNA 371a-3p test has a much better specificity than the classical serum markers, and is especially useful for detection and monitoring of HCG- and AFP-negative seminomas (146, 147).

After diagnosis, careful clinical staging is necessary in each patient to decide for the most appropriate treatment strategy, and the measurements of circulating tumor markers are an important part of this process (1, 123, 124). There is a tendency for higher levels of tumor markers to be associated with a poorer prognosis. For example, the presence of syncytiotrophoblastic cells in the subtype of seminoma can lead to a mild increase of serum HCG, but the tumor responds well to treatment regardless of the presence of these cells, so more aggressive management should be avoided.

In addition to serum markers, scrotal ultrasonography is the first line diagnostic procedure. In general, seminoma is a homogenous tumor, whereas nonseminomas usually display heterogeneous patterns, with frequent hyperechoic calcified areas (148, 149). Additional imaging procedures help to evaluate the spread of disease, including CT scan of the chest and abdomen, or MRI (148, 149, 150).

Histopathological evaluation of the orchiectomy specimen is an essential part of the clinical staging. The important risk factors are tumor size, vascular/lymphatic invasion or the presence of tumor cells in rete testis, and the presence of pure embryonal carcinoma (152, 153).

The most commonly used staging and prognostic classification system is the TNM (tumor, node, metastases) System of the International Germ Cell Cancer Collaborative Group (GCCCG) and the American Joint Committee on Cancer (AJCC) (154). The stage grouping updated by the WHO Consensus (2), is shown in Table 2.

Table 2.

Stage Grouping According to TNM Classification

Stage 0 (pTis): Germ cell neoplasia in situ
Stage IA (pT1, N0, M0, S0): Tumor limited to testis and epididymis
Stage IB (pT2-4): as IA but with vascular/lymphatic, tunica or scrotal invasion
Stage II (any pT, N1-3, M0, S0-1): Metastasis in lymph nodes, serum markers normal or moderately increased
IIA (N1): lymph nodes <2 cm
IIB (N2, S1): lymph nodes ≥2 cm but <5 cm
IIC (N3, S1): lymph nodes ≥5 cm
Stage III (any pT, any N, M1, S0-3): Distant metastasis (spread beyond regional nodes)
IIIA (M1a, S0-1): Spread to non-regional nodes or lung
IIIB (M1a, S2): as IIIA but high serum markers
IIIC (M1a-b, S3): distant metastasis to sites other than IIIA, or very high serum markers

Footnote: pT=primary tumor, N=regional lymph nodes, M=distant metastasis, S=serum tumor markers (X=unknown, not available)

Management of GCNIS and Testicular Germ Cell Cancer

Early diagnosis of testicular neoplasia at the stage of GCNIS, followed by adequate treatment of GCNIS is capable of preventing progression to invasive tumors. Unfortunately, the vast majority of cases progress unnoticed to overt tumors. However, germ cell tumors are extremely radio- and chemo-sensitive and have apparently very high propensity to apoptosis, likely mediated by p53 (155, 156). Because of this sensitivity, and thanks to cisplatin-based chemotherapy regimens, TGCT is a highly curable malignancy, with more than 90% of patients reaching a sustained complete remission (157).

We give here only very general information concerning management of testicular cancer. The reader should consult specialized oncologic and urologic literature for management options pertinent to treatment-resistant tumors and metastatic disease.

TREATMENT OF GCNIS

Spontaneous regression of GCNIS has never been described and it is anticipated that all cases of GCNIS will eventually develop into TCGT. The following management options for GCNIS are available depending on a specific situation (158):

  • Orchiectomy - is the curative treatment with the highest assured success rate. It should always be performed on a testis with GCNIS or localized tumor when the second testis is not affected by malignancy (including GCNIS).
  • Radiotherapy - low dose radiotherapy is a good alternative to orchiectomy when GCNIS is present bilaterally or in the contralateral testis, so the patient can be spared a bilateral castration and lifelong androgen replacement therapy. The efficacy of radiotherapy with doses as low as 16 Gy was demonstrated in the early studies (159). Even though the lower dose better preserves the function of Leydig cells (160), more recent studies and current EAU guidelines recommend a dose of 18 Gy, given in fractions of 2Gy (123, 161). This dose of radiation will almost always destroy normal germ cells, so radiotherapy may be delayed in patients who wish to secure natural conception of a child.
  • Chemotherapy - is not an option to treat GCNIS, because a persistence or relapse has been reported in a high proportion of patients (121, 161). Furthermore, in some cases of extragonadal germ cell tumors treated with chemotherapy, testicular GCNIS progressed to metachronous overt testicular tumors (162). However, in patients with disseminated disease receiving chemotherapy, the risk of metachronous bilateral TGCT is lower (73, 163, 164).
  • Surveillance - is potentially hazardous since it increases the risk of metachronous bilateral TGCT (164) and GCNIS may progress to invasive cancer at any time. However, watchful surveillance may be an option after careful informed discussion of risks and monitoring with ultrasound examinations, especially if the patient wishes to defer treatment temporarily for the purpose of paternity.

In men who desire fatherhood, and in all very young men, cryopreservation of semen samples should be offered, if viable spermatozoa are detected.

TREATMENT OF OVERT SEMINOMAS AND NONSEMINOMAS

As far as the treatment of invasive germ cell tumors is concerned, the reader should consult the specialized urology and oncology guidelines.

Prior to surgery, all patients with TGCT should be offered full andrological evaluation and cryopreservation of semen. Endocrinologic evaluation of the patient should also preferably be done before surgery, with the reproductive hormone profile, including serum testosterone, gonadotropins, and inhibin B, if possible. This can be done together with the above-mentioned measurements of serum tumor markers. Pre-operative semen analysis and cryopreservation is especially important in patients with bilateral tumors or only one testicle. In the patients who have azoospermia or cryptozoospermia, and in whom it was impossible to retrieve sperm pre-operatively, testicular sperm extraction (TESE) at the time of orchiectomy (‘onco-TESE’) may be attempted in specialized centers (165). The best predicting factor of sperm retrieval is a small size of the tumor, because a chance of finding tubules with ongoing spermatogenesis increases with the distance from the tumor (166).

Radical orchiectomy remains the primary surgical treatment method of choice. Primary dissection of retroperitoneal lymph nodes (RPLND) was previously commonly practiced, especially in North America. The current consensus is to perform RPLND in experienced high-volume centers, and in selected cases, mainly in patients with nonseminoma and intermediate prognosis (1, 167, 168, 169). Nerve-sparing technique is preferred in order to avoid the loss of antegrade ejaculation, which is the commonest long-term complication of RPLND (169).

Methods of post-surgical management of overt TGCT are variable, depending upon the histological type of tumor (seminoma vs nonseminoma), levels of serum markers and stage of disease and the presence of residual retroperitoneal masses (1, 115, 151, 167). Adjuvant radiotherapy, e.g., of the retroperitoneal/para-aortic field, which was previously routinely used, is no longer recommended, because of the long-term risk of secondary malignancies (1, 167, 168).

Pure seminomas have a good prognosis and around 80% of patients with stage I seminoma (tumor confined to the testis) do not require any treatment after the surgery, thus most of the centers practice a surveillance strategy (151, 168, 170). Nonseminomas have a somewhat poorer prognosis (relapse rate is around 30%) and in some centers, patients with nonseminoma and high risk of relapse are treated with one course of adjuvant chemotherapy with cisplatin and etoposide (171, 172), but most centers currently recommend a surveillance strategy (1, 151, 152, 168, 172).

The most common post-surgical management of disseminated disease is systemic chemotherapy with a combination of cytotoxic drugs. The standard first line chemotherapy regimen is BEP (bleomycin, etoposide and cisplatin), administered in 3 or 4 cycles, depending on the patient’s prognosis (173, 174). It is very important to make a dynamic assessment of the progress of treatment through the early stages of chemotherapy, thus monitoring of serum markers is obligatory. Post-chemotherapy retroperitoneal surgery should be performed in patients with a residual tumor after BEP (151). In patients with relapse after first line treatment, salvage regimens and complex surgery for residual tumors need to be performed. Overall, the management is difficult, thus it should be carried out in specialized tertiary centers (1, 167).

The majority of relapses occur within 2 years of the initial treatment but late relapses are observed in some cases, therefore individual management of each patient and lifetime follow-up is advocated in some patients (1, 151, 167). Andrological follow-up should be coordinated with the post-surgery oncological management and controls (see the section on ‘Endocrine problems and late effects’).

SEX CORD-STROMAL TUMORS OF THE TESTIS

In adults, sex cord-stromal tumors of the testis are found in less than 5% of all testicular tumors, whereas in children, these tumors are more common and account for approximately 25% of cases (123, 175, 176, 177, 178). Most of these tumors are benign, only around 5% have malignant characteristics (2). The classification of the tumors by WHO (2016) is shown in a simplified form in Table 3 (2).

Sex cord-stromal tumors are derived from testicular somatic cells; Leydig cells, and Sertoli/granulosa cells. Despite a different cell of origin, some stromal tumors are sometimes misinterpreted as seminoma. A number of features can be used to distinguish sex cord-stromal tumors from germ cell tumors; Inhibin A and B are the best serum markers for this purpose (179, 180, 181). Inhibin A appears to be a common immunohistochemical marker for sex cord-stromal tumors, including Leydig cell, Sertoli cell and juvenile granulosa cell tumors (179, 182). Sertoli cell tumors are positive for anti-Mullerian hormone (183) and GATA-4 (184), while Leydig cell tumors express steroidogenic enzymes and INSL3 (185). Other useful immunohistochemical markers, include SOX9, calretinin, CD99, SF1 (2).

Table 3.

Sex Cord-Stromal Tumors of the Testis, Adapted from WHO Classification (ref. 2)

Leydig cell tumor (8650/1)
Malignant Leydig cell tumor (8650/3)
Sertoli cell tumor (SCT) (8640/1)
Malignant SCT (8640/3)
Large cell calcifying SCT (8642/1)
Intratubular large cell calcifying SCT (8643/1)
Granulosa cell tumors
Juvenile-type granulosa cell tumor (8622/1)
Adult-type granulosa cell tumor (8620/1)
The fibroma-thecoma tumors (8600/0)
Mixed sex cord-stromal tumors (8592/1)
Unclassified sex cord-stromal tumors (8591/1)
*

Footnote to Table 2: The codes in parentheses are from the International Classification of Diseases for Oncology (ICD-O-3).

Leydig Cell Hyperplasia and Tumors

Leydig cells are located in the interstitial compartment of the testis and are involved in the development of secondary male characteristics and maintenance of spermatogenesis by secretion of testosterone. Although Leydig cells in adult men are considered to be a terminally differentiated and mitotically quiescent cell type, in various disorders of testicular function, focal or diffuse Leydig cell hyperplasia is very common. Micronodules of Leydig cells are frequently seen in certain conditions associated with severe decrease of spermatogenesis or germinal aplasia, such as the Sertoli-cell-only syndrome (Del Castillo syndrome), cryptorchidism, or Klinefelter syndrome, when the nodules can be particularly florid (186, 187). A term 'Leydig cell adenoma' is used when the size of a nodule exceeds several- fold the diameter of a seminiferous tubule. It is unknown whether Leydig cell adenomas can progress further to form overt Leydig cell tumors, but even if it were the case, it is exceedingly rare. Morphological heterogeneity of hyperplastic Leydig cells is noticeable in some cases, and it has been shown that the micronodules contain a large proportion of immature Leydig cells (186, 187).

The mechanism of Leydig cell hyperplasia and larger nodules in the human male is still poorly understood. Leydig cell nodules of variable-size are common in patients with TDS and infertility, and the functional insufficiency of Leydig cells is often reflected by decreased testosterone/LH ratio whereas patients with overt Leydig cell tumors usually have an increased testosterone/LH ratio and high estradiol (186, 188). The disruption of hypothalamo-pituitary-testicular axis leading to an excessive stimulation of Leydig cells by LH can play a central role (186). This in turn leads to an increased renewal of immature, adult-type Leydig cells from their precursors. The immature cells are characterized by low numbers of Reinke crystals, a relatively high expression of a mesenchymal factor DLK1 and low amounts of INSL3 (187, 189, 190). However, Leydig cell hyperplasia is distinct from tumors that are usually solitary. Leydig cell hyperplasia and adenomas can be easily induced in rodents by administration of estrogens, gonadotropins, and a wide range of chemical compounds. Whether or not humans would be similarly susceptible to environmental effects remains to be elucidated.

Leydig cell tumors account for up to 5% of testicular neoplasms and occur in all age groups (2, 178, 191). Approximately 20% are found in boys, most often between five and ten years of age. There is perception that the prevalence of benign Leydig cell tumors among the adults is greater than previously thought, possibly thanks to improved detection of small nonpalpable nodules by modern imaging methods. Higher incidence numbers have also been reported in fertility centers that actively scan the testes of infertile patients (192).

In a subset of cases of Leydig cell tumors, activating mutations of the LH receptor (193, 194, 195) or G proteins (196, 197) can be detected. Constitutively activating mutations of LH receptor cause early Leydig cell hyperplasia and precocious puberty (193, 195, 197). Similarly, constitutively activating mutations of Gs-protein in Leydig cells or inactivation of PKAR1A (protein kinase cyclic adenosine monophosphate-dependent regulatory type 1 alpha) lead to hyperplasia and endocrine hyperactivity (178, 197). In adult Leydig cell tumors, germline fumarate hydratase mutations have been identified in a few cases which also had hereditary leiomyomatosis and renal cell cancer (199).

Precocious puberty (so-called testotoxicosis) is the presenting symptom in most of cases of Leydig cell adenomas or tumors in children, due to the excessive androgen production, mainly testosterone that causes growth of penis, pubic hair, accelerated skeletal and muscle growth, advancement of bone age, skin changes (acne, comedos, hair greasing), and adult-type odor of sweat. Androgen secretion in the pediatric cases is gonadotropin independent, and therefore LH and FSH remain low in spite of external signs of puberty (195). Approximately 10% of the boys also have gynecomastia that is caused by estrogens produced in excess due to aromatase activity in some of the tumors or peripheral aromatization of testosterone. It is important to keep in mind that transient gynecomastia or pseudo-gynecomastia can occur in newborns, in obese boys, and at puberty as a non-pathological condition (200). In adults, gynecomastia is a frequent condition with a reported prevalence of 32–65%, depending on the age and the criteria used for definition (summarized in 201). Gynecomastia is sometimes associated with loss of libido, impotence, and infertility (201). However, Leydig cell tumor is the cause in only 1-2% of cases, and the excessive androgen secretion by these tumors rarely causes notable effects in adults (202). Malignant tumors are hormonally active only in exceptional cases (2, 178).

In children, Leydig cell tumors are always benign and can be treated with surgical enucleation when the tumor is encapsulated (203). In adults malignant Leydig cell tumors have been found in 10-15% of patients, and inguinal orchiectomy is often used (204), while testis-sparing enucleation remains an option, if there are no signs of malignancy in the frozen preparation (205). The presence of cytologic atypia, necrosis, angiolymphatic invasion, increased mitotic activity, atypical mitotic figures, infiltrative margins, extension beyond testicular parenchyma, and DNA aneuploidy are associated with metastatic behavior in Leydig cell tumors (178, 202, 206) (see Figure 5).

Figure 5. . Histology of a Leydig cell tumor.

Figure 5.

Histology of a Leydig cell tumor. The appearance of tumor cells resembles normal Leydig cells. A section stained with hematoxylin-eosin (HE).

Malignant Leydig cell tumors do not respond favorably to conventional chemotherapy and irradiation (204). Survival time has ranged from 2 months to 17 years (median, 2 years), and metastases have been detected as late as nine years after the diagnosis (178, 202). Metastatic Leydig cell tumors require salvage surgery, radiotherapy and intensive chemotherapy regimens but in most cases the outcome is poor (188, 207). Therefore, follow-up of patients with malignant Leydig cell tumors has to be life-long. The remaining testis may be irreversibly damaged by longstanding high estrogen levels, resulting in both permanent infertility and hypoandrogenism (202, 206).

Testicular Adrenal Rest Tumors (TART)

Excessive secretion of adrenocorticotropin (ACTH) in 21-hydroxylase deficiency (congenital adrenal hyperplasia, CAH) or Nelson syndrome (post adrenalectomy status) may lead to development of hyperplastic interstitial nodules called testicular adrenal rests tumors (TART) resembling Leydig cell tumor or hyperplasia (191, 208, 209). These cells are hormonally active in secreting androgens. It is important to remember that the adrenal rests are almost invariably bilateral, whereas the Leydig cell tumors are usually unilateral. Adrenal rests can be treated by appropriate glucocorticoid substitution of the patient, which leads to gradual regression of the 'tumor' in 75 percent of cases (208, 209). TART have to be distinguished from benign hyperplasia, adenomas and malignant Leydig cell tumors (185).

Histopathological and endocrine evaluation covering both testicular and adrenal steroids and pituitary gonadotropins and ACTH are important in the differential diagnosis (185, 208). It would be an error to orchidectomize the CAH patients with TART, since the tumors are always benign and only some of them continue to be active after appropriate glucocorticoid substitution. In the patients who do not respond well to glucocorticoid replacement, or develop fibrosis, testis sparing surgery with enucleation of the larger nodules can be considered, if the testicles have become so large that they cause discomfort for the patient (209).

Sertoli Cell Tumors

Sertoli cells are the somatic cells in the seminiferous epithelium giving structural, metabolic, and hormonal support to spermatogenic cells. Sertoli cells cease their proliferation at puberty. In rare infantile cases, multiple foci of proliferating Sertoli cells have been described and proposed to be early intratubular forms of Sertoli cell tumors (210). The classification of these tumors according to the WHO (2, 178), is shown in Table 3.

Sertoli cell tumors often occur as a part of multiple neoplasia syndromes (see below). Rare Sertoli cell tumors which do not belong to a syndrome are called ‘not otherwise specified’ (NOS). About 5% of these tumors are malignant and can metastasize. The age of patients ranges from 18 to 80 years, but most of them are young adults (median age 30). Out of 60 patients, only four were younger than 20 years old in the series reviewed by Young et al. (211). The tumors typically are composed of sex cord cells with tubular differentiation, with a subset of tumors hyalinized, previously classified as sclerosing variant (2). Some tumors often contain lipid droplets but do not show any endocrine activity. The molecular origin is known only in a small proportion of tumors with sclerosing appearance, in which CTNNB1 mutations causing nuclear accumulation of β-catenin were identified (212). The tumors occurred in descended testes and were always unilateral. An infiltrative margin was found in four cases, but most of the tumors were well demarcated. The tumors were hormonally inactive, and only two patients with alcoholic cirrhosis also had gynecomastia. Eighteen pediatric cases were reported from the Kiel Pediatric Tumor Registry (176), but perhaps the histopathologic pattern was somewhat different, because the age of the children was very young, ranging from 0 to 14 months (median 4 months). Juvenile Sertoli cell tumors often showed infiltrative growth into adjacent tissue, dense cellularity and considerable proliferative activity. However, after surgical excision no local recurrences and no metastases occurred. Thus, these patients have a good prognosis. These Sertoli cell tumors can be treated by orchiectomy, and retroperitoneal lymphadenectomy is indicated only when there is radiographically detected retroperitoneal involvement (213).

Calcifying Sertoli cell tumors, are frequently found in association with two distinct multiple neoplasm syndromes, Carney complex and Peutz-Jeghers syndrome, however in the latter syndrome, the tumors belong to a distinct ‘intratubular’ morphological group (2, 178, 214). Association of large-cell calcifying Sertoli cell tumors with other neoplasms, particularly heart myxomas in Carney complex and gastrointestinal tumors in Peutz-Jeghers syndrome, should be kept in mind to reach an early diagnosis of these potentially fatal diseases.

The Carney complex is characterized by skin myxomas, heart myxomas, skin pigmentations, adrenal and testicular tumors, but other tumors can also occur (215). The testicular tumors are large-cell calcifying Sertoli cell tumors that are multifocal and bilateral, and should be distinguished from teratomas (Figure 6) (178, 216, 217). The tumors appear usually during the second decade of life (218). Only one malignant case has been reported in association with Carney complex (in an adult patient), whereas seven malignant tumors were reported in other patients with large-cell calcifying Sertoli cell tumors (218). These patients were older than 25 years. The malignant cases were unilateral and solitary in contrast to bilateral and multifocal occurrence of testicular tumors in Carney complex. Large-cell calcifying Sertoli cell tumors are usually not hormonally active, although elevated levels of serum inhibin B or testosterone have been reported, but other tumors of Carney complex, including Leydig cell tumors, can cause endocrine manifestations (181, 213, 214).

Figure 6. . Large cell calcifying Sertoli cell tumor isolated from a 12-year-old boy.

Figure 6.

Large cell calcifying Sertoli cell tumor isolated from a 12-year-old boy. The neoplastic tubules contain only large pale Sertoli cells and visible calcifications in the lumen (stained with PAS). Adjacent normal tubules show advanced spermatogenesis.

Two genetic loci for Carney complex have been identified on chromosome 2p16 (196) and 17q23-24 (219, 220). Germline mutations identified in Carney complex most often occur in type I-alpha regulatory subunit of protein kinase A, PRKAR1A (221, 222). Inactivating mutations of phosphodiesterase 11A212 and phosphodiesterase 8B213 are associated with bilateral adrenocortical hyperplasia in Carney complex patients. Genetic variation of the phosphodiesterase 11A gene can modify the development of the testicular tumors (223). Molecular genetic diagnosis is available to many of these patients. However, only a part of the mutations occur in the germ line, and therefore genetic analysis should be performed on affected tissues, such as the tumors, in cases of somatic cell line mutations.

In Peutz-Jeghers syndrome Sertoli cell tumors are similar in appearance to those found in Carney complex patients, but can be distinguished by typical intratubular proliferation of lightly eosinophilic cells with prominent basement membrane deposits, and occasionally, features of ovarian sex-cord tumors with annular tubules (178). Thus, the tumors are described as intratubular large-cell hyalinizing Sertoli cell neoplasia. These tumors may have strong aromatase activity and therefore be associated with gynecomastia and advanced bone age (224). No malignant testicular tumors have been reported in Peutz-Jeghers patients, but they have a highly increased risk of other neoplasms, especially colorectal, breast, pancreatic and ovarian cancers (214). Germline loss-of-function mutations in the STK11/LKB1 gene that encodes for a serine-threonine kinase causes Peutz-Jeghers syndrome in the majority of patients, allowing molecular genetic diagnostics (214, 224, 225).

Patients with the Sertoli cell tumors should be treated conservatively during childhood to give them a possibility for sperm banking before orchiectomy comes necessary (226). Autosomal dominant inheritance should be considered in genetic counselling. The patients should be frequently controlled with ultrasound examinations to follow changes in the tumors size, and reproductive hormone measurements. If precocious puberty and/or gynecomastia appear, aromatase inhibitors and antiandrogens can be used to prevent estrogen formation and androgen action (219, 226). There are currently no clear guidelines for the treatment of metastatic Sertoli cell cancers, but orchiectomy and retroperitoneal lymph node dissection are usually performed combined with individualized chemotherapy and radiotherapy (227). Without surgery, the metastatic disease progresses and 20-month median survival time of patients was reported (IQR: 6–30 months) (227).

Granulosa Cell Tumors

Juvenile-type granulosa cell tumors are rare but are the most common somatic testicular tumors in infants and occur during the first 6 months after birth (2, 176, 178, 228). These tumors were described in patients with undescended testes with abnormal sex chromosomes and ambiguous genitalia (176). Prominent differentiation into follicles and immature nuclei distinguishes juvenile granulosa cell tumors from other Sertoli cell tumors that express tubular differentiation (178, 229). Most of the immunohistochemical markers in these tumor types are similar, e.g., inhibin, calretinin, but also distinct, e.g., expression of FOXL2 (229). Juvenile granulosa cell tumors always have a good prognosis (230). Testicular tumors do not show endocrine hyperactivity, in contrast to ovarian juvenile granulosa cell tumors. Aberrant WNT signaling (231) and stimulatory G-protein mediated signaling (232) have been linked with juvenile granulosa cell tumors. The patients should be managed comprehensively by a multidisciplinary DSD team, and the treatment of tumors may require orchiectomy.

Adult-type granulosa cell tumors are comparable to the ovarian tumors, but are extremely rare (2, 178, 233). These tumors occur in adults at an average age of 42 years. Twenty percent of the patients have shown gynecomastia due to the hormonal activity of the tumor. Most of the tumors are benign, but some malignant cases have also been reported (230). Mutations in FOXL2 have been reported in adult ovarian granulosa cell tumors but only few secondary mutations have been found in testicular granulosa cell tumors (234, 235). The treatment is primarily surgical.

Fibroma-Thecoma Tumors

These exceedingly rare neoplasms are composed of fibroblastic cells of testicular stroma or tunica albuginea (2). These tumors are reported to be benign.

Mixed and Unclassified Sex Cord-Stromal Tumors

Tumors consisting of more than one stromal or tubular component or have indeterminate morphology are classified as mixed and unclassified sex cord-stromal tumors (2). Sex cord-stromal tumors can contain combinations of Leydig, Sertoli, granulosa, and theca cells, and are therefore called mixed tumors (178). Leydig cells can be difficult to recognize in these tumors. These tumors are rare and can occur at any age. Depending on the predominant cell type the tumors may behave differently. Gynecomastia, as a sign of endocrine activity, can be found in 10% of patients (2). These tumors are always benign in children, but in adults, malignancy can be found (232). Thus, most of the patients can be treated by orchiectomy, and lymph node dissection is indicated only in cases with overt malignant features on microscopic examination.

OTHER TESTICULAR TUMORS

Other tumors occurring in the testis are divided into miscellaneous and hematolymphoid tumors (2, 178). The first group includes ovarian epithelial-type tumors, serous or mucinous cystadenomas, adenocarcinomas, Brenner tumor, xantogranuloma, and hemangioma. The hematolymphoid tumors comprise malignant lymphomas (B-cell, NK/T-cell or follicular), plasmacytoma, myeloid sarcoma and Rosai-Dorfman disease (2, 178). In addition, testicular spread of malignant acute leukemia is common in young boys, and metastases from other solid tumors including the prostate gland, colon, kidney, stomach, pancreas, and malignant melanoma can be found in the testis of adults.

ENDOCRINE PROBLEMS AND LATE EFFECTS IN TESTICULAR CANCER PATIENTS

Testis Dysfunction and Fertility Issues

Relative imbalance of androgen signaling (excess or deficiency) causes the most pronounced secondary endocrine symptoms associated with testicular tumors. Testosterone is produced by tumors, such as Leydig cell tumors, or by normal Leydig cells stimulated by large amounts of hCG from some germ cell tumors. Excess of androgens would lead to precocious puberty in children (193, 194, 195). In addition, aromatization of androgens leads to a relative excess of estrogens, which causes impairment of spermatogenesis in adults and gynecomastia at any age (224, 236).

Testicular dysfunction in young adult patients with testicular cancer, who usually are in their best reproductive age, is a serious clinical problem. Patients with TGCT have poor spermatogenesis and decreased fertility even before the overt tumor has developed (67, 68, 69, 130) and before cytotoxic treatment (237, 238). Pathological features can include oligozoospermia or azoospermia, moderately decreased testosterone and elevated LH levels. If testicular biopsies are taken, a variable degree of testicular dysgenesis or atrophy is often seen, and in some cases further complicated by the presence of GCNIS. Examination of a contralateral biopsy in a patient with a unilateral tumor may show a similar picture; with at least 5-7% risk of the presence of GCNIS cells (71, 126).

Testicular function is further disturbed by treatment of malignancy. In recent years there is growing concern about adverse late effects of irradiation and chemotherapy, which induce severe impairment of spermatogenesis and DNA damage (1, 167, 168, 239, 240). Refinement of the dosage must be considered in each patient individually, to eradicate the neoplasm with least possible damage to the endocrine function. The eradication of GCNIS or a tumor by irradiation in bilateral cancer cases leads also invariably to the disappearance of all germ cells and sterility. This underlines the importance of semen cryopreservation before treatment, which will allow assisted reproduction treatment, if needed (238, 241).

All patients treated for testicular cancer require assessment of their reproductive hormones and spermatogenic capacity by semen analysis with respect to their future fertility. If semen banking or pre-operative sperm retrieval is not possible or if the patient has azoospermia or cryptozoospermia, testicular sperm extraction (TESE) at the time of orchiectomy (‘onco-TESE’) may be the only chance of fertility, and can be attempted in specialized centers (165, 166). TESE and subsequent intracytoplasmic sperm injection (ICSI) may be also an option for fertility treatment in some cases of post-chemotherapy azoospermia (242).

Fertility preservation is a serious challenge in children and young adolescents. In the adolescent boys who are unable to ejaculate, penile vibratory stimulation or electroejaculation can be considered (243). However, there is currently no treatment for prepubertal boys. There are ongoing studies attempting to optimize protocols of cryopreservation of immature testis tissue before cytotoxic treatment or orchiectomy (244). This approach would require in vitro or in vivo induction of spermatogenesis and gametogenesis, which has not yet been achieved satisfactorily in humans, but studies in experimental animals, including nonhuman primates are promising (245).

Long-Term Sequelae and Quality of Life

All patients treated for testicular cancer require monitoring of their reproductive hormone profiles not only with respect to their fertility, if paternity is desired, but also risk of testosterone deficiency and ensuing symptoms (246, 247, 248).

An increased risk of cardiovascular disease is present in TGCT survivors who have been treated with radio- or chemotherapy (249, 250) but a recent Danish study found that the risk decreases during long-term follow-up, although it remains elevated (251). Additional problems in survivors of TGCT treated with radiotherapy or chemotherapy are increased risk of peripheral neuropathy or ototoxicity (after chemotherapy) and sexual dysfunction (after chemotherapy and radiotherapy) (239, 240, 250, 251, 252).

Survivors of metastatic TGCT have increased mortality rates when compared to the general male population. A study from Norway reported that most of the excess deaths occurred within a decade from diagnosis due to the testicular cancer, but during a longer follow-up, additional excess deaths appeared in patients treated with radiotherapy or cisplatin-based chemotherapy, mainly due to non-TGCT second cancers; primarily gastric, pancreatic or bladder tumors (253, 254). A population-based large study from the US found an increased risk of leukemia after chemotherapy and a marked excess of solid tumors in patients treated with radiotherapy, which can appear after long follow-up (255). This study confirmed previously published worrying reports from several centers, which called for reducing the use of radiotherapy and radiologic imaging procedures during the follow-up (256).

Of importance for the testicular cancer survivors are also quality of life issues related to prolonged anxiety, depression and psychological stress, and loss in socioeconomic status or unemployment, leading to excess of suicide among the patients (167, 252, 254, 257, 258, 255, 259, 260). Hence, a psychologist/social advisor should be added to the multidisciplinary team of experts caring for survivors of TGCT. Clearly, the focus of the current comprehensive care has moved from survival to survivorship of the patients after their recovery from testicular cancer.

CONCLUSION

Key take home points are shown in figure 7.

Figure 7. . Key take home points.

Figure 7.

Key take home points.

ACKNOWLEDGEMENTS

The authors thank Prof. Niels E. Skakkebæk for his mentorship, and the research and clinical teams at their departments for the contribution to the studies summarized in this review. The work was supported by grants from numerous foundations, with the biggest contributions from the Danish Cancer Society, the Lundbeck Foundation, the Svend Andersen Foundation, the Danish Advanced Technology Foundation, Sigrid Juselius Foundation and Novo Nordisk Foundation.

REFERENCES

1.
Rajpert-De Meyts E, McGlynn KA, Okamoto K, Jewett MAS, Bokemeyer C. Testicular Germ Cell Tumours. Lancet. 2016;387(10029):1762–1774. doi.org. [PubMed: 26651223] [CrossRef]
2.
Moch H, Humphrey PA, Ulbright TM, Reuter VE, (Eds). WHO Classification of Tumours of the Urinary System and Male Genital Organs, 4th Edition. IARC Press, Lyon 2016.
3.
Skakkebaek NE, Rajpert-De Meyts E, Main KM. Testicular Dysgenesis Syndrome: An increasingly common developmental disorder with environmental aspects. Hum Reprod. 2001;16:972–978. [PubMed: 11331648]
4.
Rajpert-De Meyts E. Developmental model for the pathogenesis of testicular carcinoma in situ: Environmental and genetic aspects. Hum Reprod Update. 2006;12:303–323. [PubMed: 16540528]
5.
Skakkebæk NE, Rajpert-De Meyts E, Buck Louis GM, Toppari J, Andersson AM, Eisenberg ML, Jensen TK, Jørgensen N, Swan SH, Sapra KJ, Ziebe S, Priskorn L, Juul A. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol Rev. 2016;96:55–97. [PMC free article: PMC4698396] [PubMed: 26582516]
6.
Skotheim RI, Lind GE, Monni O, Nesland JM, Abeler VM, Fosså SD, Duale N, Brunborg G, Kallioniemi O, Andrews PW, Lothe RA. Differentiation of human embryonal carcinomas in vitro and in vivo reveals expression profiles relevant to normal development. Cancer Res. 2005;65(13):5588–98. [PubMed: 15994931]
7.
Solter D. From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research. Nature Rev Genetics. 2006;7:319–327. [PubMed: 16534514]
8.
Oosterhuis JW, Looijenga LHJ. Human germ cell tumours from a developmental perspective. Nat Rev Cancer. 2019;19:522–537. [PubMed: 31413324]
9.
Busch J, Seidel C, Zengerling F. Male Extragonadal Germ Cell Tumors of the Adult. Oncol Res Treat. 2016;39(3):140–4. Review. doi. [PubMed: 27032104] [CrossRef]
10.
Aguirre D, Nieto K, Lazos M, Peña YR, Palma I, Kofman-Alfaro S, Queipo G. Extragonadal germ cell tumors are often associated with Klinefelter syndrome. Hum Pathol. 2006;37:477–480. [PubMed: 16564924]
11.
Fosså SD, Aass N, Heilo A, Daugaard G, Skakkebaek NE, Stenwig AE, Nesland JM, Looijenga LH, Oosterhuis JW. Testicular carcinoma in situ in patients with extragonadal germ-cell tumours: the clinical role of pretreatment biopsy. Ann Oncol. 2003;14(9):1412–1418. [PubMed: 12954581]
12.
Scholz M, Zehender M, Thalmann GN, Borner M, Thöni H, Studer UE. Extragonadal retroperitoneal germ cell tumor: evidence of origin in the testis. Ann Oncol. 2002;13(1):121–124. [PubMed: 11863093]
13.
Berney DM, Looijenga LH, Idrees M, Oosterhuis JW, Rajpert-De Meyts E, Ulbright TM, Skakkebaek NE. Germ cell neoplasia in situ (GCNIS): evolution of the current nomenclature for testicular pre-invasive germ cell malignancy. Histopathology. 2016;69(1):7–10. [PubMed: 26918959] [CrossRef]
14.
Berney DM, Cree I, Rao V, Moch H, Srigley JR, Tsuzuki T, Amin MB, Comperat EM, Hartmann A, Menon S, Netto GJ, Rubin MA, Turajlic S, Raspollini MR, Tickoo SK. An Introduction to the WHO 5th Edition 2022 Classification of Testicular tumours. Histopathology. 2022 May 3. doi: 10.1111/his.14675. Epub ahead of print. PMID: 35502823. [PMC free article: PMC9544657] [PubMed: 35502823] [CrossRef]
15.
Skakkebaek NE. Possible carcinoma-in-situ of the testis. Lancet. 1972;ii:516–517. [PubMed: 4115573]
16.
Skakkebaek NE, Berthelsen JG, Giwercman A, Müller J. Carcinoma-in-situ of the testis. Possible origin from gonocytes and precursor of all types of germ cell tumours except spermatocytoma. Int J Androl. 1987;10:19–28. [PubMed: 3034791]
17.
Sonne SB, Almstrup K, Dalgaard M, Juncker AS, Edsgard D, Ruban L, Harrison NJ, Schwager C, Abdollahi A, Huber PE, Brunak S, Gjerdrum LM, Moore HD, Andrews PW, Skakkebaek NE, Rajpert-De Meyts E, Leffers H. Analysis of gene expression profiles of microdissected cell populations indicates that testicular carcinoma in situ is an arrested gonocyte. Cancer Res. 2009;69:5241–5250. [PMC free article: PMC2869030] [PubMed: 19491264]
18.
Jørgensen A, Lindhardt Johansen M, Juul A, Skakkebaek NE, Main KM, Rajpert-De Meyts E. Pathogenesis of germ cell neoplasia in testicular dysgenesis and disorders of sex development. Semin Cell Dev Biol. 2015;45:124–137. dx.doi.org. [PubMed: 26410164] [CrossRef]
19.
Cools M, Pleskacova J, Stoop H, Hoebeke P, Van Laecke E, Drop SL, Lebl J, Oosterhuis JW, Looijenga LH, Wolffenbuttel KP., Mosaicism Collaborative Group. Gonadal pathology and tumor risk in relation to clinical characteristics in patients with 45,X/46,XY mosaicism. J Clin Endocrinol Metab. 2011;96:E1171–1180. [PubMed: 21508138]
20.
Matsumoto F, Matsuyama S, Matsui F, Yazawa K, Matsuoka K. Variation of gonadal dysgenesis and tumor risk in patients with 45,X/46,XY mosaicism. Urology. 2020;137:157–160. [PubMed: 31883875]
21.
Jørgensen N, Müller J, Jaubert F, Clausen OP, Skakkebæk NE. Heterogeneity of gonadoblastoma germ cells: similarities with immature germ cells, spermatogonia and testicular carcinoma in situ cells. Histopathology. 1997;30:177–186. [PubMed: 9190360]
22.
Hersmus R, Kalfa N, de Leeuw B, Stoop H, Oosterhuis JW, de Krijger R, Wolffenbuttel KP, Drop SL, Veitia RA, Fellous M, Jaubert F, Looijenga LH. FOXL2 and SOX9 as parameters of female and male gonadal differentiation in patients with various forms of disorders of sex development (DSD). J Pathol. 2008;215:31–38. [PubMed: 18348162]
23.
Guminska A, Oszukowska E, Kuzanski W, Sosnowski M, Wolski JK, Walczak-Jedrzejowska R, Marchlewska K, Niedzielski J, Kula K, Slowikowska-Hilczer J. Less advanced testicular dysgenesis is associated by a higher prevalence of germ cell neoplasia. Int J Androl. 2010;33:e153–e62. [PubMed: 19719533]
24.
Chemes HE, Venara M, Del Rey G, Arcari AJ, Musse MP, Papazian R, Forclaz V, Gottlieb S. Is a CIS phenotype apparent in children with Disorders of Sex Development? Milder testicular dysgenesis is associated with a higher risk of malignancy. Andrology. 2015;3(1):59–69. doiPMID. [PubMed: 25598272] [CrossRef]
25.
Jørgensen N, Rajpert-De Meyts E, Græm N, Müller J, Giwercman A, Skakkebaek NE. Expression of immunohistochemical markers for testicular carcinoma in situ by normal fetal human germ cells. Lab Invest. 1995;72:223–231. [PubMed: 7531795]
26.
Rajpert-De Meyts E, Nielsen JE, Skakkebæk NE, Almstrup K. Diagnostic markers for germ cell neoplasms: from placental-like alkaline phosphatase to micro-RNAs. Folia Histochem Cytobiol. 2015;53(3):177–188. [PubMed: 26306513] [CrossRef]
27.
Almstrup K, Hoei-Hansen CE, Wirkner U, Blake J, Schwager C, Ansorge W, Nielsen JE, Skakkebaek NE, Rajpert-De Meyts E, Leffers H. Embryonic stem cell-like features of testicular carcinoma in situ revealed by genome-wide gene expression profiling. Cancer Res. 2004;64:4736–4743. [PubMed: 15256440]
28.
Alagaratnam S, Lind GE, Kraggerud SM, Lothe RA, Skotheim RI. The testicular germ cell tumour transcriptome. Int J Androl. 2011;34:e133–e150. [PubMed: 21651573]
29.
Looijenga LH, Stoop H, de Leeuw HP, de Gouveia Brazao CA, Gillis AJ, van Roozendaal KE, van Zoelen EJ, Weber RF, Wolffenbuttel KP, van Dekken H, Honecker F, Bokemeyer C, Perlman EJ, Schneider DT, Kononen J, Sauter G, Oosterhuis JW. POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors. Cancer Res. 2003;63:2244–2250. [PubMed: 12727846]
30.
Hoei-Hansen CE, Nielsen JE, Almstrup K, Sonne SB, Graem N, Skakkebaek NE, Leffers H, Rajpert-De Meyts E. Transcription factor AP-2gamma is a developmentally regulated marker of testicular carcinoma in situ and germ cell tumors. Clin Cancer Res. 2004;10:8521–8530. [PubMed: 15623634]
31.
Hoei-Hansen CE, Almstrup K, Nielsen JE, Brask Sonne S, Graem N, Skakkebaek NE, Leffers H, Rajpert-De Meyts E. Stem cell pluripotency factor NANOG is expressed in human fetal gonocytes, testicular carcinoma in situ and germ cell tumours. Histopathology. 2005;47:48–56. [PubMed: 15982323]
32.
Hart AH, Hartley L, Parker K, Ibrahim M, Looijenga LH, Pauchnik M, Chow CW, Robb L. The pluripotency homeobox gene NANOG is expressed in human germ cell tumors. Cancer. 2005;104:2092–2098. [PubMed: 16206293]
33.
Rajpert-De Meyts E, Skakkebæk NE. Expression of the c-kit protein product in carcinoma in situ and invasive germ cell tumours. Int J Androl. 1994;17:85–92. [PubMed: 7517917]
34.
Bártková J, Bártek J, Lukás J, Vojtĕsek B, Stasková Z, Rejthar A, Kovarík J, Midgley CA, Lane DP. p53 protein alterations in human testicular cancer including pre-invasive intratubular germ-cell neoplasia. Int J Cancer. 1991;49:196–202. [PubMed: 1652567]
35.
West JA, Viswanathan SR, Yabuuchi A, Cunniff K, Takeuchi A, Park IH, Sero JE, Zhu H, Perez-Atayde A, Frazier AL, Surani MA, Daley GQ. A role for Lin28 in primordial germ-cell development and germ-cell malignancy. Nature. 2009;460:909–913. [PMC free article: PMC2729657] [PubMed: 19578360]
36.
Gillis AJ, Stoop H, Biermann K, van Gurp RJ, Swartzman E, Cribbes S, Ferlinz A, Shannon M, Oosterhuis JW, Looijenga LH. Expression and interdependencies of pluripotency factors LIN28, OCT3/4, NANOG and SOX2 in human testicular germ cells and tumours of the testis. Int J Androl. 2011;34:e160–e174. [PubMed: 21631526]
37.
Novotny GW, Belling KC, Bramsen JB, Nielsen JE, Bork-Jensen J, Almstrup K, Sonne SB, Kjems J, Rajpert-De Meyts E, Leffers H. MicroRNA expression profiling of carcinoma in situ cells of the testis. Endocr Relat Cancer. 2012;19:365–379. [PubMed: 22420006]
38.
Almstrup K, Lobo J, Mørup N, Belge G, Rajpert-De Meyts E, Looijenga LHJ, Dieckmann KP. Application of miRNAs in the diagnosis and monitoring of testicular germ cell tumours. Nat Rev Urol. 2020;17:201–213. [PubMed: 32157202]
39.
Kraggerud SM, Hoei-Hansen CE, Alagaratnam S, Skotheim RI, Abeler VM, Rajpert-De Meyts E, Lothe RA. Molecular characteristics of malignant ovarian germ cell tumours and comparison to testicular counterparts: Implications for pathogenesis. Endocrine Reviews. 2013;34:339–376. [PMC free article: PMC3787935] [PubMed: 23575763]
40.
Palmer RD, Barbosa-Morais NL, Gooding EL, Muralidhar B, Thornton CM, Pett MR, Roberts I, Schneider DT, Thorne N, Tavaré S, Nicholson JC, Coleman N., Children's Cancer and Leukaemia Group. Pediatric malignant germ cell tumors show characteristic transcriptome profiles. Cancer Res. 2008;68:4239–4247. [PubMed: 18519683]
41.
Palmer RD, Murray MJ, Saini HK, van Dongen S, Abreu-Goodger C, Muralidhar B, Pett MR, Thornton CM, Nicholson JC, Enright AJ, Coleman N., Children's Cancer and Leukaemia Group. Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Res. 2010;70:2911–2923. [PMC free article: PMC3000593] [PubMed: 20332240]
42.
Manivel JC, Simonton S, Wold LE, Dehner LP. Absence of intratubular germ cell neoplasia in testicular yolk sac tumors in children. A histochemical and immunohistochemical study. Arch Pathol Lab Med. 1988;112:641–645. [PubMed: 2837162]
43.
Mosbech CH, Rechnitzer C, Brok JS, Rajpert-De Meyts E, Hoei-Hansen CE. Recent advances in understanding the etiology and pathogenesis of pediatric germ cell tumors. J Ped Hematol Oncol. 2014;36:263–270. [PubMed: 24577549]
44.
Grogg JB, Schneider K, Bode PK, Wettstein MS, Kranzbühler B, Eberli D, Sulser T, Beyer J, Hermanns T, Fankhauser CD. A systematic review of treatment outcomes in localised and metastatic spermatocytic tumors of the testis. J Cancer Res Clin Oncol. 2019;145(12):3037–3045. [PubMed: 31646373]
45.
Müller J, Skakkebaek NE, Parkinson MC. The spermatocytic seminoma: views on pathogenesis. Int J Androl. 1987;10:147–156. [PubMed: 3583416]
46.
Looijenga LH, Hersmus R, Gillis AJ, Pfundt R, Stoop HJ, van Gurp RJ, Veltman J, Beverloo HB, van Drunen E, van Kessel AG, Pera RR, Schneider DT, Summersgill B, Shipley J, McIntyre A, van der Spek P, Schoenmakers E, Oosterhuis JW. Genomic and expression profiling of human spermatocytic seminomas: primary spermatocyte as tumorigenic precursor and DMRT1 as candidate chromosome 9 gene. Cancer Res. 2006;66:290–302. [PubMed: 16397242]
47.
Rajpert-De Meyts E, Jacobsen GK, Bartkova J, Aubry F, Samson M, Bartek J, Skakkebaek NE. The imunohistochemical expression pattern of Chk2, p53, p19-INK4d, MAGE-A4 and other selected antigens provides new evidence for the pre-meiotic origin of spermatocytic seminoma. Histopathology. 2003;42:217–226. [PubMed: 12605640]
48.
Lim J, Goriely A, Turner GD, Ewen KA, Jacobsen GK, Graem N, Wilkie AO, Rajpert-De Meyts E. OCT2, SSX and SAGE1 reveal the phenotypic heterogeneity of spermatocytic seminoma reflecting distinct subpopulations of spermatogonia. J Pathol. 2011;224:473–483. [PMC free article: PMC3210831] [PubMed: 21706474]
49.
Goriely A, Hansen RM, Taylor IB, Olesen IA, Jacobsen GK, McGowan SJ, Pfeifer SP, McVean GA, Rajpert-De Meyts E, Wilkie AO. Activating mutations in FGFR3 and HRAS reveal a shared genetic origin for congenital disorders and testicular tumors. Nat Genet. 2009;41:1247–1252. [PMC free article: PMC2817493] [PubMed: 19855393]
50.
Giannoulatou E, Maher GJ, Ding Z, Gillis AJM, Dorssers LCJ, Hoischen A, Rajpert-De Meyts E., WGS500 Consortium. McVean GAT, Wilkie AOM, Looijenga LHJ, Goriely A. Whole-genome sequencing of spermatocytic tumors provides insights into the mutational processes operating in the male germline. PLoS One. 2017;12(5):e0178169. doi.org. [PMC free article: PMC5439955] [PubMed: 28542371] [CrossRef]
51.
Goriely A, Wilkie AO. Paternal age effect mutations and selfish spermatogonial selection: causes and consequences for human disease. Am J Hum Genet. 2012;90:175–200. [PMC free article: PMC3276674] [PubMed: 22325359]
52.
Znaor A, Lortet-Tieulent J, Jemal A, Bray F. International variations and trends in testicular cancer incidence and mortality. Eur Urol. 2014 Jun;65(6):1095–106. [PubMed: 24268506]
53.
Znaor A, Skakkebaek NE, Rajpert-De Meyts E, Kuliš T, Laversanne M, Gurney J, Sarfati D, McGlynn KA, Bray F. Global patterns in testicular cancer incidence and mortality in 2020. Int J Cancer. 2022. doiEpub ahead of printPMID. [PubMed: 35277970] [CrossRef]
54.
Serrano T, Chevrier C, Multigner L, Cordier S, Jégou B. International geographic correlation study of the prevalence of disorders of male reproductive health. Hum Reprod. 2013;28:1974–1986. [PubMed: 23670171]
55.
Gurney JK. The puzzling incidence of testicular cancer in New Zealand: what can we learn? Andrology. 2019;7(4):394–401. [PubMed: 30663250]
56.
Jørgensen N, Vierula M, Jacobsen R, Pukkala E, Perheentupa A, Virtanen HE, Skakkebaek NE, Toppari J. Recent adverse trends in semen quality and testis cancer incidence among Finnish men. Int J Androl. 2011;34(4 Pt 2):e37–e48. [PMC free article: PMC3170483] [PubMed: 21366607]
57.
Ghazarian AA, Trabert B, Devesa SS, McGlynn KA. Recent trends in the incidence of testicular germ cell tumors in the United States. Andrology. 2015;3(1):13–8. [PMC free article: PMC4342271] [PubMed: 25331158]
58.
Møller H. Clues to the aetiology of testicular germ cell tumours from descriptive epidemiology. Eur Urol. 1993;23:8–15. [PubMed: 8386660]
59.
Lindhardt Johansen M, Hagen CP, Rajpert-De Meyts E, Kjærgaard S, Petersen BL, Skakkebæk NE, Main KM, Juul A. 45,X/46,XY mosaicism: phenotypic characteristics, growth, and reproductive function--a retrospective longitudinal study. J Clin Endocrinol Metab. 2012;97:E1540–1549. [PubMed: 22605431]
60.
Slowikowska-Hilczer J, Szarras-Czapnik M, Duranteau L, Rapp M, Walczak-Jedrzejowska R, Marchlewska K, Oszukowska E, Nordenstrom A. dsd-LIFE group. Risk of gonadal neoplasia in patients with disorders/differences of sex development. Cancer Epidemiol. 2020;69:101800. doi. [PubMed: 32905884] [CrossRef]
61.
Giwercman A, Grinsted J, Hansen B, Jensen OM, Skakkebaek NE. Testicular cancer risk in boys with maldescended testis: a cohort study. J Urol. 1987;138:1214–1216. [PubMed: 2889833]
62.
Piltoft JS, Larsen SB, Dalton SO, Johansen C, Baker JL, Cederkvist L, Andersen I. Early life risk factors for testicular cancer: a case-cohort study based on the Copenhagen School Health Records Register. Acta Oncol. 2017;56(2):220–224. [PubMed: 28080187]
63.
Dieckmann KP, Rube C, Henke RP. Association of Down's syndrome and testicular cancer. J Urol. 1997;157:1701–1704. [PubMed: 9112509]
64.
Cook MB, Akre O, Forman D, Madigan MP, Richiardi L, McGlynn KA. A systematic review and meta-analysis of perinatal variables in relation to the risk of testicular cancer--experiences of the son. Int J Epidemiol. 2010;39:1605–1618. [PMC free article: PMC2992627] [PubMed: 20660640]
65.
Maule M, Malavassi JL, Richiardi L. Age at puberty and risk of testicular cancer: a meta-analysis. Int J Androl. 2012 Dec;35(6):828–34. doiPMID. [PubMed: 22713104] [CrossRef]
66.
McGlynn KA, Petrick JL, Gamborg M, Aarestrup J, Baker JL. Childhood height and risk of testicular germ cell tumors in adulthood. Int J Cancer. 2018;143(4):767–772. [PMC free article: PMC6041140] [PubMed: 29508384]
67.
Møller H, Skakkebaek NE. Risk of testicular cancer in subfertile men: case-control study. Br Med J. 1999;318:559–562. [PMC free article: PMC27753] [PubMed: 10037628]
68.
Jacobsen R, Bostofte E, Engholm G, Hansen J, Olsen J H, Skakkebaek NE, Møller H. Risk of testicular cancer in men with abnormal semen characteristics: cohort study. Br Med J. 2000;321:789–792. [PMC free article: PMC27489] [PubMed: 11009515]
69.
Hanson HA, Anderson RE, Aston KI, Carrell DT, Smith KR, Hotaling JM. Subfertility increases risk of testicular cancer: evidence from population-based semen samples. Fertil Steril. 2016;105(2):322–8.e1. [PMC free article: PMC4744156] [PubMed: 26604070]
70.
Fosså SD, Chen J, Schonfeld SJ, McGlynn KA, McMaster ML, Gail MH, Travis LB. Risk of contralateral testicular cancer: a population-based study of 29,515 U.S. men. J Natl Cancer Inst. 2005;97:1056–1066. [PubMed: 16030303]
71.
Hoei-Hansen CE, Holm M, Rajpert-De Meyts E, Skakkebaek NE. Histological evidence of testicular dysgenesis in contralateral biopsies of 218 patients with testicular germ cell cancer. J Pathol. 2003;200:370–374. [PubMed: 12845633]
72.
Zequi Sde C, da Costa WH, Santana TB, Favaretto RL, Sacomani CA, Guimaraes GC. Bilateral testicular germ cell tumours: a systematic review. BJU Int. 2012;110:1102–1109. [PubMed: 22429248]
73.
Watson RA, Morgan RD, Joseph J, Sivakumar S, Verrill C, Tuthill M, Sullivan M, Protheroe AS. Bilateral Testicular Germ Cell Tumors: A Case-Series From a UK-Based Tertiary Referral Center Over 19 Years. Clin Genitourinary Cancer. 2018;16:e513–e516. [PubMed: 29566999]
74.
Dieckmann KP, Anheuser P, Sattler F, Von Kügelgen T, Matthies C, Ruf C. Sequential bilateral testicular tumours presenting with intervals of 20 years and more. BMC Urol. 2013;13:71. (doi. [PMC free article: PMC4028980] [PubMed: 24321309] [CrossRef]
75.
Harland SJ, Cook PA, Fosså SD, Horwich A, Mead GM, Parkinson MC, Roberts JT, Stenning SP. Intratubular germ cell neoplasia of the contralateral testis in testicular cancer: defining a high risk group. J Urol. 1998;160:1353–1357. [PubMed: 9751353]
76.
Rud CN, Daugaard G, Rajpert-De Meyts E, Skakkebaek NE, Petersen JH, Jørgensen N. Sperm concentration, testicular volume and age predict risk of carcinoma-in-situ in contralateral testis of men with testicular germ cell cancer. J Urol. 2013;190:2074–2080. [PubMed: 23770148]
77.
Jørgensen N, Rajpert-De Meyts E, Main KM, Skakkebæk NE. Testicular dysgenesis syndrome comprises some but not all cases of hypospadias and impaired spermatogenesis. Int J Androl. 2010;33:298–303. [PubMed: 20132348]
78.
Atkin NB, Baker MC. Specific chromosome change, i(12p), in testicular tumours? Lancet. 1982 Dec 11;2(8311):1349. [PubMed: 6128640]
79.
Taylor-Weiner A, Zack T, O'Donnell E, Guerriero JL, Bernard B, Reddy A, Han GC, AlDubayan S, Amin-Mansour A, Schumacher SE, Litchfield K, Turnbull C, Gabriel S, Beroukhim R, Getz G, Carter SL, Hirsch MS, Letai A, Sweeney C, Van Allen EM. Genomic evolution and chemoresistance in germ-cell tumours. Nature. 2016;540(7631):114–118. PMID. [PMC free article: PMC5553306] [PubMed: 27905446]
80.
Hoff AM, Alagaratnam S, Zhao S, Bruun J, Andrews PW, Lothe RA, Skotheim RI. Identification of Novel Fusion Genes in Testicular Germ Cell Tumors. Cancer Res. 2016;76(1):108–16. doiPMID. [PMC free article: PMC4704135] [PubMed: 26659575] [CrossRef]
81.
Tian Q. Frierson HFJr, Krystal GW, Moskaluk CA. Activating c-kit gene mutations in human germ cell tumors. Am J Pathol. 1999;154:1643–164. [PMC free article: PMC1876773] [PubMed: 10362788]
82.
McIntyre A, Summersgill B, Grygalewicz B, Gillis AJ, Stoop J, van Gurp RJ, Dennis N, Fisher C, Huddart R, Cooper C, Clark J, Oosterhuis JW, Looijenga LH, Shipley J. Amplification and overexpression of the KIT gene is associated with progression in the seminoma subtype of testicular germ cell tumors of adolescents and adults. Cancer Res. 2005;65:8085–8089. [PubMed: 16166280]
83.
Shen H, Shih J, Hollern DP, Wang L, Bowlby R, Tickoo SK, Thorsson V, Mungall AJ, Newton Y, Hegde AM, Armenia J, Sánchez-Vega F, Pluta J, Pyle LC, Mehra R, Reuter VE, Godoy G, Jones J, Shelley CS, Feldman DR, Vidal DO, Lessel D, Kulis T, Cárcano FM, Leraas KM, Lichtenberg TM, Brooks D, Cherniack AD, Cho J, Heiman DI, Kasaian K, Liu M, Noble MS, Xi L, Zhang H, Zhou W. ZenKlusen JC, Hutter CM, Felau I, Zhang J, Schultz N, Getz G, Meyerson M, Stuart JM; Cancer Genome Atlas Research Network, Akbani R, Wheeler DA, Laird PW, Nathanson KL, Cortessis VK, Hoadley KA. Integrated Molecular Characterization of Testicular Germ Cell Tumors. Cell Rep. 2018;23(11):3392–3406. [PMC free article: PMC6075738] [PubMed: 29898407]
84.
Hemminki K, Li X. Familial risk in testicular cancer as a clue to a heritable and environmental aetiology. Brit J Cancer. 2004;90:1765–1770. [PMC free article: PMC2410275] [PubMed: 15208620]
85.
Mucci LA, Hjelmborg JB, Harris JR, Czene K, Havelick DJ, Scheike T, Graff RE, Holst K, Möller S, Unger RH, McIntosh C, Nuttall E, Brandt I, Penney KL, Hartman M, Kraft P, Parmigiani G, Christensen K, Koskenvuo M, Holm NV, Heikkilä K, Pukkala E, Skytthe A, Adami HO, Kaprio J., Nordic Twin Study of Cancer. (NorTwinCan) Collaboration. Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA. 2016;315(1):68–76. Erratum in: JAMA 315(8):822, 2016. [PMC free article: PMC5498110] [PubMed: 26746459]
86.
Litchfield K, Loveday C, Levy M, Dudakia D, Rapley E, Nsengimana J, Bishop DT, Reid A, Huddart R, Broderick P, Houlston RS, Turnbull C. Large-scale Sequencing of Testicular Germ Cell Tumour (TGCT) Cases Excludes Major TGCT Predisposition Gene. Eur Urol. 2018;73(6):828–831. [PubMed: 29433971]
87.
Pluta J, Pyle LC, Nead KT, Wilf R, Li M, Mitra N, Weathers B, D'Andrea K, Almstrup K, Anson-Cartwright L, Benitez J, Brown CD, Chanock S, Chen C, Cortessis VK, Ferlin A, Foresta C, Gamulin M, Gietema JA, Grasso C, Greene MH, Grotmol T, Hamilton RJ, Haugen TB, Hauser R, Hildebrandt MAT, Johnson ME, Karlsson R, Kiemeney LA, Lessel D, Lothe RA, Loud JT, Loveday C, Martin-Gimeno P, Meijer C, Nsengimana J, Quinn DI, Rafnar T, Ramdas S, Richiardi L, Skotheim RI, Stefansson K, Turnbull C, Vaughn DJ, Wiklund F, Wu X, Yang D, Zheng T, Wells AD, Grant SFA, Rajpert-De Meyts E, Schwartz SM, Bishop DT, McGlynn KA, Kanetsky PA, Nathanson KL., Testicular Cancer Consortium. Identification of 22 susceptibility loci associated with testicular germ cell tumors. Nat Commun. 2021;12(1):4487. doi. [PMC free article: PMC8302763] [PubMed: 34301922] [CrossRef]
88.
Rapley EA, Turnbull C, Al Olama AA, Dermitzakis ET, Linger R, Huddart RA, Renwick A, Hughes D, Hines S, Seal S, Morrison J, Nsengimana J, Deloukas P., UK Testicular Cancer Collaboration. Rahman N, Bishop DT, Easton DF, Stratton MR. A genome-wide association study of testicular germ cell tumor. Nat Genet. 2009;41:807–810. [PMC free article: PMC2871592] [PubMed: 19483681]
89.
Kanetsky PA, Mitra N, Vardhanabhuti S, Li M, Vaughn DJ, Letrero R, Ciosek SL, Doody DR, Smith LM, Weaver J, Albano A, Chen C, Starr JR, Rader DJ, Godwin AK, Reilly MP, Hakonarson H, Schwartz SM, Nathanson KL. Common variation in KITLG and at 5q31.3 predisposes to testicular germ cell cancer. Nat Genet. 2009;41:811–815. [PMC free article: PMC2865677] [PubMed: 19483682]
90.
Turnbull C, Rapley EA, Seal S, Pernet D, Renwick A, Hughes D, Ricketts M, Linger R, Nsengimana J, Deloukas P, Huddart RA, Bishop DT, Easton DF, Stratton MR, Rahman N., UK Testicular Cancer Collaboration. Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer. Nat Genet. 2010;42:604–607. [PMC free article: PMC3773909] [PubMed: 20543847]
91.
Dalgaard MD, Weinhold N, Edsgärd D, Silver JD, Pers TH, Nielsen JE, Jørgensen N, Juul A, Gerds TA, Giwercman A, Giwercman YL, Cohn-Cedermark G, Virtanen HE, Toppari J, Daugaard G, Jensen TS, Brunak S, Rajpert-De Meyts E, Skakkebæk NE, Leffers H, Gupta R. A genome-wide association study of men with symptoms of testicular dysgenesis syndrome and its network biology interpretation. J Med Genet. 2012;49:58–65. [PMC free article: PMC3284313] [PubMed: 22140272]
92.
Litchfield K, Levy M, Orlando G, Loveday C, Law PJ, Migliorini G, Holroyd A, Broderick P, Karlsson R, Haugen TB, Kristiansen W, Nsengimana J, Fenwick K, Assiotis I, Kote-Jarai Z, Dunning AM, Muir K, Peto J, Eeles R, Easton DF, Dudakia D, Orr N, Pashayan N., UK Testicular Cancer Collaboration. PRACTICAL Consortium, Bishop DT, Reid A, Huddart RA, Shipley J, Grotmol T, Wiklund F, Houlston RS, Turnbull C. Identification of 19 new risk loci and potential regulatory mechanisms influencing susceptibility to testicular germ cell tumor. Nat Genet. 2017;49(7):1133–1140. [PMC free article: PMC6016736] [PubMed: 28604728]
93.
Wang Z, McGlynn KA, Rajpert-De Meyts E, Bishop DT, Chung CC, Dalgaard MD, Greene MH, Gupta R, Grotmol T, Haugen TB, Karlsson R, Litchfield K, Mitra N, Nielsen K, Pyle LC, Schwartz SM, Thorsson V, Vardhanabhuti S, Wiklund F, Turnbull C, Chanock SJ, Kanetsky PA, Nathanson KL., Testicular Cancer Consortium. Meta-analysis of five genome-wide association studies identifies multiple new loci associated with testicular germ cell tumor. Nat Genet. 2017;49(7):1141–1147. [PMC free article: PMC5490654] [PubMed: 28604732]
94.
Dolci S, Williams DE, Ernst MK, Resnick JL, Brannan CI, Lock LF, Lyman SD, Boswell HS, Donovan PJ. Requirement for mast cell growth factor for primordial germ cell survival in culture. Nature. 1991;352:809–811. [PubMed: 1715518]
95.
Gu Y, Runyan C, Shoemaker A, Surani A, Wylie C. Steel factor controls primordial germ cell survival and motility from the time of their specification in the allantois, and provides a continuous niche throughout their migration. Development. 2009;136:1295–1303. [PubMed: 19279135]
96.
Matson CK, Murphy MW, Griswold MD, Yoshida S, Bardwell VJ, Zarkower D. The mammalian doublesex homolog DMRT1 is a transcriptional gatekeeper that controls the mitosis versus meiosis decision in male germ cells. Dev Cell. 2010;19(4):612–24. [PMC free article: PMC2996490] [PubMed: 20951351]
97.
Jørgensen A, Nielsen JE, Jensen MB, Græm N, Rajpert-De Meyts E. Analysis of meiosis regulators in human gonads: a sexually dimorphic spatio-temporal expression pattern suggests involvement of DMRT1 in meiotic entry. Mol Hum Reprod. 2012;18:523–534. [PubMed: 22899867]
98.
Jørgensen A, Nielsen JE, Almstrup K, Toft BG, Petersen BL, Rajpert-De Meyts E. Dysregulation of the mitosis-meiosis switch in testicular carcinoma in situ. J Pathol. 2013;229(4):588–598. [PubMed: 23303528]
99.
Eini R, Dorssers LC, Looijenga LH. Role of stem cell proteins and microRNAs in embryogenesis and germ cell cancer. Int J Dev Biol. 2013;57:319–332. [PubMed: 23784843]
100.
Almstrup K, Nielsen JE, Mlynarska O, Jansen MT, Jørgensen A, Skakkebæk NE, Rajpert-De Meyts E. Carcinoma in situ testis displays permissive chromatin modifications similar to immature foetal germ cells. Br J Cancer. 2010;103:1269–1276. [PMC free article: PMC2967056] [PubMed: 20823885]
101.
Spiller CM, Bowles J, Koopman P. Nodal/Cripto signaling in fetal male germ cell development: implications for testicular germ cell tumors. Int J Dev Biol. 2013;57:211–219. [PubMed: 23784832]
102.
Jørgensen A, Macdonald J, Nielsen JE, Kilcoyne KR, Perlman S, Lundvall L, Langhoff Thuesen L, Juul Hare K, Frederiksen H, Andersson AM, Skakkebæk NE, Juul A, Sharpe RM, Rajpert-De Meyts E, Mitchell RT. Nodal Signaling Regulates Germ Cell Development and Establishment of Seminiferous Cords in the Human Fetal Testis. Cell Rep. 2018;25(7):1924–1937. [PubMed: 30428358]
103.
Young JC, Kerr G, Micati D, Nielsen JE, Rajpert-De Meyts E, Abud HE, Loveland KL. WNT signalling in the normal human adult testis and in male germ cell neoplasms. Hum Reprod. 2020;5(9):1991–2003. PMID. [PubMed: 32667987]
104.
Toppari J, Larsen JC, Christiansen P, Giwercman A, Grandjean P, Guillette LJ, Jegou B, Jensen TK, Jouannet P, Keiding N, Leffers H, McLachlan JA, Meyer O, Müller J. Rajpert-De Meyts E, Scheike T, Sharpe R, Sumpter J, Skakkebaek NE. Male reproductive health and environmental xenoestrogens. Environ Health Perspect. 1996;104 Suppl 4:741–803. [PMC free article: PMC1469672] [PubMed: 8880001]
105.
van den Driesche S, Kilcoyne K, Wagner I, Rebourcet D, Boyle A, Mitchell R, McKinnell C, Macpherson S, Donat R, Shukla CJ, Joergensen A, Rajpert-De Meyts E, Skakkebæk NE, Sharpe R. Experimentally induced testicular dysgenesis syndrome originates in the masculinization programming window. JCI Insight. 2017;2(6):e91204. [PMC free article: PMC5358493] [PubMed: 28352662] [CrossRef]
106.
Jørgensen A, Svingen T, Miles H, Chetty T, Stukenborg JB, Mitchell RT. Environmental Impacts on Male Reproductive Development: Lessons from Experimental Models. Horm Res Paediatr Oct 4:303-319, 2021. [PubMed: 34607330]
107.
Hardell L, Bavel B, Lindström G, Eriksson M, Carlberg M. In utero exposure to persistent organic pollutants in relation to testicular cancer risk. Int J Androl. 2006;29:228–234. [PubMed: 16371110]
108.
McGlynn KA, Quraishi SM, Graubard BI, Weber JP, Rubertone MV, Erickson RL. Persistent organochlorine pesticides and risk of testicular germ cell tumors. J Natl Cancer Inst. 2008;100:663–671. [PubMed: 18445826]
109.
Bräuner EV, Lim YH, Koch T, Uldbjerg CS, Gregersen LS, Pedersen MK, Frederiksen H, Petersen JH, Coull BA, Andersson AM, Hickey M, Skakkebæk NE, Hauser R, Juul A. Endocrine Disrupting Chemicals and Risk of Testicular Cancer: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab. 2021;106(12):e4834–e4860. [PMC free article: PMC8864757] [PubMed: 34270734]
110.
Rodprasert W, Toppari J, Virtanen HE. Endocrine Disrupting Chemicals and Reproductive Health in Boys and Men. Front Endocrinol (Lausanne). 2021;12:706532. doiPMID. [PMC free article: PMC8530230] [PubMed: 34690925] [CrossRef]
111.
Daling JR, Doody DR, Sun X, Trabert BL, Weiss NS, Chen C, Biggs ML, Starr JR, Dey SK, Schwartz SM. Association of marijuana use and the incidence of testicular germ cell tumors. Cancer. 2009;115:1215–1223. [PMC free article: PMC2759698] [PubMed: 19204904]
112.
Gurney J, Shaw C, Stanley J, Signal V, Sarfati D. Cannabis exposure and risk of testicular cancer: a systematic review and meta-analysis. BMC Cancer. 2015;15:897. doi. [PMC free article: PMC4642772] [PubMed: 26560314] [CrossRef]
113.
Holm M, Hoei-Hansen CE, Rajpert-De Meyts E, Skakkebaek NE. Increased risk of carcinoma in situ in patients with testicular germ cell cancer with ultrasonic microlithiasis in the contralateral testicle. J Urol. 2003;170:1163–1167. [PubMed: 14501716]
114.
van Casteren NJ, Looijenga LH, Dohle GR. Testicular microlithiasis and carcinoma in situ overview and proposed clinical guideline. Int J Androl. 2009;32:279–287. [PubMed: 19207616]
115.
Wang T, Liu L, Luo J, Liu T, Wei A. A Meta-Analysis of the Relationship between Testicular Microlithiasis and Incidence of Testicular Cancer. Urol J. 2015;12(2):2057–64. PMID. [PubMed: 25923148]
116.
Barbonetti A, Martorella A, Minaldi E, D'Andrea S, Bardhi D, Castellini C, Francavilla F, Francavilla S. Testicular Cancer in Infertile Men With and Without Testicular Microlithiasis: A Systematic Review and Meta-Analysis of Case-Control Studies. Front Endocrinol (Lausanne10:164. 2019. doi: 10.3389/fendo.2019.00164. PMID: 30949131. [PMC free article: PMC6437042] [PubMed: 30949131] [CrossRef]
117.
Dohle GR, Elzanaty S, van Casteren NJ. Testicular biopsy: clinical practice and interpretation. Asian J Androl. 2012;14(1):88–93. doiPMID. [PMC free article: PMC3735160] [PubMed: 22157985] [CrossRef]
118.
Kliesch S, Schmidt S, Wilborn D, Aigner C, Albrecht W, Bedke J, Beintker M, Beyersdorff D, Bokemeyer C, Busch J, Classen J, de Wit M, Dieckmann KP, Diemer T, Dieing A, Gockel M, Göckel-Beining B, Hakenberg OW, Heidenreich A, Heinzelbecker J, Herkommer K, Hermanns T, Kaufmann S, Kornmann M, Kotzerke J, Krege S, Kristiansen G, Lorch A, Müller AC, Oechsle K, Ohloff T, Oing C, Otto U, Pfister D, Pichler R, Recken H, Rick O, Rudolph Y, Ruf C, Schirren J, Schmelz H, Schmidberger H, Schrader M, Schweyer S, Seeling S, Souchon R, Winter C, Wittekind C, Zengerling F, Zermann DH, Zillmann R, Albers P. Management of Germ Cell Tumours of the Testis in Adult Patients. German Clinical Practice Guideline Part I: Epidemiology, Classification, Diagnosis, Prognosis, Fertility Preservation, and Treatment Recommendations for Localized Stages. Urol Int. 2021;105(3-4):169–180. [PMC free article: PMC8006579] [PubMed: 33412555] [CrossRef]
119.
McLachlan RI, Rajpert-De Meyts E, Hoei-Hansen CE, De Kretser DM, Skakkebaek NE. Histological evaluation of the human testis: approaches to optimizing the clinical value of the assessment: mini review. Hum Reprod. 2007;22:2–16. [PubMed: 16887924]
120.
van Casteren NJ, de Jong J, Stoop H, Steyerberg EW, de Bekker-Grob EW, Dohle GR, Oosterhuis JW, Looijenga LH. Evaluation of testicular biopsies for carcinoma in situ: immunohistochemistry is mandatory. Int J Androl. 2009;32:666–674. [PubMed: 18798762]
121.
Kier MGG, Lauritsen J, Almstrup K, Mortensen MS, Toft BG, Rajpert-De Meyts E, Skakkebæk NE, Rørth M, von der Maase H, Agerbæk M, Holm NV, Andersen KK, Dalton SO, Johansen C, Daugaard G. Screening for carcinoma in situ in the contralateral testicle in patients with testicular cancer: a population-based study. Ann Oncol. 2015;15(26):737–742. [PubMed: 25542924]
122.
Rajpert-De Meyts E, Jørgensen N, Petersen JH, Almstrup K, Aksglaede L, Lauritsen J, Rørth M, Daugaard G, Skakkebaek NE. Optimized detection of germ cell neoplasia in situ in contralateral biopsy reduces the risk of second testis cancer. BJU Int. 2022. doiEpub ahead of printPMID. [PMC free article: PMC9796833] [PubMed: 35575005] [CrossRef]
123.
Albers P, Albrecht W, Algaba F, Bokemeyer C, Cohn-Cedermark G, Fizazi K, Horwich A, Laguna MP, Nicolai N, Oldenburg J., European Association of Urology. Guidelines on testicular cancer: 2015 update. Eur Urol. 2015;68:1054–1068. [PubMed: 26297604]
124.
Gilligan T, Lin DW, Aggarwal R, Chism D, Cost N, Derweesh IH, Emamekhoo H, Feldman DR, Geynisman DM, Hancock SL, LaGrange C, Levine EG, Longo T, Lowrance W, McGregor B, Monk P, Picus J, Pierorazio P, Rais-Bahrami S, Saylor P, Sircar K, Smith DC, Tzou K, Vaena D, Vaughn D, Yamoah K, Yamzon J, Johnson-Chilla A, Keller J, Pluchino LA. Testicular Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17(12):1529–1554. doiPMID. [PubMed: 31805523] [CrossRef]
125.
Bagrodia A, Pierorazio P, Singla N, Albers P. The Complex and Nuanced Care for Early-stage Testicular Cancer: Lessons from the European Association of Urology and American Urological Association Testis Cancer Guidelines. Eur Urol. 2020;77(2):139–141. doiPMID. [PubMed: 31672465] [CrossRef]
126.
Dieckmann KP, Kulejewski M, Pichlmeier U, Loy V. Diagnosis of contralateral testicular intraepithelial neoplasia (TIN) in patients with testicular germ cell cancer: systematic two-site biopsies are more sensitive than a single random biopsy. Eur Urol. 2007;51:175–183. [PubMed: 16814456]
127.
Dieckmann KP, Heinemann V, Frey U, Pichlmeier U., German Testicular Cancer Study Group. How harmful is contralateral testicular biopsy? An analysis of serial imaging studies and a prospective evaluation of surgical complications. Eur Urol. 2005;48:662–672. [PubMed: 16009484]
128.
Dieckmann KP, Loy V. False-negative biopsies for the diagnosis of testicular intraepithelial neoplasia (TIN)--an update. Eur Urol. 2003;43:516–521. [PubMed: 12705996]
129.
Rajpert-De Meyts E, Grigor KM, Skakkebæk NE. Histopathological evaluation of testicular biopsy. In: Endocrinology of the Testis and Male Reproduction. M. Simoni, I. Huhtaniemi (Editors). Springer International Publishing, 2017, pp. 1-20, doi: 10.1007/978-3-319-29456-8_20-1. [CrossRef]
130.
Petersen PM, Skakkebaek NE, Vistisen K, Rørth M, Giwercman A. Semen quality and reproductive hormones before orchiectomy in men with testicular cancer. J Clin Oncol. 1999;17:941–947. [PubMed: 10071288]
131.
Del Giudice F, Kasman AM, De Berardinis E, Busetto GM, Belladelli F, Eisenberg ML. Association between male infertility and male-specific malignancies: systematic review and meta-analysis of population-based retrospective cohort studies. Fertil Steril. 2020;114(5):984–996. doiPMID. [PubMed: 32709378] [CrossRef]
132.
Giwercman A, Marks A, Skakkebaek NE. Carcinoma-in-situ germ-cells exfoliated from seminiferous epithelium into seminal fluid. Lancet. 1988;1(8584):530. [PubMed: 2893940]
133.
Hoei-Hansen CE, Carlsen E, Jørgensen N, Leffers H, Skakkebæk NE. Rajpert-De Meyts E. Towards a non-invasive method for early detection of testicular neoplasia in semen samples by identification of fetal germ cell-specific markers. Hum Reprod. 2007;22:167–173. [PubMed: 16920726]
134.
van Casteren NJ, Stoop H, Dohle GR, de Wit R, Oosterhuis JW, Looijenga LHJ. Noninvasive detection of testicular carcinoma in situ in semen using OCT3/4. Eur Urol. 2008;54:153–158. [PubMed: 17996359]
135.
Almstrup K, Lippert M, Mogensen HO, Nielsen JE, Hansen JD, Daugaard G, Jørgensen N, Foged NT, Skakkebæk NE, Rajpert-De Meyts E. Screening of subfertile men for testicular carcinoma in situ by an automated image analysis-based cytological test of the ejaculate. Int J Androl. 2011;34:e21–e31. [PubMed: 21696398]
136.
Radtke A, Cremers JF, Kliesch S, Riek S, Junker K, Mohamed SA, Anheuser P, Belge G, Dieckmann KP. Can germ cell neoplasia in situ be diagnosed by measuring serum levels of microRNA371a-3p? J Cancer Res Clin Oncol. 2017;143(11):2383–2392. [PMC free article: PMC5640733] [PubMed: 28819887]
137.
Pelloni M, Coltrinari G, Paoli D, Pallotti F, Lombardo F, Lenzi A, Gandini L. Differential expression of miRNAs in the seminal plasma and serum of testicular cancer patients. Endocrine. 2017 Sep;57(3):518–527. Epub 2016 Oct 28. PMID. [PubMed: 27796811] [CrossRef]
138.
Boellaard WPA, Gillis AJM, van Leenders GJLH, Stoop H, van Agthoven T, Dorssers LCJ, Dinkelman-Smit M, Boormans JL, Looijenga LHJ. Cellular origin of microRNA-371a-3p in healthy males based on systematic urogenital tract tissue evaluation. Andrology. 2019;7(4):463–468. doiPMID. [PMC free article: PMC6767197] [PubMed: 30786164] [CrossRef]
139.
Radtke A, Dieckmann KP, Grobelny F, Salzbrunn A, Oing C, Schulze W, Belge G. Expression of miRNA-371a-3p in seminal plasma and ejaculate is associated with sperm concentration. Andrology. 2019;7(4):469–474. doiPMID. [PubMed: 31310058] [CrossRef]
140.
Gilligan TD, Seidenfeld J, Basch EM, Einhorn LH, Fancher T, Smith DC, Stephenson AJ, Vaughn DJ, Cosby R, Hayes DF., American Society of Clinical Oncology. American Society of Clinical Oncology Clinical Practice Guideline on uses of serum tumor markers in adult males with germ cell tumors. J Clin Oncol. 2010;28:3388–3404. [PubMed: 20530278]
141.
Gitlin D, Perricelli A. Synthesis of serum albumin, prealbumin, alpha-fetoprotein, 1-antitrypsin and transferrin by the human yolk sac. Nature. 1970;228:995–996. [PubMed: 5529798]
142.
von Eyben FE, de Graaff WE, Marrink J, Blaabjerg O, Sleijfer DT, Koops HS, Oosterhuis JW, Petersen PH, van Echten-Arends J, de Jong B. Serum lactate dehydrogenase isoenzyme-1 activity in patients with testicular germ cell tumours correlates with the total number of copies of the short arm of chromosome 12 in the tumour. Mol Gen Genet. 1991;235:140–147. [PubMed: 1435725]
143.
Bandak M, Jørgensen N, Juul A, Lauritsen J, Gundgaard Kier MG, Mortensen MS, et al. Preorchiectomy Leydig cell dysfunction in patients with testicular cancer. Clin Genitourin Cancer 15(1):e37–43, 2016. doi: 10.1016/j.clgc.2016. 07.006. [PubMed: 27524512] [CrossRef]
144.
Palmer RD, Murray MJ, Saini HK, et al. Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Research. 2010;70:2911–2923. [PMC free article: PMC3000593] [PubMed: 20332240]
145.
Gillis AJ, Rijlaarsdam MA, Eini R, et al. Targeted serum miRNA (TSmiR) test for diagnosis and follow-up of (testicular) germ cell cancer patients: a proof of principle. Mol Oncology. 2013;7:1083–1092. [PMC free article: PMC5528443] [PubMed: 24012110]
146.
Dieckmann KP, Radtke A, Geczi L, Matthies C, Anheuser P, Eckardt U, Sommer J, Zengerling F, Trenti E, Pichler R, Belz H, Zastrow S, Winter A, Melchior S, Hammel J, Kranz J, Bolten M, Krege S, Haben B, Loidl W, Ruf CG, Heinzelbecker J, Heidenreich A, Cremers JF, Oing C, Hermanns T, Fankhauser CD, Gillessen S, Reichegger H, Cathomas R, Pichler M, Hentrich M, Eredics K, Lorch A, Wülfing C, Peine S, Wosniok W, Bokemeyer C, Belge G. Serum Levels of MicroRNA-371a-3p (M371 Test) as a New Biomarker of Testicular Germ Cell Tumors: Results of a Prospective Multicentric Study. J Clin Oncol. 2019;37(16):1412–1423. [PMC free article: PMC6544462] [PubMed: 30875280] [CrossRef]
147.
Piao J, Lafin JT, Scarpini CG, Nuño MM, Syring I, Dieckmann KP, Belge G, Ellinger J, Amatruda JF, Bagrodia A, Coleman N, Krailo MD, Frazier AL, Murray MJ. A Multi-institutional Pooled Analysis Demonstrates That Circulating miR-371a-3p Alone is Sufficient for Testicular Malignant Germ Cell Tumor Diagnosis. Clin Genitourin Cancer. 2021;19(6):469–479. [PMC free article: PMC9084514] [PubMed: 34629299] [CrossRef]
148.
Aganovic L, Cassidy F. Imaging of the scrotum. Radiol Clin North Am. 2012;50:1145–1165. [PubMed: 23122043]
149.
Cantisani V, Di Leo N, Bertolotto M, Fresilli D, Granata A, Polti G, Polito E, Pacini P, Guiban O, Del Gaudio G, Dolcetti V. DʼAndrea V, Di Pierro GB, Verrengia M, Drudi FM, Catalano C. Role of multiparametric ultrasound in testicular focal lesions and diffuse pathology evaluation, with particular regard to elastography: Review of literature. Andrology. 2021;9(5):1356–1368. [PubMed: 34114745] [CrossRef]
150.
Tsili AC, Argyropoulou MI, Astrakas LG, Ntoulia EA, Giannakis D, Sofikitis N, Tsampoulas K. Dynamic contrast-enhanced subtraction MRI for characterizing intratesticular mass lesions. AJR Am J Roentgenol. 2013;200:578–585. [PubMed: 23436847]
151.
Honecker F, Aparicio J, Berney D, Beyer J, Bokemeyer C, Cathomas R, Clarke N, Cohn-Cedermark G, Daugaard G, Dieckmann KP, Fizazi K, Fosså S, Germa-Lluch JR, Giannatempo P, Gietema JA, Gillessen S, Haugnes HS, Heidenreich A, Hemminki K, Huddart R, Jewett MAS, Joly F, Lauritsen J, Lorch A, Necchi A, Nicolai N, Oing C, Oldenburg J, Ondruš D, Papachristofilou A, Powles T, Sohaib A, Ståhl O, Tandstad T, Toner G, Horwich A. ESMO Consensus Conference on testicular germ cell cancer: diagnosis, treatment and follow-up. Ann Oncol. 2018;29(8):1658–1686. [PubMed: 30113631] [CrossRef]
152.
Daugaard G, Gundgaard MG, Mortensen MS, Agerbaek M, Holm NV, Rorth M, et al. Surveillance for stage I nonseminoma testicular cancer: outcomes and long-term follow-up in a population-based cohort. J Clin Oncol. 2014;32(34):3817–23. [PubMed: 25267754]
153.
Verrill C, Yilmaz A, Srigley JR, et al. Am J Surg Pathol. 2017;41:e22–e32. et Members of the International Society of Urological Pathology Testicular Tumor Panel. Reporting and Staging of Testicular Germ Cell Tumors: The International Society of Urological Pathology (ISUP) Testicular Cancer Consultation Conference Recommendations. [PubMed: 28368923]
154.
Sobin LH, Gospodarowicz M, Wittekind C (eds). TNM Classification of Malignant Tumours, UICC International Union Against Cancer 7th ed. Wiley-Blackwell, pp 249-254, 2009.
155.
Chresta CM, Masters JWR, Hickman JA. Hypersensitivity of human testicular tumors to etoposide-induced apoptosis is associated with functional p53 and a high bax:bcl-2 ratio. Cancer Res. 1996;56:1834–1841. [PubMed: 8620501]
156.
Timmerman DM, Eleveld TF, Gillis AJM, Friedrichs CC, Hillenius S, Remmers TL, Sriram S, Looijenga LHJ. The Role of TP53 in Cisplatin Resistance in Mediastinal and Testicular Germ Cell Tumors. Int J Mol Sci. 2021;22(21):11774. [PMC free article: PMC8583723] [PubMed: 34769213] [CrossRef]
157.
King J, Adra N, Einhorn LH. Testicular Cancer: Biology to Bedside. Cancer Res. 2021;81(21):5369–5376. [PubMed: 34380632] [CrossRef]
158.
Mortensen MS, Gundgaard MG, Daugaard G. Treatment options for carcinoma in situ testis. Int J Androl. 2011;34(4 Pt 2):e32–e36. [PubMed: 21651575]
159.
Giwercman A, von der Maase H, Berthelsen JG, Rorth M, Bertelsen A, Skakkebaek NE. Localized irradiation of testes with carcinoma in situ: effects on Leydig cell function and eradication of malignant germ cells in 20 patients. J Clin Endocrinol Metab. 1991;73:596–603. [PubMed: 1908483]
160.
Bang AK, Petersen JH, Petersen PM, Andersson AM, Daugaard G, Jørgensen N. Testosterone production is better preserved after 16 than 20 Gray irradiation treatment against testicular carcinoma in situ cells. Int J Radiat Oncol Biol Phys. 2009;75:672–676. [PubMed: 19250763]
161.
Dieckmann KP, Wilken S, Loy V, Matthies C, Kleinschmidt K, Bedke J, Martinschek A, Souchon R, Pichlmeier U, Kliesch S. Treatment of testicular intraepithelial neoplasia (intratubular germ cell neoplasia unspecified) with local radiotherapy or with platinum-based chemotherapy: a survey of the German Testicular Cancer Study Group. Ann Oncol. 2013;24(5):1332–7. [PubMed: 23293116] [CrossRef]
162.
Fosså SD, Aass N, Heilo A, Daugaard G, E, Skakkebaek N, Stenwig AE, Nesland JM, Looijenga LH, Oosterhuis JW. Testicular carcinoma in situ in patients with extragonadal germ-cell tumours: the clinical role of pretreatment biopsy. Ann Oncol. 2003;14:1412–1418. [PubMed: 12954581]
163.
Blok JM, Groot HJ, Huele EH, de Wit R, Horenblas S, Nuver J, Groenewegen G, Bosch JLHR, Witjes JA, Tromp JM, de Brouwer PJM, van den Berg HA, Vanneste BGL, Smilde TJ, Aarts MJB, Gietema JA, Meijer RP, Schaapveld M. Dose-Dependent Effect of Platinum-Based Chemotherapy on the Risk of Metachronous Contralateral Testicular Cancer. J Clin Oncol. 2021;39:319–327. [PubMed: 33119475]
164.
Hellesnes R, Myklebust TÅ, Bremnes RM, Karlsdottir Á, Kvammen Ø, Negaard HFS, Tandstad T, Wilsgaard T, Fosså SD, Haugnes HS. Metachronous Contralateral Testicular Cancer in the Cisplatin Era: A Population-Based Cohort Study. J Clin Oncol. 2021;39(4):308–318. PMID. [PubMed: 33356420]
165.
Schrader M, Müller M, Sofikitis N, Straub B, Krause H, Miller K. "Onco-TESE": testicular sperm extraction in azoospermic cancer patients before chemotherapy-new guidelines? Urology. 2003;61:421–425. [PubMed: 12597960]
166.
Suzuki K, Shin T, Shimomura Y, Iwahata T, Okada H. Spermatogenesis in tumor-bearing testes in germ cell testicular cancer patients. Hum Reprod. 2015;30(12):2853–2858. [PubMed: 26428212]
167.
Beyer J, Albers P, Altena R, Aparicio J, Bokemeyer C, Busch J, et al. Maintaining success, reducing treatment burden, focusing on survivorship: highlights from the third European consensus conference on diagnosis and treatment of germ-cell cancer. Ann Oncol. 2013;24(4):878–88. [PMC free article: PMC3603440] [PubMed: 23152360]
168.
Nichols CR, Roth B, Albers P, Einhorn LH, et al. Active surveillance is the preferred approach to clinical stage I testicular cancer. J Clin Oncol. 2013;31:3490–3493. [PubMed: 24002502]
169.
Heidenreich A, Paffenholz P, Nestler T, Pfister D. Primary and Postchemotherapy Retroperitoneal Lymphadenectomy for Testicular Cancer. Oncol Res Treat. 2018;41(6):370–378. [PubMed: 29772568] [CrossRef]
170.
Mortensen MS, Lauritsen J, Gundgaard MG, Agerbaek M, Holm NV, Christensen IJ, et al. A nationwide cohort study of stage I seminoma patients followed on a surveillance program. Eur Urol. 2014;66(6):1172–8. [PubMed: 25064686] [CrossRef]
171.
Tandstad T, Ståhl O, Håkansson U, et al. One course of adjuvant BEP in clinical stage I nonseminoma mature and expanded results from the SWENOTECA group. Ann Oncol. 2014;25:2167–2172. [PubMed: 25114021]
172.
Oldenburg J, Berney DM, Bokemeyer C, Climent MA, Daugaard G, Gietema JA, De Giorgi U, Haugnes HS, Huddart RA, Leão R, Sohaib A, Gillessen S, Powles T., ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org; EURACAN. Testicular seminoma and nonseminoma: ESMO-EURACAN Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33(4):362–375. doiPMID. [PubMed: 35065204] [CrossRef]
173.
Beyer J, Collette L, Sauvé N, Daugaard G, Feldman DR, Tandstad T, Tryakin A, Stahl O, Gonzalez-Billalabeitia E, De Giorgi U, Culine S, de Wit R, Hansen AR, Bebek M, Terbuch A, Albany C, Hentrich M, Gietema JA, Negaard H, Huddart RA, Lorch A, Cafferty FH, Heng DYC, Sweeney CJ, Winquist E, Chovanec M, Fankhauser C, Stark D, Grimison P, Necchi A, Tran B, Heidenreich A, Shamash J, Sternberg CN, Vaughn DJ, Duran I, Bokemeyer C, Patrikidou A, Cathomas R, Assele S, Gillessen S., International Germ Cell Cancer Classification Update Consortium. Survival and New Prognosticators in Metastatic Seminoma: Results From the IGCCCG-Update Consortium. J Clin Oncol. 2021;39(14):1553–1562. doiPMID. [PMC free article: PMC8099394] [PubMed: 33729863] [CrossRef]
174.
Gillessen S, Sauvé N, Collette L, Daugaard G, de Wit R, Albany C, Tryakin A, Fizazi K, Stahl O, Gietema JA, De Giorgi U, Cafferty FH, Hansen AR, Tandstad T, Huddart RA, Necchi A, Sweeney CJ, Garcia-Del-Muro X, Heng DYC, Lorch A, Chovanec M, Winquist E, Grimison P, Feldman DR, Terbuch A, Hentrich M, Bokemeyer C, Negaard H, Fankhauser C, Shamash J, Vaughn DJ, Sternberg CN, Heidenreich A, Beyer J., International Germ Cell Cancer Classification Update Consortium. Predicting Outcomes in Men With Metastatic Nonseminomatous Germ Cell Tumors (NSGCT): Results From the IGCCCG Update Consortium. J Clin Oncol. 2021;39(14):1563–1574. doiPMID. [PMC free article: PMC8099402] [PubMed: 33822655] [CrossRef]
175.
Borer JG, Tan PE, Diamond DA. The spectrum of Sertoli cell tumors in children. Urol Clin North Am. 2000;27:529–541. [PubMed: 10985152]
176.
Harms D, Kock LR. Testicular juvenile granulosa cell and Sertoli cell tumours: a clinicopathological study of 29 cases from the Kiel Paediatric Tumour Registry. Virchows Arch. 1997;430:301–309. [PubMed: 9134041]
177.
Kaplan GW, Cromie WJ, Kelalis PP, Silber I. Tank ESJr. Gonadal stromal tumors: a report of prepubertal tumor registry. J Urol. 1986;136:300–302. [PubMed: 3723681]
178.
Idrees MT, Ulbright TM, Oliva E, et al. Histopathology. 2017;70:513–521. et Members of the International Society of Urological Pathology Testicular Tumour Panel. The World Health Organization 2016 classification of testicular non-germ cell tumours: a review and update from the International Society of Urological Pathology Testis Consultation Panel. [PubMed: 27801954]
179.
Iczkowski KA, Bostwick DG, Roche PC, Cheville JC. Inhibin A is a sensitive and specific marker for testicular sex cord-stromal tumors. Mod Pathol. 1998;11:774–779. [PubMed: 9720507]
180.
Toppari J, Kaipia A, Kaleva M, et al. Inhibin gene expression in a large cell calcifying Sertoli cell tumour and serum inhibin and activin levels. APMIS. 1998;106:101–113. [PubMed: 9524568]
181.
Henley JD, Young RH, Ulbright TM. Malignant Sertoli cell tumors of the testis: a study of 13 examples of a neoplasm frequently misinterpreted as seminoma. Am J Surg Pathol. 2002;26:541–550. [PubMed: 11979085]
182.
Kommoss F, Oliva E, Bittinger F, Kirkpatrick CJ, Amin MB, Bhan AK, Young RH, Scully RE. Inhibin-alpha CD99, HEA125, PLAP, and chromogranin immunoreactivity in testicular neoplasms and the androgen insensitivity syndrome. Hum Pathol. 2000;31:1055–1061. [PubMed: 11014571]
183.
Rey R, Sabourin JC, Venara M, Long WQ, Jaubert F, Zeller WP, Duvillard P, Chemes H, Bidart JM. Anti-Mullerian hormone is a specific marker of Sertoli- and granulosa-cell origin in gonadal tumors. Hum Pathol. 2000;31:1202–1208. [PubMed: 11070112]
184.
Ketola I, Pentikainen V, Vaskivuo T, Ilvesmaki V, Herva R, Dunkel L, Tapanainen JS, Toppari J, Heikinheimo M. Expression of transcription factor GATA-4 during human testicular development and disease. J Clin Endocrinol Metab. 2000;85:3925–3931. [PubMed: 11061558]
185.
Lottrup G, Nielsen JE, Skakkebæk NE, Juul A, Rajpert-De Meyts E. Abundance of DLK1, differential expression of CYP11B1, CYP21A2, MC2R and lack of INSL3 distinguish testicular adrenal rest tumours from Leydig cell tumours. Eur J Endocrinol. 2015;172:491–499. [PubMed: 25609776]
186.
Holm M, Rajpert-De Meyts E, Andersson A-M, Skakkebæk NE. Leydig cell micronodules are a common finding in testicular biopsies from men with impaired spermatogenesis and are associated with decreased Testosterone/LH ratio. J Pathol. 2003;199:378–386. [PubMed: 12579540]
187.
Lottrup G, Nielsen JE, Maroun LL, Møller LMA, Yassin M, Leffers H, Skakkebæk NE, Rajpert-De Meyts E. Expression patterns of DLK1 and INSL3 identify stages of Leydig cell differentiation during normal development and in testicular pathologies, including testicular cancer and Klinefelter syndrome. Hum Reprod. 2014;29:1637–1650. [PubMed: 24908673]
188.
Fankhauser CD, Grogg JB, Hayoz S, et al. Risk Factors and Treatment Outcomes of 1,375 Patients with Testicular Leydig Cell Tumors: Analysis of Published Case Series Data. J Urol. 2020;203:949–956. [PubMed: 31845841]
189.
Sørensen RR, Johannsen TH, Skakkebaek NE, Rajpert-De Meyts E. Leydig cell clustering and Reinke crystal distribution in relation to hormonal function in adult patients with testicular dysgenesis syndrome (TDS) including cryptorchidism. Hormones. 2016;15:518–526. [PubMed: 28222406]
190.
Lottrup G, Belling K, Leffers H, Nielsen JE, Dalgaard MD, Juul A, Skakkebæk NE, Brunak S, Rajpert-De Meyts E. Comparison of global gene expression profiles of microdissected human foetal Leydig cells with their normal and hyperplastic adult equivalents. Mol Hum Reprod. 2017;23(5):339–354. [PubMed: 28333300]
191.
Rich MA, Keating MA. Leydig cell tumors and tumors associated with congenital adrenal hyperplasia. Urol Clin North Am. 2000;27:519–528. [PubMed: 10985151]
192.
Pozza C, Pofi R, Tenuta M, Tarsitano MG, Sbardella E, Fattorini G, Cantisani V, Lenzi A, Isidori AM, Gianfrilli D. TESTIS UNIT. Clinical presentation, management and follow-up of 83 patients with Leydig cell tumors of the testis: a prospective case-cohort study. Hum Reprod. 2019;34(8):1389–1403. doiPMID. [PMC free article: PMC6688875] [PubMed: 31532522] [CrossRef]
193.
Shenker A, Laue L, Kosugi S, Merendino JJ Jr, Minegishi T, Cutler GB Jr. A constitutively activating mutation of the luteinizing hormone receptor in familial male precocious puberty. Nature. 1993;365:652–654. [PubMed: 7692306]
194.
Liu G, Duranteau L, Carel JC, Monroe J, Doyle DA, Shenker A. Leydig-cell tumors caused by an activating mutation of the gene encoding the luteinizing hormone receptor. N Engl J Med. 1999;341:173–176. [PubMed: 10580072]
195.
Mortensen LJ, Blomberg Jensen M, Christiansen P, Rønholt AM, Jørgensen A, Frederiksen H, Nielsen JE, Loya AC, Grønkær Toft B, Skakkebæk NE, Rajpert-De Meyts E, Juul A. Germ cell neoplasia in situ and preserved fertility despite suppressed gonadotropins in a patient with testotoxicosis. J Clin Endocrinol Metab. 2017;102:4411–4416. [PubMed: 29029242]
196.
Iiri T, Herzmark P, Nakamoto JM, van Dop C, Bourne HR. Rapid GDP release from Gs alpha in patients with gain and loss of endocrine function. Nature. 1994;371:164–168. [PubMed: 8072545]
197.
Fragoso MC, Latronico AC, Carvalho FM, Zerbini MC, Marcondes JA, Araujo LM, Lando VS, Frazzatto ET, Mendonca BB, Villares SM. Activating mutation of the stimulatory G protein (gsp) as a putative cause of ovarian and testicular human stromal Leydig cell tumors. J Clin Endocrinol Metab. 1998;83:2074–2081. [PubMed: 9626141]
198.
Themmen APN, Brunner HG. Luteinizing hormone receptor mutations and sex differentiation. Eur J Endocrinol. 1996;134:533–540. [PubMed: 8664969]
199.
Carvajal-Carmona LG, Alam NA, Pollard PJ, et al. Adult Leydig cell tumors of the testis caused by germline fumarate hydratase mutations. J Clin Endocrinol Metab. 2006;91:3071–3075. [PubMed: 16757530]
200.
Mieritz MG, Raket LL, Hagen CP, Nielsen JE, Talman ML, Petersen JH, Sommer SH, Main KM, Jørgensen N, Juul A. A Longitudinal Study of Growth, Sex Steroids, and IGF-1 in Boys With Physiological Gynecomastia. J Clin Endocrinol Metab. 2015;100:3752–3759. [PubMed: 26287961]
201.
Kanakis GA, Nordkap L, Bang AK, Calogero AE, Bártfai G, Corona G, Forti G, Toppari J, Goulis DG, Jørgensen N. EAA clinical practice guidelines - gynecomastia evaluation and management. Andrology. 2019;7:778–793. [PubMed: 31099174]
202.
Kim I, Young RH, Scully RE. Leydig cell tumors of the testis. A clinicopathologic study of 40 cases and review of the literature. Am J Surg Pathol. 1985;9:177–192. [PubMed: 3993830]
203.
Rushton HG, Belman AB. Testis-sparing surgery for benign lesions of the prepubertal testis. Urol Clin North Am. 1993;20:27–37. [PubMed: 8434437]
204.
Bozzini G, Picozzi S, Gadda F, Colombo R, Decobelli O, Palou J, Colpi G, Carmignani L. Long-term follow-up using testicle-sparing surgery for Leydig cell tumor. Clin Genitourin Cancer. 2013;11(3):321–324. [PubMed: 23317518]
205.
Fankhauser CD, Roth L, Kranzbühler B, Eberli D, Bode P, Moch H, Oliveira P, Ramani V, Beyer J, Hermanns T. The Role of Frozen Section Examination During Inguinal Exploration in Men with Inconclusive Testicular Tumors: A Systematic Review and Meta-analysis. Eur Urol Focus. 2021;7(6):1400–1402. doiPMID. [PubMed: 32684510] [CrossRef]
206.
Cheville JC, Sebo TJ, Lager DJ, Bostwick DG, Farrow GM. Leydig cell tumor of the testis: a clinicopathologic, DNA content, and MIB-1 comparison of nonmetastasizing and metastasizing tumors. Am J Surg Pathol. 1998;22:1361–1367. [PubMed: 9808128]
207.
Calaway AC, Tachibana I, Masterson TA, Foster RS, Einhorn LH, Cary C. Oncologic outcomes following surgical management of clinical stage II sex cord stromal tumors. Urology. 2019;127:74–79. PMID. [PubMed: 30807775]
208.
Hamwi GJ, Gwinup G, Mostow JH, Besch PK. Activation of testicular adrenal rest tissue by prolonged excessive ACTH production. J Clin Endocrinol Metab. 1963;23:861–869. [PubMed: 14064131]
209.
Engels M, Span PN, van Herwaarden AE, Sweep FCGJ, Stikkelbroeck NMML, Claahsen-van der Grinten HL. Testicular Adrenal Rest Tumors: Current Insights on Prevalence, Characteristics, Origin, and Treatment. Endocr Rev. 2019;40:973–987. [PubMed: 30882882]
210.
Venara M, Rey R, Bergada I, Mendilaharzu H, Campo S, Chemes H. Sertoli cell proliferations of the infantile testis: an intratubular form of Sertoli cell tumor? Am J Surg Pathol. 2001;25:1237–1244. [PubMed: 11688457]
211.
Young RH, Koelliker DD, Scully RE. Sertoli cell tumors of the testis, not otherwise specified. Am J Surg Pathol. 1998;22:709–721. [PubMed: 9630178]
212.
Perrone F, Bertolotti A, Montemurro G, Paolini B, Pierotti MA, Colecchia M. Frequent mutation and nuclear localization of β-catenin in Sertoli cell tumors of the testis. Am J Surg Pathol. 2014;38(1):66–71. [PubMed: 24061522]
213.
Giglio M, Medica M, De Rose AF, Germinale F, Ravetti JL, Carmignani G, Giglio M, Medica M, De Rose AF, Germinale F, Ravetti JL, Carmignani G. Testicular Sertoli cell tumours and relative sub-types. Analysis of clinical and prognostic features. Urol Int. 2003;70:205–210. [PubMed: 12660458]
214.
Gourgari E, Saloustros E, Stratakis CA. Large-cell calcifying Sertoli cell tumors of the testes in pediatrics. Curr Opin Pediatr. 2012;24:518–22. [PMC free article: PMC4132931] [PubMed: 22732638]
215.
Carney JA, Gordon H, Carpenter PC, Shenoy BV, Go VL. The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine. 1985;64:270–283. [PubMed: 4010501]
216.
Proppe KH, Scully RE. Large-cell calcifying Sertoli cell tumor of the testis. Am J Clin Pathol. 1980;74:607–619. [PubMed: 7446466]
217.
Washecka R, Dresner MI, Honda SA. Testicular tumors in Carney’s complex. J Urol. 2002;167:1299–1302. [PubMed: 11832717]
218.
Kratzer SS, Ulbright TM, Talerman A, Srigley JR, Roth LM, Wahle GR, Moussa M, Stephens JK, Millos A, Young RH. Large cell calcifying Sertoli cell tumor of the testis. Contrasting features of six malignant and six benign tumors and a review of literature. Am J Surg Pathol. 1997;21:1271–1280. [PubMed: 9351565]
219.
Stratakis CA, Carney JA, Lin JP, Papanicolau DA, Karl M, Kastner DL, Pras E, Chrousos GP. Carney complex, a familial multiple neoplasia and lentiginosis syndrome. Analysis of 11 kindreds and linkage to the short arm of chromosome 2. J Clin Invest. 1996;97:699–705. [PMC free article: PMC507106] [PubMed: 8609225]
220.
Casey M, Mah C, Merliss AD, Kirschner LS, Taymans SE, Denio AE, Korf B, Irvine AD, Hughes A, Carney JA, Stratakis CA, Basson CT. Identification of a novel genetic locus for familial cardiac myxomas and Carney complex. Circulation. 1998;98:2560–2566. [PubMed: 9843463]
221.
Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients the Carney complex. Nat Genet. 2000;26:89–92. [PubMed: 10973256]
222.
Casey M, Vaughan CJ, He J, Hatcher CJ, Winter JM, Weremowicz S, Montgomery K, Kucherlapati R, Morton CC, Basson CT. Mutations in the protein kinase A R1alpha regulatory subunit cause familial cardiac myxomas and Carney complex. J Clin Invest. 2000;106:R31–8. [PMC free article: PMC381290] [PubMed: 10974026]
223.
Libé R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, Raffin-Sanson ML, Siegel J, Moran J, Drori-Herishanu L, Faucz FR, Lodish M, Nesterova M, Bertagna X, Bertherat J, Stratakis CA. Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with Carney complex (CNC) caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab. 2011;96(1):E208–214. [PMC free article: PMC3038483] [PubMed: 21047926]
224.
Ham S, Meachem SJ, Choong CS, Charles AK, Baynam GS, Jones TW, Samarajeewa NU, Simpson ER, Brown KA. Overexpression of aromatase associated with loss of heterozygosity of the STK11 gene accounts for prepubertal gynecomastia in boys with Peutz-Jeghers syndrome. J Clin Endocrinol Metab. 2013;98:E1979–E1987. [PubMed: 24037887]
225.
Hemminki A, Markie D, Tomlison I, Avizienyte E, Roth S, Loukola A, Bignell G, Warren W, Aminoff M, Hoglund P, Jarvinen H, Kristo P, Pelin K, Ridanpaa M, Salovaara R, Toro T, Bodmer W, Olschwang S, Olsen AS, Stratton MR, de la Chapelle A, Aaltonen LA. A serine/threonine kinase gene defective in Peutz-Jegher syndrome. Nature. 1998;391:184–187. [PubMed: 9428765]
226.
Brown B, Ram A, Clayton P, et al. Conservative management of bilateral Sertoli cell tumors of the testicle in association with the Carney complex: a case report. J Pediatr Surg. 2007;42:E13–15. [PubMed: 17848226]
227.
Grogg J, Schneider K, Bode PK, Kranzbühler B, Eberli D, Sulser T, Lorch A, Beyer J, Hermanns T, Fankhauser CD. Sertoli Cell Tumors of the Testes: Systematic Literature Review and Meta-Analysis of Outcomes in 435 Patients. Oncologist. 2020;25(7):585–590. PMID. [PMC free article: PMC7356704] [PubMed: 32043680]
228.
Hofmann M, Schlegel PG, Hippert F, Schmidt P, von-Schweinitz D, Leuschner I, Göbel U, Calaminus G, Schneider DT., MAKEI study group. Testicular sex cord stromal tumors: Analysis of patients from the MAKEI study. Pediatr Blood Cancer. 2013;60(10):1651–1655. [PubMed: 23733594]
229.
Kao CS, Cornejo KM, Ulbright TM, Young RH. Juvenile granulosa cell tumors of the testis: a clinicopathologic study of 70 cases with emphasis on its wide morphologic spectrum. Am J Surg Pathol. 2015;39(9):1159–69. [PubMed: 26076062]
230.
Grogg JB, Schneider K, Bode PK, Kranzbühler B, Eberli D, Sulser T, Beyer J, Lorch A, Hermanns T, Fankhauser CD. Risk factors and treatment outcomes of 239 patients with testicular granulosa cell tumors: a systematic review of published case series data. J Cancer Res Clin Oncol. 2020;146(11):2829–2841. PMID. [PMC free article: PMC7519920] [PubMed: 32719989]
231.
Boyer A, Paquet M, Laguë MN, Hermo L, Boerboom D. Dysregulation of WNT/CTNNB1 and PI3K/AKT signaling in testicular stromal cells causes granulosa cell tumor of the testis. Carcinogenesis. 2009;30(5):869–878. [PMC free article: PMC2675650] [PubMed: 19237610]
232.
Kalfa N, Ecochard A, Patte C, Duvillard P, Audran F, Pienkowski C, Thibaud E, Brauner R, Lecointre C, Plantaz D, Guedj AM, Paris F, Baldet P, Lumbroso S, Sultan C. Activating mutations of the stimulatory G protein in juvenile ovarian granulosa cell tumors: a new prognostic factor? J Clin Endocrinol Metab. 2006;91(5):1842–1847. [PubMed: 16507630]
233.
Jimenez-Quintero LP, Ro JY, Zavala-Pompa A, Amin MB, Tetu B, Ordonez NG, Ayala AG. Granulosa cell tumor of the adult testis: a clinicopathologic study of seven cases and a review of the literature. Hum Pathol. 1993;24:1120–1125. [PubMed: 8406422]
234.
Shah SP, Köbel M, Senz J, Morin RD, Clarke BA, Wiegand KC, Leung G, Zayed A, Mehl E, Kalloger SE, Sun M, Giuliany R, Yorida E, Jones S, Varhol R, Swenerton KD, Miller D, Clement PB, Crane C, Madore J, Provencher D, Leung P, DeFazio A, Khattra J, Turashvili G, Zhao Y, Zeng T, Glover JN, Vanderhyden B, Zhao C, Parkinson CA, Jimenez-Linan M, Bowtell DD, Mes-Masson AM, Brenton JD, Aparicio SA, Boyd N, Hirst M, Gilks CB, Marra M, Huntsman DG. Mutation of FOXL2 in granulosa-cell tumors of the ovary. N Engl J Med. 2009;360:2719–2729. [PubMed: 19516027]
235.
Lima JF, Jin L, de Araujo AR, Erikson-Johnson MR, Oliveira AM, Sebo TJ, Keeney GL, Medeiros F. FOXL2 mutations in granulosa cell tumors occurring in males. Arch Pathol Lab Med. 2012;136(7):825–828. [PubMed: 22742556]
236.
Morrish DW, Venner PM, Siy O, Barron G, Bhardwaj D, Outhet D. Mechanisms of endocrine dysfunction in patients with testicular cancer. J Natl Cancer Inst. 1990;82:412–418. [PubMed: 2304089]
237.
van Casteren NJ, Boellaard WP, Romijn JC, Dohle GR. Gonadal dysfunction in male cancer patients before cytotoxic treatment. Int J Androl. 2010;33:c73–79. [PubMed: 19538481]
238.
Rives N, Perdrix A, Hennebicq S, Saïas-Magnan J, Melin MC, Berthaut I, Barthélémy C, Daudin M, Szerman E, Bresson JL, Brugnon F, Bujan L. The semen quality of 1158 men with testicular cancer at the time of cryopreservation: results of the French National CECOS Network. J Androl. 2012;33:1394–1401. [PubMed: 22837112]
239.
Haugnes HS, Bosl GJ, Boer H, Gietema JA, Brydøy M, Oldenburg J, Dahl AA, Bremnes RM, Fosså SD. Long-term and late effects of germ cell testicular cancer treatment and implications for follow-up. J Clin Oncol. 2012;30:3752–3763. [PubMed: 23008318]
240.
Fung C, Fossa SD, Williams A, Travis LB. Long-term Morbidity of Testicular Cancer Treatment. Urol Clin North Am. 2015;42(3):393–408. [PubMed: 26216826]
241.
Brydøy M, Fosså SD, Klepp O, Bremnes RM, Wist EA, Bjøro T, Wentzel-Larsen T, Dahl O., Norwegian Urology Cancer Group (NUCG) III study group. Sperm counts and endocrinological markers of spermatogenesis in long-term survivors of testicular cancer. Br J Cancer. 2012;107:1833–1839. [PMC free article: PMC3504949] [PubMed: 23169336]
242.
Hsiao W, Stahl PJ, Osterberg EC, Nejat E, Palermo GD, Rosenwaks Z, Schlegel PN. Successful treatment of postchemotherapy azoospermia with microsurgical testicular sperm extraction: the Weill Cornell experience. J Clin Oncol. 2011;29:1607–1611. [PubMed: 21402606]
243.
Gat I, Toren A, Hourvitz A, Raviv G, Band G, Baum M, Lerner-Geva L, Inbar R, Madgar I. Sperm preservation by electroejaculation in adolescent cancer patients. Pediatr Blood Cancer. 2014;61:286–90. [PubMed: 24000134]
244.
Goossens E, Jahnukainen K, Mitchell RT, van Pelt A, Pennings G, Rives N, Poels J, Wyns C, Lane S, Rodriguez-Wallberg KA, Rives A, Valli-Pulaski H, Steimer S, Kliesch S, Braye A, Andres MM, Medrano J, Ramos L, Kristensen SG, Andersen CY, Bjarnason R, Orwig KE, Neuhaus N, Stukenborg JB.. Fertility preservation in boys: recent developments and new insights. Hum Reprod Open. 2020; 2020(3):hoaa016. doi: 10.1093/hropen/hoaa016. [PMC free article: PMC7275639] [PubMed: 32529047] [CrossRef]
245.
Diao L, Turek PJ, John CM, Fang F, Reijo Pera RA. Roles of Spermatogonial Stem Cells in Spermatogenesis and Fertility Restoration. Front Endocrinol (Lausanne). 2022 May 12;13:895528. doiPMID. [PMC free article: PMC9135128] [PubMed: 35634498] [CrossRef]
246.
Cvancarova M, Samuelsen SO, Magelssen H, Fossa SD. Reproduction rates after cancer treatment: experience from the Norwegian radium hospital. J Clin Oncol. 2009;27:334–343. [PubMed: 19075285]
247.
Bandak M, Jørgensen N, Juul A, Vogelius IR, Lauritsen J, Kier MG, Mortensen MS, Glovinski P, Daugaard G. Testosterone deficiency in testicular cancer survivors - a systematic review and meta-analysis. Andrology. 2016;4(3):382–388. [PubMed: 27009402]
248.
Bandak M, Jørgensen N, Juul A, Lauritsen J, Oturai PS, Mortensen J, Hojman P, Helge JW, Daugaard G. Leydig cell dysfunction, systemic inflammation and metabolic syndrome in long-term testicular cancer survivors. Eur J Cancer. 2017;84:9–17. [PubMed: 28772110]
249.
Willemse PM, Burggraaf J, Hamdy NA, Weijl NI, Vossen CY, van Wulften L, van Steijn-van Tol AQ, Rosendaal FR, Osanto S. Prevalence of the metabolic syndrome and cardiovascular disease risk in chemotherapy-treated testicular germ cell tumour survivors. Brit J Cancer. 2013;109:60–67. [PMC free article: PMC3708554] [PubMed: 23660945]
250.
Chovanec M, Abu Zaid M, Hanna N, El-Kouri N, Einhorn LH, Albany C. Long-term toxicity of cisplatin in germ-cell tumor survivors. Ann Oncol. 2017;28:2670–2679. [PMC free article: PMC6246726] [PubMed: 29045502]
251.
Lauritsen J, Hansen MK, Bandak M, Kreiberg MB, Skøtt JW, Wagner T, Gundgaard Kier MG, Holm NV, Agerbæk M, Gupta R, Dehlendorff C, Andersen KK, Daugaard G. Cardiovascular Risk Factors and Disease After Male Germ Cell Cancer. J Clin Oncol. 2020;38:584–592. [PubMed: 31821065]
252.
Chovanec M, Lauritsen J, Bandak M, Oing C, Kier GG, Kreiberg M, Rosenvilde J, Wagner T, Bokemeyer C, Daugaard G. Late adverse effects and quality of life in survivors of testicular germ cell tumour. Nat Rev Urol. 2021;18(4):227–245. PMID. [PubMed: 33686290]
253.
Kvammen Ø, Myklebust TÅ, Solberg A, Møller B, Klepp OH, Fosså SD, Tandstad T. Causes of inferior relative survival after testicular germ cell tumor diagnosed 1953-2015: A population-based prospective cohort study. PLoS One. 2019;14(12):e0225942. [PMC free article: PMC6919610] [PubMed: 31851716] [CrossRef]
254.
Hellesnes R, Myklebust TÅ, Fosså SD, Bremnes RM, Karlsdottir Á, Kvammen Ø, Tandstad T, Wilsgaard T, Negaard HFS, Haugnes HS. Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort. J Clin Oncol. 2021;39(32):3561–3573. PMID. [PubMed: 34388002]
255.
Milano MT, Dinh PC, Yang H, Zaid MA, Fossa SD, Feldman DR, Monahan PO, Travis LB, Fung C. Solid and Hematologic Neoplasms After Testicular Cancer: A US Population-Based Study of 24 900 Survivors. JNCI Cancer Spectr. 2020 Mar 17;4(3):pkaa017. doi: 10.1093/jncics/pkaa017. PMID: 32455335. [PMC free article: PMC7236780] [PubMed: 32455335] [CrossRef]
256.
Nichols CR, Nappi L, Kollmannsberger C, Hamilton R, Daneshmand S. Back to the Future-Moving Forward for Testicular Cancer Survivors. JNCI Cancer Spectr. 2019;4(2):pkz082. doiPMID. [PMC free article: PMC7065711] [PubMed: 32190816] [CrossRef]
257.
Dahl AA, Haaland CF, Mykletun A, Bremnes R, Dahl O, Klepp O, Wist E, Fosså SD. Study of anxiety disorder and depression in long-term survivors of testicular cancer. J Clin Oncol. 2005;23(10):2389–95. PMID. [PubMed: 15800331]
258.
Skaali T, Fossa SD, Andersson S, Langberg CW, Lehne G, Dahl AA. Is psychological distress in men recently diagnosed with testicular cancer associated with their neuropsychological test performance? Psycho-oncology. 2011;20:369–377. [PubMed: 20878832]
259.
Smith AB, Butow P, Olver I, Luckett T, Grimison P, Toner GC, Stockler MR, Hovey E, Stubbs J, Turner S, Hruby G, Gurney H, Alam M, Cox K, King MT. The prevalence, severity, and correlates of psychological distress and impaired health-related quality of life following treatment for testicular cancer: a survivorship study. J Cancer Surviv. 2016;10:223–233. [PubMed: 26178326]
260.
Kreiberg M, Bandak M, Lauritsen J, Andersen KK, Skøtt JW, Johansen C, Agerbaek M, Holm NV, Lau CJ, Daugaard G. Psychological stress in long-term testicular cancer survivors: a Danish nationwide cohort study. J Cancer Surviv. 2020;14(1):72–79. PMID. [PubMed: 31748852]
Copyright © 2000-2024, MDText.com, Inc.

This electronic version has been made freely available under a Creative Commons (CC-BY-NC-ND) license. A copy of the license can be viewed at http://creativecommons.org/licenses/by-nc-nd/2.0/.

Bookshelf ID: NBK278992PMID: 25905224

Views

  • PubReader
  • Print View
  • Cite this Page

LINKS TO WWW.ENDOTEXT.ORG

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...