Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Clin Invest ; 130(12): 6238-6241, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33196463

ABSTRACT

Some germ cell tumors (GCTs) in men develop into hematologic malignancies; however, the clonal origins of such malignancies remain unknown. In this issue of the JCI, Taylor, Donoghue, et al. unravel the clonal relationship between primary mediastinal nonseminomas (PMNs) and hematologic somatic-type malignancies (HSTMs). Whole-exome sequencing was used to construct phylogenetic trees of the PMNs and the ensuing HSTM clones. HSTMs were derived from multiple distinct clones not detected within the PMNs. Clones from PMNs and HSTMs shared a common precursor, arguably an embryonal carcinoma cell resulting from a reprogrammed primordial germ cell from the thymus. Mutational and copy number variation analysis of a large cohort of patients with PMNs also demonstrated a high prevalence of TP53 mutations not found in testicular nonseminomas. These data likely explain why patients with PMNs are frequently resistant to platinum-based chemotherapy and provide TP53 mutations as potential targets.


Subject(s)
Hematologic Neoplasms , Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , DNA Copy Number Variations , Humans , Male , Neoplasms, Germ Cell and Embryonal/genetics , Phylogeny
2.
Crit Rev Oncol Hematol ; 156: 103140, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33142194

ABSTRACT

BACKGROUND AND AIMS: Sacrococcygeal teratoma (SCT) is a rare extragonadal germ cell tumour mostly diagnosed during infancy and early childhood. Neonatal SCTs are mostly mature, but can also contain immature and/or malignant components. Recurrence of an SCT alters prognosis, especially when it is malignant, of which its mechanism is not yet fully understood. This study is a review and meta-analysis of the literature on malignant recurrences after an initially mature SCT. METHODS: A literature search was performed to identify studies describing children with SCT and presenting specific information on histology of the initial tumour as well as the recurrence. Random effect models for mature recurrence and malignant recurrence after an initially mature SCT were employed to pool study-specific percentages in order to estimate an overall percentage and its associated 95 % confidence intervals (CI). Inverse variance method, which gives more weight to larger studies, was used to pool outcomes for the different studies. RESULTS: A total of 22 articles, comprising 1516 patients with SCT, were included in the meta-analysis. The pooled proportions of mature and malignant recurrences after mature SCT were 3 % (95 % CI 1-4 %) and 5% (95 % CI 3-6 %), respectively. Fifty-seven (56 %) of a total of 102 recurrences after resection of an initially mature SCT were malignant, mostly yolk sac tumour (YST). Many recurrences occurred within 1-6 years, however some occurred as long as 20 years after initial diagnosis. CONCLUSIONS: A substantial number of recurrences of mature SCT present as a malignant tumour. Overlooking malignant components on initial pathological evaluation and the progression of mature SCT cells to malignant cells may play a role. Treatment of mature SCTs with resection alone requires thorough follow-up of at least 6 years. Future research is needed to determine whether SCTs with malignant microfoci should be treated or followed-up differently from mature or immature SCTs. In addition, the value of serum biomarkers in follow-up after SCT needs to be further evaluated.


Subject(s)
Neoplasms, Germ Cell and Embryonal , Teratoma , Child , Child, Preschool , Humans , Neoplasm Recurrence, Local , Prognosis , Sacrococcygeal Region , Teratoma/diagnosis
3.
Cancers (Basel) ; 12(10)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050470

ABSTRACT

Novel treatment options are needed for testicular germ cell tumor (TGCT) patients, particularly important for those showing or developing cisplatin resistance, the major cause of cancer-related deaths. As TGCTs pathobiology is highly related to epigenetic (de)regulation, epidrugs are potentially effective therapies. Hence, we sought to explore, for the first time, the effect of the two most recently FDA-approved HDAC inhibitors (HDACis), belinostat and panobinostat, in (T)GCT cell lines including those resistant to cisplatin. In silico results were validated in 261 patient samples and differential expression of HDACs was also observed across cell lines. Belinostat and panobinostat reduced cell viability in both cisplatin-sensitive cells (NCCIT-P, 2102Ep-P, and NT2-P) and, importantly, also in matched cisplatin-resistant subclones (NCCIT-R, 2102Ep-R, and NT2-R), with IC50s in the low nanomolar range for all cell lines. Treatment of NCCIT-R with both drugs increased acetylation, induced cell cycle arrest, reduced proliferation, decreased Ki67 index, and increased p21, while increasing cell death by apoptosis, with upregulation of cleaved caspase 3. These findings support the effectiveness of HDACis for treating TGCT patients in general, including those developing cisplatin resistance. Future studies should explore them as single or combination agents.

4.
Epigenomics ; 12(18): 1579-1592, 2020 09.
Article in English | MEDLINE | ID: mdl-32957806

ABSTRACT

Aim: Characterize DNA methyltransferases/demethylases expression in testicular germ cell tumors (TGCTs). Methods:In silico analysis of TCGA database, assessment of transcript levels of most relevant enzymes in four TGCT cell lines and validation in patient cohort (real-time quantitative polymerase chain reaction; immunohistochemistry). Results:DNMT3A, DNMT3B and TET2 were the most differentially expressed between seminomas (SEs) and nonseminomas (NSs). DNMT3B was significantly overexpressed in NS-related cell lines, and the opposite was found for TET2. Significantly higher DNMT3A/B mRNA expression was observed in NS, indicating a role for de novo methylation in reprogramming. Significantly higher TET2 protein expression was observed in SEs, suggesting active demethylation contributes for SE hypomethylated state. More differentiated histologies disclosed distinct expression patterns. Conclusion: DNA-modifying enzymes are differentially expressed between TGCT subtypes, influencing reprogramming and differentiation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins/metabolism , Neoplasms, Germ Cell and Embryonal/enzymology , Proto-Oncogene Proteins/metabolism , Testicular Neoplasms/enzymology , Adolescent , Adult , Cell Line, Tumor , Computer Simulation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Dioxygenases , Humans , Male , Middle Aged , Neoplasms, Germ Cell and Embryonal/classification , Neoplasms, Germ Cell and Embryonal/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Testicular Neoplasms/classification , Testicular Neoplasms/genetics , Young Adult , DNA Methyltransferase 3B
5.
Biomark Med ; 14(9): 785-793, 2020 06.
Article in English | MEDLINE | ID: mdl-32677456

ABSTRACT

Aim: To determine if neoadjuvant chemoradiotherapy (nCRT) affects p53 and SOX2 expression in esophageal adenocarcinoma (EAC). Materials & methods: Comparison of p53 and SOX2 expression in 100 paired pre- and post-nCRT EAC samples. Results: Aberrant p53 was largely concordant (75/83, 90%), while 13/18 (72%) pre-nCRT samples with wild-type (WT) p53 staining, showed aberrant staining in paired post-nCRT samples. Similarly, 31/45 (69%) with previous WT SOX2 showed SOX2 loss in paired post-nCRT samples, whereas aberrant SOX2 loss was concordant in 50/55 (91%) cases. The prognostic values of both markers regarding survival differ before and after nCRT. Conclusion: Aberrant expression of p53 and SOX2 staining in EAC tissue is unaffected by nCRT. Conversely, the WT-staining pattern frequently changed to aberrant expression.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Chemoradiotherapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/therapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/diagnosis , Aged , Esophageal Neoplasms/diagnosis , Female , Humans , Middle Aged , Neoadjuvant Therapy , Prognosis , Treatment Outcome
7.
United European Gastroenterol J ; 7(2): 261-269, 2019 03.
Article in English | MEDLINE | ID: mdl-31080611

ABSTRACT

Background: In early (T1) oesophageal adenocarcinoma (OAC), the histological profile of an endoscopic resection specimen plays a pivotal role in the prediction of lymph node metastasis and the potential need for oesophagectomy with lymphadenectomy. Objective: To evaluate the inter-observer agreement of the histological assessment of submucosal (pT1b) OAC. Methods: Surgical and endoscopic resection specimens with pT1b OAC were independently reviewed by three gastrointestinal pathologists. Agreement was determined by intraclass correlation coefficient for continuous variables, and Fleiss' kappa (κ) for categorical variables. Bland-Altman plots of the submucosal invasion depth were made. Results: Eighty-five resection specimens with pT1b OAC were evaluated. The agreement was good for differentiation grade (κ=0.77, 95% confidence interval (CI) 0.68-0.87), excellent for lymphovascular invasion (κ=0.88, 95% CI 0.76-1.00) and moderate for submucosal invasion depth using the Paris and Pragmatic classifications (κ=0.60, 95% CI 0.49-0.72 and κ=0.42, 95% CI 0.33-0.51, respectively). Systematic mean differences between pathologists were detected for the measurement of submucosal invasion depth, ranging from 297 µm to 602 µm. Conclusions: A substantial discordance was found between pathologists for the measurement of submucosal invasion depth in pT1b OAC. Differences may lead to an over- or underestimation of the lymph node metastasis risk, with grave implications for the treatment strategy. Review by a second gastrointestinal pathologist is recommended to improve differentiating between a favourable and an unfavourable histological profile.


Subject(s)
Adenocarcinoma/diagnosis , Esophageal Neoplasms/diagnosis , Adenocarcinoma/therapy , Adult , Aged , Aged, 80 and over , Esophageal Neoplasms/therapy , Esophagoscopy , Female , Follow-Up Studies , Histocytochemistry/methods , Histocytochemistry/standards , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Observer Variation , Pathologists
8.
Asian J Androl ; 17(3): 381-93, 2015.
Article in English | MEDLINE | ID: mdl-25791729

ABSTRACT

Malignant testicular germ cell tumors (TGCT) are the most frequent cancers in Caucasian males (20-40 years) with an 70% increasing incidence the last 20 years, probably due to combined action of (epi)genetic and (micro)environmental factors. It is expected that TGCT have carcinoma in situ(CIS) as their common precursor, originating from an embryonic germ cell blocked in its maturation process. The overall cure rate of TGCT is more than 90%, however, men surviving TGCT can present long-term side effects of systemic cancer treatment. In contrast, men diagnosed and treated for CIS only continue to live without these long-term side effects. Therefore, early detection of CIS has great health benefits, which will require an informative screening method. This review described the etiology and early pathogenesis of TGCT, as well as the possibilities of early detection and future potential of screening men at risk for TGCT. For screening, a well-defined risk profile based on both genetic and environmental risk factors is needed. Since 2009, several genome wide association studies (GWAS) have been published, reporting on single-nucleotide polymorphisms (SNPs) with significant associations in or near the genes KITLG, SPRY4, BAK1, DMRT1, TERT, ATF7IP, HPGDS, MAD1L1, RFWD3, TEX14, and PPM1E, likely to be related to TGCT development. Prenatal, perinatal, and postnatal environmental factors also influence the onset of CIS. A noninvasive early detection method for CIS would be highly beneficial in a clinical setting, for which specific miRNA detection in semen seems to be very promising. Further research is needed to develop a well-defined TGCT risk profile, based on gene-environment interactions, combined with noninvasive detection method for CIS.


Subject(s)
Carcinoma in Situ/diagnosis , Carcinoma in Situ/etiology , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/etiology , Testicular Neoplasms/diagnosis , Testicular Neoplasms/etiology , Adult , Carcinoma in Situ/genetics , Early Detection of Cancer , Gene-Environment Interaction , Genome-Wide Association Study , Humans , Male , Neoplasms, Germ Cell and Embryonal/genetics , Polymorphism, Single Nucleotide/genetics , RNA, Messenger/analysis , Risk Factors , Semen/chemistry , Testicular Neoplasms/genetics
9.
Mod Pathol ; 27(9): 1255-1266, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24457464

ABSTRACT

Testicular germ cell cancer develops from premalignant intratubular germ cell neoplasia, unclassified cells that are believed to arise from failure of normal maturation of fetal germ cells from gonocytes (OCT4(+)/MAGEA4(-)) into pre-spermatogonia (OCT4(-)/MAGEA4(+)). Intratubular germ cell neoplasia cell subpopulations based on stage of germ cell differentiation have been described, however the importance of these subpopulations in terms of invasive potential has not been reported. We hypothesized that cells expressing an immature (OCT4(+)/MAGEA4(-)) germ cell profile would exhibit an increased proliferation rate compared with those with a mature profile (OCT4(+)/MAGEA4(+)). Therefore, we performed triple immunofluorescence and stereology to quantify the different intratubular germ cell neoplasia cell subpopulations, based on expression of germ cell (OCT4, PLAP, AP2γ, MAGEA4, VASA) and proliferation (Ki67) markers, in testis sections from patients with preinvasive disease, seminoma, and non-seminoma. We compared these subpopulations with normal human fetal testis and with seminoma cells. Heterogeneity of protein expression was demonstrated in intratubular germ cell neoplasia cells with respect to gonocyte and spermatogonial markers. It included an embryonic/fetal germ cell subpopulation lacking expression of the definitive intratubular germ cell neoplasia marker OCT4, that did not correspond to a physiological (fetal) germ cell subpopulation. OCT4(+)/MAGEA4(-) cells showed a significantly increased rate of proliferation compared with the OCT4(+)/MAGEA4(+) population (12.8 versus 3.4%, P<0.0001) irrespective of histological tumor type, reflected in the predominance of OCT4(+)/MAGEA4(-) cells in the invasive tumor component. Surprisingly, OCT4(+)/MAGEA4(-) cells in patients with preinvasive disease showed significantly higher proliferation compared to those with seminoma or non-seminoma (18.1 versus 10.2 versus 7.2%, P<0.05, respectively). In conclusion, this study has demonstrated that OCT4(+)/MAGEA4(-) cells are the most frequent and most proliferative cell population in tubules containing intratubular germ cell neoplasia, which appears to be an important factor in determining invasive potential of intratubular germ cell neoplasia to seminomas.


Subject(s)
Antigens, Neoplasm/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Seminiferous Tubules/pathology , Testicular Neoplasms/metabolism , Adult , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Proliferation , Child , Fluorescent Antibody Technique, Indirect , Germinoma/metabolism , Germinoma/pathology , Humans , Immunohistochemistry , Infant , Male , Neoplasm Invasiveness , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/metabolism , Seminoma/pathology , Spermatogonia/metabolism , Testicular Neoplasms/pathology , Testis/embryology , Young Adult
10.
BMC Med Genet ; 13: 108, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23157850

ABSTRACT

BACKGROUND: The presence of the Y-chromosome or Y chromosome-derived material is seen in 4-60% of Turner syndrome patients (Chromosomal Disorders of Sex Development (DSD)). DSD patients with specific Y-chromosomal material in their karyotype, the GonadoBlastoma on the Y-chromosome (GBY) region, have an increased risk of developing type II germ cell tumors/cancer (GCC), most likely related to TSPY. The Sex determining Region on the Y gene (SRY) is located on the short arm of the Y-chromosome and is the crucial switch that initiates testis determination and subsequent male development. Mutations in this gene are responsible for sex reversal in approximately 10-15% of 46,XY pure gonadal dysgenesis (46,XY DSD) cases. The majority of the mutations described are located in the central HMG domain, which is involved in the binding and bending of the DNA and harbors two nuclear localization signals. SRY mutations have also been found in a small number of patients with a 45,X/46,XY karyotype and might play a role in the maldevelopment of the gonads. METHODS: To thoroughly investigate the presence of possible SRY gene mutations in mosaic DSD patients, we performed next generation (deep) sequencing on the genomic DNA of fourteen independent patients (twelve 45,X/46,XY, one 45,X/46,XX/46,XY, and one 46,XX/46,XY). RESULTS AND CONCLUSIONS: The results demonstrate that aberrations in SRY are rare in mosaic DSD patients and therefore do not play a significant role in the etiology of the disease.


Subject(s)
Chromosomes, Human, Y/genetics , Sex-Determining Region Y Protein/genetics , Testis/abnormalities , Turner Syndrome/genetics , Adolescent , Child , Child, Preschool , DNA Mutational Analysis/methods , Female , Gonadoblastoma/genetics , Humans , Infant , Karyotype , Male , Mosaicism , Sex Determination Analysis , Sex-Determining Region Y Protein/metabolism , Sexual Development/genetics , Transcription Factors/genetics
11.
J Pathol ; 224(4): 431-3, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21725972

ABSTRACT

Human germ cell tumours comprise a heterogeneous group of neoplasms which, based on pathobiological, genetic and clinical characteristics, can be subdivided into different entities. One of these subgroups relates to the so-called spermatocytic seminomas, benign tumours only found in the testis, preferentially in elderly men. Various developmental models for this type of germ cell tumour have been proposed and it is clear that spermatocytic seminoma has a pathogenesis independent from that of seminoma. A recent study examining expression of spermatogonial markers shows that spermatocytic seminomas are a heterogeneous group of tumours, with a supposed difference in origin, ie the majority from A(pale) or B spermatogonia, and a minority from A(dark) spermatogonia. However, this does not exclude an earlier cell of origin, possibly explaining the unique properties of this type of human germ cell tumour, with various counterparts in animals.


Subject(s)
Seminoma/etiology , Testicular Neoplasms/etiology , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Gene Expression Profiling/methods , Humans , Male , Neoplasm Proteins/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2 , Repressor Proteins/metabolism , Seminoma/metabolism , Testicular Neoplasms/metabolism
12.
Pediatr Endocrinol Rev ; 9 Suppl 1: 519-24, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22423509

ABSTRACT

A subset of patients with disorders of sex development (DSD) is at risk for malignant germ cell tumors (GCTs). The degree of gonadal differentiation (or "testicularization" in the presence of a specific part of the Y chromosome), in combination with expression of embryonic germ cell markers, and (a) Y specific gene(s) related to cell-cycle control and proliferation, determines this risk. Incompletely matured Sertoli/granulosa cells are insufficiently capable of directing the normal mitotic block/meiotic induction germ cell program, and as a result, embryonic germ cells are delayed or blocked in their normal maturation process. Thereby, they remain pluripotent and gain increased mitotic and survival characteristics, being the first step in the pathogenesis of GCTs. The patient's underlying genetic defect and phenotype might be informative in assessing the degree of gonadal "testicularization" on a clinical basis. Current knowledge allows development of an informative cancer risk assessment of DSD patients.


Subject(s)
Disorders of Sex Development/complications , Disorders of Sex Development/therapy , Neoplasms/etiology , Disorders of Sex Development/epidemiology , Female , Follow-Up Studies , Genital Neoplasms, Male/epidemiology , Genital Neoplasms, Male/etiology , Gonads/surgery , Humans , Male , Neoplasms/epidemiology , Neoplasms, Germ Cell and Embryonal/epidemiology , Neoplasms, Germ Cell and Embryonal/etiology , Practice Guidelines as Topic , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...