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.

Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].

Show details

Telomerase Activity in Neuroblastomas: A New Molecular Marker for Treatment Stratification and Prognostic Grouping

and .

Introduction: Clinical Aspects and Molecular Biology of Neuroblastoma

Neuroblastoma, a tumour derived from neural crest cells, represents the third most common pediatric cancer (accounting for approximately 7-10% of all childhood malignancies) and is the most common solid extracranial neoplasm of infancy and childhood, responsible for approximately 1-5% of all childhood cancer deaths. The clinical hallmark of neuroblastoma is heterogeneity, associated with a wide variety of likelihood of tumor progression. The most significant clinical predictors of outcome are age and stage, although an assessment of the patient's prognosis solely on the basis of clinical parameters is limited due to diverse biological tumor behavior and subsequent survival rates, even at distinct clinical stages.1,2 Therefore, the heterogeneity of this tumor entity requires cellular and molecular markers in order to distinguish the different biological characteristics. Established molecular markers such as MYCN copy number and loss of heterozygosity for chromosome 1p3236 may help predicting poor outcome.37 Based on the age-related prevalence and distinguishing genetic features of biologically favourable and unfavourable tumours, Brodeur et al8 proposed that NB are at least two or three diseases with distinct clinical and biological characteristics (Table 1). However, there is evidence that traditional prognostic parameters, such as analysis of MYCN amplification, which is considered an essential component of disease evaluation and treatment stratification, do not ensure completely accurate prognostic grouping.9–11 Identifying new prognostic markers can increase the accuracy of risk assessment and can also identify biologically relevant targets for developing new therapies.

Telomerase and Telomeres

A promising new marker is telomerase, a multicomponent ribonucleoprotein enzyme, which has been implicated in cell immortalization and tumorigenesis in almost all human tumors, including neuroblastomas.12–17 Telomerase is a ribonucleoprotein polymerase which uses an internal RNA component as a template to synthesize telomeric DNA directly onto the ends of chromosomes.18,19 These repetetive sequences are considered to be important in the protection and replication of chromosomes. Cells without telomerase activity display progressive shortening of telomeric repeats with each cell division, because lagging-strand DNA synthesis at the very end of linear chromosomes cannot be completed, a phenomenon generally referred to as the end-replication problem.20–22 Telomere shortening contributing to genetic instability is believed to be the primary signal for senescence mediated by tumor suppressor genes, such as p53 and Rb.23–26 Induction of telomerase activity in cells that have bypassed this control mechanism could give rise to clonal immortality by compensating for the loss of telomeric DNA and thus maintaining telomere length.27,28 Telomere length itself has not proven to be a good indicator of malignancy, since both stabilization and extension caused by mechanisms other than telomerase activation have been reported.29–34

Recently, the catalytic subunit hTERT (or hTRT, hEST2, TP2) of human telomerase was cloned and detection of hTERT expression by RT-PCR revealed a strong correlation with telomerase activity by the TRAP assay in the majority of tumors so far examined.35–37 Other components of the the telomerase holoenzyme complex, such as human telomerase RNA (hTR) and telomerase protein 1 (TP1, TLP1, hTEP1) seem to be expressed in both normal and tumor tissues, and expression levels of these genes revealed no or only limited correlation with telomerase activity.38–40

What Is the Role of Telomerase in Neuroblastomas?

Previous studies proposed that neuroblastomas with low telomerase activity might consist of cells that completely failed to repress telomerase activity during development, whereas tumors with high telomerase activity are likely to be derived from an immortalization event in a single cell.15,29,41,42 This condition renders clinical implications, since high telomerase activity is supposed to be accompanied by several genetic alterations and poor prognosis, whereas low or even absent telomerase activity in neuroblastomas coincides with good prognosis, and eventually, spontaneous regression.15,29 Exact prognostic implications on the basis of this two-entity model have so far been hampered by insufficient distinction of activity levels due to positivity for telomerase activity in the vast majority of neuroblastomas.29,41

In our study, a total of 133 neuroblastomas of all stages were analyzed in blind-trial fashion by a modified nonradioactive and semiquantitative TRAP (telomeric repeat amplification protocol) assay. Telomerase activity (TA) was present in 39 (29%) tumor tissues including 4/24 (17%) stage 4S neuroblastomas, 25/41 (61%) stage 4 neuroblastomas, 8/23 (35%) stage 3 neuroblastomas, 0/13 (0%) stage 2 neuroblastomas, and 2/32 (6%) stage 1 neuroblastomas. Of the 104 tumors resected initially without cytotoxic pretreatment, 31 showed TA (29%). After cytotoxic pretreatment (either initially or after tumor recurrence), TA was present in 8/29 tumors (28%). For 128/133 tumors included in this study, MYCN analysis was available and relevant MYCN amplification >3 copies43 could be observed in 21/128 (16%) samples. Data on LOH analysis of chromosome arm 1p were available for a subset of tumors (Table 2).

Correlation of TRAP and Survival for all Stages Combined

The correlation of TA status determined by the TRAP assay with the clinical data of 133 patients included in the German Neuroblastoma Trial was established under simple-blind study conditions to prevent any subjective impact on the estimation of TA levels. Clinical outcome was significantly correlated with the presence of TA distinguishing between TA positive (without further discrimination of low and high grade TA) and negative tumors. For all tumors (n=133), 5-year event-free survival (EFS) for 94 telomerase-negative tumors was 0.86 ± 0.04 versus 0.25 ± 0.08 for 39 telomerase- positive tumors at p<0.0001. 5-year overall survival (OS) for telomerase negative tumors was 0.93 ± 0.03 versus 0.34 ± 0.09 for telomerase positive tumors at p<0.0001. For initial samples (n=104), 5-year EFS for 73 telomerase-negative tumors was 0.92 ± 0.03 versus 0.33 ± 0.10 for 31 telomerase-positive tumors at p<0.0001 (Fig. 1A). 5-year OS for telomerase-negative tumors was 0.97 ± 0.02 versus 0.42 ± 0.10 for telomerase-positive tumors at p<0.0001 (Fig. 1B). Numbers for EFS and OS for localized disease (stage 1,2,3), stage 4, and stage 4S are summarized in Table 2.

Figure 1. (A) Event-free survival (EFS) and (B) overall survival (Survival) in 104 initial neuroblastomas prior to chemotherapy.

Figure 1

(A) Event-free survival (EFS) and (B) overall survival (Survival) in 104 initial neuroblastomas prior to chemotherapy. Telomerase activity (TA) negative, n=73; TA positive, n=31. p<0.0001.

Comparison of TA with MYCN Status

MYCN status was available for a total of 128 patients. The tumors of 107 patients had MYCN copy numbers £3. Patients with TA-positive tumors and MYCN copy numbers £3 in their tumors (n=22) taken before cytotoxic treatment were found to have a 5-year EFS of 0.31 ± 0.11, whereas those with TA-negative tumors (n=85) reached a 5-year EFS rate of 0.87 ± 0.04 (p<0.0001) (Fig. 2).

Figure 2. (A) Event-free survival (EFS) and overall survival (Survival) in relation to the MYCN status in 107 neuroblastomas with MYCN copy numbers £3.

Figure 2

(A) Event-free survival (EFS) and overall survival (Survival) in relation to the MYCN status in 107 neuroblastomas with MYCN copy numbers £3. TA negative, n=85; TA positive, n=22. p<0.0001.

Correlation Between TA and Survival by Age at Diagnosis (£ 1 year vs. >1 year)

OS and EFS were 0.90 ± 0.04 and 0.80 ± 0.05, respectively, for 63 patients < 1 year of age at diagnosis, compared to 0.64 ± 0.06 and 0.58 ± 0.06, respectively, for 70 patients > 1 year of age at diagnosis (p=0.0004 and p=0.0056, respectively). For patients < 1 year of age at diagnosis, OS and EFS were 0.96 ± 0.03 and 0.88 ± 0.05 for 53 TA-negative tumors, and 0.50 ± 0.16 and 0.30 ± 0.14, respectively, for 10 TA-positive tumors (p<0.0001). For patients > 1 year of age at diagnosis, OS and EFS were 0.87 ± 0.06 and 0.80 ± 0.06 for 41 TA-negative tumors, and 0.19 ± 0.08 and 0.17 ± 0.08, respectively, for 29 TA-positive tumors (p<0.0001).

Comparison of TA in TRAP Assay with Gene Expression of Telomerase Components

Catalytic Protein Subunit hTERT

From 75 neuroblastomas, frozen sections were made for extraction of both protein and RNA. For analysis, quantitative amounts of hTERT, hTR and hTEP1 gene expressions standardized on GAPDH expression were grouped on a score system ranging from 07 (see Material & Methods for details). On quantitative RT-PCR analysis on the LightCycler instrument, hTERT expression was found in 37 of 75 (49%) neuroblastomas. For correlating hTERT expression with TA in the TRAP assay, hTERT expression levels of each neuroblastoma sample was compared to hTERT expression of a dilution series (see Material & Methods) of a telomerase-positive (TRAP positive) Ewing tumor cell line. 22/75 (29.3%) neuroblastomas were positive for both telomerase activity and hTERT mRNA and 37/75 (49.3%) were negative for both TA and hTERT mRNA. One neuroblastoma (1.3%) was only positive for TA but not for hTERT mRNA, and 15 (20%) neuroblastomas were only positive for hTERT mRNA, but not for TA. Among the 22 neuroblastomas that were positive for both TA and hTERT mRNA, there was a significant statistical correlation (p<0.001) between quantitative expression levels of TA and hTERT mRNA. That is, 10 samples with intermediate or high TA levels (score 2 or 3 on a scale of 03) displayed intermediate or high hTERT mRNA levels (score 4 on a scale of 07) and, vice versa, 11 samples with low TA levels (score 1) displayed low hTERT mRNA levels (score 2 or 3). A single case of a stage 4 neuroblastoma after cytotoxic pretreatment revealed low TA (score 1) and high hTERT mRNA expression (score 5). Among the 15 samples which were positive for hTERT mRNA but negative for TA, almost all cases (n=14) had hTERT levels that were minimal (score 1). These minimal (score 1) hTERT levels always had values significantly lower than neuroblastomas with low TA and low hTERT. Furthermore, minimal hTERT levels were found in up to 50% of analyzed tumor-free tissues such as normal breast or normal liver (data not shown). However, there was a single case with no TA in TRAP assay which repeatedly displayed high hTERT mRNA levels by RT-PCR (score 4). This sample was from a stage 1 neuroblastoma of a 32-month-old (age at diagnosis) child with relapsefree clinical follow-up for 46 months. Further investigations revealed no Taq polymerase inhibitors in the TRAP assay and no MYCN amplification in this sample. Using cRNA sense and antisense probes for Northern Blots on RNA from a TA-positive Ewing's tumor and neuroblastoma cell line, the specificity of hTERT expression detected by our RT-PCR system was confirmed.

Based on 67 neuroblastomas without prior cytotoxic treatment, 5-year EFS for 37 hTERT negative tumors (score 0) was 0.85 ± 0.06, 1.0 ± 0.0 for 11 tumors with minimal (score 1) hTERT, 0.38 ± 0.17 for 9 tumors with low (score 23) hTERT, and 0.44 ± 0.17 for 10 tumors with intermediate to high (score 4) hTERT at p<0.0001. 5-year OS for 37 hTERT-negative tumors (score 0) was 0.93 ± 0.05, 1.0 ± 0.0 for 11 tumors with minimal (score 1) hTERT, 0.73 ± 0.17 for 9 tumors with low (score 23) hTERT, and 0.42 ± 0.17 for 10 tumors with intermediate to high (score 4) hTERT at p<0.0001.

RNA Component hTR

Positive hTR RNA expression at different levels was found in 74/75 neuroblastomas examined, and expression of hTR mRNA was also significantly correlated with TA (p<0.001). Based on 66 neuroblastomas without prior cytotoxic treatment, 5-year EFS for 34 tumors with negative (n=1) or low hTR expression (n=33) was 0.93 ± 0.05, 0.54 ± 0.11 for 24 tumors with intermediate hTR, and 0.54 ± 0.20 for 8 tumors with high hTR at p=0.0005. 5-year OS for 34 tumors with negative or low hTR was 0.96 ± 0.04, 0.75 ± 0.10 for 24 tumors with intermediate hTR, and 0.54 ± 0.20 for 8 tumors with high hTR at p=0.0012. From 4 of these 75 cases, slides were made from paraffin-embedded tissue for hTR in situ hybridization (Fig. 3), which revealed a strong correlation with both TA and hTR mRNA expression by RT-PCR. Interestingly, in a TA-negative and hTERT-negative (both by RTPCR and immunohistochemistry) neuroblastoma with intermixed differentiated ganglion cells, only the ganglion cells, but not the undifferentiated neuroblastic cells, displayed weak hTR in situ hybridization signals.

Figure 3. Telomerase activity (TRAP assay) and hTR RNA expression (in situ hybridization) in neuroblastomas.

Figure 3

Telomerase activity (TRAP assay) and hTR RNA expression (in situ hybridization) in neuroblastomas. Lane A: intermediate telomerase activity; lane B: high telomerase activity; lane C: no telomerase activity.

Telomerase-Associated Protein hTEP1

Positive hTEP1 mRNA expression was found in 90% of the samples, but there was no correlation between TA and HTEP1 expression levels. High HTEP1 mRNA levels were also found in two-thirds of the TA-negative neuroblastomas, and at the same time low HTEP1 mRNA levels were also associated with some TA-positive neuroblastomas with high TA levels.

hTERT mRNA Expression in ParaffinEmbedded Tissues

hTERT expression by RT-PCR from fresh-frozen and paraffin-embedded tissues was compared in 16 selected cases from the archives. For assessing tissue sample preservation, RNA from these 16 formalin-fixed and paraffin-embedded neuroblastoma tissues was initially amplified by RT-PCR (TITAN system, Roche Diagnostics, Mannheim, Germany) for the GAPDH gene. In 11/16 cases, suitable RNA for RT-PCR was available. These 11 cases included five stage 4 neuroblastomas (with intermediate or high TA and hTERT mRNA expression in corresponding fresh-frozen tissue), three stage 3 neuroblastomas (with intermediate or high TA and hTERT mRNA expression in corresponding fresh-frozen tissue) and three stage 1 or 2 neuroblastoma (with neither TA nor hTERT mRNA expression in corresponding fresh-frozen tissue). Comparison of hTERT mRNA expression from the corresponding fresh-frozen tissues revealed similar expression patterns as with the paraffin-embedded tissue. hTERT mRNA expression in these two cases was correlated with fatal outcome, whereas the hTERT mRNA-negative stage 1 neuroblastoma case is relapse and disease free after 54 months of follow-up.

Multivariate Analysis: Is TA an Independent Prognostic Marker?

Cox regression model for event-free survival was built on data of 122 patients from which the most covariates were available. Telomerase activity (negative versus positive), stage (1,2,3,4S vs. 4), serum LDH (normal vs. increased), MYCN copy number (£3 versus >3) and age (continuous) were entered as covariates. The model revealed only telomerase activity and stage, but not serum LDH, MYCN amplification or age as independent prognostic factors (Table 3).

Conclusions: Telomerase Activity as New Molecular Marker for Treatment Stratification and Prognostic Grouping

Telomerase activity was demonstrated to be significantly correlated with poor prognosis in neuroblastoma. In our study according to stage classification, multivariate analysis revealed only TA and stage as independent prognostic factors, whereas serum LDH, MYCN status, and age were not. The high accuracy with which tumors prone to unfavorable outcome can be identified makes TA a reliable prognostic tool in neuroblastoma. Although it is not proven so far that TA will not change under cytotoxic pretreatment, our data suggest that TA analysis is suitable in all neuroblastomas irrespective of their pretreatment status: comparing the number of TA-positive tumors among all neuroblastomas in this study (n=133) vs. the subset of initial samples without pretreatment (n=104), we found no difference between these two groups (39/133 equals 29.3% of all neuroblastomas vs. 31/104 equals 29.8% of initial neuroblastomas).

One particular strength of TA as a prognostic marker is the differentiation of good and poor outcome in putative “favorable” clinical or biological subtypes of NB patients. These include, for example, patients in the age group of 01 years at diagnosis and patients with nonamplified MYCN in their tumors.44 For all 63 patients < 1 year of age at diagnosis in this study combined, OS was ∼90% and EFS was ∼80%. However, for 10 of these patients with TA positive tumors, OS dropped down to ∼50%, and EFS down to ∼30%. The only favorable cases in this subset of TA-positive tumors were congenital neuroblastomas with minimal TA. These cases may represent NB derived from neuroblasts which retain the expression of fetal levels of TA because of a failure to repress telomerase, as suggested by Hiyama's NB development model.17 In the putative favorable group of patients with nonamplified MYCN, TA once again identifies good and poor prognostic subsets: for 86 initial samples without cytotoxic pretreatment, with MYCN < 3 copies, OS was ∼87% and EFS was ∼80%. However, for 20 of these patients with TA-positive tumors, OS dropped down to ∼40% and EFS down to ∼31%.

Above all, the significant correlation of clinical outcome with presence of telomerase activity strongly recommends that the analysis of telomerase activity in individual tumors should be accorded a priority at least equal to that of the determination of established markers, such as MYCN and LOH of chromosome 1p, thereby facilitating risk-directed therapy.45

Molecular Mechanisms of Telomerase Activity in Neuroblastoma

The recent cloning of the telomerase holoenzyme components hTERT, hTR and HTEP1 allows a more precise definition of the molecular mechanisms of telomerase activity in neuroblastoma. hTERT has been identified as the catalytic subunit of telomerase, and expression of hTERT mRNA is usually observed at high levels in telomerase-positive cancer cell lines and malignant tumors, but not in adjacent normal tissues. Furthermore, a strong correlation was found between telomerase activity and hTERT mRNA expression in different tumor types.39,46,48 Recently, Hiyama et al49 found in a smaller series of neuroblastoma (35 cases) hTERT mRNA expression in all 13 tumors with high TA and in 5/23 tumors with low or undetectable TA. Using a kinetic rather than end-point approach for quantification of PCR products, we could demonstrate a significant correlation between distinct levels of TA and hTERT mRNA expression for the majority of neuroblastomas in our study. Our finding that 14 neuroblastomas in this study displayed minimal expression of hTERT mRNA without detectable TA is in agreement with studies on other tumors which found that hTERT mRNA expression did not always give rise to TA.50 Recent studies by Xu et al47 provide new insights into the regulatory control of telomerase activity at the molecular level: during differentiation of HL60 promyelocytic leukaemic cells, suppression of TA is preceded by a down-regulation of hTERT mRNA, which is achieved through inhibition of its transcription. Furthermore, Xu et al showed that the onset of reduction of hTERT expression during cellular differentiation was at least in part independent of cell proliferation status and suppression of TA was unrelated to hTR and HTEP1 expression in differentiation of HL60 cells.

Diagnostic Procedures for Telomerase Detection

In the case of limited material from very small biopsy samples or formalin-fixed and paraffin-embedded tissues, for example archived material, immunohistochemical detection of hTERT by antihTERT antibody-reactive protein in tissue sections may be an indicator of telomerase activity in the tumor cells. However, so far there are only few reliable anti-hTERTantibodies for immunohistochemical detection of hTERT protein in paraffin-embedded tissues. At the moment, we do not recommend using these antibodies for diagnostic procedures in the routine setting as they require thorough standardization using several positive and negative controls (from which in corresponding fresh-frozen tissue the status of telomerase activity, the quantity of hTERT mRNA expression and hTERT protein expression by Western Blot should be known). For retrospective studies on archived material, RT-PCR to detect hTERT mRNA expression from paraffin-embedded tissues may be another option for indirect assessment of telomerase activity. To our knowledge, we have demonstrated for the first time that expression of hTERT mRNA can be determined even from paraffin-embedded tissues and that this expression seems to correlate with hTERT gene and protein expression as well as TA from corresponding fresh-frozen tumor tissue. For this analysis, however, one should keep in mind that RT-PCR from paraffin-embedded tissue derived RNA may not always allow accurate quantification of gene expression levels, which is an important limitation of directly deducing TA levels from hTERT expression. Prospective studies with larger case numbers should be performed to further evaluate the prognostic impact of hTERT RNA or protein detection in paraffin samples.

The Role of hTR and hTEP1

Focusing on the other gene components of the telomerase complex , different studies showed that the human telomerase RNA component hTR and telomerase protein HTEP1 mRNA are broadly expressed in both malignant and tumor-free normal tissue. Previous investigations furthermore revealed no significant correlation between telomerase activity and the expression of hTR or HTEP1 mRNA in the vast majority of human tumors.38–40 Other authors suggest that overexpression of hTR may be correlated with proliferative cell activity.51 So far, there is only limited data on hTR expression compared to telomerase activity in neuroblastomas. In a small series of 5 neuroblastomas and 1 ganglioneuroma, Maitra et al52 described a correlation between telomerase activity, MYCN amplification and hTR expression determined by in situ hybridization. In our series, we found a significant correlation between hTR mRNA expression levels and TA. Analysis of hTERT and hTR expression by quantitative real-time RT-PCR suggests that there is a threshold for the expression levels of these genes before TA is detectable in NB; this threshold which has to be exceeded before TA is present is probably higher for hTR as hTR mRNA expression was found in nearly all (74/75) NB by RT-PCR, but only 23 cases displayed TA. Furthermore, this finding supports the hypothesis that hTERT as the catalytic subunit of telomerase is essential for telomerase activity. On histologic sections made from paraffin-embedded tissues, NB with high TA and strong expression of hTERT and hTR by RT-PCR displayed strong nuclear staining by hTERT immunohistochemistry (IH) and hTR in situ hybridization (ISH) in more than 50% of the neuroblastic cells. TA-negative NB showed no or <1% nuclear staining for hTERT and some weak staining, in particular in ganglion cells, for hTR. Interestingly, these ganglion cells did not express hTERT protein in IH. In NB with low and intermediate TA, we found 1-10% or 10-50% positive nuclei with hTERT IH, respectively. Since there is evidence from other studies53 that differential regulation of TA and hTR may be controlled by independent mechanisms in a small subset of NB and NB cell lines, further studies are needed for analysis of regulatory pathways of TA in NB.

Detection of Telomerase Activity by the TRAP Assay: The Gold Standard

Although we found a significant correlation between TA determined by the TRAP and hTERT mRNA expression by quantitative RT-PCR, our data suggest that the TRAP assay is a more powerful molecular tool which could be complemented, but not be replaced, by analysis of hTERT expression in the clinical setting. Whereas the mere presence or absence of TA ­ irrespective of activity levels ­ is associated with either favorable or unfavorable clinical outcome, the interpretation of hTERT expression as a prognostic marker is much more difficult: first, accurate quantification of hTERT expression levels is necessary and requires correlation to a defined standard, because ­ as shown above ­ minimal hTERT levels are associated with a favourable prognosis, whereas low, intermediate or high hTERT levels predict unfavourable outcome. Second, accurate quantification requires special and expensive equipment such as new PCR systems for real-time detection of reaction kinetics and, furthermore, standardized reaction kits to permit the comparison of results between different laboratories.

As the molecular biology of neuroblastoma has led to a combined clinical and risk stratification, the search for new molecular markers for assessing the individual patient's prognosis is of vital importance, since there is evidence that analysis of MYCN amplification does not ensure completely accurate prognostic grouping,10 which unfortunately is also true for other traditional molecular and clinical markers. This study and previous investigations by our group and others suggest that analysis of telomerase activity by the TRAP assay is a powerful additional molecular tool for distinguishing neuroblastomas with good and poor prognosis. Along with other emerging markers such as gain of chromosome arm 17q,10 we propose that analysis of TA should be incorporated into the clinical investigation of each individual neuroblastoma case at the time of diagnosis.

Acknowledgments

The authors thank Christina Scheel, Dr. KarlLudwig Schäfer and Dr. Bernhard Heine for their cooperation in several studies on telomerase activity in neuroblastomas.

References

1.
Maris JM, Matthay KK. Molecular biology of neuroblastoma. J Clin Oncol. 1999;17:2264–2279. [PubMed: 10561284]
2.
Woods WG, Lemieux B, Tuchman M. Neuroblastoma represents distinct clinicalbiologic entities: A review and perspective from the Quebec Neuroblastoma Screening Project. Pediatrics. 1992;89:114–118. [PubMed: 1727993]
3.
Gallego S, Parareda A, Munell F. et al. Clinical relevance of molecular markers in neuroblastoma: Results from a single institution. Oncol Rep. 1999;6:891–896. [PubMed: 10373677]
4.
Erttmann R, Tafese T, Berthold F. et al. 10 years' neuroblastoma screening in Europe: preliminary results of a clinical and biological review from the Study Group for Evaluation of Neuroblastoma Screening in Europe (SENSE). Eur J Cancer. 1998;34:1391–1397. [PubMed: 9849422]
5.
Schwab M. Amplification of N-myc as a prognostic marker for patients with neuroblastoma. Semin Cancer Biol. 1993;4:13–18. [PubMed: 8448374]
6.
Christiansen H, Lampert F. Tumour karyotype discriminates between good and bad prognostic outcome in neuroblastoma. Br J Cancer. 1988;57:121–126. [PMC free article: PMC2246687] [PubMed: 3348945]
7.
Brodeur GM, Seeger RC, Schwab M. et al. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science. 1984;224:1121–1124. [PubMed: 6719137]
8.
Brodeur GM, Ambros P F F M. Biological aspects of neuroblastoma screening. Med.Pediatr.Oncol. 1998;31:394–400.
9.
Eggert A, Ikegaki N, Liu XG. et al. Prognostic and biological role of neurotrophin receptor TrkA and TrkB in neuroblastoma. Klin.Padiatr. 2000;212:200–205. [PubMed: 10994551]
10.
Bown N, Cotterill S, Lastowska M. et al. Gain of chromosome arm 17q and adverse outcome in patients with neuroblastoma. N Engl J Med. 1999;340:1954–1961. [PubMed: 10379019]
11.
Kawa K, Ohnuma N, Kaneko M. et al. Long-term survivors of advanced neuroblastoma with MYCN amplification: A report of 19 patients surviving diseasefree for more than 66 months. J Clin Oncol. 1999;17:3216–3220. [PubMed: 10506621]
12.
Poremba C, Scheel C, Hero B. et al. Telomerase activity and telomerase subunits gene expression patterns in neuroblastoma: a molecular and immunohistochemical study establishing prognostic tools for freshfrozen and paraffinembedded tissues. J Clin Oncol. 2000;18:2582–2592. [PubMed: 10893290]
13.
Schneider-Stock R, Epplen JT, Walter H. et al. Telomeric lengths and telomerase activity in liposarcomas. Mol Carcinog. 1999;24:144–151. [PubMed: 10078942]
14.
Umbricht CB, Sherman ME, Dome J. et al. Telomerase activity in ductal carcinoma in situ and invasive breast cancer. Oncogene. 1999;18:3407–3414. [PubMed: 10362362]
15.
Brinkschmidt C, Poremba C, Christiansen H. et al. Comparative genomic hybridization and telomerase activity analysis identify two biologically different groups of 4s neuroblastomas. Br J Cancer. 1998;77:2223–2229. [PMC free article: PMC2150403] [PubMed: 9649137]
16.
Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. Eur J Cancer. 1997;33:787–791. [PubMed: 9282118]
17.
Hiyama E, Hiyama K, Yokoyama T. et al. Correlating telomerase activity levels with human neuroblastoma outcomes. Nat Med. 1995;1:249–255. [PubMed: 7585042]
18.
Morin GB. The human telomere terminal transferase enzyme is a ribonucleoprotein that synthesizes TTAGGG repeats. Cell. 1989;59:521–529. [PubMed: 2805070]
19.
Kobayashi T, Clodi K, Yang Y. et al. Inhibition of telomerase activity correlates with a decrease in the RNA component of telomerase during differentiation. Int J Oncol. 1997;11:13–17. [PubMed: 21528174]
20.
Levy MZ, Allsopp RC, Futcher AB. et al. Telomere end-replication problem and cell aging. J Mol Biol. 1992;225:951–960. [PubMed: 1613801]
21.
Hiyama E, Hiyama K, Tatsumoto N. et al. Telomerase activity in human intestine. Int J Oncol. 1996;9:453–458. [PubMed: 21541534]
22.
Olovnikov AM. Principles of marginotomy in template synthesis of polynucleotides. Dokl Akad Nauk SSSR. 1971;201:1496–1499. [PubMed: 5158754]
23.
Harley CB. Telomere loss: mitotic clock or genetic time bomb? Mutat Res. 1991;256:271–282. [PubMed: 1722017]
24.
WynfordThomas D. Telomeres, p53 and cellular senescence. Oncol Res. 1996;8:387–398. [PubMed: 9114431]
25.
Blackburn EH. Structure and function of telomeres. Nature. 1991;350:569–573. [PubMed: 1708110]
26.
Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature. 1990;345:458–460. [PubMed: 2342578]
27.
Counter CM, Hirte HW, Bacchetti S. et al. Telomerase activity in human ovarian carcinoma [see comments] Proc Natl Acad Sci USA. 1994;91:2900–2904. [PMC free article: PMC43481] [PubMed: 8159676]
28.
De Lange T. Activation of telomerase in a human tumor. Proc Natl Acad Sci USA. 1994;91:2882–2885. [PMC free article: PMC43476] [PubMed: 8159672]
29.
Hiyama E, Hiyama K, Yokoyama T. et al. Correlating telomerase activity levels with human neuroblastoma outcomes. Nat Med. 1995;1:249–255. [PubMed: 7585042]
30.
Murnane JP, Sabatier L, Marder BA. et al. Telomere dynamics in an immortal human cell line. EMBO J. 1994;13:4953–4962. [PMC free article: PMC395436] [PubMed: 7957062]
31.
Brodeur GM, Maris JM, Yamashiro DJ. et al. Biology and genetics of human neuroblastomas. J Pediatr Hematol Oncol. 1997;19:93–101. [PubMed: 9149737]
32.
Bryan TM, Englezou A, Dalla PL. et al. Evidence for an alternative mechanism for maintaining telomere length in human tumors and tumorderived cell lines. Nat Med. 1997;3:1271–1274. [PubMed: 9359704]
33.
Belair CD, Yeager TR, Lopez PM. et al. Telomerase activity: a biomarker of cell proliferation, not malignant transformation. Proc Natl Acad Sci USA. 1997;94:13677–13682. [PMC free article: PMC28365] [PubMed: 9391085]
34.
Hiyama E, Hiyama K, Yokoyama T. et al. Length of telomeric repeats in neuroblastoma: correlation with prognosis and other biological characteristics. Jpn J Cancer Res. 1992;83:159–164. [PMC free article: PMC5918785] [PubMed: 1555997]
35.
Takakura M, Kyo S, Kanaya T. et al. Cloning of human telomerase catalytic subunit (hTERT) gene promoter and identification of proximal core promoter sequences essential for transcriptional activation in immortalized and cancer cells. Cancer Res. 1999;59:551–557. [PubMed: 9973199]
36.
Ramakrishnan S, Eppenberger U, Mueller H. et al. Expression profile of the putative catalytic subunit of the telomerase gene. Cancer Res. 1998;58:622–625. [PubMed: 9485011]
37.
Meyerson M, Counter CM, Eaton EN. et al. hEST2, the putative human telomerase catalytic subunit gene, is upregulated in tumor cells and during immortalization. Cell. 1997;90:785–795. [PubMed: 9288757]
38.
Sumida T, Hamakawa H, Sogawa K. et al. Telomerase components as a diagnostic tool in human oral lesions. Int J Cancer. 1999;80:1–4. [PubMed: 9935220]
39.
Ito H, Kyo S, Kanaya T. et al. Expression of human telomerase subunits and correlation with telomerase activity in urothelial cancer. Clin Cancer Res. 1998;4:1603–1608. [PubMed: 9676833]
40.
Avilion AA, Piatyszek MA, Gupta J. et al. Human telomerase RNA and telomerase activity in immortal cell lines and tumor tissues. Cancer Res. 1996;56:645–650. [PubMed: 8564985]
41.
Hiyama E, Hiyama K, Ohtsu K. et al. Telomerase activity in neuroblastoma: is it a prognostic indicator of clinical behaviour? Eur J Cancer. 1997;33:1932–1936. [PubMed: 9516827]
42.
Reynolds CP, Zuo JJ, Kim NW. et al. Telomerase expression in primary neuroblastomas. Eur J Cancer. 1997;33:1929–1931. [PubMed: 9516826]
43.
Katzenstein HM, Bowman LC, Brodeur GM. et al. Prognostic significance of age, MYCN oncogene amplification, tumor cell ploidy, and histology in 110 infants with stage D(S) neuroblastoma: the pediatric oncology group experience A pediatric oncology group study. J Clin Oncol. 1998;16:2007–2017. [PubMed: 9626197]
44.
Brodeur GM, Ambros P F F M. Biological aspects of neuroblastoma screening. Med Pediatr Oncol. 1998;31:394–400.
45.
Healy KC. Telomere dynamics and telomerase activation in tumor progression: prospects for prognosis and therapy. Oncol Res. 1995;7:121–130. [PubMed: 8555645]
46.
Hisatomi H, Nagao K, Kanamaru T. et al. Levels of telomerase catalytic subunit mRNA as a predictor of potential malignancy. Int J Oncol. 1999;14:727–732. [PubMed: 10087321]
47.
Xu D, Gruber A, Björkholm M. et al. Suppression of telomerase reverse transcriptase (hTERT) expression in differentiated HL60 cells: Regulatory mechanisms. Br J Cancer. 1999;80:1156–1161. [PMC free article: PMC2362380] [PubMed: 10376966]
48.
Wada E, Hisatomi H, Moritoyo T. et al. Genetic diagnostic test of hepatocellular carcinoma by telomerase catalytic subunit mRNA. Oncol Rep. 1998;5:1407–1412. [PubMed: 9769378]
49.
Hiyama E, Hiyama K, Yokoyama T. et al. Rapid detection of MYCN gene amplification and telomerase expression in neuroblastoma. Clin Cancer Res. 1999;5:601–609. [PubMed: 10100712]
50.
Tahara H, Yasui W, Tahara E. et al. Immunohistochemical detection of human telomerase catalytic component, hTERT, in human colorectal tumor and nontumor tissue sections. Oncogene. 1999;18:1561–1567. [PubMed: 10102626]
51.
Hiyama T, Yokozaki H, Kitadai Y. et al. Overexpression of human telomerase RNA is an early event in oesophageal carcinogenesis. Virchows Arch. 1999;434:483–487. [PubMed: 10394881]
52.
Maitra A, Yashima K, Rathi A. et al. The RNA component of telomerase as a marker of biologic potential and clinical outcome in childhood neuroblastic tumors. Cancer. 1999;85:741–749. [PubMed: 10091748]
53.
Choi LM, Kim NW, Zuo JJ. et al. Telomerase activity by TRAP assay and telomerase RNA (hTR) expression are predictive of outcome in neuroblastoma. Med Pediatr Oncol. 2000;35:647–650. [PubMed: 11107138]
Copyright © 2000-2013, Landes Bioscience.
Bookshelf ID: NBK6022

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...