Skip to main content
  • Research article
  • Open access
  • Published:

The emerging clinical relevance of genomic profiling in neuroendocrine tumours

Abstract

Background

Neuroendocrine tumours (NETs) arise from hormone-producing or nervous system cells and can develop from anywhere in the body. They have heterogeneous origins from skin to gastrointestinal track and a complicated histology. Thus, there is an inevitable need for genomic profiling to determine the exact genetics of each tumour for prognosis and treatment strategies to overcome the disease’s complexity. For this purpose, next-generation-sequencing (NGS) is the most reliable methodology for both germ-line and somatic studies to make a clinical diagnosis. In this study, we analyse liquid biopsies, formalin fixed paraffin embedded (FFPE) tissues, and peripheral blood samples for their ability to provide information for actionability.

Methods

A customized multi-gene panel comprised of Succinate Dehydrogenase Complex Iron Sulfur Subunit B (SDHB), Succinate Dehydrogenase Complex Subunit C (SDHC), Cell Division Cycle 73(CDC73), Calcium Sensing Receptor (CASR), Platelet Derived Growth Factor Receptor Alpha (PDGFRA), Succinate Dehydrogenase Complex Flavoprotein Subunit A (SDHA), Ret Proto-Oncogene (RET), Succinate Dehydrogenase Complex Assembly Factor 2(SDHAF2), Menin 1(MEN1), Succinate Dehydrogenase Complex Subunit D (SDHD), MYC Associated Factor X (MAX) and Protein Kinase CAMP-Dependent Type I Regulatory Subunit Alpha (PRKAR1A) genes was constructed to assess multiple specimen types including: 3 liquid biopsies, 6 FFPE tissues, and 26 peripheral blood samples from 35 unique NET patients. Quality-control and bioinformatics analyses were performed using QCI-Analyze and QCI-Interpret.

Results

The three liquid biopsies and the 6 FFPE tissue samples were evaluated for somatic mutations; while the 26 peripheral blood samples were analysed using the germ-line pipeline. Five (55.6%) of the nine patients that were studied for somatic changes carried actionable mutations related to therapy sensitivities. Through the germ-line studies, we observed a 50% positivity rate for disease predisposition with 16 variants classified according to ACMG (American College of Medical Genetics) Standards and Guidelines.

Conclusions

Genomic profiling medicine is an emerging area of clinical oncology and has become crucial for disease and patient management by providing a precision approach; this is especially true for rare diseases including rare cancers such as NETs. Notably, this study emphasized the relevance of multiple distinctive biological sample types for use in the genetic testing of cancers to help with the choice of therapy to maximize the likelihood of a positive clinical outcome.

Peer Review reports

Background

Neuroendocrine tumours (NETs) are a relatively rare group of cancers which can arise from endocrine and nervous system cells. They can originate from multiple tissues and organs; most frequently from within the lungs, pancreas and intestine. Although the gastroenteropancreatic neuroendocrine tumours group (GEP-NET) is the major class which includes pancreatic neuroendocrine tumours and carcinoids, thymus, parathyroid and lung forms are also take place in rarer cases [1]. The definition and classification of NETs has changed over time due to their complexity [2, 3]. The International Agency for Research on Cancer (IARC) and the World Health Organization (WHO) have published a site-specific criteria and terminology guide to provide uniform classifications This organ-specific approach has aided more accurate nomenclature and grading. Moreover, it has provided us with a better understanding of NETs’ organ-specific characteristics and prognostic behaviours [4, 5]. Due to the fact that NETs can develop anywhere in the body and that the organ-tissue of origin plays an important role in prognosis and response to treatments, the patients’ and the tumour’s genetic backgrounds have become more important. The conventional treatment strategies included mainly surgery. However, it is now known that a multidisciplinary approach is necessary for a more personalized and efficient therapy taking into consideration of site of origin, potential of metastasis, grade and the genetic background of the patient. Finally, the heterogeneous origins of NETs lead to a very complicated tumour histology and multiple distinctive aetiologies [6, 7].

Although the vast majority of NETs are sporadic and caused by somatic mutations, germ-line genetic changes also play a critical role in disease inheritance and prognosis [8, 9]. Therefore, a patient-specific approach is a necessity for disease management [7, 10]. Next-generation sequencing (NGS) has become the most powerful methodology for the precise diagnosis, prognosis track and development of therapeutic strategies for many types of cancers. Through the development and testing of customized gene panels for various cancer types, it has been possible to provide precision medicine. In our centre we perform NGS from liquid biopsies - one of the most popular topics in oncology - on formalin fixed paraffin embedded (FFPE) tissues, and peripheral blood samples for the accurate genomic profiling of NETs to determine their hereditary predispositions and treatment actionability.

Methods

Demographic and clinical information

Thirty-five distinctive NET patients who were referred to our center for molecular genetic testing were recruited for this study. Patient distribution by their tumor site of origin is as follows; 74.3% (n = 26) foregut (including lung, pancreas, thymus, parathyroid, stomach and medullary thyroid cancer), 5.7% (n = 2) hindgut (rectum cancer), 17.1% (n = 6) other (given in Table 1) and 2.9% both foregut (pancreas cancer) and hindgut (rectum cancer). All participants signed an informed consent, and were enrolled in accordance with the ethical standards of the institutional ethical committee (Cukurova University Faculty of Medicine Non-Invasive Clinical Research Ethics Commission) and the Helsinki declaration. Starting material types were selected in accordance with the patients’ diagnoses, clinical background and familial history. Patients with any inherited syndromes suspected were excluded from the study. Demographic information for the patients is shown in Table 1.

Table 1 Demographic information of the patients, biological sample types, sites of origin and World Health Organization (WHO) classification of NETs

Sampling and DNA isolation

Liquid biopsy samples were collected via biological sampling tubes special for ccfDNA. Tumor sites were determined to obtain FFPE tissue sections. Somatic DNA from liquid biopsies (circulating cell-free DNA, ccfDNA) and FFPE tissues were isolated by manufacturer’s instructions with modifications [QIAamp Circulating Nucleic Acid Kit and DNA FFPE Tissue Kit (Qiagen, Germany)] [11, 12]. Genomic DNA from peripheral blood was isolated via instructions provided by the kit manufacturer [QIAamp DNA Blood Midi Kit (Qiagen, Germany)]. Fluorometric measurements were made using a Qubit 3.0 fluorometer to assess the quality and quantity of the isolated DNAs. The purified DNAs were then subjected to next-generation sequencing.

Next-generation sequencing and QC

Target enrichment was performed starting with 40 ng of input gDNA from PBMCs, 100 ng of DNA from peripheral blood ccfDNA from liquid biopsies; and 250 ng of DNA from FFPE samples using a customized-designed multi-gene capture panel (Qiagen, Hilden, Germany) that consists of 12 NET-related genes. All exons and exon-intron junctions of Succinate Dehydrogenase Complex Iron Sulfur Subunit B (SDHB), Succinate Dehydrogenase Complex Subunit C (SDHC), Cell Division Cycle 73 (CDC73), Calcium Sensing Receptor (CASR), Platelet Derived Growth Factor Receptor Alpha (PDGFRA), Succinate Dehydrogenase Complex Flavoprotein Subunit A (SDHA), Ret Proto-Oncogene (RET), Succinate Dehydrogenase Complex Assembly Factor 2 (SDHAF2), Menin 1 (MEN1), Succinate Dehydrogenase Complex Subunit D (SDHD), MYC Associated Factor X (MAX) and Protein Kinase CAMP-Dependent Type I Regulatory Subunit Alpha (PRKAR1A) genes were amplified and sequenced with a minimum of 300x coverage for germ-line studies and 1500x coverage for somatic studies.

Sequencing quality control assessments were carried out using the QCI-Analyze tool, and the QCI-Interpret interface was used for the bioinformatics analyses. Multiple quality control parameters including total yield, variant frequency, forward/reverse ratio, depth of coverage and quality score were assessed for each sample type. All of the detected genetic changes were evaluated independently of the patients’ diagnoses. Next generation sequencing steps and quality control assessments were performed with optimization based on previous studies [11, 12].

Data interpretation and bioinformatics analysis

Bioinformatics analyses were made for each variant detected using multiple databases including: HGMD (Human Gene Mutation Database), ClinVar, NCBI (National Center for Biotechnology Information), VarSome (The Human Genomic Variant Search Engine), ExAC (The Exome Aggregation Consortium), 1000 Genome Frequency, ESP (Exome Sequencing Project), Ancestry, Ingenuity Knowledge Base, COSMIC, GnomAD, OMIM (Online Mendelian Inheritance in Man) and Franklin to provide as much information as possible for making each clinical interpretation. All of the in-silico predictions were performed using a minimum of 10 different analysis tools including MutationTaster, SIFT and PolyPhen 2. The pathogenicity classification of each of the detected variants was determined in compliance with ACMG guidelines and standards.

In the secondary analysis, all of the variants which met the quality control criteria were investigated in accordance with the patients’ diagnoses and clinical findings. Hereditary predispositions were determined for germ-line studies, and actionability was assessed for the detected somatic variants.

Results

Total of thirty-five patients were recruited to the study which 25.7% (n = 9) of them are for somatic and 74.3% (n = 26) of them are for germline molecular testing. Six patients of 9 somatic studies were analysed for somatic variants in FFPE samples with their NET diagnoses. Actionable mutations with regard to therapy were detected in 3 (50%) of them. Patients had six actionable mutations in PDGFRA, RET and SDHA genes. Among the liquid biopsy studies, we detected four actionable mutations within the RET oncogene in two (75%) of the three liquid biopsy specimens. Mutations were observed in the same clone for one of these patients while the other patient had the mutation in different clones. The distribution of the detected somatic alterations is given in Table 2. The overall positivity rate was 55.6% among the somatic studies. Additionally, somatic status of PDGFR variant with 49% allel frequency was confirmed by the re-evaluation with germ-line study.

Table 2 Distribution of the actionable variants which all are in confidentiality rates according to the literature and our previous clinical validation studies [11,12,13]

Through the germ-line studies, we also observed a 50% positivity rate for disease predisposition with 16 variants identified among 13 patients. Germ-line mutations were seen in the RET, KIT, SDHB, SDHC, SDHD and PDGFRA genes. The variant distributions and their classification criteria are listed in Table 3 according to the ACMG Standards and Guidelines.

Table 3 Implemented variant classification criteria of detected germ-line mutations

Additionally, variation quantiles according to grade and site of origin were given in Fig. 1. The overall mutation distribution was not distinctive between neither the different material types nor the severity of diseases.

Fig. 1
figure 1

Variant distribution versus clinical characteristics. PB: Peripheral Blood, LB: Liquid Biopsy, FFPE: Formalin Fixed Paraffin Embedded Tissue, G: Grade, PLU: Primary Location Unknown, NET: Neuroendocrine Tumour, NEC: Neuroendocrine Carcinoma

This study evaluated both the risk factors and proposed potential risk factors together with germline and somatic genomic profiling. A multi-gene panel was used to investigate the genetics underlying both germline and somatic NETs and was demonstrated to provide actionable information in greater than 50% of the cases including those were evaluated using liquid biopsy specimens.

Discussion

Comprehensive molecular genetic profiling is a must for heterogeneous group of cancers such as NETs. As a result, we detected somatic variations in RET, PDGFRA and SDHA genes; while RET, KIT, SDHB, SDHC, SDHD and PDGFRA genes were mutated in germ-line studies. The distribution of the detected variants and genes are deviational because of the complex tissue origins of NETs and the individual differences. When the genetic changes classified by the ACMG criteria and assessed together with the both biological sample type and pathological findings, the frequency becomes similar between the groups. A comprehensive grade specific study with a broaden cohort is needed due to more precise variant interpretation and molecular grade classification of NETs.

We also emphasized that not only the somatic changes play a role in cancer progression and therapeutic approach, but a complement study of germ-line genetic profiling is beneficial for the patient. Cote et al. has shown that the liquid biopsy plays a prognostic role on medullary thyroid carcinoma due to a single-variant study of RET oncogene, and can be efficiently used in monitoring the disease [14]. Cavalcanti et al. recently reported that tracking PDGFRA expressions provide as a promising anti-angiogenic target in well-differentiated NETs [15]. Knösel et al. also showed that the over-expression of KIT and PDGFRA genes are related to short survival and a negative prognostic factor in advanced pancreatic neuroendocrine tumours [16]. Niemeijer et al. reported that SDH mutations also cause other types of cancers which have neuroendocrine origins other than paraganglionic tumours. Literature of NETs points that a broaden genetic profiling of both somatic and germline mutations should be the next step to improve NET diagnostics and therapeutic approaches. Emerging the accurate material type with the proper multi-gene panels that next generation sequencing technologies offer is the key.

Conclusions

Molecular genetic testing strategies can be tailored to fit an individual patient’s specific needs. The most efficient approach including the best specimen type and target gene panel will differ for each patient group, particularly those that are rare and/or heterogeneous cancers such as NETs. Genomic profiling medicine can act as a bridge between clinicians and patients for to provide precision for the development of therapeutic algorithms. Additionally, it is also crucial to practice effective patient follow-up to ensure proper patient management for both the patients and their families.

In summation, in this study we show the significance of selecting determinative biological samples in the decision making process to provide the best possible health care service.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author upon reasonable request.

Abbreviations

NET:

Neuroendocrine Tumours

NGS:

Next Generation Sequencing

GEP-NET:

Gastroenteropancreatic Neuroendocrine Tumours

DNA:

Deoxyribonucleic Acid

ccfDNA:

Circulating cell-free DNA

gDNA:

Genomic DNA

FFPE:

Formalin Fixed Paraffin Embedded

ACMG:

American College of Medical Genetics and Genomics

SDHB:

Succinate Dehydrogenase Complex Iron Sulfur Subunit B

SDHC:

Succinate Dehydrogenase Complex Subunit C

CDC73:

Cell Division Cycle 73

CASR:

Calcium Sensing Receptor

PDGFRA:

Platelet Derived Growth Factor Receptor Alpha

SDHA:

Succinate Dehydrogenase Complex Flavoprotein Subunit A

RET:

Ret Proto-Oncogene

SDHAF2:

Succinate Dehydrogenase Complex Assembly Factor 2

MEN1:

Menin 1

SDHD:

Succinate Dehydrogenase Complex Subunit D

MAX:

MYC Associated Factor X

PRKAR1A:

Protein Kinase CAMP-Dependent Type I Regulatory Subunit Alpha

HGMD:

Human Gene Mutation Database

NCBI:

National Center for Biotechnology Information)

ExAC:

The Exome Aggregation Consortium

ESP:

Exome Sequencing Project

COSMIC:

Catalogue of Somatic Mutations

OMIM:

Online Mendelian Inheritance in Man

ACMG:

American College of Medical Genetics

IARC:

International Agency for Research on Cancer

WHO:

World Health Organization

References

  1. Oronsky B, Ma PC, Morgensztern D, Carter CA. Nothing But NET: a review of neuroendocrine tumors and carcinomas. Neoplasia. 2017;19(12):991–1002.

    Article  CAS  Google Scholar 

  2. Shah MH, Goldner WS, Halfdanarson TR, Bergsland E, Berlin JD, Halperin D, et al. NCCN guidelines insights: neuroendocrine and adrenal tumors, version 2.2018. J Natl Compr Cancer Netw. 2018;16(6):693–702.

    Article  CAS  Google Scholar 

  3. Basu B, Sirohi B, Corrie P. Systemic therapy for neuroendocrine tumours of gastroenteropancreatic origin. Endocr Relat Cancer. 2010;17(1):R75–90.

    Article  CAS  Google Scholar 

  4. Rindi G, Klimstra DS, Abedi-Ardekani B, Asa SL, Bosman FT, Brambilla E, et al. A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod Pathol. 2018;31(12):1770–86.

    Article  Google Scholar 

  5. Mafficini A, Scarpa A. Genomic landscape of pancreatic neuroendocrine tumours: the international Cancer genome consortium. J Endocrinol. 2018;236(3):R161–r7.

    Article  CAS  Google Scholar 

  6. Tsoli M, Chatzellis E, Koumarianou A, Kolomodi D, Kaltsas G. Current best practice in the management of neuroendocrine tumors. Ther Adv Endocrinol Metab. 2019;10:2042018818804698.

    Article  Google Scholar 

  7. Vijayvergia N, Boland PM, Handorf E, Gustafson KS, Gong Y, Cooper HS, et al. Molecular profiling of neuroendocrine malignancies to identify prognostic and therapeutic markers: a fox Chase Cancer Center pilot study. Br J Cancer. 2016;115(5):564–70.

    Article  CAS  Google Scholar 

  8. Mafficini A, Scarpa A. Genetics and epigenetics of Gastroenteropancreatic neuroendocrine neoplasms. Endocr Rev. 2019;40(2):506–36.

    Article  Google Scholar 

  9. Banck MS, Kanwar R, Kulkarni AA, Boora GK, Metge F, Kipp BR, et al. The genomic landscape of small intestine neuroendocrine tumors. J Clin Invest. 2013;123(6):2502–8.

    Article  CAS  Google Scholar 

  10. Astsaturov IA, Cohen SJ, Engstrom PF, Millis SZ. Profiling of 1,350 neuroendocrine tumors for identification of multiple potential drug targets. J Clin Oncol. 2014;32(15_suppl):4113.

    Article  Google Scholar 

  11. Boga I, Sonmezler O, Bisgin A. Clinical Validation of a Novel GeneReader Next Generation Sequencing System for Tumor Specific Mutations and Bioinformatics Variant Analysis. Clin Lab. 2020;66(11):2213-8.

  12. Sonmezler O, Boga I, Bisgin A. Integration of Liquid Biopsies into Clinical Laboratory Applications via NGS in Cancer Diagnostics. Clin Lab. 2020;66(5):763-9.

  13. Tang Y, Liu X, Ou ZA, He Z, Zhu Q, Wang Y, et al. Maximum allele frequency observed in plasma: a potential indicator of liquid biopsy sensitivity. Oncol Lett. 2019;18(2):2118–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Cote GJ, Evers C, Hu MI, Grubbs EG, Williams MD, Hai T, et al. Prognostic significance of circulating RET M918T mutated tumor DNA in patients with advanced medullary thyroid carcinoma. J Clin Endocrinol Metab. 2017;102(9):3591–9.

    Article  Google Scholar 

  15. Cavalcanti E, Ignazzi A, De Michele F, Caruso ML. PDGFRα expression as a novel therapeutic marker in well-differentiated neuroendocrine tumors. Cancer Biol Ther. 2019;20(4):423–30.

    Article  CAS  Google Scholar 

  16. Knösel T, Chen Y, Altendorf-Hofmann A, Danielczok C, Freesmeyer M, Settmacher U, et al. High KIT and PDGFRA are associated with shorter patients survival in gastroenteropancreatic neuroendocrine tumors, but mutations are a rare event. J Cancer Res Clin Oncol. 2012;138(3):397–403.

    Article  Google Scholar 

Download references

Acknowledgements

We would like to thank the editorial board of “BMC Cancer” for their review. We also thank to enrolled patients and their families for participation.

Funding

This study was partially supported for the establishment of next-generation-sequencing custom panel designs and pipelines for the liquid biopsy samples by grants from Cukurova University Scientific Research Projects (Project No: TAY-2017-8767). The funding source had no role in publication.

Author information

Authors and Affiliations

Authors

Contributions

All authors have read and approved the manuscript. Author contributions were as follows; conceptualization, investigation and writing-original draft by IBG; investigation, methodology, validation, visualization and writing-original draft by OS; investigation, methodology, visualization and data curation by CM; patient recruitment and clinical interpretation by GB and ZYD; conceptualization, project administration, supervision and writing – review & editing by AB.

Corresponding author

Correspondence to Bisgin Atil.

Ethics declarations

Ethics approval and consent to participate

All participants were informed, and signed consent/permissions for this research in accordance with Helsinki declaration. The parental consent form was obtained from the children included in this study. Ethics approval was received from Cukurova University Ethical Committee.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burak, G.I., Ozge, S., Cem, M. et al. The emerging clinical relevance of genomic profiling in neuroendocrine tumours. BMC Cancer 21, 234 (2021). https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-021-07961-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-021-07961-y

Keywords