Elsevier

Leukemia Research

Volume 34, Issue 8, August 2010, Pages 1091-1093
Leukemia Research

Brief communication
Detection of isocitrate dehydrogenase 1 mutation R132H in myelodysplastic syndrome by mutation-specific antibody and direct sequencing

https://doi.org/10.1016/j.leukres.2010.02.014Get rights and content

Abstract

Sequencing of the acute myeloid leukemia genome revealed somatic mutations in isocitrate dehydrogenase-1. Acute myeloid leukemia frequently develops from myelodysplastic syndrome. In order to test whether myelodysplastic syndrome also carries isocitrate dehydrogenase-1 mutations, we stained a series of bone marrow samples from patients with myelodysplastic syndrome using an antibody specific for the R132H mutation. Three out of 71 patients exhibited antibody binding to myeloid precursor cells. The presence of the R132H mutation was confirmed by DNA sequencing. We demonstrated that isocitrate dehydrogenase-1 mutations occur in myelodysplasia preceding acute myeloid leukemia and that the R132H alteration can be detected by immunohistochemistry.

Introduction

Somatic mutations in the gene encoding cytosolic NADP+-dependent isocitrate dehydrogenase 1 (IDH1) are very frequent in human glioma, but extremely rare in other solid tumors [1], [2], [3]. R132H mutation dominantly inhibits IDH1 activity and induces hypoxia-inducible factor 1 (HIF1) [4]. Thus IDH1 may function as a tumor suppressor that might contribute to tumorigenesis in part through induction of the HIF1 pathway [4].

Acute myeloid leukemia (AML) is a malignant hematopoietic neoplasia characterized by heterogeneous genomic aberrations of myeloid precursor cells resulting in maturation arrest and bone marrow (BM) infiltration by blasts. In about 45–50% of AML no chromosomal abnormalities but hidden genomic alterations and somatic mutations can be detected. Recently IDH1 R132 mutations had been found in 10% of de novo AML [5] and mutated status was significantly associated with normal karyotype suggesting that these mutations are not random and are probably important for the pathogenesis of AML.

Myelodysplastic syndrome (MDS) is another clonal bone marrow disorder that shares with AML several pathogenetic mechanisms including common chromosomal aberrations and somatic mutations. Furthermore, MDS is characterized by presence of neoplastic sub-clones of myeloid precursor cells that accumulate genomic alterations and finally progress to AML with variable frequency. At the initial stages of MDS, the clonal stem cells usually represent a minority of BM cells. It is therefore very difficult to demonstrate the neoplastic nature of MDS, and the pathogenesis of MDS in general still remains obscure.

The aim of this study was to investigate a series of MDS cases for IDH1 R132H mutation using immunohistochemistry with a recently developed R132H mutation-specific monoclonal antibody [6].

Section snippets

Tissue specimens

BM-biopsies were randomly retrieved from the archives of the Pathology Department of Institute of Pathology Heidelberg (Heidelberg, Germany). The routine diagnosis was established according to World Health Organization (WHO) criteria on formalin-fixed, paraffin-embedded tissue specimens and peripheral blood/bone marrow smears. All diagnoses were reviewed and confirmed by two of the authors (MA and TL).

Immunohistochemistry

IDH1 R132H immunostaining was performed using a Ventana BenchMark XT immuno stainer (Ventana

Results and discussion

Immunohistochemical staining of 71 BM-biopsies of MDS patients using the IDH1 R132H mutation-specific antibody revealed three cases with positive hematopoietic cells. The mutated IDH1 was detectable in the cytoplasm in of granulocyte precursor cells and in few dysplastic megakaryocytes (Fig. 1A–C). This result suggests that mutant IDH1 occurs early in the course of disease and initially only affects few myeloid precursor cells that populate bone marrow together with the residual non-neoplastic

Conflict of interest statement

The authors report no potential conflicts of interest.

Acknowledgements

The authors would like to thank Charlotte Wahrendorf, Diana Jäger and Jochen Meyer for technical assistance. This work was supported by grants from the Bundesministerium fuer Bildung und Forschung (No. BMBF01ES0730 and No. BMBF01GS0883).

Contributions: AvD was the principal investigator and takes primary responsibility for the paper. HZ, DC and AvD developed the IDH antibody. TL and MA selected cases and analyzed data. DC, CH, AvD and MA evaluated the laboratory work. AvD and MA coordinated

References (8)

  • F.E. Bleeker et al.

    IDH1 mutations at residue p.R132 (IDH1(R132)) occur frequently in high-grade gliomas but not in other solid tumors

    Hum Mutat

    (2009)
  • C. Hartmann et al.

    Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas

    Acta Neuropathol

    (2009)
  • D.W. Parsons et al.

    An integrated genomic analysis of human glioblastoma multiforme

    Science

    (2008)
  • S. Zhao et al.

    Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha

    Science

    (2009)
There are more references available in the full text version of this article.

Cited by (16)

  • A gated material as immunosensor for in-tissue detection of IDH1-R132H mutation in gliomas

    2021, Sensors and Actuators, B: Chemical
    Citation Excerpt :

    The most habitual mutation detected was R132H (89.7 %), but other mutations have also been reported, for example R132C (2.8 %), R132 K (2.8 %), R132S (1.4 %) and R132 G (1.1 %) [9]. To date, this potential biomarker has been detected by routine clinical analysis such as direct sequencing [10,11], western-blot [12] and immunohistochemistry in tumour tissue using different anti-IDH1 (R132 H) antibodies [13–15]. Although some of them present high accuracy, these techniques are time consuming and require complex and expensive equipment [16].

  • Isocitrate dehydrogenase mutation hot spots in acute lymphoblastic leukemia and oral cancer

    2012, Kaohsiung Journal of Medical Sciences
    Citation Excerpt :

    Mutations of IDH impair the ability to catalyze the conversion of isocitrate to ɑ-ketoglutarate (ɑ-KG), which decreases conversion of ɑ-KG to the metabolite D2-hydroxyglutarate. Of the three IDH isoforms (IDH1, IDH2, and IDH3), two isoforms (IDH1 and IDH2) are associated with brain tumors [3–10], acute myeloid leukemia (AML) [11–15], myelodysplastic syndromes [16,17], prostate cancer [9], acute lymphoblastic leukemia (ALL) B [9], paraganglioma [18], thyroid cancer [19], and colorectal cancer [3,20]. Like tumor suppressors, heterozygous missense mutations of IDH1 residues act in a dominant negative fashion [18].

  • Rapid and reliable detection of IDH1 R132 mutations in acute myeloid leukemia using high-resolution melting curve analysis

    2011, Clinical Biochemistry
    Citation Excerpt :

    Identification of such mutation might be useful to provide a biomarker for further stratifying patients and monitoring minimal residual disease in cytogenetically normal AML. Several methods have been established to detect IDH1 mutation, including direct DNA sequencing, PCR followed by restriction fragment length polymorphism (RFLP) gel electrophoresis, PCR-single strain conformation polymorphism (SSCP) electrophoresis, and R132H antibody detection [2–17]. Each of these methods has its own merits and limitations, including sensitivity, ease of use, and low throughput.

  • IDH mutations in glioma and acute myeloid leukemia

    2010, Trends in Molecular Medicine
    Citation Excerpt :

    For one, the AML case shares important similarities to GBM in that IDH mutation already exists in several related precursor diseases often antecedent to the development of AML, parallel to the case of low-grade gliomas that progress to become secondary GBM maintaining the IDH mutation throughout disease progression. In addition to primary AML, recurrent IDH1 were found in 5–10% of sAML, MDS and MPN [29,38,57]. MDS and MPN are also clonal proliferative blood disorders affecting the bone marrow and share many of the common cytogenetic and somatic alterations found in primary AML.

  • Emerging Immunohistochemical Biomarkers for Myeloid Neoplasms

    2023, Archives of Pathology and Laboratory Medicine
View all citing articles on Scopus
View full text