Next Article in Journal / Special Issue
The Machinery of Exosomes: Biogenesis, Release, and Uptake
Previous Article in Journal
Toll-like Receptor 9 Induced Dendritic Cell Activation Promotes Anti-Myeloperoxidase Autoimmunity and Glomerulonephritis
Previous Article in Special Issue
The Pitfall of White Blood Cell Cystine Measurement to Diagnose Juvenile Cystinosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

FISH Diagnostic Assessment of MDM2 Amplification in Liposarcoma: Potential Pitfalls and Troubleshooting Recommendations

by
Alessandro Gambella
1,2,†,
Luca Bertero
2,†,
Milena Rondón-Lagos
3,
Ludovica Verdun Di Cantogno
4,
Nelson Rangel
5,
Chiara Pitino
2,
Alessia Andrea Ricci
2,
Luca Mangherini
2,
Isabella Castellano
2 and
Paola Cassoni
2,*
1
Division of Liver and Transplant Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
2
Department of Medical Sciences, University of Turin, 10124 Turin, Italy
3
School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
4
Department of Laboratory Medicine, Azienda Ospedaliera Città della Salute e della Scienza di Torino, 10126 Turin, Italy
5
Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(2), 1342; https://doi.org/10.3390/ijms24021342
Submission received: 9 November 2022 / Revised: 4 January 2023 / Accepted: 6 January 2023 / Published: 10 January 2023

Abstract

:
MDM2 amplification represents the leading oncogenic pathway and diagnostic hallmark of liposarcoma, whose assessment is based on Fluorescence In Situ Hybridization (FISH) analysis. Despite its diagnostic relevance, no univocal interpretation criteria regarding FISH assessments of MDM2 amplification have been established so far, leading to several different approaches and potential diagnostic misinterpretations. This study aims to address the most common issues and proposes troubleshooting guidelines for MDM2 amplification assessments by FISH. We retrospectively retrieved 51 liposarcomas, 25 Lipomas, 5 Spindle Cell Lipoma/Pleomorphic Lipomas, and 2 Atypical Spindle Cell Lipomatous Tumors and the corresponding MDM2 FISH analysis. We observed MDM2 amplification in liposarcomas cases only (43 out of 51 cases) and identified three MDM2-amplified patterns (scattered (50% of cases), clustered (14% of cases), and mixed (36% of cases)) and two nonamplified patterns (low number of signals (82% of cases) and polysomic (18% of cases)). Based on these data and published evidence in the literature, we propose a set of criteria to guide MDM2 amplification analysis in liposarcoma. Kindled by the compelling importance of MDM2 assessments to improve diagnostic and therapeutic liposarcoma management, these suggestions could represent the first step to develop a univocal interpretation model and consensus guidelines.

1. Introduction

Within the neoplastic soft tissue panorama, liposarcoma (LPS) represents the most common type of adult sarcoma, accounting for almost 20% of cases worldwide [1,2]. LPS is a clinical challenge, as it presents a high recurrence rate, unsatisfactory response to available treatments, and a challenging diagnostic workup, especially if based on morphology and immunohistochemistry alone [3].
The new WHO Classification of Soft Tissue Tumors edition identifies several specific subtypes of LPS, namely Atypical Lipomatous Tumors (ALT)/Well-Differentiated LPS (WDLPS), Myxoid LPS (MLPS), Dedifferentiated LPS (DDLPS), and Pleomorphic LPS (PLPS) [4,5,6]. The most common variants are WDLPS and MLPS, while DDLPS represents the aggressive evolution of WDLPS [7]. From a pathologist perspective, WDLPS and DDLPS represent the most challenging variants, as WDLPS can present overlapping features with benign tumors such as Lipomas, while DDLPS may show extensive dedifferentiated areas that are morphologically indistinguishable from other high-grade sarcomas [8,9,10]. Additionally, these neoplasms are usually initially approached with a needle biopsy, further complicating pathologist’s evaluation because of the limited sample available. Although immunohistochemistry (IHC) represents a valuable diagnostic tool for multiple sarcoma types, unfortunately, no immunophenotype specifically defines LPS, limiting its usefulness in this setting [8,11].
Molecular testing is a reliable and informative analysis to achieve an LPS diagnosis. The amplification of the Murine Double Minute-2 (MDM2) gene emerged as an essential molecular hallmark of LPS, particularly for ALT/WDLPS and DDLPS. MDM2 is an oncogene located in the long arm of chromosome 12 (12q15) (Figure 1) and its activity is strictly related to the expression of p53, with a negative feedback-type relationship.
MDM2 acts as an E3 ubiquitin-ligase, binding p53 and promoting its ubiquitylation and consequent proteasome-dependent degradation [12] (Figure 1). Low levels of p53 activity are unable to (I) regulate the cell cycle and (II) induce apoptosis in DNA-damaged cells, leading to uncontrolled proliferation. On the other hand, p53 regulates the expression of MDM2, balancing its activity and influence on the cell cycle and proliferation. MDM2 also regulates the Retinoblastoma protein (RB) [13]. In human tumor cell lines, MDM2 enhances RB degradation through a proteasome-dependent mechanism in a similar process as seen for p53 [12].
MDM2 amplification emerged as an oncogenic pathway in several malignances, but it is mostly represented in soft tissue sarcomas [14,15,16,17] where, differently from other tumors, MDM2 amplification and p53 mutation are mutually exclusive [18,19,20,21]. In soft tissue sarcomas, the MDM2 amplification occurs mainly through the so-called double minutes chromosomes (Dmins) mechanism [22]. Dmins are small, generally acentric, and autonomously replicating chromatin bodies that act as an amplification mechanism for several oncogenes, including MDM2 [23,24,25,26]. The sarcomas showing the highest percentages of MDM2 amplification are the low-grade/periosteal osteosarcoma, ALT/WDLPS, DDLPS, and intimal sarcoma [27]. Indeed, the evaluation of the MDM2 status is fundamental in the LPS diagnostics workup, as its amplification is present in 95% of WDLPS and DDLPS cases, while benign lipomatous lesions show no amplification at all [9,10,12]. IHC for MDM2 expression is available and frequently (>95% of cases) positive in WDLS. However, previous reports found an unsatisfactory correlation between IHC for the MDM2 protein and MDM2 gene amplification status, particularly in poorly differentiated cases or in cases with MDM2 overexpression not related to gene amplification [8,11,27,28,29]. Considering the clinical consequences of misdiagnosing these lesions, the molecular analysis of MDM2 is essential, and it is usually performed with Fluorescence In Situ Hybridization (FISH) [30]. Indeed, the MDM2 amplification evaluation by FISH is a crucial and well-established assay for liposarcoma diagnostic work-up and, nowadays, is considered as a diagnostic gold standard [31,32,33]. However, despite its diagnostic relevance, no definitive consensus has ever been defined for determining the MDM2 gene status. Considering the frequency of LPS in the adult population and the diagnostic and prognostic implications, the establishment of interpretation guidelines is an unmet need of crucial importance.
In this setting, our study aims to improve the diagnostic interpretation of the FISH assessment of MDM2 amplification in LPS by evaluating the potential drawbacks and pitfalls and suggesting a potential set of diagnostic criteria to achieve a standardized evaluation of this diagnostic test.

2. Results

2.1. MDM2 Amplification Accurately Stratify Our Series

We collected and analyzed 27 DDLPS, 19 WDLPS/ALT, 3 PLPS, 2 MLPS, 25 Lipomas, 5 Spindle Cell Lipoma/Pleomorphic Lipomas (SLC/PL), and 2 Atypical Spindle Cell Lipomatous Tumors (ASCLT). In our series, 43 cases (52%) presented MDM2 amplification including 25/27 DDLPS (93%) and 18/19 WDLPS/ALT (95%).
Amplified DDLPS cases presented a mean MDM2/CEP12 ratio of 10.1 (range 4.0–20.3) further detailed in a mean MDM2 copy number per cell of 24.1 (range 13.9–30.2) and a mean CEP12 copy number per cell of 2.5 (range 1.4–3.5).
WDLPS amplified cases presented a mean MDM2/CEP12 ratio of 8.2 (range 4.8–16.0). The mean MDM2 copy number per cell was 17.9 (range 11.4–28.4), and the mean CEP12 copy number per cell was 2.4 (range 1.8–3.7). The results are detailed in Table 1.

2.2. MDM2 Amplification Patterns

MDM2 amplified cases presented a high number of MDM2 copies (mean 21; range 11–30.2), a low number of CEP12 (mean 2.5; range 1.4–3.7), and, consequently, an MDM2/CEP12 ratio > 2. MDM2 positive cases presented three distinctive FISH amplification patterns: (i) the first (50% of cases) was characterized by several scattered signals distributed over the whole nucleus (Figure 2A); (ii) the second pattern (14% of cases) presented MDM2 signals clustered in specific areas of the nucleus (Figure 2B); (iii) the third presented overlapping features of the two previous patterns, as signals were contemporarily clustered and scattered (36% of cases). Regardless of these patterns, MDM2 amplified cases also presented extra chromosomal signals that were referred to as being Dmin. These signals were so small that they could be read by the Metafer 4 in 18 of the 43 amplified cases (42%) only. Of note, the Dmin amplification could present focal areas with overlapping signals resulting in an overall blurry appearance [34,35,36,37].
On the other hand, cases resulting negative for MDM2 amplification presented two different patterns: (i) The most common one (82% of cases) showed less than three MDM2 gene signals (MDM2 < 3; mean 2.2; range 1.7–2.9). These cases were considered negative regardless of the MDM2/CEP12 ratio (Figure 2C). (ii) The second pattern (18% of cases) showed instead three or more MDM2 gene signals (MDM2 ≥ 3; mean 3.7; range 3.1–4.3) together with a gain of the chromosome 12 centromere (mean 3.1; range 2.0–3.9) (Figure 2D).
We also identified two subgroups of cells with “giant nuclei” (i.e., at least two times larger than nearby nuclei) that presented challenging features. The first subgroup consisted of chromosome 12 polysomy (more than ten MDM2 and CEP12 signals) (Figure 2E—top inset) that was negative for MDM2 amplification by ratio. The evaluation of the EGFR/CEP7 dual probe expression in this subgroup revealed EGFR/CEP7 polysomy and confirmed the cells’ polyploidy (Figure 2F). The second subgroup is represented by amplified cells with “giant nuclei” harboring increased MDM2 gene signals and was positive by ratio (Figure 2E—bottom inset).

2.3. Literature Review and Analysis

We first identified 90 studies that initially satisfied the keywords research on main databases. After a careful analysis and screening, 36 studies were eventually selected (Table 2).
From these studies, a relevant heterogeneity in the MDM2 amplification assessment clearly emerged, as the median number of evaluated nuclei per analysis was 60 but with a wide range (20 to 200). Similarly, the definition of MDM2 amplification itself varied significantly, as some studies based their evaluation upon the absolute MDM2 count (i.e., three or more signals as sufficient for a diagnosis of amplification [51,63], while others set the limit to five signals) [30,52,54,57]. More commonly, the MDM2/CEP12 ratio was used, but again with variable cut-off thresholds including >2 [10,41,44,46,47,50,59], ≥2 [8,9,42,43,48,49,56,61], or ≥2.2 [58], probably borrowing this criteria from the previous ASCO/CAP interpretation guidelines for HER2 assessment [64]. Of note, even the type of FISH probes utilized greatly differed or was not exhaustively reported. FISH probes ranged from home made [8,9,10,48] to commercially available, the latter including dual probes (LSI MDM2/CEP12) [30,44,46,47,49,50,51,52,56,57,58,59,61] but also single locus probes (LSI MDM2) that do not allow chromosome 12 polysomy evaluation [1,54,56] (Figure 3).

3. Discussion

To date, soft tissue lesions are initially approached and diagnosed mainly through the accurate combination of clinical data and morphological features of biopsy samples. This practice, although useful and accurate in several settings, presents crucial drawbacks, especially when dealing with equivocal and misleading histopathological findings, as in the LPS scenario. Indeed, LPS and, particularly, the ALT/WDLPS, DDLPS, and UPS variants present relevant overlapping morphologic features with other types of malignant sarcomas or even with benign entities, such as Lipomas, thus leading to a challenging diagnostic assessment and a broad differential diagnosis process. In this context, the evaluation of MDM2 status is of crucial support, as it represents a diagnostic hallmark of LPS. Regardless of its diagnostic relevance, no guidelines or consensus criteria regarding MDM2 gene amplification interpretation have ever been proposed, whereas previous studies assessing this molecular hallmark adopted significant methodological differences, including a broad spectrum of criteria for probe counting and diagnostic cut-off values [65,66]. This methodological heterogeneity could lead to confounding definitions and hamper the diagnostic reliability/reproducibility, eventually misleading patient clinical management.
Based on published evidence and our data, we hereby provide a set of considerations and recommendations regarding FISH interpretation criteria for MDM2 amplification assessments that should be considered in the LPS diagnostic workup (Figure 4).
#1 The analyzed nuclei must be representative of the entire lesion. The FISH analysis is performed in a small, selected area, that, however, has to be representative of the whole lesion. We recommend that an expert pathologist in soft tissue tumors determines the area for FISH analysis, supplying marked hematoxylin–eosin (H&E)-stained slides. This criterion, common to every molecular analysis, should be strictly and routinely applied to avoid misleading results due to nonrepresentative sampling.
#2 The MDM2 amplification is secondary to a Dmin-based mechanism. Dmins are extra chromosomal elements that are considered cytogenetic hallmarks of high-gene amplification. Based on our data, Dmins were the main mechanism behind MDM2 amplification in all the cases evaluated.
#3 MDM2/CEP12 ratio is essential to define MDM2 status. We recommend determining the MDM2/CEP12 ratio and consider only cases with a ratio > 2 as amplified. Generally, MDM2 amplified cases present many MDM2 gene signals together with a mean of two CEP12 signals. In our experience, observing a >2 MDM2/CEP12 ratio is necessary to differentiate amplified from polysomic cases. We did not experience cases with a ratio = 2, but we consider that this equivocal pattern could be solved by extending the cells count. Furthermore, we observed no cases with MDM2 signals < 4 and a ratio > 2. This eventuality could happen due to the loss of the CEP12 (CEP12 < 1.8), but we would have considered these cases as not amplified without any further analysis. Similarly, cases with several MDM2 signals (e.g., >6) but a <2 ratio should be considered nonamplified as well. In this scenario, the use of other enumeration probes could prove helpful in confirming cells’ polysomic nature.
#4 Ambiguous cases require attention and critical review, especially if polysomy is suspected. In ambiguous cases and, in particular, if polysomy is suspected, we recommend analyzing at least 50 nuclei to avoid MDM2 status misinterpretation. In these cases, a careful assessment of the MDM2/CEP12 ratio is crucial to properly determine the amplification status.
Screening and reviewing the literature regarding MDM2 amplification, we noticed that several studies used assessment criteria similar to the ones reported in breast cancer HER2 evaluation guidelines. Overall, we discourage this practice, as these guidelines were tailored for a different setting (breast cancer) and a diverse gene with its peculiar amplification pattern. As we already discussed, MDM2 amplification is developed through the Dmin mechanism and is usually characterized by the presence of several gene signals (MDM2 > 10) clustered or scattered over the whole nucleus, with a low centromere number (Figure 5A). Differently, HER2 amplification mainly occurs with two distinct amplification mechanisms: 30% of HER2-positive breast tumors present a Dmin amplification mechanism with a pattern similar to the ones described for MDM2 (Figure 5A), but the majority of HER2-positive breast cases (~60%) involve intrachromosomal regions called homogeneously staining regions (HSR) (Figure 5B) [67,68]. HSR have also been identified in breast cancer cell lines, including BT474, SKBR3, and JIMT-1 [69]. HSR are unusual patterns associated with the gain (CEP > 2), loss (CEP < 2), or coamplification of the centromeric region [70]. The differences could depend on the HSR extension and the presence of other cytogenetic aberrations (translocations, inversions, and deletions) involving the gene loci and the CEP. Furthermore, breast cancer HER2 assessment guidelines are based on different aims which mainly include the prognostic and predictive assessment (the selection of eligible patients for Trastuzumab treatment). Differently, LPS MDM2 amplification harbors a diagnostic value only, to date. Based on this evidence and distinct characteristics, the application of HER2 criteria to define MDM2 status could increase, rather than reduce, inappropriate interpretations and diagnosis.
Our study presents some limitations, mainly represented by the monocentric collection and analysis of our series and the absence of external validation, which is required to independently confirm our diagnostic recommendations.
This effort is especially important if we consider the evaluation of MDM2 amplification by FISH as a cornerstone of the LPS diagnostic workup, but it is still affected by the lack of consensus guidelines, potentially resulting in different and even misleading approaches. Our study represents the first attempt to solve this controversial scenario and develop formal guidance for the interpretation of this assay to optimize the diagnostic and clinical management of patients with suspected LPS.

4. Materials and Methods

This study focuses on the FISH assessment of MDM2 status in LPS by combining data acquired at our Institution and the evidence published in the literature so far.

4.1. Case Series Construction and FISH Analysis

Formalin-fixed paraffin-embedded (FFPE) tissue blocks of 83 adipocytic tumors (diagnosed from 2014 to 2019) were retrieved from the archive of the Pathology Unit of the Città della Salute e della Scienza Hospital (Turin, Italy). From each FFPE block, two 4 μm thick tissue serial sections were cut for a tissue adequacy evaluation and FISH analysis. The slides for the tissue adequacy assessment were stained with hematoxylin and eosin and reviewed by two pathologists with soft-tissue expertise (Figure 6).
The slides for FISH analysis were baked overnight at 58 °C and then deparaffinized. Later, samples were treated with the Invitrogen Spot-light tissue pretreatment kit (Invitrogen Corporation, Camarillo, CA, USA) at 98 °C for 15 min, and enzymatic digestion with a protease solution (pepsin) at 37 °C for 45 to 60 min was then performed. Finally, the sections were dehydrated in ethanol of different concentrations for the subsequent hybridization. The hybridization was performed indifferently using two commercially available dual-color probes: MDM2 (green spectrum)/CEP12 (orange spectrum) (Abbott Molecular, Chicago, IL, USA) and ZytoLight SPEC MDM2 (green spectrum)/CEN 12 (orange spectrum) (Zytovision. GmbH, Bremerhaven, Germany) (for consistency, the control probe has always been identified as CEP12). The slides were codenatured in an HYBrite System at 72 °C for 5 min (Abbott) or 75 °C for 10 min (Zytovision) and hybridized overnight at 37 °C. The slides were then washed in a 0.7xSSC/0.3% NP-40 solution at 73.5 °C for 3 min (Abbott) or in a 2xSSC/0.3% NP-40 solution at 73.5 °C for 3 min (Zytovision); then, they were dehydrated in ethanol of different concentrations, air-dried, and counterstained with 6-diamidino-2-phenylindole (DAPI). The presence of polyploidy was identified using a dual-color probe for EGFR (7p11) (orange spectrum; Abbott Molecular) and the centromere of chromosome 7 (D7Z1) (green spectrum; Abbott Molecular).
On each slide, five to ten tumor areas of interest were identified, selected, and automatically acquired with the motorized Metafer 4 Scanning System (Carl Zeiss MetaSystems GmbH. Jena, Germany) equipped with AxioImager epifluorescence microscope (one focus plane for DAPI and nine focus planes for green and red spots). An analysis of the MDM2/CEP12 and EGFR/CEP7 probe patterns was performed both with the Metafer 4 software and by counting the MDM2 and CEP12 spots on images taken through Metafer 4 and transferred into the Integrated Set of Information Systems (ISIS) software.
Successively, the MDM2 gene was evaluated on 20 to 200 nuclei in the selected representative areas. Only nuclei with both the MDM2 and CEP12 signals were assessed.
An MDM2/CEP12 ratio higher than two (MDM2/CEP12 > 2) was considered positive for MDM2 amplification, as assumed from the available literature [9,18,19,20,21,22,23,24]. Cases with a ratio equal or smaller than two were considered not MDM2 amplified regardless of the absolute number of copies (e.g., polysomic cases with a relatively high number of MDM2 copies, but with a high number of CEP12 copies too).

4.2. Literature Analysis

An extensive literature review of the published evidence regarding MDM2 status in LPS was performed by querying PubMed, Scopus, Embase, and Web of Science databases. Up to 90 papers were identified using the following keywords: (“MDM2”) AND (“amplification”) AND (“FISH”) AND (“LPS” OR “Liposarcoma”). Abstracts of conference presentations, case reports, and non-English written papers were excluded. The title and abstract of the selected papers were then screened and assessed for appropriateness, whereas references were double checked to identify potentially neglected relevant articles and ensure literature research adequacy. From each study, details about study design, material and methods, and FISH analysis outcomes were then evaluated and recorded.

5. Conclusions

In conclusion, clinical data and morphologic features are of crucial relevance to approach LPS diagnosis, but molecular techniques, such as FISH cytogenetic analysis, are increasingly required to achieve a conclusive diagnosis, particularly for specific variants such as ALT/WDLPS and DDPLS. Considering its crucial diagnostic role in the LPS diagnostic workup, MDM2 amplification assessment requires a clearly defined workflow and interpretation criteria.
Based on our experience as a tertiary referral center for LPS diagnostic assessment and considering the current literature evidence, we here proposed a set of criteria for MDM2 FISH assessment as a step towards the development of consensus-based formal guidelines.

Author Contributions

Conceptualization, A.G., L.B., I.C. and P.C.; data curation, M.R.-L., L.V.D.C., N.R., C.P., A.A.R. and L.M.; formal analysis, M.R.-L., L.V.D.C., N.R., C.P., A.A.R. and L.M.; methodology, L.V.D.C.; supervision, I.C. and P.C.; writing—original draft, A.G.; writing—review and editing, A.G., L.B., M.R.-L., L.V.D.C., N.R., C.P., A.A.R., L.M., I.C. and P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki) for experiments involving humans and within the guidelines and regulations defined by the Research Ethics Committee of the University of Turin. This study was approved by the Research Ethics Committee of the University of Turin.

Informed Consent Statement

Patient consent was waived due to the retrospective nature of the research protocol and the fact that this study had no impact at all on the patients’ care.

Data Availability Statement

The data supporting the findings of this study are not publicly available due to privacy or ethical restrictions but can be obtained upon reasonable request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marino-Enriquez, A.; Hornick, J.L.; Dal Cin, P.; Cibas, E.S.; Qian, X. Dedifferentiated liposarcoma and pleomorphic liposarcoma: A comparative study of cytomorphology and MDM2/CDK4 expression on fine-needle aspiration. Cancer Cytopathol. 2014, 122, 128–137. [Google Scholar] [CrossRef]
  2. De Vita, A.; Mercatali, L.; Recine, F.; Pieri, F.; Riva, N.; Bongiovanni, A.; Liverani, C.; Spadazzi, C.; Miserocchi, G.; Amadori, D.; et al. Current classification, treatment options, and new perspectives in the management of adipocytic sarcomas. OncoTargets Ther. 2016, 9, 6233–6246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mack, T.M. Sarcomas and other malignancies of soft tissue, retroperitoneum, peritoneum, pleura, heart, mediastinum, and spleen. Cancer 1995, 75 (Suppl. 1), 211–244. [Google Scholar] [CrossRef]
  4. Sbaraglia, M.; Bellan, E.; Tos, A.P.D. The 2020 WHO Classification of Soft Tissue Tumours: News and perspectives. Pathologica 2020, 113, 70–84. [Google Scholar] [CrossRef] [PubMed]
  5. Choi, J.H.; Ro, J.Y. The 2020 WHO Classification of Tumors of Soft Tissue: Selected Changes and New Entities. Adv. Anat. Pathol. 2021, 28, 44–58. [Google Scholar] [CrossRef]
  6. Kallen, M.E.; Hornick, J.L. The 2020 WHO Classification: What’s New in Soft Tissue Tumor Pathology? Am. J. Surg. Pathol. 2021, 45, e1–e23. [Google Scholar] [CrossRef] [PubMed]
  7. Nascimento, A.G. Dedifferentiated liposarcoma. Semin. Diagn. Pathol. 2001, 18, 263–266. [Google Scholar]
  8. Weaver, J.; Downs-Kelly, E.; Goldblum, J.R.; Turner, S.; Kulkarni, S.; Tubbs, R.R.; Rubin, B.P.; Skacel, M. Fluorescence in situ hybridization for MDM2 gene amplification as a diagnostic tool in lipomatous neoplasms. Mod. Pathol. 2008, 21, 943–949. [Google Scholar] [CrossRef] [Green Version]
  9. Weaver, J.; Goldblum, J.R.; Turner, S.; Tubbs, R.R.; Wang, W.L.; Lazar, A.J.; Rubin, B.P. Detection of MDM2 gene amplification or protein expression distinguishes sclerosing mesenteritis and retroperitoneal fibrosis from inflammatory well-differentiated liposarcoma. Mod. Pathol. 2009, 22, 66–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kimura, H.; Dobashi, Y.; Nojima, T.; Nakamura, H.; Yamamoto, N.; Tsuchiya, H.; Ikeda, H.; Sawada-Kitamura, S.; Oyama, T.; Ooi, A. Utility of fluorescence in situ hybridization to detect MDM2 amplification in liposarcomas and their morphological mimics. Int. J. Clin. Exp. Pathol. 2013, 6, 1306–1316. [Google Scholar] [PubMed]
  11. Sirvent, N.; Coindre, J.-M.; Maire, G.; Hostein, I.; Keslair, F.; Guillou, L.; Ranchere-Vince, D.; Terrier, P.; Pedeutour, F. Detection of MDM2-CDK4 Amplification by Fluorescence In Situ Hybridization in 200 Paraffin-embedded Tumor Samples: Utility in Diagnosing Adipocytic Lesions and Comparison With Immunohistochemistry and Real-time PCR. Am. J. Surg. Pathol. 2007, 31, 1476–1489. [Google Scholar] [CrossRef] [PubMed]
  12. Shi, D.; Gu, W. Dual Roles of MDM2 in the Regulation of p53: Ubiquitination Dependent and Ubiquitination Independent Mechanisms of MDM2 Repression of p53 Activity. Genes Cancer 2012, 3, 240–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jarboe, E.A.; Layfield, L.J. Cytologic features of pancreatic intraepithelial neoplasia and pancreatitis: Potential pitfalls in the diagnosis of pancreatic ductal carcinoma. Diagn. Cytopathol. 2011, 39, 575–581. [Google Scholar] [CrossRef]
  14. Kato, S.; Ross, J.S.; Gay, L.; Dayyani, F.; Roszik, J.; Subbiah, V.; Kurzrock, R. Analysis of MDM2 Amplification: Next-Generation Sequencing of Patients with Diverse Malignancies. JCO Precis. Oncol. 2018, 2, 1–14. [Google Scholar] [CrossRef]
  15. Dembla, V.; Somaiah, N.; Barata, P.; Hess, K.; Fu, S.; Janku, F.; Karp, D.D.; Naing, A.; Piha-Paul, S.A.; Subbiah, V.; et al. Prevalence of MDM2 amplification and coalterations in 523 advanced cancer patients in the MD Anderson phase 1 clinic. Oncotarget 2018, 9, 33232–33243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sciot, R. MDM2 Amplified Sarcomas: A Literature Review. Diagnostics 2021, 11, 496. [Google Scholar] [CrossRef]
  17. Urso, L.; Calabrese, F.; Favaretto, A.; Conte, P.; Pasello, G. Critical review about MDM2 in cancer: Possible role in malignant mesothelioma and implications for treatment. Crit. Rev. Oncol. 2015, 97, 220–230. [Google Scholar] [CrossRef]
  18. Ito, M.; Barys, L.; O’Reilly, T.; Young, S.; Gorbatcheva, B.; Monahan, J.; Zumstein-Mecker, S.; Choong, P.F.; Dickinson, I.; Crowe, P.; et al. Comprehensive Mapping of p53 Pathway Alterations Reveals an Apparent Role for Both SNP309 and MDM2 Amplification in Sarcomagenesis. Clin. Cancer Res. 2011, 17, 416–426. [Google Scholar] [CrossRef] [Green Version]
  19. Momand, J.; Jung, D.; Wilczynski, S.; Niland, J. The MDM2 gene amplification database. Nucleic Acids Res. 1998, 26, 3453–3459. [Google Scholar] [CrossRef] [Green Version]
  20. Traweek, R.S.; Cope, B.M.; Roland, C.L.; Keung, E.Z.; Nassif, E.F.; Erstad, D.J. Targeting the MDM2-p53 pathway in dedifferentiated liposarcoma. Front. Oncol. 2022, 12, 1006959. [Google Scholar] [CrossRef]
  21. Wade, M.; Li, Y.-C.; Wahl, G.M. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nat. Rev. Cancer 2013, 13, 83–96. [Google Scholar] [CrossRef]
  22. Fakharzadeh, S.S.; Rosenblum-Vos, L.; Murphy, M.; Hoffman, E.K.; George, D.L. Structure and Organization of Amplified DNA on Double Minutes Containing the mdm2 Oncogene. Genomics 1993, 15, 283–290. [Google Scholar] [CrossRef]
  23. Hahn, P.J. Molecular biology of double-minute chromosomes. Bioessays 1993, 15, 477–484. [Google Scholar] [CrossRef]
  24. Kuttler, F.; Mai, S. Formation of non-random extrachromosomal elements during development, differentiation and oncogenesis. Semin. Cancer Biol. 2007, 17, 56–64. [Google Scholar] [CrossRef]
  25. Gebhart, E. Double minutes, cytogenetic equivalents of gene amplification, in human neoplasia—A review. Clin. Transl. Oncol. 2005, 7, 477–485. [Google Scholar] [CrossRef]
  26. Wahl, G.M. The importance of circular DNA in mammalian gene amplification. Cancer Res. 1989, 49, 1333–1340. [Google Scholar] [PubMed]
  27. Anderson, W.J.; Hornick, J.L. Immunohistochemical correlates of recurrent genetic alterations in sarcomas. Genes Chromosomes Cancer 2019, 58, 111–123. [Google Scholar] [CrossRef]
  28. Hornick, J.L. Limited biopsies of soft tissue tumors: The contemporary role of immunohistochemistry and molecular diagnostics. Mod. Pathol. 2019, 32, 27–37. [Google Scholar] [CrossRef] [PubMed]
  29. Machado, I.; Vargas, A.C.; Maclean, F.; Llombart-Bosch, A. Negative MDM2/CDK4 immunoreactivity does not fully exclude MDM2/CDK4 amplification in a subset of atypical lipomatous tumor/ well differentiated liposarcoma. Pathol. Res. Pract. 2022, 232, 153839. [Google Scholar] [CrossRef] [PubMed]
  30. Thway, K.; Wang, J.; Swansbury, J.; Min, T.; Fisher, C. Fluorescence In Situ Hybridization for MDM2 Amplification as a Routine Ancillary Diagnostic Tool for Suspected Well-Differentiated and Dedifferentiated Liposarcomas: Experience at a Tertiary Center. Sarcoma 2015, 2015, 812089. [Google Scholar] [CrossRef] [Green Version]
  31. Vargas, A.C.; Joy, C.; Cheah, A.L.; Jones, M.; Bonar, F.; Brookwell, R.; Garrone, B.; Talbot, J.; Harraway, J.; Gill, A.J.; et al. Lessons learnt from MDM2 fluorescence in-situ hybridisation analysis of 439 mature lipomatous lesions with an emphasis on atypical lipomatous tumour/well-differentiated liposarcoma lacking cytological atypia. Histopathology 2021, 80, 369–380. [Google Scholar] [CrossRef] [PubMed]
  32. Sugiyama, K.; Washimi, K.; Sato, S.; Hiruma, T.; Sakai, M.; Okubo, Y.; Miyagi, Y.; Yokose, T. Differential diagnosis of lipoma and atypical lipomatous tumor/well-differentiated liposarcoma by cytological analysis. Diagn. Cytopathol. 2022, 50, 112–122. [Google Scholar] [CrossRef] [PubMed]
  33. Pei, J.; Flieder, D.B.; Talarchek, J.N.; Cooper, H.S.; Patchefsky, A.S.; Wei, S. Clinical Application of Chromosome Microarray Analysis in the Diagnosis of Lipomatous Tumors. Appl. Immunohistochem. Mol. Morphol. 2021, 29, 592–598. [Google Scholar] [CrossRef]
  34. Lopez-Gines, C.; Gil-Benso, R.; Ferrer-Luna, R.; Benito, R.; Serna, E.; Gonzalez-Darder, J.; Quilis, V.; Monleon, D.; Celda, B.; Cerdá-Nicolas, M. New pattern of EGFR amplification in glioblastoma and the relationship of gene copy number with gene expression profile. Mod. Pathol. 2010, 23, 856–865. [Google Scholar] [CrossRef] [Green Version]
  35. Aygun, N.; Altungoz, O. MYCN is amplified during S phase, and c-myb is involved in controlling MYCN expression and amplification in MYCN-amplified neuroblastoma cell lines. Mol. Med. Rep. 2019, 19, 345–361. [Google Scholar] [CrossRef] [PubMed]
  36. Paulsson, K.; Lassen, C.; Kuric, N.; Billström, R.; Fioretos, T.; Tanke, H.J.; Johansson, B. MYC is not overexpressed in a case of chronic myelomonocytic leukemia with MYC-containing double minutes. Leukemia 2003, 17, 813–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Cuthbert, G.; Thompson, K.; McCullough, S.; Watmore, A.; Dickinson, H.; Telford, N.; Mugneret, F.; Harrison, C.; Griffiths, M.; Bown, N. MLL amplification in acute leukaemia: A United Kingdom Cancer Cytogenetics Group (UKCCG) study. Leukemia 2000, 14, 1885–1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Rekhi, B.; Karnik, N.; Agrawal, R.; Shetty, O.; Patkar, S. Detection of MDM2 gene amplification on tissue microarray-based Fluorescence In-Situ Hybridization (FISH) in well-differentiated and dedifferentiated liposarcomas, displaying a wide morphological spectrum: A validation study at a tertiary cancer referral centre. Indian J. Pathol. Microbiol. 2022, 65, 65–75. [Google Scholar]
  39. Sarwar, S.; Mushtaq, S.; Hassan, U.; Maqbool, H.; Qazi, R. Diagnostic Utility of Fish for MDM2 in Adipocytic Neoplasms. J. Ayub. Med. Coll. Abbottabad. 2021, 33, 563–567. [Google Scholar]
  40. Knebel, C.; Neumann, J.; Schwaiger, B.J.; Karampinos, D.C.; Pfeiffer, D.; Specht, K.; Lenze, U.; Von Eisenhart-Rothe, R.; Rummeny, E.J.; Woertler, K.; et al. Differentiating atypical lipomatous tumors from lipomas with magnetic resonance imaging: A comparison with MDM2 gene amplification status. BMC Cancer 2019, 19, 309. [Google Scholar] [CrossRef] [Green Version]
  41. Stojanov, I.J.; Mariño-Enriquez, A.; Bahri, N.; Jo, V.Y.; Woo, S.-B. Lipomas of the Oral Cavity: Utility of MDM2 and CDK4 in Avoiding Overdiagnosis as Atypical Lipomatous Tumor. Head Neck Pathol. 2018, 13, 169–176. [Google Scholar] [CrossRef]
  42. Vargas, A.C.; Selinger, C.; Satgunaseelan, L.; Cooper, W.A.; Gupta, R.; Stalley, P.; Brown, W.; Soper, J.; Schatz, J.; Boyle, R.; et al. FISH analysis of selected soft tissue tumors: Diagnostic experience in a tertiary center. Asia-Pac. J. Clin. Oncol. 2018, 15, 38–47. [Google Scholar] [CrossRef] [Green Version]
  43. Michal, M.; Agaimy, A.; Contreras, A.L.; Svajdler, M.; Kazakov, D.V.; Steiner, P.; Grossmann, P.; Martinek, P.; Hadravsky, L.; Michalova, K.; et al. Dysplastic Lipoma: A Distinctive Atypical Lipomatous Neoplasm with Anisocytosis, Focal Nuclear Atypia, p53 Overexpression, and a Lack of MDM2 Gene Amplification by FISH.; A Report of 66 Cases Demonstrating Occasional Multifocality and a Rare Association with Retinoblastoma. Am. J. Surg. Pathol. 2018, 42, 1530–1540. [Google Scholar]
  44. Asif, A.; Mushtaq, S.; Hassan, U.; Akhter, N.; Hussain, M.; Azam, M.; Qazi, R. Fluorescence in Situ Hybridization (FISH) for Differential Diagnosis of Soft Tissue Sarcomas. Asian Pac. J. Cancer Prev. 2018, 19, 655–660. [Google Scholar] [CrossRef] [PubMed]
  45. Marino-Enriquez, A.; Nascimento, A.F.; Ligon, A.H.; Liang, C.; Fletcher, C.D. Atypical Spindle Cell Lipomatous Tumor: Clinicopathologic Characterization of 232 Cases Demonstrating a Morphologic Spectrum. Am. J. Surg. Pathol. 2017, 41, 234–244. [Google Scholar] [CrossRef] [PubMed]
  46. Kammerer-Jacquet, S.-F.; Thierry, S.; Cabillic, F.; Lannes, M.; Burtin, F.; Henno, S.; Dugay, F.; Bouzillé, G.; Rioux-Leclercq, N.; Belaud-Rotureau, M.-A.; et al. Differential diagnosis of atypical lipomatous tumor/well-differentiated liposarcoma and dedifferentiated liposarcoma: Utility of p16 in combination with MDM2 and CDK4 immunohistochemistry. Hum. Pathol. 2017, 59, 34–40. [Google Scholar] [CrossRef] [PubMed]
  47. Yamashita, K.; Kohashi, K.; Yamada, Y.; Nishida, Y.; Urakawa, H.; Oda, Y.; Toyokuni, S. Primary extraskeletal osteosarcoma: A clinicopathological study of 18 cases focusing on MDM2 amplification status. Hum. Pathol. 2017, 63, 63–69. [Google Scholar] [CrossRef] [PubMed]
  48. Wong, D.D.; Low, I.C.; Peverall, J.; Robbins, P.D.; Spagnolo, D.V.; Nairn, R.; Wood, D. MDM2/CDK4 gene amplification in large/deep-seated ‘lipomas’: Incidence, predictors and clinical significance. Pathology 2016, 48, 203–209. [Google Scholar] [CrossRef]
  49. Zhang, G.; Lanigan, C.P.; Goldblum, J.R.; Tubbs, R.R.; Downs-Kelly, E. Automated Bright-Field Dual-Color In Situ Hybridization for MDM2: Interobserver Reproducibility and Correlation With Fluorescence In Situ Hybridization in a Series of Soft Tissue Consults. Arch. Pathol. Lab. Med. 2016, 140, 1111–1115. [Google Scholar] [CrossRef] [Green Version]
  50. Setsu, N.; Miyake, M.; Wakai, S.; Nakatani, F.; Kobayashi, E.; Chuman, H.; Hiraoka, N.; Kawai, A.; Yoshida, A. Primary Retroperitoneal Myxoid Liposarcomas. Am. J. Surg. Pathol. 2016, 40, 1286–1290. [Google Scholar] [CrossRef] [Green Version]
  51. Inyang, A.; Thomas, D.G.; Jorns, J. Heterologous Liposarcomatous Differentiation in Malignant Phyllodes Tumor is Histologically Similar but Immunohistochemically and Molecularly Distinct from Well-differentiated Liposarcoma of Soft Tissue. Breast J. 2016, 22, 282–286. [Google Scholar] [CrossRef] [PubMed]
  52. Lyle, P.L.; Bridge, J.A.; Simpson, J.F.; Cates, J.M.; Sanders, M.E. Liposarcomatous differentiation in malignant phyllodes tumors is unassociated with MDM2 or CDK4 amplification. Histopathology 2015, 68, 1040–1045. [Google Scholar] [CrossRef] [PubMed]
  53. Creytens, D.; van Gorp, J.; Ferdinande, L.; Speel, E.J.; Libbrecht, L. Detection of MDM2/CDK4 amplification in lipomatous soft tissue tumors from formalin-fixed, paraffin-embedded tissue: Comparison of multiplex ligation-dependent probe amplification (MLPA) and fluorescence in situ hybridization (FISH). Appl. Immunohistochem. Mol. Morphol. 2015, 23, 126–133. [Google Scholar] [CrossRef] [PubMed]
  54. Jour, G.; Gullet, A.; Liu, M.; Hoch, B.L. Prognostic relevance of Fédération Nationale des Centres de Lutte Contre le Cancer grade and MDM2 amplification levels in dedifferentiated liposarcoma: A study of 50 cases. Mod. Pathol. 2014, 28, 37–47. [Google Scholar] [CrossRef] [PubMed]
  55. Clay, M.R.; Martinez, A.P.; Weiss, S.W.; Edgar, M.A. MDM2 Amplification in Problematic Lipomatous Tumors: Analysis of FISH Testing Criteria. Am. J. Surg. Pathol. 2015, 39, 1433–1439. [Google Scholar] [CrossRef] [PubMed]
  56. Ware, P.L.; Snow, A.N.; Gvalani, M.; Pettenati, M.J.; Qasem, S.A. MDM2 copy numbers in well-differentiated and dedifferentiated liposarcoma: Characterizing progression to high-grade tumors. Am. J. Clin. Pathol. 2014, 141, 334–341. [Google Scholar] [CrossRef] [Green Version]
  57. Horn, H.; Allmanritter, J.; Doglioni, C.; Marx, A.; Müller, J.; Gattenlöhner, S.; Staiger, A.M.; Rosenwald, A.; Ott, G.; Ott, M.M. Fluorescence in situ analysis of soft tissue tumor associated genetic alterations in formalin-fixed paraffin-embedded tissue. Pathol. Res. Pract. 2014, 210, 804–811. [Google Scholar] [CrossRef]
  58. Cho, J.; Lee, S.E.; Choi, Y.-L. Diagnostic Value of MDM2 and DDIT3 Fluorescence In Situ Hybridization in Liposarcoma Classification: A Single-Institution Experience. Korean J. Pathol. 2012, 46, 115–122. [Google Scholar] [CrossRef]
  59. Kashima, T.; Halai, D.; Ye, H.; Hing, S.N.; Delaney, D.; Pollock, R.; O’Donnell, P.; Tirabosco, R.; Flanagan, A.M. Sensitivity of MDM2 amplification and unexpected multiple faint alphoid 12 (alpha 12 satellite sequences) signals in atypical lipomatous tumor. Mod. Pathol. 2012, 25, 1384–1396. [Google Scholar] [CrossRef] [Green Version]
  60. Mariño-Enríquez, A.; Fletcher, C.D.; Cin, P.D.; Hornick, J.L. Dedifferentiated Liposarcoma With “Homologous” Lipoblastic (Pleomorphic Liposarcoma-like) Differentiation: Clinicopathologic and Molecular Analysis of a Series Suggesting Revised Diagnostic Criteria. Am. J. Surg. Pathol. 2010, 34, 1122–1131. [Google Scholar] [CrossRef]
  61. Weaver, J.; Rao, P.; Goldblum, J.R.; Joyce, M.J.; Turner, S.L.; Lazar, A.J.; Lopez-Terada, D.; Tubbs, R.R.; Rubin, B.P. Can MDM2 analytical tests performed on core needle biopsy be relied upon to diagnose well-differentiated liposarcoma? Mod. Pathol. 2010, 23, 1301–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Shimada, S.; Ishizawa, T.; Ishizawa, K.; Matsumura, T.; Hasegawa, T.; Hirose, T. The value of MDM2 and CDK4 amplification levels using real-time polymerase chain reaction for the differential diagnosis of liposarcomas and their histologic mimickers. Hum. Pathol. 2006, 37, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  63. Pilotti, S.; Della Torre, G.; Mezzelani, A.; Tamborini, E.; Azzarelli, A.; Sozzi, G.; Pierotti, M.A. The expression of MDM2/CDK4 gene product in the differential diagnosis of well differentiated liposarcoma and large deep-seated lipoma. Br. J. Cancer 2000, 82, 1271–1275. [Google Scholar] [CrossRef] [PubMed]
  64. Wolff, A.C.; Hammond, M.E.; Schwartz, J.N.; Hagerty, K.L.; Allred, D.C.; Cote, R.J.; Dowsett, M.; Fitzgibbons, P.L.; Hanna, W.M.; Langer, A.; et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch. Pathol. Lab. Med. 2007, 131, 18–43. [Google Scholar] [CrossRef]
  65. Dashti, N.K.; Fritchie, K.J.; Folpe, A.L. Perinephric myxoid pseudotumor of fat: A distinctive pseudoneoplasm most often associated with non-neoplastic renal disease. Hum. Pathol. 2019, 87, 37–43. [Google Scholar] [CrossRef]
  66. Ryan, S.; Visgauss, J.; Kerr, D.; Helmkamp, J.; Said, N.; Vinson, E.; O’Donnell, P.; Li, X.; Jung, S.-H.; Cardona, D.; et al. The Value of MRI in Distinguishing Subtypes of Lipomatous Extremity Tumors Needs Reassessment in the Era of MDM2 and CDK4 Testing. Sarcoma 2018, 2018, 1901896. [Google Scholar] [CrossRef] [Green Version]
  67. Vicario, R.; Peg, V.; Morancho, B.; Zacarías-Fluck, M.; Zhang, J.; Martínez-Barriocanal, A.; Jiménez, A.N.; Aura, C.; Burgués, O.; Lluch, A.; et al. Patterns of HER2 Gene Amplification and Response to Anti-HER2 Therapies. PLoS ONE 2015, 10, e0129876. [Google Scholar] [CrossRef]
  68. Nuciforo, P.; Thyparambil, S.; Aura, C.; Garrido-Castro, A.; Vilaro, M.; Peg, V.; Jimenez, J.; Vicario, R.; Cecchi, F.; Hoos, W.; et al. High HER2 protein levels correlate with increased survival in breast cancer patients treated with anti-HER2 therapy. Mol. Oncol. 2015, 10, 138–147. [Google Scholar] [CrossRef] [Green Version]
  69. Rondón-Lagos, M.; Di Cantogno, L.V.; Rangel, N.; Mele, T.; Ramírez-Clavijo, S.R.; Scagliotti, G.; Marchio, C.; Sapino, A. Unraveling the chromosome 17 patterns of FISH in interphase nuclei: An in-depth analysis of the HER2 amplicon and chromosome 17 centromere by karyotyping, FISH and M-FISH in breast cancer cells. BMC Cancer 2014, 14, 922. [Google Scholar] [CrossRef] [Green Version]
  70. Starczynski, J.; Atkey, N.; Connelly, Y.; O’Grady, T.; Campbell, F.M.; di Palma, S.; Wencyk, P.; Jasani, B.; Gandy, M.; Bartlett, J.M.S. HER2 gene amplification in breast cancer: A rogues’ gallery of challenging diagnostic cases: UKNEQAS interpretation guidelines and research recommendations. Am. J. Clin. Pathol. 2012, 137, 595–605. [Google Scholar] [CrossRef]
Figure 1. MDM2 location and function. (A) MDM2 is located in the long arm of chromosome 12, region 1, and band 5 (12q15). (B) MDM2 is an E3 ubiquitin ligase that targets p53 for ubiquitylation and subsequent proteasomal degradation.
Figure 1. MDM2 location and function. (A) MDM2 is located in the long arm of chromosome 12, region 1, and band 5 (12q15). (B) MDM2 is an E3 ubiquitin ligase that targets p53 for ubiquitylation and subsequent proteasomal degradation.
Ijms 24 01342 g001
Figure 2. Patterns of MDM2 gene status by FISH assessment. MDM2 red signals and CEP12 green signals (Abbott molecular probes). Nuclei are counterstained with 4′-6-diamino-2-phenylindole and appear blue. (A) MDM2 amplification with MDM2 > 10 and CEP12 = 2. Notably, several MDM2 gene dot-like signals are scattered over the whole nucleus. Focal areas with overlapping signals are also present. Overall, this pattern is consistent with gene localization on Dmin. (B) MDM2 amplification with several areas with crowded, overlapping signals arranged in clusters over the whole nucleus and two CEP12 signals. This pattern is typical of Dmin amplification as well. (C) No MDM2 amplification nor CEP12 augmented copies are present in interphase of tumor cells’ nuclei (negative case). (D) An increased number of both MDM2 and CEP12 signals (3–4 copies) in a polysomic sample which resulted negative for MDM2 amplification. (E) A challenging sample with the presence of “giant nuclei”: a polysomic non-MDM2-amplified nucleus with more than ten MDM2 and CEP12 signals is shown in the top inset; for comparison, the lower inset presents an MDM2 amplified nucleus from another case with MDM2 ≥ 10 and a ratio > 2. (F) EGFR (red signals) and CEP7 (green signals) probes in “giant nuclei” (same case of 3E).
Figure 2. Patterns of MDM2 gene status by FISH assessment. MDM2 red signals and CEP12 green signals (Abbott molecular probes). Nuclei are counterstained with 4′-6-diamino-2-phenylindole and appear blue. (A) MDM2 amplification with MDM2 > 10 and CEP12 = 2. Notably, several MDM2 gene dot-like signals are scattered over the whole nucleus. Focal areas with overlapping signals are also present. Overall, this pattern is consistent with gene localization on Dmin. (B) MDM2 amplification with several areas with crowded, overlapping signals arranged in clusters over the whole nucleus and two CEP12 signals. This pattern is typical of Dmin amplification as well. (C) No MDM2 amplification nor CEP12 augmented copies are present in interphase of tumor cells’ nuclei (negative case). (D) An increased number of both MDM2 and CEP12 signals (3–4 copies) in a polysomic sample which resulted negative for MDM2 amplification. (E) A challenging sample with the presence of “giant nuclei”: a polysomic non-MDM2-amplified nucleus with more than ten MDM2 and CEP12 signals is shown in the top inset; for comparison, the lower inset presents an MDM2 amplified nucleus from another case with MDM2 ≥ 10 and a ratio > 2. (F) EGFR (red signals) and CEP7 (green signals) probes in “giant nuclei” (same case of 3E).
Ijms 24 01342 g002
Figure 3. Schematic representation of the probes used for MDM2 gene evaluation and the expected FISH pattern. CEP12: centromeric probe for chromosome 12; LSI MDM2: Locus Specific Identifier for MDM2 gene; LSI MDM2/CEP12: dual probe.
Figure 3. Schematic representation of the probes used for MDM2 gene evaluation and the expected FISH pattern. CEP12: centromeric probe for chromosome 12; LSI MDM2: Locus Specific Identifier for MDM2 gene; LSI MDM2/CEP12: dual probe.
Ijms 24 01342 g003
Figure 4. Criteria settled for FISH MDM2/CEP12 analysis. * Generally, cases with MDM2 amplification have a lot of MDM2 signals, so we think that an MDM2/CEP12 ratio > 2 is better than MDM2/CEP12 ≥ 2 as a diagnostic criterion. ** This parameter supports excluding cases with loss of CEP12 (CEP12 per nucleus < 2) as amplified.
Figure 4. Criteria settled for FISH MDM2/CEP12 analysis. * Generally, cases with MDM2 amplification have a lot of MDM2 signals, so we think that an MDM2/CEP12 ratio > 2 is better than MDM2/CEP12 ≥ 2 as a diagnostic criterion. ** This parameter supports excluding cases with loss of CEP12 (CEP12 per nucleus < 2) as amplified.
Ijms 24 01342 g004
Figure 5. Gene amplification patterns. Amplified DNA can be observed in two different patterns: (A) Gene amplification in extra chromosomal entities called double minutes (Dmins). Small fragments of extra chromosomal DNA scattered over the whole nucleus are commonly observed. This pattern is characteristic of the MDM2 gene. (B) Gene amplification in intrachromosomal entities called homogeneously staining regions (HSR). HSR are chromosomal segments of various lengths but uniform staining intensity. In the chromosomal region where HSR occur, a segment of the chromosome is amplified or duplicated several times. This pattern is the most common in HER2 amplified cases.
Figure 5. Gene amplification patterns. Amplified DNA can be observed in two different patterns: (A) Gene amplification in extra chromosomal entities called double minutes (Dmins). Small fragments of extra chromosomal DNA scattered over the whole nucleus are commonly observed. This pattern is characteristic of the MDM2 gene. (B) Gene amplification in intrachromosomal entities called homogeneously staining regions (HSR). HSR are chromosomal segments of various lengths but uniform staining intensity. In the chromosomal region where HSR occur, a segment of the chromosome is amplified or duplicated several times. This pattern is the most common in HER2 amplified cases.
Ijms 24 01342 g005
Figure 6. Matched histopathological features of the representative samples presented in Figure 2. Hematoxylin and eosin stain were used to evaluate the histological characteristics of the analyzed tumors and select representative areas for FISH analysis. (A) An MDM2-amplified DDLPS showing only focal lipomatous areas (the corresponding MDM2 amplification pattern is shown in Figure 2A). (B) An MDM2-amplified WDLPS (the MDM2 amplification pattern of this cases is represented in Figure 2B); (C,D) Representative images of two lipomas that were analyzed for MDM2 amplification (the corresponding negative MDM2 FISH patterns are represented in Figure 2C,D); (E) A non-MDM2-amplified undifferentiated pleomorphic sarcoma with giant cells (the corresponding MDM2 and EGFR FISH patterns are represented in Figure 2E (including the top inset) and Figure 2F, respectively); (F) An MDM2-amplified DDLPS with scattered giant cells (the corresponding MDM2 FISH pattern is provided in Figure 2E (bottom inset)).
Figure 6. Matched histopathological features of the representative samples presented in Figure 2. Hematoxylin and eosin stain were used to evaluate the histological characteristics of the analyzed tumors and select representative areas for FISH analysis. (A) An MDM2-amplified DDLPS showing only focal lipomatous areas (the corresponding MDM2 amplification pattern is shown in Figure 2A). (B) An MDM2-amplified WDLPS (the MDM2 amplification pattern of this cases is represented in Figure 2B); (C,D) Representative images of two lipomas that were analyzed for MDM2 amplification (the corresponding negative MDM2 FISH patterns are represented in Figure 2C,D); (E) A non-MDM2-amplified undifferentiated pleomorphic sarcoma with giant cells (the corresponding MDM2 and EGFR FISH patterns are represented in Figure 2E (including the top inset) and Figure 2F, respectively); (F) An MDM2-amplified DDLPS with scattered giant cells (the corresponding MDM2 FISH pattern is provided in Figure 2E (bottom inset)).
Ijms 24 01342 g006
Table 1. LPS variants/other lipomatous tumors and corresponding MDM2 amplification rate.
Table 1. LPS variants/other lipomatous tumors and corresponding MDM2 amplification rate.
Case SeriesMDM2 Amplification
AmplifiedNot AmplifiedRatio (%)
DDLPS25225/27 (93)
ALT/WDLPS18118/19 (95)
PLPS030/3 (0)
MLPS020/2 (0)
SCL/PL050/5 (0)
ASCLT020/2 (0)
Lipoma0250/25 (0)
Total434043/83 (52)
DDLPS: Dedifferentiated LPS; LPS: liposarcomas; ALT/WDLPS: Atypical Lipomatous Tumors/Well-differentiated LPS; PLPS: Pleomorphic LPS; MLPS: Myxoid LPS; SCL/PL: Spindle Cell Lipoma/Pleomorphic Lipoma; ASCLT: Atypical Spindle Cell Lipomatous Tumor.
Table 2. Selection of the most relevant published studies evaluating FISH MDM2 amplification. IHP: in-house probe; SP: single LSI MDM2 probe; DCP: dual color probe.
Table 2. Selection of the most relevant published studies evaluating FISH MDM2 amplification. IHP: in-house probe; SP: single LSI MDM2 probe; DCP: dual color probe.
Sample SizeFISH ProbesN° Nuclei EvaluatedAmplification Diagnostic Cut OffYear of PublicationReference
38DCP40MDM2/CEP12 ≥ 22022[38]
439DCP200MDM2/CEP12 ≥ 22022[31]
20DCP20MDM2/CEP12 ≥ 22022[32]
55DCPn.a.n.a.2021[39]
35DCPn.a.n.a.2021[33]
113DCPn.a.n.a.2019[40]
17DCP100MDM2/CEP12 > 22019[41]
180DCPn.a.MDM2/CEP12 ≥ 22018[42]
66DCP100MDM2/CEP12 ≥ 22018[43]
25DCPn.a.MDM2/CEP12 > 22018[44]
232DCP200n.a.2017[45]
101DCP40MDM2/CEP12 > 22017[46]
18DCPn.a.MDM2/CEP12 > 22017[47]
140IHP-DCP200MDM2/CEP12 ≥ 22016[48]
102DCP40MDM2/CEP12 ≥ 22016[49]
5DCP100MDM2/CEP12 > 2.02016[50]
5DCPn.a.MDM2 ≥ 3
CEP12 = 2
2016[51]
347DCPn.a.2–4 CEP12 signals with ≥6 extra MDM2 signals.2015[30]
10DCP100-200MDM2 ≥ 10
Polysomy CEP12: MDM2/CEP12 ≤ 2
2015[52]
77DCP60MDM2/CEP12 > 22015[53]
50SP40MDM2 > 5.02015[54]
347DCPn.a.MDM2 ≥ 6
CEP12 = 2–4
2015[30]
301DCP200At least 15% of nuclei presenting at least 15 MDM2 signals per cell2015[55]
46DCP60MDM2/CEP12 ≥ 22014[56]
64DCP100MDM2 ≥ 5
CEP12 = 1–2
2014[57]
172IHP-DCPn.a.MDM2/CEP12 > 22013[10]
38SPn.a.n.a.2013[1]
82DCP100MDM2/CEP12 ≥ 2.22012[58]
428DCP50MDM2/CEP12 > 2.02012[59]
12IHP-DCPn.a.n.a.2010[60]
54DCPn.a.MDM2/CEP12 ≥ 2.02010[61]
41IHP-DCP40MDM2/CEP12 ≥ 2.02009[9]
130IHP-DCP40MDM2/CEP12 ≥ 2.02008[8]
200SP100MDM2 > 5 signals/cell2007[11]
71IHP-DCP100MDM2/CEP12 > 32006[62]
21SPn.a.MDM2 > 2 signals/cell2000[63]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gambella, A.; Bertero, L.; Rondón-Lagos, M.; Verdun Di Cantogno, L.; Rangel, N.; Pitino, C.; Ricci, A.A.; Mangherini, L.; Castellano, I.; Cassoni, P. FISH Diagnostic Assessment of MDM2 Amplification in Liposarcoma: Potential Pitfalls and Troubleshooting Recommendations. Int. J. Mol. Sci. 2023, 24, 1342. https://doi.org/10.3390/ijms24021342

AMA Style

Gambella A, Bertero L, Rondón-Lagos M, Verdun Di Cantogno L, Rangel N, Pitino C, Ricci AA, Mangherini L, Castellano I, Cassoni P. FISH Diagnostic Assessment of MDM2 Amplification in Liposarcoma: Potential Pitfalls and Troubleshooting Recommendations. International Journal of Molecular Sciences. 2023; 24(2):1342. https://doi.org/10.3390/ijms24021342

Chicago/Turabian Style

Gambella, Alessandro, Luca Bertero, Milena Rondón-Lagos, Ludovica Verdun Di Cantogno, Nelson Rangel, Chiara Pitino, Alessia Andrea Ricci, Luca Mangherini, Isabella Castellano, and Paola Cassoni. 2023. "FISH Diagnostic Assessment of MDM2 Amplification in Liposarcoma: Potential Pitfalls and Troubleshooting Recommendations" International Journal of Molecular Sciences 24, no. 2: 1342. https://doi.org/10.3390/ijms24021342

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop