Skip to main content

Novel TNC-PDGFD fusion in fibrosarcomatous dermatofibrosarcoma protuberans: a case report

Abstract

Background

Dermatofibrosarcoma protuberans (DFSP) is a superficial fibroblastic tumor characterized by high rate of local recurrence and low metastatic potential. Fibrosarcomatous transformation can rarely arise in DFSP either de novo or as recurrent, which represents a form of tumor progression and carries an increased risk of metastasis over classic DFSP. Cytogenetically, DFSP is characterized by a recurrent unbalanced chromosome translocation t (17;22)(q22;q13), leading to the formation of COL1A1-PDGFB fusion transcript that is present in more than 90% of cases. Alternative fusions involving the PDGFD with partners of COL6A3 or EMILIN2 have recently been documented in less than 2% of cases. Herein, we report a DFSP with fibrosarcomtous morphology harboring a novel TNC-PDGFD fusion.

Case presentation

A 54-year-old female presented with a slowly growing mass in the right thigh. Excision demonstrated a 2-cm ovoid, well-circumscribed, gray-white, mass. Microscopic examination revealed a partially encapsulated subcutaneous nodule without dermal connection. The neoplasm was composed of cellular and fairly uniform spindle cells with brisk mitoses, arranged in elongated fascicles and herringbone patterns, with focal collagenized stroma. The neoplastic cells were positive for CD34 and smooth muscle actin. Fluorescence in-situ hybridization analyses showed negative for COL1A1-PDGFB fusion as well as NTRK1/2/3 rearrangements. A subsequent RNA sequencing detected an in-frame fusion between exon 15 of TNC and exon 6 of PDGFD. This fusion was further confirmed by nested reverse transcription polymerase chain reaction amplification followed by Sanger sequencing. A diagnosis of fibrosarcomatous DFSP was rendered and the patient was in good status at a follow-up of 12 months after the operation.

Conclusions

We report a fibrosarcomatous DFSP with novel TNC-PDGFD fusion, which adds to the pathologic and genetic spectrum of PDGFD-rearranged DFSP.

Background

Dermatofibrosarcoma protuberans (DFSP) is a locally aggressive but rarely metastasizing, fibroblastic neoplasm that typically presents as a nodular and multinodular cutaneous mass on the trunk and proximal extremities of young to middle-aged adults [1, 2]. Classically, DFSP is composed of fairly uniform, mildly atypical spindle cells, often arranged in tight storiform, whorled, or cartwheel patterns. It usually originates in the dermal with subsequent infiltration the subcutaneous fat with a characteristic honeycomb appearance. By immunohistochemistry (IHC), the neoplastic cells usually express CD34 with focal expression of smooth muscle actin (SMA) sometimes observed [1, 2]. Fibrosarcomatous transformation can rarely arise in DFSP either de novo or as recurrent, which represents a form of tumor progression and carries an increased risk of metastasis over classic DFSP [3, 4]. The fibrosarcomatous component varies in proportion from less than 5% to more than 95%, and often arises in the subcutis with a nodular, rather well-demarcated growth pattern [3, 4]. The tumor cells in fibrosarcomatous areas often arrange in fascicular and herringbone-like and frequently have greater cytologic atypia, increased cellularity and mitotic activity than those in the ordinary DFSP. By IHC, fibrosarcomatous DFSP commonly exhibits diminished, even loss of CD34 expression and increased P53 expression [3, 4]. Cytogenetically, the vast majority of DFSPs (including those with fibrosarcomatous change) harbor the recurrent unbalanced chromosome translocation t (17;22)(q21;q13), commonly in the form of supernumerary ring chromosomes, resulting in the fusion of genes COL1A1 on chromosome 17q21.3 and PDGFB on 22q13 [5, 6]. It has been proposed that constitutive expression of PDGFB is the fundamental mechanism of tumorigenesis in DFSP [5, 6]. However, rare variant fusion such as COLIA2-PDGFB fusion [7] and alternative rearrangements involving the related PDGFD gene with partners of either COL6A3 or EMILIN2 have also been documented recently [8, 9].

Herein, we report a fibrosarcomatous DFSP in which a novel fusion between TNC and PDGFD genes was detected by RNA sequencing and further confirmed by nested reverse transcription polymerase chain reaction (RT-PCR) amplification followed by Sanger sequencing .

Case presentation

A previously healthy 54-year-old female presented with a slowly growing mass in the right thigh for 1 year. Enhanced computed tomography scan demonstrated a 2.2 cm, well-defined, subcutaneous nodule in the right thigh with moderately heterogeneous enhancement. The tumor was surgically removed with narrow margins. No evidence of tumor recurrence or metastasis was noted at a follow-up of 12 months after the operation.

Gross examination revealed a 2 cm, well-circumscribed nodular mass with a firm, gray-white cut surface. Low power magnification showed a well-defined, partially encapsulated subcutaneous tumor without dermal connection (Fig. 1a). The tumor was composed of cellular, fairly uniform, slightly rounded spindle cells containing scant cytoplasm and ovoid, mildly atypical nuclei with brisk mitoses (up to 10 mitotic figures per 10 high power fields). The tumor cells were arranged predominantly in elongated fascicles and herringbone architectures (Fig. 1b, c). The stroma was typical minimal with scattered round small vessels and focal depositions of thick and band-like collagen bundles (Fig. 1d). No tumor necrosis was noted. At the periphery of the mass, minor areas showing vague storiform growth of less cellular and more bland-appearing tumor cells setting in a more collagenized stroma, with occasionally entrapped mature adipose tissues, were observed (Fig. 1e).

Fig. 1
figure 1

a Low-power view demonstrating a partially encapsulated nodular mass without dermal connection. b, c Cellular and mildly atypical spindle cells with brisk mitoses (arrows), arranging in elongated fascicles and herringbone patterns. d Focal collagenized stroma. e Minor areas displaying classic dermatofibrosarcoma protuberans with storiform growth and entrapment of adipocytes. f Immunoreactivity for CD34

Immunohistochemical staining revealed the tumor cells to be positive moderately and diffusely for CD34 (Fig. 1f) and focally for SMA and negative for Cam5.2, epithelial membrane antigen (EMA), desmin, calponin, TLE-1, CD99, Stat6, anaplastic lymphoma kinase (ALK, 1A4), SOX10, and S100 protein. The Ki67 proliferation index was approximately 20%. Fluorescence in-situ hybridization (FISH) analysis was negative for fusion of the COL1A1 and PDGFB using the dual spanning probe set (Fig. 2a). Assessment for rearrangements of the NTRK1/2/3 locus using the corresponding break-apart probe sets were all negative (Fig. 2b).

Fig. 2
figure 2

Fluorescence in-situ hybridization analyses revealing negative for (a) COL1A1-PDGFB fusion using the dual spanning probe set and (b) NTRK1 rearrangement with the break-apart probe set. (c) RNA sequencing suggesting a chromosomal translocation of TNC gene exon 15 on chromosome 9 with PDGFD gene exon 6 on chromosome 11

RNA sequencing was performed on formalin-fixed paraffin-embedded (FFPE) tissue as described previously [10]. Specifically, total RNA was extracted from FFPE samples with a RNeasy FFPE kit (QIAGEN). The quantity and quality of total RNA were assessed with the KAPA Library Quantification Kit (KAPA Biosystems), and the Agilent High Sensitivity DNA Kit and Bioanalyzer 2100 (Agilent Technologies), respectively, Sequencing was performed on the Illumina HiSeq next-generation sequencing platform (Illumina). The results were then analyzed using the BLAT aligner, Factera and Socrates, respectively, as previously described [10]. A fusion product between TNC exon 15 and PDGFD exon 6 with the variant allele frequency (VAF) of 85.37% was identified. The fusion result was confirmed through manually reviewing on the Integrative Genomics Viewer (Fig. 2c). This fusion was further confirmed by nested reverse transcription polymerase chain reaction (RT-PCR) amplification (TNC forward primer 5′-TGGCTACCGATGGGATCTTC − 3′ and PDGFD reverse primer 5′-CCGAGTAATTCCTGGGAGTGC-3′) followed by Sanger sequencing (Fig. 3).

Fig. 3
figure 3

Sanger sequencing of the nested RT-PCR amplification product further confirmed the TNC-PDGFD fusion transcript

Discussion

Historically DFSP has genetically been featured by t (17;22)(q22;q13) translocation, leading to the fusion of the COL1A1 gene with the PDGFB gene [5, 6]. The COLIA1-PDGFB fusion results in the constitutive up-regulation of PDGFB expression, leading to autocrine activation of PDGF receptor β (PDGFR-β) receptor tyrosine kinase signaling and consequently drives the tumorigenesis [2, 11]. These provide a rationale for targeted therapy with tyrosine kinase inhibitors (TKIs) for unresectable or metastatic DFSPs [12]. The COL1A1-PDGFB fusion has been detected in up to 96% of DFSPs and represents a quite useful tool for the differential diagnosis of DFSP with its mimickers [1, 2, 6, 7, 13]. With the application of more sensitive detection assays, the fusion incidence appears to increase and rare cryptic fusions and alternative rearrangements involving the related PDGFD gene have also been documented [7,8,9]. In the few cases of DFSP that were negative for COL1A1-PDGFB fusion through routine FISH analysis, two recent studies using Next generation sequencing found that 40% of cases indeed had the classical COL1A1-PDGFB fusion, while more than half of cases harbored a fusion between PDGFD and either COL6A3 or EMILIN2, with the COL6A3-PDGFD fusion much more frequently encountered than the EMILIN2-PDGFD fusion [8, 9]. In this report, we describe a novel TNC-PDGFD gene fusion in a DFSP with fibrosarcomatous morphology, enhancing the genetic spectrum of DFSP.

PDGFD, located at 11q22.3, encodes a protein belonging to the same family of platelet-derived growth factor as PDGFB [14]. It has been proposed that PDGFD displays an oncogenic activity specifically through binding to and activating its cognate receptor PDGFR-β, and plays an important role in regulating tumor cell growth, migration, invasion, angiogenesis and metastasis by cross-talk with many signaling pathways in a wide array of malignancies [14, 15]. In PDGFD-rearranged DFSP, the reported genomic breakpoint was constantly located in exon 6, which retained the PDGF domain in a manner similar to rearrangements involving PDGFB [5, 8, 9]. TNC, also known as Tenascin-C, located at 9q33.1, is a member of tenascin gene family and encodes an extracellular matrix glycoprotein TNC with a spatially and temporally restricted tissue distribution [16, 17]. TNC is homohexameric with disulfide-linked subunits, and contains multiple EGF-like and fibronectin type-III domains [16, 17]. TNC has oncogenic properties through promotion of cell proliferation, migration and angiogenesis and its over-expression has been linked to a variety of malignancies [18]. Recently, fusions involving TNC have rarely been documented to occur in other neoplasms, including TNC-NRG1 fusion in a non-small cell lung carcinoma [19] and in a papillary renal cell carcinoma [20], and TNC-USP6 fusion in a primary aneurysmal bone cyst [21]. In these scenarios, TNC is functioned as a strong promoter, leading to activation of oncogenes NRG1 and USP6, and subsequently induction of tumor formation.

According to the limited cases published to date, the PDGFD-rearranged DFSPs are commonly centered in subcutaneous fat without dermal involvement [8, 9], and the COL6A3-PDGFD fusion variant shows an apparent proclivity for the breast or chest wall locations in female patients and present with classic histology and immunophenotype [8], while both the two reported cases of DFSP harboring PDFGD-EMILINE2 fusion arise in the leg and demonstrate a fibrosarcomatous morphology [9]. Studies from 2 groups by Dickson et al. [8] and Dadone-Montaudié et al. [9] have showed that PDGFD-rearranged DFSP clustered together with the group of DFSP with the classic COL1A1-PDGFB fusion upon unsupervised hierarchical clustering analysis, and demonstrated increased expression of PDGFRB mRNA by RNA sequencing. These evidence suggested that PDGFD rearrangements may function in a similar pattern of autocrine activation via PDGFR-β receptor tyrosine kinase signaling as COL1A1-PDGFB fusions, and rearrangements of PDGFD might therefore be targeted by TKIs as classical DFSP [8, 9].

Fibrosarcomatous DFSP represents morphological progression to a usually fascicular and herringbone pattern with increased risk of recurrence and metastatic potential [1,2,3,4]. The underlying oncogenic mechanism of fibrosarcomatous transformation in conventional DFSP is largely undetermined. Several different molecular genetic alterations have been proposed to account for this transformation, including genomic gains of COL1A1-PDGFB, losses of genomic material from 22q, mutations of TP53, activation of the PDGFR-β/Akt/mTOR pathway signaling, and microsatellite instability [22]. Most recently, single reports have suggested that over-expression of programmed cell death 1 ligand (PD-L1) [23] and fusion of MAP 3K7CL-ERG [24] may be implicated in the transformation of conventional DFSP to fibrosarcomatous DFSP. It’s worth noting that both the previously reported two cases of EMILIN2-PDGFD fusion DFSP exhibited a fibrosarcomatous histology and showed homozygous deletion of CDKN2A [9], which had also been identified in PGDFB-rearranged DFSPs and often observed in cases showing hypercellularity and fibrosarcomatous transformation morphology [25]. These suggest that, despite limited experiences, disruptions of the CDKN2A/CDK4/RB1 pathway may also represent an oncogenic mechanism in the clonal evolution of a subset of PDGFD-rearranged DFSP with fibrosarcomatous transformation.

The case we described here harbors a novel TNC-PDGFD gene fusion, as detected by RNA sequencing and nested RT-PCR, and shares some features with EMILIN2-PDGFD fusion DFSP, including deep-seated without dermal connection, fibrsarcomatous transformation morphology with focally abundant collagenous stroma. Similar to other reported PFGFD fusions, the PDGF receptors DNA binding domain of PDGFD is preserved in the currently documented TNC-PDGFD fusion [8, 9]. As with the EMILIN2-PDGFD fusion, it is difficult to determine with certainty if TNC-PDGFD plays a driving role in the transformation of this tumor from DFSP to fibrosarcomatous DFSP, but it is a possible candidate. Larger cohorts with functional studies will be needed to further assess the role of TNC-PDGFD and the behavior of DFSP containing this fusion. With regard to the differential diagnosis of this tumor, NTRK-rearranged tumors may be a consideration given the monomorphic spindle cells, CD34 expression, and focally sclerotic background. However, NTRK-rearranged tumors typically co-express S100 protein, often harbor NTRK1 rearrangement although rare rearrangements involving NTRK2, NTRK3, RAF1, and BRAF have been documented [1] .

In summary, we report a fibrosarcomatous DFSP with novel TNC-PDGFD fusion, which adds to the pathologic and genetic spectrum of PDGFD-rearranged DFSP. The expanded molecular spectrum provides a novel insight into DFSP oncogenesis and carries important implications for molecular diagnostics as well as potential tailored therapies.

Availability of data and materials

Records and data pertaining to both the cases are in the patient’s secure medical records in Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College. All searched data by literature review are included in this paper.

Abbreviations

ALK:

anaplastic lymphoma kinase

DFSP:

dermatofibrosarcoma protuberans

EMA:

epithelial membrane antigen

FISH:

fluorescence in-situ hybridization

IHC:

immunohistochemistry

PDGFR-β:

PDGF receptor β

PD-L1:

programmed cell death 1 ligand

RT-PCR:

reverse transcription polymerase chain reaction

SMA:

smooth muscle actin

TKIs:

tyrosine kinase inhibitors

VAF:

variant allele frequency

References

  1. WHO Classification of Tumours Editorial Board. WHO Classification of Tumours of Soft Tissue and Bone. 5th ed. Lyon, France: IARC Press; 2020.

    Google Scholar 

  2. Thway K, Noujaim J, Jones RL, Fisher C. Dermatofibrosarcoma protuberans: pathology, genetics, and potential therapeutic strategies. Ann Diagn Pathol. 2016;25:64–71. https://0-doi-org.brum.beds.ac.uk/10.1016/j.anndiagpath.2016.09.013.

    Article  PubMed  Google Scholar 

  3. Mentzel T, Beham A, Katenkamp D, Dei Tos AP, Fletcher CD. Fibrosarcomatous (“high-grade”) dermatofibrosarcoma protuberans: clinicopathologic and immunohistochemical study of a series of 41 cases with emphasis on prognostic significance. Am J Surg Pathol. 1998;22(5):576–87. https://0-doi-org.brum.beds.ac.uk/10.1097/00000478-199805000-00009.

    Article  CAS  PubMed  Google Scholar 

  4. Abbott JJ, Oliveira AM, Nascimento AG. The prognostic significance of fibrosarcomatous transformation in dermatofibrosarcoma protuberans. Am J Surg Pathol. 2006;30(4):436–43. https://0-doi-org.brum.beds.ac.uk/10.1097/00000478-200604000-00002.

    Article  PubMed  Google Scholar 

  5. Simon MP, Pedeutour F, Sirvent N, Grosgeorge J, Minoletti F, Coindre JM, et al. Deregulation of the platelet-derived growth factor B-chain gene via fusion with collagen gene COL1A1 in dermatofibrosarcoma protuberans and giant-cell fibroblastoma. Nat Genet. 1997;15(1):95–8. https://0-doi-org.brum.beds.ac.uk/10.1038/ng0197-95.

    Article  CAS  PubMed  Google Scholar 

  6. Sirvent N, Maire G, Pedeutour F. Genetics of dermatofibrosarcoma protuberans family of tumors: from ring chromosomes to tyrosine kinase inhibitor treatment. Genes Chromosom Cancer. 2003;37(1):1–19. https://0-doi-org.brum.beds.ac.uk/10.1002/gcc.10202.

    Article  CAS  PubMed  Google Scholar 

  7. Nakamura I, Kariya Y, Okada E, Yasuda M, Matori S, Ishikawa O, et al. A novel chromosomal translocation associated with COL1A2-PDGFB gene fusion in dermatofibrosarcoma protuberans: PDGF expression as a new diagnostic tool. JAMA Dermatol. 2015;151(12):1330–7. https://0-doi-org.brum.beds.ac.uk/10.1001/jamadermatol.2015.2389.

    Article  PubMed  Google Scholar 

  8. Dickson BC, Hornick JL, Fletcher CDM, Demicco EG, Howarth DJ, Swanson D, et al. Dermatofibrosarcoma protuberans with a novel COL6A3-PDGFD fusion gene and apparent predilection for breast. Genes Chromosom Cancer. 2018;57(9):437–45. https://0-doi-org.brum.beds.ac.uk/10.1002/gcc.22663.

    Article  CAS  PubMed  Google Scholar 

  9. Dadone-Montaudié B, Alberti L, Duc A, Delespaul L, Lesluyes T, Pérot G, et al. Alternative PDGFD rearrangements in dermatofibrosarcomas protuberans without PDGFB fusions. Mod Pathol. 2018;31(11):1683–93. https://0-doi-org.brum.beds.ac.uk/10.1038/s41379-018-0089-4.

    Article  CAS  PubMed  Google Scholar 

  10. Xia QY, Wang XT, Fang R, et al. Clinicopathologic and molecular analysis of the TFEB fusion variant reveals new members of TFEB translocation renal cell carcinomas (RCCs): expanding the genomic Spectrum. Am J Surg Pathol. 2020;44(4):477–89. https://0-doi-org.brum.beds.ac.uk/10.1097/PAS.0000000000001408.

    Article  PubMed  Google Scholar 

  11. Greco A, Fusetti L, Villa R, Sozzi G, Minoletti F, Mauri P, et al. Transforming activity of the chimeric sequence formed by the fusion of collagen gene COL1A1 and the platelet derived growth factor b-chain gene in dermatofibrosarcoma protuberans. Oncogene. 1998;17(10):1313–9. https://0-doi-org.brum.beds.ac.uk/10.1038/sj.onc.1202051.

    Article  CAS  PubMed  Google Scholar 

  12. Saiag P, Grob JJ, Lebbe C, Malvehy J, del Marmol V, Pehamberger H, et al. Diagnosis and treatment of dermatofibrosarcoma protuberans. European consensus-based interdisciplinary guideline. Eur J Cancer. 2015;51(17):2604–8. https://0-doi-org.brum.beds.ac.uk/10.1016/j.ejca.2015.06.108.

    Article  PubMed  Google Scholar 

  13. Karanian M, Pérot G, Coindre JM, Chibon F, Pedeutour F, Neuville A. Fluorescence in situ hybridization analysis is a helpful test for the diagnosis of dermatofibrosarcoma protuberans. Mod Pathol. 2015;28(2):230–7. https://0-doi-org.brum.beds.ac.uk/10.1038/modpathol.2014.97.

    Article  CAS  PubMed  Google Scholar 

  14. Wang Z, Kong D, Li Y, Sarkar FH. PDGF-D signaling: a novel target in cancer therapy. Curr Drug Targets. 2009;10(1):38–41. https://0-doi-org.brum.beds.ac.uk/10.2174/138945009787122914.

    Article  PubMed  Google Scholar 

  15. Wang Z, Ahmad A, Li Y, Kong D, Azmi AS, Banerjee S, et al. Emerging roles of PDGF-D signaling pathway in tumor development and progression. Biochim Biophys Acta. 2010;1806(1):122–30. https://0-doi-org.brum.beds.ac.uk/10.1016/j.bbcan.2010.04.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Midwood KS, Chiquet M, Tucker RP, Orend G. Tenascin-C at a glance. J Cell Sci. 2016;129(23):4321–7. https://0-doi-org.brum.beds.ac.uk/10.1242/jcs.190546.

    Article  CAS  PubMed  Google Scholar 

  17. Chiovaro F, Chiquet-Ehrismann R, Chiquet M. Transcriptional regulation of tenascin genes. Cell Adhes Migr. 2015;9(1–2):34–47. https://0-doi-org.brum.beds.ac.uk/10.1080/19336918.2015.1008333.

    Article  CAS  Google Scholar 

  18. Yoshida T, Akatsuka T, Imanaka-Yoshida K. Tenascin-C and integrins in cancer. Cell Adhes Migr. 2015;9(1–2):96–104. https://0-doi-org.brum.beds.ac.uk/10.1080/19336918.2015.1008332.

    Article  CAS  Google Scholar 

  19. Jonna S, Feldman RA, Swensen J, Gatalica Z, Korn WM, Borghaei H, et al. Detection of NRG1 gene fusions in solid tumors. Clin Cancer Res. 2019;25(16):4966–72. https://0-doi-org.brum.beds.ac.uk/10.1158/1078-0432.CCR-19-0160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ptáková N, Martínek P, Holubec L, Janovský V, Vančurová J, Grossmann P, et al. Identification of tumors with NRG1 rearrangement, including a novel putative pathogenic UNC5D-NRG1 gene fusion in prostate cancer by data-drilling a de-identified tumor database. Genes Chromosom Cancer. 2021;60(7):474–81. https://0-doi-org.brum.beds.ac.uk/10.1002/gcc.22942.

    Article  CAS  PubMed  Google Scholar 

  21. Šekoranja D, Zupan A, Mavčič B, Martinčič D, Salapura V, Snoj Ž, et al. Novel ASAP1-USP6, FAT1-USP6, SAR1A-USP6, and TNC-USP6 fusions in primary aneurysmal bone cyst. Genes Chromosom Cancer. 2020;59(6):357–65. https://0-doi-org.brum.beds.ac.uk/10.1002/gcc.22836.

    Article  CAS  PubMed  Google Scholar 

  22. Iwasaki T, Yamamoto H, Oda Y. Current update on the molecular biology of cutaneous sarcoma: dermatofibrosarcoma protuberans. Curr Treat Options in Oncol. 2019;20(4):29. https://0-doi-org.brum.beds.ac.uk/10.1007/s11864-019-0628-3.

    Article  Google Scholar 

  23. Tsuchihashi K, Kusaba H, Yamada Y, Okumura Y, Shimokawa H, Komoda M, et al. Programmed death-ligand 1 expression is associated with fibrosarcomatous transformation of dermatofibrosarcoma protuberans. Mol Clin Oncol. 2017;6(5):665–8. https://0-doi-org.brum.beds.ac.uk/10.3892/mco.2017.1197.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Maloney N, Bridge JA, de Abreu F, Korkolopoulou P, Sakellariou S, Linos K. A novel MAP 3K7CL-ERG fusion in a molecularly confirmed case of dermatofibrosarcoma protuberans with fibrosarcomatous transformation. J Cutan Pathol. 2019;46(7):532–7. https://0-doi-org.brum.beds.ac.uk/10.1111/cup.13469.

    Article  PubMed  Google Scholar 

  25. Eilers G, Czaplinski JT, Mayeda M, Bahri N, Tao D, Zhu M, et al. CDKN2A/p16 loss implicates CDK4 as a therapeutic target in Imatinib-resistant dermatofibrosarcoma protuberans. Mol Cancer Ther. 2015;14(6):1346–53. https://0-doi-org.brum.beds.ac.uk/10.1158/1535-7163.MCT-14-0793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

The authors are grateful to Mr. Shao-yu Chen from Anbiping Laboratory, Guangzhou, China, for facilitating FISH, nested RT-PCR, and Sanger sequencing studies.

Funding

This study was supported by Zhejiang Provincial Natural Science Foundation (LY21H160052), and Zhejiang Provincial Medicine and Health Research Foundation (2018KY246, 2019KY020).

Author information

Authors and Affiliations

Authors

Contributions

YC, YS, MZ: conception and design of the work, acquisition, analysis and interpretation of data, drafting the manuscript and revising it critically for important intellectual content and scientific integrity. YC, YS, XF, XW, XH, MZ: acquisition, analysis and interpretation of data, reading and revising the manuscript critically for important intellectual content and scientific integrity. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Ming Zhao.

Ethics declarations

Ethics approval and consent to participate

Samples were used in accordance with ethical guidelines for the use of retrospective tissue samples provided by the local ethics committee of the Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College.

Consent for publication

Written informed consent was obtained from the patient for publication of this case report and any accompanying images. A copy of the consent form is available for review by the Editor of this journal.

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

Chen, Y., Shi, Yz., Feng, Xh. et al. Novel TNC-PDGFD fusion in fibrosarcomatous dermatofibrosarcoma protuberans: a case report. Diagn Pathol 16, 63 (2021). https://0-doi-org.brum.beds.ac.uk/10.1186/s13000-021-01123-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13000-021-01123-1

Keywords