Next Article in Journal
Liquid Biopsy as a New Tool for Diagnosis and Monitoring in Renal Cell Carcinoma
Previous Article in Journal
Impact of the Addition of Daratumumab to the Frontline Treatment of Patients with Immunoglobulin Light-Chain Amyloidosis: A Single-Centre Experience
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

TFCP2 Fusion-Positive Rhabdomyosarcomas: A Report of 10 Cases and a Review of the Literature

by
Madison P. Ginn
1,
Ryan A. Denu
2,
Davis R. Ingram
3,
Khalida M. Wani
3,
Alexander J. Lazar
3,4,
Douglas J. Harrison
5,
Michael S. Nakazawa
6,
Anthony P. Conley
6,
Shreyaskumar Patel
6 and
John Andrew Livingston
5,6,*
1
Departments of Internal Medicine & Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
2
Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
3
Department of Pathology, Division of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
4
Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
5
Department of Pediatrics Patient Care, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
6
Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(9), 1441; https://doi.org/10.3390/cancers17091441
Submission received: 7 March 2025 / Revised: 4 April 2025 / Accepted: 8 April 2025 / Published: 25 April 2025
(This article belongs to the Section Clinical Research of Cancer)

Simple Summary

Rhabdomyosarcomas with TFCP2 fusions comprise an extremely rare and aggressive type of cancer. In this paper, we detail the history and treatment of 10 patients with this rare sarcoma and compare them to 53 total reported cases in the literature. TFCP2 fusion sarcomas are associated with poor outcomes, high rates of recurrence, and treatment resistance. The median overall survival of our cohort was 24.7 months, the median time to recurrence following surgery was 2.1 months, and the median time to progression during treatment with chemotherapy was 1.6 months. As ALK alterations commonly co-occur in these sarcomas, two of our patients were treated with ALK inhibitors, with initial, although short-lived, benefits. More work must be conducted to determine the best treatment course for this disease.

Abstract

Background/Objectives: The fusion of the TFCP2 gene with either EWSR1 or FUS typically results in a spindle cell and/or epithelioid variant of rhabdomyosarcoma. This is an ultra-rare type of sarcoma, with most of our knowledge about these coming from case reports and small case series. Herein, we describe the clinical characteristics and treatment course of 10 patients with TFCP2 fusion sarcomas. Methods: We identified 10 patients in our hospital system with TFCP2 fusion sarcomas and 43 previously reported cases in the literature. We assessed primary tumor characteristics, treatment regimens, and survival rates among all cases. Results: We find that TFCP2 fusion sarcomas most commonly occur in young adults (median age: 33 years) and arise in craniofacial bones (7/10, 70%). Concomitant ALK alterations and ALK overexpression is nearly universal, and two of our patients were treated with ALK inhibitors; one patient had a near complete response before eventual progression, while the other patient had progressive disease after 2 months. For most, the prognosis was poor. The median overall survival in this cohort was 24.7 months (range: 5.9–29.7 months). Four patients were treated with upfront surgery, and all four developed recurrent disease. The median time to recurrence following upfront surgery was 2.1 months (range: 0.73–6.9 months). Five patients received systemic therapy, and the median progression-free survival from the start of treatment to progression was 1.6 months (range: 0.97–2.7). We also review the 53 total cases of TFCP2 fusion sarcomas in the literature, again highlighting the dismal outcomes in this disease. Conclusions: TFCP2 fusion sarcomas are proven to be aggressive and have poor prognosis. Additional work is needed to define the optimal treatment course for TFCP2 fusion sarcomas.

1. Introduction

TFCP2 (aliases LSF and SEF) was first described as an activator of the late SV40 promoter and later found to bind globin genes and HIV-1 promoters [1,2]. TFCP2 is part of the TFCP2/Grainyhead transcription factor family. It has been implicated as an oncogene in hepatocellular carcinoma [3], pancreatic ductal adenocarcinoma [4], and breast cancer [5], although it has been hypothesized to be a tumor suppressor in melanoma [6]. TFCP2 is ubiquitously expressed and involved in cell cycle regulation and the expression of lineage-specific genes [7].
The translocation of TFCP2 (usually exon 2) with either FET family member—EWSR1 (usually exon 5) or FUS (usually exon 6)—most often results in an epithelioid variant of rhabdomyosarcoma. The resulting fusion protein retains the CP2 DNA binding and the SAM/pointed domains of TFCP2 [8]. Pathologically, they are notable for significant ALK overexpression, as well as often expressing desmin, myogenin, MyoD1, and cytokeratin [9]. This is a rare orphan disease, so most of what is known comes from case reports and small case series. TFCP2-fusion sarcomas most commonly occur in adolescent and young adult women, although one case was reported in a 72-year-old person [10]. They most commonly arise in a craniofacial bone, such as the mandible, but have also been reported in the pelvis and chest wall [11,12,13,14]. TFCP2 fusion sarcomas are associated with extremely poor prognosis [14]. Patients who present with localized disease harbor a high risk of development of distant metastases (over 50%) [15]. The median overall survival has been reported to be 29 months [15]. Furthermore, responses are rare to traditional combination chemotherapy utilized for other rhabdomyosarcomas, and median progression-free survival on these treatments is about 2 months [16].
Herein, we report the clinical characteristics, treatments, and outcomes of 10 patients with TFCP2 fusion sarcomas from a single institution.

2. Methods

2.1. Study Design

This is a retrospective study of patients seen at MD Anderson Cancer Center from 2016–2024 with a TFCP2 fusion sarcoma. Electronic medical records from the identified patients were reviewed manually. The following clinical data were collected via retrospective chart review: age, gender, race, ethnicity, date of birth, date of diagnosis, vital status, tumor grade, tumor mitotic rate, tumor stage, surgery date, systemic therapies and dates of treatment, response to treatment, imaging results, personal history of cancer, family history of cancer, and results of next-generation sequencing tests. The response to treatment was determined based on the treating clinician’s determination and description in the electronic medical record.

2.2. Ethics

This study was approved by the University of Texas MD Anderson Cancer Center (MDACC) Institutional Review Board (protocol 2022-0278, approved 9 December 2022, and protocol DR09-0245, approved 26 May 2009) and was conducted in accordance with the U.S. Common Rule. Clinical and genomic data were obtained following signed informed consent onto prospective institutional protocols or under retrospective review protocols with a limited waiver of authorization.

2.3. Data Analysis and Statistics

Statistical analyses were performed using GraphPad Prism (version 9.5.0 or higher, RRID:SCR_002798) and R (version 4.2.2 or higher). The ggplot2 package was used to construct a swimmer plot.
Overall survival (OS) was calculated from the date of the first histologic diagnosis (either pre-treatment biopsy or surgical pathology) to the date of death or the latest follow-up (whichever occurred earlier). Recurrence-free survival (RFS) was calculated in patients with initially localized disease from the date of surgery for resection of the primary tumor to the date of recurrence or the latest follow-up (whichever occurred earlier). Progression-free survival (PFS) was defined in patients with metastatic disease from the start of therapy to the date of progression or the latest follow-up (whichever occurred earlier).

2.4. Data Availability

Deidentified data leading to the reported findings in this paper are available upon request from the corresponding author.

3. Results

3.1. Clinico-Pathologic Features of TFCP2 Fusion-Positive Sarcomas

We identified 10 patients with confirmed TFCP2 fusion-positive sarcomas (Table 1). Most exhibited a fascicular spindle cell morphology (Figure 1A) and stained positive for MyoD1 (Figure 1B), desmin (Figure 1C), and ALK (Figure 1D). Seven had the FUS-TFCP2 fusion, and three had the EWSR1-TFCP2 fusion (Figure 2A). Regarding sites of disease, seven (70%) arose in craniofacial bones, one (10%) started in the acetabulum, and two (20%) started in soft tissues of the trunk (Figure 2B). The mean and median ages of diagnosis were 30.5 and 33.0 years, respectively (range: 11.8–48.1 years; Figure 2C). The mean and median sizes of the primary tumor at diagnosis were both 6.8 cm (range: 2.8–11.6 cm; Figure 2D). Five (50%) were described by the pathologist as epithelioid and spindle cell rhabdomyosarcoma, three (30%) were described as a spindle cell/sclerosing rhabdomyosarcoma, one (10%) as a high-grade rhabdoid malignant tumor, and one (10%) as a small-cell sarcoma with rhabdomyogenic/rhabdomyosarcoma differentiation. These tumors were highly proliferative; five had at least 10 mitoses per 10 high-powered fields (hpf) and one had Ki67 of >80%. Most or all were positive for ALK (Figure 2E), cytokeratins (Figure 2F), desmin (Figure 2G), MyoD1 (Figure 2H), and myogenin (Figure 2I).
Six patients (60%) presented with localized disease and four (40%) with de novo metastatic disease (Figure 2J). Most patients developed locally recurrent disease (5/10, 50%). Lymph node metastases were reported in half (5/10, 50%) of the patients in our cohort (Table 1).

3.2. Treatment

Treatment and imaging data were available for 9 of the 10 patients. The first patient, a 21-year-old woman with TFCP2 fusion sarcoma of the left frontal bone (Figure 3A) was initially treated with surgery, developed local recurrence with invasion of the underlying brain parenchyma approximately 2 months later and underwent radiation (VMAT 64.2 Gy in 30 fractions). Three months later, she developed distant recurrence in the bilateral lungs and was treated with chemotherapy with VDC, although she developed progressive disease after two cycles. She was then treated with irinotecan plus temozolomide and had a mixed response, with response in the lungs but progression in the brain. The patient died shortly after.
The second patient is a 35-year-old male with EWSR1-TFCP2 fusion sarcoma of the right occipital bone with extension into the posterior cranial fossa and involvement of the right internal jugular vein and right internal carotid artery (Figure 3B). He was treated with gemcitabine plus docetaxel and achieved a mixed response. The next-generation sequencing panel showed an ALK inversion, so he was subsequently treated with lorlatinib; this was switched to alectinib after 1 week due to his insurance no longer covering lorlatinib. The patient initially had a positive response, and the patient’s symptoms also improved. However, after about 2 months he developed progressive disease. He was then treated with vincristine plus doxorubicin plus ifosfamide (VAI) and developed progressive disease after two cycles, with widespread metastases to the bilateral lungs and axial and appendicular skeleton. He was then treated with vincristine plus irinotecan plus temozolomide for four cycles, with an initial positive response after two cycles, but then progressed after four cycles. The patient died shortly thereafter.
The third patient is a 48-year-old female with FUS-TFCP2 fusion sarcoma that likely originated in the right hip but presented with de novo metastatic disease to the bilateral lungs, liver, lymph nodes, and bone (Figure 3C). She was treated with gemcitabine plus docetaxel but developed progressive disease after two cycles. She then received VDC for a total of five cycles, with a mixed response. She relocated and transitioned her care to another cancer center, although she died approximately 2 months later.
The fourth patient is a 30-year-old female with FUS-TFCP2 fusion sarcoma of the hard palate (Figure 3D). This was resected, and she subsequently developed local recurrence. She was treated initially with vincristine plus actinomycin D plus cyclophosphamide but stopped after one cycle due to poor tolerance. She was then treated with single-agent actinomycin D but only received one cycle again due to poor tolerance to therapy. She developed local progression and received three additional cycles of VAC but stopped due to toxicity. She was administered 60.4 Gy in 28 fractions to the primary tumor. Unfortunately, her cancer continued to progress, and she was unable to tolerate further therapy and passed away 4 months after completing radiation.
The fifth patient is a 13-year-old male with FUS-TFCP2 fusion sarcoma of the right maxilla (Figure 3E). He was treated with neoadjuvant VAC, although he developed progressive disease after 10 weeks and was switched to VDC/IE as per the Children’s Oncology Protocol, ARST0431, which achieved a partial response. He received radiation (41.4 Gy to right maxilla and neck with 9 Gy boost). After the completion of planned therapy, he continued to have gross disease, and he underwent resection with right total maxillectomy, right total ethmoidectomy, sphenoidotomy, frontal sinusotomy, right infratemporal fossa dissection, and fibula free flap reconstruction. Imaging 2 months following surgery showed concern for local recurrence, with cutaneous and subcutaneous nodules in the neck and periorbital area. He was started on lorlatinib and achieved a partial response after 3 months of treatment, with complete resolution of many of the subcutaneous and cutaneous nodules along the lateral aspect of the right face. He developed progressive disease after 3 more months and was switched to entrectenib, although he developed progressive disease and worsening pain after one cycle. With no viable treatment options and given that lorlatinib appeared to have slowed disease progression, lorlatinib was restarted, and he was treated with this for about 6 months before further progression. He is now receiving comfort-focused care.
The sixth patient is an 11-year-old girl that presented with left-sided neck pain and was found to have a sarcoma arising from C1 that had extended locally to the vertebral arteries and spinal cord (Figure 3F). She was treated with VAC alternating with vincristine and irinotecan but developed progression after two cycles. She received 50.4 Gy of radiation to the tumor, which did result in a decrease in the size of the tumor. She was then treated with lorlatinib for about one month, although the disease progressed rapidly during this time, and the patient died shortly afterwards.
The seventh patient is a 35-year-old man with tumor arising from paraspinal muscles near C7 (Figure 3G). He was treated with neoadjuvant vincristine, doxorubicin, and cyclophosphamide but stopped after restaging imaging after two cycles showed progressive disease. He then received radiation with 50 Gy to the tumor and was then started on crizotinib and subsequently underwent surgical resection. Crizotinib was held during the perioperative window and resumed. Three months later, he developed recurrent disease locally and new lung metastases.
The eighth patient (Figure 3H) is a 33-year-old woman that developed a sarcoma of the right alveolar ridge. PET showed FDG-avid right cervical lymphadenopathy. She was treated with neoadjuvant vincristine, dactinomycin, and cyclophosphamide for four cycles. Restaging imaging showed progressive disease and new metastases in the lungs. She was then treated with single-agent ifosfamide (10 g/m2) with concurrent radiation for pain control and received two cycles before developing progressive disease at the first restaging imaging. She was then treated with ipilimumab and nivolumab and received two cycles, at which point first restaging imaging showed progression in the head and neck, lymph nodes, lung, liver, and bone. One of the sites of disease was encasing and starting to narrow the right internal carotid, and radiation was planned. However, the patient declined and died quickly.
The ninth patient is a 40-year-old man who presented with a mass in his upper back (Figure 3I). He was treated with VAC for 13 weeks and then underwent wide local excision. Two weeks later, he developed local recurrence and resumed VAC. He had further progression and was switched to VDC. He was started on a clinical trial with local injection of an oncolytic herpes virus but developed progressive disease after three treatments.
A swimmer’s plot summarizing the treatment of each patient is shown in Figure 4. This demonstrates the timing of each treatment and the reason for changing treatment (progression or toxicity).

3.3. Clinical Outcomes

The median overall survival in this cohort was 16.7 months (range: 5.9–24.7 months after diagnosis; Figure 5A). Six patients were treated with upfront surgery, and all six developed recurrent disease. The median time to recurrence following upfront surgery was 2.8 months (range: 0.73–7.1 months; Figure 5B). Nine patients received systemic therapy, and the median progression-free survival from the start of treatment to progression was 1.7 months (range: 0.97–6.8; Figure 5C).

3.4. Review of the Literature

In reviewing the literature, we identified 53 cases of TFCP2 fusion sarcomas, including the 10 reported herein (Table 2). These were slightly more common in females (56.6%). The FUS-TFCP2 fusion was present in 63.0%, and the EWSR1-TFCP2 fusion was present in 37.0% (Figure 6A). There was a predilection for craniofacial bones (69.8%; Figure 6B). The tumors less commonly arose from other bony sites (18.9%) and soft tissues (11.3%). The mean and median ages of diagnosis were 31.5 and 30 years, respectively (Figure 6C). There was one outlier in a 72-year-old woman [16]. The mean and median tumor sizes were 6.3 cm and 6 cm, respectively (Figure 6D). Other reported alterations included ALK overexpression as well as ALK intragenic deletions and aberrant splicing and CDKN2A and MTAP deletions (Table 2) [15]. Regarding outcomes, the median overall survival for the 43 cases with reported outcomes was 17 months (Figure 6E). For the 17 cases with localized disease at presentation with reported outcomes, the median recurrence-free survival was 4 months (range: 0.73–28 months; Figure 6F). For the 25 cases with metastatic disease treated with systemic therapy and reported outcomes, the median progression-free survival was 2 months (range: 0.96–8 months; Figure 6G).
We assessed for potential differences in patient and tumor characteristics and outcomes based on the fusion partner (e.g., EWSR1 or FUS). Tumors with the EWSR1-TFCP2 fusion may be slightly more likely to arise from soft tissues than those with FUS-TFCP2 (Supplemental Figure S1A). There was no difference in fusion partner based on the patient gender or sex, age at diagnosis, or size of primary tumor. Furthermore, there were no significant differences in patient outcomes based on the fusion partner (Supplemental Figure S1).

4. Discussion

Spindle cell/sclerosing rhabdomyosarcomas typically harbor certain genomic alterations, including rearrangements involving TFCP2, VGLL2, NCOA2, CITED2, TEAD1, and SRF, as well as the MYOD1 p.L122R mutation [17,18,19]. There are also rarer molecular events that have been reported, such as a RAB3IP-HMGA2 fusion [20]. In the present study, we sought to understand the clinical characteristics, efficacy of treatments, and clinical outcomes in patients with TFCP2 fusion sarcomas. Analysis of the 10 cases from our institution along with the additional 43 cases identified in the literature allowed us to more comprehensively assess the clinical characteristics and outcomes of this orphan disease. In general, we find that TFCP2 fusion often results in an epithelioid and spindle cell rhabdomyosarcoma that most commonly affects young women. Sarcomas do not typically involve lymph nodes, with some exceptions in specific subtypes, such as epithelioid clear cell, angiosarcoma, synovial sarcomas, and rhabdomyosarcomas. However, several cases of TFCP2 fusion sarcoma reported in the literature have noted lymph node involvement [13,15], and 5 of the 10 patients described herein had lymph node involvement. Unfortunately, outcomes for this disease are extremely poor, leading to significant loss of life-years in this young population. More than half of patients with localized disease developed recurrent disease. Assessing for the fusion is critical for the diagnosis, as we encountered multiple cases in the literature that were initially misclassified as a different malignancy, including DSRCT [8].
A recent study performed multi-omics analysis of 12 cases of RMS with TFCP2 fusion to better understand the biology of this disease [15]. Six out of eleven patients with localized disease eventually developed distant metastases, mostly in the lungs and lymph nodes. Six patients received ALK inhibitors, and their responses were poor. RNA sequencing of these tumors was notable for elevated ALK expression. Exogenous expression of the ALK variants seen in TFCP2 fusion sarcomas in p53-deficient MCF10A untransformed human breast epithelial cell lines is sufficient for transformation. Furthermore, these alterations sensitized cells to ALK inhibitors, namely ceritinib. However, exogenous expression of the TFCP2 fusion is not sufficient for transformation but does block myogenic differentiation. This study also found that many TFCP2 fusion sarcomas have CDKN2A loss. Interestingly, the genomes were quite complex and showed a homologous recombination deficiency signature, which is unusual for fusion positive sarcoma and was more reminiscent of undifferentiated complex karyotype sarcomas. Therefore, immunotherapy may be worth trialing in this disease, although enthusiasm should be quelled by the limited efficacy of immunotherapy in other sarcomas with complex karyotypes, such as leiomyosarcoma [21].
What is the optimal way to treat TFCP2 fusion sarcomas? Unfortunately, these tumors have shown limited sensitivity to conventional chemotherapies. ALK alterations, namely inversions and intragenic deletions, as well as ALK protein overexpression seen via IHC, commonly co-occur with TFCP2 fusion [15], and the upregulation of ALK activity leads to increased cell proliferation and migration in multiple cancer types [22]. The development of targeted therapies with ALK inhibitors has shown positive outcomes in many tumor types, including other sarcomas such as inflammatory myofibroblastic tumors, where ALK inhibitors have shown benefits [23,24,25,26]. Despite the prevalence of ALK alterations and overexpression, most TFCP2 fusion sarcoma patients treated with ALK inhibitors have not responded well. One patient treated with crizotinib progressed rapidly after 10 days of treatment [27]. One patient in our study progressed rapidly on lorlatinib. However, one patient in our cohort did have a near complete response, and the total duration of clinical benefit with lorlatinib was about 6 months. Future work will need to focus on the mechanisms of resistance to ALK inhibition. Borrowing from the lung cancer literature, where ALK alterations are more commonly seen, the mechanisms of resistance to ALK inhibitors include acquired resistance mutations in ALK; ALK gene amplification; the activation of downstream or bypass signaling pathways, including EGFR, MET, and MAPK; and the expression of drug efflux pumps [28,29,30,31]. These mechanisms were largely discerned with genomic profiling following resistance. However, these data are lacking in TFCP2-fusion sarcomas and are a critical area of future investigation.
Other potential ideas for future trials in this disease may come from studying the concurrent genetic aberrations, namely CDKN2A and MTAP co-deletion [15]. CDKN2A is a known tumor suppressor, and its loss may render sensitivity to CDK4/6 inhibitors. MTAP is a gene adjacent to CDKN2A on chromosome 9p and is frequently co-deleted with CDKN2A in a wide range of cancers. MTAP is an enzyme involved in the salvage of methionine and adenine. Preclinical studies identified that PRMT5 inhibition is synthetic lethal with MTAP loss [32]. Furthermore, MTAP loss also renders sensitivity to antifolates such as pemetrexed. In a single-arm phase 2 trial in urothelial carcinoma, 3 out of 7 patients showed a response to pemetrexed (ORR 43%). In addition, analysis of a historic cohort showed 4 out of 4 MTAP-deficient tumors responded to pemetrexed compared to 1/10 MTAP proficient [33]. Therefore, PRMT5 inhibitors and antifolate drugs may be potential therapeutic options to pursue in TFCP2 fusion sarcomas with MTAP deletion. Furthermore, given the reported homologous recombination deficiency signature seen [15], this may suggest trialing platinum-based chemotherapy or DNA damage pathway inhibitors such as PARP inhibitors. Lastly, small molecules have been developed that may interfere with TFCP2 binding to DNA [34,35] and may be of therapeutic interest.
There are several limitations to our study. Due to the rarity of this disease, we have a small sample size, which limits the ability to make statistically significant conclusions, although to mitigate this, we incorporated analyses of all previously published cases. This disease does not have a standardized treatment regimen and, thus, provides another layer of complexity in comparing responses to treatments, as each patient often has a unique and personalized treatment course. Next, there is potential bias in patient selection, as we included only patients seen at our institution, a large tertiary referral center.

5. Conclusions

In summary, we report our experience with 10 cases of TFCP2 fusion sarcomas and review the literature of 53 total cases. This particular subtype of sarcoma is associated with a dismal prognosis in a young population, resulting in significant loss of life-years. Our clinical data may serve as a benchmark against the efficacy of future novel therapies. ALK inhibitors have been largely unsuccessful, although one patient at our institution derived significant clinical benefit from lorlatinib. There is great need to develop novel treatment strategies for this orphan disease, as well as further investigate the genetic profiles of these tumors to determine prognostic factors, oncogenic drivers, and directed therapy targets.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cancers17091441/s1: Figure S1. Outcomes based on fusion partner.

Author Contributions

M.P.G.: conceptualization, data curation, formal analysis, investigation, writing—original draft, and writing—review and editing. R.A.D.: conceptualization, data curation, formal analysis, investigation, methodology, visualization, writing—original draft, and writing—review and editing. D.R.I.: data curation, investigation, methodology, and writing—review and editing. K.M.W.: data curation, investigation, methodology, and writing—review and editing. M.S.N.: investigation, methodology, and writing—review and editing. A.J.L.: conceptualization, resources, supervision, and writing—review and editing. D.J.H.: data curation and writing—review and editing. A.P.C.: data curation and writing—review and editing. S.P.: conceptualization, investigation, supervision, and writing—review and editing. J.A.L.: conceptualization, investigation, supervision, and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the NIH (P30 CA016672; T32 CA009666) and a Conquer Cancer—Endowed Young Investigator Award in Honor of Grant R. and Victoria A. Merryman. Any opinions, findings, and conclusions expressed in this material are those of the author(s) and do not necessarily reflect those of the American Society of Clinical Oncology® or Conquer Cancer®.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of the University of Texas MD Anderson Cancer Center (protocol DR09-0245, approved 26 May 2009; protocol LAB04-0890, approved 29 November 2004).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The raw data supporting the conclusions of this article may be made available by the authors on reasonable request to the corresponding author.

Acknowledgments

Any opinions, findings, and conclusions expressed in this material are those of the author(s) and do not necessarily reflect those of the American Society of Clinical Oncology® or Conquer Cancer®.

Conflicts of Interest

M.P.G. reports no conflicts of interest. R.A.D. reports no conflicts of interest. D.R.I. reports no conflicts of interest. K.M.W. reports no conflicts of interest. A.P.C. reports research funding from Eli Lilly, Epicentrx, Chordoma Foundation, Inhibrx, Kronos Bio, Krystal Biotech, NCI, and Roche and consultancy or advisory board participation for Aadi Biosciences, Applied Clinical Intelligence (DSMB), Deciphera, and Inhibrx. A.J.L. reports consulting and/or advisory board relationships with the following entities: AbbVie, Adaptimmune, AJCC, Astra-Zeneca, Bain Capital, Bayer, Bio-AI Health, BMS, CAP, Caris, Deciphera, Elsevier, Foghorn Therapeutics, Gothams, GSK, Illumina, Invitae/Archer DX, Iterion Therapeutics, Merck, Novartis, Nucleai, OncoKB (MSKCC), Paige, Pfizer, Regeneron, Roche/Genentech, SpringerNature, SpringWorks, Tempus, ThermoFisher, and USCAP. M.S.N. reports no conflicts of interest. S.P. reports consulting for Deciphera. J.A.L reports funding from Repare Therapeutics.

Abbreviations

IEIfosfamide and etoposide
VACVincristine, actinomycin, and cyclophosphamide
VAIVincristine, doxorubicin, and ifosfamide
VDCVincristine, doxorubicin, and cyclophosphamide

References

  1. Swendeman, S.L.; Spielholz, C.; Jenkins, N.A.; Gilbert, D.J.; Copeland, N.G.; Sheffery, M. Characterization of the genomic structure, chromosomal location, promoter, and development expression of the alpha-globin transcription factor CP2. J. Biol. Chem. 1994, 269, 11663–11671. [Google Scholar] [CrossRef] [PubMed]
  2. Yoon, J.B.; Li, G.; Roeder, R.G. Characterization of a family of related cellular transcription factors which can modulate human immunodeficiency virus type 1 transcription in vitro. Mol. Cell. Biol 1994, 14, 1776–1785. [Google Scholar] [PubMed]
  3. Yoo, B.K.; Emdad, L.; Gredler, R.; Fuller, C.; Dumur, C.I.; Jones, K.H.; Jackson-Cook, C.; Su, Z.; Chen, D.; Saxena, U.; et al. Transcription factor Late SV40 Factor (LSF) functions as an oncogene in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2010, 107, 8357–8362. [Google Scholar] [CrossRef]
  4. Dai, Y.; Cai, Y.; Zhao, J.; Liu, H.; Wang, Y.; Liu, H.; Zhang, D. TFCP2 activates beta-catenin/TCF signaling in the progression of pancreatic cancer. Oncotarget 2017, 8, 70538–70549. [Google Scholar]
  5. Mehner, C.; Hockla, A.; Miller, E.; Ran, S.; Radisky, D.C.; Radisky, E.S. Tumor cell-produced matrix metalloproteinase 9 (MMP-9) drives malignant progression and metastasis of basal-like triple negative breast cancer. Oncotarget 2014, 5, 2736–2749. [Google Scholar] [CrossRef]
  6. Goto, Y.; Yajima, I.; Kumasaka, M.; Ohgami, N.; Tanaka, A.; Tsuzuki, T.; Inoue, Y.; Fukushima, S.; Ihn, H.; Kyoya, M.; et al. Transcription factor LSF (TFCP2) inhibits melanoma growth. Oncotarget 2016, 7, 2379–2390. [Google Scholar] [CrossRef]
  7. Veljkovic, J.; Hansen, U. Lineage-specific and ubiquitous biological roles of the mammalian transcription factor LSF. Gene 2004, 343, 23–40. [Google Scholar] [CrossRef]
  8. Watson, S.; Perrin, V.; Guillemot, D.; Reynaud, S.; Coindre, J.; Karanian, M.; Guinebretiere, J.; Freneauz, P.; Le Loarer, F.; Bouvet, M.; et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J. Pathol. 2018, 245, 29–40. [Google Scholar] [CrossRef]
  9. Xu, B.; Suurmeijer, A.J.H.; Agaram, N.P.; Zhang, L.; Antonescu, C.R. Head and neck rhabdomyosarcoma with TFCP2 fusions and ALK overexpression: A clinicopathological and molecular analysis of 11 cases. Histopathology 2021, 79, 347–357. [Google Scholar] [CrossRef]
  10. Dashti, N.K.; Wehrs, R.N.; Thomas, B.C.; Nair, A.; Davila, J.; Buckner, J.C.; Martinez, A.P.; Sukov, W.; Halling, K.C.; Howe, B.M.; et al. Spindle cell rhabdomyosarcoma of bone with FUS-TFCP2 fusion: Confirmation of a very recently described rhabdomyosarcoma subtype. Histopathology 2018, 73, 514–520. [Google Scholar] [CrossRef]
  11. Le Loarer, F.; Cleven, A.H.G.; Bouvier, C.; Castex, M.; Romagosa, C.; Moreau, A.; Salas, S.; Bonhomme, B.; Gomez-Brouchet, A.; Laurent, C.; et al. A subset of epithelioid and spindle cell rhabdomyosarcomas is associated with TFCP2 fusions and common ALK upregulation. Mod. Pathol. 2020, 33, 404–419. [Google Scholar] [CrossRef] [PubMed]
  12. Chrisinger, J.S.A.; Wehrli, B.; Dickson, B.; Fasih, S.; Hirbe, A.C.; Shultz, D.B.; Zadeh, G.; Gupta, A.A.; Demicco, E.G. Epithelioid and spindle cell rhabdomyosarcoma with FUS-TFCP2 or EWSR1-TFCP2 fusion: Report of two cases. Virchows Arch. 2020, 477, 725–732. [Google Scholar] [CrossRef] [PubMed]
  13. Koutlas, I.G.; Olson, D.R.; Rawwas, J. FET(EWSR1)-TFCP2 Rhabdomyosarcoma: An Additional Example of this Aggressive Variant with Predilection for the Gnathic Bones. Head Neck Pathol. 2021, 15, 374–380. [Google Scholar] [CrossRef] [PubMed]
  14. Sivakumar, N.; Sharma, P.; Chandra, S.; Gupta, S.; Samadi, F.M.; Baghel, S. Clinicopathological and Molecular Characteristics of Intraosseous Rhabdomyosarcoma Involving Head and Neck Region: A Systematic Review and Meta-Analysis. Pediatr. Dev. Pathol. 2023, 26, 299–309. [Google Scholar] [CrossRef]
  15. Schöpf, J.; Uhrig, S.; Heilig, C.E.; Lee, K.; Walther, T.; Carazzato, A.; Dobberkau, A.M.; Weichenhan, D.; Plass, C.; Hartmann, M.; et al. Multi-omic and functional analysis for classification and treatment of sarcomas with FUS-TFCP2 or EWSR1-TFCP2 fusions. Nat. Commun. 2024, 15, 51. [Google Scholar] [CrossRef]
  16. Tagami, Y.; Sugita, S.; Kubo, T.; Iesato, N.; Emori, M.; Takada, K.; Tsujiwaki, M.; Segawa, K.; Sugawara, T.; Kikuchi, T.; et al. Spindle cell rhabdomyosarcoma in a lumbar vertebra with FUS-TFCP2 fusion. Pathol. Res. Pract. 2019, 215, 152399. [Google Scholar] [CrossRef]
  17. Alaggio, R.; Zhang, L.; Sung, Y.-S.; Huang, S.-C.; Chen, C.-L.; Bisogno, G.; Zin, A.; Agaram, N.P.; LaQuaglia, M.P.; Wexler, L.H.; et al. A Molecular Study of Pediatric Spindle and Sclerosing Rhabdomyosarcoma: Identification of Novel and Recurrent VGLL2-Related Fusions in Infantile Cases. Am. J. Surg. Pathol. 2016, 40, 224–235. [Google Scholar] [CrossRef]
  18. Rekhi, B.; Upadhyay, P.; Ramteke, M.P.; Dutt, A. MYOD1 (L122R) Mutations Are Associated with Spindle Cell and Sclerosing Rhabdomyosarcomas with Aggressive Clinical Outcomes. Mod. Pathol. 2016, 29, 1532–1540. [Google Scholar] [CrossRef]
  19. Dehner, C.A.; Broski, S.M.; Meis, J.M.; Murugan, P.; Chrisinger, J.S.A.; Sosa, C.; Petersen, M.; Halling, K.C.; Gupta, S.; Folpe, A.L. Fusion-Driven Spindle Cell Rhabdomyosarcomas of Bone and Soft Tissue: A Clinicopathologic and Molecular Genetic Study of 25 Cases. Mod. Pathol. 2023, 36, 100271. [Google Scholar] [CrossRef]
  20. De Vita, A.; Ferrari, A.; Miserocchi, G.; Vanni, S.; Domizio, C.; Fonzi, E.; Fausti, V.; Recine, F.; Bassi, M.; Campobassi, A.; et al. Identification of a novel RAB3IP-HMGA2 fusion transcript in an adult head and neck rhabdomyosarcoma. Oral Dis. 2022, 28, 2052–2054. [Google Scholar] [CrossRef]
  21. Tawbi, H.A.; Burgess, M.; Bolejack, V.; Van Tine, B.A.; Schuetze, S.M.; Hu, J.; D’Angelo, S.; Attia, S.; Riedel, R.F.; Priebat, D.A.; et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): A multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
  22. Zhao, Z.; Verma, V.; Zhang, M. Anaplastic lymphoma kinase: Role in cancer and therapy perspective. Cancer Biol. Ther. 2015, 16, 1691–1701. [Google Scholar] [CrossRef] [PubMed]
  23. Theilen, T.M.; Soerensen, J.; Bochennek, K.; Becker, M.; Schwabe, D.; Rolle, U.; Klingebiel, T.; Lehrnbecher, T. Crizotinib in ALK+ inflammatory myofibroblastic tumors—Current experience and future perspectives. Pediatr. Blood Cancer 2018, 65, 26920. [Google Scholar] [CrossRef] [PubMed]
  24. Felkai, L.; Bánusz, R.; Kovalszky, I.; Sapi, Z.; Garami, M.; Papp, G.; Karaszi, K.; Varga, E.; Csoka, M. The Presence of ALK Alterations and Clinical Relevance of Crizotinib Treatment in Pediatric Solid Tumors. Pathol. Oncol. Res. 2019, 25, 217–224. [Google Scholar] [CrossRef]
  25. Debonis, S.A.; Bongiovanni, A.; Pieri, F.; Fausti, V.; De Vita, A.; Riva, N.; Gurrieri, L.; Vanni, S.; Diano, D.; Mercatali, L.; et al. ALK-negative lung inflammatory myofibroblastic tumor in a young adult: A case report and literature review of molecular alterations. Medicine 2021, 100, 25972. [Google Scholar] [CrossRef]
  26. Butrynski, J.E.; D’Adamo, D.R.; Hornick, J.L.; Dal Cin, P.; Antonescu, C.R.; Jhanwar, S.C.; Ladanyi, M.; Capelletti, M.; Rodig, S.J.; Ramaiya, N.; et al. Crizotinib in ALK-Rearranged Inflammatory Myofibroblastic Tumor. N. Engl. J. Med. 2010, 363, 1727–1733. [Google Scholar] [CrossRef]
  27. Panferova, A.; Sinichenkova, K.Y.; Abu Jabal, M.; Usman, N.; Sharlai, A.; Roshchin, V.; Konovalov, D.; Druy, A. EWSR1-TFCP2 in an adolescent represents an extremely rare and aggressive form of intraosseous spindle cell rhabdomyosarcomas. Cold Spring Harb. Mol. Case Stud. 2022, 8, a006209. [Google Scholar] [CrossRef]
  28. Wang, W.; Xu, C.; Zhu, Y.; Liao, X.; Zhuang, W.; Du, K.; Chen, R.; Chen, Y.; Chen, G.; Fang, M. Patients Harboring ALK Rearrangement Adenocarcinoma after Acquired Resistance to Crizotinib and Transformation to SCLC: A Case Report. Oncotargets Ther. 2017, 10, 3187–3192. [Google Scholar] [CrossRef]
  29. Ignatius Ou, S.; Lee, T.K.; Young, L.; Fernandez-Rocha, M.Y.; Pavlick, D.; Schrock, A.B.; Zhu, V.W.; Milliken, J.; Ali, S.M.; Gitlitz, B.J. Dual occurrence of ALK G1202R solvent front mutation and small cell lung cancer transformation as resistance mechanisms to second generation ALK inhibitors without prior exposure to crizotinib. Pitfall of solely relying on liquid re-biopsy? Lung Cancer 2017, 106, 110–114. [Google Scholar]
  30. Dardaei, L.; Wang, H.Q.; Singh, M.; Fordjour, P.; Shaw, K.X.; Yoda, S.; Kerr, G.; Yu, K.; Liang, J.; Cao, Y. SHP2 inhibition restores sensitivity in ALK-rearranged non-small-cell lung cancer resistant to ALK inhibitors. Nat. Med. 2018, 24, 512–519. [Google Scholar] [CrossRef]
  31. Pan, Y.; Deng, C.; Qiu, Z.; Cao, C.; Wu, F. The Resistance Mechanisms and Treatment Strategies for ALK-Rearranged Non-Small Cell Lung Cancer. Front. Oncol. 2021, 11, 713530. [Google Scholar] [CrossRef] [PubMed]
  32. Kryukov, G.V.; Wilson, F.H.; Ruth, J.R.; Paulk, J.; Tsherniak, A.; Marlow, S.E.; Vazquez, F.; Weir, B.A.; Fitzgerald, M.E.; Tanaka, M.; et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science 2016, 351, 1214–1218. [Google Scholar] [CrossRef] [PubMed]
  33. Alhalabi, O.; Chen, J.; Zhang, Y.; Lu, Y.; Wang, Q.; Ramachandran, S.; Tidwell, R.S.; Han, G.; Yan, X.; Meng, J.; et al. MTAP deficiency creates an exploitable target for antifolate therapy in 9p21-loss cancers. Nat. Commun. 2022, 13, 1797. [Google Scholar] [CrossRef] [PubMed]
  34. Grant, T.J.; Bishop, J.A.; Christadore, L.M.; Barot, G.; Chin, H.G.; Woodson, S.; Kavouris, J.; Siddiq, A.; Gredler, R.; Shen, X.; et al. Antiproliferative small-molecule inhibitors of transcription factor LSF reveal oncogene addiction to LSF in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2012, 109, 4503–4508. [Google Scholar] [CrossRef]
  35. Rajasekaran, D.; Siddiq, A.; Willoughby, J.L.; Biagi, J.M.; Christadore, L.M.; Yunes, S.A.; Gredler, R.; Jariwala, N.; Robertson, C.L.; Akiel, M.A.; et al. Small molecule inhibitors of Late SV40 Factor (LSF) abrogate hepatocellular carcinoma (HCC): Evaluation using an endogenous HCC model. Oncotarget 2015, 6, 26266–26277. [Google Scholar] [CrossRef]
Figure 1. Pathologic features of TFCP2 fusion sarcomas. (A) Representative H&E image of a TFCP2 fusion-positive spindle cell/sclerosing rhabdomyosarcoma. (BD) Representative images of immunohistochemical staining for MyoD1 (B), desmin (C), and ALK (D). Scale bars = 100 μm.
Figure 1. Pathologic features of TFCP2 fusion sarcomas. (A) Representative H&E image of a TFCP2 fusion-positive spindle cell/sclerosing rhabdomyosarcoma. (BD) Representative images of immunohistochemical staining for MyoD1 (B), desmin (C), and ALK (D). Scale bars = 100 μm.
Cancers 17 01441 g001
Figure 2. Clinical and pathologic characteristics of TFCP2 fusion sarcomas. (A) Percent of patients with each of the two TFCP2 fusions, either FUS-TFCP2 or EWSR1-TFCP2. (B) Breakdown of the site of disease. (C) Distribution of the age at diagnosis. (D) Distribution of the size of the sarcoma at diagnosis. In (C,D), bars represent means ± standard deviations, and each black dot represents a different patient. (EI) Percent of tumors that stained positive for ALK (E), cytokeratins (F), desmin (G), MyoD1 (H), and myogenin (I). (J) Disease stage at diagnosis. The numbers on top of the bars indicate the absolute number of patients represented by the bar.
Figure 2. Clinical and pathologic characteristics of TFCP2 fusion sarcomas. (A) Percent of patients with each of the two TFCP2 fusions, either FUS-TFCP2 or EWSR1-TFCP2. (B) Breakdown of the site of disease. (C) Distribution of the age at diagnosis. (D) Distribution of the size of the sarcoma at diagnosis. In (C,D), bars represent means ± standard deviations, and each black dot represents a different patient. (EI) Percent of tumors that stained positive for ALK (E), cytokeratins (F), desmin (G), MyoD1 (H), and myogenin (I). (J) Disease stage at diagnosis. The numbers on top of the bars indicate the absolute number of patients represented by the bar.
Cancers 17 01441 g002
Figure 3. Patients with TFCP2 fusion sarcomas. (A) Patient 1, CT (left) and MRI (right) images from initial diagnosis showing left frontal bone tumor. (B) Patient 2, MRI demonstrating involvement of the right IJV and right ICA and extension into the posterior cranial fossa. (C) Patient 3, CT images at the time of diagnosis showing presumed primary tumor in right acetabulum (left), additional diffuse spinal metastases (middle), and diffuse hepatic metastases (right). (D) Patient 4, PET/CT showing primary FDG-avid mass of the hard palate at the time of diagnosis (left) and an image of the patient after development of recurrent disease (right). (E) Patient 5, CT images showing large tumor in axial (left), coronal (middle), and sagittal (right) planes. (F) Patient 6, sagittal (left) and coronal and sagittal (right) MRI images showing mass arising from C1. (G) Patient 7, axial and sagittal MRI images of mass arising from C7. (H) Patient 8, CT images of mass arising in right alveolar ridge in bone window (left, middle) and lymphadenopathy in soft tissue window (right). (I) Patient 9, CT at time of diagnosis (left) and local recurrence after surgery (right) with cervical lymphadenopathy (arrowhead). Red arrows highlight location of the tumors.
Figure 3. Patients with TFCP2 fusion sarcomas. (A) Patient 1, CT (left) and MRI (right) images from initial diagnosis showing left frontal bone tumor. (B) Patient 2, MRI demonstrating involvement of the right IJV and right ICA and extension into the posterior cranial fossa. (C) Patient 3, CT images at the time of diagnosis showing presumed primary tumor in right acetabulum (left), additional diffuse spinal metastases (middle), and diffuse hepatic metastases (right). (D) Patient 4, PET/CT showing primary FDG-avid mass of the hard palate at the time of diagnosis (left) and an image of the patient after development of recurrent disease (right). (E) Patient 5, CT images showing large tumor in axial (left), coronal (middle), and sagittal (right) planes. (F) Patient 6, sagittal (left) and coronal and sagittal (right) MRI images showing mass arising from C1. (G) Patient 7, axial and sagittal MRI images of mass arising from C7. (H) Patient 8, CT images of mass arising in right alveolar ridge in bone window (left, middle) and lymphadenopathy in soft tissue window (right). (I) Patient 9, CT at time of diagnosis (left) and local recurrence after surgery (right) with cervical lymphadenopathy (arrowhead). Red arrows highlight location of the tumors.
Cancers 17 01441 g003
Figure 4. Treatment of TFCP2 fusion sarcomas. Swimmer’s plot showing timeline of indicated treatments for 9 out of 10 patients for which treatment data were available. Time 0 is the date of diagnosis. VAC = vincristine, actinomycin, and cyclophosphamide. VAI = vincristine, doxorubicin, and ifosfamide. VDC = vincristine, doxorubicin, and cyclophosphamide. VI = vincristine and ifosfamide. VIT = vincristine, ifosfamide, and temozolomide.
Figure 4. Treatment of TFCP2 fusion sarcomas. Swimmer’s plot showing timeline of indicated treatments for 9 out of 10 patients for which treatment data were available. Time 0 is the date of diagnosis. VAC = vincristine, actinomycin, and cyclophosphamide. VAI = vincristine, doxorubicin, and ifosfamide. VDC = vincristine, doxorubicin, and cyclophosphamide. VI = vincristine and ifosfamide. VIT = vincristine, ifosfamide, and temozolomide.
Cancers 17 01441 g004
Figure 5. Survival outcomes in TFCP2 fusion sarcomas. (A) Overall survival of the patients with available treatment data (n = 9). (B) Recurrence-free survival was calculated in patients with initially localized disease from the date of histologic diagnosis to the date of recurrence, death, or the latest follow-up (n = 6). (C) Progression-free survival of the patients with available treatment data was calculated from the start of systemic therapy for recurrent or metastatic disease to the date of progression, death, or the latest follow-up (n = 9).
Figure 5. Survival outcomes in TFCP2 fusion sarcomas. (A) Overall survival of the patients with available treatment data (n = 9). (B) Recurrence-free survival was calculated in patients with initially localized disease from the date of histologic diagnosis to the date of recurrence, death, or the latest follow-up (n = 6). (C) Progression-free survival of the patients with available treatment data was calculated from the start of systemic therapy for recurrent or metastatic disease to the date of progression, death, or the latest follow-up (n = 9).
Cancers 17 01441 g005
Figure 6. Summary of 53 cases of TFCP2 fusion sarcomas in the literature. (A) Percent of patients with each of the two TFCP2 fusions, either FUS-TFCP2 or EWSR1-TFCP2. (B) Breakdown of the site of disease. (C) Distribution of the age at diagnosis. (D) Distribution of the size of the sarcoma at diagnosis. In (C,D), bars represent means ± standard deviations, and each black dot represents a different patient. (E) Overall survival of the cohort (n = 43). (F) Recurrence-free survival was calculated in patients with initially localized disease from the date of histologic diagnosis to the date of recurrence, death, or the latest follow-up (n = 17). (G) Progression-free survival of the cohort was calculated from the start of systemic therapy for recurrent or metastatic disease to the date of progression, death, or the latest follow-up (n = 25).
Figure 6. Summary of 53 cases of TFCP2 fusion sarcomas in the literature. (A) Percent of patients with each of the two TFCP2 fusions, either FUS-TFCP2 or EWSR1-TFCP2. (B) Breakdown of the site of disease. (C) Distribution of the age at diagnosis. (D) Distribution of the size of the sarcoma at diagnosis. In (C,D), bars represent means ± standard deviations, and each black dot represents a different patient. (E) Overall survival of the cohort (n = 43). (F) Recurrence-free survival was calculated in patients with initially localized disease from the date of histologic diagnosis to the date of recurrence, death, or the latest follow-up (n = 17). (G) Progression-free survival of the cohort was calculated from the start of systemic therapy for recurrent or metastatic disease to the date of progression, death, or the latest follow-up (n = 25).
Cancers 17 01441 g006
Table 1. Clinical characteristics of patients with TFCP2 fusion sarcomas.
Table 1. Clinical characteristics of patients with TFCP2 fusion sarcomas.
IDAge of Sarcoma DiagnosisSexFusionPathologySiteDesminMyoD1ALKMyogeninCytokeratinSize of Primary Tumor (cm)StageSite(s) of MetastasesMitosesPrimary SurgeryChemo 1st LineBest ResponseChemo 2nd LineBest ResponseRadiation?Radiation SummaryHow Was Fusion Discovered?Other MutationsPersonal History of CancerFamily History of Cancer
121FemaleFUS-TFCP2Spindle cell rhabdomyosarcomaLeft frontal bonePositivePositivePositivePositivePositive 7T3 N0 M0Brain parenchyma underlying primary site, lungs>20/10 hpfYesVDCSDIrinotecan + temozolomideMixedYes64.2 Gy in 30 fxMayo Clinic fusion panelNANone None
235MaleEWSR1-TFCP2High-grade rhabdoid malignant tumorRight occipital skull base (cerebellopontine angle)PositiveRarePositive RarePositive 2.8T2 Nx M1Lymph nodes, and lungsKi-67 > 80%NoGem/docetaxMixedLorlatinib, alecitinib (switched from lorlatinib after 5 days due to insurance issue)SDNoN/ANGS panelCDKN2A and CDKN2B loss, ALK inversionNone None
348FemaleFUS-TFCP2Poorly differentiated epithelioid and spindle cell rhabdomyosarcoma Right acetabulumPositivePositivePositiveNegativePositive 11.6IV (T3 Nx M1)Lungs, liver, lymph nodes, and bonesUnknownNoGem/docetaxPDVDCMixedNoN/AMayo Clinic fusion panelNANone Paternal grandmother and great-grandmother with stomach cancer
430FemaleFUS-TFCP2Epithelioid and spindle cell rhabdomyosarcomaHard palatePositivePositivePositive Positive Positive 3.5T2 N0 M0Locally10/10 hpfYesVACPDSingle-agent actinomycin-DPDYes60.4 Gy in 28 fxMayo Clinic fusion panelNANone Maternal grandma with liver;
maternal uncle with lung
513MaleFUS-TFCP2Small-cell sarcoma with rhabdomyogenic/rhabdomyosarcoma differentiationRight maxilla PositivePositivePositive Very rareVery rare 9.5Group III, Stage 3 (pediatric)Lymph nodes, and lungs4/10 hpfYesVACMixedVDC/IEPRYes50.4 Gy in 28 fxMayo Clinic fusion panelNANoneUnspecified cancer in paternal grandfather
611FemaleFUS-TFCP2High-grade rhabdomyosarcoma with spindle and epithelioid cellsC1PositivePositivePositivePositivePositive 7.1IV (T2b, N0, M1)Local extension to vertebral arteries, spinal cord on presentation10/10 hpfNoVincristine, irinotecan PDVI (with concurrent radiation) PRYes50.4 Gy in 28 fx concurrently with VI NGS panelNANoneNone
735MaleFUS-TFCP2Spindle cell sarcoma with rhabdomyosarcomatous differentiationC7 paraspinal musculaturePositivePositivePositivePositivePositive 4.5cT1, cN0, cM0Pulmonary mets present on presentation, enlarged on subsequent scans 8/10 hpf NoVDCPDCrizotinib PDYes50 Gy in 25 fx NGS panelNANoneMaternal grandma with breast cancer
833FemaleEWSR1-TFCP2Spindle cell sclerosing rhabdomyosarcomaRight maxilla PositivePositivePositivePositivePositive 1.7cT4, cN1, cM0 Local nodal spread and small pulmonary nodules on presentation 8/10 hpf YesVDCPDLorlatinib + ifosfamidePDYes36 Gy in 12 fxNGS panelNANoneMaternal grandfather with unspecified cancer
940MaleEWSR1-TFCP2Epithelioid and spindle cell rhabdomyosarcomaLeft trapezius massPositivePositivePositivePositivePositive 6.5Stage IV (cT2, cN1, cM0) C5 LAD10/10 hpfYesVACSDVAC (resumed post-surgery)PDYes50 Gy in 25 fx NGS panelNANoneFather with skin cancer;
paternal aunt with lung cancer
1034MaleFUS-TFCP2High-grade rhabdomyosarcoma with epithelioid and spindle cell featuresMandible and floor of mouth PositivePositiveNot testedNot testedNot tested 6.5T3 N0 M0Locally10/10 hpfYesUnknownNAUnknownNAUnknownNANGS panelNANoneNone
NA: not applicable or available.
Table 2. Review of the literature of TFCP2 fusion sarcoma cases.
Table 2. Review of the literature of TFCP2 fusion sarcoma cases.
Study AuthorPMIDFusionSarcoma Subtype, PathologyCo-Occurring Molecular AlterationsDesminMyoD1ALKMyogeninCytokeratinLocationSize (cm)Mitotic rateAge at
Sarcoma Diagnosis
SexPersonal History of Other CancersTreatment SummarySurgery?Chemo?Radiation?Overall OutcomeTime to Local RecurrenceTime to ProgressionTime to Distant RecurrenceFollow Up
GinnNAFUS-TFCP2Spindle cell rhabdomyosarcomaNAPositivePositivePositivePositivePositiveLeft frontal bone7>20/10 hpf21FemaleNoneSurgery, local recurrence; radiation, distant recurrence; VDC, progression; irinotecan/temozolomide, progression, deathYesVDC, irinotecan/temozolomide64.2 Gy in 30 fxDOD1.6 mo1.6 mo4.6 mo16.7 mo
GinnNAEWSR1-TFCP2High-grade rhabdoid malignant tumorCDKN2A and CDKN2B loss, ALK inversionPositiveRarePositiveRarePositiveRight occipital skull base (cerebellopontine angle)2.8Ki-67 > 80%35MaleNoneGem/docetaxel, mixed response; lorlatinib/alectinib, progression; VAI, progression; VIT, progression, deathNoGem/docetax, lorlatinib/alectinib, VAIN/ADODNA1.1 mo1.910.0 mo
GinnNAFUS-TFCP2Poorly differentiated epithelioid and spindle cell rhabdomyosarcomaNAPositivePositivePositiveNegativePositiveRight acetabulum11.6Unknown48FemaleNoneGem/docetaxel, progression; VDC, mixed response, deathNoGem/docetax, VDCN/ADODNA1.0 mo05.9 mo
GinnNAFUS-TFCP2Epithelioid and spindle cell rhabdomyosarcomaNAPositivePositivePositivePositivePositiveHard palate3.510/10 hpf30FemaleNoneSurgery, local recurrence; VAC, acinomycin, progression; radiation, progression, deathYesVAC, actinomycin D60.4 Gy in 28 fxDOD0.7 mo2.7 moNA24.7 mo
GinnNAFUS-TFCP2Small-cell sarcoma with rhabdomyogenic/rhabdomyosarcoma differentiationNAPositivePositivePositiveVery rareVery rareRight Maxilla9.54/10 hpf13MaleNoneVAC, progression; VDC/IE, PR, radiation, surgery, local recurrence; lorlatinib, progression; entrectenib, progression, lorlatinibYesVAC, VDC/IE50.4 Gy in 28 fxAWD6.9NA6.927.8 mo
GinnNAFUS-TFCP2High-grade rhabdomyosarcoma with epithelioid and spindle cell featuresNAPositivePositiveNot testedNot testedNot testedMandible and floor of mouth6.510/10 hpf34MaleNoneNAYesNANANA6.9 moNA6.9 mo6.9 mo
GinnNAFUS-TFCP2HIGH-GRADE RHABDOMYOSARCOMA WITH SPINDLE AND EPITHELIOID CELLSNAPositivePositivePositivePositivePositiveC17.110/10 hpf11FemaleNoneVincristine/irinotecan, progression after 2 cycles; radiation, PR, lorlatinib, progressed after 1 month, deathNoVincristine, irinotecan50.4 Gy in 28 fx concurrently with VIDODNA1.7 moNA5.8 mo
GinnNAFUS-TFCP2Spindle cell sarcoma with rhabdomyosarcomatous differentiationNAPositivePositivePositivePositivePositiveC7 paraspinal musculature4.58/10 hpf35MaleNoneNeoadjuvant VDC, progression after 2 cycles; radiation, crizotinib, resection, continued crizotinib, local and distant recurrence 3 months after resectionNoVDC50 Gy in 25 fxAWDNA1.2 moNA7.7 mo
GinnNAEWSR1-TFCP2Spindle cell sclerosing rhabdomyosarcomaNAPositivePositivePositivePositivePositiveRight maxilla1.78/10 hpf33FemaleNoneVAC, local and distant progression after 4 cycles; ifosfamide, progression after 2 cycles; ipilimumab/nivolumab, progressed after 2 cycles, deathYesVDC36 Gy in 12 fxDOD3.0 mo2.0 moNA8.6 mo
GinnNAEWSR1-TFCP2Epithelioid and spindle cell rhabdomyosarcomaNAPositivePositivePositivePositivePositiveLeft trapezius mass6.510/10 hpf40MaleNoneVAC, resection, local progression; VAC, progression; VDC, progression; clinical trial, progressionYesVAC50 Gy in 25 fxAWD7.1 mo6.8 moNA22.3 mo
Watson29431183EWSR1-TFCP2Desmoplastic small-round-cell tumor (prior to sequencing)MCFD2-PREPL; POLA2-TCIRG1; TFCP2-BRDT; CAND2-PPARG; ZNF584-TBC1D30 Chest wall 38.3F Avg survival 5.5 months
Watson29431183FUS-TFCP2Rhabdomyosarcoma Pelvic bone 26.1F
Watson29431183FUS-TFCP2Osteosarcoma Sphenoid bone 16.1F
Chrisinger32556562EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma Pos PosNegNegRight frontal bone5 × 4.6 × 4 cm>20/10 hpfmid 20 s–30 sF VDC x 3 cycles: no change; concurrent radiation with another 2 cycles, with decrease in size,
went for resection; resulted in wound dehiscence and infection, multiple OR visits with 2 skin flaps, unable to continue chemo for 4 months; at 8-month scan had mets to R acetabulum and L iliac; continued to have growth of mets, expired from disease 17 months after diagnosis
Removal of mass following chemo-XRTVDC × 3 cycles50.4 Gy in 28 fxDOD8 mo 8 mo17 mo
Chrisinger32556562FUS-TFCP2Rhabdomyosarcoma (epithelioid, spindle, and rhabdoid) PosPosPosPos Pelvic bone9.5 cm25/10 hpf20sF Multi-agent chemotherapy × 2 months with overall tumor burden reduction; repeat scans in 3 months with mixed response, concern for progression in main tumor
radiation of primary site with VI; continued on high-risk RMS protocol for 3 months; stable scans
3 months later, progression with new mets; started on temsirolimus, vinorelbine, and cyclophosphamide; passed 6 weeks later,
11 months after diagnosis
NoneHigh-risk RMS protocol (Ifosfamide, vincristine, etoposide, doxorubicin, cyclophosphamide, and dactinomycin)
Temsirolimus, vinorelbine, cyclophosphamide
50.4 Gy in 28 fxDOD 3 mo 11 mo
Dashti29758589FUS-TFCP2Spindle cell RMS PosPosPosPos Mandible2.8 cm 72M Resection of primary tumor, declined adjuvant chemoResectionDeclined ANED 2 mo
Lewin31470995FUS-TFCP2RhabdomyosarcomaPBRM1 loss (associated with PD-1 and CTLA-4/PD-1 checkpoint inhibitors; did not target due to poor ECOG status) PosPosPos Nasal cavity 23M Rapid progression through 4 cycles of anthracycline-based chemo + external beam radiation to primary sinonasal tumor; transitioned to crizotinib 250 mg BID, stable disease after 4 weeks, died 3 weeks later, 2/2 symptomatic pleural effusionNoneAnthrocycline-based chemo for 4 cycles
crizotinib 250 mg BID for ~7 weeks
Radiation to primary site, unknown amount or fxDOD 4 mo 6 mo
Agaram30720533EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosSkull >15/1027F Unknown Unknown Unknown
Agaram30720533EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosMaxilla, masticator space, sinuses, orbit, and clivus; mets to femur >15/1033F Unknown Unknown Unknown
Agaram30720533EWSR1-TFCP2Spindle cell RMS PosPosPosPosPosFemur >15/1020M Unknown Unknown Unknown
Agaram30720533FUS-TFCP2Spindle cell RMS PosPosPosPosPosIliac <15/1037F Unknown Unknown Unknown
Tagami30948206FUS-TFCP2Spindle cell RMS PosPosPosPosPosLumbar vertebra12/10 hpf70F Surgery --> docetaxel + XRT (30 Gr/10fr + 21 Gr/7fr) --> adriamycin AWD 6 mo
Le Loarer31383960FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosSphenoid bone91216F 4 cycles etop, ifos --> resection --> local relapse 2 mo later with distant mets --> 1 cycle cyclophos and doxo + XRT --> progression --> death DOD2 mo4 mo2 mo15 mo
Le Loarer31383960FUS-TFCP2Epithelioid cell RMS PosPosPosPosPosSacrum101826F 2 cycles IVADO --> progression --> 1 cycle IVE --> progression --> 1 cycle VAI DOD 2 mo 4 mo
Le Loarer31383960EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosPeritoneum 1238F 1 cycle carboplatin, paclitaxel --> progression --> 1 cycle adriamycin, holoxan --> lymphangitic carcinomatosis, death DOD 1 mo 2 mo
Le Loarer31383960EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosNegNegPosHard palate and upper lip32132M 3 cycles VAI --> local progression --> 1 cycle etoposide–carboplatin --> local progression --> 1 cycle actinomycin–cyclophosh --> local progression DOD 3 mo 8 mo
Le Loarer31383960FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosNegPosPosOrbito-temporal-sphenoid15 x 14.53220M 1 cycle doxo-ifos --> local progression --> 3 cycles VAC --> local progression --> XRT + pazopanib --> local progression DOD 1 mo 6 mo
Le Loarer31383960EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosInguinal6.5 x 4.01086M Resection --> recurrence 4 mo --> palliative --> progressed in 2 mo DOD4 mo2 mo 6 mo
Le Loarer31383960EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosFemur5.1818F 2 cycles VDC IE --> local progression --> pazopanib --> local progression DOD 2 mo 8 mo
Le Loarer31383960FUS-TFCP2Epithelioid cell RMS PosPosPosNegPosCervico-occipital5.3 × 3.7 × 4.41617F 2 cycles VDC IE + XRT (60 Gy) --> local progression --> crizotinib 500 mg/d for 1 month then switched to alectinib 1200 mg/d --> stable disease x 9 mos AWD 8 mo 15 mo
Le Loarer31383960FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosLeft occipital11.86631M Resection --> relapse at 1 mo --> 3 cycles AI --> local progression and distant mets --> death DOD1 mo3 mo3 mo6 mo
Le Loarer31383960FUS-TFCP2Spindle cell RMS PosPosPosPosposMandible4.5 × 3.22132M Surgery + adjuvant 5 cycles doxo + ifos --> local relapse at 12 mo + distant met --> 3 cycles gemcitabine, docetaxel AWD12 mo 12 mo14 mo
Le Loarer31383960FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosMandible1.6 × 1.5 × 1.31158F Surgery + adjuvant CT + XRT --> NED ANED 21 mo
Le Loarer31383960FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosMandible5.5412F Neoadjuvant chemo --> surgery, RT, adjuvant chemo --> NED ANED 21 mo
Le Loarer31383960EWSR1-TFCP2Epithelioid cell RMS PosPosNegPosPosMaxilla62611F Chemo --> radiotherapy --> progression (unknown time frame) DOD Unknown
Le Loarer31383960EWSR1-TFCP2Epithelioid cell RMS NegPosPosNegPosMandible3.4325M Surgery --> chemo --> NED ANED 20 mo
Zhong37545350FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosMandible4.5 × 3.3 × 2.2 cmKi-67 30%26M Resection --> recurrence after 2 month --> repeat surgery at 4-month mark --> recurred 2 months later --> died 3 months later DOD2 mo 9 mo
Xu33382123FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosPosPosPosMandible 22M Unknown Unknown
Xu33382123FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosNeg NegMandible 34M ANED 10 mo
Koutlas32504289EWSR1-TFCP2Spindle–epithelioid cell rhabdomyosarcoma PosPosNegPosPosMandibular7 × 1.8 × 1 cm>10/1015MFetal hepatoblastoma, mixed epithelial/mesenchymal type at 2 years with lung mets, treated with cisplatin, doxorubicin, 5FU and vincristine --> pulm recurrence 2 years later, resected, irinotecan, vincristine, erlotinib --> remission
negative for Li-Fraumeni
Resection with LN dissection --> alternating courses of vincristine, actinomycin D, and cyclophosphamide with etoposide and ifosfamide --> developed met to T7 --> radiation, switched to irinotecan and temozolamide, patient still undergoing treatment at time of paperResection with LN dissection Unknown Unknown
Panferova35768243EWSR1-TFCP2Spindle cell RMS Neg PosPosPosMandiublar64mm × 18mm × 44mmKi-67 50%16F Resection with negative margins, negative MRI --> originally treated with doxorubicin, cisplatin --> showed local relapse after 1 cycle with mets to LN, C1-C2, iliac wing (tumor progression after 2 months) --> switched to ifosfamide, vincristine, dactinomycin, progressed after 2 cycles (4.5 months after primary diagnosis) --> radiation to C1-C2 + vincristine, irinotecan, temozolamide + crizotinib (250 mg daily) --> palliative 6.5 months after primary diagnosis, death 5 months later 54 Gy total to C1/2DOD1 mo2 mo1 mo11.5 mo
Schopf38168093FUS-TFCP2Pleomorphic spindle and epithelioid cell RMSCDK2NA frameshift del; CCND2 high expression Pos Maxillary bone/palate 38M Neoadjuvant doxorubicin/ifosfamide, surgery, adjuvant RTX --> relapse at 28 mo --> surgery (5 mo), gemcitabine/docetaxel (2 mo, PD), eribulin (3 mo, MR), crizotinib (3 mo, PD), surgery (3 mo), ifosfamide (2 mo, PD) DOD28 mo N/A48 mo
Schopf38168093EWSR1-TFCP2Spindle cell RMSCDK2NA loss Pos Mediastinum 60M Surgery + XRT --> lung mets at 7 mo --> Doxorubicin/olaratumab (2 mo, PD), trabectedin (2 mo, PD), crizotinib (5 mo, MR) DODN/A 7 mo20 mo
Schopf38168093FUS-TFCP2SpindleCDK2NA loss Pos Maxilla 48M Neoadjuvant vincristine/ifosfamide/actinomycin, surgery, adjuvant vincristine/ifosfamide/actinomycin --> lung, LN, bone mets at 9 mo --> Topotecan/carboplatin/cyclophosphamide/etoposide (4 mo, PD), crizotinib (2 mo, n.e.) DODN/A 9 mo16 mo
Schopf38168093FUS-TFCP2Epithelioid cell RMSCDK2NA loss Pos Occipital/nuchal soft tissue 35M Surgery --> relapse 4 mo --> Surgery (9 mo), surgery, adjuvant RTX (12 mo, n.a.), surgery (4 mo, n.a.), surgery (1 mo), doxorubicin/ifosfamide (3 mo, SD), pazopanib (1 mo, PD), RTX (1 mo, PD), ceritinib (1 mo, n.e.) DOD4 mo 13 mo42 mo
Schopf38168093FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcomaCDK2NA frameshift del; CCND2 high expression Pos Occipital/nuchal soft tissue 40F Surgery --> relapse 7 mo --> surgery, adjuvant RTX (16 mo), surgery, adjuvant doxorubicin/ifosfamide (5 mo, n.a.), surgery (6 mo), surgery (2 mo), trabectedin/olaparib (4 mo, SD) DOD7 mo N/A42 mo
Schopf38168093FUS-TFCP2Unspecified RMSCCND2 gain and high CCND2 expression Pos Iliac bone 17F Neoadjuvant vincristine/ifosfamide/doxorubicin/actinomycin/etoposide, neoadjuvant RTX, surgery, maintenance chemotherapy with trofosfamide/idarubicin/etoposide (6 mo) --> relapse with distant mets 14 mo --> Topotecan/cyclophosphamide, Irinotecan/temozolomide (7 mo, SD), trofosfamide/idarubicin/etoposide/pazopanib (2 mo, PD), alectinib (3 mo, PD), RTX (palliative, n.e.), lorlatinib (2 mo, n.e.) DOD14 mo 14 mo34 mo
Schopf38168093FUS-TFCP2Unspecified RMS Pos Maxillary bone 14F Vincristine/doxorubicin/cyclophosphamide/ifosfamide/etoposide (4 mo, SD) --> progression at 6 mo --> Vincristine/ifosfamide/actinomycin (1.5 mo, PD), carboplatin/etoposide, maintenance chemotherapy with trofosfamide/idarubicin/etoposide, cyclophosphamide/vinblastine (3 mo, SD), RTX (palliative, n.e.) DOD 6 moN/A14 mo
Schopf38168093EWSR1-TFCP2Unspecified RMSCDK2NA loss Pos Mandible 9F Neoadjuvant ifosfamide/vincristine/actinomycin --> progression 2 mo --> surgery, maintenance therapy with crizotinib (9 mo, n.a.), vincristine/irinotecan/temozolomide/olaparib (7 mo, SD), RTX (palliative, n.a.) DODN/A2 mo 25 mo
Schopf38168093FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcomaCDK2NA loss; CCND2 high expression Pos Temporal/sphenoid bone 15F Neoadjuvant ifosfamide/vincristine/actinomycin/doxorubicin, neoadjuvant RTX --> local progression 5 mo --> surgery, temozolomide/irinotecan (2 mo, PD), surgery, gemcitabine/docetaxel (1 mo, PD) DOD 5 mo 9 mo
Schopf38168093FUS-TFCP2Spindle–epithelioid cell rhabdomyosarcomaCDKN2A loss; CCND2 gain and high CCND2 expression Pos Mandibular head/temporal bone 49F Neoadjuvant docetaxel/cisplatin/5-fluorouracil, surgery, adjuvant RTX --> distant met 12 months --> surgery (2 mo), pembrolizumab (2 mo, PD), doxorubicin/ifosfamide and hyperthermia (8 mo, PR), surgery (4 mo), paclitaxel/gemcitabine (3 mo, n.e.) DOD 12 mo36 mo
Schopf38168093EWSR1-TFCP2Epithelioid cell RMSCDK2NA loss Pos Shoulder soft tissue 25F Surgery, adjuvant RTX --> distant met 6 mo --> surgery (6 mo), surgery (2 mo), doxorubicin/ifosfamide (3 mo, SD), RTX (palliative, n.a.) LTFU 6 mo19 mo
Schopf38168093EWSR1-TFCP2Spindle cell RMSCDK2NA loss Pos Ethmoid cells/frontal sinus 58M Neoadjuvant doxorubicin/ifosfamide --> local progression 2 months --> “Ifosfamide/vincristine/doxorubicin/actinomycin,
RTX, maintenance therapy with cyclophosphamide/vinorelbine (17 mo, PR),
RTX (definitive, 5 mo, PD), topotecan/cyclophosphamide (2 mo, PD), crizotinib (1 mo, n.e.)”
DOD 2 mo 33 mo
NA: not applicable.
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

Ginn, M.P.; Denu, R.A.; Ingram, D.R.; Wani, K.M.; Lazar, A.J.; Harrison, D.J.; Nakazawa, M.S.; Conley, A.P.; Patel, S.; Livingston, J.A. TFCP2 Fusion-Positive Rhabdomyosarcomas: A Report of 10 Cases and a Review of the Literature. Cancers 2025, 17, 1441. https://doi.org/10.3390/cancers17091441

AMA Style

Ginn MP, Denu RA, Ingram DR, Wani KM, Lazar AJ, Harrison DJ, Nakazawa MS, Conley AP, Patel S, Livingston JA. TFCP2 Fusion-Positive Rhabdomyosarcomas: A Report of 10 Cases and a Review of the Literature. Cancers. 2025; 17(9):1441. https://doi.org/10.3390/cancers17091441

Chicago/Turabian Style

Ginn, Madison P., Ryan A. Denu, Davis R. Ingram, Khalida M. Wani, Alexander J. Lazar, Douglas J. Harrison, Michael S. Nakazawa, Anthony P. Conley, Shreyaskumar Patel, and John Andrew Livingston. 2025. "TFCP2 Fusion-Positive Rhabdomyosarcomas: A Report of 10 Cases and a Review of the Literature" Cancers 17, no. 9: 1441. https://doi.org/10.3390/cancers17091441

APA Style

Ginn, M. P., Denu, R. A., Ingram, D. R., Wani, K. M., Lazar, A. J., Harrison, D. J., Nakazawa, M. S., Conley, A. P., Patel, S., & Livingston, J. A. (2025). TFCP2 Fusion-Positive Rhabdomyosarcomas: A Report of 10 Cases and a Review of the Literature. Cancers, 17(9), 1441. https://doi.org/10.3390/cancers17091441

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