Open Access

Clinicopathological and molecular characteristics of abdominal desmoid tumors in the Chinese population: A single‑center report of 15 cases

  • Authors:
    • Jiongyuan Wang
    • Ning Jia
    • Qiaowei Lin
    • Yuan Huang
    • Jinglei Li
    • Quan Jiang
    • Wenshuai Liu
    • Jing Xu
    • Yingyong Hou
    • Ju Liu
    • Ming Li
    • Weiqi Lu
    • Yuhong Zhou
    • Yong Zhang
    • Hanxing Tong
  • View Affiliations

  • Published online on: November 1, 2019     https://doi.org/10.3892/ol.2019.11038
  • Pages: 6443-6450
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Desmoid tumors (DTs), derived from the abdomen, are a type of rare and aggressive borderline tumor exhibiting high recurrence and malignant potential. The aim of the present study was to investigate the clinicopathological and molecular characteristics of abdominal DT in a Chinese population and to provide clues for selecting the optimal treatment strategy for different types of abdominal DT. The clinicopathological data of 15 consecutive patients with DT was collected. Matched fresh‑frozen tumor tissues and peripheral blood samples were used to detect mutations of adenomatous polyposis coli gene (APC), β‑catenin (CTNNB1) and MutY DNA glycosylase (MUTYH) using Sanger sequencing. Pearson's test was conducted to analyze the differences between sporadic DT and familial adenomatous polyposis (FAP) associated with DT. Time to progress (TTP) and overall survival curves were estimated using the Kaplan‑Meier method and compared using the log‑rank test. A review of the patient clinicopathological characteristics revealed that FAP‑associated DT exhibited a higher rate of abdominal surgery history (P=0.011), with no significant differences in any other characteristics. Sequencing revealed that mutations in the APC, CTNNB1 and MUTYH genes were common in DT, and only one patient harbored no mutations in these genes. Survival analyses revealed that patients with FAP exhibited shorter TTP (P=0.030). Log‑rank test demonstrated a tendency towards shorter TTP in the cases where an R2 resection was performed (P=0.072) and a tendency towards poor prognosis in the cases of DT associated with FAP (P=0.087). In conclusion, Abdominal DTs were prone to occur in patients with FAP with a history of abdominal surgery. Mutations in the APC, CTNNB1 and MUTYH genes were detected in patients with DTs. To the best of our knowledge, this is the first study of abdominal DT occurrence in patients with MUTYH‑associated FAP. The prognosis of DT associated with FAP may be worse compared with that of sporadic DT.

Introduction

Desmoid tumors (DTs), also known as aggressive fibromatosis, are rare, locally infiltrative mesenchymal neoplasms that are associated with high rates of local recurrence, but do not possess the potential to metastasize (1). DTs account for <3% of all soft tissue tumors (2) and have an estimated incidence rate of 2–5 individuals per million per year in Europe (3). The exact etiology of DTs is currently unknown, but hormonal, genetic and physical factors all serve roles in their development and growth. Two different types of DTs have been described: Sporadic tumors and familial adenomatous polyposis (FAP)-associated DTs (4). Regardless of type, an important part of the biology is the disruption of the Wnt signaling pathway, leading to the accumulation of nuclear β-catenin and the inappropriate stimulation of downstream genes (4). In sporadic DT, this is often caused by a mutation in the β-catenin gene (CTNNB1) (5), whereas in FAP-associated DT, the loss of adenomatous polyposis coli (APC) function serves a role (6). Recently described biallelic mutations of MutY DNA glycosylase (MUTYH) have been demonstrated to be responsible for adenomatous polyposis with an increased risk of colorectal cancer and have been observed in 30–40% of adenomatous polyposis cases in which an APC mutation was not identified; however, no evidence has indicated thus far that MUTYH serves a role in DT (7). Further studies have revealed that the mutation site of APC, as well as that of CTNNB1, may impact the incidence, severity and prognosis of DT (810). This may be significant for the prevention and treatment of DTs.

DT can occur in any location, often involving the extremities, the trunk, including the pelvic and shoulder girdles, and the abdomen (4). For abdominal DTs, the clinical presentation depends on its primary site. In the present study, according to its location, abdominal DTs are classified into two types: Peripheral and central DT. The clinicopathological and molecular characteristics were collected from a group of patients with abdominal DTs and analyzed to explore the optimal strategies for diagnosing and management of different types of abdominal DTs.

Materials and methods

Study design, patients and samples

Tumor samples, peripheral blood samples and matched clinical data from 15 patients with DT were retrospectively collected from the Zhongshan Hospital (Shanghai, China) with informed consent. The present study was approved by the Committee for Ethical Review of Research Involving Human Subjects of Zhongshan Hospital Affiliated with Fudan University. All 15 patients received surgery and were diagnosed with DT consecutively between July, 2009 and January, 2016; no patients were excluded from the study. DTs were diagnosed by an experienced pathologist based on tumor morphology and immunohistochemistry. The expression levels of APC, CTNNB1 and MUTYH genes of 15 patients were analyzed by direct sequencing of tumor and blood samples. The clinicopathological data and follow-up outcomes were collected and analyzed.

Data retrieved included age, sex, date of surgery, anatomic site, tumor size and margin status. The results of the APC, CTNNB1 and MUTYH mutational analysis were recorded along with the type of treatments administered prior to and/or following surgery. Patients were followed to record any future recurrence of disease with the date of recurrence, treatment following recurrence, date of the last follow-up and status at last follow-up. According to the anatomic site, abdominal DTs were classified as either peripheral (derived from the abdominal wall) or central (derived from the retroperitoneal space and mesentery). Tumor size was defined as the greatest DT dimension in the surgical specimen reported by the original pathologists. Surgical excisions were considered macroscopically complete in the absence of gross residual disease. All macroscopically complete resections were classified according to the closest surgical margin, which was microscopically categorized as positive (R1, tumor within 1 mm from the inked surface) or negative (R0, the absence of tumor within 1 mm from the inked surface). Non-surgical treatments were administered in the primary or recurrent phase of the disease on an individualized basis. These included radiotherapy, chemotherapy (methotrexate and vinorelbine in the majority of cases), medical therapy/hormone agents (tamoxifen, toremifene) and non-steroidal anti-inflammatory drugs (cyclooxygenase-2 inhibitors).

Telephone and outpatient follow-up were used to collect the data. The median follow-up time following the diagnosis of DT was 60 months (range, 20–90 months), and all follow-up data were complete.

Mutational analysis

Genomic DNA was extracted from peripheral blood using a TIANamp Blood DNA kit (Tiangen Biotech Co., Ltd.) and tumor samples using a TIANamp Genomic DNA kit (Tiangen Biotech Co., Ltd.). Primers flanking all coding exons and intron-exon boundaries of APC, CTNNB1 and MUTYH were designed using Primer Premier (version 5.0; Premier Biosoft) and are presented in Table SI. Genomic DNA samples were amplified using PCR. The thermocycling conditions were as follows: Denaturation at 94°C for 5 min; 31 cycles of denaturation at 94°C for 30 sec, annealing for 30 sec (temperature was set according to the primers of each fragment) and an extension at 72°C for 1 min. A final extension step was performed at 4°C for 5 min, and the experiment was repeated 10–20 times. The PCR products were evaluated by a 2% agarose gel electrophoresis and were further purified using an AxyPrep DNA Gel Extraction kit (Corning Life Sciences) according to the manufacturer's protocol. Sanger sequencing was subsequently performed using an ABI PRISM 3730 automated sequencer (Applied Biosystems; Thermo Fisher Scientific, Inc.). Sequencing results were analyzed by Geneious software (version 5.6.7; Biomatters Ltd.). An identified mutation was verified in the corresponding region of the unaffected parents of the proband and 100 population-matched healthy controls. The mutation was described by comparison with the NCBI cDNA reference sequences (11) NM_001354897.1 for APC, NM_001098209.1 for CTNNB1 and NM_012222.2 for MUTYH.

Statistical analysis

All statistical analyses were performed using SPSS software (version 18.0; SPSS, Inc.). The primary endpoint was the time to progress (TTP) and overall survival (OS), and the event time was calculated as the time between the date of surgery and the date of relapse or death, whichever occurred first, or was censored at the date of last follow-up assessment in event-free patients. TTP curves were estimated using the Kaplan-Meier method and were compared statistically using the log-rank test. Multivariate Cox model analyses were also performed using following characteristics: Sex, tumor site, tumor size and margin status. A Fisher's exact test was performed to analyze the differences between sporadic DT and FAP-associated DT. P<0.05 was considered to indicate a statistically significant difference.

Results

Patient demographics and disease characteristics

Between July, 2009 and January, 2016, 15 patients, including nine female and six male patients, were enrolled in the present study. The demographic and disease characteristics are presented in Table I and Fig. 1. In total, nine patients were ≥30 years, and the median age was 32 years (range, 26–47 years). According to the primary site of DT, the central type accounted for 80% (12/15), whereas the peripheral type accounted for 20% (3/15) of the cases. The median size of the primary lesions was 10 cm (range, 4–17 cm). Of the 15 cases, nine were sporadic (four male and five female patients), whereas six patients had a family history of FAP and a history of abdominal injury, including surgery.

Table I.

Clinical characteristics of 15 patients with DT.

Table I.

Clinical characteristics of 15 patients with DT.

CharacteristicsCases, n (%)
Type of DT
  FAP-associated6 (42.86)
  Sporadic9 (57.14)
Age, years
  <306 (42.86)
  ≥309 (57.14)
Sex
  Male6 (42.86)
  Female9 (57.14)
History of abdominal surgery
  Yes6 (42.86)
  No9 (57.14)
Site
  Central12 (80)
  Peripheral  3 (20)
Tumor size
  <105 (33.33)
  ≥10 cm10 (66.67)
Margin status for primary surgery
  R0/R19 (60)
  R26 (40)
Recurrence or progression after primary surgery
  No3 (20)
  Yes12 (80)
Treatment after primary surgery
  Radiotherapy2 (13.33)
  Chemotherapy6 (40)
  Other (COX-2I, TAM or wait and see)7 (46.67)
Type of treatment for recurrence
  Surgery6
  Radiotherapy3
  Chemotherapy6
  Other (COX-2I, TAM or none)8

[i] DT, desmoid tumor; FAP, familial adenomatous polyposis syndrome; COX-2I, cyclooxygenase-2 inhibitor; TAM, tamoxifen.

All the patients accepted surgical resection as a treatment course, with a number of patients undergoing multiple surgeries due to recurrence. Overall, nine patients underwent complete surgical excision (R0/R1), and six patients underwent R2 resection. Due to variations in the anatomic site of the tumor, the extent of surgery was different for each case. The three patients with peripheral-type sporadic DTs underwent R0/R1 resection without combined organ resection. Among the 12 patients with central type DTs, 50% (6/12) underwent R0/R1 resection, whereas 50% (6/12) underwent R2 resection. For the central type group, combined evisceration was performed in 75% (9/12) of patients, and the resected organs included the small intestine (66.67%, 6/9), colon (33.33%, 3/9), ureter (22.22%, 2/9), appendix (11.11%, 1/9), ovary (11.11%, 1/9) and pancreas (11.11%, 1/9). Following primary surgery, only three patients did not experience recurrence or progression. In addition, six patients who underwent R2 resection received chemotherapy, two patients received radiotherapy and seven patients received Tamoxifen, a non-steroid anti-inflammatory drug (NSAID; COX-2 inhibitor) and/or waited for the outcome. Of patients who experienced recurrence and progression following primary resection, six cases underwent additional surgery, three received radiotherapy and six received chemotherapy; eight patients received Tamoxifen, NSAID (COX2 inhibitor) or waited for the outcome.

Difference between sporadic and FAP-associated DT

To reveal the differences between sporadic and FAP-associated DT, clinicopathological characteristics between the two groups were compared. The results revealed a significant difference between the two groups in past history of abdominal surgery, with patients with FAP-associated DT exhibiting a higher rate of abdominal surgery. This may suggest an increased susceptibility to the development of DT in patients with FAP that had a history of abdominal surgery. No significant differences were identified in the other characteristics (Table II).

Table II.

Differences between sporadic and FAP-associated desmoid tumors.

Table II.

Differences between sporadic and FAP-associated desmoid tumors.

CharacteristicsFAP-associated, nSporadic, nFisher's exact test P-value
Sex
  Male24>0.999
  Female45
Age, years
  20–30330.622
  ≥3036
History of abdominal surgery
  Yes520.011a
  No17
Site
  Central660.229
  Peripheral03
Tumor size, cm
  <10140.580
  ≥1055
Margin status
  R0/R1470.136
  R222
Recurrence after R0/R1 resection
  Yes440.236
  No03

a P<0.05. FAP, familial adenomatous polyposis syndrome.

Mutations in the study cohort

Among the 15 patients with DT, only one patient did not exhibit any mutations in CTNNB1, APC or MUTYH. Of the six patients with FAP-associated DT, one patient harbored a MUTYH gene mutation, whereas the others possessed APC gene mutations. In the nine patients with sporadic DT, analysis of the tumor samples revealed six mutations in the CTNNB1 gene, four mutations in the APC gene and one mutation in the MUTYH gene. In the group of patients with sporadic DT, one harbored mutations in CTNNB1, APC and MUTYH genes, whereas another carried CTNNB1 and APC mutations (Table III).

Table III.

Mutations identified in APC, CTNNB1 and MUTYH in the present study.

Table III.

Mutations identified in APC, CTNNB1 and MUTYH in the present study.

No.IncidenceGeneLocationMutationMutation type
1FamilialAPCE15 c.1875_1878delGACAFrameshift
2FamilialAPCE09 c.904C>T:p.R302XMissense
3FamilialMUTYHE15 c.1005G>C:p.Q335HMissense
4FamilialAPCE16-9 c.3927_3931delAAAGAFrameshift
5FamilialAPCE16 c.3927-3931delAAAGAFrameshift
6FamilialAPCE16-10 c.4429C>T:p.Q1477XMissense
7SporadicAPCE16 c.5465T>A:p.V1822D, c.1635G>AMissense, synonymous
8SporadicAPC c.1458T>C, c.1635G>ASynonymous
9SporadicCTNNB1E3 c.133T>C:p.S45PMissense
10SporadicAPC c.1458T>C, c.1488A>T, c.1635G>ASynonymous
CTNNB1E15c.2340C>TSynonymous
MUTYHE12c.1005 G>CSynonymous
11Sporadic None
12SporadicAPC c.1458T>C, c.1635G>ASynonymous
CTNNB1E15c.2340 C>TSynonymous
13SporadicCTNNB1E3 c.134C>T:p.S45FMissense
14SporadicCTNNB1E3 c.121:A>G:p.T41AMissense
15SporadicCTNNB1E3 c.121:A>G:p.T41AMissense

[i] APC, adenomatous polyposis coli; CTNNB1, β-catenin; MUTYH, MutY DNA glycosylase.

Survival analysis

In the follow-up to December 2016, two patients with FAP-associated DT succumbed to disease, six patients survived with progression and seven exhibited disease-free survival. The median TTP was 13 months, and median OS time was 57 months. Survival analysis revealed that patients with FAP-associated DT exhibited shorter TTP (P=0.030). Log-rank test revealed a tendency for shorter TTP in the cases that underwent R2 resection (P=0.072) and poor prognosis in the cases with FAP-associated DT (P=0.087) (Table IV; Fig. 2). In the multivariate Cox regression analysis, FAP-associated DT was revealed to be associated with a higher risk of recurrence following resection (Table V).

Table IV.

Univariate analysis of TPP and OS.

Table IV.

Univariate analysis of TPP and OS.

A, Univariate analysis of TTP

Log-rank test

Variablesχ2P-value
Sex (male vs. female)1.4010.236
Age, years (20–30 vs. ≥30)0.2420.623
Tumor size, cm (<10 vs. ≥10)0.7870.375
Site (central vs. peripheral)2.6580.103
Margin status (R0/R1 vs. R2)3.2270.072a
FAP-associated (yes vs. no)4.6840.030a
Systematic therapy (yes vs. no)0.0030.959

B, Univariate analysis of OS

Log-rank test

Variables χ2P-value

Sex (male vs. female)1.6060.205
Age, years (20–30 vs. ≥30)0.0720.788
Tumor size, cm (<10 vs. ≥10)0.1430.705
Site (central vs. peripheral)0.5730.449
Margin status (R0/R1 vs. R2)0.0720.788
FAP-associated (yes vs. no)2.9250.087
Systematic therapy (yes vs. no)1.1800.277

a P<0.05. TTP, time to progress; OS, overall survival; FAP, familial adenomatous polyposis syndrome.

Table V.

Cox regression analysis of risk factors for recurrence.

Table V.

Cox regression analysis of risk factors for recurrence.

Risk factorHR95% CIP-value
Sex1.4040.188–10.4740.740
Age1.0150.151–6.8210.998
Site0.0900.003–2.2970.145
FAP-associated0.0860.006–1.2830.075a
Margin status0.4100.062–2.7030.354
Systematic therapy15.3670.325–726.4520.165

a P<0.05. FAP, familial adenomatous polyposis syndrome; CI, confidence interval. HR, hazard ratio.

Discussion

DTs are heterogenous borderline lesions that originate from mesenchymal fibroblastic proliferation with an incidence rate of 0.03% of all neoplasms and 3.00% of soft tissue tumors (12). It is a rare condition that typically infiltrates surrounding structures and tends to recur with minimal metastatic potential (12). The majority of DTs occur sporadically, but a number of DTs are associated with FAP syndrome (13). In the present study, when clinicopathological characteristics were compared between the sporadic and FAP-associated groups, the history of abdominal surgery and prognosis were notably different. Among the 15 patients, all patients with FAP-associated DTs had a history of abdominal surgery. Clark et al (14) reported that 82% of patients with FAP-associated DTs had predisposing surgery. It has been hypothesized that surgical trauma and the process of tissue repair by fibroblasts contribute to DT development following injury. The risk of tumor development among patients with FAP following injury or surgical trauma remains unknown.

Among patients with FAP, DT is one of the most common causes of death after colorectal cancer (15). Due to the high local recurrence rate, recurrence and survival are of equal importance in the management of DT. In the present study, 2 patients with FAP-associated DTs succumbed to complications associated with the tumor. Survival analysis revealed that patients with FAP-associated DT exhibited shorter TTP and a relatively poor prognosis compared with those with sporadic DTs. These results suggest a more aggressive pathology and poor prognosis in patients with FAP-associated DT, but more evidence is required to support this hypothesis.

Alterations in the Wnt/β-catenin signaling pathway have been reported as the likely driving mechanism of tumorigenesis in the development of the disease; mutations in the β-catenin gene occur in the majority of sporadic cases, as well as mutations in APC, which regulates β-catenin degradation in FAP-associated DTs (16). In the present study, APC, CTNNB1 and MUTYH mutational analyses were performed in 15 patients, and only one patient did not exhibit any mutations in these genes. Of the six patients with FAP-associated DT, five harbored APC mutations, of which 80% were located downstream from codon 1444. Previous studies have reported that FAP-associated DTs are associated with mutations toward the 3′ end of the gene, distal to codon 1444, compared with the general FAP population, where 90% of mutations are located upstream from codon 1444 (17,18). A novel finding from the present study was that one patient with FAP-associated DT exhibited MUTYH mutations within the tumor tissues. Mutations in the MUTYH gene have been previously demonstrated in a type of colorectal polyposis referred to as MUTYH-associated polyposis (MAP), which has a relatively mild phenotype, exhibiting <100 adenomas per patient and an average age at diagnosis of 45 years (19). In certain patients with MAP, the extracolonic manifestations can also be demonstrated, but DTs are not part of the MAP tumor spectrum (20). To date, MUTYH mutations in FAP-associated DTs have not been reported in published literature or in any database. The mutation c.1005G>C has also not yet been reported. Whether it is meaningless or functionally associated with the development of DT will require further investigation. In tumors of the nine patients with sporadic DT, six mutations in the CTNNB1 gene, four mutations in the APC gene and one mutation in the MUTYH gene were identified. Three particular mutations in the CTNNB1 gene have been reported (and associated) with DT: T41A, S45F and S45P (6). A number of studies have demonstrated a significantly increased risk of disease recurrence in patients harboring an S45F mutation compared with those who had either tumors with a T41A mutation of WT tumors (21,22); however, the association between genotype and phenotype has not yet been determined. In the present study, two T41A, one S45F and one S45P mutations were identified in CTNNB1. However, due to the small sample size of the present study, the association between particular gene mutations and prognosis could not be confirmed.

Due to the lack of prospective cohort studies for rare conditions such as DTs, the optimal treatment remains to be elucidated. Surgery is the primary treatment for patients with resectable DTs (23). According to the results of the present study, resection of central-type DTs is more difficult compared with that of peripheral-type DTs due to organ involvement. Although margin status remains a controversial topic in the management of DTs (24,25), the present study demonstrated a tendency towards shorter TTP in the patients that underwent R2 resection, which may be due to the small sample size. DTs behave in an unpredictable manner, as they may remain stable, grow aggressively or even regress without intervention (26,27). Due to the heterogeneity, unpredictable natural biology and high recurrence rate of DTs, the benefit of aggressive treatment modalities remains unclear. A move towards conservative management strategies has emerged over the past decade, including hormonal therapy, NSAIDs, chemotherapy and a ‘wait and see’ approach. In the present study, following primary surgery, seven patients who accepted R0/R1 or R2 resection, received Tamoxifen, NSAID (COX2 inhibitor) and/or waited for the outcome remained alive by the end of the follow-up period. Bonvalot et al (28) revealed that patients with extra-abdominal DT with a microscopically complete surgery had a similar outcome compared with patients in a no-surgery group. However, abdominal DT has different characteristics, particularly for retroperitoneal and intra-abdominal cases, and may lead to severe morbidity, functional impairment and even mortality due to its anatomic site (12). In certain cases, resectable tumors may progress to be unresectable or involve additional viscera (12). Therefore, it is currently unknown when a ‘wait and see’ approach may be the best treatment strategy. More prospective trials are required to provide a basis for clinical management. A number of systemic therapies have been used for DT, ranging from those with low toxicity such as hormonal therapy or NSAIDs (29,30) to combination chemotherapy (31,32). In the present study, hormonal therapy, NSAIDs and chemotherapy were used alone or in combinations to treat patients with primary or recurrent DTs, but the effect was difficult to evaluate. Potential complications should be carefully considered when systemic therapy is used as, In the present study, two patients experienced gastrointestinal perforation due to chemotherapy (33).

For abdominal DTs, which are a rare condition, the management remains controversial due to the high number of variations, such as the incisional margin, site of tumor and genotype. However, surgery remains the preferred treatment for DTs. Although the present study comprised a small sample size, the analysis revealed a number of meaningful results, such as mutations in APC, CTNNB1 and MUTYH. To the best of our knowledge, the present study is the first to report a patient with MUTYH-associated FAP and an abdominal DT; abdominal DT was prone to occur in patients with FAP who had undergone abdominal surgery, and the prognosis of patients with FAP-associated DT was poor. The present study may contribute to the future development of optimal strategies for diagnosing and management of different types of abdominal DTs.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

JYW, NJ, QWL, YH, YZ and HXT conceived and designed the study. JYW and HXT drafted the manuscript. QWL, JLL, QJ and WSL performed the mutation analysis. YFH and JL collected the samples and performed pathological analysis. YH performed the enteroscopy. NJ, JX and ML collected clinical data and assisted in the follow-up. WQL and YHZ assisted in data analysis and interpretation. YZ revised the manuscript critically. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Committee for Ethical Review of Research Involving Human Subjects of Zhongshan Hospital affiliated with Fudan University. Informed consent was acquired from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

DT

desmoid tumor

FAP

familial adenomatous polyposis

TTP

time to progress

OS

overall survival

References

1 

Wu C, Nik-Amini S, Nadesan P, Stanford WL and Alman BA: Aggressive fibromatosis (desmoid tumor) is derived from mesenchymal progenitor cells. Cancer Res. 70:7690–7698. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Desmoid-type fibromatoses, ; Fletcher CDM, Bridge JA, Hogendoorn PCW and Mertens F: WHO Classification of Tumours of Soft Tissue and Bone. 4th. IARC Press; Lyon: pp. 72–73. 2013

3 

Penel N, Coindre JM, Bonvalot S, Italiano A, Neuville A, Le Cesne A, Terrier P, Ray-Coquard I, Ranchere-Vince D, Robin YM, et al: Management of desmoid tumours: A nationwide survey of labelled reference centre networks in France. Eur J Cancer. 58:90–96. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Lips DJ, Barker N, Clevers H and Hennipman A: The role of APC and beta-catenin in the aetiology of aggressive fibromatosis (desmoid tumors). Eur J Surg Oncol. 35:3–10. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Hartley JE, Church JM, Gupta S, mcGannon E and Fazio VW: Significance of incidental desmoids identified during surgery for familial adenomatous polyposis. Dis Colon Rectum. 47:334–340. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Miyoshi Y, Iwao K, Nawa G, Yoshikawa H, Ochi T and Nakamura Y: Frequent mutations in the beta-catenin gene in desmoid tumors from patients without familial adenomatous polyposis. Oncol Res. 10:591–594. 1998.PubMed/NCBI

7 

Sereno M, Merino M, López-Gómez M, Gómez-Raposo C, Zambrana Tébar F, Moreno Rubio J, Espinós J, Martín-Algarra S and Casado Sáenz E: MYH polyposis syndrome: Clinical findings, genetics issues and management. Clin Transl Oncol. 16:675–679. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Couture J, Mitri A, Lagace R, Smits R, Berk T, Bouchard HL, Fodde R, Alman B and Bapat B: A germline mutation at the extreme 3′ end of the APC gene results in a severe desmoids phenotype and is associated with overexpression of beta-catenin in the desmoid tumor. Clin Genet. 57:205–212. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Colombo C, Miceli R, Lazar AJ, Perrone F, Pollock RE, Le Cesne A, Hartgrink HH, Cleton-Jansen AM, Domont J, Bovée JV, et al: CTNNB1 45F mutation is a molecular prognosticator of increased postoperative primary desmoids tumor recurrence: An independent, multicenter validation study. Cancer. 119:3696–3702. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Van Broekhoven DL, Verhoef C, Grünhagen DJ, van Gorp JM, den Bakker MA, Hinrichs JW, de Voijs CM and van Dalen T: Prognostic value of CTNNB1 gene mutation in primary sporadic aggressive fibromatosis. Ann Surg Oncol. 22:1464–1470. 2015. View Article : Google Scholar : PubMed/NCBI

11 

CCDS Database, . https://www.ncbi.nlm.nih.gov/CCDS/CcdsBrowse.cgi

12 

Sakorafas GH, Nissotakis C and Peros G: Abdominal desmoid tumors. Surg Oncol. 16:131–142. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Clark SK and Phillips RK: Desmoids in familial adenomatous polyposis. Br J Surg. 83:1494–1504. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Clark SK, Neale KF, Landgrebe JC and Phillips RK: Desmoid tumours complicating familial adenomatous polyposis. Br J Surg. 86:1185–1189. 1999. View Article : Google Scholar : PubMed/NCBI

15 

de Campos FG, Peres RO, Imperiale AR, Seid VE, Nahas SC and Cecconello I: Evaluating causes of death in familial adenomatous polyposis. J Gastrointest Surg. 14:1943–1949. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Skubitz KM: Biology and treatment of aggressive fibromatosis or desmoid tumor. Mayo Clin Proc. 92:947–964. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Bertario L, Russo A, Sala P, Eboli M, Giarola M, D'amico F, Gismondi V, Varesco L, Pierotti MA and Radice P; Hereditary Colorectal Tumours Registry, : Hereditary Colorectal Tumours Registry: Genotype and phenotype factors as determinants of desmoid tumors in patients with familial adenomatous polyposis. Int J Cancer. 95:102–107. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Lefevre JH, Parc Y, Kerneis S, Goasguen N, Benis M, Parc R and Tiret E: Risk factors for development of desmoid tumours in familial adenomatous polyposis. Br J Surg. 95:1136–1139. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Out AA, Tops CM, Nielsen M, Weiss MM, van Minderhout IJ, Fokkema IF, Buisine MP, Claes K, Colas C, Fodde R, et al: Leiden open variation database of the MUTYH gene. Hum Mutat. 31:1205–1215. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Mazzei F, Viel A and Bignamia M: Role of MUTYH in human cancer. Mutat Res. 743-744:33–43. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Colombo C, Miceli R, Lazar AJ, Perrone F, Pollock RE, Le Cesne A, Hartgrink HH, Cleton-Jansen AM, Domont J, Bovée JV, et al: CTNNB1 45F mutation is a molecular prognosticator of increased postoperative primary desmoid tumor recurrence: An independent, multicenter validation study. Cancer. 119:3696–3702. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Lazar AJ, Tuvin D, Hajibashi S, Habeeb S, Bolshakov S, Mayordomo-Aranda E, Warneke CL, Lopez-Terrada D, Pollock RE and Lev D: Specific mutations in the beta-catenin gene (CTNNB1) correlate with local recurrence in sporadic desmoid tumors. Am J Pathol. 173:1518–1527. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Lev D, Kotilingam D, Wei C, Ballo MT, Zagars GK, Pisters PW, Lazar AA, Patel SR, Benjamin RS and Pollock RE: Optimizing treatment of desmoid tumors. J Clin Oncol. 25:1785–1791. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Mullen JT, Delaney TF, Kobayashi WK, Szymonifka J, Yeap BY, Chen YL, Rosenberg AE, Harmon DC, Choy E, Yoon SS, et al: Desmoid tumor: Analysis of prognostic factors and outcomes in a surgical series. Ann Surg Oncol. 19:4028–4035. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Crago AM, Denton B, Salas S, Dufresne A, Mezhir JJ, Hameed M, Gonen M, Singer S and Brennan MF: Prognostic nomogram for prediction of recurrence in desmoid fibromatosis. Ann Surg. 258:347–353. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Stoeckle E, Coindre JM, Longy M, Binh MB, Kantor G, Kind M, de Lara CT, Avril A, Bonichon F and Bui BN: A critical analysis of treatment strategies in desmoid tumours: A review of a series of 106 cases. Eur J Surg Onco. 35:129–134. 2009. View Article : Google Scholar

27 

Briand S, Barbier O, Biau D, Bertrand-Vasseur A, Larousserie F, Anract P and Gouin F: Wait-and-see policy as a first-line management for extra-abdominal desmoid tumors. J Bone Joint Surg Am. 96:631–638. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Bonvalot S, Eldweny H, Haddad V, Rimareix F, Missenard G, Oberlin O, Vanel D, Terrier P, Blay JY, Le Cesne A and Le Péchoux C: Extra-abdominal primary fibromatosis: Aggressive management could be avoided in a subgroup of patients. Eur J Surg Oncol. 34:462–468. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Hansmann A, Adolph C, Vogel T, Unger A and Moeslein G: High-dose tamoxifen and sulindac as first-line treatment for desmoid tumors. Cancer. 100:612–620. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Waddell WR and Kirsch WM: Testolactone, sulindac, warfarin, and vitamin K1 for unresectable desmoid tumors. Am J Surg. 161:416–421. 1991. View Article : Google Scholar : PubMed/NCBI

31 

Patel SR and Benjamin RS: Desmoid tumors respond to chemotherapy: Defying the dogma in oncology. J Clin Oncol. 24:11–12. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Patel SR, Evans HL and Benjamin RS: Combination chemotherapy in adult desmoid tumors. Cancer. 72:3244–3247. 1993. View Article : Google Scholar : PubMed/NCBI

33 

Li W, Zhou Y, Li Q, Tong H and Lu W: Intestinal perforation during chemotherapeutic treatment of intra-abdominal desmoid tumor in patients with Gardner's syndrome: Report of two cases. World J Surg Oncol. 14:1782016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang J, Jia N, Lin Q, Huang Y, Li J, Jiang Q, Liu W, Xu J, Hou Y, Liu J, Liu J, et al: Clinicopathological and molecular characteristics of abdominal desmoid tumors in the Chinese population: A single‑center report of 15 cases. Oncol Lett 18: 6443-6450, 2019
APA
Wang, J., Jia, N., Lin, Q., Huang, Y., Li, J., Jiang, Q. ... Tong, H. (2019). Clinicopathological and molecular characteristics of abdominal desmoid tumors in the Chinese population: A single‑center report of 15 cases. Oncology Letters, 18, 6443-6450. https://doi.org/10.3892/ol.2019.11038
MLA
Wang, J., Jia, N., Lin, Q., Huang, Y., Li, J., Jiang, Q., Liu, W., Xu, J., Hou, Y., Liu, J., Li, M., Lu, W., Zhou, Y., Zhang, Y., Tong, H."Clinicopathological and molecular characteristics of abdominal desmoid tumors in the Chinese population: A single‑center report of 15 cases". Oncology Letters 18.6 (2019): 6443-6450.
Chicago
Wang, J., Jia, N., Lin, Q., Huang, Y., Li, J., Jiang, Q., Liu, W., Xu, J., Hou, Y., Liu, J., Li, M., Lu, W., Zhou, Y., Zhang, Y., Tong, H."Clinicopathological and molecular characteristics of abdominal desmoid tumors in the Chinese population: A single‑center report of 15 cases". Oncology Letters 18, no. 6 (2019): 6443-6450. https://doi.org/10.3892/ol.2019.11038