JCDR - Register at Journal of Clinical and Diagnostic Research
Journal of Clinical and Diagnostic Research, ISSN - 0973 - 709X
Surgery Section DOI : 10.7860/JCDR/2016/19451.8596
Year : 2016 | Month : Oct | Volume : 10 | Issue : 10 Full Version Page : PC01 - PC05

Desmoid Tumours in Familial Adenomatous Polyposis: Review of 17 Patients from a Portuguese Tertiary Center

Marco Santos1, Anabela Rocha2, Vilma Martins3, Marisa Santos4

1 General Surgeons, Department of Surgery, General Surgery Service, Centro Hospitalar do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.
2 General Surgeons, Department of Surgery, General Surgery Service, Centro Hospitalar do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.
3 General Surgeons, Department of Surgery, General Surgery Service, Centro Hospitalar do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.
4 General Surgeons, Department of Surgery, General Surgery Service, Centro Hospitalar do Porto, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal.


NAME, ADDRESS, E-MAIL ID OF THE CORRESPONDING AUTHOR: Dr. Marco Santos, Rua São Tomé e Príncipe, nr 376, hab 3.3, 4430-228, Vila Nova de Gaia, Portugal.
E-mail: msilvasantos@gmail.com
Abstract

Introduction

Desmoid Tumours (DT) are benign tumours with an estimated incidence of 2-4 per million per year. Between 7-16% of them are associated with Familial Adenomatous Polyposis (FAP) and are mostly parietal or intra-abdominal. They are a challenge in relation to their unpredictable natural course, associated complications and difficult treatment.

Aim

The aim of the present study was to review the occurrence, management and follow-up of DT on FAP patients treated condivutively at a tertiary care center.

Materials and Methods

A retrospective review of clinical data from patients treated condivutively between 1993 and 2014. Patients’ data was gathered from clinical records. Data collection included the following variables: demographic data, genotype, FAP phenotype, data on FAP related surgery, DT diagnosis, location, size and number, DT treatment, patients’ status and follow-up data.

Results

The study population consisted of 17 patients from 9 families; with a mean age of 41 years, mostly women (59%) and most with a mutation either on codon 232 or 554. Most tumours had an intra-abdominal component (59%) with a mean size of 5cm. Fifteen patients were first treated with pharmacotherapy (Non-steroidal Anti-inflammatory Drugs (NSAIDs) and Tamoxifen). Five patients (29%) underwent surgery, 4 of them for complications of intra-abdominal tumours and 1 patient for abdominal wall tumours. Two patients underwent chemotherapy in relation to aggressive intra-abdominal disease. The mean follow-up time since diagnosis of DT was 123 months. Overall, 2 patients had remission, 11 patients had regression or stabilized disease, and 2 patients had progression. One patient died due to surgical complications.

Conclusion

Diagnosis of DT is based on clinical symptoms, without the need for screening, although imaging plays an important role once diagnosis is suspected. The treatment approach is conservative on most patients, leaving surgery for DT related complications. The follow-up of patients with DT is also based on clinical symptoms.

Keywords

Introduction

Desmoid Tumours (DT), also known as aggressive fibromatosis, is part of a group of fibroproliferative diseases. They result from the proliferation of monoclonal cells of fibroblastic origin. They don’t metastasize, hence they are not considered malignant. They can however be aggressive due to their infiltrative nature, possible multifocality and tendency to recur [15].

These tumours are rare, with an estimated incidence of 2-4 per million per year [4]. They are mostly sporadic but between 7-16% are associated with Familial Adenomatous Polyposis (FAP), as part of Gardner’s syndrome [1,6,7]. DT are an important cause of morbidity and mortality in patients with FAP, occurring in 12-15% of these patients [1,8].

FAP is related to mutations on Adenomatous Polyposis Coli gene (APC), which is a tumour suppressor gene. APC protein degrades β-catenin, which is implicated in cell proliferation regulation. The truncated APC protein is unable to perform its function appropriately [9]. The mutation site on the APC gene is associated to FAP phenotype, including DT development. The more distal the mutation (closer to 3’ end), the higher the risk of the patient being affected by DT [1,9,10].

Other factors involved in the development of DT are previous abdominal surgery and family history of DT [11,12]. Female sex has been reported as a risk factor but findings are not consistent [11,12].

Despite the possibility of DT arising in any location, DT related to FAP are mostly on the abdominal region: intra-abdominal, on the abdominal wall and “transabdominal” [1,7].

Some of them take a benign course, with indolent evolution, stabilization of growth or even remission. Others show an aggressive behaviour with rapid growth and mass effect on surrounding structures, particularly in case of intra-abdominal DT. Possible complications of intra-abdominal DT are intestinal obstruction, ischemia, haemorrhage and perforation or ureteric obstruction [1,4,8].

When DT develops in FAP patients, they are the second most common cause of mortality [13]. DT are challenging because of their unpredictable natural course, associated complications and difficult treatment. The aim of the present study was to review the occurrence, management and follow-up of DT on FAP patients treated consecutively at a tertiary center.

Materials and Methods

This was a retrospective review of a cohort of patients with FAP, treated consecutively at a Digestive Surgery Unit of a single tertiary referral center, between 1993 and 2014, who developed DT. Patients underwent clinical evaluation every 6 months.

The study was approved by the general surgery scientific commission and institutional review board. We consulted patients’ clinical records and gathered data with full anonymity.

Data collection included the following variables: demographic data (age, sex), genotype (APC mutation), FAP phenotype, data on FAP related surgery (age, type of surgery), DT diagnosis, location, size and number, DT treatment, patient’s status and follow-up data. Considering the location of tumours, if a patient had at least one tumour with an intra-abdominal component, we classified them as intra-abdominal.

Patients were also staged according to Church et al., which is a proposed staging system for intra-abdominal DT [8]. This staging system allows stratification of this set of patients by disease severity. In case of multiple DT, the clinically more relevant lesion was used to classify the disease.

Results

Our population consisted of 17 patients, belonging to 9 families, most being women (59%) with mean age of 41years. Ten patients belonging to 2 families had mutations on codon 232 and 554 (5 patients each). Five patients from different families had mutations on codons 554, 625, 1009, 1309 and 1465. The APC mutation could not be determined on 2 unrelated patients. These patients were also tested for MUTYH gene mutations and microsatellite instability with negative results. Patients with mutation on codon 232 (all from same family) had attenuated FAP. All other patients had classic FAP.

[Table/Fig-1] gives an overview of our population of FAP patients with desmoids. All patients underwent prophylactic colorectal surgery before diagnosis of DT. The FAP related surgery was a coloprotectomy with ileoanal pouch in 10 patients (59%) or a colectomy with ileorrectal anastomosis (41%) according to rectal presentation of the disease. The mean time between surgery and the first DT diagnosis was 60months (6-94months), with a mean age of 30 years at DT diagnosis.

Population overview: demographic, clinical, treatment and outcome data of FAP patients With DT.

N*SexFAP SurgeryAge (years)DT LocationDT treatmentMonths of follow-upOutcome
FAP surgeryDT diagnosis
1FIRA3738AW§NSAID’s / surgery216Progression
2FCPIP2526IA||NSAID’s216Remission
3FIRA2728IANSAID’s / tamoxifen216Stable
4MCPIP2528IANSAID’s / tamoxifen204Stable
5FCPIP3943IAtamoxifen / surgery141Stable
6MIRA3236AW / IAsurgery / chemo**/ NSAID’s146Stable
7FIRA2929AWNSAID’s144Stable
8FIRA5454IANSAID’s/ surgery36Death
9FCPIP1819AWNSAID’s / tamoxifen108Regression
10FCPIP2031IANSAID’S / tamoxifen / surgery106Remission
11MCPIP1718AWNSAID’s / tamoxifen106Progression
12MIRA1819AWNSAID’s67Stable
13FCPIP1730IANSAID’s72Stable
14MIRA3347IANSAID’s252Regression
15FCPIP2222AW / IANSAID’s/ tamoxifen / chemo30Regression
16MCPIP1819AWNSAID’s29Stable
17MCPIP1922AW-6Stable
TotalNNMeanMeanNNMeanN
1710 F7 M10 CPIP7 IRA2630 (19-54)8 - IA7 - AW2 - AW / IA15 - NSAID’s7 - tamoxifen5 – surgery2 - chemo123 (6-252)9 - stable3 - regression2 - remission2 - progression1 - death

*N- refers to number of patients

IRA – colectomy with ileorrectal anastomosis

CPIP – coloprotectomy with ileoanal pouch

§AW – abdominal wall

||IA – intra-abdominal

**chemo – chemotherapy


The diagnosis was first suspected clinically in 13 patients (76%) on palpation of an abdominal mass and then confirmed and characterized by Computed Tomography (CT) scan. Three patients presented with ileoanal pouch dysfunction and were investigated with CT scan. One patient had diagnosis during surgery for Gastrointestinal Haemorrhage (GIH).

Ten patients (59%) had intra-abdominal tumours with a mean size of 5cm, while the rest were located on the abdominal wall (41%) with a mean size of 4.5cm. One patient developed 1 intra-thoracic tumour diagnosed during follow-up. [Table/Fig-2] presents more information regarding intra-abdominal tumours.

Characterization of intra-abdominal DT at diagnosis.

Number of DTNumber of patients
15
24
>21
Staging (Church
I4
II-
III4
IV2
Time between surgery and diagnosisMonths: mean (min-max)
All patients60 (2-372)
stage I279
stage II-
stage III24
stage IV69

Fifteen patients were first treated with pharmacotherapy [Table/Fig-1,3]. Pharmacotherapy consisted of Non-steroidal Anti-inflammatory Drugs (NSAIDs) as first line of treatment– sulindac until it was discontinued in Portugal in 2008, and then celecoxib. Tamoxifen was the second line of treatment, mostly associated with NSAIDs, usually with progressive dosing as required, up to 120mg/day. Two patients didn’t tolerate NSAIDs; one because of profuse diarrhea, the other had ileoanal pouch bleeding. The former stayed on tamoxifen alone, the latter had no therapy since he remained asymptomatic and the tumour remained stable. Ten patients (59%) underwent pharmacotherapy as the sole treatment during follow-up time. The remaining patients underwent either surgery or chemotherapy, except one patient who had a recent clinically detected abdominal wall DT. He had a pericentimetric tumour and was asymptomatic. He was left untreated at the time of diagnosis, in order to assess the presence of other DT at different locations by CT scan.

Management of patients.

Staging *Treatment$Number of patients
INSAIDs2
Tamoxifen - surgery1
NSAIDs and tamoxifen –surgery1
II-0
IIINSAIDs1
NSAIDs and tamoxifen2
NSAIDs –surgery1
IVNSAIDs and tamoxifen - chemotherapy1
surgery – chemotherapy – NSAIDs1

* initial staging

$ at any point


Five patients (29%) have been submitted to DT related surgery. Despite different initial staging [Table/Fig-3], the indication for surgery was related to complications on 4 patients which had intra-abdominal DT – they all evolved to stage IV disease:

- One patient was initial stage III - slow growing of DT – and started NSAIDs at diagnosis. On the second year of DT follow-up, the patient had a small bowel perforation associated with bowel erosion by the tumour. An enterectomy was performed and the resection was considered R2. The patient died of septic complications in the late post-operative period, without anastomotic leakage.

- One patient started NSAIDs and tamoxifen once diagnosed with intra-abdominal DT. Nine months later, the patient progressed from stage I to stage IV and underwent emergent surgery for GIH– R0 resection.

- One patient was diagnosed at the time of emergent surgery. The patient underwent a R2 resection and needed chemotherapy.

- The fourth patient who underwent surgery for intra-abdominal DT was also stage I at the time of diagnosis. He was given NSAIDs but they were not tolerated (profuse diarrhea). For 7 years this patient’s therapy consisted of tamoxifen with regression of DT. She then progressed to stage IV, presenting extensive portal venous gas in relation to small bowel and mesenteric venous erosion by DT – a complete resection was performed.

- Only one patient with abdominal wall DT had surgery [Table/Fig-1], based on referred discomfort, having two DT resected with histologic free margins. Both DT recurred on this patient, who also developed an intra-abdominal and intra-thoracic DT. The patient is currently treated with NSAIDs and tamoxifen.

Both patients with histologically free margins had DT recurrence and started pharmacotherapy. One patient is in remission while the other remains stable.

Only 2 patients underwent chemotherapy in relation to aggressive intra-abdominal disease, with progressive symptoms, and no response to NSAIDs or tamoxifen. Both patients were stage IV at presentation. One patient was already described and underwent surgery as first treatment but the other patient had no indication to surgery based on aggressive disease with no chance of resection free of iatrogenic complications. Chemotherapy regimen consisted of doxorubicin and dacarbazine, on both patients. Both had regression of disease and they are asymptomatic.

The mean follow-up time since diagnosis of DT was 123 months. Information about current patient status is presented on [Table/Fig-4].

Patients status.

Patient statusNumber of patients
Dead1
Symptomatic1
Asymptomatic15

Discussion

Our population of 17 patients from a single institution is in line with DT prevalence on FAP patients. The largest series reported are from multi-institutional or national studies [11]. Nieuwenhuis et al., reported 78 patients from a national Dutch Polyposis Registry [14].

The most important risk factors for the development of DT are previous surgery, family history and the position of the mutation on the APC gene [1,912,16]. There is no current evidence associating the type of surgery and the occurrence of DT. All patients from our population have been submitted to prophylactic colectomy/coloprotectomy before the diagnosis of DT.

Ten of our patients (59%) belong to 2 families which is consistent with what is called family clustering [11,15,16]. Studies showed that tumours tended to cluster in susceptible individuals with a strong family history of DT, irrespective of the germline APC mutation [15].

It is known that the phenotype of FAP is related to the position of the APC gene mutation [9]. Regarding extra colonic manifestations of FAP, some patients are affected more severely than others, depending on the site of the mutation [10]. There is some evidence that mutations on the 3’ end of the APC gene, particularly distal to codon 1399, might be associated with a greater tendency to development of DT, but findings are not always consistent [9,10,15]. This study population had a high frequency of proximal mutations (closer to 5’ end) on the APC gene. Only 2 patients had mutations within the hotspot region between codons 1250 and 1464 [9]. This mutational pattern is not frequently reported on FAP patients with DT development [9].

According to Church et al., patients which have the shortest interval between surgery and DT appearance have the highest staging [8,17,18]. There seems to be such a relation on this study population. Patients classified as stage I have clearly the longest interval from surgery to DT diagnosis [Table/Fig-2].

The diagnosis of FAP related DT is made clinically in relation to a mass on the abdominal region, while investigating symptoms or incidentally on imaging or surgery. In this study population most DT were detected clinically, but four patients with intra-abdominal tumours were diagnosed by CT scan, during the investigation of ileoanal pouch dysfunction. Such presentation should prompt investigation and imaging is essential because DT is a possible aetiology, particularly in known affected families. CT is the most common image modality for diagnosis confirmation, identifying other DT and their characterization, such as determining the extent of invasion onto vital structures [4]. All of our patients underwent CT for this purpose. MRI might be useful when elective surgery is contemplated for an intra-abdominal DT [4]. There is no recommendation to actively search for DT on FAP patients, since they are benign and their clinical importance is related to symptoms. Although rare, it is possible for a DT to present itself as a life threatening complication, such as a GIH as reported in this study.

Treatment for DT may be challenging. Surgery has been advocated as the gold standard whenever feasible [1,3,19]. Currently, many authors consider medical treatment or even a wait-and-see approach on the management of these patients, since >50% have a slow growing or potentially regressive disease [5]. In fact, the Italian and French Sarcoma Group review, contemplate a wait-and-see approach for most patients [5].

There are some questions when considering a low threshold surgical approach. First of all, DT shows a variable behaviour and so far there are no reliable factors that predict it. Surgery itself is a possible trigger of further tumorigenesis [11]. Even though free histologic margins seem to be a prognostic factor of local recurrence, there are reports that show a less clear relation [4,5,2022]. Recurrences might occur after complete resections. Moreover, patients that develop recurrence after surgery may remain stable without further treatment [3]. As such, negative margins should be achieved only if not associated with increased morbidity.

In this study there is only one abdominal wall DT resection reported. The margins were negative but the patient had recurrence. At that time the decision to perform surgery was based on the available literature. The evidence that most patients did not have progressive disease, led us to a conservative non-surgical approach for the remainder of patients with abdominal wall DT.

However, FAP related DT is mostly intra-abdominal [13]. The usual infiltrative pattern of mesenteric DT poses difficulties regarding surgery. The required complex surgeries, most often with incomplete resections, the high risk of iatrogenic injuries, the possibility of intestinal failure and the uncertainty of results, are major drawbacks to perform surgery on such patients [15,2224]. Surgery should be planned if complications are eminently expected or actually happen.

We performed surgery only on four patients with intra-abdominal DT, all because of complications, most notably gastrointestinal haemorrhage and perforation.

There are various options for medical treatment of DT despite a lack of full knowledge of their mechanisms of action [25]. Currently there are no randomized trials comparing different options.

The most common medical treatment consists of NSAIDs. Indomethacin and sulindac (long-acting analogue of indomethacin), have been tested with variable reported responses [4,26]. Celecoxib, frequently prescribed for control of duodenal and colic adenomas, is also used for DT treatment but there is no clear evidence of its role on this setting [27].

There is some evidence that oestrogen stimulate growth of DT. As such, hormone therapy has been investigated and described as effective in the treatment of DT, particularly Tamoxifen [4,20,26]. Tested doses are as high as 120mg [26].

The combination of hormone therapy with NSAIDs is widely used since it is associated with low toxicity and possibly better outcome than isolated therapies [4]. We have used NSAIDs on most patients and used them associated with tamoxifen on patients who did not respond or did not tolerate NSAIDs. We did not use this strategy on 2 patients. One patient underwent emergent surgery for stage IV disease while the other patient had the clinical diagnosis of a small parietal asymptomatic DT three months ago and still awaits better characterization with a CT scan to evaluate the adequate therapy, including a possible wait and see approach.

Radiotherapy (RT) is considered a useful adjuvant therapy for DT, particularly when there are positive or close surgical margins. There are limitations to the use of RT in FAP patients, because most DT are located in the abdomen, and on this location, it is associated with serious side effects. We did not offer this treatment to any patient.

Imatinib has been studied, as well as other tyrosine kinase inhibitors. So far results vary between stabilization and 15% response rates. Its use is only justifiable in a trial setting [4,25].

Interferon has been scarcely reported as an optionand currently is not recommended [4,26].

Cytotoxic chemotherapy is reserved for patients with aggressive disease, who don’t respond to less toxic treatment and when DT are not amenable to resection. The major concern is its use for a benign disease on a generally young population, and its late side effects. The French Sarcoma Group has reviewed the effect of chemotherapy in patients with DT and concluded there is a clear benefit, particularly with anthracyclines, despite side effects [28]. Our regimen consisted, on both patients, of doxorubicin and dacarbazine which is a common protocol on this setting [28]. Other common protocol is methotrexate and vinblastine/vinorelbine, considered safer in terms of toxicity profile [5,28]. There are no follow-up recommendations on these patients in the literature.

Conclusion

Diagnosis of DT in PAF patients is based on clinical symptoms, without the need for screening, although imaging plays an important role once diagnosis is suspected. The treatment approach is conservative on most patients, mainly with NSAIDs and tamoxifen, leaving surgery for DT related complications. Most patients follow an indolent path. The follow-up of patients with DT is also based on clinical symptoms.

Ethical approval: Approved by Ethic Committee at Centro Hospitalar do Porto. Document available if requested.

*N- refers to number of patientsIRA – colectomy with ileorrectal anastomosisCPIP – coloprotectomy with ileoanal pouch§AW – abdominal wall||IA – intra-abdominal**chemo – chemotherapy* initial staging$ at any point

References

[1]Sakorafas GH, Nissotakis C, Peros G, Abdominal desmoid tumors Surg Oncol 2007 16(2):131-42.  [Google Scholar]

[2]Escobar C, Munker R, Thomas JO, Li BD, Burton G V, Update on desmoid tumors Ann Oncol 2012 23(3):562-69.  [Google Scholar]

[3]Bonvalot S, Desai a, Coppola S, The treatment of desmoid tumors: a stepwise clinical approach Ann Oncol 2012 23(Suppl 1(Supplement 10)):x158-66.  [Google Scholar]

[4]Devata S, Chugh R, Desmoid tumors: A comprehensive review of the evolving biology, unpredictable behavior, and myriad of management options Hematol Oncol Clin North Am 2013 27(5):989-1005.  [Google Scholar]

[5]Gronchi A, Colombo C, Le Péchoux C, Sporadic desmoid-type fibromatosis: a stepwise approach to a non-metastasising neoplasm—a position paper from the Italian and the French Sarcoma Group Ann Oncol 2014 25(3):578-83.  [Google Scholar]

[6]Nieuwenhuis MH, Casparie M, Mathus-Vliegen LMH, Dekkers OM, Hogendoorn PCW, Vasen HF a, A nation-wide study comparing sporadic and familial adenomatous polyposis-related desmoid-type fibromatoses Int J Cancer 2011 129(1):256-61.  [Google Scholar]

[7]Fallen T, Wilson M, Morlan B, Lindor NM, Desmoid tumors - a characterization of patients seen at Mayo Clinic 1976-1999 Fam Cancer 2006 5(2):191-94.  [Google Scholar]

[8]Church J, Berk T, Boman BM, Staging intra-abdominal desmoid tumors in familial adenomatous polyposis: a search for a uniform approach to a troubling disease Dis Colon Rectum 2005 48(8):1528-34.  [Google Scholar]

[9]Nieuwenhuis MH, Vasen HF a, Correlations between mutation site in APC and phenotype of familial adenomatous polyposis (FAP): a review of the literature Crit Rev Oncol Hematol 2007 61(2):153-61.  [Google Scholar]

[10]Enomoto M, Konishi M, Iwama T, Utsunomiya J, Sugihara KI, Miyaki M, The relationship between frequencies of extracolonic manifestations and the position of APC germline mutation in patients with familial adenomatous polyposis Jpn J Clin Oncol 2000 30(2):82-88.  [Google Scholar]

[11]Nieuwenhuis MH, Lefevre JH, Bülow S, Family history, surgery, and APC mutation are risk factors for desmoid tumors in familial adenomatous polyposis: an international cohort study Dis Colon Rectum 2011 54(10):1229-34.  [Google Scholar]

[12]Sinha A, Tekkis PP, Gibbons DC, Phillips RK, Clark SK, Risk factors predicting desmoid occurrence in patients with familial adenomatous polyposis: a meta-analysis Colorectal Dis 2011 13(11):1222-29.  [Google Scholar]

[13]Arvanitis ML, Jagelman DG, Fazio VW, Lavery IC, McGannon E, Mortality in patients with familial adenomatous polyposis Dis Colon Rectum 1990 33(8):639-42.  [Google Scholar]

[14]Nieuwenhuis MH, Mathus-Vliegen EM, Baeten CG, Evaluation of management of desmoid tumours associated with familial adenomatous polyposis in Dutch patients Br J Cancer 2011 104(1):37-42.  [Google Scholar]

[15]Sturt NJH, Gallagher MC, Bassett P, Evidence for genetic predisposition to desmoid tumours in familial adenomatous polyposis independent of the germline APC mutation Gut 2004 53(12):1832-36.  [Google Scholar]

[16]Bertario L, Russo A, Sala P, Genotype and phenotype factors as determinants of desmoid tumors in patients with familial adenomatous polyposis Int J Cancer 2001 95(2):102-07.  [Google Scholar]

[17]Leal RF, Silva PVVT, Lourdes M De, Ayrizono S, Fagundes JJ, Desmoid tumours in patients with FAP - Univ Campinas Arq Gastroenterol 2010 (4):373-78.  [Google Scholar]

[18]Church J, Lynch C, Neary P, LaGuardia L, Elayi E, A desmoid tumor-staging system separates patients with intra-abdominal, familial adenomatous polyposis-associated desmoid disease by behavior and prognosis Dis Colon Rectum 2008 51(6):897-901.  [Google Scholar]

[19]Clark SK, Neale KF, Landgrebe JC, Phillips RK, Desmoid tumours complicating familial adenomatous polyposis Br J Surg 1999 86(9):1185-89.  [Google Scholar]

[20]Joglekar SB, Rose PS, Sim F, Okuno S, Petersen I, Current perspectives on desmoid tumors: the mayo clinic approach Cancers (Basel) 2011 3(3):3143-55.  [Google Scholar]

[21]Ghert M, Yao X, Corbett T, Treatment and follow-up strategies in desmoid tumours: a practice guideline Curr Oncol 2014 21(4):e642-49.  [Google Scholar]

[22]Stoeckle E, Coindre JM, Longy M, A critical analysis of treatment strategies in desmoid tumours: a review of a series of 106 cases Eur J Surg Oncol 2009 35(2):129-34.  [Google Scholar]

[23]Quintini C, Ward G, Shatnawei A, Mortality of intra-abdominal desmoid tumors in patients with familial adenomatous polyposis: a single center review of 154 patients Ann Surg 2012 255(3):511-16.  [Google Scholar]

[24]Fiore M, Rimareix F, Mariani L, Desmoid-type fibromatosis: a front-line conservative approach to select patients for surgical treatment Ann Surg Oncol 2009 16(9):2587-93.  [Google Scholar]

[25]Yao X, Corbett T, Gupta a a, A systematic review of active treatment options in patients with desmoid tumours Curr Oncol 2014 21(4):e613-29.  [Google Scholar]

[26]Janinis J, Patriki M, Vini L, Aravantinos G, Whelan JS, The pharmacological treatment of aggressive fibromatosis: a systematic review Ann Oncol 2003 14(2):181-90.  [Google Scholar]

[27]Vasen HF a, Möslein G, Alonso a, Guidelines for the clinical management of familial adenomatous polyposis (FAP) Gut 2008 57(5):704-13.  [Google Scholar]

[28]Garbay D, Le Cesne A, Penel N, Chemotherapy in patients with desmoid tumors: a study from the French Sarcoma Group (FSG) Ann Oncol 2012 23(1):182-86.  [Google Scholar]