ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Dermatofibrosarcoma protuberans: Epidemiology, pathogenesis, clinical presentation, diagnosis, and staging

Dermatofibrosarcoma protuberans: Epidemiology, pathogenesis, clinical presentation, diagnosis, and staging
Literature review current through: Jan 2024.
This topic last updated: Dec 05, 2022.

INTRODUCTION — Dermatofibrosarcoma protuberans (DFSP) is an uncommon locally aggressive cutaneous soft tissue sarcoma. Approximately 85 to 90 percent of DFSPs are low grade, while the remainder contain a high-grade sarcomatous component (which is usually a fibrosarcoma, designated DFSP-FS) and are considered to be intermediate-grade sarcomas [1]. Although they rarely metastasize (fewer than 5 percent of cases), all DFSP variants have a propensity to recur locally. Recurrence potential is directly related to the extent of resection. (See "Dermatofibrosarcoma protuberans: Treatment", section on 'Importance of resection margins'.)

This topic review will focus on the epidemiology, molecular pathogenesis, histology, clinical presentation, diagnosis, and staging of DFSP. Treatment of DFSP and issues specific to DFSP arising in the head and neck region are discussed in detail elsewhere. (See "Dermatofibrosarcoma protuberans: Treatment" and "Head and neck sarcomas", section on 'Desmoids and dermatofibrosarcoma protuberans'.)

EPIDEMIOLOGY — Dermatofibrosarcoma protuberans (DFSP) is a relatively rare tumor. Estimates of the overall incidence of DFSP in the United States are 0.8 to 4.5 cases per million persons per year [2-4]. In the United States, DFSP accounts for between 1 and 6 percent of all soft tissue sarcomas [5,6] and 18 percent of all cutaneous soft tissue sarcomas [3]. In a series of 12,114 cutaneous soft tissue sarcomas derived from the Surveillance, Epidemiology and End Results (SEER) tumor registry from 1992 through 2004, DFSP was second only to Kaposi sarcoma, which accounted for 71 percent of cases [3].

The Bednar or pigmented variant (distinguished by the dispersal of melanin-containing cells in an otherwise typical DFSP) is even less common. It represents fewer than 5 percent of all DFSP cases and is more common in Black individuals [4,7-10]. The fibrosarcomatous variant of DFSP (DFSP-FS) accounts for approximately 5 to 15 percent of DFSPs [11,12].

DFSP most often arises in adults in their thirties [1,4,13-15], but it has been described in all age groups including children and older adults; rarely it presents congenitally [1,4,8,14,16-18]. In one large series, children accounted for 6 percent of all cases of DFSP [19]. The so-called giant cell fibroblastoma is considered to be the juvenile form of DFSP, given its similar morphology, identical chromosomal translocation, and CD34 positivity [20-22]. (See 'Molecular diagnostics' below and 'Immunohistochemistry (IHC)' below.)

In some reports, patients with DFSP-FS are older at presentation (median age 56 versus 37 years [23]). (See 'Fibrosarcomatous variant' below.)

DFSP is found in similar frequencies in males and females, although some large series suggest a slight male predominance [1,24-27]. DFSP is more common in Black individuals than in White individuals (eg, 7.1 versus 3.6 per million in the SEER registry series described above [4]).

MOLECULAR PATHOGENESIS — Over 90 percent of dermatofibrosarcoma protuberans (DFSPs) are characterized by a unique translocation t(17;22) (q22;q13) [28-32]. This translocation usually results in the formation of supernumerary ring chromosomes, which are derived from chromosome 22 and contain low level amplified sequences from 17q22-qter and 22q10-q13.1. Less common, a linear derivative of chromosome 22 (linear der[22]) results, which is most often unbalanced.

Both the ring chromosomes and the translocated linear der(22) contain a unique fusion gene in which the gene for platelet-derived growth factor beta polypeptide (PDGFB gene) is fused with the highly expressed collagen type 1A1 (COL1A1) gene [33,34]. This places expression of the normally inhibited PDGFB gene under the activating control of the COL1A1 promoter. The resulting PDGFB/COL1A1 fusion protein is processed to produce fully functional PDGFB, which results in continuous autocrine activation of the platelet-derived growth factor receptor b (PDGFRb), a tyrosine kinase [35-37]. This molecular alteration, which has been demonstrated in over 90 percent of DFSPs, is thought to be fundamental to the development of the tumor [6,35,38,39].

To date, 34 different PDGFB/COL1A1 fusion variants have been described; the break point in PDGFB is constant, whereas in COL1A1, the break may occur in any of the exons coding the alpha-helical region (codons 6 to 49) [6]. There appears to be no association between the different variants of the PDGFB/COL1A1 fusion gene and the histopathologic subtype or clinical characteristics [40].

In most of the reported cases of DFSP in which t(17;22) has not been found, an alternative translocation, usually (but not inevitably [41]) involving the PDGFB gene on chromosome 22, has been detected [42-44].

Highly sensitive fluorescence in situ hybridization (FISH) assays have been developed to detect the PDGFB/COL1A1 fusion transcript, the presence of which can be useful diagnostically [38]. (See 'Molecular diagnostics' below.)

In addition, the constitutive activation of the tyrosine kinase receptor PDGFRb by the PDGFB/COL1A1 fusion transcript in the majority of cases provides the rationale for targeted inhibition of this pathway as a potential treatment strategy. (See "Dermatofibrosarcoma protuberans: Treatment", section on 'Molecularly targeted therapy'.)

Fibrosarcomatous variant — The fibrosarcomatous variant of DFSP (DFSP-FS) is also characterized by a high frequency of t(17;22), which supports the theory of common histogenesis of both the DFSP and the fibrosarcomatous tissue [26,45]. In some cases, the clonal evolution from DFSP to a DFSP-FS (or less commonly, DFSP with areas of pleomorphic sarcoma) involves genomic gains in the PDGFB/COL1A1 fusion gene [11,37,46,47]. These results suggest a role for the PDGFB/COL1A1 fusion protein in sarcomatous change in DFSP over time.

However, this is not a universal finding. Others describe the early development of microsatellite instability (MSI) and the later acquisition of TP53 mutations in cases with a high-grade sarcoma component [11,48-50]. As an example, Japanese investigators analyzed MSI and TP53 mutations in 44 tumors from 36 patients with either DFSP (n = 27) or DFSP-FS (n = 9) [48]. A high rate of MSI occurred significantly more often in DFSP-FS compared with DFSP (4 of 9 versus 1 of 27 patients). Mutational analysis revealed 10 TP53 point mutations in six patients, four of which were missense mutations and six silent. Three of the four missense mutations were found in the DFSP-FS group compared with only one in the DFSP group. These results suggest that a different molecular pathophysiology underlies the acquisition of high-grade sarcomatous change.

CLINICAL PRESENTATION — In its earliest stage, dermatofibrosarcoma protuberans (DFSP) most commonly presents as an asymptomatic, indurated plaque that slowly enlarges over months to years. The tumors are covered by skin-colored, brown-yellow, red-tinged, sclerodermiform, or telangiectatic atrophic skin. Early lesions may be violet-red or blue at the margins. The Bednar pigmented variant typically contains brown pigmentation and has an irregular surface [7,9]. Less common, DFSP presents as a firm cutaneous nodule.

As the tumor slowly enlarges, it becomes raised, firm, and nodular; the surrounding skin may be telangiectatic [13]. The nodule is often fixed to the dermis but moves freely over deeper tissues. Fixation to deeper structures is observed late in the course of the disease [13]. Once nodules appear, growth is often accelerated, and the tumor may ulcerate, bleed, or become painful. Untreated, DFSP can attain massive dimensions, producing the large "protuberant" nodules for which the disease is named. However, at the time of diagnosis, most tumors are superficial (77 percent in one series of 159 patients [1]) and less than 5 cm in diameter (84 percent in the same series [1]).

The most common location for a DFSP is on the trunk and proximal extremities, generally on the chest and shoulders [4,24,27,51]. The following site distribution was observed in a series of 6,817 patients reported to the Surveillance, Epidemiology, and End Results (SEER) database between 2000 and 2010 [4]:

Trunk – 42 percent

Lower extremity – 21 percent

Upper extremity – 21 percent

Head and neck – 13 percent

Genitals – 1 percent

Rarely, DFSP arises within a preexisting scar (including a vaccination scar) or tattoo [9,52-55]. Most DFSPs arise from the dermis; however, they may also arise in the subcutaneous tissues, sometimes as a mass in the breast [56,57] or on the head [58].

The majority of DFSPs exhibit an indolent growth pattern, and in most cases, the lesion has been present for years [13,59]. Indolent growth, coupled with the rarity of DFSP and the variability in clinical appearance, contributes to diagnostic delay. As is seen with desmoid tumors, accelerated growth may occur during pregnancy [60,61]. (See "Desmoid tumors: Epidemiology, molecular pathogenesis, clinical presentation, diagnosis, and local therapy".)

The clinical signs and symptoms associated with the fibrosarcomatous variant of DFSP (DFSP-FS) are similar to those of classic DFSP, and even though DFSP-FS tends to be larger and present for a longer period of time than conventional DFSP [26], neither history nor physical examination is helpful in recognizing the presence of a sarcomatous component.

Metastatic disease — Metastases to regional lymph nodes are extremely rare, with only a few case reports in the literature [27,62]. Distant hematogenous metastases are even rarer and are most likely in patients who have had multiple local recurrences after inadequate surgical resection [25]. Repeatedly recurring tumors have an increased risk for transformation into a more malignant form (DFSP-FS). The lungs are most frequently affected, but metastases to brain, bone, and other soft tissues are reported. Although some authors report a rate of metastases as high as 5 percent, it is probably closer to 1 percent for classic (low-grade) DFSP [25,27,63].

DIAGNOSIS — Dermatofibrosarcoma protuberans (DFSP) should be suspected in any patient with a history of a firm, slow-growing cutaneous nodule. Findings on dermoscopy (epiluminescence microscopy) can help to suspect DFSP [64], but definitive diagnosis requires a core needle or incisional biopsy. For de novo previously untreated tumors, fine needle aspirate (FNA) biopsy does not satisfactorily provide enough tissue to render an accurate diagnosis [65,66]. However, FNA may be useful to establish the diagnosis of recurrent disease in a previously treated patient [66].

Histology — Histologically, DFSP is composed of monomorphic, benign-appearing (low mitotic activity) spindle cells arranged in an irregularly whorled or storiform pattern said to resemble a straw mat [67]. The cells intersect in tight right angles around central vessels, and early lesions may have a narrow tumor-free zone (Grenz Zone) between the tumor and epidermis [13]. A predominant histologic characteristic of DFSP is its capacity to invade surrounding tissues to a considerable distance from the central focus of the tumor, with tumor cells invading subcutaneous tissue in the form of irregular tentacle-like projections through the septa and fat lobules [6]. These peripheral tumor extensions may have a bland appearance with few tumor cells, and at first glance, they can appear similar to normal fibrous tracts. This feature makes it difficult to determine the true border of the lesion. This is why local recurrences are frequent after excision with an apparently wide margin.

As noted above, an uncommon variant is the Bednar tumor, in which there are melanin-containing dendritic cells interspersed among the spindle cells [6-9]. Other variants are myxoid DFSP with prominent myxoid stromal changes and a multinodular growth pattern with numerous blood vessels, which may be difficult to differentiate from a myxoid liposarcoma [68-70]. Other described variants include sclerosing DFSP and the atrophic type of DFSP [2,6,8,71].

Between 10 and 20 percent of tumors contain a high-grade sarcomatous component that is usually, but not inevitably [27], a fibrosarcoma (hence the abbreviation DFSP-FS) [1,6,72-74]. The sarcomatous component typically occupies between 20 and 80 percent of the tumor (median, 60 percent) [74]. The spindle cells in the sarcomatous areas show marked cellular and nuclear pleomorphism, and the mitotic rate is usually (but not always) higher than in the typical DFSP component [74]. Upon immunohistochemical staining (IHC), staining for CD34 is often lost in the sarcomatous component. (See 'Immunohistochemistry (IHC)' below.)

Sarcomatous change of a DFSP is a form of tumor progression. The presence of a sarcomatous component increases the tumor grade from low to intermediate, but the impact on overall prognosis is debated. Most but not all series suggest a higher rate of metastases and a worse prognosis in these patients. The impact of a sarcomatous component on local recurrence rates (especially in patients who undergo optimal surgery) is even less clear; at least some of the discrepancy in reported results reflects the adequacy of surgical excision rather than a true difference in local aggressiveness of the DFSP-FS variant. These issues are discussed in detail elsewhere. (See "Dermatofibrosarcoma protuberans: Treatment", section on 'Prognosis'.)

In most cases, examination of a hematoxylin and eosin-stained specimen by light microscopy provides an unequivocal diagnosis of DFSP. However, it may be difficult to distinguish DFSP histologically from other mesenchymal neoplasms such as a dermatofibroma (the fibrous type of benign fibrous histiocytoma), fibromatosis (desmoid tumor), fibrosarcoma, leiomyosarcoma, undifferentiated/unclassified soft tissue sarcoma (formerly known as malignant fibrous histiocytoma or undifferentiated pleomorphic sarcoma), or atypical fibroxanthoma. In such cases, IHC or even molecular diagnostic testing for the characteristic fusion gene may be helpful. Updated guidelines for management of DFSP from the National Comprehensive Cancer Network (NCCN) [75] recommend appropriate and confirmatory immunostaining in all cases of suspected DFSP. (See 'Molecular pathogenesis' above.)

Immunohistochemistry (IHC) — DFSP usually stains positively for CD34, hyaluronate, and vimentin, and negatively for factor XIIIa (table 1) [2,6,67,76,77]. CD34 is one of the most useful stains to differentiate DFSP from dermatofibroma and other soft tissue tumors. The spindle cells of DFSP stain positively with CD34, while dermatofibromas are usually CD34-negative. In fact, the finding of CD34 positivity has renewed the debate over the cellular origins of DFSP. (See 'Implications of IHC for histogenesis' below.)

Unfortunately, the reliability of CD34 staining is not absolute. The sensitivity of CD34 IHC in DFSP ranges from 84 to 100 percent [67,76-78], while between 2.5 and 5 percent of dermatofibromas stain positively [67,78]. There are also reports of loss of CD34 expression with the transition of DFSP to DFSP-FS and in myxoid DFSP [6,74,79].

A more recent study suggests utility for IHC staining for hyaluronate (the major component of extracellular matrix) and CD44 (a membrane glycoprotein thought to be its cell surface receptor) in the differential diagnosis of dermatofibroma and DFSP. The cells of dermatofibromas tend to stain intensely for CD44, while its stroma stains faintly for hyaluronate [80]. In contrast, DFSP tends to be strongly positive for hyaluronate, while CD44 is significantly reduced or absent.

Additional immunostaining with nestin, apolipoprotein D, and cathepsin K may be considered for equivocal lesions [81-83]; alternatively, FISH (fluorescence in situ hybridization) or PCR (polymerase chain reaction) can be used to identify a characteristic translocation. (See 'Molecular diagnostics' below.)

Implications of IHC for histogenesis — The exact type of dermal mesenchymal cell from which DFSP arises is debated. Until more recently, the histochemical and electron microscopic evidence seemed to point toward a fibroblastic cellular origin, because DFSP cells stain uniformly for vimentin and contain active endoplasmic reticulum that synthesizes collagen [84,85]. However, the finding of CD34 positivity in DFSP has raised the possibility that dendritic cells in the skin may be responsible, possibly those derived from circulating hematopoietic progenitor cells. CD34 is a surface antigen that is expressed on normal hematopoietic stem cells that seems to be involved in the process of leukocyte adhesion to the vascular endothelium and in the control of immune responses. (See "Overview of hematopoietic stem cells".)

IHC staining characteristics suggest that in normal skin, there appear to be at least three different kinds of dermal mesenchymal cells, other than endothelial cells [86]:

CD34-negative/factor XIIIa-negative, as in the majority of fibroblasts in the reticular dermis

CD34-negative/factor XIIIa-positive, as in dermatofibromas

CD34-positive/factor XIIIa-negative, as in DFSP

In normal skin, the CD34-positive cells are a distinct population of spindle-shaped cells that are present in a perivascular, perifollicular, and peri-sweat gland distribution, and on interstitial dermal and subcutaneous dendritic cells. It is speculated that these dendritic cells might directly evolve from circulating hematopoietic progenitor cells that have become skin-associated and that they represent the cell of origin of DFSP [87]. This is an area of ongoing investigation.

Molecular diagnostics — As noted above, over 90 percent of cases of DFSP have a characteristic unbalanced translocation t(17;22) that places the PDGFB gene under the control of the COL1A1 promoter. The fusion transcript of this chimeric gene can be identified using reverse transcription polymerase chain reaction (RT-PCR) or fluorescence in situ hybridization (FISH). Both techniques were compared on a study of 103 archival DFSPs; although both techniques had a similar specificity (100 percent), FISH was more sensitive than RT-PCR for detection of the PDGFB/COL1A1 transcript (90 versus 72 percent) [32].

Routine molecular testing is not needed in all cases. Molecular testing should be used if the diagnosis is in doubt or to predict the likelihood of response to the tyrosine kinase inhibitor imatinib [88]. The multicenter GENSARC study, conducted in 32 French sarcoma centers found that molecular diagnostics altered the diagnosis in only 5 of 50 patients with DFSP (10 percent) when cases were reviewed by an expert group of pathologists [89]. (See 'Molecular pathogenesis' above and "Dermatofibrosarcoma protuberans: Treatment", section on 'Treatment of locally advanced, recurrent, and metastatic disease'.)

DIFFERENTIAL DIAGNOSIS — The clinical differential diagnosis of dermatofibrosarcoma protuberans (DFSP) is broad, and can include a number of diseases, depending on the size and location of the tumor. The nodular form of DFSP may be clinically confused with neurofibroma, leiomyoma, epidermal (sebaceous) cyst, malignant melanoma, basal cell carcinoma, keloid, desmoid tumor, dermatofibroma, lipoma, nodular fasciitis, sarcoidosis, and other cutaneous soft tissue sarcomas including Kaposi sarcoma, fibrosarcoma/fibromyxosarcoma, liposarcoma, leiomyosarcoma, angiosarcoma, and undifferentiated/unclassified soft tissue sarcoma (previously known as malignant fibrous histiocytoma, or undifferentiated pleomorphic sarcoma). (See "Overview of benign lesions of the skin" and "Classic Kaposi sarcoma: Clinical features, staging, diagnosis, and treatment" and "AIDS-related Kaposi sarcoma: Clinical manifestations and diagnosis" and "Ganglion cysts of the wrist and hand" and "Desmoid tumors: Epidemiology, molecular pathogenesis, clinical presentation, diagnosis, and local therapy" and "Keloids and hypertrophic scars" and "Basal cell carcinoma: Epidemiology, pathogenesis, clinical features, and diagnosis" and "Overview of soft tissue musculoskeletal disorders" and "Melanoma: Clinical features and diagnosis" and "Clinical manifestations and diagnosis of sarcoidosis" and "Clinical presentation, histopathology, diagnostic evaluation, and staging of soft tissue sarcoma".)

The plaque and atrophic forms of DFSP may clinically resemble morphea (localized scleroderma), morpheaform basal cell carcinoma, anetoderma (loss of normal elastic tissue sometimes seen with systemic lupus erythematosus or antiphospholipid syndrome), or scar [90,91]. (See "Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults" and "Basal cell carcinoma: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Morpheaform/infiltrative' and "Clinical manifestations of antiphospholipid syndrome".)

STAGING SYSTEM — There is no uniformly used staging system for dermatofibrosarcoma protuberans (DFSP); three are in use:

As with other soft tissue sarcomas, many clinicians stage DFSP and the fibrosarcomatous variant of DFSP (DFSP-FS) according to the American Musculoskeletal Tumor Society (MSTS) staging system, which is based on histologic grade (using a two-tiered system, low versus high) and whether the tumor is confined to an anatomic compartment (table 2) [92]. All DFSP fall into the category of stage IA or IB disease. The designation of stage II disease in the MSTS staging system implies high-grade histology and does not apply to DFSP or DFSP-FS.

There is an American Joint Committee on Cancer (AJCC)/ Union for international Cancer Control (UICC) staging system for soft tissue sarcomas that includes DFSP. The most recent version (eighth edition, 2017) provides a separate classification system for tumors of the extremities and trunk (table 3) and those arising in the head and neck (table 4) [93,94]. (See "Clinical presentation, histopathology, diagnostic evaluation, and staging of soft tissue sarcoma", section on 'Staging'.)

In Germany, a staging system was described (the Short German Guidelines for DFSP), but the clinical utility is unclear, as the vast majority of tumors fall into the stage I category [95]:

Stage I – Local tumor involvement

Stage II – Lymph node metastases

Stage III – Distant metastases

STAGING EVALUATION — Most dermatofibrosarcoma protuberans (DFSPs) are superficial, and tumor extent and mobility as well as regional node assessment can be assessed by physical examination. Because a characteristic of DFSP is asymmetric growth and frequent finger-like extensions, determining tumor size and extent of penetration into surrounding tissues is of utmost importance, and special attention should be given to palpation around the tumor and regional lymph nodes. The history, physical examination, and review of systems largely determine the extent of workup that is required.

The physical examination should include a complete skin examination. Imaging studies are not required in every case of DFSP, but can be helpful in select cases (large or recurrent tumors, suspicion of bone invasion) to define the extent of disease.

For purposes of treatment planning, magnetic resonance imaging (MRI) is useful to determine the deep extent of the tumor, particularly with large or recurrent lesions at sites other than the head and neck or upper thorax; it is more sensitive than palpation for ascertaining the depth of invasion [16,96,97]. On conventional T1-weighted images, DFSP can be isointense, hypointense, or hyperintense compared with normal skeletal muscle and typically has a lower signal intensity than subcutaneous fat [96]. On T2-weighted images, there may be higher signal intensity as compared with fat, or an intermediate signal intensity that is similar to fat.

Computed tomography (CT) scanning is not indicated except if underlying bone involvement is suspected.

Because of the rarity of both lymphatic and hematogenous dissemination, an extensive staging workup is unnecessary unless there are suggestive aspects in the history and physical examination for metastatic disease, if adverse prognostic histologic features are present (eg, fibrosarcomatous transformation), and for recurrent tumors [98]. Although metastases are rare overall, the lungs are the most common site. (See 'Metastatic disease' above.)

Some suggest chest CT to evaluate for lung metastases if the primary lesion is advanced, recurrent, and/or intermediate grade [63]. We usually get a chest radiograph preoperatively in all patients, and we preferentially order chest CT for those with advanced, unfavorable tumors of prolonged duration, recurrent tumors, or if the tumor has sarcomatous transformation (DFSP-FS).

Guidelines for treatment of DFSP from the National Comprehensive Cancer Network (NCCN) [75] do not make any recommendations for imaging of the primary tumor or staging workup.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Soft tissue sarcoma".)

SUMMARY

Dermatofibrosarcoma protuberans (DFSP) is a rare low- to intermediate-grade cutaneous sarcoma with little metastatic potential but a significant propensity for local recurrence. The recurrence potential is directly related to the extent of resection. (See 'Introduction' above.)

Over 90 percent of DFSPs have a translocation involving chromosomes 17 and 22, t(17;22). A fusion gene results (platelet-derived growth factor-beta [PDGFB]/collagen type 1A1 [COL1A1]), which causes dysregulated expression of PDGFB and continuous autocrine activation of the platelet-derived growth factor receptor b (PDGFRb), a tyrosine kinase. This provides both a unique molecular diagnostic test for DFSP and the rationale for targeted inhibition of this pathway as a potential treatment strategy. (See 'Molecular pathogenesis' above.)

DFSP most commonly presents as an asymptomatic, skin-colored indurated plaque that slowly enlarges over months to years. As the tumor slowly enlarges, it becomes raised, firm, and nodular. The most common locations are the trunk and proximal extremities. (See 'Clinical presentation' above.)

Definitive diagnosis requires a core needle or incisional biopsy. Most of the time, the correct diagnosis can be made by review of hematoxylin and eosin-stained sections, combined with immunohistochemical stating. Molecular testing for the PDGFB/COL1A1 fusion protein may help if the diagnosis is in doubt. (See 'Diagnosis' above.)

The clinical differential diagnosis of DFSP is broad. DFSP may be clinically confused with neurofibroma, leiomyoma, epidermal cyst, malignant melanoma, morpheaform basal cell carcinoma, keloid, desmoid tumor, Kaposi sarcoma, fibrosarcoma, dermatofibroma, nodular fasciitis, or sarcoidosis. (See 'Differential diagnosis' above.)

Most clinicians stage DFSP according to the American Musculoskeletal Tumor Society (MSTS) staging system, which is based on histologic grade (using a two-tiered system, low versus high) and whether the tumor is confined to an anatomic compartment (table 2). Most tumors are IA or IB. (See 'Staging system' above.)  

Imaging studies are not required in every case of DFSP, but magnetic resonance imaging (MRI) can be helpful in defining the extent of disease in cases of a large or recurrent primary tumor. Computed tomography (CT) scanning is not indicated except if underlying bone involvement is suspected. Because of the rarity of both lymphatic and hematogenous dissemination, an extensive staging workup is usually unnecessary in the absence of clinical suspicion for metastatic disease or adverse prognostic histologic features. We usually get a chest radiograph preoperatively in all patients, and we preferentially order chest CT for those with advanced, unfavorable tumors of prolonged duration or if the tumor has sarcomatous transformation (DFSP-FS). (See 'Staging evaluation' above.)

  1. Bowne WB, Antonescu CR, Leung DH, et al. Dermatofibrosarcoma protuberans: A clinicopathologic analysis of patients treated and followed at a single institution. Cancer 2000; 88:2711.
  2. Gloster HM Jr. Dermatofibrosarcoma protuberans. J Am Acad Dermatol 1996; 35:355.
  3. Rouhani P, Fletcher CD, Devesa SS, Toro JR. Cutaneous soft tissue sarcoma incidence patterns in the U.S. : an analysis of 12,114 cases. Cancer 2008; 113:616.
  4. Kreicher KL, Kurlander DE, Gittleman HR, et al. Incidence and Survival of Primary Dermatofibrosarcoma Protuberans in the United States. Dermatol Surg 2016; 42 Suppl 1:S24.
  5. Kransdorf MJ. Malignant soft-tissue tumors in a large referral population: distribution of diagnoses by age, sex, and location. AJR Am J Roentgenol 1995; 164:129.
  6. Llombart B, Serra-Guillén C, Monteagudo C, et al. Dermatofibrosarcoma protuberans: a comprehensive review and update on diagnosis and management. Semin Diagn Pathol 2013; 30:13.
  7. Yagi Y, Ueda K, Maruyama S, Noborio R. Bednar tumor: a report of two cases. J Dermatol 2004; 31:484.
  8. Marcus JR, Few JW, Senger C, Reynolds M. Dermatofibrosarcoma protuberans and the Bednar tumor: treatment in the pediatric population. J Pediatr Surg 1998; 33:1811.
  9. Elgart GW, Hanly A, Busso M, Spencer JM. Bednar tumor (pigmented dermatofibrosarcoma protuberans) occurring in a site of prior immunization: immunochemical findings and therapy. J Am Acad Dermatol 1999; 40:315.
  10. Dupree WB, Langloss JM, Weiss SW. Pigmented dermatofibrosarcoma protuberans (Bednar tumor). A pathologic, ultrastructural, and immunohistochemical study. Am J Surg Pathol 1985; 9:630.
  11. Abbott JJ, Erickson-Johnson M, Wang X, et al. Gains of COL1A1-PDGFB genomic copies occur in fibrosarcomatous transformation of dermatofibrosarcoma protuberans. Mod Pathol 2006; 19:1512.
  12. Stacchiotti S, Pedeutour F, Negri T, et al. Dermatofibrosarcoma protuberans-derived fibrosarcoma: clinical history, biological profile and sensitivity to imatinib. Int J Cancer 2011; 129:1761.
  13. Lindner NJ, Scarborough MT, Powell GJ, et al. Revision surgery in dermatofibrosarcoma protuberans of the trunk and extremities. Eur J Surg Oncol 1999; 25:392.
  14. Love WE, Keiler SA, Tamburro JE, et al. Surgical management of congenital dermatofibrosarcoma protuberans. J Am Acad Dermatol 2009; 61:1014.
  15. Chuang TY, Su WP, Muller SA. Incidence of cutaneous T cell lymphoma and other rare skin cancers in a defined population. J Am Acad Dermatol 1990; 23:254.
  16. Thornton SL, Reid J, Papay FA, Vidimos AT. Childhood dermatofibrosarcoma protuberans: role of preoperative imaging. J Am Acad Dermatol 2005; 53:76.
  17. Pappo AS, Rao BN, Cain A, et al. Dermatofibrosarcoma protuberans: the pediatric experience at St. Jude Children's Research Hospital. Pediatr Hematol Oncol 1997; 14:563.
  18. Weinstein JM, Drolet BA, Esterly NB, et al. Congenital dermatofibrosarcoma protuberans: variability in presentation. Arch Dermatol 2003; 139:207.
  19. McKee PH, Fletcher CD. Dermatofibrosarcoma protuberans presenting in infancy and childhood. J Cutan Pathol 1991; 18:241.
  20. Shmookler BM, Enzinger FM, Weiss SW. Giant cell fibroblastoma. A juvenile form of dermatofibrosarcoma protuberans. Cancer 1989; 64:2154.
  21. Macarenco RS, Zamolyi R, Franco MF, et al. Genomic gains of COL1A1-PDFGB occur in the histologic evolution of giant cell fibroblastoma into dermatofibrosarcoma protuberans. Genes Chromosomes Cancer 2008; 47:260.
  22. Diwan AH, Skelton HG 3rd, Horenstein MG, et al. Dermatofibrosarcoma protuberans and giant cell fibroblastoma exhibit CD99 positivity. J Cutan Pathol 2008; 35:647.
  23. Connelly JH, Evans HL. Dermatofibrosarcoma protuberans. A clinicopathologic review with emphasis on fibrosarcomatous areas. Am J Surg Pathol 1992; 16:921.
  24. Fiore M, Miceli R, Mussi C, et al. Dermatofibrosarcoma protuberans treated at a single institution: a surgical disease with a high cure rate. J Clin Oncol 2005; 23:7669.
  25. Rutgers EJ, Kroon BB, Albus-Lutter CE, Gortzak E. Dermatofibrosarcoma protuberans: treatment and prognosis. Eur J Surg Oncol 1992; 18:241.
  26. Llombart B, Monteagudo C, Sanmartín O, et al. Dermatofibrosarcoma protuberans: a clinicopathological, immunohistochemical, genetic (COL1A1-PDGFB), and therapeutic study of low-grade versus high-grade (fibrosarcomatous) tumors. J Am Acad Dermatol 2011; 65:564.
  27. Cai H, Wang Y, Wu J, Shi Y. Dermatofibrosarcoma protuberans: clinical diagnoses and treatment results of 260 cases in China. J Surg Oncol 2012; 105:142.
  28. Sirvent N, Maire G, Pedeutour F. Genetics of dermatofibrosarcoma protuberans family of tumors: from ring chromosomes to tyrosine kinase inhibitor treatment. Genes Chromosomes Cancer 2003; 37:1.
  29. Nishio J, Iwasaki H, Ishiguro M, et al. Supernumerary ring chromosome in a Bednar tumor (pigmented dermatofibrosarcoma protuberans) is composed of interspersed sequences from chromosomes 17 and 22: a fluorescence in situ hybridization and comparative genomic hybridization analysis. Genes Chromosomes Cancer 2001; 30:305.
  30. Simon MP, Pedeutour F, Sirvent N, et al. Deregulation of the platelet-derived growth factor B-chain gene via fusion with collagen gene COL1A1 in dermatofibrosarcoma protuberans and giant-cell fibroblastoma. Nat Genet 1997; 15:95.
  31. Naeem R, Lux ML, Huang SF, et al. Ring chromosomes in dermatofibrosarcoma protuberans are composed of interspersed sequences from chromosomes 17 and 22. Am J Pathol 1995; 147:1553.
  32. Salgado R, Llombart B, M Pujol R, et al. Molecular diagnosis of dermatofibrosarcoma protuberans: a comparison between reverse transcriptase-polymerase chain reaction and fluorescence in situ hybridization methodologies. Genes Chromosomes Cancer 2011; 50:510.
  33. Maki RG, Awan RA, Dixon RH, et al. Differential sensitivity to imatinib of 2 patients with metastatic sarcoma arising from dermatofibrosarcoma protuberans. Int J Cancer 2002; 100:623.
  34. Sandberg AA, Bridge JA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors. Dermatofibrosarcoma protuberans and giant cell fibroblastoma. Cancer Genet Cytogenet 2003; 140:1.
  35. Sjöblom T, Shimizu A, O'Brien KP, et al. Growth inhibition of dermatofibrosarcoma protuberans tumors by the platelet-derived growth factor receptor antagonist STI571 through induction of apoptosis. Cancer Res 2001; 61:5778.
  36. McArthur GA. Dermatofibrosarcoma protuberans: a surgical disease with a molecular savior. Curr Opin Oncol 2006; 18:341.
  37. Kiuru-Kuhlefelt S, El-Rifai W, Fanburg-Smith J, et al. Concomitant DNA copy number amplification at 17q and 22q in dermatofibrosarcoma protuberans. Cytogenet Cell Genet 2001; 92:192.
  38. Patel KU, Szabo SS, Hernandez VS, et al. Dermatofibrosarcoma protuberans COL1A1-PDGFB fusion is identified in virtually all dermatofibrosarcoma protuberans cases when investigated by newly developed multiplex reverse transcription polymerase chain reaction and fluorescence in situ hybridization assays. Hum Pathol 2008; 39:184.
  39. Shimizu A, O'Brien KP, Sjöblom T, et al. The dermatofibrosarcoma protuberans-associated collagen type Ialpha1/platelet-derived growth factor (PDGF) B-chain fusion gene generates a transforming protein that is processed to functional PDGF-BB. Cancer Res 1999; 59:3719.
  40. Llombart B, Sanmartín O, López-Guerrero JA, et al. Dermatofibrosarcoma protuberans: clinical, pathological, and genetic (COL1A1-PDGFB ) study with therapeutic implications. Histopathology 2009; 54:860.
  41. Bianchini L, Maire G, Guillot B, et al. Complex t(5;8) involving the CSPG2 and PTK2B genes in a case of dermatofibrosarcoma protuberans without the COL1A1-PDGFB fusion. Virchows Arch 2008; 452:689.
  42. Gisselsson D, Höglund M, O'Brien KP, et al. A case of dermatofibrosarcoma protuberans with a ring chromosome 5 and a rearranged chromosome 22 containing amplified COL1A1 and PDGFB sequences. Cancer Lett 1998; 133:129.
  43. Sonobe H, Furihata M, Iwata J, et al. Dermatofibrosarcoma protuberans harboring t(9;22)(q32;q12.2). Cancer Genet Cytogenet 1999; 110:14.
  44. Nakamura I, Kariya Y, Okada E, et al. A Novel Chromosomal Translocation Associated With COL1A2-PDGFB Gene Fusion in Dermatofibrosarcoma Protuberans: PDGF Expression as a New Diagnostic Tool. JAMA Dermatol 2015; 151:1330.
  45. Szollosi Z, Scholtz B, Egervari K, Nemes Z. Transformed dermatofibrosarcoma protuberans: real time polymerase chain reaction detection of COL1A1-PDGFB fusion transcripts in sarcomatous areas. J Clin Pathol 2007; 60:190.
  46. Wang J, Morimitsu Y, Okamoto S, et al. COL1A1-PDGFB fusion transcripts in fibrosarcomatous areas of six dermatofibrosarcomas protuberans. J Mol Diagn 2000; 2:47.
  47. Ha SY, Lee SE, Kwon MJ, et al. PDGFB rearrangement in dermatofibrosarcoma protuberans: correlation with clinicopathologic characteristics and clinical implications. Hum Pathol 2013; 44:1300.
  48. Takahira T, Oda Y, Tamiya S, et al. Microsatellite instability and p53 mutation associated with tumor progression in dermatofibrosarcoma protuberans. Hum Pathol 2004; 35:240.
  49. Hisaoka M, Okamoto S, Morimitsu Y, et al. Dermatofibrosarcoma protuberans with fibrosarcomatous areas. Molecular abnormalities of the p53 pathway in fibrosarcomatous transformation of dermatofibrosarcoma protuberans. Virchows Arch 1998; 433:323.
  50. Sasaki M, Ishida T, Horiuchi H, MacHinami R. Dermatofibrosarcoma protuberans: an analysis of proliferative activity, DNA flow cytometry and p53 overexpression with emphasis on its progression. Pathol Int 1999; 49:799.
  51. Goldblum J, Weiss S, Folpe AL. Fibrohistiocytic tumors of intermediate malignancy. In: Enzinger and Weiss's Soft Tissue Tumors, 7th ed, Elsevier, 2019. p.424.
  52. Tanaka A, Hatoko M, Tada H, et al. Dermatofibrosarcoma protuberans arising from a burn scar of the axilla. Ann Plast Surg 2004; 52:423.
  53. Green JJ, Heymann WR. Dermatofibrosarcoma protuberans occurring in a smallpox vaccination scar. J Am Acad Dermatol 2003; 48:S54.
  54. Baker PA, O'Dowd GJ, Khan IU. Dermatofibrosarcoma protuberans arising in a decorative tattoo. Sarcoma 2005; 9:37.
  55. Hirano SA, Torosky CM. Dermatofibrosarcoma protuberans arising at a Rho(D) immune globulin injection site. Cutis 2012; 90:233.
  56. Lee SJ, Mahoney MC, Shaughnessy E. Dermatofibrosarcoma protuberans of the breast: imaging features and review of the literature. AJR Am J Roentgenol 2009; 193:W64.
  57. Wang Y, Wang Y, Chen R, et al. A Rare Malignant Disease, Dermatofibrosarcoma Protuberans of the Breast: A Retrospective Analysis and Review of Literature. Biomed Res Int 2020; 2020:8852182.
  58. Llombart B, Serra-Guillén C, Rubio L, et al. Subcutaneous dermatofibrosarcoma protuberans, a rare subtype with predilection for the head: A retrospective series of 18 cases. J Am Acad Dermatol 2017; 77:503.
  59. Mentzel T, Beham A, Katenkamp D, et al. Fibrosarcomatous ("high-grade") dermatofibrosarcoma protuberans: clinicopathologic and immunohistochemical study of a series of 41 cases with emphasis on prognostic significance. Am J Surg Pathol 1998; 22:576.
  60. Parlette LE, Smith CK, Germain LM, et al. Accelerated growth of dermatofibrosarcoma protuberans during pregnancy. J Am Acad Dermatol 1999; 41:778.
  61. Har-Shai Y, Govrin-Yehudain J, Ullmann Y, et al. Dermatofibrosarcoma protuberans appearing during pregnancy. Ann Plast Surg 1993; 31:91.
  62. Mavili ME, Gursu KG, Gokoz A. Dermatofibrosarcoma with lymph node involvement. Ann Plast Surg 1994; 32:438.
  63. Lemm D, Mügge LO, Mentzel T, Höffken K. Current treatment options in dermatofibrosarcoma protuberans. J Cancer Res Clin Oncol 2009; 135:653.
  64. Bernard J, Poulalhon N, Argenziano G, et al. Dermoscopy of dermatofibrosarcoma protuberans: a study of 15 cases. Br J Dermatol 2013; 169:85.
  65. Domanski HA, Gustafson P. Cytologic features of primary, recurrent, and metastatic dermatofibrosarcoma protuberans. Cancer 2002; 96:351.
  66. Klijanienko J, Caillaud JM, Lagacé R. Fine-needle aspiration of primary and recurrent dermatofibrosarcoma protuberans. Diagn Cytopathol 2004; 30:261.
  67. Haycox CL, Odland PB, Olbricht SM, Piepkorn M. Immunohistochemical characterization of dermatofibrosarcoma protuberans with practical applications for diagnosis and treatment. J Am Acad Dermatol 1997; 37:438.
  68. Reimann JD, Fletcher CD. Myxoid dermatofibrosarcoma protuberans: a rare variant analyzed in a series of 23 cases. Am J Surg Pathol 2007; 31:1371.
  69. Frierson HF, Cooper PH. Myxoid variant of dermatofibrosarcoma protuberans. Am J Surg Pathol 1983; 7:445.
  70. Mentzel T, Schärer L, Kazakov DV, Michal M. Myxoid dermatofibrosarcoma protuberans: clinicopathologic, immunohistochemical, and molecular analysis of eight cases. Am J Dermatopathol 2007; 29:443.
  71. BEDNAR B. Storiform neurofibromas of the skin, pigmented and nonpigmented. Cancer 1957; 10:368.
  72. Gayner SM, Lewis JE, McCaffrey TV. Effect of resection margins on dermatofibrosarcoma protuberans of the head and neck. Arch Otolaryngol Head Neck Surg 1997; 123:430.
  73. Stojadinovic A, Karpoff HM, Antonescu CR, et al. Dermatofibrosarcoma protuberans of the head and neck. Ann Surg Oncol 2000; 7:696.
  74. Goldblum JR, Reith JD, Weiss SW. Sarcomas arising in dermatofibrosarcoma protuberans: a reappraisal of biologic behavior in eighteen cases treated by wide local excision with extended clinical follow up. Am J Surg Pathol 2000; 24:1125.
  75. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology. Available at: https://www.nccn.org/professionals/physician_gls/pdf/gist.pdf (Accessed on July 25, 2023).
  76. Abenoza P, Lillemoe T. CD34 and factor XIIIa in the differential diagnosis of dermatofibroma and dermatofibrosarcoma protuberans. Am J Dermatopathol 1993; 15:429.
  77. Kamino H, Reddy VB, Gero M, Greco MA. Dermatomyofibroma. A benign cutaneous, plaque-like proliferation of fibroblasts and myofibroblasts in young adults. J Cutan Pathol 1992; 19:85.
  78. Zelger B, Sidoroff A, Stanzl U, et al. Deep penetrating dermatofibroma versus dermatofibrosarcoma protuberans. A clinicopathologic comparison. Am J Surg Pathol 1994; 18:677.
  79. Sato N, Kimura K, Tomita Y. Recurrent dermatofibrosarcoma protuberans with myxoid and fibrosarcomatous changes paralleled by loss of CD34 expression. J Dermatol 1995; 22:665.
  80. Calikoglu E, Augsburger E, Chavaz P, et al. CD44 and hyaluronate in the differential diagnosis of dermatofibroma and dermatofibrosarcoma protuberans. J Cutan Pathol 2003; 30:185.
  81. Sellheyer K, Nelson P, Krahl D. Dermatofibrosarcoma protuberans: a tumour of nestin-positive cutaneous mesenchymal stem cells? Br J Dermatol 2009; 161:1317.
  82. Lisovsky M, Hoang MP, Dresser KA, et al. Apolipoprotein D in CD34-positive and CD34-negative cutaneous neoplasms: a useful marker in differentiating superficial acral fibromyxoma from dermatofibrosarcoma protuberans. Mod Pathol 2008; 21:31.
  83. Yan X, Takahara M, Xie L, et al. Cathepsin K expression: a useful marker for the differential diagnosis of dermatofibroma and dermatofibrosarcoma protuberans. Histopathology 2010; 57:486.
  84. Allan AE, Tsou HC, Harrington A, et al. Clonal origin of dermatofibrosarcoma protuberans. J Invest Dermatol 1993; 100:99.
  85. Dominguez-Malagón HR, Ordóñez NG, Mackay B. Dermatofibrosarcoma protuberans: ultrastructural and immunocytochemical observations. Ultrastruct Pathol 1995; 19:281.
  86. Aiba S, Tabata N, Ishii H, et al. Dermatofibrosarcoma protuberans is a unique fibrohistiocytic tumour expressing CD34. Br J Dermatol 1992; 127:79.
  87. Nickoloff BJ. The human progenitor cell antigen (CD34) is localized on endothelial cells, dermal dendritic cells, and perifollicular cells in formalin-fixed normal skin, and on proliferating endothelial cells and stromal spindle-shaped cells in Kaposi's sarcoma. Arch Dermatol 1991; 127:523.
  88. Kerob D, Pedeutour F, Leboeuf C, et al. Value of cytogenetic analysis in the treatment of dermatofibrosarcoma protuberans. J Clin Oncol 2008; 26:1757.
  89. Italiano A, Di Mauro I, Rapp J, et al. Clinical effect of molecular methods in sarcoma diagnosis (GENSARC): a prospective, multicentre, observational study. Lancet Oncol 2016; 17:532.
  90. Young CR 3rd, Albertini MJ. Atrophic dermatofibrosarcoma protuberans: case report, review, and proposed molecular mechanisms. J Am Acad Dermatol 2003; 49:761.
  91. Fujimoto M, Kikuchi K, Okochi H, Furue M. Atrophic dermatofibrosarcoma protuberans: a case report and review of the literature. Dermatology 1998; 196:422.
  92. Enneking WF, Spanier SS, Goodman MA. Current concepts review. The surgical staging of musculoskeletal sarcoma. J Bone Joint Surg Am 1980; 62:1027.
  93. Yoon SS, Maki RG, Asare EA, et al. Soft tissue sarcoma of the trunk and extremities. In: AJCC Cancer Staging Manual, 8th ed, Amin MB (Ed), AJCC, Chicago 2017. p.507.
  94. O'Sullivan B, Maki R, Agulnik M, et al. Soft tissue sarcoma of the head and neck. In: AJCC Cancer Staging Manual, 8th ed, Amin MB (Ed), American Joint Committee on Cancer, Chicago 2017. p.504.
  95. Ugurel S, Kortmann RD, Mohr P, et al. Short German guidelines: dermatofibrosarcoma protuberans. J Dtsch Dermatol Ges 2008; 6 Suppl 1:S17.
  96. Torreggiani WC, Al-Ismail K, Munk PL, et al. Dermatofibrosarcoma protuberans: MR imaging features. AJR Am J Roentgenol 2002; 178:989.
  97. Serra-Guillén C, Sanmartín O, Llombart B, et al. Correlation between preoperative magnetic resonance imaging and surgical margins with modified Mohs for dermatofibrosarcoma protuberans. Dermatol Surg 2011; 37:1638.
  98. Saiag P, Grob JJ, Lebbe C, et al. Diagnosis and treatment of dermatofibrosarcoma protuberans. European consensus-based interdisciplinary guideline. Eur J Cancer 2015; 51:2604.
Topic 7747 Version 34.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟