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

Skin nodules in newborns and infants

Skin nodules in newborns and infants
Literature review current through: Jan 2024.
This topic last updated: Oct 31, 2022.

INTRODUCTION — A variety of cutaneous and subcutaneous nodules may be detected in the newborn. Most are benign and self-limited, such as cysts, subcutaneous fat necrosis, and certain soft tissue tumors. However, some may be locally aggressive or malignant, or can be a marker for other abnormalities, such as neural tube defects [1].

Cysts and tumors that present at birth or in early infancy are reviewed here. Other benign skin lesions, vesiculopustular and bullous disorders, and vascular and pigmented lesions in the newborn are discussed separately.

(See "Skin lesions in the newborn and infant".)

(See "Vesicular, pustular, and bullous lesions in the newborn and infant".)

(See "Vascular lesions in the newborn".)

(See "Congenital melanocytic nevi".)

(See "Subcutaneous fat necrosis of the newborn".)

CYSTS — Common types of cysts include dermoid cysts and epidermoid cysts. Other types, such as branchial cleft or thyroglossal duct cysts, may also occur. (See "Cutaneous developmental anomalies in the newborn and infant".)

Dermoid cysts and sinuses — Dermoid cysts are congenital subcutaneous lesions that are usually distributed along embryonic fusion lines of the facial processes or within the neural axis [2]. The most common locations are overlying the anterior fontanelle, the bregma (junction of the coronal and sagittal sutures), upper lateral region of the forehead, lateral upper eyelid (picture 1), and submental region, although lesions can occur anywhere on the scalp, face, spinal axis, or other body sites [3].

Dermoid cysts are almost always present at birth, although subtle lesions may be noticed later, for example, when trauma causes inflammation (picture 2). They present as slow-growing, asymptomatic, rubbery subcutaneous nodules 1 to 4 cm in diameter that are usually solitary. They are nonpulsatile, noncompressible, nontender masses that appear skin colored or bluish [2]. The skin overlying the cyst appears normal unless a pit or a sinus is present [3].

The characteristic histologic findings include a lining of epidermis and evidence of adnexal structures, such as hair follicles, sebaceous glands, and eccrine and apocrine sweat apparatus. Foci of keratinization and, sometimes, mesodermal skin components may be present. These features distinguish dermoid cysts from epidermoid cysts. The latter typically appear later in life on the trunk and extremities. They generally are intradermal and do not uniformly contain hair follicles and sebaceous glands.

Dermal sinus tracts may connect dermoid cysts to the skin surface or to underlying structures, such as the bone, central nervous system (CNS), or paranasal sinuses. Cutaneous sinuses are 1 to 5 mm in length and may have a small tuft of hair protruding from the orifice [2]. Connections to the CNS are most frequent with midline or nasal dermoid cysts, occurring in approximately 25 percent of cases. There are rare reports of lateral dermoid cysts with intracranial connections [4,5]. A pit or sinus is associated with approximately one-half of nasal dermoid cysts. Midline lesions, most commonly in the lumbosacral region, are often markers of spinal dysraphism, including a tethered cord [6,7].

Sinus tracts to the skin are usually detected when they become infected and drain purulent material. If the sinus or cyst communicates with the CNS, cerebrospinal fluid may drain from the lesion. This connection also allows entry of bacteria and may be detected in a patient presenting with meningitis caused by unusual organisms [8].

Patients with midline lesions should be evaluated with neuroimaging to detect CNS connections or underlying spinal dysraphism [3]. Patients with findings on neuroimaging or other clinical abnormalities should be referred to a neurosurgeon.

Dermoid cysts should be surgically excised because of the risk of associated complications; the timing of excision depends upon the site and any associated findings. The most common complication is infection, including meningitis in lesions that connect to the CNS. In addition, the cysts may adhere to the periosteum or erode underlying skull [3,9-12]. Nasal dermoids typically enlarge, resulting in damage to the nasal bones. Malignant degeneration can occur but is rare.

Thyroglossal duct cysts — Thyroglossal duct cysts are the most common form of congenital neck mass. They represent epithelial remnants of the thyroglossal tract and present characteristically as a midline mass at the level of the thyrohyoid membrane, closely associated with the hyoid bone (picture 3). Affected patients should be referred for surgical excision of the cyst if there is infection or persistent drainage. If the size of the lesion and thyroid function are not problems, they can be followed clinically. (See "Cutaneous developmental anomalies in the newborn and infant", section on 'Thyroglossal duct cysts' and "Thyroglossal duct cyst, thyroglossal duct cyst cancer, and ectopic thyroid".)

Branchial cleft cysts — Branchial cleft cysts usually arise from the first and second branchial arches. They are commonly located anterior to the sternocleidomastoid muscle on the lower third of the lateral neck (picture 4). The cysts often contain lymphoid tissue. Third branchial cleft cysts are found at the head of the clavicles. (See "Differential diagnosis of a neck mass", section on 'Branchial cleft cyst'.)

Epidermoid cysts — Epidermoid cysts, also called epidermal inclusion cysts or epidermal cysts, result when epidermal cells located within the dermis proliferate and do not communicate with the surface. They can arise from occluded pilosebaceous follicles, entrapment of epidermal cells along embryonic lines of closures, or traumatic displacement of epidermal cells into the dermis.

Epidermoid cysts usually develop after puberty, although some present in the newborn period [13]. Examples in newborns include milia, median raphe cysts, and midline anterior neck inclusion cysts (MANIC) [14]. Their histologic appearance may be similar to dermoid cysts, or they may contain stratified squamous epithelium.

SUBCUTANEOUS FAT NECROSIS — Subcutaneous fat necrosis of the newborn (SCFN) is a rare, self-limited panniculitis that affects newborns in the first few weeks of life [15]. It typically affects term or post-term newborns and usually follows perinatal complications, such as birth asphyxia, hypothermia, meconium aspiration syndrome, newborn failure to thrive, forceps delivery, and maternal high blood pressure and/or diabetes [16-18].

Clinical features of SCFN include multiple firm, nontender subcutaneous nodules or large plaques that appear one to four weeks after birth. Commonly affected sites are the cheeks, buttocks, back, and limbs; the overlying skin may be erythematous (picture 5A-B).

SCFN should be differentiated from sclerema neonatorum, which also occurs during the first few months after birth, usually in preterm newborns. It is characterized by the presence of diffuse, waxlike hardening of the skin and subcutaneous adipose tissue, which are bound to the underlying muscle and bone, resulting in respiratory and feeding difficulties and flexion contractures (table 1) [19]. Sclerema neonatorum has become exceedingly rare, likely due to advances in neonatal intensive care [20].

SCFN and sclerema neonatorum are discussed in detail separately. (See "Subcutaneous fat necrosis of the newborn" and "Sclerema neonatorum".)

BENIGN TUMORS — Most soft tissue tumors in newborns and infants are benign [21]. These include hemangiomas and other vascular tumors, fibromatoses, non-Langerhans cell histiocytoses, and cutaneous mastocytosis.

Vascular tumors — Vascular tumors occurring in neonates and infants, including infantile hemangiomas, congenital hemangiomas, tufted angiomas and kaposiform hemangioendotheliomas, and pyogenic granulomas, are discussed separately.

(See "Infantile hemangiomas: Epidemiology, pathogenesis, clinical features, and complications".)

(See "Congenital hemangiomas: Rapidly involuting congenital hemangioma (RICH), noninvoluting congenital hemangioma (NICH), and partially involuting congenital hemangioma (PICH)".)

(See "Tufted angioma, kaposiform hemangioendothelioma (KHE), and Kasabach-Merritt phenomenon (KMP)".)

(See "Pyogenic granuloma (lobular capillary hemangioma)".)

Infantile myofibromatosis — Infantile myofibromatosis, formerly known as congenital fibromatosis, is the most common fibrous tumor of infancy [22]. In three series, approximately 60 percent of cases were congenital, and 90 percent were detected during the first year of life [23-25]. Infantile myofibromas occur more commonly in boys (60 percent) [25]. In most cases, these tumors are sporadic, but rare familial cases have been described. Both autosomal recessive and autosomal dominant inheritance patterns have been reported [26-28].

Variants in the PDGFRB gene, which encodes the platelet-derived growth factor receptor-beta, have been identified as the cause of most cases of autosomal dominant infantile myofibromatosis [29-31]. Penetrance may be variable [32]. Variants in NOTCH3, the human homolog of the Drosophila melanogaster type I membrane protein Notch3, and in SRF (serum response factor) have been reported, suggesting genetic heterogeneity [33,34]. PDGFRB variants have also been identified in cases of sporadic infantile myofibromatosis [35,36].

Clinical presentation — Infantile myofibromatosis occurs in three forms, solitary (the more frequent type), multicentric, and generalized. In the solitary form, a single lesion affecting the skin or muscle is present, most commonly on the head, neck, or trunk (picture 6A-B). Multiple lesions are present in the multicentric form, which is more likely to be congenital and may affect muscles and bones as well as skin and subcutaneous tissues (picture 7) [24,25,37]. The generalized form is characterized by visceral involvement in addition to skin lesions and is associated with a poor prognosis. The most common sites of visceral involvement are lung, bone, gastrointestinal tract, heart, and liver [38]. PDGFR variants associated with infantile myofibromas may also predispose to cerebral aneurysms [35,39].

The superficial lesions are rubbery and firm, rounded nodules and may be mobile [40]. The deeper lesions are nonmobile. They range in diameter from 1 to 7 cm [24]. In some cases, the overlying skin has a red or purple discoloration that may mimic a hemangioma [40] (see "Infantile hemangiomas: Epidemiology, pathogenesis, clinical features, and complications"). The lesions may also be sclerodermoid [25]. Reticulate vascular patches with underlying atrophy have rarely been described and are more common in the multicentric form [25,41]. Skin lesions associated with the generalized form vary; rare cases of atrophic papules and widespread, hypopigmented macules have been reported [42].

Diagnosis — The diagnosis of infantile myofibromatosis is usually made on the basis of clinical features and characteristic histopathology. The histologic appearance is similar in solitary and multiple cutaneous and visceral lesions [43]. There are well-circumscribed masses of spindle-shaped cells staining as fibroblasts or smooth muscle cells, bland necrosis with stippled calcification, and plentiful thin-walled vessels. Atypia is uniformly absent.

Additional evaluation to exclude visceral involvement should be considered. While there are no established guidelines for staging or surveillance, some authors recommend screening all patients (even those with apparent solitary disease) for visceral involvement [31,44,45]. Although radiograph/bone scans, ultrasound, computed tomography (CT), fluorodeoxyglucose-positron emission tomography (FDG-PET), and magnetic resonance imaging (MRI) have all been used effectively to screen for systemic disease [25,45-47], whole-body MRI has gained popularity due to its superior soft tissue resolution [48].

One group suggests a dedicated MRI to delineate the clinically apparent lesion and a rapid whole-body MRI at diagnosis followed by imaging surveillance every three to six months for two years [45]. Tumor tissue should be sequenced for PDGFRB variants, and additional genetic evaluation may be indicated [31]. All patients should be referred for genetic counseling [31]. This has implications for not only the patient but also for family members, as first-degree relatives of patients diagnosed with infantile myofibromatosis or PDGFRB germline mutations should also undergo surveillance [31]. Due to the association of cerebral aneurysms with PDGFRB mutations, a screening brain MRI at age 15 to 18 years can be considered for patients with PDGFRB mutations.

Treatment and prognosis — Solitary lesions are often surgically excised. However, close observation of these tumors may be appropriate in cases without visceral involvement, as lesions in most cases spontaneously involute in one to two years, occasionally leaving residual calcifications or atrophic scars [24,25,37,40].

Patients with visceral involvement must be followed closely due to the higher risk of morbidity and mortality, although in many cases, a strategy of observation may be appropriate. Progressive visceral myofibromas or tumors that affect function or cause local complications due to mass effect are surgically excised or treated with chemotherapy [38,48-50]. Successful targeted therapy with PDGFR-beta tyrosine kinase inhibitors has been reported [39,51-54].

Recurrence of infantile myofibromatosis is unusual but can be delayed into adulthood [32]. Therefore, long-term follow-up is indicated for patients with the multicentric or generalized forms of myofibromatosis [45]. Recurrences can be managed by re-excision or treated with chemotherapy [55].

Though the prognosis is generally excellent, infantile myofibromatosis can rarely be progressive and fatal. Poor outcomes are greatest among infants with the generalized form with extensive visceral involvement (often due to cardiopulmonary or gastrointestinal complications), though rare reports of fatalities occurring in patients with solitary disease also exist [44].

Plaque-like myofibroblastic tumor — Plaque-like myofibroblastic tumor, first described in 2007, is a rare, benign, soft tissue tumor presenting in infants and young children as a firm, irregularly shaped dermal plaque typically located on the middle or lower back, although they can develop in other areas, such as the hips or thighs (picture 8) [56-59]. Lesions are 2 to 8 cm in diameter and reddish to brown in color. The diagnosis is based on the clinical presentation and a skin biopsy. Histologically, the tumor resembles a dermatofibroma, with a relatively well-circumscribed proliferation of spindle cells arranged in short fascicles with thin collagen bundles extending through the entire reticular dermis [56]. On immunohistochemistry, these lesions stain for factor XIIIa and smooth muscle actin, but are CD34-negative, consistent with a myofibroblastic lineage.

The differential diagnosis of plaque-like myofibroblastic tumor includes other dermal tumors, including clustered dermatofibroma, fibrous connective tissue nevus, dermatomyofibroma, and dermatofibroma protuberans (table 2) [60,61]. (See "Skin lesions in the newborn and infant", section on 'Connective tissue nevus' and "Dermatofibrosarcoma protuberans: Epidemiology, pathogenesis, clinical presentation, diagnosis, and staging" and "Overview of benign lesions of the skin".)

Treatment is surgical excision. Local recurrence has been reported [56,62].

Infantile digital fibromatosis — Infantile digital fibroma, also called "recurrent digital fibrous tumor of childhood," is a rare, benign childhood tumor that occurs almost exclusively on the digits [63,64]. It usually develops in the first year of life and typically presents as a firm, reddish-pink or skin-colored, asymptomatic nodule 3 to 35 mm in size located on the dorsolateral aspects of the fingers and toes, characteristically sparing the thumbs and great toes (picture 9) [63]. Tumors may be multiple (picture 10) and, if large, may cause deformity and functional impairment.

The diagnosis is based upon the clinical appearance and the histologic finding of a dermal nonencapsulated proliferation of spindle-shaped myofibroblasts intertwined with coarse collagen fibers, with perpendicular cell fascicles reaching the epidermis. The presence of eosinophilic intracytoplasmic inclusions that stain red with Masson's trichrome is pathognomonic. On immunohistochemistry, tumors consistently express alpha-smooth muscle actin, calponin, and desmin [63].

Because lesions may regress spontaneously over several months to years, observation is a reasonable first-line management strategy [65,66]. Lesions causing functional impairment may require surgical excision to prevent joint deformities. However, rapid recurrence after surgical excision is common [63]. Mohs micrographic surgery has been successfully used in a few cases [67,68].

Other juvenile fibromatoses — Other rare fibromatoses that occur in infancy include infantile desmoid-type fibromatosis, fibromatosis colli (congenital torticollis), fibrous hamartoma of infancy, lipofibromatosis, precalcaneal congenital fibrolipomatous hamartoma, gingival fibromatosis, juvenile hyaline fibromatosis, and infantile systemic fibromatosis [69-74]. (See "Congenital muscular torticollis: Clinical features and diagnosis" and "Soft tissue lesions of the oral cavity in children", section on 'Gingival overgrowth'.)

Juvenile xanthogranuloma — Juvenile xanthogranuloma (JXG) is a benign proliferative disorder of histiocytic cells, a non-Langerhans cell histiocytosis. It typically presents within the first two years of life and, though less common, may be congenital [75]. The most common presentation of JXG is a firm, well-circumscribed, yellow-red tumor that increases in size over the first few months of life (picture 11). Less common presentations include exophytic tumors, agminated lesions over an erythematous patch or plaque, infiltrative plaques, subcutaneous masses, or multiple lesions (picture 12). In a review of 31 congenital cases, systemic involvement occurred in 16, the majority of which had multiple cutaneous lesions [75]. The most common extracutaneous sites of involvement are the eyes, followed by the liver, bone marrow, and lung [75]. JXG is relatively common among children with neurofibromatosis type I [76] and may also be associated with Noonan syndrome [77], a related RASopathy.

The clinical presentation, diagnosis, and management of JXG are discussed in greater detail separately. (See "Juvenile xanthogranuloma (JXG)".)

Cutaneous mastocytosis — Cutaneous mastocytomas are rare lesions composed of mast cells that occur in childhood (picture 13A-B). They may be solitary or multiple. Manipulation of the lesion by rubbing or pressure may trigger mast cell mediator release with resultant flushing and, occasionally, hypotension (Darier's sign). In most cases, the caretakers have noticed this phenomenon and can describe it, without the need to disturb the lesion and induce symptoms. Cutaneous mastocytomas typically regress spontaneously by adolescence. Cutaneous and systemic mastocytosis are discussed separately. (See "Mastocytosis (cutaneous and systemic) in children: Epidemiology, clinical manifestations, evaluation, and diagnosis".)

MALIGNANT TUMORS — Malignant soft tissue tumors that present at birth or in the neonatal period are rare and over 50 percent present in the skin or subcutaneous tissue [78]. Examples include rhabdomyosarcoma, infantile fibrosarcoma (IF), neuroblastoma, and congenital leukemia [79]. IFs and malignant rhabdoid tumors appear most commonly in the first year of life [78].

Rhabdomyosarcoma — Rhabdomyosarcoma is a highly malignant skeletal muscle neoplasm that arises from embryonal mesenchyme. It is the most common soft tissue sarcoma in infants, accounting for one-third of all cases [80]. The primary cutaneous form typically presents as a single firm, subcutaneous nodule that adheres to the skin above and tissue underneath [40]. The overlying skin may be erythematous or shiny, occasionally mimicking a deep hemangioma [81]. An extremely rare neonatal presentation is alveolar rhabdomyosarcoma, a very aggressive tumor that frequently includes multiple skin and subcutaneous metastases [82,83]. (See "Rhabdomyosarcoma in childhood and adolescence: Clinical presentation, diagnostic evaluation, and staging".)

Infantile fibrosarcoma — Infantile fibrosarcoma (IF), also called congenital infantile fibrosarcoma, is a rapidly growing, poorly circumscribed spindle cell tumor of the soft tissues that may present at birth or appear subsequently (picture 14). Approximately 75 percent of patients with IF are diagnosed at less than one year of age [84]. Forty percent of cases are discovered at birth [85]. The tumor typically involves the distal extremities, followed by the trunk, although the head and neck region is affected more frequently in infants than older children [21,84,86]. It is the second most common type of soft tissue sarcoma in infants and the most common occurring on the extremities [80].

Histologically, IF is characterized by spindle cells arranged in interlacing fascicles with a high mitotic rate and positive staining for vimentin. The majority of IFs have a recurrent translocation t(12;15)(p13;q25) that results in a fusion gene involving the neurotrophic tyrosine receptor kinase-3 (NTRK3) gene, called ETV6-NTRK3 (also known as Tel-TRKC) [87-92]. This structural rearrangement results in a tropomyosin receptor kinase (TRK) fusion oncoprotein that drives uninterrupted downstream signaling messages and oncogenic transformation and tumor growth [93-95]. TRK fusions involving the NTRK1, NTRK2, and NTRK3 genes have been identified in a variety of other pediatric mesenchymal tumors but at a lower frequency than in IF [96,97]. (See "TRK fusion-positive cancers and TRK inhibitor therapy", section on 'Prevalence' and "TRK fusion-positive cancers and TRK inhibitor therapy", section on 'Basic biology'.)

Immunohistochemical methods are available to identify TRK fusions in IF and can help to differentiate IF from other pediatric fibromatous tumors [98-100]. (See "TRK fusion-positive cancers and TRK inhibitor therapy", section on 'Testing algorithms'.)

The accurate and timely identification of TRK fusions is of immediate therapeutic importance, given that TRK inhibitions have been shown to have robust and durable activity in TRK fusion-positive solid tumors, such as IF. Larotrectinib, an oral small molecule TRK inhibitor, is approved in the United States and elsewhere for adults and children (ages 28 days to 82 years) with solid tumors that have an NTRK gene fusion without a known acquired resistance mutation, that are metastatic or where surgical resection is likely to result in severe morbidity, and that have no satisfactory alternative treatments or have progressed following treatment. (See "TRK fusion-positive cancers and TRK inhibitor therapy", section on 'Efficacy of first-generation TRK inhibitors'.)

Although locally infiltrative, IF rarely metastasizes and generally has a good prognosis; more than 80 percent are cured [101-103]. However, bleeding and coagulopathy may be significant complications, resulting in high mortality [104]. Although surgical resection is the mainstay of treatment, the surgical approach has evolved over the years from the primary treatment modality to a component of a multidisciplinary approach and from mutilating operations, such as amputation, to more conservative tissue-sparing procedures utilizing initial (neoadjuvant) chemotherapy [84,90,105].

Limited data suggest promise for neoadjuvant use of larotrectinib in pediatric sarcomas, such as IF [106-108]. In a phase 1 trial of larotrectinib, all five pediatric patients with locally advanced, documented TRK fusion sarcomas (three with IF) treated with preoperative larotrectinib were able to undergo successful curative-intent surgical resection, with three achieving complete or near-complete (>98 percent treatment effect) pathologic response [106]. These three patients remained off larotrectinib between 7 and 15 months postoperatively. The remaining two patients who had viable tumors at the time of surgical resection and positive resection margins continued to receive adjuvant larotrectinib, although the duration and long-term outcomes were not reported. All four patients with initially inoperable limb sarcomas were able to undergo limb-sparing surgery. Adverse effects of larotrectinib include low grade increases in liver enzyme levels, decrease in neutrophil count, anemia, and vomiting [107].

While small case series such as this demonstrate the potential promise of neoadjuvant therapy with a TRK inhibitor, the main obstacle to broader use of this approach has been timely, preoperative identification of the presence of an NTRK gene or TRK oncogene fusion. (See "TRK fusion-positive cancers and TRK inhibitor therapy", section on 'Testing algorithms'.)

Other soft tissue sarcomas — Other soft tissue sarcomas occurring in the first year of life include malignant rhabdoid tumors, hemangiopericytoma, the Ewing sarcoma family of tumors, dermatofibrosarcoma protuberans, malignant fibrous histiocytoma, and malignant peripheral nerve sheath tumor [79,80]. (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma" and "Dermatofibrosarcoma protuberans: Epidemiology, pathogenesis, clinical presentation, diagnosis, and staging".)

Neuroblastoma — Neuroblastoma, the most common malignancy identified at birth, typically presents with an abdominal mass. Metastatic cutaneous lesions are present in approximately one-third of children with congenital neuroblastoma [40]. In one review of 208 neonates with cutaneous metastases, 17 percent were neuroblastoma metastases [109].

Lesions appear as firm, nontender, bluish-red nodules and papules distributed over the entire body ("blueberry muffin baby") and have a distinctive response to rubbing, characterized by central blanching with a surrounding halo of erythema that persists for 30 to 60 minutes [40,110]. The mechanism is thought to be vasoconstriction induced by catecholamine release [111]. (See "Epidemiology, pathogenesis, and pathology of neuroblastoma" and "Clinical presentation, diagnosis, and staging evaluation of neuroblastoma".)

Congenital leukemia — Congenital leukemia is an extremely rare disorder that typically presents at birth with hepatosplenomegaly, lymphadenopathy, central nervous system involvement, petechiae, and ecchymoses [112]. Many affected patients have red/brown or violaceous nodules caused by infiltration of the skin by leukemic cells and known as leukemia cutis (picture 15) [112,113]. The differential diagnosis of this so-called "blueberry muffin baby" appearance includes intrauterine infections, hemolytic diseases, and other tumors.

Rarely, affected infants present with a vesiculopustular eruption [114]. The diagnosis is confirmed by skin biopsy. The prognosis depends on the presence of the associated systemic leukemia [112,115]. Patients with aleukemic leukemia cutis can be managed conservatively.

Congenital self-healing reticulohistiocytosis — Congenital self-healing reticulohistiocytosis (CSHR; Hashimoto-Pritzker disease) is a single-system variant of Langerhans cell histiocytosis with involvement limited to the skin that in most cases regresses spontaneously in weeks to months of onset. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

CSHR is an uncommon disease. However, because of the high rate of spontaneous resolution and lack of clinical recognition, its incidence is likely underestimated [116]. Somatic mutations in various genes have been reported, including multiple in the mitogen-activated protein (MAP) kinase cell-signaling pathway (BRAF-V6000E, HRAS) and in PTEN [117]. Circulating mononuclear cells with the BRAF-V600E mutation are infrequently found in patients with CSHR, whereas they can be identified in the majority of patients with skin and multisystem Langerhans cell histiocytosis [118].

CSHR presents at birth or in the newborn period as solitary (25 percent of cases) or multiple cutaneous nodules that may be located anywhere on the infant (picture 16A-B) [119]. The uninodular form has also been termed "solitary Langerhans cell histiocytoma" (picture 17) [117]. The nodules often centrally ulcerate either before or shortly after birth (picture 18). Vesicles and bullae also have been reported [120]. All lesions heal within a few months, sometimes leaving an atrophic, white scar.

The diagnosis of CSHR is based upon the histopathologic evaluation of a skin biopsy. Histology shows a proliferation of mononuclear cells resembling Langerhans cells with characteristic, abundant eosinophilic cytoplasm and a lobulated, kidney-shaped nucleus. Immunostaining is positive for CD1a, CD4, CD207, and S-100 protein. Staining for E-cadherin, Ki-67, and phosphorylated histone H3 is not helpful in differentiating CSHR from disseminated Langerhans cell histiocytosis with cutaneous involvement [116]. Characteristic features seen on electron microscopy are dense bodies, regularly laminated bodies, and Birbeck granules.

Histopathologic and immunophenotypic features distinguish CSHR from other nodular neonatal lesions. In vesicular or bullous cases, congenital varicella and neonatal herpes simplex virus (HSV) must be excluded. (See "Varicella-zoster infection in the newborn" and "Neonatal herpes simplex virus infection: Clinical features and diagnosis".)

Infants with CSHR should be evaluated for extracutaneous involvement, including a complete blood count, coagulation studies, serum chemistries, liver function tests, urine osmolality, chest and bone radiographs, abdominal ultrasound, and bone marrow biopsy if necessary [121]. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

No treatment is required for CSHR. However, close clinical and laboratory monitoring is important because of the possibility of recurrence or progression [122]. A review of 126 reported cases of CSHR found a recurrence rate of 10 percent (all within the first year of life) and a mortality rate of 3 percent [123]. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

SUMMARY

Cysts – Common types of cysts include dermoid cysts, thyroglossal duct cysts, branchial cleft cysts, and epidermoid cysts. Dermoid cysts are small (1 to 4 cm in diameter), usually solitary, slow-growing, asymptomatic, rubbery, nontender masses that appear skin-colored or blue; the skin overlying the cyst appears normal unless a pit or a sinus is present (picture 1). They are commonly located over the anterior fontanelle, junction of the coronal and sagittal sutures, upper lateral region of the forehead, lateral upper eyelid, and submental region. (See 'Cysts' above.)

Subcutaneous fat necrosis – Subcutaneous fat necrosis (SCFN) is a rare condition that typically affects term or post-term newborns in the first few weeks of life, usually following perinatal complications. It is characterized by multiple firm, nontender subcutaneous nodules or large plaques over bony prominences (eg, cheeks, buttocks, back, limbs) (picture 5A). SCFN is discussed in detail separately. (See "Subcutaneous fat necrosis of the newborn".)

Benign tumors – Most soft tissue tumors in newborns and infants are benign. These include hemangiomas and other vascular tumors, fibromatoses, non-Langerhans cell histiocytoses, and cutaneous mastocytosis (see 'Benign tumors' above):

Vascular tumors – Vascular tumors occurring in neonates and infants, including infantile hemangiomas, congenital hemangiomas, tufted angiomas and kaposiform hemangioendotheliomas, and pyogenic granulomas, are discussed separately. (See "Infantile hemangiomas: Epidemiology, pathogenesis, clinical features, and complications" and "Congenital hemangiomas: Rapidly involuting congenital hemangioma (RICH), noninvoluting congenital hemangioma (NICH), and partially involuting congenital hemangioma (PICH)" and "Tufted angioma, kaposiform hemangioendothelioma (KHE), and Kasabach-Merritt phenomenon (KMP)" and "Pyogenic granuloma (lobular capillary hemangioma)".)

Infantile myofibromatosis – Infantile myofibromatosis, also known as congenital fibromatosis, is the most common fibrous tumor of infancy. It presents in most cases as a solitary lesion on the head, neck, or trunk (picture 6A-B), but can be multicentric or generalized. The diagnosis is based on a lesion biopsy. (See 'Infantile myofibromatosis' above.)

Malignant tumors – Malignant tumors affecting the skin and soft tissues of newborns and infants include infantile fibrosarcoma, rhabdomyosarcoma, neuroblastoma, congenital leukemia, and congenital self-healing reticulohistiocytosis (see 'Malignant tumors' above):

Rhabdomyosarcoma – Rhabdomyosarcoma is a highly malignant skeletal muscle neoplasm and the most common soft tissue sarcoma in infants. The primary cutaneous form typically presents as a single firm subcutaneous nodule that adheres to the skin above and tissue underneath. (See "Rhabdomyosarcoma in childhood and adolescence: Clinical presentation, diagnostic evaluation, and staging".)

Infantile fibrosarcoma – Infantile fibrosarcoma is the second most common type of soft tissue sarcoma in infants. It presents as a rapidly growing, poorly circumscribed subcutaneous mass, most frequently located on the distal extremities. Although locally infiltrative, these tumors rarely metastasize; however, bleeding and coagulopathy may be life-threatening complications. Evaluation for tropomyosin receptor kinase (TRK) fusions, which are common in infantile fibrosarcoma, is important and can guide management. Larotrectinib, a TRK inhibitor approved for the treatment of NTRK fusion-positive solid tumors in children and adults, has shown promise as neoadjuvant and adjuvant treatment of infantile fibrosarcoma. (See 'Infantile fibrosarcoma' above and "TRK fusion-positive cancers and TRK inhibitor therapy".)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Josie A Pielop, MD, who contributed to an earlier version of this topic review.

  1. Levin LE, Kinariwalla N, Behr GG, et al. Lumps and bumps: What not to miss. Pediatr Dermatol 2022; 39:679.
  2. Drolet BA. Cutaneous signs of neural tube dysraphism. Pediatr Clin North Am 2000; 47:813.
  3. Orozco-Covarrubias L, Lara-Carpio R, Saez-De-Ocariz M, et al. Dermoid cysts: a report of 75 pediatric patients. Pediatr Dermatol 2013; 30:706.
  4. Prior A, Anania P, Pacetti M, et al. Dermoid and Epidermoid Cysts of Scalp: Case Series of 234 Consecutive Patients. World Neurosurg 2018; 120:119.
  5. Yan C, Low DW. A Rare Presentation of a Dermoid Cyst with Draining Sinus in a Child: Case Report and Literature Review. Pediatr Dermatol 2016; 33:e244.
  6. Ackerman LL, Menezes AH. Spinal congenital dermal sinuses: a 30-year experience. Pediatrics 2003; 112:641.
  7. Guggisberg D, Hadj-Rabia S, Viney C, et al. Skin markers of occult spinal dysraphism in children: a review of 54 cases. Arch Dermatol 2004; 140:1109.
  8. Douvoyiannis M, Goldman DL, Abbott IR 3rd, Litman N. Posterior fossa dermoid cyst with sinus tract and meningitis in a toddler. Pediatr Neurol 2008; 39:63.
  9. Brownstein MH, Helwig EB. Subcutaneous dermoid cysts. Arch Dermatol 1973; 107:237.
  10. Baldwin HE, Berck CM, Lynfield YL. Subcutaneous nodules of the scalp: preoperative management. J Am Acad Dermatol 1991; 25:819.
  11. Mallory SB. Subcutaneous scalp lesions in children. J Am Acad Dermatol 1993; 28:131.
  12. Moses MA, Green BC, Cugno S, et al. The management of midline frontonasal dermoids: a review of 55 cases at a tertiary referral center and a protocol for treatment. Plast Reconstr Surg 2015; 135:187.
  13. Lucky AW. Transient and benign cutaneous lesions in the newborn. In: Neonatal and Infant Dermatology, 3rd ed, Eichenfield LF, Frieden IJ, Mathes EF, Zaenglein A (Eds), Elsevier, 2015. p.65.
  14. Walsh R, North J, Cordoro KM, et al. Midline anterior neck inclusion cyst: A novel superficial congenital developmental anomaly of the neck. Pediatr Dermatol 2018; 35:55.
  15. Stefanko NS, Drolet BA. Subcutaneous fat necrosis of the newborn and associated hypercalcemia: A systematic review of the literature. Pediatr Dermatol 2019; 36:24.
  16. Mahé E, Girszyn N, Hadj-Rabia S, et al. Subcutaneous fat necrosis of the newborn: a systematic evaluation of risk factors, clinical manifestations, complications and outcome of 16 children. Br J Dermatol 2007; 156:709.
  17. Oza V, Treat J, Cook N, et al. Subcutaneous fat necrosis as a complication of whole-body cooling for birth asphyxia. Arch Dermatol 2010; 146:882.
  18. Del Pozzo-Magaña BR, Ho N. Subcutaneous Fat Necrosis of the Newborn: A 20-Year Retrospective Study. Pediatr Dermatol 2016; 33:e353.
  19. Zeb A, Darmstadt GL. Sclerema neonatorum: a review of nomenclature, clinical presentation, histological features, differential diagnoses and management. J Perinatol 2008; 28:453.
  20. Spohn GP, Pietras TA, Stone MS. Delayed-Onset Sclerema Neonatorum in a Critically Ill Premature Infant. Pediatr Dermatol 2016; 33:e168.
  21. Coffin CM, Dehner LP. Soft tissue tumors in first year of life: a report of 190 cases. Pediatr Pathol 1990; 10:509.
  22. Wiswell TE, Davis J, Cunningham BE, et al. Infantile myofibromatosis: the most common fibrous tumor of infancy. J Pediatr Surg 1988; 23:315.
  23. Coffin CM, Dehner LP. Fibroblastic-myofibroblastic tumors in children and adolescents: a clinicopathologic study of 108 examples in 103 patients. Pediatr Pathol 1991; 11:569.
  24. Chung EB, Enzinger FM. Infantile myofibromatosis. Cancer 1981; 48:1807.
  25. Mashiah J, Hadj-Rabia S, Dompmartin A, et al. Infantile myofibromatosis: a series of 28 cases. J Am Acad Dermatol 2014; 71:264.
  26. Zand DJ, Huff D, Everman D, et al. Autosomal dominant inheritance of infantile myofibromatosis. Am J Med Genet A 2004; 126A:261.
  27. Bracko M, Cindro L, Golouh R. Familial occurrence of infantile myofibromatosis. Cancer 1992; 69:1294.
  28. Smith A, Orchard D. Infantile myofibromatosis: two families supporting autosomal dominant inheritance. Australas J Dermatol 2011; 52:214.
  29. Martignetti JA, Tian L, Li D, et al. Mutations in PDGFRB cause autosomal-dominant infantile myofibromatosis. Am J Hum Genet 2013; 92:1001.
  30. Cheung YH, Gayden T, Campeau PM, et al. A recurrent PDGFRB mutation causes familial infantile myofibromatosis. Am J Hum Genet 2013; 92:996.
  31. Hettmer S, Dachy G, Seitz G, et al. Genetic testing and surveillance in infantile myofibromatosis: a report from the SIOPE Host Genome Working Group. Fam Cancer 2021; 20:327.
  32. Murray N, Hanna B, Graf N, et al. The spectrum of infantile myofibromatosis includes both non-penetrance and adult recurrence. Eur J Med Genet 2017; 60:353.
  33. Lee JW. Mutations in PDGFRB and NOTCH3 are the first genetic causes identified for autosomal dominant infantile myofibromatosis. Clin Genet 2013; 84:340.
  34. Antonescu CR, Sung YS, Zhang L, et al. Recurrent SRF-RELA Fusions Define a Novel Subset of Cellular Myofibroma/Myopericytoma: A Potential Diagnostic Pitfall With Sarcomas With Myogenic Differentiation. Am J Surg Pathol 2017; 41:677.
  35. Arts FA, Sciot R, Brichard B, et al. PDGFRB gain-of-function mutations in sporadic infantile myofibromatosis. Hum Mol Genet 2017; 26:1801.
  36. Dachy G, de Krijger RR, Fraitag S, et al. Association of PDGFRB Mutations With Pediatric Myofibroma and Myofibromatosis. JAMA Dermatol 2019; 155:946.
  37. Larralde M, Hoffner MV, Boggio P, et al. Infantile myofibromatosis: report of nine patients. Pediatr Dermatol 2010; 27:29.
  38. Wu SY, McCavit TL, Cederberg K, et al. Chemotherapy for Generalized Infantile Myofibromatosis With Visceral Involvement. J Pediatr Hematol Oncol 2015; 37:402.
  39. Wenger TL, Bly RA, Wu N, et al. Activating variants in PDGFRB result in a spectrum of disorders responsive to imatinib monotherapy. Am J Med Genet A 2020; 182:1576.
  40. Wyatt AJ, Hansen RC. Pediatric skin tumors. Pediatr Clin North Am 2000; 47:937.
  41. Zhong CS, Song H, Weiss A, et al. Myofibromatosis presenting as reticulated vascular changes and subcutaneous atrophy in a patient with somatic mosaicism of PDGFRB mutation. Br J Dermatol 2018; 179:1408.
  42. Saikaly SK, Schoch JJ, Motaparthi K, et al. Generalized infantile myofibromatosis with visceral involvement presenting as diffuse hypopigmented macules at birth. Pediatr Dermatol 2021; 38:249.
  43. Briselli MF, Soule EH, Gilchrist GS. Congenital fibromatosis: report of 18 cases of solitary and 4 cases of multiple tumors. Mayo Clin Proc 1980; 55:554.
  44. Sparber-Sauer M, Vokuhl C, Seitz G, et al. Infantile myofibromatosis: Excellent prognosis but also rare fatal progressive disease. Treatment results of five Cooperative Weichteilsarkom Studiengruppe (CWS) trials and one registry. Pediatr Blood Cancer 2022; 69:e29403.
  45. Todd K, Kim HK, Szabo S, et al. Solitary myofibroma preceding the development of multicentric myofibromatosis: A report of two cases with surveillance recommendations. Pediatr Blood Cancer 2020; 67:e28266.
  46. Tang ER, Khalatbari H, Parisi MT. Utility of 18F-FDG PET/CT in Infantile Myofibromatosis. Clin Nucl Med 2019; 44:676.
  47. Naffaa L, Khalifeh I, Salman R, et al. Infantile myofibromatosis: review of imaging findings and emphasis on correlation between MRI and histopathological findings. Clin Imaging 2019; 54:40.
  48. Manisterski M, Benish M, Levin D, et al. Diverse presentation and tailored treatment of infantile myofibromatosis: A single-center experience. Pediatr Blood Cancer 2021; 68:e28769.
  49. Levine E, Fréneaux P, Schleiermacher G, et al. Risk-adapted therapy for infantile myofibromatosis in children. Pediatr Blood Cancer 2012; 59:115.
  50. Weaver MS, Navid F, Huppmann A, et al. Vincristine and Dactinomycin in Infantile Myofibromatosis With a Review of Treatment Options. J Pediatr Hematol Oncol 2015; 37:237.
  51. Mudry P, Slaby O, Neradil J, et al. Case report: rapid and durable response to PDGFR targeted therapy in a child with refractory multiple infantile myofibromatosis and a heterozygous germline mutation of the PDGFRB gene. BMC Cancer 2017; 17:119.
  52. Pond D, Arts FA, Mendelsohn NJ, et al. A patient with germ-line gain-of-function PDGFRB p.N666H mutation and marked clinical response to imatinib. Genet Med 2018; 20:142.
  53. Sramek M, Neradil J, Macigova P, et al. Effects of Sunitinib and Other Kinase Inhibitors on Cells Harboring a PDGFRB Mutation Associated with Infantile Myofibromatosis. Int J Mol Sci 2018; 19.
  54. Weller JM, Keil VC, Gielen GH, et al. PDGRFB mutation-associated myofibromatosis: Response to targeted therapy with imatinib. Am J Med Genet A 2019; 179:1895.
  55. Ebert CS Jr, Zdanski C, Ardeshirpour F, et al. Recurrent infantile myofibromatosis: a report of conservative management and discussion of treatment strategies. Ear Nose Throat J 2008; 87:E4.
  56. Moulonguet I, Biaggi A, Eschard C, et al. Plaque-Like Myofibroblastic Tumor: Report of 4 Cases. Am J Dermatopathol 2017; 39:767.
  57. Alesini F, Soda G, Gianno F, et al. Plaque-Like Myofibroblastic Tumor of Infancy: A New Case Report and Literature Review. Pediatr Dermatol 2017; 34:176.
  58. Clarke JT, Clarke LE, Miller C, et al. Plaque-like myofibroblastic tumor of infancy. Pediatr Dermatol 2007; 24:E83.
  59. Sekar T, Mushtaq J, AlBadry W, et al. Plaque-like Myofibroblastic Tumor: 2 Cases of This Unusual Dermal Tumor Which Occurs in Infancy and Early Childhood. Pediatr Dev Pathol 2018; 21:444.
  60. Krol A. Beyond infections: tumors and malformations of the diaper area. Int J Dermatol 2016; 55 Suppl 1:14.
  61. Virdi A, Baraldi C, Barisani A, et al. Plaque-like myofibroblastic tumor, a rare entity of childhood: Possible pitfalls in differential diagnosis. J Cutan Pathol 2019; 46:389.
  62. Marqueling AL, Dasher D, Friedlander SF, et al. Plaque-like myofibroblastic tumor: report of three cases. Pediatr Dermatol 2013; 30:600.
  63. Laskin WB, Miettinen M, Fetsch JF. Infantile digital fibroma/fibromatosis: a clinicopathologic and immunohistochemical study of 69 tumors from 57 patients with long-term follow-up. Am J Surg Pathol 2009; 33:1.
  64. Taylor HO, Gellis SE, Schmidt BA, et al. Infantile digital fibromatosis. Ann Plast Surg 2008; 61:472.
  65. Paloni G, Mattei I, Salmaso R, Cutrone M. Infantile digital fibromatosis. Arch Dis Child 2013; 98:308.
  66. Niamba P, Léauté-Labrèze C, Boralevi F, et al. Further documentation of spontaneous regression of infantile digital fibromatosis. Pediatr Dermatol 2007; 24:280.
  67. Campbell LB, Petrick MG. Mohs micrographic surgery for a problematic infantile digital fibroma. Dermatol Surg 2007; 33:385.
  68. Albertini JG, Welsch MJ, Conger LA, et al. Infantile digital fibroma treated with mohs micrographic surgery. Dermatol Surg 2002; 28:959.
  69. Prendiville JS. Lumps, bumps, and hamartomas. In: Neonatal and Infant Dermatology, 3rd ed, Eichenfeild LF, Frieden IJ, Mathes EF, Zanglein A (Eds), Elsevier, 2015. p.422.
  70. Al-Ibraheemi A, Martinez A, Weiss SW, et al. Fibrous hamartoma of infancy: a clinicopathologic study of 145 cases, including 2 with sarcomatous features. Mod Pathol 2017; 30:474.
  71. Saab ST, McClain CM, Coffin CM. Fibrous hamartoma of infancy: a clinicopathologic analysis of 60 cases. Am J Surg Pathol 2014; 38:394.
  72. Keltz M, DiCostanzo D, Desai P, Cohen SR. Infantile (desmoid-type) fibromatosis. Pediatr Dermatol 1995; 12:149.
  73. Boos MD, Chikwava KR, Dormans JP, et al. Lipofibromatosis: an institutional and literature review of an uncommon entity. Pediatr Dermatol 2014; 31:298.
  74. Kalgaonkar PS, Wade M, Warke C, et al. Juvenile Hyaline Fibromatosis- A Rare Autosomal Recessive Disease. J Clin Diagn Res 2017; 11:SD04.
  75. Oza VS, Stringer T, Campbell C, et al. Congenital-type juvenile xanthogranuloma: A case series and literature review. Pediatr Dermatol 2018; 35:582.
  76. Liy-Wong C, Mohammed J, Carleton A, et al. The relationship between neurofibromatosis type 1, juvenile xanthogranuloma, and malignancy: A retrospective case-control study. J Am Acad Dermatol 2017; 76:1084.
  77. Ali MM, Gilliam AE, Ruben BS, et al. Juvenile xanthogranuloma in Noonan syndrome. Am J Med Genet A 2021; 185:3048.
  78. Liszewski W, Maguiness S, Greengard E, Boull C. The incidence of pediatric malignant soft tissue tumors of the skin and subcutaneous tissue. Pediatr Dermatol 2018; 35:e427.
  79. Mondì V, Piersigilli F, Salvatori G, Auriti C. The Skin as an Early Expression of Malignancies in the Neonatal Age: A Review of the Literature and a Case Series. Biomed Res Int 2015; 2015:809406.
  80. Sultan I, Casanova M, Al-Jumaily U, et al. Soft tissue sarcomas in the first year of life. Eur J Cancer 2010; 46:2449.
  81. Megarbane H, Doz F, Manach Y, et al. Neonatal rhabdomyosarcoma misdiagnosed as a congenital hemangioma. Pediatr Dermatol 2011; 28:299.
  82. Rodriguez-Galindo C, Hill DA, Onyekwere O, et al. Neonatal alveolar rhabdomyosarcoma with skin and brain metastases. Cancer 2001; 92:1613.
  83. Grundy R, Anderson J, Gaze M, et al. Congenital alveolar rhabdomyosarcoma: clinical and molecular distinction from alveolar rhabdomyosarcoma in older children. Cancer 2001; 91:606.
  84. Sulkowski JP, Raval MV, Browne M. Margin status and multimodal therapy in infantile fibrosarcoma. Pediatr Surg Int 2013; 29:771.
  85. Chung EB, Enzinger FM. Infantile fibrosarcoma. Cancer 1976; 38:729.
  86. Blocker S, Koenig J, Ternberg J. Congenital fibrosarcoma. J Pediatr Surg 1987; 22:665.
  87. Sandberg AA, Bridge JA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors: congenital (infantile) fibrosarcoma and mesoblastic nephroma. Cancer Genet Cytogenet 2002; 132:1.
  88. Bourgeois JM, Knezevich SR, Mathers JA, Sorensen PH. Molecular detection of the ETV6-NTRK3 gene fusion differentiates congenital fibrosarcoma from other childhood spindle cell tumors. Am J Surg Pathol 2000; 24:937.
  89. Rubin BP, Chen CJ, Morgan TW, et al. Congenital mesoblastic nephroma t(12;15) is associated with ETV6-NTRK3 gene fusion: cytogenetic and molecular relationship to congenital (infantile) fibrosarcoma. Am J Pathol 1998; 153:1451.
  90. Orbach D, Brennan B, De Paoli A, et al. Conservative strategy in infantile fibrosarcoma is possible: The European paediatric Soft tissue sarcoma Study Group experience. Eur J Cancer 2016; 57:1.
  91. Chmielecki J, Bailey M, He J, et al. Genomic Profiling of a Large Set of Diverse Pediatric Cancers Identifies Known and Novel Mutations across Tumor Spectra. Cancer Res 2017; 77:509.
  92. Davis JL, Lockwood CM, Albert CM, et al. Infantile NTRK-associated Mesenchymal Tumors. Pediatr Dev Pathol 2018; 21:68.
  93. Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov 2015; 5:25.
  94. Rubin JB, Segal RA. Growth, survival and migration: the Trk to cancer. Cancer Treat Res 2003; 115:1.
  95. Tacconelli A, Farina AR, Cappabianca L, et al. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell 2004; 6:347.
  96. Albert CM, Davis JL, Federman N, et al. TRK Fusion Cancers in Children: A Clinical Review and Recommendations for Screening. J Clin Oncol 2019; 37:513.
  97. Knezevich SR, McFadden DE, Tao W, et al. A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet 1998; 18:184.
  98. Rudzinski ER, Lockwood CM, Stohr BA, et al. Pan-Trk Immunohistochemistry Identifies NTRK Rearrangements in Pediatric Mesenchymal Tumors. Am J Surg Pathol 2018; 42:927.
  99. Hechtman JF, Benayed R, Hyman DM, et al. Pan-Trk Immunohistochemistry Is an Efficient and Reliable Screen for the Detection of NTRK Fusions. Am J Surg Pathol 2017; 41:1547.
  100. Kang J, Park JW, Won JK, et al. Clinicopathological findings of pediatric NTRK fusion mesenchymal tumors. Diagn Pathol 2020; 15:114.
  101. Ferguson WS. Advances in the adjuvant treatment of infantile fibrosarcoma. Expert Rev Anticancer Ther 2003; 3:185.
  102. Loh ML, Ahn P, Perez-Atayde AR, et al. Treatment of infantile fibrosarcoma with chemotherapy and surgery: results from the Dana-Farber Cancer Institute and Children's Hospital, Boston. J Pediatr Hematol Oncol 2002; 24:722.
  103. Cecchetto G, Carli M, Alaggio R, et al. Fibrosarcoma in pediatric patients: results of the Italian Cooperative Group studies (1979-1995). J Surg Oncol 2001; 78:225.
  104. Salman M, Khoury NJ, Khalifeh I, et al. Congenital infantile fibrosarcoma: Association with bleeding diathesis. Am J Case Rep 2013; 14:481.
  105. Orbach D, Rey A, Cecchetto G, et al. Infantile fibrosarcoma: management based on the European experience. J Clin Oncol 2010; 28:318.
  106. DuBois SG, Laetsch TW, Federman N, et al. The use of neoadjuvant larotrectinib in the management of children with locally advanced TRK fusion sarcomas. Cancer 2018; 124:4241.
  107. Laetsch TW, DuBois SG, Mascarenhas L, et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol 2018; 19:705.
  108. Caldwell KJ, De La Cuesta E, Morin C, et al. A newborn with a large NTRK fusion positive infantile fibrosarcoma successfully treated with larotrectinib. Pediatr Blood Cancer 2020; 67:e28330.
  109. Isaacs H Jr. Cutaneous metastases in neonates: a review. Pediatr Dermatol 2011; 28:85.
  110. Lucky AW, McGuire J, Komp DM. Infantile neuroblastoma presenting with cutaneous blanching nodules. J Am Acad Dermatol 1982; 6:389.
  111. Schneider KM, Becker JM, Krasna IH. Neonatal neuroblastoma. Pediatrics 1965; 36:359.
  112. Handler MZ, Schwartz RA. Neonatal leukaemia cutis. J Eur Acad Dermatol Venereol 2015; 29:1884.
  113. Patel N, Price H, Bernert R. Congenital blue nodules on a 4-week-old female. Pediatr Dermatol 2013; 30:387.
  114. Papantoniou K, Sulis ML, Glick S. Vesicopustular eruption in a neonate. Pediatr Ann 2012; 41:186.
  115. Green K, Tandon S, Ahmed M, et al. Congenital acute myeloid leukemia: challenges and lessons. A 15-year experience from the UK. Leuk Lymphoma 2021; 62:688.
  116. Kapur P, Erickson C, Rakheja D, et al. Congenital self-healing reticulohistiocytosis (Hashimoto-Pritzker disease): ten-year experience at Dallas Children's Medical Center. J Am Acad Dermatol 2007; 56:290.
  117. Dupeux M, Boccara O, Frassati-Biaggi A, et al. Langerhans Cell Histiocytoma: A Benign Histiocytic Neoplasm of Diverse Lines of Terminal Differentiation. Am J Dermatopathol 2019; 41:29.
  118. Simko SJ, Garmezy B, Abhyankar H, et al. Differentiating skin-limited and multisystem Langerhans cell histiocytosis. J Pediatr 2014; 165:990.
  119. Bernstein EF, Resnik KS, Loose JH, et al. Solitary congenital self-healing reticulohistiocytosis. Br J Dermatol 1993; 129:449.
  120. Inuzuka M, Tomita K, Tokura Y, Takigawa M. Congenital self-healing reticulohistiocytosis presenting with hemorrhagic bullae. J Am Acad Dermatol 2003; 48:S75.
  121. Satter EK, High WA. Langerhans cell histiocytosis: a review of the current recommendations of the Histiocyte Society. Pediatr Dermatol 2008; 25:291.
  122. Longaker MA, Frieden IJ, LeBoit PE, Sherertz EF. Congenital "self-healing" Langerhans cell histiocytosis: the need for long-term follow-up. J Am Acad Dermatol 1994; 31:910.
  123. Larsen L, Merin MR, Konia T, Armstrong AW. Congenital self-healing reticulohistiocytosis: concern for a poor prognosis. Dermatol Online J 2012; 18:2.
Topic 5788 Version 20.0

References

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