Skip to main content

Kaposiform hemangioendothelioma: current knowledge and future perspectives

Abstract

Kaposiform hemangioendothelioma (KHE) is a rare vascular neoplasm with high morbidity and mortality. The initiating mechanism during the pathogenesis of KHE has yet to be discovered. The main pathological features of KHE are abnormal angiogenesis and lymphangiogenesis. KHEs are clinically heterogeneous and may develop into a life-threatening thrombocytopenia and consumptive coagulopathy, known as the Kasabach-Merritt phenomenon (KMP). The heterogeneity and the highly frequent occurrence of disease-related comorbidities make the management of KHE challenging. Currently, there are no medications approved by the FDA for the treatment of KHE. Multiple treatment regimens have been used with varying success, and new clinical trials are in progress. In severe patients, multiple agents with variable adjuvant therapies are given in sequence or in combination. Recent studies have demonstrated a satisfactory efficacy of sirolimus, an inhibitor of mammalian target of rapamycin, in the treatment of KHE. Novel targeted treatments based on a better understanding of the pathogenesis of KHE are needed to maximize patient outcomes and quality of life. This review summarizes the epidemiology, etiology, pathophysiology, clinical features, diagnosis and treatments of KHE. Recent new concepts and future perspectives for KHE will also be discussed.

Introduction

Kaposiform hemangioendothelioma (KHE) is a rare vascular neoplasm that is typically diagnosed in infancy or early childhood. KHE has intermediate tumor type with locally aggressive characteristics. The predominant feature of the pathology of KHE is progressive angiogenesis and lymphangiogenesis [1, 2]. Clinically, KHE has high morbidity rates, primarily due to local invasive features, compressive effects, or the life-threatening consumptive coagulopathy known as the Kasabach-Merritt phenomenon (KMP) [3,4,5]. Recent studies have rapidly expanded our basic knowledge of KHE, including the etiology, pathophysiology, diagnosis and treatment of the disorder. In this review, we describe the current knowledge and discuss future perspectives on KHE, with the aim of improving our understanding of KHE and preventing mortality and morbidity in severe cases.

Definition

Since 1940, the term ‘Kasabach-Merritt syndrome (KMS)’ has been used for patients with vascular anomalies that are associated with thrombocytopenia and coagulopathy [6]. KMS has also been widely considered a complication of ‘hemangioma’. KHE was first designated by Zukerberg and coworkers in 1993 as an entity distinct from infantile hemangioma (IH) because of its locally invasive growth, aggressive course and ‘focal Kaposi-like appearance’ [7]. In 1997, investigators from two different groups reported that the vascular lesions associated with KMS (or KMP) were not IHs as was previously believed [8, 9]. Currently, KMP is defined as profound thrombocytopenia, together with consumptive coagulopathy and hypofibrinogenemia associated only with the vascular tumors, KHE and tufted angioma (TA) (Fig. 1) [10, 11]. Conceptually, KHE and TA are part of the same neoplastic spectrum and can be present in the same biopsy specimen of the same patient [12].

Fig. 1
figure 1

A 3-month-old male infant presenting KHE associated with KMP. The tumor lesion grew progressively after the first week of age and extended through the whole thigh, scrotum and abdominal wall, with ‘extensive thrombocytopenic purpura’ as described by Kasabach and Merritt in 1940

Epidemiology and demography

Currently, there is a paucity of literature specifically addressing the incidence of KHE. In Massachusetts, the annual prevalence and incidence have been estimated at 0.91 and 0.071 per 100,000 children, respectively [13]. However, asymptomatic KHE lesions are less likely to be reported or diagnosed by pathology. Indeed, small asymptomatic or atypical KHEs can be misdiagnosed as unusual variants of IH or other vascular anomalies [14]. Therefore, the actual prevalence and incidence of KHE are most likely higher than those indicated in the limited published reports.

Previously, KHE was shown to have an equal sex predilection. However, a slight male predominance has been indicated by two large retrospective studies, both of which collected data from more than 100 patients with KHE [10, 11]. The distribution of age at onset has one peak within the first year of life when approximately 90% of KHE are evident. Approximately 50% of cutaneous lesions are visible or detectable at birth [14].

Etiology

In general, the etiology of KHE remains largely unknown. Almost all cases of KHE arise without any obvious cause. In rare scenarios, signs and symptoms of KHE/TA can worsen with trauma or infections. There is also evidence that aggravation or manipulation of the tumor either from surgery or trauma can incite KMP, whether the patient had a history of KMP or not [15,16,17]. In addition, rapid enlargement of the lesion with severe KMP development shortly after vaccination has been reported in several patients [18,19,20,21]. These phenomena raise the intriguing possibility that physical trauma and the inflammatory response might contribute to the aggravation of KHE.

It is likely that the origin of KHE is multifactorial, with genetic factors being part of the contributing triggers, although the mutations in KHE tumor are sporadic rather than germline. Rapid advances in the area of molecular genetics have enabled the identification of somatic mutations in many types of vascular anomalies. Zhou and coworkers provided evidence of a somatic translocation between chromosomes 13 and 16 at the bands of 13q14 and 16p13.3 in 10% of metaphase cells in KHE lesions with the presence of normal cells in the karyotype [22]. Remarkably, the somatic activating GNA14 c.614A > T (p.Gln205Leu) mutation was found in 1/3 of KHE specimens and in 1/4 of TA specimens, although these studies were weakened by small sample size [23]. Somatic mutations in GNAQ and its paralogues (e.g., GNA11 and GNA14) have also been identified in many other vascular tumors [24, 25], vascular malformations [26, 27] and solid tumors [28, 29].

The GNAQ family encodes Gα subunits that form a heterotrimer with Gβ and Gγ subunits and bind G-protein-coupled receptors (GPCRs). GPCRs are involved in many aspects of tumor and vascular biology [30, 31]. In addition, platelet aggregation, glucose secretion, and inflammation are among the physiological processes affected by GPCRs [32]. p.Gln205Leu substitution can induce changes in cellular morphology and render cells growth-factor independent by upregulating the MAPK/ERK1/2 pathway (Fig. 2) [23]. However, it is important to note that although GNAQ mutations have been found in KHE, we do not know whether they are causative or develop secondarily in the tumor.

Fig. 2
figure 2

G-protein-coupled receptors (GPCRs) participate in different physiological processes. The binding of ligands to GPCRs triggers a universal G protein allosteric mechanism that promotes the exchange of GDP with GTP on the α subunit of heterotrimeric G proteins. This event causes the dissociation of Gα from the dimer. Gα subunits mediate signals between GPCRs and intracellular signaling cascades. These signaling pathways include the PI3K/AKT/mTOR and MAPK/ERK pathways, both of which can mediate different biological processes, such as cell proliferation, migration and survival. Mutations in GPCRs and G proteins have been found at a very high frequency in tumor cells and endothelial cells in vascular anomalies

It is unclear how mutations in the same gene can lead to different vascular anomalies or clinical manifestations, but the mechanism may be based on the location of the mutation in the gene, the cell type(s) affected, and/or the point in development at which the mutations occur [33]. In these scenarios, the highly variable clinical presentations of KHE further reflect the complexity of gene mutations in the development of this rare disease. It is also conceivable that undetected mutations exist in KHE lesions given that many technical hurdles are still present, although these are not likely to be insurmountable in the future.

Pathophysiology

The pathophysiology of KHE may not be attributable to a single mechanism, but rather, to a combination of events that have not yet been elucidated or understood completely.

Angiogenesis and lymphangiogenesis

KHE is the result of dysregulation of both angiogenesis and lymphangiogenesis. In vivo, mouse hemangioendothelioma cells can form KHE-like, intradermal tumors. Interestingly, overexpression of prospero-related homeobox-1 (Prox-1) in mouse hemangioendothelioma cells induces an invasive phenotype in vivo, enhances the migration rate in vitro, and significantly upregulates the expression of podoplanin (D2–40) and vascular endothelial growth factor receptor-3 (VEGFR-3) [34]. Recent data indicated that KHE-derived mesenchymal stem cells (MSCs) have the capacity to support vascular network formation in vitro [35]. In addition to expressing VEGFR-3, KHE-derived MSCs also show higher levels of VEGF-C than normal lymphatic endothelial cells [35].

VEGF-C/VEGFR3 axis

The vascular endothelial growth factor-C (VEGF-C)/VEGFR3 axis in lymphatic endothelial cells (LECs) is important throughout lymphangiogenenic growth [36]. The expression of both VEGFR-3 and VEGF-C in KHE suggests that the VEGF-C/VEGFR3 axis may contribute to the aggressive behavior of KHE [37, 38]. The VEGF-C/VEGFR3 axis has been implicated in tumor progression by directly affecting tumor cells or modulating lymphangiogenesis and the immune response (Fig. 3) [39]. The VEGF-C/VEGFR-3 axis has been demonstrated to promote tumor growth in an autocrine manner [40]. In addition to lymphangiogenesis, VEGF-C/VEGFR3 signaling has also been shown to be important for angiogenesis, acting together with VEGF-A/VEGFR-2 and Dll4/Notch signaling to control angiogenesis [41]. VEGF-C/VEGFR3 axis may play an important role in chronic inflammation associated with KHE [42, 43].

Fig. 3
figure 3

VEGF-C/VEGFR3 and Ang-2/Tie-2 signaling pathways play an important role in lymphangiogenesis. The binding of VEGF-C can stimulate the activation of VEGFR-3 and induce downstream PI3K/Akt/mTOR signaling, which mediates lymphangiogenesis. VEGF-C binding to NRP-2 can form a complex with VEGFR-3, further activating the VEGFR-3 signaling that enhances the proliferation of lymphatic endothelial cells (LECs) and lymphangiogenesis. Ang-2 ligand-induced Tie-2 activation triggers Akt/mTOR signaling, which in LECs is mainly mediated by PIK3CA. Gene-targeting experiments have identified that the Ang-2/Tie-2 signaling system is needed for physiological and pathological remodeling of lymphatic vessels. Sirolimus (rapamycin), which is an inhibitor of mTOR, complements current antilymphatic strategies in the treatment of vascular anomalies, such as KHE

Angiopoietin-2/tie-2 signaling

The high serum levels of angiopoietin-2 (Ang-2) in patients with KHE raise the possibility that Ang-2 might have a paracrine effect on endothelial cells (ECs) in KHE lesions. Remarkably, Ang-2 levels dramatically decrease with sirolimus treatment in patients with KHE and KMP [44]. It is therefore possible that Ang-2 might have a role in the development of KMP in patients with KHE. The Ang/Tie2 ligand-receptor system is required for lymphatic and blood vessel development during midgestation. The Ang/Tie2 pathway controls vascular permeability, inflammation and pathological angiogenic and lymphatic responses postnatally. Ang-2 acts as a context-dependent weak Tie-2 agonist or antagonist that can inhibit the Ang-1/Tie-2 signaling axis [45]. In humans, Ang-2 levels are greatly increased during vascular remodeling, which occurs, for example, during vessel sprouting and inflammatory lymphangiogenesis [46]. This may be a potential explanation for the upregulation of Ang-2 in patients with KHE. Ang-2-induced Tie-2 activation triggers Akt/mTOR signaling, which in ECs is mainly mediated by PIK3CA (which encodes PI3K catalytic subunit-α) [47].

Platelet aggregation: a key trigger of KMP?

Intralesional platelet trapping is followed by the activation and aggregation of platelets, which then results in activation of the coagulation cascade with subsequent consumption of clotting factors. Platelet trapping has been revealed histologically in KHE with or without KMP [48]. Of interest then is the mechanism or mechanisms of increased platelet trapping in KHE vessels. One hypothesis is that EC damage or alteration in KHE can lead to exposure of extracellular matrix components, which are ligands for platelet adhesion [1]. The C-type lectin-like receptor-2 (CLEC-2) expressed on platelets is an endogenous receptor of podoplanin, which in turn is widely expressed in ECs within the KHE lesions [49, 50]. Binding of podoplanin to CLEC-2 can transmit platelet activation signals via Src family kinases, which may account for platelet aggregation in KHE [51].

Podoplanin is highly expressed in dysmorphic vessels within the lymphatic malformations, but no obvious platelet aggregation occurs in these lesions [52]. Therefore, there may also be alternative and/or additional mechanisms by which platelet trapping is triggered in KHE lesions [2, 53]. In KHE, the thrombi in the microvasculature cause vessel occlusion and prevent normal blood flow, all of which can lead to elevated shear stress. It is recognized that high shear stress induces an increased activation of platelets in vitro and in vivo, with a mechanism dependent on von Willebrand factor interacting with both its platelet binding sites, glycoprotein (GP) Ib-IX and GP IIb-IIIa [54, 55]. With regard to KHE, platelets in circulating blood may become exposed to turbulent blood flow and high shear stress that results from the architecture of the small, convoluted and thrombus-obstructed vessels within the KHE lesions. This process in turn causes further platelet trapping and activation during the active phase of KHE.

Continued platelet aggregation, together with coagulopathy and hypofibrinogenemia with elevated D-dimer (coagulation markers), eventually result in intralesional hemorrhage, which clinically manifests as very purpuric, warm, painful, and rapidly enlarged tumor lesion [3]. In patients with KHE, the coexistence of KMP always represents aggressive tumor progression. Although the pathophysiological roles of activated platelets in the KHE tissue environment are not yet fully understood, these observations raise the intriguing possibility that the activated platelets might contribute to worsening coagulopathy by promoting critical processes such as neovascularization. This hypothesis is supported by findings that platelets are activated in different tumor vasculatures [56,57,58,59]. Platelets are reservoirs of proangiogenic proteins that are mainly stored in α-granules and secreted upon physiological and pathological stimulations. Various types of tumor cells and tumor-derived ECs can activate platelets by different mechanisms. There is plenty of evidence that activated platelets exert their pleiotropic effects on many biological processes central to angiogenesis, progression, inflammation and metastasis in various tumor types [57, 60, 61]. Improving our understanding of such platelet involvement in neovascularization may potentially help in the development of alternative treatment strategies for patients with KHE.

Clinical characteristics

The manifestations of KHE are variable and range from cutaneous lesions with wide varieties of appearances to deep masses without cutaneous signs. The clinical features also differ substantially between patients with KMP and patients without KMP [13, 14, 48]. In the majority of patients, KHE is a single soft tissue mass with cutaneous findings that range from an erythematous papule, plaque, or nodule to an indurated, purple and firm tumor. With KMP, these lesions are purpuric, hot to the touch, swollen and very painful. Most patients experience progressive lesion enlargement and/or symptom progression [62,63,64]. However, a small but significant minority of KHEs do not grow [13, 48]. Approximately 12% of patients lack cutaneous involvement [14].

Complications

Complications in patients with KHE are common. The complication severity strongly depends on the age, lesion size, lesion location, lesion extension into the deep tissue and vital organs, and associated hematologic abnormalities. It is prudent for clinicians to remain vigilant of potential complications and of risk factors that may herald future complications.

Kasabach-Merritt phenomenon

KMP occurs with an estimated incidence of 42 to 71% [2, 13, 14, 64]. The thrombocytopenia is usually severe, with a median platelet count of 21 × 109/L at the initial presentation of KMP [14]. KHE lesions with KMP have progressive engorgement and purpura. KMP can lead to significant pain and secondary bleeding. As far as we know, KHE appear to be congenital as the majority of cases are diagnosed in the newborn/infancy period. It is now thought that the few cases in which adults were found to have KHE or to develop KMP, occurred because of an inciting event like trauma or pregnancy. The risk of KMP is highest for congenital KHEs with a large size (especially > 8 cm in diameter) [48, 65]. Anatomic location may also be a predictor of KMP. Clinically, intrathoracic KHEs are frequently associated with KMP [66,67,68]. The frequency of KMP in the retroperitoneal KHEs was also high [13, 14]. Evidently, intrathoracic and retroperitoneal lesions tend to be more expansive and infiltrative and are more likely to develop KMP. The identification of the most dangerous factors associated with KMP will be enormously helpful to treating clinicians.

Musculoskeletal disorders

The infiltrative nature and destructive growth patterns of KHE can cause functional limitations and pain; all of these musculoskeletal disorders may affect patients’ abilities to perform routine daily activities and eventually influence quality of life [4, 69]. Acute pain at the tumor sites is a common symptom during KMP [14]. Even in patients without KMP, musculoskeletal disorders are frequently seen in cases involving the extremities, with a majority of these lesions located on or adjacent to joints [70]. Progressive thoracolumbar scoliosis can be seen in patients with thoracic or retroperitoneal lesions [71,72,73]. It is important that these patients are diagnosed early and treated more aggressively at the outset even in the absence of KMP. In some cases, residual KHEs will continue to infiltrate surrounding tissues, erode bone and destroy joints. Infiltration of the muscles and connective tissues in patients with KHE can alter the structural matrix and mechanical properties of these tissues, leading to chronic degenerative changes. Pathologically, untreated KHE lesions are characterized by progressive fibrosis [12]. There is ample evidence for the crucial role of platelet activation and aggregation in the development of fibrosis in different tissues and organs [74, 75]. It is conceivable that platelet activation and aggregation in muscles and connective tissues during KHE infiltration can activate variable fibrotic pathways. The diffuse intraarticular and periarticular fibrosis can further aggravate muscular atrophy and lead to subluxations and flexion contracture of the involved joints. Patients may eventually suffer from recalcitrant pain and fixed contractures (Fig. 4).

Fig. 4
figure 4

Deep KHE with bone and joint destruction in a 3.5-year-old girl. The patient had been diagnosed with a left-hip KHE associated with KMP at 1 year of age. She received a partial resection at the local hospital. Although surgical excision improved the associated KMP, she exhibited a progressively decreased hip range of motion. An anteroposterior pelvis radiograph showed right hip subluxation and progressive bone erosion in the left ilium and proximal femur before referral (a, b and c). Coronal T2-weighted MRI revealed a deep lesion infiltrating the left ilium and proximal femur at the time of referral to our department (d, e)

Lymphedema

Lymphedema is the chronic, progressive swelling of subcutaneous tissue caused by inadequate lymphatic function. Lymphedema may be caused by anomalous development (primary) or injury (e.g., infection) to lymph nodes or lymphatic vessels [76]. Lymphedema may be a potential sequela of KHE, particularly tumor involving the legs [12, 77]. KHE involving the proximal extremity, particularly at or adjacent to the inguinal or axillary lymph nodes, may influence lymphatic development or damage the lymphatic vasculatures. In this scenario, it is hypothesized that the mechanical obstruction of the lymphatic flow during the acute phase of KMP may eventually lead to lymphedema [78]. In addition, active and chronic inflammation may also play a role in the development of lymphedema in patients with KHE.

Compression of vital structures

Compression of vital structures may occur in a number of settings and is far more frequently observed in patients with KMP than in patients without KMP [14]. The progressive expansion of the mass during the active phase of KMP can further compromise the vital structures. Obstructing KHEs of the airway typically involves the neck and thorax [67, 79]. KHEs involving the pancreas are extremely uncommon but have the potential to cause obstructive jaundice [80, 81]. If compression of vital structures is visualized, prompt therapy should be initiated even without KMP.

Diagnosis

The diagnosis of KHE often requires an analysis of clinical, imaging, hematological and/or histological characteristics, but even with all of these data, the diagnosis might not be readily apparent in certain cases [73]. In patients with deep KHE without KMP (bone and/or joint and so on), a definitive diagnosis is often delayed because of the non-specific and wide variety of symptoms.

Ultrasound is the modality of choice for small and superficial lesions [82]. Magnetic resonance imaging (MRI) is generally first-line assessment because the deep infiltrating nature of KHE may not be apparent on physical exam or on ultrasound. MRI with and without gadolinium has the most value in the diagnosis of KHE as well as for clearly determining the extent of involvement and response to treatment [83]. On MRI scans, KHEs usually exhibit ill-defined margins and are characterized by multiplanar involvement, diffuse enhancement, and adjacent fat stranding in unusual locations, iso-intensity relative to adjacent muscle on T1-weighted imaging, hyperintensity on T2-weighted imaging (Fig. 5). Adjacent bone and/or joint changes are common. The MRI findings of these changes include destruction of the adjacent cortex, injury of the epiphyseal region and invasion to near joints. Deep KHE with KMP should be considered in patients presenting with an unexplained severe thrombocytopenia and coagulopathy, especially in patients coexisting with cutaneous purpura and severe anemia. MRI scan of the abdomen and chest should be recommended for such patients [84, 85].

Fig. 5
figure 5

Clinical and MRI features of KHE with KMP. a A 2-month-old boy was found to have a chest wall mass after birth. The mass became progressively indurated and purpuric. The boy developed profound thrombocytopenia and consumptive coagulopathy. b Horizontal T1-weighted MRI revealed that the heterogeneous mass was isointense relative to the adjacent muscle on T1-weighted imaging. Horizontal (c) and sagittal (d) T2-weighted MRI revealed hyperintense lesions infiltrating the right lateral chest wall

Biopsy is gold standard for diagnosis and should be performed if possible and safe. Biopsy is frequently not possible or recommended in KHE with severe KMP, and can potentially worsen the coagulopathy. Biopsy specimens should be considered in patients with an uncertain diagnosis and atypical clinical manifestation (e.g., at an unusual site). The histologic hallmark of KHE is infiltrating, defined, rounded and confluent nodules, which are composed of spindle endothelial cells. These spindle endothelial cells align to form malformed lymphatic channels and slit-like vascular lumina containing erythrocytes, along with platelet thrombi, eosinophilic hyaline bodies and the extravasation of hemosiderin deposits. Immunohistochemical staining shows that endothelial cells in KHE lesions are positive both for vascular endothelial markers CD31 and CD34, lymphatic endothelial marker VEGFR-3, D2–40, lymphatic endothelial hyaluronan receptor-1 and Prox-1, but negative for glucose transporter-1(Glut-1) and human herpes virus-8 staining (Fig. 6) [2, 8, 9].

Fig. 6
figure 6

Histopathological characteristics of KHE. aHematoxylin and eosin (H&E)-stained sections of KHE (original magnification × 100). The histologic hallmark of KHE was infiltrating, defined, rounded and confluent nodules, which were composed of spindle endothelial cells. b These spindle endothelial cells aligned to form malformed lymphatic channels and a slit-like vascular lumina (× 200). Immunohistochemical staining showed that the endothelial cells in KHE lesions were positive for the vascular endothelial markers CD31 (c, × 100) and CD34 (d, × 100) and the lymphatic endothelial marker D2–40 (e, × 100) but were negative for glucose transporter-1 and human herpes virus-8 staining (data not shown). Ki-67 was noted in only a few nuclei in the lesion (f, × 100)

Differential diagnoses

The heterogeneous clinical, radiographic and laboratory findings of KHE raise an extensive differential diagnosis that includes IH, venous malformation, congenital hemangioma and Kaposiform lymphangiomatosis (KLA), among others. A definitive preoperative differential diagnosis between deep KHE and a malignant tumor (e.g., metastasis neuroblastoma and sarcoma) is also challenging in patients with spine involvement without KMP.

Infantile hemangioma

One of the most important diseases to rule out in the differential diagnosis of KHE is IH. The appearance of IH is dictated by the depth, location, and stage of evolution. A defining feature of IH is its dramatic growth (between 5.5 and 7.5 weeks of age) and development into a disorganized mass of blood vessels [86]. Cutaneous IHs are usually diagnosed clinically and frequently without needing for imaging. Imaging examinations and other investigations may be required in special situations (e.g., paraglottic or hepatic IHs). Both KHE and IH will look hypervascular on ultrasound. MRI is helpful in differentiating the difficult cases. Throughout the development of IHs, ECs in IH lesions are positive for Glut-1, which is absent in KHE and other vascular tumors [87, 88].

Congenital hemangiomas

Congenital hemangiomas are biologically and behaviorally distinct from KHEs. They are benign vascular tumors of infancy that arise in utero and are present and fully formed at birth. The 3 variants of congenital hemangiomas are rapidly involuting congenital hemangiomas (RICHs), partially involuting congenital hemangiomas (PICHs) and noninvoluting congenital hemangiomas (NICHs) [89, 90]. The defining feature of RICHs is accelerated regression, which is usually complete within the first year of life, leaving behind atrophic skin (Fig. 7). NICHs persist in a stable state without growth or involution. However, a slight enlargement of NICHs over the years has recently been reported [91]. PICHs evolve from RICHs to persistent NICH-like lesions. Congenital hemangiomas can look very much like KHE particularly in the newborn period. Congenital hemangiomas do not exhibit progressive postnatal growth. In contrast, KHE tumors that develop KMP will appear to ‘grow’ and become engorged and purpuric in the first days/weeks/months of life. Mild consumption of coagulation factors can occur but most importantly, the coagulopathy associated with congenital hemangioma is not associated with bleeding issues and tends to self resolve in 1 to 2 weeks [92]. Treatment with surgical excision, as there are no medical therapies currently, for congenital hemangiomas may be required for cosmetic reasons or to resolve persistent pain in patients with NICH [89].

Fig. 7
figure 7

Rapidly involuting congenital hemangiomas (RICH). a RICH is fully formed at birth and then involutes, mostly during the first year of life. The patient’s hematologic parameters were within the normal reference ranges. b The same RICH involuted rapidly without any treatment. By 9 months of age, the lesion had involuted completely, leaving dermal atrophy

Venous malformation

KHE with KMP should be differentiated from the clotting disorder associated with extensive venous malformations (VMs). VMs are slow-flow vascular malformations present at birth. In patients with extensive VMs, especially involving the trunk and/or extremities, localized intravascular coagulopathy can occur at baseline and be worsening by any aggravation of the malformation such as trauma or surgery. The levels of fibrinogen are low, associated with elevated D-dimer and fibrin degradation products. However, the thrombocytopenia is less profound in VMs than in KHE with KMP [93]. VMs can usually be diagnosed based on patient history, physical examination and MRI. On light microscopy, VMs are characterized by enlarged and irregular venous channels lined by a flattened layer of ECs [94].

Kaposiform lymphangiomatosis

KLA is a rare, complex lymphatic disorder with multifocal or diffuse lesions. KLAs often involve the mediastinum, lung, abdomen and multiple bones [95,96,97]. Interestingly, thrombocytopenia and coagulopathy in KLA have overlapping features with KMP in KHE. The thrombocytopenia in some KLAs is extremely severe, similar to that observed in KMP [98, 99]. Histologically, KLAs are composed of malformed lymphatic channels with dispersed and poorly marginated clusters or sheets of spindled lymphatic ECs [96, 98]. In vitro, KHE cells can support vascular network formation, whereas KLA cells appear inert in this capacity [35]. A somatic activating NRAS variant (c.182 A > G, p. Q61R) was recently identified in patients with KLA but was absent in KHE samples, thus providing a molecular means to further differentiate these two entities [100]. In addition, KLA appears to be more refractory to medical therapies, with an overall survival rate of only 34% [96].

Management

Due to the marked heterogeneity and rarity of KHEs, no validated scores have been established to assess disease severity. Consensus treatment statements by Drolet et al. [83] were published in 2013. Medical treatments with corticosteroids and/or vincristine have been recommended for the management of KHE. However, these recommendations are based on expert opinion rather than rigorous clinical studies. There is a lack of well-designed clinical trials and insufficient evidence to support existing interventions. There is also no definite treatment guideline for the long-term observation of patients with KHE [101].

Currently, the management of KHE has been based upon a review of the available evidence, expert opinions, and clinical experiences. Sirolimus has recently been suggested as a treatment option for complicated vascular anomalies and tumors in children, including KHE with or without KMP [102]. In many patients, multiple treatments are given in sequence or in combination. Notably, the treatment practices and regimens for KHE should be tailored to individual patients and guided by specific clinical circumstances. Patients with KMP should be treated aggressively with a combined regimen; monotherapy is usually not recommended. Several studies for combined regimens in the treatment of KHE are currently accruing subjects, including one randomized controlled trial (ClinicalTrials.gov identifier NCT03188068). Supportive care treatments (e.g., cryoprecipitate) are often required for patients with KMP (Table 1). Platelet transfusion should not be used unless the patient is actively bleeding or in preparation for surgery. Ideally, a patient with KHE who also exhibits severe complications or is at risk of complications should be referred to a multidisciplinary team for evaluation and treatment.

Table 1 Management options for KHEa

Pharmacological treatments

Vincristine

Haisley-Royster et al. [103] reported encouraging findings regarding the successful use of vincristine in the management of KMP. Many studies have also demonstrated the satisfactory outcomes of vincristine in treating KHE with KMP, including steroid-resistant patients [5, 104,105,106]. First-line therapy with vincristine or vincristine plus corticosteroids is recommended for cases of KHE with KMP in consensus-derived guidelines [83, 107]. First-line treatment with vincristine has an overall response rate of 72% [108]. There is mounting evidence that vincristine monotherapy is not effective in very severe patients [109, 110]. In this regard, successful use of vincristine plus ticlopidine has been reported in some cases [5, 111].

Corticosteroids

Systemic corticosteroid treatment is recommended as another first-line therapy for KMP because of its success in rapidly normalizing platelet counts [83, 107]. The sustained response, however, is variable, and many cases do not improve with corticosteroid monotherapy (with an overall response rate of 10–27%), even when higher doses are given [106, 112]. In addition, long-term corticosteroid use has undesired side effects, such as temporary growth retardation, an increased risk of infection and behavioral changes [113]. Recent studies have suggested that corticosteroids can be used in combination therapies for KMP [73, 114]. Considering the undesirable side effects of long-term daily corticosteroid treatment in children, they should be weaned off these drugs as soon as medically feasible.

Sirolimus

Since 2010, an increasing number of studies have reported the exceptional effectiveness of sirolimus and everolimus, which are well-known mTOR inhibitors, on KHE. The authors described a reduction in KHE size, an eventual normalization of platelet counts in KMP patients, and in some cases, improvements in musculoskeletal pain, function and quality of life [80, 109, 112, 114,115,116,117,118,119,120,121,122,123,124]. For patients who either did not respond to the previous treatment (e.g., corticosteroids and vincristine) or who relapsed once the dose was tapered, sirolimus therapy still exhibited a high response rate (94%) [108]. However, sirolimus alone is usually not sufficient to treat severe KMP. In patients with severe KMP, sirolimus in combination with the short-term administration of corticosteroid has been recommended (Fig. 8) [114]. Sirolimus plus steroids is now considered as a first-line therapy for the treatment of KHE with KMP (versus vincristine plus steroids). Many clinicians (and families) prefer sirolimus (plus steroids) over vincristine because vincristine requires a central line.

Fig. 8
figure 8

Congenital KHE associated with KMP on the right face. Congenital KHEs with KMP likely represent a period of temporary and partial remission shortly after birth. The signs and symptoms may alleviate spontaneously. However, rebound growth of the lesions accompanying severe KMP would reoccur within the next several days or weeks. a The parents’ photograph revealed a bluish, swollen and firm vascular mass on the right face after birth (2 days of age). Her platelet count was 7 × 109/L. Without any special treatment, the tumor became soft and was stagnant in size. Her platelet count reached a highest value of 161 × 109/L (b: 1 week of age, c: 6.5 weeks of age). Subsequently, however, the tumor became progressively enlarged and displayed obvious ecchymosis (d: 8 weeks of age). The patient’s platelet count dropped to 3 × 109/L. She was treated with a combination therapy of sirolimus (0.8 mg/m2 administered twice daily) and prednisolone (2 mg/kg/d). One (e) and 4 weeks (f) after treatment. Within 10 days of combination therapy, the girl’s platelet level normalized. The prednisolone was tapered and discontinued within the following 4 weeks, and sirolimus was continued. G, Twelve months after treatment. H, Photograph at 26 months of age (24 months of treatment) demonstrates a nearly complete involution of the lesion. Sirolimus was then tapered and discontinued

Although sirolimus is clearly efficacious, rare side effects, such as interstitial pneumonitis and Pneumocystis carinii pneumonia, may be life-threatening [125, 126]. Currently, the optimal sirolimus dose and prophylactic regimen in patients with KHE has not been established. Many authors have reported maintaining the serum levels between 8 and 15 ng/ml [114, 116, 120]. However, low-dose sirolimus (2–3 ng/ml serum levels) may be associated with low toxicity and has been demonstrated to be effective for treating patients without KMP [127]. Interestingly, there is mounting evidence that low-dose sirolimus markedly ameliorates the development of inflammation and fibrogenesis in animal models, providing a theoretical basis for its use in KHE with musculoskeletal disorders [128, 129].

Topical treatments

Several case reports and case series have reported success using topical sirolimus and tacrolimus ointment in superficial KHE/TA. The investigators have shown a good response of KHE/TA to twice daily topical application of these drugs [130, 131]. Tacrolimus is an anti-T-cell immunosuppressive drug that is FDA-approved as a topical gel formulation (available in a concentration of 0.03 and 0.1%) for the treatment of cutaneous inflammatory/fibrotic diseases [132]. The clinically important implication of topical treatments is that superficial KHE/TA can be treated with local/topical agents, thus decreasing the potential complications associated with systemic treatments. However, it is important to note that most of these cases are TA lesions rather than KHE. It is also important to ensure that there is not a deep component.

Other pharmacological therapies

Several other medicines have been used in an attempt to optimize efficacy. Ticlopidine and aspirin are specific ant-platelet-aggregating agents. Successful use of ticlopidine plus aspirin in KMP has been described [133]. Interferon-α and propranolol have also been used to treat KHE. However, the standard protocols are inadequate because responses to these agents are variable and unpredictable [134,135,136,137]. In addition, the side effects of interferon-α are significant and include spastic diplegia [138].

Invasive interventions

Elective resection of a KHE during the active phase of KMP is unusually not necessary and is ill advised. Given the young age of the patients and the vascularity of the tumors, these patients are at higher risk of blood loss and iatrogenic injury, along with worsening of the coagulopathy. Clinically, surgery is rarely an option for extensive KHEs or for patients in whom surgery will result in significant functional impairment. Conversely, surgery can be an approach for tumors in which a complete and safe resection can be performed [48]. Surgery is also an option for resection of a fibrofatty residuum or reconstruction of damaged structures [12]. Failure of pharmacotherapy may lower the threshold for the resection of a cosmetically or functionally problematic KHE [139]. In patients with KMP, arterial embolization may have a role in disease control [139]. Embolization can initially decrease blood flow from the tumor and decrease risk of high output cardiac failure. However, a remarkable limitation of embolization is the technical difficulty of cannulating very small feeder vessels in young patients. The possibility of worsening the hematological parameters by invasive interventions is also important and highlights the need for employing more established techniques in these patients.

Conclusions and future directions

Although the incidence of KHE is low, it can cause morbidity and mortality in children and adults. Consequently, prompt diagnosis and appropriate management are crucial to improving the long-term prognosis of patients. Mutations and their pathways are providing potential targets for the development of novel pharmacotherapy for KHE. The future challenge will be to dissect the mutations and signaling cascades in terms of their pharmacological relevance. It is likely that the rapid advances in basic science and translational medicine will facilitate the development of important, new, and targeted molecular treatment strategies for KHE. Further clinical studies are also needed to refine the guidelines for the standard use of therapies and follow-up in patients with KHE.

Availability of data and materials

The datasets used and/or analyzed during the current study available from the corresponding author on reasonable request.

Abbreviations

Ang-2:

Angiopoietin-2

CLEC-2:

C-type lectin-like receptor-2

CT:

Computed tomography

GP:

Glycoprotein

GPCR:

G-protein-coupled receptor

IH:

Infantile hemngioma

KHE:

Kaposiform hemangioendothelioma

KLA:

Kaposiform lymphangiomatosis

KMP:

Kasabach-Merritt phenomenon

KMS:

Kasabach-Merritt syndrome

MRI:

Magnetic resonance imaging

MSC:

Mesenchymal stem cell

NRP-2:

Neuropilin-2

Prox-1:

Prospero-related homeobox-1

TA:

Tufted angioma

VEGF-C:

Vascular endothelial growth factor-C

VEGFR-3:

Vascular endothelial growth factor receptor-3

VM:

Venous malformations

References

  1. O'Rafferty C, O'Regan GM, Irvine AD, Smith OP. Recent advances in the pathobiology and management of Kasabach-Merritt phenomenon. Brit J Haematol. 2015;171(1):38–51.

    Article  CAS  Google Scholar 

  2. Lyons LL, North PE, Mac-Moune LF, Stoler MH, Folpe AL, Weiss SW. Kaposiform hemangioendothelioma: a study of 33 cases emphasizing its pathologic, immunophenotypic, and biologic uniqueness from juvenile hemangioma. Am J Surg Pathol. 2004;28(5):559–68.

    Article  PubMed  Google Scholar 

  3. Mulliken JB, Anupindi S, Ezekowitz RAB, Mihm MC. Case 13-2004. New Engl J Med. 2004;350(17):1764–75.

    Article  CAS  PubMed  Google Scholar 

  4. Schaefer BA, Wang D, Merrow AC, Dickie BH, Adams DM. Long-term outcome for kaposiform hemangioendothelioma: a report of two cases. Pediatr Blood Cancer. 2017;64(2):284–6.

    Article  PubMed  Google Scholar 

  5. Fernandez-Pineda I, Lopez-Gutierrez JC, Chocarro G, Bernabeu-Wittel J, Ramirez-Villar GL. Long-term outcome of vincristine-aspirin-ticlopidine (VAT) therapy for vascular tumors associated with kasabach-Merritt phenomenon. Pediatr Blood Cancer. 2013;60(9):1478–81.

    Article  CAS  PubMed  Google Scholar 

  6. Kasabach H, Merritt K. Capillary hemangioma with extensive purpurareport of a case. Am J Dis Child. 1940;5(59):1093–70.

    Google Scholar 

  7. Zukerberg LR, Nickoloff BJ, Weiss SW. Kaposiform hemangioendothelioma of infancy and childhood. An aggressive neoplasm associated with Kasabach-Merritt syndrome and lymphangiomatosis. Am J Surg Pathol. 1993;17(4):321–8.

    Article  CAS  PubMed  Google Scholar 

  8. Enjolras O, Wassef M, Mazoyer E, Frieden IJ, Rieu PN, Drouet L, et al. Infants with Kasabach-Merritt syndrome do not have "true" hemangiomas. J Pediatr. 1997;130(4):631–40.

    Article  CAS  PubMed  Google Scholar 

  9. Sarkar M, Mulliken JB, Kozakewich HP, Robertson RL, Burrows PE. Thrombocytopenic coagulopathy (Kasabach-Merritt phenomenon) is associated with Kaposiform hemangioendothelioma and not with common infantile hemangioma. Plast Reconstr Surg. 1997;100(6):1377–86.

    Article  CAS  PubMed  Google Scholar 

  10. Adams DM, Brandão LR, Peterman CM, Gupta A, Patel M, Fishman S, et al. Vascular anomaly cases for the pediatric hematologist oncologists—An interdisciplinary review. Pediatr Blood Cancer. 2018;65(1):e26716 -n/a.

    Article  Google Scholar 

  11. Mahajan P, Margolin J, Iacobas I. Kasabach-Merritt Phenomenon: Classic Presentation and Management Options. Clin Med Insights Blood Disord. 2017;10:1179545X17699849.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Enjolras O, Mulliken JB, Wassef M, Frieden IJ, Rieu PN, Burrows PE, et al. Residual lesions after Kasabach-Merritt phenomenon in 41 patients. J Am Acad Dermatol. 2000;42(2 Pt 1):225–35.

    Article  CAS  PubMed  Google Scholar 

  13. Croteau SE, Liang MG, Kozakewich HP, Alomari AI, Fishman SJ, Mulliken JB, et al. Kaposiform hemangioendothelioma: atypical features and risks of Kasabach-Merritt phenomenon in 107 referrals. J Pediatr. 2013;162(1):142–7.

    Article  PubMed  Google Scholar 

  14. Ji Y, Yang K, Peng S, Chen S, Xiang B, Xu Z, et al. Kaposiform haemangioendothelioma: clinical features, complications and risk factors for Kasabach-Merritt phenomenon. Br J Dermatol. 2018;179(2):457–63.

    CAS  PubMed  Google Scholar 

  15. Karnes JC, Lee BT, Phung T, Alomari AI, Mulliken JB, Greene AK. Adult-onset kaposiform hemangioendothelioma in a posttraumatic site. Ann Plast Surg. 2009;62(4):456–8.

    Article  CAS  PubMed  Google Scholar 

  16. Goyal A, Babu SN, Kim V, Katariya S, Rao KL. Hemangioendothelioma of liver and spleen: trauma-induced consumptive coagulopathy. J Pediatr Surg. 2002;37(10):E29.

    Article  PubMed  Google Scholar 

  17. Kim CY, Nam YH, Kim GD, Oh CW. Tufted angioma in site of healed herpes zoster: isotopic response. Clin Exp Dermatol. 2006;31(5):714–5.

    Article  CAS  PubMed  Google Scholar 

  18. Zeng YP, Ma DL. Tufted angioma arising at a site of BCG vaccination. Eur J Dermatol. 2013;23(1):102–3.

    PubMed  Google Scholar 

  19. Choi JW, Na JI, Hong JS, Kwon SH, Byun SY, Cho KH, et al. Intractable tufted Angioma associated with Kasabach-Merritt syndrome. Ann Dermatol. 2013;25(1):129.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Yue X, Zhao X, Dai Y, Shu Q. Episode of Kasabach-Merritt phenomenon following Japanese encephalitis vaccination: case report. Vaccine. 2017;35(48):6594–7.

    Article  PubMed  Google Scholar 

  21. Ji Y, Chen S, Yang K, Xia C, Peng S. Development of Kasabach–Merritt phenomenon following vaccination: more than a coincidence? J Dermatol. 2018;45(10):1203–6.

    Article  PubMed  Google Scholar 

  22. Zhou S, Wang L, Panossian A, Anselmo D, Wu S, Venkatramani R. Refractory Kaposiform Hemangioendothelioma associated with the chromosomal translocation t(13;16)(q14;p13.3). Pediatr Dev Pathol. 2016;19(5):417–20.

    Article  PubMed  Google Scholar 

  23. Lim YH, Bacchiocchi A, Qiu J, Straub R, Bruckner A, Bercovitch L, et al. GNA14 somatic mutation causes congenital and sporadic vascular tumors by MAPK activation. Am J Hum Genet. 2016;99(2):443–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bean GR, Joseph NM, Folpe AL, Horvai AE, Umetsu SE. RecurrentGNA14 mutations in anastomosing haemangiomas. Histopathology. 2018;73(2):354–7.

    Article  PubMed  Google Scholar 

  25. Ayturk UM, Couto JA, Hann S, Mulliken JB, Williams KL, Huang AY, et al. Somatic activating mutations in GNAQ and GNA11 are associated with congenital Hemangioma. Am J Hum Genet. 2016;98(6):1271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shirley MD, Tang H, Gallione CJ, Baugher JD, Frelin LP, Cohen B, et al. Sturge-weber syndrome and port-wine stains caused by somatic mutation in GNAQ. N Engl J Med. 2013;368(21):1971–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Couto JA, Huang L, Vivero MP, Kamitaki N, Maclellan RA, Mulliken JB, et al. Endothelial Cells from Capillary Malformations Are Enriched for Somatic GNAQ Mutations. Plast Reconstr Surg. 2016;137(1):77e–82e.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O'Brien JM, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457(7229):599–602.

    Article  PubMed  CAS  Google Scholar 

  29. Joseph NM, Brunt EM, Marginean C, Nalbantoglu I, Snover DC, Thung SN, et al. Frequent GNAQ and GNA14 mutations in hepatic small vessel neoplasm. The Am J Surg Pathol. 2018;42(9):1201–7.

    Article  PubMed  Google Scholar 

  30. Couto JA, Ayturk UM, Konczyk DJ, Goss JA, Huang AY, Hann S, et al. A somatic GNA11 mutation is associated with extremity capillary malformation and overgrowth. Angiogenesis. 2017;20(3):303–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hutchings CJ, Koglin M, Olson WC, Marshall FH. Opportunities for therapeutic antibodies directed at G-protein-coupled receptors. Nat Rev Drug Discov. 2017;16(9):661.

    Article  CAS  PubMed  Google Scholar 

  32. Kimple AJ, Bosch DE, Giguere PM, Siderovski DP. Regulators of G-protein signaling and their Galpha substrates: promises and challenges in their use as drug discovery targets. Pharmacol Rev. 2011;63(3):728–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Greene AK, Goss JA. Vascular Anomalies. Plast Reconstr Surg. 2018;141(5):709e–17e.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dadras SS, Skrzypek A, Nguyen L, Shin JW, Schulz MM, Arbiser J, et al. Prox-1 promotes invasion of kaposiform hemangioendotheliomas. J Invest Dermatol. 2008;128(12):2798–806.

    Article  CAS  PubMed  Google Scholar 

  35. Glaser K, Dickie P, Dickie BH. Proliferative cells from Kaposiform Lymphangiomatosis lesions resemble mesenchyme stem cell–like Pericytes defective in vessel formation. J Pediatr Hematol Oncol. 2018;40(8):e495–504.

    Article  PubMed  Google Scholar 

  36. Flister MJ, Wilber A, Hall KL, Iwata C, Miyazono K, Nisato RE, et al. Inflammation induces lymphangiogenesis through up-regulation of VEGFR-3 mediated by NF-kappaB and Prox1. Blood. 2010;115(2):418–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Saito M, Gunji Y, Kashii Y, Odaka J, Yamauchi T, Kanai N, et al. Refractory kaposiform hemangioendothelioma that expressed vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3: a case report. J Pediatr Hematol Oncol. 2009;31(3):194–7.

    Article  PubMed  Google Scholar 

  38. Folpe AL, Veikkola T, Valtola R, Weiss SW. Vascular endothelial growth factor receptor-3 (VEGFR-3): a marker of vascular tumors with presumed lymphatic differentiation, including Kaposi's sarcoma, kaposiform and Dabska-type hemangioendotheliomas, and a subset of angiosarcomas. Mod Pathol. 2000;13(2):180–5.

    Article  CAS  PubMed  Google Scholar 

  39. Karpanen T, Egeblad M, Karkkainen MJ, Kubo H, Yla-Herttuala S, Jaattela M, et al. Vascular endothelial growth factor C promotes tumor lymphangiogenesis and intralymphatic tumor growth. Cancer Res. 2001;61(5):1786–90.

    CAS  PubMed  Google Scholar 

  40. Varney ML, Singh RK. VEGF-C-VEGFR3/Flt4 axis regulates mammary tumor growth and metastasis in an autocrine manner. Am J Cancer Res. 2015;5(2):616–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Lohela M, Bry M, Tammela T, Alitalo K. VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol. 2009;21(2):154–65.

    Article  CAS  PubMed  Google Scholar 

  42. Hsu M, Pan M, Hung W. Two birds, one stone: double hits on tumor growth and Lymphangiogenesis by targeting vascular endothelial growth factor receptor 3. Cells. 2019;8(3):270.

    Article  CAS  PubMed Central  Google Scholar 

  43. Varricchi G, Granata F, Loffredo S, Genovese A, Marone G. Angiogenesis and lymphangiogenesis in inflammatory skin disorders. J Am Acad Dermatol. 2015;73(1):144–53.

    Article  CAS  PubMed  Google Scholar 

  44. Le Cras TD, Mobberley-Schuman PS, Broering M, Fei L, Trenor CC, Adams DM. Angiopoietins as serum biomarkers for lymphatic anomalies. Angiogenesis. 2017;20(1):163–73.

    Article  PubMed  CAS  Google Scholar 

  45. Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov. 2017;16(9):635–61.

    Article  CAS  PubMed  Google Scholar 

  46. Saharinen P, Eklund L, Miettinen J, Wirkkala R, Anisimov A, Winderlich M, et al. Angiopoietins assemble distinct Tie2 signalling complexes in endothelial cell-cell and cell-matrix contacts. Nat Cell Biol. 2008;10(5):527–37.

    Article  CAS  PubMed  Google Scholar 

  47. Kim I, Kim HG, So JN, Kim JH, Kwak HJ, Koh GY. Angiopoietin-1 regulates endothelial cell survival through the phosphatidylinositol 3′-kinase/Akt signal transduction pathway. Circ Res. 2000;86(1):24–9.

    Article  CAS  PubMed  Google Scholar 

  48. Gruman A, Liang MG, Mulliken JB, Fishman SJ, Burrows PE, Kozakewich HPW, et al. Kaposiform hemangioendothelioma without Kasabach-Merritt phenomenon. J Am Acad Dermatol. 2005;52(4):616–22.

    Article  PubMed  Google Scholar 

  49. Arai E, Kuramochi A, Tsuchida T, Tsuneyoshi M, Kage M, Fukunaga M, et al. Usefulness of D2-40 immunohistochemistry for differentiation between kaposiform hemangioendothelioma and tufted angioma. J Cutan Pathol. 2006;33(7):492–7.

    Article  PubMed  Google Scholar 

  50. Debelenko LV, Perez-Atayde AR, Mulliken JB, Liang MG, Archibald TH, Kozakewich HP. D2-40 immunohistochemical analysis of pediatric vascular tumors reveals positivity in kaposiform hemangioendothelioma. Mod Pathol. 2005;18(11):1454–60.

    Article  PubMed  Google Scholar 

  51. Pollitt AY, Poulter NS, Gitz E, Navarro-Nunez L, Wang YJ, Hughes CE, et al. Syk and Src family kinases regulate C-type lectin receptor 2 (CLEC-2)-mediated clustering of podoplanin and platelet adhesion to lymphatic endothelial cells. J Biol Chem. 2014;289(52):35695–710.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Florez-Vargas A, Vargas SO, Debelenko LV, Perez-Atayde AR, Archibald T, Kozakewich HP, et al. Comparative analysis of D2-40 and LYVE-1 immunostaining in lymphatic malformations. Lymphology. 2008;41(3):103–10.

    CAS  PubMed  Google Scholar 

  53. Le Huu AR, Jokinen CH, Rubin BP, Mihm MC, Weiss SW, North PE, et al. Expression of prox1, lymphatic endothelial nuclear transcription factor, in Kaposiform hemangioendothelioma and tufted angioma. Am J Surg Pathol. 2010;34(11):1563–73.

    PubMed  Google Scholar 

  54. Ruggeri ZM, Orje JN, Habermann R, Federici AB, Reininger AJ. Activation-independent platelet adhesion and aggregation under elevated shear stress. Blood. 2006;108(6):1903–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ikeda Y, Handa M, Kawano K, Kamata T, Murata M, Araki Y, et al. The role of von Willebrand factor and fibrinogen in platelet aggregation under varying shear stress. J Clin Invest. 1991;87(4):1234–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Di Vito C, Navone SE, Marfia G, Abdel HL, Mancuso ME, Pecci A, et al. Platelets from glioblastoma patients promote angiogenesis of tumor endothelial cells and exhibit increased VEGF content and release. Platelets. 2017;28(6):585–94.

    Article  PubMed  CAS  Google Scholar 

  57. Wojtukiewicz MZ, Sierko E, Hempel D, Tucker SC, Honn KV. Platelets and cancer angiogenesis nexus. Cancer Metastasis Rev. 2017;36(2):249–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Janowska-Wieczorek A, Wysoczynski M, Kijowski J, Marquez-Curtis L, Machalinski B, Ratajczak J, et al. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int J Cancer. 2005;113(5):752–60.

    Article  CAS  PubMed  Google Scholar 

  59. Verheul HM, Jorna AS, Hoekman K, Broxterman HJ, Gebbink MF, Pinedo HM. Vascular endothelial growth factor-stimulated endothelial cells promote adhesion and activation of platelets. Blood. 2000;96(13):4216–21.

    Article  CAS  PubMed  Google Scholar 

  60. Olsson AK, Cedervall J. The pro-inflammatory role of platelets in cancer. Platelets. 2018;29(6):569–73.

    Article  CAS  PubMed  Google Scholar 

  61. Caine GJ, Lip GY, Stonelake PS, Ryan P, Blann AD. Platelet activation, coagulation and angiogenesis in breast and prostate carcinoma. Thromb Haemost. 2004;92(1):185–90.

    Article  CAS  PubMed  Google Scholar 

  62. Chan S, Cassarino DS. Rapidly enlarging “bruise” on the Back of an infant. JAMA Dermatol. 2013;149(11):1337.

    Article  PubMed  Google Scholar 

  63. Zhang G, Gao Y, Liu X. Kaposiform haemangioendothelioma in a nine-year-old boy with Kasabach-Merritt phenomenon. Br J Haematol. 2017;179(1):9.

    Article  PubMed  Google Scholar 

  64. Rodriguez V, Lee A, Witman PM, Anderson PA. Kasabach-Merritt Phenomenon. J Pediatr Hematol Oncol. 2009;31(7):522–6.

    Article  PubMed  Google Scholar 

  65. Mulliken JB, Young AE. Vascular birthmarks: hemangiomas and malformations: WB Saunders company; 1988.

    Google Scholar 

  66. San Miguel FL, Spurbeck W, Budding C, Horton J. Kaposiform hemangioendothelioma: a rare cause of spontaneous hemothorax in infancy. Review of the literature. J Pediatr Surg. 2008;43(1):e37–41.

    Article  PubMed  Google Scholar 

  67. Iwami D, Shimaoka S, Mochizuki I, Sakuma T. Kaposiform hemangioendothelioma of the mediastinum in a 7-month-old boy: a case report. J Pediatr Surg. 2006;41(8):1486–8.

    Article  PubMed  Google Scholar 

  68. Duan L, Renzi S, Weidman D, Waespe N, Chami R, Manson D, et al. Sirolimus Treatment of an Infant With Intrathoracic Kaposiform Hemangioendothelioma Complicated by Life-threatening Pleural and Pericardial Effusions. J Pediatr Hematol Oncol. 2020;42(1):74–78

  69. Oza VS, Mamlouk MD, Hess CP, Mathes EF, Frieden IJ. Role of Sirolimus in advanced Kaposiform Hemangioendothelioma. Pediatr Dermatol. 2016;33(2):e88–92.

    Article  PubMed  Google Scholar 

  70. Ji Y, Yang K, Chen S, Peng S, Lu G, Liu X. Musculoskeletal complication in kaposiform hemangioendothelioma without Kasabach–Merritt phenomenon: clinical characteristics and management. Cancer Manag Res. 2018;10:3325–31.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Keskindemirci G, Tugcu D, Aydogan G, Akcay A, Aktay AN, Er A, et al. Paravertebral and retroperitoneal vascular tumour presenting with Kasabach-Merritt phenomenon in childhood, Diagnosed with Magnetic Resonance Imaging. Case Rep Pediatr. 2015;2015:537530.

    PubMed  PubMed Central  Google Scholar 

  72. Zhu Y, Qiu G, Zhao H, Liang J, Shi X. Kaposiform hemangioendothelioma with adolescent thoracic scoliosis: a case report and review of literature. Eur Spine J. 2011;20(Suppl 2):S309–13.

    Article  PubMed  Google Scholar 

  73. Ji Y, Chen S, Li L, Yang K, Xia C, Li L, et al. Kaposiform hemangioendothelioma without cutaneous involvement. J Cancer Res and Clin. 2018;144(12):2475–84.

    Article  CAS  Google Scholar 

  74. Lindberg U, Svensson L, Hellmark T, Segelmark M, Shannon O. Increased platelet activation occurs in cystic fibrosis patients and correlates to clinical status. Thromb Res. 2018;162:32–7.

    Article  CAS  PubMed  Google Scholar 

  75. Zaldivar MM, Pauels K, von Hundelshausen P, Berres ML, Schmitz P, Bornemann J, et al. CXC chemokine ligand 4 (Cxcl4) is a platelet-derived mediator of experimental liver fibrosis. Hepatology. 2010;51(4):1345–53.

    Article  CAS  PubMed  Google Scholar 

  76. Schook CC, Mulliken JB, Fishman SJ, Alomari AI, Grant FD, Greene AK. Differential diagnosis of lower extremity enlargement in pediatric patients referred with a diagnosis of lymphedema. Plast Reconstr Surg. 2011;127(4):1571–81.

    Article  CAS  PubMed  Google Scholar 

  77. Hammill A, Mobberley-Schuman P, Adams D. Lymphoedema is a potential sequela of kaposiform haemangioendothelioma. Br J Dermatol. 2016;175(4):833–4.

    Article  CAS  PubMed  Google Scholar 

  78. Konczyk DJ, Goss JA, Maclellan RA, Greene AK. Association between extremity kaposiform hemangioendothelioma and lymphedema. Pediatr Dermatol. 2018;35(1):e92–3.

    Article  PubMed  Google Scholar 

  79. Kim DW, Chung JH, Ahn SH, Kwon T. Laryngeal kaposiform hemangioendothelioma: case report and literature review. Auris Nasus Larynx. 2010;37(2):258–62.

    Article  PubMed  Google Scholar 

  80. Wang C, Li Y, Xiang B, Li F, Chen S, Li L, et al. Successful Management of Pancreatic Kaposiform Hemangioendothelioma with Sirolimus. Pancreas. 2017;46(5):e39–41.

    Article  PubMed  Google Scholar 

  81. Leung M, Chao NS, Tang PM, Liu K, Chung KL. Pancreatic kaposiform hemangioendothelioma presenting with duodenal obstruction and kasabach-Merritt phenomenon: a neonate cured by whipple operation. European J Pediatr Surg Rep. 2014;2(1):7–9.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Gong X, Ying H, Zhang Z, Wang L, Li J, Ding A, et al. Ultrasonography and magnetic resonance imaging features of kaposiform hemangioendothelioma and tufted angioma. J Dermatol. 2019;46(10):835–42.

    Article  PubMed  Google Scholar 

  83. Drolet BA, Trenor CC, Brandão LR, Chiu YE, Chun RH, Dasgupta R, et al. Consensus-derived practice standards plan for complicated Kaposiform Hemangioendothelioma. J Pediatr. 2013;163(1):285–91.

    Article  PubMed  Google Scholar 

  84. Hu P, Zhou Z. Clinical and imaging features of Kaposiform Hemangioendothelioma. Br J Radiol. 2018;91(1086):20170798.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Ryu YJ, Choi YH, Cheon J, Kim WS, Kim I, Park JE, et al. Imaging findings of Kaposiform Hemangioendothelioma in children. Eur J Radiol. 2017;86:198–205.

    Article  PubMed  Google Scholar 

  86. Tollefson MM, Frieden IJ. Early growth of infantile hemangiomas: what parents' photographs tell us. Pediatrics. 2012;130(2):e314–20.

    Article  PubMed  Google Scholar 

  87. Krowchuk DP, Frieden IJ, Mancini AJ, Darrow DH, Blei F, Greene AK, et al. Clinical practice guideline for the Management of Infantile Hemangiomas. Pediatrics. 2019;143(1):e20183475.

    Article  PubMed  Google Scholar 

  88. Léauté-Labrèze C, Harper JI, Hoeger PH. Infantile haemangioma. Lancet. 2017;390(10089):85–94.

    Article  PubMed  Google Scholar 

  89. Mulliken JB, Enjolras O. Congenital hemangiomas and infantile hemangioma: missing links. J Am Acad Dermatol. 2004;50(6):875–82.

    Article  PubMed  Google Scholar 

  90. Nasseri E, Piram M, McCuaig CC, Kokta V, Dubois J, Powell J. Partially involuting congenital hemangiomas: a report of 8 cases and review of the literature. J Am Acad Dermatol. 2014;70(1):75–9.

    Article  PubMed  Google Scholar 

  91. Knöpfel N, Wälchli R, Luchsinger I, Theiler M, Weibel L, Schwieger BA. Congenital hemangioma exhibiting postnatal growth. Pediatr Dermatol. 2019;36(4):548–9.

    PubMed  Google Scholar 

  92. Baselga E, Cordisco MR, Garzon M, Lee MT, Alomar A, Blei F. Rapidly involuting congenital haemangioma associated with transient thrombocytopenia and coagulopathy: a case series. Br J Dermatol. 2008;158(6):1363–70.

    Article  CAS  PubMed  Google Scholar 

  93. Dompmartin A, Acher A, Thibon P, Tourbach S, Hermans C, Deneys V, et al. Association of localized intravascular coagulopathy with venous malformations. Arch Dermatol. 2008;144(7):873–7.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Dompmartin A, Vikkula M, Boon LM. Venous malformation: update on aetiopathogenesis, diagnosis and management. Phlebology. 2010;25(5):224–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Boland JM, Tazelaar HD, Colby TV, Leslie KO, Hartman TE, Yi ES. Diffuse pulmonary lymphatic disease presenting as interstitial lung disease in adulthood: report of 3 cases. Am J Surg Pathol. 2012;36(10):1548–54.

    Article  PubMed  Google Scholar 

  96. Croteau SE, Kozakewich HP, Perez-Atayde AR, Fishman SJ, Alomari AI, Chaudry G, et al. Kaposiform lymphangiomatosis: a distinct aggressive lymphatic anomaly. J Pediatr. 2014;164(2):383–8.

    Article  PubMed  Google Scholar 

  97. Safi F, Gupta A, Adams D, Anandan V, McCormack FX, Assaly R. Kaposiform lymphangiomatosis, a newly characterized vascular anomaly presenting with hemoptysis in an adult woman. Ann Am Thorac Soc. 2014;11(1):92–5.

    Article  PubMed  Google Scholar 

  98. Ji Y, Chen S, Peng S, Xia C, Li L. Kaposiform lymphangiomatosis and kaposiform hemangioendothelioma: similarities and differences. Orphanet J Rare Dis. 2019;14(1):165.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Kato H, Ozeki M, Fukao T, Matsuo M. MR imaging findings of vertebral involvement in Gorham-stout disease, generalized lymphatic anomaly, and kaposiform lymphangiomatosis. Jpn J Radiol. 2017;35(10):606–12.

    Article  PubMed  Google Scholar 

  100. Barclay SF, Inman KW, Luks VL, McIntyre JB, Al-Ibraheemi A, Church AJ, et al. A somatic activating NRAS variant associated with kaposiform lymphangiomatosis. Genet Med. 2019;21(7):1517–24.

    Article  CAS  PubMed  Google Scholar 

  101. Tower RL. Kaposiform haemangioendothelioma: new insights and old problems. Br J Dermatol. 2018;179(2):253–4.

    CAS  PubMed  Google Scholar 

  102. Freixo C, Ferreira V, Martins J, Almeida R, Caldeira D, Rosa M, et al. Efficacy and safety of sirolimus in the treatment of vascular anomalies: a systematic review. J Vasc Surg. 2019. https://doi.org/10.1016/j.jvs.2019.06.217.

  103. Haisley-Royster C, Enjolras O, Frieden IJ, Garzon M, Lee M, Oranje A, et al. Kasabach-Merritt phenomenon: a retrospective study of treatment with vincristine. J Pediatr Hematol Oncol. 2002;24(6):459–62.

    Article  PubMed  Google Scholar 

  104. Fahrtash F, McCahon E, Arbuckle S. Successful treatment of Kaposiform Hemangioendothelioma and tufted Angioma with vincristine. J Pediatr Hematol Oncol. 2010;32(6):506–10.

    Article  PubMed  Google Scholar 

  105. Wang Z, Li K, Yao W, Dong K, Xiao X, Zheng S. Steroid-resistant kaposiform hemangioendothelioma: a retrospective study of 37 patients treated with vincristine and long-term follow-up. Pediatr Blood Cancer. 2015;62(4):577–80.

    Article  CAS  PubMed  Google Scholar 

  106. Liu X, Li J, Qu X, Yan W, Zhang L, Zhang S, et al. Clinical outcomes for systemic corticosteroids versus vincristine in treating Kaposiform Hemangioendothelioma and tufted Angioma. Medicine. 2016;95(20):e3431.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Tlougan BE, Lee MT, Drolet BA, Frieden IJ, Adams DM, Garzon MC. Medical Management of Tumors Associated with Kasabach-Merritt Phenomenon. J Pediatr Hematol Oncol. 2013;35(8):618–22.

    Article  PubMed  Google Scholar 

  108. Peng S, Yang K, Xu Z, Chen S, Ji Y. Vincristine and sirolimus in the treatment of kaposiform haemangioendothelioma. J Paediatr Child H. 2019;55(9):1119–24.

    Article  Google Scholar 

  109. Tan X, Zhang J, Zhou S, Liu Z, Zhang T, Xia J. Successful management of steroid-resistant vascular tumors associated with the Kasabach-Merritt phenomenon using sirolimus. J Dermatol. 2018;45(5):580–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kai L, Wang Z, Yao W, Dong K, Xiao X. Sirolimus, a promising treatment for refractory Kaposiform hemangioendothelioma. J Cancer Res Clin. 2014;140(3):471–6.

    Article  CAS  Google Scholar 

  111. Lopez V, Marti N, Pereda C, Martin JM, Ramon D, Mayordomo E, et al. Successful management of Kaposiform hemangioendothelioma with Kasabach-Merritt phenomenon using vincristine and ticlopidine. Pediatr Dermatol. 2009;26(3):365–6.

    Article  PubMed  Google Scholar 

  112. Liu XH, Li JY, Qu XH, Yan WL, Zhang L, Yang C, et al. Treatment of kaposiform hemangioendothelioma and tufted angioma. Int J Cancer. 2016;139(7):1658–66.

    Article  CAS  PubMed  Google Scholar 

  113. George ME, Sharma V, Jacobson J, Simon S, Nopper AJ. Adverse effects of systemic Glucocorticosteroid therapy in infants with Hemangiomas. Arch Dermatol. 2004;140(8):963–9.

    Article  CAS  PubMed  Google Scholar 

  114. Ji Y, Chen S, Xiang B, Li K, Xu Z, Yao W, et al. Sirolimus for the treatment of progressive kaposiform hemangioendothelioma: a multicenter retrospective study. Int J Cancer. 2017;141(4):848–55.

    Article  CAS  PubMed  Google Scholar 

  115. Boccara O, Puzenat E, Proust S, Leblanc T, Lasne D, Hadj-Rabia S, et al. The effects of sirolimus on Kasabach-Merritt phenomenon coagulopathy. British J Dermatol. 2018;178(2):e114–6.

    Article  CAS  Google Scholar 

  116. Zhang G, Chen H, Gao Y, Liu Y, Wang J, Liu XY. Sirolimus for treatment of Kaposiform haemangioendothelioma with Kasabach-Merritt phenomenon: a retrospective cohort study. Brit J Dermatol. 2018;178(5):1213–4.

    Article  CAS  Google Scholar 

  117. Triana P, Dore M, Cerezo V, Cervantes M, Sánchez A, Ferrero M, et al. Sirolimus in the treatment of vascular anomalies. Eur J Pediatr Surg. 2017;27(01):086–90.

    Google Scholar 

  118. Tasani M, Ancliff P, Glover M. Sirolimus therapy for children with problematic kaposiform haemangioendothelioma and tufted angioma. Brit J Dermatol. 2017;177(6):e344–6.

    Article  CAS  Google Scholar 

  119. Wang H, Duan Y, Gao Y, Guo X. Sirolimus for vincristine-resistant Kasabach-Merritt phenomenon: report of eight patients. Pediatr Dermatol. 2017;34(3):261–5.

    Article  PubMed  Google Scholar 

  120. Adams DM, Trenor CC, Hammill AM, Vinks AA, Patel MN, Chaudry G, et al. Efficacy and safety of Sirolimus in the treatment of complicated vascular anomalies. Pediatrics. 2016;137(2):e20153257.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Nadal M, Giraudeau B, Tavernier E, Jonville-Bera A, Lorette G, Maruani A. Efficacy and safety of mammalian target of Rapamycin inhibitors in vascular anomalies: a systematic review. Acta Dermato Venereologica. 2016;96(4):448–52.

    Article  CAS  PubMed  Google Scholar 

  122. Wang Z, Li K, Dong K, Xiao X, Zheng S. Successful treatment of Kasabach-Merritt phenomenon arising from Kaposiform Hemangioendothelioma by Sirolimus. J Pediatr Hematol Oncol. 2015;37(1):72–3.

    Article  PubMed  Google Scholar 

  123. Lackner H, Karastaneva A, Schwinger W, Benesch M, Sovinz P, Seidel M, et al. Sirolimus for the treatment of children with various complicated vascular anomalies. Eur J Pediatr. 2015;174(12):1579–84.

    Article  CAS  PubMed  Google Scholar 

  124. Blatt J, Stavas J, Moats-Staats B, Woosley J, Morrell DS. Treatment of childhood kaposiform hemangioendothelioma with sirolimus. Pediatr Blood Cancer. 2010;55(7):1396–8.

    Article  PubMed  Google Scholar 

  125. Russell TB, Rinker EK, Dillingham CS, Givner LB, McLean TW. Pneumocystis Jirovecii pneumonia during Sirolimus therapy for Kaposiform Hemangioendothelioma. Pediatrics. 2018;141(Suppl 5):S421.

    Article  PubMed  Google Scholar 

  126. Ying H, Qiao C, Yang X, Lin X. A case report of 2 Sirolimus-related deaths among infants with Kaposiform Hemangioendotheliomas. Pediatrics. 2018;141(Suppl 5):S425–9.

    Article  PubMed  Google Scholar 

  127. Mariani LG, Schmitt IR, Garcia CD, Kiszewski AE. Low dose sirolimus treatment for refractory tufted angioma and congenital kaposiform hemangioendothelioma, both with Kasabach-Merritt phenomenon. Pediatr Blood Cancer. 2019;66(8):e27810.

    Article  PubMed  Google Scholar 

  128. Zhu J, Wu J, Frizell E, Liu SL, Bashey R, Rubin R, et al. Rapamycin inhibits hepatic stellate cell proliferation in vitro and limits fibrogenesis in an in vivo model of liver fibrosis. Gastroenterology. 1999;117(5):1198–204.

    Article  CAS  PubMed  Google Scholar 

  129. Wu MJ, Wen MC, Chiu YT, Chiou YY, Shu KH, Tang MJ. Rapamycin attenuates unilateral ureteral obstruction-induced renal fibrosis. Kidney Int. 2006;69(11):2029–36.

    Article  CAS  PubMed  Google Scholar 

  130. Burleigh A, Kanigsberg N, Lam JM. Topical rapamycin (sirolimus) for the treatment of uncomplicated tufted angiomas in two children and review of the literature. Pediatr Dermatol. 2018;35(5):e286–90.

    Article  PubMed  Google Scholar 

  131. Zhang X, Yang K, Chen S, Ji Y. Tacrolimus ointment for the treatment of superficial kaposiform hemangioendothelioma and tufted angioma. J Dermatol. 2019;46(10):898–901.

    Article  CAS  PubMed  Google Scholar 

  132. Paller AS, Eichenfield LF, Kirsner RS, Shull T, Jaracz E, Simpson EL. Three times weekly tacrolimus ointment reduces relapse in stabilized atopic dermatitis: a new paradigm for use. Pediatrics. 2008;122(6):e1210–8.

    Article  PubMed  Google Scholar 

  133. Moimeaux V, Taieb A, Legrain V, Meraud JP, Jimenez M, Choussat A, et al. Aspirin-ticlopidin in Kasabach-Merritt syndrome. Lancet. 1992;340(8810):55.

    Article  CAS  PubMed  Google Scholar 

  134. Wu HW, Wang X, Zhang L, Zhao HG, Wang YA, Su LX, et al. Interferon-alpha therapy for refractory kaposiform hemangioendothelioma: a single-center experience. Sci Rep. 2016;6:36261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Hermans DJ, van Beynum IM, van der Vijver RJ, Kool LJ, de Blaauw I, van der Vleuten CJ. Kaposiform hemangioendothelioma with Kasabach-Merritt syndrome: a new indication for propranolol treatment. J Pediatr Hematol Oncol. 2011;33(4):e171–3.

    Article  CAS  PubMed  Google Scholar 

  136. Chiu YE, Drolet BA, Blei F, Carcao M, Fangusaro J, Kelly ME, et al. Variable response to propranolol treatment of kaposiform hemangioendothelioma, tufted angioma, and Kasabach-Merritt phenomenon. Pediatr Blood Cancer. 2012;59(5):934–8.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Filippi L, Tamburini A, Berti E, Perrone A, Defilippi C, Favre C, et al. Successful propranolol treatment of a Kaposiform Hemangioendothelioma apparently resistant to propranolol. Pediatr Blood Cancer. 2016;63(7):1290–2.

    Article  CAS  PubMed  Google Scholar 

  138. Barlow CF, Priebe CJ, Mulliken JB, Barnes PD, Mac DD, Folkman J, et al. Spastic diplegia as a complication of interferon Alfa-2a treatment of hemangiomas of infancy. J Pediatr. 1998;132(3 Pt 1):527–30.

    Article  CAS  PubMed  Google Scholar 

  139. Wang P, Zhou W, Tao L, Zhao N, Chen XW. Clinical analysis of Kasabach-Merritt syndrome in 17 neonates. BMC Pediatr. 2014;14:146.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients and their parents for their co-operation and support, and for providing consent for publication.

Funding

This work was supported by grants from the National Natural Science Foundation of China (Grants Nos. 81400862 and 81401606), the Key Project in the Science & Technology Program of Sichuan Province (Grant No. 2019YFS0322), the Science Foundation for The Excellent Youth Scholars of Sichuan University (Grant No. 2015SU04A15), and the 1·3·5 Project for Disciplines of Excellence-Clinical Research Incubation Project of West China Hospital of Sichuan University (2019HXFH056).

Author information

Authors and Affiliations

Authors

Contributions

YJ and SYC drafted the manuscript. YJ, SYC, KYY, CCX and LL were involved in the clinical management of these patients and collected clinical details of this study. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yi Ji or Siyuan Chen.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Ethics Committee of the West China Hospital of Sichuan University. Written informed consent was obtained from the patients’ parents, according to the provisions of the Declaration of Helsinki.

Consent for publication

Written informed consent for publication this study was obtained from the patients’ parents. Copies of the signed informed consent forms are available for review by the Series Editor of Journal of Hematology & Oncology.

Competing interests

The authors declare that they have no competing interests, either financial or non-financial, that could be perceived as prejudicing the impartiality of the research reported.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ji, Y., Chen, S., Yang, K. et al. Kaposiform hemangioendothelioma: current knowledge and future perspectives. Orphanet J Rare Dis 15, 39 (2020). https://doi.org/10.1186/s13023-020-1320-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13023-020-1320-1

Keywords