Next Article in Journal
Lizard Blastema Organoid Model Recapitulates Regenerated Tail Chondrogenesis
Next Article in Special Issue
The Hypothesis of the Prolonged Cell Cycle in Turner Syndrome
Previous Article in Journal
Actin Filament in the First Cell Cycle Contributes to the Determination of the Anteroposterior Axis in Ascidian Development
Previous Article in Special Issue
Altered Cogs of the Clock: Insights into the Embryonic Etiology of Spondylocostal Dysostosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic and Molecular Determinants of Lymphatic Malformations: Potential Targets for Therapy

1
Division of Pediatric, Thoracic and Fetal Surgery, University of California Davis Medical Center, Sacramento, CA 95817, USA
2
Center for Surgical Bioengineering, Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
3
Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
*
Authors to whom correspondence should be addressed.
Submission received: 16 December 2021 / Revised: 28 January 2022 / Accepted: 1 February 2022 / Published: 8 February 2022
(This article belongs to the Special Issue Advances in Development: Focus on Rare Congenital Diseases)

Abstract

:
Lymphatic malformations are fluid-filled congenital defects of lymphatic channels occurring in 1 in 6000 to 16,000 patients. There are various types, and they often exist in conjunction with other congenital anomalies and vascular malformations. Great strides have been made in understanding these malformations in recent years. This review summarize known molecular and embryological precursors for lymphangiogenesis. Gene mutations and dysregulations implicated in pathogenesis of lymphatic malformations are discussed. Finally, we touch on current and developing therapies with special attention on targeted biotherapeutics.

1. Introduction

The lymphatic system works to maintain extracellular fluid homeostasis, aid in fat absorption and transportation, and optimize immunity. Lymphatic malformations are congenital low flow vascular malformations of the lymphatic vessels. They can be differentiated into three general categories: (1) abnormalities of lymph vessels and nodes leading to inadequate clearance of lymph and subsequent lymphedema, (2) mass lesion composed of cysts, and (3) disordered circulation of chyle in central conducting lymphatic channels [1]. This review focuses on the genetic and molecular pathogenesis of cystic mass lesions. While commonly referred to as cystic hygroma, lymphatic anomalies have extremely varied presentations other than simple cysts and may be combined with malformations of capillaries, veins, and arteries.
The overall incidence of lymphatic malformations ranges from 1 in 6000 to 1 in 16,000, with no predilection for sex or race [2]. Seventy-five percent of the cases occur primarily in the lymphatic-rich head and neck region but can exist in any part of the body, including the axilla, mediastinum, and abdomen [3]. According to the International Society for the Study of Vascular Anomalies (ISSVA), lymphatic malformations are classified by size, with discrete cysts more than 1–2 cm referred to as macrocystic, and those smaller than 1–2 cm as microcystic [4]. The lesions can vary from focal and well-defined cystic masses to infiltrative and poorly defined multicystic lesions. On histology, they are seen to be lined by single layer of flattened endothelial cells with lymphatic phenotype staining with prospero homeobox protein 1 (Prox1) and podoplanin D2-40. For clinical diagnosis, the hematoxylin and eosin-stained histology is sufficient. They also have abnormally formed smooth muscle resulting in fibrovascular vessel walls of varying thickness (Figure 1) [5]. Their lumens are filled with eosinophilic proteinaceous lymph fluid, which allows it to be easily seen on imaging. With advancements in prenatal care and imaging, lymphatic malformations are being more readily classified and diagnosed on prenatal ultrasound. Respiratory distress, recurrent infections, and deformity are the main indications for treatment. Treatment is based on surgical excision, although sclerotherapy and pharmacologic treatments, including use of immunosuppressive agents, are being studied.

2. Lymphatic Development

While the exact embryonic origin of these malformations is unclear, they are believed to be the result of developmental defects of dilated lymphatic channels [6]. There are multiple theories on lymphatic development. Initially proposed by Sabin, in the “centrifugal theory,” primordial lymphatic sacs were thought to sprout from developing central veins and propagate peripherally [7]. This model was further supported by experiments from Lewis that demonstrated detachment of portions of sub-cardinal vein from the venous system to form lymphatic ducts in rabbits [8]. McClure proposed a different theory in which lymphatic vessels form by coalescing isolated spaces in the mesenchyme, which subsequently transform into lymphatic endothelial cells (LECs) [9]. These LECs develop into a primitive lymphatic network and connect to the venous system in a “centripetal” model in which the lymphatic system develops peripherally to centrally.
Recent work in different model organisms has demonstrated variation in the origin of LECs. Avian and xenopus models have demonstrated dual origin of venous endothelial cells and mesenchymal lymphangioblasts, combining both theories [10,11]. Live imaging zebrafish embryos allowed in vivo tracking of LEC progenitors originating in the posterior cardinal vein and migrating dorsally to the thoracic duct, thus supporting Sabin’s centrifugal theory [12]. Okuda et al. demonstrated that in zebrafish, the facial lymphatic network develops through sprouting from the common cardinal vein and additional populations of lymphangioblasts recruited to this main sprout [13]. However, the formation of lymphatic networks differs between mammals and zebrafish, with lymphangiogenesis and angiogenesis temporally separated; in mice, lymphatic structure forms after lymphatic specification following angiogenesis, while in zebrafish, venous sprouts give rise to both intersegmental veins and lymphatic precursors [14]. As such, prox1 signaling to specify LEC in mouse is integral for lymphangiogenesis, but dispensable in zebrafish. Additionally, lineage-tracing studies in mice demonstrated that LECs were derived from venous origins. Particularly, in hematopoietic deficient mice, LECs from venous-derived lymph sacs proliferated into lymphatic vasculature; hematopoietic cells did not contribute to development of the lymphatic system [15].
Adding to this complexity, organ-specific lymphatic vasculature pools progenitor cells from different populations. In the murine model, skin, cervical, and thoracic lymphatic vessels were formed from Tie2 lineage venous derived LEC progenitors, while most lumbar and cardiac lymphatic vessels were formed by coalescence of isolated Tie2 negative non-venous LEC progenitors [16,17]. Mesenteric lymphatic vessels were seen to originate from two distinct sources: Tie2 positive venous endothelial cells and PDGFB and c-Kit positive hemogenic endothelial cells [18]. Lymph nodes have been suggested to arise from nestin positive precursors [19]. However, the results of these experiments with lineage tracing in mice should be interpreted with caution. The efficiency of Cre-mediated recombination may vary, therefore introducing a chance for misinterpretation of biological heterogeneity as incomplete recombination. Additionally, there may be other markers that are more specific for lymphatic lineage and able to mark cells earlier in development.
An unknown signal currently triggers polarized expression of homeobox gene Prox1 in a subpopulation of endothelial cells, committing them to lymphatic lineage and promoting budding [20]. Underexpression or overexpression of Prox1 in mice were lethal secondary to lack of lymphovascular development and edema from increased permeability, respectively [20,21]. Prox1 expression in mice occurs at E9.5 in subpopulation in cardinal vein until E14.5, and is maintained by nuclear hormone receptor Coup-TFII and SOX18 [22]. SOX18, SRY-related HMG domain family of transcription factors that bind to Prox1 proximal promoter, is integral for early maintenance of Prox1 but is not past E14.5 during lymphangiogenesis [23]. Lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1), podoplanin, and vascular endothelial growth factor receptor 3 (VEGFR3) are upregulated by these PROX1 positive progenitors [24,25,26].
Early LECs start producing secondary lymphoid chemokines such as vascular endothelial growth factor c (VEGFC) and increase expression of VEGFR3 [27]. VEGFC binds to VEGFR3, mediated by neuropilin 2 (NRP2), then activates phosphoinositide 3-kinase (PI3K)/AKT and RAF1/MEK/ERK signaling pathways, which induces LEC migration and SOX18 expression, respectively [28,29,30,31]. VEGFC is integral for initial lymphatic development, as seen by the lack of lymph vessels sprouting from committed endothelial cells in VEGFC null mice [32]. VEGFC and VEGFD, which also activates VEGFR3, need to be activated to their mature form by proteolytic cleavage [33,34,35]. Of note, VEGFD is not required for lymphangiogenesis, unlike VEGFC [36]. Collagen- and calcium-binding epidermal growth factor domains 1 (CCBE1) enhances the cleavage of VEGFC by A disintegrin and metalloprotease with thrombospondin motifs 3 (ADAMTS3) [37]. CCBE1 knock-out mice have shown similar phenotype as VEGFC null mice, with specified lymphatic progenitors failing to migrate from cardinal vein [38]. Finally, angiopoietin 2 (ANGPT2) and its tyrosine kinase receptor TIE2 are involved with maturation and patterning of these lymphatic vessels continuing postnatally [39]. Of note, small populations of Prox1 positive LECs remaining in the veins form lymph-venous valves [40].
First, morphologically visible primordial lymph sacs in humans are seen at 6 to 7 weeks gestational age, consisting of paired jugulo-axillary (which gives rise to lymph vessels of head, neck and arms) and lumbo-iliac (gives rise to lower half of the body) lymph sacs, and singular retroperitoneal lymph sac and chylocyst [41]. Starting at weeks 6.5 to 7, endothelial buds (lymphatic primordia) develop in the jugular region after arterial and venous development. These buds unite to form plexuses which further develop into paired jugular sacs and axillary lymph sacs [42]. By 8 weeks, both axillary and jugular sacs enlarge and form single communication to the internal jugular vein and to each other on either side. Jugulo-axillary lymph sacs continue to grow through week 9 predominantly in cranial and dorsolateral directions to eventually reach the subcutaneous area of the posterior neck compartment. Rudimentary bilateral thoracic ducts are seen to anastomose with left jugulo-axillary lymph sac and lumbar lymph plexuses. Lymphatic valves are starting to be formed at this time. All lymphatic primordia fuse and one continuous system is formed at about 10 weeks gestational age. Continuous peripheral development from these central primordial occurs in all trunks, and these primordial lymph sacs become less prominent as lymphatic branches continue to develop.
In the end, the lymphatic system develops into a network of thin, permeable, blind-ended capillaries that drain into larger collecting vessels complete with valves. Lymphatic capillaries consist of a single layer of LECs that lack a continuous basement membrane [43]. Initial lymphatics have discontinuous button-like junctions between endothelial cells that allow for easy uptake of interstitial fluid and macromolecules [44]. Lymph, after uptake by initial lymphatics, flows through collecting lymphatic vessels characterized by zipper-like junctions [44]. Valves allow for unidirectional transport of lymph through these collecting vessels, into lymph nodes to larger collecting ducts and back to venous circulation. Collecting vessels and onwards have basement membrane and are surrounded by pericytes and smooth muscle cells. Additional skeletal muscle contractions, arterial pulsation, and gravity provide propulsive force for lymph transport.

3. Pathogenesis

Few major theories on cystic lymphatic malformation pathogenesis have been proposed (Figure 2). One theory assumes McClure’s centripetal theory of mesenchymal origin of lymphangiogenesis and suggests primordial lymphatic sacs fail to fuse with venous system [45]. This results in isolated lymphatic canals that dilate and form cystic malformations. Zadvinskis et al. and van der Putte both demonstrated cystic lesions in the neck have missing lympho-venous connections in jugulo-axillary and jugulo-subclavian areas [42,46]. Others have assumed that tissue abnormally sequestered early in embryogenesis was the culprit. These fail to join normal central lymphatic channels and become malformations [47]. Abnormal budding of lymphatic structures may be another etiology of lymphatic malformations. Aberrant buds lose connections with lymphatic primordia and canalize to form lymph filled cysts. These structures branch and grow in a disorderly manner, forming cysts without lymphatic drainage [41]. More recently, efforts are being taken to elucidate molecular mechanisms behind this aberrant growth.

3.1. Clinical Syndromes

While lymphatic malformations can exist as solitary lesions, many of them are associated with specific syndromes and genetic mutations. Generalized lymphatic anomaly (GLA) and Gorham–Stout disease (GSD) are complicated lymphatic malformations that involve multiple body sites.
GLA most often appears in childhood with proliferation of normal mature lymphatic ducts and the formation of numerous cysts in any organ of the body. While the cysts themselves are benign, the size, critical locations, and secondary infections can cause significant morbidity and mortality [48]. Kaposiform lymphangiomatosis has been recently recognized as a subtype of GLA, with foci of spindle endothelial cells amid a background of malformed lymphatic channels [49]. It affects multiple organs, with thoracic involvement being most common. Extrathoracic disease can manifest in bone including in the extremities, and in abdominal viscera. Mortality is high with cardio-respiratory failure from thoracic disease, with only 34% overall survival, and mean interval between diagnosis and death of less than 3 years [50].
GSD is a very rare disorder characterized by proliferation of capillary-sized vascular channels, including lymphatic vessels, in bone, and nearby soft tissue resulting in progressive osteolysis. While GSD can develop in any musculoskeletal site, it most commonly occurs in the shoulder and the pelvic girdle. Skull, humerus, sternum, ribs, femur, and spine have all been seen to be affected [51,52]. Morbidity and mortality is high secondary to osteomyelitis, spinal cord involvement including paraplegia from vertebral lesions, and chylothorax from extension into pleural cavity or thoracic duct [52].
Additionally, there are mixed venous, arterial, and lymphatic vascular malformations that are associated with other anomalies. These include Klippel–Trenaunay syndrome (KTS) with capillary, venous, and lymphatic malformation and limb overgrowth, CLOVES (congenital lipomatous overgrowth, vascular malformations, epidermal nevis, spinal/skeletal anomalies/scoliosis) syndrome with capillary, venous, lymphatic, and arterio-venous malformations and lipomatous overgrowth, and CLAPO (capillary vascular malformation of the lower lip, lymphatic malformations of the head and neck, asymmetry and partial or generalized overgrowth) syndrome with lower lip capillary malformation, cervical lymphatic malformation, and asymmetrical somatic overgrowth as part of PIK3CA-related overgrowth spectrum (PROS) which results from somatic PIK3CA activation mutation. Additionally, Proteus syndrome has capillary, venous, and lymphatic malformations, and asymmetrical somatic overgrowth from AKT1 mutation.

3.2. Genetic Mutations

In certain cases of lymphatic malformation, its cause can be easily determined if it is a feature of a greater condition like KTS or CLOVES syndrome in PROS. However, pinning down the genetic source of solitary malformations has proven challenging as there is no historically known cause. Research in the past decade has helped elucidate various genes that might be involved in the development of lymphatic malformations and whether they are isolated or a part of a greater medical condition.
One such gene of interest is the PIK3CA gene, which encodes for the catalytic subunit of PI3K. The PI3K/AKT pathway is integral in lymphatic development by inducing LEC migration. As previously mentioned, PIK3CA somatic mosaic activating mutation, particularly in the catalytic p110α subunit, is seen in a number of mixed vascular malformations [53,54]. Using samples of affected tissue from Boston Children’s and Seattle Children’s hospitals, Luk et al.’s analysis revealed five activating mutations in the gene that were present at low frequencies (<10%) in patients with isolated lymphatic malformations, KTS, CLOVES, and fibro-adipose vascular anomaly (FAVA) [55]. These five mutations accounted for ~80% of cases and considering both the somatic mosaic nature of their results and proliferative nature of the PI3K pathway, this may reflect a biological mechanism where mutant cells recruit wild-type cells over the course of the overgrowth process [55,56]. Recent studies from Le Cras et al. demonstrate that LECs from patients with capillary lymphatic venous malformations with PIK3CA mutations recapitulate the patient’s lesion when injected into immunocompromised mice in a xenograft model [54]. Additionally, there may be a mutation in PIK3CD that acts in the PI3K pathway in conjunction with PIK3CA. Wang et al. demonstrated novel PIK3CD mutation in human samples of lymphatic malformation, and in vitro studies in human umbilical vein endothelial cells with these mutations demonstrated increased cell proliferation and hyperactivation of mTOR pathway [57].
AKT1 is another gene in the PI3K/AKT signaling pathway that has been seen with activating mutation associated with Proteus syndrome. As previously discussed AKT stimulates LEC migration during lymphangiogenesis. Lindhurst et al. performed DNA exome sequencing of varying samples from 29 patients with Proteus syndrome and found that 89% of tested patient samples had somatic activating mutation in the AKT1 gene [58]. Importantly, they demonstrated somatic mosaicism ranging from 1–50% of mutant alleles in different tissues and cell lines of patients with varying levels of phosphorylation. At the current time, it is unclear during which stage of development the somatic mutation occurs in embryo. Additionally, there is a wide variability in lymphatic presentation of Proteus syndrome, and there is no clear association between proportion of mutant alleles to lymphatic malformation phenotype. However, there have been associations between the frequency of cutaneous manifestations, including lymphatic malformations, with clinical severity of proteus syndrome and extra-cutaneous manifestations [59]. An additional case report describe novel somatic mosaic heterozygous duplication encompassing exon 3 to 15 of AKT1 gene in a patient with large left sided cervical cystic lymphatic malformation [60].
Similarly, the Burrows et al. case study of a patient with Parkes–Weber syndrome (PWS) uncovered the RASA1 gene as a possible genetic source for the phenotypic expression of lymphedema. In adult mice studies, the gene was revealed to be an integral part in regulating the Ras signaling pathway, as its overactivity leads to hyperplasia of the lymphatic vasculature [61]. Utilizing whole-exome sequencing and Sanger sequencing, they confirmed a frameshift mutation in RASA1 in the patient and his father. Additionally, use of investigational near-infrared fluorescence lymphatic imaging (NIRFLI) in concert with radiographic lymphangiography further affirmed the association between RASA1 and lymphatic abnormalities. Aberrant lymphatics were observed in both the PWS patient and Rasa1 knockout mice, suggesting the mutation in RASA1 and subsequent irregular Ras activity could explain such lymphatic vessel anomalies [56]. While these studies successfully unveil possible genes for further investigation, it has yet to be determined why such mutations create an isolated malformation in one individual as compared to the presence of a malformation as part of a greater syndrome in others.
Broadly, the findings presented from these various studies do not point to a definitive cause of lymphatic malformations, but rather have revealed new genetic targets to explore which can be used to investigate more effective treatments for all types of lymphatic malformations. In addition, many of these mutations including ones in the PI3K pathway are significant contributors in vascular malformations and have been identified as potential targets for pharmaceutical therapies [62]. Although these may acquire mutations at different stages of development, there are clear parallels between lymphangiogenesis and angiogenesis. This lends further evidence to using these genes as potential targets for future therapies for lymphatic malformations as well.

3.3. Gene Dysregulation

Gene dysregulation can also play a role in pathogenesis. Another gene being closely studied is VEGFC. In a PIK3CA driven microcystic lymphatic malformation mouse model, VEGFC/VEGFR3 signaling was integral in the growth of the lesion [63]. Furthermore, Han et al. investigated the relationship between Hypoxia-inducible factor-1α (HIF-1α) and VEGF receptor 3 (VEGFR-3) using samples obtained from 20 patients with microcystic, macrocystic, or mixed lymphatic malformation [64]. In brief, HIF-1α binds to HIF-1β under hypoxic conditions to trigger the HIF-1 cascade to mediate cells and their metabolism while VEGFR3 is localized to LECs and associated with lymphangiogenesis. The results indicate that both HIF-1α and VEGFR3 were upregulated in patient samples compared with normal tissues. Additionally, Han and collaborators engineered a stable, HIF-1α-overexpressing human LEC (HLEC) cell line which had increased cell migration and colony formation of HIF-1α-overexpression cells compared to control HLECs [64]. Previous studies have shown increased expression of VEGFR3 and VEGFC in various cancer cell lines under a hypoxic environment [65,66,67], and combined with the results from Han et al., suggest that the genes encoding for HIF-1α and VEGFR3 could also be involved in the malformation of lymphatic vessels leading to lymphatic malformation.
Gomez–Acevedo et al. analyzed tissue samples from 18 pediatric patients—six microcystic, seven macrocystic, and five normal controls—to determine the underlying genetic and physiologic differences between groups. Through transcriptome analysis, they discovered 426 differentially expressed genes of which 192 and 234 were upregulated in microcystic and macrocystic samples, respectively [68]. Among the overexpressed genes in microcystic samples is eukaryotic initiation factor 4A-1 (EIF4A1), which encodes for the RNA helicase subunit of the eukaryotic initiation factor 4F (eIF4F) complex [69]. As a result, proteins such as c-Myc and VEGF are produced, and in turn, promote cell survival, proliferation, and angiogenesis which could account for the abnormalities in lymph vessels. Additionally, the upregulation of metastatic suppressor NME1 points to limitations in cell spreading and motility and may explain the condensed, fibrous morphology of microcystic lymphatic malformations [68]. While the microcystic-related genes demonstrate proliferative mechanisms and imitate oncogenic processes, the upregulated genes for macrocystic samples are related to cell differentiation, embryogenesis, adipogenesis, cell adhesion, and adaptation to hypoxia. Upregulation of microfibril-associated glycoprotein 2 (MFAP2), VEGFB, and angiopoietin-like 2 (ANGPTL2) denote angiogenic events related to cell sprouting and sprout formation [68]. Particularly of interest is the abnormal expression of extracellular matrix protein dermatopontin (DPT) which modulates transforming growth factor beta (TGF-β). In conjunction with an increase in TGF-β3, its upregulation indicates that macrocystic lymphatic malformations may arise due to improper regulation and pruning of developing lymphatic networks [68]. The transcriptome differences discovered in this study illuminate differences in genetic causes and mechanisms that should be further investigated to better understand the formation of microcystic and macrocystic lymphatic malformations. While these dysregulated pathways need further studies to demonstrate causality, this is a promising area of study for potential therapeutic targets.

4. Clinical Implications

Most lymphatic malformations persist postnatally, with spontaneous regression rates between 2.3% to 41% [70]. While some minimally symptomatic lesions can be observed, most will undergo treatment for aesthetic or functional reasons.

4.1. Current Surgical Treatment

Surgical excision is the mainstay of treatment for lymphatic malformation, particularly in lesions causing functional disability secondary to compression. Airway obstruction, dysphagia and problematic bleeding should be addressed promptly, and with excision. For large prenatally diagnosed cervical lymphatic malformation, ex-utero intrapartum therapy (EXIT) is indicated to secure the airway [71]. While small or macrocystic lesions in general can be readily excised, lesions of larger size, diffuse and microcystic in nature, or involving surrounding structures such as nerves or blood vessels become very complicated and result in incomplete removal. In one case series of 63 patients, 14% had recurrence of macrocystic cervicofacial lymphatic malformation after excision, and associated factors for recurrence included bilaterality and advanced staging [72]. Cosmetic appearance, impaired functionality following excision, and scarring, in addition to other potential complications, including fistulization, infection, and dehiscence, need to be kept in mind. Carbon dioxide laser and radiofrequency ablation have been used intraoperatively as an adjunct for intraoral disease [73,74].
Percutaneous treatment with injection sclerotherapy is another option for macrocystic lymphatic malformations. Introduction of sclerosing agent induces endothelial inflammation leading to secondary occlusion, fibrosis and contraction of the structure. Picibanil (OK-432), doxycycline, and bleomycin are commonly used sclerosing agents. One randomized control study demonstrated substantial response to picibanil sclerotherapy in 94% of patients with macrocystic disease and 63% in mixed macrocystic and microcystic disease [75]. Bleomycin also demonstrated similar efficacy with 80.3% of patients with macrocystic lesions and 71.4% with mixed lesions having complete response [76]. Alcohol, doxycycline, and sodium tetradecyl sulfate have also been used, but the best sclerosing agent is still unclear [77,78]. Currently, there is no established guideline for the best choice of initial therapy between surgical excision, sclerotherapy or medical treatment. A retrospective cohort study demonstrated similar effectiveness for both initial surgical excision and sclerotherapy controlling for lesion stage and type [79].

4.2. Current Medical Treatment

Similar to surgical treatment, there is no consensus on best pharmaceutical treatment for lymphatic malformation. Sirolimus, an immunosuppressant, is currently the most commonly used agent for treatment of lymphovascular anomalies. Sirolimus is an inhibitor of mTOR, a downstream protein kinase in the PI3K pathway. While there is a lack of randomized control trial data demonstrating its effectiveness for lymphatic malformations, a number of retrospective and nonrandomized studies describe a decrease in lesion size in majority of patients with oral sirolimus [80,81]. In particular, a large prospective study by Adams et al. demonstrated disease response for mixed capillary or venous lymphatic malformation, and favorable response to microcystic lymphatic malformations with 1 year of oral sirolimus treatment. The most common side effects of oral sirolimus are related to its immunosuppressive effects including anemia, thrombocytopenia and leukopenia [82,83]. It is difficult to draw conclusions for topical sirolimus, as only 3 studies including 7 patients total have been described [81]. There are phase 2, multi-centered trials underway looking at efficacy of topical sirolimus for treatment of microcystic lymphatic malformation (NCT05050149 and NCT03972592). Other medications that have been discussed include sildenafil and propranolol, which are thought to ameliorate symptoms though vascular smooth muscle relaxation from inhibition of cGMP breakdown and reduction of lesion growth by downregulation of VEGF expression, respectively [84,85,86]. However, both have been reported in small case studies with mixed results.

5. Future Therapies

As more molecular players of the disease are being identified, these can represent potential targets for new therapeutics. One of those include PIK3CA gene, which is one of the most common mutations in cancer of all types [87]. Of note, human cancer often contains multiple oncogenic mutations along the same PI3K pathway in comparison to the heterozygous state for PROS [88]. There are a number of PI3K inhibitors that are being investigated for oncologic treatment including pan-PI3K inhibitors, such as copanlisib or buparlisib, and specific p110α inhibitors, such as alpelisib [89]. Alpelisib is currently approved for use in PIK3CA-mutated, hormone receptor positive advanced breast cancer. In a murine model of PROS/CLOVES, administration of alpelisib improved the survival, decreased tissue changes, and reduced cell proliferation of affected organs of PROS/CLOVES mice compared to wild type [90]. In a different mouse model of PROS, alpelisib again improved survival of mice, but also was able to prevent development of lymphatic malformations [91]. Alpelisib has been used in small groups of patients, with progressive reduction in tumor size, improvements in secondary organ dysfunctions including cardiac and renal function, musculoskeletal deformities and general clinical status [90,91]. However, larger scale prospective, randomized controlled trials are needed to fully evaluate the efficacy of these specific inhibitors. Topical application of PI3KA inhibitor is also under investigation, with a phase 1 clinical trial currently underway (NCT04409145).
Another strategy could include inhibiting AKT, a direct downstream serine threonine kinase of PI3K with a mosaic activating mutation that results in Proteus syndrome [58]. The pan-AKT inhibitor miransertib has been used in patients with cancer [92,93]. A dosing study of miransertib in adults and children with Proteus syndrome has been completed and case series of 2 patients has shown improvements in quality of life [94,95]. A phase 1/2 study is currently underway to establish efficacy and safety of miransertib for PROS (NCT03094832).
Lastly, photoacoustic technology, based on nonradiative absorbed energy combined with targeted lymph vessel endothelium labeling is being studied for mapping and potential therapeutic ablation. Kim et al. have used bioconjugated gold based nanoparticles with antibodies to LEC marker LYVE1 as photoacoustic and photothermal contrast agents in mice to precisely map lymphatic vasculature and malformations [96]. In addition, labeling of target cells can increase therapeutic efficacy and optimize guidance of laser treatment. In the same murine model with LYVE1 antibody conjugated gold nanoparticles, there was a 6-fold increase in laser energy with highly localized damage around zones of high concentration of gold nanoparticle clusters without deleterious effects in surrounding tissue [96]. Future studies are needed to establish bioconjugated nanoparticles as a potential targeted single diagnostic and therapeutic platform for microcystic lymphatic malformations. This will allow this theragnostic versatile tool to overcome current imaging technique limitations and possibly be utilized for other lymphatic related diseases.

6. Conclusions

While large strides into understanding the origins of lymphatic vessels and the molecular underpinnings of lymphatic malformations have been taken, the complexity of different contributions of endothelial cells and initial signaling for lymphangiogenesis is still being teased out. Further studies and investigations into understanding molecular mechanisms contributing to the specification and growth of lymphatic vessels will open doors to many new targeted therapies, including perhaps in utero therapies. Additional studies will also enrich the efforts to treat other diseases of lymphatic system including lymphedema and cancer.

Author Contributions

Conceptualization, S.Y.L. and D.L.F.; writing—original draft preparation, S.Y.L. and E.G.L.; writing—review and editing, S.Y.L., E.G.L., A.-E.S.H., M.C., A.W. and D.L.F.; visualization, S.Y.L. and M.C.; supervision, A.W. and D.L.F. All authors have read and agreed to the published version of the manuscript.

Funding

Author A.-E.S.H. was supported by the National Center for Advancing Translational Sciences, National Institutes of Health, through grant number UL1 TR001860.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to acknowledge Nora Lelivelt for her support with scientific writing. Figure 2 was created with Biorender.com.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Fishman, S.J.; Young, A.E. Slow-Flow Vascular Malformations; Oxford University Press: Oxford, UK, 2013; ISBN 978-0-19-935714-7. [Google Scholar]
  2. Dubois, J.; Thomas-Chaussé, F.; Soulez, G. Common (Cystic) Lymphatic Malformations: Current Knowledge and Management. Tech. Vasc. Interv. Radiol. 2019, 22, 100631. [Google Scholar] [CrossRef] [PubMed]
  3. Kulungowski, A.M.; Patel, M. Lymphatic Malformations. Semin. Pediatr. Surg. 2020, 29, 150971. [Google Scholar] [CrossRef] [PubMed]
  4. Merrow, A.C.; Gupta, A.; Patel, M.N.; Adams, D.M. 2014 Revised Classification of Vascular Lesions from the International Society for the Study of Vascular Anomalies: Radiologic-Pathologic Update. Radiographics 2016, 36, 1494–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Elluru, R.G.; Balakrishnan, K.; Padua, H.M. Lymphatic Malformations: Diagnosis and Management. Semin. Pediatr. Surg. 2014, 23, 178–185. [Google Scholar] [CrossRef]
  6. Brouillard, P.; Boon, L.; Vikkula, M. Genetics of Lymphatic Anomalies. J. Clin. Investig. 2014, 124, 898–904. [Google Scholar] [CrossRef]
  7. Semo, J.; Nicenboim, J.; Yaniv, K. Development of the Lymphatic System: New Questions and Paradigms. Development 2016, 143, 924–935. [Google Scholar] [CrossRef] [Green Version]
  8. Lewis, F.T. The Development of the Lymphatic System in Rabbits. Am. J. Anat. 1905, 5, 95–111. [Google Scholar] [CrossRef] [Green Version]
  9. Huntington, G.S.; McClure, C.F.W. The Anatomy and Development of the Jugular Lymph Sacs in the Domestic Cat (Felis Domestica). Am. J. Anat. 1910, 10, 177–312. [Google Scholar] [CrossRef]
  10. Ny, A.; Koch, M.; Schneider, M.; Neven, E.; Tong, R.T.; Maity, S.; Fischer, C.; Plaisance, S.; Lambrechts, D.; Héligon, C.; et al. A Genetic Xenopus Laevis Tadpole Model to Study Lymphangiogenesis. Nat. Med. 2005, 11, 998–1004. [Google Scholar] [CrossRef]
  11. Wilting, J.; Aref, Y.; Huang, R.; Tomarev, S.I.; Schweigerer, L.; Christ, B.; Valasek, P.; Papoutsi, M. Dual Origin of Avian Lymphatics. Dev. Biol. 2006, 292, 165–173. [Google Scholar] [CrossRef] [Green Version]
  12. Yaniv, K.; Isogai, S.; Castranova, D.; Dye, L.; Hitomi, J.; Weinstein, B.M. Live Imaging of Lymphatic Development in the Zebrafish. Nat. Med. 2006, 12, 711–716. [Google Scholar] [CrossRef] [PubMed]
  13. Okuda, K.S.; Astin, J.W.; Misa, J.P.; Flores, M.V.; Crosier, K.E.; Crosier, P.S. Lyve1 Expression Reveals Novel Lymphatic Vessels and New Mechanisms for Lymphatic Vessel Development in Zebrafish. Development 2012, 139, 2381–2391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Van Impel, A.; Zhao, Z.; Hermkens, D.M.A.; Roukens, M.G.; Fischer, J.C.; Peterson-Maduro, J.; Duckers, H.; Ober, E.A.; Ingham, P.W.; Schulte-Merker, S. Divergence of Zebrafish and Mouse Lymphatic Cell Fate Specification Pathways. Development 2014, 141, 1228–1238. [Google Scholar] [CrossRef] [Green Version]
  15. Srinivasan, R.S.; Dillard, M.E.; Lagutin, O.V.; Lin, F.-J.; Tsai, S.; Tsai, M.-J.; Samokhvalov, I.M.; Oliver, G. Lineage Tracing Demonstrates the Venous Origin of the Mammalian Lymphatic Vasculature. Genes Dev. 2007, 21, 2422–2432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Martinez-Corral, I.; Ulvmar, M.H.; Stanczuk, L.; Tatin, F.; Kizhatil, K.; John, S.W.M.; Alitalo, K.; Ortega, S.; Makinen, T. Nonvenous Origin of Dermal Lymphatic Vasculature. Circ. Res. 2015, 116, 1649–1654. [Google Scholar] [CrossRef] [Green Version]
  17. Klotz, L.; Norman, S.; Vieira, J.M.; Masters, M.; Rohling, M.; Dubé, K.N.; Bollini, S.; Matsuzaki, F.; Carr, C.A.; Riley, P.R. Cardiac Lymphatics Are Heterogeneous in Origin and Respond to Injury. Nature 2015, 522, 62–67. [Google Scholar] [CrossRef] [Green Version]
  18. Stanczuk, L.; Martinez-Corral, I.; Ulvmar, M.H.; Zhang, Y.; Laviña, B.; Fruttiger, M.; Adams, R.H.; Saur, D.; Betsholtz, C.; Ortega, S.; et al. CKit Lineage Hemogenic Endothelium-Derived Cells Contribute to Mesenteric Lymphatic Vessels. Cell Rep. 2015, 10, 1708–1721. [Google Scholar] [CrossRef]
  19. Koning, J.J.; Konijn, T.; Lakeman, K.A.; O’Toole, T.; Kenswil, K.J.G.; Raaijmakers, M.H.G.P.; Michurina, T.V.; Enikolopov, G.; Mebius, R.E. Nestin-Expressing Precursors Give Rise to Both Endothelial as Well as Nonendothelial Lymph Node Stromal Cells. J. Immunol. 2016, 197, 2686–2694. [Google Scholar] [CrossRef] [Green Version]
  20. Wigle, J.T.; Oliver, G. Prox1 Function Is Required for the Development of the Murine Lymphatic System. Cell 1999, 98, 769–778. [Google Scholar] [CrossRef] [Green Version]
  21. Kim, H.; Nguyen, V.P.; Petrova, T.V.; Cruz, M.; Alitalo, K.; Dumont, D.J. Embryonic Vascular Endothelial Cells Are Malleable to Reprogramming via Prox1 to a Lymphatic Gene Signature. BMC Dev. Biol. 2010, 10, 72. [Google Scholar] [CrossRef] [Green Version]
  22. Srinivasan, R.S.; Geng, X.; Yang, Y.; Wang, Y.; Mukatira, S.; Studer, M.; Porto, M.P.R.; Lagutin, O.; Oliver, G. The Nuclear Hormone Receptor Coup-TFII Is Required for the Initiation and Early Maintenance of Prox1 Expression in Lymphatic Endothelial Cells. Genes Dev. 2010, 24, 696–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. François, M.; Caprini, A.; Hosking, B.; Orsenigo, F.; Wilhelm, D.; Browne, C.; Paavonen, K.; Karnezis, T.; Shayan, R.; Downes, M.; et al. Sox18 Induces Development of the Lymphatic Vasculature in Mice. Nature 2008, 456, 643–647. [Google Scholar] [CrossRef] [PubMed]
  24. Banerji, S.; Ni, J.; Wang, S.X.; Clasper, S.; Su, J.; Tammi, R.; Jones, M.; Jackson, D.G. LYVE-1, a New Homologue of the CD44 Glycoprotein, Is a Lymph-Specific Receptor for Hyaluronan. J. Cell Biol. 1999, 144, 789–801. [Google Scholar] [CrossRef]
  25. Breiteneder-Geleff, S.; Soleiman, A.; Kowalski, H.; Horvat, R.; Amann, G.; Kriehuber, E.; Diem, K.; Weninger, W.; Tschachler, E.; Alitalo, K.; et al. Angiosarcomas Express Mixed Endothelial Phenotypes of Blood and Lymphatic Capillaries: Podoplanin as a Specific Marker for Lymphatic Endothelium. Am. J. Pathol. 1999, 154, 385–394. [Google Scholar] [CrossRef]
  26. Kaipainen, A.; Korhonen, J.; Mustonen, T.; van Hinsbergh, V.W.; Fang, G.H.; Dumont, D.; Breitman, M.; Alitalo, K. Expression of the Fms-like Tyrosine Kinase 4 Gene Becomes Restricted to Lymphatic Endothelium during Development. Proc. Natl. Acad. Sci. USA 1995, 92, 3566–3570. [Google Scholar] [CrossRef] [Green Version]
  27. Joukov, V.; Pajusola, K.; Kaipainen, A.; Chilov, D.; Lahtinen, I.; Kukk, E.; Saksela, O.; Kalkkinen, N.; Alitalo, K. A Novel Vascular Endothelial Growth Factor, VEGF-C, Is a Ligand for the Flt4 (VEGFR-3) and KDR (VEGFR-2) Receptor Tyrosine Kinases. EMBO J. 1996, 15, 290–298. [Google Scholar] [CrossRef]
  28. Mäkinen, T.; Veikkola, T.; Mustjoki, S.; Karpanen, T.; Catimel, B.; Nice, E.C.; Wise, L.; Mercer, A.; Kowalski, H.; Kerjaschki, D.; et al. Isolated Lymphatic Endothelial Cells Transduce Growth, Survival and Migratory Signals via the VEGF-C/D Receptor VEGFR-3. EMBO J. 2001, 20, 4762–4773. [Google Scholar] [CrossRef] [Green Version]
  29. Zhou, F.; Chang, Z.; Zhang, L.; Hong, Y.-K.; Shen, B.; Wang, B.; Zhang, F.; Lu, G.; Tvorogov, D.; Alitalo, K.; et al. Akt/Protein Kinase B Is Required for Lymphatic Network Formation, Remodeling, and Valve Development. Am. J. Pathol. 2010, 177, 2124–2133. [Google Scholar] [CrossRef]
  30. Deng, Y.; Atri, D.; Eichmann, A.; Simons, M. Endothelial ERK Signaling Controls Lymphatic Fate Specification. J. Clin. Invest. 2013, 123, 1202–1215. [Google Scholar] [CrossRef]
  31. Xu, Y.; Yuan, L.; Mak, J.; Pardanaud, L.; Caunt, M.; Kasman, I.; Larrivée, B.; del Toro, R.; Suchting, S.; Medvinsky, A.; et al. Neuropilin-2 Mediates VEGF-C–Induced Lymphatic Sprouting Together with VEGFR3. J. Cell Biol. 2010, 188, 115–130. [Google Scholar] [CrossRef] [Green Version]
  32. Karkkainen, M.J.; Haiko, P.; Sainio, K.; Partanen, J.; Taipale, J.; Petrova, T.V.; Jeltsch, M.; Jackson, D.G.; Talikka, M.; Rauvala, H.; et al. Vascular Endothelial Growth Factor C Is Required for Sprouting of the First Lymphatic Vessels from Embryonic Veins. Nat. Immunol. 2004, 5, 74–80. [Google Scholar] [CrossRef] [PubMed]
  33. Achen, M.G.; Roufail, S.; Domagala, T.; Catimel, B.; Nice, E.C.; Geleick, D.M.; Murphy, R.; Scott, A.M.; Caesar, C.; Makinen, T.; et al. Monoclonal Antibodies to Vascular Endothelial Growth Factor-D Block Its Interactions with Both VEGF Receptor-2 and VEGF Receptor-3. Eur. J. Biochem. 2000, 267, 2505–2515. [Google Scholar] [CrossRef] [PubMed]
  34. Joukov, V.; Sorsa, T.; Kumar, V.; Jeltsch, M.; Claesson-Welsh, L.; Cao, Y.; Saksela, O.; Kalkkinen, N.; Alitalo, K. Proteolytic Processing Regulates Receptor Specificity and Activity of VEGF-C. EMBO J. 1997, 16, 3898–3911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Stacker, S.A.; Stenvers, K.; Caesar, C.; Vitali, A.; Domagala, T.; Nice, E.; Roufail, S.; Simpson, R.J.; Moritz, R.; Karpanen, T.; et al. Biosynthesis of Vascular Endothelial Growth Factor-D Involves Proteolytic Processing Which Generates Non-Covalent Homodimers. J. Biol. Chem. 1999, 274, 32127–32136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Baldwin, M.E.; Halford, M.M.; Roufail, S.; Williams, R.A.; Hibbs, M.L.; Grail, D.; Kubo, H.; Stacker, S.A.; Achen, M.G. Vascular Endothelial Growth Factor D Is Dispensable for Development of the Lymphatic System. Mol. Cell. Biol. 2005, 25, 2441–2449. [Google Scholar] [CrossRef] [Green Version]
  37. Jeltsch, M.; Jha, S.K.; Tvorogov, D.; Anisimov, A.; Leppänen, V.-M.; Holopainen, T.; Kivelä, R.; Ortega, S.; Kärpanen, T.; Alitalo, K. CCBE1 Enhances Lymphangiogenesis via A Disintegrin and Metalloprotease With Thrombospondin Motifs-3–Mediated Vascular Endothelial Growth Factor-C Activation. Circulation 2014, 129, 1962–1971. [Google Scholar] [CrossRef] [Green Version]
  38. Roukens, M.G.; Peterson-Maduro, J.; Padberg, Y.; Jeltsch, M.; Leppänen, V.-M.; Bos, F.L.; Alitalo, K.; Schulte-Merker, S.; Schulte, D. Functional Dissection of the CCBE1 Protein. Circ. Res. 2015, 116, 1660–1669. [Google Scholar] [CrossRef] [Green Version]
  39. Gale, N.W.; Thurston, G.; Hackett, S.F.; Renard, R.; Wang, Q.; McClain, J.; Martin, C.; Witte, C.; Witte, M.H.; Jackson, D.; et al. Angiopoietin-2 Is Required for Postnatal Angiogenesis and Lymphatic Patterning, and Only the Latter Role Is Rescued by Angiopoietin-1. Dev. Cell 2002, 3, 411–423. [Google Scholar] [CrossRef]
  40. Srinivasan, R.S.; Oliver, G. Prox1 Dosage Controls the Number of Lymphatic Endothelial Cell Progenitors and the Formation of the Lymphovenous Valves. Genes Dev. 2011, 25, 2187–2197. [Google Scholar] [CrossRef] [Green Version]
  41. Wiegand, S.; Eivazi, B.; Barth, P.J.; von Rautenfeld, D.B.; Folz, B.J.; Mandic, R.; Werner, J.A. Pathogenesis of Lymphangiomas. Virchows Arch. 2008, 453, 1–8. [Google Scholar] [CrossRef]
  42. Zadvinskis, D.P.; Benson, M.T.; Kerr, H.H.; Mancuso, A.A.; Cacciarelli, A.A.; Madrazo, B.L.; Mafee, M.F.; Dalen, K. Congenital Malformations of the Cervicothoracic Lymphatic System: Embryology and Pathogenesis. Radiographics 1992, 12, 1175–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Oliver, G.; Kipnis, J.; Randolph, G.J.; Harvey, N.L. The Lymphatic Vasculature in the 21st Century: Novel Functional Roles in Homeostasis and Disease. Cell 2020, 182, 270–296. [Google Scholar] [CrossRef]
  44. Baluk, P.; Fuxe, J.; Hashizume, H.; Romano, T.; Lashnits, E.; Butz, S.; Vestweber, D.; Corada, M.; Molendini, C.; Dejana, E.; et al. Functionally Specialized Junctions between Endothelial Cells of Lymphatic Vessels. J. Exp. Med. 2007, 204, 2349–2362. [Google Scholar] [CrossRef] [PubMed]
  45. Weingast, G.R.; Hopper, K.D.; Gottesfeld, S.A.; Manco-Johnson, M.L. Congenital Lymphangiectasia with Fetal Cystic Hygroma: Report of Two Cases with Coexistent Down’s Syndrome. J. Clin. Ultrasound 1988, 16, 663–668. [Google Scholar] [CrossRef] [PubMed]
  46. Van der Putte, S.C.J. Lymphatic Malformation in Human Fetuses. Virchows Arch. A Path. Anat. Histol. 1977, 376, 233–246. [Google Scholar] [CrossRef]
  47. Phillips, H.E.; McGahan, J.P. Intrauterine Fetal Cystic Hygromas: Sonographic Detection. AJR Am. J. Roentgenol. 1981, 136, 799–802. [Google Scholar] [CrossRef] [Green Version]
  48. Van Meerhaeghe, T.; Vandenbroucke, F.; Velkeniers, B. Systemic Generalised Lymphangiomatosis: Unknown Aetiology and a Challenge to Treat. BMJ Case Rep. 2021, 14, e237331. [Google Scholar] [CrossRef]
  49. Ozeki, M.; Nozawa, A.; Kawamoto, N.; Fujino, A.; Hirakawa, S.; Fukao, T. Potential Biomarkers of Kaposiform Lymphangiomatosis. Pediatr. Blood Cancer 2019, 66, e27878. [Google Scholar] [CrossRef]
  50. Croteau, S.E.; Kozakewich, H.P.W.; Perez-Atayde, A.R.; Fishman, S.J.; Alomari, A.I.; Chaudry, G.; Mulliken, J.B.; Trenor, C.C. Kaposiform Lymphangiomatosis: A Distinct Aggressive Lymphatic Anomaly. J. Pediatr. 2014, 164, 383–388. [Google Scholar] [CrossRef] [Green Version]
  51. Ruggieri, P.; Montalti, M.; Angelini, A.; Alberghini, M.; Mercuri, M. Gorham–Stout Disease: The Experience of the Rizzoli Institute and Review of the Literature. Skelet. Radiol. 2011, 40, 1391–1397. [Google Scholar] [CrossRef]
  52. Rossi, M.; Buonuomo, P.S.; Battafarano, G.; Conforti, A.; Mariani, E.; Algeri, M.; Pelle, S.; D’Agostini, M.; Macchiaiolo, M.; De Vito, R.; et al. Dissecting the Mechanisms of Bone Loss in Gorham-Stout Disease. Bone 2020, 130, 115068. [Google Scholar] [CrossRef] [PubMed]
  53. Kurek, K.C.; Luks, V.L.; Ayturk, U.M.; Alomari, A.I.; Fishman, S.J.; Spencer, S.A.; Mulliken, J.B.; Bowen, M.E.; Yamamoto, G.L.; Kozakewich, H.P.W.; et al. Somatic Mosaic Activating Mutations in PIK3CA Cause CLOVES Syndrome. Am. J. Hum. Genet. 2012, 90, 1108–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Le Cras, T.D.; Goines, J.; Lakes, N.; Pastura, P.; Hammill, A.M.; Adams, D.M.; Boscolo, E. Constitutively Active PIK3CA Mutations Are Expressed by Lymphatic and Vascular Endothelial Cells in Capillary Lymphatic Venous Malformation. Angiogenesis 2020, 23, 425–442. [Google Scholar] [CrossRef] [PubMed]
  55. Luks, V.L.; Kamitaki, N.; Vivero, M.P.; Uller, W.; Rab, R.; Bovée, J.V.M.G.; Rialon, K.L.; Guevara, C.J.; Alomari, A.I.; Greene, A.K.; et al. Lymphatic and Other Vascular Malformative/Overgrowth Disorders Are Caused by Somatic Mutations in PIK3CA. J. Pediatr. 2015, 166, 1048–1054. [Google Scholar] [CrossRef] [Green Version]
  56. Burrows, P.E.; Gonzalez-Garay, M.L.; Rasmussen, J.C.; Aldrich, M.B.; Guilliod, R.; Maus, E.A.; Fife, C.E.; Kwon, S.; Lapinski, P.E.; King, P.D.; et al. Lymphatic Abnormalities Are Associated with RASA1 Gene Mutations in Mouse and Man. Proc. Natl. Acad. Sci. USA 2013, 110, 8621–8626. [Google Scholar] [CrossRef] [Green Version]
  57. Wang, S.; Wang, W.; Zhang, X.; Gui, J.; Zhang, J.; Guo, Y.; Liu, Y.; Han, L.; Liu, Q.; Li, Y.; et al. A Somatic Mutation in PIK3CD Unravels a Novel Candidate Gene for Lymphatic Malformation. Orphanet J. Rare Dis. 2021, 16, 208. [Google Scholar] [CrossRef]
  58. Lindhurst, M.J.; Sapp, J.C.; Teer, J.K.; Johnston, J.J.; Finn, E.M.; Peters, K.; Turner, J.; Cannons, J.L.; Bick, D.; Blakemore, L.; et al. A Mosaic Activating Mutation in AKT1 Associated with the Proteus Syndrome. N. Engl. J. Med. 2011, 365, 611–619. [Google Scholar] [CrossRef] [Green Version]
  59. Nguyen, D.; Turner, J.T.; Olsen, C.; Biesecker, L.G.; Darling, T.N. Cutaneous Manifestations of Proteus Syndrome: Correlations with General Clinical Severity. Arch. Dermatol. 2004, 140, 947–953. [Google Scholar] [CrossRef]
  60. AlAnzi, T.; Al-Mashharawi, E.; Alhashem, A. Proteus Syndrome Caused by Novel Somatic AKT1 Duplication. Saudi Med. J. 2021, 42, 95–99. [Google Scholar] [CrossRef]
  61. Vivanco, I.; Sawyers, C.L. The Phosphatidylinositol 3-Kinase–AKT Pathway in Human Cancer. Nat. Rev. Cancer 2002, 2, 489–501. [Google Scholar] [CrossRef]
  62. Kangas, J.; Nätynki, M.; Eklund, L. Development of Molecular Therapies for Venous Malformations. Basic Clin. Pharmacol. Toxicol. 2018, 123, 6–19. [Google Scholar] [CrossRef]
  63. Martinez-Corral, I.; Zhang, Y.; Petkova, M.; Ortsäter, H.; Sjöberg, S.; Castillo, S.D.; Brouillard, P.; Libbrecht, L.; Saur, D.; Graupera, M.; et al. Blockade of VEGF-C Signaling Inhibits Lymphatic Malformations Driven by Oncogenic PIK3CA Mutation. Nat. Commun. 2020, 11, 2869. [Google Scholar] [CrossRef]
  64. Han, T.; Yan, J.; Chen, H.; Ji, Y.; Chen, J.; Cui, J.; Shen, W.; Zou, J. HIF-1α Contributes to Tube Malformation of Human Lymphatic Endothelial Cells by Upregulating VEGFR-3. Int. J. Oncol. 2019, 54, 139–151. [Google Scholar] [CrossRef] [PubMed]
  65. Boussat, S.; Eddahibi, S.; Coste, A.; Fataccioli, V.; Gouge, M.; Housset, B.; Adnot, S.; Maitre, B. Expression and Regulation of Vascular Endothelial Growth Factor in Human Pulmonary Epithelial Cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2000, 279, L371–L378. [Google Scholar] [CrossRef] [Green Version]
  66. Mizukami, Y.; Li, J.; Zhang, X.; Zimmer, M.A.; Iliopoulos, O.; Chung, D.C. Hypoxia-Inducible Factor-1-Independent Regulation of Vascular Endothelial Growth Factor by Hypoxia in Colon Cancer. Cancer Res. 2004, 64, 1765–1772. [Google Scholar] [CrossRef] [Green Version]
  67. Liang, X.; Yang, D.; Hu, J.; Hao, X.; Gao, J.; Mao, Z. Hypoxia Inducible Factor-Alpha Expression Correlates with Vascular Endothelial Growth Factor-C Expression and Lymphangiogenesis/Angiogenesis in Oral Squamous Cell Carcinoma. Anticancer Res. 2008, 28, 1659–1666. [Google Scholar] [PubMed]
  68. Gomez-Acevedo, H.; Dornhoffer, J.R.; Stone, A.; Dai, Y.; Richter, G.T. Gene Expression Differences in Pediatric Lymphatic Malformations: Size Really Matters. Lymphat. Res. Biol. 2018, 16, 347–352. [Google Scholar] [CrossRef]
  69. Hinnebusch, A.G. The Scanning Mechanism of Eukaryotic Translation Initiation. Annu. Rev. Biochem. 2014, 83, 779–812. [Google Scholar] [CrossRef] [PubMed]
  70. Kalwani, N.M.; Rockson, S.G. Management of Lymphatic Vascular Malformations: A Systematic Review of the Literature. J. Vasc. Surg. Venous Lymphat. Disord. 2021, 9, 1077–1082. [Google Scholar] [CrossRef] [PubMed]
  71. Dighe, M.K.; Peterson, S.E.; Dubinsky, T.J.; Perkins, J.; Cheng, E. EXIT Procedure: Technique and Indications with Prenatal Imaging Parameters for Assessment of Airway Patency. Radiographics 2011, 31, 511–526. [Google Scholar] [CrossRef] [PubMed]
  72. Bonilla-Velez, J.; Moore, B.P.; Cleves, M.A.; Buckmiller, L.; Richter, G.T. Surgical Resection of Macrocystic Lymphatic Malformations of the Head and Neck: Short and Long-Term Outcomes. Int. J. Pediatr. Otorhinolaryngol. 2020, 134, 110013. [Google Scholar] [CrossRef] [PubMed]
  73. Glade, R.S.; Buckmiller, L.M. CO2 Laser Resurfacing of Intraoral Lymphatic Malformations: A 10-Year Experience. Int. J. Pediatr. Otorhinolaryngol. 2009, 73, 1358–1361. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, S.W.; Kauvanough, K.; Orbach, D.B.; Alomari, A.I.; Mulliken, J.B.; Rahbar, R. Long-Term Outcome of Radiofrequency Ablation for Intraoral Microcystic Lymphatic Malformation. Arch. Otolaryngol.–Head Neck Surg. 2011, 137, 1247–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Smith, M.C.; Zimmerman, M.B.; Burke, D.K.; Bauman, N.M.; Sato, Y.; Smith, R.J.H. OK-432 Collaborative Study Group Efficacy and Safety of OK-432 Immunotherapy of Lymphatic Malformations. Laryngoscope 2009, 119, 107–115. [Google Scholar] [CrossRef]
  76. Tiwari, P.; Pandey, V.; Bera, R.N.; Sharma, S.P.; Chauhan, N. Bleomycin Sclerotherapy in Lymphangiomas of the Head and Neck Region: A Prospective Study. Int. J. Oral Maxillofac. Surg. 2021, 50, 619–626. [Google Scholar] [CrossRef]
  77. Cahill, A.M.; Nijs, E.; Ballah, D.; Rabinowitz, D.; Thompson, L.; Rintoul, N.; Hedrick, H.; Jacobs, I.; Low, D. Percutaneous Sclerotherapy in Neonatal and Infant Head and Neck Lymphatic Malformations: A Single Center Experience. J. Pediatr. Surg. 2011, 46, 2083–2095. [Google Scholar] [CrossRef]
  78. Thomas, D.M.; Wieck, M.M.; Grant, C.N.; Dossa, A.; Nowicki, D.; Stanley, P.; Zeinati, C.; Howell, L.K.; Anselmo, D.M. Doxycycline Sclerotherapy Is Superior in the Treatment of Pediatric Lymphatic Malformations. J. Vasc. Interv. Radiol. 2016, 27, 1846–1856. [Google Scholar] [CrossRef]
  79. Balakrishnan, K.; Menezes, M.D.; Chen, B.S.; Magit, A.E.; Perkins, J.A. Primary Surgery vs Primary Sclerotherapy for Head and Neck Lymphatic Malformations. JAMA Otolaryngol.–Head Neck Surg. 2014, 140, 41–45. [Google Scholar] [CrossRef]
  80. Wiegand, S.; Wichmann, G.; Dietz, A. Treatment of Lymphatic Malformations with the MTOR Inhibitor Sirolimus: A Systematic Review. Lymphat. Res. Biol. 2018, 16, 330–339. [Google Scholar] [CrossRef] [Green Version]
  81. Freixo, C.; Ferreira, V.; Martins, J.; Almeida, R.; Caldeira, D.; Rosa, M.; Costa, J.; Ferreira, J. Efficacy and Safety of Sirolimus in the Treatment of Vascular Anomalies: A Systematic Review. J. Vasc. Surg. 2020, 71, 318–327. [Google Scholar] [CrossRef] [Green Version]
  82. Adams, D.M.; Trenor, C.C.; Hammill, A.M.; Vinks, A.A.; Patel, M.N.; Chaudry, G.; Wentzel, M.S.; Mobberley-Schuman, P.S.; Campbell, L.M.; Brookbank, C.; et al. Efficacy and Safety of Sirolimus in the Treatment of Complicated Vascular Anomalies. Pediatrics 2016, 137, e20153257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Nguyen, L.S.; Vautier, M.; Allenbach, Y.; Zahr, N.; Benveniste, O.; Funck-Brentano, C.; Salem, J.-E. Sirolimus and MTOR Inhibitors: A Review of Side Effects and Specific Management in Solid Organ Transplantation. Drug Saf. 2019, 42, 813–825. [Google Scholar] [CrossRef] [PubMed]
  84. Danial, C.; Tichy, A.L.; Tariq, U.; Swetman, G.L.; Khuu, P.; Leung, T.H.; Benjamin, L.; Teng, J.; Vasanawala, S.S.; Lane, A.T. An Open-Label Study to Evaluate Sildenafil for the Treatment of Lymphatic Malformations. J. Am. Acad. Dermatol. 2014, 70, 1050–1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Koshy, J.C.; Eisemann, B.S.; Agrawal, N.; Pimpalwar, S.; Edmonds, J.L. Sildenafil for Microcystic Lymphatic Malformations of the Head and Neck: A Prospective Study. Int. J. Pediatr. Otorhinolaryngol. 2015, 79, 980–982. [Google Scholar] [CrossRef]
  86. Ozeki, M.; Kanda, K.; Kawamoto, N.; Ohnishi, H.; Fujino, A.; Hirayama, M.; Kato, Z.; Azuma, E.; Fukao, T.; Kondo, N. Propranolol as an Alternative Treatment Option for Pediatric Lymphatic Malformation. Tohoku J. Exp. Med. 2013, 229, 61–66. [Google Scholar] [CrossRef] [Green Version]
  87. Samuels, Y.; Wang, Z.; Bardelli, A.; Silliman, N.; Ptak, J.; Szabo, S.; Yan, H.; Gazdar, A.; Powell, S.M.; Riggins, G.J.; et al. High Frequency of Mutations of the PIK3CA Gene in Human Cancers. Science 2004, 304, 554. [Google Scholar] [CrossRef] [Green Version]
  88. Madsen, R.R.; Knox, R.G.; Pearce, W.; Lopez, S.; Mahler-Araujo, B.; McGranahan, N.; Vanhaesebroeck, B.; Semple, R.K. Oncogenic PIK3CA Promotes Cellular Stemness in an Allele Dose-Dependent Manner. Proc. Natl. Acad. Sci. USA 2019, 116, 8380–8389. [Google Scholar] [CrossRef] [Green Version]
  89. Keppler-Noreuil, K.M.; Parker, V.E.R.; Darling, T.N.; Martinez-Agosto, J.A. Somatic Overgrowth Disorders of the PI3K/AKT/MTOR Pathway & Therapeutic Strategies. Am. J. Med. Genet. C Semin. Med. Genet. 2016, 172, 402–421. [Google Scholar] [CrossRef] [Green Version]
  90. Venot, Q.; Blanc, T.; Rabia, S.H.; Berteloot, L.; Ladraa, S.; Duong, J.-P.; Blanc, E.; Johnson, S.C.; Hoguin, C.; Boccara, O.; et al. Targeted Therapy in Patients with PIK3CA-Related Overgrowth Syndrome. Nature 2018, 558, 540–546. [Google Scholar] [CrossRef]
  91. Delestre, F.; Venot, Q.; Bayard, C.; Fraissenon, A.; Ladraa, S.; Hoguin, C.; Chapelle, C.; Yamaguchi, J.; Cassaca, R.; Zerbib, L.; et al. Alpelisib Administration Reduced Lymphatic Malformations in a Mouse Model and in Patients. Sci. Transl. Med. 2021, 13, eabg0809. [Google Scholar] [CrossRef]
  92. Lakhani, N.; Tolcher, A.W.; Rasco, D.W.; Patnaik, A.; O’Rourke, T.J.; Schwartz, B.E.; Abbadessa, G.; Kazakin, J.; Savage, R.; Wang, Y.; et al. Results of a Phase Ib Study of ARQ 092 in Combination with Carboplatin (C) plus Paclitaxel (P), or with P in Patients (Pts) with Solid Tumors. JCO 2017, 35, 2524. [Google Scholar] [CrossRef]
  93. Hyman, D.; Bonafede, M.; O’Cearbhaill, R.; Grisham, R.; Zamarin, D.; Tew, W.; Aghajanian, C.; Cadoo, K.; Friedman, C.; Savage, R.E.; et al. Abstract CT035: A Phase Ib Study of Miransertib (ARQ 092) in Combination with Anastrozole in Patients with PIK3CA or AKT1-Mutant ER+ Endometrial or Ovarian Cancer. Cancer Res. 2018, 78, CT035. [Google Scholar] [CrossRef]
  94. Keppler-Noreuil, K.M.; Sapp, J.C.; Lindhurst, M.J.; Darling, T.N.; Burton-Akright, J.; Bagheri, M.; Dombi, E.; Gruber, A.; Jarosinski, P.F.; Martin, S.; et al. Pharmacodynamic Study of Miransertib in Individuals with Proteus Syndrome. Am. J. Hum. Genet. 2019, 104, 484–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Forde, K.; Resta, N.; Ranieri, C.; Rea, D.; Kubassova, O.; Hinton, M.; Andrews, K.A.; Semple, R.; Irvine, A.D.; Dvorakova, V. Clinical Experience with the AKT1 Inhibitor Miransertib in Two Children with PIK3CA-Related Overgrowth Syndrome. Orphanet J. Rare Dis. 2021, 16, 109. [Google Scholar] [CrossRef]
  96. Kim, J.-W.; Galanzha, E.I.; Shashkov, E.V.; Moon, H.-M.; Zharov, V.P. Golden Carbon Nanotubes as Multimodal Photoacoustic and Photothermal High-Contrast Molecular Agents. Nat. Nanotechnol. 2009, 4, 688–694. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Hematoxylin and eosin stained of lymphatic malformation. (A) 40× image shows variably sized anastomosing vascular space lined by endothelial cells with stromal fibrosis of the malformation walls. (B) 100× image shows small with bland appearing endothelial cells with oval to flattened nuclei. Lymphocytes are present within both dilated lumina and septa.
Figure 1. Hematoxylin and eosin stained of lymphatic malformation. (A) 40× image shows variably sized anastomosing vascular space lined by endothelial cells with stromal fibrosis of the malformation walls. (B) 100× image shows small with bland appearing endothelial cells with oval to flattened nuclei. Lymphocytes are present within both dilated lumina and septa.
Jdb 10 00011 g001
Figure 2. Major hypothesis regarding pathogenesis of lymphatic malformations. (A) McClure’s centripetal theory of mesenchymal origin of lymphangiogenesis where primordial sacs fail to fuse with venous system. (B) Tissue abnormally sequestered early in embryogenesis and fail to join central lymphatic channels. (C) Abnormal budding of lymphatic structures. Created with Biorender.com.
Figure 2. Major hypothesis regarding pathogenesis of lymphatic malformations. (A) McClure’s centripetal theory of mesenchymal origin of lymphangiogenesis where primordial sacs fail to fuse with venous system. (B) Tissue abnormally sequestered early in embryogenesis and fail to join central lymphatic channels. (C) Abnormal budding of lymphatic structures. Created with Biorender.com.
Jdb 10 00011 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, S.Y.; Loll, E.G.; Hassan, A.-E.S.; Cheng, M.; Wang, A.; Farmer, D.L. Genetic and Molecular Determinants of Lymphatic Malformations: Potential Targets for Therapy. J. Dev. Biol. 2022, 10, 11. https://0-doi-org.brum.beds.ac.uk/10.3390/jdb10010011

AMA Style

Lee SY, Loll EG, Hassan A-ES, Cheng M, Wang A, Farmer DL. Genetic and Molecular Determinants of Lymphatic Malformations: Potential Targets for Therapy. Journal of Developmental Biology. 2022; 10(1):11. https://0-doi-org.brum.beds.ac.uk/10.3390/jdb10010011

Chicago/Turabian Style

Lee, Su Yeon, Emma Grace Loll, Abd-Elrahman Said Hassan, Mingyu Cheng, Aijun Wang, and Diana Lee Farmer. 2022. "Genetic and Molecular Determinants of Lymphatic Malformations: Potential Targets for Therapy" Journal of Developmental Biology 10, no. 1: 11. https://0-doi-org.brum.beds.ac.uk/10.3390/jdb10010011

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop