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

Sporadic lymphangioleiomyomatosis: Epidemiology and pathogenesis

Sporadic lymphangioleiomyomatosis: Epidemiology and pathogenesis
Literature review current through: Jan 2024.
This topic last updated: Dec 21, 2022.

INTRODUCTION — Lymphangioleiomyomatosis (LAM) is a rare multisystem disorder, belonging to the family of neoplasms with perivascular epithelioid differentiation (PEComa) [1], that mostly afflicts women and primarily affects the lung [2-5]. The term sporadic LAM is used for patients with LAM who do not have tuberous sclerosis complex (TSC), while TSC-LAM refers to LAM that occurs in patients with TSC. Since the 2000s, studies exploring the pathogenesis of LAM have produced ground-breaking insights into the genetic and molecular basis of LAM and have led to the development of an effective treatment.  

The epidemiology and pathophysiology of sporadic LAM will be reviewed here. The clinical evaluation and treatment of sporadic LAM and the clinical features and diagnosis of TSC and TSC-associated LAM are discussed separately. (See "Tuberous sclerosis complex associated lymphangioleiomyomatosis in adults" and "Sporadic lymphangioleiomyomatosis: Clinical presentation and diagnostic evaluation" and "Sporadic lymphangioleiomyomatosis: Treatment and prognosis" and "Tuberous sclerosis complex: Clinical features".)

EPIDEMIOLOGY — The true incidence and prevalence of sporadic LAM are unknown, as the available epidemiological data are observational and frequently include patients with tuberous sclerosis complex (TSC). Nonetheless, clinical experience and most studies confirm that the sporadic variant of LAM is rare and that it almost exclusively affects women.

Where previous estimates suggested a rate of 1 per million in the general population, regional and national registry data indicate higher rates, possibly reflecting advances in disease recognition and diagnosis [6-13]. As examples:

One study that collected data from seven countries, including the United States, the United Kingdom, and Japan, reported that the prevalence of LAM was between 3 and 8 per million women [8].

The LAM Foundation estimated a prevalence of 3 to 5 women per million women [14]. However, more than 2000 women from the United States have been diagnosed with LAM, consistent with a minimum prevalence of >10 per million (assuming United States population of females, 18 years or older of about 134 million). About 10 to 15 percent of patients registered with The LAM Foundation also report having TSC.

In a database study from Quebec, the prevalence of LAM was approximately 2 to 3 per million women [15] .

The highest rates of LAM occur in patients with TSC. Briefly, given the estimate of the worldwide prevalence of TSC of one million people (including 0.38 million females age 18 or older) and the conservative projection that 30 percent of women with TSC develop cystic changes consistent with LAM, the number of women with TSC-LAM on earth is predicted to be at least 100,000. The epidemiology of TSC is discussed separately. (See "Tuberous sclerosis complex associated lymphangioleiomyomatosis in adults", section on 'Epidemiology'.)

Although LAM can affect all races, one United States registry reported that White Americans may have a higher prevalence of LAM, especially when compared with individuals of African or Asian descent [13]. The same registry also reported that LAM was more commonly found in women of higher socioeconomic class; however, apparent racial and ethnic disparities in prevalence likely reflect differential access to healthcare rather than a true biological phenomenon.

LAM has rarely been reported in men, most often in association with definite or probable TSC [16-21]. Screening of large TSC clinics revealed cystic changes consistent with LAM in 10 to 13 percent of males [22,23]. There has been only one published case of a male with LAM with no clinical or genotypic evidence of TSC, although limitations in genetic analysis can result in "no mutation detected" in up to 15 percent of patients [19].

Traditionally, LAM was thought to only affect young premenopausal women of childbearing age. The average age at diagnosis of LAM is around 35 years. However, studies report LAM in women ranging from teenagers to octogenarians [5,7,9,13,24-29]. Prepubertal LAM is rare [30].

There are no known risk factors, other than TSC, for the development of LAM. While smoking may potentially worsen disease progression it does not appear to be a risk factor for disease induction [13].  

PATHOGENESIS — The primary histopathological abnormality in LAM is the proliferation of atypical smooth muscle-like cells (LAM cells) (picture 1A-B and picture 2). Additionally, in the lung, LAM cells are associated with multiple cysts (picture 3A-C) (see "Sporadic lymphangioleiomyomatosis: Clinical presentation and diagnostic evaluation", section on 'Tissue pathology'). Significant advances in our understanding of the pathogenesis of LAM have occurred since the early 2000s [31-33]. Excessive proliferation of LAM cells is driven by mutations in tuberous sclerosis complex (TSC) genes, in particular, TSC2. Additional factors that may contribute to cellular proliferation in sporadic LAM include aberrant stimulation of LAM cell growth by estrogen and other growth factors. Processes that promote differentiation (eg, expression of melanoma antigens), migration (estrogen), immune evasion (increased signaling through immune checkpoint inhibitors such as PD-1, or their ligands such as PD-L1), lymphangiogenesis (vascular endothelial growth factors-C and D), and tissue destruction (eg, proteases) have also been implicated in the pathogenesis of LAM. It is likely that some combination of these mechanisms best explains the multisystemic nature and unique features that occur in LAM.

The pathogenesis of TSC is discussed separately. (See "Tuberous sclerosis complex associated lymphangioleiomyomatosis in adults".)

Cellular proliferation

Tuberous sclerosis gene mutations — Since the early 2000s, reports from several groups support a dominant role for loss of function of the tuberous sclerosis proteins, tuberin or hamartin, in the pathogenesis of LAM. Tuberin is the protein product of the TSC gene, TSC2, which is located on chromosome 16p13. Tuberin complexes with hamartin, a sister protein that is encoded by the TSC1 gene located on chromosome 9q34 [34,35]. The tuberin-hamartin hetero-oligomer functions as a tumor suppressor complex that inhibits mechanistic target of rapamycin (mTOR; previously called mammalian target of rapamycin), which in turn integrates and controls cellular signals that regulate cell growth, cell size, cell survival, and autophagy (figure 1) [36-38]. Thus, when tuberin or hamartin function is lost, mTOR activity is unrestrained and these processes become dysregulated. (See "Tuberous sclerosis complex: Genetics and pathogenesis", section on 'Mechanism of tumor formation'.)

The central role of the loss of functioning tuberin in the pathogenesis of sporadic LAM is supported clinically by the nearly identical pathological presentations of the sporadic and TSC subtypes of LAM, the high proportion of women with TSC who develop cysts consistent with LAM (up to 80 percent by age 40 years) [39], and consistent findings of mutations in TSC genes and activation of downstream signals (eg, phosphorylation of ribosomal protein S6) in LAM cells derived from patients with both sporadic LAM and TSC-LAM [40-48].

Unlike patients with TSC who have germline mutations (ie, genes that are either inherited in a Mendelian fashion or occur early in embryogenesis and are present in every cell of offspring) of TSC1 or TSC2, patients with sporadic LAM have only somatic mutations (ie, mutations limited to the abnormal lesions) in TSC genes; thus, sporadic LAM is not inheritable. Only one case of generalized mosaicism for TSC2 has been reported among 61 cases of sporadic LAM [49]. Interestingly, all cases of sporadic LAM that have genetic data available are due to mutations in TSC2 genes, whereas both TSC1 and TSC2 mutations have been reported in patients with TSC-LAM [45,50]. Whereas lesions in TSC-LAM occur as a result of a germline (first hit) and a somatic (second hit) mutation in one of either TSC1 or TSC2, lesion development in sporadic LAM is associated with somatic mutations in both parental TSC alleles [40-47]. Low-level somatic mosaicism as the genesis of the “first hit” cannot be excluded (ie, some, but not all, cells become mutated during embryogenesis). Regardless of the mechanism, the end result is deficient or defective tuberin or hamartin in affected cells and dysregulated signaling through the mTOR pathway.

Genetic studies of involved tissues have only been conducted in a limited number of patients with sporadic LAM. As examples, early studies revealed somatic TSC2 mutations and loss of heterozygosity (LOH) for TSC2 in approximately one-half of renal angiomyolipomas in patients with LAM. Within individuals who had tissue available from both lung and kidney, mutations were absent in normal tissue but were identical in AML and LAM lesions [40-42]. Subsequent studies have identified identical mutations and LOH for TSC2 in multiple sites, including the lung, angiomyolipomas, chyle, and urine, suggesting clonal origins [43,47,51-53]. For those in whom TSC2 mutations are not identified, LAM may be due to mutations in TSC1 genes or other unexplored pathogenetic mechanisms. (See "Tuberous sclerosis complex: Clinical features".)

Role of estrogen — Clinical and biologic data support a critical role for female hormones in LAM induction or progression. This role is supported by the predilection of LAM for women (particularly those of childbearing age), reports of disease exacerbations during pregnancy and in response to exogenous estrogens (eg, oral contraception, fertility treatments), cyclical worsening of LAM symptoms in relationship with the menstrual cycle in a subset of LAM patients, the presence of hormonal receptors on LAM cells (picture 4), and a slower decline in lung function after menopause [54-61].

Estrogen has been the primary focus in most LAM studies exploring hormonal influences on disease pathogenesis. Estrogen promotes the proliferation of tuberin null mouse leiomyoma cells as well as immortalized LAM-like cells derived from human kidney [62]. In addition, estrogen promotes the survival and lung metastasis of tuberin-null cells in rodent models [63]. In a model of progesterone receptor driven deletion of TSC2 in the uterus, growth of myometrial tumors and secretion of proteolytic enzymes were estrogen-dependent [64,65]. Estrogen has also been shown to promote tuberin null cell invasiveness via matrix metalloproteinases, and through cooperative interactions between the extracellular signal regulated kinase (ERK) and mechanistic target of rapamycin (mTOR) pathways that result in epithelial mesenchymal transition [66,67]. (See 'Perivascular epithelioid cell (PEComa)' below.)

Prolactin is elevated in the serum of LAM patients and is expressed in LAM involved tissues. Higher levels of serum prolactin are associated with greater rates of lung function decline and pneumothorax [68]. Another study reported that loss of TSC2 enhances expression of prolactin receptors and promotes the growth and invasiveness of tuberin null cells [69].

Few studies have addressed the role of hormones other than estrogen and prolactin in LAM progression. While multiple investigators have described the presence of progesterone receptors in LAM lung, their role remains unclear [54,69,70]. Although there have been no controlled trials, the most widely quoted retrospective studies did not demonstrate a beneficial effect of progesterone on disease progression [57]. (See "Sporadic lymphangioleiomyomatosis: Treatment and prognosis", section on 'Statins, doxycycline, hormone manipulation, hydroxychloroquine, celecoxib, resveratrol'.)

Growth factor stimulation — The increased expression of growth factors including insulin like growth factor, insulin like growth factor binding protein 4 and 5, platelet-derived growth factor, fibroblast growth factor, endothelin and vascular endothelial growth factor have all been identified in serum or immunohistochemically in LAM tissue [71-74]. Although their pathogenetic role is unknown, these growth factors presumably stimulate LAM cell growth but may also play a role in tissue differentiation and migration.

Abnormal lymphangiogenesis — Lymphatic clefts (ie, malformed vessels) are present in pulmonary LAM nodules, and LAM is known to involve lymph nodes and lymphatic channels [75-77]. The thoracic duct is often extensively infiltrated by LAM at autopsy [78], suggesting a model for spread based on cycles of implantation, proliferation, and budding that allows LAM cells to "leap frog" toward the venous system in the neck and ultimately reach the pulmonary microvasculature [77,79]. LAM cell clusters (LCCs), comprised of a spherical collection of LAM cells enveloped by a single layer of lymphatic endothelial cells, are found within the lymphatic lumen and in chylous accumulations in the chest and abdomen. Chylous effusions and other lymphatic manifestations can occur in approximately 20 percent of patients with sporadic LAM [80,81]. The lymphatic manifestations of LAM are discussed separately. (see "Sporadic lymphangioleiomyomatosis: Clinical presentation and diagnostic evaluation", section on 'Cytology' and "Sporadic lymphangioleiomyomatosis: Clinical presentation and diagnostic evaluation", section on 'Lymphatic manifestations')

LAM involvement of these tissues is associated with the expression of lymphangiogenic proteins, including vascular endothelial growth factor (VEGF)-C and VEGF–D, and their receptors, VEGFR-2 and -3, as well as with other lymphatic markers, lymphatic vessel endothelial receptor-1 (LYVE-1) and podoplanin [76,80-86]. It has also been postulated that VEGF-D, which is elevated in the serum of women with LAM, may promote lymphangiogenesis and the shedding of LCCs [80,85]. (See "Sporadic lymphangioleiomyomatosis: Clinical presentation and diagnostic evaluation", section on 'Vascular endothelial growth factor-D positive'.)

Tissue destruction-cyst formation — The mechanism of cyst formation in pulmonary LAM is incompletely understood. Some groups have postulated that cyst formation is due to smooth muscle cell proliferation within/around the airways, creating a "ball-valve" obstruction that leads to distention of the terminal airspaces, or that tissue degradation is related to an imbalance of proteases and anti-proteases [87-96]. Another theory for the destructive lung remodeling in LAM is that LAM cells that invade the lung and express lymphangiogenic growth factors, such as VEGF-D, drive a program of "frustrated lymphangiogenesis" that leads to cyst formation (figure 2) [79].

Pulmonary LAM nodules contain matrix metalloproteinase (MMP)-1, -2, -3, -9, -11, and -14 as well as MMP activators and MMP inhibitors, TIMP-1 and TIMP-2 [92,94-96]. MMP-2 [97] and MMP-9 [98] have also been reported to be elevated in serum. Elevated expression of cathepsin K, a serine protease, has also been reported in LAM lung lesions [99]. It is unknown which of these and other proteases are directly involved in cyst formation, cell invasion, cell migration, differentiation, or remodeling [100].

Using quantitative microCT and histology of LAM lungs, one study demonstrated a reduced number of small airways (from generation 7 on), a phenomenon that may contribute to the airflow obstruction seen in LAM [101].

Other

Recruitment of stromal cells — Genetic studies indicate that only a fraction of cells (in some cases <10 percent) within the LAM lesions contain mutations in TSC genes, and the remaining cells are likely recruited into the LAM lesions [102]. Recruited cells include fibroblasts [103], mast cells [71], and neutrophils and lymphocytes [102]. As with all cancers, cooperative interactions between mutant cells and stromal cells play a major role in disease pathogenesis [104]. Their exact role in LAM remains undetermined.

Evasion of immune system — Tumor cells evade destruction by the innate immune system by expression of checkpoint inhibitor proteins (eg, programmed cell death protein 1 [PD-1]) that suppress T lymphocyte mediated killing [105-107]. LAM and AML cells have been reported to exhibit [108-110] increased expression of natural killer (NK) cell ligands [108] and PD-L1 [110]. Staining of PD-1 on T cells within the LAM lesion has also been reported [111]. Anti-PD-1 antibody suppresses tumor growth of TSC deficient cells in mouse models of LAM [109]. A case report of effective treatment of a PECOMA with checkpoint inhibitor therapy provides optimism that this approach could be applied to LAM [112].

Modulation of autophagy — As nutrients become limiting, cells regulate their metabolic state to sustain viability. A cellular program of bulk degradation of organelles and macromolecules called autophagy is deployed to generate substrate for generation of cellular biomass. Inappropriate activation of mTOR by TSC mutations simulates a fed state, and both stimulates proliferation and suppresses autophagy [113].

Altered cellular metabolism — mTOR is part of a complex (together with Raptor, Rictor, Sin) that forms the nexus of multiple cellular signaling networks and functions to balance metabolic supply with demand [114]. Mutations in mTOR mediate altered cellular metabolism that facilitate neoplastic growth, including a shift from oxidative phosphorylation to glycolysis as a mechanism to increase availability of metabolic substrate for generation of cellular biomass and for tolerance of anaerobic environments [115]. LAM cells take up glutamine as an alternative carbon source, which provides an adaptive mechanism to fuel mTOR activity when glucose pathways are insufficient to support it or are blocked [116]. In addition, activation of mTOR drives flux through purine and pyrimidine biosynthesis pathways and the pentose phosphate shunt pathway to provide substrate for the synthesis of polynucleotides, and activates super conserved receptor expressed in brain (SREB) which is required for the biosynthesis of fatty acids and cholesterol [117,118]. Activated mTOR stimulates ribonucleic acid (RNA) translation by regulating the assembly of the eukaryotic translation initiation factor (eIF4F) complex and phosphorylating downstream targets including eIF4F binding proteins (4E-BPs) and ribosomal protein S6 kinases [119]. Altered polyamine metabolism has also been reported in LAM cells, although several flaws in the study cast doubt on the validity of this effect [120].

Miscellaneous — Other factors that have been found to be abnormally expressed in LAM include:

High Mobility Group 2A [121]

Adipocyte phospholipase A2 [122]

Survivin [123]

Osteopontin [124]

HIF1 alpha targets

Erythropoietin [125]

Metastasis and invasion markers (eg, CD44V and CXCR4) [124,126]

Src kinase [127]

Plasminogen system [128]

Urotensin [129]

CA-125 [130]

Nuclear receptor subfamily 2 group F member 2 (NR2F2) [131]

LAM CELL ORIGIN — The origin of the LAM cells, which are included in the family of neoplasms with perivascular epithelioid differentiation (PEComa), is unknown. Theories for the source of LAM cells that populate the lung include the uterus [65,132], angiomyolipomas [133], bone marrow [134], lymphatic system [40], or the neural crest [135]. Evidence to support potential sources is described in the sections below.

Perivascular epithelioid cell (PEComa) — LAM is classified by the World Health Organization (WHO) as a lung tumor, in the perivascular epithelioid cell (ie, PEComa) family [136-138], which also includes angiomyolipomas, clear cell “sugar” tumors, and other rare neoplasms of visceral organs. The classification of LAM as a low grade neoplasm is based upon the observations that LAM cells exhibit cellular and genetic fingerprints similar to neoplasms [137], have loss of heterozygosity (LOH) for the tumor suppressor gene TSC2, can recur in transplanted lung, exhibit invasive/immune evasive/migratory/metastatic behavior [35,41,51,80,99,139], and have the capacity to destroy remote organs.

Neural crest (melanoma antigen expression) — A monoclonal antibody to human melanoma black (HMB)-45 (also known as melanosome specific antigen), derived from melanoma hybridomas, is the classic marker for the diagnosis of LAM on lung biopsy (picture 5) [140,141] but can also be found in angiomyolipomas and some uterine PEComas [142-144].

The antigen to which HMB-45 binds has total cDNA homology with the gene, premelanosome protein (PMEL17, located on chromosome 12), the product of which is involved in the synthesis of melanin and catecholamines from the amino acid tyrosine, suggesting a neuroendocrine origin.

Immunohistochemical staining of LAM lung or cells in culture have shown that pulmonary LAM cells aberrantly express the epitope for HMB-45 as well as other melanoma-derived antigens, further supporting the hypothesis that LAM cells may be of neuroendocrine origin and may be responsible for LAM cell differentiation [135,145-148]. However, immunoreactivity for HMB-45 can be variable, and even absent in LAM lesions. LAM cells also stain for other melanocytic proteins, including PNL2 [149], MART1 [145], and aPep13h [147].  

Uterus — The uterus has been suggested as the source of origin of LAM cells by multiple observations. Rats with naturally occurring Tsc2 mutations develop uterine myometrial tumors with features similar to LAM. In addition, mice with inactivation of Tsc2 in the uterus develop smooth muscle tumors that metastasize to the lungs [64,65]. Although multiple case reports of LAM in the uterus have appeared in the literature, it remains possible that the uterus is another target for metastasis rather than the primary source [132,150]. A single cell transcriptomics analysis conducted on lungs and uteri obtained from LAM patients has shown that LAM cells in lung and uterus are morphologically indistinguishable and share similar gene expression profiles and biallelic Tsc2 mutations, supporting a uterine origin of LAM cells [151].

Other — Other purported origins for the LAM cell include the lymphatics [86], and the kidney (eg, from angiomyolipomas) [133]. Data to support these origins come from studies that report that LAM cells in the lung are morphologically or genetically similar to neoplastic cells in these organs [40-44,51,80,86,152].

RESEARCH MODELS — Experimental modeling is critical for effective LAM research. While multiple cellular and animal models of TSC and LAM have been developed, all have limitations. The most commonly used cell models for LAM are ELT3 cells, a uterine leiomyoma cell line from the Eker rat which has a germ line mutation in Tsc2 [153], Tsc null murine embryonic fibroblast cell lines [154], 621-101 cells, a transformed cell line from a renal angiomyolipoma [155], and low passage mixed cultures isolated from LAM lung at the time of transplant [48]. Animal models for LAM that have been reported include uterus specific Tsc2 deletion [64], subcutaneous xenograft models [63,156], and intrapulmonary Tsc2 null cell administration through the endonasal route [157].

A LAM cell atlas has been created which facilitates collaborative research into the pathogenesis of LAM and can be found here. [158]

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

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Lymphangioleiomyomatosis (The Basics)")

PATIENT PERSPECTIVE TOPIC — Patient perspectives are provided for selected disorders to help clinicians better understand the patient experience and patient concerns. These narratives may offer insights into patient values and preferences not included in other UpToDate topics. (See "Patient perspective: Lymphangioleiomyomatosis (LAM)".)

SUMMARY

Lymphangioleiomyomatosis (LAM) is a rare multisystem disease with a female predilection that is characterized by cyst formation in the lungs and the proliferation of immature smooth muscle with features of perivascular epithelioid cells (LAM cells) in affected organs. The term sporadic LAM is used for patients with LAM who do not have tuberous sclerosis complex (TSC), while TSC-LAM refers to LAM that is associated with TSC. (See 'Introduction' above.)

Estimates of the prevalence of sporadic LAM vary, ranging from 1 to 14 per million women in the general population with the highest rates in women with TSC (more than 30 percent). (See 'Epidemiology' above and "Tuberous sclerosis complex associated lymphangioleiomyomatosis in adults", section on 'Clinical manifestations'.)

The exact pathogenesis of LAM is unknown, but appears to involve excessive proliferation of LAM cells due to loss of functioning TSC genes. Loss of heterozygosity and somatic mutations for the TSC2 gene are found in affected tissue in the majority of sporadic LAM cases, but TSC mutations are not present in the germ line, so sporadic LAM cannot be inherited. The absence of tuberin prevents formation of the hamartin-tuberin complex, which regulates mechanistic (previously mammalian) target of rapamycin (mTOR), a molecule that controls cell growth and cell size. (See 'Tuberous sclerosis gene mutations' above and "Tuberous sclerosis complex: Clinical features".)

Hormonal fluxes (eg, pregnancy, exogenous estrogen) play a key role in the pathogenesis of LAM. In particular, an important role for estrogen is suggested by the presence of estrogen receptors on LAM cells and a slower decline in lung function after menopause. Estrogen may also play a role in cell invasion and migration. (See 'Role of estrogen' above.)

Abnormal lymphangiogenesis may play role in cell migration and disease progression as evidenced by lymphatic differentiation within LAM lesions and extensive involvement of lymphatic channels by LAM cells. (See 'Abnormal lymphangiogenesis' above.)

The mechanism of cyst formation in LAM lung is unclear but may involve bronchial obstruction, tissue destruction by proteases, and/or dysregulated lymphangiogenesis. (See 'Tissue destruction-cyst formation' above.)

LAM pathogenesis also involves recruitment of stromal cells, metabolic reprogramming of the LAM cells (eg, upregulation of glycolysis, increased synthesis of lipids, protein, and nucleotides), immune evasion by upregulation of immune checkpoint proteins, promotion of cell survival, and inhibition of autophagy. (See 'Other' above.)

The exact origin of LAM cells that populate the lung is unknown. LAM is included in the family of neoplasms with perivascular epithelioid differentiation (PEComa). Accumulating evidence suggests the uterus to be a promising organ of origin of LAM cells. Other evidence, such as cell surface positivity for the human melanoma black (HMB)-45 antigen, suggests a neuroendocrine origin. (See 'Lam cell origin' above and 'Research models' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Talmadge E King, Jr, MD, who contributed to earlier versions of this topic review.

  1. Thway K, Fisher C. PEComa: morphology and genetics of a complex tumor family. Ann Diagn Pathol 2015; 19:359.
  2. Kalassian KG, Doyle R, Kao P, et al. Lymphangioleiomyomatosis: new insights. Am J Respir Crit Care Med 1997; 155:1183.
  3. Sullivan EJ. Lymphangioleiomyomatosis: a review. Chest 1998; 114:1689.
  4. NHLBI Workshop Summary. Report of workshop on lymphangioleiomyomatosis. National Heart, Lung, and Blood Institute. Am J Respir Crit Care Med 1999; 159:679.
  5. Johnson S. Rare diseases. 1. Lymphangioleiomyomatosis: clinical features, management and basic mechanisms. Thorax 1999; 54:254.
  6. Moss J, Avila NA, Barnes PM, et al. Prevalence and clinical characteristics of lymphangioleiomyomatosis (LAM) in patients with tuberous sclerosis complex. Am J Respir Crit Care Med 2001; 164:669.
  7. Hancock E, Osborne J. Lymphangioleiomyomatosis: a review of the literature. Respir Med 2002; 96:1.
  8. Harknett EC, Chang WY, Byrnes S, et al. Use of variability in national and regional data to estimate the prevalence of lymphangioleiomyomatosis. QJM 2011; 104:971.
  9. Baldi BG, Freitas CS, Araujo MS, et al. Clinical course and characterisation of lymphangioleiomyomatosis in a Brazilian reference centre. Sarcoidosis Vasc Diffuse Lung Dis 2014; 31:129.
  10. Park HY, Nam HS, Chung MP, et al. A nationwide survey of lymphangioleiomyomatosis in Korea: recent increase in newly diagnosed patients. J Korean Med Sci 2010; 25:1182.
  11. Hayashida M, Seyama K, Inoue Y, et al. The epidemiology of lymphangioleiomyomatosis in Japan: a nationwide cross-sectional study of presenting features and prognostic factors. Respirology 2007; 12:523.
  12. Johnson SR, Tattersfield AE. Clinical experience of lymphangioleiomyomatosis in the UK. Thorax 2000; 55:1052.
  13. Ryu JH, Moss J, Beck GJ, et al. The NHLBI lymphangioleiomyomatosis registry: characteristics of 230 patients at enrollment. Am J Respir Crit Care Med 2006; 173:105.
  14. The LAM Foundation. https://www.thelamfoundation.org/Newly-Diagnosed/Learning-About-Lam/About-LAM (Accessed on January 30, 2020).
  15. Kristof AS, Li PZ, Major P, Landry JS. Lymphangioleiomyomatosis and Tuberous Sclerosis Complex in Quebec: Prevalence and Health-care Utilization. Chest 2015; 148:444.
  16. Aubry MC, Myers JL, Ryu JH, et al. Pulmonary lymphangioleiomyomatosis in a man. Am J Respir Crit Care Med 2000; 162:749.
  17. Miyake M, Tateishi U, Maeda T, et al. Pulmonary lymphangioleiomyomatosis in a male patient with tuberous sclerosis complex. Radiat Med 2005; 23:525.
  18. Kim NR, Chung MP, Park CK, et al. Pulmonary lymphangioleiomyomatosis and multiple hepatic angiomyolipomas in a man. Pathol Int 2003; 53:231.
  19. Schiavina M, Di Scioscio V, Contini P, et al. Pulmonary lymphangioleiomyomatosis in a karyotypically normal man without tuberous sclerosis complex. Am J Respir Crit Care Med 2007; 176:96.
  20. Wakida K, Watanabe Y, Kumasaka T, et al. Lymphangioleiomyomatosis in a Male. Ann Thorac Surg 2015; 100:1105.
  21. Yamanaka S, Mizobuchi T, Kurihara M. Two Kinds of Cystic Lung Lesions with Pulmonary Lymphangioleiomyomatosis in a Male. Ann Thorac Cardiovasc Surg 2017; 23:36.
  22. Muzykewicz DA, Sharma A, Muse V, et al. TSC1 and TSC2 mutations in patients with lymphangioleiomyomatosis and tuberous sclerosis complex. J Med Genet 2009; 46:465.
  23. Adriaensen ME, Schaefer-Prokop CM, Duyndam DA, et al. Radiological evidence of lymphangioleiomyomatosis in female and male patients with tuberous sclerosis complex. Clin Radiol 2011; 66:625.
  24. Restrepo-Gualteros SM, Rodriguez-Martinez CE, Jaramillo-Barberi LE, et al. Adolescent form of sporadic lymphangioleiomyomatosis (S-LAM). Allergol Immunopathol (Madr) 2015; 43:111.
  25. Richter MJ, Baumgartner R, Leuchte HH. [Chylothorax in an 87-year-old patient]. Internist (Berl) 2013; 54:105.
  26. Kebria M, Black D, Borelli C, et al. Primary retroperitoneal lymphangioleiomyomatosis in a postmenopausal woman: a case report and review of the literature. Int J Gynecol Cancer 2007; 17:528.
  27. Zanella A, Toppan P, Nitti D, Lise M. Pulmonary lymphangioleiomyomatosis: a case report in postmenopausal woman treated with pleurodesis and progesterone (medroxyprogesterone acetate). Tumori 1996; 82:96.
  28. Nagy B, Nábrády Z, Nemes Z. Pulmonary lymphangiomyomatosis in a preadolescent girl. N Engl J Med 1998; 338:473.
  29. Ho TB, Hull JH, Hughes NC. An 86-year-old female with lymphangioleiomyomatosis. Eur Respir J 2006; 28:1065.
  30. Ciftci AO, Sanlialp I, Tanyel FC, Buyukpamukçu N. The association of pulmonary lymphangioleiomyomatosis with renal and hepatic angiomyolipomas in a prepubertal girl: a previously unreported entity. Respiration 2007; 74:335.
  31. Harari S, Torre O, Cassandro R, Moss J. The changing face of a rare disease: lymphangioleiomyomatosis. Eur Respir J 2015; 46:1471.
  32. Taveira-DaSilva AM, Moss J. Clinical features, epidemiology, and therapy of lymphangioleiomyomatosis. Clin Epidemiol 2015; 7:249.
  33. Meraj R, Wikenheiser-Brokamp KA, Young LR, McCormack FX. Lymphangioleiomyomatosis: new concepts in pathogenesis, diagnosis, and treatment. Semin Respir Crit Care Med 2012; 33:486.
  34. Green AJ, Smith M, Yates JR. Loss of heterozygosity on chromosome 16p13.3 in hamartomas from tuberous sclerosis patients. Nat Genet 1994; 6:193.
  35. Goncharova EA, Goncharov DA, Lim PN, et al. Modulation of cell migration and invasiveness by tumor suppressor TSC2 in lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2006; 34:473.
  36. Rosner M, Hanneder M, Siegel N, et al. The tuberous sclerosis gene products hamartin and tuberin are multifunctional proteins with a wide spectrum of interacting partners. Mutat Res 2008; 658:234.
  37. Sengupta S, Peterson TR, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell 2010; 40:310.
  38. Sarbassov DD, Ali SM, Kim DH, et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 2004; 14:1296.
  39. Cudzilo CJ, Szczesniak RD, Brody AS, et al. Lymphangioleiomyomatosis screening in women with tuberous sclerosis. Chest 2013; 144:578.
  40. Smolarek TA, Wessner LL, McCormack FX, et al. Evidence that lymphangiomyomatosis is caused by TSC2 mutations: chromosome 16p13 loss of heterozygosity in angiomyolipomas and lymph nodes from women with lymphangiomyomatosis. Am J Hum Genet 1998; 62:810.
  41. Carsillo T, Astrinidis A, Henske EP. Mutations in the tuberous sclerosis complex gene TSC2 are a cause of sporadic pulmonary lymphangioleiomyomatosis. Proc Natl Acad Sci U S A 2000; 97:6085.
  42. Astrinidis A, Khare L, Carsillo T, et al. Mutational analysis of the tuberous sclerosis gene TSC2 in patients with pulmonary lymphangioleiomyomatosis. J Med Genet 2000; 37:55.
  43. Cai X, Pacheco-Rodriguez G, Fan QY, et al. Phenotypic characterization of disseminated cells with TSC2 loss of heterozygosity in patients with lymphangioleiomyomatosis. Am J Respir Crit Care Med 2010; 182:1410.
  44. Yu J, Astrinidis A, Henske EP. Chromosome 16 loss of heterozygosity in tuberous sclerosis and sporadic lymphangiomyomatosis. Am J Respir Crit Care Med 2001; 164:1537.
  45. Sato T, Seyama K, Fujii H, et al. Mutation analysis of the TSC1 and TSC2 genes in Japanese patients with pulmonary lymphangioleiomyomatosis. J Hum Genet 2002; 47:20.
  46. Ando K, Fujino N, Mitani K, et al. Isolation of individual cellular components from lung tissues of patients with lymphangioleiomyomatosis. Am J Physiol Lung Cell Mol Physiol 2016; 310:L899.
  47. Fujita A, Ando K, Kobayashi E, et al. Detection of low-prevalence somatic TSC2 mutations in sporadic pulmonary lymphangioleiomyomatosis tissues by deep sequencing. Hum Genet 2016; 135:61.
  48. Goncharova EA, Goncharov DA, Eszterhas A, et al. Tuberin regulates p70 S6 kinase activation and ribosomal protein S6 phosphorylation. A role for the TSC2 tumor suppressor gene in pulmonary lymphangioleiomyomatosis (LAM). J Biol Chem 2002; 277:30958.
  49. Ogórek B, Hamieh L, Lasseter K, et al. Generalised mosaicism for TSC2 mutation in isolated lymphangioleiomyomatosis. Eur Respir J 2019; 54.
  50. Jones AC, Shyamsundar MM, Thomas MW, et al. Comprehensive mutation analysis of TSC1 and TSC2-and phenotypic correlations in 150 families with tuberous sclerosis. Am J Hum Genet 1999; 64:1305.
  51. Crooks DM, Pacheco-Rodriguez G, DeCastro RM, et al. Molecular and genetic analysis of disseminated neoplastic cells in lymphangioleiomyomatosis. Proc Natl Acad Sci U S A 2004; 101:17462.
  52. Bittmann I, Rolf B, Amann G, Löhrs U. Recurrence of lymphangioleiomyomatosis after single lung transplantation: new insights into pathogenesis. Hum Pathol 2003; 34:95.
  53. Bittmann I, Dose TB, Müller C, et al. Lymphangioleiomyomatosis: recurrence after single lung transplantation. Hum Pathol 1997; 28:1420.
  54. Berger U, Khaghani A, Pomerance A, et al. Pulmonary lymphangioleiomyomatosis and steroid receptors. An immunocytochemical study. Am J Clin Pathol 1990; 93:609.
  55. Ohori NP, Yousem SA, Sonmez-Alpan E, Colby TV. Estrogen and progesterone receptors in lymphangioleiomyomatosis, epithelioid hemangioendothelioma, and sclerosing hemangioma of the lung. Am J Clin Pathol 1991; 96:529.
  56. Matsui K, Takeda K, Yu ZX, et al. Downregulation of estrogen and progesterone receptors in the abnormal smooth muscle cells in pulmonary lymphangioleiomyomatosis following therapy. An immunohistochemical study. Am J Respir Crit Care Med 2000; 161:1002.
  57. Taveira-DaSilva AM, Stylianou MP, Hedin CJ, et al. Decline in lung function in patients with lymphangioleiomyomatosis treated with or without progesterone. Chest 2004; 126:1867.
  58. Brunelli A, Catalini G, Fianchini A. Pregnancy exacerbating unsuspected mediastinal lymphangioleiomyomatosis and chylothorax. Int J Gynaecol Obstet 1996; 52:289.
  59. Yano S. Exacerbation of pulmonary lymphangioleiomyomatosis by exogenous oestrogen used for infertility treatment. Thorax 2002; 57:1085.
  60. Kanemura H, Jinta T, Tamura T. Pulmonary lymphangioleiomyomatosis and fertility treatment. QJM 2018; 111:123.
  61. Gupta N, Lee HS, Ryu JH, et al. The NHLBI LAM Registry: Prognostic Physiologic and Radiologic Biomarkers Emerge From a 15-Year Prospective Longitudinal Analysis. Chest 2019; 155:288.
  62. Yu J, Astrinidis A, Howard S, Henske EP. Estradiol and tamoxifen stimulate LAM-associated angiomyolipoma cell growth and activate both genomic and nongenomic signaling pathways. Am J Physiol Lung Cell Mol Physiol 2004; 286:L694.
  63. Yu JJ, Robb VA, Morrison TA, et al. Estrogen promotes the survival and pulmonary metastasis of tuberin-null cells. Proc Natl Acad Sci U S A 2009; 106:2635.
  64. Prizant H, Taya M, Lerman I, et al. Estrogen maintains myometrial tumors in a lymphangioleiomyomatosis model. Endocr Relat Cancer 2016; 23:265.
  65. Prizant H, Sen A, Light A, et al. Uterine-specific loss of Tsc2 leads to myometrial tumors in both the uterus and lungs. Mol Endocrinol 2013; 27:1403.
  66. Glassberg MK, Elliot SJ, Fritz J, et al. Activation of the estrogen receptor contributes to the progression of pulmonary lymphangioleiomyomatosis via matrix metalloproteinase-induced cell invasiveness. J Clin Endocrinol Metab 2008; 93:1625.
  67. Gu X, Yu JJ, Ilter D, et al. Integration of mTOR and estrogen-ERK2 signaling in lymphangioleiomyomatosis pathogenesis. Proc Natl Acad Sci U S A 2013; 110:14960.
  68. Terasaki Y, Yahiro K, Pacheco-Rodriguez G, et al. Effects of prolactin on TSC2-null Eker rat cells and in pulmonary lymphangioleiomyomatosis. Am J Respir Crit Care Med 2010; 182:531.
  69. Alkharusi A, Lesma E, Ancona S, et al. Role of Prolactin Receptors in Lymphangioleiomyomatosis. PLoS One 2016; 11:e0146653.
  70. Gao L, Yue MM, Davis J, et al. In pulmonary lymphangioleiomyomatosis expression of progesterone receptor is frequently higher than that of estrogen receptor. Virchows Arch 2014; 464:495.
  71. Inoue Y, King TE Jr, Barker E, et al. Basic fibroblast growth factor and its receptors in idiopathic pulmonary fibrosis and lymphangioleiomyomatosis. Am J Respir Crit Care Med 2002; 166:765.
  72. Clements D, Asprey SL, McCulloch TA, et al. Analysis of the oestrogen response in an angiomyolipoma derived xenograft model. Endocr Relat Cancer 2009; 16:59.
  73. Lesma E, Grande V, Carelli S, et al. Isolation and growth of smooth muscle-like cells derived from tuberous sclerosis complex-2 human renal angiomyolipoma: epidermal growth factor is the required growth factor. Am J Pathol 2005; 167:1093.
  74. Ding M, Bruick RK, Yu Y. Secreted IGFBP5 mediates mTORC1-dependent feedback inhibition of IGF-1 signalling. Nat Cell Biol 2016; 18:319.
  75. Davis JM, Hyjek E, Husain AN, et al. Lymphatic endothelial differentiation in pulmonary lymphangioleiomyomatosis cells. J Histochem Cytochem 2013; 61:580.
  76. Gupta R, Kitaichi M, Inoue Y, et al. Lymphatic manifestations of lymphangioleiomyomatosis. Lymphology 2014; 47:106.
  77. Seyama K, Kumasaka T, Kurihara M, et al. Lymphangioleiomyomatosis: a disease involving the lymphatic system. Lymphat Res Biol 2010; 8:21.
  78. Cornog JL Jr, Enterline HT. Lymphangiomyoma, a benign lesion of chyliferous lymphatics synonymous with lymphangiopericytoma. Cancer 1966; 19:1909.
  79. Henske EP, McCormack FX. Lymphangioleiomyomatosis - a wolf in sheep's clothing. J Clin Invest 2012; 122:3807.
  80. Kumasaka T, Seyama K, Mitani K, et al. Lymphangiogenesis-mediated shedding of LAM cell clusters as a mechanism for dissemination in lymphangioleiomyomatosis. Am J Surg Pathol 2005; 29:1356.
  81. Kumasaka T, Seyama K, Mitani K, et al. Lymphangiogenesis in lymphangioleiomyomatosis: its implication in the progression of lymphangioleiomyomatosis. Am J Surg Pathol 2004; 28:1007.
  82. Young L, Lee HS, Inoue Y, et al. Serum VEGF-D a concentration as a biomarker of lymphangioleiomyomatosis severity and treatment response: a prospective analysis of the Multicenter International Lymphangioleiomyomatosis Efficacy of Sirolimus (MILES) trial. Lancet Respir Med 2013; 1:445.
  83. Seyama K, Kumasaka T, Souma S, et al. Vascular endothelial growth factor-D is increased in serum of patients with lymphangioleiomyomatosis. Lymphat Res Biol 2006; 4:143.
  84. Glasgow CG, Avila NA, Lin JP, et al. Serum vascular endothelial growth factor-D levels in patients with lymphangioleiomyomatosis reflect lymphatic involvement. Chest 2009; 135:1293.
  85. Mitani K, Kumasaka T, Takemura H, et al. Cytologic, immunocytochemical and ultrastructural characterization of lymphangioleiomyomatosis cell clusters in chylous effusions of patients with lymphangioleiomyomatosis. Acta Cytol 2009; 53:402.
  86. Yue M, Pacheco G, Cheng T, et al. Evidence Supporting a Lymphatic Endothelium Origin for Angiomyolipoma, a TSC2(-) Tumor Related to Lymphangioleiomyomatosis. Am J Pathol 2016; 186:1825.
  87. Fukuda Y, Kawamoto M, Yamamoto A, et al. Role of elastic fiber degradation in emphysema-like lesions of pulmonary lymphangiomyomatosis. Hum Pathol 1990; 21:1252.
  88. Corrin B, Liebow AA, Friedman PJ. Pulmonary lymphangiomyomatosis. A review. Am J Pathol 1975; 79:348.
  89. Sobonya RE, Quan SF, Fleishman JS. Pulmonary lymphangioleiomyomatosis: quantitative analysis of lesions producing airflow limitation. Hum Pathol 1985; 16:1122.
  90. McCormack FX. Lymphangioleiomyomatosis: a clinical update. Chest 2008; 133:507.
  91. Abbott GF, Rosado-de-Christenson ML, Frazier AA, et al. From the archives of the AFIP: lymphangioleiomyomatosis: radiologic-pathologic correlation. Radiographics 2005; 25:803.
  92. Matsui K, Takeda K, Yu ZX, et al. Role for activation of matrix metalloproteinases in the pathogenesis of pulmonary lymphangioleiomyomatosis. Arch Pathol Lab Med 2000; 124:267.
  93. Krymskaya VP, Shipley JM. Lymphangioleiomyomatosis: a complex tale of serum response factor-mediated tissue inhibitor of metalloproteinase-3 regulation. Am J Respir Cell Mol Biol 2003; 28:546.
  94. Zhe X, Yang Y, Jakkaraju S, Schuger L. Tissue inhibitor of metalloproteinase-3 downregulation in lymphangioleiomyomatosis: potential consequence of abnormal serum response factor expression. Am J Respir Cell Mol Biol 2003; 28:504.
  95. Odajima N, Betsuyaku T, Nasuhara Y, et al. Matrix metalloproteinases in blood from patients with LAM. Respir Med 2009; 103:124.
  96. Goncharova EA, Goncharov DA, Fehrenbach M, et al. Prevention of alveolar destruction and airspace enlargement in a mouse model of pulmonary lymphangioleiomyomatosis (LAM). Sci Transl Med 2012; 4:154ra134.
  97. Chang WY, Cane JL, Blakey JD, et al. Clinical utility of diagnostic guidelines and putative biomarkers in lymphangioleiomyomatosis. Respir Res 2012; 13:34.
  98. Pimenta SP, Baldi BG, Acencio MM, et al. Doxycycline use in patients with lymphangioleiomyomatosis: safety and efficacy in metalloproteinase blockade. J Bras Pneumol 2011; 37:424.
  99. Chilosi M, Pea M, Martignoni G, et al. Cathepsin-k expression in pulmonary lymphangioleiomyomatosis. Mod Pathol 2009; 22:161.
  100. Bühling F, Groneberg D, Welte T. Proteases and their role in chronic inflammatory lung diseases. Curr Drug Targets 2006; 7:751.
  101. Verleden SE, Vanstapel A, De Sadeleer L, et al. Quantitative analysis of airway obstruction in lymphangioleiomyomatosis. Eur Respir J 2020; 56.
  102. Badri KR, Gao L, Hyjek E, et al. Exonic mutations of TSC2/TSC1 are common but not seen in all sporadic pulmonary lymphangioleiomyomatosis. Am J Respir Crit Care Med 2013; 187:663.
  103. Clements D, Dongre A, Krymskaya VP, Johnson SR. Wild type mesenchymal cells contribute to the lung pathology of lymphangioleiomyomatosis. PLoS One 2015; 10:e0126025.
  104. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144:646.
  105. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015; 373:123.
  106. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med 2015; 373:1627.
  107. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 2017; 389:255.
  108. Osterburg AR, Nelson RL, Yaniv BZ, et al. NK cell activating receptor ligand expression in lymphangioleiomyomatosis is associated with lung function decline. JCI Insight 2016; 1:e87270.
  109. Liu HJ, Lizotte PH, Du H, et al. TSC2-deficient tumors have evidence of T cell exhaustion and respond to anti-PD-1/anti-CTLA-4 immunotherapy. JCI Insight 2018; 3.
  110. Maisel K, Merrilees MJ, Atochina-Vasserman EN, et al. Immune Checkpoint Ligand PD-L1 Is Upregulated in Pulmonary Lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2018; 59:723.
  111. Liu HJ, Krymskaya VP, Henske EP. Immunotherapy for Lymphangioleiomyomatosis and Tuberous Sclerosis: Progress and Future Directions. Chest 2019; 156:1062.
  112. Lattanzi M, Deng FM, Chiriboga LA, et al. Durable response to anti-PD-1 immunotherapy in epithelioid angiomyolipoma: a report on the successful treatment of a rare malignancy. J Immunother Cancer 2018; 6:97.
  113. Yu J, Parkhitko AA, Henske EP. Mammalian target of rapamycin signaling and autophagy: roles in lymphangioleiomyomatosis therapy. Proc Am Thorac Soc 2010; 7:48.
  114. Choo AY, Kim SG, Vander Heiden MG, et al. Glucose addiction of TSC null cells is caused by failed mTORC1-dependent balancing of metabolic demand with supply. Mol Cell 2010; 38:487.
  115. Sun Q, Chen X, Ma J, et al. Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth. Proc Natl Acad Sci U S A 2011; 108:4129.
  116. Fu V, Moroishi T, Guan KL. Glycoholics Anonymous: Cancer Sobers Up with mTORC1. Cancer Cell 2016; 29:432.
  117. Ben-Sahra I, Hoxhaj G, Ricoult SJ, et al. mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle. Science 2016; 351:728.
  118. Ben-Sahra I, Howell JJ, Asara JM, Manning BD. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 2013; 339:1323.
  119. Bhat M, Robichaud N, Hulea L, et al. Targeting the translation machinery in cancer. Nat Rev Drug Discov 2015; 14:261.
  120. Tang Y, El-Chemaly S, Taveira-Dasilva A, et al. Alterations in Polyamine Metabolism in Patients With Lymphangioleiomyomatosis and Tuberous Sclerosis Complex 2-Deficient Cells. Chest 2019; 156:1137.
  121. D'Armiento J, Shiomi T, Marks S, et al. Mesenchymal Tumorigenesis Driven by TSC2 Haploinsufficiency Requires HMGA2 and Is Independent of mTOR Pathway Activation. Cancer Res 2016; 76:844.
  122. Li C, Zhang E, Sun Y, et al. Rapamycin-insensitive up-regulation of adipocyte phospholipase A2 in tuberous sclerosis and lymphangioleiomyomatosis. PLoS One 2014; 9:e104809.
  123. Carelli S, Lesma E, Paratore S, et al. Survivin expression in tuberous sclerosis complex cells. Mol Med 2007; 13:166.
  124. Pacheco-Rodriguez G, Steagall WK, Crooks DM, et al. TSC2 loss in lymphangioleiomyomatosis cells correlated with expression of CD44v6, a molecular determinant of metastasis. Cancer Res 2007; 67:10573.
  125. Ikeda Y, Taveira-DaSilva AM, Pacheco-Rodriguez G, et al. Erythropoietin-driven proliferation of cells with mutations in the tumor suppressor gene TSC2. Am J Physiol Lung Cell Mol Physiol 2011; 300:L64.
  126. Clements D, Markwick LJ, Puri N, Johnson SR. Role of the CXCR4/CXCL12 axis in lymphangioleiomyomatosis and angiomyolipoma. J Immunol 2010; 185:1812.
  127. Tyryshkin A, Bhattacharya A, Eissa NT. SRC kinase is a novel therapeutic target in lymphangioleiomyomatosis. Cancer Res 2014; 74:1996.
  128. Zhe X, Yang Y, Schuger L. Imbalanced plasminogen system in lymphangioleiomyomatosis: potential role of serum response factor. Am J Respir Cell Mol Biol 2005; 32:28.
  129. Kristof AS, You Z, Han YS, Giaid A. Protein expression of urotensin II, urotensin-related peptide and their receptor in the lungs of patients with lymphangioleiomyomatosis. Peptides 2010; 31:1511.
  130. Glasgow CG, Pacheco-Rodriguez G, Steagall WK, et al. CA-125 in Disease Progression and Treatment of Lymphangioleiomyomatosis. Chest 2018; 153:339.
  131. Kim W, Giannikou K, Dreier JR, et al. A genome-wide association study implicates NR2F2 in lymphangioleiomyomatosis pathogenesis. Eur Respir J 2019; 53.
  132. Hayashi T, Kumasaka T, Mitani K, et al. Prevalence of uterine and adnexal involvement in pulmonary lymphangioleiomyomatosis: a clinicopathologic study of 10 patients. Am J Surg Pathol 2011; 35:1776.
  133. Henske EP. Metastasis of benign tumor cells in tuberous sclerosis complex. Genes Chromosomes Cancer 2003; 38:376.
  134. Krymskaya VP, McCormack FX. Lymphangioleiomyomatosis: A Monogenic Model of Malignancy. Annu Rev Med 2017; 68:69.
  135. Delaney SP, Julian LM, Stanford WL. The neural crest lineage as a driver of disease heterogeneity in Tuberous Sclerosis Complex and Lymphangioleiomyomatosis. Front Cell Dev Biol 2014; 2:69.
  136. Travis WD, Brambilla EW, Burke AP, et al. WHO Classification of Tumours of the Lung, Pleura, Thymus, and Heart, IARC Press, Lyon 2015.
  137. McCormack FX, Travis WD, Colby TV, et al. Lymphangioleiomyomatosis: calling it what it is: a low-grade, destructive, metastasizing neoplasm. Am J Respir Crit Care Med 2012; 186:1210.
  138. Martignoni G, Pea M, Reghellin D, et al. Molecular pathology of lymphangioleiomyomatosis and other perivascular epithelioid cell tumors. Arch Pathol Lab Med 2010; 134:33.
  139. Karbowniczek M, Astrinidis A, Balsara BR, et al. Recurrent lymphangiomyomatosis after transplantation: genetic analyses reveal a metastatic mechanism. Am J Respir Crit Care Med 2003; 167:976.
  140. Chu SC, Horiba K, Usuki J, et al. Comprehensive evaluation of 35 patients with lymphangioleiomyomatosis. Chest 1999; 115:1041.
  141. Grzegorek I, Lenze D, Chabowski M, et al. Immunohistochemical evaluation of pulmonary lymphangioleiomyomatosis. Anticancer Res 2015; 35:3353.
  142. Yavuz E, Cakr C, Tuzlal S, et al. Uterine perivascular epithelioid cell tumor coexisting with pulmonary lymphangioleiomyomatosis and renal angiomyolipoma: a case report. Appl Immunohistochem Mol Morphol 2008; 16:405.
  143. Vang R, Kempson RL. Perivascular epithelioid cell tumor ('PEComa') of the uterus: a subset of HMB-45-positive epithelioid mesenchymal neoplasms with an uncertain relationship to pure smooth muscle tumors. Am J Surg Pathol 2002; 26:1.
  144. Aydin H, Magi-Galluzzi C, Lane BR, et al. Renal angiomyolipoma: clinicopathologic study of 194 cases with emphasis on the epithelioid histology and tuberous sclerosis association. Am J Surg Pathol 2009; 33:289.
  145. Klarquist J, Barfuss A, Kandala S, et al. Melanoma-associated antigen expression in lymphangioleiomyomatosis renders tumor cells susceptible to cytotoxic T cells. Am J Pathol 2009; 175:2463.
  146. Matsumoto Y, Horiba K, Usuki J, et al. Markers of cell proliferation and expression of melanosomal antigen in lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 1999; 21:327.
  147. Valencia JC, Steagall WK, Zhang Y, et al. Antibody αPEP13h reacts with lymphangioleiomyomatosis cells in lung nodules. Chest 2015; 147:771.
  148. Gilbert ER, Eby JM, Hammer AM, et al. Positioning ganglioside D3 as an immunotherapeutic target in lymphangioleiomyomatosis. Am J Pathol 2013; 183:226.
  149. Zhe X, Schuger L. Combined smooth muscle and melanocytic differentiation in lymphangioleiomyomatosis. J Histochem Cytochem 2004; 52:1537.
  150. Taveira-Dasilva AM, Rabel A, Gochuico BR, et al. Prevalence of uterine leiomyomas in lymphangioleiomyomatosis. Fertil Steril 2011; 96:711.
  151. Guo M, Yu JJ, Perl AK, et al. Single-Cell Transcriptomic Analysis Identifies a Unique Pulmonary Lymphangioleiomyomatosis Cell. Am J Respir Crit Care Med 2020; 202:1373.
  152. Hammes SR, Krymskaya VP. Targeted approaches toward understanding and treating pulmonary lymphangioleiomyomatosis (LAM). Horm Cancer 2013; 4:70.
  153. Howe SR, Gottardis MM, Everitt JI, Walker C. Estrogen stimulation and tamoxifen inhibition of leiomyoma cell growth in vitro and in vivo. Endocrinology 1995; 136:4996.
  154. Zhang H, Bajraszewski N, Wu E, et al. PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. J Clin Invest 2007; 117:730.
  155. Lee PS, Tsang SW, Moses MA, et al. Rapamycin-insensitive up-regulation of MMP2 and other genes in tuberous sclerosis complex 2-deficient lymphangioleiomyomatosis-like cells. Am J Respir Cell Mol Biol 2010; 42:227.
  156. Atochina-Vasserman EN, Abramova E, James ML, et al. Pharmacological targeting of VEGFR signaling with axitinib inhibits Tsc2-null lesion growth in the mouse model of lymphangioleiomyomatosis. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1447.
  157. Lesma E, Eloisa C, Isaia E, et al. Development of a lymphangioleiomyomatosis model by endonasal administration of human TSC2-/- smooth muscle cells in mice. Am J Pathol 2012; 181:947.
  158. Du Y, Guo M, Wu Y, et al. Lymphangioleiomyomatosis (LAM) Cell Atlas. Thorax 2023; 78:85.
Topic 4335 Version 49.0

References

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