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

Erythropoietic protoporphyria and X-linked protoporphyria

Erythropoietic protoporphyria and X-linked protoporphyria
Literature review current through: Jan 2024.
This topic last updated: Sep 20, 2023.

INTRODUCTION — Erythropoietic protoporphyria (EPP) is an inherited cutaneous porphyria characterized by painful, nonblistering photosensitivity usually first noted in early childhood and occurring acutely after sunlight exposure but leaving little residual skin damage. The cutaneous phenotype can result from altered activity of one of two enzymes in the heme biosynthetic pathway, either a deficiency of ferrochelatase (FECH), which causes EPP; or a gain-of-function mutation of the erythroid-specific form of delta-aminolevulinic acid synthase (ALAS2), which causes X-linked protoporphyria (XLP). In one family, protoporphyria was caused by a pathogenic variant in CLPX, which encodes a mitochondrial chaperone that regulates ALAS2 [1]. An acquired, adult-onset form of EPP has also been described, in which a clone of cells with a pathogenic variant in the FECH gene expands in the setting of a myeloproliferative or myelodysplastic syndrome. The term "protoporphyria" includes both EPP and XLP.

The pathogenesis, clinical features, and treatment of EPP and XLP will be discussed here.

Other cutaneous porphyrias, as well as a general overview, are presented separately:

Overview – (See "Porphyrias: An overview".)

PCT and HEP diagnosis – (See "Porphyria cutanea tarda and hepatoerythropoietic porphyria: Pathogenesis, clinical manifestations, and diagnosis".)

PCT and HEP management – (See "Porphyria cutanea tarda and hepatoerythropoietic porphyria: Management and prognosis".)

CEP – (See "Congenital erythropoietic porphyria".)

HCP – (See "Hereditary coproporphyria".)

VP – (See "Variegate porphyria".)

PATHOGENESIS — EPP and XLP result from the accumulations of protoporphyrin, initially in the bone marrow, and then in erythrocytes and plasma, leading to nonblistering cutaneous photosensitivity.

Genetics — The characteristic protoporphyria phenotype can be due either to loss-of-function mutations in the gene for ferrochelatase (FECH), causing EPP, or, less commonly, to gain-of-function mutations in the gene for the erythroid form of delta-aminolevulinic acid synthase (ALAS2), causing XLP (figure 1) [2,3]. Protoporphyria was reported in one family to be caused by a pathogenic variant in CLPX suggests that additional very rare genetic mechanisms are possible [1].

FECH catalyzes insertion of iron into protoporphyrin to form heme and the insertion of zinc into most of the remaining small amount of protoporphyrin. (See "Porphyrias: An overview", section on 'Enzymes and intermediates'.)

In most protoporphyria families, the disease is EPP (OMIM #177000) and is due to pathogenic variants in the FECH gene, which encodes the final enzyme in the heme biosynthetic pathway, ferrochelatase (figure 1). Inheritance is autosomal recessive because biallelic loss-of-function variants are required to cause the disease.

Less commonly, the clinical phenotype can occur with gain-of-function mutations in ALAS2, which encodes the initial enzyme of the heme biosynthetic pathway in developing red blood cells. This form of protoporphyria is termed X-linked protoporphyria (XLP) or X-linked erythropoietic protoporphyria (XLEPP; OMIM #300752) because ALAS2 is located on the X chromosome. Inheritance is X-linked.

A case report has described EPP in a family with a pathogenic variant in CLPX, which encodes a mitochondrial chaperone that regulates the abundance of ALAS2 [1]. A dominantly acting mutation in CLPX that interfered with its ATPase activity caused ALAS2 to be stabilized and its activity increased, with overproduction of delta-aminolevulinic acid (ALA) and accumulation of protoporphyrin.

Rarely, adults can develop late onset EPP in the setting of a myeloproliferative neoplasm or myelodysplastic syndrome, in which a clone of erythroid precursor cells carrying a pathogenic variant in FECH expands and is responsible for a significant portion of hematopoiesis [4-6].

EPP due to FECH variant — EPP results from reduction of FECH activity to less than approximately 30 percent of normal, due to loss-of-function variants affecting both FECH alleles (located on chromosome 18). Most patients have a rare and severe loss-of-function FECH variant (>100 described in different families) in trans to a common hypomorphic (or "low expression") c.315 FECH variant on the other allele. The hypomorphic variant itself has no phenotype, even if homozygous.

The many severe loss-of-function FECH variants that have been described include missense, nonsense, and splice site mutations and deletions that lead to absent or substantially reduced enzyme activity. FECH deficiency impairs the formation of zinc protoporphyrin as well as heme synthesis in bone marrow red blood cell precursors (erythroblasts and reticulocytes); as a result, protoporphyrin accumulates mostly in its metal-free form in EPP.

The most common molecular basis for EPP is compound heterozygosity for a severe FECH variant inherited from one parent and the hypomorphic variant, IVS3-48T/C, inherited from the other parent [7]. The IVS3-48T/C variant results in increased use of an aberrant splice site and production of an mRNA that is more prone to degradation, leading to a decreased amount of FECH enzyme. In a French study, this allele occurred in approximately 10 percent of White individuals [8]; it is more common in people from Japan and China, and very rare in people from Africa [7-10]. This IVS3-48T/C variant is commonly described as pathogenic because it contributes importantly to reducing FECH activity in EPP, an autosomal recessive disease. Because it is common in individuals without EPP, it is often detected by whole genome sequencing or on genetic screening panels for porphyrias when not present in trans to a severe FECH variant.

Rarely, EPP is due to severe variants affecting both FECH alleles, in the absence of the hypomorphic c.315-48T>C FECH variant; but at least one of the affected alleles must be able to produce a small amount of functioning FECH enzyme [11-15]. EPP may be more severe and risk of liver disease higher in such families.  

In the past, inheritance of EPP was often described as autosomal dominant with variable penetrance. This erroneous designation occurred because heterozygous, severe FECH variants had been identified in EPP patients, but the common hypomorphic c.315-48T>C FECH variant had not yet been identified.

XLP due to ALAS2 gain-of-function mutations — XLP was recognized after some families with an EPP phenotype but lacking pathogenic variants in FECH were noted to have an X-linked inheritance pattern. Affected individuals also had a higher proportion of zinc protoporphyrin in their erythrocytes relative to patients with EPP due to FECH variants. These observations led to the identification of a pathogenic variant in the only heme biosynthetic gene located on the X chromosome, the erythroid form of delta-aminolevulinic acid synthase (ALAS2) [16]. These families were classified as having XLP, also called X-linked EPP (XLEPP).

ALAS is the initial enzyme in the heme biosynthetic pathway (figure 1), and ALAS2 variants responsible for XLP cause C-terminal deletions that lead to enzymatic gain of function. ALA and downstream heme pathway intermediates are overproduced. The resulting excess protoporphyrin in bone marrow erythroblasts and reticulocytes greatly exceeds the amount needed for heme synthesis, and because FECH activity is not deficient, more of the excess protoporphyrin is liganded with zinc than in EPP. (See 'Erythrocyte protoporphyrin' below.)

ALAS2 is transcribed only in bone marrow erythroblasts. In contrast to the gain-of-function mutations in XLP, loss-of-function mutations of ALAS2 have been described in cases of sex-linked sideroblastic anemia. A ubiquitous (housekeeping) form of ALAS (ALAS1) is produced in all tissues including the liver. Naturally occurring ALAS1 pathogenic variants have not been described. (See "Causes and pathophysiology of the sideroblastic anemias", section on 'X-linked sideroblastic anemia (ALAS2 mutation)'.)

EPP due to CLPX mutation — In 2017, a family was reported with autosomal dominant protoporphyria associated with a point mutation in the CLPX gene, identified by whole exome sequencing after no pathogenic variants were detected in FECH or ALAS2 [1]. ClpX is a mitochondrial AAA+ ATPase, and together with ClpP, it forms the AAA+ protease ClpXP.

Under normal conditions, ALAS2 is activated by ClpX via its unfoldase activity. A negative feedback loop is constituted by ClpP-mediated degradation of ALAS2 in response to high heme levels. The CLPX variant in this family inactivated the ATPase activity of the enzyme, thereby making active ALAS2 unavailable to ClpP for degradation, in turn leading to an increase of ALAS2 and accumulation of ALA and protoporphyrin [17].

Protoporphyrin accumulation — The FECH enzyme exists as a homodimer in the inner mitochondrial membrane and is essential for the final step of heme synthesis, which is the insertion of iron into the protoporphyrin IX ring (figure 1) [18-20]. FECH also catalyzes the insertion of zinc into most of the small amount of protoporphyrin IX that normally remains after completion of heme synthesis.

Insufficient FECH activity results in excessive accumulation of protoporphyrin that lacks iron or other metals, particularly zinc (metal-free protoporphyrin). The excess plasma and erythrocyte protoporphyrin in EPP is primarily metal-free protoporphyrin produced by bone marrow reticulocytes. Production of protoporphyrin by the bone marrow may increase further in EPP under conditions in which erythropoiesis is stimulated, leading to further increases in plasma and erythrocyte protoporphyrin levels. The liver had been proposed as an additional source of excess protoporphyrin, but this is now considered unlikely [21].

In XLP, there is increased generation of the protoporphyrin substrate for FECH; the amount of protoporphyrin exceeds the capacity of normal FECH activity. The functioning FECH enzyme can utilize zinc to form zinc protoporphyrin. Thus, in XLP, zinc protoporphyrin makes up a greater proportion of excess protoporphyrin than seen with EPP. However, metal-free protoporphyrin predominates in most XLP cases, suggesting that the capacity of normal FECH activity to insert zinc into protoporphyrin is exceeded.

Total, metal-free, and zinc protoporphyrin are most conveniently measured in circulating erythrocytes in EPP and XLP. Plasma protoporphyrin is a separate pool that reflects both bone marrow production and liver uptake of protoporphyrin. (See 'Diagnostic evaluation' below.)

Accumulation of metal-free protoporphyrin is a distinctive feature of EPP (>85 percent of total erythrocyte protoporphyrin) and is accounted for by FECH deficiency, and less than approximately 15 percent of the total protoporphyrin is in the form of zinc protoporphyrin.

Accumulation of metal-free protoporphyrin is also characteristic of XLP, which is due to ALAS2 gain-of-function mutations. However, a greater proportion of zinc protoporphyrin (approximately 15 to 50 percent of total erythrocyte protoporphyrin) is characteristic of XLP.

Protoporphyrin that accumulates in other erythrocyte disorders, such as iron deficiency, lead poisoning, anemia of chronic disease, and hemolytic disorders, is predominantly zinc protoporphyrin, since FECH activity is not appreciably decreased relative to the amount of available substrate (protoporphyrin) in these conditions.

Metal-free protoporphyrin in erythrocytes is bound to hemoglobin, and light irradiation may promote its release from hemoglobin (and from erythrocytes), perhaps thereby increasing plasma porphyrin levels and photosensitivity [22]. Metal-free protoporphyrin diffuses from circulating erythrocytes into plasma more readily than does zinc protoporphyrin, most of which remains within erythrocytes for their full life span [23]. It is also likely that bone marrow reticulocytes contribute protoporphyrin directly to the plasma pool.

As a result of diffusion of metal-free porphyrin from circulating erythrocytes as they age, the total protoporphyrin content declines with increasing red blood cell age in EPP, and "fluorocytes" seen on peripheral blood smears by fluorescence microscopy are younger erythrocytes that contain large amounts of metal-free protoporphyrin [2,23-25].

The excess protoporphyrin in plasma in EPP and XLP is bound to albumin and is taken up by the liver for secretion into bile, which is the only mechanism for removing this water-insoluble dicarboxyl porphyrin from the body (table 1) [22,24,26]. After entering the small intestine, some protoporphyrin may be absorbed and undergo enterohepatic circulation.

Several transporters are involved in heme biosynthesis and protoporphyrin disposition, and some are potential therapeutic targets. As examples:

Glycine is needed in large amounts for heme synthesis and is transported into mitochondria by glycine transporter 1 (GlyT1). Bitopertin, a selective GlyT1 inhibitor, reduced erythrocyte protoporphyrin levels and improved liver fibrosis in a mouse model of EPP and is under development for treatment of human EPP and XLP [27].

ABCG2 (ATP-binding cassette transporter subfamily G member 2), which transports protoporphyrin out of mitochondria and cells, is also a potential therapeutic target because ABCG2 deficiency reduces transport of intracellular protoporphyrin into plasma and bile and protects against hepatotoxicity and phototoxicity in mice with EPP [28].

Iron in EPP and XLP — Patients with EPP or XLP often have borderline iron deficiency, and it has been suggested that the excess protoporphyrin in EPP may downregulate iron absorption or lead to a redistribution of iron stores [29-31]. Although iron absorption was considered to be impaired [29,30], elemental iron absorption has been found to be normal and hepcidin levels appropriately low in these patients. For example, a study that assessed iron absorption in eight individuals with EPP compared with nine controls did not find evidence for impaired iron absorption or increased hepcidin levels in the individuals with EPP [32].

Preclinical studies suggest that iron status may modulate porphyrin accumulation in EPP and XLP. As an example, iron is important for formation of the non-catalytic iron-sulfur cluster in FECH and may thereby enhance post-translational stability of the FECH protein [33]. Consequently, iron deficiency might further impair FECH stability. Furthermore, because iron is a substrate for FECH, its deficiency in EPP might increase protoporphyrin accumulation, as in other iron deficiency states. Conversely, however, iron can upregulate ALAS2 and might increase protoporphyrin production [34]. Therefore, it is unclear whether iron deficiency is harmful or beneficial in patients with EPP or XLP. (See 'Iron' below.)

Effects on skin — Protoporphyrin is hydrophobic and deposited in lipid layers such as cell membranes; therefore, its tissue and subcellular distribution may differ from the hydrophilic porphyrins that accumulate and cause blistering manifestations in other cutaneous porphyrias [35]. Endothelial cells in the dermis are considered the primary cellular targets underlying phototoxicity in EPP [36,37].  

Porphyrins are photoactive and absorb light and enter an excited energy state, also known as a triplet form [38]. They can then transfer energy to dissolved oxygen to form the superoxide ion (O2-), which in turn can form hydroxyl ions (OH-). These highly oxidizing species of oxygen can interact with many biological molecules, such as proteins, lipids, and DNA, and can form adducts with carbon-carbon double bonds [35,39,40]. Porphyrins absorb wavelengths of light especially in the 400 to 420 nm range (ie, the Soret peak for porphyrins), which is in the visible light range and close to the range of long wavelength ultraviolet light (ie, UVA; range 315 to 400 nm). Patients with EPP and other cutaneous porphyrias are sensitive to sunlight and to some extent fluorescent and even incandescent indoor lights. Window glass transmits this light (but not short wave ultraviolet light [ie, UVB]) and therefore is not protective. Porphyrin photoreactivity causes tissue damage through lipid peroxidation, oxidation of nucleic acids and polypeptides, complement activation, and mast cell degranulation [39,41-44].

Liver damage — As noted below, protoporphyric hepatopathy is a rare but potentially severe complication of EPP, affecting fewer than 5 percent of patients. (See 'Liver disease' below.)

It is not clear what predisposes some patients and not others to develop hepatopathy. In some cases, protoporphyric hepatopathy was precipitated by another cause of liver disease, such as excess alcohol consumption or viral hepatitis, which might initially impair protoporphyrin excretion [45,46]. Iron deficiency might also contribute by impairing the conversion of protoporphyrin to heme in the bone marrow, resulting in increasing amounts of protoporphyrin circulating to the liver. However, this possibility has not been well studied. Studies in a mouse model of EPP have shown that a deficiency of ABCG2, which transports protoporphyrin from erythrocytes into plasma and from hepatocytes into bile, protects against development of liver damage and suggests that protoporphyrin is especially damaging to cholangiocytes [47].

When it does occur, hepatopathy results from accumulation of protoporphyrin in the liver in amounts that are damaging to hepatocytes and cholangiocytes [48]. This results in reduced biliary excretion of protoporphyrin and a progressive further buildup of protoporphyrin in the liver, plasma, and erythrocytes. Protoporphyrin has been shown to be cholestatic in an animal model [49]. Hepatopathy develops in association with a severe FECH mutation and also in patients with ALAS2 mutations, but no clear relationship to specific mutations has been found [50,51]. In a mouse model with inherited FECH deficiency, the development of hepatopathy is influenced by genetic background, but important modifier genes were not identified [52].

As liver damage and impaired biliary excretion progress, protoporphyrin levels in plasma, erythrocytes, and liver increase further, causing more severe photosensitivity and further liver damage [53]. Erythrocyte survival is reduced by splenic enlargement, which may stimulate erythropoiesis and further increases in marrow protoporphyrin overproduction [11,54]. A vicious cycle results from worsening hepatopathy and increased protoporphyrin production and retention.

EPIDEMIOLOGY — EPP was first comprehensively described in 1961 [55]. Since this initial description, EPP has been reported worldwide [56,57]. It is now recognized as the most common porphyria in children and the third most common in adults, after porphyria cutanea tarda (PCT) and acute intermittent porphyria (AIP) [58].

The incidence is similar in males and females.

Prevalence estimates for the general population based on surveys have ranged from 1:75,000 in the Netherlands to 1:200,000 in Wales [59,60]. A UK Biobank study of FECH variants suggests that EPP is more underrecognized than previously thought (prevalence, 1 in 17,000) [61].

EPP is very rare in Africa and is more common in East Asian countries than in European and North American countries, related to differing prevalence of the common hypomorphic FECH allele in individuals living in these regions. (See 'EPP due to FECH variant' above.)

XLP was initially described as a variant form of EPP without pathogenic variants in FECH and was characterized genetically in 2008; XLP comprises up to 10 percent of those with the protoporphyria phenotype [16,62].

CLINICAL FEATURES

Overview of clinical features — EPP and XLP typically present in early childhood with painful photosensitivity, although the cause of the photosensitivity these patients experience is often overlooked until later in life. In a review of over 100 cases in the United States, the average age of presentation was under four years [58]. Presentation is similar in males and females.

Children may be misdiagnosed as having an allergic reaction or primary angioedema. (See 'Differential diagnosis' below.)

In a large series from the United Kingdom (UK), the median ages at onset and diagnosis were 1 and 12 years, respectively [63]. In the review of over 100 cases in the United States, the average delay between presenting symptoms and ultimate diagnosis of EPP was 13 years [58]. Nearly 40 percent had seen at least five clinicians before the diagnosis was made; 22 percent had seen more than 10 clinicians.

The predominant clinical manifestation in EPP and XLP is painful, nonblistering cutaneous photosensitivity that differs distinctly from the chronic, blistering skin manifestations of the other cutaneous porphyrias. (See 'Cutaneous symptoms' below.)

Hepatobiliary complications include protoporphyrin-containing gallstones and, in less than 5 percent of cases, severe liver failure. (See 'Hepatobiliary manifestations' below.)

Many patients with EPP will have normal results on routine laboratory testing, with the common exception of mild hypochromic microcytic anemia, accompanied by low serum ferritin and low transferrin saturation. This iron deficiency phenotype is poorly understood. (See 'Anemia' below.)

The defining laboratory manifestation of EPP is a marked elevation of total erythrocyte protoporphyrin that is mostly (85 to 100 percent) metal-free; the defining finding in XLP is marked elevation of total erythrocyte protoporphyrin that is approximately 50 to 85 percent metal-free protoporphyrin. (See 'Diagnostic evaluation' below.)

Cutaneous symptoms — In most cases, photosensitivity is first experienced in infancy or early childhood [55,56,63,64]. Patients report onset of symptoms with exposure to sunlight, often within minutes.

Some patients can tolerate longer exposure periods, which is not well explained. Many patients note a "priming" effect, whereby sunlight sensitivity is increased by greater than usual exposure the previous day [65].

Prodromal symptoms are those that occur initially with sunlight exposure and are reversible if exposure ceases; these may include itching, tingling, stinging, heat, or mild burning [66-68].

Patients may describe themselves as "shadow jumpers" because they habitually avoid direct sunlight and try to respond quickly to these early cutaneous symptoms. With more prolonged exposure they develop severe burning pain accompanied or followed by redness, swelling, mild blistering, and systemic symptoms that may persist for several days (table 2). In one case series, the median time to onset of symptoms and signs (swelling, redness) following sun exposure was 20 minutes, and the median time to resolution of well-developed symptoms was three days [63]. In addition to sunlight, including sunlight passing through window glass or a car windshield, symptoms can also be elicited by non-sun exposures such as fluorescent lights and operating room lights. Time to prodrome during sunlight exposure has become a useful symptom endpoint in clinical trials of new therapies for EPP and XLP [67].  

When these symptoms resolve, there is little or no residual scarring. Patients learn to avoid sunlight and only rarely present to clinicians with findings such as edema, petechiae, telangiectasia, and scarring (picture 1 and picture 2). Therefore, physical findings on examination are usually absent.

There may be subtle findings such as [69-71]:

Cobblestone-like thickening or lichenification of the skin on the backs of the hands, especially over the knuckles

A waxy or leathery texture, petechiae, ecchymoses and minor scarring on the face

Vertical grooving of the lips; and loss of lunulae of fingernails

These changes result from repeated exposure to light and generally are seen in patients who experience more sun exposure (often children). Bullae, vesicles, and crusts, which are frequent in blistering cutaneous porphyrias, of which porphyria cutanea tarda (PCT) is the most common, are uncommon in EPP [48].

Variation in the severity of symptoms among different individuals relates in part to levels of erythrocyte protoporphyrin (see 'Erythrocyte protoporphyrin' below) and to inherent differences in pigmentation (Fitzpatrick skin type). Symptoms change little with age. Unexplained decreases in porphyrin levels and symptoms may occur during pregnancy [63,72,73]. Variations in skin manifestations over time are primarily related to the degree of sun exposure (eg, milder symptoms in winter) or to "priming" (more pronounced symptoms after the second of two sunlight exposures on consecutive days) [63,65].

Impaired quality of life — Patients learn to successfully avoid sunlight or fluorescent light and the painful photosensitivity that results from such exposure. However, these compensatory behaviors greatly impair daily activities and quality of life. Parents and caregivers of children with EPP and XLP also learn how to protect them from sunlight exposure. This requires considerable adjustments in lifestyle and may limit educational and employment opportunities. Diagnosis and recognition of the cause of symptoms is often delayed, which compounds quality of life issues, particularly in children.  

Hepatobiliary manifestations

Gallstones — The risk of gallstones is increased in patients with EPP and XLP; presentation is similar to patients without protoporphyria [55,74]. Gallstones were recognized in 8 percent of patients in one series [63]. Protoporphyria should be considered as a possible cause of gallstones in children [48]. Development of stones containing protoporphyrin is related to the overproduction of this water-insoluble porphyrin and its excretion exclusively in bile [75].

Liver disease — A cholestatic form of liver disease, referred to as protoporphyric hepatopathy, is the most serious complication of EPP and XLP. This complication is rare, occurring in fewer than 5 percent of patients [59,63]. Higher prevalence in some series may reflect referral bias or possibly closer monitoring and earlier detection. In contrast, most patients with uncomplicated EPP have normal liver function tests, and, although data are limited, little or no elevation in liver protoporphyrin concentration. Hepatopathy may be more likely in individuals with higher protoporphyrin levels; however, prospective data to support this impression are seldom available.

Protoporphyric hepatopathy can present initially as an asymptomatic chronic liver disease with mild abnormalities in serum transaminases (aminotransferases) and may progress to cirrhosis and hepatic failure. It may also present acutely at an advanced stage with severe right upper quadrant pain, jaundice, nausea, vomiting, and markedly abnormal liver chemistries [74,76].

The abdominal pain may be mistakenly attributed to gallstones, which may be present but asymptomatic. Such patients who present acutely are very likely to already have underlying chronic liver disease or cirrhosis with splenomegaly and other evidence of portal hypertension [54,77]. Severe hepatopathy may lead to the initial diagnosis of EPP in a patient with a long history of previously unexplained photosensitivity. As noted above, hepatopathy often leads to worsening photosensitivity, as greater amounts of porphyrins accumulate in plasma and erythrocytes. (See 'Liver damage' above.)

A diagnosis of protoporphyric hepatopathy is established by liver biopsy to demonstrate characteristic histological findings and exclude other more common liver diseases. If measured, hepatic protoporphyrin content is markedly elevated.  

The significance of persistent aminotransferase elevations in patients with protoporphyria is uncertain and should prompt diagnostic evaluation for other causes of liver disease. Liver enzyme elevations in the protoporphyrias have been reported in 25 to 27 percent of cases in large series [78]. However, most of these patients were not fully confirmed or evaluated for other primary causes of liver disease. A more detailed study of a large cohort in the Netherlands found that only 6.2 percent of patients had elevated liver enzymes, 29 percent had liver steatosis, and 9.6 percent had significant fibrosis by vibration controlled transient elastography [79]. Moreover, liver enzyme elevations in EPP patients were only slightly more common than expected relative to individuals without protoporphyria, and steatosis and increased liver stiffness were not more common than expected. These results suggest that early liver dysfunction due to protoporphyria, or early stages of hepatopathy likely to progress to end stage liver disease, is not common. An unexplained finding was mild splenomegaly affecting 22 percent, often accompanied by mild thrombocytopenia; these may be early findings of chronic liver disease that deserve further study [79].  

Peripheral neuropathy — Peripheral neuropathy resembling that seen in the acute porphyrias may develop in late stages of protoporphyric hepatopathy and may progress to respiratory failure [80,81]. To our knowledge, EPP with peripheral neuropathy has been reported only once in the absence of liver failure, but the diagnosis of EPP in that case was not convincing [82]. (See "Acute intermittent porphyria: Pathogenesis, clinical features, and diagnosis", section on 'Peripheral neuropathy'.)

Vitamin D deficiency and osteoporosis — Patients with EPP or XLP are predisposed to vitamin D insufficiency as a result of sun avoidance, which may lead to osteoporosis [83,84]. A Dutch study showed a high prevalence of vitamin D deficiency in individuals with EPP (46 percent); vitamin D deficiency was more common in males and correlated with severity of EPP [83]. Vitamin D supplementation is advised. (See 'Routine monitoring and interventions' below.)

Anemia — Some patients with EPP or XLP have a mild hypochromic microcytic anemia. This is often accompanied by low or borderline low serum ferritin levels and decreased transferrin saturation [85]. These features suggest the presence of iron deficiency without blood loss, and there is often an unexplained poor response to oral iron administration. (See 'Routine monitoring and interventions' below.)

Some individuals with EPP or XLP have ring sideroblasts on the iron-stained bone marrow aspirate smear, indicative of sideroblastic anemia. (See "Sideroblastic anemias: Diagnosis and management", section on 'Autosomal recessive forms'.)

Pregnancy — For unclear reasons, pregnancy may be associated with decreases in protoporphyrin levels and improvement in photosensitivity in patients with EPP or XLP [63,72,73]. This was demonstrated in a series of 32 pregnancies in 19 Swedish women with EPP; photosensitivity during pregnancy was reduced, unchanged, or increased in 53, 44, and 3 percent, respectively [86]. The postpartum period was associated with reduced photosensitivity in one-third and no change in two-thirds. All newborns were healthy, with one later diagnosis of EPP.

DIAGNOSTIC EVALUATION

When to suspect — Delays in diagnosis are especially concerning in EPP and XLP; these delays may be greater than with any other type of porphyria [63]. (See 'Overview of clinical features' above.)

The index of suspicion for EPP and XLP is often low because there are many causes of photosensitivity (table 2). Moreover, in contrast to other cutaneous porphyrias, skin findings are usually minimal and transient. Delay may also result from measuring urine porphyrins (which are not elevated) rather than total erythrocyte protoporphyrin. Plasma porphyrins are usually but not always elevated in EPP and XLP and are much lower than in erythrocytes. Plasma porphyrin levels become markedly elevated in protoporphyric hepatopathy.

The diagnosis of EPP or XLP should be considered in any patient with photosensitivity that is primarily acute and nonblistering. The diagnosis is readily established or excluded by measurement of total erythrocyte protoporphyrin followed by fractionation, if elevated, to determine the proportions of metal-free and zinc protoporphyrin. (See 'Erythrocyte protoporphyrin' below.)

Erythrocyte protoporphyrin — As with other porphyrias, our approach to diagnosis is to perform a sensitive screening test first, in this case total erythrocyte protoporphyrin, followed by fractionation (into metal-free and zinc protoporphyrin) using the same sample if the total is elevated (algorithm 1). Elevation of total erythrocyte protoporphyrin level with a predominance of metal-free protoporphyrin confirms the diagnosis of EPP or XLP biochemically. We then measure plasma porphyrins and perform genetic testing in all patients. (See 'Molecular/genetic testing' below.)

Urinary and fecal porphyrin testing is not required in the evaluation for EPP or XLP. If done, total porphyrins in feces may be normal or modestly increased, and consist mostly of protoporphyrin (table 1). Urinary porphyrins, delta-aminolevulinic acid (ALA), and porphobilinogen (PBG) are normal, with the exception of patients with protoporphyric hepatopathy, in whom urinary porphyrins, especially coproporphyrin, may be increased, as occurs in liver disease from any cause.

Normal range and fractionation — The normal range for total erythrocyte protoporphyrin is up to approximately 80 mcg/dL. The most reliable assays extract and measure all erythrocyte porphyrins fluorometrically and express the total as protoporphyrin. Erythrocyte porphyrins in health and disease are almost entirely protoporphyrin, with the exception of patients with congenital erythropoietic porphyria (see "Congenital erythropoietic porphyria"). The upper limit of normal varies with age and among laboratories, but this is not problematic for diagnosis of EPP or XLP because values in these patients are markedly elevated, in the range of 300 to 8000 mcg/dL (table 3).

An increase in total erythrocyte protoporphyrin is nonspecific, and therefore it is essential to fractionate the total and report the proportions of zinc protoporphyrin and metal-free protoporphyrin. An increase in erythrocyte zinc protoporphyrin is seen in many conditions, including iron deficiency, lead poisoning, anemia of chronic disease, and hemolytic disorders [87,88]. However, in EPP and most cases of XLP, the increase in protoporphyrin is predominantly in metal-free protoporphyrin [89]. Ultimately, genetic testing should be relied upon for differentiating EPP and XLP. (See 'Molecular/genetic testing' below.)

EPP – In EPP due to pathogenic variants in FECH, the excess protoporphyrin in erythrocytes is almost always >85 percent metal-free protoporphyrin and <15 percent zinc protoporphyrin.

XLP – In XLP due to ALAS2 gain-of-function mutations, a predominance of metal-free protoporphyrin is typically seen as well, but the proportion of zinc protoporphyrin is usually 15 to 50 percent. This greater abundance of zinc protoporphyrin can almost always differentiate XLP from EPP, but confirmation by genetic testing is advised.

Selecting a testing laboratory — Confusion among laboratories about terminology and methods for measuring erythrocyte porphyrins can complicate testing for EPP and XLP [89,90]. This occurs because some major laboratories that do not focus on diagnosis of protoporphyrias use a particular fluorescence instrument (hematofluorometer) for measuring erythrocyte protoporphyrin that was originally developed for screening for lead poisoning and is tuned to measure only zinc protoporphyrin; however, these laboratories may incorrectly report the results as “erythrocyte protoporphyrin” (implying that total erythrocyte protoporphyrin was measured) or “free erythrocyte protoporphyrin” (implying metal-free protoporphyrin) when in fact only zinc protoporphyrin was measured. Therefore, one must select a laboratory that measures total erythrocyte protoporphyrin and can then fractionate metal-free and zinc protoporphyrin.

Certified laboratories that can perform this fractionation include:

United StatesThe University of Texas Medical Branch (UTMB) Porphyria Center and Mayo Clinic Laboratories [91-93].

Europe – Appropriate laboratories at specialist centers in Europe can be accessed through the website of the International Porphyria Network (IPNET) (https://porphyrianet.org/en/content/worldwide-network).

Plasma porphyrins — The total plasma porphyrin concentration is elevated in most patients with EPP and XLP, but less so than in other cutaneous porphyrias, and it may be normal in milder cases. In addition, plasma porphyrins in EPP and XLP are especially sensitive to light and may degrade rapidly during sample processing [94]. Therefore, plasma porphyrin measurement should not be used alone as a screening test for protoporphyria.

Fluorescence scanning of plasma is performed in addition to measurement of plasma porphyrins. In EPP or XLP, the fluorescence of plasma (diluted at neutral pH) shows a peak at a wavelength of approximately 634 nm. However, this may be absent in mild cases. This fluorescence peak near 634 nm differs from other porphyrias (eg, peak near 626 nm in variegate porphyria [VP], peak near 620 nm in porphyria cutanea tarda [PCT] and congenital erythropoietic porphyria [CEP]) (table 3) [59].

In individuals with EPP and XLP, plasma porphyrin levels correlate roughly with the levels of total erythrocyte protoporphyrin. The latter is believed to remain fairly constant throughout life. Limited longitudinal data suggest that variations in total erythrocyte protoporphyrin of approximately 25 percent over time may not be of concern; plasma protoporphyrin levels are much more variable [95]. This is likely to be because the plasma pool of protoporphyrin turns over more rapidly. Plasma porphyrin is also likely to be the source of protoporphyrin uptake by the liver. Therefore, a higher plasma level may indicate a greater risk for hepatic complications. (See 'Hepatobiliary manifestations' above.)

Diagnosis — The diagnosis of protoporphyria (EPP and XLP) is made by demonstrating both of the following (table 4 and algorithm 1):

Increased total erythrocyte protoporphyrin (usually 300 to 8000 mcg/dL; normal <80 mcg/dL)

Increased percentage of erythrocyte metal-free protoporphyrin rather than zinc protoporphyrin

In EPP, metal-free protoporphyrin generally represents >85 percent of total porphyrins. In XLP, metal-free protoporphyrin generally represents 50 to 85 percent of total porphyrins.

Measurement of erythrocyte total protoporphyrin rather than molecular testing is advised for initial screening of patients with nonblistering photosensitivity, in part because finding the common hypomorphic c.315-48T>C FECH variant is an incidental benign finding in approximately 10 percent of individuals of European background and is even more common in individuals from East Asia.

Distinction between EPP and XLP using molecular (genetic) testing is not required for diagnosis or therapy, but it is confirmatory and should be done on all patients to characterize the disease and to inform genetic counseling and testing of family members. (See 'Molecular/genetic testing' below.)

Molecular/genetic testing — We perform genetic testing in all patients with EPP and XLP to confirm the diagnosis and enable genetic counseling and testing of relatives. (See 'Diagnosis' above and 'Genetic counseling and testing of relatives' below.)

In most families, EPP results from a rare, severe pathogenic variant in FECH and a hypomorphic c.315-48T>C FECH variant on the other allele (in trans) that is common in the general population but itself has no phenotype. Thus, the risk for EPP in future generations depends on transmission of the severe variant as well as on carriage of the common hypomorphic c.315-48T>C variant by present and future partners. Very rarely, EPP results from inheritance of a severe FECH variant from each parent; at least one of these variants must allow some FECH enzyme to be produced for heme synthesis.

Genetic testing of FECH is widely available. Genetic counseling is appropriate especially for individuals of childbearing potential. (See 'Genetic counseling and testing of relatives' below.)

Tissue biopsy — Biopsy is rarely required in the evaluation of EPP or XLP, with the exception of liver biopsy to confirm a diagnosis of protoporphyric hepatopathy and exclude other causes of liver disease. The following findings may be seen if biopsy is performed:

Skin – In the skin, the findings may differ depending on the stage of photosensitivity. An acute inflammatory reaction may be seen microscopically with extravasation of red cells, deposition of PAS-positive material in the perivascular spaces, and proliferation of the epidermal basal membrane [42,64,96-99]. Immunohistochemistry shows deposition of immunoglobulins and complement [100]. These changes are not specific for EPP and, except for absence of subepidermal blisters, may not be distinguishable from changes seen in the other cutaneous porphyrias.

Liver – Early stages of protoporphyric hepatopathy in terms of increased protoporphyrin content, damage to hepatocytes and cholangiocytes, and fibrosis have been little studied [101]. With advanced disease, the liver appears black on gross examination due to marked deposition of protoporphyrin and bilirubin [53]. Histologically, there is usually micronodular cirrhosis with cholestatic features and marked deposition of protoporphyrin seen as dark brown pigment that forms inclusions that are birefringent on polarizing microscopy with a "Maltese cross" appearance [102-104]. By electron microscopy, protoporphyrin appears as crystalline deposits mainly in hepatocytes but also in Kupffer cells and bile canaliculi, accompanied by ultrastructural damage to the endoplasmic reticulum, mitochondria, and cell membrane [101,102,105-107].

Bone marrow – In the bone marrow, fluorescence may be seen as a result of protoporphyrin accumulation in erythroid precursors. This fluorescence is maximal in reticulocytes [8,9,82].

DIFFERENTIAL DIAGNOSIS — The differential diagnosis of EPP and XLP is extensive (table 2). Diagnosis is often delayed, and most such individuals are regarded as having sensitivity to sunlight of unknown cause until elevation of metal-free protoporphyrin is documented.

Polymorphous light eruption – Polymorphous light eruption (PMLE; sometimes called "sun poisoning" or "sun allergy") is a common photodermatosis that typically occurs during the first three decades of life. Like EPP or XLP, the symptoms occur on sun-exposed areas, and family members may have similar symptoms. Unlike EPP or XLP, individuals with PMLE generally have discrete lesions such as pruritic papules, papulovesicles, or plaques, and the lesions develop later than EPP or XLP (hours to days rather than minutes); individuals with PMLE do not have elevations of erythrocyte total and metal-free protoporphyrin as measured by a reliable laboratory. (See 'Diagnostic evaluation' above and "Polymorphous light eruption".)

Solar urticaria – Solar urticaria is a condition in which exposure to sunlight causes urticaria. Like EPP and XLP, symptoms often develop within minutes. Unlike EPP and XLP, the symptoms of solar urticaria are often pruritic rather than painful. Unlike EPP and XLP, individuals with solar urticaria do not have elevations of erythrocyte protoporphyrin. (See "Physical (inducible) forms of urticaria", section on 'Solar urticaria'.)

Drug-induced photosensitivity – Drug-induced photosensitivity, also called phototoxicity, results when a sensitizing agent has been ingested or applied to the skin. Like EPP or XLP, patients may have painful erythema soon after sun exposure, often within minutes. Unlike EPP or XLP, phototoxic reactions are associated with use of the photosensitizing agent, and erythrocyte protoporphyrin is not elevated. (See "Photosensitivity disorders (photodermatoses): Clinical manifestations, diagnosis, and treatment", section on 'Phototoxicity' and "Drug eruptions", section on 'Photosensitivity reactions'.)

Sunburn – Sunburn is a transient inflammatory skin response to ultraviolet radiation from sunlight or artificial sources. Sunburn can occur in individuals without an underlying dermatologic condition, with sensitivity dependent on the degree of skin pigmentation. Like EPP or XLP, sunburn is a transient, painful, erythematous reaction that often does not cause blistering or scarring, and members of a family may have similar sensitivities. Unlike EPP or XLP, most individuals, even those who sunburn easily, do not develop symptoms upon normal daily sunlight exposure. Unlike EPP or XLP, individuals with sunburn do not have elevations of erythrocyte protoporphyrin [68]. (See "Sunburn".)

Additional discussions of the general approach to a patient with photosensitivity are presented separately. (See "Overview of cutaneous photosensitivity: Photobiology, patient evaluation, and photoprotection" and "Photosensitivity disorders (photodermatoses): Clinical manifestations, diagnosis, and treatment".)

MANAGEMENT — No effective way of lowering circulating porphyrin levels in individuals with EPP or XLP has been established. Patients learn to avoid sunlight and fluorescent light as much as possible, but compensatory behaviors greatly impair daily activities and quality of life and limit educational and employment opportunities and recreational activities. (See 'Impaired quality of life' above.)

Other photoprotective interventions are limited, as discussed below. (See 'Photoprotection' below.)

Photoprotection

Sun and UV light avoidance — Protection from sunlight is a cornerstone of EPP management. The use of protective clothing and hats is beneficial for most patients when outdoors. Patients learn from experience to alter behavior, such as seeking shaded areas when outdoors, and refer to themselves as "shadow jumpers."

Broad spectrum sunscreen preparations that are protective against long wave ultraviolet (UVA) light may be of some benefit, but these do not protect against wavelengths of light in the visible range. Those that are protective against UVB are not beneficial. As in other cutaneous porphyrias, window glass is not protective when indoors (or in a vehicle), and patients may equip house and car windows with protective tinted glass. Patients may require clinician letters for state permission to use adequately tinted glass in their automobiles. Many patients also find it necessary to avoid indoor lighting. (See "Selection of sunscreen and sun-protective measures".)

Other sources of UVA and visible light such as operating room lights may also cause phototoxic reactions [108]. Protection from light during surgical procedures using light filters has been best studied in the setting of liver transplantation, which is a prolonged operation in patients with hepatopathy and particularly high porphyrin levels [109] (see 'Liver transplantation' below). Complications in patients without hepatopathy undergoing more routine surgical procedures are uncommon.

We do not recommend topical application of dihydroxyacetone and lawsone (naphthoquinone), because of concerns about potential adverse cosmetic effects and carcinogenic potential with long-term use [110-112].

Although avoiding light is highly beneficial in preventing symptoms of EPP and XLP, it greatly limits options for employment and prevents engagement in many pleasurable outdoor activities with family and friends, thereby substantially reducing some important life opportunities. Therefore, it is important to consider other therapies that may increase light tolerance and allow patients to have more normal lifestyles.

Afamelanotide — Afamelanotide is a synthetic analogue of alpha-melanocyte stimulating hormone (alpha-MSH), a naturally occurring hormone that increases skin pigmentation by increasing melanin production, and reduces free radical formation and cytokine production [113,114]. Afamelanotide increases sunlight tolerance in EPP and XLP, by these and possibly other mechanisms [115].

For adults with EPP or XLP, we suggest afamelanotide, unless their lifestyle and/or employment needs do not require greater sunlight tolerance. Pediatric administration has not been evaluated and pediatric dose implants are not available. Afamelanotide became available in parts of Europe in 2009 and was approved by the US Food and Drug Administration in October of 2019 [116,117]. Access is limited in the United States due to the high cost and restriction by the manufacturer of physicians authorized to administer the drug.

Afamelanotide is administered as a controlled-release 16 mg implant injected subcutaneously every other month, and can be used especially during the summer months.

Evidence for the efficacy of afamelanotide includes the following:

Two multicenter, randomized trials in individuals 18 years or older with EPP or XLP found that afamelanotide substantially reduced photosensitivity and improved sunlight tolerance [114]. In a European trial (74 patients followed for nine months), the duration of pain-free time in direct sunlight was longer with afamelanotide versus placebo (median 6.0 versus 0.8 hours) and the number of phototoxic reactions was lower in the afamelanotide group (77 versus 146). In the United States (US) trial (94 patients followed for six months) the duration of pain-free time in direct sunlight was longer in the afamelanotide group (median 69.4 versus 40.8 hours); the greater number of hours in the United States trial reflects a longer portion of the day in which sunlight tolerance was assessed. In both trials, quality of life improved with afamelanotide therapy. Some participants described this treatment as 'life changing' in terms of activities they could engage in during treatment, which previously were not possible. Real-world studies have also observed improved quality of life with afamelanotide [118].

In a retrospective study of 115 individuals with EPP who received afamelanotide at one of two porphyria centers for up to six years, 74 percent found the therapy to be effective [119]. Of the remaining individuals, 23 percent had to discontinue therapy for pregnancy or financial reasons, and 3 percent found the therapy to be ineffective. The mean quality of life scores for the group rose from 32 to 74 percent during the first six months of therapy and remained high (69 to 91 percent) for the duration of the study. Melanin density also increased but was challenging to interpret due to reduced therapy during the winter and increased sun exposure with effective therapy in some patients. The most frequent reasons for drug discontinuation were financial restrictions and pregnancy. This study population represented two-thirds of all individuals with EPP at these centers.

Time to prodrome was not assessed in the initial afamelanotide clinical trials. However, a later retrospective interview study of patients from the United States and the Netherlands found that the time to prodrome was substantially improved during afamelanotide treatment [67].

Toxicities of afamelanotide appear to be minimal; the most common adverse events are nausea and headache [114,119]. Temporary skin darkening is expected [120]. Pigmented moles may become more prominent during treatment, so skin examinations by a dermatologist are recommended every six months [121].

However, clinical experience is limited. European product information for afamelanotide states it is not to be used during pregnancy or in individuals with liver or kidney impairment, as there are no long term data regarding safety [120]. Preclinical data indicate that hepatic uptake of the drug is minimal, so adverse effects on the liver seem unlikely. Retrospective observations in patients with protoporphyria suggest that afamelanotide may lower rather than increase levels of erythrocyte protoporphyrin and serum transaminases [122]. It is possible that increased sunlight tolerance and exposure leads to an increase in photodegradation of protoporphyrin during circulation through the skin, thereby lowering protoporphyrin levels. However, this remains to be convincingly demonstrated. (See 'Protoporphyrin accumulation' above.)

Dersimelagon — Dersimelagon is an investigational orally administered small molecule that selectively activates the melanocortin-1 receptor (MC1R). In a 2023 trial that randomly assigned 93 individuals with EPP and nine with XLP to receive dersimelagon (100 mg daily or 300 mg daily) or placebo for 16 weeks, those assigned to dersimelagon had a significant improvement in sunlight tolerance [66]. The time to prodromal skin symptoms improved by 20 minutes in the placebo group, versus 74 minutes in the dersimelagon 100 mg group and 83 minutes in the dersimelagon 300 mg group. The incidence of phototoxic pain also decreased (from 7.5 events in 16 weeks with placebo, to 3.3 events in 16 weeks with dersimelagon 100 mg, to 3.5 events in 16 weeks with dersimelagon 300 mg). Adverse effects were mostly mild and transient (nausea, headache, freckles, skin hyperpigmentation). If it becomes available, dersimelagon may be preferred by some patients because it is administered orally rather than as a subcutaneous implant.

Beta-carotene — Some patients note increased tolerance to sunlight with beta-carotene use, especially in the summer [123]. The proposed protective mechanism is quenching of oxygen free radicals by beta-carotene. Although we suggest that most patients with EPP or XLP try oral beta carotene, most do not find this therapy to be very beneficial, and it causes skin discoloration. Some patients believe that they benefit and save on cost by taking beta-carotene mostly in spring and summer, which permits greater sunlight exposure and acquisition of a suntan that provides further sunlight protection.

Evidence supporting the efficacy of beta-carotene includes several small studies and case series [124-132]. In the largest series with 133 patients, 22 reported no effect and 111 reported moderate to substantial improvement in sunlight tolerance. A single randomized crossover trial enrolled 14 children; this showed that compared with placebo, individuals receiving beta-carotene had a small increase in time spent outdoors (equivalent to approximately 13 minutes daily) but no major difference in symptoms reported in diaries [133].

We advise using pharmaceutical grade medication (brand name, Lumitene) developed specifically for treating EPP. It was originally available by prescription but is now available over the counter. For adults, we use oral doses of 30 to 300 mg (1 to 10 capsules) daily, and titrate to maintain serum carotene levels in the range of 600 to 800 mcg/dL or a tolerable degree of yellowish skin pigmentation, which is most prominent on the palms of the hands. Carotene levels can be assessed three to four weeks after a dose change.

Doses in children are in the range of 30 to 150 mg/day (1 to 5 capsules). Capsules may be opened and the contents mixed into orange or tomato juice to aid administration. Most individuals notice a protective effect within one to three months. If the patient is not experiencing reduced symptoms with an adequate carotene level after three months, the therapy may be discontinued. High-dose beta-carotene supplementation may increase the risk of lung cancer among current smokers [134].

Plasma and erythrocyte protoporphyrin levels are not affected by beta carotene administration. (See 'Protoporphyrin accumulation' above.)

Mild, dose-related skin discoloration, especially on the palms of the hands, is expected because an effective dose is associated with carotenemia.

Other agents — A number of other methods of photoprotection have been employed, with variable success.

Narrow-band UVB phototherapy that provides exposure to UVB in the range of 311 to 313 nm, which stimulates melanin formation but does not activate porphyrins, has been described as beneficial [135]. (See "UVB phototherapy (broadband and narrowband)".)

Oral cysteine, which is available as a nutritional supplement, may improve light tolerance in EPP by quenching excited oxygen species at doses of 500 mg twice daily [136,137]. However, experience is limited, and we have not used this approach. N-acetylcysteine, a closely related product was shown to be ineffective in two double blind crossover placebo-controlled trials [138,139].

We do not use vitamin C to treat EPP, as it did not show benefit in a double blind randomized controlled trial [140], and therefore is not recommended.

At present, we also do not recommend oral cimetidine, which has been used in acute hepatic porphyrias, but in the absence of evidence for efficacy and safety. Cimetidine inhibits hepatic cytochrome P450 enzymes, and thereby ameliorates hepatic porphyria in an animal model (rodents treated with chemicals that are activated by these enzymes). Cimetidine inhibition of bone marrow ALAS2 has been suggested but not demonstrated. In a 2016 report, decreased photosensitivity in response to oral cimetidine was described in three children with EPP [141]. However, this was not a formal pilot study or trial, and decreases in protoporphyrin levels were not documented. A double-blind placebo-controlled trial is underway in the United States to determine if cimetidine is effective and safe in patients with protoporphyria.

Iron — Iron supplementation in patients with EPP and XLP is controversial. As noted above, the reason for mild iron deficiency commonly seen in these patients is not understood. In a 2015 study, patients were able to absorb iron well from the gastrointestinal tract and did not have inappropriately high levels of hepcidin [32]. (See 'Iron in EPP and XLP' above.)

Some authorities feel that because iron can upregulate ALAS2, iron deficiency may be protective and iron supplementation is contraindicated [34]. Some patients have had unexplained worsening of photosensitivity with iron therapy, although increases in porphyrin levels were not documented [142-145]. In such cases it is possible that correction of iron deficiency may have transiently increased erythropoiesis, with a resulting short term increase in protoporphyrin production. However, in one patient with EPP, intravenous iron sucrose lowered porphyrin levels and improved sunlight tolerance [146]. It is generally agreed that patients with XLP benefit from iron replacement [147]. Larger systematic studies of iron therapy are needed.

Until further data become available, we use iron supplementation (eg, ferrous sulfate 325 mg one to three times daily) in patients with clearly low levels of serum ferritin (ie, less than approximately 15 to 20 ng/mL). The goal is to treat anemia, improve fatigue and subtle cognitive impairment that may be caused by iron deficiency [148]; porphyrin accumulation may be reduced. Future systematic studies may alter this practice.

Pain control/antiinflammatory agents — Initial prodromal symptoms resolve rapidly if the patient escapes from sun to shade [67]. Acute cutaneous symptoms from more prolonged exposure generally resolve without treatment within hours or days, but before resolution patients may have severe pain and systemic symptoms and may be bedridden. Treatment with nonsteroidal antiinflammatory drugs (NSAIDs) and even opioids may provide little relief. Some patients report that cold compresses may be helpful.

Routine monitoring and interventions — Individuals with EPP and XLP should have at least annual monitoring of the following [90,121]:

Liver function tests

Complete blood count (CBC)

Ferritin

Vitamin D

RBC and plasma porphyrins

The rationale for testing and appropriate interventions based on the results include:

Liver function – This monitoring may help in the early detection of protoporphyric hepatopathy. Additionally, monitoring of hepatic function may help identify other possible causes of liver disease that may precipitate protoporphyric hepatopathy. However, this practice has not been studied prospectively, and it is not yet possible to identify those patients at risk of developing protoporphyria-related liver disease. Elevations in liver transaminases can be seen in as many as 27 percent of EPP patients [121]. Evaluation for other causes of liver disease is often incomplete so the true prevalence of early protoporphyric hepatopathy in EPP and XLP is unknown.

If liver function tests are abnormal, the evaluation should exclude other causes of liver disease, and imaging is recommended to look for evidence of chronic liver disease, such as splenomegaly possibly related to portal hypertension. Any reversible causes of liver dysfunction that might precipitate hepatopathy, including excess alcohol use, should be identified and treated. There is no association of EPP with hepatitis C virus (HCV), other viral infections or other causes of liver disease. However, elevated liver function tests should prompt the same workup as in patients without EPP, and include testing for hepatitis B and C infection and for more uncommon causes such as Wilson disease, alpha-1-antitrypsin deficiency and hemochromatosis. Steatotic liver diseases due to alcohol or metabolic factors such as obesity and diabetes are likely causes of abnormal liver function tests in patients with EPP or XLP, and may prompt confirmation by liver biopsy. Liver biopsy is generally required to establish a diagnosis of protoporphyric hepatopathy but its early recognition has been little studied [149].

Liver protection – Vaccination to prevent hepatitis A and B is important in EPP or XLP, as viral hepatitis might precipitate protoporphyric hepatopathy. (See "Immunizations for adults with chronic liver disease".)

Additionally, patients should avoid excess exposure to alcohol and hepatotoxic drugs. As a precaution, we also avoid use of drugs or hormone preparations that impair hepatic excretory function, and severe calorie restriction, which is harmful in acute hepatic porphyrias.

Iron status – CBC and ferritin should be monitored, since EPP and XLP are often accompanied by a poorly explained "iron deficiency" phenotype. Patients with XLP generally benefit from correction of iron deficiency, but iron treatment in EPP is more controversial, because there is evidence that iron may upregulate ALAS2 and increase production and circulating levels of protoporphyrin [90,121]. However, severe iron deficiency, especially with significant anemia, should be treated if identified. (See "Treatment of iron deficiency anemia in adults".)

Porphyrin levels – Erythrocyte and plasma porphyrin levels should be measured at least annually for early detection of increases in levels possibly due to liver disease or change in iron status. Of note, on average, porphyrin levels are higher in XLP than in EPP, and individuals with higher total erythrocyte porphyrin levels are likely to be at greater risk for hepatopathy. Patients should be encouraged to enroll in longitudinal studies being conducted by the Porphyrias Consortium in the United States and the International Porphyria Network (IPNET) in Europe in order to gain knowledge and develop better guidelines for long-term management.

Vitamin D – Patients with EPP are very likely to develop vitamin D deficiency because of sunlight avoidance [90,121]. Measurements of serum 25-hydroxy vitamin D are performed annually.

We ensure that patients have a daily intake of 800 international units of vitamin D and 1000 mg of calcium; typically, this requires vitamin D supplementation because sunlight avoidance is extreme. Individuals with vitamin D deficiency despite this intake may require higher doses of vitamin D.

Treatment of gallstones and protoporphyric hepatopathy

Gallstones — Management of gallstones is the same as in patients without EPP. Cholecystectomy for symptomatic gallstones may be needed during childhood. (See "Approach to the management of gallstones".)

In asymptomatic patients with normal liver chemistries, a screening ultrasound to detect gallstones is not recommended because cholecystectomy is not recommended for asymptomatic cholelithiasis [121].

Management of protoporphyric hepatopathy — In a 2023 document, experts from the Porphyrias Consortium (part of the National Institutes of Health [NIH] Rare Diseases Clinical Research Network [RDCRN]) examined important questions in the diagnosis and management of liver complications of protoporphyria and recognized that evidence was often insufficient for strong recommendations [78]. Little has been published on this very rare condition other than case reports and small series of patients describing efforts to bridge patients with life-threatening disease to liver transplantation. Medical interventions have sometimes led to remission, but well designed trials to examine efficacy and safety of particular treatments are lacking. Moreover, porphyrin levels are rarely measured at most centers, so it seldom has been possible to ascertain whether or not short term biochemical improvements are occurring with treatment in these acutely affected patients.

A combination of treatments is often used for patients with severe, decompensated hepatopathy. The aim is to reduce the amount of protoporphyrin entering plasma and delivered to the liver and also ameliorate its toxic effects, allowing the liver a chance to recover sufficiently to bridge patients to liver transplantation [150].

A regimen for patients with severe and rapidly progressing hepatopathy may include all of the following [151]. If at all possible, plasma and erythrocyte porphyrin levels, in addition to liver chemistries, should be measured frequently to assess ongoing effectiveness of treatment.

Hemin – Intravenous hemin has been found to reduce plasma porphyrin levels presumably by reducing protoporphyrin production by the marrow, but the mechanism is not established [152-155]. Hemin is administered at a dose of 4 mg/kg body weight daily for at least four days, as discussed in more detail separately. (See "Acute intermittent porphyria: Management", section on 'Indications and mechanism of action'.)

Plasmapheresis – This can reduce markedly elevated plasma porphyrin levels up to 40 percent and thereby reduce liver uptake of protoporphyrin from the plasma compartment [156,157]. Erythrocyte exchange has also been used because the amount of protoporphyrin in erythrocytes greatly exceeds that in plasma [158]. However, this may be less successful because the aim in the treatment of hepatopathy is to lower the level of plasma protoporphyrin, which turns over rapidly and may largely come directly from the marrow as well as from circulating erythrocytes. Evidence in mice with EPP suggests that retention of excess protoporphyrin within erythrocytes and hepatocytes may protect against development of hepatopathy [47].

Treatment of anemia – It is important to correct anemia by erythrocyte transfusions because, even if well tolerated, anemia can stimulate erythropoiesis and might increase overproduction of protoporphyrin by the bone marrow [153,159]. An increased hemoglobin level after erythrocyte transfusions (eg, >10 g/dL) can decrease erythropoietin levels and reduce erythropoiesis. Response may be less in patients with shortened red cell survival due to splenomegaly [160].

UDCAUrsodeoxycholic acid (UDCA) may increase biliary excretion of protoporphyrin. Chenodeoxycholic acid should be avoided because of potential hepatotoxicity [161,162]. UDCA is administered as 10 mg/kg daily, in two divided doses.

Cholestyramine – Given at a dose of 4 grams one to two times daily can be used to interrupt the enterohepatic circulation of protoporphyrin and thereby reduce plasma protoporphyrin levels [163,164].

Vitamin E – Given orally at a dose of 400 international units daily can be used to reduce oxidative damage to hepatocytes [165].

These treatments are administered in combination based on experience in patients with advanced, life-threatening decompensation, which is the most common presentation described in the literature. Some patients recover from an acute episode or are bridged to liver transplantation. However, high quality evidence for efficacy of these treatments alone or in combination are lacking, due to the rarity and the acute, life-threatening presentation of this hepatic complication. Frequent measurements to document short term improvement in porphyrin levels are also usually lacking.  

There is less published experience in treating patients with hepatopathy that is progressing slowly but causing few symptoms. Such patients may already have established cirrhosis. Oral agents such as UDCA, cholestyramine, and vitamin E may be most practical, with repeated measurements of liver chemistries and plasma and erythrocyte porphyrin levels to assess response.  

Liver transplantation — Any patient with EPP who develops cirrhosis or severe protoporphyric hepatopathy should be referred for evaluation for possible liver transplantation. (See "Liver transplantation in adults: Patient selection and pretransplantation evaluation".)

Liver transplantation for protoporphyric hepatopathy was first reported in 1980; subsequent experience has expanded to more than 40 cases [166,167]. Compilation of cases in the United States and European transplant registries has shown that even though liver disease commonly recurs, overall survival is comparable to that of patients transplanted for other liver diseases [152,168-170]. As an example, in the United States, survival of adults who underwent liver transplantation for EPP was 85 percent at one year and 69 percent at five years [169]. Biliary complications occurred in 45 percent of patients, which is higher than in other liver transplant recipients. This is attributed to high levels of protoporphyrin in bile and damaging effects of "toxic bile" on cholangiocytes. Therefore, construction of a Roux loop to more safely transport "toxic bile" has been recommended in preference to duct-to-duct anastomosis [152,168,169].

The perioperative course in these patients is often complicated by severe motor neuropathy, as was first described in a case report [81]. This may be prevented or lessened in severity with the use of intravenous hemin and plasmapheresis prior to liver transplantation, although controlled studies are lacking. Dosages of hemin have been similar to those used in acute porphyrias (eg, 4 mg/kg body weight daily). (See "Acute intermittent porphyria: Management", section on 'Indications and mechanism of action'.)

Because these patients have very high plasma and erythrocyte porphyrin levels at the time of surgery, there is a risk of severe phototoxicity to the skin and visceral surfaces from the surgical lighting. Protective filters can prevent this phototoxicity. Three types of filters have been studied: CLS-200-X and TA-81 from Madico Inc, and 61011 from Reflective SA. CLS-200-X has the least visual distortion, while TA-81 and 61011 offer more protection. These authors recommended 61011 filters as the best compromise between visual distortion for the surgeon and light protection [109].

Liver transplantation restores normal liver function, including hepatic excretion of protoporphyrin, but it does not correct the metabolic abnormality in the bone marrow, which continues to produce excessive amounts of protoporphyrin. Patients should be monitored closely to avoid anemia, pronounced iron deficiency, and other factors that may cause the bone marrow to produce greater amounts of protoporphyrin. Recurrent liver disease has been reported as early as eight months after transplantation. There is anecdotal evidence that chronic plasmapheresis and intravenous hemin can stabilize and improve recurrent protoporphyric hepatopathy after liver transplantation [171].

Patients with recurrent hepatopathy should be evaluated for hematopoietic cell transplantation to avoid loss of the grafted liver [152,172,173]. (See 'Hematopoietic stem cell transplantation' below.)

Hematopoietic stem cell transplantation — Indications for hematopoietic stem cell transplantation in EPP are complex, in part because predictors of developing hepatopathy are lacking [170,174]. In patients with protoporphyric hepatopathy, sequential liver transplantation and HSCT may be appropriate in order to prevent future damage to the allograft liver, if a suitable hematopoietic stem cell donor is available [172]. In patients who recover from hepatopathy and have minimal or no fibrosis, HSCT may be performed without liver transplantation, which if successful will essentially cure the EPP or XLP and prevent a recurrence of hepatopathy [83,170].

Transplantation was first performed in a patient with adult onset EPP in the setting of leukemia and resulted in resolution of the EPP phenotype [175]. Subsequently, additional reports have described individuals with protoporphyria who had developed severe cholestasis, with documented resolution of the disease in many, although some individuals did not experience hematopoietic stem cell engraftment [150,169,172,176].

It is not possible with available data to predict which patients with EPP are at greatest risk to develop cholestatic liver disease, for whom early transplantation would be appropriate [174]. Decisions regarding the choice of donor are at the discretion of the transplant center.

Genetic counseling and testing of relatives — A confirmed genetic diagnosis of EPP or XLP provides a rational basis for genetic counseling. The details of the counseling and genetic testing differ for these protoporphyrias:

EPP – Inheritance of EPP, which is due to biallelic pathogenic variants in the FECH gene, is autosomal recessive. (See 'EPP due to FECH variant' above.)

For families in which a child has EPP due to a severe FECH variant plus the common hypomorphic c.315-48T>C FECH variant, the likelihood of having another child who inherits the severe variant is approximately 50 percent, and of having another affected child (who inherits both the severe variant and the hypomorphic allele) is approximately 25 percent.

For patients with EPP due to a severe FECH mutation plus the hypomorphic c.315-48T>C FECH variant, the likelihood of their having an affected child will depend on whether the other parent carries the hypomorphic c.315-48T>C variant, which is more common in some populations than others (see 'EPP due to FECH variant' above). Testing the other parent can determine this likelihood. If the other parent's FECH genes are both normal, half the children will inherit the severe FECH mutation (which may be passed on to future generations); however, they will not have EPP because they also inherited one normal FECH allele.

XLP – Inheritance of XLP (due to an ALAS2 gain-of-function mutation) is sex linked. Males who inherit the mutation are affected. Less commonly, females who inherit the mutation are affected if skewed lyonization (random X chromosome inactivation) results in preferential inactivation of the unaffected X chromosome. (See 'XLP due to ALAS2 gain-of-function mutations' above.)

For families in which a child has XLP due to an ALAS2 gain-of-function mutation, the mother is an obligate carrier. The likelihood of these parents having another child who inherits the mutation is approximately 50 percent; of those who inherit the mutation, sons will have XLP and most daughters will be carriers. The father with a normal ALAS2 allele will not transmit the disease.

For males with XLP, the mutation will be transmitted only to daughters, so no sons will have XLP. All daughters will have one ALAS2 gain-of-function mutation (and may or may not be affected) and will transmit the mutation to half of their children, resulting in disease in male children who inherit the mutation and carrier status in most female children who inherit the mutation. Some females will be affected due to skewed lyonization.

Additional information about these inheritance patterns and genetic counseling issues for children is presented separately. (See "Genetic testing", section on 'Testing children' and "Inheritance patterns of monogenic disorders (Mendelian and non-Mendelian)", section on 'Mendelian inheritance patterns'.)

PROGNOSIS — EPP and XLP have been described in the past as diseases that cause non-life-threatening photosensitivity that can be managed by avoiding sunlight. Life expectancy generally is normal, unless porphyric hepatopathy develops. However, although these diseases may not shorten life expectancy, they have a greater impact on quality of life than do other cutaneous porphyrias and other diseases that cause photosensitivity because pain in EPP and XLP is more acute and intense and necessitates alterations in lifestyle and employment [63,177]. These adverse consequences are accentuated in children and adults who are undiagnosed and as yet have no explanation for their symptoms.

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: Porphyria" and "Society guideline links: Photosensitivity disorders (photodermatoses)".)

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 topic (see "Patient education: Erythropoietic protoporphyria and X-linked protoporphyria (The Basics)")

SUMMARY AND RECOMMENDATIONS

Genetics and pathogenesis – Erythropoietic protoporphyria (EPP) and X-linked protoporphyria (XLP) result from accumulations of protoporphyrin in bone marrow reticulocytes and in turn in the circulation, leading to nonblistering cutaneous photosensitivity. In EPP, the characteristic phenotype is due to loss-of-function mutations in the gene for ferrochelatase (FECH) (figure 1), or, in the less common condition XLP, to gain-of-function mutations in the gene for the erythroid form of delta-aminolevulinic acid synthase (ALAS2). A single family has been described with protoporphyria due to a variant in CLPX, which encodes a protein that inactivates ALAS2. (See 'Pathogenesis' above.)

Prevalence – EPP is the most common porphyria in children and the third most common in adults, after porphyria cutanea tarda and acute intermittent porphyria. The incidence is similar in males and females. EPP is very rare in Africa and is more common in East Asia than in White European or American populations. (See 'Epidemiology' above.)

Clinical features – The predominant clinical manifestation in protoporphyria is painful, nonblistering cutaneous photosensitivity, often within minutes of sun exposure. Vesiculation sometimes occurs after prolonged exposure, and subtle residual skin changes are seen with repeated exposure over months or years. Most patients avoid sunlight and have no physical findings. Hepatobiliary complications include protoporphyrin-containing gallstones and, in less than 5 percent of cases, severe liver failure. Patients are at risk for vitamin D deficiency due to sun avoidance, and often have mild iron deficiency anemia, which is unexplained. Symptoms may improve during pregnancy. (See 'Clinical features' above.)

Evaluation – EPP and XLP should be considered in any patient with acute and primarily nonblistering photosensitivity; delays in diagnosis are common. The primary screening test is total erythrocyte (red blood cell) protoporphyrin; if this is elevated, fractionation is used to determine the proportions of metal-free and zinc protoporphyrin (algorithm 1 and table 4). The diagnosis is made if total erythrocyte protoporphyrin is elevated with a predominance of erythrocyte metal-free protoporphyrin (usually >85 percent in EPP and 50 to 85 percent in XLP). Genetic testing is confirmatory and important for genetic counseling and is strongly advised, especially for differentiating EPP and XLP. (See 'Diagnostic evaluation' above.)

Differential diagnosis – The differential diagnosis of EPP and XLP includes other photosensitivity disorders such as polymorphous light eruption, solar urticaria, drug-induced phototoxicity, and sunburn (table 2). (See 'Differential diagnosis' above.)

Treatment

Protection from sunlight – Protection from sunlight is a cornerstone of EPP management. Patients learn to avoid sunlight exposure, but this limits occupational and recreational opportunities and impairs quality of life. The use of protective clothing, hats, and protective tinted automobile window glass is essential for most patients when outdoors or driving. Other strong sources of light including operating room lights should be used with caution.

Afamelanotide and beta carotene – For adults with EPP or XLP, we suggest afamelanotide to improve sunlight tolerance and quality of life (Grade 2B). This is administered as a subcutaneous implant every other month. Although less effective, we also suggest beta-carotene for patients of all ages with EPP or XLP (Grade 2C). These therapies do not directly alter porphyrin levels. (See 'Photoprotection' above.)

Dersimelagon – Dersimelagon is under investigation but is not clinically available. (See 'Dersimelagon' above.)

Monitoring – Individuals with EPP and XLP should have monitoring of liver function, complete blood count (CBC), ferritin, erythrocyte and plasma protoporphyrin levels, and vitamin D levels at least once per year. Vitamin D supplementation is usually required. If liver chemistries are persistently abnormal, investigation for other etiologies of liver disease should follow. Vaccination to prevent hepatitis A and B is important. (See 'Routine monitoring and interventions' above.)

Liver and hematopoietic stem cell transplantation – A combination of treatments is often used for patients with decompensated hepatopathy. Any patient with EPP and advanced protoporphyric hepatopathy should be referred for evaluation for possible liver transplantation. In some individuals with severe hepatopathy, sequential liver transplantation and hematopoietic stem cell transplantation may be appropriate. (See 'Treatment of gallstones and protoporphyric hepatopathy' above.)

Genetic testing and counseling – Genetic testing and counseling are appropriate for individuals with EPP or XLP and their first-degree relatives. (See 'Genetic counseling and testing of relatives' above.)

  1. Yien YY, Ducamp S, van der Vorm LN, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A 2017; 114:E8045.
  2. Bottomley SS, Tanaka M, Everett MA. Diminished erythroid ferrochelatase activity in protoporphyria. J Lab Clin Med 1975; 86:126.
  3. Bonkowsky HL, Bloomer JR, Ebert PS, Mahoney MJ. Heme synthetase deficiency in human protoporphyria. Demonstration of the defect in liver and cultured skin fibroblasts. J Clin Invest 1975; 56:1139.
  4. Goodwin RG, Kell WJ, Laidler P, et al. Photosensitivity and acute liver injury in myeloproliferative disorder secondary to late-onset protoporphyria caused by deletion of a ferrochelatase gene in hematopoietic cells. Blood 2006; 107:60.
  5. Sarkany RP, Ross G, Willis F. Acquired erythropoietic protoporphyria as a result of myelodysplasia causing loss of chromosome 18. Br J Dermatol 2006; 155:464.
  6. Fassi DE, Jønch, AE, et al. PB1998: AZACYTIDINE AND ALLOGENOUS STEM CELL TRANSPLANTATION IN LATE-ONSET ERYTHROPOIETIC PROTOPORPHYRIA. Hemasphere 2023; 7 (Suppl):e6610528.
  7. Gouya L, Puy H, Robreau AM, et al. The penetrance of dominant erythropoietic protoporphyria is modulated by expression of wildtype FECH. Nat Genet 2002; 30:27.
  8. Gouya L, Puy H, Lamoril J, et al. Inheritance in erythropoietic protoporphyria: a common wild-type ferrochelatase allelic variant with low expression accounts for clinical manifestation. Blood 1999; 93:2105.
  9. Gouya L, Deybach JC, Lamoril J, et al. Modulation of the phenotype in dominant erythropoietic protoporphyria by a low expression of the normal ferrochelatase allele. Am J Hum Genet 1996; 58:292.
  10. Gouya L, Martin-Schmitt C, Robreau AM, et al. Contribution of a common single-nucleotide polymorphism to the genetic predisposition for erythropoietic protoporphyria. Am J Hum Genet 2006; 78:2.
  11. Sarkany RP, Cox TM. Autosomal recessive erythropoietic protoporphyria: a syndrome of severe photosensitivity and liver failure. QJM 1995; 88:541.
  12. Risheg H, Chen FP, Bloomer JR. Genotypic determinants of phenotype in North American patients with erythropoietic protoporphyria. Mol Genet Metab 2003; 80:196.
  13. Wiman A, Floderus Y, Harper P. Novel mutations and phenotypic effect of the splice site modulator IVS3-48C in nine Swedish families with erythropoietic protoporphyria. J Hum Genet 2003; 48:70.
  14. Parker M, Corrigall AV, Hift RJ, Meissner PN. Molecular characterization of erythropoietic protoporphyria in South Africa. Br J Dermatol 2008; 159:182.
  15. Schneider-Yin X, Mamet R, Minder EI, Schoenfeld N. Biochemical and molecular diagnosis of erythropoietic protoporphyria in an Ashkenazi Jewish family. J Inherit Metab Dis 2008; 31 Suppl 2:S363.
  16. Whatley SD, Ducamp S, Gouya L, et al. C-terminal deletions in the ALAS2 gene lead to gain of function and cause X-linked dominant protoporphyria without anemia or iron overload. Am J Hum Genet 2008; 83:408.
  17. Whitman JC, Paw BH, Chung J. The role of ClpX in erythropoietic protoporphyria. Hematol Transfus Cell Ther 2018; 40:182.
  18. Nakahashi Y, Taketani S, Okuda M, et al. Molecular cloning and sequence analysis of cDNA encoding human ferrochelatase. Biochem Biophys Res Commun 1990; 173:748.
  19. Whitcombe DM, Carter NP, Albertson DG, et al. Assignment of the human ferrochelatase gene (FECH) and a locus for protoporphyria to chromosome 18q22. Genomics 1991; 11:1152.
  20. Taketani S, Inazawa J, Nakahashi Y, et al. Structure of the human ferrochelatase gene. Exon/intron gene organization and location of the gene to chromosome 18. Eur J Biochem 1992; 205:217.
  21. Scholnick P, Marver HS, Schmid R. Erythropoietic protoporphyria: evidence for multiple sites of excess protoporphyrin formation. J Clin Invest 1971; 50:203.
  22. Sandberg S, Talstad I, Høvding G, Bjelland N. Light-induced release of protoporphyrin, but not of zinc protoporphyrin, from erythrocytes in a patient with greatly elevated erythrocyte protoporphyrin. Blood 1983; 62:846.
  23. Piomelli S, Lamola AA, Poh-Fitzpatrick MF, et al. Erythropoietic protoporphyria and lead intoxication: the molecular basis for difference in cutaneous photosensitivity. I. Different rates of disappearance of protoporphyrin from the erythrocytes, both in vivo and in vitro. J Clin Invest 1975; 56:1519.
  24. Sassaroli M, da Costa R, Väänänen H, et al. Distribution of erythrocyte free porphyrin content in erythropoietic protoporphyria. J Lab Clin Med 1992; 120:614.
  25. Clark KG, Nicholson DC. Erythrocyte protoporphyrin and iron uptake in erythropoietic protoporphyria. Clin Sci 1971; 41:363.
  26. Sandberg S, Brun A. Light-induced protoporphyrin release from erythrocytes in erythropoietic protoporphyria. J Clin Invest 1982; 70:693.
  27. Wu M, Ducamp S, Xiang Y, et al. Bitopertin, a Selective Glycine Transporter 1 Inhibitor, Reduced Protoporphyrin IX (PPIX) Level and Improved Liver Fibrosis in a Mouse Model of Erythropoietic Protoporphyria (EPP). Blood 2022; 140 (Suppl 1):8192.
  28. Zhu J, Lei S, Lu J, et al. Metabolism-guided development of Ko143 analogs as ABCG2 inhibitors. Eur J Med Chem 2023; 259:115666.
  29. Holme SA, Worwood M, Anstey AV, et al. Erythropoiesis and iron metabolism in dominant erythropoietic protoporphyria. Blood 2007; 110:4108.
  30. Lyoumi S, Abitbol M, Andrieu V, et al. Increased plasma transferrin, altered body iron distribution, and microcytic hypochromic anemia in ferrochelatase-deficient mice. Blood 2007; 109:811.
  31. Delaby C, Lyoumi S, Ducamp S, et al. Excessive erythrocyte PPIX influences the hematologic status and iron metabolism in patients with dominant erythropoietic protoporphyria. Cell Mol Biol (Noisy-le-grand) 2009; 55:45.
  32. Bossi K, Lee J, Schmeltzer P, et al. Homeostasis of iron and hepcidin in erythropoietic protoporphyria. Eur J Clin Invest 2015; 45:1032.
  33. Crooks DR, Ghosh MC, Haller RG, et al. Posttranslational stability of the heme biosynthetic enzyme ferrochelatase is dependent on iron availability and intact iron-sulfur cluster assembly machinery. Blood 2010; 115:860.
  34. Barman-Aksözen J, Minder EI, Schubiger C, et al. In ferrochelatase-deficient protoporphyria patients, ALAS2 expression is enhanced and erythrocytic protoporphyrin concentration correlates with iron availability. Blood Cells Mol Dis 2015; 54:71.
  35. Sandberg S, Romslo I. Porphyrin-induced photodamage at the cellular and the subcellular level as related to the solubility of the porphyrin. Clin Chim Acta 1981; 109:193.
  36. Brun A, Sandberg S. Mechanisms of photosensitivity in porphyric patients with special emphasis on erythropoietic protoporphyria. J Photochem Photobiol B 1991; 10:285.
  37. Hussain Z, Qi Q, Zhu J, et al. Protoporphyrin IX-induced phototoxicity: Mechanisms and therapeutics. Pharmacol Ther 2023; 248:108487.
  38. Kochevar IE. Primary processes in photobiology and photosensitization. In: Photoimmunology: An Update, Krutman J, Elmats C (Eds), Blackwell, New York 1995. p.19.
  39. Goldstein BD, Harber LC. Erythropoietic protoporphyria: lipid peroxidation and red cell membrane damage associated with photohemolysis. J Clin Invest 1972; 51:892.
  40. De Goeij AF, Van Steveninck J. Photodynamic effects of protoporphyrin on cholesterol and unsaturated fatty acids in erythrocyte membranes in protoporphyria and in normal red blood cells. Clin Chim Acta 1976; 68:115.
  41. Poh-Fitzpatrick MB. Molecular and cellular mechanisms of porphyrin photosensitization. Photodermatol 1986; 3:148.
  42. Gigli I, Schothorst AA, Soter NA, Pathak MA. Erythropoietic protoporphyria. Photoactivation of the complement system. J Clin Invest 1980; 66:517.
  43. Lim HW, Gigli I. Role of complement in porphyrin-induced photosensitivity. J Invest Dermatol 1981; 76:4.
  44. Lim HW, Poh-Fitzpatrick MB, Gigli I. Activation of the complement system in patients with porphyrias after irradiation in vivo. J Clin Invest 1984; 74:1961.
  45. Poh-Fitzpatrick MB, Whitlock RT, Leftkowitch JH. Changes in protoporphyrin distribution dynamics during liver failure and recovery in a patient with protoporphyria and Epstein-Barr viral hepatitis. Am J Med 1986; 80:943.
  46. Bonkovsky HL, Schned AR. Fatal liver failure in protoporphyria. Synergism between ethanol excess and the genetic defect. Gastroenterology 1986; 90:191.
  47. Wang P, Sachar M, Lu J, et al. The essential role of the transporter ABCG2 in the pathophysiology of erythropoietic protoporphyria. Sci Adv 2019; 5:eaaw6127.
  48. Cox TM. Protoporphyria. In: Porphyrin Handbook, Part II, Kadish KM, Smith K, Guilard R (Eds), Academic Press, San Diego 2003. p.121.
  49. Avner DL, Lee RG, Berenson MM. Protoporphyrin-induced cholestasis in the isolated in situ perfused rat liver. J Clin Invest 1981; 67:385.
  50. Minder EI, Gouya L, Schneider-Yin X, Deybach JC. A genotype-phenotype correlation between null-allele mutations in the ferrochelatase gene and liver complication in patients with erythropoietic protoporphyria. Cell Mol Biol (Noisy-le-grand) 2002; 48:91.
  51. Bloomer J, Bruzzone C, Zhu L, et al. Molecular defects in ferrochelatase in patients with protoporphyria requiring liver transplantation. J Clin Invest 1998; 102:107.
  52. Lyoumi S, Abitbol M, Rainteau D, et al. Protoporphyrin retention in hepatocytes and Kupffer cells prevents sclerosing cholangitis in erythropoietic protoporphyria mouse model. Gastroenterology 2011; 141:1509.
  53. Bloomer JR. The liver in protoporphyria. Hepatology 1988; 8:402.
  54. Key NS, Rank JM, Freese D, et al. Hemolytic anemia in protoporphyria: possible precipitating role of liver failure and photic stress. Am J Hematol 1992; 39:202.
  55. MAGNUS IA, JARRETT A, PRANKERD TA, RIMINGTON C. Erythropoietic protoporphyria. A new porphyria syndrome with solar urticaria due to protoporphyrinaemia. Lancet 1961; 2:448.
  56. DeLeo VA, Poh-Fitzpatrick M, Mathews-Roth M, Harber LC. Erythropoietic protoporphyria. 10 years experience. Am J Med 1976; 60:8.
  57. Poh-Fitzpatrick MB. Erythropoietic porphyrias: current mechanistic, diagnostic, and therapeutic considerations. Semin Hematol 1977; 14:211.
  58. Lala SM, Naik H, Balwani M. Diagnostic Delay in Erythropoietic Protoporphyria. J Pediatr 2018; 202:320.
  59. Elder GH, Smith SG, Smyth SJ. Laboratory investigation of the porphyrias. Ann Clin Biochem 1990; 27 ( Pt 5):395.
  60. Went LN, Klasen EC. Genetic aspects of erythropoietic protoporphyria. Ann Hum Genet 1984; 48:105.
  61. Dickey AK, Quick C, Ducamp S, et al. Evidence in the UK Biobank for the underdiagnosis of erythropoietic protoporphyria. Genet Med 2021; 23:140.
  62. Balwani M, Doheny D, Bishop DF, et al. Loss-of-function ferrochelatase and gain-of-function erythroid-specific 5-aminolevulinate synthase mutations causing erythropoietic protoporphyria and x-linked protoporphyria in North American patients reveal novel mutations and a high prevalence of X-linked protoporphyria. Mol Med 2013; 19:26.
  63. Holme SA, Anstey AV, Finlay AY, et al. Erythropoietic protoporphyria in the U.K.: clinical features and effect on quality of life. Br J Dermatol 2006; 155:574.
  64. Peterka ES, Fusaro RM, Goltz RW. Erythropoietic protoporphyria. II. Histological and histochemical studies of cutaneous lesions. Arch Dermatol 1965; 92:357.
  65. Poh-Fitzpatrick MB. The "priming phenomenon" in the acute phototoxicity of erythropoietic protoporphyria. J Am Acad Dermatol 1989; 21:311.
  66. Balwani M, Bonkovsky HL, Levy C, et al. Dersimelagon in Erythropoietic Protoporphyrias. N Engl J Med 2023; 388:1376.
  67. Wensink D, Langendonk JG, Overbey JR, et al. Erythropoietic protoporphyria: time to prodrome, the warning signal to exit sun exposure without pain-a patient-reported outcome efficacy measure. Genet Med 2021; 23:1616.
  68. Raef HS, Rebeiz L, Leaf RK, et al. Light-Related Cutaneous Symptoms of Erythropoietic Protoporphyria and Associations With Light Sensitivity Measurements. JAMA Dermatol 2023; 159:204.
  69. Baart de la Faille H, Bijlmer-Iest JC, van Hattum J, et al. Erythropoietic protoporphyria: clinical aspects with emphasis on the skin. Curr Probl Dermatol 1991; 20:123.
  70. Schmidt H, Snitker G, Thomsen K, Lintrup J. Erythropoietic protoporphyria. A clinical study based on 29 cases in 14 families. Arch Dermatol 1974; 110:58.
  71. Bopp C, Bakos L, da Graça Busko M. Erythropoietic protoporphyria. Int J Biochem 1980; 12:909.
  72. Madu AE, Whittaker SJ. Erythropoietic protoporphyria in pregnancy. J Obstet Gynaecol 2006; 26:687.
  73. Poh-Fitzpatrick MB. Human protoporphyria: reduced cutaneous photosensitivity and lower erythrocyte porphyrin levels during pregnancy. J Am Acad Dermatol 1997; 36:40.
  74. Doss MO, Frank M. Hepatobiliary implications and complications in protoporphyria, a 20-year study. Clin Biochem 1989; 22:223.
  75. Cripps DJ, Scheuer PJ. Hepatobiliary changes in erythropoietic protoporphyria. Arch Pathol 1965; 80:500.
  76. Bloomer JR, Phillips MJ, Davidson DL, et al. Hepatic disease in erythropoietic protoporphyria. Am J Med 1975; 58:869.
  77. Bloomer JR, Rank JM, Payne WD, et al. Follow-up after liver transplantation for protoporphyric liver disease. Liver Transpl Surg 1996; 2:269.
  78. Levy C, Dickey AK, Wang B, et al. Evidence-based consensus guidelines for the diagnosis and management of protoporphyria-related liver dysfunction in erythropoietic protoporphyria and X-linked protoporphyria. Hepatology 2023.
  79. Wensink D, Coenen S, Wilson JHP, et al. Liver involvement in patients with erythropoietic protoporphyria. Dig Liver Dis 2022; 54:515.
  80. Herbert A, Corbin D, Williams A, et al. Erythropoietic protoporphyria: unusual skin and neurological problems after liver transplantation. Gastroenterology 1991; 100:1753.
  81. Rank JM, Carithers R, Bloomer J. Evidence for neurological dysfunction in end-stage protoporphyric liver disease. Hepatology 1993; 18:1404.
  82. Hengstman GJ, de Laat KF, Jacobs B, van Engelen BG. Sensorimotor axonal polyneuropathy without hepatic failure in erythropoietic protoporphyria. J Clin Neuromuscul Dis 2009; 11:72.
  83. Spelt JM, de Rooij FW, Wilson JH, Zandbergen AA. Vitamin D deficiency in patients with erythropoietic protoporphyria. J Inherit Metab Dis 2010; 33 Suppl 3:S1.
  84. Biewenga M, Matawlie RHS, Friesema ECH, et al. Osteoporosis in patients with erythropoietic protoporphyria. Br J Dermatol 2017; 177:1693.
  85. Wahlin S, Floderus Y, Stål P, Harper P. Erythropoietic protoporphyria in Sweden: demographic, clinical, biochemical and genetic characteristics. J Intern Med 2011; 269:278.
  86. Wahlin S, Marschall HU, Fischler B. Maternal and fetal outcome in Swedish women with erythropoietic protoporphyria. Br J Dermatol 2013; 168:1311.
  87. Hastka J, Lasserre JJ, Schwarzbeck A, et al. Zinc protoporphyrin in anemia of chronic disorders. Blood 1993; 81:1200.
  88. Anderson KE, Sassa S, Peterson CM, Kappas A. Increased erythrocyte uroporphyrinogen-l-synthetase, delta-aminolevulinic acid dehydratase and protoporphyrin in hemolytic anemias. Am J Med 1977; 63:359.
  89. Gou EW, Balwani M, Bissell DM, et al. Pitfalls in Erythrocyte Protoporphyrin Measurement for Diagnosis and Monitoring of Protoporphyrias. Clin Chem 2015; 61:1453.
  90. Leaf RK, Dickey AK. How I treat erythropoietic protoporphyria and X-linked protoporphyria. Blood 2023; 141:2921.
  91. http://www.mayomedicallaboratories.com/test-catalog/Overview/88886.
  92. http://www.mayomedicallaboratories.com/test-catalog/Overview/31893.
  93. https://www.utmb.edu/internalmedicine/divisions/gastroenterology/research/research-programs/porphyria-program (Accessed on January 26, 2022).
  94. Poh-Fitzpatrick MB, DeLeo VA. Rates of plasma porphyrin disappearance in fluorescent vs. red incandescent light exposure. J Invest Dermatol 1977; 69:510.
  95. Gou E, Weng C, Greene T, et al. Longitudinal Analysis of Erythrocyte and Plasma Protoporphyrin Levels in Patients with Protoporphyria. J Appl Lab Med 2018; 3:213.
  96. Epstein JH, Tuffanelli DL, Epstein WL. Cutaneous changes in the porphyrias. A microscopic study. Arch Dermatol 1973; 107:689.
  97. Ryan EA, Madill GT. Electron microscopy of the skin in erythropoietic protoporphyria. Br J Dermatol 1968; 80:561.
  98. Ryan EA. Histochemistry of the skin in erythropoietic protoporphyria. Br J Dermatol 1966; 78:501.
  99. Sasai Y. Erythropoietic protoporphyria. Histochemical study of hyaline material. Acta Derm Venereol 1973; 53:179.
  100. Lim HW. Pathophysiology of cutaneous lesions in porphyrias. Semin Hematol 1989; 26:114.
  101. MacDonald DM, Germain D, Perrot H. The histopathology and ultrastructure of liver disease in erythropoietic protoporphyria. Br J Dermatol 1981; 104:7.
  102. Klatskin G, Bloomer JR. Birefringence of hepatic pigment deposits in erythropoietic protoporphyria. Specificity of polarization microscopy in the identification of hepatic protoporphyrin deposits. Gastroenterology 1974; 67:294.
  103. Matilla A, Molland EA. A light and electron microscopic study of the liver in case of erythrohepatic protoporphyria and in griseofulvin-induced porphyria in mice. J Clin Pathol 1974; 27:698.
  104. Bloomer JR, Enriquez R. Evidence that hepatic crystalline deposits in a patient with protoporphyria are composed of protoporphyrin. Gastroenterology 1982; 82:569.
  105. Barnes HD, Hurworth E, Millar JH. Erythropoietic porphyrin hepatitis. J Clin Pathol 1968; 21:157.
  106. Cripps DJ, Scheuer PJ. Hepatobiliary changes in erythropoietic protoporphyria. Arch Pathol 1965; 80:500.
  107. Rademakers LH, Cleton MI, Kooijman C, et al. Ultrastructural aspects of the liver in erythrohepatic protoporphyria. Curr Probl Dermatol 1991; 20:154.
  108. Balwani M, Bloomer J, Desnick R. Erythropoietic Protoporphyria, Autosomal Recessive. In: GeneReviews, Pagon RA, Adam MP, Ardinger HH, et al (Eds), University of Washington, Seattle, Seattle 2012.
  109. Wahlin S, Srikanthan N, Hamre B, et al. Protection from phototoxic injury during surgery and endoscopy in erythropoietic protoporphyria. Liver Transpl 2008; 14:1340.
  110. Fusaro RM, Johnson JA. Protection against long ultraviolet and/or visible light with topical dihydroxyacetone. Implications for the mechanism of action of the sunscreen combination, dihydroxyacetone/naphthoquinone. Dermatologica 1975; 150:346.
  111. Fusaro RM, Rice EG. The maillard reaction for sunlight protection. Ann N Y Acad Sci 2005; 1043:174.
  112. Rice EG. Dihydroxyacetone naphthoquinone protection against photosensitivity. Dermatologica 1976; 153:38.
  113. Minder EI. Afamelanotide, an agonistic analog of α-melanocyte-stimulating hormone, in dermal phototoxicity of erythropoietic protoporphyria. Expert Opin Investig Drugs 2010; 19:1591.
  114. Langendonk JG, Balwani M, Anderson KE, et al. Afamelanotide for Erythropoietic Protoporphyria. N Engl J Med 2015; 373:48.
  115. Luger TA, Böhm M. An α-MSH analog in erythropoietic protoporphyria. J Invest Dermatol 2015; 135:929.
  116. Biba E. Protection: the sunscreen pill. Nature 2014; 515:S124.
  117. https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-increase-pain-free-light-exposure-patients-rare-disorder (Accessed on October 08, 2019).
  118. Ceresnie MS, Mohney L, Ko D, et al. Association of quality of life measures with afamelanotide treatment in patients with erythropoietic protoporphyria and x-linked protoporphyria: A retrospective cohort study. J Am Acad Dermatol 2023; 88:880.
  119. Biolcati G, Marchesini E, Sorge F, et al. Long-term observational study of afamelanotide in 115 patients with erythropoietic protoporphyria. Br J Dermatol 2015; 172:1601.
  120. http://ec.europa.eu/health/documents/community-register/2014/20141222130241/anx_130241_en.pdf (Accessed on August 04, 2015).
  121. Dickey AK, Naik H, Keel SB, et al. Evidence-based consensus guidelines for the diagnosis and management of erythropoietic protoporphyria and X-linked protoporphyria. J Am Acad Dermatol 2023; 89:1227.
  122. Minder AE, Schneider-Yin X, Zulewski H, et al. Afamelanotide Is Associated with Dose-Dependent Protective Effect from Liver Damage Related to Erythropoietic Protoporphyria. Life (Basel) 2023; 13.
  123. Köpcke W, Krutmann J. Protection from sunburn with beta-Carotene--a meta-analysis. Photochem Photobiol 2008; 84:284.
  124. Mathews-Roth MM, Pathak MA, Fitzpatrick TB, et al. Beta-carotene as a photoprotective agent in erythropoietic protoporphyria. N Engl J Med 1970; 282:1231.
  125. Baart de la Faille H, Suurmond D, Went LN, et al. -Carotene as a treatment for photohypersensitivity due to erythropoietic protoporphyria. Dermatologica 1972; 145:389.
  126. Mathews-Roth MM, Pathak UA, Fitzpatrick TB, et al. Beta-carotene as an oral photoprotective agent in erythropoietic protoporphyria. JAMA 1974; 228:1004.
  127. Beckert E, Metz J. [Erythropoietic protoporthyria. Clinical aspects and therapy]. Fortschr Med 1976; 94:1981.
  128. Goerz G, Ippen H. [Treatment of photodermatoses with carotinoids (author's transl)]. Dtsch Med Wochenschr 1977; 102:1051.
  129. Mathews-Roth MM, Pathak MA, Fitzpatrick TB, et al. Beta carotene therapy for erythropoietic protoporphyria and other photosensitivity diseases. Arch Dermatol 1977; 113:1229.
  130. Thomsen K, Schmidt H, Fischer A. Beta-carotene in erythropoietic protoporphyria: 5 years' experience. Dermatologica 1979; 159:82.
  131. Lehmann P, Scharffetter K, Kind P, Goerz G. [Erythropoietic protoporphyria: synopsis of 20 patients]. Hautarzt 1991; 42:570.
  132. Minder EI, Schneider-Yin X, Steurer J, Bachmann LM. A systematic review of treatment options for dermal photosensitivity in erythropoietic protoporphyria. Cell Mol Biol (Noisy-le-grand) 2009; 55:84.
  133. Corbett MF, Herxheimer A, Magnus IA, et al. The long term treatment with beta-carotene in erythropoietic protoporphyria: a controlled trial. Br J Dermatol 1977; 97:655.
  134. Tanvetyanon T, Bepler G. Beta-carotene in multivitamins and the possible risk of lung cancer among smokers versus former smokers: a meta-analysis and evaluation of national brands. Cancer 2008; 113:150.
  135. Collins P, Ferguson J. Narrow-band UVB (TL-01) phototherapy: an effective preventative treatment for the photodermatoses. Br J Dermatol 1995; 132:956.
  136. Mathews-Roth MM, Rosner B, Benfell K, Roberts JE. A double-blind study of cysteine photoprotection in erythropoietic protoporphyria. Photodermatol Photoimmunol Photomed 1994; 10:244.
  137. Mathews-Roth MM, Rosner B. Long-term treatment of erythropoietic protoporphyria with cysteine. Photodermatol Photoimmunol Photomed 2002; 18:307.
  138. Bijlmer-Iest JC, Baart de la Faille H, van Asbeck BS, et al. Protoporphyrin photosensitivity cannot be attenuated by oral N-acetylcysteine. Photodermatol Photoimmunol Photomed 1992-1993; 9:245.
  139. Norris PG, Baker CS, Roberts JE, Hawk JL. Treatment of erythropoietic protoporphyria with N-acetylcysteine. Arch Dermatol 1995; 131:354.
  140. Boffa MJ, Ead RD, Reed P, Weinkove C. A double-blind, placebo-controlled, crossover trial of oral vitamin C in erythropoietic protoporphyria. Photodermatol Photoimmunol Photomed 1996; 12:27.
  141. Tu JH, Sheu SL, Teng JM. Novel Treatment Using Cimetidine for Erythropoietic Protoporphyria in Children. JAMA Dermatol 2016; 152:1258.
  142. Gordeuk VR, Brittenham GM, Hawkins CW, et al. Iron therapy for hepatic dysfunction in erythropoietic protoporphyria. Ann Intern Med 1986; 105:27.
  143. Holme SA, Thomas CL, Whatley SD, et al. Symptomatic response of erythropoietic protoporphyria to iron supplementation. J Am Acad Dermatol 2007; 56:1070.
  144. Milligan A, Graham-Brown RA, Sarkany I, Baker H. Erythropoietic protoporphyria exacerbated by oral iron therapy. Br J Dermatol 1988; 119:63.
  145. McClements BM, Bingham A, Callender ME, Trimble ER. Erythropoietic protoporphyria and iron therapy. Br J Dermatol 1990; 122:423.
  146. Bentley DP, Meek EM. Clinical and biochemical improvement following low-dose intravenous iron therapy in a patient with erythropoietic protoporphyria. Br J Haematol 2013; 163:289.
  147. Landefeld C, Kentouche K, Gruhn B, et al. X-linked protoporphyria: Iron supplementation improves protoporphyrin overload, liver damage and anaemia. Br J Haematol 2016; 173:482.
  148. Prasad AN, Prasad C. Iron deficiency; non-hematological manifestations. Prog Food Nutr Sci 1991; 15:255.
  149. Anstey AV, Hift RJ. Liver disease in erythropoietic protoporphyria: insights and implications for management. Postgrad Med J 2007; 83:739.
  150. Wahlin S, Aschan J, Björnstedt M, et al. Curative bone marrow transplantation in erythropoietic protoporphyria after reversal of severe cholestasis. J Hepatol 2007; 46:174.
  151. Sood G, Anderson K. Porphyrias. In: Evidence-Based Hematology, Crowther M, Ginsberg J, Schunemann H, et al. (Eds), Wiley, Hoboken 2008. p.229.
  152. Dowman JK, Gunson BK, Mirza DF, et al. UK experience of liver transplantation for erythropoietic protoporphyria. J Inherit Metab Dis 2011; 34:539.
  153. Bloomer JR, Pierach CA. Effect of hematin administration to patients with protoporphyria and liver disease. Hepatology 1982; 2:817.
  154. Lamon JM, Poh-Fitzpatrick MB, Lamola AA. Hepatic protoporphyrin production in human protoporphyria. Effects of intravenous hematin and analysis of erythrocyte protoporphyrin distribution. Gastroenterology 1980; 79:115.
  155. Reichheld JH, Katz E, Banner BF, et al. The value of intravenous heme-albumin and plasmapheresis in reducing postoperative complications of orthotopic liver transplantation for erythropoietic protoporphyria. Transplantation 1999; 67:922.
  156. Wahlin S, Harper P. Pretransplant albumin dialysis in erythropoietic protoporphyria: a costly detour. Liver Transpl 2007; 13:1614.
  157. Krishnamurthy P, Xie T, Schuetz JD. The role of transporters in cellular heme and porphyrin homeostasis. Pharmacol Ther 2007; 114:345.
  158. Eichbaum QG, Dzik WH, Chung RT, Szczepiorkowski ZM. Red blood cell exchange transfusion in two patients with advanced erythropoietic protoporphyria. Transfusion 2005; 45:208.
  159. van Wijk HJ, van Hattum J, Baart de la Faille H, et al. Blood exchange and transfusion therapy for acute cholestasis in protoporphyria. Dig Dis Sci 1988; 33:1621.
  160. PORTER FS, LOWE BA. CONGENITAL ERYTHROPOIETIC PROTOPORPHYRIA. I. CASE REPORTS, CLINICAL STUDIES AND PORPHYRIN ANALYSES IN TWO BROTHERS. Blood 1963; 22:521.
  161. Gross U, Frank M, Doss MO. Hepatic complications of erythropoietic protoporphyria. Photodermatol Photoimmunol Photomed 1998; 14:52.
  162. Rademakers LH, Cleton MI, Kooijman C, et al. Early involvement of hepatic parenchymal cells in erythrohepatic protoporphyria? An ultrastructural study of patients with and without overt liver disease and the effect of chenodeoxycholic acid treatment. Hepatology 1990; 11:449.
  163. Ishibashi A, Ogata R, Sakisaka S, et al. Erythropoietic protoporphyria with fatal liver failure. J Gastroenterol 1999; 34:405.
  164. Stathers GM. Porphyrin-binding effect of cholestyramine. Results of in-vitro and in-vivo studies. Lancet 1966; 2:780.
  165. Komatsu H, Ishii K, Imamura K, et al. A case of erythropoietic protoporphyria with liver cirrhosis suggesting a therapeutic value of supplementation with alpha-tocopherol. Hepatol Res 2000; 18:298.
  166. Wells MM, Golitz LE, Bender BJ. Erythropoietic protoporphyria with hepatic cirrhosis. Arch Dermatol 1980; 116:429.
  167. Anstey AV, Hift RJ. Liver disease in erythropoietic protoporphyria: insights and implications for management. Gut 2007; 56:1009.
  168. Wahlin S, Stal P, Adam R, et al. Liver transplantation for erythropoietic protoporphyria in Europe. Liver Transpl 2011; 17:1021.
  169. McGuire BM, Bonkovsky HL, Carithers RL Jr, et al. Liver transplantation for erythropoietic protoporphyria liver disease. Liver Transpl 2005; 11:1590.
  170. Singal AK, Parker C, Bowden C, et al. Liver transplantation in the management of porphyria. Hepatology 2014; 60:1082.
  171. Do KD, Banner BF, Katz E, et al. Benefits of chronic plasmapheresis and intravenous heme-albumin in erythropoietic protoporphyria after orthotopic liver transplantation. Transplantation 2002; 73:469.
  172. Rand EB, Bunin N, Cochran W, et al. Sequential liver and bone marrow transplantation for treatment of erythropoietic protoporphyria. Pediatrics 2006; 118:e1896.
  173. Windon AL, Tondon R, Singh N, et al. Erythropoietic protoporphyria in an adult with sequential liver and hematopoietic stem cell transplantation: A case report. Am J Transplant 2018; 18:745.
  174. Wahlin S, Harper P. The role for BMT in erythropoietic protoporphyria. Bone Marrow Transplant 2010; 45:393.
  175. Poh-Fitzpatrick MB, Wang X, Anderson KE, et al. Erythropoietic protoporphyria: altered phenotype after bone marrow transplantation for myelogenous leukemia in a patient heteroallelic for ferrochelatase gene mutations. J Am Acad Dermatol 2002; 46:861.
  176. Ardalan ZS, Chandran S, Vasudevan A, et al. Management of Patients With Erythropoietic Protoporphyria-Related Progressive Liver Disease. Liver Transpl 2019; 25:1620.
  177. Jong CT, Finlay AY, Pearse AD, et al. The quality of life of 790 patients with photodermatoses. Br J Dermatol 2008; 159:192.
Topic 7125 Version 52.0

References

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