Protoporphyria

Back

Background

The term protoporphyria now encompasses two clinically similar disorders that most often result from hereditary mutations in one of three different genes. The most common is erythropoietic protoporphyria (EPP). It is an inherited disorder caused by partial deficiency in mitochondrial ferrochelatase (FECH), the terminal enzyme of heme biosynthesis. The resultant accumulated excess of its substrate, metal-free protoporphyrin, causes two principal manifestations: (1) an acute cutaneous photosensitivity typically first appearing during childhood and (2) hepatobiliary disease.[1, 2, 3, 4, 5]

The predominant genotype associated with phenotypic expression of EPP is one mutant ferrochelatase allele (FECH) encoding a defective enzyme protein with little or no function, paired with a relatively common polymorphic allele (IVS3-48T>C) with low gene expression that only mildly affects heme synthesis.[6, 7] This type of inheritance has been termed pseudodominant[8, 9] or semidominant,[10] but is often referred to as autosomal recessive[11] in that anomalies in both paired FECH alleles are required for disease expression, even though one alteration causes marked reduction or abrogation of residual enzyme activity while the polymorphism causes little clinically noticeable harm when unpaired with a severely dysfunctional allele. Infrequently, two deleterious FECH mutations are paired in a recessive genotype of EPP that may impart a higher risk for hepatic dysfunction.[12, 13] Rarely, acquired somatic mutation or deletion of a ferrochelatase gene secondary to myelodysplastic or myeloproliferative disorders leads to an adult-onset protoporphyric disorder.[14, 15, 16]

A far less frequent type (< 10%) of hereditary protoporphyria, now recognized as a separate disorder, is X-linked dominant protoporphyria (XLDPP, OMIM 300752), or simply XLP. XLP arises from C-terminal deletions or alterations in the gene encoding the erythroid-specific enzyme 5-aminolevulinic acid synthase-2 (ALAS2), increased function of which leads to overproduction of protoporphyrin.[11, 17] Like EPP, XLP is caused by bone marrow heme synthetic dysfunction, but most often results in a greater ratio of accumulated erythrocyte zinc-protoporphyrin to metal-free protoporphyrin than is typical for EPP. XLP manifests as an acute, childhood-onset, cutaneous photosensitivity indistinguishable from that of EPP, but appears to have a higher risk for hepatic dysfunction.[17] An adult-onset case of XLP in an 89-year-old man with evolving myelodysplasia exhibited somatic mosaicism in erythroid hematopoietic cells associated with an ALAS2 mutation that predicted a C-terminal deletion.[18]

Mutation in a third gene, CLPX, which encodes the mitochondrial AAA+ unfoldase ClpX, has been linked to a familial disorder with biochemical and clinical features of protoporphyria in individuals without either FECH or ALAS2 mutations.[19] ClpX acts to control ALAS activation and degradation during heme synthesis. Defective CLPX leads to increased ALAS post-translational stability, which results in excess accumulation of erythrocyte protoporphyrin.[19]

Pathophysiology

Protoporphyrin is a lipophilic molecule capable of transformation to excited states by absorption of light energy. Excited-state protoporphyrin mediates photoxidative damage to biomolecular targets in the skin,[20] resulting in immediate phototoxic symptoms variously described as tingling, stinging, or burning that may be followed by the appearance of erythema, edema, and purpura.[1, 20] Excess protoporphyrin is formed during maturation of erythroid cells in the bone marrow and present at the highest levels in reticulocytes and young erythrocytes.[21] Metal-free protoporphyrin escapes from red blood cells into the plasma, from which it is cleared by the liver and secreted into bile. Protoporphyrin-rich bile facilitates gallstone formation.[22] Toxic effects of protoporphyrin deposition in the liver may lead to life-threatening hepatic dysfunction.[22, 23, 24] 99

Etiology

The cytogenetic location of the ferrochelatase gene is 18q21.3.[25] Loss of activity by as much as 50% as the result of one FECH mutant gene is generally insufficient to cause overt disease when its complementary allele has normal function.[6] FECH genotypes composed of either two mutant alleles (< 1-4% of cases) or one mutation and a variant allele with a specific intronic single nucleotide polymorphism (IVS3-48C) (~82-94% of cases) have been found in most symptomatic individuals.[9, 12, 13, 26, 27] This polymorphism enhances aberrant splicing and rapid degradation of FECH mRNA, with resultant low expression.[7] The allele frequency of this polymorphism varies widely in diverse populations studied, as follows:

The pairing of a mutated allele encoding a severely impaired enzyme protein with this low-expressing polymorphic allele typically yields enzyme activity diminished to less than 30% of normal, low enough to cause protoporphyrin accumulation. Individuals with no FECH mutation but who are heterozygous for this polymorphism typically do not have sufficiently diminished FECH activity to cause clinical abnormalities. Individuals with no FECH mutation, but who are homozygous for this polymorphism, may exhibit slightly abnormal erythrocyte protoporphyrin levels and mild photosensitivity.[32]

Adult-onset protoporphyric photosensitivity and increased protoporphyrin levels have been associated with an acquired somatic mutation or deletion of a FECH gene due to myelodysplastic or myeloproliferative disorders.[14, 15, 16]

Eight families were described in 2008[17] with a protoporphyric disorder indistinguishable clinically from the predominant form of the disease, but without FECH mutations, that is now called X-linked dominant protoporphyria or X-linked protoporphyria (XLDPP, XLP or XLEPP [OMIM 300752]). Two different C-terminal deletions in the gene encoding the erythroid-specific isoform of aminolevulinic acid synthase were identified among these families. The locus for this gene was identified on the X-chromosome, and the inheritance pattern in the families was consistent with X-linked dominant transmission. Both mutations caused a marked increase in activity of ALAS2 that eventuated in large accumulations of erythrocyte metal-free protoporphyrin and zinc-protoporphyrin. Seventeen percent of affected individuals in that study exhibited overt liver disease (40% of affected males), a significantly greater number than the 2-5% of individuals with ferrochelatase-deficient protoporphyria who develop this complication.

Additional cases of XLP and novel associated gain-of-function ALAS2 mutations have subsequently been recognized.[9, 11, 18, 33, 34, 35] A higher prevalence (~10%) of XLP was found among 226 North American individuals with the protoporphyria phenotype; this is 2-5 times greater than observed among Western Europeans previously studied.[11]

Epidemiology

Frequency

United States

Until the recently established registry for protoporphyria sponsored by the American Porphyria Foundation collects sufficient data, accurate enumeration in the United States cannot be provided, but it is probably similar to data from European countries. A study of 226 North American individuals exhibiting the protoporphyria phenotype identified 22 with ALAS2 mutations and 187 with FECH anomalies.[33] The ALAS2/FECH ratio of approximately 10% in this study is greater than ratios reported elsewhere.

International

Estimates of one EPP case in populations of 75,000-200,000 have been reported for several Western European populations and in the South African population of European ancestry.[3, 36, 37, 38] XLP remains rare but has been identified in increasing numbers.[9, 11, 17, 33, 34, 35, 39]

Race

EPP has been reported most often in people with white heritage, but it has also been reported in persons with Japanese, Chinese, East Indian, or north or central African ancestry. XLP has been identified chiefly among individuals of Western European ancestry but also in an African American and Pacific Islander[11] and a Japanese boy.[34]

Sex

EPP and XLP occur in both males and females.

Age

Photocutaneous symptoms usually appear during childhood,[1] but they also may be noted for the first time in adult life.[14, 15, 16, 18, 39] Gallstones may become symptomatic in young adulthood or in middle age.[1] Liver failure and its complications, sufficiently severe to result in liver transplantation and/or death, may develop in children and adolescents as well as adults.[22, 24, 40, 41, 42]

Prognosis

In the absence of hepatic failure, individuals with EPP have normal life expectancies.

Painful cutaneous photosensitivity reduces the sunlight tolerance of individuals with protoporphyria and may influence their lifestyles over entire lifetimes.[1]

An increased prevalence of cholelithiasis in both men and women can result in signs and symptoms of gallstone disease at relatively early ages.[1]

Hepatotoxic effects of excess protoporphyrin deposition have led to liver dysfunction that progressed to life-threatening severity in approximately 2-5% of known cases of protoporphyria.[3]

History

Skin manifestations after sunlight exposure typically begin during infancy or childhood, most often involving dorsal hands, the face and ears, and, occasionally, legs and dorsal feet, after short periods of exposure. If exposure is promptly discontinued, visible skin lesions may not ensue. Longer exposure, or multiple exposures on sequential days, can elicit swelling with or without redness in the exposed skin that evolves into sheets of petechiae. This exquisitely painful reaction resolves over several days to leave skin that may appear normal. The onset after sun exposure varies but may be within 10 minutes. Older children and adults, particularly those with darker skin, may handle being outdoors for an hour or more.

Patients with protoporphyria who report skin pain but have minimal findings may be considered malingerers until an acute reaction is observed. Gallstones may remain silent or evoke reports of indigestion and/or right upper quadrant abdominal pain consistent with symptomatic cholelithiasis. Individuals with protoporphyria associated with hepatotoxicity may report loss of appetite, nausea, vomiting, weakness and fatigue, anorexia, malaise, weight changes, increasing abdominal girth, abdominal pain, jaundice, and increasing photosensitivity.

Physical Examination

The acute phototoxic reaction typically includes edema, erythema, and petechiae. Blisters, crusted erosions, and scarring may occur but are less florid and less frequent than in other porphyrias. Chronic changes include shallow, elongated depressions in facial skin, especially over the nose; perioral furrowing; and prematurely aged skin of the dorsal hands, often most prominent over the knuckles. In more severe cases, sclerodermalike waxy induration or a cobblestone texture of facial and hand skin may develop. Mechanical fragility, when present, is less severe than in other porphyrias; hypertrichosis is infrequent.

Some individuals with autosomal recessive (two deleterious FECH mutations) erythropoietic protoporphyria (EPP) exhibit palmar keratoderma that often worsens in summer and remits in winter.[43, 44] Neuropathy has been observed in some of these individuals, but not severe liver dysfunction, suggesting that protoporphyrin hepatotoxicity may be mitigated in this subset of recessive protoporphyria patients by efficient hepatobiliary protoporphyrin excretion mechanisms yet to be elucidated.[44]

With progressive liver dysfunction, hepatosplenomegaly and jaundice may develop, as may signs of increasing cutaneous photosensitivity. End-stage liver disease is signaled by intense jaundice, ascites, vomiting, fever, encephalopathy, axonal polyneuropathy that may progress to paresis and respiratory failure, hemorrhage from esophageal varices, and extreme photosensitivity.

Complications

Severe neurological dysfunction (eg, encephalopathy, axonal polyneuropathy, respiratory failure) characteristic of acute porphyria attacks has been observed in patients with end-stage hepatic failure associated with erythropoietic protoporphyria (EPP).[45, 46, 47]

Pregnancy does not cause worsening of EPP; photosensitivity may actually improve during gestation.[37]

Laboratory Studies

Protoporphyrin concentration is elevated in red blood cells, plasma, bile, and feces. The diagnosis is usually made by finding the abnormal levels in erythrocytes and plasma. Urinary porphyrin levels are normal in patients without liver dysfunction. Abnormal coproporphyrinuria develops when liver function is deteriorating.[48, 49]

The screening test for either type of protoporphyria is measurement of total blood porphyrin, which includes both metal-free protoporphyrin and zinc protoporphyrin. Levels that are 5-50 times the upper limit of the normal range are diagnostic. Separate determination of zinc protoporphyrin as a fraction of the total is helpful for a preliminary differentiation between erythropoietic protoporphyria (EPP) and X-linked protoporphyria. In EPP, zinc protoporphyrin constitutes approximately 5% of the total protoporphyrin in blood, whereas in X-linked protoporphyria, it constitutes 20-40%. The determination directs attention to the appropriate gene for mutation analysis. Some reference laboratories measure only zinc protoporphyrin but label it, misleadingly, as “free protoporphyrin.” The level of zinc protoporphyrin is elevated in patients with iron deficiency and lead poisoning. The test is not useful in screening for protoporphyria.

Obtain a serum liver function panel at diagnosis. Monitor indices of liver function at 6- to 12-month intervals if baseline values are normal. If liver function is abnormal, complicating factors (eg, gallstones, viral hepatitis, alcohol or drug abuse, other toxic, infectious, immunologic, or metabolic storage disorders) should be excluded by appropriate testing.

Perform a hematological assessment. Microcytic anemia, which may occur in 20-60% of individuals with protoporphyria,[50] often associated with mild iron deficiency, may be found.[1, 51] Iron therapies have resulted in both beneficial[52, 53, 54] and adverse[55, 56] effects in EPP. Iron supplementation has been beneficial in some cases of XLP.[35]

A lifetime of sunlight avoidance predisposes to vitamin D deficiency among individuals with protoporphyria.[57, 58] Determine serum 25-hydroxyvitamin D level at diagnosis and at intervals as needed to monitor response to supplementation, if indicated.

Imaging Studies

If cholelithiasis is suspected, abdominal ultrasonography or other imaging procedures are indicated.

Bone mineral density studies may reveal osteopenic or osteoporotic changes due to vitamin D deficiency as a result of chronic sunlight avoidance.[58]

Other Tests

Impending liver failure may be signaled by progressively rising levels of urinary coproporphyrin.[49] Urinary porphyrin levels are within normal limits in persons with uncomplicated protoporphyria. Protoporphyrin, being lipophilic, is not excreted by renal mechanisms and does not normally appear in urine. Coproporphyrin, which accumulates as a result of liver disease, has intermediate water solubility, and levels become abnormally elevated in the urine of patients developing protoporphyrin-induced hepatotoxicity.[48]

Procedures

In the event of overt liver dysfunction, liver biopsy is indicated. Some experts suggest that individuals with genotypes associated with higher risk of liver disease, such as a "null-allele" FECH mutation that encodes an enzyme with essentially no residual activity, biallelic FECH mutations, one of the ALAS2 increased-function mutations, or a family history of protoporphyric liver disease, should undergo liver biopsy even before liver function tests become abnormal.[24, 36] The presence of other risk factors for liver disease, such as viral hepatitis, hemochromatosis, or alcoholic or nonalcoholic fatty liver, increases the weight of argument for earlier liver biopsy.

Liver transplantation may be life-saving, but does not cure protoporphyria because the source of most of the excess protoporphyrin is bone marrow erythropoiesis. Continued overproduction of protoporphyrin eventually leads to its deposition in the engrafted liver, which may again become dysfunctional.[42] Bone marrow or peripheral stem cell transplantation can be curative, but attendant risks have restricted use of these procedures to a limited number of highly selected cases.[15, 35, 41, 59, 60] Research in animal models has shown promising developments in gene therapy strategies that may eventually be transferrable to humans.

Histologic Findings

Light microscopy examination of the acute skin reaction shows nonspecific perivascular and interstitial neutrophilic dermal infiltrates. Subepidermal blisters similar to those seen in porphyria cutanea tarda can be seen in early lesions of erythropoietic protoporphyria (EPP). These blisters are cell-poor and inflammatory infiltrate is sparse. “Caterpillar bodies” may be seen in the basal layer epidermis overlying blisters. Ultrastructural findings in the acute reaction include damage of endothelial cells with extravasation of intravascular contents and degranulated mast cells.[61]

Biopsy specimens of chronically damaged skin show deposition of hyaline masses in the upper dermis and markedly thickened walls of upper dermal capillaries.[62] The deposition can be extensive and involve the surrounding dermis to mimic a colloid milium. The hyaline material surrounding blood vessels is periodic acid-Schiff–positive and diastase-resistant. Ultrastructural findings in chronically damaged skin include replicated basal laminae around dermal vessels, degranulated mast cells, and amorphous dermal deposits.[62] Direct immunofluorescence studies show deposition of immunoglobulins and complement in and around upper dermal vessel walls and, to a lesser extent, at the dermoepidermal junction.[62]

Liver biopsy typically reveals brown pigment in hepatocytes, Kupffer cells, portal macrophages, and small biliary structures.[22, 42] Many of these protoporphyrin deposits are crystalline when examined under electron microscopy and birefringent when examined under polarization microscopy.[22, 42] Cirrhotic changes are seen in advanced disease, including fibrous expansion of portal areas and regenerative nodules.[22, 42]

Medical Care

For protoporphyria uncomplicated by hepatobiliary disease, the major problem is lifelong cutaneous photosensitivity. Avoiding sunlight is the mainstay of management.

Opaque topical sunscreens or ultraviolet (UV) B phototherapy may improve the tolerance of light. Symptom prevention was impossible until 2006 when afamelanotide, a melanocyte-stimulating hormone analogue that induces epidermal melanin skin-tanning and has an anti-inflammatory effect on the skin, became available. Afamelanotide is a newer therapy based on the observation that sun-related symptoms are inversely related to skin pigmentation in people with protoporphyria.[63, 64, 65] This congener of α-melanocyte–stimulating hormone increases production of eumelanin. It is supplied as a sustained-release subcutaneous implant. Skin darkening starts within a few days after placement of the implant and persists for 3-4 weeks.[66]

US approval of afamelanotide was based on 2 multicenter, randomized, double-blind, placebo-controlled trials in patients residing in the European Union (n=74) and the United States (n=94). Patients were randomly assigned, in a 1:1 ratio, to receive a subcutaneous implant containing either afamelanotide or placebo every 60 days (a total of 5 implants in the European Union study and 3 in the US study). The type and duration of sun exposure, number and severity of phototoxic reactions, and adverse events were recorded over the respective 180-day and 270-day study periods. In the US study, the duration of pain-free time after 6 months was longer in the afamelanotide group (median, 69.4 h vs 40.8 h in the placebo group; P = .04). In the European Union study, the duration of pain-free time after 9 months was also longer in the afamelanotide group than in the placebo group (median, 6 h vs 0.8 h; P = .005), and the number of phototoxic reactions was lower in the afamelanotide group (77 vs 146, P = .04).[67]

Note the following treatment measures for photosensitivity:

Anemia, if present in EPP, most often is mild but may have features of iron deficiency. Iron supplementation has both improved[52, 53, 54] and worsened[55, 56] EPP. In one X-linked dominant protoporphyria patient, iron therapy was followed by improvements in protoporphyrin overload, liver damage, and anemia.[35] Cholelithiasis is managed surgically. Liver dysfunction is an ominous development for which medical remedies are not consistently effective. Progressive intractable liver insufficiency is an indication for liver transplantation.[22, 24, 76]

Although adverse reactions to porphyrinogenic drugs known to exacerbate acute hepatic porphyrias are not characteristic of protoporphyria, avoid or administer with caution drugs with cholestatic properties, such as estrogenic hormones. Assess the risk-to-benefit ratio for each individual with protoporphyria when considering use of cholestatic therapies.

Immunization against viral hepatitis agents should be offered.

Supplementation with cholecalciferol (vitamin D3) should be given if serum vitamin D levels are low and/or bone density studies reveal osteopenia or osteoporosis.

Medical approaches to reversing protoporphyric liver dysfunction are not well established, owing to inconsistent or uncertain efficacy and experience in relatively few cases. These include the following:

Surgical Care

Failure of medical reversal of protoporphyrin-induced hepatic decompensation warrants liver transplantation. Operating room illumination has caused acute phototoxic damage to skin and internal organs during liver transplantation for protoporphyric liver failure.[45, 46, 60, 84] This procedure requires several hours' exposure of skin and internal organs to intense visible light at a time when the patient's accumulated protoporphyrin levels are typically extremely high, causing severe photosensitivity. Preoperative exchange transfusions, plasmapheresis, and/or infusion of a heme analogue may lower the circulating burden of protoporphyrin in the blood, reducing intraoperative phototoxic potential.[85] These treatments may also aid postoperatively in retarding the development of protoporphyrin hepatotoxicity in the engrafted liver.[80, 81]

It has been recommended that a specific flexible yellow filter excluding wavelengths below 470 nm be used over operating room lamps to optimally attenuate phototoxic damage to patients with EPP during liver transplantation, yet provide acceptable illumination for the surgeons.[86] They found the risk of phototoxic injury in endoscopic, laparoscopic, and non–liver transplant surgery in such patients to be low and recommended that protective measures, including filters for operating room lamps, should be reserved for liver transplantation or for other prolonged surgical procedures in cholestatic patients. Case-by-case judgment was advised before any procedure involving intense lighting, considering duration of irradiation, light emission spectrum, and patient variables such as evidence of cholestasis or liver impairment, after burning pain, edema, and erythema developed in an arm exposed to unfiltered surgical lighting after a one-hour procedure performed on a man whose erythrocyte protoporphyrin 18 months prior was 98.3 μM/L (~ 5532 μg/dL).[87]

Biliary complications (stones, sludge, strictures) are more frequent after liver transplantation for EPP compared with the general population of transplant patients; therefore, consideration of Roux loop reconstruction is recommended.[42, 60]

Adverse reactions to anesthetic agents problematic in acute hepatic porphyrias are not characteristic of protoporphyria.

Consultations

Consultation with a hematologist should be sought for management of anemia, particularly before instituting iron supplementation, or if hypertransfusion, exchange transfusion, or plasmapheresis is considered. Rarely, bone marrow transplantation may have a role in the management of selected patients with severe manifestations.[41, 59]

Referral to specialists at a comprehensive liver center should be arranged at the earliest signs of liver decompensation for assistance in evaluation and management of progressive liver dysfunction. If liver transplantation becomes necessary, a successful outcome is more likely if the procedure is performed before the patient is gravely debilitated.

Referral to a medical geneticist can aid in counseling patients and families about risks of inheriting or transmitting the mutations and polymorphisms associated with protoporphyrias.[88, 89]

Preoperative consultation with anesthesiologists and biomedical engineers is essential concerning use of appropriate filters over operating room lighting during liver transplantation or for other procedures for protoporphyria patients with high circulating protoporphyrin levels and/or anticipated severe photosensitivity.

Diet

Do not severely curtail carbohydrate intake; a "glucose effect" may beneficially modulate abnormal heme synthesis.[90] Limit use of ethanol; alcohol excess has been implicated in fatal protoporphyria associated with liver failure.[91] Vitamin D and calcium-rich foods or supplements may reduce the incidence of osteopenia or osteoporosis associated with chronic sunlight avoidance.

Activity

Sunlight avoidance is mandatory. Recommend adjustment of outdoor activities to avoid midday sunlight. Stylish and comfortable sun-protective clothing is commercially available that can reduce time constraints on many outdoor sports or activities. Specialized programs for photosensitive children can be found that offer safe and healthy recreational experiences, even a summer camp organized by the Xeroderma Pigmentosum Society.

Medication Summary

Synthetic beta-carotene (Lumitene), an oral photoprotective agent, is available as a nonprescription product. Cysteine showed benefit in clinical trials, but is rarely used. Pyridoxine was reported effective in two cases. H1-receptor blockade may reduce symptoms due to mast cell histamine release during acute phototoxic reactions if established prior to exposure. Whether H2-receptor antagonists reproducibly slow porphyrin production in various porphyrias remains unproven.

Afamelanotide is a synthetic alpha-melanocyte–stimulating hormone analogue that increases the production of eumelanin in skin, thus producing a tan. Afamelanotide binds to the melanocortin-1 receptor (MC1R). MC1R signaling up-regulates the synthesis of eumelanin and pheomelanin, induces antioxidant activity, enhances DNA repair processes, and modulates inflammation. It is administered as a subcutaneous implant containing 16 mg of afamelanotide. It is approved in the European Union and the United States for use in adults with erythropoietic protoporphyria (EPP).

Liver dysfunction warrants individualized design of therapeutic regimens that may include the administration of enteric sorbents to promote protoporphyrin excretion, bile acids to enhance porphyrin clearance from the liver, and hematin to repress porphyrin production. Combinations of these and other adjunctive agents and modalities may moderate the urgency presented by a failing organ, allowing orderly preparation for an optimal transplantation.

Afamelanotide (Scenesse)

Clinical Context:  Erythropoietic protoporphyria is a rare disorder caused by mutations leading to impaired activity of ferrochelatase, an enzyme involved in heme production; decreased ferrochelatase activity leads to protoporphyrin IX (PPIX) accumulation in the body; light reaching the skin can react with PPIX causing intense skin pain and skin changes (eg, redness, thickening). Afamelanotide, a melanocortin-1 receptor (MC1-R) agonist, increases production of eumelanin (the most common type of melanin) in the skin independent of exposure to sunlight or artificial light sources. It is indicated to increase pain-free light exposure in adults with history of phototoxic reactions from erythropoietic protoporphyria (EPP).

Class Summary

Alpha melanocyte stimulating hormone analogs increase production of eumelanin (the most common type of melanin) in the skin independent of exposure to sunlight or artificial light sources. 

Beta carotene (Lumitene)

Clinical Context:  The exact mechanism of action is not completely elucidated. The patient must become carotenemic before effects are observed. More than one internal light screen may be responsible for effects. This agent may provide a limited level of photoprotection. It causes yellowing of skin (carotenoderma). Any photoprotection afforded increases slowly over a 4- to 6-week period after drug therapy is commenced. When discontinued, skin color and benefit diminish over several weeks.

Class Summary

Beta-carotene is a scavenger of singlet-exited oxygen and is believed to interfere with the efficiency of porphyrin-sensitized photoxidative damage in the skin. Ingestion of beta-carotene at recommended doses produces carotenodermia after several weeks. Increasing tolerance of sunlight develops during this loading period. Tolerance diminishes over several weeks when treatment is stopped. Only some individuals with EPP reported that beta-carotene provided photoprotection; others reported no noticeable benefit.

Fexofenadine (Allegra)

Clinical Context:  Fexofenadine is a nonsedating second-generation medication with fewer adverse effects than first-generation medications. It competes with histamine for H1 receptors in the GI tract, blood vessels, and respiratory tract, reducing hypersensitivity reactions. Fexofenadine does not sedate. It is available in daily and twice-daily preparations.

Class Summary

H1-receptor antagonists modulate effects of histamine in skin. If taken prior to anticipated strong sunlight exposure that cannot be avoided, acute reactions may be attenuated to some extent; minimal benefit is expected if taken afterward.

Cimetidine (Tagamet)

Clinical Context:  Cimetidine is an H2 antagonist, which, when combined with an H1-type, may be useful in treating itching and flushing in urticaria. Porphyria-specific usage for inhibiting overproduction of porphyrins is experimental.

Class Summary

These agents produce blockade of H2 receptors.

Cholestyramine (Questran)

Clinical Context:  Cholestyramine is a polymeric resin that binds bile acids, porphyrins, and other molecules to form nonabsorbable complexes that are excreted unchanged in feces. It adsorbs many drugs and nutrients; long-term use requires proper timing of oral drugs and may warrant supplementation of vitamins D, E, A, and K.

Activated charcoal (Actidose)

Clinical Context:  Activated charcoal prevents absorption by adsorbing porphyrin in the intestine. Multidose charcoal may interrupt enterohepatic recirculation and enhance elimination by enterocapillary exsorption. It does not dissolve in water. It adsorbs many medications and nutrients; long-term use requires proper timing of oral drugs and may warrant supplementation of vitamins D, E, A, and K.

Class Summary

Agents that bind protoporphyrin in the intestinal lumen promote its excretion by interrupting enterohepatic recirculation, thereby reducing the porphyrin load presented to the liver for clearance.

Ursodiol (Actigall)

Clinical Context:  Ursodiol has been shown to promote bile flow in cholestatic conditions associated with a patent extrahepatic biliary system. It decreases the cholesterol content of bile and therefore reduces bile stone and sludge formation.

Class Summary

Increasing bile flow enhances secretion of protoporphyrin by the liver into the enteric tract and clearance from the body.

Hemin (Panhematin)

Clinical Context:  Hemin in an enzyme inhibitor derived from processed red blood cells and is an iron-containing metalloporphyrin. It was previously known as hematin, a term used to describe the chemical reaction product of hemin and sodium carbonate solution.

It has an anticoagulant effect and may cause thrombophlebitis at the infusion site. It must be reconstituted from lyophilized powder. Reconstitute it with human serum albumin 25% (132 mL of 25% human serum albumin to 1 vial of hemin [301 mg heme]).

Class Summary

Intravenous infusion of a heme analogue may repress heme synthesis in liver and bone marrow cells, thereby reducing rate of protoporphyrin overproduction.

Author

Jose A Plaza, MD, Director of Dermatopathology, Department of Pathology, Froedtert Hospital; Associate Professor, Department of Pathology, Section of Dermatopathology, Medical College of Wisconsin

Disclosure: Nothing to disclose.

Coauthor(s)

,

Disclosure: Nothing to disclose.

Specialty Editors

David F Butler, MD, Former Section Chief of Dermatology, Central Texas Veterans Healthcare System; Professor of Dermatology, Texas A&M University College of Medicine; Founding Chair, Department of Dermatology, Scott and White Clinic

Disclosure: Nothing to disclose.

Edward F Chan, MD, Clinical Assistant Professor, Department of Dermatology, University of Pennsylvania School of Medicine

Disclosure: Nothing to disclose.

Chief Editor

William D James, MD, Paul R Gross Professor of Dermatology, Vice-Chairman, Residency Program Director, Department of Dermatology, University of Pennsylvania School of Medicine

Disclosure: Received income in an amount equal to or greater than $250 from: Elsevier; WebMD.

Additional Contributors

Maureen B Poh-Fitzpatrick, MD, Professor Emerita of Dermatology and Special Lecturer, Columbia University College of Physicians and Surgeons

Disclosure: Nothing to disclose.

Acknowledgements

Günter Burg, MD Professor and Chairman Emeritus, Department of Dermatology, University of Zürich School of Medicine; Delegate of The Foundation for Modern Teaching and Learning in Medicine Faculty of Medicine, University of Zürich, Switzerland

Günter Burg, MD is a member of the following medical societies: American Academy of Dermatology, American Dermatological Association, International Society for Dermatologic Surgery, North American Clinical Dermatologic Society, and Pacific Dermatologic Association

Disclosure: Nothing to disclose.

References

  1. DeLeo VA, Poh-Fitzpatrick M, Mathews-Roth M, Harber LC. Erythropoietic protoporphyria. 10 years experience. Am J Med. 1976 Jan. 60(1):8-22. [View Abstract]
  2. Baart de la Faille H, Bijlmer-Iest JC, van Hattum J, Koningsberger J, Rademakers LH, van Weelden H. Erythropoietic protoporphyria: clinical aspects with emphasis on the skin. Curr Probl Dermatol. 1991. 20:123-34. [View Abstract]
  3. Todd DJ. Erythropoietic protoporphyria. Br J Dermatol. 1994 Dec. 131(6):751-66. [View Abstract]
  4. Bissell DM, Anderson KE, Bonkovsky HL. Porphyria. N Engl J Med. 2017 Aug 31. 377 (9):862-872. [View Abstract]
  5. Karim Z, Lyoumi S, Nicolas G, Deybach JC, Gouya L, Puy H. Porphyrias: A 2015 update. Clin Res Hepatol Gastroenterol. 2015 Sep. 39 (4):412-25. [View Abstract]
  6. 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 Mar 15. 93(6):2105-10. [View Abstract]
  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 Jan. 30(1):27-8. [View Abstract]
  8. Schmitt C, Ducamp S, Gouya L, Deybach JC, Puy H. [Inheritance in erythropoietic protoporphyria]. Pathol Biol (Paris). 2010 Oct. 58(5):372-80. [View Abstract]
  9. Whatley SD, Mason NG, Holme SA, Anstey AV, Elder GH, Badminton MN. Molecular epidemiology of erythropoietic protoporphyria in the U.K. Br J Dermatol. 2010 Mar. 162(3):642-6. [View Abstract]
  10. Frank J, Poblete-Gutiérrez P. Delayed diagnosis and diminished quality of life in erythropoietic protoporphyria: results of a cross-sectional study in Sweden. J Intern Med. 2011 Mar. 269(3):270-4. [View Abstract]
  11. Balwani M, Doheny D, Bishop DF, Nazarenko I, Yasuda M, Dailey HA. 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. 19:26-35. [View Abstract]
  12. Whatley SD, Mason NG, Khan M, et al. Autosomal recessive erythropoietic protoporphyria in the United Kingdom: prevalence and relationship to liver disease. J Med Genet. 2004 Aug. 41(8):e105. [View Abstract]
  13. 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 Jan. 78(1):2-14. [View Abstract]
  14. 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 Jan 1. 107(1):60-2. [View Abstract]
  15. Poh-Fitzpatrick MB, Wang X, Anderson KE, Bloomer JR, Bolwell B, Lichtin AE. Erythropoietic protoporphyria: altered phenotype after bone marrow transplantation for myelogenous leukemia in a patient heteroallelic for ferrochelatase gene mutations. J Am Acad Dermatol. 2002 Jun. 46(6):861-6. [View Abstract]
  16. Aplin C, Whatley SD, Thompson P, et al. Late-onset erythropoietic porphyria caused by a chromosome 18q deletion in erythroid cells. J Invest Dermatol. 2001 Dec. 117(6):1647-9. [View Abstract]
  17. Whatley SD, Ducamp S, Gouya L, Grandchamp B, Beaumont C, Badminton MN. 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 Sep. 83(3):408-14. [View Abstract]
  18. Livideanu CB, Ducamp S, Lamant L, Gouya L, Rauzy OB, Deybach JC. Late-onset X-linked dominant protoporphyria: an etiology of photosensitivity in the elderly. J Invest Dermatol. 2013 Jun. 133(6):1688-90. [View Abstract]
  19. Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, 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 Sep 19. 114 (38):E8045-E8052. [View Abstract]
  20. Poh-Fitzpatrick MB. Porphyrin-sensitized cutaneous photosensitivity: pathogenesis and treatment. Clin Dermatol. 1985 Apr-Jun. 3(2):41-82. [View Abstract]
  21. Piomelli S, Lamola AA, Poh-Fitzpatrick MF, Seaman C, Harber LC. 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 Dec. 56(6):1519-27. [View Abstract]
  22. Bloomer JR. The liver in protoporphyria. Hepatology. 1988 Mar-Apr. 8(2):402-7. [View Abstract]
  23. Gross U, Frank M, Doss MO. Hepatic complications of erythropoietic protoporphyria. Photodermatol Photoimmunol Photomed. 1998 Apr. 14(2):52-7. [View Abstract]
  24. Anstey AV, Hift RJ. Liver disease in erythropoietic protoporphyria: insights and implications for management. Gut. 2007 Jul. 56(7):1009-18. [View Abstract]
  25. Brenner DA, Didier JM, Frasier F, Christensen SR, Evans GA, Dailey HA. A molecular defect in human protoporphyria. Am J Hum Genet. 1992 Jun. 50(6):1203-10. [View Abstract]
  26. Saruwatari H, Ueki Y, Yotsumoto S, Shimada T, Fukumaru S, Kanekura T, et al. Genetic analysis of the ferrochelatase gene in eight Japanese patients from seven families with erythropoietic protoporphyria. J Dermatol. 2006 Sep. 33(9):603-8. [View Abstract]
  27. Yasuda M, Chen B, Desnick RJ. Recent advances on porphyria genetics: Inheritance, penetrance & molecular heterogeneity, including new modifying/causative genes. Mol Genet Metab. 2018 Nov 30. [View Abstract]
  28. Nakano H, Nakano A, Toyomaki Y, Ohashi S, Harada K, Moritsugu R, et al. Novel ferrochelatase mutations in Japanese patients with erythropoietic protoporphyria: high frequency of the splice site modulator IVS3-48C polymorphism in the Japanese population. J Invest Dermatol. 2006 Dec. 126 (12):2717-9. [View Abstract]
  29. Risheg H, Chen FP, Bloomer JR. Genotypic determinants of phenotype in North American patients with erythropoietic protoporphyria. Mol Genet Metab. 2003 Sep-Oct. 80(1-2):196-206. [View Abstract]
  30. Lau K, Lam C. DNA-based diagnosis of erythropoietic protoporphyria in two families and the frequency of a low-expression FECH allele in a Chinese population. Clin Chim Acta. 2009. 400:132-4.
  31. 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(2):70-6. [View Abstract]
  32. Mizawa M, Makino T, Nakano H, Sawamura D, Shimizu T. Incomplete erythropoietic protoporphyria caused by a splice site modulator homozygous IVS3-48C polymorphism in the ferrochelatase gene. Br J Dermatol. 2016 Jan. 174 (1):172-5. [View Abstract]
  33. Ducamp S, Schneider-Yin X, de Rooij F, Clayton J, Fratz EJ, Rudd A. Molecular and functional analysis of the C-terminal region of human erythroid-specific 5-aminolevulinic synthase associated with X-linked dominant protoporphyria (XLDPP). Hum Mol Genet. 2013 Apr 1. 22(7):1280-8. [View Abstract]
  34. Ninomiya Y, Kokunai Y, Tanizaki H, Akasaka E, Nakano H, Moriwaki S. X-linked dominant protoporphyria: The first reported Japanese case. J Dermatol. 2015 Sep 21. 42:1-5. [View Abstract]
  35. Landefeld C, Kentouche K, Gruhn B, Stauch T, Rößler S, Schuppan D, et al. X-linked protoporphyria: Iron supplementation improves protoporphyrin overload, liver damage and anaemia. Br J Haematol. 2015 Jul 20. [View Abstract]
  36. Lecha M, Puy H, Deybach J-C. Erythropoietic protoporphyria. Orphanet Journal of Rare Diseases. 10 September 2009. 4:1-19.
  37. Poh-Fitzpatrick MB. Human protoporphyria: reduced cutaneous photosensitivity and lower erythrocyte porphyrin levels during pregnancy. J Am Acad Dermatol. 1997 Jan. 36(1):40-3. [View Abstract]
  38. Whalin S, Floderus Y, Stal P, Harper P. Erythropietic protoporphyria in Sweden: demographic, clinical, biochemical and genetic characteristics. J Intern Med. 2010. 269:278-288.
  39. Livideanu CB, Ducamp S, Lamant L, Gouya L, Rauzy OB, Deybach JC, et al. Late-onset X-linked dominant protoporphyria: an etiology of photosensitivity in the elderly. J Invest Dermatol. 2013 Jun. 133 (6):1688-90. [View Abstract]
  40. Cripps DJ, Gilbert LA, Goldfarb SS. Erythropoietic protoporphyria: juvenile protoporphyrin hepatopathy cirrhosis and death. J Pediatr. 1977 Nov. 91(5):744-8. [View Abstract]
  41. Rand EB, Bunin N, Cochran W, Ruchelli E, Olthoff KM, Bloomer JR. Sequential liver and bone marrow transplantation for treatment of erythropoietic protoporphyria. Pediatrics. 2006 Dec. 118(6):e1896-9. [View Abstract]
  42. McGuire BM, Bonkovsky HL, Carithers RL Jr, et al. Liver transplantation for erythropoietic protoporphyria liver disease. Liver Transpl. 2005 Dec. 11(12):1590-6. [View Abstract]
  43. Méndez M, Poblete-Gutiérrez P, Morán-Jiménez MJ, Rodriguez ME, Garrido-Astray MC, Fontanellas A. A homozygous mutation in the ferrochelatase gene underlies erythropoietic protoporphyria associated with palmar keratoderma. Br J Dermatol. 2009 Jun. 160(6):1330-4. [View Abstract]
  44. Holme SA, Whatley SD, Roberts AG, Anstey AV, Elder GH, Ead RD. Seasonal palmar keratoderma in erythropoietic protoporphyria indicates autosomal recessive inheritance. J Invest Dermatol. 2009 Mar. 129(3):599-605. [View Abstract]
  45. Shehade SA, Chalmers RJ, Prescott RJ. Predictable and unpredictable hazards of erythropoietic protoporphyria. Clin Exp Dermatol. 1991 May. 16(3):185-7. [View Abstract]
  46. Herbert A, Corbin D, Williams A, Thompson D, Buckels J, Elias E. Erythropoietic protoporphyria: unusual skin and neurological problems after liver transplantation. Gastroenterology. 1991 Jun. 100(6):1753-7. [View Abstract]
  47. Rank JM, Carithers R, Bloomer J. Evidence for neurological dysfunction in end-stage protoporphyric liver disease. Hepatology. 1993 Dec. 18(6):1404-9. [View Abstract]
  48. Gross U, Frank M, Doss MO. Hepatic complications of erythropoietic protoporphyria. Photodermatol Photoimmunol Photomed. 1998 Apr. 14(2):52-7. [View Abstract]
  49. Doss MO, Frank M. Hepatobiliary implications and complications in protoporphyria, a 20-year study. Clin Biochem. 1989 Jun. 22(3):223-9. [View Abstract]
  50. Holme SA, Worwood M, Anstey AV, Elder GH, Badminton MN. Erythropoiesis and iron metabolism in dominant erythropoietic protoporphyria. Blood. 2007 Dec 1. 110 (12):4108-10. [View Abstract]
  51. Mathews-Roth MM. Letter: Anemia in erythropoietic protoporphyria. JAMA. 1974 Nov 11. 230(6):824. [View Abstract]
  52. Gordeuk VR, Brittenham GM, Hawkins CW, Mukhtar H, Bickers DR. Iron therapy for hepatic dysfunction in erythropoietic protoporphyria. Ann Intern Med. 1986 Jul. 105(1):27-31. [View Abstract]
  53. Bentley DP, Meek EM. Clinical and biochemical improvement following low-dose intravenous iron therapy in a patient with erythropoietic protoporphyria. Br J Haematol. 2013 Oct. 163 (2):289-91. [View Abstract]
  54. Holme SA, Thomas CL, Whatley SD, Bentley DP, Anstey AV, Badminton MN. Symptomatic response of erythropoietic protoporphyria to iron supplementation. J Am Acad Dermatol. 2007 Jun. 56 (6):1070-2. [View Abstract]
  55. McClements BM, Bingham A, Callender ME, Trimble ER. Erythropoietic protoporphyria and iron therapy. Br J Dermatol. 1990 Mar. 122(3):423-4. [View Abstract]
  56. Milligan A, Graham-Brown RA, Sarkany I, Baker H. Erythropoietic protoporphyria exacerbated by oral iron therapy. Br J Dermatol. 1988 Jul. 119 (1):63-6. [View Abstract]
  57. Spelt JM, de Rooij FW, Wilson JH, Zandbergen AA. Vitamin D deficiency in patients with erythropoietic protoporphyria. J Inherit Metab Dis. 2010 Dec. 33 Suppl 3:S1-4. [View Abstract]
  58. Allo G, del Carmen Garrido-Astray M, De Salamanca RE, Martínez G, Hawkins F. Bone mineral density and vitamin D levels in erythropoietic protoporphyria. Endocrine. 2013 Dec. 44(3):803-7. [View Abstract]
  59. Wahlin S, Aschan J, Bjornstedt M, Broomé U, Harper P. Curative bone marrow transplantation in erythropoietic protoporphyria after reversal of severe cholestasis. J Hepatol. 2007 Jan. 46(1):174-9. [View Abstract]
  60. Wahlin S, Stal P, Adam R, Karam V, Porte R, Seehofer D. Liver transplantation for erythropoietic protoporphyria in Europe. Liver Transpl. 2011 Sep. 17(9):1021-6. [View Abstract]
  61. Wolff K, Honigsmann H, Rauschmeier W, Schuler G, Pechlaner R. Microscopic and fine structural aspects of porphyrias. Acta Derm Venereol Suppl (Stockh). 1982. 100:17-28. [View Abstract]
  62. Epstein JH, Tuffanelli DL, Epstein WL. Cutaneous changes in the porphyrias. A microscopic study. Arch Dermatol. 1973 May. 107(5):689-98. [View Abstract]
  63. Minder EI. Afamelanotide, an agonistic analog of a-melanocyte-stimulating hormone, in dermal phototoxicity of erythropoietic protoporphyria. Expert Opin Investig Drugs. 2010 Dec. 19(12):1591-602. [View Abstract]
  64. Kim ES, Garnock-Jones KP. Afamelanotide: a review in erythropoietic protoporphyria. Am J Clin Dermatol. 2016 Apr. 17 (2):179-85. [View Abstract]
  65. Minder EI, Barman-Aksoezen J, Schneider-Yin X. Pharmacokinetics and pharmacodynamics of afamelanotide and its clinical use in treating dermatologic disorders. Clin Pharmacokinet. 2017 Aug. 56 (8):815-823. [View Abstract]
  66. SCENESSE (afamelanotide) implant, for subcutaneous use [package insert]. West Menlo Park, Calif: CLINUVEL, INC. October 2019. Available at
  67. Langendonk JG, Balwani M, Anderson KE, Bonkovsky HL, Anstey AV, Bissell DM, et al. Afamelanotide for Erythropoietic Protoporphyria. N Engl J Med. 2015 Jul 2. 373 (1):48-59. [View Abstract]
  68. Johnson JA, Fusaro RM. Broad-spectrum photoprotection: the roles of tinted auto windows, sunscreens and browning agents in the diagnosis and treatment of photosensitivity. Dermatology. 1992. 185(4):237-41. [View Abstract]
  69. Roelandts R. Photo(chemo)therapy and general management of erythropoietic protoporphyria. Dermatology. 1995. 190(4):330-1. [View Abstract]
  70. Mathews-Roth MM, Pathak UA, Fitzpatrick TB, Harber LC, Kass EH. Beta-carotene as an oral photoprotective agent in erythropoietic protoporphyria. JAMA. 1974 May 20. 228(8):1004-8. [View Abstract]
  71. Mathews-Roth MM. Carotenoid functions in photoprotection and cancer prevention. J Environ Pathol Toxicol Oncol. 1990 Jul-Oct. 10(4-5):181-92. [View Abstract]
  72. Mathews-Roth MM, Rosner B. Long-term treatment of erythropoietic protoporphyria with cysteine. Photodermatol Photoimmunol Photomed. 2002 Dec. 18(6):307-9. [View Abstract]
  73. Yamamoto S, Hirano Y, Horie Y. Cimetidine reduces erythrocyte protoporphyrin in erythropoietic protoporphyria. Am J Gastroenterol. 1993 Sep. 88(9):1465-6. [View Abstract]
  74. Tu JH, Sheu SL, Teng JM. Novel Treatment Using Cimetidine for Erythropoietic Protoporphyria in Children. JAMA Dermatol. 2016 Nov 1. 152 (11):1258-1261. [View Abstract]
  75. Tishler PV, Rosner B. Treatment of erythropoietic protoporphyria with the oral sorbent colestipol: A proof-of-concept clinical trial. J Amer Acad Dermatol. 2014. 70 (2):391-2.
  76. Bloomer JR, Rank JM, Payne WD, et al. Follow-up after liver transplantation for protoporphyric liver disease. Liver Transpl Surg. 1996 Jul. 2(4):269-75. [View Abstract]
  77. McCullough AJ, Barron D, Mullen KD, et al. Fecal protoporphyrin excretion in erythropoietic protoporphyria: effect of cholestyramine and bile acid feeding. Gastroenterology. 1988 Jan. 94(1):177-81. [View Abstract]
  78. Gorchein A, Foster GR. Liver failure in protoporphyria: long-term treatment with oral charcoal. Hepatology. 1999 Mar. 29(3):995-6. [View Abstract]
  79. van Wijk HJ, van Hattum J, Baart de la Faille H, van den Berg JW, Edixhoven-Bosdijk A, Wilson JH. Blood exchange and transfusion therapy for acute cholestasis in protoporphyria. Dig Dis Sci. 1988 Dec. 33(12):1621-5. [View Abstract]
  80. Dellon ES, Szczepiorkowski ZM, Dzik WH, et al. Treatment of recurrent allograft dysfunction with intravenous hematin after liver transplantation for erythropoietic protoporphyria. Transplantation. 2002 Mar 27. 73(6):911-5. [View Abstract]
  81. Do KD, Banner BF, Katz E, Szymanski IO, Bonkovsky HL. Benefits of chronic plasmapheresis and intravenous heme-albumin in erythropoietic protoporphyria after orthotopic liver transplantation. Transplantation. 2002 Feb 15. 73(3):469-72. [View Abstract]
  82. 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 Nov. 18(3):298-309. [View Abstract]
  83. Eichbaum QG, Dzik WH, Chung RT, Szczepiorkowski ZM. Red blood cell exchange transfusion in two patients with advanced erythropoietic protoporphyria. Transfusion. 2005 Feb. 45(2):208-13. [View Abstract]
  84. Dowman JK, Gunson BK, Mirza DF, Badminton MN, Newsome PN,. UK experience of liver transplantation for erythropoietic protoporphyria. J Inherit Metab Dis. 2011 Apr. 34(2):539-45. [View Abstract]
  85. Meerman L, Verwer R, Slooff MJ, et al. Perioperative measures during liver transplantation for erythropoietic protoporphyria. Transplantation. 1994 Jan. 57(1):155-8. [View Abstract]
  86. Wahlin S, Srikanthan N, Hamre B, Harper P, Brun A. Protection from phototoxic injury during surgery and endoscopy in erythropoietic protoporphyria. Liver Transpl. 2008 Sep. 14(9):1340-6. [View Abstract]
  87. Graham-Brown MP, Ilchyshyn A. Development of acute phototoxic reaction during surgery in a patient with erythropoietic protoporphyria. Clin Exp Dermatol. 2013 Jul. 38 (5):566-8; quiz 568. [View Abstract]
  88. Morris SD, Mason NG, Elder GH, Hawk JL, Sarkany RP. Ferrochelatase gene polymorphism analysis for accurate genetic counselling in erythropoietic protoporphyria. Br J Dermatol. 2002 Sep. 147(3):572-4. [View Abstract]
  89. Schneider-Yin X, Gouya L, Meier-Weinand A, Deybach JC, Minder EI. New insights into the pathogenesis of erythropoietic protoporphyria and their impact on patient care. Eur J Pediatr. 2000 Oct. 159(10):719-25. [View Abstract]
  90. Redeker AG, Sterling RE. The "glucose effect" in erythropoietic protoporphyria. Arch Intern Med. 1968 May. 121(5):446-8. [View Abstract]
  91. Bonkovsky HL, Schned AR. Fatal liver failure in protoporphyria. Synergism between ethanol excess and the genetic defect. Gastroenterology. 1986 Jan. 90(1):191-201. [View Abstract]