Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
2.
Ital J Pediatr ; 49(1): 156, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37996925

ABSTRACT

BACKGROUND: Erythropoietic protoporphyria is a rare disorder which represents an important health problem in children, causing painful photosensitivity. Little is known on the correlation between genetic profile and clinical manifestations. The standard of care for Erythropoietic protoporphyria is based on avoiding sun and using sun protections, but recent literature has suggested that cimetidine may have a role in improving sun sensitivity. Herein we report our case series describing the successful use of cimetidine and analyzing potential phenotype-genotype correlations. CASE PRESENTATION: This case series describes five patients presented to our Rheumatology Service complaining sun sensitivity. Blood exams and genetic analysis were consistent with the diagnosis of erythropoietic protoporphyria. Four of 5 patients received cimetidine in addition to standard therapies and the effect of treatment was evaluated by Erythropoietic Protoporphyria - Quality of Life questionnaire. CONCLUSIONS: Erythropoietic protoporphyria usually manifests in early childhood after a short sun exposure. Skin manifestations are the main reason for investigations, although sometimes they can be more subtle, leading to a significant diagnostic delay. Skin diseases in children can have profound effects on their family and social relationships. A treatment with cimetidine appears to be an excellent therapeutic option in children with Erythropoietic protoporphyria.


Subject(s)
Photosensitivity Disorders , Protoporphyria, Erythropoietic , Child , Humans , Child, Preschool , Protoporphyria, Erythropoietic/diagnosis , Protoporphyria, Erythropoietic/therapy , Protoporphyria, Erythropoietic/complications , Ferrochelatase/genetics , Cimetidine , Quality of Life , Delayed Diagnosis , Photosensitivity Disorders/etiology
3.
J Pediatr Hematol Oncol ; 45(7): 416-422, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37539993

ABSTRACT

BACKGROUND: Erythropoietic protoporphyria (EPP) is a rare inherited disease of heme biosynthesis resulting in the accumulation of protoporphyrin, characterized by liver failure in a minority of cases. Although liver transplant (LT) is the therapeutic strategy for advanced hepatic disease, it does not correct the primary defect, which leads to recurrence in liver graft. Thus, hematopoietic stem cell transplantation (HSCT) is an approach for treating EPP. METHODS: We aim to describe the first sequential LT and HSCT for EPP performed in Latin America, besides reviewing the present-day literature. RESULTS: The patient, a 13-year-old female with a history of photosensitivity, presented with symptoms of cholestatic and hepatopulmonary syndrome and was diagnosed with EPP. Liver biopsy demonstrated cirrhosis. She was submitted to a successful LT and showed improvement of respiratory symptoms. However, she had disease recurrence on the liver graft. She underwent a myeloablative HSCT using a matched unrelated donor, conditioning with BuCy (busulfan and cyclophosphamide), and GvHD (graft vs. host disease) prophylaxis with ATG (thymoglobulin), tacrolimus and methotrexate. Neutrophil engraftment occurred on D+18. She has presented mixed chimerism, but normalization of PP levels, being 300 days after HSCT, in good state of health and normal liver function. CONCLUSIONS: Consecutive LT and HSCT for EPP is a procedure that has been described in 10 cases in the literature and, even though these patients are a highly diversified population, studies have shown favorable results. This concept of treatment should be considered in patients with established liver disease.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Liver Diseases , Liver Transplantation , Protoporphyria, Erythropoietic , Female , Humans , Adolescent , Bone Marrow Transplantation , Protoporphyria, Erythropoietic/therapy , Protoporphyria, Erythropoietic/pathology , Hematopoietic Stem Cell Transplantation/methods , Liver Transplantation/methods , Transplantation Conditioning
4.
Pharmacol Ther ; 248: 108487, 2023 08.
Article in English | MEDLINE | ID: mdl-37392940

ABSTRACT

Protoporphyrin IX (PPIX) is an intermediate in the heme biosynthesis pathway. Abnormal accumulation of PPIX due to certain pathological conditions such as erythropoietic protoporphyria and X-linked protoporphyria causes painful phototoxic reactions of the skin, which can significantly impact daily life. Endothelial cells in the skin have been proposed as the primary target for PPIX-induced phototoxicity through light-triggered generation of reactive oxygen species. Current approaches for the management of PPIX-induced phototoxicity include opaque clothing, sunscreens, phototherapy, blood therapy, antioxidants, bone marrow transplantation, and drugs that increase skin pigmentation. In this review, we discuss the present understanding of PPIX-induced phototoxicity including PPIX production and disposition, conditions that lead to PPIX accumulation, symptoms and individual differences, mechanisms, and therapeutics.


Subject(s)
Endothelial Cells , Protoporphyria, Erythropoietic , Humans , Endothelial Cells/metabolism , Protoporphyrins/pharmacology , Protoporphyrins/metabolism , Protoporphyria, Erythropoietic/metabolism , Protoporphyria, Erythropoietic/pathology , Protoporphyria, Erythropoietic/therapy , 5-Aminolevulinate Synthetase
5.
Exp Dermatol ; 32(7): 1042-1047, 2023 07.
Article in English | MEDLINE | ID: mdl-37052136

ABSTRACT

Severe skin pain when exposed to long wave ultraviolet radiation or visible light is the main symptom of erythropoietic protoporphyria (EPP). Treatment options for EPP are inadequate and new treatments are needed but hampered by the lack of valid efficacy outcomes. Phototesting with well-defined illumination of the skin can be performed reliably. We aimed to provide an overview of phototest procedures used to evaluate EPP treatments. Systematic searches of Embase, MEDLINE and the Cochrane Library were performed. Searches identified 11 studies using photosensitivity as efficacy outcome. The studies used eight different phototest protocols. Illuminations were performed with a filtered high-pressure mercury arc, or a xenon arc lamp equipped with monochromator or filters. Some used broadband, others narrowband illumination. In all protocols phototests were performed on the hands or the back. Endpoints were minimal dose required to induce either first symptom of discomfort, erythema, urticaria or intolerable pain. Other endpoints were change in erythema intensity or diameter of any type of flare after exposure compared to before. In conclusion, protocols displayed extensive variability in illumination set-up and evaluation of phototest reactions. Implementation of a standardized phototest method will allow more consistent and reliable outcome evaluation in future therapeutic research of protoporphyric photosensitivity.


Subject(s)
Photosensitivity Disorders , Protoporphyria, Erythropoietic , Humans , Protoporphyria, Erythropoietic/therapy , Ultraviolet Rays , Skin , Erythema
6.
Blood ; 141(24): 2921-2931, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36898083

ABSTRACT

Erythropoietic protoporphyria (EPP) is an inherited cutaneous porphyria caused by reduced expression of ferrochelatase, the enzyme that catalyzes the final step in heme biosynthesis. The resultant accumulation of protoporphyrin IX leads to severe, painful cutaneous photosensitivity, as well as potentially life-threatening liver disease in a small percentage of patients. X-linked protoporphyria (XLP) is clinically similar to EPP but results from increased activity of δ-aminolevulinic acid synthase 2, the first step in heme biosynthesis in the bone marrow, and also causes protoporphyrin accumulation. Although historically the management of EPP and XLP (collectively termed protoporphyria) centered around avoidance of sunlight, novel therapies have recently been approved or are in development, which will alter the therapeutic landscape for these conditions. We present 3 patient cases, highlighting key treatment considerations in patients with protoporphyria, including (1) approach to photosensitivity, (2) managing iron deficiency in protoporphyria, and (3) understanding hepatic failure in protoporphyria.


Subject(s)
Liver Diseases , Photosensitivity Disorders , Protoporphyria, Erythropoietic , Humans , Protoporphyria, Erythropoietic/therapy , Protoporphyria, Erythropoietic/complications , Ferrochelatase/genetics , Ferrochelatase/metabolism , Photosensitivity Disorders/etiology , Photosensitivity Disorders/therapy , Protoporphyrins , Heme/metabolism
7.
J Am Acad Dermatol ; 89(6): 1227-1237, 2023 12.
Article in English | MEDLINE | ID: mdl-36041558

ABSTRACT

Erythropoietic protoporphyria and X-linked protoporphyria are rare genetic photodermatoses. Limited expertise with these disorders among physicians leads to diagnostic delays. Here, we present evidence-based consensus guidelines for the diagnosis, monitoring, and management of erythropoietic protoporphyria and X-linked protoporphyria. A systematic literature review was conducted, and reviewed among subcommittees of experts, divided by topic. Consensus on guidelines was reached within each subcommittee and then among all members of the committee. The appropriate biochemical and genetic testing to establish the diagnosis is reviewed in addition to the interpretation of results. Prevention of symptoms, management of acute phototoxicity, and pharmacologic and nonpharmacologic treatment options are discussed. The importance of ongoing monitoring for liver disease, iron deficiency, and vitamin D deficiency is discussed with management guidance. Finally, management of pregnancy and surgery and the safety of other therapies are summarized. We emphasize that these are multisystemic disorders that require longitudinal monitoring. These guidelines provide a structure for evidence-based diagnosis and management for practicing physicians. Early diagnosis and management of these disorders are essential, particularly given the availability of new and emerging therapies.


Subject(s)
Dermatitis, Phototoxic , Genetic Diseases, X-Linked , Liver Diseases , Practice Guidelines as Topic , Protoporphyria, Erythropoietic , Humans , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/therapy , Genetic Diseases, X-Linked/genetics , Protoporphyria, Erythropoietic/diagnosis , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy
8.
Mol Genet Metab ; 135(3): 215-220, 2022 03.
Article in English | MEDLINE | ID: mdl-35034844

ABSTRACT

BACKGROUND: Erythropoietic protoporphyria (EPP) patients suffer from painful phototoxicity. Sunlight-avoiding behaviour has not yet been quantified objectively in EPP patients. OBJECTIVE: To study total white light exposure obtained with an actigraph device, before and during afamelanotide treatment, in EPP patients compared to healthy controls. Effects on circadian rhythm, pain and sleep were also investigated. METHODS: Adult EPP patients visiting the Porphyria Center Rotterdam of the Erasmus MC were included in this single-center longitudinal case-control open-label intervention study. Controls were age and place of residence matched. Participants wore an actigraph (Actiwatch Pro) during two weeks for multiple periods. Afamelanotide was given to EPP patients as part of standard care. RESULTS: Twenty-six EPP patients and 23 matched controls participated. Controls were statistically significantly more exposed to white light than EPP patients off treatment during autumn (95.4%), spring (69.9%), and summer (105.4%; p = 0.01). EPP patients on afamelanotide treatment had 71.6% more light exposure during spring compared to EPP patients off treatment (p < 0.01). Afamelanotide treatment resulted in a reduction of painful moments in the morning (6.5% decrease) and the evening (8.1% decrease; p < 0.05). Bedtime differed between EPP patients off treatment, controls and EPP patients on treatment (23:45 h ± 1:51 versus 23:02 ± 1:41 and 23:14 ± 1:29, respectively; p < 0.0001). CONCLUSION: Actigraphy is a useful method to objectively measure white light exposure and treatment effects in EPP. In EPP patients afamelanotide treatment is associated with increased white light exposure during spring, and overall less pain. Treatment with afamelanotide is also associated with normalization of circadian rhythm.


Subject(s)
Dermatitis, Phototoxic , Protoporphyria, Erythropoietic , Adult , Case-Control Studies , Circadian Rhythm , Humans , Pain/drug therapy , Protoporphyria, Erythropoietic/therapy
10.
Pediatr Transplant ; 25(6): e13966, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33405342

ABSTRACT

BACKGROUND: EPP is a rare disorder of heme biosynthesis in which patients present with disabling photosensitivity. A subset of patients develop severe liver disease with progressive liver failure necessitating an OLT. A HCT can potentially cure EPP by replacing the native bone marrow, which is the primary site of heme synthesis. However, due to concerns for inherent risks of treatment-related toxicities, the use of HCT has been reserved for patients undergoing an OLT to avoid disease recurrence in the hepatic graft. Data for HCT in EPP are lacking, particularly in the pediatric population. CASE (METHODS/RESULTS): We present the case of a 12-year-old patient with EPP photosensitivity and cirrhosis, whom we successfully treated with pre-emptive allogeneic HCT, significantly improving the patient's quality of life. We used a matched-unrelated donor bone marrow-derived graft. Our patient achieved full donor peripheral blood chimerism and has not had any evidence of GVHD. In addition to resolution of photosensitivity, our patient had reversal of liver fibrosis which we feel was largely due to intervention at an early stage of compensated cirrhosis. CONCLUSION: Our case highlights the successful application of a known RIC regimen to this rare disorder that was well tolerated with sustained donor engraftment. It also emphasizes the importance of timing for HCT in patients with EPP and liver fibrosis. HCT should be considered early in pediatric patients with EPP-hepatopathy to prevent progression to liver failure and need for OLT with lifelong immunosuppression.


Subject(s)
Hematopoietic Stem Cell Transplantation , Liver Cirrhosis/surgery , Protoporphyria, Erythropoietic/therapy , Child , Humans , Protoporphyria, Erythropoietic/genetics , Transplantation Conditioning
11.
Hematology Am Soc Hematol Educ Program ; 2020(1): 400-410, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33275677

ABSTRACT

The porphyrias are a family of metabolic disorders caused by defects in the activity of one of the enzymes in the heme biosynthetic pathway. Acute intermittent porphyria (AIP), caused by autosomal dominant mutations in the gene encoding hydroxymethylbilane synthase, can lead to hepatocyte overaccumulation and systemic distribution of the proximal porphyrin precursors, 5-aminolevulinic acid (ALA) and porphobilinogen (PBG). ALA and PBG are toxic to neurons and extrahepatic tissue and cause the neurovisceral clinical manifestations of AIP. Management of AIP includes awareness and avoidance of triggering factors, infusions of hemin for severe acute attacks, and, if indicated for chronic suppressive therapy, maintenance treatment with hemin or givosiran, a small interfering RNA molecule that antagonizes ALA synthase 1 transcripts. Erythropoietic protoporphyria (EPP) is most commonly caused by autosomal recessive mutations in the gene encoding ferrochelatase (FECH), the heme pathway terminal enzyme. FECH deficiency leads to erythrocyte overaccumulation and high plasma levels of lipophilic protoporphyrins that photoactivate in the skin, causing burning pain and erythema. Protoporphyrins excreted in the bile can cause gallstones, cholestasis, fibrosis, and ultimately liver failure. Management of EPP includes skin protection and afamelanotide, an α-melanocyte stimulating hormone analog that increases melanin pigment and reduces photoactivation. Liver transplantation may be necessary for severe EPP-induced liver complications. Because AIP and EPP arise from defects in the heme biosynthetic pathway, hematologists are often consulted to evaluate and manage suspected or proven porphyrias. A working knowledge of these disorders increases our confidence and effectiveness as consultants and medical providers.


Subject(s)
Porphyria, Acute Intermittent/diagnosis , Protoporphyria, Erythropoietic/diagnosis , Adult , Disease Management , Female , Heme/genetics , Humans , Mutation , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/pathology , Porphyria, Acute Intermittent/therapy , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/pathology , Protoporphyria, Erythropoietic/therapy , Young Adult
12.
Nucleic Acids Res ; 48(9): 4658-4671, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32313951

ABSTRACT

Erythropoietic protoporphyria (EPP) is a rare genetic disease in which patients experience acute phototoxic reactions after sunlight exposure. It is caused by a deficiency in ferrochelatase (FECH) in the heme biosynthesis pathway. Most patients exhibit a loss-of-function mutation in trans to an allele bearing a SNP that favors aberrant splicing of transcripts. One viable strategy for EPP is to deploy splice-switching oligonucleotides (SSOs) to increase FECH synthesis, whereby an increase of a few percent would provide therapeutic benefit. However, successful application of SSOs in bone marrow cells is not described. Here, we show that SSOs comprising methoxyethyl-chemistry increase FECH levels in cells. We conjugated one SSO to three prototypical targeting groups and administered them to a mouse model of EPP in order to study their biodistribution, their metabolic stability and their FECH splice-switching ability. The SSOs exhibited distinct distribution profiles, with increased accumulation in liver, kidney, bone marrow and lung. However, they also underwent substantial metabolism, mainly at their linker groups. An SSO bearing a cholesteryl group increased levels of correctly spliced FECH transcript by 80% in the bone marrow. The results provide a promising approach to treat EPP and other disorders originating from splicing dysregulation in the bone marrow.


Subject(s)
Ferrochelatase/genetics , Oligonucleotides/administration & dosage , Protoporphyria, Erythropoietic/metabolism , RNA Splicing , Albumins/metabolism , Animals , Bone Marrow/metabolism , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Ferrochelatase/metabolism , Humans , K562 Cells , Mice , Oligonucleotides/blood , Oligonucleotides/chemistry , Oligonucleotides/pharmacokinetics , Polymorphism, Single Nucleotide , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy , RNA Splice Sites , Tissue Distribution
13.
Photodiagnosis Photodyn Ther ; 29: 101582, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31809913

ABSTRACT

BACKGROUND: Erythropoietic protoporphyria (EPP) is a rare, genetic disease with reduced ferrochelatase activity causing protoporphyrine IX (PpIX) to accumulate in erythrocytes. PpIX activation by daylight causes skin erythema, edema, burning, and stinging. No treatment exists to reduce PpIX. AIM: To introduce a method that reduces PpIX in erythrocytes to relieve skin symptoms in patients with EPP. METHOD: A case series of 7 patients with EPP constituted this explorative study. Erythrocyte PpIX was inactivated by illuminating the patients' heparinized blood outside their body, then returning it to the patient. About 3 litres of blood was illuminated with 630 nm light, 20 J/cm2. The effect was measured as a reduction in erythrocyte PpIX. The patients reported the number of minutes in daylight tolerated before and after intervention. RESULTS: This procedure reduced PpIX by about 30 % and daylight tolerance was, on average, increased by 14 times. The subsequently excreted photoproducts resulted in some liver toxicity. Three treatments during spring and early summer were sufficient to reduce the patients' symptoms throughout the year in Northern Europe. CONCLUSION: Extracorporeal erythrocyte photodynamic therapy is the first treatment to successfully reduce the amount of PpIX in the blood of EPP patients, thus "normalizing" their daylight tolerance.


Subject(s)
Extracorporeal Circulation/methods , Photochemotherapy/methods , Protoporphyria, Erythropoietic/blood , Protoporphyria, Erythropoietic/therapy , Protoporphyrins/metabolism , Adult , Female , Humans , Male , Middle Aged
14.
Ann Hematol ; 99(2): 385-388, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31773213
15.
Liver Transpl ; 25(11): 1620-1633, 2019 11.
Article in English | MEDLINE | ID: mdl-31469227

ABSTRACT

Erythropoietic protoporphyria (EPP) is an inherited metabolic disorder of heme synthesis resulting from overproduction of protoporphyrin IX (PPIX), which can lead to progressive liver disease characterized by recurrent EPP crises and end-stage liver disease. We used the Australian Transplant Registry to identify 5 patients referred for liver transplantation between 2008 and 2017. A total of 4 patients had EPP secondary to ferrochelatase deficiency, and 1 patient had X-linked EPP. No patient had follow-up with a specialist prior to the diagnosis of progressive liver disease. There were 3 patients who underwent orthotopic liver transplantation, whereas 2 died while on the transplant waiting list. Parenteral PPIX-lowering therapy was used in 4 patients and was effective in 3 patients, although 2 of these had rebound porphyria and worsening liver function following a decrease in the intensity of therapy. Early disease recurrence in the allograft following transplantation occurred in 2 patients requiring red cell exchange (RCE) to successfully attain and maintain low PPIX levels, but RCE was associated with hemosiderosis in 1 patient. Allogeneic stem cell transplantation (AlloSCT) was performed in 2 patients. One failed engraftment twice, whereas the second rejected the first graft but achieved full donor chimerism with a second graft and increased immunosuppression. In conclusion, our observations suggest that progressive liver disease needs parenteral PPIX-lowering treatment with the intensity adjusted to achieve a target Erc-PPIX level. Because EPP liver disease is universally recurrent, AlloSCT should be considered in all patients with adequate immunosuppression to facilitate engraftment. RCE appears to be effective for recurrent EPP liver disease but is associated with an increased risk of iron overload.


Subject(s)
End Stage Liver Disease/therapy , Graft Rejection/epidemiology , Liver Transplantation , Protoporphyria, Erythropoietic/pathology , Stem Cell Transplantation , Waiting Lists/mortality , Adolescent , Adult , Allografts/pathology , Disease Progression , End Stage Liver Disease/mortality , End Stage Liver Disease/pathology , Female , Graft Rejection/pathology , Humans , Infant , Liver/pathology , Male , Middle Aged , Protoporphyria, Erythropoietic/mortality , Protoporphyria, Erythropoietic/therapy , Recurrence , Registries/statistics & numerical data , Transplantation, Homologous , Young Adult
17.
Mol Genet Metab ; 128(3): 304-308, 2019 11.
Article in English | MEDLINE | ID: mdl-31076252

ABSTRACT

Deficiency in ferrochelatase (FECH), the last enzyme in the heme biosynthetic pathway, leads to an accumulation of protoporphyrin IX (PPIX) that causes a severely painful phototoxic reaction of the skin in patients with erythropoietic protoporphyria (EPP). Besides phototoxicity of the skin, EPP patients often present with symptoms of iron deficiency in form of a microcytic and hypochromic anemia with low serum iron and ferritin. In addition, elevated aminolevulinic acid synthase 2 (ALAS2) both at the mRNA and protein levels have been observed among EPP patients. ALAS is the first enzyme in the pathway and exists in two isoforms, whereby the isoform 2 (ALAS2) is expressed exclusively in erythropoiesis. The mRNA of ALAS2 contains an iron response element (IRE) at its 5'UTR. When iron is limited, iron response element binding protein 2 (IRP2) binds to the IRE of ALAS2 mRNA and suppresses its translation. In this study, we demonstrated that iron deprivation increased the amount of ALAS2 mRNA as well as the ratio of ALAS2 to FECH mRNAs in cultured erythroleukemic K562 cells. At the protein level, however, iron deprivation in the cell line caused reductions in both enzymes as shown by the Western blot analysis. A comparable increase in the ratio of ALAS2 to FECH mRNAs was also found in EPP patients indicating an imbalance in heme biosynthesis. As iron cannot be completely missing from an organism, we assume that in EPP patients, a certain amount of ALAS2 mRNA is translated despite a partial deficiency of FECH. The increase in ALAS2 enzyme contributes to the accumulation in PPIX in the patients. Targeted inhibition of ALAS2 could therefore be a treatment option for EPP.


Subject(s)
5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Iron/metabolism , Protoporphyria, Erythropoietic/enzymology , Biosynthetic Pathways , Ferrochelatase/genetics , Humans , Iron/blood , Iron Regulatory Protein 2/metabolism , Iron-Regulatory Proteins/metabolism , K562 Cells , Protoporphyria, Erythropoietic/therapy , Protoporphyrins/metabolism
18.
Mol Genet Metab ; 128(3): 298-303, 2019 11.
Article in English | MEDLINE | ID: mdl-30704898

ABSTRACT

Erythropoietic Protoporphyria (EPP) and X-linked Protoporphyria (XLP) are rare, genetic photodermatoses resulting from defects in enzymes of the heme-biosynthetic pathway. EPP results from the partial deficiency of ferrochelatase, and XLP results from gain-of-function mutations in erythroid specific ALAS2. Both disorders result in the accumulation of erythrocyte protoporphyrin, which is released in the plasma and taken up by the liver and vascular endothelium. The accumulated protoporphyrin is activated by sunlight exposure, generating singlet oxygen radical reactions leading to tissue damage and excruciating pain. About 2-5% of patients develop clinically significant liver dysfunction due to protoporphyrin deposition in bile and/or hepatocytes which can advance to cholestatic liver failure requiring transplantation. Clinically these patients present with acute, severe, non-blistering phototoxicity within minutes of sun-exposure. Anemia is seen in about 47% of patients and about 27% of patients will develop abnormal serum aminotransferases. The diagnosis of EPP and XLP is made by detection of markedly increased erythrocyte protoporphyrin levels with a predominance of metal-free protoporphyrin. Genetic testing by sequencing the FECH or ALAS2 gene confirms the diagnosis. Treatment is limited to sun-protection and there are no currently available FDA-approved therapies for these disorders. Afamelanotide, a synthetic analogue of α-melanocyte stimulating hormone was found to increase pain-free sun exposure and improve quality of life in adults with EPP. It has been approved for use in the European Union since 2014 and is not available in the U.S. In addition to the development of effective therapeutics, future studies are needed to establish the role of iron and the risks related to the development of hepatopathy in these patients.


Subject(s)
Disease Management , Genes, X-Linked , Porphyrias, Hepatic/genetics , Porphyrias, Hepatic/physiopathology , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/physiopathology , 5-Aminolevulinate Synthetase/genetics , Anemia/etiology , Clinical Trials as Topic , Dermatitis, Phototoxic , Heme/metabolism , Humans , Liver Diseases/etiology , Liver Diseases/physiopathology , Porphyrias, Hepatic/complications , Porphyrias, Hepatic/therapy , Protoporphyria, Erythropoietic/complications , Protoporphyria, Erythropoietic/therapy
19.
Ann Dermatol Venereol ; 146(2): 143-159, 2019 Feb.
Article in French | MEDLINE | ID: mdl-30709634

ABSTRACT

The porphyrias are a group of metabolic disorders resulting from an innate abnormality in haem biosynthesis, and the clinical settings of which vary according to the genetic enzyme abnormality in question. These are genetic disorders with autosomal dominant or recessive inheritance of varying penetrance, and whose clinical expression differs according to the preferential location of haem precursors. Different classifications have been proposed according to genetic inheritance, the enzyme anomaly at issue, and clinical expression. The clinical classification distinguishes between acute porphyria (acute intermittent porphyria, porphyria variegata, hereditary coproporphyria), bullous cutaneous porphyrias (porphyria cutanea tarda, porphyria variegata and hereditary coproporphyria), painful photosensitive acute cutaneous porphyrias (erythropoietic protoporphyria and X-linked dominant protoporphyria), and rare recessive porphyrias (congenital erythropoietic porphyria, Doss porphyria, hepatoerythropoietic porphyria and harderoporphyria). Treatment depends on the clinical expression of the disorder.


Subject(s)
Porphyrias , Skin Diseases, Metabolic , Biopsy , Coproporphyria, Hereditary/diagnosis , Coproporphyria, Hereditary/genetics , Coproporphyria, Hereditary/therapy , Diagnosis, Differential , Heme/biosynthesis , Humans , Photosensitivity Disorders/complications , Photosensitivity Disorders/diagnosis , Photosensitivity Disorders/therapy , Porphyria Cutanea Tarda/diagnosis , Porphyria Cutanea Tarda/genetics , Porphyria Cutanea Tarda/therapy , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/therapy , Porphyrias/classification , Porphyrias/diagnosis , Porphyrias/genetics , Porphyrias/therapy , Protoporphyria, Erythropoietic/diagnosis , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy , Skin/pathology , Skin Diseases, Metabolic/classification , Skin Diseases, Metabolic/diagnosis , Skin Diseases, Metabolic/genetics , Skin Diseases, Metabolic/therapy
20.
Am J Hum Genet ; 104(2): 341-347, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30712775

ABSTRACT

Erythropoietic protoporphyria (EPP) is a hereditary disease characterized by a deficiency in ferrochelatase (FECH) activity. FECH activity is responsible for the accumulation of protoporphyrin IX (PPIX). Without etiopathogenic treatment, EPP manifests as severe photosensitivity. 95% of affected individuals present a hypomorphic FECH allele trans to a loss-of-function (LOF) FECH mutation, resulting in a reduction in FECH activity in erythroblasts below a critical threshold. The hypomorphic allele promotes the use of a cryptic acceptor splice site, generating an aberrant FECH mRNA, which is responsible for the reduced level of wild-type FECH mRNA and, ultimately, FECH activity. We have previously identified an antisense oligonucleotide (AON), AON-V1 (V1), that redirects splicing to the physiological acceptor site and reduces the accumulation of PPIX. Here, we developed a specific strategy that uses transferrin receptor 1 (TRF1) as a Trojan horse to deliver V1 to erythroid progenitors. We designed a bifunctional peptide (P1-9R) including a TFR1-targeting peptide coupled to a nine-arginine cell-penetrating peptide (CPP) that facilitates the release of the AON from TFR1 in endosomal vesicles. We demonstrated that the P1-9R/V1 nanocomplex promotes the efficient and prolonged redirection of splicing towards the physiological splice site and subsequent normalization of WT FECH mRNA and protein levels. Finally, the P1-9R/V1 nanocomplex increases WT FECH mRNA production and significantly decreases PPIX accumulation in primary cultures of differentiating erythroid progenitors from an overt EPP-affected individual. P1-9R is a method designed to target erythroid progenitors and represents a potentially powerful tool for the in vivo delivery of therapeutic DNA in many erythroid disorders.


Subject(s)
Antigens, CD/metabolism , Cell-Penetrating Peptides/metabolism , Erythroid Precursor Cells/metabolism , Genetic Therapy/methods , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy , Receptors, Transferrin/metabolism , Antigens, CD/administration & dosage , Antigens, CD34/metabolism , Cell Line , Cell-Penetrating Peptides/administration & dosage , Erythroblasts/cytology , Erythroblasts/metabolism , Ferrochelatase/genetics , Ferrochelatase/metabolism , Humans , Ligands , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protoporphyrins/metabolism , RNA, Messenger , Receptors, Transferrin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...