The acute hepatic porphyrias (AHPs) are inherited disorders of heme biosynthesis characterized by life-threatening acute neurovisceral attacks precipitated by factors that upregulate hepatic 5-aminolevulinic acid synthase 1 (ALAS1) activity. Induction of hepatic ALAS1 leads to the accumulation of porphyrin precursors, in particular 5-aminolevulinic acid (ALA), which is thought to be the neurotoxic mediator leading to acute attack symptoms such as severe abdominal pain and autonomic dysfunction. Patients may also develop debilitating chronic symptoms and long-term medical complications, including kidney disease and an increased risk of hepatocellular carcinoma. Exogenous heme is the historical treatment for attacks and exerts its therapeutic effect by inhibiting hepatic ALAS1 activity. The pathophysiology of acute attacks provided the rationale to develop an RNA interference therapeutic that suppresses hepatic ALAS1 expression. Givosiran is a subcutaneously administered N-acetylgalactosamine–conjugated small interfering RNA against ALAS1 that is taken up nearly exclusively by hepatocytes via the asialoglycoprotein receptor. Clinical trials established that the continuous suppression of hepatic ALAS1 mRNA via monthly givosiran administration effectively reduced urinary ALA and porphobilinogen levels and acute attack rates and improved quality of life. Common side effects include injection site reactions and increases in liver enzymes and creatinine. Givosiran was approved by the US Food and Drug Administration and European Medicines Agency in 2019 and 2020, respectively, for the treatment of patients with AHP. Although givosiran has the potential to decrease the risk of chronic complications, long-term data on the safety and effects of sustained ALAS1 suppression in patients with AHP are lacking.

The porphyrias are a group of metabolic diseases, each caused by a defect in a specific enzyme in the heme biosynthetic pathway, resulting in the accumulation of pathway intermediates. Disease phenotype, whether neurologic and/or photocutaneous symptoms, is determined by the site and composition of heme intermediates that accumulate. Porphyrias are classified pathophysiologically as either hepatic or erythropoietic based on the primary site of overproduction and accumulation of heme pathway intermediates.1 The acute hepatic porphyrias (AHPs) include 3 autosomal dominant disorders (acute intermittent porphyria [AIP], hereditary coproporphyria, and variegate porphyria) and the ultrarare autosomal recessive disorder, 5-aminolevulinic acid dehydrogenase deficiency porphyria (ADP), although the relevant sites of heme intermediate overproduction in ADP remain unclear.2,3 The enzyme deficiencies in each AHP are shown in Figure 1.

Figure 1.

Hepatic heme biosynthetic pathway and associated AHPs. Heme biosynthesis involves the conversion of glycine and succinyl CoA into heme via 8 enzymatic steps (enzymes are shown in italics). The first and rate-limiting enzyme, 5-aminolevulinic acid synthase 1 (ALAS1), uses pyridoxal 5-phosphate (or active vitamin B6) as a cofactor to condense glycine and succinyl CoA into 5-aminolevulinic acid (ALA). Two molecules of ALA are then condensed to form the monopyrrole porphobilinogen (PBG) through the action of the second enzyme, 5-aminolevulinic acid dehydratase. ALA and PBG are porphyrin precursors, and ALA in particular is believed to be neurotoxic. The third enzyme, hydroxymethylbilane synthase (HMBS), converts PBG to the linear tetrapyrrole, hydroxymethylbilane, which is then converted to the circular tetrapyrrole, uroporphyrinogen III, via uroporphyrinogen synthase. The subsequent 2 enzymatic reactions modify the side chains of the porphyrinogens, whereas the penultimate enzyme, protoporphyrinogen oxidase, oxidizes protoporphyrinogen IX to protoporphyrin IX. Finally, ferrochelatase inserts ferrous iron into protoporphyrin IX to form heme. In hepatocytes, heme synthesis is controlled by ALAS1 activity, which is regulated by the end-product, heme. Under heme-replete conditions, heme represses ALAS1 activity, whereas under heme-deficient conditions, this negative feedback inhibition is lost and heme synthesis is induced. The AHPs, shown in red, include 4 disorders that are caused by partial deficiencies of the corresponding heme biosynthetic enzymes.

Figure 1.

Hepatic heme biosynthetic pathway and associated AHPs. Heme biosynthesis involves the conversion of glycine and succinyl CoA into heme via 8 enzymatic steps (enzymes are shown in italics). The first and rate-limiting enzyme, 5-aminolevulinic acid synthase 1 (ALAS1), uses pyridoxal 5-phosphate (or active vitamin B6) as a cofactor to condense glycine and succinyl CoA into 5-aminolevulinic acid (ALA). Two molecules of ALA are then condensed to form the monopyrrole porphobilinogen (PBG) through the action of the second enzyme, 5-aminolevulinic acid dehydratase. ALA and PBG are porphyrin precursors, and ALA in particular is believed to be neurotoxic. The third enzyme, hydroxymethylbilane synthase (HMBS), converts PBG to the linear tetrapyrrole, hydroxymethylbilane, which is then converted to the circular tetrapyrrole, uroporphyrinogen III, via uroporphyrinogen synthase. The subsequent 2 enzymatic reactions modify the side chains of the porphyrinogens, whereas the penultimate enzyme, protoporphyrinogen oxidase, oxidizes protoporphyrinogen IX to protoporphyrin IX. Finally, ferrochelatase inserts ferrous iron into protoporphyrin IX to form heme. In hepatocytes, heme synthesis is controlled by ALAS1 activity, which is regulated by the end-product, heme. Under heme-replete conditions, heme represses ALAS1 activity, whereas under heme-deficient conditions, this negative feedback inhibition is lost and heme synthesis is induced. The AHPs, shown in red, include 4 disorders that are caused by partial deficiencies of the corresponding heme biosynthetic enzymes.

Close modal

Patients with AHP present with episodic attacks of severe abdominal pain and acute autonomic and other neurologic dysfunction (eg, seizures and posterior reversible encephalopathy).4 Most of the symptomatic patients are female, with disease onset after puberty.5-7 Various factors precipitate the acute attacks by increasing the expression of hepatic 5-aminolevulinic acid synthase 1 (ALAS1), either by directly promoting its transcription (eg, dieting or exogenous/endogenous progesterone8-11) and/or by consuming heme and increasing heme demand in the liver12 (eg, cytochrome p450-inducing drugs or exogenous/endogenous progesterone13,14). When ALAS1 activity is markedly increased, the respective enzyme deficiencies create a metabolic bottleneck, leading to insufficient hepatic heme production and in turn loss of the negative feedback inhibition of heme on ALAS1.15 Consequently, ALAS1 activity is further upregulated, and the porphyrin precursors, ALA and porphobilinogen (PBG), accumulate in the liver. Several hypotheses have been proposed to explain the neurologic dysfunction in the AHPs.12 Porphyrin precursors, ALA in particular, are likely neurotoxic,16-18 and their hepatic accumulation19,20 is thought to be a major mediator of neurotoxicity in acute attacks. Symptomatic improvement after an acute attack correlates with decreased ALA levels.21 Some patients demonstrate persistent elevations in ALA and PBG after an attack. The pathophysiologic implication of this finding to the neurologic complications and why some patients develop chronic neurologic symptoms remain open questions. In variegate porphyria and hereditary coproporphyria, porphyrins (in addition to porphyrin precursors) accumulate, which are photosensitizers and account for the cutaneous photosensitivity of these 2 AHP subtypes.

Disease penetrance in the AHPs is low, with most of the genetically diagnosed patients never experiencing an acute attack. AIP, the most common AHP, is caused by heterozygous pathogenic variants in hydroxymethylbilane synthase (HMBS), and the prevalence of pathogenic HMBS variants is ∼1/1700 Caucasians22,23 while the estimated disease penetrance is ∼1% in the general population. The disease penetrance is notably higher, ∼20% to 40% in families with symptomatic AIP heterozygotes, suggesting that genetic modifiers and/or environmental factors modulate susceptibility to acute attacks.23-25 Most symptomatic patients have only a few attacks in their lifetime, however ∼5% of symptomatic patients with AIP experience recurrent attacks.26,27 Importantly, patients with AIP with recurrent attacks (≥4 attacks/year) commonly report chronic symptoms between attacks.5,28 A contemporary natural history study of mostly patients with AIP with recurrent attacks found that ≥80% of patients reported chronic symptoms when not having an attack (eg, pain, mood/sleep disturbances, gastrointestinal/bladder symptoms) and high healthcare use.29,30 Hepatic ALAS1 mRNA and ALA and PBG levels often remain elevated between attacks, particularly in AIP, suggesting that a component of the chronic symptoms suffered by patients may reflect active disease.30 Moreover, AHPs are associated with long-term coexisting illnesses, including chronic kidney disease, hypertension, chronic neuropathy, and liver disease, which may manifest as elevated aminotransferase levels, fibrosis, cirrhosis, and/or hepatocellular carcinoma.5,31 

Treatment of an AHP attack includes addressing symptoms and complications of the disease (eg, pain management), eliminating known triggers that increase hepatic ALAS1 activity and precipitate or worsen an attack, and intravenous carbohydrate (glucose) and hemin loading.32,33 Hemin (heme arginate in Europe and lyophilized hematin in the United States) is thought to act by repressing hepatic ALAS1 activity and hence decreasing the overproduction of ALA and PBG. Treatment reduces urinary ALA and PBG levels and improves symptoms in most patients after 3 to 4 daily infusions.34-40 Common side effects of hemin include infusion-site phlebitis, partially ameliorated by reconstituting the drug in albumin rather than sterile water,41 and iron overload after multiple treatments42 (100 mg of hemin contains ∼8 mg of elemental iron43).

Management of patients suffering from recurrent attacks is challenging. Case series support some benefit from off-label prophylactic or “on demand” infusions of hemin in the outpatient setting given at various dosing frequencies; however, some patients continue to suffer attacks and develop chronic pain.5,39,44 Moreover, this approach often requires central venous access, which itself carries risk. Recurrent attacks during the luteal phase of the menstrual cycle have been treated with gonadotropin-releasing hormone analogs to suppress ovulation, with a mixed response45,46 and at the expense of potential bone mineral density loss and postmenopausal symptoms. Liver transplantation is a last-resort treatment option for select patients with recurrent attacks that are refractory to treatment and lead to poor quality of life, in whom the benefit is felt to outweigh its associated risks of morbidity and mortality.47-49 RNA interference (RNAi; givosiran) against ALAS1 offers another option for these patients and is the focus of this review and the only US Food and Drug Administration (FDA)–approved preventative therapy currently available for the AHPs. Its therapeutic benefit in treating ongoing acute attacks in patients remains unexplored.

The central role of hepatic ALAS1 induction causing elevated ALA and PBG levels in the pathogenesis of the acute attacks provided the rationale to develop a RNAi therapeutic targeting hepatic ALAS1 for the prevention and treatment of acute attacks in AHPs. RNAi therapeutics leverage a naturally occurring regulatory process in which double-stranded RNAs are cleaved into short fragments known as short interfering RNAs (siRNAs), which subsequently bind to mRNAs in a sequence-specific manner and trigger their degradation.50 

To facilitate preclinical studies, a highly potent siRNA duplex specific for the murine Alas1 mRNA (Alas1-siRNA) was identified and encapsulated in proprietary biodegradable lipid nanoparticles (LNPs) for efficient hepatic delivery following intravenous administration.51 As shown in Figure 2, the LNPs consist of ionizable cationic lipids that are neutral at physiologic pH and acquire apolipoprotein E on their surface while in circulation, which targets them for uptake by low-density lipoprotein receptors that are primarily expressed on hepatocytes52-54 (Figure 2). After clathrin-mediated endocytosis and acidification of the endocytic vesicle, the ionizable lipids on the surfaces of the LNP become positively charged and interact with anionic phospholipids in the endosomal membrane, leading to LNP-endosomal membrane fusion.55 Subsequently, Alas1-siRNA is released into the hepatocyte cytoplasm and is loaded into the RNA-induced silencing complex (RISC), where the double strands unwind and the guide strand loads onto the RISC complex while the passenger strand gets degraded (Figure 2). The guide strand-loaded RISC complex binds Alas1 mRNA in a complementary manner and degrades the target mRNA, ultimately leading to decreased ALAS1 protein expression.

Figure 2.

Mechanism of LNP-mediated Alas1 silencing in hepatocytes. The LNPs encapsulating the double-stranded Alas1-siRNA consist of cationic, ionizable lipids that are positively charged at acidic pH but neutral at physiologic pH. While in circulation, the LNPs acquire apolipoprotein E on their surface, which is bound primarily by low-density lipoprotein receptors that are highly expressed on the surface of hepatocytes. This induces clathrin-mediated endocytosis and the formation of early endosomes. As the endosomes mature and the pH becomes highly acidic, the ionizable lipids turn positive and bind to the negative lipids that are exposed on the endosomal membrane, leading to membrane fusion and disruption of the LNP. This allows escape of the Alas1-siRNA into the hepatocyte cytoplasm, where it is loaded onto the RISC. The passenger (sense) strand is degraded, leaving only the guide (antisense) strand on the RISC, which binds endogenous Alas1 mRNA in a complementary manner and degrades it. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 2.

Mechanism of LNP-mediated Alas1 silencing in hepatocytes. The LNPs encapsulating the double-stranded Alas1-siRNA consist of cationic, ionizable lipids that are positively charged at acidic pH but neutral at physiologic pH. While in circulation, the LNPs acquire apolipoprotein E on their surface, which is bound primarily by low-density lipoprotein receptors that are highly expressed on the surface of hepatocytes. This induces clathrin-mediated endocytosis and the formation of early endosomes. As the endosomes mature and the pH becomes highly acidic, the ionizable lipids turn positive and bind to the negative lipids that are exposed on the endosomal membrane, leading to membrane fusion and disruption of the LNP. This allows escape of the Alas1-siRNA into the hepatocyte cytoplasm, where it is loaded onto the RISC. The passenger (sense) strand is degraded, leaving only the guide (antisense) strand on the RISC, which binds endogenous Alas1 mRNA in a complementary manner and degrades it. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

Proof of concept of this RNAi therapeutic was established using a murine model of AIP that has ∼30% of normal HMBS enzymatic activity. Treatment of these mice with serial injections of phenobarbital, a prototypic porphyrinogenic agent, induces hepatic Alas1 expression and leads to massive accumulation of plasma and urinary ALA and PBG, akin to a clinical acute attack in patients.56 

When AIP mice were administered a single infusion of the lead LNP-encapsulated Alas1-siRNA (LNP-Alas1) and then challenged with serial phenobarbital injections, hepatic Alas1 and plasma and urinary ALA and PBG remained at baseline levels, demonstrating that LNP-Alas1 completely prevented the acute attack.51 LNP-Alas1 was also highly effective in treating an ongoing acute attack, as a single infusion into AIP mice pretreated with phenobarbital rapidly decreased the elevated plasma ALA and PBG concentrations. Importantly, hepatic heme content appeared unaltered by a single therapeutic dose of LNP-Alas1, as heme saturation levels or activities of representative hepatic hemoproteins, tryptophan dioxygenase and CYP2E1, were maintained.51 

Subsequent efforts were directed to develop an Alas1-siRNA conjugated to trivalent N-acetyl galactosamine (GalNAc), as this approach permits subcutaneous administration. As depicted in Figure 3, the GalNAc-conjugated Alas1-siRNA (GalNAc-Alas1) is recognized and bound by asialoglycoprotein receptors that are abundantly and nearly exclusively expressed on hepatocytes. This induces clathrin-mediated endocytosis and transport of the GalNAc-Alas1 to the endosomal compartment, where its acidic pH causes the siRNA to be released from asialoglycoprotein receptors, which are then rapidly recycled to the hepatocyte cell surface. Alas1-targeted siRNA is released into the cytosol, loaded onto the RISC complex, and thereafter, represses Alas1 mRNA expression (Figure 3).

Figure 3.

Mechanism of GalNAc-conjugated siRNA-mediated silencing of hepatocyte Alas1. Alas1-siRNA is conjugated to trivalent GalNAc, which is recognized and bound by asialoglycoprotein receptors that are abundantly and near-exclusively expressed on the surface of hepatocytes. This induces clathrin-mediated endocytosis and the formation of early endosomes, which mature into late endosomes that are highly acidic. The lower pH results in the release of the Alas1-siRNA from the asialoglycoprotein receptors, which are then rapidly recycled to the hepatocyte cell surface. Alas1-siRNAs then escape to the hepatocyte cytoplasm and are loaded onto RISC. The passenger (sense) strand is degraded, leaving only the guide (antisense) strand on the RISC, which binds endogenous Alas1 mRNA in a complementary manner and degrades it. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 3.

Mechanism of GalNAc-conjugated siRNA-mediated silencing of hepatocyte Alas1. Alas1-siRNA is conjugated to trivalent GalNAc, which is recognized and bound by asialoglycoprotein receptors that are abundantly and near-exclusively expressed on the surface of hepatocytes. This induces clathrin-mediated endocytosis and the formation of early endosomes, which mature into late endosomes that are highly acidic. The lower pH results in the release of the Alas1-siRNA from the asialoglycoprotein receptors, which are then rapidly recycled to the hepatocyte cell surface. Alas1-siRNAs then escape to the hepatocyte cytoplasm and are loaded onto RISC. The passenger (sense) strand is degraded, leaving only the guide (antisense) strand on the RISC, which binds endogenous Alas1 mRNA in a complementary manner and degrades it. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

GalNAc-Alas1 also effectively prevented acute attacks in the AIP mice when administered subcutaneously once a week for 4 consecutive weeks (Figure 4). Remarkably, these 4 doses provided significant protection against the induction of an acute attack for at least 6 weeks (Figure 4). The long-lasting activity of GalNAc-conjugated siRNAs has been attributed to their stability in highly acidic endosomal compartments, which serve as long-term depots for the siRNA.57 Notably, a single GalNAc-Alas1 dose was highly effective in treating an ongoing attack, as it drove down the elevated plasma ALA and PBG concentrations much more rapidly and effectively compared with 2 consecutive daily intravenous doses of hemin (4.0 mg/kg per day) (Figure 5). Based on these promising results, a GalNAc-conjugated siRNA targeting the human ALAS1 sequence (givosiran) was developed and evaluated for its safety in rodents and nonhuman primates before advancing to clinical trials.

Figure 4.

GalNAc-Alas1 effectively prevents phenobarbital-induced attacks in AIP mice. AIP mice were subcutaneously administered GalNAc-Alas1 (denoted by black triangles [▼]; 3 mg/kg per dose) once a week for 4 consecutive weeks and then challenged with serial phenobarbital injections (denoted by arrows [↓]; doses were 110, 120, and 130 mg/kg per day) at weeks (W) 0, 2, 4, and 6 after the last siRNA dose to determine the effectiveness and durability of GalNAc-Alas1 to prevent attacks. Blood samples were collected (depicted by X marks), and plasma ALA and PBG concentrations were determined. Data shown are means + standard deviations. ∗P < .01 and ∗∗P < .005 vs control AIP mice that were phenobarbital-induced after treatment with saline (no GalNAc-Alas1), t-test (n = 3-4).

Figure 4.

GalNAc-Alas1 effectively prevents phenobarbital-induced attacks in AIP mice. AIP mice were subcutaneously administered GalNAc-Alas1 (denoted by black triangles [▼]; 3 mg/kg per dose) once a week for 4 consecutive weeks and then challenged with serial phenobarbital injections (denoted by arrows [↓]; doses were 110, 120, and 130 mg/kg per day) at weeks (W) 0, 2, 4, and 6 after the last siRNA dose to determine the effectiveness and durability of GalNAc-Alas1 to prevent attacks. Blood samples were collected (depicted by X marks), and plasma ALA and PBG concentrations were determined. Data shown are means + standard deviations. ∗P < .01 and ∗∗P < .005 vs control AIP mice that were phenobarbital-induced after treatment with saline (no GalNAc-Alas1), t-test (n = 3-4).

Close modal
Figure 5.

GalNAc-Alas1 effectively treats ongoing induced attacks in AIP mice. AIP mice were induced with a combination of serial phenobarbital injections and diethoxycarbonyl-1,4-dihydrocollidine (DDC) (denoted by arrows [↓]; phenobarbital doses were 90, 100, 110, and 90 mg/kg per day, whereas DDC dose was 20 mg/kg per day) for 4 consecutive days to markedly elevate plasma ALA and PBG concentrations and treated with either a single subcutaneous dose of GalNAc-Alas1 (denoted by a black triangle [▼]; 3 mg/kg), 2 daily infusions of hemin (represented by white triangles [▽]; 4 mg/kg), or saline (controls), as shown. Blood samples were collected (represented by X marks) at the indicated times, and plasma ALA and PBG concentrations were determined. Data shown are means + standard deviations (n = 3-5).

Figure 5.

GalNAc-Alas1 effectively treats ongoing induced attacks in AIP mice. AIP mice were induced with a combination of serial phenobarbital injections and diethoxycarbonyl-1,4-dihydrocollidine (DDC) (denoted by arrows [↓]; phenobarbital doses were 90, 100, 110, and 90 mg/kg per day, whereas DDC dose was 20 mg/kg per day) for 4 consecutive days to markedly elevate plasma ALA and PBG concentrations and treated with either a single subcutaneous dose of GalNAc-Alas1 (denoted by a black triangle [▼]; 3 mg/kg), 2 daily infusions of hemin (represented by white triangles [▽]; 4 mg/kg), or saline (controls), as shown. Blood samples were collected (represented by X marks) at the indicated times, and plasma ALA and PBG concentrations were determined. Data shown are means + standard deviations (n = 3-5).

Close modal

Because ALAS1 is a nonsecretory protein, one challenge in monitoring the therapeutic effectiveness of the Alas1-siRNA in patients was the inability to directly assess ALAS1 mRNA levels without serial liver biopsies. To overcome this challenge, a circulating extracellular mRNA detection (cERD) assay that measures Alas1 mRNA expression levels in serum and urinary exosomes was developed.58 This was based on previous studies showing that hepatocytes secrete specific mRNAs in exosomes, which are then released into the serum or urine and reflect levels of mRNA expression in the liver.59,60 Indeed, serum and urinary exosome Alas1 mRNA expression in AIP mice faithfully reflected those in the liver, both after phenobarbital induction and GalNAc-Alas1 treatment.58 This cERD assay permits assessment of basal hepatic ALAS1 mRNA status in patients with AHPs and related disorders,2,4 and importantly, allows for noninvasive monitoring of hepatic ALAS1 mRNA levels after RNAi-mediated silencing.

Clinical studies established the effectiveness of givosiran to continuously suppress hepatic ALAS1 and urinary ALA and PBG levels and to reduce acute attack rates in the AHPs. A multicenter, randomized, placebo-controlled phase 1 trial of givosiran for AHP was conducted in 3 parts (supplemental Table 1, available on Blood website).61 In parts A and B, single- and multiple-dose cohort studies in asymptomatic patients with AHP with elevated urinary ALA and PBG levels (so-called “chronic high excretors”) demonstrated dose-dependent reductions from baseline in urinary exosome ALAS1 mRNA, ALA, and PBG levels, which were sustained up to day 70. Urinary exosome ALAS1 mRNA was measured using the aforementioned cERD assay as a surrogate for hepatic ALAS1 mRNA.58 As expected, urinary exosome ALAS1 mRNA and urinary ALA measurements correlated closely with one another. Part C was conducted in patients with recurrent attacks, defined as ≥2 attacks in the previous 6 months or on prophylactic hemin therapy. Patients were randomized (3:1) to receive givosiran or placebo dosed quarterly or monthly. Urinary exosome ALAS1 mRNA decreased at all dosing levels, although monthly dosing led to more sustained reductions in ALAS1 mRNA and urinary ALA and PBG levels compared with quarterly dosing. This corresponded clinically to a reduction in the mean annualized attack rate in the givosiran-treated group compared with the placebo (7.2 vs 16.7) and a 48% reduction in annualized hemin usage. Antidrug antibodies were rare and did not affect safety or efficacy.62,63 The most common adverse events were injection site reactions, abdominal pain, nausea, diarrhea, and nasopharyngitis.

A phase 3, randomized, double-blind, placebo-controlled trial of 94 patients with AHP with recurrent attacks (ENVISION study) randomly assigned patients 1:1 to receive placebo or givosiran (2.5 mg/kg subcutaneously monthly) (supplemental Table 1).63 Patients with AIP who were treated with givosiran showed a 74% reduction (P < .001) in the mean annualized attack rate, a reduction in hemin usage, and improved quality of life as measured by the patient global impression of change questionnaire compared with patients receiving placebo. Fatigue, nausea, decreased estimated GFR (eGFR), and increased creatinine and alanine transaminase (ALT) were more common in patients treated with givosiran compared with patients receiving placebo. A decline in kidney function was seen in 5 patients in the givosiran-treated group. Renal biopsies in 2 of these patients showed underlying porphyria-related kidney disease. ALT elevations greater than threefold the upper limit of normal occurred in 14.6% of patients treated with givosiran compared with 2.2% of patients receiving placebo. The drug was discontinued per protocol in one patient who developed an ALT elevation greater than 8 times the upper limit of normal.

A 24-month efficacy analysis showed sustained reductions in urinary ALA and PBG levels and in the annualized attack rate in givosiran-treated patients and a significant decrease in the annualized attack rate in patients who crossed over from placebo to active drug.64 Sustained reductions in urinary ALA and PBG levels are associated with decreases in hemin use, pain, opioid use, and improved quality of life. Hepatic adverse events characterized primarily as elevations in ALT were seen in 18% of patients treated with givosiran and were mild to moderate in severity. These transaminase elevations typically occurred within the first 3 to 6 months of treatment and resolved with ongoing treatment. Twenty-two percent of patients treated with givosiran had a decline in eGFR and an increase in creatinine, which were not progressive.64 It was speculated that givosiran may accumulate in kidney tubules and lead to decreased Alas1 in animal models.65 However, preclinical studies with high doses of givosiran in rodents showed no effect of Alas1 mRNA expression in the kidney.66 

Important efficacy outcomes and side effects/toxicity of givosiran from these initial clinical studies61,63,64 relevant to the practicing clinician are summarized in Table 1.

Based on the positive clinical trial data, givosiran (Givlaari) was approved by the FDA in November 2019 for adults with AHP at a dose of 2.5 mg/kg subcutaneously monthly.67 It was approved by the European Medicines Agency in 2020 for patients 12 years of age and older. Listed precautions include monitoring for anaphylaxis, injection site reactions, and hepatic and renal toxicity. Givosiran is not recommended for women who are pregnant or breastfeeding because of limited safety information. Data collection through a global registry is ongoing to characterize the natural history and management of patients with AHP and to further characterize the real-world safety and efficacy of givosiran.68 

Clinical experience in France suggests that the biologic activity of givosiran, reflected in the median reduction in urinary ALA levels from baseline, is highest in patients treated earlier in their disease course.69 Notably, in a subset of patients reported in this small cohort study, givosiran dosing frequency was adapted (given less frequently than monthly) based on monthly measurements of urinary ALA levels without compromising treatment efficacy. Some patients had prolonged and stably low ALA levels without recurrent givosiran treatment. Based on this experience, the authors suggested the potential for personalizing treatment approaches in the future with this therapeutic.69 

Real-world clinical experience demonstrates a transient decrease in kidney function in most patients treated with givosiran with no signs of drug-induced injury.70 The decline in renal function was higher in patients with lower baseline eGFR. Renal biopsy of 1 patient showed normal tubular ALAS1 expression. The authors hypothesize that alterations in intrarenal hemodynamics may be a contributing factor. Given the available data, close monitoring of kidney function is indicated in patients treated with givosiran. Clinical experience from France reported ALT elevations in 32% of their patients treated with givosiran.69 Treatment was interrupted only if the ALT measured >5× the upper limit of normal. Among their patients with ALT elevations, all had resolution of their ALT elevations with continued dosing.

Several case series have reported elevated plasma homocysteine levels in patients with AHP receiving givosiran.71-74 Hyperhomocysteinemia has also been reported in patients with AHP in the absence of givosiran exposure.75,76 Vitamin B6 supplementation has been reported to reduce or normalize homocysteine levels in both patients with AHP receiving givosiran and those not receiving givosiran.71,73 The precise mechanism underlying the hyperhomocysteinemia remains unknown.

Although givosiran provides significant therapeutic benefit to patients with AHP, several relevant questions surrounding this therapeutic approach remain.

Are there negative consequences of sustained ALAS1 suppression?

By reducing hepatic ALAS1 activity, givosiran might theoretically reduce hepatic heme content and consequently the activity of heme-dependent hepatic proteins, such as the cytochrome p450 enzymes. This could, in turn, decrease the metabolism of concurrently administered medications. In a single drug-drug interaction study of 9 patients with biochemical evidence of AIP who remained asymptomatic, givosiran moderately inhibited CYP1A2 and CYP2D6, weakly inhibited CYP3A4 and CYP2C19, and had no effect on CYP2C9.77 CYP2D6 catalyzes the metabolism of a number of medications, including several antidepressants and neuroleptics. Some patients with AHP are treated with multiple medications to address the signs and symptoms of their disease. Whether clinically relevant drug-drug interactions will emerge on an individual basis in this polypharmacy setting remains a concern, so caution is advised in patients who require concomitant CYP1A2 or CYP2D6 substrates.

Why are plasma homocysteine levels elevated in patients with AHP, how does givosiran affect homocysteine metabolism, and what is the clinical significance of this hyperhomocysteinemia?

The observation of a co-increase in plasma homocysteine and methionine levels72,73 has led to the hypothesis that hyperhomocysteinemia in the AHPs, potentially exacerbated by givosiran, is because of hepatocyte heme deficiency leading to reduced function of cystathionine beta-synthase (CBS).74 CBS is a key homocysteine-metabolizing enzyme that requires heme for its full enzymatic activity. The finding of normal or minimally decreased exosomal ALAS1 mRNA levels in patients treated with givosiran suggests that hepatocytes are not heme deficient. The activities of CBS and cystathionine gamma lyase, another homocysteine-metabolizing enzyme, are also dependent on vitamin B6 as a cofactor.78 Vitamin B6 deficiency is reported in patients with AHP,76,79 perhaps reflecting the chronic hyperactivity of hepatic 5-aminolevulinic acid synthase, which also requires vitamin B6 (as a cofactor) or a nutritional deficiency of vitamin B6. Further investigation is required to understand homocysteine metabolism in the AHPs and how givosiran affects this. The clinical consequences of hyperhomocysteinemia in AHPs and the utility of monitoring plasma homocysteine and vitamin supplementation (ie, B vitamins and folate) are areas open for investigation.

Will givosiran affect the long-term complications of chronic pain and chronic kidney and liver disease in the AHPs?

In patients with AHP, chronic pain and chronic kidney and liver disease are linked to elevations in ALA and PBG levels.5,31,48,80 In the ENVISION trial, givosiran therapy reduced pain during attack-free periods compared with placebo,63 suggesting that a component of the chronic pain suffered by patients may be responsive to targeting ALAS1 activity. Recent studies have implicated ALA in the tubular injury in porphyria-associated kidney disease.31,81 ALA is freely filtered by the kidneys and reabsorbed in the proximal tubule by human peptide transporter 2.82 Patients with AHP harboring variants in peptide transporter 2 that result in increased renal ALA reabsorption are likely to have more severe chronic kidney disease compared with those with variants with decreased affinity for ALA.81 Any renal benefit of givosiran will have to outweigh the risk. Worsening chronic kidney disease after long-term givosiran therapy was reported in several patients with AHP with preexisting chronic kidney disease.70 An interesting feature of the hepatocellular carcinoma observed in patients with AHP, in contrast to non-AHP patients, is that it often occurs in the absence of a background of fibrosis or cirrhosis, suggesting a unique tumorigenesis. An informative study of 4 hepatocellular carcinomas developing in 3 patients with AIP demonstrated somatic mutations in the second HMBS allele in all 4 tumors and PBG (and presumably ALA) accumulation in the tumors.83 More broadly, ALA has been shown to induce oxidative stress and DNA damage in vitro.84,85 Collectively, these data suggest that long-term suppression of hepatic ALAS1 and the resulting normalization of ALA (and PBG) may prevent or ameliorate some of the chronic complications in AHP. Additional studies are needed to confirm this possibility.

Is there a role for givosiran in the treatment of acute attacks in patients with AHP?

Givosiran is FDA-approved for the prevention of acute attacks in AHP. Although murine studies support a role for givosiran in the treatment of acute attacks,51 this remains untested in patients. Givosiran treatment results in a rapid and dose-dependent reduction in urinary ALA and PBG toward the upper limit of normal in patients with AHP within hours of dosing,62 suggesting it is likely to be efficacious in acute attacks. Given the lack of clinical studies, acute attacks should be treated as per standard of care.

Can givosiran be safely administered to pregnant individuals?

There are no data on the use of givosiran in pregnant persons to evaluate its risk to the pregnancy and fetus, nor on whether the drug is present in human breast milk. Females of childbearing age should be counseled on this lack of data and provided contraception options before prescribing givosiran. In females who are planning pregnancy and who are medically stable, givosiran can be discontinued before pregnancy planning with close monitoring of symptoms. Hemin therapy has been given safely to pregnant women and could be considered.

Several alternative therapeutic approaches aimed at restoring deficient HMBS enzymatic activity in the liver are currently being developed for AIP, including mRNA-based gene therapy, enzyme replacement, and molecular chaperone therapy approaches. To date, clinical trials establishing efficacy in human disease are lacking for these therapies.

Initial gene therapy efforts using liver-targeted adeno-associated virus (AAV) to deliver a normal copy of the human HMBS (hHMBS) cDNA into AIP mice completely prevented the induction of acute biochemical attacks.86,87 However, phase I/II clinical trials in human patients with AIP failed to show clinical and/or biochemical benefit (ie, lowering of urinary ALA and PBG), presumably because of insufficient hepatocyte transduction.88 Therefore, subsequent efforts were directed to improve transgene expression by optimizing the promoter/enhancer of the therapeutic vector or by introducing sequence variants within the hHMBS cDNA that enhance HMBS activity.89,90 

More recently, the effectiveness of LNP-mediated delivery of a mRNA encoding wild-type hHMBS (LNP-HMBS-mRNA) was evaluated in AIP mice.91 Intravenous administration of a single dose of LNP-HMBS mRNA completely prevented the induction of a phenobarbital-precipitated acute attack, and its protective effects were not diminished on repeated dosing. LNP-HMBS mRNA also rapidly and effectively decreased urinary ALA and PBG levels that were markedly elevated during a phenobarbital-induced attack. Treatment with LNP-HMBS mRNA prevented phenobarbital-induced systolic hypertension and mitochondrial insufficiency, which was previously shown to be present in AIP mouse livers, brains, and muscles.91-93 

Although early efforts to replace the deficient HMBS enzyme using recombinant hHMBS enzyme were not successful in phase I/II clinical trials, recent preclinical studies have shown that conjugation of the hHMBS enzyme to apolipoprotein A1 (ApoA1), the major protein component of high-density lipoprotein that mediates its uptake by hepatocytes, prevents against phenobarbital-induced acute attacks when administered intravenously or subcutaneously into AIP mice.94 

An alternative therapeutic approach that is still in the early preclinical stages is stabilizing the normal HMBS enzyme that is encoded by the nonmutated wild-type HMBS allele using allosteric molecular chaperones. Preclinical studies in the AIP mice showed modest increases (two- to threefold) of hepatic HMBS activity and incomplete protection against phenobarbital-induced acute attacks.95 Current efforts are directed to design more potent molecular chaperone candidates.

In the AHPs, RNAi technology provided the unique opportunity to suppress hepatocyte ALAS1 mRNA with high specificity. Givosiran is highly effective in reducing recurrent acute attacks and ameliorating chronic symptoms in patients with AHP. As of the end of 2022, 520 patients worldwide were receiving commercial drug.96 RNA therapeutics provide opportunities for the future development of AHP treatments, as exemplified by the mRNA-based gene therapy currently being developed for AIP. Additional long-term follow-up data are needed to fully understand the impact and safety of these therapies.

The Porphyrias Consortium (PC) is part of the Rare Diseases Clinical Research Network, which is funded by the National Institutes of Health (NIH) and led by the NIH National Center for Advancing Translational Sciences (NCATS) through its Division of Rare Diseases Research Innovation. PC is funded under grant number U54DK083909 as a collaboration between NIH NCATS and the NIH National Institute of Diabetes and Digestive and Kidney Diseases.

Contribution: M.Y., S.K., and M.B. drafted and edited the manuscript and approved the submission.

Conflict-of-interest disclosure: M.Y. is a named inventor of intellectual property titled “siRNA-mediated therapy for the AHPs,” filed through the Icahn School of Medicine at Mount Sinai (ISMMS) and licensed to Alnylam Pharmaceuticals; as a coinventor, is entitled to a portion of the payments received by ISMMS; and also serves as a consultant for Alnylam Japan. S.K. is on an advisory board for Disc Medicine and has consulted for Alnylam Pharmaceuticals. M.B. has participated in advisory boards for Alnylam Pharmaceuticals, Recordati Rare Diseases, Disc Medicine, and Mitsubishi-Tanabe, and is a member of the scientific advisory board for the ELEVATE registry for AHPs sponsored by Alnylam Pharmaceuticals.

Correspondence: Manisha Balwani, Division of Medical Genetics and Genomics, Department of Genetics and Genomic Sciences and Medicine, Icahn School of Medicine at Mount Sinai, 1428 Madison Ave, 1st floor, New York, NY 10029; e-mail: manisha.balwani@mssm.edu.

1.
Balwani
M
,
Desnick
RJ
.
The porphyrias: advances in diagnosis and treatment
.
Blood
.
2012
;
120
(
23
):
4496
-
4504
.
2.
Lahiji
AP
,
Anderson
KE
,
Chan
A
,
Simon
A
,
Desnick
RJ
,
Ramanujam
VMS
.
5-Aminolevulinate dehydratase porphyria: update on hepatic 5-aminolevulinic acid synthase induction and long-term response to hemin
.
Mol Genet Metab
.
2020
;
131
(
4
):
418
-
423
.
3.
Thunell
S
,
Henrichson
A
,
Floderus
Y
, et al
.
Liver transplantation in a boy with acute porphyria due to aminolaevulinate dehydratase deficiency
.
Eur J Clin Chem Clin Biochem
.
1992
;
30
(
10
):
599
-
606
.
4.
Bonkovsky
HL
,
Dixon
N
,
Rudnick
S
.
Pathogenesis and clinical features of the acute hepatic porphyrias (AHPs)
.
Mol Genet Metab
.
2019
;
128
(
3
):
213
-
218
.
5.
Bonkovsky
HL
,
Maddukuri
VC
,
Yazici
C
, et al
.
Acute porphyrias in the USA: features of 108 subjects from porphyrias consortium
.
Am J Med
.
2014
;
127
(
12
):
1233
-
1241
.
6.
Fraunberg
M
,
Pischik
E
,
Udd
L
,
Kauppinen
R
.
Clinical and biochemical characteristics and genotype-phenotype correlation in 143 Finnish and Russian patients with acute intermittent porphyria
.
Medicine (Baltimore)
.
2005
;
84
(
1
):
35
-
47
.
7.
Schuurmans
MM
,
Schneider-Yin
X
,
Rufenacht
UB
, et al
.
Influence of age and gender on the clinical expression of acute intermittent porphyria based on molecular study of porphobilinogen deaminase gene among Swiss patients
.
Mol Med
.
2001
;
7
(
8
):
535
-
542
.
8.
Podvinec
M
,
Handschin
C
,
Looser
R
,
Meyer
UA
.
Identification of the xenosensors regulating human 5-aminolevulinate synthase
.
Proc Natl Acad Sci U S A
.
2004
;
101
(
24
):
9127
-
9132
.
9.
Virbasius
JV
,
Scarpulla
RC
.
Activation of the human mitochondrial transcription factor A gene by nuclear respiratory factors: a potential regulatory link between nuclear and mitochondrial gene expression in organelle biogenesis
.
Proc Natl Acad Sci U S A
.
1994
;
91
(
4
):
1309
-
1313
.
10.
Scassa
ME
,
Guberman
AS
,
Varone
CL
,
Canepa
ET
.
Phosphatidylinositol 3-kinase and Ras/mitogen-activated protein kinase signaling pathways are required for the regulation of 5-aminolevulinate synthase gene expression by insulin
.
Exp Cell Res
.
2001
;
271
(
2
):
201
-
213
.
11.
Handschin
C
,
Lin
J
,
Rhee
J
, et al
.
Nutritional regulation of hepatic heme biosynthesis and porphyria through PGC-1alpha
.
Cell
.
2005
;
122
(
4
):
505
-
515
.
12.
Phillips
JD
.
Heme biosynthesis and the porphyrias
.
Mol Genet Metab
.
2019
;
128
(
3
):
164
-
177
.
13.
Murray
M
.
Microsomal cytochrome P450-dependent steroid metabolism in male sheep liver. Quantitative importance of 6 beta-hydroxylation and evidence for the involvement of a P450 from the IIIA subfamily in the pathway
.
J Steroid Biochem Mol Biol
.
1991
;
38
(
5
):
611
-
619
.
14.
Murray
M
.
Participation of a cytochrome P450 enzyme from the 2C subfamily in progesterone 21-hydroxylation in sheep liver
.
J Steroid Biochem Mol Biol
.
1992
;
43
(
6
):
591
-
593
.
15.
Lathrop
JT
,
Timko
MP
.
Regulation by heme of mitochondrial protein transport through a conserved amino acid motif
.
Science
.
1993
;
259
(
5094
):
522
-
525
.
16.
Monteiro
HP
,
Bechara
EJ
,
Abdalla
DS
.
Free radicals involvement in neurological porphyrias and lead poisoning
.
Mol Cell Biochem
.
1991
;
103
(
1
):
73
-
83
.
17.
Lindblad
B
,
Lindstedt
S
,
Steen
G
.
On the enzymic defects in hereditary tyrosinemia
.
Proc Natl Acad Sci U S A
.
1977
;
74
(
10
):
4641
-
4645
.
18.
Felitsyn
N
,
McLeod
C
,
Shroads
AL
,
Stacpoole
PW
,
Notterpek
L
.
The heme precursor delta-aminolevulinate blocks peripheral myelin formation
.
J Neurochem
.
2008
;
106
(
5
):
2068
-
2079
.
19.
Dowman
JK
,
Gunson
BK
,
Bramhall
S
,
Badminton
MN
,
Newsome
PN
.
Liver transplantation from donors with acute intermittent porphyria
.
Ann Intern Med
.
2011
;
154
(
8
):
571
-
572
.
20.
Solis
C
,
Martinez-Bermejo
A
,
Naidich
TP
, et al
.
Acute intermittent porphyria: studies of the severe homozygous dominant disease provides insights into the neurologic attacks in acute porphyrias
.
Arch Neurol
.
2004
;
61
(
11
):
1764
-
1770
.
21.
Bissell
DM
.
Treatment of acute hepatic porphyria with hematin
.
J Hepatol
.
1988
;
6
(
1
):
1
-
7
.
22.
Nordmann
Y
,
Puy
H
,
Da Silva
V
, et al
.
Acute intermittent porphyria: prevalence of mutations in the porphobilinogen deaminase gene in blood donors in France
.
J Intern Med
.
1997
;
242
(
3
):
213
-
217
.
23.
Chen
B
,
Solis-Villa
C
,
Hakenberg
J
, et al
.
Acute intermittent porphyria: predicted pathogenicity of HMBS variants indicates extremely low penetrance of the autosomal dominant disease
.
Hum Mutat
.
2016
;
37
(
11
):
1215
-
1222
.
24.
Lenglet
H
,
Schmitt
C
,
Grange
T
, et al
.
From a dominant to an oligogenic model of inheritance with environmental modifiers in acute intermittent porphyria
.
Hum Mol Genet
.
2018
;
27
(
7
):
1164
-
1173
.
25.
Baumann
K
,
Kauppinen
R
.
Penetrance and predictive value of genetic screening in acute porphyria
.
Mol Genet Metab
.
2020
;
130
(
1
):
87
-
99
.
26.
Schmitt
C
,
Lenglet
H
,
Yu
A
, et al
.
Recurrent attacks of acute hepatic porphyria: major role of the chronic inflammatory response in the liver
.
J Intern Med
.
2018
;
284
(
1
):
78
-
91
.
27.
Elder
G
,
Harper
P
,
Badminton
M
,
Sandberg
S
,
Deybach
JC
.
The incidence of inherited porphyrias in Europe
.
J Inherit Metab Dis
.
2013
;
36
(
5
):
849
-
857
.
28.
Naik
H
,
Stoecker
M
,
Sanderson
SC
,
Balwani
M
,
Desnick
RJ
.
Experiences and concerns of patients with recurrent attacks of acute hepatic porphyria: a qualitative study
.
Mol Genet Metab
.
2016
;
119
(
3
):
278
-
283
.
29.
Cassiman
D
,
Kauppinen
R
,
Monroy
S
, et al
.
EXPLORE B: a prospective, long-term natural history study of patients with acute hepatic porphyria with chronic symptoms
.
J Inherit Metab Dis
.
2022
;
45
(
6
):
1163
-
1174
.
30.
Gouya
L
,
Ventura
P
,
Balwani
M
, et al
.
EXPLORE: a prospective, multinational, natural history study of patients with acute hepatic porphyria with recurrent attacks
.
Hepatology
.
2020
;
71
(
5
):
1546
-
1558
.
31.
Pallet
N
,
Mami
I
,
Schmitt
C
, et al
.
High prevalence of and potential mechanisms for chronic kidney disease in patients with acute intermittent porphyria
.
Kidney Int
.
2015
;
88
(
2
):
386
-
395
.
32.
Anderson
KE
,
Bloomer
JR
,
Bonkovsky
HL
, et al
.
Recommendations for the diagnosis and treatment of the acute porphyrias
.
Ann Intern Med
.
2005
;
142
(
6
):
439
-
450
.
33.
Balwani
M
,
Wang
B
,
Anderson
KE
, et al
.
Acute hepatic porphyrias: recommendations for evaluation and long-term management
.
Hepatology
.
2017
;
66
(
4
):
1314
-
1322
.
34.
Watson
CJ
,
Pierach
CA
,
Bossenmaier
I
,
Cardinal
R
.
Use of hematin in the acute attack of the “inducible” hepatic prophyrias
.
Adv Intern Med
.
1978
;
23
:
265
-
286
.
35.
Pierach
CA
,
Bossenmaier
I
,
Cardinal
R
,
Weimer
M
,
Watson
CJ
.
Hematin therapy in porphyric attacks
.
Klin Wochenschr
.
1980
;
58
(
16
):
829
-
832
.
36.
Lamon
JM
,
Frykholm
BC
,
Hess
RA
,
Tschudy
DP
.
Hematin therapy for acute porphyria
.
Medicine (Baltimore)
.
1979
;
58
(
3
):
252
-
269
.
37.
McColl
KE
,
Moore
MR
,
Thompson
GG
,
Goldberg
A
.
Treatment with haematin in acute hepatic porphyria
.
Q J Med
.
1981
;
50
(
198
):
161
-
174
.
38.
Mustajoki
P
,
Nordmann
Y
.
Early administration of heme arginate for acute porphyric attacks
.
Arch Intern Med
.
1993
;
153
(
17
):
2004
-
2008
.
39.
Anderson
KE
,
Collins
S
.
Open-label study of hemin for acute porphyria: clinical practice implications
.
Am J Med
.
2006
;
119
(
9
):
801.e19
-
801.e24
.
40.
Herrick
AL
,
McColl
KE
,
Moore
MR
,
Cook
A
,
Goldberg
A
.
Controlled trial of haem arginate in acute hepatic porphyria
.
Lancet
.
1989
;
1
(
8650
):
1295
-
1297
.
41.
Anderson
KE
,
Bonkovsky
HL
,
Bloomer
JR
,
Shedlofsky
SI
.
Reconstitution of hematin for intravenous infusion
.
Ann Intern Med
.
2006
;
144
(
7
):
537
-
538
.
42.
Willandt
B
,
Langendonk
JG
,
Biermann
K
, et al
.
Liver fibrosis associated with iron accumulation due to long-term heme-arginate treatment in acute intermittent porphyria: a case series
.
JIMD Rep
.
2016
;
25
:
77
-
81
.
43.
Puy
H
,
Gouya
L
,
Deybach
JC
.
Porphyrias
.
Lancet
.
2010
;
375
(
9718
):
924
-
937
.
44.
Marsden
JT
,
Guppy
S
,
Stein
P
, et al
.
Audit of the use of regular haem arginate infusions in patients with acute porphyria to prevent recurrent symptoms
.
JIMD Rep
.
2015
;
22
:
57
-
65
.
45.
Anderson
KE
,
Spitz
IM
,
Bardin
CW
,
Kappas
A
.
A gonadotropin releasing hormone analogue prevents cyclical attacks of porphyria
.
Arch Intern Med
.
1990
;
150
(
7
):
1469
-
1474
.
46.
Yamamori
I
,
Asai
M
,
Tanaka
F
,
Muramoto
A
,
Hasegawa
H
.
Prevention of premenstrual exacerbation of hereditary coproporphyria by gonadotropin-releasing hormone analogue
.
Intern Med
.
1999
;
38
(
4
):
365
-
368
.
47.
Singal
AK
,
Parker
C
,
Bowden
C
,
Thapar
M
,
Liu
L
,
McGuire
BM
.
Liver transplantation in the management of porphyria
.
Hepatology
.
2014
;
60
(
3
):
1082
-
1089
.
48.
Yasuda
M
,
Erwin
AL
,
Liu
LU
, et al
.
Liver transplantation for acute intermittent porphyria: biochemical and pathologic studies of the explanted liver
.
Mol Med
.
2015
;
21
(
1
):
487
-
495
.
49.
Dowman
JK
,
Gunson
BK
,
Mirza
DF
, et al
.
Liver transplantation for acute intermittent porphyria is complicated by a high rate of hepatic artery thrombosis
.
Liver Transpl
.
2012
;
18
(
2
):
195
-
200
.
50.
Fire
A
,
Xu
S
,
Montgomery
MK
,
Kostas
SA
,
Driver
SE
,
Mello
CC
.
Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans
.
Nature
.
1998
;
391
(
6669
):
806
-
811
.
51.
Yasuda
M
,
Gan
L
,
Chen
B
, et al
.
RNAi-mediated silencing of hepatic Alas1 effectively prevents and treats the induced acute attacks in acute intermittent porphyria mice
.
Proc Natl Acad Sci U S A
.
2014
;
111
(
21
):
7777
-
7782
.
52.
Beisiegel
U
,
Weber
W
,
Ihrke
G
,
Herz
J
,
Stanley
KK
.
The LDL-receptor-related protein, LRP, is an apolipoprotein E-binding protein
.
Nature
.
1989
;
341
(
6238
):
162
-
164
.
53.
Heeren
J
,
Grewal
T
,
Jackle
S
,
Beisiegel
U
.
Recycling of apolipoprotein E and lipoprotein lipase through endosomal compartments in vivo
.
J Biol Chem
.
2001
;
276
(
45
):
42333
-
42338
.
54.
Yan
X
,
Kuipers
F
,
Havekes
LM
, et al
.
The role of apolipoprotein E in the elimination of liposomes from blood by hepatocytes in the mouse
.
Biochem Biophys Res Commun
.
2005
;
328
(
1
):
57
-
62
.
55.
Schlich
M
,
Palomba
R
,
Costabile
G
, et al
.
Cytosolic delivery of nucleic acids: the case of ionizable lipid nanoparticles
.
Bioeng Transl Med
.
2021
;
6
(
2
):
e10213
.
56.
Lindberg
RL
,
Porcher
C
,
Grandchamp
B
, et al
.
Porphobilinogen deaminase deficiency in mice causes a neuropathy resembling that of human hepatic porphyria
.
Nat Genet
.
1996
;
12
(
2
):
195
-
199
.
57.
Brown
CR
,
Gupta
S
,
Qin
J
, et al
.
Investigating the pharmacodynamic durability of GalNAc-siRNA conjugates
.
Nucleic Acids Res
.
2020
;
48
(
21
):
11827
-
11844
.
58.
Chan
A
,
Liebow
A
,
Yasuda
M
, et al
.
Preclinical development of a subcutaneous ALAS1 RNAi therapeutic for treatment of hepatic porphyrias using circulating RNA quantification
.
Mol Ther Nucleic Acids
.
2015
;
4
(
11
):
e263
.
59.
Sehgal
A
,
Chen
Q
,
Gibbings
D
,
Sah
DW
,
Bumcrot
D
.
Tissue-specific gene silencing monitored in circulating RNA
.
RNA
.
2014
;
20
(
2
):
143
-
149
.
60.
Taylor
DD
,
Gercel-Taylor
C
.
The origin, function, and diagnostic potential of RNA within extracellular vesicles present in human biological fluids
.
Front Genet
.
2013
;
4
:
142
.
61.
Sardh
E
,
Harper
P
,
Balwani
M
, et al
.
Phase 1 trial of an RNA interference therapy for acute intermittent porphyria
.
N Engl J Med
.
2019
;
380
(
6
):
549
-
558
.
62.
Agarwal
S
,
Simon
AR
,
Goel
V
, et al
.
Pharmacokinetics and pharmacodynamics of the small interfering ribonucleic acid, givosiran, in patients with acute hepatic porphyria
.
Clin Pharmacol Ther
.
2020
;
108
(
1
):
63
-
72
.
63.
Balwani
M
,
Sardh
E
,
Ventura
P
, et al
.
Phase 3 trial of RNAi therapeutic givosiran for acute intermittent porphyria
.
N Engl J Med
.
2020
;
382
(
24
):
2289
-
2301
.
64.
Ventura
P
,
Bonkovsky
HL
,
Gouya
L
, et al
.
Efficacy and safety of givosiran for acute hepatic porphyria: 24-month interim analysis of the randomized phase 3 ENVISION study
.
Liver Int
.
2022
;
42
(
1
):
161
-
172
.
65.
Goma-Garces
E
,
Perez-Gomez
MV
,
Ortiz
A
.
Givosiran for acute intermittent porphyria
.
N Engl J Med
.
2020
;
383
(
20
):
1989
.
66.
Balwani
M
,
Sardh
E
,
Gouya
L
.
Givosiran for acute intermittent porphyria. Reply
.
N Engl J Med
.
2020
;
383
(
20
):
1989
-
1990
.
67.
FDA approves givosiran for acute hepatic porphyria
.
News release. US Food and Drug Administration
. 20 November 2019. Accessed 1 December 2022. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-givosiran-acute-hepatic-porphyria.
68.
US National Library of Medicine
.
ELEVATE, a registry of patients with acute hepatic porphyria (AHP)
. Updated 21 March 2023. Accessed 1 December 2022. https://clinicaltrials.gov/ct2/show/NCT04883905.
69.
Poli
A
,
Schmitt
C
,
Moulouel
B
, et al
.
Givosiran in acute intermittent porphyria: a personalized medicine approach
.
Mol Genet Metab
.
2022
;
135
(
3
):
206
-
214
.
70.
Lazareth
H
,
Poli
A
,
Bignon
Y
, et al
.
Renal function decline with small interfering RNA silencing aminolevulinic acid synthase 1 (ALAS1)
.
Kidney Int Rep
.
2021
;
6
(
7
):
1904
-
1911
.
71.
Ricci
A
,
Marcacci
M
,
Cuoghi
C
,
Pietrangelo
A
,
Ventura
P
.
Hyperhomocysteinemia in patients with acute porphyrias: a possible effect of ALAS1 modulation by siRNAm therapy and its control by vitamin supplementation
.
Eur J Intern Med
.
2021
;
92
:
121
-
123
.
72.
To-Figueras
J
,
Wijngaard
R
,
Garcia-Villoria
J
, et al
.
Dysregulation of homocysteine homeostasis in acute intermittent porphyria patients receiving heme arginate or givosiran
.
J Inherit Metab Dis
.
2021
;
44
(
4
):
961
-
971
.
73.
Vassiliou
D
,
Sardh
E
.
Homocysteine elevation in givosiran treatment: suggested ALAS1 siRNA effect on cystathionine beta-synthase
.
J Intern Med
.
2021
;
290
(
4
):
928
-
930
.
74.
Ventura
P
,
Sardh
E
,
Longo
N
, et al
.
Hyperhomocysteinemia in acute hepatic porphyria (AHP) and implications for treatment with givosiran
.
Expert Rev Gastroenterol Hepatol
.
2022
;
16
(
9
):
879
-
894
.
75.
To-Figueras
J
,
Lopez
RM
,
Deulofeu
R
,
Herrero
C
.
Preliminary report: hyperhomocysteinemia in patients with acute intermittent porphyria
.
Metabolism
.
2010
;
59
(
12
):
1809
-
1810
.
76.
Ventura
P
,
Corradini
E
,
Di Pierro
E
, et al
.
Hyperhomocysteinemia in patients with acute porphyrias: a potentially dangerous metabolic crossroad?
.
Eur J Intern Med
.
2020
;
79
:
101
-
107
.
77.
Vassiliou
D
,
Sardh
E
,
Harper
P
, et al
.
A drug-drug interaction study evaluating the effect of givosiran, a small interfering ribonucleic acid, on cytochrome P450 activity in the liver
.
Clin Pharmacol Ther
.
2021
;
110
(
5
):
1250
-
1260
.
78.
Zuhra
K
,
Augsburger
F
,
Majtan
T
,
Szabo
C
.
Cystathionine-beta-synthase: molecular regulation and pharmacological inhibition
.
Biomolecules
.
2020
;
10
(
5
):
697
.
79.
Hamfelt
A
,
Wetterberg
L
.
Pyridoxal phosphate in acute intermittent porphyria
.
Ann N Y Acad Sci
.
1969
;
166
(
1
):
361
-
364
.
80.
Stewart
MF
.
Review of hepatocellular cancer, hypertension and renal impairment as late complications of acute porphyria and recommendations for patient follow-up
.
J Clin Pathol
.
2012
;
65
(
11
):
976
-
980
.
81.
Tchernitchko
D
,
Tavernier
Q
,
Lamoril
J
, et al
.
A variant of peptide transporter 2 predicts the severity of porphyria-associated kidney disease
.
J Am Soc Nephrol
.
2017
;
28
(
6
):
1924
-
1932
.
82.
Doring
F
,
Walter
J
,
Will
J
, et al
.
Delta-aminolevulinic acid transport by intestinal and renal peptide transporters and its physiological and clinical implications
.
J Clin Invest
.
1998
;
101
(
12
):
2761
-
2767
.
83.
Molina
L
,
Zhu
J
,
Trepo
E
, et al
.
Bi-allelic hydroxymethylbilane synthase inactivation defines a homogenous clinico-molecular subtype of hepatocellular carcinoma
.
J Hepatol
.
2022
;
77
(
4
):
1038
-
1046
.
84.
De Siervi
A
,
Vazquez
ES
,
Rezaval
C
,
Rossetti
MV
,
del Batlle
AM
.
Delta-aminolevulinic acid cytotoxic effects on human hepatocarcinoma cell lines
.
BMC Cancer
.
2002
;
2
:
6
.
85.
Onuki
J
,
Chen
Y
,
Teixeira
PC
, et al
.
Mitochondrial and nuclear DNA damage induced by 5-aminolevulinic acid
.
Arch Biochem Biophys
.
2004
;
432
(
2
):
178
-
187
.
86.
Unzu
C
,
Sampedro
A
,
Mauleon
I
, et al
.
Sustained enzymatic correction by rAAV-mediated liver gene therapy protects against induced motor neuropathy in acute porphyria mice
.
Mol Ther
.
2011
;
19
(
2
):
243
-
250
.
87.
Yasuda
M
,
Bishop
DF
,
Fowkes
M
,
Cheng
SH
,
Gan
L
,
Desnick
RJ
.
AAV8-mediated gene therapy prevents induced biochemical attacks of acute intermittent porphyria and improves neuromotor function
.
Mol Ther
.
2010
;
18
(
1
):
17
-
22
.
88.
D'Avola
D
,
Lopez-Franco
E
,
Sangro
B
, et al
.
Phase I open label liver-directed gene therapy clinical trial for acute intermittent porphyria
.
J Hepatol
.
2016
;
65
(
4
):
776
-
783
.
89.
Serrano-Mendioroz
I
,
Sampedro
A
,
Alegre
M
,
Enriquez de Salamanca
R
,
Berraondo
P
,
Fontanellas
A
.
An inducible promoter responsive to different porphyrinogenic stimuli improves gene therapy vectors for acute intermittent porphyria
.
Hum Gene Ther
.
2018
;
29
(
4
):
480
-
491
.
90.
Serrano-Mendioroz
I
,
Sampedro
A
,
Serna
N
, et al
.
Bioengineered PBGD variant improves the therapeutic index of gene therapy vectors for acute intermittent porphyria
.
Hum Mol Genet
.
2018
;
27
(
21
):
3688
-
3696
.
91.
Jiang
L
,
Berraondo
P
,
Jerico
D
, et al
.
Systemic messenger RNA as an etiological treatment for acute intermittent porphyria
.
Nat Med
.
2018
;
24
(
12
):
1899
-
1909
.
92.
Homedan
C
,
Laafi
J
,
Schmitt
C
, et al
.
Acute intermittent porphyria causes hepatic mitochondrial energetic failure in a mouse model
.
Int J Biochem Cell Biol
.
2014
;
51
:
93
-
101
.
93.
Homedan
C
,
Schmitt
C
,
Laafi
J
, et al
.
Mitochondrial energetic defects in muscle and brain of a Hmbs-/- mouse model of acute intermittent porphyria
.
Hum Mol Genet
.
2015
;
24
(
17
):
5015
-
5023
.
94.
Cordoba
KM
,
Serrano-Mendioroz
I
,
Jerico
D
, et al
.
Recombinant porphobilinogen deaminase targeted to the liver corrects enzymopenia in a mouse model of acute intermittent porphyria
.
Sci Transl Med
.
2022
;
14
(
627
):
eabc0700
.
95.
Bustad
HJ
,
Toska
K
,
Schmitt
C
, et al
.
A pharmacological chaperone therapy for acute intermittent porphyria
.
Mol Ther
.
2020
;
28
(
2
):
677
-
689
.
96.
Alnylam announces preliminary fourth quarter and full year 2022 global net product revenues and provides additional updates
.
News release
. Alnylam; updated 8 January 2023. Accessed 1 March 2023. https://investors.alnylam.com/press-release?id=27161.

Author notes

M.Y. and S.K. are joint first authors.

The online version of this article contains a data supplement.

Supplemental data

Sign in via your Institution