Fact-checked by Grok 2 weeks ago

Propionic acidemia

Propionic acidemia (PA), also known as propionyl-CoA carboxylase deficiency, is a rare autosomal recessive caused by pathogenic variants in the PCCA or PCCB genes, resulting in deficient activity of the mitochondrial propionyl-CoA carboxylase (PCC). This deficiency impairs the catabolism of the isoleucine, , , and , as well as odd-chain fatty acids and , leading to the toxic accumulation of and related metabolites such as 3-hydroxypropionate, methylcitrate, and propionylcarnitine. The disorder manifests primarily in the neonatal period or later under catabolic stress, with symptoms including poor feeding, , , , seizures, , and , which can progress to coma or death if untreated. The incidence of PA varies globally, estimated at 1 in 20,000 to 1 in 250,000 live births, with higher rates in specific populations such as the of (up to 1 in 1,000) and certain Middle Eastern and communities. Diagnosis is typically achieved through detecting elevated C3-acylcarnitine levels, followed by confirmatory enzymatic assays or molecular of PCCA and PCCB. Long-term complications in survivors include developmental delays, (affecting 32%-76% of cases), growth impairment, (7%-39%), and , though early intervention has reduced mortality from over 40% in the 1980s to 7%-18% in recent decades. Management involves acute treatment of metabolic crises with intravenous glucose, hydration, L-carnitine supplementation, and ammonia scavengers or as needed, alongside chronic strategies such as protein-restricted diets using medical foods low in propiogenic , to reduce gut bacterial propionate production, and monitoring for complications. In severe cases, may be considered to improve metabolic control and . Emerging therapies, such as the investigational mRNA-based mRNA-3927, show promise in preclinical and early-phase trials for reducing decompensation events by restoring function.

Overview and Genetics

Definition and Classification

Propionic acidemia (PA) is an autosomal recessive inherited metabolic disorder characterized by a deficiency in the enzyme propionyl-CoA carboxylase (PCC), which impairs the catabolism of specific substrates and leads to the accumulation of toxic organic acids. This deficiency disrupts the breakdown of branched-chain amino acids such as isoleucine and valine, as well as methionine, threonine, odd-chain fatty acids, and the cholesterol side chain, resulting in elevated levels of propionyl-CoA and its derivatives in blood and urine. PA is classified as an organic aciduria, or , within the broader category of , specifically as a biotin-dependent carboxylase deficiency . are distinguished by the accumulation of organic acids due to defects in or pathways, often presenting with acute metabolic crises. Unlike urea cycle disorders or oxidation defects, PA falls under the organic acidurias due to its primary involvement in propionate metabolism. In normal propionate , propionyl-CoA—generated from the of , , , , odd-chain fatty acids, and —is carboxylated by to form D-methylmalonyl-CoA, a precursor that enters the after and . This mitochondrial enzyme requires as a cofactor and functions as a heterododecamer composed of α and β subunits, ensuring efficient in the first committed step of the propionate pathway. PA is biochemically distinct from related disorders such as , as it features accumulation of propionyl-CoA and without elevated levels, whereas involves a downstream defect in leading to buildup.

Genetic Basis and Mutations

Propionic acidemia is inherited in an autosomal recessive manner, which requires an individual to inherit two mutated alleles, one from each carrier parent, to manifest the disorder. The condition arises from pathogenic variants in either the PCCA or PCCB gene, which encode the alpha and beta subunits, respectively, of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). The PCCA gene is located on chromosome 13q32.3, while the PCCB gene resides on chromosome 3q22.3. Over 100 pathogenic variants have been identified in each gene, with missense substitutions, nonsense mutations, small deletions/insertions, and splicing defects being the most common types. For instance, the PCCA variant c.425G>A (p.Gly142Asp) and the PCCB variant c.1534C>T (p.Arg512Cys) are recurrent examples reported in multiple affected individuals. These mutations disrupt the assembly or function of the heteromeric enzyme, leading to deficient activity. Genotype-phenotype correlations indicate that biallelic null , such as large deletions or early truncating variants, are associated with severe neonatal-onset forms of , whereas certain missense may result in milder, late-onset presentations. Complementation studies have classified into two main groups: (pccA) for PCCA variants and /C (pccBC) for PCCB variants, highlighting the role of interallelic complementation in enzyme assembly, particularly within PCCB where subgroup B affects the N-terminal biotin-binding and subgroup C impacts the C-terminal region. Carrier frequencies for PCCA or PCCB variants vary by population, with elevated rates observed in certain ethnic groups such as the (due to a founder PCCB mutation) or populations. is recommended for at-risk families to assess recurrence risks, which stand at 25% for each when both parents are carriers. options include molecular analysis of PCCA and PCCB variants via or , enabling early diagnosis in pregnancies at risk.

Clinical Manifestations

Symptoms and Signs

Propionic acidemia most commonly manifests in the neonatal period with symptoms such as poor feeding, , , , , seizures, and , typically appearing within the first week of life. These early signs can rapidly progress to and if untreated. In cases with later onset, symptoms often include recurrent episodes of , developmental delays, , and acute encephalopathic crises, which may be triggered by illness or and present with escalating to . Neurological signs encompass , seizures, and, in chronic cases, such as , , and choreoathetosis. Physical signs frequently observed include persistent , , and manifestations of , such as reduced and abnormal heart rhythms. may contribute to and during crises. Gastrointestinal signs are prominent, featuring feeding difficulties and an elevated risk of , alongside recurrent vomiting that can lead to .

Disease Forms and Onset

Propionic acidemia manifests in distinct clinical forms differentiated primarily by the timing of onset and overall severity, with the neonatal-onset form being the most prevalent. The neonatal-onset form, representing approximately 50%-60% of diagnosed cases, typically emerges within the first few days to weeks of life in otherwise healthy newborns who suddenly exhibit poor feeding, , lethargy, hypotonia, and . This severe variant progresses rapidly to life-threatening complications such as , seizures, , and coma without immediate intervention, carrying a high risk of mortality in the acute phase. In contrast, the late-onset or intermittent form is milder and generally appears after the neonatal period, often beyond infancy, with symptoms triggered by catabolic stressors like infections, , or . Affected individuals may experience episodic decompensations characterized by , , , and acute neurological disturbances, though baseline function can remain relatively stable between episodes. A less common atypical presentation mimics , featuring erythematous, scaly skin lesions in periorificial and acral distributions accompanied by alopecia and , which has been documented in isolated cases of propionic acidemia. Factors such as the underlying genotype and degree of residual propionyl-CoA carboxylase enzyme activity significantly influence the form and onset; severe neonatal cases correlate with very low residual enzyme activity, often approaching negligible levels, whereas modestly higher activity can postpone symptom emergence. Untreated, these forms can evolve from initial acute crises into chronic patterns involving recurrent decompensations alongside long-term neurodevelopmental challenges, including developmental delays and cognitive impairments.

Pathophysiology

Enzymatic Defect

Propionic acidemia is caused by a deficiency in the mitochondrial propionyl-CoA carboxylase (PCC), a biotin-dependent carboxylase that catalyzes the of propionyl-CoA to D-methylmalonyl-CoA using as the CO₂ donor and ATP as the energy source. This reaction is the committed step in the catabolic pathway for propionyl-CoA, preventing its accumulation and enabling its conversion to for entry into the tricarboxylic acid () cycle. PCC is structured as a large heterododecamer composed of six α subunits and six β subunits ((α₆β₆)), with a total molecular weight of approximately 750 kDa. The α subunit, encoded by the PCCA gene, has a molecular weight of about 72 kDa and is responsible for biotinylation through its biotin carboxylase (BC) and biotin carboxyl carrier protein (BCCP) domains. Biotin is covalently attached to the BCCP domain by holocarboxylase synthetase. The β subunit, encoded by the PCCB gene, weighs approximately 56 kDa and contains the carboxytransferase (CT) domain that performs the actual transfer of the carboxyl group to propionyl-CoA. Assembly of the holoenzyme requires proper folding, mitochondrial import of the α subunit, post-translational cleavage of the β subunit, and covalent attachment of biotin to the α subunit's BCCP domain. In normal metabolism, plays a critical role in the catabolism of propionyl-CoA derived from several sources, including the breakdown of the amino acids , , , and ; the side chain of ; odd-chain fatty acids; and propionate produced by gut bacteria such as species. The exhibits kinetic parameters suited to physiological conditions, with a Km for propionyl-CoA of approximately 0.29 mM and for of 3.0 mM, ensuring efficient processing of these substrates during or high-protein intake. Deficiencies in are classified as complete apoenzyme defects, where in PCCA or PCCB impair subunit , , or stability, resulting in near-total loss of enzymatic activity, or partial defects where attachment is compromised, leading to reduced but detectable activity. In mutant forms, are often altered, with increased Km values for substrates indicating diminished affinity and catalytic efficiency, which exacerbates propionyl-CoA buildup even at normal substrate levels. Rarely, cases of PA exhibit responsiveness due to specific in PCCA or PCCB that impair binding to the , allowing high-dose to partially restore activity. This is distinct from multiple carboxylase deficiency caused by holocarboxylase synthetase defects.

Metabolic Accumulation and Effects

In propionic acidemia (), deficiency of propionyl-CoA carboxylase () blocks the of propionyl-CoA to D-methylmalonyl-CoA, leading to the accumulation of propionyl-CoA and its downstream derivatives, including , 3-hydroxypropionate, methylcitrate, and propionylcarnitine. This reaction, normally facilitated by , is represented as: \text{Propionyl-CoA} + \text{CO}_2 + \text{ATP} \rightarrow \text{D-methylmalonyl-CoA} + \text{ADP} + \text{P}_i The buildup of these metabolites disrupts normal metabolic pathways, primarily causing due to the acidic nature of and from impaired oxidation and alternative energy production during catabolic states. Methylcitrate, in particular, inhibits key enzymes in the tricarboxylic acid (TCA) cycle, exacerbating energy deficits. Secondary metabolic effects include , mediated by the of N-acetylglutamate synthase by propionyl-CoA in liver mitochondria, which reduces N-acetylglutamate levels essential for activating synthetase in the . This inhibition occurs at physiologically relevant concentrations (Ki = 0.71 mM), aligning with serum levels observed in PA patients during crises. Additionally, accumulated metabolites induce mitochondrial dysfunction by impairing (e.g., state 3 and uncoupled rates), reducing ATP production, and decreasing mitochondrial , particularly in heart and brain tissues. arises from generation and mitochondrial permeability transition, contributing to cellular damage and energy deficits in vulnerable organs. Recent studies (as of 2025) highlight that chronic propionate elevation may induce neurological deficits through aberrant protein propionylation, affecting modifications and neuronal . These disruptions manifest in multi-organ toxicity: involves injury from energy failure and , leading to acute ; cardiotoxicity results in due to impaired and calcium in cardiac mitochondria; and hepatic stems from perturbations and accumulation in liver cells. The gut microbiota further aggravates propionate load, contributing approximately 25% of daily propionate via bacterial fermentation, with (e.g., reduced butyrate-producers like Roseburia and ) worsening during infections through increased transit time and bacterial overgrowth.

Diagnosis

Screening Methods

Newborn screening (NBS) for propionic acidemia is routinely performed using (MS/MS) on dried blood spots collected shortly after birth to detect elevated levels of C3-acylcarnitine (propionylcarnitine), the primary biochemical marker indicative of the disorder. This approach allows for early, presymptomatic identification, as C3-acylcarnitine accumulates due to the enzymatic defect in propionyl-CoA carboxylase. Expanded NBS programs incorporating MS/MS were implemented in the early in many regions, with propionic acidemia added to the U.S. Recommended Uniform Screening Panel in 2006, resulting in its inclusion across all U.S. states and territories by the mid-2010s. Similar protocols have been adopted in numerous countries worldwide, enabling population-wide detection. Screening protocols typically flag samples with C3-acylcarnitine levels exceeding laboratory-specific cutoffs, such as >0.4 µmol/L, though these thresholds vary (e.g., 0.5–1.0 µmol/L in some programs) to balance . The ratio of C3 to (C2) may also be evaluated to improve accuracy. Positive screens prompt immediate referral for confirmatory testing, often within 24–48 hours, to initiate timely intervention if confirmed. For at-risk families, presymptomatic screening of siblings of affected individuals involves biochemical analysis of acylcarnitine profiles in blood, similar to NBS methods, to detect elevated levels before symptom onset. This targeted approach is recommended for newborns not captured by routine NBS or older siblings in families with a known . Prenatal screening in high-risk pregnancies, such as those with a previously affected child, utilizes invasive procedures like (CVS) at 10–13 weeks or at 15–20 weeks to assess fetal activity (propionyl-CoA carboxylase) or identify known familial genetic mutations. These methods provide definitive but carry a small risk of . Despite their effectiveness, screening methods have limitations, including false positives arising from maternal , which can elevate C3-acylcarnitine in the newborn due to transplacental effects, or from other metabolic disorders like . Such cases, accounting for up to 40% of initial positives in some programs, necessitate prompt follow-up to differentiate true disease from benign or acquired conditions.

Diagnostic Confirmation

Confirmation of propionic acidemia (PA) following initial suspicion or typically involves a combination of biochemical, enzymatic, and genetic tests to establish the diagnosis definitively. These methods focus on detecting the underlying defect in (PCC) activity and the accumulation of characteristic metabolites, while excluding similar disorders. Enzyme assays measure activity, which is deficient in due to in the PCCA or PCCB genes. This is commonly performed on cultured fibroblasts, lymphocytes (leukocytes), or occasionally erythrocytes, where normal activity exceeds 80% of control values, while affected individuals show less than 10% activity, often approaching 0-5%. assays are preferred for their reliability and ability to support complementation studies to distinguish PCCA from PCCB defects. Biochemical confirmation includes organic acid analysis in urine using gas chromatography-mass spectrometry (GC-MS), which reveals elevated levels of 3-hydroxypropionic acid, methylcitric acid, and tiglylglycine as hallmarks of impaired propionate . Additionally, plasma acylcarnitine profiling via confirms the diagnosis by demonstrating elevated C3-carnitine (propionylcarnitine), a secondary marker of PCC deficiency. These patterns provide rapid supportive evidence, with elevations typically several-fold above normal ranges during acute or interictal states. Genetic testing involves targeted sequencing of the PCCA and PCCB genes to identify biallelic pathogenic variants, confirming the in approximately 90-97% of cases depending on the gene. Variants are classified according to American College of and (ACMG) guidelines as pathogenic or likely pathogenic, enabling family counseling and prenatal . This approach is particularly useful when assays are inconclusive or unavailable. To exclude differential diagnoses, a trial of supplementation may be administered to rule out holocarboxylase synthetase deficiency or biotinidase deficiency, which can present with overlapping and organic aciduria but respond to therapy. Non-response to , combined with specific metabolite profiles (e.g., absence of elevated 3-methylcrotonylglycine or ), supports PA over these biotin-responsive carboxylase deficiencies.

Management

Acute Crisis Management

Acute crisis management in propionic acidemia focuses on rapid stabilization during episodes of metabolic , typically triggered by factors such as or , to prevent and correct life-threatening imbalances. Hospitalization is essential for intensive care, where intravenous () glucose is administered at rates of 8-10 mg/kg/min in neonates and infants, 6-7 mg/kg/min in children, and lower rates in older patients to suppress endogenous protein breakdown and provide caloric support. and correction is prioritized to address and , using rehydration with fluid infusion of approximately 150 mL/kg per 24 hours, planned over 48 hours with adjusted (e.g., 2 g/L) and (e.g., 1.5 g/L) solutions. For severe exceeding 400-500 µmol/L, extracorporeal dialysis such as continuous venovenous hemodiafiltration (CVVHDF) is recommended to rapidly remove toxic metabolites, alongside or for refractory . Pharmacologic interventions include ammonia scavengers like (250 mg/kg bolus followed by 250 mg/kg/day ) and, in severe cases, concurrent sodium phenylbutyrate/phenylacetate to enhance nitrogen excretion. L-carnitine is given at 100 mg/kg bolus initially, then 100-200 mg/kg/day or orally (maximum 5 g/day) to facilitate conjugation and elimination of propionyl groups. Infection control is critical, as illnesses often precipitate crises and gut microbiota can exacerbate propionate production; prompt administration of broad-spectrum antibiotics is advised for suspected infections, alongside antipyretics like for fever above 38°C. All oral protein intake should be halted during the acute phase to minimize substrate load. Close monitoring is maintained throughout, with serial measurements of blood gases, levels every 3-6 hours, electrolytes, , and acylcarnitine profiles to guide therapy adjustments and assess response.

Chronic Management Strategies

Chronic management of propionic acidemia focuses on maintaining metabolic stability, promoting normal growth and development, and preventing acute decompensations through ongoing preventive measures. This involves a tailored approach that minimizes the accumulation of toxic metabolites while ensuring adequate nutrition and monitoring for complications. Dietary therapy is central to long-term care, emphasizing a low-protein diet that restricts intake of propiogenic amino acids such as isoleucine, valine, methionine, and threonine to reduce propionate production. Natural protein intake is typically restricted to 0.6-1.2 g/kg/day depending on age and tolerance, supplemented with specialized medical formulas that provide a balanced profile of essential amino acids with restricted levels of the propiogenic amino acids (isoleucine, valine, threonine, and methionine) and additional calories from carbohydrate- and fat-based sources to meet energy needs of 120-150% of recommended daily allowances. This regimen is individualized based on age, growth parameters, and biochemical markers, with regular adjustments by a metabolic dietitian to avoid malnutrition or excesses that could trigger crises. Supplementation plays a key role in detoxification and addressing potential deficiencies. L-carnitine is routinely administered at 100-200 mg/kg/day orally in divided doses to conjugate and excrete propionyl-CoA, helping to maintain normal plasma carnitine levels and mitigate risks. Biotin supplementation (e.g., 5-10 mg/day) may be trialed in patients with partial responsiveness, as it can enhance residual propionyl-CoA carboxylase activity, though it is discontinued if no biochemical improvement is observed. Monitoring for and correcting deficiencies, such as , is essential, particularly in those with gastrointestinal issues affecting absorption. Antibiotics such as (10-20 mg/kg/day cyclically for 7-10 days every 1-3 months) may be used to reduce propionate production by . Regular monitoring ensures early detection of metabolic instability or complications. Plasma amino acids, acylcarnitines, , and urine organic acids are assessed every 3-6 months, alongside growth evaluations, nutritional markers (e.g., , prealbumin), and organ function tests such as for cardiac health and renal function panels. These intervals may be more frequent in infants or during periods of rapid growth, with home tools like samples for acylcarnitines used in some cases to guide adjustments. A multidisciplinary team approach is vital for comprehensive care, involving metabolic specialists, dietitians, neurologists, cardiologists, and other providers as needed for organ-specific concerns like developmental delays or feeding difficulties. Coordination facilitates holistic support, including tube placement if oral intake is inadequate, and transition planning to adult services. In patients with frequent decompensations or severe complications, may be considered to restore function and enhance quality of life. Illness management plans, or sick-day protocols, are critical to prevent during infections or stressors. These include increasing intake (e.g., glucose polymers at 10-25% solutions) to 150% of calories while temporarily reducing protein, ensuring , and escalating to intravenous glucose (8-12 mg/kg/min) if oral measures fail, all while continuing carnitine supplementation. Families are educated on recognizing early signs of , such as or , to initiate these protocols promptly at home or seek urgent care.

Prognosis and Complications

Long-term Prognosis

With early diagnosis through and aggressive management, survival rates for individuals with propionic acidemia have improved significantly, reaching approximately 60% to adulthood for early-onset cases and over 90% for late-onset forms. Neonatal-onset carries a higher of early mortality, with historical indicating 20-30% mortality in the first year despite interventions, though recent advances have reduced overall mortality to 7-18%. For patients receiving , suggest a of around 40 years. Developmental outcomes remain challenging, with intellectual disability affecting 50-75% of survivors (typically IQ <70), manifesting as cognitive delays, motor impairments, and the need for special education in most school-aged patients. Late-onset presentations are associated with better cognitive preservation compared to neonatal forms, and early interventions such as dietary therapy can enhance achievement of developmental milestones. Neurologic sequelae, including speech delays and hypotonia, contribute to reduced independence in daily activities for a majority of affected individuals. Key prognostic factors include residual propionyl-CoA carboxylase (PCC) activity, where higher levels correlate with milder phenotypes and delayed symptom onset. Adherence to low-protein diets and carnitine supplementation, alongside minimizing metabolic crises (occurring at rates of 0.5 per patient-year), positively influences long-term trajectories. Access to liver transplantation further improves survival and metabolic stability in severe cases. Quality of life is impacted by recurrent hospitalizations, with a 2025 study using U.S. claims data (2015-2022) indicating rates of 0.3-0.6 per patient-year for propionic acidemia-related events, particularly in younger children, alongside frequent metabolic decompensations that disrupt daily functioning. Natural history studies highlight persistent multisystem involvement, underscoring the need for multidisciplinary care to optimize outcomes.

Associated Complications

Propionic acidemia is associated with a range of long-term multisystemic complications, particularly in patients with recurrent metabolic decompensations, affecting up to one-third of individuals across various organ systems. These complications arise from chronic exposure to toxic metabolites and often manifest in childhood or adolescence, contributing to significant morbidity. Neurological complications are prevalent in approximately 33-40% of patients and include basal ganglia damage, which can lead to movement disorders such as and . Optic nerve atrophy occurs in 11-25% of cases, typically with onset around age 13, while affects 13-53% of individuals, presenting with various seizure types. Elevated propionate levels contribute to neurodegeneration through mechanisms like mitochondrial dysfunction, oxidative stress, and disruption of glutamate metabolism, as highlighted in recent analyses. Cardiac issues develop in 20-30% of patients, primarily manifesting as cardiomyopathy (prevalence 7-39%) and arrhythmias, including prolonged QT intervals observed in up to 70% of cases. These are often linked to carnitine deficiency and the toxic effects of accumulated metabolites like propionyl-CoA, leading to mitochondrial impairment and ion channel alterations, with median onset around 7-14 years. Endocrine and metabolic complications encompass osteoporosis, noted in adult patients due to chronic metabolic stress, and pancreatitis, affecting 3-18%. Growth retardation is common, impacting 16% of cases and stemming from persistent acidosis and nutritional challenges. Hematologic complications involve bone marrow suppression, resulting in anemia (up to 82%), thrombocytopenia (35%), neutropenia (29%), or pancytopenia (6-15%), particularly during metabolic crises. Recurrent decompensations serve as a key risk factor, exacerbating the burden of these complications by increasing the frequency and severity of neurological and other sequelae, as evidenced by longitudinal studies.

Epidemiology

Global Prevalence

Propionic acidemia is an ultra-rare inherited metabolic disorder with a global birth incidence estimated at approximately 1 in 100,000 live births. This rate varies widely across regions, ranging from 1:20,000 to 1:250,000 live births depending on population genetics and screening practices. In Western populations, incidence is typically reported between 1:50,000 and 1:500,000 births, with an overall estimate of around 1:100,000 to 1:150,000. Newborn screening (NBS) programs have revealed higher detection rates in populations with comprehensive implementation, such as 1:129,792 to 1:733,000 in the United States based on tandem mass spectrometry data. In Europe, birth prevalence ranges from 0.32 to 2.20 per 100,000 live births (approximately 1:312,500 to 1:45,455), reflecting improved ascertainment through expanded NBS since the early 2000s. These figures underscore the role of screening in identifying cases that might otherwise go undiagnosed, particularly mild or late-onset forms. International registries, such as the European Network for Intoxication-Type Metabolic Diseases (E-IMD), provide aggregated data supporting these estimates and highlight stable underlying incidence despite increased detections. Underreporting remains a significant challenge in low-resource areas lacking universal NBS, where many cases may present clinically only after severe decompensation, leading to underestimation of true global burden. The expansion of NBS programs post-2000 has resulted in trends of higher reported incidence without evidence of changing underlying prevalence, coupled with improved early intervention and survival rates.

Population Variations

Propionic acidemia exhibits notable variations in prevalence across populations with high rates of consanguinity or founder effects. The highest incidence is observed in the Inuit population of Greenland, estimated at 1 in 1,000 live births, attributed to a prevalent founder mutation (1540insCCC) in the PCCB gene. In certain Amish and Mennonite communities in North America, the disorder occurs at an estimated frequency of 1 in 4,244 live births due to a founder mutation in the PCCB gene (c.1606A>G; p.Asn536Asp). This mutation leads to a spectrum of phenotypes, ranging from neonatal onset with metabolic crises to milder presentations diagnosed in adulthood, including and developmental delays in a significant proportion of affected individuals. In contrast, the prevalence in these communities is substantially higher than the global average, attributed to the historical isolation and endogamous marriage practices that amplify the founder effect. Regional disparities are evident in the , particularly , where consanguineous marriages contribute to an elevated incidence of 1 in 2,000 to 5,000 in certain tribes, compared to 1 in 20,000 to 45,000 across the broader Middle Eastern and North African region. These high rates are linked to homozygous mutations resulting from parental relatedness, often presenting with severe neonatal forms characterized by acute metabolic decompensation, , and increased susceptibility to . Ethnic-specific mutations further influence severity; for instance, the PCCA c.1897_1900del variant, identified in Turkish populations from southern and southeastern regions, is associated with profound developmental delays, , seizures, and multisystem complications like and renal failure. In East Asian populations, propionic acidemia is rarer, with incidences reported as low as 1 in 114,820 in and 1 in 189,671 in , , and 1 in 313,000 in , though some regional screening data suggest variability up to 1 in 41,000 in for milder forms. Phenotypic differences are pronounced when comparing Middle Eastern cohorts, which often feature more aggressive neonatal presentations due to limited early screening, to Western populations where enables detection of milder or late-onset cases, reducing early mortality. Socioeconomic factors exacerbate these disparities, as access to specialized metabolic care in developing regions leads to poorer long-term outcomes, including higher rates of and death from untreated crises, whereas developed settings benefit from proactive management and lower rates.

History and Future Directions

Historical Milestones

Propionic acidemia was first described in 1961 by Childs et al. as ketotic hyperglycinemia, a condition characterized by elevated levels, , and neurological symptoms in affected infants, based on clinical observations in a family with multiple impacted siblings. This initial report highlighted the metabolic disturbance without identifying the underlying biochemical defect, marking the beginning of recognition for this rare inborn error of metabolism. In 1969, Hsia et al. pinpointed the enzymatic defect to impaired propionate carboxylation, demonstrating deficient oxidation of propionate in patient fibroblasts and establishing the link to propionyl-CoA carboxylase (PCC) deficiency. This breakthrough was followed in 1970 by Gompertz et al., who identified the accumulation of propionic acid in blood and urine during acute crises, renaming the disorder propionic acidemia and confirming its role in acidosis and ketosis. These findings shifted focus from hyperglycinemia to organic acid metabolism, enabling more targeted diagnostic approaches. During the 1980s, advances in led to the of the genes encoding PCC subunits: the PCCA gene in 1987 by Lamhonwah et al., followed by the PCCB gene in 1991, which together established the genetic basis of the disorder as autosomal recessive mutations affecting alpha or beta subunits of the enzyme. The 1990s saw the introduction of programs utilizing to detect elevated propionylcarnitine, alongside standardized dietary management protocols emphasizing restriction of propiogenic amino acids (, , , ) to prevent crises. In the early 2000s, further characterization revealed distinct complementation groups (pccA, pccB, pccC) based on somatic cell fusion studies, correlating with mutations in PCCA or PCCB, while rare biotin-responsive variants were recognized, often linked to holocarboxylase synthetase defects treatable with high-dose biotin supplementation.

Recent Research and Therapies

Recent advancements in the treatment of propionic acidemia (PA) have focused on innovative therapeutic approaches aimed at addressing the underlying enzyme deficiency in propionyl-CoA carboxylase (PCC). One promising strategy involves mRNA-based therapy, exemplified by Moderna's investigational mRNA-3927, which encodes the alpha subunit of PCC (PCCA) to restore enzyme function in the liver. In a phase 1/2 open-label trial (NCT04159103), interim data from 16 pediatric and adult patients across five dose levels demonstrated dose-dependent increases in hepatic PCC activity following intravenous administration, with peak activity observed after the third dose. Additionally, treatment led to significant reductions in disease-related metabolites, including 2-methylcitric acid (up to 47% decrease), 3-hydroxypropionate (up to 84% decrease), propionylcarnitine (up to 53% decrease), and n-propionylglycine (up to 57% decrease) from baseline in most participants. The therapy was generally well-tolerated, with no dose-limiting toxicities reported, though common adverse events included pyrexia, diarrhea, and vomiting; furthermore, the frequency of metabolic decompensation events decreased from 50% pre-treatment to 12.5% during the treatment period, indicating a relative risk reduction of 70%. Gene therapy represents another frontier, with preclinical studies utilizing (AAV) vectors to deliver PCCA or PCCB genes for hepatic expression, aiming for long-term correction of the metabolic defect. The National Institutes of Health's Platform Vector Gene Therapy (PaVe-GT) program has prioritized PA, particularly PCCA-related forms, developing AAV-based constructs that have shown efficacy in restoring PCC activity and improving survival in neonatal murine models of the disease. Systemic administration of an AAV44.9 encoding human PCCA in a lethal PA model resulted in normalized propionylcarnitine levels and prevented metabolic crises, supporting progression toward clinical translation. As of 2025, these efforts remain in preclinical stages, with early-phase human trials anticipated by 2026 under regulatory guidance to optimize design and dosing for and durability. Other investigational pipelines include small-molecule approaches to modulate metabolic pathways. BBP-671, developed by BridgeBio Pharma (now Therapeutics), is an oral pantothenate activator designed to boost levels and alleviate toxic metabolite accumulation in PA; a phase 1 trial (NCT04836494) evaluating its , , and in healthy volunteers and PA patients is ongoing, with initial dosing completed in 2022 and no major safety signals reported to date. Complementing this, BCAT2 inhibitors from Agios Pharmaceuticals target 2 to limit propionyl-CoA precursor formation, reducing downstream toxicity; these compounds are in early lead optimization for PA and related organic acidemias, with preclinical data indicating potential to lower metabolite burdens without disrupting . Liver transplantation, an established intervention since the 1990s, continues to serve as a viable option for stabilizing severe cases by providing functional from donor hepatocytes. Data from 2025 registries, including the , report graft survival rates exceeding 80% at five years post-transplant in pediatric patients, with overall patient survival approaching 97% at one year and 85% at ten years, significantly mitigating recurrent decompensations and improving . Natural history studies have informed these developments by elucidating disease burden and guiding research. A 2025 U.S. claims-based analysis of over 300 PA patients revealed high rates of metabolic (31.4% experiencing at least one event over 2.7 years) and elevated healthcare utilization, underscoring the need for targeted therapies. Ongoing trials, such as the NIH's prospective study (NCT02890342), are investigating neurological s like elevated propionate levels and their correlation with cognitive and motor impairments, with preliminary 2025 findings linking persistent hyperpropionemia to dysfunction in PA cohorts.

References

  1. [1]
    Propionic Acidemia - GeneReviews® - NCBI Bookshelf - NIH
    May 17, 2012 · Neonatal-onset PA, the most common form, is characterized by a healthy newborn with poor feeding and decreased arousal in the first few days of life.
  2. [2]
    Propionic Acidemia: Overview, Etiology and Pathophysiology ...
    Sep 23, 2024 · Propionic acidemia is an autosomal recessive, inherited, metabolic disorder that is caused by a defective form of the enzyme propionyl-coenzyme A (CoA) ...
  3. [3]
    Propionic Acidemia - Symptoms, Causes, Treatment | NORD
    Apr 17, 2020 · Propionic acidemia is a rare metabolic disorder affecting from 1/20,000 to 1/250,000 individuals in various regions of the world. It is ...
  4. [4]
    Propionic acidemia - Orphanet
    Propionic acidemia (PA) is an organic aciduria caused by the deficient activity of the propionyl Coenzyme A carboxylase and is characterized by life threatening ...
  5. [5]
    Entry - #606054 - PROPIONIC ACIDEMIA - OMIM - (OMIM.ORG)
    Propionic acidemia is an autosomal recessive metabolic disorder caused by defective functioning in the mitochondrial enzyme propionyl CoA carboxylase (PCC), ...
  6. [6]
    Overview of mutations in the PCCA and PCCB genes ... - PubMed
    24 mutations in the PCCA gene and 29 in the PCCB gene have been reported, most of them single base substitutions causing amino acid replacements and a variety ...
  7. [7]
    Propionic Acidemia: Diagnosis and Neuroimaging Findings of This ...
    The main features of this disorder are based on recurrent vomiting, ketosis, hypotonia, difficult feeding, lethargy, hyperglycinemia, episodes of seizure and ...<|control11|><|separator|>
  8. [8]
    Acrodermatitis enteropathica-like cutaneous lesions in organic ...
    Cutaneous lesions resembling acrodermatitis enteropathica were present in two infants with methylmalonic acidemia and in one infant with propionic acidemia.
  9. [9]
    Propionyl-CoA Carboxylase Deficiency in a Patient with Biotin ...
    Nov 1, 1977 · The activity of this enzyme in fibroblasts derived from the patient and grown in media low in biotin was 4% of normal.
  10. [10]
  11. [11]
  12. [12]
    Inhibition by propionyl-coenzyme A of N-acetylglutamate synthetase ...
    In the search for the mechanism by which hyperammonemia complicates propionic and methylmalonic acidemia the effects of a series of acyl-coenzyme A (CoA) ...
  13. [13]
  14. [14]
    Disturbance of bioenergetics and calcium homeostasis provoked by ...
    Propionic acidemia is caused by lack of propionyl-CoA carboxylase activity. It is biochemically characterized by accumulation of propionic (PA) and ...
  15. [15]
  16. [16]
    Pathophysiological mechanisms of complications associated with ...
    Jul 22, 2023 · Most research suggests that propionyl-CoA and its metabolites can impair mitochondrial energy metabolism and cause cellular damage by inducing ...Missing: effects | Show results with:effects
  17. [17]
  18. [18]
    Altered gut microbiome diversity and function in patients with ...
    Propionic acidemia (PA) is an inherited metabolic disorder of propionate metabolism, where the gut microbiota may play a role in pathophysiology and ...
  19. [19]
  20. [20]
    Screening for Methylmalonic and Propionic Acidemia - NIH
    Feb 7, 2022 · Wisconsin's newborn screening program implemented second-tier testing on specimens with elevated propionylcarnitine (C3) to aid in the ...
  21. [21]
    Recommended Uniform Screening Panel - HRSA
    Jul 11, 2024 · It is recommended that every newborn be screened for all disorders on the RUSP. Most states screen for the majority of disorders on the RUSP; ...
  22. [22]
    Tracking Louisiana Newborn Screening Conditions
    Date Added to the Recommended Uniform Screening Panel, Date Added to the ... Propionic Acidemia, 2006, 2006. Methylmalonic Acidemia (methylmalonyl-CoA ...
  23. [23]
    Newborn screening information for propionic acidemia | Baby Health
    Early Signs · Poor feeding · Vomiting · Loss of appetite · Weak muscle tone (known as hypotonia) · Sleeping longer or more often · Tiredness.
  24. [24]
    Implementation of second-tier tests in newborn screening for the ...
    Apr 30, 2021 · 3; C3 and C3/C2 values were clearly above the cut-off values in MUT, CblA, CblB, CblC, and propionic acidemia while patients with SUCLA2 or ...
  25. [25]
    [PDF] Newborn Screening ACT Sheet - Elevated C3 Acylcarnitine
    May 24, 2024 · Propionic acidemia (PA); is caused by a defect in propionyl-CoA carboxylase, which converts propionyl-CoA to methylmalonyl-CoA; MMA results from ...Missing: tandem mass spectrometry
  26. [26]
    Diagnosis - Propionic Acidemia Foundation
    If the baby isn't screened at birth, late onset cases may present themselves throughout the first few months of life with failure to thrive, vomiting, trouble ...Missing: neonatal | Show results with:neonatal
  27. [27]
    Biochemical and genetic approaches to the prenatal diagnosis of ...
    Oct 7, 2020 · Propionic acidemia (PA) is a serious metabolic disorder, and different approaches have been applied to its prenatal diagnosis.
  28. [28]
  29. [29]
  30. [30]
  31. [31]
  32. [32]
    Proposed guidelines for the diagnosis and management of ...
    Methylmalonic and propionic acidemia (MMA/PA) are inborn errors of metabolism characterized by accumulation of propionic acid and/or methylmalonic acid due ...
  33. [33]
  34. [34]
    Methylmalonic and Propionic Acidemias: Clinical Management Update
    Recent clinical studies and management guidelines for the treatment of the organic acidopathies methylmalonic acidemia (MMA) and propionic acidemia (PA) ...
  35. [35]
    [PDF] For healthcare professionals Propionic Aciduria - E-IMD
    Recent studies indicate long-term survival rates of. ~60% in early-onset PA and >90% for late-onset PA. Most surviving individuals have varying degrees of ...<|separator|>
  36. [36]
    Survival of propionic acidemia patients with liver transplant - NIH
    May 27, 2024 · At 33 years of age the survival probability was only 62% among PA patients and 98% in the general population. Based on the survival ...
  37. [37]
    Propionic acidemia: clinical course and outcome in 55 pediatric and ...
    Jan 10, 2013 · Although improved acute and long-term management have increased the survival rates within the last decades, the neurologic outcome of PA ...
  38. [38]
    Clinical burden of propionic acidemia in the United States: a claims ...
    Jul 15, 2025 · Patients with propionic acidemia (PA) may face recurrent metabolic decompensation events (MDEs) and multisystemic complications.
  39. [39]
    Propionic acidemia: clinical course and outcome in 55 pediatric and ...
    Jan 10, 2013 · Propionic acidemia is an inherited disorder caused by deficiency of propionyl-CoA carboxylase. Although it is one of the most frequent organic ...
  40. [40]
    Elevated propionate and its association with neurological ... - Frontiers
    Mar 18, 2025 · This review examines propionate synthesis, its physiological role, its metabolism in healthy individuals and those with PA, and the pathological link
  41. [41]
    Cardiac Complications of Propionic and Other Inherited Organic ...
    PCC activity was determined to be <2% normal activity in the fibroblasts. ... propionic acidemia amenable to gene therapy testing. Mol Ther. (2013) 21:1316 ...Missing: erythrocytes | Show results with:erythrocytes
  42. [42]
    Propionic aciduria (PA) - Diseases - E-IMD
    Estimates of incidence in Western populations have ranged from 1:50,000 to 1:500,000 births, and overall incidence is believed to be ~ 1:100-150,000.Missing: prevalence | Show results with:prevalence
  43. [43]
    Natural history of propionic acidemia in the Amish population - PMC
    Propionic acidemia (PA) in the Amish is caused by a homozygous pathogenic variant (c.1606A>G; p.Asn536Asp) in the PCCB gene. Amish patients can have borderline ...
  44. [44]
    Propionic Acidemia diagnosed in Amish adults and pregnancy ...
    The Amish of North America have an increased prevalence of PA due to a founder variant in the PCCB gene. The Amish PA phenotype is variable, and some ...
  45. [45]
    Systematic literature review and meta-analysis on the epidemiology ...
    Feb 13, 2019 · Our systematic literature review and meta-analysis confirm that PA is an ultra-rare disorder, with similar detection rates across all regions.Missing: resource | Show results with:resource
  46. [46]
    Clinical features of 27 Turkish Propionic acidemia patients with 12 ...
    In this study, we investigate the mutation spectrum of PCCAPCCB genes and phenotypic features of 27 Turkish patients with PA from the South and Southeast parts ...Missing: ethnic 1897_1900del
  47. [47]
    Prevalence of propionic acidemia in China
    Sep 9, 2023 · The diagnosis of PA is confirmed through the analysis of mutations in the PCCA and PCCB genes, as well as the measurement of PCC enzyme activity ...Background · Correlation Between... · Therapeutic Interventions...Missing: factors | Show results with:factors
  48. [48]
    Prevalence of propionic acidemia in China - PMC - NIH
    Sep 9, 2023 · High incidence of propionic acidemia in greenland is due to a ... Phenotypes and pathogenic variations in two cases of propionic acidemia.
  49. [49]
    Genetics of Propionic Acidemia (Propionyl CoA Carboxylase ...
    Feb 17, 2019 · In 1969, Hsia et al described the underlying defect in propionate carboxylation that occurs in patients with ketotic hyperglycinemia.<|control11|><|separator|>
  50. [50]
    PROPIONYL-CoA CARBOXYLASE, ALPHA SUBUNIT; PCCA - OMIM
    Propionyl-CoA carboxylase (PCC) catalyzes the first step in the catabolism of propionyl-CoA. PCC is composed of 2 nonidentical subunits, alpha and beta.
  51. [51]
    PROPIONYL-CoA CARBOXYLASE, BETA SUBUNIT; PCCB - OMIM
    Propionyl-CoA is an important intermediate in the metabolism of several amino acids and is also produced by oxidation of odd-numbered fatty acids.
  52. [52]
    Biotin-responsive propionic acidemia presenting as the rumination ...
    Biotin-responsive propionic acidemia presenting as the rumination syndrome. J Pediatr. 1978 Mar;92(3):439-41. doi: 10.1016/s0022-3476(78)80441-7.Missing: discovery | Show results with:discovery
  53. [53]
    Interim analyses of a first-in-human phase 1/2 mRNA trial ... - Nature
    Apr 3, 2024 · Propionic acidaemia is a rare disorder caused by defects in the propionyl-coenzyme A carboxylase α or β (PCCA or PCCB) subunits that leads to ...
  54. [54]
    Moderna Reports Phase I/II Data for Propionic Acidemia mRNA ...
    Apr 3, 2024 · These preliminary findings are encouraging and suggest that Moderna's mRNA-3927 may have clinical benefits for propionic acidemia patients.
  55. [55]
    The National Institutes of Health Platform Vector Gene Therapy ...
    Mar 20, 2023 · The diseases selected in PaVe-GT include two organic acidemias, propionyl-CoA carboxylase alpha subunit propionic acidemia (PCCA-related PA) and ...
  56. [56]
    Systemic gene therapy using an AAV44.9 vector rescues a neonatal ...
    Venditti and colleagues use the AAV44.9 vector to treat a severe murine model of propionic acidemia, a lethal metabolic disorder.Missing: preclinical trials
  57. [57]
    BGTC: Creating a Roadmap for Rare Disease Gene Therapy - FNIH
    The AMP BGTC remains focused on completing a streamlined, start-to-finish approval pathway guide for AAV gene therapies and is adapting to what has been learned ...
  58. [58]
    Study Details | NCT04836494 | ClinicalTrials.gov - Clinical Trials
    The purpose of this study is to assess the safety, tolerability, PK and PD of BBP-671 in healthy volunteers and patients with Propionic Acidemia or ...
  59. [59]
    Dosing of First Patient in Phase 1 Trial of BBP-671 | BridgeBio
    Aug 18, 2022 · It is being developed as a potential therapy for diseases in which CoA metabolism is deficient, including propionic acidemia (PA), methylmalonic ...
  60. [60]
    Propionic Acidemia Pipeline Insights 2025: Emerging Therapies
    May 21, 2025 · DelveInsight's "Propionic Acidemia - Pipeline Insight, 2025" provides a thorough evaluation of the current clinical development landscape, ...
  61. [61]
    Agios Pharmaceuticals divulges new BCAT2 inhibitors - BioWorld
    Jun 8, 2023 · ... acidemia, maple syrup urine disease, methylmalonic acidemia and propionic acidemia. An exemplified compound inhibited human BCAT2 and BCAT1 ...
  62. [62]
    The Evolving Trend of Liver Transplantation in Metabolic Diseases ...
    Oct 10, 2025 · ... 2025, with an overall survival rate at follow-up exceeding 96%. 2 Liver Transplantation in IMD. 2.1 Classification of IMD Potentially Leading ...
  63. [63]
    Solid organ transplantation in methylmalonic acidemia and ...
    Dec 19, 2022 · Although acute rejection occurs in about 40% of patients, chronic rejection is rare in LT, and current 10-year outcomes denote 85% survival.
  64. [64]
    Natural History, Physiology, Microbiome and Biochemistry Studies of ...
    The study objectives will be to describe the natural history of propionic acidemia in the US patients by delineating the spectrum of phenotypes and querying for ...
  65. [65]
    Elevated propionate and its association with neurological ...
    Mar 19, 2025 · This review examines propionate synthesis, its physiological role, its metabolism in healthy individuals and those with PA, and the pathological link