Fact-checked by Grok 2 weeks ago

Enzyme replacement therapy

Enzyme replacement therapy (ERT) is a medical treatment designed to address congenital deficiencies, particularly in lysosomal storage disorders (LSDs), by administering purified recombinant enzymes intravenously to restore partial enzymatic function and alleviate associated symptoms. This approach compensates for the body's inability to produce sufficient functional enzymes, which leads to the accumulation of substrates in cells and tissues. ERT has become a cornerstone therapy for several rare genetic conditions, with over a dozen FDA-approved products for various LSDs as of 2025, improving through regular infusions, though it is not curative and requires lifelong administration. The mechanism of ERT relies on the uptake of infused enzymes by target cells, often facilitated by modifications such as mannose-6-phosphate tagging to direct them to lysosomes, where they catalyze the breakdown of accumulated macromolecules like glycosaminoglycans or . Treatments are typically delivered via weekly or biweekly intravenous infusions lasting 1–4 hours, dosed by body weight, and can be administered in clinical settings or at home under supervision. Enzymes are produced using technology in cell lines such as ovary cells or plant cells to ensure high purity and scalability. Historically, ERT's development began in the with plasma-derived enzymes for conditions like alpha-1-antitrypsin deficiency, but recombinant forms gained prominence in the 1990s, starting with imiglucerase for approved by the FDA in 1994. By the early 2000s, multiple FDA-approved products expanded its applications, marking a shift from supportive care—such as blood transfusions or surgeries—to disease-modifying interventions for LSDs. As of 2025, ongoing advancements include next-generation enzymes with enhanced stability and targeted delivery to overcome tissue barriers. ERT is primarily applied to LSDs, a group of over 50 disorders caused by lysosomal enzyme defects, including (treated with imiglucerase, velaglucerase alfa, or taliglucerase alfa), Pompe disease (alglucosidase alfa, avalglucosidase alfa, or cipaglucosidase alfa with miglustat), (agalsidase alfa or agalsidase beta), and such as MPS I (laronidase), MPS II (idursulfase), and MPS VI (galsulfase). These therapies target specific enzyme deficiencies to reduce organ enlargement, improve cardiac and pulmonary function, and enhance mobility in affected patients. Early initiation, ideally through , maximizes benefits by preventing irreversible damage. Clinical efficacy varies by disease but generally includes reductions in substrate accumulation, such as decreased urinary glycosaminoglycans in patients, alongside improvements in endurance (e.g., via 6-minute walk test), joint , and overall . For instance, in , ERT significantly shrinks liver and spleen volumes and stabilizes bone health. Long-term studies, including those up to 2025, confirm sustained stability in motor function for conditions like IVA when started in adulthood. Despite its successes, ERT faces limitations, including poor penetration into the due to the blood-brain barrier, rendering it less effective for cognitive aspects of severe LSDs like MPS I and II. Challenges also encompass infusion-associated reactions (affecting up to 50% of patients), potential development of neutralizing antibodies that diminish efficacy, and restricted access to hard-to-reach tissues like and . Emerging strategies, such as and induction, aim to address these issues in recent clinical trials.

Overview and Mechanism

Definition and Principles

Enzyme replacement (ERT) is a targeted medical intervention involving the exogenous administration of purified recombinant to compensate for deficient or dysfunctional endogenous , particularly in genetic disorders such as lysosomal storage diseases (LSDs). These recombinant are typically produced using biotechnological methods, such as expression in mammalian cell lines like cells, to ensure proper and functionality mimicking the natural human . The aims to restore enzymatic activity within affected cells, thereby addressing the root cause of substrate accumulation that leads to cellular and . The foundational principles of ERT revolve around substrate clearance and reduction of pathological accumulations through restored hydrolytic activity. In LSDs, deficient enzymes result in the buildup of undegraded macromolecules within lysosomes, causing progressive cellular damage; ERT supplies functional enzymes that are internalized by target cells via , primarily through the mannose-6-phosphate receptor, and directed to lysosomes where they catalyze the breakdown of these s. This process prevents further lysosomal distension and mitigates downstream effects like and , promoting metabolic without altering the patient's . The efficacy depends on the enzyme's ability to access affected s and maintain sustained activity, highlighting the importance of periodic infusions to sustain therapeutic levels. ERT primarily targets lysosomal hydrolases, a class of enzymes critical for catabolizing complex biomolecules in metabolic pathways. These hydrolases, such as acid in or A in , function within the acidic lysosomal environment to hydrolyze glycosphingolipids, glycoproteins, and mucopolysaccharides, ensuring the recycling of cellular components. By replacing these deficient hydrolases, ERT facilitates the resumption of normal degradative processes, reducing the metabolic burden on cells. Candidacy for ERT requires confirmation of an deficiency through diagnostic methods, including enzyme activity assays on leukocytes, fibroblasts, or , which measure residual hydrolytic function, and to identify pathogenic mutations via techniques like next-generation sequencing. of affected tissues may also be used to assess substrate accumulation and correlate it with enzyme levels, ensuring the therapy is appropriate for the underlying genetic defect. Early is essential, as irreversible damage limits therapeutic benefits.

Biochemical Mechanism

Enzyme replacement therapy (ERT) relies on the delivery of recombinant lysosomal enzymes that are engineered to mimic the natural mannose-6-phosphate (M6P) targeting signal found on endogenous lysosomal hydrolases. These enzymes are produced with high-mannose N-glycans modified by the addition of M6P residues in the Golgi apparatus, catalyzed by GlcNAc-1-phosphotransferase and an uncovering enzyme, which exposes the phosphate for recognition. This tagging enables the recombinant enzymes to bind to M6P receptors on the cell surface, primarily the cation-independent mannose-6-phosphate receptor (CI-MPR), facilitating uptake into target cells via clathrin-mediated endocytosis. Once internalized, the enzyme-receptor complex is transported within clathrin-coated vesicles to early endosomes, where the mildly acidic environment promotes dissociation of the from the receptor. The receptors recycle back to the trans-Golgi network or plasma membrane, while the unbound enzymes progress to late endosomes and subsequently fuse with through the action of vesicular trafficking proteins such as Rab7 and SNARE complexes. In the , the enzymes undergo proteolytic processing to their mature, active forms, restoring degradative capacity against accumulated substrates; for instance, in , recombinant enzymes like idursulfase facilitate the breakdown of glycosaminoglycans such as dermatan and heparan sulfates, preventing lysosomal distension and cellular dysfunction. Pharmacokinetically, infused recombinant enzymes exhibit short plasma half-lives, typically ranging from 26 minutes for galsulfase to 1.5–3.6 hours for laronidase, due to rapid binding to M6P receptors and subsequent cellular uptake. This leads to preferential distribution to highly vascularized tissues such as the liver and , where uptake can achieve significant enzyme activity restoration, while shows more modest accumulation owing to limited vascular access. The short circulation time necessitates frequent intravenous infusions to maintain therapeutic levels, with clearance primarily mediated by receptor-dependent rather than renal or hepatic . A key limitation of ERT is the poor penetration across the in adults, as the tight endothelial junctions restrict access to the , resulting in negligible delivery to at conventional doses. This confines therapeutic effects to peripheral tissues and , leaving CNS manifestations of diseases like VII untreated unless high-dose regimens (e.g., 20 mg/kg) are employed, which can achieve 1–3% of wild-type activity in the but at the risk of systemic overload. strategies, such as , are explored to circumvent this barrier for CNS-involved lysosomal storage disorders.

Historical Development

Early Discoveries

The discovery of lysosomes in 1955 by provided the foundational understanding necessary for recognizing lysosomal storage disorders (LSDs) as conditions caused by enzyme deficiencies leading to substrate accumulation within cells. Building on this, researchers in the and identified specific enzyme deficiencies in various LSDs, such as beta-glucuronidase in VII (MPS VII), identified in 1973, and other hydrolases in related disorders. In 1964, de Duve proposed the concept of enzyme replacement therapy (ERT) as a potential treatment, suggesting that exogenous enzyme administration could restore lysosomal function and alleviate storage pathology. This idea marked the initial theoretical framework for ERT, emphasizing the need for enzymes capable of cellular uptake and targeting to lysosomes. In the early , preclinical experiments in animal models began to test the feasibility of ERT, focusing on intravenous to demonstrate clearance and uptake. Initial studies in mice with induced or genetic deficiencies showed that infused lysosomal enzymes could be taken up by cells via , reducing accumulated substrates in tissues like liver and . For instance, experiments with beta-glucuronidase in MPS VII models, established in the late , illustrated partial clearance of glycosaminoglycans in affected organs, providing proof-of-concept for systemic delivery. These studies highlighted the potential of ERT to mitigate storage but also revealed limitations, such as short in circulation. By the , initial human applications emerged through case reports of ERT for non-lysosomal enzyme deficiencies, prefiguring broader use in LSDs. Notably, polyethylene glycol-modified (PEG-ADA) was administered to patients with (SCID) due to ADA deficiency, achieving partial immune restoration and metabolic correction in early trials starting around 1986. These efforts demonstrated clinical tolerability and efficacy in reducing toxic metabolites, though sustained benefits required repeated dosing. Early preclinical work consistently identified key challenges, including enzyme instability during circulation and inefficient targeting to affected tissues, particularly those protected by barriers like the blood-brain barrier. Researchers noted that unmodified enzymes were rapidly cleared by the liver without reaching lysosomes, prompting investigations into modifications for improved mannose-6-phosphate receptor , a elucidated in the late 1970s and early 1980s. These hurdles underscored the need for optimized formulations to enhance delivery and longevity in vivo.

Key Milestones and Approvals

The landmark approval of enzyme replacement therapy (ERT) occurred in 1991 when the U.S. (FDA) granted approval to alglucerase (Ceredase), a placental-derived form of , for the treatment of type 1 . This therapy, produced by Genzyme Corporation, marked the first regulatory endorsement of ERT for a lysosomal storage disorder, demonstrating clinical benefits such as reduced spleen and liver volumes in patients. The approval was facilitated by the , which incentivized development for rare diseases, and relied on pivotal phase 3 trials showing sustained improvements in hematologic parameters. In the , a significant shift toward recombinant production revolutionized ERT manufacturing, addressing supply limitations of human-derived enzymes like Ceredase. This culminated in the FDA approval of imiglucerase (Cerezyme), a recombinant human expressed in ovary (CHO) cells, also for type 1 . The use of CHO cells enabled scalable, consistent production with human-like , improving safety and efficacy over placental sources, and Cerezyme quickly became the standard , with over 8,000 patients enrolled in long-term safety studies by the early 2000s. The 2000s saw expansions into additional lysosomal storage disorders, broadening ERT's clinical scope. In 2003, the FDA approved agalsidase beta (Fabrazyme) for , a recombinant alpha-galactosidase A produced in CHO cells that reduced globotriaosylceramide accumulation in vascular , based on phase 3 data from 58 patients showing histologic improvements. That same year, laronidase (Aldurazyme) received approval for mucopolysaccharidosis type I ( I), including Hurler and Hurler-Scheie forms, as a recombinant alpha-L-iduronidase that improved forced in a placebo-controlled of 45 patients. These approvals highlighted ERT's versatility across enzyme deficiencies, with subsequent extensions to pediatric populations. More recent milestones include the 2017 FDA approval of vestronidase alfa (Mepsevii) for MPS VII (Sly syndrome), a recombinant beta-glucuronidase that addressed a previously untreatable rare disorder, supported by phase 3 evidence of reduced glycosaminoglycan levels in 12 patients. As of 2025, ongoing clinical trials are exploring CNS-targeted ERT variants, such as fusion proteins designed to cross the blood-brain barrier, including tividenofusp alfa for Hunter syndrome (MPS II), which received priority review by the FDA in July 2025 based on phase 2/3 data showing cognitive stabilization, although the review was extended in October 2025 to April 2026. Parallel production advancements have enhanced ERT efficacy through glycoengineering, particularly the addition of mannose-6-phosphate (M6P) glycans to facilitate lysosomal targeting via cation-independent mannose-6-phosphate receptor-mediated uptake. Techniques like enzymatic remodeling in cell-free systems or engineered phosphotransferase expression in producer cells have increased M6P content—up to 15-fold in some cases—improving cellular delivery and therapeutic outcomes, as seen in next-generation ERTs for Pompe disease. These innovations, building on recombinant platforms, continue to refine ERT for broader disease applications.

Clinical Applications

Targeted Diseases

Enzyme replacement therapy (ERT) primarily targets lysosomal storage diseases (LSDs), more than 70 inherited metabolic disorders caused by deficiencies in lysosomal , resulting in the progressive accumulation of undegraded within cells. This accumulation disrupts lysosomal function and leads to cellular damage, inflammation, and multisystem organ dysfunction, including neurological impairment, skeletal abnormalities, and cardiopulmonary complications. ERT is particularly suitable for these conditions because it supplies the missing enzyme to degrade accumulated substrates, thereby alleviating substrate buildup and mitigating downstream organ pathology, though it is most effective for non-neurological manifestations due to limited blood-brain barrier penetration. Among LSDs, ERT has been developed for several subtypes of (MPS), including MPS I (Hurler and Scheie syndromes), MPS II (), MPS IVA (Morquio A syndrome), MPS VI (Maroteaux-Lamy syndrome), and MPS VII (Sly syndrome), as well as , , and Pompe disease. In , deficiency of leads to accumulation of glucosylceramide in macrophages, causing , bone lesions, and . Similarly, in Pompe disease, acid alpha-glucosidase deficiency results in buildup in lysosomes, particularly affecting skeletal and , leading to and . involves alpha-galactosidase A deficiency, with globotriaosylceramide accumulation in vascular , contributing to renal failure, strokes, and cardiac issues, while MPS disorders feature buildup, resulting in coarse facial features, joint stiffness, and corneal clouding. Eligibility for ERT in these LSDs emphasizes early intervention to prevent irreversible damage, often identified through programs that detect enzyme deficiencies via or fluorometric assays for conditions like Pompe and I. Biomarkers such as elevated chitotriosidase levels in , which reflect activation and disease burden in , further aid in and suitability for initiation. Beyond LSDs, similar replacement strategies are applied in other enzyme deficiencies, such as augmentation therapy with purified for to protect against lung by inhibiting neutrophil elastase; this FDA-approved therapy demonstrates biochemical efficacy and slowing of lung density loss, though clinical benefits continue to be evaluated in long-term studies.

Specific Enzyme Therapies

Imiglucerase for Gaucher Type 1 Imiglucerase, a recombinant form of the enzyme , is approved for the treatment of type 1, a lysosomal characterized by deficient enzyme activity leading to glucocerebroside accumulation in macrophages. In phase III clinical trials, imiglucerase treatment resulted in a significant reduction in spleen volume by 30-50% within the first year, with mean reductions of approximately 33% observed across adult patients receiving doses of 30-60 U/kg every two weeks. Long-term data from observational studies and registries indicate sustained improvements in bone health, including decreased incidence of bone crises, increased density, and resolution of in a majority of patients after 5-10 years of therapy. Additional recombinant options include velaglucerase alfa and taliglucerase alfa, which show comparable efficacy in reducing substrate accumulation and improving hematologic parameters. Agalsidase Beta for Fabry Disease Agalsidase beta, a recombinant α-galactosidase A, targets by hydrolyzing globotriaosylceramide (GL-3) accumulated in vascular and other tissues. Randomized controlled trials and long-term extensions have demonstrated stabilization of renal function, with estimated (eGFR) maintained in patients without baseline impairment over 4-5 years of at 1 mg/kg every two weeks, contrasting with decline in untreated cohorts. Cardiac benefits include reduction in left ventricular (LV) mass index, with meta-analyses of clinical studies showing greater LV mass regression in agalsidase beta-treated patients compared to or alternative therapies, particularly in males with classic after 1-2 years. Laronidase for Mucopolysaccharidosis Type I (MPS I) Laronidase, recombinant human α-L-iduronidase, addresses MPS I (Hurler and Scheie syndromes) by degrading glycosaminoglycans (GAGs) such as dermatan and heparan sulfate. Phase III randomized trials in patients aged 6-43 years showed enhancements in pulmonary function, with forced vital capacity (FVC) increasing by an average of 5-6% from baseline after 26 weeks compared to placebo, and sustained improvements over 3-4 years in open-label extensions. In children with attenuated MPS I, long-term treatment promotes growth, with height velocity increasing by 35-192% in those aged 8-12 years after one year, alongside reductions in urinary GAG levels by approximately 50-60% as a biomarker of substrate clearance. As of 2025, biosimilar versions of laronidase have been evaluated in phase III trials, showing equivalent efficacy in maintaining urinary GAG reductions. Alglucosidase Alfa for Pompe Disease Alglucosidase alfa, recombinant human acid α-glucosidase, treats Pompe disease by replenishing the deficient responsible for breakdown in lysosomes. In infantile-onset Pompe disease, clinical trials and extensions report improved ventilator-free survival, with approximately 83% of treated infants alive without invasive ventilation at 18 months versus 25% in historical controls, reflecting cardiac and respiratory benefits over 2-3 years. For late-onset cohorts, a randomized phase III trial demonstrated stabilization of pulmonary function and improved walking distance (6-minute walk test) after 18 months at 20 mg/kg every two weeks, with evidence of reduced muscle accumulation in responders. Newer formulations, such as avalglucosidase alfa approved in 2021, offer enhanced uptake and potentially better outcomes in both forms. Other Specific Therapies for MPS Disorders For MPS II (), idursulfase reduces urinary levels by 50-60% and improves joint mobility and growth in clinical trials. Galsulfase for MPS VI (Maroteaux-Lamy) similarly decreases accumulation, enhancing endurance as measured by 6-minute walk tests over 1-2 years. Elosulfase alfa for MPS IVA (Morquio A) improves walking distance and pulmonary function in phase III studies, while vestronidase alfa for MPS VII (Sly) reduces levels and stabilizes disease progression. Comparative Efficacy Across Therapies Meta-analyses of enzyme replacement therapies for lysosomal storage diseases reveal variable success in substrate reduction and clinical outcomes, with urinary GAG or accumulated substrate levels decreasing by 60-80% in responsive conditions like MPS I and , though efficacy is lower in neuronopathic or late-stage presentations due to blood-brain barrier limitations. Overall, these therapies consistently demonstrate organ-specific benefits, such as visceral and skeletal improvements in and MPS I, renal and cardiac stabilization in Fabry, and respiratory gains in Pompe, but response rates differ by disease and patient age at initiation. As of 2025, ongoing research into next-generation ERT, including improved stability and delivery methods, continues to address limitations in tissue penetration and immune responses.

Delivery and Administration

Routes and Methods

Enzyme replacement therapy (ERT) is predominantly administered via , which serves as the standard route for delivering recombinant enzymes to target tissues throughout the body. This method involves a slow drip over 1 to 4 hours to reduce the risk of infusion-related reactions, with infusion durations typically around 3 hours for many formulations. Infusions can be performed in clinical settings or at home, where home administration has been shown to be safe and effective for conditions like and , potentially easing the lifelong treatment burden. ERT enzymes are commonly supplied as lyophilized powders in single-dose , which are reconstituted prior to . Reconstitution typically involves adding sterile to the , followed by dilution in 0.9% (normal saline) to achieve the desired concentration for . Stabilizers such as , , and citrate salts are included in these formulations to maintain enzyme stability during storage and preparation, with often used at concentrations around 340 mg per to prevent degradation. Alternative routes are under investigation to address limitations of IV delivery, particularly for (CNS) involvement. , involving direct injection into the , is experimental for CNS diseases like MPS III ( type D), where it has shown potential to drive uptake across the blood-brain barrier in preclinical models. Subcutaneous routes are also being piloted for select s, offering advantages like self-administration, though they remain non-standard due to challenges in and are primarily explored for proteins like C1-esterase inhibitor in related replacement therapies. Patient preparation for ERT infusions includes with antihistamines, such as diphenhydramine, administered 1 to 24 hours prior to reduce risks. For patients requiring frequent dosing, venous access is often facilitated by implanted ports (port-a-caths), which support long-term administration in lysosomal storage disorders by minimizing vein damage from repeated punctures.

Dosing and Monitoring

Enzyme replacement therapy (ERT) dosing is typically weight-based and administered intravenously at regular intervals, with most regimens occurring every 1 to 2 weeks to maintain therapeutic enzyme levels. For type 1 , imiglucerase is dosed at 60 units per kilogram of body weight every 2 weeks, while velaglucerase alfa and taliglucerase alfa follow similar 60 units/kg every-other-week schedules. In Pompe disease, alglucosidase alfa is given at 20 mg/kg every 2 weeks over approximately 4 hours. For (MPS), laronidase for MPS I is 0.58 mg/kg weekly, idursulfase for MPS II is 0.5 mg/kg weekly, and galsulfase for MPS VI is 1 mg/kg weekly. These standard doses are often individualized based on disease severity and patient response, with initial infusion rates adjusted gradually to minimize reactions. Dose adjustments in ERT involve guided by clinical outcomes and biomarkers, differing slightly between pediatric and adult patients due to body weight scaling and growth considerations. In disorders, doses may be increased if urinary (GAG) levels remain elevated, targeting a reduction of at least 50% from baseline. For , adjustments are made based on chitotriosidase or glucosylsphingosine (lyso-Gb1) levels, with pediatric dosing scaled by weight to achieve similar exposure as adults. In Pompe disease, higher doses (up to 40 mg/kg every 2 weeks) have been explored for non-responders, particularly in infants, to improve muscle and respiratory function. Anti-drug antibodies, which develop in up to 20-30% of patients across ERTs, can neutralize activity and necessitate dose escalation or switching products to restore efficacy. Monitoring ERT effectiveness relies on a combination of biochemical assays, clinical assessments, and to evaluate stabilization or . Enzyme activity is assessed via trough levels in plasma, ensuring sustained pharmacodynamic effects between infusions, particularly in Pompe disease where glucose tetrasaccharide (Glc4) serves as a key for clearance. In , biomarkers like lyso-Gb1 and chitotriosidase are tracked quarterly, alongside levels and platelet counts, to confirm reductions in substrate accumulation. For , urinary levels and 6-minute walk test (6MWT) distances are primary metrics, with MRI used to measure liver and volumes for progression. Quality-of-life scales, such as the , and respiratory function tests (e.g., forced ) are incorporated to assess broader functional impacts, with evaluations typically every 3-6 months.

Risks and Complications

Adverse Effects

Enzyme replacement therapy (ERT) commonly elicits infusion-related reactions, which are typically mild and occur in varying incidences depending on the disease, reported from approximately 10% up to over 50% of patients during the initial infusions. These reactions often include fever, chills, and , resulting from release triggered by the exogenous activating immune cells and inflammatory pathways. Such symptoms usually manifest within minutes to hours of and are more frequent in the early phase, affecting up to 53% of patients in certain lysosomal storage disorders like . Organ-specific adverse effects of ERT can include transient elevations in liver enzymes in certain therapies, such as for Niemann-Pick disease, indicative of mild in hepatic tissue, alongside general symptoms such as and gastrointestinal disturbances like , , , and . These liver enzyme increases are generally self-limited and resolve without , with no evidence of progression to severe . In patients with , long-term ERT may initially exacerbate bone pain flares due to substrate mobilization and inflammatory responses in affected skeletal sites, though overall skeletal manifestations improve over time. Rare severe reactions, such as , have been reported in post-marketing surveillance, occurring in less than 1% of cases. Management of these non-immune adverse effects focuses on symptom-directed interventions, such as administering acetaminophen or other antipyretics for fever and chills, slowing the rate, or temporarily halting therapy. with antipyretics and antihistamines prior to infusions can prevent recurrence in susceptible patients, with incidence rates derived from clinical trials and post-marketing data guiding individualized adjustments. Monitoring during the first several infusions is essential to mitigate risks, ensuring most patients tolerate ERT long-term with minimal disruption.

Immune Response Challenges

One major challenge in enzyme replacement therapy (ERT) is the development of anti-drug (ADAs), which occur in a significant proportion of patients. In infantile-onset Pompe disease, approximately 90% of patients treated with alglucosidase alfa develop IgG against the recombinant enzyme, with a subset progressing to high sustained antibody titers (HSAT) that neutralize activity. IgE can also form, contributing to allergic responses, though IgG predominates in impairing therapeutic delivery. Similar is observed in other lysosomal storage disorders, such as , where up to 50% of male patients develop neutralizing ADAs during ERT with agalsidase beta. As of 2025, studies continue to explore ADA formation in patients on ERT, highlighting predictors like baseline biomarkers. The primary mechanism driving ADA formation involves the immune system's recognition of recombinant enzymes as foreign antigens, particularly due to differences in post-translational modifications like . Recombinant enzymes, often produced in non-human cell lines such as ovary (CHO) cells, exhibit glycosylation patterns—such as high-mannose structures or altered sialylation—that deviate from human endogenous forms, triggering B-cell activation and production. These foreign glycans facilitate to T-helper cells, amplifying humoral responses and leading to sustained ADA secretion that binds and inactivates the infused . ADAs significantly compromise ERT efficacy by accelerating enzyme clearance, thereby reducing its and tissue uptake. In patients with high-titer ADAs, this can result in attenuated clinical responses, such as diminished improvements in cardiac or motor outcomes in Pompe disease, with some studies reporting up to 50% less reduction in accumulation compared to low-titer patients. Additionally, ADAs exacerbate infusion-associated reactions, increasing their frequency and severity through immune complex formation and complement activation. To mitigate these immune challenges, induction (ITI) protocols have been developed, often involving immunosuppressive agents like rituximab (anti-CD20 ) combined with to deplete B cells and suppress T-cell help, achieving ADA titer reductions in 68-100% of cross-reactive immunologic material (CRIM)-negative Pompe patients in reported cohorts. Enzyme switching represents another strategy; for instance, in , transitioning from imiglucerase (CHO-derived) to velaglucerase alfa (human cell line-derived) has shown lower ADA incidence (0% vs. 23.5% in comparative trials), owing to more native-like that evades immune detection. Ongoing monitoring of ADA titers guides these interventions to restore therapeutic .

Alternative and Complementary Approaches

Other Therapies for Enzyme Deficiencies

In addition to enzyme replacement therapy (ERT), several established non-ERT approaches address deficiencies, particularly in lysosomal storage disorders (LSDs), by targeting substrate accumulation, providing donor-derived enzymes, or stabilizing proteins. These therapies offer distinct mechanisms and outcomes, often complementing or serving as alternatives to ERT depending on disease stage, patient age, and organ involvement. For instance, while ERT delivers exogenous enzymes intravenously, other options like oral agents or one-time procedures aim to mitigate lifelong infusion needs or improve (CNS) penetration. Substrate reduction therapy (SRT) employs small-molecule inhibitors to decrease the synthesis of substrates that accumulate due to enzyme deficiency, thereby reducing lysosomal overload upstream of the enzymatic block. In type 1, oral agents such as inhibit glucosylceramide synthase, leading to a 12% reduction in liver volume and 19% in volume after 12 months of treatment, with non-inferiority to ERT in maintaining organ volumes over 24 months. Eliglustat, a newer SRT, achieves similar , with 77% of patients meeting composite clinical outcomes (including 17% liver volume reduction) after one year and 85% stability comparable to ERT in switch trials. SRT provides an oral alternative for ERT-intolerant patients but is associated with higher gastrointestinal side effects, such as diarrhea in 79% of users, limiting its use in severe or pediatric cases. Hematopoietic stem cell transplantation (HSCT) offers a potentially curative option by engrafting donor stem cells that produce functional enzymes, which are secreted and taken up by recipient tissues via cross-correction, including macrophages providing sustained enzyme supply. For type I (MPS I, ), HSCT in patients under 2.5 years old stabilizes psychomotor development, improves cognitive function, and achieves 62% 10-year survival, with superior metabolic correction compared to ERT. Unlike ERT, HSCT penetrates the blood-brain barrier, addressing CNS manifestations, and is more cost-effective long-term (approximately $70,000–$205,000 total versus $218,000 annually for ERT). However, it carries risks including and up to 10% mortality with modern protocols, making it suitable primarily for young, pre-symptomatic patients. Pharmacological chaperones represent another targeted strategy, using small molecules to bind and stabilize misfolded mutant enzymes, facilitating their proper trafficking to lysosomes and enhancing residual activity. In , is approved for patients with amenable mutations (affecting 35–50% of cases), reducing globotriaosylceramide (GL-3) inclusions and lyso-Gb3 levels while maintaining renal function and decreasing cardiac mass more effectively than ERT over 18–24 months. This oral avoids intravenous but is limited to specific genetic variants confirmed by assays. Supportive care focuses on symptomatic management of complications arising from enzyme deficiencies, employing multidisciplinary interventions to alleviate , improve mobility, and prevent secondary issues without directly addressing the underlying defect. For example, in , chronic is controlled with analgesics or anticonvulsants, while in or MPS disorders, orthopedic surgeries correct skeletal deformities like bone lesions or joint contractures. These measures enhance but do not alter disease progression, often serving as adjuncts to disease-modifying therapies. Comparatively, ERT requires lifelong biweekly infusions and excels in peripheral symptom control but falters in CNS and bone penetration, whereas HSCT provides a one-time potential cure with broader organ benefits at the cost of procedural risks. SRT and pharmacological chaperones offer convenient oral dosing with efficacy akin to ERT for in select LSDs like Gaucher and Fabry, though they may cause more tolerability issues and are less suitable for advanced disease. Combination approaches, such as SRT following ERT stabilization (e.g., ) or chaperones with ERT, demonstrate synergistic effects in enhancing enzyme activity and tissue substrate reduction, improving outcomes in crossover studies.

Emerging Research Directions

Recent advancements in enzyme replacement therapy (ERT) are focusing on overcoming limitations in (CNS) penetration, particularly for lysosomal storage disorders (LSDs) like type III (MPS III), where neurological symptoms predominate. Intrathecal infusions have emerged as a promising route for direct CNS delivery, with clinical studies demonstrating improved activity and reduced substrate accumulation in the without significant systemic exposure. For instance, trials in MPS IIIA patients using intrathecal recombinant human N-sulfoglucosamine sulfohydrolase have shown tolerability and preliminary efficacy in stabilizing cognitive decline. Complementing these efforts, blood-brain barrier (BBB) shuttles and hybrid approaches with (AAV) vectors are under investigation to enhance systemic ERT's CNS access; preclinical models of MPS III have reported up to 50% restoration of activity in brain tissues following AAV-mediated delivery combined with ERT infusions. Efforts to develop next-generation enzymes aim to mitigate and extend therapeutic duration. Deimmunized variants, engineered to remove immunogenic epitopes, and plant-produced enzymes, such as those expressed in cells, have shown reduced antibody formation in preclinical and I/II trials for , with up to 80% lower anti-drug incidence compared to mammalian cell-derived counterparts. , the covalent attachment of chains, further prolongs plasma half-life—extending it from hours to days in formulations like pegunigalsidase alfa, which achieves stable over 24 months in clinical use and improves uptake in the heart and kidneys. These modifications collectively lower dosing and enhance efficacy while minimizing infusion-related reactions. Integrating gene editing technologies like / with ERT represents a hybrid strategy for achieving both immediate symptom relief and long-term correction of enzyme deficiencies. In preclinical models of MPS I, CRISPR-assisted editing of hematopoietic stem cells has been combined with ERT infusions to enable cross-correction via enzyme secretion from edited cells, resulting in sustained lysosomal function restoration without repeated dosing. This tandem approach addresses ERT's transient effects by providing a one-time genetic fix alongside ongoing enzymatic support, with early trials demonstrating up to 40% editing efficiency in patient-derived cells for LSDs. Combination therapies pairing ERT with are advancing through clinical pilots, particularly for Pompe disease, where muscle weakness limits standard ERT's reach. Phase I/II trials of AAV-mediated GAA gene transfer as an adjunct to ERT have reported dose-dependent increases in clearance and respiratory function, with one-year follow-up showing 20-30% improvement in six-minute walk distance in late-onset patients. Similarly, personalized dosing models leveraging () are being explored to optimize ERT regimens; in , AI-driven algorithms analyzing biomarkers and patient responses have enabled tailored infusions, maintaining stable disease control while reducing annual dosing volume by up to 25% in a phase II . Looking ahead, challenges persist in broadening access, especially for rare LSDs in low-resource settings, where high costs—often exceeding $300,000 annually per —restrict availability despite programs. As of 2025, regulatory expansions for compassionate use of next-gen ERTs in ultra-rare LSDs, such as those presented at international congresses, signal progress, but equitable global distribution remains a priority through initiatives like subsidized manufacturing and to developing regions. Overall, these directions hold potential to transform ERT into a more durable, accessible paradigm for deficiencies.

References

  1. [1]
    Enzyme Replacement Therapy - LiverTox - NCBI Bookshelf - NIH
    Mar 10, 2016 · Enzyme replacement therapy is typically used to replace a missing or deficient enzyme in a person with an inherited enzyme deficiency syndrome.
  2. [2]
    Enzyme-Replacement Therapies for Lysosomal Storage Diseases
    “Enzyme replacement therapy” (ERT) refers to a group of nine commercially available glycoprotein products, each intended to augment or replace the activity of ...
  3. [3]
    Enzyme replacement therapy: efficacy and limitations
    Nov 16, 2018 · Enzyme replacement therapy (ERT), based on the periodic intravenous administration of specific enzymes produced with recombinant DNA technology, ...
  4. [4]
    Improving the treatment of Pompe disease with enzyme replacement ...
    Apr 16, 2025 · This review describes the limitations of alglucosidase alfa and focuses on the strategies used to overcome these limitations, including the ...
  5. [5]
    Long-term outcomes of elosulfase alfa enzyme replacement therapy ...
    Oct 30, 2025 · Real-world data collected from MARS suggest that patients with MPS IVA who initiated ERT in adulthood remained stable over 7 years of follow-up.
  6. [6]
    Clinical insights in enzyme replacement therapy for metabolic ...
    Although enzyme replacement therapy has substantially improved outcomes for patients with lysosomal storage disorders, limitations of this therapy have become ...
  7. [7]
  8. [8]
    Lysosomal Storage Disease - StatPearls - NCBI Bookshelf - NIH
    1) Enzyme Replacement Therapy (ERT): Human recombinant enzymes are manufactured in Chinese hamster ovary(CHO) cells using DNA technology.Introduction · Evaluation · Treatment / Management · Complications
  9. [9]
    Lysosomal Enzyme Replacement Therapies - PMC - NIH
    A class of small molecules reduce the accumulation of substrates by inhibiting their biosynthesis; hence they are known as substrate reduction therapy [94]. One ...
  10. [10]
    Full article: Mannose-6-phosphate glycan for lysosomal targeting
    May 29, 2022 · Enzyme replacement therapy (ERT) for lysosomal storage diseases exploits this M6P-MPR-dependent endocytosis to deliver recombinant enzymes to ...
  11. [11]
    Toward Engineering the Mannose 6-Phosphate Elaboration ... - MDPI
    Recombinant enzyme in ERT is generally thought to require a mannose 6-phosphate (M6P) targeting signal for endocytosis into patient cells and for intracellular ...2. Materials And Methods · 2.8. Idua Uptake Assays On... · 3. Results
  12. [12]
    Mannose-6-phosphate pathway: A review on its role in lysosomal ...
    The above described process is known as the M6P-dependent pathway and is responsible for transporting most lysosomal enzymes.
  13. [13]
    Enzyme replacement therapy: efficacy and limitations - PMC
    Nov 16, 2018 · The infused enzymes have a short half-life in the circulation due to rapid binding to M6P receptors and uptake into visceral organs.
  14. [14]
    Pharmacokinetic and Pharmacodynamic Evaluation of Elosulfase ...
    Sep 19, 2014 · Enzyme replacement therapies for MPS diseases generally have a short plasma elimination half-life, from 26 min for galsulfase to 1.5 to 3.6 h ...
  15. [15]
    Overcoming the blood-brain barrier with high-dose enzyme ... - PNAS
    Oct 3, 2005 · However, improvement in brain is limited by the blood-brain barrier except in the newborn period.Missing: limitations | Show results with:limitations
  16. [16]
    Targeting the central nervous system in lysosomal storage diseases
    Mar 1, 2023 · One limitation that is common to all these strategies is the inability, or limited ability, of therapeutic agents to cross the blood-brain ...
  17. [17]
    Lysosomes as a therapeutic target | Nature Reviews Drug Discovery
    Sep 2, 2019 · Discovered in the 1950s by Christian de Duve, lysosomes are membrane-bound vesicles containing numerous hydrolytic enzymes that can break down ...
  18. [18]
    Enzyme replacement therapy for lysosomal diseases - PubMed - NIH
    In 1964, Christian de Duve first suggested that enzyme replacement might prove therapeutic for lysosomal storage diseases (LSDs).
  19. [19]
    Enzyme replacement therapy – a brief history - Fabry Disease - NCBI
    The concept of enzyme replacement therapy for lysosomal storage diseases was enunciated by de Duve in 1964. However, much cell biology had to be learned ...Missing: 1960s | Show results with:1960s
  20. [20]
    Feline mucopolysaccharidosis VII due to beta-glucuronidase ...
    Feline mucopolysaccharidosis VII due to beta-glucuronidase deficiency. Vet Pathol. 1994 Jul;31(4):435-43.Missing: discovery date
  21. [21]
    Enzyme Replacement Therapy with Polyethylene Glycol-Adenosine ...
    In April 1986, we became involved in a clinical trial of an untested approach to enzyme replacement therapy for ADA deficiency in a gravely ill child with SCID ...
  22. [22]
    Review of Treatment for Adenosine Deaminase Deficiency (ADA ...
    Sep 22, 2022 · Treatment with enzyme replacement therapy (ERT) was developed in the 1970s and became the treatment for ADA SCID by the 1980s. It remains an ...
  23. [23]
    Enzyme Therapy: Current Challenges and Future Perspectives - PMC
    Enzymes have risen as promising therapeutic tools for different pathologies, from metabolic deficiencies, such as fibrosis conditions, ocular pathologies or ...
  24. [24]
    Enzyme replacement therapy: current challenges and drug delivery ...
    Enzyme replacement therapy(ERT) can be a lucrative niche for orphan indications, leading to quicker regulatory feedback and entry into clinical trials with ...<|control11|><|separator|>
  25. [25]
    Search Orphan Drug Designations and Approvals - FDA
    For replacement therapy in patients with Gaucher's disease type I. Orphan ... Ceredase. Marketing Approval Date: 04/05/1991. Approved Labeled Indication ...
  26. [26]
    COMPANY NEWS; F.D.A. Approves Genzyme's Drug
    Apr 9, 1991 · The new drug, alglucerase, replaces an enzyme that is missing in people who have the hereditary disorder, the F.D.A. said. The agency had made ...
  27. [27]
    ERT Approval Anniversary | Gaucher Disease Blog
    In 1991, the FDA approved Genzyme's ERT drug Ceredase (alglucerase) for patients with Gaucher disease type 1. Patients nationwide with Gaucher disease ...
  28. [28]
    Search Orphan Drug Designations and Approvals - FDA
    Cerezyme. Date Designated: 11/05/1991. Orphan Designation: Replacement therapy in patients with types I, II, and III Gaucher's disease. Orphan Designation ...Missing: Ceredase | Show results with:Ceredase
  29. [29]
    Purple Book Database of Licensed Biological Products
    Cerezyme, Rx. Proper Name imiglucerase. BLA Number 020367. Applicant Genzyme Corporation. Original Approval Date May 23, 1994. Date of First Licensure. Return ...
  30. [30]
    Search Orphan Drug Designations and Approvals - FDA
    Marketing approved: ; Fabrazyme · 04/24/2003 · For use in patients with Fabry disease to reduce globotriaosylceramide (GL-3) deposition in capillary endothelium of ...
  31. [31]
    Search Orphan Drug Designations and Approvals - FDA
    Marketing approved: ; Aldurazyme · 04/30/2003 · Treatment for patients with Hurler and Hurler-Scheie forms of Mucopolysaccharidosis I (MPS I) and for patients with ...
  32. [32]
    [PDF] CENTER FOR DRUG EVALUATION AND RESEARCH
    Nov 15, 2017 · Mepsevii (vestronidase alfa-vjbk) is indicated in pediatric and adult patients for the treatment of Mucopolysaccharidosis type VII (MPS VII, Sly ...
  33. [33]
    Ultragenyx Announces FDA Approval of MEPSEVII™ (vestronidase ...
    Nov 15, 2017 · MEPSEVII is an enzyme replacement therapy designed to replace the deficient lysosomal enzyme beta-glucuronidase in MPS VII patients. "The ...
  34. [34]
    Mannose-6-phosphate glycan for lysosomal targeting - NIH
    May 29, 2022 · Enzyme replacement therapy (ERT) for lysosomal storage diseases exploits this M6P-MPR-dependent endocytosis to deliver recombinant enzymes to ...
  35. [35]
    Therapeutic Approaches in Lysosomal Storage Diseases - PMC - NIH
    Enzyme replacement therapy (ERT) consists of the intravenous administration of a properly glycosylated and functional form of the enzyme impaired in the disease ...
  36. [36]
    Newborn Screening for Lysosomal Storage Diseases - PMC - NIH
    Treatment options include enzyme replacement therapy (3), hematopoietic stem cell transplantation (4), small molecular weight drugs (5), and gene therapy ...
  37. [37]
    Accuracy of chitotriosidase activity and CCL18 concentration in ...
    Plasma chitotriosidase activity decreases dramatically after initiation of enzyme replacement therapy (ERT) and rises again when the treatment is stopped.<|control11|><|separator|>
  38. [38]
    Intravenous Alpha-1 Antitrypsin Therapy for Alpha-1 ... - NIH
    This review provides an update on the evidence for the clinical efficacy of intravenous AAT therapy for patients with AATD-related emphysema.
  39. [39]
    Imiglucerase in the management of Gaucher disease type 1 - NIH
    Oct 14, 2016 · Visceral parameters. Results from clinical trials. In the pivotal clinical trial, spleen volume was reduced by a mean of 33% in all patients ...
  40. [40]
    Improvement of bone disease by imiglucerase (Cerezyme) therapy ...
    With imiglucerase treatment, patients should anticipate resolution of BC, rapid improvement in BP, increases in BMD, and decreased skeletal complications.
  41. [41]
    Sustained, long-term renal stabilization after 54 months of ... - PubMed
    Apr 4, 2007 · Long-term agalsidase beta therapy stabilizes renal function in patients without renal involvement at baseline, maintains reduction of plasma GL-3, and sustains ...
  42. [42]
    Agalsidase alfa versus agalsidase beta for the treatment of Fabry ...
    A greater biochemical response, also in the presence of antibodies, and better reduction in left ventricular mass was observed with agalsidase beta. fabry ...Methods · Left Ventricular Mass · Lysogb3Missing: phase | Show results with:phase<|control11|><|separator|>
  43. [43]
    Growth in individuals with attenuated mucopolysaccharidosis type I ...
    Jul 23, 2022 · Data from an open-label study of laronidase showed that growth rate increased by 35%–192% in five children age 8–12 years with attenuated MPS ...2 Methods · 3 Results · 3.3 Mps I Height-For-Age...<|control11|><|separator|>
  44. [44]
    Early Treatment with Alglucosidase Alfa Prolongs Long Term ...
    Over the course of the study, alglucosidase alfa treatment markedly improved overall survival, as well as ventilator-free and invasive ventilator-free survival.
  45. [45]
    A Randomized Study of Alglucosidase Alfa in Late-Onset Pompe's ...
    Apr 15, 2010 · Treatment with alglucosidase alfa was associated with improved walking distance and stabilization of pulmonary function over an 18-month period.
  46. [46]
    Efficacy of different treatment strategies in patients ... - PubMed Central
    May 2, 2025 · In patients with MPS I, the respiratory function of patients treated with laronidase (0.58 mg/kg/week) increased by 5.6, 95% CI[2.40, 8.80] ...Missing: growth | Show results with:growth
  47. [47]
    Management of infusion-related reactions to enzyme replacement ...
    Close monitoring of patients receiving ERT is essential. Use of an adapted ERT infusion regimen with premedication resulted in improvement of signs and ...<|control11|><|separator|>
  48. [48]
    Home treatment with enzyme replacement therapy for ... - PubMed
    ERT for MPS I applied at home is safe and might alleviate the burden of life-long intravenous treatment in these patients.
  49. [49]
    Home infusion program with enzyme replacement therapy for Fabry ...
    Jun 22, 2017 · Despite the encouraging clinical outcomes achieved with ERT, a life-long intravenous (IV) treatment with an every other week schedule, may ...
  50. [50]
    [PDF] Reference ID: 5411279 - accessdata.fda.gov
    Reconstitute each 400 unit vial of Cerezyme by slowly injecting 10.2 mL of Sterile Water for Injection, USP, down the inside wall of each vial. 3. Roll and tilt ...Missing: formulation | Show results with:formulation
  51. [51]
    [PDF] 1 This label may not be the latest approved by FDA. For current ...
    Reconstitute each 5 mg vial of Fabrazyme by slowly injecting 1.1 mL of Sterile Water for Injection, USP down the inside wall of each vial. Roll and tilt each ...Missing: formulation | Show results with:formulation
  52. [52]
    Enzyme Replacement Therapy for Mucopolysaccharidosis IIID using ...
    There is currently no cure or effective treatment available for mucopolysaccharidosis type IIID (MPS IIID, Sanfilippo syndrome type D), a lysosomal storage ...
  53. [53]
    Intrathecal enzyme replacement therapy: Successful treatment of ...
    We studied whether application of high concentrations of therapeutic enzymes via intrathecal (IT) injections could successfully drive uptake across the ...
  54. [54]
    Subcutaneous Administration of Biotherapeutics - PMC - NIH
    Subcutaneous administration has proven effective, safe, well-tolerated, generally preferred by patients and healthcare providers.
  55. [55]
    Phase II study results of a replacement therapy for hereditary ...
    Subcutaneous (SC) administration of C1‐INH offers several potential advantages including easier access for self‐administration and more consistent PKs. The ...
  56. [56]
    Safety of enzyme replacement therapy - Fabry Disease - NCBI - NIH
    Premedication, generally with paracetamol, oral antihistamines and/or corticosteroids given 1–24 hours before the infusion, has prevented subsequent reactions ...
  57. [57]
    The use of port-a-caths in adult patients with Lysosomal Storage ...
    Oct 21, 2017 · Port-a-cath is a widely used device in patients with long-term venous access demand such as frequent or continuous administration of medications ...
  58. [58]
    Findings - Enzyme-Replacement Therapies for Lysosomal Storage ...
    Reported ages ranged from 9 to 76 years with the length of follow-up ranging from 20, weeks up to 234 weeks. The ERT dose in all studies ranged from 0.2 mg/kg, ...
  59. [59]
    Glucosylsphingosine (Lyso-Gb1): An Informative Biomarker in the ...
    Nov 29, 2022 · Recently, glucosylsphingosine (lyso-Gb1), a sensitive and specific GD biomarker, has been recommended for patient monitoring. Furthermore, ...
  60. [60]
    Dose selection for biological enzyme replacement therapy indicated ...
    Two important considerations for successful dose selection include (1) identifying appropriate disease‐specific endpoints, including pharmacodynamic (PD) end ...Missing: candidacy | Show results with:candidacy
  61. [61]
    Effects of Enzyme Replacement Therapy and Antidrug Antibodies in ...
    The recommended dosage for agalsidase-α is 0.2 mg/kg intravenously every 2 weeks, and the advised dosage for agalsidase-β is 1.0 mg/kg intravenously every 2 ...
  62. [62]
    Glucose tetrasaccharide as a biomarker for monitoring the ... - PubMed
    A tetraglucose oligomer, Glcalpha1-6Glcalpha1-4Glcalpha1-4Glc, designated Glc4, has been shown to be a putative biomarker for the diagnosis of Pompe disease.Missing: parameters | Show results with:parameters<|separator|>
  63. [63]
    Low-dose Gene Therapy Reduces the Frequency of Enzyme ...
    Oct 25, 2016 · Reduction of urinary GAGs is a sensitive and reliable biomarker of lysosomal storage clearance and therapeutic efficacy in LSDs.Missing: parameters | Show results with:parameters
  64. [64]
    S3.1 Are there ERT defined guidelines for Pompe disease? - PMC
    Nowadays, general recommendations includes monitoring of limb muscles and respiratory functions every 6 months using timed tests and functional scales. Muscle ...
  65. [65]
    Biomarkers for nonclinical infusion reactions in marketed ...
    Nonclinical IRs after a first dose are associated with cytokine release. •. Nonclinical IRs after several doses are associated with ADA. •. ADA-mediated IRs ...
  66. [66]
    The effect of enzyme replacement therapy on bone crisis ... - PubMed
    ERT is associated with a reduction in bone crisis and bone pain in patients with GD type 1. This study shows that significant improvements in symptoms of ...Missing: initial | Show results with:initial
  67. [67]
    Table 6, Drugs potentially used for the treatment of ERT infusion ...
    Table 6Drugs potentially used for the treatment of ERT infusion-associated reactions ; Moderate, Acetaminophen Ibuprofen, Chlorpheniramine (IV), Hydrocortisone ( ...Missing: management | Show results with:management
  68. [68]
    Immune responses to alglucosidase in infantile Pompe disease - NIH
    Approximately 90% of IOPD patients treated with alglucosidase alfa develop IgG antibodies [5, 20], with a large majority developing immunological tolerance with ...Missing: prevalence | Show results with:prevalence<|control11|><|separator|>
  69. [69]
    Predicting the Development of Anti-Drug Antibodies against ... - MDPI
    Enzyme replacement therapy (ERT) with recombinant alpha-galactosidase A is available, but approximately 50% of male patients with classical FD develop ...Missing: prevalence | Show results with:prevalence
  70. [70]
    Immunogenicity of Biotherapeutics: Causes and Association with ...
    Immunogenic response to therapeutic molecules can generate anti-drug antibodies (ADAs), which can be either neutralizing or nonneutralizing. Neutralizing ...
  71. [71]
    Current status of the immunogenicity of enzyme replacement ...
    In patients with Fabry disease (FD), treatment with enzyme replacement therapy (ERT), may trigger the formation of anti-drug antibodies (ADAs).
  72. [72]
    effects of antibody formation on enzyme replacement therapy - Nature
    Jun 30, 2016 · In adults with Pompe disease, antibody formation does not interfere with rhGAA efficacy in the majority of patients, is associated with IARs, and may be ...
  73. [73]
    Advances in Immune Tolerance Induction in Enzyme Replacement ...
    Apr 25, 2024 · Strategies focusing on immunomodulation have shown promise in inducing immune tolerance to ERT, leading to improved overall survival rates.
  74. [74]
    Velaglucerase alfa enzyme replacement therapy compared with ...
    Dec 19, 2012 · Enzyme replacement therapy for Gaucher disease (GD) has been available since 1991. This study compared the efficacy and safety of ...Abstract · Methods · Results · Discussion
  75. [75]
    Enzyme Replacement or Substrate Reduction? A Review of ... - NIH
    Conclusion: Enzyme replacement therapy is preferred for treating type 1 Gaucher disease and substrate replacement therapy may be considered in patients who do ...
  76. [76]
    Hematopoietic stem cell transplantation for mucopolysaccharidoses
    HSCT is considered the standard of care for those with MPS IH and an optional treatment for Hurler/Scheie syndrome (MPS IH/S) and Scheie syndrome (MPS-IS) ( ...
  77. [77]
    Migalastat: A Review in Fabry Disease - PMC - PubMed Central
    Feb 26, 2019 · Migalastat is an important treatment option for Fabry disease in patients with migalastat-amenable GLA mutations.
  78. [78]
    Lysosomal Storage Disease - UF Health
    Jun 12, 2025 · Enzyme-replacement therapy (ERT) · Eight LSDs are treated with ERT – Gaucher disease, Fabry disease, mucopolysaccharidoses types I,II, IV and VI, ...What are lysosomal storage... · LSD treatmentMissing: targeted | Show results with:targeted
  79. [79]
    Mucopolysaccharidoses and the blood–brain barrier
    Sep 19, 2022 · One of the main disadvantages of this therapy is that the adult BBB, unlike the neonatal one, does not transport lysosomal enzymes into the ...
  80. [80]
    Enhancing CNS Entry of Systemically-Delivered AAV Vectors for ...
    Sep 30, 2019 · This project aims to lead to the development of safe and effective approaches that can enhance the neurological benefit of systemic AAV9 gene ...<|separator|>
  81. [81]
    Pegunigalsidase alfa: a novel, pegylated recombinant alpha ...
    Apr 12, 2024 · Produced in plant cells, pegunigalsidase alfa exhibits enhanced stability, prolonged half-life, and reduced immunogenicity due to pegylation. A ...Missing: enzymes longer
  82. [82]
    Pegunigalsidase alfa, a novel PEGylated enzyme replacement ...
    Mar 4, 2019 · Pegunigalsidase alfa, a novel PEGylated enzyme replacement therapy for Fabry disease, provides sustained plasma concentrations and favorable pharmacodynamics.Missing: deimmunized | Show results with:deimmunized
  83. [83]
    Current status of the immunogenicity of enzyme replacement ...
    May 26, 2025 · This article reviews different aspects of immunogenicity and its impact on the generation of neutralizing antibodies, which may compromise the efficacy of ERT ...
  84. [84]
    CRISPR/Cas9 technology in the modeling of and treatment of ...
    This review concentrates on the application of this technique in the treatment of MPS, particularly MPS I, and modeling of disease-causing mutations.
  85. [85]
    CRISPR/Cas-Based Ex Vivo Gene Therapy and Lysosomal Storage ...
    Emerging approaches aim to edit the genome by leveraging enzymes such as deaminases and reverse transcriptase in CRISPR/Cas-based systems, which are referred to ...
  86. [86]
    Phase I study of liver depot gene therapy in late-onset Pompe disease
    Feb 17, 2023 · This Phase I clinical trial of liver depot gene therapy for Pompe disease is the first report of systemic gene therapy to treat Pompe disease. A ...
  87. [87]
    Current avenues of gene therapy in Pompe disease - PMC
    While enzyme replacement therapy has dramatically changed the outcome of patients with the disease, this strategy has several limitations. Gene therapy in Pompe ...
  88. [88]
    (PDF) A Feasibility Open-Labeled Clinical Trial Using a Second ...
    Jun 5, 2024 · Results: The second-generation AI-based personalized regimen was associated with stable responses to ERT in patients with GD1. The SF-36 quality ...
  89. [89]
    Enzyme Replacement Therapy Market Size & Outlook, 2025-2033
    The high cost also makes it difficult for patients in lower-income regions to access these therapies, hindering global market penetration. As a result ...
  90. [90]
    In the Press - Chiesi Global Rare Diseases
    Sep 9, 2025 · In ERT-switch patients, scores remained stable, with a slight trend toward improvement by month 24. All MSSI domains showed positive changes as ...
  91. [91]