Biotech
875125-19-2
Nomacopan is an investigational, second-generation biopharmaceutical agent representing a novel class of immunomodulators with a unique, dual-action mechanism. Originating from a protein found in the saliva of the Ornithodoros moubata tick, this recombinant small peptide is engineered to simultaneously inhibit two distinct and potent inflammatory pathways: the terminal complement component C5 and the lipid mediator leukotriene B4 (LTB4). This bifunctional activity distinguishes it from all currently approved complement inhibitors and forms the basis of its therapeutic rationale across a wide spectrum of autoimmune and inflammatory diseases.
Pharmacologically, Nomacopan prevents the cleavage of C5, thereby blocking the generation of the pro-inflammatory anaphylatoxin C5a and the formation of the cell-lysing Membrane Attack Complex (MAC). Concurrently, it employs a unique "ligand capture" mechanism to sequester and neutralize LTB4, a powerful chemoattractant for neutrophils. This combined blockade offers a more comprehensive approach to suppressing inflammation than targeting either pathway alone.
The clinical development of Nomacopan, led by Akari Therapeutics, has been characterized by a strategic evolution. Initial investigations demonstrated significant efficacy and a favorable safety profile in Paroxysmal Nocturnal Hemoglobinuria (PNH) and the autoimmune blistering skin disease Bullous Pemphigoid (BP). However, a portfolio reprioritization has shifted the company's focus toward indications with the highest unmet medical need and a clearer path to market. The lead clinical programs are now in Hematopoietic Stem Cell Transplant-Associated Thrombotic Microangiopathy (HSCT-TMA), a rare and often fatal condition with no approved treatments, and Geographic Atrophy (GA), a leading cause of blindness.
To overcome the short half-life of the parent molecule, a long-acting version, PAS-nomacopan, has been developed using PASylation® technology. This next-generation asset is being advanced for GA, where its potential for infrequent intravitreal injections (up to every three months) and its unique ability to potentially mitigate the risk of choroidal neovascularization (a side effect of other complement inhibitors) position it as a potential best-in-class therapy.
Across more than 30 cumulative patient-years of clinical exposure, Nomacopan has consistently been well-tolerated, with no drug-related serious adverse events reported in key trials. Manufacturing has been significantly optimized through a partnership with Wacker Biotech, whose proprietary ESETEC® technology has increased production yields by up to 12-fold, ensuring commercial viability. The program is strongly supported by global regulatory agencies, having received numerous Orphan Drug, Fast Track, and Rare Pediatric Disease designations from the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This comprehensive regulatory support, combined with a robust scientific foundation and a focused clinical strategy, positions Nomacopan as a significant therapeutic candidate with the potential to redefine the standard of care in several severe diseases.
This section establishes the fundamental identity of Nomacopan, detailing its nomenclature, classification, biological origin, and molecular structure.
Nomacopan is a biopharmaceutical drug, classified as a 'Biotech' type and categorized as a Recombinant Protein Inhibitor of Complement Factor 5.[1] The drug's identity is formally established through several key identifiers that are used globally in scientific literature, regulatory filings, and chemical databases.
The progression of its nomenclature reflects a typical maturation process for a biopharmaceutical asset, moving from descriptive research names to a formally adopted nonproprietary name. Its earliest identifiers were descriptive of its origin and function, such as "recombinant Ornithodoros moubata complement inhibitor" or "recombinant OmCI".[2] As it entered formal development, it was assigned codes like rEV576 and AK576.[2] For a period, it was also known by the proposed U.S. brand name Coversin.[2] The official International Nonproprietary Name (INN) and United States Adopted Name (USAN) is Nomacopan, a designation that signifies its status as a mature clinical candidate undergoing late-stage evaluation.[4]
| Identifier/Property | Details | Source(s) |
|---|---|---|
| Generic Name | Nomacopan | 1 |
| DrugBank ID | DB14991 | 1 |
| CAS Number | 875125-19-2 | 1 |
| UNII | 79V7Q9VIWQ | 1 |
| Type | Biotech; Recombinant Protein | 1 |
| Molecular Formula | $C_{720}H_{1107}N_{197}O_{244}S_{11}$ | 8 |
| Molecular Weight | 16779.41 Da (Average) | 8 |
| Origin | Recombinant protein derived from Ornithodoros moubata (soft tick) saliva | 2 |
| Key Synonyms | Coversin, rEV576, recombinant OmCI, AK576, Protein rEV576 | 2 |
Nomacopan is a compelling example of nature-inspired drug discovery. It is a recombinant small protein originally derived from a natural protein discovered in the saliva of the Ornithodoros moubata tick.[2] This tick, a parasite, evolved this protein as a sophisticated defense mechanism to modulate the host's immune and inflammatory responses, allowing it to feed for extended periods without detection or provoking an immune attack.[7] This evolutionary pre-optimization has resulted in a molecule with high potency and specificity for its human targets.
For clinical and commercial purposes, Nomacopan is produced using recombinant DNA technology in an Escherichia coli expression system.[4] The choice of an E. coli production platform for a non-glycosylated protein of this size offers significant manufacturing advantages, including faster production cycles and potentially lower costs compared to more complex mammalian cell culture systems required for larger biologics like monoclonal antibodies.
Structurally, Nomacopan is a single-chain polypeptide with a molecular weight of approximately 16,740 Da.7 Its complete primary structure consists of a sequence of 150 amino acids 4:
DSESDCTGSE PVDAFQAFSE GKEAYVLVRS TDPKARDCLK GEPAGEKQDN TLPVMMTFKN GTDWASTDWT FTLDGAKVTA TLGNLTQNRE VVYDSQSHHC HVDKVEKEVP DYEMWMLDAG GLEVEVECCR QKLEELASGR NQMYPHLKDC
The protein's tertiary structure is stabilized by three intramolecular disulfide bridges, which are critical for its proper folding and biological activity. These bridges link cysteine residues at the following positions: 6 to 128, 38 to 150, and 100 to 129.[4]
The therapeutic potential of Nomacopan is rooted in its unique and innovative pharmacological profile. Unlike other complement-modulating therapies, it is a bifunctional inhibitor that concurrently targets two separate but often interconnected inflammatory pathways.
Nomacopan's primary differentiator is its dual mechanism of action, simultaneously inhibiting the terminal complement protein C5 and the pro-inflammatory lipid mediator leukotriene B4 (LTB4).[9] These two pathways are central drivers of inflammation and tissue damage in a host of autoimmune and inflammatory diseases. The pathways can be activated independently by different triggers, meaning that blockade of one may be insufficient to fully control the pathological process.[10] By targeting both, Nomacopan offers a more comprehensive and potentially more potent anti-inflammatory effect with a single molecular entity.
This dual functionality is not an artificial construct but an evolutionary adaptation. Phylogenetic analysis suggests that the ancestral protein first evolved the ability to bind and inhibit LTB4, and later acquired the C5-inhibiting function.[11] This indicates a strong selective advantage for the tick in simultaneously suppressing both of these co-located inflammatory mediators at the site of feeding, a principle that has been translated into a therapeutic strategy.[7] This dual blockade is the scientific rationale for investigating Nomacopan in diseases where both C5 and LTB4 are implicated, such as Bullous Pemphigoid and traumatic injury.[12]
Nomacopan acts at the final, critical step of the complement cascade. It targets and binds to a unique site on the complement C5 protein.[2] This physical interaction sterically hinders the C5 convertase enzyme, blocking the cleavage of C5 into its two potent effector fragments:
By preventing the generation of C5b, Nomacopan effectively halts the formation of the Membrane Attack Complex (MAC), a pore-forming structure (composed of C5b-9) that inserts into cell membranes and causes direct cell lysis and tissue destruction.[2]
This mechanism is comprehensive, as it prevents the formation of both the key inflammatory mediator (C5a) and the lytic effector (MAC). This is a critical distinction from other classes of complement modulators. For instance, a C5a receptor antagonist would block C5a-mediated inflammation but would have no effect on MAC-driven cell destruction, which is the primary pathology in diseases like Paroxysmal Nocturnal Hemoglobinuria (PNH).[2] Nomacopan's ability to block both arms of the terminal complement pathway underpins its efficacy in both inflammatory and hemolytic complement-mediated disorders.
In addition to its action on the complement system, Nomacopan independently inhibits LTB4, a potent lipid mediator derived from arachidonic acid that plays a crucial role in initiating and amplifying inflammatory responses, primarily by recruiting and activating neutrophils.[2]
Nomacopan's mode of LTB4 inhibition is distinct and highly specific. It functions via a "ligand capture" mechanism, whereby the LTB4 fatty acid molecule is tightly bound and sequestered within a hydrophobic cavity inside the protein's structure ($K_D$ 0.2nM).[9] This action effectively removes LTB4 from the biological milieu, acting as a molecular sponge. By depleting free LTB4, Nomacopan disrupts the chemotactic gradient that draws inflammatory cells to sites of injury and prevents LTB4 from activating its G protein-coupled receptors, BLT1 and BLT2, on the surface of these cells.[10]
This ligand-capture approach offers potential advantages over other strategies for targeting the LTB4 pathway, such as receptor antagonists or synthesis inhibitors. By physically removing the ligand, it ensures a complete blockade of signaling through both of its receptors. Furthermore, it avoids potential off-target effects associated with synthesis inhibitors, such as the shunting of arachidonic acid metabolism towards the production of other pro-inflammatory molecules like prostanoids.[11] The clinical importance of this second mechanism is underscored by preclinical studies in which an engineered version of Nomacopan that only blocks LTB4 was shown to be effective in an animal model of pemphigoid disease.[14]
The physical and pharmacokinetic properties of Nomacopan have been pivotal in shaping its clinical application and have driven the strategic development of a next-generation, long-acting formulation to expand its therapeutic potential.
Nomacopan is a relatively small recombinant protein with a molecular weight of 16.7 kDa.[7] Its biophysical characteristics lend themselves to a variety of formulation and delivery options, including subcutaneous, intravenous, topical, and inhaled routes of administration.[7] This versatility provides a platform for potential future development in a range of localized diseases.
In its clinical development to date, the primary route of administration has been subcutaneous injection.[17] The ability for patients to self-administer the drug subcutaneously at home is a significant advantage in the management of chronic diseases, offering greater convenience and reducing the healthcare burden compared to therapies requiring regular intravenous infusions in a clinical setting, such as the first-generation C5 inhibitor eculizumab.[15]
A key pharmacokinetic characteristic of the parent Nomacopan molecule is its very short plasma half-life when it is not bound to its target, C5.[21] This property is typical for small proteins that are subject to rapid renal clearance. Consequently, to maintain therapeutically effective concentrations and ensure continuous blockade of the C5 and LTB4 pathways, frequent dosing is required.[21]
Clinical trials have primarily employed once-daily subcutaneous dosing regimens.[18] The Phase 3 trial in Bullous Pemphigoid (ARREST-BP) was designed to evaluate two different once-daily doses: 15 mg and 45 mg.[17] This dose-ranging was informed by pharmacokinetic modeling, which predicted that these regimens would achieve at least 90% inhibition of terminal complement activity in 75% and 98% of patients, respectively.[22] While daily self-injection is an improvement over intravenous therapy, the need for frequent administration represents a pharmacokinetic liability that can impact long-term patient adherence. This limitation was the primary impetus for developing a long-acting version of the drug.
To address the short half-life of the parent molecule and unlock new therapeutic opportunities, Akari Therapeutics has developed PAS-nomacopan, a long-acting formulation engineered using PASylation® technology.[23] This technology, developed by XL-protein, involves the genetic fusion of a long, unstructured polypeptide composed of Proline, Alanine, and Serine (PAS) residues to the therapeutic protein.[21]
In the case of PAS-nomacopan, a 600-residue PAS polypeptide is conjugated to the N-terminus of the Nomacopan molecule.[21] This modification significantly increases the effective size and hydrodynamic volume of the drug, which in turn slows its rate of clearance via renal filtration.[21] This PEG-like effect dramatically extends the drug's plasma half-life. Preclinical pharmacokinetic studies of PAS-nomacopan administered via intravitreal (IVT) injection in rabbits demonstrated a half-life in the vitreous and retina of 5-6 days.[25] This data supports modeling that suggests a potential clinical dosing interval of three months or longer in humans for the treatment of retinal diseases like Geographic Atrophy (GA).[23]
The development of PAS-nomacopan represents a pivotal strategic evolution for the program. It transforms the asset from a daily-dosed therapy for systemic diseases into a potential best-in-class treatment for chronic, localized conditions where infrequent administration is a major clinical and commercial differentiator.
The clinical development of Nomacopan has spanned multiple therapeutic areas, reflecting the broad pathogenic roles of the complement and leukotriene pathways. The program's history reveals a strategic evolution, marked by initial broad exploration followed by a deliberate focusing of resources on indications with the highest unmet need and clearest path to regulatory approval.
Nomacopan has been evaluated in a diverse range of complement- and LTB4-mediated diseases. Major clinical programs have been conducted in Paroxysmal Nocturnal Hemoglobinuria (PNH), Bullous Pemphigoid (BP), and various Thrombotic Microangiopathies (TMA), including the high-priority indication of Hematopoietic Stem Cell Transplant-Associated TMA (HSCT-TMA).[13] Early-phase studies also explored its use in allergic and drug-induced keratoconjunctivitis.[1]
A notable feature of the development history is the termination or withdrawal of several Phase 3 trials. Importantly, these decisions were not based on safety or efficacy concerns but were described as strategic business decisions resulting from a portfolio reprioritization.[28] This pattern indicates a calculated pivot away from more competitive or challenging development areas toward indications where Nomacopan has the greatest potential to become a first-in-class or best-in-class therapy. The current late-stage focus is on pediatric HSCT-TMA and, with the long-acting formulation, Geographic Atrophy.[29]
| NCT Identifier | Indication | Phase | Status | Key Outcome / Rationale for Status |
|---|---|---|---|---|
| NCT03829449 | Paroxysmal Nocturnal Hemoglobinuria (PNH) | 3 | Terminated | The CAPSTONE trial was terminated. Efficacy in reducing transfusion dependence was shown in earlier and long-term studies.20 |
| NCT04035733 | Bullous Pemphigoid (BP) | 2a | Completed | Demonstrated good safety and therapeutic potential in 9 elderly patients, meeting primary and secondary endpoints.32 |
| NCT05061771 | Bullous Pemphigoid (BP) | 3 | Withdrawn | The ARREST-BP trial was withdrawn prior to completion as part of a portfolio reprioritization.1 |
| NCT04784455 | HSCT-Associated Thrombotic Microangiopathy (Pediatric) | 3 | Terminated | Terminated due to a business decision for portfolio reprioritization. Positive case study data emerged from the trial.28 |
PNH, a rare blood disorder characterized by MAC-mediated destruction of red blood cells, was one of the first major indications pursued for Nomacopan. Its efficacy was evaluated in the Phase 2 COBALT trial and the Phase 3 CAPSTONE trial (NCT03829449).[19] Patients from these studies were eligible to enroll in the CONSERVE long-term safety and efficacy study.[3]
The clinical data generated in PNH were strong. Across studies involving 19 patients and over 30 cumulative patient-years of treatment, self-administered subcutaneous Nomacopan demonstrated a substantial reduction in transfusion dependence, a key clinical endpoint.[20] In a long-term analysis of a cohort of previously transfusion-dependent patients, all six (100%) achieved and maintained transfusion independence for six months or more.[3] Throughout treatment, patients showed complete inhibition of terminal complement activity and the drug was well tolerated.[3]
Despite these positive results, the Phase 3 PNH program was discontinued. This was likely a commercial and strategic decision rather than a clinical one. The PNH market is well-established and highly competitive, dominated by long-acting C5 inhibitors like ravulizumab, which offers dosing every eight weeks.[27] Competing as a new entrant with a daily-dosed therapy, even one with strong efficacy, would have been a significant commercial challenge. Akari appears to have strategically reallocated its resources to orphan diseases with no approved therapies, where the unmet need is higher and the path to market is clearer.
BP is a severe autoimmune blistering skin disease of the elderly, in which both C5 and LTB4 are known to be key pathogenic drivers.[12] This provided a strong scientific rationale for the use of a dual inhibitor like Nomacopan.
A multicenter, single-arm Phase 2a trial (NCT04035733) was conducted in nine patients (median age 75) with mild-to-moderate BP.[18] The results were highly encouraging. The trial met its primary safety endpoint, with no drug-related serious adverse events reported.[33] On efficacy measures, 7 of the 9 patients (77.8%) responded to treatment, showing a clinically significant reduction in the Bullous Pemphigoid Disease Area Index (BPDAI) activity score over 42 days. Three of these responders experienced a reduction of 80% or greater, nearing complete remission.[18]
Based on this positive data, a Phase 3 study, ARREST-BP (NCT05061771), was initiated.[17] However, this trial was later withdrawn as part of the company's broader portfolio review.[1] While the clinical program in BP is currently on hold, the strong proof-of-concept data remains a valuable asset. Akari has secured intellectual property for this use, with the European Patent Office granting a patent for Nomacopan in the treatment of autoimmune blistering diseases, preserving the opportunity for future development or partnership.[30]
HSCT-TMA has emerged as Akari's lead clinical indication and represents the company's most focused effort toward securing a first regulatory approval. HSCT-TMA is a severe and life-threatening complication of stem cell transplantation with no approved treatments and a mortality rate approaching 80% in severe cases.[39] This high unmet medical need makes it an ideal target for an orphan drug development program.
An initial Phase 3 study in pediatric HSCT-TMA (NCT04784455) was initiated but later terminated as part of the portfolio reprioritization.[28] However, the trial provided valuable early proof-of-concept. A case study of a 6-year-old patient with severe HSCT-TMA treated in the study was presented at major medical congresses. The patient, treated with Nomacopan, experienced resolution of gut pathology and thrombocytopenia and was ultimately discharged from the hospital, with no adverse events related to the drug.[36]
Building on this experience and extensive regulatory engagement, Akari is advancing a refined late-stage clinical program. The company is proceeding with a new registrational Phase 3 open-label, single-arm study in pediatric HSCT-TMA, with enrollment expected to begin by the end of 2023.[29] In parallel, a Phase 3 double-blind, placebo-controlled clinical trial in adult HSCT-TMA is planned to begin enrollment in 2024.[29] This focused, dual-population approach in an area of dire unmet need, backed by robust regulatory support, represents the core of the company's current clinical strategy.
The versatile, dual-action mechanism of Nomacopan has prompted research across several other therapeutic areas.
A comprehensive evaluation of safety data from across the Nomacopan clinical development program reveals a consistently favorable and manageable safety profile, which is a cornerstone of its value proposition, particularly in the treatment of frail and vulnerable patient populations.
Nomacopan has been administered to patients across multiple trials, accumulating over 30 cumulative patient-years of safety data.[20] A consistent finding across all studies has been that the drug is well tolerated.
The most robust long-term safety data comes from the PNH program, which included the Phase 2 COBALT trial, the Phase 3 CAPSTONE trial, and the long-term CONSERVE extension study.[20] In an analysis of 19 PNH patients treated for a median of 18.5 months, there were no reported major adverse vascular events (MAVEs), a critical safety consideration in this patient population. Only a single serious adverse event (a urinary tract infection) was considered possibly related to Nomacopan.[20]
Similarly, the Phase 2a study in nine elderly patients (median age 75) with Bullous Pemphigoid, a population with significant comorbidities, met its primary safety endpoint.[18] There were no Common Terminology Criteria for Adverse Events (CTCAE) grade 3, 4, or 5 adverse events that were deemed by investigators to be related or possibly related to Nomacopan treatment.[18] This strong safety record in a frail population is a significant de-risking factor for the drug's continued development.
| Adverse Event (AE) | Indication | Frequency / No. of Patients | Severity | Investigator-Assessed Relationship to Drug | Source(s) |
|---|---|---|---|---|---|
| Serious AEs | |||||
| Urinary Tract Infection | PNH | 1 of 19 | Serious | Possibly Related | 20 |
| Localized infection (knee) | BP | 1 of 9 | Severe | No Association | 18 |
| Condition reaggravated (BP) | BP | 2 of 9 | Moderate to Severe | No Association | 18 |
| Most Frequent AEs | |||||
| Injection site reactions | PNH | Commonly reported | Mild, self-limiting | Related | 19 |
| Urinary Tract Infection (UTI) | BP | 3 of 9 (33.3%) | Mild | No Association | 18 |
| Peripheral edema | BP | 2 of 9 (22.2%) | Not specified | No Association | 18 |
| Pruritus | BP | 2 of 9 (22.2%) | Not specified | No Association | 18 |
| Headache | BP | 2 of 9 (22.2%) | Not specified | No Association | 18 |
The overall adverse event profile of Nomacopan appears benign. The most frequently reported AEs have been mild and generally consistent with events expected in the underlying patient populations. In the PNH trials, where patients self-administered the drug subcutaneously, the most common AEs were mild and transient injection site reactions.[19]
In the Phase 2a BP trial, several non-serious AEs were reported in more than one patient, including urinary tract infections (3 patients), reaggravation of BP (2 patients), peripheral edema (2 patients), pruritus (2 patients), and headache (2 patients).[18] However, after careful review, the clinical investigators determined that none of these events had an association with Nomacopan treatment.[18] This suggests that the dual inhibition of C5 and LTB4 does not appear to introduce novel or unexpected toxicities beyond the known risks of the drug class.
The inhibition of the terminal complement pathway is a well-established therapeutic strategy, but it carries a known class-specific risk: an increased susceptibility to infections caused by encapsulated bacteria, most notably Neisseria meningitidis.[15] The MAC is a primary defense mechanism against such organisms, and its blockade can leave patients vulnerable.
The clinical development program for Nomacopan has incorporated standard and robust risk mitigation strategies to address this. Protocols for clinical trials consistently require that participants either have up-to-date immunizations against N. meningitidis or be willing to receive vaccination and/or prophylactic antibiotic therapy for the duration of treatment.[12] This proactive risk management has proven effective. Across all clinical studies to date, there have been no reported cases of meningococcal infection in patients treated with Nomacopan.[20] This demonstrates that while the risk is real, it is manageable with appropriate medical oversight, which is a critical factor for the drug's overall risk-benefit assessment.
The successful development of a biologic therapy depends not only on its clinical performance but also on the ability to manufacture it reliably, consistently, and cost-effectively. The manufacturing strategy for Nomacopan has been a key area of focus, characterized by strategic partnerships and significant technological advancements to ensure commercial viability.
Nomacopan is a recombinant protein produced in a microbial expression system.[4] Its manufacture is managed through a long-term partnership with Wacker Biotech GmbH, a leading contract development and manufacturing organization (CDMO).[46] The production process leverages Wacker's proprietary ESETEC® expression technology.[47]
The ESETEC® platform is based on engineered strains of E. coli K12 that have been modified to secrete correctly folded recombinant proteins directly into the culture medium during fermentation.[24] This is a significant advantage over conventional intracellular E. coli expression systems, where proteins often accumulate inside the cell as insoluble, misfolded aggregates (inclusion bodies). Such systems require complex and costly downstream processing steps, including cell disruption (homogenization), solubilization, and protein refolding, which can be inefficient and lead to lower final yields. The ESETEC® secretory system bypasses these steps, greatly simplifying the purification process and making the entire manufacturing workflow more efficient and cost-effective.[49] This technology is used for both the parent Nomacopan and the long-acting PAS-nomacopan.[48]
A critical milestone in the Nomacopan program has been the substantial improvement of its manufacturing process. Through continuous investment and optimization of the ESETEC® technology, including the development of new bacterial strains, Wacker and Akari have achieved a dramatic increase in production yield.
Reports indicate that a "third generation" manufacturing process has increased the final yield of Nomacopan by at least 5-fold compared to the previous process.[30] Another announcement highlighted a remarkable 12-fold increase in yield, while comparative studies confirmed that the quality and purity of the protein remained consistently high.[47]
This multi-fold increase in yield is a transformative development. It significantly lowers the projected commercial cost of goods (COGS), which enhances the drug's potential profitability and market competitiveness. Crucially, Akari has secured agreement from major regulatory bodies—including the U.S. FDA, the U.K.'s MHRA, and Poland's URPL—to use this new, higher-yielding process in its pivotal clinical studies.[46] This is a strategically important step, as it ensures that the drug submitted for marketing approval will be the one produced by the most efficient and commercially scalable process, thereby avoiding the need for costly and time-consuming post-approval manufacturing changes.
Akari Therapeutics operates on a capital-efficient model, leveraging the expertise of specialized external partners rather than building in-house manufacturing capabilities. This strategy is central to its ability to advance a complex biologic through late-stage development.
The cornerstone of this strategy is the partnership with Wacker Biotech GmbH, which serves as the CDMO for both Nomacopan and PAS-nomacopan.[23] This relationship provides Akari with access to the state-of-the-art ESETEC® technology and Wacker's extensive experience in GMP-compliant manufacturing of therapeutic proteins.[47]
The development of the long-acting PAS-nomacopan also involves a key technology partnership with XL-protein GmbH, the German biotech company that developed and commercializes the PASylation® platform.[24] By licensing this technology, Akari was able to efficiently create a next-generation version of its lead asset with a vastly improved pharmacokinetic profile. These strategic partnerships are integral to the Nomacopan program, providing the technological foundation and manufacturing capacity necessary for clinical trials and potential commercialization.
The development pathway of Nomacopan has been significantly shaped and supported by extensive engagement with global regulatory authorities. The numerous special designations it has received underscore its potential to address serious and life-threatening diseases with high unmet medical needs, while the company's clinical strategy points to a clear and focused path forward.
Nomacopan has been granted an impressive array of special designations by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA). These designations are awarded to promising therapies for rare or serious conditions and provide significant incentives, including scientific support from regulators, extended market exclusivity, and expedited review pathways. The breadth of these designations highlights a strong regulatory consensus on the drug's potential value.
| Indication | Agency | Designation | Source(s) |
|---|---|---|---|
| HSCT-TMA | FDA | Orphan Drug Designation | 4 |
| FDA | Fast Track Designation | 29 | |
| FDA | Rare Pediatric Disease Designation | 29 | |
| EMA | Orphan Drug Designation (Positive Opinion) | 29 | |
| Bullous Pemphigoid | FDA | Orphan Drug Designation | 4 |
| FDA | Fast Track Designation | 53 | |
| EMA | Orphan Drug Designation | 53 | |
| Guillain-Barre Syndrome | FDA | Orphan Drug Designation | 4 |
| Paroxysmal Nocturnal Hemoglobinuria | EMA | Orphan Drug Designation | 4 |
This robust collection of designations provides a significant strategic advantage. Orphan Drug status grants 7 years (FDA) and 10 years (EMA) of market exclusivity post-approval. Fast Track designation facilitates more frequent communication with the FDA and allows for a rolling review of the marketing application. The Rare Pediatric Disease designation for pediatric HSCT-TMA makes Akari eligible to receive a Priority Review Voucher upon approval, which can be used to expedite the review of a future drug or be sold to another company for a substantial sum.
Following a period of broad clinical exploration, Akari Therapeutics has refined its strategy to focus on a "twin-pillar" approach that balances near-term opportunities with long-term, high-value potential.
Nomacopan is a highly differentiated biopharmaceutical asset defined by its novel, dual-action mechanism targeting both complement C5 and leukotriene B4. Its journey through clinical development has been marked by a strategic pivot from competitive markets to areas of profound unmet medical need, demonstrating a disciplined and focused approach by its developer. The drug has consistently shown a favorable safety profile across multiple studies and in vulnerable patient populations. Its efficacy has been demonstrated in several indications, providing strong proof-of-concept for its unique mechanism.
Critical advancements in manufacturing have dramatically improved production yields, solidifying the drug's commercial viability. This technical progress, combined with a well-executed regulatory strategy that has secured numerous valuable designations from the FDA and EMA, has significantly de-risked the path forward.
While regulatory and clinical hurdles remain, the program's current trajectory is clear and compelling. The focus on securing a first approval in the orphan indication of HSCT-TMA, coupled with the ambitious development of a potentially best-in-class, long-acting formulation for the mass-market indication of Geographic Atrophy, represents a sophisticated strategy to maximize the asset's value. Nomacopan stands as a significant therapeutic candidate, poised with the potential to offer a new standard of care for patients with severe and life-threatening inflammatory diseases.
Published at: October 24, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.