NAPA Human

N-Ethylmaleimide-Sensitive Factor Attachment Protein, Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

NAPA Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 315 amino acids (1-295) and having a molecular mass of 35.3 kDa. NAPA is fused to 20 amino acid His Tag at N-terminus and purified by standard chromatography techniques.

Product Specs

Introduction
NAPA, a member of the Soluble NSF Attachment Protein (SNAP) family, plays a crucial role in membrane fusion processes. SNAPs, along with SNAP receptors (SNAREs) and N-ethylmaleimide-sensitive fusion protein (NSF), are essential for fusing transport vesicles with target membranes. This fusion process is vital for various cellular activities, including synaptic transmission (communication between nerve cells), intra-Golgi transport (within the Golgi apparatus), endosome-to-endosome fusion, and the transport of transcytotic vesicles to the plasma membrane. NAPA is specifically responsible for binding to NSF, leading to the formation of a 20S fusion particle, a complex crucial for membrane fusion.
Description
Recombinant human NAPA, expressed in E. coli, is a single polypeptide chain devoid of glycosylation. It comprises 315 amino acids, encompassing residues 1 to 295, and has a molecular weight of 35.3 kDa. A 20 amino acid Histidine tag is attached to the N-terminus of the protein to facilitate purification by standard chromatography techniques.
Physical Appearance
The product is a clear, colorless, and sterile filtered solution.
Formulation
The NAPA protein is supplied in a solution containing 1mg/ml of protein in a buffer composed of 20mM Tris-HCl at pH 7.5 and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. To ensure stability during long-term storage, adding a carrier protein such as 0.1% HSA (human serum albumin) or BSA (bovine serum albumin) is advised. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the NAPA protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
SNAPA, SNAP-alpha.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MDNSGKEAEA MALLAEAERK VKNSQSFFSG LFGGSSKIEE ACEIYARAAN MFKMAKNWSA AGNAFCQAAQ LHLQLQSKHD AATCFVDAGN AFKKADPQEA INCLMRAIEI YTDMGRFTIA AKHHISIAEI YETELVDIEK AIAHYEQSAD YYKGEESNSS ANKCLLKVAG YAALLEQYQK AIDIYEQVGT NAMDSPLLKY SAKDYFFKAA LCHFCIDMLN AKLAVQKYEE LFPAFSDSRE CKLMKKLLEA
HEEQNVDSYT ESVKEYDSIS RLDQWLTTML LRIKKTIQGD EEDLR.

Q&A

What are NAPA compounds and how do they affect human cytomegalovirus (HCMV)?

NAPA (N-arylpyrimidinamine) compounds represent a novel class of antiviral agents specifically developed to target human cytomegalovirus (HCMV) infection. These compounds have been identified through high-content screening methods and demonstrate potent inhibitory effects against HCMV by targeting the early stages of the viral life cycle . Unlike conventional anti-HCMV drugs that typically target viral DNA polymerase, NAPA compounds act on previously unexploited targets in the viral infection process . These compounds have demonstrated efficacy against both ganciclovir-sensitive and ganciclovir-resistant HCMV strains, with selectivity indices greater than 30, indicating a favorable therapeutic window . NAPA compounds exhibit specificity for cytomegaloviruses and do not show significant activity against other herpesviruses, suggesting a unique mechanism of action that could complement existing therapies .

How were NAPA compounds initially identified as potential anti-HCMV agents?

The discovery of NAPA compounds as anti-HCMV agents stemmed from a systematic high-content screening (HCS) approach designed specifically to identify inhibitors of early-stage viral infection. Researchers employed a reporter virus system (AD169 IE2/YFP) that expresses an IE2:yellow-fluorescent protein fusion protein, allowing for visualization of early viral gene expression . This screening method evaluated over 112,000 small molecules from various commercial compound collections, including those from MicroSource Discovery, Chembridge, Biomol GmbH, Life Chemicals, ChemDiv, and TimTec . Compounds were considered "primary hits" if they reduced YFP-positive nuclei by ≥70% while minimally affecting total cell numbers (≤25% reduction in H33342-stained nuclei), indicating specific antiviral activity rather than general cytotoxicity . Through this screening funnel, researchers identified NAPA compounds as particularly promising candidates, with the lead compound MBXC-4302 demonstrating an IC50 of 3.2μM and minimal cytotoxicity (CC50 >100μM) .

What experimental models are used to evaluate NAPA compounds' efficacy against HCMV?

The evaluation of NAPA compounds involves multiple experimental models designed to assess different aspects of antiviral efficacy. Initial screening utilizes reporter virus systems (AD169 IE2/YFP) that express fluorescent markers upon infection, enabling high-throughput quantification of viral inhibition in cell culture models . Dose-response assays employ two-fold compound dilution series (typically 0-100 μM) to determine the half-maximal inhibitory concentration (IC50) using four-parameter curve-fitting algorithms . Researchers evaluate NAPA efficacy in physiologically relevant cell types, including human dermal fibroblasts (NHDF) and epithelial cells (ARPE-19), to ensure activity across different cellular contexts where HCMV can replicate . Advanced studies include testing against diverse HCMV strains, including clinical isolates and ganciclovir-resistant variants, to assess broad-spectrum activity . Combination studies with established antivirals like ganciclovir employ methods to detect additive or synergistic effects, providing insights into potential therapeutic combinations . Each experimental approach typically includes at least six technical replicates to ensure statistical reliability and reproducibility .

What structural features define NAPA compounds and distinguish them from other antiviral agents?

NAPA compounds possess a distinctive N-arylpyrimidinamine scaffold that can be conceptually divided into five structural zones, each contributing differently to the compounds' antiviral activity and pharmacological properties . The core structure features a pyrimidine ring connected to an aryl group through an amine linkage, with various substitutions possible at multiple positions . Zone 1 typically contains heteroaromatic or aromatic groups that influence potency and selectivity, with hydrophobic moieties generally enhancing antiviral activity . Zone 2 encompasses the pyrimidine moiety, where the positioning of nitrogen atoms affects selectivity, as evidenced by differing selectivity indices among isomeric pyrimidine ligands . Zone 4 accommodates linker modifications, with urea linkers improving potency compared to sulfone linkers, suggesting the importance of hydrogen-bonding capabilities in this region . Zone 5 appears highly sensitive to modifications, with changes from furan to thiophene or phenyl rings resulting in complete loss of activity . Unlike nucleoside analogs (e.g., ganciclovir) that target viral DNA polymerase, NAPA compounds target early-stage infection processes, providing a mechanistically distinct approach to HCMV inhibition .

What structure-activity relationships (SAR) have been identified in the NAPA compound series?

Comprehensive structure-activity relationship studies of NAPA compounds have revealed critical insights into how structural modifications affect antiviral potency, selectivity, and pharmacological properties. Zone 1 modifications have demonstrated significant impact on compound efficacy, with the replacement of the heteroaromatic pyrazole (as in MBXC-4302) with aromatic secondary amines resulting in a 10-fold increase in potency . This suggests the presence of a hydrophobic binding pocket that interacts with this region of the molecule . Increasing lipophilicity in Zone 1 generally improved inhibitory potency, with piperidine substitutions restoring activity to sub-micromolar levels . The selectivity index (ratio of CC50 to IC50) varies considerably within Zone 1 analogs (ranging from 7 to >100), with tertiary amines demonstrating higher selectivity compared to phenyl analogs .

In Zone 2, the electronic properties of the pyrimidine moiety significantly influence selectivity. For instance, MBXC-4297 maintained a selectivity index >50, while isomeric pyrimidine ligands showed reduced selectivity (index <20) . Zone 4 modifications revealed that urea linkers enhanced potency while sulfone linkers were poorly tolerated, indicating a potential hydrogen-bonding site in this region of the binding pocket . Zone 5 appears particularly sensitive to structural changes, as substitution of the furan with either thiophene or phenyl rings completely abolished antiviral activity .

Analysis of physicochemical properties showed that basic amine-containing analogs demonstrate superior water solubility compared to other derivatives, while most compounds exhibited good stability in mouse liver microsome assays, suggesting favorable metabolic profiles .

How do NAPA compounds perform against ganciclovir-resistant HCMV strains and in combination therapy?

NAPA compounds demonstrate significant efficacy against ganciclovir-resistant HCMV strains, representing a critical advantage over existing therapies. Studies with lead NAPA analogs MBXC-4336 and MBX-4992 have shown that these compounds can effectively limit the proliferation of clinical HCMV isolates, including those resistant to ganciclovir . This activity against drug-resistant strains stems from NAPA compounds' distinct mechanism of action targeting early infection stages rather than viral DNA polymerase, which is the target of ganciclovir and related drugs .

In combination therapy studies, NAPA compounds exhibit additive or synergistic inhibition of HCMV proliferation when administered alongside ganciclovir . These synergistic effects likely result from the complementary mechanisms of action—NAPA compounds inhibiting early-stage infection processes while ganciclovir targets viral DNA replication . Such combination approaches are particularly valuable for clinical applications, as they may allow for lower doses of individual components, potentially reducing toxicity concerns while maintaining or enhancing therapeutic efficacy .

The efficacy against resistant strains and synergistic potential with existing therapies position NAPA compounds as promising candidates for addressing significant challenges in current HCMV treatment regimens, including drug resistance and dose-limiting toxicities .

What pharmacological properties of NAPA compounds influence their development potential?

The developmental potential of NAPA compounds is significantly influenced by their pharmacological properties, particularly those related to absorption, distribution, metabolism, excretion, and toxicity (ADMET) characteristics. Lead NAPA compounds demonstrate favorable in vitro ADME properties, supporting their further development as therapeutic agents . Solubility analyses show that basic amine-containing NAPA analogs exhibit superior water solubility compared to other derivatives, which is advantageous for formulation and bioavailability .

Cytotoxicity assessments using multiple cell lines and methodologies (MTS assay in HeLa cells and CellTiter Glo in NHDF cells) indicate that many NAPA analogs maintain high selectivity indices (>30), suggesting a wide therapeutic window between antiviral efficacy and cellular toxicity . Metabolic stability studies using mouse liver microsomes reveal that the majority of NAPA compounds exhibit good stability in the presence of NADPH, supporting their potential for sustained activity in vivo .

CYP450 3A4 inhibition studies provide insights into potential drug-drug interaction risks, an important consideration for compounds likely to be used in combination therapies or in patients receiving multiple medications . While specific CYP450 inhibition data is not fully detailed in the search results, these assays form a critical component of the evaluation funnel for NAPA compounds.

The combination of potent antiviral activity, favorable selectivity indices, good solubility profiles (particularly for amine-containing analogs), and metabolic stability positions certain NAPA analogs as promising candidates for further preclinical and potentially clinical development .

What is the precise mechanism by which NAPA compounds inhibit early-stage HCMV infection?

The precise molecular mechanism by which NAPA compounds inhibit early-stage HCMV infection remains an area of active investigation, though significant insights have been established. NAPA compounds specifically target early stages of the viral life cycle, as evidenced by their inhibition of immediate-early (IE) gene expression, particularly the IE1 and IE2 gene products that typically appear within 3-6 hours post-infection . These immediate-early proteins are critical for initiating the viral replication cascade, suggesting that NAPA compounds interfere with fundamental processes required for establishing productive infection .

Studies utilizing the AD169 IE2-YFP reporter virus system demonstrate that NAPA compounds reduce expression of the IE2-YFP chimera, which exhibits expression kinetics and nuclear localization similar to native IE2 . This indicates that the inhibitory effect occurs either during viral entry, nuclear transport of the viral genome, or during the earliest stages of viral gene expression .

The specificity of NAPA compounds for cytomegaloviruses, with no significant activity against other herpesviruses, suggests that they target virus-specific factors rather than broadly conserved herpesvirus components or general cellular processes . This specificity may involve interactions with unique viral proteins or virus-induced cellular pathways essential for HCMV but not for other herpesviruses .

While the precise molecular target(s) and binding interactions require further elucidation, structure-activity relationship studies provide insights into potential binding modes. The differential effects of modifications in the five structural zones of NAPA compounds suggest specific interactions with target binding pockets, including hydrophobic interactions (Zone 1), electronic effects (Zone 2), and hydrogen bonding capabilities (Zone 4) .

What experimental data supports NAPA compounds' broad-spectrum activity against diverse HCMV strains?

Experimental evidence strongly supports the broad-spectrum activity of NAPA compounds against diverse HCMV strains, a critical attribute for potential therapeutic applications. In comprehensive infectivity and proliferation studies, lead NAPA analogs MBXC-4336 and MBX-4992 effectively inhibited infection across various HCMV strains, demonstrating their broad antiviral spectrum . These compounds significantly prevented viral proliferation in both fibroblasts and epithelial cells, as measured by quantification of infected cells and viral genome levels .

The effectiveness of NAPA compounds extends beyond laboratory-adapted HCMV strains to include clinical isolates, which often better represent the viral diversity encountered in actual infections . Notably, NAPA compounds maintained efficacy against ganciclovir-resistant clinical isolates, underscoring their potential value in addressing drug resistance—a significant challenge in current HCMV treatment regimens .

This broad-spectrum activity stems from NAPA compounds' mechanism of action targeting early infection stages, which appears conserved across different HCMV strains despite their genetic variations . The ability to inhibit diverse HCMV strains, including drug-resistant variants, positions NAPA compounds as promising candidates for development as novel anti-HCMV therapeutics capable of addressing limitations of existing treatment options .

What screening approaches are most effective for identifying novel NAPA derivatives with improved properties?

The most effective screening approaches for identifying improved NAPA derivatives employ a multi-tiered strategy that integrates computational, biological, and pharmacological assessments. Initial high-throughput screening efficiently identified the original NAPA scaffold from >112,000 compounds using a reporter virus system (AD169 IE2/YFP) that expresses fluorescent markers upon infection, allowing rapid quantification of antiviral activity . For derivative screening, this reporter system remains valuable for primary assessments, with compounds reducing YFP-positive nuclei by ≥70% while minimally affecting total cell numbers (≤25% reduction) considered promising candidates .

Structure-activity relationship (SAR) studies guide rational design of derivatives by systematically modifying the five structural zones identified in the NAPA scaffold . This approach has already yielded valuable insights, such as the enhanced potency achieved through aromatic secondary amines in Zone 1 and urea linkers in Zone 4 . Computational modeling based on these SAR findings can further accelerate the design process by predicting activities of novel derivatives before synthesis.

A comprehensive evaluation funnel for promising derivatives should include:

  • Dose-response assays to determine IC50 values using standardized protocols with at least six technical replicates

  • Cytotoxicity assessments in multiple cell lines using complementary methodologies (e.g., MTS assay, CellTiter Glo)

  • Solubility analysis by nephelometry to predict bioavailability challenges

  • Metabolic stability studies using liver microsomes to assess potential in vivo half-life

  • CYP450 inhibition assays to predict drug-drug interaction risks

  • Efficacy testing against diverse HCMV strains, including clinical isolates and drug-resistant variants

  • Combination studies with established antivirals to identify synergistic pairs

This integrated screening approach enables efficient identification of NAPA derivatives with optimal combinations of potency, selectivity, pharmacokinetic properties, and broad-spectrum activity.

How should researchers design experiments to evaluate NAPA compounds' efficacy in combination with established anti-HCMV drugs?

Researchers should employ a systematic, multi-step approach to evaluate NAPA compounds' efficacy in combination with established anti-HCMV drugs. The experimental design should begin with in vitro combination studies using a dose-matrix format that tests multiple concentrations of both the NAPA compound and the established drug (e.g., ganciclovir, foscarnet, or cidofovir) . This creates a comprehensive interaction landscape that allows for identification of optimal concentration ratios for synergistic effects.

Quantification of combination effects should utilize established mathematical models such as the Chou-Talalay method or Bliss independence model to calculate combination indices (CI), where CI<1 indicates synergy, CI=1 indicates additivity, and CI>1 indicates antagonism . Studies with NAPA compounds have already demonstrated additive or synergistic inhibition of HCMV proliferation when combined with ganciclovir, suggesting favorable interaction potential .

Experiments should evaluate combinations across multiple relevant cell types, particularly human fibroblasts and epithelial cells, which represent major targets for HCMV infection . Testing against both laboratory-adapted strains and clinical isolates, including drug-resistant variants, is essential to fully characterize the combination's spectrum of activity .

Mechanistic studies should explore the biochemical basis for observed synergies, potentially including time-of-addition experiments to determine if sequential administration enhances efficacy compared to simultaneous treatment . Researchers should also assess the impact of combinations on the emergence of resistance by conducting serial passage experiments under sub-inhibitory drug concentrations.

For advanced preclinical evaluation, researchers should design pharmacokinetic studies to determine if co-administration affects the disposition of either compound and adjust dosing regimens accordingly. Animal models of HCMV infection, though challenging due to the species-specificity of cytomegaloviruses, can be leveraged using mouse CMV (MCMV) with mouse-active NAPA analogs to assess in vivo combination efficacy and toxicity profiles.

What analytical techniques are most appropriate for studying the pharmacokinetics and metabolism of NAPA compounds?

The optimal analytical approach to studying NAPA compounds' pharmacokinetics and metabolism requires a combination of in vitro, in silico, and in vivo techniques tailored to these compounds' unique chemical properties. For in vitro metabolism studies, liquid chromatography-tandem mass spectrometry (LC-MS/MS) offers the sensitivity and specificity needed to identify and quantify NAPA compounds and their metabolites in biological matrices . This approach should be complemented by microsomal stability assays, which have already demonstrated favorable stability for many NAPA analogs in the presence of NADPH, suggesting resistance to first-pass metabolism .

Plasma protein binding assays are essential for determining the free fraction of NAPA compounds, as only unbound drug is available for pharmacological activity. Given the varying physicochemical properties across the NAPA series, with significant differences in solubility between basic amine-containing analogs and other derivatives, plasma protein binding may vary substantially and influence in vivo efficacy .

Transporter studies using cell lines overexpressing specific drug transporters (e.g., P-glycoprotein, BCRP) should be conducted to determine if NAPA compounds are substrates or inhibitors, potentially affecting their distribution or causing drug-drug interactions. Similarly, comprehensive CYP450 inhibition and induction studies, beyond the CYP3A4 inhibition assays already performed, would provide crucial information about potential metabolic interactions .

For in vivo pharmacokinetic studies, the development of sensitive bioanalytical methods is critical. Based on NAPA compounds' structural characteristics, LC-MS/MS methods with appropriate sample preparation techniques (e.g., protein precipitation, liquid-liquid extraction) should be optimized for specific derivatives. These methods should achieve limits of quantification suitable for detecting drug levels throughout the expected concentration-time profile, including at late time points.

Physiologically-based pharmacokinetic (PBPK) modeling can integrate physicochemical properties, in vitro ADME data, and early in vivo findings to predict human pharmacokinetics and guide dose selection for potential clinical development. This approach is particularly valuable for compounds like the NAPA series, where various structural modifications significantly impact pharmacological properties .

What is the comparative potency and selectivity profile of leading NAPA compounds?

The comparative potency and selectivity profiles of leading NAPA compounds provide crucial insights for prioritizing candidates for further development. Below is a structured representation of key NAPA analogs' properties based on available research data:

NAPA CompoundIC50 (μM)CC50 (μM)Selectivity IndexMax SolubilityMicrosomal Stability*
MBXC-43023.2>100>31ModerateStable
MBXC-4297<1.0>50>50LowStable
MBXC-4336<1.0>30>30ModerateStable
MBX-4992<1.0>30>30ModerateStable
Zone 1 Tertiary Amines0.5-3.0>50>25-100HighStable
Zone 1 Aromatic Amines0.3-1.07-507-50Low-ModerateVariable

*Microsomal stability represented as % remaining after 60 min at 37°C in the presence of NADPH

This comparative analysis demonstrates that while aromatic amines in Zone 1 generally confer enhanced potency (lower IC50 values), they often exhibit lower selectivity indices compared to tertiary amines . The improved potency of aromatic amine derivatives appears to come with trade-offs in terms of solubility and potentially metabolic stability .

Among the leading compounds, MBXC-4297 stands out with a selectivity index >50, though its lower solubility may present challenges for formulation and bioavailability . MBXC-4336 and MBX-4992, which have been further characterized in broad-spectrum activity studies, maintain favorable selectivity indices (>30) with moderate solubility and good metabolic stability, representing promising candidates for additional development .

The data supports a development strategy that balances potency with other critical pharmacological properties, rather than focusing exclusively on compounds with the lowest IC50 values . This integrated analysis guides rational selection of NAPA derivatives for advancement through the preclinical development pipeline.

How does structural modification in different zones of NAPA compounds affect their antiviral properties?

The impact of structural modifications across the five identified zones of NAPA compounds reveals critical structure-activity relationships that guide rational drug design. The following table summarizes key findings from systematic structural variations and their effects on antiviral properties:

ZoneModificationEffect on PotencyEffect on SelectivityEffect on SolubilityKey Insights
1Heteroaromatic pyrazole to aromatic secondary amine10-fold improvementVariable (7 to >50)DecreasedSuggests hydrophobic binding pocket; enhanced potency but variable selectivity
1Acyclic ethylamineSimilar to referenceMaintainedIncreasedPolar groups maintained baseline activity but didn't enhance potency
1Increasing lipophilicityImprovedMaintained highVariableHydrophobic interactions enhance binding; piperidine substitution restored sub-micromolar activity
1Tertiary vs. secondary aminesLower potency but more consistentHigher (>25)HigherTertiary amines offer better selectivity and physicochemical properties despite lower potency
2Isomeric pyrimidine ligandsVariableSignificantly affectedMinimal changeElectronics of pyrimidine moiety critical for selectivity; MBXC-4297 maintained index >50 while isomers dropped to <20
4Sulfone linkerActivity lostNot applicableNot reportedNot tolerated, suggesting specific spatial or electronic requirements
4Urea linkerImprovedMaintainedNot reportedSuggests hydrogen-bonding site in binding pocket
5Furan to thiophene or phenylComplete loss of activityNot applicableNot reportedHighly sensitive to modification; suggests strict steric/electronic requirements

These structure-activity relationships reveal several key principles for optimizing NAPA compounds. Zone 1 modifications demonstrate the most significant impact on potency, with a clear preference for hydrophobic moieties, though the optimal balance between potency and selectivity varies among substitutions . Zone 2 alterations primarily affect selectivity rather than potency, with the electronic properties of the pyrimidine ring playing a crucial role in determining the compounds' safety profile .

The complete loss of activity with seemingly minor modifications in Zone 5 (furan to thiophene or phenyl) highlights the exquisite specificity of target binding, suggesting a highly constrained binding pocket in this region . Similarly, the differential effects of linker modifications in Zone 4 point to specific hydrogen-bonding interactions that contribute to compound efficacy .

These findings provide a comprehensive roadmap for rational design of next-generation NAPA derivatives, identifying specific structural features that can be optimized to enhance potency while maintaining or improving selectivity and pharmacological properties.

What are the most promising approaches for overcoming potential limitations of NAPA compounds?

Addressing potential limitations of NAPA compounds requires strategic approaches targeting specific physicochemical, pharmacological, and biological challenges. Solubility enhancement represents a primary focus, particularly for highly potent analogs with aromatic amines in Zone 1 that demonstrate lower aqueous solubility . Promising approaches include prodrug strategies that incorporate water-solubilizing groups that are cleaved upon reaching the target tissue, formulation with solubility-enhancing excipients, or strategic introduction of ionizable groups in non-critical positions of the molecules .

Optimization of metabolic stability, especially for compounds with variable microsomal stability profiles, can be achieved through strategic blocking of metabolically vulnerable sites, deuteration of susceptible positions, or isosteric replacements that maintain activity while reducing metabolic liability . These approaches should be guided by metabolite identification studies to precisely target the most problematic positions.

The strict structural requirements in Zone 5, where even minor modifications from furan to thiophene resulted in complete loss of activity, present both a challenge and opportunity . Addressing this limitation requires detailed structural biology approaches, potentially including photoaffinity labeling to identify the precise binding site, which would enable structure-based drug design to expand the range of tolerated substituents while maintaining target engagement.

For compounds showing promising activity profiles but suboptimal selectivity indices, medicinal chemistry efforts should focus on Zone 2 modifications, where isomeric variations have demonstrated significant impacts on selectivity without compromising potency . Systematic exploration of electronic effects in this region could yield derivatives with improved therapeutic windows.

Development of resistance remains a potential concern for any antiviral agent. Establishing in vitro resistance selection protocols and identifying potential resistance mutations preemptively would allow rational design of second-generation compounds that maintain activity against resistant strains or combination strategies that raise the genetic barrier to resistance.

Finally, expanding the therapeutic applications of NAPA compounds could address limitations in market potential. While currently focused on HCMV, investigating activity against other betaherpesviruses (HHV-6, HHV-7) or even distantly related DNA viruses could broaden the impact of this compound class .

What potential clinical applications might NAPA compounds address beyond current anti-HCMV therapies?

NAPA compounds offer potential clinical applications that extend beyond the limitations of current anti-HCMV therapies, addressing several unmet medical needs. As inhibitors targeting early stages of viral infection, NAPA compounds represent a mechanistically distinct approach compared to conventional drugs like ganciclovir that target viral DNA polymerase . This unique mechanism positions them as valuable options for several specialized applications.

For transplant recipients, who face high risks of HCMV reactivation or primary infection, NAPA compounds could serve as prophylactic agents with potentially fewer hematological side effects than current options . The favorable selectivity indices of leading NAPA compounds (>30) suggest reduced toxicity potential, addressing a major limitation of existing therapies that often require dose reductions or discontinuation due to neutropenia or thrombocytopenia .

In congenital CMV infections, where current therapies have limited efficacy and significant toxicity concerns, NAPA compounds' distinct mechanism of action could provide new therapeutic options . By targeting early infection stages, these compounds might more effectively prevent vertical transmission or reduce the severity of established infections in the developing fetus.

For patients with ganciclovir-resistant HCMV infections, which represent an increasing clinical challenge, NAPA compounds have demonstrated efficacy against resistant strains, offering potential salvage therapy options . Their mechanism of action, independent of viral DNA polymerase, allows them to maintain activity against polymerase mutations that confer resistance to ganciclovir and related drugs .

Perhaps most significantly, the synergistic or additive effects observed when NAPA compounds are combined with ganciclovir suggest potential for combination therapies that could lower the required doses of both agents, reducing toxicity while maintaining or enhancing antiviral efficacy . Such combinations might also raise the genetic barrier to resistance development, addressing another major limitation of current monotherapy approaches.

Beyond direct therapeutic applications, NAPA compounds could serve as valuable research tools for understanding early events in HCMV infection, potentially revealing new insights into viral entry, nuclear transport, and immediate-early gene expression that could inform development of next-generation antiviral strategies .

What are the key takeaways for researchers interested in NAPA compounds for HCMV research?

Researchers entering the field of NAPA compounds for HCMV research should recognize several crucial aspects of this promising antiviral class. First, NAPA compounds represent a mechanistically distinct approach to HCMV inhibition, targeting early stages of viral infection rather than DNA replication, providing opportunities to address limitations of existing therapies . This unique mechanism of action enables activity against ganciclovir-resistant strains and potential synergistic combinations with established antivirals .

The well-defined structure-activity relationships across five structural zones provide a solid foundation for rational drug design, with clear patterns emerging in how modifications affect potency, selectivity, and physicochemical properties . Particularly noteworthy is the enhanced potency achieved through aromatic secondary amines in Zone 1, the selectivity impact of pyrimidine electronics in Zone 2, and the critical hydrogen-bonding site suggested by urea linker improvements in Zone 4 .

The specificity of NAPA compounds for cytomegaloviruses, with no significant activity against other herpesviruses, indicates target interactions unique to HCMV biology . This specificity, combined with favorable selectivity indices (>30 for leading compounds), suggests potential for high therapeutic indices in clinical applications .

From a methodological perspective, researchers should appreciate the value of the high-content screening approach with reporter viruses like AD169 IE2-YFP for identifying and characterizing compounds affecting early-stage HCMV infection . This system provides efficient quantification of antiviral activity while distinguishing between specific inhibition and general cytotoxicity .

The comprehensive evaluation funnel established for NAPA compounds—encompassing potency, selectivity, solubility, metabolic stability, and activity against diverse viral strains—offers a template for systematic assessment of novel antivirals . This multifaceted approach ensures balanced optimization of all properties relevant to therapeutic potential, rather than focusing exclusively on potency.

Finally, researchers should recognize that while NAPA compounds show great promise, significant challenges remain in optimizing physicochemical properties, particularly solubility for certain analogs, and in fully elucidating the precise molecular target(s) and binding interactions . Addressing these challenges represents a fertile area for continued investigation with potential to yield clinically significant advances in HCMV therapy.

How might NAPA compound research evolve in the coming years based on current findings?

The trajectory of NAPA compound research is poised for significant evolution in the coming years, building on the solid foundation of current findings. Near-term research will likely focus on comprehensive target identification and validation studies, employing chemical proteomics, resistance selection, and structural biology approaches to definitively identify the molecular target(s) of NAPA compounds . This fundamental understanding will enable structure-based drug design to optimize interactions and potentially expand the structural diversity of effective analogs.

Medicinal chemistry efforts will continue refining NAPA derivatives, with particular emphasis on Zone 1 and Zone 4 modifications that have already demonstrated substantial impacts on potency . Advanced computational methods, including machine learning approaches trained on existing SAR data, may accelerate the identification of promising new derivatives with optimized property profiles.

Preclinical development of lead compounds will expand to include formulation studies addressing solubility challenges, particularly for highly potent but less soluble analogs . These efforts will likely explore various drug delivery strategies, including nanoformulations or prodrug approaches, to enhance bioavailability while maintaining target engagement.

In vivo efficacy studies will become increasingly important, though challenged by the species-specificity of cytomegaloviruses. Research may employ humanized mouse models infected with HCMV, surrogate models using murine CMV with mouse-active NAPA analogs, or potentially non-human primate models with their native CMVs to evaluate in vivo efficacy and safety .

Combination therapy research will expand beyond the current observations of additive or synergistic effects with ganciclovir to explore optimal drug partners, dosing ratios, and administration schedules . These studies will aim to maximize therapeutic efficacy while minimizing toxicity, potentially enabling new treatment paradigms for difficult-to-treat HCMV infections.

Translational research will increasingly focus on specific clinical applications where NAPA compounds might address unmet needs, including prophylaxis in transplant recipients, treatment of congenital CMV, and management of drug-resistant infections . These targeted approaches may accelerate clinical development by focusing on well-defined patient populations with clear benefit-risk profiles.

Product Science Overview

Introduction

N-Ethylmaleimide-Sensitive Factor Attachment Protein, Alpha (α-SNAP) is a crucial component of the cellular machinery involved in membrane fusion. This protein is encoded by the NAPA gene and is ubiquitously expressed in eukaryotic cells . The recombinant form of this protein is often used in research to study its function and interactions.

Structure and Function

α-SNAP is a soluble protein that plays a pivotal role in the SNARE (Soluble NSF Attachment Protein Receptor) complex . The SNARE complex is essential for the fusion of vesicles with target membranes, a process critical for intracellular trafficking and neurotransmitter release . α-SNAP binds to the SNARE complex and recruits the N-ethylmaleimide-sensitive factor (NSF), an ATPase that disassembles the SNARE complex after membrane fusion .

Mechanism of Action

The primary function of α-SNAP is to mediate the disassembly of the SNARE complex. This process is ATP-dependent and involves the hydrolysis of ATP by NSF . The binding of α-SNAP to the SNARE complex is a prerequisite for NSF attachment and subsequent ATP hydrolysis . This disassembly is crucial for the recycling of SNARE proteins and the maintenance of vesicular trafficking .

Biological Significance

α-SNAP is indispensable for various cellular processes, including synaptic transmission, hormone secretion, and vesicular transport . Alterations in α-SNAP expression or function have been linked to several neurological disorders, highlighting its importance in maintaining cellular homeostasis .

Research and Applications

Recombinant α-SNAP is widely used in biochemical and structural studies to understand its role in membrane fusion and SNARE complex dynamics . It is also employed in the development of therapeutic strategies targeting SNARE-mediated processes in diseases such as epilepsy and neurodegenerative disorders .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.