Synonym |
N-Me-Val-Leu-anilide |
Species |
Human |
Protein Accession |
NA |
Purity |
Greater than 95% as determined by SDS-PAGE |
Endotoxin Level |
Less than 1 EU/μg |
Biological Activity |
Not determined |
Expression System |
E. coli |
Fusion Tag |
His tag |
Predicted Molecular Mass |
354.4 kDa |
Formulation |
Lyophilized from a 0.2 μm filtered solution in PBS |
Reconstitution |
It is recommended to reconstitute the lyophilized product with sterile water at a concentration
of 0.1 mg/ml, which can then be further diluted into other aqueous solutions |
Storage & Stability |
Store lyophilized product at -20°C. Upon reconstitution, store at 4°C for up to one week or at
-20°C for longer. Avoid repeated freeze-thaw cycles. |
FAQ
What is N-Me-Val-Leu-anilide and what are its potential applications in the pharmaceutical
industry?
N-Me-Val-Leu-anilide is a synthetic compound that falls within the category of peptide
derivatives. This compound is comprised of a sequence of amino acids—namely, N-methyl valine, leucine,
and anilide. Such compounds are of significant interest in the pharmaceutical industry due to their
structural similarity to naturally occurring peptides and proteins, making them potential candidates for
drug development. Peptides are critical biological molecules that often play central roles in various
physiological functions, and as such, have been targeted for therapeutic intervention in a wide range of
medical conditions.
One of the main potential applications of N-Me-Val-Leu-anilide is in the
development of drugs that can modulate specific biological functions by either mimicking or inhibiting
natural peptide substrates. Peptides can act as signaling molecules, hormones, or enzyme substrates, and
slight modifications in their sequence can drastically alter their interaction with biological receptors
or enzymes. N-Me-Val-Leu-anilide's structural backbone can be optimized to enhance binding affinity or
selectivity towards these biological targets, making it a versatile lead compound in drug
design.
Furthermore, N-Me-Val-Leu-anilide may also serve as a probe in biochemistry and molecular
biology research. The compound can be used to study the behavior of peptide-binding proteins or enzymes,
providing insights into fundamental biological processes. This can facilitate the elucidation of
mechanisms underlying various diseases, which in turn can lead to the identification of new therapeutic
targets. Additionally, the chemical synthesis of N-Me-Val-Leu-anilide allows for the introduction of
various chemical groups that can expand its utility in research, such as adding fluorophores for imaging
studies or biotin tags for affinity purification.
The stability and specificity of peptide
derivatives like N-Me-Val-Leu-anilide offer an advantage over traditional small molecules, as they tend
to have a benign safety profile and reduced off-target effects. This makes them attractive candidates
for drug development, particularly for chronic conditions where long-term medication administration is
required. However, challenges such as peptide bioavailability and rapid degradation in vivo must be
overcome. Techniques such as cyclic peptide design, pegylation, and encapsulation in nanocarriers are
explored to enhance the pharmacokinetic properties of peptide-based drugs.
Overall,
N-Me-Val-Leu-anilide represents a promising tool in the arena of drug discovery and biological research.
Its potential applications are vast, encompassing therapeutic development, mechanistic studies, and the
novel design of bioactive molecules. As research continues to advance the understanding of peptide
chemistry and biology, compounds like N-Me-Val-Leu-anilide will likely play an even greater role in
innovative therapeutic strategies.
How does the structure of N-Me-Val-Leu-anilide influence its
biological activity?
The biological activity of N-Me-Val-Leu-anilide is intricately linked to its
structure, which is defined by its sequence of amino acids—N-methyl valine, leucine, and anilide. This
sequence and its chemical modifications determine how the compound interacts with biological targets,
which in turn affects its potential utility as a therapeutic agent. One fundamental aspect of its
structure is the presence of the N-methyl group. Methylation of the amino group can enhance the
compound's stability by protecting it from proteolytic enzymes that typically degrade peptides. This
increased stability can lead to prolonged activity in biological systems, which is highly desirable in
drug development.
Moreover, the structure of N-Me-Val-Leu-anilide could also affect its
solubility and permeability, impacting its bioavailability. The lipophilic side chains of valine and
leucine may promote membrane permeability, potentially facilitating the compound's ability to cross
cellular barriers and interact with intracellular targets. This feature is important for compounds
intended to act within cells rather than extracellular or on the cell surface. However, increased
lipophilicity can sometimes lead to poor water solubility, which would necessitate formulation
strategies to enhance solubility while maintaining biological efficacy.
The specific arrangement
of amino acids, along with the chemical environment within N-Me-Val-Leu-anilide, influences its
three-dimensional conformation. This conformational preference is crucial as it dictates the compound's
ability to bind to specific receptors or enzymes with high affinity and selectivity. The precise fit
into these biological targets can activate or inhibit the receptor or enzyme's function, resulting in
the modulation of a particular physiological pathway. Such specificity is beneficial in reducing
off-target effects and improving the therapeutic index of potential drugs.
Another noteworthy
feature is the anilide moiety at the end of the sequence. Anilides are known to impart various
properties, such as influencing the electronic distribution within the molecule, which could affect
binding interactions with target proteins. This property might enhance the compound’s affinity to
certain receptor sites or alter its activity profile, making N-Me-Val-Leu-anilide a favorable candidate
for modifying biological responses.
In addition to its potential as a therapeutic agent, the
structural characteristics of N-Me-Val-Leu-anilide can be capitalized upon in drug delivery systems. By
conjugating N-Me-Val-Leu-anilide to drug carriers or incorporating it into nanoparticles, its stability
and cellular uptake may be further improved. This conjugation provides an opportunity to develop
advanced delivery systems that target specific tissues or cells in the body, thereby optimizing the
therapeutic effects while minimizing systemic side effects.
Overall, the structure of
N-Me-Val-Leu-anilide plays a critical role in its biological activity, impacting stability,
permeability, binding affinity, and specificity. These factors collectively define its potential
applications in therapeutics and research, and continuous advancements in peptide chemistry will further
enhance the ability to tailor such compounds for targeted biological effects.
What are the
challenges associated with the development of N-Me-Val-Leu-anilide as a therapeutic
agent?
Developing N-Me-Val-Leu-anilide as a therapeutic agent involves addressing several
challenges inherent in peptide-based drug design and delivery. One significant issue is the molecular
stability of peptides, as they are vulnerable to enzymatic degradation in the gastrointestinal tract and
bloodstream. Proteases can rapidly degrade unmodified peptides, leading to a short half-life and reduced
efficacy when administered orally or by other common routes. For N-Me-Val-Leu-anilide, which includes a
methylated amino acid, there may be some inherent stability improvements, yet proteolytic degradation
remains a concern that necessitates additional modifications or delivery strategies to prolong its
activity.
Another challenge is associated with the pharmacokinetic properties of peptide-based
drugs. While peptides generally have good tissue penetration and an ability to target specific pathways,
their overall bioavailability is often limited due to rapid clearance by the kidneys and poor membrane
permeability. Compounds like N-Me-Val-Leu-anilide need to be designed or formulated in a way that
maximizes retention in the bloodstream and ensures consistent delivery to the desired tissue or cellular
targets. This can involve intricate formulation strategies, including encapsulation in liposomes,
cyclodextrin inclusion complexes, or other nanoparticles.
The route of administration also poses
a challenge. Peptide-based therapeutics generally require parenteral administration (e.g., intravenous
or subcutaneous injections) due to their poor oral bioavailability. For broader patient compliance and
market potential, developing non-invasive delivery methods such as transdermal patches, inhalation
formulations, or advanced oral delivery systems might be necessary. These alternative methods must
overcome barriers such as skin permeability, pulmonary absorption, and protection from the harsh
gastrointestinal environment.
Cost of production is another consideration in developing
peptide-based drugs like N-Me-Val-Leu-anilide. Peptide synthesis can be expensive and time-consuming,
especially when high purity and yields are required for therapeutic applications. Advancements in
synthesis technology, such as solid-phase peptide synthesis (SPPS) and microwave-assisted synthesis, can
help reduce costs and production times, but these still represent a significant portion of the
development budget.
Formulation and stability issues also present hurdles; peptides can be
sensitive to temperature, pH, and mechanical stress. Therefore, N-Me-Val-Leu-anilide needs to be
formulated with suitable excipients to stabilize the compound both during storage and after
administration, ensuring that it maintains its activity until it reaches the target
site.
Finally, regulatory hurdles are stringent for peptide-based therapeutics, where
comprehensive preclinical and clinical testing is required to establish safety, efficacy,
pharmacokinetics, and pharmacodynamics. N-Me-Val-Leu-anilide would need to demonstrate not only
therapeutic benefits but also a favorable safety profile with minimal side effects. Addressing these
challenges involves extensive research and collaboration between chemists, pharmacologists, and
regulatory experts to bring a successful therapeutic agent to market.
In sum, despite the
promising potential of N-Me-Val-Leu-anilide as a therapeutic agent, significant challenges regarding
stability, bioavailability, administration routes, cost, formulation, and regulatory requirements need
to be addressed. Concerted efforts in research and development can help overcome these obstacles, paving
the way for innovative treatments using this peptide derivative.
Why is the specificity of
N-Me-Val-Leu-anilide important in therapeutics, and how can it be achieved?
The specificity of
N-Me-Val-Leu-anilide in therapeutics is paramount because it dictates the compound's ability to
selectively bind to a particular biological target—be it a receptor, enzyme, or protein—without
affecting others. This preferential interaction is crucial for maximizing therapeutic efficacy while
minimizing potential off-target effects and adverse reactions. Achieving high specificity can
significantly improve the safety and tolerability profiles of a drug, allowing for higher doses if
necessary and reducing the risk of side effects associated with the intervention.
One strategy to
attain specificity involves meticulous structural design focused on optimizing the compound's molecular
interactions with its intended target. This includes ensuring that N-Me-Val-Leu-anilide's amino acid
sequence and overall conformation precisely complement the binding site of the target, mirroring the
'lock and key' model. Computational modeling and structure-activity relationship (SAR) studies can be
invaluable in this process, facilitating an understanding of how minor alterations in the compound’s
structure influence binding affinity and specificity.
Another avenue is the incorporation of
chemical modifications that enhance specificity. Derivatization, such as the addition of protective
groups or the substitution of specific atoms or functional groups, can amplify the interaction with the
target site. It can also prevent the compound from being recognized and acted upon by unintended targets
or enzymes. For N-Me-Val-Leu-anilide, maintaining the methylation on valine or introducing other
modifications on the anilide group could tailor its binding characteristics further.
Specificity
can also be achieved via a high-throughput screening of peptide libraries, which can pinpoint variants
that exhibit desired binding properties to the target protein. This method is efficient in identifying
lead candidates with optimal specificity from a large pool of random peptide sequences. Once identified,
these leads can undergo iterative cycles of optimization for further refinement.
Incorporating
technologies like phage display and affinity maturation can amplify the specificity of
N-Me-Val-Leu-anilide. These techniques allow for the selection and enrichment of peptide sequences that
bind with high affinity and specificity to a target of interest. Affinity maturation involves successive
rounds of mutation and selection to enhance the binding characteristics of the lead peptide while
maintaining or even enhancing specificity.
In addition, site-specific conjugation or fusion of
N-Me-Val-Leu-anilide with targeting moieties such as antibodies or aptamers can enhance specificity.
These conjugates can direct the compound to the desired cells or tissues, thereby improving localization
and minimizing systemic exposure. This approach is particularly beneficial for targeting disease sites
or cell-specific markers in conditions like cancer or autoimmune diseases.
Overall, the
specificity of N-Me-Val-Leu-anilide in therapeutics can be achieved through a combination of rational
design, chemical modification, high-throughput screening, advanced selection technologies, and targeted
delivery strategies. These approaches collectively ensure that the compound exerts its therapeutic
effects precisely where needed, improving treatment outcomes and patient safety in clinical
settings.
What are potential side effects that could arise from using N-Me-Val-Leu-anilide, and
how might these be mitigated?
As with any therapeutic compound, the use of N-Me-Val-Leu-anilide
could potentially lead to side effects, which arise when the compound interacts with unintended pathways
or when its interaction with the intended target has broader physiological impacts. Understanding and
mitigating these side effects is an essential step in the drug development process, aimed at ensuring
both efficacy and safety for the end user.
One potential side effect of peptide-based
therapeutics, including N-Me-Val-Leu-anilide, is immunogenicity. Peptides, being foreign entities
introduced into the body, might trigger an immune response that results in the formation of antibodies
against the compound. This immunogenic potential is a key concern since it could render the treatment
ineffective over time or provoke adverse immune responses. To mitigate this, strategies such as
modifying the peptide sequence to decrease its antigenicity, using human-derived sequences, or employing
immunosuppressive regimens may be considered.
Off-target effects represent another category of
potential side effects. These occur when the compound interacts with proteins or receptors other than
the intended target, leading to unintended physiological effects. Addressing off-target interactions
involves refining the specificity of N-Me-Val-Leu-anilide through chemical modifications,
high-throughput screening, and affinity maturation, ensuring the compound is highly selective for its
target protein.
Pharmacokinetic-related side effects are also possible, arising from the
compound's distribution, metabolism, and excretion profiles. Rapid clearance might necessitate higher
dosages or more frequent administration, increasing the risk of toxicity and side effects. On the other
hand, reduced clearance can result in accumulation and toxicity. These issues can be mitigated by
optimizing the administration route (e.g., using controlled-release formulations) or altering the
chemical structure to improve pharmacokinetic properties while maintaining
efficacy.
Dose-dependent side effects must also be considered. High concentrations of peptides
can sometimes lead to aggregation or precipitation, especially if solubility issues are present, causing
localized reactions or site-specific toxicity. This can be mitigated through proper formulation
strategies that enhance solubility or stabilize the compound in solution, such as co-solvents,
surfactants, or encapsulation technologies.
Additionally, local injection site reactions or
hypersensitivity can occur with peptide therapeutics, particularly for those administered parenterally.
These can range from mild irritation to severe inflammatory responses. To mitigate these reactions,
formulation optimization to reduce irritants, the use of buffer systems that match physiological pH, and
ensuring that the peptide is free from impurities or endotoxins are essential
considerations.
Finally, individual variability in patient response is always a concern. Factors
such as genetic differences, pre-existing conditions, concomitant medications, and overall health can
influence how a patient reacts to N-Me-Val-Leu-anilide. Personalized medicine approaches, such as
pharmacogenomic screenings, can help tailor treatments to individual patients, optimizing efficacy while
minimizing side effects.
In conclusion, understanding the potential side effects of
N-Me-Val-Leu-anilide and implementing strategic mitigation practices are crucial for its successful
development as a therapeutic agent. Through scientific investigation and technological advancements,
these challenges can be addressed, leading to safer and more effective treatments.