Synonym |
Fmoc-Val-Cit-PAB-PNP |
Species |
Human |
Protein Accession |
NA |
Purity |
> 95% |
Endotoxin Level |
< 1 EU/ug |
Biological Activity |
NA |
Expression System |
Chemical synthesis |
Fusion Tag |
NA |
Predicted Molecular Mass |
NA |
Formulation |
Supplied as a lyophilized powder |
Reconstitution |
Reconstitute in water to a concentration of 0.1-1.0 mg/ml |
Storage & Stability |
Store at -20°C. Stable for up to 12 months from date of receipt |
FAQ
What is Fmoc-Val-Cit-PAB-PNP and what is its significance in peptide synthesis?
Fmoc-Val-Cit-PAB-PNP
is a specialized chemical compound used extensively in the field of peptide synthesis and drug delivery,
particularly in the development of antibody-drug conjugates (ADCs). The compound's structure comprises
Fmoc (9-fluorenylmethoxycarbonyl) protecting group, Val (valine), Cit (citrulline), PAB
(para-aminobenzyl) linker, and PNP (p-nitrophenol) leaving group. Each component of this compound serves
a unique and crucial role in making it an optimal choice for advanced bioconjugation applications.
Specifically, the Fmoc group is utilized as a protecting group for amines, which can be selectively
removed to facilitate step-wise synthesis without compromising the integrity of the peptide. Valine and
citrulline are amino acids that provide structural significance and enhance the stability of the linker,
improving the efficacy and specificity in targeted delivery systems.
The PAB linker is notable
for its ability to act as a self-immolating spacer. This means that upon enzymatic cleavage of a peptide
bond, the PAB linker undergoes structural rearrangement that leads to its own degradation and releases
the attached payload. Such a mechanism is particularly important in drug delivery applications, ensuring
that the active drug is only released at the target site, thereby minimizing systemic toxicity and side
effects. The PNP leaving group facilitates efficient chemical reactions, often used in the final
deprotection or coupling reaction, due to its ability to be a good leaving group, enhancing the rate of
reaction and overall yield.
The significance of Fmoc-Val-Cit-PAB-PNP in peptide synthesis is
its multifunctionality and adaptability in creating complex peptide-drug conjugates with high precision
and efficiency. Its design is tailored to overcome common challenges in drug delivery, such as ensuring
stability in the bloodstream, achieving precise targeting, and enabling controlled release. Hence, this
compound is of great interest to researchers developing next-generation therapeutic agents where
precision targeting and safety are paramount. The meticulous design of Fmoc-Val-Cit-PAB-PNP underscores
its importance in advancing medicinal chemistry and facilitating breakthroughs in targeted therapies.
What are the potential applications of Fmoc-Val-Cit-PAB-PNP in medicine?
Fmoc-Val-Cit-PAB-PNP has burgeoning potential applications in medicine, particularly in the
development and improvement of therapeutic modalities such as targeted drug delivery systems and
antibody-drug conjugates (ADCs). ADCs are an innovative class of therapeutics that combine the
selectivity of monoclonal antibodies with the potent cytotoxicity of small-molecule drugs. These
conjugates leverage the high specificity of antibodies to deliver cytotoxic agents directly to cancer
cells, sparing normal tissues, and thus, reducing systemic toxicity. The role of Fmoc-Val-Cit-PAB-PNP in
this domain is particularly significant due to its highly effective linker and release mechanism.
In the context of ADCs, Fmoc-Val-Cit-PAB-PNP serves as a critical component for linking the drug
payload to the antibody. The enzymatically cleavable dipeptide linker, Val-Cit, in the compound is
designed to be stable in the bloodstream but undergoes cleavage in the presence of specific enzymes—such
as cathepsins—that are overexpressed in tumor cells. Once the linker is cleaved, it triggers the PAB
group to self-immolate and release the active drug precisely at the site of action. This targeted
release mechanism is crucial in minimizing off-target effects and improving the therapeutic index of
ADCs. Moreover, the Fmoc protecting group facilitates the stepwise synthesis of such complex molecules,
aiding in the assembly of sophisticated drug conjugates.
Beyond oncology,
Fmoc-Val-Cit-PAB-PNP's targeted release capability is also being explored in treating other diseases
where precise drug delivery could enhance therapeutic outcomes. For instance, in autoimmune diseases,
where treatment needs to be confined to specific tissues to reduce systemic immunosuppression, this
compound offers a promising approach. Similarly, in infectious diseases, conjugates designed to deliver
antimicrobial agents directly to infected cells can potentially increase efficacy while minimizing
damage to the host’s healthy cells.
Furthermore, Fmoc-Val-Cit-PAB-PNP may have utility in
developing diagnostic tools. By attaching imaging agents instead of cytotoxic drugs, researchers could
create conjugates that enable precise visualization of disease sites, aiding in early detection and
monitoring therapeutic responses. Thus, Fmoc-Val-Cit-PAB-PNP is pivotal in the ongoing evolution of
precision medicine, offering a versatile platform for developing tailored therapies that maximize
efficacy while minimizing adverse effects.
How does Fmoc-Val-Cit-PAB-PNP contribute to the
field of targeted cancer therapy?
The contribution of Fmoc-Val-Cit-PAB-PNP to the field of targeted
cancer therapy is profound and multifaceted, primarily through its application in the creation of
antibody-drug conjugates (ADCs). Targeted cancer therapies aim to specifically attack cancer cells while
sparing normal, healthy cells, thereby reducing side effects and improving therapeutic outcomes.
Fmoc-Val-Cit-PAB-PNP plays an integral role in realizing this goal due to its unique chemical properties
and structure.
The use of ADCs represents a significant advancement in cancer treatment,
combining the specificity of monoclonal antibodies with the potency of small-molecule chemotherapeutics.
In this regard, the Fmoc-Val-Cit-PAB-PNP compound is especially valuable for its linker technology that
facilitates the selective release of the drug at the tumor site. The peptide linkage Val-Cit in
Fmoc-Val-Cit-PAB-PNP is tailored to be cleaved by specific proteases like cathepsins, which are
overexpressed in many tumor types. This selective cleavage leads to a cascade of reactions, including
the self-immolative breakdown of the PAB linker, culminating in the release of the cytotoxic agent right
at the tumor site while maintaining stability in systemic circulation.
This precise targeting
capability translates into a more effective kill rate of cancer cells while minimizing damage to
surrounding healthy tissue, a balance that is critical in cancer therapy. The Fmoc group aids the
synthetic process, ensuring that complex drug conjugates can be built methodically and efficiently, thus
supporting the production of structurally diverse and potent ADCs. Beyond enhancing the delivery and
efficacy of chemotherapeutics, Fmoc-Val-Cit-PAB-PNP is also under exploration for delivering novel
therapeutic agents, like small interfering RNAs (siRNAs) or next-generation kinase inhibitors, which
could further expand the therapeutic arsenal against cancer.
Moreover, this compound may
facilitate the development of personalized medicine strategies. With the ability to fine-tune the ADC
components to target specific biomarkers expressed on cancer cells, therapies can be customized to
individual patients' tumor profiles, leading to more successful and personalized treatment regimens.
Therefore, Fmoc-Val-Cit-PAB-PNP is not just a chemical compound abetting cancer therapy but a linchpin
in advancing precision oncology, offering hope for more effective treatment options and improved patient
outcomes.
What are the challenges in using Fmoc-Val-Cit-PAB-PNP for new drug development?
While Fmoc-Val-Cit-PAB-PNP offers significant advantages in targeted drug delivery, particularly in
creating ADCs for cancer treatment, there are several challenges involved in using this compound for new
drug development. Each phase of developing novel therapeutics using such complex conjugates presents
distinct hurdles, from synthesis to clinical application.
One major challenge is the synthesis
and scale-up production of ADCs using Fmoc-Val-Cit-PAB-PNP. Synthesizing ADCs involves creating a stable
link between the antibody and the drug via the linker, which in this case includes the
Fmoc-Val-Cit-PAB-PNP. The complexity of accurately synthesizing such a compound lies in the need for
precise control over reactions, ensuring that the linker is both stable enough to survive the
bloodstream and reactive enough to release the drug at the right time and location. The Fmoc protecting
group requires specific conditions for removal, necessitating careful orchestration of each step in
synthesis to maintain yield and purity. Scaling the production from the laboratory to industrial levels
without compromising the consistency of these components is a significant challenge.
Another
challenge is the variability in expression levels of the enzymes required for the linker cleavage across
different patients and tumor types, which can affect the predictability and efficiency of drug release.
While such enzyme-selective linkers as Val-Cit are designed to be cleaved by specific proteases (e.g.,
cathepsins), the heterogeneity of tumor expression can lead to inconsistent therapeutic outcomes.
Moreover, ensuring the ADC reaches the tumor site in optimal concentration without premature breakdown
or clearance by the immune system is a complex delivery challenge that continues to necessitate
sophisticated engineering and validation.
From a regulatory and safety perspective, addressing
the potential immunogenicity of ADC components and unintended interactions within the body requires
extensive characterization and testing. This includes understanding the pharmacokinetics and potential
off-target effects, which must be thoroughly evaluated through pre-clinical and clinical studies.
Additionally, the high cost and resource-intensive nature of developing ADCs with Fmoc-Val-Cit-PAB-PNP
can be prohibitive, requiring a delicate balance between innovation and feasible manufacturing
processes.
Thus, while Fmoc-Val-Cit-PAB-PNP holds great promise for enhancing drug targeting
capabilities, the challenges in ensuring consistent production, targeting specificity, therapeutic
efficacy, and safety must be strategically addressed to successfully leverage its full potential in new
drug development.
What are the advantages of using Fmoc-Val-Cit-PAB-PNP in comparison to other
linkers in ADCs?
Fmoc-Val-Cit-PAB-PNP stands out among various linkers used in ADCs for several
advantageous features that address critical aspects of drug design, including stability, release
mechanism, and adaptability in synthesis and application. Compared to other linkers, these attributes
offer significant potential in delivering safer and more effective targeted therapies.
One
significant advantage is its stability in the bloodstream contrasted with its efficacy at the target
site. The stability of an ADC is paramount to its effectiveness, as premature release of the cytotoxic
payload can result in systemic toxicity. The Fmoc-Val-Cit-PAB-PNP linker is constructed with a Val-Cit
dipeptide sequence that remains stable under physiological conditions but is efficiently cleaved by
specific lysosomal enzymes such as cathepsins inside the target cancer cells. This enzymatic sensitivity
ensures that the payload release occurs precisely where needed, thereby minimizing collateral damage to
healthy cells and enhancing the therapeutic index.
Another notable advantage is the
self-immolative nature of the PAB moiety within the linker. Upon enzymatic cleavage of the Val-Cit bond
within tumor cells, the PAB unit undergoes spontaneous decomposition, facilitating the controlled
release of the drug payload. This self-immolative property not only ensures a rapid and complete drug
release but also favors the use of more potent cytotoxic agents that depend on accurate delivery and
release mechanisms to be safely used in the bloodstream.
Furthermore, the Fmoc protecting group
is a particularly useful feature during synthesis, allowing for the stepwise conjugation process that
ensures high precision and reduced risk of undesired side reactions. This level of control and
predictability in synthesis offers manufacturers the ability to consistently produce high-purity ADCs
with specific design features tailored to meet therapeutic goals.
In comparison to other linker
technologies, such as non-cleavable linkers or those dependent on different physicochemical triggers for
activation, the enzymatic cleavability of Fmoc-Val-Cit-PAB-PNP provides a greater level of biological
specificity and adaptability to diverse therapeutic scenarios. This is particularly advantageous in the
treatment of heterogeneous tumors, where variability in molecular markers is common. The enhanced
specificity and reduction of off-target effects also translate into improved clinical safety profiles,
which are critical in drug approval and patient acceptance. Thus, the distinct chemical and functional
properties of Fmoc-Val-Cit-PAB-PNP render it a superior choice for designing next-generation ADCs aimed
at improving the outcomes in cancer and potentially other therapeutic areas.