Peptides are crucial building blocks in medicinal chemistry, composed of amino acids linked by peptide bonds. They play vital roles in biological processes and serve as important drug targets. Understanding peptide structure, synthesis, and interactions is key to developing effective therapies.

Peptide-based drug discovery has led to breakthroughs in treating various diseases. From peptide to antimicrobial and anticancer peptides, these versatile molecules offer high specificity and low toxicity. Overcoming challenges like poor bioavailability and rapid clearance is essential for advancing peptide-based therapies.

Peptide structure and composition

Amino acid building blocks

Top images from around the web for Amino acid building blocks
Top images from around the web for Amino acid building blocks
  • Peptides are composed of amino acids, which are organic compounds containing an amino group, a carboxyl group, and a unique side chain (R group)
  • There are 20 standard amino acids, each with a specific side chain that determines its chemical properties and interactions
  • Amino acids are joined together by peptide bonds, formed through a condensation reaction between the carboxyl group of one amino acid and the amino group of another

Primary structure of peptides

  • The of a peptide refers to the linear sequence of amino acids connected by peptide bonds
  • The order of amino acids in the primary structure is determined by the genetic code and is crucial for the peptide's function and properties
  • The N-terminus of a peptide has a free amino group, while the C-terminus has a free carboxyl group

Secondary structures: α-helices and β-sheets

  • Secondary structures are regular, repeating patterns of local folding within a peptide chain, stabilized by hydrogen bonds between the backbone atoms
  • The α-helix is a common , characterized by a right-handed spiral conformation with 3.6 amino acids per turn, stabilized by hydrogen bonds between the carbonyl oxygen of one amino acid and the amino hydrogen of another located four residues ahead
  • β-sheets are another type of secondary structure, formed by extended peptide chains that are aligned side-by-side and stabilized by hydrogen bonds between the backbone atoms of adjacent strands, creating a pleated sheet-like structure (parallel or antiparallel)

Tertiary structure and folding

  • refers to the three-dimensional arrangement of a peptide chain, resulting from the folding and interaction of secondary structure elements
  • Folding is driven by various non-covalent interactions, such as hydrogen bonds, van der Waals forces, hydrophobic interactions, and electrostatic interactions between amino acid side chains
  • The tertiary structure of a peptide is crucial for its biological function, as it determines the peptide's binding sites, catalytic activity, and interactions with other molecules
  • Misfolding of peptides can lead to aggregation and the formation of insoluble fibrils, which are associated with various diseases (Alzheimer's, Parkinson's)

Peptide synthesis

Solid-phase peptide synthesis (SPPS)

  • SPPS is a method for synthesizing peptides by anchoring the growing peptide chain to a solid support (resin) and adding amino acids sequentially
  • The C-terminal amino acid is attached to the resin, and subsequent amino acids are coupled to the N-terminus of the growing chain using protecting groups and coupling reagents
  • SPPS enables the synthesis of longer peptides with higher purity compared to solution-phase methods, as the resin-bound peptide can be easily washed and purified after each coupling step

Protecting groups and coupling reagents

  • Protecting groups are chemical moieties used to temporarily block reactive functional groups (amino, carboxyl) during peptide synthesis to prevent unwanted side reactions
  • Common protecting groups include Fmoc (9-fluorenylmethoxycarbonyl) for the N-terminus and tBu (tert-butyl) for side chains, which can be selectively removed under specific conditions
  • Coupling reagents activate the carboxyl group of the incoming amino acid, facilitating its reaction with the N-terminus of the growing peptide chain
  • Examples of coupling reagents include carbodiimides (DCC, EDC), phosphonium salts (PyBOP), and uronium salts (HBTU, HATU)

Purification and characterization techniques

  • After synthesis, peptides need to be purified to remove truncated sequences, deletion products, and other impurities
  • Reversed-phase high-performance liquid chromatography (RP-HPLC) is commonly used for peptide purification, separating peptides based on their hydrophobicity
  • (MALDI-TOF, ESI-MS) is used to characterize the purified peptides, confirming their molecular weight and sequence
  • Circular dichroism (CD) spectroscopy and nuclear magnetic resonance (NMR) spectroscopy can provide information about the secondary structure and conformation of peptides

Challenges in peptide synthesis

  • Peptide synthesis can be challenging due to the potential for side reactions, such as racemization, aspartimide formation, and peptide aggregation
  • Difficult sequences, such as those containing multiple hydrophobic or β-branched amino acids, can lead to incomplete coupling and low yields
  • Longer peptides (>50 amino acids) are more prone to aggregation and can be difficult to synthesize and purify
  • Chemical modifications, such as cyclization, stapling, or the incorporation of non-natural amino acids, may require specialized strategies and reagents

Peptide-based drug discovery

Therapeutic targets for peptide drugs

  • Peptides can be designed to target various receptors, enzymes, and other proteins involved in disease pathways
  • G protein-coupled receptors (GPCRs) are common targets for peptide drugs, as they play crucial roles in signaling and are implicated in numerous diseases (diabetes, cancer, cardiovascular disorders)
  • Peptides can also target ion channels, transporters, and protein-protein interactions, modulating their activity and downstream effects
  • Examples of peptide drug targets include the glucagon-like peptide-1 receptor (GLP-1R) for diabetes, the μ-opioid receptor for pain management, and the vascular endothelial growth factor (VEGF) for cancer therapy

Peptide libraries and screening methods

  • Peptide libraries are collections of diverse peptide sequences that can be screened for binding or activity against a specific target
  • Libraries can be generated by chemical synthesis (combinatorial libraries) or biological methods (phage display, mRNA display)
  • High-throughput screening (HTS) techniques, such as fluorescence polarization, surface plasmon resonance (SPR), and enzyme-linked immunosorbent assay (ELISA), are used to identify peptides with desired properties from large libraries
  • Iterative rounds of screening, selection, and amplification can be employed to enrich for peptides with high and specificity for the target

Peptidomimetics and peptide analogs

  • are compounds that mimic the structure and function of peptides but have improved pharmacological properties, such as increased stability, bioavailability, and potency
  • Peptide analogs are modified versions of natural peptides, incorporating non-natural amino acids, backbone modifications, or structural constraints to enhance their drug-like properties
  • Examples of peptidomimetic strategies include the use of D-amino acids, β-amino acids, peptoids, and small molecule mimics
  • Peptide analogs can be designed to improve receptor , reduce off-target effects, and increase resistance to proteolytic degradation

Peptide drug delivery systems

  • Peptide drugs often face challenges in delivery due to their poor oral bioavailability, rapid clearance, and limited permeability across biological barriers
  • Various drug delivery systems have been developed to overcome these limitations and improve the therapeutic efficacy of peptide drugs
  • Liposomes and nanoparticles can encapsulate peptides, protecting them from degradation and facilitating their transport across membranes
  • Cell-penetrating peptides (CPPs) can be conjugated to to enhance their cellular uptake and intracellular delivery
  • Sustained-release formulations, such as hydrogels and microspheres, can provide controlled and prolonged release of peptide drugs, reducing the frequency of administration

Peptide-protein interactions

Peptide binding sites on proteins

  • Peptides can bind to specific sites on proteins, such as active sites, allosteric sites, or protein-protein interaction interfaces
  • Binding sites are typically characterized by complementary shapes, electrostatic interactions, and hydrophobic contacts between the peptide and protein surfaces
  • The specificity and affinity of peptide-protein interactions are determined by the amino acid composition, sequence, and conformation of the peptide and the corresponding binding site on the protein
  • Examples of peptide binding sites include the substrate-binding pocket of enzymes, the ligand-binding domain of receptors, and the groove of major histocompatibility complex (MHC) molecules

Peptide-mediated signaling pathways

  • Peptides can act as ligands for cell surface receptors, triggering intracellular signaling cascades that regulate various cellular processes, such as proliferation, differentiation, and apoptosis
  • Peptide hormones and neuropeptides bind to specific GPCRs, leading to the activation of G proteins and downstream effector molecules (adenylyl cyclase, phospholipase C)
  • Peptide growth factors, such as insulin-like growth factor (IGF) and epidermal growth factor (EGF), bind to receptor tyrosine kinases (RTKs), initiating cascades that control gene expression and cell fate
  • Peptides can also modulate signaling pathways by interfering with protein-protein interactions, such as the binding of adaptor proteins or the formation of signaling complexes

Peptide inhibitors and agonists

  • Peptides can be designed to act as inhibitors or agonists of specific protein targets, modulating their activity and downstream effects
  • Peptide inhibitors can block the binding of natural ligands, substrates, or protein partners, preventing the activation of the target protein
  • Examples of peptide inhibitors include the HIV-1 fusion inhibitor enfuvirtide, which blocks the interaction between the viral envelope protein and the host cell membrane, and the cancer therapeutic cilengitide, which inhibits the αvβ3 and αvβ5 integrins involved in angiogenesis
  • Peptide agonists can mimic the effect of natural ligands, activating the target protein and inducing a biological response
  • Examples of peptide agonists include the GLP-1 receptor agonist exenatide for the treatment of type 2 diabetes and the parathyroid hormone (PTH) analog teriparatide for the treatment of osteoporosis

Peptide-based protein-protein interaction modulators

  • Protein-protein interactions (PPIs) play crucial roles in many biological processes and are attractive targets for therapeutic intervention
  • Peptides can be designed to modulate PPIs by mimicking the binding interface of one protein partner, competing with the natural interaction
  • Peptide-based PPI inhibitors can disrupt the formation of protein complexes, preventing their functional activity and downstream effects
  • Examples of peptide-based PPI modulators include the p53-MDM2 interaction inhibitor PMI, which reactivates the tumor suppressor function of p53, and the BCL-2 family inhibitor ABT-737, which induces apoptosis in cancer cells
  • Peptide-based PPI stabilizers can also be developed to promote the formation of specific protein complexes, enhancing their activity and biological effects

Peptide stability and metabolism

Enzymatic degradation of peptides

  • Peptides are susceptible to degradation by various proteolytic enzymes, such as peptidases and proteases, which cleave peptide bonds at specific sites
  • Endopeptidases, such as trypsin and chymotrypsin, cleave peptide bonds within the peptide chain, while exopeptidases, such as aminopeptidases and carboxypeptidases, remove amino acids from the N-terminus or C-terminus, respectively
  • The stability of peptides towards enzymatic degradation depends on their amino acid composition, sequence, and structure
  • Peptides containing certain amino acids, such as proline, glycine, and D-amino acids, are more resistant to proteolysis due to their conformational constraints or lack of recognition by enzymes

Strategies for improving peptide stability

  • Several strategies can be employed to enhance the stability of peptides and protect them from enzymatic degradation
  • Incorporation of non-natural amino acids, such as D-amino acids, β-amino acids, or unnatural side chain modifications, can reduce the susceptibility of peptides to proteolysis
  • Cyclization of peptides, either head-to-tail or through disulfide bridges, can improve their stability by constraining their conformation and reducing the accessibility of cleavage sites
  • Peptide stapling, which involves the introduction of covalent cross-links between amino acid side chains, can stabilize α-helical conformations and increase resistance to proteolysis
  • PEGylation, the attachment of polyethylene glycol (PEG) chains to peptides, can increase their size, hydrophilicity, and steric hindrance, reducing their clearance and enzymatic degradation

Peptide half-life and clearance

  • The half-life of a peptide refers to the time required for its concentration to decrease by 50% in the body
  • Peptides often have short half-lives due to rapid clearance by the kidneys, liver, and other organs, as well as enzymatic degradation in the blood and tissues
  • The clearance of peptides is influenced by their size, charge, hydrophobicity, and binding to plasma proteins
  • Strategies to extend the half-life of peptides include increasing their size (PEGylation, fusion to albumin or Fc domains), reducing their renal filtration (charge modification, glycosylation), and improving their binding to plasma proteins (fatty acid conjugation)

Peptide prodrugs and bioconjugation

  • Peptide prodrugs are inactive precursors that are converted to the active peptide drug by enzymatic or chemical transformations in the body
  • Prodrug strategies can be used to improve the stability, bioavailability, and targeting of peptide drugs
  • Examples of peptide prodrugs include esterification of carboxyl groups to increase lipophilicity and oral absorption, and conjugation to targeting moieties (antibodies, aptamers) for site-specific delivery
  • Bioconjugation involves the covalent attachment of peptides to other molecules, such as polymers, lipids, or small molecules, to modify their pharmacokinetic and pharmacodynamic properties
  • Examples of peptide bioconjugates include peptide-drug conjugates (PDCs) for targeted delivery of cytotoxic agents, and peptide-oligonucleotide conjugates (POCs) for improved cellular uptake and gene silencing

Peptide-based therapies

Peptide hormones and neurotransmitters

  • Peptide hormones are endogenous signaling molecules that are produced by endocrine glands and released into the bloodstream to regulate various physiological processes
  • Examples of peptide hormones include insulin (glucose metabolism), glucagon (blood sugar regulation), oxytocin (social bonding, lactation), and vasopressin (water balance, blood pressure)
  • Peptide are signaling molecules that are released by neurons and bind to receptors on target cells to modulate synaptic transmission and neuronal activity
  • Examples of peptide neurotransmitters include endorphins (pain modulation, reward), substance P (pain sensation, inflammation), and neuropeptide Y (appetite, stress response)
  • Synthetic analogs of peptide hormones and neurotransmitters can be developed as therapeutic agents to treat disorders associated with their dysregulation, such as diabetes, obesity, and neurological diseases

Antimicrobial and anticancer peptides

  • Antimicrobial peptides (AMPs) are a diverse group of peptides that exhibit broad-spectrum activity against bacteria, fungi, and viruses
  • AMPs typically have a cationic charge and amphipathic structure, which allows them to interact with and disrupt the negatively charged cell membranes of microorganisms
  • Examples of AMPs include defensins, cathelicidins, and magainins, which are found in various organisms as part of their innate immune defense
  • Anticancer peptides (ACPs) are peptides that selectively target and kill cancer cells while sparing healthy cells
  • ACPs can act through various mechanisms, such as membrane disruption, apoptosis induction, and inhibition of angiogenesis and metastasis
  • Examples of ACPs include the proapoptotic peptide (KLAKLAK)2, the antiangiogenic peptide anginex, and the cell-penetrating peptide Tat-KLA

Peptide vaccines and immunotherapies

  • Peptide vaccines are designed to elicit an immune response against specific antigens, such as viral proteins or tumor-associated antigens, by presenting peptide epitopes to the immune system
  • Peptide vaccines can be used for the prevention or treatment of infectious diseases, cancer, and autoimmune disorders
  • Examples of peptide vaccines include the HPV vaccine (cervical cancer), the influenza vaccine (seasonal flu), and the gp100 vaccine (melanoma)
  • Peptide-based immunotherapies leverage the ability of peptides to modulate the immune system, either by stimulating an immune response or suppressing an overactive immune reaction
  • Examples of peptide immunotherapies include the T cell receptor (TCR) mimic peptides for cancer treatment, the tolerogenic peptides for autoimmune diseases, and the checkpoint inhibitor peptides for immune activation

Clinical applications and challenges

  • Peptide-based therapies have been successfully applied to the treatment of various diseases, such as diabetes (insulin, GLP-1 receptor agonists), cancer (goserelin, octreotide), and HIV (enfuvirtide)
  • Peptides offer several advantages as therapeutic agents, including high specificity, low toxicity, and the ability to target protein-protein interactions and other challenging targets
  • However, peptide-based therapies also face challenges, such as poor oral bioavailability, rapid clearance, and immunogenicity
  • Strategies to overcome these challenges include the use of peptide anal

Key Terms to Review (19)

Acetylation: Acetylation is a biochemical modification process where an acetyl group ($$C_2H_3O$$) is transferred to a molecule, commonly affecting proteins, nucleic acids, and small molecules. This process can alter the function, activity, or localization of the modified substances, playing a crucial role in various biological processes such as gene expression and enzyme activity.
Affinity: Affinity refers to the strength of the interaction between a ligand and its target receptor or protein. It plays a crucial role in determining how effectively a drug can bind to its target, influencing the overall efficacy and potency of the therapeutic agent. Understanding affinity is essential when designing drugs that will interact with specific biological targets, allowing for better therapeutic outcomes.
Frederick Sanger: Frederick Sanger was a renowned British biochemist known for his groundbreaking contributions to the field of protein sequencing and DNA sequencing. His work in developing methods for determining the amino acid sequences in peptides revolutionized the understanding of protein structure and function. Sanger's innovative techniques, particularly the Sanger sequencing method, paved the way for advancements in molecular biology and genetics.
Hormones: Hormones are chemical messengers produced by glands in the endocrine system that travel through the bloodstream to target organs, where they regulate various physiological processes. They play crucial roles in metabolism, growth and development, tissue function, and mood regulation, acting as key components in maintaining homeostasis within the body.
Mass Spectrometry: Mass spectrometry is an analytical technique used to measure the mass-to-charge ratio of ions, allowing for the identification and quantification of molecules in a sample. This method is essential in various fields for characterizing chemical compounds, analyzing biomolecules, and determining molecular structures, making it invaluable in early drug development, chemical property assessments, and exploring complex mixtures such as natural products.
Neurotransmitters: Neurotransmitters are chemical messengers that transmit signals across synapses from one neuron to another or to a target cell, playing a crucial role in the nervous system. They influence a variety of physiological functions, including mood, sleep, and muscle contraction, and can have excitatory or inhibitory effects on neuronal activity. These molecules are essential for communication within the brain and throughout the body.
Nmr spectroscopy: NMR spectroscopy, or nuclear magnetic resonance spectroscopy, is a powerful analytical technique used to determine the structure of organic compounds by observing the magnetic properties of atomic nuclei. This method provides detailed information about the molecular structure, dynamics, and environment of atoms, making it an essential tool in various fields including medicinal chemistry, where understanding molecular interactions is crucial for drug development and design.
Oligopeptides: Oligopeptides are short chains of amino acids, typically consisting of 2 to 20 amino acid residues linked by peptide bonds. These small peptides play crucial roles in various biological processes, including hormone regulation, cellular signaling, and immune response. Their unique properties and functions make them essential components in the study of proteins and biological interactions.
Peptidomimetics: Peptidomimetics are synthetic compounds that mimic the structure and function of peptides, often designed to enhance biological activity and stability while overcoming the limitations of natural peptides. By modifying the peptide backbone or side chains, these molecules can improve oral bioavailability, resistance to enzymatic degradation, and selectivity for specific biological targets. This approach is crucial in drug development, where peptidomimetics can serve as more effective therapeutics than their natural counterparts.
Phosphorylation: Phosphorylation is the process of adding a phosphate group (PO₄³⁻) to a molecule, typically a protein, which can alter the molecule's function and activity. This modification plays a crucial role in various cellular processes, including signal transduction pathways, where it can activate or deactivate proteins and enzymes, ultimately affecting cellular responses. Additionally, phosphorylation is vital in regulating peptide functions and interactions within cells.
Polypeptides: Polypeptides are long chains of amino acids linked together by peptide bonds, which form the building blocks of proteins. These chains can vary in length and sequence, leading to a diverse range of structures and functions essential for biological processes. Polypeptides can fold into specific three-dimensional shapes, which are crucial for their activity and interaction with other molecules.
Primary structure: Primary structure refers to the unique sequence of amino acids that make up a peptide or protein. This sequence is determined by the genetic code and is crucial for the protein's overall shape and function, as even a single change in the amino acid sequence can significantly alter how a peptide behaves.
Robert H. Grubbs: Robert H. Grubbs is a prominent American chemist known for his significant contributions to the field of organic chemistry, particularly in the development of metathesis reactions. His work has greatly influenced the synthesis of peptides and other complex molecules, making it easier to create and manipulate these substances in medicinal chemistry and related fields.
Secondary structure: Secondary structure refers to the local folded structures that form within a polypeptide chain due to interactions between the backbone atoms in the amino acid sequence. These structures are primarily stabilized by hydrogen bonds and can manifest as alpha helices and beta sheets, both of which are crucial for the overall stability and functionality of proteins derived from peptides.
Selectivity: Selectivity refers to the ability of a drug or compound to preferentially bind to a specific target, such as a receptor or enzyme, while minimizing interactions with other targets. This characteristic is crucial for enhancing therapeutic efficacy and reducing side effects, making it a central concept in drug design and optimization processes. Understanding selectivity is essential for developing drugs that provide maximum therapeutic benefit while limiting undesirable effects on non-target systems.
Solid-phase synthesis: Solid-phase synthesis is a method used to construct molecules by attaching them to a solid support, which simplifies purification and isolation of the desired products. This technique is especially useful in combinatorial chemistry, allowing for the rapid generation of diverse chemical libraries, as well as in the synthesis of peptides, where sequences can be assembled efficiently on a solid substrate.
Solution-phase synthesis: Solution-phase synthesis is a method of chemical synthesis that involves the formation of compounds in a liquid solution rather than on a solid support. This approach allows for a wide range of reactions to occur, facilitating the creation of complex molecules like peptides through simpler and more efficient processes, while also providing the ability to easily monitor and manipulate reaction conditions.
Tertiary structure: Tertiary structure refers to the overall three-dimensional shape of a polypeptide chain, determined by interactions between the side chains of the amino acids. This level of structure is crucial for the protein's functionality, as it dictates how the protein will interact with other molecules. The tertiary structure is stabilized by various types of bonds and interactions, including hydrogen bonds, ionic bonds, hydrophobic interactions, and disulfide bridges, which play significant roles in the properties and behavior of peptides.
Therapeutic peptides: Therapeutic peptides are short chains of amino acids that have specific biological activities and can be used in medical applications to treat various diseases. These peptides can mimic naturally occurring hormones, neurotransmitters, or other bioactive molecules, providing a mechanism for targeted therapy. Their unique properties make them suitable for drug development, as they can offer high specificity and lower side effects compared to traditional small molecule drugs.
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.