Cyclic Peptides in Pipeline: Comparison
Please note this is a comparison between Version 1 by Carla Fernandes and Version 2 by Camila Xu.

Cyclic peptides are molecules that are already used as drugs in therapies approved for various pharmacological activities, for example, as antibiotics, antifungals, anticancer, and immunosuppressants. Interest in these molecules has been growing due to the improved pharmacokinetic and pharmacodynamic properties of the cyclic structure over linear peptides and by the evolution of chemical synthesis, computational, and in vitro methods.

  • bioactivity
  • clinical trials
  • cyclic peptides
  • cyclization
  • pipeline

1. Introduction

Peptides, molecules that contain two or more residues of amino acids linked by an amide bond [1], can be considered to fall between small molecules and large biological molecules, such as proteins or antibodies [2]. Peptides have several advantages over small molecules used in conventional therapy, such as high selectivity, potency, biotarget specificity, few side effects, and low accumulation in tissues [3][4][3,4]. When compared to proteins and antibodies, peptides have the advantage of a lower immunogenicity [2].
Over the years, the therapeutic potential of peptides has been exploited for a broad spectrum of biological activities, such as antimicrobial, antihypertensive, antioxidant, anticancer, antidiabetic, and anti-inflammatory, among others, which attract the attention of the pharmaceutical [5][6][7][5,6,7], cosmeceutical [8], and nutraceutical [9][10][9,10] industries.
The era of therapeutic peptides began with the first medical administration of insulin in the 1920s [11]. This discovery revolutionized the treatment of patients with type I diabetes, and peptides were seen as potential therapeutic tools [12]. About 40 years later, the first hormones used in clinical practice, oxytocin, and vasopressin, were synthesized [13]. Some industrial groups have dived into this field, and the interest in this type of molecules increased [14]. While the advantages of using these molecules were explored, their limitations also emerged at a time when the discovery and development of small molecules was at its peak. This circumstance has led to the stagnation of research on peptides as drugs. Despite this, peptides continued to be explored as tools for the study of targets, and in the 1980s, the interest in these molecules returned backed by biotechnology companies [14]. Since then, the tendency of approved peptides as therapeutic agents has been increasing [5].
In addition to their therapeutic potential, peptides can also be used in imaging and disease diagnosis [5]. For example, it is known that peptides composed of arginine-glycine-aspartate (RGD) moiety have an affinity to bind to integrins, which are heterodimeric receptors that play pivotal roles in cells. By binding to integrin, RGD peptides can prevent angiogenesis, a process involved in diseases, such as cancer and rheumatoid arthritis [15][16][17][15,16,17]. These peptides can also be used for tumor imaging when linked to radioisotopes or even to create tumor-targeted drug delivery systems reducing the adverse effects inherent to conventional chemotherapy [18][19][18,19].
Until May 2023, one hundred and fourteen peptides have been approved by the regulatory authorities as therapeutic agents (Figure 1), which included pharmaceuticals and diagnostic tools [20].
Figure 1.
Evolution of approved peptide drugs over the years.
Among the approved peptides, the cyclic peptides represent 46% of the total approvals (Figure 2A) [21]. Gramicidin S (antibiotic) was the first cyclic peptide to be used as a drug. Its discovery in 1944 by Gause and Brazhnikova during the Second World War and its use in Soviet military hospitals revolutionized the field of cyclic peptides [22][23][22,23]. Other interesting examples are telavancin, dalbavancin, and oritavancin (semi-synthetic cyclic lipoglycopeptide antibiotics) [24], anidulafungin (from the class of echinocandin antifungals) [25], lanreotide, pasireotide, and romidepsin (anticancer drugs), and linaclotide (derived from an enterotoxin for gastrointestinal (GI) disorders) [26]. The last approved cyclic peptide was rezafungin (antifungal analog of anidulafungin), being approved by Food and Drug Administration (FDA) in 2023 [27]. This drug is administered orally and has a half-life of 30 h, which represents an advance in pharmacokinetic characteristics in comparison with other peptide drugs. In fact, regarding the routes of administration of approved peptides (Figure 2B), parenteral administration is the most frequent, of which the intravenous route is the most recurrent [21].
Figure 2. Comparison between approved cyclic and linear peptides (A). Routes of administration of approved peptides (B). IV: Intravenous; IM: Intramuscular; SC: Subcutaneous.
Another relevant application of peptides is in drug delivery, considering the good hydrophilicity/hydrophobicity ratio, as well as intra and intermolecular interaction of amino acids by weak non-covalent bonds, which makes them capable of organizing themselves to form nanostructures. Peptide nanostructures have demonstrated a great drug load capacity and drug protection and are responsive to external stimuli [28]. Recently, new cyclic peptides exhibiting nanospherical structures demonstrated the ability to form stable complexes with short-interfering RNA (siRNA), proving to be a promising tool in nucleic acid delivery for cancer treatment, as an example [29]. Moreover, cyclic peptides, such as vancomycin, teicoplanin, and ristocetin (macrocyclic antibiotics), in addition to their therapeutic actions, were also explored as chiral stationary phases for chromatographic applications [30][31][30,31]. This application of macrocyclic antibiotics in liquid chromatography was introduced by Armstrong et al. in 1994 [32]. The high number of stereogenic centers and the macrocyclic structure of these peptides allow a variety of interactions with the analytes to enantioseparate and the possibility of forming inclusion complexes, which contribute to their high capacity of chiral recognition [33][34][33,34]. Cyclic peptides can be obtained from natural sources, both terrestrial and marine [35][36][35,36]. With regard to terrestrial sources, these can be from animal origin [37], such as venoms [38] (a rich source of bioactive peptides revised in [39]), plants [40], microorganisms [41], among others). Examples of peptides obtained from terrestrial sources are the antibiotics vancomycin (isolated from the soil bacterium Amycolatopsis orientalis) [42], daptomycin (from the soil bacterium Streptomyces roseoporus) [43], teixobactin (from the soil bacterium Eleftheria terrae) [44], and apamin (isolated from bee Apis mellifera) [45]. Bioactive marine cyclic peptides can be found in marine tunicates [46][47][46,47], sponges [47][48][47,48], algae [49], bacteria [50][51][50,51], cyanobacteria [52][53][52,53], fungi [54][55][54,55], and other invertebrates [56], including symbionts [57] and non-symbiotic microorganisms, such as sponge-associated fungi [58][59][58,59]. Marine-derived peptides display a broad spectrum of bioactivities [60][61][60,61], mainly anticancer [62] and antimicrobial [63] being one of the research topics that gives a very high output, with a considerable increase in the number of publications (268 per year), from 2010 to 2020 [64]. To highlight that, one marine cyclic peptide-derived drug has reached the market—ziconotide [65] (the first FDA-approved marine peptide, in 2004). Chemical synthesis is also a remarkable source of peptides, which allows obtaining an appropriate amount of compound to carry out further large-scale biological assays, including studies of the mechanism of action, pharmacokinetics, toxicity, and others [66][67][68][69][66,67,68,69]. In the literature, several reports can be found describing synthetic routes for peptides, including very large and complex structures [70][71][70,71]. In addition, the synthesis allows for obtaining the structurally diverse analogs and derivatives of the natural peptides with improved properties for structure–activity relationship (SAR) studies [72]. The synthetic strategies for molecular modifications can include the following: (1) incorporation of non-proteinogenic amino acids to prevent proteolysis of peptides [73]; (2) acetylation of the N-terminus of short peptides to increase peptidase stability in serum and, consequently, enhance the half-life [74]; (3) glycosylation to improve protein–protein interaction, protein permeability, metabolic stability, and bioavailability [75]; lipidation to enable the binding to a carrier serum protein and, consequently, enhance the half-life, among others [76][77][76,77]. Typically, there are two strategies to synthesize peptides: solution-phase and solid-phase peptide synthesis (SPPS) [63]. They include two key steps in the formation of a peptide bond between two amino acids: the activation of the carboxyl group by coupling agents and the use of temporary protecting groups to direct the reaction to the desired direction [78]. The two most used strategies are fluorenylmethyloxycarbonyl (Fmoc)/tert-butyl (tBu) and tert-butyloxycarbonyl (Boc)/benzyl (Bn) strategies [79]. In addition to classical peptide synthesis techniques [69], significant efforts have been carried out for the introduction of sustainable and innovative processes for synthesis and purification methodologies [80][81][80,81]. Despite several advantages of peptides, they also have characteristics that are disadvantageous when they are used for drug development, mainly concerning pharmacokinetic issues: (1) they have a short half-life in the plasma because of the action of peptidases; (2) are easily degraded by enzyme actions and pH hydrolysis on the GI tract, which makes them not bioavailable orally; (3) have low membrane permeability, which makes its passage through membranes on absorption locals and intracellular biotargets difficult [2]. Nevertheless, the pharmacokinetic properties of these molecules can be improved with some strategies of molecular modifications, such as conjugation with polyethylene glycol (PEG), albumin, or proteins, as well as other approaches, such as cyclization [14]. Cyclization of peptides has proven to be an asset in enhancing the advantages of linear peptides, but also a way to overcome their disadvantages. Cyclization of a peptide reduces the spatial vibrations of the molecule leading to a decrease in conformational changes. In addition, cyclization induces an increase in the surface area available for interaction with the biological target. These two reasons lead to an increase in binding affinity and selectivity to the target [4]. In addition to pharmacodynamic considerations, the pharmacokinetic properties of peptides can also be improved by cyclization, as the rigidification of the structure leads to a lowering of the energy barrier required for the peptide to adapt to the membrane environment and bind to transport proteins to enter the cell by passive diffusion or active transport. Thus, cyclization can improve the absorption and membrane permeability of peptides [5]. Cyclization of peptides also gives greater metabolic stability, as cyclic peptides are resistant to the action of exopeptidases, due to the lack of terminal amine and carboxylic acid groups, and endopeptidases, by blocking the access to the cleavage site [19]. Although cyclization may improve the pharmacokinetic properties of peptides, it is important to highlight that for many cyclic peptides, the poor pharmacokinetic parameters are one of the main reasons for failure in phase I/II trials [82]. Through chemical synthesis, it is possible to carry out molecular modifications to obtain derivatives with improved characteristics [14].

2. Cyclic Peptides in the Pipeline

Currently, there are various cyclic peptides in clinical development to cover such diverse medical conditions as cancer, infectious diseases, and hematological disorders, among others. Table 1 summarizes 27 cyclic peptides in clinical trials, including their generic names, therapeutic indication(s), current stage of development, name of the pharmaceutical company responsible for the development, and source (natural or synthetic).
Table 1.
Cyclic peptides currently in clinical trials.
Video Production Service