Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jun 1.
Published in final edited form as: Curr Opin Struct Biol. 2023 May 12;80:102603. doi: 10.1016/j.sbi.2023.102603

Cell-permeable chameleonic peptides: exploiting conformational dynamics in de novo cyclic peptide design

Theresa A Ramelot 1, Jonathan Palmer 2,3, Gaetano T Montelione 1,*, Gaurav Bhardwaj 2,3,*
PMCID: PMC10923192  NIHMSID: NIHMS1900376  PMID: 37178478

Abstract

Membrane-traversing peptides offer opportunities for targeting intracellular proteins and oral delivery. Despite progress in understanding the mechanisms underlying membrane traversal in natural cell-permeable peptides, there are still several challenges to developing designed membrane-traversing peptides with diverse shapes and sizes. Conformational flexibility appears to be a key determinant of membrane permeability of large macrocycles. We review recent developments in the design and validation of chameleonic cyclic peptides, which can switch between alternative conformations to enable improved permeability through cell membranes, while still maintaining reasonable solubility and exposed polar functional groups for target protein binding. Finally, we discuss the principles, strategies, and practical considerations for rational design, discovery, and validation of permeable chameleonic peptides.

Introduction

Advances in high-throughput screening methods, rational design, and formulation strategies have enabled the development of peptide-based lead candidates against diverse therapeutic targets considered non-ideal for conventional drug modalities [15]. However, the limited cell permeability, oral bioavailability, and pharmacokinetics of peptides pose challenges to their translation from research tools to therapeutics [6,7]. Consequently, there has been a renewed focus on identifying general principles for developing peptides with enhanced cell penetration, oral bioavailability, and physiologic stability, while retaining reasonable solubility and target binding specificity. Naturally occurring cell-penetrating peptides exploit direct translocation, endocytosis, and passive diffusion for cell permeability (reviewed in ref [8]). The underlying chemical and structural requirements for leveraging these routes rely on size, shape, polarity, and chemical composition. The role of amphipathic and polycationic cell-penetrating peptides in endocytosis has been reviewed extensively [812]. Here we focus on passive permeability and, specifically, on the role of conformational switching in passive membrane traversal.

Generally, peptides with low polar surface area (PSA) show an improved passive permeability [6,1316]. Low PSA can be achieved by masking backbone amides through internal hydrogen bonding, N-methylation, or shielding by bulky nonpolar amino-acid side chains [7,1621]. Many research groups over the last two decades have focused on exploring the limits of passive permeability and defining the features that promote passive diffusion [14,1822]. The rates of passive permeability depend on size, polarity, and conformational flexibility [6,13,15,23]. The constraints on these properties are conceptually similar to those of small molecules, defined by Lipinsky’s rule of five (Ro5) [24]. However, peptides are generally larger and have more hydrogen bond donors and acceptors than the limits prescribed by the Ro5. Features governing their permeability are more complex [7,8]. Although most of these studies focused on small macrocycles composed of 4–7 amino acids, recent work by Lokey and coworkers has identified larger membrane-traversing peptides with molecular weights (MW) greater than 1000 Da [25,26]. We recently described a de novo design approach for accurately designing membrane-traversing macrocycles with diverse shapes and sizes between 6–12 amino acids and MW up to 1400 Da, which have conformations with backbone amides forming internal hydrogen bonds [27].

Unfortunately, the same features that increase passive permeability also lead to excessive hydrophobicity, resulting in poor solubility and decreased target selectivity [28]. To address this problem, chameleonic peptides are under development that can adopt significantly different conformations in the aqueous and membrane environments: the more polar conformation inside the cell with an exposed backbone allows for hydrogen bonding interactions with the solvent or the target of interest, and a less polar state within the membrane with all (or most) polar groups satisfied or shielded favors membrane traversal. Cyclosporin is the prototypical example, with chameleonic character that enables it to overcome the permeability barriers [29]. Multiple structural and in silico molecular dynamics (MD) studies of the 11-amino acid macrocycle cyclosporin in different solvents and membranes have highlighted the critical role of alternate conformations and conformational dynamics in its permeability [30,31]. However, developing new chameleonic membrane-traversing peptides in medium-to-high throughput will require structure-guided computational tools, high-throughput methods for parallel permeability measurements, and methods for structural characterization of multiple conformations.

Computational design of conformationally-switching peptides

In silico methods that calculate the difference in solvation free energy for a predicted low-energy conformer (or ensemble of conformers) going from a high-dielectric to low-dielectric solvents have been used to predict the cell permeability of macrocycles [3234]. Monovich and coworkers combined such methods with analysis of intermolecular hydrogen bonds to design and synthesize orally bioavailable thioether-cyclized macrocycles between 6–8 amino acids [35,36]. Accurate low-energy conformations required for such permeability predictions can be generated using de novo design approaches, MD simulations, or deep-learning-guided models [3739].

De novo peptide design methods offer opportunities to systematically evaluate structural features that could improve permeability. We have previously described computational methods for accurately designing peptides composed of canonical and noncanonical amino acids using Rosetta [38]. This approach relies on extensive in silico backbone sampling followed by the design of amino acid sequences that minimize the energy—as calculated by the Rosetta energy function—for each selected backbone. We recently expanded this method to design 6–12 amino acid membrane-traversing macrocycles that included N-methylated amino acids; a subset of which were designed to switch conformations in a solvent-dependent manner (Figure 1A-C) [27]. Generally, computational design methods rely on sampling structures with a single deep well in their energy landscapes [38]. In contrast, for chameleonic peptide design, we focused on generating designs with two or three distinct isoenergetic states in their structure-energy landscapes (Figure 1C). Other key criteria is to have one of the conformations with all backbone amides hydrogen-bonded or N-methylated for favorable membrane permeability [27]. We relied on cis-trans isomerization of peptide bonds for achieving multiple conformations and switching in our designed macrocycles. However, other strategies could also be employed to drive conformation-switching, including metal binding [40], light-sensitive cross-linkers [41], pH-dependent changes to sidechain protonation, or oligomerization. These switching mechanisms may involve different timescales and require appropriate experimental methods for validating multiple states. Improvements in the accuracy of energy functions are needed to further increase confidence in the calculations of isoenergetic states and improve the predictability of peptide designs with multiple local energy minima.

Figure 1. Computational Design of Chameleonic Peptides.

Figure 1.

A. Proposed method for passive permeability in chameleonic peptides. Macrocyclic peptides exist as an equilibrium of conformations. When contacting a hydrophobic cell membrane, the equilibrium shifts towards the lowest PSA conformation, providing passive permeability. B. Computational design of macrocyclic peptides begins by generating thousands, and sometimes millions, of designs. This library of designs is filtered by scoring metrics. C. Energy prediction landscapes help identify chameleonic peptides. Traditional design calls for a “funnel” shape, resulting in a single low energy minima. Chameleonic design looks for two or more deep energy minima. D. Transwell assays provide in vitro data on permeability. Peptides added to the donor well diffuse across the membrane and into the acceptor well. Mass spectrometry measures the amount of crossing to estimate permeability. E. Macrocyclic peptides that demonstrate permeability are structurally validated using NMR and X-ray crystallography. NMR is capable of capturing the signatures of multiple peptide conformations as they interconvert in solution. In some cases, different solvents can be used to shift the equilibrium towards one conformation or another. Similarly, different crystallography solvents can be used to capture static images of chameleonic peptides in alternative conformations.

MD simulations provide an orthogonal approach to Rosetta-based methods and enable studies of chameleonic character [4246]. Lin and coworkers have used MD simulations in their successful structure prediction and design of chameleonic peptide macrocycles [42,47], including peptides with N-methylated amino acids to promote membrane permeability [48]. MD trajectories can also predict the relative populations of conformers in solution as was done for cyclosporin [37,42,43,49]. In principle, MD can be used to model the conformations of peptides in different solvents or membranes with different compositions and predict permeability [50]; however, such calculations or simulations are computationally very intensive and cannot currently be used to screen thousands of design models [43]. Going forward, approaches that combine MD simulations with machine learning (ML) could provide the basis for rapid and accurate design of chameleonic cell-permeable macrocycles [37].

ML-based structure prediction networks for larger proteins have seen tremendous success over the last few years [51,52]. Although these deep learning methods may not generalize to small peptides as they are trained on larger protein structures composed mostly of natural L-amino acids [51,52], recent work by Gulsevin and Meiler suggests some limited predictive value for predicting small, rigid polypeptide structures [53]. Approaches that combine information and training from small molecules and proteins could guide ML methods for designing new peptides with desired properties, such as membrane permeability and oral bioavailability. Data from computationally-intensive QM calculations or MD simulations can also be used as training sets to develop ML models that can predict the states populated by a given amino acid sequence without running computationally expensive molecular simulations for each new peptide [37]. It remains to be seen how the accuracy of such methods translates to sequences that differ substantially from the training set in their chemical and structural diversity. Another potential approach is to skip structure prediction and train models that predict a property, such as membrane permeability, from the primary sequence alone. However, it may be challenging to deconvolute the diverse mechanisms and structural principles that underlie permeability from such sequence-driven property prediction models. Given the recent transformative success of generative models for larger proteins [54], an important area to explore will be the development of such models—simultaneously trained on proteins, small molecules, and peptides—to hallucinate polypeptides with desired chemical, structural, and functional properties.

Experimental methods for testing permeability:

There are several approaches for measuring cell penetration of peptides by tagging them with fluorescent dyes or transcriptional modulators [55], and tracking the intracellular localization by fluorescence microscopy [56,57], flow cytometry [58], fluorescence correlation spectroscopy [59], or some transcriptional readout [57,60]. However, there are concerns about the throughput of these assays and the influence of the tag on the permeability, solubility, and overall structure of the peptide being tested. Newer methods that use smaller tags [61,62] and rely on fluorescence measurements or cell sorting address some of these concerns, and enable higher throughput in permeability measurements. Alternatively, mass spectrometry (MS)-based transwell assays, such as Parallel Artificial Membrane Permeation Assay (PAMPA), enable tag-free measurement of permeability across a lipid membrane barrier between the donor and acceptor wells [63] (Figure 1D). MS-based quantification allows for high-throughput permeability measurements of thousands of peptides; however, PAMPA can be only be used for measuring passive membrane traversal. In contrast, transwell Caco-2 and MDCK assays use intestinal epithelial cells and canine kidney cells as permeation barriers [64] to appropriately account for efflux in permeability measurements, and are very useful in predicting oral bioavailability in vivo. Overall, these diverse set of established methods for measuring permeability and traversal across different cell types provide the basis for developing design principles for accessing specific cell and tissue types.

Dynamic structural analysis of chameleonic peptides

Structure determination can provide validation and new insights critical for de novo protein design (Figure 1E). Often chameleonic peptides can be crystallized from solvents of different polarities, and 3D conformations determined using X-ray crystallography. However, the crystal lattice may favor particular conformations, and it may not be possible to capture the full range of conformations present in the solution. Solution NMR studies can provide accurate structures of multiple conformations in dynamic equilibrium, as well as additional information about populations, relative free energies, and rates of interconversion between conformational states. Particularly for studies of multiple conformations, many research groups have turned to NMR studies in polar and nonpolar solvents.

NMR is especially useful for assessing intramolecular hydrogen bonds, which are essential for the characterization of chameleonic states. These include measurements of amide proton temperature coefficients by variable temperature NMR, amide hydrogen/deuterium exchange (HDX) rates, and in the case of a faster exchange, amide proton saturation transfer [65,66]. In some cases, hydrogen-bond analysis alone may provide the key insights needed for guiding design.

Solvent-dependent conformational changes are easiest to identify by 1D 1H NMR in cases where two or more distinct populations interconvert slowly on the chemical shift timescale (e.g., longer lived than ~ 10 ms for typical 1H chemical shift differences between conformers at 600 MHz), resulting in at least double the number of expected resonances (Figure 2A). Chameleonic peptides involving peptide bond isomerization typically have slow enough exchange rates to provide a discrete set of resonances for each peptide bond conformer. Relative populations of different conformations at equilibrium can typically be determined by integration of peaks, and conformational switching can be monitored by comparing spectral data recorded in different solvents and temperatures.

Figure 2. Solvent-Dependent Conformational Switching.

Figure 2.

A. 1D 1H-NMR data for chameleonic cyclic peptide D9.16 collected in CDCl3 and DMSO-d6 at 20 oC and 800 MHz. B. 2D ROESY (250 ms mixing time) data in CDCl3 with schematic showing how CYANA was used to model two conformations simultaneously from the complete ROESY peak list, chemical shifts, and 3JHNHA -based dihedral restraints, using tandem cyclic peptide sequences representing the between the trans-trans (55%) and cis-trans (45%) conformations (lower case residues are D-amino acids, * denotes N-methylated amino-acid positions, cP is cis L-Pro-8, and (xxx)3 denotes a 9-residue sterically-transparent linker). Molar percentages given in parenthesis were determined from averaged peak integrations of the 1D spectra. C. Design models and experimentally-determine structures (X-ray and NMR) for different conformational states of D9.16. The design models are in the top row. The middle row shows the NMR structure in polar solvent DMSO-d6 and the X-ray crystal structure, which both match the trans-cis design model (upper right) with 3 unsatisfied hydrogen bonds. In non-polar solvent (CDCl3), two slowly-exchanging states in equilibrium were observed (bottom panel), a trans-cis state that matches the structures observed in DMSO-d6, X-ray crystallography, and the trans-cis design model, and a trans-trans state that matches closely the trans-trans design model, with all backbone NHs satisfied. These studies show that the population of the trans-cis state observed in the X-ray crystal structure decreases from 100% in polar solvent to 45% in non-polar solvent, as 3 NH groups are buried by intra-peptide hydrogen bonds, providing the basis for conformational-switch-mediated membrane permeability.

When there is only a single set of chemical shifts(Figure 2A), NMR structure determination can be pursued with traditional approaches using restraint-satisfaction structure-generation methods and software such as CYANA [67], CNS [68], Xplor-NIH [69], and Surflex [70]. NMR-derived data typically includes chemical shift assignments, NOESY/ROESY distance restraints, and dihedral angles constraints from 3J coupling constants. Residual dipolar coupling (RDCs), amide proton temperature coefficient, and amide hydrogen exchange data are also used, as reviewed recently [66]. For chameleonic peptide D9.16 in DMSO, a standard CYANA protocol was used to interpret and calibrate NOE-based constraints and to model an ensemble of low energy conformers that satisfy the NOE and dihedral restraints (Figure 2B) [27].

In the case of slow-exchanging multiple conformations, structure analysis is further complicated because the data for both conformations are present in NMR data collected from a single sample (Figure 2B). Chemical shifts and NOE/ROE restraints need to be separated for each set of slow-exchanging isomeric conformers. If the ratios of two switching conformations are close to unity, NOEs for both forms can be deconvoluted from NOESY or ROESY peak lists simultaneously, as was done for cyclic peptide D9.16 in CDCl3 (Figure 2). Difficulties arise when one conformation is significantly less populated than the other, as the subsequent calibration of NOE peak volumes (or intensities) into distance restraints needs to be appropriately population-weighted. However, it is often impossible to determine structures for low abundance conformations, where through-bond (TOCSY) and through-space (NOESY) cross peaks may be obscured by the more abundant conformer.

Even when the data can be interpreted as a single conformation that satisfies all of the NMR-based distance restraints, the molecule may in fact adopt multiple conformations in fast exchange, resulting in population-weighted averaging for all the peaks in the NMR spectra. This is further complicated by the fact that peptide systems may provide only a small number of NOESY/ROESY peaks, resulting in a sparse network of distance restraints, which can be satisfied by a single-conformation model even when the underlying system is dynamic. One potentially powerful approach to this challenge is NMR Analysis of Molecular Flexibility in Solution (NAMFIS) [71], in which modeling is used to generate a ‘complete’ structural ensemble, and back-calculated NMR data (e.g. NOEs and J coupling) are used to rule in or rule out members of this predicted ensemble and to fit relative populations, as in recent studies of macrocyclic drugs [72,73]. In a similar way, advanced conformational sampling by the program ForceGen can be used to obtain an ensemble of NMR-validated structures [70,74]. A related approach by the Lokey lab used only 1H and 13C chemical shift data, without any NOESY/ROESY data, to simultaneously determine the structures of both major and minor population cyclic peptides [75]. Here, Quantum Mechanics (QM) was used to back-calculate population-weighted chemical shifts for a family of MD-generated conformers, allowing identification of the conformational ensemble best-fitting the NMR data. Such MD and QM approaches are computationally expensive, but can exploit ergotic ensemble-averaged chemical shifts. However, it is important to ensure comprehensive conformational sampling when using such modeling methods.

Another challenge for NMR studies is determination of molecular topology and templates for non-canonical amino acids, such as N-methylated and D-amino acids, and cyclization. The Automated Topology Builder (ATB) server has been developed to address some of these problems [76]. Larger polypeptides and miniproteins have greater spectral overlap, making simultaneous structural analysis of multiple conformations even more challenging. This problem is addressed in proteins by biosynthetic enrichment with 13C and 15N, allowing for additional dimensional separation in 3D/4D NMR data collection. Unfortunately, in the case of synthetic chameleonic peptides containing non-canonical amino acids, it can be prohibitively expensive to obtain labeled precursors for incorporation into peptides.

Although there are many advantages in NMR studies of slowly-exchanging multiple conformations, these methods are generally slow and expensive. HDX-MS is another powerful approach for characterizing the solvent-exposure of amide protons, as highlighted by recent studies of cyclosporin [77,78]. Here, conformers of cyclosporin analogs were separated using liquid chromatography and further separated in the gas phase by ion mobility (IM) or shape differential mobility spectroscopy (DMS), prior to MS/MS detection [77,78]. HDX-MS was used to determine the amount of gas-phase HDX, which correlated with differences in intermolecular hydrogen bonding. A related approach is pulsed HDX-MS, which has been used to investigate amide hydrogen-bonding behavior in proteins and peptides (reviewed in [79]). These MS approaches can potentially be extended to high-throughput analysis of cyclic peptides, providing key insights prior to more extensive NMR studies.

Conclusions

Conformational flexibility has emerged as a key feature for peptides with enhanced passive permeability [6,73]. Chameleonic character provides a path towards addressing the critical challenge of balancing the overall hydrophobicity required to cross non-polar cellular membranes with reasonable solubility and polarity required to bind therapeutic targets. However, translating the idea of conformational flexibility to a broadly-applicable design principle or a prediction metric for membrane-traversing peptides will require designing and testing tens of thousands of peptides for permeability and conformational flexibility. Recent advances in structure-guided design methods have enabled precise design of membrane-traversing and orally bioavailable peptides [27,35]. These computational methods enable rapid enumeration of the structural and chemical diversity of chameleonic peptides; however, they will need to be integrated with high-throughput methods for measuring membrane permeability and chameleonic behavior. Advances in the peptide library screening approaches [2,80] and high-throughput methods for measuring permeability [26,61,62] have been leveraged to diversify validated membrane-traversing peptides and explore their mutational tolerance. Biophysical studies of the conformational dynamics of chameleonic behavior present unique opportunities and challenges, as outlined above. High-throughput HDX-MS studies can potentially be leveraged as an initial coarse-grained filter for structural flexibility prior to more extensive characterization with NMR and X-ray crystallography. Future studies that combine rational design with high-throughput experimental methods will be instrumental in generating extensive scaffold sets of membrane-traversing chameleonic peptides. While the application of machine learning for peptide design is still in its infancy [53], we will see a larger role going forward of such methods in both rational design and NMR data interpretation. The key will be to integrate these diverse approaches to develop chameleonic peptides that can bind to intracellular therapeutic targets with high potency and selectivity while retaining membrane permeability, solubility, and metabolic stability.

Acknowledgments

We thank Prof. P. Güntert for helpful advice on handling non-canonical amino acid residues in CYANA. We thank members of the Montelione lab, Bhardwaj lab, and the UW Institute for Protein Design for helpful discussions. This research was supported by NIH NIGMS grant 5T32GM007750-43 (to JP) and grant R35-GM141818 (to GTM) from the National Institutes of Health. GB is supported by funds from DARPA, DTRA, HHMI Emerging Pathogens Initiative, and start-up support from the UW Department of Medicinal Chemistry.

Footnotes

G.T.M. is a cofounder, shareholder, and consultant of Nexomics Biosciences, Inc., a structural biology contract research organization. G.B. is the scientific cofounder and shareholder of Vilya, biotech company focused peptide drug discovery.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Drucker DJ: Advances in oral peptide therapeutics. Nat Rev Drug Discov 2020, 19:277–289. [DOI] [PubMed] [Google Scholar]
  • 2.Vinogradov AA, Yin Y, Suga H: Macrocyclic Peptides as Drug Candidates: Recent Progress and Remaining Challenges. J Am Chem Soc 2019, 141:4167–4181. [DOI] [PubMed] [Google Scholar]
  • 3.Hosseinzadeh P, Watson PR, Craven TW, Li X, Rettie S, Pardo-Avila F, Bera AK, Mulligan VK, Lu P, Ford AS, et al. : Anchor extension: a structure-guided approach to design cyclic peptides targeting enzyme active sites. Nat Commun 2021, 12:3384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Mulligan VK, Workman S, Sun T, Rettie S, Li X, Worrall LJ, Craven TW, King DT, Hosseinzadeh P, Watkins AM, et al. : Computationally designed peptide macrocycle inhibitors of New Delhi metallo-β-lactamase 1. Proc Natl Acad Sci U S A 2021, 118:e2012800118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cardote TAF, Ciulli A: Cyclic and Macrocyclic Peptides as Chemical Tools To Recognise Protein Surfaces and Probe Protein-Protein Interactions. ChemMedChem 2016, 11:787–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Over B, Matsson P, Tyrchan C, Artursson P, Doak BC, Foley MA, Hilgendorf C, Johnston SE, Lee MD 4th, Lewis RJ, et al. : Structural and conformational determinants of macrocycle cell permeability. Nat Chem Biol 2016, 12:1065–1074. [DOI] [PubMed] [Google Scholar]
  • 7.Nielsen DS, Shepherd NE, Xu W, Lucke AJ, Stoermer MJ, Fairlie DP: Orally Absorbed Cyclic Peptides. Chem Rev 2017, 117:8094–8128. [DOI] [PubMed] [Google Scholar]
  • 8.Peraro L, Kritzer J: Getting in: emerging methods and design principles for cell-penetrant peptides. Angew Chem Int Ed Engl 2018, doi: 10.1002/ange.201801361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bechara C, Sagan S: Cell-penetrating peptides: 20 years later, where do we stand? FEBS Lett 2013, 587:1693–1702. [DOI] [PubMed] [Google Scholar]
  • 10.Kauffman WB, Fuselier T, He J, Wimley WC: Mechanism Matters: A Taxonomy of Cell Penetrating Peptides. Trends Biochem Sci 2015, 40:749–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Futaki S, Suzuki T, Ohashi W, Yagami T, Tanaka S, Ueda K, Sugiura Y: Arginine-rich Peptides: AN ABUNDANT SOURCE OF MEMBRANE-PERMEABLE PEPTIDES HAVING POTENTIAL AS CARRIERS FOR INTRACELLULAR PROTEIN DELIVERY*. J Biol Chem 2001, 276:5836–5840. [DOI] [PubMed] [Google Scholar]
  • 12.Appelbaum JS, LaRochelle JR, Smith BA, Balkin DM, Holub JM, Schepartz A: Arginine topology controls escape of minimally cationic proteins from early endosomes to the cytoplasm. Chem Biol 2012, 19:819–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Doak BC, Over B, Giordanetto F, Kihlberg J: Oral druggable space beyond the rule of 5: insights from drugs and clinical candidates. Chem Biol 2014, 21:1115–1142. [DOI] [PubMed] [Google Scholar]
  • 14.Hewitt WM, Leung SSF, Pye CR, Ponkey AR, Bednarek M, Jacobson MP, Lokey RS: Cell-permeable cyclic peptides from synthetic libraries inspired by natural products. J Am Chem Soc 2015, 137:715–721. [DOI] [PubMed] [Google Scholar]
  • 15.Pye CR, Hewitt WM, Schwochert J, Haddad TD, Townsend CE, Etienne L, Lao Y, Limberakis C, Furukawa A, Mathiowetz AM, et al. : Nonclassical Size Dependence of Permeation Defines Bounds for Passive Adsorption of Large Drug Molecules. J Med Chem 2017, 60:1665–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.White TR, Renzelman CM, Rand AC, Rezai T, McEwen CM, Gelev VM, Turner RA, Linington RG, Leung SSF, Kalgutkar AS, et al. : On-resin N-methylation of cyclic peptides for discovery of orally bioavailable scaffolds. Nat Chem Biol 2011, 7:810–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Furukawa A, Townsend CE, Schwochert J, Pye CR, Bednarek MA, Lokey RS: Passive Membrane Permeability in Cyclic Peptomer Scaffolds Is Robust to Extensive Variation in Side Chain Functionality and Backbone Geometry. J Med Chem 2016, 59:9503–9512. [DOI] [PubMed] [Google Scholar]
  • 18.Biron E, Chatterjee J, Ovadia O, Langenegger D, Brueggen J, Hoyer D, Schmid HA, Jelinek R, Gilon C, Hoffman A, et al. : Improving oral bioavailability of peptides by multiple N-methylation: somatostatin analogues. Angew Chem Int Ed Engl 2008, 47:2595–2599. [DOI] [PubMed] [Google Scholar]
  • 19.Wang CK, Northfield SE, Colless B, Chaousis S, Hamernig I, Lohman R-J, Nielsen DS, Schroeder CI, Liras S, Price DA, et al. : Rational design and synthesis of an orally bioavailable peptide guided by NMR amide temperature coefficients. Proc Natl Acad Sci U S A 2014, 111:17504–17509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ovadia O, Greenberg S, Chatterjee J, Laufer B, Opperer F, Kessler H, Gilon C, Hoffman A: The effect of multiple N-methylation on intestinal permeability of cyclic hexapeptides. Mol Pharm 2011, 8:479–487. [DOI] [PubMed] [Google Scholar]
  • 21.Bockus AT, Lexa KW, Pye CR, Kalgutkar AS, Gardner JW, Hund KCR, Hewitt WM, Schwochert JA, Glassey E, Price DA, et al. : Probing the Physicochemical Boundaries of Cell Permeability and Oral Bioavailability in Lipophilic Macrocycles Inspired by Natural Products. J Med Chem 2015, 58:4581–4589. [DOI] [PubMed] [Google Scholar]
  • 22.Wang CK, Northfield SE, Swedberg JE, Colless B, Chaousis S, Price DA, Liras S, Craik DJ: Exploring experimental and computational markers of cyclic peptides: Charting islands of permeability. Eur J Med Chem 2015, 97:202–213. [DOI] [PubMed] [Google Scholar]
  • 23.Doak BC, Zheng J, Dobritzsch D, Kihlberg J: How Beyond Rule of 5 Drugs and Clinical Candidates Bind to Their Targets. J Med Chem 2016, 59:2312–2327. [DOI] [PubMed] [Google Scholar]
  • 24.Lipinski CA, Lombardo F, Dominy BW, Feeney PJ: Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 1997, 23:3–25. [DOI] [PubMed] [Google Scholar]
  • 25.Furukawa A, Schwochert J, Pye CR, Asano D, Edmondson QD, Turmon AC, Klein VG, Ono S, Okada O, Lokey RS: Drug-Like Properties in Macrocycles above MW 1000: Backbone Rigidity versus Side-Chain Lipophilicity. Angew Chem Int Ed Engl 2020, 59:21571–21577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kelly CN, Townsend CE, Jain AN, Naylor MR, Pye CR, Schwochert J, Lokey RS: Geometrically Diverse Lariat Peptide Scaffolds Reveal an Untapped Chemical Space of High Membrane Permeability. J Am Chem Soc 2021, 143:705–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bhardwaj G, O’Connor J, Rettie S, Huang Y-H, Ramelot TA, Mulligan VK, Alpkilic GG, Palmer J, Bera AK, Bick MJ, et al. : Accurate de novo design of membrane-traversing macrocycles. Cell 2022, doi: 10.1016/j.cell.2022.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Naylor MR, Ly AM, Handford MJ, Ramos DP, Pye CR, Furukawa A, Klein VG, Noland RP, Edmondson Q, Turmon AC, et al. : Lipophilic Permeability Efficiency Reconciles the Opposing Roles of Lipophilicity in Membrane Permeability and Aqueous Solubility. J Med Chem 2018, 61:11169–11182. [DOI] [PubMed] [Google Scholar]
  • 29.Wang CK, Swedberg JE, Harvey PJ, Kaas Q, Craik DJ: Conformational Flexibility Is a Determinant of Permeability for Cyclosporin. J Phys Chem B 2018, 122:2261–2276. [DOI] [PubMed] [Google Scholar]
  • 30.Corbett KM, Ford L, Warren DB, Pouton CW, Chalmers DK: Cyclosporin Structure and Permeability: From A to Z and Beyond. J Med Chem 2021, 64:13131–13151. [DOI] [PubMed] [Google Scholar]
  • 31.Limbach MN, Antevska A, Oluwatoba DS, Gray ALH, Carroll XB, Hoffmann CM, Wang X, Voehler MW, Steren CA, Do TD: Atomic View of Aqueous Cyclosporine A: Unpacking a Decades-Old Mystery. J Am Chem Soc 2022, 144:12602–12607. [DOI] [PubMed] [Google Scholar]
  • 32.Leung SSF, Mijalkovic J, Borrelli K, Jacobson MP: Testing physical models of passive membrane permeation. J Chem Inf Model 2012, 52:1621–1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rezai T, Bock JE, Zhou MV, Kalyanaraman C, Scott Lokey R, Jacobson MP: Conformational Flexibility, Internal Hydrogen Bonding, and Passive Membrane Permeability: Successful in Silico Prediction of the Relative Permeabilities of Cyclic Peptides. Journal of the American Chemical Society 2006, 128:14073–14080. [DOI] [PubMed] [Google Scholar]
  • 34.Kamenik AS, Kraml J, Hofer F, Waibl F, Quoika PK, Kahler U, Schauperl M, Liedl KR: Macrocycle Cell Permeability Measured by Solvation Free Energies in Polar and Apolar Environments. J Chem Inf Model 2020, 60:3508–3517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Golosov AA, Flyer AN, Monovich LG: Design and Discovery of Orally Bioavailable Macrocycles: Toward Orally Bioavailable Peptide Therapeutics. In Approaching the Next Inflection in Peptide Therapeutics: Attaining Cell Permeability and Oral Bioavailability. American Chemical Society; 2022:199–222. [Google Scholar]
  • 36.Golosov AA, Flyer AN, Amin J, Babu C, Gampe C, Li J, Liu E, Nakajima K, Nettleton D, Patel TJ, et al. : Design of Thioether Cyclic Peptide Scaffolds with Passive Permeability and Oral Exposure. J Med Chem 2021, 64:2622–2633. [DOI] [PubMed] [Google Scholar]
  • 37.Miao J, Descoteaux ML, Lin Y-S: Structure prediction of cyclic peptides by molecular dynamics + machine learning. Chem Sci 2021, 12:14927–14936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hosseinzadeh P, Bhardwaj G, Mulligan VK, Shortridge MD, Craven TW, Pardo-Avila F, Rettie SA, Kim DE, Silva D-A, Ibrahim YM, et al. : Comprehensive computational design of ordered peptide macrocycles. Science 2017, 358:1461–1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sindhikara D, Johnston J: Roles of Conformations on Predictions of Peptide Properties. In Approaching the Next Inflection in Peptide Therapeutics: Attaining Cell Permeability and Oral Bioavailability. . American Chemical Society; 2022:103–135. [Google Scholar]
  • 40.Mulligan VK, Kang CS, Sawaya MR, Rettie S, Li X, Antselovich I, Craven TW, Watkins AM, Labonte JW, DiMaio F, et al. : Computational design of mixed chirality peptide macrocycles with internal symmetry. Protein Sci 2020, 29:2433–2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cataldi E, Raschig M, Gutmann M, Geppert PT, Ruopp M, Schock M, Gerwe H, Bertermann R, Meinel L, Finze M, et al. : Amber Light Control of Peptide Secondary Structure by a Perfluoroaromatic Azobenzene Photoswitch. Chembiochem 2022, [DOI] [PubMed] [Google Scholar]
  • 42.McHugh SM, Rogers JR, Yu H, Lin Y-S: Insights into How Cyclic Peptides Switch Conformations. J Chem Theory Comput 2016, 12:2480–2488. [DOI] [PubMed] [Google Scholar]
  • 43.Damjanovic J, Miao J, Huang H, Lin Y-S: Elucidating Solution Structures of Cyclic Peptides Using Molecular Dynamics Simulations. Chem Rev 2021, 121:2292–2324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Witek J, Keller BG, Blatter M, Meissner A, Wagner T, Riniker S: Kinetic Models of Cyclosporin A in Polar and Apolar Environments Reveal Multiple Congruent Conformational States. J Chem Inf Model 2016, 56:1547–1562. [DOI] [PubMed] [Google Scholar]
  • 45.Kamenik AS, Linker SM, Riniker S: Matching Simulations and Experiments of Conformationally Flexible Cyclic Peptides: Steps toward a Holistic View of Passive Membrane Permeability. In Approaching the Next Inflection in Peptide Therapeutics: Attaining Cell Permeability and Oral Bioavailability. . American Chemical Society; 2022:137–154. [Google Scholar]
  • 46.Sugita M, Sugiyama S, Fujie T, Yoshikawa Y, Yanagisawa K, Ohue M, Akiyama Y: Large-Scale Membrane Permeability Prediction of Cyclic Peptides Crossing a Lipid Bilayer Based on Enhanced Sampling Molecular Dynamics Simulations. J Chem Inf Model 2021, 61:3681–3695. [DOI] [PubMed] [Google Scholar]
  • 47.McHugh SM, Yu H, Slough DP, Lin Y-S: Mapping the sequence-structure relationships of simple cyclic hexapeptides. Phys Chem Chem Phys 2017, 19:3315–3324. [DOI] [PubMed] [Google Scholar]
  • 48.Slough DP, Yu H, McHugh SM, Lin Y-S: Toward accurately modeling N-methylated cyclic peptides. Phys Chem Chem Phys 2017, 19:5377–5388. [DOI] [PubMed] [Google Scholar]
  • 49.Ono S, Naylor MR, Townsend CE, Okumura C, Okada O, Lee H-W, Lokey RS: Cyclosporin A: Conformational Complexity and Chameleonicity. J Chem Inf Model 2021, 61:5601–5613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dickson CJ, Hornak V, Bednarczyk D, Duca JS: Using Membrane Partitioning Simulations To Predict Permeability of Forty-Nine Drug-Like Molecules. J Chem Inf Model 2019, 59:236–244. [DOI] [PubMed] [Google Scholar]
  • 51.Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, Tunyasuvunakool K, Bates R, Žídek A, Potapenko A, et al. : Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596:583–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Baek M, DiMaio F, Anishchenko I, Dauparas J, Ovchinnikov S, Lee GR, Wang J, Cong Q, Kinch LN, Schaeffer RD, et al. : Accurate prediction of protein structures and interactions using a three-track neural network. Science 2021, 373:871–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.McDonald EF, Jones T, Plate L, Meiler J, Gulsevin A: Benchmarking AlphaFold2 on peptide structure prediction. Structure 2023, 31:111–119.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Watson JL, Juergens D, Bennett NR, Trippe BL, Yim J, Eisenach HE, Ahern W, Borst AJ, Ragotte RJ, Milles LF, et al. : Broadly applicable and accurate protein design by integrating structure prediction networks and diffusion generative models. bioRxiv 2022, doi: 10.1101/2022.12.09.519842. [DOI] [Google Scholar]
  • 55.Yu P, Liu B, Kodadek T: A high-throughput assay for assessing the cell permeability of combinatorial libraries. Nat Biotechnol 2005, 23:746–751. [DOI] [PubMed] [Google Scholar]
  • 56.Bird GH, Mazzola E, Opoku-Nsiah K, Lammert MA, Godes M, Neuberg DS, Walensky LD: Biophysical determinants for cellular uptake of hydrocarbon-stapled peptide helices. Nat Chem Biol 2016, 12:845–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Holub JM, Larochelle JR, Appelbaum JS, Schepartz A: Improved assays for determining the cytosolic access of peptides, proteins, and their mimetics. Biochemistry 2013, 52:9036–9046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Qian Z, Martyna A, Hard RL, Wang J, Appiah-Kubi G, Coss C, Phelps MA, Rossman JS, Pei D: Discovery and Mechanism of Highly Efficient Cyclic Cell-Penetrating Peptides. Biochemistry 2016, 55:2601–2612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.LaRochelle JR, Cobb GB, Steinauer A, Rhoades E, Schepartz A: Fluorescence correlation spectroscopy reveals highly efficient cytosolic delivery of certain penta-arg proteins and stapled peptides. J Am Chem Soc 2015, 137:2536–2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kwon Y-U, Kodadek T: Quantitative comparison of the relative cell permeability of cyclic and linear peptides. Chem Biol 2007, 14:671–677. [DOI] [PubMed] [Google Scholar]
  • 61.Mientkiewicz KM, Peraro L, Kritzer JA: Parallel Screening Using the Chloroalkane Penetration Assay Reveals Structure-Penetration Relationships. ACS Chem Biol 2021, doi: 10.1021/acschembio.1c00434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Peier A, Ge L, Boyer N, Frost J, Duggal R, Biswas K, Edmondson S, Hermes JD, Yan L, Zimprich C, et al. : NanoClick: A High Throughput, Target-Agnostic Peptide Cell Permeability Assay. ACS Chem Biol 2021, 16:293–309. [DOI] [PubMed] [Google Scholar]
  • 63.Kansy M, Senner F, Gubernator K: Physicochemical high throughput screening: parallel artificial membrane permeation assay in the description of passive absorption processes. J Med Chem 1998, 41:1007–1010. [DOI] [PubMed] [Google Scholar]
  • 64.Hilgers AR, Conradi RA, Burton PS: Caco-2 cell monolayers as a model for drug transport across the intestinal mucosa. Pharm Res 1990, 7:902–910. [DOI] [PubMed] [Google Scholar]
  • 65.Dempsey CE: Hydrogen exchange in peptides and proteins using NMR spectroscopy. Prog Nucl Magn Reson Spectrosc 2001, 39:135–170. [Google Scholar]
  • 66.Adaligil E, Fairbrother WJ: NMR Spectroscopy for Studying Peptide Conformations and Cell Permeability. In Approaching the Next Inflection in Peptide Therapeutics: Attaining Cell Permeability and Oral Bioavailability. . American Chemical Society; 2022:155–177. [Google Scholar]
  • 67.Güntert P, Buchner L: Combined automated NOE assignment and structure calculation with CYANA. J Biomol NMR 2015, 62:453–471. [DOI] [PubMed] [Google Scholar]
  • 68.Brünger AT, Adams PD, Clore GM, DeLano WL, Gros P, Grosse-Kunstleve RW, Jiang JS, Kuszewski J, Nilges M, Pannu NS, et al. : Crystallography & NMR system: A new software suite for macromolecular structure determination. Acta Crystallogr D Biol Crystallogr 1998, 54:905–921. [DOI] [PubMed] [Google Scholar]
  • 69.Schwieters C, Kuszewski J, Mariusclore G: Using Xplor–NIH for NMR molecular structure determination. Prog Nucl Magn Reson Spectrosc 2006, 48:47–62. [Google Scholar]
  • 70.Jain AN, Cleves AE, Gao Q, Wang X, Liu Y, Sherer EC, Reibarkh MY: Complex macrocycle exploration: parallel, heuristic, and constraint-based conformer generation using ForceGen. J Comput Aided Mol Des 2019, 33:531–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cicero DO, Barbato G, Bazzo R: NMR analysis of molecular flexibility in solution: A new method for the study of complex distributions of rapidly exchanging conformations. Application to a 13-residue peptide with an 8-residue loop. J Am Chem Soc 1995, 117:1027–1033. [Google Scholar]
  • 72.Wieske LHE, Atilaw Y, Poongavanam V, Erdélyi M, Kihlberg J: Going Viral: An Investigation into the Chameleonic Behaviour of Antiviral Compounds. Chemistry 2022, [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Danelius E, Poongavanam V, Peintner S, Wieske LHE, Erdélyi M, Kihlberg J: Solution Conformations Explain the Chameleonic Behaviour of Macrocyclic Drugs. Chemistry 2020, 26:5231–5244. [DOI] [PubMed] [Google Scholar]
  • 74.Cleves AE, Jain AN: ForceGen 3D structure and conformer generation: from small lead-like molecules to macrocyclic drugs. J Comput Aided Mol Des 2017, 31:419–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Nguyen QNN, Schwochert J, Tantillo DJ, Scott Lokey R: Using 1 H and 13 C NMR chemical shifts to determine cyclic peptide conformations: a combined molecular dynamics and quantum mechanics approach. Phys Chem Chem Phys 2018, 20:14003–14012. [DOI] [PubMed] [Google Scholar]
  • 76.Kuschert S, Stroet M, Chin YK-Y, Conibear AC, Jia X, Lee T, Bartling CRO, Strømgaard K, Güntert P, Rosengren KJ, et al. : Facilitating the structural characterisation of non-canonical amino acids in biomolecular NMR. Magnetic Resonance Discussions 2022, doi: 10.5194/mr-2022-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hyung S-J, Feng X, Che Y, Stroh JG, Shapiro M: Detection of conformation types of cyclosporin retaining intramolecular hydrogen bonds by mass spectrometry. Anal Bioanal Chem 2014, 406:5785–5794. [DOI] [PubMed] [Google Scholar]
  • 78.Lam KHB, Le Blanc JCY, Campbell JL: Separating Isomers, Conformers, and Analogues of Cyclosporin using Differential Mobility Spectroscopy, Mass Spectrometry, and Hydrogen–Deuterium Exchange. Anal Chem 2020, 92:11053–11061. [DOI] [PubMed] [Google Scholar]
  • 79.James EI, Murphree TA, Vorauer C, Engen JR, Guttman M: Advances in Hydrogen/Deuterium Exchange Mass Spectrometry and the Pursuit of Challenging Biological Systems. Chem Rev 2022, 122:7562–7623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Goto Y, Suga H: The RaPID Platform for the Discovery of Pseudo-Natural Macrocyclic Peptides. Acc Chem Res 2021, 54:3604–3617. [DOI] [PubMed] [Google Scholar]

RESOURCES