You are on page 1of 16

European Journal of Medicinal Chemistry 223 (2021) 113635

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Modified histidine containing amphipathic ultrashort antifungal


peptide, His[2-p-(n-butyl)phenyl]-Trp-Arg-OMe exhibits potent
anticryptococcal activity
Krishna K. Sharma a, Ravikant Ravi a, Indresh Kumar Maurya b, Akshay Kapadia a,
Shabana I. Khan c, Vinod Kumar d, Kulbhushan Tikoo d, Rahul Jain a, *
a
Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
b
Department of Microbial Technology, Panjab University, Sector 25, Chandigarh 160 014, India
c
National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, Mississippi 38677, United States
d
Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar 160 062, Punjab India

a r t i c l e i n f o a b s t r a c t

Article history: In pursuit of ultrashort peptide-based antifungals, a new structural class, His(2-aryl)-Trp-Arg is reported.
Received 22 January 2021 Structural changes were investigated on His-Trp-Arg scaffold to demonstrate the impact of charge and
Received in revised form lipophilic character on the biological activity. The presence and size of the aryl moiety on imidazole of
31 May 2021
histidine modulated overall amphiphilic character, and biological activity. Peptides exhibited IC50 of 0.37
Accepted 6 June 2021
Available online 12 June 2021
e9.66 mg/mL against C. neoformans. Peptide 14f [His(2-p-(n-butyl)phenyl)-Trp-Arg-OMe] exhibited two-
fold potency (IC50 ¼ 0.37 mg/mL, MIC ¼ 0.63 mg/mL) related to amphotericin B, without any cytotoxic
effects up to 10 mg/mL. Peptide 14f act by nuclear fragmentation, membranes permeabilization,
Keywords:
Antifungal peptides (AFPs)
disruption and pore formations in the microbial cells as determined by the mechanistic studies
Cryptococcus neoformans employing Trp-quenching, CLSM, SEM, and HR-TEM. The amalgamation of short sequence, presence of
Membrane-disruption appropriate aryl group on L-histidine, potent anticryptococcal activity, no cytotoxicity, and detailed
Membrane selectivity mechanistic studies directed to the identification of 14f as a new antifungal structural lead.
Amphiphilicity © 2021 Elsevier Masson SAS. All rights reserved.
Cytotoxicity

1. Introduction unwanted drug-drug interactions, poor solubility, and importantly


drug resistance that makes management a difficult and challenging
Global incidences of fungal infections are increasing at an task [6e8]. A palliative solution to this conundrum is the discovery
alarming rate and pose an urgent necessity to discover new classes of and development of newer classes of antifungal agents with alter-
antifungals. Invasive fungal infections being potentially pathogenic, native mechanism of action [9,10].
takes a huge toll on human population mainly targeting immuno- Cationic antimicrobial peptides (CAMPs) bid encouraging pros-
compromised individuals suffering from AIDS, cancer and organ pects for the medicinal chemistry research of newer antifungal
transplant receivers [1e3]. Cryptococcosis is one of the most agents [11e13]. Optimal balance of cationic charge vs lipophilicity
worldwide distributed infection responsible for more than 600,000 ratio is considered to be important for a potent CAMP [14e16].
deaths per year and presents a substantial therapeutic challenge due Their mechanisms of action are still debatable, though, its activity is
to increasing resistance cases to clinically used antifungal medica- attributed to membrane infiltration and disruption of the pathogen
tions [1e5]. Clinical therapeutic interventions are mainly depend on cells [17]. In terms of selectivity, CAMPs being positively charged,
azoles (fluconazole), fluoropyrimidines (5-fluorocytosine), allyl- preferentially bind to negatively charged microbial membranes,
amines (terbinafine), echinocandins (caspofungin) and macrocyclic and composition of cell surface of susceptible cells also played
polyenes (amphotericin B). Clinically available treatments have importantly role. In case of fungal strain compared to bacteria, cell
shortcomings such as side effects, non-optimal pharmacokinetics, wall composition can also play important role for its susceptibility
[18,19]. Therefore, unique mechanism of action makes this class of
compound less likely to develop resistance. There has been an
* Corresponding author. evolution in designing short synthetic peptide-based derivatives
E-mail address: rahuljain@niper.ac.in (R. Jain). from the long sequence natural AMPs with a view to improve

https://doi.org/10.1016/j.ejmech.2021.113635
0223-5234/© 2021 Elsevier Masson SAS. All rights reserved.
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

proteolytic stability and enhanced bioactivity [20,21]. A compara- using tryptophan quenching, confocal laser scanning microscopy
tive sequence analysis of natural as well as synthetically derived (CLSM), and electron microscopic examinations clearly demon-
antimicrobial peptides show that cationic amino acids (Arg, His and strate the membrane-disruption and intracellular localization
Lys), and lipophilic amino acids (Trp, Phe, Ile and Val) have been mechanism of action of the peptide 14f.
repeatedly present in randomized manner [22,23]. This gives a
clear idea of the necessity of incorporating amino acid residues that 2. Results and discussion
optimally balance both the charge and lipophilicity in the designed
peptides [24e26]. 2.1. Chemistry and synthesis
In our quest to develop ultrashort peptide/mimetics-based
novel lead antimicrobial probes, we examined the prerequisite The synthetic strategy employed to prepare the designed tri-
features in the naturally occurring and synthetic peptides [27e32]. peptides has been summarized in Schemes 1-3. Initially, N-a-Boc-
Further, peptide scaffolds possessing þ2/þ3 sites and tuned with 2-aryl-L-histidines (4a-e) were synthesized in four steps (Scheme
lipophilic character imparts an improved antifungal and antibac- 1) using a previously reported protocol [40].
terial activity [27,29e32]. Naturally occurring cationic peptides The method employed for arylboronic acids mediated arylation
such as the histidine-rich histatins & clavanins, and tryptophan- of the imidazole moiety of the histidine, proceeds through a direct
rich indolicidin & tritrpticin scaffolds display potent antifungal regioselective homolytic mechanism, which involves the addition
activity, encouraging us to design ultrashort peptide sequence(s) by of an aryl radical to electron deficient position of a histidine ring
judiciously using histidine and tryptophan residues [33e38]. It is followed by re-aromatization, affording the arylated derivatives
also known that naturally occurring cationic peptides containing (2a-e) in 42e55% yields. Acidolysis by refluxing with methanolic
cationic amino acids such lysine and arginine at the end terminus HCl provided the deprotected 2-aryl-L-histidine dihydrochloride
display activity [14e16]. So, we decided to use histidine, tryptophan salts (3a-e) in 92e96% yields. N-a-Boc protection using di-tert-
and arginine amino acids in the designed sequence by placing butyl dicarbonate in basic reaction medium provided by 4 N NaOH,
lipophilic tryptophan centrally and arginine at the C-terminus. furnished the desired N-a-Boc-2-aryl-L-histidine scaffolds (4a-e) in
Histidine was placed at the N-terminus to tune amphiphilicity of 91e95% yields for incorporation in the designed tripeptide skel-
the designed sequence by using imidazole ring derivatization. eton. Next, to synthesize the starting material for Series 1, L-Arg-OH
Structural modifications on the aromatic ring of histidine was also was reacted with methanolic HCl to afford methyl ester derivative
envisioned to generate amphiphilicity and stability in a sterically (6). Similarly for Series 2, L-Arg-OH after reacting with benzylamine
complex functional architecture [31,39]. Recently, ring-modified using 1,10 -carbonyldiimidazole (CDI) in H2O afforded L-Arg-NHBzl
histidine scaffolds proposed themselves as medicinally important (7) [45]. This reaction is stereo conservative and the side chain
the building blocks for peptide/mimetic-based design and discov- functional groups remained unaffected under the employed re-
ery [40e43]. It was envisioned that placement of C-2 substituted agents and reaction conditions (Scheme 2).
aromatic moieties on the imidazole ring of histidine would The synthesis of the tripeptides was accomplished in two steps.
generate lateral amphiphilicity in the membrane-active peptide Initially, Boc-L-Trp-OH was coupled with L-Arg-OMe/NHBzl (6 or 7)
sequence, which could possibly penetrate deeper into the lipid using a racemization free and microwave-mediated solution phase
chains of the fungal cell membranes. On the other hand, tryptophan peptide synthesis protocol [46,47], by employing DIC and HOBt as
residue provides a lipophilic bulk and cell membrane interaction coupling combination in anhydrous DMF to provided Boc protected
properties to the His-Trp-Arg sequence [44]. Next, C-terminus dipeptides derivatives (8 and 9).
modification of arginine was carried out to mask the anionic These dipeptides were further subjected to acidolysis using 6 N
character of the carboxyl group with methyl ester (less lipophilic) methanolic HCl to furnish Boc-deprotected L-Trp-L-Arg-OMe/
and benzylamide (higher lipophilicity) groups to modulate the NHBzl derivatives (10 and 11). The coupling of respective arylated L-
lipophilicity of the designed peptide scaffolds. histidine scaffolds (4a-f) were carried out with the deprotected
We designed His(aryl)-Trp-Arg-OMe/NHBzl scaffolds by dipeptides using solution phase peptide synthesis to afford the
reasoning that arginine methyl ester (cationicity with masked respective Boc-L-His(2-aryl)-L-Trp-L-Arg-OMe/NHBzl derivatives
anionic character), arginine benzylamide (cationicity with masked (12a-f and 13a-f). Acidolysis using 6 N methanolic HCl furnished
anionic character and lipophilicity) and tryptophan (lipophilicity) desired Boc-deprotected L-His(2-aryl)-L-Trp-L-Arg-OMe/NHBzl
along with ring-arylated histidine derivatives (to tune amphiphi- peptides (14a-f and 15a-f) (Scheme 3).
licity) would create a milieu of balanced charge-to-bulk ratio in
quest of potential ultrashort antimicrobials (Fig. 1). Position of the 2.2. In vitro antimicrobial activity
amino acids in the designed peptide scaffolds was assigned using
the nature of amino acids and to tune the characteristics of In the synthesized tripeptides, three structural manipulations
membrane-active antimicrobial peptides. To systematically allocate on the lead skeleton His-Trp-Arg were performed to demonstrate
the charge and bulk distribution, the lipophilic tryptophan was effect of charge and lipophilic character imparted through synthetic
centrally placed with cationic modified arginine at the C-terminus derivatization on the biological activity of the peptides. C2 position
and amphiphilic ring-modified histidine at the N-terminus (Fig. 1). of imidazole ring was substituted by phenyl, p-tolyl, p-ethylphenyl,
Additionally, the use of terminally placed modified arginine and biphenyl and p-(n-butyl)phenyl to investigate the effect of
histidine provide hindrance towards the peptidases to develop increasing lipophilicity with constant positive charge residues.
shorter bioactive His(aryl)-Trp-Arg-OMe/NHBzl peptides. Here, we Next, to evaluate the effect of bulkiness at the C-terminus, two
report the design, synthesis, biological and cytotoxicity evaluation different series of tripeptides with C-terminus methyl ester and
of ring-modified histidine, tryptophan and arginine containing benzylamide groups were synthesized. To get more insight of the
His(aryl)-Trp-Arg class that exhibit potent antifungal activities structure activity relationship (SAR) study, it was also planned to
against C. neoformans and moderate antibacterial activities against biologically investigate Boc moiety containing peptides 12a-f and
a type of staph bacteria, methicillin-resistant Staphylococcus aureus 13a-f to observe the effect of presence of aliphatic bulk and
(MSRA),and vancomycin-resistant Enterococci (VRE). The initial decreased cationicity at the N-terminus along with final peptides
biological activity assessment resulted in the identification of a lead 14a-f and 15a-f. Synthesized peptides (12a-f, 13a-f, 14a-f &15a-f)
peptide 14f. Detailed mechanistic investigations were performed were tested for in vitro antimicrobial activity against various fungal
2
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Fig. 1. Generalized scaffolds of the designed and synthesized peptides.

Scheme 1. Reagents and conditions: (i) 1 (1 equiv.), AreB(OH)2 (2 equiv.), (NH4)2S2O8 (2 equiv.), AgNO3 (0.2 equiv.), TFA (1.5 equiv.), CH2Cl2/H2O, rt, 12e18 h; (ii) 6 N HCl, 100  C,
12 h; (iii) (Boc)2O (3 equiv.), 4 N NaOH, 1,4-dioxane/H2O (3; 2), 24 h.

Scheme 2. Reaction and conditions: (i) HCl gas in MeOH, rt, 2 h; (ii) 5 (1 equiv.), benzylamine (1.5 equiv.), CDI (1.5 equiv.), H2O, rt, 12 h.

strains such as C. albicans, A. fumigatus, C. neoformans, and bacterial comparison to bacterial strains (Table 2). The results depict that the
strains such as MRSA, E. coli, K. pneumonia, P. aeruginosa, and VRE variation in the substituents at the aryl ring of the terminally placed
up to 20 mg/mL, and beyond that peptides were considered as L-histidine, resulting in varied lipophilicity greatly influenced the
inactive (NA) [48,49]. The synthesized peptides exhibited prom- potency. In series 1, Boc-protected peptide 12a without any sub-
ising activity against C. neoformans and activity values have been stitution on histidine residue was found inactive while peptides
presented in Table 1, whereas a few peptides showed moderate 12b-f containing ring-arylated histidines displayed IC50 values in
activity against MRSA and VRE and results are summarized in the range of 1.99 mg/mL to 9.66 mg/mL. Peptide 12f also exhibited
Table 2. Synthesized peptides were inactive against C. albicans, A. MIC value of 2.50 mg/mL against C. neoformans.
fumigatus, and K. pneumonia. At the same time, Boc-deprotected peptides 14a-f exhibited
From the bioactivity results, it could be inferred that the tested high antifungal activity as compared to its Boc-protected pre-
peptides showed preferential biological action against yeast, cursors 12a-f. Peptide 14a with additional charge (þ3) at the N-
C. neoformans of IC50 values of 0.37e9.66 mg/mL (Table 1) in terminus due to Boc-deprotection of 12a exhibited IC50 and MIC

3
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Scheme 3. Reagents and conditions: (i) 6 or 7 (1 equiv.), Boc-L-Trp-OH (1.5 equiv.), DIC (1.5 equiv.), DIEA (4 equiv.), HOBt (1.5 equiv.), DMF, 60  C, MW, 30 min; (ii) 6 N HCl in MeOH,
rt, 20 min; (iii) 10 or 11 (1 equiv.), Boc-L-His(2-aryl)-OH (1.5 equiv.), DIC (1.5 equiv.), DIEA (4 equiv.), HOBt (1.5 equiv.), DMF, 60  C, MW, 30 min.

values of 2.41 mg/mL and 10 mg/mL, respectively. Peptide 14f con- combination of OMe and Boc group of series 1 peptides, possibly
taining a bulkier p-(n-butyl)phenyl aliphatic-aromatic pendant due to better tuning of charge versus lipophilicity. In peptides 14a-f,
attached to histidine ring exhibited the most potent and promising without Boc moiety and an increase in lipophilic character modu-
activity with an IC50 value of 0.37 mg/mL and MIC of 0.63 mg/mL. lated by the aryl moiety of histidine lead to higher antifungal ac-
Peptide 14f was twice as potent related to the clinical agent, tivity compared to the Boc group containing peptides 12a-f,
amphotericin B (IC50 and MIC of 0.69 and 1.25 mg/mL, respectively). possibly due to better tuning of amphiphilicity and results in the
Peptides 14d (p-ethylphenyl) and 14e (biphenyl) also showed high identification of the most promising peptide 14f. In the nutshell,
antifungal activities with IC50 values of 1.85 and 1.51 mg/mL, peptides 12f, 13e, 13f, 14e, 14f, 15e and 15f produced higher activity
respectively and identical MIC value of 2.50 mg/mL. Hence, a and selectivity against C. neoformans.
obvious pattern demonstrating the role of lipophilicity and cationic These SAR results as depicted in Fig. 2 steered this study to infer
charge sites was observed in the antifungal activity for peptides that the bulky substitution at the C-2 position of histidine residue
12a-f and 14a-f having Boc/H and methylester at the N- and C- has a more obvious role in modulating the amphiphilicity and the
terminus, respectively. potency of the peptides.
In series 2, peptide 13a (R1 ¼ H, R2 ¼ NHBzl) with Boc-group The tested peptides also produced moderate activity against
intact was inactive while other Boc-protected peptides 13b-f MRSA (IC50 values ranged in 4.44e17.5 mg/mL and MIC values be-
showed a linear trend of increase in activity with lipophilicity tween 5 and 20 mg/mL) and VRE (IC50 values between 7.38 and
provided by ring-modification of histidine. The peptide 13b-f 15.4 mg/mL and MIC values ranged in 10e20 mg/mL). Specifically,
exhibited IC50 values ranged between 1.5 mg/mL to 7.93 mg/mL with peptides 13e and 13f showed IC50 values of 4.55 and 9.13 mg/mL,
MIC values ranged in 2.5 mg/mL to 5 mg/mL. Peptide 13d (p-ethyl- respectively, with identical MIC value of 10 mg/mL against MRSA.
phenyl) emerged as most potent amongst 13a-f with IC50 and MIC The same peptides also displayed IC50 values of 8.82 and 7.38 mg/
values of 1.5 and 2.5 mg/mL, respectively. In contrast, all Boc- mL, respectively and identical MIC value of 10 mg/mL for VRE
deprotected peptides 15a-f showed activity with IC50 values (Table 2). Furthermore, peptide 15e displayed IC50 and MIC value of
ranged between 1.17 mg/mL to 4.92 mg/mL and MIC values between 4.44 and 5 mg/mL against MRSA, while producing noticeable IC50
5 mg/mL to 10 mg/mL. Herein, peptides with bulkier aryl moiety 15d- and MICs of 8.33 and 20 mg/mL, respectively against VRE. Some
f [p-ethylphenyl, biphenyl and p-(n-butyl)phenyl] exhibited less peptides also indicated low activity against E. coli and P. aeruginosa.
potency at MIC values compared to their Boc-protected derivatives Aromatic lipophilic bulky groups such as ethylphenyl, biphenyl and
(13d-f). Activity results suggests that both NHBzl and Boc moiety butylphenyl at the imidazole ring of histidine also imparted anti-
exhibited more potent antifungal activity as compared to the bacterial activity against MRSA and VRE. (Table 2). The results of

4
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Table 1
In vitro anticryptococcal activities of synthetic peptides against C. neoformans.

Entry R1 R2 R3 C. neoformans CTXa (mg/mL)

IC50 MIC

12a H Boc OMe >20 e >10


12b Phenyl Boc OMe 8.85 >20 >10
12c p-Tolyl Boc OMe 7.66 >20 >10
12d p-Ethylphenyl Boc OMe 9.66 >20 >10
12e Biphenyl Boc OMe 8.06 >20 >10
12f p-(n-Butyl)phenyl Boc OMe 1.99 2.5 >10
13a H Boc NHBzl >20 e >10
13b Phenyl Boc NHBzl 7.93 >20 >10
13c p-Tolyl Boc NHBzl 4.34 5 >10
13d p-Ethylphenyl Boc NHBzl 1.5 2.5 >10
13e Biphenyl Boc NHBzl 2.15 2.5 >10
13f p-(n-Butyl)phenyl Boc NHBzl 1.98 2.5 >10
14a H H OMe 2.41 10 >10
14b Phenyl H OMe 9.23 >20 >10
14c p-Tolyl H OMe 8.98 >20 >10
14d p-Ethylphenyl H OMe 1.85 2.5 >10
14e Biphenyl H OMe 1.51 2.5 >10
14f p-(n-Butyl)phenyl H OMe 0.37 0.63 >10
15a H H NHBzl 4.92 10 >10
15b Phenyl H NHBzl 5.31 10 >10
15c p-Tolyl H NHBzl 3.36 5 >10
15d p-Ethylphenyl H NHBzl 2.26 10 >10
15e Biphenyl H NHBzl 1.17 5 >10
15f p-(n-Butyl)phenyl H NHBzl 2.27 5 >10
Amphotericin B 0.69 1.25

IC50 and MIC values in mg/mL.


a
Highest tested concentration of 10 mg/mL.

antibacterial activities provides a starting designed structural with methyl ester (less bulky) and hydrophobic benzylamide (more
template for further fine tuning in quest of new scaffolds against bulky) motifs at the C-termini exhibited a linear increases in ac-
pathogenic and challenging VRE and MRSA strains [50,51]. tivity with increasing aromatic bulk. In general, increase in lipo-
philic bulk [p-(n-butyl)phenyl > biphenyl > p-ethylphenyl > p-
2.3. Cytotoxicity assay tolyl > phenyl > H] results in incremental increase in bioactivity
against C. neoformans. The results of this study further reinforces
The cytotoxicity is one of the most important parameters for the the essentiality of optimal charge-to-bulk ratio (ideal balance of
determination of the potential of membrane disruptive antifungal amphiphilicity) for promising antifungal activity.
peptides as both fungal cells and humans are eukaryotes. Peptides
(series 1e2) were evaluated for in vitro cytotoxicity against
noncancerous mammalian cell line, VERO using the neutral red Table 2
uptake assay [52]. The results as summarizied in Table 1 confirms In vitro anti-bacterial activities of peptides.
that all peptides were exhibited no toxicity up to 10.0 mg/mL. These
Entry MSRA VRE E. coli P. aeruginosa
results indicate that antifungal activity is exhibited by peptide-
mediated effects and not due to the cell cytotoxicity. IC50 MIC IC50 MIC IC50 MIC IC50 MIC

12e 16.3 >20 e e >20 e >20 e


2.4. Molecular properties and activity correlation analysis 12f 9.29 10 7.82 20 17.88 >20 >20 e
13c 17.5 20 >20 e >20 e >20 e
13d 8.93 20 14.9 >20 >20 e >20 e
For a better understanding at the molecular level and to eval- 13e 4.56 10 8.82 10 >20 e 19.78 e
uate the effect of lipophilic changes on the antifungal action of the 13f 9.13 10 7.38 10 >20 e >20 e
peptides, and a relationship was formed between the retention 14d >20 e >20 e 17.36 >20 >20 e
14e 7.81 10 13.2 20 >20 e >20 e
times (tR), measured by analytical C-18 RP-HPLC and predicted
14f 8.74 10 15.4 20 15.57 >20 >20 e
ClogP values using ACD software [53]. 15d 18.4 20 13.5 20 >20 e >20 e
The study was conducted on peptides 14a-f and 15a-f. The 15e 4.44 5 8.33 20 >20 e 18.7 >20
correlation of activity (IC50 values against C. neoformans) and lip- 15f 4.46 10 9.99 20 16.26 >20 17.22 >20
ophilicity (tR and ClogP) of the tested peptides are summarized in Cpx 0.09 0.25 0.15 0.63 0.007 0.031 0.09 0.50

Table 3. The results reveal that peptides 14a-f and 15a-f substituted Cpx: Ciprofloxacin, IC50 and MIC values in mg/mL.

5
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Fig. 2. Structure activity relationship study of the synthesized peptides.

Table 3 membranes (EYPC/cholesterol) and Stern-Volmer plots were


Correlation between molecular properties and anticryptococcal activity of peptides. generated (Fig. 3) [55]. The fluorescence of the indole ring of
Entry CLogPa tR (min) C. neoformans IC50 (mg/mL) tryptophan decreased due to quenching in a concentration reliant
approach in the absence and presence of liposomes, using acryl-
14a 2.19 15.013 2.41
14b 0.10 17.892 9.24
amide to the peptide solution, and without any other effects. The
14c 0.39 18.652 8.98 values for the Stern-Volmer quenching constant (Ksv) in the
14d 0.92 19.982 1.85 absence of liposomes was 153.40 for 14f. For bioactive peptide,
14e 1.78 21.909 1.51 Stern-Volmer quenching constant was decreased in the presence of
14f 1.98 22.383 0.37
EYPC/EYPG (7:3, w/w Ksv ¼ 6.68 for 14f) and remained almost
15a 1.17 20.202 4.92
15b 0.92 20.749 5.31 similar in the presence of EYPC/cholesterol (10:1, w/w Ksv ¼ 145.69
15c 1.42 20.955 3.36 for 14f). These results suggest that the tryptophan fluorescent
15d 1.95 22.837 2.26 indole ring was submerged in the bilayers, and therefore became
15e 2.81 23.438 1.17
unavailable for quenching. These results illustrate that peptide af-
15f 3.00 24.533 2.27
finity for liposomes was EYPC/EYPG > EYPC/cholesterol, and clearly
NA: not active. demonstrated that the peptides gets effectively implanted in EYPC/
b
Retention times (tR) are mentioned using a C-18 column.
a EYPG than EYPC/cholesterol, and that indicating higher selectivity
ClogP values using ACD/LogP software.
ratio of the peptides 12f, 13d, 14e, 14f and 15e towards mimics of
bacterial cell membranes in comparison to mammalian cells.
By using in vitro activity, cytotoxicity results, molecular prop-
erties and activity correlation analysis, we chose peptide 14f for 2.6. Peptide architecture study
thorough evaluation, including membrane selectivity study, pep-
tide architecture study, and mechanism of action investigations. Bioactivity of the amphiphilic antimicrobial peptides sometimes
depends upon the mixed interplay of hydrophobic and hydrophilic
2.5. Tryptophan quenching study interactions between the bioactive peptide and the membranes
[56]. Recently, it has been discovered that intermolecular in-
The correlation between antimicrobial activity and cytotoxicity teractions among the peptides themselves play an important role in
is vested on host versus microbe membrane selectivity. If peptides determining their activity. Hydrophobic interactions involving pp
show activity by membrane-targeting then membrane selectivity is stacking and hydrophilic interactions between the cationic groups
of utmost importance [54]. The cationic property of peptides present within the peptide skeleton are responsible for molecular
potentially contributes to cell selectivity because the surface of self-assembling, supramolecular aggregation and gel-formation
pathogenic membranes is more negatively charged in comparison properties [57].
to that of mammalian cells. The amphipathic character of a peptide
is the main component driving the interaction with microbial cell 2.6.1. Dynamic light scattering study
membranes compared to that of the mammalian membranes. We To investigate the nature of intermolecular interactions of 14f,
noted antibacterial activity is some of the compounds (Table 2). we decided to study the particle size and dispersion of the peptide
Upon the basis of activity profile of peptides, we selected five most in solution state using Zetasizer by performing dynamic light
promising antibacterial peptides 12f, 13d, 14e, 14f and 15e for scattering experiments (Fig. 4) [58e60]. For comparative purposes,
tryptophan fluorescence quenching study and to elucidate their we also conducted the same experiment on the inactive peptide
interactions with synthetic mimics of membrane. The interactions 12a.
of the selected peptides towards pathogen was evaluated to A time dependent particle size analysis of peptides 14f and 12a
differentiate between the interaction with mammalian mem- was carried out using dynamic light scattering experiments. Hy-
branes, and the outer layer of bacterial surface. drodynamic radius of 90% peptide population D(90) in PBS was
Quenching of the fluorescence of tryptophan by acrylamide was considered, and the results revealed that the particle size of 14f
studied using synthetic small unilamellar vesicles (SUVs) i.e. increases slightly from 2073 nm to 2792 nm whereas the particle
negatively charged (EYPC/EYPG) and the zwitterionic model size of 12a decreased from 802 nm to 108 nm on incubation from
6
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

tryptophan residue, which were evaluated by monitoring the


fluorescence emission spectra (Fig. 5) [58e60]. Upon investigation,
initially, peptides 14f and 12a displayed a prominent emission peak
around 335 nm representing the tryptophan residue on excitation
at 280 nm.
After 24 h, 14f displayed increase in emission in fluorescence,
which is consistent with extended p-stacked aggregate formation
of tryptophan residue possibly due to the presence of ring-arylation
at the histidine residue resulting in additional p-stacked aggrega-
tion. In the case of 12a, the increase in fluorescence was less
compared to 14f due to the absence of ring-arylation.

2.6.3. Scanning electron microscopy (SEM) study


To have a microscopic insight of the peptide assembly, we
investigated the morphology of the active (14f) and control peptide
(12a). SEM study revealed that the peptide 14f upon incubation for
24 h formed unorganized aggregates (Fig. 6A and B) in comparison
to that of 12a (Fig. 6C), which was quite homogenously dispersed in
the phosphate saline buffer (PBS) solution. The SEM results and
fluorescence emission sepctroscopic study indicate that peptide 14f
forms random aggregates.
The peptide architecture studies indiactes that the synthetic
amphiphilic peptide 14f exhibits self-aggregation properties at the
pathogen surface. For better understanding, we designed other
microscopic experiments to further investigate mechanism of ac-
tion studies.

2.7. Mechanism of action study

We investigated the thorough mechanism of action by


employing various microscopic approaches. We anticipate that
determination of mechanism of action of 14f could guide in further
design of potent analogues with defined mode of action in
combating microbes. The CLSM examination was conducted to
understand the various actions of 14f on the fungal permeabiliza-
tion, intracellular localization and binding to genetic materials in
the fungal cells. The SEM study was carried out to study the effect of
active peptide on the cellular morphological changes, and the high-
resolution transmission electron microscopy (HR-TEM) study hel-
ped to observed the ultrastrucural actions of active peptide 14f on
cell surface of fungi.

2.7.1. Confocal laser scanning microscopic study


Red fluorescent propidium iodide (PI) is not permeable to intact
cell membrane, and DNA binding dye. If interacting peptide per-
meabilizes cell wall and membrane, causing in compromised fungal
membrane permeability thereby permitting PI to enter inside the
dead cells and provide red fluorescence. Under confocal micro-
Fig. 3. Stern-Volmer plots for the Trp fluorescence quenching. A) In the absence of
scopic visualization, the uptake of PI after treatment by fungal cells
vesicles, B) Plots in the presence of EYPC/EYPG (7:3 w/w), and C) In the presence of
EYPC/cholesterol (10:1 w/w). with 14f at MIC (1 h) showed red fluorescence due to per-
meabilization of cell surface (Fig. 4B), although the untreated cells
did not possess red fluorescence due to undamaged cell surface
0 to 4 h (Fig. 5). The polydispersity index (Ð) for 14f sparingly in- (Fig. 4A).
creases from 0.333 to 0.584, whereas for 12a it increases from 0.383 These results indicate that PI entered intracellularly within the
to 0.922. The experiments indicate higher dispersion, homogeneity cells after treatment with 14f as treated cells were unable to
and uniformity within the solution for 12a in comparison to 14f. regulate the chemical movement from its cell surface.
However, inactive peptide 12a is better distributed within the so- To study intracellular localization of 14f in C. neoformans, we
lution. To further understand peptide architecture, fluorescence prepared FITC-labeled peptides (fluorescein isothiocyanate; Ex
spectroscopic and electron microscopy techniques were employed. 480 nm) 14f and 12a. The confocal investigation of FITC-labeled 12a
revealed that it can not cross the cell surface due to the intact cell
2.6.2. Fluorescence spectroscopic study surface as confirmed by the absence of green fluorescence (Fig. 7C).
Tryptophan fluorescence emission study is a useful method for FITC-labeled 14f permeated from cell membrane barriers via per-
investigating the interaction of the synthetic peptides and their meabilization and disruption after treatment with fungal cells, and
assemblies [29]. One plausible interaction within the molecule able to enter inside fungal cells by exhibiting the green fluorescence
could be due to the formation of p-stacked aggregates of the (Fig. 7D). These results confirm the ability of 14f to cross membrane
7
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Fig. 4. Dynamic light scattering studies (A) time dependent particle size distribution showing D(90) of the peptides 14f and 12a in PBS at pH 7.4, and (B) polydispersity index (Ð)
analysis.

Fig. 5. Fluorescence emission spectra of 10 mM samples of 14f and 12a, respectively at 24 h overlaid with emission at 0 h.

Fig. 6. SEM micrographs showing aggregated state of 14f in comparison to homogenously dispersed 12a after 24 h incubation. Scale of image A (10.4 mm x 7.50 k, 5 mm), image B
(10.4 mm x 8.50 k, 5 mm), image C (10.6 mm x 7.50 k, 5 mm).

barriers and sebsequent interaction with the intracellular targets 2.7.2. Electron microscopic study
resulting in the death of cells. The detailed mechanistic investigation of the potent peptide 14f
Blue fluorescnent DAPI (40 ,6-diamidino-2-phenylindole) re- was investigated by SEM and HR-TEM with a view to gain addi-
leases fluorescence after selective binding to the AT regions of DNA tional understandings about the mechanism of action. SEM exam-
while entering in live cells. The DAPI dye is utilized for nuclear or ination was carried out to observe changes in the cryptococcal cell
chromatin fragmentation of DNA, and alterations in fluorescence surface morphology after treatment with 14f at its MIC. The un-
intensity indicate the magnitude of DNA damage triggered by the treated cells have smooth outer cell surface when compared with
active peptides. Treated fungal cells upon DAPI staining and treat- treated cells with 14f (Fig. 8A). Peptide 14f caused prominent cell
ment with 14f showed the split and less blue fluorescence intensity wall breakage or cell surface damage, and permeabilization and
in the cryptococcal cells compared with untreated cells (Fig. 7E and hole(s) formation was evidently noticeable with the release of
F). From these results, killing of fungal cells by 14f is also contrib- intracellular material outside cells (Fig. 8BeD). Possibly, a cationic
uted to its interaction with genetic material such as DNA after amphiphilic peptide first interact with the negatively charged
membrane permeabilization causing the nuclear fragmentation. fungal cells by electronic interactions and gets aggregated over

8
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Fig. 7. Confocal fluorescence microscopic imagesto examine the impact of 14f at MIC: (A and B) membrane permeabilization examination using PI after 1 h; (C and D) FITC-labeled
14f after 1 h and FITC-labeled control peptide; (E and F) peptide treatment using DAPI after 1 h. Images in (A) and (E) are fungal cells (ATCC350) incubated alone, and image in (C)
FITC-labeled control. Images in (B), (D), and (F) correspond to active peptide treatment after 1 h of incubation.

fungal surface in comparison to zwitterionic mammalian mem-


branes, thereby exhibiting preferential selectivity to fungal strains.
Further, amphiphilic peptide form pores over cell surface, resulting
in hole formation thereby increasing permiabilization. It is inter-
pretatedthat 14f treated fungal cells can not withstand the various
effects exerted by peptides, leading to extensive disruption of cell
surface chracterstics, and permeability through pore(s) formation.
In the HR-TEM examination of cells, 14f treatment (at MIC
concentration) resulted in disruptive effects on capsule, cell wall
and the plasma membrane.
Untreated fungal cells exhibit undamaged membranes including
capsule under the HR-TEM investigation. Fig. 9A showed intact cell
morphology of a group of fungal cells, while Fig. 9B and C centered
on a single cell imagining and exhibited undamaged cell surface
including capsule, cell wall and plasma membrane under untreated
conditions. Fig. 9DeF exhibits the rainbow images of the untreated
fungal cells. The peptide 14f treated fungal cells demonstrate se-
vere disruption of capsule (Fig. 9F and L), cell wall [(Fig. 9C and F
(intact) and Fig. 9I and L (disrupted)] and plasma membrane
resulting in membrane permeabilization leading to fungal cell lysis
Fig. 8. Morphological examination of cells with 14f and untreated cells using SEM. (Fig. 9GeI). This examination clearly reveals that 14f peptide act by
Images (A and B) represent untreated cells (10000  , 10 mm; 7500  , 10.1 mm, 5 mm). interacting with cell surface as cryptococcus capsule was disrupted
Images (C and D) represent treated cells treated with 14f (7500  , 10.1 mm; 7500  , followed by other membranes (please see marked arrows, Fig. 9I
10.5 mm, 5 mm).

9
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

Fig. 9. Morphological examination of cells treated with 14f and untreated using HR-TEM. Images (AeF) represent untreated cells; rainbow images D-F generated from A-C. Images
(GeL) represent cells treated with 14f; rainbow images J-L generated from G-I. The scale bar of images A and G is 0.5 mm, images B and H is 0.2 mm and for other images it is 100 nm.

10
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

and L). It is known that Cryptococcal cells produce a characteristic patterns when suitable: br ¼ broad, s ¼ singlet, d ¼ doublet,
polysaccharide capsule that provide protection against environ- dd ¼ doublet of doublets, t ¼ triplet, q ¼ quadruplet, m ¼ multiplet.
mental stress and predators. Capsule provides protection inside Coupling constants (J) were reported in hertz unit (Hz). The final
host through reducing host immune responses by down regulating peptides were purified through Shimadzu preparative HPLC using a
inflammatory cytokines and inhibiting monocytes [18,19]. There- Luna 10 mM C-18 (250 mm  21.2 mm, 10 mm) column. The purity of
fore, alteration in capsule can makes C. neoformans cells susceptible peptides was assessed on a Shimadzu SPD-M20A analytical HPLC
for killing through various means, including drugs. The rainbow using a supelcosil™ LC-18 (25 cm  4.6 mm ID, 5 mm), and final
images were generated from corresponding images to enhance purity assessed, using a gradient run of 955% (1 mL/min; A, 0.1%
examination clarity (Fig. 9J-L). TFA in H2O/B, 0.1% TFA in CH3CN), and detection using PDA detector
The mechanism of action studies by using CLSM, SEM, and HR- at 220 or 254 nm; all biologically evaluated peptides exhibited
TEM indicate that 14f is a surface or membrane active peptide and 95% purity. The high-resolution mass of the synthesized peptides
act by infiltrating the fungal cellular membranes, thus killing the were analyzed on a MAXIS Bruker spectrometer using ESI-TOF
fungal pathogen. Selectivity of this class of peptides towards method.
C. neoformans cells might be due to disruption of specialized
capsule and cell wall composition. Additionally, the positively 4.1.1. General synthesis of N-a-Boc-2-aryl-L-histidines (4a-e)
charged bioactive peptide interacts with the negatively charged In a flask, starting material (1, 1.0 equiv.) was subjected in CH2Cl2
fungal membrane by electrostatic interactions in comparison to (5 mL), and CF3CO2H (1.5 equiv.) was added and stirred for 10 min.
zwitterionic mammalian membranes and exhibit preferential Thereafter, AgNO3 (0.2 equiv.) dissolved in H2O (5 mL) was added in
selectivity to cryptococcal strain. flask and followed by addition of arylboronic acid (1.5 equiv.). Next,
(NH4)2S2O8 (2.0 equiv.) was added at ambient conditions for
3. Conclusions 12e18 h and reaction monitored through TLC. After completion, the
pH of the reaction was adjusted (pH ¼ 11e12) by the NH4OH, and
In conclusion, design and synthesis of new scaffold of His(2-Ar)- CH2Cl2 was evaporated under vacuum, and extraction of the reac-
Trp-Arg of the amphipathic peptides composed of C-2 arylated tion mixture with EtOAc. The organic layer was washed with brine
histidine along with cationic arginine and hydrophobic tryptophan and dried using Na2SO4and crude mixture purified by flash chro-
is reported. These peptides have demonstrated high potency matography to isolate N-a-trifluoroacetyl-2-aryl-L-histidine methyl
against C. neoformans fungal strain, take the lead to identification of esters (2a-f) in 42e55% yields [40]. 2-Aryl-L-histidine.2HCl salts
bioactive peptide 14f as the most potent, which is two-fold more (3a-f) were produced by refluxing a solution of 2a-f in 6 N HCl for
potent than amphotericin B. Some other peptides (13e, 13f, 15e and 12 h in 92e96% yields. Derivatives of 2-Aryl-L-histidine salts (3a-f, 1
15f) of this class also provide a starting template for desiging new equiv.) were suspended in water-dioxane (1:2) and pH of solution
leads against pathogenic and challenging VRE and MRSA strains. adjusted to 12 with 4 N NaOH, and then (Boc)2O (1.5 equiv.) was
Short sequential size facilitates the ease of synthesis along with added to the reaction. After 15 min, pH was again adjusted to 12
lower production cost in this class of peptides. The peptides and another (Boc)2O (1.5 equiv.) was added, and stirred (12 h, RT).
exhibited no apparent mammalian cytotoxicity confirmed by Vero Solvent removed and methanol (10 mL) was added, and stirred
Cells experiment. It is noted that variation in lipophilicity, partic- again for 12 h. The Reaction oncentrated under vacuum and fol-
ularly imparted by substituent at the imidazole ring played a vital lowed by pH adjustment of resulting N-a-Boc-L-His(2-aryl)-O-Naþ
role in antifungal potency and selectivity. The potency is found to with aq. KHSO4 to pH 3 gave free N-a-Boc-2-aryl-L-histidines. After
be related to the bulkiness of the substituent attached to the concentrated reaction under vacuum and the mixture was extrac-
imidazole ring. Membrane selectivity study with tryptophan fluo- ted through tert-butanol, afforded N-a-Boc-2-aryl-L-histidines (4a-
rescence quenching and mechanistic investigations by electron e) in 91e95% yields.
microscopy prove that 14f is selective to fungal cell surface and
disrupt the fungal membranes resulting in killing of pathogenic 4.1.2. Synthesis of L-Arg-OMe (6)
cells. This study experimentally strengthen the concept of the role L-Arg-OH (5, 1 equiv.) was purged with HCl gas in MeOH for 2 h
of suitably modified amino acids, their sequence, and importance of at RT and stirred for 8 h at 10  Cto afford pure L-Arg-OMe (6) in 95%
the synthetically tuned amphiphilicity in the antifunal peptide yields.
design. Indentification of the lead peptide 14f provided insights in
the design, amino acids placement in a sequence, and role of the 4.1.3. Synthesis of L-Arg-NHBzl (7)
modified amino acids. This study opens additional avenues for the In a dry flask, L-Arg-OH (5, 1 equiv.), CDI (1.5 equiv.) and ben-
discovery of ultrashort, membrane-disruptive, and selective zylamine (1.5 equiv.) was added and mixed, and allowed to stir at
peptide-based antifungal probes. ambient temperature for 12 h. The complete removal of solvent
under reduced pressure produced crude product, which was puri-
4. Experimental fied on basic alumina column to afford L-Arg-NHBzl (7) in 72%
yields [45].
4.1. General chemistry
4.1.4. Synthesis of N-a-Boc-L-Trp-L-Arg-OMe (8) and N-a-Boc-L-
All chemicals were purchased from commercial sources such as Trp-L-Arg-NHBzl (9)
Sigma-Aldrich, Acros Organics, Chem-Impex, Himedia, Avra and In a dry MW vial (10 mL) equiped with a magnetic stir bar, L-
Alfa-Aesar. Solution phase peptide synthesis was performed on Arg-OMe/NHBzl (1 equiv.) and DIEA (4 equiv.) was added togather.
CEM Microwave Discover® using MW assisted solution phase Next, Boc-L-Trp-OH (1.5 equiv.), DIC (1.5 equiv.) and HOBt (1.5
peptide synthesis protocol. 1H NMR were obtained on AVANCE III equiv.) were added to vial in DMF (5 mL). The reaction mixture was
400 Bruker (400 MHz) andchemical shifts are stated in parts per placed under MW heating (CEM Discover® microwave reactor) for
million (ppm, d scale) and are referenced to residual solvent peak in 30 min at 60  C. The reaction mixture was diluted with methanol
the NMR solvent or TMS. 13C NMR was recorded on AVANCE III 400 for homogenization and purified through column chromatography
Bruker (100 MHz) and decoupled by broad band decoupling. The system to afford pure Boc-L-Trp-L-Arg-OMe/NHBzl (8 and 9) in 78%
following abbreviations were utilized to present peptides peak and 71% yields, respectively.
11
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

4.1.5. General synthesis of L-Trp-L-Arg-OMe (10) and L-Trp-L-Arg- J ¼ 7.84, 1H), 7.50 (d, J ¼ 7.16, 3H), 7.36 (d, J ¼ 8.04, 1H), 7.19 (s, 1H),
NHBzl (11) 7.13e7.09 (m, 1H), 7.05e7.01 (m, 1H), 4.66 (t, J ¼ 7.02, 1H), 4.41e4.38
The dipeptides, Boc-L-Trp-L-Arg-OMe/NHBzl (8 or 9) and 6 N (m, 2H), 3.66 (s, 3H), 3.27 (d, J ¼ 6.44, 1H), 3.22 (t, J ¼ 6.52, 1H), 3.17
methanolic HCl (5 mL) were stirred at ambient temperature for (d, J ¼ 9.28, 1H), 3.12 (t, J ¼ 7.10, 2H), 3.06e2.99 (m, 1H), 2.78 (q,
20 min, and thenconcentrated under vacuum to get 10 and 11 in J ¼ 7.60, 7.52, 2H), 1.83e1.78 (m, 1H), 1.68e1.65 (m, 1H), 1.56e1.52
92% and 93% yields, respectively. (m, 2H), 1.38 (s, 9H), 1.30 (t, J ¼ 7.60, 3H); 13C NMR [100 MHz,
CD3OD]: d 172.9, 171.8, 171.4, 157.2, 156.5, 149.5, 136.6, 130.4, 129.0,
4.1.6. General procedure of the synthesis of N-a-Boc-L-His(2-aryl)- 128.9, 127.3, 126.5, 126.3, 123.5, 121.1, 118.5, 117.8, 111.0, 108.8, 79.9,
L-Trp-L-Arg-OMe (12a-f) and N-a-Boc-L-His(2-aryl)-L-Trp-L-Arg- 53.4, 51.9, 51.7, 51.5, 40.4, 28.3, 28.2, 27.3, 27.1, 27.0, 24.5, 14.2;
NHBzl (13a-f) HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 716.3884, found:
In a dry MW vial (10 mL) equiped with a magnetic stir bar, 10 or 716.3868; HPLC: tR ¼ 24.1 min, 96.2% purity.
11 (1 equiv.) was added and then DIEA (4 equiv.), DIC (1.5 equiv.), N-a-Boc-L-His(2-biphenyl)-L-Trp-L-Arg-OMe (12e). Yield:
HOBt (1.5 equiv.), dry DMF (3 mL) and N-a-Boc-2-aryl-L-histidines 60%. 1H NMR [400 MHz, CD3OD]: d 8.00 (d, J ¼ 6.32, 2H), 7.86 (d,
(4a-f, 1.5 equiv.) were added, and MW irradiation was applied at J ¼ 6.80, 2H), 7.72 (d, J ¼ 7.52, 2H), 7.58 (d, J ¼ 7.40, 1H), 7.51 (t,
60  C (30 min). Solvent was evaporated under reduced pressure J ¼ 7.46, 2H), 7.42 (t, J ¼ 7.28, 1H), 7.35 (d, J ¼ 8.08, 1H), 7.19 (s, 2H),
and the crude peptide was precipitated using cold diethyl ether. 7.10 (t, J ¼ 7.42, 1H), 7.02 (J ¼ 7.36, 1H), 4.66 (t, J ¼ 6.70, 1H), 4.42, (br.
The peptides were purified on preparative-HPLC system using a C- s, 2H), 3.65 (s, 3H), 3.26 (t, J ¼ 7.30, 3H), 3.20e3.08 (m, 2H),
18 RP-column with CH3CN and water a solvent system containing 3.06e3.00 (m, 1H), 1.80 (t, J ¼ 6.54, 1H), 1.65e1.52 (m, 3H), 1.38 (s,
0.08% TFA to yield Boc-L-His(2-aryl)-L-Trp-L-Arg-OMe/NHBzl (12a- 9H); 13C NMR [100 MHz, CD3OD]: d 172.9, 171.8, 171.7, 157.1, 145.9,
f and 13a-f). 139.3, 136.6, 133.3, 130.5, 128.7, 127.9, 127.5, 127.4, 126.6, 126.5,
123.4, 121.0, 118.5, 117.8, 110.9, 108.8, 79.7, 54.4, 53.8, 51.8, 51.4, 40.4,
4.1.7. General synthetic procedure for L-His(2-aryl)-L-Trp-L-Arg- 29.3, 28.2, 27.1, 24.5, 22.3; HRMS(ESI-TOF): calculated for m/z
OMe (14a-f) and L-His(2-aryl)-L-Trp-L-Arg-NHBzl (15a-f) [M þ Hþ]: 764.3884, found: 764.3884; HPLC: tR ¼ 26.1 min, 99.8%
N-a-Boc-L-His(2-aryl)-L-Trp-L-Arg-OMe/NHBzl (12a-f or 13a-f) purity.
upon reaction with 6 N HCl (5 mL) at 25  C for 15e20 min resulted N-a-Boc-L-His[(2-p-(n-butylphenyl)]-L-Trp-L-Arg-OMe (12f).
in the removal of Boc group to give the desired tripeptides (14a-f Yield: 59% 1H NMR [400 MHz, CD3OD]: d 7.87e7.80 (m, 2H), 7.55 (d,
and 15a-f). J ¼ 7.76, 1H), 7.51e7.47 (m, 2H), 7.41 (d, J ¼ 8.12, 1H), 7.28 (s, 1H),
7.22 (d, J ¼ 4.32, 1H), 7.15 (t, J ¼ 7.12, 1H), 7.06 (t, J ¼ 7.08, 1H), 4.64 (t,
4.1.8. Characterization of synthesized peptides J ¼ 7.56, 1H), 4.43 (t, J ¼ 6.96, 1H), 4.33e4.29 (m, 1H), 3.64 (s, 3H),
N-a-Boc-L-His-L-Trp-L-Arg-OMe (12a). Yield: 56%. 1H NMR 3.25e3.18 (m, 3H), 3.10 (t, J ¼ 6.90, 2H), 2.72 (t, J ¼ 7.54, 2H),
[400 MHz, CD3OD]: d 8.73 (s, 1H), 7.61 (d, J ¼ 7.84, 1H), 7.37 (d, 1.80e1.74 (m, 1H), 1.67e1.59 (m, 3H), 1.49e1.44 (m, 2H), 1.39e1.32
J ¼ 8.08, 1H), 7.19 (d, J ¼ 6.80, 2H), 7.12 (t, J ¼ 7.54, 1H), 7.04 (d, (m, 11H), 0.93 (t, J ¼ 7.36, 3H); 13C NMR [100 MHz, CD3OD]: d 173.2,
J ¼ 7.44, 1H), 4.67 (t, J ¼ 6.90, 1H), 4.44 (t, J ¼ 6.68, 1H), 4.35 (t, 172.9, 172.5, 156.8, 153.9, 148.4, 144.7, 136.3, 129.7, 129.7, 127.2,
J ¼ 6.78, 1H), 3.6849 (s, 3H), 3.30e3.11 (m, 5H), 2.9607 (dd, 126.9, 126.4, 123.9, 121.5, 119.9, 118.9, 117.8, 111.4, 108.4, 80.8, 54.6,
J ¼ 15.26, 8.34, 1H), 1.91e1.85 (m, 1H), 1.73e1.57 (m, 3H), 1.39 (s, 53.5, 52.3, 52.1, 40.4, 34.9, 32.9, 28.1, 27.2, 27.1, 27.1, 24.3, 21.7, 13.0;
9H); 13C NMR [100 MHz, CD3OD]: d 172.7, 171.8, 169.7, 157.2, 156.2, HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 744.4197, found:
136.6, 133.4, 129.6, 127.4, 123.5, 121.1, 118.5, 117.8, 117.1, 111.0, 108.8, 744.4180; HPLC: tR ¼ 27.1 min, 96.4% purity.
79.9, 54.4, 53.5, 51.8, 51.5, 40.4, 29.3, 28.7, 28.2, 27.1, 24.6; N-a-Boc-L-His-L-Trp-L-Arg-NHBzl (13a). Yield: 55%. 1H NMR
HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 612.3258, found: [400 MHz, CD3OD]: d 8.70 (s, 1H), 7.58 (d, J ¼ 7.72, 1H), 7.33 (d,
612.3259; HPLC: tR ¼ 20.1 min, 95.3% purity. J ¼ 8.08, 1H), 7.30e7.26 (m, 2H), 7.23e7.18 (m, 4H), 7.15 (s, 1H), 7.10
N-a-Boc-L-His(2-phenyl)-L-Trp-L-Arg-OMe (12b). Yield: 63%. (t, J ¼ 7.34, 1H), 7.02 (t, J ¼ 7.34, 1H), 4.63 (t, J ¼ 6.60, 1H), 4.31e4.23
1
H NMR [400 MHz, CD3OD]: d 7.93 (d, J ¼ 6.64, 2H), 7.65 (d, J ¼ 7.32, (m, 4H), 3.26e3.20 (m, 2H), 3.10e3.06 (m, 3H), 2.96e2.90 (m, 1H),
3H), 7.58 (d, J ¼ 8.20, 1H), 7.35 (d, J ¼ 8.04, 1H), 7.27 (s, 1H), 7.20 (d, 1.85e1.78 (m, 1H), 1.63e1.48 (m, 3H), 1.36 (s, 9H) 13C NMR
J ¼ 8.16, 1H), 7.10 (t, J ¼ 7.48, 1H), 7.02 (t, J ¼ 7.24, 1H), 4.66 (t, [100 MHz, CD3OD]: d 172.9, 171.9, 171.6, 157.1, 156.3, 138.2, 136.6,
J ¼ 6.90, 1H), 4.44e4.38 (m, 2H), 3.65 (s, 3H), 3.28e3.17 (m, 3H), 133.4, 129.6, 128.1, 127.2, 127.0, 126.8, 123.4, 121.3, 118.7, 117.9, 117.1,
3.13e3.03 (m, 3H), 1.83e1.80 (m, 1H), 1.68e1.64 (m, 1H), 1.57e1.52 111.1, 108.8, 79.9, 54.6, 53.6, 53.0, 42.6, 40.4, 28.5, 27.1, 27.0, 26.7,
(m, 2H), 1.37 (s, 9H); 13C NMR [100 MHz, CD3OD]: d 172.9, 171.7, 24.7; HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 687.3731, found:
171.3, 157.1, 156.1, 144.5, 136.6, 132.1, 130.39, 129.4, 127.3, 126.4, 687.3725; HPLC: tR ¼ 22.2 min, 95.4% purity.
123.4, 122.9, 121.1, 118.5, 118.0, 117.8, 111.0, 108.8, 79.8, 54.4, 53.4, N-a-Boc-L-His(2-phenyl)-L-Trp-L-Arg-NHBzl (13b). Yield:
51.9, 51.4, 40.3, 28.2, 27.3, 27.1, 24.6; HRMS(ESI-TOF), calculated for 62%. 1H NMR [400 MHz, CD3OD]: d 7.92 (d, J ¼ 6.64, 2H), 7.68e7.62
m/z [M þ Hþ]: 688.3571, found: 688.3552; HPLC: tR ¼ 22.1 min, (m, 3H), 7.57 (d, J ¼ 7.88, 1H), 7.34 (d, J ¼ 8.00, 1H), 7.30e7.27 (m,
99.8%. 3H), 7.23e7.21 (m, 3H), 7.17 (s, 1H), 7.11 (t, J ¼ 7.42, 1H), 7.03 (t,
N-a-Boc-L-His(2-tolyl)-L-Trp-L-Arg-OMe (12c). Yield: 65%. 1H J ¼ 7.14, 1H), 4.66 (t, J ¼ 6.60, 1H), 4.41 (t, J ¼ 6.78, 1H), 4.33e4.29 (m,
NMR [400 MHz, CD3OD]: d 7.80 (t, J ¼ 7.62,2H), 7.58 (d, J ¼ 7.04, 1H), 1H), 4.26e4.24 (m, 2H), 3.27e3.14 (m, 3H), 3.11e2.99 (m, 3H),
7.49e7.47 (m, 3H), 7.35 (d, J ¼ 8.16, 1H), 7.11 (t, J ¼ 7.14, 1H), 7.03 (t, 1.85e1.80 (m, 1H), 1.62e1.57 (m, 1H), 1.49 (d, J ¼ 3.68, 2H), 1.37 (s,
J ¼ 6.76, 1H), 4.66 (t, J ¼ 7.00, 1H), 4.42e4.39 (m, 2H), 3.67 (s, 3H), 9H) 13C NMR [100 MHz, CD3OD]: d 172.9, 171.8, 171.6, 157.1, 156.4,
3.27e3.19 (m, 2H), 3.15e2.99 (m, 4H), 2.47 (s, 3H), 1.86e1.81 (m, 144.4, 138.2, 136.6, 132.1, 130.5, 129.5, 128.1, 127.2, 127.0, 126.8,
1H), 1.69e1.65 (m, 1H), 1.57e1.52 (m, 2H), 1.38 (s, 9H); 13C NMR 126.7, 126.4, 123.4, 122.8, 121.2, 118.7, 117.8, 111.1, 108.8, 79.8, 54.7,
[100 MHz, CD3OD]: d 174.3, 173.2, 172.8, 158.6, 157.9, 144.6, 131.9, 53.6, 53.0, 42.6, 40.4, 28.5, 27.1, 26.7, 26.6, 24.7; HRMS(ESI-TOF):
131.5, 131.5, 127.7, 127.6, 127.5, 124.9, 122.5, 121.6, 119.9, 119.2, 112.4, calculated for m/z [M þ Hþ]: 763.4044, found: 763.4034; HPLC:
110.2, 81.0, 54.0, 53.3, 53.1, 52.9, 41.8, 29.6, 28.7, 28.6, 26.0, 21.5; tR ¼ 24.2 min, 96.8% purity.
HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 702.3727, found: N-a-Boc-L-His(2-tolyl)-L-Trp-L-Arg-NHBzl (13c). Yield: 66%.
1
702.3724; HPLC: tR ¼ 23.3 min, 96.2%. H NMR [400 MHz, CD3OD]: d 7.84e7.78 (m, 2H), 7.51e7.45 (m, 3H),
N-a-Boc-L-His(2-ethylphenyl)-L-Trp-L-Arg-OMe (12d). Yield: 7.39 (t, J ¼ 7.02, 1H), 7.32e7.27 (m, 4H), 7.21e7.14 (m, 4H), 7.08 (t,
61%. 1H NMR [400 MHz, CD3OD]: d 7.84 (t, J ¼ 6.88,2H), 7.58 (d, J ¼ 8.52, 1H), 4.66 (t, J ¼ 7.12, 1H), 4.40e4.36 (m, 1H), 4.34e4.21 (m,
12
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

2H), 4.10 (s, 1H), 3.50e3.45 (m, 1H), 3.26e3.25 (m, 1H), 3.21e3.16 122.9, 120.8, 120.2, 117.8, 117.6, 116.3, 110.2, 106.4, 53.4, 51.1, 50.8,
(m, 2H), 3.07e3.02 (m, 2H), 2.45 (s, 3H), 1.79e1.53 (m, 4H), 1.33 (s, 50.2, 38.7, 26.4, 25.4, 24.8, 22.3; HRMS(ESI-TOF): calculated for m/z
9H); 13C NMR [100 MHz, CD3OD]: d 171.9, 171.7, 170.9, 156.5, 153.4, [M þ Hþ]: 588.3047, found: 588.3029; HPLC: tR ¼ 17.8 min, 96.1%
145.5, 135.1, 131.4, 130.1, 129.4, 128.2, 127.0, 126.8, 126.8, 126.5, 126.1, purity.
125.1, 123.7, 121.0, 119.0, 117.7, 111.4, 108.3, 79.6, 54.8, 54.6, 53.5, L-His(2-tolyl)-L-Trp-L-Arg-OMe (14c). Yield: 67%. 1H NMR
42.5, 40.3, 28.1, 27.1, 26.7, 26.3, 24.2, 20.3; HRMS(ESI-TOF): calcu- [400 MHz, CD3OD]: d 7.89 (br. s, 2H), 7.57e7.55 (m, 2H), 7.46 (br. s,
lated for m/z [M þ Hþ]: 777.4200, found: 777.4193; HPLC: 2H), 7.30 (d, J ¼ 7.56, 1H), 7.23e7.20 (m, 1H), 7.06 (t, J ¼ 6.56, 1H),
tR ¼ 25.2 min, 95.6% purity. 6.98 (t, J ¼ 5.34, 1H), 4.73e4.71 (m, 2H), 4.30 (br. s, 1H), 3.63 (s, 3H),
N-a-Boc-L-His(2-ethylphenyl)-L-Trp-L-Arg-NHBzl (13d). 3.14e2.83 (m, 6H), 2.45 (s, 3H), 1.67e1.50 (m, 4H); 13C NMR
Yield: 58%. 1H NMR [400 MHz, CD3OD]: d 7.83 (d, J ¼ 8.00, 2H), 7.57 [100 MHz, CD3OD]: d 172.9, 172.4, 171.6, 157.5, 149.3, 143.5, 136.6,
(d, J ¼ 7.80, 1H), 7.47 (d, J ¼ 8.28, 2H), 7.34 (d, J ¼ 8.08, 1H), 134.1, 130.2, 128.0, 127.4, 127.2, 127.1, 118.5, 117.1, 116.9, 111.0, 108.6,
7.30e7.26 (m, 2H), 7.23e7.18 (m, 5H), 7.11 (t, J ¼ 7.20, 1H), 7.03 (t, 58.7, 53.3, 52.4, 51.8, 40.8, 30.5, 29.3, 29.1, 22.2, 20.4; HRMS(ESI-
J ¼ 7.40, 1H), 4.66 (t, J ¼ 7.20, 1H), 4.40 (t, J ¼ 7.10, 1H), 4.33e4.30 (m, TOF): calculated for m/z [M þ Hþ]: 602.3203, found: 602.3198;
1H), 4.25 (d, J ¼ 2.40, 2H), 3.26 (d, J ¼ 6.52, 2H), 3.19e3.14 (m, 1H), HPLC: tR ¼ 18.6 min, 95.6% purity.
3.09 (t, J ¼ 6.76, 2H), 3.02e2.97 (m, 1H), 2.75 (q, J ¼ 7.60, 7.56, 2H) L-His(2-ethylphenyl)-L-Trp-L-Arg-OMe (14d). Yield: 64%. 1H
1.84e1.78 (m, 1H), 1.64e1.57 (m, 1H), 1.52e1.46 (m, 2H), 1.36 (s, 9H), NMR [400 MHz, CD3OD]: d 7.95e7.88 (m, 2H), 7.61e7.51 (m, 4H),
1.28 (t, J ¼ 7.60, 3H); 13C NMR [100 MHz, CD3OD]: d 172.9, 171.9, 7.34 (d, J ¼ 8.00, 1H), 7.24 (s, 1H), 7.10 (t, J ¼ 7.40, 1H), 7.01 (t, J ¼ 7.30,
171.7, 157.1, 156.3, 149.5, 144.6, 138.1, 136.5, 130.1, 129.0, 128.1, 127.2, 1H), 4.54 (t, J ¼ 6.00, 1H), 4.32 (br. s, 1H), 3.65 (s, 3H), 3.53e3.42 (m,
127.0, 126.8, 126.4, 123.5, 121.3, 120.2, 118.8, 117.9, 117.6, 111.2, 108.7, 3H), 3.23e3.17 (m, 1H), 3.09e3.03 (m, 2H), 2.81e2.74 (m, 2H)
80.0, 54.7, 53.7, 53.1, 42.6, 40.4, 28.5, 28.3, 27.1, 26.1, 24.7, 14.2; 1.76e1.65 (m, 2H), 1.56e1.50 (m, 1H), 1.45e1.37 (m, 1H), 1.32e1.26
HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 791.4357, found: (m, 3H); 13C NMR [100 MHz, CD3OD]: d 173.3, 171.7, 167.4, 157.1,
791.4333; HPLC: tR ¼ 26.2 min, 95.8% purity. 149.7, 145.6, 136.6, 129.1, 129.0, 127.1, 126.7, 123.7, 121.1, 119.6, 118.5,
N-a-Boc-L-His(2-biphenyl)-L-Trp-L-Arg-NHBzl (13e). Yield: 117.8, 111.0, 108.5, 54.8, 53.1, 52.3, 51.7, 40.7, 29.2, 28.5, 28.2, 26.7,
61%. 1H NMR [400 MHz, CD3OD]: d 8.00 (d, J ¼ 8.08, 2H), 7.90 (d, 24.7, 14.4; HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 616.3360,
J ¼ 8.32, 2H), 7.71 (d, J ¼ 7.32, 2H), 7.57 (d, J ¼ 7.92, 1H), 7.53 (t, found: 616.3357; HPLC: tR ¼ 19.9 min, 97.0% purity.
J ¼ 7.46, 2H), 7.45 (d, J ¼ 7.12, 1H), 7.33 (d, J ¼ 8.08, 1H), 7.30 (s, 1H), L-His(2-biphenyl)-L-Trp-L-Arg-OMe (14e). Yield: 56%. 1H NMR
7.26 (d, J ¼ 7.04, 2H), 7.21 (t, J ¼ 2.94, 2H), 7.19 (d, J ¼ 4.44, 2H), 7.10 [400 MHz, CD3OD]: d 8.10 (br. s, 2H), 7.93 (br. s, 2H), 7.73 (br. s, 2H),
(t, J ¼ 7.42, 1H), 7.02 (t, J ¼ 7.44, 1H), 4.66 (t, J ¼ 6.72, 1H), 4.42 (t, 7.64e7.59 (m, 2H), 7.50e7.42 (m, 3H), 7.32e7.25 (m, 2H), 7.07 (t,
J ¼ 6.72, 1H), 4.34e4.30 (m, 1H), 4.25 (s 2H), 3.27e3.15 (m, 3H), J ¼ 7.32, 1H), 6.98 (br. s, 1H), 4.48e4.45 (m, 1H), 4.33 (br. s, 2H), 3.62
3.11e3.03 (m, 3H), 1.85e1.79 (m, 1H), 1.66e1.44 (m, 4H), 1.36 (s, (s, 3H), 3.21e3.16 (m, 3H), 3.04e3.01 (m, 3H), 1.72e1.51 (m, 4H); 13C
9H); 13C NMR [100 MHz, CD3OD]: d 173.0, 171.8, 171.6, 157.1, 156.3, NMR [100 MHz, CD3OD]: d 176.2, 172.5, 171.1, 155.0, 146.8, 140.1,
148.0, 145.0, 144.2, 139.0, 138.2, 136.6, 130.4, 128.8, 128.2, 128.1, 138.8, 137.4, 131.3, 129.1, 129.0, 128.3, 127.6, 127.5, 127.5, 127.4, 126.7,
127.8, 127.2, 126.9, 126.9, 126.8, 126.7, 123.4, 121.5, 121.2, 118.7, 118.0, 125.8, 123.6, 121.7, 119.0, 117.7, 111.7, 107.8, 54.5, 53.3, 52.5, 52.2,
117.8, 111.1, 108.7, 79.8, 54.7, 53.6, 53.0, 42.6, 40.4, 30.5, 29.3, 28.5, 40.1, 30.2, 29.7, 27.7, 25.0; HRMS(ESI-TOF): calculated for m/z
27.1, 24.7; HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 839.4357, [M þ Hþ]: 664.3360, found: 664.3345; HPLC: tR ¼ 20.9 min, 96.8%
found: 839.4339; HPLC: tR ¼ 27.5 min, 99.8% purity. purity.
N-a-Boc-L-His[(2-p-(n-butylphenyl)]-L-Trp-L-Arg-NHBzl L-His[(2-p-(n-butylphenyl)]-L-Trp-L-Arg-OMe (14f). Yield:
(13f). Yield: 64%. 1H NMR [400 MHz, CD3OD]: d 7.93e7.82 (m, 2H), 55%. 1H NMR [400 MHz, CD3OD]: d 7.93 (d, J ¼ 7.68, 2H), 7.59 (d,
7.56 (d, J ¼ 7.68, 1H), 7.50e7.45 (m, 3H), 7.34e7.31 (m, 1H), J ¼ 7.76, 1H), 7.45 (s, 1H), 7.49 (d, J ¼ 7.64, 2H), 7.35 (d, J ¼ 8.00, 1H),
7.28e7.18 (m, 6H), 7.10 (t, J ¼ 7.54, 1H), 7.02 (t, J ¼ 7.08, 1H), 4.65 (t, 7.26 (d, J ¼ 6.80, 1H), 7.09 (t, J ¼ 7.38, 1H), 7.01 (t, J ¼ 7.32, 1H), 4.72
J ¼ 6.74, 1H), 4.40 (t, J ¼ 6.62, 1H), 4.33e4.29 (m, 1H), 4.24 (d, (t, J ¼ 7.12, 1H), 4.33e4.30 (m, 2H), 3.63 (s, 3H), 3.54e3.43 (m, 2H),
J ¼ 7.72, 2H), 3.42e3.36 (m, 1H), 3.25 (d, J ¼ 6.68, 1H), 3.18e3.08 (m, 3.23e3.18 (m, 1H), 3.09e3.02 (m, 2H), 2.74 (t, J ¼ 7.38, 2H),
3H), 3.03e2.97 (m, 1H), 2.75e2.70 (m, 2H), 1.82e1.78 (m, 1H), 1.73e1.71 (m, 1H), 1.69e1.63 (m, 3H), 1.53e1.49 (m, 2H), 1.41e1.35
1.66e1.61 (m, 3H), 1.42e1.41 (m, 2H), 1.37 (s, 9H), 1.29 (s, 2H), (m, 2H), 0.95 (t, J ¼ 7.48, 3H); 13C NMR [100 MHz, CD3OD]: d 173.3,
0.98e0.93 (m, 3H); 13C NMR [100 MHz, CD3OD]: d 173.3, 172.9, 171.8, 167.5, 157.0, 148.3, 145.6, 136.5, 129.5, 127.1, 126.9, 126.5, 123.7,
171.8, 157.1, 156.4, 148.1, 144.6, 138.2, 136.5, 130.1, 129.5, 128.1, 127.0, 121.1, 120.1, 119.4, 118.5, 117.8, 111.0, 108.5, 54.88, 52.4, 51.7, 51.6,
126.8, 126.3, 126.2, 123.4, 121.2, 120.2, 118.7, 117.8, 117.6, 111.1, 108.7, 40.5, 35.1, 33.1, 28.1, 27.3, 26.5, 24.6, 21.8, 12.9; HRMS(ESI-TOF):
79.8, 54.7, 53.0, 51.7, 42.6, 40.4, 35.0, 33.0, 29.3, 28.5, 27.1, 24.7, 21.9 calculated for m/z [M þ Hþ]: 644.3673, found: 644.3660; HPLC:
12.8; HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 819.4670, tR ¼ 22.3 min, 97.1% purity.
found: 819.4665; HPLC: tR ¼ 28.4 min, 99.8% purity. L-His-L-Trp-L-Arg-NHBzl (15a). Yield: 62%. 1H NMR [400 MHz,
L-His-L-Trp-L-Arg-OMe (14a). Yield: 58%. 1H NMR [400 MHz, CD3OD]: d 8.81 (br. s, 1H), 7.64 (d, J ¼ 7.04, 1H), 7.49e7.43 (m, 2H),
CD3OD]: d 8.85 (br. s, 1H), 7.64e7.44 (m, 2H), 7.35e7.26 (m, 2H), 7.33e7.23 (m, 6H), 7.08 (t, J ¼ 7.16, 1H), 7.00 (t, J ¼ 7.10, 1H),
7.10e7.02 (m, 2H), 4.49 (br. s, 2H), 4.27 (br. s, 1H), 3.68 (s, 3H), 4.41e4.24 (m, 5H), 3.78e3.63 (m, 1H), 3.43 (br. s, 1H), 3.15 (br. s,
3.48e3.45 (m, 2H), 3.25e3.19 (m, 4H), 2.02e1.66 (m, 4H); 13C NMR 3H), 2.81 (s, 1H), 1.84e1.58 (m, 4H); 13C NMR [100 MHz, CD3OD]:
[100 MHz, CD3OD]: d 171.8, 168.7, 167.4, 157.1, 132.1, 127.8, 126.2, d 171.6, 170.0, 167.4, 157.1, 138.3, 136.6, 134.6, 128.1, 127.1126.8, 121.3,
123.8, 121.1, 119.0, 118.5, 117.8, 111.0, 52.3, 51.7, 51.7, 40.7, 29.3, 27.4, 118.6, 118.1, 111.0, 108.7, 54.8, 51.7, 42.6, 29.3, 28.7, 27.4, 26.4, 24.9;
26.4, 24.8; HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 512.2734, HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 587.3206, found:
found: 512.2726; HPLC: tR ¼ 15.0 min, 95.8% purity. 587.3196; HPLC: tR ¼ 20.2 min, 95.4% purity.
L-His(2-phenyl)-L-Trp-L-Arg-OMe (14b). Yield: 65%. 1H NMR L-His(2-phenyl)-L-Trp-L-Arg-NHBzl (15b). Yield: 66%. 1H NMR
[400 MHz, CD3OD]: d 7.85 (t, J ¼ 7.48, 2H), 7.71e7.64 (m, 3H) [400 MHz, CD3OD]: d 7.92e7.85 (m, 2H), 7.75e7.63 (m, 4H),
7.50e7.40 (m, 3H), 7.18 (t, J ¼ 7.28, 2H), 7.08 (t, J ¼ 7.16, 1H), 4.61 (t, 7.49e7.31 (m, 6H), 7.18e7.09 (m, 4H), 4.36 (br. s, 1H), 4.05 (s, 2H),
J ¼ 7.80, 1H), 4.37e4.33 (m, 1H), 4.00e3.96 (m, 1H), 3.52 (s, 3H), 3.93e3.87 (m, 2H), 3.50e3.46 (m, 1H), 3.24e3.06 (m, 3H), 2.91 (br.
3.38e3.34 (m, 2H), 3.21e3.13 (m, 2H), 2.98e2.89 (m, 2H), 1.50e1.39 s, 2H), 1.52e1.34 (m, 2H), 1.21e1.13 (m, 2H); 13C NMR [100 MHz,
(m, 2H), 1.25e1.16 (m, 2H); 13C NMR [100 MHz, CD3OD]: d 171.2, CD3OD]: d 172.8, 171.9, 167.7156.4, 145.4, 137.6, 135.9, 132.7, 129.7,
170.9, 166.0, 154.9, 143.9, 134.5, 131.2, 128.1, 125.4, 125.2, 124.6, 128.6, 127.4, 127.1, 127.0, 126.6, 126.3, 124.3, 122.3, 122.1, 119.5, 119.3,
13
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

118.0, 111.9, 108.0, 54.9, 53.6, 51.9, 42.9, 40.4, 28.3, 26.9, 26.4, 24.1; albicans (ATCC 10231), Aspergillus fumigatus (ATCC 1022), Crypto-
HRMS(ESI-TOF): calculated for m/z [M þ Hþ]: 663.3519, found: coccus neoformans (ATCC 90113), and bacterial strains of MRSA
663.3510; HPLC: tR ¼ 20.7 min, 96.8% purity. (ATCC 33591), Escherichia coli (ATCC 35218), Klebsiella pneumoniae
L-His(2-tolyl)-L-Trp-L-Arg-NHBzl (15c). Yield: 63%. 1H NMR (ATCC BAA-1705), Pseudomonas aeruginosa (ATCC 27853), and VRE
[400 MHz, CD3OD]: d 7.87 (d, J ¼ 7.76, 2H), 7.51e7.44 (m, 4H), 7.34 (ATCC MP-1), at the highest concentration of 20 mg/mL. Suscepti-
(d, J ¼ 8.00, 1H), 7.29e7.22 (m, 4H), 7.16e7.09 (m, 3H), 7.02 (t, bility of bacterial and fungal to the tested peptides was evaluated
J ¼ 7.78, 1H), 4.32 (t, J ¼ 6.14, 2H), 4.20e4.12 (m, 3H), 3.49e3.39 (m, according to a modified CLSI protocol. For pharmacological evalu-
2H), 3.16 (d, J ¼ 6.92, 2H), 3.06 (t, J ¼ 6.76, 2H), 2.44 (s, 3H), ation, all tested peptides through serial dilution in 20% DMSO/sa-
1.75e1.72 (m, 2H), 1.63e1.59 (m, 2H); 13C NMR [100 MHz, CD3OD]: line, added in duplicate to 96-well plates, and inocula were made
d 174.9, 171.0, 168.8, 156.1, 145.6, 139.0, 138.2, 130.1, 128.1, 128.0, by correcting the OD630 of microbe suspensions in incubation
126.9, 126.8, 126.7, 126.5, 123.6, 121.2, 118.6, 117.9, 117.7, 111.0, 108.6, broth [Sabouraud Dextrose (Difco) for C. neoformans, cation
57.0, 55.4, 53.5, 42.6, 40.6, 31.7, 29.3, 28.7, 24.7, 20.0; HRMS(ESI- adjusted Mueller-20 Hinton (Difco) at pH 7.3 for non-mycobacterial
TOF): calculated for m/z [M þ Hþ]: 677.3676, found: 677.3670; bacteria to afford: C. neoformans 1.5  103 CFU/mL, non-
HPLC: tR ¼ 20.9 min, 96.1% purity. mycobacterial bacteria 5  105 CFU/mL, and for detailed test pro-
L-His(2-ethylphenyl)-L-Trp-L-Arg-NHBzl (15d). Yield: 61%. 1H cedure see SI [61].
NMR [400 MHz, CD3OD]: d 7.93 (br. s, 2H), 7.54e7.47 (m, 4H),
7.31e7.18 (m, 7H), 7.06e6.98 (m, 2H), 4.56 (br. s, 1H), 4.41e4.20 (m,
4.4. Cytotoxicity assay
4H), 3.31 (br. s, 2H), 3.20e2.92 (m, 4H), 2.74 (br. s, 2H), 1.77e1.65
(m, 2H), 1.53e1.42 (m, 2H), 1.25 (s, 3H); 13C NMR [100 MHz,
Cytotoxicity of tested synthetic tripeptides were analyzedin a
CD3OD]: d 174.2, 173.0, 172.6, 157.2, 155.3, 149.7, 138.3, 136.6, 129.1,
mammalian cell line to evaluate itssafety profile. All cells were
128.1, 127.0, 126.8, 121.3, 111.1, 108.5, 57.1, 53.5, 51.9, 42.7, 40.9, 28.7,
obtained from ATCC, and the in vitro mammalian cell cytotoxicity of
28.5, 27.4, 27.1, 24.9, 21.5, 14.6; HRMS(ESI-TOF): calculated for m/z
these synthetic peptides was carried out against non-cancerous
[M þ Hþ]: 691. 3832, found: 691.3826; HPLC: tR ¼ 22.8 min, 95.6%
kidney cells VERO. 25,000 cells/well were seeded and incubated
purity.
for 24 h for checked for confluency. Tested synthetic peptide
L-His(2-biphenyl)-L-Trp-L-Arg-NHBzl (15e). Yield: 58%. 1H
samples were added, and the plates were again incubated for 48 h.
NMR [400 MHz, CD3OD]: d 7.75 (d, J ¼ 7.92, 2H), 7.55 (d, J ¼ 5.68,
The number of viable cells were established according to the
2H), 7.43 (d, J ¼ 6.76, 3H), 7.29 (d, J ¼ 6.76, 3H), 7.16e7.07 (m, 6H),
modified version of neutral red assay, and doxorubicin and DMSO
6.92e6.87 (m, 3H), 6.73 (t, J ¼ 7.34, 1H), 4.63 (t, J ¼ 7.90, 1H), 4.33 (t,
were used as the positive and negative control, respectively, and for
J ¼ 6.76, 1H), 3.94 (t, J ¼ 6.76, 1H), 3.78 (s, 2H), 3.50e3.42 (m, 1H),
detailed test procedure see SI [52].
3.22e3.11 (m, 2H), 2.96 (br. s, 1H), 2.81 (br. s, 2H), 1.51e1.50 (m, 1H),
1.39e1.38 (m, 1H), 1.24e1.22 (m, 1H), 1.14e1.12 (m, 1H); 13C NMR
[100 MHz, CD3OD]: d 171.5, 170.2166.2154.7, 143.1, 142.5, 136.7, 4.5. Tryptophan quenching study
135.9, 134.1, 127.5, 126.8, 126.0, 125.7, 125.6, 125.3, 125.2, 125.0,
124.7, 122.5, 120.1, 119.3, 118.0, 117.6, 116.1, 110.2, 106.2, 53.4, 52.0, For quenching study, L-a-phosphatidylcholine (EYPC), L-a-
50.3, 41.1, 38.8, 26.6, 25.1, 25.0, 22.6; HRMS(ESI-TOF): calculated for phosphatidyl-D,L-glycerol (EYPG), acrylamide and cholesterol were
m/z [M þ Hþ]: 739.3832, found: 739.3827; HPLC: tR ¼ 23.4 min, procured from Sigma Aldrich. Briefly, SUVs were prepared consist
96.2% purity. of EYPC, EYPG and/or cholesterol, and the bacterial SUVs (EYP-
L-His[(2-p-(n-butylphenyl)]-L-Trp-L-Arg-NHBzl (15f). Yield: C:EYPG, 7:3) and mammalian cell membranes SUVs (EYPC:choles-
67%. 1H NMR [400 MHz, CD3OD]: d 7.89 (d, J ¼ 8.04, 2H), 7.55e7.42 terol, 10:1) developed by dissolving in chloroform, and followed by
(m, 5H), 7.34 (d, J ¼ 7.46, 1H), 7.28e7.21 (m, 3H), 7.15e7.08 (m, 3H), air dying. The fluorescence quenching was calculated using Stern-
7.01 (t, J ¼ 7.44, 1H), 4.52 (t, J ¼ 6.72, 1H), 4.33 (t, J ¼ 6.12, 1H), 4.18 (t, Volmer equation: F0/F ¼ 1 þ KSV [Q] and for detailed test pro-
J ¼ 7.20, 1H), 4.10 (s, 2H), 3.52e3.45 (m, 2H), 3.20 (d, J ¼ 7.12, 2H), cedure see SI [54,55].
3.06 (t, J ¼ 6.52, 2H), 2.74e2.67 (m, 2H), 1.76e1.70 (m, 1H),
1.61e1.56 (m, 4H), 1.36e1.30 (m, 3H), 0.91 (t, J ¼ 7.34, 3H); 13C NMR
4.6. Dynamic light scattering study
[100 MHz, CD3OD]: d 174.6, 173.2, 169.1, 158.2, 149.8, 146.9, 139.3,
137.7, 131.1, 131.1, 129.7, 128.4, 128.2, 127.9, 125.2, 122.9, 121.3, 121.3,
Hydrodynamic sizes of the synthetic peptides was assessed
120.7, 120.3, 119.3, 112.8, 109.6, 56.4, 55.0, 53.2, 44.0, 41.9, 36.4, 34.4,
through a Zetasizer nano-ZS (Malvern Instruments, UK). For size
29.7, 28.4, 27.9, 25.9, 23.2, 14.4; HRMS(ESI-TOF): calculated for m/z
analysis, peptides (1 mg) were dissolved in PBS pH 7.4 (1.0 mL) by
[M þ Hþ]: 719.4145, found: 719.4131; HPLC: tR ¼ 24.5 min, 95.4%
ultrasonic treatment and run for measurements at specified time
purity.
intervals. Experimental recordings were conducted at 25  C (90
and 60 s), and the viscosity and refractive index were considered of
4.2. FITC labelling of peptides
water 0.89 cp and 1.330, respectively. All readings were taken in
triplicates and an average of the three was compiled as the final
In a dry round bottom flask, pyridine/DMF/dichloromethane
reading for further observations.
(12:7:5, 1 mL), 14f or 12a (0.012 mmol, 1 equiv.) was added at
ambient temperature, and then FITC (1.2 equiv.) was added. The
reaction mixture was stirred for 15 h at room temperature. Reaction 4.7. Fluorescence spectroscopic study
mixture was conc under vacuum and purified using CH2Cl2:me-
thanol (95e92:5e8) using neutral alumina to get FITC-labeled A fluorescence quenching experiment for determining trypto-
peptides (14f and 12a). phan aggregation and stacking was evaluated with the Cary Eclipse
spectrofluorometer a magnetically stirred at 25  C (300 mL cuvette,
4.3. Assay for in vitro antimicrobial activity 1 cm) and thermostated cuvette using Ex 280 nm (bandwidth
5 nm). Here, 600 nm/min scan speed was used with the emission
The antimicrobial activities of all synthesized peptides (12a-f scan from 300 to 600 nm (bandwidth 10 nm). and the average of
and 15a-f) were evaluated against the fungal strains of Candida three scans was used with 5 min of equilibration period.
14
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

4.8. SEM study MIC minimum inhibitory concentration


Amp B amphotericin B
Sample preparation for this study was carried out by using the CTX cytotoxicity
peptides 14f and 12a (1 mg). Peptides were suspended in PBSpH 7.4 SI selectivity index
(1.0 mL) by ultrasonic irradiation and incubated at 37  C for 24 h. MRSA Methicillin-resistant S. aureus
The peptide solutions were placed on the coverslips previously VRE Vancomycin-resistant Enterococcus
attached to the SEM holder adhered by adhesive tape, and samples PBS phosphate buffer saline
were dried with carbon dioxide up to the critical point. In last step, PI propidium iodide
sample preparations used sputter gold coating, and finally peptides FITC fluorescein isothiocyanate
were examined using SEM (Hitachi Se3400 N, Japan) [31]. DAPI 40 ,6-diamidino-2-phenylindole
Cpx ciprofloxacin
4.9. CLSM and electron microscopic study SEM scanning electron microscopy
CLSM confocal laser scanning microscopy
In confocal and electron microscopic imaging studies, Ex exciataion
C. neoformans (ATCC350) cells (~1  104 CFU/mL) were used in a Em emission
RPMI-1640 medium containing peptide 14f at the MIC. HRTEM high resolution transmission electron microscopy
For PI study, after incubation (1 h, 30  C, shaking 200 rpm) with HMDS hexamethyldisilizane
14f, fungal cells were collected by centrifugation, and resuspended RP-HPLC reverse-phase high pressure liquid chromatography
in PBS (pH 7.4). Next, the funal cellsamples were observed by
confocal microscope at >560 nm wavelength (Olympus Fluoview Appendix A. Supplementary data
FV1000 SPD; Japan). Untreated cryptococcal cells served as a con-
trol in this study. Supplementary data to this article can be found online at
For DAPI study, after incubation, fungal cells were collected by https://doi.org/10.1016/j.ejmech.2021.113635.
centrifugation and resuspended in PBS, and DAPI added (3.01 mM,
10 min). The untreated fungalcells used for confocal imaging with References
DAPI used as a control. For confocal imaging with DAPI, samples
were examined with Ex/Em; 350/470 nm [31,62]. [1] M.A. Pfaller, S.A. Messer, R.N. Jones, M. Castanheira, Antifungal susceptibilities
of Candida, Cryptococcus neoformans and Aspergillus fumigatus from the Asia
For FITC-labeled peptide study, MIC of FITC labeled peptides was
and Western Pacific region: data from the SENTRY antifungal surveillance
also considered same as its parent peptide. After incubation with program (2010e2012), J. Antibiot. 68 (2015) 556e561.
labeled peptides for 1 h, fungal cells were collected and resus- [2] G.D. Brown, D.W. Denning, N.A. Gow, S.M. Levitz, M.G. Netea, T.C. White,
pended in PBS, then examined under confocal microscope (Ex/Em; Hidden killers: human fungal infections, Sci. Transl. Med. 4 (2012),
165rv13165rv13.
488/515 nm). In this study, 12a served as control for examination of [3] N.P. Wiederhold, Antifungal resistance: current trends and future strategies to
fungal cells [31,63]. combat, Infect. Drug Resist. 10 (2017) 249e259.
For morphological examination using SEM, the prepared sam- [4] R.C. May, N.R. Stone, D.L. Wiesner, T. Bicanic, K. Nielsen, Cryptococcus: from
environmental saprophyte to global pathogen, Nat. Rev. Microbiol. 14 (2016)
ples were dehydrated, and then protected with coverslip with 106e117.
hexamethyldisilizane (HMDS), and samples were dried at 25  C, [5] K. Sharma, S. Aaghaz, K. Shenmar, R. Jain, Short antimicrobial peptides, Recent
and then followed by gold coating and examined using the SEM Pat. Anti-Infect. Drug Discov. 13 (2018) 12e52.
[6] World Health Organization, Antimicrobial Resistance Global Report on Sur-
(JEOL 6100) [31,64]. For ultrastructure study, semithin and ultrathin veillance: 2014 Summary, 2014 (please see: https://apps.who.int/iris/
section was made using ultramicrotome (Ultramicotome Lecia EM bitstream/handle/10665/112647/WHO_HSE_PED_AIP_2014.2_eng.pdf.
UC6), and then sections of treated and control samples were placed (Accessed 15 January 2021).
[7] C. Coelho, A. Casadevall, Cryptococcal therapies and drug targets: the old, the
on the copper grid of 3.05 mm diameter (200 mess). Staining was
new and the promising, Cell Microbiol. 18 (2016) 792e799.
done with uranyl acetate and lead acetate, and finally observations [8] T. Roemer, D.J. Krysan, Antifungal drug development: challenges, unmet
made usingthe HRTEM (FEI Technai G2 F20, 120 kV) [31,65], for clinical needs, and new approaches, CSH Perspect. Med. 4 (2014), 019703.
[9] L. Ostrosky-Zeichner, A. Casadevall, J.N. Galgiani, F.C. Odds, J.H. Rex, An insight
detailed test procedure please see SI.
into the antifungal pipeline: selected new molecules and beyond, Nat. Rev.
Drug Discov. 9 (2010) 719e727.
Declaration of competing interest [10] D.W. Denning, M.J. Bromley, How to bolster the antifungal pipeline, Science
347 (2015) 1414e1416.
[11] C.D. Fjell, J.A. Hiss, R.E. Hancock, G. Schneider, Designing antimicrobial pep-
The authors declare that they have no known competing tides: form follows function, Nat. Rev. Drug Discov. 11 (2011) 37e51.
financial interests or personal relationships that could have [12] J. Cruz, C. Ortiz, F. Guzman, R. Fern andez-Lafuente, R. Torres, Antimicrobial
appeared to influence the work reported in this paper. peptides: promising compounds against pathogenic microorganisms, Curr.
Med. Chem. 21 (2014) 2299e2321.
[13] D.A. Phoenix, S.R. Dennison, F. Harris, Cationic antimicrobial peptides, in:
Acknowledgements Antimicrobial Peptides, Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim,
Germany, 2013, pp. 39e81.
[14] J.P.S. Powers, R.E.W. Hancock, The relationship between peptide structure and
K.K.S. thanks the University Grant Commission (UGC), New antibacterial activity, Peptides 24 (2003) 1681e1691.
Delhi (India) for the award of senior research fellowship (UGC-NET- [15] K.L. Brown, R.E. Hancock, Cationic host defense (antimicrobial) peptides, Curr.
SRF). Opin. Immunol. 18 (2006) 24e30.
[16] M. Wenzel, A.I. Chiriac, A. Otto, D. Zweytick, C. May, C. Schumacher, R. Gust,
H.B. Albada, M. Penkova, U. Kra €mer, R. Erdmann, Small cationic antimicrobial
Abbreviations peptides delocalize peripheral membrane proteins, Proc. Natl. Acad. Sci. U.S.A.
111 (2014) E1409eE1418.
[17] K.A. Brogden, Antimicrobial peptides: pore formers or metabolic inhibitors in
CAMP cationic antimicrobial peptide
bacteria? Nat. Rev. Microbiol. 3 (2005) 238e250.
AFP antifungal peptides [18] R. Garcia-Rubio, H.C. de Oliveira, J. Rivera, N. Trevijano-Contador, The fungal
DIEA N,N-diisopropylethylamine cell wall: Candida, cryptococcus, and Aspergillus species, Front. Microbiol. 10
CDI 1,10 -Carbonyldiimidazole (2020) 2993.
[19] T.R. O'Meara, J.A. Alspaugh, The Cryptococcus neoformans capsule: a sword
DIC N,N0 -diisopropylcarbodiimide and a shield, Clin. Microbiol. Rev. 25 (2012) 387e408.
HOBt 1-Hydroxybenzotriazole [20] J.D. Steckbeck, B. Deslouches, R.C. Montelaro, Antimicrobial peptides: new

15
K.K. Sharma, R. Ravi, I.K. Maurya et al. European Journal of Medicinal Chemistry 223 (2021) 113635

drugs for bad bugs? Expet Opin. Biol. Ther. 14 (2014) 11e14. [42] K.K. Sharma, M. Mandloi, R. Jain, Regioselective copper-catalyzed N(1)-(het-
[21] W.M. Hewitt, S.S. Leung, C.R. Pye, A.R. Ponkey, M. Bednarek, M.P. Jacobson, ero) arylation of protected histidine, Org. Biomol. Chem. 14 (2016)
R.S. Lokey, Cell-permeable cyclic peptides from synthetic libraries inspired by 8937e8941.
natural products, J. Am. Chem. Soc. 137 (2015) 715e721. [43] K.K. Sharma, M. Mandloi, R. Jain, Regioselective access to 1, 2-diaryl histidines
[22] G. Wang, B. Mishra, K. Lau, T. Lushnikova, R. Golla, X. Wang, Antimicrobial via copper-catalyzed N(1)-arylation of 2-aryl histidines, Eur. J. Org Chem.
peptides in 2014, Pharmaceuticals 8 (2015) 123e150. (2017) 984e988.
[23] J. Zhu, P.W. Luther, Q. Leng, A.J. Mixson, Synthetic histidine-rich peptides [44] M. Nichols, M. Kuljanin, M. Nategholeslam, T. Hoang, S. Vafaei, B. Tomberli,
inhibit Candida species and other fungi in vitro: role of endocytosis and C.G. Gray, L. DeBruin, M. Jelokhani-Niaraki, Dynamic turn conformation of a
treatment implications, Antimicrob. Agents Chemother. 50 (2006) short tryptophan-rich cationic antimicrobial peptide and its interaction with
2797e2805. phospholipid membranes, J. Phys. Chem. B 117 (2013) 14697e14708.
[24] M.B. Strøm, B.E. Haug, M.L. Skar, W. Stensen, T. Stiberg, J.S. Svendsen, The [45] R.K. Sharma, R. Jain, Unprecedented 1, 10 -carbonyldiimidazole-mediated
pharmacophore of short cationic antibacterial peptides, J. Med. Chem. 46 amidation of unprotected a-amino acids in water, Synlett 4 (2007) 603e606.
(2003) 1567e1570. [46] A. Mahindra, K.K. Sharma, R. Jain, Rapid microwave-assisted solution-phase
[25] A. Matejuk, Q. Leng, M.D. Begum, M.C. Woodle, P. Scaria, S.T. Chou, A.J. Mixson, peptide synthesis, Tetrahedron Lett. 53 (2012) 6931e6935.
Peptide-based antifungal therapies against emerging infections, Drugs Future [47] A. Mahindra, K. Nooney, S. Uraon, K.K. Sharma, R. Jain, Microwave-assisted
35 (2010) 197e232. solution phase peptide synthesis in neat water, RSC Adv. 3 (2013)
[26] C. Lawyer, S. Pai, M. Watabe, P. Borgia, T. Mashimo, L. Eagleton, K. Watabe, 16810e16816.
Antimicrobial activity of a 13 amino acid tryptophan-rich peptide derived [48] Reference Method for Broth Dilution Antifungal Susceptibility Testing of
from a putative porcine precursor protein of a novel family of antibacterial Yeasts: Approved Standard, second ed., National Committee for Clinical Lab-
peptides, FEBS Lett. 390 (1996) 95e98. oratory Standards, Wayne, PA, 2002. M27-A2.
[27] R.K. Sharma, R.P. Reddy, W. Tegge, R. Jain, Discovery of Trp-His and His-Arg [49] S. Kagan, D. Ickowicz, M. Shmuel, Y. Altschuler, E. Sionov, M. Pitusi, A. Weiss,
analogues as new structural classes of short antimicrobial peptides, J. Med. S. Farber, A.J. Domb, I. Polacheck, Toxicity mechanisms of amphotericin B and
Chem. 52 (2009) 7421e7431. its neutralization by conjugation with arabinogalactan, Antimicrob, Agents
[28] R.K. Sharma, S. Sundriyal, N. Wangoo, W. Tegge, R. Jain, New antimicrobial Chemother 56 (2012) 5603e5611.
hexapeptides: synthesis, antimicrobial activities, cytotoxicity, and mecha- [50] S.B. Levy, B. Marshall, Antibacterial resistance worldwide: causes, challenges
nistic studies, ChemMedChem 5 (2010) 86e95. and responses, Nat. Med. 10 (2004) S122eS129.
[29] A. Mahindra, N. Bagra, N. Wangoo, S.I. Khan, M.R. Jacob, R. Jain, Discovery of [51] C.A. Arias, B.E. Murray, The rise of the Enterococcus: beyond vancomycin
short peptides exhibiting high potency against Cryptococcus neoformans, ACS resistance, Nat. Rev. Microbiol. 10 (2012) 266e278.
Med. Chem. Lett. 5 (2014) 315e320. [52] E. Borenfreund, H. Babich, N. Martin-Alguacil, Rapid chemosensitivity assay
[30] A. Mahindra, K.K. Sharma, D. Rathore, S.I. Khan, M.R. Jacob, R. Jain, Synthesis with human normal and tumor cells in vitro, in Vitro Cell, Dev. Biol. 26 (1990)
and antimicrobial activities of His (2-aryl)-Arg and Trp-His (2-aryl) classes of 1030e1034.
dipeptidomimetics, MedChemComm 5 (2014) 671e676. [53] K. Valko, C. Bevan, D. Reynolds, Chromatographic lipophilicity index by fast-
[31] K.K. Sharma, I.K. Maurya, S.I. Khan, M.R. Jacob, V. Kumar, K. Tikoo, R. Jain, gradient RP-HPLC: a high-throughput alternative to log P/log D, Anal. Chem.
Discovery of a membrane-active, ring-modified histidine containing ultra- 69 (1997) 2022e2029.
short amphiphilic peptide that exhibits potent inhibition of cryptococcus [54] M.R. Eftink, C.A. Ghiron, Fluorescence quenching studies with proteins, Anal.
neoformans, J. Med. Chem. 60 (2017) 6607e6621. Biochem. 114 (1981) 199e227.
[32] K. Shenmar, K.K. Sharma, N. Wangoo, I.K. Maurya, V. Kumar, S.I. Khan, [55] J.H. Morrissey, Morrissey Laboratory Protocol for Preparing Phospholipid
M.R. Jacob, K. Tikoo, R. Jain, Synthesis, stability and mechanistic studies of Vesicles (SUV) by Sonication, 2001 (please see: http://tf7.org/suv.pdf.
potent anticryptococcal hexapeptides, Eur. J. Med. Chem. 132 (2017) (Accessed 11 February 2020).
192e203. [56] E. Kokkoli, A. Mardilovich, A. Wedekind, E.L. Rexeisen, A. Garg, J.A. Craig, Self-
[33] P.A. Raj, M. Edgerton, M.J. Levine, Salivary histatin 5: dependence of sequence, assembly and applications of biomimetic and bioactive peptide-amphiphiles,
chain length, and helical conformation for candidacidal activity, J. Biol. Chem. Soft Matter 2 (2006) 1015e1024.
265 (1990) 3898e3905. [57] L.M. Yin, M.A. Edwards, J. Li, C.M. Yip, C.M. Deber, Roles of lipophilicity and
[34] F.G. Oppenheim, T. Xu, F.M. McMillian, S.M. Levitz, R.D. Diamond, G.D. Offner, charge distribution of cationic antimicrobial peptides in peptide-membrane
R.F. Troxler, Histatins, a novel family of histidine-rich proteins in human pa- interactions, J. Biol. Chem. 287 (2012) 7738e7745.
rotid secretion. Isolation, characterization, primary structure, and fungistatic [58] M. Hughes, S. Debnath, C.W. Knapp, R.V. Ulijn, Antimicrobial properties of
effects on Candida albicans, J. Biol. Chem. 263 (1988) 7472e7477. enzymatically triggered self-assembling aromatic peptide amphiphiles, Bio-
[35] J. Zhu, P.W. Luther, Q. Leng, A.J. Mixson, Synthetic histidine-rich peptides mater, Sci 1 (2013) 1138e1142.
inhibit Candida species and other fungi in vitro: role of endocytosis and [59] H. Sarig, S. Rotem, L. Ziserman, D. Danino, A. Mor, Impact of self-assembly
treatment implications, Antimicrob. Agents Chemother. 50 (2006) properties on antibacterial activity of short acyl-lysine oligomers, Anti-
2797e2805. microb. Agents Chemother. 52 (2008) 4308e4314.
[36] C. Lawyer, S. Pai, M. Watabe, P. Borgia, T. Mashimo, L. Eagleton, K. Watabe, [60] A. Bonucci, E. Caldaroni, E. Balducci, R. Pogni, A spectroscopic study of the
Antimicrobial activity of a 13 amino acid tryptophan-rich peptide derived aggregation state of the human antimicrobial peptide LL-37 in bacterial versus
from a putative porcine precursor protein of a novel family of antibacterial host cell model membranes, Biochemistry 54 (2015) 6760e6768.
peptides, FEBS Lett. 390 (1996) 95e98. [61] Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow
[37] D.J. Schibli, R.F. Epand, H.J. Vogel, R.M. Epand, Tryptophan-rich antimicrobial Aerobically, seventh ed., National Committee for Clinical Laboratory Stan-
peptides: comparative properties and membrane interactions, Biochem. Cell. dards, Wayne, PA, 2006. M7-A7.
Biol. 80 (2002) 667e677. [62] D.H. Kim, D.G. Lee, K.L. Kim, Y. Lee, Internalization of tenecin 3 by a fungal
[38] D.I. Chan, E.J. Prenner, H.J. Vogel, Tryptophan-and arginine-rich antimicrobial cellular process is essential for its fungicidal effect on Candida albicans, Eur. J.
peptides: structures and mechanisms of action, Biochim. Biophys. Acta Bio- Biochem. 268 (2001) 4449e4458.
membr. 1758 (2006) 1184e1202. [63] C. Park, D.G. Lee, Melittin induces apoptotic features in Candida albicans,
[39] L. Gentilucci, R. De Marco, L. Cerisoli, Chemical modifications designed to Biochem. Biophys. Res. Commun. 394 (2010) 170e172.
improve peptide stability: incorporation of non-natural amino acids, pseudo- [64] D. Mares, Electron microscopy of Microsporum cookei after ‘in vitro’ treat-
peptide bonds, and cyclization, Curr. Pharmaceut. Des. 16 (2010) 3185e3203. ment with protoanemonin: a combined SEM and TEM study, Mycopathologia
[40] A. Mahindra, R. Jain, Regiospecific direct C-H arylation at the 2-position of L- 108 (1989) 37e46.
histidine using arylboronic acids, Synlett 23 (2012) 1759e1764. [65] M. Borgers, M.A. Van De Ven, J. Van Cutsem, Structural degeneration of
[41] A. Mahindra, N. Bagra, R. Jain, Palladium-Catalyzed Regioselective C-5 aryla- Aspergillus fumigatus after exposure to saperconazole, Med. Mycol. 27 (1989)
tion of protected L-histidine: microwave-assisted CeH activation adjacent to 381e389.
donor arm, J. Org. Chem. 78 (2013) 10954e10959.

16

You might also like