Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Feb 14.
Published in final edited form as: ACS Infect Dis. 2020 Jan 16;6(2):159–167. doi: 10.1021/acsinfecdis.9b00380

Re-Engineering of Yohimbine’s Biological Activity through Ring Distortion: Identification and Structure–Activity Relationships of a New Class of Antiplasmodial Agents

Nicholas G Paciaroni 1, David L Perry II 2, Verrill M Norwood IV 3, Claribel Murillo-Solano 4, Jennifer Collins 5, Srinivasarao Tenneti 6, Debopam Chakrabarti 7, Robert W Huigens III 8
PMCID: PMC7346850  NIHMSID: NIHMS1600192  PMID: 31913597

Abstract

Select natural products are ideal starting points for ring distortion, or the dramatic altering of inherently complex molecules through short synthetic pathways, to generate an array of novel compounds with diverse skeletal architectures. A major goal of our ring distortion approach is to re-engineer the biological activity of indole alkaloids to identify new compounds with diverse biological activities in areas of significance to human health and medicine. In this study, we re-engineered the biological activity of the indole alkaloid yohimbine through ring rearrangement and ring cleavage synthesis pathways to discover new series of antiplasmodial agents. One new compound, Y7j, was found to demonstrate good potency against chloroquine-resistant Plasmodium falciparum Dd2 cells (EC50 = 0.33 μM) without eliciting cytotoxicity against HepG2 cells (EC50 > 40 μM). Y7j demonstrated stage-specific action against parasites at the late ring/trophozoite stage. A series of analogues was synthesized to gain structure—activity relationship insights, and we learned that both benzyl groups of Y7j are required for activity and fine-tuning of antiplasmodial activities could be accomplished by changing substitution patterns on the benzyl moieties. This study demonstrates the potential for ring distortion to drive new discoveries and change paradigms in chemical biology and drug discovery.

Keywords: yohimbine, malaria, drug discovery, ring distortion, indole alkaloids

Graphical Abstract

graphic file with name nihms-1600192-f0001.jpg


Malaria continues to be a major global health concern. Malaria is a mosquito-borne infectious disease caused by a variety of Plasmodium parasites, with Plasmodium falciparum being the most deadly to humans. In 2017, the World Health Organization (WHO) reported 219 million new cases of malaria, an increase of 2 million from 2016.1 Additionally, malaria-related deaths totaled 435,000, resulting in $12 billion per year with associated healthcare costs. Current clinically used antiplasmodial therapeutics have been developed from drugs that were initially discovered over 30 years ago and primarily belong to one of three drug classes: artemisinins, aminoquinolines, or antifolates.2 To make matters worse, each of these drug classes have garnered resistance from the P. falciparum parasite.3 In several parts of the world where malaria has reached epidemic proportions, artemisinin-based combination therapies (ACTs) used for uncomplicated P. falciparum are showing increasing signs of ineffectiveness.4,5 New antimalarial agents that operate through novel mechanisms are needed to combat widespread resistance to existing drugs.6,7

Natural products have been a primary source of therapeutic agents due to their outstanding ability to bind and modulate numerous biological targets critical to disease.8,9 Complex natural products, such as taxol, morphine, and vancomycin, form highly specific binding modes with their respective protein targets due to their complex and chiral molecular architectures that display an array of functional groups critical for such interactions. Despite the many advances and curative natural products that have been introduced into the clinic, there has been a paradigm shift to the high-throughput screening (HTS) of synthetic compound libraries as the primary approach driving current drug discovery efforts.1012 It is well established that these compound libraries are composed of structurally simple organic molecules, which have served well for drugging certain biological targets (e.g., kinases); however, these compound libraries lack chemical diversity13 and have been implicated in failures to identify viable therapeutic lead compounds for more sophisticated targets (e.g., protein—protein interactions,14 transcription factors15).

Innovative strategies to address chemical diversity concerns by increasing the population of complex small molecules within screening libraries have been pursued. The most established approaches include diversity-oriented synthesis (DOS)1618 and biology-oriented synthesis (BIOS),19,20 which have been pioneered by Schreiber and Waldmann, respectively. These strategies aim at generating complex libraries of small molecules by building structural complexity in subsequent synthetic reactions, starting from simple building blocks, and have unveiled a variety of biological discoveries of therapeutic relevance. Interestingly, Schreiber and co-workers have discovered multiple series of new antimalarial agents using DOS.2123

Complementary approaches to DOS and BIOS have been developed,2427 including Complexity-to-Diversity/ring distortion of available natural products.2835 In this approach, natural products are subjected to an array of chemoselective reactions aimed at ring distortion, or the dramatic altering of inherently complex ring systems of select natural products, to generate architecturally unique scaffolds. A major goal of ring distortion is to identify complex small molecules that display biological activity that is distinct from the parent natural product and is of significance in disease and human health. Ring distortion efforts have been reported for a diversity of natural products, including: gibberellic acid,28 abietic acid,29 pleuromutulin,30,34 quinine,28 sinomenine,31 yohimbine,32 lycorine,33 adrenosterone,28 and other steroids.36

Our laboratory recently reported a tryptoline-enabled ring distortion campaign of yohimbine, a complex indole alkaloid that is commercially available on the decagram scale.32,35 Utilizing the extended tryptoline substructure of yohimbine, we were able to rapidly generate an array of unique, complex, and diverse small molecules through a series of highly selective ring-cleavage and oxidative indole rearrangement reactions. Using this approach, our ring distortion efforts have culminated in a growing library of small molecules, which are being subjected to various biological screens to identify new active scaffolds relevant to important disease areas.32,37 Our work presented here details the identification and structure—activity relationship (SAR) studies regarding a new series of antiplasmodial agents derived from the ring distortion of yohimbine.

RESULTS AND DISCUSSION

An initial library of 70 yohimbine-derived ring-distorted compounds was screened for antiplasmodial activity against a chloroquine-resistant P. falciparum strain (Dd2) in an unbiased, cell-based SYBR Green I assay3840 (Figure 1). From the initial screen, compounds from the Y1 and Y7 series demonstrated inhibitory activity against Dd2 cells (>40% at 2 μM). Validation of active hits and structurally related analogues were subsequently subjected to dose–response experiments to generate comparative EC50 values to determine overall potency and gain initial insights into SAR. Interestingly, all hit compounds arose from two of the ring-distorted scaffolds accessed from yohimbine, including the Y1 series (6 analogues; synthesized in three steps from yohimbine; EC50 = 1.60–5.18 μM against Dd2) and the Y7 series (8 analogues; synthesized in four steps from yohimbine; EC50 values ranging from 0.33 to 2.57 μM against Dd2) (Figure 2).

Figure 1.

Figure 1.

Complex and diverse scaffolds rapidly synthesized from the indole alkaloid yohimbine. Initial hit compounds possessing antiplasmodial activities are highlighted; however, yohimbine was found to demonstrate no antiplasmodial activity. The remainder of the yohimbine-derived scaffolds (Y2–Y6) were found to be inactive against P. falciparum Dd2 cells during initial screens.

Figure 2.

Figure 2.

Antiplasmodial activities of Y1 and Y7 series analogues, which were found to be active during initial screens against Dd2 cells. Testing of related analogues provides initial SAR insights and informs follow-up chemical synthesis investigations.

From the initial screen, dibenzylated spiro-oxindole analogue Y7j proved to be the most potent analogue of the library (EC50 = 0.33 μM, Figure 2) and was also found to have no cytotoxicity against HepG2 cells at 40 μM (highest test concentration), demonstrating a clear selectivity for plasmodial cell targeting (selectivity index >121; determined by [Hep G2, EC50]/[Dd2, EC50]). In addition, the parent indole alkaloid yohimbine was completely inactive against Dd2 cells, EC50 of 76.7 μM, demonstrating a re-engineering of its biological activities that occurred through the dramatic altering of its molecular architecture.

To further investigate the antiplasmodial activity of Y7j, we evaluated the developmental stage-specific action of this small molecule by microscopy and flow cytometry.41,42 Precise delineation of the timing of action of an inhibitor provides valuable insight into the developmental growth and clinical clearance of the parasite. Tightly synchronized parasites were treated at 6 h post-invasion of the merozoites with a 5× EC50 concentration of Y7j (Figure 3A). Microscopic evaluation of Giemsa-stained-thin smears and flow cytometric assessments were done at 12 h intervals. Negative controls represented infected red blood cells exposed to vehicle DMSO (0.1%) only. As can be seen from Figure 3A, compared to untreated cultures, Y7j inhibited the parasite’s late ring/trophozoite stage (18 h post-invasion, HPI).43 The untreated control culture matures as expected through the trophozoite/schizont (30 HPI) and segmenter (42 HPI) and reappears as rings after reinvasion (54 HPI) with a concomitant increase in parasitemia (see flow cytometry data, Figure 3B,C). Although at the trophozoite stage (30 h) the parasite mass increased, there was significant vacuolization noticed in the parasites exposed to Y7j. Interestingly, Y1f also demonstrated stage-specific action at the late ring/trophozoite phase in Dd2 cells and vacuolization, similar to Y7j (see the Supporting Information). These two ring distortion scaffolds, Y1 and Y7, are very different from each other structurally and likely operate through different primary modes of action against plasmodial cells.

Figure 3.

Figure 3.

Characterization of stage-specific activity of Y7j in Dd2 cells. Tightly synchronized Dd2 cultures were treated at 6 h time points with Y7j at 5× EC50. Blood smears and Giemsa staining were also taken at each time point to verify (A) phenotype and (B) flow cytometry. Flow cytometry was performed to quantify the stage of action for Y7j at the time of harvesting, following a 6 HPI treatment. For flow cytometric analysis, collected samples were fixed in 0.04% glutaraldehyde in PBS, permeabilized with 0.25% Triton X-100, treated with RNase (50 μg/mL), and stained with YOYO-1. Data acquisition was performed in a CytoFLEX (Beckman) flow cytometer. (C) Parasitemia of Y7j treated and vehicle untreated cultures. Notes: Dihydroartemisinin (DHA), an inhibitor of the late ring/trophozoite stage, was tested as a positive control alongside Y7j (not shown). HPI, hours post-invasion of red blood cells by merozoites.

To determine if Y7j elicits it’s antiplasmodial activity through a parasitocidal or parasitostatic mechanism, we performed a series of kill kinetic experiments in Dd2 cultures.41,42 We treated asynchronous parasite cultures with 5× EC50 concentration of the compound for different periods of time (6, 12, 24, and 48 h). After washing out the compound at these time points, the growth of the culture was evaluated for 96 h. As is evident from these kill kinetic experiments (Figure 4), the most significant reduction in parasitemia observed following a 24 h treatment with Y7j suggests this compound is a fast-acting parasitocidal agent with rapid clearance of the remaining parasites 48 h post-treatment. Similar to Y7j, significant killing of Dd2 cells was observed following a 24 h treatment of Y1f (see the Supporting Information).

Figure 4.

Figure 4.

Rate of killing and parasitocidal/parasitostatic determination of Y7j in Dd2 cultures. To evaluate the rate of killing for Y7j, asynchronous cultures were treated with 5× EC50 (Y7j concentrations) for (A) 6, (B) 12, (C) 24, and (D) 48 h. After each treatment, cultures were washed three times in RPMI, then resuspended in culture media, and monitored daily for parasite growth. Note: DMSO concentration was 0.15% for these experiments. Dihydroartemisinin (DHA) was used as a positive control for rapid parasite killing (test concentration = 50 nM; 5× EC50) in these experiments.

With the interesting antiplasmodial activity we discovered during the initial investigations, we worked to optimize the synthetic route to Y7j and related analogues from yohimbine (Y). Our goals were to explore the structure–activity relationship profiles related to the Y7 series and, ideally, improve antiplasmodial activities. We were able to utilize our previous protocol32 to scale up the synthesis of Y7a (925 mg) from Y via treatment with N-chlorosuccinimide (NCS) and subsequent addition of sodium methoxide (NaOMe), which afforded 925 mg of Y7a in a single run (76% yield over 2 steps, Figure 5). Y7a proved to be an important intermediate as we strategically used the protected amide to selectively monoalkylate the free hydroxyl group of Y7a using sodium hydride and benzyl bromides to synthesize a focused series of compounds for antiplasmodial investigations, following treatment with aqueous trifluoroacetic acid (analogues Y7aa to Y7ad, average yield = 52% over two steps; Figure 5). In addition, Y7a was also used to generate key intermediate Y7b upon treatment with trifluoroacetic acid (TFA; 96% yield, scaled up to 1.63 g of Y7b in a single run). With both Y7a and Y7b in hand, our goals were to synthesize focused subclasses of analogues to gain detailed SAR information for these new antiplasmodial agents. This chemistry provided synthetic routes to diverse mono- and dibenzylated analogues of Y7j for biological evaluation.

Figure 5.

Figure 5.

Chemical synthesis of spirocyclic antiplasmodial agents (Y7j analogues), general SAR trends, and the top four most potent analogues from these studies are highlighted.

Symmetrically functionalized derivatives of Y7j were synthesized by treating Y7b with sodium hydride, followed by two equivalents of (hetero)benzyl bromides to afford analogues Y7m–Y7v (average yield = 46%; 10 analogues). This allowed for the incorporation of various heterocyclic bioisosteres44 of the phenyl moieties in Y7j, including: pyridines (Y7o, Y7q), pyrazines (Y7t, Y7v), thiophene (Y7p), and furan (Y7r) heterocycles (Figure 5). Complementary diversifications were carried out through mono-N-alkylation of Y7b via treatment with various alkyl bromides and potassium carbonate in N,N-dimethylformamide to afford Y7ae–Y7ai (average yield = 76%; 5 analogues). The free secondary hydroxyl of the resulting amides (Y7ae–Y7ai) was then deprotonated with sodium hydride and alkylated with (hetero)aromatic benzyl bromides to generate mixed functionalized derivatives Y7aj–Y7am (average yield = 71%; 4 analogues).

With new Y7j and Y1f analogues in hand (see the Supporting Information for additional Y1f analogues; Y1i, Y1j), all compounds were subjected to dose–response experiments to determine EC50 values against Dd2 cells. The two new Y1f analogues were found to be inactive (Y1i, Y1j; EC50 > 20 μM), and analogues related to the Y1 series were no longer pursued during these investigations. On the other hand, six new Y7j analogues demonstrated submicromolar antiplasmodial activities with thiophene analogue Y7p (EC50 = 0.32 μM) and 2-chlorobenzyl analogue Y7n (EC50 = 0.52 μM) being the most potent new analogues synthesized against Dd2 cells (Figure 5 and Table 1).

Table 1.

Summary of Antiplasmodial Activities and Cytotoxicity against HepG2 (Liver) Cells for Y and Select Y1 and Y7 Analoguesa

Y code EC50 Dd2 cells (μM) EC50 3D7 cells (μM) EC50 HepG2 cells (μM) selectivity index (SI) stage-specific activity
Y 76.7 ± 0.74 - >100 - n.a.
Y1c 2.58 ± 0.07 - - - -
Y1e 2.98 ± 0.20 - - - -
Y1f 1.60 ± 0.16 - 18.9 - A
Y1g 3.18 ± 0.11 - - - -
Y1h 3.47 ± 0.11 - - - -
Y7e 1.50 ± 0.09 - - - -
Y7f 1.41 ± 0.23 - - - -
Y7g 0.79 ± 0.06 0.74 ± 0.07 >40 >51 A
Y7h 1.05 ± 0.08 - - - -
Y7i 0.85 ± 0.05 - - - -
Y7j 0.33 ± 0.03 0.35 ± 0.03 >40 >121 A
Y7k 0.64 ± 0.07 0.68 ± 0.07 - - -
Y7l 2.57 ± 0.05 - - - -
Y7m 1.51 ± 0.06 - - - -
Y7n 0.52 ± 0.04 0.56 ± 0.04 >40 >77 -
Y7o 3.51 ± 0.32 - - - -
Y7p 0.32 ± 0.02 0.49 ± 0.05 >40 >125 A
Y7q 0.87 ± 0.04 0.91 ± 0.03 >40 >46 -
Y7r 0.65 ± 0.07 0.68 ± 0.09 >40 >62 -
Y7s 1.32 ± 0.07 1.13 ± 0.09 - - -
Y7t 3.08 ± 0.20 4.31 ± 0.16 - - -
Y7u 0.89 ± 0.05 0.80 ± 0.09 >40 >45 -
Y7aj 2.38 ± 0.24 - - - -
Y7ak 0.94 ± 0.10 - - - -
Y7am 2.71 ± 0.07 3.00 ± 0.13 - - -
a

All concentrations are reported in micromolar (μM). Notes: “A” = stage-specific activity at the late ring/trophozoite phase of the asexual blood stage of P. falciparum. “n.a.” = yohimbine (Y), which is inactive against P. falciparum. “-” = noted for analogues that were not tested for stage-specific activity. Selectivity index (SI) was generated according to EC50 against HepG2 cells divided by EC50 against Dd2 cells. All values reported in this table resulted from three independent experiments.

From these studies, an interesting structure–activity relationship profile was revealed for the new antiplasmodial agent Y7j and related analogues. The most prominent structural feature regarding antiplasmodial activity of Y7j is the requirement of both 2-chloro-4-methoxybenzyl groups, as removal of either group led to a complete loss in antiplasmodial activity (e.g., Y7ab, EC50 > 20 μM; Figure 6). This SAR information could not have been obtained without the new synthetic efforts described in Figure 4, which allowed for a complementary series of monobenzylated analogues for biological investigations. In total, seven monoalkylated/benzylated analogues of Y7j (containing either a free amide or a free hydroxyl group) were investigated during these studies, and each of these analogues reported an EC50 > 20 μM (e.g., Y7aa and Y7ae). In addition, subtle structural changes were made to Y7j through alkylation of Y7b (Figure 5) with alternative benzyl and heterobenzyl starting materials. Removal of the methoxy groups at the 4-position of both benzyl moieties of Y7j resulted in a 1.6-fold loss in antiplasmodial activity against Dd2 cells (Y7n, EC50 = 0.52 μM), while removal of the 2-chlorine atom of Y7j resulted in a 4.5-fold loss in antiplasmodial activity (Y7m, EC50 = 1.51 μM).

Figure 6.

Figure 6.

Detailed structure-activity relationship profiles regarding the antiplasmodial activities of Y7j.

Additional substitutions with six-membered heterocycles containing nitrogen also resulted in losses in activity compared to Y7j (see Figure 6 for detailed SAR profiling). Interestingly, the only new compound to demonstrate equipotent activity against Dd2 cells compared to Y7j was thiophene analogue Y7p (EC50 = 0.32 μM; demonstrated stage-specific activity at the late ring/trophozoite phase of asexual blood stage against Dd2 cells, similar to Y7j; Table 1), as previously mentioned. In addition to SAR, seven Y7 analogues were counter-screened in dose–response experiments against wild-type 3D7 P. falciparum cells, and each analogue demonstrated near equipotent activity when comparing EC50 values to those generated against chloroquine-resistant Dd2 cells (Table 1). Additionally, the same seven Y7 analogues demonstrated no cytotoxicity against HepG2 cells (EC50 > 40 μM; selectivity indexes >45 to >125; Table 1). These combined analyses point to a clear SAR, and we hypothesize that the structural requirements for active analogues related to Y7j result from such analogues interacting with a well-defined biological target. On the basis of our SAR profile, we predict these agents to elicit their activities against a new antiplasmodial target. Additional mechanistic studies are required to substantiate this hypothesis and are currently being pursued.

In conclusion, we identified two diverse and complex scaffolds that possess antiplasmodial activities from ring distortion efforts of the indole alkaloid yohimbine. One new antiplasmodial compound, Y7j, was found to demonstrate good potency against chloroquine-resistant P. falciparum Dd2 cells (EC50 = 0.33 μM) without eliciting cytotoxicity against HepG2 cells (EC50 > 40 μM; selectivity index >121). In addition, Y7j demonstrated stage-specific action against P. falciparum at the late ring/trophozoite stage, preventing an increase in parasitemia as determined by flow cytometry, and rapid parasitocidal activity against Dd2 cultures following 24 h of treatment. A series of focused analogues were also synthesized to gain detailed structure–activity relationship insights, and we learned that both benzyl groups of Y7j are required for activity and that fine-tuning of antiplasmodial activities could be accomplished by changing substitution patterns on the benzyl moieties; however, the only new analogue to demonstrate equipotent activity to Y7j was thiophene bioisostere Y7p. Interestingly, the parent indole alkaloid yohimbine demonstrated no antiplasmodial activities, and our active scaffolds are the result of a re-engineering of yohimbine’s biological activity. This work demonstrates the potential for the ring distortion of indole alkaloids to drive new discoveries in chemical biology and drug discovery.

METHODS

Please see the Supporting Information for the details of the experimental methods and chemical synthesis protocols.

Supplementary Material

Yohimbine Re-engineering to Antiplasmodial Agents SI

ACKNOWLEDGMENTS

We acknowledge the University of Florida and the National Institute of General Medical Sciences of the National Institutes of Health for providing support for this work (R35GM128621 to R.W.H.). High-resolution mass spectrometry (HRMS) was performed for all new compounds synthesized at the Mass Spectrometry Research and Education Center in the Chemistry Department at the University of Florida.

ABBREVIATIONS

3D7

chloroquine-sensitive P. falciuarum strain

ACTs

artemisinin-based combination therapies

BIOS

biology-oriented synthesis

Dd2

chloroquine-resistant P. falciuarum strain

DHA

dihydroartemisinin

DMF

dimethylformamide

DMSO

dimethyl sulfoxide

DOS

diversity-oriented synthesis

EC50

concentration of a compound that gives half-maximal response

equiv

equivalents

HepG2

a human liver cancer cell line

HPI

hour post-invasion

HTS

high-throughput screening

K2CO3

potassium carbonate

mg

milligrams

NaH

sodium hydride

NaOMe

sodium methoxide

NCS

N-chlorosuccinimide

nM

nanomolar

SAR

structure–activity relationship

SI

selectivity index

TFA

trifluoroacetic acid

THF

tetrahydrofuran

μM

micromolar

WHO

World Health Organization

Footnotes

The authors declare no competing financial interest.

Supporting Information

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.9b00380.

All experimental methods, chemical synthesis protocols, dose–response curves for select yohimbine-derived antiplasmodial agents in chloroquine-resistant Dd2 cells (Supporting Figure 1), dose–response curves for select yohimbine-derived antiplasmodial agents in chloroquine-sensitive (3D7) cells (Supporting Figure 2), additional kill kinetic data, stage-specific activity for Y1f, full characterization data (with tabulations of NMR spectra), 1H and 13C NMR spectra for all new compounds, and 2-D NMR spectra for select derivatives (PDF)

Contributor Information

Nicholas G. Paciaroni, University of Florida, Gainesville, Florida.

David L. Perry, II, University of Central Florida, Orlando, Florida.

Verrill M. Norwood, IV, University of Florida, Gainesville, Florida.

Claribel Murillo-Solano, University of Central Florida, Orlando, Florida.

Jennifer Collins, University of Central Florida, Orlando, Florida.

Srinivasarao Tenneti, University of Florida, Gainesville, Florida.

Debopam Chakrabarti, University of Central Florida, Orlando, Florida.

Robert W. Huigens, III, University of Florida, Gainesville, Florida.

REFERENCES

  • (1).World Health Organization. (accessed September 30, 2019) World Malaria Report; https://www.who.int/en/news-room/fact-sheets/detail/malaria.
  • (2).Grimberg BT, and Mehlotra RK (2011) Expanding the antimalarial drug arsenal – Now, but how? Pharmaceuticals 4, 681–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (3).Escalante AA, Smith DL, and Kim Y (2009) The dynamics of mutations associated with anti-malarial drug resistance in Plasmodium falciparum. Trends Parasitol. 25, 557–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (4).Cui W (2011) WHO urges the phasing out of artemisinin based monotherapy for malaria to reduce resistance. BMJ. 342, d2793. [DOI] [PubMed] [Google Scholar]
  • (5).Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, Sreng S, Anderson JM, Mao S, Sam B, Sopha C, Chuor CM, Nguon C, Sovannaroth S, Pukrittayakamee S, Jittamala P, Chotivanich K, Chutasmit K, Suchatsoonthorn C, Runcharoen R, Hien TT, Thuy-Nhien NT, Thanh NV, Phu NH, Htut Y, Han KT, Aye KH, Mokuolu OA, Olaosebikan RR, Folaranmi OO, Mayxay M, Khanthavong M, Hongvanthong B, Newton PN, Onyamboko MA, Fanello CI, Tshefu AK, Mishra N, Valecha N, Phyo AP, Nosten F, Yi P, Tripura R, Borrmann S, Bashraheil M, Peshu J, Faiz MA, Ghose A, Hossain MA, Samad R, Rahman MR, Hasan MM, Islam A, Miotto O, Amato R, MacInnis B, Stalker J, Kwiatkowski DP, Bozdech Z, Jeeyapant A, Cheah PY, Sakulthaew T, Chalk J, Intharabut B, Silamut K, Lee SJ, Vihokhern B, Kunasol C, Imwong M, Tarning J, Taylor WJ, Yeung S, Woodrow CJ, Flegg JA, Das D, Smith J, Venkatesan M, Plowe CV, Stepniewska K, Guerin PJ, Dondorp AM, Day NP, and White NJ (2014) Tracking Resistance to Artemisinin Collaboration (TRAC). Spread of artemisinin resistance in Plasmodium falciparum malaria. N. Engl. J. Med. 371, 411–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (6).Flannery EL, Chatterjee AK, and Winzeler EA (2013) Antimalarial drug discovery – approaches and progress towards new medicines. Nat. Rev. Microbiol. 11, 849–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (7).Burrows JN, Duparc S, Gutteridge WE, Hooft van Huijsduijnen R, Kaszubska W, Macintyre F, Mazzuri S, Möhrle JJ, and Wells TNC (2017) New developments in anti-malarial target candidate and product profiles. Malar. J. 16, 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (8).Newman DJ, and Cragg GM (2016) Natural products as sources of new drugs from 1981 to 2014. J. Nat. Prod. 79, 629–661. [DOI] [PubMed] [Google Scholar]
  • (9).Böttcher T, Pitscheider M, and Sieber SA (2010) Natural products and their biological targets: proteomic and metabolomics labelling strategies. Angew. Chem., Int. Ed, 49, 2680–2698. [DOI] [PubMed] [Google Scholar]
  • (10).Swinney DC, and Anthony J (2011) How were new medicines discovered? Nat. Rev. Drug Discovery 10, 507–519. [DOI] [PubMed] [Google Scholar]
  • (11).Macarron R, Banks MN, Bojanic D, Burns DJ, Cirovic DA, Garyantes T, Green DV, Hertzberg RP, Janzen WP, Paslay JW, Schopfer U, and Sittampalam GS (2011) Impact of high-throughput screening on biomedical research. Nat. Rev. Drug Discovery 10, 188–195. [DOI] [PubMed] [Google Scholar]
  • (12).Hughes JP, Rees S, Kalindjian SB, and Philpott KL (2011) Principles of early drug discovery. Br. J. Pharmacol. 162, 1239–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (13).Payne DJ, Gwynn MN, Holmes DJ, and Pompliano DL (2007) Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat. Rev. Drug Discovery 6, 29–40. [DOI] [PubMed] [Google Scholar]
  • (14).Dömling A (2008) Small molecular weight protein-protein interaction antagonists: an insurmountable challenge? Curr. Opin. Chem. Biol. 12, 281–291. [DOI] [PubMed] [Google Scholar]
  • (15).Grivas PD, and Papavassiliou AG (2013) Transcriptional corepressors in cancer: emerging targets for therapeutic intervention. Cancer 119, 1120–1128. [DOI] [PubMed] [Google Scholar]
  • (16).Schreiber SL (2009) Organic Chemistry: Molecular Diversity by Design. Nature 457, 153–154. [DOI] [PubMed] [Google Scholar]
  • (17).Galloway WRJD, Isidro-Llobet A, and Spring DR (2010) Diversity-oriented synthesis as a tool for the discovery of novel biologically active small molecules. Nat. Commun. 1, 1–13. [DOI] [PubMed] [Google Scholar]
  • (18).Shang S, and Tan DS (2005) Advancing chemistry and biology through diversity-oriented synthesis of natural product-like libraries. Curr. Opin. Chem. Biol. 9, 248–258. [DOI] [PubMed] [Google Scholar]
  • (19).Wetzel S, Bon RS, Kumar K, and Waldmann H (2011) Biology-Oriented Synthesis. Angew. Chem., Int. Ed. 50, 10800–10826. [DOI] [PubMed] [Google Scholar]
  • (20).van Hattum H, and Waldmann H (2014) Biology-oriented synthesis: harnessing the power of evolution. J. Am. Chem. Soc. 136, 11853–11859. [DOI] [PubMed] [Google Scholar]
  • (21).Heidebrecht RW, Mulrooney C, Austin CP, Barker RH, Beaudoin JA, Cheng KC, Comer E, Dandapani S, Dick J, Duvall JR, Ekland EH, Fidock DA, Fitzgerald ME, Foley M, Guha R, Hinkson P, Kramer M, Lukens AK, Masi D, Marcaurelle LA, Su XZ, Thomas CJ, Weïwer M, Wiegand RC, Wirth D, Xia M, Yuan J, Zhao J, Palmer M, Munoz B, and Schreiber SL (2012) Diversity-oriented synthesis yields a novel lead for the treatment of malaria. ACS Med. Chem. Lett. 3, 112–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (22).Comer E, Beaudoin JA, Kato N, Fitzgerald ME, Heidebrecht RW, Lee M. d., Masi D, Mercier M, Mulrooney C, Muncipinto G, Rowley A, Crespo-Llado K, Serrano AE, Lukens AK, Wiegand RC, Wirth DF, Palmer MA, Foley MA, Munoz B, Scherer CA, Duvall JR, and Schreiber SL (2014) Diversity-oriented synthesis-facilitated medicinal chemistry: toward the development of novel antimalarial agents. J. Med. Chem. 57, 8496–8502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (23).Kato N, Comer E, Sakata-Kato T, Sharma A, Sharma M, Maetani M, Bastien J, Brancucci NM, Bittker JA, Corey V, Clarke D, Derbyshire ER, Dornan GL, Duffy S, Eckley S, Itoe MA, Koolen KMJ, Lewis TA, Lui PS, Lukens AK, Lund E, March S, Meibalan E, Meier BC, McPhail JA, Mitasev B, Moss EL, Sayes M, Van Gessel Y, Wawer MJ, Yoshinaga T, Zeeman A-M, Avery VM, Bhatia SN, Burke JE, Catteruccia F, Clardy JC, Clemons PA, Dechering KJ, Duvall JR, Foley MA, Gusovsky F, Kocken CHM, Marti M, Morningstar ML, Munoz B, Neafsey DE, Sharma A, Winzeler EA, Wirth DF, Scherer CA, and Schreiber SL (2016) Diversity-oriented synthesis yields novel multistage antimalarial inhibitors. Nature 538, 344–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (24).Balthaser BR, Maloney MC, Beeler AB, Porco JA Jr,, and Snyder JK. (2011) Remodelling of the natural product fumagillol employing a reaction discovery approach. Nat. Chem. 3, 969–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (25).Aquino C, Sarkar M, Chalmers MJ, Mendes K, Kodadek T, and Micalizio G (2012) A Biomimetic polyketide-inspired approach to small molecule ligand discovery. Nat. Chem. 4, 99–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (26).McLeod MC, Singh G, Plampin JN III, Rane D, Wang JL, Day VW, and Aubé J (2014) Probing chemical space with alkaloid-inspired libraries. Nat. Chem. 6, 133–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (27).Usanov DL, Chan AI, Maianti JP, and Liu DR (2018) Second-generation DNA-templated macrocycle libraries for the discovery of bioactive small molecules. Nat. Chem. 10, 704–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (28).Huigens RW III, Morrison KC, Hicklin RW, Flood TA, Richter MF, and Hergenrother PJ (2013) A ring-distortion strategy to construct stereochemically complex and structurally diverse compounds from natural products. Nat. Chem. 5, 195–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (29).Rafferty RJ, Hicklin RW, Maloof KA, and Hergenrother PJ (2014) Synthesis of complex and diverse compounds through ring distortion of abietic acid. Angew. Chem., Int. Ed. 53, 220–224. [DOI] [PubMed] [Google Scholar]
  • (30).Hicklin RW, Lopez Silvia TL, and Hergenrother PJ (2014) Synthesis of bridged oxafenestranes from pleuromutilin. Angew. Chem., Int. Ed. 53, 9880–9883. [DOI] [PubMed] [Google Scholar]
  • (31).Garcia A, Drown BS, and Hergenrother PJ (2016) Access to a structurally complex compound collection via ring distortion of the alkaloid sinomenine. Org. Lett. 18, 4852–4855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (32).Paciaroni NG, Ratnayake R, Matthews JH, Norwood VM IV, Arnold AC, Dang LH, Luesch H, and Huigens RW III (2017) A tryptoline ring-distortion strategy leads to complex and diverse biologically active molecules from the indole alkaloid yohimbine. Chem. - Eur. J. 23, 4327–4335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (33).Tasker SZ, Cowfer AE, and Hergenrother PJ (2018) Preparation of structurally diverse compounds from the natural product lycorine. Org. Lett. 20, 5894–5898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (34).Llabani E, Hicklin RW, Lee H-Y, Motika SE, Crawford LA, Weerapana E, and Hergenrother PJ (2019) Diverse compounds from pleuromutilin lead to a thioredoxin inhibitor and inducer of ferroptosis. Nat. Chem. 11, 521–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (35).Norwood VM IV, and Huigens RW III (2019) Harnessing the Chemistry of the Indole Heterocycle to Drive Discoveries in Biology and Medicine. ChemBioChem 20, 2273–2297. [DOI] [PubMed] [Google Scholar]
  • (36).Charaschanya M, and Aubé J (2018) Reagent-controlled regiodivergent ring expansions of steroids. Nat. Commun. 9, 934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (37).Huigens RW III, and Paciaroni NG (2017) Preparation of yohimbine analogues for the treatment of diseases. PCT Int. Appl. WO 2017190038 A1 20171102.
  • (38).Smilkstein M, Sriwilaijaroen N, Kelly JX, Wilairat P, and Riscoe M (2004) Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screening. Antimicrob. Agents Chemother. 48, 1803–1806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (39).Bennett TN, Paguio M, Gligorijevic B, Seudieu C, Kosar AD, Davidson E, and Roepe PD (2004) Novel, rapid, and inexpensive cell-based quantification of antimalarial drug efficacy. Antimicrob. Agents Chemother. 48, 1807–1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (40).Johnson JD, Dennull RA, Gerena L, Lopez-Sanchez M, Roncal NE, and Waters NC (2007) Assessment and continued validation of the malaria SYBR green I-based fluorescence assay for use in malaria drug screening. Antimicrob. Agents Chemother. 51, 1926–1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (41).Roberts BF, Iyamu ID, Lee S, Lee E, Ayong L, Kyle DE, Yuan Y, Manetsch R, and Chakrabarti D (2016) Spirocyclic chromanes exhibit antiplasmodial activities and inhibit all intraerythrocytic life cycle stages. Int. J. Parasitol.: Drugs Drug Resist. 6, 85–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (42).Roberts BF, Zheng Y, Cleaveleand J, Lee S, Lee E, Ayong L, Yuan Y, and Chakrabarti D (2017) 4-Nitro styrylquino-line is an antimalarial inhibiting multiple stages of Plasmodium falciparum asexual life cycle. Int. J. Parasitol.: Drugs Drug Resist. 7, 120–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (43).Bozdech Z, Zhu J, Joachimiak MP, Cohen FE, Pulliam B, and DeRisi JL (2003) Expression profiling of the schizont and trophozoite stages of Plasmodium falciparum with a long-oligonucleotide microarray. Genome Biol. 4, R9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (44).Meanwell NA (2011) Synopsis of Some Recent Tactical Application of Bioisosteres in Drug Design. J. Med. Chem. 54, 2529–2591. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Yohimbine Re-engineering to Antiplasmodial Agents SI

RESOURCES