Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Antroquinonol A: Scalable Synthesis and Preclinical Biology of a Phase 2 Drug Candidate
My Activity
  • Open Access
Research Article

Antroquinonol A: Scalable Synthesis and Preclinical Biology of a Phase 2 Drug Candidate
Click to copy article linkArticle link copied!

View Author Information
Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
§ Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
*E-mail: [email protected] (T.G.M.D.).
*E-mail: [email protected] (P.S.B.).
Open PDFSupporting Information (3)

ACS Central Science

Cite this: ACS Cent. Sci. 2016, 2, 1, 27–31
Click to copy citationCitation copied!
https://doi.org/10.1021/acscentsci.5b00345
Published December 23, 2015

Copyright © 2015 American Chemical Society. This publication is licensed under these Terms of Use.

Abstract

Click to copy section linkSection link copied!

The fungal-derived Taiwanese natural product antroquinonol A has attracted both academic and commercial interest due to its reported exciting biological properties. This reduced quinone is currently in phase II trials (USA and Taiwan) for the treatment of non-small-cell lung carcinoma (NSCLC) and was recently granted orphan drug status by the FDA for the treatment of pancreatic cancer and acute myeloid leukemia. Pending successful completion of human clinical trials, antroquinonol is expected to be commercialized under the trade name Hocena. A synthesis-enabled biological re-examination of this promising natural product, however, reveals minimal in vitro and in vivo antitumor activity in preclinical models.

Copyright © 2015 American Chemical Society

Antroquinonol A (1, Figure 1A) is a quinone-containing natural product reported to have remarkable medicinal potential in the areas of oncology, immunology, and even diabetes. (1) More than ten publications have delineated the exciting activities of 1, and as a consequence, an investigational new drug (IND) application was filed by Golden Biotech Corp. in 2010. (2) It is currently in phase II clinical trials (in the USA and Taiwan) (3) for the treatment of non-small-cell lung cancer (NSCLC; phase II began in January 2014) (3) and has been granted orphan drug status by the FDA for the treatment of pancreatic cancer and acute myeloid leukemia. Given the excitement surrounding this natural product (4) coupled with its relative structural simplicity, an effort was launched to pursue its synthesis and biological evaluation as part of the academic–industrial symbiosis between Scripps and Bristol-Myers Squibb (BMS). (5) In this communication, an enantioselective, scalable, and modular synthesis of 1 is reported. Access to copious amounts of pure 1 enabled a detailed biological reevaluation that is in contrast to the reported preclinical efficacy of the compound in a nude mouse model harboring human hepatoma xenografts and may temper the enthusiasm surrounding this natural product.

Figure 1

Figure 1. Graphical summary of retrosynthetic analyses explored for the synthesis of antroquinonol A (1).

At the outset of this project, no synthesis of 1 existed and its isolation from the rare Taiwanese fungus Antrodia camphorata was not a practical means for studying pure antroquinonol at BMS. (6) A set of plans, ranging in level of ambition and precedent, were evaluated (Figure 1) before settling on the substituted quinone 10 as our final starting point. Six representative blueprints are illustrated and can be divided conceptually into approaches that build the hydroxy enone of 1 via cyclization strategies or via the semireduction of a quinone system. Among the ring-building approaches, Danheiser-type annulation (2), (7) stepwise conjugate addition/annulation (3 + 4), (8) and Diels–Alder (5 + 6) (9) strategies were all pursued, leading either to lengthy sequences, low-yielding key steps, or unstable starting materials (4 and 6 rapidly isomerize). Inspired by the related structure of Coenzyme Q3 (7), extensive efforts to controllably reduce such systems were explored, to no avail. (10) Attempted allylic alcohol isomerization (11) by utilizing 1,4-diol 8 was not possible due to the inherent instability of such systems (spontaneous aromatization). Finally, efforts centering on a tandem 1,2-/1,4-addition (12) to silyl-dienone 9 (13) failed due to the tendency of silyl lithium reagents to reduce quinones rather than add to them. Collectively, these failures led us to the simplest possible approach: a conjugate addition to a substituted quinone. Reimagining this quinone as quinone–monoketal 10 gives the starting material “directionality”, allowing selective introduction of the nucleophile and electrophile through a Michael addition and subsequent 1,2-alkylation. (14) This strategy inherently makes the synthesis modular, allowing a host of nucleophiles and electrophiles to be quickly appended to any desired quinone starting material.

Scheme 1 depicts the fully optimized six-step, enantioselective, scalable synthesis of antroquinonol A (1). Each step was meticulously studied (see Supporting Information for tables of screened conditions), and as a result, gram quantities of 1 were generated for biological screening. The synthesis commences with the formation of quinon–monoketal 12 from commercial benzaldehyde 11. Baeyer–Villiger and dearomative oxidations followed by a trans-ketalization gave the desired quinone–monoacetal in an overall yield of 64%. (15) Extensive screening of the identity of the ketal protecting group (see Scheme 1B) and also of enantioselective conjugate addition conditions (16) led to a one-pot procedure that produced the vicinally difunctionalized product 14 with moderate yield and high stereoselectivity. Conjugate addition product 13 was also prepared in order to confirm absolute stereochemistry via X-ray crystallography. L-Selectride proved best in producing the 4,5-cis stereochemistry of the natural product with a dr of 3:1. Hydrolysis turned out to be a significant challenge, with most acidic conditions leading to a complex mixture of elimination products. Montmorillonite K10 clay was a sufficiently mild proton source for hydrolysis of ketal 14, producing enantioenriched (+)-antroquinonol A (1) in an overall yield of 13% and with 96% ee. Over one gram of the natural product has been prepared to date.

Scheme 1

Scheme 1. Total Synthesis of (+)-Antroquinonol A (1)a

Scheme aReagents and conditions: (a) H2O2 (1.0 equiv), H2SO4 (0.1 equiv), MeOH, 0 °C, 2 h (98%); (b) PIDA (2.0 equiv), MeOH, 23 °C, (89%); (c) 2,2-dimethyl-1,3-propanediol (2.5 equiv), PPTS (0.15 equiv), toluene, 60 °C, 1 h (75%); (d) Cu(OTf)2 (0.05 equiv), phosphoramidite ligand SI2 (0.1 equiv), Me2Zn (2.5 equiv), toluene, −30 °C, 18 h; LiHMDS (1.5 equiv), HMPA (2.5 equiv), farnesyl bromide (2.0 equiv), 0 °C, 3 h; (e) L-Selectride (2.5 equiv), toluene, −78 °C to −20 °C, 6 h; (f) Montmorillonite K10 Clay (30 wt %), DCM, 23 °C, 2 h. PIDA = (diacetoxyiodo)benzene, PPTS = pyridinium p-toluenesulfonate, LiHMDS = lithium bis(trimethylsilyl)amide, HMPA = hexamethylphosphoramide, L-Selectride = lithium tri-sec-butylborohydride solution, DCM = dichloromethane.

With synthetic (+)-1 in hand, we began testing the compound against a panel of human tumor cell lines in vitro, previously reported in the literature for the natural product. As outlined in Table 1, natural antroquinonol (1) is reported to have low micromolar activity against the MDA-MB-231 breast, HepG2 hepatocellular, and LNCaP prostate tumor cell lines and low nanomolar activity versus the Hep 3B hepatocellular carcinoma (HCC) cell line. (1, 17) The reported IC50 value in the lung tumor cell line (A549) for the natural product is 25 μM. (2c) In our hands, 1 is ∼3–70-fold less cytotoxic against these cell lines except for the A549 cell line, where it is ∼4-fold more potent than previously reported. The activity of 1 in the Hep 3B cell line was particularly striking, where it is significantly less potent when compared to the natural product. To be thorough, the enantiomer of 1, (−)-antroquinonol A, was tested in a similar panel, and, much like the natural product, high micromolar activity was seen for the Hep 3B and A549 cell lines (Table S6). The observed and reported high micromolar activity of 1 against a NSCLC cell line (A549) is somewhat puzzling since the natural product is in phase II trials for the treatment of this tumor type. It was indicated in the package submitted to the FDA for phase I studies that “In vivo study in NOD/SCID mice with A549 subcutaneous xenografts consistently showed tumor growth suppression after 2 weeks of oral 30 and 60 mg/kg antroquinonol treatment.” (18) In addition, the natural product is reported to have in vivo activity in a Hep 3B tumor model (vide infra). (19) Because of the promising efficacy in this hepatocellular model and the limited in vitro activity against the A549 cell line, we reasoned that an active metabolite(s) could potentially be contributing to the observed in vivo activity seen with 1.

Table 1. Oncology Panel in Vitro Data
entrycell lineIC50 μM reported in the lit.aIC50 μM for 1b (72 h)c,dIC50 μM for 15 (72 h)c,e
1MDA-MB-2312.6 ± 0.05f19 ± 1.6 >25
2HepG24.3 ± 0.03f >25 >25
3LNCaP6.1 ± 0.07f22 ± 5.4 >25
4Hep 3B0.13 ± 0.02f8.9 ± 2.1 >25
5PANC-1 (48 h)c19g,h >25 >25
6AsPC-1 (48 h)c20g,h >25 >25
7A549 (12 h)c25i6.7 ± 2.510.8 ± 5.8
8H44125i >25e >25
a

Source of cell line reported in the literature. MDA-MB-231: CCRC-60425. HepG2: BCRC-60025. LNCaP: CCRC-60088. Hep 3B: BCRC-60434. PANC-1: ATCC. AsPC-1: ATCC. A549: KMUH. H441: KMUH.

b

Source of cell lines reported in this paper: ATCC.

c

Incubation time.

d

Triplicate data.

e

Duplicate data.

f

Reference 1.

g

Reference 2f.

h

SRB assay.

i

Reference 2c.

When 1 was subjected to metabolite identification/biotransformation studies using human, rat, and mouse liver microsomes, it was rapidly converted to a major metabolite across all three species (Figure 2), (20) the structure of which was tentatively assigned as the acid 15 based on MS/MS and 1H NMR data.

Figure 2

Figure 2. A. Antroquinonol A metabolite (15) profile in MsLM. X = sample matrix peaks, not parent related. B. Reagents and conditions: (a) LiHMDS (1.5 equiv), HMPA (2.5 equiv), allyl bromide (2.5 equiv), THF, 0 °C, 6 h (44%); (b) L-Selectride (2.0 equiv), toluene, −78 °C to −20 °C, 6 h (74%); (c) Montmorillonite K10 clay (30 wt %), DCM, 23 °C, 2 h (49%); (d) TBAF (2.0 equiv), THF, 23 °C, 2 h (99%); (e) PIDA (1.1 equiv), TEMPO (0.2 equiv), THF, 23 °C, 4 h; (f) NaClO2 (7 equiv), NaH2PO4 (7 equiv), 2-methyl-2-butene (20 equiv), t-BuOH, H2O, 23 °C, 12 h (58%). MsLM = mouse liver microsomes.

In order to confirm this structure and produce enough material for biological evaluation, a synthesis of 15 was undertaken. As a testament to the modularity of the synthetic route to 1, acid 15 was easily accessed through an identical strategy. Simply replacing farnesyl bromide with an oxidized derivative, followed by alkylation, reduction, and hydrolysis conditions, furnished antroquinonol analogue 17. Not surprisingly, chemoselective oxidation of the primary alcohol in the presence of the partially reduced quinone core of antroquinonol proved challenging. Employing TEMPO as a catalytic oxidant proved successful. (21) Subsequent oxidation of the sensitive aldehyde under Pinnick conditions gave the desired acid metabolite 15, which spectroscopically matched the isolated material from liver microsomes (vide supra).

Compound 15 was subjected to the same tumor cell line panel as 1; however, little to no cytotoxicity was observed. (22) Further metabolism studies where 1 was incubated with human, rat, and mouse hepatocytes showed that compound 15 is most likely rapidly oxidized and degraded to a known inactive metabolite (Met2) most likely via mitochondrial β-oxidation (Figure S2). (23a-23c)

Although the source of the Hep 3B tumor cell lines used in the literature is different from the one described herein (see Table 1 footnote), we decided to conduct an in vivo study with synthetic antroquinonol (1) based on the reported efficacy of the natural product. (19) Before embarking on such a study, a pharmacokinetic (PK) study of 1 in mouse was performed to determine the exposure of the compound when dosed either orally (po) or intraperitoneally (ip). A single 50 mg/kg dose of 1 provided the PK profile depicted in Table 2, in which exposures following oral administration were significantly lower compared to those from ip dosing.

Table 2. Mouse Pharmacokinetic Data
PK parametersip dosingapo dosinga
Cmax (nM)457 ± 4579 ± 52
Tmax (h)4 ± 32 ± 1
AUClast (nM·h)2290 ± 142266 ± 88
a

Corn oil was used as the dosing vehicle.

An in vivo efficacy study was conducted with 1 using Hep 3B HCC tumor xenografts implanted subcutaneously in female NSG mice. Despite literature reports of statistically significant antitumor activity observed with the natural product, (2a, 19) compound 1 was inactive (<70% tumor growth inhibition, TGI) in our hands when administered daily ip for 14 consecutive days (Figure 3). Paclitaxel served as a positive control in this study and was clearly efficacious (79% TGI) when administered on its optimal preclinical dosing regimen (24 mg/kg, qd×5, iv). Compound 1 was generally well-tolerated with no overt signs of toxicity and minimal weight loss (∼4%).

Figure 3

Figure 3. In vivo antitumor activity of synthetic antroquinonol in the Hep 3B HCC xenograft model. Last measurement in the control group includes only one animal, since all other mice in this group had to be euthanized due to severe tumor necrosis. Standard error of the mean (SE) is calculated using the average deviation built-in formula in Quantrix spreadsheets as part of IDBS’s EWorkBook Suite.

In summary, a concise, six-step, scalable, enantioselective synthesis of (+)-antroquinonol (1) has enabled an extensive reinvestigation of some of its reported preclinical biological properties. As outlined in this manuscript, compound 1 demonstrated only micromolar activity in a panel of select tumor cell lines. While the reported Hep 3B cytotoxicity data was intriguing, it was significantly less potent in our panel, therefore it was not unexpected that the compound was inactive in the xenograft model derived from this cell line. The modular synthetic route enabled us to prepare and evaluate a major metabolite identified from biotransformation studies; this compound was inactive and therefore unlikely contributing to the reported in vivo activity. Despite differences in the cell lines used for the in vitro assays (24) and the manner in which the in vivo studies were conducted (e.g., the different initial tumor sizes), the lack of efficacy in an in vivo preclinical model would be a cause for concern if the compound were to be advanced into the clinic for treating hepatocellular carcinoma. As indicated earlier, antroquinonol is currently in phase II clinical trials for the treatment of NSCLC. However, the reported in vitro IC50 for antroquinonol A (1) as well as the data with synthetic antroquinonol (Table 1) suggest that the activity of the parent compound 1 for this tumor cell line is in the high micromolar range. Based on the in vitro cytotoxicity data for the A549 cell line, metabolic stability data in microsomes, and mouse PK profile, very high exposures (25) or the presence of a yet unknown active metabolite may be necessary for 1 to show efficacy in treating NSCLC in the clinic.

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acscentsci.5b00345.

  • Crystallographic data (CIF)

  • Experimental procedures and analytical data (PDF)

  • NMR spectra (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Authors
    • T. G. Murali Dhar - Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States Email: [email protected]
    • Phil S. Baran - Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States Email: [email protected]
  • Authors
    • Matthew T. Villaume - Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
    • Eran Sella - Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
    • Garrett Saul - Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
    • Robert M. Borzilleri - Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
    • Joseph Fargnoli - Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
    • Kathy A. Johnston - Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
    • Haiying Zhang - Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
    • Mark P. Fereshteh - Departments of Discovery Chemistry, Oncology Discovery and Leads Discovery & Optimization, Preclinical Optimization, Bristol-Myers Squibb Co., Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
  • Notes
    The authors declare no competing financial interest.

Acknowledgment

Click to copy section linkSection link copied!

Financial support for this work was provided by NSF (predoctoral fellowship to M.T.V.) and George E. Hewitt Foundation (postdoctoral fellowship to E.S.). The authors thank Prof. Arnold L. Rheingold and Dr. Curtis E. Moore for X-ray crystallographic analysis. We thank Dr. Joel C. Barrish for useful discussions, Sarah Traeger for NMR analysis of the metabolite, Robin Moore, Paul Elzinga, and Georgia Cornelius for PK support, and Christopher Mulligan for in vivo support.

References

Click to copy section linkSection link copied!

This article references 25 other publications.

  1. 1

    Isolation and structural determination of antroquinonol A (1):

    Lee, T.; Lee, C.; Tsou, W.; Liu, S.; Kuo, M.; Wen, W. Planta Med. 2007, 73, 1412 DOI: 10.1055/s-2007-990232
  2. 2

    Biological activity studies of 1:

    (a) Chiang, P. C.; Lin, S. C.; Pan, S. L.; Kuo, C. H.; Tsai, I. L.; Kuo, M. T.; Wen, W. C.; Chen, P.; Guh, J. H. Biochem. Pharmacol. 2010, 79, 162 DOI: 10.1016/j.bcp.2009.08.022
    (b) Kumar, K. J.; Chu, F. H.; Hsieh, H. W.; Liao, J. W.; Li, W. H.; Lin, J. C.; Shaw, J. F.; Wang, S. Y. J. Ethnopharmacol. 2011, 136, 168 DOI: 10.1016/j.jep.2011.04.030
    (c) Kumar, V. B.; Yuan, T. C.; Liou, J. W.; Yang, C. J.; Sung, P. J.; Weng, C. F. Mutat. Res., Fundam. Mol. Mech. Mutagen. 2011, 707, 42 DOI: 10.1016/j.mrfmmm.2010.12.009
    (d) Tsai, P. Y.; Ka, S. M.; Chao, T. K.; Chang, J. M.; Lin, S. H.; Li, C. Y.; Kuo, M. T.; Chen, P.; Chen, A. Free Radical Biol. Med. 2011, 50, 1503 DOI: 10.1016/j.freeradbiomed.2011.02.029
    (e) Tsai, P. Y.; Ka, S. M.; Chang, J. M.; Lai, J. H.; Dai, M. S.; Jheng, H. L.; Kuo, M. T.; Chen, P.; Chen, A. Arthritis Rheum. 2012, 64, 232 DOI: 10.1002/art.33328
    (f) Yu, C. C.; Chiang, P. C.; Lu, P. H.; Kuo, M. T.; Wen, W. C.; Chen, P.; Guh, J. H. J. Nutr. Biochem. 2012, 23, 900 DOI: 10.1016/j.jnutbio.2011.04.015
    (g) Yang, S. M.; Ka, S. M.; Hua, K. F.; Wu, T. H.; Chuang, Y. P.; Lin, Y. W.; Yang, F. L.; Wu, S. H.; Yang, S. S.; Lin, S. H.; Chang, J. M.; Chen, A. Free Radical Biol. Med. 2013, 61, 285 DOI: 10.1016/j.freeradbiomed.2013.03.024
    (h) Chen, C. K.; Kang, J. J.; Wen, W. C.; Chiang, H. F.; Lee, S. S. J. Nat. Prod. 2014, 77, 1061 DOI: 10.1021/np400670a
    (i) Ho, C. L.; Wang, J. L.; Lee, C. C.; Cheng, H. Y.; Wen, W. C.; Cheng, H. H.; Chen, M. C. Biomed. Pharmacother. 2014, 68, 1007 DOI: 10.1016/j.biopha.2014.09.008
    (j) Hsu, C. Y.; Sulake, R. S.; Huang, P. K.; Shih, H. Y.; Sie, H. W.; Lai, Y. K.; Chen, C.; Weng, C. F. Br. J. Pharmacol. 2015, 172, 38 DOI: 10.1111/bph.12828
    (k) Lee, W. T.; Lee, T. H.; Cheng, C. H.; Chen, K. C.; Chen, Y. C.; Lin, C. W. Food Chem. Toxicol. 2015, 78, 33 DOI: 10.1016/j.fct.2015.01.012
  3. 3

    ClinicalTrials.gov Identifier: NCT02047344.

  4. 4
    GoldenBiotech’s new cancer drug Hocena has been awarded the “Research and Development Innovation Award” in Taipei Biotech Awards 2014. http://www.goldenbiotech.com.tw/en/newslist.html, accessed on July 18, 2015.
  5. 5
    Michaudel, Q.; Ishihara, Y.; Baran, P. S. Acc. Chem. Res. 2015, 48, 712 DOI: 10.1021/ar500424a
  6. 6

    Three total syntheses of 1 were reported while this manuscript was in preparation:

    (a) Hsu, C. S.; Chou, H. H.; Fang, J. M. Org. Biomol. Chem. 2015, 13, 5510 DOI: 10.1039/C5OB00411J
    (b) Sulake, R. S.; Chen, C. Org. Lett. 2015, 17, 1138 DOI: 10.1021/acs.orglett.5b00046
    (c) Sulake, R. S.; Lin, H. H.; Hsu, C. Y.; Weng, C. F.; Chen, C. J. Org. Chem. 2015, 80, 6044 DOI: 10.1021/acs.joc.5b00345
  7. 7
    (a) Magomedov, N. A.; Ruggiero, P. L.; Tang, Y. J. Am. Chem. Soc. 2004, 126, 1624 DOI: 10.1021/ja0399066
    (b) Matsumoto, T.; Hamura, T.; Miyamoto, M.; Suzuki, K. Tetrahedron Lett. 1998, 39, 4853 DOI: 10.1016/S0040-4039(98)00920-4
  8. 8
    Beckwith, A. L. J.; Chai, C. L. L. Tetrahedron 1993, 49, 7871 DOI: 10.1016/S0040-4020(01)88012-1
  9. 9
    Brailsford, J. A.; Lauchli, R.; Shea, K. J. Org. Lett. 2009, 11, 5330 DOI: 10.1021/ol902173g
  10. 10
    Kündig, E. P.; Enriquez-Garcia, A. Beilstein J. Org. Chem. 2008, 4, 37 DOI: 10.3762/bjoc.4.37
  11. 11
    Mantilli, L.; Gerard, D.; Torche, S.; Besnard, C.; Mazet, C. Angew. Chem., Int. Ed. 2009, 48, 5143 DOI: 10.1002/anie.200901863
  12. 12
    Solomon, M.; Jamison, C. L.; McCormick, M.; Liotta, D. J. Am. Chem. Soc. 1988, 110, 3702 DOI: 10.1021/ja00219a079
  13. 13
    Koreeda, M.; Koo, S. Tetrahedron Lett. 1990, 31, 831 DOI: 10.1016/S0040-4039(00)94639-2
  14. 14
    Meister, A. C.; Sauter, P. F.; Brase, S. Eur. J. Org. Chem. 2013, 2013, 7110 DOI: 10.1002/ejoc.201300752
  15. 15
    Pirrung, M. C.; Nunn, D. S. Tetrahedron Lett. 1992, 33, 6591 DOI: 10.1016/S0040-4039(00)60993-0
  16. 16
    Imbos, R.; Brilman, M. H. G.; Pineschi, M.; Feringa, B. L. Org. Lett. 1999, 1, 623 DOI: 10.1021/ol990707u
  17. 17

    The IC50 values for the natural product against the Hep3B and HepG2 carcinoma cell lines are reported as 0.13 ± 0.02 μM and 4.3 ± 0.03 μM respectively in ref 1, as indicated in Table 1. The GI50′ values for 1 against the same cell lines are listed as >30 μM and 0.22 μM respectively in ref 2a. The cytotoxicity assay protocol used in this paper and ref 1 employs 10% FBS and a 72 h incubation time with compound (antroquinonol) using MTS or MTT stain. The protocol outlined in ref 2a employs 5% FBS, 48 h incubation time, and an SRB stain. It is clear from the discussion (vide supra) that the protocol employed in this paper to determine cytotoxicity closely mirrors the conditions used in ref 1.

  18. 18

    ClinicalTrials.gov Identifier: NCT01134016.

  19. 19

    For discussion regarding the in vivo activity see ref 2a, page 170, left column, third paragraph. See supplementary material of ref 2a for in vivo data. Initial tumor sizes were not identical: 50–70 mm3 (lit.) (2a) vs 75–100 mm3 (BMS).

  20. 20

    Data shown for mouse liver microsomes.

  21. 21
    De Mico, A.; Margarita, R.; Parlanti, L.; Vescovi, A.; Piancatelli, G. J. Org. Chem. 1997, 62, 6974 DOI: 10.1021/jo971046m
  22. 22

    The Caco-2 value for 15 (Pc A → B, 62 nm/s, and B → A, 63 nm/s) suggests that cell permeability may not be an issue with 15.

  23. 23
    (a)

    Met2 (Figure S2; structure based on MS–MS data) is a reported metabolite of antroquinonol

    .
    (b)

    Met2 is inactive in the H838 tumor cell line (IC50 > 100 μM vs antroquinonol IC50 of ∼ 3 μM).

    Ho, C.-L.; Wang, J.-L.; Lee, C.-C.; Cheng, H.-Y.; Wen, W.-C.; Cheng, H. H-Y.; Chen, M. C-M. Biomed. Pharmacother. 2014, 68, 1007 DOI: 10.1016/j.biopha.2014.09.008
    (c)

    Synthetic antroquinonol appears to have a relatively better in vitro metabolic stability in human hepatocytes compared to human liver microsomes.

    .
  24. 24

    A thorough investigation of the antiproliferative activity against a large panel of human tumor cell lines was not performed.

  25. 25

    Antroquinonol is being administered to patients at 200 mg tid in the ongoing phase II clinical trial (ref 3).

Cited By

Click to copy section linkSection link copied!

This article is cited by 34 publications.

  1. David S. Peters, Cody Ross Pitts, Kyle S. McClymont, Thomas P. Stratton, Cheng Bi, Phil S. Baran. Ideality in Context: Motivations for Total Synthesis. Accounts of Chemical Research 2021, 54 (3) , 605-617. https://doi.org/10.1021/acs.accounts.0c00821
  2. Marianne Piochon, Pauline M. L. Coulon, Armand Caulet, Marie-Christine Groleau, Eric Déziel, Charles Gauthier. Synthesis and Antimicrobial Activity of Burkholderia-Related 4-Hydroxy-3-methyl-2-alkenylquinolines (HMAQs) and Their N-Oxide Counterparts. Journal of Natural Products 2020, 83 (7) , 2145-2154. https://doi.org/10.1021/acs.jnatprod.0c00171
  3. Jacob C. Timmerman, John L. Wood. Synthesis and Biological Evaluation of Hippolachnin A Analogues. Organic Letters 2018, 20 (13) , 3788-3792. https://doi.org/10.1021/acs.orglett.8b01381
  4. Jordan T. Koehn, Estela S. Magallanes, Benjamin J. Peters, Cheryle N. Beuning, Allison A. Haase, Michelle J. Zhu, Christopher D. Rithner, Dean C. Crick, Debbie C. Crans. A Synthetic Isoprenoid Lipoquinone, Menaquinone-2, Adopts a Folded Conformation in Solution and at a Model Membrane Interface. The Journal of Organic Chemistry 2018, 83 (1) , 275-288. https://doi.org/10.1021/acs.joc.7b02649
  5. Bo-Bo Zhang, Peng-Fei Hu, Jing Huang, Yong-Dan Hu, Lei Chen, and Gan-Rong Xu . Current Advances on the Structure, Bioactivity, Synthesis, and Metabolic Regulation of Novel Ubiquinone Derivatives in the Edible and Medicinal Mushroom Antrodia cinnamomea. Journal of Agricultural and Food Chemistry 2017, 65 (48) , 10395-10405. https://doi.org/10.1021/acs.jafc.7b04206
  6. Mei-Chuan Chen, Ting-Yu Cho, Yueh-Hsiung Kuo, and Tzong-Huei Lee . Meroterpenoids from a Medicinal Fungus Antrodia cinnamomea. Journal of Natural Products 2017, 80 (9) , 2439-2446. https://doi.org/10.1021/acs.jnatprod.7b00223
  7. Maoqun Tian, Ming Yan, and Phil S. Baran . 11-Step Total Synthesis of Araiosamines. Journal of the American Chemical Society 2016, 138 (43) , 14234-14237. https://doi.org/10.1021/jacs.6b09701
  8. Artiom Cernijenko, Rune Risgaard, and Phil S. Baran . 11-Step Total Synthesis of (−)-Maoecrystal V. Journal of the American Chemical Society 2016, 138 (30) , 9425-9428. https://doi.org/10.1021/jacs.6b06623
  9. Xiang Li. Natural products as inhibitors against pancreatic cancer cell proliferation and invasion: possible mechanisms. American Journal of Cancer Research 2024, 14 (6) , 2695-2713. https://doi.org/10.62347/XLZX8935
  10. Ardalan Pasdaran, Bahareh Hassani, Ali Tavakoli, Ekaterina Kozuharova, Azadeh Hamedi. A Review of the Potential Benefits of Herbal Medicines, Small Molecules of Natural Sources, and Supplements for Health Promotion in Lupus Conditions. Life 2023, 13 (7) , 1589. https://doi.org/10.3390/life13071589
  11. Thomas A. K. Prescott, Rowena Hill, Eduard Mas-Claret, Ester Gaya, Edie Burns. Fungal Drug Discovery for Chronic Disease: History, New Discoveries and New Approaches. Biomolecules 2023, 13 (6) , 986. https://doi.org/10.3390/biom13060986
  12. Feng-Wei Guo, Tian-Yi Zhou, Yong Qu, Mei-Yan Wei, Guang-Ying Chen, Yu-Cheng Gu, Chang-Yun Wang, Chang-Lun Shao. Highly efficient and scalable total syntheses and stereochemical assignment of potent anti-inflammatory pesimquinolones I and J. Organic Chemistry Frontiers 2023, 10 (4) , 852-858. https://doi.org/10.1039/D2QO01788A
  13. Susan Miller, Walter Moos, Barbara Munk, Stephen Munk, Charles Hart, David Spellmeyer. Drug discovery: Chaos can be your friend or your enemy. 2023, 417-511. https://doi.org/10.1016/B978-0-12-824304-6.00012-2
  14. Fernandez Francesca, Aust Caitlin, Lye Sarah, Griffiths Lyn Robyn. Antroquinonol administration in animal preclinical studies for Alzheimer's disease (AD): A new avenue for modifying progression of AD pathophysiology. Brain, Behavior, & Immunity - Health 2022, 21 , 100435. https://doi.org/10.1016/j.bbih.2022.100435
  15. Minghua Jiang, Zhenger Wu, Lan Liu, Senhua Chen. The chemistry and biology of fungal meroterpenoids (2009–2019). Organic & Biomolecular Chemistry 2021, 19 (8) , 1644-1704. https://doi.org/10.1039/D0OB02162H
  16. Yi Kuang, Bin Li, Zilong Wang, Xue Qiao, Min Ye. Terpenoids from the medicinal mushroom Antrodia camphorata : chemistry and medicinal potential. Natural Product Reports 2021, 38 (1) , 83-102. https://doi.org/10.1039/D0NP00023J
  17. Jing‐Kai Huang, Kak‐Shan Shia. Development of a Cross‐Conjugated Vinylogous [4+2] Anionic Annulation and Application to the Total Synthesis of Natural Antibiotic (±)‐ABX. Angewandte Chemie 2020, 132 (16) , 6602-6607. https://doi.org/10.1002/ange.201914657
  18. Jing‐Kai Huang, Kak‐Shan Shia. Development of a Cross‐Conjugated Vinylogous [4+2] Anionic Annulation and Application to the Total Synthesis of Natural Antibiotic (±)‐ABX. Angewandte Chemie International Edition 2020, 59 (16) , 6540-6545. https://doi.org/10.1002/anie.201914657
  19. Xiao-Feng Liu, Yong-Jun Xia, Phoency F-H Lai, Yao Zhang, Zhen-Wei Yi, Chun-Liang Xie, Yi-Qiu Hong, Lian-Zhong Ai. An increase in cell membrane permeability in the in situ extractive fermentation improves the production of antroquinonol from Antrodia camphorata S-29. Journal of Industrial Microbiology and Biotechnology 2020, 47 (2) , 197-207. https://doi.org/10.1007/s10295-020-02258-8
  20. Keita Sakamoto, Haruhiko Fuwa. Total Synthesis of a Marine Macrolide Natural Product, Iriomoteolide-2a: The Fundamental Role of Total Synthesis in Natural Product Chemistry. Journal of Synthetic Organic Chemistry, Japan 2019, 77 (8) , 831-840. https://doi.org/10.5059/yukigoseikyokaishi.77.831
  21. Keita Sakamoto, Akihiro Hakamata, Arihiro Iwasaki, Kiyotake Suenaga, Masashi Tsuda, Haruhiko Fuwa. Total Synthesis, Stereochemical Revision, and Biological Assessment of Iriomoteolide‐2a. Chemistry – A European Journal 2019, 25 (36) , 8528-8542. https://doi.org/10.1002/chem.201900813
  22. Bo-Bo Zhang, Yu-Yan Guan, Peng-Fei Hu, Lei Chen, Gan-Rong Xu, Liming Liu, Peter C. K. Cheung. Production of bioactive metabolites by submerged fermentation of the medicinal mushroom Antrodia cinnamomea : recent advances and future development. Critical Reviews in Biotechnology 2019, 39 (4) , 541-554. https://doi.org/10.1080/07388551.2019.1577798
  23. Alexander A. Braddock, Emmanuel A. Theodorakis. Marine Spirotetronates: Biosynthetic Edifices That Inspire Drug Discovery. Marine Drugs 2019, 17 (4) , 232. https://doi.org/10.3390/md17040232
  24. Hairui Yang, Xu Bai, Henan Zhang, Jingsong Zhang, Yingying Wu, Chuanhong Tang, Yanfang Liu, Yan Yang, Zhendong Liu, Wei Jia, Wenhan Wang. Antrodin C, an NADPH Dependent Metabolism, Encourages Crosstalk between Autophagy and Apoptosis in Lung Carcinoma Cells by Use of an AMPK Inhibition-Independent Blockade of the Akt/mTOR Pathway. Molecules 2019, 24 (5) , 993. https://doi.org/10.3390/molecules24050993
  25. Xiaoming Wang, Chao Du, Benke Hong, Xiaoguang Lei. Total synthesis of (±)-antroquinonol. Organic & Biomolecular Chemistry 2019, 17 (7) , 1754-1757. https://doi.org/10.1039/C8OB02494D
  26. Venkatachalam Angamuthu, M. Shanmugavadivu, G. Nagarajan, Bharath Kumar Velmurugan. Pharmacological activities of antroquinonol- Mini review. Chemico-Biological Interactions 2019, 297 , 8-15. https://doi.org/10.1016/j.cbi.2018.10.009
  27. Mairi M. Littleson, Christopher M. Baker, Anne J. Dalençon, Elizabeth C. Frye, Craig Jamieson, Alan R. Kennedy, Kenneth B. Ling, Matthew M. McLachlan, Mark G. Montgomery, Claire J. Russell, Allan J. B. Watson. Scalable total synthesis and comprehensive structure–activity relationship studies of the phytotoxin coronatine. Nature Communications 2018, 9 (1) https://doi.org/10.1038/s41467-018-03443-1
  28. Lizhi Zhu, Wenjing Ma, Mengxun Zhang, Magnolia Muk-Lan Lee, Wing-Yan Wong, Brandon Dow Chan, Qianqian Yang, Wing-Tak Wong, William Chi-Shing Tai, Chi-Sing Lee. Scalable synthesis enabling multilevel bio-evaluations of natural products for discovery of lead compounds. Nature Communications 2018, 9 (1) https://doi.org/10.1038/s41467-018-03546-9
  29. Chin-Sheng Hung, Sheng-Chao Wang, Yi-Ting Yen, Tzong-Huei Lee, Wu-Che Wen, Ruo-Kai Lin. Hypermethylation of CCND2 in Lung and Breast Cancer Is a Potential Biomarker and Drug Target. International Journal of Molecular Sciences 2018, 19 (10) , 3096. https://doi.org/10.3390/ijms19103096
  30. Hiromu Hattori, Joel Roesslein, Patrick Caspers, Katja Zerbe, Hideki Miyatake‐Ondozabal, Daniel Ritz, Georg Rueedi, Karl Gademann. Total Synthesis and Biological Evaluation of the Glycosylated Macrocyclic Antibiotic Mangrolide A. Angewandte Chemie 2018, 130 (34) , 11186-11190. https://doi.org/10.1002/ange.201805770
  31. Hiromu Hattori, Joel Roesslein, Patrick Caspers, Katja Zerbe, Hideki Miyatake‐Ondozabal, Daniel Ritz, Georg Rueedi, Karl Gademann. Total Synthesis and Biological Evaluation of the Glycosylated Macrocyclic Antibiotic Mangrolide A. Angewandte Chemie International Edition 2018, 57 (34) , 11020-11024. https://doi.org/10.1002/anie.201805770
  32. Anja Schüffler. Secondary Metabolites of Basidiomycetes. 2018, 231-275. https://doi.org/10.1007/978-3-319-71740-1_8
  33. Susan M. Miller, Walter H. Moos, Barbara H. Munk, Stephen A. Munk. Drug discovery. 2017, 183-279. https://doi.org/10.1016/B978-0-08-100625-2.00010-6
  34. Che‐Sheng Hsu, Jim‐Min Fang. Synthesis of (+)‐Antroquinonol and Analogues by Using Enantioselective Michael Reactions of Benzoquinone Monoketals. European Journal of Organic Chemistry 2016, 2016 (22) , 3809-3816. https://doi.org/10.1002/ejoc.201600542
Open PDF

ACS Central Science

Cite this: ACS Cent. Sci. 2016, 2, 1, 27–31
Click to copy citationCitation copied!
https://doi.org/10.1021/acscentsci.5b00345
Published December 23, 2015

Copyright © 2015 American Chemical Society. This publication is licensed under these Terms of Use.

Article Views

12k

Altmetric

-

Citations

Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Graphical summary of retrosynthetic analyses explored for the synthesis of antroquinonol A (1).

    Scheme 1

    Scheme 1. Total Synthesis of (+)-Antroquinonol A (1)a

    Scheme aReagents and conditions: (a) H2O2 (1.0 equiv), H2SO4 (0.1 equiv), MeOH, 0 °C, 2 h (98%); (b) PIDA (2.0 equiv), MeOH, 23 °C, (89%); (c) 2,2-dimethyl-1,3-propanediol (2.5 equiv), PPTS (0.15 equiv), toluene, 60 °C, 1 h (75%); (d) Cu(OTf)2 (0.05 equiv), phosphoramidite ligand SI2 (0.1 equiv), Me2Zn (2.5 equiv), toluene, −30 °C, 18 h; LiHMDS (1.5 equiv), HMPA (2.5 equiv), farnesyl bromide (2.0 equiv), 0 °C, 3 h; (e) L-Selectride (2.5 equiv), toluene, −78 °C to −20 °C, 6 h; (f) Montmorillonite K10 Clay (30 wt %), DCM, 23 °C, 2 h. PIDA = (diacetoxyiodo)benzene, PPTS = pyridinium p-toluenesulfonate, LiHMDS = lithium bis(trimethylsilyl)amide, HMPA = hexamethylphosphoramide, L-Selectride = lithium tri-sec-butylborohydride solution, DCM = dichloromethane.

    Figure 2

    Figure 2. A. Antroquinonol A metabolite (15) profile in MsLM. X = sample matrix peaks, not parent related. B. Reagents and conditions: (a) LiHMDS (1.5 equiv), HMPA (2.5 equiv), allyl bromide (2.5 equiv), THF, 0 °C, 6 h (44%); (b) L-Selectride (2.0 equiv), toluene, −78 °C to −20 °C, 6 h (74%); (c) Montmorillonite K10 clay (30 wt %), DCM, 23 °C, 2 h (49%); (d) TBAF (2.0 equiv), THF, 23 °C, 2 h (99%); (e) PIDA (1.1 equiv), TEMPO (0.2 equiv), THF, 23 °C, 4 h; (f) NaClO2 (7 equiv), NaH2PO4 (7 equiv), 2-methyl-2-butene (20 equiv), t-BuOH, H2O, 23 °C, 12 h (58%). MsLM = mouse liver microsomes.

    Figure 3

    Figure 3. In vivo antitumor activity of synthetic antroquinonol in the Hep 3B HCC xenograft model. Last measurement in the control group includes only one animal, since all other mice in this group had to be euthanized due to severe tumor necrosis. Standard error of the mean (SE) is calculated using the average deviation built-in formula in Quantrix spreadsheets as part of IDBS’s EWorkBook Suite.

  • References


    This article references 25 other publications.

    1. 1

      Isolation and structural determination of antroquinonol A (1):

      Lee, T.; Lee, C.; Tsou, W.; Liu, S.; Kuo, M.; Wen, W. Planta Med. 2007, 73, 1412 DOI: 10.1055/s-2007-990232
    2. 2

      Biological activity studies of 1:

      (a) Chiang, P. C.; Lin, S. C.; Pan, S. L.; Kuo, C. H.; Tsai, I. L.; Kuo, M. T.; Wen, W. C.; Chen, P.; Guh, J. H. Biochem. Pharmacol. 2010, 79, 162 DOI: 10.1016/j.bcp.2009.08.022
      (b) Kumar, K. J.; Chu, F. H.; Hsieh, H. W.; Liao, J. W.; Li, W. H.; Lin, J. C.; Shaw, J. F.; Wang, S. Y. J. Ethnopharmacol. 2011, 136, 168 DOI: 10.1016/j.jep.2011.04.030
      (c) Kumar, V. B.; Yuan, T. C.; Liou, J. W.; Yang, C. J.; Sung, P. J.; Weng, C. F. Mutat. Res., Fundam. Mol. Mech. Mutagen. 2011, 707, 42 DOI: 10.1016/j.mrfmmm.2010.12.009
      (d) Tsai, P. Y.; Ka, S. M.; Chao, T. K.; Chang, J. M.; Lin, S. H.; Li, C. Y.; Kuo, M. T.; Chen, P.; Chen, A. Free Radical Biol. Med. 2011, 50, 1503 DOI: 10.1016/j.freeradbiomed.2011.02.029
      (e) Tsai, P. Y.; Ka, S. M.; Chang, J. M.; Lai, J. H.; Dai, M. S.; Jheng, H. L.; Kuo, M. T.; Chen, P.; Chen, A. Arthritis Rheum. 2012, 64, 232 DOI: 10.1002/art.33328
      (f) Yu, C. C.; Chiang, P. C.; Lu, P. H.; Kuo, M. T.; Wen, W. C.; Chen, P.; Guh, J. H. J. Nutr. Biochem. 2012, 23, 900 DOI: 10.1016/j.jnutbio.2011.04.015
      (g) Yang, S. M.; Ka, S. M.; Hua, K. F.; Wu, T. H.; Chuang, Y. P.; Lin, Y. W.; Yang, F. L.; Wu, S. H.; Yang, S. S.; Lin, S. H.; Chang, J. M.; Chen, A. Free Radical Biol. Med. 2013, 61, 285 DOI: 10.1016/j.freeradbiomed.2013.03.024
      (h) Chen, C. K.; Kang, J. J.; Wen, W. C.; Chiang, H. F.; Lee, S. S. J. Nat. Prod. 2014, 77, 1061 DOI: 10.1021/np400670a
      (i) Ho, C. L.; Wang, J. L.; Lee, C. C.; Cheng, H. Y.; Wen, W. C.; Cheng, H. H.; Chen, M. C. Biomed. Pharmacother. 2014, 68, 1007 DOI: 10.1016/j.biopha.2014.09.008
      (j) Hsu, C. Y.; Sulake, R. S.; Huang, P. K.; Shih, H. Y.; Sie, H. W.; Lai, Y. K.; Chen, C.; Weng, C. F. Br. J. Pharmacol. 2015, 172, 38 DOI: 10.1111/bph.12828
      (k) Lee, W. T.; Lee, T. H.; Cheng, C. H.; Chen, K. C.; Chen, Y. C.; Lin, C. W. Food Chem. Toxicol. 2015, 78, 33 DOI: 10.1016/j.fct.2015.01.012
    3. 3

      ClinicalTrials.gov Identifier: NCT02047344.

    4. 4
      GoldenBiotech’s new cancer drug Hocena has been awarded the “Research and Development Innovation Award” in Taipei Biotech Awards 2014. http://www.goldenbiotech.com.tw/en/newslist.html, accessed on July 18, 2015.
    5. 5
      Michaudel, Q.; Ishihara, Y.; Baran, P. S. Acc. Chem. Res. 2015, 48, 712 DOI: 10.1021/ar500424a
    6. 6

      Three total syntheses of 1 were reported while this manuscript was in preparation:

      (a) Hsu, C. S.; Chou, H. H.; Fang, J. M. Org. Biomol. Chem. 2015, 13, 5510 DOI: 10.1039/C5OB00411J
      (b) Sulake, R. S.; Chen, C. Org. Lett. 2015, 17, 1138 DOI: 10.1021/acs.orglett.5b00046
      (c) Sulake, R. S.; Lin, H. H.; Hsu, C. Y.; Weng, C. F.; Chen, C. J. Org. Chem. 2015, 80, 6044 DOI: 10.1021/acs.joc.5b00345
    7. 7
      (a) Magomedov, N. A.; Ruggiero, P. L.; Tang, Y. J. Am. Chem. Soc. 2004, 126, 1624 DOI: 10.1021/ja0399066
      (b) Matsumoto, T.; Hamura, T.; Miyamoto, M.; Suzuki, K. Tetrahedron Lett. 1998, 39, 4853 DOI: 10.1016/S0040-4039(98)00920-4
    8. 8
      Beckwith, A. L. J.; Chai, C. L. L. Tetrahedron 1993, 49, 7871 DOI: 10.1016/S0040-4020(01)88012-1
    9. 9
      Brailsford, J. A.; Lauchli, R.; Shea, K. J. Org. Lett. 2009, 11, 5330 DOI: 10.1021/ol902173g
    10. 10
      Kündig, E. P.; Enriquez-Garcia, A. Beilstein J. Org. Chem. 2008, 4, 37 DOI: 10.3762/bjoc.4.37
    11. 11
      Mantilli, L.; Gerard, D.; Torche, S.; Besnard, C.; Mazet, C. Angew. Chem., Int. Ed. 2009, 48, 5143 DOI: 10.1002/anie.200901863
    12. 12
      Solomon, M.; Jamison, C. L.; McCormick, M.; Liotta, D. J. Am. Chem. Soc. 1988, 110, 3702 DOI: 10.1021/ja00219a079
    13. 13
      Koreeda, M.; Koo, S. Tetrahedron Lett. 1990, 31, 831 DOI: 10.1016/S0040-4039(00)94639-2
    14. 14
      Meister, A. C.; Sauter, P. F.; Brase, S. Eur. J. Org. Chem. 2013, 2013, 7110 DOI: 10.1002/ejoc.201300752
    15. 15
      Pirrung, M. C.; Nunn, D. S. Tetrahedron Lett. 1992, 33, 6591 DOI: 10.1016/S0040-4039(00)60993-0
    16. 16
      Imbos, R.; Brilman, M. H. G.; Pineschi, M.; Feringa, B. L. Org. Lett. 1999, 1, 623 DOI: 10.1021/ol990707u
    17. 17

      The IC50 values for the natural product against the Hep3B and HepG2 carcinoma cell lines are reported as 0.13 ± 0.02 μM and 4.3 ± 0.03 μM respectively in ref 1, as indicated in Table 1. The GI50′ values for 1 against the same cell lines are listed as >30 μM and 0.22 μM respectively in ref 2a. The cytotoxicity assay protocol used in this paper and ref 1 employs 10% FBS and a 72 h incubation time with compound (antroquinonol) using MTS or MTT stain. The protocol outlined in ref 2a employs 5% FBS, 48 h incubation time, and an SRB stain. It is clear from the discussion (vide supra) that the protocol employed in this paper to determine cytotoxicity closely mirrors the conditions used in ref 1.

    18. 18

      ClinicalTrials.gov Identifier: NCT01134016.

    19. 19

      For discussion regarding the in vivo activity see ref 2a, page 170, left column, third paragraph. See supplementary material of ref 2a for in vivo data. Initial tumor sizes were not identical: 50–70 mm3 (lit.) (2a) vs 75–100 mm3 (BMS).

    20. 20

      Data shown for mouse liver microsomes.

    21. 21
      De Mico, A.; Margarita, R.; Parlanti, L.; Vescovi, A.; Piancatelli, G. J. Org. Chem. 1997, 62, 6974 DOI: 10.1021/jo971046m
    22. 22

      The Caco-2 value for 15 (Pc A → B, 62 nm/s, and B → A, 63 nm/s) suggests that cell permeability may not be an issue with 15.

    23. 23
      (a)

      Met2 (Figure S2; structure based on MS–MS data) is a reported metabolite of antroquinonol

      .
      (b)

      Met2 is inactive in the H838 tumor cell line (IC50 > 100 μM vs antroquinonol IC50 of ∼ 3 μM).

      Ho, C.-L.; Wang, J.-L.; Lee, C.-C.; Cheng, H.-Y.; Wen, W.-C.; Cheng, H. H-Y.; Chen, M. C-M. Biomed. Pharmacother. 2014, 68, 1007 DOI: 10.1016/j.biopha.2014.09.008
      (c)

      Synthetic antroquinonol appears to have a relatively better in vitro metabolic stability in human hepatocytes compared to human liver microsomes.

      .
    24. 24

      A thorough investigation of the antiproliferative activity against a large panel of human tumor cell lines was not performed.

    25. 25

      Antroquinonol is being administered to patients at 200 mg tid in the ongoing phase II clinical trial (ref 3).

  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acscentsci.5b00345.

    • Crystallographic data (CIF)

    • Experimental procedures and analytical data (PDF)

    • NMR spectra (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.