Download as pdf or txt
Download as pdf or txt
You are on page 1of 39

European Journal of Medicinal Chemistry xxx (xxxx) xxx

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


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

Review article

A review: Biologically active 3,4-heterocycle-fused coumarins


Fatemeh Salehian a, Hamid Nadri b, Leili Jalili-Baleh a, Leila Youseftabar-Miri c,
Syed Nasir Abbas Bukhari d, Alireza Foroumadi a, e, Tuba Tüylü Küçükkilinç f,
Mohammad Sharifzadeh g, Mehdi Khoobi a, e, *
a
Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, 1417614411, Iran
b
Faculty of Pharmacy, and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
c
Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
d
Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Aljouf, Sakaka, 2014, Saudi Arabia
e
Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
f
Hacettepe University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
g
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences,
Tehran, Iran

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

Article history: The combination of heterocycles offers a new opportunity to create novel multicyclic compounds having
Received 28 August 2020 improved biological activity. Coumarins are ubiquitous natural heterocycle widely adopted in the design
Received in revised form of various biologically active compounds. Fusing different heterocycles with coumarin ring is one of the
15 November 2020
interesting approaches to generating novel hybrid molecules having highlighted biological activities. In
Accepted 15 November 2020
Available online xxx
the efforts to develop heterocyclic-fused coumarins, a wide range of 3,4-heterocycle-fused coumarins
have been introduced bearing outstanding biological activity. The effect of heterocycles annulation at
3,4-positions of coumarin ring on the biological activity of the target structures were discussed. This
Keywords:
Coumarin
review focuses on the important progress of 3,4-heterocycle-fused coumarins providing better insight for
Antibacterial medicinal chemists on the design and preparation of biologically active heterocycle-fused coumarins
Antiviral with a significant therapeutic effect in the future.
Antioxidant © 2020 Elsevier Masson SAS. All rights reserved.
Antipsychotic
Anticholinesterase

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1. Pyrrolocoumarin and indolocoumarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.1. MDR reversal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.2. Antitumor activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.3. Anti-retroviral activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.4. DYRK1A inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.2. Furocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.2.1. Antifungal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.2.2. Antiprofilerative activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.2.3. Antioxidant activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.2.4. Estrogen receptor modulators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.3. Thienocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.3.1. Antibacterial and antifungal activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.3.2. RNA-binding activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.3.3. Antiproliferative activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

* Corresponding author. The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, 1417614411, Iran.
E-mail addresses: m-khoobi@tums.ac.ir, mehdi.khoobi@gmail.com (M. Khoobi).

https://doi.org/10.1016/j.ejmech.2020.113034
0223-5234/© 2020 Elsevier Masson SAS. All rights reserved.

Please cite this article as: F. Salehian, H. Nadri, L. Jalili-Baleh et al., A review: Biologically active 3,4-heterocycle-fused coumarins, European
Journal of Medicinal Chemistry, https://doi.org/10.1016/j.ejmech.2020.113034
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

1.4. Selenocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00


1.4.1. Anticancer and antioxidant properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Coumarin-fused five-membered rings with two heteroatoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1. Pyrazolocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1.1. Antibacterial and antifungal activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1.2. Anticancer property . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2. Imidazolocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2.1. Anticancer property . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Coumarin-fused saturated six-membered ring . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1. Tetrahydropyridocoumarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1.1. Antifungal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1.2. Antipsychotic activity (dopamine D4 receptor antagonists) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1.3. Bronchodilatory activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Protoberberine derivatives (polycyclic coumarin-based compounds) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.1. Antibacterial and antifungal activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. Coumarin-fused unsaturated six-membered heterocycle with one heteroatom . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1. Pyridinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1.1. Antibacterial and antifungal activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1.2. Anti-inflammatory and analgesic activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1.3. Antimalarial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.1.4. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.2. Pyridonocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.2.1. Anticancer property . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.2.2. Fibrotic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.3. Quinolinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.3.1. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.4. Iminopyridinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.4.1. Antibacterial and antifungal activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.5. Benzofuro[3,2-b]pyridinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.5.1. Anti-inflammatory and antimicrobial activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.6. Pyranocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.6.1. Antiproliferative property . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.6.2. Anticholinesterase activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.7. Benzopyranocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.7.1. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.8. Dioxo-pyranocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.8.1. Antibacterial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.8.2. Antifungal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.9. Pyranoquinolino coumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.9.1. Anticholinesterase activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.10. Dihydropyrano pyrrolocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.10.1. Antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.11. Pyranopyrimidinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.11.1. Antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.12. Spirooxindoles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.12.1. Antibacterial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5.12.2. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. Coumarin-fused unsaturated six-membered rings with two heteroatoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1. Pyrimidinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1.1. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1.2. Antithrombotic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1.3. Antibacterial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.1.4. Antioxidant activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.2. Imidazopyrimidinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.2.1. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.3. Pyrimidopyrimidinocoumarin derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6.3.1. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7. Coumarin fused seven-membered ring . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.1. Thiazepinocoumarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
7.1.1. Anticancer and antioxidant activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Research ethics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Declaration of competing interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

2
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Abbreviations F oxysporum, Fusarium Oxysporum


G candidum, Geotrichum candidum
A mali, Alternaria mali GSK glycogen synthase kinase-3
A solani, Alternaria solani HIF1a Hypoxia inducible factor
A flavus, Aspergillus flavus M luteus, Micrococcus luteus
A niger, Aspergillus niger MIC minimum inhibitory concentration
A ochraceus Wilhelm, Aspergillus ochraceus Wilhelm MSCs Mesenchymal stem cells
B cereus, Bacillus cereus P chrysogenum Thom, Penicillium chrysogenum Thom
B subtilis, Bacillus subtilis P italicum, Penicillium italicum; PPA, Polyphosphoric
B cinerea, Botrytis cinerea acid
C albicans, Candida albicans P mirabilis, Proteus mirabilis
CDI 1,10 -Carbonyldiimidazole P aeruginosa, Pseudomonas aurignosa
CDK cyclin-dependent kinases Py Pyridine;
CHO Chinese hamster ovary Rh Solani, Rhizoctonia solani
C capsici, Colletotrichum capsici S enteritidis, Salmonella Enteritidis
DOX Doxorubicin SAR Structure-activity relationship;
DPPF 1,10 -Bis(diphenylphosphino)ferrocene S brevicaulis, Scopulariopsis brevicaulis
DYRK1A Dual-specificity tyrosine phosphorylation-regulated S marcescens, Serratia marcescens
kinase 1A SPHK1 sphingosine kinase 1
EAC Ehrlich ascites carcinoma S aureus, Staphylococcus aureus
ECA Ethyl cyanoacetate TFA Trifluoroacetic acid
E coli, Echerichia coli T. rubrum Trichophyton rubrum
E aerogenes, Enterobacter aerogenes

1. Introduction

The development or discovery of effective and inexpensive


drugs against disease is one of the main goals of researchers in the
field of medicinal chemistry. Over the years, great attention has
been paid to coumarin and its derivatives, which are versatile
molecules exhibiting a wide variety of biological properties
including antimicrobial, antiviral, anticancer, antioxidant, anti-
inflammatory, anti-tuberculosis, anti-influenza, anti-Alzheimer,
and anti-hyperlipidemia activities [1e15]. Coumarins (1) belong to
a-benzopyrone heterocyclic systems derived from Coumarouna
Fig. 2. Various types of heterocycle coupling with coumarin scaffold.

Fig. 3. The biological activities of 3,4-heterocycle fused coumarin derivatives (X is a


Fig. 1. Examples of clinically used coumarin derivatives.
hetero atom).
3
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Fig. 4. Representative of possible fusion of pyrrole/indole to the coumarin ring.

Scheme 2. Synthesis of pyrrolo[3,4-b]coumarins as antitumor agents.

Fig. 5. Structure and biological activities of pyrrolo[3,4-b]coumarin.

Fig. 7. Indolo[3,4-b]coumarins as anti-retroviral agent.

Scheme 1. Synthesis of Ningalin B derivative as MDR reversal agent.

Scheme 3. Synthesis of indolo[3,4-b]coumarins as DYRK1A inhibitor.

odorata Aube (Dipteryx odorata) first time [16]. They occur naturally
as a secondary metabolite in several plants, microorganisms, and
essential oils [17e19]. They have become a hot point in the me-
Fig. 6. Lamellarin I as MDR reversal agent.
dicinal chemistry because of their various pharmacological poten-
tials and less harmful effects against normal cells. The clinical
applications of certain members of this heterocycle have been

4
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Scheme 5. Synthesis of furocoumarins as anti-proliferative agents.

Fig. 8. Structure and biological activities of furo[3,2-c] coumarins.


Table 2
Anti-proliferative activity of furocoumarins 37a-37d.

Comp. No. X R IC50 (mM)

HCT-15 MCF-7

37a O CH3 212 112


37b O CH2CH3 10 16
37c N CH2CH2CH2CH2 8 19
37d N (CH3)2 252 50

Fig. 9. Chemical structure of Pterophyllin 2.

Fig. 10. Coumestrol as antiproliferative agent.

Scheme 4. Synthesis of furocoumarins.

reported extensively. For example, Phenprocoumon (2), Warfarin


(3), and Acenocoumarol (4) are considered as potent anticoagulant
agents (Fig. 1) [16,20]. Armillarisin A (5) is an antibiotic and Scheme 6. Synthesis of furocoumarin derivatives possessing anticancer activity.
Hymecromone (6) is a choleretic and antispasmodic agent [21].
Ensaculin (7) is also a well-known coumarin-based structure hav-
ing valuable anti-dementia activity on the neurotransmitter system Heterocyclic-fused coumarins are important and interesting
with an acceptable pharmacodynamic profile [22]. It could be pharmacophoric moieties existing in many natural or synthetic
concluded that the pharmacological properties and therapeutic biologically active compounds [24]. They have provoked much in-
applications of coumarins strongly depend on the pattern of sub- terest in researchers owing to diverse biological activities including
stitution [16]. antimicrobial, antiviral, anticancer, anticholinesterase, antipsy-
The presence of two or more heterocyclic nuclei in one molecule chotic, anti-malarial, dopamine D4 receptor inhibition, adenosine
can enhance the biological effects of the target system or generate receptor binding ability, anti-fibrotic, anti-coagulant, estrogen re-
new biological properties. When different heterocyclic rings are ceptor modulation, anti-inflammatory, analgesic, bronchodilation,
linked to coumarin either as a substituent or as a fused component SPHK1 inhibition and DYRK1A inhibition (Fig. 3). Despite several
into lactone (8,9) or benzene (10,11) ring, a synergistic effect of both reviews on the biological aspects of coumarins, none of them has
the rings is obtained in the target system (Fig. 2) [23]. The coupling focused specifically on 3,4-heterocycle fused coumarins and their
of the coumarin ring with different heterocycles has produced biological activities. In the present review, we try to find a corre-
novel hybrid molecules bearing highlighted biological activities. lation between the type of heterocycle fused at 3 and 4 positions of

Table 1
Antifungal effect of compound 34.

Pathogen EC50 of compound 34

Botrytis cinerea R1 ¼ Isopropoxy, R2 ¼ Me > R1 ¼ Allyloxy, R2 ¼ H > R1 ¼ Allyloxy, R2 ¼ Me > Oshtol


Collecterichum capsica R1 ¼ Ethoxy, R2 ¼ Me > R1 ¼ Allyloxy, R2 ¼ H > Oshtol
Rhizoctorzia solani R1 ¼ OEt, R2 ¼ H > Oshtol > R1 ¼ OMe, R2 ¼ Me > R1 ¼ OEt, R2 ¼ Me > R1 ¼ Allyloxy, R2 ¼ H

5
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 3
Anticancer activity of furocoumarins 40a-40c.

Comp. No. R1 R2 R3 ED50 (mg/mL)

MCF-7 ZR-75-1 SK-BR-3

40a Me H Me 0.60 0.25 0.18


40b Et H Me 0.45 0.18 0.10
40c H H Me 4.00 4.00
40d H Cl Me 5.0 8.6 4.5 Fig. 12. Furocoumarin as Estrogen receptor modulator.
40e H OMe Me 5.0 >10 8.6
40f Me H H 5.5 >10 8.5
40g Et H H >10
40h H H H >10
40i H Cl H 7.0 >10 >10 1.1.1. MDR reversal activity
40j H OMe H 3.4 8.5 4.0 A major obstacle in the successful chemotherapy of cancers is
Tamoxifen 5.0 3.6 5.0 MDR. Extensive studies have been carried out during the past few
decades to improve the ability of chemotherapy by overcoming
MDR. The major mechanism of MDR in mammalian cells includes
the overexpression of human P-glycoprotein (P-gp, a plasma
membrane protein) by the MDR1 gene. Some alkaloids with the
core of pyrrolo[3,4-b]coumarin like lamellarins and ningalins acted
as nontoxic inhibitors of MDR pumps in different cancer cell lines
and could reverse MDR at noncytotoxic concentrations by inhibit-
ing P-gp-mediated drug efflux.
Boger et al. described a three-step synthesis protocol including
(1) N-alkylation, (2) lactonization and (3) decarboxylation started
from tetrasubstituted pyrrole (16) and phenethyl bromide (17),
resulted in permethyl ether of ningalin B (18) (Scheme 1) [26,27].
Fig. 11. Furocoumarin derivatives as antioxidant agents. Ningalin B (19) was synthesized by the demethylation of compound
18 with BBr3. Then, the cytotoxicity and MDR reverse activity of the
products (18 and 19) against L1210, HCT116, and HCT116/VM46 cell
coumarins and corresponding biological activities. This review is
lines were evaluated [28]. The results of the cytotoxic assay
classified according to the size (including five-, six- and seven-
exhibited that ningalin B (19) was 2.5- and 5-times more active
membered ring), type of heterocycles (including saturated and
than the O-methyl derivative (18) against HCT116 and L1210,
unsaturated), and the number (mono, di, and tri-) or nature of
heteroatoms on the ring (including nitrogen, oxygen, sulfur and
selenium heteroatoms) fused to 3 and 4 positions of coumarin. Each
section is fully discussed and compared in terms of the biological
activities of the compounds.
2. Coumarin-fused five-membered ring with one heteroatom
(Pyrrole, Indole, Furan, Thiophene, Selenophene).

1.1. Pyrrolocoumarin and indolocoumarin

A fusion of pyrrole or indole to the different sites of the


coumarin ring results in diverse molecular structures. There are
two possible sites for the fusion of coumarin with pyrrole or indole
ring producing pyrrolo[3,4-b]coumarin (12), pyrrolo[4,3-b] Fig. 13. Structure and bioactivities of thienocoumarins.
coumarin (13), indolo[3,4-b]coumarin (14), and indolo[4,3-b]
coumarin (15) derivatives (Fig. 4).

The pyrrolo[3,4-b]coumarin or chromeno[3,4-b]pyrrol-4(3H)-


one derivatives are one of the well-known natural marine alkaloids
and synthetic compounds having an appreciable biological signif-
icance (Fig. 5) [25e40]. Pyrrolocoumarins showed diverse biolog-
ical activities including multidrug resistance (MDR) reversal
[26e28], antitumor, and anti-retroviral activities. They were also
introduced as potent DYRK1A inhibitors [29e40]. The synthesis and
biological activities of this valuable coumarin-fused heterocycle are
discussed below. Scheme 7. Synthesis of thieno[3,2-c]coumarin derivatives as antimicrobial agents.

6
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 4
Antibacterial and antifungal activity of compounds 44e47.

Inhibition Zone (mm)

Comp. No. 1 2 3 4 5 6 7 8 9 10 11 12

44 9 0 0 10 8 8 13 0 0 0 8 0
45 10 0 0 10 11 0 14 0 10 16 13 13 Fig. 14. Thienocoumarin as RNA binding agent.
46 8 0 0 0 0 0 10 0 0 0 0 0
47a 0 0 0 0 0 0 13 10 12 0 10 11
47b 0 0 0 0 0 0 13 0 0 0 0 8
47c 0 0 0 9 0 0 0 0 0 0 0 0
47d 9 0 9 0 0 0 0 0 0 0 0 10
47e 10 0 9 9 0 0 13 0 0 0 0 0
Chloramphenicol 18 22 18 18 20 20 20 36 44 28 20 24

Organisms: 1) S. aureus, 2) B. cereus, 3) E. coli, 4) P. aeruginos, 5) S. marcescens, 6)


M. luteus, 7) C. albicancs, 8) T. rubrum, 9) A. flavus, 10) F. oxysporum, 11) S. brevicaulis,
and 12) G. candidum.

respectively. However, the MDR reversal activity of compound 18


was more effective than that of ningalin B and verapamil as the
standard inhibitors. On the other hand, compound 18 showed a
significant capability to reverse MDR by resensitizing a resistant
human colon cancer cell line (HCT116/VM46) to vinblastine (IC50
value: 0.002 mM) and doxorubicin (IC50 value: 0.01 mM) at 1 mM.
Lamellarin I (20), isolated from Didemnum sp., exhibited high Scheme 9. Synthesis of thienocoumarins as antiproliferative agents.

activity against MDR P388/Schabel cells at a non-toxic concentra-


tion (Fig. 6) [29]. The cytotoxicity of doxorubicin (DOX), vinblastine,
observed for most of the lamellarins. Interestingly, lamellarin N
and daunorubicin against MDR cell lines was effectively increased
(25) and dehydrolamellarin J (26) (Scheme 2) not only were
by compound 20. The MDR reversal activity of compound 20 was 9-
significantly more active than the other lamellarins and even than
to 16- fold more than that of verapamil at 2 mM. It was also found
etoposide as the positive control against most the cancer cell lines,
that compound 20 could modulate the resistance by inhibition of
but also exhibited low toxicity against fibroblast cell line (MRC-5
the P-gp pump function.
IC50 > 97 mM).
Lamellarin D (compound 24) has been also reported as a potent
1.1.2. Antitumor activity antiproliferative agent against various tumor cells, such as human
A six-step reaction sequence was utilized for the preparation of prostate cancer cells (DU-145, LNCaP) and human chronic mye-
lamellarins (23) prepared from benzylisoquinoline derivative (21) logenous leukemia cells (K562) [32,33]. It has been also introduced
and compound 22 (Scheme 2) [30,31]. Anticancer activity of the as a potent mitochondrial topoisomerase I inhibitor [34,35]. It has
prepared lamellarins were evaluated against different cancerous been confirmed by various studies that the presence of full pen-
cell lines such as T47D and MDA-MB-231 (breast cancer cells), KB tacyclic pharmacophore, the planarity of the structure resulted
(mouth carcinoma cells), HuCCA-1 (cholangiocarcinoma cells), from saturation of the C5eC6 bond, and the presence of hydroxyl
HepG2 and S102 (hepatocellular carcinoma cells), Hela (cervical groups at C7, C8 and C20 are crucial for remarkable cytotoxic and
cancer cells), A549 (lung carcinoma cells), H69AR (multidrug topoisomerase I inhibition activity [31].
resistant lung cancer), HL-60 and P388 (leukemic cells), and MRC-5
fibroblast cells. The prepared lamellarins exhibited significant 1.1.3. Anti-retroviral activity
cytotoxic activity. Especially the cytotoxic activity of lamellarins K HIV-1 integrase, a multi-domain enzyme, is required for the
(23) against H69AR and HuCCA-1 cell lines (IC50: 4.3 and 3.4 mM, integration of retroviral DNA into the host genome. It is an
respectively) was much more than that of the positive control, endorsed target for developing anti-HIV agents. Sulfated marine
etoposide (anticancer drug, IC50: 45.9 and 6.8 mM, respectively). alkaloids have been introduced as an inhibitor of HIV-1 integrase
Lamellarin D (24) showed remarkable cytotoxic activity against all and HIV-1 replication [36]. Lamellarin a 20-sulfate (27) was
the cell lines (IC50: 0.00008e3.2 mM). It was more toxic than the
standard drug, etoposide, against all the tested cell lines, except
MDA-MB-231 and S102 cell lines. No selective toxicity was

Scheme 8. Synthesis of thienocoumarins as antimicrobial agents. Fig. 15. Selenocoumarins and their bioactivities.

7
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Fig. 16. Selenocoumarin derivatives as anti-proliferative and antioxidant agents as well as MMP inhibitors.

isolated from unidentified ascidian and evaluated against pre- lipophilic groups like eCH3, eCF3, eC6H4OH and 4-MeOC6H4 on
integration complexes and HIV. Acceptable inhibitory activity was indoline moiety spontaneously decreased the kinase inhibition
obtained for the compound (Fig. 7) [36]. The screening of the in- activity of the target compounds.
hibition of viral replication revealed that compound 25 could
inhibit HIV replication in early steps with IC50 value of 8.0 mM. 1.2. Furocoumarin derivatives

1.1.4. DYRK1A inhibitors Furocoumarins are found in a vast number of natural and syn-
The dual-specificity tyrosine phosphorylation-regulated kinase thetic compounds with potential biological and pharmacological
1A (DYRK1A) is one of the important target in neurodegenerative activities [41]. Furocoumarins have gained interest due to the
diseases [37]. Researchers have designed various compounds as antioxidant, cytotoxic, antifungal, NF-kB pathway inhibition and
DYRK1A inhibitors to treat neurological disorders. Several DYRK1A estrogen receptor modulating activity. They have been candidate
inhibitors have been reported so far [37e39]. Neagoie et al. pre- for skin diseases treating, such as psoriasis [42]. Compounds having
pared a series of lamellarin D analogs from the reaction of 2- furo[3,2-c]coumarin scaffold have shown interesting biological
hydroxyphenylboronic acid derivatives (29) with indole de- activities encouraging researchers to find a new way for simple
rivatives (28) through a Suzuki crosscoupling reaction, and a one- preparation of this valuable framework (Fig. 8) [43e51].
pot deprotection/lactonization step (Scheme 3). The inhibitory ef-
fect of the indolocoumarins (30) was investigated against topo- 1.2.1. Antifungal activity
isomerase I and kinases (CDK5, GSK3, and DYRK1A). The study Pterophyllin 2 (31) is a furo[3,2-c]coumarin derivative isolated
showed that the presence of the hydroxyl group and its position at from methanolic extract of the Ekebergia pterophylla. It has
the C-2/C-3 and C-10 could strongly affect the both activities. showed desirable inhibitory activity against phytopathogenic fungi
Among all the prepared indolocoumarins, compound 30a bearing (Fig. 9) [43a].
hydroxyl group at C-3 and C-10 positions exhibited a strong topo- Furo[3,2-c]chromen-4-ones (34) were synthesized via the re-
isomerase I inhibitory activity (IC50: 38.5 nM) and weak kinase action of 4-hydroxycoumarins (32) with a-chloroketones (33) un-
activity (DYRK1A IC50: 0.3 mM). Although compound 30b bearing der microwave irradiation (Scheme 4) [43band43c]. The products
hydroxyl group at C-2 showed no significant topoisomerase inhi- showed antifungal activity against Botrytis cinerea, Collecterichum
bition activity, a selective and efficient DYRK1A inhibition effect capsica, Alternaria solani, Gibberella zeae, and Rhizoctorzia solani at
was observed with IC50 value of 67 nM. The molecular modeling the concentration of 50 mg/mL (Table 1). The results revealed that
also revealed that the planar skeleton of compound 30b is the main incorporation of the substituents like isopropoxy and allyloxy on
part creating a strong binding with the ATP active site of DYRK1A the coumarin nucleus affected the antifungal activity, so that these
similar to lamellarin D. In addition, the presence of bulky and compounds had 100% inhibition against Botrytis cinerea, and
8
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Scheme 11. Synthesis of pyrazolocoumarin derivatives as antibacterial and antifungal


agents.
Fig. 17. Structure and biological activities of pyrazolocoumarins.

Fig. 18. Pyrazolocoumarins as anticancer agents.


Scheme 10. Synthesis of aminopyrazolocoumarin

DNA ¼ 8.1  103 M1, K37c-DNA ¼ 1.1  104 M1, K37b-


showed more activity than oshtole as a positive control (92.8%). BSA ¼ 5.1  104 M1, and K37c-BSA ¼ 7.6  104 M1). The acceptable
Isopropoxy-substituted derivative of compound 34 with R2 ¼ Me anticancer activity of compounds 37b and 37c was referred to the
showed the strongest activity against Botrytis cinerea (EC50: 1.72 mg/ desirable interaction and binding affinity of the compounds with
mL), as compared to Osthole (EC50: 6.45 mg/mL). Allyloxy- DNA.
substituted compound with R2 ¼ H exhibited a better antifungal Coumestrol (38) is a benzofuranocoumarin derivative (Fig. 10).
activity against Botrytis cinerea and Collecterichum capsica rather This naturally occurring antitumor compound is a protein kinase
than osthole. In the case of Collecterichum capsica, OEt-substituted CKII inhibitor against MCF-7 and HCT116 cells with 50% and 30%
compound with R2 ¼ Me revealed the higher antifungal activity growth inhibition at 50 mM, respectively [45b]. Cho and co-workers
(EC50: 6.02 mg/mL) than allyloxy-substituted analogue with studied the antitumor mechanism of compound 38 in hypoxic
R2 ¼ Me (EC50: 8.74 mg/mL) and Osthol (EC50: 16.83 mg/mL). Also, prostate PC-3 cancer cells targeting hypoxia inducible factor 1a
the OEt-substituted compound with R2 ¼ H was the most effective (HIF1a) and sphingosine kinase 1 (SPHK1) signaling [46]. MTT assay
inhibitor of Rhizoctorzia solani (EC50: 3.57 mg/mL); whereas revealed that compound 38 could inhibit the activity of SPHK1, HIF-
allyloxy-substituted compound with R2 ¼ H had weaker antifungal 1a, pAKT, and pGSK-3b expressions in a time- and dose-dependent
activity (EC50: 8.87 mg/mL) than osthole (EC50: 6.88 mg/mL). manner in hypoxic PC-3 cells. The results disclosed that compound
38 suppressed HIF-1 a accumulation via the SPHK1 suppression
pathway in hypoxic PC-3 cells.
1.2.2. Antiprofilerative activity Neo-tanshinlactone (40a) isolated from Salvia miltiorrhiza dis-
A series of furo[3,2-c]coumarin derivatives were prepared from played potent and selective anti-breast cancer activity (Scheme 6)
the reaction of compound 35 with alcohols or amines (36) in the [47a]. Compounds 40a showed 10-fold better activity and more
presence of Et3N as a catalyst and their potential anti-proliferative than 20-fold higher selectivity against ER þ cell lines (MCF-7 and
activity was then screened against MCF-7 breast cancer cell line and ZR-75-1) than tamoxifen. Wang et al. synthesized compounds 40a-
HCT-15 colon cancer cells (Scheme 5) [44]. The cytotoxicity results 40j from substituted a-naphthols (39) and evaluated their anti-
showed that replacing of 2-methyl group (37a) with 2-ethoxy cancer activity against several human tumor cell lines (MCF-7, ZR-
group (37b) improved the activity by 22- and 7-times on HCT-15 75-1, MDA MB-231, HS587-1, SK-BR-3, A431, SW620, and KB)
and MCF-7, respectively (Table 2). Compound 37c bearing 2- [47b]. The results of the best compounds are presented in Table 3.
pyrrolidinyl group at furan ring exhibited 31.5- and 2.6-fold more The SAR studies revealed that the presence of the substitution at 4-
active than compound 37d bearing 2-dimethylamine substituent position of the ring A was important for the anticancer activity. The
on HCT-15 and MCF-7, respectively. Based on the results, com- order of the activity among compounds substituted at the 4-
pounds 37b bearing ethoxy group (X ¼ O, R ¼ CH2CH3) and 37c position of the ring A (40a-40c) was ethyl (40b) ~ methyl
having pyrrolidinyl moiety (X ¼ N, R ¼ eCH2CH2CH2CH2-) were the (40a) > no substituent (40c). The presence of the substituents like
most active compounds against HCT-15 and MCF-7 cell lines. The Cl or OMe at position 6 of the ring B had no significant effect on the
anti-proliferative activity of the tested compounds against both the activity. Also, the presence of methyl group on the furan moiety
cell lines was found in order of 37b > 37a and 37c > 37d. Besides, (ring D) was critical for the anti-breast cancer activity of the target
both compounds 37b and 37c diminished HCT-15 cancer cell pro- compounds (40a-40e) resulting in better activity than unsub-
liferation more than that of MCF-7. Compounds 37b and 37c had stituted analogs (40f-40j).
appropriate DNA and BSA-binding affinity (K37b-
9
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Fig. 19. Structure and bioactivity of the imidazolocoumarin.

Scheme 13. Tetrahydropyridocoumarin derivatives as antifungal agents.

1.2.4. Estrogen receptor modulators


Psoralidin (43), isolated from Psoralea corylifolia (an estrogenic
plant metabolite), is a benzofurocoumarin derivative having a wide
range of biological activities such as regulation of insulin signaling,
antibacterial, antioxidant, anti-inflammatory, anticancer, and anti-
depressant activities [49,50]. Liu et al. demonstrated that this
Scheme 12. Synthesis of imidazolocoumarin derivatives as anticancer agents.
compound is a potent estrogen receptor (ER) signaling agonist
(Fig. 12) [51]. They described that compound 43 could activate the
1.2.3. Antioxidant activity classical pathway of ER-signaling in both ER-positive human breast
Wedelolactone (41), a phenolic derivative of Coumestan (42), is (MCF-7) and endometrial (Ishikawa) cell lines as well as non-
a natural furocoumarin having potent antioxidant properties human cultured cells expressing shortly either ERa or ERb
(Fig. 11) [48]. Compound 41 could efficiently protect Mesenchymal (EC50 ¼ 3.68 and 6.88 mM, respectively). The relative binding af-
stem cells (MSCs) from hydroxyl radical-mediated oxidative dam- finity of psoralidin to both ERa and ERb was 0.56% and 0.042%,
age. Wedelolactone (41) could increase the viability of hydroxyl respectively. IC50 values to replace the binding of E2 to both re-
radical-treated MSC cells. The antioxidant activity of this furo- ceptors were obtained 1.03 and 24.6 l mM, respectively.
coumarin was 1.62 times higher than that of the standard antiox-
idant (Trolox). The antioxidant mechanism of wedelolactone (41) 1.3. Thienocoumarin derivatives
has been ascribed as the direct radical scavenging via a single
electron transfer to radical adduct formation pathway. The results Thiophene structure is present in many natural products and
showed that the catechol moiety of the compound was essential to several pharmacologically active compounds. Thieno-fused
bind and scavenge DPPH radical for both of the pathways. Com- coumarin derivatives have gained much attention due to their
pound 41 has been suggested as an effective therapeutic agent for wide range of bioactivities such as antibacterial, antifungal, and
MSCs transplantation in bone-related diseases, especially osteo- anticancer activities (Fig. 13) [52e55].
porosis, due to the phytoestrogenic and antioxidant effects.

1.3.1. Antibacterial and antifungal activities


A series of thieno[3,2-c]coumarin derivatives (44e47) were
synthesized from 4-hydroxycoumarin (32) (Scheme 7) [52]. The
antibacterial activity of the products was evaluated against
different strains of bacteria (such as S. aureus, B. cereus, E. coli, P.
aeruginosa, S. marcescens, M. luteus) and fungus (C. albicancs, T.
rubrum, A. flavus, F. oxysporum, S. brevicaulis, G. candidum) (Table 4).
The aminothienocoumarin carboxylate (44) and carbohydrazide
(45) exhibited similar activity against P. aeruginosa (inhibition
zone ¼ 10 mm), less active than chloramphenicol as standard drug
(inhibition zone ¼ 18 mm). Replacement of the ester group (44)
with the carbohydrazide group led to the higher antifungal activity,
without any significant changes in the antibacterial activity. Ami-
noester derivative (44) exhibited remarkable activity against
Fig. 20. Structure and biological properties of tetrahydropyrido[3,4-c]coumarins. S. aureus, P. aeruginosa, S. marcescens, M. luteus, C. albicans and
10
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 5
Antifungal activity of the pyridocoumarin derivatives.

Comp. No. The percentage of the growth reduction of bacteria and the mycelium of the fungus or mold in comparison with the control

Xanthomonas malvacearum Fusarium moniliform Rhizoctonia solani Powdery mildew Phytophthora infection Gray mold

80a 9 18 30 68 83 32
80b 9 6 2 0 0 21
80c 9 14 0 8 0 0
81a 0 20 13 38 83 0
81b 18 10 0 52 83 11
81c 0 22 15 0 0 0

Fig. 23. Structure and biological activities of protoberberine derivatives.

Fig. 21. Tetrahydropyridocoumarins as antipsychotic agents.

Table 6
Receptor binding data for the pyridocoumarins 83e86.

Ki (nM)a
1 2 3
Comp. No. R R R D4 D2 D3 D2/D4

83a OMe H H 1.5 436 60 291


83b Me H H 3.6 774 338 216
83c OMe 4-Cl H 8.7 >5800 2780 667
83d Me 4-Cl H 11 1170 e 106
83e OH 4-Cl H 2.4 2780 317 1160
83f OMe 4-F H 4.3 >5800 548 1350
83g Me 4-F H 7.5 >5800 2210 773
83h OMe 2-Cl H 34 >5800 2620 171
83i OMe 3-Cl H 17 5500 2630 324
83j OMe 3-Cl 4-Cl 61 >5800 e 95
83k OMe 4-Me H 2.6 1170 491 450
83l OMe 4-OMe H 84 >5800 e 69
83m OMe 4-CF3 H 8.2 3700 966 451
84 e e e >3300 e e e
85 e e e 7.9 >5800 591 734
86 e e e 74 >5800 e 78
a
Affinities for cloned human dopamine receptors.

Scheme 14. Synthesis of protoberberine derivatives of coumarin as antimicrobial


Fig. 22. Tetrahydropyridocoumarins as potential anticholinergic bronchodilators. agents.

11
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Scheme 16. Synthesis of pyridinocoumarin as antibacterial and antifungal agents.

additional nitrogen atom (47c-e) in the amine substitution of


thiophene ring, only compound 47d considerably expanded the
spectrum of the activity against more strains (such as P. aeruginosa,
S. marcescens, S. aureus), comparable to that of 47c and 47e. The
order of antibacterial activity against P. aeruginosa was
Fig. 24. Structure and biological activities pyridinocoumarins. 47d > 44 ¼ 45 > 47c > 46 ¼ 47a ¼ 47b ¼ 47e. The high antibacterial
activity of the compound 47d could be possibly due to the presence
of key sulfonamide pharmacophore.
Furthermore, among the prepared compounds, carbohydrazide
derivative (45) bearing hydrazone-substituted thiophene moiety
displayed considerable antifungal activity, whereas amino-
thienocoumarin carboxylate (44) with ester-substituted thiophene
and chloroacetyl amino derivative (46) with amide-substituted
thiophene, showed relatively low antifungal activity. The valuable
antifungal activity of compound 45 could be possibly attributed to
the pivotal role of hydrazide motif as versatile building block of
various antifungal agents [54]. Among compounds possessing
aminoacetyl derivatives (47a-47e) obtained from the nucleophilic
substitution of chloroacetyl amino compound (46), only compound
47a was the most active compound. The order of antifungal activity
was 45 > 47a > 44 > 46 > 47e¼ 47b¼ 47d > 47c.
Scheme 15. Synthesis of pyridinocoumarin derivatives as antimicrobial agents.
In another study, benzothieno[2,3-c]chromen-6-one derivatives
(50a-e) were synthesized via condensation of substituted phenols
(49) with compound 48 and subsequently photocyclization reac-
S. brevicaulis; while, addition of a chloroacetyl group reduced
antibacterial and antifungal activities of the compound (46) con- tion (Scheme 8) [55]. The antibacterial activity assay of benzothieno
[2,3-c]chromen-6-one derivatives (50a-e) against S. aureus, E. coli,
firming the important role of the free amine group of thiophene
ring on the antimicrobial activity of the target compound. This may B. subtilis, and S. typhosa showed the relationship between the
inhibitory effect and concentration of the compounds. The tested
be due to the effect of the amine groups producing positive charge
leading to the strongly attachment of the compound to the nega- compounds exhibited a much higher inhibitory effect on the
growth of bacteria at higher concentrations. The best inhibitory
tively charged membrane of bacteria [53]. Compound 46 was just
active against S. aureus and C. albicans, less than the mother com- effect on the growth of bacteria was obtained using 5 mg/mL of
compound 50a (R ¼ H and R1 ¼ NO2) against E. coli and S. typhosa
pound (44). Nucleophilic substitution reaction of compound 46
with aromatic amines resulted in the formation of arylamino acetyl (inhibition zone: 15e18 mm). The methyl-substituted compound
(50b, R ¼ H and R1 ¼ Me) also showed more inhibitory activity at
derivatives (47a-e). Although, compounds 47d and 47e exhibited
moderate antibacterial activity against S. aureus, compounds 47a, 5 mg/mL against B. subtilis compared with other bacterial species.
Presumably, the electronic effects of the substitution groups were
and 47b showed no antibacterial activity against all the tested
species of bacteria. Among arylaminoacetyl congeners possessing responsible for a better internalization of the compounds through

Table 7
Antibacterial and antifungal activities of compounds 93a-93g.

MIC (mg/mL)

Comp. No. R R1 R2 R3 1 2 3 4 5 6

93a H H H H 200 250 125 200 500 1000


93b H Me OMe H 100 62.5 250 200 500 1000
93c H Me H Benzo 62.5 200 250 250 500 1000
93d Me H H Benzo 100 125 62.5 100 >1000 1000
93e H Cl OMe H 200 62.5 200 250 500 >1000
93f H Cl H Benzo 62.5 200 62.5 100 1000 500
93g Me H OMe H 250 125 250 250 >1000 1000
Ampicillin 250 250 100 100 NT NT
Griseofulvin NT NT NT NT 100 500

Organisms: 1) S. aureus, 2) B. subtilis, 3) E. coli, 4) S. typhimurium, 5) A. niger, 6) C. albicans; NT: not tested.

12
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 8
Antibacterial and antifungal activities of compounds 96a-96l.

Comp. No. R1 R2 R3 R4 R5 Zone of inhibition (mm)

1 2 3 4 5 6 7 8

96a H H Cl H Cl 16 19 21 19 RD RD -ve -ve


96b OH H H Cl Cl 8 10 12 10 -ve -ve RD RD
96c OH Br H Cl Cl 21 20 19 22 -ve -ve -ve -ve
96d OH I H Cl Cl 16 8 18 14 RD RD þve þve
96e OH H OH H Cl 10 14 19 12 RD -ve RD RD
96f OH H Me Cl Cl 7 14 8 10 þve þve þve þve
96g OH Cl OH Cl Cl 18 22 22 20 -ve -ve RD -ve
96h OH I Me Cl Cl 15 12 9 12 þve þve þve þve
96i OH H H Cl OH 19 18 21 20 -ve -ve -ve -ve
96j OH Br H Cl OH 18 16 19 12 RD RD -ve RD
96k OH I H Cl OH 19 22 20 16 -ve -ve -ve -ve
96l OH I H Me OH 15 18 18 12 RD -ve -ve RD
Penicillin 24 24 22 22 NA NA NA NA
Nystatin NA NA NA NA -ve -ve -ve -ve

Organisms: 1) S. aureus, 2) B. subtilis, 3) E. coli, 4) S. typhimurium, 5) A. niger, 6) A. flavus, 7) F. moniliforme, 8) P. chrysogenum; þve, Growth; -ve: No growth; RD: reduced growth;
NA: not applicable.

Scheme 17. Synthesis of polyheterocyclic derivatives of pyridinocoumarin.

Scheme 19. Synthesis of pyridinocoumarin as an antimalarial agent.

Fig. 25. Polyheterocyclic derivatives of pyridinocoumarin as antibacterial agents.

Scheme 20. Synthesis of the pyridinocoumarins as anticancer agents.


Scheme 18. Synthesis of pyridinocoumarins as anti-inflammatory and analgesic
agents.

13
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Scheme 22. Synthesis of quinolinocoumarin derivatives.

1.3.2. RNA-binding activity


Fig. 26. Structure and pharmacological properties of pyridonocoumarins
Fukuzumi and co-workers performed a study on small mole-
cules binding to the precursor of miR-29a (pre-miR-29a) by the
fluorescent indicator displacement (FID) assay and subsequently
surface plasmon resonance (SPR) assay. The results suggested that
4H-thieno[3,2-c]chromen-4-one 51 (Fig. 14) could potentially be an
important structural motif to bind to pre-miR-29a and could be
considered as a suitable molecule for the detailed quantitative
structure-activity relationship (QSAR) studies [57].

1.3.3. Antiproliferative activity


Wittine et al. reported synthesis of thieno[3,2-c]coumarin de-
rivatives (53 and 54) from 4-hydroxycoumarin (32) (Scheme 9). The
prepared compounds were evaluated as an antiproliferative agent
against various human tumor cell lines including cervical carci-
Scheme 21. Synthesis of the pyridonocoumarin derivative.
noma (HeLa), colorectal adenocarcinoma-derived metastatic
(SW620), pancreatic carcinoma (MiaPaCa-2), breast epithelial
adenocarcinoma-derived metastatic (MCF-7) and hepatocellular
carcinoma (HepG2) as well as against normal diploid human fi-
broblasts (BJ) [58]. Antiproliferative activity of the coumarin[3,2-c]
thiophene hydroxamic acid derivatives (53a and 53b) against the
tested cell lines were more than coumarin[3,2-c]thiophene ureido
derivatives (54). The order of growth-inhibitory effects of the
hydroxamic acid derivatives against all the tested tumor cell lines
was 53b > 53c. In particular, the hydroxamic acid derivatives (53b
and 53c) exhibited high and selective antiproliferative effects on
HeLa cell lines with IC50 values of 8.84 and 9.47 mM, respectively.
Among coumarin[3,2-c]thiophene ureido derivatives, compound
54 showed remarkable selective cytotoxicity effect on Hela cell
lines (IC50: 9.43 mM).
Fig. 27. Pyridonocoumarin as fibrotic agent.

Fig. 28. Structure and biological activity of quinolinocoumarin.

the outer membrane of the Gram-negative or Gram-positive bac-


teria [56]. Scheme 23. Synthesis of quinolinocoumarin derivatives as anticancer agents.

14
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

1.4. Selenocoumarin derivatives

Selenium is essential for cell metabolism as a component of


glutathione peroxidase and other enzyme systems. The anticancer
effects of compounds bearing selenium were reported against
various cancer cell lines [59]. Two regioisomers of seleno-
coumarins, selenopheno[3,2-c] and [2,3-c]coumarins, exhibited
various pharmacological activities like antioxidant and anticancer
activities (Fig. 15) [60,63]. Fig. 29. Structure and biological activities of iminopyridinocoumarins

1.4.1. Anticancer and antioxidant properties


Two regioisomers of selenocoumarins (58e62) were prepared
from 3-bromocoumarin (55) or 4-(trifluoromethanesulfonyl)
coumarin (56) (Fig. 16) [60]. The compounds were then evaluated
for their cytotoxic activity against various tumor cell lines including
MES-SA (human uterus sarcoma), CCL-8 (mouse sarcoma), MCF-7
(human breast adenocarcinoma), MDA-MB-435s (human mela-
noma), HT-1080 (human fibrosarcoma), MG-22A (mouse hepa-
toma) and normal cell line of NIH3T3 (mouse fibroblasts). The
Scheme 25. Synthesis of iminopyridinocoumarin as an antibacterial and antifungal
potency of the compounds for matrix metalloproteinases inhibition agent.
was also investigated [60]. Most of the compounds showed no
significant toxicity against all the cell lines. However, compound 60
showed more cytotoxic activity against MES-SA cells (IC50: 9.4 mM) increased. Selenolocoumarin functionalized with morpholine and
than sodium selenite as reference compound (Na2SeO3 IC50: 4-pyridyl amine group (63) having acceptable cytotoxic activity
29.0 mM). Selenophenocoumarins were much less toxic (LD50: against MCF-7 comparable with sodium selenite (IC50: 18.0 mM)
334e2000 mg/kg) than sodium selenite (LD50: 105 mg/kg). Among exhibited the highest selectivity for inhibition of MMP-2 (54%)
the different derivatives, a few compounds were able to scavenge compared to N-isobutyl-N-(4-methoxyphenylsulfonyl)-glycylhy-
free radicals. N-Methylpiperazyl aminomethyl derivatives (60 and droxamic acid as the standard inhibitor. Methylpiperazyl
61) showed mild peroxyl radical scavenging activity (IC50: 62.0 and substituted selenophenocoumarin (60) also inhibited angiogenesis
87.0 mM, respectively). Compound 62 (R ¼ CH2N(CH2CH2)2O, by 84% [60].
Ar ¼ 3-Pyridyl) was more active in peroxyl radical scavenging (IC50: In another study, the antiproliferative effect of the regioisomeric
4.0 mM) than compounds 60 and 61. This could be due to the high derivatives of selenocoumarins (60e62) was also screened against
potential of pyridyl or aminopyridyl group to improve the antiox- HepG2, MCF-7, MH-22A (mouse hepatocarcinoma) tumor cell lines
idant activity of the target compound [61,62]. Besides, by intro- (Fig. 16) [63]. The results showed that activity of the compounds
ducing the aminopyridyl group in the selenopheno[3,2-c]coumarin, against tumor cell lines depended on their ability to induce
the inhibition of matrix metalloproteinase (MMP-2) was selectively apoptosis. The compounds inhibited antioxidant enzyme system
activities (SOD, GPx and TrxR) and could diminish the ROS con-
centration (LD50: 572e2000 and 334e2000 mg/kg for selenopheno
[3,2-c] (60) and -[2,3-c]coumarins (61), respectively). The com-
pounds showed selectivity regarding redox enzyme activity con-
firming the important role of ROS in the mechanism of antitumor
action.

2. Coumarin-fused five-membered rings with two


Scheme 24. Synthesis of quinolino[3,2-c]coumarin derivatives as an anticancer agent. heteroatoms

2.1. Pyrazolocoumarin derivatives


Table 9
Cytotoxic activity of compounds 129a-129m assessed by the MTT reduction assay.
The wide spectrum of biological activities of pyrazole as one of
Comp. No. R IC50 (mM) the outstanding heterocyclic compounds including antimicrobial,
Hela MCF-7 K562 LS180 antifungal, antioxidant, antiviral, anti-tuberculosis, anti-
hyperglycemic, anti-inflammatory, and antitumoral activities has
129a 2-NO2 29.8 58.6 25.4 >100
129b 2-Cl >100 >100 >100 >100
129c 2-OMe >100 >100 >100 >100
129d 3-OMe >100 >100 >100 >100
129e 2-Me >100 >100 >100 >100
129f 4-NO2 88.2 >100 43.8 >100
129g 3-Br 83.6 31.4 38.8 >100
129h 4-Br 47.3 46.2 47.5 >100
129i 3-Cl >100 71.7 50.7 >100
129j 3-Me >100 52.2 57.4 >100
129k 4-Cl 37.7 41.8 37.5 >100
129l 4-OMe 98.5 62 >100 >100
129m 4-Me 43.6 66.7 56.2 >100
DOX e 0.256 0.137 0.157 0.180 Fig. 30. Structure and biological activities of the benzofuro[3,2-b]pyridinocoumarins.

15
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Fig. 32. Pyrano[3,2-c]benzopyran-5-ones as antiproliferative agents.


Scheme 26. Synthesis of benzofuro[3,2-b]pyridinocoumarins as anti-inflammatory
and antimicrobial agents.

encouraged many researchers to fuse this heterocycle to coumarin


derivatives [64e71]. Pyrazolocoumarin derivatives comprising of
coumarin and pyrazole moieties exhibited antibacterial, antifungal,
and anticancer activities (Fig. 17) [72,75].

2.1.1. Antibacterial and antifungal activities


Mulwad et al. synthesized a series of pyrazolocoumarin de-
rivatives (65a-65b) from 3-formyl-4-hydroxycoumarin (64) and
evaluated their antibacterial activities against S. aureus as a Gram-
positive and S. typhi as a Gram-negative bacterial strain (Scheme
10) [72]. The study showed that the phenyl substitution on pyr-
azole ring (65b) decreased the antibacterial activity against both
the bacteria (MIC value of 65a and 65b: 100 and 200 mg/mL,
respectively). Antimicrobial agents perform their activity via
various mechanisms including inhibition of the cell wall, protein or
nuclei acid synthesis, inhibition of the metabolic pathways in
bacteria synthesis or depolarization of the cell membrane [73].
Pyrazole skeleton is one of the well-known lead structures
inspiring medicinal chemists to design new generation of antimi-
crobial agents. It has been reported that attachment of the phenyl
or benzyl groups as lipophilic substituents to the nitrogen atom of
the pyrazole ring mostly resulted in the more antibacterial activity
than small substituents like hydrogen and methyl [74]; however,
Mulwad et al. were obtained different results reversely. The reason
for the different results in the studies may be attributed to the Scheme 27. Synthesis of pyranocoumarin derivatives as anticholinesterase agents.
different action mechanism of the compounds which could be
interesting for the researcher in the field of medicinal chemistry to
study on the rational antimicrobial mechanism of pyr- Table 10
azolocoumarins. It has been also reported that there is a threshold The anticholinesterase activities of compounds 141e142.

of lipophilicity for bacterial membrane permeabilization improving Comp. No. R IC50 (AChE, mM) IC50 (BuChE, mM) Selectivity
the antimicrobial activity, but further increase in the hydropho- 141a 2-F 2.9 36 12.4
bicity results in a loss of antimicrobial activity [75]. It seems more 141b 3-F 2.8 >50 >17.8
quantitative structure-activity relationship (QSAR) should be 141c 4-F 0.038 1.84 48.4
considered by medicinal chemist to better perceive the role of the 141d 2-Cl 1.5 4.8 3.2
141e 2-Me 3.7 17.8 4.8
substations in the activity of the compounds.
141f 3-Cl 0.48 21.8 5.2
El-Dean et al. synthesized a series of pyrazolo[4,3-c]coumarins 141g 3-Me 1.9 5.5 2.6
(68 and 69) from 4- hydroxycoumarin (32) in 3 steps (Scheme 11) 141h 3,4-Cl 0.044 1.27 28.9
[52]. The antibacterial and antifungal activities of the prepared 141i 2,4-Cl 0.831 0.566 0.68
142a 2-F 1.8 41 22.8
compounds were investigated against different strains of bacteria
142b 3-F 2.0 >50 >25
(S. aureus, B. cereus, E. coli, P. aeruginosa, S. marcescens and M. luteus) 142c 4-F 2.17 2.17 1
and fungi (C. albicans, T. rubrum, A. flavus, F. oxysporum, S. 142d 2-Cl 1 11.2 11.2
142e 3-Cl 7.4 38.4 5.2
142f 3-Me 0.79 14.3 18.1
Donepezil 0.014 5.38 384.3

Selectivity of AChE: IC50 (BuChE)/IC50 (AChE).

brevicaulis, and G. candidum). The synthesized compounds (68 and


69) exhibited lower antimicrobial activity than chloramphenicol
and clotrimazole, as standard drugs for antibacterial and antifungal
assay, respectively. Compound 68 showed higher antifungal activ-
ity than compound 69. Although, compound 68 showed no
Fig. 31. Different pharmacological activities of pyranocoumarin derivatives.

16
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

antibacterial activity against all the bacterial species, compound 69


exhibited antibacterial activities against S. aureus, E. coli, and
P. aeruginosa. The results confirmed that the dominant mechanism
responsible for antibacterial and antifungal activities in this study
might be different for the compounds. The increase in the per-
centage of the nitrogen elements could be a reasonable explanation
of the improved antifungal activity of compound 68 rather than
compound 69. Improved electron density and more cationic Scheme 28. Synthesis of benzopyranocoumarin derivatives as anticancer agents.
structure could result in strong interactions with cell membrane
which could be one of the possible reasons for better antifungal
activity of compound 68 than compound 69.

2.1.2. Anticancer property


A series of substituted pyrazolocoumarins (73e75) were syn-
thesized via a two-step reaction including (1) the reaction of 4-
chloro-3-formylcoumarin (70) with phenylhydrazine hydrochlo-
ride (71) and (2) ring cyclization by pyridine with the aid of
piperidine as catalyst (Fig. 18) [76]. The anticancer activity of the
coumarinepyrazole hybrids (73) was investigated in human pros- Fig. 34. Structure and biological activity of dioxo-pyranocoumarin.
tate (DU-145), lung adenocarcinoma (A549), and cervical cancer
cell lines (HeLa) using MTT assay. Doxorubicin (DOX) and nocoda-
2.2.1. Anticancer property
zole were applied as standard drugs. Most of the compounds were
Han et al. synthesized a series of chromeno[3,4-d]imidazole-
inactive against the cancerous cell lines. The results revealed that
4(1H)-one derivatives (75) from compound 74 and evaluated their
compound 73a bearing para methoxy phenyl group was the most
anticancer activities on U87-MG, HCT116, and PC-3 cell lines
active compound against A549 (IC50: 27.0 mM) without any signif-
(Scheme 12) [80]. This study showed that among all the derivatives
icant cytotoxic activity against other cancerous cell lines (DU-145
having simple phenyl substituent attached to the N1, the cytotoxic
IC50: 175.0 and HeLa IC50: 190.0 mM). Compound 73b having methyl
activity of the pyridine-4-yl (75a) or quinolin-3-yl (75b) derivatives
group at meta position of the phenyl ring showed moderate cyto-
was more than other substituted compounds such as pyridine-3-yl,
toxic activity against A549 (IC50: 43.0 mM). None of the prepared
3-NH2-C6H4, 2-Me-C6H4, 4-OH-C6H4, and 3-NHSO2-(4-FeC6H4)e
compounds showed appropriate cytotoxic effect against DU-145
C6H4. When the substituent was changed from simple phenyl group
(IC50  100), except compound 73c and 73d having mild toxicity
to 4-CH2CN substituted phenyl group, the anticancer activity of the
(IC50: 53.0 and 60.0 mM, respectively).
target compound was slightly improved. Among this group, pyri-
dine-3-yl (75c) or quinolin-3-yl (75d) derivatives exhibited
2.2. Imidazolocoumarin derivatives appreciable cytotoxic activity against HCT116 cells (IC50: 29.37 and
28.4 mM, respectively), confirming the key role of the nitrogen atom
Imidazole plays an essential role in the development of new on the heterocyclic substituent as hydrogen acceptor site in
drugs in medicinal chemistry, especially for cancer treatment [77]. appropriate binding with the corresponding target. The SAR study
Owing to the valuable biological profiles of the coumarin and also demonstrated the important role of cyano group on the phenyl
imidazole scaffolds, hybridization of these interesting structures ring attached to the N-1, possibly as a hydrogen acceptor center
has encouraged researchers to prepare the compounds having improving the interaction of the compound with the corresponding
improved synergistic effects (Fig. 19) [78e80]. There is only one target. Among all the substituents attached to the N-1, 4-
report related to the anticancer activity of the imidazolocoumarins CH2CNeC6H4 (75c and 75d) and 4-C(CH3)2CNeC6H4 (75e and
and therefore various derivatives of this scaffold could be consid- 75f) showed the best performance in cytotoxic activity. Compound
ered as potential candidate for other biological activities. 75e bearing pyridine-3-yl substituent at C-6 position showed lower
anticancer activity against PC-3 cells (IC50: 38.20 mM) than com-
pound 75f with quinoline-3-yl substituent at the C-6 position (IC50:
19.76 mM). In addition, when compound 76 converted to compound
77, anticancer activity improved against HCT116 (IC50 value
changed from 20.40 to 1.40 mM). Compound 77 showed excellent
anticancer activity against a broad spectrum of the cell lines (IC50:
1.40, 3.55, 1.70, 3.50, and 9.50 mM against HCT116, SW620, MCF-7,
CT26, and A549, respectively). This research also revealed that
compound 77 bearing 4-C(CH3)2CNeC6H4 at N-1 position, methyl

Fig. 33. Structure and biological activity of benzopyranocoumarin. scheme 29. Synthesis of dioxopyranocoumarins as antibacterial agents.

17
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Scheme 32. Synthesis of pyrano[3,2-c]chromene-2,5-dione as antifungal agent.

condensation of b-ketoester 78 with aenaphthol (39) or m-cersol


(79), respectively, and evaluated the fungicidal activities against
Xanthomonas malvacearum, Fusarium manilliform, Rhizoctonia sol-
anis, Powdery mildew, Phytophthora and Gray mold (Scheme 13,
Table 5) [82]. Tetrahydrobenzopyridocoumarin derivatives (80)
showed higher activity than the series of tetrahydropyr-
idocoumarins (81). Compounds 80a and 81a having unprotected
amine group (R ¼ H) showed the highest activity among all the
Scheme 30. Synthesis of dioxopyranocoumarin derivatives used as antibacterial
agents.
compounds against most of the fungi. N-Acetylation of compound
80a led to a significant decrease in the fungicidal activity (80b). N-
Acetylation of compound 81a also resulted in a significant decrease
group at N-3 position, tert-butoxy and methyl groups at C-2 of the in the antifungal activity of compound 81b against Fusarium man-
imidazole moiety, and quinoline-3-yl at the C-6 position of illiform and Rhizoctonia solanis. Furthermore, N-methylation led to
coumarin was the best anticancer agent. The flow cytometry the lower activity of compounds 80c and 81c against Powdery
experiment demonstrated that compound 77 could arrest the cell mildew and Phytophthora infection than their analogs (80a and 81a).
cycle of SW620 at G0/G1 and induce apoptosis in a concentration- The results clearly confirmed the crucial role of the free amine on
dependent manner. Further in vivo anticancer experiments on antifungal activity of the target compounds. It has been suggested
subcutaneous HCT116 tumor model in nude mice showed that that the free NH is essential for the high potency of the compounds,
compound 77 could inhibit tumor growth by 52.96% at 80 mg/kg/ while the protection of NH by groups changing the electronic
48 h after 20 days. contribution of the nitrogen and tetrahydropyridin part decreases
the antifungal activity. The presence of methyl substituent at po-
3. Coumarin-fused saturated six-membered ring sition 8 of the coumarin moiety (81a) showed lower potential to
improve antifungal activity than the 7,8-benzocoumarin analogs
3.1. Tetrahydropyridocoumarin (80a) indicating the key role of the lipophilic phenyl group resulting

Tetrahydropyridine scaffold is found in numerous natural and


synthetic compounds exhibiting interest pharmacological proper-
ties such as neuroprotective effect [81]. Tetrahydropyrido[2,3-c]
coumarin derivatives are considered as potential antifungal, anti-
psychotic, and bronchodilator agents (Fig. 20) [82e84].

3.1.1. Antifungal activity


Mandal et al. synthesized a series of substituted tetrahydro[3,4-
c] and benzo[h]tetrahydro pyrido[3,4-c] coumarins via

Scheme 31. Synthesis of dioxopyranocoumarin derivative. Fig. 35. Structure and biological activity of pyranoquinolinocoumarins.

18
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

in the more penetration via permeation mechanism and subse-


quently more activity (see Table 6).

2.1.2. Antipsychotic activity (dopamine D4 receptor antagonists)


Pyridine-fused coumarin derivatives have been synthesized
from different phenols and beketoesters and introduced as selec-
tive human dopamine D4 receptor antagonists (Fig. 21) [83]. The
affinity of the synthesized tetrahydrochromeno[3,4-c]pyridin-5-
ones (82) towards the dopamine D2, D3, and D4 receptors have
been determined in vitro by assessing their ability to displace the
dopamine ligand [3H]-spiperone from cloned human dopamine
receptor subtypes expressed in Chinese hamster ovary (CHO) K-
1 cells. The type of the substituents at different positions of the
phenyl or benzyl parts and the length of the side chain affected
significantly receptor binding activity of the target compounds. The
binding activity decreased by the elimination of the methylene side
chain (83a compared with 84) or replacement of the phenyl group
with pyridine (83a compared with 86). Compound 85 possessing Fig. 36. Structure and biological activity of dihydropyranopyrrolocoumarin
additional carbon in the chain length showed improved selectivity derivatives.
for D4 versus D2 receptor in comparison to compound 83a. The
ability of the best active compounds to block stimulation of mito-
genesisintrinsic activity in transfected CHO 10001 cells with DA D4
receptors was also investigated to study the intrinsic activity of the
compounds in comparison with quinpirole as a full agonist of
dopamine. The compounds having 20% or less intrinsic activity
(83b, 83g, 83k, and 83m) were antagonists, while compounds
showing more than 20% intrinsic activity (83a, 83c, 83e, 83f, 83i,
and 85) were partial agonists.

3.1.3. Bronchodilatory activity


A series of 3-substituted benzopyrano[3,4-c]pyridin-5-ones Scheme 34. Synthesis of dihydropyranopyrrolocoumarin derivatives as antimicrobial
were synthesized and introduced as a potential anticholinergic agents.

bronchodilator confirmed by several in vivo studies (pig and dog


models) (Fig. 22) [79]. Among various derivatives of 87 bearing
various amine groups attached to the carbon a (*), the most potent 4. Protoberberine derivatives (polycyclic coumarin-based
bronchodilators were compounds containing the 3-azabicyclo compounds)
[3.2.2]non-3-yl and methoxy groups (87a and 87b ID50: 35.0 and
113.0 mg/kg for dog model, respectively). The results showed that Protoberberines, a member of alkaloids, have attracted consid-
the steric hindrance in the carbon a position is critical for the su- erable attention over the decades because of the important bio-
perior bronchodilator activity. Addition of a methyl group on the logical activities and their presence in many traditional medicines
carbon a improved often the potency and pulmonary selectivity of as active components [85e89]. Coumarin analogs of proto-
the compound. Compound 88a without substituent on coumarin berberine exhibited antibacterial and antifungal activities (Fig. 23)
ring and having noncyclic amine group showed the highest pul- [90].
monary selectivity (21.6).

4.1. Antibacterial and antifungal activities

Jadhav et al. prepared a series of protoberberines (90a-f) from


coumarin-4-acetic acid 89 via five-step reaction including (1)
esterification, (2) amidation with 2-phenethylamines, (3) Bischler-
Napieralski, (4) reduction and (5) Mannich reactions (Scheme 14)
[90]. The results of the gel electrophoresis technique for DNA
cleavage assay showed that all the tested compounds (90a-f) were
inactive against E. coli, but compounds 90e and 90f bearing 7,8-
benzo substituent on the coumarin moiety were capable to cleav-
age DNA of S. aureus and A. niger. It may be due to the presence of
the angularly fused cyclic skeleton preventing the growth of the
pathogenic organism. This study confirmed the critical role of the
additional benzene ring fused to the coumarin skeleton in cleavage
of the genomic DNA of the bacteria as well as its effect on the lip-
ophilicity of the system leading to more penetration of the target
compounds via permeation mechanism and subsequently more
antibacterial activity.
Scheme 33. Synthesis of pyranoquinolinocoumarin derivatives.

19
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Fig. 37. Structure and biological activity of pyranopyrimidino coumarin derivatives.

5. Coumarin-fused unsaturated six-membered heterocycle


with one heteroatom
Scheme 36. Synthesis of pyranopyrimidinocoumarin derivatives as antimicrobial
agents.
5.1. Pyridinocoumarin derivatives

Pyridine and its derivatives are important chemical compounds Table 11


Antibacterial activity of some pyranopyrimidinocoumarin derivatives.
because of their diverse range of biological applications in different
fields [91]. Pyridinocoumarin core produced by the fusion of Comp. Inhibition zone (mm)
coumarin and pyridine parts exhibited a wide range of biological 1 2 3 4 5 6 7 8 9 10 11 12 13
activities [92] such as antimicrobial [93e95], anti-inflammatory a
160a 28 11 19 10 20 10 e e 12 11 20 e e
and analgesic [96], antimalarial [97], anticancer (Fig. 24) [98]. 160bb 18 e 19 e e e 18 20 e e e 16 18
166aa 22 18 16 10 13 12 e e 21 12 20 e e
167aa 29 22 18 18 21 20 e e 14 16 18 e e
5.1.1. Antibacterial and antifungal activities 167bb 21 e 22 e e e 20 19 e e e 14 12
A series of pyridine-fused coumarin derivatives 93a-g were 169bb 18 e 17 e e e 19 19 e e e 19 15
synthesized via Mannich-type reaction between pyridinium salts 170ab 20 e 22 e e e 21 20 e e e 18 19
170bb 20 e 22 e e e 20 19 e e e 19 17
(91) and 4-hydroxycoumarin derivative (92) or 4-chloro-3- 170cb 23 e 22 e e e 23 22 e e e 19 19
formylcoumarin (70) and evaluated for their antibacterial and 170db 23 e 22 e e e 25 23 e e e 19 20
antifungal activities against Gram-positive bacteria (B. subtilis and 171b 20 e 22 e e e 21 22 e e e 20 19
S. aureus), Gram-negative bacteria (E. coli and S. typhimurium) and Amoxicillin 29 12 20 11 36 10 e e e e e e e
Ampicillin 26 e 25 e e e 26 27 e e e e e
fungi (A. niger and C. albicans) (Scheme 15) [93]. The results showed
Mycostatin e e e e e e e e 12 20 26 22 24
that the antimicrobial potency of the compounds strongly depen-
ded on the pattern of the substituents (Table 7). All the compounds Organisms: 1) S. aureus, 2) B. subtilis, 3) B. cereus, 4) P. aeruginosa, 5) E.coli, 6)
E. aerogenes, 7) S. marcescens, 8) P. merabitis, 9) A. niger, 10) P. italicum, 11)
exhibited significant activity against Gram-positive bacteria (MIC F. Oxysporum, 12) A. ochraceus Wilhelm, 13) P. chrysogenum Thom.
250 mM) compared with ampicillin as the reference drug (MIC: a
Amoxicillin was used as the reference compound.
250 mM). Introduction of methoxy as electron releasing group and b
Ampicillin was used as the reference drug.
methyl as lipophilic part (93b or 93g) improved the activity of the
compounds against Gram-positive bacteria compared with simple
derivative (93a). Also, benzo and chloro groups as lipophilic could (95), compounds 96c, 96i and 96k showed equipotent antifungal
significantly improve the antimicrobial activity of the target com- activity as compared to nystatin against all fungi including A. niger,
pounds (93d-93f). The compound bearing both chloro substituent A. flavus, F. moniliforme, P. chrysogenum with MIC value of 100 mg/mL
on the phenyl ring and additional benzene ring in coumarin scaf- (Scheme 16, Table 8) [94]. Compounds 96c and 96g exhibited good
fold (93f) exhibited the highest activity towards both E. coli and inhibitory effect as potent as penicillin against S. typhi and E. coli,
S. aureus (MIC: 62.5 mg/mL), as compared to the standard drug, respectively. Compounds 96g and 96k exhibited better antibacte-
ampicillin. Furthermore, compounds 93g showed better activity rial profile against B. subtilis than the other compounds and pro-
(MIC: 200 mg/mL) than griseofulvin as reference drug (MIC: 500 mg/ duced an inhibition zone similar to that of Penicillin. Compound
mL) against fungal pathogen C. albicans. 96g having 1,3-di-hydroxyl and 2,4,5-tri-chloro groups in the
In another study, among different derivatives of pyridocoumarin
having pyrazole moiety (96a-96l) synthesized by the reaction of 4-
hydroxycoumarin derivative (94) with a,beunsaturated ketones

Scheme 35. Synthesis of pyranopyrimidino coumarin derivatives. Fig. 38. Structure and biological activities of spirooxindolocoumarin derivatives.

20
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

compound. The order of the analgesic activity was similar to the


order of the anti-inflammatory potency. Also, compound 103d
displayed lower toxicity than diclofenac potassium as the standard
drug (LD50: 2368 and 1988 mg/kg, respectively).

5.1.3. Antimalarial activity


Gorlitzer et al. synthesized pyridine-fused derivative of
coumarin (106) from compound 104 and N,N-diethyl-1,4-
pentadiamine (105) and evaluated its antimalarial activity. Com-
pound 106 showed the antimalarial activity against the
chloroquine-resistant plasmodium falciparum strain Dd2 with IC50
value of 1100 nM (IC50 for chloroquine as reference drug: 140 nM)
Scheme 37. Synthesis of spirooxindolocoumarin derivatives as antibacterial and (Scheme 19) [97].
anticancer agents.

5.1.4. Anticancer activity


Various pyridinocoumarin derivatives were synthesized from
phenyl ring displayed higher antibacterial activity than the other the reaction of 4-aminocoumarin (107) with various a,b-unsatu-
compounds. rated carbonyl compounds (108e111). Anticancer activity of the
In another study, polyheterocyclic derivatives of pyr- prepared compounds (112e115) was evaluated against Ehrlich As-
idinocoumarin were synthesized through the reaction of 3- cites Carcinoma (EAC) cells (Scheme 20). The results showed
aminopyrazol-5-ones (97) with salicylaldehydes (98) and acetyl- acceptable antitumor activity of the pyridinocoumarin derivatives
acetic ester (99). The prepared compounds (100) showed antibac- (112e115) against EAC cells (IC50: 0.12e0.13 mM) which was more
terial activity against different Gram-positive strains (S. epidermidis than 5-fluorouracil as the standard drug (IC50: 0.38 mM) [98]. The
and S. aureus) (Scheme 17) [95]. Compound 100a exhibited the high cytotoxic activity of the compounds could be ascribed to the
most activity against S. epidermidis and methicillin-resistant great effect of the pyridine, quinolone, or indandione ring through
S. aureus (MIC: 6.3 and 25.0 mM, respectively) and compound various mechanisms like growth inhibition, apoptosis, or
100b displayed significant antibacterial property against
S. epidermidis (MIC: 25.0 mM) (Fig. 25).

5.1.2. Anti-inflammatory and analgesic activities


Hosni et al. synthesized benzopyranopyridine derivatives (103)
via Michael reaction of compound 101 with malononitrile (102)
followed by hydrolysis. The anti-inflammatory and analgesic ac-
tivities of the products were tested (Scheme 18) [96]. It was
revealed that both pyridine nucleus and five-membered ring
bearing one heteroatom in furanyl benzopyranopyridine de-
rivatives were essential for anti-inflammatory and analgesic activ-
ities. Among the tested compounds, compound 103d showed high
protection against induced edema, more than 89%, with high per-
centage of the plasma prostaglandin (PGE2) inhibition, comparable
with diclofenac as the reference drug (more than 62%). The degree
of anti-inflammatory potency was 103d > 103a > 103b > 103c. The
analgesic activity of the compounds on adult male albino rats
confirmed the high potency of compound 103d which was nearly 2
times more than diclofenac as the standard anti-analgesic

Fig. 39. Structure and biological activities of pyrimidinocoumarin derivatives. Scheme 38. Pyrimidinocoumarins as anticancer agents.

21
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

5.2. Pyridonocoumarin derivatives

Pirfenidone (PFD, 116), 5-methyl-1-phenylpyridine-2(1H)-one,


is used for the treatment of Idiopathic Pulmonary Fibrosis (IPF)
(Fig. 26). The 2-pyridone scaffold is ubiquitous in many natural and
synthetic compounds displaying oncogenic properties. 5-Pyridyl-2-
oxopyridine derivatives such as milrinone (117) and amrinone (118)
have also shown great potential for the treatment of congestive
heart failure [102e104]. The valuable features of pyridine-2(1H)-
one framework together with the interesting biological activity of
Scheme 39. Synthesis of pyrimidine-fused coumarin derivatives as anticancer agents. the coumarin derivatives encouraged many medicinal chemists to
design and prepare new multicyclic compounds bearing both
scaffolds. Pyridonocoumarin derivatives have been screened for
their biological activities and some of them exhibited anticancer
angiogenesis inhibition on the anticancer activity of the target
and fibrotic potency [105,106].
compounds as reported previously [99e101]. All the compounds
had similar cytotoxic activity and the electronic properties of the
substituents on the pyridine moiety had no effect on the activity of 5.2.1. Anticancer property
the products. El-Said et al. reported a series of pyridinocoumarin as PFD an-
alogs anticancer derivatives against EAC cell lines (Scheme 21)
[105]. They found that cyclization of compound 119 at positions 3
and 4 of the coumarin ring with malononitrile (102) resulted in a
reduction of pyridinocoumarin 120 activity (IC50: 245.7 mM).
Compound 120 had no cytotoxic effect against EAC cell lines
compared to positive control doxorubicin (IC50: 68.13 mg/mL).

5.2.2. Fibrotic activity


Despite the reported anti-fibrotic activity of PFD alone, the
annulation of PFD to coumarin ring (121) led to an increased fibrotic
potential on lung fibrosis in the rat model (Hydroxyproline
content ¼ 193.07 mmol/Lung) and toxicity on liver and kidney
functions (Fig. 27) [106]. Also, compound 121 showed a reduction
in LDH (lactate dehydrogenase) activity, protein concentration, and
total cell count (macrophages, neutrophils, and eosinophils)
compared with PFD alone.

5.3. Quinolinocoumarin derivatives

The diverse pharmacological properties of quinoline and its


derivatives have attracted more attention to employ this scaffold in
the design of new multicyclic heterocycles. quinoline derivatives
fused with various heterocycles have displayed wide pharmaco-
Scheme 40. Synthesis of pyrimidinocoumarin derivatives as antithrombotic agents. logical activity such as anticancer, antimycobacterial, antimicrobial,

Table 12
In vitro antiplatelet potency of compounds 183, 185e157 against arachidonic acid (AA), U46619, and ADP induced aggregation in rabbit PRP and inhibitory potency against
thrombin induced clot retraction in rat PRP.

Comp. No. R R0 Inhibition of platelet aggregation IC50 (mM) Inhibition of clot retraction IC50 (mM)

ADP 3 mM AA 50 mM U46619 1 mM Thrombin 20 U/mL

183a e OMe e e e e
183b e SMe e e e e
183c e Pyrrolidino e e e e
183d e Morpjolino e e e
185a NH2 H e e e e
185b NH2 NH2 e e e e
185c NH2 OMe e e e e
185d NH2 SMe e e e e
185e NH2 Pyrrolidino e e e e
185f NH2 Morpjolino e e e e
186a SMe Pyrrolidino e e e e
186b SMe Piperidino e e e e
186c SMe Morpjolino e e e
187a Pyrrolidino SO2Me 13 15 4 13
187b Piperidino SO2Me 18 16 21 27
187c Morpjolino SO2Me 16 21 3 37
Aspirin - e >500 61 e e

22
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Fig. 42. Structure and biological activities of thiazepinocoumarin.


Fig. 40. Structure and biological activity of imidazopyrimidinocoumarin derivatives.

(R1 ¼ 4-Me and R2 ¼ 3-OMe-C6H4) showed the most activity against


anticonvulsant, anti-inflammatory, and cardiovascular activities A-549 cell lines (IC50: 0.05 mM). However, compound 126c was
[107]. There is just one report related to the biological activity of inactive against both the cell lines (IC50 > 100.0 mM). Compound
benzopyranoquinolinone scaffold, a class of heterocycles produced 126a was also oxidized by 2,3-dichloro-5,6-dicyano-1,4-
from combination of coumarin and quinoline moieties (Fig. 28) benzoquinone (DDQ) resulting in the formation of a pyridine ring
[108e110]. (127). However, the aromatization process led to a significant
decrease in the cytotoxic activity (IC50 > 100.0 mM). The order of
5.3.1. Anticancer activity anticancer activity against MCF-7 was 126a > 126b > 126c > 126d
A series of 6H-1-benzopyrano[4,3-b]quinolin-6-ones (123) were and also against A-549 was 126b > 126a > 126c > 126d.
synthesized via the reaction of 4-chloro-3-formylcoumarin (70) In another study, 7H-chromeno[4,3-b]quinoline-6,8-dione de-
with different anilines (122) (Scheme 22) [108]. The anti- rivatives (129a-129m) were synthesized from 1,3-cyclohexadione
proliferative property of the products was evaluated in vitro (128), benzaldehydes (125), and 4-hydroxycoumarin (32), and
against four human cancer cell lines including myeloid (K562), their cytotoxic activity were evaluated against four different human
breast (MDA-MB 231), colon (Colo-205), and neuroblast (IMR32). cancer cell lines; HeLa (human cervical adenocarcinoma), K562
Compounds 123a, 123b, 123c, 123g, 123h, 123i and 123j exhibited (human chronic myelogenous leukemia), LS180 (human colon
good activity against MDA-MB 231 (IC50 ~ 8.0e16.0 mM), and adenocarcinoma), and MCF-7 (human breast adenocarcinoma)
compounds 123a, 123c, 123g and 123j were active against Colo-205 (Scheme 24) [110]. The tested compounds showed weak activity
(IC50~ 11.0e18.0 mM). Among all the tested compounds, compound against LS180 cells (IC50 > 100.0 mM) and mild activity against Hela,
123c was the most active compound against Colo-205 and MDA- K562, and MCF-7 cell lines (IC50: 25.0e100.0 mM) compared to the
MB 231 cell lines with IC50 values of 11.0 and 8.0 mM, respec- DOX as the reference drug (IC50 < 0.26 mM) (Table 9). The results
tively. All the tested compounds showed moderate activity against demonstrated that the substituent group on the phenyl ring
IMR32 (43.0 > IC50 > 20.0 mM), and were more active than hermine affected the cytotoxic activity significantly. For example, compound
as reference compound (IC50 > 45.0 mM). Compound 123e showed 129a with a nitro group at the ortho position of the phenyl ring
significant activity against IMR32 (IC50: 23.0 mM). It has been sug- showed the highest activity against K562 and Hela cell lines (IC50:
gested that sirtuin inhibition activity could be the possible mech- 25.0 and 29.0 mM, respectively), while compounds 129b-129e were
anism of anticancer activity of the compounds against Sirt1. the weakest derivatives against all four human cell lines
Compound 123d showed 48% inhibition of Sirt1, much less than (IC50 > 100.0 mM). Also, calcium channel antagonist activity of the
suramin as standard drug having 79% inhibition at 10 mM. tested compounds was evaluated and only 3-Cl substituted com-
A family of 7,12-dihydro-6H-chromeno[4,3-b]quinoline de- pound (129i) exhibited 3 folds stronger activity (IC50: 599 nM) than
rivatives (126) was prepared using 4-hydroxycoumarin (32), aro- the other derivatives which was nearly 7 folds weaker than nifed-
matic amines (124), and aldehydes (125) (Scheme 23) [109]. The ipine (as the reference drug, IC50: 91.4 nM).
antitumor activity of the products was investigated against A-549
and MCF-7 cell lines using convenient MTT assay. Some of the 5.4. Iminopyridinocoumarin derivatives
compounds were stronger than cisplatin as the reference drug
against both the cell lines. Among tested compounds, compound Iminopyridine complexes such as osmium (II) arene iminopyr-
126a (R1 ¼ 4-Me and R2 ¼ Ph) was the most active compound idine complexes have gained much interest as strong anticancer
against MCF-7 cells with IC50 value of 0.52 mM, and compound 126b agents having IC50 values in nanomolar range [111]. Iminopyr-
idinocoumarin is a class of the fused coumarin scaffolds, which
exhibited valuable pharmacological properties such as antibacterial
and antifungal activities (Fig. 29). The synthesis and biological ac-
tivities of this valuable compound are discussed below [112].

5.4.1. Antibacterial and antifungal activities


Iminopyridinocoumarin derivative (132) was prepared from
dimethylaminoethylene derivative 130 and 2-aminopyridine (131).
Compound 132 showed strong antibacterial and antifungal activ-
ities against B. subtilis, S. aureus, E. coli, and A. niger compared with
streptomycin and mycostatine (MIC: 50 mg/mL) as the references
(Scheme 25) [112]. Although no study was performed or suggested
Fig. 41. Structure and biological activity of pyramidopyrimidinocoumarin for the appropriate antimicrobial effect of the target compound
23
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

exhibiting valuable antimicrobial and anti-inflammation properties


(Fig. 30) [122].

5.5.1. Anti-inflammatory and antimicrobial activities


Khan et al. synthesized polycyclic heterocycles 135 having
benzofuranopyridine and coumarin rings from various 4-
bromomethylcoumarins 133, salicylonitrile 134, and orthoesters
via a three-step reaction including (1) allylic substitution, (2)
intramolecular condensation and (3) ring closure reaction. The
authors evaluated anti-inflammatory and analgesic activities of the
Scheme 41. Synthesis of pyrimidinocoumarin derivatives as antibacterial agents.
products (Scheme 26) [122]. LD50 values of the prepared com-
pounds were more than 800 mg/kg body weight. Among the tested
(132), it seems that the presence of nitrogen-rich heterocycle and group, compound 135a showed significant anti-inflammatory ef-
free amine controlling the charge and subsequently affinity of the fects at 200 and 300 mg/kg dose level (81% and 89% inhibition,
compound to the cell membrane and also ion chelation ability of respectively). The evaluation of the analgesic activity of the com-
the compound play key roles in the antimicrobial activity. pounds by acetic acid-induced abdominal constriction revealed
that compound 135b was the most potent compound with 78%
inhibition of inflammation at 300 mg/kg dose level (maximum
5.5. Benzofuro[3,2-b]pyridinocoumarin derivatives
possible effect ¼ 24.4%). Also, all the tested compounds exhibited
antimicrobial activity against Pseudomonas cichorii (MIC: 25 mg/
Benzofuran scaffolds have drawn much attention due to their
mL), and no activity was observed against Micrococcus aureus. All
wide range of biological activities and potential applications as
the compounds could also inhibit the growth of Aspergillus fumi-
pharmacological molecules. Diverse biological activities of benzo-
gatus at 25 mg/mL and Penicillium wortmannin at a concentration of
furan derivatives have been reported in the literature including
100 mg/mL.
antimicrobial [113], analgesic, anti-inflammatory [114], antipara-
sitic [115], antihyperglycemic [116], antitumor, and kinase inhibi-
tion activities [117]. 5.6. Pyranocoumarin derivatives
Also, pyridine derivatives possess exciting antibacterial, anti-
viral, antioxidant, anti-diabetic, anti-cancer, anti-malarial, anal- Pyran derivatives are the nucleus of many compounds with
gesic potency, anti-amoebic, enzyme inhibition, anti-inflammatory wide biological and pharmacological properties such as anticancer,
activities [118]. Studies on the biological activities of the benzo- anti-microbial, antifungal, anti-osteosarcoma, and antihistaminic
furano[3,2-b]pyridines have shown interesting biological activities activities [123]. Also, pyrano-fused heterocycles are an important
[119e121] inspiring researchers to fuse this valuable heterocycle to class of structural motif in many natural products and synthetic
other biologically active frameworks. The fusion of benzofur- drugs with antifungal, insecticidal, anticancer, anti-HIV, anti-in-
anopyridine unit with coumarin gave rise to a pentacyclic system flammatory, and antibacterial activities [124,125]. Different

Scheme 42. Synthesis of pyrimidin-annulated coumarin derivatives.

24
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

nanomolar concentration (IC50: 0.038 mM) and higher BuChE/AChE


selectivity (Sl ¼ 48.4). The docking study demonstrated that pyr-
anocoumarin part of the compound 141c interacts with the cata-
lytic active site (CAS) of the AChE and N-benzyl pyridinium part
interacts with the peripheral anionic site (PAS) of the enzyme.

5.7. Benzopyranocoumarin derivatives

Scheme 43. Synthesis of imidazopyridinocoumarin as an anticancer agent.


Benzopyran ring is an important group of organic compounds
displaying pharmacological properties such as antimicrobials,
antitumor, anti-inflammatory, anticancer, anti-HIV, and anticoag-
ulants activities [126]. There is only one report related to the bio-
logical activity of this group of compounds (Fig. 33) [127].

5.7.1. Anticancer activity


Benzopyrano[3,2-c]chromene-6,8-diones (143) were synthe-
sized through three-component reaction between 4-
hydroxycoumarin (32) to 1,3-cyclohexadione (128) and benzalde-
Scheme 44. Synthesis of pyrimidopyrimidinocoumarin as an anticancer agent. hydes (125) (Scheme 28) [127]. In vitro cytotoxic studies of the
compounds were performed against four different human cancer
cell lines (Raji, HeLa, LS180, and MCF-7). According to the results, all
the compounds showed weak-to-moderate anti-tumor activities
with IC50 values ranging from 49.0 to more than 100 mM. No
meaningful relationship was observed between the effect of the
substituents and the cytotoxic effect of the compounds. Among the
prepared compounds, derivatives containing methyl group in po-
sition 2, 3, or 4 of the phenyl ring showed the weakest activities
(IC50 > 100 mM) against all the cell lines except for Raji cells with
Scheme 45. Synthesis of thiazepine-annulated coumarins.
IC50 value of 73e80 mM. The results also revealed that the presence
of methoxy group, especially at the meta position of the phenyl ring
pharmacological activities have been described for the compounds in compound 143 (R ¼ 3-OMe), resulted in partially more cytotoxic
bearing the pyrano[3,2-c]chromene-5(4H)-one nucleus (Fig. 31) activity against all the cell lines (IC50: 49e99 mM). Owing to the
[124,125]. high potential of furocoumarin scaffolds to bind to the DNA re-
ported previously, it could be assumed for the target compounds
(143) to have the possible ability for interaction with nucleobases in
5.6.1. Antiproliferative property DNA causing the cytotoxic effect and possibly the presence of
A series of halogenated 2,4-diaryl-4H,5H-pyrano[3,2-c]benzo- electron donor group improved the interaction [128].
pyran-5-ones (136) were prepared and their anticancer activity was
evaluated against MCF-7 cell lines. All compounds showed a 5.8. Dioxo-pyranocoumarin derivatives
remarkable antiproliferative effect against MCF-7 (Fig. 32) [124]. All
of the compounds were more potent than the reference drug ful- The pyranone (pyron) scaffold as one of the most abundant
vestrant (ICI 182780, Estrogen receptor antagonist). Nevertheless, heterocycles in naturally occurring organic compounds exhibited
all the compounds had no appropriate estrogenicity or affinity to- various biological activities such as antitumor, anti-inflammatory,
wards recombinant human estrogen receptors alpha protein. antibacterial, antityrosinase, and anti-HIV activities [129]. The
fusion of 2H-pyran-2-one ring to the coumarin ring produced a
5.6.2. Anticholinesterase activity series of pyrano[3,2-c]chromene-2,5-dione (dioxo-pyr-
Pyranocoumarin derivatives have been introduced as a suitable anocoumarin) reported as antibacterial and antifungal agents
pharmacophore for the preparation of novel cholinesterase in- (Fig. 34) [130e132].
hibitors (ChEIs). Two series of pyrano[3,2-c]chromene derivatives
bearing N-benzyl pyridinium part (140) were synthesized in the 5.8.1. Antibacterial activity
presence of magnetic nanocatalyst via a three-step procedure Tetrazoles have exhibited a wide range of biological and phar-
including (1) reaction between pyridine-3 or 4-carbaldehydes (137) macological properties such as antibacterial, antifungal, anticancer,
and malononitrile (103), (2) reaction with 4-hydroxycoumarin (32) antiviral, antimalarial, and anti-tubercular activities. This valuable
and (3) the reaction of compound 138 with benzyl chlorides (139) heterocycle are considered as one of the key pharmacophore
(Scheme 27). Then, the products (140) were evaluated against encouraging medicinal chemist to employ it in the drug design and
acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) discovery [133]. A series of pyrano[3,2-c]chromene-2,5-dione de-
(Table 10) [125]. Both derivatives showed significant inhibitory rivatives having tetrazole moiety (146) were synthesized via a
activity against AChE with IC50 values less than 7.4 mM. The study three-step reaction including (1) Knoevenagel condensation be-
revealed that anti-ChE activity and AChE/BuChE selectivity index tween compound 144 and ethyl cyanoacetate (ECA), (2) cyclization
strongly depends on the type and position of substituent on the by polyphosphoric acid (PPA), and (3) 1,3-dipolar reaction between
benzyl pendent. In term of the AChE inhibitory activity, 3- compound 145 and NaN3 (Scheme 29) [130]. Antibacterial activity
pyridinium analogs were more active than 4-pyridinium analogs of the products (146) was screened against S. aureus, S. typhi and
except 3-Cl and 4-F substituted derivatives. Despite the similar E. coli. The MIC values of the compounds against S. aureus, S. typhi
range of BuChE inhibitory activity of both 4-F analogs (141c and and E. coli (100e150 mg/mL) were far inferior to that of cloxacillin
142c), compound 141c showed the highest anti-AChE activity at a (MIC: 10.0 mg/mL), ciprofloxacin (MIC: 5.0 mg/mL) or gentamycin
25
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

(MIC: 5.0 mg/mL) as the standard drugs. known pharmacophore inhibiting AChE aroused the interest of
Pyranobenzopyran derivatives (151e153) were also prepared the researchers to synthesize new pyranoquinoline-fused
through a three-step synthetic method including (1) Knovenagel coumarin derivatives having high anticholinesterase activity
condensation of compound 63a with diethyl malonate (147), (2) (Fig. 35) [138].
Michael addition of compound 148 and ethyl aceto acetate (EAA,
149) or acetyl acetone (150), and (3) pechmann condensation of 151
with EAA (Scheme 30) [131]. Antibacterial activity of the pyr- 5.9.1. Anticholinesterase activity
anobenzopyran derivatives (151e153) against S. aureus, E. coli and A series of tetrahydrochromeno[30 ,4’:5,6]pyrano[2,3-b]quino-
S. typhi was compared with dioxo-pyranocoumarin 148 and the lin-6(7H)-one derivatives (162) were prepared via a two-step re-
standard drugs. All the prepared compounds showed no appro- action including a three-component reaction between
priate antimicrobial activity (MIC > 110.0 mM) compared with the benzaldehydes (125), malononitrile (102), and 4-hydroxycoumarin
standard drugs (ciprofloxacin MIC: 5.0; cloxacillin MIC:10.0 and (32) to yield compound 160 which was then reacted with cyclo-
gentamycin MIC: 5 mM). Compound 151 showed better antibacterial hexanone (161) via €nder
friedla reaction. The pyr-
activity against S. aureus which could be due to intrinsic antimi- anoquinolinocoumarins 162 were screened against AChE and BChE
crobial property of catechol moiety in the structure of the com- and their activity was compared with tacrine as reference drug
pound than that of the mother analogue [134].Compound 153 also with IC50 value of 410 and 32.0 nM against AChE and BuChE,
showed better antibacterial activity against both S. aureus, S. typhi respectively (Scheme 33) [138]. Among them, 4-fluorophenyl
strains than compound 148 which could be due to the presence of substituted derivative (162a) was the most active compound
the fused pyranonebenzopyranone system [135]. Compound 152 against AChE (IC50: 5.00 nM), while benzodioxole-5-yl substituted
showed higher activity against E. coli than other strains. Compound (162b) showed high activity against BuChE (IC50: 570.0 nM). By
153 having more lipophilic methyl substituted group showed replacing the aromatic substitutions (R ¼ phenyl, 4-pyridyl, 2-
acceptable activity against both Gram positive and Gram negative thienyl) with a methyl group, more inhibitory potency against
strains (S. aureus, S. typhi and E. coli). BChE and less inhibitory activity against AChE were obtained.
In another study, compound 155 was prepared by the reaction of Docking studies on the active compound showed both occupation
3-formyl-4-hydroxy coumarin 62a and hippuric acid 154. Com- of PAS and CAS of AChE and mixed-type kinetic mode of the
pound 155 exhibited appropriate antibacterial activity towards enzyme inhibition. It was revealed that p-p stacking between
S. typhi (MIC: 50 mg/mL) (Scheme 31) [78]. Comparison between phenyl moiety of compound 162a (R ¼ 4-fluorophenyl group) and
antibacterial activity of compound 148 and compound 155 showed the phenyl side chain of Tyr334 as well as hydrogen bonding be-
that addition of the amide bond could improve the antimicrobial tween the carbonyl group of the coumarin ring and the hydroxyl
activity of the target compounds. group of Tyr121 were the main factor stabilizing the complex of
ligand-protein in the active site.
5.8.2. Antifungal activity
Zhang and his coworkers described synthesis and antifungal
activity of pyrano[3,2-c]chromene-2,5-dione derivatives (158 and 4.10. Dihydropyrano pyrrolocoumarin derivatives
159) against five phytopathogenic fungi (B. cinerea, C. capsici,
A. solani, A. mali, and Rh. solani) [132]. Various derivatives of pyrano The 4H-pyran derivatives have attracted immense interest in
[3,2-c]chromene-2,5-dione were synthesized by the reaction of 4- medicinal chemistry due to their various pharmacological and
hydroxycoumarin derivatives (32) with beketoesters (156 or 157) biological activities, such as antimicrobial, mutagenicity, anti-
(Scheme 32). All derivatives of compound 158 exhibited more proliferative, antitumor activities [139]. Heterocycles having fused
antifungal activity against B. cinerea (inhibition rate: 35e67%) than dihydropyranopyrrole nucleus have attracted much attention due
azoxystrobin as reference drug (inhibition rate: 34%), except com- to the synergistically improved effect of the heterocycles. There is
pound 158a (R1 ¼ O(CH2)3CH3, R2 ¼ H, R3 ¼ Me) and 158b only one report on dihydropyrano[2,3-b]pyrrole derivatives fused
(R1 ¼ OMe, R2 ¼ H, R3 ¼ Me). Compound 158c (R1 ¼ H, R2 ¼ Et, to the coumarin ring as antimicrobial agents (Fig. 36) [140].
R3 ¼ Me) showed superior antifungal activity against B. cinerea,
C. capsici, A. solani, more active than the standard drug. Among
compounds 158, only one derivative substituted by R1 ¼ H, R2 ¼ Et, 5.10.1. Antimicrobial activity
and R3 ¼ Me showed better antifungal activity against B. cinerea Al-Haiza et al. synthesized compound 165 via the reaction of
and C. capsica (EC50 values of 0.141 and 0.115 mM, respectively) compound 163 and 4-bromophenacyl bromide (164) (Scheme 34)
compared to azoxystrobin (the control, EC50: 0.158 and 0.222 mM, [140]. The antibacterial and antifungal activities of the compounds
respectively). The results revealed that the presence of lipophilic were evaluated against Gram-positive bacterium (S. aureus,
ethyl group at 4-position of the compound (R2 ¼ Et) has more effect B. subtilis and B. cereus), Gram-negative bacterium (E. coli, P. aeru-
than other substituents on the antifungal activity. Compound 158b ginosa, and E. aerogenes), and fungi (A. niger, P. italicum, and
was more active than compound 158d confirming the crucial effect F. oxysporum). The study revealed that the ring-closing and for-
of CF3 group (R2 ¼ CF3) to improve the antifungal activity of the mation of dihydropyranopyrrolo moiety decreased the antibacte-
target compounds. The results of the study confirmed that the level rial and antifungal activities of the target structure (165) compared
of the hydrophobicity generated by substituents at 3,4-positions with that of the mother compound (163). The order of the anti-
had great effect on the activity of the dioxo-pyranocoumarins. microbial activity was 163 > 165. Compound 165 showed signifi-
cant antibacterial activity against B. subtilis, more than amoxicillin
5.9. Pyranoquinolino coumarin derivatives as the standard drug. However, compound 165 exhibited lower
antifungal activity against all the fungi compared to mycostatine as
The pyranoquinolines are one of the attractive scaffolds having a standard drug. Only one pyranopyrrolocoumarin was prepared in
various biological effects such as anticancer [136], psychotropic, this study limiting appropriate interpretation regarding parameters
anti-inflammatory, and antibacterial activities [137]. The great po- affecting the antimicrobial activity of the compound. However, it
tential of 5,6,7,8-tetrahydroquinolin-4-amine (THQA) as an seems that the presence of the free amine is crucial to reach great
important part of Tacrin analogs and coumarin moiety as a well- antibacterial activity.
26
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

5.11. Pyranopyrimidinocoumarin derivatives pyranocoumarin and spirooxindole scaffolds in medicinal chemis-


try, a series of pyranocoumarin derivatives bearing spirooxindole
Pyrano[2,3-d]pyrimidine derivatives possess various pharma- moiety have been synthesized and introduced as promising can-
cological activities including antifungal, antitumor, hep- didates for drug discovery such as antibacterial and anticancer
atoprotective, and antihypertensive activities [141]. In the light of compounds (Fig. 38) [147,148].
the valuable antimicrobial activity of this heterocycle, pyranopyr-
imidinocoumarin derivatives were prepared via fusing pyranopyr- 5.12.1. Antibacterial activity
imidine moiety with the coumarin at 3 and 4 positions of the ring. A series of pyrannocoumarin derivatives bearing spirooxindole
The new heterocyclic systems possessed notable biological activ- moiety was synthesized via a one-pot three-component reaction
ities such as antibacterial and antifungal properties (Fig. 37) between 4-hydroxycoumarin (32), isatin (172), and malononitrile
[140,142,143]. (102). The antibacterial activities of the prepared compounds were
evaluated against S. aureus, S. epidermidis, E. coli, P. aeruginosa
5.11.1. Antimicrobial activity (Scheme 37) [147]. The compounds showed good to moderate
Al-Haiza et al. and El-Agrody et al. used pyrano derivatives of antibacterial activity. The presence of halo group (173b, X ¼ Br) on
coumarin (160) as an intermediate for the preparation of pyr- coumarin ring improved the activity. Also, the length of the R group
anopyrimidino coumarin derivatives (168 and 167) and evaluated had great effect on the MIC of the target compounds. Compound
their activity against Gram-positive bacteria (S. aureus, B. subtilis, B. 173g with long-chain alkyl halide-functionalized oxindole exhibi-
cereus) and Gram-negative bacteria (E. coli, P. aeruginosa, ted maximum antibacterial activity (MIC: 62.5 mg/mL) as compared
E. aerogenes) (Scheme 35, Table 11) [131,140,142]. The biological to the others (MIC value: 125e512 mg/mL) revealing the important
screening indicated that cyclization of compound 160 had no sig- role of the lipophilicity controlling the penetration and activity of
nificant impact on antibacterial activity of compound 166 and 167. the compounds.
However, among the prepared compounds having a 4-
bromophenyl group (160a, 166a and 167a), compound 167a 5.12.2. Anticancer activity
showed the best antibacterial activity towards S. aureus, B. subtilis, The cytotoxic activity of the pyrannocoumarins 173 was also
P. aurignosa, and E. aerogenes compared to the standard drug evaluated against U87 human glioma cells. All the derivatives
amoxicillin [140]. In addition, the antibacterial evaluation of the showed week to moderate cytotoxic activity. Compounds 173a-
pyranopyrimidinocoumarin derivatives with R ¼ CH3 showed 173band 173e showed the same cytotoxic activity (IC50: 78 mM) and
moderate antibacterial activity of the compounds (166b and 167b) compound 173g was the best active compound (IC50: 39.0 mM), but
against all the tested bacteria (S. aureus, B. cereus, S. marcescens, P. lower than carmustine as the standard drug (IC50: 3.9 mg/mL)
merabitis) compared to ampicillin as the reference drug [143]. (Scheme 37) [148]. It has been suggested that the presence of long
The authors also examined the antifungal activity of the pyr- lipophilic moiety could increase the anticancer activity of the
anopyrimidinocoumarin derivatives (166 and 167) against A. niger, compounds, which could be probably due to the higher hydro-
P. italicum, and F. oxysporum. Compound 167a showed the most phobic character of the compounds resulting in the high cellular
activity against A. niger compared to the standard fungicide uptake.
mycostatine [140]. The antifungal activity of compounds 160b,
166b, and 167b was also investigated against A. ochraceus Wilhelm 6. Coumarin-fused unsaturated six-membered rings with two
and P. chrysogenum Thom which was lower than mycostatine as the heteroatoms
standard drug [143].
A series of pyranotriazolopyrimidine derivatives (169a-d) were 6.1. Pyrimidinocoumarin derivatives
synthesized by hydrazinolysis of compound 168. Corresponding
triazolopyrimidine (170) were then synthesized via cyclization of Pyrimidine, as a privileged scaffold in a wide range of hetero-
compound 169 by appropriate carboxylic acid chlorides/diethyl cyclic compounds, showed a broad spectrum of activities including
oxalate. The pyrimidotriazine scaffold (171) was similarly prepared antimicrobial, anti-viral, anti-inflammatory, antihypertensive, anti-
by reaction of compound 168 and triethylorthoformate (Scheme HIV, and anticancer activities [149]. There are several works
36) [142,143]. The biological screening of the synthesized com- reporting the biological activity of pyrimidines in fused structure,
pounds (169, 170 and 171) against various bacteria (S. aureus, especially anticancer activity with various mechanisms. Pyr-
B. cereus, S. marcescens, and P. merabitis) and fungal strains imidinocoumarins were widely evaluated as antibacterial, anti-
(A. ochraceus Wilhelm, P. chrysogenum Thom) revealed that all the cancer, antioxidant, and antithrombotic agents (Fig. 39)
compounds had similar antibacterial activity compared to ampi- [78,148e153].
cillin as the reference drug (Table 11). Compounds 168 and 169
showed nearly the same antifungal activity against A. ochraceus 6.1.1. Anticancer activity
Wilhelm and P. chrysogenum Thom. Compounds 170a-d and 171 A series of mono- and di-substituted 2-phenyl-benzopyr-
were the most active compounds against A. ochraceus Wilhelm and anopyrimidines (PBPP) (176, 178, 179) were synthesized via ternary
P. chrysogenum Thom, while compounds 160b, 167b, 168, and 169b condensation of salicylaldehyde (99) or its derivative (174), ECA,
exhibited moderate activity against A. ochraceus Wilhelm and and benzaldehyde (125) in the presence of CH3COONH4 (Scheme
P. chrysogenum Thom. 38) [150]. The selectivity of the synthesized compounds to telo-
meric G-quadruplex DNA and their inhibitory effect on telomerase
5.12. Spirooxindoles activity were evaluated. The results showed that the di-substituted
PBPP derivatives (179) with two cationic side chains had higher G-
Spirooxindole framework is considered as an important struc- quadruplex stabilizing property than the monosubstituted de-
tural motif and is available in several medicinal agents and natural rivatives (176 and 178) due to the great impact of two alkylamino
products [144]. Spirooxindole derivatives exhibited a wide spec- side chains on a strong interaction with G-quadruplex and their
trum of biological activity including antimicrobial, anti- large planar scaffold. The cytotoxic activity of the compounds was
inflammatory, antitumor, antimalarials, anti-HIV and antiviral, also evaluated against normal cell line (ECV-304) and a series of
anticancer activities [145,146]. Taking account of the application of telomerase positive cancer cell lines (A549, HepG2, and K562).
27
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13
Classification of 3,4-heterocyclic fused coumarins based on the structure and biological properties.

C$NO. Chemical structure Biological activity Result Ref.

Coumarin-fused five-membered heterocycle with one heteroatom


18 MDR reversal HCT116/VM46 IC50: 40 mM [28]

19 In vitro Cytotoxic L1210 IC50: 10 mM [28]

27 Anti-retroviral IC50: 8.0 mM [36]

30a Topoisomerase I inhibitor IC50: 38.5 nM [40]

30b DYRK1A inhibitor IC50: 0.07 mM [40]

34 Antifungal Botrytis cinerea EC50: 1.72 mM [43]

37d Anticancer HCT-15 IC50: 8.0 mM [44]

40b Anticancer SK-BR-3 ED50: 0.10 mM [47b]

41 Antioxidant 1.62 times more than trolox [48]

43 Estrogen receptor modulator IC50: 1.03 mM [51]

28
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13 (continued )

C$NO. Chemical structure Biological activity Result Ref.

44 Antifungal F. oxysporum Inhibition Zone: 16 mm [52]

47d Antimicrobial G. candidum Inhibition Zone: 10 mm [52]

51 RNA-binding microRNA-29a [55]

53b Antiproliferative Hela IC50: 8.84 mM [58]

62 Anticancer MCF-7 IC50: 41.0 mM [60]

Antioxidant IC50: 4 mM
Coumarin-fused five-membered heterocycle with two heteroatom
65a Antibacterial MIC: 100 mM [72]

S. aureus and
S. typhi
68 Antibacterial Candida albicans Inhibition Zone: 14 mm [52]

69 Antifungal Candida albicans Inhibition Zone: 10 mm [52]

73a Anticancer A549 IC50: 27.0 mM [76]

77 Anticancer HCT116 IC50: 1.4 mM [80]

(continued on next page)

29
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13 (continued )

C$NO. Chemical structure Biological activity Result Ref.

Coumarin-fused saturated 6-membered heterocycle


80a Antifungal Powdery mildew 68% [82]

86 Antipsychotic Ki: 1.5 nM [83]

(Dopamine D4 receptor antagonists)


87a Bronchodilator ID50: 35 mg/kg [84]

90e Antimicrobial genomic cleavage [90]

S. aureus and
A. niger
Coumarin-fused unsaturated 6-membered heterocycle with one heteroatoms
93f Antimicrobial MIC: 62.5 mM [93]

S. aureus and E. coli


96g Antibacterial Inhibition Zone: 22 mm [91]

B. subtilis and E. coli


100a Antibacterial MIC: 6.3 mM [95]

S. epidermidis

30
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13 (continued )

C$NO. Chemical structure Biological activity Result Ref.

103d Anti-inflammatory 62% [96]

Analgesic
106 Antimalarial plasmodium falciparum IC50: 1100 nM [97]

112 Anticancer EAC IC50: 0.12 mM [98]

113 Anticancer EAC IC50: 0.12 mM [98]

114 Anticancer EAC IC50: 0.13 mM [98]

115 Anticancer EAC IC50: 0.13 mM [98]

121 Anticancer EAC IC50: 245.7 mM [105]

123 Anticancer MDA-MB IC50: 8e16 mM [108]

126a Anticancer IC50 against A-549: 0.14 mM [109]

129a Anticancer A-549 IC50: 0.14 mM [110]

(continued on next page)

31
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13 (continued )

C$NO. Chemical structure Biological activity Result Ref.

132 Antimicrobial MIC: 50 mg/mL [112]

B. subtilis, S. aureus, E. coli, and A. niger


135a Anti-inflammatory 89% [122]

Antimicrobial
Pseudomonas chinchori, Aspergillus fumigatus MIC: 25 mM
and and
Penicillium wortmanni MIC: 100 mM
141c Anticholinesterase AChE IC50: 0.038 mM [125]

142 Anticancer MCF-7 IC50: 49 mM [127]

146 Antibacterial S.aureus, E.coli and S.typhi MIC: 150 mM and 100 mM [130]

152 Antibacterial E.coli MIC: 105 mM [131]

155 Antibacterial S.typhi MIC: 50 mM [72]

158 Antibacterial EC50: 0.141 and 0.115 mM [132]

B. cinerea and C. capsica

32
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13 (continued )

C$NO. Chemical structure Biological activity Result Ref.

162a Anticholinesterase AChE IC50: 5.0 nM [138]

165 Antibacterial MIC: 15 mM [140]

Bacillus subtilis
167a Antibacterial Bacillus subtilis Inhibition zone: 22 mm [140]

Antifungal Aspergillus niger Inhibition zone: 14 mm


170d Antibacterial S. marcescens Inhibition zone: 25 mm [142]

171 Antimicrobial chrysogenum Thom, chrysogenum Thom and Aspergillus ochraceus Inhibition zone: 22 mm [143]

Wilhelm and 20 mm
173 Antibacterial U87 IC50: 39 mM [147,148]

Anticancer
Coumarin-fused unsaturated 6-membered heterocycle with two heteroatoms
179 Anticancer HepG2 IC50: 2.1 mM [150]

181 Anticancer HepG2 IC50: 18.8 mM [150]

187c Anticancer U46619 HepG2 IC50: 3.0 mM [152]

(continued on next page)

33
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Table 13 (continued )

C$NO. Chemical structure Biological activity Result Ref.

188 Antibacterial S. aureus MIC: 100 mM [72]

194 Antimicrobial C. albicans, S. aureus Inhibition zone: 19 mm [153]

Antioxidant 4 times more than vitamin E


198b Anticancer HepG2 IC50: 36.0 mM [151]

200 Anticancer HepG2 IC50: 58.0 mM [151]

Coumarin-fused 7-membered heterocycle


203c Anticancer HepG2 IC50: 0.29 mM [158]
Antioxidant IC50: 0.29 mM (DPPH assay)

Telomerase inhibition activity of the derivatives and their effect on as the standard antithrombotic drug. Also, the results showed that
the cancer cell lines revealed that compound 179 was the most the presence of any substituents (except for R’ ¼ SO2CH3) at posi-
promising one as a telomerase inhibitor and telomeric G-quad- tion 2 of the pyrimidine ring resulted in the lower fibrin clotting
ruplex binding ligand and had significant selectivity over normal retraction and also antiplatelet activity than the unsubstituted
cells (IC50: 2.10 and 15.5 mM against HepG2 and normal cell lines, analogue (Up to 1 mM, Table 12). Furthermore, it was cleared that
respectively). Additionally, cellular experiments displayed a in vivo antithrombotic activity had no relation with in vitro inhi-
notable decrease in the cell population growth as well as a short- bition of platelet. Only compounds 183d and 187aec could signif-
ening of the telomere length. It was also found that compound 179 icantly prevent the thromboembolic paralysis and they were more
accelerated the senescence of K562 cancer cell lines. potent than aspirin; but less powerful than the conventional anti-
A pyrimidine-fused coumarin derivative was constructed form coagulant warfarin. Compound 187c displayed 3 times more potent
the acetylation of 2-amino-5-bromo-4-hydroxxy-5H-chromeno- activity than compounds 187a and 187b.
[4,3-d]-pyrimidine-5-one 180 (Scheme 39) [151]. The molecules
have been evaluated as anticancer agents against HepG2 cell line.
Results revealed that compound 181 had higher cytotoxic activity
(IC50: 18.80 mg) compared to compound 180. 6.1.3. Antibacterial activity
A series of pyrimidine-fused coumarin derivatives (188) was
prepared from compound 63a via a two-step reaction and their
6.1.2. Antithrombotic activity antibacterial activities was evaluated against Gram-positive
Pyrimidinocoumarin was reported as antiplatelet and antith- (S. aureus) and Gram-negative bacteria (S. typhi) (Scheme 41)
rombotic compounds [152]. Compounds 183a-c were obtained by [78]. All compounds 188 showed similar activity against S. aureus
oxidation of compound 182 with KMnO4 (Scheme 40) [152]. (MIC value of 100 mg/mL), but the order of antibacterial activity
Compounds 185a-f were synthesized from the reaction of com- against S. typhi was X ¼ O > NH > S.
pound 184 (R’ ¼ CN) with amidine or guanidine in ethanol and Also, a series of pyrimidine-annulated coumarins (190e196)
compounds 186a-c were prepared via a three-step reaction were synthesized by reaction between 3-cyano-4-chlorocoumarin
including (1) reaction between compound 184 (R’ ¼ COOCH3) and (184a) and heteroarylamine (189) in one pot condition and then
S-methylisothiourea, (2) chlorination with POCl3, and (3) replace- they were evaluated as antimicrobial agents using the disk diffu-
ment of chlorine with cycloamine. Also, oxidation of the SCH 3 sion method (Scheme 42) [153]. The reduction in bacterial growth
group of compounds 186a-c with m-chloroperbenzoic acid pro- was observed for all the compounds except for compounds 195 and
duced compounds 187a-c. Inhibition of the platelet aggregation 196. Just compound 195 showed fungistatic activity against A. niger
and clot retraction of compounds 183e187 were studied using ADP- (growth inhibition zone: 15.0 mm), as potent as the standard
, arachidonic acid-, and the stable TXA2-receptor agonist U46619- antibiotic nystastin (growth inhibition zone: 17.0 mm). Compound
induced platelet aggregation in rabbit platelet-rich plasma and 194 exhibited strong antimicrobial activity against various infec-
against thrombin-induced clot retraction in rat-platelet-rich tious microorganisms including E. coli, K. pneumoniae, P. aeruginosa,
plasma. Some of the compounds exhibited broad-spectrum of an- S. aureus, S. enteritidis, A. niger, C. albicans and P. aeruginosa without
tiplatelet activity in vitro against ADP- AA- and U46619-induced any selectivity (growth inhibition zone: 17.0, 18.0, 19.0, 18.0, 16.0,
aggregation and had higher potency in comparison with aspirin 19.0, and 16.0 mm, respectively).
34
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

6.1.4. Antioxidant activity 7.1.1. Anticancer and antioxidant activities


The antioxidant capacity of compounds 190e196 was also Our group prepared a series of thiazepine-annulated coumarins
determined spectrophotometrically using phosphomolybdenum (203) from 4-hydroxycoumarin derivatives (32) [158]. The in-
method. Compound 194 showed four times more activity than that termediates (201) were reacted with 2-aminoethanethiol hydro-
of the standard drug a-tocopherol acetate (Fig. 42) [153]. The de- chloride (202) to yield compounds 203 (Scheme 45). The
gree of antioxidant capacity, in descending order, was 194 > 195 > antioxidant and cytotoxic activities of the thiazepine-fused cou-
196 > 193 > 190 > 189 > 192. After hydrolysis of compounds 190 marins were evaluated. Compounds 203a and 203b showed the
and 192 producing compounds 191 and 193, respectively, the highest antioxidant power determined using the DPPH colorimetric
antioxidant activity was increased (193 > 192 and 191 > 190) which method (IC50: 0.12 mM), but the activity was lower than quercetin
could be due to the higher ability of the compounds in the acidic H- as a standard drug (IC50: 7.92 mM). The study revealed that thia-
atom transfer. zepine annulation improved the radical scavenging ability of the
mother compounds. Compound 203c with respectable DPPH
6.2. Imidazopyrimidinocoumarin derivatives radical scavenging activity (IC50: 0.29 mM) exhibited the most
potent scavenging activity confirmed by FRAP assay (FRAP
Imidazo[1,2-a]pyrimidines have attracted a great deal of interest value ¼ 186.4 mM Fe2þ/100 mg), more potent than quercetin (FRAP
among researchers in the field of medicinal chemistry because of value ¼ 53.9 mM Fe2þ/100 mg). Moreover, the result of cytotoxic
their broad spectrum of biological activities such as antimicrobial, evaluation displayed that all derivatives of compound 201 had no
antifungal, antiviral, anticancer, anti-tubercular, anti-inflammatory, significant cytotoxic activity against all the tested cell lines; while
parasiticidal, benzodiazepine receptor stimulating, calcium chan- fusing of thiazepine ring at 3 and 4 positions of the coumarin ring
nel blocking, and P38 MAP kinase inhibitory activities [154]. Imi- resulted in the formation of hybrid structures having good cyto-
dazopyrimidinocoumarins are tetracyclic fused heterocycles with toxic activity against all the tested cell lines. Compound 203c was at
anticancer activity (Fig. 40) [151]. least two times stronger than etoposide as a standard drug against
human cancer cell lines such as SK-N-MC, MDA-MB 231, and MCF-7.
6.2.1. Anticancer activity
8. Conclusion
The cyclization of compound 180 by 4-substituted phenacyl
bromide 197 yielded imidazopyrimidinocoumarin 198. Compound
Coumarins are the heterocyclic structures currently being of
198a showed more cytotoxic activity than compound 198b. The
great interest in the development of new lead structures bearing
order of toxicity against HePG2 cell line was 198a>198b (Scheme
valuable biological properties. In this work, we try to review the
43) [151].
effect of the heterocycles annulation at 3,4-position of the
coumarin ring on the biological activity of the target structures.
6.3. Pyrimidopyrimidinocoumarin derivatives
This review provides a better insight into medicinal chemists on the
potential biologically active heterocyclic-fused coumarin de-
Pyrimido[1,2-a]pyrimidines are one of the famous scaffold in
rivatives to design new coumarin analogs with a more significant
various therapeutically potential compounds like bronchodilator,
therapeutic effect. So, it will help in understanding the effect of
vasodilator, anticancer, and anti-hypertensive agents [155]. The
various substituents and heterocyclic rings fused to 3,4-position of
effect of the annulation of this heterocyclic ring with coumarin
coumarin on the biological activities of the target compounds. The
scaffold on the biological properties of the target compounds was
biological properties of different 3,4-heterocyclic-fused coumarins
evaluated. It was found that pyramidopyrimidinocoumarins could
along with chemical structures are schematically reviewed in
be considered as potential antitumor agents (Fig. 41) [151].
Table 13.
All studied activities like antibacterial, antifungal, anticancer,
6.3.1. Anticancer activity anti-proliferative, antipsychotic, antimalarial, antithrombotic, es-
Sherif et al. synthesized compound 201 as a pyrimidopyr- trogen receptor modulating, RNA-binding, antioxidant, and
imidinocoumarin derivative from the cyclization of the compound DYRK1A inhibitor have been discussed hereunder separated
180a with methyl acrylate (199) and assessed its anticancer activity headings. Antibacterial and antifungal activities were the most
against HePG2 cancer cell lines (Scheme 44) [151]. The results studied analyses performed on the heterocycles fused to 3 and 4
showed that the activity of compound 200 did not significantly positions of the coumarin ring. Heterocyclic rings annulation at 3
change compared to compound 180a. Comparison of the cytotoxic and 4 positions of coumarin ring generally produced an improved
activity of all the compounds prepared in the study performed by synergistic effect. It has been revealed that the antimicrobial ac-
Sherif et al. revealed that order of the cytotoxic activity was 181b tivity of the heterocyclic fused-coumarins could be efficiently
[ 198a>200 > 198b confirming the important role of the type of modulated by the pattern of appropriate substitutions. The pres-
the heterocycle fused at 3,4-positions of the coumarin ring. ence of the fused cyclic skeleton like 7,8-benzo substituent on the
coumarin ring could improve the potent antibacterial activity of the
7. Coumarin fused seven-membered ring target compounds; while, omitting or protection of the free amine
groups caused deterioration of the antimicrobial activity. Lip-
7.1. Thiazepinocoumarin ophilicity also played a crucial role on the antimicrobial activity of
the target compounds through improved permeation of the target
Thiazepine and its derivatives are pharmacologically important compounds. Some of the studies suffered from lack of the deriva-
compounds having various biological properties such as anticancer tization limiting the appropriate QSAR and more studies are
and anti-inflammatory effect [156]. The aryl- and heteroaryl-fused necessary to comprehensively understand the antimicrobial
1,4-thiazepines are privileged structure with fascinating pharma- mechanism of the 3,4-heterocycle-fused coumarins.
cological properties, such as calcium channel blocking, antipsy- The anticancer effect was another most studied biological ac-
chotic, anti-inflammatory, antibacterial and antiviral activities tivity. Table 13 shows the structure of all the heterocycles fused to
[157]. There is only one report about thiazepine-fused coumarin 3,4-position of the coumarin ring with anticancer potency. Pyrrole,
derivatives as anticancer and antioxidant agents (Fig. 42) [158]. furan, thiophene, benzofuran, selenophene, pyrazole,
35
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

hydroimidazole, pyridine, hydropyridine, pyran, spiroxindole, py- Acknowledgement


rimidine and thiazepine are heterocycles fused at 3,4-position of
the coumarin rings. Pyrimidine derivatives, especially amino- The authors thank The Institute of Pharmaceutical Sciences
pyrimidine showed great potential to endow the target compounds (TIPS).
with remarkable cytotoxic activity. Lipophilic substituents on spi-
rooxindol, pyran and imidazole moieties fused to the coumarin ring References
increased anticancer activity of the target structures. In furo-
coumarin derivatives, the alkoxy and hydroxyl enriched com- [1] M. Solanki, An Introduction of Coumarins as Bioactive Heterocycles, Publish
World, 2013.
pounds had better potency compared with alkyl analogs. Moreover, [2] D.A. Ostrov, J.A. Hernandez Prada, P.E. Corsino, K.A. Finton, N. Le, T.C. Rowe,
fluorine group as the unique electron-withdrawing group (F) Discovery of novel DNA gyrase inhibitors by high-throughput virtual
showed more effect on the cytotoxic activity of the quinolinocou- screening, Antimicrob, Agents Chemother 51 (2007) 3688e3698.
[3] N.A. Gormley, G. Orphanides, A. Meyer, P.M. Cullis, A. Maxwell, The inter-
marin derivatives than Cl, OMe, or Me substituents. Also,
action of coumarin antibiotics with fragments of the DNA gyrase B protein,
hydroxamic acid and ureido groups could improve the cytotoxic Biochem 35 (1996) 5083e5092.
activity of the thienocoumarin derivatives. There are still gaps in [4] M. Curini, F. Epifano, F. Maltese, M.C. Marcotullio, S.P. Gonzales,
the anticancer mechanism of various reported 3,4-heterocycle- J.C. Rodriguez, Synthesis of Collinin, an antiviral coumarin, Aust. J. Chem. 56
(2003) 59e60.
fused coumarins and more studies should be performed to ratio- [5] Y. Kashman, K.R. Gustafson, R.W. Fuller, J.H. Cardellina, J.B. McMahon,
nally design appropriate target structures. M.J. Currens, R.W. Buckheit, S.H. Hughes, G.M. Cragg, M.R. Boyd, The cala-
Indolocoumarins, pyrano- and pyranoquinolinocoumarins were nolides, a novel HIV-inhibitory class of coumarin derivatives from the
tropical rainforest tree, Calophyllum lanigerum, J. Med. Chem. 35 (1992)
the only 3,4-heterocycle-fused coumarin scaffolds studied in 2735e2743.
neurodegenerative diseases. Pyrano- or pyranoquinolinocoumarins [6] Y. Shikishima, Y. Takaishi, G. Honda, M. Ito, Y. Takfda, O.K. Kodzhimatov,
showed appropriate ChE inhibition activity and promising features O. Ashurmetov, K.H. Lee, Chemical constituents of Prangos tschimganica;
structure elucidation and absolute configuration of coumarin and fur-
to be candidate molecules for further studies as anti-Alzheimer’s anocoumarin derivatives with anti-HIV activity, Chem. Pharm. Bull. 49
disease agents. (2001) 877e880.
3,4-Furano, seleno, thiazepino, pyridino, and benzofurano-fused [7] M.E. Riveiro, A. Moglioni, R. Vazquez, N. Gomez, G. Facorro, L. Piehl, E.R. de
Celis, C. Shayo, C. Davio, Structural insights into hydroxycoumarin-induced
coumarins were studied as antioxidant and anti-inflammatory apoptosis in U-937 cells, Bioorg, Med. Chem. 16 (2008) 2665e2675.
agents so far. The compounds bearing hydroxyl, alkoxy, pyridyl or [8] K.C. Fylaktakidou, D.J. Hadjipavlou-Litina, K.E. Litinas, D.N. Nicolaides, Nat-
aminopyridyl groups showed great potential as promising ural and synthetic coumarin derivatives with anti-inflammatory/antioxidant
activities, Curr. Pharmaceut. Des. 10 (2004) 3813e3833.
antioxidants.
[9] Y. Bansal, P. Sethi, G. Bansal, Coumarin: a potential nucleus for anti-
In sum, the synthetic methods, biological properties and SAR inflammatory molecules, Med. Chem. Res. 22 (2013) 3049e3060.
studies were discussed in this review to pave the way for further [10] A. Manvar, A. Bavishi, A. Radadiya, J. Patel, V. Vora, N. Dodia, K. Rawal,
rational development of heterocycle-fused coumarins. There are A. Shah, Diversity oriented design of various hydrazides and their in vitro
evaluation against Mycobacterium tuberculosis H37Rv strains, Bioorg. Med.
still various type of the reported 3,4-heterocycle-fused coumarins Chem. Lett 21 (2011) 4728e4731.
that no study has been performed on their biological activities. [11] J.Y. Yeh, M.S. Coumar, J.T. Horng, H.Y. Shiao, F.M. Kuo, H.L. Lee, I.C. Chen,
Moreover, coumarin scaffold could be easily functionalized at 3 and ChW. Chang, W.F. Tang, S.N. Tseng, C.J. Chen, ShR. Shih, J.T.A. Hsu, C.C. Liao,
Y.S. Chao, H.P. Hsieh, Anti-influenza drug discovery: structure-activity rela-
4 positions providing appropriate condition for annulation with tionship and mechanistic insight into novel angelicin derivatives, J. Med.
other heterocycles. Unsaturated bond in lactone ring of the Chem. 53 (2010) 1519e1533.
coumarin ring is also ideal for direct annulation with various het- [12] P. Anand, B. Singh, N. Singh, A review on coumarins as acetylcholinesterase
inhibitors for Alzheimer’s disease, Bioorg. Med. Chem. 20 (2012) 1175e1180.
erocycles via CeH activation without prefunctionalization pro- [13] L. Piazzi, A. Cavalli, F. Colizzi, F. Belluti, M. Bartolini, F. Mancinni,
ducing new coumarin-based derivatives which could have probably M. Recanatini, V. Andrisana, A. Rampa, Multi-target-directed coumarin de-
significant biological activity. This review tries to open a new ho- rivatives: hAChE and BACE1 inhibitors as potential anti-Alzheimer com-
pounds, Bioorg. Med. Chem. Lett 18 (2008) 423e426.
rizon for research on 3,4-heterocycle-fused coumarins and to
[14] B. Yuce, O. Danis, A. Ogan, G. Sener, M. Bulut, A. Yarat, Antioxidative and lipid
encourage medicinal chemists to use the promising heterocycle- lowering effects of 7,8-dihydroxy-3-(4-methylphenyl) coumarin in hyper-
fused coumarins for further biological studies. lipidemic rats, Arzneim.-Forsch Drug Res. 59 (2009) 129e134.
[15] G.R. Madhavan, V. Balraju, B. Mallesham, R. Chakrabarti, V.B. Lohray, Novel
coumarin derivatives of heterocyclic compounds as lipid-lowering agents,
Bioorg. Med. Chem. Lett 13 (2003) 2547e2551.
Funding [16] S. Sandhu, Y. Bansal, O. Silakari, G. Bansal, Coumarin hybrids as novel ther-
apeutic agents, Bioorg. Med. Chem. 22 (2014) 3806e3814.
Funding was received for this work. [17] S. Vilar, E. Quezada, L. Santana, E. Uriarte, M. Yanez, N. Fraiz, C. Alcaide,
E. Cano, F. Orallo, Design, synthesis, and vasorelaxant and platelet anti-
All of the sources of funding for the work described in this aggregatory activities of coumarin-resveratrol hybrids, Bioorg. Med. Chem.
publication are acknowledged below: Lett 16 (2006) 257e261.
[18] K.N. Venugopala, V. Rashmi, B. Odhav, Review on natural coumarin lead
compounds for their pharmacological activity, BioMed Res. Int. (2013)
Research ethics 963248.
[19] (a) M.I. Hussain, S. Qamar Abbas, M.J. Reigosa, Activities and novel applica-
tions of secondary metabolite coumarins, Planta Daninha 36 (2018),
We further confirm that any aspect of the work covered in this e018174040;
manuscript that has involved human patients has been conducted (b) H. Fan, J. Peng, M.T. Hamann, J.F. Hu, Lamellarins and related pyrrole-
derived alkaloids from marine organisms, Chem. Rev. 108 (2008) 264e287.
with the ethical approval of all relevant bodies and that such ap- [20] L. Wu, X. Wang, W. Xu, F. Farzaneh, R. Xu, The structure and pharmacological
provals are acknowledged within the manuscript. functions of coumarins and their derivatives, Curr. Med. Chem. 16 (2009)
4236e4260.
[21] M.V. Kulkarni, G.M. Kulkarni, C.H. Lin, C.M. Sun, Recent advances in cou-
marins and 1-azacoumarins as versatile biodynamic agents, Curr. Med.
Declaration of competing interest
Chem. 13 (2006) 2795e2818.
[22] L. Heide, The aminocoumarins: biosynthesis and biology, Nat. Prod. Rep. 26
We wish to confirm that there are no known conflicts of interest (2009) 1241e1250.
associated with this publication and there has been no significant [23] F.G. Medina, J.G. Marrero, M. Macias-Alonso, M.C. Gonzalez, I. Cordova-
Guerrero, A.G.T. Garcia, S. Osegueda-Robles, Coumarin heterocyclic de-
financial support for this work that could have influenced its rivatives: chemical synthesis and biological activity, Nat. Prod. Rep. 32
outcome. (2015) 1472e1507.

36
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

[24] S. Penta, V.R. Rao, G. Satish, A. Sharma, Advances in Structure and Activity production in human breast cancer and colon cancer cells, Food Chem. 141
Relationship of Coumarin Derivatives, Elsevier Inc, 2016. (2013) 381e388.
[25] X. Yang, Zh Chen, W. Zhong, Synthesis of chromeno[3,4-b]pyrrol-4(3H)-ones [46] S.Y. Cho, S. Cho, E. Park, B. Kim, E.J. Sohn, B. Oh, E.O. Lee, H.J. Lee, S.H. Kim,
through the domino cyclization of 3-aminocoumarins with arylglyoxal Coumestrol suppresses hypoxia inducible factor 1a by inhibiting ROS
monohydrates, Eur. J. Org Chem. (2017) 2258e2265. mediated sphingosine kinase 1 in hypoxic PC-3 prostate cancer cells, Bioorg.
[26] K. Hasse, A.C. Willis, M.G. Banwell, A total synthesis of the marine alkaloid Med. Chem. Lett 24 (2014) 2560e2564.
Ningalin B from (S)-Proline, Aust. J. Chem. 62 (2009) 683e691. [47] (a) X. Wang, K.F. Bastow, C.-M. Sun, Y.-L. Lin, H.-J. Yu, M.-J. Don, T.-S. Wu,
[27] J.L. Bullington, R.R. Wolff, P.F. Jackson, Regioselective preparation of 2- S. Nakamura, K.-K. Lee, Antitumor agents. 239. Isolation, structure elucida-
substituted 3,4-diaryl pyrroles: a concise total synthesis of Ningalin B, tion, total synthesis, and anti-breast cancer activity of neo-tanshinlactone
J. Org. Chem. 67 (2002) 9439e9442. from Salvia miltiorrhiza, J. Med. Chem. 47 (2004) 5816e5819;
[28] D.L. Boger, D.R. Soenen, C.W. Boyce, M.P. Hedrick, Q. Jin, Total synthesis of (b) X. Wang, K. Nakagawa-Goto, K.F. Bastow, M.J. Don, Y.L. Lin, T.Sh Wu,
Ningalin B utilizing a heterocyclic azadiene Diels-Alder reaction and dis- K.H. Lee, Antitumor agents. 254. Synthesis and biological evaluation of novel
covery of a new class of potent multidrug resistant (MDR) reversal agents, neo-tanshinlactone analogues as potent anti-breast cancer agents, J. Med.
J. Org. Chem. 65 (2000) 2479e2483. Chem. 49 (2006) 5631e5634.
[29] A.R. Quesada, M.D.G. Gr avalos, J.L.F. Puentes, Polyaromatic alkaloids from [48] X. Li, T. Wang, J. Liu, Y. Liu, J. Zhang, J. Lin, Zh Zhao, D. Chen, Effect and
marine invertebrates as cytotoxic compounds and inhibitors of multidrug mechanism of wedelolactone as antioxidant-coumestan on .OH-treated
resistance caused by P-glycoprotein, Br. J. Canc. 74 (1996) 677e682. mesenchymal stem cells, Arabian J. Chem. 13 (2020) 184e192.
[30] T. Fukuda, F. Ishibashi, M. Iwao, Synthesis and biological activity of lamellarin [49] D. Xin, H. Wang, J. Yang, Y.-F. Su, G.-W. Fan, Y.-F. Wang, Y. Zhu, X.-M. Gao,
alkaloids: an ovverview, Heterocycles 83 (2011) 491e529. Phytoestrogens from Psoralea corylifolia reveal estrogen receptor-subtype
[31] M. Chittchang, P. Batsomboon, S. Ruchirawat, P. Ploypradith, Cytotoxicities selectivity, Phytomed. Int. J. Phytother. Phytopharmacol. 17 (2010) 126e131.
and structure-activity relationships of natural and unnatural lamellarins [50] S.-H. Lim, T.-Y. Ha, J. Ahn, S. Kim, Estrogenic activities of Psoralea corylifolia L.
toward cancer cell lines, ChemMedChem 4 (2009) 457e465. seed extracts and main constituents, Phytomed. Int. J. Phytother. Phyto-
[32] M. Vanhuyse, J. Kluza, C. Tardy, G. Otero, C. Cuevas, C. Bailly, A. Lansiaux, pharmacol. 18 (2011) 425e430.
D. Lamellarin, A novel pro-apoptotic agent from marine origin insensitive to [51] X. Liu, J.W. Nam, Y.S. Song, A.N.I. Viswanath, A.N. Pae, Y.-S. Kil, H.D. Kim,
P-glycoprotein-mediated drug efflux, Canc. Lett. 221 (2005) 165e175. J.H. Park, E.-K. Seo, M. Chang, Psoralidin, a coumestan analogue, as a novel
[33] N. Zhang, D. Wang, Y. Zhu, J. Wang, H. Lin, Inhibition effects of lamellarin D potent estrogen receptor signaling molecule isolated from Psoralea cor-
on human leukemia K562 cell proliferation and underlying mechanisms, ylifolia, Bioorg. Med. Chem. Lett 24 (2014) 1403e1406.
Asian Pac. J. Cancer Prev. APJCP 15 (2014) 9915e9919. [52] A.M.K. El-Dean, R.M. Zaki, A.A. Geies, S.M. Radwan, M.S. Tolba, Synthesis and
[34] M. Facompre , C. Tardy, C. Bal-Mahieu, P. Colson, C. Perez, I. Manzanares, antimicrobial activity of new heterocyclic compounds containing thieno[3,2-
C. Cuevas, C. Bailly, D. Lamellarin, A novel potent inhibitor of topoisomerase c]coumarin and pyrazolo[4,3-c]coumarin frameworks, Russ. J. Bioorg. Chem.
I, Canc. Res. 63 (2003) 7392e7399. 39 (2013) 553e564.
[35] J. Kluza, M.-A. Gallego, A. Loyens, J.-C. Beauvillain, J.-M.F. Sousa- Farco, [53] K. Matsuzaki, K. Sugishita, N. Fujii, K. Miyajima, Molecular basis for mem-
C. Cuevas, P. Marchetti, C. Bailly, Cancer cell mitochondria are direct pro- brane selectivity of an antimicrobial peptide, Magainin 2, Biochem 34 (1995)
apoptotic targets for the marine antitumor drug Lamellarin D, Canc. Res. 66 3423e3429.
(2006) 3177e3187. [54] (a) Y.Y. Wu, W.B. Shao, J.J. Zhu, Z.-Q. Long, L.W. Liu, P.Y. Wang, Z. Li, S. Yang,
[36] M.V.R. Reddy, M.R. Rao, D. Rhodes, M.S.T. Hansen, K. Rubins, F.D. Bushman, Novel 1,3,4-oxadiazole-2-carbohydrazides as prospective agricultural anti-
Y. Venkateswarlu, D.J. Faulkner, Lamellarin a 20-sulfate, an inhibitor of HIV-1 fungal agents potentially targeting succinate dehydrogenase, J. Agric. Food
integrase active against HIV-1 virus in cell culture, J. Med. Chem. 42 (1999) Chem. 67 (2019) 13892e13903;
1901e1907. (b) X. Yu, P. Teng, Y.L. Zhang, Z.J. Xu, M.Zh Zhang, W.H. Zhang, Design,
[37] K.A. Koo, N.D. Kim, Y.S. Chon, M.-S. Jung, B.-J. Lee, J.H. Kim, W.-J. Song, QSAR synthesis and antifungal activity evaluation of coumarin-3-carboxamide
analysis of pyrazolidine-3,5-diones derivatives as DYRK1A inhibitors, Bioorg. derivatives, Fitoterapia 127 (2018) 387e395.
Med. Chem. Lett 19 (2009) 2324e2328. [55] F.H. Havaldar, S. Bhise, S. Burudkar, A facile synthesis of 10-methoxy-4,8-
[38] (a) A. Foucourt, D. Hedou, C. Dubouilh-Benard, L. Desire, A.-S. Casagrande, dinitro-6H-benzothieno[2,3-c]chromen-6-one, J. Serb. Chem. Soc. 69 (2004)
B. Leblond, N. Loaec, L. Meijer, T. Besson, Design and synthesis of thiazolo 527e532.
[5,4-f]quinazolines as DYRK1A inhibitors, part I,, Molecules 19 (2014) [56] (a) F. Hamadi, H. Latrache, H. Zahir, A. Elghmari, M. Timinouni, M. Ellouali,
15546e15571; The relationship between Escherichia coli surface functional groups’
(b) J.A. Beutler, T.C. McKee, R.W. Fuller, M. Tischler, J.H. Cardellina, composition and their physicochemical properties, Braz. J. Microbiol. 39
K.M. Snader, T.G. McCloud, M.R. Boyd, Frequent occurrence of HIV-inhibitory (2008) 10e15;
sulphated polysaccharides in marine invertebrates, Antiviral Chem. Che- (b) I. Malík, M. Bukovský, F. Andriamainty, J. Galisinova , Antimicrobial ac-
mother. 4 (1993) 167e172. tivity of meta-alkoxyphenylcarbamates containing substituted N-phenyl-
[39] H. Falke, A. Chaikuad, A. Becker, N. Loaec, O. Lozach, S. Abu Jhaisha, piperazine fragment, Braz. J. Microbiol. 43 (2012) 959e965.
W. Becker, P.G. Jones, L. Preu, K. Baumann, S. Knapp, L. Meijer, C. Kunick, 10- [57] T. Fukuzumi, A. Murata, H. Aikawa, Y. Harada, K. Nakatani, Exploratory study
Iodo-11H-indolo[3,2-c]quinoline-6-carboxylic acids are selective inhibitors on the RNA-binding structural motifs by library screening targeting pre-
of DYRK1A, J. Med. Chem. 58 (2015) 3131e3143. miRNA-29a, Chem. Eur J. 21 (2015) 16859e16867.
[40] C. Neagoie, E. Vedrenne, F. Buron, J.Y. Me rour, S. Rosca, S. Bourg, O. Lozach, [58] K. Wittine, I. Ratkaj, K. Benci, T. Suhina, L. Mandi c, N. Ili
c, S.K. Pavelic,
L. Meijer, B. Baldeyrou, A. Lansiaux, S. Routier, Synthesis of chromeno[3,4-b] K. Paveli c, M. Mintas, The novel coumarin[3,2-c]thiophene and its
indoles as Lamellarin D analogues: a novel DYRK1A inhibitor class, Eur. J. hydroxamic acid and ureido derivatives: synthesis and cytostatic activity
Med. Chem. 49 (2012) 379e396. evaluations, Med. Chem. Res. 25 (2016) 728e737.
[41] A. Dey, A. Hajra, FeCl3/ZnI2-Catalyzed regioselective synthesis of angularly [59] M. Bjo € rnstedt, S. Kumar, A. Holmgren, Selenodiglutathione is a highly effi-
fused furans, Org. Biomol. Chem. 15 (2017) 8084e8090. cient oxidant of reduced thioredoxin substrate for mammalian thioredoxin
[42] (a) E.C. Gaudino, S. Tagliapietra, K. Martina, G. Palmisano, G. Cravotto, Recent reductase, J. Biol. Chem. 267 (1992) 8030e8034.
advances and perspectives in the synthesis of bioactive coumarins, RSC Adv. [60] P. Arsenyan, J. Vasiljeva, I. Shestakova, I. Domracheva, E. Jaschenko,
6 (2016) 46394e46405; N. Romanchikova, A. Leonchiks, Z. Rudevica, S. Belyakov, Selenopheno[3,2-
(b) M. Borgatti, A. Chilin, L. Piccagli, I. Lampronti, N. Bianchi, I. Mancini, c]- and [2,3-c]coumarins: synthesis, cytotoxicity, angiogenesis inhibition,
G. Marzaro, F. dall’Acqua, A. Guiotto, R. Gambari, Development of a novel and antioxidant properties, C. R, Chimie 18 (2015) 399e409.
furocoumarin derivative inhibiting NF-kB dependent biological functions: [61] Y. Hu, J. Zhang, C. Yu, Q. Li, F. Dong, G. Wang, Z. Guo, Synthesis, character-
design, synthesis and biological effects, Eur. J. Med. Chem. 46 (2011) ization, and antioxidant properties of novel inulin derivatives with amino-
4870e4877. pyridine group, Int. J. Biol. Macromol. 70 (2014) 44e49.
[43] (a) D.A. Mulholland, S.E. Iourine, D.A.H. Taylor, F.M. Dean, Coumarins from [62] Q. Li, C. Zhang, W. Tan, G. Gu, Z. Guo, Novel amino-pyridine functionalized
Ekebergia pterophylla, Phytochemistry (Oxf.) 47 (1998) 1641e1644; chitosan quaternary ammonium derivatives: design, synthesis, and antioxi-
(b) R. Zhang, Z. Xu, W. Yin, P. Liu, W. Zhang, Microwave-assisted synthesis dant activity, Molecules 22 (2017) 156.
and antifungal activities of polysubstituted furo[3,2-c]chromen-4-ones and [63] I. Domracheva, I. Kanepe-Lapsa, L. Jackevica, J. Vasiljeva, P. Arsenyan, Sele-
7,8,9,10-tetrahydro-6h-benzofuro[3,2-c]chromen-6-ones, Synth. Commun. nopheno quinolinones and coumarins promote cancer cell apoptosis by ROS
44 (2014) 3257e3263; depletion and caspase-7 activation, Life Sci. 186 (2017) 92e101.
(c) M.Z. Zhang, R.R. Zhang, J.Q. Wang, X. Yu, Y.L. Zhang, Q.Q. Wang, [64] N.M. Abunada, H.M. Hassaneen, N.G. Kandile, O.A. Miqdad, Synthesis and
W.H. Zhang, Microwave-assisted synthesis and antifungal activity of novel antimicrobial activity of some new pyrazole, fused pyrazolo[3,4-d]-pyrimi-
fused Osthole derivatives, Eur. J. Med. Chem. 124 (2016) 10e16. dine and pyrazolo[4,3-e][1,2,4]-triazolo[1,5-c]pyrimidine derivatives, Mole-
[44] M. Rajabi, Z. Hossaini, M.A. Khalilzadeh, S. Datta, M. Halder, S.A. Mousa, cules 13 (2008) 1501e1517.
Synthesis of a new class of furo[3,2-c]coumarins and its anticancer activity, [65] Kh Karrouchi, S. Radi, Y. Ramli, J. Taoufik, Y.N. Mabkhot, F.A. Al-aizari,
J. Photochem. Photobiol. B Biol. 148 (2015) 66e72. M. Ansar, Synthesis and pharmacological activities of pyrazole derivatives: a
[45] (a) J.H. Lee, B.W. Lee, J.H. Kim, T.S. Jeong, M.J. Kim, W.S. Lee, K.H. Park, LDL- review, Molecules 23 (2018) 134e218.
Antioxidant pterocarpans from roots of Glycine max (L.) Merr, J. Agric. Food [66] S.A.H. El-Feky, Z.K. Abd El-Samii, N.A. Osman, J. Lashine, M.A. Kamel,
Chem. 54 (2006) 2057e2063; H.K. Thabet, Synthesis, molecular docking and anti-inflammatory screening
(b) Y.H. Lee, H.J. Yuk, K.H. Park, Y.S. Bae, Coumestrol induces senescence of novel quinoline incorporated pyrazole derivatives using the Pfitzinger
through protein kinase CKII inhibition-mediated reactive oxygen species reaction II, Bioorg. Chem. 58 (2015) 104e116.

37
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

[67] C.B. Sangani, D.C. Mungra, M.P. Patel, R.G. Patel, Synthesis and in vitro anti- [92] L.J. Nunez-Vergara, J.A. Squella, P.A. Navarrete-Encrina, E. Vicente-Garcia,
microbial screening of new pyrano[4,3-b]pyrane derivatives of 1H-pyrazole, S. Preciado, R. Lavilla, Chromenopyridines: promising scaffolds for medicinal
Chin. Chem. Lett. 23 (2012) 57e60. and biological chemistry, Curr. Med. Chem. 18 (2011) 4761e4785.
[68] J.B. Shi, W.J. Tang, X.B. Qi, R. Li, X.H. Liu, Novel pyrazole-5-carboxamide and [93] A.A. Patel, H.B. Lad, K.R. Pandya, C.V. Patel, D.I. Brahmbhatt, Synthesis of a
pyrazole-pyrimidine derivatives: synthesis and anticancer activity, Eur. J. new series of 2-(2-oxo-2H-chromen-3-yl)-5H-chromeno[4,3-b]pyridin-5-
Med. Chem. 90 (2015) 889e896. ones by two facile methods and evaluation of their antimicrobial activity,
[69] P. Horrocks, M.R. Pickard, H.H. Parekh, S.P. Patel, R.B. Pathak, Synthesis and Med. Chem. Res. 22 (2013) 4745e4754.
biological evaluation of 3-(4-chlorophenyl)-4-substituted pyrazole de- [94] B.S. Dawane, S.G. Konda, R.G. Bodade, R.B. Bhosale, An efficient one-pot
rivatives, Org. Biomol. Chem. 11 (2013) 4891e4898. synthesis of some new 2,4-diaryl pyrido[3,2-c]coumarins as potent antimi-
[70] M. Mamaghani, R. Hossein Nia, F. Shirini, Kh Tabatabaeian, M. Rassa, An crobial agents, J. Heterocycl. Chem. 47 (2010) 237e241.
efficient and eco-friendly synthesis and evaluation of antibactrial activity of [95] L.V. Frolova, I. Malik, P.Y. Uglinskii, S. Rogelj, A. Kornienko, I.V. Magedov,
pyrano[2,3-c] pyrazole derivatives, Med. Chem. Res. 24 (2015) 1916e1926. Multicomponent synthesis of 2,3-dihydrochromeno[4,3-d]pyrazolo[3,4-b]
[71] H. Kumar, D. Saini, S. Jain, N. Jain, Pyrazole scaffold: a remarkable tool in the pyridine-1,6-diones: a novel heterocyclic scaffold with antibacterial activity,
development of anticancer agents, Eur. J. Med. Chem. 70 (2013) 248e258. Tetrahedron Lett. 52 (2011) 6643e6645.
[72] V.V. Mulwad, J.M. Shirodkar, Synthesis of some of the antibacterial com- [96] H.M. Hosni, M.M. Abdulla, Anti-inflammatory and analgesic activities of
pounds from 4-hydroxycoumarins: Part II, Indian J. Chem. 41B (2002) some newly synthesized pyridinedicarbonitrile and benzopyranopyridine
1263e1267. derivatives, Acta Pharm. 58 (2008) 175e186.
[73] W.C. Reygaert, An overview of the antimicrobial resistance mechanisms of [97] K. Gorlitzer, Ch Enge, P.G. Jones, H. Jomaa, J. Wiesner, Benzo[c][2,7]naph-
bacteria, AIMS Microbiol 4 (2018) 482e501. thyridin-2-yl-, 5-yl- and 2,5-diyl-novaldiamine- Synthesis and investigation
[74] A. Tanitame, Y. Oyamada, K. Ofuji, M. Fujimoto, N. Iwai, Y. Hiyama, et al., of anti-malarial activity, Pharmazie 61 (2006) 975e980.
Synthesis and antibacterial activity of a novel series of potent DNA gyrase [98] W.S. Hamama, M.E. Ibrahim, A.E. Metwalli, H.H. Zoorob, New synthetic
inhibitors. Pyrazole derivatives, J. Med. Chem. 47 (2004) 3693e3696. approach to coumarino[4,3-b]pyridine systems and potential cytotoxic
[75] (a) E. Glukhov, L.L. Burrows, C.M. Deber, Membrane interactions of designed evaluation, Med. Chem. Res. 23 (2014) 2615e2621.
cationic antimicrobial peptides: the two thresholds, Biopolymers 89 (2008) [99] M. El-Naggar, H. Almahli, H.S. Ibrahim, W.M. Eldehna, H.A. Abdel-Aziz, Pyr-
360e371; idine-ureas as potential anticancer agents: synthesis and in vitro biological
(b) Y. Chen, M.T. Guarnieri, A.I. Vasil, M.L. Vasil, C.T. Mant, R.S. Hodges, Role evaluation, Molecules 23 (2018) 1459e1474.
of peptide hydrophobicity in the mechanism of action of alpha-helical [100] H. Sachdeva, J. Mathur, A. Guleria, Indole derivatives as potential anticancer
antimicrobial peptides, Antimicrob. Agents Chemother. 51 (2007) agents: a review, J. Chil. Chem. Soc. 65 (2020) 4900e4907.
1398e1406; [101] V.R. Solomon, H. Lee, Quinoline as a privileged scaffold in cancer drug dis-
(c) I.A. Edwards, A.G. Elliott, A.M. Kavanagh, J. Zuegg, M.A.T. Blaskovich, covery, Curr. Med. Chem. 18 (2011) 1488e1508.
M.A. Cooper, Contribution of amphipathicity and hydrophobicity to the [102] A.H. Abadi, T.M. Ibrahim, KhM. Abouzid, J. Lehmann, H.N. Tinsley, B.D. Gary,
antimicrobial activity and cytotoxicity of b-hairpin peptides, ACS Infect. Dis. G.A. Piazza, Design, synthesis and biological evaluation of novel pyridine
2 (2016) 442e450. derivatives as anticancer agents and phosphodiesterase 3 inhibitors, Bioorg.
[76] J.A. Kumar, G. Saidachary, G. Mallesham, B. Sridhar, N. Jain, S.V. Kalivendi, Med. Chem. 17 (2009) 5974e5982.
V.J. Rao, B.C. Raju, Synthesis, anticancer activity and photophysical properties [103] J. Cheng, J.P. Grande, Cyclic nucleotide phosphodiesterase (PDE) inhibitors:
of novel substituted 2-oxo-2H-chromenylpyrazolecarboxylates, Eur. J. Med. novel therapeutic agents for progressive renal disease, Exp. Biol. Med. 232
Chem. 65 (2013) 389e402, 65. (2007) 38e51.
[77] S. Baroniya, Z. Anwer, P.K. Sharma, R. Dudhe, N. Kumar, Recent advancement [104] T. Murata, K. Shimizu, M. Narita, V.C. Manganiello, T. Tagawa, Characteriza-
in imidazole as anticancer agents: a review, Der Pharm. Sin. 1 (2010) tion of phosphodiesterase 3 in human malignant melanoma cell line, Anti-
172e182. cancer Res. 22 (2003) 3171e3174.
[78] Y.-F. Li, G.-F. Wang, P.-L. He, W.-G. Huang, F.-H. Zhu, H.-Y. Gao, W. Tang, [105] M.S. El-Said, M.G. El-Gazzar, M.S. Al-Dosari, M.M. Ghorab, Synthesis, anti-
Y. Luo, C.-L. Feng, L.-P. Shi, Y.-D. Ren, W. Lu, J.-P. Zuo, Synthesis and anti- cancer activity and radiosensitizing evaluation of some new 2-pyridone
hepatitis B virus activity of novel benzimidazole derivatives, J. Med. Chem. derivatives, Arzneimittelforschung 62 (2012) 149e156.
49 (2006) 4790e4794. [106] M.M.F. Ismail, E. Noaman, Novel Pirfenidone analogs as antifibrotic agents:
[79] Y. Hu, Y. Shen, X. Wu, X. Tu, G.X. Wang, Synthesis and biological evaluation of synthesis and antifibrotic evaluation of 2-pyridones and fused pyridones,
coumarin derivatives containing imidazole skeleton as potential antibacte- Med. Chem. Res. 14 (2005) 382e403.
rial agents, Eur. J. Med. Chem. 143 (2018) 958e969. [107] S. Kumar, S. Bawa, H. Gupta, Biological activities of quinoline derivatives,
[80] X. Han, J. Luo, F. Wu, X.Y. Hou, G. Yan, M. Zhou, M. Zhang, Ch Pu, R. Li, Mini Rev. Med. Chem. 9 (2009) 1648e1654.
Synthesis and biological evaluation of novel 2,3-dihydrochromeno[3,4-d] [108] N. Mulakayala, D. Rambabu, M.R. Raja, C.M.C. Suresh Kumar, A.M. Kalle,
imidazole-4(1H)-one derivatives as potent anticancer cell proliferation and G. Rama Krishna, C.M. Reddy, M.V. Basaveswara Rao, M. Pal, Ultrasound
migration agents, Eur. J. Med. Chem. 114 (2016) 232e243. mediated catalyst free synthesis of 6H-1-benzopyrano [4,3-b]quinolin-6-
[81] D. Barker, Pyridines and their benzo derivatives: Reactivity of reduced ones leading to novel quinolone derivatives: their evaluation as potential
compounds 72 (2008) 171e216. anti-cancer agents, Bioorg. Med. Chem. 20 (2012) 759e768.
[82] T.K. Mandal, A.T. Soldatenkov, E.A. Ageev, E.V. Stashenko, E.N. Denisov, [109] Z. Chen, J. Bi, W. Su, Synthesis and antitumor activity of novel coumarin
E.I. Andreeva, N.S. Prostakov, Synthesis and fungicidal activity of substituted derivatives via a three-component reaction in water, Chin. J. Chem. 31
tetrahydro-[3,4-c]- and benzo[h]tetrahydropyrido[3,4-c]coumarins, Pharm. (2013) 507e514.
Chem. J. 24 (1990) 145e149. [110] R. Miri, R. Motamedi, M.R. Rezaei, Q.R. Firuzi, A. Javidnia, A. Shafiee, Design,
[83] (a) D.T. Connor, S.R. Miller, P.C. Unangst, L.D. Wise, Antipsychotic Method synthesis and evaluation of cytotoxicity of novel chromeno[4,3-b]quinoline
Utilizing Certain Tetrahydrochromeno[3,4-C]pyridin-5-Ones, 1998, derivatives, Arch. Pharm. Chem. Life Sci. 2 (2011) 111e118.
p. 5760050. U.S. Patent, No; [111] Y. Fu, M.J. Romero, A. Habtemariam, M.E. Snowden, L. Song, G.J. Clarkson,
(b) P.C. Unangst, T. Capiris, D.T. Connor, T.G. Heffner, R.G. Mackenzie, B. Qamar, A.M. Pizarro, P.R. Unwin, P.J. Sadler, The contrasting chemical
S.R. Miller, T.A. Pugsley, L.D. Wise, Chromeno[3,4-c]pyridin-5-ones: selective reactivity of potent isoelectronic iminopyridine and azopyridine osmium(II)
human dopamine D4 receptor antagonists as potential antipsychotic agents, arene anticancer complexes, Chem. Sci. 3 (2012) 2485e2494.
J. Med. Chem. 40 (1997) 2688e2693. [112] F. Al-Omran, A.Z.A. Elassar, A.A. El-Khair, Synthesis and biological effects of
[84] D.T. Connor, P.C. Unangst, C.F. Schwender, R.J. Sorenson, M.E. Carethers, new derivatives of azines incorporating coumarin, J. Heterocycl. Chem. 40
C. Puchalski, R.E. Brown, M.P. Finkel, 3-(Aminoalkyl)-1,2,3,4-tetrahydro-5H- (2003) 249e254.
[1]benzopyrano[3,4-c]pyridin-5-ones as potential anticholinergic broncho- [113] M. Koca, S. Servi, C. Kirilmis, M. Ahmedzade, C. Kazaz, B. Ozbek,€ €
G. Otük,
dilators, J. Med. Chem. 32 (1989) 683e688. Synthesis and antimicrobial activity of some novel derivatives of benzo-
[85] K.W. Bentley, The Isoquinoline Alkaloids, Harwood Academic Publishers, furan: part 1. Synthesis and antimicrobial activity of (benzofuran-2-yl)(3-
Amsterdam, 1998, pp. 219e264. phenyl-3-methylcyclobutyl) ketoxime derivatives, Eur. J. Med. Chem. 40
[86] A. Capasso, S. Piacente, N. De Tommasi, L. Rastrelli, C. Pizza, The effect of (2005) 1351e1358.
isoquinoline alkaloids on opiate withdrawal, Curr. Med. Chem. 13 (2006) [114] Y.-S. Xie, D. Kumar, V.D.V. Bodduri, P.S. Tarani, B.-X. Zhao, J.-Y. Miao, K. Jang,
807e812. D.-S. Shin, Microwave-assisted parallel synthesis of benzofuran-2-
[87] K.W. Bentley, b-Phenylethylamines and the isoquinoline alkaloids, Nat. Prod. carboxamide derivatives bearing anti-inflammatory, analgesic and antipy-
Rep. 18 (2001) 148e170. retic agents, Tetrahedron Lett. 55 (2014) 2796e2800.
[88] D.S. Bhakuni, S. Jain, The Alkaloids, vol. 28, Academic Press, New York, 1986, [115] M. The venin, S. Thoret, P. Grellier, J. Dubois, Synthesis of polysubstituted
pp. 95e171. benzofuran derivatives as novel inhibitors of parasitic growth, Bioorg. Med.
[89] K. Iwasa, M. Moriyasu, T. Yamori, T. Turuo, D.-U. Lee, W. Wiegrebe, In Vitro Chem. 21 (2013) 4885e4892.
cytotoxicity of the protoberberine-type alkaloids, J. Nat. Prod. 64 (2001) [116] R. Motamedi, A. Shafiee, M.R. Rezai, O.R. Firuzi, N. Edraki, R. Miri, Oxidative
896e898. aromatization, cytotoxic activity evaluation and conformational study of
[90] V.B. Jadhav, S.K. Nayak, T.N.G. Row, M.V. Kulkarni, Synthesis, structure and novel 7-aryl-10, 11-dihydro-7H-chromeno [4,3-b]quinoline-6, 8(9H, 12H)-
DNA cleavage studies of coumarin analogues of tetrahydroisoquinoline and dione derivatives, Iran, J. Pharm. Res. 13 (2014) 103e114.
protoberberine alkaloids, Eur. J. Med. Chem. 45 (2010) 3575e3580. [117] (a) M.-A. Bazin, L. Bodero, C. Tomasoni, B. Rousseau, C. Roussakis,
[91] E.V.L. da-Cunha, I.M. Fechine, D.N. Guedes, J.M. Barbosa-Filho, M.S. da Silva, P. Marchand, Synthesis and antiproliferative activity of benzofuran-based
Protoberberine alkaloids, Alkaloids - Chem. Biol. 62 (2005) 1e75. analogs of cercosporamide against non-small cell lung cancer cell lines,

38
F. Salehian, H. Nadri, L. Jalili-Baleh et al. European Journal of Medicinal Chemistry xxx (xxxx) xxx

Eur. J. Med. Chem. 69 (2013) 823e832; M. Ghandi, A. Foroumadi, A. Shafiee, Design, synthesis, docking study and
(b) F. Xie, H. Zhu, H. Zhang, Q. Lang, L. Tang, Q. Huang, L. Yu, In vitro and biological evaluation of some novel tetrahydrochromeno [3’,4’:5,6]pyrano
in vivo characterization of a benzofuran derivative, a potential anticancer [2,3-b]quinolin-6(7H)-one derivatives against acetyl- and butyr-
agent, as a novel Aurora B kinase inhibitor, Eur. J. Med. Chem. 89 (2015) ylcholinesterase, Eur. J. Med. Chem. 68 (2013) 291e300.
310e319. [139] S. Damavandi, R. Sandaroos, Novel synthetic route to pyrano[2,3-b]pyrrole
[118] A.A. Altaf, A. Shahzad, Z. Gul, N. Rasool, A. Badshah, B. Lal, E. Khan, A review derivatives, Synth, React. Inorg. Met. Org. Chem. 42 (2012) 621e627.
on the medicinal importance of pyridine derivatives, J. Drug Design Med. [140] M.A. Al-Haiza, M.S. Mostafa, M.Y. El-Kady, Synthesis and biological evalua-
Chem. 1 (2015) 1e11. tion of some new coumarin derivatives, Molecules 8 (2003) 275e286.
[119] K. Gorlitzer, C. Kramer, Potential antiallergic agents- synthesis and reactions [141] W.S. Shehab, A.F. El-Farargy, A.O. Abdelhamid, M.A. Aziz, Synthesis and
of 1,4-dihydro-4-oxo-(1)benzofuro(3,2-b)pyridin-2-carboxylic acid esters, biological application of pyranopyrimidine derivatives catalyzed by efficient
Pharmazie 55 (2000) 587e594. nanoparticles and their nucleoside analogues, Synth. Commun. 49 (2019)
[120] W.W.K.R. Mederski, M. Osswald, D. Dorsch, M. Christadler, C.J. Schmitges, 3560e3572.
C. Wilm, Benzofuro[3,2-b]pyridines as mixed ETA/ETB and selective ETB [142] A.M. El-Agrody, M.S. Abd El-Latif, N.A. El-Hady, A.H. Fakery, A.H. Bedair,
endothelin receptor antagonists, Bioorg. Med. Chem. Lett 9 (1999) 619e622. Heteroaromatization with 4-hydroxycoumarin part II: synthesis of some
[121] L.E.J. Kennis, F.P. Bischoff, C.J. Mertens, C.J. Love, F.A.F. Van den Keybus, new pyrano[2,3-d]pyrimidines, [1,2,4]triazolo[1,5-c]pyrimidines and pyr-
S. Pieters, M. Braeken, A.A.H.P. Megens, J.E. Leysen, New 2-substituted imido[1,6-b]-[1,2,4]triazine derivatives, Molecules 6 (2001) 519e527.
1,2,3,4-tetrahydrobenzofuro[3,2-c]pyridine having highly active and potent [143] A.H. Bedair, N.A. El-Hady, M.S. Abd El-Latif, A.H. Fakery, A.M. El-Agrody, 4-
central a2-antagonistic activity as potential antidepressants, Bioorg. Med. Hydroxycoumarin in heterocyclic synthesis Part III. Synthesis of some new
Chem. Lett 10 (2000) 71e74. pyrano[2,3-d]pyrimidine, 2-substituted[1,2,4]triazolo[1,5-c]pyrimidine and
[122] I.A. Khan, M.V. Kulkarni, M. Gopal, M.S. Shahabuddin, C.M. Sun, Synthesis pyrimido[1,6-b][1,2,4] triazine derivatives, Il Farmaco 55 (2000) 708e714.
and biological evaluation of novel angularly fused polycyclic coumarins, [144] rez, J. Ga
J. Quiroga, S. Portillo, A. Pe lvez, R. Abonia, B. Insuasty, An efficient
Bioorg. Med. Chem. Lett 15 (2005) 3584e3587. synthesis of pyrazolo[3,4-b]pyridine-4-spiroindolinones by a three-
[123] K.A. Kumar, R. Nagamallu, V.K. Govindappa, D.M. Lokeshwari, Pyrans: het- component reaction of 5-aminopyrazoles, isatin, and cyclic b-diketones,
erocycles of chemical and biological interest, J. Chem. Pharmaceut. Res. 7 Tetrahedron Lett. 52 (2011) 2664e2666.
(2015) 693e700. [145] G. Shanthi, P.T. Perumal, An InCl3 catalyzed facile one-pot synthesis of novel
[124] Y. Jacquot, B. Refouvelet, L. Bermont, G.L. Adessi, G. Leclercq, A. Xicluna, dispiro[cyclopent-30 -ene]bisoxindoles, Tetrahedron Lett. 49 (2008)
Synthesis and cytotoxic activity of new 2,4-diaryl-4H,5H-pyrano[3,2-c]ben- 7139e7142.
zopyran-5-ones on MCF-7 cells, Pharmazie 57 (2002) 233e237. [146] K. Rad-Moghadam, L. Youseftabar-Miri, Synthesis of novel spiro
[125] M. Khoobi, M. Alipour, A. Sakhteman, H. Nadri, A. Moradi, M. Ghandi, [dihydropyridine-oxindole] compounds in water, Synlett 14 (2010)
S. Emami, A. Foroumadi, A. Shafiee, Design, synthesis, biological evaluation 1969e1973.
and docking study of 5-oxo-4,5-dihydropyrano[3,2-c]chromene derivatives [147] M. Kidwai, A. Jain, S. Singh, V. Nemaysh, P.M. Luthra, An investigatory study
as acetylcholinesterase and butyrylcholinesterase inhibitors, Eur. J. Med. of antibacterial activity of functionalized spirooxindoles, Indian J. Chem. 53B
Chem. 68 (2013) 260e269. (2014) 399e411.
[126] R. Pratap, V. Ji Ram, Natural and synthetic chromenes, fused chromenes, and [148] M. Kidwai, A. Jain, V. Nemaysh, R. Kumar, P.M. Luthra, Efficient entry to
versatility of dihydrobenzo[h]chromenes in organic synthesis, Chem. Rev. diversely functionalized spirooxindoles from isatin and their biological ac-
114 (2014) 10476e10526. tivity, Med. Chem. Res. 22 (2013) 2717e2723.
[127] A. Shafiee, R. Motamedi, O. Firuzi, S. Meili, A.R. Mehdipour, R. Miri, Synthesis [149] A.R. Bhat, Biological activity of pyrimidine derivativies: a review, Org. Med.
and cytotoxic activity of novel benzopyrano[3,2-c]chromene-6,8-dione de- Chem. I. J. 2 (2017) 1e4.
rivatives, Med. Chem. Res. 20 (2011) 466e474. [150] W.B. Wu, S.-H. Chen, J.-Q. Hou, J.-H. Tan, T.-M. Ou, S.-L. Huang, D. Li, L.-Q. Gu,
[128] (a) L.P. Christensen, Polyphenols and polyphenol-derived compounds from Z.-S. Huang, Disubstituted 2-phenyl-benzopyranopyrimidine derivatives as a
plants and contact dermatitis, in: second ed., in: R.R. Watson, V.R. Preedy, new type of highly selective ligands for telomeric G-quadruplex DNA, Org.
S. Zibadi (Eds.), Polyphenols: Prevention and Treatment of Human Disease, Biomol. Chem. 9 (2011) 2975e2986.
vol. 2, 2018, pp. 349e384; [151] M.H. Sherif, A.M. Yossef, Synthesis and anticancer evaluation of some fused
(b) B.R. Scott, M.A. Pathak, G.R. Mohn, Molecular and genetic basis of furo- coumarino-[4,3-d]-pyrimidine derivatives, Res. Chem. Intermed. 41 (2015)
coumarin reactions, Mutat. Res. Genet. Toxicol. 39 (1976) 29e74. 383e390.
[129] G. Obi, J.C. Chukwujekwu, F.R. van Heerden, Synthesis and antimicrobial [152] O. Bruno, C. Brullo, S. Schenone, F. Bondavalli, A. Ranise, M. Tognolini,
activity of new prenylated 2-pyrone derivatives, Synth. Commun. 50 (2020) M. Impicciatore, V. Ballabeni, E. Barocelli, Synthesis, antiplatelet and
726e734. antithrombotic activities of new 2-substituted benzopyrano[4,3-d]pyr-
[130] V.V. Mulwad, R.B. Pawar, A.C. Chaskar, Synthesis and antibacterial activity of imidin-4-cycloamines and 4-amino/cycloamino-benzopyrano[4,3-d]pyr-
new tetrazole derivatives, J. Kor. Chem. Soc. 52 (2008) 249e256. imidin-5-ones, Bioorg. Med. Chem. 14 (2006) 121e130.
[131] V.V. Mulwad, A.C. Chaskar, B.P. Langi, Synthesis of pyranobenzopyran de- [153] N. Radulovic, G. Stojanovic, R. Vukicevic, V. Dekic, B. Dekic, R. Palic, New 3,4-
rivatives as a potent antibacterial agent, J. Kor. Chem. Soc. 52 (2008) annelated coumarin derivatives: synthesis, antimicrobial activity, antioxi-
663e667. dant capacity, and molecular modeling, Monatsh. Chem. 137 (2006)
[132] R.R. Zhang, J. Liu, Y. Zhang, M.-Q. Hou, M.-Z. Zhang, F. Zhou, W.-H. Zhang, 1477e1486.
Microwave-assisted synthesis and antifungal activity of novel coumarin [154] R. Aeluri, M. Alla, S. Polepalli, N. Jain, Synthesis and antiproliferative activity
derivatives: pyrano[3,2-c]chromene-2,5-diones, Eur. J. Med. Chem. 116 of imidazo[1,2-a]pyrimidine Mannich bases, Eur. J. Med. Chem. 100 (2015)
(2016) 76e83. 18e23.
[133] Ch Gao, L. Chang, Zh Xu, X.-F. Yan, Ch Ding, F. Zhao, X. Wu, L.-Sh Feng, Recent [155] R. Abdel-Rahman, K. El-mahdy, Biological evaluation of pyrimidopyrimidines
advances of tetrazole derivatives as potential anti-tubercular and anti- as multi-targeted small molecule inhibitors and resistance modifying agents,
malarial agents, Eur. J. Med. Chem. 163 (2019) 404e412. Heterocycles 85 (2012) 2391e2414.
[134] B. Liu, C. Zhou, Z. Zhang, J.D. Roland, B.P. Lee, Antimicrobial property of [156] F. Vairoletti, A. Medeiros, P. Font an, J. Melendrez, C. Tabarez, G. Salinas,
halogenated catechols, Chem. Eng. J. 403 (2021) 126340. J. Franco, M.A. Comini, J. Saldan ~ a, V. Jancik, G. Mahler, C. Saiz, Synthesis of
[135] J.H. Roireau, R.J. Rosano, N.C. Lazzara, T. Chen, J. Bajsa-Hirschel, K.K. Schrader, bicyclic 1,4-thiazepines as novel anti-Trypanosoma brucei brucei agents, Med.
et al., Synthesis of pyranopyrans related to diplopyrone and evaluation as Chem. Commun. 10 (2019) 1481e1487.
antibacterials and herbicides, J. Agric. Food Chem. 68 (2020) 9906e9916. [157] H. Ding, Y. Zhang, M. Bian, W. Yao, C. Ma, Concise assembly of highly
[136] K. Piyush, S. Kuldeep, R.Md Azizur, H.S. Misbahul, W. Pranay, A review of substituted furan-fused 1,4-thiazepines and their Diels-Alder reactions with
benzopyran derivatives in pharmacotherapy of breast cancer, Asian J. benzynes, J. Org. Chem. 73 (2008) 578e584.
Pharmaceut. Clin. Res. 11 (2018) 43e46. [158] M. Khoobi, A. Foroumadi, S. Emami, M. Safavi, G. Dehghan, B.H. Alizadeh,
[137] F. Schmitt, R. Schobert, B. Biersack, New pyranoquinoline derivatives as A. Ramazani, S.K. Ardestani, A. Shafiee, Coumarin-based bioactive com-
vascular-disrupting anticancer agents, Med. Chem. Res. 28 (2019) pounds: facile synthesis and biological evaluation of coumarin-fused 1,4-
1694e1703. thiazepines, Chem. Biol. Drug Des. 78 (2011) 580e586.
[138] M. Khoobi, M. Alipour, A. Moradi, A. Sakhteman, H. Nadri, S.F. Razavi,

39

You might also like